1
|
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
|
2
|
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
|
3
|
Li L, Feldman BJ. Characterization and lineage tracing of a mouse adipose depot reveal properties conserved with human supraclavicular brown adipose tissue. Stem Cell Reports 2025:102509. [PMID: 40409261 DOI: 10.1016/j.stemcr.2025.102509] [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: 09/13/2024] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/25/2025] Open
Abstract
Enhancing energy utilization by increasing the number or activity of beige adipocytes has the potential to be of therapeutic benefit for a broad range of metabolic disorders. However, knowledge gaps in our understanding of the mouse versus human developmental origins of beige fat have inhibited the generation of robust preclinical models, leaving a barrier to the success of therapies. Here, we report that a distinct inguinal beige adipose tissue (ibAT) depot lineage traces to the same Prx1+ cell origins as inguinal white adipose tissue (iWAT) but maintains higher thermogenic activity and capability during aging. We discovered that ibAT has the morphological appearance of human supraclavicular brown adipose tissue (scBAT) and, importantly, conserved molecular markers and developmental origins with human scBAT. Our findings reveal a distinct mouse beige adipose tissue depot and provide a preclinical model of human beige adipose tissue development and maintenance.
Collapse
Affiliation(s)
- Liang Li
- Department of Pediatrics, University of California, San Francisco (UCSF) School of Medicine, San Francisco, CA 94158, USA; Department of Pediatrics, Yale School of Medicine, New Haven, CT 06520, USA.
| | - Brian J Feldman
- Department of Pediatrics, University of California, San Francisco (UCSF) School of Medicine, San Francisco, CA 94158, USA; Department of Pediatrics, Yale School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
4
|
Han X, Feng T, Yang Y, Zhu Z, Shao F, Sun L, Yin Y, Zhang W. Intestinal Epithelial-Derived Exosomes Under Cold Stimulation Promote Adipose Thermogenesis. Metabolites 2025; 15:324. [PMID: 40422900 DOI: 10.3390/metabo15050324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2025] [Revised: 05/09/2025] [Accepted: 05/12/2025] [Indexed: 05/28/2025] Open
Abstract
Background: Whether intestinal epithelial cells can regulate distant adipose tissue remains a mystery. Methods: Cold-stimulated intestinal epithelial cell-derived exosomes (Cold IEC-Exo) play a pivotal role in enhancing adipose thermogenesis and metabolic homeostasis, as demonstrated in this study. Results: IEC-Exo can accumulate in adipose tissue. Compared with IEC-Exo derived from room temperature mice (RT IEC-Exo), Cold IEC-Exo significantly enhanced the thermogenesis of adipose. In vitro, Cold IEC-Exo directly stimulated thermogenesis in primary adipocytes by elevating oxygen consumption rate, proton leak, and fatty acid uptake, with no effect on glucose uptake. Small RNA sequencing identified miR-674-3p as a key mediator enriched in Cold IEC-Exo. miR-674-3p mimicry replicated Cold IEC-Exo effects, augmenting Ucp1 expression, mitochondrial uncoupling, and fatty acid utilization in adipocytes. Local overexpression of miR-674-3p in BAT and sWAT via AAV in vivo enhanced thermogenesis and attenuated diet-induced glucose intolerance. Conclusions: These findings establish that Cold IEC-Exo, via miR-674-3p transfer, drive adipose thermogenic activation and mitigate metabolic dysfunction, highlighting their therapeutic potential in obesity-related disorders.
Collapse
Affiliation(s)
- Xue Han
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Tiange Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Yaxu Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Ziming Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Fangyu Shao
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Lijun Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Yue Yin
- Department of Pharmacology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Weizhen Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| |
Collapse
|
5
|
Nisoli E, Cinti S. What defines a cell type? Perspectives from adipocyte biology. Int J Obes (Lond) 2025; 49:751-754. [PMID: 39627607 PMCID: PMC12095039 DOI: 10.1038/s41366-024-01696-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 05/23/2025]
Affiliation(s)
- Enzo Nisoli
- Center for Study and Research on Obesity, Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy.
| | - Saverio Cinti
- Center of Obesity, Marche Polytechnic University, Ancona, Italy
| |
Collapse
|
6
|
Yao X, Mai X, Tian Y, Liu Y, Jin G, Li Z, Chen S, Dai X, Huang L, Fan Z, Pan G, Pan X, Li X, Yu MC, Sun J, Ou J, Chen H, Xie L. Skeletal muscle-specific Bambi deletion induces hypertrophy and oxidative switching coupling with adipocyte thermogenesis against metabolic disorders. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1352-1368. [PMID: 39821828 DOI: 10.1007/s11427-023-2586-x] [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: 02/04/2024] [Accepted: 04/01/2024] [Indexed: 01/19/2025]
Abstract
Skeletal muscle plays a significant role in both local and systemic energy metabolism. The current investigation aims to explore the role of the Bambi gene in skeletal muscle, focusing on its implications for muscle hypertrophy and systemic metabolism. We hypothesize that skeletal muscle-specific deletion of Bambi induces muscle hypertrophy, improves metabolic performance, and activates thermogenic adipocytes via the reprogramming of progenitor of iWAT, offering potential therapeutic strategies for metabolic syndromes. Leveraging the Chromatin immunoprecipitation (ChIP)-seq and bioinformatics analysis, Bambi gene is shown to be a direct target of HIF2α, which is further confirmed by ChIP-qPCR and promoter luciferase assay. Skeletal muscle-specific Bambi deletion led to significant muscle hypertrophy and improved metabolic parameters, even under high-fat diet conditions. This deletion induced metabolic reprogramming of stromal vascular fractions (SVFs) into thermogenic adipocytes, contributing to systemic metabolic improvements, potentially through the secretory factor. Notably, mice with skeletal muscle-specific Bambi deletion demonstrate resistance to high-fat diet-induced metabolic disorders, highlighting a potential therapeutic pathway for metabolic syndrome management. Thus, skeletal muscle-specific deletion of Bambi triggers muscle growth, enhances metabolic performance, and activates thermogenic adipocytes. These findings suggest Bambi as a novel therapeutic target for metabolic syndromes, providing new insights into the interaction between muscle hypertrophy and systemic metabolic improvement. The study underscores the potential of manipulating muscle physiology to regulate whole-body metabolism, offering a novel perspective on treating metabolic disorders.
Collapse
Affiliation(s)
- Xiangping Yao
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Xudong Mai
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Ye Tian
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Yifan Liu
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, 524023, China
| | - Guanghui Jin
- Department of Hepatic Surgery and Liver transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-sen University; Organ Transplantation Research Center of Guangdong Province, Guangdong province engineering laboratory for transplantation medicine; Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, China
| | - Ze Li
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, China
| | - Shujie Chen
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Xiaoshuang Dai
- BGI Institute of Applied Agriculture, BGI-Shenzhen, Shenzhen, 518120, China
| | - Liujing Huang
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Zijing Fan
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, China
| | - Guihua Pan
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Xiaohan Pan
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong SAR, 999077, China
| | - Xiangmin Li
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China
| | - Michael C Yu
- Department of Biological Sciences, State University of New York at Buffalo, Buffalo, 14260, USA
| | - Jia Sun
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Jingxing Ou
- Department of Hepatic Surgery and Liver transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-sen University; Organ Transplantation Research Center of Guangdong Province, Guangdong province engineering laboratory for transplantation medicine; Guangdong Provincial Key Laboratory of Liver Disease Research, Guangzhou, 510630, China.
| | - Hong Chen
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Liwei Xie
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, 510070, China.
- School of Public Health, Xinxiang Medical University, Xinxiang, 453003, China.
- Institute of Aging Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, 524023, China.
- Department of Internal Medicine, Shunde Women and Children's Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, 528300, China.
- College of Life and Health Sciences, Guangdong Industry Polytechnic, Guangzhou, 510300, China.
| |
Collapse
|
7
|
Liu J, Jiang Y, Liu T, Chen C, Chui L, Cui A, Zhang X, Wang X, Wang Y, Yang C, Zhang Y, Wu T, Yang S, Huang J, Tao C, Zhao J, Wang Y. Single-nucleus RNA sequencing defines adipose tissue subpopulations that contribute to Tibetan pig cold adaptation. BMC Biol 2025; 23:107. [PMID: 40275312 PMCID: PMC12023645 DOI: 10.1186/s12915-025-02211-0] [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: 08/17/2024] [Accepted: 04/09/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Thermogenic beige adipocyte displays a remarkable ability in mammals to adapt to cold environments, but the underlying cellular mechanisms remain unclear, especially in pigs that lack functional UCP1. RESULTS Multilocular beige adipocytes were observed in both Tibetan pigs (cold-tolerant) and Bama pigs (cold-sensitive) after short-term cold exposure (4 ℃ for 3 days). Through single-nucleus RNA sequencing of adipose tissues, including subcutaneous inguinal adipose tissues (IAT) and perirenal adipose tissues (PAT), from both pig breeds at room temperature and cold treatment conditions, we discovered two cell subpopulations specific to Tibetan pigs, PDGFRα+EBF2High in IAT and ADIPOQ+HIF1AHigh in both depots. PDGFRα+EBF2High cells were characterized as potential beige precursors, while ADIPOQ+HIF1AHigh cells were found to express highly thermogenic-related genes. Despite the decrease of the lipogenic subpopulation and the increase of the lipolytic and the thermogenic subpopulations observed in both pig breeds upon cold treatment, Tibetan pigs exhibited stronger cellular and molecular responses compared to Bama pigs. Remarkably, cold-induced de novo beige adipogenesis and white adipocyte browning, likely occurred in Tibetan pigs, while Bama pigs relied more heavily on white browning. Moreover, BMP7, which was highly expressed in the PDGFRα+EBF2High subpopulation, positively regulates porcine beige thermogenic capacity. CONCLUSIONS Our data offers a comprehensive and unprecedented perspective on the heterogeneity and plasticity of adipose tissues of pigs and broadens the understanding of beige fat biology in mammals.
Collapse
Affiliation(s)
- Jiali Liu
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing, 100193, China
| | - Yao Jiang
- National Animal Husbandry Service, Beijing, 100125, China
| | - Tianxia Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Lnstitute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
| | - Chuanhe Chen
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing, 100193, China
| | - Linya Chui
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing, 100193, China
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Along Cui
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Lnstitute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Xueping Zhang
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing, 100193, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Xiao Wang
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing, 100193, China
| | - Yu Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Lnstitute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
| | - Chunhuai Yang
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing, 100193, China
| | - Ying Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Lnstitute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Tianwen Wu
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing, 100193, China
| | - Shulin Yang
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing, 100193, China
| | - Jiaojiao Huang
- College of Life Sciences, Qingdao Agricultural University, Qingdao, 266109, China
| | - Cong Tao
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing, 100193, China.
| | - Jianguo Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Lnstitute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya, 572025, China.
| | - Yanfang Wang
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing, 100193, China.
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya, 572025, China.
| |
Collapse
|
8
|
Asahi R, Udagawa H, Oshiro R, Nakajima S, Kanzawa N, Sano K, Shimizu Y, Okamura T, Fujimi TJ. Histidine and soy isoflavones co-ingestion induces browning of white adipose tissue and promotes lipolysis in female rats. Exp Anim 2025; 74:239-250. [PMID: 39675964 PMCID: PMC12044354 DOI: 10.1538/expanim.24-0138] [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: 10/31/2024] [Accepted: 12/04/2024] [Indexed: 12/17/2024] Open
Abstract
Beige adipocytes arise from white adipocytes in response to cold or other stimuli, known as browning of white adipose. Beige adipocytes play a role similar to that of brown adipocytes, express high levels of uncoupling protein 1 (UCP1), and are responsible for energy consumption via heat production, thus aiding in fat loss. Although histidine (His) and soy isoflavones (Iso) co-ingestion reportedly reduces food intake, body weight, and fat accumulation in female rats, the underlying mechanism remains unclear. Therefore, this study aimed to elucidate the mechanisms whereby histidine and soy isoflavones (His-Iso) co-ingestion suppresses fat accumulation. Female rats were fed a control diet or diet containing Iso, His, or His-Iso for 2 weeks, followed by sampling of periovarian white adipose tissue (poWAT) and retroperitoneal white adipose tissue (rWAT) and adipocyte morphology analysis. Additionally, the expression of browning- and lipid metabolism-related genes was examined. Histochemical analysis revealed the presence of multilocular lipid droplets, representative of beige adipocytes, in the poWAT and rWAT of rats in the His-Iso co-ingestion group. Quantitative PCR analysis showed that His-Iso co-ingestion upregulated brown adipocyte and beige adipocyte markers, including UCP1, indicating that His-Iso intake induces beige adipocytes. Moreover, His-Iso co-ingestion upregulated genes related to fatty acid oxidation (carnitine palmitoyl transferase 1A) and lipolysis (adipose triglyceride lipase) in WATs. In conclusion, His-Iso co-ingestion increases UCP1 expression and morphological changes to beige adipocytes, and suppresses fat accumulation by promotion of lipolysis and fatty acid oxidation in WAT.
Collapse
Affiliation(s)
- Riku Asahi
- Department of Registered Dietitians, Faculty of Health and Nutrition, Bunkyo University, 1100 Namegaya, Chigasaki-shi, Kanagawa 253-8550, Japan
| | - Haruhide Udagawa
- Department of Registered Dietitians, Faculty of Health and Nutrition, Bunkyo University, 1100 Namegaya, Chigasaki-shi, Kanagawa 253-8550, Japan
| | - Remiko Oshiro
- Department of Registered Dietitians, Faculty of Health and Nutrition, Bunkyo University, 1100 Namegaya, Chigasaki-shi, Kanagawa 253-8550, Japan
| | - Shigeru Nakajima
- Department of Registered Dietitians, Faculty of Health and Nutrition, Bunkyo University, 1100 Namegaya, Chigasaki-shi, Kanagawa 253-8550, Japan
| | - Nobuyuki Kanzawa
- Department of Materials and Life Science, Faculty of Science and Technology, Sophia University, 7-1 Kioi, Chiyoda-ku, Tokyo 102-8554, Japan
| | - Kaori Sano
- Department of Chemistry, Faculty of Science, Josai University, 1-1 Keyakidai Sakado-shi, Saitama 350-0295, Japan
| | - Yukiko Shimizu
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Tadashi Okamura
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Takahiko J Fujimi
- Department of Registered Dietitians, Faculty of Health and Nutrition, Bunkyo University, 1100 Namegaya, Chigasaki-shi, Kanagawa 253-8550, Japan
| |
Collapse
|
9
|
Wu D, Eeda V, Maria Z, Rawal K, Wang A, Herlea-Pana O, Babu Undi R, Lim HY, Wang W. Targeting IRE1α improves insulin sensitivity and thermogenesis and suppresses metabolically active adipose tissue macrophages in male obese mice. eLife 2025; 13:RP100581. [PMID: 40244655 PMCID: PMC12005715 DOI: 10.7554/elife.100581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025] Open
Abstract
Overnutrition engenders the expansion of adipose tissue and the accumulation of immune cells, in particular, macrophages, in the adipose tissue, leading to chronic low-grade inflammation and insulin resistance. In obesity, several proinflammatory subpopulations of adipose tissue macrophages (ATMs) identified hitherto include the conventional 'M1-like' CD11C-expressing ATM and the newly discovered metabolically activated CD9-expressing ATM; however, the relationship among ATM subpopulations is unclear. The ER stress sensor inositol-requiring enzyme 1α (IRE1α) is activated in the adipocytes and immune cells under obesity. It is unknown whether targeting IRE1α is capable of reversing insulin resistance and obesity and modulating the metabolically activated ATMs. We report that pharmacological inhibition of IRE1α RNase significantly ameliorates insulin resistance and glucose intolerance in male mice with diet-induced obesity. IRE1α inhibition also increases thermogenesis and energy expenditure, and hence protects against high fat diet-induced obesity. Our study shows that the 'M1-like' CD11c+ ATMs are largely overlapping with but yet non-identical to CD9+ ATMs in obese white adipose tissue. Notably, IRE1α inhibition diminishes the accumulation of obesity-induced metabolically activated ATMs and 'M1-like' ATMs, resulting in the curtailment of adipose inflammation and ensuing reactivation of thermogenesis, without augmentation of the alternatively activated M2 macrophage population. Our findings suggest the potential of targeting IRE1α for the therapeutic treatment of insulin resistance and obesity.
Collapse
Affiliation(s)
- Dan Wu
- Department of Genetics, Heersink School of Medicine, UAB Comprehensive Diabetes Center, University of Alabama at BirminghamBirminghamUnited States
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Sciences CenterOklahoma CityUnited States
| | - Venkateswararao Eeda
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Sciences CenterOklahoma CityUnited States
| | - Zahra Maria
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Sciences CenterOklahoma CityUnited States
| | - Komal Rawal
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Sciences CenterOklahoma CityUnited States
| | | | - Oana Herlea-Pana
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Sciences CenterOklahoma CityUnited States
| | - Ram Babu Undi
- Department of Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Sciences CenterOklahoma CityUnited States
| | - Hui-Ying Lim
- Department of Genetics, Heersink School of Medicine, UAB Comprehensive Diabetes Center, University of Alabama at BirminghamBirminghamUnited States
- Department of Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Sciences CenterOklahoma CityUnited States
| | - Weidong Wang
- Department of Genetics, Heersink School of Medicine, UAB Comprehensive Diabetes Center, University of Alabama at BirminghamBirminghamUnited States
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Sciences CenterOklahoma CityUnited States
| |
Collapse
|
10
|
Youn D, Kim B, Jeong D, Lee JY, Kim S, Sumberzul D, Ginting RP, Lee MW, Song JH, Park YS, Kim Y, Oh CM, Lee M, Cho J. Cross-talks between Metabolic and Translational Controls during Beige Adipocyte Differentiation. Nat Commun 2025; 16:3373. [PMID: 40204764 PMCID: PMC11982337 DOI: 10.1038/s41467-025-58665-x] [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: 06/06/2024] [Accepted: 03/26/2025] [Indexed: 04/11/2025] Open
Abstract
Whether and how regulatory events at the translation stage shape the cellular and metabolic features of thermogenic adipocytes is hardly understood. In this study, we report two hitherto unidentified cross-talk pathways between metabolic and translational regulation in beige adipocytes. By analysing temporal profiles of translation activity and protein level changes during precursor-to-beige differentiation, we found selective translational down-regulation of OXPHOS component-coding mRNAs. The down-regulation restricted to Complexes I, III, IV, and V, is coordinated with enhanced translation of TCA cycle genes, engendering distinct stoichiometry of OXPHOS and TCA cycle components and altering the related metabolic activities in mitochondria of thermogenic adipocytes. Our high-resolution description of ribosome positioning unveiled potentiated ribosome pausing at glutamate codons. The increased stalling is attributable to remodelled glutamate metabolism that decreases glutamates for tRNA charging during pan-adipocyte differentiation. The ribosome pauses decrease protein synthesis and mRNA stability of glutamate codon-rich genes, such as actin cytoskeleton-associated genes.
Collapse
Affiliation(s)
- Daehwa Youn
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Boseon Kim
- Soonchunhyang Institute of Medi-bio Science, Soonchunhyang University, Cheonan, 31151, Republic of Korea
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Dahee Jeong
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
- Soonchunhyang Institute of Medi-bio Science, Soonchunhyang University, Cheonan, 31151, Republic of Korea
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Ju Yeon Lee
- Digital Omics Research Center, Korea Basic Science Institute, Ochang, 28119, Republic of Korea
| | - Seha Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Dulguun Sumberzul
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Rehna Paula Ginting
- Soonchunhyang Institute of Medi-bio Science, Soonchunhyang University, Cheonan, 31151, Republic of Korea
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Min-Woo Lee
- Soonchunhyang Institute of Medi-bio Science, Soonchunhyang University, Cheonan, 31151, Republic of Korea
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Ju Hwan Song
- Digital Omics Research Center, Korea Basic Science Institute, Ochang, 28119, Republic of Korea
| | - Ye Seul Park
- Digital Omics Research Center, Korea Basic Science Institute, Ochang, 28119, Republic of Korea
| | - Yumin Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Chang-Myung Oh
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Mihye Lee
- Soonchunhyang Institute of Medi-bio Science, Soonchunhyang University, Cheonan, 31151, Republic of Korea.
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, 31151, Republic of Korea.
| | - Jun Cho
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea.
| |
Collapse
|
11
|
Alcover S, Ramos-Regalado L, Girón G, Muñoz-García N, Vilahur G. HDL-Cholesterol and Triglycerides Dynamics: Essential Players in Metabolic Syndrome. Antioxidants (Basel) 2025; 14:434. [PMID: 40298782 PMCID: PMC12024175 DOI: 10.3390/antiox14040434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/27/2025] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
Metabolic syndrome (MetS) is a complex cluster of interrelated metabolic disorders that significantly elevate the risk of cardiovascular disease, making it a pressing public health concern worldwide. Among the key features of MetS, dyslipidemia-characterized by altered levels of high-density lipoprotein cholesterol (HDL-C) and triglycerides (TG)-plays a crucial role in the disorder's progression. This review aims to elucidate the intricate interplay between HDL-C and TG within the context of lipid metabolism and cardiovascular health, while also addressing the detrimental impact of various cardiovascular risk factors and associated comorbidities. The dynamics of HDL-C and TG are explored, highlighting their reciprocal relationship and respective contributions to the pathophysiology of MetS. Elevated levels of TGs are consistently associated with reduced concentrations of HDL-C, resulting in a lipid profile that promotes the development of vascular disease. Specifically, as TG levels rise, the protective cardiovascular effects of HDL-C are diminished, leading to the increased accumulation of pro-atherogenic TG-rich lipoproteins and low-density lipoprotein particles within the vascular wall, contributing to the progression of atheromas, which can ultimately result in significant ischemic cardiovascular events. Ultimately, this paper underscores the significance of HDL and TG as essential targets for therapeutic intervention, emphasizing their potential in effectively managing MetS and reducing cardiovascular risk.
Collapse
Affiliation(s)
- Sebastià Alcover
- Research Institute Sant Pau (IR SANT PAU), 08041 Barcelona, Spain; (S.A.); (L.R.-R.); (G.G.); (N.M.-G.)
- Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Lisaidy Ramos-Regalado
- Research Institute Sant Pau (IR SANT PAU), 08041 Barcelona, Spain; (S.A.); (L.R.-R.); (G.G.); (N.M.-G.)
- Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Gabriela Girón
- Research Institute Sant Pau (IR SANT PAU), 08041 Barcelona, Spain; (S.A.); (L.R.-R.); (G.G.); (N.M.-G.)
- Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Natàlia Muñoz-García
- Research Institute Sant Pau (IR SANT PAU), 08041 Barcelona, Spain; (S.A.); (L.R.-R.); (G.G.); (N.M.-G.)
- Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Gemma Vilahur
- Research Institute Sant Pau (IR SANT PAU), 08041 Barcelona, Spain; (S.A.); (L.R.-R.); (G.G.); (N.M.-G.)
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
12
|
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
|
13
|
Passini FS, Bornstein B, Rubin S, Kuperman Y, Krief S, Masschelein E, Mehlman T, Brandis A, Addadi Y, Shalom SHO, Richter EA, Yardeni T, Tirosh A, De Bock K, Zelzer E. Piezo2 in sensory neurons regulates systemic and adipose tissue metabolism. Cell Metab 2025; 37:987-1000.e6. [PMID: 39919739 DOI: 10.1016/j.cmet.2024.12.016] [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: 04/14/2024] [Revised: 10/23/2024] [Accepted: 12/31/2024] [Indexed: 02/09/2025]
Abstract
Systemic metabolism ensures energy homeostasis through inter-organ crosstalk regulating thermogenic adipose tissue. Unlike the well-described inductive role of the sympathetic system, the inhibitory signal ensuring energy preservation remains poorly understood. Here, we show that, via the mechanosensor Piezo2, sensory neurons regulate morphological and physiological properties of brown and beige fat and prevent systemic hypermetabolism. Targeting runt-related transcription factor 3 (Runx3)/parvalbumin (PV) sensory neurons in independent genetic mouse models resulted in a systemic metabolic phenotype characterized by reduced body fat and increased insulin sensitivity and glucose tolerance. Deletion of Piezo2 in PV sensory neurons reproduced the phenotype, protected against high-fat-diet-induced obesity, and caused adipose tissue browning and beiging, likely driven by elevated norepinephrine levels. Finding that brown and beige fat are innervated by Runx3/PV sensory neurons expressing Piezo2 suggests a model in which mechanical signals, sensed by Piezo2 in sensory neurons, protect energy storage and prevent a systemic hypermetabolic phenotype.
Collapse
Affiliation(s)
- Fabian S Passini
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel.
| | - Bavat Bornstein
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Sarah Rubin
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Yael Kuperman
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Sharon Krief
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Evi Masschelein
- Department of Health Sciences and Technology, Laboratory of Exercise and Health, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | - Tevie Mehlman
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Alexander Brandis
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Yoseph Addadi
- MICC Cell Observatory, Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Shira Huri-Ohev Shalom
- Bert Strassburger Metabolic Center for Preventive Medicine, Sheba Medical Center, Tel-Hashomer, Israel
| | - Erik A Richter
- Department of Nutrition, Exercise, and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Tal Yardeni
- Bert Strassburger Metabolic Center for Preventive Medicine, Sheba Medical Center, Tel-Hashomer, Israel
| | - Amir Tirosh
- The Dalia and David Arabov Endocrinology and Diabetes Research Center, Institute of Endocrinology, Sheba Medical Center, Tel-Hashomer, Israel; Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Katrien De Bock
- Department of Health Sciences and Technology, Laboratory of Exercise and Health, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | - Elazar Zelzer
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
14
|
Vinnai BÁ, Arianti R, Fischer-Posovszky P, Wabitsch M, Fésüs L, Kristóf E. The importance of thiamine availability in the thermogenic competency of human adipocytes. Mol Cell Endocrinol 2025; 599:112483. [PMID: 39884417 DOI: 10.1016/j.mce.2025.112483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/20/2025] [Accepted: 01/25/2025] [Indexed: 02/01/2025]
Abstract
Brown and beige adipocytes express uncoupling protein 1 (UCP1), which is located in the inner mitochondrial membrane and facilitates the dissipation of excess energy as heat. The activation of thermogenic adipocytes is a potential therapeutic target for treating type 2 diabetes mellitus, obesity, and related co-morbidities. Therefore, identifying novel approaches to stimulate the function of these adipocytes is crucial for advancing therapeutic strategies. Currently, there are limited amount of human adipocyte cell line models available to study the regulatory mechanisms of browning and key players in thermogenesis. The Simpson-Golabi-Behmel syndrome (SGBS) preadipocyte cell line has been proven as a valuable model to investigate human adipocyte biology. In this study, we investigated how excess thiamine (vitamin B1), and the inhibition of thiamine transporters affect the expression of thermogenic markers and functional parameters during adrenergic stimulation in SGBS adipocytes. We found that limiting thiamine availability by pharmacological inhibitors impeded the dibutyryl-cAMP (db-cAMP)-dependent induction of thiamine transporter 1 and 2 (encoded by SLC19A2 and SLC19A3), UCP1, PGC1a, and other browning markers, as well as proton leak respiration which is associated with UCP1-dependent heat generation. Contrarily, excess thiamine enhanced the db-cAMP-dependent induction of thiamine transporters, while UCP1, PGC1a, and other browning markers were upregulated. In addition, abundant amounts of thiamine increased the basal, unstimulated coupled and uncoupled respiration, and the expression of mitochondrial complex subunits. Our study highlights the critical role of excess thiamine in the thermogenic activation of SGBS adipocytes and its potential to enhance thermogenesis.
Collapse
Affiliation(s)
- Boglárka Ágnes Vinnai
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, H-4032, Debrecen, Hungary; Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, H-4032, Debrecen, Hungary
| | - Rini Arianti
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, H-4032, Debrecen, Hungary; Universitas Muhammadiyah Bangka Belitung, 33134, Pangkalpinang, Indonesia
| | - Pamela Fischer-Posovszky
- German Center for Child and Adolescent Health (DZKJ), Partner Site Ulm, Ulm, Germany; Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Martin Wabitsch
- German Center for Child and Adolescent Health (DZKJ), Partner Site Ulm, Ulm, Germany; Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - László Fésüs
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, H-4032, Debrecen, Hungary
| | - Endre Kristóf
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, H-4032, Debrecen, Hungary.
| |
Collapse
|
15
|
Pessoa VFDS, Hecht M, Nitz N, Hagström L. Adipose Tissue in Chagas Disease: A Neglected Component of Pathogenesis. Pathogens 2025; 14:339. [PMID: 40333112 PMCID: PMC12030347 DOI: 10.3390/pathogens14040339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 05/09/2025] Open
Abstract
Chagas disease (CD), caused by the protozoan T. cruzi, is a serious public health issue with high morbidity and mortality rates. Approximately 7 million people are infected, mostly in Latin America. The pathogenesis is multifactorial and poorly elucidated, particularly regarding the role of adipose tissue (AT). This review aims to explore the complex relationship between T. cruzi and AT, focusing on the possible role of this tissue in CD, as well as to explore the impact of diet on the progression of the disease. T. cruzi infects adipocytes, affecting their function. Chronic infection alters adipose physiology, contributing to systemic inflammation and metabolic disturbances. Adipokines are dysregulated, while markers of inflammation and oxidative stress increase within AT during CD. Additionally, the immune response and clinical aspects of CD may be influenced by the host's diet. High-fat diets (HFDs) impact parasite burden and cardiac pathology in murine models. The complex interaction among T. cruzi infection, AT dysfunction, and dietary factors underscore the complexity of CD pathogenesis. Despite accumulating evidence suggesting the role of AT in CD, further research is needed to elucidate its clinical implications. Understanding the bidirectional relationship between AT and T. cruzi infection may offer insights into disease progression and potential therapeutic targets, highlighting the importance of considering adipose physiology in CD management strategies.
Collapse
Affiliation(s)
- Vitória França dos Santos Pessoa
- Interdisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasilia, Brasília 70910-900, Brazil; (V.F.d.S.P.); (M.H.); (N.N.)
| | - Mariana Hecht
- Interdisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasilia, Brasília 70910-900, Brazil; (V.F.d.S.P.); (M.H.); (N.N.)
| | - Nadjar Nitz
- Interdisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasilia, Brasília 70910-900, Brazil; (V.F.d.S.P.); (M.H.); (N.N.)
| | - Luciana Hagström
- Interdisciplinary Laboratory of Biosciences, Faculty of Medicine, University of Brasilia, Brasília 70910-900, Brazil; (V.F.d.S.P.); (M.H.); (N.N.)
- Faculty of Physical Education, University of Brasília, Brasília 70910-900, Brazil
| |
Collapse
|
16
|
Ji Y, Liu S, Zhang Y, Min Y, Wei L, Guan C, Yu H, Zhang Z. Lysine crotonylation in disease: mechanisms, biological functions and therapeutic targets. Epigenetics Chromatin 2025; 18:13. [PMID: 40119392 PMCID: PMC11929287 DOI: 10.1186/s13072-025-00577-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/24/2025] [Indexed: 03/24/2025] Open
Abstract
Lysine crotonylation (Kcr), a previously unknown post-translational modification (PTM), plays crucial roles in regulating cellular processes, including gene expression, chromatin remodeling, and cellular metabolism. Kcr is associated with various diseases, including neurodegenerative disorders, cancer, cardiovascular conditions, and metabolic syndromes. Despite advances in identifying crotonylation sites and their regulatory enzymes, the molecular mechanisms by which Kcr influences disease progression remain poorly understood. Understanding the interplay between Kcr and other acylation modifications may reveal opportunities for developing targeted therapies. This review synthesizes current research on Kcr, focusing on its regulatory mechanisms and disease associations, and highlights insights into future exploration in epigenetics and therapeutic interventions.
Collapse
Affiliation(s)
- Yu Ji
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University & State Key Lab of Digestive Health & National Clinical Research Center for Digestive Diseases, Beijing, 100050, China
| | - Shanshan Liu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University & State Key Lab of Digestive Health & National Clinical Research Center for Digestive Diseases, Beijing, 100050, China
| | - Yiqiao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University & State Key Lab of Digestive Health & National Clinical Research Center for Digestive Diseases, Beijing, 100050, China
| | - Yiyang Min
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University & State Key Lab of Digestive Health & National Clinical Research Center for Digestive Diseases, Beijing, 100050, China
| | - Luyang Wei
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University & State Key Lab of Digestive Health & National Clinical Research Center for Digestive Diseases, Beijing, 100050, China
| | - Chengjian Guan
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University & State Key Lab of Digestive Health & National Clinical Research Center for Digestive Diseases, Beijing, 100050, China.
| | - Huajing Yu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University & State Key Lab of Digestive Health & National Clinical Research Center for Digestive Diseases, Beijing, 100050, China.
| | - Zhongtao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University & State Key Lab of Digestive Health & National Clinical Research Center for Digestive Diseases, Beijing, 100050, China.
| |
Collapse
|
17
|
Li Y, Shi HX, Li J, Du H, Jia R, Liang YH, Huang XY, Gao XL, Gun SB, Yang QL. Adaptive Thermogenesis and Lipid Metabolism Modulation in Inguinal and Perirenal Adipose Tissues of Hezuo Pigs in Response to Low-Temperature Exposure. Cells 2025; 14:392. [PMID: 40136641 PMCID: PMC11941736 DOI: 10.3390/cells14060392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 03/27/2025] Open
Abstract
In mammals, exposure to low temperatures induces white adipose tissue (WAT) browning and alters lipid metabolism to promote thermogenesis, thereby maintaining body temperature. However, this response varies across different adipose depots. In this study, Hezuo pigs were exposed to either room temperature (23 ± 2 °C) or low temperature (-15 ± 2 °C) for periods of 12 h, 24 h, 48 h, 5 d, 10 d, and 15 d. Inguinal fat (IF) and perirenal fat (PF) were collected and analyzed using hematoxylin and eosin (HE) staining, transmission electron microscopy, RT-qPCR, and RNA-seq. Following cryoexposure, our results demonstrated a significant increase in adipocyte number and a corresponding decrease in cross-sectional area in both IF and PF groups from 24 h to 10 d. While adipocyte numbers were elevated at 12 h and 15 d, these changes were not statistically significant. Moreover, lipid droplets and mitochondria were more abundant, and the mRNA expression levels of thermogenic genes UCP3 and PGC-1α were significantly higher compared to the control group during the 24 h-10 d cold exposure period. No significant changes were observed in the other groups. RNA-seq data indicated that the lipid metabolism of IF and PF peaked on day 5 of low-temperature treatment. In IF tissue, lipid metabolism is mainly regulated by genes such as FABP4, WNT10B, PCK1, PLIN1, LEPR, and ADIPOQ. These genes are involved in the classical lipid metabolism pathway and provide energy for cold adaptation. In contrast, in PF tissue, genes like ATP5F1A, ATP5PO, SDHB, NDUFS8, SDHA, and COX5A play roles within the neurodegenerative disease pathway, and PF tissue has a positive impact on the process related to degenerative diseases. Further investigation is needed to clarify the functions of these candidate genes in lipid metabolism in Hezuo pigs and to explore the genetic mechanisms underlying the cold-resistance traits in local pig populations.
Collapse
Affiliation(s)
- Yao Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Y.L.); (H.-X.S.); (J.L.); (H.D.); (R.J.); (Y.-H.L.); (X.-Y.H.); (X.-L.G.)
| | - Hai-Xia Shi
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Y.L.); (H.-X.S.); (J.L.); (H.D.); (R.J.); (Y.-H.L.); (X.-Y.H.); (X.-L.G.)
| | - Jie Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Y.L.); (H.-X.S.); (J.L.); (H.D.); (R.J.); (Y.-H.L.); (X.-Y.H.); (X.-L.G.)
| | - Hong Du
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Y.L.); (H.-X.S.); (J.L.); (H.D.); (R.J.); (Y.-H.L.); (X.-Y.H.); (X.-L.G.)
| | - Rui Jia
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Y.L.); (H.-X.S.); (J.L.); (H.D.); (R.J.); (Y.-H.L.); (X.-Y.H.); (X.-L.G.)
| | - Yu-Hao Liang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Y.L.); (H.-X.S.); (J.L.); (H.D.); (R.J.); (Y.-H.L.); (X.-Y.H.); (X.-L.G.)
| | - Xiao-Yu Huang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Y.L.); (H.-X.S.); (J.L.); (H.D.); (R.J.); (Y.-H.L.); (X.-Y.H.); (X.-L.G.)
| | - Xiao-Li Gao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Y.L.); (H.-X.S.); (J.L.); (H.D.); (R.J.); (Y.-H.L.); (X.-Y.H.); (X.-L.G.)
| | - Shuang-Bao Gun
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Y.L.); (H.-X.S.); (J.L.); (H.D.); (R.J.); (Y.-H.L.); (X.-Y.H.); (X.-L.G.)
- Gansu Modern Pig Rearing Engineering and Technology Research Center, Lanzhou 730070, China
- Gansu Diebu Juema Pig Science and Technology Backyard, Gannan 740070, China
| | - Qiao-Li Yang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Y.L.); (H.-X.S.); (J.L.); (H.D.); (R.J.); (Y.-H.L.); (X.-Y.H.); (X.-L.G.)
- Gansu Diebu Juema Pig Science and Technology Backyard, Gannan 740070, China
| |
Collapse
|
18
|
Rodríguez-Díaz A, Diéguez C, López M, Freire-Agulleiro Ó. FAcTs on fire: Exploring thermogenesis. ADVANCES IN GENETICS 2025; 113:172-198. [PMID: 40409797 DOI: 10.1016/bs.adgen.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2025]
Abstract
Thermoregulation is a fundamental biological process that allows birds and mammals to maintain a stable internal temperature despite environmental fluctuations, a mechanism shaped by millions of years of evolution. Non-shivering thermogenesis (NST), primarily driven by brown adipose tissue (BAT), plays a central role in thermoregulation by not only helping maintain energy homeostasis but also influencing broader metabolic and physiological processes. Recent research has revealed that BAT thermogenesis is regulated by peripheral hormones and at a central level, with key hypothalamic energy-sensing pathways-such as AMP-activated protein kinase (AMPK) and endoplasmic reticulum (ER) stress-playing critical roles. Beyond its metabolic functions, BAT and NST have emerged as important contributors to tumor biology, offering novel therapeutic strategies for metabolic and oncological diseases. This review explores the intricate mechanisms underpinning NST, including UCP1-dependent thermogenesis and alternative pathways such as creatine cycling, calcium-dependent thermogenesis, and lipid cycling. Emerging evidence further highlights BAT's potential in to modulate tumor metabolism, with pharmacological and genetic approaches showing promise in reshaping the tumor microenvironment. This growing body of knowledge offers exciting prospects for targeting BAT thermogenesis in treating obesity and other metabolic diseases.
Collapse
Affiliation(s)
- Amanda Rodríguez-Díaz
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Madrid, Spain.
| | - Carlos Diéguez
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Madrid, Spain
| | - Miguel López
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Madrid, Spain
| | - Óscar Freire-Agulleiro
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Madrid, Spain.
| |
Collapse
|
19
|
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
|
20
|
Gao Z, Yu Y, Eckel‐Mahan K, Kolonin MG. Caloric Restriction and Telomere Preservation in TERT Knockout Adipocyte Progenitors Does Not Rescue Mice From Metabolic Dysfunction due to a TERT Function in Adipocyte Mitochondria. Aging Cell 2025; 24:e14499. [PMID: 39932851 PMCID: PMC11896407 DOI: 10.1111/acel.14499] [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: 11/04/2024] [Revised: 12/20/2024] [Accepted: 01/09/2025] [Indexed: 02/13/2025] Open
Abstract
Inactivation of telomerase (TERT) in adipocyte progenitor cells (APC) expedites telomere attrition, and the onset of diabetes in mice fed high-fat diet (HFD), which promotes APC over-proliferation and replicative senescence. Here, we show that time-restricted feeding or caloric restriction in the postnatal development of mice subsequently subjected to HFD prevents telomere attrition but not glucose intolerance. This metabolic effect of dietary intervention was not observed for mice with TERT KO in endothelial or myeloid cells. To characterize the telomere-independent effects of TERT in the APC lineage, we analyzed mice with TERT knockout in mature adipocytes (AD-TERT-KO), which do not proliferate and avoid telomere attrition. Analysis of adipocytes from AD-TERT-KO mice indicated reliance on glycolysis and decreased mitochondrial oxidative metabolism. We show that AD-TERT-KO mice have reduced cold tolerance and metabolism abnormality indicating a defect in adaptive thermogenesis, characteristic of aging. Conversely, ectopic TERT expression in brown adipocytes-induced mitochondrial oxidation and thermogenic gene expression. We conclude that TERT plays an important non-canonical function in the mitochondria of adipocytes.
Collapse
Affiliation(s)
- Zhanguo Gao
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Yongmei Yu
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Kristin Eckel‐Mahan
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| | - Mikhail G. Kolonin
- The Brown Foundation Institute of Molecular MedicineUniversity of Texas Health Science CenterHoustonTexasUSA
| |
Collapse
|
21
|
Qiu J, Khedr MA, Pan M, Ferreira CR, Chen J, Snyder MM, Ajuwon KM, Yue F, Kuang S. Ablation of FAM210A in Brown Adipocytes of Mice Exacerbates High-Fat Diet-Induced Metabolic Dysfunction. Diabetes 2025; 74:282-294. [PMID: 39602358 PMCID: PMC11842609 DOI: 10.2337/db24-0294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 11/24/2024] [Indexed: 11/29/2024]
Abstract
Thermogenesis of brown adipose tissue (BAT) provides metabolic benefits against pathologic conditions, such as type 2 diabetes, obesity, cardiovascular disease, and cancer. The thermogenic function of BAT relies on mitochondria, but whether mitochondrial remodeling is required for the beneficial effects of BAT remains unclear. We recently identified FAM210A as a BAT-enriched mitochondrial protein essential for cold-induced thermogenesis through the modulation of OPA1-dependent cristae remodeling. Here, we report a key role of FAM210A in the systemic response to a high-fat diet (HFD). We discovered that an HFD suppressed FAM210A expression, associated with excessive OPA1 cleavage in BAT. Ucp1-Cre-driven BAT-specific Fam210a knockout (Fam210aUKO) similarly elevated OPA1 cleavage, accompanied by whitening of BAT. When subjected to an HFD, Fam210aUKO mice gained similar fat mass as sibling control mice but developed glucose intolerance, insulin resistance, and liver steatosis. The metabolic dysfunction was associated with overall increased lipid content in both the liver and BAT. Additionally, Fam210aUKO leads to inflammation in white adipose tissue. These data demonstrate that FAM210A in BAT is necessary for counteracting HFD-induced metabolic dysfunction but not obesity. ARTICLE HIGHLIGHTS FAM210A regulates cold-induced mitochondrial remodeling through control of OPA1 cleavage, but whether it also plays a role in high-fat diet (HFD)-induced cristae remodeling is unknown. We asked if an HFD would alter the FAM210A level and OPA1 cleavage in brown adipose tissue (BAT) and how FAM210A loss of function would affect diet-induced obesity in mice. We found that an HFD diminished FAM210A expression and accelerated OPA1 cleavage in BAT, and Fam210a knockout exacerbated HFD-induced whitening of BAT, cold intolerance, liver steatosis, white adipose tissue inflammation, and metabolic dysfunction. Our work reveals a physiologic role of FAM210A-mediated BAT mitochondrial remodeling in systemic adaptation to an HFD and suggests that BAT mitochondria may be targeted to treat diet-induced metabolic dysfunction.
Collapse
Affiliation(s)
- Jiamin Qiu
- Department of Animal Sciences, Purdue University, West Lafayette, IN
| | - Mennatallah A. Khedr
- Department of Animal Sciences, Purdue University, West Lafayette, IN
- Department of Orthopaedic Surgery, School of Medicine, Duke University, Durham, NC
| | - Meijin Pan
- Department of Animal Sciences, Purdue University, West Lafayette, IN
| | | | - Jingjuan Chen
- Department of Animal Sciences, Purdue University, West Lafayette, IN
- Department of Orthopaedic Surgery, School of Medicine, Duke University, Durham, NC
| | - Madigan M. Snyder
- Department of Animal Sciences, Purdue University, West Lafayette, IN
- Department of Biological Sciences, Purdue University, West Lafayette, IN
| | - Kolapo M. Ajuwon
- Department of Animal Sciences, Purdue University, West Lafayette, IN
| | - Feng Yue
- Department of Animal Sciences, Purdue University, West Lafayette, IN
- Department of Animal Sciences, University of Florida, Gainesville, FL
| | - Shihuan Kuang
- Department of Animal Sciences, Purdue University, West Lafayette, IN
- Department of Orthopaedic Surgery, School of Medicine, Duke University, Durham, NC
- Institute for Cancer Research, Purdue University, West Lafayette, IN
| |
Collapse
|
22
|
Zou M, Tanabe K, Amo-Shiinoki K, Kohno D, Kagawa S, Shirasawa H, Ikeda K, Taguchi A, Ohta Y, Okuya S, Yamada T, Kitamura T, Masutani H, Tanizawa Y. Txnip deficiency causes a susceptibility to acute cold stress with brown fat dysfunction in mice. J Biol Chem 2025; 301:108293. [PMID: 39947474 PMCID: PMC11938133 DOI: 10.1016/j.jbc.2025.108293] [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: 05/24/2024] [Revised: 10/29/2024] [Accepted: 11/11/2024] [Indexed: 03/29/2025] Open
Abstract
Mammals adaptively regulate energy metabolism in response to environmental changes such as starvation and cold circumstances. Thioredoxin-interacting protein (Txnip), known as a redox regulator, serves as a nutrient sensor regulating energy homeostasis. Txnip is essential for mice to adapt to starvation, but its role in adapting to cold circumstances remains unclear. Here, we identified Txnip as a pivotal factor for maintaining nonshivering thermogenesis in mice. Txnip protein levels in brown adipose tissue (BAT) were upregulated by the acute cold exposure. Txnip-deficient (Txnip-/-) mice acclimated to thermoneutrality (30 °C) exhibited significant BAT enlargement and triglyceride accumulation with downregulation of BAT signature and metabolic gene expression. Upon acute cold exposure (5 °C), Txnip-/- mice showed a rapid decline in BAT surface temperatures with the failure of increasing metabolic respiration, developing lethal hypothermia. The BAT dysfunction and cold susceptibility in Txnip-/- mice were corrected by acclimation to 16 °C, protecting the mice from life-threatening hypothermia. Transcriptomic and metabolomic analysis using dissected BAT revealed that despite preserving glycolysis, the BAT of Txnip-/- mice failed to activate the catabolism of branched-chain amino acids and fatty acids in response to acute cold stress. These findings illustrate that Txnip is required for maintaining basal BAT function and ensuring cold-induced thermogenesis.
Collapse
Affiliation(s)
- Meng Zou
- Division of Endocrinology, Metabolism, Hematological Sciences and Therapeutics, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Katsuya Tanabe
- Division of Endocrinology, Metabolism, Hematological Sciences and Therapeutics, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan.
| | - Kikuko Amo-Shiinoki
- Division of Endocrinology, Metabolism, Hematological Sciences and Therapeutics, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Daisuke Kohno
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, Japan
| | - Syota Kagawa
- Department of Natural Products Chemistry, Daiichi University of Pharmacy, Fukuoka, Japan
| | - Hideki Shirasawa
- Division of Endocrinology, Metabolism, Hematological Sciences and Therapeutics, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Kenji Ikeda
- Department of Molecular Endocrinology and Metabolism, Tokyo Medical and Dental University, Tokyo, Japan
| | - Akihiko Taguchi
- Division of Endocrinology, Metabolism, Hematological Sciences and Therapeutics, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Yasuharu Ohta
- Division of Endocrinology, Metabolism, Hematological Sciences and Therapeutics, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Shigeru Okuya
- Health Administration Center, Organization for University Education, Yamaguchi University, Yamaguchi, Japan
| | - Tetsuya Yamada
- Department of Molecular Endocrinology and Metabolism, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tadahiro Kitamura
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, Japan
| | - Hiroshi Masutani
- Department of Clinical Laboratory Sciences, Faculty of Health Care, Tenri University, Tenri, Nara, Japan
| | - Yukio Tanizawa
- Division of Endocrinology, Metabolism, Hematological Sciences and Therapeutics, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| |
Collapse
|
23
|
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
|
24
|
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
|
25
|
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
|
26
|
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
|
27
|
Abdulla A, Sadida HQ, Jerobin J, Elfaki I, Mir R, Mirza S, Singh M, Macha MA, Uddin S, Fakhro K, Bhat AA, Akil ASAS. Unraveling molecular interconnections and identifying potential therapeutic targets of significance in obesity-cancer link. JOURNAL OF THE NATIONAL CANCER CENTER 2025; 5:8-27. [PMID: 40040878 PMCID: PMC11873641 DOI: 10.1016/j.jncc.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/16/2024] [Accepted: 11/11/2024] [Indexed: 03/06/2025] Open
Abstract
Obesity, a global health concern, is associated with severe health issues like type 2 diabetes, heart disease, and respiratory complications. It also increases the risk of various cancers, including melanoma, endometrial, prostate, pancreatic, esophageal adenocarcinoma, colorectal carcinoma, renal adenocarcinoma, and pre-and post-menopausal breast cancer. Obesity-induced cellular changes, such as impaired CD8+ T cell function, dyslipidemia, hypercholesterolemia, insulin resistance, mild hyperglycemia, and fluctuating levels of leptin, resistin, adiponectin, and IL-6, contribute to cancer development by promoting inflammation and creating a tumor-promoting microenvironment rich in adipocytes. Adipocytes release leptin, a pro-inflammatory substance that stimulates cancer cell proliferation, inflammation, and invasion, altering the tumor cell metabolic pathway. Adiponectin, an insulin-sensitizing adipokine, is typically downregulated in obese individuals. It has antiproliferative, proapoptotic, and antiangiogenic properties, making it a potential cancer treatment. This narrative review offers a comprehensive examination of the molecular interconnections between obesity and cancer, drawing on an extensive, though non-systematic, survey of the recent literature. This approach allows us to integrate and synthesize findings from various studies, offering a cohesive perspective on emerging themes and potential therapeutic targets. The review explores the metabolic disturbances, cellular alterations, inflammatory responses, and shifts in the tumor microenvironment that contribute to the obesity-cancer link. Finally, it discusses potential therapeutic strategies aimed at disrupting these connections, offering valuable insights into future research directions and the development of targeted interventions.
Collapse
Affiliation(s)
- Alanoud Abdulla
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Research Program, Sidra Medicine, Doha, Qatar
| | - Hana Q. Sadida
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Research Program, Sidra Medicine, Doha, Qatar
| | - Jayakumar Jerobin
- Qatar Metabolic Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Imadeldin Elfaki
- Department of Biochemistry, Faculty of Science, University of Tabuk, Tabuk, Saudi Arabia
| | - Rashid Mir
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Sameer Mirza
- Department of Chemistry, College of Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Mayank Singh
- Department of Medical Oncology (Lab.), Dr. BRAIRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Muzafar A. Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Pulwama, Jammu and Kashmir, India
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Laboratory of Animal Research Center, Qatar University, Doha, Qatar
| | - Khalid Fakhro
- Department of Human Genetics, Sidra Medicine, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
- Department of Genetic Medicine, Weill Cornell Medicine, Doha, Qatar
| | - Ajaz A. Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Research Program, Sidra Medicine, Doha, Qatar
| | - Ammira S. Al-Shabeeb Akil
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Research Program, Sidra Medicine, Doha, Qatar
| |
Collapse
|
28
|
Wu D, Eeda V, Maria Z, Rawal K, Wang A, Herlea-Pana O, Undi RB, Lim HY, Wang W. Targeting IRE1α improves insulin sensitivity and thermogenesis and suppresses metabolically active adipose tissue macrophages in male obese mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.17.603931. [PMID: 39071288 PMCID: PMC11275733 DOI: 10.1101/2024.07.17.603931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Overnutrition engenders the expansion of adipose tissue and the accumulation of immune cells, in particular, macrophages, in the adipose tissue, leading to chronic low-grade inflammation and insulin resistance. In obesity, several proinflammatory subpopulations of adipose tissue macrophages (ATMs) identified hitherto include the conventional "M1-like" CD11C-expressing ATM and the newly discovered metabolically activated CD9-expressing ATM; however, the relationship among ATM subpopulations is unclear. The ER stress sensor inositol-requiring enzyme 1α (IRE1α) is activated in the adipocytes and immune cells under obesity. It is unknown whether targeting IRE1α is capable of reversing insulin resistance and obesity and modulating the metabolically activated ATMs. We report that pharmacological inhibition of IRE1α RNase significantly ameliorates insulin resistance and glucose intolerance in male mice with diet-induced obesity. IRE1α inhibition also increases thermogenesis and energy expenditure, and hence protects against high fat diet-induced obesity. Our study shows that the "M1-like" CD11c+ ATMs are largely overlapping with but yet non-identical to CD9+ ATMs in obese white adipose tissue. Notably, IRE1α inhibition diminishes the accumulation of obesity-induced metabolically activated ATMs and "M1-like" ATMs, resulting in the curtailment of adipose inflammation and ensuing reactivation of thermogenesis, without augmentation of the alternatively activated M2 macrophage population. Our findings suggest the potential of targeting IRE1α for the therapeutic treatment of insulin resistance and obesity.
Collapse
Affiliation(s)
- Dan Wu
- Department of Genetics, Heersink School of Medicine, UAB Comprehensive Diabetes Center, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, Alabama, 35233, United States
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Science Center, 941 Stanton L. Young Boulevard, Oklahoma City, Oklahoma 73104, United States
| | - Venkateswararao Eeda
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Science Center, 941 Stanton L. Young Boulevard, Oklahoma City, Oklahoma 73104, United States
| | - Zahra Maria
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Science Center, 941 Stanton L. Young Boulevard, Oklahoma City, Oklahoma 73104, United States
| | - Komal Rawal
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Science Center, 941 Stanton L. Young Boulevard, Oklahoma City, Oklahoma 73104, United States
| | - Audrey Wang
- Indian Springs School, 190 Woodward Dr, Pelham, Alabama 35124
| | - Oana Herlea-Pana
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Science Center, 941 Stanton L. Young Boulevard, Oklahoma City, Oklahoma 73104, United States
| | - Ram Babu Undi
- Department of Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Science Center, 941 Stanton L. Young Boulevard, Oklahoma City, Oklahoma 73104, United States
| | - Hui-Ying Lim
- Department of Genetics, Heersink School of Medicine, UAB Comprehensive Diabetes Center, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, Alabama, 35233, United States
- Department of Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Science Center, 941 Stanton L. Young Boulevard, Oklahoma City, Oklahoma 73104, United States
| | - Weidong Wang
- Department of Genetics, Heersink School of Medicine, UAB Comprehensive Diabetes Center, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, Alabama, 35233, United States
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Science Center, 941 Stanton L. Young Boulevard, Oklahoma City, Oklahoma 73104, United States
| |
Collapse
|
29
|
Cammisotto V, Valeriani E, Pignatelli P, Violi F. Nicotinamide Adenine Dinucleotide Phosphate Oxidases and Metabolic Dysfunction-Associated Steatotic Liver Disease. Antioxidants (Basel) 2025; 14:83. [PMID: 39857417 PMCID: PMC11763266 DOI: 10.3390/antiox14010083] [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: 11/08/2024] [Revised: 01/01/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is characterized by lipid accumulation in the liver due to an excess in their supplies or an impairment in their management. While some patients remain stable for years, a proportion of them progress up to steatohepatitis (MASH). MASLD links with systemic pathways being associated with metabolic and non-metabolic diseases. Although liver lipid accumulation represents the first hit for MASLD, the pathophysiology of its development and progression to MASH remains not completely understood. Oxidative stress has received particular attention in recent years, as most of the oxidative process occurs in the liver, which is also the target of oxidative stress-induced damage. Growing evidence linked the activity of nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOX) to the increased liver production of reactive oxygen species up to liver damage and fibrosis. NOX acts both in hepatocytes and in non-parenchymal hepatic cells, contributing to hepatocyte lipotoxicity, impaired hepatic microcirculation, hepatic stellate, and mesenchymal stem cells activation and proliferation. This review aims to summarize the current knowledge on the involvement of oxidative stress in the MASLD-MASH transition, focusing on the role of NOX isoforms, and to suggest targeting NOX as a therapeutic approach in MASLD.
Collapse
Affiliation(s)
- Vittoria Cammisotto
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy; (V.C.); (P.P.); (F.V.)
| | - Emanuele Valeriani
- Department of General Surgery and Surgical Specialty, Sapienza University of Rome, 00185 Rome, Italy
- Department of Infectious Disease, Azienda Ospedaliero-Universitaria Policlinico Umberto I, 00161 Rome, Italy
| | - Pasquale Pignatelli
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy; (V.C.); (P.P.); (F.V.)
| | - Francesco Violi
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy; (V.C.); (P.P.); (F.V.)
| |
Collapse
|
30
|
Su D, Jiang T, Song Y, Li D, Zhan S, Zhong T, Guo J, Li L, Zhang H, Wang L. Identification of a distal enhancer of Ucp1 essential for thermogenesis and mitochondrial function in brown fat. Commun Biol 2025; 8:31. [PMID: 39789228 PMCID: PMC11718246 DOI: 10.1038/s42003-025-07468-3] [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: 08/20/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025] Open
Abstract
Uncoupling protein 1 (UCP1) is a crucial protein located in the mitochondrial inner membrane that mediates nonshivering thermogenesis. However, the molecular mechanisms by which enhancer-promoter chromatin interactions control Ucp1 transcriptional regulation in brown adipose tissue (BAT) are unclear. Here, we employed circularized chromosome conformation capture coupled with next-generation sequencing (4C-seq) to generate high-resolution chromatin interaction profiles of Ucp1 in interscapular brown adipose tissue (iBAT) and epididymal white adipose tissue (eWAT) and revealed marked changes in Ucp1 chromatin interaction between iBAT and eWAT. Next, we identified four iBAT-specific active enhancers of Ucp1, and three of them were activated by cold stimulation. Transcriptional repression of the Ucp1-En4 or Ucp1-En6 region significantly downregulated Ucp1 and impaired mitochondrial function in brown adipocytes. Furthermore, depletion of the cohesin subunit RAD21 decreased the interaction intensity between Ucp1-En4 and the Ucp1 promoter and downregulated Ucp1. EBF2 cooperated with the acetyltransferase CBP to regulate Ucp1-En4 activity and increase Ucp1 transcriptional activity. In vivo, lentivirus-mediated repression of Ucp1-En4 was injected into iBAT, resulting in impacted iBAT thermogenic capacity and impaired iBAT mitochondrial function under cold acclimation conditions. Studying the functional enhancers regulating Ucp1 expression in iBAT will provide important insights into the regulatory mechanisms of BAT activity.
Collapse
Affiliation(s)
- Duo Su
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Tingting Jiang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Yulong Song
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Die Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Siyuan Zhan
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Tao Zhong
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Jiazhong Guo
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Li Li
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Hongping Zhang
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China
| | - Linjie Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China.
- Key Laboratory of Livestock and Poultry Multi-omics, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China.
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China.
- Key Laboratory of Agricultural Bioinformatics, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China.
| |
Collapse
|
31
|
Luk C, Bridge KI, Warmke N, Simmons KJ, Drozd M, Moran A, MacCannell ADV, Cheng CW, Straw S, Scragg JL, Smith J, Ozber CH, Wilkinson CG, Skromna A, Makava N, Prag HA, Simon Futers T, Brown OI, Bruns AF, Walker AM, Watt NT, Mughal R, Griffin KJ, Yuldasheva NY, Limumpornpetch S, Viswambharan H, Sukumar P, Beech DJ, Vidal-Puig A, Witte KK, Murphy MP, Hartley RC, Wheatcroft SB, Cubbon RM, Roberts LD, Kearney MT, Haywood NJ. Paracrine role of endothelial IGF-1 receptor in depot-specific adipose tissue adaptation in male mice. Nat Commun 2025; 16:170. [PMID: 39747815 PMCID: PMC11696296 DOI: 10.1038/s41467-024-54669-1] [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: 11/23/2021] [Accepted: 11/18/2024] [Indexed: 01/04/2025] Open
Abstract
During recent decades, changes in lifestyle have led to widespread nutritional obesity and its related complications. Remodelling adipose tissue as a therapeutic goal for obesity and its complications has attracted much attention and continues to be actively explored. The endothelium lines all blood vessels and is close to all cells, including adipocytes. The endothelium has been suggested to act as a paracrine organ. We explore the role of endothelial insulin-like growth factor-1 receptor (IGF-1R), as a paracrine modulator of white adipose phenotype. We show that a reduction in endothelial IGF-1R expression in the presence of high-fat feeding in male mice leads to depot-specific beneficial white adipose tissue remodelling, increases whole-body energy expenditure and enhances insulin sensitivity via a non-cell-autonomous paracrine mechanism. We demonstrate that increased endothelial malonate may be contributory and that malonate prodrugs have potentially therapeutically relevant properties in the treatment of obesity-related metabolic disease.
Collapse
Affiliation(s)
- Cheukyau Luk
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Katherine I Bridge
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Nele Warmke
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Katie J Simmons
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- School of Biomedical Sciences, Faculty of Biological Sciences & Astbury Centre, University of Leeds, Leeds, UK
| | - Michael Drozd
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Amy Moran
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Amanda D V MacCannell
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Chew W Cheng
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Sam Straw
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Jason L Scragg
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Jessica Smith
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Claire H Ozber
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- Division of Gastroenterology & Surgery, Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Chloe G Wilkinson
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- North West Genomic Laboratory Hub, Manchester University NHS Foundation Trust, St Mary's Hospital, Oxford Road, Manchester, UK
| | - Anna Skromna
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Natallia Makava
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Hiran A Prag
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - T Simon Futers
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Oliver I Brown
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Alexander-Francisco Bruns
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Andrew Mn Walker
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Nicole T Watt
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Romana Mughal
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- Department of Optometry and Vision Sciences, University of Huddersfield, Huddersfield, UK
| | - Kathryn J Griffin
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Nadira Y Yuldasheva
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Sunti Limumpornpetch
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- Division of Internal Medicine, Cardiology Unit, Faculty of Medicine Prince of Songkla University, Songkhla, Thailand
| | - Hema Viswambharan
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Piruthivi Sukumar
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - David J Beech
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | | | - Klaus K Witte
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | | | - Stephen B Wheatcroft
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Richard M Cubbon
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Lee D Roberts
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Mark T Kearney
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK.
| | - Natalie J Haywood
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| |
Collapse
|
32
|
Kim JG, Park J, Baek S, Won S, Cho J. Effects of Melissa officinalis Extracts on Obesity and Anxiety. Clin Nutr Res 2025; 14:65-77. [PMID: 39968272 PMCID: PMC11832291 DOI: 10.7762/cnr.2025.14.1.65] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/22/2025] [Accepted: 01/30/2025] [Indexed: 02/20/2025] Open
Abstract
Obesity is a significant global health concern that not only increases metabolic disorders risks but also impacts mental health, particularly affecting women due to hormonal fluctuations and societal pressures. This study investigated anti-obesity and anti-anxiety effects of lemon balm (Melissa officinalis) extracts in female C57BL/6 mouse (n = 16, 17 weeks old) fed a high-fat diet (HFD). We compared 2 extracts method: distilled water (LBD, n = 5) and 80% ethanol (LBE, n = 6), administered via oral gavage (200 mg/kg/day) for 6 weeks alongside HFD. Both extract groups showed lower weight increase ratio compared to the control group in experiment period (n = 5) (LBD: 27.74%, LBE: 29.71% vs. Control: 51.88%, p < 0.05). The extracts significantly decreased mesenteric white adipose tissue (mWAT) among WATs examined (mWAT and parametrial white adipose tissue [pWAT]). While both LBD and LBE reduced fatty acid synthase (FAS) mRNA expression in pWAT, only LBD reduced peroxisome proliferator-activated receptor gamma and FAS mRNA expression in mWAT. In elevated plus maze behavioral experiments, the LBD group displayed reduced anxiety-like behavior, spending significantly more time and travelling greater distances in the open arms compared to other groups (p < 0.05), independent of brain inflammatory markers. Our findings demonstrate lemon balm extracts simultaneously address both obesity and anxiety-like behaviors in female mice, with extraction solvent-dependent variations in efficacy and mechanism of action. These results suggest potential therapeutic applications for lemon balm as a functional food ingredient, particularly for women experiencing concurrent obesity and anxiety symptoms.
Collapse
Affiliation(s)
- Jin Gyeom Kim
- Department of Food and Nutrition, Chungnam National University, Daejeon 34134, Korea
| | - Jiye Park
- Department of Food and Nutrition, Chungnam National University, Daejeon 34134, Korea
| | - Sooyeon Baek
- Department of Food and Nutrition, Chungnam National University, Daejeon 34134, Korea
| | - Seunghyun Won
- Department of Food and Nutrition, Chungnam National University, Daejeon 34134, Korea
| | - Jin Cho
- Department of Food and Nutrition, Chungnam National University, Daejeon 34134, Korea
| |
Collapse
|
33
|
Blüher M. Understanding Adipose Tissue Dysfunction. J Obes Metab Syndr 2024; 33:275-288. [PMID: 39734091 PMCID: PMC11704217 DOI: 10.7570/jomes24013] [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: 04/25/2024] [Revised: 06/08/2024] [Accepted: 12/16/2024] [Indexed: 12/31/2024] Open
Abstract
Diseases affecting adipose tissue (AT) function include obesity, lipodystrophy, and lipedema, among others. Both a lack of and excess AT are associated with increased risk for developing diseases including type 2 diabetes mellitus, hypertension, obstructive sleep apnea, and some types of cancer. However, individual risk of developing cardiometabolic and other 'obesity-related' diseases is not entirely determined by fat mass. Rather than excess fat accumulation, AT dysfunction may represent the mechanistic link between obesity and comorbid diseases. There are people who remain metabolically healthy despite obesity, whereas people with normal weight or very low subcutaneous AT mass may develop typically obesity-related diseases. AT dysfunction is characterized by adipocyte hypertrophy, impaired subcutaneous AT expandability (ectopic fat deposition), hypoxia, a variety of stress, inflammatory processes, and the release of proinflammatory, diabetogenic, and atherogenic signals. Genetic and environmental factors might contribute to AT heterogeneity either alone or via interaction with intrinsic biological factors. However, many questions remain regarding the mechanisms of AT dysfunction initiation and whether and how it could be reversed. Do AT signatures define clinically relevant subtypes of obesity? Is the cellular composition of AT associated with variation in obesity phenotypes? What roles do environmental compounds play in the manifestation of AT dysfunction? Answers to these and other questions may explain AT disease mechanisms and help to define strategies for improving AT health. This review focuses on recent advances in our understanding of AT biology.
Collapse
Affiliation(s)
- 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
| |
Collapse
|
34
|
AKYÜREK F, TUNCEZ AKYÜREK F, ŞENGÜL BAĞ F. Relationship between uncoupling protein 1 (UCP1) levels and psoriasis. Turk J Med Sci 2024; 55:215-222. [PMID: 40104293 PMCID: PMC11913510 DOI: 10.55730/1300-0144.5960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 02/18/2025] [Accepted: 12/30/2024] [Indexed: 03/20/2025] Open
Abstract
Background/aim Psoriasis is a common chronic autoimmune skin disease. Comorbidities increase the mortality risk of the disease. The aim of this study was to investigate the changes in uncoupling protein 1 (UCP1) level in psoriasis patients and evaluate its possible role in the pathogenesis of the disease, focusing on disease severity (Psoriasis Area and Severity Index), dyslipidemia, inflammation, and cardiovascular risk. Materials and methods This study included 30 psoriasis patients and 30 healthy individuals as a control group. Serum UCP1 was measured using an ELISA test kit. The laboratory results of psoriasis patients and healthy controls were compared. Results UCP1 level was a significant candidate marker for the prediction of psoriatic disease (AUC: 0.708, 95% CI: 0.577-0.819, p = 0.002) with sensitivity of 66.67%, specificity of 76.67%, negative predictive value of 69.7%, and positive predictive value of 74.1%. Simple logistic regression analysis showed that an individual with a UCP1 value below 7.561 had a 73% lower probability (OR: 0.27, 95% CI: 0.08-0.94, p = 0.039) of developing psoriasis than an individual with a UCP1 value above 7.561. Among the biochemical parameters, the high-sensitivity C-reactive protein and triglyceride levels of the patients were significantly higher compared to those of the healthy controls while their high-density lipoprotein levels were lower. Conclusion According to the sensitivity (66.67%) and specificity (76.67%) of UCP1, it may be a valuable candidate marker in the diagnosis of psoriasis patients in symptomatic and asymptomatic phases. Further work is needed to substantiate these findings.
Collapse
Affiliation(s)
- Fikret AKYÜREK
- Department of Medical Biochemistry, Faculty of Medicine, Selçuk University, Konya,
Turkiye
| | - Fatma TUNCEZ AKYÜREK
- Department of Dermatology, Faculty of Medicine, Selçuk University, Konya,
Turkiye
| | - Fatma ŞENGÜL BAĞ
- Department of Biochemistry, Faculty of Pharmacy, Adıyaman University, Adıyaman,
Turkiye
| |
Collapse
|
35
|
Zhao Z, Hu L, Song B, Jiang T, Wu Q, Lin J, Li X, Cai Y, Li J, Qian B, Liu S, Lang J, Yang Z. Constitutively active receptor ADGRA3 signaling induces adipose thermogenesis. eLife 2024; 13:RP100205. [PMID: 39718208 DOI: 10.7554/elife.100205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2024] Open
Abstract
The induction of adipose thermogenesis plays a critical role in maintaining body temperature and improving metabolic homeostasis to combat obesity. β3-adrenoceptor (β3-AR) is widely recognized as a canonical β-adrenergic G-protein-coupled receptor (GPCR) that plays a crucial role in mediating adipose thermogenesis in mice. Nonetheless, the limited expression of β3-AR in human adipocytes restricts its clinical application. The objective of this study was to identify a GPCR that is highly expressed in human adipocytes and to explore its potential involvement in adipose thermogenesis. Our research findings have demonstrated that the adhesion G-protein-coupled receptor A3 (ADGRA3), an orphan GPCR, plays a significant role in adipose thermogenesis through its constitutively active effects. ADGRA3 exhibited high expression levels in human adipocytes and mouse brown fat. Furthermore, the knockdown of Adgra3 resulted in an exacerbated obese phenotype and a reduction in the expression of thermogenic markers in mice. Conversely, Adgra3 overexpression activated the adipose thermogenic program and improved metabolic homeostasis in mice without exogenous ligand. We found that ADGRA3 facilitates the biogenesis of beige human or mouse adipocytes in vitro. Moreover, hesperetin was identified as a potential agonist of ADGRA3, capable of inducing adipocyte browning and ameliorating insulin resistance in mice. In conclusion, our study demonstrated that the overexpression of constitutively active ADGRA3 or the activation of ADGRA3 by hesperetin can induce adipocyte browning by Gs-PKA-CREB axis. These findings indicate that the utilization of hesperetin and the selective overexpression of ADGRA3 in adipose tissue could serve as promising therapeutic strategies in the fight against obesity.
Collapse
Affiliation(s)
- Zewei Zhao
- Shenzhen Key Laboratory of Systems Medicine for inflammatory diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
| | - Longyun Hu
- Shenzhen Key Laboratory of Systems Medicine for inflammatory diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
| | - Bigui Song
- Shenzhen Key Laboratory of Systems Medicine for inflammatory diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
| | - Tao Jiang
- Shenzhen Key Laboratory of Systems Medicine for inflammatory diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
| | - Qian Wu
- Shenzhen Key Laboratory of Systems Medicine for inflammatory diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
| | - Jiejing Lin
- Shenzhen Key Laboratory of Systems Medicine for inflammatory diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
| | - Xiaoxiao Li
- Shenzhen Key Laboratory of Systems Medicine for inflammatory diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
| | - Yi Cai
- Shenzhen Key Laboratory of Systems Medicine for inflammatory diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
| | - Jin Li
- Shenzhen Key Laboratory of Systems Medicine for inflammatory diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
| | - Bingxiu Qian
- Shenzhen Key Laboratory of Systems Medicine for inflammatory diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
| | - Siqi Liu
- Shenzhen Key Laboratory of Systems Medicine for inflammatory diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
| | - Jilu Lang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Zhonghan Yang
- Shenzhen Key Laboratory of Systems Medicine for inflammatory diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, China
| |
Collapse
|
36
|
Vatner DE, Zhang J, Vatner SF. Brown adipose tissue enhances exercise performance and healthful longevity. Aging (Albany NY) 2024; 16:13442-13451. [PMID: 39699442 PMCID: PMC11723650 DOI: 10.18632/aging.206179] [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: 11/13/2024] [Accepted: 12/02/2024] [Indexed: 12/20/2024]
Abstract
Brown adipose tissue (BAT), a major subtypes of adipose tissues, is known for thermogenesis and promoting healthful longevity. Our hypothesis is that BAT protects against impaired healthful longevity, i.e., obesity, diabetes, cardiovascular disorders, cancer, Alzheimer's disease, and reduced exercise tolerance. While most prior studies have shown that exercise regulates BAT activation and improves BAT density, relatively few have shown that BAT increases exercise performance. In contrast, our recent studies with the regulator of G protein signaling 14 (RGS14) knockout (KO) model of extended longevity showed that it enhances exercise performance, mediated by its more potent BAT, compared with BAT from wild type mice. For example, when the BAT from RGS14 KO mice is transplanted to WT mice, their exercise capacity is enhanced at 3 days after BAT transplantation, whereas BAT transplantation from WT to WT mice increased exercise performance, but only at 8 weeks after transplantation. The goal of this research perspective is to review the role of BAT in mediating healthful longevity, specifically exercise capacity. In view of the ability of BAT to mediate healthful longevity and enhance exercise performance, it is likely that a pharmaceutical analog of BAT will become a novel therapeutic modality.
Collapse
Affiliation(s)
- Dorothy E. Vatner
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Jie Zhang
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Stephen F. Vatner
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| |
Collapse
|
37
|
Mutlu HH, Koç Ada S, Uzunlulu M, Mutlu HH, Sargın M, Oğuz A. A comparison of brown fat tissue related hormone levels in metabolically healthy and unhealthy individuals with obesity. Endocrine 2024; 86:1025-1034. [PMID: 39008201 PMCID: PMC11554687 DOI: 10.1007/s12020-024-03960-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 07/06/2024] [Indexed: 07/16/2024]
Abstract
PURPOSE One of the key functions of brown adipose tissue is its positive impact on metabolism. This study aimed to examine the potential involvement of brown fat-related hormones in the development of metabolically healthy obesity. Specifically, we sought to compare the levels of NRG4, FGF21, and irisin between metabolically healthy and unhealthy individuals with obesity. METHODS Patients with BMI ≥ 30 kg/m2 and aged between 20 and 50 years were included in the study. Among these patients, those who did not have any metabolic syndrome criteria except for increased waist circumference were defined as metabolically healthy obese. Age, gender, BMI, body fat, and muscle mass, matched metabolically healthy and unhealthy obese groups were compared in terms of FGF21, irisin, and NRG4 levels. RESULTS Metabolically healthy and unhealthy obese groups were similar in terms of age and gender. There was no difference between the two groups in terms of BMI, weight, total body fat, muscle, fat-free mass, distribution of body fat and muscle mass. No statistically significant difference was found between irisin, NRG4, and FGF21 levels between metabolically healthy and unhealthy individuals with obesity. It was found that irisin had a significant inverse correlation with BMI and body fat percentage. CONCLUSION The present study showed no difference between metabolically healthy and unhealthy obese individuals in terms of irisin, FGF21, and NRG4 levels. The weak association between irisin and BMI and body fat percentage may suggest a potential link between irisin with metabolic health.
Collapse
Affiliation(s)
- Hacer Hicran Mutlu
- Department of Family Medicine, Faculty of Medicine, Istanbul Medeniyet University, Istanbul, Turkey.
| | - Saniye Koç Ada
- Department of Biochemistry, Faculty of Medicine, Istanbul Medeniyet University, Istanbul, Turkey
| | - Mehmet Uzunlulu
- Department of Internal Medicine, Faculty of Medicine, Istanbul Medeniyet University, Istanbul, Turkey
| | - Hasan Hüseyin Mutlu
- Department of Family Medicine, Faculty of Medicine, Health Sciences University, Istanbul, Turkey
| | - Mehmet Sargın
- Department of Family Medicine, Faculty of Medicine, Istanbul Medeniyet University, Istanbul, Turkey
| | - Aytekin Oğuz
- Department of Internal Medicine, Faculty of Medicine, Istanbul Medeniyet University, Istanbul, Turkey
| |
Collapse
|
38
|
Lai TH, Hwang JS, Ngo QN, Lee DK, Kim HJ, Kim DR. A comparative assessment of reference genes in mouse brown adipocyte differentiation and thermogenesis in vitro. Adipocyte 2024; 13:2330355. [PMID: 38527945 PMCID: PMC10965104 DOI: 10.1080/21623945.2024.2330355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 03/06/2024] [Indexed: 03/27/2024] Open
Abstract
Adipogenic differentiation and thermogenesis in brown adipose tissue (BAT) undergo dynamic processes, altering phenotypes and gene expressions. Proper reference genes in gene expression analysis are crucial to mitigate experimental variances and ensure PCR efficacy. Unreliable reference genes can lead to erroneous gene expression quantification, resulting in data misinterpretation. This study focused on identifying suitable reference genes for mouse brown adipocyte research, utilizing brown adipocytes from the Ucp1-luciferase ThermoMouse model. Comparative analysis of gene expression data under adipogenesis and thermogenesis conditions was conducted, validating 13 housekeeping genes through various algorithms, including DeltaCq, BestKeeper, geNorm, Normfinder, and RefFinder. Tbp and Rer1 emerged as optimal references for Ucp1 and Pparg expression in brown adipogenesis, while Tbp and Ubc were ideal for the expression analysis of these target genes in thermogenesis. Conversely, certain conventional references, including Actb, Tubb5, and Gapdh, proved unstable as reference genes under both conditions. These findings stress the critical consideration of reference gene selection in gene expression analysis within specific biological systems to ensure accurate conclusions.
Collapse
Affiliation(s)
- Trang Huyen Lai
- Department of Biochemistry and Convergence Medical Sciences and Institute of Medical Science, Gyeongsang National University, College of Medicine, Jinju, South Korea
| | - Jin Seok Hwang
- Department of Biochemistry and Convergence Medical Sciences and Institute of Medical Science, Gyeongsang National University, College of Medicine, Jinju, South Korea
| | - Quang Nhat Ngo
- Department of Biochemistry and Convergence Medical Sciences and Institute of Medical Science, Gyeongsang National University, College of Medicine, Jinju, South Korea
| | - Dong-Kun Lee
- Department of Physiology and Convergence Medical Sciences and Institute of Medical Science, Gyeongsang National University, College of Medicine, Jinju, South Korea
| | - Hyun Joon Kim
- Department of Anatomy and Convergence Medical Sciences and Institute of Medical Science, Gyeongsang National University, College of Medicine, Jinju, South Korea
| | - Deok Ryong Kim
- Department of Biochemistry and Convergence Medical Sciences and Institute of Medical Science, Gyeongsang National University, College of Medicine, Jinju, South Korea
| |
Collapse
|
39
|
Zhu Y, Yang R, Deng Z, Deng B, Zhao K, Dai C, Wei G, Wang Y, Zheng J, Ren Z, Lv W, Xiao Y, Mei Z, Song T. Adipose Tissue-Resident Sphingomonas Paucimobilis Suppresses Adaptive Thermogenesis by Reducing 15-HETE Production and Inhibiting AMPK Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2310236. [PMID: 39476363 DOI: 10.1002/advs.202310236] [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: 12/27/2023] [Revised: 09/26/2024] [Indexed: 12/19/2024]
Abstract
Obesity represents a low-grade chronic inflammation status, which is associated with compromised adaptive thermogenesis. However, the mechanisms underlying the defective activation of thermogenesis in chronic inflammation remain unclear. Here, a chronic inflammatory model is first estabolished by injecting mice with low-dose lipopolysaccharide (LPS) before cold exposure, and then it is verified that LPS treatment can decrease the core body temperature of mice and alter the microbial distribution in epididymal white adipose tissue (eWAT). An adipose tissue-resident bacterium Sphingomonas paucimobilis is identified as a potential inhibitor on the activation of brown fat and browning of inguinal WAT, resulting in defective adaptive thermogenesis. Mechanically, LPS and S. paucimobilis inhibit the production and release of 15-HETE by suppressing its main metabolic enzyme 12 lipoxygenase (12-LOX) and 15- Hydroxyeicosatetraenoic acid (15-HETE) rescues the impaired thermogenesis. Interestingly, 15-HETE directly binds to AMP-activated protein kinase α (AMPKα) and elevates the phosphorylation of AMPK, leading to the activation of uncoupling protein 1 (UCP1) and mitochondrial oxidative phosphorylation (OXPHOS) complexes. Further analysis with human obesity subjects reveals that individuals with high body mass index displayed lower 15-HETE levels. Taken together, this work improves the understanding of how chronic inflammation impairs adaptive thermogenesis and provides novel targets for alleviating obesity.
Collapse
Affiliation(s)
- Yucheng Zhu
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Ruiqi Yang
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhangchao Deng
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Bohua Deng
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Kun Zhao
- Department of Endocrinology, the Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, China
| | - Chen Dai
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - 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
| | - YanJiang Wang
- Beijing Chao-yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jinshui Zheng
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhuqing Ren
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Wentao Lv
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Yingping Xiao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310021, China
| | - Zhinan Mei
- College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Tongxing Song
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| |
Collapse
|
40
|
Niu Z, Hildebrand S, Kappes S, Ali ME, Vogel M, Mikhael M, Ran D, Kozak J, Wiedner M, Richter DF, Lamprecht A, Pfeifer A. Enhanced browning of adipose tissue by mirabegron-microspheres. J Control Release 2024; 375:601-613. [PMID: 39278357 DOI: 10.1016/j.jconrel.2024.09.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 09/18/2024]
Abstract
Thermogenic brown adipose tissue (BAT) has emerged as an attractive target for combating obesity. However, pharmacological activation of energy expenditure by BAT and/or induction of browning of white adipose tissue (WAT) has been hampered by cardiovascular side effects. To address these concerns, we developed polylactide-co-glycolide acid (PLGA) microspheres loaded with mirabegron (MIR), a selective beta-3 adrenergic receptor (ADRB3) agonist, to achieve sustained local induction and activation of thermogenic adipocytes. MIR-loaded PLGA microspheres (MIR-MS) effectively activated brown adipocytes and enhanced the thermogenic program in white adipocytes. Moreover, treating isolated inguinal WAT (iWAT) with MIR-MS resulted in increased expression of browning markers and elevated lipolysis mainly via ADRB3. In mice, injection of MIR-MS over four weeks induced browning of iWAT at the injection site. Importantly, local MIR-MS injection successfully mitigated unwanted cardiovascular risks, including high systolic blood pressure (SBP) and heart rate, as compared to MIR-treated mice. Finally, injecting MIR-MS into human subcutaneous WAT led to a significant induction of lipolysis and an increase in the expression of thermogenic marker uncoupling protein 1 (UCP1). Taken together, our findings indicate that MIR-MS function as a local drug release system that induces browning of human and murine subcutaneous WAT while mitigating undesirable cardiovascular effects.
Collapse
Affiliation(s)
- Zheming Niu
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany
| | - Staffan Hildebrand
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany
| | - Sebastian Kappes
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Bonn, Germany
| | - Mohamed Ehab Ali
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Bonn, Germany
| | - Matthias Vogel
- Pharmacogenomic, Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany
| | - Mickel Mikhael
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany
| | - Danli Ran
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany
| | - Jan Kozak
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Bonn, Germany
| | - Maria Wiedner
- Institut ID, Beethoven Clinic, Plastic and Aesthetic Surgery Cologne, Cologne, Germany
| | - Dirk F Richter
- Institut ID, Beethoven Clinic, Plastic and Aesthetic Surgery Cologne, Cologne, Germany
| | - Alf Lamprecht
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Bonn, Germany.
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany.
| |
Collapse
|
41
|
Kim YJ, Lee SG, Jang SI, Kim WK, Oh KJ, Bae KH, Kim HJ, Seong JK. Lactate utilization in Lace1 knockout mice promotes browning of inguinal white adipose tissue. Exp Mol Med 2024; 56:2491-2502. [PMID: 39511428 PMCID: PMC11612233 DOI: 10.1038/s12276-024-01324-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: 01/04/2024] [Revised: 06/02/2024] [Accepted: 07/02/2024] [Indexed: 11/15/2024] Open
Abstract
Recent studies have focused on identifying novel genes involved in the browning process of inguinal white adipose tissue (iWAT). In this context, we propose that the mitochondrial ATPase gene lactation elevated 1 (Lace1) utilizes lactate to regulate the browning capacity of iWAT, specifically in response to challenge with CL-316,243 (CL), a beta3-adrenergic receptor (β3-AR) agonist. The mice were injected with CL over a span of 3 days and exposed to cold temperatures (4-6 °C) for 1 week. The results revealed a significant increase in Lace1 expression levels during beige adipogenesis. Additionally, a strong positive correlation was observed between Lace1 and Ucp1 mRNA expression in iWAT under browning stimulation. To further explore this phenomenon, we subjected engineered Lace1 KO mice to CL and cold challenges to validate their browning potential. Surprisingly, Lace1 KO mice presented increased oxygen consumption and heat generation upon CL challenge and cold exposure, along with increased expression of genes related to brown adipogenesis. Notably, deletion of Lace1 led to increased lactate uptake and browning in iWAT under CL challenge compared with those of the controls. These unique phenomena stem from increased lactate release due to the inactivation of pyruvate dehydrogenase (PDH) in the hearts of Lace1 KO mice.
Collapse
Affiliation(s)
- Youn Ju Kim
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Korea Model animal Priority Center (KMPC), Seoul National University, Seoul, Republic of Korea
- Division of Endocrine and Kidney Disease Research, Department of Chronic Disease Convergence Research, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Sang Gyu Lee
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
- Korea Model animal Priority Center (KMPC), Seoul National University, Seoul, Republic of Korea
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, Republic of Korea
| | - Su In Jang
- Korea Model animal Priority Center (KMPC), Seoul National University, Seoul, Republic of Korea
| | - Won Kon Kim
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Kyoung-Jin Oh
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Kwang-Hee Bae
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Hye Jin Kim
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.
- Korea Model animal Priority Center (KMPC), Seoul National University, Seoul, Republic of Korea.
| | - Je Kyung Seong
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.
- Korea Model animal Priority Center (KMPC), Seoul National University, Seoul, Republic of Korea.
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, Republic of Korea.
- Interdisciplinary Program for Bioinformatics, Program for Cancer Biology, BIO-MAX/N-Bio Institute, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
42
|
Yao Z, Liang S, Chen J, Zhang H, Chen W, Li H. Dietary Lactate Intake and Physical Exercise Synergistically Reverse Brown Adipose Tissue Whitening to Ameliorate Diet-Induced Obesity. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024. [PMID: 39486070 DOI: 10.1021/acs.jafc.4c06899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Physical exercise represents an effective strategy for combating obesity via brown adipose tissue (BAT) activation, but the mechanism remains unclear. In this study, we demonstrated that the cooperation between lactate and adrenoceptor signaling regulated BAT activity during exercise. The lactate receptor GPR81 was highly expressed in the BAT of lean mice, whereas its expression was markedly decreased in obese mice. Notably, the level of GPR81 in BAT could be upregulated by exercise. The blockade of lactate production or GPR81 significantly impaired exercise-induced BAT activation. In addition, dietary lactate intake enhanced the efficacy of physical exercise in alleviating BAT whitening in obese mice, as evidenced by the improved mitochondrial ultrastructure, reduced lipid droplets, increased UCP1 expression, and elevated mitochondrial DNA content. Further data indicated that norepinephrine triggered UCP1 activation through both the cAMP/PKA and Ca2+/CaMK pathways during exercise, while lactate mediated this process via the GPR81-Ca2+/CaMK cascade. Our findings unveil a novel mechanism in the regulation of BAT function by physical exercise, providing a promising lifestyle intervention to improve metabolic health.
Collapse
Affiliation(s)
- Zhijie Yao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Shuxiao Liang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Jinxiang Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Haitao Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
43
|
Mohaghegh N, Ahari A, Buttles C, Davani S, Hoang H, Huang Q, Huang Y, Hosseinpour B, Abbasgholizadeh R, Cottingham AL, Farhadi N, Akbari M, Kang H, Khademhosseini A, Jucaud V, Pearson RM, Hassani Najafabadi A. Simvastatin-Loaded Polymeric Nanoparticles: Targeting Inflammatory Macrophages for Local Adipose Tissue Browning in Obesity Treatment. ACS NANO 2024; 18:27764-27781. [PMID: 39342648 DOI: 10.1021/acsnano.4c10742] [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] [Indexed: 10/01/2024]
Abstract
Obesity is defined as chronic, low-grade inflammation within specific tissues. Given the escalating prevalence of obesity among individuals of all ages, obesity has reached epidemic proportions, posing an important public health challenge. Despite significant advancements in treating obesity, conventional approaches remain largely ineffective or involve severe side effects, thus underscoring the pressing need to explore and develop treatment approaches. Targeted and local immunomodulation using nanoparticles (NPs) can influence fat production and utilization processes. Statins, known for their anti-inflammatory properties, show the potential for mitigating obesity-related inflammation. A localized delivery option offers several advantages over oral and parenteral delivery methods. Here, we developed simvastatin (Sim) encapsulated within PLGA NPs (Sim-NP) for localized delivery of Sim to adipose tissues (ATs) for immunomodulation to treat obesity. In vitro experiments revealed the strong anti-inflammatory effects of Sim-NPs, which resulted in enhanced modulation of macrophage (MΦ) polarization and induction of AT browning. We then extended our investigation to an in vivo mouse model of high-fat-diet (HFD)-induced obesity. Sim-NP administration led to the controlled release of Sim within AT, directly impacting MΦ activity and inducing AT browning while inducing weight loss. Our findings demonstrated that Sim-NP administration effectively inhibited the progression of obesity-related inflammation, controlled white fat production, and enhanced AT modulation. These results highlight the potential of Sim-NP as a potent nanotherapy for treating obesity by modulating the immune system.
Collapse
Affiliation(s)
- Neda Mohaghegh
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Amir Ahari
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
- Department of Surgery, University of California-Los Angeles, Los Angeles, California 90095, United States
| | - Claire Buttles
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
- Department of Biology, Indiana University Bloomington, Bloomington, Indiana 47405, United States
| | - Saya Davani
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
- Department of Neurobiology, Physiology, and Behavior, University of California Davis, Briggs Hall, Davis, California 95616, United States
| | - Hanna Hoang
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California 90024, United States
| | - Qiang Huang
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Yixuan Huang
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Bahareh Hosseinpour
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
- Department of Chemistry, Carleton University, Ottawa, Ontario K1S 5B6, Canada
| | - Reza Abbasgholizadeh
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Andrea L Cottingham
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| | - Neda Farhadi
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Mohsen Akbari
- Department of Mechanical Engineering, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
- Biotechnology Center, Silesian University of Technology, Gliwice 44-100, Poland
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, Los Angeles, California 90064, United States
| | - Ryan M Pearson
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| | | |
Collapse
|
44
|
Gong D, Lei J, He X, Hao J, Zhang F, Huang X, Gu W, Yang X, Yu J. Keys to the switch of fat burning: stimuli that trigger the uncoupling protein 1 (UCP1) activation in adipose tissue. Lipids Health Dis 2024; 23:322. [PMID: 39342273 PMCID: PMC11439242 DOI: 10.1186/s12944-024-02300-z] [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: 05/25/2024] [Accepted: 09/14/2024] [Indexed: 10/01/2024] Open
Abstract
As one of the main pathogenic factors of cardiovascular and cerebrovascular diseases, the incidence of metabolic diseases such as adiposity and metabolic dysfunction-associated steatotic liver disease (MASLD) is increasing annually. It is urgent and crucial to find more therapeutic targets to treat these diseases. Mainly expressed in brown adipocytes, mitochondrial uncoupling protein 1 (UCP1) is key to the thermogenesis of classical brown adipose tissue (BAT). Furthermore, white adipose tissue (WAT) is likely to express more UCP1 and subsequently acquire the ability to undergo thermogenesis under certain stimuli. Therefore, targeting and activating UCP1 to promote increased BAT thermogenesis and browning of WAT are helpful in treating metabolic diseases, such as adiposity and MASLD. In this case, the stimuli that activate UCP1 are emerging. Therefore, we summarize the thermogenic stimuli that have activated UCP1 in recent decades, among which cold exposure is one of the stimuli first discovered to activate BAT thermogenesis. As a convenient and efficient therapy with few side effects and good metabolic benefits, physical exercise can also activate the expression of UCP1 in adipose tissue. Notably, for the first time, we have summarized and demonstrated the stimuli of traditional Chinese medicines that can activate UCP1, such as acupuncture, Chinese herbal formulas, and Chinese medicinal herbs. Moreover, pharmacological agents, functional foods, food ingredients, and the gut microbiota are also commonly associated with regulating and activating UCP1. The identification and analysis of UCP1 stimuli can greatly facilitate our understanding of adipose tissue thermogenesis, including the browning of WAT. Thus, it is more conducive to further research and therapy for glucose and lipid metabolism disorders.
Collapse
Affiliation(s)
- Dihong Gong
- Yunnan University of Chinese Medicine, Kunming, 650000, Yunnan, China
| | - Juanhong Lei
- Yunnan University of Chinese Medicine, Kunming, 650000, Yunnan, China
| | - Xudong He
- Yunnan University of Chinese Medicine, Kunming, 650000, Yunnan, China
| | - Junjie Hao
- Yunnan University of Chinese Medicine, Kunming, 650000, Yunnan, China
| | - Fan Zhang
- Yunnan University of Chinese Medicine, Kunming, 650000, Yunnan, China
| | - Xinya Huang
- Yunnan University of Chinese Medicine, Kunming, 650000, Yunnan, China
| | - Wen Gu
- Yunnan University of Chinese Medicine, Kunming, 650000, Yunnan, China
| | - Xingxin Yang
- Yunnan University of Chinese Medicine, Kunming, 650000, Yunnan, China.
| | - Jie Yu
- Yunnan University of Chinese Medicine, Kunming, 650000, Yunnan, China.
| |
Collapse
|
45
|
Yonemoto E, Ihara R, Tanaka E, Mitani T. Cocoa extract induces browning of white adipocytes and improves glucose intolerance in mice fed a high-fat diet. Biosci Biotechnol Biochem 2024; 88:1188-1198. [PMID: 39025807 DOI: 10.1093/bbb/zbae105] [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: 04/02/2024] [Accepted: 07/12/2024] [Indexed: 07/20/2024]
Abstract
Cocoa extract (CE) offers several health benefits, such as antiobesity and improved glucose intolerance. However, the mechanisms remain unclear. Adipose tissue includes white adipose tissue (WAT) and brown adipose tissue. Brown adipose tissue leads to body fat reduction by metabolizing lipids to heat via uncoupling protein 1 (UCP1). The conversion of white adipocytes into brown-like adipocytes (beige adipocytes) is called browning, and it contributes to the anti-obesity effect and improved glucose tolerance. This study aimed to evaluate the effect of CE on glucose tolerance in terms of browning. We found that dietary supplementation with CE improved glucose intolerance in mice fed a high-fat diet, and it increased the expression levels of Ucp1 and browning-associated gene in inguinal WAT. Furthermore, in primary adipocytes of mice, CE induced Ucp1 expression through β3-adrenergic receptor stimulation. These results suggest that dietary CE improves glucose intolerance by inducing browning in WAT.
Collapse
MESH Headings
- Animals
- Diet, High-Fat/adverse effects
- Glucose Intolerance/drug therapy
- Glucose Intolerance/metabolism
- Cacao/chemistry
- Plant Extracts/pharmacology
- Mice
- Uncoupling Protein 1/metabolism
- Uncoupling Protein 1/genetics
- Male
- Adipocytes, White/drug effects
- Adipocytes, White/metabolism
- Mice, Inbred C57BL
- Adipose Tissue, White/drug effects
- Adipose Tissue, White/metabolism
- Receptors, Adrenergic, beta-3/metabolism
- Receptors, Adrenergic, beta-3/genetics
- Adipose Tissue, Brown/drug effects
- Adipose Tissue, Brown/metabolism
- Adipocytes, Brown/drug effects
- Adipocytes, Brown/metabolism
Collapse
Affiliation(s)
- Eito Yonemoto
- D ivision of Food Science and Biotechnology, Graduated School of Science and Technology, Shinshu University, Kamiina, Nagano, Japan
| | - Risa Ihara
- Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, Kamiina, Nagano, Japan
| | - Emi Tanaka
- D ivision of Food Science and Biotechnology, Graduated School of Science and Technology, Shinshu University, Kamiina, Nagano, Japan
| | - Takakazu Mitani
- D ivision of Food Science and Biotechnology, Graduated School of Science and Technology, Shinshu University, Kamiina, Nagano, Japan
- Department of Agricultural and Life Sciences, Faculty of Agriculture, Shinshu University, Kamiina, Nagano, Japan
| |
Collapse
|
46
|
Yang M, Ge J, Liu YL, Wang HY, Wang ZH, Li DP, He R, Xie YY, Deng HY, Peng XM, Wang WS, Liu JD, Zhu ZZ, Yu XF, Maretich P, Kajimura S, Pan RP, Chen Y. Sortilin-mediated translocation of mitochondrial ACSL1 impairs adipocyte thermogenesis and energy expenditure in male mice. Nat Commun 2024; 15:7746. [PMID: 39232011 PMCID: PMC11374900 DOI: 10.1038/s41467-024-52218-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024] Open
Abstract
Beige fat activation involves a fuel switch to fatty acid oxidation following chronic cold adaptation. Mitochondrial acyl-CoA synthetase long-chain family member 1 (ACSL1) localizes in the mitochondria and plays a key role in fatty acid oxidation; however, the regulatory mechanism of the subcellular localization remains poorly understood. Here, we identify an endosomal trafficking component sortilin (encoded by Sort1) in adipose tissues that shows dynamic expression during beige fat activation and facilitates the translocation of ACSL1 from the mitochondria to the endolysosomal pathway for degradation. Depletion of sortilin in adipocytes results in an increase of mitochondrial ACSL1 and the activation of AMPK/PGC1α signaling, thereby activating beige fat and preventing high-fat diet (HFD)-induced obesity and insulin resistance. Collectively, our findings indicate that sortilin controls adipose tissue fatty acid oxidation by substrate fuel selection during beige fat activation and provides a potential targeted approach for the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Min Yang
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Ge
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Lian Liu
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huan-Yu Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhi-Han Wang
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dan-Pei Li
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui He
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Yu Xie
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong-Yan Deng
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xue-Min Peng
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wen-She Wang
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia-Dai Liu
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zeng-Zhe Zhu
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xue-Feng Yu
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China
| | - Pema Maretich
- Research Laboratory of Electronics and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Shingo Kajimura
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Howard Hughes Medical Institute, Boston, MA, USA
| | - Ru-Ping Pan
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Nuclear Medicine, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Chen
- Division of Endocrinology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Research Group of Endocrinology & Metabolism, Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Branch of National Clinical Research Center for Metabolic Diseases, Hubei, China.
| |
Collapse
|
47
|
Undrakhbayar E, Zhang XY, Wang CZ, Wang DH. The function of brown adipose tissue at different sites of the body in Brandt's voles during cold acclimation. Comp Biochem Physiol A Mol Integr Physiol 2024; 295:111655. [PMID: 38723743 DOI: 10.1016/j.cbpa.2024.111655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 05/06/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024]
Abstract
Ambient temperatures have great impacts on thermoregulation of small mammals. Brown adipose tissue (BAT), an obligative thermogenic tissue for small mammals, is localized not only in the interscapular depot (iBAT), but also in supraclavicular, infra/subscapular, cervical, paravertebral, and periaortic depots. The iBAT is known for its cold-induced thermogenesis, however, less has been paid attention to the function of BAT at other sites. Here, we investigated the function of BAT at different sites of the body during cold acclimation in a small rodent species. As expected, Brandt's voles (Lasiopodomys brandtii) consumed more food and reduced the body mass gain when they were exposed to cold. The voles increased resting metabolic rate and maintained a relatively lower body temperature in the cold (36.5 ± 0.27 °C) compared to those in the warm condition (37.1 ± 0.36 °C). During cold acclimation, the uncoupling protein 1 (UCP1) increased in aBAT (axillary), cBAT (anterior cervical), iBAT (interscapular), nBAT (supraclavicular), and sBAT (suprascapular). The levels of proliferating cell nuclear antigen (PCNA), a marker for cell proliferation, were higher in cBAT and iBAT in the cold than in the warm group. The pAMPK/AMPK and pCREB/CREB were increased in cBAT and iBAT during cold acclimation, respectively. These data indicate that these different sites of BAT play the cold-induced thermogenic function for small mammals.
Collapse
Affiliation(s)
- Enkhbat Undrakhbayar
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xue-Ying Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Chen-Zhu Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China
| | - De-Hua Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing 100049, China; School of Life Sciences, Shandong University, Qingdao 266237, China.
| |
Collapse
|
48
|
Shi R, He M, Peng Y, Xia X. Homotherapy for heteropathy: Interleukin-41 and its biological functions. Immunology 2024; 173:1-13. [PMID: 38594835 DOI: 10.1111/imm.13791] [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: 12/03/2023] [Accepted: 03/29/2024] [Indexed: 04/11/2024] Open
Abstract
Interleukin-41 (IL-41) is a newly discovered cytokine, named Cometin, Subfatin, meteorin-like transcription (Metrnl), and so forth. It is widely expressed in animals and can exert its biological roles through autocrine and paracrine forms. It has functions such as anti-inflammatory, improving body metabolism, regulating immunity, regulating fat metabolism and participates in the process of autoimmune disease or inflammatory injury. It plays an important role in psoriasis, diabetes, Crohn's disease (CD), osteoarthritis, Kawasaki disease (KD), Graves' disease, autoimmune hepatitis, infertility, obesity, sepsis, cardiovascular diseases and respiratory diseases. This paper reviews the biological functions of IL-41, the relationship between IL-41 and diseases, the effects of IL-41 in the cytokine network and the possible signalling pathways. In order to explore the same target or the same drug for the treatment of different diseases from the perspective of homotherapy for heteropathy, cytokine strategies based on IL-41 have been put forward for the precise treatment of immune diseases and inflammatory diseases. It is worth noting that IL-41 related preparations for lung protection and smoking cessation are interesting research fields.
Collapse
Affiliation(s)
- Runfeng Shi
- The First Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Meixin He
- School of Public Health, Southern Medical University, Guangzhou, China
| | - Yongzheng Peng
- Department of Transfusion Medicine and Laboratory Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Xu Xia
- Southern Medical University Library, Guangzhou, China
| |
Collapse
|
49
|
Li X, Pham K, Ysaguirre J, Mahmud I, Tan L, Wei B, Shao LJ, Elizondo M, Habib R, Elizondo F, Sesaki H, Lorenzi PL, Sun K. Mechanistic insights into metabolic function of dynamin-related protein 1. J Lipid Res 2024; 65:100633. [PMID: 39182608 PMCID: PMC11426057 DOI: 10.1016/j.jlr.2024.100633] [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: 04/13/2024] [Revised: 07/17/2024] [Accepted: 08/13/2024] [Indexed: 08/27/2024] Open
Abstract
Dynamin-related protein 1 (DRP1) plays crucial roles in mitochondrial and peroxisome fission. However, the mechanisms underlying the functional regulation of DRP1 in adipose tissue during obesity remain unclear. To elucidate the metabolic and pathological significance of diminished DRP1 in obese adipose tissue, we utilized adipose tissue-specific DRP1 KO mice challenged with a high-fat diet. We observed significant metabolic dysregulations in the KO mice. Mechanistically, DRP1 exerts multifaceted functions in mitochondrial dynamics and endoplasmic reticulum (ER)-lipid droplet crosstalk in normal mice. Loss of function of DRP1 resulted in abnormally giant mitochondrial shapes, distorted mitochondrial membrane structure, and disrupted cristae architecture. Meanwhile, DRP1 deficiency induced the retention of nascent lipid droplets in ER, leading to perturbed overall lipid dynamics in the KO mice. Collectively, dysregulation of the dynamics of mitochondria, ER, and lipid droplets contributes to whole-body metabolic disorders, as evidenced by perturbations in energy metabolites. Our findings demonstrate that DRP1 plays diverse and critical roles in regulating energy metabolism within adipose tissue during the progression of obesity.
Collapse
Affiliation(s)
- Xin Li
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Katherine Pham
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Jazmin Ysaguirre
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Iqbal Mahmud
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Lin Tan
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Bo Wei
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Long J Shao
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Maryam Elizondo
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Rabie Habib
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Fathima Elizondo
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Hiromi Sesaki
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Philip L Lorenzi
- Metabolomics Core Facility, Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kai Sun
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, Texas, USA; Graduate Program in Biochemistry and Cellular Biology, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, Texas, USA.
| |
Collapse
|
50
|
Velez‐delValle C, Hernandez‐Mosqueira CP, Castro‐Rodriguez LI, Vazquez‐Sandoval A, Marsch‐Moreno M, Kuri‐Harcuch W. Gene expression and characterization of clonally derived murine embryonic brown and brite adipocytes. FEBS Open Bio 2024; 14:1503-1525. [PMID: 38972757 PMCID: PMC11492321 DOI: 10.1002/2211-5463.13861] [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: 01/12/2024] [Revised: 05/29/2024] [Accepted: 06/25/2024] [Indexed: 07/09/2024] Open
Abstract
White adipocytes store energy, while brown and brite adipocytes release heat via nonshivering thermogenesis. In this study, we characterized two murine embryonic clonal preadipocyte lines, EB5 and EB7, each displaying unique gene marker expression profiles. EB5 cells differentiate into brown adipocytes, whereas EB7 cells into brite (also known as beige) adipocytes. To draw a comprehensive comparison, we contrasted the gene expression patterns, adipogenic capacity, as well as carbohydrate and lipid metabolism of these cells to that of F442A, a well-known white preadipocyte and adipocyte model. We found that commitment to differentiation in both EB5 and EB7 cells can be induced by 3-Isobutyl-1-methylxanthine/dexamethasone (Mix/Dex) and staurosporine/dexamethasone (St/Dex) treatments. Additionally, the administration of rosiglitazone significantly enhances the brown and brite adipocyte phenotypes. Our data also reveal the involvement of a series of genes in the transcriptional cascade guiding adipogenesis, pinpointing GSK3β as a critical regulator for both EB5 and EB7 adipogenesis. In a developmental context, we observe that, akin to brown fat progenitors, brite fat progenitors make their appearance in murine development by 11-12 days of gestation or potentially earlier. This result contributes to our understanding of adipocyte lineage specification during embryonic development. In conclusion, EB5 and EB7 cell lines are valuable for research into adipocyte biology, providing insights into the differentiation and development of brown and beige adipocytes. Furthermore, they could be useful for the characterization of drugs targeting energy balance for the treatment of obesity and metabolic diseases.
Collapse
Affiliation(s)
- Cristina Velez‐delValle
- Department of Cell BiologyCenter for Research and Advanced Studies (Cinvestav)Mexico CityMexico
| | | | | | | | - Meytha Marsch‐Moreno
- Department of Cell BiologyCenter for Research and Advanced Studies (Cinvestav)Mexico CityMexico
| | - Walid Kuri‐Harcuch
- Department of Cell BiologyCenter for Research and Advanced Studies (Cinvestav)Mexico CityMexico
| |
Collapse
|