1
|
Wu L, Deng L, Xu X, Chang H, Liu C, Wu J, Zhang C, Wang R, Gao R, Chen H, Ye-Lehmann S, Zhang Z, Zhu T, Chen C. Astrocyte-derived extracellular vesicular NFIA mediates obesity-associated cognitive impairment. J Neuroinflammation 2025; 22:145. [PMID: 40448146 DOI: 10.1186/s12974-025-03473-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Accepted: 05/21/2025] [Indexed: 06/02/2025] Open
Abstract
BACKGROUND The escalating global prevalence of cognitive decline associated with obesity represents a significant public health challenge. Emerging evidence implicates astrocyte-derived extracellular vesicles (ADEVs) as key mediators in the pathogenesis of neurodegenerative disease, positioning them as potential therapeutic targets. However, the precise mechanistic role of ADEVs in the pathological processes underlying obesity-related cognitive impairment remains poorly understood. METHODS We established an obese mouse model by feeding mice a 60% high-fat diet (HFD) and assessed cognitive function through a series of behavioral tests. To investigate the role of extracellular vesicles (EVs), we inhibited EVs secretion by intraperitoneally administering GW4869, a neutral sphingomyelinase-2 (nSMase2) inhibitor, to 12-week HFD-fed male mice. Using comprehensive proteomic sequencing of brain-derived EVs, we identified NFIA as a potentially candidate protein. A series of in vivo and in vitro experiments were then conducted to confirmed the astrocytic origin of NFIA and neuronal uptake of ADEVs. Further, ADEVs isolated from primary cultured astrocytes under high glucose conditions were administered to both wild-type mice and primary cultured neurons to demonstrate their mediating role. Additionally, we developed adeno-associated virus (AAV) constructs to specifically knockdown the target gene Nfia of astrocyte to validate these findings. RESULTS Following 16 weeks of HFD feeding, obese mice exhibited significant cognitive impairment, which was significantly alleviated by GW4869 administration through inhibition of ceramide-dependent EVs secretion. Proteomic analysis revealed a marked upregulation of NFIA protein in brain-derived EVs from obese mice, with astrocytes identified as the predominant cellular origin. ADEVs containing NFIA has been found to specifically accumulated in the hippocampal neurons both in vivo and in vitro. As expected, ADEVs isolated from high glucose-treated primary astrocytes induced substantial cognitive decline in healthy adult mice and caused synaptic injury in primary cultured neurons. Of note, astrocyte-specific knockdown of the Nfia gene resulted in improved synaptic function and ameliorated cognitive impairment in obese mice. CONCLUSIONS These findings demonstrated that elevated levels of NFIA packaged within ADEVs contributed to hippocampal synaptic injury under obesity-induced stress condition. The mechanistic insight may provide potential therapeutic targets for addressing obesity-related cognitive decline.
Collapse
Affiliation(s)
- Lining Wu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Liyun Deng
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Xiaolin Xu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Haiqing Chang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Changliang Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Jiahui Wu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Changteng Zhang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Ruiqun Wang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Rui Gao
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Hai Chen
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Shixin Ye-Lehmann
- Diseases and Hormones of the Nervous System, Unité INSERM U1195, University of Paris-Scalay, Bicêtre Hosptial, Bât. Grégory Pincus, Le Kremlin-Bicêtre, France
| | - Zhi Zhang
- Department of Anesthesiology, The First Affiliated Hospital of University of Science and Technology of China, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P.R. China
- The Center for Advanced Interdisciplinary Science and Biomedicine, Institute of Health and Medicine, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P.R. China
| | - Tao Zhu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China.
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China.
| | - Chan Chen
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China.
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China.
| |
Collapse
|
2
|
Li M, Zhu Q, Yang H, Hu Y, Zhao L, Zhao Y. Identification of key genes regulating brown adipose tissue thermogenesis in goat kids ( Capra hircus) by using weighted gene co-expression network analysis. Front Vet Sci 2025; 12:1525437. [PMID: 40438410 PMCID: PMC12116553 DOI: 10.3389/fvets.2025.1525437] [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: 11/09/2024] [Accepted: 04/28/2025] [Indexed: 06/01/2025] Open
Abstract
Brown adipose tissue (BAT) is crucial for the maintenance of body temperature in newborn animals through non-shivering thermogenesis (NST). However, which kind key genes involved in the regulation of BAT thermogenesis and the internal regulation mechanism of heat production in goat BAT were still unclear. In this study, we analyzed the perirenal adipose tissue transcriptome of Dazu black goats from 0, 7, 14, 21 and 28 days after birth using weighted gene co-expression network analysis (WGCNA) to identify key genes involved in the thermogenesis of BAT. Genes were classified into 22 co-expression modules by WGCNA. The turquoise module exhibited high gene expression in D0, with generally lower expression in the later dates. This pattern is consistent with the rapid color, morphological, and thermogenic changes observed in perirenal adipose tissue shortly after birth. GO functional annotation revealed that the genes in the turquoise module were significantly enriched in the mitochondrion, mitochondrial protein-containing complex, cytoplasm, and mitochondrial inner membrane. KEGG pathway enrichment analysis indicated that these genes were predominantly enriched in the signaling pathways of oxidative phosphorylation, thermogenesis, and TCA cycle. By combining the gene co-expression network analysis of the turquoise module genes and the differentially expression genes (DEG) analysis, we identified 5 candidate key genes (ACO2, MRPS27, IMMT, MRPL12, and TUFM) involved in regulation of perirenal adipose tissue thermogenesis. This finding offer candidate genes that in the regulation of BAT thermogenesis and body temperature maintenance in goat kids.
Collapse
Affiliation(s)
| | | | | | | | - Le Zhao
- Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Yongju Zhao
- Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing, China
| |
Collapse
|
3
|
Gu Y, He W, Li W, Cai J, Wang Z, Li K, Qin G, Gu X, Lin X, Ma L, Xiao X, Hou Y, Luo T. Arctiin, a lignan compound, enhances adipose tissue browning and energy expenditure by activating the adenosine A 2A receptor. Mol Med 2025; 31:188. [PMID: 40369420 PMCID: PMC12079995 DOI: 10.1186/s10020-025-01249-8] [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: 02/08/2025] [Accepted: 05/06/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND The activation of brown adipose tissue (BAT) or the browning of white adipose tissue (WAT) represents a promising therapeutic strategy for obesity. Arctiin (ARC), a lignan compound known for its anti-inflammatory, anti-tumor, and hypoglycemic properties, has not been fully elucidated regarding its effects and mechanisms on obesity. METHODS In the present study, we established both high-fat diet-induced obese mouse models and mature adipocyte cultures to comprehensively investigate the therapeutic effects of ARC on obesity. Systemic energy metabolism and thermogenic capacity were assessed through metabolic cage monitoring and cold stimulation tests. Histopathological alterations in adipose tissues were examined using hematoxylin and eosin (H&E) staining, while key gene expression in adipocytes was determined by Western blotting (WB), immunohistochemistry, and immunofluorescence staining. To further elucidate the molecular mechanisms underlying ARC's anti-obesity effects, we employed an integrated approach combining network pharmacology analysis, molecular docking simulations, cellular thermal shift assay (CETSA), and WB to identify potential molecular targets and delineate the associated signaling pathways modulated by ARC treatment. RESULTS In diet-induced obese mice, ARC administration at doses of 20 and 60 mg/kg/day ameliorated metabolic dysfunction through enhanced WAT browning and increased energy expenditure. In C3H10T1/2-induced adipocytes, ARC upregulated the protein expression of uncoupling protein 1 (UCP1), peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), and other brown-specific marker genes, promoting mitochondrial function and browning of adipocytes. Mechanistically, our findings suggest that ARC may promote adipocyte browning via the A2AR-cyclic AMP (cAMP)-protein kinase A (PKA) signaling pathway. CONCLUSION In summary, ARC exerts protective effects against obesity by promoting the browning of white adipocytes and holds promise as a potentially beneficial therapeutic agent for the treatment of obesity.
Collapse
Affiliation(s)
- Yuanfeng Gu
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Wenjun He
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Wenxuan Li
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Jingshu Cai
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Zhuyun Wang
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Kemeng Li
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Guangcheng Qin
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
- Laboratory Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiaojie Gu
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Xiaojing Lin
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Li Ma
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Xiaoqiu Xiao
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China.
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China.
| | - Yi Hou
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China.
- Laboratory of Traditional Chinese Medicine, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing, 400016, China.
| | - Ting Luo
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
4
|
Kaur G, Lamb T, Tjitropranoto A, Rahman I. Single-cell transcriptomics identifies a dampened neutrophil function and accentuated T-cell cytotoxicity in tobacco flavored e-cigarette exposed mouse lungs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.17.638715. [PMID: 40027777 PMCID: PMC11870523 DOI: 10.1101/2025.02.17.638715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
E-cigarettes (e-cigs) are a public health concern for young adults due to their popularity and evidence for increased oxidative stress and immunotoxicity. Yet an extensive study defining the cell-specific immune changes upon exposure to flavored e-cigs remains elusive. To understand the immunological lung landscape upon acute nose-only exposure of C57BL/6J to flavored e-cig aerosols we performed single-cell RNA sequencing (scRNA seq). scRNA profiles of 71,725 cells were generated from control and treatment groups (n=2/sex/group). A distinct phenotype of Ly6G-neutrophils was identified in lungs exposed to tobacco flavored e-cig aerosol which demonstrated dampened IL-1 mediated and pattern recognition signaling as compared to air controls. Differential gene expression analyses identified dysregulation of T-cell mediated pro-inflammation ( Cct7 , Cct8 ) and stress-response signals ( Neurl3 , Stap1 , Cirbp and Htr2c) in the lungs of mice exposed to e-cig aerosols, with pronounced effects for tobacco flavor. Flow cytometry analyses and cytokine/chemokine assessments within the lungs corroborated the scRNA seq data, demonstrating a significant increase in T-cell percentages and levels of T-cell associated cytokine/chemokines in the lungs of tobacco-flavored aerosol exposed mice. Increased levels of Klra4 and Klra8 expression also suggest an enhanced natural killer (NK) cell activity in this mouse group. Overall, this is a pilot study identifying increase in the percentages of Ly6G-neutrophils that may be responsible for dampened innate immune responses and heightened T-cell cytotoxicity in lungs of tobacco-flavored e-cig aerosol exposed mice. In addition, we provide preliminary evidence for sex-specific changes in the transcriptional landscape of mouse lungs upon exposure to e-cig aerosol, an area that warrants further study.
Collapse
|
5
|
Yang Y, Wu J, Zhou W, Ji G, Dang Y. Protein posttranslational modifications in metabolic diseases: basic concepts and targeted therapies. MedComm (Beijing) 2024; 5:e752. [PMID: 39355507 PMCID: PMC11442990 DOI: 10.1002/mco2.752] [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: 05/25/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 10/03/2024] Open
Abstract
Metabolism-related diseases, including diabetes mellitus, obesity, hyperlipidemia, and nonalcoholic fatty liver disease, are becoming increasingly prevalent, thereby posing significant threats to human health and longevity. Proteins, as the primary mediators of biological activities, undergo various posttranslational modifications (PTMs), including phosphorylation, ubiquitination, acetylation, methylation, and SUMOylation, among others, which substantially diversify their functions. These modifications are crucial in the physiological and pathological processes associated with metabolic disorders. Despite advancements in the field, there remains a deficiency in contemporary summaries addressing how these modifications influence processes of metabolic disease. This review aims to systematically elucidate the mechanisms through which PTM of proteins impact the progression of metabolic diseases, including diabetes, obesity, hyperlipidemia, and nonalcoholic fatty liver disease. Additionally, the limitations of the current body of research are critically assessed. Leveraging PTMs of proteins provides novel insights and therapeutic targets for the prevention and treatment of metabolic disorders. Numerous drugs designed to target these modifications are currently in preclinical or clinical trials. This review also provides a comprehensive summary. By elucidating the intricate interplay between PTMs and metabolic pathways, this study advances understanding of the molecular mechanisms underlying metabolic dysfunction, thereby facilitating the development of more precise and effective disease management strategies.
Collapse
Affiliation(s)
- Yunuo Yang
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Jiaxuan Wu
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Wenjun Zhou
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Guang Ji
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Yanqi Dang
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| |
Collapse
|
6
|
Dada A, Habibi J, Naz H, Chen D, Lastra G, Bostick BP, Whaley-Connell A, Hill MA, Sowers JR, Jia G. Enhanced ECCD36 signaling promotes skeletal muscle insulin resistance in female mice. Am J Physiol Endocrinol Metab 2024; 327:E533-E543. [PMID: 39196801 PMCID: PMC11482271 DOI: 10.1152/ajpendo.00246.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/19/2024] [Accepted: 08/24/2024] [Indexed: 08/30/2024]
Abstract
Consumption of a Western diet (WD) increases CD36 expression in vascular, hepatic, and skeletal muscle tissues promoting lipid metabolic disorders and insulin resistance. We further examined the role of endothelial cell-specific CD36 (ECCD36) signaling in contributing to skeletal muscle lipid metabolic disorders, insulin resistance, and their underlying molecular mechanisms. Female ECCD36 wild-type (ECCD36+/+) and knock-out (ECCD36-/-) mice, aged 6 wk, were provided with either a WD or a standard chow diet for a duration of 16 wk. ECCD36+/+ WD mice were characterized by elevated fasting plasma glucose and insulin levels, increased homeostatic model assessment for insulin resistance, and glucose intolerance that was blunted in ECCD36-/- mice. Improved insulin sensitivity in ECCD36-/- mice was characterized by increased phosphoinositide 3-kinases/protein kinase B signaling that further augmented glucose transporter type 4 expression and glucose uptake. Meanwhile, 16 wk of WD feeding also increased skeletal muscle free fatty acid (FFA) and lipid accumulation, without any observed changes in plasma FFA levels. These lipid metabolic disorders were blunted in ECCD36-/- mice. Moreover, ECCD36 also mediated in vitro palmitic acid-induced lipid accumulation in cultured ECs, subsequently leading to the release of FFAs into the culture media. Furthermore, consumption of a WD increased FFA oxidation, mitochondrial dysfunction, impaired mitochondrial respiratory, skeletal muscle fiber type transition, and fibrosis. These WD-induced abnormalities were blunted in ECCD36-/- mice. These findings demonstrate that endothelial-specific ECCD36 signaling participates in skeletal muscle FFA uptake, ectopic lipid accumulation, mitochondrial dysfunction, insulin resistance, and associated skeletal muscle dysfunction in diet-induced obesity.NEW & NOTEWORTHY ECCD36 exerts "extra endothelial cell" actions in skeletal muscle insulin resistance. ECCD36 is a major mediator of Western diet-induced lipid metabolic disorders and insulin resistance in skeletal muscle. Mitochondrial dysfunction is associated with diet-induced CD36 activation and related skeletal muscle insulin resistance.
Collapse
Affiliation(s)
- Austin Dada
- Department of Medicine-Endocrinology and Metabolism, University of Missouri School of Medicine, Columbia, Missouri, United States
| | - Javad Habibi
- Department of Medicine-Endocrinology and Metabolism, University of Missouri School of Medicine, Columbia, Missouri, United States
- Research Service, Harry S Truman Memorial Veterans Hospital, Research Service, Columbia, Missouri, United States
| | - Huma Naz
- Department of Medicine-Endocrinology and Metabolism, University of Missouri School of Medicine, Columbia, Missouri, United States
- Research Service, Harry S Truman Memorial Veterans Hospital, Research Service, Columbia, Missouri, United States
| | - Dongqing Chen
- Department of Medicine-Endocrinology and Metabolism, University of Missouri School of Medicine, Columbia, Missouri, United States
- Research Service, Harry S Truman Memorial Veterans Hospital, Research Service, Columbia, Missouri, United States
| | - Guido Lastra
- Department of Medicine-Endocrinology and Metabolism, University of Missouri School of Medicine, Columbia, Missouri, United States
- Research Service, Harry S Truman Memorial Veterans Hospital, Research Service, Columbia, Missouri, United States
| | - Brian P Bostick
- Department of Medicine-Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, Missouri, United States
| | - Adam Whaley-Connell
- Department of Medicine-Endocrinology and Metabolism, University of Missouri School of Medicine, Columbia, Missouri, United States
- Research Service, Harry S Truman Memorial Veterans Hospital, Research Service, Columbia, Missouri, United States
- Department of Medicine-Nephrology and Hypertension, University of Missouri School of Medicine, Columbia, Missouri, United States
| | - Michael A Hill
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, United States
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
| | - James R Sowers
- Department of Medicine-Endocrinology and Metabolism, University of Missouri School of Medicine, Columbia, Missouri, United States
- Research Service, Harry S Truman Memorial Veterans Hospital, Research Service, Columbia, Missouri, United States
- Department of Medicine-Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, Missouri, United States
- Department of Medicine-Nephrology and Hypertension, University of Missouri School of Medicine, Columbia, Missouri, United States
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, United States
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
| | - Guanghong Jia
- Department of Medicine-Endocrinology and Metabolism, University of Missouri School of Medicine, Columbia, Missouri, United States
- Research Service, Harry S Truman Memorial Veterans Hospital, Research Service, Columbia, Missouri, United States
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
| |
Collapse
|
7
|
Mao L, Lu J, Hou Y, Nie T. Directly targeting PRDM16 in thermogenic adipose tissue to treat obesity and its related metabolic diseases. Front Endocrinol (Lausanne) 2024; 15:1458848. [PMID: 39351529 PMCID: PMC11439700 DOI: 10.3389/fendo.2024.1458848] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 08/28/2024] [Indexed: 10/04/2024] Open
Abstract
Obesity is increasing globally and is closely associated with a range of metabolic disorders, including metabolic associated fatty liver disease, diabetes, and cardiovascular diseases. An effective strategy to combat obesity involves stimulating brown and beige adipocyte thermogenesis, which significantly enhances energy expenditure. Recent research has underscored the vital role of PRDM16 in the development and functionality of thermogenic adipocytes. Consequently, PRDM16 has been identified as a potential therapeutic target for obesity and its related metabolic disorders. This review comprehensively examines various studies that focus on combating obesity by directly targeting PRDM16 in adipose tissue.
Collapse
Affiliation(s)
- Liufeng Mao
- The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jinli Lu
- The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yunliang Hou
- The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, China
| | - Tao Nie
- School of Basic Medicine, Hubei University of Arts and Science, Xiangyang, China
| |
Collapse
|
8
|
He C, An Y, Shi L, Huang Y, Zhang H, Fu W, Wang M, Shan Z, Du Y, Xie J, Huang Z, Sun W, Zhao Y, Zhao B. Xiasangju alleviate metabolic syndrome by enhancing noradrenaline biosynthesis and activating brown adipose tissue. Front Pharmacol 2024; 15:1371929. [PMID: 38576483 PMCID: PMC10993144 DOI: 10.3389/fphar.2024.1371929] [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: 01/17/2024] [Accepted: 03/06/2024] [Indexed: 04/06/2024] Open
Abstract
Metabolic syndrome (MetS) is a clinical condition associated with multiple metabolic risk factors leading to type 2 diabetes mellitus and other metabolic diseases. Recent evidence suggests that modulating adipose tissue to adaptive thermogenesis may offer therapeutic potential for MetS. Xiasangju (XSJ) is a marketed drug and dietary supplement used for the treatment of metabolic disease with anti-inflammatory activity. This study investigated the therapeutic effects of XSJ and the underlying mechanisms affecting the activation of brown adipose tissue (BAT) in MetS. The results revealed that XSJ ameliorated MetS by enhancing glucose and lipid metabolism, leading to reduced body weight and abdominal circumference, decreased adipose tissue and liver index, and improved blood glucose tolerance. XSJ administration stimulated catecholamine biosynthesis, increasing noradrenaline (NA) levels and activating NA-mediated proteins in BAT. Thus, BAT enhanced thermogenesis and oxidative phosphorylation (OXPHOS). Moreover, XSJ induced changes in gut microbiota composition, with an increase in Oscillibacter abundance and a decrease in Bilophila, Candidatus Stoquefichus, Holdemania, Parasutterella and Rothia. XSJ upregulated the proteins associated with intestinal tight junctions corresponding with lower serum lipopolysaccharide (LPS), tumor necrosis factor α (TNF-α) monocyte chemoattractant protein-1 (MCP-1) and interleukin-6 (IL-6) levels to maintain NA signaling transport. In summary, XSJ may alleviate MetS by promoting thermogenesis in BAT to ultimately boost energy metabolism through increasing NA biosynthesis, strengthening intestinal barrier integrity and reducing low-grade inflammation. These findings suggest XSJ has potential as a natural therapeutic agent for the treatment of MetS.
Collapse
Affiliation(s)
- Changhao He
- Department of Pharmacology of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yongcheng An
- Department of Pharmacology of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Lu Shi
- Department of Pharmacology of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
- Central Laboratories, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Yan Huang
- College of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Huilin Zhang
- Department of Pharmacology of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Wanxin Fu
- College of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Menglu Wang
- Department of Pharmacology of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Ziyi Shan
- College of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yuhang Du
- Department of Pharmacology of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jiamei Xie
- Department of Pharmacology of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhiyun Huang
- Guangzhou Baiyunshan Xingqun Pharmaceutical Co., Ltd, Guangzhou, China
| | - Weiguang Sun
- Guangzhou Baiyunshan Xingqun Pharmaceutical Co., Ltd, Guangzhou, China
| | - Yonghua Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, China
| | - Baosheng Zhao
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
9
|
Santa K, Kumazawa Y, Watanabe K, Nagaoka I. The Recommendation of the Mediterranean-styled Japanese Diet for Healthy Longevity. Endocr Metab Immune Disord Drug Targets 2024; 24:1794-1812. [PMID: 38343059 DOI: 10.2174/0118715303280097240130072031] [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: 08/31/2023] [Revised: 12/08/2023] [Accepted: 01/05/2024] [Indexed: 10/22/2024]
Abstract
The Mediterranean diet, listed as the intangible cultural heritage of humanity by UNESCO, is known as healthy and consumed worldwide. The Japanese diet is also listed and considered healthy. This narrative review compares the Mediterranean diet with its Japanese counterpart. Research has reported that people in Mediterranean regions, such as Italy and Greece, have one-third of the mortality ratio from cardiovascular diseases compared to people in the United States and Northern Europe because of the difference in eating habits. Therefore, Mediterranean diets are considered as healthy. A typical Western diet containing high amounts of fat, sugar, and calories is responsible for several diseases like metabolic syndrome and obesity, which are induced by chronic inflammation. In contrast, Mediterranean and Japanese diets contain them only less. The similarity between Mediterranean and Japanese diets is the substantial intake of vegetables, beans, and fish. On the other hand, the Mediterranean diet consumes large amounts of olive oil, especially polyphenol-rich extra virgin olive oil and dairy products, but meat consumption is relatively small. In contrast, the Japanese diet does not use oil and fat, contains abundant fermented foods, and consumes seaweed. Japan is known for its longevity, and people think that a well-balanced diet daily is good for preventing and curing illness. In this regard, finding non-disease conditions, so-called "ME-BYO," and curing them before the manifestation of diseases is becoming more common. In this review, we discuss the healthy eating habit, "The Mediterranean-styled Japanese diet," which prevents ME-BYO condition and reduces the risk of various diseases. The Mediterranean-styled Japanese diet, a hybrid of Mediterranean and Japanese diets, reduces the risk of various diseases by suppressing chronic inflammation. This nutritional intervention prevents ME-BYO and is beneficial for healthy longevity. Hence, a Mediterranean-styled Japanese diet might be helpful for healthy longevity in Japan and around the world.
Collapse
Affiliation(s)
- Kazuki Santa
- Department of Biotechnology, Tokyo College of Biotechnology, Ota-ku, Tokyo, Japan
| | - Yoshio Kumazawa
- Vino Science Japan, Inc., Kawasaki, Japan
- Department of Biochemistry and Systems Biomedicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Kenji Watanabe
- Center for Kampo Medicine, Keio University, Shinjuku-ku, Tokyo, Japan
- Yokohama University of Pharmacy, Yokohama, Japan
| | - Isao Nagaoka
- Department of Biochemistry and Systems Biomedicine, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
- Faculty of Medical Science, Juntendo University, Urayasu, Japan
| |
Collapse
|
10
|
Hiraike Y, Saito K, Oguchi M, Wada T, Toda G, Tsutsumi S, Bando K, Sagawa J, Nagano G, Ohno H, Kubota N, Kubota T, Aburatani H, Kadowaki T, Waki H, Yanagimoto S, Yamauchi T. NFIA in adipocytes reciprocally regulates mitochondrial and inflammatory gene program to improve glucose homeostasis. Proc Natl Acad Sci U S A 2023; 120:e2308750120. [PMID: 37487068 PMCID: PMC10401007 DOI: 10.1073/pnas.2308750120] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 06/20/2023] [Indexed: 07/26/2023] Open
Abstract
Adipose tissue is central to regulation of energy homeostasis. Adaptive thermogenesis, which relies on mitochondrial oxidative phosphorylation (Ox-Phos), dissipates energy to counteract obesity. On the other hand, chronic inflammation in adipose tissue is linked to type 2 diabetes and obesity. Here, we show that nuclear factor I-A (NFIA), a transcriptional regulator of brown and beige adipocytes, improves glucose homeostasis by upregulation of Ox-Phos and reciprocal downregulation of inflammation. Mice with transgenic expression of NFIA in adipocytes exhibited improved glucose tolerance and limited weight gain. NFIA up-regulates Ox-Phos and brown-fat-specific genes by enhancer activation that involves facilitated genomic binding of PPARγ. In contrast, NFIA in adipocytes, but not in macrophages, down-regulates proinflammatory cytokine genes to ameliorate adipose tissue inflammation. NFIA binds to regulatory region of the Ccl2 gene, which encodes proinflammatory cytokine MCP-1 (monocyte chemoattractant protein-1), to down-regulate its transcription. CCL2 expression was negatively correlated with NFIA expression in human adipose tissue. These results reveal the beneficial effect of NFIA on glucose and body weight homeostasis and also highlight previously unappreciated role of NFIA in suppressing adipose tissue inflammation.
Collapse
Affiliation(s)
- Yuta Hiraike
- Division for Health Service Promotion, The University of Tokyo, Tokyo113-0033, Japan
- The University of Tokyo Excellent Young Researcher Program, The University of Tokyo, Tokyo113-8654, Japan
| | - Kaede Saito
- Division for Health Service Promotion, The University of Tokyo, Tokyo113-0033, Japan
| | - Misato Oguchi
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo113-8655, Japan
| | - Takahito Wada
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo113-8655, Japan
| | - Gotaro Toda
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo113-8655, Japan
| | - Shuichi Tsutsumi
- Genome Science and Medicine Laboratory, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo153-8904, Japan
| | - Kana Bando
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe650-0047, Japan
| | - Junji Sagawa
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima734-8551, Japan
| | - Gaku Nagano
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima734-8551, Japan
| | - Haruya Ohno
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima734-8551, Japan
| | - Naoto Kubota
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo113-8655, Japan
- Department of Clinical Nutrition Therapy, The University of Tokyo Hospital, Tokyo113-8655, Japan
| | - Tetsuya Kubota
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo113-8655, Japan
- Division of Diabetes and Metabolism, The Institute of Medical Science, Asahi Life Foundation, Tokyo103-0002, Japan
- National Institutes of Biomedical Innovation, Health and Nutrition, Tokyo162-8636, Japan
| | - Hiroyuki Aburatani
- Genome Science and Medicine Laboratory, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo153-8904, Japan
| | | | - Hironori Waki
- Department of Diabetes and Endocrinology, Akita University Graduate School of Medicine, Akita010-8543, Japan
| | - Shintaro Yanagimoto
- Division for Health Service Promotion, The University of Tokyo, Tokyo113-0033, Japan
| | - Toshimasa Yamauchi
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo113-8655, Japan
| |
Collapse
|