1
|
Gao D, Bing C, Griffiths HR. Disrupted adipokine secretion and inflammatory responses in human adipocyte hypertrophy. Adipocyte 2025; 14:2485927. [PMID: 40176539 PMCID: PMC11980453 DOI: 10.1080/21623945.2025.2485927] [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: 07/02/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 04/04/2025] Open
Abstract
Adipocyte hypertrophy is a critical contributor to obesity-induced inflammation and insulin resistance. This study employed a human adipocyte hypertrophy model to investigate the adipokine release, inflammatory responses, and the intracellular singling pathways. Hypertrophic adipocytes exhibited increased lipid content and lipolysis, a decline of anti-inflammatory adipokine adiponectin release and an increase of pro-inflammatory adipokine leptin release compared to mature adipocytes. Moreover, TNFα and LPS exacerbated the decrease in adiponectin secretion by hypertrophic adipocytes while promoting the secretion of leptin, MCP-1 and IL-6, which is associated with impaired activation of p38 and JNK MAPK and persistent activation of ERK and IκBα in hypertrophic adipocytes. These altered adipokine secretions and inflammatory responses within hypertrophic adipocytes may contribute to adipocyte dysfunction in human obesity.
Collapse
Affiliation(s)
- Dan Gao
- Institute of Molecular and Translational Medicine, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Ministry of Education, Key Laboratory of Environment and Genes Related to Diseases Xi’an Jiaotong University, Xi’an, China
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Center, Xi’an, China
| | - Chen Bing
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | | |
Collapse
|
2
|
Agadagba SK, Yau SY, Liang Y, Dalton K, Thompson B. Bidirectional causality of physical exercise in retinal neuroprotection. Neural Regen Res 2025; 20:3400-3415. [PMID: 39688575 PMCID: PMC11974656 DOI: 10.4103/nrr.nrr-d-24-00942] [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: 08/16/2024] [Revised: 10/21/2024] [Accepted: 11/16/2024] [Indexed: 12/18/2024] Open
Abstract
Physical exercise is recognized as an effective intervention to improve mood, physical performance, and general well-being. It achieves these benefits through cellular and molecular mechanisms that promote the release of neuroprotective factors. Interestingly, reduced levels of physical exercise have been implicated in several central nervous system diseases, including ocular disorders. Emerging evidence has suggested that physical exercise levels are significantly lower in individuals with ocular diseases such as glaucoma, age-related macular degeneration, retinitis pigmentosa, and diabetic retinopathy. Physical exercise may have a neuroprotective effect on the retina. Therefore, the association between reduced physical exercise and ocular diseases may involve a bidirectional causal relationship whereby visual impairment leads to reduced physical exercise and decreased exercise exacerbates the development of ocular disease. In this review, we summarize the evidence linking physical exercise to eye disease and identify potential mediators of physical exercise-induced retinal neuroprotection. Finally, we discuss future directions for preclinical and clinical research in exercise and eye health.
Collapse
Affiliation(s)
- Stephen K. Agadagba
- Center for Eye and Vision Research Limited, 17W, Hong Kong Science Park, Hong Kong Special Administrative Region, China
| | - Suk-yu Yau
- Center for Eye and Vision Research Limited, 17W, Hong Kong Science Park, Hong Kong Special Administrative Region, China
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Ying Liang
- Center for Eye and Vision Research Limited, 17W, Hong Kong Science Park, Hong Kong Special Administrative Region, China
| | - Kristine Dalton
- Center for Eye and Vision Research Limited, 17W, Hong Kong Science Park, Hong Kong Special Administrative Region, China
- School of Optometry and Vision Science, University of Waterloo, Waterloo, ON, Canada
| | - Benjamin Thompson
- Center for Eye and Vision Research Limited, 17W, Hong Kong Science Park, Hong Kong Special Administrative Region, China
- School of Optometry and Vision Science, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
3
|
Huang W, Zhu W, Lin Y, Chan FKL, Xu Z, Ng SC. Roseburia hominis improves host metabolism in diet-induced obesity. Gut Microbes 2025; 17:2467193. [PMID: 39976263 PMCID: PMC11845086 DOI: 10.1080/19490976.2025.2467193] [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: 07/24/2024] [Revised: 02/05/2025] [Accepted: 02/10/2025] [Indexed: 02/21/2025] Open
Abstract
Next-generation live biotherapeutics are promising to aid the treatment of obesity and metabolic diseases. Here, we reported a novel anti-obesity probiotic candidate, Roseburia hominis, that was depleted in stool samples of obese subjects compared with lean controls, and its abundance was negatively correlated with body mass index and serum triglycerides. Supplementation of R. hominis prevented body weight gain and disorders of glucose and lipid metabolism, prevented fatty liver, inhibited white adipose tissue expansion and brown adipose tissue whitening in mice fed with high-fat diet, and boosted the abundance of lean-related species. The effects of R. hominis could be partially attributed to the production of nicotinamide riboside and upregulation of the Sirtuin1/mTOR signaling pathway. These results indicated that R. hominis is a promising candidate for the development of next-generation live biotherapeutics for the prevention of obesity and metabolic diseases.
Collapse
Affiliation(s)
- Wenli Huang
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Wenyi Zhu
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Yu Lin
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Francis K. L. Chan
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhilu Xu
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Siew C. Ng
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
4
|
Wang H, Qin Y, Niu J, Chen H, Lu X, Wang R, Han J. Evolving perspectives on evaluating obesity: from traditional methods to cutting-edge techniques. Ann Med 2025; 57:2472856. [PMID: 40077889 PMCID: PMC11912248 DOI: 10.1080/07853890.2025.2472856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
Objective: This review examines the evolution of obesity evaluation methods, from traditional anthropometric indices to advanced imaging techniques, focusing on their clinical utility, limitations, and potential for personalized assessment of visceral adiposity and associated metabolic risks. Methods: A comprehensive analysis of existing literature was conducted, encompassing anthropometric indices (BMI, WC, WHR, WHtR, NC), lipid-related metrics (LAP, VAI, CVAI, mBMI), and imaging technologies (3D scanning, BIA, ultrasound, DXA, CT, MRI). The study highlights the biological roles of white, brown, and beige adipocytes, emphasizing visceral adipose tissue (VAT) as a critical mediator of metabolic diseases. Conclusion: Although BMI and other anthropometric measurements are still included in the guidelines, indicators that incorporate lipid metabolism information can more accurately reflect the relationship between metabolic diseases and visceral obesity. At the same time, the use of more modern medical equipment, such as ultrasound, X-rays, and CT scans, allows for a more intuitive assessment of the extent of visceral obesity.
Collapse
Affiliation(s)
- Heyue Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yaxin Qin
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jinzhu Niu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Haowen Chen
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xinda Lu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Rui Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jianli Han
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
5
|
Zhou J, Guo Y, Liu X, Yuan W. Bioinformatics analysis identifies key secretory protein-encoding differentially expressed genes in adipose tissue of metabolic syndrome. Adipocyte 2025; 14:2446243. [PMID: 39819282 DOI: 10.1080/21623945.2024.2446243] [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: 08/14/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/19/2025] Open
Abstract
The objective of this study was to identify key secretory protein-encoding differentially expressed genes (SP-DEGs) in adipose tissue in female metabolic syndrome, thus detecting potential targets in treatment. We examined gene expression profiles in 8 women with metabolic syndrome and 7 healthy, normal body weight women. A total of 143 SP-DEGs were screened, including 83 upregulated genes and 60 downregulated genes. GO analyses of these SP-DEGs included proteolysis, angiogenesis, positive regulation of endothelial cell proliferation, immune response, protein processing, positive regulation of neuroblast proliferation, cell adhesion and ER to Golgi vesicle-mediated transport. KEGG pathway analysis of the SP-DEGs were involved in the TGF-beta signalling pathway, cytokine‒cytokine receptor interactions, the hippo signalling pathway, Malaria. Two modules were identified from the PPI network, namely, Module 1 (DNMT1, KDM1A, NCoR1, and E2F1) and Module 2 (IL-7 R, IL-12A, and CSF3). The gene DNMT1 was shared between the network modules and the WGCNA brown module. According to the single-gene GSEA results, DNMT1 was significantly positively correlated with histidine metabolism and phenylalanine metabolism. This study identified 7 key SP-DEGs in adipose tissue. DNMT1 was selected as the central gene in the development of metabolic syndrome and might be a potential therapeutic target.
Collapse
Affiliation(s)
- Jiandong Zhou
- Department of Nephrology, Shanghai General Hospital of Nanjing Medical University, Shanghai, China
| | - Yunshan Guo
- Department of Nephrology, Shanghai General Hospital of Nanjing Medical University, Shanghai, China
| | - Xuan Liu
- Department of Nephrology, Shanghai General Hospital of Nanjing Medical University, Shanghai, China
| | - Weijie Yuan
- Department of Nephrology, Shanghai General Hospital of Nanjing Medical University, Shanghai, China
| |
Collapse
|
6
|
Todorović N, Kuzmanovic J, Javorac D, Ostojic SM. Role of molecular hydrogen in obesity treatment: modulation of GLP-1, irisin, and PGC-1α for improved metabolism. Med Gas Res 2025; 15:442-443. [PMID: 40251025 PMCID: PMC12054667 DOI: 10.4103/mgr.medgasres-d-24-00146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/03/2025] [Accepted: 01/17/2025] [Indexed: 04/20/2025] Open
Affiliation(s)
- Nikola Todorović
- Applied Bioenergetics Lab, Faculty of Sport and PE, University of Novi Sad, Novi Sad, Serbia
| | - Jovan Kuzmanovic
- Applied Bioenergetics Lab, Faculty of Sport and PE, University of Novi Sad, Novi Sad, Serbia
| | - Dejan Javorac
- Applied Bioenergetics Lab, Faculty of Sport and PE, University of Novi Sad, Novi Sad, Serbia
| | - Sergej M. Ostojic
- Applied Bioenergetics Lab, Faculty of Sport and PE, University of Novi Sad, Novi Sad, Serbia
- Department of Nutrition and Public Health, University of Agder, Kristiansand, Norway
- Faculty of Health Sciences, University of Pécs, Pécs, Hungary
| |
Collapse
|
7
|
Wang T, Tang C, Xiao M, He M, Li Y, Li X. Characteristics of lipid accumulation induced by high-altitude environment improve the total antioxidant capacity of Ophiocordyceps sinensis. Food Chem 2025; 480:143812. [PMID: 40112714 DOI: 10.1016/j.foodchem.2025.143812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 02/26/2025] [Accepted: 03/07/2025] [Indexed: 03/22/2025]
Abstract
Ophiocordyceps sinensis is a traditional Chinese herbal medicine and nutritional supplement, which is rich in functional components beneficial to health. The biosynthesis of these components is affected by environment factors, especially lipids. This study analyzed the lipid mass spectrometry characteristics and total antioxidant capacity (TAC) of O. sinensis in 5 different altitude environments and explored the important contributions of environmental factors. The pathway of glycerophospholipid metabolism and biosynthesis of secondary metabolites in O. sinensis was activated by altitude (AM) and mean annual temperature (MAT) at high altitudes. This stimulated the degradation of triglycerides (TG) and the biosynthesis of phosphatidylcholine (PC), and phosphatidylethanolamine (PE), promoted the accumulation of free radical scavenging (FRS) abilities and antioxidant components (AC), and increased its TAC. This study reflects the important role of high altitude environment on lipid metabolism and the formation of bioactive components in O. sinensis and provides a scientific basis for exploring its medicinal value and nutritional value.
Collapse
Affiliation(s)
- Tao Wang
- State Key Laboratory of Plateau Ecology and Agriculture, College of Animal Husbandry and Veterinary Sciences, Qinghai University, Xining 810016, China
| | - Chuyu Tang
- State Key Laboratory of Plateau Ecology and Agriculture, College of Animal Husbandry and Veterinary Sciences, Qinghai University, Xining 810016, China
| | - Mengjun Xiao
- State Key Laboratory of Plateau Ecology and Agriculture, College of Animal Husbandry and Veterinary Sciences, Qinghai University, Xining 810016, China
| | - Min He
- State Key Laboratory of Plateau Ecology and Agriculture, College of Animal Husbandry and Veterinary Sciences, Qinghai University, Xining 810016, China
| | - Yuling Li
- State Key Laboratory of Plateau Ecology and Agriculture, College of Animal Husbandry and Veterinary Sciences, Qinghai University, Xining 810016, China
| | - Xiuzhang Li
- State Key Laboratory of Plateau Ecology and Agriculture, College of Animal Husbandry and Veterinary Sciences, Qinghai University, Xining 810016, China.
| |
Collapse
|
8
|
Ma J, Wang S, Zhang P, Zheng S, Li X, Li J, Pei H. Emerging roles for fatty acid oxidation in cancer. Genes Dis 2025; 12:101491. [PMID: 40290117 PMCID: PMC12022645 DOI: 10.1016/j.gendis.2024.101491] [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: 06/22/2024] [Accepted: 11/09/2024] [Indexed: 04/30/2025] Open
Abstract
Fatty acid oxidation (FAO) denotes the mitochondrial aerobic process responsible for breaking down fatty acids (FAs) into acetyl-CoA units. This process holds a central position in the cancer metabolic landscape, with certain tumor cells relying primarily on FAO for energy production. Over the past decade, mounting evidence has underscored the critical role of FAO in various cellular processes such as cell growth, epigenetic modifications, tissue-immune homeostasis, cell signal transduction, and more. FAO is tightly regulated by multiple evolutionarily conserved mechanisms, and any dysregulation can predispose to cancer development. In this view, we summarize recent findings to provide an updated understanding of the multifaceted roles of FAO in tumor development, metastasis, and the response to cancer therapy. Additionally, we explore the regulatory mechanisms of FAO, laying the groundwork for potential therapeutic interventions targeting FAO in cancers within the metabolic landscape.
Collapse
Affiliation(s)
- Jialin Ma
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Shuxian Wang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Pingfeng Zhang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Sihao Zheng
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Xiangpan Li
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Juanjuan Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Huadong Pei
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| |
Collapse
|
9
|
Mizunoe Y, Kumagai M, Fukai H, Hachiya K, Otani Y, Nozaki Y, Tezuka K, Kobayashi M, Haeno H, Saeki K, Murayama Y, Shimano H, Higami Y. Caloric restriction alters NCOA2 splicing to regulate lipid metabolism in subcutaneous white adipose tissue. Biochem Biophys Res Commun 2025; 765:151871. [PMID: 40267838 DOI: 10.1016/j.bbrc.2025.151871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 04/25/2025]
Abstract
Caloric restriction (CR) promotes longevity and metabolic health by modulating gene expression and cellular processes. However, the role of alternative mRNA splicing in CR-induced metabolic adaptation remains underexplored. In this study, we analyzed RNA sequencing data from the subcutaneous white adipose tissue of CR mice. We identified 6058 differentially expressed genes, with significant upregulation of lipid metabolism pathway genes, such as Elovl6, Fasn, and Srebp1c. We also detected 400 CR-associated alternative splicing events, with the skipped exon and retained intron events predominantly affecting lipid biosynthesis and energy metabolism. Among these events, Ncoa2, a nuclear receptor coactivator involved in lipid metabolism, exhibited increased exon 13 inclusion under CR, favoring the expression of the full-length isoform. Functional assays revealed that full-length NCOA2 enhanced PPARγ-mediated transcriptional activation, while the truncated Δ-NCOA2 isoform exhibited altered coactivator activity. Δ-NCOA2 was found to lack an LXXL motif critical for nuclear receptor interactions, potentially modifying its function. Taken together, these findings indicate that CR-induced alternative splicing fine-tunes metabolic and transcriptional networks, thereby contributing to lipid homeostasis and energy adaptation. Our study highlights a novel regulatory layer by which CR modulates metabolism through coordinated transcriptional and splicing alterations, offering new insights into the molecular mechanisms underlying the beneficial effects of CR on aging and metabolic health. Further investigations are warranted to determine the tissue-specificity of the CR-induced splicing changes and their potential implications for metabolic disorders and lifespan extension.
Collapse
Affiliation(s)
- Yuhei Mizunoe
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo, 125-8585, Japan.
| | - Mitsuki Kumagai
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo, 125-8585, Japan.
| | - Hiroto Fukai
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo, 125-8585, Japan.
| | - Kazuki Hachiya
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo, 125-8585, Japan.
| | - Yuina Otani
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo, 125-8585, Japan.
| | - Yuka Nozaki
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo, 125-8585, Japan.
| | - Kyo Tezuka
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo, 125-8585, Japan.
| | - Masaki Kobayashi
- Department of Nutrition and Food Science, Graduate School of Humanities and Sciences, Ochanomizu University, Tokyo, 112-8610, Japan; Institute for Human Life Science, Ochanomizu University, Bunkyo-ku, Tokyo, 112-8610, Japan.
| | - Hiroshi Haeno
- Research Institute for Biomedical Sciences (RIBS), Tokyo University of Science, Noda, Chiba, 278-0022, Japan.
| | - Koichi Saeki
- Research Institute for Biomedical Sciences (RIBS), Tokyo University of Science, Noda, Chiba, 278-0022, Japan.
| | - Yuki Murayama
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan.
| | - Hitoshi Shimano
- Department of Endocrinology and Metabolism, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, 305-8575, Japan.
| | - Yoshikazu Higami
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo, 125-8585, Japan; Division of Cell Fate Regulation, Research Institute for Biomedical Science (RIBS), Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo, 125-8585, Japan.
| |
Collapse
|
10
|
Binmahfouz LS, Al Otaibi A, Binmahfouz NS, Abdel-Naim AB, Eid BG, Shaik RA, Bagher AM. Luteolin modulates the TGFB1/PI3K/PTEN axis in hormone-induced uterine leiomyomas: Insights from a rat model. Eur J Pharmacol 2025; 996:177439. [PMID: 40043870 DOI: 10.1016/j.ejphar.2025.177439] [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: 09/19/2024] [Revised: 01/28/2025] [Accepted: 02/26/2025] [Indexed: 03/09/2025]
Abstract
Uterine leiomyomas (UL), or fibroids, are non-cancerous tumors of the uterine smooth muscle, affecting approximately 70% of women of reproductive age. They are the most prevalent solid tumors in the gynecological tract and a major indication for hysterectomy. The pathogenesis of UL involves uterine inflammation, uncontrolled cell division, and suppressed apoptosis. This study evaluated the protective effects of luteolin, a flavonoid known for its anti-inflammatory and antioxidant properties, against diethylstilbestrol and progesterone-induced UL in female rats. Twenty-four female Wistar rats were divided into four groups: (1) control, (2) luteolin (10 mg/kg, PO), (3) UL (diethylstilbestrol 1.35 mg/kg + progesterone 1 mg/kg, SC), and (4) UL + luteolin (10 mg/kg). The treatment duration was five weeks. Histological analyses were performed using hematoxylin and eosin (H&E) staining and Masson's Trichrome staining to evaluate uterine architecture and fibrosis. Histological results demonstrated normal uterine architecture in the control and luteolin groups, with marked neoplastic cell proliferation and fibrosis in the UL group, significantly mitigated by luteolin treatment. Luteolin reduced uterine weights and exhibited antioxidant, anti-inflammatory, pro-apoptotic, and anti-proliferative effects. Immunohistochemical analysis revealed that luteolin significantly reduced α-SMA protein expression, suggesting its role in modulating fibrotic pathways by inhibiting TGF-β1 and PI3K and enhancing PTEN production. These findings highlight luteolin's potential as a non-invasive therapeutic option for UL and suggest the need for further clinical studies to establish its efficacy, optimize dosage, and evaluate its safety profile in humans.
Collapse
Affiliation(s)
- Lenah S Binmahfouz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, 21589, Saudi Arabia.
| | - Abdullah Al Otaibi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, 21589, Saudi Arabia; Department of Pharmaceutical Care, Maternal and Children Specialist Hospital, Jeddah, 23816, Saudi Arabia
| | - Najlaa S Binmahfouz
- Department of Anatomical Histopathology, East Jeddah General Hospital, Jeddah, 22253, Saudi Arabia
| | - Ashraf B Abdel-Naim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Basma G Eid
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Rasheed A Shaik
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Amina M Bagher
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| |
Collapse
|
11
|
Fang L, Li W, Zhao H, Wang W, Gao H, Wang P, Zhang X, Lv R, Xu F, Chen J, Lyu L, Chen Y. Irisin alleviates steroid-induced vascular dysfunction by regulating the αVβ5-c-Abl-Caveolin-1 signaling pathway. Biochem Pharmacol 2025; 236:116870. [PMID: 40086515 DOI: 10.1016/j.bcp.2025.116870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 03/07/2025] [Accepted: 03/11/2025] [Indexed: 03/16/2025]
Abstract
Steroid-induced avascular necrosis of the femoral head (SANFH) is a progressive degenerative disease of the hip, primarily due to glucocorticoid (GC)-induced endothelial cell (EC) injury and compromised blood supply. Irisin is an EC-protective mytokine whose receptor is the integrin αVβ5. Caveolin-1 (CAV-1), a major component of caveolae, causes endothelial dysfunction when phosphorylated. However, the role of irisin and CAV-1 in SANFH remains unclear. In our study, irisin levels decreased but CAV-1 phosphorylation increased in human and mouse SANFH samples. Intraperitoneal irisin injection (250 μg/kg daily) notably reduced GC-induced osteonecrosis, vascular abnormalities, and CAV-1 phosphorylation in SANFH mice. In cultured ECs, GC induced CAV-1 phosphorylation by activating c-Abl via the glucocorticoid receptor, and irisin inhibited GC-induced phosphorylation of c-Abl and CAV-1 via the integrin αVβ5. Inhibition of integrin αVβ5 also abolished the protective effects of irisin on ERK and eNOS signalling, viability, angiogenesis, and migration in ECs. Therefore, our findings indicate that irisin has a protective role against vascular dysfunction in SANFH, possibly mediated by the inhibition of GC-triggered c-Abl-CAV-1 phosphorylation through integrin αVβ5. These findings provide insights into the potential therapeutic applications of irisin in SANFH.
Collapse
Affiliation(s)
- Lijun Fang
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Wenqiang Li
- Department of Emergency Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hua Zhao
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, China
| | - Wei Wang
- School of Public Health, Shandong University, Jinan, China
| | - Hongmei Gao
- Department of Cardiology, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Pengqi Wang
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xinzhi Zhang
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ruijuan Lv
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, China; Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Feng Xu
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, China; Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China.
| | - Jiazheng Chen
- Department of Orthopaedics, Peking University Third Hospital, Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China.
| | - Linmao Lyu
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, China; Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China.
| | - Yuguo Chen
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, China; Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China; Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
12
|
Yaligar J, Rengaraj A, Le GTT, Leow MKS, Eriksson JG, Ashokkumar B, Velan SS. Fatty Acylcarnitine Metabolism in Brown/Beige and White Fats by 13C HRMAS NMR Spectroscopy With Metabolic Interventions. NMR IN BIOMEDICINE 2025; 38:e70040. [PMID: 40230060 DOI: 10.1002/nbm.70040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/16/2025]
Abstract
White adipose tissue (WAT) and brown adipose tissue (BAT) have distinct structural and physiological characteristics and serve opposing functions in the body. WAT primarily stores energy, whereas BAT is metabolically active and positively influences metabolic health, contributing to energy expenditure, reduced fat accumulation and enhanced mitochondrial metabolism. Recently, both classical BAT and beige fat (or inducible/recruitable BAT) that arises from the browning of WAT have attracted clinical interest as potential targets for improving mitochondrial metabolism and managing obesity-related metabolic disorders. Currently, there is a lack of specific metabolic biomarkers for characterizing classical BAT and beige fat tissues, which are essential for evaluating mitochondrial oxidative metabolism and screening browning agents for therapeutic use. In this study, we investigated the potential metabolic biomarker fatty acylcarnitine in interscapular BAT (iBAT) from the interscapular region and beige adipose tissue from the inguinal region of the animal using ex vivo 13C high-resolution magic angle spinning (HRMAS) NMR spectroscopy. We examined how mitochondrial oxidative metabolism was altered in response to a high-fat diet (HFD) and how it was restored through iBAT activation using stimuli such as cold exposure and β3-adrenergic receptor (β3-AR) agonist, CL-316,243 treatment. We identified fatty acylcarnitine as a potential metabolic marker present in iBAT and beige tissues, whereas it was absent in iWAT. The concentration of fatty acylcarnitine significantly decreased in HFD-fed animals due to impaired lipolysis resulting in inefficient shuttling of fatty acids for mitochondrial β oxidation. However, its concentration increased or was restored in response to iBAT activation through either β3-AR agonist treatment or cold exposure, indicating a high-energy metabolic state with enhanced mitochondrial metabolism in iBAT. Fatty acylcarnitine shows promise as a biomarker for evaluating mitochondrial metabolism and for screening potential browning agents and nutraceuticals capable of inducing the browning of WAT.
Collapse
Affiliation(s)
- Jadegoud Yaligar
- Institute for Human Development and Potential, Agency for Science Technology and Research (A*STAR), Singapore
- Laboratory of Molecular Imaging, Institute of Bioengineering and Bioimaging, A*STAR, Singapore
- Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Anantharaj Rengaraj
- Institute for Human Development and Potential, Agency for Science Technology and Research (A*STAR), Singapore
- Laboratory of Molecular Imaging, Institute of Bioengineering and Bioimaging, A*STAR, Singapore
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, India
| | - Giang Thi Thu Le
- Institute for Human Development and Potential, Agency for Science Technology and Research (A*STAR), Singapore
- Laboratory of Molecular Imaging, Institute of Bioengineering and Bioimaging, A*STAR, Singapore
| | - Melvin Khee-Shing Leow
- Institute for Human Development and Potential, Agency for Science Technology and Research (A*STAR), Singapore
- Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
- Cardiovascular and Metabolic Diseases Program, Duke-NUS Medical School, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Division of Medicine, Department of Endocrinology, Tan Tock Seng Hospital, Singapore
| | - Johan G Eriksson
- Institute for Human Development and Potential, Agency for Science Technology and Research (A*STAR), Singapore
- Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | - S Sendhil Velan
- Institute for Human Development and Potential, Agency for Science Technology and Research (A*STAR), Singapore
- Laboratory of Molecular Imaging, Institute of Bioengineering and Bioimaging, A*STAR, Singapore
- Human Magnetic Resonance Centre, Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| |
Collapse
|
13
|
Qi G, Tang H, Hu J, Kang S, Qin S. Potential role of tanycyte-derived neurogenesis in Alzheimer's disease. Neural Regen Res 2025; 20:1599-1612. [PMID: 38934388 PMCID: PMC11688558 DOI: 10.4103/nrr.nrr-d-23-01865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/19/2024] [Accepted: 04/17/2024] [Indexed: 06/28/2024] Open
Abstract
Tanycytes, specialized ependymal cells located in the hypothalamus, play a crucial role in the generation of new neurons that contribute to the neural circuits responsible for regulating the systemic energy balance. The precise coordination of the gene networks controlling neurogenesis in naive and mature tanycytes is essential for maintaining homeostasis in adulthood. However, our understanding of the molecular mechanisms and signaling pathways that govern the proliferation and differentiation of tanycytes into neurons remains limited. This article aims to review the recent advancements in research into the mechanisms and functions of tanycyte-derived neurogenesis. Studies employing lineage-tracing techniques have revealed that the neurogenesis specifically originating from tanycytes in the hypothalamus has a compensatory role in neuronal loss and helps maintain energy homeostasis during metabolic diseases. Intriguingly, metabolic disorders are considered early biomarkers of Alzheimer's disease. Furthermore, the neurogenic potential of tanycytes and the state of newborn neurons derived from tanycytes heavily depend on the maintenance of mild microenvironments, which may be disrupted in Alzheimer's disease due to the impaired blood-brain barrier function. However, the specific alterations and regulatory mechanisms governing tanycyte-derived neurogenesis in Alzheimer's disease remain unclear. Accumulating evidence suggests that tanycyte-derived neurogenesis might be impaired in Alzheimer's disease, exacerbating neurodegeneration. Confirming this hypothesis, however, poses a challenge because of the lack of long-term tracing and nucleus-specific analyses of newborn neurons in the hypothalamus of patients with Alzheimer's disease. Further research into the molecular mechanisms underlying tanycyte-derived neurogenesis holds promise for identifying small molecules capable of restoring tanycyte proliferation in neurodegenerative diseases. This line of investigation could provide valuable insights into potential therapeutic strategies for Alzheimer's disease and related conditions.
Collapse
Affiliation(s)
- Guibo Qi
- Department of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Han Tang
- Department of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jianian Hu
- Department of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Siying Kang
- Department of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Song Qin
- Department of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
14
|
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
|
15
|
Nozu T, Miyagishi S, Ishioh M, Takakusaki K, Okumura T. Irisin prevents visceral hypersensitivity and colonic hyperpermeability in a rat model of irritable bowel syndrome. Peptides 2025; 188:171394. [PMID: 40154794 DOI: 10.1016/j.peptides.2025.171394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/03/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
Visceral hypersensitivity and impaired gut barrier function, accompanied by minor inflammation, are crucial components of the pathophysiology of irritable bowel syndrome (IBS). Research has demonstrated that corticotropin-releasing factor (CRF) and toll-like receptor 4 (TLR4) signaling mutually activate to produce proinflammatory cytokines, which modulate these gastrointestinal changes. Irisin, a myokine, has been shown to inhibit TLR4-proinflammatory cytokine signaling, thereby improving inflammation driven by obesity and metabolic syndrome. Based on this, we hypothesized that irisin could improve visceral hypersensitivity and impaired gut barrier function induced by lipopolysaccharide (LPS) or CRF (IBS rat models), and tested this hypothesis. The visceral pain threshold, triggered by colonic balloon distention, was assessed by electrophysiologically monitoring abdominal muscle contractions in male Sprague-Dawley rats. Colonic permeability was evaluated by measuring the amount of Evans blue dye absorbed within the colonic tissue. Intraperitoneal irisin prevented LPS-induced visceral hypersensitivity and colonic hyperpermeability in a dose-dependent manner. Irisin also prevented CRF-induced gastrointestinal alterations. The beneficial effects of irisin in the LPS model were reversed by compound C, an AMP-activated protein kinase (AMPK) inhibitor; NG-nitro-L-arginine methyl ester, a nitric oxide (NO) synthesis inhibitor; sulpiride or domperidone, a dopamine D2 receptor antagonist; atropine and intracisternal injection of SB-334867, a selective orexin 1 receptor antagonist. Overall, these findings suggest that irisin improves visceral sensation and colonic barrier function through AMPK, NO and dopamine D2, cholinergic and brain orexin signaling in IBS model. Thus, irisin may be a promising therapeutic agent for treating IBS.
Collapse
Affiliation(s)
- Tsukasa Nozu
- Department of Regional Medicine and Education, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan; Center for Medical Education, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan; Department of General Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan.
| | - Saori Miyagishi
- Department of General Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan.
| | - Masatomo Ishioh
- Department of General Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan.
| | - Kaoru Takakusaki
- Division of Neuroscience, Department of Physiology, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan.
| | - Toshikatsu Okumura
- Department of General Medicine, Asahikawa Medical University, 2-1-1-1 Midorigaoka-Higashi, Asahikawa, Hokkaido 078-8510, Japan.
| |
Collapse
|
16
|
Kim E, Tanzi RE, Choi SH. Therapeutic potential of exercise-hormone irisin in Alzheimer's disease. Neural Regen Res 2025; 20:1555-1564. [PMID: 38993140 PMCID: PMC11688551 DOI: 10.4103/nrr.nrr-d-24-00098] [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: 01/24/2024] [Revised: 05/20/2024] [Accepted: 06/04/2024] [Indexed: 07/13/2024] Open
Abstract
Irisin is a myokine that is generated by cleavage of the membrane protein fibronectin type III domain-containing protein 5 (FNDC5) in response to physical exercise. Studies reveal that irisin/FNDC5 has neuroprotective functions against Alzheimer's disease, the most common form of dementia in the elderly, by improving cognitive function and reducing amyloid-β and tau pathologies as well as neuroinflammation in cell culture or animal models of Alzheimer's disease. Although current and ongoing studies on irisin/FNDC5 show promising results, further mechanistic studies are required to clarify its potential as a meaningful therapeutic target for alleviating Alzheimer's disease. We recently found that irisin treatment reduces amyloid-β pathology by increasing the activity/levels of amyloid-β-degrading enzyme neprilysin secreted from astrocytes. Herein, we present an overview of irisin/FNDC5's protective roles and mechanisms against Alzheimer's disease.
Collapse
Affiliation(s)
- Eunhee Kim
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
| | - Rudolph E. Tanzi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
| | - Se Hoon Choi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
17
|
Zhang Y, Gao X, Liu C, Yang Q, Huang X, Li Y, Gun S. Proteomics reveals genetic mechanisms of cold resistance in Hezuo pig liver tissue. J Proteomics 2025; 316:105420. [PMID: 40064419 DOI: 10.1016/j.jprot.2025.105420] [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: 11/21/2024] [Revised: 03/01/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025]
Abstract
Cold stress poses a significant challenge to pig farming in northern China, leading to reduced productivity and, in severe cases, even mortality. However, the mechanisms underlying cold resistance in pigs are not well understood. To explore the genetic mechanism of cold resistance in pigs under low-temperature conditions, the cold-tolerant Hezuo pig was selected as a model. DIA proteomics analysis was performed on liver tissues from Hezuo pigs after 24 h of exposure to low-temperature treatments. The results showed that approximately 149 differential abundance proteins (DAPs) were detected (95 up-regulated and 54 down-regulated). GO analysis showed that these DAPs were mainly associated with lipid metabolism, vesicle fusion, and membrane function. KEGG analysis showed that these DAPs were primarily enriched in lipid metabolism-related pathways such as cholesterol metabolism and vitamin digestion and absorption. Comprehensive analysis identified APOA4, APOA2, SREBF2, ATP23, STX2, USO1, ETFA, RAB11FIP1, ETNPPL, and SGMS1 as potential key proteins involved in cold resistance mechanisms. The mRNA expression of the genes for two key candidate proteins (APOA4 and SREBF2), which are involved in lipid metabolism, was analyzed using qRT-PCR, revealing a significant up-regulation after low-temperature treatment. These findings provide significant insights into the mechanisms of cold resistance in animals and may serve as candidate markers for further studies on cold tolerance. SIGNIFICANCE: Cold resistance is one of the key traits in pigs and involves multiple complex coordinated regulatory mechanisms. However, its genetic mechanisms are not completely understood. In this study, a DIA proteomics approach was used to identify proteins and pathways associated with cold resistance in the liver of low-temperature-treated Hezuo pigs. These findings offer novel candidate proteins and key pathways for investigating the molecular mechanisms of cold resistance in Hezuo pigs, providing a base for further elucidating the mechanisms of cold tolerance in pigs.
Collapse
Affiliation(s)
- Yali Zhang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Xiaoli Gao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Chao Liu
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Qiaoli Yang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Xiaoyu Huang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Yajuan Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Shuangbao Gun
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; Gansu Innovations Center for Swine Production Engineering and Technology, Lanzhou 730070, China.
| |
Collapse
|
18
|
Singh A, Chaudhary R. Potentials of peroxisome proliferator-activated receptor (PPAR) α, β/δ, and γ: An in-depth and comprehensive review of their molecular mechanisms, cellular Signalling, immune responses and therapeutic implications in multiple diseases. Int Immunopharmacol 2025; 155:114616. [PMID: 40222274 DOI: 10.1016/j.intimp.2025.114616] [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: 01/22/2025] [Revised: 03/21/2025] [Accepted: 04/01/2025] [Indexed: 04/15/2025]
Abstract
Peroxisome proliferator-activated receptors (PPARs), ligand-activated transcription factors, have emerged as a key regulator of various biological processes, underscoring their relevance in the pathophysiology and treatment of numerous diseases. PPARs are primarily recognized for their critical role in lipid and glucose metabolism, which underpins their therapeutic applications in managing type 2 diabetes mellitus. Beyond metabolic disorders, they have gained attention for their involvement in immune modulation, making them potential targets for autoimmune-related inflammatory diseases. Furthermore, PPAR's ability to regulate proliferation, differentiation, and apoptosis has positioned them as promising candidates in oncology. Their anti-inflammatory and anti-fibrotic properties further highlight their potential in dermatological and cardiovascular conditions, where dysregulated inflammatory responses contribute to disease progression. Recent advancements have elucidated the molecular mechanisms of different PPAR isoforms, including their regulation of key signalling pathways such as NF-κB and MAPK, which are crucial in inflammation and cellular stress responses. Additionally, their interactions with co-factors and post-translational modifications further diversify their functional roles. The therapeutic potential of various PPAR agonists has been extensively explored, although challenges related to side effects and target specificity remain. This growing body of evidence underscores the significance of PPARs in understanding the molecular basis of diseases and advancing therapeutic interventions, paving way for targeted treatment approach across a wide spectrum of medical conditions. Here, we provide a comprehensive and detailed perspective of PPARs and their potential across different health conditions to advance our understanding, elucidate underlying mechanisms, and facilitate the development of potential treatment strategies.
Collapse
Affiliation(s)
- Alpana Singh
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Rishabh Chaudhary
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India.
| |
Collapse
|
19
|
Santana-Oliveira DA, Souza-Tavares H, Fernandes-da-Silva A, Marinho TS, Silva-Veiga FM, Daleprane JB, Souza-Mello V. Obesity prevention by different exercise protocols (HIIT or MICT) involves beige adipocyte recruitment and improved mitochondrial dynamics in high-fat-fed mice. Mol Cell Endocrinol 2025; 602:112533. [PMID: 40157711 DOI: 10.1016/j.mce.2025.112533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
AIM This study evaluated the effects of high-intensity interval training (HIIT) and moderate-intensity continuous training (MICT) on UCP1-dependent and UCP1-independent thermogenic and mitochondrial dynamics markers in the inguinal sWAT of high-fat-fed mice. METHODS Sixty male C57BL/6 mice (3 months old) were divided into six experimental groups: control diet (C), C + HIIT (C-HIIT), C + MICT (C-MICT), high-fat diet (HF), HF + HIIT (HF-HIIT) and HF + MICT (HF-MICT). The diet and exercise protocols started simultaneously and lasted ten weeks. RESULTS HIIT and MICT prevented body mass gain and fat pad expansion, improved insulin sensitivity, and induced browning in C-fed and HF-fed animals. Chronic intake of a HF diet caused adipocyte hypertrophy with a proinflammatory adipokine profile and impaired the expression of thermogenic and mitochondrial dynamics markers. However, both exercise intensities increased anti-inflammatory adipokine concentrations and improved gene markers of mitochondrial dynamics, resulting in sustained UCP1-dependent and UCP1-independent thermogenic markers and maintenance of the beige phenotype in inguinal sWAT. The principal component analysis placed all trained groups opposite the HF group and near the C group, ensuring the effectiveness of HIIT and MICT to prevent metabolic alterations. CONCLUSIONS This study provides reliable evidence that, regardless of intensity, exercise is a strategy to prevent obesity by reducing body fat accumulation and inducing browning. The anti-inflammatory adipokine profile and the increased expression of UCP1-dependent and UCP1-independent thermogenic markers sustained active beige adipocytes and mitochondrial enhancement to halt metabolic disturbances due to HF-feeding in exercised mice.
Collapse
Affiliation(s)
- Daiana Araujo Santana-Oliveira
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology. Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Henrique Souza-Tavares
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology. Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Aline Fernandes-da-Silva
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology. Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Thatiany Souza Marinho
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology. Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Flavia Maria Silva-Veiga
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology. Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Julio Beltrame Daleprane
- Laboratory for Studies of Interactions Between Nutrition and Genetics (LEING), Institute of Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Vanessa Souza-Mello
- Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Biomedical Center, Institute of Biology. Rio de Janeiro State University, Rio de Janeiro, Brazil.
| |
Collapse
|
20
|
Constant B, Kamzolas I, Yang X, Guo J, Rodriguez-Fdez S, Mali I, Rodriguez-Cuenca S, Petsalaki E, Vidal-Puig A, Li W. Distinct signalling dynamics of BMP4 and BMP9 in brown versus white adipocytes. Sci Rep 2025; 15:15971. [PMID: 40335635 DOI: 10.1038/s41598-025-99122-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 04/17/2025] [Indexed: 05/09/2025] Open
Abstract
Adipocyte dysfunction contributes to lipotoxicity and cardiometabolic diseases. Bone morphogenetic protein 4 (BMP4) is expressed in white adipocytes and remodels white adipose tissue, while liver-derived BMP9, a key circulating BMP, influences adipocyte lipid metabolism. The gene sets regulated by BMP4 and BMP9 signalling in mature adipocytes remain unclear. Here, we directly compare BMP4 and BMP9 signalling in mature brown and white adipocytes. While both BMPs showed comparable potency across adipocyte types, RNA sequencing analysis revealed extensive gene regulation, with many more differentially expressed genes and suppression of critical metabolic pathways in white adipocytes. Although BMP4 and BMP9 induced inhibitors of BMP and GDF signalling in both adipocytes, they selectively upregulated several TGF-β family receptors and BMP4 expression only in white adipocytes. These findings underscore a central role of BMP signalling in adipocyte homeostasis and suggest both BMP4 and BMP9 as regulators of white adipocyte plasticity with potential therapeutic implications.
Collapse
Affiliation(s)
- Benjamin Constant
- Department of Medicine, VPD Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
| | - Ioannis Kamzolas
- MRC Institute of Metabolic Science, MRC Metabolic Diseases Unit, Addenbrooke's Hospital, University of Cambridge, Box 289, Cambridge, CB2 0QQ, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Xudong Yang
- Department of Medicine, VPD Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
| | - Jingxu Guo
- Department of Medicine, VPD Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
| | - Sonia Rodriguez-Fdez
- Department of Medicine, VPD Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
- MRC Institute of Metabolic Science, MRC Metabolic Diseases Unit, Addenbrooke's Hospital, University of Cambridge, Box 289, Cambridge, CB2 0QQ, UK
| | - Iman Mali
- Department of Medicine, VPD Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK
- MRC Institute of Metabolic Science, MRC Metabolic Diseases Unit, Addenbrooke's Hospital, University of Cambridge, Box 289, Cambridge, CB2 0QQ, UK
| | - Sergio Rodriguez-Cuenca
- MRC Institute of Metabolic Science, MRC Metabolic Diseases Unit, Addenbrooke's Hospital, University of Cambridge, Box 289, Cambridge, CB2 0QQ, UK
| | - Evangelia Petsalaki
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Antonio Vidal-Puig
- Department of Medicine, VPD Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK.
- MRC Institute of Metabolic Science, MRC Metabolic Diseases Unit, Addenbrooke's Hospital, University of Cambridge, Box 289, Cambridge, CB2 0QQ, UK.
- CIBERDEN, Centro de Investigacion Principe Felipe, Valencia, Spain.
| | - Wei Li
- Department of Medicine, VPD Heart and Lung Research Institute, School of Clinical Medicine, University of Cambridge, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK.
| |
Collapse
|
21
|
Renu K. Understanding myokine Irisin's function in oral cancer: therapeutic and prognostic consequences. Expert Rev Anticancer Ther 2025. [PMID: 40338275 DOI: 10.1080/14737140.2025.2504471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 05/06/2025] [Accepted: 05/07/2025] [Indexed: 05/09/2025]
Affiliation(s)
- Kaviyarasi Renu
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| |
Collapse
|
22
|
Pena Calderin E, Zheng JJ, Boyd NL, Lynch W, Sansbury B, Spite M, Hill BG, Hellmann J. Exercise-Stimulated Resolvin Biosynthesis in the Adipose Tissue Is Abrogated by High-Fat Diet-Induced Adrenergic Deficiency. Arterioscler Thromb Vasc Biol 2025. [PMID: 40336478 DOI: 10.1161/atvbaha.124.322234] [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: 11/25/2024] [Accepted: 04/24/2025] [Indexed: 05/09/2025]
Abstract
BACKGROUND Diet-induced white adipose tissue inflammation is associated with insulin resistance and metabolic perturbations. Conversely, exercise protects against the development of diet-induced chronic inflammation and insulin resistance independent of weight loss; however, the mechanisms remain largely unknown. We have recently shown that through adrenergic stimulation of macrophages, exercise promotes resolution of acute peritoneal inflammation by enhancing the biosynthesis of specialized proresolving lipid mediators. In this study, we sought to determine whether exercise stimulates proresolving pathways in adipose tissue and whether this response is modified by diet. Specifically, we hypothesized that exercise stimulates proresolving pathways by adrenergic signaling, which is inhibited by high-fat diet, priming the development of chronic inflammation in the adipose tissue. METHODS To explore the dietary dependence of the proresolving effects of exercise, mice were fed either a control or high-fat diet for 2 weeks before, and throughout, a 4-week period of daily treadmill running. Glucose handling, body weight and composition, lipemia, and exercise performance were evaluated at the end of the feeding and exercise interventions. Likewise, changes in catecholamines and their biosynthetic enzymes were measured along with adipose tissue specialized proresolving lipid mediator levels and macrophage phenotype and abundance. RESULTS When compared with sedentary controls, macrophages isolated from mice exposed to 4 weeks of exercise display elevated expression of the specialized proresolving lipid mediator biosynthetic enzyme Alox15, while adipose tissue specialized proresolving lipid mediator levels and anti-inflammatory CD301+ M2 macrophages increased. These changes were dependent upon diet as 6 weeks of feeding with high-fat diet abrogated the proresolving effect of exercise when compared with control diet-fed animals. Interestingly, exercise-induced epinephrine production was inhibited by high-fat diet, which diminished the expression of the epinephrine biosynthetic enzyme PNMT (phenylethanolamine N-methyltransferase) in adrenal glands. CONCLUSIONS Taken together, these results suggest that a diet high in fat diminishes the proresolving effects of exercise in the adipose tissue via decreasing the biosynthesis of catecholamines.
Collapse
Affiliation(s)
- Ernesto Pena Calderin
- Division of Environmental Medicine, Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, University of Louisville School of Medicine, KY. (E.P.C., J.-J.Z., N.L.B., W.L., B.S., B.G.H., J.H.)
- Department of Physiology, University of Louisville School of Medicine, KY. (E.P.C.)
| | - Jing-Juan Zheng
- Division of Environmental Medicine, Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, University of Louisville School of Medicine, KY. (E.P.C., J.-J.Z., N.L.B., W.L., B.S., B.G.H., J.H.)
| | - Nolan L Boyd
- Division of Environmental Medicine, Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, University of Louisville School of Medicine, KY. (E.P.C., J.-J.Z., N.L.B., W.L., B.S., B.G.H., J.H.)
| | - Will Lynch
- Division of Environmental Medicine, Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, University of Louisville School of Medicine, KY. (E.P.C., J.-J.Z., N.L.B., W.L., B.S., B.G.H., J.H.)
| | - Brian Sansbury
- Division of Environmental Medicine, Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, University of Louisville School of Medicine, KY. (E.P.C., J.-J.Z., N.L.B., W.L., B.S., B.G.H., J.H.)
| | - Matthew Spite
- Department of Anesthesiology, Perioperative and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (M.S.)
| | - Bradford G Hill
- Division of Environmental Medicine, Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, University of Louisville School of Medicine, KY. (E.P.C., J.-J.Z., N.L.B., W.L., B.S., B.G.H., J.H.)
| | - Jason Hellmann
- Division of Environmental Medicine, Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, University of Louisville School of Medicine, KY. (E.P.C., J.-J.Z., N.L.B., W.L., B.S., B.G.H., J.H.)
| |
Collapse
|
23
|
Wu J, Tang J, Huang D, Wang Y, Zhou E, Ru Q, Xu G, Chen L, Wu Y. Study on the comorbid mechanisms of sarcopenia and late-life depression. Behav Brain Res 2025; 485:115538. [PMID: 40122287 DOI: 10.1016/j.bbr.2025.115538] [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: 10/15/2024] [Revised: 03/06/2025] [Accepted: 03/09/2025] [Indexed: 03/25/2025]
Abstract
The increasing global aging population has brought greater focus to age-related diseases, particularly muscle-brain comorbidities such as sarcopenia and late-life depression. Sarcopenia, defined by the gradual loss of muscle mass and function, is notably prevalent among older individuals, while late-life depression profoundly affects their mental health and overall well-being. Epidemiological evidence suggests a high co-occurrence of these two conditions, although the precise biological mechanisms linking them remain inadequately understood. This review synthesizes the existing body of literature on sarcopenia and late-life depression, examining their definitions, prevalence, clinical presentations, and available treatments. The goal is to clarify the potential connections between these comorbidities and offer a theoretical framework for the development of future preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Jiale Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Jun Tang
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Di Huang
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Yu Wang
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Enyuan Zhou
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Guodong Xu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China.
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China.
| |
Collapse
|
24
|
Xu J, Wang H, Han B, Zhang X. Mechanisms through which laparoscopic sleeve gastrectomy mitigates atherosclerosis risk: a focus on visceral adipose tissue. Eur J Med Res 2025; 30:370. [PMID: 40336107 DOI: 10.1186/s40001-025-02635-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 04/23/2025] [Indexed: 05/09/2025] Open
Abstract
Bariatric surgery is currently considered the key treatment method for patients with obesity and related complications. Among the various surgeries, laparoscopic sleeve gastrectomy (LSG) is the most widely used. Obesity is a multifactor chronic disease characterized by the accumulation of visceral adipose tissue (VAT), leading to susceptibility to cardiac metabolic diseases. Many mechanisms, including abnormal lipid metabolism, insulin resistance, inflammation, endothelial dysfunction, adipocytokine imbalance and inflammasome activation, have been identified as the basis for the relationship between obesity and atherosclerosis. Bariatric surgery, such as LSG, reduces the risk of atherosclerosis in people living with obesity by reducing energy intake, disrupting energy balance and reducing the secretion of intestinal hormones to intervene in these risk factors. This review explores the current understanding of how LSG affects VAT and its impact on the risk of atherosclerosis.
Collapse
Affiliation(s)
- Juan Xu
- General Surgery Department, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, 030032, China.
| | - Heyue Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Bin Han
- Shanxi Provincial People's Hospital, Taiyuan, 030032, China
| | - Xiaomin Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| |
Collapse
|
25
|
Zhang C, Wang B, Zheng J, Zhang Y, Han P, Ge Z, An Y, Wen C, Yuan Y, Jin J, Wang X, Wu J, Li X. Nuclear IL-1α Triggers Pyroptosis in Porcine Intramuscular Preadipocytes. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:10799-10813. [PMID: 40275457 DOI: 10.1021/acs.jafc.5c00112] [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: 04/26/2025]
Abstract
Pigs are vital for meat production, and their intramuscular fat content significantly impacts pork quality and flavor. Using single-cell RNA sequencing (scRNA-seq) on porcine intramuscular fat, we found that adipose progenitor cells express highly inflammatory genes, including IL-1α, during adipogenic differentiation. IL-1α, a pro-inflammatory cytokine present in various cell types, can translocate to the nucleus via its nuclear localization sequence (NLS), yet its nuclear function remains unclear. By constructing an inactivated NLS carrier for IL-1α, we discovered that nuclear IL-1α promotes pyroptosis in porcine intramuscular adipocytes through the IRF2-NOCT-ROS pathway. This study reveals a novel role of nuclear IL-1α in pyroptosis regulation, providing insights into enhancing the intramuscular fat content and improving pork quality.
Collapse
Affiliation(s)
- Chen Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Shaanxi 712100, People's Republic of China
| | - Bin Wang
- Department of Thoracic Surgery, First Medical Center of PLA General Hospital. Haidian District, Beijing 100000, People's Republic of China
| | - Juan Zheng
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Shaanxi 712100, People's Republic of China
| | - Yining Zhang
- College of Veterinary Medicine, Northwest A&F University, Shaanxi 712100, People's Republic of China
| | - Peiyuan Han
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Shaanxi 712100, People's Republic of China
| | - Zihao Ge
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Shaanxi 712100, People's Republic of China
| | - Yalong An
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Shaanxi 712100, People's Republic of China
| | - Chenglong Wen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Shaanxi 712100, People's Republic of China
| | - Yexian Yuan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Shaanxi 712100, People's Republic of China
| | - Jianjun Jin
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Shaanxi 712100, People's Republic of China
| | - Xinglong Wang
- College of Veterinary Medicine, Northwest A&F University, Shaanxi 712100, People's Republic of China
| | - Jiangwei Wu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Shaanxi 712100, People's Republic of China
| | - Xiao Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Shaanxi 712100, People's Republic of China
| |
Collapse
|
26
|
Naderi J, Johnson AK, Thakkar H, Chandravanshi B, Ksiazek A, Anand A, Vincent V, Tran A, Kalimireddy A, Singh P, Sood A, Das A, Talbot CL, Distefano IA, Maschek JA, Cox J, Li Y, Summers SA, Atkinson DJ, Turapov T, Ratcliff JA, Fung J, Shabbir A, Shabeer Yassin M, Shiow SATE, Holland WL, Pitt GS, Chaurasia B. Ceramide-induced FGF13 impairs systemic metabolic health. Cell Metab 2025; 37:1206-1222.e8. [PMID: 40169001 DOI: 10.1016/j.cmet.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 12/02/2024] [Accepted: 03/05/2025] [Indexed: 04/03/2025]
Abstract
Ceramide accumulation impairs adipocytes' ability to efficiently store and utilize nutrients, leading to energy and glucose homeostasis deterioration. Using a comparative transcriptomic screen, we identified the non-canonical, non-secreted fibroblast growth factor FGF13 as a ceramide-regulated factor that impairs adipocyte function. Obesity robustly induces FGF13 expression in adipose tissue in mice and humans and is positively associated with glycemic indices of type 2 diabetes. Pharmacological or genetic inhibition of ceramide biosynthesis reduces FGF13 expression. Using mice with loss and gain of function of FGF13, we demonstrate that FGF13 is both necessary and sufficient to impair energy and glucose homeostasis independent of ceramides. Mechanistically, FGF13 exerts these effects by inhibiting mitochondrial content and function, metabolic elasticity, and caveolae formation, which cumulatively impairs glucose utilization and thermogenesis. These studies suggest the therapeutic potential of targeting FGF13 to prevent and treat metabolic diseases.
Collapse
Affiliation(s)
- Jamal Naderi
- Division of Endocrinology, Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
| | - Amanda Kelsey Johnson
- Division of Endocrinology, Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
| | - Himani Thakkar
- Division of Endocrinology, Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
| | - Bhawna Chandravanshi
- Division of Endocrinology, Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
| | - Alec Ksiazek
- Division of Endocrinology, Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
| | - Ajay Anand
- Division of Endocrinology, Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
| | - Vinnyfred Vincent
- Division of Endocrinology, Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
| | - Aaron Tran
- Division of Endocrinology, Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
| | - Anish Kalimireddy
- Division of Endocrinology, Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
| | - Pratibha Singh
- Division of Endocrinology, Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
| | - Ayushi Sood
- Division of Endocrinology, Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
| | - Aasthika Das
- Division of Endocrinology, Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
| | - Chad Lamar Talbot
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT 84112, USA
| | - Isabella A Distefano
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - J Alan Maschek
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT 84112, USA
| | - James Cox
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84112, USA
| | - Ying Li
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT 84112, USA
| | - Scott A Summers
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84112, USA; Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT 84112, USA
| | - Donald J Atkinson
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT 84112, USA
| | - Tursun Turapov
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT 84112, USA
| | - Jason A Ratcliff
- Iowa Institute of Human Genetics, University of Iowa, Iowa City, IA 52242, USA
| | - Javis Fung
- Department of Surgery, University Surgical Cluster, National University Health System, Singapore 119228, Singapore
| | - Asim Shabbir
- Department of Surgery, University Surgical Cluster, National University Health System, Singapore 119228, Singapore
| | - M Shabeer Yassin
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Sue-Anne Toh Ee Shiow
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - William L Holland
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84112, USA; Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT 84112, USA
| | - Geoffrey S Pitt
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Bhagirath Chaurasia
- Division of Endocrinology, Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
27
|
Bolcato V, Poloni TE, Basile G, Davin A, Ferrari RR, Negro G, Ceretti A, Guaita A, Tronconi LP. Brain donation rules in Italy and worldwide: overview of a cutting-edge topic for human brain research. Neurol Sci 2025:10.1007/s10072-025-08214-7. [PMID: 40327175 DOI: 10.1007/s10072-025-08214-7] [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: 04/05/2025] [Accepted: 04/24/2025] [Indexed: 05/07/2025]
Abstract
Neuropathological examination of the brain and its biochemical analyses are fundamental to neuroscience studies and public health decisions, but are dependent on the effectiveness of regulations and operational protocols. The article discusses opportunities and limits of Italian regulation on body donation in relation to the specific requirements of neuropathology and brain sciences, in comparison with the regulations of other countries. Some crucial issues emerge, widely shared in the various regulatory contexts. The main aspect is the willingness to donate, consciously expressed by the subject by signing an informed consent or through the formulation of advanced directives. The donation of a single organ, the brain in particular, does not necessarily imply the donation of the entire body, which should be considered separately. In the specific case of the brain, particular attention is given to reducing the post-mortem interval, in order to obtain tissues suitable for research. Consequently, the centres that deal with the brain and brain banking must have experience and expertise in handling nervous tissue, and do not necessarily have to deal with the management of the entire body. These aspects, still little addressed in Italy, are the basis to develop an effective brain banking activity, which can only develop by integrating post-mortem body donation with specific rules for brain banking without which Italian neuroscience will be penalised in the coming years.
Collapse
Affiliation(s)
- Vittorio Bolcato
- Astolfi Associates Legal Firm, Milan & Maria Beatrice Hospital, GVM Care and Research, Florence, Italy
| | - Tino Emanuele Poloni
- Department of Neurology and Neuropathology, Golgi Cenci Foundation, Abbiategrasso, MI, Italy.
- Department of Rehabilitation, ASP Golgi Redaelli, Abbiategrasso, MI, Italy.
| | - Giuseppe Basile
- Department of Biomedical Sciences and Public Health, University "Politecnica Delle Marche" of Ancona, Ancona, AN, Italy
| | - Annalisa Davin
- Golgi Cenci Foundation, Laboratory of Neurobiology and Neurogenetics, Abbiategrasso, MI, Italy
| | - Riccardo Rocco Ferrari
- Golgi Cenci Foundation, Laboratory of Neurobiology and Neurogenetics, Abbiategrasso, MI, Italy
| | - Giulia Negro
- Department of Neurology and Neuropathology, Golgi Cenci Foundation, Abbiategrasso, MI, Italy
- Department of Rehabilitation, ASP Golgi Redaelli, Abbiategrasso, MI, Italy
| | - Arcangelo Ceretti
- Department of Neurology and Neuropathology, Golgi Cenci Foundation, Abbiategrasso, MI, Italy
- Department of Rehabilitation, ASP Golgi Redaelli, Abbiategrasso, MI, Italy
| | - Antonio Guaita
- Golgi Cenci Foundation, Laboratory of Neurobiology and Neurogenetics, Abbiategrasso, MI, Italy
| | - Livio Pietro Tronconi
- Department of Life and Human Sciences, European University of Rome, Rome, Italy
- Scientific Directorate, GVM Care and Research, Maria Cecilia Hospital, Cotignola, RA, Italy
| |
Collapse
|
28
|
Oliveri C, Xourafa A, Morabito N, Di Giovanni A, Lupo E, Basile G, Gaudio A, Catalano A. Calf circumference predicts changes of bone mineral density in postmenopausal osteoporotic women receiving denosumab. Aging Clin Exp Res 2025; 37:141. [PMID: 40323522 PMCID: PMC12053332 DOI: 10.1007/s40520-025-02989-7] [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: 01/13/2025] [Accepted: 02/19/2025] [Indexed: 05/08/2025]
Abstract
BACKGROUND Aging is associated with deterioration of muscle and bone health, resulting in increased fragility fracture risk. It is not known whether muscle mass and strength could impact the osteoporosis pharmacological response. AIM The aim of this study was to analyze the association between muscle mass and strength with the response to denosumab in osteoporosis. METHODS Postmenopausal women at high fracture risk receiving denosumab (60 mg subcutaneously administered every 6 months) were considered. The likelihood of sarcopenia was estimated by administering the SARC-F questionnaire, muscle mass and performance were assessed by measuring calf circumference (CC) and hand grip strength, respectively. Bone mineral density (BMD) was measured by dual energy X-ray absorptiometry. RESULTS 130 women (age 70.2 ± 9.4 years) were recruited. Baseline BMD T-score values were - 2.6 ± 1.1 SD and - 2.3 ± 0.7 SD at lumbar spine and femoral neck, respectively; while CC and grip strength were 31.9 ± 2.9 cm and 22.7 ± 6.7 kg, respectively. The SARC-F score was associated with the 10-year probability of major osteoporotic fracture (r = 0.21, p < 0.05). The CC was positively associated with the T-score values of both lumbar spine (r = 0.262, p = 0.034) and femoral neck (r = 0.359, p = 0.004). Denosumab administration (treatment duration 43 months), lead to BMD improvement by + 9.6% at the lumbar spine and + 7.3% at the femoral neck (pall < 0.05). After adjustment for comorbidities, fracture risk and treatment duration, the CC (β = 1.76, SE = 0.82, p = 0.03) and the baseline femoral BMD (β = - 94.19, SE = 26.09, p = 0.0009) were independently associated with femoral BMD gain over time. CONCLUSION In postmenopausal osteoporotic women, the CC was positively and independently associated with denosumab treatment response.
Collapse
Affiliation(s)
- Cecilia Oliveri
- Unit and School of Geriatrics, Department of Clinical and Experimental Medicine, University Hospital of Messina, Via C. Valeria, 98125, Messina, Italy.
| | | | - Nunziata Morabito
- Unit and School of Geriatrics, Department of Clinical and Experimental Medicine, University Hospital of Messina, Via C. Valeria, 98125, Messina, Italy
| | - Adele Di Giovanni
- Unit and School of Geriatrics, Department of Clinical and Experimental Medicine, University Hospital of Messina, Via C. Valeria, 98125, Messina, Italy
| | - Elisa Lupo
- Unit and School of Geriatrics, Department of Clinical and Experimental Medicine, University Hospital of Messina, Via C. Valeria, 98125, Messina, Italy
| | - Giorgio Basile
- Unit and School of Geriatrics, Department of Clinical and Experimental Medicine, University Hospital of Messina, Via C. Valeria, 98125, Messina, Italy
| | - Agostino Gaudio
- Department of Clinical and Experimental Medicine, University Hospital of Catania, Catania, Italy.
| | - Antonino Catalano
- Unit and School of Geriatrics, Department of Clinical and Experimental Medicine, University Hospital of Messina, Via C. Valeria, 98125, Messina, Italy.
| |
Collapse
|
29
|
Yilmaz FÇ, Türkoğlu S, Erman F. Are breast milk and serum irisin levels affected by the BMI and nutritional status? A prospective observational study. Eur J Pediatr 2025; 184:326. [PMID: 40325267 PMCID: PMC12053133 DOI: 10.1007/s00431-025-06154-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 04/19/2025] [Accepted: 04/24/2025] [Indexed: 05/07/2025]
Abstract
The influence of maternal nutritional status and anthropometric measurements on breast milk and serum irisin levels remains unclear. This study is the first to explore this relationship. This study aims to investigate the association between maternal BMI and nutritional status in the first month postpartum and their impact on breast milk and serum irisin levels. Forty-five mothers and their infants participated. Anthropometric measurements were taken at one month postpartum, maternal dietary intake was recorded over three days, and breast milk and serum irisin levels were analyzed. Overweight and obese mothers had lower breast milk irisin levels but higher serum irisin levels. A positive correlation was observed between breast milk irisin levels and infant birth weight. Additionally, serum irisin levels were positively associated with infant weight and height at one month. Maternal fiber intake was positively correlated with breast milk irisin levels, whereas fat intake showed a negative correlation. Moreover, higher folate, B12, and zinc intake were linked to increased breast milk and serum irisin levels. CONCLUSION Maternal BMI and nutritional status significantly influence breast milk and serum irisin levels. Promoting healthy eating habits and maintaining an optimal body weight before and after pregnancy may enhance irisin levels, potentially supporting infant growth and metabolic health. WHAT IS KNOWN • Irisin is a myokine involved in energy metabolism and insulin sensitivity, and breast milk composition is influenced by maternal BMI and nutritional status. • The effect of maternal obesity on breast milk irisin levels remains insufficiently understood. WHAT IS NEW • This is the first study to evaluate the association between maternal BMI, nutritional status, and both breast milk and serum irisin levels. • Overweight and obese mothers exhibit lower breast milk irisin but higher serum irisin levels, and maternal intake of fiber, fat, folate, vitamin B12, and zinc is significantly associated with irisin levels.
Collapse
Affiliation(s)
- Feray Çağiran Yilmaz
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Dicle University, Diyarbakır, Turkey.
- Department of Dietetic, Faculty of Health Sciences, Dicle University, Diyarbakır, Turkey.
| | - Semra Türkoğlu
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Firat University, Elazig, Turkey
| | - Fazilet Erman
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Firat University, Elazig, Turkey
| |
Collapse
|
30
|
Xu H, Shi J, Yu W, Sun S, Zhou H, Wang L, Ren J, Gu Z, Lu Q, Zhang Y. TBC1D15 protects alcohol-induced liver injury in female mice through PLIN5-mediated mitochondrial and lipid droplet contacting. Metabolism 2025:156290. [PMID: 40334909 DOI: 10.1016/j.metabol.2025.156290] [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: 11/13/2024] [Revised: 04/06/2025] [Accepted: 05/02/2025] [Indexed: 05/09/2025]
Abstract
OBJECTIVE Alcohol-induced hepatic steatosis and mitochondrial dysfunction are progressive conditions contributing to the development of alcoholic liver disease (ALD), often leading to cirrhosis and hepatocellular carcinoma. TBC1D15, a Rab7 GTPase-activating protein (GAP), has been implicated in mitochondrial homeostasis, however, its role in ALD remains elusive. This study aimed to investigate the functional role of TBC1D15 in ALD and elucidate the underlying mechanisms. METHODS Female TBC1D15flox/flox mice and hepatocyte-specific overexpression of TBC1D15 mice were fed a Lieber-DeCarli ethanol diet, which progressively increasing ethanol dosages over 8 weeks. Liver tissues were assessed using histology, transmission electron microscopy, immunofluorescence, immunoblotting, and real-time PCR techniques. RESULTS TBC1D15 levels were markedly decreased in human ALD samples and primary hepatocytes exposed to ethanol. Hepatocyte-specific TBC1D15 overexpression attenuated alcohol-induced body weight loss, improved survival, and alleviated liver injury, lipid droplet (LD) accumulation, and hepatocyte apoptosis. TBC1D15 overexpression also protected against alcohol-induced mitochondrial dysfunction and enhanced mitochondrial fatty acid β-oxidation (FAO) by promoting interactions between mitochondria and LDs in the face of alcohol exposure. Mechanistically, TBC1D15 was translocated to mitochondrial membranes in hepatocytes in response to alcohol exposure, where it recruited PLIN5 through its 10-180 aa domain. This interaction promoted mitochondria-LD contacts and facilitated PKA-induced nuclear translocation of PLIN5. Furthermore, TBC1D15 upregulated protein levels of PPARα, PGC1α and CPT1α in hepatocytes following alcohol challenge, an effect that was nullified by PKA inhibition. CONCLUSION TBC1D15 plays a promising protective role in ALD injury by enhancing mitochondrial function and FAO, potentially through its interaction with PLIN5 and modulation of mitochondria-LD contacts via PKA-mediated nuclear translocation of PLIN5. These findings identify TBC1D15 as a potential therapeutic target for ALD.
Collapse
Affiliation(s)
- Haixia Xu
- Department of Cardiology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China; Department of Cardiology, Zhongshan Hospital, Fudan University; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Jiayu Shi
- Department of Cardiology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Wenjun Yu
- Department of Cardiology, Zhongshan Hospital, Fudan University; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Shiqun Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Haoxiong Zhou
- Department of Gastroenterology, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China
| | - Lu Wang
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China; State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Air Force Medical University, Xi'an 710032, China
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital, Fudan University; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China.
| | - Zhifeng Gu
- Department of Cardiology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
| | - Qi Lu
- Department of Cardiology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
| | - Yingmei Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University; Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China.
| |
Collapse
|
31
|
Zhang K, Du Y, Yang S, Sun G. Irisin suppressed the progression of TBI via modulating AMPK/MerTK/autophagy and SYK/ROS/inflammatory signaling. Sci Rep 2025; 15:15583. [PMID: 40320408 PMCID: PMC12050266 DOI: 10.1038/s41598-025-00066-7] [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: 08/28/2024] [Accepted: 04/24/2025] [Indexed: 05/08/2025] Open
Abstract
Irisin is a hormone-like peptide secreted by muscle tissues and generated by hydrolysis of type III fibronectin domain-containing protein 5 by proteolytic hydrolases. Whether Irisin has a potential protective role in traumatic brain injury (TBI). In this study, we will investigate the relevant research progress of Irisin's protective role in traumatic brain injury (TBI) in recent years in terms of attenuating oxidative stress, inhibiting pyroptosis, suppressing inflammatory response, and improving autophagy, with the aim of providing valuable references for the diagnosis and treatment of traumatic brain injury (TBI). Utilize bioinformatics analysis to study the interactions between genes in TBI (Traumatic Brain Injury). Construct a TBI mouse model to observe the effects of Irisin on TBI. The Morris water maze test is used to assess the learning and spatial memory abilities of mice, TUNEL fluorescence is used to detect cell apoptosis, Nissl staining is employed to observe the survival of hippocampal neurons in mice, and HE staining is used to observe the extent of brain injury in mice. Western blot is used to detect protein expression in both in vivo and in vitro experiments. Q-PCR is employed to detect the levels of proteins related to autophagy/pyroptosis/inflammation. Irisin promotes MerTK overexpression by enhancing AMPK activation. Irisin can increase the expression of LC3I and Beclin-1 proteins, indicating the promotion of autophagic response. Additionally, Irisin reduces ROS levels and decreases SYK expression, thereby inhibiting the inflammatory response. Irisin improves the learning and spatial memory abilities of TBI mice and reduces cell apoptosis, as well as decreases hippocampal neuron death. HE staining shows that the brain injury in mice treated with Irisin is significantly alleviated. Irisin can enhance the expression of phosphorylated AMPK and phosphorylated MerTK proteins, promote autophagic response, and inhibit pyroptosis/inflammatory response. Correction experiments confirmed that after stimulation with an AMPK agonist, the expression of phosphorylated MerTK protein is significantly increased, autophagic response is enhanced, and pyroptosis/inflammatory response is weakened. When treated with a MerTK inhibitor during AMPK agonist stimulation, the autophagic response is weakened while pyroptosis/inflammatory response is enhanced. Irisin can inhibit the progression of traumatic brain injury by regulating AMPK/MerTK/autophagy and SYK/ROS/inflammatory signaling.
Collapse
Affiliation(s)
- Kuo Zhang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang City, 050000, Hebei Province, China
- Department of Neurosurgery, Hebei General Hospital, Shijiazhuang City, 050000, Hebei Province, China
| | - Yihui Du
- Department of Neurosurgery, Hebei General Hospital, Shijiazhuang City, 050000, Hebei Province, China
| | - Sihui Yang
- Department of Neurosurgery, Hebei General Hospital, Shijiazhuang City, 050000, Hebei Province, China
| | - Guozhu Sun
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang City, 050000, Hebei Province, China.
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, 215 Heping West Road, Xinhua District, Shijiazhuang City, 050000, Hebei Province, China.
| |
Collapse
|
32
|
Liu S, Cao H, Wang Z, Zhu J, An X, Zhang L, Song Y. Single-cell transcriptomics reveals extracellular matrix remodeling and collagen dynamics during lactation in sheep mammary gland. Int J Biol Macromol 2025:143669. [PMID: 40319976 DOI: 10.1016/j.ijbiomac.2025.143669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 04/13/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
The mammary gland is a dynamic organ with diverse cell populations that maintain glandular homeostasis, particularly during lactation. However, the cellular architecture and molecular mechanisms underlying lactational remodeling in the sheep mammary gland remain incompletely understood. Given similarities in mammary stromal structure, sheep serve as a valuable model for studying lactational changes relevant to the human breast, which experiences collagen loss and sagging during lactation. Utilizing single-cell transcriptomics (scRNA-seq), we mapped the sheep mammary gland's cellular landscape at postpartum days 60 and 150, identifying seven major cell types, including six distinct epithelial clusters. These clusters revealed differentiation among luminal progenitors, hormone-sensing, and myoepithelial cells across peak and late lactation stages. Transcriptomic analysis highlighted pivotal roles for epithelial integrity and ECM remodeling, with myoepithelial cells centrally involved in these processes. We observed significant collagen remodeling driven by fibroblast-epithelial crosstalk and ECM reorganization during late lactation. Comparative analysis with human mammary epithelial cells showed conserved basal and myoepithelial cell populations, while luminal cells diverged across species. This study provides insights into lactation biology and ECM remodeling, offering a framework to inform future studies on lactational adaptation and its implications for human health.
Collapse
Affiliation(s)
- Shujuan Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Heran Cao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Zhanhang Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Junru Zhu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Xiaopeng An
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| | - Lei Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| | - Yuxuan Song
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| |
Collapse
|
33
|
Castro-Rodríguez LI, Velez-delValle C, Hernández-Mosqueira CP, Kuri-Harcuch W. Spot-14 and its paralog Spot-14R regulate expression of metabolic and thermogenic pathway genes in murine brown and beige adipocytes. FEBS Lett 2025. [PMID: 40317955 DOI: 10.1002/1873-3468.70052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 01/24/2025] [Accepted: 03/07/2025] [Indexed: 05/07/2025]
Abstract
Spot 14 (S14), encoded by Thrsp, is a thyroid hormone-responsive transcriptional activator that regulates lipogenesis, though its mechanisms remain unclear. We aimed to study the role of S14 on gene expression in adipocytes. We analyzed Thrsp and its paralog Mid1ip1 in brown (EB5), beige (EB7), and white (F442A) adipocytes. Thrsp expression was higher in EB5 and EB7 than in F442A and increased with thyroid hormone T3 in EB5 and EB7 but decreased in F442A. Mid1ip1 expression rose moderately in EB5 and EB7, influencing lipid metabolism genes. Silencing Thrsp upregulated Mid1ip1 in EB7 and reduced thermogenic gene expression in EB5 and EB7. These findings underscore the roles of Thrsp and Mid1ip1 in metabolic and thermogenic pathways, highlighting the responsiveness of S14 to thyroid hormones and nutrient signals. Impact statement This study reveals that Thyroid Hormone-Induced Protein 8 (THRSP), also known as Spot-14, and its paralog Spot-14R, regulate metabolic and thermogenic gene expression differently in brown and beige adipocytes. These findings provide insights into adipocyte metabolism, offering potential targets for obesity and metabolic disorder treatments.
Collapse
Affiliation(s)
| | - Cristina Velez-delValle
- Department of Cell Biology, Center for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | | | - Walid Kuri-Harcuch
- Department of Cell Biology, Center for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| |
Collapse
|
34
|
Machado IF, Palmeira CM, Rolo AP. Sestrin2 is a central regulator of mitochondrial stress responses in disease and aging. Ageing Res Rev 2025; 109:102762. [PMID: 40320152 DOI: 10.1016/j.arr.2025.102762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 04/09/2025] [Accepted: 04/30/2025] [Indexed: 05/08/2025]
Abstract
Mitochondria supply most of the energy for cellular functions and coordinate numerous cellular pathways. Their dynamic nature allows them to adjust to stress and cellular metabolic demands, thus ensuring the preservation of cellular homeostasis. Loss of normal mitochondrial function compromises cell survival and has been implicated in the development of many diseases and in aging. Although exposure to continuous or severe stress has adverse effects on cells, mild mitochondrial stress enhances mitochondrial function and potentially extends health span through mitochondrial adaptive responses. Over the past few decades, sestrin2 (SESN2) has emerged as a pivotal regulator of stress responses. For instance, SESN2 responds to genotoxic, oxidative, and metabolic stress, promoting cellular defense against stress-associated damage. Here, we focus on recent findings that establish SESN2 as an orchestrator of mitochondrial stress adaptation, which is supported by its involvement in the integrated stress response, mitochondrial biogenesis, and mitophagy. Additionally, we discuss the integral role of SESN2 in mediating the health benefits of exercise as well as its impact on skeletal muscle, liver and heart injury, and aging.
Collapse
Affiliation(s)
- Ivo F Machado
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CiBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Institute of Interdisciplinary Research, Doctoral Program in Experimental Biology and Biomedicine (PDBEB), University of Coimbra, Coimbra, Portugal
| | - Carlos M Palmeira
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CiBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Anabela P Rolo
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CiBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Department of Life Sciences, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
35
|
Chung E, Zhang D, Gonzalez Porras M, Hsu CG. TREM2 as a regulator of obesity-induced cardiac remodeling: mechanisms and therapeutic insights. Am J Physiol Heart Circ Physiol 2025; 328:H1073-H1082. [PMID: 40152357 DOI: 10.1152/ajpheart.00075.2025] [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: 01/29/2025] [Revised: 02/17/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
Obesity and type 2 diabetes mellitus (T2DM) are global health challenges that significantly increase the risk of cardiovascular diseases (CVD). Advances in immunometabolism have identified triggering receptor expressed on myeloid cells 2 (TREM2) as a key regulator of macrophage function, lipid metabolism, and inflammation resolution. Although extensively studied in neurodegenerative diseases, TREM2's role in metabolic disorders and cardiovascular health is an emerging area of research. This review explores TREM2's molecular structure and functions, emphasizing its contributions to immunometabolic regulation in obesity and T2DM. Evidence from preclinical models demonstrates that TREM2 modulates macrophage-driven inflammatory responses, lipid clearance, plaque stability, fibrosis, and myocardial remodeling. Translational findings suggest that TREM2 expression correlates with cardiometabolic outcomes, underscoring its potential as a therapeutic target. Key knowledge gaps include TREM2's temporal dynamics during disease progression, sex-specific effects, and interactions with recruited or resident macrophage activation in obesity and T2DM. Integrating mechanistic and translational insights is critical to harness TREM2's immunoregulatory potential for improving CVD outcomes in metabolic disorders.
Collapse
Affiliation(s)
- Eunhee Chung
- Department of Kinesiology, University of Texas at San Antonio, San Antonio, Texas, United States
| | - David Zhang
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, Texas, United States
| | - Maria Gonzalez Porras
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, Texas, United States
| | - Chia George Hsu
- Department of Kinesiology, University of Texas at San Antonio, San Antonio, Texas, United States
| |
Collapse
|
36
|
Freedman F, Marsk R, Yan J, Karlsson L, Sandborgh-Englund G. Dental outcomes after gastric bypass and sleeve gastrectomy: a register-based study. Surg Obes Relat Dis 2025; 21:570-579. [PMID: 39710527 DOI: 10.1016/j.soard.2024.12.001] [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: 04/08/2024] [Revised: 11/21/2024] [Accepted: 12/03/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND Bariatric surgery has been shown to cause a negative impact on oral health, as reflected by postsurgical increase of caries-related dental interventions. OBJECTIVES The aim of this study was to compare dental intervention rates after Roux-en-Y gastric bypass (RYGB) and sleeve gastrectomy (SG). SETTING Nationwide and register-based (Sweden). METHODS This 2-staged matched cohort study included all adults who underwent RYGB (n = 26,594) or SG (n = 3416) between 2011 and 2015, registered in the Scandinavian Obesity Surgery Register. Propensity score matching was used to match SG patients to RYGB patients, based on several covariates. The follow-up time was 3 years after surgery. The dental variables were collected from the Dental Health Register, including tooth extractions, restorative interventions (dental fillings), and endodontic interventions (root canal treatment). RESULTS In total, 3317 RYGB and 3317 SG patients were included. Both groups showed increased dental event rates postoperatively. RYGB patients had significantly higher event rates compared with SG postoperatively regarding all interventions, restorative and endodontic interventions. CONCLUSIONS The negative effect on dental outcomes in terms of dental fillings and tooth extractions were higher after RYGB than after SG. The reasons are not clear. More research is needed to replicate these findings, to understand the mechanisms, and further delineate the significance of the surgical method.
Collapse
Affiliation(s)
- Freja Freedman
- Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Richard Marsk
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.
| | - Jane Yan
- Division of Biostatistics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lena Karlsson
- Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
37
|
Yang H, Xia R, Teame T, Meng D, Li S, Wang T, Ding Q, Yao Y, Xu X, Yang Y, Ran C, Zhang Y, Li S, Niemann B, Guan LL, Zhang Z, Zhou Z. Activation of Gut Microbiota-HIF1α Axis Effectively Restores Resistance to Aeromonas veronii Caused by Improper Administration of AiiO-AIO6. J Nutr 2025; 155:1429-1441. [PMID: 40064423 DOI: 10.1016/j.tjnut.2025.03.007] [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: 01/15/2025] [Revised: 02/26/2025] [Accepted: 03/02/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Feeding adult zebrafish a diet supplemented with quenching enzyme AiiO-AIO6 (AIO6) for 3 wk improved the growth performance and disease resistance. However, when the feeding period was extended to 8 wk, zebrafish's disease resistance to Aeromonas veronii decreased. OBJECTIVES We investigated the mechanisms of the reduced disease resistance of zebrafish induced by feeding on an AIO6 supplemented diet for a long term (8 wk) and assessed the effectiveness of feed additives in restoring the low disease resistance. METHODS One-month-old (adult) zebrafish were fed with a basal diet and the basal diet supplemented with AIO6 (10 U/g) for 8 wk (experiment 1). Furthermore, the zebrafish larvae model (experiment 2) was developed and used to study the mechanisms of how AIO6 affected disease resistance (experiment 3). We also investigated the effectiveness of selected prebiotic tributyrin, β-glucan or mannan in activating gut microbiota- HIF1α to restore the low disease resistance of adult zebrafish fed with AIO6 for 8 wk (experiment 4). Lastly, the effects of Bacillus subtilis in activating the gut microbiota-HIF1α and improving the low disease resistance of zebrafish larvae induced by AIO6 were examined (experiment 5). RESULTS Feeding adult zebrafish with AIO6 for 8 wk promoted growth but disordered the gut microbiota and reduced disease resistance. The zebrafish larvae model confirmed that feeding AIO6 for 2 d increased disease resistance, whereas 7 d decreased the resistance by suppressing HIF1α. Using a germ-free zebrafish larvae model, we also demonstrated that AIO6-induced gut microbiota mediated inhibition of HIF1α. Furthermore, zebrafish fed on the AIO6-containing diet supplement with tributyrin, β-glucan, mannan, or Bacillus subtilis activated the gut microbiota-HIF1α axis to reverse the low resistance caused by AIO6. CONCLUSIONS Activating the gut microbiota-HIF1α axis has a vital role in improving intestinal health and restores the low resistance to Aeromonas veronii caused by improper administration of dietary AIO6 in zebrafish.
Collapse
Affiliation(s)
- Hongwei Yang
- China-Norway Joint Lab on Fish Gut Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China; Institute of Marine Sciences, Shantou University, Shantou, China
| | - Rui Xia
- China-Norway Joint Lab on Fish Gut Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China; Kunpeng Institute of Modern Agriculture at Foshan, Chinese Academy of Agricultural Sciences, Foshan, China
| | - Tsegay Teame
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China; Department of Aquaculture and Fisheries, Tigray Agricultural Research Institute (TARI), Mekelle, Tigray, Ethiopia
| | - Delong Meng
- China-Norway Joint Lab on Fish Gut Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Shenghui Li
- China-Norway Joint Lab on Fish Gut Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Tiantian Wang
- China-Norway Joint Lab on Fish Gut Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Qianwen Ding
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yuanyuan Yao
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xiaoqing Xu
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yalin Yang
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Chao Ran
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yaqing Zhang
- Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing, China
| | - Shengkang Li
- Institute of Marine Sciences, Shantou University, Shantou, China
| | - Benjamin Niemann
- Faculty of Land and Food Systems, The University of British Columbia, Vancouver, Canada
| | - Le Luo Guan
- Faculty of Land and Food Systems, The University of British Columbia, Vancouver, Canada
| | - Zhen Zhang
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China; Faculty of Land and Food Systems, The University of British Columbia, Vancouver, Canada.
| | - Zhigang Zhou
- China-Norway Joint Lab on Fish Gut Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China.
| |
Collapse
|
38
|
Ge T, Zou R, Zhang M, Hu J, He K, Li G, Zhang T, Fan X. Natural products alleviate atrial fibrillation by modulating mitochondrial quality control. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156555. [PMID: 40056631 DOI: 10.1016/j.phymed.2025.156555] [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: 11/13/2024] [Revised: 02/02/2025] [Accepted: 02/21/2025] [Indexed: 03/10/2025]
Abstract
BACKGROUND Atrial fibrillation (AF), one of the most common cardiac arrhythmias, is associated with high mortality rates and significant healthcare burdens. Mitochondrial homeostasis has recently emerged as a critical factor in AF pathogenesis but remains at the experimental stage. Current drug and surgical treatments for AF often involve side effects and require ongoing treatment plan evaluation and adjustment. In contrast, natural products (NPs), which have been utilized in China for over 2,000 years, show remarkable efficacy in treating AF and are receiving growing attention. PURPOSE We aimed to investigate the regulatory effects of NPs on mitochondrial quality control (MQC) and their impact on AF occurrence and progression. By constructing a novel NP-mitochondria-AF axis, we propose a framework to translate experimental findings into clinical practice and identify potential therapeutic strategies for AF. METHODS Databases such as PubMed, Web of Science, and China National Knowledge Infrastructure were searched (up to October 2024) using the following keywords: "atrial fibrillation," "traditional Chinese medicine," "mitochondrial biogenesis," "mitochondrial dynamics," "mitophagy," "apoptosis," "oxidative stress," "inflammation," and "Ca2+ concentration." NP targets were identified using the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform, while disease targets were retrieved from Online Mendelian Inheritance in Man, GeneCards, and Therapeutic Target Database. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis was performed using the Metascape database. Protein-protein interactions were analyzed using the STRING database, and core monomers and hub genes were identified using Cytoscape 3.7.2. RESULTS We found a strong relationship between mitochondrial homeostasis and AF development. KEGG pathway analysis indicated that commonly used NPs regulate mitochondrial homeostasis, affecting AF progression through various hub genes, including protein kinase B-alpha (AKT1), jun proto-oncogene (JUN), and tumor necrosis factor (TNF). Molecular docking analysis revealed that NP core monomers exhibited binding affinities to hub genes below -5 kcal/mol and to transforming growth factor-β (TGF-β) below -7 kcal/mol. CONCLUSION NPs, including traditional Chinese medicine (TCM) compounds, TCM monomers, and traditional Chinese patent medicines, alleviate AF by modulating MQC with minimal side effects and high efficacy. These findings highlight the therapeutic potential of NPs as promising candidates for AF treatment and further underscore the importance of MQC in AF pathogenesis.
Collapse
Affiliation(s)
- Teng Ge
- School of Second Clinical Medical, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Rongjun Zou
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou 510120, Guangdong, PR China; Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong, PR China; Guangdong Provincial Key Laboratory of TCM Emergency Research, Guangzhou 510120, Guangdong, PR China
| | - Miao Zhang
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Jinlin Hu
- School of Second Clinical Medical, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Kunyang He
- School of Second Clinical Medical, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Guanmou Li
- Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong, PR China
| | - Tong Zhang
- Heart Failure Center, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510120, Guangdong, PR China.
| | - Xiaoping Fan
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou 510120, Guangdong, PR China; Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, the Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong, PR China; Guangdong Provincial Key Laboratory of TCM Emergency Research, Guangzhou 510120, Guangdong, PR China.
| |
Collapse
|
39
|
Khalaf F, Barayan D, Saldanha S, Jeschke MG. Metabolaging: a new geroscience perspective linking aging pathologies and metabolic dysfunction. Metabolism 2025; 166:156158. [PMID: 39947519 DOI: 10.1016/j.metabol.2025.156158] [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: 11/06/2024] [Revised: 01/31/2025] [Accepted: 02/09/2025] [Indexed: 02/16/2025]
Abstract
With age, our metabolic systems undergo significant alterations, which can lead to a cascade of adverse effects that are implicated in both metabolic disorders, such as diabetes, and in the body's ability to respond to acute stress and trauma. To elucidate the metabolic imbalances arising from aging, we introduce the concept of "metabolaging." This framework encompasses the broad spectrum of metabolic disruptions associated with the hallmarks of aging, including the functional decline of key metabolically active organs, like the adipose tissue. By examining how these organs interact with essential nutrient-sensing pathways, "metabolaging" provides a more comprehensive view of the systemic metabolic imbalances that occur with age. This concept extends to understanding how age-related metabolic disturbances can influence the response to acute stressors, like burn injuries, highlighting the interplay between metabolic dysfunction and the ability to handle severe physiological challenges. Finally, we propose potential interventions that hold promise in mitigating the effects of metabolaging and its downstream consequences.
Collapse
Affiliation(s)
- Fadi Khalaf
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Dalia Barayan
- David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada; Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Sean Saldanha
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Marc G Jeschke
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; David Braley Research Institute, Hamilton, Ontario, Canada; Hamilton Health Sciences, Hamilton, Ontario, Canada; Department of Surgery, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
40
|
Ismaiah MJ, Lo EKK, Chen C, Tsui JSJ, Johnson-Hill WA, Felicianna, Zhang F, Leung HKM, Oger C, Durand T, Lee JCY, El-Nezami H. Alpha-aminobutyric acid administration suppressed visceral obesity and modulated hepatic oxidized PUFA metabolism via gut microbiota modulation. Free Radic Biol Med 2025; 232:86-96. [PMID: 40032028 DOI: 10.1016/j.freeradbiomed.2025.02.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/17/2025] [Accepted: 02/20/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND High-fat diet (HFD) is associated with visceral obesity due to disruption in the lipid metabolism and gut dysbiosis. These symptoms may contribute to hepatic steatosis and the formation of oxidized polyunsaturated fatty acids (PUFAs). Alpha-aminobutyric acid (ABA) is an amino-acid derived metabolite, and its concentration has been correlated with several metabolic conditions and gut microbiome diversity while its direct effects on visceral obesity, lipid metabolism and the gut microbiota are not well understood. This study was designed to investigate the effect of physiological dose of ABA on diet-induced visceral obesity and lipid metabolism dysregulation by examining the fatty acids and oxidized PUFAs profile in the liver as well as the gut microbiota. RESULTS ABA administration reduced visceral obesity by 28 % and lessened adipocyte hypertrophy. The expression of liver Cd36 was lowered by more than 50 % as well as the saturated and monounsaturated FA concentration. Notably, the desaturation index for C16 and C18 FAs that are correlated with adiposity were reduced. The concentration of several DHA-derived oxidized PUFAs were also enhanced. Faecal metagenomics sequencing revealed enriched abundance of Leptogranulimonas caecicola and Bacteroides sp. ZJ-18 and were positively correlated with several DHA- and ALA-derived oxidized PUFAs in ABA group. CONCLUSION Our study revealed the modulatory effect of physiological dose of ABA on attenuating visceral obesity, reducing hepatic steatosis, and promoting the production of anti-inflammatory oxidized PUFAs that were potentially mediated by the gut microbiota.
Collapse
Affiliation(s)
- Marsena Jasiel Ismaiah
- School of Biological Sciences, The University of Hong Kong, Pok Fu Lam Road, Hong Kong Special Administrative Region of China
| | - Emily Kwun Kwan Lo
- School of Biological Sciences, The University of Hong Kong, Pok Fu Lam Road, Hong Kong Special Administrative Region of China
| | - Congjia Chen
- School of Biological Sciences, The University of Hong Kong, Pok Fu Lam Road, Hong Kong Special Administrative Region of China
| | - Jacob Shing-Jie Tsui
- School of Biological Sciences, The University of Hong Kong, Pok Fu Lam Road, Hong Kong Special Administrative Region of China
| | - Winifred Audrey Johnson-Hill
- School of Biological Sciences, The University of Hong Kong, Pok Fu Lam Road, Hong Kong Special Administrative Region of China
| | - Felicianna
- School of Biological Sciences, The University of Hong Kong, Pok Fu Lam Road, Hong Kong Special Administrative Region of China
| | - Fangfei Zhang
- School of Biological Sciences, The University of Hong Kong, Pok Fu Lam Road, Hong Kong Special Administrative Region of China
| | - Hoi Kit Matthew Leung
- School of Biological Sciences, The University of Hong Kong, Pok Fu Lam Road, Hong Kong Special Administrative Region of China
| | - Camille Oger
- Institut des Biomolécules Max Mousseron, UMR5247, CNRS, ENSCM, Université de Montpellier, F-34093, Montpellier, France
| | - Thierry Durand
- Institut des Biomolécules Max Mousseron, UMR5247, CNRS, ENSCM, Université de Montpellier, F-34093, Montpellier, France
| | - Jetty Chung-Yung Lee
- School of Biological Sciences, The University of Hong Kong, Pok Fu Lam Road, Hong Kong Special Administrative Region of China.
| | - Hani El-Nezami
- School of Biological Sciences, The University of Hong Kong, Pok Fu Lam Road, Hong Kong Special Administrative Region of China; Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, Kuopio, FI-70211, Finland
| |
Collapse
|
41
|
Kruglikov IL, Scherer PE. Regulation of the terminal complement cascade in adipose tissue for control of its volume, cellularity, and fibrosis. Obesity (Silver Spring) 2025; 33:839-850. [PMID: 40134146 PMCID: PMC12015659 DOI: 10.1002/oby.24270] [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: 11/12/2024] [Revised: 12/28/2024] [Accepted: 01/26/2025] [Indexed: 03/27/2025]
Abstract
White adipose tissue (WAT) is a reservoir for various pathogens and their products, such as lipopolysaccharides. Therefore, it must be equipped with a defense mechanism connected with the activation of innate immunity. This explains the phenomenon that adipocytes express components of the classical and alternative complement pathways, which can be activated even in the absence of opportunistic pathogens. Terminal stages of the complement pathway are related to the production of membrane attack complexes and, thus, can cause lysis of pathogens, as well as autolysis of host adipocytes, contributing to the regulation of the cellularity in WAT. Complement-induced autolysis of adipocytes is counteracted by a number of cellular defense mechanisms. This versatility of activation and suppression processes enables a broad range of adaptability to physiological contexts, ranging from the development of hypertrophic WAT to lipodystrophy. Pathogen-induced activation of the complement pathway in WAT also induces a profibrotic phenotype. These processes may also be involved in the regulation of insulin resistance in adipocytes. This explains the dual immune/metabolic role of the complement pathway in WAT: the pathway is an integral part of the immune response but also potently involved in the control of volume and cellularity of WAT under both physiological and pathological conditions.
Collapse
Affiliation(s)
| | - Philipp E. Scherer
- Touchstone Diabetes CenterUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| |
Collapse
|
42
|
Lecoutre S, Rebière C, Maqdasy S, Lambert M, Dussaud S, Abatan JB, Dugail I, Gautier EL, Clément K, Marcelin G. Enhancing adipose tissue plasticity: progenitor cell roles in metabolic health. Nat Rev Endocrinol 2025; 21:272-288. [PMID: 39757324 DOI: 10.1038/s41574-024-01071-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/27/2024] [Indexed: 01/07/2025]
Abstract
Adipose tissue demonstrates considerable plasticity and heterogeneity, enabling metabolic, cellular and structural adaptations to environmental signals. This adaptability is key for maintaining metabolic homeostasis. Impaired adipose tissue plasticity can lead to abnormal adipose tissue responses to metabolic cues, which contributes to the development of cardiometabolic diseases. In chronic obesity, white adipose tissue undergoes pathological remodelling marked by adipocyte hypertrophy, chronic inflammation and fibrosis, which are linked to local and systemic insulin resistance. Research data suggest that the capacity for healthy or unhealthy white adipose tissue remodelling might depend on the intrinsic diversity of adipose progenitor cells (APCs), which sense and respond to metabolic cues. This Review highlights studies on APCs as key determinants of adipose tissue plasticity, discussing differences between subcutaneous and visceral adipose tissue depots during development, growth and obesity. Modulating APC functions could improve strategies for treating adipose tissue dysfunction and metabolic diseases in obesity.
Collapse
Affiliation(s)
- Simon Lecoutre
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France.
| | - Clémentine Rebière
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Salwan Maqdasy
- Department of Medicine, Karolinska Institutet Hospital, Stockholm, Sweden
| | - Mélanie Lambert
- Institut National de la Santé et de la Recherche Médicale, Bobigny, France
- Labex Inflamex, Université Sorbonne Paris Nord, Alliance Sorbonne Paris Cité, Bobigny, France
| | - Sébastien Dussaud
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Jimon Boniface Abatan
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Isabelle Dugail
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Emmanuel L Gautier
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Karine Clément
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France.
- Department of Nutrition, Pitie-Salpêtriere Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France.
| | - Geneviève Marcelin
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France.
| |
Collapse
|
43
|
Li Z, Cheng W, Gao K, Liang S, Ke L, Wang M, Fan J, Li D, Zhang P, Xu Z, Li N. Pyroptosis: A spoiler of peaceful coexistence between cells in degenerative bone and joint diseases. J Adv Res 2025; 71:227-262. [PMID: 38876191 DOI: 10.1016/j.jare.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 05/23/2024] [Accepted: 06/07/2024] [Indexed: 06/16/2024] Open
Abstract
BACKGROUND As people age, degenerative bone and joint diseases (DBJDs) become more prevalent. When middle-aged and elderly people are diagnosed with one or more disorders such as osteoporosis (OP), osteoarthritis (OA), and intervertebral disc degeneration (IVDD), it often signals the onset of prolonged pain and reduced functionality. Chronic inflammation has been identified as the underlying cause of various degenerative diseases, including DBJDs. Recently, excessive activation of pyroptosis, a form of programed cell death (PCD) mediated by inflammasomes, has emerged as a primary driver of harmful chronic inflammation. Consequently, pyroptosis has become a potential target for preventing and treating DBJDs. AIM OF REVIEW This review explored the physiological and pathological roles of the pyroptosis pathway in bone and joint development and its relation to DBJDs. Meanwhile, it elaborated the molecular mechanisms of pyroptosis within individual cell types in the bone marrow and joints, as well as the interplay among different cell types in the context of DBJDs. Furthermore, this review presented the latest compelling evidence supporting the idea of regulating the pyroptosis pathway for DBJDs treatment, and discussed the potential, limitations, and challenges of various therapeutic strategies involving pyroptosis regulation. KEY SCIENTIFIC CONCEPTS OF REVIEW In summary, an interesting identity for the unregulated pyroptosis pathway in the context of DBJDs was proposed in this review, which was undertaken as a spoiler of peaceful coexistence between cells in a degenerative environment. Over the extended course of DBJDs, pyroptosis pathway perpetuated its activity through crosstalk among pyroptosis cascades in different cell types, thus exacerbating the inflammatory environment throughout the entire bone marrow and joint degeneration environment. Correspondingly, pyroptosis regulation therapy emerged as a promising option for clinical treatment of DBJDs.
Collapse
Affiliation(s)
- Zhichao Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Wenxiang Cheng
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Kuanhui Gao
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Songlin Liang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Liqing Ke
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Mengjie Wang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Jilin Fan
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Dandan Li
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050011, China
| | - Peng Zhang
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Faculty of Biomedical Engineering, Shenzhen University of Advanced Technology, Shenzhen 518000, China; Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, 518000 China; Shandong Zhongke Advanced Technology Co., Ltd., Jinan, 250300 China.
| | - Zhanwang Xu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Nianhu Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| |
Collapse
|
44
|
Bat-Erdene B, He M, Dong J, Li Y, Ta D. Therapeutic Effects of Different Ultrasound Intensity Stimulation on Brown Adipose Tissue for the Treatment of Type 2 Diabetes. ULTRASOUND IN MEDICINE & BIOLOGY 2025; 51:830-840. [PMID: 39924417 DOI: 10.1016/j.ultrasmedbio.2025.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 02/11/2025]
Abstract
Type 2 diabetes (T2D) is a persistent illness that has a high incidence rate. Still, there is no conclusive evidence on effectively improving blood sugar levels in patients through physical therapy. This study examined the regulatory effects of different intensities of low-intensity pulsed ultrasound (LIPUS) on T2D by stimulating brown adipose tissue (BAT). Eight-week-old C57BL/6J mice were divided into six groups (n = 10 per group): Control sham (C-Sham), Control-LIPUS (C-LIPUS), T2D-sham (T2D-Sham), T2D groups treated with LIPUS at spatial average-temporal-average intensity (Isata) of 60mW/cm² (T2D-L-60), 80mW/cm² (T2D-L-80), and 100mW/cm² (T2D-L-100). T2D models were induced by intraperitoneal injection of 40 mg/kg streptozotocin (STZ) three times after 12 wks of high-fat diet (HFD). The T2D-LIPUS group received LIPUS stimulation for 20 minutes per day for 6 weeks. The LIPUS stimulation had a duty cycle of 20%, a frequency of 1 MHz, and Isata of 60mW/cm², 80mW/cm², 100mW/cm². Subsequently, glucose tolerance tests (GTT) and insulin tolerance tests (ITT) were performed, and body fat content in mice was analyzed using nuclear magnetic resonance (NMR). Metabolic changes were monitored using metabolic cages. The results indicated that 80mW/cm² intensity level significantly improved glucose tolerance, insulin sensitivity, and metabolic function after LIPUS exposure. Significant reductions in body fat content and enhanced thermogenesis were observed, highlighting the potential of LIPUS in T2D management. This provides the basis for the dose study of LIPUS in the treatment of T2D.
Collapse
Affiliation(s)
- Badamgarav Bat-Erdene
- Department of Biomedical Engineering, School of Information Science and Technology, Fudan University, Shanghai, China
| | - Min He
- Department of Biomedical Engineering, School of Information Science and Technology, Fudan University, Shanghai, China.
| | - Jingsong Dong
- Department of Biomedical Engineering, School of Information Science and Technology, Fudan University, Shanghai, China
| | - Ying Li
- Department of Biomedical Engineering, School of Information Science and Technology, Fudan University, Shanghai, China
| | - Dean Ta
- Department of Biomedical Engineering, School of Information Science and Technology, Fudan University, Shanghai, China; Academy for Engineering and Technology, Fudan University, Shanghai, China; State Key Laboratory of Integrated Chips and Systems, Fudan University, Shanghai, China; Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
45
|
Newport ME, Wilson P, Lowes S, Behrends M, Coons A, Bowman J, Bates HE. Photoperiod influences visceral adiposity and the adipose molecular clock independent of temperature in wild-derived Peromyscus leucopus. FASEB Bioadv 2025; 7:e70006. [PMID: 40330430 PMCID: PMC12050962 DOI: 10.1096/fba.2024-00115] [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: 07/18/2024] [Revised: 02/08/2025] [Accepted: 02/17/2025] [Indexed: 05/08/2025] Open
Abstract
Physiology is closely synchronized to daily and seasonal light/dark cycles. Humans artificially extend daylight and experience irregular light schedules, resulting in dysregulation of metabolism and body mass. In rodents, winter-like conditions (cold and short photoperiod) can alter energy balance and adipose tissue mass. To determine if photoperiod alone, independent of temperature, is a strong enough signal to regulate adiposity, we compared the effects of long and short photoperiod at thermoneutrality on adiposity and WAT gene expression in photoperiod-sensitive, F1 generation wild-derived adult male white-footed mice (Peromyscus leucopus). Mice were housed in long-day (16:8 light:dark) or short-day (8:16 light:dark) photoperiod conditions at thermoneutrality (27°C) for 4 weeks with the extended light being provided through artificial lighting. Photoperiod did not impact body weight or calorie consumption. However, mice housed in long photoperiod with extended artificial light selectively developed greater visceral WAT mass without changing subcutaneous WAT or interscapular BAT mass. This was accompanied by a decrease in Adrβ3 and Ucp1 mRNA expression in visceral WAT with no change in Pgc1a, Lpl, or Hsl. Expression of Per1, Per2, and Nr1d1 mRNA in visceral WAT differed between long and short photoperiods over time when aligned to circadian time but not onset of darkness, indicating alterations in clock gene expression with photoperiod. These findings suggest that extended photoperiod through artificial light can promote visceral fat accumulation alone, independent of temperature, supporting that artificial light may play a role in obesity.
Collapse
Affiliation(s)
| | - Paul Wilson
- Department of BiologyTrent UniversityPeterboroughOntarioCanada
| | - Shanna Lowes
- Department of BiologyTrent UniversityPeterboroughOntarioCanada
| | - Marthe Behrends
- Department of BiologyTrent UniversityPeterboroughOntarioCanada
| | - Alexis Coons
- Department of BiologyTrent UniversityPeterboroughOntarioCanada
| | - Jeff Bowman
- Department of BiologyTrent UniversityPeterboroughOntarioCanada
- Wildlife Research and Monitoring SectionOntario Ministry of Natural ResourcesPeterboroughOntarioCanada
| | - Holly E. Bates
- Department of BiologyTrent UniversityPeterboroughOntarioCanada
| |
Collapse
|
46
|
Sousa-Filho CPB, Petrovic N. No UCP1 in the kidney. Mol Metab 2025; 95:102127. [PMID: 40120980 PMCID: PMC11995138 DOI: 10.1016/j.molmet.2025.102127] [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: 02/19/2025] [Revised: 03/05/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025] Open
Abstract
OBJECTIVES Several recent studies have indicated the presence of UCP1 in the kidney, challenging the paradigm that UCP1 is only found in brown and beige adipocytes and broadening the (patho)physiological significance of UCP1. The kidney localization has been the direct result of immunohistochemical investigations and an inferred outcome from multiple lines of reporter mice. These findings require confirmation and further physiological characterization. METHODS We examined UCP1 expression in the kidney using immunohistochemistry and qPCR. Transversal sections through or near the kidney hilum, consistently including perirenal brown fat and adjacent kidney tissue, were analyzed with four UCP1 antibodies. RESULTS In addition to detecting UCP1 in perirenal adipose tissue, we observed distinct immunopositive structures in the kidney with our in-house UCP1-antibody, 'C10', in apparent agreement with earlier reports. To corroborate this, we tested the C10-antibody on kidney sections from UCP1-ablated mice but found equal reactivity in these UCP1-negative tissues. We then tested the widely used antibody ab10983, previously employed in kidney studies. Also here, the positive signal persisted in UCP1-ablated mice, clearly invalidating earlier findings. UCP1 qPCR studies also failed to detect UCP1 mRNA above background. Finally, two highly specific antibodies, E9Z2V and EPR20381, accurately detected UCP1 in perirenal adipose tissue but showed no signal in the kidney. CONCLUSIONS When appropriate controls are implemented, there is no evidence for the presence of UCP1 in the kidney. Consequently, this conclusion also implies that the results from UCP1 reporter mice, specifically regarding kidney expression of the UCP1 gene - though possibly applicable to other tissues - require reconfirmation before being accepted as evidence for the presence of UCP1 in non-adipose tissues.
Collapse
Affiliation(s)
| | - Natasa Petrovic
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden.
| |
Collapse
|
47
|
Emont MP, Essene AL, Gulko A, Bozadjieva-Kramer N, Jacobs C, Nagesh S, Seeley RJ, Tsai LT, Rosen ED. Semaglutide and bariatric surgery induce distinct changes in the composition of mouse white adipose tissue. Mol Metab 2025; 95:102126. [PMID: 40139440 PMCID: PMC11999362 DOI: 10.1016/j.molmet.2025.102126] [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: 01/16/2025] [Revised: 03/13/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
Adipose tissue is a central player in energy balance and glucose homeostasis, expanding in the face of caloric overload in order to store energy safely. If caloric overload continues unabated, however, adipose tissue becomes dysfunctional, leading to systemic metabolic compromise in the form of insulin resistance and type 2 diabetes. Changes in adipose tissue during the development of metabolic disease are varied and complex, made all the more so by the heterogeneity of cell types within the tissue. Here we present detailed comparisons of atlases of murine WAT in the setting of diet-induced obesity, as well as after weight loss induced by either vertical sleeve gastrectomy (VSG) or treatment with the GLP-1 receptor agonist semaglutide. We focus on identifying populations of cells that return to a lean-like phenotype versus those that persist from the obese state, and examine pathways regulated in these cell types across conditions. These data provide a resource for the study of the cell type changes in WAT during weight loss, and paint a clearer picture of the differences between adipose tissue from lean animals that have never been obese, versus those that have.
Collapse
Affiliation(s)
- Margo P Emont
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Harvard Medical School, Boston, MA, USA.
| | - Adam L Essene
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Anton Gulko
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Nadejda Bozadjieva-Kramer
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA; Veterans Affairs Ann Arbor Healthcare System, Research Service, Ann Arbor, MI, USA
| | - Christopher Jacobs
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Soumya Nagesh
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Randy J Seeley
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Linus T Tsai
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Evan D Rosen
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
48
|
Shirai T, Uemichi K, Iwai R, Shinkai H, Iwata T, Tanimura R, Sugiyama S, Takemasa T. Systemic effect of combined functional overload and endurance-type swimming exercise on whole body metabolism in mice. Am J Physiol Endocrinol Metab 2025; 328:E695-E710. [PMID: 40248969 DOI: 10.1152/ajpendo.00433.2024] [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: 10/28/2024] [Revised: 02/06/2025] [Accepted: 03/26/2025] [Indexed: 04/19/2025]
Abstract
In this study, we examined the effects of concurrent functional overload and endurance exercise on muscle hypertrophy, mitochondrial function, and systemic adaptations in male mice. The mice were assigned to three groups: Sham (Sham), overload-induced hypertrophy (OL), and overload with concurrent 60-min free swimming (5 times/wk) (OL + Swim), for 4 wk. Although OL promoted muscle hypertrophy and protein synthesis through the Akt/mammalian/mechanistic target of rapamycin (mTOR) signaling pathway, the addition of swimming (OL + Swim) attenuated these effects, resulting in less pronounced muscle growth and a smaller increase in myofiber cross-sectional area. Notably, the OL + Swim group exhibited enhanced mitochondrial activity and glycogen content compared with the OL group. Both the OL and OL + Swim groups showed elevated rates of protein synthesis, with a significant upregulation of AMP-activated kinase (AMPK) and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) in the OL + Swim group, suggesting enhanced mitochondrial biogenesis and adaptation. Concurrent training also resulted in systemic benefits, including reduced inguinal and epididymal white adipocyte size, improved mitochondrial enzyme activities in adipose and liver tissues, and higher levels of fibronectin type III domain containing protein 5 (FNDC5), fibroblast growth factor 21 (FGF21), and brain-derived neurotrophic factor (BDNF) in serum, which contributed to enhanced muscle protein synthesis in cultured muscle cells. These results highlight the trade-offs between muscle hypertrophy and metabolic health in mice and underscore the importance of balanced training regimens to optimize overall metabolic health and muscle function. Our results provide further insight into how concurrent strength and endurance training can be optimized for health and performance benefits.NEW & NOTEWORTHY This study provides novel insights into the mechanisms underlying the interference effect that occurs in concurrent training, highlighting the potential systemic benefits of combining resistance and endurance exercises. Despite a reduction in muscle hypertrophy, concurrent training enhances metabolic adaptations and systemic health markers and offers a comprehensive approach to improving both muscle and metabolic fitness.
Collapse
Affiliation(s)
- Takanaga Shirai
- Department of Human Sciences, Kanagawa University, Kanagawa, Japan
- Research Fellow of Japan Society for Promotion Science, Tokyo, Japan
- Institute of Health and Sport Sciences, University of Tsukuba, Ibaraki, Japan
| | - Kazuki Uemichi
- Research Fellow of Japan Society for Promotion Science, Tokyo, Japan
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Ryoto Iwai
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Hayato Shinkai
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Tomohiro Iwata
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Riku Tanimura
- Research Fellow of Japan Society for Promotion Science, Tokyo, Japan
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Shunsuke Sugiyama
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Tohru Takemasa
- Institute of Health and Sport Sciences, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
49
|
Liang Y, Zhu Z, Lu Y, Ma C, Li J, Yu K, Wu J, Che X, Liu X, Huang X, Li P, Chen FJ. Cytoskeleton regulates lipid droplet fusion and lipid storage by controlling lipid droplet movement. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159610. [PMID: 40189192 DOI: 10.1016/j.bbalip.2025.159610] [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: 11/20/2024] [Revised: 03/17/2025] [Accepted: 04/02/2025] [Indexed: 04/13/2025]
Abstract
Lipid droplets (LDs) are highly dynamic organelles that maintain cellular lipid homeostasis through size and number control. In adipose tissue, CIDEC plays a crucial role in LD fusion and lipid homeostasis. However, the regulatory factors and mechanisms of LD fusion remain largely unknown. Here, we established a high-throughput LD phenotypic screen on a compound library consisting of 2010 small molecules, and identified 11 cytoskeleton inhibitors that negatively regulate LD size. Using specific inhibitors against each of the three types of cytoskeleton, our data showed that the disruption of microtubules and microfilaments but not intermediate filaments limits CIDEC-mediated LD fusion and growth by reducing LD movement and LD-LD contact. The collective effect of microtubule inhibitors results in a small LD phenotype which favors lipolysis upon activation of cAMP-PKA pathway in adipocytes. Our findings demonstrate that cytoskeleton is involved in the process of LD fusion and growth, indicating their role in lipid storage metabolism. One-Sentence Summary: Cytoskeleton regulates lipid droplet fusion and lipid storage.
Collapse
Affiliation(s)
- Yan Liang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, State Key Laboratory of Genetics and Development of Complex Phenotypes, Institute of Metabolism and Integrative Biology, School of Life Sciences, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Zanzan Zhu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, State Key Laboratory of Genetics and Development of Complex Phenotypes, Institute of Metabolism and Integrative Biology, School of Life Sciences, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Yiming Lu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, State Key Laboratory of Genetics and Development of Complex Phenotypes, Institute of Metabolism and Integrative Biology, School of Life Sciences, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Chengxin Ma
- Shanghai Key Laboratory of Metabolic Remodeling and Health, State Key Laboratory of Genetics and Development of Complex Phenotypes, Institute of Metabolism and Integrative Biology, School of Life Sciences, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Jiacheng Li
- Shanghai Key Laboratory of Metabolic Remodeling and Health, State Key Laboratory of Genetics and Development of Complex Phenotypes, Institute of Metabolism and Integrative Biology, School of Life Sciences, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Kuan Yu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, State Key Laboratory of Genetics and Development of Complex Phenotypes, Institute of Metabolism and Integrative Biology, School of Life Sciences, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Jin Wu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, State Key Laboratory of Genetics and Development of Complex Phenotypes, Institute of Metabolism and Integrative Biology, School of Life Sciences, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Xinmeng Che
- Shanghai Key Laboratory of Metabolic Remodeling and Health, State Key Laboratory of Genetics and Development of Complex Phenotypes, Institute of Metabolism and Integrative Biology, School of Life Sciences, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Xu Liu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, State Key Laboratory of Genetics and Development of Complex Phenotypes, Institute of Metabolism and Integrative Biology, School of Life Sciences, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Xiaoxiao Huang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, State Key Laboratory of Genetics and Development of Complex Phenotypes, Institute of Metabolism and Integrative Biology, School of Life Sciences, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200438, China
| | - Peng Li
- Shanghai Key Laboratory of Metabolic Remodeling and Health, State Key Laboratory of Genetics and Development of Complex Phenotypes, Institute of Metabolism and Integrative Biology, School of Life Sciences, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200438, China; State Key Laboratory of Membrane Biology and Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Shanghai Qi Zhi Institute, Shanghai 200030, China
| | - Feng-Jung Chen
- Shanghai Key Laboratory of Metabolic Remodeling and Health, State Key Laboratory of Genetics and Development of Complex Phenotypes, Institute of Metabolism and Integrative Biology, School of Life Sciences, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200438, China; Shanghai Qi Zhi Institute, Shanghai 200030, China.
| |
Collapse
|
50
|
Zhong Y, Yan J, Lei Y, Zhang R, Abudurexiti A, Qi S, Hou W, Ma X. Lactucin and lactucopicrin ameliorate obesity in high-fat diet fed mice by promoting white adipose tissue browning through the activation of the AMPK/SIRT1/PGC-1α pathway. J Nutr Biochem 2025; 139:109851. [PMID: 39909319 DOI: 10.1016/j.jnutbio.2025.109851] [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: 06/20/2024] [Revised: 01/21/2025] [Accepted: 01/29/2025] [Indexed: 02/07/2025]
Abstract
Lactucin and lactucopicrin are the characteristic lipid-lowering active components found in Cichorium glandulosum. However, their effects and underlying mechanisms in obesity remain unclear. In the present study, C57BL/6J mice were simultaneously subjected to a high-fat diet (HFD) and treated with drugs to investigate the impacts of lactucin and lactucopicrin on HFD-induced obese mice. The results demonstrated that in HFD obese mice, lactucin and lactucopicrin significantly decreased body weight and the weights of adipose tissues, improved serum metabolic parameters, and increased the content of irisin. Regarding the intermediate metabolites of intestinal flora, which are closely associated with white adipose tissue (WAT) browning, lactucin and lactucopicrin treatment led to a reduction in the levels of 12-α-OH/non-12-α-OH bile acids (BAs) and also tended to enhance the levels of short-chain fatty acids (SCFAs). qRT-PCR results indicated that lactucin and lactucopicrin treatment elevated the expression levels of genes related to beige fat markers, thermogenesis, mitochondrial biogenesis, and lipolysis in WAT, as well as those of thermogenesis and lipolysis genes in brown adipose tissue (BAT). Western blot analysis revealed that lactucin and lactucopicrin up-regulated the expression of uncoupling protein 1 (UCP1), the core protein in thermogenesis, in both WAT and BAT. Moreover, they also up-regulated the expression levels of AMP-activated kinase (AMPK), sirtuin 1 (SIRT1), and PPARγ coactivator 1-alpha (PGC-1α), which are key pathway proteins involved in WAT browning. Furthermore, 16S rRNA sequencing results showed that in HFD obese mice, lactucin and lactucopicrin improved the composition and function of the intestinal microbiota. In conclusion, lactucin and lactucopicrin may promote WAT browning by activating the AMPK/SIRT1/PGC-1α pathway, thereby ameliorating obesity in HFD mice.
Collapse
Affiliation(s)
- Yewei Zhong
- College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | - Junlin Yan
- College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | - Yi Lei
- College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | - Rui Zhang
- College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | | | - Shuwen Qi
- College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | - Wenhui Hou
- College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | - Xiaoli Ma
- College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China.
| |
Collapse
|