1
|
Gong H, Qi Y, Wu X, Wu L, Liu W, Chen H, Qiu J, Wen H, Shen Z, Wang Z, Zhang M, Xu Z, Xu Z, Sun B, Li X, Zhao Q. UCP1-inspired mitochondrial uncouplers: Design, synthesis and thermogenic activity studies. Bioorg Chem 2025; 161:108466. [PMID: 40319812 DOI: 10.1016/j.bioorg.2025.108466] [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: 12/05/2024] [Revised: 04/09/2025] [Accepted: 04/09/2025] [Indexed: 05/07/2025]
Abstract
Given that uncoupling protein 1 (UCP1) serves as the principal thermogenic effector in adipose tissue, and motivated by recent breakthroughs in its structural characterization, we tried to obtain the precursor compounds with optimal docking mode, tryptophan compounds, by virtual screening. Following the initial structural refinement of these derivatives, we synthesized a series of tryptophan-based compounds aimed at enhancing their functional properties. Notably, the tryptophan-derived compound ZGL-18 was found to effectively induce lipid consumption without causing toxicity in brown adipocytes at a concentration of 100 μmol/L. In addition, administration of ZGL-18 stimulates brown adipocytes, trigger a reduction in mitochondrial membrane potential. In vivo experiments showed that ZGL-18 at a dose of 100 mg/kg significantly enhanced thermogenesis and cold tolerance in mice with preserving core and skin temperature. ZGL-18 was found to efficiently enhance respiration as well as energy expenditure in cold environments, in addition to passing, and was non-toxic at a dose of 1000 mg/kg. Furthermore, we preliminarily confirmed the binding mode of ZGL-18 to UCP1 by molecular docking and molecular dynamics simulation. These results suggest that ZGL-18 might be a promising candidate for drug development targeting UCP1.
Collapse
Affiliation(s)
- Hao Gong
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, People's Republic of China
| | - Yiming Qi
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, People's Republic of China
| | - Xinyi Wu
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, People's Republic of China
| | - Limeng Wu
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, People's Republic of China
| | - Wenjie Liu
- College of Life and Health Sciences, Northeastern University, Shenyang 110819, People's Republic of China
| | - Huanhua Chen
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, People's Republic of China
| | - Jingsong Qiu
- State Key Laboratory of Digital Medical Engineering, Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Sanya, Hainan 572025, People's Republic of China
| | - Han Wen
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, People's Republic of China
| | - Zixian Shen
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, People's Republic of China
| | - Zhiya Wang
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, People's Republic of China
| | - Mingzuo Zhang
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, People's Republic of China
| | - Zonghe Xu
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, People's Republic of China
| | - Zihua Xu
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, People's Republic of China
| | - Bohang Sun
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China.
| | - Xiang Li
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, People's Republic of China.
| | - Qingchun Zhao
- Department of Pharmacy, General Hospital of Northern Theater Command, Shenyang 110840, People's Republic of China.
| |
Collapse
|
2
|
Mohaghegh N, Iyer A, Wang E, Balajam NZ, Kang H, Akbari M, Barnhill MS, Khademhosseini A, Pearson RM, Hassani Najafabadi A. Apigenin-loaded nanoparticles for obesity intervention through immunomodulation and adipocyte browning. J Control Release 2025; 382:113670. [PMID: 40187647 DOI: 10.1016/j.jconrel.2025.113670] [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: 12/11/2024] [Revised: 03/04/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025]
Abstract
Obesity is characterized by a significant imbalance in adipose tissue macrophages (ATMs), shifting from anti-inflammatory M2 to pro-inflammatory M1 phenotypes, contributing to chronic low-grade inflammation and metabolic dysfunction. This study explores the potential of nanoparticle (NP)-mediated immunomodulation to address obesity-related inflammation, adipocyte browning, and metabolic dysfunction. Apigenin (Api), a natural compound with notable anti-inflammatory properties, was encapsulated within poly(lactic-co-glycolic acid) (PLGA) NPs (Api-NPs) for localized delivery to adipose tissues (ATs). Api-NPs demonstrated favorable physicochemical properties and sustained release profiles. In vitro, Api-NPs, increased M2 macrophage (MΦ) polarization, reduced inflammatory markers, and promoted adipocyte browning. In a high-fat diet (HFD)-induced obesity mouse model, Api-NP administration effectively modulated MΦ polarization and enhanced AT browning, leading to marked reductions in body weight and AT mass. Our findings indicate that Api-NP treatment mitigates obesity-related inflammation and promotes beneficial changes in AT composition and function. Importantly, histological evaluations confirmed the absence of toxicity in major organs, reinforcing the safety profile of this approach. These results position Api-NPs as a promising novel therapeutic strategy for obesity management, integrating immune modulation and localized drug delivery to address the complexities of obesity and its associated inflammatory processes.
Collapse
Affiliation(s)
- Neda Mohaghegh
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Anjali Iyer
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
| | - Ethan Wang
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Heemin Kang
- Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Mohsen Akbari
- Laboratory for Innovations in MicroEngineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC, Canada
| | | | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Ryan M Pearson
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA.
| | | |
Collapse
|
3
|
Kalim F, Sivaraman G, Vankhede H, Ramesh A, Raja SO, Gulyani A. A red-shifted donor-acceptor hemicyanine-based probe for mitochondrial pH in live cells. J Mater Chem B 2025; 13:5550-5557. [PMID: 40243010 DOI: 10.1039/d4tb01839g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
pH dynamically regulates diverse cellular functions and processes. At the inner mitochondrial membrane (IMM), nanoscale pH gradients generated by the electron transport chain (ETC) play a critical role in contributing to mitochondrial membrane potential that drives ATP synthesis and thermogenesis. However, tools to decouple pH gradients from the overall IMM potential in living cells are limited. This study integrates a fluorescent "benzo-indole" chromophore with a pH-sensitive "phenol" moiety into a single covalent skeleton to build a sensitive, red-shifted, cell-permeable pH probe (Mito-pH2). Mito-pH2 localizes inside mitochondria with high specificity presumably to the mitochondrial inner membrane by virtue of being an amphiphilic cation and can report dynamic changes in mitochondrial pH in living cells. Our design ensures that Mito-pH2 exhibits pH-sensitive dual-excitation and dual-emission peaks enabling ratiometric pH-sensing. Furthermore, Mito-pH2 reports an increase in pH in the pH range of 3-9 through a striking colour change from yellow to purple making it a sensitive all-purpose colorimetric pH probe. A combination of DFT calculations and spectroscopy shed light on likely sensing mechanisms including photophysics. Quantitative live-cell fluorescence imaging reveals that Mito-pH2 can detect dynamic changes in mitochondrial pH upon extracellular pH modulation with little or no measurable cytotoxicity during live imaging. Red-emitting Mito-pH2 opens new avenues of quantitative mapping of physiological mitochondrial membrane pH and significantly enhances the repertoire of environment-sensitive and low-toxicity mitochondrial probes that link mitochondrial state and micro-environment.
Collapse
Affiliation(s)
- Fouzia Kalim
- National Centre for Biological Sciences, GKVK Post, Bellary Road, Bengaluru 560065, India
| | - Gandhi Sivaraman
- Institute for Stem Cell Science and Regenerative Medicine, GKVK Post, Bellary Road, Bengaluru 560065, India.
- Department of Chemistry, Gandhigram Rural Institute-Deemed to be University, Gandhigram, Dindigul 624302, Tamil Nadu, India
| | - Himanshu Vankhede
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Central University Post, Prof. C.R. Rao Road, Gachibowli, Hyderabad, Telangana 500046, India
| | - Arati Ramesh
- National Centre for Biological Sciences, GKVK Post, Bellary Road, Bengaluru 560065, India
| | - Sufi O Raja
- Institute for Stem Cell Science and Regenerative Medicine, GKVK Post, Bellary Road, Bengaluru 560065, India.
- School of Chemical and Biotechnology, SASTRA Deemed University, Tirumalaisamudram, Thanjavur 613401, Tamilnadu, India
| | - Akash Gulyani
- Institute for Stem Cell Science and Regenerative Medicine, GKVK Post, Bellary Road, Bengaluru 560065, India.
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Central University Post, Prof. C.R. Rao Road, Gachibowli, Hyderabad, Telangana 500046, India
| |
Collapse
|
4
|
Gu Y, He W, Li W, Cai J, Wang Z, Li K, Qin G, Gu X, Lin X, Ma L, Xiao X, Hou Y, Luo T. Arctiin, a lignan compound, enhances adipose tissue browning and energy expenditure by activating the adenosine A 2A receptor. Mol Med 2025; 31:188. [PMID: 40369420 PMCID: PMC12079995 DOI: 10.1186/s10020-025-01249-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 05/06/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND The activation of brown adipose tissue (BAT) or the browning of white adipose tissue (WAT) represents a promising therapeutic strategy for obesity. Arctiin (ARC), a lignan compound known for its anti-inflammatory, anti-tumor, and hypoglycemic properties, has not been fully elucidated regarding its effects and mechanisms on obesity. METHODS In the present study, we established both high-fat diet-induced obese mouse models and mature adipocyte cultures to comprehensively investigate the therapeutic effects of ARC on obesity. Systemic energy metabolism and thermogenic capacity were assessed through metabolic cage monitoring and cold stimulation tests. Histopathological alterations in adipose tissues were examined using hematoxylin and eosin (H&E) staining, while key gene expression in adipocytes was determined by Western blotting (WB), immunohistochemistry, and immunofluorescence staining. To further elucidate the molecular mechanisms underlying ARC's anti-obesity effects, we employed an integrated approach combining network pharmacology analysis, molecular docking simulations, cellular thermal shift assay (CETSA), and WB to identify potential molecular targets and delineate the associated signaling pathways modulated by ARC treatment. RESULTS In diet-induced obese mice, ARC administration at doses of 20 and 60 mg/kg/day ameliorated metabolic dysfunction through enhanced WAT browning and increased energy expenditure. In C3H10T1/2-induced adipocytes, ARC upregulated the protein expression of uncoupling protein 1 (UCP1), peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), and other brown-specific marker genes, promoting mitochondrial function and browning of adipocytes. Mechanistically, our findings suggest that ARC may promote adipocyte browning via the A2AR-cyclic AMP (cAMP)-protein kinase A (PKA) signaling pathway. CONCLUSION In summary, ARC exerts protective effects against obesity by promoting the browning of white adipocytes and holds promise as a potentially beneficial therapeutic agent for the treatment of obesity.
Collapse
Affiliation(s)
- Yuanfeng Gu
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Wenjun He
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Wenxuan Li
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Jingshu Cai
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Zhuyun Wang
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Kemeng Li
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Guangcheng Qin
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
- Laboratory Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xiaojie Gu
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Xiaojing Lin
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Li Ma
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China
| | - Xiaoqiu Xiao
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China.
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China.
| | - Yi Hou
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China.
- Laboratory of Traditional Chinese Medicine, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing, 400016, China.
| | - Ting Luo
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
5
|
Mori MP, Lozoya OA, Brooks AM, Bortner CD, Nadalutti CA, Ryback B, Rickard BP, Overchuk M, Rizvi I, Rogasevskaia T, Huang KT, Hasan P, Hajnóczky G, Santos JH. Mitochondrial membrane hyperpolarization modulates nuclear DNA methylation and gene expression through phospholipid remodeling. Nat Commun 2025; 16:4029. [PMID: 40301431 PMCID: PMC12041266 DOI: 10.1038/s41467-025-59427-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 04/23/2025] [Indexed: 05/01/2025] Open
Abstract
Maintenance of the mitochondrial inner membrane potential (ΔΨm) is critical for many aspects of mitochondrial function. While ΔΨm loss and its consequences are well studied, little is known about the effects of mitochondrial hyperpolarization. In this study, we used cells deleted of ATP5IF1 (IF1), a natural inhibitor of the hydrolytic activity of the ATP synthase, as a genetic model of increased resting ΔΨm. We found that the nuclear DNA hypermethylates when the ΔΨm is chronically high, regulating the transcription of mitochondrial, carbohydrate and lipid genes. These effects can be reversed by decreasing the ΔΨm and recapitulated in wild-type (WT) cells exposed to environmental chemicals that cause hyperpolarization. Surprisingly, phospholipid changes, but not redox or metabolic alterations, linked the ΔΨm to the epigenome. Sorted hyperpolarized WT and ovarian cancer cells naturally depleted of IF1 also showed phospholipid remodeling, indicating this as an adaptation to mitochondrial hyperpolarization. These data provide a new framework for how mitochondria can impact epigenetics and cellular biology to influence health outcomes, including through chemical exposures and in disease states.
Collapse
Affiliation(s)
- Mateus Prates Mori
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Durham, NC, USA
| | - Oswaldo A Lozoya
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Durham, NC, USA
| | - Ashley M Brooks
- Biostatistics and Computational Biology Branch, Integrative Bioinformatics Support Group, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Durham, NC, USA
| | - Carl D Bortner
- Flow Cytometry Center, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Durham, NC, USA
| | - Cristina A Nadalutti
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Durham, NC, USA
| | - Birgitta Ryback
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Brittany P Rickard
- Curriculum in Toxicology & Environmental Medicine, University of North Carolina (UNC), Chapel Hill, NC, USA
| | - Marta Overchuk
- Department of Biomedical Engineering, North Carolina State University, Raleigh, NC, USA
| | - Imran Rizvi
- Department of Biomedical Engineering, North Carolina State University, Raleigh, NC, USA
- Lineberger Comprehensive Cancer Center, UNC, Chapel Hill, NC, USA
| | | | - Kai Ting Huang
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Prottoy Hasan
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - György Hajnóczky
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Janine H Santos
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Durham, NC, USA.
| |
Collapse
|
6
|
Liu L, Cui H, Dong N, Zhu X, Li S, Ma X, Niu D. Effects of phosphatidylethanolamine on intramuscular fat deposition and key gene identification by transcriptome sequencing in broiler chickens. Poult Sci 2025; 104:104914. [PMID: 39983529 PMCID: PMC11889968 DOI: 10.1016/j.psj.2025.104914] [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: 11/29/2024] [Revised: 02/03/2025] [Accepted: 02/16/2025] [Indexed: 02/23/2025] Open
Abstract
High-intensity breeding of broilers has increased meat yield while reducing meat quality. Properly enhancing intramuscular fat (IMF) content is an effective strategy to improve chicken meat quality and boost consumer preference. Given that the role of phosphatidylethanolamine (PE) in chicken IMF metabolism remains unclear, we investigated the effects of PE on the meat quality, as well as the differentiation and gene expression regulation of chicken intramuscular adipocytes (IMAs). PE supplementation significantly increased the a* value of meat color and reduced shear force (P < 0.05); however, it did not exert a significant effect on the pH value 45 min post-slaughter (P > 0.05). After treating intramuscular adipocytes IMAs with 25, 50, and 100 µM PE, 100 µM PE supplement markedly enhanced lipid deposition and the expression of genes related to adipogenic differentiation. Differentially expressed genes (DEGs) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Set Enrichment Analysis (GESA) were used to identify key genes involved in PE regulation of chicken IMA deposition. A total of 1,182 DEGs were identified, with genes S1PR3, FABP4, PLIN2, APOA1, PPARG and CD36 recognized as hub genes associated with the triglycerides (TG) content of IMAs. PPAR signaling pathway, terpenoid backbone biosynthesis, cytokine-cytokine receptor interaction, and neuroactive ligand-receptor interaction pathway were significantly enriched in PE group. This study reveals the role of PE in chicken IMAs differentiation and lipid deposition, providing a theoretical foundation for further research into the impact of PE on IMF accumulation in broiler chickens.
Collapse
Affiliation(s)
- Lu Liu
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Hangzhou, Zhejiang 311300, China
| | - Houxue Cui
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Hangzhou, Zhejiang 311300, China
| | - Nanxi Dong
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Hangzhou, Zhejiang 311300, China
| | - Xianghua Zhu
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Hangzhou, Zhejiang 311300, China
| | - Sihong Li
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Hangzhou, Zhejiang 311300, China
| | - Xiang Ma
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Hangzhou, Zhejiang 311300, China
| | - Dong Niu
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Hangzhou, Zhejiang 311300, China.
| |
Collapse
|
7
|
Kim SH, Park WY, Kim B, Kim JH, Song G, Park JY, Jiao W, Jung SJ, Ahn KS, Kwak HJ, Um JY. FXR-ApoC2 pathway activates UCP1-mediated thermogenesis by promoting the browning of white adipose tissues. J Biol Chem 2025; 301:108181. [PMID: 39798876 PMCID: PMC11871442 DOI: 10.1016/j.jbc.2025.108181] [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: 08/21/2024] [Revised: 12/17/2024] [Accepted: 12/27/2024] [Indexed: 01/15/2025] Open
Abstract
FXR, encoded by Nh1r4, is a nuclear receptor crucial in regulating bile acid, lipid, and glucose metabolism. Prior research has indicated that activating FXR in the liver and small intestine may offer protection against obesity and metabolic diseases. This study demonstrates the essential role of the FXR-ApoC2 pathway in promoting the browning of white adipose tissue (WAT). Increased FXR by treatment with the FXR agonist farnesol upregulated beige adipocyte markers, including UCP1, PGC1α, and PRDM16, and increased the FXR target gene, ApoC2, in beige adipocytes and cold-exposed mice. However, these effects were not observed in mature white adipocytes. Remarkably, the knockdown of FXR results in a significantly reduced expression of UCP1, PGC1α, PRDM16, and ApoC2 in beige adipocytes. While studying the interaction between the nuclear receptor RXRα and FXR in transcription regulation, it was found that the knockdown of RXRα did not control the expression of FXR under beige adipogenesis. We further investigated whether the expression of beige-related markers could be altered under ApoC2 overexpression to ascertain the mechanism of action of FXR in relation to ApoC2 regulation. The overexpression of ApoC2 in both preadipocytes and beige adipocytes led to a significant increase in the expression of UCP1 and PGC1α. These results indicate that the FXR-mediated ApoC2 pathway is essential in the browning of WAT by inducing beige adipogenesis from preadipocytes.
Collapse
Affiliation(s)
- Sang Hee Kim
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Woo Yong Park
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea; Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Beomsu Kim
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Jin-Hyung Kim
- Department of Biomedical and Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
| | - Gahee Song
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea; Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea; Kyung Hee Institute of Convergence Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Ja Yeon Park
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Wenjun Jiao
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Se Jin Jung
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea; Kyung Hee Institute of Convergence Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hyun Jeong Kwak
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul Republic of Korea
| | - Jae-Young Um
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea; Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea; Kyung Hee Institute of Convergence Korean Medicine, Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
8
|
Xu P, Zhu Z, Zhou R, Wei Y, Xu F, Shen L, Wang Z, Xue Y, Yu X, Zhao J, Yan J, Jin Y, Fang P, Shang W. Huanglian-Jiedu decoction promotes adipose thermogenesis in obese mice by suppressing the expression of HDAC3. JOURNAL OF ETHNOPHARMACOLOGY 2025; 336:118760. [PMID: 39216772 DOI: 10.1016/j.jep.2024.118760] [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: 05/13/2024] [Revised: 08/06/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Huanglian Jiedu Decoction (HLJDD) is an ancient formula of traditional Chinese medicine that is commonly utilized in a range of disorders, and it has been shown to have pharmacological effects on glucose and lipid metabolism. However, the specific mechanism of HLJDD for the treatment of obesity and related metabolic disorders remains to be further investigated. AIM OF THE STUDY It has been thought that encouraging adipose thermogenesis to raise the body's energy expenditure is a useful tactic for improving metabolic abnormalities and losing weight. In this study, we investigated the ability and underlying mechanisms of HLJDD to regulate fat cell thermogenesis to improve energy expenditure in obesity. METHODS The obese mouse model was established on a high-fat diet for 12 weeks. All mice were divided into NC, HFD, HFD with HLJDD of a low dose (2.25 g/kg/d), and HFD with HLJDD of a high dose (4.5 g/kg/d) groups and kept for 4 weeks. In vitro experiments were conducted to evaluate the effects of 5% and 10% HLJDD-containing serum on differentiated 3T3-L1 cells and HDAC3-knocking-down 3T3-L1 cells. RESULTS The results showed that HLJDD treatment significantly improved glucose and insulin tolerance and decreased the adipocyte radius of WATs, as well as increased energy consumption in obese mice. Besides, HLJDD treatment dramatically increased the levels of thermogenic genes UCP-1 and PGC-1α while suppressing HDAC3 levels in WATs and 3T3-L1 adipocytes. Importantly, the effects of HLJDD on PGC-1α and UCP-1 were blocked in HDAC3 knockdown adipocytes. CONCLUSIONS Therefore, these results suggest that HLJDD enhanced adipose thermogenesis and improved energy expenditure by inhibiting HDAC3, thereby increasing UCP-1 and PGC-1α expression. These findings amplified the mechanisms of HLJDD and its potential to treat obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Pingyuan Xu
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ziwei Zhu
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ruonan Zhou
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yaping Wei
- Department of Endocrinology, Changzhou Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Changzhou China
| | - Fangyuan Xu
- Department of Endocrinology, Changzhou Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Changzhou China
| | - Lixuan Shen
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ziwei Wang
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yingying Xue
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xizhong Yu
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Juan Zhao
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jing Yan
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yu Jin
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Penghua Fang
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Wenbin Shang
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
9
|
Chen S, Nie K, Wang H, Gao Y, Jiang X, Su H, Wang Z, Tang Y, Lu F, Dong H, Li J. Wu-Mei-Wan enhances brown adipose tissue function and white adipose browning in obese mice via upregulation of HSF1. Chin Med 2025; 20:1. [PMID: 39754217 PMCID: PMC11697821 DOI: 10.1186/s13020-024-01053-2] [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: 08/28/2024] [Accepted: 12/25/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND This research aims to explore the anti-obesity potential of Wu-Mei-Wan (WMW), particularly its effects on adipose tissue regulation in obese mice induced by a high-fat diet (HFD). The study focuses on understanding the role of heat shock factor 1 (HSF1) in mediating these effects. METHODS HFD-induced obese mice were treated with WMW. Body weight, food intake, and histopathological analysis of adipose tissue were conducted. Brown adipose tissue (BAT) activity was evaluated using Positron Emission Tomography, and ultrastructural changes were examined via transmission electron microscopy. Proteomic analysis identified targets of WMW in obesity treatment. HSF1 expression was inhibited to confirm its role. Molecular docking studied interactions between WMW and HSF1. Short-chain fatty acids (SCFAs) in the intestines were measured to determine if WMW's effects on HSF1 are mediated through SCFAs. Protein expression was assessed using western blot, immunohistochemistry, immunofluorescence and RT-qPCR were employed to detect the mRNA levels. Statistical analyses included t-tests, ANOVA, and non-parametric tests like the Mann-Whitney U test or Kruskal-Wallis test. RESULTS WMW significantly mitigates the adverse effects of a HFD on body weight and glucose metabolism in obese mice. Both low-dose WMW and high-dose WMW treatments led to reduced weight gain and improved glucose tolerance, with low-dose WMW showing more pronounced effects. WMW also reversed structural damage in BAT, enhancing mitochondrial integrity and thermogenic function, particularly at the low dose. Additionally, WMW treatment promoted the browning of WAT, evidenced by increased expression of key thermogenic proteins such as UCP1 and PGC-1α. The increase in HSF1 expression in both BAT and WAT, observed with WMW treatment, was crucial for these beneficial effects, as inhibition of HSF1 negated the positive outcomes. Furthermore, WMW treatment led to elevated levels of short-chain fatty acids SCFAs in the intestines, which are associated with increased HSF1 expression. CONCLUSIONS WMW represents a potent therapeutic strategy for obesity, promoting metabolic health and beneficial modulation of adipose tissue through an HSF1-dependent pathway.
Collapse
Affiliation(s)
- Shen Chen
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Kexin Nie
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Hongzhan Wang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Yang Gao
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Xinyue Jiang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Hao Su
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Zhi Wang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Yueheng Tang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Fuer Lu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Hui Dong
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Jingbin Li
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| |
Collapse
|
10
|
Hidrobo MS, Höring M, Brunner S, Liebisch G, Schweizer S, Klingenspor M, Schreiber R, Zechner R, Burkhardt R, Ecker J. Cold-induced phosphatidylethanolamine synthesis in liver and brown adipose tissue of mice. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159562. [PMID: 39214167 DOI: 10.1016/j.bbalip.2024.159562] [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: 04/05/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Increasing energy expenditure in brown adipose (BAT) tissue by cold-induced lipolysis is discussed as a potential strategy to counteract imbalanced lipid homeostasis caused through unhealthy lifestyle and cardiometabolic disease. Yet, it is largely unclear how liberated fatty acids (FA) are metabolized. We investigated the liver and BAT lipidome of mice housed for 1 week at thermoneutrality, 23 °C and 4 °C using quantitative mass spectrometry-based lipidomics. Housing at temperatures below thermoneutrality triggered the generation of phosphatidylethanolamine (PE) in both tissues. Particularly, the concentrations of PE containing polyunsaturated fatty acids (PUFA) in their acyl chains like PE 18:0_20:4 were increased at cold. Investigation of the plasma's FA profile using gas chromatography coupled to mass spectrometry revealed a negative correlation of PUFA with unsaturated PE in liver and BAT indicating a flux of FA from the circulation into these tissues. Beta-adrenergic stimulation elevated intracellular levels of PE 38:4 and PE 40:6 in beige wildtype adipocytes, but not in adipose triglyceride lipase (ATGL)-deficient cells. These results imply an induction of PE synthesis in liver, BAT and thermogenic adipocytes after activation of the beta-adrenergic signaling cascade.
Collapse
Affiliation(s)
- Maria Soledad Hidrobo
- ZIEL Institute for Food & Health, Research Group Lipid Metabolism, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| | - Marcus Höring
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Sarah Brunner
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Sabine Schweizer
- ZIEL Institute for Food & Health, Research Group Lipid Metabolism, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| | - Martin Klingenspor
- Chair of Molecular Nutritional Medicine, TUM School of Life Sciences, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| | - Renate Schreiber
- Institute of Molecular Biosciences, University of Graz, Heinrichstraße 31/2, 8010 Graz, Austria
| | - Rudolf Zechner
- Institute of Molecular Biosciences, University of Graz, Heinrichstraße 31/2, 8010 Graz, Austria
| | - Ralph Burkhardt
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Josef Ecker
- ZIEL Institute for Food & Health, Research Group Lipid Metabolism, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany; Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany.
| |
Collapse
|
11
|
AKYÜREK F, TUNCEZ AKYÜREK F, ŞENGÜL BAĞ F. Relationship between uncoupling protein 1 (UCP1) levels and psoriasis. Turk J Med Sci 2024; 55:215-222. [PMID: 40104293 PMCID: PMC11913510 DOI: 10.55730/1300-0144.5960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 02/18/2025] [Accepted: 12/30/2024] [Indexed: 03/20/2025] Open
Abstract
Background/aim Psoriasis is a common chronic autoimmune skin disease. Comorbidities increase the mortality risk of the disease. The aim of this study was to investigate the changes in uncoupling protein 1 (UCP1) level in psoriasis patients and evaluate its possible role in the pathogenesis of the disease, focusing on disease severity (Psoriasis Area and Severity Index), dyslipidemia, inflammation, and cardiovascular risk. Materials and methods This study included 30 psoriasis patients and 30 healthy individuals as a control group. Serum UCP1 was measured using an ELISA test kit. The laboratory results of psoriasis patients and healthy controls were compared. Results UCP1 level was a significant candidate marker for the prediction of psoriatic disease (AUC: 0.708, 95% CI: 0.577-0.819, p = 0.002) with sensitivity of 66.67%, specificity of 76.67%, negative predictive value of 69.7%, and positive predictive value of 74.1%. Simple logistic regression analysis showed that an individual with a UCP1 value below 7.561 had a 73% lower probability (OR: 0.27, 95% CI: 0.08-0.94, p = 0.039) of developing psoriasis than an individual with a UCP1 value above 7.561. Among the biochemical parameters, the high-sensitivity C-reactive protein and triglyceride levels of the patients were significantly higher compared to those of the healthy controls while their high-density lipoprotein levels were lower. Conclusion According to the sensitivity (66.67%) and specificity (76.67%) of UCP1, it may be a valuable candidate marker in the diagnosis of psoriasis patients in symptomatic and asymptomatic phases. Further work is needed to substantiate these findings.
Collapse
Affiliation(s)
- Fikret AKYÜREK
- Department of Medical Biochemistry, Faculty of Medicine, Selçuk University, Konya,
Turkiye
| | - Fatma TUNCEZ AKYÜREK
- Department of Dermatology, Faculty of Medicine, Selçuk University, Konya,
Turkiye
| | - Fatma ŞENGÜL BAĞ
- Department of Biochemistry, Faculty of Pharmacy, Adıyaman University, Adıyaman,
Turkiye
| |
Collapse
|
12
|
Yang Y, Luo L, Li Y, Shi X, Li C, Chai J, Jiang S, Zheng R. Succinic Acid Improves the Metabolism of High-Fat Diet-Induced Mice and Promotes White Adipose Browning. Nutrients 2024; 16:3828. [PMID: 39599615 PMCID: PMC11597198 DOI: 10.3390/nu16223828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
Succinic acid plays a crucial role as an essential intermediate in the mitochondrial tricarboxylic acid cycle in mitochondria. In recent years, growing evidence has supported the the important role of succinic acid in fat metabolism. Therefore, we aimed to investigate the effects of succinic acid on adipose tissue metabolism and insulin sensitivity in high-fat diet (HFD)-induced obese mice and try to explore its potential mechanism. We found that the addition of succinic acid (40 mM) to drinking water inhibited the hypertrophy of inguinal white adipose tissue (iWAT) in HFD-induced mice. Furthermore, succinic acid supplementation enhanced insulin sensitivity and improved their glucose tolerance in obese mice. Interestingly, succinic acid supplementation improved lipid metabolism in HFD-fed mice, as shown by decreased serum levels of TG, TC, LDL-C, and increased HDL-C. In addition, succinic acid supplementation increased the expression of browning markers and mitochondria-related genes in iWAT. Further studies showed that the addition of succinic acid to drinking water promotes the browning of iWAT by activating the PI3K-AKT/MAPK signaling pathway. These results suggest that succinic acid has the potential to be used as an effective component for dietary intervention and may, therefore, play an important role in ameliorating and preventing obesity and associated metabolic diseases caused by HFD.
Collapse
Affiliation(s)
- Yuxuan Yang
- Agricultural Ministry Key Laboratory of Swine Breeding and Genetics & Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China; (Y.Y.); (L.L.); (Y.L.); (X.S.); (C.L.); (J.C.); (S.J.)
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Liang Luo
- Agricultural Ministry Key Laboratory of Swine Breeding and Genetics & Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China; (Y.Y.); (L.L.); (Y.L.); (X.S.); (C.L.); (J.C.); (S.J.)
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Yiqi Li
- Agricultural Ministry Key Laboratory of Swine Breeding and Genetics & Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China; (Y.Y.); (L.L.); (Y.L.); (X.S.); (C.L.); (J.C.); (S.J.)
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Xiangda Shi
- Agricultural Ministry Key Laboratory of Swine Breeding and Genetics & Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China; (Y.Y.); (L.L.); (Y.L.); (X.S.); (C.L.); (J.C.); (S.J.)
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Chen Li
- Agricultural Ministry Key Laboratory of Swine Breeding and Genetics & Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China; (Y.Y.); (L.L.); (Y.L.); (X.S.); (C.L.); (J.C.); (S.J.)
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Jin Chai
- Agricultural Ministry Key Laboratory of Swine Breeding and Genetics & Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China; (Y.Y.); (L.L.); (Y.L.); (X.S.); (C.L.); (J.C.); (S.J.)
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Siwen Jiang
- Agricultural Ministry Key Laboratory of Swine Breeding and Genetics & Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China; (Y.Y.); (L.L.); (Y.L.); (X.S.); (C.L.); (J.C.); (S.J.)
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Rong Zheng
- Agricultural Ministry Key Laboratory of Swine Breeding and Genetics & Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China; (Y.Y.); (L.L.); (Y.L.); (X.S.); (C.L.); (J.C.); (S.J.)
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| |
Collapse
|
13
|
Yao Z, Liang S, Chen J, Zhang H, Chen W, Li H. Dietary Lactate Intake and Physical Exercise Synergistically Reverse Brown Adipose Tissue Whitening to Ameliorate Diet-Induced Obesity. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024. [PMID: 39486070 DOI: 10.1021/acs.jafc.4c06899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Physical exercise represents an effective strategy for combating obesity via brown adipose tissue (BAT) activation, but the mechanism remains unclear. In this study, we demonstrated that the cooperation between lactate and adrenoceptor signaling regulated BAT activity during exercise. The lactate receptor GPR81 was highly expressed in the BAT of lean mice, whereas its expression was markedly decreased in obese mice. Notably, the level of GPR81 in BAT could be upregulated by exercise. The blockade of lactate production or GPR81 significantly impaired exercise-induced BAT activation. In addition, dietary lactate intake enhanced the efficacy of physical exercise in alleviating BAT whitening in obese mice, as evidenced by the improved mitochondrial ultrastructure, reduced lipid droplets, increased UCP1 expression, and elevated mitochondrial DNA content. Further data indicated that norepinephrine triggered UCP1 activation through both the cAMP/PKA and Ca2+/CaMK pathways during exercise, while lactate mediated this process via the GPR81-Ca2+/CaMK cascade. Our findings unveil a novel mechanism in the regulation of BAT function by physical exercise, providing a promising lifestyle intervention to improve metabolic health.
Collapse
Affiliation(s)
- Zhijie Yao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Shuxiao Liang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Jinxiang Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Haitao Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
14
|
Farrand ZM, Galbreath KE, Teeter KC. Evidence of Intraspecific Adaptive Variation in the American Pika (Ochotona princeps) on a Continental Scale Using a Target Enrichment and Mitochondrial Genome Skimming Approach. Mol Ecol 2024:e17557. [PMID: 39425616 DOI: 10.1111/mec.17557] [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: 05/31/2024] [Revised: 08/23/2024] [Accepted: 09/26/2024] [Indexed: 10/21/2024]
Abstract
Montane landscapes present an array of abiotic challenges that drive adaptive evolution amongst organisms. These adaptations can promote habitat specialisation, which may heighten the risk of extirpation from environmental change. For example, higher metabolic rates in an endothermic species may contribute to heightened cold tolerance, whilst simultaneously limiting heat tolerance. Here, using the climate-sensitive American pika (Ochotona princeps), we test for evidence of intraspecific adaptive variation amongst environmental gradients across the Intermountain West of North America. We leveraged results from previous studies on pika adaptation to generate a custom nuclear target enrichment design to sequence several hundred candidate genes related to cold, hypoxia and dietary detoxification. We also applied a 'genome skimming' approach to sequence mitochondrial DNA. Using genotype-environment association tests, we identified rare genomic variants associated with elevation and temperature variation amongst populations. Amongst mitochondrial genes, we identified intraspecific variation in selective signals and significant changes to the amino acid property equilibrium constant, which may relate to electron transport chain efficiency. These results illustrate a complex dynamic of adaptive variation amongst O. princeps where lineages and populations have adapted to unique regional conditions. Some of the clearest signals of selection were in a genetic lineage that includes pikas of the Great Basin region, which is also where recent localised extirpations have taken place and highlights the risk of losing adaptive alleles during environmental change.
Collapse
Affiliation(s)
- Zachery M Farrand
- Department of Entomology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Biology, Northern Michigan University, Marquette, Michigan, USA
| | - Kurt E Galbreath
- Department of Biology, Northern Michigan University, Marquette, Michigan, USA
| | - Katherine C Teeter
- Department of Biology, Northern Michigan University, Marquette, Michigan, USA
| |
Collapse
|
15
|
Korbecki J, Bosiacki M, Kupnicka P, Barczak K, Ziętek P, Chlubek D, Baranowska-Bosiacka I. Biochemistry and Diseases Related to the Interconversion of Phosphatidylcholine, Phosphatidylethanolamine, and Phosphatidylserine. Int J Mol Sci 2024; 25:10745. [PMID: 39409074 PMCID: PMC11477190 DOI: 10.3390/ijms251910745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/20/2024] Open
Abstract
Phospholipids are crucial structural components of cells. Phosphatidylcholine and phosphatidylethanolamine (both synthesized via the Kennedy pathway) and phosphatidylserine undergo interconversion. The dysregulation of this process is implicated in various diseases. This paper discusses the role of enzymes involved in the interconversion of phosphatidylcholine, phosphatidylethanolamine, and phosphatidylserine, specifically phosphatidylethanolamine N-methyltransferase (PEMT), phosphatidylserine synthases (PTDSS1 and PTDSS2), and phosphatidylserine decarboxylase (PISD), with a focus on their biochemical properties. Additionally, we describe the effects of the deregulation of these enzymes and their roles in both oncological and non-oncological diseases, including nonalcoholic fatty liver disease (NAFLD), Alzheimer's disease, obesity, insulin resistance, and type II diabetes. Current knowledge on inhibitors of these enzymes as potential therapeutic agents is also reviewed, although in most cases, inhibitors are yet to be developed. The final section of this article presents a bioinformatic analysis using the GEPIA portal to explore the significance of these enzymes in cancer processes.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Anatomy and Histology, Collegium Medicum, University of Zielona Góra, Zyty 28, 65-046 Zielona Góra, Poland;
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (M.B.); (P.K.); (D.C.)
| | - Mateusz Bosiacki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (M.B.); (P.K.); (D.C.)
| | - Patrycja Kupnicka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (M.B.); (P.K.); (D.C.)
| | - Katarzyna Barczak
- Department of Conservative Dentistry and Endodontics, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland;
| | - Paweł Ziętek
- Department of Orthopaedics, Traumatology and Orthopaedic Oncology, Pomeranian Medical University, Unii Lubelskiej 1, 71-252 Szczecin, Poland;
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (M.B.); (P.K.); (D.C.)
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (M.B.); (P.K.); (D.C.)
| |
Collapse
|
16
|
Crivelli SM, Gaifullina A, Chatton JY. Exploring the role of mitochondrial uncoupling protein 4 in brain metabolism: implications for Alzheimer's disease. Front Neurosci 2024; 18:1483708. [PMID: 39381683 PMCID: PMC11459774 DOI: 10.3389/fnins.2024.1483708] [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: 08/20/2024] [Accepted: 09/06/2024] [Indexed: 10/10/2024] Open
Abstract
The brain's high demand for energy necessitates tightly regulated metabolic pathways to sustain physiological activity. Glucose, the primary energy substrate, undergoes complex metabolic transformations, with mitochondria playing a central role in ATP production via oxidative phosphorylation. Dysregulation of this metabolic interplay is implicated in Alzheimer's disease (AD), where compromised glucose metabolism, oxidative stress, and mitochondrial dysfunction contribute to disease progression. This review explores the intricate bioenergetic crosstalk between astrocytes and neurons, highlighting the function of mitochondrial uncoupling proteins (UCPs), particularly UCP4, as important regulators of brain metabolism and neuronal function. Predominantly expressed in the brain, UCP4 reduces the membrane potential in the inner mitochondrial membrane, thereby potentially decreasing the generation of reactive oxygen species. Furthermore, UCP4 mitigates mitochondrial calcium overload and sustains cellular ATP levels through a metabolic shift from mitochondrial respiration to glycolysis. Interestingly, the levels of the neuronal UCPs, UCP2, 4 and 5 are significantly reduced in AD brain tissue and a specific UCP4 variant has been associated to an increased risk of developing AD. Few studies modulating the expression of UCP4 in astrocytes or neurons have highlighted protective effects against neurodegeneration and aging, suggesting that pharmacological strategies aimed at activating UCPs, such as protonophoric uncouplers, hold promise for therapeutic interventions in AD and other neurodegenerative diseases. Despite significant advances, our understanding of UCPs in brain metabolism remains in its early stages, emphasizing the need for further research to unravel their biological functions in the brain and their therapeutic potential.
Collapse
Affiliation(s)
| | | | - Jean-Yves Chatton
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
17
|
Decker ST, Funai K. Mitochondrial membrane lipids in the regulation of bioenergetic flux. Cell Metab 2024; 36:1963-1978. [PMID: 39178855 PMCID: PMC11374467 DOI: 10.1016/j.cmet.2024.07.024] [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: 01/24/2024] [Revised: 06/12/2024] [Accepted: 07/31/2024] [Indexed: 08/26/2024]
Abstract
Oxidative phosphorylation (OXPHOS) occurs through and across the inner mitochondrial membrane (IMM). Mitochondrial membranes contain a distinct lipid composition, aided by lipid biosynthetic machinery localized in the IMM and class-specific lipid transporters that limit lipid traffic in and out of mitochondria. This unique lipid composition appears to be essential for functions of mitochondria, particularly OXPHOS, by its effects on direct lipid-to-protein interactions, membrane properties, and cristae ultrastructure. This review highlights the biological significance of mitochondrial lipids, with a particular spotlight on the role of lipids in mitochondrial bioenergetics. We describe pathways for the biosynthesis of mitochondrial lipids and provide evidence for their roles in physiology, their implications in human disease, and the mechanisms by which they regulate mitochondrial bioenergetics.
Collapse
Affiliation(s)
- Stephen Thomas Decker
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT, USA
| | - Katsuhiko Funai
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
18
|
Deng Y, Dong Y, Zhang S, Feng Y. Targeting mitochondrial homeostasis in the treatment of non-alcoholic fatty liver disease: a review. Front Pharmacol 2024; 15:1463187. [PMID: 39290869 PMCID: PMC11405192 DOI: 10.3389/fphar.2024.1463187] [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: 07/11/2024] [Accepted: 08/20/2024] [Indexed: 09/19/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease worldwide, and its prevalence is rapidly increasing. Antioxidants, lipid-lowering medications, and lifestyle interventions are the most commonly used treatment options for NAFLD, but their efficacy in inhibiting steatosis progression is limited and their long-term ineffectiveness and adverse effects have been widely reported. Therefore, it is important to gain a deeper understanding of the pathogenesis of NAFLD and to identify more effective therapeutic approaches. Mitochondrial homeostasis governs cellular redox biology, lipid metabolism, and cell death, all of which are crucial to control hepatic function. Recent findings have indicated that disruption of mitochondrial homeostasis occurs in the early stage of NAFLD and mitochondrial dysfunction reinforces disease progression. In this review, we summarize the physical roles of the mitochondria and describe their response and dysfunction in the context of NAFLD. We also discuss the drug targets associated with the mitochondria that are currently in the clinical trial phase of exploration. From our findings, we hope that the mitochondria may be a promising therapeutic target for the treatment of NAFLD.
Collapse
Affiliation(s)
- Yalan Deng
- Department of Science and Technology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yuan Dong
- Department of Science and Technology, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Sitian Zhang
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yingmei Feng
- Department of Science and Technology, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
19
|
Fu Y, Wang Q, Tang Z, Liu G, Guan G, Lyu J. Cordycepin Ameliorates High Fat Diet-Induced Obesity by Modulating Endogenous Metabolism and Gut Microbiota Dysbiosis. Nutrients 2024; 16:2859. [PMID: 39275176 PMCID: PMC11396883 DOI: 10.3390/nu16172859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/14/2024] [Accepted: 08/24/2024] [Indexed: 09/16/2024] Open
Abstract
BACKGROUND Numerous metabolic illnesses have obesity as a risk factor. The composition of the gut microbiota and endogenous metabolism are important factors in the onset and progression of obesity. Recent research indicates that cordycepin (CRD), derived from fungi, exhibits anti-inflammatory and antioxidant properties, showing potential in combating obesity. However, further investigation is required to delineate its precise impacts on endogenous metabolism and gut microbiota. METHODS In this work, male C57BL/6J mice were used as models of obesity caused by a high-fat diet (HFD) and given CRD. Mice's colon, liver, and adipose tissues were stained with H&E. Serum metabolome analysis and 16S rRNA sequencing elucidated the effects of CRD on HFD-induced obese mice and identified potential mediators for its anti-obesity effects. RESULTS CRD intervention alleviated HFD-induced intestinal inflammation, improved blood glucose levels, and reduced fat accumulation. Furthermore, CRD supplementation demonstrated the ability to modulate endogenous metabolic disorders by regulating the levels of key metabolites, including DL-2-aminooctanoic acid, inositol, and 6-deoxyfagomine. CRD influenced the abundance of important microbiota such as Parasutterella, Alloprevotella, Prevotellaceae_NK3B31_group, Alistipes, unclassified_Clostridia_vadinBB60_group, and unclassified_Muribaculaceae, ultimately leading to the modulation of endogenous metabolism and the amelioration of gut microbiota disorders. CONCLUSIONS According to our research, CRD therapies show promise in regulating fat accumulation and stabilizing blood glucose levels. Furthermore, through the modulation of gut microbiota composition and key metabolites, CRD interventions have the dual capacity to prevent and ameliorate obesity.
Collapse
Affiliation(s)
- Yifeng Fu
- Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Qiangfeng Wang
- Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Zihan Tang
- Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Gang Liu
- Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Guiping Guan
- Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| | - Jin Lyu
- Department of Pathology, The First People's Hospital of Foshan, Foshan 528000, China
| |
Collapse
|
20
|
Chen X, Han L, Xu W. Dissecting causal relationships between gut microbiota, blood metabolites, and glioblastoma multiforme: a two-sample Mendelian randomization study. Front Microbiol 2024; 15:1403316. [PMID: 39021629 PMCID: PMC11251919 DOI: 10.3389/fmicb.2024.1403316] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 06/18/2024] [Indexed: 07/20/2024] Open
Abstract
Background Given the increasing interest in the role of gut microbiota in glioblastoma multiforme (GBM), our objective was to examine the potential causal relationship between gut microbiota and GBM, as well as the mediating effects of specific metabolites. Methods A bidirectional two-sample Mendelian randomization (MR) analysis was conducted to investigate the associations between 196 microbial taxa and GBM. A two-step MR technique was used to identify significant mediators in this relationship. Subsequently, a mediation analysis was performed to explore and quantify the mediating effects of specific metabolites on the causal relationship between gut microbiota and GBM. Results Five taxa showed significant associations with GBM. Among them, family Victivallaceae [odds ratio (OR): 1.95; 95% confidence interval (CI): 1.21, 3.13; p = 0.005] and genus Lactococcus (OR: 1.81; 95% CI: 1.04, 3.15; p = 0.036) were positively correlated with the risk of GBM, while phylum Cyanobacteria had a protective effect against GBM (OR: 0.45; 95% CI: 0.22, 0.89; p = 0.021). The mediation analysis revealed that the connections among family Victivallaceae, genus Lactococcus, phylum Cyanobacteria and GBM were mediated by Methyl-4-hydroxybenzoate sulfate, phosphoethanolamine and dehydroepiandrosterone sulfate. Each of these accounted for 7.27, 7.98, and 8.65%, respectively. Conclusion Our study provides evidence supporting a potential causal association between certain gut microbiota taxa and GBM. The study highlights the central role of gut microbiota in GBM pathogenesis and their interactions with vital serum metabolites. This paves the way for potential novel therapeutic interventions in GBM management.
Collapse
Affiliation(s)
- Xuan Chen
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, China
| | - Lihui Han
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, China
| | - Wenzhe Xu
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
- Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| |
Collapse
|
21
|
Hu Q, Xu Y, Xiao T, Peng R, Li Z, Xu G, Yu B, Li J, Li ZY, Hou H, Lin Y, Cao J, Liu N, Zha ZG, Gui T, Zhang HT, Cai Y. Trim21 Regulates the Postnatal Development and Thermogenesis of Brown Adipose Tissue. Adv Biol (Weinh) 2024; 8:e2300510. [PMID: 38085135 DOI: 10.1002/adbi.202300510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/08/2023] [Indexed: 03/16/2024]
Abstract
Brown adipose tissue undergoes rapid postnatal development to mature and plays a crucial role in thermoregulation and energy expenditure, which protects against cold and obesity. Herein, it is shown that the expression of Trim21 mRNA level of interscapular brown adipose tissue elevates after birth, and peaks at P14 (postnatal day 14). Trim21 depletion severely impairs the maturation of interscapular brown adipose tissue, decreases the expression of a series of thermogenic genes, and reduces energy expenditure. Consistently, the loss of Trim21 also leads to a suppression of white adipose tissue "browning", in response to cold exposure and a β-adrenergic agonist, CL316,243. In addition, Trim21-/- mice are more prone to high-fat diet-induced obesity compared with the control littermates. Taken together, the study for the first time reveals a critical role of Trim21 in regulating iBAT postnatal development and thermogenesis.
Collapse
Affiliation(s)
- Qinxiao Hu
- Department of Bone and Joint Surgery, the First Affiliated Hospital of Jinan University, Key Laboratory of Regenerative Medicine of Ministry of Education of Jinan University, Guangzhou, Guangdong, 510630, China
| | - Yidi Xu
- Department of Bone and Joint Surgery, the First Affiliated Hospital of Jinan University, Key Laboratory of Regenerative Medicine of Ministry of Education of Jinan University, Guangzhou, Guangdong, 510630, China
| | - Teng Xiao
- Department of Bone and Joint Surgery, the First Affiliated Hospital of Jinan University, Key Laboratory of Regenerative Medicine of Ministry of Education of Jinan University, Guangzhou, Guangdong, 510630, China
| | - Rui Peng
- Department of Bone and Joint Surgery, the First Affiliated Hospital of Jinan University, Key Laboratory of Regenerative Medicine of Ministry of Education of Jinan University, Guangzhou, Guangdong, 510630, China
| | - Zhenwei Li
- Department of Bone and Joint Surgery, the First Affiliated Hospital of Jinan University, Key Laboratory of Regenerative Medicine of Ministry of Education of Jinan University, Guangzhou, Guangdong, 510630, China
- Department of Orthopedics, the Second Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, 233002, China
| | - Guisheng Xu
- Department of Joint and Sports Medicine, The First People's Hospital of Zhaoqing, Zhaoqing, Guangdong, 526000, China
| | - Bo Yu
- Department of Bone and Joint Surgery, the First Affiliated Hospital of Jinan University, Key Laboratory of Regenerative Medicine of Ministry of Education of Jinan University, Guangzhou, Guangdong, 510630, China
| | - Jianping Li
- The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Zhen-Yan Li
- Department of Bone and Joint Surgery, the First Affiliated Hospital of Jinan University, Key Laboratory of Regenerative Medicine of Ministry of Education of Jinan University, Guangzhou, Guangdong, 510630, China
| | - Huige Hou
- Department of Bone and Joint Surgery, the First Affiliated Hospital of Jinan University, Key Laboratory of Regenerative Medicine of Ministry of Education of Jinan University, Guangzhou, Guangdong, 510630, China
| | - Yuning Lin
- Department of Joint and Sports Medicine, The First People's Hospital of Zhaoqing, Zhaoqing, Guangdong, 526000, China
| | - Jiahui Cao
- The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Ning Liu
- Department of Bone and Joint Surgery, the First Affiliated Hospital of Jinan University, Key Laboratory of Regenerative Medicine of Ministry of Education of Jinan University, Guangzhou, Guangdong, 510630, China
| | - Zhen-Gang Zha
- Department of Bone and Joint Surgery, the First Affiliated Hospital of Jinan University, Key Laboratory of Regenerative Medicine of Ministry of Education of Jinan University, Guangzhou, Guangdong, 510630, China
| | - Tao Gui
- Department of Bone and Joint Surgery, the First Affiliated Hospital of Jinan University, Key Laboratory of Regenerative Medicine of Ministry of Education of Jinan University, Guangzhou, Guangdong, 510630, China
| | - Huan-Tian Zhang
- Department of Bone and Joint Surgery, the First Affiliated Hospital of Jinan University, Key Laboratory of Regenerative Medicine of Ministry of Education of Jinan University, Guangzhou, Guangdong, 510630, China
| | - Yuebo Cai
- Department of Bone and Joint Surgery, the First Affiliated Hospital of Jinan University, Key Laboratory of Regenerative Medicine of Ministry of Education of Jinan University, Guangzhou, Guangdong, 510630, China
- Department of Orthopedics, the Affiliated Shunde Hospital of Jinan University, Shunde, Guangdong, 528300, China
| |
Collapse
|
22
|
Mota-Rojas D, Ghezzi MD, Hernández-Ávalos I, Domínguez-Oliva A, Casas-Alvarado A, Lendez PA, Ceriani MC, Wang D. Hypothalamic Neuromodulation of Hypothermia in Domestic Animals. Animals (Basel) 2024; 14:513. [PMID: 38338158 PMCID: PMC10854546 DOI: 10.3390/ani14030513] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/12/2024] Open
Abstract
When an organism detects decreases in their core body temperature, the hypothalamus, the main thermoregulatory center, triggers compensatory responses. These responses include vasomotor changes to prevent heat loss and physiological mechanisms (e.g., shivering and non-shivering thermogenesis) for heat production. Both types of changes require the participation of peripheral thermoreceptors, afferent signaling to the spinal cord and hypothalamus, and efferent pathways to motor and/or sympathetic neurons. The present review aims to analyze the scientific evidence of the hypothalamic control of hypothermia and the central and peripheral changes that are triggered in domestic animals.
Collapse
Affiliation(s)
- Daniel Mota-Rojas
- Neurophysiology, Behavior and Animal Welfare Assessment, DPAA, Universidad Autónoma Metropolitana (UAM), Mexico City 04960, Mexico
| | - Marcelo Daniel Ghezzi
- Animal Welfare Area, Faculty of Veterinary Sciences (FCV), Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPBA), GIB, Tandil 7000, Buenos Aires, Argentina
| | - Ismael Hernández-Ávalos
- Clinical Pharmacology and Veterinary Anesthesia, Biological Sciences Department, FESC, Universidad Nacional Autónoma de México, Cuautitlán 54714, Mexico
| | - Adriana Domínguez-Oliva
- Neurophysiology, Behavior and Animal Welfare Assessment, DPAA, Universidad Autónoma Metropolitana (UAM), Mexico City 04960, Mexico
| | - Alejandro Casas-Alvarado
- Neurophysiology, Behavior and Animal Welfare Assessment, DPAA, Universidad Autónoma Metropolitana (UAM), Mexico City 04960, Mexico
| | - Pamela Anahí Lendez
- Anatomy Area, Faculty of Veterinary Sciences, Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPBA), GIB/CISAPA, Tandil 7000, Buenos Aires, Argentina
| | - María Carolina Ceriani
- Anatomy Area, Faculty of Veterinary Sciences, Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPBA), GIB/CISAPA, Tandil 7000, Buenos Aires, Argentina
| | - Dehua Wang
- School of Life Sciences, Shandong University, Qingdao 266237, China
| |
Collapse
|
23
|
Mori MP, Lozoya O, Brooks AM, Grenet D, Nadalutti CA, Ryback B, Huang KT, Hasan P, Hajnóczky G, Santos JH. Mitochondrial membrane potential regulates nuclear DNA methylation and gene expression through phospholipid remodeling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.12.575075. [PMID: 38260521 PMCID: PMC10802563 DOI: 10.1101/2024.01.12.575075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Maintenance of the mitochondrial inner membrane potential (ΔΨM) is critical for many aspects of mitochondrial function, including mitochondrial protein import and ion homeostasis. While ΔΨM loss and its consequences are well studied, little is known about the effects of increased ΔΨM. In this study, we used cells deleted of ATPIF1, a natural inhibitor of the hydrolytic activity of the ATP synthase, as a genetic model of mitochondrial hyperpolarization. Our data show that chronic ΔΨM increase leads to nuclear DNA hypermethylation, regulating transcription of mitochondria, carbohydrate and lipid metabolism genes. Surprisingly, remodeling of phospholipids, but not metabolites or redox changes, mechanistically links the ΔΨM to the epigenome. These changes were also observed upon chemical exposures and reversed by decreasing the ΔΨM, highlighting them as hallmark adaptations to chronic mitochondrial hyperpolarization. Our results reveal the ΔΨM as the upstream signal conveying the mitochondrial status to the epigenome to regulate cellular biology, providing a new framework for how mitochondria can influence health outcomes in the absence of canonical dysfunction.
Collapse
Affiliation(s)
| | | | - Ashley M. Brooks
- Biostatistics and Computational Biology Branch, Integrative Bioinformatics Support Group, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), 111 TW Alexander drive, Research Triangle Park, NC, 27709
| | - Dagoberto Grenet
- Mechanistic Toxicology Branch, Division of Translational Toxicology
| | | | - Birgitta Ryback
- Dana Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Kai Ting Huang
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, US
| | - Prottoy Hasan
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, US
| | - Gyӧrgy Hajnóczky
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, US
| | - Janine H. Santos
- Mechanistic Toxicology Branch, Division of Translational Toxicology
| |
Collapse
|
24
|
Makio T, Simmen T. Not So Rare: Diseases Based on Mutant Proteins Controlling Endoplasmic Reticulum-Mitochondria Contact (MERC) Tethering. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2024; 7:25152564241261228. [PMID: 39070058 PMCID: PMC11273598 DOI: 10.1177/25152564241261228] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/12/2024] [Accepted: 05/27/2024] [Indexed: 07/30/2024]
Abstract
Mitochondria-endoplasmic reticulum contacts (MERCs), also called endoplasmic reticulum (ER)-mitochondria contact sites (ERMCS), are the membrane domains, where these two organelles exchange lipids, Ca2+ ions, and reactive oxygen species. This crosstalk is a major determinant of cell metabolism, since it allows the ER to control mitochondrial oxidative phosphorylation and the Krebs cycle, while conversely, it allows the mitochondria to provide sufficient ATP to control ER proteostasis. MERC metabolic signaling is under the control of tethers and a multitude of regulatory proteins. Many of these proteins have recently been discovered to give rise to rare diseases if their genes are mutated. Surprisingly, these diseases share important hallmarks and cause neurological defects, sometimes paired with, or replaced by skeletal muscle deficiency. Typical symptoms include developmental delay, intellectual disability, facial dysmorphism and ophthalmologic defects. Seizures, epilepsy, deafness, ataxia, or peripheral neuropathy can also occur upon mutation of a MERC protein. Given that most MERC tethers and regulatory proteins have secondary functions, some MERC protein-based diseases do not fit into this categorization. Typically, however, the proteins affected in those diseases have dominant functions unrelated to their roles in MERCs tethering or their regulation. We are discussing avenues to pharmacologically target genetic diseases leading to MERC defects, based on our novel insight that MERC defects lead to common characteristics in rare diseases. These shared characteristics of MERCs disorders raise the hope that they may allow for similar treatment options.
Collapse
Affiliation(s)
- Tadashi Makio
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Thomas Simmen
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
25
|
Kubatzky KF, Gao Y, Yu D. Post-translational modulation of cell signalling through protein succinylation. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:1260-1285. [PMID: 38213532 PMCID: PMC10776603 DOI: 10.37349/etat.2023.00196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 08/22/2023] [Indexed: 01/13/2024] Open
Abstract
Cells need to adapt their activities to extra- and intracellular signalling cues. To translate a received extracellular signal, cells have specific receptors that transmit the signal to downstream proteins so that it can reach the nucleus to initiate or repress gene transcription. Post-translational modifications (PTMs) of proteins are reversible or irreversible chemical modifications that help to further modulate protein activity. The most commonly observed PTMs are the phosphorylation of serine, threonine, and tyrosine residues, followed by acetylation, glycosylation, and amidation. In addition to PTMs that involve the modification of a certain amino acid (phosphorylation, hydrophobic groups for membrane localisation, or chemical groups like acylation), or the conjugation of peptides (SUMOylation, NEDDylation), structural changes such as the formation of disulphide bridge, protein cleavage or splicing can also be classified as PTMs. Recently, it was discovered that metabolites from the tricarboxylic acid (TCA) cycle are not only intermediates that support cellular metabolism but can also modify lysine residues. This has been shown for acetate, succinate, and lactate, among others. Due to the importance of mitochondria for the overall fitness of organisms, the regulatory function of such PTMs is critical for protection from aging, neurodegeneration, or cardiovascular disease. Cancer cells and activated immune cells display a phenotype of accelerated metabolic activity known as the Warburg effect. This metabolic state is characterised by enhanced glycolysis, the use of the pentose phosphate pathway as well as a disruption of the TCA cycle, ultimately causing the accumulation of metabolites like citrate, succinate, and malate. Succinate can then serve as a signalling molecule by directly interacting with proteins, by binding to its G protein-coupled receptor 91 (GPR91) and by post-translationally modifying proteins through succinylation of lysine residues, respectively. This review is focus on the process of protein succinylation and its importance in health and disease.
Collapse
Affiliation(s)
- Katharina F. Kubatzky
- Department of Infectious Diseases, Medical Faculty Heidelberg, Medical Microbiology and Hygiene, Heidelberg University, 69120 Heidelberg, Germany
- Department of Infectious Diseases, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Yue Gao
- Department of Infectious Diseases, Medical Faculty Heidelberg, Medical Microbiology and Hygiene, Heidelberg University, 69120 Heidelberg, Germany
- Department of Infectious Diseases, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Dayoung Yu
- Department of Infectious Diseases, Medical Faculty Heidelberg, Medical Microbiology and Hygiene, Heidelberg University, 69120 Heidelberg, Germany
- Department of Infectious Diseases, University Hospital Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
26
|
Wong CP, Iwaniec UT, Turner RT. Brown adipose tissue but not tibia exhibits a dramatic response to acute reduction in environmental temperature in growing male mice. Bone Rep 2023; 19:101706. [PMID: 37637756 PMCID: PMC10448410 DOI: 10.1016/j.bonr.2023.101706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/04/2023] [Accepted: 08/07/2023] [Indexed: 08/29/2023] Open
Abstract
Mice are typically housed at room temperature (∼22 °C), which is well below their thermoneutral zone and results in cold stress. Chronic cold stress leads to increased adaptive thermogenesis and reductions in cancellous bone volume and bone marrow adipose tissue mass in long bones of growing mice. There is strong evidence that increased neuronal activity initiates the metabolic response of intrascapular brown adipose tissue (BAT) to cold stress, but it is less clear whether bone is regulated through a similar mechanism. Therefore, we compared the short-term response of BAT and whole tibia to a reduction in environmental temperature. To accomplish this, we transferred a group of 6-week-old male mice from 32 °C to 22 °C housing and sacrificed the mice 24 h later. Age-matched controls were maintained at 32 °C. We then evaluated expression levels of a panel of genes related to adipocyte differentiation and fat metabolism in BAT and tibia, and a panel of genes related to bone metabolism in tibia. The decrease in housing temperature resulted in changes in expression levels for 47/86 genes related to adipocyte differentiation and fat metabolism in BAT, including 9-fold and 17-fold increases in Ucp1 and Dio2, respectively. In contrast, only 1/86 genes related to adipocyte differentiation and fat metabolism and 4/84 genes related to bone metabolism were differentially expressed in tibia. These findings suggest that bone, although innervated with sensory and sympathetic neurons, does not respond as rapidly as BAT to changes in environmental temperature.
Collapse
Affiliation(s)
- Carmen P. Wong
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, USA
| | - Urszula T. Iwaniec
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, USA
- Center for Healthy Aging Research, Oregon State University, Corvallis, OR 97331, USA
| | - Russell T. Turner
- Skeletal Biology Laboratory, School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, USA
- Center for Healthy Aging Research, Oregon State University, Corvallis, OR 97331, USA
| |
Collapse
|
27
|
Ji T, Fang B, Zhang M, Liu Y. Succinate Enhances Lipolysis and Decreases Adipocytes Size in Both Subcutaneous and Visceral Adipose Tissue from High-Fat-Diet-Fed Obese Mice. Foods 2023; 12:4285. [PMID: 38231706 DOI: 10.3390/foods12234285] [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: 11/08/2023] [Revised: 11/15/2023] [Accepted: 11/22/2023] [Indexed: 01/19/2024] Open
Abstract
Obesity is a risk factor for many chronic diseases related to the overexpansion of adipose tissue during obesity, leading to metabolic dysfunction and ectopic lipids. Previous studies reported a close relationship between succinate and obesity and its co-morbidities, and studies have also reported on its anti-obesity potential. To confirm its efficacy in obesity interventions, we supplemented mice with obesity induced by a high-fat diet with succinate (1.5% m/v in drinking water) for 11 weeks without changing the diet. After succinate supplementation, the changes in body weight, adipose tissue deposition, glucose tolerance, energy expenditure and lipid metabolism were evaluated. It was found that succinate supplementation significantly decreased subcutaneous adipose tissue (HFD: 4239.3 ± 211.2 mg; HFD-SA: 3268.9 ± 265.7 mg. p < 0.05), triglyceride contents (decreased by 1.53 mmol/g and 0.39 mmol/g in eWAT and ingWAT, respectively, p < 0.05) and NEFA (decreased by 1.41 μmol/g and 1.31 μmol/g in eWAT and ingWAT, respectively, p < 0.05). The adipocytes' sizes all significantly decreased in both subcutaneous and visceral adipose tissue (the proportion of adipocytes with diameters larger than 100 μm in eWAT and ingWAT decreased by 16.83% and 11.96%, respectively. p < 0.05). Succinate significantly enhanced lipolysis in adipose tissue (eWAT: Adrb3, Hsl and Plin1; ingWAT: Hsl and CPT1a; p < 0.05), whereas the expression of lipogenesis-related genes remained unchanged (p > 0.05). Succinate supplementation also enhanced the activity of BAT by stimulating the expression of Ucp1 and Cidea (p < 0.05). Our results reported that succinate has a potential beneficial effect on obesity pathogenesis but cannot efficiently decrease bodyweight.
Collapse
Affiliation(s)
- Tengteng Ji
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China
| | - Bing Fang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China
| | - Ming Zhang
- School of Food Science and Chemical Engineering, Beijing Technology and Business University, Beijing 100048, China
| | - Yaqiong Liu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China
| |
Collapse
|
28
|
Cui X, Liu H, Shi T, Zhao Q, Li F, Lv W, Yu C, Huang H, Tang QQ, Pan D. IFI27 Integrates Succinate and Fatty Acid Oxidation to Promote Adipocyte Thermogenic Adaption. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301855. [PMID: 37544897 PMCID: PMC10558685 DOI: 10.1002/advs.202301855] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 06/16/2023] [Indexed: 08/08/2023]
Abstract
Mitochondria are the pivot organelles to control metabolism and energy homeostasis. The capacity of mitochondrial metabolic adaptions to cold stress is essential for adipocyte thermogenesis. How brown adipocytes keep mitochondrial fitness upon a challenge of cold-induced oxidative stress has not been well characterized. This manuscript shows that IFI27 plays an important role in cristae morphogenesis, keeping intact succinate dehydrogenase (SDH) function and active fatty acid oxidation to sustain thermogenesis in brown adipocytes. IFI27 protein interaction map identifies SDHB and HADHA as its binding partners. IFI27 physically links SDHB to chaperone TNF receptor associated protein 1 (TRAP1), which shields SDHB from oxidative damage-triggered degradation. Moreover, IFI27 increases hydroxyacyl-CoA dehydrogenase trifunctional multienzyme complex subunit alpha (HADHA) catalytic activity in β-oxidation pathway. The reduced SDH level and fatty acid oxidation in Ifi27-knockout brown fat results in impaired oxygen consumption and defective thermogenesis. Thus, IFI27 is a novel regulator of mitochondrial metabolism and thermogenesis.
Collapse
Affiliation(s)
- Xuan Cui
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Haojie Liu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Ting Shi
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Qingwen Zhao
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Feiyan Li
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Wenjing Lv
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Chao Yu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Haiyan Huang
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Qi-Qun Tang
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Dongning Pan
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| |
Collapse
|
29
|
Abstract
Studies of rare human genetic disorders of mitochondrial phospholipid metabolism have highlighted the crucial role that membrane phospholipids play in mitochondrial bioenergetics and human health. The phospholipid composition of mitochondrial membranes is highly conserved from yeast to humans, with each class of phospholipid performing a specific function in the assembly and activity of various mitochondrial membrane proteins, including the oxidative phosphorylation complexes. Recent studies have uncovered novel roles of cardiolipin and phosphatidylethanolamine, two crucial mitochondrial phospholipids, in organismal physiology. Studies on inter-organellar and intramitochondrial phospholipid transport have significantly advanced our understanding of the mechanisms that maintain mitochondrial phospholipid homeostasis. Here, we discuss these recent advances in the function and transport of mitochondrial phospholipids while describing their biochemical and biophysical properties and biosynthetic pathways. Additionally, we highlight the roles of mitochondrial phospholipids in human health by describing the various genetic diseases caused by disruptions in their biosynthesis and discuss advances in therapeutic strategies for Barth syndrome, the best-studied disorder of mitochondrial phospholipid metabolism.
Collapse
Affiliation(s)
- Alaumy Joshi
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA
| | - Travis H. Richard
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA
| | - Vishal M. Gohil
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
30
|
Cho YK, Lee S, Lee J, Doh J, Park JH, Jung YS, Lee YH. Lipid remodeling of adipose tissue in metabolic health and disease. Exp Mol Med 2023; 55:1955-1973. [PMID: 37653032 PMCID: PMC10545718 DOI: 10.1038/s12276-023-01071-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/30/2023] [Accepted: 06/05/2023] [Indexed: 09/02/2023] Open
Abstract
Adipose tissue is a dynamic and metabolically active organ that plays a crucial role in energy homeostasis and endocrine function. Recent advancements in lipidomics techniques have enabled the study of the complex lipid composition of adipose tissue and its role in metabolic disorders such as obesity, diabetes, and cardiovascular disease. In addition, adipose tissue lipidomics has emerged as a powerful tool for understanding the molecular mechanisms underlying these disorders and identifying bioactive lipid mediators and potential therapeutic targets. This review aims to summarize recent lipidomics studies that investigated the dynamic remodeling of adipose tissue lipids in response to specific physiological changes, pharmacological interventions, and pathological conditions. We discuss the molecular mechanisms of lipid remodeling in adipose tissue and explore the recent identification of bioactive lipid mediators generated in adipose tissue that regulate adipocytes and systemic metabolism. We propose that manipulating lipid-mediator metabolism could serve as a therapeutic approach for preventing or treating obesity-related metabolic diseases.
Collapse
Affiliation(s)
- Yoon Keun Cho
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sumin Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jaewon Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Junsang Doh
- Department of Materials Science and Engineering, Research Institute of Advanced Materials, Institute of Engineering Research, Bio-MAX Institute, Soft Foundry Institute, Seoul National University, Seoul, Republic of Korea
| | - Joo-Hong Park
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Young-Suk Jung
- College of Pharmacy, Pusan National University, Busan, Republic of Korea
| | - Yun-Hee Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
31
|
Bienboire-Frosini C, Wang D, Marcet-Rius M, Villanueva-García D, Gazzano A, Domínguez-Oliva A, Olmos-Hernández A, Hernández-Ávalos I, Lezama-García K, Verduzco-Mendoza A, Gómez-Prado J, Mota-Rojas D. The Role of Brown Adipose Tissue and Energy Metabolism in Mammalian Thermoregulation during the Perinatal Period. Animals (Basel) 2023; 13:2173. [PMID: 37443971 DOI: 10.3390/ani13132173] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/21/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Hypothermia is one of the most common causes of mortality in neonates, and it could be developed after birth because the uterus temperature is more elevated than the extrauterine temperature. Neonates use diverse mechanisms to thermoregulate, such as shivering and non-shivering thermogenesis. These strategies can be more efficient in some species, but not in others, i.e., altricials, which have the greatest difficulty with achieving thermoneutrality. In addition, there are anatomical and neurological differences in mammals, which may present different distributions and amounts of brown fat. This article aims to discuss the neuromodulation mechanisms of thermoregulation and the importance of brown fat in the thermogenesis of newborn mammals, emphasizing the analysis of the biochemical, physiological, and genetic factors that determine the distribution, amount, and efficiency of this energy resource in newborns of different species. It has been concluded that is vital to understand and minimize hypothermia causes in newborns, which is one of the main causes of mortality in neonates. This would be beneficial for both animals and producers.
Collapse
Affiliation(s)
- Cécile Bienboire-Frosini
- Department of Molecular Biology and Chemical Communication, Research Institute in Semiochemistry and Applied Ethology (IRSEA), 84400 Apt, France
| | - Dehua Wang
- School of Life Sciences, Shandong University, Qingdao 266237, China
| | - Míriam Marcet-Rius
- Animal Behaviour and Welfare Department, Research Institute in Semiochemistry and Applied Ethology (IRSEA), 84400 Apt, France
| | - Dina Villanueva-García
- Division of Neonatology, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico
| | - Angelo Gazzano
- Department of Veterinary Sciences, University of Pisa, 56124 Pisa, Italy
| | - Adriana Domínguez-Oliva
- Neurophysiology, Behavior and Animal Welfare Assessment, DPAA, Universidad Autónoma Metropolitana, Xochimilco Campus, Mexico City 04960, Mexico
| | - Adriana Olmos-Hernández
- Division of Biotechnology-Bioterio and Experimental Surgery, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra (INR-LGII), Mexico City 14389, Mexico
| | - Ismael Hernández-Ávalos
- Clinical Pharmacology and Veterinary Anesthesia, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México (UNAM), Cuautitlán Izcalli 54714, Mexico
| | - Karina Lezama-García
- Neurophysiology, Behavior and Animal Welfare Assessment, DPAA, Universidad Autónoma Metropolitana, Xochimilco Campus, Mexico City 04960, Mexico
| | - Antonio Verduzco-Mendoza
- Division of Biotechnology-Bioterio and Experimental Surgery, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra (INR-LGII), Mexico City 14389, Mexico
| | - Jocelyn Gómez-Prado
- Neurophysiology, Behavior and Animal Welfare Assessment, DPAA, Universidad Autónoma Metropolitana, Xochimilco Campus, Mexico City 04960, Mexico
| | - Daniel Mota-Rojas
- Neurophysiology, Behavior and Animal Welfare Assessment, DPAA, Universidad Autónoma Metropolitana, Xochimilco Campus, Mexico City 04960, Mexico
| |
Collapse
|
32
|
Pistritu DV, Vasiliniuc AC, Vasiliu A, Visinescu EF, Visoiu IE, Vizdei S, Martínez Anghel P, Tanca A, Bucur O, Liehn EA. Phospholipids, the Masters in the Shadows during Healing after Acute Myocardial Infarction. Int J Mol Sci 2023; 24:8360. [PMID: 37176067 PMCID: PMC10178977 DOI: 10.3390/ijms24098360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
Phospholipids are major components of cell membranes with complex structures, high heterogeneity and critical biological functions and have been used since ancient times to treat cardiovascular disease. Their importance and role were shadowed by the difficulty or incomplete available research methodology to study their biological presence and functionality. This review focuses on the current knowledge about the roles of phospholipids in the pathophysiology and therapy of cardiovascular diseases, which have been increasingly recognized. Used in singular formulation or in inclusive combinations with current drugs, phospholipids proved their positive and valuable effects not only in the protection of myocardial tissue, inflammation and fibrosis but also in angiogenesis, coagulation or cardiac regeneration more frequently in animal models as well as in human pathology. Thus, while mainly neglected by the scientific community, phospholipids present negligible side effects and could represent an ideal target for future therapeutic strategies in healing myocardial infarction. Acknowledging and understanding their mechanisms of action could offer a new perspective into novel therapeutic strategies for patients suffering an acute myocardial infarction, reducing the burden and improving the general social and economic outcome.
Collapse
Affiliation(s)
- Dan-Valentin Pistritu
- Victor Babes’ National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania
| | | | - Anda Vasiliu
- Victor Babes’ National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania
| | - Elena-Florentina Visinescu
- Faculty of Human Medicine, Carol Davila University of Medicine and Pharmacy, 37 Dionisie Lupu Street, 020021 Bucharest, Romania
| | - Ioana-Elena Visoiu
- Faculty of Human Medicine, Carol Davila University of Medicine and Pharmacy, 37 Dionisie Lupu Street, 020021 Bucharest, Romania
| | - Smaranda Vizdei
- Faculty of Human Medicine, Carol Davila University of Medicine and Pharmacy, 37 Dionisie Lupu Street, 020021 Bucharest, Romania
| | - Paula Martínez Anghel
- Victor Babes’ National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania
- Business Academy Aarhus, 30 Sønderhøj, 8260 Viby J, Denmark
| | - Antoanela Tanca
- Victor Babes’ National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania
- Faculty of Human Medicine, Carol Davila University of Medicine and Pharmacy, 37 Dionisie Lupu Street, 020021 Bucharest, Romania
| | - Octavian Bucur
- Victor Babes’ National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania
- Viron Molecular Medicine Institute, 201 Washington Street, Boston, MA 02108, USA
| | - Elisa Anamaria Liehn
- Victor Babes’ National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania
- Institute for Molecular Medicine, University of Southern Denmark, 25 J.B Winsløws Vej, 5230 Odense, Denmark
- National Heart Center Singapore, 5 Hospital Dr., Singapore 169609, Singapore
| |
Collapse
|