1
|
Hao Y, Hu J, Zhang Z, Guan Q, Wang J, Tao Y, Cheng J, Fan Y. Sirt6 deficiency exacerbates angiotensin II-induced lipid nephrotoxicity by affecting PLD6-derived cardiolipin metabolism in podocytes. Cell Signal 2025; 133:111858. [PMID: 40355014 DOI: 10.1016/j.cellsig.2025.111858] [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: 12/21/2024] [Revised: 04/22/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND AND AIMS Perturbation of cardiolipin (CL) metabolism is associated with lipid nephrotoxicity. Recent findings provide new insights into the roles of CL-modulating proteins as critical determinants of podocyte function in chronic kidney disease (CKD). We previously demonstrated that Sirtuin 6 (Sirt6) is a compelling target inhibiting Angiotensin II (Ang II)-induced lipid dysregulation in podocytes. However, whether Sirt6 regulates podocyte CL metabolism is unknown. METHODS Renal biopsy specimens of patients with hypertensive nephropathy (HN) were used in this study. Podocyte Sirt6-specific knockout mice were generated using the Cre-loxP system. The effect of Sirt6 on mitochondrial CL metabolism, especially the peroxidation and hydrolysis of CL, was investigated in Ang II-infusion mice and Ang II-induced cultured podocytes. RESULTS Sirt6 and outer mitochondrial membrane protein phospholipase D family member 6 (PLD6) were decreased in the glomeruli of patients with HN. Ang II downregulated Sirt6 and PLD6 expression in podocytes in vitro and in vivo. Podocyte-specific deletion of Sirt6 exacerbated lipid droplets formation, CL accumulation and peroxidation, aggravated Ang II-induced mitochondrial dysfunction and cell apoptosis. Mechanically, Sirt6 maintained podocyte CL homeostasis, at least in part through PLD6 signaling-mediated CL metabolism. In addition, cardiolipin antioxidant Szeto-Schiller Peptide 31 (SS-31) treatment inhibited Ang II-induced lipid accumulation and CL peroxidation in podocytes. CONCLUSIONS Our findings shed light on Sirt6's regulatory mechanisms on podocyte CL metabolism and suggest exploiting the Sirt6-PLD6 axis as a potential therapeutic target for protecting against lipid nephrotoxicity.
Collapse
Affiliation(s)
- Yiqun Hao
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Jijia Hu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Zongwei Zhang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Qian Guan
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Juan Wang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yu Tao
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jing Cheng
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Yanqin Fan
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
2
|
Ko JH, Jung TW, Pyo MK, Lim DS, Gwon HJ, Abd El-Aty AM, Yağan R, Shin JW, Lee MY, Song JH, Jeong JH. Senkyunolide A mitigates lipid deposition in hyperlipidemic hepatocytes through SIRT6-mediated suppression of oxidative stress and fetuin-A. Biochem Biophys Res Commun 2025; 771:152046. [PMID: 40403679 DOI: 10.1016/j.bbrc.2025.152046] [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: 05/14/2025] [Accepted: 05/17/2025] [Indexed: 05/24/2025]
Abstract
Senkyunolide A (SenA), a phthalide compound isolated from celery seed essential oil, is known for its anti-inflammatory and antioxidant properties in various disease models. However, its impact on hepatic lipid metabolism and associated molecular mechanisms remain unclear. This study investigated the protective effects of SenA against lipotoxicity-induced hepatic steatosis and explored its underlying pathways. Lipid accumulation was assessed via Oil Red O staining, whereas protein expression was analyzed via Western blotting. Apoptosis was evaluated through TUNEL staining, caspase-3 activity, and cell viability assays. Oxidative stress was measured via DCFDA-based ROS detection and assays for MDA and H2O2. The role of SIRT6 was examined through siRNA-mediated knockdown. SenA treatment significantly reduced lipid deposition, apoptosis, oxidative stress, and fetuin-A expression in palmitate-treated hepatocytes. It also increased the expression of SIRT6 and Nrf2, two key regulators of metabolic and redox homeostasis. Silencing SIRT6 diminished these protective effects, indicating its essential role in mediating SenA activity. These findings suggest that SenA mitigates hepatic steatosis by modulating oxidative stress and metabolic dysfunction via SIRT6/Nrf2 signaling. As a natural bioactive compound, SenA offers promise for the development of safer therapeutic strategies for managing nonalcoholic fatty liver disease (NAFLD), which has broader relevance to metabolic health.
Collapse
Affiliation(s)
- Jun Hwi Ko
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - Tae Woo Jung
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Min Kyung Pyo
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - Do Su Lim
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - Hyeon Ji Gwon
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - A M Abd El-Aty
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt; Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum, 25240, Turkey.
| | - Rıdvan Yağan
- Dr. Filiz Dolunay Family Health Center Unit Number:61 Erzurum, Turkey
| | - Jong Wook Shin
- Department of Internal Medicine, Division of Allergy and Respiratory Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Moo Yeol Lee
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Jin-Ho Song
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea.
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea.
| |
Collapse
|
3
|
Guo S, Zhao Y, Wang Y, Lin M, Luan Q, Hu Z, Zhao X, Tian X, Wang Z, Yao J. OTUB1 enhances fatty acid oxidation in APAP-induced liver injury by mediating ACSL5 deubiquitination. Biochem Pharmacol 2025; 237:116957. [PMID: 40280245 DOI: 10.1016/j.bcp.2025.116957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/22/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
Overdosing on acetaminophen (APAP) is the primary cause of drug-induced liver injury. Recent studies have demonstrated that dysregulated lipid metabolism, particularly decreased fatty acid oxidation (FAO), is a key contributor to APAP-induced acute liver injury (AILI). OTU domain-containing ubiquitin aldehyde-binding protein 1 (OTUB1), a crucial member of the OTU deubiquitinase family, has been involved in the metabolic progression of multiple diseases. Nevertheless, its involvement in AILI as well as FAO remains unclear. Here, we aimed to elucidate the effects of OTUB1 on the regulation of FAO in AILI. Our investigation revealed decreased OTUB1 expression in AILI. OTUB1 overexpression not only alleviated liver injury but also improved FAO in vivo and in vitro. Conversely, opposite biochemical changes were observed in hepatocytes with OTUB1 knockdown. Mechanistically, long-chain acyl-CoA synthase 5 (ACSL5), which plays a crucial role in regulating FAO, was identified as a novel substrate of OTUB1 in AILI via mass spectrometry analysis. OTUB1 interacts with ACSL5 and promotes its deubiquitination and stability. Moreover, the protective effect of OTUB1 on FAO in AILI occurred via the deubiquitination of ACSL5. Overall, the present study revealed that the OTUB1-ACSL5 axis plays an essential role in regulating FAO during AILI progression and might be a novel target for therapeutic intervention.
Collapse
Affiliation(s)
- Shuyu Guo
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Yan Zhao
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Yue Wang
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Musen Lin
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qinrong Luan
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Zhehao Hu
- Department of Pharmacology, Dalian Medical University, Dalian, China
| | - Xuzi Zhao
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaofeng Tian
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Zhecheng Wang
- Department of Pharmacology, Dalian Medical University, Dalian, China.
| | - Jihong Yao
- Department of Pharmacology, Dalian Medical University, Dalian, China.
| |
Collapse
|
4
|
Zhang B, Fan Z, Liu X, Wu Y, Cheng L, Wang L, Liu H. Bisphenol AF induces lipid metabolism disorders, oxidative stress and upregulation of heat shock protein 70 in zebrafish. Comp Biochem Physiol C Toxicol Pharmacol 2025; 293:110164. [PMID: 40020955 DOI: 10.1016/j.cbpc.2025.110164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 02/11/2025] [Accepted: 02/24/2025] [Indexed: 03/03/2025]
Abstract
Bisphenol AF (BPAF) is a widespread endocrine disruptor in the environment, and the use of BPAF has been strongly associated with the development of several diseases. In this study, we investigated the effects of BPAF on growth, development, oxidative stress and lipid metabolism in zebrafish. We chose the concentrations based on the measured LC50 at 96 h post-fertilization (96 hpf), and the zebrafish embryos were exposed to three different concentrations (0.125, 0.5 and 2 μmol/L). The findings indicated that BPAF exposure in zebrafish leaded to alterations in heart rate, body length and hatching rate, as well as an accumulation of red blood cells in the heart. Additionally, BPAF exposure resulted in increased levels of neutrophils, reactive oxygen species (ROS) and malondialdehyde (MDA), and decreased activity of antioxidant enzymes (superoxide dismutase (SOD) and catalase (CAT)), thus disturbing the balance between oxidative and antioxidative systems. BPAF promoted fatty acid catabolism and inhibited fatty acid synthesis, ultimately leading to a reduction in fatty acid content. Mechanistically, RNA-seq analysis and RT-qPCR revealed a significant upregulation of heat shock protein 70 (hsp70) after BPAF exposure. Inhibition of hsp70 with VER-155008 ameliorated BPAF-induced oxidative stress. These data provided a novel approach to investigate BPAF-induced oxidative stress and suggested that regulation of hsp70 is a crucial target for alleviating this process.
Collapse
Affiliation(s)
- Bingya Zhang
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical University, Bengbu 233030, PR China; Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu 233030, PR China; Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu 233030, PR China
| | - Zhonghua Fan
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical University, Bengbu 233030, PR China; Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu 233030, PR China; Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu 233030, PR China
| | - Xuan Liu
- School of Public Health, Bengbu Medical University, Bengbu 233030, PR China
| | - Yuanyuan Wu
- School of Public Health, Bengbu Medical University, Bengbu 233030, PR China
| | - Lin Cheng
- School of Public Health, Bengbu Medical University, Bengbu 233030, PR China
| | - Li Wang
- School of Public Health, Bengbu Medical University, Bengbu 233030, PR China.
| | - Hui Liu
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical University, Bengbu 233030, PR China; Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu 233030, PR China; Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu 233030, PR China.
| |
Collapse
|
5
|
Fan XC, Wang J. Hederagenin promotes SIRT6 to attenuate epidural scar formation by aggravating PRMT1 deacetylation. Bone Joint Res 2025; 14:516-526. [PMID: 40511498 PMCID: PMC12163784 DOI: 10.1302/2046-3758.146.bjr-2024-0287.r2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/16/2025] Open
Abstract
Aims The formation of a postoperative epidural scar induced by epidural fibrosis is the main reason for recurrence of lumbar disc herniation after laminectomy. Hederagenin (HE) has been found to be widely present in various medicinal plants and has various pharmacological functions. This study aimed to investigate the effect and regulatory mechanism of HE on epidural scar formation. Methods Transforming growth factor beta 1 (TGF-β1)-stimulated epidural scar fibroblasts were used as an in vitro cell model. Based on that, HE treatment was carried out along with sirtuin-6 (SIRT6) silence or protein arginine N-methyltransferase 1 (PRMT1) overexpression. The interaction between SIRT6 and PRMT1 was evaluated by pulldown and co-immunoprecipitation (CoIP) assays. Then, cell proliferation, apoptosis, and fibrosis were measured by Cell Counting Kit (CCK)-8, flow cytometry, and western blotting. Moreover, the effects of receptor activator of nuclear factor-κB ligand (RANKL) supplementation and endoplasmic reticulum (ER) stress were also evaluated by supplementing recombinant protein and specific inhibitor or activator. Results HE depressed cell proliferation and fibrosis, while inducing apoptosis of epidural fibroblasts. Meanwhile, HE promoted SIRT6 expression which suppressed PRMT1 acetylation and protein stability. Additionally, HE induced ER stress and upregulated RANKL in epidural fibroblasts via mediating SIRT6/PRMT1 axis. Conclusion Generally, the therapeutic role of HE treatment on epidural scar formation was exerted by regulating SIRT6/PRMT1 axis-mediated ER stress and RANKL pathway. This study provides evidence of a novel therapeutic measure for epidural scar formation.
Collapse
Affiliation(s)
- Xiao-Chen Fan
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education and Institute of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiao Tong University, Xi'an, China
- Department of Traditional Chinese Medicine, Honghui Hospital, Xi’an, China
| | - Jue Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education and Institute of Biomedical Engineering, School of Life Science and Technology, Xi'an Jiao Tong University, Xi'an, China
| |
Collapse
|
6
|
Jiang Y, Zhang J, Liu W, Qian X, Zhuang X, Hu C. Scutellariae Radix and Coptidis Rhizoma improve NAFLD via regulation of SIRT6/ACSL5 pathway and SCD1. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119834. [PMID: 40254111 DOI: 10.1016/j.jep.2025.119834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 04/06/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The herbal pair Scutellariae Radix-Coptidis Rhizoma (SR-CR) has been widely used in Traditional Chinese Medicine (TCM) for treating metabolic disorders, including nonalcoholic fatty liver disease (NAFLD) -related conditions. Its traditional use highlights its potential in addressing the multifaceted pathogenesis of NAFLD, though the underlying mechanisms remain unclear. AIM OF THE STUDY To evaluate the therapeutic efficacy of the SR-CR herbal pair in alleviating NAFLD and to elucidate its mechanisms of action, with a specific focus on lipid metabolism pathways. MATERIALS AND METHODS The therapeutic effects of SR-CR were assessed using a high-fat diet (HFD)-induced NAFLD rat model and HepG2 cell model. Multi-omics analyses were employed to identify molecular targets and pathways, while affinity ultrafiltration-mass spectrometry characterized bioactive constituents. Findings were validated in vivo and in vitro via Western blot and immunofluorescence. Protein-constituent interactions were further characterized by surface plasmon resonance and molecular docking. RESULTS SR-CR significantly alleviated NAFLD symptoms in HFD-fed rats by reducing hepatic lipid accumulation, inflammation, and hepatocyte ballooning while normalizing biochemical indicators. Mechanistic studies revealed that SR-CR regulates the SIRT6/ACSL5 pathway and SCD1, both critical to lipid metabolism. Scutellariae Radix (SR) and its major constituent, baicalin, enhanced ACSL5 activity via SIRT6-mediated deacetylation, promoting fatty acid oxidation and intracellular lipid utilization. Coptidis Rhizoma (CR) and its primary component, berberine, inhibited SCD1, thereby reducing de novo lipogenesis. These complementary effects synergistically enhanced energy expenditure and reduced lipid synthesis. CONCLUSION The SR-CR herbal pair effectively alleviates HFD-induced NAFLD by synergistically modulating lipid metabolism, enhancing energy expenditure, and reducing de novo lipogenesis through the regulation of the SIRT6/ACSL5 pathway and SCD1. These findings provide molecular evidence for the traditional use of SR-CR in treating metabolic disorders and highlight its potential as a plant-based therapeutic for NAFLD.
Collapse
Affiliation(s)
- Yuanye Jiang
- Department of Gastroenterology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, 200062, China
| | - Jiaqi Zhang
- Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Wangzhenzu Liu
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiaojing Qian
- Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Xiaoyu Zhuang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Cheng Hu
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
7
|
Li L, Zeng Y, Cheng G, Yang H. Acetylation and deacetylation dynamics in stress response to cancer and infections. Semin Immunol 2025; 78:101957. [PMID: 40288003 DOI: 10.1016/j.smim.2025.101957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025]
Abstract
In response to stress stimuli, cells have evolved various mechanisms to integrate internal and external signals to achieve dynamic homeostasis. Lysine acetyltransferase (KATs) and deacetyltransferase (KDACs) are the key modulators of epigenetic modifications, enabling cells to modulate cellular responses through the acetylation and deacetylation of both histone and nonhistone proteins. Understanding the signaling pathways involved in cellular stress response, along with the roles of KATs and KDACs may pave the way for the development of novel therapeutic strategies. This review discusses the molecular mechanisms of acetylation and deacetylation in stress responses related to tumorigenesis, viral and bacterial infections. In tumorigenesis section, we focused on the tumor cells' intrinsic and external molecules and signaling pathways regulated by acetylation and deacetylation modification. In viral and bacterial infections, we summarized the update research on acetylation and deacetylation modification in viral and bacterial infections, which systematical introduction on this topic is not too much. Additionally, we provide an overview of current therapeutic interventions and clinical trials involving KAT and KDAC inhibitors in the treatment of cancer, as well as viral and bacterial infection-related diseases.
Collapse
Affiliation(s)
- Lili Li
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu 215123, China; Guangzhou National Laboratory, Guangzhou, Guangdong 510005, China
| | - Yanqiong Zeng
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu 215123, China
| | - Genhong Cheng
- Guangzhou National Laboratory, Guangzhou, Guangdong 510005, China
| | - Heng Yang
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, Jiangsu 215123, China
| |
Collapse
|
8
|
Zhang L, Liu S, Zhao Q, Liu X, Zhang Q, Liu M, Zhao W. The role of ubiquitination and deubiquitination in the pathogenesis of non-alcoholic fatty liver disease. Front Immunol 2025; 16:1535362. [PMID: 40292292 PMCID: PMC12021615 DOI: 10.3389/fimmu.2025.1535362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/19/2025] [Indexed: 04/30/2025] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is one of the most common chronic liver diseases and is closely associated with metabolic abnormalities. The causes of NAFLD are exceedingly complicated, and it is known that a variety of signaling pathways, endoplasmic reticulum stress, and mitochondrial dysfunction play a role in the pathogenesis of NAFLD. Recent studies have shown that ubiquitination and deubiquitination are involved in the regulation of the NAFLD pathophysiology. Protein ubiquitination is a dynamic and diverse post-translational alteration that affects various cellular biological processes. Numerous disorders, including NAFLD, exhibit imbalances in ubiquitination and deubiquitination. To highlight the significance of this post-translational modification in the pathogenesis of NAFLD and to aid in the development of new therapeutic approaches for the disease, we will discuss the role of enzymes involved in the processes of ubiquitination and deubiquitination, specifically E3 ubiquitin ligases and deubiquitinating enzymes that are important in the regulation of NAFLD.
Collapse
Affiliation(s)
- Lihui Zhang
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Zhengzhou, Henan, China
| | - Sutong Liu
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Zhengzhou, Henan, China
| | - Qing Zhao
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiaoyan Liu
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Qiang Zhang
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Minghao Liu
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Zhengzhou, Henan, China
| | - Wenxiao Zhao
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Zhengzhou, Henan, China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Zhengzhou, Henan, China
| |
Collapse
|
9
|
Ru Z, Li S, Wang M, Ni Y, Qiao H. Exploring Immune-Related Ferroptosis Genes in Thyroid Cancer: A Comprehensive Analysis. Biomedicines 2025; 13:903. [PMID: 40299520 PMCID: PMC12024864 DOI: 10.3390/biomedicines13040903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Accepted: 04/05/2025] [Indexed: 04/30/2025] Open
Abstract
Background: The increasing incidence and poor outcomes of recurrent thyroid cancer highlight the need for innovative therapies. Ferroptosis, a regulated cell death process linked to the tumour microenvironment (TME), offers a promising antitumour strategy. This study explored immune-related ferroptosis genes (IRFGs) in thyroid cancer to uncover novel therapeutic targets. Methods: CIBERSORTx and WGCNA were applied to data from TCGA-THCA to identify hub genes. A prognostic model composed of IRFGs was constructed using LASSO Cox regression. Pearson correlation was employed to analyse the relationships between IRFGs and immune features. Single-cell RNA sequencing (scRNA-seq) revealed gene expression in cell subsets, and qRT-PCR was used for validation. Results: Twelve IRFGs were identified through WGCNA, leading to the classification of thyroid cancer samples into three distinct subtypes. There were significant differences in patient outcomes among these subtypes. A prognostic risk score model was developed based on six key IRFGs (ACSL5, HSD17B11, CCL5, NCF2, PSME1, and ACTB), which were found to be closely associated with immune cell infiltration and immune responses within the TME. The prognostic risk score was identified as a risk factor for thyroid cancer outcomes (HR = 14.737, 95% CI = 1.95-111.65; p = 0.009). ScRNA-seq revealed the predominant expression of these genes in myeloid cells, with differential expression validated using qRT-PCR in thyroid tumour and normal tissues. Conclusions: This study integrates bulk and single-cell RNA sequencing data to identify IRFGs and construct a robust prognostic model, offering new therapeutic targets and improving prognostic evaluation for thyroid cancer patients.
Collapse
Affiliation(s)
- Zixuan Ru
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; (Z.R.)
| | - Siwei Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, China;
| | - Minnan Wang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; (Z.R.)
| | - Yanan Ni
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; (Z.R.)
| | - Hong Qiao
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China; (Z.R.)
- NHC Key Laboratory of Etiology and Epidemiology, Harbin Medical University, Harbin 150081, China
| |
Collapse
|
10
|
Lu Y, Yang J, Wu Q, Wang X. The Role and Molecular Pathways of SIRT6 in Senescence and Age-related Diseases. Adv Biol (Weinh) 2025; 9:e2400469. [PMID: 39913122 DOI: 10.1002/adbi.202400469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 12/10/2024] [Indexed: 02/07/2025]
Abstract
SIRT6 is a NAD+-dependent histone deacetylase with crucial roles in controlling DNA damage repair, telomere homeostasis, oxidative stress, autophagy, and other cellular processes, and it has long been recognized as a longevity-associated protein. This review details its anti-aging-related mechanisms. First, SIRT6 facilitates DNA repair pathways and maintains genome stability by deacetylating histone H3 at K56, K9, and K18 residues, in addition to participating in DNA damage repair through mono-ADP-ribosylation and other mechanisms. Second, SIRT6 preserves telomere integrity and mitigates cellular senescence by reducing oxidative stress-induced damage through the regulation of reactive oxygen species (ROS), inhibition of inflammation, and other pathways. Furthermore, SIRT6 promotes autophagy, slowing cellular senescence via the modulation of various signaling pathways, including AMPK, IGF-Akt-mTOR, H133Y, IL-1β, and mitochondrial autophagy-related proteins. Finally, SIRT6 regulates multiple signaling pathways, such asNF-κB, FOXO, and AMPK, to counteract the aging process. This review particularly delves into the interplay between SIRT6 and various diseases, including tumors, cardiovascular diseases (e.g., atherosclerosis, heart failure), metabolic diseases (e.g., type 2 diabetes, dyslipidemia, gluconeogenesis, osteoporosis), and neurodegenerative diseases (e.g., Alzheimer's disease). Moreover, recent advancements in SIRT6-regulated compounds (e.g., C3G, BZBS, Fisetin, FNDC5, Lycorine hydrochloride, and Ergothioneine) are discussed as potential therapeutic agents for these mediated diseases.
Collapse
Affiliation(s)
- Yi Lu
- School of Basic Medicine, Dali University, Dali, Yunnan, 671000, China
| | - Junye Yang
- School of Basic Medicine, Dali University, Dali, Yunnan, 671000, China
| | - Qiuju Wu
- College of General Education, Guangxi Vocational University of Agriculture, Nanning, Guangxi, 530007, China
| | - Xiaobo Wang
- School of Basic Medicine, Dali University, Dali, Yunnan, 671000, China
| |
Collapse
|
11
|
Choi MG, Lee NY, Koo JH. Stabilizing hepatic fatty acid oxidation: Editorial on "USP29 alleviates the progression of MASLD by stabilizing ACSL5 through K48 deubiquitination". Clin Mol Hepatol 2025; 31:592-595. [PMID: 39501572 PMCID: PMC12016596 DOI: 10.3350/cmh.2024.0971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 04/18/2025] Open
Affiliation(s)
- Myeung Gi Choi
- College of Pharmacy, Seoul National University, Seoul, Korea
| | - Na Young Lee
- College of Pharmacy, Seoul National University, Seoul, Korea
| | - Ja Hyun Koo
- College of Pharmacy, Seoul National University, Seoul, Korea
- Research Institute of Pharmaceutical Sciences and Natural Products Research Institute, Seoul National University, Seoul, Korea
| |
Collapse
|
12
|
Zhang F, Yue K, Sun S, Lu S, Jia G, Zha Y, Zhang S, Chou CJ, Liao C, Li X, Duan Y. Targeting Histone Deacetylase 11 with a Highly Selective Inhibitor for the Treatment of MASLD. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412903. [PMID: 39976110 PMCID: PMC12005767 DOI: 10.1002/advs.202412903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/26/2025] [Indexed: 02/21/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) represents the most prevalent chronic liver disorder globally. Due to its intricate pathogenesis and the current lack of efficacious pharmacological interventions, there is a pressing need to discover novel therapeutic targets and agents for MASLD treatment. Herein, it is found that histone deacetylase 11 (HDAC11), a subtype of HDAC family, is markedly overexpressed in both in vitro and in vivo models of MASLD. Furthermore, the knockdown of HDAC11 is observed to mitigate lipid accumulation in hepatic cells. A highly selective HDAC11 inhibitor, B6, which exhibits favorable pharmacokinetic property and liver distribution, is further designed and synthesized. Integrating RNA-seq data with in vivo and in vitro experiments, B6 is found to inhibit de novo lipogenesis (DNL) and promote fatty acid oxidation, thus mitigating hepatic lipid accumulation and pathological symptoms in MASLD mice. Further omics analysis and experiments reveal that B6 enhances the phosphorylation of AMPKα1 at Thr172 through the inhibition of HDAC11, consequently modulating DNL and fatty acid oxidation in the liver. In summary, this study identifies HDAC11 as a potential therapeutic target in MASLD and reports the discovery of a highly selective HDAC11 inhibitor with favorable drug-like properties for the treatment of MASLD.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Cardiologythe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230001China
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education InstitutesHefei University of TechnologyHefei230031China
| | - Kairui Yue
- Key Laboratory of Marine DrugsChinese Ministry of EducationSchool of Medicine and PharmacyOcean University of China5 Yushan RoadQingdao266003China
| | - Simin Sun
- Key Laboratory of Marine DrugsChinese Ministry of EducationSchool of Medicine and PharmacyOcean University of China5 Yushan RoadQingdao266003China
| | - Shengyuan Lu
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education InstitutesHefei University of TechnologyHefei230031China
| | - Geng Jia
- Key Laboratory of Marine DrugsChinese Ministry of EducationSchool of Medicine and PharmacyOcean University of China5 Yushan RoadQingdao266003China
| | - Yang Zha
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education InstitutesHefei University of TechnologyHefei230031China
| | - Shuang Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education InstitutesHefei University of TechnologyHefei230031China
| | - C. James Chou
- Department of Drug Discovery and Biomedical SciencesCollege of PharmacyMedical University of South CarolinaCharlestonSC29425USA
| | - Chenzhong Liao
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education InstitutesHefei University of TechnologyHefei230031China
| | - Xiaoyang Li
- Key Laboratory of Marine DrugsChinese Ministry of EducationSchool of Medicine and PharmacyOcean University of China5 Yushan RoadQingdao266003China
- Marine Biomedical Research Institute of QingdaoQingdaoShandong266071China
| | - Yajun Duan
- Department of Cardiologythe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230001China
| |
Collapse
|
13
|
Mejía-Guzmán JE, Belmont-Hernández RA, Chávez-Tapia NC, Uribe M, Nuño-Lámbarri N. Metabolic-Dysfunction-Associated Steatotic Liver Disease: Molecular Mechanisms, Clinical Implications, and Emerging Therapeutic Strategies. Int J Mol Sci 2025; 26:2959. [PMID: 40243565 PMCID: PMC11988898 DOI: 10.3390/ijms26072959] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Metabolic-dysfunction-associated steatotic liver disease (MASLD), previously known as non-alcoholic fatty liver disease (NAFLD), is a highly prevalent metabolic disorder characterized by hepatic steatosis in conjunction with at least one cardiometabolic risk factor, such as obesity, type 2 diabetes, hypertension, or dyslipidemia. As global rates of obesity and metabolic syndrome continue to rise, MASLD is becoming a major public health concern, with projections indicating a substantial increase in prevalence over the coming decades. The disease spectrum ranges from simple steatosis to metabolic-dysfunction-associated steatohepatitis (MASH), fibrosis, cirrhosis, and hepatocellular carcinoma, contributing to significant morbidity and mortality worldwide. This review delves into the molecular mechanisms driving MASLD pathogenesis, including dysregulation of lipid metabolism, chronic inflammation, oxidative stress, mitochondrial dysfunction, and gut microbiota alterations. Recent advances in research have highlighted the role of genetic and epigenetic factors in disease progression, as well as novel therapeutic targets such as peroxisome proliferator-activated receptors (PPARs), fibroblast growth factors, and thyroid hormone receptor beta agonists. Given the multifaceted nature of MASLD, a multidisciplinary approach integrating early diagnosis, molecular insights, lifestyle interventions, and personalized therapies is critical. This review underscores the urgent need for continued research into innovative treatment strategies and precision medicine approaches to halt MASLD progression and improve patient outcomes.
Collapse
Affiliation(s)
- Jeysson E. Mejía-Guzmán
- Translational Research Unit, Medica Sur Clinic & Foundation, Mexico City 14050, Mexico; (J.E.M.-G.); (R.A.B.-H.); (N.C.C.-T.)
| | - Ramón A. Belmont-Hernández
- Translational Research Unit, Medica Sur Clinic & Foundation, Mexico City 14050, Mexico; (J.E.M.-G.); (R.A.B.-H.); (N.C.C.-T.)
- Postgraduate Program in Experimental Biology, División de Ciencias Básicas y de la Salud (DCBS), Universidad Autonoma Metropolitana-Iztapalapa, Mexico City 09340, Mexico
| | - Norberto C. Chávez-Tapia
- Translational Research Unit, Medica Sur Clinic & Foundation, Mexico City 14050, Mexico; (J.E.M.-G.); (R.A.B.-H.); (N.C.C.-T.)
- Obesity and Digestive Diseases Unit, Medica Sur Clinic & Foundation, Mexico City 14050, Mexico;
| | - Misael Uribe
- Obesity and Digestive Diseases Unit, Medica Sur Clinic & Foundation, Mexico City 14050, Mexico;
| | - Natalia Nuño-Lámbarri
- Translational Research Unit, Medica Sur Clinic & Foundation, Mexico City 14050, Mexico; (J.E.M.-G.); (R.A.B.-H.); (N.C.C.-T.)
- Surgery Department, Faculty of Medicine, The National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico
| |
Collapse
|
14
|
Wang M, Mu G, Qiu B, Wang S, Tao C, Mao Y, Zhao X, Liu J, Chen K, Li Z, Wang W, Yang E, Yang Y. Competitive antagonism of KAT7 crotonylation against acetylation affects procentriole formation and colorectal tumorigenesis. Nat Commun 2025; 16:2379. [PMID: 40064919 PMCID: PMC11893896 DOI: 10.1038/s41467-025-57546-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Accurate procentriole formation is critical for centriole duplication. However, the holistic transcriptional regulatory mechanisms underlying this process remain elusive. Here, we show that KAT7 crotonylation, facilitated by the crotonyltransferase hMOF, competes against its acetylation regulated by the deacetylase HDAC2 at the K432 residue upon DNA damage stimulation. This competition diminishes its histone acetyltransferase activity, leading to the inhibition of procentriole formation in colorectal cancer cells. Mechanistically, the reduction of KAT7 histone acetyltransferase activity by the antagonistic effect of KAT7 crotonylation against its acetylation decreases the gene expression associated with procentriole formation by modulating the enrichment of H3K14ac at their promoters and plays an important role in colorectal tumorigenesis. Furthermore, KAT7 crotonylation and acetylation are associated with the prognosis in colorectal cancer patients. Collectively, our findings uncover a previously unidentified role of KAT7 in the regulation of procentriole formation and colorectal tumorigenesis via competitive antagonism of its crotonylation against acetylation.
Collapse
Affiliation(s)
- Meng Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Beijing, 100191, China
| | - Guanqun Mu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Bingquan Qiu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Shuo Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Changyu Tao
- Department of Human Anatomy, Histology & Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Yutong Mao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Xinhui Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Jiansong Liu
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Keyu Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Ziyu Li
- Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Weibin Wang
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Ence Yang
- Department of Medical Bioinformatics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
| | - Yang Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
- Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Beijing, 100191, China.
| |
Collapse
|
15
|
Fan S, Chen W, Li Y, Guo K, Tang H, Ye J, Zhou Z, Tan M, Wei H, Huang X, Huang K, Ke X. Qige Decoction attenuated non-alcoholic fatty liver disease through regulating SIRT6-PPARα-mediated fatty acid oxidation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156395. [PMID: 39855055 DOI: 10.1016/j.phymed.2025.156395] [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: 04/17/2024] [Revised: 11/24/2024] [Accepted: 01/12/2025] [Indexed: 01/27/2025]
Abstract
BACKGROUND Sirtuin 6 (SIRT6), a potential therapeutic target for non-alcoholic fatty liver disease (NAFLD), has been shown to regulate fatty acid oxidation (FAO) by interacting with peroxisome proliferator-activated receptor α (PPARα). However, the impact of SIRT6-PPARα pathway on NAFLD phenotype has not yet been reported. Qige decoction (QG), a traditional Chinese medicine (TCM) formula, is widely applied to treat disorders of glycolipid metabolism. Our previous experiments showed that QG reduced hepatic steatosis and provided preliminary evidence that QG may promote FAO. However, a thorough understanding of molecular mechanisms by which QG regulates FAO requires further investigation. PURPOSE To investigate the role of SIRT6-PPARα signalling pathway on NAFLD phenotype and explore the mechanism by which QG improves NAFLD and its relationship with FAO regulated by SIRT6-PPARα signalling pathway. METHODS In vivo study, NAFLD mice induced by high fat diet (HFD) were divided into two parts. The first part involved four groups: control (CON), model (MOD), PPARα agonist (WY-14,643, WY), and SIRT6 inhibitor (OSS-128,167, OS) groups. The second part involved five groups: CON group, MOD group, positive drug (POS) group, low dose QG (QGL) group, and high dose QG (QGH) group. Widely-targeted lipidomic were performed by UHPLC-QTOF/MS to analyse differential lipids (DELs) in the liver, while differentially expressed genes (DEGs) were analysed by transcriptome analysis on the Illumina sequencing platform. In vitro study, co-immunoprecipitation and dual luciferase assay were employed to further identify the molecular mechanisms of SIRT6-PPARα interaction. The lentiviral vector, TG assay, and acetyl-CoA assay were used to clarify the indispensable role of the SIRT6-PPARα signalling pathway on QG amelioration of lipid accumulation in vitro. RESULTS Down-regulation of SIRT6 inhibited PPARα-mediated FAO and aggravated lipid accumulation in hepatocytes both in vivo and in vitro. SIRT6 bound to PPARα in HepG2 cells; however, SIRT6 activation of the PPARα promoter was not detected. Along with QG reduced hepatocyte lipid accumulation, SIRT6-PPARα signalling pathway was upregulated in vivo and in vitro. However, the alleviating effect of QG on lipid accumulation was blocked by SIRT6 silencing in vitro. CONCLUSION This study verified that SIRT6-PPARα signalling pathway inhibition exacerbated NAFLD dyslipidaemia and hepatic steatosis. In addition, this study provided the first in-depth analysis of the molecular mechanisms by which QG ameliorates NFALD, involving promotion of FAO through activation of the SIRT6-PPARα signalling pathway. Our study offers significant insights for the clinical application of QG.
Collapse
Affiliation(s)
- Simin Fan
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 51000 Guangdong, PR China; First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou 510410 Guangdong, PR China; The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405 Guangdong, PR China
| | - Wei Chen
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405 Guangdong, PR China
| | - Yanfang Li
- First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou 510410 Guangdong, PR China
| | - Kaixin Guo
- First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou 510410 Guangdong, PR China
| | - Hui Tang
- First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou 510410 Guangdong, PR China
| | - Jintong Ye
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405 Guangdong, PR China
| | - Zunming Zhou
- First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou 510410 Guangdong, PR China
| | - Meiao Tan
- First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou 510410 Guangdong, PR China
| | - Haoyang Wei
- First Clinical School of Guangzhou University of Chinese Medicine, Guangzhou 510410 Guangdong, PR China
| | - Xiwen Huang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405 Guangdong, PR China
| | - Keer Huang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405 Guangdong, PR China.
| | - Xuehong Ke
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405 Guangdong, PR China.
| |
Collapse
|
16
|
Chen F, Xu W, Tang M, Tian Y, Shu Y, He X, Zhou L, Liu Q, Zhu Q, Lu X, Zhang J, Zhu WG. hnRNPA2B1 deacetylation by SIRT6 restrains local transcription and safeguards genome stability. Cell Death Differ 2025; 32:382-396. [PMID: 39511404 PMCID: PMC11893882 DOI: 10.1038/s41418-024-01412-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/15/2024] Open
Abstract
Repair of double strand breaks (DSBs) by RNA-binding proteins (RBPs) is vital for ensuring genome integrity. DSB repair is accompanied by local transcriptional repression in the vicinity of transcriptionally active genes, but the mechanism by which RBPs regulate transcriptional regulation is unclear. Here, we demonstrated that RBP hnRNPA2B1 functions as a RNA polymerase-associated factor that stabilizes the transcription complex under physiological conditions. Following a DSB, hnRNPA2B1 is released from damaged chromatin, reducing the efficiency of RNAPII complex assembly, leading to local transcriptional repression. Mechanistically, SIRT6 deacetylates hnRNPA2B1 at K113/173 residues, enforcing its rapid detachment from DSBs. This process disrupts the integrity of the RNAPII complex on active chromatin, which is a pre-requisite for transient but complete repression of local transcription. Functionally, the overexpression of an acetylation mimic stabilizes the transcription complex and facilitates the functioning of the transcription machinery. hnRNPA2B1 acetylation status was negatively correlated with SIRT6 expression, and acetylation mimic enhanced radio-sensitivity in vivo. Our findings demonstrate that hnRNPA2B1 is crucial for transcriptional repression. We have uncovered the missing link between DSB repair and transcriptional regulation in genome stability maintenance, highlighting the potential of hnRNPA2B1 as a therapeutic target.
Collapse
Affiliation(s)
- Feng Chen
- Department of Biochemistry and Molecular Biology, International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
| | - Wenchao Xu
- Department of Biochemistry and Molecular Biology, International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
| | - Ming Tang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuan Tian
- Department of Biochemistry and Molecular Biology, International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
| | - Yuxin Shu
- Department of Biochemistry and Molecular Biology, International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
- School of Basic Medical Sciences, Wannan Medical College, Wuhu, Anhui, China
| | - Xingkai He
- Department of Biochemistry and Molecular Biology, International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
| | - Linmin Zhou
- Department of Biochemistry and Molecular Biology, International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
| | - Qi Liu
- Department of Biochemistry and Molecular Biology, International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
| | - Qian Zhu
- Department of Biochemistry and Molecular Biology, International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
| | - Xiaopeng Lu
- Department of Biochemistry and Molecular Biology, International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
| | - Jun Zhang
- Department of Biochemistry and Molecular Biology, International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China.
| | - Wei-Guo Zhu
- Department of Biochemistry and Molecular Biology, International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China.
- School of Basic Medical Sciences, Wannan Medical College, Wuhu, Anhui, China.
| |
Collapse
|
17
|
Ran Q, Huang M, Wang L, Li Y, Wu W, Liu X, Chen J, Yang M, Han K, Guo X. Integrated bioinformatics and multi-omics to investigate the mechanism of Rhododendron molle Flos-induced hepatotoxicity. JOURNAL OF ETHNOPHARMACOLOGY 2025; 341:119308. [PMID: 39746411 DOI: 10.1016/j.jep.2024.119308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 12/11/2024] [Accepted: 12/29/2024] [Indexed: 01/04/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Drug-induced liver injury (DILI) is an important and common adverse drug event. Rhododendron molle Flos (RMF), as one of toxic Traditional Chinese medicines (TCMs), holds a prominent position in clinical practice for treating rheumatoid arthritis. However, the toxicity of RMF limits its safe. Most of the concerns are about its rapid neurotoxicity and cardiotoxicity, with less attention paid to its hepatotoxicity, and the mechanism of which is still unclear. AIM OF THE STUDY To reveal the mechanism of RMF-induced hepatotoxicity by bioinformatics and multi-omics. MATERIALS AND METHODS Rats were intragastric administered RMF at doses of 0.8 g/kg, 0.4 g/kg, and 0.2 g/kg once daily for 2 weeks. Initially, hepatotoxicity was then evaluated using liver function enzymes, antioxidant enzymes, and histopathology. Subsequently, network toxicology, transcriptomics, and metabolomics were used to identify the genes and metabolites. In addition, molecular docking and Western blot were employed to verify toxic components and key targets. RESULTS RMF caused abnormal levels of ALT, γ-GT, TBIL, and TBA in the serum of rats, as well as abnormal levels of MDA, GSH-Px, and SOD in the liver, leading to inflammatory infiltration of liver cells, with a dose-dependent manner. RMF disordered the steroid hormone biosynthesis, pyruvate metabolism, fatty acid biosynthesis, and arachidonic acid metabolism. Six key targets were identified- UGT1A6, CYP2E1, ACOT1, ACSL5, CTH, and PKLR, along with their corresponding metabolites, namely 17β-estradiol, estriol, arachidonic acid, octadecanoic acid, and pyruvic acid. The hepatotoxicity could be attributed to five diterpenoid components, including grayanotoxin-III, rhodojaponin (RJ)-I, RJ-II, RJ-III, and RJ-V. CONCLUSIONS This study comprehensively identified the toxic components, upstream targets, and downstream metabolites of RMF-induced liver toxicity, providing a basis for evaluating and monitoring liver function in patients during clinical application.
Collapse
Affiliation(s)
- Qiang Ran
- Department of Orthopedics, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China.
| | - Mengjun Huang
- National-Local Joint Engineering Research Center for Innovative Targeted Drugs, Chongqing University of Arts and Sciences, Chongqing, 402160, China.
| | - Lijuan Wang
- Department of Pathology, Chongqing Traditional Chinese Medicine Hospital (the First Affiliated Hospital of Chongqing College of Traditional Chinese Medicine), Chongqing, 400021, China.
| | - Yanyan Li
- Department of Pharmacy, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China.
| | - Wenhui Wu
- Department of Preparation Center, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China.
| | - Xia Liu
- Department of Pharmacy, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China.
| | - Juan Chen
- Department of Preparation Center, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China.
| | - Min Yang
- Department of Preparation Center, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China.
| | - Keqing Han
- Department of Pharmacy, Shaanxi Provincial Tuberculosis Prevention and Control Hospital (the Fifth People's Hospital of Shaanxi Province), Xi'an, Shanxi, 710100, China.
| | - Xiaohong Guo
- Department of Preparation Center, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China.
| |
Collapse
|
18
|
Yang Q, He WH, Xie L, Chen T, Liu RF, Hu JJ, Guo JY, Tan GZ, Wu FL, Gu P, Chen P, Chen Y. Oral administration of astilbin mitigates acetaminophen-induced acute liver injury in mice by modulating the gut microbiota. Acta Pharmacol Sin 2025; 46:416-429. [PMID: 39313515 PMCID: PMC11747501 DOI: 10.1038/s41401-024-01383-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/16/2024] [Accepted: 08/18/2024] [Indexed: 09/25/2024]
Abstract
Acetaminophen (APAP) overdose-induced acute liver injury (ALI) is characterized by extensive oxidative stress, and the clinical interventions for this adverse effect remain limited. Astilbin is an active compound found in the rhizome of Smilax glabra Roxb. with anti-inflammatory and antioxidant activities. Due to its low oral bioavailability, astilbin can accumulate in the intestine, which provides a basis for the interaction between astilbin and gut microbiota (GM). In the present study we investigated the protective effects of astilbin against APAP-induced ALI by focusing on the interaction between astilbin and GM. Mice were treated with astilbin (50 mg·kg-1·d-1, i.g.) for 7 days. After the last administration of astilbin for 2 h, the mice received APAP (300 mg/kg, i.g.) to induce ALI. We showed that oral administration of astilbin significantly alleviated APAP-induced ALI by altering the composition of GM and enriching beneficial metabolites including hydroxytyrosol (HT). GM depletion using an "antibiotics cocktail" or paraoral administration of astilbin abolished the hepatoprotective effects of astilbin. On the other hand, administration of HT (10 mg/kg, i.g.) caused similar protective effects in APAP-induced ALI mice. Transcriptomic analysis of the liver tissue revealed that HT inhibited reactive oxygen species and inflammation-related signaling in APAP-induced ALI; HT promoted activation of the Nrf2 signaling pathway to combat oxidative stress following APAP challenge in a sirtuin-6-dependent manner. These results highlight that oral astilbin ameliorates APAP-induced ALI by manipulating the GM and metabolites towards a more favorable profile, and provide an alternative therapeutic strategy for alleviating APAP-induced ALI.
Collapse
Affiliation(s)
- Qin Yang
- Department of Gastroenterology, The Seventh Affiliated Hospital of Southern Medical University, Foshan, 528244, China
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Wen-Hao He
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Li Xie
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Tao Chen
- Department of Physiology, School of Basic Medical Sciences, Gannan Medical University, Ganzhou, 341000, China
| | - Ruo-Fan Liu
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jia-Jia Hu
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jia-Yin Guo
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Guo-Zhu Tan
- Department of Gastroenterology, The Seventh Affiliated Hospital of Southern Medical University, Foshan, 528244, China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Fu-Ling Wu
- Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Peng Gu
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Peng Chen
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Yu Chen
- Department of Gastroenterology, The Seventh Affiliated Hospital of Southern Medical University, Foshan, 528244, China.
| |
Collapse
|
19
|
Fan Z, Wei X, Zhu X, Du Y. Sirtuins in kidney homeostasis and disease: where are we now? Front Endocrinol (Lausanne) 2025; 15:1524674. [PMID: 39911234 PMCID: PMC11794115 DOI: 10.3389/fendo.2024.1524674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 12/31/2024] [Indexed: 02/07/2025] Open
Abstract
Sirtuins, identified as (NAD+)- dependent class III histone deacetylases, engage in a spectrum of biological functions, encompassing DNA damage repair, oxidative stress, immune modulation, mitochondrial homeostasis, apoptosis and autophagy. Sirtuins play an apoptosis role in regulating cellular operations and overall organism health. Mounting data indicate that dysregulated sirtuin expression is linked to the onset of renal diseases. Effective modulation of sirtuins expression and activity has been shown to improve renal function and attenuate the advancement of kidney diseases. In this review, we present a comprehensive overview of the biological impacts of sirtuins and their molecular targets in regulating renal diseases. Additionally, we detail advancements in elucidating sirtuin roles in the pathophysiology of both chronic and acute renal disorders. We review compounds that modulate sirtuin activity through activation or inhibition, potentially improving outcomes in renal disease. In summary, strategic manipulation of sirtuin activity represents a prospective therapeutic approach for renal diseases.
Collapse
Affiliation(s)
| | | | | | - Yujun Du
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
20
|
Wang H, Zhang L, Bai X, Wang H, Sun H. Propofol Protects against Pyroptosis of Photoreceptors in Subretinal Hemorrhage via Regulating SIRT6/NLRP3 Signaling. Crit Rev Eukaryot Gene Expr 2025; 35:75-85. [PMID: 39957594 DOI: 10.1615/critreveukaryotgeneexpr.2024056605] [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: 02/18/2025]
Abstract
Subretinal hemorrhage-induced neurotoxicity is a key cause of vision loss in age-related macular degeneration (AMD). The purpose of this study is to investigate the effects of Propofol on neurotoxicity. Oxygen glucose deprivation (OGD) was used to establish in vitro subretinal hemorrhage model. Gene expression was determined using reverse transcription-quantitative polymerase chain reaction and western blot. Cytokine release was determined using enzyme-linked immunosorbent assay. The interaction between sirtuin 6 (SIRT6) and NLR family pyrin domain containing 3 (NLRP3) was detected using co-immunoprecipitation assay. Cellular function was determined using cell counting kit-8 assay, lactate dehydrogenase assay, and terminal deoxynucleotidyl transferase dUTP nick end labeling assay. Propofol suppressed the inflammatory response induced by OGD. Moreover, Propofol inhibited the neurotoxicity and pyroptosis of photoreceptors. Propofol mediated the overexpression of SIRT6, which was downregulated in AMD. Inhibition of SIRT6 alleviated its deacetylation of NLRP3. Additionally, SIRT6 deficiency antagonized the effects of Propofol and promoted the neurotoxicity and pyroptosis of photoreceptors. Taken together, Propofol protects against subretinal hemorrhage-induced neurotoxicity and pyroptosis of photoreceptors via promoting SIRT6-mediated deacetylation of NLRP3.
Collapse
Affiliation(s)
| | - Limei Zhang
- Hebei Eye Hospital, Hebei Treatment Center for Eye Disease, Hebei Key Laboratory of Ophthalmology
| | - Xiaoli Bai
- Department of Anesthesiology, Hebei Eye Hospital, Xingtai City, Hebei Province 054001, China
| | - Hao Wang
- Department of Neurosurgery, Xingtai People's Hospital, Xingtai City, Hebei Province 054001, China
| | - Hao Sun
- Department of Anesthesiology, Hebei Eye Hospital, Xingtai City, Hebei Province 054001, China
| |
Collapse
|
21
|
Hu S, Wang Z, Zhu K, Shi H, Qin F, Zhang T, tian S, Ji Y, Zhang J, Qin J, She Z, Zhang X, Zhang P, Li H. USP29 alleviates the progression of MASLD by stabilizing ACSL5 through K48 deubiquitination. Clin Mol Hepatol 2025; 31:147-165. [PMID: 39355870 PMCID: PMC11791544 DOI: 10.3350/cmh.2024.0478] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/29/2024] [Accepted: 10/02/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND/AIMS Metabolic dysfunction-associated steatotic liver disease (MASLD) is a chronic liver disease characterized by hepatic steatosis. Ubiquitin-specific protease 29 (USP29) plays pivotal roles in hepatic ischemiareperfusion injury and hepatocellular carcinoma, but its role in MASLD remains unexplored. Therefore, the aim of this study was to reveal the effects and underlying mechanisms of USP29 in MASLD progression. METHODS USP29 expression was assessed in liver samples from MASLD patients and mice. The role and molecular mechanism of USP29 in MASLD were assessed in high-fat diet-fed and high-fat/high-cholesterol diet-fed mice and palmitic acid and oleic acid treated hepatocytes. RESULTS USP29 protein levels were significantly reduced in mice and humans with MASLD. Hepatic steatosis, inflammation and fibrosis were significantly exacerbated by USP29 deletion and relieved by USP29 overexpression. Mechanistically, USP29 significantly activated the expression of genes related to fatty acid β-oxidation (FAO) under metabolic stimulation, directly interacted with long-chain acyl-CoA synthase 5 (ACSL5) and repressed ACSL5 degradation by increasing ACSL5 K48-linked deubiquitination. Moreover, the effect of USP29 on hepatocyte lipid accumulation and MASLD was dependent on ACSL5. CONCLUSION USP29 functions as a novel negative regulator of MASLD by stabilizing ACSL5 to promote FAO. The activation of the USP29-ACSL5 axis may represent a potential therapeutic strategy for MASLD.
Collapse
Affiliation(s)
- Sha Hu
- Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Zhouxiang Wang
- Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Kun Zhu
- Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Hongjie Shi
- Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Fang Qin
- Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Tuo Zhang
- Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Song tian
- Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Yanxiao Ji
- Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Jianqing Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Juanjuan Qin
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhigang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaojing Zhang
- Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Peng Zhang
- Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
| | - Hongliang Li
- Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| |
Collapse
|
22
|
Li Y, Han Q, Liu Y, Yin J, Ma J. Role of the histone deacetylase family in lipid metabolism: Structural specificity and functional diversity. Pharmacol Res 2024; 210:107493. [PMID: 39491635 DOI: 10.1016/j.phrs.2024.107493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/23/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
Lipids play crucial roles in signal transduction. Lipid metabolism is associated with several transcriptional regulators, including peroxisome proliferator activated receptor γ, sterol regulatory element-binding protein 1, and acetyl-CoA carboxylase. In recent years, increasing evidence has suggested that members of the histone deacetylase (HDAC) family play key roles in lipid metabolism. However, the mechanisms by which each member of this family regulates lipid metabolism remain unclear. This review discusses the latest research on the roles played by HDACs in fat metabolism. The role of HDACs in obesity, diabetes, and atherosclerosis has also been discussed. In addition, the interaction of HDACs with the gut microbiome and circadian rhythm has been reviewed, and the future development trend in HDACs has been predicted, which may potentiate therapeutic application of targeted HDACs in related metabolic diseases.
Collapse
Affiliation(s)
- Yunxia Li
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China; College of Animal Science and Technology, Hunan Agriculture University, Changsha 410128, China
| | - Qi Han
- College of Animal Science and Technology, Hunan Agriculture University, Changsha 410128, China
| | - Yuxin Liu
- College of Animal Science and Technology, Hunan Agriculture University, Changsha 410128, China
| | - Jie Yin
- College of Animal Science and Technology, Hunan Agriculture University, Changsha 410128, China.
| | - Jie Ma
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China.
| |
Collapse
|
23
|
Wan W, Wei R, Xu B, Cao H, Zhi Y, Guo F, Liu H, Li B, Wu J, Gao Y, Zhang K. Qiwei Jinggan Ling regulates oxidative stress and lipid metabolism in alcoholic liver disease by activating AMPK. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156125. [PMID: 39388920 DOI: 10.1016/j.phymed.2024.156125] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/24/2024] [Accepted: 07/09/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Alcoholic liver disease (ALD) is a severe public health concern worldwide and there is still a lack of effective treatments. Qiwei Jinggan Ling (QJL) has protective effects against various liver injuries, but its pharmacological action on ALD has received little attention. PURPOSE To investigate the effect and mechanism of QJL on ALD in vivo and in vitro. METHODS In vivo, an ALD mouse model was established by alcohol combined with a high-fat diet (HFD) and treated with QJL. Biochemical indicators, HE staining, and Oil Red O staining were employed to assess hepatic oxidative stress, steatosis, and alcohol metabolism. RNA sequencing analysis was performed, and the results were verified by qRT-PCR and Western blot to elucidate the hepatoprotective mechanism of QJL. In vitro, HepG2 cells were co-stimulated with NaOA (sodium oleate) and EtOH (ethanol), followed by intervention with Compound C (CC, AMPK inhibitor) and QJL-containing serum. Oil Red O, BODIPY (boron-dipyrromethene), and ROS (reactive oxygen species) staining were applied to validate the efficacy and mechanism of QJL-containing serum. The expression of AMP-activated protein kinase (AMPK) pathway-related factors was analyzed through qRT-PCR and Western blot for additional corroboration. Moreover, the key pharmacodynamic components of QJL were identified by UPLC-MS/MS and molecular docking. RESULTS In vivo, QJL ameliorated liver structural disorders, steatosis, oxidative stress, and impaired alcohol metabolism, as indicated by biochemical indicators and histopathological assays. RNA sequencing analysis revealed that QJL reversed the expression of genes related to alcohol metabolism, fatty acid metabolism, and cholesterol metabolism. The results of qRT-PCR and Western blot were in line with those of RNA sequencing. Furthermore, it was discovered that QJL significantly upregulated the expression of p-AMPK and downregulated the expression of sterol regulatory element binding transcription factor 1 (SREBP-1c). In vitro, biochemical indicators and staining assays demonstrated that QJL-containing serum inhibited lipid accumulation and oxidative stress. The qRT-PCR and Western blot analysis revealed that QJL-containing serum markedly enhanced the expression of p-AMPK and carnitine palmitoyltransferase 1a (Cpt1a), while suppressing the expression of SREBP-1c, fatty acid synthase (Fasn), and acetyl-coenzyme A carboxylase 1 (ACC-1). However, CC inhibited the above pharmacological activities of QJL-containing serum. Additionally, (2S)-Liquiritigenin, Glycyrrhetinate, Isovitexin, Taxifolin, and Yohimbine were proved to be the key active components of QJL. CONCLUSION QJL had the potential to be a therapeutic drug for ALD by activating the AMPK pathway, thereby regulating lipid metabolism and inhibiting oxidative stress.
Collapse
Affiliation(s)
- Weimin Wan
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Riming Wei
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Baoling Xu
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, Guangxi, China; Department of Emergency, The Second Affiliated Hospital of Guilin Medical University, Guilin 541199, Guangxi, China
| | - Houkang Cao
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Yueping Zhi
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Fengyue Guo
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Haiping Liu
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Bo Li
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Jianzhao Wu
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Ya Gao
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, Guangxi, China
| | - Kefeng Zhang
- Pharmacology Laboratory of Prevention and Treatment of High Incidence of Disease, Guilin Medical University, Guilin 541199, Guangxi, China.
| |
Collapse
|
24
|
Wade H, Pan K, Zhang B, Zheng W, Su Q. Mechanistic role of long non-coding RNAs in the pathogenesis of metabolic dysfunction-associated steatotic liver disease and fibrosis. EGASTROENTEROLOGY 2024; 2:e100115. [PMID: 39872125 PMCID: PMC11729351 DOI: 10.1136/egastro-2024-100115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), previously referred to as non-alcoholic fatty liver disease, encompasses a broad range of hepatic metabolic disorders primarily characterised by the disruption of hepatic lipid metabolism, hepatic lipid accumulation and steatosis. Severe cases of MASLD might progress to metabolic dysfunction-associated steatohepatitis, characterised by hepatic inflammation, hepatocyte ballooning degeneration, activation of hepatic stellate cells (HSCs) and fibrogenesis. It may further progress to hepatocellular carcinoma. In the liver, long non-coding RNAs (lncRNAs) target multiple metabolic pathways in hepatocytes, HSCs, and Kupffer cells at different stages of MASLD and liver fibrosis. In this study, we overview recent findings on the potential role of lncRNAs in the pathogenesis of MASLD and liver fibrosis via modulation of de novo lipid synthesis, fatty acid β-oxidation, lipotoxicity, oxidative stress, metabolic inflammation, mammalian target of rapamycin signalling, apoptosis, ubiquitination and fibrogenesis. We critically assess the literature reports that investigate the complex interplay between lncRNA, microRNA and key mediators in liver injury, in both human participants and animal models of MASLD and liver fibrosis. We also highlight the therapeutic potential of lncRNAs in chronic liver diseases.
Collapse
Affiliation(s)
- Henry Wade
- School of Biological Sciences, Queen’s University Belfast, Belfast, UK
| | - Kaichao Pan
- Endocrinology Group, Advocate Illinois Masonic Medical Center, Chicago, Illinois, USA
| | - Bingrui Zhang
- School of Biological Sciences, Queen’s University Belfast, Belfast, UK
| | - Wenhua Zheng
- Faculty of Health Science, University of Macau, Macau, China
| | - Qiaozhu Su
- School of Biological Sciences, Queen’s University Belfast, Belfast, UK
| |
Collapse
|
25
|
Huang J, Su J, Wang H, Chen J, Tian Y, Zhang J, Feng T, Di L, Lu X, Sheng H, Zhu Q, Chen X, Wang J, He X, Yerkinkazhina Y, Xie Z, Shu Y, Kang T, Tang H, Qian J, Zhu WG. Discovery of Novel PROTAC SIRT6 Degraders with Potent Efficacy against Hepatocellular Carcinoma. J Med Chem 2024; 67:17319-17349. [PMID: 39323022 DOI: 10.1021/acs.jmedchem.4c01223] [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: 09/27/2024]
Abstract
Sirtuin 6 (SIRT6), a member of the SIRT family, plays essential roles in the regulation of metabolism, inflammation, aging, DNA repair, and cancer development, making it a promising anticancer drug target. Herein, we present our use of proteolysis-targeting chimera (PROTAC) technology to formulate a series of highly potent and selective SIRT6 degraders. One of the degraders, SZU-B6, induced the near-complete degradation of SIRT6 in both SK-HEP-1 and Huh-7 cell lines and more potently inhibited hepatocellular carcinoma (HCC) cell proliferation than the parental inhibitors. In preliminary mechanistic studies, SZU-B6 hampered DNA damage repair, promoting the cellular radiosensitization of cancer cells. Our SIRT6 degrader SZU-B6 displayed promising antitumor activity, particularly when combined with the well-known kinase inhibitor sorafenib or irradiation in an SK-HEP-1 xenograft mouse model. Our results suggest that these PROTACs might constitute a potent therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Jinbo Huang
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Health Science Centre School of Basic Medical Sciences, Shenzhen University, Shenzhen 518055, China
- Shenzhen University School of Pharmacy, Shenzhen University Medical School, Shenzhen 518055, China
- National Engineering Research Centrer for Biotechnology, Shenzhen 518055, China
| | - Jiajie Su
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Health Science Centre School of Basic Medical Sciences, Shenzhen University, Shenzhen 518055, China
- Shenzhen University School of Pharmacy, Shenzhen University Medical School, Shenzhen 518055, China
| | - Haiyu Wang
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Health Science Centre School of Basic Medical Sciences, Shenzhen University, Shenzhen 518055, China
- Shenzhen University School of Pharmacy, Shenzhen University Medical School, Shenzhen 518055, China
| | - Jiayi Chen
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Health Science Centre School of Basic Medical Sciences, Shenzhen University, Shenzhen 518055, China
- Shenzhen University School of Pharmacy, Shenzhen University Medical School, Shenzhen 518055, China
| | - Yuan Tian
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Health Science Centre School of Basic Medical Sciences, Shenzhen University, Shenzhen 518055, China
- Shenzhen University School of Pharmacy, Shenzhen University Medical School, Shenzhen 518055, China
| | - Jun Zhang
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Health Science Centre School of Basic Medical Sciences, Shenzhen University, Shenzhen 518055, China
- Shenzhen University School of Pharmacy, Shenzhen University Medical School, Shenzhen 518055, China
| | - Tingting Feng
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Health Science Centre School of Basic Medical Sciences, Shenzhen University, Shenzhen 518055, China
| | - Longjiang Di
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiaopeng Lu
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Health Science Centre School of Basic Medical Sciences, Shenzhen University, Shenzhen 518055, China
- Shenzhen University School of Pharmacy, Shenzhen University Medical School, Shenzhen 518055, China
| | - Hao Sheng
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Health Science Centre School of Basic Medical Sciences, Shenzhen University, Shenzhen 518055, China
| | - Qian Zhu
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Health Science Centre School of Basic Medical Sciences, Shenzhen University, Shenzhen 518055, China
- Shenzhen University School of Pharmacy, Shenzhen University Medical School, Shenzhen 518055, China
| | - Xinyun Chen
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Health Science Centre School of Basic Medical Sciences, Shenzhen University, Shenzhen 518055, China
| | - Jingchao Wang
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Health Science Centre School of Basic Medical Sciences, Shenzhen University, Shenzhen 518055, China
| | - Xingkai He
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Health Science Centre School of Basic Medical Sciences, Shenzhen University, Shenzhen 518055, China
| | - Yerkezhan Yerkinkazhina
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Health Science Centre School of Basic Medical Sciences, Shenzhen University, Shenzhen 518055, China
| | - Zhongyi Xie
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Health Science Centre School of Basic Medical Sciences, Shenzhen University, Shenzhen 518055, China
| | - Yuxin Shu
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Health Science Centre School of Basic Medical Sciences, Shenzhen University, Shenzhen 518055, China
- School of Basic Medical Sciences, Wannan Medical College, Wuhu, Anhui 241002, China
| | - Tianshu Kang
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Health Science Centre School of Basic Medical Sciences, Shenzhen University, Shenzhen 518055, China
| | - Huangqi Tang
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Health Science Centre School of Basic Medical Sciences, Shenzhen University, Shenzhen 518055, China
| | - Jinqin Qian
- Department of Urology, Peking University First Hospital, Beijing 100035, China
| | - Wei-Guo Zhu
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Health Science Centre School of Basic Medical Sciences, Shenzhen University, Shenzhen 518055, China
- Shenzhen University School of Pharmacy, Shenzhen University Medical School, Shenzhen 518055, China
- School of Basic Medical Sciences, Wannan Medical College, Wuhu, Anhui 241002, China
| |
Collapse
|
26
|
Yang Y, Wu J, Zhou W, Ji G, Dang Y. Protein posttranslational modifications in metabolic diseases: basic concepts and targeted therapies. MedComm (Beijing) 2024; 5:e752. [PMID: 39355507 PMCID: PMC11442990 DOI: 10.1002/mco2.752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 10/03/2024] Open
Abstract
Metabolism-related diseases, including diabetes mellitus, obesity, hyperlipidemia, and nonalcoholic fatty liver disease, are becoming increasingly prevalent, thereby posing significant threats to human health and longevity. Proteins, as the primary mediators of biological activities, undergo various posttranslational modifications (PTMs), including phosphorylation, ubiquitination, acetylation, methylation, and SUMOylation, among others, which substantially diversify their functions. These modifications are crucial in the physiological and pathological processes associated with metabolic disorders. Despite advancements in the field, there remains a deficiency in contemporary summaries addressing how these modifications influence processes of metabolic disease. This review aims to systematically elucidate the mechanisms through which PTM of proteins impact the progression of metabolic diseases, including diabetes, obesity, hyperlipidemia, and nonalcoholic fatty liver disease. Additionally, the limitations of the current body of research are critically assessed. Leveraging PTMs of proteins provides novel insights and therapeutic targets for the prevention and treatment of metabolic disorders. Numerous drugs designed to target these modifications are currently in preclinical or clinical trials. This review also provides a comprehensive summary. By elucidating the intricate interplay between PTMs and metabolic pathways, this study advances understanding of the molecular mechanisms underlying metabolic dysfunction, thereby facilitating the development of more precise and effective disease management strategies.
Collapse
Affiliation(s)
- Yunuo Yang
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Jiaxuan Wu
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Wenjun Zhou
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Guang Ji
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Yanqi Dang
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| |
Collapse
|
27
|
Zou D, Liao J, Xiao M, Liu L, Dai D, Xu M. Impaired SUMOylation of FoxA1 promotes nonalcoholic fatty liver disease through down-regulation of Sirt6. Cell Death Dis 2024; 15:674. [PMID: 39277582 PMCID: PMC11401847 DOI: 10.1038/s41419-024-07054-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 09/03/2024] [Accepted: 09/03/2024] [Indexed: 09/17/2024]
Abstract
Abnormal SUMOylation is implicated in non-alcoholic fatty liver disease (NAFLD) progression. Forkhead box protein A1 (FoxA1) has been shown to protect liver from steatosis, which was down-regulated in NAFLD. This study elucidated the role of FoxA1 deSUMOylation in NAFLD. NAFLD models were established in high-fat diet (HFD)-induced mice and palmitate acid (PAL)-treated hepatocytes. Hepatic steatosis was evaluated by biochemical and histological methods. Lipid droplet formation was determined by BODIPY and Oil red O staining. Target molecule levels were analyzed by RT-qPCR, Western blotting, and immunohistochemistry staining. SUMOylation of FoxA1 was determined by Ni-NTA pull-down assay and SUMOylation assay Ultra Kit. Protein interaction and ubiquitination were detected by Co-IP. Gene transcription was assessed by ChIP and dual luciferase reporter assays. Liver FoxA1 knockout mice developed severe liver steatosis, which could be ameliorated by sirtuin 6 (Sirt6) overexpression. Nutritional stresses reduced Sumo2/3-mediated FoxA1 SUMOylation at lysine residue K6, which promoted lipid droplet formation by repressing fatty acid β-oxidation. Moreover, Sirt6 was a target gene of FoxA1, and Sirt6 transcription activity was restrained by deSUMOylation of FoxA1 at site K6. Furthermore, nutritional stresses-induced deSUMOylation of FoxA1 promoted the ubiquitination and degradation of FoxA1 with assistance of murine double minute 2 (Mdm2). Finally, activating FoxA1 SUMOylation delayed the progression of NAFLD in mice. DeSUMOylation of FoxA1 at K6 promotes FoxA1 degradation and then inhibits Sirt6 transcription, thereby suppressing fatty acid β-oxidation and facilitating NAFLD development. Our findings suggest that FoxA1 SUMOylation activation might be a promising therapeutic strategy for NAFLD.
Collapse
Affiliation(s)
- Dongmei Zou
- The Department of Pediatric, Shenzhen Children's Hospital, China Medical University, Shenzhen, 518038, Guangdong Province, China
| | - Jinwen Liao
- The Department of Pediatric, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City, (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, 518172, Guangdong Province, China
| | - Min Xiao
- The Department of Pediatric, Shenzhen Children's Hospital, China Medical University, Shenzhen, 518038, Guangdong Province, China
| | - Liang Liu
- The Department of Pediatric, Shenzhen Children's Hospital, China Medical University, Shenzhen, 518038, Guangdong Province, China
| | - Dongling Dai
- The Department of Pediatric, Shenzhen Children's Hospital, China Medical University, Shenzhen, 518038, Guangdong Province, China
| | - Mingguo Xu
- The Department of Pediatric, Shenzhen Children's Hospital, China Medical University, Shenzhen, 518038, Guangdong Province, China.
- The Department of Pediatric, The Third People's Hospital of Longgang District Shenzhen, Shenzhen, 518112, Guangdong Province, China.
| |
Collapse
|
28
|
Wang JX, Zhang YY, Qian YC, Qian YF, Jin AH, Wang M, Luo Y, Qiao F, Zhang ML, Chen LQ, Du ZY. Inhibition of mitochondrial citrate shuttle alleviates metabolic syndromes induced by high-fat diet. Am J Physiol Cell Physiol 2024; 327:C737-C749. [PMID: 39069827 DOI: 10.1152/ajpcell.00194.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 06/07/2024] [Accepted: 06/23/2024] [Indexed: 07/30/2024]
Abstract
The mitochondrial citrate shuttle, which relies on the solute carrier family 25 member 1 (SLC25A1), plays a pivotal role in transporting citrate from the mitochondria to the cytoplasm. This shuttle supports glycolysis, lipid biosynthesis, and protein acetylation. Previous research has primarily focused on SLC25A1 in pathological models, particularly high-fat diet (HFD)-induced obesity. However, the impact of SLC25A1 inhibition on nutrient metabolism under HFD remains unclear. To address this gap, we used zebrafish (Danio rerio) and Nile tilapia (Oreochromis niloticus) to evaluate the effects of inhibiting Slc25a1. In zebrafish, we administered Slc25a1-specific inhibitors (CTPI-2) for 4 wk, whereas Nile tilapia received intraperitoneal injections of dsRNA to knock down slc25a1b for 7 days. Inhibition of the mitochondrial citrate shuttle effectively protected zebrafish from HFD-induced obesity, hepatic steatosis, and insulin resistance. Of note, glucose tolerance was unaffected. Inhibition of Slc25a1 altered hepatic protein acetylation patterns, with decreased cytoplasmic acetylation and increased mitochondrial acetylation. Under HFD conditions, Slc25a1 inhibition promoted fatty acid oxidation and reduced hepatic triglyceride (TAG) accumulation by deacetylating carnitine palmitoyltransferase 1a (Cpt1a). In addition, Slc25a1 inhibition triggered acetylation-induced inactivation of Pdhe1α, leading to a reduction in glucose oxidative catabolism. This was accompanied by enhanced glucose uptake and storage in zebrafish livers. Furthermore, Slc25a1 inhibition under HFD conditions activated the SIRT1/PGC1α pathway, promoting mitochondrial proliferation and enhancing oxidative phosphorylation for energy production. Our findings provide new insights into the role of nonhistone protein acetylation via the mitochondrial citrate shuttle in the development of hepatic lipid deposition and hyperglycemia caused by HFD.NEW & NOTEWORTHY The mitochondrial citrate shuttle is a crucial physiological process for maintaining metabolic homeostasis. In the present study, we found that inhibition of mitochondrial citrate shuttle (Slc25a1) could alleviate metabolic syndromes induced by high-fat diet (HFD) through remodeling hepatic protein acetylation modification. Briefly, Slc25a1 inhibition reduces hepatic triglyceride deposition by deacetylating Cpt1a and reduces glucose oxidative catabolism by acetylating Pdhe1α. Our study provides new insights into the treatment of diet-induced metabolic syndromes.
Collapse
Affiliation(s)
- Jun-Xian Wang
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, People's Republic of China
| | - Yan-Yu Zhang
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, People's Republic of China
| | - Yu-Cheng Qian
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, People's Republic of China
| | - Yi-Fan Qian
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, People's Republic of China
| | - An-Hui Jin
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, People's Republic of China
| | - Mai Wang
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, People's Republic of China
| | - Yuan Luo
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, People's Republic of China
| | - Fang Qiao
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, People's Republic of China
| | - Mei-Ling Zhang
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, People's Republic of China
| | - Li-Qiao Chen
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, People's Republic of China
| | - Zhen-Yu Du
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, People's Republic of China
| |
Collapse
|
29
|
Hong SH, Yu X, Zhu Y, Chen Y. Liver epigenomic signature associated with chronic oxidative stress in a mouse model of glutathione deficiency. Chem Biol Interact 2024; 398:111093. [PMID: 38830566 PMCID: PMC11223951 DOI: 10.1016/j.cbi.2024.111093] [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: 04/20/2024] [Revised: 05/17/2024] [Accepted: 05/31/2024] [Indexed: 06/05/2024]
Abstract
Oxidative stress is intimately involved in the pathogenesis of fatty liver disease (FLD). A major factor contributing to oxidative stress is the depletion of the ubiquitous antioxidant glutathione (GSH). Unexpectedly, chronic GSH deficiency renders glutamate-cysteine ligase modifier subunit (Gclm)-null mice protected from fatty liver injuries. Epigenetic regulation serves as an important cellular mechanism in modulating gene expression and disease outcome in FLD, although it is not well understood how systemic redox imbalance modifies the liver epigenome. In the current study, utilizing the Gclm-null mouse model, we aimed to elucidate redox-associated epigenomic changes and their implications in liver stress response. We performed high-throughput array-based DNA methylation profiling (MeDIP array) in 22,327 gene promoter regions (from -1300 bp to +500 bp of the Transcription Start Sites) in the liver and peripheral blood cells. Results from the MeDIP array demonstrate that, although global methylation enrichment in gene promoters did not change, low GSH resulted in prevalent demethylation at the individual promoter level. Such an effect likely attributed to a declined availability of the methyl donor S-adenosyl methionine (SAM) in Gclm-null liver. Functional enrichment analysis of liver target genes is suggestive of a potential role of epigenetic mechanisms in promoting cellular survival and lipid homeostasis in Gclm-null liver. In comparison with the liver tissue, MeDIP array in peripheral blood cells revealed a panel of 19 gene promoters that are candidate circulating biomarkers for hepatic epigenomic changes associated with chronic GSH deficiency. Collectively, our results provided new insights into the in vivo interplay between liver redox state and DNA methylation status. The current study laid the groundwork for future epigenetic/epigenomic investigations in experimental settings or human populations under conditions of liver oxidative stress induced by environmental or dietary challenges.
Collapse
Affiliation(s)
- Seong Hwi Hong
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, 06510, USA
| | - Xiaoqing Yu
- Department of Biostatistics, Yale School of Public Health, Yale University, New Haven, CT, 06510, USA
| | - Yong Zhu
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, 06510, USA
| | - Ying Chen
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, 06510, USA.
| |
Collapse
|
30
|
Zhuang W, Chen Z, Shu X, Zhang J, Zhu R, Shen M, Chen J, Zheng X. Establishment of a Steatosis Model in LMH Cells, Chicken Embryo Hepatocytes, and Liver Tissues Based on a Mixture of Sodium Oleate and Palmitic Acid. Animals (Basel) 2024; 14:2173. [PMID: 39123699 PMCID: PMC11311026 DOI: 10.3390/ani14152173] [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: 06/14/2024] [Revised: 07/16/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Research on hepatic steatosis in animal husbandry has been a prominent area of study. Developing an appropriate in vitro cellular steatosis model is crucial for comprehensively investigating the mechanisms involved in liver lipid deposition in poultry and for identifying potential interventions to address abnormalities in lipid metabolism. The research on the methods of in vitro liver steatosis in chickens, particularly the effects of different fat mixtures, is still lacking. In this study, LMH cells were utilized to investigate the effects of OA, SO, PA, SP, and their pairwise combinations on steatosis development, with the aim of identifying the optimal conditions for inducing steatosis. Analysis of triglyceride (TG) content in LMH cells revealed that OA and SP had limited efficacy in increasing TG content, while a combination of SO and PA in a 1:2 ratio exhibited the highest TG content. Moreover, Oil Red O staining results in LMH cells demonstrated that the combination treatment had a more pronounced induction effect compared to 0.375 mM SO. Additionally, RNA-seq analysis showed that 0.375 mM SO significantly influenced the expression of genes associated with fatty acid metabolism compared to the control group, whereas the combination of SO and PA led to an enrichment of key GO terms associated with programmed cell death. These findings suggest that varying conditions of cellular steatosis could lead to distinct disruptions in gene expression. The optimal conditions for inducing steatosis in LMH cells were also tested on chicken embryonic liver cells and embryos. TG detection and Oil Red O staining assays showed that the combination of SO and PA successfully induced steatosis. However, the gene expression pattern differed from that of LMH cells. This study lays the foundations for further investigations into avian hepatic steatosis.
Collapse
Affiliation(s)
- Wuchao Zhuang
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (W.Z.); (Z.C.); (X.S.); (J.Z.); (R.Z.); (M.S.)
| | - Ziwei Chen
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (W.Z.); (Z.C.); (X.S.); (J.Z.); (R.Z.); (M.S.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Xin Shu
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (W.Z.); (Z.C.); (X.S.); (J.Z.); (R.Z.); (M.S.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Jilong Zhang
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (W.Z.); (Z.C.); (X.S.); (J.Z.); (R.Z.); (M.S.)
| | - Runbang Zhu
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (W.Z.); (Z.C.); (X.S.); (J.Z.); (R.Z.); (M.S.)
| | - Manman Shen
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (W.Z.); (Z.C.); (X.S.); (J.Z.); (R.Z.); (M.S.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Jianfei Chen
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (W.Z.); (Z.C.); (X.S.); (J.Z.); (R.Z.); (M.S.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| | - Xiaotong Zheng
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (W.Z.); (Z.C.); (X.S.); (J.Z.); (R.Z.); (M.S.)
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang 212100, China
| |
Collapse
|
31
|
Huang B, Yu Z, Cui D, Du F. MAPKAP1 orchestrates macrophage polarization and lipid metabolism in fatty liver-enhanced colorectal cancer. Transl Oncol 2024; 45:101941. [PMID: 38692197 PMCID: PMC11070763 DOI: 10.1016/j.tranon.2024.101941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 03/02/2024] [Accepted: 03/16/2024] [Indexed: 05/03/2024] Open
Abstract
Various factors, including fatty liver and macrophage alterations, influence colorectal cancer (CRC). This study explores the mechanistic role of fatty liver in CRC progression, focusing on macrophage polarization and lipid metabolism. A murine fatty liver model was created with a high-fat diet (HFD), and CRC was induced using AOM and DSS. Single-cell transcriptome sequencing (scRNA-seq) identified MAPKAP1 as a critical gene promoting CRC via M2 macrophage polarization and lipid metabolism reprogramming. Prognosis analysis on the TCGA-CRC dataset confirmed MAPKAP1's significance. In vitro and in vivo experiments demonstrated that EVs from fatty liver cells enhanced MAPKAP1 expression, accelerating CRC development and metastasis. HFD exacerbated CRC, but fatty acid inhibitors delayed progression. Fatty liver upregulates MAPKAP1, driving M2 macrophage polarization and lipid metabolism changes, worsening CRC. These findings suggest potential therapeutic strategies for CRC, particularly targeting lipid metabolism and macrophage-mediated tumor promotion.
Collapse
Affiliation(s)
- Bo Huang
- Department of Hypertension, The Affiliated Hospital of Guizhou Medical University, No.28, Guimedical Street, Yunyan District, Guiyang City, Guizhou Province, PR China.
| | - Zhenqiu Yu
- Department of Hypertension, The Affiliated Hospital of Guizhou Medical University, No.28, Guimedical Street, Yunyan District, Guiyang City, Guizhou Province, PR China.
| | - Dejun Cui
- Department of Gastroenterology, Guizhou Provincial People's Hospital, PR China.
| | - Fawang Du
- Department of Hypertension, The Affiliated Hospital of Guizhou Medical University, No.28, Guimedical Street, Yunyan District, Guiyang City, Guizhou Province, PR China
| |
Collapse
|
32
|
Xue X, Wang L, Wu R, Li Y, Liu R, Ma Z, Jia K, Zhang Y, Li X. Si-Wu-Tang alleviates metabolic dysfunction-associated fatty liver disease by inhibiting ACSL4-mediated arachidonic acid metabolism and ferroptosis in MCD diet-fed mice. Chin Med 2024; 19:79. [PMID: 38844978 PMCID: PMC11157816 DOI: 10.1186/s13020-024-00953-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/31/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Metabolic dysfunction-associated fatty liver disease (MAFLD) is a prevalent chronic liver disease worldwide. Si-Wu-Tang (SWT), a traditional Chinese medicine decoction has shown therapeutic effects on various liver diseases. However, the hepatoprotective effects and underlying mechanism of SWT on MAFLD remain unclear. METHODS First, a methionine-choline-deficient (MCD) diet-fed mice model was used and lipidomic analysis and transcriptomic analysis were performed. The contents of total iron ions, ferrous ions, and lipid peroxidation were detected and Prussian blue staining was performed to confirm the protective effects of SWT against ferroptosis. Finally, chemical characterization and network pharmacological analysis were employed to identify the potential active ingredients. RESULTS Serological and hepatic histopathological findings indicated SWT's discernible therapeutic impact on MCD diet-induced MAFLD. Lipidomic analysis revealed that SWT improved intrahepatic lipid accumulation by inhibiting TG synthesis and promoting TG transport. Transcriptomic analysis suggested that SWT ameliorated abnormal FA metabolism by inhibiting FA synthesis and promoting FA β-oxidation. Then, ferroptosis phenotype experiments revealed that SWT could effectively impede hepatocyte ferroptosis, which was induced by long-chain acyl-CoA synthetase 4 (ACSL4)-mediated esterification of arachidonic acid (AA). Finally, chemical characterization and network pharmacological analysis identified that paeoniflorin and other active ingredients might be responsible for the regulative effects against ferroptosis and MAFLD. CONCLUSION In conclusion, our study revealed the intricate mechanism through which SWT improved MCD diet-induced MAFLD by targeting FA metabolism and ferroptosis in hepatocytes, thus offering a novel therapeutic approach for the treatment of MAFLD and its complications.
Collapse
Affiliation(s)
- Xiaoyong Xue
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Le Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Ruiyu Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China
| | - Yufei Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China
| | - Runping Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China
| | - Zhi Ma
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Kexin Jia
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yinhao Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xiaojiaoyang Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
33
|
Tian C, Huang R, Xiang M. SIRT1: Harnessing multiple pathways to hinder NAFLD. Pharmacol Res 2024; 203:107155. [PMID: 38527697 DOI: 10.1016/j.phrs.2024.107155] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/04/2024] [Accepted: 03/21/2024] [Indexed: 03/27/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) encompasses hepatic steatosis, non-alcoholic steatohepatitis (NASH), fibrosis, cirrhosis, and hepatocellular carcinoma. It is the primary cause of chronic liver disorders, with a high prevalence but no approved treatment. Therefore, it is indispensable to find a trustworthy therapy for NAFLD. Recently, mounting evidence illustrates that Sirtuin 1 (SIRT1) is strongly associated with NAFLD. SIRT1 activation or overexpression attenuate NAFLD, while SIRT1 deficiency aggravates NAFLD. Besides, an array of therapeutic agents, including natural compounds, synthetic compounds, traditional Chinese medicine formula, and stem cell transplantation, alleviates NALFD via SIRT1 activation or upregulation. Mechanically, SIRT1 alleviates NAFLD by reestablishing autophagy, enhancing mitochondrial function, suppressing oxidative stress, and coordinating lipid metabolism, as well as reducing hepatocyte apoptosis and inflammation. In this review, we introduced the structure and function of SIRT1 briefly, and summarized the effect of SIRT1 on NAFLD and its mechanism, along with the application of SIRT1 agonists in treating NAFLD.
Collapse
Affiliation(s)
- Cheng Tian
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Rongrong Huang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ming Xiang
- Department of Pharmacology, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
34
|
Fu Y, Wang Z, Qin H. Examining the Pathogenesis of MAFLD and the Medicinal Properties of Natural Products from a Metabolic Perspective. Metabolites 2024; 14:218. [PMID: 38668346 PMCID: PMC11052500 DOI: 10.3390/metabo14040218] [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: 03/17/2024] [Revised: 04/06/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Metabolic-associated fatty liver disease (MAFLD), characterized primarily by hepatic steatosis, has become the most prevalent liver disease worldwide, affecting approximately two-fifths of the global population. The pathogenesis of MAFLD is extremely complex, and to date, there are no approved therapeutic drugs for clinical use. Considerable evidence indicates that various metabolic disorders play a pivotal role in the progression of MAFLD, including lipids, carbohydrates, amino acids, and micronutrients. In recent years, the medicinal properties of natural products have attracted widespread attention, and numerous studies have reported their efficacy in ameliorating metabolic disorders and subsequently alleviating MAFLD. This review aims to summarize the metabolic-associated pathological mechanisms of MAFLD, as well as the natural products that regulate metabolic pathways to alleviate MAFLD.
Collapse
Affiliation(s)
| | | | - Hong Qin
- Department of Nutrition and Food Hygiene, Xiangya School of Public Health, Central South University, Changsha 410006, China; (Y.F.); (Z.W.)
| |
Collapse
|
35
|
Lai Y, Gao Y, Lin J, Liu F, Yang L, Zhou J, Xue Y, Li Y, Chang Z, Li J, Chao T, Chen J, Cheng X, Gao X, Li X, Lu F, Chu Q, Wang W. Dietary elaidic acid boosts tumoral antigen presentation and cancer immunity via ACSL5. Cell Metab 2024; 36:822-838.e8. [PMID: 38350448 DOI: 10.1016/j.cmet.2024.01.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 12/12/2023] [Accepted: 01/20/2024] [Indexed: 02/15/2024]
Abstract
Immunomodulatory effects of long-chain fatty acids (LCFAs) and their activating enzyme, acyl-coenzyme A (CoA) synthetase long-chain family (ACSL), in the tumor microenvironment remain largely unknown. Here, we find that ACSL5 functions as an immune-dependent tumor suppressor. ACSL5 expression sensitizes tumors to PD-1 blockade therapy in vivo and the cytotoxicity mediated by CD8+ T cells in vitro via regulation of major histocompatibility complex class I (MHC-I)-mediated antigen presentation. Through screening potential substrates for ACSL5, we further identify that elaidic acid (EA), a trans LCFA that has long been considered harmful to human health, phenocopies to enhance MHC-I expression. EA supplementation can suppress tumor growth and sensitize PD-1 blockade therapy. Clinically, ACSL5 expression is positively associated with improved survival in patients with lung cancer, and plasma EA level is also predictive for immunotherapy efficiency. Our findings provide a foundation for enhancing immunotherapy through either targeting ACSL5 or metabolic reprogramming of antigen presentation via dietary EA supplementation.
Collapse
Affiliation(s)
- Yongfeng Lai
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Gao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junhong Lin
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Fangfang Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liguo Yang
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Zhou
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Xue
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Li
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Zhenzhen Chang
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Li
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China
| | - Tengfei Chao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xianfu Gao
- Shanghai ProfLeader Biotech Co., Ltd, Shanghai, China
| | - Xiong Li
- Department of Gynecology & Obstetrics, the Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fujia Lu
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China.
| | - Qian Chu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Weimin Wang
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China; Cell Architecture Research Institute, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
36
|
Wang S, Zhang X, Hou Y, Zhang Y, Chen J, Gao S, Duan H, Gu S, Yu S, Cai Y. SIRT6 activates PPARα to improve doxorubicin-induced myocardial cell aging and damage. Chem Biol Interact 2024; 392:110920. [PMID: 38395252 DOI: 10.1016/j.cbi.2024.110920] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 01/26/2024] [Accepted: 02/18/2024] [Indexed: 02/25/2024]
Abstract
The Sirtuins family, formally known as the Silent Information Regulator Factors, constitutes a highly conserved group of histone deacetylases. Recent studies have illuminated SIRT6's role in doxorubicin (DOX)-induced oxidative stress and apoptosis within myocardial cells. Nevertheless, the extent of SIRT6's impact on DOX-triggered myocardial cell aging and damage remains uncertain, with the associated mechanisms yet to be fully understood. In our research, we examined the influence of SIRT6 on DOX-induced cardiomyocyte senescence using β-galactosidase and γ-H2AX staining. Additionally, we gauged the mRNA expression of senescence-associated genes, namely p16, p21, and p53, through Real-time PCR. Employing ELISA assay kits, MDA, and total SOD activity assay kits, we measured inflammatory factors TNF-α, IL-6, and IL-1β, alongside oxidative stress-related indicators. The results unequivocally indicated that SIRT6 overexpression robustly inhibited DOX-induced cardiomyocyte senescence. Furthermore, we established that SIRT6 overexpression suppressed the inflammatory response and oxidative stress induced by DOX in cardiomyocytes. Conversely, silencing SIRT6 exacerbated DOX-induced cardiomyocyte injury. Our investigations further unveiled that SIRT6 upregulated the expression of genes CD36, CPT1, LCAD, MCAD associated with fatty acid oxidation through its interaction with PPARα, thereby exerting anti-aging effects. In vivo, the overexpression of SIRT6 was observed to restore DOX-induced declines in EF and FS to normal levels in mice. Echocardiography and HE staining revealed the restoration of cardiomyocyte alignment, affording protection against DOX-induced myocardial senescence and injury. The findings from this study suggest that SIRT6 holds significant promise as a therapeutic target for mitigating DOX-induced cardiomyopathy.
Collapse
Affiliation(s)
- Shulin Wang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xuan Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Afffliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yanhong Hou
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Afffliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yuliang Zhang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jiamin Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Afffliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shuhan Gao
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Afffliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Huiying Duan
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Afffliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shaoju Gu
- Laboratory Animal Centre, Guangzhou Medical University, Guangzhou, China.
| | - Shanshan Yu
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.
| | - Yi Cai
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, China; Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Afffliated Hospital, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
37
|
Samoilova EM, Romanov SE, Chudakova DA, Laktionov PP. Role of sirtuins in epigenetic regulation and aging control. Vavilovskii Zhurnal Genet Selektsii 2024; 28:215-227. [PMID: 38680178 PMCID: PMC11043508 DOI: 10.18699/vjgb-24-26] [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: 11/12/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 05/01/2024] Open
Abstract
Advances in modern healthcare in developed countries make it possible to extend the human lifespan, which is why maintaining active longevity is becoming increasingly important. After the sirtuin (SIRT) protein family was discovered, it started to be considered as a significant regulator of the physiological processes associated with aging. SIRT has deacetylase, deacylase, and ADP-ribosyltransferase activity and modifies a variety of protein substrates, including chromatin components and regulatory proteins. This multifactorial regulatory system affects many processes: cellular metabolism, mitochondrial functions, epigenetic regulation, DNA repair and more. As is expected, the activity of sirtuin proteins affects the manifestation of classic signs of aging in the body, such as cellular senescence, metabolic disorders, mitochondrial dysfunction, genomic instability, and the disruption of epigenetic regulation. Changes in the SIRT activity in human cells can also be considered a marker of aging and are involved in the genesis of various age-dependent disorders. Additionally, experimental data obtained in animal models, as well as data from population genomic studies, suggest a SIRT effect on life expectancy. At the same time, the diversity of sirtuin functions and biochemical substrates makes it extremely complicated to identify cause-and-effect relationships and the direct role of SIRT in controlling the functional state of the body. However, the SIRT influence on the epigenetic regulation of gene expression during the aging process and the development of disorders is one of the most important aspects of maintaining the homeostasis of organs and tissues. The presented review centers on the diversity of SIRT in humans and model animals. In addition to a brief description of the main SIRT enzymatic and biological activity, the review discusses its role in the epigenetic regulation of chromatin structure, including the context of the development of genome instability associated with aging. Studies on the functional connection between SIRT and longevity, as well as its effect on pathological processes associated with aging, such as chronic inflammation, fibrosis, and neuroinflammation, have been critically analyzed.
Collapse
Affiliation(s)
- E M Samoilova
- Novosibirsk State University, Novosibirsk, Russia Engelhardt Institute of Molecular Biology of the Russian Academy of Sciences, Moscow, Russia
| | - S E Romanov
- Novosibirsk State University, Novosibirsk, Russia Institute of Molecular and Cellular Biology of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - D A Chudakova
- Federal Center of Brain Research and Neurotechnologies of the Federal Medical Biological Agency of Russia, Moscow, Russia
| | - P P Laktionov
- Novosibirsk State University, Novosibirsk, Russia Institute of Molecular and Cellular Biology of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
38
|
Li J, Ni Y, Zhang Y, Liu H. GBA3 promotes fatty acid oxidation and alleviates non-alcoholic fatty liver by increasing CPT2 transcription. Aging (Albany NY) 2024; 16:4591-4608. [PMID: 38428407 PMCID: PMC10968678 DOI: 10.18632/aging.205616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 01/23/2024] [Indexed: 03/03/2024]
Abstract
BACKGROUND Excessive lipids accumulation and hepatocytes death are prominent characteristics of non-alcoholic fatty liver disease (NAFLD). Nonetheless, the precise pathophysiological mechanisms are not fully elucidated. METHODS HepG2 cells stimulated with palmitic acids and rats fed with high-fat diet were used as models for NAFLD. The impact of Glucosylceramidase Beta 3 (GBA3) on fatty acid oxidation (FAO) was assessed using Seahorse metabolic analyzer. Lipid content was measured both in vitro and in vivo. To evaluate NAFLD progression, histological analysis was performed along with measurements of inflammatory factors and liver enzyme levels. Western blot and immunohistochemistry were employed to examine the activity levels of necroptosis. Flow cytometry and reactive oxygen species (ROS) staining were utilized to assess levels of oxidative stress. RESULTS GBA3 promoted FAO and enhanced the mitochondrial membrane potential without affecting glycolysis. These reduced the lipid accumulation. Rats supplemented with GBA3 exhibited lower levels of inflammatory factors and liver enzymes, resulting in a slower progression of NAFLD. GBA3 overexpression reduced ROS and the ratio of cell apoptosis. Phosphorylation level was reduced in the essential mediator, MLKL, implicated in necroptosis. Mechanistically, as a transcriptional coactivator, GBA3 promoted the expression of Carnitine Palmitoyltransferase 2 (CPT2), which resulted in enhanced FAO. CONCLUSIONS Increased FAO resulting from GBA3 reduced oxidative stress and the production of ROS, thereby inhibiting necroptosis and delaying the progression of NAFLD. Our research offers novel insights into the potential therapeutic applications of GBA3 and FAO in the management and treatment of NAFLD.
Collapse
Affiliation(s)
- Juyi Li
- Department of Endocrinology, Geriatrics Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230001, Anhui, China
| | - Yingqun Ni
- Department of Endocrinology, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230001, Anhui, China
| | - Yuanyuan Zhang
- Department of Endocrinology, Geriatrics Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230001, Anhui, China
| | - Huaizhen Liu
- Department of Endocrinology, Geriatrics Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230001, Anhui, China
| |
Collapse
|
39
|
Wang R, Zhang J, Ren H, Qi S, Xie L, Xie H, Shang Z, Liu C. Dysregulated palmitic acid metabolism promotes the formation of renal calcium-oxalate stones through ferroptosis induced by polyunsaturated fatty acids/phosphatidic acid. Cell Mol Life Sci 2024; 81:85. [PMID: 38345762 PMCID: PMC10861707 DOI: 10.1007/s00018-024-05145-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 01/04/2024] [Accepted: 01/25/2024] [Indexed: 02/15/2024]
Abstract
The pathogenesis of renal calcium-oxalate (CaOx) stones is complex and influenced by various metabolic factors. In parallel, palmitic acid (PA) has been identified as an upregulated lipid metabolite in the urine and serum of patients with renal CaOx stones via untargeted metabolomics. Thus, this study aimed to mechanistically assess whether PA is involved in stone formation. Lipidomics analysis of PA-treated renal tubular epithelial cells compared with the control samples revealed that α-linoleic acid and α-linolenic acid were desaturated and elongated, resulting in the formation of downstream polyunsaturated fatty acids (PUFAs). In correlation, the levels of fatty acid desaturase 1 and 2 (FADS1 and FADS2) and peroxisome proliferator-activated receptor α (PPARα) in these cells treated with PA were increased relative to the control levels, suggesting that PA-induced upregulation of PPARα, which in turn upregulated these two enzymes, forming the observed PUFAs. Lipid peroxidation occurred in these downstream PUFAs under oxidative stress and Fenton Reaction. Furthermore, transcriptomics analysis revealed significant changes in the expression levels of ferroptosis-related genes in PA-treated renal tubular epithelial cells, induced by PUFA peroxides. In addition, phosphatidyl ethanolamine binding protein 1 (PEBP1) formed a complex with 15-lipoxygenase (15-LO) to exacerbate PUFA peroxidation under protein kinase C ζ (PKC ζ) phosphorylation, and PKC ζ was activated by phosphatidic acid derived from PA. In conclusion, this study found that the formation of renal CaOx stones is promoted by ferroptosis of renal tubular epithelial cells resulting from PA-induced dysregulation of PUFA and phosphatidic acid metabolism, and PA can promote the renal adhesion and deposition of CaOx crystals by injuring renal tubular epithelial cells, consequently upregulating adhesion molecules. Accordingly, this study provides a new theoretical basis for understanding the correlation between fatty acid metabolism and the formation of renal CaOx stones, offering potential targets for clinical applications.
Collapse
Affiliation(s)
- Rui Wang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Jingdong Zhang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Haotian Ren
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Shiyong Qi
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Linguo Xie
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Haijie Xie
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhiqun Shang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China.
| | - Chunyu Liu
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China.
| |
Collapse
|
40
|
Jin Q, Ma F, Liu T, Yang L, Mao H, Wang Y, Peng L, Li P, Zhan Y. Sirtuins in kidney diseases: potential mechanism and therapeutic targets. Cell Commun Signal 2024; 22:114. [PMID: 38347622 PMCID: PMC10860260 DOI: 10.1186/s12964-023-01442-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 12/12/2023] [Indexed: 02/15/2024] Open
Abstract
Sirtuins, which are NAD+-dependent class III histone deacetylases, are involved in various biological processes, including DNA damage repair, immune inflammation, oxidative stress, mitochondrial homeostasis, autophagy, and apoptosis. Sirtuins are essential regulators of cellular function and organismal health. Increasing evidence suggests that the development of age-related diseases, including kidney diseases, is associated with aberrant expression of sirtuins, and that regulation of sirtuins expression and activity can effectively improve kidney function and delay the progression of kidney disease. In this review, we summarise current studies highlighting the role of sirtuins in renal diseases. First, we discuss sirtuin family members and their main mechanisms of action. We then outline the possible roles of sirtuins in various cell types in kidney diseases. Finally, we summarise the compounds that activate or inhibit sirtuin activity and that consequently ameliorate renal diseases. In conclusion, targeted modulation of sirtuins is a potential therapeutic strategy for kidney diseases. Video Abstract.
Collapse
Affiliation(s)
- Qi Jin
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fang Ma
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tongtong Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liping Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Huimin Mao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuyang Wang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liang Peng
- China-Japan Friendship Hospital, Institute of Clinical Medical Sciences, Beijing, China.
| | - Ping Li
- China-Japan Friendship Hospital, Institute of Clinical Medical Sciences, Beijing, China.
| | - Yongli Zhan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
41
|
Theys C, Vanderhaeghen T, Van Dijck E, Peleman C, Scheepers A, Ibrahim J, Mateiu L, Timmermans S, Vanden Berghe T, Francque SM, Van Hul W, Libert C, Vanden Berghe W. Loss of PPARα function promotes epigenetic dysregulation of lipid homeostasis driving ferroptosis and pyroptosis lipotoxicity in metabolic dysfunction associated Steatotic liver disease (MASLD). FRONTIERS IN MOLECULAR MEDICINE 2024; 3:1283170. [PMID: 39086681 PMCID: PMC11285560 DOI: 10.3389/fmmed.2023.1283170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 12/14/2023] [Indexed: 08/02/2024]
Abstract
Metabolic Dysfunction Associated Steatotic Liver Disease (MASLD) is a growing epidemic with an estimated prevalence of 20%-30% in Europe and the most common cause of chronic liver disease worldwide. The onset and progression of MASLD are orchestrated by an interplay of the metabolic environment with genetic and epigenetic factors. Emerging evidence suggests altered DNA methylation pattern as a major determinant of MASLD pathogenesis coinciding with progressive DNA hypermethylation and gene silencing of the liver-specific nuclear receptor PPARα, a key regulator of lipid metabolism. To investigate how PPARα loss of function contributes to epigenetic dysregulation in MASLD pathology, we studied DNA methylation changes in liver biopsies of WT and hepatocyte-specific PPARα KO mice, following a 6-week CDAHFD (choline-deficient, L-amino acid-defined, high-fat diet) or chow diet. Interestingly, genetic loss of PPARα function in hepatocyte-specific KO mice could be phenocopied by a 6-week CDAHFD diet in WT mice which promotes epigenetic silencing of PPARα function via DNA hypermethylation, similar to MASLD pathology. Remarkably, genetic and lipid diet-induced loss of PPARα function triggers compensatory activation of multiple lipid sensing transcription factors and epigenetic writer-eraser-reader proteins, which promotes the epigenetic transition from lipid metabolic stress towards ferroptosis and pyroptosis lipid hepatoxicity pathways associated with advanced MASLD. In conclusion, we show that PPARα function is essential to support lipid homeostasis and to suppress the epigenetic progression of ferroptosis-pyroptosis lipid damage associated pathways towards MASLD fibrosis.
Collapse
Affiliation(s)
- Claudia Theys
- Protein Chemistry, Proteomics and Epigenetic Signaling (PPES), Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Tineke Vanderhaeghen
- Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | | | - Cedric Peleman
- Laboratory of Experimental Medicine and Pediatrics, Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Pathophysiology Lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Anne Scheepers
- Center of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Joe Ibrahim
- Center of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Ligia Mateiu
- Center of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Steven Timmermans
- Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Tom Vanden Berghe
- Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Pathophysiology Lab, Infla-Med Centre of Excellence, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Sven M. Francque
- Laboratory of Experimental Medicine and Pediatrics, Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem, Belgium
| | - Wim Van Hul
- Center of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Claude Libert
- Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Wim Vanden Berghe
- Protein Chemistry, Proteomics and Epigenetic Signaling (PPES), Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
42
|
Jin S, Chen P, Yang J, Li D, Liu X, Zhang Y, Xia Q, Li Y, Chen G, Li Y, Tong Y, Yu W, Fan X, Lin H. Phocaeicola vulgatus alleviates diet-induced metabolic dysfunction-associated steatotic liver disease progression by downregulating histone acetylation level via 3-HPAA. Gut Microbes 2024; 16:2309683. [PMID: 38312099 PMCID: PMC10854360 DOI: 10.1080/19490976.2024.2309683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 01/19/2024] [Indexed: 02/06/2024] Open
Abstract
Diet-induced metabolic dysfunction-associated steatotic liver disease (MASLD) is a prevalent metabolic disorder with limited effective interventions available. A novel approach to address this issue is through gut microbiota-based therapy. In our study, we utilized multi-omics analysis to identify Phocaeicola vulgatus (P. vulgatus) as a potential probiotic for the treatment of MASLD. Our findings from murine models clearly illustrate that the supplementation of P. vulgatus mitigates the development of MASLD. This beneficial effect is partly attributed to the metabolite 3-Hydroxyphenylacetic acid (3-HPAA) produced by P. vulgatus, which reduces the acetylation levels of H3K27 and downregulates the transcription of Squalene Epoxidase (SQLE), a rate-limiting enzyme in steroid biosynthesis that promotes lipid accumulation in liver cells. This study underscores the significant role of P. vulgatus in the development of MASLD and the critical importance of its metabolite 3-HPAA in regulating lipid homeostasis. These findings offer a promising avenue for early intervention therapy in the context of MASLD.
Collapse
Affiliation(s)
- Shengxi Jin
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Peng Chen
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jing Yang
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Duguang Li
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaolong Liu
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiyin Zhang
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiming Xia
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiling Li
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Guoqiao Chen
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yixuan Li
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yifan Tong
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Weihua Yu
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoxiao Fan
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui Lin
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| |
Collapse
|
43
|
Dong T, Li J, Liu Y, Zhou S, Wei X, Hua H, Tang K, Zhang X, Wang Y, Wu Z, Gao C, Zhang H. Roles of immune dysregulation in MASLD. Biomed Pharmacother 2024; 170:116069. [PMID: 38147736 DOI: 10.1016/j.biopha.2023.116069] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/14/2023] [Accepted: 12/21/2023] [Indexed: 12/28/2023] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common chronic liver disease worldwide. Its occurrence and progression involve the process from simple hepatic steatosis to metabolic dysfunction associated steatohepatitis (MASH), which could develop into advanced liver fibrosis, cirrhosis, or hepatocellular carcinoma (HCC). Growing evidences support that the pathogenesis and progression of MASLD are closely related to immune system dysfunction. This review aims to summarize the association of MASLD with immune disorders and the prospect of using immunotherapy for MASLD.
Collapse
Affiliation(s)
- Tingyu Dong
- The Second Clinical Medical College of Anhui Medical University, Hefei 230032, China; Department of Biochemistry and Molecular Biology, Metabolic Disease Research Center, School of Basic Medicine, Anhui Medical University, Hefei 230032, China
| | - Jiajin Li
- The Second Clinical Medical College of Anhui Medical University, Hefei 230032, China; Department of Biochemistry and Molecular Biology, Metabolic Disease Research Center, School of Basic Medicine, Anhui Medical University, Hefei 230032, China
| | - Yuqing Liu
- Department of Biochemistry and Molecular Biology, Metabolic Disease Research Center, School of Basic Medicine, Anhui Medical University, Hefei 230032, China
| | - Shikai Zhou
- The Second Clinical Medical College of Anhui Medical University, Hefei 230032, China
| | - Xiang Wei
- Department of Biochemistry and Molecular Biology, Metabolic Disease Research Center, School of Basic Medicine, Anhui Medical University, Hefei 230032, China
| | - Hongting Hua
- Department of Otorhinolaryngology Head and Neck Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Kechao Tang
- Department of Biochemistry and Molecular Biology, Metabolic Disease Research Center, School of Basic Medicine, Anhui Medical University, Hefei 230032, China
| | - Xiaomin Zhang
- Department of Biochemistry and Molecular Biology, Metabolic Disease Research Center, School of Basic Medicine, Anhui Medical University, Hefei 230032, China
| | - Yiming Wang
- Department of Biochemistry and Molecular Biology, Metabolic Disease Research Center, School of Basic Medicine, Anhui Medical University, Hefei 230032, China
| | - Zhen Wu
- Department of Biochemistry and Molecular Biology, Metabolic Disease Research Center, School of Basic Medicine, Anhui Medical University, Hefei 230032, China
| | - Chaobing Gao
- Department of Otorhinolaryngology Head and Neck Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China.
| | - Huabing Zhang
- Department of Biochemistry and Molecular Biology, Metabolic Disease Research Center, School of Basic Medicine, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
44
|
Quan J, Wen X, Su G, Zhong Y, Huang T, Xiong Z, Huang J, Lv Y, Li S, Luo S, Luo C, Cai X, Lai X, Xiang Y, Zheng SG, Shao Y, Lin H, Gao X, Tang J, Lai T. Epithelial SIRT6 governs IL-17A pathogenicity and drives allergic airway inflammation and remodeling. Nat Commun 2023; 14:8525. [PMID: 38135684 PMCID: PMC10746710 DOI: 10.1038/s41467-023-44179-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Dysregulation of IL-17A is closely associated with airway inflammation and remodeling in severe asthma. However, the molecular mechanisms by which IL-17A is regulated remain unclear. Here we identify epithelial sirtuin 6 (SIRT6) as an epigenetic regulator that governs IL-17A pathogenicity in severe asthma. Mice with airway epithelial cell-specific deletion of Sirt6 are protected against allergen-induced airway inflammation and remodeling via inhibiting IL-17A-mediated inflammatory chemokines and mesenchymal reprogramming. Mechanistically, SIRT6 directly interacts with RORγt and mediates RORγt deacetylation at lysine 192 via its PPXY motifs. SIRT6 promotes RORγt recruitment to the IL-17A gene promoter and enhances its transcription. In severe asthma patients, high expression of SIRT6 positively correlates with airway remodeling and disease severity. SIRT6 inhibitor (OSS_128167) treatment significantly attenuates airway inflammation and remodeling in mice. Collectively, these results uncover a function for SIRT6 in regulating IL-17A pathogenicity in severe asthma, implicating SIRT6 as a potential therapeutic target for severe asthma.
Collapse
Affiliation(s)
- Jingyun Quan
- Department of Respiratory and Critical Care Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523710, China
- Department of Health Management & Physical Examination Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Xiaoxia Wen
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Guomei Su
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Yu Zhong
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Tong Huang
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Zhilin Xiong
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Jiewen Huang
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Yingying Lv
- Department of Respiratory and Critical Care Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523710, China
| | - Shihai Li
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Shuhua Luo
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Chaole Luo
- Department of Respiratory and Critical Care Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523710, China
| | - Xin Cai
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Xianwen Lai
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Yuanyuan Xiang
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Song Guo Zheng
- Dongguan Key Laboratory of Chronic Inflammatory Diseases, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523710, China
| | - Yiming Shao
- Department of Respiratory and Critical Care Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523710, China
| | - Haitao Lin
- Department of Health Management & Physical Examination Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Xiao Gao
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
| | - Jing Tang
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
| | - Tianwen Lai
- Department of Respiratory and Critical Care Medicine, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, 523710, China.
- Institute of Respiratory Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China.
| |
Collapse
|
45
|
Zhang J, Chen F, Tian Y, Xu W, Zhu Q, Li Z, Qiu L, Lu X, Peng B, Liu X, Gan H, Liu B, Xu X, Zhu WG. PARylated PDHE1α generates acetyl-CoA for local chromatin acetylation and DNA damage repair. Nat Struct Mol Biol 2023; 30:1719-1734. [PMID: 37735618 DOI: 10.1038/s41594-023-01107-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 08/21/2023] [Indexed: 09/23/2023]
Abstract
Chromatin relaxation is a prerequisite for the DNA repair machinery to access double-strand breaks (DSBs). Local histones around the DSBs then undergo prompt changes in acetylation status, but how the large demands of acetyl-CoA are met is unclear. Here, we report that pyruvate dehydrogenase 1α (PDHE1α) catalyzes pyruvate metabolism to rapidly provide acetyl-CoA in response to DNA damage. We show that PDHE1α is quickly recruited to chromatin in a polyADP-ribosylation-dependent manner, which drives acetyl-CoA generation to support local chromatin acetylation around DSBs. This process increases the formation of relaxed chromatin to facilitate repair-factor loading, genome stability and cancer cell resistance to DNA-damaging treatments in vitro and in vivo. Indeed, we demonstrate that blocking polyADP-ribosylation-based PDHE1α chromatin recruitment attenuates chromatin relaxation and DSB repair efficiency, resulting in genome instability and restored radiosensitivity. These findings support a mechanism in which chromatin-associated PDHE1α locally generates acetyl-CoA to remodel the chromatin environment adjacent to DSBs and promote their repair.
Collapse
Affiliation(s)
- Jun Zhang
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, China
| | - Feng Chen
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, China
| | - Yuan Tian
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, China
| | - Wenchao Xu
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, China
| | - Qian Zhu
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, China
| | - Zhenhai Li
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, China
| | - Lingyu Qiu
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, China
| | - Xiaopeng Lu
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, China
| | - Bin Peng
- Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Cell Biology and Medical Genetics, Shenzhen University Medical School, Shenzhen, China
| | - Xiangyu Liu
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, China
| | - Haiyun Gan
- Shenzhen Key Laboratory of Synthetic Genomics, Guangdong Provincial Key Laboratory of Synthetic Genomics, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Baohua Liu
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, China
- Shenzhen Key Laboratory for Systemic Aging and Intervention, National Engineering Research Center for Biotechnology (Shenzhen), Shenzhen University Medical School, Shenzhen, China
| | - Xingzhi Xu
- Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Cell Biology and Medical Genetics, Shenzhen University Medical School, Shenzhen, China
| | - Wei-Guo Zhu
- International Cancer Center, Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Marshall Laboratory of Biomedical Engineering, Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, China.
| |
Collapse
|
46
|
Wang Y, Liu T, Cai Y, Liu W, Guo J. SIRT6's function in controlling the metabolism of lipids and glucose in diabetic nephropathy. Front Endocrinol (Lausanne) 2023; 14:1244705. [PMID: 37876546 PMCID: PMC10591331 DOI: 10.3389/fendo.2023.1244705] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/21/2023] [Indexed: 10/26/2023] Open
Abstract
Diabetic nephropathy (DN) is a complication of diabetes mellitus (DM) and the main cause of excess mortality in patients with type 2 DM. The pathogenesis and progression of DN are closely associated with disorders of glucose and lipid metabolism. As a member of the sirtuin family, SIRT6 has deacetylation, defatty-acylation, and adenosine diphosphate-ribosylation enzyme activities as well as anti-aging and anticancer activities. SIRT6 plays an important role in glucose and lipid metabolism and signaling, especially in DN. SIRT6 improves glucose and lipid metabolism by controlling glycolysis and gluconeogenesis, affecting insulin secretion and transmission and regulating lipid decomposition, transport, and synthesis. Targeting SIRT6 may provide a new therapeutic strategy for DN by improving glucose and lipid metabolism. This review elaborates on the important role of SIRT6 in glucose and lipid metabolism, discusses the potential of SIRT6 as a therapeutic target to improve glucose and lipid metabolism and alleviate DN occurrence and progression of DN, and describes the prospects for future research.
Collapse
Affiliation(s)
- Ying Wang
- Country Renal Research Institution of Beijing University of Chinese Medicine, Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Tongtong Liu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuzi Cai
- Country Renal Research Institution of Beijing University of Chinese Medicine, Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Weijing Liu
- Country Renal Research Institution of Beijing University of Chinese Medicine, Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Jing Guo
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
47
|
Shi Y, Qi W. Histone Modifications in NAFLD: Mechanisms and Potential Therapy. Int J Mol Sci 2023; 24:14653. [PMID: 37834101 PMCID: PMC10572202 DOI: 10.3390/ijms241914653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/03/2023] [Accepted: 09/09/2023] [Indexed: 10/15/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a progressive condition that encompasses a spectrum of liver disorders, beginning with the simple steatosis, progressing to nonalcoholic steatohepatitis (NASH), and possibly leading to more severe diseases, including liver cirrhosis and hepatocellular carcinoma (HCC). In recent years, the prevalence of NAFLD has increased due to a shift towards energy-dense dietary patterns and a sedentary lifestyle. NAFLD is also strongly associated with metabolic disorders such as obesity and hyperlipidemia. The progression of NAFLD could be influenced by a variety of factors, such as diet, genetic factors, and even epigenetic factors. In contrast to genetic factors, epigenetic factors, including histone modifications, exhibit dynamic and reversible features. Therefore, the epigenetic regulation of the initiation and progression of NAFLD is one of the directions under intensive investigation in terms of pathogenic mechanisms and possible therapeutic interventions. This review aims to discuss the possible mechanisms and the crucial role of histone modifications in the framework of epigenetic regulation in NAFLD, which may provide potential therapeutic targets and a scientific basis for the treatment of NAFLD.
Collapse
Affiliation(s)
- Yulei Shi
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Wei Qi
- Gene Editing Center, School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
48
|
Tian Y, Feng T, Hou T, Zhu WG. Protocol to purify the histone deacetylase SIRT6 and assess its activity in vitro. STAR Protoc 2023; 4:102206. [PMID: 36995934 PMCID: PMC10090442 DOI: 10.1016/j.xpro.2023.102206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/16/2023] [Accepted: 03/08/2023] [Indexed: 03/30/2023] Open
Abstract
The histone deacetylase known as sirtuin 6 (SIRT6) deacetylates both histone and non-histone proteins but has low deacetylase activity in vitro. Here, we present a protocol to monitor SIRT6-mediated deacetylation of long-chain acyl-CoA synthase 5 in the presence of palmitic acid. We describe the purification of His-SIRT6 and a Flag-tagged substrate. We then detail a deacetylation assay protocol that can be widely applied to study other SIRT6-mediated deacetylation events and the effect of SIRT6 mutations on its activity. For complete details on the use and execution of this protocol, please refer to Hou et al. (2022).1.
Collapse
|
49
|
Li C, Wang F, Cui L, Li S, Zhao J, Liao L. Association between abnormal lipid metabolism and tumor. Front Endocrinol (Lausanne) 2023; 14:1134154. [PMID: 37305043 PMCID: PMC10248433 DOI: 10.3389/fendo.2023.1134154] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 05/05/2023] [Indexed: 06/13/2023] Open
Abstract
Metabolic Reprogramming is a sign of tumor, and as one of the three major substances metabolism, lipid has an obvious impact. Abnormal lipid metabolism is related to the occurrence of various diseases, and the proportion of people with abnormal lipid metabolism is increasing year by year. Lipid metabolism is involved in the occurrence, development, invasion, and metastasis of tumors by regulating various oncogenic signal pathways. The differences in lipid metabolism among different tumors are related to various factors such as tumor origin, regulation of lipid metabolism pathways, and diet. This article reviews the synthesis and regulatory pathways of lipids, as well as the research progress on cholesterol, triglycerides, sphingolipids, lipid related lipid rafts, adipocytes, lipid droplets, and lipid-lowering drugs in relation to tumors and their drug resistance. It also points out the limitations of current research and potential tumor treatment targets and drugs in the lipid metabolism pathway. Research and intervention on lipid metabolism abnormalities may provide new ideas for the treatment and survival prognosis of tumors.
Collapse
Affiliation(s)
- Chunyu Li
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong First Medical University, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Institute of Nephrology, Jinan, China
| | - Fei Wang
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong First Medical University, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Institute of Nephrology, Jinan, China
| | - Lili Cui
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong First Medical University, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Institute of Nephrology, Jinan, China
| | - Shaoxin Li
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong First Medical University, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Institute of Nephrology, Jinan, China
| | - Junyu Zhao
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong First Medical University, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Institute of Nephrology, Jinan, China
- Department of Endocrinology and Metabology, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lin Liao
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong First Medical University, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Institute of Nephrology, Jinan, China
- Department of Endocrinology and Metabology, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
50
|
Zhang L, Wang E, Peng G, Wang Y, Huang F. Comprehensive Proteome and Acetyl-Proteome Atlas Reveals Hepatic Lipid Metabolism in Layer Hens with Fatty Liver Hemorrhagic Syndrome. Int J Mol Sci 2023; 24:ijms24108491. [PMID: 37239836 DOI: 10.3390/ijms24108491] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
The feeding of high-energy and low-protein diets often induces fatty liver hemorrhagic syndrome (FLHS) in laying hens. However, the mechanism of hepatic fat accumulation in hens with FLHS remains uncertain. In this research, a comprehensive hepatic proteome and acetyl-proteome analysis was performed in both normal and FLHS-affected hens. The results indicated that the upregulated proteins were primarily associated with fat digestion and absorption, the biosynthesis of unsaturated fatty acids, and glycerophospholipid metabolism, while the downregulated proteins were mainly related to bile secretion and amino acid metabolism. Furthermore, the significant acetylated proteins were largely involved in ribosome and fatty acid degradation, and the PPAR signaling pathway, while the significant deacetylated proteins were related to valine, leucine, and isoleucine degradation in laying hens with FLHS. Overall, these results demonstrate that acetylation inhibits hepatic fatty acid oxidation and transport in hens with FLHS, and mainly exerts its effects by affecting protein activity rather than expression. This study provides new nutritional regulation options to alleviate FLHS in laying hens.
Collapse
Affiliation(s)
- Li Zhang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Enling Wang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Gang Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Yi Wang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Feiruo Huang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|