1
|
Tu S, Dong Y, Li C, Jiang M, Duan L, Zhang W, Chen X. Phosphatidylcholine Ameliorates Palmitic Acid-Induced Lipotoxicity by Facilitating Endoplasmic Reticulum and Mitochondria Contacts in Intervertebral Disc Degeneration. JOR Spine 2025; 8:e70062. [PMID: 40171442 PMCID: PMC11956213 DOI: 10.1002/jsp2.70062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 02/21/2025] [Accepted: 03/23/2025] [Indexed: 04/03/2025] Open
Abstract
Background Intervertebral disc degeneration (IDD) is a prevalent musculoskeletal disorder with substantial socioeconomic impacts. Despite its high prevalence, the pathogenesis of IDD remains unclear, and effective pharmacological interventions are lacking. This study aimed to investigate metabolic alterations in IDD and explore potential therapeutic targets by analyzing lipotoxicity-related mechanisms in nucleus pulposus (NP) cells. Methods Metabolomics and magnetic resonance spectroscopy were utilized to profile metabolic changes in NP tissues from advanced-stage IDD. Transcriptomics and metabolomics integration were performed to identify key regulatory pathways. In vitro experiments using human NP cells exposed to palmitic acid were conducted to evaluate endoplasmic reticulum (ER) stress, mitochondrial dysfunction, lipid droplet accumulation, and senescence. Phosphatidylcholine supplementation was tested for its ability to mitigate lipotoxicity, with ER-mitochondria interactions and mitochondrial oxidation capacity assessed as mechanistic endpoints. Results Our findings revealed an abnormal lipotoxic condition in NP cells from advanced-stage IDD. Furthermore, we identified abnormal accumulation of triglycerides and palmitic acid in NP cells from IDD. The palmitic acid accumulation resulted in endoplasmic reticulum stress, mitochondrial damage, lipid droplet accumulation, and senescence of NP cells. By integrating transcriptomics and metabolomics analyses, we discovered that phosphatidylcholine plays a role in regulating palmitic acid-induced lipotoxicity. Notably, phosphatidylcholine level was found to be low in the endoplasmic reticulum and mitochondria of advanced-stage NP cells. Phosphatidylcholine treatment alleviated palmitic acid-induced lipid droplet accumulation and senescence of NP cells by modulating ER-mitochondria contacts and mitochondrial oxidation capacity. Conclusion Phosphatidylcholine emerges as a potential therapeutic agent to counteract lipotoxic stress by modulating organelle interactions and mitochondrial function. These findings advance our understanding of IDD pathogenesis and provide a novel metabolic target for therapeutic development.
Collapse
Affiliation(s)
- Shuangshuang Tu
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- College of Pharmacy, Anhui Xinhua UniversityHefeiChina
| | - Yijun Dong
- Department of OrthopedicsThe First Affiliated Hospital of USTC, Provincial Hospital Affiliated to Anhui Medical UniversityHefeiChina
| | - Chuanfu Li
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Clinical College of Anhui Medical UniversityHefeiChina
| | - Mingxin Jiang
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- School of Clinical MedicineAnhui University of Science and TechnologyHuainanChina
| | - Liqun Duan
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Wenzhi Zhang
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Xi Chen
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| |
Collapse
|
2
|
Wang L, Hou P, Ma W, Jin R, Wei X, Li X, He H, Wang H. Unveiling EXOC4/SEC8: a key player in enhancing antiviral immunity by inhibiting the FBXL19-STING1-SQSTM1 signaling axis. Autophagy 2025. [PMID: 40413753 DOI: 10.1080/15548627.2025.2511077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 05/14/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025] Open
Abstract
As a core aptamer for anti-DNA viral immunity, STING1 (stimulator of interferon response cGAMP interactor 1) is tightly regulated to ensure the proper functioning of the natural antiviral immune response. However, many mechanisms underlying the regulation of STING1 remain largely unknown. In this study, we identify EXOC4/SEC8 (exocyst complex component 4) as a novel positive regulator of DNA virus-triggered type I interferon signaling responses through stabilizing STING1, thereby inhibiting DNA viral replication. Mechanistically, EXOC4 suppresses K27-linked ubiquitination of STING1 at K338, K347, and K370 catalyzed by the E3 ligase FBXL19 (F-box and leucine rich repeat protein 19), thereby preventing ubiquitinated-STING1 from recognition by SQSTM1 (sequestosome 1) for autophagic degradation. Importantly, mice conditionally knocked out for Exoc4/Sec8 are more susceptible to herpes simplex virus type 1 (HSV-1) infection and exhibit more severe lung pathology compared to control mice. This further confirms the important role of EXOC4/SEC8 in antiviral natural immunity. Taken together, our study reveals the importance of EXOC4/SEC8 in promoting STING1-centered antiviral natural immunity and highlights its potential as an anti-DNA viral therapeutic target.
Collapse
Affiliation(s)
- Lin Wang
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
- Key Laboratory of Animal Resistant Biology of Shandong, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Peili Hou
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
- Key Laboratory of Animal Resistant Biology of Shandong, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Wenqing Ma
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
- Key Laboratory of Animal Resistant Biology of Shandong, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Rong Jin
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Xinxin Wei
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Xingyu Li
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Hongbin He
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
- Key Laboratory of Animal Resistant Biology of Shandong, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| | - Hongmei Wang
- Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
- Key Laboratory of Animal Resistant Biology of Shandong, College of Life Sciences, Shandong Normal University, Jinan, Shandong, China
| |
Collapse
|
3
|
Song K, Heng L, Yan N. STING: a multifaced player in cellular homeostasis. Hum Mol Genet 2025:ddae175. [PMID: 40292755 DOI: 10.1093/hmg/ddae175] [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: 10/28/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 04/30/2025] Open
Abstract
The stimulator of interferon gene (STING) is an important innate immune mediator of the cytoplasmic DNA sensing pathway. As a mediator known for its role in the immune response to infections, STING is also surprisingly at the center of a variety of non-infectious human diseases, including cancer, autoimmune diseases and neurodegenerative diseases. Recent studies have shown that STING has many signaling activities, including type I interferon (IFN-I) and other IFN-independent activities, many of which are poorly understood. STING also has the unique property of being continuous transported from the ER to the Golgi then to the lysosome. Mutations of STING or trafficking cofactors are associated with human diseases affecting multiple immune and non-immune organs. Here, we review recent advances in STING trafficking and signaling mechanisms based in part on studies of STING-associated monogenic inborn error diseases.
Collapse
Affiliation(s)
- Kun Song
- Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390, United States
| | - Lyu Heng
- Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390, United States
| | - Nan Yan
- Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390, United States
| |
Collapse
|
4
|
Tan Y, Miao L, Wang C, Wang H, Li Y, Huang Y, Teng H, Tian Y, Yang G, Zeng X, Li J. The Role and Mechanism of TRIM13 Regulation of TRAF6 Ubiquitination in the Synergy of Inflammatory Responses and Neurotoxicity Induced by METH and HIV- 1 Tat Protein in Astrocytes. Neurotox Res 2025; 43:21. [PMID: 40192895 DOI: 10.1007/s12640-025-00743-5] [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: 07/07/2024] [Revised: 02/13/2025] [Accepted: 03/28/2025] [Indexed: 05/03/2025]
Abstract
Methamphetamine (METH) abuse and HIV infection are major public health concerns worldwide. While both METH and HIV- 1 Tat proteins can induce neurotoxicity and synergistic effects on the nervous system, the mechanisms by which they act synergistically remain unclear. Our recent research shows that neuroinflammation plays an important role in neurotoxicity induced by METH and HIV- 1 Tat proteins, but the regulatory mechanism has not been clarified. Tripartite Motif Containing 13 (TRIM13) is a protein known to regulate the inflammatory response through ubiquitination of Tumor Necrosis Factor Receptor Associated Factor 6 (TRAF6). This study investigated the role of TRIM13 and TRAF6 in the inflammatory response of U- 87 MG cells induced by METH and HIV- 1 Tat proteins. U- 87 MG cells were treated with 2 mM METH and/or 100 nM HIV- 1 Tat protein. Western blot (WB), immunofluorescence (IF), and co-immunoprecipitation (Co-IP) experiments were employed to elucidate the role of TRIM13 and TRAF6. The results demonstrated that METH and HIV- 1 Tat protein could synergistically induce an inflammatory response in U- 87 MG cells. Furthermore, the knockdown of TRIM13 significantly enhanced this inflammatory response, while the inhibition of TRAF6 significantly weakened it. Additionally, the study revealed that TRIM13 could degrade TRAF6 via ubiquitination. In conclusion, this study suggests that TRIM13 regulates TRAF6 ubiquitination to dampen the inflammatory response of U- 87 MG cells induced by METH and HIV- 1 Tat proteins. These findings highlight TRIM13 and TRAF6 as potential targets for therapeutic intervention in the context of METH and HIV- 1 Tat protein-induced inflammatory responses and neurotoxic effects.
Collapse
Affiliation(s)
- Yi Tan
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong District, Kunming, Yunnan, 650500, People's Republic of China
| | - Lin Miao
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong District, Kunming, Yunnan, 650500, People's Republic of China
| | - Chan Wang
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong District, Kunming, Yunnan, 650500, People's Republic of China
| | - Haowei Wang
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong District, Kunming, Yunnan, 650500, People's Republic of China
| | - Yi Li
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong District, Kunming, Yunnan, 650500, People's Republic of China
| | - Yizhen Huang
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong District, Kunming, Yunnan, 650500, People's Republic of China
| | - Hanxin Teng
- Department of Pathogen Biology and Immunology, School of Basic Medical Science, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong District, Kunming, Yunnan, People's Republic of China
| | - Yunqing Tian
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong District, Kunming, Yunnan, 650500, People's Republic of China
| | - Genmeng Yang
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong District, Kunming, Yunnan, 650500, People's Republic of China.
| | - Xiaofeng Zeng
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong District, Kunming, Yunnan, 650500, People's Republic of China.
| | - Juan Li
- Department of Pathogen Biology and Immunology, School of Basic Medical Science, Kunming Medical University, 1168 West Chunrong Road, Yuhua Avenue Chenggong District, Kunming, Yunnan, People's Republic of China.
| |
Collapse
|
5
|
Zhang J, Wu Y, Wang Y, Wang J, Ye Y, Yin H, Sun N, Qin B, Sun N. TRIM35 Negatively Regulates the cGAS-STING-Mediated Signaling Pathway by Attenuating K63-Linked Ubiquitination of STING. Inflammation 2025; 48:855-869. [PMID: 39088122 DOI: 10.1007/s10753-024-02093-4] [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: 04/14/2024] [Revised: 06/21/2024] [Accepted: 06/27/2024] [Indexed: 08/02/2024]
Abstract
The cGAS-STING-mediated antiviral response plays an important role in the defense against DNA virus infection. Tripartite motif protein 35 (TRIM35), an E3 ubiquitin ligase, was identified as a positive regulator of RLR-mediated antiviral signaling in our previous study, but the effect of TRIM35 on the cGAS-STING signaling pathway has not been elucidated. Herein, we showed that TRIM35 negatively regulates the cGAS-STING signaling pathway by directly targeting STING. TRIM35 overexpression significantly inhibited the cGAMP-triggered phosphorylation of TBK1 and IRF3, attenuating IFN-β expression and the downstream antiviral response. Mechanistically, TRIM35 colocalized and directly interacted with STING in the cytoplasm. TRM35 removed K63-linked ubiquitin from STING through the C36 and C44 sites in the RING domain, which impaired the interaction of STING with TBK1 or IKKε. In addition, we demonstrated that the RING domain is a key region for the antiviral effects of TIRM35. These results collectively indicate that TRIM35 negatively regulates type I interferon (IFN-I) production by targeting and deubiquitinating STING. TRIM35 may be a potential therapeutic target for controlling viral infection.
Collapse
Affiliation(s)
- Jikai Zhang
- Xuzhou Medical University, Xuzhou, China
- Department of Pathogen Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, School of Basic Medical Sciences, Xuzhou Medical University, Xuzhou, China
| | - Yuhao Wu
- Xuzhou Medical University, Xuzhou, China
| | - Yiwen Wang
- Xuzhou Medical University, Xuzhou, China
| | - Jing Wang
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yinlin Ye
- Xuzhou Medical University, Xuzhou, China
| | - Hang Yin
- Xuzhou Medical University, Xuzhou, China
| | - Ningye Sun
- Xuzhou Medical University, Xuzhou, China
| | | | - Nan Sun
- Xuzhou Medical University, Xuzhou, China.
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China.
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
6
|
Zhou P, Zhang Q, Yang Y, Chen D, Jongkaewwattana A, Jin H, Zhou H, Luo R. Avian TRIM13 attenuates antiviral innate immunity by targeting MAVS for autophagic degradation. Autophagy 2025; 21:754-770. [PMID: 39508267 DOI: 10.1080/15548627.2024.2426114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/29/2024] [Accepted: 11/02/2024] [Indexed: 11/15/2024] Open
Abstract
MAVS (mitochondrial antiviral signaling protein) is a crucial adaptor in antiviral innate immunity that must be tightly regulated to maintain immune homeostasis. In this study, we identified the duck Anas platyrhynchos domesticus TRIM13 (ApdTRIM13) as a novel negative regulator of duck MAVS (ApdMAVS) that mediates the antiviral innate immune response. Upon infection with RNA viruses, ApdTRIM13 expression increased, and it specifically binds to ApdMAVS through its TM domain, facilitating the degradation of ApdMAVS in a manner independent of E3 ligase activity. Furthermore, ApdTRIM13 recruits the autophagic cargo receptor duck SQSTM1 (ApdSQSTM1), which facilitates its interaction with ApdMAVS independent of ubiquitin signaling, and subsequently delivers ApdMAVS to phagophores for degradation. Depletion of ApdSQSTM1 reduces ApdTRIM13-mediated autophagic degradation of ApdMAVS, thereby enhancing the antiviral immune response. Collectively, our findings reveal a novel mechanism by which ApdTRIM13 regulates type I interferon production by targeting ApdMAVS for selective autophagic degradation mediated by ApdSQSTM1, providing insights into the crosstalk between selective autophagy and innate immune responses in avian species.Abbreviation: 3-MA: 3-methyladenine; ATG5: autophagy related 5; baf A1: bafilomycin A1; BECN1: beclin 1; CALCOCO2/NDP52: calcium binding and coiled-coil domain 2; CARD: caspase recruitment domain; co-IP: co-immunoprecipitation; DEFs: duck embryonic fibroblasts; DTMUV: duck Tembusu virus; eGFP: enhanced green fluorescent protein; hpi: hours post infection; IFIH1/MDA5: interferon induced with helicase C domain 1; IFN: interferon; IKBKE/IKKε: inhibitor of nuclear factor kappa B kinase subunit epsilon; IP: immunoprecipitation; IRF7: interferon regulatory factor 7; ISRE: interferon-stimulated response element; mAb: monoclonal antibody; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MAVS: mitochondrial antiviral signaling protein; MOI: multiplicity of infection; NBR1: NBR1 autophagy cargo receptor; NFKB: nuclear factor kappa B; pAb: polyclonal antibody; poly(I:C): Polyriboinosinic polyribocytidylic acid; RIGI: RNA sensor RIG-I; RLR: RIGI-like-receptor; SeV: sendai virus; siRNA: small interfering RNA; SQSTM1/p62: sequestosome 1; TAX1BP1: Tax1 binding protein 1; TBK1: TANK binding kinase 1; TCID50: 50% tissue culture infectious dose; TM: tansmembrane; TOLLIP: toll interacting protein; TRIM: tripartite motif containing; UBA: ubiquitin-associated domain; Ub: ubiquitin; VSV: vesicular stomatitis virus; WT: wild type.
Collapse
Affiliation(s)
- Peng Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Wuhan, China
| | - Qingxiang Zhang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Wuhan, China
| | - Yueshan Yang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Wuhan, China
| | - Dong Chen
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Wuhan, China
| | - Anan Jongkaewwattana
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | - Hui Jin
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Wuhan, China
| | - Hongbo Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Wuhan, China
| | - Rui Luo
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China
- Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Wuhan, China
| |
Collapse
|
7
|
Deng C, Chen D, Yang L, Zhang Y, Jin C, Li Y, Lin Q, Luo M, Zheng R, Huang B, Liu S. The role of cGAS-STING pathway ubiquitination in innate immunity and multiple diseases. Front Immunol 2025; 16:1522200. [PMID: 40028324 PMCID: PMC11868049 DOI: 10.3389/fimmu.2025.1522200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/20/2025] [Indexed: 03/05/2025] Open
Abstract
The cGAS-STING pathway is essential in innate immunity, especially in antiviral responses and cellular stress management. cGAS acts as a cytoplasmic DNA sensor by initiating the synthesis of the second messenger cyclic GMP-AMP synthase (cGAMP), which subsequently activates the STING pathway, leading to the production of type I interferons and other cytokines, as well as the activation of inflammatory mediators. Recent studies have demonstrated that ubiquitination changes closely regulate the function of the cGAS-STING pathway. Ubiquitination modifications influence the stability and activity of cGAS and STING, while also influencing the accuracy of the immune response by adjusting their degradation and signal intensity. E3 ubiquitin ligase specifically facilitates the degradation or modulates the signaling of cGAS-STING-associated proteins via ubiquitination alterations. Furthermore, the ubiquitination of the cGAS-STING pathway serves distinct functions in various cell types and engages with NF-κB, IRF3/7, autophagy, and endoplasmic reticulum stress. This ubiquitin-mediated regulation is crucial for sustaining the balance of innate immunity, while excessive or inadequate ubiquitination can result in autoimmune disorders, cancers, and viral infections. An extensive examination of the ubiquitination process within the cGAS-STING pathway elucidates its specific regulatory mechanisms in innate immunity and identifies novel targets for the intervention of associated diseases.
Collapse
Affiliation(s)
- Chunyan Deng
- Department of Hematology and Oncology, Shenzhen Children ‘s Hospital, Shenzhen, China
| | - Dongyan Chen
- Department of Hematology and Oncology, Shenzhen Children ‘s Hospital, Shenzhen, China
| | - Liang Yang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yubiao Zhang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Cheng Jin
- Department of Hematology and Oncology, Shenzhen Children ‘s Hospital, Shenzhen, China
| | - Yue Li
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Qihong Lin
- Department of Hematology and Oncology, Shenzhen Children ‘s Hospital, Shenzhen, China
| | - Mingjing Luo
- Department of Hematology and Oncology, Shenzhen Children ‘s Hospital, Shenzhen, China
| | - Ruihao Zheng
- Department of Hematology and Oncology, Shenzhen Children ‘s Hospital, Shenzhen, China
| | - Baozhen Huang
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Sixi Liu
- Department of Hematology and Oncology, Shenzhen Children ‘s Hospital, Shenzhen, China
| |
Collapse
|
8
|
You J, Xu A, Wang Y, Tu G, Huang R, Wu S. The STING signaling pathways and bacterial infection. Apoptosis 2025; 30:389-400. [PMID: 39428409 DOI: 10.1007/s10495-024-02031-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2024] [Indexed: 10/22/2024]
Abstract
As antibiotic-resistant bacteria continue to emerge frequently, bacterial infections have become a significant and pressing challenge to global public health. Innate immunity triggers the activation of host responses by sensing "non-self" components through various pattern recognition receptors (PRRs), serving as the first line of antibacterial defense. Stimulator of interferon genes (STING) is a PRR that binds with cyclic dinucleotides (CDN) to exert effects against bacteria, viruses, and cancer by inducing the production of type I interferon and inflammatory cytokines, and facilitating regulated cell death. Currently, drugs targeting the STING signaling pathway are predominantly applied in the fields of modulating host immune defense against cancer and viral infections, with relatively limited application in treating bacterial infections. Given the significant immunomodulatory functions of STING in the interaction between bacteria and hosts, this review summarizes the research progress on STING signaling pathways and their roles in bacterial infection, as well as the novel functions of STING modulators, aiming to offer insights for the development of antibacterial drugs.
Collapse
Affiliation(s)
- Jiayi You
- Department of Medical Microbiology, School of Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, 215123, China
| | - Ailing Xu
- Department of Medical Microbiology, School of Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, 215123, China
| | - Ye Wang
- Department of Medical Microbiology, School of Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, 215123, China
| | - Guangmin Tu
- Department of Medical Microbiology, School of Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, 215123, China
| | - Rui Huang
- Department of Medical Microbiology, School of Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, 215123, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-Infective Medicine, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, 215123, China
| | - Shuyan Wu
- Department of Medical Microbiology, School of Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, 215123, China.
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Key Laboratory of Pathogen Bioscience and Anti-Infective Medicine, Suzhou Medical College of Soochow University, Suzhou, Jiangsu Province, 215123, China.
| |
Collapse
|
9
|
Wang M, Qin Y, Ai X, Liu X. RBM15-dependent m6A modification mediates progression of non-small cell lung cancer cells. Mol Med 2024; 30:267. [PMID: 39716068 DOI: 10.1186/s10020-024-01018-z] [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: 05/10/2024] [Accepted: 11/28/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is the predominant form of lung cancer, contributing significantly to global health and economic challenges. This study elucidated the role of RBM15 in NSCLC progression through its involvement in m6A modifications. METHODS RBM15 levels in NSCLC tissues and cells were assessed via RT-qPCR and Western blotting. The impact of RBM15 knockdown on NSCLC proliferation, invasion, and migration was evaluated using CCK-8, colony formation, and Transwell assays. Expression levels of KLF1, TRIM13, and ANXA8 were determined by RT-qPCR and Western blot. m6A methylation levels were analyzed, while RIP and MeRIP assays were employed to explore the interaction between YTHDF1/YTHDF2/m6A and KLF1/TRIM13, as well as KLF1 binding to the ANXA8 promoter. The ubiquitination of ANXA8 was examined through ubiquitination assays. Xenograft and metastasis models were utilized to assess RBM15's role in vivo. RESULTS RBM15 was found to be overexpressed in NSCLC. Silencing RBM15 led to decreased cell proliferation, invasion, and migration of NSCLC cells. RBM15 upregulated KLF1 and downregulated TRIM13 via YTHDF1/YTHDF2, resulting in the promotion of ANXA8 expression. KLF1 overexpression or TRIM13 downregulation partially reversed the suppressive effects of RBM15 knockdown on NSCLC cell proliferation. ANXA8, upregulated in NSCLC, mitigated the inhibitory effects of RBM15 silencing on malignant behaviors. In vivo, RBM15 downregulation hindered NSCLC cell proliferation and metastasis by modulating the KLF1-TRIM13/ANXA8 axis. CONCLUSION RBM15-mediated m6A methylation enhances KLF1 expression and suppresses TRIM13 via YTHDF1/YTHDF2, thereby promoting ANXA8 and facilitating NSCLC progression. These findings provide novel insights and potential therapeutic targets for NSCLC treatment.
Collapse
Affiliation(s)
- Man Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Jilin University, 1 Xinmin Street, Changchun, 130021, Jilin, China
| | - Yujiao Qin
- Department of Respiratory Medicine, The First Affiliated Hospital of Jilin University, 1 Xinmin Street, Changchun, 130021, Jilin, China
| | - Xiaoqi Ai
- Department of Respiratory Medicine, The First Affiliated Hospital of Jilin University, 1 Xinmin Street, Changchun, 130021, Jilin, China
| | - Xiuhua Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Jilin University, 1 Xinmin Street, Changchun, 130021, Jilin, China.
| |
Collapse
|
10
|
Weng C, Jin R, Jin X, Yang Z, He C, Zhang Q, Xu J, Lv B. Exploring the Mechanisms, Biomarkers, and Therapeutic Targets of TRIM Family in Gastrointestinal Cancer. Drug Des Devel Ther 2024; 18:5615-5639. [PMID: 39654601 PMCID: PMC11626976 DOI: 10.2147/dddt.s482340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 11/23/2024] [Indexed: 12/12/2024] Open
Abstract
Gastrointestinal region (GI) cancers are closely linked to the ubiquitination system, with the E3 ubiquitin ligase playing a crucial role by targeting various substrates. As E3 ubiquitin ligases, proteins of tripartite motif (TRIM) family play a role in cancer signaling, development, apoptosis, and formation. These proteins regulate diverse biological activities and signaling pathways. This study comprehensively outlines the functions of TRIM proteins in gastrointestinal physiology, contributing to our knowledge of the molecular pathways involved in gastrointestinal tumors. Gastrointestinal region (GI) cancers are closely linked to the ubiquitination system, with the E3 ubiquitin ligase playing a crucial role by targeting various substrates.
Collapse
Affiliation(s)
- Chunyan Weng
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, People’s Republic of China
| | - Rijuan Jin
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, People’s Republic of China
| | - Xiaoliang Jin
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, People’s Republic of China
| | - Zimei Yang
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, People’s Republic of China
| | - Chenghai He
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, People’s Republic of China
- Department of Gastroenterology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Qiuhua Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, People’s Republic of China
| | - Jingli Xu
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, People’s Republic of China
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Bin Lv
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, People’s Republic of China
| |
Collapse
|
11
|
Xiang Y, Li C, Wang Z, Feng J, Zhang J, Yang Y, Zhou J, Zhang J. TRIM13 Reduces Damage to Alveolar Epithelial Cells in COPD by Inhibiting Endoplasmic Reticulum Stress-Induced ER-Phagy. Lung 2024; 202:821-830. [PMID: 39382594 PMCID: PMC11541378 DOI: 10.1007/s00408-024-00753-8] [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: 07/19/2024] [Accepted: 09/20/2024] [Indexed: 10/10/2024]
Abstract
PURPOSE Tripartite motif-containing protein 13 (TRIM13) directly or indirectly participates in autophagy and apoptosis. However, it remains unclear whether TRIM13 participates in chronic obstructive pulmonary disease (COPD) progression. This study aimed to reveal the molecular mechanisms through which TRIM13 regulates alveolar epithelial cell injury in COPD to provide new molecular targets for COPD treatment. METHODS The TRIM13 expression levels were determined in clinical COPD patients and a rat emphysema model. A cigarette smoke-induced model of endoplasmic reticulum stress (ERS) and endoplasmic reticulum autophagy (ER-phagy) was developed using A549 cells, and the effects of TRIM13 gene overexpression/knockdown on ERS, ER-phagy, and cell apoptosis were assessed in these cells. RESULTS TRIM13 expression was significantly decreased in the lung tissues of COPD patients and rats with emphysema. Moreover, the apoptosis level was significantly increased in the lung tissues of rats with emphysema. TRIM13 gene overexpression reduced the expression levels of ERS-related molecules (GRP78, GRP94, XBP-1, and eIF2a) in the COPD model; it also lowered the ER-phagy level, as evidenced by decreased number of autolysosomes observed by transmission electron microscopy, improved endoplasmic reticulum structure, reduced LC3-II/LC3-I and Beclin1 expression levels, and increased expression level of the autophagy inhibitory molecule Bcl-2. TRIM13 gene knockdown, however, led to opposite results. CONCLUSION TRIM13 expression attenuated alveolar epithelial cell injury in COPD by inhibiting ERS-induced ER-phagy.
Collapse
Affiliation(s)
- Yaling Xiang
- Department of Respiratory Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Chuntao Li
- Department of Respiratory Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Zhiyuan Wang
- Department of Respiratory Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Jiagang Feng
- Department of Respiratory Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Jiaqiang Zhang
- Department of Respiratory Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Yue Yang
- Department of Respiratory Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Jinbiao Zhou
- Department of Respiratory Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Jianqing Zhang
- Department of Respiratory Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China.
| |
Collapse
|
12
|
Sayyad Z, Acharya D, Gack MU. TRIM Proteins: Key Regulators of Immunity to Herpesvirus Infection. Viruses 2024; 16:1738. [PMID: 39599852 PMCID: PMC11599090 DOI: 10.3390/v16111738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024] Open
Abstract
Herpesviruses are ubiquitous DNA viruses that can establish latency and cause a range of mild to life-threatening diseases in humans. Upon infection, herpesviruses trigger the activation of several host antiviral defense programs that play critical roles in curbing virus replication and dissemination. Recent work from many groups has integrated our understanding of TRIM (tripartite motif) proteins, a specific group of E3 ligase enzymes, as pivotal orchestrators of mammalian antiviral immunity. In this review, we summarize recent advances in the modulation of innate immune signaling by TRIM proteins during herpesvirus infection, with a focus on the detection of herpes simplex virus type 1 (HSV-1, a prototype herpesvirus) by cGAS-STING, RIG-I-like receptors, and Toll-like receptors. We also review the latest progress in understanding the intricate relationship between herpesvirus replication and TRIM protein-regulated autophagy and apoptosis. Finally, we discuss the maneuvers used by HSV-1 and other herpesviruses to overcome TRIM protein-mediated virus restriction.
Collapse
Affiliation(s)
| | - Dhiraj Acharya
- Florida Research and Innovation Center, Cleveland Clinic, 9801 SW Discovery Way, Port St. Lucie, FL 34987, USA;
| | - Michaela U. Gack
- Florida Research and Innovation Center, Cleveland Clinic, 9801 SW Discovery Way, Port St. Lucie, FL 34987, USA;
| |
Collapse
|
13
|
Li W, Lin Y, Wang X, Yang H, Ding Y, Chen Z, He Z, Zhang J, Zhao L, Jiao P. Chicken UFL1 Restricts Avian Influenza Virus Replication by Disrupting the Viral Polymerase Complex and Facilitating Type I IFN Production. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1479-1492. [PMID: 38477617 DOI: 10.4049/jimmunol.2300613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 02/12/2024] [Indexed: 03/14/2024]
Abstract
During avian influenza virus (AIV) infection, host defensive proteins promote antiviral innate immunity or antagonize viral components to limit viral replication. UFM1-specific ligase 1 (UFL1) is involved in regulating innate immunity and DNA virus replication in mammals, but the molecular mechanism by which chicken (ch)UFL1 regulates AIV replication is unclear. In this study, we first identified chUFL1 as a negative regulator of AIV replication by enhancing innate immunity and disrupting the assembly of the viral polymerase complex. Mechanistically, chUFL1 interacted with chicken stimulator of IFN genes (chSTING) and contributed to chSTING dimerization and the formation of the STING-TBK1-IRF7 complex. We further demonstrated that chUFL1 promoted K63-linked polyubiquitination of chSTING at K308 to facilitate chSTING-mediated type I IFN production independent of UFMylation. Additionally, chUFL1 expression was upregulated in response to AIV infection. Importantly, chUFL1 also interacted with the AIV PA protein to inhibit viral polymerase activity. Furthermore, chUFL1 impeded the nuclear import of the AIV PA protein and the assembly of the viral polymerase complex to suppress AIV replication. Collectively, these findings demonstrate that chUFL1 restricts AIV replication by disrupting the viral polymerase complex and facilitating type I IFN production, which provides new insights into the regulation of AIV replication in chickens.
Collapse
Affiliation(s)
- Weiqiang Li
- College of Veterinary Medicine, Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China; and
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, Guangzhou, China
| | - Yu Lin
- College of Veterinary Medicine, Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China; and
| | - Xiyi Wang
- College of Veterinary Medicine, Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China; and
| | - Huixing Yang
- College of Veterinary Medicine, Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China; and
| | - Yangbao Ding
- College of Veterinary Medicine, Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China; and
| | - Zuxian Chen
- College of Veterinary Medicine, Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China; and
| | - Zhuoliang He
- College of Veterinary Medicine, Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China; and
| | - Junsheng Zhang
- College of Veterinary Medicine, Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China; and
| | - Luxiang Zhao
- College of Veterinary Medicine, Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China; and
| | - Peirong Jiao
- College of Veterinary Medicine, Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, China; and
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, Guangzhou, China
| |
Collapse
|
14
|
Yang B, Pei J, Lu C, Wang Y, Shen M, Qin X, Huang Y, Yang X, Zhao X, Ma S, Song Z, Liang Y, Wang H, Wang J. RNF144A promotes antiviral responses by modulating STING ubiquitination. EMBO Rep 2023; 24:e57528. [PMID: 37955227 PMCID: PMC10702816 DOI: 10.15252/embr.202357528] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 10/18/2023] [Accepted: 10/23/2023] [Indexed: 11/14/2023] Open
Abstract
Stimulator of interferon (IFN) genes (STING, also named MITA, ERIS, MPYS, or TMEM173) plays an essential role in DNA virus- or cytosolic DNA-triggered innate immune responses. Here, we demonstrate that the RING-in-between RING (RBR) E3 ubiquitin ligase family member RING-finger protein (RNF) 144A interacts with STING and promotes its K6-linked ubiquitination at K236, thereby enhancing STING translocation from the ER to the Golgi and downstream signaling pathways. The K236R mutant of STING displays reduced activity in promoting innate immune signal transduction. Overexpression of RNF144A upregulates HSV-1- or cytosolic DNA-induced immune responses, while knockdown of RNF144A expression has the opposite effect. In addition, Rnf144a-deficient cells exhibit impaired DNA virus- or cytosolic DNA-triggered signaling, and RNF144A protects mice from DNA virus infection. In contrast, RNF144A does not affect RNA virus- or cytosolic RNA-triggered innate immune responses. Taken together, our findings identify a new positive regulator of DNA virus- or cytosolic DNA-triggered signaling pathways and a critical ubiquitination site important for fully functional STING during antiviral responses.
Collapse
Affiliation(s)
- Bo Yang
- Xinxiang Key Laboratory of Inflammation and ImmunologyXinxiang Medical UniversityXinxiangChina
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory MedicineXinxiang Medical UniversityXinxiangChina
- Henan Key Laboratory of Immunology and Targeted DrugXinxiang Medical UniversityXinxiangChina
| | - Jinyong Pei
- Xinxiang Key Laboratory of Inflammation and ImmunologyXinxiang Medical UniversityXinxiangChina
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory MedicineXinxiang Medical UniversityXinxiangChina
- Henan Key Laboratory of Immunology and Targeted DrugXinxiang Medical UniversityXinxiangChina
| | - Chen Lu
- Xinxiang Key Laboratory of Inflammation and ImmunologyXinxiang Medical UniversityXinxiangChina
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory MedicineXinxiang Medical UniversityXinxiangChina
- Henan Key Laboratory of Immunology and Targeted DrugXinxiang Medical UniversityXinxiangChina
| | - Yi Wang
- Xinxiang Key Laboratory of Inflammation and ImmunologyXinxiang Medical UniversityXinxiangChina
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory MedicineXinxiang Medical UniversityXinxiangChina
- Henan Key Laboratory of Immunology and Targeted DrugXinxiang Medical UniversityXinxiangChina
| | - Mengyang Shen
- Xinxiang Key Laboratory of Inflammation and ImmunologyXinxiang Medical UniversityXinxiangChina
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory MedicineXinxiang Medical UniversityXinxiangChina
- Henan Key Laboratory of Immunology and Targeted DrugXinxiang Medical UniversityXinxiangChina
| | - Xiao Qin
- Xinxiang Key Laboratory of Inflammation and ImmunologyXinxiang Medical UniversityXinxiangChina
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory MedicineXinxiang Medical UniversityXinxiangChina
- Henan Key Laboratory of Immunology and Targeted DrugXinxiang Medical UniversityXinxiangChina
| | - Yulu Huang
- Xinxiang Key Laboratory of Inflammation and ImmunologyXinxiang Medical UniversityXinxiangChina
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory MedicineXinxiang Medical UniversityXinxiangChina
- Henan Key Laboratory of Immunology and Targeted DrugXinxiang Medical UniversityXinxiangChina
| | - Xi Yang
- Xinxiang Key Laboratory of Inflammation and ImmunologyXinxiang Medical UniversityXinxiangChina
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory MedicineXinxiang Medical UniversityXinxiangChina
- Henan Key Laboratory of Immunology and Targeted DrugXinxiang Medical UniversityXinxiangChina
| | - Xin Zhao
- Department of Laboratory MedicineThe Third Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
| | - Shujun Ma
- Xinxiang Key Laboratory of Inflammation and ImmunologyXinxiang Medical UniversityXinxiangChina
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory MedicineXinxiang Medical UniversityXinxiangChina
- Henan Key Laboratory of Immunology and Targeted DrugXinxiang Medical UniversityXinxiangChina
| | - Zhishan Song
- Xinxiang Key Laboratory of Inflammation and ImmunologyXinxiang Medical UniversityXinxiangChina
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory MedicineXinxiang Medical UniversityXinxiangChina
- Henan Key Laboratory of Immunology and Targeted DrugXinxiang Medical UniversityXinxiangChina
| | - Yinming Liang
- Henan Key Laboratory of Immunology and Targeted DrugXinxiang Medical UniversityXinxiangChina
- Ping Yuan LaboratoryXinxiangChina
| | - Hui Wang
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory MedicineXinxiang Medical UniversityXinxiangChina
- Henan Key Laboratory of Immunology and Targeted DrugXinxiang Medical UniversityXinxiangChina
| | - Jie Wang
- Xinxiang Key Laboratory of Inflammation and ImmunologyXinxiang Medical UniversityXinxiangChina
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory MedicineXinxiang Medical UniversityXinxiangChina
- Henan Key Laboratory of Immunology and Targeted DrugXinxiang Medical UniversityXinxiangChina
| |
Collapse
|
15
|
Dudley-Fraser J, Rittinger K. It's a TRIM-endous view from the top: the varied roles of TRIpartite Motif proteins in brain development and disease. Front Mol Neurosci 2023; 16:1287257. [PMID: 38115822 PMCID: PMC10728303 DOI: 10.3389/fnmol.2023.1287257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/13/2023] [Indexed: 12/21/2023] Open
Abstract
The tripartite motif (TRIM) protein family members have been implicated in a multitude of physiologies and pathologies in different tissues. With diverse functions in cellular processes including regulation of signaling pathways, protein degradation, and transcriptional control, the impact of TRIM dysregulation can be multifaceted and complex. Here, we focus on the cellular and molecular roles of TRIMs identified in the brain in the context of a selection of pathologies including cancer and neurodegeneration. By examining each disease in parallel with described roles in brain development, we aim to highlight fundamental common mechanisms employed by TRIM proteins and identify opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Jane Dudley-Fraser
- Molecular Structure of Cell Signalling Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Katrin Rittinger
- Molecular Structure of Cell Signalling Laboratory, The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
16
|
Christianson JC, Jarosch E, Sommer T. Mechanisms of substrate processing during ER-associated protein degradation. Nat Rev Mol Cell Biol 2023; 24:777-796. [PMID: 37528230 DOI: 10.1038/s41580-023-00633-8] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2023] [Indexed: 08/03/2023]
Abstract
Maintaining proteome integrity is essential for long-term viability of all organisms and is overseen by intrinsic quality control mechanisms. The secretory pathway of eukaryotes poses a challenge for such quality assurance as proteins destined for secretion enter the endoplasmic reticulum (ER) and become spatially segregated from the cytosolic machinery responsible for disposal of aberrant (misfolded or otherwise damaged) or superfluous polypeptides. The elegant solution provided by evolution is ER-membrane-bound ubiquitylation machinery that recognizes misfolded or surplus proteins or by-products of protein biosynthesis in the ER and delivers them to 26S proteasomes for degradation. ER-associated protein degradation (ERAD) collectively describes this specialized arm of protein quality control via the ubiquitin-proteasome system. But, instead of providing a single strategy to remove defective or unwanted proteins, ERAD represents a collection of independent processes that exhibit distinct yet overlapping selectivity for a wide range of substrates. Not surprisingly, ER-membrane-embedded ubiquitin ligases (ER-E3s) act as central hubs for each of these separate ERAD disposal routes. In these processes, ER-E3s cooperate with a plethora of specialized factors, coordinating recognition, transport and ubiquitylation of undesirable secretory, membrane and cytoplasmic proteins. In this Review, we focus on substrate processing during ERAD, highlighting common threads as well as differences between the many routes via ERAD.
Collapse
Affiliation(s)
- John C Christianson
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK.
| | - Ernst Jarosch
- Max-Delbrück-Centrer for Molecular Medicine in Helmholtz Association, Berlin-Buch, Germany
| | - Thomas Sommer
- Max-Delbrück-Centrer for Molecular Medicine in Helmholtz Association, Berlin-Buch, Germany.
- Institute for Biology, Humboldt Universität zu Berlin, Berlin, Germany.
| |
Collapse
|
17
|
Lai M, Liu M, Li D, Zhang R, Wang L, Li X, Chen S, Wu S, Xiao S, Wei L, Wang X, Nan A, Zeng X. circELMOD3 increases and stabilizes TRIM13 by sponging miR-6864-5p and direct binding to inhibit HCC progression. iScience 2023; 26:107818. [PMID: 37752955 PMCID: PMC10518715 DOI: 10.1016/j.isci.2023.107818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/17/2023] [Accepted: 08/31/2023] [Indexed: 09/28/2023] Open
Abstract
Many circular RNAs (circRNAs) have been identified to be associated with hepatocellular carcinoma (HCC) progression. We aim to explore the diagnostic potential, functions, and mechanism of circELMOD3 in HCC. Differentially expressed circRNAs in HCC and its paired adjacent tissues were identified by RNA sequencing. circELMOD3 was downregulated in HCC tissues and was related to clinicopathological characteristics of HCC patients. Additionally, plasma circELMOD3 was shown to be a highly sensitive and non-invasive biomarker to distinguish HCC from healthy controls. Functional assays showed that circELMOD3 inhibited proliferation and induced apoptosis of HCC cells both in vitro and in vivo. Mechanistically, RNA antisense purification (RAP) and luciferase reporter assays verified that circELMOD3 functioned as a sponge for miR-6864-5p leading to increased expression of its target gene TRIM13. Interestingly, RNA stability test demonstrated that circELMOD3 overexpression led to enhanced stability of its directly bound TRIM13 mRNA, which in turn co-activated the p53 signaling pathway.
Collapse
Affiliation(s)
- Mingshuang Lai
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Meiliang Liu
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Deyuan Li
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Ruirui Zhang
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Lijun Wang
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Xiaofei Li
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Sixian Chen
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Siqian Wu
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Suyang Xiao
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Liling Wei
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Xiaogang Wang
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Aruo Nan
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Xiaoyun Zeng
- School of Public Health, Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi, China
- Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning 530021, Guangxi, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, Guangxi, China
| |
Collapse
|
18
|
Dean ST, Ishikawa C, Zhu X, Walulik S, Nixon T, Jordan JK, Henderson S, Wyder M, Salomonis N, Wunderlich M, Greis KD, Starczynowski DT, Volk AG. Repression of TRIM13 by chromatin assembly factor CHAF1B is critical for AML development. Blood Adv 2023; 7:4822-4837. [PMID: 37205848 PMCID: PMC10469560 DOI: 10.1182/bloodadvances.2022009438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/22/2023] [Accepted: 04/18/2023] [Indexed: 05/21/2023] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive blood cancer that stems from the rapid expansion of immature leukemic blasts in the bone marrow. Mutations in epigenetic factors represent the largest category of genetic drivers of AML. The chromatin assembly factor CHAF1B is a master epigenetic regulator of transcription associated with self-renewal and the undifferentiated state of AML blasts. Upregulation of CHAF1B, as observed in almost all AML samples, promotes leukemic progression by repressing the transcription of differentiation factors and tumor suppressors. However, the specific factors regulated by CHAF1B and their contributions to leukemogenesis are unstudied. We analyzed RNA sequencing data from mouse MLL-AF9 leukemic cells and bone marrow aspirates, representing a diverse collection of pediatric AML samples and identified the E3 ubiquitin ligase TRIM13 as a target of CHAF1B-mediated transcriptional repression associated with leukemogenesis. We found that CHAF1B binds the promoter of TRIM13, resulting in its transcriptional repression. In turn, TRIM13 suppresses self-renewal of leukemic cells by promoting pernicious entry into the cell cycle through its nuclear localization and catalytic ubiquitination of cell cycle-promoting protein, CCNA1. Overexpression of TRIM13 initially prompted a proliferative burst in AML cells, which was followed by exhaustion, whereas loss of total TRIM13 or deletion of its catalytic domain enhanced leukemogenesis in AML cell lines and patient-derived xenografts. These data suggest that CHAF1B promotes leukemic development, in part, by repressing TRIM13 expression and that this relationship is necessary for leukemic progression.
Collapse
Affiliation(s)
- Sarai T. Dean
- Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Chiharu Ishikawa
- Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- College of Medicine, University of Cincinnati, Cincinnati, OH
| | - Xiaoqin Zhu
- Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- College of Medicine, University of Cincinnati, Cincinnati, OH
| | - Sean Walulik
- Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Timothy Nixon
- Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- College of Medicine, University of Cincinnati, Cincinnati, OH
| | - Jessica K. Jordan
- Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Samantha Henderson
- Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Michael Wyder
- Department of Cancer Biology, Proteomics Laboratory, University of Cincinnati, Cincinnati, OH
| | - Nathan Salomonis
- Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- College of Medicine, University of Cincinnati, Cincinnati, OH
- Department of Cancer Biology, Proteomics Laboratory, University of Cincinnati, Cincinnati, OH
| | - Mark Wunderlich
- Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Kenneth D. Greis
- College of Medicine, University of Cincinnati, Cincinnati, OH
- Department of Cancer Biology, Proteomics Laboratory, University of Cincinnati, Cincinnati, OH
| | - Daniel T. Starczynowski
- Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- College of Medicine, University of Cincinnati, Cincinnati, OH
| | - Andrew G. Volk
- Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- College of Medicine, University of Cincinnati, Cincinnati, OH
| |
Collapse
|
19
|
Huang J, Yu Z, Li X, Yang M, Fang Q, Li Z, Wang C, Chen T, Cao X. E3 ligase HECTD3 promotes RNA virus replication and virus-induced inflammation via K33-linked polyubiquitination of PKR. Cell Death Dis 2023; 14:396. [PMID: 37402711 DOI: 10.1038/s41419-023-05923-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 06/17/2023] [Accepted: 06/23/2023] [Indexed: 07/06/2023]
Abstract
Uncontrolled viral replication and excessive inflammation are the main causes of death in the host infected with virus. Hence inhibition of intracellular viral replication and production of innate cytokines, which are the key strategies of hosts to fight virus infections, need to be finely tuned to eliminate viruses while avoid harmful inflammation. The E3 ligases in regulating virus replication and subsequent innate cytokines production remain to be fully characterized. Here we report that the deficiency of the E3 ubiquitin-protein ligase HECTD3 results in accelerated RNA virus clearance and reduced inflammatory response both in vitro and in vivo. Mechanistically, HECTD3 interacts with dsRNA-dependent protein kinase R (PKR) and mediates Lys33-linkage of PKR, which is the first non-proteolytic ubiquitin modification for PKR. This process disrupts the dimerization and phosphorylation of PKR and subsequent EIF2α activation, which results in the acceleration of virus replication, but promotes the formation of PKR-IKK complex and subsequent inflammatory response. The finding suggests HECTD3 is the potential therapeutic target for simultaneously restraining RNA virus replication and virus-induced inflammation once pharmacologically inhibited.
Collapse
Affiliation(s)
- Jiaying Huang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Zhou Yu
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China.
- National Key Laboratory of Immunity and Inflammation & Institute of Immunology, Navy Medical University, Shanghai, 200433, China.
| | - Xuelian Li
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China
| | - Mingjin Yang
- National Key Laboratory of Immunity and Inflammation & Institute of Immunology, Navy Medical University, Shanghai, 200433, China
| | - Qian Fang
- National Key Laboratory of Immunity and Inflammation & Institute of Immunology, Navy Medical University, Shanghai, 200433, China
| | - Zheng Li
- National Key Laboratory of Immunity and Inflammation & Institute of Immunology, Navy Medical University, Shanghai, 200433, China
| | - Chunmei Wang
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China
| | - Taoyong Chen
- National Key Laboratory of Immunity and Inflammation & Institute of Immunology, Navy Medical University, Shanghai, 200433, China
| | - Xuetao Cao
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- National Key Laboratory of Immunity and Inflammation, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, Jiangsu, China.
- National Key Laboratory of Immunity and Inflammation & Institute of Immunology, Navy Medical University, Shanghai, 200433, China.
- Institute of Immunology, College of Life Science, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
20
|
Kim J, Kim HS, Chung JH. Molecular mechanisms of mitochondrial DNA release and activation of the cGAS-STING pathway. Exp Mol Med 2023; 55:510-519. [PMID: 36964253 PMCID: PMC10037406 DOI: 10.1038/s12276-023-00965-7] [Citation(s) in RCA: 132] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 12/15/2022] [Indexed: 03/26/2023] Open
Abstract
In addition to constituting the genetic material of an organism, DNA is a tracer for the recognition of foreign pathogens and a trigger of the innate immune system. cGAS functions as a sensor of double-stranded DNA fragments and initiates an immune response via the adaptor protein STING. The cGAS-STING pathway not only defends cells against various DNA-containing pathogens but also modulates many pathological processes caused by the immune response to the ectopic localization of self-DNA, such as cytosolic mitochondrial DNA (mtDNA) and extranuclear chromatin. In addition, macrophages can cause inflammation by forming a class of protein complexes called inflammasomes, and the activation of the NLRP3 inflammasome requires the release of oxidized mtDNA. In innate immunity related to inflammasomes, mtDNA release is mediated by macropores that are formed on the outer membrane of mitochondria via VDAC oligomerization. These macropores are specifically formed in response to mitochondrial stress and tissue damage, and the inhibition of VDAC oligomerization mitigates this inflammatory response. The rapidly expanding area of research on the mechanisms by which mtDNA is released and triggers inflammation has revealed new treatment strategies not only for inflammation but also, surprisingly, for neurodegenerative diseases such as amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Jeonghan Kim
- Department of Biochemistry, The Catholic University of Korea College of Medicine, Seoul, 06591, South Korea.
| | - Ho-Shik Kim
- Department of Biochemistry, The Catholic University of Korea College of Medicine, Seoul, 06591, South Korea
| | - Jay H Chung
- Laboratory of Obesity and Aging Research, Cardiovascular Branch, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
21
|
Pan J, Fei CJ, Hu Y, Wu XY, Nie L, Chen J. Current understanding of the cGAS-STING signaling pathway: Structure, regulatory mechanisms, and related diseases. Zool Res 2023; 44:183-218. [PMID: 36579404 PMCID: PMC9841179 DOI: 10.24272/j.issn.2095-8137.2022.464] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 12/27/2022] [Indexed: 01/04/2023] Open
Abstract
The innate immune system protects the host from external pathogens and internal damage in various ways. The cGAS-STING signaling pathway, comprised of cyclic GMP-AMP synthase (cGAS), stimulator of interferon genes (STING), and downstream signaling adaptors, plays an essential role in protective immune defense against microbial DNA and internal damaged-associated DNA and is responsible for various immune-related diseases. After binding with DNA, cytosolic cGAS undergoes conformational change and DNA-linked liquid-liquid phase separation to produce 2'3'-cGAMP for the activation of endoplasmic reticulum (ER)-localized STING. However, further studies revealed that cGAS is predominantly expressed in the nucleus and strictly tethered to chromatin to prevent binding with nuclear DNA, and functions differently from cytosolic-localized cGAS. Detailed delineation of this pathway, including its structure, signaling, and regulatory mechanisms, is of great significance to fully understand the diversity of cGAS-STING activation and signaling and will be of benefit for the treatment of inflammatory diseases and cancer. Here, we review recent progress on the above-mentioned perspectives of the cGAS-STING signaling pathway and discuss new avenues for further study.
Collapse
Affiliation(s)
- Jing Pan
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang 315211, China
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, Zhejiang 315832, China
- Zhejiang Key Laboratory of Marine Bioengineering, Ningbo University, Ningbo, Zhejiang 315832, China
| | - Chen-Jie Fei
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang 315211, China
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, Zhejiang 315832, China
- Zhejiang Key Laboratory of Marine Bioengineering, Ningbo University, Ningbo, Zhejiang 315832, China
| | - Yang Hu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang 315211, China
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, Zhejiang 315832, China
- Zhejiang Key Laboratory of Marine Bioengineering, Ningbo University, Ningbo, Zhejiang 315832, China
| | - Xiang-Yu Wu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang 315211, China
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, Zhejiang 315832, China
- Zhejiang Key Laboratory of Marine Bioengineering, Ningbo University, Ningbo, Zhejiang 315832, China
| | - Li Nie
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang 315211, China
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, Zhejiang 315832, China
- Zhejiang Key Laboratory of Marine Bioengineering, Ningbo University, Ningbo, Zhejiang 315832, China. E-mail:
| | - Jiong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, Zhejiang 315211, China
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Meishan Campus, Ningbo University, Ningbo, Zhejiang 315832, China
- Zhejiang Key Laboratory of Marine Bioengineering, Ningbo University, Ningbo, Zhejiang 315832, China. E-mail:
| |
Collapse
|
22
|
Zhang Z, Zhou H, Ouyang X, Dong Y, Sarapultsev A, Luo S, Hu D. Multifaceted functions of STING in human health and disease: from molecular mechanism to targeted strategy. Signal Transduct Target Ther 2022; 7:394. [PMID: 36550103 PMCID: PMC9780328 DOI: 10.1038/s41392-022-01252-z] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 10/25/2022] [Accepted: 11/09/2022] [Indexed: 12/24/2022] Open
Abstract
Since the discovery of Stimulator of Interferon Genes (STING) as an important pivot for cytosolic DNA sensation and interferon (IFN) induction, intensive efforts have been endeavored to clarify the molecular mechanism of its activation, its physiological function as a ubiquitously expressed protein, and to explore its potential as a therapeutic target in a wide range of immune-related diseases. With its orthodox ligand 2'3'-cyclic GMP-AMP (2'3'-cGAMP) and the upstream sensor 2'3'-cGAMP synthase (cGAS) to be found, STING acquires its central functionality in the best-studied signaling cascade, namely the cGAS-STING-IFN pathway. However, recently updated research through structural research, genetic screening, and biochemical assay greatly extends the current knowledge of STING biology. A second ligand pocket was recently discovered in the transmembrane domain for a synthetic agonist. On its downstream outputs, accumulating studies sketch primordial and multifaceted roles of STING beyond its cytokine-inducing function, such as autophagy, cell death, metabolic modulation, endoplasmic reticulum (ER) stress, and RNA virus restriction. Furthermore, with the expansion of the STING interactome, the details of STING trafficking also get clearer. After retrospecting the brief history of viral interference and the milestone events since the discovery of STING, we present a vivid panorama of STING biology taking into account the details of the biochemical assay and structural information, especially its versatile outputs and functions beyond IFN induction. We also summarize the roles of STING in the pathogenesis of various diseases and highlight the development of small-molecular compounds targeting STING for disease treatment in combination with the latest research. Finally, we discuss the open questions imperative to answer.
Collapse
Affiliation(s)
- Zili Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Haifeng Zhou
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Xiaohu Ouyang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Yalan Dong
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Alexey Sarapultsev
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049, Ekaterinburg, Russia
| | - Shanshan Luo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Desheng Hu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
- Key Laboratory of Biological Targeted Therapy, The Ministry of Education, 430022, Wuhan, China.
- Clinical Research Center of Cancer Immunotherapy, 430022, Hubei, Wuhan, China.
| |
Collapse
|
23
|
Liu J, Rui K, Peng N, Luo H, Zhu B, Zuo X, Lu L, Chen J, Tian J. The cGAS-STING pathway: Post-translational modifications and functional implications in diseases. Cytokine Growth Factor Rev 2022; 68:69-80. [PMID: 36151014 DOI: 10.1016/j.cytogfr.2022.09.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 09/12/2022] [Indexed: 01/30/2023]
Abstract
Recent studies have illustrated the functional significance of DNA recognition in the activation of innate immune responses among a variety of diseases. The cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway has been found to be modulated by post-translational modifications and can regulate the immune response via type I IFNs. Accumulating evidence indicates a pivotal role of cGAS-STING signaling, being protective or pathogenic, in the development of diseases. Thus, a comprehensive understanding of the post-translational modifications of cGAS-STING pathway and their role in disease development will provide insights in predicting individual disease outcomes and developing appropriate therapies. In this review, we will discuss the regulation of the cGAS-STING pathway and its implications in disease pathologies, as well as pharmacologic strategies to target the cGAS-STING pathway for therapeutic intervention.
Collapse
Affiliation(s)
- Jun Liu
- Institute of Medical Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, China; Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Ke Rui
- Institute of Medical Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, China; Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| | - Na Peng
- Department of Rheumatology, the Second People's Hospital, China Three Gorges University, Yichang, China
| | - Hui Luo
- Department of Rheumatology and immunology, Xiangya Hospital, Central South University, Changsha, China
| | - Bo Zhu
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiaoxia Zuo
- Department of Rheumatology and immunology, Xiangya Hospital, Central South University, Changsha, China
| | - Liwei Lu
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong; Chongqing International Institute for Immunology, China
| | - Jixiang Chen
- Department of Gastrointestinal Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| | - Jie Tian
- Institute of Medical Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, China; Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China.
| |
Collapse
|
24
|
Nishitsuji H, Iwahori S, Ohmori M, Shimotohno K, Murata T. Ubiquitination of SARS-CoV-2 NSP6 and ORF7a Facilitates NF-κB Activation. mBio 2022; 13:e0097122. [PMID: 35856559 PMCID: PMC9426613 DOI: 10.1128/mbio.00971-22] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 07/01/2022] [Indexed: 11/20/2022] Open
Abstract
Patients with severe coronavirus disease 2019 tend to have high levels of proinflammatory cytokines, which eventually lead to cytokine storm and the development of acute respiratory distress syndrome. However, the detailed molecular mechanisms of proinflammatory cytokine production remain unknown. Here, we screened severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) genes and found that nonstructural protein 6 (NSP6) and open reading frame 7a (ORF7a) activated the NF-κB pathway. NSP6 and ORF7a interacted with transforming growth factor β-activated kinase 1 (TAK1), and knockout (KO) of TAK1 or NF-κB essential modulator (NEMO) abolished NF-κB activation by NSP6 and ORF7a. Interestingly, K61 of NSP6 was conjugated to K63-linked polyubiquitin chains by the E3 ubiquitin ligase tripartite motif-containing 13, and this polyubiquitination of NSP6 appeared crucial for recruitment of NEMO to the NSP6-TAK1 complex and NF-κB activation. On the other hand, ring finger protein 121 (RNF121) was required for the polyubiquitination of ORF7a. Knockdown of RNF121 significantly decreased ORF7a binding of TAK1 and NEMO, resulting in the suppression of NF-κB activation. Taken together, our results provide novel molecular insights into the pathogenesis of SARS-CoV-2 and the host immune response to SARS-CoV-2 infection. IMPORTANCE The detailed molecular basis of the induction of proinflammatory cytokines and chemokines by SARS-CoV-2 is unclear, although such induction is clearly related to the severity of COVID-19. Here, we show that SARS-CoV-2 NSP6 and ORF7a lead to NF-κB activation through associations with TAK1. K63-linked polyubiquitination of NSP6 and ORF7a by TRIM13 and RNF121, respectively, appears essential for NF-κB activation. These results suggest that inhibition of the NSP6 and ORF7a gene products may reduce the severity of COVID-19 symptoms by decreasing proinflammatory cytokine levels.
Collapse
Affiliation(s)
- Hironori Nishitsuji
- Department of Virology and Parasitology, Fujita Health University School of Medicine, Aichi, Japan
| | - Satoko Iwahori
- Department of Virology and Parasitology, Fujita Health University School of Medicine, Aichi, Japan
| | - Mariko Ohmori
- Department of Virology and Parasitology, Fujita Health University School of Medicine, Aichi, Japan
| | - Kunitada Shimotohno
- The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Chiba, Japan
| | - Takayuki Murata
- Department of Virology and Parasitology, Fujita Health University School of Medicine, Aichi, Japan
| |
Collapse
|
25
|
Kang J, Wu J, Liu Q, Wu X, Zhao Y, Ren J. Post-Translational Modifications of STING: A Potential Therapeutic Target. Front Immunol 2022; 13:888147. [PMID: 35603197 PMCID: PMC9120648 DOI: 10.3389/fimmu.2022.888147] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/11/2022] [Indexed: 12/18/2022] Open
Abstract
Stimulator of interferon genes (STING) is an endoplasmic-reticulum resident protein, playing essential roles in immune responses against microbial infections. However, over-activation of STING is accompanied by excessive inflammation and results in various diseases, including autoinflammatory diseases and cancers. Therefore, precise regulation of STING activities is critical for adequate immune protection while limiting abnormal tissue damage. Numerous mechanisms regulate STING to maintain homeostasis, including protein-protein interaction and molecular modification. Among these, post-translational modifications (PTMs) are key to accurately orchestrating the activation and degradation of STING by temporarily changing the structure of STING. In this review, we focus on the emerging roles of PTMs that regulate activation and inhibition of STING, and provide insights into the roles of the PTMs of STING in disease pathogenesis and as potential targeted therapy.
Collapse
Affiliation(s)
- Jiaqi Kang
- Research Institute of General Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jie Wu
- Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China
| | - Qinjie Liu
- Research Institute of General Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xiuwen Wu
- Research Institute of General Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yun Zhao
- Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China
| | - Jianan Ren
- Research Institute of General Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|