1
|
Zhang Z, Wang X, Zhao C, Zhu H, Liao X, Tsai HI. STING and metabolism-related diseases: Roles, mechanisms, and applications. Cell Signal 2025; 132:111833. [PMID: 40294833 DOI: 10.1016/j.cellsig.2025.111833] [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: 01/23/2025] [Revised: 04/08/2025] [Accepted: 04/22/2025] [Indexed: 04/30/2025]
Abstract
The stimulator of interferon genes (STING) pathway plays a critical role in innate immunity, acting as a central mediator that links cytosolic DNA sensing to inflammatory signaling. STING not only responds to cellular metabolic states but also actively regulates key metabolic processes, including glycolysis, lipid metabolism, and redox balance. This bidirectional interaction underscores the existence of a dynamic feedback mechanism between STING signaling and metabolic pathways, which is essential for maintaining cellular homeostasis. This review provides a comprehensive analysis, beginning with an in-depth overview of the classical STING signaling pathway, followed by a detailed examination of its reciprocal regulation of various metabolic pathways. Additionally, it explores the role and mechanisms of STING signaling in metabolic disorders, including obesity, diabetes, and atherosclerosis. By integrating these insights into the mutual regulation between STING and its metabolism, novel therapeutic strategies targeting this pathway in metabolic diseases have been proposed.
Collapse
Affiliation(s)
- Zhengyang Zhang
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China; School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Xirui Wang
- Department of Biomedical Engineering, School of Medical Imaging, Xuzhou Medical University, Xuzhou 221000, China
| | - Chuangchuang Zhao
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China; School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Haitao Zhu
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China; Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Xiang Liao
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China.
| | - Hsiang-I Tsai
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang 212001, China; Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| |
Collapse
|
2
|
Yang H, Xia Y, Ma Y, Gao M, Hou S, Xu S, Wang Y. Inhibition of the cGAS-STING pathway: contributing to the treatment of cerebral ischemia-reperfusion injury. Neural Regen Res 2025; 20:1900-1918. [PMID: 38993125 PMCID: PMC11691458 DOI: 10.4103/nrr.nrr-d-24-00015] [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: 01/05/2024] [Revised: 03/05/2024] [Accepted: 05/02/2024] [Indexed: 07/13/2024] Open
Abstract
The cGAS-STING pathway plays an important role in ischemia-reperfusion injury in the heart, liver, brain, and kidney, but its role and mechanisms in cerebral ischemia-reperfusion injury have not been systematically reviewed. Here, we outline the components of the cGAS-STING pathway and then analyze its role in autophagy, ferroptosis, cellular pyroptosis, disequilibrium of calcium homeostasis, inflammatory responses, disruption of the blood-brain barrier, microglia transformation, and complement system activation following cerebral ischemia-reperfusion injury. We further analyze the value of cGAS-STING pathway inhibitors in the treatment of cerebral ischemia-reperfusion injury and conclude that the pathway can regulate cerebral ischemia-reperfusion injury through multiple mechanisms. Inhibition of the cGAS-STING pathway may be helpful in the treatment of cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Hang Yang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Yulei Xia
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Yue Ma
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Mingtong Gao
- Department of Emergency, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong Province, China
| | - Shuai Hou
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Shanshan Xu
- Department of Emergency, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong Province, China
| | - Yanqiang Wang
- Department of Neurology II, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong Province, China
| |
Collapse
|
3
|
Zhang H, Xu X, Li S, Huang H, Zhang K, Li W, Wang X, Yang J, Yin X, Qu C, Ni J, Dong X. Advances in nanoplatform-based multimodal combination therapy activating STING pathway for enhanced anti-tumor immunotherapy. Colloids Surf B Biointerfaces 2025; 250:114573. [PMID: 39983453 DOI: 10.1016/j.colsurfb.2025.114573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 01/24/2025] [Accepted: 02/16/2025] [Indexed: 02/23/2025]
Abstract
Activation of the cyclic GMP-AMP synthase(cGAS)-stimulator of interferon genes (STING) has great potential to promote antitumor immunity. As a major effector of the cell to sense and respond to the aberrant presence of cytoplasmic double-stranded DNA (dsDNA), inducing the expression and secretion of type I interferons (IFN) and STING, cGAS-STING signaling pathway establishes an effective natural immune response, which is one of the fundamental mechanisms of host defense in organisms. In addition to the release of heterologous DNA due to pathogen invasion and replication, mitochondrial damage and massive cell death can also cause abnormal leakage of the body's own dsDNA, which is then recognized by the DNA receptor cGAS and activates the cGAS-STING signaling pathway. However, small molecule STING agonists suffer from rapid excretion, low bioavailability, non-specificity and adverse effects, which limits their therapeutic efficacy and in vivo application. Various types of nano-delivery systems, on the other hand, make use of the different unique structures and surface modifications of nanoparticles to circumvent the defects of small molecule STING agonists such as fast metabolism and low bioavailability. Also, the nanoparticles are precisely directed to the focal site, with their own appropriate particle size combined with the characteristics of passive or active targeting. Herein, combined with the cGAS-STING pathway to activate the immune system and kill tumor tissues directly or indirectly, which help maximize the use of the functions of chemotherapy, photothermal therapy(PTT), chemodynamic therapy(CDT), and radiotherapy(RT). In this review, we will discuss the mechanism of action of the cGAS-STING pathway and introduce nanoparticle-mediated tumor combination therapy based on the STING pathway. Collectively, the effective multimodal nanoplatform, which can activate cGAS-STING pathway for enhanced anti-tumor immunotherapy, has promising avenue clinical applications for cancer treatment.
Collapse
Affiliation(s)
- Huizhong Zhang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xiaohan Xu
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Shiman Li
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Huating Huang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Ke Zhang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Wenjing Li
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xinzhu Wang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jingwen Yang
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xingbin Yin
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Changhai Qu
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jian Ni
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Xiaoxv Dong
- School of Chinese Material Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
4
|
Zou Y, Tao Z, Li P, Yang J, Xu Q, Xu X, Miao Z, Zhao X. Clemastine attenuates subarachnoid haemorrhage pathology in a mouse model via Nrf2/SQSTM1-mediated autophagy. Br J Pharmacol 2025; 182:2730-2753. [PMID: 40052261 DOI: 10.1111/bph.17465] [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: 05/29/2024] [Revised: 12/31/2024] [Accepted: 01/03/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND AND PURPOSE Subarachnoid haemorrhage (SAH) is an uncommon and severe subtype of stroke, but the availability of drugs for its treatment is limited. Enhanced autophagy is believed to attenuate SAH pathology; however, autophagy level is tentatively up-regulated and then down-regulated after SAH onset in mice. Clemastine, a first-generation histamine H1R antagonist, is believed to persistently enhance autophagy. However, the precise mechanism of clemastine in the treatment of SAH remains largely elusive. EXPERIMENTAL APPROACH Haemoglobin-induced neuron injury model and autologous-blood-injected SAH-model mice were used to investigate the effects of clemastine in vitro and in vivo, respectively. The expressions of Nrf2/Keap1 and autophagy-related proteins were detected using western blotting and immunofluorescence. Neuronal injury and hyperoxide level were measured via Fluoro-Jade C and dihydroethidium staining. Neurological behaviours were evaluated using modified Garcia Scale, beam balance test, Morris water maze, Y-maze and novel object recognition test. The structures of autophagosomes and mitochondria were visualised using transmission electron microscope. The binding sites of clemastine was predicted and verified using database and drug affinity-responsive target stability. KEY RESULTS Clemastine ameliorated SAH pathogenesis in vivo and in vitro. Moreover, the intraperitoneal injection of clemastine and its oral administration reduced neuronal death and improved cognitive deficits in SAH-model mice. Mechanistically, clemastine directly bound to muscarinic acetylcholine receptor M4, prevented Nrf2 degradation via Nrf2/Keap1/SQSTM1 pathway and promoted Nrf2 nuclear translocation, thus enhancing autophagy-related gene transcription and autophagy activation. CONCLUSIONS AND IMPLICATIONS Clemastine can attenuate SAH pathology via the activation of Nrf2/SQSTM1 autophagy and could be a useful therapeutic in the context of SAH.
Collapse
Affiliation(s)
- Yan Zou
- Department of Neurosurgery, Jiangnan University Medical Center, Wuxi, China
| | - Zhen'xing Tao
- Neuroscience Center, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Peng'peng Li
- Neuroscience Center, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Jie'qiong Yang
- Neuroscience Center, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Qin'yi Xu
- Department of Hepatobiliary Surgery, Jiangnan University Medical Center, Wuxi, China
| | - Xing Xu
- Department of Neurosurgery, Jiangnan University Medical Center, Wuxi, China
| | - Zeng'li Miao
- Department of Neurosurgery, Jiangnan University Medical Center, Wuxi, China
- Wuxi Neurosurgical Institute, Wuxi, China
| | - Xu'dong Zhao
- Department of Neurosurgery, Jiangnan University Medical Center, Wuxi, China
- Wuxi Neurosurgical Institute, Wuxi, China
- Department of Clinical Medicine, Medical College, Nantong University, Nantong, China
| |
Collapse
|
5
|
Liu Y, Hu Y, Shan ZL. Mitochondrial DNA release mediates metabolic-associated steatohepatitis via activation of inflammatory pathways. Shijie Huaren Xiaohua Zazhi 2025; 33:344-360. [DOI: 10.11569/wcjd.v33.i5.344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/25/2025] [Accepted: 05/19/2025] [Indexed: 05/28/2025] Open
Affiliation(s)
- Ying Liu
- Gannan Institute of Medical Innovation and Translational Medicine, Gannan Medical University, Ganzhou 431000, Jiangxi Province, China
| | - Yang Hu
- Gannan Institute of Medical Innovation and Translational Medicine, Gannan Medical University, Ganzhou 431000, Jiangxi Province, China
| | - Zhao-Liang Shan
- Gannan Institute of Medical Innovation and Translational Medicine, Gannan Medical University, Ganzhou 431000, Jiangxi Province, China
| |
Collapse
|
6
|
Zhou Q, Luo J, Chai X, Yang J, Zhong S, Zhang Z, Chang X, Wang H. Therapeutic targeting the cGAS-STING pathway associated with protein and gene: An emerging and promising novel strategy for aging-related neurodegenerative disease. Int Immunopharmacol 2025; 156:114679. [PMID: 40252469 DOI: 10.1016/j.intimp.2025.114679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 04/11/2025] [Accepted: 04/13/2025] [Indexed: 04/21/2025]
Abstract
Neurodegenerative diseases (NDDs) represent a rapidly escalating global health challenge, contributing significantly to the worldwide disease burden and posing substantial threats to public health systems across nations. Among the many risk factors for neurodegeneration, aging is the major risk factor. In the context of aging, multiple factors lead to the release of endogenous DNA (especially mitochondrial DNA, mtDNA), which is an important trigger for the activation of the cGAS-STING innate immune pathway. Recent studies have identified an increasing role for activation of the cGAS-STING signaling pathway as a driver of senescence-associated secretory phenotypes (SASPs) in aging and NDDs. The cGAS-STING pathway mediates the immune sensing of DNA and is a key driver of chronic inflammation and functional decline during the aging process. Blocking cGAS-STING signaling may reduce the inflammatory response by preventing mtDNA release and enhancing mitophagy. Targeted inhibition of the cGAS-STING pathway by biological macromolecules such as natural products shows promise in therapeutic strategies for age-related NDDs. This review aims to systematically and comprehensively introduces the role of the cGAS-STING pathway in age-related NDDs in the context of aging while revealing the molecular mechanisms of the cGAS-STING pathway and its downstream signaling pathways and to develop more targeted and effective therapeutic strategies for NDDs.
Collapse
Affiliation(s)
- Qiongli Zhou
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China
| | - Jinghao Luo
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China
| | - Xueting Chai
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China
| | - Jirui Yang
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China
| | - Shiyin Zhong
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China
| | - Zhimin Zhang
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China
| | - Xuhong Chang
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China
| | - Hui Wang
- Department of Toxicology, School of Public Health, Lanzhou University, Gansu 730000, China.
| |
Collapse
|
7
|
Barros D, Ferreira BH, Garcia-Gonzalez P, Carbone F, Luka M, Leite-Pinheiro F, Machado MD, Nikolaou T, Pilotti A, Goguet E, Antas P, Mendes A, Zhang L, Cresci M, Galliot L, Gigan JP, Reverendo M, Su B, Narita M, Paton AW, Paton JC, Rocchi S, Rieux-Laucat F, Argüello RJ, Nal B, Liang Y, Ménager M, Gatti E, Almeida CR, Pierre P. Induction of the ISR by AB5 subtilase cytotoxin drives type-I IFN expression in pDCs via STING activation. Proc Natl Acad Sci U S A 2025; 122:e2421258122. [PMID: 40388626 DOI: 10.1073/pnas.2421258122] [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: 10/15/2024] [Accepted: 04/17/2025] [Indexed: 05/21/2025] Open
Abstract
We demonstrate that exposure to the AB5 subtilase cytotoxin (SubAB) induces the unfolded protein response (UPR) in human peripheral blood mononuclear cells, concomitant with a proinflammatory response across distinct cell subsets. Notably, SubAB selectively induces type-I interferon (IFN) expression in plasmacytoid dendritic cells, acting synergistically with Toll-like receptor 7 stimulation. The induction of type-I IFN in response to SubAB relies on stimulator of interferon genes (STING) activation, coupled with protein synthesis inhibition mediated by protein kinase R-like endoplasmic reticulum kinase (PERK) and phosphorylation of the eukaryotic translation initiation factor 2 subunit-alpha. By impeding mRNA translation through the integrated stress response, SubAB precipitates the downregulation of the negative innate signaling feedback regulator Tax1-binding protein 1. This downregulation is necessary to unleash TANK-binding kinase 1 signaling associated with STING activation. These findings shed light on how UPR-inducing conditions may regulate the immune system during infection or pathogenesis.
Collapse
Affiliation(s)
- Daniela Barros
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Beatriz H Ferreira
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
- Centre for Research in Ceramics and Composite Materials (CICECO)-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Aveiro 3810-193, Portugal
| | - Paulina Garcia-Gonzalez
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Francesco Carbone
- Université de Paris Cité, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM U1163, Paris F-75015, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM, Paris F-75015, France
| | - Marine Luka
- Université de Paris Cité, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM U1163, Paris F-75015, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM, Paris F-75015, France
| | - Fátima Leite-Pinheiro
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
| | - Mariana D Machado
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
| | - Theopisti Nikolaou
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Angelo Pilotti
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Eliot Goguet
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Paulo Antas
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
| | - Andreia Mendes
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Lichen Zhang
- School of Medical Technology, Henan Key Laboratory of Immunology and Targeted Therapy, Xinxiang Medical University, Xinxiang 453003, People's Republic of China
| | - Marina Cresci
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Lou Galliot
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Julien P Gigan
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Marisa Reverendo
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
| | - Bing Su
- Shanghai Institute of Immunology, Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Miwako Narita
- Laboratory of Hematology and Oncology, Graduate School of Health Sciences, Niigata University, Niigata 951-8518, Japan
| | - Adrienne W Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA 5005, Australia
| | - James C Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA 5005, Australia
| | | | - Frédéric Rieux-Laucat
- Université de Paris Cité, Imagine Institute Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, Paris 75015, France
| | - Rafael J Argüello
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Béatrice Nal
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Yinming Liang
- School of Medical Technology, Henan Key Laboratory of Immunology and Targeted Therapy, Xinxiang Medical University, Xinxiang 453003, People's Republic of China
| | - Mickaël Ménager
- Université de Paris Cité, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM U1163, Paris F-75015, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM, Paris F-75015, France
| | - Evelina Gatti
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
| | - Catarina R Almeida
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
| | - Philippe Pierre
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro 3810-193, Portugal
- Aix Marseille Université, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), U2, UMR7280 and U1104, Marseille 13288 Cedex 9, France
- School of Medical Technology, Henan Key Laboratory of Immunology and Targeted Therapy, Xinxiang Medical University, Xinxiang 453003, People's Republic of China
| |
Collapse
|
8
|
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
|
9
|
Dhapola R, Paidlewar M, Kumari S, Sharma P, Vellingiri B, Medhi B, HariKrishnaReddy D. cGAS-STING and neurodegenerative diseases: A molecular crosstalk and therapeutic perspective. Int Immunopharmacol 2025; 159:114902. [PMID: 40403503 DOI: 10.1016/j.intimp.2025.114902] [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: 02/21/2025] [Revised: 05/05/2025] [Accepted: 05/15/2025] [Indexed: 05/24/2025]
Abstract
Neurodegenerative disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), Amyotrophic Lateral Sclerosis (ALS), Multiple Sclerosis (MS) and Frontotemporal Dementia (FTD) share key pathological features, including neuroinflammation, oxidative stress, mitochondrial dysfunction, autophagic dysfunction, and DNA damage. By identifying cytosolic DNA and triggering the type I interferon response, the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway regulates neuroinflammation. Dysregulated cGAS-STING signaling has been linked to neuroinflammation and neuronal degeneration across multiple neurodegenerative conditions. In many neurodegenerative disorders, neuroinflammation is mediated by the cGAS-STING pathway. Mitochondrial malfunction and impaired autophagy cause cytosolic DNA buildup in Huntington's, Parkinson's, and Alzheimer's diseases, which activates cGAS-STING and drives chronic inflammation. This pathway is triggered by TDP-43 pathology and nucleic acid dysregulation in ALS and FTD, which leads to neuronal destruction. Both central demyelination and peripheral immunological responses are linked to cGAS-STING activation in multiple sclerosis. Various inhibitors, such as RU.521, H-151, and naturally occurring compounds like metformin, potentially attenuate cGAS-STING-mediated neuroinflammation and associated pathologies. H-151 significantly decreased the expression of pro-inflammatory markers in murine macrophage J774 cells activated with cGAMP: TNF-α by 68 %, IFN-β by 84 %, and CXCL10 by 96 %. cGAS-STING inhibitors target neuroinflammation, offering a disease-modifying approach unlike current symptomatic treatments. However, challenges like blood-brain barrier penetration, off-target effects, and immune suppression hinder clinical translation, necessitating optimized drug delivery and immune modulation. With a focus on its potential for future clinical applications, this review explores the role of the cGAS-STING pathway in neurodegeneration and new treatment approaches.
Collapse
Affiliation(s)
- Rishika Dhapola
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda 151401, Punjab, India
| | - Mohit Paidlewar
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda 151401, Punjab, India
| | - Sneha Kumari
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda 151401, Punjab, India
| | - Prajjwal Sharma
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda 151401, Punjab, India
| | - Balachandar Vellingiri
- Human Cytogenetics and Stem Cell Laboratory, Department of Zoology, School of Basic Sciences, Central University of Punjab, 151401 Bathinda, Punjab, India
| | - Bikash Medhi
- Department of Pharmacology, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Dibbanti HariKrishnaReddy
- Advanced Pharmacology and Neuroscience Laboratory, Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda 151401, Punjab, India.
| |
Collapse
|
10
|
Li L, He Y, Chen Y, Zhou X. cGAS-STING Pathway's Impact on Intestinal Barrier. J Gastroenterol Hepatol 2025. [PMID: 40377214 DOI: 10.1111/jgh.16974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 03/09/2025] [Accepted: 04/03/2025] [Indexed: 05/18/2025]
Abstract
Intestinal inflammation and increased permeability have been linked to metabolic dysregulation in patients with compromised intestinal barrier function. Among the pathways, garnering attention is the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway. Upon binding to double-stranded DNA (dsDNA), cGAS catalyzes the conversion of ATP and GTP into cyclic GMP-AMP (cGAMP). Subsequently, cGAMP binds to STING, triggering the activation of tank-binding kinase 1 (TBK1), which activates interferon regulatory factor 3 (IRF3), thus inducing the production of type I interferon. Activated TBK1 can also induce the activation of nuclear factor κB (NF-κB), thus mediating the production of proinflammatory cytokines. The effects of this process vary among innate and adaptive immune cells, as well as intestinal epithelial cells (IECs). This review aims to elucidate the impact and role of the cGAS-STING pathway on intestinal barrier function.
Collapse
Affiliation(s)
- Liqi Li
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yingge He
- Department of Thyroid and Breast Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Yu Chen
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China
| | - Xiaoshu Zhou
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
11
|
Wang XT, Zhu X, Lian ZH, Liu Q, Yan HH, Qiu Y, Ge XY. AUP1 and UBE2G2 complex targets STING signaling and regulates virus-induced innate immunity. mBio 2025; 16:e0060225. [PMID: 40237449 PMCID: PMC12077101 DOI: 10.1128/mbio.00602-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 03/18/2025] [Indexed: 04/18/2025] Open
Abstract
Stimulator of interferon genes (STING) is an endoplasmic reticulum (ER) signaling adaptor that is essential for the host immune response triggered by DNA pathogens. Precise regulation of STING is crucial for maintaining a balanced immune response and preventing harmful autoinflammation. Activation of STING requires its translocation from the ER to the Golgi apparatus. However, the mechanisms that maintain STING in its resting state remain largely unclear. Here, we find that deficiency of the ancient ubiquitous protein 1 (AUP1) causes spontaneous activation of STING and enhances the expression of type I interferons (IFNs) under resting conditions. Furthermore, deficiency of UBE2G2, a cofactor of AUP1, also promotes the abnormal activation of STING. AUP1 deficiency significantly enhances STING signaling induced by DNA virus, and AUP1 deficiency exhibits increased resistance to DNA virus infection in vitro and in vivo. Mechanistically, AUP1 may form a complex with UBE2G2 to interact with STING, preventing its exit from the ER membrane. Notably, infection with the RNA virus vesicular stomatitis virus (VSV) promotes the accumulation of lipid droplets (LDs) and AUP1 proteins. Additionally, AUP1 deficiency markedly inhibits the replication of VSV because AUP1 deficiency reduces lipid accumulation and alters the expression of lipid metabolism genes, such as carnitine palmitoyltransferase 1A (CPT1A), monoglyceride lipase (MGLL), and sterol regulatory element-binding transcription factor 1 (SREBF1). This study uncovers the essential roles of AUP1 in the STING signaling pathway and lipid metabolism pathway, highlighting its dual role in regulating virus replication.IMPORTANCEThe stimulator of interferon genes (STING) signaling cascade plays an essential role in coordinating innate immunity against DNA pathogens and autoimmunity. Precise regulation of the innate immune response is essential for maintaining homeostasis. In this study, we demonstrate that ancient ubiquitous protein 1 (AUP1) and UBE2G2 act as negative regulators of the innate immune response by targeting STING. Notably, AUP1 interacts with STING to retain STING in the endoplasmic reticulum (ER), preventing STING translocation and thereby limiting STING signaling in the resting state. In addition, deficiency of AUP1 markedly inhibits the replication of DNA virus and RNA virus. Our findings provide new insights into the regulation of STING signaling and confirm AUP1 has a dual role in regulating virus replication.
Collapse
Affiliation(s)
- Xin-Tao Wang
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, Hunan, China
| | - Xi Zhu
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, Hunan, China
| | - Zhong-Hao Lian
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, Hunan, China
| | - Qian Liu
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, Hunan, China
| | - Hui-Hui Yan
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, Hunan, China
| | - Ye Qiu
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, Hunan, China
| | - Xing-Yi Ge
- Hunan Provincial Key Laboratory of Medical Virology, Institute of Pathogen Biology and Immunology, College of Biology, Hunan University, Changsha, Hunan, China
| |
Collapse
|
12
|
Acharya D, Sayyad Z, Hoenigsperger H, Hirschenberger M, Zurenski M, Balakrishnan K, Zhu J, Gableske S, Kato J, Zhang SY, Casanova JL, Moss J, Sparrer KMJ, Gack MU. TRIM23 mediates cGAS-induced autophagy in anti-HSV defense. Nat Commun 2025; 16:4418. [PMID: 40360474 PMCID: PMC12075517 DOI: 10.1038/s41467-025-59338-5] [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: 07/16/2024] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
The cGAS-STING pathway, well-known to elicit interferon (IFN) responses, is also a key inducer of autophagy upon virus infection or other stimuli. Whereas the mediators for cGAS-induced IFN responses are well characterized, much less is known about how cGAS elicits autophagy. Here, we report that TRIM23, a unique TRIM protein harboring both ubiquitin E3 ligase and GTPase activity, is crucial for cGAS-STING-dependent antiviral autophagy. Genetic ablation of TRIM23 impairs autophagic control of HSV-1 infection. HSV-1 infection or cGAS-STING stimulation induces TBK1-mediated TRIM23 phosphorylation at S39, which triggers TRIM23 autoubiquitination and GTPase activity and ultimately elicits autophagy. Fibroblasts from a patient with herpes simplex encephalitis heterozygous for a dominant-negative, kinase-inactivating TBK1 mutation fail to activate autophagy by TRIM23 and cGAS-STING. Our results thus identify the cGAS-STING-TBK1-TRIM23 axis as a key autophagy defense pathway and may stimulate new therapeutic interventions for viral or inflammatory diseases.
Collapse
Affiliation(s)
- Dhiraj Acharya
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, USA
- Department of Microbiology, The University of Chicago, Chicago, IL, USA
| | - Zuberwasim Sayyad
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, USA
| | | | | | - Matthew Zurenski
- Department of Microbiology, The University of Chicago, Chicago, IL, USA
| | - Kannan Balakrishnan
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, USA
| | - Junji Zhu
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, USA
| | - Sebastian Gableske
- Department of Microbiology, The University of Chicago, Chicago, IL, USA
- Eisai GmbH, Frankfurt am Main, Germany
| | - Jiro Kato
- The Critical Care Medicine and Pulmonary Branch; National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Shen-Ying Zhang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- Howard Hughes Medical Institute, New York, NY, USA
| | - Joel Moss
- The Critical Care Medicine and Pulmonary Branch; National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Konstantin M J Sparrer
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany
| | - Michaela U Gack
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, USA.
- Department of Microbiology, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
13
|
Glass AM, Navas-Martin S. Interferon-induced protein ISG15 in the central nervous system, quo vadis? FEBS Lett 2025. [PMID: 40353372 DOI: 10.1002/1873-3468.70063] [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: 12/23/2024] [Revised: 04/03/2025] [Accepted: 04/17/2025] [Indexed: 05/14/2025]
Abstract
The ubiquitin-like interferon (IFN)-stimulated gene 15 (ISG15) is a unique molecular effector that functions both intra- and extracellularly. Central to its pleiotropic nature is the ability to coordinate cellular responses following its conjugation to target proteins via ISGylation or in its free form. The activity of ISG15 is highly context-dependent: in the case of viral infections, ISG15 can serve as a pro- or antiviral factor. While ISG15 has been studied extensively, several gaps persist in our understanding of its role in dysregulated immune homeostasis. In particular, the role of ISG15 in the central nervous system (CNS), which has traditionally been considered an immune-privileged site, remains ill-defined. Interestingly, elevated ISG15 expression is observed in the CNS following instances of brain injury, autoimmunity, neurodegeneration, and viral infection. In this review, we seek to provide a comprehensive analysis of these studies as they pertain to ISG15 and its potential roles in the CNS. Furthermore, we discuss questions and challenges in the field while highlighting ISG15 as a potential diagnostic biomarker or therapeutic target. Impact statement While ISG15 has been studied extensively, several gaps remain in our understanding of its role in dysregulated immune homeostasis and its impact within the central nervous system (CNS). In this review, we provide a comprehensive analysis of the emerging roles of ISG15 in brain injury, autoimmunity, neurodegeneration, and viral infection within the CNS.
Collapse
Affiliation(s)
- Adam M Glass
- Department of Microbiology and Immunology, Centers for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Sonia Navas-Martin
- Department of Microbiology and Immunology, Centers for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
14
|
Zhang S, Shan X, Qu G, Sun A, Xing J, Liu Q, Liu Z, Wang Y, Zhang X, Cao X, Zeng Y, Wang J, Wang C, Hu J. Lactiplantibacillus plantarum extracellular vesicles exert anti-PEDV effects through STING-dependent autophagy. BMC Microbiol 2025; 25:271. [PMID: 40329214 PMCID: PMC12057047 DOI: 10.1186/s12866-025-04019-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: 12/08/2024] [Accepted: 05/02/2025] [Indexed: 05/08/2025] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) infection causes severe gastrointestinal and lethal disease in piglets, leading to huge economic losses for the swine industry worldwide. Recent studies have emphasized probiotics can regulate innate immunity and cellular functions through interaction with intestinal epithelial cells via extracellular vesicles (EVs) as effective carriers. The cGAS-STING signaling pathway is crucial for inducing type I interferons (IFNs) to establish antiviral innate immunity. It also triggers cellular autophagy, which helps maintain intracellular environmental homeostasis. In our study, we found that Lactiplantibacillus plantarum extracellular vesicles (LpEVs) significantly activated the cGAS-STING signaling pathway in porcine intestinal epithelial cells (IPEC-J2), thereby enhancing antiviral immune responses. Notably, compared to the untreated control group, 10 μg/mL LpEVs retained the capacity to activate the cGAS-STING pathway, but their activation efficacy was significantly lower than that of 2.5 μg/mL, suggesting a potential feedback regulatory mechanism at higher concentrations. Furthermore, 10 μg/mL LpEVs regulated cGAS-STING activation through autophagy induction, and this autophagic response was STING-dependent. Additionally, LpEVs at concentrations of 2.5, 5, and 10 μg/mL all significantly inhibited the proliferation of PEDV. However, 10 μg/mL LpEVs exhibited a stronger inhibitory effect on PEDV replication compared to 2.5 or 5 μg/mL doses, and this enhanced antiviral activity was closely associated with autophagy. Our findings elucidate the underlying mechanism of antiviral effects of probiotics through regulating innate immunity and autophagy, which highlights the critical role of LpEVs in preventing PEDV infection as a potential antiviral agent.
Collapse
Grants
- 20240601058RC Jilin Province Science and Technology Project
- 20240601058RC Jilin Province Science and Technology Project
- 20240601058RC Jilin Province Science and Technology Project
- 20240601058RC Jilin Province Science and Technology Project
- 20240601058RC Jilin Province Science and Technology Project
- 20240601058RC Jilin Province Science and Technology Project
- 21A20261, 32273043 National Natural Science Foundation of China
- 21A20261, 32273043 National Natural Science Foundation of China
- 21A20261, 32273043 National Natural Science Foundation of China
- 21A20261, 32273043 National Natural Science Foundation of China
- 21A20261, 32273043 National Natural Science Foundation of China
- 21A20261, 32273043 National Natural Science Foundation of China
- 21A20261, 32273043 National Natural Science Foundation of China
- 21A20261, 32273043 National Natural Science Foundation of China
- 21A20261, 32273043 National Natural Science Foundation of China
- 21A20261, 32273043 National Natural Science Foundation of China
- S202310193135, s202410193009 Jilin province college students' innovation and entrepreneurship training program project
- S202310193135, s202410193009 Jilin province college students' innovation and entrepreneurship training program project
- S202310193135, s202410193009 Jilin province college students' innovation and entrepreneurship training program project
Collapse
Affiliation(s)
- Shihan Zhang
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Changchun, 130118, China
- Animal Medical College, Jilin Agricultural University, Changchun, 130118, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Xin Shan
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Changchun, 130118, China
- Animal Medical College, Jilin Agricultural University, Changchun, 130118, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Guanglong Qu
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Changchun, 130118, China
- Animal Medical College, Jilin Agricultural University, Changchun, 130118, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Anqi Sun
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Changchun, 130118, China
- Animal Medical College, Jilin Agricultural University, Changchun, 130118, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Junhong Xing
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Changchun, 130118, China
- Animal Medical College, Jilin Agricultural University, Changchun, 130118, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Qiuyuan Liu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Zixuan Liu
- Animal Medical College, Jilin Agricultural University, Changchun, 130118, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Yibing Wang
- Animal Medical College, Jilin Agricultural University, Changchun, 130118, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Xinyue Zhang
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Changchun, 130118, China
- Animal Medical College, Jilin Agricultural University, Changchun, 130118, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Xin Cao
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Changchun, 130118, China
- Animal Medical College, Jilin Agricultural University, Changchun, 130118, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Yan Zeng
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Changchun, 130118, China
- Animal Medical College, Jilin Agricultural University, Changchun, 130118, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Jianzhong Wang
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Changchun, 130118, China
- Animal Medical College, Jilin Agricultural University, Changchun, 130118, China
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Chunfeng Wang
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Changchun, 130118, China.
- Animal Medical College, Jilin Agricultural University, Changchun, 130118, China.
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China.
| | - Jingtao Hu
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Changchun, 130118, China.
- Animal Medical College, Jilin Agricultural University, Changchun, 130118, China.
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
15
|
Yang X, Lv L, Zhang Y, Zhang Z, Zeng S, Zhang X, Wang Q, Dorf M, Li S, Fu B. ATP2A2 regulates STING1/MITA-driven signal transduction including selective autophagy. Autophagy 2025:1-16. [PMID: 40265346 DOI: 10.1080/15548627.2025.2496786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 04/08/2025] [Accepted: 04/18/2025] [Indexed: 04/24/2025] Open
Abstract
STING1/MITA not only induces innate immune responses but also triggers macroautophagy/autophagy to selectively degrade signaling molecules. However, the molecular mechanisms regulating STING1-mediated selective autophagy remain unclear. Here, we first report that ATP2A2 directly interacts with STING1, regulating STING1-mediated innate immune response by modulating its polymerization and trafficking, thereby inhibiting DNA virus infection. Notably, while screening for reticulophagy receptors involved in STING1-mediated selective autophagy, we identified SEC62 as an important receptor protein in STING1-mediated reticulophagy. Mechanistically, SEC62 strengthens its interaction with STING1 upon activation and concurrently facilitates STING1-mediated reticulophagy upon starvation, which are dependent on ATP2A2. Furthermore, knocking down SEC62 in WT cells inhibits STING1-mediated MAP1LC3B/LC3B lipidation and autophagosome formation, an effect that is lost in ATP2A2 knockout cells, suggesting that SEC62's role in STING1-mediated selective autophagy is ATP2A2 dependent. Thus, our findings identify the reticulophagy receptor SEC62 as a novel receptor protein regulating STING1-mediated selective autophagy, providing new insight into the mechanism regarding a reticulophagy receptor in the process of STING1-induced selective autophagy.Abbrevations: aa: amino acids; AP-MS: affinity tag purification-mass spectrometry; ATP2A1: ATPase sarcoplasmic/endoplasmic reticulum Ca2+ transporting 1; ATP2A2: ATPase sarcoplasmic/endoplasmic reticulum Ca2+ transporting 2; ATP2A3: ATPase sarcoplasmic/endoplasmic reticulum Ca2+ transporting 3; CANX: calnexin; CCPG1: cell cycle progression 1; CGAS: cyclic GMP-AMP synthase; ctDNA: calf thymus DNA; dsRNA: double-stranded RNA; diABZI: diamidobenzimidazole; ER: endoplasmic reticulum; ERGIC: ER-Golgi intermediate compartment; EBSS: Earle's Balanced Salt Solution; EV: empty vector; FL: full length; GOLGA2/GM130: golgin A2; HSV-1: herpes simplex virus type 1; IRF3: interferon regulatory factor 3; IFNs: type I interferons; ISD: interferon stimulatory DNA; KO: knockout; MAVS: mitochondrial antiviral signaling protein; MOI: multiplicity of infection; poly(I:C): polyinosinic-polycytidylic acid; NBR1: NBR1 autophagy cargo receptor; PRR: pattern recognition receptor; reticulophagy: selective autophagic degradation of the ER; RETREG1/FAM134B: reticulophagy regulator 1; RIGI: RNA sensor RIG-I; RTN3L: reticulon 3; SEC62: SEC62 homolog, preprotein translocation factor; SeV: Sendai virus; STIM1: stromal interaction molecule 1; STING1/MITA: stimulator of interferon response cGAMP interactor 1; TBK1: TANK binding kinase 1; TEX264: testis expressed 264, ER-phagy receptor; TMX1: thioredoxin related transmembrane protein 1; VSV: vesicular stomatitis virus; VACV: vaccinia virus; ZMPSTE24: zinc metallopeptidase STE24.
Collapse
Affiliation(s)
- Xue Yang
- Department of Rheumatology and Immunology, State Key Laboratory of Virology and Biosafety, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Linyue Lv
- Department of Rheumatology and Immunology, State Key Laboratory of Virology and Biosafety, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Yuelan Zhang
- Department of Rheumatology and Immunology, State Key Laboratory of Virology and Biosafety, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Zhuyou Zhang
- Department of Rheumatology and Immunology, State Key Laboratory of Virology and Biosafety, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Shaowei Zeng
- Department of Rheumatology and Immunology, State Key Laboratory of Virology and Biosafety, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Xinyi Zhang
- Department of Rheumatology and Immunology, State Key Laboratory of Virology and Biosafety, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Qinyang Wang
- Department of Rheumatology and Immunology, State Key Laboratory of Virology and Biosafety, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Martin Dorf
- Department of Microbiology & Immunobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Shitao Li
- Department of Microbiology and Immunology, Tulane University, New Orleans, LA, USA
| | - Bishi Fu
- Department of Rheumatology and Immunology, State Key Laboratory of Virology and Biosafety, Zhongnan Hospital, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| |
Collapse
|
16
|
Jiang Q, Chen Z, Jiang J, Chen Q, Lan H, Zhu J, Mao W. The role of cGAS-STING in remodeling the tumor immune microenvironment induced by radiotherapy. Crit Rev Oncol Hematol 2025; 209:104658. [PMID: 39956501 DOI: 10.1016/j.critrevonc.2025.104658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/06/2025] [Accepted: 02/11/2025] [Indexed: 02/18/2025] Open
Abstract
The activation of the cGAS-STING pathway occurs when tumor cell DNA is damaged by ionizing radiation. Once triggered, this pathway reshapes the tumor immune microenvironment by promoting the maturation, activation, polarization, and immune-killing capacity of immune cells, as well as by inducing the release of interferons and the expression of immune-related genes. In addition, the gut microbiota and various mechanisms of programmed cell death interact with the cGAS-STING pathway, further influencing its function in remodeling the immune microenvironment after radiotherapy. Therefore, investigating the mechanisms of the cGAS-STING pathway in reshaping the tumor immune microenvironment post-radiotherapy can not only optimize the efficacy of combined radiotherapy and immunotherapy but also provide new research directions and potential targets for cancer treatment.
Collapse
Affiliation(s)
- Qingyu Jiang
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310000, China; Zhejiang Chinese Medical University, Hangzhou 310053, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310000, China
| | - Zhiheng Chen
- Department of Oncology, Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing 31400, China
| | - Jin Jiang
- Department of Oncology, Affiliated Hospital of Jiaxing University, The First Hospital of Jiaxing, Jiaxing 31400, China
| | - Qianping Chen
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310000, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310000, China
| | - Huiyin Lan
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310000, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310000, China
| | - Ji Zhu
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310000, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310000, China.
| | - Wei Mao
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310000, China; Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310000, China; Zhejiang Key Laboratory of Radiation Oncology, Hangzhou 310000, China.
| |
Collapse
|
17
|
Giordano L, Ware SA, Lagranha CJ, Kaufman BA. Mitochondrial DNA signals driving immune responses: Why, How, Where? Cell Commun Signal 2025; 23:192. [PMID: 40264103 PMCID: PMC12012978 DOI: 10.1186/s12964-025-02042-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/14/2025] [Indexed: 04/24/2025] Open
Abstract
There has been a recent expansion in our understanding of DNA-sensing mechanisms. Mitochondrial dysfunction, oxidative and proteostatic stresses, instability and impaired disposal of nucleoids cause the release of mitochondrial DNA (mtDNA) from the mitochondria in several human diseases, as well as in cell culture and animal models. Mitochondrial DNA mislocalized to the cytosol and/or the extracellular compartments can trigger innate immune and inflammation responses by binding DNA-sensing receptors (DSRs). Here, we define the features that make mtDNA highly immunogenic and the mechanisms of its release from the mitochondria into the cytosol and the extracellular compartments. We describe the major DSRs that bind mtDNA such as cyclic guanosine-monophosphate-adenosine-monophosphate synthase (cGAS), Z-DNA-binding protein 1 (ZBP1), NOD-, LRR-, and PYD- domain-containing protein 3 receptor (NLRP3), absent in melanoma 2 (AIM2) and toll-like receptor 9 (TLR9), and their downstream signaling cascades. We summarize the key findings, novelties, and gaps of mislocalized mtDNA as a driving signal of immune responses in vascular, metabolic, kidney, lung, and neurodegenerative diseases, as well as viral and bacterial infections. Finally, we define common strategies to induce or inhibit mtDNA release and propose challenges to advance the field.
Collapse
Affiliation(s)
- Luca Giordano
- Center for Metabolism and Mitochondrial Medicine, Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA.
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Cardio-Pulmonary Institute (CPI), Justus-Liebig-University, Giessen, Germany.
| | - Sarah A Ware
- Center for Metabolism and Mitochondrial Medicine, Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Claudia J Lagranha
- Center for Metabolism and Mitochondrial Medicine, Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brett A Kaufman
- Center for Metabolism and Mitochondrial Medicine, Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
- Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
18
|
Luo C, Ma C, Xu G, Lu C, Ma J, Huang Y, Nie L, Yu C, Xia Y, Liu Z, Zhu Y, Liu S. Hepatitis B surface antigen hijacks TANK-binding kinase 1 to suppress type I interferon and induce early autophagy. Cell Death Dis 2025; 16:304. [PMID: 40234418 PMCID: PMC12000394 DOI: 10.1038/s41419-025-07605-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 03/19/2025] [Accepted: 03/27/2025] [Indexed: 04/17/2025]
Abstract
There are close links between innate immunity and autophagy. However, the crosstalk between innate immunity and autophagy in host cells infected with hepatitis B virus (HBV) remains unclear. Here, we reported that HBsAg suppressed type I interferon production and induced the accumulation of autophagosomes. HBsAg boosted TANK-binding kinase 1 (TBK1) phosphorylation and depressed interferon regulatory factor 3 (IRF3) phosphorylation ex vivo and in vivo. Mechanistic studies showed that HBsAg interaction with the kinase domain (KD) of TBK1 augmented its dimerization but disrupted TBK1-IRF3 complexes. Using the TBK1 inhibitor, BX795, we discovered that HBsAg-enhanced TBK1 dimerization, promoting sequestosome-1 (p62) phosphorylation, was necessary for HBV-induced autophagy and HBV replication. Moreover, HBsAg blocked autophagosome-lysosome fusion by inhibiting the synaptosomal-associated protein 29 (SNAP29) promoter. Notably, liver tissues from HBsAg transgenic mice or chronic HBV patients revealed that IFNβ signaling was inhibited and incomplete autophagy was induced. These findings suggest a novel mechanism by which HBsAg targets TBK1 to inhibit type I interferon and induce early autophagy, possibly leading to persistent HBV infection. Molecular mechanisms of HBsAg suppression of the IFNβ signaling pathway and triggering of early autophagy. HBsAg targets the kinase domain of TBK1, thereby disrupting the TBK1-IRF3 complex and inhibiting type I interferon production. On the other hand, HBsAg enhances TBK1 dimerization and phosphorylation, which upregulates the phosphorylation of p62 to induce p62-mediated autophagy. Furthermore, HBV infection causes the accumulation of autophagosomes. This is achieved by HBsAg suppressing the SNAP29 promoter activity, which blocks autophagosome-lysosome fusion.
Collapse
Affiliation(s)
- Chuanjin Luo
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Caijiao Ma
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Gang Xu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Chengbo Lu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - June Ma
- Department of Clinical Laboratory, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Huang
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Longyu Nie
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Chen Yu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yongfang Xia
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Zhiqiang Liu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Ying Zhu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China.
| | - Shi Liu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China.
| |
Collapse
|
19
|
Terraza-Silvestre E, Villamuera R, Bandera-Linero J, Letek M, Oña-Sánchez D, Ramón-Barros C, Moyano-Jimeno C, Pimentel-Muiños FX. An unconventional autophagic pathway that inhibits ATP secretion during apoptotic cell death. Nat Commun 2025; 16:3409. [PMID: 40210636 PMCID: PMC11986000 DOI: 10.1038/s41467-025-58619-3] [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: 02/16/2024] [Accepted: 03/26/2025] [Indexed: 04/12/2025] Open
Abstract
Mobilisation of Damage-Associated Molecular Patterns (DAMPs) determines the immunogenic properties of apoptosis, but the mechanisms that control DAMP exposure are still unclear. Here we describe an unconventional autophagic pathway that inhibits the release of ATP, a critical DAMP in immunogenic apoptosis, from dying cells. Mitochondrial BAK activated by BH3-only molecules interacts with prohibitins and stomatin-1 through its latch domain, indicating the existence of an interactome specifically assembled by unfolded BAK. This complex engages the WD40 domain of the autophagic effector ATG16L1 to induce unconventional autophagy, and the resulting LC3-positive vesicles contain ATP. Functional interference with the pathway increases ATP release during cell death, reduces ATP levels remaining in the apoptotic bodies, and improves phagocyte activation. These results reveal that an unconventional component of the autophagic burst that often accompanies apoptosis sequesters intracellular ATP to prevent its release, thus favouring the immunosilent nature of apoptotic cell death.
Collapse
Affiliation(s)
- Elena Terraza-Silvestre
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Nicolás Cabrera, 1, 28049, Madrid, Spain
| | - Raquel Villamuera
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Nicolás Cabrera, 1, 28049, Madrid, Spain
| | - Julia Bandera-Linero
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Nicolás Cabrera, 1, 28049, Madrid, Spain
| | - Michal Letek
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Nicolás Cabrera, 1, 28049, Madrid, Spain
- Departamento de Biología Molecular, Área de Microbiología, Universidad de León, 24071, León, Spain
| | - Daniel Oña-Sánchez
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Nicolás Cabrera, 1, 28049, Madrid, Spain
| | - Cristina Ramón-Barros
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Nicolás Cabrera, 1, 28049, Madrid, Spain
| | - Clara Moyano-Jimeno
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Nicolás Cabrera, 1, 28049, Madrid, Spain
| | - Felipe X Pimentel-Muiños
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Nicolás Cabrera, 1, 28049, Madrid, Spain.
| |
Collapse
|
20
|
Canova PN, Katzenell S, Cerón S, Charron AJ, Pesola JM, Oh HS, Coen DM, Knipe DM, Leib DA. Regulation of the innate immune response in human neurons by ICP34.5 maintains herpes simplex virus 1 latency. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.04.647253. [PMID: 40291710 PMCID: PMC12026746 DOI: 10.1101/2025.04.04.647253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Herpes simplex virus 1 (HSV-1) establishes latent infections in sensory neurons, from which HSV sporadically reactivates due to external stress and other stimuli. Latency and reactivation are studied through in vivo models in a variety of hosts, as well as in vitro models using primary neurons, and neurons derived from pluripotent stem cells (iPSCs). These systems behave disparately, but the reasons remain unknown. The interferon (IFN)-based neuronal innate immune response is critical in controlling HSV-1 replication and HSV-1 counters these responses in part through infectedcell protein 34.5 (ICP34.5). ICP34.5 also promotes neurovirulence by preventing host translational shutoff and interfering with host cell autophagy. Here we demonstrate in a human iPSC neuronal model that sustaining host translation is the key activity of ICP34.5 for enhancement of reactivation. Specifically, our data shows that ICP34.5 was key for maintenance of HSV-1 latency. While interaction of ICP34.5 with the autophagy regulator Beclin 1 was important for maintaining latency, this was not due to modulation of bulk autophagy. Our work from primary mouse neurons suggested that the major effect of ICP34.5 on latency maintenance occurs in an IRF3/7-dependent manner. Notably, the role of ICP34.5 in regulating latency and reactivation differs between neurons derived from human iPSCs (iNeurons) and primary mouse trigeminal (TG) neurons. This highlights the importance of selecting an appropriate neuronal model and validating experimental outcomes in multiple models.
Collapse
|
21
|
Yang D, Peng N, Zhang H, Qiu Z, Xu L, Pan M. Cordycepin ameliorates autoimmunity by promoting STING degradation via autophagy pathway. Br J Pharmacol 2025; 182:1546-1560. [PMID: 39675775 DOI: 10.1111/bph.17425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/23/2024] [Accepted: 11/18/2024] [Indexed: 12/17/2024] Open
Abstract
BACKGROUND AND PURPOSE Stimulator of interferon response cGAMP interactor 1 (STING), a central hub protein of cyclic GMP-AMP synthase (cGAS)-STING signalling pathway, has a crucial role in regulating type I interferons (IFNs) production and response. Recent studies indicate that excessive activation of STING is strongly associated with autoimmune diseases, including systemic lupus erythematosus (SLE). Searching immunomodulators that negatively regulate STING might greatly contribute to the suppression of autoimmunity. EXPERIMENTAL APPROACH The peripheral blood mononuclear cells (PBMCs) of SLE patients, Hela cells, L929 cells and bone marrow-derived macrophages (BMDMs) from mice were used as in vitro models. While, Trex1 KO mouse autoimmune disease model was used as in vivo model. After treatment with cordycepin, a nucleoside from Cordyceps mushrooms, type I IFNs production and response were determined by western blotting, real-time polymerase chain reaction (PCR), dual-luciferase assay, enzyme-linked immunosorbent assay (ELISA), haematoxylin-eosin staining and RNA-seq. KEY RESULTS Cordycepin inhibited type I IFNs production and response in human and murine systems following cGAS-STING signalling activation. Importantly, cordycepin markedly attenuates the autoinflammatory and autoimmune responses in Trex1 KO BMDMs and Trex1 KO mice. Furthermore, cordycepin effectively suppressed the production of type I IFNs and interferon-stimulated genes (ISGs) in the PBMCs of SLE patients. Mechanistically, cordycepin promoted STING degradation via autophagy pathway upon DNA stimulation. CONCLUSION AND IMPLICATIONS This study shows that cordycepin promotes STING autophagic degradation to alleviate autoimmunity upon DNA stimulation. Cordycepin might be a potential therapeutic candidate for alleviating aberrant type I IFNs in autoimmune and autoinflammatory diseases.
Collapse
Affiliation(s)
- Daidi Yang
- Department of Ophthalmology, The First People's Hospital of Wuhu, Wuhu, China
| | - Niannian Peng
- School of Pharmacy, Jiangsu Food and Pharmaceutical Science College, Huaian, China
| | - Hongqian Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Zuocheng Qiu
- Guangdong Provincial Key Laboratory of Speed Capability Research, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Lingxiao Xu
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mingyu Pan
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Department of Biomedical Science, City University of Hong Kong, Kowloon, Hong Kong
| |
Collapse
|
22
|
Liang P, Li Z, Chen Z, Chen Z, He F, Jin T, Cao Y, Yang K. HER2 regulates autophagy and promotes migration in gastric cancer cells through the cGAS-STING pathway. Anticancer Drugs 2025; 36:306-318. [PMID: 39869353 PMCID: PMC11884795 DOI: 10.1097/cad.0000000000001680] [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: 12/10/2024] [Accepted: 12/10/2024] [Indexed: 01/28/2025]
Abstract
In gastric cancer, the relationship between human epidermal growth factor receptor 2 (HER2), the cyclic GMP-AMP synthase-stimulator of the interferon genes (cGAS-STING) pathway, and autophagy remains unclear. This study examines whether HER2 regulates autophagy in gastric cancer cells via the cGAS-STING signaling pathway, influencing key processes such as cell proliferation and migration. Understanding this relationship could uncover new molecular targets for diagnosis and treatment. Through lentiviral transfection, cell counting kit-8 assays, colony formation, transwell migration, scratch assays, and siRNA, we found that HER2 overexpression suppresses the cGAS-STING pathway, inhibits autophagy, and enhances the migratory ability of gastric cancer cells. In contrast, HER2 knockdown activates the cGAS-STING pathway, promotes autophagy, and reduces cell migration. We further observed that the inhibition of autophagy using chloroquine (CQ) increases the migration ability of HER2-overexpressing cells. Moreover, interfering with STING expression reversed the migration defects caused by HER2 knockdown, underscoring the critical role of the cGAS-STING pathway in HER2-regulated cell migration. We also revealed that high STING expression in gastric cancer is significantly associated with poor prognosis. STING expression was identified as an independent prognostic factor for survival (hazard ratio, 1.942; 95% confidence interval, 1.06-3.54; P = 0.031). These results highlight the importance of HER2-driven regulation of autophagy through the cGAS-STING pathway in gastric cancer progression and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Panping Liang
- Department of General Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zedong Li
- Department of General Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zhengwen Chen
- Department of General Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zehua Chen
- Department of General Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Fengjun He
- Department of General Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Jin
- Department of General Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yuwei Cao
- Department of General Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Kun Yang
- Department of General Surgery and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
23
|
Li C, Ma A, Bai Y, Liu Z, Tian L, Wang Z, Ma H, Chen Z, Gao Z, Feng S, Fu P. TRIM21 promotes type I interferon by inhibiting the autophagic degradation of STING via p62/SQSTM1 ubiquitination in systemic lupus erythematosus. Acta Biochim Biophys Sin (Shanghai) 2025. [PMID: 40165656 DOI: 10.3724/abbs.2025046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025] Open
Abstract
The cGAS-STING signaling pathway serves as a pivotal surveillance mechanism for cytosolic double-stranded DNA (dsDNA) detection in mammalian systems. While STING-mediated type I interferon production is crucial for host defense, sustained activation of this pathway contributes to autoimmune pathologies, including systemic lupus erythematosus (SLE). Maintaining immune homeostasis requires precise regulation of STING activity to prevent hyperactivation. Our study identifies TRIM21 as a novel positive regulator of cGAS-STING signaling in SLE pathogenesis. Our results demonstrate that TRIM21 overexpression stabilizes STING by suppressing autophagic degradation, whereas TRIM21 depletion accelerates this clearance process. Mechanistically, TRIM21 catalyzes the K63-linked polyubiquitylation of the selective autophagy receptor p62/SQSTM1, disrupting its interaction with STING. This post-translational modification prevents the sequestration of STING into autophagosomes, thereby stabilizing the adaptor protein and amplifying downstream type I interferon responses. Our findings reveal a previously unrecognized regulatory circuit in which TRIM21 orchestrates cross-talk between ubiquitin signaling and autophagy to control STING turnover. The TRIM21-p62 axis represents a potential therapeutic target for attenuating pathological interferon production in STING-dependent autoimmune disorders. This work advances our understanding of immune regulation by demonstrating how E3 ligase-mediated ubiquitin modifications modulate cargo recognition in selective autophagy pathways. The identified mechanism provides new insights into the molecular interplay between protein ubiquitylation and autophagic degradation in maintaining the innate immune balance, offering novel perspectives for developing targeted therapies against interferonopathies associated with cGAS-STING hyperactivity.
Collapse
Affiliation(s)
- Chen Li
- Department of Rheumatology and Clinical Immunology, the Second Affiliated Hospital of Kunming Medical University, Kunming 650032, China
- Department of Scientific Research, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming 650118, China
| | - Ang Ma
- Department of Rheumatology and Clinical Immunology, the Second Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Yu Bai
- Department of Urology, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming 650118, China
| | - Zitao Liu
- Department of Scientific Research, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming 650118, China
| | - Linghan Tian
- Department of Scientific Research, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming 650118, China
| | - Ziyuan Wang
- Cancer Institute, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital, Kunming 650118, China
| | - Huaishun Ma
- Department of Scientific Research, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming 650118, China
| | - Zhengpu Chen
- Department of Scientific Research, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming 650118, China
| | - Zhengheng Gao
- Department of Health Management and Tumor Screening Center, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming 650118, China
| | - Shijie Feng
- Department of Rheumatology and Clinical Immunology, the Second Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Ping Fu
- Department of Rheumatology and Clinical Immunology, the Second Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| |
Collapse
|
24
|
Shen Y, Huang W, Nie J, Zhang L. Progress Update on STING Agonists as Vaccine Adjuvants. Vaccines (Basel) 2025; 13:371. [PMID: 40333245 PMCID: PMC12030840 DOI: 10.3390/vaccines13040371] [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: 02/18/2025] [Revised: 03/18/2025] [Accepted: 03/27/2025] [Indexed: 05/09/2025] Open
Abstract
Low antigen immunogenicity poses a significant challenge in vaccine development, often leading to inadequate immune responses and reduced vaccine efficacy. Therefore, the discovery of potent immune-enhancing adjuvants is crucial. STING (stimulator of interferon genes) agonists are a promising class of adjuvants which have been identified in various immune cells and are activated in response to DNA fragments, triggering a broad range of type-I interferon-dependent immune responses. Integrating STING agonists with vaccine components is an ideal strategy to bolster vaccine-induced immunity to infections and cancer cells. Several STING agonists are currently under investigation in preclinical studies and clinical trials; however, some have shown limited efficacy, while others exhibit off-target effects. To ensure safety, they are typically delivered with carriers that exhibit high biocompatibility and insolubility. In this review, we present the latest research on natural and synthetic STING agonists that have been effectively used in vaccine development, and summarize their application in adjuvant preventive and therapeutic vaccines. Additionally, we discuss the safety of STING agonists as vaccine adjuvants by reviewing potential delivery strategies. Overall, incorporating STING agonists into vaccine formulations represents a significant advancement in vaccine research with the potential to significantly enhance immune responses and improve vaccine efficacy. However, ongoing research is still required to identify the most effective and safe delivery strategies for STING agonists, as well as to evaluate their long-term safety and efficacy in clinical trials.
Collapse
Affiliation(s)
- Yanru Shen
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China; (Y.S.); (W.H.); (J.N.)
| | - Weijin Huang
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China; (Y.S.); (W.H.); (J.N.)
- WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing 102629, China
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products and NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing 102629, China
| | - Jianhui Nie
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China; (Y.S.); (W.H.); (J.N.)
- WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing 102629, China
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products and NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing 102629, China
| | - Li Zhang
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China; (Y.S.); (W.H.); (J.N.)
- WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing 102629, China
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products and NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing 102629, China
| |
Collapse
|
25
|
Zhang R, Yu C, Zeh HJ, Kroemer G, Klionsky DJ, Tang D, Kang R. TAX1BP1-dependent autophagic degradation of STING1 impairs anti-tumor immunity. Autophagy 2025:1-22. [PMID: 40000606 DOI: 10.1080/15548627.2025.2471736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/02/2024] [Accepted: 02/21/2025] [Indexed: 02/27/2025] Open
Abstract
The activation of STING1 can lead to the production and secretion of cytokines, initiating antitumor immunity. Here, we screened an ion channel ligand library and identified tetrandrine, a bis-benzylisoquinoline alkaloid, as an immunological adjuvant that enhances antitumor immunity by preventing the autophagic degradation of the STING1 protein. This tetrandrine effect is independent of its known function as a calcium or potassium channel blocker. Instead, tetrandrine inhibits lysosomal function, impairing cathepsin maturation, and autophagic degradation. Proteomic analysis of lysosomes identified TAX1BP1 as a novel autophagic receptor for the proteolysis of STING1. TAX1BP1 recognizes STING1 through the physical interaction of its coiled-coil domain with the cyclic dinucleotide binding domain of STING1. Systematic mutation of lysine (K) residues revealed that K63-ubiquitination of STING1 at the K224 site ignites TAX1BP1-dependent STING1 degradation. Combined treatment with tetrandrine and STING1 agonists promotes antitumor immunity by converting "cold" pancreatic cancers into "hot" tumors. This process is associated with enhanced cytokine release and increased infiltration of cytotoxic T-cells into the tumor microenvironment. The antitumor immunity mediated by tetrandrine and STING1 agonists is limited by neutralizing antibodies to the type I interferon receptor or CD8+ T cells. Thus, these findings establish a potential immunotherapeutic strategy against pancreatic cancer by preventing the autophagic degradation of STING1.
Collapse
Affiliation(s)
- Ruoxi Zhang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Chunhua Yu
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Herbert J Zeh
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Department of Biology, Pôle de Biologie, Institut du Cancer Paris CARPEM, Paris, France
| | - Daniel J Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
26
|
Zhang X, Chen Y, Liu X, Li G, Zhang S, Zhang Q, Cui Z, Qin M, Simon HU, Terzić J, Kocic G, Polić B, Yin C, Li X, Zheng T, Liu B, Zhu Y. STING in cancer immunoediting: Modeling tumor-immune dynamics throughout cancer development. Cancer Lett 2025; 612:217410. [PMID: 39826670 DOI: 10.1016/j.canlet.2024.217410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/16/2024] [Accepted: 12/21/2024] [Indexed: 01/22/2025]
Abstract
Cancer immunoediting is a dynamic process of tumor-immune system interaction that plays a critical role in cancer development and progression. Recent studies have highlighted the importance of innate signaling pathways possessed by both cancer cells and immune cells in this process. The STING molecule, a pivotal innate immune signaling molecule, mediates DNA-triggered immune responses in both cancer cells and immune cells, modulating the anti-tumor immune response and shaping the efficacy of immunotherapy. Emerging evidence has shown that the activation of STING signaling has dual opposing effects in cancer progression, simultaneously provoking and restricting anti-tumor immunity, and participating in every phase of cancer immunoediting, including immune elimination, equilibrium, and escape. In this review, we elucidate the roles of STING in the process of cancer immunoediting and discuss the dichotomous effects of STING agonists in the cancer immunotherapy response or resistance. A profound understanding of the sophisticated roles of STING signaling pathway in cancer immunoediting would potentially inspire the development of novel cancer therapeutic approaches and overcome the undesirable protumor effects of STING activation.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, People's Republic of China; Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Yan Chen
- Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Xi Liu
- Department of Cardiology, ordos central hospital, Ordos, People's Republic of China
| | - Guoli Li
- Department of Colorectal and Anal Surgery, Chifeng Municipal Hospital, Chifeng Clinical Medical School of Inner Mongolia Medical University, Chifeng, People's Republic of China
| | - Shuo Zhang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, People's Republic of China
| | - Qi Zhang
- Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Zihan Cui
- Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Minglu Qin
- Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland; Institute of Biochemistry, Brandenburg Medical School, Neuruppin, 16816, Germany
| | - Janoš Terzić
- Laboratory for Cancer Research, University of Split School of Medicine, Split, Croatia
| | - Gordana Kocic
- Department of Biochemistry, Faculty of Medicine, University of Nis, 18000 Nis, Serbia
| | - Bojan Polić
- University of Rijeka Faculty of Medicine, Croatia
| | - Chengliang Yin
- Faculty of Medicine, Macau University of Science and Technology, 999078, Macao.
| | - Xiaobo Li
- Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China.
| | - Tongsen Zheng
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, No.150 Haping Road, Nangang District, Harbin, Heilongjiang, People's Republic of China.
| | - Bing Liu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, People's Republic of China; School of Stomatology, Harbin Medical University, Harbin, 150001, People's Republic of China.
| | - Yuanyuan Zhu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, People's Republic of China; Department of Pathology, Harbin Medical University, Harbin, 150081, People's Republic of China.
| |
Collapse
|
27
|
Zhang X, Liu J, Zhong S, Zhang Z, Zhou Q, Yang J, Chang X, Wang H. Exposure to Manganese Induces Autophagy-Lysosomal Pathway Dysfunction-Mediated Tauopathy by Activating the cGAS-STING Pathway in the Brain. ENVIRONMENT & HEALTH (WASHINGTON, D.C.) 2025; 3:199-212. [PMID: 40012869 PMCID: PMC11851216 DOI: 10.1021/envhealth.4c00176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 02/28/2025]
Abstract
Manganese (Mn) exposure leads to pathological accumulation of Tau-associated neurodegenerative disease and has become a major public health concern. However, the precise mechanism underlying this effect remains unclear. Here, the mechanism by which Mn induces dysfunction of autophagy-lysosomal pathway-mediated tauopathy by activating the cGAS-STING pathway was explored both in vitro and in vivo. Mn exposure induced tauopathy in microglia and in mice while activating the cGAS-STING pathway, inducing type I interferon production, and impairing the degradation function of the autophagy-lysosomal pathway. Importantly, inactivation of the cGAS-STING pathway rescued the degradation activity of the autophagy-lysosomal pathway, while tauopathy was markedly attenuated, as shown in both cGAS-knockout and STING-knockout BV2 microglia and in mice. Moreover, the autophagy inhibitor 3-methyladenine (3-MA) restored the impaired degradation activity of the autophagy-lysosomal pathway by inactivating the cGAS-STING pathway, thereby clearing Tau aggregation. Taken together, these results indicate that Mn exposure induces tauopathy by impairing the function of the autophagy-lysosomal pathway through the activation of the cGAS-STING pathway. Thus, this study identifies a novel mechanism by which Mn exposure induces Tau aggregation, which in turn triggers potential neurotoxicity, providing a foundation for future drug target research.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Toxicology, School of
Public Health, Lanzhou University, Gansu 730000, China
| | - Jingjing Liu
- Department of Toxicology, School of
Public Health, Lanzhou University, Gansu 730000, China
| | - Shiyin Zhong
- Department of Toxicology, School of
Public Health, Lanzhou University, Gansu 730000, China
| | - Zhimin Zhang
- Department of Toxicology, School of
Public Health, Lanzhou University, Gansu 730000, China
| | - Qiongli Zhou
- Department of Toxicology, School of
Public Health, Lanzhou University, Gansu 730000, China
| | - Jirui Yang
- Department of Toxicology, School of
Public Health, Lanzhou University, Gansu 730000, China
| | - Xuhong Chang
- Department of Toxicology, School of
Public Health, Lanzhou University, Gansu 730000, China
| | - Hui Wang
- Department of Toxicology, School of
Public Health, Lanzhou University, Gansu 730000, China
| |
Collapse
|
28
|
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
|
29
|
Xu Y, Wang Q, Wang J, Qian C, Wang Y, Lu S, Song L, He Z, Liu W, Wan W. The cGAS-STING pathway activates transcription factor TFEB to stimulate lysosome biogenesis and pathogen clearance. Immunity 2025; 58:309-325.e6. [PMID: 39689715 DOI: 10.1016/j.immuni.2024.11.017] [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: 05/24/2024] [Revised: 09/27/2024] [Accepted: 11/14/2024] [Indexed: 12/19/2024]
Abstract
Induction of autophagy is an ancient function of the cyclic GMP-AMP (cGAMP) synthase (cGAS)-stimulator of interferon genes (STING) pathway through which autophagic cargoes are delivered to lysosomes for degradation. However, whether lysosome function is also modulated by the cGAS-STING pathway remains unknown. Here, we discovered that the cGAS-STING pathway upregulated lysosomal activity by stimulating lysosome biogenesis independently of the downstream protein kinase TANK-binding kinase 1 (TBK1). STING activation enhanced lysosome biogenesis through inducing the nuclear translocation of transcription factor EB (TFEB) as well as its paralogs transcription factor E3 (TFE3) and microphthalmia-associated transcription factor (MITF). STING-induced lipidation of GABA type A receptor-associated protein (GABARAP), an autophagy-related protein, on STING vesicles was responsible for TFEB activation. Membrane-bound GABARAP sequestered the GTPase-activating protein folliculin (FLCN) and FLCN-interacting protein (FNIP) complex to block its function toward the Rag GTPases Ras-related GTP-binding C and D (RagC and RagD), abolishing mechanistic target of rapamycin (mTOR) complex 1 (mTORC1)-dependent phosphorylation and inactivation of TFEB. Functionally, STING-induced lysosome biogenesis within cells facilitated the clearance of cytoplasmic DNA and invading pathogens. Thus, our findings reveal that induction of lysosome biogenesis is another important function of the cGAS-STING pathway.
Collapse
Affiliation(s)
- Yinfeng Xu
- Laboratory of Basic Biology, Hunan First Normal University, Changsha 410205, Hunan, China.
| | - Qian Wang
- Department of Thoracic Surgery of Sir Run Run Shaw Hospital, and Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Jun Wang
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Chuying Qian
- Department of Thoracic Surgery of Sir Run Run Shaw Hospital, and Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Yusha Wang
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Sheng Lu
- Department of Thoracic Surgery of Sir Run Run Shaw Hospital, and Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Lijiang Song
- Department of Thoracic Surgery of Sir Run Run Shaw Hospital, and Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Zhengfu He
- Department of Thoracic Surgery of Sir Run Run Shaw Hospital, and Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China
| | - Wei Liu
- Department of Metabolic Medicine, International Institutes of Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, Zhejiang, China.
| | - Wei Wan
- Department of Thoracic Surgery of Sir Run Run Shaw Hospital, and Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, Zhejiang, China.
| |
Collapse
|
30
|
Lee JS, Dittmar M, Miller J, Li M, Ayyanathan K, Ferretti M, Hulahan J, Whig K, Etwebi Z, Griesman T, Schultz DC, Cherry S. Pressure to evade cell-autonomous innate sensing reveals interplay between mitophagy, IFN signaling, and SARS-CoV-2 evolution. Cell Rep 2025; 44:115115. [PMID: 39708319 DOI: 10.1016/j.celrep.2024.115115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 11/07/2024] [Accepted: 12/05/2024] [Indexed: 12/23/2024] Open
Abstract
SARS-CoV-2 emerged, and continues to evolve, to efficiently infect humans worldwide. SARS-CoV-2 evades early innate recognition, interferon signaling occurring only in bystander cells. How the virus continues to evolve in the face of innate responses has important consequences, but the pathways involved are incompletely understood. Here, we find that autophagy genes regulate innate immune signaling, impacting the basal set point of interferons and, thus, permissivity to infection. Mechanistically, autophagy (mitophagy) genes negatively regulate MAVS, and this low basal level of MAVS is efficiently antagonized by SARS-CoV-2 ORF9b, blocking interferon activation in infected cells. However, loss of autophagy increased MAVS and overcomes ORF9b-mediated antagonism. This has driven the evolution of SARS-CoV-2 to express more ORF9b, allowing SARS-CoV-2 to replicate under conditions of increased MAVS signaling. Altogether, we find a critical role of mitophagy in the regulation of innate immunity and uncover an evolutionary trajectory of SARS-CoV-2 ORF9b to overcome host defenses.
Collapse
Affiliation(s)
- Jae Seung Lee
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mark Dittmar
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jesse Miller
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Minghua Li
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kasirajan Ayyanathan
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Max Ferretti
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jesse Hulahan
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kanupriya Whig
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zienab Etwebi
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Trevor Griesman
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David C Schultz
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sara Cherry
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Microbiology, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
31
|
Hyde VR, Zhou C, Fernandez JR, Chatterjee K, Ramakrishna P, Lin A, Fisher GW, Çeliker OT, Caldwell J, Bender O, Sauer PJ, Lugo-Martinez J, Bar DZ, D'Aiuto L, Shemesh OA. Anti-herpetic tau preserves neurons via the cGAS-STING-TBK1 pathway in Alzheimer's disease. Cell Rep 2025; 44:115109. [PMID: 39753133 DOI: 10.1016/j.celrep.2024.115109] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 08/06/2024] [Accepted: 12/03/2024] [Indexed: 02/01/2025] Open
Abstract
Alzheimer's disease (AD) diagnosis relies on the presence of extracellular β-amyloid (Aβ) and intracellular hyperphosphorylated tau (p-tau). Emerging evidence suggests a potential link between AD pathologies and infectious agents, with herpes simplex virus 1 (HSV-1) being a leading candidate. Our investigation, using metagenomics, mass spectrometry, western blotting, and decrowding expansion pathology, detects HSV-1-associated proteins in human brain samples. Expression of the herpesvirus protein ICP27 increases with AD severity and strongly colocalizes with p-tau but not with Aβ. Modeling in human brain organoids shows that HSV-1 infection elevates tau phosphorylation. Notably, p-tau reduces ICP27 expression and markedly decreases post-infection neuronal death from 64% to 7%. This modeling prompts investigation into the cGAS-STING-TBK1 pathway products, nuclear factor (NF)-κB and IRF-3, which colocalizes with ICP27 and p-tau in AD. Furthermore, experimental activation of STING enhances tau phosphorylation, while TBK1 inhibition prevents it. Together, these findings suggest that tau phosphorylation acts as an innate immune response in AD, driven by cGAS-STING.
Collapse
Affiliation(s)
- Vanesa R Hyde
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Chaoming Zhou
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Juan R Fernandez
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Krishnashis Chatterjee
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Pururav Ramakrishna
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Amanda Lin
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Gregory W Fisher
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Chemistry, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Orhan Tunç Çeliker
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jill Caldwell
- Department of Psychiatry, Western Psychiatric Institute and Clinic, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Omer Bender
- Department of Oral Biology, Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Peter Joseph Sauer
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Jose Lugo-Martinez
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Daniel Z Bar
- Department of Oral Biology, Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Leonardo D'Aiuto
- Department of Psychiatry, Western Psychiatric Institute and Clinic, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Or A Shemesh
- School of Pharmacy, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112102, Israel; Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.
| |
Collapse
|
32
|
Zhao P, Yin S, Qiu Y, Sun C, Yu H. Ferroptosis and pyroptosis are connected through autophagy: a new perspective of overcoming drug resistance. Mol Cancer 2025; 24:23. [PMID: 39825385 PMCID: PMC11740669 DOI: 10.1186/s12943-024-02217-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 12/25/2024] [Indexed: 01/20/2025] Open
Abstract
Drug resistance is a common challenge in clinical tumor treatment. A reduction in drug sensitivity of tumor cells is often accompanied by an increase in autophagy levels, leading to autophagy-related resistance. The effectiveness of combining chemotherapy drugs with autophagy inducers/inhibitors has been widely confirmed, but the mechanisms are still unclear. Ferroptosis and pyroptosis can be affected by various types of autophagy. Therefore, ferroptosis and pyroptosis have crosstalk via autophagy, potentially leading to a switch in cell death types under certain conditions. As two forms of inflammatory programmed cell death, ferroptosis and pyroptosis have different effects on inflammation, and the cGAS-STING signaling pathway is also involved. Therefore, it also plays an important role in the progression of some chronic inflammatory diseases. This review discusses the relationship between autophagy, ferroptosis and pyroptosis, and attempts to uncover the reasons behind the evasion of tumor cell death and the nature of drug resistance.
Collapse
Affiliation(s)
- Peng Zhao
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Shuangshuang Yin
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yuling Qiu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China.
| | - Changgang Sun
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, 261053, China.
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261041, China.
| | - Haiyang Yu
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
33
|
Cao Y, He X, Liu Z, Miao L, Zhu B. The potential of melatonin in sepsis-associated acute kidney injury: Mitochondrial protection and cGAS-STING signaling pathway. Heliyon 2025; 11:e41501. [PMID: 39850412 PMCID: PMC11755053 DOI: 10.1016/j.heliyon.2024.e41501] [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: 07/27/2024] [Revised: 12/15/2024] [Accepted: 12/24/2024] [Indexed: 01/25/2025] Open
Abstract
Melatonin (Mel) is known for various biological function, such as antioxidant and anti-inflammatory capabilities, as well as its ability to modulate immune responses, which can protect mitochondria and improve the prognosis of sepsis-associated acute kidney injury (SA-AKI). However, there is a multitude of theories regarding how Mel exerts its immune-modulating functions, with no consensus reached as of yet. We propose the protective effects of Mel on mitochondria are closely related to the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway in the immune-inflammatory response. We intraperitoneally injected H151 and Mel into SA-AKI mouse models to interfere the cGAS-STING signaling pathway. By comparing behavioral, pathological, and molecular biology results, we discovered that Mel could reduce cGAS-STING signaling pathway while greatly relieving kidney damage and function. In addition, Mel-treated mice showed a significant increase in autophagosome formations, which might be linked to the cGAS-STING signaling pathway. Our findings suggest that Mel protection on kidney injury in SA-AKI mice is partially attributed to the inhibition of the cGAS-STING signaling pathway.
Collapse
Affiliation(s)
- Yuchun Cao
- Department of Critical Care Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, Jiangsu, China
| | - Xiaofang He
- Department of Critical Care Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, Jiangsu, China
| | - Zeyuan Liu
- Department of Critical Care Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, Jiangsu, China
| | - Liying Miao
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, Jiangsu, China
| | - Bin Zhu
- Department of Critical Care Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, 213000, Jiangsu, China
| |
Collapse
|
34
|
Fischer TD, Bunker EN, Zhu PP, Le Guerroué F, Hadjian M, Dominguez-Martin E, Scavone F, Cohen R, Yao T, Wang Y, Werner A, Youle RJ. STING induces HOIP-mediated synthesis of M1 ubiquitin chains to stimulate NF-κB signaling. EMBO J 2025; 44:141-165. [PMID: 39578541 PMCID: PMC11696098 DOI: 10.1038/s44318-024-00291-2] [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: 10/24/2023] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 11/24/2024] Open
Abstract
STING activation by cyclic dinucleotides induces IRF3- and NF-κB-mediated gene expression in mammals, as well as lipidation of LC3B at Golgi-related membranes. While mechanisms of the IRF3 response are well understood, the mechanisms of NF-κB activation via STING remain unclear. We report here that STING activation induces linear/M1-linked ubiquitin chain (M1-Ub) formation and recruitment of the LUBAC E3 ligase, HOIP, to LC3B-associated Golgi membranes where ubiquitin is also localized. Loss of HOIP prevents formation of M1-Ub chains and reduces STING-induced NF-κB and IRF3 signaling in human THP1 monocytes and mouse bone marrow-derived macrophages, without affecting STING activation. STING-induced LC3B lipidation is not required for M1-Ub chain formation or for immune-related gene expression, but the recently reported STING function in neutralizing Golgi pH may be involved. Thus, LUBAC synthesis of M1-linked ubiquitin chains mediates STING-induced innate immune signaling.
Collapse
Affiliation(s)
- Tara D Fischer
- Biochemistry Section, Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| | - Eric N Bunker
- Biochemistry Section, Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Peng-Peng Zhu
- Biochemistry Section, Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - François Le Guerroué
- Biochemistry Section, Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- Single Cell Biomarkers UTechS, Institut Pasteur, Université Paris Cité, Paris, France
| | - Mahan Hadjian
- Biochemistry Section, Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Eunice Dominguez-Martin
- Biochemistry Section, Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Francesco Scavone
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Robert Cohen
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Tingting Yao
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Yan Wang
- Mass Spectrometry, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Achim Werner
- Stem Cell Biochemistry Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Richard J Youle
- Biochemistry Section, Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
35
|
Sharma R, Wu K, Han K, Russo AC, Dagur PK, Combs CA, Yao X, Levine SJ, Sack MN. BLOC1S1 Control of Vacuolar Organelle Fidelity Modulates Murine T H2 Cell Immunity and Allergy Susceptibility. Allergy 2024. [PMID: 39737471 DOI: 10.1111/all.16461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 11/15/2024] [Accepted: 12/09/2024] [Indexed: 01/01/2025]
Abstract
BACKGROUND The levels of biogenesis of lysosome organelles complex 1 subunit 1 (BLOC1S1) control mitochondrial and endolysosome organelle homeostasis and function. Reduced fidelity of these vacuolar organelles is increasingly being recognized as important in instigating cell-autonomous immune cell activation. We reasoned that exploring the role of BLOC1S1 in CD4+ T cells may further advance our understanding of regulatory events linked to mitochondrial and/or endolysosomal function in adaptive immunity. METHODS CD4+ T cells were analyzed from control and CD4+ T-cell-specific BLOC1S1 knockout mice. Polarization profiles were assayed using biochemical and molecular signatures, and signaling pathways were disrupted pharmacologically or via siRNA. Mouse models of airway and skin inflammation were generated by Ovalbumin and MC903 exposure, respectively. RESULTS TH2 regulator GATA3 and phosphorylated STAT6 were preferentially induced in BLOC1S1-depleted primary CD4+ T (TKO) cells. The levels of IL-4, IL-5, and IL-13 were markedly induced in the absence of BLOC1S1. At the organelle level, mitochondrial DNA leakage evoked cGAS-STING and NF-κB pathway activation with subsequent TH2 polarization. The induction of autophagy with rapamycin reduced cytosolic mtDNA and reversed these TH2 signatures. Furthermore, genetic knockdown of STING and NF-κB inhibition ameliorated this immune regulatory cascade in TKO cells. Finally, at a functional level, TKO mice displayed an increased susceptibility to allergic conditions, including dermatitis and allergic asthma. CONCLUSIONS BLOC1S1 depletion in mouse CD4+ T cells mediated disruption of mitochondrial integrity to initiate a predominant TH2-responsive phenotype via STING-NF-κB-driven signaling of the canonical TH2 regulatory program.
Collapse
Affiliation(s)
- Rahul Sharma
- Laboratory of Mitochondrial Biology and Metabolism, NHLBI, NIH, Bethesda, Maryland, USA
| | - Kaiyuan Wu
- Cardiovascular Branch, NHLBI, NIH, Bethesda, Maryland, USA
| | - Kim Han
- Laboratory of Mitochondrial Biology and Metabolism, NHLBI, NIH, Bethesda, Maryland, USA
| | - Anna Chiara Russo
- Laboratory of Mitochondrial Biology and Metabolism, NHLBI, NIH, Bethesda, Maryland, USA
| | | | | | - Xianglan Yao
- Critical Care Medicine and Pulmonary Branch, Bethesda, Maryland, USA
| | - Stewart J Levine
- Critical Care Medicine and Pulmonary Branch, Bethesda, Maryland, USA
| | - Michael N Sack
- Laboratory of Mitochondrial Biology and Metabolism, NHLBI, NIH, Bethesda, Maryland, USA
- Cardiovascular Branch, NHLBI, NIH, Bethesda, Maryland, USA
| |
Collapse
|
36
|
Wang H, Zuo Q, Li X, Liu Y, Gan L, Wang L, Rao Y, Pan R, Dong J. p62 Binding to Protein Kinase C Regulates HIV-1 gp120 V3 Loop Induced Microglial Inflammation. Inflammation 2024:10.1007/s10753-024-02229-6. [PMID: 39731677 DOI: 10.1007/s10753-024-02229-6] [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: 07/18/2024] [Revised: 11/12/2024] [Accepted: 12/22/2024] [Indexed: 12/30/2024]
Abstract
The main pathogenic mechanism of HIV-associated neurocognitive disorders (HAND) is neuronal apoptosis induced by inflammatory mediators, in which microglial inflammation plays a crucial role. However, the exact pathogenic mechanism remains unclear. Previous studies have shown that the HIV-1 gp120 V3 loop can trigger inflammation in CHME-5 microglia. p62 is a post-translational modified multidomain protein that is involved in the regulation of autophagy and is closely related to neuroinflammation. In this study, we found that p62 knockout down-regulated the expression of MCP-1, IL-6 and COX-2, and improved the inflammation of HIV-1 gp120 V3 loop induced microglia, while overexpression of p62 up-regulated the expression of MCP-1, IL-6 and COX-2, and promoted the inflammation of microglia. In addition, protein kinase C (PKC) knockout down-regulated the expression of MCP-1, IL-6 and COX-2 and inhibited the activation of IKK/ NF-κ B pathway, while tumor necrosis factor receptor-associated factor 6 (TRAF6) knockout had no significant effect on the expression of MCP-1, IL-6 and COX-2. Co-immunoprecipitation showed that p62 was bound and interacted with PKC. Inhibition of IKK/ NF-κ B pathway can down-regulate the expression of MCP-1, IL-6 and COX-2, and improve the inflammatory response of microglia. Our research further found that inhibition of IKK/ NF-κ B can decrease the expression of Caspase-3 and reduce the apoptosis of neurons in the co-culture of CHME-5 microglia and primary mouse neurons. The results of this study suggest that HIV-1 gp120 V3 loop induced CHME-5 microglial inflammation may be activated by the direct binding of p62 and PKC through the IKK/ NF-κ B signaling pathway, and these findings provide an important reference for the prevention and treatment of HAND.
Collapse
Affiliation(s)
- Huili Wang
- Department of Pathophysiology, Key Laboratory of the State Administration of Traditional Chinese Medicine, Medical College of Jinan University, Guangzhou, Guangdong Province, China
| | - Qin Zuo
- Department of Pathophysiology, Key Laboratory of the State Administration of Traditional Chinese Medicine, Medical College of Jinan University, Guangzhou, Guangdong Province, China
| | - Xinyi Li
- Department of Pathophysiology, Key Laboratory of the State Administration of Traditional Chinese Medicine, Medical College of Jinan University, Guangzhou, Guangdong Province, China
| | - Yuanyuan Liu
- Department of Pathophysiology, Key Laboratory of the State Administration of Traditional Chinese Medicine, Medical College of Jinan University, Guangzhou, Guangdong Province, China
| | - Limeng Gan
- Department of Pathophysiology, Key Laboratory of the State Administration of Traditional Chinese Medicine, Medical College of Jinan University, Guangzhou, Guangdong Province, China
| | - Linlin Wang
- Department of Pathophysiology, Key Laboratory of the State Administration of Traditional Chinese Medicine, Medical College of Jinan University, Guangzhou, Guangdong Province, China
| | - Yin Rao
- Department of Pathophysiology, Key Laboratory of the State Administration of Traditional Chinese Medicine, Medical College of Jinan University, Guangzhou, Guangdong Province, China
| | - Rui Pan
- Department of Orthopedics, The First Affiliated Hospital, Medical College of Jinan University, Guangzhou, Guangdong Province, China
| | - Jun Dong
- Department of Pathophysiology, Key Laboratory of the State Administration of Traditional Chinese Medicine, Medical College of Jinan University, Guangzhou, Guangdong Province, China.
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
37
|
Paludan SR, Pradeu T, Pichlmair A, Wray KB, Mikkelsen JG, Olagnier D, Mogensen TH. Early host defense against virus infections. Cell Rep 2024; 43:115070. [PMID: 39675007 DOI: 10.1016/j.celrep.2024.115070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/25/2024] [Accepted: 11/22/2024] [Indexed: 12/17/2024] Open
Abstract
Early host defense eliminates many viruses before infections are established while clearing others so they remain subclinical or cause only mild disease. The field of immunology has been shaped by broad concepts, including the pattern recognition theory that currently dominates innate immunology. Focusing on early host responses to virus infections, we analyze the literature to build a working hypothesis for the principles that govern the early line of cellular antiviral defense. Aiming to ultimately arrive at a criteria-based theory with strong explanatory power, we propose that both controlling infection and limiting inflammation are key drivers for the early cellular antiviral response. This response, which we suggest is exerted by a set of "microbe- and inflammation-restricting mechanisms," directly restrict viral replication while also counteracting inflammation. Exploring the mechanisms and physiological importance of the early layer of cellular antiviral defense may open further lines of research in immunology.
Collapse
Affiliation(s)
- Søren R Paludan
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark; Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Thomas Pradeu
- CNRS UMR 5164 ImmunoConcept, University of Bordeaux, Bordeaux, France; Department of Biological and Medical Sciences, University of Bordeaux, Bordeaux, France; Chapman University, Orange, CA, USA
| | - Andreas Pichlmair
- Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark; Technical University of Munich, School of Medicine, Institute of Virology, Munich, Germany; German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - K Brad Wray
- Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark; Centre for Science Studies, Aarhus University, Aarhus, Denmark; Aarhus Institute of Advanced Studies, Aarhus University, Aarhus, Denmark
| | - Jacob Giehm Mikkelsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark
| | - David Olagnier
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark
| | - Trine H Mogensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark; Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
38
|
Hamamoto K, Liang X, Ito A, Lanza M, Bui V, Zhang J, Opozda DM, Hattori T, Chen L, Haddock D, Imamura F, Wang HG, Takahashi Y. Unveiling the physiological impact of ESCRT-dependent autophagosome closure by targeting the VPS37A ubiquitin E2 variant-like domain. Cell Rep 2024; 43:115016. [PMID: 39607828 PMCID: PMC11748760 DOI: 10.1016/j.celrep.2024.115016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 09/05/2024] [Accepted: 11/11/2024] [Indexed: 11/30/2024] Open
Abstract
Macroautophagy (autophagy) involves the formation of phagophores that mature into autophagosomes. The impact of inhibiting autophagosome closure remains unclear. Here, we report the generation and analysis of mice with impaired autophagosome closure by targeting the ubiquitin E2 variant-like (UEVL) β strands of the endosomal sorting complex required for transport (ESCRT) I subunit VPS37A. The VPS37A UEVL mutation (Δ43-139) impairs bulk autophagic flux without disrupting ESCRT-I complex assembly and endosomal function. Homozygous mutant mice exhibit signs of autophagy impairment, including p62/SQSTM1 and ubiquitinated protein accumulation, neuronal dysfunction, growth retardation, antioxidant gene upregulation, and tissue abnormalities. However, about half of the mutant neonates survive to adulthood without severe liver injury. LC3 proximity proteomics reveals that the VPS37A UEVL mutation leads to active TANK-binding kinase 1 (TBK1) accumulation on phagophores, resulting in increased p62 phosphorylation and inclusion formation. These findings reveal a previously unappreciated role of LC3-conjugated phagophores in facilitating protein aggregation and sequestration, potentially alleviating proteotoxicity.
Collapse
Affiliation(s)
- Kouta Hamamoto
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Xinwen Liang
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Ayako Ito
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Matthew Lanza
- Department of Comparative Medicine, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Van Bui
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Jiawen Zhang
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - David M Opozda
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Tatsuya Hattori
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Longgui Chen
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - David Haddock
- Department of Pathology and Biochemistry, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Fumiaki Imamura
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Hong-Gang Wang
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | - Yoshinori Takahashi
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| |
Collapse
|
39
|
Gentili M, Carlson RJ, Liu B, Hellier Q, Andrews J, Qin Y, Blainey PC, Hacohen N. Classification and functional characterization of regulators of intracellular STING trafficking identified by genome-wide optical pooled screening. Cell Syst 2024; 15:1264-1277.e8. [PMID: 39657680 DOI: 10.1016/j.cels.2024.11.004] [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: 03/03/2024] [Revised: 08/05/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024]
Abstract
Stimulator of interferon genes (STING) traffics across intracellular compartments to trigger innate responses. Mutations in factors regulating this process lead to inflammatory disorders. To systematically identify factors involved in STING trafficking, we performed a genome-wide optical pooled screen (OPS). Based on the subcellular localization of STING in 45 million cells, we defined 464 clusters of gene perturbations based on their cellular phenotypes. A secondary, higher-dimensional OPS identified 73 finer clusters. We show that the loss of the gene of unknown function C19orf25, which clustered with USE1, a protein involved in Golgi-to-endoplasmic reticulum (ER) transport, enhances STING signaling. Additionally, HOPS deficiency delayed STING degradation and consequently increased signaling. Similarly, GARP/RIC1-RGP1 loss increased STING signaling by delaying STING Golgi exit. Our findings demonstrate that genome-wide genotype-phenotype maps based on high-content cell imaging outperform other screening approaches and provide a community resource for mining factors that impact STING trafficking and other cellular processes.
Collapse
Affiliation(s)
| | - Rebecca J Carlson
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Massachusetts Institute of Technology, Department of Health Sciences and Technology, Cambridge, MA, USA
| | - Bingxu Liu
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | - Yue Qin
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Paul C Blainey
- Massachusetts Institute of Technology, Department of Health Sciences and Technology, Cambridge, MA, USA; Massachusetts Institute of Technology, Department of Biological Engineering, Cambridge, MA, USA; Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA, USA.
| | - Nir Hacohen
- Massachusetts Institute of Technology, Department of Health Sciences and Technology, Cambridge, MA, USA; Massachusetts General Hospital, Krantz Family Center for Cancer Research, Boston, MA, USA.
| |
Collapse
|
40
|
Nishimura S, Linares JF, L'Hermitte A, Duran A, Cid-Diaz T, Martinez-Ordoñez A, Ruiz-Martinez M, Kudo Y, Marzio A, Heikenwalder M, Roberts LR, Diaz-Meco MT, Moscat J. Opposing regulation of the STING pathway in hepatic stellate cells by NBR1 and p62 determines the progression of hepatocellular carcinoma. Mol Cell 2024; 84:4660-4676.e10. [PMID: 39423823 PMCID: PMC12006816 DOI: 10.1016/j.molcel.2024.09.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 06/18/2024] [Accepted: 09/20/2024] [Indexed: 10/21/2024]
Abstract
Hepatocellular carcinoma (HCC) emerges from chronic inflammation, to which activation of hepatic stellate cells (HSCs) contributes by shaping a pro-tumorigenic microenvironment. Key to this process is p62, whose inactivation leads to enhanced hepatocarcinogenesis. Here, we show that p62 activates the interferon (IFN) cascade by promoting STING ubiquitination by tripartite motif protein 32 (TRIM32) in HSCs. p62, binding neighbor of BRCA1 gene 1 (NBR1) and STING, triggers the IFN cascade by displacing NBR1, which normally prevents the interaction of TRIM32 with STING and its subsequent activation. Furthermore, NBR1 also antagonizes STING by promoting its trafficking to the endosome-lysosomal compartment for degradation independent of autophagy. Of functional relevance, NBR1 deletion completely reverts the tumor-promoting function of p62-deficient HSCs by rescuing the inhibited STING-IFN pathway, thus enhancing anti-tumor responses mediated by CD8+ T cells. Therefore, NBR1 emerges as a synthetic vulnerability of p62 deficiency in HSCs by promoting the STING/IFN pathway, which boosts anti-tumor CD8+ T cell responses to restrain HCC progression.
Collapse
Affiliation(s)
- Sadaaki Nishimura
- Department of Pathology and Laboratory Medicine and Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Juan F Linares
- Department of Pathology and Laboratory Medicine and Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Antoine L'Hermitte
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Angeles Duran
- Department of Pathology and Laboratory Medicine and Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Tania Cid-Diaz
- Department of Pathology and Laboratory Medicine and Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Anxo Martinez-Ordoñez
- Department of Pathology and Laboratory Medicine and Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Marc Ruiz-Martinez
- Department of Pathology and Laboratory Medicine and Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Yotaro Kudo
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Antonio Marzio
- Department of Pathology and Laboratory Medicine and Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Mathias Heikenwalder
- German Cancer Research Center (DKFZ), Division of Chronic Inflammation and Cancer, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; University of Tuebingen, Faculty of Medicine, Institute for Interdisciplinary Research on Cancer Metabolism and Chronic Inflammation, M3-Research Center for Malignome, Metabolome and Microbiome, Otfried-Müller-Straße 37, 72076 Tübingen, Germany
| | - Lewis R Roberts
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Mayo Clinic Cancer Center, Rochester, MN 55905, USA
| | - Maria T Diaz-Meco
- Department of Pathology and Laboratory Medicine and Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA.
| | - Jorge Moscat
- Department of Pathology and Laboratory Medicine and Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
41
|
Soh LJ, Lee SY, Roebuck MM, Wong PF. Unravelling the interplay between ER stress, UPR and the cGAS-STING pathway: Implications for osteoarthritis pathogenesis and treatment strategy. Life Sci 2024; 357:123112. [PMID: 39378929 DOI: 10.1016/j.lfs.2024.123112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 09/20/2024] [Accepted: 10/03/2024] [Indexed: 10/10/2024]
Abstract
Osteoarthritis (OA) is a debilitating chronic degenerative disease affecting the whole joint organ leading to pain and disability. Cellular stress and injuries trigger inflammation and the onset of pathophysiological changes ensue after irreparable damage and inability to resolve inflammation, impeding the completion of the healing process. Extracellular matrix (ECM) degradation leads to dysregulated joint tissue metabolism. The reparative effort induces the proliferation of hypertrophic chondrocytes and matrix protein synthesis. Aberrant protein synthesis leads to endoplasmic reticulum (ER) stress and chondrocyte apoptosis with consequent cartilage matrix loss. These events in a vicious cycle perpetuate inflammation, hindering the restoration of normal tissue homeostasis. Recent evidence suggests that inflammatory responses and chondrocyte apoptosis could be caused by the activation of the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signalling axis in response to DNA damage. It has been reported that there is a crosstalk between ER stress and cGAS-STING signalling in cellular senescence and other diseases. Based on recent evidence, this review discusses the role of ER stress, Unfolded Protein Response (UPR) and cGAS-STING pathway in mediating inflammatory responses in OA.
Collapse
Affiliation(s)
- Li-Jen Soh
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Siam-Yee Lee
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Margaret M Roebuck
- Department of Musculoskeletal & Ageing Science, Institute of Life Course & Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK; Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool L3 9TA, UK
| | - Pooi-Fong Wong
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia.
| |
Collapse
|
42
|
Kong L, Xu P, Shen N, Li W, Li R, Tao C, Wang G, Zhang Y, Sun W, Hu W, Liu X. STING orchestrates microglia polarization via interaction with LC3 in autophagy after ischemia. Cell Death Dis 2024; 15:824. [PMID: 39537618 PMCID: PMC11560960 DOI: 10.1038/s41419-024-07208-1] [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: 03/24/2024] [Revised: 10/25/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
Autophagy has both protective and pathogenetic effects on injury caused by cerebral ischemia/reperfusion (I/R). Our previous research has indicated that stimulator of interferon genes (STING) could orchestrate microglia polarization following middle cerebral artery occlusion. However, it remains largely unexplored whether STING balances microglial polarization by regulating autophagy in brain I/R injury. Here, STING was observed to show an up-regulation in the microglia from mice subjected to experimental ischemic stroke. Strikingly, the deletion of STING led to the significant skewness of microglia activated by ischemia from a pro- to anti-inflammatory state and substantially alleviated ischemia-induced infarction and neuronal injury. In addition, STING-null mice can restore long-term neurobehavioral function. Then, the crosstalk between neuroinflammation and microglia autophagy was analyzed. The differential activity of autophagy in wild-type and STING-knockout (KO) mice or primary microglia was largely reversed when STING was restored in microglia. Irritating autophagy by rapamycin skewed the anti‑inflammatory state induced by STING-KO to a pro‑inflammatory state in microglia. Furthermore, microtubule-associated protein light-chain-3 (LC3) was identified as the key factor in the STING regulation of autophagy by glutathione-S-transferase (GST) pull-down analysis. Mechanically, STING can directly interact with LC3 through the STING transmembrane domain (1-139aa). Herein, current data determine the pivotal role of autophagy, specifically via LC3 protein, in the regulation of microglial phenotypic transformation by STING. These findings may provide a possible treatment target for delaying the progression of ischemic stroke.
Collapse
Affiliation(s)
- Lingqi Kong
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Pengfei Xu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
| | - Nan Shen
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Wenyu Li
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Rui Li
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Chunrong Tao
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Guoping Wang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Yan Zhang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Wen Sun
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Wei Hu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
| | - Xinfeng Liu
- Department of Neurology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
| |
Collapse
|
43
|
Zhang Y, Zou M, Wu H, Zhu J, Jin T. The cGAS-STING pathway drives neuroinflammation and neurodegeneration via cellular and molecular mechanisms in neurodegenerative diseases. Neurobiol Dis 2024; 202:106710. [PMID: 39490400 DOI: 10.1016/j.nbd.2024.106710] [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: 07/03/2024] [Revised: 09/27/2024] [Accepted: 10/18/2024] [Indexed: 11/05/2024] Open
Abstract
Neurodegenerative diseases (NDs) are a type of common chronic progressive disorders characterized by progressive damage to specific cell populations in the nervous system, ultimately leading to disability or death. Effective treatments for these diseases are still lacking, due to a limited understanding of their pathogeneses, which involve multiple cellular and molecular pathways. The triggering of an immune response is a common feature in neurodegenerative disorders. A critical challenge is the intricate interplay between neuroinflammation, neurodegeneration, and immune responses, which are not yet fully characterized. In recent years, the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon gene (STING) pathway, a crucial immune response for intracellular DNA sensing, has gradually gained attention. However, the specific roles of this pathway within cellular types such as immune cells, glial and neuronal cells, and its contribution to ND pathogenesis, remain not fully elucidated. In this review, we systematically explore how the cGAS-STING signaling links various cell types with related cellular effector pathways under the context of NDs for multifaceted therapeutic directions. We emphasize the discovery of condition-dependent cellular heterogeneity in the cGAS-STING pathway, which is integral for understanding the diverse cellular responses and potential therapeutic targets. Additionally, we review the pathogenic role of cGAS-STING activation in Parkinson's disease, ataxia-telangiectasia, and amyotrophic lateral sclerosis. We focus on the complex bidirectional roles of the cGAS-STING pathway in Alzheimer's disease, Huntington's disease, and multiple sclerosis, revealing their double-edged nature in disease progression. The objective of this review is to elucidate the pivotal role of the cGAS-STING pathway in ND pathogenesis and catalyze new insights for facilitating the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Yuxin Zhang
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Meijuan Zou
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Hao Wu
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jie Zhu
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China; Department of Neurobiology, Care Sciences & Society, Karolinska Institute, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Tao Jin
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
44
|
Yin J, Lai P, Zhu L, Ma J. Angiopoietin 1 Relieves Osteolysis by Promoting Macrophage Mitophagy Through the TBK1-SQSTM1 Pathway to Inhibit AIM2 Inflammasome-Mediated Pyroptosis. Appl Biochem Biotechnol 2024; 196:7908-7927. [PMID: 38662322 DOI: 10.1007/s12010-024-04961-z] [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: 04/16/2024] [Indexed: 04/26/2024]
Abstract
Osteolysis resulting from wear particles and subsequent aseptic loosening is a leading cause of revision surgery of artificial joints. The underlying pathogenesis of particle-induced osteolysis (PPO) has remained largely uncertain. Addressing how to mitigate osteolysis caused by wear particles presents a significant challenge for orthopedic surgeons. This study aimed to explore the molecular mechanism by which Angiopoietin (Ang-1) inhibits osteoclast activation to alleviate osteolysis. RAW264.7 mouse macrophages were stimulated with LPS or RANKL to induce osteoclast formation. Additionally, titanium (Ti) particles (50 mg) were subperiosteally implanted around the cranial suture of mice to establish a calvarial osteolysis model. Ang-1, a member of the pro-angiogenic factor protein family and an important inflammatory regulator molecule, was utilized in this model. TRAP staining was utilized to detect osteoclast activation, while a western blot was conducted to identify key proteins associated with mitophagy and pyroptosis. Scanning electron microscopy was employed to observe the morphology and dimensions of Ti particles. Additionally, a combination of micro-CT, H&E, Masson's trichrome, and immunohistochemical staining techniques were applied to analyze the calvarial samples. Results indicated that Ang-1 could inhibit LPS- or RANKL-induced osteoclastogenesis and alleviate Ti particle-induced calvarial osteolysis in mice. TBK-1, a key signaling molecule involved in initiating mitophagy, was found to be mechanistically enhanced by Ang-1 through promoting TBK-1 phosphorylation in macrophages. This process inhibited AIM2 inflammasome-mediated pyroptosis and impeded osteoclastogenesis. Overall, this research uncovers a novel mechanism by which Ang-1 can attenuate inflammatory osteolysis, potentially offering a new therapeutic approach for PPO.
Collapse
Affiliation(s)
- Jian Yin
- Department of Orthopedics, Shanghai General Hospital of Nanjing Medical University, Songjiang, Shanghai, 201600, China
- Department of Orthopedics, the Affiliated Jiangning Hospital With Nanjing Medical University, Nanjing, 211100, China
| | - Peng Lai
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Songjiang, Shanghai, 201600, China
| | - Libo Zhu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Songjiang, Shanghai, 201600, China
| | - Jinzhong Ma
- Department of Orthopedics, Shanghai General Hospital of Nanjing Medical University, Songjiang, Shanghai, 201600, China.
- Department of Orthopedics, the Affiliated Jiangning Hospital With Nanjing Medical University, Nanjing, 211100, China.
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Songjiang, Shanghai, 201600, China.
| |
Collapse
|
45
|
Jiang H, Guo Y, Wang Q, Wang Y, Peng D, Fang Y, Yan L, Ruan Z, Zhang S, Zhao Y, Zhang W, Shang W, Feng Z. The dysfunction of complement and coagulation in diseases: the implications for the therapeutic interventions. MedComm (Beijing) 2024; 5:e785. [PMID: 39445002 PMCID: PMC11496570 DOI: 10.1002/mco2.785] [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: 03/31/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024] Open
Abstract
The complement system, comprising over 30 proteins, is integral to the immune system, and the coagulation system is critical for vascular homeostasis. The activation of the complement and coagulation systems involves an organized proteolytic cascade, and the overactivation of these systems is a central pathogenic mechanism in several diseases. This review describes the role of complement and coagulation system activation in critical illness, particularly sepsis. The complexities of sepsis reveal significant knowledge gaps that can be compared to a profound abyss, highlighting the urgent need for further investigation and exploration. It is well recognized that the inflammatory network, coagulation, and complement systems are integral mechanisms through which multiple factors contribute to increased susceptibility to infection and may result in a disordered immune response during septic events in patients. Given the overlapping pathogenic mechanisms in sepsis, immunomodulatory therapies currently under development may be particularly beneficial for patients with sepsis who have concurrent infections. Herein, we present recent findings regarding the molecular relationships between the coagulation and complement pathways in the advancement of sepsis, and propose potential intervention targets related to the crosstalk between coagulation and complement, aiming to provide more valuable treatment of sepsis.
Collapse
Affiliation(s)
- Honghong Jiang
- Faculty of Pediatrics, the Seventh Medical Center of Chinese PLA General HospitalNational Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing Key Laboratory of Pediatric Organ FailureBeijingChina
| | - Yiming Guo
- Department of Biological Science, The Dietrich School of Arts and SciencesUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Qihang Wang
- Department of Obstetrics and GynecologyThe Seventh Medical Center of Chinese PLA General HospitalBeijingChina
| | - Yiran Wang
- Department of Obstetrics and GynecologyThe sixth Medical Center of Chinese PLA General HospitalBeijingChina
| | - Dingchuan Peng
- School of MedicineSouth China University of TechnologyGuangzhouChina
| | - Yigong Fang
- Institute of Acupuncture and MoxibustionChina Academy of Chinese Medical SciencesBeijingChina
| | - Lei Yan
- Faculty of Pediatrics, the Seventh Medical Center of Chinese PLA General HospitalNational Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing Key Laboratory of Pediatric Organ FailureBeijingChina
| | - Zhuolin Ruan
- Department of Obstetrics and Gynecology,Chinese PLA General HospitalBeijingChina
| | - Sheng Zhang
- Faculty of Pediatrics, the Seventh Medical Center of Chinese PLA General HospitalNational Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing Key Laboratory of Pediatric Organ FailureBeijingChina
| | - Yong Zhao
- Department of Obstetrics and GynecologyThe Seventh Medical Center of Chinese PLA General HospitalBeijingChina
| | - Wendan Zhang
- Faculty of Pediatrics, the Seventh Medical Center of Chinese PLA General HospitalNational Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing Key Laboratory of Pediatric Organ FailureBeijingChina
| | - Wei Shang
- Faculty of Pediatrics, the Seventh Medical Center of Chinese PLA General HospitalNational Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing Key Laboratory of Pediatric Organ FailureBeijingChina
- Department of Obstetrics and GynecologyThe Seventh Medical Center of Chinese PLA General HospitalBeijingChina
| | - Zhichun Feng
- Faculty of Pediatrics, the Seventh Medical Center of Chinese PLA General HospitalNational Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing Key Laboratory of Pediatric Organ FailureBeijingChina
| |
Collapse
|
46
|
Kim HG, Kim JH, Yu T, Cho JY. Functional Involvement of TANK-Binding Kinase 1 in the MyD88-Dependent NF- κB Pathway Through Syk. Mediators Inflamm 2024; 2024:8634515. [PMID: 39493293 PMCID: PMC11531359 DOI: 10.1155/2024/8634515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 10/03/2024] [Accepted: 10/07/2024] [Indexed: 11/05/2024] Open
Abstract
Inflammation is a vital immune defense mechanism regulated by Toll-like receptors (TLRs) and the nuclear factor-kappa B (NF-κB) pathway. TANK-binding kinase 1 (TBK1) is central to immunity and inflammation and influences antiviral responses and cellular processes. However, the precise role of TBK1 in modulating the NF-κB pathway through interactions with other proteins, such as spleen tyrosine kinase (Syk), remains poorly understood. As dysregulation of TBK1 and NF-κB can lead to a variety of diseases, they are important therapeutic targets. In this work, inflammatory processes involving the TBK1-Syk-NF-κB pathway were elucidated using lipopolysaccharide (LPS)-induced macrophages; human embryonic kidney 293 (HEK293) cells overexpressing MyD88, TBK1, and Syk proteins and their mutants; and real-time polymerase chain reaction (PCR), immunoblotting analyses, and kinase assays. TBK1 was activated in LPS-, poly I:C-, and Pam3CSK-stimulated macrophages. Transcript levels of TNF, NOS2, and IL1B were increased in cells overexpressing TBK1 but not in cells overexpressing TBK1 K38A. The transcription of TNF, NOS2, and IL1B and NF-κB luciferase activity were inhibited by silencing TBK1 in LPS-stimulated RAW264.7 cells and MyD88-transfected HEK293 cells. Syk was the key mediator of the TBK1-dependent NF-κB pathway and bound directly to the coiled coil domain of TBK1, which was necessary to activate Syk and the Syk-p85 pathway. This research advances the understanding of the role of TBK1 in NF-κB signaling, emphasizing Syk as a key mediator. The interaction between TBK1 and Syk has potential for precise immune modulation that can be applied to treat immune-related disorders.
Collapse
Affiliation(s)
- Han Gyung Kim
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Ji Hye Kim
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Tao Yu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Jae Youl Cho
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
47
|
Xu T, Zhong X, Luo N, Ma W, Hao P. Review of Excessive Cytosolic DNA and Its Role in AIM2 and cGAS-STING Mediated Psoriasis Development. Clin Cosmet Investig Dermatol 2024; 17:2345-2357. [PMID: 39464745 PMCID: PMC11512523 DOI: 10.2147/ccid.s476785] [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: 05/27/2024] [Accepted: 10/07/2024] [Indexed: 10/29/2024]
Abstract
In psoriasis, keratinocytes are triggered by factors, such as infection or tissue damage, to release DNA, which thereby activates plasmacytoid dendritic cells and macrophages to induce inflammation, thickened epidermis, and parakeratosis. The recognition of double-stranded (ds)DNA facilitates the activation of cytoplasmic DNA sensors absent in melanoma 2 (AIM2) inflammasome assembly and cyclic guanosine monophosphate adenosine monophosphate (cGAMP) synthase (cGAS) - stimulator of interferon gene (STING) pathway, both of which play a pivotal role in mediating the inflammatory response and driving the progression of psoriasis. Additionally, secreted proinflammatory cytokines can stimulate further DNA release from keratinocytes. Notably, the activation of AIM2 and cGAS-STING signaling pathways also mediates programmed cell death, potentially enhancing DNA overproduction. As a result, excessive DNA can activate these pathways, amplifying persistent inflammatory responses that contribute to the maintenance of psoriasis. Several studies have validated that targeting DNA and its mediated activation of AIM2 and cGAS-STING offers promising therapeutic strategies for psoriasis. Here, we postulate a hypothesis that excessive cytosolic DNA can activate AIM2 and cGAS-STING, mediating inflammation and programmed cell death, ultimately fostering DNA accumulation and contributing to the development of psoriasis.
Collapse
Affiliation(s)
- Tongtong Xu
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Xiaojing Zhong
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Nana Luo
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Wenyi Ma
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Pingsheng Hao
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| |
Collapse
|
48
|
Luo R, Wang T, Lan J, Lu Z, Chen S, Sun Y, Qiu HJ. The multifaceted roles of selective autophagy receptors in viral infections. J Virol 2024; 98:e0081424. [PMID: 39212450 PMCID: PMC11494948 DOI: 10.1128/jvi.00814-24] [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] [Indexed: 09/04/2024] Open
Abstract
Selective autophagy is a protein clearance mechanism mediated by evolutionarily conserved selective autophagy receptors (SARs), which specifically degrades misfolded, misassembled, or metabolically regulated proteins. SARs help the host to suppress viral infections by degrading viral proteins. However, viruses have evolved sophisticated mechanisms to counteract, evade, or co-opt autophagic processes, thereby facilitating viral replication. Therefore, this review aims to summarize the complex mechanisms of SARs involved in viral infections, specifically focusing on how viruses exploit strategies to regulate selective autophagy. We present an updated understanding of the various critical roles of SARs in viral pathogenesis. Furthermore, newly discovered evasion strategies employed by viruses are discussed and the ubiquitination-autophagy-innate immune regulatory axis is proposed to be a crucial pathway to control viral infections. This review highlights the remarkable flexibility and plasticity of SARs in viral infections.
Collapse
Affiliation(s)
- Rui Luo
- State Key Laboratory for Animal Disease Control and Prevention, National African Swine Fever Para-Reference Laboratory, National High Containment Facilities for Animal Diseases Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Tao Wang
- State Key Laboratory for Animal Disease Control and Prevention, National African Swine Fever Para-Reference Laboratory, National High Containment Facilities for Animal Diseases Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Jing Lan
- State Key Laboratory for Animal Disease Control and Prevention, National African Swine Fever Para-Reference Laboratory, National High Containment Facilities for Animal Diseases Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
- College of Animal Sciences, Yangtze University, Jingzhou, China
| | - Zhanhao Lu
- State Key Laboratory for Animal Disease Control and Prevention, National African Swine Fever Para-Reference Laboratory, National High Containment Facilities for Animal Diseases Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Shengmei Chen
- State Key Laboratory for Animal Disease Control and Prevention, National African Swine Fever Para-Reference Laboratory, National High Containment Facilities for Animal Diseases Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
- School of Life Science Engineering, Foshan University, Foshan, China
| | - Yuan Sun
- State Key Laboratory for Animal Disease Control and Prevention, National African Swine Fever Para-Reference Laboratory, National High Containment Facilities for Animal Diseases Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Hua-Ji Qiu
- State Key Laboratory for Animal Disease Control and Prevention, National African Swine Fever Para-Reference Laboratory, National High Containment Facilities for Animal Diseases Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
- College of Animal Sciences, Yangtze University, Jingzhou, China
- School of Life Science Engineering, Foshan University, Foshan, China
| |
Collapse
|
49
|
Islam S, Islam MM, Akhand MRN, Park BY, Akanda MR. Recent advancements in cGAS-STING activation, tumor immune evasion, and therapeutic implications. Med Oncol 2024; 41:291. [PMID: 39419913 DOI: 10.1007/s12032-024-02539-7] [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: 08/15/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024]
Abstract
The cGAS-STING signaling pathway is indeed a pivotal component of the immune system and serve as a crucial link between innate and adaptive immune responses. STING is involved in the cellular response to pathogen invasion and DNA damage, and which has important consequences for host defense mechanisms and cancer regulation. Ongoing research aiming to modulate the cGAS-STING pathway for improved clinical outcomes in cancer and autoimmune diseases is underway. Indeed, the interaction between the cGAS-STING pathway and immune evasion mechanisms is a complex and critical aspect of cancer biology. Pathogens and various host factors can exploit this pathway to reduce the effectiveness of cancer therapies, particularly immunotherapies. Thus, immunotherapies or combination therapies may assist in overcoming the immune suppression and improving clinical outcomes. This review explores recent advancements in understanding the cGAS-STING signaling pathway, with particular emphasis on its activation mechanisms and role in tumor immune evasion. The dual role of the pathway in boosting immune responses while simultaneously enabling tumors to evade the immune system makes it a crucial target for innovative cancer treatment approaches.Please confirm if the author names are presented accurately and in the correct sequence (given name, middle name/initial, family name). Author 2 Given name: [Md Mazedul] Last name [Islam], Author 3 Given name: [Mst Rubaiat Nazneen] Last name [Akhand] and Author 5 Given name: [Md Rashedunnabi] Last name [Akanda]. Also, kindly confirm the details in the metadata are correct.AQ1: Here Author 4 given name: [Byung-Yong] Last name [Park] is missing. Metadata are correct.
Collapse
Affiliation(s)
- Saiful Islam
- Department of Physiology, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | - Md Mazedul Islam
- Faculty of Veterinary, Animal and Biomedical Sciences, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | | | - Byung-Yong Park
- Institute of Animal Transplantation, College of Veterinary Medicine, Jeonbuk National University, Iksan, 54596, South Korea
| | - Md Rashedunnabi Akanda
- Department of Pharmacology and Toxicology, Sylhet Agricultural University, Sylhet, 3100, Bangladesh.
| |
Collapse
|
50
|
Kumari D, Kaur S, Dandekar MP. Intricate Role of the Cyclic Guanosine Monophosphate Adenosine Monophosphate Synthase-Stimulator of Interferon Genes (cGAS-STING) Pathway in Traumatic Brain Injury-Generated Neuroinflammation and Neuronal Death. ACS Pharmacol Transl Sci 2024; 7:2936-2950. [PMID: 39416963 PMCID: PMC11475349 DOI: 10.1021/acsptsci.4c00310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 10/19/2024]
Abstract
The secondary insult in the aftermath of traumatic brain injury (TBI) causes detrimental and self-perpetuating alteration in cells, resulting in aberrant function and the death of neuronal cells. The secondary insult is mainly driven by activation of the neuroinflammatory pathway. Among several classical pathways, the cGAS-STING pathway, a primary neuroinflammatory route, encompasses the cyclic GMP-AMP synthase (cGAS), stimulator of interferon genes (STING), and downstream signaling adaptor. Recently, the cGAS-STING research domain has gained exponential attention. The aberrant stimulation of cGAS-STING machinery and corresponding neuroinflammation have also been reported after TBI. In addition to the critical contribution to neuroinflammation, the cGAS-STING signaling also provokes neuronal cell death through various cell death mechanisms. This review highlights the structural and molecular mechanisms of the cGAS-STING machinery associated with TBI. We also focus on the intricate relationship and framework between cGAS-STING signaling and cell death mechanisms (autophagy, apoptosis, pyroptosis, ferroptosis, and necroptosis) in the aftermath of TBI. We suggest that the targeting of cGAS-STING signaling may open new therapeutic strategies to combat neuroinflammation and neurodegeneration in TBI.
Collapse
Affiliation(s)
- Deepali Kumari
- Department of Biological
Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana 500037, India
| | - Simranjit Kaur
- Department of Biological
Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana 500037, India
| | - Manoj P. Dandekar
- Department of Biological
Sciences (Pharmacology and Toxicology), National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana 500037, India
| |
Collapse
|