1
|
Fan JJ, Hu C, Hu M, Dong WS, Li K, Ye YJ, Zhang X. A brief overview of the E3 ubiquitin ligase: TRIM7. Cell Signal 2025:111886. [PMID: 40419231 DOI: 10.1016/j.cellsig.2025.111886] [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: 04/13/2025] [Revised: 05/17/2025] [Accepted: 05/19/2025] [Indexed: 05/28/2025]
Abstract
TRIM7, a member of the E3 ubiquitin ligase family, has garnered significant attentions in different research fields since its discovery. This enzyme plays indispensable roles in various pathophysiological processes through ubiquitination-mediated degradation of diverse protein substrates. This review systematically summarizes the current knowledge on the protein structure and biological functions of TRIM7. Structurally, TRIM7 features a conserved RBCC motif (RING, B-box, and coiled-coil domains) coupled with a variable C-terminal region that dictates the substrate specificity. In infectious contexts, TRIM7 is required for the pathogen-specific regulation, and exerts paradoxical effects by either promoting host defense or facilitating viral pathogenesis depending on pathogen type. Within oncology, TRIM7 manifests tumor-suppressive properties through regulating metastasis, apoptosis, and tumor immunology. In addition, it might serve as a reliable biomarker for monitoring the progression of idiopathic pulmonary fibrosis and also inhibits the progression of atherosclerosis. In summary, TRIM7 plays critical roles in different pathophysiological processes, and it might be a predictive and therapeutic target in certain human diseases.
Collapse
Affiliation(s)
- Jun-Jie Fan
- Department of Geriatrics, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Can Hu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Research Center for Medical Imaging in Hubei Province, Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Min Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Wen-Sheng Dong
- Department of Geriatrics, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Kang Li
- Department of Geriatrics, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Yun-Jia Ye
- Department of Geriatrics, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Xin Zhang
- Department of Geriatrics, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China.
| |
Collapse
|
2
|
Thanos JM, Campbell OC, Cowan MN, Bruch KR, Moore KA, Ennerfelt HE, Natale NR, Mangalmurti A, Kerur N, Lukens JR. STING deletion protects against amyloid β-induced Alzheimer's disease pathogenesis. Alzheimers Dement 2025; 21:e70305. [PMID: 40410932 PMCID: PMC12101966 DOI: 10.1002/alz.70305] [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: 02/05/2025] [Revised: 04/10/2025] [Accepted: 04/28/2025] [Indexed: 05/26/2025]
Abstract
INTRODUCTION While immune dysfunction has been increasingly linked to Alzheimer's disease (AD) progression, many major innate immune signaling molecules have yet to be explored in AD pathogenesis using genetic targeting approaches. METHODS To investigate a role for the key innate immune adaptor molecule, stimulator of interferon genes (STING), in AD, we deleted Sting1 in the 5xFAD mouse model of AD-related amyloidosis and evaluated the effects on pathology, neuroinflammation, gene expression, and cognition. RESULTS Genetic ablation of STING in 5xFAD mice led to improved control of amyloid beta (Aβ) plaques, alterations in microglial activation status, decreased levels of neuritic dystrophy, and protection against cognitive decline. Moreover, rescue of neurological disease in STING-deficient 5xFAD mice was characterized by reduced expression of type I interferon signaling genes in both microglia and excitatory neurons. DISCUSSION These findings reveal critical roles for STING in Aβ-driven neurological disease and suggest that STING-targeting therapeutics may offer promising strategies to treat AD. HIGHLIGHTS Stimulator of interferon genes (STING) deficiency in the 5xFAD mouse model of Alzheimer's disease-related amyloidosis results in decreased amyloid beta (Aβ) deposition and altered microglial activation status. Protection against amyloidosis in STING-deficient 5xFAD mice is associated with decreased expression of genes involved in type I IFN signaling, improved neuronal health, and reduced levels of oxidative stress. Loss of STING in 5xFAD mice leads to improved spatial learning and memory.
Collapse
Affiliation(s)
- Jessica M. Thanos
- Department of Neuroscience, Center for Brain Immunology and Glia (BIG)University of VirginiaCharlottesvilleVirginiaUSA
- Neuroscience Graduate ProgramUniversity of VirginiaCharlottesvilleVirginiaUSA
- Brain Immunology and Glia Graduate Training ProgramUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Olivia C. Campbell
- Department of Neuroscience, Center for Brain Immunology and Glia (BIG)University of VirginiaCharlottesvilleVirginiaUSA
| | - Maureen N. Cowan
- Department of Neuroscience, Center for Brain Immunology and Glia (BIG)University of VirginiaCharlottesvilleVirginiaUSA
| | - Katherine R. Bruch
- Department of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Katelyn A. Moore
- Department of Neuroscience, Center for Brain Immunology and Glia (BIG)University of VirginiaCharlottesvilleVirginiaUSA
| | - Hannah E. Ennerfelt
- Department of Neurology and Neurological SciencesStanford UniversityPalo AltoCaliforniaUSA
| | - Nick R. Natale
- Department of Neuroscience, Center for Brain Immunology and Glia (BIG)University of VirginiaCharlottesvilleVirginiaUSA
- Neuroscience Graduate ProgramUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Aman Mangalmurti
- Department of Neuroscience, Center for Brain Immunology and Glia (BIG)University of VirginiaCharlottesvilleVirginiaUSA
- Neuroscience Graduate ProgramUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Nagaraj Kerur
- Department of Ophthalmology and Visual Sciencesthe Ohio State University Wexner Medical CenterColumbusOhioUSA
- Ohio State Havener Eye Institutethe Ohio State University Wexner Medical CenterColumbusOhioUSA
- Department of Microbial Infection and Immunitythe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - John R. Lukens
- Department of Neuroscience, Center for Brain Immunology and Glia (BIG)University of VirginiaCharlottesvilleVirginiaUSA
- Neuroscience Graduate ProgramUniversity of VirginiaCharlottesvilleVirginiaUSA
- Brain Immunology and Glia Graduate Training ProgramUniversity of VirginiaCharlottesvilleVirginiaUSA
| |
Collapse
|
3
|
Herbst M, Köksal H, Brunn S, Zanetti D, Domocos I, De Stefani V, Gatti M, Vivalda F, Pereira P, Nater M, Cecconi V, Sartori AA, van den Broek M. Cancer-cell-derived cGAMP limits the activity of tumor-associated CD8 + T cells. Cell Rep 2025; 44:115510. [PMID: 40178978 DOI: 10.1016/j.celrep.2025.115510] [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: 11/05/2024] [Revised: 02/19/2025] [Accepted: 03/12/2025] [Indexed: 04/05/2025] Open
Abstract
Using a mouse tumor model with inducible cancer-cell-intrinsic cyclic GMP-AMP (cGAMP) synthase (cGAS) expression, we show that cancer-cell-derived cGAMP is essential and sufficient to trigger a sustained type I interferon response within the tumor microenvironment. This leads to improved CD8+ T cell-dependent tumor restriction. However, cGAMP limits the proliferation, survival, and function of stimulator of IFN genes (STING)-expressing, but not of STING-deficient, CD8+ T cells. In vivo, STING deficiency in CD8+ T cells enhances tumor restriction. Consequently, cancer-cell-derived cGAMP both drives and limits the anti-tumor potential of CD8+ T cells. Mechanistically, T cell-intrinsic STING is associated with pro-apoptotic and antiproliferative gene signatures. Our findings suggest that STING signaling acts as a checkpoint in CD8+ T cells that balances tumor immunity.
Collapse
Affiliation(s)
- Michael Herbst
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Hakan Köksal
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Silvan Brunn
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Dominik Zanetti
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Ioana Domocos
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Viola De Stefani
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Marco Gatti
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Francesca Vivalda
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Paulo Pereira
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Marc Nater
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Virginia Cecconi
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Alessandro A Sartori
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Maries van den Broek
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
4
|
Wu S, Bu X, Chen D, Wu X, Wu H, Caiyin Q, Qiao J. Molecules-mediated bidirectional interactions between microbes and human cells. NPJ Biofilms Microbiomes 2025; 11:38. [PMID: 40038292 PMCID: PMC11880406 DOI: 10.1038/s41522-025-00657-2] [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: 10/06/2024] [Accepted: 01/22/2025] [Indexed: 03/06/2025] Open
Abstract
Complex molecules-mediated interactions, which are based on the bidirectional information exchange between microbes and human cells, enable the defense against diseases and health maintenance. Recently, diverse single-direction interactions based on active metabolites, immunity factors, and quorum sensing signals have largely been summarized separately. In this review, according to a simplified timeline, we proposed the framework of Molecules-mediated Bidirectional Interactions (MBI) between microbe and humans to decipher and understand their intricate interactions systematically. About the microbe-derived interactions, we summarized various molecules, such as short-chain fatty acids, bile acids, tryptophan catabolites, and quorum sensing molecules, and their corresponding human receptors. Concerning the human-derived interactions, we reviewed the effect of human molecules, including hormones, cytokines, and other circulatory metabolites on microbial characteristics and phenotypes. Finally, we discussed the challenges and trends for developing and deciphering molecule-mediated bidirectional interactions and their potential applications in the guard of human health.
Collapse
Affiliation(s)
- Shengbo Wu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China
- Zhejiang Institute of Tianjin University, Shaoxing, Shaoxing, 312300, Zhejiang, China
| | - Xueying Bu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China
| | - Danlei Chen
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China
- Zhejiang Institute of Tianjin University, Shaoxing, Shaoxing, 312300, Zhejiang, China
| | - Xueyan Wu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China
| | - Hao Wu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China.
- Zhejiang Institute of Tianjin University, Shaoxing, Shaoxing, 312300, Zhejiang, China.
| | - Qinggele Caiyin
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China.
- Zhejiang Institute of Tianjin University, Shaoxing, Shaoxing, 312300, Zhejiang, China.
- Key Laboratory of Systems Bioengineering, Ministry of Education (Tianjin University), Tianjin, 300072, China.
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, 300072, China.
| | - Jianjun Qiao
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China.
- Zhejiang Institute of Tianjin University, Shaoxing, Shaoxing, 312300, Zhejiang, China.
- Key Laboratory of Systems Bioengineering, Ministry of Education (Tianjin University), Tianjin, 300072, China.
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, 300072, China.
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin, 300072, China.
| |
Collapse
|
5
|
Pian Y, OuYang X. TRIM32 positively regulates c-di-GMP-Induced type I interferon signaling pathway in Listeria monocytogenes infection. Microbes Infect 2025:105499. [PMID: 40049511 DOI: 10.1016/j.micinf.2025.105499] [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: 10/24/2024] [Revised: 02/13/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025]
Abstract
Listeria monocytogenes (Lm) poses a significant threat to human health. TRIM32, an E3 ubiquitin ligase, plays a critical role in regulating immune responses to pathogen infections. Previous studies have shown that TRIM32 deficiency significantly impairs IFN-β production. In this study, we demonstrate that TRIM32 enhances IFN-β release upon activation by cyclic di-GMP (c-di-GMP). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses revealed that TRIM32 deficiency upregulates genes associated with metabolic pathways while downregulating those involved in cytokine signaling and inflammatory responses. Western blot analysis further indicated a significant reduction in ERK and JNK phosphorylation in splenocytes and peritoneal macrophages, suggesting that TRIM32 modulates the MAPK signaling pathway. Additionally, the duration of p38, STAT, and TBK1 phosphorylation was shortened in bone marrow-derived macrophages. Collectively, these findings highlight the role of TRIM32 in enhancing the host immune response against Lm infection.
Collapse
Affiliation(s)
- Yaya Pian
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China.
| | - Xuan OuYang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| |
Collapse
|
6
|
Gomez-Diaz C, Greulich W, Wefers B, Wang M, Bolsega S, Effern M, Varga DP, Han Z, Chen M, Bérouti M, Leonardi N, Schillinger U, Holzmann B, Liesz A, Roers A, Hölzel M, Basic M, Wurst W, Hornung V. RNase T2 restricts TLR13-mediated autoinflammation in vivo. J Exp Med 2025; 222:e20241424. [PMID: 39853306 PMCID: PMC11758920 DOI: 10.1084/jem.20241424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 11/18/2024] [Accepted: 12/20/2024] [Indexed: 01/26/2025] Open
Abstract
RNA-sensing TLRs are strategically positioned in the endolysosome to detect incoming nonself RNA. RNase T2 plays a critical role in processing long, structured RNA into short oligoribonucleotides that engage TLR7 or TLR8. In addition to its positive regulatory role, RNase T2 also restricts RNA recognition through unknown mechanisms, as patients deficient in RNase T2 suffer from neuroinflammation. Consistent with this, mice lacking RNase T2 exhibit interferon-dependent neuroinflammation, impaired hematopoiesis, and splenomegaly. However, the mechanism by which RNase T2 deficiency unleashes inflammation in vivo remains unknown. Here, we report that the inflammatory phenotype found in Rnaset2-/- mice is completely reversed in the absence of TLR13, suggesting aberrant accumulation of an RNA ligand for this receptor. Interestingly, this TLR13-driven inflammatory phenotype is also fully present in germ-free mice, suggesting a role for RNase T2 in limiting erroneous TLR13 activation by an as yet unidentified endogenous ligand. These results establish TLR13 as a potential self-sensor that is kept in check by RNase T2.
Collapse
Affiliation(s)
- Carlos Gomez-Diaz
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Wilhelm Greulich
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Benedikt Wefers
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
- TUM School of Life Sciences, Technische Universität München, Freising-Weihenstephan, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen Site Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Meiyue Wang
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Silvia Bolsega
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Maike Effern
- Institute of Experimental Oncology, Medical Faculty, University Hospital Bonn, Bonn, Germany
| | - Daniel P. Varga
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Zhe Han
- Institute for Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Minyi Chen
- Institute for Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Marleen Bérouti
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Natascia Leonardi
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Ulrike Schillinger
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Bernhard Holzmann
- Department of Surgery, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Arthur Liesz
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Axel Roers
- Institute for Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Hölzel
- Institute of Experimental Oncology, Medical Faculty, University Hospital Bonn, Bonn, Germany
| | - Marijana Basic
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
- TUM School of Life Sciences, Technische Universität München, Freising-Weihenstephan, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen Site Munich, Munich, Germany
| | - Veit Hornung
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
7
|
Li J, Yu J, Shen A, Lai S, Liu Z, He TS. The RNA-binding proteins regulate innate antiviral immune signaling by modulating pattern recognition receptors. Virol J 2024; 21:225. [PMID: 39304943 PMCID: PMC11414252 DOI: 10.1186/s12985-024-02503-x] [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: 12/22/2023] [Accepted: 09/12/2024] [Indexed: 09/22/2024] Open
Abstract
Viral infections pose significant threats to human health, leading to a diverse spectrum of infectious diseases. The innate immune system serves as the primary barrier against viruses and bacteria in the early stages of infection. A rapid and forceful antiviral innate immune response is triggered by distinguishing between self-nucleic acids and viral nucleic acids. RNA-binding proteins (RBPs) are a diverse group of proteins which contain specific structural motifs or domains for binding RNA molecules. In the last decade, numerous of studies have outlined that RBPs influence viral replication via diverse mechanisms, directly recognizing viral nucleic acids and modulating the activity of pattern recognition receptors (PRRs). In this review, we summarize the functions of RBPs in regulation of host-virus interplay by controlling the activation of PRRs, such as RIG-I, MDA5, cGAS and TLR3. RBPs are instrumental in facilitating the identification of viral RNA or DNA, as well as viral structural proteins within the cellular cytoplasm and nucleus, functioning as co-receptor elements. On the other hand, RBPs are capable of orchestrating the activation of PRRs and facilitating the transmission of antiviral signals to downstream adaptor proteins by post-translational modifications or aggregation. Gaining a deeper comprehension of the interaction between the host and viruses is crucial for the development of novel therapeutics targeting viral infections.
Collapse
Affiliation(s)
- Jianguo Li
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
- Center for Immunology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, China
- Graduate School, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Jingge Yu
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
- Department of Blood Transfusion, Jingmen Central Hospital, Jingmen, China
| | - Ao Shen
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
- Graduate School, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Suwen Lai
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Zhiping Liu
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China.
- Center for Immunology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, China.
| | - Tian-Sheng He
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China.
- Center for Immunology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, China.
| |
Collapse
|
8
|
Shao C, Chen J, Qiang B, Ye J, Yan F, Zhu Y. The role of cGAS-STING signaling in the development and therapy of head and neck squamous cell carcinoma. Front Immunol 2024; 15:1451305. [PMID: 39295867 PMCID: PMC11408205 DOI: 10.3389/fimmu.2024.1451305] [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: 06/19/2024] [Accepted: 08/19/2024] [Indexed: 09/21/2024] Open
Abstract
The cGAS-STING signaling pathway plays a critical role in innate immunity and defense against viral infections by orchestrating intracellular and adaptive immune responses to DNA. In the context of head and neck squamous cell carcinoma (HNSCC), this pathway has garnered significant attention due to its potential relevance in disease development and progression. HNSCC is strongly associated with risk factors such as smoking, heavy alcohol consumption, and human papillomavirus (HPV) infection. The presence or absence of HPV in HNSCC patients has been shown to have a profound impact on patient survival and prognosis, possibly due to the distinct biological characteristics of HPV-associated tumors. This review aims to provide a comprehensive overview of the current therapeutic approaches and challenges in HNSCC management, as well as the involvement of cGAS-STING signaling and its potential in the therapy of HNSCC. In addition, by advancing the present understanding of the mechanisms underlying this pathway, Activation of cGAS-STING-dependent inflammatory signaling downstream of chromosomal instability can exert both anti-tumoral and pro-tumoral effects in a cell-intrinsic manner, suggesting individualized therapy is of great importance. However, further exploration of the cGAS-STING signaling pathway is imperative for the effective management of HNSCC.
Collapse
Affiliation(s)
- Chengze Shao
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
- Department of Oncology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
| | - Jiawen Chen
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Bi Qiang
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
| | - Junmei Ye
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Fangrong Yan
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yongbo Zhu
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
| |
Collapse
|
9
|
Woo MS, Mayer C, Binkle-Ladisch L, Sonner JK, Rosenkranz SC, Shaposhnykov A, Rothammer N, Tsvilovskyy V, Lorenz SM, Raich L, Bal LC, Vieira V, Wagner I, Bauer S, Glatzel M, Conrad M, Merkler D, Freichel M, Friese MA. STING orchestrates the neuronal inflammatory stress response in multiple sclerosis. Cell 2024; 187:4043-4060.e30. [PMID: 38878778 DOI: 10.1016/j.cell.2024.05.031] [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: 01/16/2024] [Revised: 04/08/2024] [Accepted: 05/16/2024] [Indexed: 07/28/2024]
Abstract
Inflammation-induced neurodegeneration is a defining feature of multiple sclerosis (MS), yet the underlying mechanisms remain unclear. By dissecting the neuronal inflammatory stress response, we discovered that neurons in MS and its mouse model induce the stimulator of interferon genes (STING). However, activation of neuronal STING requires its detachment from the stromal interaction molecule 1 (STIM1), a process triggered by glutamate excitotoxicity. This detachment initiates non-canonical STING signaling, which leads to autophagic degradation of glutathione peroxidase 4 (GPX4), essential for neuronal redox homeostasis and thereby inducing ferroptosis. Both genetic and pharmacological interventions that target STING in neurons protect against inflammation-induced neurodegeneration. Our findings position STING as a central regulator of the detrimental neuronal inflammatory stress response, integrating inflammation with glutamate signaling to cause neuronal cell death, and present it as a tractable target for treating neurodegeneration in MS.
Collapse
Affiliation(s)
- Marcel S Woo
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christina Mayer
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lars Binkle-Ladisch
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jana K Sonner
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sina C Rosenkranz
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Artem Shaposhnykov
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicola Rothammer
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Volodymyr Tsvilovskyy
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Svenja M Lorenz
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
| | - Lukas Raich
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lukas C Bal
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Vanessa Vieira
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ingrid Wagner
- Department of Pathology and Immunology, Division of Clinical Pathology, Faculty of Medicine, University and University Hospital of Geneva, Geneva, Switzerland
| | - Simone Bauer
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
| | - Doron Merkler
- Department of Pathology and Immunology, Division of Clinical Pathology, Faculty of Medicine, University and University Hospital of Geneva, Geneva, Switzerland
| | - Marc Freichel
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
10
|
Zhao X, Zhang Y, Trejo-Cerro O, Kaplan E, Li Z, Albertsboer F, El Hammiri N, Mariz FC, Banks L, Ottonello S, Müller M. A safe and potentiated multi-type HPV L2-E7 nanoparticle vaccine with combined prophylactic and therapeutic activity. NPJ Vaccines 2024; 9:119. [PMID: 38926425 PMCID: PMC11208501 DOI: 10.1038/s41541-024-00914-z] [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: 12/21/2023] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Persistent infection with high-risk human papillomavirus (HPV) is widely recognized as the primary cause of cervical and other malignant cancers. There are six licensed prophylactic vaccines available against HPV, but none of them shows any significant therapeutic effect on pre-existing infections or lesions. Thus, a prophylactic vaccine also endowed with therapeutic activity would afford protection regardless of the vaccine recipients HPV-infection status. Here, we describe the refinement and further potentiation of a dual-purpose HPV nanoparticle vaccine (hereafter referred to as cPANHPVAX) relying on eight different HPV L2 peptide epitopes and on the E7 oncoantigens from HPV16 and 18. cPANHPVAX not only induces anti-HPV16 E7 cytotoxic T-cell responses in C57BL/6 mice, but also anti-HPV18 E7 T-cell responses in transgenic mice with the A2.DR1 haplotype. These cytotoxic responses add up to a potent, broad-coverage humoral (HPV-neutralizing) response. cPANHPVAX safety was further improved by deletion of the pRb-binding domains of E7. Our dual-purpose vaccine holds great potential for clinical translation as an immune-treatment capable of targeting active infections as well as established HPV-related malignancies, thus benefiting both uninfected and infected individuals.
Collapse
Affiliation(s)
- Xueer Zhao
- Tumorvirus-specific Vaccination Strategies, German Cancer Research Center, Heidelberg, Germany.
| | - Yueru Zhang
- Tumorvirus-specific Vaccination Strategies, German Cancer Research Center, Heidelberg, Germany
| | - Oscar Trejo-Cerro
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Ecem Kaplan
- Tumorvirus-specific Vaccination Strategies, German Cancer Research Center, Heidelberg, Germany
| | - Zhe Li
- B Cell Immunology, German Cancer Research Center, Heidelberg, Germany
| | - Femke Albertsboer
- Tumorvirus-specific Vaccination Strategies, German Cancer Research Center, Heidelberg, Germany
| | - Neyla El Hammiri
- Tumorvirus-specific Vaccination Strategies, German Cancer Research Center, Heidelberg, Germany
| | - Filipe Colaço Mariz
- Tumorvirus-specific Vaccination Strategies, German Cancer Research Center, Heidelberg, Germany
| | - Lawrence Banks
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Simone Ottonello
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Martin Müller
- Tumorvirus-specific Vaccination Strategies, German Cancer Research Center, Heidelberg, Germany.
| |
Collapse
|
11
|
En A, Bogireddi H, Thomas B, Stutzman AV, Ikegami S, LaForest B, Almakki O, Pytel P, Moskowitz IP, Ikegami K. Pervasive nuclear envelope ruptures precede ECM signaling and disease onset without activating cGAS-STING in Lamin-cardiomyopathy mice. Cell Rep 2024; 43:114284. [PMID: 38814785 PMCID: PMC11290591 DOI: 10.1016/j.celrep.2024.114284] [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: 10/03/2023] [Revised: 04/14/2024] [Accepted: 05/13/2024] [Indexed: 06/01/2024] Open
Abstract
Nuclear envelope (NE) ruptures are emerging observations in Lamin-related dilated cardiomyopathy, an adult-onset disease caused by loss-of-function mutations in Lamin A/C, a nuclear lamina component. Here, we test a prevailing hypothesis that NE ruptures trigger the pathological cGAS-STING cytosolic DNA-sensing pathway using a mouse model of Lamin cardiomyopathy. The reduction of Lamin A/C in cardio-myocyte of adult mice causes pervasive NE ruptures in cardiomyocytes, preceding inflammatory transcription, fibrosis, and fatal dilated cardiomyopathy. NE ruptures are followed by DNA damage accumulation without causing immediate cardiomyocyte death. However, cGAS-STING-dependent inflammatory signaling remains inactive. Deleting cGas or Sting does not rescue cardiomyopathy in the mouse model. The lack of cGAS-STING activation is likely due to the near absence of cGAS expression in adult cardiomyocytes at baseline. Instead, extracellular matrix (ECM) signaling is activated and predicted to initiate pro-inflammatory communication from Lamin-reduced cardiomyocytes to fibroblasts. Our work nominates ECM signaling, not cGAS-STING, as a potential inflammatory contributor in Lamin cardiomyopathy.
Collapse
Affiliation(s)
- Atsuki En
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Graduate School of Nanobioscience, Yokohama City University, Yokohama, Kanagawa 236-0027, Japan
| | - Hanumakumar Bogireddi
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Briana Thomas
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Alexis V Stutzman
- Department of Pediatrics, the University of Chicago, Chicago, IL 60637, USA
| | - Sachie Ikegami
- Department of Pediatrics, the University of Chicago, Chicago, IL 60637, USA
| | - Brigitte LaForest
- Department of Pediatrics, the University of Chicago, Chicago, IL 60637, USA
| | - Omar Almakki
- Department of Pediatrics, the University of Chicago, Chicago, IL 60637, USA
| | - Peter Pytel
- Department of Pathology, the University of Chicago, Chicago, IL 60637, USA
| | - Ivan P Moskowitz
- Department of Pediatrics, the University of Chicago, Chicago, IL 60637, USA; Department of Pathology, the University of Chicago, Chicago, IL 60637, USA; Department of Human Genetics, the University of Chicago, Chicago, IL 60637, USA
| | - Kohta Ikegami
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
| |
Collapse
|
12
|
Hui S, Kan W, Qin S, He P, Zhao J, Li H, Bai J, Wen J, Mou W, Hou M, Wei Z, Lin L, Xiao X, Xu G, Bai Z. Glycyrrhiza uralensis polysaccharides ameliorates cecal ligation and puncture-induced sepsis by inhibiting the cGAS-STING signaling pathway. Front Pharmacol 2024; 15:1374179. [PMID: 38904004 PMCID: PMC11188434 DOI: 10.3389/fphar.2024.1374179] [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: 02/15/2024] [Accepted: 05/06/2024] [Indexed: 06/22/2024] Open
Abstract
Ethnopharmacological relevance: G. uralensis Fisch. (Glycyrrhiza uralensis) is an ancient and widely used traditional Chinese medicine with good efficacy in clearing heat and detoxifying action. Studies suggest that Glycyrrhiza Uralensis Polysaccharides (GUP), one of the major components of G. uralensis, has anti-inflammatory, anti-cancer and hepatoprotective effects., but its exact molecular mechanism has not been explored in depth. Aim of the study: Objectives of our research are about exploring the anti-inflammatory role of GUP and the mechanisms of its action. Materials and methods: ELISA kits, Western blotting, immunofluorescence, quantitative real-time PCR, immunoprecipitation and DMXAA-mediated STING activation mice models were performed to investigate the role of GUP on the cGAS-STING pathway. To determine the anti-inflammatory effects of GUP, cecal ligation and puncture (CLP) sepsis models were employed. Results: GUP could effectively inhibit the activation of the cGAS-STING signaling pathway accompany by a decrease the expression of type I interferon-related genes and inflammatory factors in BMDMs, THP-1, and human PBMCs. Mechanistically, GUP does not affect the oligomerization of STING, but affects the interaction of STING with TBK1 and TBK1 with IRF3. Significantly, GUP had great therapeutic effects on DMXAA-induced agonist experiments in vivo as well as CLP sepsis in mice. Conclusion: Our studies suggest that GUP is an effective inhibitor of the cGAS-STING pathway, which may be a potential medicine for the treatment of inflammatory diseases mediated by the cGAS-STING pathway.
Collapse
Affiliation(s)
- Siwen Hui
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- China Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Wen Kan
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- China Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Shuanglin Qin
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- China Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Ping He
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- China Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jia Zhao
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- China Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Hui Li
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- China Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jun Bai
- Department of Neurosurgery, General Hospital of Chinese People Liberty Army, Beijing, China
| | - Jincai Wen
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- China Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Wenqing Mou
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- China Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Manting Hou
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- China Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Ziying Wei
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- China Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Li Lin
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- China Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiaohe Xiao
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- China Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Guang Xu
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- China Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Zhaofang Bai
- Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
- China Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
13
|
En A, Bogireddi H, Thomas B, Stutzman A, Ikegami S, LaForest B, Almakki O, Pytel P, Moskowitz IP, Ikegami K. Pervasive nuclear envelope ruptures precede ECM signaling and disease onset without activating cGAS-STING in Lamin-cardiomyopathy mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.28.555134. [PMID: 37693381 PMCID: PMC10491116 DOI: 10.1101/2023.08.28.555134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Nuclear envelope (NE) ruptures are emerging observations in Lamin-related dilated cardiomyopathy, an adult-onset disease caused by loss-of-function mutations in Lamin A/C, a nuclear lamina component. Here, we tested a prevailing hypothesis that NE ruptures trigger pathological cGAS-STING cytosolic DNA-sensing pathway, using a mouse model of Lamin-cardiomyopathy. Reduction of Lamin A/C in cardiomyocytes of adult mice caused pervasive NE ruptures in cardiomyocytes, preceding inflammatory transcription, fibrosis, and fatal dilated cardiomyopathy. NE ruptures were followed by DNA damage accumulation without causing immediate cardiomyocyte death. However, cGAS-STING-dependent inflammatory signaling remained inactive. Deleting cGas or Sting did not rescue cardiomyopathy. The lack of cGAS-STING activation was likely due to the near absence of cGAS expression in adult cardiomyocytes at baseline. Instead, extracellular matrix (ECM) signaling was activated and predicted to initiate pro-inflammatory communication from Lamin-reduced cardiomyocytes to fibroblasts. Our work nominates ECM signaling, not cGAS-STING, as a potential inflammatory contributor in Lamin-cardiomyopathy.
Collapse
Affiliation(s)
- Atsuki En
- Division of Molecular Cardvascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Graduate School of Nanobioscience, Yokohama City University, Yokohama, Kanagawa, Japan
| | - Hanumakumar Bogireddi
- Division of Molecular Cardvascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Briana Thomas
- Division of Molecular Cardvascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Alexis Stutzman
- Department of Pediatrics, The University of Chicago, Chicago, IL, USA
| | - Sachie Ikegami
- Department of Pediatrics, The University of Chicago, Chicago, IL, USA
| | - Brigitte LaForest
- Department of Pediatrics, The University of Chicago, Chicago, IL, USA
| | - Omar Almakki
- Department of Pediatrics, The University of Chicago, Chicago, IL, USA
| | - Peter Pytel
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Ivan P Moskowitz
- Department of Pediatrics, The University of Chicago, Chicago, IL, USA
- Department of Pathology, The University of Chicago, Chicago, IL, USA
- Department of Human Genetics, The University of Chicago, Chicago, IL, USA
| | - Kohta Ikegami
- Division of Molecular Cardvascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
14
|
Li Q, Jia M, Song H, Peng J, Zhao W, Zhang W. Astaxanthin Inhibits STING Carbonylation and Enhances Antiviral Responses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1188-1195. [PMID: 38391298 DOI: 10.4049/jimmunol.2300306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 01/19/2024] [Indexed: 02/24/2024]
Abstract
STING-mediated DNA sensing pathway plays a crucial role in the innate antiviral immune responses. Clarifying its regulatory mechanism and searching STING agonists has potential clinical implications. Although multiple STING agonists have been developed to target cancer, there are few for the treatment of infectious diseases. Astaxanthin, a natural and powerful antioxidant, serves many biological functions and as a potential candidate drug for many diseases. However, how astaxanthin combats viruses and whether astaxanthin regulates the cyclic GMP-AMP synthase-STING pathway remains unclear. In this study, we showed that astaxanthin markedly inhibited HSV-1-induced lipid peroxidation and inflammatory responses and enhanced the induction of type I IFN in C57BL/6J mice and mouse primary peritoneal macrophages. Mechanistically, astaxanthin inhibited HSV-1 infection and oxidative stress-induced STING carbonylation and consequently promoted STING translocation to the Golgi apparatus and oligomerization, which activated STING-dependent host defenses. Thus, our study reveals that astaxanthin displays a strong antiviral activity by targeting STING, suggesting that astaxanthin might be a promising STING agonist and a therapeutic target for viral infectious diseases.
Collapse
Affiliation(s)
- Qizhao Li
- Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Mutian Jia
- Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Hui Song
- Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jun Peng
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Wei Zhao
- Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Weifang Zhang
- Key Laboratory for Experimental Teratology of the Chinese Ministry of Education, and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
15
|
Becker B, Wottawa F, Bakr M, Koncina E, Mayr L, Kugler J, Yang G, Windross SJ, Neises L, Mishra N, Harris D, Tran F, Welz L, Schwärzler J, Bánki Z, Stengel ST, Ito G, Krötz C, Coleman OI, Jaeger C, Haller D, Paludan SR, Blumberg R, Kaser A, Cicin-Sain L, Schreiber S, Adolph TE, Letellier E, Rosenstiel P, Meiser J, Aden K. Serine metabolism is crucial for cGAS-STING signaling and viral defense control in the gut. iScience 2024; 27:109173. [PMID: 38496294 PMCID: PMC10943449 DOI: 10.1016/j.isci.2024.109173] [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: 11/15/2023] [Revised: 12/27/2023] [Accepted: 02/06/2024] [Indexed: 03/19/2024] Open
Abstract
Inflammatory bowel diseases are characterized by the chronic relapsing inflammation of the gastrointestinal tract. While the molecular causality between endoplasmic reticulum (ER) stress and intestinal inflammation is widely accepted, the metabolic consequences of chronic ER stress on the pathophysiology of IBD remain unclear. By using in vitro, in vivo models, and patient datasets, we identified a distinct polarization of the mitochondrial one-carbon metabolism and a fine-tuning of the amino acid uptake in intestinal epithelial cells tailored to support GSH and NADPH metabolism upon ER stress. This metabolic phenotype strongly correlates with IBD severity and therapy response. Mechanistically, we uncover that both chronic ER stress and serine limitation disrupt cGAS-STING signaling, impairing the epithelial response against viral and bacterial infection and fueling experimental enteritis. Consequently, the antioxidant treatment restores STING function and virus control. Collectively, our data highlight the importance of serine metabolism to allow proper cGAS-STING signaling and innate immune responses upon gut inflammation.
Collapse
Affiliation(s)
- Björn Becker
- Luxembourg Institute of Health, Department of Cancer Research, Luxembourg, Luxembourg
| | - Felix Wottawa
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Mohamed Bakr
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Eric Koncina
- Faculty of Science, Technology and Medicine, Department of Life Sciences and Medicine, Université du Luxembourg, Luxembourg, Luxembourg
| | - Lisa Mayr
- Department of Internal Medicine I, Gastroenterology, Hepatology, Metabolism & Endocrinology, Medical University of Innsbruck, Innsbruck, Austria
| | - Julia Kugler
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Guang Yang
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | | | - Laura Neises
- Luxembourg Institute of Health, Department of Cancer Research, Luxembourg, Luxembourg
| | - Neha Mishra
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Danielle Harris
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Florian Tran
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
- Department of Internal Medicine I, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Lina Welz
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
- Department of Internal Medicine I, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Julian Schwärzler
- Department of Internal Medicine I, Gastroenterology, Hepatology, Metabolism & Endocrinology, Medical University of Innsbruck, Innsbruck, Austria
| | - Zoltán Bánki
- Institute of Virology, Department of Hygiene, Microbiology and Public Health, Medical University of Innsbruck, Innsbruck, Austria
| | - Stephanie T. Stengel
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Go Ito
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Christina Krötz
- Luxembourg Institute of Health, Department of Cancer Research, Luxembourg, Luxembourg
| | - Olivia I. Coleman
- Chair of Nutrition and Immunology, TUM School of Life Sciences, Technical University of Munich, Luxembourg, Luxembourg
| | - Christian Jaeger
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Dirk Haller
- Chair of Nutrition and Immunology, TUM School of Life Sciences, Technical University of Munich, Luxembourg, Luxembourg
- ZIEL-Institute for Food & Health, Technical University of Munich, 85354 Freising, Germany
| | | | - Richard Blumberg
- Gastroenterology Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Arthur Kaser
- Division of Gastroenterology and Hepatology, Department of Medicine, Addenbrooke’s Hospital, University of Cambridge, Cambridge, England, UK
| | - Luka Cicin-Sain
- Helmholtz Zentrum für Infektionsforschung, Braunschweig, Germany
| | - Stefan Schreiber
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
- Department of Internal Medicine I, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Timon E. Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Metabolism & Endocrinology, Medical University of Innsbruck, Innsbruck, Austria
| | - Elisabeth Letellier
- Faculty of Science, Technology and Medicine, Department of Life Sciences and Medicine, Université du Luxembourg, Luxembourg, Luxembourg
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| | - Johannes Meiser
- Luxembourg Institute of Health, Department of Cancer Research, Luxembourg, Luxembourg
| | - Konrad Aden
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
- Department of Internal Medicine I, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Campus Kiel, 24105 Kiel, Germany
| |
Collapse
|
16
|
Li J, Canham SM, Wu H, Henault M, Chen L, Liu G, Chen Y, Yu G, Miller HR, Hornak V, Brittain SM, Michaud GA, Tutter A, Broom W, Digan ME, McWhirter SM, Sivick KE, Pham HT, Chen CH, Tria GS, McKenna JM, Schirle M, Mao X, Nicholson TB, Wang Y, Jenkins JL, Jain RK, Tallarico JA, Patel SJ, Zheng L, Ross NT, Cho CY, Zhang X, Bai XC, Feng Y. Activation of human STING by a molecular glue-like compound. Nat Chem Biol 2024; 20:365-372. [PMID: 37828400 PMCID: PMC10907298 DOI: 10.1038/s41589-023-01434-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 09/02/2023] [Indexed: 10/14/2023]
Abstract
Stimulator of interferon genes (STING) is a dimeric transmembrane adapter protein that plays a key role in the human innate immune response to infection and has been therapeutically exploited for its antitumor activity. The activation of STING requires its high-order oligomerization, which could be induced by binding of the endogenous ligand, cGAMP, to the cytosolic ligand-binding domain. Here we report the discovery through functional screens of a class of compounds, named NVS-STGs, that activate human STING. Our cryo-EM structures show that NVS-STG2 induces the high-order oligomerization of human STING by binding to a pocket between the transmembrane domains of the neighboring STING dimers, effectively acting as a molecular glue. Our functional assays showed that NVS-STG2 could elicit potent STING-mediated immune responses in cells and antitumor activities in animal models.
Collapse
Affiliation(s)
- Jie Li
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Stephen M Canham
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA.
| | - Hua Wu
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Martin Henault
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Lihao Chen
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Guoxun Liu
- Novartis Institutes for BioMedical Research, San Diego, CA, USA
| | - Yu Chen
- Novartis Institutes for BioMedical Research, San Diego, CA, USA
| | - Gary Yu
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Howard R Miller
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Viktor Hornak
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | | | | | - Antonin Tutter
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Wendy Broom
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | | | | | | | - Helen T Pham
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | | | - George S Tria
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | | | - Markus Schirle
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Xiaohong Mao
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | | | - Yuan Wang
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | | | - Rishi K Jain
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | | | - Sejal J Patel
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Lianxing Zheng
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Nathan T Ross
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Charles Y Cho
- Novartis Institutes for BioMedical Research, San Diego, CA, USA
| | - Xuewu Zhang
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Xiao-Chen Bai
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Yan Feng
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA.
| |
Collapse
|
17
|
Winstone L, Jung Y, Wu Y. DDX41: exploring the roles of a versatile helicase. Biochem Soc Trans 2024; 52:395-405. [PMID: 38348889 PMCID: PMC10903454 DOI: 10.1042/bst20230725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 02/29/2024]
Abstract
DDX41 is a DEAD-box helicase and is conserved across species. Mutations in DDX41 have been associated with myeloid neoplasms, including myelodysplastic syndrome and acute myeloid leukemia. Though its pathogenesis is not completely known, DDX41 has been shown to have many cellular roles, including in pre-mRNA splicing, innate immune sensing, ribosome biogenesis, translational regulation, and R-loop metabolism. In this review, we will summarize the latest understandings regarding the various roles of DDX41, as well as highlight challenges associated with drug development to target DDX41. Overall, understanding the molecular and cellular mechanisms of DDX41 could help develop novel therapeutic options for DDX41 mutation-related hematologic malignancies.
Collapse
Affiliation(s)
- Lacey Winstone
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Yohan Jung
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Yuliang Wu
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| |
Collapse
|
18
|
Ge J, Zhang L. RNF5: inhibiting antiviral immunity and shaping virus life cycle. Front Immunol 2024; 14:1324516. [PMID: 38250078 PMCID: PMC10796512 DOI: 10.3389/fimmu.2023.1324516] [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: 10/19/2023] [Accepted: 12/15/2023] [Indexed: 01/23/2024] Open
Abstract
RNF5 is an E3 ubiquitin ligase involved in various physiological processes such as protein localization and cancer progression. Recent studies have shown that RNF5 significantly inhibits antiviral innate immunity by promoting the ubiquitination and degradation of STING and MAVS, which are essential adaptor proteins, as well as their downstream signal IRF3. The abundance of RNF5 is delicately regulated by both host factors and viruses. Host factors have been found to restrict RNF5-mediated ubiquitination, maintaining the stability of STING or MAVS through distinct mechanisms. Meanwhile, viruses have developed ingenious strategies to hijack RNF5 to ubiquitinate and degrade immune proteins. Moreover, recent studies have revealed the multifaceted roles of RNF5 in the life cycle of various viruses, including SARS-CoV-2 and KSHV. Based on these emerging discoveries, RNF5 represents a novel means of modulating antiviral immunity. In this review, we summarize the latest research on the roles of RNF5 in antiviral immunity and virus life cycle. This comprehensive understanding could offer valuable insights into exploring potential therapeutic applications focused on targeting RNF5 during viral infections.
Collapse
Affiliation(s)
- Junyi Ge
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Leiliang Zhang
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
19
|
Verhoeven J, Jacobs KA, Rizzollo F, Lodi F, Hua Y, Poźniak J, Narayanan Srinivasan A, Houbaert D, Shankar G, More S, Schaaf MB, Dubroja Lakic N, Ganne M, Lamote J, Van Weyenbergh J, Boon L, Bechter O, Bosisio F, Uchiyama Y, Bertrand MJ, Marine JC, Lambrechts D, Bergers G, Agrawal M, Agostinis P. Tumor endothelial cell autophagy is a key vascular-immune checkpoint in melanoma. EMBO Mol Med 2023; 15:e18028. [PMID: 38009521 DOI: 10.15252/emmm.202318028] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 10/25/2023] [Accepted: 10/30/2023] [Indexed: 11/29/2023] Open
Abstract
Tumor endothelial cells (TECs) actively repress inflammatory responses and maintain an immune-excluded tumor phenotype. However, the molecular mechanisms that sustain TEC-mediated immunosuppression remain largely elusive. Here, we show that autophagy ablation in TECs boosts antitumor immunity by supporting infiltration and effector function of T-cells, thereby restricting melanoma growth. In melanoma-bearing mice, loss of TEC autophagy leads to the transcriptional expression of an immunostimulatory/inflammatory TEC phenotype driven by heightened NF-kB and STING signaling. In line, single-cell transcriptomic datasets from melanoma patients disclose an enriched InflammatoryHigh /AutophagyLow TEC phenotype in correlation with clinical responses to immunotherapy, and responders exhibit an increased presence of inflamed vessels interfacing with infiltrating CD8+ T-cells. Mechanistically, STING-dependent immunity in TECs is not critical for the immunomodulatory effects of autophagy ablation, since NF-kB-driven inflammation remains functional in STING/ATG5 double knockout TECs. Hence, our study identifies autophagy as a principal tumor vascular anti-inflammatory mechanism dampening melanoma antitumor immunity.
Collapse
Affiliation(s)
- Jelle Verhoeven
- Cell Death Research and Therapy Laboratory, Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Kathryn A Jacobs
- Cell Death Research and Therapy Laboratory, Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Francesca Rizzollo
- Cell Death Research and Therapy Laboratory, Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Francesca Lodi
- Laboratory of Translational Genetics, Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Yichao Hua
- Laboratory of Tumor Microenvironment and Therapeutic Resistance Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | - Joanna Poźniak
- Department of Oncology, KU Leuven, Leuven, Belgium
- Laboratory for Molecular Cancer Biology, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Adhithya Narayanan Srinivasan
- Cell Death Research and Therapy Laboratory, Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Diede Houbaert
- Cell Death Research and Therapy Laboratory, Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Gautam Shankar
- Laboratory of Translational Cell and Tissue Research, Department of Pathology, KULeuven and UZ Leuven, Leuven, Belgium
- Department of Pathology, UZLeuven, Leuven, Belgium
| | - Sanket More
- Cell Death Research and Therapy Laboratory, Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Marco B Schaaf
- Cell Death Research and Therapy Laboratory, Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Nikolina Dubroja Lakic
- Laboratory of Translational Cell and Tissue Research, Department of Pathology, KULeuven and UZ Leuven, Leuven, Belgium
- Department of Pathology, UZLeuven, Leuven, Belgium
| | - Maarten Ganne
- Cell Death Research and Therapy Laboratory, Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jochen Lamote
- Department of Oncology, KU Leuven, Leuven, Belgium
- Laboratory for Molecular Cancer Biology, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Johan Van Weyenbergh
- Laboratory of Clinical and Epidemiological Virology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Louis Boon
- Polpharma Biologics, Utrecht, The Netherlands
| | - Oliver Bechter
- Department of General Medical Oncology, UZ Leuven, Leuven, Belgium
| | - Francesca Bosisio
- Laboratory of Translational Cell and Tissue Research, Department of Pathology, KULeuven and UZ Leuven, Leuven, Belgium
- Department of Pathology, UZLeuven, Leuven, Belgium
| | - Yasuo Uchiyama
- Department of Cellular and Molecular Neuropathology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Mathieu Jm Bertrand
- VIB Center for Inflammation Research, Ghent University, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Jean Christophe Marine
- Laboratory for Molecular Cancer Biology, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Diether Lambrechts
- Laboratory of Translational Genetics, Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Gabriele Bergers
- Laboratory of Tumor Microenvironment and Therapeutic Resistance Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | - Madhur Agrawal
- Cell Death Research and Therapy Laboratory, Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Patrizia Agostinis
- Cell Death Research and Therapy Laboratory, Center for Cancer Biology, VIB, Leuven, Belgium
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
20
|
Wright MJ, Bai G. Bacterial second messenger cyclic di-AMP in streptococci. Mol Microbiol 2023; 120:791-804. [PMID: 37898560 DOI: 10.1111/mmi.15187] [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: 08/04/2023] [Revised: 10/10/2023] [Accepted: 10/15/2023] [Indexed: 10/30/2023]
Abstract
Cyclic dimeric adenosine monophosphate (c-di-AMP) has been well studied in bacteria, including those of the genus Streptococcus, since the first recognition of this dinucleotide in 2008. Streptococci possess a sole diadenylate cyclase, CdaA, and distinct c-di-AMP phosphodiesterases. Interestingly, cdaA is required for viability of some streptococcal species but not all when streptococci are grown in standard laboratory media. Bacteria of this genus also have distinct c-di-AMP effector proteins, diverse c-di-AMP-signaling pathways, and subsequent biological outcomes. In streptococci, c-di-AMP may influence bacterial growth, morphology, biofilm formation, competence program, drug resistance, and bacterial pathogenesis. c-di-AMP secreted by streptococci has also been shown to interact with the mammalian host and induces immune responses including type I interferon production. In this review, we summarize the reported c-di-AMP networks in seven species of the genus Streptococcus, which cause diverse clinical manifestations, and propose future perspectives to investigate the signaling molecule in these streptococcal pathogens.
Collapse
Affiliation(s)
- Michael J Wright
- Department of Internal Medicine, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Guangchun Bai
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, New York, USA
| |
Collapse
|
21
|
Patil V, Hernandez-Franco JF, Yadagiri G, Bugybayeva D, Dolatyabi S, Feliciano-Ruiz N, Schrock J, Suresh R, Hanson J, Yassine H, HogenEsch H, Renukaradhya GJ. Characterization of the Efficacy of a Split Swine Influenza A Virus Nasal Vaccine Formulated with a Nanoparticle/STING Agonist Combination Adjuvant in Conventional Pigs. Vaccines (Basel) 2023; 11:1707. [PMID: 38006039 PMCID: PMC10675483 DOI: 10.3390/vaccines11111707] [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: 09/07/2023] [Revised: 10/09/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
Swine influenza A viruses (SwIAVs) are pathogens of both veterinary and medical significance. Intranasal (IN) vaccination has the potential to reduce flu infection. We investigated the efficacy of split SwIAV H1N2 antigens adsorbed with a plant origin nanoparticle adjuvant [Nano11-SwIAV] or in combination with a STING agonist ADU-S100 [NanoS100-SwIAV]. Conventional pigs were vaccinated via IN and challenged with a heterologous SwIAV H1N1-OH7 or 2009 H1N1 pandemic virus. Immunologically, in NanoS100-SwIAV vaccinates, we observed enhanced frequencies of activated monocytes in the blood of the pandemic virus challenged animals and in tracheobronchial lymph nodes (TBLN) of H1N1-OH7 challenged animals. In both groups of the virus challenged pigs, increased frequencies of IL-17A+ and CD49d+IL-17A+ cytotoxic lymphocytes were observed in Nano11-SwIAV vaccinates in the draining TBLN. Enhanced frequency of CD49d+IFNγ+ CTLs in the TBLN and blood of both the Nano11-based SwIAV vaccinates was observed. Animals vaccinated with both Nano11-based vaccines had upregulated cross-reactive secretory IgA in the lungs and serum IgG against heterologous and heterosubtypic viruses. However, in NanoS100-SwIAV vaccinates, a slight early reduction in the H1N1 pandemic virus and a late reduction in the SwIAV H1N1-OH7 load in the nasal passages were detected. Hence, despite vast genetic differences between the vaccine and both the challenge viruses, IN vaccination with NanoS100-SwIAV induced antigen-specific moderate levels of cross-protective immune responses.
Collapse
Affiliation(s)
- Veerupaxagouda Patil
- Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691, USA; (V.P.); (G.Y.); (D.B.); (S.D.); (N.F.-R.); (J.S.); (R.S.); (J.H.)
| | - Juan F. Hernandez-Franco
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA;
| | - Ganesh Yadagiri
- Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691, USA; (V.P.); (G.Y.); (D.B.); (S.D.); (N.F.-R.); (J.S.); (R.S.); (J.H.)
| | - Dina Bugybayeva
- Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691, USA; (V.P.); (G.Y.); (D.B.); (S.D.); (N.F.-R.); (J.S.); (R.S.); (J.H.)
| | - Sara Dolatyabi
- Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691, USA; (V.P.); (G.Y.); (D.B.); (S.D.); (N.F.-R.); (J.S.); (R.S.); (J.H.)
| | - Ninoshkaly Feliciano-Ruiz
- Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691, USA; (V.P.); (G.Y.); (D.B.); (S.D.); (N.F.-R.); (J.S.); (R.S.); (J.H.)
| | - Jennifer Schrock
- Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691, USA; (V.P.); (G.Y.); (D.B.); (S.D.); (N.F.-R.); (J.S.); (R.S.); (J.H.)
| | - Raksha Suresh
- Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691, USA; (V.P.); (G.Y.); (D.B.); (S.D.); (N.F.-R.); (J.S.); (R.S.); (J.H.)
| | - Juliette Hanson
- Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691, USA; (V.P.); (G.Y.); (D.B.); (S.D.); (N.F.-R.); (J.S.); (R.S.); (J.H.)
| | - Hadi Yassine
- Biomedical Research Center, Research Institute in Doha, Qatar University, QU-NRC, Building H10, Zone 5, Room D101, Doha P.O. Box 2713, Qatar;
| | - Harm HogenEsch
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47907, USA;
| | - Gourapura J. Renukaradhya
- Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691, USA; (V.P.); (G.Y.); (D.B.); (S.D.); (N.F.-R.); (J.S.); (R.S.); (J.H.)
| |
Collapse
|
22
|
Wu X, Zhou L, Ye C, Zha Z, Li C, Feng C, Zhang Y, Jin Q, Pan J. Destruction of self-derived PAMP via T3SS2 effector VopY to subvert PAMP-triggered immunity mediates Vibrio parahaemolyticus pathogenicity. Cell Rep 2023; 42:113261. [PMID: 37847589 DOI: 10.1016/j.celrep.2023.113261] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 05/20/2023] [Accepted: 09/28/2023] [Indexed: 10/19/2023] Open
Abstract
Cyclic di-guanosine monophosphate (c-di-GMP) is a unique bacterial second messenger but is hijacked by host cells during bacterial infection as a pathogen-associated molecular pattern (PAMP) to trigger STING-dependent immune responses. Here, we show that upon infection, VopY, an effector of Vibrio parahaemolyticus, is injected into host cells by type III secretion system 2 (T3SS2), a secretion system unique to its pathogenic strains and indispensable for enterotoxicity. VopY is an EAL-domain-containing phosphodiesterase and is capable of hydrolyzing c-di-GMP. VopY expression in host cells prevents the activation of STING and STING-dependent downstream signaling triggered by c-di-GMP and, consequently, suppresses type I interferon immune responses. The presence of VopY in V. parahaemolyticus enables it to cause both T3SS2-dependent enterotoxicity and cytotoxicity. These findings uncover the destruction of self-derived PAMPs by injecting specific effectors to suppress PAMP-triggered immune responses as a unique strategy for bacterial pathogens to subvert immunity and cause disease.
Collapse
Affiliation(s)
- Xuan Wu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Lantian Zhou
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Chen Ye
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Zhenzhong Zha
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Chuchu Li
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Chao Feng
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Yue Zhang
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Qian Jin
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Jianyi Pan
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| |
Collapse
|
23
|
Wang Y, De Labastida Rivera F, Edwards CL, Frame TC, Engel JA, Bukali L, Na J, Ng SS, Corvino D, Montes de Oca M, Bunn PT, Soon MS, Andrew D, Loughland JR, Zhang J, Amante FH, Barber BE, McCarthy JS, Lopez JA, Boyle MJ, Engwerda CR. STING activation promotes autologous type I interferon-dependent development of type 1 regulatory T cells during malaria. J Clin Invest 2023; 133:e169417. [PMID: 37781920 PMCID: PMC10541195 DOI: 10.1172/jci169417] [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: 02/03/2023] [Accepted: 08/08/2023] [Indexed: 10/03/2023] Open
Abstract
The development of highly effective malaria vaccines and improvement of drug-treatment protocols to boost antiparasitic immunity are critical for malaria elimination. However, the rapid establishment of parasite-specific immune regulatory networks following exposure to malaria parasites hampers these efforts. Here, we identified stimulator of interferon genes (STING) as a critical mediator of type I interferon production by CD4+ T cells during blood-stage Plasmodium falciparum infection. The activation of STING in CD4+ T cells by cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) stimulated IFNB gene transcription, which promoted development of IL-10- and IFN-γ-coproducing CD4+ T (type I regulatory [Tr1]) cells. The critical role for type I IFN signaling for Tr1 cell development was confirmed in vivo using a preclinical malaria model. CD4+ T cell sensitivity to STING phosphorylation was increased in healthy volunteers following P. falciparum infection, particularly in Tr1 cells. These findings identified STING expressed by CD4+ T cells as an important mediator of type I IFN production and Tr1 cell development and activation during malaria.
Collapse
Affiliation(s)
- Yulin Wang
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Griffith University, School of Environment and Science, Nathan, Australia
| | | | - Chelsea L. Edwards
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- University of Queensland, School of Medicine, Brisbane, Australia
| | - Teija C.M. Frame
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- University of Queensland, School of Medicine, Brisbane, Australia
| | | | - Luzia Bukali
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- University of Queensland, School of Medicine, Brisbane, Australia
| | - Jinrui Na
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- University of Queensland, School of Medicine, Brisbane, Australia
| | - Susanna S. Ng
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Institute of Experimental Oncology, University Hospital Bonn, Bonn, Germany
| | - Dillon Corvino
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Institute of Experimental Oncology, University Hospital Bonn, Bonn, Germany
| | - Marcela Montes de Oca
- York Biomedical Research Institute, Hull York Medical School, University of York, York, United Kingdom
| | - Patrick T. Bunn
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Megan S.F. Soon
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Dean Andrew
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | | | - Jia Zhang
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Fiona H. Amante
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | | | - James S. McCarthy
- Victorian Infectious Diseases Services, Doherty Institute, University of Melbourne, Melbourne, Australia
| | - J. Alejandro Lopez
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Griffith University, School of Environment and Science, Nathan, Australia
| | - Michelle J. Boyle
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Life Sciences Division, Burnet Institute, Melbourne, Australia
| | | |
Collapse
|
24
|
Bichet MC, Adderley J, Avellaneda-Franco L, Magnin-Bougma I, Torriero-Smith N, Gearing LJ, Deffrasnes C, David C, Pepin G, Gantier MP, Lin RCY, Patwa R, Moseley GW, Doerig C, Barr JJ. Mammalian cells internalize bacteriophages and use them as a resource to enhance cellular growth and survival. PLoS Biol 2023; 21:e3002341. [PMID: 37883333 PMCID: PMC10602308 DOI: 10.1371/journal.pbio.3002341] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 09/20/2023] [Indexed: 10/28/2023] Open
Abstract
There is a growing appreciation that the direct interaction between bacteriophages and the mammalian host can facilitate diverse and unexplored symbioses. Yet the impact these bacteriophages may have on mammalian cellular and immunological processes is poorly understood. Here, we applied highly purified phage T4, free from bacterial by-products and endotoxins to mammalian cells and analyzed the cellular responses using luciferase reporter and antibody microarray assays. Phage preparations were applied in vitro to either A549 lung epithelial cells, MDCK-I kidney cells, or primary mouse bone marrow derived macrophages with the phage-free supernatant serving as a comparative control. Highly purified T4 phages were rapidly internalized by mammalian cells and accumulated within macropinosomes but did not activate the inflammatory DNA response TLR9 or cGAS-STING pathways. Following 8 hours of incubation with T4 phage, whole cell lysates were analyzed via antibody microarray that detected expression and phosphorylation levels of human signaling proteins. T4 phage application led to the activation of AKT-dependent pathways, resulting in an increase in cell metabolism, survival, and actin reorganization, the last being critical for macropinocytosis and potentially regulating a positive feedback loop to drive further phage internalization. T4 phages additionally down-regulated CDK1 and its downstream effectors, leading to an inhibition of cell cycle progression and an increase in cellular growth through a prolonged G1 phase. These interactions demonstrate that highly purified T4 phages do not activate DNA-mediated inflammatory pathways but do trigger protein phosphorylation cascades that promote cellular growth and survival. We conclude that mammalian cells are internalizing bacteriophages as a resource to promote cellular growth and metabolism.
Collapse
Affiliation(s)
- Marion C. Bichet
- School of Biological Sciences, Monash University, Clayton, Australia
- ACTALIA, Food Safety Department, Saint-Lô, France
- University of Lorraine, CNRS, LCPME, Vandœuvre-lès-Nancy, France
| | - Jack Adderley
- School of Health and Biomedical Science, RMIT University, Bundoora, Australia
| | | | | | | | - Linden J. Gearing
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, Australia
| | - Celine Deffrasnes
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Cassandra David
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Genevieve Pepin
- Medical Biology Department, Université du Québec à Trois-Rivières, Trois-Rivières, Québec, Canada
| | - Michael P. Gantier
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, Australia
| | - Ruby CY Lin
- Centre for Infectious Diseases and Microbiology; The Westmead Institute for Medical Research, Westmead, Australia
| | - Ruzeen Patwa
- School of Biological Sciences, Monash University, Clayton, Australia
| | - Gregory W. Moseley
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Christian Doerig
- School of Health and Biomedical Science, RMIT University, Bundoora, Australia
| | - Jeremy J. Barr
- School of Biological Sciences, Monash University, Clayton, Australia
| |
Collapse
|
25
|
Tsui V, Lyu R, Novakovic S, Stringer JM, Dunleavy JE, Granger E, Semple T, Leichter A, Martelotto LG, Merriner DJ, Liu R, McNeill L, Zerafa N, Hoffmann ER, O’Bryan MK, Hutt K, Deans AJ, Heierhorst J, McCarthy DJ, Crismani W. Fancm has dual roles in the limiting of meiotic crossovers and germ cell maintenance in mammals. CELL GENOMICS 2023; 3:100349. [PMID: 37601968 PMCID: PMC10435384 DOI: 10.1016/j.xgen.2023.100349] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 03/30/2023] [Accepted: 06/02/2023] [Indexed: 08/22/2023]
Abstract
Meiotic crossovers are required for accurate chromosome segregation and producing new allelic combinations. Meiotic crossover numbers are tightly regulated within a narrow range, despite an excess of initiating DNA double-strand breaks. Here, we reveal the tumor suppressor FANCM as a meiotic anti-crossover factor in mammals. We use unique large-scale crossover analyses with both single-gamete sequencing and pedigree-based bulk-sequencing datasets to identify a genome-wide increase in crossover frequencies in Fancm-deficient mice. Gametogenesis is heavily perturbed in Fancm loss-of-function mice, which is consistent with the reproductive defects reported in humans with biallelic FANCM mutations. A portion of the gametogenesis defects can be attributed to the cGAS-STING pathway after birth. Despite the gametogenesis phenotypes in Fancm mutants, both sexes are capable of producing offspring. We propose that the anti-crossover function and role in gametogenesis of Fancm are separable and will inform diagnostic pathways for human genomic instability disorders.
Collapse
Affiliation(s)
- Vanessa Tsui
- DNA Repair and Recombination Laboratory, St Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia
- The Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Parkville, VIC, Australia
| | - Ruqian Lyu
- Bioinformatics and Cellular Genomics, St Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia
- Melbourne Integrative Genomics, Faculty of Science, The University of Melbourne, Parkville, VIC, Australia
| | - Stevan Novakovic
- DNA Repair and Recombination Laboratory, St Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia
| | - Jessica M. Stringer
- Ovarian Biology Laboratory, Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, Australia
| | - Jessica E.M. Dunleavy
- Male Infertility and Germ Cell Biology Group, School of BioSciences and the Bio21 Institute, Faculty of Science, The University of Melbourne, Parkville, VIC, Australia
| | - Elissah Granger
- DNA Repair and Recombination Laboratory, St Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia
| | - Tim Semple
- Single Cell Innovation Laboratory, Centre for Cancer Research, University of Melbourne, Parkville, VIC, Australia
| | - Anna Leichter
- Single Cell Innovation Laboratory, Centre for Cancer Research, University of Melbourne, Parkville, VIC, Australia
| | - Luciano G. Martelotto
- Single Cell Innovation Laboratory, Centre for Cancer Research, University of Melbourne, Parkville, VIC, Australia
| | - D. Jo Merriner
- Male Infertility and Germ Cell Biology Group, School of BioSciences and the Bio21 Institute, Faculty of Science, The University of Melbourne, Parkville, VIC, Australia
| | - Ruijie Liu
- Bioinformatics and Cellular Genomics, St Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia
- Melbourne Integrative Genomics, Faculty of Science, The University of Melbourne, Parkville, VIC, Australia
| | - Lucy McNeill
- DNA Repair and Recombination Laboratory, St Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia
| | - Nadeen Zerafa
- Ovarian Biology Laboratory, Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, Australia
| | - Eva R. Hoffmann
- DNRF Center for Chromosome Stability, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Moira K. O’Bryan
- Male Infertility and Germ Cell Biology Group, School of BioSciences and the Bio21 Institute, Faculty of Science, The University of Melbourne, Parkville, VIC, Australia
| | - Karla Hutt
- Ovarian Biology Laboratory, Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, Australia
| | - Andrew J. Deans
- The Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Parkville, VIC, Australia
- Genome Stability Unit, St Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia
| | - Jörg Heierhorst
- The Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Parkville, VIC, Australia
- Molecular Genetics Unit, St Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia
| | - Davis J. McCarthy
- Bioinformatics and Cellular Genomics, St Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia
- Melbourne Integrative Genomics, Faculty of Science, The University of Melbourne, Parkville, VIC, Australia
| | - Wayne Crismani
- DNA Repair and Recombination Laboratory, St Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia
- The Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
26
|
Chen T, Feng Y, Sun W, Zhao G, Wu H, Cheng X, Zhao F, Zhang L, Zheng Y, Zhan P, Zhao W, Liu B, Gao C. The nucleotide receptor STING translocates to the phagosomes to negatively regulate anti-fungal immunity. Immunity 2023; 56:1727-1742.e6. [PMID: 37379835 DOI: 10.1016/j.immuni.2023.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 04/26/2023] [Accepted: 06/02/2023] [Indexed: 06/30/2023]
Abstract
STING (stimulator of interferon genes) exerts protective cellular responses to viral infection via induction of interferon production and autophagy. Here, we report the role of STING in modulating the immune responses toward fungal infection. Upon Candida albicans stimulation, STING transited alongside the endoplasmic reticulum (ER) to the phagosomes. In phagosomes, STING directly bound with Src via the N-terminal 18 amino acids of STING, and this binding prevented Src from recruiting and phosphorylating Syk. Consistently, Syk-associated signaling and production of pro-inflammatory cytokines and chemokines were increased in mouse BMDCs (bone-marrow-derived dendritic cells) lacking STING with fungal treatment. STING deficiency improved anti-fungal immunity in systemic C. albicans infection. Importantly, administration of the N-terminal 18-aa (amino acid) peptide of STING improved host outcomes in disseminated fungal infection. Overall, our study identifies a previously unrecognized function of STING in negatively regulating anti-fungal immune responses and offers a potential therapeutic strategy for controlling C. albicans infection.
Collapse
Affiliation(s)
- Tian Chen
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Pathogenic Biology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China
| | - Yiting Feng
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China
| | - Wanwei Sun
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China
| | - Guimin Zhao
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China
| | - Han Wu
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China
| | - Xiaochen Cheng
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China
| | - Fabao Zhao
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, P.R. China
| | - Lei Zhang
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China
| | - Yi Zheng
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China
| | - Peng Zhan
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, P.R. China
| | - Wei Zhao
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Pathogenic Biology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China
| | - Bingyu Liu
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China
| | - Chengjiang Gao
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan 250012, Shandong, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan 250012, Shandong, P.R. China.
| |
Collapse
|
27
|
Chauvin SD, Stinson WA, Platt DJ, Poddar S, Miner JJ. Regulation of cGAS and STING signaling during inflammation and infection. J Biol Chem 2023; 299:104866. [PMID: 37247757 PMCID: PMC10316007 DOI: 10.1016/j.jbc.2023.104866] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 05/31/2023] Open
Abstract
Stimulator of interferon genes (STING) is a sensor of cyclic dinucleotides including cyclic GMP-AMP, which is produced by cyclic GMP-AMP synthase (cGAS) in response to cytosolic DNA. The cGAS-STING signaling pathway regulates both innate and adaptive immune responses, as well as fundamental cellular functions such as autophagy, senescence, and apoptosis. Mutations leading to constitutive activation of STING cause devastating human diseases. Thus, the cGAS-STING pathway is of great interest because of its role in diverse cellular processes and because of the potential therapeutic implications of targeting cGAS and STING. Here, we review molecular and cellular mechanisms of STING signaling, and we propose a framework for understanding the immunological and other cellular functions of STING in the context of disease.
Collapse
Affiliation(s)
- Samuel D Chauvin
- Departments of Medicine and Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - W Alexander Stinson
- Departments of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Derek J Platt
- Department Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Subhajit Poddar
- Departments of Medicine and Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jonathan J Miner
- Departments of Medicine and Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA; Departments of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA; Department Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA; Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA.
| |
Collapse
|
28
|
Dharshika C, Gonzales J, Chow A, Morales-Soto W, Gulbransen BD. Stimulator of interferon genes (STING) expression in the enteric nervous system and contributions of glial STING in disease. Neurogastroenterol Motil 2023; 35:e14553. [PMID: 37309618 PMCID: PMC10266835 DOI: 10.1111/nmo.14553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/26/2023] [Accepted: 02/07/2023] [Indexed: 06/14/2023]
Abstract
BACKGROUND Appropriate host-microbe interactions are essential for enteric glial development and subsequent gastrointestinal function, but the potential mechanisms of microbe-glial communication are unclear. Here, we tested the hypothesis that enteric glia express the pattern recognition receptor stimulator of interferon genes (STING) and communicate with the microbiome through this pathway to modulate gastrointestinal inflammation. METHODS In situ transcriptional labeling and immunohistochemistry were used to examine STING and IFNβ expression in enteric neurons and glia. Glial-STING KO mice (Sox10CreERT2+/- ;STINGfl/fl ) and IFNβ ELISA were used to characterize the role of enteric glia in canonical STING activation. The role of glial STING in gastrointestinal inflammation was assessed in the 3% DSS colitis model. RESULTS Enteric glia and neurons express STING, but only enteric neurons express IFNβ. While both the myenteric and submucosal plexuses produce IFNβ with STING activation, enteric glial STING plays a minor role in its production and seems more involved in autophagy processes. Furthermore, deleting enteric glial STING does not affect weight loss, colitis severity, or neuronal cell proportions in the DSS colitis model. CONCLUSION Taken together, our data support canonical roles for STING and IFNβ signaling in the enteric nervous system through enteric neurons but that enteric glia do not use these same mechanisms. We propose that enteric glial STING may utilize alternative signaling mechanisms and/or is only active in particular disease conditions. Regardless, this study provides the first glimpse of STING signaling in the enteric nervous system and highlights a potential avenue of neuroglial-microbial communication.
Collapse
Affiliation(s)
- Christine Dharshika
- Department of Physiology, Neuroscience Program, Michigan State University, 567 Wilson Road, East Lansing, MI, 48824 USA
- College of Human Medicine, Michigan State University, 804 Service Road, East Lansing, MI, 48824 USA
| | - Jacques Gonzales
- Department of Physiology, Neuroscience Program, Michigan State University, 567 Wilson Road, East Lansing, MI, 48824 USA
| | - Aaron Chow
- Department of Physiology, Neuroscience Program, Michigan State University, 567 Wilson Road, East Lansing, MI, 48824 USA
| | - Wilmarie Morales-Soto
- Department of Physiology, Neuroscience Program, Michigan State University, 567 Wilson Road, East Lansing, MI, 48824 USA
| | - Brian D. Gulbransen
- Department of Physiology, Neuroscience Program, Michigan State University, 567 Wilson Road, East Lansing, MI, 48824 USA
| |
Collapse
|
29
|
Balka KR, Venkatraman R, Saunders TL, Shoppee A, Pang ES, Magill Z, Homman-Ludiye J, Huang C, Lane RM, York HM, Tan P, Schittenhelm RB, Arumugam S, Kile BT, O'Keeffe M, De Nardo D. Termination of STING responses is mediated via ESCRT-dependent degradation. EMBO J 2023:e112712. [PMID: 37139896 DOI: 10.15252/embj.2022112712] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 04/14/2023] [Accepted: 04/17/2023] [Indexed: 05/05/2023] Open
Abstract
cGAS-STING signalling is induced by detection of foreign or mislocalised host double-stranded (ds)DNA within the cytosol. STING acts as the major signalling hub, where it controls production of type I interferons and inflammatory cytokines. Basally, STING resides on the ER membrane. Following activation STING traffics to the Golgi to initiate downstream signalling and subsequently to endolysosomal compartments for degradation and termination of signalling. While STING is known to be degraded within lysosomes, the mechanisms controlling its delivery remain poorly defined. Here we utilised a proteomics-based approach to assess phosphorylation changes in primary murine macrophages following STING activation. This identified numerous phosphorylation events in proteins involved in intracellular and vesicular transport. We utilised high-temporal microscopy to track STING vesicular transport in live macrophages. We subsequently identified that the endosomal complexes required for transport (ESCRT) pathway detects ubiquitinated STING on vesicles, which facilitates the degradation of STING in murine macrophages. Disruption of ESCRT functionality greatly enhanced STING signalling and cytokine production, thus characterising a mechanism controlling effective termination of STING signalling.
Collapse
Affiliation(s)
- Katherine R Balka
- Department of Biochemistry and Molecular Biology, Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Vic., Australia
| | - Rajan Venkatraman
- Department of Biochemistry and Molecular Biology, Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Vic., Australia
| | - Tahnee L Saunders
- Ubiquitin Signalling Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Vic., Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Vic., Australia
| | - Angus Shoppee
- Department of Biochemistry and Molecular Biology, Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Vic., Australia
- Department of Biochemistry and Molecular Biology, Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Vic., Australia
| | - Ee Shan Pang
- Department of Biochemistry and Molecular Biology, Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Vic., Australia
- Department of Biochemistry and Molecular Biology, Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Vic., Australia
| | - Zoe Magill
- Department of Biochemistry and Molecular Biology, Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Vic., Australia
- Department of Biochemistry and Molecular Biology, Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Vic., Australia
| | - Jihane Homman-Ludiye
- Monash Micro Imaging, Biomedicine Discovery Institute, Monash University, Clayton, Vic., Australia
| | - Cheng Huang
- Department of Biochemistry and Molecular Biology, Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Vic., Australia
- Monash Proteomics and Metabolomics Facility, Monash University, Clayton, Vic., Australia
| | - Rachael M Lane
- Department of Biochemistry and Molecular Biology, Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Vic., Australia
| | - Harrison M York
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, Vic., Australia
| | - Peck Tan
- Department of Biochemistry and Molecular Biology, Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Vic., Australia
- Department of Biochemistry and Molecular Biology, Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Vic., Australia
| | - Ralf B Schittenhelm
- Department of Biochemistry and Molecular Biology, Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Vic., Australia
- Monash Proteomics and Metabolomics Facility, Monash University, Clayton, Vic., Australia
| | - Senthil Arumugam
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, Vic., Australia
- European Molecular Biological Laboratory Australia (EMBL Australia), Monash University, Clayton/Melbourne, Vic., Australia
| | - Benjamin T Kile
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Meredith O'Keeffe
- Department of Biochemistry and Molecular Biology, Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Vic., Australia
- Department of Biochemistry and Molecular Biology, Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Vic., Australia
| | - Dominic De Nardo
- Department of Biochemistry and Molecular Biology, Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Vic., Australia
| |
Collapse
|
30
|
Yan J, Qiao G, Yin Y, Wang E, Xiao J, Peng Y, Yu J, Du Y, Li Z, Wu H, Liu M, Tu J, Zhang Y, Feng H. Black carp RNF5 inhibits STING/IFN signaling through promoting K48-linked ubiquitination and degradation of STING. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 145:104712. [PMID: 37100266 DOI: 10.1016/j.dci.2023.104712] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/15/2023] [Accepted: 04/16/2023] [Indexed: 05/09/2023]
Abstract
Ubiquitination is one of the important post-translational modifications (PTMs) of proteins that plays a vital role in regulating substrate degradation to ensure cellular homeostasis. Ring finger protein 5 (RNF5) is an essential E3 ubiquitin ligase for inhibiting STING-mediated interferon (IFN) signaling in mammals. Nevertheless, the function of RNF5 in STING/IFN pathway remains obscure in teleost. Here, we reported that over-expression of black carp RNF5 (bcRNF5) inhibited STING-mediated transcription activity of bcIFNa, DrIFNφ1, NF-κB and ISRE promoters and antiviral activity against SVCV. Moreover, knockdown of bcRNF5 increased the expression of host genes, including bcIFNa, bcIFNb, bcILβ, bcMX1 and bcViperin, and also enhanced the antiviral capability of host cells. Immunofluorescence (IF) and Co-immunoprecipitation (Co-IP) assay confirmed that bcRNF5 was mainly localized in the cytoplasm and interacted with bcSTING. The expression level of bcSTING protein was attenuated by co-expressed bcRNF5 and MG132 treatment rescued this attenuating effect, suggesting that bcRNF5-mediated bcSTING degradation was dependent on the proteasome pathway. Subsequent, Co-IP and immunoblot (IB) experiments identified that bcRNF5 triggered the K48-linked but not K63-linked ubiquitination of bcSTING. Altogether, above results conclude that RNF5 suppresses STING/IFN signaling by enhancing K48-linked ubiquitination and protease degradation of STING in black carp.
Collapse
Affiliation(s)
- Jun Yan
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China; College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, China
| | - Guoxia Qiao
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Yuqi Yin
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Enhui Wang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Jun Xiao
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China.
| | - Yuqing Peng
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Jiamin Yu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Yuting Du
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Zhiming Li
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Hui Wu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Meiling Liu
- College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, China
| | - Jiagang Tu
- College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Yongan Zhang
- College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Hao Feng
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China.
| |
Collapse
|
31
|
MacLauchlan S, Kushwaha P, Tai A, Chen J, Manning C, Swarnkar G, Abu-Amer Y, Fitzgerald KA, Sharma S, Gravallese EM. STING-dependent interferon signatures restrict osteoclast differentiation and bone loss in mice. Proc Natl Acad Sci U S A 2023; 120:e2210409120. [PMID: 37023130 PMCID: PMC10104545 DOI: 10.1073/pnas.2210409120] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 02/14/2023] [Indexed: 04/07/2023] Open
Abstract
Stimulator of interferon genes (STING) is a key mediator of type-I interferon (IFN-I) signaling in response to a variety of stimuli, but the contribution of STING to homeostatic processes is not fully characterized. Previous studies showed that ligand activation of STING limits osteoclast differentiation in vitro through the induction of IFNβ and IFN-I interferon-stimulated genes (ISGs). In a disease model (SAVI) driven by the V154M gain-of-function mutation in STING, fewer osteoclasts form from SAVI precursors in response to receptor activator of NF-kappaB ligand (RANKL) in an IFN-I-dependent manner. Due to the described role of STING-mediated regulation of osteoclastogenesis in activation settings, we sought to determine whether basal STING signaling contributes to bone homeostasis, an unexplored area. Using whole-body and myeloid-specific deficiency, we show that STING signaling prevents trabecular bone loss in mice over time and that myeloid-restricted STING activity is sufficient for this effect. STING-deficient osteoclast precursors differentiate with greater efficiency than wild types. RNA sequencing of wild-type and STING-deficient osteoclast precursor cells and differentiating osteoclasts reveals unique clusters of ISGs including a previously undescribed ISG set expressed in RANKL naïve precursors (tonic expression) and down-regulated during differentiation. We identify a 50 gene tonic ISG signature that is STING dependent and shapes osteoclast differentiation. From this list, we identify interferon-stimulated gene 15 (ISG15) as a tonic STING-regulated ISG that limits osteoclast formation. Thus, STING is an important upstream regulator of tonic IFN-I signatures shaping the commitment to osteoclast fates, providing evidence for a nuanced and unique role for this pathway in bone homeostasis.
Collapse
Affiliation(s)
- Susan MacLauchlan
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Priyanka Kushwaha
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Albert Tai
- Department of Immunology, Tufts University School of Medicine, Boston, MA02111
| | - Jia Chen
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Catherine Manning
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Gaurav Swarnkar
- Department of Orthopedics and Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO63110
| | - Yousef Abu-Amer
- Department of Orthopedics and Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO63110
| | - Katherine A. Fitzgerald
- Department of Medicine, Program in Innate Immunity, University of Massachusetts Chan Medical School, Worcester, MA01655
| | - Shruti Sharma
- Department of Immunology, Tufts University School of Medicine, Boston, MA02111
| | - Ellen M. Gravallese
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA02115
| |
Collapse
|
32
|
Chen NN, Zhang H, Zhu QS, Zeng T, Dai W, Zhou YL, Xin GF, Wu BD, Gong SJ, Jiang ZY, You QD, Xu XL. Development of Orally Bioavailable Amidobenzimidazole Analogues Targeting Stimulator of Interferon Gene (STING) Receptor. J Med Chem 2023; 66:5584-5610. [PMID: 37027512 DOI: 10.1021/acs.jmedchem.2c02046] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
Abstract
Stimulator of interferon gene (STING) is a critical adaptor protein that has a pivotal role in triggering inherent immune responses to infection. STING-linked interferon production has been involved in anti-inflammation, anti-infection, and antitumor immunity. Herein, a series of amidobenzimidazole analogues as STING agonists were profiled for potency and drug-like properties. By structure-based modification and optimization based on mono-aminobenzimidazole (ABZI), analogues with nanomolar STING agonistic activities were obtained. Among them, compounds D59 and D61 significantly increased the transcription of IFN-β and proinflammatory cytokine CXCL10, as well as dramatically induced the phosphorylation of STING downstream proteins in THP1 cells. Furthermore, compound D61 exhibited favorable pharmacokinetic properties and metabolic stabilities. In a CT-26 syngeneic mice-bearing tumor model, D61 effectively inhibited tumor growth with good tolerance when administered via intratumoral, intravenous, intraperitoneal, and oral routes. This research on orally bioavailable amidobenzimidazole analogues expands the diversity of chemical structures of agonists for STING-mediated immunotherapy.
Collapse
Affiliation(s)
- Nan-Nan Chen
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Han Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qiang-Sheng Zhu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Ting Zeng
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Wei Dai
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Ye-Ling Zhou
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Guo-Feng Xin
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Bei-Duo Wu
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Si-Jia Gong
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zheng-Yu Jiang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qi-Dong You
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiao-Li Xu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
33
|
Song J, Yang RR, Chang J, Liu YD, Lu CH, Chen LF, Guo H, Zhang YH, Fan ZS, Zhou JY, Zhou GZ, Zhang KK, Luo XM, Chen KX, Jiang HL, Zhang SL, Zheng MY. Discovery and characterization of a novel cGAS covalent inhibitor for the treatment of inflammatory bowel disease. Acta Pharmacol Sin 2023; 44:791-800. [PMID: 36229599 PMCID: PMC10043009 DOI: 10.1038/s41401-022-01002-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 09/19/2022] [Indexed: 11/09/2022]
Abstract
Cyclic GMP-AMP synthase (cGAS), a cytosolic DNA sensor, acts as a nucleotidyl transferase that catalyzes ATP and GTP to form cyclic GMP-AMP (cGAMP) and plays a critical role in innate immunity. Hyperactivation of cGAS-STING signaling contributes to hyperinflammatory responses. Therefore, cGAS is considered a promising target for the treatment of inflammatory diseases. Herein, we report the discovery and identification of several novel types of cGAS inhibitors by pyrophosphatase (PPiase)-coupled activity assays. Among these inhibitors, 1-(1-phenyl-3,4-dihydro-1H-pyrrolo[1,2-a]pyrazin-2-yl)prop-2-yn-1-one (compound 3) displayed the highest potency and selectivity at the cellular level. Compound 3 exhibited better inhibitory activity and pathway selectivity than RU.521, which is a selective cGAS inhibitor with anti-inflammatory effects in vitro and in vivo. Thermostability analysis, nuclear magnetic resonance and isothermal titration calorimetry assays confirmed that compound 3 directly binds to the cGAS protein. Mass spectrometry and mutation analysis revealed that compound 3 covalently binds to Cys419 of cGAS. Notably, compound 3 demonstrated promising therapeutic efficacy in a dextran sulfate sodium (DSS)-induced mouse colitis model. These results collectively suggest that compound 3 will be useful for understanding the biological function of cGAS and has the potential to be further developed for inflammatory disease therapies.
Collapse
Affiliation(s)
- Jia Song
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Rui-Rui Yang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Shanghai Institute for Advanced Immunochemical Studies, and School of Life Science and Technology, Shanghai Tech University, Shanghai, 200031, China
| | - Jie Chang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Ya-Dan Liu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Cheng-Hao Lu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Li-Fan Chen
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hao Guo
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ying-Hui Zhang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zi-Sheng Fan
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jing-Yi Zhou
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Gui-Zhen Zhou
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ke-Ke Zhang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xiao-Min Luo
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Kai-Xian Chen
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Hua-Liang Jiang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China.
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Shanghai Institute for Advanced Immunochemical Studies, and School of Life Science and Technology, Shanghai Tech University, Shanghai, 200031, China.
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Su-Lin Zhang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Ming-Yue Zheng
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
34
|
Abstract
The discovery of cGAMP in 2012 filled an important gap in our understanding of innate immune signaling. It has been known for over a century that DNA can induce immune responses, but the underlying mechanism was not clear. With the identification of STING as a key player in interferon induction, the DNA detector that activates STING was the last missing link in TBK1-IRF3 signaling. Somewhat unexpectedly, it turns out that nature relays the DNA danger signal through a small molecule. cGAMP is a cyclic dinucleotide produced from cyclodimerization of ATP and GTP upon detection of cytosolic DNA by cGAS, a previously uncharacterized protein, to promote the assembly of the STING signalosome. This article covers a personal account of the discovery of cGAMP, a short history of the relevant nucleotide chemistry, and a summary of the latest development in this field of research in chemistry. It is the author's hope that, with a historic perspective, the readers can better appreciate the synergy between chemistry and biology in drug development.
Collapse
Affiliation(s)
- Chuo Chen
- Department of Biochemistry, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9038, USA
| |
Collapse
|
35
|
Myristic acid as a checkpoint to regulate STING-dependent autophagy and interferon responses by promoting N-myristoylation. Nat Commun 2023; 14:660. [PMID: 36750575 PMCID: PMC9905541 DOI: 10.1038/s41467-023-36332-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 01/23/2023] [Indexed: 02/09/2023] Open
Abstract
Stimulator of interferon gene (STING)-triggered autophagy is crucial for the host to eliminate invading pathogens and serves as a self-limiting mechanism of STING-induced interferon (IFN) responses. Thus, the mechanisms that ensure the beneficial effects of STING activation are of particular importance. Herein, we show that myristic acid, a type of long-chain saturated fatty acid (SFA), specifically attenuates cGAS-STING-induced IFN responses in macrophages, while enhancing STING-dependent autophagy. Myristic acid inhibits HSV-1 infection-induced innate antiviral immune responses and promotes HSV-1 replication in mice in vivo. Mechanistically, myristic acid enhances N-myristoylation of ARF1, a master regulator that controls STING membrane trafficking. Consequently, myristic acid facilitates STING activation-triggered autophagy degradation of the STING complex. Thus, our work identifies myristic acid as a metabolic checkpoint that contributes to immune homeostasis by balancing STING-dependent autophagy and IFN responses. This suggests that myristic acid and N-myristoylation are promising targets for the treatment of diseases caused by aberrant STING activation.
Collapse
|
36
|
Wang X, Yang C, Wang X, Miao J, Chen W, Zhou Y, Xu Y, An Y, Cheng A, Ye W, Chen M, Song D, Yuan X, Wang J, Qian P, Ruohao Wu A, Zhang ZY, Liu K. Driving axon regeneration by orchestrating neuronal and non-neuronal innate immune responses via the IFNγ-cGAS-STING axis. Neuron 2023; 111:236-255.e7. [PMID: 36370710 PMCID: PMC9851977 DOI: 10.1016/j.neuron.2022.10.028] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 09/20/2022] [Accepted: 10/18/2022] [Indexed: 11/13/2022]
Abstract
The coordination mechanism of neural innate immune responses for axon regeneration is not well understood. Here, we showed that neuronal deletion of protein tyrosine phosphatase non-receptor type 2 sustains the IFNγ-STAT1 activity in retinal ganglion cells (RGCs) to promote axon regeneration after injury, independent of mTOR or STAT3. DNA-damage-induced cGAMP synthase (cGAS)-stimulator of interferon genes (STINGs) activation is the functional downstream signaling. Directly activating neuronal STING by cGAMP promotes axon regeneration. In contrast to the central axons, IFNγ is locally translated in the injured peripheral axons and upregulates cGAS expression in Schwann cells and infiltrating blood cells to produce cGAMP, which promotes spontaneous axon regeneration as an immunotransmitter. Our study demonstrates that injured peripheral nervous system (PNS) axons can direct the environmental innate immune response for self-repair and that the neural antiviral mechanism can be harnessed to promote axon regeneration in the central nervous system (CNS).
Collapse
Affiliation(s)
- Xu Wang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China,Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China,Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China,Biomedical Research Institute, Shenzhen Peking University–The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China,Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, Guangdong 518057, China,Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou 510515, China,These authors contributed equally
| | - Chao Yang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China,Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China,Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China,Biomedical Research Institute, Shenzhen Peking University–The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China,Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, Guangdong 518057, China,Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou 510515, China,These authors contributed equally
| | - Xuejie Wang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China,Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Jinmin Miao
- Department of Medicinal Chemistry and Molecular Pharmacology, Department of Chemistry, Center for Cancer Research and Institute for Drug Discovery, Purdue University, West Lafayette, IN, USA
| | - Weitao Chen
- Biomedical Research Institute, Shenzhen Peking University–The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
| | - Yiren Zhou
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Ying Xu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China,Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yongyan An
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Aifang Cheng
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China,Department of Ocean Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Wenkang Ye
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China,Department of Ocean Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Mengxian Chen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Dong Song
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China,Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Xue Yuan
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Jiguang Wang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China,Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Peiyuan Qian
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China,Department of Ocean Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Angela Ruohao Wu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China,Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China,Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong, China,Center for Aging Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Zhong-Yin Zhang
- Department of Medicinal Chemistry and Molecular Pharmacology, Department of Chemistry, Center for Cancer Research and Institute for Drug Discovery, Purdue University, West Lafayette, IN, USA
| | - Kai Liu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China; Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China; Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China; Biomedical Research Institute, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China; Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, Guangdong 518057, China; Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou 510515, China.
| |
Collapse
|
37
|
Gao Z, Gao Z, Zhang H, Hou S, Zhou Y, Liu X. Targeting STING: From antiviral immunity to treat osteoporosis. Front Immunol 2023; 13:1095577. [PMID: 36741390 PMCID: PMC9891206 DOI: 10.3389/fimmu.2022.1095577] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 12/30/2022] [Indexed: 01/19/2023] Open
Abstract
The cGAS-STING signaling pathway can trigger innate immune responses by detecting dsDNA from outside or within the host. In addition, the cGAS-STING signaling pathway has emerged as a critical mediator of the inflammatory response and a new target for inflammatory diseases. STING activation leads to dimerization and translocation to the endoplasmic reticulum Golgi intermediate compartment or Golgi apparatus catalyzed by TBK1, triggers the production of IRF3 and NF-κB and translocates to the nucleus to induce a subsequent interferon response and pro-inflammatory factor production. Osteoporosis is a degenerative bone metabolic disease accompanied by chronic sterile inflammation. Activating the STING/IFN-β signaling pathway can reduce bone resorption by inhibiting osteoclast differentiation. Conversely, activation of STING/NF-κB leads to the formation of osteoporosis by increasing bone resorption and decreasing bone formation. In addition, activation of STING inhibits the generation of type H vessels with the capacity to osteogenesis, thereby inhibiting bone formation. Here, we outline the mechanism of action of STING and its downstream in osteoporosis and discuss the role of targeting STING in the treatment of osteoporosis, thus providing new ideas for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Zhonghua Gao
- Department of Geriatrics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhongguo Gao
- Department of Medical Laboratory Technology, School of Biomedical Engineering, Hubei University of Medicine, Shiyan, Hubei, China
| | - Hao Zhang
- Department of Geriatrics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shoubo Hou
- Department of General Practice, General Hospital of Central Theater Command, Wuhan, Hubei, China
| | - Yunhua Zhou
- Department of Wound Repair Surgery, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China,*Correspondence: Yunhua Zhou, ; Xiangjie Liu,
| | - Xiangjie Liu
- Department of Geriatrics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China,*Correspondence: Yunhua Zhou, ; Xiangjie Liu,
| |
Collapse
|
38
|
Yang J, Ding H, Shuai B, Zhang Y, Zhang Y. Mechanism and effects of STING-IFN-I pathway on nociception: A narrative review. Front Mol Neurosci 2023; 15:1081288. [PMID: 36683857 PMCID: PMC9846240 DOI: 10.3389/fnmol.2022.1081288] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/05/2022] [Indexed: 01/05/2023] Open
Abstract
Since the discovery of STING in 2008, numerous studies have investigated its functions in immunity, inflammation, and cancer. STING activates downstream molecules including IFN-I, NLRP3, and NF-κB. The STING-IFN-I pathway plays a vital role in nociception. After receiving the upstream signal, STING is activated and induces the expression of IFN-I, and after paracrine and autocrine signaling, IFN-I binds to IFN receptors. Subsequently, the activity of ion channels is inhibited by TYK2, which induces an acute antinociceptive effect. JAK activates PIK3 and MAPK-MNK-eIF4E pathways, which sensitize nociceptors in the peripheral nervous system. In the mid-late stage, the STING-IFN-I pathway activates STAT, increases pro-inflammatory and anti-inflammatory cytokines, inhibits ER-phagy, and promotes microglial M1-polarization in the central nervous system, leading to central sensitization. Thus, the STING-IFN-I pathway may exert complex effects on nociception at various stages, and these effects require further comprehensive elucidation. Therefore, in this review, we systematically summarized the mechanisms of the STING-IFN-I pathway and discussed its function in nociception.
Collapse
Affiliation(s)
- Jinghan Yang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Ding
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Shuai
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Zhang
- Department of Pain, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,*Correspondence: Yan Zhang,
| | - Yan Zhang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
39
|
Villemin C, Six A, Neville BA, Lawley TD, Robinson MJ, Bakdash G. The heightened importance of the microbiome in cancer immunotherapy. Trends Immunol 2023; 44:44-59. [PMID: 36464584 DOI: 10.1016/j.it.2022.11.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/28/2022] [Accepted: 11/08/2022] [Indexed: 12/03/2022]
Abstract
The human microbiome is recognized as a key factor in health and disease. This has been further corroborated by identifying changes in microbiome composition and function as a novel hallmark in cancer. These effects are exerted through microbiome interactions with host cells, impacting a wide variety of developmental and physiological processes. In this review, we discuss some of the latest findings on how the bacterial component of the microbiome can influence outcomes for different cancer immunotherapy modalities, highlighting identified mechanisms of action. We also address the clinical efforts to utilize this knowledge to achieve better responses to immunotherapy. A refined understanding of microbiome variations in patients and microbiome-host interactions with cancer therapies is essential to realize optimal clinical responses.
Collapse
Affiliation(s)
| | - Anne Six
- Microbiotica Ltd., Cambridge, UK
| | | | - Trevor D Lawley
- Microbiotica Ltd., Cambridge, UK; Host-Microbiota Interactions Laboratory, Wellcome Sanger Institute, Hinxton, UK
| | | | | |
Collapse
|
40
|
Zhang Z, Zhou H, Ouyang X, Dong Y, Sarapultsev A, Luo S, Hu D. Multifaceted functions of STING in human health and disease: from molecular mechanism to targeted strategy. Signal Transduct Target Ther 2022; 7:394. [PMID: 36550103 PMCID: PMC9780328 DOI: 10.1038/s41392-022-01252-z] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 10/25/2022] [Accepted: 11/09/2022] [Indexed: 12/24/2022] Open
Abstract
Since the discovery of Stimulator of Interferon Genes (STING) as an important pivot for cytosolic DNA sensation and interferon (IFN) induction, intensive efforts have been endeavored to clarify the molecular mechanism of its activation, its physiological function as a ubiquitously expressed protein, and to explore its potential as a therapeutic target in a wide range of immune-related diseases. With its orthodox ligand 2'3'-cyclic GMP-AMP (2'3'-cGAMP) and the upstream sensor 2'3'-cGAMP synthase (cGAS) to be found, STING acquires its central functionality in the best-studied signaling cascade, namely the cGAS-STING-IFN pathway. However, recently updated research through structural research, genetic screening, and biochemical assay greatly extends the current knowledge of STING biology. A second ligand pocket was recently discovered in the transmembrane domain for a synthetic agonist. On its downstream outputs, accumulating studies sketch primordial and multifaceted roles of STING beyond its cytokine-inducing function, such as autophagy, cell death, metabolic modulation, endoplasmic reticulum (ER) stress, and RNA virus restriction. Furthermore, with the expansion of the STING interactome, the details of STING trafficking also get clearer. After retrospecting the brief history of viral interference and the milestone events since the discovery of STING, we present a vivid panorama of STING biology taking into account the details of the biochemical assay and structural information, especially its versatile outputs and functions beyond IFN induction. We also summarize the roles of STING in the pathogenesis of various diseases and highlight the development of small-molecular compounds targeting STING for disease treatment in combination with the latest research. Finally, we discuss the open questions imperative to answer.
Collapse
Affiliation(s)
- Zili Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Haifeng Zhou
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Xiaohu Ouyang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Yalan Dong
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
| | - Alexey Sarapultsev
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049, Ekaterinburg, Russia
| | - Shanshan Luo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Desheng Hu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
- Key Laboratory of Biological Targeted Therapy, The Ministry of Education, 430022, Wuhan, China.
- Clinical Research Center of Cancer Immunotherapy, 430022, Hubei, Wuhan, China.
| |
Collapse
|
41
|
Structural insights into a shared mechanism of human STING activation by a potent agonist and an autoimmune disease-associated mutation. Cell Discov 2022; 8:133. [PMID: 36513640 DOI: 10.1038/s41421-022-00481-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 09/23/2022] [Indexed: 12/14/2022] Open
Abstract
Stimulator of interferon gene (STING) is increasingly exploited for the potential in cancer immunotherapy, yet its mechanism of activation remains not fully understood. Herein, we designed a novel STING agonist, designated as HB3089 that exhibits robust and durable anti-tumor activity in tumor models across various cancer types. Cryo-EM analysis reveals that HB3089-bound human STING has structural changes similar to that of the STING mutant V147L, a constitutively activated mutant identified in patients with STING-associated vasculopathy with onset in infancy (SAVI). Both structures highlight the conformational changes of the transmembrane domain (TMD), but without the 180°-rotation of the ligand binding domain (LBD) previously shown to be required for STING activation. Further structure-based functional analysis confirmed a new STING activation mode shared by the agonist and the SAVI-related mutation, in which the connector linking the LBD and the TMD senses the activation signal and controls the conformational changes of the LBD and the TMD for STING activation. Together, our findings lead to a new working model for STING activation and open a new avenue for the rationale design of STING-targeted therapies either for cancer or autoimmune disorders.
Collapse
|
42
|
The Relationship between Reactive Oxygen Species and the cGAS/STING Signaling Pathway in the Inflammaging Process. Int J Mol Sci 2022; 23:ijms232315182. [PMID: 36499506 PMCID: PMC9735967 DOI: 10.3390/ijms232315182] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/08/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
During Inflammaging, a dysregulation of the immune cell functions is generated, and these cells acquire a senescent phenotype with an increase in pro-inflammatory cytokines and ROS. This increase in pro-inflammatory molecules contributes to the chronic inflammation and oxidative damage of biomolecules, classically observed in the Inflammaging process. One of the most critical oxidative damages is generated to the host DNA. Damaged DNA is located out of the natural compartments, such as the nucleus and mitochondria, and is present in the cell's cytoplasm. This DNA localization activates some DNA sensors, such as the cGAS/STING signaling pathway, that induce transcriptional factors involved in increasing inflammatory molecules. Some of the targets of this signaling pathway are the SASPs. SASPs are secreted pro-inflammatory molecules characteristic of the senescent cells and inducers of ROS production. It has been suggested that oxidative damage to nuclear and mitochondrial DNA generates activation of the cGAS/STING pathway, increasing ROS levels induced by SASPs. These additional ROS increase oxidative DNA damage, causing a loop during the Inflammaging. However, the relationship between the cGAS/STING pathway and the increase in ROS during Inflammaging has not been clarified. This review attempt to describe the potential connection between the cGAS/STING pathway and ROS during the Inflammaging process, based on the current literature, as a contribution to the knowledge of the molecular mechanisms that occur and contribute to the development of the considered adaptative Inflammaging process during aging.
Collapse
|
43
|
Lv X, Xin S, Zheng W, Xu T, Sun Y. microRNA-27c negatively regulates NF-κB and IRF3 signaling pathway via targeting MITA in miiuy croaker. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 137:104522. [PMID: 36049570 DOI: 10.1016/j.dci.2022.104522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/13/2022] [Accepted: 08/24/2022] [Indexed: 06/15/2023]
Abstract
As a non-coding RNA with regulatory functions, microRNAs(miRNAs) can regulate gene expression and participate in a variety of physiological and pathological processes. In recent years, although there have been many studies on miRNA, the regulation mechanisms of miRNA in teleost fish have not been fully elucidated. In this study, it was first predicted that MITA is the target of miR-27c through bioinformatics, and it was confirmed by dual fluorescence experiments. Then we found that miR-27c can inhibit the expression of MITA at the mRNA and protein levels, thereby promoting the NF-κB or IRF3 pathway. It is speculated that miR-27c plays an important role in the innate immunity of teleost fish. This study will help to further understand miRNAs regulatory mechanism in teleost fish.
Collapse
Affiliation(s)
- Xing Lv
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Shiying Xin
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Weiwei Zheng
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Tianjun Xu
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China; Laboratory of Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education, 201306, China; National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, 201306, China.
| | - Yuena Sun
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education, 201306, China; National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, 201306, China.
| |
Collapse
|
44
|
Mansouri S, Gogoi H, Patel S, Katikaneni DS, Singh A, Aybar-Torres A, de Lartigue G, Jin L. MPYS Modulates Fatty Acid Metabolism and Immune Tolerance at Homeostasis Independent of Type I IFNs. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:2114-2132. [PMID: 36261171 PMCID: PMC9679991 DOI: 10.4049/jimmunol.2200158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/15/2022] [Indexed: 01/04/2023]
Abstract
MPYS/STING (stimulator of IFN genes) senses cyclic dinucleotides (CDNs), generates type I IFNs, and plays a critical role in infection, inflammation, and cancer. In this study, analyzing genotype and haplotype data from the 1000 Genomes Project, we found that the R71H-G230A-R293Q (HAQ) MPYS allele frequency increased 57-fold in East Asians compared with sub-Saharan Africans. Meanwhile, the G230A-R293Q (AQ) allele frequency decreased by 98% in East Asians compared with sub-Saharan Africans. We propose that the HAQ and AQ alleles underwent a natural selection during the out-of-Africa migration. We used mouse models of HAQ and AQ to investigate the underlying mechanism. We found that the mice carrying the AQ allele, which disappeared in East Asians, had normal CDN-type I IFN responses. Adult AQ mice, however, had less fat mass than did HAQ or wild-type mice on a chow diet. AQ epididymal adipose tissue had increased regulatory T cells and M2 macrophages with protein expression associated with enhanced fatty acid oxidation. Conditional knockout mice and adoptive cell transfer indicate a macrophage and regulatory T cell-intrinsic role of MPYS in fatty acid metabolism. Mechanistically, AQ/IFNAR1-/- mice had a similar lean phenotype as for the AQ mice. MPYS intrinsic tryptophan fluorescence revealed that the R71H change increased MPYS hydrophilicity. Lastly, we found that the second transmembrane (TM) and the TM2-TM3 linker region of MPYS interact with activated fatty acid, fatty acyl-CoA. In summary, studying the evolution of the human MPYS gene revealed an MPYS function in modulating fatty acid metabolism that may be critical during the out-of-Africa migration.
Collapse
Affiliation(s)
- Samira Mansouri
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL
| | - Himanshu Gogoi
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL
| | - Seema Patel
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL
| | - Divya S. Katikaneni
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL
| | - Arashdeep Singh
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL; and
- Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, FL
| | - Alexandra Aybar-Torres
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL
| | - Guillaume de Lartigue
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL; and
- Center for Integrative Cardiovascular and Metabolic Disease, University of Florida, Gainesville, FL
| | - Lei Jin
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL
| |
Collapse
|
45
|
Patil V, Hernandez-Franco JF, Yadagiri G, Bugybayeva D, Dolatyabi S, Feliciano-Ruiz N, Schrock J, Hanson J, Ngunjiri J, HogenEsch H, Renukaradhya GJ. A split influenza vaccine formulated with a combination adjuvant composed of alpha-D-glucan nanoparticles and a STING agonist elicits cross-protective immunity in pigs. J Nanobiotechnology 2022; 20:477. [PMID: 36369044 PMCID: PMC9652892 DOI: 10.1186/s12951-022-01677-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 10/16/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Swine influenza A viruses (SwIAVs) pose an economic and pandemic threat, and development of novel effective vaccines is of critical significance. We evaluated the performance of split swine influenza A virus (SwIAV) H1N2 antigens with a plant-derived nanoparticle adjuvant alone (Nano-11) [Nano11-SwIAV] or in combination with the synthetic stimulator of interferon genes (STING) agonist ADU-S100 (NanoS100-SwIAV). Specific pathogen free (SPF) pigs were vaccinated twice via intramuscular (IM) or intradermal (ID) routes and challenged with a virulent heterologous SwIAV H1N1-OH7 virus. RESULTS Animals vaccinated IM or ID with NanoS100-SwIAV had significantly increased cross-reactive IgG and IgA titers in serum, nasal secretion and bronchoalveolar lavage fluid at day post challenge 6 (DPC6). Furthermore, NanoS100-SwIAV ID vaccinates, even at half the vaccine dose compared to their IM vaccinated counterparts, had significantly increased frequencies of CXCL10+ myeloid cells in the tracheobronchial lymph nodes (TBLN), and IFNγ+ effector memory T-helper/memory cells, IL-17A+ total T-helper/memory cells, central and effector memory T-helper/memory cells, IL-17A+ total cytotoxic T-lymphocytes (CTLs), and early effector CTLs in blood compared with the Nano11-SwIAV group demonstrating a potential dose-sparing effect and induction of a strong IL-17A+ T-helper/memory (Th17) response in the periphery. However, the frequencies of IFNγ+ late effector CTLs and effector memory T-helper/memory cells, IL-17A+ total CTLs, late effector CTLs, and CXCL10+ myeloid cells in blood, as well as lung CXCL10+ plasmacytoid dendritic cells were increased in NanoS100-SwIAV IM vaccinated pigs. Increased expression of IL-4 and IL-6 mRNA was observed in TBLN of Nano-11 based IM vaccinates following challenge. Furthermore, the challenge virus load in the lungs and nasal passage was undetectable in NanoS100-SwIAV IM vaccinates by DPC6 along with reduced macroscopic lung lesions and significantly higher virus neutralization titers in lungs at DPC6. However, NanoS100-SwIAV ID vaccinates exhibited significant reduction of challenge virus titers in nasal passages and a remarkable reduction of challenge virus in lungs. CONCLUSIONS Despite vast genetic difference (77% HA gene identity) between the H1N2 and H1N1 SwIAV, the NanoS100 adjuvanted vaccine elicited cross protective cell mediated immune responses, suggesting the potential role of this combination adjuvant in inducing cross-protective immunity in pigs.
Collapse
Affiliation(s)
- V. Patil
- grid.261331.40000 0001 2285 7943Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - J. F. Hernandez-Franco
- grid.169077.e0000 0004 1937 2197Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN USA
| | - G. Yadagiri
- grid.261331.40000 0001 2285 7943Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - D. Bugybayeva
- grid.261331.40000 0001 2285 7943Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA ,International Center for Vaccinology, Kazakh National Agrarian Research University (KazNARU), Almaty, Kazakhstan
| | - S. Dolatyabi
- grid.261331.40000 0001 2285 7943Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - N. Feliciano-Ruiz
- grid.261331.40000 0001 2285 7943Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - J. Schrock
- grid.261331.40000 0001 2285 7943Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - J. Hanson
- grid.261331.40000 0001 2285 7943Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - J. Ngunjiri
- grid.261331.40000 0001 2285 7943Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| | - H. HogenEsch
- grid.169077.e0000 0004 1937 2197Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN USA
| | - G. J. Renukaradhya
- grid.261331.40000 0001 2285 7943Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691 USA
| |
Collapse
|
46
|
Li Y, Qiao X, Hou L, Liu X, Li Q, Jin Y, Li Y, Wang L, Song L. A stimulator of interferon gene (CgSTING) involved in antimicrobial immune response of oyster Crassostrea gigas. FISH & SHELLFISH IMMUNOLOGY 2022; 128:82-90. [PMID: 35917891 DOI: 10.1016/j.fsi.2022.07.059] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/17/2022] [Accepted: 07/19/2022] [Indexed: 06/15/2023]
Abstract
The stimulator of interferon gene (STING), an intracellular sensor of cyclic dinucleotides, is critical to the innate immune response, especially the induction of type I interferon (IFN) during pathogenic infection. A STING homologue (CgSTING) regulating the expression of IFN-like protein (CgIFNLP) was previously identified in the Pacific oyster Crassostrea gigas, and its involvement in antibacterial immunity was further investigated in the present study. The mRNA transcripts of CgSTING were ubiquitously detected in all the three subpopulations of haemocytes with the highest expression in semi-granulocytes. After the stimulation with Vibrio splendidus, the mRNA expression of CgSTING in haemocytes was significantly up-regulated and peaked at 72 h, which was 12.91-fold of that in control group (p < 0.01). The CgSTING protein was mainly located in the cytoplasm of haemocytes. After the expression of CgSTING was knocked down (0.12-fold of that in control group, p < 0.05) by RNAi, the mRNA expression levels of interleukin17-1 (CgIL17-1), interleukin17-3 (CgIL17-3), interleukin17-4 (CgIL17-4), defensins (Cgdefh1, Cgdefh2), big defensin (CgBigDef1), interferon-like protein (CgIFNLP), tumor necrosis factor (CgTNF) and nuclear factor-κB (CgRel) all decreased significantly at 12 h after V. splendidus stimulation, which was 0.12-fold-0.72-fold (p < 0.05) of that in control group, respectively. The positive signals of CgRel were observed in the haemocyte nucleus after V. splendidus stimulation. The nuclear translocation of CgRel was suppressed in CgSTING-RNAi oysters, and the green signals of CgRel were mainly observed in the haemocyte cytoplasm after V. splendidus stimulation. Furthermore, the number of V. splendidus in the haemolymph of CgSTING-RNAi oysters increased significantly, which was 26.78-fold (p < 0.01) of that in the control group at 12 h after V. splendidus stimulation. These results indicated that CgSTING played important role in the immune defense against bacterial infection by inducing the expressions of cytokines and defensins.
Collapse
Affiliation(s)
- Youjing Li
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Functional Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Xue Qiao
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Functional Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Lilin Hou
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Functional Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Xiyang Liu
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Functional Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Qing Li
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Functional Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - YuHao Jin
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Functional Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Yinan Li
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Functional Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China
| | - Lingling Wang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Southern Laboratory of Ocean Science and Engineering (Guangdong, Zhuhai), Zhuhai, 519000, China; Functional Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Linsheng Song
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Southern Laboratory of Ocean Science and Engineering (Guangdong, Zhuhai), Zhuhai, 519000, China; Functional Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China.
| |
Collapse
|
47
|
Zhang Q, Chen C, Xia B, Xu P. Chemical regulation of the cGAS-STING pathway. Curr Opin Chem Biol 2022; 69:102170. [PMID: 35753220 DOI: 10.1016/j.cbpa.2022.102170] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/05/2022] [Accepted: 05/10/2022] [Indexed: 11/25/2022]
Abstract
Nucleic acids represent a major class of pathogen and damage signatures, recognized by a variety of host sensors to initiate signaling cascades and immune responses, such as mechanisms of RLR-MAVS, cGAS-STING, TLR-TRIF, and AIM2 inflammasome. Yet, an outstanding challenge is understanding how nucleic acid sensing initiates immune responses and its tethering in various infectious, cancerous, autoimmune, and inflammatory diseases. However, the discovery and application of a plethora of small molecule compounds have substantially facilitated this process. This review provides an overview and recent development of the innate DNA-sensing pathway of cGAS-STING and highlights the multiple agonists and inhibitors in fine-tuning the pathway that can be exploited to improve disease treatment, focusing primarily on crucial pathway components and regulators.
Collapse
Affiliation(s)
- Qian Zhang
- MOE Laboratory of Biosystems Homeostasis and Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China
| | - Chen Chen
- MOE Laboratory of Biosystems Homeostasis and Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China
| | - Bing Xia
- Department of Thoracic Cancer, Affiliated Hangzhou Cancer Hospital, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Pinglong Xu
- MOE Laboratory of Biosystems Homeostasis and Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China; Cancer Center, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
48
|
Sun L, Zheng W, Sun Y, Xu T. Long non-coding RNA LTCONS7822 positively regulates innate immunity by targeting MITA in miiuy croaker (Miichthys miiuy). FISH & SHELLFISH IMMUNOLOGY 2022; 125:285-291. [PMID: 35595061 DOI: 10.1016/j.fsi.2022.05.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 06/15/2023]
Abstract
Accumulated studies have shown that long non-coding RNA (lncRNA) is considered a critical regulatory factor in mammals, with a length greater than 200 nucleotides, and it can participate in gene imprinting, dose compensation, transcription enhancement, and antisense regulation. Most of the above studies are carried out in mammals, and there are very few studies on lncRNA of lower vertebrates. Here, we report a novel lncRNA, LTCONS7822, which can play a positive regulatory effect on antiviral immunity in miiuy croaker, Miichthys miiuy. Our results show that the levels of lncRNA LTCONS7822 were significantly increased after poly (I:C) stimulation. Further study, we found that lncRNA LTCONS7822 could positively regulate MITA at the post-transcriptional level. In addition, the dual-luciferase reporter assay analysis showed that the positive regulatory effect of lncRNA LTCONS7822 on NF-κB and IRF3 signaling pathways presented the dose and time-dependent manner. Western blotting experiments proved that lncRNA LTCONS7822 has a positive regulatory effect on MITA. Collectively, our study provided new information to enrich the immune regulation network of lncRNA in teleost fish.
Collapse
Affiliation(s)
- Lingping Sun
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Weiwei Zheng
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Yuena Sun
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education, China; National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, China.
| | - Tianjun Xu
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China; Laboratory of Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education, China; National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, China.
| |
Collapse
|
49
|
Singh RS, Vidhyasagar V, Yang S, Arna AB, Yadav M, Aggarwal A, Aguilera AN, Shinriki S, Bhanumathy KK, Pandey K, Xu A, Rapin N, Bosch M, DeCoteau J, Xiang J, Vizeacoumar FJ, Zhou Y, Misra V, Matsui H, Ross SR, Wu Y. DDX41 is required for cGAS-STING activation against DNA virus infection. Cell Rep 2022; 39:110856. [PMID: 35613581 PMCID: PMC9205463 DOI: 10.1016/j.celrep.2022.110856] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 04/07/2022] [Accepted: 05/02/2022] [Indexed: 12/27/2022] Open
Abstract
Upon binding double-stranded DNA (dsDNA), cyclic GMP-AMP synthase (cGAS) is activated and initiates the cGAS-stimulator of IFN genes (STING)-type I interferon pathway. DEAD-box helicase 41 (DDX41) is a DEAD-box helicase, and mutations in DDX41 cause myelodysplastic syndromes (MDSs) and acute myeloid leukemia (AML). Here, we show that DDX41-knockout (KO) cells have reduced type I interferon production after DNA virus infection. Unexpectedly, activations of cGAS and STING are affected in DDX41 KO cells, suggesting that DDX41 functions upstream of cGAS. The recombinant DDX41 protein exhibits ATP-dependent DNA-unwinding activity and ATP-independent strand-annealing activity. The MDS/AML-derived mutant R525H has reduced unwinding activity but retains normal strand-annealing activity and stimulates greater cGAS dinucleotide-synthesis activity than wild-type DDX41. Overexpression of R525H in either DDX41-deficient or -proficient cells results in higher type I interferon production. Our results have led to the hypothesis that DDX41 utilizes its unwinding and annealing activities to regulate the homeostasis of dsDNA and single-stranded DNA (ssDNA), which, in turn, regulates cGAS-STING activation. cGAS is activated by dsDNA. Singh et al. find DDX41 regulates cGAS activation through unwinding and annealing activities on dsDNA and ssDNA, respectively, and MDS/AML patient mutant R525H causes overactivation of innate immune response due to its unbalanced activities. This DDX41-cGAS-STING pathway may be related to molecular pathogenesis of MDS/AML.
Collapse
Affiliation(s)
- Ravi Shankar Singh
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon SK S7N 5E5, Canada
| | | | - Shizhuo Yang
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon SK S7N 5E5, Canada
| | - Ananna Bhadra Arna
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon SK S7N 5E5, Canada
| | - Manisha Yadav
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon SK S7N 5E5, Canada
| | - Aanchal Aggarwal
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon SK S7N 5E5, Canada
| | - Alexya N Aguilera
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Satoru Shinriki
- Department of Molecular Laboratory Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | | | - Kannupriya Pandey
- Vaccine and Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
| | - Aizhang Xu
- Saskatchewan Cancer Agency, Saskatoon, SK S7N 5E5, Canada
| | - Noreen Rapin
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Mark Bosch
- Saskatchewan Cancer Agency, Saskatoon, SK S7N 5E5, Canada
| | - John DeCoteau
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Jim Xiang
- Saskatchewan Cancer Agency, Saskatoon, SK S7N 5E5, Canada
| | - Franco J Vizeacoumar
- Saskatchewan Cancer Agency, Saskatoon, SK S7N 5E5, Canada; Department of Pathology and Laboratory Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Yan Zhou
- Vaccine and Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada; Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Vikram Misra
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Hirotaka Matsui
- Department of Molecular Laboratory Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Susan R Ross
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Yuliang Wu
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon SK S7N 5E5, Canada.
| |
Collapse
|
50
|
Pang ES, Daraj G, Balka KR, De Nardo D, Macri C, Hochrein H, Masterman KA, Tan PS, Shoppee A, Magill Z, Jahan N, Bafit M, Zhan Y, Kile BT, Lawlor KE, Radford KJ, Wright MD, O’Keeffe M. Discordance in STING-Induced Activation and Cell Death Between Mouse and Human Dendritic Cell Populations. Front Immunol 2022; 13:794776. [PMID: 35281062 PMCID: PMC8914948 DOI: 10.3389/fimmu.2022.794776] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 02/03/2022] [Indexed: 12/27/2022] Open
Abstract
Stimulator of Interferon Genes (STING) is a cytosolic sensor of cyclic dinucleotides (CDNs). The activation of dendritic cells (DC) via the STING pathway, and their subsequent production of type I interferon (IFN) is considered central to eradicating tumours in mouse models. However, this contribution of STING in preclinical murine studies has not translated into positive outcomes of STING agonists in phase I & II clinical trials. We therefore questioned whether a difference in human DC responses could be critical to the lack of STING agonist efficacy in human settings. This study sought to directly compare mouse and human plasmacytoid DCs and conventional DC subset responses upon STING activation. We found all mouse and human DC subsets were potently activated by STING stimulation. As expected, Type I IFNs were produced by both mouse and human plasmacytoid DCs. However, mouse and human plasmacytoid and conventional DCs all produced type III IFNs (i.e., IFN-λs) in response to STING activation. Of particular interest, all human DCs produced large amounts of IFN-λ1, not expressed in the mouse genome. Furthermore, we also found differential cell death responses upon STING activation, observing rapid ablation of mouse, but not human, plasmacytoid DCs. STING-induced cell death in murine plasmacytoid DCs occurred in a cell-intrinsic manner and involved intrinsic apoptosis. These data highlight discordance between STING IFN and cell death responses in mouse and human DCs and caution against extrapolating STING-mediated events in mouse models to equivalent human outcomes.
Collapse
Affiliation(s)
- Ee Shan Pang
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Ghazal Daraj
- Translational Research Institute, Mater Research-University of Queensland, Woolloongabba, QLD, Australia
| | - Katherine R. Balka
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Dominic De Nardo
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Christophe Macri
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | | | - Kelly-Anne Masterman
- Translational Research Institute, Mater Research-University of Queensland, Woolloongabba, QLD, Australia
| | - Peck S. Tan
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Angus Shoppee
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Zoe Magill
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Nazneen Jahan
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Mariam Bafit
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Yifan Zhan
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Benjamin T. Kile
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Kate E. Lawlor
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, VIC, Australia
| | - Kristen J. Radford
- Translational Research Institute, Mater Research-University of Queensland, Woolloongabba, QLD, Australia
| | - Mark D. Wright
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
| | - Meredith O’Keeffe
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- *Correspondence: Meredith O’Keeffe,
| |
Collapse
|