101
|
Emerging role of the cGAS-STING signaling pathway in autoimmune diseases: Biologic function, mechanisms and clinical prospection. Autoimmun Rev 2022; 21:103155. [PMID: 35902046 DOI: 10.1016/j.autrev.2022.103155] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 07/21/2022] [Indexed: 12/15/2022]
Abstract
The cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) signaling pathway, as vital component of innate immune system, acts a vital role in distinguishing invasive pathogens and cytosolic DNA. Cytosolic DNA sensor cGAS first binds to cytosolic DNA and catalyze synthesis of cyclic guanosine monophosphate-adenosine monophosphate (cGAMP), which is known as the secondmessenger. Next, cGAMP activates the adaptor protein STING, triggering a molecular chain reaction to stimulate cytokines including interferons (IFNs). Recently, many researches have revealed that the regulatory role of cGAS-STING signaling pathway in autoimmune diseases (AIDs) such as Rheumatoid arthritis (RA), Aicardi Goutières syndrome (AGS) and systemic lupus erythematosus (SLE). Moreover, accumulated evidence showed inhibition of the cGAS-STING signaling pathway can remarkably suppress joint swelling and inflammatory cell infiltration in RA mice. Therefore, in this review, we describe the molecular properties, biologic function and mechanisms of the cGAS-STING signaling pathway in AIDs. In addition, potential clinical applications especially selective small molecule inhibitors targeting the cGAS-STING signaling pathway are also discussed.
Collapse
|
102
|
Hussain B, Xie Y, Jabeen U, Lu D, Yang B, Wu C, Shang G. Activation of STING Based on Its Structural Features. Front Immunol 2022; 13:808607. [PMID: 35928815 PMCID: PMC9343627 DOI: 10.3389/fimmu.2022.808607] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
The cGAS-cGAMP-STING pathway is an important innate immune signaling cascade responsible for the sensing of abnormal cytosolic double-stranded DNA (dsDNA), which is a hallmark of infection or cancers. Recently, tremendous progress has been made in the understanding of the STING activation mechanism from various aspects. In this review, the molecular mechanism of activation of STING protein based on its structural features is briefly discussed. The underlying molecular mechanism of STING activation will enable us to develop novel therapeutics to treat STING-associated diseases and understand how STING has evolved to eliminate infection and maintain immune homeostasis in innate immunity.
Collapse
Affiliation(s)
- Behzad Hussain
- The Key Laboratory of Medical Molecular Cell Biology of Shanxi Province, The Institutes of Biomedical Sciences, Shanxi University, Taiyuan, China
| | - Yufeng Xie
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Uzma Jabeen
- Institute of Microbiology, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Defen Lu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Bo Yang
- Shanxi Provincial Key Laboratory of Protein Structure Determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
- Shanxi Provincial Key Laboratory for Major Infectious Disease Response, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - Changxin Wu
- The Key Laboratory of Medical Molecular Cell Biology of Shanxi Province, The Institutes of Biomedical Sciences, Shanxi University, Taiyuan, China
| | - Guijun Shang
- Shanxi Provincial Key Laboratory of Protein Structure Determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
- Shanxi Provincial Key Laboratory for Major Infectious Disease Response, Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
- *Correspondence: Guijun Shang,
| |
Collapse
|
103
|
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: 14] [Impact Index Per Article: 4.7] [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
|
104
|
Gao J, Zheng M, Wu X, Zhang H, Su H, Dang Y, Ma M, Wang F, Xu J, Chen L, Liu T, Chen J, Zhang F, Yang L, Xu Q, Hu X, Wang H, Fei Y, Chen C, Liu H. CDK inhibitor Palbociclib targets STING to alleviate autoinflammation. EMBO Rep 2022; 23:e53932. [PMID: 35403787 PMCID: PMC9171422 DOI: 10.15252/embr.202153932] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 03/18/2022] [Accepted: 03/25/2022] [Indexed: 12/29/2022] Open
Abstract
Aberrant activation of stimulator of interferon genes (STING) is tightly associated with multiple types of disease, including cancer, infection, and autoimmune diseases. However, the development of STING modulators for the therapy of STING-related diseases is still an unmet clinical need. We employed a high-throughput screening approach based on the interaction of small-molecule chemical compounds with recombinant STING protein to identify functional STING modulators. Intriguingly, the cyclin-dependent protein kinase (CDK) inhibitor Palbociclib was found to directly bind STING and inhibit its activation in both mouse and human cells. Mechanistically, Palbociclib targets Y167 of STING to block its dimerization, its binding with cyclic dinucleotides, and its trafficking. Importantly, Palbociclib alleviates autoimmune disease features induced by dextran sulphate sodium or genetic ablation of three prime repair exonuclease 1 (Trex1) in mice in a STING-dependent manner. Our work identifies Palbociclib as a novel pharmacological inhibitor of STING that abrogates its homodimerization and provides a basis for the fast repurposing of this Food and Drug Administration-approved drug for the therapy of autoinflammatory diseases.
Collapse
Affiliation(s)
- Jiani Gao
- Department of Thoracic SurgeryShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
- Clinical and Translational Research CenterShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Mengge Zheng
- Clinical and Translational Research CenterShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Xiangyang Wu
- Clinical and Translational Research CenterShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Hang Zhang
- Department of Optical Science and EngineeringShanghai Engineering Research Center of Ultra‐Precision Optical ManufacturingKey Laboratory of Micro and Nano Photonic Structures (Ministry of Education)Fudan UniversityShanghaiChina
| | - Hang Su
- Department of Thoracic SurgeryShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Yifang Dang
- Clinical and Translational Research CenterShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
- Shanghai Key Laboratory of TuberculosisShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Mingtong Ma
- Clinical and Translational Research CenterShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Fei Wang
- Clinical and Translational Research CenterShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Junfang Xu
- Clinical and Translational Research CenterShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Li Chen
- Clinical and Translational Research CenterShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Tianhao Liu
- Clinical and Translational Research CenterShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Jianxia Chen
- Clinical and Translational Research CenterShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
- Shanghai Key Laboratory of TuberculosisShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Fan Zhang
- Clinical and Translational Research CenterShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Li Yang
- Clinical and Translational Research CenterShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Qinghua Xu
- Clinical and Translational Research CenterShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Xuefei Hu
- Department of Thoracic SurgeryShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Heyong Wang
- Central LaboratoryShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Yiyan Fei
- Department of Optical Science and EngineeringShanghai Engineering Research Center of Ultra‐Precision Optical ManufacturingKey Laboratory of Micro and Nano Photonic Structures (Ministry of Education)Fudan UniversityShanghaiChina
| | - Chang Chen
- Department of Thoracic SurgeryShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
| | - Haipeng Liu
- Clinical and Translational Research CenterShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
- Shanghai Key Laboratory of TuberculosisShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
- Central LaboratoryShanghai Pulmonary HospitalTongji University School of MedicineShanghaiChina
- Institute of Nuclear MedicineTongji University School of MedicineShanghaiChina
| |
Collapse
|
105
|
Hong Z, Mei J, Guo H, Zhu J, Wang C. Intervention of cGAS‒STING signaling in sterile inflammatory diseases. J Mol Cell Biol 2022; 14:mjac005. [PMID: 35084490 PMCID: PMC9122663 DOI: 10.1093/jmcb/mjac005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/21/2021] [Accepted: 11/24/2021] [Indexed: 11/12/2022] Open
Abstract
Sterile inflammation characterized by unresolved chronic inflammation is well established to promote the progression of multiple autoimmune diseases, metabolic disorders, neurodegenerative diseases, and cardiovascular diseases, collectively termed 'sterile inflammatory diseases'. By recognizing host-derived DNA, cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS) activates endoplasmic reticulum-associated stimulator of interferon genes (STING), which leads to the induction of type I interferons and inflammatory cytokines or immunogenic cell death that promotes sterile inflammation. Additionally, the DNA/cGAS-independent mode of STING activation has also been characterized in the progression of several sterile inflammatory diseases. This review focuses on the molecular mechanism of cGAS-dependent and cGAS-independent STING signaling under various disease conditions, particularly highlighting the diverse initiators upon this signaling pathway. We also summarize recent advances in the discovery of antagonists targeting cGAS and STING and the evaluation of their efficiencies in preclinical models. Finally, we discuss potential differences in the clinical applications of the specific antagonists, which may shed light on the precision therapeutic interventions.
Collapse
Affiliation(s)
- Ze Hong
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Jiahao Mei
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Hanli Guo
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Juanjuan Zhu
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Chen Wang
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
106
|
Kang J, Wu J, Liu Q, Wu X, Zhao Y, Ren J. Post-Translational Modifications of STING: A Potential Therapeutic Target. Front Immunol 2022; 13:888147. [PMID: 35603197 PMCID: PMC9120648 DOI: 10.3389/fimmu.2022.888147] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/11/2022] [Indexed: 12/18/2022] Open
Abstract
Stimulator of interferon genes (STING) is an endoplasmic-reticulum resident protein, playing essential roles in immune responses against microbial infections. However, over-activation of STING is accompanied by excessive inflammation and results in various diseases, including autoinflammatory diseases and cancers. Therefore, precise regulation of STING activities is critical for adequate immune protection while limiting abnormal tissue damage. Numerous mechanisms regulate STING to maintain homeostasis, including protein-protein interaction and molecular modification. Among these, post-translational modifications (PTMs) are key to accurately orchestrating the activation and degradation of STING by temporarily changing the structure of STING. In this review, we focus on the emerging roles of PTMs that regulate activation and inhibition of STING, and provide insights into the roles of the PTMs of STING in disease pathogenesis and as potential targeted therapy.
Collapse
Affiliation(s)
- Jiaqi Kang
- Research Institute of General Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jie Wu
- Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China
| | - Qinjie Liu
- Research Institute of General Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xiuwen Wu
- Research Institute of General Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
- *Correspondence: Yun Zhao, ; Jianan Ren, ; Xiuwen Wu,
| | - Yun Zhao
- Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Yun Zhao, ; Jianan Ren, ; Xiuwen Wu,
| | - Jianan Ren
- Research Institute of General Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
- *Correspondence: Yun Zhao, ; Jianan Ren, ; Xiuwen Wu,
| |
Collapse
|
107
|
Liu J, Yuan L, Ruan Y, Deng B, Yang Z, Ren Y, Li L, Liu T, Zhao H, Mai R, Chen J. Novel CRBN-Recruiting Proteolysis-Targeting Chimeras as Degraders of Stimulator of Interferon Genes with In Vivo Anti-Inflammatory Efficacy. J Med Chem 2022; 65:6593-6611. [PMID: 35452223 DOI: 10.1021/acs.jmedchem.1c01948] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The activation of the cyclic GMP-AMP synthase-stimulator of interferon gene (STING) pathway has been associated with the pathogenesis of many autoimmune and inflammatory disorders, and small molecules targeting STING have emerged as a new therapeutic strategy for the treatment of these diseases. While several STING inhibitors have been identified with potent anti-inflammatory effects, we would like to explore STING degraders based on the proteolysis-targeting chimera (PROTAC) technology as an alternative strategy to target the STING pathway. Thus, we designed and synthesized a series of STING protein degraders based on a small-molecule STING inhibitor (C-170) and pomalidomide (a CRBN ligand). These compounds demonstrated moderate STING-degrading activities. Among them, SP23 achieved the highest degradation potency with a DC50 of 3.2 μM. Importantly, SP23 exerted high anti-inflammatory efficacy in a cisplatin-induced acute kidney injury mouse model by modulating the STING signaling pathway. Taken together, SP23 represents the first PROTAC degrader of STING deserving further investigation as a new anti-inflammatory agent.
Collapse
Affiliation(s)
- Jin Liu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Lin Yuan
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Yong Ruan
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Bulian Deng
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Zicao Yang
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Yichang Ren
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Ling Li
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Ting Liu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Huiting Zhao
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Ruiyao Mai
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Jianjun Chen
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
108
|
Zhou J, Cui X, Xie Y, Zhang M, Gao J, Zhou X, Ding J, Cen S. Identification of Ziyuglycoside II from natural products library as a novel STING agonist. ChemMedChem 2022; 17:e202100719. [PMID: 35293138 DOI: 10.1002/cmdc.202100719] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 02/11/2022] [Indexed: 11/05/2022]
Abstract
Given the emerging pivotal roles of STING (stimulator of interferon genes) in host pathogen defense and immune-oncology, STING is regarded as a promising target for drug development. CDNs (cyclic dinucleotides) are the first-generation STING agonists. However, their poor metabolic stability and membrane permeability utterly limits therapeutic applications. By contrast, small molecule STING agonists show superiority of properties such as molecular weight, polar character, and delivery diversity. The quest for the potent small molecular agonist of human STING remains ongoing. In our study, through an IRF/IFN pathway-targeted cell-based screen of natural products library, we identified a small-molecular STING agonist Ziyuglycoside II, termed as ST12, with potent stimulation of IRF/IFN pathway and NF-κB pathway. Furthermore, its binding to the C-terminal domain of human STING detected by bio-layer interferometry technique, indicating that ST12 is a human STING agonist. Further tanimoto similarity analyze with existing small-molecule STING agonists indicates that ST12 represents a lead compound with a novel core-structure for the further optimization. Insert abstract text here.
Collapse
Affiliation(s)
- Jinming Zhou
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Immunology, Nanwei Road, 100050, Beijing, CHINA
| | - Xiangling Cui
- Institute of Medicinal biotechnology, Medicinal chemistry, CHINA
| | - Yongli Xie
- Institute of Medicinal biotechnology, Medicinal chemistry, CHINA
| | - Min Zhang
- Zhejiang Normal University, College of Chemistry and Life Science, CHINA
| | - Jieke Gao
- Zhejiang Normal University, College of Chemistry and Life Science, CHINA
| | - Xujun Zhou
- Zhejiang Normal University, College of Chemistry and Life Science, CHINA
| | - Jiwei Ding
- Institute of Medicinal Biotechnology, Medicinal chemistry, CHINA
| | - Shan Cen
- Institute of Medicinal Biotechnology, Immune, CHINA
| |
Collapse
|
109
|
Gan Y, Li X, Han S, Liang Q, Ma X, Rong P, Wang W, Li W. The cGAS/STING Pathway: A Novel Target for Cancer Therapy. Front Immunol 2022; 12:795401. [PMID: 35046953 PMCID: PMC8761794 DOI: 10.3389/fimmu.2021.795401] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/08/2021] [Indexed: 12/12/2022] Open
Abstract
As a DNA receptor, cyclic GMP-AMP synthase (cGAS) plays a crucial role in the immune system by recognizing abnormal DNA in the cytoplasm and activating the stimulator of interferon genes (STING) signaling pathway. This signaling cascade reaction leads to an immune response produced by type I interferon and other immune mediators. Recent advances in research have enhanced our current understanding of the potential role of the cGAS/STING pathway in anticancer therapy; however, in some cases, chronic STING activation may promote tumorigenesis. The present review article discusses the biological mechanisms of the cGAS/STING pathway, its dichotomous role in tumors, and the latest advances with respect to STING agonists and antagonists.
Collapse
Affiliation(s)
- Yu Gan
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Xiaoying Li
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Shuangze Han
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Qi Liang
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China.,Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoqian Ma
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China.,Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Pengfei Rong
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China.,Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Wei Wang
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China.,Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Wei Li
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, China.,Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
110
|
Rech L, Abdellatif M, Pöttler M, Stangl V, Mabotuwana N, Hardy S, Rainer PP. Small molecule STING inhibition improves myocardial infarction remodeling. Life Sci 2022; 291:120263. [PMID: 34971697 DOI: 10.1016/j.lfs.2021.120263] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/07/2021] [Accepted: 12/18/2021] [Indexed: 01/31/2023]
Abstract
AIMS Myocardial infarction (MI) is a major global cause of death. Massive cell death leads to inflammation, which is necessary for ensuing wound healing. Extensive inflammation, however, promotes infarct expansion and adverse remodeling. The DNA sensing receptor cyclic GMP-AMP synthase and its downstream signaling effector stimulator of interferon genes (cGAS-STING) is central in innate immune reactions in infections or autoimmunity. Cytosolic double-strand DNA activates the pathway and down-stream inflammatory responses. Recent papers demonstrated that this pathway is also active following MI and that its genetic targeting improves outcome. Thus, we investigated if pharmacologic pathway inhibition is protective after MI in order to test its translational potential. MAIN METHODS We investigated novel and selective small-molecule STING inhibitors that inhibit STING palmitoylation and multimerization and thereby downstream pathway activation in a preclinical murine MI model. We assessed structural and functional cardiac remodeling, infarct expansion and fibrosis, as well as cardiomyocyte hypertrophy and the expression of inflammatory genes. KEY FINDINGS Pharmacologic STING inhibition did not reduce mortality due to myocardial rupture in non-reperfused MI. Infarct size at day one was comparable. However, three weeks of pharmacologic STING inhibition after reperfused MI decreased infarct expansion and scarring, increased left ventricular systolic function to levels approaching normal values, and reduced myocardial hypertrophy. SIGNIFICANCE Selective small-molecule STING inhibition after myocardial infarction has the potential to improve wound healing responses and pathological remodeling and thereby attenuate the development of ischemic heart failure.
Collapse
Affiliation(s)
- Lavinia Rech
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Mahmoud Abdellatif
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Maria Pöttler
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Verena Stangl
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Nishani Mabotuwana
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria; School of Medicine and Public Health, The University of Newcastle, Callaghan, Australia; Hunter Medical Research Institute, New Lambton Heights, Australia
| | - Sean Hardy
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria; School of Medicine and Public Health, The University of Newcastle, Callaghan, Australia; Hunter Medical Research Institute, New Lambton Heights, Australia
| | - Peter P Rainer
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria; BioTechMed Graz, Graz, Austria.
| |
Collapse
|
111
|
David C, Frémond ML. Lung Inflammation in STING-Associated Vasculopathy with Onset in Infancy (SAVI). Cells 2022; 11:318. [PMID: 35159128 PMCID: PMC8834229 DOI: 10.3390/cells11030318] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 12/15/2022] Open
Abstract
STING-associated vasculopathy with onset in infancy (SAVI) is a type I interferonopathy caused by gain-of-function mutations in STING1 encoding stimulator of interferon genes (STING) protein. SAVI is characterized by severe inflammatory lung disease, a feature not observed in previously described type I interferonopathies i.e., Mendelian autoinflammatory disorders defined by constitutive activation of the type I interferon (IFN) pathway. Molecular defects in nucleic acid metabolism or sensing are central to the pathophysiology of these diseases, with such defects occurring at any step of the tightly regulated pathway of type I IFN production and signaling (e.g., exonuclease loss of function, RNA-DNA hybrid accumulation, constitutive activation of adaptor proteins such as STING). Among over 30 genotypes, SAVI and COPA syndrome, whose pathophysiology was recently linked to a constitutive activation of STING signaling, are the only type I interferonopathies presenting with predominant lung involvement. Lung disease is the leading cause of morbidity and mortality in these two disorders which do not respond to conventional immunosuppressive therapies and only partially to JAK1/2 inhibitors. In human silicosis, STING-dependent sensing of self-DNA following cell death triggered by silica exposure has been found to drive lung inflammation in mice and human models. These recent findings support a key role for STING and nucleic acid sensing in the homeostasis of intrinsic pulmonary inflammation. However, mechanisms by which monogenic defects in the STING pathway lead to pulmonary damages are not yet fully elucidated, and an improved understanding of such mechanisms is fundamental to improved future patient management. Here, we review the recent insights into the pathophysiology of SAVI and outline our current understanding of self-nucleic acid-mediated lung inflammation in humans.
Collapse
Affiliation(s)
- Clémence David
- Université de Paris, Imagine Institute, Laboratory of Neurogenetics and Neuroinflammation, 24 Boulevard du Montparnasse, 75015 Paris, France
| | - Marie-Louise Frémond
- Université de Paris, Imagine Institute, Laboratory of Neurogenetics and Neuroinflammation, 24 Boulevard du Montparnasse, 75015 Paris, France
- Paediatric Immunology-Hematology and Rheumatology Department, Hôpital Necker-Enfants Malades, APHP.Centre-Université de Paris, 24 Boulevard du Montparnasse, 75015 Paris, France
| |
Collapse
|
112
|
The cGAS-STING signaling in cardiovascular and metabolic diseases: Future novel target option for pharmacotherapy. Acta Pharm Sin B 2022; 12:50-75. [PMID: 35127372 PMCID: PMC8799861 DOI: 10.1016/j.apsb.2021.05.011] [Citation(s) in RCA: 140] [Impact Index Per Article: 46.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/05/2021] [Accepted: 04/15/2021] [Indexed: 12/12/2022] Open
Abstract
The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling exert essential regulatory function in microbial-and onco-immunology through the induction of cytokines, primarily type I interferons. Recently, the aberrant and deranged signaling of the cGAS-STING axis is closely implicated in multiple sterile inflammatory diseases, including heart failure, myocardial infarction, cardiac hypertrophy, nonalcoholic fatty liver diseases, aortic aneurysm and dissection, obesity, etc. This is because of the massive loads of damage-associated molecular patterns (mitochondrial DNA, DNA in extracellular vesicles) liberated from recurrent injury to metabolic cellular organelles and tissues, which are sensed by the pathway. Also, the cGAS-STING pathway crosstalk with essential intracellular homeostasis processes like apoptosis, autophagy, and regulate cellular metabolism. Targeting derailed STING signaling has become necessary for chronic inflammatory diseases. Meanwhile, excessive type I interferons signaling impact on cardiovascular and metabolic health remain entirely elusive. In this review, we summarize the intimate connection between the cGAS-STING pathway and cardiovascular and metabolic disorders. We also discuss some potential small molecule inhibitors for the pathway. This review provides insight to stimulate interest in and support future research into understanding this signaling axis in cardiovascular and metabolic tissues and diseases.
Collapse
Key Words
- AA, amino acids
- AAD, aortic aneurysm and dissection
- AKT, protein kinase B
- AMPK, AMP-activated protein kinase
- ATP, adenosine triphosphate
- Ang II, angiotensin II
- CBD, C-binding domain
- CDG, c-di-GMP
- CDNs, cyclic dinucleotides
- CTD, C-terminal domain
- CTT, C-terminal tail
- CVDs, cardiovascular diseases
- Cardiovascular diseases
- Cys, cysteine
- DAMPs, danger-associated molecular patterns
- Damage-associated molecular patterns
- DsbA-L, disulfide-bond A oxidoreductase-like protein
- ER stress
- ER, endoplasmic reticulum
- GTP, guanosine triphosphate
- HAQ, R71H-G230A-R293Q
- HFD, high-fat diet
- ICAM-1, intracellular adhesion molecule 1
- IFN, interferon
- IFN-I, type 1 interferon
- IFNAR, interferon receptors
- IFNIC, interferon-inducible cells
- IKK, IκB kinase
- IL, interleukin
- IRF3, interferon regulatory factor 3
- ISGs, IRF-3-dependent interferon-stimulated genes
- Inflammation
- LBD, ligand-binding pocket
- LPS, lipopolysaccharides
- MI, myocardial infarction
- MLKL, mixed lineage kinase domain-like protein
- MST1, mammalian Ste20-like kinases 1
- Metabolic diseases
- Mitochondria
- NAFLD, nonalcoholic fatty liver disease
- NASH, nonalcoholic steatohepatitis
- NF-κB, nuclear factor-kappa B
- NLRP3, NOD-, LRR- and pyrin domain-containing protein 3
- NO2-FA, nitro-fatty acids
- NTase, nucleotidyltransferase
- PDE3B/4, phosphodiesterase-3B/4
- PKA, protein kinase A
- PPI, protein–protein interface
- Poly: I.C, polyinosinic-polycytidylic acid
- ROS, reactive oxygen species
- SAVI, STING-associated vasculopathy with onset in infancy
- SNPs, single nucleotide polymorphisms
- STIM1, stromal interaction molecule 1
- STING
- STING, stimulator of interferon genes
- Ser, serine
- TAK1, transforming growth factor β-activated kinase 1
- TBK1, TANK-binding kinase 1
- TFAM, mitochondrial transcription factor A
- TLR, Toll-like receptors
- TM, transmembrane
- TNFα, tumor necrosis factor-alpha
- TRAF6, tumor necrosis factor receptor-associated factor 6
- TREX1, three prime repair exonuclease 1
- YAP1, Yes-associated protein 1
- cGAMP, 2′,3′-cyclic GMP–AMP
- cGAS
- cGAS, cyclic GMP–AMP synthase
- dsDNA, double-stranded DNA
- hSTING, human stimulator of interferon genes
- mTOR, mammalian target of rapamycin
- mtDNA, mitochondrial DNA
Collapse
|
113
|
Li Q, Tian S, Liang J, Fan J, Lai J, Chen Q. Therapeutic Development by Targeting the cGAS-STING Pathway in Autoimmune Disease and Cancer. Front Pharmacol 2021; 12:779425. [PMID: 34867409 PMCID: PMC8634458 DOI: 10.3389/fphar.2021.779425] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 10/18/2021] [Indexed: 01/03/2023] Open
Abstract
DNA immune recognition regulation mediated by the cGAS-STING pathway plays an important role in immune functions. Under normal physiological conditions, cGAS can recognize and bind to invading pathogen DNA and activate the innate immune response. On the other hand, abnormal activation of cGAS or STING is closely related to autoimmune diseases. In addition, activation of STING proteins as a bridge connecting innate immunity and adaptive immunity can effectively restrain tumor growth. Therefore, targeting the cGAS-STING pathway can alleviate autoimmune symptoms and be a potential drug target for treating cancer. This article summarizes the current progress on cGAS-STING pathway modulators and lays the foundation for further investigating therapeutic development in autoimmune diseases and tumors.
Collapse
Affiliation(s)
- Qiumei Li
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou, China
| | - Shuoran Tian
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou, China
| | - Jiadi Liang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou, China
| | - Jiqiang Fan
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou, China
| | - Junzhong Lai
- The Cancer Center, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Qi Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou, China
| |
Collapse
|
114
|
Rech L, Rainer PP. The Innate Immune cGAS-STING-Pathway in Cardiovascular Diseases - A Mini Review. Front Cardiovasc Med 2021; 8:715903. [PMID: 34381828 PMCID: PMC8349977 DOI: 10.3389/fcvm.2021.715903] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 07/01/2021] [Indexed: 12/12/2022] Open
Abstract
Inflammation plays a central role in cardiovascular diseases (CVD). One pathway under investigation is the innate immune DNA sensor cyclic GMP-AMP synthase (cGAS) and its downstream receptor stimulator of interferon genes (STING). cGAS-STING upregulates type I interferons in response to pathogens. Recent studies show that also self-DNA may activate cGAS-STING, for instance, DNA released from nuclei or mitochondria during obesity or myocardial infarction. Here, we focus on emerging evidence describing the interaction of cGAS-STING with cardiovascular risk factors and disease. We also touch on translational therapeutic opportunities and potential further investigations.
Collapse
Affiliation(s)
- Lavinia Rech
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Peter P Rainer
- Division of Cardiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria.,BioTechMed Graz, Graz, Austria
| |
Collapse
|
115
|
Hong Z, Ma T, Liu X, Wang C. cGAS-STING pathway: post-translational modifications and functions in sterile inflammatory diseases. FEBS J 2021; 289:6187-6208. [PMID: 34310043 DOI: 10.1111/febs.16137] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/12/2021] [Accepted: 07/22/2021] [Indexed: 02/06/2023]
Abstract
Cytoplasmic microbial and host aberrant DNAs act as danger signals and trigger host immune responses. Upon recognition, the cytosolic DNA sensor cyclic GMP-AMP synthase (cGAS) catalyzes the production of a second messenger 2'3'-cGAMP, which activates endoplasmic reticulum (ER)-associated stimulator of interferon (IFN) genes (STING) and ultimately leads to the induction of type I IFNs and inflammatory genes that collectively initiate host immune defense against microbial invasion. Inappropriate activation or suppression of this signaling pathway has been implicated in the development of some autoimmune diseases, sterile inflammation, and cancers. In this review, we describe how the activity of cGAS and STING is regulated by host post-translational modifications and summarize the recent advances of cell-specific cGAS-STING activation and its association in sterile inflammatory diseases. We also discuss key outstanding questions in the field, including how our knowledge of cGAS-STING pathway could be translated into clinical applications.
Collapse
Affiliation(s)
- Ze Hong
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Tianchi Ma
- Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Xing Liu
- Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Chen Wang
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
116
|
Jin Z, Zhu Z. The role of TRIM proteins in PRR signaling pathways and immune-related diseases. Int Immunopharmacol 2021; 98:107813. [PMID: 34126340 DOI: 10.1016/j.intimp.2021.107813] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 05/13/2021] [Accepted: 05/23/2021] [Indexed: 12/25/2022]
Abstract
Pattern recognition receptors (PRRs) are a kind of recognition molecules mainly expressed on innate immune cells. PRRs recognize one or more kinds of pathogen-associated molecular patterns (PAMPs), inducing the production of interleukin (IL), tumor necrosis factor (TNF), interferon (IFN) and other related cytokines to aggravate immune-related diseases. PPR signaling pathways play an important role in both innate and adaptive immune system, and they are easy to be activated or regulated. Tripartite motif (TRIM) proteins are a group of highly conserved proteins in structure. Most of TRIM proteins contain RING domain, which is thought to play a role in ubiquitination. TRIM proteins are involved in viral immunity, inflammatory response, autophagy, and tumor growth. In this review, we focus on the regulation of TRIM proteins on PRR signaling pathways and their roles in immune-related diseases.
Collapse
Affiliation(s)
- Zheng Jin
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - Zhenhua Zhu
- Department of Orthopaedic Trauma, The Third Affiliated Hospital of Southern, Medical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
117
|
Abstract
cGAS (cytosolic DNA sensor cyclic AMP-GMP synthase)-STING (stimulator of interferon genes) signaling is critical for sensing cytosolic DNA to initiate host immune responses against invading pathogens and cancer. However, inappropriate activation of STING signaling causes severe and often fatal autoimmune or autoinflammatory diseases. Hence, STING is an attractive drug target for the treatment of STING-driven autoimmune and inflammatory disorders. Therefore, there is a need to identify lead compounds that effectively inhibit human STING for further drug development. Here, we identified and characterized a STING-specific inhibitor SN-011 with high efficiency, specificity, and safety, paving the way for therapeutically manipulating STING-mediated clinical diseases. Cytosolic DNA activates cGAS (cytosolic DNA sensor cyclic AMP-GMP synthase)-STING (stimulator of interferon genes) signaling, which triggers interferon and inflammatory responses that help defend against microbial infection and cancer. However, aberrant cytosolic self-DNA in Aicardi–Goutière’s syndrome and constituently active gain-of-function mutations in STING in STING-associated vasculopathy with onset in infancy (SAVI) patients lead to excessive type I interferons and proinflammatory cytokines, which cause difficult-to-treat and sometimes fatal autoimmune disease. Here, in silico docking identified a potent STING antagonist SN-011 that binds with higher affinity to the cyclic dinucleotide (CDN)-binding pocket of STING than endogenous 2′3′-cGAMP. SN-011 locks STING in an open inactive conformation, which inhibits interferon and inflammatory cytokine induction activated by 2′3′-cGAMP, herpes simplex virus type 1 infection, Trex1 deficiency, overexpression of cGAS-STING, or SAVI STING mutants. In Trex1−/− mice, SN-011 was well tolerated, strongly inhibited hallmarks of inflammation and autoimmunity disease, and prevented death. Thus, a specific STING inhibitor that binds to the STING CDN-binding pocket is a promising lead compound for STING-driven disease.
Collapse
|
118
|
Zhang R, Kang R, Tang D. The STING1 network regulates autophagy and cell death. Signal Transduct Target Ther 2021; 6:208. [PMID: 34078874 PMCID: PMC8172903 DOI: 10.1038/s41392-021-00613-4] [Citation(s) in RCA: 138] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 04/01/2021] [Accepted: 04/08/2021] [Indexed: 01/18/2023] Open
Abstract
Cell death and immune response are at the core of life. In past decades, the endoplasmic reticulum (ER) protein STING1 (also known as STING or TMEM173) was found to play a fundamental role in the production of type I interferons (IFNs) and pro-inflammatory cytokines in response to DNA derived from invading microbial pathogens or damaged hosts by activating multiple transcription factors. In addition to this well-known function in infection, inflammation, and immunity, emerging evidence suggests that the STING1-dependent signaling network is implicated in health and disease by regulating autophagic degradation or various cell death modalities (e.g., apoptosis, necroptosis, pyroptosis, ferroptosis, mitotic cell death, and immunogenic cell death [ICD]). Here, we outline the latest advances in our understanding of the regulating mechanisms and signaling pathways of STING1 in autophagy and cell death, which may shed light on new targets for therapeutic interventions.
Collapse
Affiliation(s)
- Ruoxi Zhang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
119
|
Elmanfi S, Yilmaz M, Ong WWS, Yeboah KS, Sintim HO, Gürsoy M, Könönen E, Gürsoy UK. Bacterial Cyclic Dinucleotides and the cGAS-cGAMP-STING Pathway: A Role in Periodontitis? Pathogens 2021; 10:675. [PMID: 34070809 PMCID: PMC8226932 DOI: 10.3390/pathogens10060675] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 01/07/2023] Open
Abstract
Host cells can recognize cytosolic double-stranded DNAs and endogenous second messengers as cyclic dinucleotides-including c-di-GMP, c-di-AMP, and cGAMP-of invading microbes via the critical and essential innate immune signaling adaptor molecule known as STING. This recognition activates the innate immune system and leads to the production of Type I interferons and proinflammatory cytokines. In this review, we (1) focus on the possible role of bacterial cyclic dinucleotides and the STING/TBK1/IRF3 pathway in the pathogenesis of periodontal disease and the regulation of periodontal immune response, and (2) review and discuss activators and inhibitors of the STING pathway as immune response regulators and their potential utility in the treatment of periodontitis. PubMed/Medline, Scopus, and Web of Science were searched with the terms "STING", "TBK 1", "IRF3", and "cGAS"-alone, or together with "periodontitis". Current studies produced evidence for using STING-pathway-targeting molecules as part of anticancer therapy, and as vaccine adjuvants against microbial infections; however, the role of the STING/TBK1/IRF3 pathway in periodontal disease pathogenesis is still undiscovered. Understanding the stimulation of the innate immune response by cyclic dinucleotides opens a new approach to host modulation therapies in periodontology.
Collapse
Affiliation(s)
- Samira Elmanfi
- Department of Periodontology, Institute of Dentistry, University of Turku, 20520 Turku, Finland; (S.E.); (M.G.); (E.K.)
| | - Mustafa Yilmaz
- Department of Periodontology, Faculty of Dentistry, Biruni University, 34010 Istanbul, Turkey;
| | - Wilson W. S. Ong
- Department of Chemistry and Purdue Institute for Drug Discovery and Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, Indiana, IN 47907, USA; (W.W.S.O.); (K.S.Y.)
| | - Kofi S. Yeboah
- Department of Chemistry and Purdue Institute for Drug Discovery and Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, Indiana, IN 47907, USA; (W.W.S.O.); (K.S.Y.)
| | - Herman O. Sintim
- Department of Chemistry and Purdue Institute for Drug Discovery and Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, Indiana, IN 47907, USA; (W.W.S.O.); (K.S.Y.)
| | - Mervi Gürsoy
- Department of Periodontology, Institute of Dentistry, University of Turku, 20520 Turku, Finland; (S.E.); (M.G.); (E.K.)
| | - Eija Könönen
- Department of Periodontology, Institute of Dentistry, University of Turku, 20520 Turku, Finland; (S.E.); (M.G.); (E.K.)
- Oral Health Care, Welfare Division, City of Turku, 20520 Turku, Finland
| | - Ulvi K. Gürsoy
- Department of Periodontology, Institute of Dentistry, University of Turku, 20520 Turku, Finland; (S.E.); (M.G.); (E.K.)
| |
Collapse
|
120
|
Wang X, Liu Y, Han X, Zou G, Zhu W, Shen H, Liu H. Small molecule approaches to treat autoimmune and inflammatory diseases (Part II): Nucleic acid sensing antagonists and inhibitors. Bioorg Med Chem Lett 2021; 44:128101. [PMID: 33984476 DOI: 10.1016/j.bmcl.2021.128101] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 05/05/2021] [Accepted: 05/05/2021] [Indexed: 02/06/2023]
Abstract
Nucleic acid sensing pathways play an important role in the innate immune system, protecting hosts against infections. However, a large body of evidence supports a close association between aberrant activation of those pathways and autoimmune and inflammatory diseases. Part II of the digest series on small molecule approaches to autoimmune and inflammatory diseases concentrates on recent advances with respect to small molecule antagonists or inhibitors of the nucleic acid sensing pathways, including endosomal TLRs, NLRP3 inflammasome and cGAS-STING.
Collapse
Affiliation(s)
- Xiaoqing Wang
- Department of Medicinal Chemistry, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai 201203, China
| | - Yafei Liu
- Department of Medicinal Chemistry, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai 201203, China
| | - Xingchun Han
- Department of Medicinal Chemistry, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai 201203, China
| | - Ge Zou
- Department of Medicinal Chemistry, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai 201203, China
| | - Wei Zhu
- Department of Medicinal Chemistry, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai 201203, China
| | - Hong Shen
- Department of Medicinal Chemistry, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai 201203, China
| | - Haixia Liu
- Department of Medicinal Chemistry, Roche Innovation Center Shanghai, Roche Pharma Research and Early Development, Shanghai 201203, China.
| |
Collapse
|
121
|
Chu L, Li C, Li Y, Yu Q, Yu H, Li C, Meng W, Zhu J, Wang Q, Wang C, Cui S. Perillaldehyde Inhibition of cGAS Reduces dsDNA-Induced Interferon Response. Front Immunol 2021; 12:655637. [PMID: 33968056 PMCID: PMC8100446 DOI: 10.3389/fimmu.2021.655637] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/24/2021] [Indexed: 02/02/2023] Open
Abstract
Cyclic GMP-AMP synthase (cGAS), serving as a primary sensor of intracellular DNA, is essential to initiate anti-microbial innate immunity. Inappropriate activation of cGAS by self-DNA promotes severe autoinflammatory diseases such as Aicardi-Goutières syndrome (AGS); thus, inhibition of cGAS may provide therapeutic benefit in anti-autoimmunity. Here we report that perillaldehyde (PAH), a natural monoterpenoid compound derived from Perilla frutescens, suppresses cytosolic-DNA-induced innate immune responses by inhibiting cGAS activity. Mice treated with PAH are more susceptible to herpes simplex virus type 1 (HSV-1) infection. Moreover, administration with PAH markedly ameliorates self-DNA-induced autoinflammatory responses in a mouse model of AGS. Collectively, our study reveals that PAH can effectively inhibit cGAS-STING signaling and could be developed toward the treatment of cGAS-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Lei Chu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Chenhui Li
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yongxing Li
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Qiuya Yu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Huansha Yu
- Experimental Animal Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chunhui Li
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Wei Meng
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Juanjuan Zhu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Quanyi Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Chen Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Shufang Cui
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
122
|
Prabakaran T, Troldborg A, Kumpunya S, Alee I, Marinković E, Windross SJ, Nandakumar R, Narita R, Zhang BC, Carstensen M, Vejvisithsakul P, Marqvorsen MHS, Iversen MB, Holm CK, Østergaard LJ, Pedersen FS, Pisitkun T, Behrendt R, Pisitkun P, Paludan SR. A STING antagonist modulating the interaction with STIM1 blocks ER-to-Golgi trafficking and inhibits lupus pathology. EBioMedicine 2021; 66:103314. [PMID: 33813142 PMCID: PMC8047499 DOI: 10.1016/j.ebiom.2021.103314] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 03/15/2021] [Accepted: 03/15/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Nucleic acids are potent stimulators of type I interferon (IFN-I) and antiviral defense, but may also promote pathological inflammation. A range of diseases are characterized by elevated IFN-I, including systemic lupus erythematosus (lupus). The DNA-activated cGAS-STING pathway is a major IFN-I-inducing pathway, and activation of signaling is dependent on trafficking of STING from the ER to the Golgi. METHODS Here we used cell culture systems, a mouse lupus model, and material from lupus patients, to explore the mode of action of a STING antagonistic peptide, and its ability to modulate disease processes. FINDINGS We report that the peptide ISD017 selectively inhibits all known down-stream activities of STING, including IFN-I, inflammatory cytokines, autophagy, and apoptosis. ISD017 blocks the essential trafficking of STING from the ER to Golgi through a mechanism dependent on the STING ER retention factor STIM1. Importantly, ISD017 blocks STING activity in vivo and ameliorates disease development in a mouse model for lupus. Finally, ISD017 treatment blocks pathological cytokine responses in cells from lupus patients with elevated IFN-I levels. INTERPRETATION These data hold promise for beneficial use of STING-targeting therapy in lupus. FUNDING The Novo Nordisk Foundation, The European Research Council, The Lundbeck Foundation, European Union under the Horizon 2020 Research, Deutsche Forschungsgemeinschaft, Chulalongkorn University.
Collapse
Affiliation(s)
| | - Anne Troldborg
- Department of Biomedicine, Aarhus University, Aarhus DK-8000, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus DK-8000, Denmark; Department of Rheumatology, Aarhus University Hospital, Aarhus N 8200, Denmark
| | - Sarinya Kumpunya
- Center of Excellence in Systems Biology, Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Inter-Department Program of Biomedical Sciences, Faculty of Graduate, Chulalongkorn University, Bangkok, Thailand
| | - Isara Alee
- Center of Excellence in Systems Biology, Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Inter-Department Program of Biomedical Sciences, Faculty of Graduate, Chulalongkorn University, Bangkok, Thailand
| | - Emilija Marinković
- Institute for Immunology, Faculty of Medicine, Technical University Dresden, Dresden, Germany
| | - Samuel J Windross
- Department of Biomedicine, Aarhus University, Aarhus DK-8000, Denmark
| | - Ramya Nandakumar
- Department of Biomedicine, Aarhus University, Aarhus DK-8000, Denmark
| | - Ryo Narita
- Department of Biomedicine, Aarhus University, Aarhus DK-8000, Denmark
| | - Bao-Cun Zhang
- Department of Biomedicine, Aarhus University, Aarhus DK-8000, Denmark
| | - Mikkel Carstensen
- Department of Biomedicine, Aarhus University, Aarhus DK-8000, Denmark
| | - Pichpisith Vejvisithsakul
- Department of Biomedicine, Aarhus University, Aarhus DK-8000, Denmark; Section for Translational Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | | | - Marie B Iversen
- Department of Biomedicine, Aarhus University, Aarhus DK-8000, Denmark
| | - Christian K Holm
- Department of Biomedicine, Aarhus University, Aarhus DK-8000, Denmark
| | - Lars J Østergaard
- Department of Clinical Medicine, Aarhus University, Aarhus DK-8000, Denmark; Department of Infectious Diseases, Aarhus University Hospital, Aarhus N 8200, Denmark
| | - Finn Skou Pedersen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus DK-8000, Denmark
| | - Trairak Pisitkun
- Center of Excellence in Systems Biology, Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Rayk Behrendt
- Institute for Immunology, Faculty of Medicine, Technical University Dresden, Dresden, Germany
| | - Prapaporn Pisitkun
- Section for Translational Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand; Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Søren R Paludan
- Department of Biomedicine, Aarhus University, Aarhus DK-8000, Denmark.
| |
Collapse
|
123
|
Gong W, Lu L, Zhou Y, Liu J, Ma H, Fu L, Huang S, Zhang Y, Zhang A, Jia Z. The novel STING antagonist H151 ameliorates cisplatin-induced acute kidney injury and mitochondrial dysfunction. Am J Physiol Renal Physiol 2021; 320:F608-F616. [PMID: 33615891 DOI: 10.1152/ajprenal.00554.2020] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Stimulator of interferon genes (STING) is an important adaptor in cytosolic DNA-sensing pathways. A recent study found that the deletion of STING ameliorated cisplatin-induced acute kidney injury (AKI), suggesting that STING could serve as a potential target for AKI therapy. Up to now, a series of small-molecule STING inhibitors/antagonists have been identified. However, none of the research was performed to explore the role of human STING inhibitors in AKI. Here, we investigated the effect of a newly generated covalent antagonist, H151, which targets both human and murine STING, in cisplatin-induced AKI. We found that H151 treatment significantly ameliorated cisplatin-induced kidney injury as shown by the improvement of renal function, kidney morphology, and renal inflammation. In addition, tubular cell apoptosis and increased renal tubular injury marker neutrophil gelatinase-associated lipocalin induced by cisplatin were also effectively attenuated in H151-treated mice. Moreover, the mitochondrial injury caused by cisplatin was also reversed as evidenced by improved mitochondrial morphology, restored mitochondrial DNA content, and reversed mitochondrial gene expression. Finally, we observed enhanced mitochondrial DNA levels in the plasma of patients receiving platinum-based chemotherapy compared with healthy controls, which could potentially activate STING signaling. Taken together, these findings suggested that H151 could be a potential therapeutic agent for treating AKI possibly through inhibiting STING-mediated inflammation and mitochondrial injury.NEW & NOTEWORTHY Although various stimulator of interferon genes (STING) inhibitors have been identified, no research was performed to investigate the role of human STING inhibitors in AKI. Here, we evaluated the effect of H151 targeting both human and murine STING on cisplatin-induced AKI and observed a protection against renal injury possibly through ameliorating inflammation and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Wei Gong
- Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Lingling Lu
- Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yu Zhou
- Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Jiaye Liu
- Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Haoyang Ma
- Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Lvhan Fu
- Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Songming Huang
- Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yue Zhang
- Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Aihua Zhang
- Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Zhanjun Jia
- Nanjing Key Lab of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
124
|
Paul BD, Snyder SH, Bohr VA. Signaling by cGAS-STING in Neurodegeneration, Neuroinflammation, and Aging. Trends Neurosci 2021; 44:83-96. [PMID: 33187730 PMCID: PMC8662531 DOI: 10.1016/j.tins.2020.10.008] [Citation(s) in RCA: 159] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/22/2020] [Accepted: 10/15/2020] [Indexed: 12/11/2022]
Abstract
Recognition of foreign or misplaced nucleic acids is one of the principal modes by which the immune system detects pathogenic entities. When cytosolic DNA is sensed, a signal is relayed via the cGAS-STING pathway: this involves the activation of cyclic GMP-AMP (cGMP-AMP) synthase (cGAS) and generation of the cyclic dinucleotide cGAMP, followed by the induction of stimulator of interferon genes (STING). The cGAS-STING pathway responds to viral, bacterial, and self-DNA. Whereas it generally mediates immune surveillance and is often neuroprotective, excessive engagement of the system can be deleterious. This is relevant in aging and age-related neurological diseases, where neuroinflammation contributes to disease progression. This review focuses on cGAS-STING signaling in aging, neurodegeneration, and neuroinflammation, and on therapeutic implications.
Collapse
Affiliation(s)
- Bindu D Paul
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Solomon H Snyder
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Vilhelm A Bohr
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|
125
|
Abstract
The cGAS-STING signalling pathway has emerged as a key mediator of inflammation in the settings of infection, cellular stress and tissue damage. Underlying this broad involvement of the cGAS-STING pathway is its capacity to sense and regulate the cellular response towards microbial and host-derived DNAs, which serve as ubiquitous danger-associated molecules. Insights into the structural and molecular biology of the cGAS-STING pathway have enabled the development of selective small-molecule inhibitors with the potential to target the cGAS-STING axis in a number of inflammatory diseases in humans. Here, we outline the principal elements of the cGAS-STING signalling cascade and discuss the general mechanisms underlying the association of cGAS-STING activity with various autoinflammatory, autoimmune and degenerative diseases. Finally, we outline the chemical nature of recently developed cGAS and STING antagonists and summarize their potential clinical applications.
Collapse
|
126
|
Yang D, Wang S, Huang X, Ma W, Xue Z, Zhao L, Ouyang H, He J. Pharmacokinetic comparison of 15 active compositions in rat plasma after oral administration of raw and honey‐processed
Aster tataricus
extracts. J Sep Sci 2020; 44:908-921. [DOI: 10.1002/jssc.202001020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/19/2020] [Accepted: 11/27/2020] [Indexed: 01/18/2023]
Affiliation(s)
- Dongyue Yang
- State Key Laboratory of Component‐based Chinese Medicine Tianjin University of Traditional Chinese Medicine Tianjin P.R. China
| | - Songrui Wang
- State Key Laboratory of Component‐based Chinese Medicine Tianjin University of Traditional Chinese Medicine Tianjin P.R. China
| | - Xuhua Huang
- State Key Laboratory of Component‐based Chinese Medicine Tianjin University of Traditional Chinese Medicine Tianjin P.R. China
| | - Wenjuan Ma
- State Key Laboratory of Component‐based Chinese Medicine Tianjin University of Traditional Chinese Medicine Tianjin P.R. China
| | - Zixiang Xue
- State Key Laboratory of Component‐based Chinese Medicine Tianjin University of Traditional Chinese Medicine Tianjin P.R. China
| | - Lulu Zhao
- State Key Laboratory of Component‐based Chinese Medicine Tianjin University of Traditional Chinese Medicine Tianjin P.R. China
| | - Huizi Ouyang
- State Key Laboratory of Component‐based Chinese Medicine Tianjin University of Traditional Chinese Medicine Tianjin P.R. China
| | - Jun He
- State Key Laboratory of Component‐based Chinese Medicine Tianjin University of Traditional Chinese Medicine Tianjin P.R. China
| |
Collapse
|
127
|
Liu Y, Lu X, Qin N, Qiao Y, Xing S, Liu W, Feng F, Liu Z, Sun H. STING, a promising target for small molecular immune modulator: A review. Eur J Med Chem 2020; 211:113113. [PMID: 33360799 DOI: 10.1016/j.ejmech.2020.113113] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 12/13/2020] [Accepted: 12/14/2020] [Indexed: 12/19/2022]
Abstract
Stimulator of interferon genes (STING) plays a crucial role in human innate immune system, which is gradually concerned following the emerging immunotherapy. Activated STING induces the production of type I interferons (IFNs) and proinflammatory cytokines through STING-TBK1-IRF3/NF-κB pathway, which could be applied into the treatment of infection, inflammation, and tumorigenesis. Here, we provided a detailed summary of STING from its structure, function and regulation. Especially, we illustrated the canonical or noncanonical cyclic dinucleotides (CDNs) and synthetic small molecules for STING activation or inhibition and their efficacy in related diseases. Importantly, we particularly emphasized the discovery, development and modification of STING agonist or antagonist, attempting to enlighten reader's mind for enriching small molecular modulator of STING. In addition, we summarized biological evaluation methods for the assessment of small molecules activity.
Collapse
Affiliation(s)
- Yijun Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Xin Lu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Nan Qin
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Yuting Qiao
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Shuaishuai Xing
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Wenyuan Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Feng Feng
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, People's Republic of China; Jiangsu Food and Pharmaceuticals Science College, Institute of Food and Pharmaceuticals Research, 223005, People's Republic of China
| | - Zongliang Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, PR China.
| | - Haopeng Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China.
| |
Collapse
|
128
|
Sun B, Zeng H, Liang J, Zhang L, Hu H, Wang Q, Meng W, Li C, Ye F, Wang C, Zhu J. NSUN5 Facilitates Viral RNA Recognition by RIG-I Receptor. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 205:3408-3418. [PMID: 33177158 DOI: 10.4049/jimmunol.1901455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 10/14/2020] [Indexed: 11/19/2022]
Abstract
The RIG-I receptor induces the innate antiviral responses upon sensing RNA viruses. The mechanisms through which RIG-I optimizes the strength of the downstream signaling remain incompletely understood. In this study, we identified that NSUN5 could potentiate the RIG-I innate signaling pathway. Deficiency of NSUN5 enhanced RNA virus proliferation and inhibited the induction of the downstream antiviral genes. Consistently, NSUN5-deficient mice were more susceptible to RNA virus infection than their wild-type littermates. Mechanistically, NSUN5 bound directly to both viral RNA and RIG-I, synergizing the recognition of dsRNA by RIG-I. Collectively, to our knowledge, this study characterized NSUN5 as a novel RIG-I coreceptor, playing a vital role in restricting RNA virus infection.
Collapse
Affiliation(s)
- Boyue Sun
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Haoyang Zeng
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Jiaqian Liang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Lele Zhang
- Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; and
| | - Haiyang Hu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Quanyi Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Wei Meng
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Chenhui Li
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Fuqiang Ye
- Department of Disease Control and Prevention, Center for Disease Control and Prevention of Eastern Theater Command, Nanjing 210002, China
| | - Chen Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China;
| | - Juanjuan Zhu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China;
| |
Collapse
|
129
|
Balka KR, De Nardo D. Molecular and spatial mechanisms governing STING signalling. FEBS J 2020; 288:5504-5529. [PMID: 33237620 DOI: 10.1111/febs.15640] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/12/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022]
Abstract
Detection of microbial nucleic acids via innate immune receptors is critical for establishing host defence against pathogens. The DNA-sensing cGAS-STING pathway has gained increasing attention in the last decade as a key pathway for combating viral and bacterial infections. cGAS-STING activation primarily promotes the secretion of antiviral type I IFNs via the key transcription factor, IRF3. In addition, cGAS-STING signalling also elicits proinflammatory cytokines through NF-κB activity. Activation of IRF3 and NF-κB is mediated by the chief signalling receptor protein STING. Interestingly, STING undergoes significant trafficking events across multiple subcellular locations, which regulates both the activation of downstream signalling pathways, as well as appropriate termination of the responses. Studies to date have provided a comprehensive view of the regulation and role of the IRF3-IFN pathway downstream of STING. However, many aspects of STING signalling remain relatively poorly defined. This review will explore the current understanding of the mechanisms through which STING elicits inflammatory and antimicrobial responses, focusing on the precise signalling and intracellular trafficking events that occur. We will also discuss exciting and emerging concepts in the field, including the importance of IFN-independent STING responses for host defence and during STING-related disease.
Collapse
Affiliation(s)
- Katherine R Balka
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Vic., Australia
| | - Dominic De Nardo
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Vic., Australia
| |
Collapse
|
130
|
Cui S, Yu Q, Chu L, Cui Y, Ding M, Wang Q, Wang H, Chen Y, Liu X, Wang C. Nuclear cGAS Functions Non-canonically to Enhance Antiviral Immunity via Recruiting Methyltransferase Prmt5. Cell Rep 2020; 33:108490. [PMID: 33296647 DOI: 10.1016/j.celrep.2020.108490] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 10/11/2020] [Accepted: 11/13/2020] [Indexed: 12/19/2022] Open
Abstract
Cyclic guanosine monophosphate (GMP)-AMP synthase (cGAS), upon sensing cytosolic DNA, catalyzes the production of cyclic guanosine monophosphate-adenosine monophosphate (cGAMP), which activates STING-TBK1-IRF3 signaling. cGAS is also present in the nucleus, but the relevant nuclear function or mechanism remains largely unknown. Here, we report that nuclear cGAS is indispensable for inducing cytokines and chemokines triggered by RNA/DNA viruses. Unexpectedly, the DNA-binding/nucleotidyltransferase activity of cGAS is dispensable for RNA-virus-induced genes expression. cGAS deficiency does not affect the phosphorylation, dimerization, or nuclear translocation of IRF3 induced by double-stranded RNA (dsRNA). Mechanistically, nuclear-localized cGAS interacts with protein arginine methyltransferase 5 (Prmt5), which catalyzes the symmetric dimethylation of histone H3 arginine 2 at Ifnb and Ifna4 promoters, thus facilitating the access of IRF3. Deficiency of Prmt5 or disrupting its catalytic activity suppresses the production of type I interferons (IFNs), impairing the host defenses against RNA/DNA virus infections. Taken together, our study uncovers a non-canonical function of nuclear-localized cGAS in innate immunity via regulating histone arginine modification.
Collapse
Affiliation(s)
- Shufang Cui
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing 211198, China
| | - Qiuya Yu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing 211198, China
| | - Lei Chu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing 211198, China
| | - Ye Cui
- Division of Immunology, Department of Pediatrics, The Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ming Ding
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing 211198, China
| | - Quanyi Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing 211198, China
| | - Hongyun Wang
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yu Chen
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Xing Liu
- The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Chen Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing 211198, China.
| |
Collapse
|
131
|
Small molecules targeting the innate immune cGAS‒STING‒TBK1 signaling pathway. Acta Pharm Sin B 2020; 10:2272-2298. [PMID: 33354501 PMCID: PMC7745059 DOI: 10.1016/j.apsb.2020.03.001] [Citation(s) in RCA: 191] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/20/2020] [Accepted: 02/28/2020] [Indexed: 12/19/2022] Open
Abstract
Multiple cancer immunotherapies including chimeric antigen receptor T cell and immune checkpoint inhibitors (ICIs) have been successfully developed to treat various cancers by motivating the adaptive anti-tumor immunity. Particularly, the checkpoint blockade approach has achieved great clinic success as evidenced by several U.S. Food and Drug Administration (FDA)-approved anti-programmed death receptor 1/ligand 1 or anti-cytotoxic T lymphocyte associated protein 4 antibodies. However, the majority of cancers have low clinical response rates to these ICIs due to poor tumor immunogenicity. Indeed, the cyclic guanosine monophosphate-adenosine monophosphate synthase‒stimulator of interferon genes‒TANK-binding kinase 1 (cGAS‒STING‒TBK1) axis is now appreciated as the major signaling pathway in innate immune response across different species. Aberrant signaling of this pathway has been closely linked to multiple diseases, including auto-inflammation, virus infection and cancers. In this perspective, we provide an updated review on the latest progress on the development of small molecule modulators targeting the cGAS‒STING‒TBK1 signaling pathway and their preclinical and clinical use as a new immune stimulatory therapy. Meanwhile, highlights on the clinical candidates, limitations and challenges, as well as future directions in this field are also discussed. Further, small molecule inhibitors targeting this signaling axis and their potential therapeutic use for various indications are discussed as well.
Collapse
Key Words
- ABZI, amidobenzimidazole
- ACMA, 9-amino-6-chloro-2-methoxyacridine
- AMP, adenosine monophosphate
- ATP, adenosine triphosphate
- Anti-tumor
- BNBC, 6-bromo-N-(naphthalen-1-yl)benzo[d][1,3]dioxole-5-carboxamide
- CBD, cyclic dinucleotide-binding domain
- CDA, cyclic diadenosine monophosphate (c-di-AMP)
- CDG, cyclic diguanosine monophosphate (c-di-GMP)
- CDN, cyclic dinucleotide
- CMA, 10-carboxymethyl-9-acridanone
- CTD, C-terminal domain
- CTLA-4, cytotoxic T lymphocyte associated protein 4
- CTT, C-terminal tail
- CXCL, chemokine (C-X-C motif) ligand
- DC50, concentration for 50% degradation
- DCs, dendritic cells
- DMXAA, 5,6-dimethylxanthenone-4-acetic acid
- DSDP, dispiro diketopiperzine
- EM, cryo-electron microscopy
- ENPP1, ecto-nucleotide pyrophosphatase/phosphodiesterase
- ER, endoplasmic reticulum
- FAA, flavone-8-acetic acid
- FDA, U.S. Food and Drug Administration
- FP, fluorescence polarization
- GMP, guanosine monophosphate
- GTP, guanosine triphosphate
- HCQ, hydrochloroquine
- HTS, high throughput screening
- ICI, immune checkpoint inhibitor
- IKK, IκB kinase
- IO, immune-oncology
- IRF3, interferon regulatory factor 3
- ISG, interferon stimulated gene
- ITC, isothermal titration calorimetry
- Immunotherapy
- KD, kinase domain
- LBD, ligand-binding domain
- MDCK, Madin–Darby canine kidney
- MG, Mangostin
- MI, maximum induction
- MLK, mixed lineage kinase
- MinEC5×, minimum effective concentration for inducing 5-fold luciferase activity
- NF-κB, nuclear factor-κB
- Ntase, nucleotidyl transferase
- PBMCs, peripheral-blood mononuclear cells
- PD-1, programmed death receptor 1
- PD-L1, programmed death ligand 1
- PDE, phosphodiesterases
- PDK1, 3-phosphoinositide-dependent protein kinase 1
- PPi, pyrophosphoric acid
- PROTACs, proteolysis targeting chimeras
- PRRs, pattern recognition receptors
- QC, quinacrine
- SAR, structure–activity relationship
- SDD, scaffold and dimerization domain
- STAT, signal transducer and activator of transcription
- STING
- STING, stimulator of interferon genes
- Small molecule modulators
- TBK1
- TBK1, TANK-binding kinase 1
- THIQCs, tetrahydroisoquinolone acetic acids
- TNFRSF, tumor necrosis factor receptor superfamily
- ULD, ubiquitin-like domain
- VHL, von Hippel–Lindau
- cAIMP, cyclic adenosine-inosine monophosphate
- cGAMP, cyclic guanosine monophosphate-adenosine monophosphate
- cGAS
- cGAS, cyclic guanosine monophosphate-adenosine monophosphate synthase
- dsDNA, double-stranded DNA
- i.t., intratumoral
Collapse
|
132
|
Liu K, Lan Y, Li X, Li M, Cui L, Luo H, Luo L. Development of small molecule inhibitors/agonists targeting STING for disease. Biomed Pharmacother 2020; 132:110945. [PMID: 33254439 DOI: 10.1016/j.biopha.2020.110945] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/22/2020] [Accepted: 10/24/2020] [Indexed: 01/07/2023] Open
Abstract
Cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS) -stimulator of interferon genes (STING) signaling pathway is the primary immune response pathway in the cytoplasm. Pharmacological regulation of the STING pathway has good characteristics in both structure and function, which plays a significant role in the immunotherapy of autoimmune diseases, autoinflammatory diseases, and cancer. In this review, we summarized the activation of STING signaling pathway, the STING-related diseases, the development principle and the latest progress of inhibitors and agonists targeting STING. Our review demonstrates that STING signal pathway is a promising drug target, providing effective clues and correct guidance for the discovery of novel small molecule inhibitors/agonists that targeted STING for cancer, autoimmune, and inflammatory diseases.
Collapse
Affiliation(s)
- Kaifeng Liu
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, China
| | - Yongqi Lan
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, China
| | - Xiaoling Li
- Animal Experiment Center of Guangdong Medical University, Zhanjiang, 524023, China
| | - Mingyue Li
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Liao Cui
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Hui Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, 524023, China
| | - Lianxiang Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, 524023, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong, 524023, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, Guangdong, 524023, China.
| |
Collapse
|
133
|
Wang S, Wang L, Qin X, Turdi S, Sun D, Culver B, Reiter RJ, Wang X, Zhou H, Ren J. ALDH2 contributes to melatonin-induced protection against APP/PS1 mutation-prompted cardiac anomalies through cGAS-STING-TBK1-mediated regulation of mitophagy. Signal Transduct Target Ther 2020; 5:119. [PMID: 32703954 PMCID: PMC7378833 DOI: 10.1038/s41392-020-0171-5] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 03/25/2020] [Accepted: 04/15/2020] [Indexed: 12/26/2022] Open
Abstract
Ample clinical evidence suggests a high incidence of cardiovascular events in Alzheimer's disease (AD), although neither precise etiology nor effective treatment is available. This study was designed to evaluate cardiac function in AD patients and APP/PS1 mutant mice, along with circulating levels of melatonin, mitochondrial aldehyde dehydrogenase (ALDH2) and autophagy. AD patients and APP/PS1 mice displayed cognitive and myocardial deficits, low levels of circulating melatonin, ALDH2 activity, and autophagy, ultrastructural, geometric (cardiac atrophy and interstitial fibrosis) and functional (reduced fractional shortening and cardiomyocyte contraction) anomalies, mitochondrial injury, cytosolic mtDNA buildup, apoptosis, and suppressed autophagy and mitophagy. APP/PS1 mutation downregulated cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING) levels and TBK1 phosphorylation, while promoting Aβ accumulation. Treatment with melatonin overtly ameliorated unfavorable APP/PS1-induced changes in cardiac geometry and function, apoptosis, mitochondrial integrity, cytosolic mtDNA accumulation (using both immunocytochemistry and qPCR), mitophagy, and cGAS-STING-TBK1 signaling, although these benefits were absent in APP/PS1/ALDH2-/- mice. In vitro evidence indicated that melatonin attenuated APP/PS1-induced suppression of mitophagy and cardiomyocyte function, and the effect was negated by the nonselective melatonin receptor blocker luzindole, inhibitors or RNA interference of cGAS, STING, TBK1, and autophagy. Our data collectively established a correlation among cardiac dysfunction, low levels of melatonin, ALDH2 activity, and autophagy in AD patients, with compelling support in APP/PS1 mice, in which melatonin rescued myopathic changes by promoting cGAS-STING-TBK1 signaling and mitophagy via an ALDH2-dependent mechanism.
Collapse
Affiliation(s)
- Shuyi Wang
- Department of Emergency, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, 82071, USA
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, 200032, China
| | - Lin Wang
- Department of Geriatrics, Xijing Hospital, The Air Force Military Medical University, Xi'an, China
| | - Xing Qin
- Department of Cardiology, Xijing Hospital, The Air Force Military Medical University, Xi'an, 710032, China
| | - Subat Turdi
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, 82071, USA
| | - Dongdong Sun
- Department of Cardiology, Xijing Hospital, The Air Force Military Medical University, Xi'an, 710032, China
| | - Bruce Culver
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, 82071, USA
| | - Russel J Reiter
- Department of Cellular and Structural Biology, UT Health San Antonio, San Antonio, TX, USA
| | - Xiaoming Wang
- Department of Geriatrics, Xijing Hospital, The Air Force Military Medical University, Xi'an, China.
| | - Hao Zhou
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, 82071, USA.
- Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, 100853, China.
| | - Jun Ren
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, 82071, USA.
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, 200032, China.
- Department of Geriatrics, Xijing Hospital, The Air Force Military Medical University, Xi'an, China.
| |
Collapse
|
134
|
Abstract
For centuries, people believed that bats possessed sinister powers. Bats are thought to be ancestral hosts to many deadly viruses affecting humans including Ebola, rabies, and most recently SARS-CoV-2 coronavirus. However, bats themselves tolerate these viruses without ill effects. The second power that bats have is their longevity. Bats live much longer than similar-sized land mammals. Here we review how bats' ability to control inflammation may be contributing to their longevity. The underlying mechanisms may hold clues to developing new treatments for age-related diseases. Now may be the time to use science to exploit the secret powers of bats for human benefit.
Collapse
Affiliation(s)
- Vera Gorbunova
- Departments of Biology and Medicine, University of Rochester, Rochester, NY 14627, USA.
| | - Andrei Seluanov
- Departments of Biology and Medicine, University of Rochester, Rochester, NY 14627, USA
| | - Brian K Kennedy
- Departments of Biochemistry and Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore; Centre for Healthy Longevity, National University Health System, Singapore 117609, Singapore; Singapore Institute of Clinical Sciences, A(∗)STAR, Singapore 117609, Singapore.
| |
Collapse
|
135
|
Talotta R, Atzeni F, Laska MJ. Therapeutic peptides for the treatment of systemic lupus erythematosus: a place in therapy. Expert Opin Investig Drugs 2020; 29:845-867. [PMID: 32500750 DOI: 10.1080/13543784.2020.1777983] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Studies in vitro and in vivo have identified several peptides that are potentially useful in treating systemic lupus erythematosus (SLE). The rationale for their use lies in the cost-effective production, high potency, target selectivity, low toxicity, and a peculiar mechanism of action that is mainly based on the induction of immune tolerance. Three therapeutic peptides have entered clinical development, but they have yielded disappointing results. However, some subsets of patients, such as those with the positivity of anti-dsDNA antibodies, appear more likely to respond to these medications. AREAS COVERED This review evaluates the potential use of therapeutic peptides for SLE and gives an opinion on how they may offer advantages for SLE treatment. EXPERT OPINION Given their acceptable safety profile, therapeutic peptides could be added to agents traditionally used to treat SLE and this may offer a synergistic and drug-sparing effect, especially in selected patient populations. Moreover, they could temporarily be utilized to manage SLE flares, or be administered as a vaccine in subjects at risk. Efforts to ameliorate bioavailability, increase the half-life and prevent immunogenicity are ongoing. The formulation of hybrid compounds, like peptibodies or peptidomimetic small molecules, is expected to yield renewed treatments with a better pharmacologic profile and increased efficacy.
Collapse
Affiliation(s)
- Rossella Talotta
- Department of Clinical and Experimental Medicine, Rheumatology Unit, Azienda Ospedaliera "Gaetano Martino", University of Messina , Messina, Italy
| | - Fabiola Atzeni
- Department of Clinical and Experimental Medicine, Rheumatology Unit, Azienda Ospedaliera "Gaetano Martino", University of Messina , Messina, Italy
| | | |
Collapse
|
136
|
Weng JK. Plant Solutions for the COVID-19 Pandemic and Beyond: Historical Reflections and Future Perspectives. MOLECULAR PLANT 2020; 13:803-807. [PMID: 32442649 PMCID: PMC7237358 DOI: 10.1016/j.molp.2020.05.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 05/18/2020] [Accepted: 05/18/2020] [Indexed: 05/20/2023]
Affiliation(s)
- Jing-Ke Weng
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
137
|
Chen L, Quan H, Xu Z, Wang H, Xia Y, Lou L, Yang W. A modular biomimetic strategy for the synthesis of macrolide P-glycoprotein inhibitors via Rh-catalyzed C-H activation. Nat Commun 2020; 11:2151. [PMID: 32358512 PMCID: PMC7195407 DOI: 10.1038/s41467-020-16084-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 04/09/2020] [Indexed: 11/09/2022] Open
Abstract
One of the key challenges to overcome multidrug resistance (MDR) in cancer is the development of more effective and general strategies to discover bioactive scaffolds. Inspired by natural products, we describe a strategy to achieve this goal by modular biomimetic synthesis of scaffolds of (Z)-allylic-supported macrolides. Herein, an Rh(III)-catalyzed native carboxylic acid-directed and solvent-free C−H activation allylation with high stereoselectivity and chemoselectivity is achieved. The generated poly-substituted allylic alcohol as a multifunctional and biomimetic building block is crucial for the synthesis of (Z)-allylic-supported macrolides. Moreover, the unique allylic-supported macrolides significantly potentiate the sensitivity of tumor cells to cytotoxic agents such as vinorelbine and doxetaxel by reversing p170-glycoprotein-mediated MDR. Our findings will inspire the evolution of synthetic chemistry and open avenues for expedient and diversified synthesis of bioactive macrocyclic molecules. One strategy to address multidrug resistance in cancer is the development of modular methods to access bioactive scaffolds. Here, the authors report a Rh(III)-catalyzed carboxylic acid-directed C(sp2)−H allylation and apply it to the modular synthesis of (Z)-allylic macrolides which enhance antitumoral drug activity.
Collapse
Affiliation(s)
- Lu Chen
- Chinese Academy of Sciences Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Haitian Quan
- Chinese Academy of Sciences Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhongliang Xu
- Chinese Academy of Sciences Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hao Wang
- Chinese Academy of Sciences Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuanzhi Xia
- College of Chemistry and Materials Engineering, Wenzhou University, Wenzhou, 325035, China
| | - Liguang Lou
- Chinese Academy of Sciences Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Weibo Yang
- Chinese Academy of Sciences Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China. .,Key Laboratory for Functional Material, Educational Department of Liaoning Province, University of Science and Technology Liaoning, Anshan, 114051, China.
| |
Collapse
|
138
|
Wan D, Jiang W, Hao J. Research Advances in How the cGAS-STING Pathway Controls the Cellular Inflammatory Response. Front Immunol 2020; 11:615. [PMID: 32411126 PMCID: PMC7198750 DOI: 10.3389/fimmu.2020.00615] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 03/17/2020] [Indexed: 12/19/2022] Open
Abstract
Double-stranded DNA (dsDNA) sensor cyclic-GMP-AMP synthase (cGAS) along with the downstream stimulator of interferon genes (STING) acting as essential immune-surveillance mediators have become hot topics of research. The intrinsic function of the cGAS-STING pathway facilitates type-I interferon (IFN) inflammatory signaling responses and other cellular processes such as autophagy, cell survival, senescence. cGAS-STING pathway interplays with other innate immune pathways, by which it participates in regulating infection, inflammatory disease, and cancer. The therapeutic approaches targeting this pathway show promise for future translation into clinical applications. Here, we present a review of the important previous works and recent advances regarding the cGAS-STING pathway, and provide a comprehensive understanding of the modulatory pattern of the cGAS-STING pathway under multifarious pathologic states.
Collapse
Affiliation(s)
- Dongshan Wan
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.,Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Wei Jiang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Junwei Hao
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
139
|
Shi M, Zhang P, Vora SM, Wu H. Higher-order assemblies in innate immune and inflammatory signaling: A general principle in cell biology. Curr Opin Cell Biol 2020; 63:194-203. [PMID: 32272435 DOI: 10.1016/j.ceb.2020.03.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 03/08/2020] [Accepted: 03/09/2020] [Indexed: 12/12/2022]
Abstract
Higher-order supramolecular complexes-dubbed signalosomes carry out key signaling and effector functions in innate immunity and inflammation. In this review, we present several recently discovered signalosomes that are formed either by stable protein-protein interactions or by dynamic liquid-liquid phase separation. Structural features of these signalosomes are highlighted to elucidate their functions and biological insights.
Collapse
Affiliation(s)
- Ming Shi
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China.
| | - Pengfei Zhang
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Setu M Vora
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
140
|
Zhang H, You QD, Xu XL. Targeting Stimulator of Interferon Genes (STING): A Medicinal Chemistry Perspective. J Med Chem 2019; 63:3785-3816. [DOI: 10.1021/acs.jmedchem.9b01039] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Han Zhang
- State Key Laboratory of Natural Medicines and Jiang Su 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 Jiang Su 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 Jiang Su 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
|
141
|
Antitumor astins originate from the fungal endophyte Cyanodermella asteris living within the medicinal plant Aster tataricus. Proc Natl Acad Sci U S A 2019; 116:26909-26917. [PMID: 31811021 DOI: 10.1073/pnas.1910527116] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Medicinal plants are a prolific source of natural products with remarkable chemical and biological properties, many of which have considerable remedial benefits. Numerous medicinal plants are suffering from wildcrafting, and thus biotechnological production processes of their natural products are urgently needed. The plant Aster tataricus is widely used in traditional Chinese medicine and contains unique active ingredients named astins. These are macrocyclic peptides showing promising antitumor activities and usually containing the highly unusual moiety 3,4-dichloroproline. The biosynthetic origins of astins are unknown despite being studied for decades. Here we show that astins are produced by the recently discovered fungal endophyte Cyanodermella asteris We were able to produce astins in reasonable and reproducible amounts using axenic cultures of the endophyte. We identified the biosynthetic gene cluster responsible for astin biosynthesis in the genome of C. asteris and propose a production pathway that is based on a nonribosomal peptide synthetase. Striking differences in the production profiles of endophyte and host plant imply a symbiotic cross-species biosynthesis pathway for astin C derivatives, in which plant enzymes or plant signals are required to trigger the synthesis of plant-exclusive variants such as astin A. Our findings lay the foundation for the sustainable biotechnological production of astins independent from aster plants.
Collapse
|
142
|
Wu J, Zhao L, Hu H, Li W, Li Y. Agonists and inhibitors of the STING pathway: Potential agents for immunotherapy. Med Res Rev 2019; 40:1117-1141. [DOI: 10.1002/med.21649] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/15/2019] [Accepted: 11/21/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Jun‐Jun Wu
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical BiologyDepartment of ChemistryTsinghua University Beijing China
| | - Lang Zhao
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical BiologyDepartment of ChemistryTsinghua University Beijing China
| | - Hong‐Guo Hu
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical BiologyDepartment of ChemistryTsinghua University Beijing China
| | - Wen‐Hao Li
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical BiologyDepartment of ChemistryTsinghua University Beijing China
| | - Yan‐Mei Li
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical BiologyDepartment of ChemistryTsinghua University Beijing China
- Beijing Institute for Brain Disorders Beijing China
- Center for Synthetic and Systems BiologyTsinghua University Beijing China
| |
Collapse
|
143
|
Sintim HO, Mikek CG, Wang M, Sooreshjani MA. Interrupting cyclic dinucleotide-cGAS-STING axis with small molecules. MEDCHEMCOMM 2019; 10:1999-2023. [PMID: 32206239 PMCID: PMC7069516 DOI: 10.1039/c8md00555a] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 08/13/2019] [Indexed: 12/19/2022]
Abstract
The cyclic dinucleotide-cGAS-STING axis plays important roles in host immunity. Activation of this signaling pathway, via cytosolic sensing of bacterial-derived c-di-GMP/c-di-AMP or host-derived cGAMP, leads to the production of inflammatory interferons and cytokines that help resolve infection. Small molecule activators of the cGAS-STING axis have the potential to augment immune response against various pathogens or cancer. The aberrant activation of this pathway, due to gain-of-function mutations in any of the proteins that are part of the signaling axis, could lead to various autoimmune diseases. Inhibiting various nodes of the cGAS-STING axis could provide relief to patients with autoimmune diseases. Many excellent reviews on the cGAS-STING axis have been published recently, and these have mainly focused on the molecular details of the cGAS-STING pathway. This review however focuses on small molecules that can be used to modulate various aspects of the cGAS-STING pathway, as well as other parallel inflammatory pathways.
Collapse
Affiliation(s)
- Herman O Sintim
- Department of Chemistry , Purdue University , 560 Oval Drive , West Lafayette , IN 47907 , USA .
- Institute for Drug Discovery , Purdue University , 720 Clinic Drive , West Lafayette , IN 47907 , USA
- Purdue Institute of Inflammation and Infectious Diseases , Purdue University , West Lafayette , IN 47907 , USA
| | - Clinton G Mikek
- Department of Chemistry , Purdue University , 560 Oval Drive , West Lafayette , IN 47907 , USA .
| | - Modi Wang
- Department of Chemistry , Purdue University , 560 Oval Drive , West Lafayette , IN 47907 , USA .
| | - Moloud A Sooreshjani
- Department of Chemistry , Purdue University , 560 Oval Drive , West Lafayette , IN 47907 , USA .
| |
Collapse
|
144
|
Feng X, Liu D, Li Z, Bian J. Bioactive modulators targeting STING adaptor in cGAS-STING pathway. Drug Discov Today 2019; 25:230-237. [PMID: 31758915 DOI: 10.1016/j.drudis.2019.11.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 11/05/2019] [Accepted: 11/14/2019] [Indexed: 12/21/2022]
Abstract
The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING)-pathway triggers innate immune responses by recognizing cytosolic DNA. Recent studies revealed that STING adaptor associates with various diseases, and several modulators targeting STING have been identified including three agonists that have entered clinical trials for treating cancer over the past 2 years. In particular, the efficacy of STING agonists and/or antagonists suggests adaptor STING as a potential therapeutic target for diverse diseases. Herein, we summarize the latest advances in understanding STING functioning and provide an overview of recent STING modulator discoveries, including structural details and the potential therapeutic applications of these modulators.
Collapse
Affiliation(s)
- Xi Feng
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Dongyu Liu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zhiyu Li
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Jinlei Bian
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
145
|
Cui X, Zhang R, Cen S, Zhou J. STING modulators: Predictive significance in drug discovery. Eur J Med Chem 2019; 182:111591. [PMID: 31419779 PMCID: PMC7172983 DOI: 10.1016/j.ejmech.2019.111591] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/10/2019] [Accepted: 08/05/2019] [Indexed: 12/19/2022]
Abstract
Cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS) - stimulator of interferon genes (STING) signaling pathway plays the critical role in the immune response to DNA. Pharmacological modulation of the STING pathway has been well characterized both from structural and functional perspectives, which paves the way for the drug design of small modulators by medicinal chemists. Here, we outline recent progress in studies on the STING pathway, the structure and biological function of STING, the STING related disease, as well as the rationale and progress in the development of STING modulators. Our review demonstrates that STING is a promising drug target, and providing clues for the discovery of novel STING agonists and antagonists for the potential treatment of various disease including microbial infectious diseases, cancer, and autoimmune disease.
Collapse
Affiliation(s)
- Xiangling Cui
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Department of Chemistry, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, PR China,Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing, China
| | - Rongyu Zhang
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Department of Chemistry, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, PR China,Drug Development and Innovation Center, College of Chemistry and Life Sciences, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, PR China
| | - Shan Cen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing, China
| | - Jinming Zhou
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Department of Chemistry, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, PR China; Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing, China; Drug Development and Innovation Center, College of Chemistry and Life Sciences, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, PR China.
| |
Collapse
|
146
|
The triggers of the cGAS-STING pathway and the connection with inflammatory and autoimmune diseases. INFECTION GENETICS AND EVOLUTION 2019; 77:104094. [PMID: 31689545 DOI: 10.1016/j.meegid.2019.104094] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/18/2019] [Accepted: 10/23/2019] [Indexed: 12/13/2022]
Abstract
Cyclic GMP-AMP synthase (cGAS) is a cytosolic nucleic acid sensor that can bind to dsDNA. It maintains an autoinhibited state in the absence of cytosolic dsDNA, while when activated, it in turn activates its adaptor protein STING, ultimately triggering a cascade that produces inflammatory cytokines and type I interferons (IFNs). With further research, additional types of nucleic acids have been found to be activators of the cGAS-STING pathway. The cGAS-STING pathway can provide protection or resistance against infections; however, improper or overactivation might cause severe inflammatory pathologies, including autoimmunity. This article systematically reviews the latest research progress on the axis, including categorical pathway triggers, the connection with autoimmune disease and drug therapy progress.
Collapse
|
147
|
Vanpouille-Box C, Hoffmann JA, Galluzzi L. Pharmacological modulation of nucleic acid sensors - therapeutic potential and persisting obstacles. Nat Rev Drug Discov 2019; 18:845-867. [PMID: 31554927 DOI: 10.1038/s41573-019-0043-2] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2019] [Indexed: 02/08/2023]
Abstract
Nucleic acid sensors, primarily TLR and RLR family members, as well as cGAS-STING signalling, play a critical role in the preservation of cellular and organismal homeostasis. Accordingly, deregulated nucleic acid sensing contributes to the origin of a diverse range of disorders, including infectious diseases, as well as cardiovascular, autoimmune and neoplastic conditions. Accumulating evidence indicates that normalizing aberrant nucleic acid sensing can mediate robust therapeutic effects. However, targeting nucleic acid sensors with pharmacological agents, such as STING agonists, presents multiple obstacles, including drug-, target-, disease- and host-related issues. Here, we discuss preclinical and clinical data supporting the potential of this therapeutic paradigm and highlight key limitations and possible strategies to overcome them.
Collapse
Affiliation(s)
- Claire Vanpouille-Box
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Jules A Hoffmann
- University of Strasbourg Institute for Advanced Studies, Strasbourg, France.,CNRS UPR 9022, Institute for Molecular and Cellular Biology, Strasbourg, France.,Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, China
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA. .,Sandra and Edward Meyer Cancer Center, New York, NY, USA. .,Department of Dermatology, Yale School of Medicine, New Haven, CT, USA. .,Université Paris Descartes, Paris, France.
| |
Collapse
|
148
|
Kumar V. A STING to inflammation and autoimmunity. J Leukoc Biol 2019; 106:171-185. [PMID: 30990921 DOI: 10.1002/jlb.4mir1018-397rr] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 03/08/2019] [Accepted: 03/11/2019] [Indexed: 12/19/2022] Open
Abstract
Various intracellular pattern recognition receptors (PRRs) recognize cytosolic pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). Cyclic GMP-AMP synthase (cGAS), a cytosolic PRR, recognizes cytosolic nucleic acids including dsDNAs. The recognition of dsDNA by cGAS generates cyclic GMP-AMP (GAMP). The cGAMP is then recognized by STING generating type 1 IFNs and NF-κB-mediated generation of pro-inflammatory cytokines and molecules. Thus, cGAS-STING signaling mediated recognition of cytosolic dsDNA causing the induction of type 1 IFNs plays a crucial role in innate immunity against cytosolic pathogens, PAMPs, and DAMPs. The overactivation of this system may lead to the development of autoinflammation and autoimmune diseases. The article opens with the introduction of different PRRs involved in the intracellular recognition of dsDNA and gives a brief introduction of cGAS-STING signaling. The second section briefly describes cGAS as intracellular PRR required to recognize intracellular nucleic acids (dsDNA and CDNs) and the formation of cGAMP. The cGAMP acts as a second messenger to activate STING- and TANK-binding kinase 1-mediated generation of type 1 IFNs and the activation of NF-κB. The third section of the article describes the role of cGAS-STING signaling in the induction of autoinflammation and various autoimmune diseases. The subsequent fourth section describes both chemical compounds developed and the endogenous negative regulators of cGAS-STING signaling required for its regulation. Therapeutic targeting of cGAS-STING signaling could offer new ways to treat inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Vijay Kumar
- Children's Health Queensland Clinical Unit, School of Clinical Medicine, Faculty of Medicine, Mater Research, University of Queensland, Brisbane, Queensland, Australia.,School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|