1
|
Xue L, Liu R, Qiu T, Zhuang H, Li H, Zhang L, Yin R, Jiang T. Design, synthesis, and activity evaluation of novel STING inhibitors based on C170 and H151. Eur J Med Chem 2025; 290:117533. [PMID: 40157312 DOI: 10.1016/j.ejmech.2025.117533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 03/15/2025] [Accepted: 03/18/2025] [Indexed: 04/01/2025]
Abstract
Stimulating the STING signaling pathway for immune system activation is considered a promising strategy for cancer treatment. However, activating the STING pathway can lead to adverse effects, as aberrant activation or specific mutations in STING may result in autoimmune and inflammatory diseases. Therefore, the development of STING inhibitors is equally important. In this study, we first introduced hydroxyl groups into the STING inhibitors C170 and H151, creating functional sites for further modification. Then the introduction of various substituents resulted in the identification of more potent inhibitors, Y2 and HY2, which effectively suppressed the activation of the STING pathway in THP1 and RAW264.7 cells. Compounds Y2 and HY2 demonstrated potent anti-inflammatory effects in mice cisplatin-induced acute kidney injury models by inhibiting the STING pathway. Collectively, Y2 and HY2 warrant further investigation as novel anti-inflammatory agents.
Collapse
Affiliation(s)
- Liang Xue
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| | - Ruixue Liu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| | - Tingting Qiu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| | - Huiying Zhuang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| | - Hongwei Li
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| | - Lican Zhang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| | - Ruijuan Yin
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237, China; Marine Biomedical Research Institute of Qingdao, Qingdao, 266237, China.
| | - Tao Jiang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao, 266237, China; Center for Innovative Drug Discovery, Greater Bay Area Institute of Precision Medicine (Guangzhou), Guangzhou, 511455, China.
| |
Collapse
|
2
|
Liu Y, Carmona‐Rivera C, Seto NL, Oliveira CB, Patino‐Martinez E, Baumer Y, Powell‐Wiley TM, Mehta N, Hasni S, Zhang X, Kaplan MJ. Role of STING Deficiency in Amelioration of Mouse Models of Lupus and Atherosclerosis. Arthritis Rheumatol 2025; 77:547-559. [PMID: 39605244 PMCID: PMC12039466 DOI: 10.1002/art.43062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/10/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024]
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE) is a systemic autoimmune syndrome characterized by autoreactive responses to nucleic acids, dysregulation of the type I interferon (IFN-I) pathway, and accelerated atherosclerosis. The stimulator of IFN genes (STING), a cytosolic DNA sensor, has pathogenic implications in various inflammatory diseases. However, its specific role in SLE pathogenesis, particularly in tissue damage, remains unclear. This study aimed to elucidate the role of STING in murine models of Toll-like receptor 7 (TLR7)-driven lupus and atherosclerosis. METHODS A TLR7-driven lupus model was induced using imiquimod (IMQ) in wild-type (WT) and STING knockout (Sting1-/-) mice on a B6 background. Mice were assessed for organ involvement, serum autoantibodies, and innate and adaptive immune responses. Additionally, Sting1-/- mice were backcrossed to apolipoprotein E knockout (Apoe-/-) mice, and both Apoe-/- and Apoe-/-Sting1-/- mice were fed a high-fat chow diet to induce atherosclerosis. Phenotypic assessments were conducted. RESULTS Compared with IMQ-treated WT mice, Sting1-/- mice exhibited reduced disease severity in the lupus-like phenotype, characterized by decreased splenomegaly, lower renal immune complex deposition and renal damage, diminished expansion of myeloid cells, and reduced activation of T and B lymphocytes. IMQ-induced DNA release associated with IFN-β production and subsequent IFN-induced responses were attenuated in Sting1-/- mice. DNase I treatment mitigated IMQ-induced proinflammatory responses in WT mice but had no effect in Sting1-/- mice. Furthermore, STING deficiency conferred protection against vascular damage and reduced atherosclerosis burden, accompanied by decreased IFN-I production. Human monocyte-derived macrophages treated with IFN-I significantly internalized more acetylated low-density lipoprotein when compared with untreated cells, whereas an association between oxidized nucleic acids and disease activity and vascular damage was found in human SLE. CONCLUSION These findings highlight a pathogenic role of STING and downstream IFN responses in TLR7-driven autoimmunity, vascular damage and atherosclerosis, supporting a therapeutic potential for STING inhibition in SLE treatment. Further research is warranted to elucidate the mechanisms underlying STING's involvement in these processes and to explore the feasibility of targeting STING as a therapeutic strategy in SLE and related autoimmune disorders.
Collapse
Affiliation(s)
- Yudong Liu
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China, and National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIHBethesdaMaryland
| | - Carmelo Carmona‐Rivera
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIHBethesdaMaryland
| | - Nickie L. Seto
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIHBethesdaMaryland
| | | | | | - Yvonne Baumer
- National Heart Lung and Blood Institute, NIHBethesdaMaryland
| | - Tiffany M. Powell‐Wiley
- National Heart Lung and Blood Institute, NIH and National Institute on Minority Health and Health Disparities, NIHBethesdaMaryland
| | - Nehal Mehta
- National Heart Lung and Blood Institute, NIHBethesdaMaryland
| | - Sarfaraz Hasni
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIHBethesdaMaryland
| | - Xuan Zhang
- Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Graduate School of Peking Union Medical College, Chinese Academy of Medical SciencesBeijingChina
| | - Mariana J. Kaplan
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIHBethesdaMaryland
| |
Collapse
|
3
|
Shen A, Wang X, Chen Q, Zhang Y, Wang F, Li Y, Liu Z, Deng L, Ouyang W, Geng M, Song Z, Xie Z, Zhang A. Discovery of Potent STING Inhibitors Bearing a Difluorobenzodioxol Structural Motif as Potent Anti-Inflammatory Agents. J Med Chem 2025; 68:8907-8932. [PMID: 40188441 DOI: 10.1021/acs.jmedchem.5c00580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2025]
Abstract
Given the critical role of STING in autoimmune and inflammatory disorders, the development of targeted small-molecule inhibitors has been a promising strategy for the treatment of these diseases. Nevertheless, the currently reported STING inhibitors suffer from limited structural diversity, species sensitivity, and poor activity; therefore, none are suitable for clinical investigation. Herein, we performed a structural modification campaign on the tool compound 6 (H-151) based on its potential metabolic hotspots. Compound 66, bearing a difluorobenzodioxol moiety, was identified as one of the most potent STING inhibitors with IC50 values of 116 and 96.3 nM for h- and m-STING, respectively. This compound exhibited a notable enhancement in metabolic properties, especially in terms of metabolic stability. A mechanism study verified that 66 engaged with STING in a covalent manner akin to that of 6. In both the cisplatin-induced acute kidney injury and TREX1 D18N mouse models, 66 significantly alleviated tissue injury and inflammation.
Collapse
Affiliation(s)
- Ancheng Shen
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- State Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Xiyuan Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qingxuan Chen
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- State Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China
| | - Yan Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Fang Wang
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yuqiang Li
- Shanghai Artificial Intelligence Laboratory, Shanghai 200433, China
| | - Zhiguo Liu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Liufu Deng
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- State Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China
| | - Wanli Ouyang
- Shanghai Artificial Intelligence Laboratory, Shanghai 200433, China
| | - Meiyu Geng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
| | - Zilan Song
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- State Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China
| | - Zuoquan Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ao Zhang
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- State Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Shanghai Artificial Intelligence Laboratory, Shanghai 200433, China
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
| |
Collapse
|
4
|
Feng Q, Xu X, Zhang S. cGAS-STING pathway in systemic lupus erythematosus: biological implications and therapeutic opportunities. Immunol Res 2024; 72:1207-1216. [PMID: 39096420 DOI: 10.1007/s12026-024-09525-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 07/25/2024] [Indexed: 08/05/2024]
Abstract
The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway has been identified as a significant modulator of inflammation in various clinical contexts, including infection, cellular stress, and tissue injury. The extensive participation of the cGAS-STING pathway can be attributed to its ability to detect and control the cellular reaction to DNAs originating from both microorganisms and hosts. These DNAs are well recognized as molecules linked with potential risks. At physiological levels, the STING signaling system exhibits protective effects. However, prolonged stimulation of this pathway contributes to autoimmune disorder pathogenesis. The present paper provides an overview of the activation mechanism of the cGAS-STING signaling pathways and their associated significant functions, as well as therapeutic interventions in the context of systemic lupus erythematosus (SLE). The primary objective is to enhance our comprehension of SLE and facilitate more effective diagnosis and treatment strategies for this condition.
Collapse
Affiliation(s)
- Qun Feng
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, 130017, China
| | - Xiaolin Xu
- Cardiology Department, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Shoulin Zhang
- Nephropathy Department, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130021, China.
| |
Collapse
|
5
|
Ribeiro ARS, Neuper T, Horejs-Hoeck J. The Role of STING-Mediated Activation of Dendritic Cells in Cancer Immunotherapy. Int J Nanomedicine 2024; 19:10685-10697. [PMID: 39464674 PMCID: PMC11512692 DOI: 10.2147/ijn.s477320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 09/16/2024] [Indexed: 10/29/2024] Open
Abstract
The signaling pathway that comprises cyclic guanosine monophosphate-adenosine monophosphate (cGAMP or GMP-AMP) synthase (cGAS) and Stimulator of Interferon Genes (STING) is emerging as a druggable target for immunotherapy, with tumor-resident dendritic cells (DC) playing a critical role in mediating its effects. The STING receptor is part of the DNA-sensing cellular machinery, that can trigger the secretion of pro-inflammatory mediators, priming effector T cells and initiating specific antitumor responses. Yet, recent studies have highlighted the dual role of STING activation in the context of cancer: STING can either promote antitumor responses or enhance tumor progression. This dichotomy often depends on the cell type in which cGAS-STING signaling is induced and the activation mode, namely acute versus chronic. Of note, STING activation at the DC level appears to be particularly important for tumor eradication. This review outlines the contribution of the different conventional and plasmacytoid DC subsets and describes the mechanisms underlying STING-mediated activation of DCs in cancer. We further highlight how the STING pathway plays an intricate role in modulating the function of DCs embedded in tumor tissue. Additionally, we discuss the strategies being employed to harness STING activation for cancer treatment, such as the development of synthetic agonists and nano-based delivery systems, spotlighting the current techniques used to prompt STING engagement specifically in DCs.
Collapse
Affiliation(s)
- Ana R S Ribeiro
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg (CCS), Salzburg, 5020, Austria
| | - Theresa Neuper
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg (CCS), Salzburg, 5020, Austria
- Center for Tumor biology and Immunology (CTBI), Salzburg, 5020, Austria
| | - Jutta Horejs-Hoeck
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
- Cancer Cluster Salzburg (CCS), Salzburg, 5020, Austria
- Center for Tumor biology and Immunology (CTBI), Salzburg, 5020, Austria
| |
Collapse
|
6
|
Yang Y, Ren C, Xu X, Yang X, Shao W. Decoding the connection between SLE and DNA Sensors: A comprehensive review. Int Immunopharmacol 2024; 137:112446. [PMID: 38878488 DOI: 10.1016/j.intimp.2024.112446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/06/2024] [Accepted: 06/06/2024] [Indexed: 07/11/2024]
Abstract
Systemic lupus erythematosus (SLE) is recognized as a prevalent autoimmune disorder characterized by a multifaceted pathogenesis potentially influenced by a combination of environmental factors, genetic predisposition, and hormonal regulation. The continuous study of immune system activation is especially intriguing. Analysis of blood samples from individuals with SLE reveals an abnormal increase in interferon levels, along with the existence of anti-double-stranded DNA antibodies. This evidence suggests that the development of SLE may be initiated by innate immunity. The presence of abnormal dsDNA fragments can activate DNA sensors within cells, particularly immune cells, leading to the initiation of downstream signaling cascades that result in the upregulation of relevant cytokines and the subsequent initiation of adaptive immune responses, such as B cell differentiation and T cell activation. The intricate pathogenesis of SLE results in DNA sensors exhibiting a wide range of functions in innate immune responses that are subject to variation based on cell types, developmental processes, downstream effector signaling pathways and other factors. The review aims to reorganize how DNA sensors influence signaling pathways and contribute to the development of SLE according to current studies, with the aspiration of furnishing valuable insights for future investigations into the pathological mechanisms of SLE and potential treatment approaches.
Collapse
Affiliation(s)
- Yuxiang Yang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China; Medical School of Tianjin University, Tianjin, China; School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Changhuai Ren
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China; Medical School of Tianjin University, Tianjin, China
| | - Xiaopeng Xu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China; Medical School of Tianjin University, Tianjin, China
| | - Xinyi Yang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China; Medical School of Tianjin University, Tianjin, China; School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Wenwei Shao
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China; Medical School of Tianjin University, Tianjin, China; State Key Laboratory of Advanced Medical Materials and Devices, Tianjin University, Tianjin, China.
| |
Collapse
|
7
|
Tsokos GC. The immunology of systemic lupus erythematosus. Nat Immunol 2024; 25:1332-1343. [PMID: 39009839 DOI: 10.1038/s41590-024-01898-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/17/2024] [Indexed: 07/17/2024]
Abstract
Understanding the pathogenesis and clinical manifestations of systemic lupus erythematosus (SLE) has been a great challenge. Reductionist approaches to understand the nature of the disease have identified many pathogenetic contributors that parallel clinical heterogeneity. This Review outlines the immunological control of SLE and looks to experimental tools and approaches that are improving our understanding of the complex contribution of interacting genetics, environment, sex and immunoregulatory factors and their interface with processes inherent to tissue parenchymal cells. Efforts to advance precision medicine in the care of patients with SLE along with treatment strategies to correct the immune system hold hope and are also examined.
Collapse
Affiliation(s)
- George C Tsokos
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
8
|
Lin W, Szabo C, Liu T, Tao H, Wu X, Wu J. STING trafficking activates MAPK-CREB signaling to trigger regulatory T cell differentiation. Proc Natl Acad Sci U S A 2024; 121:e2320709121. [PMID: 38985760 PMCID: PMC11260101 DOI: 10.1073/pnas.2320709121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 06/12/2024] [Indexed: 07/12/2024] Open
Abstract
The Type-I interferon (IFN-I) response is the major outcome of stimulator of interferon genes (STING) activation in innate cells. STING is more abundantly expressed in adaptive T cells; nevertheless, its intrinsic function in T cells remains unclear. Intriguingly, we previously demonstrated that STING activation in T cells activates widespread IFN-independent activities, which stands in contrast to the well-known STING-mediated IFN response. Here, we have identified that STING activation induces regulatory T cells (Tregs) differentiation independently of IRF3 and IFN. Specifically, the translocation of STING from the endoplasmic reticulum to the Golgi activates mitogen-activated protein kinase (MAPK) activity, which subsequently triggers transcription factor cAMP response element-binding protein (CREB) activation. The activation of the STING-MAPK-CREB signaling pathway induces the expression of many cytokine genes, including interleukin-2 (IL-2) and transforming growth factor-beta 2 (TGF-β2), to promote the Treg differentiation. Genetic knockdown of MAPK p38 or pharmacological inhibition of MAPK p38 or CREB markedly inhibits STING-mediated Treg differentiation. Administration of the STING agonist also promotes Treg differentiation in mice. In the Trex1-/- autoimmune disease mouse model, we demonstrate that intrinsic STING activation in CD4+ T cells can drive Treg differentiation, potentially counterbalancing the autoimmunity associated with Trex1 deficiency. Thus, STING-MAPK-CREB represents an IFN-independent signaling axis of STING that may have profound effects on T cell effector function and adaptive immunity.
Collapse
Affiliation(s)
- Wei Lin
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH44195
| | - Claudia Szabo
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH44195
| | - Tao Liu
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH44195
| | - Huangheng Tao
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH44195
| | - Xianfang Wu
- Infection Biology Program, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH44195
| | - Jianjun Wu
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH44195
| |
Collapse
|
9
|
Chang L. Harnessing cGAS-STING axis for therapeutic benefits in systemic lupus erythematosus. Int J Rheum Dis 2024; 27:e15256. [PMID: 38982864 DOI: 10.1111/1756-185x.15256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/17/2024] [Accepted: 06/21/2024] [Indexed: 07/11/2024]
Abstract
The cyclic GMP-AMP synthase (cGAS), a prominent intracellular DNA sensor in mammalian cells, controls the innate immune response and the stimulator of interferon genes (STING)-mediated synthesis of pro-inflammatory cytokines, such as type-I interferon (IFN-I). For decades, IFN-I has been hypothesized to be essential in the development of systemic lupus erythematosus (SLE), a chronic multisystem autoimmunity characterized by immune complex (IC) deposition in small vessels. Recent findings revealed that the activation of the cGAS-STING pathway by self-DNA would propagate the autoimmune responses via upregulating IFN-I production in SLE. In this review, we aimed to provide a comprehensive outlook of the role of the cGAS-STING pathway in SLE pathobiology, as well as, a better understanding of current therapeutic opportunities targeting this axis.
Collapse
Affiliation(s)
- Liu Chang
- Department of Rheumatology, Henan Provincial Hospital of Traditional Chinese Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
10
|
Vejvisithsakul PP, Thumarat C, Leelahavanichkul A, Hirankan N, Pisitkun T, Paludan SR, Pisitkun P. Elucidating the function of STING in systemic lupus erythematosus through the STING Goldenticket mouse mutant. Sci Rep 2024; 14:13968. [PMID: 38886451 PMCID: PMC11183220 DOI: 10.1038/s41598-024-64495-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 06/09/2024] [Indexed: 06/20/2024] Open
Abstract
The complexity of systemic lupus erythematosus (SLE) arises from intricate genetic and environmental interactions, with STING playing a pivotal role. This study aims to comprehend the function of STING using the pristane-induced lupus (PIL) model in Sting missense mutant mice (Goldenticket or StingGt), which contrasts with previous research using Sting knockout mice. Investigating two-month-old StingGt mice over six months post-PIL induction, we observed a profound reduction in autoimmune markers, including antinuclear and anti-dsDNA antibodies, germinal center B cells, and plasma cells, compared to their wild-type counterparts. A pivotal finding was the marked decrease in IL-17-producing T cells. Notably, the severity of lupus nephritis and pulmonary hemorrhages was significantly diminished in StingGt mice. These findings demonstrate that different genetic approaches to interfere with STING signaling can lead to contrasting outcomes in SLE pathogenesis, which highlights the need for a nuanced understanding of the role of STING in drug development for SLE. In summary, the loss of Sting function in Goldenticket mutant mice rescued autoimmune phenotypes in PIL. This study reveals the critical nature of STING in SLE, suggesting that the method of STING modulation significantly influences disease phenotypes and should be a key consideration in developing targeted therapies.
Collapse
Affiliation(s)
- Pichpisith Pierre Vejvisithsakul
- Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Chisanu Thumarat
- Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Asada Leelahavanichkul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Translational Research in Inflammation and Immunology Research Unit (TRIRU), Department of Microbiology, Chulalongkorn University, Bangkok, Thailand
| | - Nattiya Hirankan
- Centre of Excellent in Immunology and Immune-Mediated Diseases, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Trairak Pisitkun
- Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Prapaporn Pisitkun
- Program in 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.
| |
Collapse
|
11
|
Alee I, Chantawichitwong P, Leelahavanichkul A, Paludan SR, Pisitkun T, Pisitkun P. The STING inhibitor (ISD-017) reduces glomerulonephritis in 129.B6.Fcgr2b-deficient mice. Sci Rep 2024; 14:11020. [PMID: 38745067 PMCID: PMC11094069 DOI: 10.1038/s41598-024-61597-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 05/07/2024] [Indexed: 05/16/2024] Open
Abstract
The absence of stimulator of interferon genes (STING) in 129.B6.Fcgr2b-deficient mice rescue lupus phenotypes. The administration of a STING inhibitor (ISD017) into the young 129.B6.Fcgr2b-deficient mice prevents lupus nephritis development. This study mainly aimed to evaluate the effects of STING inhibition (ISD107) on established SLE in mice to prove that ISD017 could be a good therapeutic drug to reverse the already set-up autoimmunity and kidney impairment. Twenty-four-week-old Fcgr2b-deficient mice were treated with cyclophosphamide (25 mg/kg, intraperitoneal, once per week), ISD017 (10 mg/kg, intraperitoneal, three times per week), or control vehicle for 8 weeks, and were analyzed for phenotypes. Both ISD017 and cyclophosphamide treatment increased long-term survival and reduced the severity of glomerulonephritis in Fcgr2b-deficient mice. While cyclophosphamide reduced activated B cells (B220+GL-7+), ISD017 decreased activated T cells (CD4+CD69+) and neutrophils (Ly6c+Ly6g+) in Fcgr2b-deficient mice. In addition, ISD017 reduced IL-1β and interferon-inducible genes. In summary, ISD017 treatment in symptomatic 129.B6.Fcgr2b-deficient mice reduced the severity of glomerulonephritis and increased long-term survival. ISD017 worked comparably to cyclophosphamide for treating lupus nephritis in 129.B6.Fcgr2b-deficient mice. ISD017 reduced activated T cells and neutrophils, while cyclophosphamide targeted activated B cells. These results suggested that STING inhibitors can potentially be a new therapeutic drug for treating lupus.
Collapse
Affiliation(s)
- Isara Alee
- Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Medical Sciences Program, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Papasara Chantawichitwong
- Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Graduated Program in Molecular Medicine, Faculty of Science, Mahidol University, Salaya, Thailand
| | - Asada Leelahavanichkul
- Center of Excellence in Translational Research in Inflammation and Immunology (CETRII), Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Søren R Paludan
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Trairak Pisitkun
- Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
- Epithelial Systems Biology Laboratory, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Prapaporn Pisitkun
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
12
|
Hopkins JW, Sulka KB, Sawden M, Carroll KA, Brown RD, Bunnell SC, Poltorak A, Tai A, Reed ER, Sharma S. STING promotes homeostatic maintenance of tissues and confers longevity with aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.04.588107. [PMID: 38645182 PMCID: PMC11030237 DOI: 10.1101/2024.04.04.588107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Local immune processes within aging tissues are a significant driver of aging associated dysfunction, but tissue-autonomous pathways and cell types that modulate these responses remain poorly characterized. The cytosolic DNA sensing pathway, acting through cyclic GMP-AMP synthase (cGAS) and Stimulator of Interferon Genes (STING), is broadly expressed in tissues, and is poised to regulate local type I interferon (IFN-I)-dependent and independent inflammatory processes within tissues. Recent studies suggest that the cGAS/STING pathway may drive pathology in various in vitro and in vivo models of accelerated aging. To date, however, the role of the cGAS/STING pathway in physiological aging processes, in the absence of genetic drivers, has remained unexplored. This remains a relevant gap, as STING is ubiquitously expressed, implicated in multitudinous disorders, and loss of function polymorphisms of STING are highly prevalent in the human population (>50%). Here we reveal that, during physiological aging, STING-deficiency leads to a significant shortening of murine lifespan, increased pro-inflammatory serum cytokines and tissue infiltrates, as well as salient changes in histological composition and organization. We note that aging hearts, livers, and kidneys express distinct subsets of inflammatory, interferon-stimulated gene (ISG), and senescence genes, collectively comprising an immune fingerprint for each tissue. These distinctive patterns are largely imprinted by tissue-specific stromal and myeloid cells. Using cellular interaction network analyses, immunofluorescence, and histopathology data, we show that these immune fingerprints shape the tissue architecture and the landscape of cell-cell interactions in aging tissues. These age-associated immune fingerprints are grossly dysregulated with STING-deficiency, with key genes that define aging STING-sufficient tissues greatly diminished in the absence of STING. Changes in immune signatures are concomitant with a restructuring of the stromal and myeloid fractions, whereby cell:cell interactions are grossly altered and resulting in disorganization of tissue architecture in STING-deficient organs. This altered homeostasis in aging STING-deficient tissues is associated with a cross-tissue loss of homeostatic tissue-resident macrophage (TRM) populations in these tissues. Ex vivo analyses reveal that basal STING-signaling limits the susceptibility of TRMs to death-inducing stimuli and determines their in situ localization in tissue niches, thereby promoting tissue homeostasis. Collectively, these data upend the paradigm that cGAS/STING signaling is primarily pathological in aging and instead indicate that basal STING signaling sustains tissue function and supports organismal longevity. Critically, our study urges caution in the indiscriminate targeting of these pathways, which may result in unpredictable and pathological consequences for health during aging.
Collapse
Affiliation(s)
- Jacob W. Hopkins
- Department of Immunology, Tufts University, Boston, MA 02111
- Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111
| | - Katherine B. Sulka
- Department of Immunology, Tufts University, Boston, MA 02111
- Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111
| | - Machlan Sawden
- Department of Immunology, Tufts University, Boston, MA 02111
- Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111
| | - Kimberly A. Carroll
- Department of Immunology, Tufts University, Boston, MA 02111
- Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111
| | - Ronald D. Brown
- Department of Neurobiology and Behavior, Cornell University, Ithaca, NY, 12853
| | | | | | - Albert Tai
- Department of Immunology, Tufts University, Boston, MA 02111
- Data Intensive Studies Center, Tufts University, Medford, MA, 02155
| | - Eric R. Reed
- Data Intensive Studies Center, Tufts University, Medford, MA, 02155
| | - Shruti Sharma
- Department of Immunology, Tufts University, Boston, MA 02111
| |
Collapse
|
13
|
Li Q, Wu P, Du Q, Hanif U, Hu H, Li K. cGAS-STING, an important signaling pathway in diseases and their therapy. MedComm (Beijing) 2024; 5:e511. [PMID: 38525112 PMCID: PMC10960729 DOI: 10.1002/mco2.511] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 02/15/2024] [Accepted: 02/21/2024] [Indexed: 03/26/2024] Open
Abstract
Since cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway was discovered in 2013, great progress has been made to elucidate the origin, function, and regulating mechanism of cGAS-STING signaling pathway in the past decade. Meanwhile, the triggering and transduction mechanisms have been continuously illuminated. cGAS-STING plays a key role in human diseases, particularly DNA-triggered inflammatory diseases, making it a potentially effective therapeutic target for inflammation-related diseases. Here, we aim to summarize the ancient origin of the cGAS-STING defense mechanism, as well as the triggers, transduction, and regulating mechanisms of the cGAS-STING. We will also focus on the important roles of cGAS-STING signal under pathological conditions, such as infections, cancers, autoimmune diseases, neurological diseases, and visceral inflammations, and review the progress in drug development targeting cGAS-STING signaling pathway. The main directions and potential obstacles in the regulating mechanism research and therapeutic drug development of the cGAS-STING signaling pathway for inflammatory diseases and cancers will be discussed. These research advancements expand our understanding of cGAS-STING, provide a theoretical basis for further exploration of the roles of cGAS-STING in diseases, and open up new strategies for targeting cGAS-STING as a promising therapeutic intervention in multiple diseases.
Collapse
Affiliation(s)
- Qijie Li
- Sichuan province Medical and Engineering Interdisciplinary Research Center of Nursing & Materials/Nursing Key Laboratory of Sichuan ProvinceWest China Hospital, Sichuan University/West China School of NursingSichuan UniversityChengduSichuanChina
| | - Ping Wu
- Department of Occupational DiseasesThe Second Affiliated Hospital of Chengdu Medical College (China National Nuclear Corporation 416 Hospital)ChengduSichuanChina
| | - Qiujing Du
- Sichuan province Medical and Engineering Interdisciplinary Research Center of Nursing & Materials/Nursing Key Laboratory of Sichuan ProvinceWest China Hospital, Sichuan University/West China School of NursingSichuan UniversityChengduSichuanChina
| | - Ullah Hanif
- Sichuan province Medical and Engineering Interdisciplinary Research Center of Nursing & Materials/Nursing Key Laboratory of Sichuan ProvinceWest China Hospital, Sichuan University/West China School of NursingSichuan UniversityChengduSichuanChina
| | - Hongbo Hu
- Center for Immunology and HematologyState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduSichuanChina
| | - Ka Li
- Sichuan province Medical and Engineering Interdisciplinary Research Center of Nursing & Materials/Nursing Key Laboratory of Sichuan ProvinceWest China Hospital, Sichuan University/West China School of NursingSichuan UniversityChengduSichuanChina
| |
Collapse
|
14
|
Wolf C, Lim EL, Mokhtari M, Kind B, Odainic A, Lara-Villacanas E, Koss S, Mages S, Menzel K, Engel K, Dückers G, Bernbeck B, Schneider DT, Siepermann K, Niehues T, Goetzke CC, Durek P, Minden K, Dörner T, Stittrich A, Szelinski F, Guerra GM, Massoud M, Bieringer M, de Oliveira Mann CC, Beltrán E, Kallinich T, Mashreghi MF, Schmidt SV, Latz E, Klughammer J, Majer O, Lee-Kirsch MA. UNC93B1 variants underlie TLR7-dependent autoimmunity. Sci Immunol 2024; 9:eadi9769. [PMID: 38207055 DOI: 10.1126/sciimmunol.adi9769] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 12/22/2023] [Indexed: 01/13/2024]
Abstract
UNC93B1 is critical for trafficking and function of nucleic acid-sensing Toll-like receptors (TLRs) TLR3, TLR7, TLR8, and TLR9, which are essential for antiviral immunity. Overactive TLR7 signaling induced by recognition of self-nucleic acids has been implicated in systemic lupus erythematosus (SLE). Here, we report UNC93B1 variants (E92G and R336L) in four patients with early-onset SLE. Patient cells or mouse macrophages carrying the UNC93B1 variants produced high amounts of TNF-α and IL-6 and upon stimulation with TLR7/TLR8 agonist, but not with TLR3 or TLR9 agonists. E92G causes UNC93B1 protein instability and reduced interaction with TLR7, leading to selective TLR7 hyperactivation with constitutive type I IFN signaling. Thus, UNC93B1 regulates TLR subtype-specific mechanisms of ligand recognition. Our findings establish a pivotal role for UNC93B1 in TLR7-dependent autoimmunity and highlight the therapeutic potential of targeting TLR7 in SLE.
Collapse
Affiliation(s)
- Christine Wolf
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany
| | - Ee Lyn Lim
- Max Planck Institute for Infection Biology, Berlin 10117, Germany
| | - Mohammad Mokhtari
- Gene Center, Systems Immunology, Ludwig-Maximilians-Universität Munich, Munich 81377, Germany
| | - Barbara Kind
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany
| | - Alexandru Odainic
- Institute of Innate Immunity, University of Bonn, Bonn 53127, Germany
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection & Immunity, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Eusebia Lara-Villacanas
- Department of Pediatrics, Klinikum Dortmund, University Witten/Herdecke, Dortmund 44145, Germany
| | - Sarah Koss
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany
| | - Simon Mages
- Gene Center, Systems Immunology, Ludwig-Maximilians-Universität Munich, Munich 81377, Germany
| | - Katharina Menzel
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany
| | - Kerstin Engel
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany
| | - Gregor Dückers
- Department of Pediatrics, Helios Klinik Krefeld, Krefeld 47805, Germany
| | - Benedikt Bernbeck
- Department of Pediatrics, Klinikum Dortmund, University Witten/Herdecke, Dortmund 44145, Germany
| | - Dominik T Schneider
- Department of Pediatrics, Klinikum Dortmund, University Witten/Herdecke, Dortmund 44145, Germany
| | | | - Tim Niehues
- Department of Pediatrics, Helios Klinik Krefeld, Krefeld 47805, Germany
| | - Carl Christoph Goetzke
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, Berlin 10117, Germany
- Deutsches Rheuma-Forschungszentrum (DRFZ), an institute of the Leibniz Association, Berlin 10117, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin 10178, Germany
| | - Pawel Durek
- Deutsches Rheuma-Forschungszentrum (DRFZ), an institute of the Leibniz Association, Berlin 10117, Germany
| | - Kirsten Minden
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, Berlin 10117, Germany
- Deutsches Rheuma-Forschungszentrum (DRFZ), an institute of the Leibniz Association, Berlin 10117, Germany
| | - Thomas Dörner
- Deutsches Rheuma-Forschungszentrum (DRFZ), an institute of the Leibniz Association, Berlin 10117, Germany
- Department of Medicine, Rheumatology and Clinical Immunology, Charite-Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Anna Stittrich
- Labor Berlin Charité-Vivantes GmbH, Department of Human Genetics, Berlin 13353, Germany
| | - Franziska Szelinski
- Deutsches Rheuma-Forschungszentrum (DRFZ), an institute of the Leibniz Association, Berlin 10117, Germany
- Department of Medicine, Rheumatology and Clinical Immunology, Charite-Universitätsmedizin Berlin, Berlin 10117, Germany
| | - Gabriela Maria Guerra
- Deutsches Rheuma-Forschungszentrum (DRFZ), an institute of the Leibniz Association, Berlin 10117, Germany
| | - Mona Massoud
- Deutsches Rheuma-Forschungszentrum (DRFZ), an institute of the Leibniz Association, Berlin 10117, Germany
| | - Markus Bieringer
- Department of Cardiology and Nephrology, HELIOS Klinikum Berlin-Buch, Berlin 13125, Germany
| | | | - Eduardo Beltrán
- Institute for Clinical Neuroimmunology, BioMedizinisches Zentrum, Ludwig-Maximilians-Universität Munich, Munich 82152, Germany
| | - Tilmann Kallinich
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, Berlin 10117, Germany
- Deutsches Rheuma-Forschungszentrum (DRFZ), an institute of the Leibniz Association, Berlin 10117, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin 10178, Germany
| | - Mir-Farzin Mashreghi
- Deutsches Rheuma-Forschungszentrum (DRFZ), an institute of the Leibniz Association, Berlin 10117, Germany
| | - Susanne V Schmidt
- Institute of Innate Immunity, University of Bonn, Bonn 53127, Germany
| | - Eicke Latz
- Institute of Innate Immunity, University of Bonn, Bonn 53127, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn 53175, Germany
| | - Johanna Klughammer
- Gene Center, Systems Immunology, Ludwig-Maximilians-Universität Munich, Munich 81377, Germany
| | - Olivia Majer
- Max Planck Institute for Infection Biology, Berlin 10117, Germany
| | - Min Ae Lee-Kirsch
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany
- University Center for Rare Diseases, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany
| |
Collapse
|
15
|
Mi X, Lai K, Yan L, Wu H, Wei S. A comprehensive analysis of type 1 interferon gene signatures in systematic lupus erythematosus and prediction of the crucial susceptible factor for Sjögren syndrome. Clin Exp Med 2023; 23:4731-4743. [PMID: 37672133 DOI: 10.1007/s10238-023-01154-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 07/23/2023] [Indexed: 09/07/2023]
Abstract
This study aimed to determine the role of IFN-1 gene signatures in SLE and their association with Sjögren syndrome (SS). Publicly available data from the Gene Expression Omnibus database were used to construct the models. The random forest tree model was used to screen key IFN-1 gene signatures, and consensus clustering algorithms were used for unsupervised cluster analysis of these signatures. CIBERSORT and gene set variation analyses were used to evaluate the relative immune cell infiltration and enriched molecular pathways of the samples, respectively. Weighted gene co-expression network analysis was used to identify the co-expression modules and hub genes. Finally, univariate and multivariate logistic regression models were used to evaluate differences in clinical and laboratory characteristics between the different groups. The role of IFN-1 gene signatures in SLE was comprehensively assessed, which revealed an IFN-1 gene signature including six genes that could easily distinguish SLE patients and healthy individuals and identified two distinct IFN-1 subtypes exhibiting significant differences in clinical characteristics, immune microenvironment, and biological functional pathways. The SLE disease activity index, lower lymphocyte count, nucleotide oligomerization domain (NOD)-like receptor signaling pathway, and dendritic cell activation were strongly correlated with the IFN-1 gene signatures. In addition, we found that IFN-1 gene signatures in SLE may be an important susceptibility factor for SS, and the NOD-like receptor signaling pathway was identified as a common pathway. This study provides a comprehensive evaluation of the IFN-1 gene signatures, which may provide a new direction for the understanding of SLE and SS and help in the selection of optimal strategies for personalized immunotherapy.
Collapse
Affiliation(s)
- Xiangbin Mi
- Department of Dermatology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Kuan Lai
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Lu Yan
- Department of Dermatology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Hang Wu
- Department of Dermatology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shanshan Wei
- Department of Dermatology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
16
|
Liu Y, Wei FZ, Zhan YW, Wang R, Mo BY, Lin SD. TLR9 regulates the autophagy-lysosome pathway to promote dendritic cell maturation and activation by activating the TRAF6-cGAS-STING pathway. Kaohsiung J Med Sci 2023; 39:1200-1212. [PMID: 37850718 DOI: 10.1002/kjm2.12769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 10/19/2023] Open
Abstract
Dysregulated maturation and activation of dendritic cells (DCs) play a significant role in the progression of systemic lupus erythematosus (SLE). The autophagy-lysosome pathway has been identified as a potential mechanism to inhibit DC activation and maturation, but its precise workings remain unclear. We investigated the role and regulatory mechanism of TLR9 in modulating the autophagy-lysosome pathway and DCs activation. The mRNA and protein expressions were assessed using qRT-PCR and/or western blot. NZBW/F1 mice was used to construct a lupus nephritis (LN) model in vivo. Cell apoptosis was analyzed by TUNEL staining. Flow cytometry was adopted to analyze DCs surface markers. Lyso-tracker red staining was employed to analyze lysosome acidification. Levels of anti-dsDNA, cytokines, C3, C4, urine protein and urine creatinine were examined by ELISA. The results showed that TLR9 was markedly increased in SLE patients, and its expression was positively correlated with SLEDAI scores and dsDNA level. Conversely, TLR9 expression showed a negative correlation with C3 and C4 levels. Loss-of function experiments demonstrated that TLR9 depletion exerted a substantial inhibition of renal injury, inflammation, and DCs numbers. Additionally, upregulation of TLR9 promoted DCs maturation and activation through activation of autophagy and lysosome acidification. Further investigation revealed that TLR9 targeted TRAF6 to activate the cGAS-STING pathway. Rescue experiments revealed that inactivation of the cGAS/STING signaling pathway could reverse the promoting effects of TLR9 upregulation on DCs maturation, activation, and autophagy-lysosome pathway. Overall, our findings suggested that TLR9 activated the autophagy-lysosome pathway to promote DCs maturation and activation by activating TRAF6-cGAS-STING pathway, thereby promoting SLE progression.
Collapse
Affiliation(s)
- Ying Liu
- Department of Rheumatology & Immunology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, People's Republic of China
| | - Fang-Zhi Wei
- Traditional Chinese Medicine Department, Boao Yiling Life Care Center, Qionghai, Hainan Province, People's Republic of China
| | - Yu-Wei Zhan
- Department of Rheumatology & Immunology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, People's Republic of China
| | - Ru Wang
- Experimental Center, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, People's Republic of China
| | - Bi-Yao Mo
- Department of Rheumatology & Immunology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, People's Republic of China
| | - Shu-Dian Lin
- Department of Rheumatology & Immunology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, People's Republic of China
| |
Collapse
|
17
|
Damasceno LEA, Cunha TM, Cunha FQ, Sparwasser T, Alves-Filho JC. A clinically-relevant STING agonist restrains human T H17 cell inflammatory profile. Int Immunopharmacol 2023; 124:111007. [PMID: 37778170 DOI: 10.1016/j.intimp.2023.111007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 09/13/2023] [Accepted: 09/25/2023] [Indexed: 10/03/2023]
Abstract
The STING signaling pathway has gained attention over the last few years due to its ability to incite antimicrobial and antitumoral immunity. Conversely, in mouse models of autoimmunity such as colitis and multiple sclerosis, where TH17 cells are implicated in tissue inflammation, STING activation has been associated with the attenuation of immunogenic responses. In this line, STING was found to limit murine TH17 pro-inflammatory program in vitro. Here we demonstrate that 2'3'-c-di-AM(PS)2(Rp,Rp), a STING agonist that has been undergoing clinical trials for antitumor immunotherapy, activates the STING signalosome in differentiating human TH17 cells. Of particular interest, 2'3'-c-di-AM(PS)2(Rp,Rp) reduces IL-17A production and IL23R expression by human TH17 cells while it favors the generation of regulatory T (Treg) cells. These findings suggest that STING agonists may be promising approaches for treating human TH17-mediated chronic inflammation.
Collapse
Affiliation(s)
- Luis Eduardo Alves Damasceno
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil; Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University, Mainz 55131, Germany
| | - Thiago Mattar Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil
| | - Fernando Queiroz Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil
| | - Tim Sparwasser
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University, Mainz 55131, Germany
| | - José Carlos Alves-Filho
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil; Center for Research in Inflammatory Diseases, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14049-900, Brazil.
| |
Collapse
|
18
|
Fan X, Song X, Chen W, Liang H, Nakatsukasa H, Zhang D. cGAS‐STING signaling in cancer: Regulation and therapeutic targeting. MEDCOMM – ONCOLOGY 2023; 2. [DOI: 10.1002/mog2.49] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 07/24/2023] [Indexed: 01/04/2025]
Abstract
AbstractImmunotherapy has revolutionized antitumor therapy. Since the discovery of stimulators of interferon genes (STING), efforts have been made to elucidate their mechanisms and physiological functions and explore the potential of STING as a therapeutic target in immune‐related diseases and malignant tumors. In recent years, STING agonists have become a popular research topic. Activation of the cyclic GMP–AMP synthase (cGAS)‐STING pathway produces large amounts of type I interferons, which play key roles in activating innate and acquired immune responses. The cGAS‐STING pathway influences almost all aspects of tumorigenesis and has great antitumor potential. In addition, the activation of the cGAS‐STING pathway is associated with tumor regression, prolonged survival of patients with cancer, and enhanced immunotherapy. Given the positive role of STING in antitumor immunity, the development of STING‐targeted drugs is important. In this review, we summarize the activation and potential mechanisms of the cGAS‐STING pathway, discuss the association of the cGAS‐STING pathway with tumors and autoimmune diseases, and highlight research progress, clinical applications, and combination drug strategies for STING agonists.
Collapse
Affiliation(s)
- Xinzou Fan
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital Sichuan University Chengdu Sichuan China
| | - Xiaoshuang Song
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital Sichuan University Chengdu Sichuan China
| | - Wenjing Chen
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital Sichuan University Chengdu Sichuan China
| | - Hantian Liang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital Sichuan University Chengdu Sichuan China
| | - Hiroko Nakatsukasa
- Laboratory of Microbiology and Immunology, Graduate School of Pharmaceutical Sciences Chiba University Chiba Japan
| | - Dunfang Zhang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital Sichuan University Chengdu Sichuan China
| |
Collapse
|
19
|
Jia X, Tan L, Chen S, Tang R, Chen W. Monogenic lupus: Tracing the therapeutic implications from single gene mutations. Clin Immunol 2023; 254:109699. [PMID: 37481012 DOI: 10.1016/j.clim.2023.109699] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/21/2023] [Accepted: 07/18/2023] [Indexed: 07/24/2023]
Abstract
Monogenic lupus, a distinctive variant of systemic lupus erythematosus (SLE), is characterized by early onset, family-centric clustering, and heightened disease severity. So far, over thirty genetic variations have been identified as single-gene etiology of SLE and lupus-like phenotypes. The critical role of these gene mutations in disrupting various immune pathways is increasingly recognized. In particular, single gene mutation-driven dysfunction within the innate immunity, notably deficiencies in the complement system, impedes the degradation of free nucleic acid and immune complexes, thereby promoting activation of innate immune cells. The accumulation of these components in various tissues and organs creates a pro-inflammatory microenvironment, characterized by a surge in pro-inflammatory cytokines, chemokines, reactive oxygen species, and type I interferons. Concurrently, single gene mutation-associated defects in the adaptive immune system give rise to the emergence of autoreactive T cells, hyperactivated B cells and plasma cells. The ensuing spectrum of cytokines and autoimmune antibodies drives systemic disease manifestations, primarily including kidney, skin and central nervous system-related phenotypes. This review provides a thorough overview of the single gene mutations and potential consequent immune dysregulations in monogenic lupus, elucidating the pathogenic mechanisms of monogenic lupus. Furthermore, it discusses the recent advances made in the therapeutic interventions for monogenic lupus.
Collapse
Affiliation(s)
- Xiuzhi Jia
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Li Tan
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Sixiu Chen
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Ruihan Tang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China.
| | - Wei Chen
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China.
| |
Collapse
|
20
|
Song C, Hu Z, Xu D, Bian H, Lv J, Zhu X, Zhang Q, Su L, Yin H, Lu T, Li Y. STING signaling in inflammaging: a new target against musculoskeletal diseases. Front Immunol 2023; 14:1227364. [PMID: 37492580 PMCID: PMC10363987 DOI: 10.3389/fimmu.2023.1227364] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 06/20/2023] [Indexed: 07/27/2023] Open
Abstract
Stimulator of Interferon Gene (STING) is a critical signaling linker protein that plays a crucial role in the intrinsic immune response, particularly in the cytoplasmic DNA-mediated immune response in both pathogens and hosts. It is also involved in various signaling processes in vivo. The musculoskeletal system provides humans with morphology, support, stability, and movement. However, its aging can result in various diseases and negatively impact people's lives. While many studies have reported that cellular aging is a leading cause of musculoskeletal disorders, it also offers insight into potential treatments. Under pathological conditions, senescent osteoblasts, chondrocytes, myeloid cells, and muscle fibers exhibit persistent senescence-associated secretory phenotype (SASP), metabolic disturbances, and cell cycle arrest, which are closely linked to abnormal STING activation. The accumulation of cytoplasmic DNA due to chromatin escape from the nucleus following DNA damage or telomere shortening activates the cGAS-STING signaling pathway. Moreover, STING activation is also linked to mitochondrial dysfunction, epigenetic modifications, and impaired cytoplasmic DNA degradation. STING activation upregulates SASP and autophagy directly and indirectly promotes cell cycle arrest. Thus, STING may be involved in the onset and development of various age-related musculoskeletal disorders and represents a potential therapeutic target. In recent years, many STING modulators have been developed and used in the study of musculoskeletal disorders. Therefore, this paper summarizes the effects of STING signaling on the musculoskeletal system at the molecular level and current understanding of the mechanisms of endogenous active ligand production and accumulation. We also discuss the relationship between some age-related musculoskeletal disorders and STING, as well as the current status of STING modulator development.
Collapse
Affiliation(s)
- Chenyu Song
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Zhuoyi Hu
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Dingjun Xu
- Department of Orthopaedics, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Zhejiang, China
| | - Huihui Bian
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Juan Lv
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Xuanxuan Zhu
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Qiang Zhang
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Li Su
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Heng Yin
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Tong Lu
- Department of Critical Care Medicine, Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, China
| | - Yinghua Li
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| |
Collapse
|
21
|
Floyd W, Pierpoint M, Su C, Patel R, Luo L, Deland K, Wisdom AJ, Zhu D, Ma Y, DeWitt SB, Williams NT, Lazarides AL, Somarelli JA, Corcoran DL, Eward WC, Cardona DM, Kirsch DG. Atrx deletion impairs CGAS/STING signaling and increases sarcoma response to radiation and oncolytic herpesvirus. J Clin Invest 2023; 133:e149310. [PMID: 37200088 PMCID: PMC10313374 DOI: 10.1172/jci149310] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/16/2023] [Indexed: 05/20/2023] Open
Abstract
ATRX is one of the most frequently altered genes in solid tumors, and mutation is especially frequent in soft tissue sarcomas. However, the role of ATRX in tumor development and response to cancer therapies remains poorly understood. Here, we developed a primary mouse model of soft tissue sarcoma and showed that Atrx-deleted tumors were more sensitive to radiation therapy and to oncolytic herpesvirus. In the absence of Atrx, irradiated sarcomas had increased persistent DNA damage, telomere dysfunction, and mitotic catastrophe. Our work also showed that Atrx deletion resulted in downregulation of the CGAS/STING signaling pathway at multiple points in the pathway and was not driven by mutations or transcriptional downregulation of the CGAS/STING pathway components. We found that both human and mouse models of Atrx-deleted sarcoma had a reduced adaptive immune response, markedly impaired CGAS/STING signaling, and increased sensitivity to TVEC, an oncolytic herpesvirus that is currently FDA approved for the treatment of aggressive melanomas. Translation of these results to patients with ATRX-mutant cancers could enable genomically guided cancer therapy approaches to improve patient outcomes.
Collapse
Affiliation(s)
- Warren Floyd
- Department of Pharmacology and Cancer Biology, and
| | | | - Chang Su
- Department of Pharmacology and Cancer Biology, and
| | - Rutulkumar Patel
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - Lixia Luo
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - Katherine Deland
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - Amy J. Wisdom
- Department of Radiation Oncology, Brigham and Women’s Hospital/Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Daniel Zhu
- Department of Pharmacology and Cancer Biology, and
| | - Yan Ma
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | | | - Nerissa T. Williams
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | | | - Jason A. Somarelli
- Department of Sarcoma, Moffitt Cancer Center, Tampa, Florida, USA
- Duke Cancer Institute, Durham, North Carolina, USA
| | - David L. Corcoran
- Center for Genomic and Computational Biology, Duke University, Durham, North Carolina, USA
| | | | - Diana M. Cardona
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | - David G. Kirsch
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Radiation Oncology and
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
22
|
Mohan C, Zhang T, Putterman C. Pathogenic cellular and molecular mediators in lupus nephritis. Nat Rev Nephrol 2023:10.1038/s41581-023-00722-z. [PMID: 37225921 DOI: 10.1038/s41581-023-00722-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2023] [Indexed: 05/26/2023]
Abstract
Kidney involvement in patients with systemic lupus erythematosus - lupus nephritis (LN) - is one of the most important and common clinical manifestations of this disease and occurs in 40-60% of patients. Current treatment regimens achieve a complete kidney response in only a minority of affected individuals, and 10-15% of patients with LN develop kidney failure, with its attendant morbidity and considerable prognostic implications. Moreover, the medications most often used to treat LN - corticosteroids in combination with immunosuppressive or cytotoxic drugs - are associated with substantial side effects. Advances in proteomics, flow cytometry and RNA sequencing have led to important new insights into immune cells, molecules and mechanistic pathways that are instrumental in the pathogenesis of LN. These insights, together with a renewed focus on the study of human LN kidney tissue, suggest new therapeutic targets that are already being tested in lupus animal models and early-phase clinical trials and, as such, are hoped to eventually lead to meaningful improvements in the care of patients with systemic lupus erythematosus-associated kidney disease.
Collapse
Affiliation(s)
- Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, TX, USA.
| | - Ting Zhang
- Division of Rheumatology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chaim Putterman
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel.
- Division of Rheumatology and Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
23
|
Zhang S, Zheng R, Pan Y, Sun H. Potential Therapeutic Value of the STING Inhibitors. Molecules 2023; 28:3127. [PMID: 37049889 PMCID: PMC10096477 DOI: 10.3390/molecules28073127] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/22/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
The stimulator of interferon genes (STING) is a critical protein in the activation of the immune system in response to DNA. It can participate the inflammatory response process by modulating the inflammation-preferred translation program through the STING-PKR-like endoplasmic reticulum kinase (PERK)-eIF2α pathway or by inducing the secretion of type I interferons (IFNs) and a variety of proinflammatory factors through the recruitment of TANK-binding kinase 1 (TBK1) and interferon regulatory factor 3 (IRF3) or the regulation of the nuclear factor kappa-B (NF-κB) pathway. Based on the structure, location, function, genotype, and regulatory mechanism of STING, this review summarizes the potential value of STING inhibitors in the prevention and treatment of infectious diseases, psoriasis, systemic lupus erythematosus, non-alcoholic fatty liver disease, and other inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Shangran Zhang
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Runan Zheng
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Yanhong Pan
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacy, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210009, China
| | - Hongbin Sun
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Chongqing Innovation Institute of China Pharmaceutical University, Chongqing 401135, China
| |
Collapse
|
24
|
Kalantari P, Shecter I, Hopkins J, Pilotta Gois A, Morales Y, Harandi BF, Sharma S, Stadecker MJ. The balance between gasdermin D and STING signaling shapes the severity of schistosome immunopathology. Proc Natl Acad Sci U S A 2023; 120:e2211047120. [PMID: 36943884 PMCID: PMC10068786 DOI: 10.1073/pnas.2211047120] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 02/03/2023] [Indexed: 03/23/2023] Open
Abstract
There is significant disease heterogeneity among mouse strains infected with the helminth Schistosoma mansoni. Here, we uncover a unique balance in two critical innate pathways governing the severity of disease. In the low-pathology setting, parasite egg-stimulated dendritic cells (DCs) induce robust interferon (IFN)β production, which is dependent on the cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING) cytosolic DNA sensing pathway and results in a Th2 response with suppression of proinflammatory cytokine production and Th17 cell activation. IFNβ induces signal transducer and activator of transcription (STAT)1, which suppresses CD209a, a C-type lectin receptor associated with severe disease. In contrast, in the high-pathology setting, enhanced DC expression of the pore-forming protein gasdermin D (Gsdmd) results in reduced expression of cGAS/STING, impaired IFNβ, and enhanced pyroptosis. Our findings demonstrate that cGAS/STING signaling represents a unique mechanism inducing protective type I IFN, which is counteracted by Gsdmd.
Collapse
Affiliation(s)
- Parisa Kalantari
- Department of Immunology, Tufts University School of Medicine, Boston, MA02111
- Department of Veterinary and Biomedical Sciences, Center for Molecular Immunology and Infectious Disease, The Pennsylvania State University, University Park, PA16802
| | - Ilana Shecter
- Department of Immunology, Tufts University School of Medicine, Boston, MA02111
| | - Jacob Hopkins
- Department of Immunology, Tufts University School of Medicine, Boston, MA02111
| | - Andrea Pilotta Gois
- Department of Immunology, Tufts University School of Medicine, Boston, MA02111
| | - Yoelkys Morales
- Department of Immunology, Tufts University School of Medicine, Boston, MA02111
| | - Bijan F. Harandi
- Department of Immunology, Tufts University School of Medicine, Boston, MA02111
| | - Shruti Sharma
- Department of Immunology, Tufts University School of Medicine, Boston, MA02111
| | - Miguel J. Stadecker
- Department of Immunology, Tufts University School of Medicine, Boston, MA02111
| |
Collapse
|
25
|
Harsini S, Rezaei N. Autoimmune diseases. Clin Immunol 2023. [DOI: 10.1016/b978-0-12-818006-8.00001-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
26
|
Kumpunya S, Thim-uam A, Thumarat C, Leelahavanichkul A, Kalpongnukul N, Chantaravisoot N, Pisitkun T, Pisitkun P. cGAS deficiency enhances inflammasome activation in macrophages and inflammatory pathology in pristane-induced lupus. Front Immunol 2022; 13:1010764. [PMID: 36591278 PMCID: PMC9800982 DOI: 10.3389/fimmu.2022.1010764] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
Introduction Type I interferon (IFN) plays a vital role in the pathogenesis of systemic lupus erythematosus. Cyclic GMP AMP synthase (cGAS) is a cytosolic DNA sensor that recognizes dsDNA and creates cGAMP to activate STING-mediated type I IFN production. The activation of STING induces lupus disease in Fcgr2b deficient mice through the differentiation of dendritic cells. In contrast, Cgas-deficient mice could be generated more autoantibody production and proteinuria in pristane-induced lupus (PIL). These data suggested that the other dsDNA sensors could be involved in lupus development mechanisms. Methods This study aimed to identify the cGAS-mediated mechanisms contributing to lupus pathogenesis in PIL. The Cgas-deficient and WT mice were induced lupus disease with pristane and subsequently analyzed autoantibody, histopathology, and immunophenotypes. The lung tissues were analyzed with the expression profiles by RT-PCR and western blot. The bone marrow-derived macrophages were stimulated with inflammasome activators and observed pyroptosis. Results The Cgas-/- mice developed more severe pulmonary hemorrhage and autoantibody production than WT mice. The activated dendritic cells, IFN-g-, and IL-17a-producing T helper cells, and infiltrated macrophages in the lung were detected in Cgas-/- mice higher than in WT mice. We observed an increase in expression of Aim2, Casp11, and Ifi16 in the lung and serum IL-1a but IL-1b in pristane-injected Cgas-/- mice. The rise of Caspase-11 in the lung of pristane-injected Cgas-/- mice suggested noncanonical inflammasome activation. The activation of AIM2 and NLRP3 inflammasomes in bone marrow-derived macrophages (BMDMs) enhanced the number of dead cells in Cgas-/- mice compared with WT mice. Activation of the inflammasome significantly induced pyroptosis in Cgas-/- BMDMs. The dsDNA level, but not mitochondrial DNA, increased dramatically in pristane-injected Cgas-/- mice suggesting the dsDNA could be a ligand activating inflammasomes. The cGAS agonist-induced BMDM activation in the Cgas-/- mice indicated that the activation of DNA sensors other than cGAS enhanced activated macrophages. Conclusion These findings suggested that cGAS hampers the unusual noncanonical inflammasome activation through other DNA sensors.
Collapse
Affiliation(s)
- Sarinya Kumpunya
- Interdisciplinary Program of Biomedical Sciences, Graduate School, Chulalongkorn University, Bangkok, Thailand
| | - Arthid Thim-uam
- Division of Biochemistry, School of Medical Sciences, University of Phayao, Phayao, Thailand
| | - Chisanu Thumarat
- Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Asada Leelahavanichkul
- Center of Excellence in Translational Research in Inflammation and Immunology (CETRII), Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Nuttiya Kalpongnukul
- Interdisciplinary Program of Biomedical Sciences, Graduate School, Chulalongkorn University, Bangkok, Thailand,Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Naphat Chantaravisoot
- Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand,Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Trairak Pisitkun
- Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand,Epithelial Systems Biology Laboratory, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States,*Correspondence: Prapaporn Pisitkun, ; Trairak Pisitkun,
| | - Prapaporn Pisitkun
- Program in 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,*Correspondence: Prapaporn Pisitkun, ; Trairak Pisitkun,
| |
Collapse
|
27
|
The cGAS-STING pathway and cancer. NATURE CANCER 2022; 3:1452-1463. [PMID: 36510011 DOI: 10.1038/s43018-022-00468-w] [Citation(s) in RCA: 255] [Impact Index Per Article: 85.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 10/17/2022] [Indexed: 12/14/2022]
Abstract
The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway has emerged as a critical innate immune pathway that, following engagement by DNA, promotes distinct immune effector responses that can impact virtually all aspects of tumorigenesis, from malignant cell transformation to metastasis. Here we address how natural tumor-associated processes and traditional cancer therapies are shaped by cGAS-STING signaling, and how this contributes to beneficial or detrimental outcomes of cancer. We consider current efforts to target the cGAS-STING axis in tumors and highlight new frontiers in cGAS-STING biology to inspire thinking about their connection to cancer.
Collapse
|
28
|
Zhuang H, Hudson E, Han S, Arja RD, Hui W, Lu L, Reeves WH. Microvascular lung injury and endoplasmic reticulum stress in systemic lupus erythematosus-associated alveolar hemorrhage and pulmonary vasculitis. Am J Physiol Lung Cell Mol Physiol 2022; 323:L715-L729. [PMID: 36255715 PMCID: PMC9744657 DOI: 10.1152/ajplung.00051.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 10/13/2022] [Accepted: 10/13/2022] [Indexed: 12/14/2022] Open
Abstract
Human COPA mutations affecting retrograde Golgi-to-endoplasmic reticulum (ER) protein transport cause diffuse alveolar hemorrhage (DAH) and ER stress ("COPA syndrome"). Patients with SLE also can develop DAH. C57BL/6 (B6) mice with pristane-induced lupus develop monocyte-dependent DAH indistinguishable from human DAH, whereas BALB/c mice are resistant. We examined Copa and ER stress in pristane-induced lupus. Copa expression, ER stress, vascular injury, and apoptosis were assessed in mice and COPA was quantified in blood from patients with SLE. Copa mRNA and protein expression were impaired in B6 mice with pristane-induced DAH, but not in pristane-treated BALB/c mice. An ER stress response (increased Hsp5a/BiP, Ddit3/CHOP, Eif2a, and spliced Xbp1) was seen in lungs from pristane-treated B6, but not BALB/c, mice. Resistance of BALB/c mice to DAH was overcome by treating them with low-dose thapsigargin plus pristane. CB6F1 mice did not develop DAH or ER stress, suggesting that susceptibility was recessive. Increased pulmonary expression of von Willebrand factor (Vwf), a marker of endothelial injury, and the chemokine Ccl2 in DAH suggested that pristane promotes lung microvascular injury and monocyte recruitment. Consistent with that possibility, lung endothelial cells and infiltrating bone marrow-derived cells from pristane-treated B6 mice expressed BiP and showed evidence of apoptosis (annexin-V and activated caspase-3 staining). COPA expression also was low in patients with SLE with lung involvement. Pristane-induced DAH may be initiated by endothelial injury, resulting in ER stress, apoptosis of lung endothelial cells, and recruitment of myeloid cells that propagate lung injury. The pathogenesis of DAH in SLE and COPA syndrome may overlap.
Collapse
Affiliation(s)
- Haoyang Zhuang
- Division of Rheumatology, Allergy, & Clinical Immunology, University of Florida, Gainesville, Florida
| | - Erin Hudson
- Division of Rheumatology, Allergy, & Clinical Immunology, University of Florida, Gainesville, Florida
| | - Shuhong Han
- Division of Rheumatology, Allergy, & Clinical Immunology, University of Florida, Gainesville, Florida
| | - Rawad Daniel Arja
- Division of Rheumatology, Allergy, & Clinical Immunology, University of Florida, Gainesville, Florida
| | - Winnie Hui
- Division of Rheumatology, Allergy, & Clinical Immunology, University of Florida, Gainesville, Florida
| | - Li Lu
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, Florida
| | - Westley H Reeves
- Division of Rheumatology, Allergy, & Clinical Immunology, University of Florida, Gainesville, Florida
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, Florida
| |
Collapse
|
29
|
Vila IK, Guha S, Kalucka J, Olagnier D, Laguette N. Alternative pathways driven by STING: From innate immunity to lipid metabolism. Cytokine Growth Factor Rev 2022; 68:54-68. [PMID: 36085258 DOI: 10.1016/j.cytogfr.2022.08.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 08/29/2022] [Indexed: 01/30/2023]
Abstract
The Stimulator of Interferon Genes (STING) is a major adaptor protein that is central to the initiation of type I interferon responses and proinflammatory signalling. STING-dependent signalling is triggered by the presence of cytosolic nucleic acids that are generated following pathogen infection or cellular stress. Beyond this central role in controlling immune responses through the production of cytokines and chemokines, recent reports have uncovered inflammation-independent STING functions. Amongst these, a rapidly growing body of evidence demonstrates a key role of STING in controlling metabolic pathways at several levels. Since immunity and metabolic homeostasis are tightly interconnected, these findings deepen our understanding of the involvement of STING in human pathologies. Here, we discuss these findings and reflect on their impact on our current understanding of how nucleic acid immunity controls homeostasis and promotes pathological outcomes.
Collapse
Affiliation(s)
- Isabelle K Vila
- Institut de Génétique Humaine, Univ Montpellier, CNRS, Montpellier, France.
| | - Soumyabrata Guha
- Institut de Génétique Humaine, Univ Montpellier, CNRS, Montpellier, France
| | - Joanna Kalucka
- Aarhus University, Department of Biomedicine, Aarhus, Denmark
| | - David Olagnier
- Aarhus University, Department of Biomedicine, Aarhus, Denmark
| | - Nadine Laguette
- Institut de Génétique Humaine, Univ Montpellier, CNRS, Montpellier, France.
| |
Collapse
|
30
|
Mitrofanova A, Fontanella A, Tolerico M, Mallela S, Molina David J, Zuo Y, Boulina M, Kim JJ, Santos J, Ge M, Sloan A, Issa W, Gurumani M, Pressly J, Ito M, Kretzler M, Eddy S, Nelson R, Merscher S, Burke G, Fornoni A. Activation of Stimulator of IFN Genes (STING) Causes Proteinuria and Contributes to Glomerular Diseases. J Am Soc Nephrol 2022; 33:2153-2173. [PMID: 36198430 PMCID: PMC9731637 DOI: 10.1681/asn.2021101286] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 09/06/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The signaling molecule stimulator of IFN genes (STING) was identified as a crucial regulator of the DNA-sensing cyclic GMP-AMP synthase (cGAS)-STING pathway, and this signaling pathway regulates inflammation and energy homeostasis under conditions of obesity, kidney fibrosis, and AKI. However, the role of STING in causing CKD, including diabetic kidney disease (DKD) and Alport syndrome, is unknown. METHODS To investigate whether STING activation contributes to the development and progression of glomerular diseases such as DKD and Alport syndrome, immortalized human and murine podocytes were differentiated for 14 days and treated with a STING-specific agonist. We used diabetic db/db mice, mice with experimental Alport syndrome, C57BL/6 mice, and STING knockout mice to assess the role of the STING signaling pathway in kidney failure. RESULTS In vitro, murine and human podocytes express all of the components of the cGAS-STING pathway. In vivo, activation of STING renders C57BL/6 mice susceptible to albuminuria and podocyte loss. STING is activated at baseline in mice with experimental DKD and Alport syndrome. STING activation occurs in the glomerular but not the tubulointerstitial compartment in association with autophagic podocyte death in Alport syndrome mice and with apoptotic podocyte death in DKD mouse models. Genetic or pharmacologic inhibition of STING protects from progression of kidney disease in mice with DKD and Alport syndrome and increases lifespan in Alport syndrome mice. CONCLUSION The activation of the STING pathway acts as a mediator of disease progression in DKD and Alport syndrome. Targeting STING may offer a therapeutic option to treat glomerular diseases of metabolic and nonmetabolic origin or prevent their development, progression, or both.
Collapse
Affiliation(s)
- Alla Mitrofanova
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, Florida
- Department of Surgery, University of Miami, Miller School of Medicine, Miami, Florida
| | - Antonio Fontanella
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, Florida
| | - Matthew Tolerico
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, Florida
| | - Shamroop Mallela
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, Florida
| | - Judith Molina David
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, Florida
| | - Yiqin Zuo
- Department of Pathology, University of Miami Medical Group, Miller School of Medicine, Miami, Florida
| | - Marcia Boulina
- Diabetes Research Institute, University of Miami, Miller School of Medicine, Miami, Florida
| | - Jin-Ju Kim
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, Florida
| | - Javier Santos
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, Florida
| | - Mengyuan Ge
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, Florida
| | - Alexis Sloan
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, Florida
| | - Wadih Issa
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, Florida
| | - Margaret Gurumani
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, Florida
| | - Jeffrey Pressly
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, Florida
| | - Marie Ito
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, Florida
| | - Matthias Kretzler
- Division of Nephrology, Departments of Internal Medicine and Computational Medicine and Bioinformatics, University of Michigan School of Medicine, Ann Arbor, Michigan
| | - Sean Eddy
- Division of Nephrology, Departments of Internal Medicine and Computational Medicine and Bioinformatics, University of Michigan School of Medicine, Ann Arbor, Michigan
| | - Robert Nelson
- Chronic Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, Arizona
| | - Sandra Merscher
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, Florida
| | - George Burke
- Department of Surgery, University of Miami, Miller School of Medicine, Miami, Florida
- Diabetes Research Institute, University of Miami, Miller School of Medicine, Miami, Florida
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, Florida
| |
Collapse
|
31
|
Shen A, Chen M, Chen Q, Liu Z, Zhang A. Recent advances in the development of STING inhibitors: an updated patent review. Expert Opin Ther Pat 2022; 32:1131-1143. [PMID: 36332188 DOI: 10.1080/13543776.2022.2144220] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
INTRODUCTION STING is at the center of the cGAS-STING signaling and acts as the hub of the innate immune system. Hyper-activation of STING has been observed in various severe autoimmune diseases, such as AGS, SLE, and many other diseases including neurological and metabolic disorders. Therefore, STING has been considered as a promising target. In recent years, several STING inhibitors have been claimed in patents. AREAS COVERED Small-molecule STING inhibitors reported in patents (disclosed before May 2022 through the public database at https://worldwide.espacenet.com) were summarized in this review and the available structure-activity relationships (SARs) and molecular mechanisms of action were presented. EXPERT OPINION Compared with STING agonists, the development of STING inhibitors is still in its infancy and no candidates have entered clinical investigation stage. Fortunately, patent applications are appearing at an increasing rate and a few of them have been validated in vivo, thus providing valuable insights for further structural optimization. More efforts are urgently needed since it is not clear yet that inhibitors targeting STING can solely exert sufficient therapeutic effects on autoimmune diseases, and the toxicity profile of such inhibitors is unknown as well. Therefore, it is extremely important to identify a selective and efficacious STING inhibitor for clinical evaluation to provide proof-of-concept for this approach.
Collapse
Affiliation(s)
- Ancheng Shen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Pharm-X Center, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Mingjie Chen
- Pharm-X Center, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Qingxuan Chen
- Pharm-X Center, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Zhiguo Liu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ao Zhang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Pharm-X Center, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
32
|
Zhang M, Zou Y, Zhou X, Zhou J. Inhibitory targeting cGAS-STING-TBK1 axis: Emerging strategies for autoimmune diseases therapy. Front Immunol 2022; 13:954129. [PMID: 36172373 PMCID: PMC9511411 DOI: 10.3389/fimmu.2022.954129] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
The cGAS-STING signaling plays an integral role in the host immune response, and the abnormal activation of cGAS-STING is highly related to various autoimmune diseases. Therefore, targeting the cGAS-STING-TBK1 axis has become a promising strategy in therapy of autoimmune diseases. Herein, we summarized the key pathways mediated by the cGAS-STING-TBK1 axis and various cGAS-STING-TBK1 related autoimmune diseases, as well as the recent development of cGAS, STING, or TBK1 selective inhibitors and their potential application in therapy of cGAS-STING-TBK1 related autoimmune diseases. Overall, the review highlights that inhibiting cGAS-STING-TBK1 signaling is an attractive strategy for autoimmune disease therapy.
Collapse
Affiliation(s)
- Min Zhang
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Department of Chemistry, Zhejiang Normal University, Jinhua, China
- Drug development and innovation center, College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Yan Zou
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Department of Chemistry, Zhejiang Normal University, Jinhua, China
- Drug development and innovation center, College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Xujun Zhou
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Department of Chemistry, Zhejiang Normal University, Jinhua, China
- Drug development and innovation center, College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Jinming Zhou
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Department of Chemistry, Zhejiang Normal University, Jinhua, China
- Drug development and innovation center, College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, China
- *Correspondence: Jinming Zhou,
| |
Collapse
|
33
|
Skopelja-Gardner S, An J, Elkon KB. Role of the cGAS-STING pathway in systemic and organ-specific diseases. Nat Rev Nephrol 2022; 18:558-572. [PMID: 35732833 PMCID: PMC9214686 DOI: 10.1038/s41581-022-00589-6] [Citation(s) in RCA: 155] [Impact Index Per Article: 51.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/18/2022] [Indexed: 12/21/2022]
Abstract
Cells are equipped with numerous sensors that recognize nucleic acids, which probably evolved for defence against viruses. Once triggered, these sensors stimulate the production of type I interferons and other cytokines that activate immune cells and promote an antiviral state. The evolutionary conserved enzyme cyclic GMP-AMP synthase (cGAS) is one of the most recently identified DNA sensors. Upon ligand engagement, cGAS dimerizes and synthesizes the dinucleotide second messenger 2',3'-cyclic GMP-AMP (cGAMP), which binds to the endoplasmic reticulum protein stimulator of interferon genes (STING) with high affinity, thereby unleashing an inflammatory response. cGAS-binding DNA is not restricted by sequence and must only be >45 nucleotides in length; therefore, cGAS can also be stimulated by self genomic or mitochondrial DNA. This broad specificity probably explains why the cGAS-STING pathway has been implicated in a number of autoinflammatory, autoimmune and neurodegenerative diseases; this pathway might also be activated during acute and chronic kidney injury. Therapeutic manipulation of the cGAS-STING pathway, using synthetic cyclic dinucleotides or inhibitors of cGAMP metabolism, promises to enhance immune responses in cancer or viral infections. By contrast, inhibitors of cGAS or STING might be useful in diseases in which this pro-inflammatory pathway is chronically activated.
Collapse
Affiliation(s)
| | - Jie An
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Keith B Elkon
- Department of Medicine, University of Washington, Seattle, WA, USA.
- Department of Immunology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
34
|
Zhao J, Xiao R, Zeng R, He E, Zhang A. Small molecules targeting cGAS-STING pathway for autoimmune disease. Eur J Med Chem 2022; 238:114480. [DOI: 10.1016/j.ejmech.2022.114480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/14/2022] [Accepted: 05/17/2022] [Indexed: 11/26/2022]
|
35
|
Zou YR, Davidson A. Nucleic Acid Sensing and Systemic Lupus Erythematosus: The Danger of Self. THE JOURNAL OF IMMUNOLOGY 2022; 209:431-433. [DOI: 10.4049/jimmunol.2200129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Abstract
This Pillars of Immunology article is a commentary on “Chromatin–IgG complexes activate B cells by dual engagement of IgM and Toll-like receptors,” a pivotal article written by E. A. Leadbetter, I. R. Rifkin, A. M. Hohlbaum, B. C. Beaudette, M. J. Shlomchik and A. Marshak-Rothstein, and published in Nature in 2002. https://www.nature.com/articles/416603a.
Collapse
Affiliation(s)
- Yong-Rui Zou
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, NY
| | - Anne Davidson
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, NY
| |
Collapse
|
36
|
Hagan RS, Gomez JC, Torres-Castillo J, Martin JR, Doerschuk CM. TBK1 Is Required for Host Defense Functions Distinct from Type I IFN Expression and Myeloid Cell Recruitment in Murine Streptococcus pneumoniae Pneumonia. Am J Respir Cell Mol Biol 2022; 66:671-681. [PMID: 35358404 PMCID: PMC9163639 DOI: 10.1165/rcmb.2020-0311oc] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 01/19/2022] [Indexed: 11/24/2022] Open
Abstract
Bacterial pneumonia induces the rapid recruitment and activation of neutrophils and macrophages into the lung, and these cells contribute to bacterial clearance and other defense functions. TBK1 (TANK-binding kinase 1) performs many functions, including activation of the type I IFN pathway and regulation of autophagy and mitophagy, but its contribution to antibacterial defenses in the lung is unclear. We previously showed that lung neutrophils upregulate mRNAs for TBK1 and its accessory proteins during Streptococcus pneumoniae pneumonia, despite low or absent expression of type I IFN in these cells. We hypothesized that TBK1 performs key antibacterial functions in pneumonia apart from type I IFN expression. Using TBK1 null mice, we show that TBK1 contributes to antibacterial defenses and promotes bacterial clearance and survival. TBK1 null mice express lower concentrations of many cytokines in the infected lung. Conditional deletion of TBK1 with LysMCre results in TBK1 deletion from macrophages but not neutrophils. LysMCre TBK1 mice have no defect in cytokine expression, implicating a nonmacrophage cell type as a key TBK1-dependent cell. TBK1 null neutrophils have no defect in recruitment to the infected lung but show impaired activation of p65/NF-κB and STAT1 and lower expression of reactive oxygen species, IFNγ, and IL12p40. TLR1/2 and 4 agonists each induce phosphorylation of TBK1 in neutrophils. Surprisingly, neutrophil TBK1 activation in vivo does not require the adaptor STING. Thus, TBK1 is a critical component of STING-independent antibacterial responses in the lung, and TBK1 is necessary for multiple neutrophil functions.
Collapse
Affiliation(s)
- Robert S. Hagan
- Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine
- Marsico Lung Institute, and
| | - John C. Gomez
- Marsico Lung Institute, and
- Center for Airways Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jose Torres-Castillo
- Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine
- Marsico Lung Institute, and
| | - Jessica R. Martin
- Marsico Lung Institute, and
- Center for Airways Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Claire M. Doerschuk
- Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine
- Marsico Lung Institute, and
- Center for Airways Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
37
|
Kong X, Zuo H, Huang HD, Zhang Q, Chen J, He C, Hu Y. STING as an emerging therapeutic target for drug discovery: Perspectives from the global patent landscape. J Adv Res 2022; 44:119-133. [PMID: 35636721 PMCID: PMC9936525 DOI: 10.1016/j.jare.2022.05.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 04/15/2022] [Accepted: 05/15/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND The STimulator of INterferon Genes (STING) plays an essential role in the innate immune system by inducing the expression of type I interferons (IFNs) and inflammatory cytokines upon sensing cytosolic DNA. Although modulating STING has shown promise as a potential treatment for cancers and inflammatory and autoimmune diseases in substantial pre-clinical studies, current preliminary clinical results of STING agonists have demonstrated limited anti-tumor efficacy. Currently, there is ongoing R&D targeting STING and focusing on the delivery of next-generation therapeutics. Whereas no comprehensive analysis on the STING patent landscape has been conducted to fill the gap between basic research progress and drug development and commercialization. AIM OF REVIEW This study summarized the current agents in the clinical stage and global patenting profiles to help identify the current status, development trends, and emerging technologies of the nascent field of STING modulation. KEY SCIENTIFIC CONCEPTS OF REVIEW Rapidly increasing R&D efforts and outcomes targeting STING were indicated by the recently increasing number and pharmacologic classes of drug candidates in clinic as well as in emergent technological patenting activities. Despite the overall fragmental ownership of patents, several pioneers that have advanced the clinical evaluation of novel STING agonists have established the basis of STING-relevant inventions through their influential patents in the field. These patents also facilitated progress on novel STING modulators, relevant delivery systems, pharmaceutical compositions, and combination strategies with the potential for further enhancing therapeutic outcomes by targeting STING.
Collapse
Affiliation(s)
- Xiangjun Kong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China,Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Macao 999078, China
| | - Huali Zuo
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong-Shenzhen, Shenzhen 518172, China,School of Life and Health Sciences, The Chinese University of Hong Kong-Shenzhen, Shenzhen 518172, China,School of Computer Science and Technology, University of Science and Technology of China, Hefei 230027, China
| | - Hsien-Da Huang
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong-Shenzhen, Shenzhen 518172, China,School of Life and Health Sciences, The Chinese University of Hong Kong-Shenzhen, Shenzhen 518172, China
| | - Qianru Zhang
- School of Pharmacy, Key Lab of the Basic Pharmacology of the Ministry of Education, Zunyi Medical University, Guizhou 563000, China
| | - Jiayu Chen
- Department of Biochemistry and Molecular Biology, Zunyi Medical University, Guizhou 563000, China
| | - Chengwei He
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Yuanjia Hu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China; Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Macao 999078, China.
| |
Collapse
|
38
|
Wobma H, Shin DS, Chou J, Dedeoğlu F. Dysregulation of the cGAS-STING Pathway in Monogenic Autoinflammation and Lupus. Front Immunol 2022; 13:905109. [PMID: 35693769 PMCID: PMC9186411 DOI: 10.3389/fimmu.2022.905109] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 05/02/2022] [Indexed: 01/19/2023] Open
Abstract
One of the oldest mechanisms of immune defense against pathogens is through detection of foreign DNA. Since human DNA is compartmentalized into the nucleus, its presence in the cytosol heralds a potential threat. The cGAS-STING pathway is one of the most important cytosolic DNA sensing pathways and leads to interferon signaling, inflammasome activation, autophagy, and cell death. While STING signaling is protective at physiologic levels, chronic activation of this pathway can instead drive autoinflammation and autoimmunity. Here we discuss several monogenic disorders of the STING pathway that highlight its impact on both innate and adaptive immunity in the progressive loss of tolerance. The potential relevance of STING signaling in systemic lupus erythematosus is then discussed with a focus on future avenues for monitoring and targeting this pathway.
Collapse
|
39
|
Fritsch LE, Ju J, Gudenschwager Basso EK, Soliman E, Paul S, Chen J, Kaloss AM, Kowalski EA, Tuhy TC, Somaiya RD, Wang X, Allen IC, Theus MH, Pickrell AM. Type I Interferon Response Is Mediated by NLRX1-cGAS-STING Signaling in Brain Injury. Front Mol Neurosci 2022; 15:852243. [PMID: 35283725 PMCID: PMC8916033 DOI: 10.3389/fnmol.2022.852243] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 02/02/2022] [Indexed: 01/05/2023] Open
Abstract
Background Inflammation is a significant contributor to neuronal death and dysfunction following traumatic brain injury (TBI). Recent evidence suggests that interferons may be a key regulator of this response. Our studies evaluated the role of the Cyclic GMP-AMP Synthase-Stimulator of Interferon Genes (cGAS-STING) signaling pathway in a murine model of TBI. Methods Male, 8-week old wildtype, STING knockout (-/-), cGAS -/-, and NLRX1 -/- mice were subjected to controlled cortical impact (CCI) or sham injury. Histopathological evaluation of tissue damage was assessed using non-biased stereology, which was complemented by analysis at the mRNA and protein level using qPCR and western blot analysis, respectively. Results We found that STING and Type I interferon-stimulated genes were upregulated after CCI injury in a bi-phasic manner and that loss of cGAS or STING conferred neuroprotection concomitant with a blunted inflammatory response at 24 h post-injury. cGAS -/- animals showed reduced motor deficits 4 days after injury (dpi), and amelioration of tissue damage was seen in both groups of mice up to 14 dpi. Given that cGAS requires a cytosolic damage- or pathogen-associated molecular pattern (DAMP/PAMP) to prompt downstream STING signaling, we further demonstrate that mitochondrial DNA is present in the cytosol after TBI as one possible trigger for this pathway. Recent reports suggest that the immune modulator NLR containing X1 (NLRX1) may sequester STING during viral infection. Our findings show that NLRX1 may be an additional regulator that functions upstream to regulate the cGAS-STING pathway in the brain. Conclusions These findings suggest that the canonical cGAS-STING-mediated Type I interferon signaling axis is a critical component of neural tissue damage following TBI and that mtDNA may be a possible trigger in this response.
Collapse
Affiliation(s)
- Lauren E. Fritsch
- Translational Biology, Medicine, and Health Graduate Program, Virginia Polytechnic Institute and State University, Roanoke, VA, United States
| | - Jing Ju
- Molecular and Cellular Biology Graduate Program, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | | | - Eman Soliman
- Department of Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Swagatika Paul
- Biomedical and Veterinary Sciences Graduate Program, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Jiang Chen
- Molecular and Cellular Biology Graduate Program, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Alexandra M. Kaloss
- Department of Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Elizabeth A. Kowalski
- Department of Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Taylor C. Tuhy
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Rachana Deven Somaiya
- Translational Biology, Medicine, and Health Graduate Program, Virginia Polytechnic Institute and State University, Roanoke, VA, United States
| | - Xia Wang
- Department of Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Irving Coy Allen
- Department of Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Michelle H. Theus
- Department of Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Alicia M. Pickrell
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| |
Collapse
|
40
|
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
|
41
|
Al-Hakeim HK, Hadi HH, Jawad GA, Maes M. Intersections between Copper, β-Arrestin-1, Calcium, FBXW7, CD17, Insulin Resistance and Atherogenicity Mediate Depression and Anxiety Due to Type 2 Diabetes Mellitus: A Nomothetic Network Approach. J Pers Med 2022; 12:jpm12010023. [PMID: 35055338 PMCID: PMC8779500 DOI: 10.3390/jpm12010023] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/14/2021] [Accepted: 12/15/2021] [Indexed: 02/06/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is frequently accompanied by affective disorders with a prevalence of comorbid depression of around 25%. Nevertheless, the biomarkers of affective symptoms including depression and anxiety due to T2DM are not well established. The present study delineated the effects of serum levels of copper, zinc, β-arrestin-1, FBXW7, lactosylceramide (LacCer), serotonin, calcium, magnesium on severity of depression and anxiety in 58 men with T2DM and 30 healthy male controls beyond the effects of insulin resistance (IR) and atherogenicity. Severity of affective symptoms was assessed using the Hamilton Depression and Anxiety rating scales. We found that 61.7% of the variance in affective symptoms was explained by the multivariate regression on copper, β-arrestin-1, calcium, and IR coupled with atherogenicity. Copper and LacCer (positive) and calcium and BXW7 (inverse) had significant specific indirect effects on affective symptoms, which were mediated by IR and atherogenicity. Copper, β-arrestin-1, and calcium were associated with affective symptoms above and beyond the effects of IR and atherogenicity. T2DM and affective symptoms share common pathways, namely increased atherogenicity, IR, copper, and β-arrestin-1, and lowered calcium, whereas copper, β-arrestin-1, calcium, LacCer, and FBXW7 may modulate depression and anxiety symptoms by affecting T2DM.
Collapse
Affiliation(s)
- Hussein Kadhem Al-Hakeim
- Department of Chemistry, College of Science, University of Kufa, Najaf 54001, Iraq; (H.K.A.-H.); (H.H.H.); (G.A.J.)
| | - Hadi Hasan Hadi
- Department of Chemistry, College of Science, University of Kufa, Najaf 54001, Iraq; (H.K.A.-H.); (H.H.H.); (G.A.J.)
| | - Ghoufran Akeel Jawad
- Department of Chemistry, College of Science, University of Kufa, Najaf 54001, Iraq; (H.K.A.-H.); (H.H.H.); (G.A.J.)
| | - Michael Maes
- Department of Psychiatry, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, P.O. Box 281, Geelong, VIC 3220, Australia
- Correspondence:
| |
Collapse
|
42
|
Liu X, Ding Y, Zheng X, Huang H, Shi L, Yang X, Wei J, Li Y, Kao W, Zhang F, Qian J. Small RNAs encoded by human endogenous retrovirus K overexpressed in PBMCs may contribute to the diagnosis and evaluation of systemic lupus erythematosus as novel biomarkers. Hum Mol Genet 2021; 31:1407-1416. [PMID: 34761271 DOI: 10.1093/hmg/ddab327] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/09/2021] [Accepted: 11/03/2021] [Indexed: 11/13/2022] Open
Abstract
This study aimed to identify the genes and small RNAs (sRNAs) expressed by the human endogenous retrovirus K (HERV-K) HML2 and their associations with the immune process of systemic lupus erythematosus (SLE). RNA-Seq data including 99 SLE patients and 18 controls (GSE72420) was obtained from the Gene Expression Omnibus. Differentially expressed genes (DEGs) as well as HML2-DEGs between SLE patients and normal controls were identified. Five HML2-DEGs involved in immune-regulating function were identified using weighted gene co-expression network analysis (WGCNA). The associations between these genes and the proportions of immune cells were determined by CIBERSORT. Ten candidate HML2-encoded sRNAs were identified based on specific criteria, and three of them were further validated in SLE patients by qRT-PCR. The diagnostic values of these three sRNAs were evaluated in SLE and lupus nephritis (LN). This study suggested that HML2 genes and their encoded sRNAs might be involved in the immune regulation and progress of SLE. These potential sRNAs might function as regulatory molecules and diagnostic biomarkers of SLE and LN.
Collapse
Affiliation(s)
- Xinyi Liu
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, Harbin, 150081, China.,Heilongjiang Provincial Key Laboratory of Infection and Immunity, Pathogen Biology, Harbin, 150081, China
| | - Yanjun Ding
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, Harbin, 150081, China.,Heilongjiang Provincial Key Laboratory of Infection and Immunity, Pathogen Biology, Harbin, 150081, China
| | - Xiaoqiu Zheng
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, Harbin, 150081, China.,Heilongjiang Provincial Key Laboratory of Infection and Immunity, Pathogen Biology, Harbin, 150081, China
| | - He Huang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Liyu Shi
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, Harbin, 150081, China.,Heilongjiang Provincial Key Laboratory of Infection and Immunity, Pathogen Biology, Harbin, 150081, China
| | - Xiaolan Yang
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, Harbin, 150081, China.,Heilongjiang Provincial Key Laboratory of Infection and Immunity, Pathogen Biology, Harbin, 150081, China
| | - Jing Wei
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, Harbin, 150081, China.,Heilongjiang Provincial Key Laboratory of Infection and Immunity, Pathogen Biology, Harbin, 150081, China
| | - Yang Li
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Wenping Kao
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, Harbin, 150081, China.,Heilongjiang Provincial Key Laboratory of Infection and Immunity, Pathogen Biology, Harbin, 150081, China
| | - Fengmin Zhang
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, Harbin, 150081, China.,Heilongjiang Provincial Key Laboratory of Infection and Immunity, Pathogen Biology, Harbin, 150081, China
| | - Jun Qian
- Wu Lien-Teh Institute, Department of Microbiology, Harbin Medical University, Harbin, 150081, China.,Heilongjiang Provincial Key Laboratory of Infection and Immunity, Pathogen Biology, Harbin, 150081, China
| |
Collapse
|
43
|
Hou P, Lin Y, Li Z, Lu R, Wang Y, Tian T, Jia P, Zhang X, Cao L, Zhou Z, Li C, Gu J, Guo D. Autophagy receptor CCDC50 tunes the STING-mediated interferon response in viral infections and autoimmune diseases. Cell Mol Immunol 2021; 18:2358-2371. [PMID: 34453126 PMCID: PMC8484562 DOI: 10.1038/s41423-021-00758-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 08/04/2021] [Indexed: 02/07/2023] Open
Abstract
DNA sensing and timely activation of interferon (IFN)-mediated innate immunity are crucial for the defense against DNA virus infections and the clearance of abnormal cells. However, overactivation of immune responses may lead to tissue damage and autoimmune diseases; therefore, these processes must be intricately regulated. STING is the key adaptor protein, which is activated by cyclic GMP-AMP, the second messenger derived from cGAS-mediated DNA sensing. Here, we report that CCDC50, a newly identified autophagy receptor, tunes STING-directed type I IFN signaling activity by delivering K63-polyubiquitinated STING to autolysosomes for degradation. Knockout of CCDC50 significantly increases herpes simplex virus 1 (HSV-1)- or DNA ligand-induced production of type I IFN and proinflammatory cytokines. Ccdc50-deficient mice show increased production of IFN, decreased viral replication, reduced cell infiltration, and improved survival rates compared with their wild-type littermates when challenged with HSV-1. Remarkably, the expression of CCDC50 is downregulated in systemic lupus erythematosus (SLE), a chronic autoimmune disease. CCDC50 levels are negatively correlated with IFN signaling pathway activation and disease severity in human SLE patients. CCDC50 deficiency potentiates the cGAS-STING-mediated immune response triggered by SLE serum. Thus, our findings reveal the critical role of CCDC50 in the immune regulation of viral infections and autoimmune diseases and provide insights into the therapeutic implications of CCDC50 manipulation.
Collapse
Affiliation(s)
- Panpan Hou
- MOE Key Laboratory of Tropical Disease Control, Centre for Infection and Immunity Study (CIIS), School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Yuxin Lin
- MOE Key Laboratory of Tropical Disease Control, Centre for Infection and Immunity Study (CIIS), School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Zibo Li
- MOE Key Laboratory of Tropical Disease Control, Centre for Infection and Immunity Study (CIIS), School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Ruiqing Lu
- MOE Key Laboratory of Tropical Disease Control, Centre for Infection and Immunity Study (CIIS), School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Yicheng Wang
- MOE Key Laboratory of Tropical Disease Control, Centre for Infection and Immunity Study (CIIS), School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Tian Tian
- The Center for Applied Genomics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Penghui Jia
- MOE Key Laboratory of Tropical Disease Control, Centre for Infection and Immunity Study (CIIS), School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Xi Zhang
- Division of Rheumatology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Liu Cao
- MOE Key Laboratory of Tropical Disease Control, Centre for Infection and Immunity Study (CIIS), School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Zhongwei Zhou
- MOE Key Laboratory of Tropical Disease Control, Centre for Infection and Immunity Study (CIIS), School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Chunmei Li
- MOE Key Laboratory of Tropical Disease Control, Centre for Infection and Immunity Study (CIIS), School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Jieruo Gu
- Division of Rheumatology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Deyin Guo
- MOE Key Laboratory of Tropical Disease Control, Centre for Infection and Immunity Study (CIIS), School of Medicine, Sun Yat-sen University, Shenzhen, China.
| |
Collapse
|
44
|
Abstract
Innate immunity is regulated by a broad set of evolutionary conserved receptors to finely probe the local environment and maintain host integrity. Besides pathogen recognition through conserved motifs, several of these receptors also sense aberrant or misplaced self-molecules as a sign of perturbed homeostasis. Among them, self-nucleic acid sensing by the cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING) pathway alerts on the presence of both exogenous and endogenous DNA in the cytoplasm. We review recent literature demonstrating that self-nucleic acid detection through the STING pathway is central to numerous processes, from cell physiology to sterile injury, auto-immunity and cancer. We address the role of STING in autoimmune diseases linked to dysfunctional DNAse or related to mutations in DNA sensing pathways. We expose the role of the cGAS/STING pathway in inflammatory diseases, neurodegenerative conditions and cancer. Connections between STING in various cell processes including autophagy and cell death are developed. Finally, we review proposed mechanisms to explain the sources of cytoplasmic DNA.
Collapse
Affiliation(s)
| | - Nicolas Riteau
- Experimental and Molecular Immunology and Neurogenetics Laboratory (INEM), Centre National de la Recherche Scientifique (CNRS), UMR7355 and University of Orleans, Orleans, France
| |
Collapse
|
45
|
Regulation of cGAS-STING pathway - Implications for systemic lupus erythematosus. RHEUMATOLOGY AND IMMUNOLOGY RESEARCH 2021; 2:173-184. [PMID: 36465073 PMCID: PMC9524788 DOI: 10.2478/rir-2021-0023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/07/2021] [Indexed: 12/22/2022]
Abstract
Abstract
Type I interferon (IFN-I) is implicated in the pathogenesis of systemic lupus erythematosus (SLE) and the closely associated monogenic autoinflammatory disorders termed the “interferonopathies.” Recently, the cytosolic DNA sensor cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS) and its downstream signaling adaptor stimulator of interferon genes (STING) have been identified as having important, if not central, roles in driving IFN-I expression in response to self-DNA. This review highlights the many ways in which this pathway is regulated in order to prevent self-DNA recognition and underlines the importance of maintaining tight control in order to prevent autoimmune disease. We will discuss the murine and human studies that have implicated the cGAS-STING pathway as being an important contributor to breakdown in tolerance in SLE and highlight the potential therapeutic application of this knowledge for the treatment of SLE.
Collapse
|
46
|
Anastasiou M, Newton GA, Kaur K, Carrillo-Salinas FJ, Smolgovsky SA, Bayer AL, Ilyukha V, Sharma S, Poltorak A, Luscinskas FW, Alcaide P. Endothelial STING controls T cell transmigration in an IFNI-dependent manner. JCI Insight 2021; 6:e149346. [PMID: 34156982 PMCID: PMC8410041 DOI: 10.1172/jci.insight.149346] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/17/2021] [Indexed: 12/15/2022] Open
Abstract
The stimulator of IFN genes (STING) protein senses cyclic dinucleotides released in response to double-stranded DNA and functions as an adaptor molecule for type I IFN (IFNI) signaling by activating IFNI-stimulated genes (ISG). We found impaired T cell infiltration into the peritoneum in response to TNF-α in global and EC-specific STING-/- mice and discovered that T cell transendothelial migration (TEM) across mouse and human endothelial cells (EC) deficient in STING was strikingly reduced compared with control EC, whereas T cell adhesion was not impaired. STING-/- T cells showed no defect in TEM or adhesion to EC, or immobilized endothelial cell-expressed molecules ICAM1 and VCAM1, compared with WT T cells. Mechanistically, CXCL10, an ISG and a chemoattractant for T cells, was dramatically reduced in TNF-α-stimulated STING-/- EC, and genetic loss or pharmacologic antagonisms of IFNI receptor (IFNAR) pathway reduced T cell TEM. Our data demonstrate a central role for EC-STING during T cell TEM that is dependent on the ISG CXCL10 and on IFNI/IFNAR signaling.
Collapse
Affiliation(s)
- Marina Anastasiou
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, USA.,Department of Internal Medicine, University of Crete Medical School, Crete, Greece
| | - Gail A. Newton
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Kuljeet Kaur
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, USA
| | | | - Sasha A. Smolgovsky
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, USA.,Tufts Graduate School for Biomedical Sciences Immunology Program, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Abraham L. Bayer
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, USA.,Tufts Graduate School for Biomedical Sciences Immunology Program, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Vladimir Ilyukha
- Petrozavodsk State University, Petrozavodsk, Republic of Karelia, Russia
| | - Shruti Sharma
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, USA.,Tufts Graduate School for Biomedical Sciences Immunology Program, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Alexander Poltorak
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, USA.,Tufts Graduate School for Biomedical Sciences Immunology Program, Tufts University School of Medicine, Boston, Massachusetts, USA.,Petrozavodsk State University, Petrozavodsk, Republic of Karelia, Russia
| | - Francis W. Luscinskas
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Pilar Alcaide
- Department of Immunology, Tufts University School of Medicine, Boston, Massachusetts, USA.,Tufts Graduate School for Biomedical Sciences Immunology Program, Tufts University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
47
|
Serrano Nájera G, Narganes Carlón D, Crowther DJ. TrendyGenes, a computational pipeline for the detection of literature trends in academia and drug discovery. Sci Rep 2021; 11:15747. [PMID: 34344904 PMCID: PMC8333311 DOI: 10.1038/s41598-021-94897-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 07/08/2021] [Indexed: 02/07/2023] Open
Abstract
Target identification and prioritisation are prominent first steps in modern drug discovery. Traditionally, individual scientists have used their expertise to manually interpret scientific literature and prioritise opportunities. However, increasing publication rates and the wider routine coverage of human genes by omic-scale research make it difficult to maintain meaningful overviews from which to identify promising new trends. Here we propose an automated yet flexible pipeline that identifies trends in the scientific corpus which align with the specific interests of a researcher and facilitate an initial prioritisation of opportunities. Using a procedure based on co-citation networks and machine learning, genes and diseases are first parsed from PubMed articles using a novel named entity recognition system together with publication date and supporting information. Then recurrent neural networks are trained to predict the publication dynamics of all human genes. For a user-defined therapeutic focus, genes generating more publications or citations are identified as high-interest targets. We also used topic detection routines to help understand why a gene is trendy and implement a system to propose the most prominent review articles for a potential target. This TrendyGenes pipeline detects emerging targets and pathways and provides a new way to explore the literature for individual researchers, pharmaceutical companies and funding agencies.
Collapse
Affiliation(s)
- Guillermo Serrano Nájera
- Division of Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - David Narganes Carlón
- Division of Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
- Division of Population Health and Genomics, Ninewells Hospital, School of Medicine, University of Dundee, Dundee, DD1 9SY, UK
- Exscientia Ltd, Dundee One, River Court, 5 West Victoria Dock Road, Dundee, DD1 3JT, UK
| | - Daniel J Crowther
- Exscientia Ltd, Dundee One, River Court, 5 West Victoria Dock Road, Dundee, DD1 3JT, UK.
| |
Collapse
|
48
|
Crucial role of stimulator of interferon genes-dependent signaling in house dust mite extract-induced IgE production. Sci Rep 2021; 11:13157. [PMID: 34162937 PMCID: PMC8222396 DOI: 10.1038/s41598-021-92561-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/04/2021] [Indexed: 01/04/2023] Open
Abstract
Stimulator of interferon genes (STING) is a DNA sensor that responds to pathogens and induces type I interferon production. Herein, the role of STING in house dust mite extract (HDM)-induced allergic asthma was investigated. C57BL/6 wild-type (WT) and Sting−/− mice were intratracheally sensitized with HDM, and the bronchoalveolar lavage fluid (BALF), sera, lungs, and mediastinal lymph nodes (MLNs) were analyzed. The total and HDM-specific serum IgE levels were lower in Sting−/− mice than in WT mice. B cell and IgE-positive B cell proportion in BALF and MLNs, respectively, was significantly lower in Sting−/− mice than in WT mice. Additionally, cyclic GMP-AMP, a STING ligand, augmented total and HDM-specific serum IgE levels and B cell proportion in BALF when applied in combination with HDM. To elucidate the role of STING in IgE production, follicular helper T (Tfh) cells, which are involved in B cell maturation, were investigated. Tfh cell proportion in MLNs decreased in Sting−/− mice, and IL-4 and IL-13 production by HDM-restimulated MLN cells from HDM-sensitized mice was decreased in Sting−/− mice compared with WT mice. Thus, STING plays an important role in the maturation and class switching of IgE-producing B cells in allergic inflammation via Tfh cells.
Collapse
|
49
|
Wang H, Hu DQ, Xiao Q, Liu YB, Song J, Liang Y, Ruan JW, Wang ZZ, Li JX, Pan L, Wang MC, Zeng M, Shi LL, Xu K, Ning Q, Zhen G, Yu D, Wang DY, Wenzel SE, Liu Z. Defective STING expression potentiates IL-13 signaling in epithelial cells in eosinophilic chronic rhinosinusitis with nasal polyps. J Allergy Clin Immunol 2021; 147:1692-1703. [PMID: 33340608 DOI: 10.1016/j.jaci.2020.12.623] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 11/25/2020] [Accepted: 12/02/2020] [Indexed: 01/06/2023]
Abstract
BACKGROUND Stimulator of interferon genes (STING) activation favors effective innate immune responses against viral infections. Its role in chronic rhinosinusitis with nasal polyps (CRSwNP) remains unknown. OBJECTIVE Our aim was to explore the expression, regulation, and function of STING in CRSwNP. METHODS STING expression in sinonasal mucosal samples was analyzed by means of quantitative RT-PCR, immunohistochemistry, flow cytometry, and Western blotting. Regulation and function of STING expression were explored by using cultured primary human nasal epithelial cells (HNECs) and cells of the line BEAS-2B in vitro. RESULTS STING expression was reduced in eosinophilic nasal polyps compared with that in noneosinophilic nasal polyps and control tissues. STING was predominantly expressed by epithelial cells in nasal tissue and was downregulated by IL-4 and IL-13 in a signal transducer and activator of transcription 6 (STAT6)-dependent manner. HNECs derived from eosinophilic polyps displayed compromised STING-dependent type I interferon production but heightened IL-13-induced STAT6 activation and CCL26 production as compared with HNECs from noneosinophilic polyps and control tissues, which were rescued by exogenous STING overexpression. Knocking down or overexpressing STING decreased or enhanced expression of suppressor of cytokine signaling 1 (SOCS1) in BEAS-2B cells, respectively, independent of the canonic STING pathway elements TBK1 and IRF3. Knocking down SOCS1 abolished the inhibitory effect of STING on IL-13 signaling in BEAS-2B cells. STING expression was positively correlated with SOCS1 expression but negatively correlated with CCL26 expression in nasal epithelial cells from patients with CRSwNP. CONCLUSIONS Reduced STING expression caused by the type 2 milieu not only impairs STING-dependent type I interferon production but also amplifies IL-13 signaling by decreasing SOCS1 expression in nasal epithelial cells in eosinophilic CRSwNP.
Collapse
Affiliation(s)
- Hai Wang
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dan-Qing Hu
- Department of Infectious Disease, Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiao Xiao
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi-Bo Liu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Song
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuxia Liang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Respiratory Diseases of Ministry of Health, Wuhan, China
| | - Jian-Wen Ruan
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhe-Zheng Wang
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing-Xian Li
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Pan
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meng-Chen Wang
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Zeng
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li-Li Shi
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Xu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qin Ning
- Department of Infectious Disease, Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guohua Zhen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Respiratory Diseases of Ministry of Health, Wuhan, China
| | - Di Yu
- Department of Immunology and Infection Disease, John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - De-Yun Wang
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Sally E Wenzel
- University of Pittsburgh Asthma Institute at University of Pittsburgh Medical Center, Department of Environmental and Occupational Health, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pa
| | - Zheng Liu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
50
|
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: 50] [Impact Index Per Article: 12.5] [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
|