201
|
Du H, Kou M, Deng W, Zhou X, Zhang X, Huang Z, Ren B, Cai X, Xu S, Chen Y, Chen L, Chen C, Bao H, Chen Q, Li D. DWL-4-140: A allene small molecule targeting STING that alleviates lupus-like phenotype in Trex1 -/- mice. Biomed Pharmacother 2023; 165:115188. [PMID: 37480829 DOI: 10.1016/j.biopha.2023.115188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/14/2023] [Accepted: 07/18/2023] [Indexed: 07/24/2023] Open
Abstract
The innate immune system plays a critical role in the host response against pathogenic microbial infection. However, aberrant activation of the innate immune pathways is a characteristic feature of various diseases. Thus, targeted drugs must be developed based on the understanding of the innate immune signaling pathways. This study demonstrated that an allene small molecule (DWL-4-140) can efficiently and selectively exert regulatory effects on the stimulator of interferon genes (STING), resulting in the downregulation of DNA-induced interferon responses. Mechanistically, DWL-4-140 targeted the cyclized nucleotide-binding domain (CBD) of STING, inhibiting the assembly of the STING multimeric complex and the recruitment of downstream signaling mediators. In addition to downregulating the 10-carboxymethyl-9-acridanone-induced production of inflammatory factors, DWL-4-140 alleviated the pathological features of Trex1 deletion-induced lupus in mice. Thus, this study demonstrated that DWL-4-140 pharmacologically inhibits STING with potential therapeutic applications in auto-inflammatory diseases.
Collapse
Affiliation(s)
- Hekang Du
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, PR China
| | - Meng Kou
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, PR China; Cord Blood Bank Centre, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, PR China
| | - Weili Deng
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, PR China; Key Laboratory of Coal to Ethylene Glycol and Its Related Technology, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, PR China
| | - Xueyuan Zhou
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, PR China
| | - Xiaoxiong Zhang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, PR China
| | - Zhengrong Huang
- Department of Integrative Medicine, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province 350117, PR China
| | - Bowen Ren
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, PR China
| | - Xingting Cai
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, PR China
| | - Shan Xu
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, PR China
| | - Yu Chen
- Cancer Bio-immunotherapy Center, Clinical Oncology School of Fujian Medical University & Fujian Cancer Hospital, Fuzhou, Fujian Province, PR China; Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian Province, PR China
| | - Lizhu Chen
- Cancer Bio-immunotherapy Center, Clinical Oncology School of Fujian Medical University & Fujian Cancer Hospital, Fuzhou, Fujian Province, PR China; Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian Province, PR China
| | - Chuanben Chen
- Cancer Bio-immunotherapy Center, Clinical Oncology School of Fujian Medical University & Fujian Cancer Hospital, Fuzhou, Fujian Province, PR China; Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian Province, PR China.
| | - Hongli Bao
- Key Laboratory of Coal to Ethylene Glycol and Its Related Technology, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, PR China.
| | - Qi Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, PR China.
| | - Daliang Li
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University Qishan Campus, College Town, Fuzhou, Fujian Province 350117, PR China.
| |
Collapse
|
202
|
Zou Y, Zhang M, Zhou J. Recent trends in STING modulators: Structures, mechanisms, and therapeutic potential. Drug Discov Today 2023; 28:103694. [PMID: 37393985 DOI: 10.1016/j.drudis.2023.103694] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 06/05/2023] [Accepted: 06/26/2023] [Indexed: 07/04/2023]
Abstract
The cyclic GMP-AMP synthase stimulator (cGAS)-stimulator of interferon gene (STING) signaling pathway has an integral role in the host immune response through DNA sensing followed by inducing a robust innate immune defense program. STING has become a promising therapeutic target associated with multiple diseases, including various inflammatory diseases, cancer, and infectious diseases, among others. Thus, modulators of STING are regarded as emerging therapeutic agents. Recent progress has been made in STING research, including recently identified STING-mediated regulatory pathways, the development of a new STING modulator, and the new association of STING with disease. In this review, we focus on recent trends in the development of STING modulators, including structures, mechanisms, and clinical application.
Collapse
Affiliation(s)
- Yan Zou
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Department of Chemistry, Zhejiang Normal University, 688 Yingbin Road, Jinhua 321004, China; Drug Development and Innovation Center, College of Chemistry and Life Sciences, Zhejiang Normal University, 688 Yingbin Road, Jinhua 321004, China
| | - Min Zhang
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Department of Chemistry, Zhejiang Normal University, 688 Yingbin Road, Jinhua 321004, China; Drug Development and Innovation Center, College of Chemistry and Life Sciences, Zhejiang Normal University, 688 Yingbin Road, Jinhua 321004, China
| | - Jinming Zhou
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Department of Chemistry, Zhejiang Normal University, 688 Yingbin Road, Jinhua 321004, China; Drug Development and Innovation Center, College of Chemistry and Life Sciences, Zhejiang Normal University, 688 Yingbin Road, Jinhua 321004, China.
| |
Collapse
|
203
|
Tokgun PE, Karagenc N, Karasu U, Tokgun O, Turel S, Demiray A, Akca H, Yüksel S. Treatment of STING-associated vasculopathy with onset in infancy in patients carrying a novel mutation in the TMEM173 gene with the JAK3-inhibitor tofacitinib. Arch Rheumatol 2023; 38:461-467. [PMID: 38046254 PMCID: PMC10689023 DOI: 10.46497/archrheumatol.2023.9927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/09/2022] [Indexed: 10/18/2023] Open
Abstract
OBJECTIVES This study aimed to reveal the genetic background of patients in the two-generation family suffering from rheumatoid arthritis, psoriatic arthropathy pain, scratches, and bruises. PATIENTS AND METHODS A clinical exome sequencing analysis was performed in 10 individuals in the same family using the Sophia Genetics clinical exome solution kit. RESULTS A novel V194L mutation in the TMEM173 gene was identified in three members of the family. Two of the family members were treated with the JAK3 inhibitor tofacitinib and recovered completely one month after the treatment. CONCLUSION The V194L mutation was reported for the first time in this study, and a positive response was achieved with tofacitinib.
Collapse
Affiliation(s)
- Pervin Elvan Tokgun
- Department of Medical Genetics, Pamukkale University Faculty of Medicine, Denizli, Türkiye
| | - Nedim Karagenc
- Department of Medical Genetics, Pamukkale University Faculty of Medicine, Denizli, Türkiye
| | - Uğur Karasu
- Department of Internal Medicine, Division of Rheumatology, Pamukkale University Faculty of Medicine, Denizli, Türkiye
| | - Onur Tokgun
- Department of Medical Genetics, Pamukkale University Faculty of Medicine, Denizli, Türkiye
| | - Samet Turel
- Department of Medical Genetics, Pamukkale University Faculty of Medicine, Denizli, Türkiye
| | - Aydın Demiray
- Department of Medical Genetics, Pamukkale University Faculty of Medicine, Denizli, Türkiye
| | - Hakan Akca
- Department of Medical Genetics, Pamukkale University Faculty of Medicine, Denizli, Türkiye
| | - Selçuk Yüksel
- Department of Pediatric Nephrology and Pediatric Rheumatology, Pamukkale University Faculty of Medicine, Denizli, Türkiye
| |
Collapse
|
204
|
Zhou J, Zhuang Z, Li J, Feng Z. Significance of the cGAS-STING Pathway in Health and Disease. Int J Mol Sci 2023; 24:13316. [PMID: 37686127 PMCID: PMC10487967 DOI: 10.3390/ijms241713316] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway plays a significant role in health and disease. In this pathway, cGAS, one of the major cytosolic DNA sensors in mammalian cells, regulates innate immunity and the STING-dependent production of pro-inflammatory cytokines, including type-I interferon. Moreover, the cGAS-STING pathway is integral to other cellular processes, such as cell death, cell senescence, and autophagy. Activation of the cGAS-STING pathway by "self" DNA is also attributed to various infectious diseases and autoimmune or inflammatory conditions. In addition, the cGAS-STING pathway activation functions as a link between innate and adaptive immunity, leading to the inhibition or facilitation of tumorigenesis; therefore, research targeting this pathway can provide novel clues for clinical applications to treat infectious, inflammatory, and autoimmune diseases and even cancer. In this review, we focus on the cGAS-STING pathway and its corresponding cellular and molecular mechanisms in health and disease.
Collapse
Affiliation(s)
- Jinglin Zhou
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University Qishan Campus, Fuzhou 350117, China
| | - Zhan Zhuang
- Key Laboratory of College of First Clinical Medicine, College of First Clinical Medicine, Fujian Medical University, Taijiang Campus, Fuzhou 350001, China
| | - Jiamian Li
- Key Laboratory of College of First Clinical Medicine, College of First Clinical Medicine, Fujian Medical University, Taijiang Campus, Fuzhou 350001, China
| | - Zhihua Feng
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, College of Life Science, Fujian Normal University Qishan Campus, Fuzhou 350117, China
| |
Collapse
|
205
|
Gagne S, Sivaraman V, Akoghlanian S. Interferonopathies masquerading as non-Mendelian autoimmune diseases: pattern recognition for early diagnosis. Front Pediatr 2023; 11:1169638. [PMID: 37622085 PMCID: PMC10445166 DOI: 10.3389/fped.2023.1169638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 07/05/2023] [Indexed: 08/26/2023] Open
Abstract
Type I interferonopathies are a broad category of conditions associated with increased type I interferon gene expression and include monogenic autoinflammatory diseases and non-Mendelian autoimmune diseases such as dermatomyositis and systemic lupus erythematosus. While a wide range of clinical presentations among type I interferonopathies exists, these conditions often share several clinical manifestations and implications for treatment. Presenting symptoms may mimic non-Mendelian autoimmune diseases, including vasculitis and systemic lupus erythematosus, leading to delayed or missed diagnosis. This review aims to raise awareness about the varied presentations of monogenic interferonopathies to provide early recognition and appropriate treatment to prevent irreversible damage and improve quality of life and outcomes in this unique patient population.
Collapse
Affiliation(s)
- Samuel Gagne
- Division of Pediatric Rheumatology, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Vidya Sivaraman
- Division of Pediatric Rheumatology, Nationwide Children’s Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University, Columbus, OH, United States
| | - Shoghik Akoghlanian
- Division of Pediatric Rheumatology, Nationwide Children’s Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
206
|
Gao KM, Nündel K, Chiang K, Yin X, Utz PJ, Fitzgerald K, Marshak-Rothstein A. Activation of Autoreactive Lymphocytes in the Lung by STING Gain-of-function Mutation Radioresistant Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.28.551002. [PMID: 37546720 PMCID: PMC10402118 DOI: 10.1101/2023.07.28.551002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Gain-of-function mutations in the dsDNA sensing adaptor STING lead to a severe autoinflammatory syndrome known as STING-associated vasculopathy with onset in Infancy (SAVI). SAVI patients develop interstitial lung disease (ILD) and commonly produce anti-nuclear antibodies (ANAs), indicative of concomitant autoimmunity. Mice heterozygous for the most common SAVI mutation, V154M (VM), also develop ILD, triggered by nonhematopoietic VM cells, but exhibit severe peripheral lymphopenia, low serum Ig titers and fail to produce autoantibodies. In contrast, we now show that lethally irradiated VM mice reconstituted with WT stem cells (WT→VM chimeras) develop ANAs and lung-reactive autoantibodies associated with accumulation of activated lymphocytes and formation of germinal centers in lung tissues. Moreover, when splenocytes from WT→VM chimeras were adoptively transferred into unmanipulated Rag1 -/- mice, donor T cells accumulated in the lung. Overall, these findings demonstrate that expression of the VM mutation in non-hematopoietic cells can promote the activation of immunocompetent autoreactive lymphocytes. Summary Chimeric mice expressing STING only in non-hematopoietic cells develop systemic and lung directed autoimmunity which recapitulates what is seen in pediatric patients with SAVI disease.
Collapse
|
207
|
Gao KM, Chiang K, Korkmaz FT, Janardhan HP, Trivedi CM, Quinton LJ, Gingras S, Fitzgerald KA, Marshak-Rothstein A. Expression of a STING Gain-of-function Mutation in Endothelial Cells Initiates Lymphocytic Infiltration of the Lungs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.27.550897. [PMID: 37547024 PMCID: PMC10402179 DOI: 10.1101/2023.07.27.550897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Patients afflicted with STING gain-of-function mutations frequently present with debilitating interstitial lung disease ( ILD ) that is recapitulated in mice expressing the STING V154M mutation ( VM ). Prior radiation chimera studies revealed an unexpected and critical role for non-hematopoietic cells in the initiation of ILD. To identify STING-expressing non-hematopoietic cell types relevant to ILD, we generated a conditional knock-in ( CKI ) model in which expression of the VM allele was directed to hematopoietic cells, fibroblasts, epithelial cells, or endothelial cells. Only endothelial cell-targeted expression of the mutant allele resulted in the recruitment of immune cells to the lung and the formation of bronchus-associated lymphoid tissue, as seen in the parental VM strain. These findings reveal the importance of endothelial cells as instigators of STING-driven lung disease and suggest that therapeutic targeting of STING inhibitors to endothelial cells could potentially mitigate inflammation in the lungs of SAVI patients or patients afflicted with other ILD-related disorders. Summary Patients with STING gain-of-function (GOF) mutations develop life-threatening lung autoinflammation. In this study, Gao et al. utilize a mouse model of conditional STING GOF to demonstrate a role for endothelial STING GOF in initiating immune cell recruitment into lung tissues of SAVI mice.
Collapse
|
208
|
Todosenko N, Khaziakhmatova O, Malashchenko V, Yurova K, Bograya M, Beletskaya M, Vulf M, Gazatova N, Litvinova L. Mitochondrial Dysfunction Associated with mtDNA in Metabolic Syndrome and Obesity. Int J Mol Sci 2023; 24:12012. [PMID: 37569389 PMCID: PMC10418437 DOI: 10.3390/ijms241512012] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 07/22/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Metabolic syndrome (MetS) is a precursor to the major health diseases associated with high mortality in industrialized countries: cardiovascular disease and diabetes. An important component of the pathogenesis of the metabolic syndrome is mitochondrial dysfunction, which is associated with tissue hypoxia, disruption of mitochondrial integrity, increased production of reactive oxygen species, and a decrease in ATP, leading to a chronic inflammatory state that affects tissues and organ systems. The mitochondrial AAA + protease Lon (Lonp1) has a broad spectrum of activities. In addition to its classical function (degradation of misfolded or damaged proteins), enzymatic activity (proteolysis, chaperone activity, mitochondrial DNA (mtDNA)binding) has been demonstrated. At the same time, the spectrum of Lonp1 activity extends to the regulation of cellular processes inside mitochondria, as well as outside mitochondria (nuclear localization). This mitochondrial protease with enzymatic activity may be a promising molecular target for the development of targeted therapy for MetS and its components. The aim of this review is to elucidate the role of mtDNA in the pathogenesis of metabolic syndrome and its components as a key component of mitochondrial dysfunction and to describe the promising and little-studied AAA + LonP1 protease as a potential target in metabolic disorders.
Collapse
Affiliation(s)
- Natalia Todosenko
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia; (N.T.); (O.K.); (V.M.); (K.Y.); (M.B.); (M.B.); (M.V.); (N.G.)
| | - Olga Khaziakhmatova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia; (N.T.); (O.K.); (V.M.); (K.Y.); (M.B.); (M.B.); (M.V.); (N.G.)
| | - Vladimir Malashchenko
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia; (N.T.); (O.K.); (V.M.); (K.Y.); (M.B.); (M.B.); (M.V.); (N.G.)
| | - Kristina Yurova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia; (N.T.); (O.K.); (V.M.); (K.Y.); (M.B.); (M.B.); (M.V.); (N.G.)
| | - Maria Bograya
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia; (N.T.); (O.K.); (V.M.); (K.Y.); (M.B.); (M.B.); (M.V.); (N.G.)
| | - Maria Beletskaya
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia; (N.T.); (O.K.); (V.M.); (K.Y.); (M.B.); (M.B.); (M.V.); (N.G.)
| | - Maria Vulf
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia; (N.T.); (O.K.); (V.M.); (K.Y.); (M.B.); (M.B.); (M.V.); (N.G.)
| | - Natalia Gazatova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia; (N.T.); (O.K.); (V.M.); (K.Y.); (M.B.); (M.B.); (M.V.); (N.G.)
| | - Larisa Litvinova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia; (N.T.); (O.K.); (V.M.); (K.Y.); (M.B.); (M.B.); (M.V.); (N.G.)
- Laboratory of Cellular and Microfluidic Technologies, Siberian State Medical University, 634050 Tomsk, Russia
| |
Collapse
|
209
|
Rwandamuriye FX, Evans CW, Wylie B, Norret M, Vitali B, Ho D, Nguyen D, Roper EA, Wang T, Hepburn MS, Sanderson RW, Pfirrmann M, Fear VS, Forbes CA, Wyatt K, Ryan AL, Johns TG, Phillips MB, Hodder R, Leslie C, Kennedy BF, Zemek RM, Iyer KS, Lesterhuis WJ. A surgically optimized intraoperative poly(I:C)-releasing hydrogel prevents cancer recurrence. Cell Rep Med 2023; 4:101113. [PMID: 37467718 PMCID: PMC10394259 DOI: 10.1016/j.xcrm.2023.101113] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 05/10/2023] [Accepted: 06/18/2023] [Indexed: 07/21/2023]
Abstract
Recurrences frequently occur following surgical removal of primary tumors. In many cancers, adjuvant therapies have limited efficacy. Surgery provides access to the tumor microenvironment, creating an opportunity for local therapy, in particular immunotherapy, which can induce local and systemic anti-cancer effects. Here, we develop a surgically optimized biodegradable hyaluronic acid-based hydrogel for sustained intraoperative delivery of Toll-like receptor 3 agonist poly(I:C) and demonstrate that it significantly reduces tumor recurrence after surgery in multiple mouse models. Mechanistically, poly(I:C) induces a transient interferon alpha (IFNα) response, reshaping the tumor/wound microenvironment by attracting inflammatory monocytes and depleting regulatory T cells. We demonstrate that a pre-existing IFN signature predicts response to the poly(I:C) hydrogel, which sensitizes tumors to immune checkpoint therapy. The safety, immunogenicity, and surgical feasibility are confirmed in a veterinary trial in canine soft tissue tumors. The surgically optimized poly(I:C)-loaded hydrogel provides a safe and effective approach to prevent cancer recurrence.
Collapse
Affiliation(s)
| | - Cameron W Evans
- School of Molecular Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Ben Wylie
- Telethon Kids Institute, The University of Western Australia, Nedlands, WA, Australia
| | - Marck Norret
- School of Molecular Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Breana Vitali
- Telethon Kids Institute, The University of Western Australia, Nedlands, WA, Australia
| | - Diwei Ho
- School of Molecular Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Dat Nguyen
- Telethon Kids Institute, The University of Western Australia, Nedlands, WA, Australia
| | - Ellise A Roper
- Telethon Kids Institute, The University of Western Australia, Nedlands, WA, Australia
| | - Tao Wang
- Telethon Kids Institute, The University of Western Australia, Nedlands, WA, Australia
| | - Matt S Hepburn
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA, Australia; Centre for Medical Research, The University of Western Australia, Crawley, WA, Australia; Department of Electrical, Electronic and Computer Engineering, School of Engineering, The University of Western Australia, Crawley, WA, Australia
| | - Rowan W Sanderson
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA, Australia; Centre for Medical Research, The University of Western Australia, Crawley, WA, Australia; Department of Electrical, Electronic and Computer Engineering, School of Engineering, The University of Western Australia, Crawley, WA, Australia
| | - Maren Pfirrmann
- Telethon Kids Institute, The University of Western Australia, Nedlands, WA, Australia; Department of Medical BioSciences, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Vanessa S Fear
- Telethon Kids Institute, The University of Western Australia, Nedlands, WA, Australia
| | - Catherine A Forbes
- Telethon Kids Institute, The University of Western Australia, Nedlands, WA, Australia
| | - Ken Wyatt
- Perth Veterinary Specialists, Osborne Park, WA, Australia; Murdoch Veterinary School, Murdoch University, Murdoch, WA, Australia
| | - Anne L Ryan
- Telethon Kids Institute, The University of Western Australia, Nedlands, WA, Australia; Department of Oncology, Hematology and Tissue and Cellular Therapies, Perth Children's Hospital, Perth, WA, Australia
| | - Terrance G Johns
- Telethon Kids Institute, The University of Western Australia, Nedlands, WA, Australia
| | - Marianne B Phillips
- Telethon Kids Institute, The University of Western Australia, Nedlands, WA, Australia; Department of Oncology, Hematology and Tissue and Cellular Therapies, Perth Children's Hospital, Perth, WA, Australia
| | - Rupert Hodder
- Department of Surgery, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Connull Leslie
- Department of Anatomical Pathology, PathWest Laboratory Medicine, QEII Medical Centre, Nedlands, WA, Australia
| | - Brendan F Kennedy
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA, Australia; Centre for Medical Research, The University of Western Australia, Crawley, WA, Australia; Department of Electrical, Electronic and Computer Engineering, School of Engineering, The University of Western Australia, Crawley, WA, Australia
| | - Rachael M Zemek
- Telethon Kids Institute, The University of Western Australia, Nedlands, WA, Australia
| | | | | |
Collapse
|
210
|
Chauvin SD, Stinson WA, Platt DJ, Poddar S, Miner JJ. Regulation of cGAS and STING signaling during inflammation and infection. J Biol Chem 2023; 299:104866. [PMID: 37247757 PMCID: PMC10316007 DOI: 10.1016/j.jbc.2023.104866] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 05/31/2023] Open
Abstract
Stimulator of interferon genes (STING) is a sensor of cyclic dinucleotides including cyclic GMP-AMP, which is produced by cyclic GMP-AMP synthase (cGAS) in response to cytosolic DNA. The cGAS-STING signaling pathway regulates both innate and adaptive immune responses, as well as fundamental cellular functions such as autophagy, senescence, and apoptosis. Mutations leading to constitutive activation of STING cause devastating human diseases. Thus, the cGAS-STING pathway is of great interest because of its role in diverse cellular processes and because of the potential therapeutic implications of targeting cGAS and STING. Here, we review molecular and cellular mechanisms of STING signaling, and we propose a framework for understanding the immunological and other cellular functions of STING in the context of disease.
Collapse
Affiliation(s)
- Samuel D Chauvin
- Departments of Medicine and Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - W Alexander Stinson
- Departments of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Derek J Platt
- Department Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Subhajit Poddar
- Departments of Medicine and Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jonathan J Miner
- Departments of Medicine and Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA; Departments of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA; Department Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA; Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA.
| |
Collapse
|
211
|
Seok JK, Kim M, Kang HC, Cho YY, Lee HS, Lee JY. Beyond DNA sensing: expanding the role of cGAS/STING in immunity and diseases. Arch Pharm Res 2023:10.1007/s12272-023-01452-3. [PMID: 37354378 DOI: 10.1007/s12272-023-01452-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 06/13/2023] [Indexed: 06/26/2023]
Abstract
Cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) synthase (cGAS) is a DNA sensor that elicits a robust type I interferon response by recognizing ubiquitous danger-associated molecules. The cGAS/stimulator of interferon genes (cGAS/STING) is activated by endogenous DNA, including DNA released from mitochondria and extranuclear chromatin, as well as exogenous DNA derived from pathogenic microorganisms. cGAS/STING is positioned as a key axis of autoimmunity, the inflammatory response, and cancer progression, suggesting that the cGAS/STING signaling pathway represents an efficient therapeutic target. Based on the accumulated evidence, we present insights into the prevention and treatment of cGAS/STING-related chronic immune and inflammatory diseases. This review presents the current state of clinical and nonclinical development of modulators targeting cGAS/STING, providing useful information on the design of therapeutic strategies.
Collapse
Affiliation(s)
- Jin Kyung Seok
- College of Pharmacy, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Minhyuk Kim
- College of Pharmacy, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Han Chang Kang
- College of Pharmacy, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Yong-Yeon Cho
- College of Pharmacy, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Hye Suk Lee
- College of Pharmacy, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Joo Young Lee
- College of Pharmacy, The Catholic University of Korea, Bucheon, 14662, Republic of Korea.
| |
Collapse
|
212
|
Ong WWS, Dayal N, Chaudhuri R, Lamptey J, Sintim HO. STING antagonists, synthesized via Povarov-Doebner type multicomponent reaction. RSC Med Chem 2023; 14:1101-1113. [PMID: 37360395 PMCID: PMC10285770 DOI: 10.1039/d3md00061c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 03/23/2023] [Indexed: 09/22/2024] Open
Abstract
The cGAS-STING axis plays an important role in protecting higher organisms against invading pathogens or cancer by promoting the production of cytokines and interferons. However, persistent or uncontrolled activation of this pathway could lead to inflamed environments, which is detrimental to the host in the long run. Persistent activation of STING is known to be the cause of STING-associated vasculopathy with onset in infancy (SAVI) and activated STING is believed to play important roles in worsening various diseased states, such as traumatic brain injury, diabetic kidney disease and colitis. Thus, antagonists of STING could play important roles in managing various inflammatory diseases. Herein, we report the discovery of small molecule STING inhibitors, HSD1077 and analogs, which are facilely synthesized via a Povarov-Doebner type three-component reaction involving an amine, ketone, and aldehyde. Structure-activity relationship, SAR, studies indicate that both the 3H-pyrazolo[4,3-f]quinoline and pyrazole moieties in HSD1077 are critical for STING binding. At concentrations as low as 20 nM, HSD1077 suppressed type-1 interferon expression in both murine RAW macrophages and human THP-1 monocytes upon treatment with 100 μM 2'-3' cGAMP. Compounds containing the 3H-pyrazolo[4,3-f]quinoline moiety have the potential to be translated into anti-inflammatory compounds via STING inhibition.
Collapse
Affiliation(s)
- Wilson W S Ong
- Department of Chemistry, Purdue University 560 Oval Drive West Lafayette IN 47907 USA
| | - Neetu Dayal
- Department of Chemistry, Purdue University 560 Oval Drive West Lafayette IN 47907 USA
| | - Riddhi Chaudhuri
- Department of Chemistry, Purdue University 560 Oval Drive West Lafayette IN 47907 USA
| | - Jones Lamptey
- Department of Chemistry, Purdue University 560 Oval Drive West Lafayette IN 47907 USA
| | - Herman O Sintim
- Department of Chemistry, Purdue University 560 Oval Drive West Lafayette IN 47907 USA
- Purdue Institute for Drug Discovery, Purdue University 720 Clinic Drive West Lafayette IN 47907 USA
- Purdue Institute of Inflammation, Immunology, and Infectious Disease West Lafayette IN 47907 USA
| |
Collapse
|
213
|
Price-Kuehne F, Omoyinmi E, Younes M, Edwards M, Eleftheriou D, Brogan P. Case report: marfan syndrome (MFS) mimicking cutaneous vasculitis. Front Pediatr 2023; 11:1205255. [PMID: 37397156 PMCID: PMC10308279 DOI: 10.3389/fped.2023.1205255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 05/30/2023] [Indexed: 07/04/2023] Open
Abstract
Marfan syndrome (MFS) is an autosomal dominant connective tissue disorder caused by variants in the extracellular microfibril fibrillin (FBN1) gene. Here we report an FBN1 variant in a child with an unusual skin rash mimicking cutaneous vasculitis, and mild aortic root dilatation. The case was complicated by lack of typical skeletal MFS phenotype; and severe needle phobia preventing any blood testing for workup of suspected vasculitis. Therefore inflammatory markers, autoantibody profile and general hematology/biochemistry results were unknown. Diagnosis of MFS was made via genetic testing of a saliva sample alone using a next-generation sequencing (NGS) targeted gene panel designed to screen for monogenic forms of vasculitis and noninflammatory vasculopathic mimics. This revealed the patient was heterozygous for a pathogenic frameshift variant in FBN1; NM_000138, c.1211delC, p.(Pro404Hisfs*44), predicted to cause premature protein truncation leading to loss of function. The variant has not been detected in control populations and has previously been detected in individuals with MFS. This rapid diagnosis significantly impacted the patient management: avoidance of invasive investigations; avoidance of unnecessary immunosuppression; facilitating genetic counselling of the index case and family; and directly informing lifelong monitoring and ongoing treatment for aortic root involvement from MFS. This case further emphasizes the diagnostic utility of NGS early in the diagnostic workup of paediatric patients referred with suspected vasculitis, and we emphasize that MFS can present with cutaneous vasculitic-like features in the absence of the typical Marfanoid skeletal phenotype.
Collapse
Affiliation(s)
- Fiona Price-Kuehne
- Infection, Immunity and Inflammation Department, University College London Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Ebun Omoyinmi
- Infection, Immunity and Inflammation Department, University College London Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Maha Younes
- Clinical Genetics and Genomics Laboratory, Royal Brompton Hospital, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Matthew Edwards
- Clinical Genetics and Genomics Laboratory, Royal Brompton Hospital, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Despina Eleftheriou
- Infection, Immunity and Inflammation Department, University College London Great Ormond Street Institute of Child Health, London, United Kingdom
- Rheumatology Department, Great Ormond Street Hospital NHS Foundations Trust, London, United Kingdom
| | - Paul Brogan
- Infection, Immunity and Inflammation Department, University College London Great Ormond Street Institute of Child Health, London, United Kingdom
- Rheumatology Department, Great Ormond Street Hospital NHS Foundations Trust, London, United Kingdom
| |
Collapse
|
214
|
Xie R, Fan J, Wen J, Jin K, Zhan J, Yuan S, Tang Y, Nie X, Wen Z, Li H, Chen C, Wang DW. LncRNA ZNF593-AS alleviates diabetic cardiomyopathy via suppressing IRF3 signaling pathway. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 32:689-703. [PMID: 37215148 PMCID: PMC10199406 DOI: 10.1016/j.omtn.2023.04.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 04/28/2023] [Indexed: 05/24/2023]
Abstract
Diabetes could directly induce cardiac injury, leading to cardiomyopathy. However, treatment strategies for diabetic cardiomyopathy remain limited. ZNF593-AS knockout and cardiomyocyte-specific transgenic mice were constructed. In addition, high-fat diet (HFD)-induced diabetic mouse model and db/db mice, another classic diabetic mouse model, were employed. ZNF593-AS was silenced using GapmeR, a modified antisense oligonucleotide, while overexpressed using a recombinant adeno-associated virus serotype 9-mediated gene delivery system. Transcriptome sequencing, RNA pull-down assays, and RNA immunoprecipitation assays were also performed to investigate the underlying mechanisms. ZNF593-AS expression was decreased in diabetic hearts. ZNF593-AS attenuated the palmitic acid-induced apoptosis of cardiomyocytes in vitro. In HFD-induced diabetic mice, ZNF593-AS deletion aggravated cardiac dysfunction and enhanced cardiac apoptosis and inflammation. In contrast, HFD-induced cardiac dysfunction was improved in ZNF593-AS transgenic mice. Consistently, ZNF593-AS exerted the same cardioprotective effects in db/db mice. Mechanistically, ZNF593-AS directly interacted with the functional domain of interferon regulatory factor 3 (IRF3), and suppressed fatty acid-induced phosphorylation and activation of IRF3, contributing to the amelioration of cardiac cell death and inflammation. In conclusion, our results identified the protective role of ZNF593-AS in diabetic cardiomyopathy, suggesting a novel potential therapeutic target.
Collapse
Affiliation(s)
- Rong Xie
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jiahui Fan
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jianpei Wen
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Kunying Jin
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jiabing Zhan
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shuai Yuan
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yuyan Tang
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiang Nie
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zheng Wen
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Huaping Li
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chen Chen
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Dao Wen Wang
- Division of Cardiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
215
|
Xie W, Patel DJ. Structure-based mechanisms of 2'3'-cGAMP intercellular transport in the cGAS-STING immune pathway. Trends Immunol 2023; 44:450-467. [PMID: 37147228 PMCID: PMC11824902 DOI: 10.1016/j.it.2023.04.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 05/07/2023]
Abstract
Upon activation by double-stranded DNA (dsDNA), the cytosolic dsDNA sensor cyclic GMP-AMP synthase (cGAS) synthesizes the diffusible cyclic dinucleotide 2'3'-cGAMP (cyclic GMP-AMP), which subsequently binds to the adaptor STING, triggering a cascade of events leading to an inflammatory response. Recent studies have highlighted the role of 2'3'-cGAMP as an 'immunotransmitter' between cells, a process facilitated by gap junctions as well as by specialized membrane-spanning importer and exporter channels. This review highlights recent advances from a structural perspective of intercellular trafficking of 2'3'-cGAMP, with particular emphasis on the binding of importer SLC19A1 to 2'3'-cGAMP, as well as the significance of associated folate nutrients and antifolate therapeutics. This provides a path forward for structure-guided understanding of the transport cycle in immunology, as well as for candidate targeting approaches towards therapeutic intervention in inflammation.
Collapse
Affiliation(s)
- Wei Xie
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 311027, China; Department of Infectious Diseases, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China.
| | - Dinshaw J Patel
- Structural Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
216
|
Shen H, Jin L, Zheng Q, Ye Z, Cheng L, Wu Y, Wu H, Jon TG, Liu W, Pan Z, Mao Z, Wang Y. Synergistically targeting synovium STING pathway for rheumatoid arthritis treatment. Bioact Mater 2023; 24:37-53. [PMID: 36582350 PMCID: PMC9761476 DOI: 10.1016/j.bioactmat.2022.12.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 10/23/2022] [Accepted: 12/01/2022] [Indexed: 12/13/2022] Open
Abstract
Rheumatoid arthritis (RA) is a common autoimmune disease leading to pain, disability, and even death. Although studies have revealed that aberrant activation of STING was implicated in various autoimmune diseases, the role of STING in RA remains unclear. In the current study, we demonstrated that STING activation was pivotal in RA pathogenesis. As the accumulation of dsDNA, a specific stimulus for STING, is a feature of RA, we developed a spherical polyethyleneimine-coated mesoporous polydopamine nanoparticles loaded with STING antagonist C-176 (PEI-PDA@C-176 NPs) for treating RA. The fabricated NPs with biocompatibility had high DNA adsorption ability and could effectively inhibit the STING pathway and inflammation in macrophages. Intra-articular administration of PEI-PDA@C-176 NPs could effectively reduce joint damage in mice models of dsDNA-induced arthritis and collagen-induced arthritis by inhibiting STING pathway. We concluded that materials with synergistic effects of STING inhibition might be an efficacious strategy to treat RA.
Collapse
Affiliation(s)
- Haotian Shen
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Lulu Jin
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Qiangqiang Zheng
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Ziqiang Ye
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Linxiang Cheng
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yuxu Wu
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, N1 Shangchen Road, Yiwu, Zhejiang, 322000, China
| | - Honghao Wu
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Tae Gyong Jon
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Wenduo Liu
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Zongyou Pan
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yue Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| |
Collapse
|
217
|
Adeva-Andany MM, Funcasta-Calderón R, Fernández-Fernández C, Ameneiros-Rodríguez E, Vila-Altesor M, Castro-Quintela E. The metabolic effects of APOL1 in humans. Pflugers Arch 2023:10.1007/s00424-023-02821-z. [PMID: 37261508 PMCID: PMC10233197 DOI: 10.1007/s00424-023-02821-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 05/04/2023] [Accepted: 05/21/2023] [Indexed: 06/02/2023]
Abstract
Harboring apolipoprotein L1 (APOL1) variants coded by the G1 or G2 alleles of the APOL1 gene increases the risk for collapsing glomerulopathy, focal segmental glomerulosclerosis, albuminuria, chronic kidney disease, and accelerated kidney function decline towards end-stage kidney disease. However, most subjects carrying APOL1 variants do not develop the kidney phenotype unless a second clinical condition adds to the genotype, indicating that modifying factors modulate the genotype-phenotype correlation. Subjects with an APOL1 high-risk genotype are more likely to develop essential hypertension or obesity, suggesting that carriers of APOL1 risk variants experience more pronounced insulin resistance compared to noncarriers. Likewise, arterionephrosclerosis (the pathological correlate of hypertension-associated nephropathy) and glomerulomegaly take place among carriers of APOL1 risk variants, and these pathological changes are also present in conditions associated with insulin resistance, such as essential hypertension, aging, and diabetes. Insulin resistance may contribute to the clinical features associated with the APOL1 high-risk genotype. Unlike carriers of wild-type APOL1, bearers of APOL1 variants show impaired formation of lipid droplets, which may contribute to inducing insulin resistance. Nascent lipid droplets normally detach from the endoplasmic reticulum into the cytoplasm, although the proteins that enable this process remain to be fully defined. Wild-type APOL1 is located in the lipid droplet, whereas mutated APOL1 remains sited at the endoplasmic reticulum, suggesting that normal APOL1 may participate in lipid droplet biogenesis. The defective formation of lipid droplets is associated with insulin resistance, which in turn may modulate the clinical phenotype present in carriers of APOL1 risk variants.
Collapse
Affiliation(s)
- María M Adeva-Andany
- Nephrology Division, Internal Medicine Department, Hospital General Juan Cardona, c/ Pardo Bazán s/n, 15406, Ferrol, Spain.
| | - Raquel Funcasta-Calderón
- Nephrology Division, Internal Medicine Department, Hospital General Juan Cardona, c/ Pardo Bazán s/n, 15406, Ferrol, Spain
| | - Carlos Fernández-Fernández
- Nephrology Division, Internal Medicine Department, Hospital General Juan Cardona, c/ Pardo Bazán s/n, 15406, Ferrol, Spain
| | - Eva Ameneiros-Rodríguez
- Nephrology Division, Internal Medicine Department, Hospital General Juan Cardona, c/ Pardo Bazán s/n, 15406, Ferrol, Spain
| | - Matilde Vila-Altesor
- Nephrology Division, Internal Medicine Department, Hospital General Juan Cardona, c/ Pardo Bazán s/n, 15406, Ferrol, Spain
| | - Elvira Castro-Quintela
- Nephrology Division, Internal Medicine Department, Hospital General Juan Cardona, c/ Pardo Bazán s/n, 15406, Ferrol, Spain
| |
Collapse
|
218
|
Fogel LA, Bigley TM, Cole JJ, Estabrook MM, Kitcharoensakkul M. Aicardi-Goutières syndrome presenting with pneumocystis jirovecii pneumonia. Pediatr Allergy Immunol 2023; 34:e13968. [PMID: 37366204 DOI: 10.1111/pai.13968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 05/15/2023] [Accepted: 05/17/2023] [Indexed: 06/28/2023]
Affiliation(s)
- Leslie A Fogel
- Division of Rheumatology/Immunology, Department of Pediatrics, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Tarin M Bigley
- Division of Rheumatology/Immunology, Department of Pediatrics, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Jordan J Cole
- Division of Neurology, Department of Pediatrics, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Michele M Estabrook
- Division of Infectious Diseases, Department of Pediatrics, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Maleewan Kitcharoensakkul
- Division of Rheumatology/Immunology, Department of Pediatrics, Washington University in St. Louis, St. Louis, Missouri, USA
- Division of Allergy and Pulmonary Medicine, Department of Pediatrics, Washington University in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
219
|
Abstract
2'3'-cGAMP is a key molecule in the cGAS-STING pathway. This cyclic dinucleotide is produced by the cytosolic DNA sensor cGAS in response to the presence of aberrant dsDNA in the cytoplasm which is associated with microbial invasion or cellular damage. 2'3'-cGAMP acts as a second messenger and activates STING, the central hub of DNA sensing, to induce type-I interferons and pro-inflammatory cytokines necessary for responses against infection, cancer or cellular stress. Classically, detection of pathogens or danger by pattern recognition receptors (PRR) was thought to signal and induce the production of interferon and pro-inflammatory cytokines in the cell where sensing occurred. These interferon and cytokines then signal in both an autocrine and paracrine manner to induce responses in neighboring cells. Deviating from this dogma, recent studies have identified multiple mechanisms by which 2'3'-cGAMP can travel to neighboring cells where it activates STING independent of DNA sensing by cGAS. This observation is of great importance, as the cGAS-STING pathway is involved in immune responses against microbial invaders and cancer while its dysregulation drives the pathology of a wide range of inflammatory diseases to which antagonists have been elusive. In this review, we describe the fast-paced discoveries of the mechanisms by which 2'3'-cGAMP can be transported. We further highlight the diseases where they are important and detail how this change in perspective can be applied to vaccine design, cancer immunotherapies and treatment of cGAS-STING associated disease.
Collapse
Affiliation(s)
- Henry T. W. Blest
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Lise Chauveau
- Institut de Recherche en Infectiologie de Montpellier (IRIM) - CNRS UMR 9004, Université de Montpellier, Montpellier, France
| |
Collapse
|
220
|
Coderch C, Arranz-Herrero J, Nistal-Villan E, de Pascual-Teresa B, Rius-Rocabert S. The Many Ways to Deal with STING. Int J Mol Sci 2023; 24:ijms24109032. [PMID: 37240378 DOI: 10.3390/ijms24109032] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
The stimulator of interferon genes (STING) is an adaptor protein involved in the activation of IFN-β and many other genes associated with the immune response activation in vertebrates. STING induction has gained attention from different angles such as the potential to trigger an early immune response against different signs of infection and cell damage, or to be used as an adjuvant in cancer immune treatments. Pharmacological control of aberrant STING activation can be used to mitigate the pathology of some autoimmune diseases. The STING structure has a well-defined ligand binding site that can harbor natural ligands such as specific purine cyclic di-nucleotides (CDN). In addition to a canonical stimulation by CDNs, other non-canonical stimuli have also been described, whose exact mechanism has not been well defined. Understanding the molecular insights underlying the activation of STING is important to realize the different angles that need to be considered when designing new STING-binding molecules as therapeutic drugs since STING acts as a versatile platform for immune modulators. This review analyzes the different determinants of STING regulation from the structural, molecular, and cell biology points of view.
Collapse
Affiliation(s)
- Claire Coderch
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Boadilla del Monte, Spain
| | - Javier Arranz-Herrero
- Transplant Immunology Unit, National Center of Microbiology, Instituto de Salud Carlos III, 28220 Majadahonda, Spain
- Departamento CC, Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Boadilla del Monte, Spain
- Institute of Applied Molecular Medicine (IMMA), Department of Basic Medical Sciences, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Boadilla del Monte, Spain
| | - Estanislao Nistal-Villan
- Departamento CC, Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Boadilla del Monte, Spain
- Institute of Applied Molecular Medicine (IMMA), Department of Basic Medical Sciences, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Boadilla del Monte, Spain
| | - Beatriz de Pascual-Teresa
- Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Boadilla del Monte, Spain
| | - Sergio Rius-Rocabert
- Departamento CC, Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Boadilla del Monte, Spain
- Institute of Applied Molecular Medicine (IMMA), Department of Basic Medical Sciences, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Urbanización Montepríncipe, 28668 Boadilla del Monte, Spain
| |
Collapse
|
221
|
McCarty E, Yu J, Ninh VK, Calcagno DM, Lee J, King KR. Single cell transcriptomics of bone marrow derived macrophages reveals Ccl5 as a biomarker of direct IFNAR-independent responses to DNA sensing. Front Immunol 2023; 14:1199730. [PMID: 37275883 PMCID: PMC10232813 DOI: 10.3389/fimmu.2023.1199730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/02/2023] [Indexed: 06/07/2023] Open
Abstract
Introduction The type I interferon (IFN) response is an innate immune program that mediates anti-viral, anti-cancer, auto-immune, auto-inflammatory, and sterile injury responses. Bone marrow derived macrophages (BMDMs) are commonly used to model macrophage type I IFN responses, but the use of bulk measurement techniques obscures underlying cellular heterogeneity. This is particularly important for the IFN response to immune stimulatory double-stranded DNA (dsDNA) because it elicits overlapping direct and indirect responses, the latter of which depend on type I IFN cytokines signaling via the IFN alpha receptor (IFNAR) to upregulate expression of interferon stimulated genes (ISGs). Single cell transcriptomics has emerged as a powerful tool for revealing functional variability within cell populations. Methods Here, we use single cell RNA-Seq to examine BMDM heterogeneity at steady state and after immune-stimulatory DNA stimulation, with or without IFNAR-dependent amplification. Results We find that many macrophages express ISGs after DNA stimulation. We also find that a subset of macrophages express ISGs even if IFNAR is inhibited, suggesting that they are direct responders. Analysis of this subset reveals Ccl5 to be an IFNAR-independent marker gene of direct DNA sensing cells. Discussion Our studies provide a method for studying direct responders to IFN-inducing stimuli and demonstrate the importance of characterizing BMDM models of innate immune responses with single cell resolution.
Collapse
Affiliation(s)
- Emily McCarty
- Department of Bioengineering, Jacobs School of Engineering, University of California San Diego, La Jolla, CA, United States
| | - Justin Yu
- Department of Bioengineering, Jacobs School of Engineering, University of California San Diego, La Jolla, CA, United States
| | - Van K. Ninh
- Department of Bioengineering, Jacobs School of Engineering, University of California San Diego, La Jolla, CA, United States
| | - David M. Calcagno
- Department of Bioengineering, Jacobs School of Engineering, University of California San Diego, La Jolla, CA, United States
| | - Jodi Lee
- Department of Bioengineering, Jacobs School of Engineering, University of California San Diego, La Jolla, CA, United States
| | - Kevin R. King
- Department of Bioengineering, Jacobs School of Engineering, University of California San Diego, La Jolla, CA, United States
- Division of Cardiovascular Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
222
|
García-Martínez K, Chen J, Jones J, Woo A, Aucapina A, Brito I, Leifer CA. Stimulator of interferon genes is required for Toll-Like Receptor-8 induced interferon response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.15.540812. [PMID: 37292640 PMCID: PMC10245589 DOI: 10.1101/2023.05.15.540812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The innate immune system is equipped with multiple receptors to detect microbial nucleic acids and induce type I interferon (IFN) to restrict viral replication. When dysregulated these receptor pathways induce inflammation in response to host nucleic acids and promote development and persistence of autoimmune diseases like Systemic Lupus Erythematosus (SLE). IFN production is regulated by the Interferon Regulatory Factor (IRF) transcription factor family of proteins that function downstream of several innate immune receptors such as Toll-like receptors (TLRs) and Stimulator of Interferon Genes (STING). Although both TLRs and STING activate the same downstream molecules, the pathway by which TLRs and STING activate IFN response are thought to be independent. Here we show that STING plays a previously undescribed role in human TLR8 signaling. Stimulation with the TLR8 ligands induced IFN secretion in primary human monocytes, and inhibition of STING reduced IFN secretion from primary monocytes from 8 healthy donors. We demonstrate that TLR8-induced IRF activity was reduced by STING inhibitors. Moreover, TLR8-induced IRF activity was blocked by inhibition or loss of IKKε, but not TBK1. Bulk RNA transcriptomic analysis supported a model where TLR8 induces transcriptional responses associated with SLE that can be downregulated by inhibition of STING. These data demonstrate that STING is required for full TLR8-to-IRF signaling and provide evidence for a new framework of crosstalk between cytosolic and endosomal innate immune receptors, which could be leveraged to treat IFN driven autoimmune diseases. Background High levels of type I interferon (IFN) is characteristic of multiple autoimmune diseases, and while TLR8 is associated with autoimmune disease and IFN production, the mechanisms of TLR8-induced IFN production are not fully understood. Results STING is phosphorylated following TLR8 signaling, which is selectively required for the IRF arm of TLR8 signaling and for TLR8-induced IFN production in primary human monocytes. Conclusion STING plays a previously unappreciated role in TLR8-induced IFN production. Significance Nucleic acid-sensing TLRs contribute to development and progression of autoimmune disease including interferonopathies, and we show a novel role for STING in TLR-induced IFN production that could be a therapeutic target.
Collapse
|
223
|
Haşlak F, Kılıç Könte E, Aslan E, Şahin S, Kasapçopur Ö. Type I Interferonopathies in Childhood. Balkan Med J 2023; 40:165-174. [PMID: 37161741 PMCID: PMC10175883 DOI: 10.4274/balkanmedj.galenos.2023.2023-4-78] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/11/2023] Open
Abstract
Type 1 interferonopathy is a novel context reflecting a group of inborn disorders sharing common pathway disturbances. This group of diseases is characterized by autoimmunity and autoinflammation caused by an upregulation of type 1 interferons (IFN)s due to certain genetic mutations. Several features are common in most of the diseases in this group, such as vasculitic skin changes, including chilblains, panniculitis, interstitial lung disease, basal ganglion calcifications, neuromotor impairments, epilepsy, stroke, and recurrent fever. Family history and consanguineous marriage are also common. IFN signature is a useful diagnostic tool and is positive in almost all patients with type 1 interferonopathies. Although IFN signature is a sensitive test, its specificity is relatively low. It can also be positive in viral infections and several connective tissue diseases. Therefore, next-generation sequence methods, whole exome sequencing (WES) in particular, are required for the ultimate diagnosis. The optimal treatment regime is still under debate due to a lack of clinical trials. Although high-dose steroids, anti-IL-1 and anti-IL-6 treatments, and reverse transcriptase inhibitors are used, JAK inhibitors are highly promising. Additionally, monoclonal antibodies against IFN-alpha and interferon-α receptor (IFNAR) are currently underway.
Collapse
Affiliation(s)
- Fatih Haşlak
- Department of Pediatric Rheumatology, İstanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine, İstanbul, Turkey
| | - Elif Kılıç Könte
- Department of Pediatric Rheumatology, İstanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine, İstanbul, Turkey
| | - Esma Aslan
- Department of Pediatric Rheumatology, İstanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine, İstanbul, Turkey
| | - Sezgin Şahin
- Department of Pediatric Rheumatology, İstanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine, İstanbul, Turkey
| | - Özgür Kasapçopur
- Department of Pediatric Rheumatology, İstanbul University-Cerrahpaşa, Cerrahpaşa Faculty of Medicine, İstanbul, Turkey
| |
Collapse
|
224
|
Ouyang Z, Xu J, Liu T, Lin S, Sun Y, Huang Y, Zheng Z, Zeng G, Li C, Li S, Ding Y. STING/TBK1 Regulates Inflammation in Macrophages and Titanium Particles-Induced Osteolysis. ACS Biomater Sci Eng 2023. [PMID: 37134278 DOI: 10.1021/acsbiomaterials.2c01509] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Inflammatory response in macrophages on account of prostheses-derived wear particles is the leading cause of artificial joint failure. However, the mechanism by which wear particles initiate macrophage inflammation has not been fully elucidated. Previous research studies have identified TANK-binding kinase 1 (TBK1) and stimulator of interferon genes (STING) as potential factors in inflammation and autoimmune diseases. Here, we found that both TBK1 and STING were increased in synovium from aseptic loosening (AL) patients and were activated in titanium particles (TiPs)-stimulated macrophages. Lentivirus-mediated knockdown of TBK or STING significantly inhibited the inflammatory effects of macrophages, while overexpression of TBK or STING exerted opposite results. In concrete, STING/TBK1 promoted the activation of NF-κB and IRF3 pathways and macrophage M1 polarization. For further validation, a mice cranial osteolysis model was constructed for in vivo assays, and we found that STING-overexpressed lentivirus injection exacerbated osteolysis and inflammation, which was counteracted by TBK1-knockdown injection. In conclusion, STING/TBK1 enhanced TiP-induced macrophage inflammation and osteolysis via orchestrating the activation of NF-κB and IRF3 pathways and M1 polarization, which suggested STING/TBK1 as potential therapeutic targets for preventing AL of prostheses.
Collapse
Affiliation(s)
- Zhuji Ouyang
- Department of Orthopedic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yanjiang West Road, Yuexiu District, Guangzhou 510120, China
| | - Jing Xu
- Department of Orthopedic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yanjiang West Road, Yuexiu District, Guangzhou 510120, China
| | - Taihe Liu
- Department of Orthopedic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yanjiang West Road, Yuexiu District, Guangzhou 510120, China
| | - Sipeng Lin
- Department of Orthopedic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yanjiang West Road, Yuexiu District, Guangzhou 510120, China
| | - Yujun Sun
- Department of Orthopedic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yanjiang West Road, Yuexiu District, Guangzhou 510120, China
| | - Yuhsi Huang
- Department of Orthopedic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yanjiang West Road, Yuexiu District, Guangzhou 510120, China
| | - Zhongcan Zheng
- Department of Orthopedic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yanjiang West Road, Yuexiu District, Guangzhou 510120, China
| | - Gang Zeng
- Department of Orthopedic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yanjiang West Road, Yuexiu District, Guangzhou 510120, China
| | - Changchuan Li
- Department of Orthopedic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yanjiang West Road, Yuexiu District, Guangzhou 510120, China
| | - Shixun Li
- Department of Orthopedic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yanjiang West Road, Yuexiu District, Guangzhou 510120, China
| | - Yue Ding
- Department of Orthopedic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, No. 107, Yanjiang West Road, Yuexiu District, Guangzhou 510120, China
| |
Collapse
|
225
|
Kida Y, Doi T, Imanaka Y, Amatya VJ, Shime N, Okada S. Lethal Interstitial Lung Disease Associated with a Gain-of-Function Mutation in IFIH1. J Clin Immunol 2023:10.1007/s10875-023-01494-8. [PMID: 37126154 DOI: 10.1007/s10875-023-01494-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 04/13/2023] [Indexed: 05/02/2023]
Affiliation(s)
- Yoshiko Kida
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takehiko Doi
- Department of Pediatrics, Graduate School of Biomedical and Health Science, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima-Shi, Hiroshima, 734-8551, Japan.
| | - Yusuke Imanaka
- Department of Pediatrics, Graduate School of Biomedical and Health Science, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima-Shi, Hiroshima, 734-8551, Japan
| | - Vishwa Jeet Amatya
- Department of Pathology, Graduate School of Biomedical and Health Science, Hiroshima University, Hiroshima, Japan
| | - Nobuaki Shime
- Department of Emergency and Critical Care Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Satoshi Okada
- Department of Pediatrics, Graduate School of Biomedical and Health Science, Hiroshima University, 1-2-3 Kasumi, Minami-Ku, Hiroshima-Shi, Hiroshima, 734-8551, Japan
| |
Collapse
|
226
|
Jeremiah N, Ferran H, Antoniadou K, De Azevedo K, Nikolic J, Maurin M, Benaroch P, Manel N. RELA tunes innate-like interferon I/III responses in human T cells. J Exp Med 2023; 220:e20220666. [PMID: 36820829 PMCID: PMC9998965 DOI: 10.1084/jem.20220666] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 11/11/2022] [Accepted: 01/10/2023] [Indexed: 02/24/2023] Open
Abstract
In innate immune cells, intracellular sensors such as cGAS-STING stimulate type I/III interferon (IFN) expression, which promotes antiviral defense and immune activation. However, how IFN-I/III expression is controlled in adaptive cells is poorly understood. Here, we identify a transcriptional rheostat orchestrated by RELA that confers human T cells with innate-like abilities to produce IFN-I/III. Despite intact cGAS-STING signaling, IFN-I/III responses are stunted in CD4+ T cells compared with dendritic cells or macrophages. We find that lysine residues in RELA tune the IFN-I/III response at baseline and in response to STING stimulation in CD4+ T cells. This response requires positive feedback driven by cGAS and IRF7 expression. By combining RELA with IRF3 and DNA demethylation, IFN-I/III production in CD4+ T cells reaches levels observed in dendritic cells. IFN-I/III production provides self-protection of CD4+ T cells against HIV infection and enhances the elimination of tumor cells by CAR T cells. Therefore, innate-like functions can be tuned and leveraged in human T cells.
Collapse
Affiliation(s)
- Nadia Jeremiah
- Institut Curie, Paris Sciences et Lettres Research University, INSERM U932, Paris, France
| | - Hermine Ferran
- Institut Curie, Paris Sciences et Lettres Research University, INSERM U932, Paris, France
| | - Konstantina Antoniadou
- Institut Curie, Paris Sciences et Lettres Research University, INSERM U932, Paris, France
| | - Kevin De Azevedo
- Institut Curie, Paris Sciences et Lettres Research University, INSERM U932, Paris, France
| | - Jovan Nikolic
- Institut Curie, Paris Sciences et Lettres Research University, INSERM U932, Paris, France
| | - Mathieu Maurin
- Institut Curie, Paris Sciences et Lettres Research University, INSERM U932, Paris, France
| | - Philippe Benaroch
- Institut Curie, Paris Sciences et Lettres Research University, INSERM U932, Paris, France
| | - Nicolas Manel
- Institut Curie, Paris Sciences et Lettres Research University, INSERM U932, Paris, France
| |
Collapse
|
227
|
Aluri J, Cooper MA. Somatic mosaicism in inborn errors of immunity: Current knowledge, challenges, and future perspectives. Semin Immunol 2023; 67:101761. [PMID: 37062181 PMCID: PMC11321052 DOI: 10.1016/j.smim.2023.101761] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 04/03/2023] [Accepted: 04/05/2023] [Indexed: 04/18/2023]
Abstract
Inborn errors of immunity (IEI) are a diverse group of monogenic disorders of the immune system due to germline variants in genes important for the immune response. Over the past decade there has been increasing recognition that acquired somatic variants present in a subset of cells can also lead to immune disorders or 'phenocopies' of IEI. Discovery of somatic mosaicism causing IEI has largely arisen from investigation of seemingly sporadic cases of IEI with predominant symptoms of autoinflammation and/or autoimmunity in which germline disease-causing variants are not detected. Disease-causing somatic mosaicism has been identified in genes that also cause germline IEI, such as FAS, and in genes without significant corresponding germline disease, such as UBA1 and TLR8. There are challenges in detecting low-level somatic variants, and it is likely that the extent of the somatic mosaicism causing IEI is largely uncharted. Here we review the field of somatic mosaicism leading to IEI and discuss challenges and methods for somatic variant detection, including diagnostic approaches for molecular diagnoses of patients.
Collapse
Affiliation(s)
- Jahnavi Aluri
- Department of Pediatrics, Division of Rheumatology/Immunology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Megan A Cooper
- Department of Pediatrics, Division of Rheumatology/Immunology, Washington University in St. Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
228
|
Ji Y, Luo Y, Wu Y, Sun Y, Zhao L, Xue Z, Sun M, Wei X, He Z, Wu SA, Lin LL, Lu Y, Chang L, Chen F, Chen S, Qian W, Xu X, Chen S, Pan D, Zhou Z, Xia S, Hu CCA, Liang T, Qi L. SEL1L-HRD1 endoplasmic reticulum-associated degradation controls STING-mediated innate immunity by limiting the size of the activable STING pool. Nat Cell Biol 2023; 25:726-739. [PMID: 37142791 PMCID: PMC10185471 DOI: 10.1038/s41556-023-01138-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 03/17/2023] [Indexed: 05/06/2023]
Abstract
Stimulator of interferon genes (STING) orchestrates the production of proinflammatory cytokines in response to cytosolic double-stranded DNA; however, the pathophysiological significance and molecular mechanism underlying the folding and maturation of nascent STING protein at the endoplasmic reticulum (ER) remain unknown. Here we report that the SEL1L-HRD1 protein complex-the most conserved branch of ER-associated degradation (ERAD)-is a negative regulator of the STING innate immunity by ubiquitinating and targeting nascent STING protein for proteasomal degradation in the basal state. SEL1L or HRD1 deficiency in macrophages specifically amplifies STING signalling and immunity against viral infection and tumour growth. Mechanistically, nascent STING protein is a bona fide substrate of SEL1L-HRD1 in the basal state, uncoupled from ER stress or its sensor inositol-requiring enzyme 1α. Hence, our study not only establishes a key role of SEL1L-HRD1 ERAD in innate immunity by limiting the size of the activable STING pool, but identifies a regulatory mechanism and therapeutic approach to targeting STING.
Collapse
Affiliation(s)
- Yewei Ji
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Yuan Luo
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yating Wu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yao Sun
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lianfeng Zhao
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhen Xue
- Graduate Program in Nutrition, Cornell University, Ithaca, NY, USA
| | - Mengqi Sun
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoqiong Wei
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Zinan He
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shuangcheng Alivia Wu
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Liangguang Leo Lin
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - You Lu
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Lei Chang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fei Chen
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Siyu Chen
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Qian
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Xiaoxi Xu
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Shengnuo Chen
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dongli Pan
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhangsen Zhou
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Sheng Xia
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Chih-Chi Andrew Hu
- Center for Translational Research in Hematologic Malignancies, Houston Methodist Cancer Center, Houston Methodist Research Institute, Houston, TX, USA
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ling Qi
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
229
|
Baris A, Fraile-Bethencourt E, Eubanks J, Khou S, Anand S. Thymidine phosphorylase facilitates retinoic acid inducible gene-I induced endothelial dysfunction. Cell Death Dis 2023; 14:294. [PMID: 37100811 PMCID: PMC10131517 DOI: 10.1038/s41419-023-05821-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 04/13/2023] [Accepted: 04/19/2023] [Indexed: 04/28/2023]
Abstract
Activation of nucleic acid sensors in endothelial cells (ECs) has been shown to drive inflammation across pathologies including cancer, atherosclerosis and obesity. We previously showed that enhancing cytosolic DNA sensing by inhibiting three prime exonuclease 1 (TREX1) in ECs led to EC dysfunction and impaired angiogenesis. Here we show that activation of a cytosolic RNA sensor, Retinoic acid Induced Gene 1 (RIG-I) diminishes EC survival, angiogenesis and triggers tissue specific gene expression programs. We discovered a RIG-I dependent 7 gene signature that affects angiogenesis, inflammation and coagulation. Among these, we identified the thymidine phosphorylase TYMP as a key mediator of RIG-I induced EC dysfunction via its regulation of a subset of interferon stimulated genes. Our RIG-I induced gene signature was also conserved in the context of human diseases - in lung cancer vasculature and herpesvirus infection of lung endothelial cells. Pharmacological or genetic inhibition of TYMP rescues RIG-I induced EC death, migration arrest and restores sprouting angiogenesis. Interestingly, using RNAseq we identified a gene expression program that was RIG-I induced but TYMP dependent. Analysis of this dataset indicated that IRF1 and IRF8 dependent transcription is diminished in RIG-I activated cells when TYMP is inhibited. Functional RNAi screen of our TYMP dependent EC genes, we found that a group of 5 genes - Flot1, Ccl5, Vars2, Samd9l and Ube2l6 are critical for endothelial cell death mediated by RIG-I activation. Our observations identify mechanisms by which RIG-I drives EC dysfunction and define pathways that can be pharmacologically targeted to ameliorate RIG-I induced vascular inflammation.
Collapse
Affiliation(s)
- Adrian Baris
- Department of Cell, Developmental & Cancer Biology, Knight Cancer Institute, 2720 S Moody Avenue, Oregon Health & Science University, Portland, OR, 97201, USA
| | - Eugenia Fraile-Bethencourt
- Department of Cell, Developmental & Cancer Biology, Knight Cancer Institute, 2720 S Moody Avenue, Oregon Health & Science University, Portland, OR, 97201, USA
| | - Jaiden Eubanks
- Department of Cell, Developmental & Cancer Biology, Knight Cancer Institute, 2720 S Moody Avenue, Oregon Health & Science University, Portland, OR, 97201, USA
| | - Sokchea Khou
- Department of Cell, Developmental & Cancer Biology, Knight Cancer Institute, 2720 S Moody Avenue, Oregon Health & Science University, Portland, OR, 97201, USA
| | - Sudarshan Anand
- Department of Cell, Developmental & Cancer Biology, Knight Cancer Institute, 2720 S Moody Avenue, Oregon Health & Science University, Portland, OR, 97201, USA.
- Department of Radiation Medicine, Knight Cancer Institute, 2720 S Moody Avenue, Oregon Health & Science University, Portland, OR, 97201, USA.
| |
Collapse
|
230
|
Ji W, Zhang L, Ma C, Xu X, Li S, Xia H, Zhou W, Liu X. Newly synthesized AIFM1 determines the hypersensitivity of T lymphocytes to STING activation-induced cell apoptosis. Cell Rep 2023; 42:112327. [PMID: 37000625 DOI: 10.1016/j.celrep.2023.112327] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 01/02/2023] [Accepted: 03/17/2023] [Indexed: 04/01/2023] Open
Abstract
STING is a well-known signaling adaptor essential for sensing cytosolic dsDNA to produce type I interferon. Although the detailed underlying mechanisms remain enigmatic, recent studies show that STING activation can lead to T lymphocyte apoptosis. Here, we report that AIFM1 facilitates STING activation-induced cell apoptosis in T lymphocytes. Mechanistically, AIFM1 is upregulated after STING activation in T cells but not in HEK293T-STING and THP-1 cells, rendering T cells more sensitive to apoptosis. In contrast to the canonical role of AIFM1 in the caspase-independent parthanatos, the function of AIFM1 is operated by the formation of an AIFM1/IRF3/BAX complex and mitochondrial outer membrane permeabilization, which cause cytochrome c release and caspase activation. Furthermore, supplementation with newly synthesized AIFM1 can reconstitute STING activation-induced cell apoptosis in HEK293T-STING and THP-1 cells. Our study identifies AIFM1 as a key regulating factor determining the hypersensitivity of T lymphocytes to STING activation-induced cell apoptosis.
Collapse
Affiliation(s)
- Wangsheng Ji
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Science, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China; Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital of Henan University, Henan University, Kaifeng 475000, China
| | - Lianfei Zhang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Science, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Chengxin Ma
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Science, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xiaoyu Xu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Science, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Shuai Li
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Science, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Huan Xia
- Department of Infectious Diseases, Nankai University Second People's Hospital, Tianjin 300071, China
| | - Weihong Zhou
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Science, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xinqi Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Science, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China.
| |
Collapse
|
231
|
Luo J, Lu C, Chen Y, Wu X, Zhu C, Cui W, Yu S, Li N, Pan Y, Zhao W, Yang Q, Yang X. Nuclear translocation of cGAS orchestrates VEGF-A-mediated angiogenesis. Cell Rep 2023; 42:112328. [PMID: 37027305 DOI: 10.1016/j.celrep.2023.112328] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 12/20/2022] [Accepted: 03/17/2023] [Indexed: 04/08/2023] Open
Abstract
Cyclic GMP-AMP synthase (cGAS) senses cytosolic incoming DNA and consequently activates stimulator of interferon response cGAMP interactor 1 (STING) to mount immune response. Here, we show nuclear cGAS could regulate VEGF-A-mediated angiogenesis in an immune-independent manner. We found VEGF-A stimulation induces cGAS nuclear translocation via importin-β pathway. Moreover, nuclear cGAS subsequently regulates miR-212-5p-ARPC3 cascade to modulate VEGF-A-mediated angiogenesis through affecting cytoskeletal dynamics and VEGFR2 trafficking from trans-Golgi network (TGN) to plasma membrane via a regulatory feedback loop. In contrast, cGAS deficiency remarkably impairs VEGF-A-mediated angiogenesis in vivo and in vitro. Furthermore, we found strong association between the expression of nuclear cGAS and VEGF-A, and the malignancy and prognosis in malignant glioma, suggesting that nuclear cGAS might play important roles in human pathology. Collectively, our findings illustrated the function of cGAS in angiogenesis other than immune surveillance, which might be a potential therapeutic target for pathological angiogenesis-related diseases.
Collapse
Affiliation(s)
- Juanjuan Luo
- Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Chunjiao Lu
- Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Yang Chen
- Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Xuewei Wu
- Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Chenchen Zhu
- Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Wei Cui
- College of Life Science and Biopharmaceutical of Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Shicang Yu
- Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Ningning Li
- The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Yihang Pan
- The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, China
| | - Weijiang Zhao
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Qingkai Yang
- Institute of Cancer Stem Cell, Dalian Medical University, Dalian, Liaoning 116044, China.
| | - Xiaojun Yang
- Shantou University Medical College, Shantou, Guangdong 515041, China.
| |
Collapse
|
232
|
Kong L, Sui C, Chen T, Zhang L, Zhao W, Zheng Y, Liu B, Cheng X, Gao C. The ubiquitin E3 ligase TRIM10 promotes STING aggregation and activation in the Golgi apparatus. Cell Rep 2023; 42:112306. [PMID: 36972172 DOI: 10.1016/j.celrep.2023.112306] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 01/30/2023] [Accepted: 03/08/2023] [Indexed: 03/28/2023] Open
Abstract
STING is an endoplasmic reticulum-resident protein regulating innate immunity. After binding with cyclic guanosine monophosphate-AMP (cGAMP), STING translocates from the endoplasmic reticulum (ER) to the Golgi apparatus to stimulate TBK1 and IRF3 activation, leading to expression of type I interferon. However, the exact mechanism concerning STING activation remains largely enigmatic. Here, we identify tripartite motif 10 (TRIM10) as a positive regulator of STING signaling. TRIM10-deficient macrophages exhibit reduced type I interferon production upon double-stranded DNA (dsDNA) or cGAMP stimulation and decreased resistance to herpes simplex virus 1 (HSV-1) infection. Additionally, TRIM10-deficient mice are more susceptible to HSV-1 infection and exhibit faster melanoma growth. Mechanistically, TRIM10 associates with STING and catalyzes K27- and K29-linked polyubiquitination of STING at K289 and K370, which promotes STING trafficking from the ER to the Golgi apparatus, formation of STING aggregates, and recruitment of TBK1 to STING, ultimately enhancing the STING-dependent type I interferon response. Our study defines TRIM10 as a critical activator in cGAS-STING-mediated antiviral and antitumor immunity.
Collapse
Affiliation(s)
- Lingli Kong
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong 250012, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Chao Sui
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong 250012, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Tian Chen
- Department of Pathogenic Biology, School of Biomedical Sciences, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Lei Zhang
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong 250012, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Wei Zhao
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong 250012, P.R. China; Department of Pathogenic Biology, School of Biomedical Sciences, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yi Zheng
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong 250012, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Bingyu Liu
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong 250012, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xiaochen Cheng
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong 250012, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong 250012, P.R. China.
| | - Chengjiang Gao
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong 250012, P.R. China; Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong 250012, P.R. China.
| |
Collapse
|
233
|
Kanazawa N, Ishii T, Takita Y, Nishikawa A, Nishikomori R. Efficacy and safety of baricitinib in Japanese patients with autoinflammatory type I interferonopathies (NNS/CANDLE, SAVI, And AGS). Pediatr Rheumatol Online J 2023; 21:38. [PMID: 37087470 PMCID: PMC10122451 DOI: 10.1186/s12969-023-00817-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 04/06/2023] [Indexed: 04/24/2023] Open
Abstract
BACKGROUND This study evaluated the efficacy and safety of baricitinib (Janus kinase-1/2 inhibitor), in adult and pediatric Japanese patients with Nakajo-Nishimura syndrome/chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature (NNS/CANDLE), stimulator of interferon genes-associated vasculopathy with onset during infancy (SAVI), or Aicardi-Goutières syndrome (AGS). METHODS A Phase 2/3, multicenter, open-label study (NCT04517253) was conducted across 52 weeks. Primary efficacy endpoint assessed the change in mean daily diary score (DDS) from baseline to the end of primary treatment period. Other efficacy endpoints included change in mean DDS to the end of maintenance period, daily corticosteroid use, Physician's Global Assessment of Disease Activity (PGA) scores, and daily symptom-specific score (DSSS) from baseline to primary and maintenance treatment periods. All treatment-emergent adverse events (TEAEs) that occurred postdosing were recorded. RESULTS Overall, 9 patients (5 with NNS, 3 with SAVI, and 1 with AGS) were enrolled; 55.6% were females, mean age was 26 years, and mean corticosteroid use/weight was 0.2 mg/kg. At the end of primary treatment period, mean DDS decreased from baseline in patients with NNS/CANDLE (0.22) and SAVI (0.21) and increased in the patient with AGS (0.07). At the end of maintenance treatment period, mean DDS decreased from baseline in patients with NNS/CANDLE (0.18) and SAVI (0.27) and increased in the patient with AGS (0.04). Mean percent corticosteroid use decreased by 18.4% in 3 out of 5 patients with NNS/CANDLE and 62.9% in 1 out of 3 patients with SAVI. Mean PGA score decreased from baseline in patients with NNS/CANDLE (1.60), SAVI (1.33), and AGS (1.0), and mean DSSS improved from baseline. All patients reported ≥ 1 TEAE. Frequently reported AEs included BK polyomavirus detection (3; 33.3%), increased blood creatine phosphokinase (2; 22.2%), anemia (2; 22.2%), and upper respiratory tract infection (2; 22.2%). Three (33.3%) patients reported serious adverse events, 1 of which was related to study drug. One patient with SAVI died due to intracranial hemorrhage, which was not related to study drug. CONCLUSION Baricitinib may offer a potential therapeutic option for patients with NNS/CANDLE, SAVI, and AGS, with a positive benefit/risk profile in a vulnerable patient population with multiple comorbidities. TRIAL REGISTRATION NLM clinicaltrials.gov, NCT04517253 . Registered 18 August 2020.
Collapse
Affiliation(s)
- Nobuo Kanazawa
- Department of Dermatology, Hyogo Medical University, Nishinomiya, Japan
| | - Taeko Ishii
- Eli Lilly Japan K.K, Lilly Plaza One Bldg., 5-1-28, Isogamidori, Chuo-ku, Kobe, 651-0086, Japan.
| | - Yasushi Takita
- Eli Lilly Japan K.K, Lilly Plaza One Bldg., 5-1-28, Isogamidori, Chuo-ku, Kobe, 651-0086, Japan
| | - Atsushi Nishikawa
- Eli Lilly Japan K.K, Lilly Plaza One Bldg., 5-1-28, Isogamidori, Chuo-ku, Kobe, 651-0086, Japan
| | - Ryuta Nishikomori
- Department of Pediatrics and Child Health, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
234
|
Liu S, Yang B, Hou Y, Cui K, Yang X, Li X, Chen L, Liu S, Zhang Z, Jia Y, Xie Y, Xue Y, Li X, Yan B, Wu C, Deng W, Qi J, Lu D, Gao GF, Wang P, Shang G. The mechanism of STING autoinhibition and activation. Mol Cell 2023; 83:1502-1518.e10. [PMID: 37086726 DOI: 10.1016/j.molcel.2023.03.029] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 10/21/2022] [Accepted: 03/30/2023] [Indexed: 04/24/2023]
Abstract
2',3'-cGAMP, produced by the DNA sensor cGAS, activates stimulator of interferon genes (STING) and triggers immune response during infection. Tremendous effort has been placed on unraveling the mechanism of STING activation. However, little is known about STING inhibition. Here, we found that apo-STING exhibits a bilayer with head-to-head as well as side-by-side packing, mediated by its ligand-binding domain (LBD). This type of assembly holds two endoplasmic reticulum (ER) membranes together not only to prevent STING ER exit but also to eliminate the recruitment of TBK1, representing the autoinhibited state of STING. Additionally, we obtained the filament structure of the STING/2',3'-cGAMP complex, which adopts a bent monolayer assembly mediated by LBD and transmembrane domain (TMD). The active, curved STING polymer could deform ER membrane to support its ER exit and anterograde transportation. Our data together provide a panoramic vision regarding STING autoinhibition and activation, which adds substantially to current understanding of the cGAS-STING pathway.
Collapse
Affiliation(s)
- Sheng Liu
- Shanxi Provincial Key Laboratory of Protein Structure Determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan 030012, China; CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; Cryo-EM Center, Southern University of Science and Technology, Shenzhen 518055, China
| | - Bo Yang
- Shanxi Provincial Key Laboratory of Protein Structure Determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan 030012, China; College of Life Sciences, Shanxi Agricultural University, Taiyuan 030031, China; Shanxi Provincial Key Laboratory for Major Infectious Disease Response, Taiyuan 030012, China
| | - Yingxiang Hou
- Shanxi Provincial Key Laboratory of Protein Structure Determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan 030012, China; The Key Laboratory of Medical Molecular Cell Biology of Shanxi Province, Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
| | - Kaige Cui
- Shanxi Provincial Key Laboratory of Protein Structure Determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan 030012, China; Shanxi Provincial Key Laboratory for Major Infectious Disease Response, Taiyuan 030012, China
| | - Xiaozhu Yang
- Shanxi Provincial Key Laboratory of Protein Structure Determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan 030012, China; Shanxi Provincial Key Laboratory for Major Infectious Disease Response, Taiyuan 030012, China
| | - Xiaoxiong Li
- Shanxi Provincial Key Laboratory of Protein Structure Determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan 030012, China; Shanxi Provincial Key Laboratory for Major Infectious Disease Response, Taiyuan 030012, China
| | - Lianwan Chen
- National Key Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Shichao Liu
- Shanxi Provincial Key Laboratory of Protein Structure Determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan 030012, China; Shanxi Provincial Key Laboratory for Major Infectious Disease Response, Taiyuan 030012, China
| | - Zhichao Zhang
- Shanxi Provincial Key Laboratory of Protein Structure Determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan 030012, China; Shanxi Provincial Key Laboratory for Major Infectious Disease Response, Taiyuan 030012, China
| | - Yuanyuan Jia
- Shanxi Provincial Key Laboratory of Protein Structure Determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan 030012, China; Shanxi Provincial Key Laboratory for Major Infectious Disease Response, Taiyuan 030012, China
| | - Yufeng Xie
- Shanxi Provincial Key Laboratory of Protein Structure Determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan 030012, China; CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Ying Xue
- Shanxi Provincial Key Laboratory of Protein Structure Determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan 030012, China; Shanxi Provincial Key Laboratory for Major Infectious Disease Response, Taiyuan 030012, China
| | - Xiaomei Li
- Shanxi Provincial Key Laboratory of Protein Structure Determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan 030012, China
| | - Bingxue Yan
- Shanxi Provincial Key Laboratory of Protein Structure Determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan 030012, China
| | - Changxin Wu
- The Key Laboratory of Medical Molecular Cell Biology of Shanxi Province, Institutes of Biomedical Sciences, Shanxi University, Taiyuan 030006, China
| | - Wen Deng
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Jianxun Qi
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Life Science Academy, Beijing 102209, China
| | - Defen Lu
- Shanxi Provincial Key Laboratory of Protein Structure Determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan 030012, China; College of Life Sciences, Shanxi Agricultural University, Taiyuan 030031, China.
| | - George F Gao
- Shanxi Provincial Key Laboratory of Protein Structure Determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan 030012, China; CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Peiyi Wang
- Cryo-EM Center, Southern University of Science and Technology, Shenzhen 518055, China.
| | - Guijun Shang
- Shanxi Provincial Key Laboratory of Protein Structure Determination, Shanxi Academy of Advanced Research and Innovation, Taiyuan 030012, China; College of Life Sciences, Shanxi Agricultural University, Taiyuan 030031, China; Shanxi Provincial Key Laboratory for Major Infectious Disease Response, Taiyuan 030012, China.
| |
Collapse
|
235
|
Fu L, Lu K, Jiao Q, Chen X, Jia F. The Regulation and Double-Edged Roles of the Deubiquitinase OTUD5. Cells 2023; 12:cells12081161. [PMID: 37190070 DOI: 10.3390/cells12081161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/09/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
OTUD5 (OTU Deubiquitinase 5) is a functional cysteine protease with deubiquitinase activity and is a member of the ovarian tumor protease (OTU) family. OTUD5 is involved in the deubiquitination of many key proteins in various cellular signaling pathways and plays an important role in maintaining normal human development and physiological functions. Its dysfunction can affect physiological processes, such as immunity and DNA damage repair, and it can even lead to tumors, inflammatory diseases and genetic disorders. Therefore, the regulation of OTUD5 activity and expression has become a hot topic of research. A comprehensive understanding of the regulatory mechanisms of OTUD5 and its use as a therapeutic target for diseases is of great value. Herein, we review the physiological processes and molecular mechanisms of OTUD5 regulation, outline the specific regulatory processes of OTUD5 activity and expression, and link OTUD5 to diseases from the perspective of studies on signaling pathways, molecular interactions, DNA damage repair and immune regulation, thus providing a theoretical basis for future studies.
Collapse
Affiliation(s)
- Lin Fu
- School of Basic Medicine, Qingdao University, Qingdao 266072, China
| | - Kun Lu
- School of Basic Medicine, Qingdao University, Qingdao 266072, China
| | - Qian Jiao
- School of Basic Medicine, Qingdao University, Qingdao 266072, China
| | - Xi Chen
- School of Basic Medicine, Qingdao University, Qingdao 266072, China
| | - Fengju Jia
- School of Nursing, Qingdao University, Qingdao 266072, China
| |
Collapse
|
236
|
He X, Sun Y, Lu J, Naz F, Ma S, Liu J. Cytoplasmic DNAs: Sources, sensing, and roles in the development of lung inflammatory diseases and cancer. Front Immunol 2023; 14:1117760. [PMID: 37122745 PMCID: PMC10130589 DOI: 10.3389/fimmu.2023.1117760] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/29/2023] [Indexed: 05/02/2023] Open
Abstract
Cytoplasmic DNA is emerging as a pivotal contributor to the pathogenesis of inflammatory diseases and cancer, such as COVID-19 and lung carcinoma. However, the complexity of various cytoplasmic DNA-related pathways and their crosstalk remains challenging to distinguish their specific roles in many distinct inflammatory diseases, especially for the underlying mechanisms. Here, we reviewed the latest findings on cytoplasmic DNA and its signaling pathways in inflammatory lung conditions and lung cancer progression. We found that sustained activation of cytoplasmic DNA sensing pathways contributes to the development of common lung diseases, which may result from external factors or mutations of key genes in the organism. We further discussed the interplays between cytoplasmic DNA and anti-inflammatory or anti-tumor effects for potential immunotherapy. In sum, this review aids in understanding the roles of cytoplasmic DNAs and exploring more therapeutic strategies.
Collapse
Affiliation(s)
- Xintong He
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining, China
- College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, United Kingdom
| | - Ye Sun
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining, China
- College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, United Kingdom
| | - Jianzhang Lu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining, China
- College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, United Kingdom
| | - Faiza Naz
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining, China
| | - Shenglin Ma
- Hangzhou Cancer Institution, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Jian Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining, China
- College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, United Kingdom
- Cancer Center, Zhejiang University, Hangzhou, China
- Biomedical and Heath Translational Research Center of Zhejiang Province, Haining, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
237
|
MacLauchlan S, Kushwaha P, Tai A, Chen J, Manning C, Swarnkar G, Abu-Amer Y, Fitzgerald KA, Sharma S, Gravallese EM. STING-dependent interferon signatures restrict osteoclast differentiation and bone loss in mice. Proc Natl Acad Sci U S A 2023; 120:e2210409120. [PMID: 37023130 PMCID: PMC10104545 DOI: 10.1073/pnas.2210409120] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 02/14/2023] [Indexed: 04/07/2023] Open
Abstract
Stimulator of interferon genes (STING) is a key mediator of type-I interferon (IFN-I) signaling in response to a variety of stimuli, but the contribution of STING to homeostatic processes is not fully characterized. Previous studies showed that ligand activation of STING limits osteoclast differentiation in vitro through the induction of IFNβ and IFN-I interferon-stimulated genes (ISGs). In a disease model (SAVI) driven by the V154M gain-of-function mutation in STING, fewer osteoclasts form from SAVI precursors in response to receptor activator of NF-kappaB ligand (RANKL) in an IFN-I-dependent manner. Due to the described role of STING-mediated regulation of osteoclastogenesis in activation settings, we sought to determine whether basal STING signaling contributes to bone homeostasis, an unexplored area. Using whole-body and myeloid-specific deficiency, we show that STING signaling prevents trabecular bone loss in mice over time and that myeloid-restricted STING activity is sufficient for this effect. STING-deficient osteoclast precursors differentiate with greater efficiency than wild types. RNA sequencing of wild-type and STING-deficient osteoclast precursor cells and differentiating osteoclasts reveals unique clusters of ISGs including a previously undescribed ISG set expressed in RANKL naïve precursors (tonic expression) and down-regulated during differentiation. We identify a 50 gene tonic ISG signature that is STING dependent and shapes osteoclast differentiation. From this list, we identify interferon-stimulated gene 15 (ISG15) as a tonic STING-regulated ISG that limits osteoclast formation. Thus, STING is an important upstream regulator of tonic IFN-I signatures shaping the commitment to osteoclast fates, providing evidence for a nuanced and unique role for this pathway in bone homeostasis.
Collapse
Affiliation(s)
- Susan MacLauchlan
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Priyanka Kushwaha
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Albert Tai
- Department of Immunology, Tufts University School of Medicine, Boston, MA02111
| | - Jia Chen
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Catherine Manning
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Gaurav Swarnkar
- Department of Orthopedics and Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO63110
| | - Yousef Abu-Amer
- Department of Orthopedics and Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, MO63110
| | - Katherine A. Fitzgerald
- Department of Medicine, Program in Innate Immunity, University of Massachusetts Chan Medical School, Worcester, MA01655
| | - Shruti Sharma
- Department of Immunology, Tufts University School of Medicine, Boston, MA02111
| | - Ellen M. Gravallese
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA02115
| |
Collapse
|
238
|
Hu B, Ma JX, Duerfeldt AS. The cGAS-STING pathway in diabetic retinopathy and age-related macular degeneration. Future Med Chem 2023; 15:717-729. [PMID: 37166075 PMCID: PMC10194038 DOI: 10.4155/fmc-2022-0301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 03/28/2023] [Indexed: 05/12/2023] Open
Abstract
Diabetic retinopathy and age-related macular degeneration are common retinal diseases with shared pathophysiology, including oxidative stress-induced inflammation. Cellular mechanisms responsible for converting oxidative stress into retinal damage are ill-defined but have begun to clarify. One common outcome of retinal oxidative stress is mitochondrial damage and subsequent release of mitochondrial DNA into the cytosol. This leads to activation of the cGAS-STING pathway, resulting in interferon release and disease-amplifying inflammation. This review summarizes the evolving link between aberrant cGAS-STING signaling and inflammation in common retinal diseases and provides prospective for targeting this system in diabetic retinopathy and age-related macular degeneration. Further defining the roles of this system in the retina is expected to reveal new disease pathology and novel therapeutic approaches.
Collapse
Affiliation(s)
- Bo Hu
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, MN 55414, USA
| | - Jian-Xing Ma
- Department of Biochemistry, Wake Forest University School of Medicine, Winston Salem, NC 27101, USA
| | - Adam S Duerfeldt
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, MN 55414, USA
| |
Collapse
|
239
|
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
|
240
|
Vornholz L, Isay SE, Kurgyis Z, Strobl DC, Loll P, Mosa MH, Luecken MD, Sterr M, Lickert H, Winter C, Greten FR, Farin HF, Theis FJ, Ruland J. Synthetic enforcement of STING signaling in cancer cells appropriates the immune microenvironment for checkpoint inhibitor therapy. SCIENCE ADVANCES 2023; 9:eadd8564. [PMID: 36921054 PMCID: PMC10017047 DOI: 10.1126/sciadv.add8564] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 01/26/2023] [Indexed: 06/18/2023]
Abstract
Immune checkpoint inhibitors (ICIs) enhance anticancer immunity by releasing repressive signals into tumor microenvironments (TMEs). To be effective, ICIs require preexisting immunologically "hot" niches for tumor antigen presentation and lymphocyte recruitment. How the mutational landscape of cancer cells shapes these immunological niches remains poorly defined. We found in human and murine colorectal cancer (CRC) models that the superior antitumor immune response of mismatch repair (MMR)-deficient CRC required tumor cell-intrinsic activation of cGAS-STING signaling triggered by genomic instability. Subsequently, we synthetically enforced STING signaling in CRC cells with intact MMR signaling using constitutively active STING variants. Even in MMR-proficient CRC, genetically encoded gain-of-function STING was sufficient to induce cancer cell-intrinsic interferon signaling, local activation of antigen-presenting cells, recruitment of effector lymphocytes, and sensitization of previously "cold" TMEs to ICI therapy in vivo. Thus, our results introduce a rational strategy for modulating cancer cell-intrinsic programs via engineered STING enforcement to sensitize resistant tumors to ICI responsiveness.
Collapse
Affiliation(s)
- Larsen Vornholz
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany
- TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Sophie E. Isay
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany
- TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Zsuzsanna Kurgyis
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany
- TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
- Department of Dermatology and Allergology, Technical University of Munich, Munich, Germany
| | - Daniel C. Strobl
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany
- TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
- Institute of Computational Biology, Department of Computational Health, Helmholtz Center Munich, Neuherberg, Germany
| | - Patricia Loll
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany
- TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
| | - Mohammed H. Mosa
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt/Main, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt/Main, Germany
| | - Malte D. Luecken
- Institute of Computational Biology, Department of Computational Health, Helmholtz Center Munich, Neuherberg, Germany
- Institute of Lung Health and Immunity (LHI), Helmholtz Center Munich, Neuherberg, Germany
- Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Michael Sterr
- Institute of Diabetes and Regeneration Research, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Stem Cell Research, Helmholtz Center Munich, Neuherberg, Germany
- School of Medicine, Technical University of Munich, Munich, Germany
| | - Christof Winter
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany
- TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Munich partner site, Germany
- German Cancer Consortium (DKTK), Frankfurt/Mainz partner site, Germany
| | - Florian R. Greten
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt/Main, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt/Main, Germany
- German Cancer Consortium (DKTK), Frankfurt/Mainz partner site, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Henner F. Farin
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt/Main, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt/Main, Germany
- German Cancer Consortium (DKTK), Frankfurt/Mainz partner site, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Fabian J. Theis
- Institute of Computational Biology, Department of Computational Health, Helmholtz Center Munich, Neuherberg, Germany
- Department of Mathematics, Technical University of Munich, Munich, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany
| | - Jürgen Ruland
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany
- TranslaTUM, Center for Translational Cancer Research, Technical University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Munich partner site, Germany
- German Cancer Consortium (DKTK), Frankfurt/Mainz partner site, Germany
- German Center for Infection Research (DZIF), Munich partner site, Germany
| |
Collapse
|
241
|
Zang R, Xue L, Zhang M, Peng X, Li X, Du K, Shi C, Liu Y, Lin Y, Han W, Yu R, Wang Q, Yang J, Wang X, Jiang T. Design and syntheses of a bimolecular STING agonist based on the covalent STING antagonist. Eur J Med Chem 2023; 250:115184. [PMID: 36758305 DOI: 10.1016/j.ejmech.2023.115184] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/23/2023] [Accepted: 02/02/2023] [Indexed: 02/06/2023]
Abstract
Cyclic GMP-AMP synthase and stimulator of interferon genes (cGAS-STING) signaling stimulators, an essential innate immunity component, monitor invading pathogen DNA and damaged self-DNA, making them an appealing target for drug development. The natural STING agonist, 2'3'-cGAMP, mounts and stabilizes the STING homodimer to trigger an antiviral or antitumor immune responses. However, cyclic-dinucleotide-based STING agonists show limited clinical effects owing to their short half-lives. To explore whether STING-dimer stabilizers could trigger STING signaling instead of cyclic dinucleotide-based molecules, we analyzed the structural characteristics of STING to design and synthesize a series of compounds based on the covalent STING inhibitor C-170, three of which were 23, 26, and 27, exhibited STING-dependent immune activation, both in vitro and in vivo. Compound 23 could act synergistically with cGAMP and other STING agonists as a promising moderate STING agonist. This indicates that promoting STING dimerization is a promising strategy for designing next-generation STING agonists.
Collapse
Affiliation(s)
- Ruochen Zang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Department of Clinical Laboratory, Qilu Hospital of Shandong University (Qingdao), Qingdao, 266100, China
| | - Liang Xue
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Drugs and Bioproducts and Innovation Center for Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266003, China
| | - Meifang Zhang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Xiaoyue Peng
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Xionghao Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Kaixin Du
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Chuanqin Shi
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Center of Translational Medicine, ZiBo Central Hospital, Zibo, 255036, China
| | - Yuqian Liu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Yuxi Lin
- Institute of Cancer Biology and Drug Screening, School of Life Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Wenwei Han
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, 266071, China
| | - Rilei Yu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
| | - Qian Wang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University (Qingdao), Qingdao, 266100, China; Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Jinbo Yang
- Marine Drug Screening and Evaluation Platform, Qingdao National Laboratory for Marine Science and Technology, Ocean University of China, Qingdao, 266071, China
| | - Xin Wang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Marine Drug Screening and Evaluation Platform, Qingdao National Laboratory for Marine Science and Technology, Ocean University of China, Qingdao, 266071, China.
| | - Tao Jiang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Drugs and Bioproducts and Innovation Center for Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266003, China.
| |
Collapse
|
242
|
Fang R, Jiang Q, Jia X, Jiang Z. ARMH3-mediated recruitment of PI4KB directs Golgi-to-endosome trafficking and activation of the antiviral effector STING. Immunity 2023; 56:500-515.e6. [PMID: 36921576 DOI: 10.1016/j.immuni.2023.02.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/30/2022] [Accepted: 01/26/2023] [Indexed: 03/15/2023]
Abstract
The cGAS-STING pathway mediates cytoplasmic DNA-triggered innate immunity. STING activation is initiated by cyclic-GMP-AMP (cGAMP)-induced translocation from the endoplasmic reticulum and sulfated glycosaminoglycans-induced polymerization at the Golgi. Here, we examine the mechanisms underlying STING transport and activation beyond the Golgi. A genome-wide CRISPR-Cas9 screen identified Armadillo-like helical domain-containing protein 3 (ARMH3) as critical for STING activation. Upon cGAMP-triggered translocation, ARMH3 interacted with STING at the Golgi and recruited phosphatidylinositol 4-kinase beta (PI4KB) to synthesize PI4P, which directed STING Golgi-to-endosome trafficking via PI4P-binding proteins AP-1 and GGA2. Disrupting PI4P-dependent lipid transport through RNAi of other PI4P-binding proteins impaired STING activation. Consistently, disturbed lipid composition inhibited STING activation, whereas aberrantly elevated cellular PI4P led to cGAS-independent STING activation. Armh3fl/fllLyzCre/Cre mice were susceptible to DNA virus challenge in vivo. Thus, ARMH3 bridges STING and PIK4B to generate PI4P for STING transportation and activation, an interaction conserved in all eukaryotes.
Collapse
Affiliation(s)
- Run Fang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Qifei Jiang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Xinying Jia
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Zhengfan Jiang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China.
| |
Collapse
|
243
|
Wang X, Zhang H, Wang Y, Bramasole L, Guo K, Mourtada F, Meul T, Hu Q, Viteri V, Kammerl I, Konigshoff M, Lehmann M, Magg T, Hauck F, Fernandez IE, Meiners S. DNA sensing via the cGAS/STING pathway activates the immunoproteasome and adaptive T-cell immunity. EMBO J 2023; 42:e110597. [PMID: 36912165 PMCID: PMC10106989 DOI: 10.15252/embj.2022110597] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/09/2023] [Accepted: 02/20/2023] [Indexed: 03/14/2023] Open
Abstract
The immunoproteasome is a specialized type of proteasome involved in MHC class I antigen presentation, antiviral adaptive immunity, autoimmunity, and is also part of a broader response to stress. Whether the immunoproteasome is regulated by DNA stress, however, is not known. We here demonstrate that mitochondrial DNA stress upregulates the immunoproteasome and MHC class I antigen presentation pathway via cGAS/STING/type I interferon signaling resulting in cell autonomous activation of CD8+ T cells. The cGAS/STING-induced adaptive immune response is also observed in response to genomic DNA and is conserved in epithelial and mesenchymal cells of mice and men. In patients with idiopathic pulmonary fibrosis, chronic activation of the cGAS/STING-induced adaptive immune response in aberrant lung epithelial cells concurs with CD8+ T-cell activation in diseased lungs. Genetic depletion of the immunoproteasome and specific immunoproteasome inhibitors counteract DNA stress induced cytotoxic CD8+ T-cell activation. Our data thus unravel cytoplasmic DNA sensing via the cGAS/STING pathway as an activator of the immunoproteasome and CD8+ T cells. This represents a novel potential pathomechanism for pulmonary fibrosis that opens new therapeutic perspectives.
Collapse
Affiliation(s)
- Xinyuan Wang
- Comprehensive Pneumology Center (CPC), Member of the German Center for Lung Research (DZL), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Munich, Germany.,State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Huabin Zhang
- Neurosurgical Research, Department of Neurosurgery, University Hospital and Walter-Brendel-Centre of Experimental Medicine, Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany.,The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuqin Wang
- Research Center Borstel/Leibniz Lung Center, Borstel, Germany.,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Borstel, Germany.,Institute of Experimental Medicine, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Laylan Bramasole
- Research Center Borstel/Leibniz Lung Center, Borstel, Germany.,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Borstel, Germany.,Institute of Experimental Medicine, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Kai Guo
- Research Center Borstel/Leibniz Lung Center, Borstel, Germany.,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Borstel, Germany.,Institute of Experimental Medicine, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Fatima Mourtada
- Research Center Borstel/Leibniz Lung Center, Borstel, Germany.,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Borstel, Germany.,Institute of Experimental Medicine, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Thomas Meul
- Comprehensive Pneumology Center (CPC), Member of the German Center for Lung Research (DZL), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Munich, Germany
| | - Qianjiang Hu
- Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), University Hospital Grosshadern, Ludwig-Maximilians-University, Munich, Germany
| | - Valeria Viteri
- Comprehensive Pneumology Center (CPC), Member of the German Center for Lung Research (DZL), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Munich, Germany
| | - Ilona Kammerl
- Comprehensive Pneumology Center (CPC), Member of the German Center for Lung Research (DZL), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Munich, Germany
| | - Melanie Konigshoff
- Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), University Hospital Grosshadern, Ludwig-Maximilians-University, Munich, Germany.,Division of Pulmonary, Allergy and Critical Care Medicine, School of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mareike Lehmann
- Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), University Hospital Grosshadern, Ludwig-Maximilians-University, Munich, Germany
| | - Thomas Magg
- Division of Pediatric Immunology and Rheumatology, Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Fabian Hauck
- Division of Pediatric Immunology and Rheumatology, Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Isis E Fernandez
- Comprehensive Pneumology Center (CPC), Member of the German Center for Lung Research (DZL), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Munich, Germany.,Department of Medicine V, University Hospital, LMU Munich, Munich, Germany
| | - Silke Meiners
- Comprehensive Pneumology Center (CPC), Member of the German Center for Lung Research (DZL), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Munich, Germany.,Research Center Borstel/Leibniz Lung Center, Borstel, Germany.,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Borstel, Germany.,Institute of Experimental Medicine, Christian-Albrechts-University Kiel, Kiel, Germany
| |
Collapse
|
244
|
Dermatologic Manifestations of Noninflammasome-Mediated Autoinflammatory Diseases. JID INNOVATIONS 2023; 3:100176. [PMID: 36876221 PMCID: PMC9982332 DOI: 10.1016/j.xjidi.2022.100176] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/14/2022] [Accepted: 11/21/2022] [Indexed: 12/23/2022] Open
Abstract
Autoinflammatory diseases (AIDs) arise from disturbances that alter interactions of immune cells and tissues. They give rise to prominent (auto)inflammation in the absence of aberrant autoantibodies and/or autoreactive T cells. AIDs that are predominantly caused by changes in the inflammasome pathways, such as the NLRP3- or pyrin-associated inflammasome, have gained substantial attention over the last years. However, AIDs resulting primarily from other changes in the defense system of the innate immune system are less well-studied. These noninflammasome-mediated AIDs relate to, for example, disturbance in the TNF or IFN signaling pathways or aberrations in genes affecting the IL-1RA. The spectrum of clinical signs and symptoms of these conditions is vast. Thus, recognizing early cutaneous signs constitutes an important step in differential diagnoses for dermatologists and other physicians. This review provides an overview of the pathogenesis, clinical presentation, and available treatment options highlighting dermatologic aspects of noninflammasome-mediated AIDs.
Collapse
Key Words
- AID, autoinflammatory disease
- ANCA, antineutrophil cytoplasmic antibody
- AOSD, adult-onset Still disease
- BASDAI, Bath Ankylosing Spondylitis Activity Index
- CANDLE, chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature
- CAPS, cryopyrin-associated periodic syndrome
- CRD, cysteine-rich domain
- DIRA, deficiency of IL-1RA
- DITRA, deficiency of IL-36RA
- ER, endoplasmic reticulum
- ESR, erythrocyte sedimentation rate
- FMF, familial Mediterranean fever
- M-CSF, macrophage colony-stimulating factor
- MAS, macrophage activation syndrome
- NET, neutrophil extracellular trap
- NOS, nitrous oxide
- NSAID, nonsteroidal anti-inflammatory drug
- NUD, neutrophilic urticarial dermatosis
- PFAPA, periodic fever, aphthous stomatitis, pharyngitis, and adenitis
- PKR, protein kinase R
- PRAAS, proteosome-associated autoinflammatory disease
- SAPHO, synovitis, acne, pustulosis, hyperostosis, osteitis syndrome
- SAVI, STING-associated vasculopathy with onset in infancy
- STAT, signal transducer and activator of transcription
- SchS, Schnitzler syndrome
- TNFR, TNF receptor
- TRAPS, TNF receptor‒associated autoinflammatory disease
- Th17, T helper 17
- VAS, Visual Analog Scale
- sTNFR, soluble TNF receptor
Collapse
|
245
|
Tcheurekdjian N, Tenbelian N, Isada CM, Honda K, Treisman G, Tcheurekdjian H. Unique ulcerative undefined autoinflammatory disease mistaken for factitious disorder. Ann Allergy Asthma Immunol 2023; 130:359-362. [PMID: 36586584 DOI: 10.1016/j.anai.2022.12.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/16/2022] [Accepted: 12/21/2022] [Indexed: 12/31/2022]
Affiliation(s)
| | | | - Carlos M Isada
- Department of Infectious Disease, Cleveland Clinic, Cleveland, Ohio
| | - Kord Honda
- Departments of Dermatology and Pathology, Case Western Reserve University, Cleveland, Ohio
| | - Glenn Treisman
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, Maryland
| | - Haig Tcheurekdjian
- Allergy/Immunology Associates, Inc, Cleveland, Ohio; Departments of Medicine and Pediatrics, Case Western Reserve University, Cleveland, Ohio.
| |
Collapse
|
246
|
Miner JJ, Fitzgerald KA. A path towards personalized medicine for autoinflammatory and related diseases. Nat Rev Rheumatol 2023; 19:182-189. [PMID: 36750685 PMCID: PMC9904876 DOI: 10.1038/s41584-022-00904-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2022] [Indexed: 02/09/2023]
Abstract
The human genome project led to the advancement of genetic technologies and genomic medicine for a variety of human diseases, including monogenic autoimmune and autoinflammatory diseases. As a result, the genome of an individual can now be rapidly sequenced at a low cost, and this technology is beginning to change the practice of rheumatology. In this Perspective, we describe how new sequencing technologies combined with careful clinical phenotyping have led to the discovery of rare rheumatic diseases and their corresponding disease-causing mutations. Additionally, we explore ways in which single-gene mutations, including somatic mutations, are creating opportunities to develop personalized medicines. To illustrate this idea, we focus on diseases affecting the TREX1-cGAS-STING pathway, which is associated with monogenic autoinflammatory diseases and vasculopathies. For many of the affected patients and families, there is an urgent, unmet need for the development of personalized therapies. New innovations related to small molecular inhibitors and gene therapies have the potential to benefit these families, and might help drive further innovations that could prove useful for patients with more common forms of autoimmunity and autoinflammation.
Collapse
Affiliation(s)
- Jonathan J Miner
- Departments of Medicine and Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Katherine A Fitzgerald
- Program in Innate Immunity, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
247
|
Ramanathan S, Brilot F, Irani SR, Dale RC. Origins and immunopathogenesis of autoimmune central nervous system disorders. Nat Rev Neurol 2023; 19:172-190. [PMID: 36788293 DOI: 10.1038/s41582-023-00776-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2023] [Indexed: 02/16/2023]
Abstract
The field of autoimmune neurology is rapidly evolving, and recent discoveries have advanced our understanding of disease aetiologies. In this article, we review the key pathogenic mechanisms underlying the development of CNS autoimmunity. First, we review non-modifiable risk factors, such as age, sex and ethnicity, as well as genetic factors such as monogenic variants, common variants in vulnerability genes and emerging HLA associations. Second, we highlight how interactions between environmental factors and epigenetics can modify disease onset and severity. Third, we review possible disease mechanisms underlying triggers that are associated with the loss of immune tolerance with consequent recognition of self-antigens; these triggers include infections, tumours and immune-checkpoint inhibitor therapies. Fourth, we outline how advances in our understanding of the anatomy of lymphatic drainage and neuroimmune interfaces are challenging long-held notions of CNS immune privilege, with direct relevance to CNS autoimmunity, and how disruption of B cell and T cell tolerance and the passage of immune cells between the peripheral and intrathecal compartments have key roles in initiating disease activity. Last, we consider novel therapeutic approaches based on our knowledge of the immunopathogenesis of autoimmune CNS disorders.
Collapse
Affiliation(s)
- Sudarshini Ramanathan
- Translational Neuroimmunology Group, Kids Neuroscience Centre, Children's Hospital at Westmead, Sydney, New South Wales, Australia
- Sydney Medical School, Faculty of Medicine and Health and Brain and Mind Centre, University of Sydney, Sydney, New South Wales, Australia
- Department of Neurology, Concord Hospital, Sydney, New South Wales, Australia
| | - Fabienne Brilot
- Translational Neuroimmunology Group, Kids Neuroscience Centre, Children's Hospital at Westmead, Sydney, New South Wales, Australia
- School of Medical Science, Faculty of Medicine and Health and Brain and Mind Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Sarosh R Irani
- Oxford Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Russell C Dale
- Translational Neuroimmunology Group, Kids Neuroscience Centre, Children's Hospital at Westmead, Sydney, New South Wales, Australia.
- Sydney Medical School, Faculty of Medicine and Health and Brain and Mind Centre, University of Sydney, Sydney, New South Wales, Australia.
- TY Nelson Department of Paediatric Neurology, Children's Hospital Westmead, Sydney, New South Wales, Australia.
| |
Collapse
|
248
|
Liang J, Yin H. STAM transports STING oligomers into extracellular vesicles, down-regulating the innate immune response. J Extracell Vesicles 2023; 12:e12316. [PMID: 36946680 PMCID: PMC10032202 DOI: 10.1002/jev2.12316] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 02/13/2023] [Accepted: 03/08/2023] [Indexed: 03/23/2023] Open
Abstract
Stimulator of interferon genes (STING) mediates the innate immune response against damaged endogenous double-strand DNA and exogenous virus infection. The location of STING is critical to the accurate control of defence signalling pathways. Recently, the effects of extracellular vesicles (EVs) in the regulation of innate immune signalling have been reported. Nevertheless, the particular roles played by STING in EVs and the related mechanisms have remained largely unknown. Herein, we report that when STING was activated in cells, EVs derived from these cells carried STING oligomers. Signal transducing adapter molecule 1 (STAM) was found to be a STING transporter that directly interacted with STING and facilitated STING transport into EVs. Importantly, the translocation of STING into EVs was a mechanism by which STING was degraded, suppressing the innate immune response. In summary, we elucidated the mechanism and function of the translocation of STING into EVs, adding to the understanding of STING activity regulation.
Collapse
Affiliation(s)
- Jiaqi Liang
- Key Laboratory of Bioorganic Phosphorous Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, School of Pharmaceutical SciencesTsinghua UniversityBeijingChina
| | - Hang Yin
- Key Laboratory of Bioorganic Phosphorous Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, School of Pharmaceutical SciencesTsinghua UniversityBeijingChina
- Beijing Advanced Innovation Center for Structural BiologyTsinghua UniversityBeijingChina
- Tsinghua‐Peking Center for Life SciencesTsinghua UniversityBeijingChina
| |
Collapse
|
249
|
Ryan TAJ, O’Neill LAJ. An Emerging Role for Type I Interferons as Critical Regulators of Blood Coagulation. Cells 2023; 12:778. [PMID: 36899914 PMCID: PMC10001161 DOI: 10.3390/cells12050778] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 02/22/2023] [Accepted: 02/28/2023] [Indexed: 03/05/2023] Open
Abstract
Type I interferons (IFNs) are central mediators of anti-viral and anti-bacterial host defence. Detection of microbes by innate immune cells via pattern recognition receptors (PRRs), including Toll-like receptors (TLRs) and cGAS-STING, induces the expression of type I IFN-stimulated genes. Primarily comprising the cytokines IFN-α and IFN-β, type I IFNs act via the type I IFN receptor in an autocrine or exocrine manner to orchestrate rapid and diverse innate immune responses. Growing evidence pinpoints type I IFN signalling as a fulcrum that not only induces blood coagulation as a core feature of the inflammatory response but is also activated by components of the coagulation cascade. In this review, we describe in detail recent studies identifying the type I IFN pathway as a modulator of vascular function and thrombosis. In addition, we profile discoveries showing that thrombin signalling via protease-activated receptors (PARs), which can synergize with TLRs, regulates the host response to infection via induction of type I IFN signalling. Thus, type I IFNs can have both protective (via maintenance of haemostasis) and pathological (facilitating thrombosis) effects on inflammation and coagulation signalling. These can manifest as an increased risk of thrombotic complications in infection and in type I interferonopathies such as systemic lupus erythematosus (SLE) and STING-associated vasculopathy with onset in infancy (SAVI). We also consider the effects on coagulation of recombinant type I IFN therapies in the clinic and discuss pharmacological regulation of type I IFN signalling as a potential mechanism by which aberrant coagulation and thrombosis may be treated therapeutically.
Collapse
Affiliation(s)
- Tristram A. J. Ryan
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin, Ireland
| | | |
Collapse
|
250
|
Zhang L, Wei X, Wang Z, Liu P, Hou Y, Xu Y, Su H, Koci MD, Yin H, Zhang C. NF-κB activation enhances STING signaling by altering microtubule-mediated STING trafficking. Cell Rep 2023; 42:112185. [PMID: 36857187 DOI: 10.1016/j.celrep.2023.112185] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 01/12/2023] [Accepted: 02/14/2023] [Indexed: 03/02/2023] Open
Abstract
It is widely known that stimulator of interferon genes (STING) can trigger nuclear factor κB (NF-κB) signaling. However, whether and how the NF-κB pathway affects STING signaling remains largely unclear. Here, we report that Toll-like receptor (TLR)-, interleukin-1 receptor (IL-1R)-, tumor necrosis factor receptor (TNFR)-, growth factor receptor (GF-R)-, and protein kinase C (PKC)-mediated NF-κB signaling activation dramatically enhances STING-mediated immune responses. Mechanistically, we find that STING interacts with microtubules, which plays a crucial role in STING intracellular trafficking. We further uncover that activation of the canonical NF-κB pathway induces microtubule depolymerization, which inhibits STING trafficking to lysosomes for degradation. This leads to increased levels of activated STING that persist for a longer period of time. The synergy between NF-κB and STING triggers a cascade-amplified interferon response and robust host antiviral defense. In addition, we observe that several gain-of-function mutations of STING abolish the microtubule-STING interaction and cause abnormal STING trafficking and ligand-independent STING autoactivation. Collectively, our data demonstrate that NF-κB activation enhances STING signaling by regulating microtubule-mediated STING trafficking.
Collapse
Affiliation(s)
- Lulu Zhang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Xubiao Wei
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Zhimeng Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Peiyuan Liu
- School of Life Science, Tianjin University, Tianjin, China
| | - Yanfei Hou
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Yifang Xu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Huili Su
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Matthew D Koci
- Prestage Department of Poultry Science, College of Agriculture and Life Sciences, North Carolina State University, Raleigh, NC, USA
| | - Hang Yin
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China.
| | - Conggang Zhang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China.
| |
Collapse
|