701
|
Dai J, Huang YJ, He X, Zhao M, Wang X, Liu ZS, Xue W, Cai H, Zhan XY, Huang SY, He K, Wang H, Wang N, Sang Z, Li T, Han QY, Mao J, Diao X, Song N, Chen Y, Li WH, Man JH, Li AL, Zhou T, Liu ZG, Zhang XM, Li T. Acetylation Blocks cGAS Activity and Inhibits Self-DNA-Induced Autoimmunity. Cell 2019; 176:1447-1460.e14. [PMID: 30799039 DOI: 10.1016/j.cell.2019.01.016] [Citation(s) in RCA: 255] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 11/27/2018] [Accepted: 01/04/2019] [Indexed: 12/30/2022]
Abstract
The presence of DNA in the cytoplasm is normally a sign of microbial infections and is quickly detected by cyclic GMP-AMP synthase (cGAS) to elicit anti-infection immune responses. However, chronic activation of cGAS by self-DNA leads to severe autoimmune diseases for which no effective treatment is available yet. Here we report that acetylation inhibits cGAS activation and that the enforced acetylation of cGAS by aspirin robustly suppresses self-DNA-induced autoimmunity. We find that cGAS acetylation on either Lys384, Lys394, or Lys414 contributes to keeping cGAS inactive. cGAS is deacetylated in response to DNA challenges. Importantly, we show that aspirin can directly acetylate cGAS and efficiently inhibit cGAS-mediated immune responses. Finally, we demonstrate that aspirin can effectively suppress self-DNA-induced autoimmunity in Aicardi-Goutières syndrome (AGS) patient cells and in an AGS mouse model. Thus, our study reveals that acetylation contributes to cGAS activity regulation and provides a potential therapy for treating DNA-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Jiang Dai
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China; State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Yi-Jiao Huang
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Xinhua He
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Ming Zhao
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Xinzheng Wang
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Zhao-Shan Liu
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Wen Xue
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Hong Cai
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Xiao-Yan Zhan
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Shao-Yi Huang
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China; State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Kun He
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Hongxia Wang
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Na Wang
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Zhihong Sang
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Tingting Li
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Qiu-Ying Han
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Jie Mao
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Xinwei Diao
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China; Department of Pathology, Xinqiao Hospital, 3(rd) Military Medical University, Chongqing 400037, China
| | - Nan Song
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Yuan Chen
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Wei-Hua Li
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Jiang-Hong Man
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Ai-Ling Li
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Tao Zhou
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China
| | - Zheng-Gang Liu
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Xue-Min Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China; State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China; Cancer Research Institute of Jilin University, the First Hospital of Jilin University, Changchun, Jilin Province 130021, China.
| | - Tao Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China; State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, 27 Tai-Ping Road, Beijing 100850, China.
| |
Collapse
|
702
|
Pei J, Zhang Y, Luo Q, Zheng W, Li W, Zeng X, Li Q, Quan J. STAT3 inhibition enhances CDN-induced STING signaling and antitumor immunity. Cancer Lett 2019; 450:110-122. [PMID: 30790684 DOI: 10.1016/j.canlet.2019.02.029] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/08/2019] [Accepted: 02/14/2019] [Indexed: 01/03/2023]
Abstract
Cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway is a key regulator in innate immunity and has emerged as a promising drug target in cancer treatment, but the utility of this pathway in therapeutic development is complicated by its dichotomous roles in tumor development and immunity. The activation of the STING pathway and the induced antitumor immunity could be attenuated by the feedback activation of IL-6/STAT3 pathway. Here we reported that STAT3 inhibition significantly enhanced the intensity and duration of STING signaling induced by the STING agonist c-diAM(PS)2. Such sensitization effect of STAT3 inhibition on STING signaling depended on STING rather than cGAS, which was mediated by simultaneously upregulating the positive modulators and downregulating the negative modulators of the STING pathway. Furthermore, the combination treatment with the STAT3 inhibitor and STING agonist markedly regressed tumor growth in syngeneic mice by increasing CD8+ T cells and reducing regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) in the tumor microenvironment. Our work provides a rationale for the combination of STAT3 inhibitors and STING agonists in cancer immunotherapy.
Collapse
Affiliation(s)
- Jianwen Pei
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Yibo Zhang
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Qinhong Luo
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Wenlv Zheng
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Wanxuan Li
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Xin Zeng
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Qinkai Li
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Junmin Quan
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China.
| |
Collapse
|
703
|
Luksch H, Stinson WA, Platt DJ, Qian W, Kalugotla G, Miner CA, Bennion BG, Gerbaulet A, Rösen-Wolff A, Miner JJ. STING-associated lung disease in mice relies on T cells but not type I interferon. J Allergy Clin Immunol 2019; 144:254-266.e8. [PMID: 30772497 DOI: 10.1016/j.jaci.2019.01.044] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 01/23/2019] [Accepted: 01/28/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND Monogenic interferonopathies are thought to be mediated by type I interferon. For example, a gain-of-function mutation in stimulator of interferon genes (STING; N153S) upregulates type I interferon-stimulated genes and causes perivascular inflammatory lung disease in mice. The equivalent mutation in human subjects also causes lung disease, which is thought to require signaling through the cyclic GMP-AMP synthase (cGAS)-STING pathway and subsequent activation of interferon regulatory factors (IRFs) 3 and 7, type I interferon, and interferon-stimulated genes. OBJECTIVE We set out to define the roles of cGAS, IRF3, IRF7, the type I interferon receptor (IFN-α and IFN-β receptor subunit 1 [IFNAR1]), T cells, and B cells in spontaneous lung disease in STING N153S mice. METHODS STING N153S mice were crossed to animals lacking cGAS, IRF3/IRF7, IFNAR1, adaptive immunity, αβ T cells, and mature B cells. Mice were evaluated for spontaneous lung disease. Additionally, bone marrow chimeric mice were assessed for lung disease severity and survival. RESULTS Lung disease in STING N153S mice developed independently of cGAS, IRF3/IRF7, and IFNAR1. Bone marrow transplantation revealed that certain features of STING N153S-associated disease are intrinsic to the hematopoietic compartment. Recombination-activating gene 1 (Rag1)-/- STING N153S mice that lack adaptive immunity had no lung disease, and T-cell receptor β chain (Tcrb)-/- STING N153S animals only had mild disease. STING N153S led to a reduction in percentages and numbers of naive and regulatory T cells, as well as an increased frequency of cytokine-producing effector T cells. CONCLUSION Spontaneous lung disease in STING N153S mice develops independently of type I interferon signaling and cGAS. STING N153S relies primarily on T cells to promote lung disease in mice.
Collapse
Affiliation(s)
- Hella Luksch
- Department of Pediatrics, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - W Alexander Stinson
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Mo
| | - Derek J Platt
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, Mo
| | - Wei Qian
- Department of Medicine, Washington University School of Medicine, St Louis, Mo
| | - Gowri Kalugotla
- Department of Medicine, Washington University School of Medicine, St Louis, Mo
| | - Cathrine A Miner
- Department of Medicine, Washington University School of Medicine, St Louis, Mo
| | - Brock G Bennion
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Mo
| | | | - Angela Rösen-Wolff
- Department of Pediatrics, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| | - Jonathan J Miner
- Department of Medicine, Washington University School of Medicine, St Louis, Mo; Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Mo; Department of Molecular Microbiology, Washington University School of Medicine, St Louis, Mo.
| |
Collapse
|
704
|
Abstract
Inborn errors of immunity usually not only result in immunodeficiency but may also manifest as immune dysregulation in the form of autoinflammation, autoimmunity, or sometimes malignancy. One of the most recently discovered monogenic disorder of immune dysregulation is COPA syndrome. COPA syndrome is an inherited autoimmune disorder caused by mutations in COPA gene. COPA gene encodes for α subunit of the COP1 protein, which is involved in the reverse vesicular protein transport from Golgi apparatus to the endoplasmic reticulum (ER). The inheritance pattern of COPA syndrome is autosomal dominant, and the patients typically present with interstitial lung disease with pulmonary hemorrhage and subsequently develop arthritis. Immunological features involve autoantibody formation, elevated expression of IL-1β and IL-6, and increase in the number of Th17 cells. Molecular pathophysiology of COPA syndrome is not clearly understood. However, it is known that accumulation of unfolded proteins in ER leads to ER stress, which is an indirect result of aberrant vesicular transport of proteins from Golgi apparatus to ER and defective cellular autophagy. ER stress induces inflammation and is responsible for pathogenesis of a large number of chronic inflammatory diseases. Unfolded protein response process responds to improperly folded proteins and defends against stress in ER to ensure the fidelity of the protein folding. It maintains the expression of stress-response genes and causes initiation of inflammatory signaling pathways essential for the innate immunity. Mutation in COPA gene associated with defective protein sorting to ER has unearthed a new primary immunodeficiency disease with a unique clinical phenotype. This review highlights the clinical and molecular aspects of COPA syndrome.
Collapse
Affiliation(s)
- Rajni Kumrah
- Pediatric Allergy and Immunology Unit, Department of Pediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India,
| | - Babu Mathew
- Pediatric Allergy and Immunology Unit, Department of Pediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India,
| | - Pandiarajan Vignesh
- Pediatric Allergy and Immunology Unit, Department of Pediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India,
| | - Surjit Singh
- Pediatric Allergy and Immunology Unit, Department of Pediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India,
| | - Amit Rawat
- Pediatric Allergy and Immunology Unit, Department of Pediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India,
| |
Collapse
|
705
|
Xu M, Liu PP, Li H. Innate Immune Signaling and Its Role in Metabolic and Cardiovascular Diseases. Physiol Rev 2019; 99:893-948. [PMID: 30565509 DOI: 10.1152/physrev.00065.2017] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The innate immune system is an evolutionarily conserved system that senses and defends against infection and irritation. Innate immune signaling is a complex cascade that quickly recognizes infectious threats through multiple germline-encoded cell surface or cytoplasmic receptors and transmits signals for the deployment of proper countermeasures through adaptors, kinases, and transcription factors, resulting in the production of cytokines. As the first response of the innate immune system to pathogenic signals, inflammatory responses must be rapid and specific to establish a physical barrier against the spread of infection and must subsequently be terminated once the pathogens have been cleared. Long-lasting and low-grade chronic inflammation is a distinguishing feature of type 2 diabetes and cardiovascular diseases, which are currently major public health problems. Cardiometabolic stress-induced inflammatory responses activate innate immune signaling, which directly contributes to the development of cardiometabolic diseases. Additionally, although the innate immune elements are highly conserved in higher-order jawed vertebrates, lower-grade jawless vertebrates lack several transcription factors and inflammatory cytokine genes downstream of the Toll-like receptors (TLRs) and retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs) pathways, suggesting that innate immune signaling components may additionally function in an immune-independent way. Notably, recent studies from our group and others have revealed that innate immune signaling can function as a vital regulator of cardiometabolic homeostasis independent of its immune function. Therefore, further investigation of innate immune signaling in cardiometabolic systems may facilitate the discovery of new strategies to manage the initiation and progression of cardiometabolic disorders, leading to better treatments for these diseases. In this review, we summarize the current progress in innate immune signaling studies and the regulatory function of innate immunity in cardiometabolic diseases. Notably, we highlight the immune-independent effects of innate immune signaling components on the development of cardiometabolic disorders.
Collapse
Affiliation(s)
- Meng Xu
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan , China ; Medical Research Center, Zhongnan Hospital of Wuhan University , Wuhan , China ; Animal Experiment Center, Wuhan University , Wuhan , China ; Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ontario , Canada
| | - Peter P Liu
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan , China ; Medical Research Center, Zhongnan Hospital of Wuhan University , Wuhan , China ; Animal Experiment Center, Wuhan University , Wuhan , China ; Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ontario , Canada
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan , China ; Medical Research Center, Zhongnan Hospital of Wuhan University , Wuhan , China ; Animal Experiment Center, Wuhan University , Wuhan , China ; Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, Ontario , Canada
| |
Collapse
|
706
|
A Human Gain-of-Function STING Mutation Causes Immunodeficiency and Gammaherpesvirus-Induced Pulmonary Fibrosis in Mice. J Virol 2019; 93:JVI.01806-18. [PMID: 30463976 DOI: 10.1128/jvi.01806-18] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 11/15/2018] [Indexed: 12/11/2022] Open
Abstract
We previously generated STING N153S knock-in mice that have a human disease-associated gain-of-function mutation in STING. Patients with this mutation (STING N154S in humans) develop STING-associated vasculopathy with onset in infancy (SAVI), a severe pediatric autoinflammatory disease characterized by pulmonary fibrosis. Since this mutation promotes the upregulation of antiviral type I interferon-stimulated genes (ISGs), we hypothesized that STING N153S knock-in mice may develop more severe autoinflammatory disease in response to a virus challenge. To test this hypothesis, we infected heterozygous STING N153S mice with murine gammaherpesvirus 68 (γHV68). STING N153S mice were highly vulnerable to infection and developed pulmonary fibrosis after infection. In addition to impairing CD8+ T cell responses and humoral immunity, STING N153S also promoted the replication of γHV68 in cultured macrophages. In further support of a combined innate and adaptive immunodeficiency, γHV68 infection was more severe in Rag1-/- STING N153S mice than in Rag1-/- littermate mice, which completely lack adaptive immunity. Thus, a gain-of-function STING mutation creates a combined innate and adaptive immunodeficiency that leads to virus-induced pulmonary fibrosis.IMPORTANCE A variety of human rheumatologic disease-causing mutations have recently been identified. Some of these mutations are found in viral nucleic acid-sensing proteins, but whether viruses can influence the onset or progression of these human diseases is less well understood. One such autoinflammatory disease, called STING-associated vasculopathy with onset in infancy (SAVI), affects children and leads to severe lung disease. We generated mice with a SAVI-associated STING mutation and infected them with γHV68, a common DNA virus that is related to human Epstein-Barr virus. Mice with the human disease-causing STING mutation were more vulnerable to infection than wild-type littermate control animals. Furthermore, the STING mutant mice developed lung fibrosis similar to that of patients with SAVI. These findings reveal that a human STING mutation creates severe immunodeficiency, leading to virus-induced lung disease in mice.
Collapse
|
707
|
Srikanth S, Woo JS, Wu B, El-Sherbiny YM, Leung J, Chupradit K, Rice L, Seo GJ, Calmettes G, Ramakrishna C, Cantin E, An DS, Sun R, Wu TT, Jung JU, Savic S, Gwack Y. The Ca 2+ sensor STIM1 regulates the type I interferon response by retaining the signaling adaptor STING at the endoplasmic reticulum. Nat Immunol 2019; 20:152-162. [PMID: 30643259 PMCID: PMC6340781 DOI: 10.1038/s41590-018-0287-8] [Citation(s) in RCA: 271] [Impact Index Per Article: 45.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 11/19/2018] [Indexed: 12/17/2022]
Abstract
Stimulator of interferon genes (STING) is an endoplasmic reticulum (ER) signaling adaptor that is essential for the type I interferon response to DNA pathogens. Aberrant activation of STING is linked to the pathology of autoimmune and autoinflammatory diseases. The rate-limiting step for the activation of STING is its translocation from the ER to the ER-Golgi intermediate compartment. Here, we found that deficiency in the Ca2+ sensor stromal interaction molecule 1 (STIM1) caused spontaneous activation of STING and enhanced expression of type I interferons under resting conditions in mice and a patient with combined immunodeficiency. Mechanistically, STIM1 associated with STING to retain it in the ER membrane, and coexpression of full-length STIM1 or a STING-interacting fragment of STIM1 suppressed the function of dominant STING mutants that cause autoinflammatory diseases. Furthermore, deficiency in STIM1 strongly enhanced the expression of type I interferons after viral infection and prevented the lethality of infection with a DNA virus in vivo. This work delineates a STIM1-STING circuit that maintains the resting state of the STING pathway.
Collapse
Affiliation(s)
- Sonal Srikanth
- Department of Physiology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA.
| | - Jin Seok Woo
- Department of Physiology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Beibei Wu
- Department of Physiology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Yasser M El-Sherbiny
- National Institute for Health Research-Leeds Biomedical Research Centre and Leeds Institute of Rheumatic and Musculoskeletal Medicine, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, UK
- Clinical Pathology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- School of Science and Technology, Department of Biosciences, Nottingham Trent University, Nottingham, UK
| | - Jennifer Leung
- Department of Physiology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Koollawat Chupradit
- Division of Hematology-Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- School of Nursing, University of California at Los Angeles, Los Angeles, CA, USA
- UCLA AIDS Institute, Los Angeles, CA, USA
| | - Laura Rice
- Leeds Institute of Biomedical and Clinical Sciences, University of Leeds, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, UK
| | - Gil Ju Seo
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Guillaume Calmettes
- Department of Medicine (Cardiology), David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Chandran Ramakrishna
- Department of Molecular Immunology, City of Hope Beckman Research Institute, Duarte, CA, USA
| | - Edouard Cantin
- Department of Molecular Immunology, City of Hope Beckman Research Institute, Duarte, CA, USA
| | - Dong Sung An
- Division of Hematology-Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- School of Nursing, University of California at Los Angeles, Los Angeles, CA, USA
- UCLA AIDS Institute, Los Angeles, CA, USA
| | - Ren Sun
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA, USA
| | - Ting-Ting Wu
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, CA, USA
| | - Jae U Jung
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Sinisa Savic
- National Institute for Health Research-Leeds Biomedical Research Centre and Leeds Institute of Rheumatic and Musculoskeletal Medicine, Wellcome Trust Brenner Building, St James's University Hospital, Leeds, UK
- Department of Clinical Immunology and Allergy, St James's University Hospital, Leeds, UK
| | - Yousang Gwack
- Department of Physiology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA.
| |
Collapse
|
708
|
Dansako H, Imai H, Ueda Y, Satoh S, Shimotohno K, Kato N. High-level expression of STING restricts susceptibility to HBV by mediating type III IFN induction. FASEB Bioadv 2019; 1:67-80. [PMID: 32123822 PMCID: PMC6996391 DOI: 10.1096/fba.1022] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 09/21/2018] [Accepted: 09/28/2018] [Indexed: 01/05/2023] Open
Abstract
Hepatitis B virus (HBV) is a hepatotropic DNA virus causing hepatic diseases such as chronic hepatitis, liver cirrhosis, and hepatocellular carcinoma. To study HBV, human hepatoma HepG2 cells are currently used as an HBV infectious cell culture model worldwide. HepG2 cells exhibit susceptibility to HBV by exogenously expressing sodium taurocholate cotransporting polypeptide (NTCP). We herein demonstrated that human immortalized hepatocyte NKNT-3 cells exhibited susceptibility to HBV by exogenously expressing NTCP (NKNT-3/NTCP cells). By comparing cyclic GMP-AMP synthetase (cGAS)-stimulator of interferon genes (STING) signaling pathway in several NKNT-3/NTCP cell-derived cell clones, we found that STING was highly expressed in cell clones exhibiting resistance but not susceptibility to HBV. High-level expression of STING was implicated in HBV-triggered induction of type III IFN and a pro-inflammatory cytokine, IL-6. In contrast, RNAi-mediated knockdown of STING inhibited type III IFN induction and restored the levels of HBV total transcript in an HBV-infected cell clone exhibiting resistance to HBV. These results suggest that STING regulates susceptibility to HBV by its expression levels. STING may thus be a novel target for anti-HBV strategies.
Collapse
Affiliation(s)
- Hiromichi Dansako
- Department of Tumor VirologyOkayama University Graduate School of Medicine, Dentistry and Pharmaceutical SciencesOkayamaJapan
| | - Hirotaka Imai
- Department of Tumor VirologyOkayama University Graduate School of Medicine, Dentistry and Pharmaceutical SciencesOkayamaJapan
| | - Youki Ueda
- Department of Tumor VirologyOkayama University Graduate School of Medicine, Dentistry and Pharmaceutical SciencesOkayamaJapan
| | - Shinya Satoh
- Department of Tumor VirologyOkayama University Graduate School of Medicine, Dentistry and Pharmaceutical SciencesOkayamaJapan
| | - Kunitada Shimotohno
- Research Center for Hepatitis and Immunology, National Center for Global Health and MedicineIchikawaJapan
| | - Nobuyuki Kato
- Department of Tumor VirologyOkayama University Graduate School of Medicine, Dentistry and Pharmaceutical SciencesOkayamaJapan
| |
Collapse
|
709
|
Beck DB, Aksentijevich I. Biochemistry of Autoinflammatory Diseases: Catalyzing Monogenic Disease. Front Immunol 2019; 10:101. [PMID: 30766537 PMCID: PMC6365650 DOI: 10.3389/fimmu.2019.00101] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 01/14/2019] [Indexed: 12/20/2022] Open
Abstract
Monogenic autoinflammatory disorders are a group of conditions defined by systemic or localized inflammation without identifiable causes, such as infection. In contrast to classical primary immunodeficiencies that manifest with impaired immune responses, these disorders are due to defects in genes that regulate innate immunity leading to constitutive activation of pro-inflammatory signaling. Through studying patients with rare autoinflammatory conditions, novel mechanisms of inflammation have been identified that bare on our understanding not only of basic signaling in inflammatory cells, but also of the pathogenesis of more common inflammatory diseases and have guided treatment modalities. Autoinflammation has further been implicated as an important component of cardiovascular, neurodegenerative, and metabolic syndromes. In this review, we will focus on a subset of inherited enzymatic deficiencies that lead to constitutive inflammation, and how these rare diseases have provided insights into diverse areas of cell biology not restricted to immune cells. In this way, Mendelian disorders of the innate immune system, and in particular loss of catalytic activity of enzymes in distinct pathways, have expanded our understanding of the interplay between many seemingly disparate cellular processes. We also explore the overlap between autoinflammation, autoimmunity, and immunodeficiency, which has been increasingly recognized in patients with dysregulated immune responses.
Collapse
Affiliation(s)
- David B Beck
- Metabolic, Cardiovascular and Inflammatory Disease Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Ivona Aksentijevich
- Metabolic, Cardiovascular and Inflammatory Disease Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
710
|
Wang Z, Sheng C, Yao C, Chen H, Wang D, Chen S. The EF-Hand Protein CALML6 Suppresses Antiviral Innate Immunity by Impairing IRF3 Dimerization. Cell Rep 2019; 26:1273-1285.e5. [PMID: 30699354 DOI: 10.1016/j.celrep.2019.01.030] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 12/09/2018] [Accepted: 01/08/2019] [Indexed: 02/07/2023] Open
Abstract
The transcription factor IRF3 is phosphorylated in response to viral infection, and it subsequently forms a homodimer and translocates into the nucleus to induce the transcription of genes important for antiviral immunity, such as type I interferons (IFNs). This multistep process is essential for host defense against viral infection, but its regulation remains elusive. Here, we report that the EF-hand protein calmodulin-like 6 (CALML6) directly bound to the phosphorylated serine-rich (SR) region of IRF3 and impaired its dimerization and nuclear translocation. Enforced CALML6 expression suppressed viral infection-induced production of IFN-β and expression of IFN-stimulated genes (ISGs), whereas CALML6 deficiency had the opposite effect. In addition, impaired IFN-β and ISG expression in bone-marrow-derived macrophages and tissues of CALML6 transgenic mice promoted viral replication. These findings identify a phosphorylation-dependent negative feedback loop that maintains the homeostasis of antiviral innate immunity.
Collapse
Affiliation(s)
- Ziyang Wang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, Guangdong, China
| | - Chunjie Sheng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, Guangdong, China
| | - Chen Yao
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, Guangdong, China
| | - Hongyuan Chen
- Department of Pathogen Biology and Immunology, School of Basic Course, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China
| | - Dan Wang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, Guangdong, China
| | - Shuai Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, Guangdong, China.
| |
Collapse
|
711
|
Georgin-Lavialle S, Fayand A, Rodrigues F, Bachmeyer C, Savey L, Grateau G. Autoinflammatory diseases: State of the art. Presse Med 2019; 48:e25-e48. [PMID: 30686513 DOI: 10.1016/j.lpm.2018.12.003] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Autoinflammatory diseases are characterized by innate immunity abnormalities. In autoinflammatory diseases (AID), inflammatory blood biomarkers are elevated during crisis without infection and usually without autoantibodies. The first 4 described AID were familial Mediterranean fever, cryopyrin-associated periodic fever syndrome (CAPS) or NLRP3-associated autoinflammatory disease (NRLP3-AID), mevalonate kinase deficiency (MKD) and TNFRSF1A-receptor associated periodic fever syndrome (TRAPS). Since their description 20 years ago, and with the progresses of genetic analysis, many new diseases have been discovered; some with recurrent fever, others with predominant cutaneous symptoms or even immune deficiency. After describing the 4 historical recurrent fevers, some polygenic inflammatory diseases will also be shortly described such as Still disease and periodic fever with adenitis, pharyngitis and aphtous (PFAPA) syndrome. To better explore AID, some key anamnesis features are crucial such as the family tree, the age at onset, crisis length and organs involved in the clinical symptoms. An acute phase response is mandatory in crisis.
Collapse
Affiliation(s)
- Sophie Georgin-Lavialle
- AP-HP, hôpital Tenon, Sorbonne université, service de médecine interne, centre de référence des maladies auto-inflammatoires et des amyloses d'origine inflammatoire (CEREMAIA), 75020 Paris, France; Assistance publique-Hôpitaux de Paris, hôpital Trousseau, université Pierre-et-Marie-Curie (UPMC)-Paris 6, Inserm UMRS_933, 75012 Paris, France.
| | - Antoine Fayand
- AP-HP, hôpital Tenon, Sorbonne université, service de médecine interne, centre de référence des maladies auto-inflammatoires et des amyloses d'origine inflammatoire (CEREMAIA), 75020 Paris, France
| | - François Rodrigues
- AP-HP, hôpital Tenon, Sorbonne université, service de médecine interne, centre de référence des maladies auto-inflammatoires et des amyloses d'origine inflammatoire (CEREMAIA), 75020 Paris, France
| | - Claude Bachmeyer
- AP-HP, hôpital Tenon, Sorbonne université, service de médecine interne, centre de référence des maladies auto-inflammatoires et des amyloses d'origine inflammatoire (CEREMAIA), 75020 Paris, France
| | - Léa Savey
- AP-HP, hôpital Tenon, Sorbonne université, service de médecine interne, centre de référence des maladies auto-inflammatoires et des amyloses d'origine inflammatoire (CEREMAIA), 75020 Paris, France
| | - Gilles Grateau
- AP-HP, hôpital Tenon, Sorbonne université, service de médecine interne, centre de référence des maladies auto-inflammatoires et des amyloses d'origine inflammatoire (CEREMAIA), 75020 Paris, France; Assistance publique-Hôpitaux de Paris, hôpital Trousseau, université Pierre-et-Marie-Curie (UPMC)-Paris 6, Inserm UMRS_933, 75012 Paris, France
| |
Collapse
|
712
|
How to prescribe a genetic test for the diagnosis of autoinflammatory diseases? Presse Med 2019; 48:e49-e59. [PMID: 30665783 DOI: 10.1016/j.lpm.2018.08.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 08/07/2018] [Indexed: 01/14/2023] Open
Abstract
The systemic autoinflammatory disorders (SAIDs) are associated with dysregulation of the innate immune system, affecting pro-inflammatory cytokines and apoptosis pathways. The spectrum of SAIDs continues to grow with over 30 different disorders identified to date. The main indication for genetic referral is when a patient presents with clinical symptoms consistent with one or more of the SAIDs. Thus, in making a referral for DNA screening, clinical information that supports the choice for screening of one or more SAIDs genes is required. Many of the SAIDs can display overlapping, partial or atypical symptoms, which makes the differential diagnosis extremely difficult and thus heavily dependent on genetic testing. Various attempts have been aimed at improving the efficiency of SAIDs diagnosis by proposing a set of clinical criteria to guide the genetic analysis of the SAIDs. In the last decade, due to application of the next-generation sequencing (NGS) the genetic diagnosis in patients with SAIDs have greatly improved; novel diseases and disease-associated genes have been identified and remarkable progress has been made in the genetic characterization of the undiagnosed patients and the sporadic cases. To date more than 800 variants have been recorded on the Infevers database, an online repository for DNA changes in genes associated with SAIDs (http://fmf.igh.cnrs.fr/ISSAID/infevers/). Recently, it has been updated with the new guidelines for classification of genetic variants pathogenicity in the in four most recognised SAIDs genes: MEFV, TNFRSF1A, NLRP3 and MVK.
Collapse
|
713
|
Novel proteasome assembly chaperone mutations in PSMG2/PAC2 cause the autoinflammatory interferonopathy CANDLE/PRAAS4. J Allergy Clin Immunol 2019; 143:1939-1943.e8. [PMID: 30664889 DOI: 10.1016/j.jaci.2018.12.1012] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 11/30/2018] [Accepted: 12/07/2018] [Indexed: 12/24/2022]
|
714
|
Uggenti C, Lepelley A, Crow YJ. Self-Awareness: Nucleic Acid-Driven Inflammation and the Type I Interferonopathies. Annu Rev Immunol 2019; 37:247-267. [PMID: 30633609 DOI: 10.1146/annurev-immunol-042718-041257] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Recognition of foreign nucleic acids is the primary mechanism by which a type I interferon-mediated antiviral response is triggered. Given that human cells are replete with DNA and RNA, this evolutionary strategy poses an inherent biological challenge, i.e., the fundamental requirement to reliably differentiate self-nucleic acids from nonself nucleic acids. We suggest that the group of Mendelian inborn errors of immunity referred to as the type I interferonopathies relate to a breakdown of self/nonself discrimination, with the associated mutant genotypes involving molecules playing direct or indirect roles in nucleic acid signaling. This perspective begs the question as to the sources of self-derived nucleic acids that drive an inappropriate immune response. Resolving this question will provide fundamental insights into immune tolerance, antiviral signaling, and complex autoinflammatory disease states. Here we develop these ideas, discussing type I interferonopathies within the broader framework of nucleic acid-driven inflammation.
Collapse
Affiliation(s)
- Carolina Uggenti
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom;
| | - Alice Lepelley
- Laboratory of Neurogenetics and Neuroinflammation, Institut Imagine, Paris 75015, France
| | - Yanick J Crow
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom; .,Laboratory of Neurogenetics and Neuroinflammation, Institut Imagine, Paris 75015, France.,Paris Descartes University, Sorbonne-Paris-Cité, Paris 75006, France
| |
Collapse
|
715
|
Sarhan J, Liu BC, Muendlein HI, Weindel CG, Smirnova I, Tang AY, Ilyukha V, Sorokin M, Buzdin A, Fitzgerald KA, Poltorak A. Constitutive interferon signaling maintains critical threshold of MLKL expression to license necroptosis. Cell Death Differ 2019; 26:332-347. [PMID: 29786074 PMCID: PMC6329789 DOI: 10.1038/s41418-018-0122-7] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 04/08/2018] [Accepted: 04/10/2018] [Indexed: 12/12/2022] Open
Abstract
Interferons (IFNs) are critical determinants in immune-competence and autoimmunity, and are endogenously regulated by a low-level constitutive feedback loop. However, little is known about the functions and origins of constitutive IFN. Recently, lipopolysaccharide (LPS)-induced IFN was implicated as a driver of necroptosis, a necrotic form of cell death downstream of receptor-interacting protein (RIP) kinase activation and executed by mixed lineage kinase like-domain (MLKL) protein. We found that the pre-established IFN status of the cell, instead of LPS-induced IFN, is critical for the early initiation of necroptosis in macrophages. This pre-established IFN signature stems from cytosolic DNA sensing via cGAS/STING, and maintains the expression of MLKL and one or more unknown effectors above a critical threshold to allow for MLKL oligomerization and cell death. Finally, we found that elevated IFN-signaling in systemic lupus erythematosus (SLE) augments necroptosis, providing a link between pathological IFN and tissue damage during autoimmunity.
Collapse
Affiliation(s)
- Joseph Sarhan
- Medical Scientist Training Program (MSTP), Tufts University School of Medicine, Boston, MA, 02111, USA
- Graduate Program in Immunology, Tufts University Sackler School of Biomedical Sciences, Boston, MA, 02111, USA
| | - Beiyun C Liu
- Graduate Program in Immunology, Tufts University Sackler School of Biomedical Sciences, Boston, MA, 02111, USA
| | - Hayley I Muendlein
- Graduate Program in Genetics, Tufts University Sackler School of Biomedical Sciences, Boston, MA, 02111, USA
| | - Chi G Weindel
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX, 77808, USA
| | - Irina Smirnova
- Department of Immunology, Tufts University School of Medicine, Boston, MA, 02111, USA
| | - Amy Y Tang
- Department of Immunology, Tufts University School of Medicine, Boston, MA, 02111, USA
| | - Vladimir Ilyukha
- Petrozavodsk State University, Petrozavodsk, Republic of Karelia, 185910, Russia
| | - Maxim Sorokin
- National Research Center, Kurchatov Institute, Moscow, Russian Federation
| | - Anton Buzdin
- National Research Center, Kurchatov Institute, Moscow, Russian Federation
- I.M. Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | - Katherine A Fitzgerald
- Program in Innate Immunity, University of Massachusetts Medical School, Worcester, MA, 01605, USA
- Centre for Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, NTNU, 7491, Trondheim, Norway
| | - Alexander Poltorak
- Graduate Program in Immunology, Tufts University Sackler School of Biomedical Sciences, Boston, MA, 02111, USA.
- Graduate Program in Genetics, Tufts University Sackler School of Biomedical Sciences, Boston, MA, 02111, USA.
- Department of Immunology, Tufts University School of Medicine, Boston, MA, 02111, USA.
- Petrozavodsk State University, Petrozavodsk, Republic of Karelia, 185910, Russia.
| |
Collapse
|
716
|
Zhu Y, Deng J, Nan ML, Zhang J, Okekunle A, Li JY, Yu XQ, Wang PH. The Interplay Between Pattern Recognition Receptors and Autophagy in Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1209:79-108. [PMID: 31728866 DOI: 10.1007/978-981-15-0606-2_6] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Pattern recognition receptors (PRRs) are sensors of exogenous and endogenous "danger" signals from pathogen-associated molecular patterns (PAMPs), and damage associated molecular patterns (DAMPs), while autophagy can respond to these signals to control homeostasis. Almost all PRRs can induce autophagy directly or indirectly. Toll-like receptors (TLRs), Nod-like receptors (NLRs), retinoic acid-inducible gene-I-like receptors (RLRs), and cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) pathway can induce autophagy directly through Beclin-1 or LC3-dependent pathway, while the interactions with the receptor for advanced glycation end products (RAGE)/high mobility group box 1 (HMGB1), CD91/Calreticulin, and TLRs/HSPs are achieved by protein, Ca2+, and mitochondrial homeostasis. Autophagy presents antigens to PRRs and helps to clean the pathogens. In addition, the induced autophagy can form a negative feedback regulation of PRRs-mediated inflammation in cell/disease-specific manner to maintain homeostasis and prevent excessive inflammation. Understanding the interaction between PRRs and autophagy in a specific disease will promote drug development for immunotherapy. Here, we focus on the interactions between PRRs and autophagy and how they affect the inflammatory response.
Collapse
Affiliation(s)
- Yun Zhu
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Institute of Pediatrics, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China.,School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Jian Deng
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Mei-Ling Nan
- Advanced Medical Research Institute, Shandong University, Jinan, 250012, Shandong, China
| | - Jing Zhang
- Advanced Medical Research Institute, Shandong University, Jinan, 250012, Shandong, China
| | - Akinkunmi Okekunle
- The Postgraduate College, University of Ibadan, Ibadan, 200284, Nigeria.,Department of Epidemiology and Medical Statistics, College of Medicine, University of Ibadan, Ibadan, 200284, Nigeria
| | - Jiang-Yuan Li
- Key Laboratory for Cell and Gene Engineering of Zhejiang Province, College of Life Sciences, Zhejiang University, Hangzhou, 310058, People's Republic of China
| | - Xiao-Qiang Yu
- School of Biological Sciences, University of Missouri-Kansas City, Kansas City, MO, 64110-2499, USA
| | - Pei-Hui Wang
- Advanced Medical Research Institute, Shandong University, Jinan, 250012, Shandong, China. .,School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong, China.
| |
Collapse
|
717
|
Hu Q, Zhou Q, Wu J, Wu X, Ren J. The Role of Mitochondrial DNA in the Development of Ischemia Reperfusion Injury. Shock 2019; 51:52-59. [PMID: 30286034 DOI: 10.1097/shk.0000000000001190] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Ischemia/reperfusion (I/R) injury is a common occurrence resulting from acute mesenteric ischemia, traumatic or septic shock, burns, and surgical procedures that can lead to multiple organ failure and high mortality in critically ill patients. Mitochondria are often considered the cellular power factory via their capacity for ATP generation. Recently, mitochondria have been further identified as vital regulators of cell death, inflammation, and oxidative stress, all of which can aggravate I/R injury. Studies have indicated that mitochondrial DNA (mtDNA) damage leads to mitochondrial dysfunction and aggravates I/R injury. mtDNA is emerging as an agonist of the innate immune system that influences inflammatory pathology during I/R injury. In addition, when mtDNA is released into the cytoplasm, extracellular milieu, or circulation, it can activate multiple pattern-recognition receptors to trigger type I interferon and pro-inflammatory responses. Here, we review the emerging role of mtDNA in I/R injury to highlight novel mechanistic insights and discuss the pathophysiological relevance of mitochondrial biology.
Collapse
Affiliation(s)
- Qiongyuan Hu
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
- Lab for Trauma and Surgical Infection, Nanjing, China
| | - Quan Zhou
- Center for Reproductive Medicine, Drum Tower Clinic Medical College of Nanjing Medical University, Nanjing, China
| | - Jie Wu
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
- Lab for Trauma and Surgical Infection, Nanjing, China
| | - Xiuwen Wu
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
- Lab for Trauma and Surgical Infection, Nanjing, China
| | - Jianan Ren
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
- Lab for Trauma and Surgical Infection, Nanjing, China
| |
Collapse
|
718
|
Toxic epidermal necrolysis-like dermatomyositis associated with antimelanoma differentiation antigen 5. JAAD Case Rep 2018; 5:91-93. [PMID: 30581943 PMCID: PMC6297259 DOI: 10.1016/j.jdcr.2018.10.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
719
|
STING-dependent sensing of self-DNA drives silica-induced lung inflammation. Nat Commun 2018; 9:5226. [PMID: 30523277 PMCID: PMC6283886 DOI: 10.1038/s41467-018-07425-1] [Citation(s) in RCA: 203] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 10/28/2018] [Indexed: 12/11/2022] Open
Abstract
Silica particles induce lung inflammation and fibrosis. Here we show that stimulator of interferon genes (STING) is essential for silica-induced lung inflammation. In mice, silica induces lung cell death and self-dsDNA release in the bronchoalveolar space that activates STING pathway. Degradation of extracellular self-dsDNA by DNase I inhibits silica-induced STING activation and the downstream type I IFN response. Patients with silicosis have increased circulating dsDNA and CXCL10 in sputum, and patients with fibrotic interstitial lung disease display STING activation and CXCL10 in the lung. In vitro, while mitochondrial dsDNA is sensed by cGAS-STING in dendritic cells, in macrophages extracellular dsDNA activates STING independent of cGAS after silica exposure. These results reveal an essential function of STING-mediated self-dsDNA sensing after silica exposure, and identify DNase I as a potential therapy for silica-induced lung inflammation. Silica particles induce intereukin-1 (IL-1) response to contribute to lung inflammation, but the underlying mechanism is unclear. Here the authors show that silica induces cell death and release of mitochondria and genomic DNA, which are sensed by STING with or without involving cGAS, respectively, for IL-1 induction and lung inflammation.
Collapse
|
720
|
G3BP1 promotes DNA binding and activation of cGAS. Nat Immunol 2018; 20:18-28. [PMID: 30510222 DOI: 10.1038/s41590-018-0262-4] [Citation(s) in RCA: 218] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 10/17/2018] [Indexed: 01/05/2023]
Abstract
Cyclic GMP-AMP synthase (cGAS) is a key sensor responsible for cytosolic DNA detection. Here we report that GTPase-activating protein SH3 domain-binding protein 1 (G3BP1) is critical for DNA sensing and efficient activation of cGAS. G3BP1 enhanced DNA binding of cGAS by promoting the formation of large cGAS complexes. G3BP1 deficiency led to inefficient DNA binding by cGAS and inhibited cGAS-dependent interferon (IFN) production. The G3BP1 inhibitor epigallocatechin gallate (EGCG) disrupted existing G3BP1-cGAS complexes and inhibited DNA-triggered cGAS activation, thereby blocking DNA-induced IFN production both in vivo and in vitro. EGCG administration blunted self DNA-induced autoinflammatory responses in an Aicardi-Goutières syndrome (AGS) mouse model and reduced IFN-stimulated gene expression in cells from a patient with AGS. Thus, our study reveals that G3BP1 physically interacts with and primes cGAS for efficient activation. Furthermore, EGCG-mediated inhibition of G3BP1 provides a potential treatment for cGAS-related autoimmune diseases.
Collapse
|
721
|
Adang LA, Frank DB, Gilani A, Takanohashi A, Ulrick N, Collins A, Cross Z, Galambos C, Helman G, Kanaan U, Keller S, Simon D, Sherbini O, Hanna BD, Vanderver AL. Aicardi goutières syndrome is associated with pulmonary hypertension. Mol Genet Metab 2018; 125:351-358. [PMID: 30219631 PMCID: PMC6880931 DOI: 10.1016/j.ymgme.2018.09.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 09/06/2018] [Accepted: 09/06/2018] [Indexed: 12/18/2022]
Abstract
While pulmonary hypertension (PH) is a potentially life threatening complication of many inflammatory conditions, an association between Aicardi Goutières syndrome (AGS), a rare genetic cause of interferon (IFN) overproduction, and the development of PH has not been characterized to date. We analyzed the cardiac function of individuals with AGS enrolled in the Myelin Disorders Bioregistry Project using retrospective chart review (n = 61). Additional prospective echocardiograms were obtained when possible (n = 22). An IFN signature score, a marker of systemic inflammation, was calculated through the measurement of mRNA transcripts of type I IFN-inducible genes (interferon signaling genes or ISG). Pathologic analysis was performed as available from autopsy samples. Within our cohort, four individuals were identified to be affected by PH: three with pathogenic gain-of-function mutations in the IFIH1 gene and one with heterozygous TREX1 mutations. All studied individuals with AGS were noted to have elevated IFN signature scores (Mann-Whitney p < .001), with the highest levels in individuals with IFIH1 mutations (Mann-Whitney p < .0001). We present clinical and histologic evidence of PH in a series of four individuals with AGS, a rare interferonopathy. Importantly, IFIH1 and TREX1 may represent a novel cause of PH. Furthermore, these findings underscore the importance of screening all individuals with AGS for PH.
Collapse
Affiliation(s)
- Laura A Adang
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| | - David B Frank
- Division of Cardiology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ahmed Gilani
- Department of Pathology, University of Colorado, Children's Hospital Colorado, Aurora, CO, USA
| | - Asako Takanohashi
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Nicole Ulrick
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Abigail Collins
- Division of Pediatric Neurology, Colorado Children's Hospital, Aurora, CO, USA
| | - Zachary Cross
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Csaba Galambos
- Department of Pathology, University of Colorado, Children's Hospital Colorado, Aurora, CO, USA
| | - Guy Helman
- Murdoch Children's Research Institute, Parkville, Melbourne, Australia
| | - Usama Kanaan
- Division of Pediatric Cardiology, Emory University, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Stephanie Keller
- Division of Pediatric Neurology, Emory University, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Dawn Simon
- Division of pediatric pulmonology, Children's Healthcare of Atlanta, Emory University, Atlanta, GA, USA
| | - Omar Sherbini
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Brian D Hanna
- Division of Cardiology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Adeline L Vanderver
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
722
|
Abstract
PURPOSE OF REVIEW Advances in sequencing techniques and systematic cohort-analysis of patients with autoinflammatory phenotypes have enabled a burst in the recognition of new autoinflammatory diseases and contributed to the description of the mechanisms involved in autoinflammation. This review focuses on new genetic and mechanistic discoveries that have broadened the definition of autoinflammatory diseases in the context of the established landscape, providing new therapeutic opportunities and avenues for further discoveries. RECENT FINDINGS Mechanistic insights of inflammatory diseases open opportunities for new targeted therapies. Advances in high-throughput screening of small-molecule inhibitors accelerate the discovery of new and more specific therapeutic options. Recent evidence establishes IL-18 as a driver of macrophage activation, emerging as a new biomarker and therapeutic target. Finally, the identification of escape of nonsense-mediated decay as the genetic mechanism resulting in a monogenic immune-dysregulatory disease, unveils a possibility for future discoveries. SUMMARY Recent mechanistic findings in autoinflammatory diseases as well as the identification of specific biomarkers and discovery of new diseases, continue to pave the way for ever more specific targeted approaches. These therapies are not only applicable to monogenic autoinflammatory syndromes but also for other diseases in which the same pathways are dysregulated.
Collapse
|
723
|
Malsin ES, Kamp DW. The mitochondria in lung fibrosis: friend or foe? Transl Res 2018; 202:1-23. [PMID: 30036495 DOI: 10.1016/j.trsl.2018.05.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 05/24/2018] [Accepted: 05/27/2018] [Indexed: 02/07/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) and other forms of lung fibrosis are age-associated diseases with increased deposition of mesenchymal collagen that promotes respiratory malfunction and eventual death from respiratory failure. Our understanding of the pathobiology underlying pulmonary fibrosis is incomplete and current therapies available to slow or treat lung fibrosis are limited. Evidence reviewed herein demonstrates key involvement of mitochondrial dysfunction in diverse pulmonary cell populations, including alveolar epithelial cells (AEC), fibroblasts, and macrophages and/or immune cells that collectively advances the development of pulmonary fibrosis. The mitochondria have an important role in regulating whether fibrogenic stimuli results in the return of normal healthy function ("friend") or the development of pulmonary fibrosis ("foe"). In particular, we summarize the evidence suggesting that AEC mitochondrial dysfunction is important in mediating lung fibrosis signaling via mechanisms involving imbalances in the levels of reactive oxygen species, endoplasmic reticulum stress response, mitophagy, apoptosis and/or senescence, and inflammatory signaling. Further, we review the emerging evidence suggesting that dysfunctional mitochondria in AECs and other cell types play crucial roles in modulating nearly all aspects of the 9 hallmarks of aging in the context of pulmonary fibrosis as well as some novel molecular pathways that have recently been identified. Finally, we discuss the potential translational aspects of these studies as well as the key knowledge gaps necessary for better informing our understanding of the pathobiology of the mitochondria in mediating pulmonary fibrosis. We reason that targeting deficient mitochondria-derived pathways may provide innovative future treatment strategies that are urgently needed for lung fibrosis.
Collapse
Affiliation(s)
- Elizabeth S Malsin
- Department of Medicine, Division of Pulmonary & Critical Care Medicine, Jesse Brown VA Medical Center and Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - David W Kamp
- Department of Medicine, Division of Pulmonary & Critical Care Medicine, Jesse Brown VA Medical Center and Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
724
|
STING-mediated type-I interferons contribute to the neuroinflammatory process and detrimental effects following traumatic brain injury. J Neuroinflammation 2018; 15:323. [PMID: 30463579 PMCID: PMC6247615 DOI: 10.1186/s12974-018-1354-7] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 10/30/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) represents a major cause of disability and death worldwide with sustained neuroinflammation and autophagy dysfunction contributing to the cellular damage. Stimulator of interferon genes (STING)-induced type-I interferon (IFN) signalling is known to be essential in mounting the innate immune response against infections and cell injury in the periphery, but its role in the CNS remains unclear. We previously identified the type-I IFN pathway as a key mediator of neuroinflammation and neuronal cell death in TBI. However, the modulation of the type-I IFN and neuroinflammatory responses by STING and its contribution to autophagy and neuronal cell death after TBI has not been explored. METHODS C57BL/6J wild-type (WT) and STING-/- mice (8-10-week-old males) were subjected to controlled cortical impact (CCI) surgery and brains analysed by QPCR, Western blot and immunohistochemical analyses at 2 h or 24 h. STING expression was also analysed by QPCR in post-mortem human brain samples. RESULTS A significant upregulation in STING expression was identified in late trauma human brain samples that was confirmed in wild-type mice at 2 h and 24 h after CCI. This correlated with an elevated pro-inflammatory cytokine profile with increased TNF-α, IL-6, IL-1β and type-I IFN (IFN-α and IFN-β) levels. This expression was suppressed in the STING-/- mice with a smaller lesion volume in the knockout animals at 24 h post CCI. Wild-type mice also displayed increased levels of autophagy markers, LC3-II, p62 and LAMP2 after TBI; however, STING-/- mice showed reduced LAMP2 expression suggesting a role for STING in driving dysfunctional autophagy after TBI. CONCLUSION Our data implicates a detrimental role for STING in mediating the TBI-induced neuroinflammatory response and autophagy dysfunction, potentially identifying a new therapeutic target for reducing cellular damage in TBI.
Collapse
|
725
|
Thompson CD, Matta B, Barnes BJ. Therapeutic Targeting of IRFs: Pathway-Dependence or Structure-Based? Front Immunol 2018; 9:2622. [PMID: 30515152 PMCID: PMC6255967 DOI: 10.3389/fimmu.2018.02622] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 10/25/2018] [Indexed: 12/12/2022] Open
Abstract
The interferon regulatory factors (IRFs) are a family of master transcription factors that regulate pathogen-induced innate and acquired immune responses. Aberration(s) in IRF signaling pathways due to infection, genetic predisposition and/or mutation, which can lead to increased expression of type I interferon (IFN) genes, IFN-stimulated genes (ISGs), and other pro-inflammatory cytokines/chemokines, has been linked to the development of numerous diseases, including (but not limited to) autoimmune and cancer. What is currently lacking in the field is an understanding of how best to therapeutically target these transcription factors. Many IRFs are regulated by post-translational modifications downstream of pattern recognition receptors (PRRs) and some of these modifications lead to activation or inhibition. We and others have been able to utilize structural features of the IRFs in order to generate dominant negative mutants that inhibit function. Here, we will review potential therapeutic strategies for targeting all IRFs by using IRF5 as a candidate targeting molecule.
Collapse
Affiliation(s)
- Cherrie D Thompson
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, Feinstein Institute for Medical Research, Manhasset, NY, United States
| | - Bharati Matta
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, Feinstein Institute for Medical Research, Manhasset, NY, United States
| | - Betsy J Barnes
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, Feinstein Institute for Medical Research, Manhasset, NY, United States
| |
Collapse
|
726
|
|
727
|
Martinez-Quiles N, Goldbach-Mansky R. Updates on autoinflammatory diseases. Curr Opin Immunol 2018; 55:97-105. [PMID: 30453204 DOI: 10.1016/j.coi.2018.09.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 09/14/2018] [Indexed: 12/14/2022]
Abstract
Autoinflammatory diseases are hyperinflammatory, immune dysregulatory diseases caused by innate immune cells dysregulation that present typically in the perinatal period with systemic and organ-targeted inflammation, but with improved genetic testing and the development of diagnostic criteria, milder and later-onset forms are being detected in adulthood. While the discovery of gain-of-function mutations in innate sensors linked to the production of proinflammatory cytokines provided the bases for anti-cytokine therapies that changed disease and patient outcomes, the field is expanding with the increasing discovery of disease-causing loss-of-function mutations in genes with cellular house-keeping functions that affect cell homeostasis and when dysregulated trigger innate inflammatory pathways. This review focuses on updates on molecular pathways and diseases that cause predominantly IL-1β and Type-I IFN-mediated autoinflammatory diseases.
Collapse
Affiliation(s)
- Narcisa Martinez-Quiles
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine and Gregorio Marañón Health Research Institute, Madrid, Spain; Translational Autoinflammatory Diseases Section, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Raphaela Goldbach-Mansky
- Translational Autoinflammatory Diseases Section, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA.
| |
Collapse
|
728
|
Abstract
PURPOSE OF REVIEW Given the recent increase in the profile and use of Janus kinase inhibitors (JAKinibs) in adult patients with rheumatic diseases, we aimed to review the current evidence accruing for use in paediatric rheumatology patients. RECENT FINDINGS Significant advances have been made in the management of rheumatic diseases in the past two decades. The introduction of biologic agents in both adults and children has provided significant improvements to patient outcomes and led to better quality of life. Moreover, responses to similar agents allude to common effector pathways operating across juvenile and adult synovitis especially. However, inefficacy and intolerance of these agents leads to a subset of children with limited treatment options. Since 2012, Janus kinase (JAK) inhibitors (JAKinibs), a novel group of oral small molecule inhibitors, have demonstrated their efficacy in several forms of adult inflammatory arthritis, such as rheumatoid arthritis (RA) and psoriatic arthritis (PsA). There are hopes that these successes will be transferable to the paediatric population. In the following review, we discuss the development and progress of JAKinibs in this regard.
Collapse
Affiliation(s)
- S. A. Kerrigan
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UG12 8TA UK
| | - I. B. McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UG12 8TA UK
| |
Collapse
|
729
|
Pescarmona R, Belot A, Villard M, Besson L, Lopez J, Mosnier I, Mathieu AL, Lombard C, Garnier L, Frachette C, Walzer T, Viel S. Comparison of RT-qPCR and Nanostring in the measurement of blood interferon response for the diagnosis of type I interferonopathies. Cytokine 2018; 113:446-452. [PMID: 30413290 DOI: 10.1016/j.cyto.2018.10.023] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 10/03/2018] [Accepted: 10/29/2018] [Indexed: 12/25/2022]
Abstract
Type I interferonopathies are characterized by an increase of circulating type I interferon (IFN) concentration. Type I interferonopathies refer to rare Mendelian genetic disorders such as Aicardi-Goutières Syndrome (AGS) as well as more frequent and polygenic auto-immune diseases like systemic lupus erythematosus (SLE). Yet, detection of type I IFN in these patients remains challenging as its amount is usually very low in patients' sera. Thus, the detection of interferon-stimulating genes has been proposed as an alternative for the detection of this cytokine but sensitivy, specificity and predictive values of the assay have not been reported so far. In this study, we propose two different methods based on Nanostring or RT-qPCR to measure in the clinical routine the IFN response, defined as a set of transcripts that are systemically induced by IFNs. The IFN signature is composed of 6 IFN stimulated genes (ISGs) and has a strong predictive value for the diagnosis of type I interferonopathies. The use of this simple test might represent a gold standard for the evaluation of various autoimmune diseases. Moreover, this test could also be used to monitor patients treated with drugs targeting type I IFN pathway. When comparing both methods - Nanostring and qPCR - in terms of analytical performance, they provided similar results but Nanostring was quicker, easier to multiplex, and almost fully-automated, which represent a more reliable assay for the daily clinical practice.
Collapse
Affiliation(s)
- Rémi Pescarmona
- CIRI, Centre International de Recherche en Infectiologie - International Center for Infectiology Research, Lyon, France; Inserm, U1111 Lyon, France; Ecole Normale Supérieure de Lyon, Lyon, France; Université Lyon 1, Lyon, France; CNRS, UMR5308, Lyon, France; Service d'Immunologie biologique, Hospices Civils de Lyon and Université Claude-Bernard Lyon 1, Lyon, France.
| | - Alexandre Belot
- CIRI, Centre International de Recherche en Infectiologie - International Center for Infectiology Research, Lyon, France; Inserm, U1111 Lyon, France; Ecole Normale Supérieure de Lyon, Lyon, France; Université Lyon 1, Lyon, France; CNRS, UMR5308, Lyon, France; Service de néphrologie rhumatologie dermatologie pédiatriques, Hospices Civils de Lyon, Université Claude-Bernard Lyon 1, Lyon, France; Centre de référence des rhumatismes inflammatoires et maladies auto-immunes systémiques rares de l'enfant RAISE, Hôpital Femme Mère Enfant, Bron, France
| | - Marine Villard
- CIRI, Centre International de Recherche en Infectiologie - International Center for Infectiology Research, Lyon, France; Inserm, U1111 Lyon, France; Ecole Normale Supérieure de Lyon, Lyon, France; Université Lyon 1, Lyon, France; CNRS, UMR5308, Lyon, France; Service d'Immunologie biologique, Hospices Civils de Lyon and Université Claude-Bernard Lyon 1, Lyon, France
| | - Laurie Besson
- CIRI, Centre International de Recherche en Infectiologie - International Center for Infectiology Research, Lyon, France; Inserm, U1111 Lyon, France; Ecole Normale Supérieure de Lyon, Lyon, France; Université Lyon 1, Lyon, France; CNRS, UMR5308, Lyon, France
| | - Jonathan Lopez
- Université Lyon 1, Lyon, France; Service de Biochimie et Biologie moléculaire, Hospices Civils de Lyon, Université Claude-Bernard Lyon 1, Lyon, France; CRCL, Centre de Recherche en Cancérologie de Lyon, Lyon, France; Inserm, U1052 Lyon, France; CNRS, U5286 Lyon, France
| | - Isabelle Mosnier
- Service de Biochimie et Biologie moléculaire, Hospices Civils de Lyon, Université Claude-Bernard Lyon 1, Lyon, France
| | - Anne-Laure Mathieu
- CIRI, Centre International de Recherche en Infectiologie - International Center for Infectiology Research, Lyon, France; Inserm, U1111 Lyon, France; Ecole Normale Supérieure de Lyon, Lyon, France; Université Lyon 1, Lyon, France; CNRS, UMR5308, Lyon, France; Centre de référence des rhumatismes inflammatoires et maladies auto-immunes systémiques rares de l'enfant RAISE, Hôpital Femme Mère Enfant, Bron, France
| | - Christine Lombard
- Service d'Immunologie biologique, Hospices Civils de Lyon and Université Claude-Bernard Lyon 1, Lyon, France
| | - Lorna Garnier
- Service d'Immunologie biologique, Hospices Civils de Lyon and Université Claude-Bernard Lyon 1, Lyon, France
| | - Cécile Frachette
- Service de néphrologie rhumatologie dermatologie pédiatriques, Hospices Civils de Lyon, Université Claude-Bernard Lyon 1, Lyon, France
| | - Thierry Walzer
- CIRI, Centre International de Recherche en Infectiologie - International Center for Infectiology Research, Lyon, France; Inserm, U1111 Lyon, France; Ecole Normale Supérieure de Lyon, Lyon, France; Université Lyon 1, Lyon, France; CNRS, UMR5308, Lyon, France; Centre de référence des rhumatismes inflammatoires et maladies auto-immunes systémiques rares de l'enfant RAISE, Hôpital Femme Mère Enfant, Bron, France
| | - Sébastien Viel
- CIRI, Centre International de Recherche en Infectiologie - International Center for Infectiology Research, Lyon, France; Inserm, U1111 Lyon, France; Ecole Normale Supérieure de Lyon, Lyon, France; Université Lyon 1, Lyon, France; CNRS, UMR5308, Lyon, France; Service d'Immunologie biologique, Hospices Civils de Lyon and Université Claude-Bernard Lyon 1, Lyon, France; Centre de référence des rhumatismes inflammatoires et maladies auto-immunes systémiques rares de l'enfant RAISE, Hôpital Femme Mère Enfant, Bron, France
| |
Collapse
|
730
|
Design of amidobenzimidazole STING receptor agonists with systemic activity. Nature 2018; 564:439-443. [PMID: 30405246 DOI: 10.1038/s41586-018-0705-y] [Citation(s) in RCA: 576] [Impact Index Per Article: 82.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 10/15/2018] [Indexed: 02/05/2023]
Abstract
Stimulator of interferon genes (STING) is a receptor in the endoplasmic reticulum that propagates innate immune sensing of cytosolic pathogen-derived and self DNA1. The development of compounds that modulate STING has recently been the focus of intense research for the treatment of cancer and infectious diseases and as vaccine adjuvants2. To our knowledge, current efforts are focused on the development of modified cyclic dinucleotides that mimic the endogenous STING ligand cGAMP; these have progressed into clinical trials in patients with solid accessible tumours amenable to intratumoral delivery3. Here we report the discovery of a small molecule STING agonist that is not a cyclic dinucleotide and is systemically efficacious for treating tumours in mice. We developed a linking strategy to synergize the effect of two symmetry-related amidobenzimidazole (ABZI)-based compounds to create linked ABZIs (diABZIs) with enhanced binding to STING and cellular function. Intravenous administration of a diABZI STING agonist to immunocompetent mice with established syngeneic colon tumours elicited strong anti-tumour activity, with complete and lasting regression of tumours. Our findings represent a milestone in the rapidly growing field of immune-modifying cancer therapies.
Collapse
|
731
|
Abstract
The autoinflammatory diseases encompass approximately 30 monogenic disorders in which inborn errors in the innate immune system lead to episodic systemic inflammation. Largely mediated by dysregulation of myeloid cells, interleukin (IL)-1β, type I interferon, and NF-κB, these disorders have rapidly expanded over the past several years, and increasing numbers of patients identified. Crossover disorders, bridging autoinflammation and immunodeficiency, have recently been described. This article focuses on the clinical presentation of IL-1 and interferon-driven autoinflammatory disorders, and discusses novel diseases with features of immunodeficiency. Approaches to the clinical diagnosis, genetic testing, and treatment of these disorders are addressed.
Collapse
Affiliation(s)
- Lori Broderick
- Division of Allergy, Immunology and Rheumatology, Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive MC 0760, La Jolla, CA 92093, USA.
| |
Collapse
|
732
|
Nathan N, Berdah L, Borensztajn K, Clement A. Chronic interstitial lung diseases in children: diagnosis approaches. Expert Rev Respir Med 2018; 12:1051-1060. [PMID: 30345849 DOI: 10.1080/17476348.2018.1538795] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Introduction: Children interstitial lung disease (chILD) is a heterogeneous group of rare respiratory disorders characterized by inflammatory and fibrotic changes of the lung parenchyma. They include ILD related to exposure/environment insults, ILD related to systemic diseases processes, ILD related to primary lung parenchyma dysfunctions and ILD specific to infancy. Areas covered: This review provides an update on chILD pathophysiology and diagnosis approaches in immunocompetent children. It includes current information on genetic causes. Expert commentary: ChILD covers a large spectrum of entities with heterogeneous disease expression. Various classifications have been reported, but none of them seems completely satisfactory. Recently, progress in molecular genetics has allowed identifying some genetic contributors, with, so far, a lack of correlations between gene disorders and disease expression. Despite improvements in patient management, chILD prognosis is still burdened by significant morbidity and mortality. Ongoing international collaborations will allow gathering larger longitudinal cohorts of patients to improve disease knowledge and personalized care. The overall goal is to help the children with ILD to reach the adulthood transition in a better condition, and to structure genetic counseling for their family.
Collapse
Affiliation(s)
- Nadia Nathan
- a Service de pneumologie pédiatrique, Centre national de référence des maladies respiratoires rares RespiRare , Hôpital Armand Trousseau, Assistance Publique Hôpitaux de Paris (AP-HP) , Paris , France.,b Sorbonne Université and Inserm UMRS933 , Paris , France
| | - Laura Berdah
- a Service de pneumologie pédiatrique, Centre national de référence des maladies respiratoires rares RespiRare , Hôpital Armand Trousseau, Assistance Publique Hôpitaux de Paris (AP-HP) , Paris , France
| | | | - Annick Clement
- a Service de pneumologie pédiatrique, Centre national de référence des maladies respiratoires rares RespiRare , Hôpital Armand Trousseau, Assistance Publique Hôpitaux de Paris (AP-HP) , Paris , France.,b Sorbonne Université and Inserm UMRS933 , Paris , France
| |
Collapse
|
733
|
Alperin JM, Ortiz-Fernández L, Sawalha AH. Monogenic Lupus: A Developing Paradigm of Disease. Front Immunol 2018; 9:2496. [PMID: 30459768 PMCID: PMC6232876 DOI: 10.3389/fimmu.2018.02496] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Accepted: 10/09/2018] [Indexed: 12/13/2022] Open
Abstract
Monogenic lupus is a form of systemic lupus erythematosus (SLE) that occurs in patients with a single gene defect. This rare variant of lupus generally presents with early onset severe disease, especially affecting the kidneys and central nervous system. To date, a significant number of genes have been implicated in monogenic lupus, providing valuable insights into a very complex disease process. Throughout this review, we will summarize the genes reported to be associated with monogenic lupus or lupus-like diseases, and the pathogenic mechanisms affected by the mutations involved upon inducing autoimmunity.
Collapse
Affiliation(s)
- Jessie M Alperin
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Lourdes Ortiz-Fernández
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Amr H Sawalha
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States.,Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
734
|
Abstract
In mammals, cytosolic detection of nucleic acids is critical in initiating innate antiviral responses against invading pathogens (like bacteria, viruses, fungi and parasites). These programs are mediated by multiple cytosolic and endosomal sensors and adaptor molecules (c-GAS/STING axis and TLR9/MyD88 axis, respectively) and lead to the production of type I interferons (IFNs), pro-inflammatory cytokines, and chemokines. While the identity and role of multiple pattern recognition receptors (PRRs) have been elucidated, such immune surveillance systems must be tightly regulated to limit collateral damage and prevent aberrant responses to self- and non-self-nucleic acids. In this review, we discuss recent advances in our understanding of how cytosolic sensing of DNA is controlled during inflammatory immune responses.
Collapse
Affiliation(s)
- Takayuki Abe
- Department of Systems Biology, Columbia University, New York, NY, United States; Department of Microbiology and Immunology, Columbia University, New York, NY, United States
| | - Sagi D Shapira
- Department of Systems Biology, Columbia University, New York, NY, United States; Department of Microbiology and Immunology, Columbia University, New York, NY, United States.
| |
Collapse
|
735
|
Hausmann JS. Targeting cytokines to treat autoinflammatory diseases. Clin Immunol 2018; 206:23-32. [PMID: 30394352 DOI: 10.1016/j.clim.2018.10.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 10/18/2018] [Accepted: 10/23/2018] [Indexed: 01/07/2023]
Abstract
Autoinflammatory diseases are rare group of conditions manifested by recurrent fevers, systemic inflammation, and dysfunctions of the innate immune system. These conditions are characterized by overproduction or lack of inhibition of various cytokines, and the advent of biologic drugs that block specific cytokines involved in these conditions has revolutionized their treatment. In this review, I will discuss the most common autoinflammatory conditions of adulthood including familial Mediterranean fever (FMF), cryopyrin-associated periodic syndrome (CAPS), mevalonate kinase deficiency/hyperimmunoglobulinemia D Syndrome (MKD/HIDS), TNF receptor-associated autoinflammatory syndrome (TRAPS), and systemic juvenile idiopathic arthritis/adult-onset Still's disease (SJIA/AOSD). I will discuss how IL-1, IL-6, IL-18, and TNF play pathogenic roles in these conditions and will review the evidence behind cytokine blockade for these diseases. Throughout the paper, I will reflect on gaps in knowledge of autoinflammatory diseases and will highlight the latest advances and newest drugs in development.
Collapse
Affiliation(s)
- Jonathan S Hausmann
- Autoinflammatory Disease Center, Beth Israel Deaconess Medical Center, 110 Francis Street, Suite 4b, Boston, MA 02215, United States; Autoinflammatory Diseases Clinic, Boston Children's Hospital, 300 Longwood Avenue, Fegan 6, Boston, MA 02115, United States; Harvard Medical School, Boston, United States.
| |
Collapse
|
736
|
Chen Y, Zhou Z, Min W. Mitochondria, Oxidative Stress and Innate Immunity. Front Physiol 2018; 9:1487. [PMID: 30405440 PMCID: PMC6200916 DOI: 10.3389/fphys.2018.01487] [Citation(s) in RCA: 221] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Accepted: 10/02/2018] [Indexed: 12/22/2022] Open
Abstract
Canonical functions of mitochondria include the regulation of cellular survival, orchestration of anabolic and metabolic pathways, as well as reactive oxygen species (ROS) signaling. Recent discoveries, nevertheless, have demonstrated that mitochondria are also critical elements to stimulate innate immune signaling cascade that is able to intensify the inflammation upon cytotoxic stimuli beyond microbial infection. Here we review the expanding research field of mitochondria and oxidative stress in innate immune system to highlight the new mechanistic insights and discuss the pathological relevance of mitochondrial dysregulation induced aberrant innate immune responses in a growing list of sterile inflammatory diseases.
Collapse
Affiliation(s)
- Yuxin Chen
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Zhongyang Zhou
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wang Min
- Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Department of Pathology and the Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
737
|
Allenbach Y, Toquet S, Landon-Cardinal O, Benveniste O. Reply: A child with severe juvenile dermatomyositis treated with ruxolitinib. Brain 2018; 141:e81. [DOI: 10.1093/brain/awy256] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Yves Allenbach
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS_974, Center of Research in Myology, AP-HP, Department of Internal Medicine and Clinical Immunology, DHU I2B, Pitié-Salpêtrière Hospital, Paris, France
| | - Ségolène Toquet
- Department of Internal Medicine, Clinical Immunology and Infectious Diseases, Robert Debré Hospital, University Hospital, Reims, France
| | - Océane Landon-Cardinal
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS_974, Center of Research in Myology, AP-HP, Department of Internal Medicine and Clinical Immunology, DHU I2B, Pitié-Salpêtrière Hospital, Paris, France
| | - Olivier Benveniste
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS_974, Center of Research in Myology, AP-HP, Department of Internal Medicine and Clinical Immunology, DHU I2B, Pitié-Salpêtrière Hospital, Paris, France
| |
Collapse
|
738
|
Wang J, Dai M, Cui Y, Hou G, Deng J, Gao X, Liao Z, Liu Y, Meng Y, Wu L, Yao C, Wang Y, Qian J, Guo Q, Ding H, Qu B, Shen N. Association of Abnormal Elevations in IFIT3 With Overactive Cyclic GMP-AMP Synthase/Stimulator of Interferon Genes Signaling in Human Systemic Lupus Erythematosus Monocytes. Arthritis Rheumatol 2018; 70:2036-2045. [PMID: 29806091 DOI: 10.1002/art.40576] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Accepted: 05/24/2018] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Increasing evidence indicates that the cyclic GMP-AMP synthase/stimulator of interferon genes (cGAS/STING) signaling pathway has a critical pathogenic role in systemic lupus erythematosus (SLE). Expression levels of the interferon (IFN)-inducible gene IFIT3 are elevated in SLE patients. However, it is still not clear how IFIT3 contributes to the pathogenesis of SLE. This study was undertaken to investigate the activation of the cGAS/STING signaling pathway in human SLE monocytes, and to determine how elevated expression of IFIT3 could contribute to overactive cGAS/STING signaling in patients with SLE. METHODS Monocytes from SLE patients or healthy controls were examined for activity of the cGAS/STING signaling pathway and expression levels of IFIT3. Correlations between cGAS/STING signaling activity and SLE clinical features were analyzed. Gain- or loss-of-function experiments were used to determine the role of IFIT3 in cGAS/STING signaling. Coimmunoprecipitation assays were used to identify the interaction between IFIT3 and other proteins. RESULTS The cGAS/STING signaling pathway was found to have enhanced activity in monocytes from SLE patients compared to healthy controls, as indicated by the higher expression of IFNβ downstream. Levels of IFIT3 were significantly elevated in human SLE monocytes, and this was positively correlated with the activity of the cGAS/STING signaling pathway. In vitro, the expression of VACV70-induced IFNβ was reduced by knockdown of IFIT3, whereas overexpression of IFIT3 produced an opposite effect. Finally, IFIT3 was found to interact with both STING and TANK-binding kinase 1. CONCLUSION These findings suggest that IFIT3 is one of the genes that contributes to the overactive cGAS/STING signaling pathway in human SLE monocytes. IFIT3 may therefore serve as a novel therapeutic target for blocking the production of type I IFN and other proinflammatory cytokines by the cGAS/STING signaling pathway in patients with SLE.
Collapse
Affiliation(s)
- Jiehua Wang
- Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Min Dai
- Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yange Cui
- Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Guojun Hou
- Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jun Deng
- Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xin Gao
- Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhuojun Liao
- Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ya Liu
- Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yao Meng
- Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lingling Wu
- Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chao Yao
- Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yan Wang
- Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jie Qian
- Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiang Guo
- Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Huihua Ding
- Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bo Qu
- Shanghai Institute of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Nan Shen
- Shanghai Institute of Rheumatology, and China-Australia Centre for Personalised Immunology, Renji Hospital, School of Medicine, and Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
739
|
Özen S, Sönmez HE, Demir S. Pediatric forms of vasculitis. Best Pract Res Clin Rheumatol 2018; 32:137-147. [PMID: 30526893 DOI: 10.1016/j.berh.2018.09.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 07/23/2018] [Accepted: 09/14/2018] [Indexed: 12/20/2022]
Abstract
Primary vasculitides that affect children are a challenging and complex group of disorders that may involve any system of the body and lead to significant morbidity and mortality. In recent years, there have been significant advances in the field of childhood vasculitides, including the development of classification criteria and outcome assessment. Although some forms of vasculitis occur in both children and adults, considerable differences exist between childhood and adult vasculitides; we review childhood vasculitides, thus highlighting their differences with the adult forms of the disease. We will also discuss monogenic forms of vasculitis, such as deficiency of adenosine deaminase type 2 (DADA2) and haploinsufficiency of A20 (HA20).
Collapse
Affiliation(s)
- Seza Özen
- Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine, Ankara, Turkey.
| | - Hafize Emine Sönmez
- Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Selcan Demir
- Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
740
|
Kato Y, Park J, Takamatsu H, Konaka H, Aoki W, Aburaya S, Ueda M, Nishide M, Koyama S, Hayama Y, Kinehara Y, Hirano T, Shima Y, Narazaki M, Kumanogoh A. Apoptosis-derived membrane vesicles drive the cGAS-STING pathway and enhance type I IFN production in systemic lupus erythematosus. Ann Rheum Dis 2018; 77:1507-1515. [PMID: 29945921 PMCID: PMC6161667 DOI: 10.1136/annrheumdis-2018-212988] [Citation(s) in RCA: 197] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 05/18/2018] [Accepted: 06/01/2018] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Despite the importance of type I interferon (IFN-I) in systemic lupus erythematosus (SLE) pathogenesis, the mechanisms of IFN-I production have not been fully elucidated. Recognition of nucleic acids by DNA sensors induces IFN-I and interferon-stimulated genes (ISGs), but the involvement of cyclic guanosine monophosphate (GMP)-AMP synthase (cGAS) and stimulator of interferon genes (STING) in SLE remains unclear. We studied the role of the cGAS-STING pathway in the IFN-I-producing cascade driven by SLE serum. METHODS We collected sera from patients with SLE (n=64), patients with other autoimmune diseases (n=31) and healthy controls (n=35), and assayed them using a cell-based reporter system that enables highly sensitive detection of IFN-I and ISG-inducing activity. We used Toll-like receptor-specific reporter cells and reporter cells harbouring knockouts of cGAS, STING and IFNAR2 to evaluate signalling pathway-dependent ISG induction. RESULTS IFN-I bioactivity and ISG-inducing activities of serum were higher in patients with SLE than in patients with other autoimmune diseases or healthy controls. ISG-inducing activity of SLE sera was significantly reduced in STING-knockout reporter cells, and STING-dependent ISG-inducing activity correlated with disease activity. Double-stranded DNA levels were elevated in SLE. Apoptosis-derived membrane vesicles (AdMVs) from SLE sera had high ISG-inducing activity, which was diminished in cGAS-knockout or STING-knockout reporter cells. CONCLUSIONS AdMVs in SLE serum induce IFN-I production through activation of the cGAS-STING pathway. Thus, blockade of the cGAS-STING axis represents a promising therapeutic target for SLE. Moreover, our cell-based reporter system may be useful for stratifying patients with SLE with high ISG-inducing activity.
Collapse
Affiliation(s)
- Yasuhiro Kato
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Immunopathology, WPI Immunology Frontier Research Center (iFReC), Osaka University, Osaka, Japan
- Japan Agency for Medical Research and Development – Core Research for Evolutional Science and Technology (AMED–CREST), Osaka University, Osaka, Japan
| | - JeongHoon Park
- Department of Immunopathology, WPI Immunology Frontier Research Center (iFReC), Osaka University, Osaka, Japan
- Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hyota Takamatsu
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Immunopathology, WPI Immunology Frontier Research Center (iFReC), Osaka University, Osaka, Japan
- Japan Agency for Medical Research and Development – Core Research for Evolutional Science and Technology (AMED–CREST), Osaka University, Osaka, Japan
- Japan Science and Technology–Core Research for Evolutional Science and Technology (JST–CREST), Osaka University, Osaka, Japan
| | - Hachirou Konaka
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Immunopathology, WPI Immunology Frontier Research Center (iFReC), Osaka University, Osaka, Japan
- Japan Agency for Medical Research and Development – Core Research for Evolutional Science and Technology (AMED–CREST), Osaka University, Osaka, Japan
| | - Wataru Aoki
- Japan Science and Technology–Core Research for Evolutional Science and Technology (JST–CREST), Osaka University, Osaka, Japan
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Syunsuke Aburaya
- Japan Science and Technology–Core Research for Evolutional Science and Technology (JST–CREST), Osaka University, Osaka, Japan
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Mitsuyoshi Ueda
- Japan Science and Technology–Core Research for Evolutional Science and Technology (JST–CREST), Osaka University, Osaka, Japan
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Masayuki Nishide
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Immunopathology, WPI Immunology Frontier Research Center (iFReC), Osaka University, Osaka, Japan
- Japan Agency for Medical Research and Development – Core Research for Evolutional Science and Technology (AMED–CREST), Osaka University, Osaka, Japan
| | - Shohei Koyama
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Immunopathology, WPI Immunology Frontier Research Center (iFReC), Osaka University, Osaka, Japan
- Japan Agency for Medical Research and Development – Core Research for Evolutional Science and Technology (AMED–CREST), Osaka University, Osaka, Japan
- Japan Science and Technology–Core Research for Evolutional Science and Technology (JST–CREST), Osaka University, Osaka, Japan
| | - Yoshitomo Hayama
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Immunopathology, WPI Immunology Frontier Research Center (iFReC), Osaka University, Osaka, Japan
- Japan Agency for Medical Research and Development – Core Research for Evolutional Science and Technology (AMED–CREST), Osaka University, Osaka, Japan
| | - Yuhei Kinehara
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Immunopathology, WPI Immunology Frontier Research Center (iFReC), Osaka University, Osaka, Japan
- Japan Agency for Medical Research and Development – Core Research for Evolutional Science and Technology (AMED–CREST), Osaka University, Osaka, Japan
| | - Toru Hirano
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Immunopathology, WPI Immunology Frontier Research Center (iFReC), Osaka University, Osaka, Japan
| | - Yoshihito Shima
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Immunopathology, WPI Immunology Frontier Research Center (iFReC), Osaka University, Osaka, Japan
| | - Masashi Narazaki
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Immunopathology, WPI Immunology Frontier Research Center (iFReC), Osaka University, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Immunopathology, WPI Immunology Frontier Research Center (iFReC), Osaka University, Osaka, Japan
- Japan Agency for Medical Research and Development – Core Research for Evolutional Science and Technology (AMED–CREST), Osaka University, Osaka, Japan
- Japan Science and Technology–Core Research for Evolutional Science and Technology (JST–CREST), Osaka University, Osaka, Japan
| |
Collapse
|
741
|
Mensa-Vilaró A, Bravo García-Morato M, de la Calle-Martin O, Franco-Jarava C, Martínez-Saavedra MT, González-Granado LI, González-Roca E, Fuster JL, Alsina L, Mutchinick OM, Balderrama-Rodríguez A, Ramos E, Modesto C, Mesa-Del-Castillo P, Ortego-Centeno N, Clemente D, Souto A, Palmou N, Remesal A, Leslie KS, Gómez de la Fuente E, Yadira Bravo Gallego L, Campistol JM, Dhouib NG, Bejaoui M, Dutra LA, Terreri MT, Mosquera C, González T, Cañellas J, García-Ruiz de Morales JM, Wouters CH, Bosque MT, Cham WT, Jiménez-Treviño S, de Inocencio J, Bloomfield M, Pérez de Diego R, Martínez-Pomar N, Rodríguez-Pena R, González-Santesteban C, Soler-Palacín P, Casals F, Yagüe J, Allende LM, Rodríguez-Gallego JC, Colobran R, Martínez-Martínez L, López-Granados E, Aróstegui JI. Unexpected relevant role of gene mosaicism in patients with primary immunodeficiency diseases. J Allergy Clin Immunol 2018; 143:359-368. [PMID: 30273710 DOI: 10.1016/j.jaci.2018.09.009] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 08/21/2018] [Accepted: 09/08/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND Postzygotic de novo mutations lead to the phenomenon of gene mosaicism. The 3 main types are called somatic, gonadal, and gonosomal mosaicism, which differ in terms of the body distribution of postzygotic mutations. Mosaicism has been reported occasionally in patients with primary immunodeficiency diseases (PIDs) since the early 1990s, but its real involvement has not been systematically addressed. OBJECTIVE We sought to investigate the incidence of gene mosaicism in patients with PIDs. METHODS The amplicon-based deep sequencing method was used in the 3 parts of the study that establish (1) the allele frequency of germline variants (n = 100), (2) the incidence of parental gonosomal mosaicism in families with PIDs with de novo mutations (n = 92), and (3) the incidence of mosaicism in families with PIDs with moderate-to-high suspicion of gene mosaicism (n = 36). Additional investigations evaluated body distribution of postzygotic mutations, their stability over time, and their characteristics. RESULTS The range of allele frequency (44.1% to 55.6%) was established for germline variants. Those with minor allele frequencies of less than 44.1% were assumed to be postzygotic. Mosaicism was detected in 30 (23.4%) of 128 families with PIDs, with a variable minor allele frequency (0.8% to 40.5%). Parental gonosomal mosaicism was detected in 6 (6.5%) of 92 families with de novo mutations, and a high incidence of mosaicism (63.9%) was detected among families with moderate-to-high suspicion of gene mosaicism. In most analyzed cases mosaicism was found to be both uniformly distributed and stable over time. CONCLUSION This study represents the largest performed to date to investigate mosaicism in patients with PIDs, revealing that it affects approximately 25% of enrolled families. Our results might have serious consequences regarding treatment and genetic counseling and reinforce the use of next-generation sequencing-based methods in the routine analyses of PIDs.
Collapse
Affiliation(s)
- Anna Mensa-Vilaró
- Department of Immunology-CDB, Hospital Clínic-IDIBAPS, Barcelona, Spain.
| | | | | | - Clara Franco-Jarava
- Department of Immunology, Hospital Universitari Vall d'Hebron, Vall d'Hebron Research Institute, Barcelona, Spain; Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Barcelona, Spain
| | | | - Luis I González-Granado
- Primary Immunodeficiencies Unit, Department of Pediatrics, Hospital Universitario 12 de Octubre, Madrid, Spain; Instituto de Investigación i+12, Hospital Universitario 12 de Octubre, Madrid, Spain; Universidad Complutense, Madrid, Spain
| | - Eva González-Roca
- Department of Immunology-CDB, Hospital Clínic-IDIBAPS, Barcelona, Spain
| | - Jose Luis Fuster
- Department of Hematology, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Laia Alsina
- Department of Allergy and Clinical Immunology, Hospital Sant Joan de Deu, Esplugues, Spain; Institut de Recerca Pediàtrica Hospital Sant Joan de Déu, Esplugues, Spain
| | - Osvaldo M Mutchinick
- Department of Genetics, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | | | - Eduardo Ramos
- Department of Pediatrics, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Consuelo Modesto
- Department of Pediatric Rheumatology, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Pablo Mesa-Del-Castillo
- Department of Pediatric Rheumatology, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| | | | - Daniel Clemente
- Department of Pediatric Rheumatology, Hospital Universitario Niño Jesús, Madrid, Spain
| | - Alejandro Souto
- Department of Rheumatology, Hospital Clinico Universitario de Santiago, Santiago de Compostela, Spain
| | - Natalia Palmou
- Department of Rheumatology, Hospital Universitario Marques de Valdecilla, Santander, Spain
| | - Agustín Remesal
- Department of Pediatric Rheumatology, Hospital Universitario La Paz, Madrid, Spain
| | - Kieron S Leslie
- Department of Dermatology, University of California San Francisco, San Francisco, Calif
| | | | | | | | - Naouel Guirat Dhouib
- Pediatric Immuno-Hematology Unit, Bone Marrow Transplantation Center, Tunis, Tunisia
| | - Mohamed Bejaoui
- Pediatric Immuno-Hematology Unit, Bone Marrow Transplantation Center, Tunis, Tunisia
| | - Lívia Almeida Dutra
- Division of General Neurology, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
| | - Maria Teresa Terreri
- Department of Pediatrics, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
| | - Catalina Mosquera
- Department of Pediatric Rheumatology, Universidad El Bosque, Bogota, Colombia
| | - Tatiana González
- Department of Pediatric Rheumatology, Universidad de Cartagena, Cartagena, Colombia
| | - Jerónima Cañellas
- Department of Rheumatology, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | | | - Carine H Wouters
- Departments of Pediatric Rheumatology, Microbiology and Immunology, University Hospitals Leuven, KU University of Leuven, Leuven, Belgium
| | - María Teresa Bosque
- Department of Rheumatology, Hospital Clinico Universitario Lozano Blesa, Zaragoza, Spain
| | - Weng Tarng Cham
- Department of Pediatric Rheumatology, Sunway Medical Centre, Kuala Lumpur, Malaysia
| | | | - Jaime de Inocencio
- Instituto de Investigación i+12, Hospital Universitario 12 de Octubre, Madrid, Spain; Department of Pediatric Rheumatology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Markéta Bloomfield
- Department of Immunology, 2(nd) Faculty of Medicine, Charles University and University Hospital in Motol, Prague, Czech Republic
| | - Rebeca Pérez de Diego
- Laboratory of Immunogenetics of Diseases, IdiPAZ Institute for Health Research, Hospital Universitario La Paz, Madrid, Spain; Innate Immunity Group, IdiPAZ Institute for Health Research, Hospital Universitario La Paz, Madrid, Spain
| | | | | | | | - Pere Soler-Palacín
- Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Barcelona, Spain; Pediatric Infectious Diseases and Immunodeficiencies Unit, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca, Barcelona, Spain
| | - Ferran Casals
- Department of Genomics, Universitat Pompeu Fabra, Barcelona, Spain
| | - Jordi Yagüe
- Department of Immunology-CDB, Hospital Clínic-IDIBAPS, Barcelona, Spain
| | - Luis M Allende
- Instituto de Investigación i+12, Hospital Universitario 12 de Octubre, Madrid, Spain; Universidad Complutense, Madrid, Spain; Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | - Roger Colobran
- Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Barcelona, Spain; Immunology Division, Department of Clinical and Molecular Genetics, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca, Barcelona, Spain; Department of Cell Biology, Physiology and Immunology, Autonomous University of Barcelona, Barcelona, Spain
| | | | | | - Juan I Aróstegui
- Department of Immunology-CDB, Hospital Clínic-IDIBAPS, Barcelona, Spain.
| |
Collapse
|
742
|
NLRP3 inflammasome activation in inflammaging. Semin Immunol 2018; 40:61-73. [PMID: 30268598 DOI: 10.1016/j.smim.2018.09.001] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/17/2018] [Accepted: 09/18/2018] [Indexed: 02/06/2023]
Abstract
The process of aging is associated with the appearance of low-grade subclinical inflammation, termed inflammaging, that can accelerate age-related diseases. In Western societies the age-related inflammatory response can additionally be aggravated by an inflammatory response related to modern lifestyles and excess calorie consumption, a pathophysiologic inflammatory response that was coined metaflammation. Here, we summarize the current knowledge of mechanisms that drive both of these processes and focus our discussion the emerging concept that a key innate immune pathway, the NLRP3 inflammasome, is centrally involved in the recognition of triggers that appear during physiological aging and during metabolic stress. We further discuss how these processes are involved in the pathogenesis of common age-related pathologies and highlight potential strategies by which the detrimental inflammatory responses could be pharmacologically addressed.
Collapse
|
743
|
Matz KM, Guzman RM, Goodman AG. The Role of Nucleic Acid Sensing in Controlling Microbial and Autoimmune Disorders. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 345:35-136. [PMID: 30904196 PMCID: PMC6445394 DOI: 10.1016/bs.ircmb.2018.08.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Innate immunity, the first line of defense against invading pathogens, is an ancient form of host defense found in all animals, from sponges to humans. During infection, innate immune receptors recognize conserved molecular patterns, such as microbial surface molecules, metabolites produces during infection, or nucleic acids of the microbe's genome. When initiated, the innate immune response activates a host defense program that leads to the synthesis proteins capable of pathogen killing. In mammals, the induction of cytokines during the innate immune response leads to the recruitment of professional immune cells to the site of infection, leading to an adaptive immune response. While a fully functional innate immune response is crucial for a proper host response and curbing microbial infection, if the innate immune response is dysfunctional and is activated in the absence of infection, autoinflammation and autoimmune disorders can develop. Therefore, it follows that the innate immune response must be tightly controlled to avoid an autoimmune response from host-derived molecules, yet still unencumbered to respond to infection. In this review, we will focus on the innate immune response activated from cytosolic nucleic acids, derived from the microbe or host itself. We will depict how viruses and bacteria activate these nucleic acid sensing pathways and their mechanisms to inhibit the pathways. We will also describe the autoinflammatory and autoimmune disorders that develop when these pathways are hyperactive. Finally, we will discuss gaps in knowledge with regard to innate immune response failure and identify where further research is needed.
Collapse
Affiliation(s)
- Keesha M Matz
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - R Marena Guzman
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Alan G Goodman
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States; Paul G. Allen School for Global Animal Health, College of Veterinary Medicine, Washington State University, Pullman, WA, United States.
| |
Collapse
|
744
|
The classification, genetic diagnosis and modelling of monogenic autoinflammatory disorders. Clin Sci (Lond) 2018; 132:1901-1924. [PMID: 30185613 PMCID: PMC6123071 DOI: 10.1042/cs20171498] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 07/30/2018] [Accepted: 08/07/2018] [Indexed: 12/13/2022]
Abstract
Monogenic autoinflammatory disorders are an increasingly heterogeneous group of conditions characterised by innate immune dysregulation. Improved genetic sequencing in recent years has led not only to the discovery of a plethora of conditions considered to be 'autoinflammatory', but also the broadening of the clinical and immunological phenotypic spectra seen in these disorders. This review outlines the classification strategies that have been employed for monogenic autoinflammatory disorders to date, including the primary innate immune pathway or the dominant cytokine implicated in disease pathogenesis, and highlights some of the advantages of these models. Furthermore, the use of the term 'autoinflammatory' is discussed in relation to disorders that cross the innate and adaptive immune divide. The utilisation of next-generation sequencing (NGS) in this population is examined, as are potential in vivo and in vitro methods of modelling to determine pathogenicity of novel genetic findings. Finally, areas where our understanding can be improved are highlighted, such as phenotypic variability and genotype-phenotype correlations, with the aim of identifying areas of future research.
Collapse
|
745
|
LeMaire SA, Zhang L, Luo W, Ren P, Azares AR, Wang Y, Zhang C, Coselli JS, Shen YH. Effect of Ciprofloxacin on Susceptibility to Aortic Dissection and Rupture in Mice. JAMA Surg 2018; 153:e181804. [PMID: 30046809 PMCID: PMC6233654 DOI: 10.1001/jamasurg.2018.1804] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 04/22/2018] [Indexed: 12/12/2022]
Abstract
Importance Fluoroquinolones are among the most commonly prescribed antibiotics. Recent clinical studies indicated an association between fluoroquinolone use and increased risk of aortic aneurysm and dissection (AAD). This alarming association has raised concern, especially in patients with AAD with risk of rupture and in individuals at risk for developing AAD. Objective To examine the effect of ciprofloxacin on AAD development in mice. Design, Setting, and Participants In a mouse model of moderate, sporadic AAD, 4-week-old male and female C57BL/6J mice were challenged with a high-fat diet and low-dose angiotensin infusion (1000 ng/min/kg). Control unchallenged mice were fed a normal diet and infused with saline. After randomization, challenged and unchallenged mice received ciprofloxacin (100 mg/kg/d) or vehicle through daily gavage during angiotensin or saline infusion. Aortic aneurysm and dissection development and aortic destruction were compared between mice. The direct effects of ciprofloxacin on aortic smooth muscle cells were examined in cultured cells. Results No notable aortic destruction was observed in unchallenged mice that received ciprofloxacin alone. Aortic challenge induced moderate aortic destruction with development of AAD in 17 of 38 mice (45%) and severe AAD in 9 (24%) but no rupture or death. However, challenged mice that received ciprofloxacin had severe aortic destruction and a significantly increased incidence of AAD (38 of 48 [79%]; P = .001; χ2 = 10.9), severe AAD (32 of 48 [67%]; P < .001; χ2 = 15.7), and rupture and premature death (7 of 48 [15%]; P = .01; χ2 = 6.0). The increased AAD incidence was observed in different aortic segments and was similar between male and female mice. Compared with aortic tissues from challenged control mice, those from challenged mice that received ciprofloxacin showed decreased expression of lysyl oxidase, an enzyme that is critical in the assembly and stabilization of elastic fibers and collagen. These aortas also showed increased matrix metalloproteinase levels and activity, elastic fiber fragmentation, and aortic cell injury. In cultured smooth muscle cells, ciprofloxacin treatment significantly reduced lysyl oxidase expression and activity, increased matrix metalloproteinase expression and activity, suppressed cell proliferation, and induced cell death. Furthermore, ciprofloxacin-a DNA topoisomerase inhibitor-caused nuclear and mitochondrial DNA damage and the release of DNA into the cytosol, subsequently inducing mitochondrial dysfunction, reactive oxygen species production, and activation of the cytosolic DNA sensor STING, which we further showed was involved in the suppression of lysyl oxidase expression and induction of matrix metalloproteinase expression. Conclusions and Relevance Ciprofloxacin increases susceptibility to aortic dissection and rupture in a mouse model of moderate, sporadic AAD. Ciprofloxacin should be used with caution in patients with aortic dilatation, as well as in those at high risk for AAD.
Collapse
Affiliation(s)
- Scott A. LeMaire
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
- Department of Cardiovascular Surgery, Texas Heart Institute, Houston
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas
| | - Lin Zhang
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
- Department of Cardiovascular Surgery, Texas Heart Institute, Houston
| | - Wei Luo
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
- Department of Cardiovascular Surgery, Texas Heart Institute, Houston
| | - Pingping Ren
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
- Department of Cardiovascular Surgery, Texas Heart Institute, Houston
| | | | - Yidan Wang
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
- Department of Cardiovascular Surgery, Texas Heart Institute, Houston
| | - Chen Zhang
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
- Department of Cardiovascular Surgery, Texas Heart Institute, Houston
| | - Joseph S. Coselli
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
- Department of Cardiovascular Surgery, Texas Heart Institute, Houston
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas
| | - Ying H. Shen
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas
- Department of Cardiovascular Surgery, Texas Heart Institute, Houston
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
746
|
Nrf2 negatively regulates STING indicating a link between antiviral sensing and metabolic reprogramming. Nat Commun 2018; 9:3506. [PMID: 30158636 PMCID: PMC6115435 DOI: 10.1038/s41467-018-05861-7] [Citation(s) in RCA: 204] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 08/02/2018] [Indexed: 12/25/2022] Open
Abstract
The transcription factor Nrf2 is a critical regulator of inflammatory responses. If and how Nrf2 also affects cytosolic nucleic acid sensing is currently unknown. Here we identify Nrf2 as an important negative regulator of STING and suggest a link between metabolic reprogramming and antiviral cytosolic DNA sensing in human cells. Here, Nrf2 activation decreases STING expression and responsiveness to STING agonists while increasing susceptibility to infection with DNA viruses. Mechanistically, Nrf2 regulates STING expression by decreasing STING mRNA stability. Repression of STING by Nrf2 occurs in metabolically reprogrammed cells following TLR4/7 engagement, and is inducible by a cell-permeable derivative of the TCA-cycle-derived metabolite itaconate (4-octyl-itaconate, 4-OI). Additionally, engagement of this pathway by 4-OI or the Nrf2 inducer sulforaphane is sufficient to repress STING expression and type I IFN production in cells from patients with STING-dependent interferonopathies. We propose Nrf2 inducers as a future treatment option in STING-dependent inflammatory diseases. Understanding how regulators of inflammation affect nucleic acid sensing is important for targeting research against inflammatory diseases and conditions. Here, the authors identify Nrf2 as an important negative regulator of STING and suggest a link between metabolic reprogramming and antiviral cytosolic DNA sensing in human cells.
Collapse
|
747
|
Ozen S, Batu ED. Vasculitis Pathogenesis: Can We Talk About Precision Medicine? Front Immunol 2018; 9:1892. [PMID: 30154798 PMCID: PMC6102378 DOI: 10.3389/fimmu.2018.01892] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 07/31/2018] [Indexed: 12/27/2022] Open
Abstract
Precision medicine is designing the medical care by taking into account the individual variability for each person. We have tried to address whether the existing data may guide precision medicine in primary systemic vasculitides (PSV). We have reviewed genome-wide association studies (GWAS) data, lessons from monogenic mimics of these diseases, and biomarker studies in immunoglobulin A vasculitis/Henoch–Schönlein purpura, Kawasaki disease, anti-neutrophil cytoplasmic antibody-associated vasculitis, polyarteritis nodosa (PAN), Takayasu arteritis, and Behçet’s disease (BD). GWAS provide insights about the pathogenesis of PSV while whole exome sequencing studies lead to discovery of monogenic vasculitides, phenotype of which could mimic other types of vasculitis such as PAN and BD. Monogenic vasculitides form a subgroup of vasculitis which are caused by single gene alterations and discovery of these diseases has enabled more specific therapies in these patients. With increasing number of studies on biomarkers, new targets for treatment appear and better and structured follow-up of PSV patients will become possible. Proteomics and metabolomics studies are required to better categorize our patients with PSV so that we can manage them appropriately and offer more targeted therapy.
Collapse
Affiliation(s)
- Seza Ozen
- Division of Rheumatology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Ezgi Deniz Batu
- Division of Rheumatology, Department of Pediatrics, University of Health Sciences, Ankara Training and Research Hospital, Ankara, Turkey
| |
Collapse
|
748
|
Abstract
Microbial nucleic acids are major signatures of invading pathogens, and their recognition by various host pattern recognition receptors (PRRs) represents the first step toward an efficient innate immune response to clear the pathogens. The nucleic acid-sensing PRRs are localized at the plasma membrane, the cytosol, and/or various cellular organelles. Sensing of nucleic acids and signaling by PRRs involve recruitment of distinct signaling components, and PRRs are intensively regulated by cellular organelle trafficking. PRR-mediated innate immune responses are also heavily regulated by posttranslational modifications, including phosphorylation, polyubiquitination, sumoylation, and glutamylation. In this review, we focus on our current understanding of recognition of microbial nucleic acid by PRRs, particularly on their regulation by organelle trafficking and posttranslational modifications. We also discuss how sensing of self nucleic acids and dysregulation of PRR-mediated signaling lead to serious human diseases.
Collapse
Affiliation(s)
- Ming-Ming Hu
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China; ,
| | - Hong-Bing Shu
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China; ,
| |
Collapse
|
749
|
Nitro-fatty acids are formed in response to virus infection and are potent inhibitors of STING palmitoylation and signaling. Proc Natl Acad Sci U S A 2018; 115:E7768-E7775. [PMID: 30061387 PMCID: PMC6099880 DOI: 10.1073/pnas.1806239115] [Citation(s) in RCA: 200] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Several chronic inflammatory conditions have recently been shown to depend on abnormally high activity of the signaling protein stimulator of IFN genes (STING). These conditions include examples from systemic lupus erythematosus, Aicardi–Goutiéres syndrome, and STING-associated vasculopathy with onset in infancy. The involvement of STING in these diseases points to an unmet demand to identify inhibitors of STING signaling, which could form the basis of anti-STING therapeutics. With this report, we identify distinct endogenously formed lipid species as potent inhibitors of STING signaling—and propose that these lipids could have pharmaceutical potential for treatment of STING-dependent inflammatory diseases. The adaptor molecule stimulator of IFN genes (STING) is central to production of type I IFNs in response to infection with DNA viruses and to presence of host DNA in the cytosol. Excessive release of type I IFNs through STING-dependent mechanisms has emerged as a central driver of several interferonopathies, including systemic lupus erythematosus (SLE), Aicardi–Goutières syndrome (AGS), and stimulator of IFN genes-associated vasculopathy with onset in infancy (SAVI). The involvement of STING in these diseases points to an unmet need for the development of agents that inhibit STING signaling. Here, we report that endogenously formed nitro-fatty acids can covalently modify STING by nitro-alkylation. These nitro-alkylations inhibit STING palmitoylation, STING signaling, and subsequently, the release of type I IFN in both human and murine cells. Furthermore, treatment with nitro-fatty acids was sufficient to inhibit production of type I IFN in fibroblasts derived from SAVI patients with a gain-of-function mutation in STING. In conclusion, we have identified nitro-fatty acids as endogenously formed inhibitors of STING signaling and propose for these lipids to be considered in the treatment of STING-dependent inflammatory diseases.
Collapse
|
750
|
Labrousse M, Kevorkian-Verguet C, Boursier G, Rowczenio D, Maurier F, Lazaro E, Aggarwal M, Lemelle I, Mura T, Belot A, Touitou I, Sarrabay G. Mosaicism in autoinflammatory diseases: Cryopyrin-associated periodic syndromes (CAPS) and beyond. A systematic review. Crit Rev Clin Lab Sci 2018; 55:432-442. [PMID: 30035647 DOI: 10.1080/10408363.2018.1488805] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Autoinflammatory diseases (AIDs) are conditions related to defective regulation of the innate immune system. Sanger sequencing of the causative genes has long been the reference for confirming the diagnosis. However, for many patients with a typical AID phenotype, the genetic cause remains unknown. A pioneering study in 2005 demonstrated mosaicism in patients with cryopyrin-associated periodic syndromes (CAPS); the authors found somatic mosaicism in 69% of their cohort of Sanger-negative CAPS patients. We aim to address the extent to which mosaicism contributes to the etiology of AIDs and its impact on phenotype. We retrieved English-language publications reporting mosaicism in AIDs by querying PubMed with no restriction on date and we surveyed French reference centers. We provide a comprehensive clinical and genetic picture of mosaicism in AIDs. We estimate that the proportion of CAPS-like patients presenting mosaicism ranges from 0.5% to 19%. We also discuss the possible links between the proportion of mutated alleles and various clinical features. This review reevaluates the contribution of mosaic DNA variants in CAPS. Mosaicism may be more common than anticipated in other AIDs. No significant difference was demonstrated between variant allele frequency and clinical phenotype. Such knowledge has implications for the development of guidelines for genetic diagnosis, genetic counseling of affected families and effective patient care.
Collapse
Affiliation(s)
- Marielle Labrousse
- a Department of Medical Genetics, Rare Diseases and Personalized Medicine, CHU Montpellier , Stem Cells, Cellular Plasticity, Regenerative Medicine and Immunotherapies, INSERM, Univ Montpellier , Montpellier , France
| | | | - Guilaine Boursier
- a Department of Medical Genetics, Rare Diseases and Personalized Medicine, CHU Montpellier , Stem Cells, Cellular Plasticity, Regenerative Medicine and Immunotherapies, INSERM, Univ Montpellier , Montpellier , France
| | - Dorota Rowczenio
- c National Amyloidosis Centre , Centre for Amyloidosis and Acute Phase Proteins, Division of Medicine , London , UK
| | - François Maurier
- d Department of Internal Medicine , Hôpitaux Privés , Metz , France
| | - Estibaliz Lazaro
- e Internal Medicine Department , Haut-Lévêque Hospital , Pessac , France
| | - Manjari Aggarwal
- f Pediatric Rheumatology Unit , Centre for Child Health, Sir Ganga Ram Hospital , New Delhi , India
| | - Irène Lemelle
- g Hemato Onco Pediatrics Department , CHRU Nancy , Vandoeuvre les Nancy , France
| | - Thibault Mura
- h Department of Biostatistics , University Hospital of Montpellier , Montpellier , France
| | - Alexandre Belot
- b Pediatric Nephrology, Rheumatology, Dermatology , University of Lyon , Lyon , France
| | - Isabelle Touitou
- a Department of Medical Genetics, Rare Diseases and Personalized Medicine, CHU Montpellier , Stem Cells, Cellular Plasticity, Regenerative Medicine and Immunotherapies, INSERM, Univ Montpellier , Montpellier , France
| | - Guillaume Sarrabay
- a Department of Medical Genetics, Rare Diseases and Personalized Medicine, CHU Montpellier , Stem Cells, Cellular Plasticity, Regenerative Medicine and Immunotherapies, INSERM, Univ Montpellier , Montpellier , France
| |
Collapse
|