601
|
Savic S, Caseley EA, McDermott MF. Moving towards a systems-based classification of innate immune-mediated diseases. NATURE REVIEWS. RHEUMATOLOGY 2020. [PMID: 32107482 DOI: 10.1038/s41584-020-0377-5)] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Autoinflammation as a distinct disease category was first reported in 1999 as a group of monogenic disorders characterized by recurrent episodes of systemic and organ-specific inflammation, known as periodic fever syndromes. Since this original description, the focus has shifted considerably to the inclusion of complex multifactorial conditions with an autoinflammatory basis. Furthermore, the boundaries of what are considered to be autoinflammatory disorders are constantly evolving and currently encompass elements of immunodeficiency and autoimmunity. Notable developments in the intervening 20 years include substantial progress in understanding how the different inflammasomes are activated, how infection is sensed by the innate immune system and how intracellular signalling systems are consequently activated and integrated with many different cellular functions in the autoinflammatory process. With these developments, the field of autoinflammation is moving from a gene-centric view of innate immune-mediated disease towards a systems-based concept, which describes how various convergent pathways, including pyrin and the actin cytoskeleton, protein misfolding and cellular stress, NF-κB dysregulation and interferon activation, contribute to the autoinflammatory process. The development and adoption of a systems-based concept of systemic autoinflammatory diseases is anticipated to have implications for the development of treatments that target specific components of the innate immune system.
Collapse
Affiliation(s)
- Sinisa Savic
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, St James's University Hospital, Leeds, UK. .,National Institute for Health Research-Leeds Biomedical Research Centre, Chapel Allerton Hospital, Leeds, UK. .,Department of Clinical Immunology and Allergy, St James's University Hospital, Leeds, UK.
| | - Emily A Caseley
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, St James's University Hospital, Leeds, UK
| | - Michael F McDermott
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, St James's University Hospital, Leeds, UK.
| |
Collapse
|
602
|
Jing Y, Dai X, Yang L, Kang D, Jiang P, Li N, Cheng J, Li J, Miller H, Ren B, Gong Q, Yin W, Liu Z, Mattila PK, Ning Q, Sun J, Yu B, Liu C. STING couples with PI3K to regulate actin reorganization during BCR activation. SCIENCE ADVANCES 2020; 6:eaax9455. [PMID: 32494627 PMCID: PMC7176427 DOI: 10.1126/sciadv.aax9455] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 01/24/2020] [Indexed: 05/10/2023]
Abstract
The adaptor protein, STING (stimulator of interferon genes), has been rarely studied in adaptive immunity. We used Sting KO mice and a patient's mutated STING cells to study the effect of STING deficiency on B cell development, differentiation, and BCR signaling. We found that STING deficiency promotes the differentiation of marginal zone B cells. STING is involved in BCR activation and negatively regulates the activation of CD19 and Btk but positively regulates the activation of SHIP. The activation of WASP and accumulation of F-actin were enhanced in Sting KO B cells upon BCR stimulation. Mechanistically, STING uses PI3K mediated by the CD19-Btk axis as a central hub for controlling the actin remodeling that, in turn, offers feedback to BCR signaling. Overall, our study provides a mechanism of how STING regulates BCR signaling via feedback from actin reorganization, which contributes to positive regulation of STING on the humoral immune response.
Collapse
Affiliation(s)
- Yukai Jing
- Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Medical Laboratory, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Dai
- Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lu Yang
- Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Danqing Kang
- Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Panpan Jiang
- Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Na Li
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
| | - Jiali Cheng
- Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingwen Li
- Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heather Miller
- Department of Intracellular Pathogens, National Institute of Allergy and Infectious Diseases, Hamilton, MT 59840, USA
| | - Boxu Ren
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
- Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, China
| | - Quan Gong
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
- Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, China
| | - Wei Yin
- Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zheng Liu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pieta K. Mattila
- Institute of Biomedicine, Unit of Pathology, and MediCity Research Laboratories, University of Turku, Turku, Finland
| | - Qin Ning
- Department of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinqiao Sun
- Department of Clinical Immunology, Children’s Hospital of Fudan University, Shanghai, China
| | - Bing Yu
- Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Corresponding author. (B.Y.); (C.L.)
| | - Chaohong Liu
- Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Corresponding author. (B.Y.); (C.L.)
| |
Collapse
|
603
|
Riley JS, Tait SW. Mitochondrial DNA in inflammation and immunity. EMBO Rep 2020; 21:e49799. [PMID: 32202065 PMCID: PMC7132203 DOI: 10.15252/embr.201949799] [Citation(s) in RCA: 543] [Impact Index Per Article: 108.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/31/2020] [Accepted: 03/03/2020] [Indexed: 12/14/2022] Open
Abstract
Mitochondria are cellular organelles that orchestrate a vast range of biological processes, from energy production and metabolism to cell death and inflammation. Despite this seemingly symbiotic relationship, mitochondria harbour within them a potent agonist of innate immunity: their own genome. Release of mitochondrial DNA into the cytoplasm and out into the extracellular milieu activates a plethora of different pattern recognition receptors and innate immune responses, including cGAS‐STING, TLR9 and inflammasome formation leading to, among others, robust type I interferon responses. In this Review, we discuss how mtDNA can be released from the mitochondria, the various inflammatory pathways triggered by mtDNA release and its myriad biological consequences for health and disease.
Collapse
Affiliation(s)
- Joel S Riley
- Cancer Research UK Beatson Institute, Glasgow, UK.,Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Stephen Wg Tait
- Cancer Research UK Beatson Institute, Glasgow, UK.,Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
604
|
Abstract
The technological advances in diagnostics and therapy of primary immunodeficiency are progressing at a fast pace. This review examines recent developments in the field of inborn errors of immunity, from their definition to their treatment. We will summarize the challenges posed by the growth of next-generation sequencing in the clinical setting, touch briefly on the expansion of the concept of inborn errors of immunity beyond the classic immune system realm, and finally review current developments in targeted therapies, stem cell transplantation, and gene therapy.
Collapse
Affiliation(s)
- Giorgia Bucciol
- Inborn Errors of Immunity, Department of Immunology, Microbiology and Transplantation, KU Leuven, Herestraat 49, Leuven, 3000, Belgium.,Childhood Immunology, Department of Pediatrics, University Hospitals Leuven, ERN-RITA Core Member, Herestraat 49, Leuven, 3000, Belgium
| | - Isabelle Meyts
- Inborn Errors of Immunity, Department of Immunology, Microbiology and Transplantation, KU Leuven, Herestraat 49, Leuven, 3000, Belgium.,Childhood Immunology, Department of Pediatrics, University Hospitals Leuven, ERN-RITA Core Member, Herestraat 49, Leuven, 3000, Belgium
| |
Collapse
|
605
|
Demirkaya E, Sahin S, Romano M, Zhou Q, Aksentijevich I. New Horizons in the Genetic Etiology of Systemic Lupus Erythematosus and Lupus-Like Disease: Monogenic Lupus and Beyond. J Clin Med 2020; 9:E712. [PMID: 32151092 PMCID: PMC7141186 DOI: 10.3390/jcm9030712] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/12/2020] [Accepted: 02/21/2020] [Indexed: 02/05/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a clinically and genetically heterogeneous autoimmune disease. The etiology of lupus and the contribution of genetic, environmental, infectious and hormonal factors to this phenotype have yet to be elucidated. The most straightforward approach to unravel the molecular pathogenesis of lupus may rely on studies of patients who present with early-onset severe phenotypes. Typically, they have at least one of the following clinical features: childhood onset of severe disease (<5 years), parental consanguinity, and presence of family history for autoimmune diseases in a first-degree relative. These patients account for a small proportion of patients with lupus but they inform considerable knowledge about cellular pathways contributing to this inflammatory phenotype. In recent years with the aid of new sequencing technologies, novel or rare pathogenic variants have been reported in over 30 genes predisposing to SLE and SLE-like diseases. Future studies will likely discover many more genes with private variants associated to lupus-like phenotypes. In addition, genome-wide association studies (GWAS) have identified a number of common alleles (SNPs), which increase the risk of developing lupus in adult age. Discovery of a possible shared immune pathway in SLE patients, either with rare or common variants, can provide important clues to better understand this complex disorder, it's prognosis and can help guide new therapeutic approaches. The aim of this review is to summarize the current knowledge of the clinical presentation, genetic diagnosis and mechanisms of disease in patents with lupus and lupus-related phenotypes.
Collapse
Affiliation(s)
- Erkan Demirkaya
- Schulich School of Medicine & Dentistry, Department of Paediatrics, Division of Paediatric Rheumatology, University of Western Ontario, London, ON N6A 5W9, Canada;
| | - Sezgin Sahin
- Van Training and Research Hospital, Department of Paediatric Rheumatology, 65000 Van, Turkey;
| | - Micol Romano
- Schulich School of Medicine & Dentistry, Department of Paediatrics, Division of Paediatric Rheumatology, University of Western Ontario, London, ON N6A 5W9, Canada;
- Department of Pediatric Rheumatology, ASST-PINI-CTO, 20122 Milano, Italy
| | - Qing Zhou
- Life Sciences Institute, Zhejiang University, Hang Zhou 310058, China;
| | - Ivona Aksentijevich
- Inflammatory Disease Section, National Human Genome Research Institute, Bethesda, MD 20892, USA;
| |
Collapse
|
606
|
Zhang H, Zeng L, Xie M, Liu J, Zhou B, Wu R, Cao L, Kroemer G, Wang H, Billiar TR, Zeh HJ, Kang R, Jiang J, Yu Y, Tang D. TMEM173 Drives Lethal Coagulation in Sepsis. Cell Host Microbe 2020; 27:556-570.e6. [PMID: 32142632 DOI: 10.1016/j.chom.2020.02.004] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/14/2020] [Accepted: 02/10/2020] [Indexed: 12/14/2022]
Abstract
The discovery of TMEM173/STING-dependent innate immunity has recently provided guidance for the prevention and management of inflammatory disorders. Here, we show that myeloid TMEM173 occupies an essential role in regulating coagulation in bacterial infections through a mechanism independent of type I interferon response. Mechanistically, TMEM173 binding to ITPR1 controls calcium release from the endoplasmic reticulum in macrophages and monocytes. The TMEM173-dependent increase in cytosolic calcium drives Gasdermin D (GSDMD) cleavage and activation, which triggers the release of F3, the key initiator of blood coagulation. Genetic or pharmacological inhibition of the TMEM173-GSDMD-F3 pathway blocks systemic coagulation and improves animal survival in three models of sepsis (cecal ligation and puncture or bacteremia with Escherichia coli or Streptococcus pneumoniae infection). The upregulation of the TMEM173 pathway correlates with the severity of disseminated intravascular coagulation and mortality in patients with sepsis. Thus, TMEM173 is a key regulator of blood clotting during lethal bacterial infections.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Ling Zeng
- Wound Trauma Medical Center, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Min Xie
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jiao Liu
- The Third Affiliated Hospital, Protein Modification and Degradation Lab of Guangzhou and Guangdong, Guangzhou Medical University, Guang Zhou, Guangdong 510600, China
| | - Borong Zhou
- The Third Affiliated Hospital, Protein Modification and Degradation Lab of Guangzhou and Guangdong, Guangzhou Medical University, Guang Zhou, Guangdong 510600, China
| | - Runliu Wu
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lizhi Cao
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Centre de Recherche des Cordeliers, Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, 94800 Villejuif, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, 75015 Paris, France; Suzhou Institute for Systems Medicine, Chinese Academy of Sciences, Suzhou, Jiangsu 215163, China; Department of Women's and Children's Health, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Haichao Wang
- Laboratory of Emergency Medicine, North Shore University Hospital and the Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Herbert J Zeh
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jianxin Jiang
- Wound Trauma Medical Center, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing 400042, China.
| | - Yan Yu
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| | - Daolin Tang
- The Third Affiliated Hospital, Protein Modification and Degradation Lab of Guangzhou and Guangdong, Guangzhou Medical University, Guang Zhou, Guangdong 510600, China; Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
607
|
Sönmez HE, Karaaslan C, de Jesus AA, Batu ED, Anlar B, Sözeri B, Bilginer Y, Karaguzel D, Ayvaz DC, Tezcan I, Goldbach-Mansky R, Ozen S. A clinical score to guide in decision making for monogenic type I IFNopathies. Pediatr Res 2020; 87:745-752. [PMID: 31641281 PMCID: PMC8425764 DOI: 10.1038/s41390-019-0614-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 08/13/2019] [Accepted: 10/01/2019] [Indexed: 01/09/2023]
Abstract
OBJECTIVE To develop a set of clinical criteria that identifies patients with a potential autoinflammatory IFNopathy. METHODS Based on a literature review, a set of clinical criteria identifying genetically confirmed monogenic IFNopathies was selected. For validation, the clinical score was assessed in healthy controls (HCs) and 18 disease controls, including 2 known autoimmune IFNopathies, juvenile systemic lupus erythematosus (JSLE, n = 4) and dermatomyositis (JDM, n = 4); adenosine deaminase 2 deficiency (DADA2, n = 4); and oligoarticular juvenile idiopathic arthritis (oJIA, n = 6). We assessed an IFN score (IRG-S) in whole blood by NanoString using a previously published 28-gene-IRG-S and a reduced 6-gene-IRG-S. RESULTS The 12 patients with a possible IFNopathy had higher clinical scores (3-5) than the patients with sJLE, JDM, DADA2, and oJIA and in HCs. Both the 28-IRG-S and 6-IRG-S were significantly higher in the autoinflammatory IFNopathy patients compared to HCs and oJIA and DADA2 patients but not different from patients with JSLE and JDM. Subsequently, genetic analysis revealed mutations in genes previously reported in genes related to the IFN pathway in 9 of the 12 patients. CONCLUSION We developed a clinical score to identify patients with possible autoinflammatory IFNopathies. A clinical score was associated with a high IRG-S and may serve to identify patients with an autoinflammatory IFNopathy.
Collapse
Affiliation(s)
- Hafize Emine Sönmez
- Division of Rheumatology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Cagatay Karaaslan
- Department of Biology, Molecular Biology Section, Hacettepe University Faculty of Science, Ankara, Turkey
| | - Adriana A. de Jesus
- Translational Autoinflammatory Diseases Section (TADS), Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ezgi Deniz Batu
- Division of Rheumatology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Banu Anlar
- Division of Neurology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Betül Sözeri
- Division of Rheumatology, Department of Pediatrics, Umraniye Research and Training Hospital, Istanbul, Turkey
| | - Yelda Bilginer
- Division of Rheumatology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Dilara Karaguzel
- Department of Biology, Molecular Biology Section, Hacettepe University Faculty of Science, Ankara, Turkey
| | - Deniz Cagdas Ayvaz
- Division of Immunology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Ilhan Tezcan
- Division of Immunology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Raphaela Goldbach-Mansky
- Translational Autoinflammatory Diseases Section (TADS), Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Seza Ozen
- Division of Rheumatology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey.
| |
Collapse
|
608
|
Stolberg NG, Verbsky JW. Autoinflammatory Disorders with Perinatal Onset. Clin Perinatol 2020; 47:41-52. [PMID: 32000928 DOI: 10.1016/j.clp.2019.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Autoinflammatory disorders are rare genetic defects that result in inflammation in the absence of an infectious or autoimmune disease. Although very rare, these disorders can occur in the perinatal period, and recognizing their presentation is important because there are often long-term complications and effective targeted therapies for these disorders. Most of these disorders present with rash, fevers, and laboratory evidence of inflammation. Importantly, these disorders can now be separated into their pathophysiologic mechanisms of action, which can also guide therapies. The article reviews the different mechanisms of autoinflammatory disorders and highlights those disorders that can present in the newborn period.
Collapse
Affiliation(s)
- Nissim G Stolberg
- Division of Rheumatology, Department of Pediatrics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - James W Verbsky
- Division of Rheumatology, Department of Pediatrics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA.
| |
Collapse
|
609
|
Bader V, Winklhofer KF. Mitochondria at the interface between neurodegeneration and neuroinflammation. Semin Cell Dev Biol 2020; 99:163-171. [DOI: 10.1016/j.semcdb.2019.05.028] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/28/2019] [Accepted: 05/29/2019] [Indexed: 12/15/2022]
|
610
|
Moving towards a systems-based classification of innate immune-mediated diseases. Nat Rev Rheumatol 2020; 16:222-237. [DOI: 10.1038/s41584-020-0377-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2020] [Indexed: 02/07/2023]
|
611
|
Massa D, Baran M, Bengoechea JA, Bowie AG. PYHIN1 regulates pro-inflammatory cytokine induction rather than innate immune DNA sensing in airway epithelial cells. J Biol Chem 2020; 295:4438-4450. [PMID: 32102850 PMCID: PMC7135979 DOI: 10.1074/jbc.ra119.011400] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 01/23/2020] [Indexed: 12/16/2022] Open
Abstract
Animal cells use pattern-recognition receptors (PRRs) to detect specific pathogens. Pathogen detection mounts an appropriate immune response, including interferon and cytokine induction. The intracellular PRR-signaling pathways that detect DNA viruses have been characterized, particularly in myeloid cells. In these pathways, cGMP-AMP synthase (cGAS) and the pyrin and HIN domain family member (PYHIN) protein interferon-γ–inducible protein 16 (IFI16) detect DNA and signal via stimulator of interferon genes protein (STING). However, although airway epithelial cells are frontline sentinels in detecting pathogens, information on how they respond to DNA viruses is limited, and the roles of PYHIN proteins in these cells are unknown. Here, we examined expression and activities of cGAS, STING, and PYHINs in human lung epithelial cells. A549 epithelial cells, commonly used for RNA-sensing studies, failed to respond to DNA because they lacked STING expression, and ectopic STING expression restored a cGAS-dependent DNA response in these cells. In contrast, NuLi-1 immortalized human bronchial epithelial cells did express STING, which was activated after DNA stimulation and mediated DNA-dependent gene induction. PYHIN1, which like IFI16 has been proposed to be a viral DNA sensor, was the only PYHIN protein expressed in both airway epithelial cell types. However, rather than having a role in DNA sensing, PYHIN1 induced proinflammatory cytokines in response to interleukin-1 (IL-1) or tumor necrosis factor α (TNFα) stimulation. Of note, PYHIN1, via its HIN domain, directly induced IL-6 and TNFα transcription, revealing that PYHIN proteins play a role in proinflammatory gene induction in airway epithelial cells.
Collapse
Affiliation(s)
- Davide Massa
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Marcin Baran
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Jose A Bengoechea
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Andrew G Bowie
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
612
|
Imanishi T, Saito T. T Cell Co-stimulation and Functional Modulation by Innate Signals. Trends Immunol 2020; 41:200-212. [PMID: 32035763 DOI: 10.1016/j.it.2020.01.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 01/10/2020] [Accepted: 01/10/2020] [Indexed: 12/22/2022]
Abstract
Pattern recognition receptors (PRRs), such as Toll-like receptors (TLRs), NOD-like receptors (NLRs), and RIG-I-like receptors (RLRs), play a pivotal role in the initiation of innate immune responses. Certain PRRs are also expressed by CD4+ and CD8+ T cells, where they function to provide co-stimulatory signals for their activation and differentiation. Recently, stimulator of interferon genes (STING) was found to be highly expressed in CD4+ and CD8+ T cells and to modulate T cell function. STING signaling inhibits cell growth and stimulates type I interferon (IFN-I) responses in T cells through reciprocal regulation between T cell receptor (TCR) and STING signals. Here, we propose a model whereby innate signals by TLRs and STING regulate TCR signals and T cell functions.
Collapse
Affiliation(s)
- Takayuki Imanishi
- Laboratory for Cell Signaling, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Kanagawa 230-0045, Japan.
| | - Takashi Saito
- Laboratory for Cell Signaling, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Kanagawa 230-0045, Japan; Laboratory for Cell Signaling, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
613
|
Yu ZX, Song HM. Toward a better understanding of type I interferonopathies: a brief summary, update and beyond. World J Pediatr 2020; 16:44-51. [PMID: 31377974 DOI: 10.1007/s12519-019-00273-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/24/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUNDS Type I interferonopathy is a group of autoinflammatory disorders associated with prominent enhanced type I interferon signaling. The mechanisms are complex, and the clinical phenotypes are diverse. This review briefly summarized the recent progresses of type I interferonopathy focusing on the clinical and molecular features, pathogeneses, diagnoses and potential therapies. DATA SOURCES Original research articles and literature reviews published in PubMed-indexed journals. RESULTS Type I interferonopathies include Aicardi-Goutières syndrome, spondyloenchondro-dysplasia with immune dysregulation, stimulator of interferon genes-associated vasculopathy with onset in infancy, X-linked reticulate pigmentary disorder, ubiquitin-specific peptidase 18 deficiency, chronic atypical neutrophilic dermatitis with lipodystrophy, and Singleton-Merten syndrome originally. Other disorders including interferon-stimulated gene 15 deficiency and DNAse II deficiency are believed to be interferonopathies as well. Intracranial calcification, skin vasculopathy, interstitial lung disease, failure to thrive, skeletal development problems and autoimmune features are common. Abnormal responses to nucleic acid stimuli and defective regulation of protein degradation are main mechanisms in disease pathogenesis. First generation Janus kinase inhibitors including baricitinib, tofacitinib and ruxolitinib are useful for disease control. Reverse transcriptase inhibitors seem to be another option for Aicardi-Goutières syndrome. CONCLUSIONS Tremendous progress has been made for the discovery of type I interferonopathies and responsible genes. Janus kinase inhibitors and other agents have potential therapeutic roles. Future basic, translational and clinical studies towards disease monitoring and powerful therapies are warranted.
Collapse
Affiliation(s)
- Zhong-Xun Yu
- Department of Pediatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Hong-Mei Song
- Department of Pediatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
614
|
Krutzke S, Rietschel C, Horneff G. Baricitinib in therapy of COPA syndrome in a 15-year-old girl. Eur J Rheumatol 2020; 7:S78-S81. [PMID: 31449490 PMCID: PMC7004263 DOI: 10.5152/eurjrheum.2019.18177] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 03/24/2019] [Indexed: 12/18/2022] Open
Abstract
COPA syndrome is a newly discovered hereditary immunodeficiency affecting the lung, kidneys, and joints. The mutated gene encodes the α subunit of the coatomer complex I, a protein transporter from the Golgi back to the endoplasmic reticulum. The impaired return of proteins leads to intracellular stress. The syndrome is an autoimmune and autoinflammatory disease that can be grouped among the interferonopathies. The knowledge about COPA syndrome and its treatment is still limited. In this paper, we describe an additional patient, a 15-year-old girl with rheumatoid factor-positive polyarthritis and rheumatoid nodules since the age of 2, who developed interstitial lung disease. The detected mutation c.698G>A was causing the disease. The patient presented with symmetric polyarthritis on wrists, fingers, and hip and ankle joints, with significant functional impairment, and high disease activity. Laboratory parameters demonstrated chronic inflammation, hypergammaglobulinemia, high titre ANA (antinuclear antibodies) and CCP (anti-citrullinated protein) antibodies, and rheumatoid factors. Therapies with various DMARDs (Disease Modifying Anti-Rheumatic Drugs) and biologicals failed. Upon baricitinib application, the clinical activity decreased dramatically with disappearance of joint pain and morning stiffness and significant decrease of joint swelling. A low disease activity was reached after 12 months, with complete disappearance of rheumatoid nodules. In contrast to IL-1 (interleukin-1), IL-6, and TNF (tumor necrosis factor) inhibitors, baricitinib was very successful, probably because baricitinib acts as a JAK-1/2 (janus kinase-1/2) inhibitor in the IFNα/β (inteferone α/β) pathway. A relatively higher dose in children is necessary. COPA syndrome represents a novel disorder of intracellular transport. Reviewing published literature on COPA syndrome, in addition to our patient, there were 31 cases further described.
Collapse
Affiliation(s)
- Sophia Krutzke
- Department of General Paediatrics, Centre for Paediatric Rheumatology, Clinic Sankt Augustin, Sankt Augustin, Germany
| | - Christoph Rietschel
- Department for Pediatric Rheumatology, Clementine Kinderhospital, Frankfurt, Germany
| | - Gerd Horneff
- Department of Pediatric and Adolescents medicine, University Hospital of Cologne, Cologne, Germany
| |
Collapse
|
615
|
Toward a better understanding of type I interferonopathies: a brief summary, update and beyond. WORLD JOURNAL OF PEDIATRICS : WJP 2020. [PMID: 31377974 DOI: 10.1007/s12519-019-00273-z)] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
BACKGROUNDS Type I interferonopathy is a group of autoinflammatory disorders associated with prominent enhanced type I interferon signaling. The mechanisms are complex, and the clinical phenotypes are diverse. This review briefly summarized the recent progresses of type I interferonopathy focusing on the clinical and molecular features, pathogeneses, diagnoses and potential therapies. DATA SOURCES Original research articles and literature reviews published in PubMed-indexed journals. RESULTS Type I interferonopathies include Aicardi-Goutières syndrome, spondyloenchondro-dysplasia with immune dysregulation, stimulator of interferon genes-associated vasculopathy with onset in infancy, X-linked reticulate pigmentary disorder, ubiquitin-specific peptidase 18 deficiency, chronic atypical neutrophilic dermatitis with lipodystrophy, and Singleton-Merten syndrome originally. Other disorders including interferon-stimulated gene 15 deficiency and DNAse II deficiency are believed to be interferonopathies as well. Intracranial calcification, skin vasculopathy, interstitial lung disease, failure to thrive, skeletal development problems and autoimmune features are common. Abnormal responses to nucleic acid stimuli and defective regulation of protein degradation are main mechanisms in disease pathogenesis. First generation Janus kinase inhibitors including baricitinib, tofacitinib and ruxolitinib are useful for disease control. Reverse transcriptase inhibitors seem to be another option for Aicardi-Goutières syndrome. CONCLUSIONS Tremendous progress has been made for the discovery of type I interferonopathies and responsible genes. Janus kinase inhibitors and other agents have potential therapeutic roles. Future basic, translational and clinical studies towards disease monitoring and powerful therapies are warranted.
Collapse
|
616
|
Tang X, Li H, Liu H, Xu H, Yang H, Liu J, Zhao S. Etiologic spectrum of interstitial lung diseases in Chinese children older than 2 years of age. Orphanet J Rare Dis 2020; 15:25. [PMID: 31969166 PMCID: PMC6977247 DOI: 10.1186/s13023-019-1270-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 12/03/2019] [Indexed: 01/08/2023] Open
Abstract
Background Childhood interstitial lung diseases (ILD) (chILD) refer to a rare heterogeneous group of disorders. Global collaborations have been working on the etiologies and classification scheme of chILD. With the development of medical technologies, some new diseases were identified to be associated with chILD and its etiologic spectrum is expanding. The aim of this study is to describe the etiologic spectrum of chILD in children older than 2 years of age and summarize the approaches to diagnosis of chILD. Methods We made a retrospective analysis of children older than 2 years of age with chILD who referred to Beijing Children’s Hospital from 21 provinces all over China from 2013 to 2018. After excluding pulmonary infection, congenital heart disease, bronchopulmonary dysplasia, bronchiolitis obliterans and bronchiectasis, 133 patients were included and categorized by etiology. Clinical manifestations, high-resolution computed tomography, laboratory data, genetic data and pathologic findings were all collected and reviewed. Results Systemic disease associated ILD were the most common causes, accounting for 49.6% of the patients, followed by alveolar structure disorder-associated ILD (27%), exposure related ILD (13.5%), and disorders masquerading as ILD (3.8%). In systemic disease associated ILD, in addition to common etiologies such as vasculitis (10.5%) and connective tissue diseases (9.0%), primary immunodeficiency diseases (PID) associated ILD (9.8%), interstitial pneumonia with autoimmune features (6.8%), and metabolic diseases (6.8%) were not rarely found. Some newly reported etiologies such as STING–associated vasculopathy with onset in infancy, COPA syndrome and STAT3 mutation were included in PID associated ILD. Genetic tests contributed to 15% of the diagnoses which mainly distributed in PID associated ILD, metabolic diseases and surfactant dysfunction disorders, and contributed to the final diagnoses more than lung biopsies (13.5%) and biopsies of rashes or other tissues (12%). Conclusions This study first demonstrated an etiologic spectrum of chILD in Chinese children older than 2 years of age and summarized the approaches to diagnosis. The etiologic spectrum of chILD is expanding with more genetic etiologies being recognized.
Collapse
Affiliation(s)
- Xiaolei Tang
- Department of Respiratory Medicine, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, China, No. 56 Nailishi Road, Xicheng District, Beijing, 100045, China
| | - Huimin Li
- Department of Respiratory Medicine, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, China, No. 56 Nailishi Road, Xicheng District, Beijing, 100045, China
| | - Hui Liu
- Department of Respiratory Medicine, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, China, No. 56 Nailishi Road, Xicheng District, Beijing, 100045, China
| | - Hui Xu
- Department of Respiratory Medicine, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, China, No. 56 Nailishi Road, Xicheng District, Beijing, 100045, China
| | - Haiming Yang
- Department of Respiratory Medicine, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, China, No. 56 Nailishi Road, Xicheng District, Beijing, 100045, China
| | - Jinrong Liu
- Department of Respiratory Medicine, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, China, No. 56 Nailishi Road, Xicheng District, Beijing, 100045, China
| | - Shunying Zhao
- Department of Respiratory Medicine, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, China, No. 56 Nailishi Road, Xicheng District, Beijing, 100045, China.
| |
Collapse
|
617
|
Fragoulis GE, McInnes IB, Siebert S. JAK-inhibitors. New players in the field of immune-mediated diseases, beyond rheumatoid arthritis. Rheumatology (Oxford) 2020; 58:i43-i54. [PMID: 30806709 PMCID: PMC6390879 DOI: 10.1093/rheumatology/key276] [Citation(s) in RCA: 232] [Impact Index Per Article: 46.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 08/03/2018] [Indexed: 02/06/2023] Open
Abstract
Janus kinase (JAK)/signal transducers and activators of transcription (STATs) are a group of molecules associated with one of the major pathways through which many cytokines exert and integrate their function, and as such they are increasingly recognized as playing critical role in the pathogenesis subserving various immune-mediated diseases, including RA, PsA, SpAs, IBD, skin disorders (e.g. alopecia areata, atopic dermatitis), single-gene disorders like interferonopathies, and others. JAKs are the key initiating players of the JAK/STAT pathway. Upon binding of their respective effector molecules (cytokines, IFNs, growth factors and others) to type I and type II receptors, JAKs are activated, and through phosphorylation of themselves and of other molecules (including STATs), they mediate signal transduction to the nucleus. A class of drugs—called JAK inhibitors or JAKinibs—that block one or more JAKs has been developed in the last decade, and now numbers >20 members. Although, so far, JAK inhibitors have been marketed only for RA and PsA, these drugs have been tested in phase 2 and phase 3 clinical trials for other inflammatory conditions and beyond. In this review, we summarize the clinical data, including efficacy and safety, available for JAK inhibitors used in some immune-mediated conditions other than RA.
Collapse
Affiliation(s)
- George E Fragoulis
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Iain B McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Stefan Siebert
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| |
Collapse
|
618
|
Qu H, Li L, Wang TL, Seckin T, Segars J, Shih IM. Epithelial Cells in Endometriosis and Adenomyosis Upregulate STING Expression. Reprod Sci 2020; 27:1276-1284. [PMID: 32046461 DOI: 10.1007/s43032-019-00127-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 11/09/2019] [Indexed: 01/10/2023]
Abstract
In response to cytosolic DNA, stimulator of interferon gene (STING) initiates and orchestrates host's innate immunity by inducing type I interferon. Since endometriosis is a chronic inflammatory disorder, we sought to determine whether STING pathway is activated in ectopic endometrium in comparison to eutopic endometrium. Immunohistochemistry was employed in evaluating the expression levels of STING in normal endometrium, endometriosis, and adenomyosis. The density of CD45+ intraepithelial lymphocytes was correlated with STING expression levels. A total of 39 cases of endometriosis and/or adenomyosis with normal endometrium were analyzed. Among them, 32 had adenomyosis, 26 had endometriosis, and 19 have both lesions. STING protein expression is mainly evident in the cytoplasm of epithelial cells but much less in stromal cells. Based on H-score, we found that the STING expression levels were significantly higher in the epithelial cells of adenomyosis and endometriosis than in eutopic endometrium (132.7 ± 12.20, 119.6 ± 12.57 vs. 19.74 ± 5.96, p < 0.0001). There was no significant difference in STING expression level between endometriosis and adenomyosis. More intraepithelial lymphocytes were detected in endometriosis and adenomyosis lesions than endometrium (5.60 ± 0.70%, 4.95 ± 0.54% vs. 1.25 ± 0.12%, p < 0.0001). A positive correlation between STING expression and intraepithelial lymphocytic infiltrate was observed (p < 0.0001). In summary, STING was upregulated in the epithelium of ectopic endometrium as compared to eutopic endometrium. Its expression levels correlate with the degree of intraepithelial lymphocyte infiltration, suggesting a role in promoting chronic inflammation of ectopic endometrium.
Collapse
Affiliation(s)
- Hong Qu
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Obstetrics and Gynecology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Lihong Li
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Departments of Oncology and Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Tian-Li Wang
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Departments of Oncology and Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Tamer Seckin
- Lenox Hill Hospital and Zucker School of Medicine at Hofstra/Northwell , New York, USA
| | - James Segars
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ie-Ming Shih
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA. .,Departments of Oncology and Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA.
| |
Collapse
|
619
|
Padilla-Salinas R, Sun L, Anderson R, Yang X, Zhang S, Chen ZJ, Yin H. Discovery of Small-Molecule Cyclic GMP-AMP Synthase Inhibitors. J Org Chem 2020; 85:1579-1600. [PMID: 31829590 DOI: 10.1021/acs.joc.9b02666] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cyclic guanosine monophosphate-adenosine monophosphate (GMP-AMP) (cGAS), a cytosolic DNA sensor, plays an important role in the type I interferon response. DNA from either invading microbes or self-origin triggers the enzymatic activity of cGAS. Aberrant activation of cGAS is associated with various autoimmune disorders. Only one selective probe exists for inhibiting cGAS in cells, while others are limited by their poor cellular activity or specificity, which underscores the urgency for discovering new cGAS inhibitors. Here, we describe the development of new small-molecule human cGAS (hcGAS) inhibitors (80 compounds synthesized) with high binding affinity in vitro and cellular activity. Our studies show CU-32 and CU-76 selectively inhibit the DNA pathway in human cells but have no effect on the RIG-I-MAVS or Toll-like receptor pathways. CU-32 and CU-76 represent a new class of hcGAS inhibitors with activity in cells and provide a new chemical scaffold for designing probes to study cGAS function and development of autoimmune therapeutics.
Collapse
Affiliation(s)
- Rosaura Padilla-Salinas
- Department of Biochemistry and BioFrontiers Institute , University of Colorado Boulder , Boulder 80309 , Colorado , United States
| | - Lijun Sun
- Department of Molecular Biology , Howard Hughes Medical Institute , Department of Immunology , and Animal Resource Center , University of Texas Southwestern Medical Center , Dallas 75390-9148 , Texas , United States
| | - Rachel Anderson
- Department of Biochemistry and BioFrontiers Institute , University of Colorado Boulder , Boulder 80309 , Colorado , United States
| | - Xikang Yang
- School of Pharmaceutical Sciences, Tsinghua University-Peking University Joint Center of Life Science , Tsinghua University , Beijing 100082 , China
| | - Shuting Zhang
- School of Pharmaceutical Sciences, Tsinghua University-Peking University Joint Center of Life Science , Tsinghua University , Beijing 100082 , China
| | - Zhijian J Chen
- Department of Molecular Biology , Howard Hughes Medical Institute , Department of Immunology , and Animal Resource Center , University of Texas Southwestern Medical Center , Dallas 75390-9148 , Texas , United States
| | - Hang Yin
- Department of Biochemistry and BioFrontiers Institute , University of Colorado Boulder , Boulder 80309 , Colorado , United States.,School of Pharmaceutical Sciences, Tsinghua University-Peking University Joint Center of Life Science , Tsinghua University , Beijing 100082 , China
| |
Collapse
|
620
|
Dai Y, Liu X, Zhao Z, He J, Yin Q. Stimulator of Interferon Genes-Associated Vasculopathy With Onset in Infancy: A Systematic Review of Case Reports. Front Pediatr 2020; 8:577918. [PMID: 33425809 PMCID: PMC7786402 DOI: 10.3389/fped.2020.577918] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 11/18/2020] [Indexed: 01/07/2023] Open
Abstract
Objective: To summarize and analyze the manifestations of stimulator of interferon genes (STING)-associated vasculopathy with onset in infancy (SAVI). Methods: A systematic literature review was performed including cases from January 1, 2014, to February 1, 2020, using PubMed, OVID, CNKI, and WanFang. This included all the literature containing comparatively complete clinical data. Statistical analysis was performed using SPSS 20.0 to analyze the difference in age of onset, severity of skin lesions, and respiratory symptoms between SAVI patients with p.N154S and p.V155M mutations. Results: A total of 25 papers were included reporting on 51 individuals, of whom 17 had familiar inheritance of their mutation. Patients included 27 males and 24 females, and 8 fatal cases were observed. A total of 10 mutation sites have been reported in the STING gene, with p.V155M being the most prevalent. We identified SAVI as an early-onset disease with a median age of onset of 3 months after birth. Skin lesions were the most common symptoms of SAVI, found in 94.1% (48/51) of patients, while 76% (19/25) who had undergone a skin biopsy showed vasculopathy. Involvement of the lungs was identified in 68.6% (35/51) of patients, while only 22.2% (4/18) who had undergone a lung biopsy showed vasculopathy. Of 20 patients, 19 had increased immunoglobulin, mainly IgG. Furthermore, 45.1% (23/51) of patients had a positive low titer or were transiently positive for antinuclear antibodies. Of the 18 patients treated with JAK inhibitors, 6 relapsed and 2 died of acute respiratory failure caused by viral infection. Patients with p.N154S mutation had an earlier disease onset (p = 0.002) and more severe skin lesions (p < 0.001) than those patients with p.V155M mutation. Conclusion: SAVI is an early-onset disease accompanied by skin and lung lesions whose clinical presentation varies among patients with different genotypes. Therapeutic effects of JAK inhibitors are unsatisfactory.
Collapse
Affiliation(s)
- YunFan Dai
- Department of Respiratory, National Children's Medical Center, China National Clinical Research Center for Respiratory Diseases, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - XiuYun Liu
- Department of Respiratory, National Children's Medical Center, China National Clinical Research Center for Respiratory Diseases, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - ZhiPeng Zhao
- Department of Respiratory, National Children's Medical Center, China National Clinical Research Center for Respiratory Diseases, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - JianXin He
- Department of Respiratory, National Children's Medical Center, China National Clinical Research Center for Respiratory Diseases, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - QingQin Yin
- Department of Respiratory, National Children's Medical Center, China National Clinical Research Center for Respiratory Diseases, Beijing Children's Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
621
|
Ablasser A, Hur S. Regulation of cGAS- and RLR-mediated immunity to nucleic acids. Nat Immunol 2020; 21:17-29. [PMID: 31819255 DOI: 10.1038/s41590-019-0556-1] [Citation(s) in RCA: 229] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 10/29/2019] [Indexed: 12/13/2022]
Abstract
Pathogen-derived nucleic acids are crucial signals for innate immunity. Despite the structural similarity between those and host nucleic acids, mammalian cells have been able to evolve powerful innate immune signaling pathways that originate from the detection of cytosolic nucleic acid species, one of the most prominent being the cGAS-STING pathway for DNA and the RLR-MAVS pathway for RNA, respectively. Recent advances have revealed a plethora of regulatory mechanisms that are crucial for balancing the activity of nucleic acid sensors for the maintenance of overall cellular homeostasis. Elucidation of the various mechanisms that enable cells to maintain control over the activity of cytosolic nucleic acid sensors has provided new insight into the pathology of human diseases and, at the same time, offers a rich and largely unexplored source for new therapeutic targets. This Review addresses the emerging literature on regulation of the sensing of cytosolic DNA and RNA via cGAS and RLRs.
Collapse
Affiliation(s)
- Andrea Ablasser
- Global Health Institute, Swiss Federal Institute of Technology, Lausanne, Switzerland.
| | - Sun Hur
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA.
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
622
|
Tang X, Xu H, Zhou C, Peng Y, Liu H, Liu J, Li H, Yang H, Zhao S. STING-Associated Vasculopathy with Onset in Infancy in Three Children with New Clinical Aspect and Unsatisfactory Therapeutic Responses to Tofacitinib. J Clin Immunol 2020. [PMID: 31705453 DOI: 10.1007/s10875-019-00690-9)] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
PURPOSE STING-associated vasculopathy with onset in infancy (SAVI) is a new rare auto-inflammatory disease. The purpose of this study is to report new cases and summarize the manifestations and outcome of SAVI. METHODS We made a retrospective analysis of three pediatric patients diagnosed with SAVI between March 2016 and July 2018 in Beijing Children's Hospital. RESULTS Three patients comprised one boy and two girls. The median age of onset was 4 months. All patients had the same de novo heterozygous mutation (c.463G>A, p. V155M) of TMEM173. All patients presented with interstitial lung disease and one coexisted with diffuse alveolar hemorrhage. Rashes were presented in two patients. Other clinical manifestations include febrile attacks, failure to thrive, arthritis, myositis, cerebrovascular involvement, ureteral calculus, gastroesophageal reflux, and malnutrition. Ground-glass opacities were the most common features of chest computed tomography, followed with cysts and reticular opacities. Transbronchial lung biopsy was performed in one patient revealing pulmonary vasculitis. Skin biopsy was performed in one patient with changes of vasculitis. All patients were treated with corticosteroids and two patients received combined treatment of tofacitinib. The therapeutic effects of tofacitinib were limited on interstitial lung disease in both patients and were poor on rashes in one patient. One patient under the treatment of tofacitinib died. CONCLUSIONS New clinical aspect of diffuse alveolar hemorrhage is first reported to be associated with SAVI. Unsatisfactory therapeutic effects of tofacitinib are observed in this study and further evaluations are needed.
Collapse
Affiliation(s)
- Xiaolei Tang
- Department of Respiratory Medicine, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, No. 56 Nailishi Road, Xicheng District, Beijing, 100045, China
| | - Hui Xu
- Department of Respiratory Medicine, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, No. 56 Nailishi Road, Xicheng District, Beijing, 100045, China
| | - Chunju Zhou
- Department of Pathology, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
| | - Yun Peng
- Department of Radiology, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
| | - Hui Liu
- Department of Respiratory Medicine, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, No. 56 Nailishi Road, Xicheng District, Beijing, 100045, China
| | - Jinrong Liu
- Department of Respiratory Medicine, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, No. 56 Nailishi Road, Xicheng District, Beijing, 100045, China
| | - Huimin Li
- Department of Respiratory Medicine, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, No. 56 Nailishi Road, Xicheng District, Beijing, 100045, China
| | - Haiming Yang
- Department of Respiratory Medicine, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, No. 56 Nailishi Road, Xicheng District, Beijing, 100045, China
| | - Shunying Zhao
- Department of Respiratory Medicine, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, No. 56 Nailishi Road, Xicheng District, Beijing, 100045, China.
| |
Collapse
|
623
|
Li J, An S, Du Z. Familial Interstitial Lung Disease Caused by Mutation of the STING1 Gene. Front Pediatr 2020; 8:543. [PMID: 33014937 PMCID: PMC7505928 DOI: 10.3389/fped.2020.00543] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 07/28/2020] [Indexed: 12/23/2022] Open
Abstract
Mutations that affect the STING1 (TMEM173) gene cause a rare autoinflammatory syndrome, which is known as STING-associated vasculopathy with onset in infancy (SAVI) and which was initially described in 2014 (1). Thus far, only four reports have been conducted regarding families affected with SAVI in the literature. In this article, the clinical, laboratory, and genetic characteristics of two generations (three cases) of SAVI are described. Unlike previously reported cases that were caused by STING1 mutation, the initial and major clinical manifestations of the mentioned cases are largely identified in the lungs with interstitial lung disease (ILD), and the evidence of typical extrapulmonary symptoms of early-onset systemic inflammation (e.g., cutaneous vasculopathy) were minimal except for the proband, who was diagnosed with arthritis 8 years after onset. In addition, a younger sibling showed no symptoms. Such reports are rarely related to mutations in STING1. The proband was examined with bronchoscopy and alveolar lavage to determine the cause. This study emphasizes that, in the clinical assessment of interstitial pneumonia in children, the possibility of STING1 mutation should be considered, especially in patients with arthritis in addition.
Collapse
Affiliation(s)
- Jinying Li
- Department of Pediatrics, Beijing Children's Hospital, Capital Medical University, Beijing, China.,Department of Respiratory, Children's Hospital of Hebei Province, Shijiazhuang, China
| | - Shuhua An
- Department of Respiratory, Children's Hospital of Hebei Province, Shijiazhuang, China
| | - Zhongdong Du
- Department of Pediatrics, Beijing Children's Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
624
|
Luo W, Wang Y, Zhang L, Ren P, Zhang C, Li Y, Azares AR, Zhang M, Guo J, Ghaghada KB, Starosolski ZA, Rajapakshe K, Coarfa C, Li Y, Chen R, Fujiwara K, Abe JI, Coselli JS, Milewicz DM, LeMaire SA, Shen YH. Critical Role of Cytosolic DNA and Its Sensing Adaptor STING in Aortic Degeneration, Dissection, and Rupture. Circulation 2019; 141:42-66. [PMID: 31887080 DOI: 10.1161/circulationaha.119.041460] [Citation(s) in RCA: 189] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Sporadic aortic aneurysm and dissection (AAD), caused by progressive aortic smooth muscle cell (SMC) loss and extracellular matrix degradation, is a highly lethal condition. Identifying mechanisms that drive aortic degeneration is a crucial step in developing an effective pharmacologic treatment to prevent disease progression. Recent evidence has indicated that cytosolic DNA and abnormal activation of the cytosolic DNA sensing adaptor STING (stimulator of interferon genes) play a critical role in vascular inflammation and destruction. Here, we examined the involvement of this mechanism in aortic degeneration and sporadic AAD formation. METHODS The presence of cytosolic DNA in aortic cells and activation of the STING pathway were examined in aortic tissues from patients with sporadic ascending thoracic AAD. The role of STING in AAD development was evaluated in Sting-deficient (Stinggt/gt) mice in a sporadic AAD model induced by challenging mice with a combination of a high-fat diet and angiotensin II. We also examined the direct effects of STING on SMC death and macrophage activation in vitro. RESULTS In human sporadic AAD tissues, we observed the presence of cytosolic DNA in SMCs and macrophages and significant activation of the STING pathway. In the sporadic AAD model, Stinggt/gt mice showed significant reductions in challenge-induced aortic enlargement, dissection, and rupture in both the thoracic and abdominal aortic regions. Single-cell transcriptome analysis revealed that aortic challenge in wild-type mice induced the DNA damage response, the inflammatory response, dedifferentiation and cell death in SMCs, and matrix metalloproteinase expression in macrophages. These changes were attenuated in challenged Stinggt/gt mice. Mechanistically, nuclear and mitochondrial DNA damage in SMCs and the subsequent leak of DNA to the cytosol activated STING signaling, which induced cell death through apoptosis and necroptosis. In addition, DNA from damaged SMCs was engulfed by macrophages in which it activated STING and its target interferon regulatory factor 3, which directly induced matrix metalloproteinase-9 expression. We also found that pharmacologically inhibiting STING activation partially prevented AAD development. CONCLUSIONS Our findings indicate that the presence of cytosolic DNA and subsequent activation of cytosolic DNA sensing adaptor STING signaling represent a key mechanism in aortic degeneration and that targeting STING may prevent sporadic AAD development.
Collapse
Affiliation(s)
- Wei Luo
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, M.Z., J.G., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, J.G., J.S.C., S.A.L., Y.H.S.), Texas Heart Institute, Houston
| | - Yidan Wang
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, M.Z., J.G., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, J.G., J.S.C., S.A.L., Y.H.S.), Texas Heart Institute, Houston
| | - Lin Zhang
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, M.Z., J.G., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, J.G., J.S.C., S.A.L., Y.H.S.), Texas Heart Institute, Houston
| | - Pingping Ren
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, M.Z., J.G., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, J.G., J.S.C., S.A.L., Y.H.S.), Texas Heart Institute, Houston
| | - Chen Zhang
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, M.Z., J.G., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, J.G., J.S.C., S.A.L., Y.H.S.), Texas Heart Institute, Houston
| | - Yanming Li
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, M.Z., J.G., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, J.G., J.S.C., S.A.L., Y.H.S.), Texas Heart Institute, Houston
| | - Alon R Azares
- Molecular Cardiology Research Lab (A.R.A.), Texas Heart Institute, Houston
| | - Michelle Zhang
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, M.Z., J.G., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
| | - Jiao Guo
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, M.Z., J.G., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, J.G., J.S.C., S.A.L., Y.H.S.), Texas Heart Institute, Houston
| | - Ketan B Ghaghada
- Department of Pediatric Radiology, Texas Children's Hospital, Houston (K.B.G., Z.A.S.)
| | | | - Kimal Rajapakshe
- Department of Molecular and Cellular Biology (K.R., C.C.), Baylor College of Medicine, Houston, TX
| | - Cristian Coarfa
- Dan L. Duncan Cancer Center (C.C.), Baylor College of Medicine, Houston, TX
| | - Yumei Li
- Human Genome Sequencing Center (Yumei Li, R.C.), Baylor College of Medicine, Houston, TX
| | - Rui Chen
- Department of Biochemistry and Molecular Biology (R.C.), Baylor College of Medicine, Houston, TX
- Department of Molecular and Human Genetics (R.C.), Baylor College of Medicine, Houston, TX
- Human Genome Sequencing Center (Yumei Li, R.C.), Baylor College of Medicine, Houston, TX
| | - Keigi Fujiwara
- Department of Biostatistics and Division of Internal Medicine, Department of Cardiology Research, The University of Texas MD Anderson Cancer Center, Houston (K.F., J.A.)
| | - Jun-Ichi Abe
- Department of Biostatistics and Division of Internal Medicine, Department of Cardiology Research, The University of Texas MD Anderson Cancer Center, Houston (K.F., J.A.)
| | - Joseph S Coselli
- Cardiovascular Research Institute (J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, M.Z., J.G., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, J.G., J.S.C., S.A.L., Y.H.S.), Texas Heart Institute, Houston
| | - Dianna M Milewicz
- Division of Medical Genetics, Department of Internal Medicine, The University of Texas Health Science Center at Houston (D.M.M.)
| | - Scott A LeMaire
- Cardiovascular Research Institute (J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, M.Z., J.G., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, J.G., J.S.C., S.A.L., Y.H.S.), Texas Heart Institute, Houston
| | - Ying H Shen
- Cardiovascular Research Institute (J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, M.Z., J.G., J.S.C., S.A.L., Y.H.S.), Baylor College of Medicine, Houston, TX
- Department of Cardiovascular Surgery (W.L., Y.W., L.Z., P.R., C.Z., Yanming Li, J.G., J.S.C., S.A.L., Y.H.S.), Texas Heart Institute, Houston
| |
Collapse
|
625
|
Marino A, Tirelli F, Giani T, Cimaz R. Periodic fever syndromes and the autoinflammatory diseases (AIDs). J Transl Autoimmun 2019; 3:100031. [PMID: 32743516 PMCID: PMC7388371 DOI: 10.1016/j.jtauto.2019.100031] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 12/13/2019] [Accepted: 12/13/2019] [Indexed: 12/14/2022] Open
Abstract
Innate immune system represents the ancestral defense against infectious agents preserved along the evolution and species; it is phylogenetically older than the adaptive immune system, which exists only in the vertebrates. Cells with phagocytic activity such as neutrophils, macrophages, and natural killer (NK) cells play a key role in innate immunity. In 1999 Kastner et al. first introduced the term “autoinflammation” describing two diseases characterized by recurrent episodes of systemic inflammation without any identifiable infectious trigger: Familial Mediterranean Fever (FMF) and TNF Receptor Associated Periodic Syndrome (TRAPS). Autoinflammatory diseases (AIDs) are caused by self-directed inflammation due to an alteration of innate immunity leading to systemic inflammatory attacks typically in an on/off mode. In addition to inflammasomopathies, nuclear factor (NF)-κB-mediated disorders (also known as Rhelopathies) and type 1 interferonopathies are subjects of more recent studies. This review aims to provide an overview of the field with the most recent updates (see “Most recent developments in..” paragraphs) and a description of the newly identified AIDs. Autoinflammatory diseases are caused by self-directed inflammation. Alteration of innate immunity leads to systemic inflammation attacks. The autoinflammatory field is exponentially expanding. The advances in AIDs have led to new insights into immune system understanding. Autoimmunity and autoinflammation features may be simultaneously present.
Collapse
Affiliation(s)
- Achille Marino
- Department of Pediatrics, Desio Hospital, ASST Monza, Desio, MB, Italy.,Biomedical Sciences, University of Florence, Florence, Italy
| | - Francesca Tirelli
- Rheumatology Unit, Meyer Children's Hospital, University of Florence, Florence, Italy
| | - Teresa Giani
- Rheumatology Unit, Meyer Children's Hospital, University of Florence, Florence, Italy.,Department of Medical Biotechnology, University of Siena, Siena, Italy
| | - Rolando Cimaz
- Department of Clinical Sciences and Community Health, University of Milano, Milan, Italy
| |
Collapse
|
626
|
Demirkaya E, Arici ZS, Romano M, Berard RA, Aksentijevich I. Current State of Precision Medicine in Primary Systemic Vasculitides. Front Immunol 2019; 10:2813. [PMID: 31921111 PMCID: PMC6927998 DOI: 10.3389/fimmu.2019.02813] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 11/15/2019] [Indexed: 12/12/2022] Open
Abstract
Precision medicine (PM) is an emerging data-driven health care approach that integrates phenotypic, genomic, epigenetic, and environmental factors unique to an individual. The goal of PM is to facilitate diagnosis, predict effective therapy, and avoid adverse reactions specific for each patient. The forefront of PM is in oncology; nonetheless, it is developing in other fields of medicine, including rheumatology. Recent studies on elucidating the genetic architecture of polygenic and monogenic rheumatological diseases have made PM possible by enabling physicians to customize medical treatment through the incorporation of clinical features and genetic data. For complex inflammatory disorders, the prevailing paradigm is that disease susceptibility is due to additive effects of common reduced-penetrance gene variants and environmental factors. Efforts have been made to calculate cumulative genetic risk score (GRS) and to relate specific susceptibility alleles for use of target therapies. The discovery of rare patients with single-gene high-penetrance mutations informed our understanding of pathways driving systemic inflammation. Here, we review the advances in practicing PM in patients with primary systemic vasculitides (PSVs). We summarize recent genetic studies and discuss current knowledge on the contribution of epigenetic factors and extracellular vesicles (EVs) in disease progression and treatment response. Implementation of PM in PSVs is a developing field that will require analysis of a large cohort of patients to validate data from genomics, transcriptomics, metabolomics, proteomics, and epigenomics studies for accurate disease profiling. This multi-omics approach to study disease pathogeneses should ultimately provide a powerful tool for stratification of patients to receive tailored optimal therapies and for monitoring their disease activity.
Collapse
Affiliation(s)
- Erkan Demirkaya
- Division of Paediatric Rheumatology, Department of Paediatrics, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Zehra Serap Arici
- Department of Paediatric Rheumatology, Sanliurfa Training and Research Hospital, Sanliurfa, Turkey
| | - Micol Romano
- Division of Paediatric Rheumatology, Department of Paediatrics, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada.,Department of Pediatric Rheumatology, Istituto Ortopedico Gaetano Pini, Milan, Italy
| | - Roberta Audrey Berard
- Division of Paediatric Rheumatology, Department of Paediatrics, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Ivona Aksentijevich
- Inflammatory Disease Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
627
|
Cao Y, Jiang LP. The Challenge of Diagnosing SAVI: Case Studies. PEDIATRIC ALLERGY IMMUNOLOGY AND PULMONOLOGY 2019; 32:167-172. [PMID: 32140288 DOI: 10.1089/ped.2019.1054] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 11/11/2019] [Indexed: 12/17/2022]
Abstract
Background: Stimulator of interferon genes (STING)-associated vasculopathy with onset in infancy (SAVI) was first described in 2014 as a type I interferonopathy resulting from heterozygous mutations in the transmembrane protein 173 (TMEM173) gene. SAVI is characterized by the neonatal onset of systemic inflammation, severe cutaneous vasculopathy, and interstitial lung disease. Janus kinase inhibitors are considered effective therapeutics. We sought to describe 2 patients who were diagnosed with SAVI only at postmortem to increase awareness of this disorder. Methods: Clinical data were collected, and Sanger sequencing of the TMEM173 gene was performed in 2 patients suspected of SAVI. This article reviews details of these cases and lessons learned from clinical review and postmortem studies. Results: Two male children shared similar manifestations, including recurrent skin abscesses in winter, skin lesions, and recurrent respiratory tract infections, since birth. Computed tomography of the chest revealed pulmonary fibrosis, but no mutations in relevant genes (including ABCA3 and SFTPC) were discovered in patient 1 (P1). Joint pain was significant in P2 and he was diagnosed with arthritis. Antibiotic treatment yielded little improvement and did not prevent progression. Finally, P1 and P2 died of respiratory and circulatory failure in 2016 and 2012, respectively. In 2018, mutations (P1: c.463G>A, p.V155M; and P2: c.461A>G, p.N154S) in exon 5 of the TMEM173 gene were discovered, confirming the diagnosis of SAVI. Conclusions: The experience with these 2 patients suggests that SAVI should be considered in children with systemic inflammation, chilblain skin lesions, and pulmonary fibrosis, and TMEM173 gene analysis can be beneficial in the diagnosis of SAVI.
Collapse
Affiliation(s)
- Yao Cao
- Clinical Immunology Laboratory, Pediatric Research Institute, Chongqing Key Laboratory of Child Infection and Immunity, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Li-Ping Jiang
- Clinical Immunology Laboratory, Pediatric Research Institute, Chongqing Key Laboratory of Child Infection and Immunity, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, P.R. China
| |
Collapse
|
628
|
Xi Q, Wang M, Jia W, Yang M, Hu J, Jin J, Chen X, Yin D, Wang X. Design, Synthesis, and Biological Evaluation of Amidobenzimidazole Derivatives as Stimulator of Interferon Genes (STING) Receptor Agonists. J Med Chem 2019; 63:260-282. [DOI: 10.1021/acs.jmedchem.9b01567] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Qiumu Xi
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P. R. China
| | - Mingjin Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P. R. China
| | - Wenqiang Jia
- Department of Medicinal Chemistry, Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P. R. China
| | - Mingjian Yang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P. R. China
| | - Jinping Hu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P. R. China
| | - Jing Jin
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P. R. China
| | - Xiaoguang Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P. R. China
| | - Dali Yin
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P. R. China
- Department of Medicinal Chemistry, Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P. R. China
| | - Xiaojian Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P. R. China
- Department of Medicinal Chemistry, Beijing Key Laboratory of Active Substances Discovery and Druggability Evaluation, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P. R. China
| |
Collapse
|
629
|
Zhang H, You QD, Xu XL. Targeting Stimulator of Interferon Genes (STING): A Medicinal Chemistry Perspective. J Med Chem 2019; 63:3785-3816. [DOI: 10.1021/acs.jmedchem.9b01039] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Han Zhang
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qi-Dong You
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiao-Li Xu
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
630
|
Keskitalo S, Haapaniemi E, Einarsdottir E, Rajamäki K, Heikkilä H, Ilander M, Pöyhönen M, Morgunova E, Hokynar K, Lagström S, Kivirikko S, Mustjoki S, Eklund K, Saarela J, Kere J, Seppänen MRJ, Ranki A, Hannula-Jouppi K, Varjosalo M. Novel TMEM173 Mutation and the Role of Disease Modifying Alleles. Front Immunol 2019; 10:2770. [PMID: 31866997 PMCID: PMC6907089 DOI: 10.3389/fimmu.2019.02770] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/12/2019] [Indexed: 02/02/2023] Open
Abstract
Upon binding to pathogen or self-derived cytosolic nucleic acids cyclic GMP-AMP synthase (cGAS) triggers the production of cGAMP that further activates transmembrane protein STING. Upon activation STING translocates from ER via Golgi to vesicles. Monogenic STING gain-of-function mutations cause early-onset type I interferonopathy, with disease presentation ranging from fatal vasculopathy to mild chilblain lupus. Molecular mechanisms underlying the variable phenotype-genotype correlation are presently unclear. Here, we report a novel gain-of-function G207E STING mutation causing a distinct phenotype with alopecia, photosensitivity, thyroid dysfunction, and features of STING-associated vasculopathy with onset in infancy (SAVI), such as livedo reticularis, skin vasculitis, nasal septum perforation, facial erythema, and bacterial infections. Polymorphism in TMEM173 and IFIH1 showed variable penetrance in the affected family, implying contribution to varying phenotype spectrum. The G207E mutation constitutively activates inflammation-related pathways in vitro, and causes aberrant interferon signature and inflammasome activation in patient PBMCs. Treatment with Janus kinase 1 and 2 (JAK1/2) inhibitor baricitinib was beneficiary for a vasculitic ulcer, induced hair regrowth and improved overall well-being in one patient. Protein-protein interactions propose impaired cellular trafficking of G207E mutant. These findings reveal the molecular landscape of STING and propose common polymorphisms in TMEM173 and IFIH1 as likely modifiers of the phenotype.
Collapse
Affiliation(s)
- Salla Keskitalo
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Emma Haapaniemi
- Research Programs Unit, Molecular Neurology and Biomedicum Stem Cell Centre, University of Helsinki, Helsinki, Finland.,Department of Hematology and Regenerative Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Elisabet Einarsdottir
- Molecular Neurology Research Program, University of Helsinki and Folkhälsan Institute of Genetics, Helsinki, Finland.,Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Kristiina Rajamäki
- Faculty of Medicine, University of Helsinki, Clinicum, Helsinki, Finland
| | - Hannele Heikkilä
- Department of Dermatology and Allergology, Skin and Allergy Hospital, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Mette Ilander
- Hematology Research Unit Helsinki, Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland.,Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland
| | - Minna Pöyhönen
- Department of Clinical Genetics, University of Helsinki, Helsinki University Hospital, Helsinki, Finland.,Department of Medical and Clinical Genetics, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
| | - Ekaterina Morgunova
- Department of Hematology and Regenerative Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Kati Hokynar
- Clinical Research Institute HUCH Ltd., Helsinki, Finland
| | - Sonja Lagström
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Sirpa Kivirikko
- Department of Clinical Genetics, University of Helsinki, Helsinki University Hospital, Helsinki, Finland.,Department of Medical and Clinical Genetics, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
| | - Satu Mustjoki
- Hematology Research Unit Helsinki, Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland.,Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland
| | - Kari Eklund
- Faculty of Medicine, University of Helsinki, Clinicum, Helsinki, Finland.,Department of Rheumatology, Helsinki University Hospital, Helsinki, Finland
| | - Janna Saarela
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Juha Kere
- Molecular Neurology Research Program, University of Helsinki and Folkhälsan Institute of Genetics, Helsinki, Finland.,Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden.,School of Basic and Medical Biosciences, King's College London, Guy's Hospital, London, United Kingdom
| | - Mikko R J Seppänen
- Rare Disease Center, Children's Hospital, University of Helsinki, Helsinki University Hospital, Helsinki, Finland.,Immunodeficiency Unit, Inflammation Center, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
| | - Annamari Ranki
- Department of Dermatology and Allergology, Skin and Allergy Hospital, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Katariina Hannula-Jouppi
- Molecular Neurology Research Program, University of Helsinki and Folkhälsan Institute of Genetics, Helsinki, Finland.,Department of Dermatology and Allergology, Skin and Allergy Hospital, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Markku Varjosalo
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
631
|
Wu J, Zhao L, Hu H, Li W, Li Y. Agonists and inhibitors of the STING pathway: Potential agents for immunotherapy. Med Res Rev 2019; 40:1117-1141. [DOI: 10.1002/med.21649] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/15/2019] [Accepted: 11/21/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Jun‐Jun Wu
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical BiologyDepartment of ChemistryTsinghua University Beijing China
| | - Lang Zhao
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical BiologyDepartment of ChemistryTsinghua University Beijing China
| | - Hong‐Guo Hu
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical BiologyDepartment of ChemistryTsinghua University Beijing China
| | - Wen‐Hao Li
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical BiologyDepartment of ChemistryTsinghua University Beijing China
| | - Yan‐Mei Li
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical BiologyDepartment of ChemistryTsinghua University Beijing China
- Beijing Institute for Brain Disorders Beijing China
- Center for Synthetic and Systems BiologyTsinghua University Beijing China
| |
Collapse
|
632
|
Tang X, Shen Y, Zhou C, Yang H, Liu H, Li H, Liu J, Zhao S. Surfactant protein C dysfunction with new clinical insights for diffuse alveolar hemorrhage and autoimmunity. Pediatr Investig 2019; 3:201-206. [PMID: 32851322 PMCID: PMC7331417 DOI: 10.1002/ped4.12162] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 11/12/2019] [Indexed: 12/18/2022] Open
Abstract
IMPORTANCE Surfactant protein C (SP-C) dysfunction is a rare disease associated with interstitial lung disease. Early therapies may improve outcomes but the diagnosis is often delayed owing to variability of manifestations. OBJECTIVE To investigate the manifestations and outcomes of SP-C dysfunction. METHODS We retrospectively analyzed the records of five pediatric patients who were diagnosed with SP-C dysfunction between February 2014 and April 2017 at Beijing Children's Hospital. RESULTS The five patients included two boys and three girls with a median age at diagnosis of 1.3 years. All patients presented with interstitial lung disease and had a heterozygous SFTPC mutation, including an I73T mutation in three patients, a V39L mutation in one patient, and a Y104H mutation in one patient. In addition to common respiratory manifestations, hemoptysis and anemia were observed in one patient with the I73T mutation. Elevated levels of autoantibodies and a large number of hemosiderin-laden macrophages in bronchoalveolar lavage fluid were found in two patients with the I73T mutation, suggesting the presence of diffuse alveolar hemorrage and autoimmunity. Chest high-resolution computed tomography features included ground-glass opacities, reticular opacities, cysts, and pleural thickening. Transbronchial lung biopsy was performed in one patient with the I73T mutation, which revealed the presence of some hemosiderin-laden macrophages in alveolar spaces. All patients received treatment with corticosteroids; two received combined treatment with hydroxychloroquine. During follow-up, the two patients who received hydroxychloroquine showed improved symptoms; of the remaining three patients, two died after their families refused further treatment, while the final patient was lost to follow-up. INTERPRETATION This is the first report to describe a new phenotype of diffuse alveolar hemorrhage with autoimmunity in patients with I73T SFTPC mutation. Treatment with hydroxychloroquine should be considered for patients with SP-C dysfunction.
Collapse
Affiliation(s)
- Xiaolei Tang
- The Second Department of Respiratory MedicineBeijing Children's HospitalCapital Medical UniversityNational Center for Children's HealthBeijingChina
| | - Yuelin Shen
- The Second Department of Respiratory MedicineBeijing Children's HospitalCapital Medical UniversityNational Center for Children's HealthBeijingChina
| | - Chunju Zhou
- Department of PathologyBeijing Children's HospitalCapital Medical UniversityNational Center for Children's HealthBeijingChina
| | - Haiming Yang
- The Second Department of Respiratory MedicineBeijing Children's HospitalCapital Medical UniversityNational Center for Children's HealthBeijingChina
| | - Hui Liu
- The Second Department of Respiratory MedicineBeijing Children's HospitalCapital Medical UniversityNational Center for Children's HealthBeijingChina
| | - Huimin Li
- The Second Department of Respiratory MedicineBeijing Children's HospitalCapital Medical UniversityNational Center for Children's HealthBeijingChina
| | - Jinrong Liu
- The Second Department of Respiratory MedicineBeijing Children's HospitalCapital Medical UniversityNational Center for Children's HealthBeijingChina
| | - Shunying Zhao
- The Second Department of Respiratory MedicineBeijing Children's HospitalCapital Medical UniversityNational Center for Children's HealthBeijingChina
| |
Collapse
|
633
|
Sintim HO, Mikek CG, Wang M, Sooreshjani MA. Interrupting cyclic dinucleotide-cGAS-STING axis with small molecules. MEDCHEMCOMM 2019; 10:1999-2023. [PMID: 32206239 PMCID: PMC7069516 DOI: 10.1039/c8md00555a] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 08/13/2019] [Indexed: 12/19/2022]
Abstract
The cyclic dinucleotide-cGAS-STING axis plays important roles in host immunity. Activation of this signaling pathway, via cytosolic sensing of bacterial-derived c-di-GMP/c-di-AMP or host-derived cGAMP, leads to the production of inflammatory interferons and cytokines that help resolve infection. Small molecule activators of the cGAS-STING axis have the potential to augment immune response against various pathogens or cancer. The aberrant activation of this pathway, due to gain-of-function mutations in any of the proteins that are part of the signaling axis, could lead to various autoimmune diseases. Inhibiting various nodes of the cGAS-STING axis could provide relief to patients with autoimmune diseases. Many excellent reviews on the cGAS-STING axis have been published recently, and these have mainly focused on the molecular details of the cGAS-STING pathway. This review however focuses on small molecules that can be used to modulate various aspects of the cGAS-STING pathway, as well as other parallel inflammatory pathways.
Collapse
Affiliation(s)
- Herman O Sintim
- Department of Chemistry , Purdue University , 560 Oval Drive , West Lafayette , IN 47907 , USA .
- Institute for Drug Discovery , Purdue University , 720 Clinic Drive , West Lafayette , IN 47907 , USA
- Purdue Institute of Inflammation and Infectious Diseases , Purdue University , West Lafayette , IN 47907 , USA
| | - Clinton G Mikek
- Department of Chemistry , Purdue University , 560 Oval Drive , West Lafayette , IN 47907 , USA .
| | - Modi Wang
- Department of Chemistry , Purdue University , 560 Oval Drive , West Lafayette , IN 47907 , USA .
| | - Moloud A Sooreshjani
- Department of Chemistry , Purdue University , 560 Oval Drive , West Lafayette , IN 47907 , USA .
| |
Collapse
|
634
|
Abstract
The pathogenesis of autoinflammatory diseases has shed light on the concept of inflammation in general and on our understanding of the role of the innate immune system. The autoinflammatory diseases have a large spectrum with varying features of inflammation. The most common autoinflammatory diseases are those associated with periodic fevers. The delay in diagnosis of these four common diseases (familial Mediterranean fever, cryopyrin-associated periodic fever syndrome, mevalonate kinase deficiency, and TNF receptor-associated periodic fever syndrome) results in secondary amyloidosis of the kidney. The new work towards classification criteria for these diseases is presented. Recently a group of autoinflammatory diseases that are associated with vasculitis have also been identified. These are stimulators of interferon genes (STING)-associated vasculopathy of infancy (SAVI), which is a monogenic defect associated with excessive activity in interferon alpha and deficiency of adenosine deaminase 2, which is characterized by a polyarteritis nodosa-like picture. These monogenic diseases are now in our differential diagnosis of vasculitides. Secondary amyloidosis is a complication of autoinflammatory diseases. Understanding the inflammatory mechanisms in these diseases has led to the use of targeted biologics for this complication. It is hoped that enlightening the mechanisms underlying these monogenic autoinflammatory diseases will also teach us about the pathways in common diseases.
Collapse
Affiliation(s)
- Seza Ozen
- Department of Pediatrics, Hacettepe University, Ankara, Turkey.
| |
Collapse
|
635
|
Barrat FJ, Crow MK, Ivashkiv LB. Interferon target-gene expression and epigenomic signatures in health and disease. Nat Immunol 2019; 20:1574-1583. [PMID: 31745335 PMCID: PMC7024546 DOI: 10.1038/s41590-019-0466-2] [Citation(s) in RCA: 349] [Impact Index Per Article: 58.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 07/10/2019] [Indexed: 02/06/2023]
Abstract
Multiple type I interferons and interferon-γ (IFN-γ) are expressed under physiological conditions and are increased by stress and infections, and in autoinflammatory and autoimmune diseases. Interferons activate the Jak-STAT signaling pathway and induce overlapping patterns of expression, called 'interferon signatures', of canonical interferon-stimulated genes (ISGs) encoding molecules important for antiviral responses, antigen presentation, autoimmunity and inflammation. It has now become clear that interferons also induce an 'interferon epigenomic signature' by activating latent enhancers and 'bookmarking' chromatin, thus reprogramming cell responses to environmental cues. The interferon epigenomic signature affects ISGs and other gene sets, including canonical targets of the transcription factor NF-κB that encode inflammatory molecules, and is involved in the priming of immune cells, tolerance and the training of innate immune memory. Here we review the mechanisms through which interferon signatures and interferon epigenomic signatures are generated, as well as the expression and functional consequences of these signatures in homeostasis and autoimmune diseases, including systemic lupus erythematosus, rheumatoid arthritis and systemic sclerosis.
Collapse
Affiliation(s)
- Franck J Barrat
- Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY, USA
| | - Mary K Crow
- Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Lionel B Ivashkiv
- Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA.
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY, USA.
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
636
|
Feng X, Liu D, Li Z, Bian J. Bioactive modulators targeting STING adaptor in cGAS-STING pathway. Drug Discov Today 2019; 25:230-237. [PMID: 31758915 DOI: 10.1016/j.drudis.2019.11.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 11/05/2019] [Accepted: 11/14/2019] [Indexed: 12/21/2022]
Abstract
The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING)-pathway triggers innate immune responses by recognizing cytosolic DNA. Recent studies revealed that STING adaptor associates with various diseases, and several modulators targeting STING have been identified including three agonists that have entered clinical trials for treating cancer over the past 2 years. In particular, the efficacy of STING agonists and/or antagonists suggests adaptor STING as a potential therapeutic target for diverse diseases. Herein, we summarize the latest advances in understanding STING functioning and provide an overview of recent STING modulator discoveries, including structural details and the potential therapeutic applications of these modulators.
Collapse
Affiliation(s)
- Xi Feng
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Dongyu Liu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zhiyu Li
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Jinlei Bian
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
637
|
Lian Y, Duffy KJ, Yang J. STING Activation and its Application in Immuno-Oncology. Curr Top Med Chem 2019; 19:2205-2227. [DOI: 10.2174/1568026619666191010155903] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/29/2019] [Accepted: 09/05/2019] [Indexed: 12/19/2022]
Abstract
Recent regulatory approval of several immune checkpoint inhibitors has ushered in a new era
of cancer immunotherapies with the promise of achieving a durable response. This represents a paradigm
shift in cancer treatment from directly targeting tumor cells to harnessing the power of a patient’s
own immune system to destroy them. The cGAS-STING pathway is the major cytosolic dsDNA sensing
pathway that plays a pivotal role in the innate antitumor immune response. With a fundamentally different
mode of action (MOA) than immune checkpoint modulators, STING activation can potentially enhance
tumor immunogenicity and improve patient responses as a single agent or by synergizing with
existing anti-cancer drugs. Therefore, there has been intense interest from the pharmaceutical industry
and academic institutions in the search for potent STING agonists as immunotherapies in oncology. In
this article, we review briefly the cGAS-STING pathway and STING agonists that are in the clinical and
preclinical studies, summarize recently disclosed patent applications and published journal articles in the
field and cover both cyclic dinucleotide (CDN) analogs and non-nucleic acid derived STING agonists.
Collapse
Affiliation(s)
- Yiqian Lian
- Department of Medicinal Chemistry, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, United States
| | - Kevin J. Duffy
- Department of Medicinal Chemistry, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, United States
| | - Jingsong Yang
- Immuno-Oncology and Combinations Research Unit, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, United States
| |
Collapse
|
638
|
Cui X, Zhang R, Cen S, Zhou J. STING modulators: Predictive significance in drug discovery. Eur J Med Chem 2019; 182:111591. [PMID: 31419779 PMCID: PMC7172983 DOI: 10.1016/j.ejmech.2019.111591] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/10/2019] [Accepted: 08/05/2019] [Indexed: 12/19/2022]
Abstract
Cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS) - stimulator of interferon genes (STING) signaling pathway plays the critical role in the immune response to DNA. Pharmacological modulation of the STING pathway has been well characterized both from structural and functional perspectives, which paves the way for the drug design of small modulators by medicinal chemists. Here, we outline recent progress in studies on the STING pathway, the structure and biological function of STING, the STING related disease, as well as the rationale and progress in the development of STING modulators. Our review demonstrates that STING is a promising drug target, and providing clues for the discovery of novel STING agonists and antagonists for the potential treatment of various disease including microbial infectious diseases, cancer, and autoimmune disease.
Collapse
Affiliation(s)
- Xiangling Cui
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Department of Chemistry, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, PR China,Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing, China
| | - Rongyu Zhang
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Department of Chemistry, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, PR China,Drug Development and Innovation Center, College of Chemistry and Life Sciences, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, PR China
| | - Shan Cen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing, China
| | - Jinming Zhou
- Key Laboratory of the Ministry of Education for Advanced Catalysis Materials, Department of Chemistry, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, PR China; Institute of Medicinal Biotechnology, Chinese Academy of Medical Science, Beijing, China; Drug Development and Innovation Center, College of Chemistry and Life Sciences, Zhejiang Normal University, 688 Yingbin Road, Jinhua, 321004, PR China.
| |
Collapse
|
639
|
Hong Y, Nanthapisal S, Omoyinmi E, Olbrich P, Neth O, Speckmann C, Lucena JM, Gilmour K, Worth A, Klein N, Eleftheriou D, Brogan P. Secondary C1q Deficiency in Activated PI3Kδ Syndrome Type 2. Front Immunol 2019; 10:2589. [PMID: 31781101 PMCID: PMC6859795 DOI: 10.3389/fimmu.2019.02589] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 10/18/2019] [Indexed: 11/13/2022] Open
Abstract
Monogenic forms of vasculitis are rare but increasingly recognized. Furthermore, genetic immunodeficiency is increasingly associated with inflammatory immune dysregulatory features, including vasculitis. This case report describes a child of non-consanguineous parents who presented with chronic digital vasculitis early in life, is of short stature, has facial dysmorphia, immunodeficiency (low serum IgA, high serum IgM), recurrent bacterial infections, lymphoproliferation, absence of detectable serum C1q, and low classical complement pathway activity. We identified a previously reported de novo heterozygous pathogenic splice mutation in PIK3R1 (c.1425 + 1G > A), resulting in the skipping of exon 11 of the p85α subunit of phosphatidylinositol 3-kinase and causing activated PI3Kδ syndrome type II (APDS2). This explained the phenotype, with the exception of digital vasculitis and C1q deficiency, which have never been described in association with APDS2. No mutations were identified in C1QA, B, or C, their promoter regions, or in any other complement component. Functional studies indicated normal monocytic C1q production and release, suggesting that the observed C1q deficiency was caused by peripheral consumption of C1q. Since C1q deficiency has never been associated with APDS2, we assessed C1q levels in two unrelated patients with genetically confirmed APDS2 and confirmed C1q deficiency in those two cases as well. This observation suggests C1q deficiency to be an inherent but previously unrecognized feature of APDS2. We speculate that the consumption of C1q is driven by increased apoptotic bodies derived from immune cellular senescence, combined with elevated IgM production (both inherent features of APDS2). Secondary C1q deficiency in APDS2 may further contribute to immunodeficiency and could also be associated with inflammatory immune dysregulatory phenotypes, such as the digital vasculitis observed in our case.
Collapse
Affiliation(s)
- Ying Hong
- Infection, Immunology and Inflammation Research & Teaching Department, University College London Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Sira Nanthapisal
- Infection, Immunology and Inflammation Research & Teaching Department, University College London Great Ormond Street Institute of Child Health, London, United Kingdom
- Department of Pediatrics, Thammasat University, Bangkok, Thailand
| | - Ebun Omoyinmi
- Infection, Immunology and Inflammation Research & Teaching Department, University College London Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Peter Olbrich
- Paediatric Infectious Diseases, Rheumatology and Immunology Unit, Institut of Biomedicine of Seville, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Olaf Neth
- Paediatric Infectious Diseases, Rheumatology and Immunology Unit, Institut of Biomedicine of Seville, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Carsten Speckmann
- Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
| | - Jose Manuel Lucena
- Unidad de Inmunología, Hospital Universitario Virgen del Rocio, Seville, Spain
| | - Kimberly Gilmour
- Clinical Immunology Laboratory, Great Ormond Street Hospital NHS Foundation Trust, London, United Kingdom
| | - Austen Worth
- Clinical Immunology Laboratory, Great Ormond Street Hospital NHS Foundation Trust, London, United Kingdom
| | - Nigel Klein
- Infection, Immunology and Inflammation Research & Teaching Department, University College London Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Despina Eleftheriou
- Infection, Immunology and Inflammation Research & Teaching Department, University College London Great Ormond Street Institute of Child Health, London, United Kingdom
- ARUK Centre for Adolescent Rheumatology, UCL, London, United Kingdom
| | - Paul Brogan
- Infection, Immunology and Inflammation Research & Teaching Department, University College London Great Ormond Street Institute of Child Health, London, United Kingdom
| |
Collapse
|
640
|
Neely J, Rychkov D, Paranjpe M, Waterfield M, Kim S, Sirota M. Gene Expression Meta-Analysis Reveals Concordance in Gene Activation, Pathway, and Cell-Type Enrichment in Dermatomyositis Target Tissues. ACR Open Rheumatol 2019; 1:657-666. [PMID: 31872188 PMCID: PMC6917332 DOI: 10.1002/acr2.11081] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 08/23/2019] [Indexed: 01/21/2023] Open
Abstract
Objective We conducted a comprehensive gene expression meta‐analysis in dermatomyositis (DM) muscle and skin tissues to identify shared disease‐relevant genes and pathways across tissues. Methods Six publicly available data sets from DM muscle and two from skin were identified. Meta‐analysis was performed by first processing data sets individually then cross‐study normalization and merging creating tissue‐specific gene expression matrices for subsequent analysis. Complementary single‐gene and network analyses using Significance Analysis of Microarrays (SAM) and Weighted Gene Co‐expression Network Analysis (WGCNA) were conducted to identify genes significantly associated with DM. Cell‐type enrichment was performed using xCell. Results There were 544 differentially expressed genes (FC ≥ 1.3, q < 0.05) in muscle and 300 in skin. There were 94 shared upregulated genes across tissues enriched in type I and II interferon (IFN) signaling and major histocompatibility complex (MHC) class I antigen‐processing pathways. In a network analysis, we identified eight significant gene modules in muscle and seven in skin. The most highly correlated modules were enriched in pathways consistent with the single‐gene analysis. Additional pathways uncovered by WGCNA included T‐cell activation and T‐cell receptor signaling. In the cell‐type enrichment analysis, both tissues were highly enriched in activated dendritic cells and M1 macrophages. Conclusion There is striking similarity in gene expression across DM target tissues with enrichment of type I and II IFN pathways, MHC class I antigen‐processing, T‐cell activation, and antigen‐presenting cells. These results suggest IFN‐γ may contribute to the global IFN signature in DM, and altered auto‐antigen presentation through the class I MHC pathway may be important in disease pathogenesis.
Collapse
Affiliation(s)
- Jessica Neely
- Department of Pediatrics, University of California, San Francisco
| | - Dmitry Rychkov
- Department of Pediatrics, University of California, San Francisco
| | - Manish Paranjpe
- Department of Pediatrics, University of California, San Francisco
| | | | - Susan Kim
- Department of Pediatrics, University of California, San Francisco
| | - Marina Sirota
- Department of Pediatrics, University of California, San Francisco
| |
Collapse
|
641
|
STING-Associated Vasculopathy with Onset in Infancy in Three Children with New Clinical Aspect and Unsatisfactory Therapeutic Responses to Tofacitinib. J Clin Immunol 2019; 40:114-122. [PMID: 31705453 DOI: 10.1007/s10875-019-00690-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/10/2019] [Indexed: 10/25/2022]
Abstract
PURPOSE STING-associated vasculopathy with onset in infancy (SAVI) is a new rare auto-inflammatory disease. The purpose of this study is to report new cases and summarize the manifestations and outcome of SAVI. METHODS We made a retrospective analysis of three pediatric patients diagnosed with SAVI between March 2016 and July 2018 in Beijing Children's Hospital. RESULTS Three patients comprised one boy and two girls. The median age of onset was 4 months. All patients had the same de novo heterozygous mutation (c.463G>A, p. V155M) of TMEM173. All patients presented with interstitial lung disease and one coexisted with diffuse alveolar hemorrhage. Rashes were presented in two patients. Other clinical manifestations include febrile attacks, failure to thrive, arthritis, myositis, cerebrovascular involvement, ureteral calculus, gastroesophageal reflux, and malnutrition. Ground-glass opacities were the most common features of chest computed tomography, followed with cysts and reticular opacities. Transbronchial lung biopsy was performed in one patient revealing pulmonary vasculitis. Skin biopsy was performed in one patient with changes of vasculitis. All patients were treated with corticosteroids and two patients received combined treatment of tofacitinib. The therapeutic effects of tofacitinib were limited on interstitial lung disease in both patients and were poor on rashes in one patient. One patient under the treatment of tofacitinib died. CONCLUSIONS New clinical aspect of diffuse alveolar hemorrhage is first reported to be associated with SAVI. Unsatisfactory therapeutic effects of tofacitinib are observed in this study and further evaluations are needed.
Collapse
|
642
|
Abstract
Over the past decade, preclinical and clinical research have confirmed the essential role of interferons for effective host immunological responses to malignant cells. Type I interferons (IFNα and IFNβ) directly regulate transcription of >100 downstream genes, which results in a myriad of direct (on cancer cells) and indirect (through immune effector cells and vasculature) effects on the tumour. New insights into endogenous and exogenous activation of type I interferons in the tumour and its microenvironment have given impetus to drug discovery and patient evaluation of interferon-directed strategies. When combined with prior observations or with other effective modalities for cancer treatment, modulation of the interferon system could contribute to further reductions in cancer morbidity and mortality. This Review discusses new interferon-directed therapeutic opportunities, ranging from cyclic dinucleotides to genome methylation inhibitors, angiogenesis inhibitors, chemoradiation, complexes with neoantigen-targeted monoclonal antibodies, combinations with other emerging therapeutic interventions and associations of interferon-stimulated gene expression with patient prognosis - all of which are strategies that have or will soon enter translational clinical evaluation.
Collapse
|
643
|
The triggers of the cGAS-STING pathway and the connection with inflammatory and autoimmune diseases. INFECTION GENETICS AND EVOLUTION 2019; 77:104094. [PMID: 31689545 DOI: 10.1016/j.meegid.2019.104094] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/18/2019] [Accepted: 10/23/2019] [Indexed: 12/13/2022]
Abstract
Cyclic GMP-AMP synthase (cGAS) is a cytosolic nucleic acid sensor that can bind to dsDNA. It maintains an autoinhibited state in the absence of cytosolic dsDNA, while when activated, it in turn activates its adaptor protein STING, ultimately triggering a cascade that produces inflammatory cytokines and type I interferons (IFNs). With further research, additional types of nucleic acids have been found to be activators of the cGAS-STING pathway. The cGAS-STING pathway can provide protection or resistance against infections; however, improper or overactivation might cause severe inflammatory pathologies, including autoimmunity. This article systematically reviews the latest research progress on the axis, including categorical pathway triggers, the connection with autoimmune disease and drug therapy progress.
Collapse
|
644
|
Figueras-Nart I, Mascaró JM, Solanich X, Hernández-Rodríguez J. Dermatologic and Dermatopathologic Features of Monogenic Autoinflammatory Diseases. Front Immunol 2019; 10:2448. [PMID: 31736939 PMCID: PMC6828938 DOI: 10.3389/fimmu.2019.02448] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 10/01/2019] [Indexed: 12/12/2022] Open
Abstract
Autoinflammatory diseases include disorders with a monogenic cause and also complex conditions associated to polygenic or multifactorial factors. An increased number of both monogenic and polygenic autoinflammatory conditions have been identified during the last years. Although skin manifestations are often predominant in monogenic autoinflammatory diseases, clinical and histopathological information regarding their dermatological involvement is still scarce. Monogenic autoinflammatory diseases with cutaneous expression can be classified based on the predominant lesion: (1) maculopapular rashes or inflammatory plaques; (2) urticarial rashes; (3) pustular, pyogenic or neutrophilic dermatosis-like rashes; (4) panniculitis or subcutaneous nodules; (5) vasculitis or vasculopathy; (6) hyperkeratotic lesions; (7) hyperpigmented lesions; (8) bullous lesions; and (9) aphthous lesions. By using this classification, this review intends to provide clinical and histopathological knowledge about cutaneous involvement in monogenic autoinflammatory diseases.
Collapse
Affiliation(s)
- Ignasi Figueras-Nart
- Department of Dermatology, Bellvitge Hospital, University of Barcelona, Barcelona, Spain
| | - José M Mascaró
- Department of Dermatology, Hospital Clinic, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Xavier Solanich
- Department of Internal Medicine, Bellvitge Hospital, University of Barcelona, Barcelona, Spain
| | - José Hernández-Rodríguez
- Clinical Unit of Autoinflammatory Diseases and Vasculitis Research Unit, Department of Autoimmune Diseases, Hospital Clinic, IDIBAPS, University of Barcelona, Barcelona, Spain
| |
Collapse
|
645
|
Mammen AL, Allenbach Y, Stenzel W, Benveniste O. 239th ENMC International Workshop: Classification of dermatomyositis, Amsterdam, the Netherlands, 14-16 December 2018. Neuromuscul Disord 2019; 30:70-92. [PMID: 31791867 DOI: 10.1016/j.nmd.2019.10.005] [Citation(s) in RCA: 176] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 10/21/2019] [Indexed: 12/29/2022]
Affiliation(s)
- Andrew L Mammen
- Muscle Disease Unit, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, 50 South Drive, Building 50, Room 1146, MD 20892, United States.
| | - Yves Allenbach
- Department of Internal Medicine and Clinical Immunology, Pitié Salpetrière Hospital, AP-HP Sorbonne University, Paris, France
| | - Werner Stenzel
- Department of Neuropathology, Charité-Universitatsmedizin, Berlin, Germany
| | - Olivier Benveniste
- Department of Internal Medicine and Clinical Immunology, Pitié Salpetrière Hospital, AP-HP Sorbonne University, Paris, France
| | | |
Collapse
|
646
|
Van Horebeek L, Dubois B, Goris A. Somatic Variants: New Kids on the Block in Human Immunogenetics. Trends Genet 2019; 35:935-947. [PMID: 31668909 DOI: 10.1016/j.tig.2019.09.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/23/2019] [Accepted: 09/27/2019] [Indexed: 01/21/2023]
Abstract
Somatic variants are not inherited but acquired during an individual's lifetime, and individuals are increasingly considered as complex mosaics of genetically distinct cells. Whereas this concept is long-recognized in cancer, this review focuses on the growing role of somatic variants in immune cells in nonmalignant immune-related disorders, such as primary immunodeficiency and autoimmune diseases. Older case reports described somatic variants early in development, leading to large numbers of affected cells and severe phenotypes. Thanks to technological evolution, it is now feasible to detect somatic variants occurring later in life and affecting fewer cells. Hence, only recently is the scale at which somatic variants contribute to monogenic diseases being uncovered and is their contribution to complex diseases being explored systematically.
Collapse
Affiliation(s)
- L Van Horebeek
- KU Leuven Department of Neurosciences, Laboratory for Neuroimmunology, 3000 Leuven, Belgium; Leuven Brain Institute, 3000 Leuven, Belgium
| | - B Dubois
- KU Leuven Department of Neurosciences, Laboratory for Neuroimmunology, 3000 Leuven, Belgium; Leuven Brain Institute, 3000 Leuven, Belgium; University Hospitals Leuven, Department of Neurology, 3000 Leuven, Belgium
| | - A Goris
- KU Leuven Department of Neurosciences, Laboratory for Neuroimmunology, 3000 Leuven, Belgium; Leuven Brain Institute, 3000 Leuven, Belgium.
| |
Collapse
|
647
|
Lemos H, Mohamed E, Huang L, Chandler PR, Ou R, Pacholczyk R, Mellor AL. Stimulator of interferon genes agonists attenuate type I diabetes progression in NOD mice. Immunology 2019; 158:353-361. [PMID: 31557322 DOI: 10.1111/imm.13122] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/09/2019] [Accepted: 09/16/2019] [Indexed: 12/27/2022] Open
Abstract
Reagents that activate the signaling adaptor stimulator of interferon genes (STING) suppress experimentally induced autoimmunity in murine models of multiple sclerosis and arthritis. In this study, we evaluated STING agonists as potential reagents to inhibit spontaneous autoimmune type I diabetes (T1D) onset in non-obese diabetic (NOD) female mice. Treatments with DNA nanoparticles (DNPs), which activate STING when cargo DNA is sensed, delayed T1D onset and reduced T1D incidence when administered before T1D onset. DNP treatment elevated indoleamine 2,3 dioxygenase (IDO) activity, which regulates T-cell immunity, in spleen, pancreatic lymph nodes and pancreas of NOD mice. Therapeutic responses to DNPs were partially reversed by inhibiting IDO and DNP treatment synergized with insulin therapy to further delay T1D onset and reduce T1D incidence. Treating pre-diabetic NOD mice with cyclic guanyl-adenyl dinucleotide (cGAMP) to activate STING directly delayed T1D onset and stimulated interferon-αβ (IFN-αβ), while treatment with cyclic diguanyl nucleotide (cdiGMP) did not delay T1D onset or induce IFN-αβ in NOD mice. DNA sequence analyses revealed that NOD mice possess a STING polymorphism that may explain differential responses to cGAMP and cdiGMP. In summary, STING agonists attenuate T1D progression and DNPs enhance therapeutic responses to insulin therapy.
Collapse
Affiliation(s)
- Henrique Lemos
- Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle-upon-Tyne, UK
| | - Eslam Mohamed
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Lei Huang
- Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle-upon-Tyne, UK
| | - Phillip R Chandler
- Biochemistry and Molecular Biology, Georgia Cancer Center, Augusta University, Augusta, GA, USA
| | - Rong Ou
- Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle-upon-Tyne, UK
| | - Rafal Pacholczyk
- Biochemistry and Molecular Biology, Georgia Cancer Center, Augusta University, Augusta, GA, USA
| | - Andrew L Mellor
- Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle-upon-Tyne, UK
| |
Collapse
|
648
|
Su T, Zhang Y, Valerie K, Wang XY, Lin S, Zhu G. STING activation in cancer immunotherapy. Theranostics 2019; 9:7759-7771. [PMID: 31695799 PMCID: PMC6831454 DOI: 10.7150/thno.37574] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 09/02/2019] [Indexed: 12/19/2022] Open
Abstract
Cancer immunotherapy modulates and leverages the host immune system to treat cancer. The past decade has witnessed historical advancement of cancer immunotherapy. A myriad of approaches have been explored to elicit or augment anticancer innate immunity and/or adaptive immunity. Recently, activation of stimulator of interferon (IFN) genes (STING), an intracellular receptor residing in the endoplasmic reticulum, has shown great potential to enhance antitumor immunity through the induction of a variety of pro-inflammatory cytokines and chemokines, including type I IFNs. A number of natural and synthetic STING agonists have been discovered or developed, and tested in preclinical models and in the clinic for the immunotherapy of diseases such as cancer and infectious diseases. Cyclic dinucleotides (CDNs), such as cyclic dimeric guanosine monophosphate (c-di-GMP), cyclic dimeric adenosine monophosphate (c-di-AMP), and cyclic GMP-AMP (cGAMP), are a class of STING agonists that can elicit immune responses. However, natural CDNs are hydrophilic small molecules with negative charges and are susceptible to enzymatic degradation, leading to low bioavailability in target tissues yet unwanted toxicities and narrow therapeutic windows. Drug delivery systems, coupled with nucleic acid chemistry, have been exploited to address these challenges. Here, we will discuss the underlying immunological mechanisms and approaches to STING activation, with a focus on the delivery of STING agonists, for cancer immunotherapy.
Collapse
Affiliation(s)
- Ting Su
- Department of Rehabilitation Medicine, Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Pharmaceutics and Center for Pharmaceutical Engineering and Sciences, School of Pharmacy, Richmond, VA, 23298, USA
| | - Yu Zhang
- Department of Rehabilitation Medicine, Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Pharmaceutics and Center for Pharmaceutical Engineering and Sciences, School of Pharmacy, Richmond, VA, 23298, USA
| | - Kristoffer Valerie
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23298, USA
- Department of Radiation Oncology, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Xiang-Yang Wang
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23298, USA
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, 23298, USA
- Institute of Molecular Medicine, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Shuibin Lin
- Department of Rehabilitation Medicine, Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Guizhi Zhu
- Department of Pharmaceutics and Center for Pharmaceutical Engineering and Sciences, School of Pharmacy, Richmond, VA, 23298, USA
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23298, USA
- Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA, 23219, USA
| |
Collapse
|
649
|
Balci S, Ekinci RMK, de Jesus AA, Goldbach-Mansky R, Yilmaz M. Baricitinib experience on STING-associated vasculopathy with onset in infancy: A representative case from Turkey. Clin Immunol 2019; 212:108273. [PMID: 31626957 DOI: 10.1016/j.clim.2019.108273] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 10/10/2019] [Accepted: 10/10/2019] [Indexed: 11/26/2022]
Abstract
Stimulator of interferon genes associated vasculopathy with onset in infancy (SAVI), caused by heterozygote gain-of-function mutations in TMEM173, is characterized by fever attacks with ulcerating cutaneous manifestations on cold-sensitive areas and interstitial lung disease. A six-month-old boy was admitted to our hospital with fever, cough, and rash on the external surface of both upper and lower extremities. Respiratory symptoms consistent with ILD developed and skin lesions evolved to eschar formation particularly on acral regions. Ultimately, diagnosis of SAVI was confirmed at the age of 10 months due to the high level of interferon-score and a heterozygous N154S mutation in TMEM173. Since systemic corticosteroid and ruxolitinib were not effective, baricitinib was initiated at the age of 15 months, resulting in alleviation of fever attacks, cutaneous manifestations and respiratory symptoms within 2 months. In conclusion, we reported an infant diagnosed with SAVI at the age of 10 months and treated with baricitinib.
Collapse
Affiliation(s)
- Sibel Balci
- Department of Pediatric Rheumatology, Cukurova University Faculty of Medicine, Adana, Turkey.
| | | | - Adriana Almeida de Jesus
- Translational Autoinflammatory Disease Studies (TADS), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD, USA.
| | - Raphaela Goldbach-Mansky
- Translational Autoinflammatory Disease Studies (TADS), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD, USA.
| | - Mustafa Yilmaz
- Department of Pediatric Rheumatology, Cukurova University Faculty of Medicine, Adana, Turkey; Department of Pediatric Allergy and Immunology, Cukurova University Faculty of Medicine, Adana, Turkey
| |
Collapse
|
650
|
Xu J, Zhang L, Xu Y, Zhang H, Gao J, Wang Q, Tian Z, Xuan L, Chen H, Wang Y. PP2A Facilitates Porcine Reproductive and Respiratory Syndrome Virus Replication by Deactivating irf3 and Limiting Type I Interferon Production. Viruses 2019; 11:v11100948. [PMID: 31618847 PMCID: PMC6832233 DOI: 10.3390/v11100948] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/11/2019] [Accepted: 10/11/2019] [Indexed: 12/12/2022] Open
Abstract
Protein phosphatase 2A (PP2A), a major serine/threonine phosphatase in mammalian cells, is known to regulate the kinase-driven intracellular signaling pathways. Emerging evidences have shown that the PP2A phosphatase functions as a bona-fide therapeutic target for anticancer therapy, but it is unclear whether PP2A affects a porcine reproductive and respiratory syndrome virus infection. In the present study, we demonstrated for the first time that inhibition of PP2A activity by either inhibitor or small interfering RNA duplexes in target cells significantly reduced their susceptibility to porcine reproductive and respiratory syndrome virus (PRRSV) infection. Further analysis revealed that inhibition of PP2A function resulted in augmented production of type I interferon (IFN). The mechanism is that inhibition of PP2A activity enhances the levels of phosphorylated interferon regulatory factor 3, which activates the transcription of IFN-stimulated genes. Moreover, inhibition of PP2A activity mainly blocked PRRSV replication in the early stage of viral life cycle, after virus entry but before virus release. Using type I IFN receptor 2 specific siRNA in combination with PP2A inhibitor, we confirmed that the effect of PP2A on viral replication within target cells was an interferon-dependent manner. Taken together, these findings demonstrate that PP2A serves as a negative regulator of host cells antiviral responses and provides a novel therapeutic target for virus infection.
Collapse
Affiliation(s)
- Jiayu Xu
- State Key Laboratory of Veterinary Biotechnology, Heilongjiang Provincial Key Laboratory of Laboratory Animal and Comparative Medicine, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Lu Zhang
- State Key Laboratory of Veterinary Biotechnology, Heilongjiang Provincial Key Laboratory of Laboratory Animal and Comparative Medicine, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Yunfei Xu
- State Key Laboratory of Veterinary Biotechnology, Heilongjiang Provincial Key Laboratory of Laboratory Animal and Comparative Medicine, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - He Zhang
- State Key Laboratory of Veterinary Biotechnology, Heilongjiang Provincial Key Laboratory of Laboratory Animal and Comparative Medicine, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Junxin Gao
- State Key Laboratory of Veterinary Biotechnology, Heilongjiang Provincial Key Laboratory of Laboratory Animal and Comparative Medicine, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Qian Wang
- State Key Laboratory of Veterinary Biotechnology, Heilongjiang Provincial Key Laboratory of Laboratory Animal and Comparative Medicine, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Zhijun Tian
- State Key Laboratory of Veterinary Biotechnology, Heilongjiang Provincial Key Laboratory of Laboratory Animal and Comparative Medicine, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China
| | - Lv Xuan
- Department of public health policy, University of California, Irvine, CA 92697, USA
| | - Hongyan Chen
- State Key Laboratory of Veterinary Biotechnology, Heilongjiang Provincial Key Laboratory of Laboratory Animal and Comparative Medicine, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
| | - Yue Wang
- State Key Laboratory of Veterinary Biotechnology, Heilongjiang Provincial Key Laboratory of Laboratory Animal and Comparative Medicine, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
| |
Collapse
|