Inviting an author to review:
Find an author and click ‘Invite to review selected article’ near their name.
Search for authorsSearch for similar articles
3
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Role of CD14+ monocyte-derived oxidised mitochondrial DNA in the inflammatory interferon type 1 signature in juvenile dermatomyositis

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Objectives

          To define the host mechanisms contributing to the pathological interferon (IFN) type 1 signature in Juvenile dermatomyositis (JDM).

          Methods

          RNA-sequencing was performed on CD4 +, CD8 +, CD14 + and CD19 + cells sorted from pretreatment and on-treatment JDM (pretreatment n=10, on-treatment n=11) and age/sex-matched child healthy-control (CHC n=4) peripheral blood mononuclear cell (PBMC). Mitochondrial morphology and superoxide were assessed by fluorescence microscopy, cellular metabolism by 13C glucose uptake assays, and oxidised mitochondrial DNA (oxmtDNA) content by dot-blot. Healthy-control PBMC and JDM pretreatment PBMC were cultured with IFN-α, oxmtDNA, cGAS-inhibitor, TLR-9 antagonist and/or n-acetyl cysteine (NAC). IFN-stimulated gene (ISGs) expression was measured by qPCR. Total numbers of patient and controls for functional experiments, JDM n=82, total CHC n=35.

          Results

          Dysregulated mitochondrial-associated gene expression correlated with increased ISG expression in JDM CD14+ monocytes. Altered mitochondrial-associated gene expression was paralleled by altered mitochondrial biology, including ‘megamitochondria’, cellular metabolism and a decrease in gene expression of superoxide dismutase ( SOD)1. This was associated with enhanced production of oxidised mitochondrial (oxmt)DNA. OxmtDNA induced ISG expression in healthy PBMC, which was blocked by targeting oxidative stress and intracellular nucleic acid sensing pathways. Complementary experiments showed that, under in vitro experimental conditions, targeting these pathways via the antioxidant drug NAC, TLR9 antagonist and to a lesser extent cGAS-inhibitor, suppressed ISG expression in pretreatment JDM PBMC.

          Conclusions

          These results describe a novel pathway where altered mitochondrial biology in JDM CD14+ monocytes lead to oxmtDNA production and stimulates ISG expression. Targeting this pathway has therapeutical potential in JDM and other IFN type 1-driven autoimmune diseases.

          Related collections

          Most cited references71

          • Record: found
          • Abstract: found
          • Article: not found

          Mitochondrial reactive oxygen species (ROS) and ROS-induced ROS release.

          Byproducts of normal mitochondrial metabolism and homeostasis include the buildup of potentially damaging levels of reactive oxygen species (ROS), Ca(2+), etc., which must be normalized. Evidence suggests that brief mitochondrial permeability transition pore (mPTP) openings play an important physiological role maintaining healthy mitochondria homeostasis. Adaptive and maladaptive responses to redox stress may involve mitochondrial channels such as mPTP and inner membrane anion channel (IMAC). Their activation causes intra- and intermitochondrial redox-environment changes leading to ROS release. This regenerative cycle of mitochondrial ROS formation and release was named ROS-induced ROS release (RIRR). Brief, reversible mPTP opening-associated ROS release apparently constitutes an adaptive housekeeping function by the timely release from mitochondria of accumulated potentially toxic levels of ROS (and Ca(2+)). At higher ROS levels, longer mPTP openings may release a ROS burst leading to destruction of mitochondria, and if propagated from mitochondrion to mitochondrion, of the cell itself. The destructive function of RIRR may serve a physiological role by removal of unwanted cells or damaged mitochondria, or cause the pathological elimination of vital and essential mitochondria and cells. The adaptive release of sufficient ROS into the vicinity of mitochondria may also activate local pools of redox-sensitive enzymes involved in protective signaling pathways that limit ischemic damage to mitochondria and cells in that area. Maladaptive mPTP- or IMAC-related RIRR may also be playing a role in aging. Because the mechanism of mitochondrial RIRR highlights the central role of mitochondria-formed ROS, we discuss all of the known ROS-producing sites (shown in vitro) and their relevance to the mitochondrial ROS production in vivo. Copyright © 2014 the American Physiological Society.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Regulation and function of the cGAS-STING pathway of cytosolic DNA sensing.

            The recognition of microbial nucleic acids is a major mechanism by which the immune system detects pathogens. Cyclic GMP-AMP (cGAMP) synthase (cGAS) is a cytosolic DNA sensor that activates innate immune responses through production of the second messenger cGAMP, which activates the adaptor STING. The cGAS-STING pathway not only mediates protective immune defense against infection by a large variety of DNA-containing pathogens but also detects tumor-derived DNA and generates intrinsic antitumor immunity. However, aberrant activation of the cGAS pathway by self DNA can also lead to autoimmune and inflammatory disease. Thus, the cGAS pathway must be properly regulated. Here we review the recent advances in understanding of the cGAS-STING pathway, focusing on the regulatory mechanisms and roles of this pathway in heath and disease.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Mitochondrial DNA Stress Primes the Antiviral Innate Immune Response

              Mitochondrial DNA (mtDNA) is normally present at thousands of copies per cell and is packaged into several hundred higher-order structures termed nucleoids 1 . The abundant mtDNA-binding protein, transcription factor A mitochondrial (TFAM), regulates nucleoid architecture, abundance, and segregation 2 . Complete mtDNA depletion profoundly impairs oxidative phosphorylation (OXPHOS), triggering calcium-dependent stress signaling and adaptive metabolic responses 3 . However, the cellular responses to mtDNA instability, a physiologically relevant stress observed in many human diseases and aging, remain ill-defined 4 . Here we show that moderate mtDNA stress elicited by TFAM deficiency engages cytosolic antiviral signaling to enhance the expression of a subset of interferon-stimulated genes (ISG). Mechanistically, we have found that aberrant mtDNA packaging promotes escape of mtDNA into the cytosol, where it engages the DNA sensor cGAS and promotes STING-IRF3-dependent signaling to elevate ISG expression, potentiate type I interferon responses, and confer broad viral resistance. Furthermore, we demonstrate that herpesviruses induce mtDNA stress, which potentiates antiviral signaling and type I interferon responses during infection. Our results further demonstrate that mitochondria are central participants in innate immunity, identify mtDNA stress as a cell-intrinsic trigger of antiviral signaling, and suggest that cellular monitoring of mtDNA homeostasis cooperates with canonical virus sensing mechanisms to fully license antiviral innate immunity.
                Bookmark

                Author and article information

                Journal
                Ann Rheum Dis
                Ann Rheum Dis
                annrheumdis
                ard
                Annals of the Rheumatic Diseases
                BMJ Publishing Group (BMA House, Tavistock Square, London, WC1H 9JR )
                0003-4967
                1468-2060
                May 2023
                23 December 2022
                : 82
                : 5
                : 658-669
                Affiliations
                [1 ] departmentInfection, Immunity and Inflammation Research and Teaching Department , UCL Great Ormond Street Institute of Child Health , London, UK
                [2 ] departmentCentre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH , UCL , London, UK
                [3 ] departmentNIHR Biomedical Research Centre , Great Ormond Street Hospital , London, UK
                [4 ] departmentDevelopmental Biology and Cancer Research & Teaching Department , UCL Great Ormond Street Institute of Child Health , London, UK
                [5 ] departmentCentre for Rheumatology Research, Division of Medicine , University College London , London, UK
                [6 ] departmentExperimental and Personalised Medicine, Genetics and Genomic Medicine , UCL Great Ormond Street Institute of Child Health , London, UK
                [7 ] departmentGenetics and Genomic Medicine Research & Teaching Department , UCL Great Ormond Street Institute of Child Health , London, UK
                [8 ] departmentRheumatology , Great Ormond Street Hospital NHS Trust , London, UK
                Author notes
                [Correspondence to ] Dr Meredyth G Ll Wilkinson, Infection, Immunity and Inflammation Research and Teaching Department, UCL, London, WC1N1EH, UK; meredyth.wilkinson.14@ 123456ucl.ac.uk ; Dr Elizabeth C Rosser, Department of Rheumatology, University College London, London, UK; e.rosser@ 123456ucl.ac.uk

                SE, CTD, ECR and LRW are joint senior authors.

                Author information
                http://orcid.org/0000-0002-7972-8066
                http://orcid.org/0000-0002-1431-7047
                http://orcid.org/0000-0002-1712-9957
                http://orcid.org/0000-0002-8742-8311
                http://orcid.org/0000-0003-0177-7623
                http://orcid.org/0000-0002-3929-948X
                http://orcid.org/0000-0001-8434-4465
                http://orcid.org/0000-0003-0583-5237
                http://orcid.org/0000-0002-1237-0557
                http://orcid.org/0000-0002-5819-4210
                http://orcid.org/0000-0003-0892-9204
                http://orcid.org/0000-0002-7044-5801
                http://orcid.org/0000-0003-4800-4695
                http://orcid.org/0000-0002-7495-1429
                Article
                ard-2022-223469
                10.1136/ard-2022-223469
                10176342
                36564154
                343fb67b-5c18-48b1-8730-fb1e24e5bab5
                © Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY. Published by BMJ.

                This is an open access article distributed in accordance with the Creative Commons Attribution 4.0 Unported (CC BY 4.0) license, which permits others to copy, redistribute, remix, transform and build upon this work for any purpose, provided the original work is properly cited, a link to the licence is given, and indication of whether changes were made. See: https://creativecommons.org/licenses/by/4.0/.

                History
                : 11 October 2022
                : 01 December 2022
                Funding
                Funded by: FundRef http://dx.doi.org/10.13039/100001927, Advanced Medical Research Foundation;
                Award ID: MRF- 057-0001-RG-ROSS-C0797
                Funded by: FundRef http://dx.doi.org/10.13039/501100012041, Versus Arthritis;
                Award ID: 14518
                Award ID: 20164
                Award ID: 21552
                Award ID: 21593
                Award ID: 22936
                Funded by: FundRef http://dx.doi.org/10.13039/501100000317, Action Medical Research;
                Award ID: SP4252
                Funded by: FundRef http://dx.doi.org/10.13039/100018208, Cure JM Foundation;
                Award ID: GOSH042019
                Funded by: FundRef http://dx.doi.org/10.13039/100016580, Kennedy Trust for Rheumatology Research;
                Award ID: KENN 21 22 09
                Funded by: NIHR Biomedical Research Centre, Great Ormond Street Hospital;
                Award ID: 18DS03
                Award ID: 18IR33
                Funded by: FundRef http://dx.doi.org/10.13039/501100000265, Medical Research Council;
                Award ID: MR/N003322/1
                Funded by: FundRef http://dx.doi.org/10.13039/501100000852, Henry Smith Charity;
                Funded by: FundRef http://dx.doi.org/10.13039/100010269, Wellcome Trust;
                Award ID: 085860
                Funded by: Myositis UK;
                Funded by: FundRef http://dx.doi.org/10.13039/501100001279, Great Ormond Street Hospital Charity;
                Award ID: V1268
                Categories
                Myositis
                1506
                2311
                2496
                Custom metadata
                unlocked

                Immunology
                dermatomyositis,autoimmune diseases,inflammation
                Immunology
                dermatomyositis, autoimmune diseases, inflammation

                Comments

                Comment on this article