1
|
Goldbach-Mansky R, Alehashemi S, de Jesus AA. Emerging concepts and treatments in autoinflammatory interferonopathies and monogenic systemic lupus erythematosus. Nat Rev Rheumatol 2025; 21:22-45. [PMID: 39623155 DOI: 10.1038/s41584-024-01184-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2024] [Indexed: 12/22/2024]
Abstract
Over the past two decades, the number of genetically defined autoinflammatory interferonopathies has steadily increased. Aicardi-Goutières syndrome and proteasome-associated autoinflammatory syndromes (PRAAS, also known as CANDLE) are caused by genetic defects that impair homeostatic intracellular nucleic acid and protein processing respectively. Research into these genetic defects revealed intracellular sensors that activate type I interferon production. In SAVI and COPA syndrome, genetic defects that cause chronic activation of the dinucleotide sensor stimulator of interferon genes (STING) share features of lung inflammation and fibrosis; and selected mutations that amplify interferon-α/β receptor signalling cause central nervous system manifestations resembling Aicardi-Goutières syndrome. Research into the monogenic causes of childhood-onset systemic lupus erythematosus (SLE) demonstrates the pathogenic role of autoantibodies to particle-bound extracellular nucleic acids that distinguishes monogenic SLE from the autoinflammatory interferonopathies. This Review introduces a classification for autoinflammatory interferonopathies and discusses the divergent and shared pathomechanisms of interferon production and signalling in these diseases. Early success with drugs that block type I interferon signalling, new insights into the roles of cytoplasmic DNA or RNA sensors, pathways in type I interferon production and organ-specific pathology of the autoinflammatory interferonopathies and monogenic SLE, reveal novel drug targets that could personalize treatment approaches.
Collapse
Affiliation(s)
- Raphaela Goldbach-Mansky
- Translational Autoinflammatory Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Sara Alehashemi
- Translational Autoinflammatory Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Adriana A de Jesus
- Translational Autoinflammatory Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
2
|
Liu Z, Fu Q, Shao Y, Duan X. The role of mitochondrial DNA copy number in autoimmune disease: a bidirectional two sample mendelian randomization study. Front Immunol 2024; 15:1409969. [PMID: 39464879 PMCID: PMC11502960 DOI: 10.3389/fimmu.2024.1409969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 09/23/2024] [Indexed: 10/29/2024] Open
Abstract
Background Mitochondrial DNA (mtDNA) plays an important role in autoimmune diseases (AD), yet the relationship between mitochondria and autoimmune disease is controversial. This study employed bidirectional Mendelian randomization (MR) to explore the causal relationship between mtDNA copy number and 13 ADs (including ankylosing spondylitis [AS], Crohn's disease [CD], juvenile rheumatoid arthritis [JRA], polymyalgia rheumatica [PMR], psoriasis [PSO], rheumatoid arthritis [RA], Sjogren's syndrome [SS], systemic lupus erythematosus [SLE], thyrotoxicosis, type 1 diabetes mellitus [T1DM], ulcerative colitis [UC], and vitiligo). Methods A two-sample MR analysis was performed to assess the causal relationship between mtDNA copy number and AD. Genome-wide association study (GWAS) for mtDNA copy number were obtained from the UK Biobank (UKBB), while those associated with AD were sourced from the FinnGen Biobank. Inverse variance weighting (IVW) was the primary analysis method, complemented by three sensitivity analyses (MR-Egger, weighted median, weighted mode) to validate the results. Results IVW MR analysis identified significant associations between mtDNA copy number and CD (OR=2.51, 95% CI 1.56-4.22, P<0.001), JRA (OR=1.87, 95% CI 1.17-7.65, P=0.022), RA (OR=1.71, 95%CI 1.18-2.47, P=0.004), thyrotoxicosis (OR=0.51, 95% CI0.27-0.96, P=0.038), and T1DM (OR=0.51, 95% CI 0.27-0.96, P=0.038). Sensitivity analyses indicated no horizontal pleiotropy. Conclusions Our study revealed a potential causal relationship between mtDNA copy number and ADs, indicating that these markers may be relevant in exploring new therapeutic approaches.
Collapse
Affiliation(s)
- Zhekang Liu
- Rheumatology and Immunology Department, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Qingan Fu
- Cardiovascular Medicine Department, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yijia Shao
- Rheumatology and Immunology Department, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xinwang Duan
- Rheumatology and Immunology Department, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
3
|
Michailidou D, Giaglis S, Dale GL. The platelet-mitochondria nexus in autoimmune and musculoskeletal diseases. Clin Immunol 2024; 267:110350. [PMID: 39218194 DOI: 10.1016/j.clim.2024.110350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 08/15/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
Platelets are crucial for thrombosis and hemostasis. Importantly, they contain mitochondria that are responsible for energy generation and therefore vital for platelet survival and activation. Activated platelets can release mitochondria that may be free or encapsulated in platelet extracellular vesicles (EVs). Extruded mitochondria are a well-known source of mitochondrial DNA, and mitochondrial antigens that can be targeted by autoantibodies forming immune complexes (IC). Interaction of IC with the platelet cell surface FcγRIIA receptor results in platelet activation and release of platelet granule components. In this review, we summarize how platelets and mitochondria may contribute to the pathogenesis of different autoimmune and musculoskeletal diseases. Targeting key drivers of mitochondrial extrusion may ultimately lead to urgently needed targeted pharmacological interventions for treating inflammation and thrombotic diathesis, and halting organ damage in some of these rheumatological conditions.
Collapse
Affiliation(s)
- Despina Michailidou
- Division of Rheumatology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Division of Rheumatology, Oklahoma City VA Health Care System, Oklahoma City, OK, USA.
| | - Stavros Giaglis
- Laboratory for Experimental Rheumatology, Department of Biomedicine, University Hospital Basel, Basel, Switzerland; Department of Rheumatology, University Hospital Basel, Basel, Switzerland
| | - George L Dale
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
4
|
Poznyak AV, Orekhov NA, Churov AV, Starodubtseva IA, Beloyartsev DF, Kovyanova TI, Sukhorukov VN, Orekhov AN. Mitochondrial Dysfunction in Systemic Lupus Erythematosus: Insights and Therapeutic Potential. Diseases 2024; 12:226. [PMID: 39329895 PMCID: PMC11430897 DOI: 10.3390/diseases12090226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/03/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disorder characterized by the presence of various serum autoantibodies and multi-system effects, predominantly affecting young female patients. The pathogenesis of SLE involves a combination of genetic factors, environmental triggers, and pathogen invasions that disrupt immune cell activation, leading to the release of autoantibodies and chronic inflammation. Mitochondria, as the primary cellular powerhouses, play a crucial role in SLE development through their control of energy generation, reactive oxygen species (ROS) production, and cellular apoptotic pathways. Dysregulation of mitochondrial structure and function can contribute to the immune dysregulation, oxidative stress, and inflammation seen in SLE. Recent research has highlighted the impact of mitochondrial dysfunction on various immune cells involved in SLE pathogenesis, such as T-lymphocytes, B-lymphocytes, neutrophils, and plasmacytoid dendritic cells. Mitochondrial dysfunction in these immune cells leads to increased ROS production, disrupted mitophagy, and alterations in energy metabolism, contributing to immune dysregulation and inflammation. Moreover, genetic variations in mitochondrial DNA (mtDNA) and abnormalities in mitochondrial dynamics have been linked to the pathogenesis of SLE, exacerbating oxidative stress and immune abnormalities. Targeting mitochondrial function has emerged as a promising therapeutic approach for SLE. Drugs such as sirolimus, N-acetylcysteine, coenzyme Q10, and metformin have shown potential in restoring mitochondrial homeostasis, reducing oxidative stress, and modulating immune responses in SLE. These agents have demonstrated efficacy in preclinical models and clinical studies by improving disease activity, reducing autoantibody titers, and ameliorating organ damage in SLE patients. In conclusion, this review underscores the critical role of mitochondria in the pathogenesis of SLE and the potential of targeting mitochondrial dysfunction as a novel therapeutic strategy for improving outcomes in SLE patients. Further investigation into the mechanisms underlying mitochondrial involvement in SLE and the development of targeted mitochondrial therapies hold promise for advancing SLE treatment and enhancing patient care.
Collapse
Affiliation(s)
- Anastasia V Poznyak
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, 121609 Moscow, Russia
| | - Nikolay A Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia
| | - Alexey V Churov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia
- Russian Gerontology Clinical Research Centre, Institute on Aging Research, Pirogov Russian National Research Medical University, Russian Federation, 16 1st Leonova Street, 129226 Moscow, Russia
| | - Irina A Starodubtseva
- Department of Polyclinic Therapy, N.N. Burdenko Voronezh State Medical University, 10 Studencheskaya Street, 394036 Voronezh, Russia
| | - Dmitry F Beloyartsev
- Vascular Surgery Department, A.V. Vishnevsky National Medical Research Center of Surgery, 27 Bolshaya Serpukhovskaya Street, 117997 Moscow, Russia
| | - Tatiana I Kovyanova
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, 121609 Moscow, Russia
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia
| | - Vasily N Sukhorukov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia
| | - Alexander N Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia
| |
Collapse
|
5
|
Gürcan D, Baysoy E, Kaleli-Can G. Anti-IgG Doped Melanin Nanoparticles Functionalized Quartz Tuning Fork Immunosensors for Immunoglobulin G Detection: In Vitro and In Silico Study. SENSORS (BASEL, SWITZERLAND) 2024; 24:4319. [PMID: 39001098 PMCID: PMC11243786 DOI: 10.3390/s24134319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/29/2024] [Accepted: 06/30/2024] [Indexed: 07/16/2024]
Abstract
The quartz tuning fork (QTF) is a promising instrument for biosensor applications due to its advanced properties such as high sensitivity to physical quantities, cost-effectiveness, frequency stability, and high-quality factor. Nevertheless, the fork's small size and difficulty in modifying the prongs' surfaces limit its wide use in experimental research. Our study presents the development of a QTF immunosensor composed of three active layers: biocompatible natural melanin nanoparticles (MNPs), glutaraldehyde (GLU), and anti-IgG layers, for the detection of immunoglobulin G (IgG). Frequency shifts of QTFs after MNP functionalization, GLU activation, and anti-IgG immobilization were measured with an Asensis QTF F-master device. Using QTF immunosensors that had been modified under optimum conditions, the performance of QTF immunosensors for IgG detection was evaluated. Accordingly, a finite element method (FEM)-based model was produced using the COMSOL Multiphysics software program (COMSOL License No. 2102058) to simulate the effect of deposited layers on the QTF resonance frequency. The experimental results, which demonstrated shifts in frequency with each layer during QTF surface functionalization, corroborated the simulation model predictions. A modelling error of 0.05% was observed for the MNP-functionalized QTF biosensor compared to experimental findings. This study validated a simulation model that demonstrates the advantages of a simulation-based approach to optimize QTF biosensors, thereby reducing the need for extensive laboratory work.
Collapse
Affiliation(s)
- Dilhan Gürcan
- Department of Biomedical Engineering, İzmir Democracy University, İzmir 35140, Türkiye
| | - Engin Baysoy
- Department of Biomedical Engineering, Bahçeşehir University, İstanbul 34353, Türkiye
| | - Gizem Kaleli-Can
- Department of Biomedical Engineering, İzmir Democracy University, İzmir 35140, Türkiye
| |
Collapse
|
6
|
Li L, Liu F, Feng C, Chen Z, Zhang N, Mao J. Role of mitochondrial dysfunction in kidney disease: Insights from the cGAS-STING signaling pathway. Chin Med J (Engl) 2024; 137:1044-1053. [PMID: 38445370 PMCID: PMC11062705 DOI: 10.1097/cm9.0000000000003022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Indexed: 03/07/2024] Open
Abstract
ABSTRACT Over the past decade, mitochondrial dysfunction has been investigated as a key contributor to acute and chronic kidney disease. However, the precise molecular mechanisms linking mitochondrial damage to kidney disease remain elusive. The recent insights into the cyclic guanosine monophosphate-adenosine monophosphate (GMP-AMP) synthetase (cGAS)-stimulator of interferon gene (STING) signaling pathway have revealed its involvement in many renal diseases. One of these findings is that mitochondrial DNA (mtDNA) induces inflammatory responses via the cGAS-STING pathway. Herein, we provide an overview of the mechanisms underlying mtDNA release following mitochondrial damage, focusing specifically on the association between mtDNA release-activated cGAS-STING signaling and the development of kidney diseases. Furthermore, we summarize the latest findings of cGAS-STING signaling pathway in cell, with a particular emphasis on its downstream signaling related to kidney diseases. This review intends to enhance our understanding of the intricate relationship among the cGAS-STING pathway, kidney diseases, and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Lu Li
- Department of Nephrology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang 310052, China
| | - Fei Liu
- Department of Nephrology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang 310052, China
| | - Chunyue Feng
- Department of Nephrology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang 310052, China
| | - Zhenjie Chen
- Department of Pediatric Intensive Care Unit, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang 310052, China
| | - Nan Zhang
- Department of Pediatric Intensive Care Unit, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang 310052, China
| | - Jianhua Mao
- Department of Nephrology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang 310052, China
| |
Collapse
|
7
|
Hensel IV, Éliás S, Steinhauer M, Stoll B, Benfatto S, Merkt W, Krienke S, Lorenz HM, Haas J, Wildemann B, Resnik-Docampo M. SLE serum induces altered goblet cell differentiation and leakiness in human intestinal organoids. EMBO Mol Med 2024; 16:547-574. [PMID: 38316934 PMCID: PMC10940301 DOI: 10.1038/s44321-024-00023-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/21/2023] [Accepted: 01/05/2024] [Indexed: 02/07/2024] Open
Abstract
Human intestinal epithelial cells are the interface between luminal content and basally residing immune cells. They form a tight monolayer that constantly secretes mucus creating a multilayered protective barrier. Alterations in this barrier can lead to increased permeability which is common in systemic lupus erythematosus (SLE) patients. However, it remains unexplored how the barrier is affected. Here, we present an in vitro model specifically designed to examine the effects of SLE on epithelial cells. We utilize human colon organoids that are stimulated with serum from SLE patients. Combining transcriptomic with functional analyses revealed that SLE serum induced an expression profile marked by a reduction of goblet cell markers and changed mucus composition. In addition, organoids exhibited imbalanced cellular composition along with enhanced permeability, altered mitochondrial function, and an interferon gene signature. Similarly, transcriptomic analysis of SLE colon biopsies revealed a downregulation of secretory markers. Our work uncovers a crucial connection between SLE and intestinal homeostasis that might be promoted in vivo through the blood, offering insights into the causal connection of barrier dysfunction and autoimmune diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Wolfgang Merkt
- Division of Rheumatology, Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Stefan Krienke
- Division of Rheumatology, Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Hanns-Martin Lorenz
- Division of Rheumatology, Department of Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Jürgen Haas
- Molecular Neuroimmunology Group, Department of Neurology, University of Heidelberg, Heidelberg, Germany
| | - Brigitte Wildemann
- Molecular Neuroimmunology Group, Department of Neurology, University of Heidelberg, Heidelberg, Germany
| | | |
Collapse
|
8
|
Moore R, Wang T, Macêdo MB, Lood C. Reply. Arthritis Rheumatol 2024; 76:489-490. [PMID: 37830244 DOI: 10.1002/art.42727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 09/25/2023] [Indexed: 10/14/2023]
Affiliation(s)
| | - Ting Wang
- University of Washington, Seattle, WA
| | | | | |
Collapse
|
9
|
Hansen FM, Kremer LS, Karayel O, Bludau I, Larsson NG, Kühl I, Mann M. Mitochondrial phosphoproteomes are functionally specialized across tissues. Life Sci Alliance 2024; 7:e202302147. [PMID: 37984987 PMCID: PMC10662294 DOI: 10.26508/lsa.202302147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 11/03/2023] [Accepted: 11/07/2023] [Indexed: 11/22/2023] Open
Abstract
Mitochondria are essential organelles whose dysfunction causes human pathologies that often manifest in a tissue-specific manner. Accordingly, mitochondrial fitness depends on versatile proteomes specialized to meet diverse tissue-specific requirements. Increasing evidence suggests that phosphorylation may play an important role in regulating tissue-specific mitochondrial functions and pathophysiology. Building on recent advances in mass spectrometry (MS)-based proteomics, we here quantitatively profile mitochondrial tissue proteomes along with their matching phosphoproteomes. We isolated mitochondria from mouse heart, skeletal muscle, brown adipose tissue, kidney, liver, brain, and spleen by differential centrifugation followed by separation on Percoll gradients and performed high-resolution MS analysis of the proteomes and phosphoproteomes. This in-depth map substantially quantifies known and predicted mitochondrial proteins and provides a resource of core and tissue-specific mitochondrial proteins (mitophos.de). Predicting kinase substrate associations for different mitochondrial compartments indicates tissue-specific regulation at the phosphoproteome level. Illustrating the functional value of our resource, we reproduce mitochondrial phosphorylation events on dynamin-related protein 1 responsible for its mitochondrial recruitment and fission initiation and describe phosphorylation clusters on MIGA2 linked to mitochondrial fusion.
Collapse
Affiliation(s)
- Fynn M Hansen
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Laura S Kremer
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ozge Karayel
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Isabell Bludau
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Nils-Göran Larsson
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Inge Kühl
- Department of Cell Biology, Institute of Integrative Biology of the Cell, UMR9198, CEA, CNRS, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| |
Collapse
|
10
|
Robert M, Scherlinger M. Platelets are a major player and represent a therapeutic opportunity in systemic lupus erythematosus. Joint Bone Spine 2024; 91:105622. [PMID: 37495075 DOI: 10.1016/j.jbspin.2023.105622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/04/2023] [Accepted: 07/12/2023] [Indexed: 07/28/2023]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by immune dysregulation and organ injury with a premature mortality due to cardiovascular diseases. Platelets, that are primarily known for their role in hemostasis, have been shown to play an active role in the pathogenesis and in the progression of immune-mediated inflammatory diseases. Here we summarize the evidence of their roles in SLE pathogenesis which supports the development of targeted treatments. Platelets and their precursors, the megakaryocytes, are intrinsically different in SLE patients compared with healthy controls. Different triggers related to innate and adaptive immunity activate platelets which release extracellular vesicles, soluble factors and interact with immune cells, thereby perpetuating inflammation. Platelets are involved in organ damage in SLE, especially in lupus nephritis and participate in the heightened cardiovascular mortality. They also play a clear role in antiphospholipid syndrome which can be associated with both thrombocytopenia and thrombosis. To tackle platelet activation and their interactions with immune cells now constitute promising therapeutic strategies in SLE.
Collapse
Affiliation(s)
- Marie Robert
- Service de médecine interne et immunologie clinique, centre hospitalier universitaire Édouard-Herriot, hospices civils de Lyon, Lyon, France
| | - Marc Scherlinger
- Service de rhumatologie, centre hospitalier universitaire de Strasbourg, 1, avenue Molière, 67098 Strasbourg, France; Laboratoire d'immuno-rhumatologie moléculaire, Institut national de la santé et de la recherche médicale (Inserm) UMR S 1109, Strasbourg, France; Centre national de référence des maladies auto-immunes et systémiques rares, Est/Sud-Ouest (RESO), France.
| |
Collapse
|
11
|
Shin YH, Bang S, Park SM, Ma X, Cassilly C, Graham D, Xavier R, Clardy J. Revisiting Coley's Toxins: Immunogenic Cardiolipins from Streptococcus pyogenes. J Am Chem Soc 2023; 145:21183-21188. [PMID: 37738205 PMCID: PMC10557101 DOI: 10.1021/jacs.3c07727] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Indexed: 09/24/2023]
Abstract
Coley's toxins, an early and enigmatic form of cancer (immuno)therapy, were based on preparations of Streptococcus pyogenes. As part of a program to explore bacterial metabolites with immunomodulatory potential, S. pyogenes metabolites were assayed in a cell-based immune assay, and a single membrane lipid, 18:1/18:0/18:1/18:0 cardiolipin, was identified. Its activity was profiled in additional cellular assays, which showed it to be an agonist of a TLR2-TLR1 signaling pathway with a 6 μM EC50 and robust TNF-α induction. A synthetic analog with switched acyl chains had no measurable activity in immune assays. The identification of a single immunogenic cardiolipin with a restricted structure-activity profile has implications for immune regulation, cancer immunotherapy, and poststreptococcal autoimmune diseases.
Collapse
Affiliation(s)
- Yern-Hyerk Shin
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School and Blavatnik Institute, Boston, Massachusetts 02115, United States
| | - Sunghee Bang
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School and Blavatnik Institute, Boston, Massachusetts 02115, United States
| | - Sung-Moo Park
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Department
of Molecular Biology and Center for the Study of Inflammatory Bowel
Disease, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Xiao Ma
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School and Blavatnik Institute, Boston, Massachusetts 02115, United States
| | - Chelsi Cassilly
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School and Blavatnik Institute, Boston, Massachusetts 02115, United States
| | - Daniel Graham
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Department
of Molecular Biology and Center for the Study of Inflammatory Bowel
Disease, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Ramnik Xavier
- Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Department
of Molecular Biology and Center for the Study of Inflammatory Bowel
Disease, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
| | - Jon Clardy
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School and Blavatnik Institute, Boston, Massachusetts 02115, United States
| |
Collapse
|
12
|
Pelletier M, Breton Y, Allaeys I, Becker Y, Benson T, Boilard E. Platelet extracellular vesicles and their mitochondrial content improve the mitochondrial bioenergetics of cellular immune recipients. Transfusion 2023; 63:1983-1996. [PMID: 37642274 DOI: 10.1111/trf.17524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 08/12/2023] [Accepted: 08/12/2023] [Indexed: 08/31/2023]
Abstract
BACKGROUND Mitochondria play a critical role in the production of cell energy and the regulation of cell death. Therefore, mitochondria orchestrate numerous cell effector functions, including fine-tuning the immune system. While mitochondria are mainly found intracellularly, they can escape the confine of the cell during the process of extracellular vesicle release. Platelets patrol blood vessels to ensure vasculature integrity and to support the immune system. In blood, platelets are the primary source of circulating mitochondria. Activated platelets produce extracellular vesicles, including a subset of mitochondria-containing vesicles. STUDY DESIGN AND METHODS We characterized mitochondrial functions in platelet-derived extracellular vesicles, and examined whether they could impact the bioenergetics of cellular immune recipients using an extracellular flux analyzer to measure real-time bioenergetics. RESULTS We validated that extracellular vesicles derived from activated platelets contain the necessary mitochondrial machinery to respirate and generate energy. Moreover, neutrophils and monocytes efficiently captured platelet-derived extracellular vesicles, enhancing their mitochondrial fitness. This process required functional mitochondria from donor platelets, as it was abolished by the inactivation of extracellular mitochondria using mitochondrial poison. DISCUSSION Together, the data suggest that extracellular mitochondria produced by platelets may support other metabolic functions through transcellular bioenergetics.
Collapse
Affiliation(s)
- Martin Pelletier
- Infectious and Immune Diseases Axis, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, Québec, Canada
- Centre de Recherche ARThrite - Arthrite, Recherche, Traitements, Université Laval, Québec, Québec, Canada
| | - Yann Breton
- Infectious and Immune Diseases Axis, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, Québec, Canada
- Centre de Recherche ARThrite - Arthrite, Recherche, Traitements, Université Laval, Québec, Québec, Canada
| | - Isabelle Allaeys
- Infectious and Immune Diseases Axis, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, Québec, Canada
- Centre de Recherche ARThrite - Arthrite, Recherche, Traitements, Université Laval, Québec, Québec, Canada
| | - Yann Becker
- Infectious and Immune Diseases Axis, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, Québec, Canada
- Centre de Recherche ARThrite - Arthrite, Recherche, Traitements, Université Laval, Québec, Québec, Canada
| | - Tom Benson
- Mitrix Bio Inc., Pleasanton, California, USA
| | - Eric Boilard
- Infectious and Immune Diseases Axis, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, Québec, Canada
- Centre de Recherche ARThrite - Arthrite, Recherche, Traitements, Université Laval, Québec, Québec, Canada
| |
Collapse
|
13
|
Duvvuri B, Pachman LM, Hermanson P, Wang T, Moore R, Ding-Hwa Wang D, Long A, Morgan GA, Doty S, Tian R, Sancak Y, Lood C. Role of mitochondria in the myopathy of juvenile dermatomyositis and implications for skeletal muscle calcinosis. J Autoimmun 2023; 138:103061. [PMID: 37244073 PMCID: PMC10330803 DOI: 10.1016/j.jaut.2023.103061] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/21/2023] [Accepted: 05/08/2023] [Indexed: 05/29/2023]
Abstract
OBJECTIVES To elucidate mechanisms contributing to skeletal muscle calcinosis in patients with juvenile dermatomyositis. METHODS A well-characterized cohorts of JDM (n = 68), disease controls (polymyositis, n = 7; juvenile SLE, n = 10, and RNP + overlap syndrome, n = 12), and age-matched health controls (n = 17) were analyzed for circulating levels of mitochondrial (mt) markers including mtDNA, mt-nd6, and anti-mitochondrial antibodies (AMAs) using standard qPCR, ELISA, and novel-in-house assays, respectively. Mitochondrial calcification of affected tissue biopsies was confirmed using electron microscopy and energy dispersive X-ray analysis. A human skeletal muscle cell line, RH30, was used to generate an in vitro calcification model. Intracellular calcification is measured by flow cytometry and microscopy. Mitochondria were assessed for mtROS production and membrane potential by flow cytometry and real-time oxygen consumption rate by Seahorse bioanalyzer. Inflammation (interferon-stimulated genes) was measured by qPCR. RESULTS In the current study, patients with JDM exhibited elevated levels of mitochondrial markers associated with muscle damage and calcinosis. Of particular interest are AMAs predictive of calcinosis. Human skeletal muscle cells undergo time- and dose-dependent accumulation of calcium phosphate salts with preferential localization to mitochondria. Calcification renders skeletal muscle cells mitochondria stressed, dysfunctional, destabilized, and interferogenic. Further, we report that inflammation induced by interferon-alpha amplifies mitochondrial calcification of human skeletal muscle cells via the generation of mitochondrial reactive oxygen species (mtROS). CONCLUSIONS Overall, our study demonstrates the mitochondrial involvement in the skeletal muscle pathology and calcinosis of JDM and mtROS as a central player in the calcification of human skeletal muscle cells. Therapeutic targeting of mtROS and/or upstream inducers, such as inflammation, may alleviate mitochondrial dysfunction, leading to calcinosis. AMAs can potentially identify patients with JDM at risk for developing calcinosis.
Collapse
Affiliation(s)
- Bhargavi Duvvuri
- Division of Rheumatology, University of Washington, Seattle, WA, USA.
| | - Lauren M Pachman
- Division of Pediatric Rheumatology, Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; CureJM Center of Excellence, Ann & Robert H. Lurie Children's Hospital of Chicago and the Stanley Manne Simpson-Quarrey Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Payton Hermanson
- Division of Rheumatology, University of Washington, Seattle, WA, USA
| | - Ting Wang
- Division of Rheumatology, University of Washington, Seattle, WA, USA
| | - Richard Moore
- Cedars Sinai Med Ctr, Division of Rheumatology, Los Angeles, CA, USA
| | | | - Aaron Long
- Department of Pharmacology, University of Washington, Seattle, WA, USA
| | - Gabrielle A Morgan
- Division of Pediatric Rheumatology, Department of Pediatrics, Ann & Robert H. Lurie Children's Hospital of Chicago, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; CureJM Center of Excellence, Ann & Robert H. Lurie Children's Hospital of Chicago and the Stanley Manne Simpson-Quarrey Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Rong Tian
- Mitochondria and Metabolism Center, University of Washington School of Medicine, Seattle, WA, USA
| | - Yasemin Sancak
- Department of Pharmacology, University of Washington, Seattle, WA, USA
| | - Christian Lood
- Division of Rheumatology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
14
|
Moore RE, Wang T, Duvvuri B, Feser ML, Deane KD, Solomon JJ, Lee Nelson J, Demoruelle MK, Lood C. Prediction of Erosive Disease Development by Antimitochondrial Antibodies in Rheumatoid Arthritis. Arthritis Rheumatol 2023; 75:890-899. [PMID: 36580020 PMCID: PMC10238559 DOI: 10.1002/art.42428] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 12/15/2022] [Accepted: 12/20/2022] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Mitochondria are found in the extracellular space in rheumatoid arthritis (RA). However, whether mitochondria are a source of autoantigens in RA has not been carefully addressed. Thus, we undertook this study to investigate the presence and significance of antimitochondrial antibodies (AMAs) in patients with RA. METHODS AMAs were measured in serum samples from 3 cross-sectional cohorts of RA patients (n = 95, n = 192, and n = 117) and healthy individuals (n = 38, n = 72, and n = 50) using a flow cytometry-based assay. Further, AMAs were detected using an anti-mitofusin-1 (anti-MFN-1) IgG enzyme-linked immunosorbent assay and Western blot analysis. A longitudinal inception cohort, followed up for a median of 8 years, was used to study disease progression. RESULTS AMA levels were elevated in RA patients from all 3 cohorts as compared to healthy individuals (P < 0.001, P < 0.05, and P < 0.01), with a range of 14-26% positivity. Levels of anti-MFN-1 antibodies correlated with AMA levels (r = 0.31, P = 0.006) and were elevated in RA patients as compared to healthy individuals (P < 0.001). The presence of AMAs was associated with erosive disease (P < 0.05) and interstitial lung disease (P < 0.01). Further, AMA levels were found to predict erosive disease (odds ratio [OR] 4.59, P = 0.006) and joint space narrowing (OR 3.08, P = 0.02) independent of anti-citrullinated protein antibodies. Finally, anti-MFN-1 antibodies identified seronegative patients developing erosive disease (OR 9.33; P = 0.02). CONCLUSION Our findings demonstrate the presence of novel autoantibodies targeting mitochondria in the setting of RA. AMAs were used to stratify patients based on disease phenotype and to predict development of erosive disease, including in patients with seronegative disease. Our results highlight the essential role of mitochondria in the pathogenesis of RA and suggest a possible benefit of therapies targeting mitochondrial-mediated inflammation and clearance in these patients.
Collapse
Affiliation(s)
- Richard E. Moore
- Division of Rheumatology, University of Washington, Seattle, WA, USA
| | - Ting Wang
- Division of Rheumatology, University of Washington, Seattle, WA, USA
| | - Bhargavi Duvvuri
- Division of Rheumatology, University of Washington, Seattle, WA, USA
| | - Marie L. Feser
- Division of Rheumatology, University of Colorado-Denver, Aurora, CO, USA
| | - Kevin D. Deane
- Division of Rheumatology, University of Colorado-Denver, Aurora, CO, USA
| | - Joshua J. Solomon
- Center for Interstitial Lung Disease, National Jewish Health, Denver, CO, USA
| | - J. Lee Nelson
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Christian Lood
- Division of Rheumatology, University of Washington, Seattle, WA, USA
| |
Collapse
|
15
|
Liedtke V, Rose L, Hiemann R, Nasser A, Rödiger S, Bonaventura A, Winkler L, Sowa M, Stöckle M, Schierack P, Junker K, Roggenbuck D. Over-Expression of LEDGF/p75 in HEp-2 Cells Enhances Autoimmune IgG Response in Patients with Benign Prostatic Hyperplasia-A Novel Diagnostic Approach with Therapeutic Consequence? Int J Mol Sci 2023; 24:ijms24076166. [PMID: 37047137 PMCID: PMC10093878 DOI: 10.3390/ijms24076166] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/20/2023] [Accepted: 03/23/2023] [Indexed: 04/14/2023] Open
Abstract
Lens epithelium-derived growth factor splice variant of 75 kDa (LEDGF/p75) is an autoantigen over-expressed in solid tumors and acts as a stress-related transcriptional co-activator. Participation of autoimmune responses in the pathophysiology of benign prostatic hyperplasia (PBH) and a corresponding immunosuppressive therapy by TNFalpha antagonists has been recently suggested. Thus, autoAb testing could aid in the diagnosis of BPH patients profiting from such therapy. We generated CRISPR/Cas9 modified HEp-2 LEDGF knock-out (KO) and HEp-2 LEDGF/p75 over-expressing (OE) cells and examined IgG autoantibody reactivity to LEDGF/p75 in patients with prostate cancer (PCa, n = 89), bladder cancer (BCa, n = 116), benign prostatic hyperplasia (BPH, n = 103), and blood donors (BD, n = 60) by indirect immunofluorescence assay (IFA). Surprisingly, we could not detect elevated binding of autoAbs against LEDGF/p75 in cancer patients, but autoAb reactivity to LEDGF/p75 OE cells in about 50% of patients with BPH was unexpectedly significantly increased. Furthermore, a line immunoassay enabling the detection of 18 different autoAbs revealed a significantly increased occurrence of anti-dsDNA autoAbs in 34% of BPH patients in contrast to tumor patients and BD. This finding was confirmed by anti-mitochondrial (mDNA) autoAb detection with the Crithidia luciliae immunofluorescence test, which also showed a significantly higher prevalence (34%) of anti-mDNA autoAbs in BPH. In summary, our study provided further evidence for the occurrence of autoimmune responses in BPH. Furthermore, LEDGF/p75 over-expression renders HEp-2 cells more autoantigenic and an ideal target for autoAb analysis in BPH with a potential therapy consequence.
Collapse
Affiliation(s)
- Victoria Liedtke
- Faculty Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany
| | - Laura Rose
- GA Generic Assays GmbH, 15827 Blankenfelde-Mahlow, Germany
| | - Rico Hiemann
- Faculty Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany
| | | | - Stefan Rödiger
- Faculty Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany
| | - Alena Bonaventura
- Department of Urology and Pediatric Urology, Saarland University, 66424 Homburg, Germany
| | - Laura Winkler
- Faculty Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany
| | - Mandy Sowa
- GA Generic Assays GmbH, 15827 Blankenfelde-Mahlow, Germany
| | - Michael Stöckle
- Department of Urology and Pediatric Urology, Saarland University, 66424 Homburg, Germany
| | - Peter Schierack
- Faculty Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany
| | - Kerstin Junker
- Department of Urology and Pediatric Urology, Saarland University, 66424 Homburg, Germany
| | - Dirk Roggenbuck
- Faculty Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany
- GA Generic Assays GmbH, 15827 Blankenfelde-Mahlow, Germany
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany
| |
Collapse
|
16
|
Xian H, Karin M. Oxidized mitochondrial DNA: a protective signal gone awry. Trends Immunol 2023; 44:188-200. [PMID: 36739208 DOI: 10.1016/j.it.2023.01.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/10/2023] [Accepted: 01/13/2023] [Indexed: 02/05/2023]
Abstract
Despite the emergence of mitochondria as key regulators of innate immunity, the mechanisms underlying the generation and release of immunostimulatory alarmins by stressed mitochondria remains nebulous. We propose that the major mitochondrial alarmin in myeloid cells is oxidized mitochondrial DNA (Ox-mtDNA). Fragmented Ox-mtDNA enters the cytosol where it activates the NLRP3 inflammasome and generates IL-1β, IL-18, and cGAS-STING to induce type I interferons and interferon-stimulated genes. Inflammasome activation further enables the circulatory release of Ox-mtDNA by opening gasdermin D pores. We summarize new data showing that, in addition to being an autoimmune disease biomarker, Ox-mtDNA converts beneficial transient inflammation into long-lasting immunopathology. We discuss how Ox-mtDNA induces short- and long-term immune activation, and highlight its homeostatic and immunopathogenic functions.
Collapse
Affiliation(s)
- Hongxu Xian
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego (UCSD), La Jolla, CA 92093, USA
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego (UCSD), La Jolla, CA 92093, USA.
| |
Collapse
|
17
|
Abstract
PURPOSE OF REVIEW Autoantibodies are cornerstone biomarkers in systemic lupus erythematosus (SLE), an autoimmune disease characterized by autoantibody-mediated tissue damage. Autoantibodies can inform about disease susceptibility, clinical course, outcomes and the cause of SLE. Identifying pathogenic autoantibodies in SLE, however, remains a significant challenge. This review summarizes recent advances in the field of autoantibodies in SLE. RECENT FINDINGS High-throughput technologies and innovative hypothesis have been applied to identify autoantibodies linked to pathogenic pathways in SLE. This work has led to the discovery of functional autoantibodies targeting key components is SLE pathogenesis (e.g. DNase1L3, cytokines, extracellular immunoregulatory receptors), as well as the identification of endogenous retroelements and interferon-induced proteins as sources of autoantigens in SLE. Others have reinvigorated the study of mitochondria, which has antigenic parallels with bacteria, as a trigger of autoantibodies in SLE, and identified faecal IgA to nuclear antigens as potential biomarkers linking gut permeability and microbial translocation in SLE pathogenesis. Recent studies showed that levels of autoantibodies against dsDNA, C1q, chromatin, Sm and ribosomal P may serve as biomarkers of proliferative lupus nephritis, and identified novel autoantibodies to several unique species of Ro52 overexpressed by SLE neutrophils. SUMMARY Autoantibodies hold promise as biomarkers of pathogenic mechanisms in SLE.
Collapse
Affiliation(s)
- Eduardo Gómez-Bañuelos
- Division of Rheumatology, The Johns Hopkins University School of Medicine. Baltimore, MD, 21224. U.S.A
| | - Andrea Fava
- Division of Rheumatology, The Johns Hopkins University School of Medicine. Baltimore, MD, 21224. U.S.A
| | - Felipe Andrade
- Division of Rheumatology, The Johns Hopkins University School of Medicine. Baltimore, MD, 21224. U.S.A
| |
Collapse
|
18
|
Becker YLC, Duvvuri B, Fortin PR, Lood C, Boilard E. The role of mitochondria in rheumatic diseases. Nat Rev Rheumatol 2022; 18:621-640. [PMID: 36175664 DOI: 10.1038/s41584-022-00834-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2022] [Indexed: 11/09/2022]
Abstract
The mitochondrion is an intracellular organelle thought to originate from endosymbiosis between an ancestral eukaryotic cell and an α-proteobacterium. Mitochondria are the powerhouses of the cell, and can control several important processes within the cell, such as cell death. Conversely, dysregulation of mitochondria possibly contributes to the pathophysiology of several autoimmune diseases. Defects in mitochondria can be caused by mutations in the mitochondrial genome or by chronic exposure to pro-inflammatory cytokines, including type I interferons. Following the release of intact mitochondria or mitochondrial components into the cytosol or the extracellular space, the bacteria-like molecular motifs of mitochondria can elicit pro-inflammatory responses by the innate immune system. Moreover, antibodies can target mitochondria in autoimmune diseases, suggesting an interplay between the adaptive immune system and mitochondria. In this Review, we discuss the roles of mitochondria in rheumatic diseases such as systemic lupus erythematosus, antiphospholipid syndrome and rheumatoid arthritis. An understanding of the different contributions of mitochondria to distinct rheumatic diseases or manifestations could permit the development of novel therapeutic strategies and the use of mitochondria-derived biomarkers to inform pathogenesis.
Collapse
Affiliation(s)
- Yann L C Becker
- Centre de Recherche ARThrite-Arthrite, Recherche et Traitements, Université Laval, Québec, QC, Canada
- Centre de Recherche du CHU de Québec-Université Laval, Axe Maladies infectieuses et immunitaires, Québec, QC, Canada
- Département de microbiologie et immunologie, Université Laval, Québec, QC, Canada
| | - Bhargavi Duvvuri
- Division of Rheumatology, University of Washington, Seattle, WA, USA
| | - Paul R Fortin
- Centre de Recherche ARThrite-Arthrite, Recherche et Traitements, Université Laval, Québec, QC, Canada
- Centre de Recherche du CHU de Québec-Université Laval, Axe Maladies infectieuses et immunitaires, Québec, QC, Canada
- Division of Rheumatology, Department of Medicine, CHU de Québec-Université Laval, Québec, QC, Canada
| | - Christian Lood
- Division of Rheumatology, University of Washington, Seattle, WA, USA.
| | - Eric Boilard
- Centre de Recherche ARThrite-Arthrite, Recherche et Traitements, Université Laval, Québec, QC, Canada.
- Centre de Recherche du CHU de Québec-Université Laval, Axe Maladies infectieuses et immunitaires, Québec, QC, Canada.
- Département de microbiologie et immunologie, Université Laval, Québec, QC, Canada.
| |
Collapse
|
19
|
De Benedittis G, Latini A, Colafrancesco S, Priori R, Perricone C, Novelli L, Borgiani P, Ciccacci C. Alteration of Mitochondrial DNA Copy Number and Increased Expression Levels of Mitochondrial Dynamics-Related Genes in Sjögren's Syndrome. Biomedicines 2022; 10:2699. [PMID: 36359219 PMCID: PMC9687724 DOI: 10.3390/biomedicines10112699] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 07/26/2023] Open
Abstract
Sjögren's syndrome (SS) is a chronic autoimmune multifactorial disease characterized by inflammation and lymphocytic infiltration of the exocrine glands. Several studies have highlighted the involvement of oxidative stress in this pathology, suggesting that it could induce mitochondrial dysfunctions. Mitochondria could have a role in inflammatory and immune processes. Since the mitochondrial DNA (mtDNA) copy number could change in response to physiological or environmental stimuli, this study aimed to evaluate possible alterations in the mtDNA copy number in SS. We have analyzed the amount of mtDNA in the peripheral blood of 74 SS patients and 61 healthy controls by qPCR. Then, since mitochondrial fusion and fission play a crucial role in maintaining the number of mitochondria, we investigated the expression variability of the genes most commonly involved in mitochondrial dynamics in a subgroup of SS patients and healthy controls. Interestingly, we observed a highly significant decrease in mtDNA copies in the SS patients compared to healthy controls (p = 1.44 × 10-12). Expression levels of mitochondrial fission factor (MFF), mitofusin-1 (MFN1), and mitochondrial transcription factor A (TFAM) genes were analyzed, showing a statistically significant increase in the expression of MFF (p = 0.003) and TFAM (p = 0.022) in the SS patients compared to healthy controls. These results give further insight into the possible involvement of mitochondrial dysfunctions in SS disease.
Collapse
Affiliation(s)
- Giada De Benedittis
- Genetics Section, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Andrea Latini
- Genetics Section, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Serena Colafrancesco
- Division of Rheumatology, Department of Clinical Internal, Anaesthesiologic and Cardiovascular Sciences, Sapienza University, 00133 Rome, Italy
| | - Roberta Priori
- Division of Rheumatology, Department of Clinical Internal, Anaesthesiologic and Cardiovascular Sciences, Sapienza University, 00133 Rome, Italy
- UniCamillus—Saint Camillus International University of Health Sciences, 00131 Rome, Italy
| | - Carlo Perricone
- Rheumatology Department of Medicine, University of Perugia, Piazzale Giorgio Menghini 1, 06129 Perugia, Italy
| | - Lucia Novelli
- UniCamillus—Saint Camillus International University of Health Sciences, 00131 Rome, Italy
| | - Paola Borgiani
- Genetics Section, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Cinzia Ciccacci
- UniCamillus—Saint Camillus International University of Health Sciences, 00131 Rome, Italy
| |
Collapse
|
20
|
Quintero-González DC, Muñoz-Urbano M, Vásquez G. Mitochondria as a key player in systemic lupus erythematosus. Autoimmunity 2022; 55:497-505. [PMID: 35978536 DOI: 10.1080/08916934.2022.2112181] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Systemic lupus erythematosus (SLE) is a heterogeneous, multisystemic autoimmune disease with a broad clinical spectrum. Loss of self-tolerance and chronic inflammation are critical markers of SLE pathogenesis. Although alterations in adaptive immunity are widely recognized, increasing reports indicate the role of mitochondrial dysfunction in activating pathogenic pathways involving the innate immune system. Among these, disarrangements in mitochondrial DNA copy number and heteroplasmy percentage are related to SLE activity. Furthermore, increased oxidative stress contributes to post-translational changes in different molecules (proteins, nucleic acids, and lipids), release of oxidized mitochondrial DNA through a pore of voltage-dependent anion channel oligomers, and spontaneous mitochondrial antiviral signaling protein oligomerization. Finally, a reduction in mitophagy, apoptosis induction, and NETosis has been reported in SLE. Most of these pathways lead to persistent and inappropriate exposure to oxidized mitochondrial DNA, which can stimulate plasmacytoid dendritic cells, enhance autoreactive lymphocyte activation, and release increased amounts of interferons through stimulation of toll-like receptors and cytosolic DNA sensors. Likewise, abnormal T-cell receptor activation, decreased regulatory T cells, enhanced Th17 phenotypes, and increased monocyte maturation to dendritic cells have also been observed in SLE. Targeting the players involved in mitochondrial damage can ultimately help.
Collapse
Affiliation(s)
| | - Marcela Muñoz-Urbano
- Rheumatology Section, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - G Vásquez
- Rheumatology Section, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia.,Grupo de Inmunología Celular e Inmunogenética (GICIC), Universidad de Antioquia, Medellín, Colombia
| |
Collapse
|
21
|
Boilard E, Bellio M. Platelet extracellular vesicles and the secretory interactome join forces in health and disease. Immunol Rev 2022; 312:38-51. [PMID: 35899405 DOI: 10.1111/imr.13119] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Extracellular vesicles (EVs) are small membrane-bound vesicles released by cells under various conditions. They are found in the extracellular milieu in all biological fluids. As the concentrations, contents, and origin of EVs can change during inflammation, the assessment of EVs can be used as a proxy of cellular activation. Here, we review the literature regarding EVs, more particularly those released by platelets and their mother cells, the megakaryocytes. Their cargo includes cytokines, growth factors, organelles (mitochondria and proteasomes), nucleic acids (messenger and non-coding RNA), transcription factors, and autoantigens. EVs may thus contribute to intercellular communication by facilitating exchange of material between cells. EVs also interact with other molecules secreted by cells. In autoimmune diseases, EVs are associated with antibodies secreted by B cells. By definition, EVs necessarily comprise a phospholipid moiety, which is thus the target of secreted phospholipases also abundantly expressed in the extracellular milieu. We discuss how platelet-derived EVs, which represent the majority of the circulating EVs, may contribute to immunity through the activity of their cargo or in combination with the secretory interactome.
Collapse
Affiliation(s)
- Eric Boilard
- Département de microbiologie-immunologie, Faculté de médecine, Université Laval, Québec, QC, Canada.,Axe maladies infectieuses et immunitaires, Centre de recherche du CHU de Québec - Université Laval, Québec, QC, Canada.,Centre de recherche ARThrite, Université Laval, Québec, QC, Canada
| | - Marie Bellio
- Département de microbiologie-immunologie, Faculté de médecine, Université Laval, Québec, QC, Canada.,Axe maladies infectieuses et immunitaires, Centre de recherche du CHU de Québec - Université Laval, Québec, QC, Canada.,Centre de recherche ARThrite, Université Laval, Québec, QC, Canada
| |
Collapse
|
22
|
Zhao L, Hu X, Xiao F, Zhang X, Zhao L, Wang M. Mitochondrial impairment and repair in the pathogenesis of systemic lupus erythematosus. Front Immunol 2022; 13:929520. [PMID: 35958572 PMCID: PMC9358979 DOI: 10.3389/fimmu.2022.929520] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/28/2022] [Indexed: 12/12/2022] Open
Abstract
Nucleic acid autoantibodies, increase type I interferon (IFN-α) levels, and immune cell hyperactivation are hallmarks of systemic lupus erythematosus (SLE). Notably, immune cell activation requires high level of cellular energy that is predominately generated by the mitochondria. Mitochondrial reactive oxygen species (mROS), the byproduct of mitochondrial energy generation, serves as an essential mediator to control the activation and differentiation of cells and regulate the antigenicity of oxidized nucleoids within the mitochondria. Recently, clinical trials on normalization of mitochondrial redox imbalance by mROS scavengers and those investigating the recovery of defective mitophagy have provided novel insights into SLE prophylaxis and therapy. However, the precise mechanism underlying the role of oxidative stress-related mitochondrial molecules in skewing the cell fate at the molecular level remains unclear. This review outlines distinctive mitochondrial functions and pathways that are involved in immune responses and systematically delineates how mitochondrial dysfunction contributes to SLE pathogenesis. In addition, we provide a comprehensive overview of damaged mitochondrial function and impaired metabolic pathways in adaptive and innate immune cells and lupus-induced organ tissues. Furthermore, we summarize the potential of current mitochondria-targeting drugs for SLE treatment. Developing novel therapeutic approaches to regulate mitochondrial oxidative stress is a promising endeavor in the search for effective treatments for systemic autoimmune diseases, particularly SLE.
Collapse
Affiliation(s)
- Like Zhao
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xianda Hu
- Beijing Tibetan Hospital, China Tibetology Research Center, Beijing, China
| | - Fei Xiao
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Clinical Biobank, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Xuan Zhang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lidan Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science and Technology, Beijing, China
- *Correspondence: Min Wang, ; Lidan Zhao,
| | - Min Wang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Min Wang, ; Lidan Zhao,
| |
Collapse
|
23
|
Rasquinha MT, Lasrado N, Petro-Turnquist E, Weaver E, Venkataraman T, Anderson D, Laserson U, Larman HB, Reddy J. PhIP-Seq Reveals Autoantibodies for Ubiquitously Expressed Antigens in Viral Myocarditis. BIOLOGY 2022; 11:biology11071055. [PMID: 36101433 PMCID: PMC9312229 DOI: 10.3390/biology11071055] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/09/2022] [Accepted: 07/11/2022] [Indexed: 12/01/2022]
Abstract
Simple Summary Myocarditis is the inflammation of the heart muscle, and viral infections are a common cause of this disease. Myocarditis in some patients can progress to dilated cardiomyopathy (DCM). The mouse model of coxsackievirus B3 (CVB3) is commonly used to understand this disease progression in DCM patients. In this paper, we have attempted to analyze antibodies for heart antigens that could be produced as a result of heart damage in animals infected with CVB3 using a technique called Phage ImmunoPrecipitation Sequencing (PhIP-Seq). The analyses led us to identify antibodies for several proteins that were not previously reported that may have relevance to human disease. Abstract Enteroviruses such as group B coxsackieviruses (CVB) are commonly suspected as causes of myocarditis that can lead to dilated cardiomyopathy (DCM), and the mouse model of CVB3 myocarditis is routinely used to understand DCM pathogenesis. Mechanistically, autoimmunity is suspected due to the presence of autoantibodies for select antigens. However, their role continues to be enigmatic, which also raises the question of whether the breadth of autoantibodies is sufficiently characterized. Here, we attempted to comprehensively analyze the autoantibody repertoire using Phage ImmunoPrecipitation Sequencing (PhIP-Seq), a versatile and high-throughput platform, in the mouse model of CVB3 myocarditis. First, PhIP-Seq analysis using the VirScan library revealed antibody reactivity only to CVB3 in the infected group but not in controls, thus validating the technique in this model. Second, using the mouse peptide library, we detected autoantibodies to 32 peptides from 25 proteins in infected animals that are ubiquitously expressed and have not been previously reported. Third, by using ELISA as a secondary assay, we confirmed antibody reactivity in sera from CVB3-infected animals to cytochrome c oxidase assembly factor 4 homolog (COA4) and phosphoinositide-3-kinase adaptor protein 1 (PIK3AP1), indicating the specificity of antibody detection by PhIP-Seq technology. Fourth, we noted similar antibody reactivity patterns in CVB3 and CVB4 infections, suggesting that the COA4- and PIK3AP1-reactive antibodies could be common to multiple CVB infections. The specificity of the autoantibodies was affirmed with influenza-infected animals that showed no reactivity to any of the antigens tested. Taken together, our data suggest that the autoantibodies identified by PhIP-Seq may have relevance to CVB pathogenesis, with a possibility that similar reactivity could be expected in human DCM patients.
Collapse
Affiliation(s)
- Mahima T. Rasquinha
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (M.T.R.); (N.L.)
| | - Ninaad Lasrado
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (M.T.R.); (N.L.)
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Erika Petro-Turnquist
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (E.P.-T.); (E.W.)
| | - Eric Weaver
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (E.P.-T.); (E.W.)
| | - Thiagarajan Venkataraman
- Division of Immunology, Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA;
| | - Daniel Anderson
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Uri Laserson
- Department of Genetics and Genomic Sciences and Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - H. Benjamin Larman
- Division of Immunology, Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA;
- Correspondence: (H.B.L.); (J.R.); Tel.: +1-(410)-614-6525 (H.B.L); +1-(402)-472-8541 (J.R.)
| | - Jay Reddy
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA; (M.T.R.); (N.L.)
- Correspondence: (H.B.L.); (J.R.); Tel.: +1-(410)-614-6525 (H.B.L); +1-(402)-472-8541 (J.R.)
| |
Collapse
|
24
|
Maleki F, Salimi M, Shirkoohi R, Rezaei M. Mitotherapy in doxorubicin induced cardiotoxicity: A promising strategy to reduce the complications of treatment. Life Sci 2022; 304:120701. [PMID: 35690107 DOI: 10.1016/j.lfs.2022.120701] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/30/2022] [Accepted: 06/06/2022] [Indexed: 11/19/2022]
Abstract
AIMS Doxorubicin is a potent and broad-spectrum antineoplastic medication prescribed for both solid and hematological malignancies. Despite its value, the clinical use of doxorubicin is limited due to cardio-oncologic complication and cardiotoxic adverse effect. Among the mechanisms proposed for its toxicity, mitochondrial dysfunction has gained more attention. Therefore, if damaged mitochondria are replaced by normal efficient mitochondria, cardiac toxicity is expected to be reduced or improved. In this way, we have studied the efficiency of transplantation of freshly isolated rat liver mitochondria in neonatal rat cardiomyocytes that have been damaged by doxorubicin. MATERIALS AND METHODS For this purpose, isolated mitochondria were characterized using mitochondrial complex II, membrane potential and swelling evaluations, and also fluorescence and electron microscopy. Afterward, the effect of mitotherapy on the damaged cardiomyocytes was investigated by using annexin V/PI staining, MTT, ROS, MMP, lipid peroxidation, GSH and ATP evaluations. KEY FINDINGS AND SIGNIFICANCE Transplanted mitochondria could remarkably enter the neonatal rat cardiomyocytes. Addition of mitochondria to the damaged cardiomyocytes, significantly increased cell viability by reducing the level of reactive oxygen species and lipid peroxidation, increasing of ∆Ψ, ATP and GSH contents and decreasing of apoptotic and necrotic cell death. Our results showed that mitotherapy has a significant restorative effect on cardiotoxicity induced by doxorubicin, which promises a better future to reduce the complications of cancer treatment.
Collapse
Affiliation(s)
- Farshid Maleki
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mona Salimi
- Physiology and Pharmacology Department, Pasteur Institute of Iran, Tehran, Iran
| | - Reza Shirkoohi
- Cancer Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Rezaei
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
25
|
Linge CP, Jern A, Tydén H, Gullstrand B, Yan H, Welinder C, Kahn R, Jönssen A, Semple JW, Bengtsson AA. Enrichment of complement, immunoglobulins and autoantibody targets in the proteome of platelets from patients with Systemic Lupus Erythematosus (SLE). Thromb Haemost 2022; 122:1486-1501. [PMID: 35419777 PMCID: PMC9420555 DOI: 10.1055/a-1825-2915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Background
Systemic lupus erythematosus (SLE) is a complex disease characterized by autoimmunity toward apoptotic cells, excessive amounts of circulating immune complexes, and complement activation. A decreased platelet size has been observed in SLE and their nonhemostatic functions may play an active role in the disease. The main objective of this study was to find clues that could explain their decreased size and functional role, analyzing the entire platelet proteome.
Methods
Platelets were isolated from 23 patients with SLE. The five individuals with the highest and lowest average platelet forward scatter were selected for further analysis. Platelet protein content was analyzed using liquid chromatography with tandem mass spectrometry (LC-MS/MS) and compared with platelets from five healthy controls. Data are available via ProteomeXchange with identifier PXD031202.
Results
Out of 2,572 proteins identified, 396 had significantly different levels (ANOVA
q
-value ≤ 0.01). Forty proteins, including immunoglobulin-, complement- and phosphatidylserine-binding proteins had higher abundance in platelets from SLE patients, largely independent of size (fold difference of ≥1.5 and a
t
-test
p
-value of ≤0.05 as cut-off). Functional characterization revealed increased degranulation and skewed hemostatic balance in platelets from SLE patients. In the SLE proteome, immunoglobulin proteins were negatively correlated to serum complement C3 and C4 and the highest relative levels were detected in platelets of normal size.
Conclusion
Platelets from SLE patients shared a specific protein profile, including immunoglobulins, complement proteins, and autoantigens, largely independent of the platelet size and in agreement with an integrated role for platelets in SLE.
Collapse
Affiliation(s)
- Carl Petrus Linge
- Department of Clinical Sciences Lund, Lund University Section for Molecular Skeletal Biology and Rheumatology, Lund, Sweden
| | - Andreas Jern
- Department of Clinical Sciences, Lund University Section for Molecular Skeletal Biology and Rheumatology, Lund, Sweden
| | - Helena Tydén
- Department of Clinical Sciences, Lund University Section for Molecular Skeletal Biology and Rheumatology, Lund, Sweden
| | - Birgitta Gullstrand
- Department of Clinical Sciences, Lund University Section for Molecular Skeletal Biology and Rheumatology, Lund, Sweden
| | - Hong Yan
- BioMS, Swedish National Infrastructure for Biological Mass Spectrometry, Lund, Sweden
| | - Charlotte Welinder
- Department of Clinical Sciences Lund, Lund University Department of Oncology and Pathology, Lund, Sweden
| | - Robin Kahn
- Wallenberg Center for Molecular Medicin, Lund University Faculty of Medicine, Lund, Sweden.,Paediatrics, Lund University Faculty of Medicine, Lund, Sweden
| | - Andreas Jönssen
- Department of Clinical Sciences Lund, Lund University Section for Molecular Skeletal Biology and Rheumatology, Lund, Sweden
| | - John W Semple
- Transfusion Medicine, Lunds Universitet, Lund, Sweden
| | - Anders A Bengtsson
- Department of Clinical Sciences, Lund University Section for Molecular Skeletal Biology and Rheumatology, Lund, Sweden
| |
Collapse
|
26
|
Gaur PS, R N, Anuja AK, Singh MK, Rai MK, Muhammed R, Sahu AK, Agarwal V, Gupta L. Anti-mitochondrial antibodies in Indian patients with idiopathic inflammatory myopathies. Int J Rheum Dis 2022; 25:659-668. [PMID: 35384316 DOI: 10.1111/1756-185x.14320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 01/27/2022] [Accepted: 03/18/2022] [Indexed: 12/01/2022]
Abstract
AIMS Anti-mitochondrial antibodies (AMAs) are associated with distinct clinical phenotypes including cardiac and hepatic manifestations in idiopathic inflammatory myopathies (IIMs). This article studies the prevalence, clinical characteristics and outcomes of AMA in Indian patients with IIM. METHODS Patients (97: 81 adult, 16 juvenile) clinically diagnosed with polymyositis or antibody-negative IIM were retrieved from the MyoCite bio-archive. They were tested for myositis-specific autoantibodies / myositis autoantibodies (MSAs/MAAs) using line immunoassay and antinuclear antibodies and AMAs using immunofluorescence assay (IFA). Patients were also screened for cardiac biomarkers (cardiac troponin I [c-TnI] and N terminal-pro brain natriuretic peptide [NT-pro-BNP] using immunometric immunoassay technique and enhanced chemiluminescence assay testing respectively) and hepatic manifestations using AMA testing. Results were formulated after carrying out analytical tests. RESULTS Of the cohort, 5 adults (6.2%) (M:F 0:1) with a median age and disease duration of 37 years and 2 months respectively, tested AMA+ while the children tested negative. Dermatomyositis was the commonest phenotype, with amyopathic forms being common, often with MSA positivity. Cancer-associated myositis and polymyositis were also seen. AMA positivity is associated with Gottron's sign and calcinosis. Comparable levels of C-TnI and NT-pro-BNP and AMA testing in patients help to rule out subclinical cardiac and hepatic involvement respectively. CONCLUSION Anti-mitochondrial antibodies are rare (6.2%) in different subtypes of IIM in the Indian population, and often coexist with MSAs. Their negative association with cardiac and hepatic involvement and probable association with Gottron's sign and calcinosis merit further investigation and long-term follow-up to understand the entire spectrum of the disease.
Collapse
Affiliation(s)
| | - Naveen R
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Anamika Kumari Anuja
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Mantabya Kumar Singh
- Department of Nephrology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Mohit Kumar Rai
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Rizwan Muhammed
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Ankit Kumar Sahu
- Department of Cardiology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Vikas Agarwal
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Latika Gupta
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India.,Department of Rheumatology, Royal Wolverhampton Hospitals NHS Trust, Wolverhampton, UK.,City Hospital, Sandwell and West Birmingham Hospitals NHS Trust, Birmingham, UK.,Division of Musculoskeletal and Dermatological Sciences, Centre for Musculoskeletal Research, School of Biological Sciences, The University of Manchester, Manchester, UK
| |
Collapse
|
27
|
Becker YL, Gagné JP, Julien AS, Lévesque T, Allaeys I, Gougeard N, Rubio V, Boisvert FM, Jean D, Wagner E, Poirier GG, Fortin PR, Boilard É. Identification of mitofusin 1 and complement component 1q subcomponent-binding protein as mitochondrial targets in systemic lupus erythematosus. Arthritis Rheumatol 2022; 74:1193-1203. [PMID: 35128841 DOI: 10.1002/art.42082] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 12/28/2021] [Accepted: 02/01/2022] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Mitochondria are organelles that possess several bacterial features such as a double-stranded genome with hypomethylated CpG islets, formylated proteins, and cardiolipin-containing membranes. In systemic lupus erythematosus (SLE), mitochondria and their inner components are released into the extracellular space, potentially eliciting a pro-inflammatory response by the immune system. While cardiolipin and mitochondrial DNA and RNA are confirmed targets of autoantibodies, other antigenic mitochondrial proteins in SLE remain to be identified. Herein, we aim to characterize the protein repertoire recognized by anti-mitochondrial antibodies (AMA) in SLE patients. METHODS Using shotgun proteomic profiling, we identified 1345 proteins, 431 of which were associated with the mitochondrial proteome. Immunoreactivities to several of these candidates were assessed by direct ELISA in serum samples from a local cohort (healthy: n=30, SLE: n=87) and associated with demographic and disease characteristics. RESULTS We determined that IgGs to the C1q-binding protein (C1qBP) are significantly elevated in SLE patients included in our cohort (p=0.049) and are associated with positivity for lupus anticoagulant (p=0.049). IgG against the mitochondrial protein mitofusin 1 (Mfn1) displayed promising performances in the prediction of SLE diagnoses (aOR: 2.99, 95%CI: 1.39-6.43, p=0.0044) in our cohort. Moreover, anti-Mfn1 were associated with positivity to anti-phospholipids (p=0.011) and anti-dsDNA (p=0.0005). CONCLUSION This study presents the mitochondrial repertoire targeted in SLE, indicating that autoantibodies can recognize secreted and/or surface proteins of mitochondrial origin. Profiling of the AMA repertoire in large prospective cohorts may improve our knowledge on mitochondrial biomarkers and their usefulness for patient stratification.
Collapse
Affiliation(s)
- Yann Lc Becker
- Centre de Recherche ARThrite - Arthrite, Recherche et Traitements, Université Laval, Québec, Qc, Canada.,Axe Maladies infectieuses et immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Québec, Qc, Canada.,Département de microbiologie et immunologie, Université Laval, Québec, Qc, Canada
| | - Jean-Philippe Gagné
- Axe Maladies infectieuses et immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Québec, Qc, Canada.,Laboratoire d'Immunologie et Histocompatibilité, CHU de Québec-Université Laval, Département de Médecine de Laboratoire, Québec, Qc, Canada
| | - Anne-Sophie Julien
- Département de mathématiques et statistique, Université Laval, Québec, Qc, Canada
| | - Tania Lévesque
- Centre de Recherche ARThrite - Arthrite, Recherche et Traitements, Université Laval, Québec, Qc, Canada.,Axe Maladies infectieuses et immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Québec, Qc, Canada.,Département de microbiologie et immunologie, Université Laval, Québec, Qc, Canada
| | - Isabelle Allaeys
- Centre de Recherche ARThrite - Arthrite, Recherche et Traitements, Université Laval, Québec, Qc, Canada.,Axe Maladies infectieuses et immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Québec, Qc, Canada.,Département de microbiologie et immunologie, Université Laval, Québec, Qc, Canada
| | - Nadine Gougeard
- Structural Enzymopathology Unit, Instituto de Biomedicina de Valencia of the CSIC (IBV-CSIC), Valencia, Spain.,Group 739, Centro de Investigación Biomédica en Red para Enfermedades Raras (CIBERER-ISCIII), Madrid, Spain
| | - Vicente Rubio
- Structural Enzymopathology Unit, Instituto de Biomedicina de Valencia of the CSIC (IBV-CSIC), Valencia, Spain.,Group 739, Centro de Investigación Biomédica en Red para Enfermedades Raras (CIBERER-ISCIII), Madrid, Spain
| | | | - Dominique Jean
- Department of Anatomy and Cell Biology, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Eric Wagner
- Département de microbiologie et immunologie, Université Laval, Québec, Qc, Canada.,Laboratoire d'Immunologie et Histocompatibilité, CHU de Québec-Université Laval, Département de Médecine de Laboratoire, Québec, Qc, Canada
| | - Guy G Poirier
- Axe Maladies infectieuses et immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Québec, Qc, Canada.,Département de biologie moléculaire, de biochimie médicale et de pathologie, Faculté de Médecine, Université Laval, Québec, Qc, Canada
| | - Paul R Fortin
- Centre de Recherche ARThrite - Arthrite, Recherche et Traitements, Université Laval, Québec, Qc, Canada.,Division de Rhumatologie, Département de Médecine, CHU de Québec - Université Laval, Québec, Qc, Canada
| | - Éric Boilard
- Centre de Recherche ARThrite - Arthrite, Recherche et Traitements, Université Laval, Québec, Qc, Canada.,Axe Maladies infectieuses et immunitaires, Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Québec, Qc, Canada.,Département de microbiologie et immunologie, Université Laval, Québec, Qc, Canada
| |
Collapse
|
28
|
Colapietro F, Lleo A, Generali E. Antimitochondrial Antibodies: from Bench to Bedside. Clin Rev Allergy Immunol 2022; 63:166-177. [PMID: 34586589 PMCID: PMC8480115 DOI: 10.1007/s12016-021-08904-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2021] [Indexed: 01/13/2023]
Abstract
Anti-mitochondrial antibodies (AMA) are directed against the E2 subunits of the 2-oxo acid dehydrogenase complexes (PDC-E2) and are the typical biomarkers of primary biliary cholangitis (PBC), being present in 90-95% of patients, with increasing sensitivity at increasing titers. Albeit being highly specific for PBC diagnosis, AMA can be detected in less than 1% of healthy subjects, and thus the management subjects with no sign or symptom of liver disease is still a challenge and data concerning clinical risk of developing PBC in this subgroup of patients are controversial. Moreover, AMA can also be detected in patients affected by overlap syndrome, as well as hepatic diseases (i.e., NASH and viral hepatitis), while the association with autoimmune diseases, in particular Sjögren's syndrome, systemic sclerosis, and systemic lupus erythematosus, is well established. Furthermore, new associations are being identified with inflammatory myositis and heart disease. AMA are directed towards the pyruvate dehydrogenase multi enzyme complex (PDC-E2) subunit, which represents an epithelial specific autoantigen for PBC. This review focuses on the main characteristics of AMA, their association with autoimmune diseases and liver diseases.
Collapse
Affiliation(s)
- Francesca Colapietro
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Division of Internal Medicine and Hepatology, Department of Gastroenterology, Humanitas Research Hospital IRCCS, Rozzano, Milan, Italy
| | - Ana Lleo
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.
- Division of Internal Medicine and Hepatology, Department of Gastroenterology, Humanitas Research Hospital IRCCS, Rozzano, Milan, Italy.
| | - Elena Generali
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- Division of Internal Medicine and Hepatology, Department of Gastroenterology, Humanitas Research Hospital IRCCS, Rozzano, Milan, Italy
| |
Collapse
|
29
|
Brunekreef T, Limper M, Melchers R, Mathsson-Alm L, Dias J, Hoefer I, Haitjema S, van Laar JM, Otten H. Microarray testing in patients with systemic lupus erythematosus identifies a high prevalence of CpG DNA-binding antibodies. Lupus Sci Med 2021; 8:8/1/e000531. [PMID: 34725184 PMCID: PMC8562534 DOI: 10.1136/lupus-2021-000531] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 10/11/2021] [Indexed: 12/13/2022]
Abstract
Objective Many autoantibodies are known to be associated with SLE, although their role in clinical practice is limited because of low sensitivity and weak associations with clinical manifestations. There has been great interest in the discovery of new autoantibodies to use in clinical practice. In this study, we investigated 57 new and known antibodies and their potential for diagnostics or risk stratification. Methods Between 2014 and 2017, residual sera of all anti-dsDNA tests in the UMC Utrecht were stored in a biobank. This included sera of patients with SLE, patients with a diagnosis of another immune-mediated inflammatory disease (IMID), patients with low (non-IMID) or medium levels of clinical suspicion of SLE but no IMID diagnosis (Rest), and self-reported healthy blood bank donors. Diagnosis and (presence of) symptoms at each blood draw were retrospectively assessed in the patient records with the Utrecht Patient-Oriented Database using a newly developed text mining algorithm. Sera of patients were analysed for the presence of 57 autoantibodies with a custom-made immunofluorescent microarray. Signal intensity cut-offs for all antigens on the microarray were set to the 95th percentile of the non-IMID control group. Differences in prevalence of autoantibodies between patients with SLE and control groups were assessed. Results Autoantibody profiles of 483 patients with SLE were compared with autoantibody profiles of 1397 patients from 4 different control groups. Anti-dsDNA was the most distinguishing feature between patients with SLE and other patients, followed by antibodies against Cytosine-phosphate-Guanine (anti-CpG) DNA motifs (p<0.0001). Antibodies against CMV (cytomegalovirus) and ASCA (anti-Saccharomyces cerevisiae antibodies) were more prevalent in patients with SLE with (a history of) lupus nephritis than patients with SLE without nephritis. Conclusion Antibodies against CpG DNA motifs are prevalent in patients with SLE. Anti-CMV antibodies are associated with lupus nephritis.
Collapse
Affiliation(s)
- Tammo Brunekreef
- Rheumatology & Clinical Immunology, UMC Utrecht, Utrecht, The Netherlands
| | - Maarten Limper
- Rheumatology & Clinical Immunology, UMC Utrecht, Utrecht, The Netherlands
| | - Rowena Melchers
- Center of Translational Immunology, UMC Utrecht, Utrecht, The Netherlands
| | | | - Jorge Dias
- ImmunoDiagnostics Division, Thermo Fisher Scientific, Uppsala, Sweden
| | - Imo Hoefer
- Clinical Diagnostic Laboratory, UMC Utrecht, Utrecht, The Netherlands
| | - Saskia Haitjema
- Clinical Diagnostic Laboratory, UMC Utrecht, Utrecht, The Netherlands
| | - Jacob M van Laar
- Rheumatology & Clinical Immunology, UMC Utrecht, Utrecht, The Netherlands
| | - Henny Otten
- Center of Translational Immunology, UMC Utrecht, Utrecht, The Netherlands
| |
Collapse
|
30
|
Zanini G, De Gaetano A, Selleri V, Savino G, Cossarizza A, Pinti M, Mattioli AV, Nasi M. Mitochondrial DNA and Exercise: Implications for Health and Injuries in Sports. Cells 2021; 10:cells10102575. [PMID: 34685555 PMCID: PMC8533813 DOI: 10.3390/cells10102575] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/21/2021] [Accepted: 09/24/2021] [Indexed: 12/31/2022] Open
Abstract
Recently, several studies have highlighted the tight connection between mitochondria and physical activity. Mitochondrial functions are important in high-demanding metabolic activities, such as endurance sports. Moreover, regular training positively affects metabolic health by increasing mitochondrial oxidative capacity and regulating glucose metabolism. Exercise could have multiple effects, also on the mitochondrial DNA (mtDNA) and vice versa; some studies have investigated how mtDNA polymorphisms can affect the performance of general athletes and mtDNA haplogroups seem to be related to the performance of elite endurance athletes. Along with several stimuli, including pathogens, stress, trauma, and reactive oxygen species, acute and intense exercise also seem to be responsible for mtDNA release into the cytoplasm and extracellular space, leading to the activation of the innate immune response. In addition, several sports are characterized by a higher frequency of injuries, including cranial trauma, associated with neurological consequences. However, with regular exercise, circulating cell-free mtDNA levels are kept low, perhaps promoting cf-mtDNA removal, acting as a protective factor against inflammation.
Collapse
Affiliation(s)
- Giada Zanini
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (G.Z.); (A.D.G.); (V.S.); (M.P.)
| | - Anna De Gaetano
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (G.Z.); (A.D.G.); (V.S.); (M.P.)
- National Institute for Cardiovascular Research-INRC, 40126 Bologna, Italy; (A.C.); (A.V.M.)
| | - Valentina Selleri
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (G.Z.); (A.D.G.); (V.S.); (M.P.)
| | - Gustavo Savino
- Department of Public Healthcare, Sports Medicine Service, Azienda USL of Modena, 41121 Modena, Italy;
| | - Andrea Cossarizza
- National Institute for Cardiovascular Research-INRC, 40126 Bologna, Italy; (A.C.); (A.V.M.)
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Marcello Pinti
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (G.Z.); (A.D.G.); (V.S.); (M.P.)
| | - Anna Vittoria Mattioli
- National Institute for Cardiovascular Research-INRC, 40126 Bologna, Italy; (A.C.); (A.V.M.)
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Milena Nasi
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
- Correspondence: ; Tel.: +39-059-205-5422
| |
Collapse
|
31
|
Wincup C, Sawford N, Rahman A. Pathological mechanisms of abnormal iron metabolism and mitochondrial dysfunction in systemic lupus erythematosus. Expert Rev Clin Immunol 2021; 17:957-967. [PMID: 34263712 PMCID: PMC8452144 DOI: 10.1080/1744666x.2021.1953981] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 07/07/2021] [Indexed: 12/19/2022]
Abstract
Introduction: Systemic lupus erythematosus [SLE] is a chronic, autoimmune condition characterized by the formation of autoantibodies directed against nuclear components and by oxidative stress. Recently, a number of studies have demonstrated the essential role of iron in the immune response and there is growing evidence that abnormal iron homeostasis can occur in the chronic inflammatory state seen in SLE. Not only is iron vital for hematopoiesis, it is also important for a number of other key physiological processes, in particular in maintaining healthy mitochondrial function.Areas covered: In this review, we highlight the latest understanding with regards to how patients with SLE may be at risk of cellular iron depletion as a result of both absolute and functional iron deficiency. Furthermore, we aim to explain the latest evidence of mitochondrial dysfunction in the pathogenesis of the disease.Expert opinion: Growing evidence suggests that both abnormal iron homeostasis and subsequent mitochondrial dysfunction can impair effector immune cell function. Through a greater understanding of these abnormalities, therapeutic options that directly target iron and mitochondria may ultimately represent novel treatment targets that may translate into clinical care of patients with SLE in the near future.
Collapse
Affiliation(s)
- Chris Wincup
- Department of Rheumatology, Division of Medicine, University College London, London, UK
| | - Natalie Sawford
- Department of Rheumatology, Division of Medicine, University College London, London, UK
| | - Anisur Rahman
- Department of Rheumatology, Division of Medicine, University College London, London, UK
| |
Collapse
|
32
|
Abnormal Mitochondrial Physiology in the Pathogenesis of Systemic Lupus Erythematosus. Rheum Dis Clin North Am 2021; 47:427-439. [PMID: 34215372 DOI: 10.1016/j.rdc.2021.05.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disorder characterized by abnormalities within the innate and adaptive immune systems. Activation and proliferation of a wide array of immune cells require significant up-regulation in cellular energy metabolism, with the mitochondria playing an essential role in the initiation and maintenance of this response. This article highlights how abnormal mitochondrial function may occur in SLE and focuses on how energy metabolism, oxidative stress, and impaired mitochondrial repair play a role in the pathogenesis of the disease. How this may represent an appealing novel therapeutic target for future drug therapy in SLE also is discussed.
Collapse
|
33
|
Melki I, Allaeys I, Tessandier N, Lévesque T, Cloutier N, Laroche A, Vernoux N, Becker Y, Benk-Fortin H, Zufferey A, Rollet-Labelle E, Pouliot M, Poirier G, Patey N, Belleannee C, Soulet D, McKenzie SE, Brisson A, Tremblay ME, Lood C, Fortin PR, Boilard E. Platelets release mitochondrial antigens in systemic lupus erythematosus. Sci Transl Med 2021; 13:13/581/eaav5928. [PMID: 33597264 DOI: 10.1126/scitranslmed.aav5928] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 03/20/2020] [Accepted: 10/02/2020] [Indexed: 12/13/2022]
Abstract
The accumulation of DNA and nuclear components in blood and their recognition by autoantibodies play a central role in the pathophysiology of systemic lupus erythematosus (SLE). Despite the efforts, the sources of circulating autoantigens in SLE are still unclear. Here, we show that in SLE, platelets release mitochondrial DNA, the majority of which is associated with the extracellular mitochondrial organelle. Mitochondrial release in patients with SLE correlates with platelet degranulation. This process requires the stimulation of platelet FcγRIIA, a receptor for immune complexes. Because mice lack FcγRIIA and murine platelets are completely devoid of receptor capable of binding IgG-containing immune complexes, we used transgenic mice expressing FcγRIIA for our in vivo investigations. FcγRIIA expression in lupus-prone mice led to the recruitment of platelets in kidneys and to the release of mitochondria in vivo. Using a reporter mouse with red fluorescent protein targeted to the mitochondrion, we confirmed platelets as a source of extracellular mitochondria driven by FcγRIIA and its cosignaling by the fibrinogen receptor α2bβ3 in vivo. These findings suggest that platelets might be a key source of mitochondrial antigens in SLE and might be a therapeutic target for treating SLE.
Collapse
Affiliation(s)
- Imene Melki
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC G1V 4G2, Canada.,Faculté de Médecine and Centre de Recherche ARThrite, Université Laval, Québec, QC G1V 4G2, Canada
| | - Isabelle Allaeys
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC G1V 4G2, Canada.,Faculté de Médecine and Centre de Recherche ARThrite, Université Laval, Québec, QC G1V 4G2, Canada
| | - Nicolas Tessandier
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC G1V 4G2, Canada.,Faculté de Médecine and Centre de Recherche ARThrite, Université Laval, Québec, QC G1V 4G2, Canada
| | - Tania Lévesque
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC G1V 4G2, Canada.,Faculté de Médecine and Centre de Recherche ARThrite, Université Laval, Québec, QC G1V 4G2, Canada
| | - Nathalie Cloutier
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC G1V 4G2, Canada
| | - Audrée Laroche
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC G1V 4G2, Canada.,Faculté de Médecine and Centre de Recherche ARThrite, Université Laval, Québec, QC G1V 4G2, Canada
| | - Nathalie Vernoux
- Axe Neurosciences du Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval et Département de Médecine Moléculaire de l'Université Laval, Québec, QC G1V 4G2, Canada
| | - Yann Becker
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC G1V 4G2, Canada.,Faculté de Médecine and Centre de Recherche ARThrite, Université Laval, Québec, QC G1V 4G2, Canada
| | - Hadrien Benk-Fortin
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC G1V 4G2, Canada.,Faculté de Médecine and Centre de Recherche ARThrite, Université Laval, Québec, QC G1V 4G2, Canada
| | - Anne Zufferey
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC G1V 4G2, Canada.,Faculté de Médecine and Centre de Recherche ARThrite, Université Laval, Québec, QC G1V 4G2, Canada
| | - Emmanuelle Rollet-Labelle
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC G1V 4G2, Canada.,Faculté de Médecine and Centre de Recherche ARThrite, Université Laval, Québec, QC G1V 4G2, Canada
| | - Marc Pouliot
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC G1V 4G2, Canada.,Faculté de Médecine and Centre de Recherche ARThrite, Université Laval, Québec, QC G1V 4G2, Canada
| | - Guy Poirier
- Department of Molecular Biology, Medical Biochemistry, and Pathology, Faculty of Medicine, Université Laval, Quebec, QC G1V 4G2, Canada
| | - Natacha Patey
- Centre Hospitalier Universitaire de Sainte-Justine, Faculté de Médecine, Département de pathologie et biologie cellulaire, Université de Montréal, Montréal, QC H3T 1C5, Canada
| | - Clemence Belleannee
- Department of Obstetrics, Gynecology and Reproduction, Centre hospitalier universitaire de Québec-Université Laval et Département de médecine moléculaire de l'Université Laval, Québec, QC G1V 4G2, Canada
| | - Denis Soulet
- Department of Molecular Biology, Medical Biochemistry, and Pathology, Faculty of Medicine, Université Laval, Quebec, QC G1V 4G2, Canada
| | - Steven E McKenzie
- Cardeza Foundation for Hematologic Research, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Alain Brisson
- UMR-CBMN CNRS-Université de Bordeaux-IPB, Pessac 33600, France
| | - Marie-Eve Tremblay
- Axe Neurosciences du Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval et Département de Médecine Moléculaire de l'Université Laval, Québec, QC G1V 4G2, Canada.,Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | - Christian Lood
- Division of Rheumatology, Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Paul R Fortin
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC G1V 4G2, Canada. .,Faculté de Médecine and Centre de Recherche ARThrite, Université Laval, Québec, QC G1V 4G2, Canada.,Division of Rheumatology, Department of Medicine, Centre hospitalier universitaire de Québec-Université Laval, Québec, QC G1V 4G2, Canada
| | - Eric Boilard
- Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, QC G1V 4G2, Canada. .,Faculté de Médecine and Centre de Recherche ARThrite, Université Laval, Québec, QC G1V 4G2, Canada
| |
Collapse
|
34
|
Abstract
Extracellular vesicles (EVs) are a means of cell-to-cell communication and can facilitate the exchange of a broad array of molecules between adjacent or distant cells. Platelets are anucleate cells derived from megakaryocytes and are primarily known for their role in maintaining hemostasis and vascular integrity. Upon activation by a variety of agonists, platelets readily generate EVs, which were initially identified as procoagulant particles. However, as both platelets and their EVs are abundant in blood, the role of platelet EVs in hemostasis may be redundant. Moreover, findings have challenged the significance of platelet-derived EVs in coagulation. Looking beyond hemostasis, platelet EV cargo is incredibly diverse and can include lipids, proteins, nucleic acids, and organelles involved in numerous other biological processes. Furthermore, while platelets cannot cross tissue barriers, their EVs can enter lymph, bone marrow, and synovial fluid. This allows for the transfer of platelet-derived content to cellular recipients and organs inaccessible to platelets. This review highlights the importance of platelet-derived EVs in physiological and pathological conditions beyond hemostasis.
Collapse
Affiliation(s)
- Florian Puhm
- Centre de recherche du CHU de Québec, Department of infectious diseases and immunity, Québec, QC, Canada
- Université Laval and Centre de recherche ARThrite, Québec, QC, Canada
| | - Eric Boilard
- Centre de recherche du CHU de Québec, Department of infectious diseases and immunity, Québec, QC, Canada
- Université Laval and Centre de recherche ARThrite, Québec, QC, Canada
| | - Kellie R Machlus
- Division of Hematology, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
35
|
Romo-Tena J, Kaplan MJ. Immunometabolism in the pathogenesis of systemic lupus erythematosus: an update. Curr Opin Rheumatol 2020; 32:562-571. [PMID: 32826478 PMCID: PMC10463177 DOI: 10.1097/bor.0000000000000738] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW To provide an update on state-of-the-art evidence on the role of immunometabolism reprogramming in the pathogenesis of systemic lupus erythematosus (SLE). RECENT FINDINGS Mitochondrial dysfunction and enhanced oxidative stress, along with specific defects in other metabolic pathways, can promote dysregulation of innate and adaptive immune responses in SLE. These abnormalities appear to be driven by genetic and epigenetic factors, modulated by stochastic events. In addition to extensive descriptions of abnormalities in immunometabolism of lupus lymphocytes, recent studies support the critical role of dysregulation of metabolic pathways in innate immune cells including neutrophils, macrophages and dendritic cells, in SLE pathogenesis. Recent abnormalities described in lipid metabolism have been associated with SLE disease activity and related damage. Promising therapeutic strategies that target these metabolic abnormalities have recently been described in SLE. SUMMARY Fundamental new insights regarding the role of mitochondrial dysfunction in innate immune dysregulation in SLE pathogenesis have recently emerged. Defects in specific molecular pathways pertinent to immunometabolism in SLE have been described. New insights in translational medicine and promising therapeutic targets have been proposed based on these recent findings.
Collapse
Affiliation(s)
- Jorge Romo-Tena
- Systemic Autoimmunity Branch, Intramural Research Program, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Medical Science PhD Program, School of Medicine, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Mariana J. Kaplan
- Systemic Autoimmunity Branch, Intramural Research Program, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
36
|
Becker YL, Julien AS, Godbout A, Boilard É, Fortin PR. Pilot study of anti-mitochondrial antibodies in antiphospholipid syndrome. Lupus 2020; 29:1623-1629. [PMID: 32787553 DOI: 10.1177/0961203320944481] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Mitochondria are intracellular organelles of bacterial origin capable of stimulating the immune system when released into the extracellular milieu. We previously reported the expression of anti-mitochondrial antibodies (AMA) targeting whole organelles (AwMA), mitochondrial DNA (AmtDNA) or mitochondrial RNA (AmtRNA) in patients with systemic lupus erythematosus (SLE). Antiphospholipid syndrome (APS) is an autoimmune condition that may be independent of, or associated with, other diseases, usually SLE. This study aimed to detect AMA in patients with APS and to explore the association with clinical features of APS. METHODS AwMA-, AmtDNA- and AmtRNA-IgG and -IgM were detected in a pilot study (healthy controls n = 30 and APS patients n = 24) by direct ELISA, and their levels were associated with demographic and disease characteristics. RESULTS AmtDNA-IgM and AmtRNA-IgG and IgM were elevated in APS compared to healthy controls (p = 0.009, p = 0.0005 and p = 0.01, respectively). AwMA-IgG were increased in patients positive for lupus anticoagulant (median ± interquartile range = 0.36 ± 0.31 vs. 0.14 ± 0.08, p = 0.008), and optical density values for AwMA-IgM were correlated with titres of IgM against cardiolipin (rs = 0.51, p = 0.01). An increment of 0.1 unit of AmtDNA-IgM levels was associated with reduced prior reporting of arterial events (odds ratio = 0.86; 95% confidence interval 0.74-1.00; p = 0.047). CONCLUSION Our pilot study suggests that AMA are represented within the autoantibody repertoire in APS and may display different associations with the clinical manifestations of the disease. Further studies should focus on reproducing these preliminary results by following AMA levels through time in larger prospective cohorts.
Collapse
Affiliation(s)
- Yann Lc Becker
- Centre de Recherche ARThrite - Arthrite, Recherche et Traitements, Université Laval, Quebec, Canada
| | - Anne-Sophie Julien
- Département de mathématiques et statistique, Université Laval, Quebec, Canada
| | - Alexandra Godbout
- Centre de Recherche ARThrite - Arthrite, Recherche et Traitements, Université Laval, Quebec, Canada.,Axe maladies infectieuses et inflammatoires, Centre de recherche du CHU de Québec - Université Laval, Quebec, Canada
| | - Éric Boilard
- Centre de Recherche ARThrite - Arthrite, Recherche et Traitements, Université Laval, Quebec, Canada.,Axe maladies infectieuses et inflammatoires, Centre de recherche du CHU de Québec - Université Laval, Quebec, Canada
| | - Paul R Fortin
- Axe maladies infectieuses et inflammatoires, Centre de recherche du CHU de Québec - Université Laval, Quebec, Canada.,Division de Rhumatologie, Département de Médecine, CHU de Québec - Université Laval, Quebec, Canada
| |
Collapse
|
37
|
Iwasaki Y, Takeshima Y, Fujio K. Basic mechanism of immune system activation by mitochondria. Immunol Med 2020; 43:142-147. [PMID: 32393116 DOI: 10.1080/25785826.2020.1756609] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Almost 160 years after the discovery of mitochondria, they are known for their production of energy and are called "the powerhouse of the cell". Recently, immune-metabolism has been revealed as a key factor controlling immune cell proliferation and differentiation. Resting lymphocytes generate energy through oxidative phosphorylation and fatty acid oxidation, whereas activated lymphocytes rapidly shift to glycolysis. Oxidative phosphorylation (OXPHOS) as well as mitochondrial reactive oxygen species (mtROS) generated through the electron transport chain (ETC) are involved in many immune cell functions. Moreover, mitochondria are dynamic organelles that can provide immunogenic molecules, such as mitochondrial DNA (mtDNA) resulting in innate immune system activation. Here, we describe the role of mitochondria in immune system regulation, highlighting metabolism-dependent and other immunogenic aspects.
Collapse
Affiliation(s)
- Yukiko Iwasaki
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yusuke Takeshima
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
38
|
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: 489] [Impact Index Per Article: 97.8] [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
|
39
|
Pisetsky DS, Spencer DM, Mobarrez F, Fuzzi E, Gunnarsson I, Svenungsson E. The binding of SLE autoantibodies to mitochondria. Clin Immunol 2020; 212:108349. [PMID: 31982644 PMCID: PMC10538439 DOI: 10.1016/j.clim.2020.108349] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 01/22/2020] [Accepted: 01/23/2020] [Indexed: 02/08/2023]
Abstract
Systemic lupus erythematosus (SLE) is a prototypic autoimmune disease characterized by immune complexes. Because these complexes contain mitochondrial components, we assessed the presence of antibodies to whole mitochondria (wMITO) using an ELISA in which mitochondria from mouse liver are bound to microtiter plates pre-coated with poly-l-lysine. Studies with this ELISA demonstrated that SLE plasmas contain abundant anti-wMITO activity. While digestion with DNase 1 did not affect anti-wMITO activity, adsorption of plasma on DNA affinity columns could reduce binding activity. Assay for anti-mitochondrial antibodies (AMA) by immunofluorescence and an ELISA with the M2 antigen (2-oxo-acid dehydrogenase protein complex) showed a low frequency of positivity, indicating that AMA and anti-wMITO are distinct specificities. In the study of 204 patients with SLE, the levels of anti-wMITO were higher in active SLE and correlated with levels of anti-DNA. These findings suggest that anti-wMITO can form immune complexes with mitochondria which may drive pathogenesis.
Collapse
Affiliation(s)
- David S Pisetsky
- Division of Rheumatology and Immunology, Duke University Medical Center, Durham, NC, United States of America; Medical Research Service, VA Medical Center, Durham, NC, United States of America.
| | - Diane M Spencer
- Division of Rheumatology and Immunology, Duke University Medical Center, Durham, NC, United States of America
| | - Fariborz Mobarrez
- Unit of Rheumatology, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden; Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Enrico Fuzzi
- Unit of Rheumatology, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden; Division of Rheumatology, Department of Medicine, University of Padua, Padua, Italy
| | - Iva Gunnarsson
- Unit of Rheumatology, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Elisabet Svenungsson
- Unit of Rheumatology, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
40
|
Dard L, Blanchard W, Hubert C, Lacombe D, Rossignol R. Mitochondrial functions and rare diseases. Mol Aspects Med 2020; 71:100842. [PMID: 32029308 DOI: 10.1016/j.mam.2019.100842] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/26/2019] [Accepted: 12/27/2019] [Indexed: 12/19/2022]
Abstract
Mitochondria are dynamic cellular organelles responsible for a large variety of biochemical processes as energy transduction, REDOX signaling, the biosynthesis of hormones and vitamins, inflammation or cell death execution. Cell biology studies established that 1158 human genes encode proteins localized to mitochondria, as registered in MITOCARTA. Clinical studies showed that a large number of these mitochondrial proteins can be altered in expression and function through genetic, epigenetic or biochemical mechanisms including the interaction with environmental toxics or iatrogenic medicine. As a result, pathogenic mitochondrial genetic and functional defects participate to the onset and the progression of a growing number of rare diseases. In this review we provide an exhaustive survey of the biochemical, genetic and clinical studies that demonstrated the implication of mitochondrial dysfunction in human rare diseases. We discuss the striking diversity of the symptoms caused by mitochondrial dysfunction and the strategies proposed for mitochondrial therapy, including a survey of ongoing clinical trials.
Collapse
Affiliation(s)
- L Dard
- Bordeaux University, 33000, Bordeaux, France; INSERM U1211, 33000, Bordeaux, France; CELLOMET, CGFB-146 Rue Léo Saignat, Bordeaux, France
| | - W Blanchard
- Bordeaux University, 33000, Bordeaux, France; INSERM U1211, 33000, Bordeaux, France; CELLOMET, CGFB-146 Rue Léo Saignat, Bordeaux, France
| | - C Hubert
- Bordeaux University, 33000, Bordeaux, France; INSERM U1211, 33000, Bordeaux, France
| | - D Lacombe
- Bordeaux University, 33000, Bordeaux, France; INSERM U1211, 33000, Bordeaux, France; CHU de Bordeaux, Service de Génétique Médicale, F-33076, Bordeaux, France
| | - R Rossignol
- Bordeaux University, 33000, Bordeaux, France; INSERM U1211, 33000, Bordeaux, France; CELLOMET, CGFB-146 Rue Léo Saignat, Bordeaux, France.
| |
Collapse
|
41
|
Wirestam L, Arve S, Linge P, Bengtsson AA. Neutrophils-Important Communicators in Systemic Lupus Erythematosus and Antiphospholipid Syndrome. Front Immunol 2019; 10:2734. [PMID: 31824510 PMCID: PMC6882868 DOI: 10.3389/fimmu.2019.02734] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 11/07/2019] [Indexed: 12/15/2022] Open
Abstract
Systemic lupus erythematosus (SLE) and antiphospholipid syndrome (APS) are two autoimmune diseases that can occur together or separately. Insights into the pathogenesis have revealed similarities, such as development of autoantibodies targeting subcellular antigens as well as a shared increased risk of cardiovascular morbidity, potentially due to mutual pathologic mechanisms. In this review, we will address the evidence implicating neutrophils in the pathogenesis of these conditions, highlighting their shared features. The neutrophil is the most abundant leukocyte, recognized for its role in infectious and inflammatory diseases, but dysregulation of neutrophil effector functions, including phagocytosis, oxidative burst and formation of neutrophil extracellular traps (NETs) may also contribute to an autoimmune process. The phenotype of neutrophils in SLE and APS differs from neutrophils of healthy individuals, where neutrophils in SLE and APS are activated and prone to aggregate. A specific subset of low-density neutrophils with different function compared to normal-density neutrophils can also be found within the peripheral blood mononuclear cell (PBMC) fraction after density gradient centrifugation of whole blood. Neutrophil phagocytosis is required for regular clearance of cell remnants and nuclear material. Reactive oxygen species (ROS) released by neutrophils during oxidative burst are important for immune suppression and impairment of ROS production is seen in SLE. NETs mediate pathology in both SLE and APS via several mechanisms, including exposure of autoantigens, priming of T-cells and activation of autoreactive B-cells. NETs are also involved in cardiovascular events by forming a pro-thrombotic scaffolding surface. Lastly, neutrophils communicate with other cells by producing cytokines, such as Interferon (IFN) -α, and via direct cell-cell contact. Physiological neutrophil effector functions are necessary to prevent autoimmunity, but in SLE and APS these are altered.
Collapse
Affiliation(s)
- Lina Wirestam
- Section of Rheumatology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Sabine Arve
- Section of Rheumatology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Petrus Linge
- Section of Rheumatology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Anders A Bengtsson
- Section of Rheumatology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| |
Collapse
|
42
|
Zhong F, Liang S, Zhong Z. Emerging Role of Mitochondrial DNA as a Major Driver of Inflammation and Disease Progression. Trends Immunol 2019; 40:1120-1133. [PMID: 31744765 DOI: 10.1016/j.it.2019.10.008] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 10/15/2019] [Accepted: 10/17/2019] [Indexed: 12/13/2022]
Abstract
Inflammation benefits the host by promoting the elimination of invading pathogens and clearance of cellular debris after tissue injury. Inflammation also stimulates tissue repair and regeneration to restore homeostasis and organismal health. Emerging evidence suggests that mitochondrial DNA (mtDNA), the only form of non-nuclear DNA in eukaryotic cells, is a major activator of inflammation when leaked out from stressed mitochondria. Here, we review the current understanding on the role of mtDNA in innate immunity, discussing how dysregulated mtDNA metabolism can promote chronic inflammation and disease progression.
Collapse
Affiliation(s)
- Fei Zhong
- Laboratory of Molecular Virology and Immunology, College of Animal Science and Technology and College of Veterinary Medicine, Agricultural University of Hebei, Baoding, Hebei 071000, China
| | - Shuang Liang
- Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Zhenyu Zhong
- Department of Immunology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd., Dallas, TX 75390, USA; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA.
| |
Collapse
|
43
|
Qiu CC, Caricchio R, Gallucci S. Triggers of Autoimmunity: The Role of Bacterial Infections in the Extracellular Exposure of Lupus Nuclear Autoantigens. Front Immunol 2019; 10:2608. [PMID: 31781110 PMCID: PMC6857005 DOI: 10.3389/fimmu.2019.02608] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 10/21/2019] [Indexed: 12/12/2022] Open
Abstract
Infections are considered important environmental triggers of autoimmunity and can contribute to autoimmune disease onset and severity. Nucleic acids and the complexes that they form with proteins—including chromatin and ribonucleoproteins—are the main autoantigens in the autoimmune disease systemic lupus erythematosus (SLE). How these nuclear molecules become available to the immune system for recognition, presentation, and targeting is an area of research where complexities remain to be disentangled. In this review, we discuss how bacterial infections participate in the exposure of nuclear autoantigens to the immune system in SLE. Infections can instigate pro-inflammatory cell death programs including pyroptosis and NETosis, induce extracellular release of host nuclear autoantigens, and promote their recognition in an immunogenic context by activating the innate and adaptive immune systems. Moreover, bacterial infections can release bacterial DNA associated with other bacterial molecules, complexes that can elicit autoimmunity by acting as innate stimuli of pattern recognition receptors and activating autoreactive B cells through molecular mimicry. Recent studies have highlighted SLE disease activity-associated alterations of the gut commensals and the expansion of pathobionts that can contribute to chronic exposure to extracellular nuclear autoantigens. A novel field in the study of autoimmunity is the contribution of bacterial biofilms to the pathogenesis of autoimmunity. Biofilms are multicellular communities of bacteria that promote colonization during chronic infections. We review the very recent literature highlighting a role for bacterial biofilms, and their major components, amyloid/DNA complexes, in the generation of anti-nuclear autoantibodies and their ability to stimulate the autoreactive immune response. The best studied bacterial amyloid is curli, produced by enteric bacteria that commonly cause infections in SLE patients, including Escherichia coli and Salmonella spps. Evidence suggests that curli/DNA complexes can trigger autoimmunity by acting as danger signals, molecular mimickers, and microbial chaperones of nucleic acids.
Collapse
Affiliation(s)
- Connie C Qiu
- Laboratory of Dendritic Cell Biology, Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Roberto Caricchio
- Division of Rheumatology, Department of Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Stefania Gallucci
- Laboratory of Dendritic Cell Biology, Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
44
|
Becker Y, Marcoux G, Allaeys I, Julien AS, Loignon RC, Benk-Fortin H, Rollet-Labelle E, Rauch J, Fortin PR, Boilard E. Autoantibodies in Systemic Lupus Erythematosus Target Mitochondrial RNA. Front Immunol 2019; 10:1026. [PMID: 31134086 PMCID: PMC6524553 DOI: 10.3389/fimmu.2019.01026] [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: 01/15/2019] [Accepted: 04/23/2019] [Indexed: 12/22/2022] Open
Abstract
The mitochondrion supplies energy to the cell and regulates apoptosis. Unlike other mammalian organelles, mitochondria are formed by binary fission and cannot be directly produced by the cell. They contain numerous copies of a compact circular genome that encodes RNA molecules and proteins involved in mitochondrial oxidative phosphorylation. Whereas, mitochondrial DNA (mtDNA) activates the innate immune system if present in the cytosol or the extracellular milieu, it is also the target of circulating autoantibodies in systemic lupus erythematosus (SLE). However, it is not known whether mitochondrial RNA is also recognized by autoantibodies in SLE. In the present study, we evaluated the presence of autoantibodies targeting mitochondrial RNA (AmtRNA) in SLE. We quantified AmtRNA in an inducible model of murine SLE. The AmtRNA were also determined in SLE patients and healthy volunteers. AmtRNA titers were measured in both our induced model of murine SLE and in human SLE, and biostatistical analyses were performed to determine whether the presence and/or levels of AmtRNA were associated with clinical features expressed by SLE patients. Both IgG and IgM classes of AmtRNA were increased in SLE patients (n = 86) compared to healthy controls (n = 30) (p < 0.0001 and p = 0.0493, respectively). AmtRNA IgG levels correlated with anti-mtDNA-IgG titers (rs = 0.54, p < 0.0001) as well as with both IgG and IgM against β-2-glycoprotein I (anti-β2GPI; rs = 0.22, p = 0.05), and AmtRNA-IgG antibodies were present at higher levels when patients were positive for autoantibodies to double-stranded-genomic DNA (p < 0.0001). AmtRNA-IgG were able to specifically discriminate SLE patients from healthy controls, and were negatively associated with plaque formation (p = 0.04) and lupus nephritis (p = 0.03). Conversely, AmtRNA-IgM titers correlated with those of anti-β2GPI-IgM (rs = 0.48, p < 0.0001). AmtRNA-IgM were higher when patients were positive for anticardiolipin antibodies (aCL-IgG: p = 0.01; aCL-IgM: p = 0.002), but AmtRNA-IgM were not associated with any of the clinical manifestations assessed. These findings identify mtRNA as a novel mitochondrial antigen target in SLE, and support the concept that mitochondria may provide an important source of circulating autoantigens in SLE.
Collapse
Affiliation(s)
- Yann Becker
- Département de microbiologie et immunologie, Faculté de Médecine de l'Université Laval, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
| | - Geneviève Marcoux
- Département de microbiologie et immunologie, Faculté de Médecine de l'Université Laval, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
| | - Isabelle Allaeys
- Département de microbiologie et immunologie, Faculté de Médecine de l'Université Laval, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
| | - Anne-Sophie Julien
- Département de mathématiques et statistiques, Université Laval, Québec City, QC, Canada
| | - Renée-Claude Loignon
- Division de Rhumatologie, Département de Médecine, CHU de Québec-Université Laval, Québec City, QC, Canada
| | - Hadrien Benk-Fortin
- Département de microbiologie et immunologie, Faculté de Médecine de l'Université Laval, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
| | - Emmanuelle Rollet-Labelle
- Département de microbiologie et immunologie, Faculté de Médecine de l'Université Laval, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
| | - Joyce Rauch
- Division of Rheumatology, Department of Medicine, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Paul R Fortin
- Département de microbiologie et immunologie, Faculté de Médecine de l'Université Laval, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada.,Division de Rhumatologie, Département de Médecine, CHU de Québec-Université Laval, Québec City, QC, Canada.,Axe maladies infectieuses et inflammatoires, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
| | - Eric Boilard
- Département de microbiologie et immunologie, Faculté de Médecine de l'Université Laval, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada.,Axe maladies infectieuses et inflammatoires, Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
| |
Collapse
|