1
|
Wu Y, Deshpande A, Geraci N, Budde P, Sellers V, Velisetty P, Sun C, Strand F, Bhavsar C, Niewold TB, Jensen MA, Kalatskaya I, Sarin KY, Fiorentino D, Bender AT. TLR7/8 Activation in Immune Cells and Muscle by RNA-Containing Immune Complexes: Role in Inflammation and the Pathogenesis of Myositis. Arthritis Rheumatol 2025; 77:190-201. [PMID: 39279150 PMCID: PMC11782111 DOI: 10.1002/art.42989] [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: 04/11/2024] [Revised: 07/31/2024] [Accepted: 09/10/2024] [Indexed: 09/18/2024]
Abstract
OBJECTIVE Activation of endosomal toll-like receptors (TLRs) is one possible driver of inflammation in idiopathic inflammatory myopathies (IIM). We investigated the potential contribution of TLR7 and TLR8 to IIM pathogenesis. METHODS Activation of TLR7/8 in healthy donor peripheral blood mononuclear cells (PBMCs) by immune complexes from patients with IIMs and lupus was tested. Autoantibody profiling of patient IgG samples was performed using a 1581 antigen array. TLR7 and/or TLR8 activation by RNA molecules associated with autoantibodies was assessed. Gene expression in human myoblasts and satellite cells following treatment with supernatants from TLR7/8-activated PBMCs was evaluated by NanoString. C57BL/6 mice were dosed intramuscularly with the TLR7/8 agonist R848 and single-cell RNA-sequencing was performed on the muscle to ascertain the cell types responding to TLR7/8 activation and the downstream effects. RESULTS Overall, 69 patients with IIMs were included with representation of dermatomyositis, polymyositis, and inclusion body myositis subsets. Immune complexes from patients with IIMs, as well as autoantibody-associated RNAs histidyl-transfer RNA, Y1, Y4, and U1, activated PBMCs to produce interferon-α and IL-6 via TLR7/8. Several canonical (Ro60, Ro52, and HIST1H4A) and novel (IL-36RN) autoreactivities correlated highly with TLR7/8 activation. Supernatants from TLR7/8-activated PBMCs had a negative impact on human myoblasts and satellite cells. Endothelial cells were activated by R848 in mouse muscle in vivo in addition to immune cells such as monocytes and macrophages. CONCLUSION Our results suggest that patients with IIMs have autoantibodies in their blood causing TLR7/8 activation, which leads to inflammation in muscles with potential deleterious effects.
Collapse
Affiliation(s)
- Yin Wu
- EMD SeronoBillericaMassachusetts
| | - Aditee Deshpande
- EMD SeronoBillericaMassachusetts
- Present address:
Novo NordiskLexington MA
| | | | | | - Vera Sellers
- EMD SeronoBillericaMassachusetts
- Present address:
NovartisCambridge MA
| | | | | | | | | | | | | | | | | | | | - Andrew T. Bender
- EMD SeronoBillericaMassachusetts
- Present address:
AbbVieWorcesterMassachusetts
| |
Collapse
|
2
|
Lessard LER, Robert M, Fenouil T, Mounier R, Landel V, Carlesimo M, Hot A, Chazaud B, Laumonier T, Streichenberger N, Gallay L. Contribution of major histocompatibility complex class II immunostaining in distinguishing idiopathic inflammatory myopathy subgroups: A histopathological cohort study. J Neuropathol Exp Neurol 2024; 83:1060-1075. [PMID: 39283714 DOI: 10.1093/jnen/nlae098] [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] [Indexed: 11/21/2024] Open
Abstract
Idiopathic inflammatory myopathies (IIM) are rare, acquired muscle diseases; their diagnosis of is based on clinical, serological, and histological criteria. MHC-I-positive immunostaining, although non-specific, is used as a marker for IIM diagnosis; however, the significance of major histocompatibility complex (MHC)-II immunostaining in IIM remains debated. We investigated patterns of MHC-II immunostaining in myofibers and capillaries in muscle biopsies from 103 patients with dermatomyositis ([DM], n = 31), inclusion body myositis ([IBM], n = 24), anti-synthetase syndrome ([ASyS], n = 10), immune-mediated necrotizing myopathy ([IMNM], n = 18), or overlap myositis ([OM], n = 20). MHC-II immunostaining of myofibers was abnormal in 63/103 of patients (61%) but the patterns differed according to the IIM subgroup. They were diffuse in IBM (96%), negative in IMNM (83%), perifascicular in ASyS (70%), negative (61%) or perifascicular (32%) in DM, and either clustered (40%), perifascicular (30%), or diffuse heterogeneous (15%) in OM. Capillary MHC-II immunostaining also identified quantitative (capillary dropout, n = 47/88, 53%) and qualitative abnormalities, that is, architectural abnormalities, including dilated and leaky capillaries, (n = 79/98, 81%) in all IIM subgroups. Thus, MHC-II myofiber expression patterns allow distinguishing among IIM subgroups. We suggest the addition of MHC-II immunostaining to routine histological panels for IIM diagnosis.
Collapse
Affiliation(s)
- Lola E R Lessard
- Service d'Electroneuromyographie et de pathologies neuromusculaires, Hôpital Neurologique, GHE, Hospices Civils de Lyon, Lyon, France
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, CNRS UMR 5261, Inserm U1315, Université Claude Bernard Lyon 1, Lyon, France
| | - Marie Robert
- Service de Médecine interne et immunologie clinique, Centre Hospitalier Universitaire Édouard Herriot, Hospices Civils de Lyon, Lyon, France
| | - Tanguy Fenouil
- Institut de Pathologie Multisite des Hospices Civils de Lyon-Site Est, GHE, Hospices Civils de Lyon, Lyon, France
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Lyon, France
| | - Rémi Mounier
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, CNRS UMR 5261, Inserm U1315, Université Claude Bernard Lyon 1, Lyon, France
| | - Véréna Landel
- Direction de la Recherche en Santé, Hospices Civils de Lyon, Lyon, France
| | - Marie Carlesimo
- Institut de Pathologie Multisite des Hospices Civils de Lyon-Site Est, GHE, Hospices Civils de Lyon, Lyon, France
| | - Arnaud Hot
- Service de Médecine interne et immunologie clinique, Centre Hospitalier Universitaire Édouard Herriot, Hospices Civils de Lyon, Lyon, France
| | - Bénédicte Chazaud
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, CNRS UMR 5261, Inserm U1315, Université Claude Bernard Lyon 1, Lyon, France
| | - Thomas Laumonier
- Laboratoire "Cell Therapy & Musculoskeletal Disorders", Département de Chirurgie Orthopédique, Hôpital Universitaire et Faculté de Médecine, Genève, Switzerland
| | - Nathalie Streichenberger
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, CNRS UMR 5261, Inserm U1315, Université Claude Bernard Lyon 1, Lyon, France
- Institut de Pathologie Multisite des Hospices Civils de Lyon-Site Est, GHE, Hospices Civils de Lyon, Lyon, France
| | - Laure Gallay
- Service de Médecine interne et immunologie clinique, Centre Hospitalier Universitaire Édouard Herriot, Hospices Civils de Lyon, Lyon, France
- Laboratoire "Cell Therapy & Musculoskeletal Disorders", Département de Chirurgie Orthopédique, Hôpital Universitaire et Faculté de Médecine, Genève, Switzerland
| |
Collapse
|
3
|
Sherman MA, Farhadi PN, Pak K, Trieu EP, Sarkar K, Targoff IN, Neely ML, Mammen AL, Rider LG. Myositis-Associated Autoantibodies in Patients With Juvenile Myositis Are Associated With Refractory Disease and Mortality. Arthritis Rheumatol 2024; 76:963-972. [PMID: 38272842 PMCID: PMC11136598 DOI: 10.1002/art.42813] [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: 11/16/2023] [Revised: 01/08/2024] [Accepted: 01/22/2024] [Indexed: 01/27/2024]
Abstract
OBJECTIVE Myositis-associated autoantibodies (MAAs) have been associated with overlap myositis, certain disease manifestations such as interstitial lung disease (ILD), and worse prognosis in the idiopathic inflammatory myopathies. MAAs overall remain largely uncharacterized in patients with juvenile-onset myositis. Moreover, it is unknown whether the number of MAAs is associated with disease severity. METHODS Patients with juvenile myositis in cross-sectional natural history studies who underwent testing for myositis autoantibodies were included. Demographics, myositis autoantibodies, clinical characteristics, medications received, and outcomes of those with and without MAAs were compared. Multivariable logistic regression was performed to determine whether the number of MAAs detected was associated with severe disease features. RESULTS Among 551 patients, 36% had an MAA and 13% had more than one MAA. Among those who were MAA positive, there was a higher frequency of overlap myositis (18% vs 5.9%, P < 0.001). MAA positivity was associated with certain clinical features, including Raynaud phenomenon (odds ratio [OR] 2.44, 95% confidence interval [CI] 1.41-4.28) and ILD (OR 3.43, 95% CI 1.75-6.96), as well as a chronic disease course (OR 1.72, 95% CI 1.10-2.72) and mortality (OR 3.76, 95% CI 1.72-8.43). The number of MAAs was also associated with mortality (OR 1.83, 95% CI 1.16-2.86). CONCLUSION MAAs were prevalent in a large cohort of patients with juvenile myositis. ILD, refractory disease, and mortality were associated with MAA positivity. Prospective studies are needed to determine whether early detection of MAAs may lead to improved outcomes for patients with juvenile myositis.
Collapse
Affiliation(s)
- Matthew A. Sherman
- Muscle Disease Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Payam Noroozi Farhadi
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Katherine Pak
- Muscle Disease Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Edward P. Trieu
- Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Kakali Sarkar
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Ira N. Targoff
- Veterans Affairs Medical Center, University of Oklahoma Health Sciences Center, and Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Megan L. Neely
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Andrew L. Mammen
- Muscle Disease Unit, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Lisa G. Rider
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | | |
Collapse
|
4
|
Văcăraş V, Vulturar R, Chiş A, Damian L. Inclusion body myositis, viral infections, and TDP-43: a narrative review. Clin Exp Med 2024; 24:91. [PMID: 38693436 PMCID: PMC11062973 DOI: 10.1007/s10238-024-01353-9] [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: 03/21/2024] [Accepted: 04/15/2024] [Indexed: 05/03/2024]
Abstract
The ubiquitous RNA-processing molecule TDP-43 is involved in neuromuscular diseases such as inclusion body myositis, a late-onset acquired inflammatory myopathy. TDP-43 solubility and function are disrupted in certain viral infections. Certain viruses, high viremia, co-infections, reactivation of latent viruses, and post-acute expansion of cytotoxic T cells may all contribute to inclusion body myositis, mainly in an age-shaped immune landscape. The virally induced senescent, interferon gamma-producing cytotoxic CD8+ T cells with increased inflammatory, and cytotoxic features are involved in the occurrence of inclusion body myositis in most such cases, in a genetically predisposed host. We discuss the putative mechanisms linking inclusion body myositis, TDP-43, and viral infections untangling the links between viruses, interferon, and neuromuscular degeneration could shed a light on the pathogenesis of the inclusion body myositis and other TDP-43-related neuromuscular diseases, with possible therapeutic implications.
Collapse
Affiliation(s)
- Vitalie Văcăraş
- Department of Neurosciences, "Iuliu Haţieganu" University of Medicine and Pharmacy, Cluj-Napoca, 43, Victor Babeş St, 400012, Cluj-Napoca, Romania
- Neurology Department of Cluj, County Emergency Hospital, 3-5, Clinicilor St, 400347, Cluj-Napoca, Romania
| | - Romana Vulturar
- Department of Molecular Sciences, "Iuliu Haţieganu" University of Medicine and Pharmacy Cluj-Napoca, 6, Pasteur St, 400349, Cluj-Napoca, Romania
- Cognitive Neuroscience Laboratory, University Babeş-Bolyai, 30, Fântânele St, 400294, Cluj-Napoca, Romania
- Association for Innovation in Rare Inflammatory, Metabolic, Genetic Diseases INNOROG, 30E, Făgetului St, 400497, Cluj-Napoca, Romania
| | - Adina Chiş
- Department of Molecular Sciences, "Iuliu Haţieganu" University of Medicine and Pharmacy Cluj-Napoca, 6, Pasteur St, 400349, Cluj-Napoca, Romania.
- Cognitive Neuroscience Laboratory, University Babeş-Bolyai, 30, Fântânele St, 400294, Cluj-Napoca, Romania.
- Association for Innovation in Rare Inflammatory, Metabolic, Genetic Diseases INNOROG, 30E, Făgetului St, 400497, Cluj-Napoca, Romania.
| | - Laura Damian
- Association for Innovation in Rare Inflammatory, Metabolic, Genetic Diseases INNOROG, 30E, Făgetului St, 400497, Cluj-Napoca, Romania
- Department of Rheumatology, Centre for Rare Autoimmune and Autoinflammatory Diseases, Emergency, Clinical County Hospital Cluj, 2-4, Clinicilor St, 400006, Cluj-Napoca, Romania
- CMI Reumatologie Dr. Damian, 6-8, Petru Maior St, 400002, Cluj-Napoca, Romania
| |
Collapse
|
5
|
Poli MC. Proteasome disorders and inborn errors of immunity. Immunol Rev 2024; 322:283-299. [PMID: 38071420 DOI: 10.1111/imr.13299] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 03/20/2024]
Abstract
Inborn errors of immunity (IEI) or primary immune deficiencies (PIDD) are caused by variants in genes encoding for molecules that are relevant to the innate or adaptive immune response. To date, defects in more than 450 different genes have been identified as causes of IEI, causing a constellation of heterogeneous clinical manifestations ranging from increased susceptibility to infection, to autoimmunity or autoinflammation. IEI that are mainly characterized by autoinflammation are broadly classified according to the inflammatory pathway that they predominantly perturb. Among autoinflammatory IEI are those characterized by the transcriptional upregulation of type I interferon genes and are referred to as interferonopathies. Within the spectrum of interferonopathies, genetic defects that affect the proteasome have been described to cause autoinflammatory disease and represent a growing area of investigation. This review is focused on describing the clinical, genetic, and molecular aspects of IEI associated with mutations that affect the proteasome and how the study of these diseases has contributed to delineate therapeutic interventions.
Collapse
Affiliation(s)
- M Cecilia Poli
- Faculty of Medicine, Clínica Alemana Universidad del Desarrollo, Santiago, Chile
- Unit of Immunology and Rheumatology Hospital Roberto del Río, Santiago, Chile
| |
Collapse
|
6
|
Tanaka Y, Kusuda M, Yamaguchi Y. Interferons and systemic lupus erythematosus: Pathogenesis, clinical features, and treatments in interferon-driven disease. Mod Rheumatol 2023; 33:857-867. [PMID: 36440704 DOI: 10.1093/mr/roac140] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/24/2022] [Accepted: 11/09/2022] [Indexed: 08/27/2023]
Abstract
Type I interferons (IFNs) have recently received a lot of attention with the elucidation of the pathogenesis of systemic lupus erythematosus (SLE). Type I IFNs are associated with many SLE symptoms and play a role in the pathogenesis of autoimmune diseases that may occur concurrently with SLE, such as Sjögren's syndrome, antiphospholipid syndrome, myositis, scleroderma, and interferonopathy. Type I IFNs could be the link between these diseases. However, direct measurement of type I IFN levels and the IFN gene signature is currently unavailable in clinical practice. This review discusses type I IFN signalling in SLE, investigates the role of type I IFN in the clinical manifestations and symptoms associated with SLE and other IFN-related diseases, and discusses the clinical tests that can be used to diagnose SLE and measure disease activity. In addition, the role of type I IFN-blocking therapies as potential treatments for SLE is discussed.
Collapse
Affiliation(s)
- Yoshiya Tanaka
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | | | | |
Collapse
|
7
|
Rodríguez-Carrio J, Burska A, Conaghan PG, Dik WA, Biesen R, Eloranta ML, Cavalli G, Visser M, Boumpas DT, Bertsias G, Wahren-Herlenius M, Rehwinkel J, Frémond ML, Crow MK, Ronnblom L, Vital E, Versnel M. Association between type I interferon pathway activation and clinical outcomes in rheumatic and musculoskeletal diseases: a systematic literature review informing EULAR points to consider. RMD Open 2023; 9:e002864. [PMID: 36882218 PMCID: PMC10008483 DOI: 10.1136/rmdopen-2022-002864] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 02/13/2023] [Indexed: 03/09/2023] Open
Abstract
BACKGROUND Type I interferons (IFN-I) contribute to a broad range of rheumatic and musculoskeletal diseases (RMDs). Compelling evidence suggests that the measurement of IFN-I pathway activation may have clinical value. Although several IFN-I pathway assays have been proposed, the exact clinical applications are unclear. We summarise the evidence on the potential clinical utility of assays measuring IFN-I pathway activation. METHODS A systematic literature review was conducted across three databases to evaluate the use of IFN-I assays in diagnosis and monitor disease activity, prognosis, response to treatment and responsiveness to change in several RMDs. RESULTS Of 366 screened, 276 studies were selected that reported the use of assays reflecting IFN-I pathway activation for disease diagnosis (n=188), assessment of disease activity (n=122), prognosis (n=20), response to treatment (n=23) and assay responsiveness (n=59). Immunoassays, quantitative PCR (qPCR) and microarrays were reported most frequently, while systemic lupus erythematosus (SLE), rheumatoid arthritis, myositis, systemic sclerosis and primary Sjögren's syndrome were the most studied RMDs. The literature demonstrated significant heterogeneity in techniques, analytical conditions, risk of bias and application in diseases. Inadequate study designs and technical heterogeneity were the main limitations. IFN-I pathway activation was associated with disease activity and flare occurrence in SLE, but their incremental value was uncertain. IFN-I pathway activation may predict response to IFN-I targeting therapies and may predict response to different treatments. CONCLUSIONS Evidence indicates potential clinical value of assays measuring IFN-I pathway activation in several RMDs, but assay harmonisation and clinical validation are urged. This review informs the EULAR points to consider for the measurement and reporting of IFN-I pathway assays.
Collapse
Affiliation(s)
- Javier Rodríguez-Carrio
- Area of Immunology, University of Oviedo, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Asturias, Spain
| | - Agata Burska
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds & NIHR Leeds Biomedical Research Centre, Leeds, UK
| | - P G Conaghan
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds & NIHR Leeds Biomedical Research Centre, Leeds, UK
| | - Willem A Dik
- Laboratory Medical Immunology, department of Immunology, Erasmus MC University Medical Center Rotterdam, The Netherlands
| | - Robert Biesen
- Department of Rheumatology, Charité University Medicine Berlin, Berlin, Germany
| | - Maija-Leena Eloranta
- Department of Medical Sciences, Rheumatology, Uppsala University, Uppsala, Sweden
| | - Giulio Cavalli
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, Vita-Salute San Raffaele University, Milan, Italy
| | - Marianne Visser
- EULAR, PARE Patient Research Partners, Amsterdam, The Netherlands
| | - Dimitrios T Boumpas
- Department of Internal Medicine, University of Crete, Medical School, Heraklion, Greece
| | - George Bertsias
- Department of Rheumatology-Clinical Immunology, University of Crete, Medical School, Heraklion, Greece
| | - Marie Wahren-Herlenius
- Karolinska Institutet, Division of Rheumatology, Stockholm, Sweden
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, Norway
| | - Jan Rehwinkel
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, UK
| | - Marie-Louise Frémond
- Université de Paris Cité, Hôpital Necker-Enfants Malades, Immuno-Hématologie et Rhumatologie pédiatriques, Paris, France
| | - Mary K Crow
- Hospital for Special Surgery, Weill Cornell Medical College, Mary Kirkland Center for Lupus Research, New York, USA
| | - Lars Ronnblom
- Department of Medical Sciences, Rheumatology, Uppsala University, Uppsala, Sweden
| | - Ed Vital
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds & NIHR Leeds Biomedical Research Centre, Leeds, UK
| | - Marjan Versnel
- Department of Immunology, Erasmus MC University Medical Center Rotterdam, The Netherlands
| |
Collapse
|
8
|
Zhou M, Cheng X, Zhu W, Jiang J, Zhu S, Wu X, Liu M, Fang Q. Activation of
cGAS‐STING
pathway – A possible cause of myofiber atrophy/necrosis in dermatomyositis and immune‐mediated necrotizing myopathy. J Clin Lab Anal 2022; 36:e24631. [PMID: 36030554 PMCID: PMC9550984 DOI: 10.1002/jcla.24631] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/08/2019] [Accepted: 06/22/2022] [Indexed: 11/09/2022] Open
Abstract
Objective The objective was to investigate the expression of the cGAS‐STING pathway‐associated protein in idiopathic inflammatory myopathy (IIM) and to investigate whether it is related to myofiber atrophy/necrosis in patients with dermatomyositis and immune‐mediated necrotizing myopathy. Material and Methods Muscle specimens obtained by open biopsy from 26 IIM patients (14 with dermatomyositis (DM), 8 with immune‐mediated necrotizing myopathy (IMNM), and 4 with other types of IIM), 4 dystrophinopathy, and 9 control patients were assessed for expression of cGAS‐STING pathway members via Western blot, quantitative real‐time PCR analysis (qRT‐PCR), and immunochemistry. Meanwhile, analysis its location distribution througn immunochemistry. Results Compared to the control group, the expression of cGAS, STING, and related molecules was obviously increased in muscle samples of IIM patients. Upregulated cGAS and STING were mainly located in the vascular structure, inflammatory infiltrates, and atrophic and necrotic fibers. While comparing to the Dys patients, the mRNA level of cGAS, STING, and TNF‐a was upregulated, meanwhile, the protein of the TBK1, P‐TBK1, and P‐IRF3 associated with interferon upregulation was overexpressed through Western blot in IMNM and DM. Considering that cGAS and STING are located in necrotic and Mx1‐positive atrophic fibers, it is really possible that the cGAS‐STING pathway may lead to fibers atrophy/necrosis by producing IFNs. Conclusion The cGAS‐STING pathway was activated in the muscle samples of IIM patients and its activation may be the reason of myofiber atrophy and necrosis in DM and IMNM patients.
Collapse
Affiliation(s)
- Meichen Zhou
- Department of Neurology First Affiliated Hospital of Soochow University Suzhou China
| | - Xiaoxiao Cheng
- Department of Neurology First Affiliated Hospital of Soochow University Suzhou China
| | - Wenhua Zhu
- Department of Neurology Huashan hospital Shanghai China
| | - Jianhua Jiang
- Department of Neurology First Affiliated Hospital of Soochow University Suzhou China
| | - Sijia Zhu
- Department of Neurology First Affiliated Hospital of Soochow University Suzhou China
| | - Xuan Wu
- Department of Neurology Affiliated Hospital of Yangzhou University Yangzhou China
| | - Meirong Liu
- Department of Neurology First Affiliated Hospital of Soochow University Suzhou China
| | - Qi Fang
- Department of Neurology First Affiliated Hospital of Soochow University Suzhou China
| |
Collapse
|
9
|
Lago SG, Tomasik J, van Rees GF, Rustogi N, Vázquez-Bourgon J, Papiol S, Suarez-Pinilla P, Crespo-Facorro B, Bahn S. Peripheral lymphocyte signaling pathway deficiencies predict treatment response in first-onset drug-naïve schizophrenia. Brain Behav Immun 2022; 103:37-49. [PMID: 35381347 DOI: 10.1016/j.bbi.2022.03.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 03/12/2022] [Accepted: 03/31/2022] [Indexed: 12/29/2022] Open
Abstract
Despite being a major cause of disability worldwide, the pathophysiology of schizophrenia and molecular basis of treatment response heterogeneity continue to be unresolved. Recent evidence suggests that multiple aspects of pathophysiology, including genetic risk factors, converge on key cell signaling pathways and that exploration of peripheral blood cells might represent a practical window into cell signaling alterations in the disease state. We employed multiplexed phospho-specific flow cytometry to examine cell signaling epitope expression in peripheral blood mononuclear cell (PBMC) subtypes in drug-naïve schizophrenia patients (n = 49) relative to controls (n = 61) and relate these changes to serum immune response proteins, schizophrenia polygenic risk scores and clinical effects of treatment, including drug response and side effects, over the longitudinal course of antipsychotic treatment. This revealed both previously characterized (Akt1) and novel cell signaling epitopes (IRF-7 (pS477/pS479), CrkL (pY207), Stat3 (pS727), Stat3 (pY705) and Stat5 (pY694)) across PBMC subtypes which were associated with schizophrenia at disease onset, and correlated with type I interferon-related serum molecules CD40 and CXCL11. Alterations in Akt1 and IRF-7 (pS477/pS479) were additionally associated with polygenic risk of schizophrenia. Finally, changes in Akt1, IRF-7 (pS477/pS479) and Stat3 (pS727) predicted development of metabolic and cardiovascular side effects following antipsychotic treatment, while IRF-7 (pS477/pS479) and Stat3 (pS727) predicted early improvements in general psychopathology scores measured using the Brief Psychiatric Rating Scale (BPRS). These findings suggest that peripheral blood cells can provide an accessible surrogate model for intracellular signaling alterations in schizophrenia and have the potential to stratify subgroups of patients with different clinical outcomes or a greater risk of developing metabolic and cardiovascular side effects following antipsychotic therapy.
Collapse
Affiliation(s)
- Santiago G Lago
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, United Kingdom
| | - Jakub Tomasik
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, United Kingdom
| | - Geertje F van Rees
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, United Kingdom
| | - Nitin Rustogi
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, United Kingdom
| | - Javier Vázquez-Bourgon
- Department of Psychiatry, Marqués de Valdecilla University Hospital, IDIVAL, School of Medicine, University of Cantabria, Santander, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Santander, Spain
| | - Sergi Papiol
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Barcelona, Spain; Institute of Psychiatric Phenomics and Genomics, University Hospital, Ludwig Maximilian University, Munich, Germany; Department of Psychiatry and Psychotherapy, University Hospital, Ludwig Maximilian University, Munich, Germany
| | - Paula Suarez-Pinilla
- Department of Psychiatry, Marqués de Valdecilla University Hospital, IDIVAL, School of Medicine, University of Cantabria, Santander, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Santander, Spain
| | - Benedicto Crespo-Facorro
- Department of Psychiatry, Marqués de Valdecilla University Hospital, IDIVAL, School of Medicine, University of Cantabria, Santander, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Santander, Spain; Department of Psychiatry, School of Medicine, University Hospital Virgen del Rocio, IBiS, Sevilla, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Sevilla, Spain
| | - Sabine Bahn
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
10
|
Gallay L, Fermon C, Lessard L, Weiss-Gayet M, Viel S, Streichenberger N, Corpet A, Mounier R, Gitiaux C, Mouchiroud G, Chazaud B. Involvement of Type-I Interferon Signaling in Muscle Stem Cell Proliferation During Dermatomyositis. Neurology 2022; 98:e2108-e2119. [DOI: 10.1212/wnl.0000000000200271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 02/08/2022] [Indexed: 11/15/2022] Open
Abstract
Background and objective:The idiopathic inflammatory myopathy Dermatomyositis (DM) is an acquired disease that combines muscle, lung and skin impairments. DM patients show a wide range of severity of proximal skeletal muscle weakness, associated with inflammatory infiltrates, vasculitis, and capillary dropout, perifascicular myofiber atrophy. Moreover, DM muscles show signs of muscle regeneration. Since muscle stem cells (MuSCs) are responsible for myofiber repair, we asked wether the proliferative properties of muscle stem cells (MuSCs) are altered in DM muscle. We investigated the role of type-I interferon (IFN-I) in this process since DM is associated with sustained inflammation with high IFN-I levels.Methods:MuSCs isolated from normal, adult and juvenile DM muscles were grown in culture and were analyzed in vitro for their proliferating properties, their myogenic capacities and their senescence. Gain and loss of function experiments were performed to assess the role of IFN-I signaling in the prolfierative capacities of MuSCs.Results:MuSCs derived from 8 DM adult patients (DM-MuSCs) (5 severe form and 3 mild form, established from histological evaluation), from 3 juvenile DM patients and from normal muscle were used to analyze their myogenesis in vitro. DM-MuSCs exhibited strongly reduced proliferating capacities as compared with healthy MuSCs (-31 to -43% for severe and mild DM, respectively), leading to poor myotube formation (-36 to -71%). DM-MuSCs were enriched in senescent, beta-galactosidase positive cells, explaining partly the proliferation defect. Gain and loss of function experiments were performed to assess the role of IFN-I on the proliferative capacity of MuSCs. High concentrations of IFN-I decreased the proliferation of healthy MuSCs. Similarly, conditioned-medium from DM-MuSCs decreased the proliferation of healthy MuSC (-15 to -22%), suggesting the delivery of an autocrine effector. Then, pharmacological blockade of the IFN signaling (using ruxolitinib or anti-IFN-receptor antibodies) in DM-MuSCs rescued their proliferation up to the control values.Discussion:These results show that autocrine IFN-I signaling prevents MuSC expansion, leading to muscle repair deficit. This process may explain the persistent muscle weakness observed in severe DM patients.
Collapse
|
11
|
Graf M, von Stuckrad SL, Uruha A, Klotsche J, Zorn-Pauly L, Unterwalder N, Buttgereit T, Krusche M, Meisel C, Burmester GR, Hiepe F, Biesen R, Kallinich T, Stenzel W, Schneider U, Rose T. SIGLEC1 enables straightforward assessment of type I interferon activity in idiopathic inflammatory myopathies. RMD Open 2022; 8:rmdopen-2021-001934. [PMID: 35177553 PMCID: PMC8860073 DOI: 10.1136/rmdopen-2021-001934] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 01/23/2022] [Indexed: 11/30/2022] Open
Abstract
Objective To evaluate sialic acid binding Ig-like lectin 1 (SIGLEC1) expression on monocytes by flow cytometry as a type I interferon biomarker in idiopathic inflammatory myopathies (IIM). Methods We performed a retrospective analysis of adult and paediatric patients with the diagnosis of IIM. SIGLEC1 expression was assessed by flow cytometry and was compared with Physician Global Assessment or Childhood Myositis Assessment Scale disease activity scores. Mann Whitney U test and receiver operating characteristic curves were used for cross-sectional data analysis (n=96), two-level mixed-effects linear regression model for longitudinal analyses (n=26, 110 visits). Response to treatment was analysed in 14 patients within 12 months, using Wilcoxon test. SIGLEC1 was compared with interferon-stimulated gene 15/MxA status by immunohistochemical staining of muscle biopsies (n=17). Results 96 patients with adult (a) and juvenile (j) dermatomyositis (DM, n=38), antisynthetase syndrome (AS, n=19), immune-mediated necrotising myopathy (IMNM, n=8), inclusion body myositis (IBM, n=9) and overlap myositis (n=22) were included. SIGLEC1 distinguished significantly between active and inactive disease with an area under the curve of 0.92 (95% CI 0.83 to 1) in DM and correlated with disease activity longitudinally (aDM: standardised beta=0.54, p<0.001; jDM: standardised beta=−0.70, p<0.001). Response to treatment in DM was associated with a decreasing SIGLEC1 (p<0.01, Wilcoxon test). SIGLEC1 was found upregulated in 8 of 19 patients with AS, 2 of 9 patients with IBM but not in IMNM. Conclusion SIGLEC1 is a candidate biomarker to assess type I interferon activity in IIM and proved useful for monitoring disease activity and response to treatment in juvenile and adult DM.
Collapse
Affiliation(s)
- Manuel Graf
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sae Lim von Stuckrad
- Department of Pediatric Pneumology, Immunology and Critical Care Medicine and SPZ (Center for Chronically Sick Children), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Akinori Uruha
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Department of Neurology, Tokyo Metropolitan Neurological Hospital, Fuchu, Tokyo, Japan
| | - Jens Klotsche
- German Rheumatism Research Center Berlin - a Leibniz Institute (DRFZ), Berlin, Germany
| | - Lydia Zorn-Pauly
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nadine Unterwalder
- Department of Immunology, Labor Berlin - Charité Vivantes GmbH, Berlin, Germany
| | - Thomas Buttgereit
- Department of Dermatology and Allergy, Dermatological Allergology, Allergie-Centrum-Charité, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Martin Krusche
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christian Meisel
- Department of Immunology, Labor Berlin - Charité Vivantes GmbH, Berlin, Germany
| | - Gerd R Burmester
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Falk Hiepe
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Robert Biesen
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Tilmann Kallinich
- Department of Pediatric Pneumology, Immunology and Critical Care Medicine and SPZ (Center for Chronically Sick Children), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,German Rheumatism Research Center Berlin - a Leibniz Institute (DRFZ), Berlin, Germany
| | - Werner Stenzel
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Udo Schneider
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Thomas Rose
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
12
|
Cooles FAH, Isaacs JD. The interferon gene signature as a clinically relevant biomarker in autoimmune rheumatic disease. THE LANCET. RHEUMATOLOGY 2022; 4:e61-e72. [PMID: 38288732 DOI: 10.1016/s2665-9913(21)00254-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 07/21/2021] [Accepted: 07/29/2021] [Indexed: 12/19/2022]
Abstract
The interferon gene signature (IGS) is derived from the expression of interferon-regulated genes and is classically increased in response to type I interferon exposure. A raised whole blood IGS has increasingly been reported in rheumatic diseases as sequencing technology has advanced. Although its role remains unclear, we explore how a raised IGS can function as a clinically relevant biomarker, independent of whether it is a bystander effect or a key pathological process. For example, a raised IGS can act as a diagnostic biomarker when predicting rheumatoid arthritis in patients with arthralgia and anti-citrullinated protein antibodies, or predicting systemic lupus erythematous (SLE) in those with antinuclear antibodies; a theragnostic biomarker when predicting response for patients receiving disease modifying therapy, such as rituximab in rheumatoid arthritis; a biomarker of disease activity (early rheumatoid arthritis, dermatomyositis, systemic sclerosis, SLE); or finally a predictor of clinical characteristics, such as lupus nephritis in SLE or disease burden in primary Sjögren's syndrome. A high IGS does not uniformly predict worse clinical phenotypes across all diseases, as demonstrated by a reduced disease burden in primary Sjögren's syndrome, nor does it predict a universally poorer response to all therapies, as shown in rheumatoid arthritis. This dichotomy highlights both the complexity of type I interferon signalling in vivo and the current lack of standardisation when calculating the IGS. The IGS as a biomarker warrants further exploration, with beneficial clinical applications anticipated in multiple rheumatic diseases.
Collapse
Affiliation(s)
- Faye A H Cooles
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - John D Isaacs
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK; Musculoskeletal Unit, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.
| |
Collapse
|
13
|
Ll Wilkinson MG, Deakin CT, Papadopoulou C, Eleftheriou D, Wedderburn LR. JAK inhibitors: a potential treatment for JDM in the context of the role of interferon-driven pathology. Pediatr Rheumatol Online J 2021; 19:146. [PMID: 34563217 PMCID: PMC8466894 DOI: 10.1186/s12969-021-00637-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/28/2021] [Indexed: 12/29/2022] Open
Abstract
Juvenile Idiopathic Inflammatory Myopathies (IIM) are a group of rare diseases that are heterogeneous in terms of pathology that can include proximal muscle weakness, associated skin changes and systemic involvement. Despite options for treatment, many patients continue to suffer resistant disease and lasting side-effects. Advances in the understanding of the immunopathology and genetics underlying IIM may specify new therapeutic targets, particularly where conventional treatment has not achieved a clinical response. An upregulated type I interferon signature is strongly associated with disease and could be a prime target for developing more specific therapeutics. There are multiple components of the IFN pathway that could be targeted for blockade therapy.Downstream of the cytokine receptor complexes are the Janus kinase-signal transducers and activators of transcription (JAK-STAT) pathway, which consists of JAK1-3, TYK2, and STAT1-6. Therapeutic inhibitors have been developed to target components of this pathway. Promising results have been observed in case studies reporting the use of the JAK inhibitors, Baricitinib, Tofacitinib and Ruxolitinib in the treatment of refractory Juvenile Dermatomyositis (JDM). There is still the question of safety and efficacy for the use of JAK inhibitors in JDM that need to be addressed by clinical trials. Here we review the future for the use of JAK inhibitors as a treatment for JDM.
Collapse
Affiliation(s)
- Meredyth G Ll Wilkinson
- Infection, Immunity and Inflammation Programme Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK.
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, University College London, London, UK.
- NIHR Biomedical Research Centre at GOSH, London, UK.
| | - Claire T Deakin
- Infection, Immunity and Inflammation Programme Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, University College London, London, UK
- NIHR Biomedical Research Centre at GOSH, London, UK
| | - Charalampia Papadopoulou
- Infection, Immunity and Inflammation Programme Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
- Rheumatology, Great Ormond Street Hospital, Great Ormond Street, London, UK
| | - Despina Eleftheriou
- Infection, Immunity and Inflammation Programme Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
- Rheumatology, Great Ormond Street Hospital, Great Ormond Street, London, UK
| | - Lucy R Wedderburn
- Infection, Immunity and Inflammation Programme Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
- Centre for Adolescent Rheumatology Versus Arthritis at UCL UCLH and GOSH, University College London, London, UK
- NIHR Biomedical Research Centre at GOSH, London, UK
- Rheumatology, Great Ormond Street Hospital, Great Ormond Street, London, UK
| |
Collapse
|
14
|
Bolko L, Jiang W, Tawara N, Landon‐Cardinal O, Anquetil C, Benveniste O, Allenbach Y. The role of interferons type I, II and III in myositis: A review. Brain Pathol 2021; 31:e12955. [PMID: 34043262 PMCID: PMC8412069 DOI: 10.1111/bpa.12955] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 03/19/2021] [Indexed: 12/27/2022] Open
Abstract
The classification of idiopathic inflammatory myopathies (IIM) is based on clinical, serological and histological criteria. The identification of myositis-specific antibodies has helped to define more homogeneous groups of myositis into four dominant subsets: dermatomyositis (DM), antisynthetase syndrome (ASyS), sporadic inclusion body myositis (sIBM) and immune-mediated necrotising myopathy (IMNM). sIBM and IMNM patients present predominantly with muscle involvement, whereas DM and ASyS patients present additionally with other extramuscular features, such as skin, lung and joints manifestations. Moreover, the pathophysiological mechanisms are distinct between each myositis subsets. Recently, interferon (IFN) pathways have been identified as key players implicated in the pathophysiology of myositis. In DM, the key role of IFN, especially type I IFN, has been supported by the identification of an IFN signature in muscle, blood and skin of DM patients. In addition, DM-specific antibodies are targeting antigens involved in the IFN signalling pathways. The pathogenicity of type I IFN has been demonstrated by the identification of mutations in the IFN pathways leading to genetic diseases, the monogenic interferonopathies. This constitutive activation of IFN signalling pathways induces systemic manifestations such as interstitial lung disease, myositis and skin rashes. Since DM patients share similar features in the context of an acquired activation of the IFN signalling pathways, we may extend underlying concepts of monogenic diseases to acquired interferonopathy such as DM. Conversely, in ASyS, available data suggest a role of type II IFN in blood, muscle and lung. Indeed, transcriptomic analyses highlighted a type II IFN gene expression in ASyS muscle tissue. In sIBM, type II IFN appears to be an important cytokine involved in muscle inflammation mechanisms and potentially linked to myodegenerative features. For IMNM, currently published data are scarce, suggesting a minor implication of type II IFN. This review highlights the involvement of different IFN subtypes and their specific molecular mechanisms in each myositis subset.
Collapse
Affiliation(s)
- Loïs Bolko
- Division of RheumatologyHopital Maison BlancheReimsFrance
| | - Wei Jiang
- Department of Internal Medicine and Clinical ImmunlogySorbonne UniversitéPitié‐Salpêtrière University HospitalParisFrance
- Centre de Recherche en MyologieUMRS974Institut National de la Santé et de la Recherche MédicaleAssociation Institut de MyologieSorbonne UniversitéParisFrance
| | - Nozomu Tawara
- Department of Internal Medicine and Clinical ImmunlogySorbonne UniversitéPitié‐Salpêtrière University HospitalParisFrance
- Centre de Recherche en MyologieUMRS974Institut National de la Santé et de la Recherche MédicaleAssociation Institut de MyologieSorbonne UniversitéParisFrance
| | - Océane Landon‐Cardinal
- Division of RheumatologyCentre hospitalier de l'Université de Montréal (CHUM)CHUM Research CenterMontréalQCCanada
- Department of MedicineUniversité de MontréalMontréalQCCanada
| | - Céline Anquetil
- Department of Internal Medicine and Clinical ImmunlogySorbonne UniversitéPitié‐Salpêtrière University HospitalParisFrance
- Centre de Recherche en MyologieUMRS974Institut National de la Santé et de la Recherche MédicaleAssociation Institut de MyologieSorbonne UniversitéParisFrance
| | - Olivier Benveniste
- Department of Internal Medicine and Clinical ImmunlogySorbonne UniversitéPitié‐Salpêtrière University HospitalParisFrance
- Centre de Recherche en MyologieUMRS974Institut National de la Santé et de la Recherche MédicaleAssociation Institut de MyologieSorbonne UniversitéParisFrance
| | - Yves Allenbach
- Department of Internal Medicine and Clinical ImmunlogySorbonne UniversitéPitié‐Salpêtrière University HospitalParisFrance
- Centre de Recherche en MyologieUMRS974Institut National de la Santé et de la Recherche MédicaleAssociation Institut de MyologieSorbonne UniversitéParisFrance
| |
Collapse
|
15
|
Chasset F, Dayer JM, Chizzolini C. Type I Interferons in Systemic Autoimmune Diseases: Distinguishing Between Afferent and Efferent Functions for Precision Medicine and Individualized Treatment. Front Pharmacol 2021; 12:633821. [PMID: 33986670 PMCID: PMC8112244 DOI: 10.3389/fphar.2021.633821] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 03/03/2021] [Indexed: 12/19/2022] Open
Abstract
A sustained increase in type I interferon (IFN-I) may accompany clinical manifestations and disease activity in systemic autoimmune diseases (SADs). Despite the very frequent presence of IFN-I in SADs, clinical manifestations are extremely varied between and within SADs. The present short review will address the following key questions associated with high IFN-I in SADs in the perspective of precision medicine. 1) What are the mechanisms leading to high IFN-I? 2) What are the predisposing conditions favoring high IFN-I production? 3) What is the role of IFN-I in the development of distinct clinical manifestations within SADs? 4) Would therapeutic strategies targeting IFN-I be helpful in controlling or even preventing SADs? In answering these questions, we will underlie areas of incertitude and the intertwined role of autoantibodies, immune complexes, and neutrophils.
Collapse
Affiliation(s)
- François Chasset
- Department of Dermatology and Allergology, Faculty of Medicine, AP-HP, Tenon Hospital, Sorbonne University, Paris, France
| | - Jean-Michel Dayer
- Emeritus Professor of Medicine, School of Medicine, Geneva University, Geneva, Switzerland
| | - Carlo Chizzolini
- Department of Pathology and Immunology, School of Medicine, Geneva University, Geneva, Switzerland
| |
Collapse
|
16
|
Melki I, Devilliers H, Gitiaux C, Bondet V, Duffy D, Charuel JL, Miyara M, Bokov P, Kheniche A, Kwon T, Authier FJ, Allenbach Y, Belot A, Bodemer C, Bourrat E, Dumaine C, Fabien N, Faye A, Frémond ML, Hadchouel A, Kitabayashi N, Lepelley A, Martin-Niclos MJ, Mudumba S, Musset L, Quartier P, Rice GI, Seabra L, Uettwiller F, Uggenti C, Viel S, Rodero MP, Crow YJ, Bader-Meunier B. Anti-MDA5 juvenile idiopathic inflammatory myopathy: a specific subgroup defined by differentially enhanced interferon-α signalling. Rheumatology (Oxford) 2021; 59:1927-1937. [PMID: 31755959 DOI: 10.1093/rheumatology/kez525] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/03/2019] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVES JDM and juvenile overlap myositis represent heterogeneous subtypes of juvenile idiopathic inflammatory myopathy (JIIM). Chronic evolution can occur in up to 60% of cases, and morbidity/mortality is substantial. We aimed to describe the clinical, biological, histological and type I IFN status in JIIM associated with anti-melanoma differentiation-associated protein 5 (anti-MDA5) autoantibodies at presentation (group 1) in comparison with other JIIM (group 2). METHODS This was a retrospective and prospective study of patients with JIIM ascertained from three French paediatric rheumatology reference centres between 2013 and 2019. Muscle biopsies were reviewed. Type I interferon pathway activity was assessed by dosage of IFNα serum protein and the expression of IFN-stimulated genes. RESULTS Sixty-four patients were included, 13 in group 1 (54% JDM and 46% juvenile overlap myositis) and 51 in group 2 (76% JDM and 24% juvenile overlap myositis). Group 1 patients demonstrated more arthritis, skin ulcerations, lupus features and interstitial lung disease, and a milder muscular involvement. Serum IFNα levels were higher in group 1 than 2, and decreased after treatment or improvement in both groups. Outcome was similar in both groups. Unconventional treatment (more than two lines) was required in order to achieve remission, especially when skin ulceration was reported. CONCLUSION This study indicates a higher frequency of arthritis, skin ulcerations and interstitial lung disease, but milder muscular involvement, in JIIM with positive anti-MDA5 autoantibodies compared with other JIIM. Our data support an important role of systemic IFNα in disease pathology, particularly in the anti-MDA5 auto-antibody-positive subgroup. In severe and refractory forms of JIIM, IFNα may represent a therapeutic target.
Collapse
Affiliation(s)
- Isabelle Melki
- Laboratory of Neurogenetics and Neuroinflammation, Imagine Institute, Paris.,General Paediatrics, Infectious Disease and Internal Medicine Department, Reference center for Rheumatic, AutoImmune and Systemic diseases in children (RAISE), Hôpital Robert Debré, AP-HP, Paris.,Paediatric Hematology-Immunology and Rheumatology Department, Reference center for Rheumatic, AutoImmune and Systemic diseases in children (RAISE), Hôpital Necker-Enfants Malades, AP-HP, Paris
| | - Hervé Devilliers
- Centre Hospitalier Universitaire de Dijon, Hôpital François-Mitterrand, Service de Médecine Interne 2 et Centre d'Investigation Clinique, Inserm CIC 1432, Dijon
| | - Cyril Gitiaux
- Reference Centre for Neuromuscular Diseases, Necker-Enfants Malades Hospital, AP-HP.5, Paris.,Department of Paediatric Neurophysiology, Necker-Enfants Malades Hospital, AP-HP.5, Paris University, Paris.,INSERM U955-Team 10 'Biology of the Neuromuscular System', Paris Est-Creteil University, Creteil
| | - Vincent Bondet
- Immunobiology of Dendritic Cells, Institut Pasteur, Paris.,INSERM U1223, Paris
| | - Darragh Duffy
- Immunobiology of Dendritic Cells, Institut Pasteur, Paris.,INSERM U1223, Paris
| | - Jean-Luc Charuel
- Department of Immunology, Assistance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris
| | - Makoto Miyara
- Department of Immunology, Assistance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris
| | - Plamen Bokov
- Paediatric Physiology Department, Hôpital Robert Debré, AP-HP, Paris.,Université Paris Diderot, Paris
| | - Ahmed Kheniche
- Paediatric Radiology Department, Hôpital Robert Debré, AP-HP, Paris
| | - Theresa Kwon
- Nephrology Department, Hôpital Robert Debré, AP-HP, Paris
| | - François Jérôme Authier
- INSERM U955-Team 10 'Biology of the Neuromuscular System', Paris Est-Creteil University, Creteil.,Reference Centre for Neuromuscular Diseases, Henri Mondor University Hospital, Paris
| | - Yves Allenbach
- Département de médecine Interne et Immunologie Clinique, Centre de Référence Maladies Neuro-Musculaires, DHUi2B, AP-HP, GH Pitié-Salpêtrière, Paris.,Centre de Recherche en Myologie, UMRS 974 UPMC - INSERM, Paris
| | - Alexandre Belot
- Service de néphrologie, rhumatologie et dermatologie pédiatriques, Reference centre for Rheumatic, AutoImmune and Systemic diseases in children (RAISE), Filière des maladies autoimmunes et autoinflammatoires rares (FAI2R), Hôpital Femme Mère-Enfant, hospices civils de Lyon, Lyon.,Université de Lyon, Bron cedex, France.,Inserm U1111, Lyon
| | - Christine Bodemer
- National Reference Centre for Genodermatosis and Rare Diseases of the Skin (MAGEC).,Department of Dermatology, Necker-Enfants Malades Hospital, APHP5, Paris.,Imagine Institute, Inserm U 1163, Paris University, Paris
| | - Emmanuelle Bourrat
- General Paediatrics, Infectious Disease and Internal Medicine Department, Reference center for Rheumatic, AutoImmune and Systemic diseases in children (RAISE), Hôpital Robert Debré, AP-HP, Paris
| | - Cécile Dumaine
- General Paediatrics, Infectious Disease and Internal Medicine Department, Reference center for Rheumatic, AutoImmune and Systemic diseases in children (RAISE), Hôpital Robert Debré, AP-HP, Paris
| | - Nicole Fabien
- Université de Lyon, Bron cedex, France.,Department of Immunology, Reference centre for Rheumatic, AutoImmune and Systemic diseases in children (RAISE) Filière des maladies autoimmunes et autoinflammatoires rares (FAI2R), Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Lyon
| | - Albert Faye
- General Paediatrics, Infectious Disease and Internal Medicine Department, Reference center for Rheumatic, AutoImmune and Systemic diseases in children (RAISE), Hôpital Robert Debré, AP-HP, Paris.,Université Paris Diderot, Paris
| | - Marie-Louise Frémond
- Laboratory of Neurogenetics and Neuroinflammation, Imagine Institute, Paris.,Paediatric Hematology-Immunology and Rheumatology Department, Reference center for Rheumatic, AutoImmune and Systemic diseases in children (RAISE), Hôpital Necker-Enfants Malades, AP-HP, Paris
| | - Alice Hadchouel
- Paris University, Paris.,Paediatric Pulmonology, University Hospital Necker-Enfants Malades, AP-HP, Paris, France
| | - Naoki Kitabayashi
- Laboratory of Neurogenetics and Neuroinflammation, Imagine Institute, Paris
| | - Alice Lepelley
- Laboratory of Neurogenetics and Neuroinflammation, Imagine Institute, Paris
| | | | | | - Lucile Musset
- Department of Immunology, Assistance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris
| | - Pierre Quartier
- Paediatric Hematology-Immunology and Rheumatology Department, Reference center for Rheumatic, AutoImmune and Systemic diseases in children (RAISE), Hôpital Necker-Enfants Malades, AP-HP, Paris.,Imagine Institute, Inserm U 1163, Paris University, Paris
| | - Gillian I Rice
- Division of Evolution and Genomic Sciences, School of Biological Sciences, University of Manchester, Manchester, UK
| | - Luis Seabra
- Laboratory of Neurogenetics and Neuroinflammation, Imagine Institute, Paris
| | - Florence Uettwiller
- Paediatric Hematology-Immunology and Rheumatology Department, Reference center for Rheumatic, AutoImmune and Systemic diseases in children (RAISE), Hôpital Necker-Enfants Malades, AP-HP, Paris.,Transversal Unit of Allergology and Rheumatology, CHRU Tours, Tours, France
| | - Carolina Uggenti
- Laboratory of Neurogenetics and Neuroinflammation, Imagine Institute, Paris.,Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Sebastien Viel
- Université de Lyon, Bron cedex, France.,Inserm U1111, Lyon.,Department of Immunology, Reference centre for Rheumatic, AutoImmune and Systemic diseases in children (RAISE) Filière des maladies autoimmunes et autoinflammatoires rares (FAI2R), Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Lyon
| | - Mathieu P Rodero
- Laboratory of Neurogenetics and Neuroinflammation, Imagine Institute, Paris.,Chimie & Biologie, Modélisation et Immunologie pour la Thérapie (CBMIT), Université Paris Descartes, CNRS, UMR8601, Paris, France
| | - Yanick J Crow
- Laboratory of Neurogenetics and Neuroinflammation, Imagine Institute, Paris.,Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Brigitte Bader-Meunier
- Paediatric Hematology-Immunology and Rheumatology Department, Reference center for Rheumatic, AutoImmune and Systemic diseases in children (RAISE), Hôpital Necker-Enfants Malades, AP-HP, Paris.,Imagine Institute, Inserm U 1163, Paris University, Paris
| |
Collapse
|
17
|
Parkes JE, Thoma A, Lightfoot AP, Day PJ, Chinoy H, Lamb JA. MicroRNA and mRNA profiling in the idiopathic inflammatory myopathies. BMC Rheumatol 2020; 4:25. [PMID: 32529172 PMCID: PMC7285612 DOI: 10.1186/s41927-020-00125-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 03/20/2020] [Indexed: 12/12/2022] Open
Abstract
Background The idiopathic inflammatory myopathies (IIMs) are heterogeneous autoimmune conditions of skeletal muscle inflammation and weakness. MicroRNAs (miRNAs) are short, non-coding RNA which regulate gene expression of target mRNAs. The aim of this study was to profile miRNA and mRNA in IIM and identify miRNA-mRNA relationships which may be relevant to disease. Methods mRNA and miRNA in whole blood samples from 7 polymyositis (PM), 7 dermatomyositis (DM), 5 inclusion body myositis and 5 non-myositis controls was profiled using next generation RNA sequencing. Gene ontology and pathway analyses were performed using GOseq and Ingenuity Pathway Analysis. Dysregulation of miRNAs and opposite dysregulation of predicted target mRNAs in IIM subgroups was validated using RTqPCR and investigated by transfecting human skeletal muscle cells with miRNA mimic. Results Analysis of differentially expressed genes showed that interferon signalling, and anti-viral response pathways were upregulated in PM and DM compared to controls. An anti-Jo1 autoantibody positive subset of PM and DM (n = 5) had more significant upregulation and predicted activation of interferon signalling and highlighted T-helper (Th1 and Th2) cell pathways. In miRNA profiling miR-96-5p was significantly upregulated in PM, DM and the anti-Jo1 positive subset. RTqPCR replicated miR-96-5p upregulation and predicted mRNA target (ADK, CD28 and SLC4A10) downregulation. Transfection of a human skeletal muscle cell line with miR-96-5p mimic resulted in significant downregulation of ADK. Conclusion MiRNA and mRNA profiling identified dysregulation of interferon signalling, anti-viral response and T-helper cell pathways, and indicates a possible role for miR-96-5p regulation of ADK in pathogenesis of IIM.
Collapse
Affiliation(s)
- Joanna E Parkes
- Centre for Epidemiology, Division of Population Health, Health Services Research & Primary Care, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.,Stopford Building, University of Manchester, Oxford Road M13 9PT, Manchester, UK
| | - Anastasia Thoma
- Musculoskeletal Science & Sports Medicine Research Centre, School of Healthcare Science, Manchester Metropolitan University, Manchester, UK
| | - Adam P Lightfoot
- Musculoskeletal Science & Sports Medicine Research Centre, School of Healthcare Science, Manchester Metropolitan University, Manchester, UK
| | - Philip J Day
- Manchester Institute of Biotechnology, University of Manchester, Manchester, UK.,Division of Evolution & Genomic Sciences, University of Manchester, Manchester, UK
| | - Hector Chinoy
- Centre for Musculoskeletal Research, Division of Musculoskeletal & Dermatological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.,National Institute for Health Research Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.,Department of Rheumatology, Salford Royal NHS Foundation Trust, Manchester Academic Health Science Centre, Salford, UK
| | - Janine A Lamb
- Centre for Epidemiology, Division of Population Health, Health Services Research & Primary Care, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| |
Collapse
|
18
|
Pin A, Monasta L, Taddio A, Piscianz E, Tommasini A, Tesser A. An Easy and Reliable Strategy for Making Type I Interferon Signature Analysis Comparable among Research Centers. Diagnostics (Basel) 2019; 9:113. [PMID: 31487897 PMCID: PMC6787630 DOI: 10.3390/diagnostics9030113] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 08/30/2019] [Accepted: 09/03/2019] [Indexed: 01/13/2023] Open
Abstract
Interferon-stimulated genes (ISGs) are a set of genes whose transcription is induced by interferon (IFN). The measure of the expression of ISGs enables calculating an IFN score, which gives an indirect estimate of the exposition of cells to IFN-mediated inflammation. The measure of the IFN score is proposed for the screening of monogenic interferonopathies, like the Aicardi-Goutières syndrome, or to stratify subjects with systemic lupus erythematosus to receive IFN-targeted treatments. Apart from these scenarios, there is no agreement on the diagnostic value of the score in distinguishing IFN-related disorders from diseases dominated by other types of cytokines. Since the IFN score is currently measured in several research hospitals, merging experiences could help define the potential of scoring IFN inflammation in clinical practice. However, the IFN score calculated at different laboratories may be hardly comparable due to the distinct sets of IFN-stimulated genes assessed and to different controls used for data normalization. We developed a reliable approach to minimize the inter-laboratory variability, thereby providing shared strategies for the IFN signature analysis and allowing different centers to compare data and merge their experiences.
Collapse
Affiliation(s)
- Alessia Pin
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34127 Trieste, Italy.
| | - Lorenzo Monasta
- Clinical Epidemiology and Public Health Research Unit, Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", 34137 Trieste, Italy.
| | - Andrea Taddio
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34127 Trieste, Italy.
- Department of Paediatrics, Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", 34137 Trieste, Italy.
| | - Elisa Piscianz
- Department of Advanced Diagnostic and Clinical Trials, Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", 34137 Trieste, Italy.
| | - Alberto Tommasini
- Department of Paediatrics, Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", 34137 Trieste, Italy.
| | - Alessandra Tesser
- Department of Advanced Diagnostic and Clinical Trials, Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", 34137 Trieste, Italy.
| |
Collapse
|
19
|
Xie S, Luo H, Zhang H, Zhu H, Zuo X, Liu S. Discovery of Key Genes in Dermatomyositis Based on the Gene Expression Omnibus Database. DNA Cell Biol 2018; 37:982-992. [PMID: 30383435 DOI: 10.1089/dna.2018.4256] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The aim of this study was to identify biomarkers of dermatomyositis (DM). The analysis was conducted by retrieving DM-related cDNA microarray data sets from public databases. Gene ontology, Kyoto encyclopedia of genes and genomes, and protein-protein interaction analyses were performed, together with quantitative PCR-based detection of biomarkers in muscle tissue after stimulation with serum from patients with DM or healthy controls. Our analysis of five microarray data sets identified 20 common differentially expressed genes that are closely associated with DM. PCR analysis showed that mRNAs of IFITM2, LY6E, DDX58, and IFI6 were expressed at significantly higher levels in the muscle tissue of patients with DM than in normal muscle tissues. These mRNAs were also upregulated in human skeletal muscle cells stimulated with the serum from patients with DM. The results of integrated analyses of the DM microarray data and the mRNA levels of genes showed significant differences between the muscle tissues of DM patients and controls, which could indicate key pathogenic genes and novel therapeutic targets for DM.
Collapse
Affiliation(s)
- Shuoshan Xie
- 1 Rheumatology Department, Xiangya Hospital, Central South University , Changsha, China
| | - Hui Luo
- 1 Rheumatology Department, Xiangya Hospital, Central South University , Changsha, China
| | - Huali Zhang
- 2 Department of Pathophysiology, Xiangya School of Medicine, Central South University , Changsha, China
| | - Honglin Zhu
- 1 Rheumatology Department, Xiangya Hospital, Central South University , Changsha, China
| | - Xiaoxia Zuo
- 1 Rheumatology Department, Xiangya Hospital, Central South University , Changsha, China
| | - Sijia Liu
- 1 Rheumatology Department, Xiangya Hospital, Central South University , Changsha, China
| |
Collapse
|
20
|
Rodríguez-Alba JC, Girón-Pérez DA, Romero-Ramírez H, Pelayo R, Santos-Argumedo L. Regulatory IFN-γ-producing killer dendritic cells are enhanced in B6.MLR-Fas lpr /J lupus-prone mice. Eur J Immunol 2018; 48:1851-1860. [PMID: 30289564 DOI: 10.1002/eji.201847547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 08/22/2018] [Accepted: 10/04/2018] [Indexed: 11/09/2022]
Abstract
A novel cell population denominated IFN-γ-producing killer dendritic cells (IKDCs) have been recently described. These cells are lymphocytes lacking B- or T- receptors, but they can be identified by the presence of B220+ CD38+ CD49b+ and low CD11c, among other cell surface markers. The main characteristics of IKDCs are the production of IFN-γ and the ability to spontaneously kill tumor cells. We found that this population increases in B6.MLR-Faslpr /J mice. Interestingly, IKDCs increase with age and are more abundant in mice older than 6 months onward. To analyze whether these cells have any role in the induction of the lupus-like phenotype in the B6.MLR-Faslpr /J mice, IKDCs were purified and transferred into 6-month-old B6.MRL-Faslpr /J mice, then the presence of anti-nuclear antibodies (ANAS) and anti-dsDNA antibodies were analyzed 2 and 4 months after the transfer. The results showed a reduction in the levels of these autoantibodies and increased survival of these mice, indicating that these cells may have a regulatory function. In vitro assays demonstrated that IKDCs reduced the proliferation of both autoreactive B and T cells, suggesting that these may be the mechanisms used by these cells to ameliorate the lupus-like phenotype in the B6.MRL-Faslpr /J mice.
Collapse
Affiliation(s)
- Juan Carlos Rodríguez-Alba
- Unidad de Citometría de Flujo, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa, Veracruz, México
| | | | | | - Rosana Pelayo
- Centro de Investigación Biomédica de Oriente-CIBIOR, IMSS, Puebla, Mexico
| | | |
Collapse
|
21
|
Soponkanaporn S, Deakin CT, Schutz PW, Marshall LR, Yasin SA, Johnson CM, Sag E, Tansley SL, McHugh NJ, Wedderburn LR, Jacques TS. Expression of myxovirus-resistance protein A: a possible marker of muscle disease activity and autoantibody specificities in juvenile dermatomyositis. Neuropathol Appl Neurobiol 2018; 45:410-420. [PMID: 29770465 PMCID: PMC6563435 DOI: 10.1111/nan.12498] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 05/01/2018] [Indexed: 12/15/2022]
Abstract
Aims To evaluate the relationship between expression of myxovirus‐resistance protein A (MxA) protein on muscle biopsies by immunohistochemistry and disease activity in juvenile dermatomyositis (JDM) patients. Also, another aim was to investigate whether the expression of MxA is related with myositis‐specific autoantibodies (MSA) status in JDM patients. Methods 103 patients (median aged 6.3, interquartile range 0.5–15.9) enrolled in the Juvenile Dermatomyositis Cohort and Biomarker Study (JDCBS). Muscle biopsies were stained with MxA and scored. Clinical data at initial presentation were collected and autoantibodies were analysed. Multiple linear regression analysis was performed to estimate the association between MxA expression on muscle fibres and muscle disease activity, and MSA status. Results Expression of MxA protein on JDM samples was identified in 61.2%. There was a significant association between MxA scores and Childhood Myositis Assessment Scale (CMAS) (P = 0.002), and Manual Muscle Testing of Eight Muscles (MMT8) (P = 0.026). CMAS and MMT8 scores were significantly lower in the group of patients with strong MxA expression. MxA scores differed according to MSA subgroups (P = 0.002). Patients with positive nuclear matrix protein 2 autoantibodies had strong MxA expression, whereas anti‐melanoma differentiation‐associated gene 5 positive patients had no or weak MxA expression. Conclusions This study reveals the significant association between level of MxA expression on muscle fibres and clinical measures of muscular disease activity in JDM patients and MSA status. This confirms type I interferonopathies in muscle fibres of JDM patients which could help with improving treatment outcome in JDM patients and underscoring the distinct pathophysiological pathways in different MSA status.
Collapse
Affiliation(s)
- S Soponkanaporn
- Infection, Immunity and Inflammation Programme, UCL Great Ormond Street Institute of Child Health, London, UK.,Division of Rheumatology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - C T Deakin
- Infection, Immunity and Inflammation Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - P W Schutz
- Infection, Immunity and Inflammation Programme, UCL Great Ormond Street Institute of Child Health, London, UK.,Division of Neuropathology, Vancouver General Hospital, Vancouver, BC, Canada.,Department of Pathology, University of British Columbia, Vancouver, BC, Canada
| | - L R Marshall
- Infection, Immunity and Inflammation Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - S A Yasin
- Infection, Immunity and Inflammation Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - C M Johnson
- Infection, Immunity and Inflammation Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - E Sag
- Infection, Immunity and Inflammation Programme, UCL Great Ormond Street Institute of Child Health, London, UK.,Department of Pediatric Rheumatology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - S L Tansley
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | - N J McHugh
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | - L R Wedderburn
- Infection, Immunity and Inflammation Programme, UCL Great Ormond Street Institute of Child Health, London, UK.,Rheumatology Unit, Great Ormond Street Hospital for Children, London, UK.,NIHR Biomedical Research Centre at Great Ormond Street Hospital for Children, NHS Foundation Trust and University College London, London, UK.,Arthritis Research UK Centre for Adolescent Rheumatology at UCL, UCLH and GOSH, London, UK
| | - T S Jacques
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
22
|
Castañeda-Delgado JE, Bastián-Hernandez Y, Macias-Segura N, Santiago-Algarra D, Castillo-Ortiz JD, Alemán-Navarro AL, Martínez-Tejada P, Enciso-Moreno L, Garcia-De Lira Y, Olguín-Calderón D, Trouw LA, Ramos-Remus C, Enciso-Moreno JA. Type I Interferon Gene Response Is Increased in Early and Established Rheumatoid Arthritis and Correlates with Autoantibody Production. Front Immunol 2017; 8:285. [PMID: 28373872 PMCID: PMC5357778 DOI: 10.3389/fimmu.2017.00285] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Accepted: 02/28/2017] [Indexed: 12/21/2022] Open
Abstract
Background Rheumatoid arthritis (RA) is an inflammatory debilitating disease that affects the joints in the early and productive phases of an individual’s life. Several cytokines have been linked to the disease pathogenesis and are known to contribute to the inflammatory state characteristic of RA. The participation of type I interferon (IFN) in the pathogenesis of the disease has been already described as well as the identity of the genes that are regulated by this molecule, which are collectively known as the type I IFN signature. These genes have several functions associated with apoptosis, transcriptional regulation, protein degradation, Th2 cell induction, B cell proliferation, etc. This article evaluated the expression of several genes of the IFN signature in different stages of disease and their correlation with the levels of anticitrullinated protein antibodies (ACPA) anticarbamylated protein (Anti-CarP) antibodies. Methods Samples from individuals with early and established RA, high-risk individuals (ACPA+ and ACPA−), and healthy controls were recruited at “Unidad de Artritis y Rheumatismo” (Rheumatism and Arthritis Unit) in Guadalajara Jalisco Mexico. Determinations of ACPA were made with Eurodiagnostica ACPA plus kit. Anti-CarP determinations were made according to previously described protocols. RNA was isolated, and purity and integrity were determined according to RNA integrity number >6. Gene expression analysis was made by RT-qPCR using specific primers for mRNAs of the type I IFN signature. Relative gene expression was calculated according to Livak and Schmitgen. Results Significant differences in gene expression were identified when comparing the different groups for MXA and MXB (P < 0.05), also when comparing established RA and ACPA− in both IFIT 1 and G15. An increased expression of ISG15 was identified (P < 0.05), and a clear tendency toward increase was identified for HERC5. EPSTRI1, IFI6, and IFI35 were found to be elevated in the chronic/established RA and early RA (P < 0.05). Significant correlations were identified for the IFN signature genes with the levels of ACPA and anti-CarP (P < 0.05). Conclusion Our data confirm previous observations in the role of IFN signature and the pathogenesis of RA. Also, we provide evidence of an association between several genes of the IFN signature (that regulate Th2 cells and B cell proliferation) with the levels of anti-CarP antibodies and ACPA.
Collapse
Affiliation(s)
- Julio E Castañeda-Delgado
- Medical research Unit of Zacatecas, Mexican Institute of Social Security, UIMZ-IMSS, Zacatecas, Mexico; National Council of Science and Technology, CONACYT, Catedras-CONACYT, Zacatecas, Mexico
| | - Yadira Bastián-Hernandez
- Medical research Unit of Zacatecas, Mexican Institute of Social Security, UIMZ-IMSS, Zacatecas, Mexico; National Council of Science and Technology, CONACYT, Catedras-CONACYT, Zacatecas, Mexico
| | - Noe Macias-Segura
- Medical research Unit of Zacatecas, Mexican Institute of Social Security, UIMZ-IMSS, Zacatecas, Mexico; Departamento de fisiología y farmacología, centro de ciencias básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, Aguascalientes, Mexico
| | - David Santiago-Algarra
- Medical research Unit of Zacatecas, Mexican Institute of Social Security, UIMZ-IMSS , Zacatecas , Mexico
| | - Jose D Castillo-Ortiz
- Unidad de Investigación en Enfermedades Crónico-Degenerativas , Guadalajara, Jalisco , México
| | - Ana L Alemán-Navarro
- Medical research Unit of Zacatecas, Mexican Institute of Social Security, UIMZ-IMSS , Zacatecas , Mexico
| | - Pedro Martínez-Tejada
- General Hospital: "Emilio Varela Lujan", Mexican Institute of Social Security, IMSS , Zacatecas , Mexico
| | - Leonor Enciso-Moreno
- Medical research Unit of Zacatecas, Mexican Institute of Social Security, UIMZ-IMSS , Zacatecas , Mexico
| | - Yolanda Garcia-De Lira
- Medical research Unit of Zacatecas, Mexican Institute of Social Security, UIMZ-IMSS , Zacatecas , Mexico
| | - Diana Olguín-Calderón
- Medical research Unit of Zacatecas, Mexican Institute of Social Security, UIMZ-IMSS , Zacatecas , Mexico
| | - Leendert A Trouw
- Department of Rheumatology, Leiden University Medical Center , Leiden , Netherlands
| | | | - Jose A Enciso-Moreno
- Medical research Unit of Zacatecas, Mexican Institute of Social Security, UIMZ-IMSS , Zacatecas , Mexico
| |
Collapse
|
23
|
The host defense peptide LL-37 a possible inducer of the type I interferon system in patients with polymyositis and dermatomyositis. J Autoimmun 2017; 78:46-56. [DOI: 10.1016/j.jaut.2016.12.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 12/01/2016] [Accepted: 12/06/2016] [Indexed: 11/22/2022]
|
24
|
Serum level of DNase1l3 in patients with dermatomyositis/polymyositis, systemic lupus erythematosus and rheumatoid arthritis, and its association with disease activity. Clin Exp Med 2016; 17:459-465. [DOI: 10.1007/s10238-016-0448-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 12/19/2016] [Indexed: 11/26/2022]
|