1
|
Covert LT, Osman A, Truskey GA. Interferon-β-Induced Injury During Pediatric Muscle Differentiation: Insight Into Juvenile Dermatomyositis Pathogenesis. ACR Open Rheumatol 2025; 7:e11760. [PMID: 39439064 DOI: 10.1002/acr2.11760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/20/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024] Open
Abstract
OBJECTIVE Juvenile dermatomyositis (JDM) involves up-regulated type I interferons (IFNs), including IFNβ, yet pathologic mechanisms remain poorly understood. We aimed to characterize the functional and structural effects of IFNβ on in vitro human pediatric myoblast growth and differentiation in a three-dimensional skeletal muscle model (myobundles). METHODS Myobundles fabricated from myoblasts of a healthy pediatric donor were exposed to IFNβ at 0 to 5,600 IU/mL during growth (days 1-4), differentiation (days 4-11), and/or mature (days 11-18) periods. To assess myobundle structure and function, contractile force, kinetics, and fatigue were measured at day 18 with subsequent immunohistochemistry. RESULTS Myobundles were not functionally affected by IFNβ exposure during growth period alone. However, when IFNβ exposure continued through differentiation, myobundles became dysfunctional (P < 0.0001). IFNβ during differentiation or mature periods alone resulted in dose-dependent decreases in contractility, with greater decrease in the differentiation alone group (P < 0.0001). Twitch kinetics and fatigue remained largely unchanged when myobundles were exposed to IFNβ only during growth, yet twitch time slowed (P < 0.005) and fatigue decreased (P < 0.002) when myobundles were exposed during differentiation or mature stages alone. Nuclei density and myofiber size and organization also decreased when IFNβ was added during differentiation period alone. CONCLUSION IFNβ decreases pediatric myobundle contractile function most significantly during differentiation of myoblasts to myotubes. Function is not affected when IFNβ exposure is limited to myoblast proliferation alone. These findings implicate a pathologic role for IFNβ in JDM by impairing myoblast differentiation, leading to subsequent loss of function and ongoing need for muscle regeneration and repair.
Collapse
|
2
|
Covert LT, Prinz JA, Swain-Lenz D, Dvergsten J, Truskey GA. Genetic changes from type I interferons and JAK inhibitors: clues to drivers of juvenile dermatomyositis. Rheumatology (Oxford) 2024; 63:SI240-SI248. [PMID: 38317053 PMCID: PMC11381683 DOI: 10.1093/rheumatology/keae082] [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: 11/09/2023] [Revised: 12/21/2023] [Accepted: 01/18/2024] [Indexed: 02/07/2024] Open
Abstract
OBJECTIVE To better understand the pathogenesis of juvenile dermatomyositis (JDM), we examined the effect of the cytokines type I interferons (IFN I) and JAK inhibitor drugs (JAKi) on gene expression in bioengineered pediatric skeletal muscle. METHODS Myoblasts from three healthy pediatric donors were used to create three-dimensional skeletal muscle units termed myobundles. Myobundles were treated with IFN I, either IFNα or IFNβ. A subset of IFNβ-exposed myobundles was treated with JAKi tofacitinib or baricitinib. RNA sequencing analysis was performed on all myobundles. RESULTS Seventy-six myobundles were analysed. Principal component analysis showed donor-specific clusters of gene expression across IFNα and IFNβ-exposed myobundles in a dose-dependent manner. Both cytokines upregulated interferon response and proinflammatory genes; however, IFNβ led to more significant upregulation. Key downregulated pathways involved oxidative phosphorylation, fatty acid metabolism and myogenesis genes. Addition of tofacitinib or baricitinib moderated the gene expression induced by IFNβ, with partial reversal of upregulated inflammatory and downregulated myogenesis pathways. Baricitinib altered genetic profiles more than tofacitinib. CONCLUSION IFNβ leads to more pro-inflammatory gene upregulation than IFNα, correlating to greater decrease in contractile protein gene expression and reduced contractile force. JAK inhibitors, baricitinib more so than tofacitinib, partially reverse IFN I-induced genetic changes. Increased IFN I exposure in healthy bioengineered skeletal muscle leads to IFN-inducible gene expression, inflammatory pathway enrichment, and myogenesis gene downregulation, consistent with what is observed in JDM.
Collapse
Affiliation(s)
- Lauren T Covert
- Department of Pediatrics, Duke University Health System, Durham, NC, USA
| | - Joseph A Prinz
- Sequencing and Genomics Technologies Core Facility, School of Medicine, Duke University, Durham, NC, USA
| | - Devjanee Swain-Lenz
- Sequencing and Genomics Technologies Core Facility, School of Medicine, Duke University, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, USA
| | - Jeffrey Dvergsten
- Department of Pediatrics, Duke University Health System, Durham, NC, USA
| | - George A Truskey
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| |
Collapse
|
3
|
Pinal-Fernandez I, Muñoz-Braceras S, Casal-Dominguez M, Pak K, Torres-Ruiz J, Musai J, Dell’Orso S, Naz F, Islam S, Gutierrez-Cruz G, Cano MD, Matas-Garcia A, Padrosa J, Tobías-Baraja E, Garrabou G, Aldecoa I, Espinosa G, Simeon-Aznar CP, Guillen-Del-Castillo A, Gil-Vila A, Trallero-Araguas E, Christopher-Stine L, Lloyd TE, Liewluck T, Naddaf E, Stenzel W, Greenberg SA, Grau JM, Selva-O’Callaghan A, Milisenda JC, Mammen AL. Pathogenic autoantibody internalization in myositis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.15.24301339. [PMID: 38313303 PMCID: PMC10836124 DOI: 10.1101/2024.01.15.24301339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
Objectives Myositis is a heterogeneous family of autoimmune muscle diseases. As myositis autoantibodies recognize intracellular proteins, their role in disease pathogenesis has been unclear. This study aimed to determine whether myositis autoantibodies reach their autoantigen targets within muscle cells and disrupt the normal function of these proteins. Methods Confocal immunofluorescence microscopy was used to localize antibodies and other proteins of interest in myositis muscle biopsies. Bulk RNA sequencing was used to study the transcriptomic profiles of 668 samples from patients with myositis, disease controls, and healthy controls. Antibodies from myositis patients were introduced into cultured myoblasts by electroporation and the transcriptomic profiles of the treated myoblasts were studied by bulk RNA sequencing. Results In patients with myositis autoantibodies, antibodies accumulated inside myofibers in the same subcellular compartment as the autoantigen. Each autoantibody was associated with effects consistent with dysfunction of its autoantigen, such as the derepression of genes normally repressed by Mi2/NuRD in patients with anti-Mi2 autoantibodies, the accumulation of RNAs degraded by the nuclear RNA exosome complex in patients with anti-PM/Scl autoantibodies targeting this complex, and the accumulation of lipids within myofibers of anti-HMGCR-positive patients. Internalization of patient immunoglobulin into cultured myoblasts recapitulated the transcriptomic phenotypes observed in human disease, including the derepression of Mi2/NuRD-regulated genes in anti-Mi2-positive dermatomyositis and the increased expression of genes normally degraded by the nuclear RNA exosome complex in anti-PM/Scl-positive myositis. Conclusions In myositis, autoantibodies are internalized into muscle fibers, disrupt the biological function of their autoantigen, and mediate the pathophysiology of the disease.
Collapse
Affiliation(s)
- Iago Pinal-Fernandez
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sandra Muñoz-Braceras
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Maria Casal-Dominguez
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Katherine Pak
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jiram Torres-Ruiz
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
- Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Jon Musai
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Stefania Dell’Orso
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Faiza Naz
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shamima Islam
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Gustavo Gutierrez-Cruz
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Maria Dolores Cano
- Muscle Research Unit, Internal Medicine Service, Hospital Clinic, Barcelona, Spain
| | - Ana Matas-Garcia
- Muscle Research Unit, Internal Medicine Service, Hospital Clinic, Barcelona, Spain
- Barcelona University, Barcelona, Spain
- CIBERER and IDIBAPS, Barcelona, Spain
| | | | - Esther Tobías-Baraja
- Muscle Research Unit, Internal Medicine Service, Hospital Clinic, Barcelona, Spain
- Barcelona University, Barcelona, Spain
- CIBERER and IDIBAPS, Barcelona, Spain
| | - Gloria Garrabou
- Muscle Research Unit, Internal Medicine Service, Hospital Clinic, Barcelona, Spain
- Barcelona University, Barcelona, Spain
- CIBERER and IDIBAPS, Barcelona, Spain
| | - Iban Aldecoa
- Pathology, Neurological Tissue Bank. Hospital Clinic of Barcelona-CDB-IDIBAPS/FCRB-University of Barcelona, Barcelona, Spain
| | - Gerard Espinosa
- Barcelona University, Barcelona, Spain
- Department of Autoimmune Diseases, Reference Centre for Systemic Autoimmune Diseases (UEC/CSUR) of the Catalan and Spanish Health Systems-Member of ERN-ReCONNET, Hospital Clinic, Barcelona, Spain
| | - Carmen Pilar Simeon-Aznar
- Systemic Autoimmune Disease Section, Vall d’Hebron Institute of Research, Barcelona, Spain
- Autonomous University of Barcelona, Barcelona, Spain
| | - Alfredo Guillen-Del-Castillo
- Systemic Autoimmune Disease Section, Vall d’Hebron Institute of Research, Barcelona, Spain
- Autonomous University of Barcelona, Barcelona, Spain
| | - Albert Gil-Vila
- Systemic Autoimmune Disease Section, Vall d’Hebron Institute of Research, Barcelona, Spain
- Autonomous University of Barcelona, Barcelona, Spain
| | - Ernesto Trallero-Araguas
- Systemic Autoimmune Disease Section, Vall d’Hebron Institute of Research, Barcelona, Spain
- Autonomous University of Barcelona, Barcelona, Spain
| | - Lisa Christopher-Stine
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thomas E. Lloyd
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Teerin Liewluck
- Division of Neuromuscular Medicine, Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Elie Naddaf
- Division of Neuromuscular Medicine, Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Werner Stenzel
- Department of Neuropathology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Steven A. Greenberg
- Department of Neurology, Brigham and Women’s Hospital and Boston Children’s Hospital, Harvard Medical School, MA, USA
| | - Josep Maria Grau
- Muscle Research Unit, Internal Medicine Service, Hospital Clinic, Barcelona, Spain
- Barcelona University, Barcelona, Spain
- CIBERER and IDIBAPS, Barcelona, Spain
| | - Albert Selva-O’Callaghan
- Systemic Autoimmune Disease Section, Vall d’Hebron Institute of Research, Barcelona, Spain
- Autonomous University of Barcelona, Barcelona, Spain
| | - Jose C. Milisenda
- Muscle Research Unit, Internal Medicine Service, Hospital Clinic, Barcelona, Spain
- Barcelona University, Barcelona, Spain
- CIBERER and IDIBAPS, Barcelona, Spain
| | - Andrew L. Mammen
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Autonomous University of Barcelona, Barcelona, Spain
| |
Collapse
|
4
|
Covert LT, Patel H, Osman A, Duncan L, Dvergsten J, Truskey GA. Effect of type I interferon on engineered pediatric skeletal muscle: a promising model for juvenile dermatomyositis. Rheumatology (Oxford) 2024; 63:209-217. [PMID: 37094222 PMCID: PMC10765138 DOI: 10.1093/rheumatology/kead186] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/24/2023] [Accepted: 04/14/2023] [Indexed: 04/26/2023] Open
Abstract
OBJECTIVE To investigate pathogenic mechanisms underlying JDM, we defined the effect of type I IFN, IFN-α and IFN-β, on pediatric skeletal muscle function and expression of myositis-related proteins using an in vitro engineered human skeletal muscle model (myobundle). METHODS Primary myoblasts were isolated from three healthy pediatric donors and used to create myobundles that mimic functioning skeletal muscle in structural architecture and physiologic function. Myobundles were exposed to 0, 5, 10 or 20 ng/ml IFN-α or IFN-β for 7 days and then functionally tested under electrical stimulation and analyzed immunohistochemically for structural and myositis-related proteins. Additionally, IFN-β-exposed myobundles were treated with Janus kinase inhibitors (JAKis) tofacitinib and baricitinib. These myobundles were also analyzed for contractile force and immunohistochemistry. RESULTS IFN-β, but not IFN-α, was associated with decreased contractile tetanus force and slowed twitch kinetics. These effects were reversed by tofacitinib and baricitinib. Type I IFN paradoxically reduced myobundle fatigue, which did not reverse after JAKi. Additionally, type I IFN correlated with MHC I upregulation, which normalized after JAKi treatment, but expression of myositis-specific autoantigens Mi-2, melanocyte differentiation-associated protein 5 and the endoplasmic reticulum stress marker GRP78 were variable and donor specific after type I IFN exposure. CONCLUSION IFN-α and IFN-β have distinct effects on pediatric skeletal muscle and these effects can partially be reversed by JAKi treatment. This is the first study illustrating effective use of a three-dimensional human skeletal muscle model to investigate JDM pathogenesis and test novel therapeutics.
Collapse
Affiliation(s)
- Lauren T Covert
- Department of Pediatrics, Duke University Health System, Durham, NC, USA
| | - Hailee Patel
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Alaa Osman
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Lavonia Duncan
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Jeffrey Dvergsten
- Department of Pediatrics, Duke University Health System, Durham, NC, USA
| | - George A Truskey
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| |
Collapse
|
5
|
Jeong HN, Lee TG, Park HJ, Yang Y, Oh SH, Kang SW, Choi YC. Transcriptome analysis of skeletal muscle in dermatomyositis, polymyositis, and dysferlinopathy, using a bioinformatics approach. Front Neurol 2023; 14:1328547. [PMID: 38125829 PMCID: PMC10731051 DOI: 10.3389/fneur.2023.1328547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023] Open
Abstract
Background Polymyositis (PM) and dermatomyositis (DM) are two distinct subgroups of idiopathic inflammatory myopathies. Dysferlinopathy, caused by a dysferlin gene mutation, usually presents in late adolescence with muscle weakness, degenerative muscle changes are often accompanied by inflammatory infiltrates, often resulting in a misdiagnosis as polymyositis. Objective To identify differential biological pathways and hub genes related to polymyositis, dermatomyositis and dysferlinopathy using bioinformatics analysis for understanding the pathomechanisms and providing guidance for therapy development. Methods We analyzed intramuscular ribonucleic acid (RNA) sequencing data from seven dermatomyositis, eight polymyositis, eight dysferlinopathy and five control subjects. Differentially expressed genes (DEGs) were identified by using DESeq2. Enrichment analyses were performed to understand the functions and enriched pathways of DEGs. A protein-protein interaction (PPI) network was constructed, and clarified the gene cluster using the molecular complex detection tool (MCODE) analysis to identify hub genes. Results A total of 1,048, 179 and 3,807 DEGs were detected in DM, PM and dysferlinopathy, respectively. Enrichment analyses revealed that upregulated DEGs were involved in type 1 interferon (IFN1) signaling pathway in DM, antigen processing and presentation of peptide antigen in PM, and cellular response to stimuli in dysferlinopathy. The PPI network and MCODE cluster identified 23 genes related to type 1 interferon signaling pathway in DM, 4 genes (PDIA3, HLA-C, B2M, and TAP1) related to MHC class 1 formation and quality control in PM, and 7 genes (HSPA9, RPTOR, MTOR, LAMTOR1, LAMTOR5, ATP6V0D1, and ATP6V0B) related to cellular response to stress in dysferliniopathy. Conclusion Overexpression of genes related to the IFN1 signaling pathway and major histocompatibility complex (MHC) class I formation was identified in DM and PM, respectively. In dysferlinopathy, overexpression of HSPA9 and the mTORC1 signaling pathway genes was detected.
Collapse
Affiliation(s)
- Ha-Neul Jeong
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Taek Gyu Lee
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Republic of Korea
| | - Hyung Jun Park
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young Yang
- Research Institute of Women's Disease, Sookmyumg Women's University, Seoul, Republic of Korea
| | - Seung-Hun Oh
- Department of Neurology, CHA Bundang Medical Center, School of Medicine, CHA University, Seongnam-si, Republic of Korea
| | - Seong-Woong Kang
- Department of Rehabilitation Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
- Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young-Chul Choi
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
6
|
Olie CS, Pinto-Fernández A, Damianou A, Vendrell I, Mei H, den Hamer B, van der Wal E, de Greef JC, Raz V, Kessler BM. USP18 is an essential regulator of muscle cell differentiation and maturation. Cell Death Dis 2023; 14:231. [PMID: 37002195 PMCID: PMC10066380 DOI: 10.1038/s41419-023-05725-z] [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: 10/27/2022] [Revised: 02/14/2023] [Accepted: 03/07/2023] [Indexed: 04/03/2023]
Abstract
The ubiquitin proteasomal system is a critical regulator of muscle physiology, and impaired UPS is key in many muscle pathologies. Yet, little is known about the function of deubiquitinating enzymes (DUBs) in the muscle cell context. We performed a genetic screen to identify DUBs as potential regulators of muscle cell differentiation. Surprisingly, we observed that the depletion of ubiquitin-specific protease 18 (USP18) affected the differentiation of muscle cells. USP18 depletion first stimulated differentiation initiation. Later, during differentiation, the absence of USP18 expression abrogated myotube maintenance. USP18 enzymatic function typically attenuates the immune response by removing interferon-stimulated gene 15 (ISG15) from protein substrates. However, in muscle cells, we found that USP18, predominantly nuclear, regulates differentiation independent of ISG15 and the ISG response. Exploring the pattern of RNA expression profiles and protein networks whose levels depend on USP18 expression, we found that differentiation initiation was concomitant with reduced expression of the cell-cycle gene network and altered expression of myogenic transcription (co) factors. We show that USP18 depletion altered the calcium channel gene network, resulting in reduced calcium flux in myotubes. Additionally, we show that reduced expression of sarcomeric proteins in the USP18 proteome was consistent with reduced contractile force in an engineered muscle model. Our results revealed nuclear USP18 as a critical regulator of differentiation initiation and maintenance, independent of ISG15 and its role in the ISG response.
Collapse
Affiliation(s)
- Cyriel Sebastiaan Olie
- Human Genetics department, Leiden University Medical Centre, 2333ZC, Leiden, The Netherlands
| | - Adán Pinto-Fernández
- Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7FZ, UK
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Andreas Damianou
- Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7FZ, UK
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Iolanda Vendrell
- Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7FZ, UK
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - Hailiang Mei
- Sequencing Analysis Support Core, Leiden University Medical Centre, 2333ZC, Leiden, The Netherlands
| | - Bianca den Hamer
- Human Genetics department, Leiden University Medical Centre, 2333ZC, Leiden, The Netherlands
| | - Erik van der Wal
- Human Genetics department, Leiden University Medical Centre, 2333ZC, Leiden, The Netherlands
| | - Jessica C de Greef
- Human Genetics department, Leiden University Medical Centre, 2333ZC, Leiden, The Netherlands
| | - Vered Raz
- Human Genetics department, Leiden University Medical Centre, 2333ZC, Leiden, The Netherlands.
| | - Benedikt M Kessler
- Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Roosevelt Drive, Oxford, OX3 7FZ, UK.
- Target Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK.
| |
Collapse
|
7
|
Abstract
PURPOSE OF REVIEW This review summarizes and comments on current knowledge in dermatomyositis. RECENT FINDINGS The 2018 European Neuromuscular Centre classification of dermatomyositis has been challenging by the discovery of clinicopathological features associated with dermatomyositis-specific antibody (DMSA) that were not incorporated in the original criteria. These features include but may not be limited to the presence of perifascicular necrosis in anti-Mi-2 dermatomyositis; presence of diffuse nonperifascicular sarcoplasmic myxovirus resistance protein A expression in anti-MDA5 dermatomyositis; and dermatomyositis sine dermatitis in anti-NXP-2 dermatomyositis. Variations and subclassifications within the same DMSA subtypes are observed: anti-MDA5 dermatomyositis is clinically subcategorized into good, intermediate, and poor prognostic subgroups; concurrent anti-CCAR1 and anti-TIF1-γ positivity identify anti-TIF1-γ-positive patient with a lower risk for cancer-associated myositis. Owing to distinct IFN1-signaling pathway activation in dermatomyositis, JAK-STAT inhibitor - the pathway-targeted therapy, have been studied with promising results in refractory dermatomyositis and some new-onset dermatomyositis. In addition, the potential serum biomarkers for IFN1 pathway activation are being investigated for their performance in monitoring the disease activity and the efficacy of the treatment. SUMMARY DMSA, evidence of prominent IFN1 pathway activation, and risk/severity-associated biomarkers would likely play major roles in future dermatomyositis classification, disease monitoring, and treatment decision.
Collapse
Affiliation(s)
- Jantima Tanboon
- Department of Pathology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Department of Neuromuscular Research, National Institute of Neuroscience (NIN), National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Ichizo Nishino
- Department of Neuromuscular Research, National Institute of Neuroscience (NIN), National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
- Department of Genome Medicine Development
- Department of Clinical Genome Analysis, Medical Genome Center (MGC), National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| |
Collapse
|
8
|
Huang P, Tang L, Zhang L, Ren Y, Peng H, Xiao Y, Xu J, Mao D, Liu L, Liu L. Identification of Biomarkers Associated With CD4+ T-Cell Infiltration With Gene Coexpression Network in Dermatomyositis. Front Immunol 2022; 13:854848. [PMID: 35711463 PMCID: PMC9196312 DOI: 10.3389/fimmu.2022.854848] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 04/27/2022] [Indexed: 12/19/2022] Open
Abstract
Background Dermatomyositis is an autoimmune disease characterized by damage to the skin and muscles. CD4+ T cells are of crucial importance in the occurrence and development of dermatomyositis (DM). However, there are few bioinformatics studies on potential pathogenic genes and immune cell infiltration of DM. Therefore, this study intended to explore CD4+ T-cell infiltration–associated key genes in DM and construct a new model to predict the level of CD4+ T-cell infiltration in DM. Methods GSE46239, GSE142807, GSE1551, and GSE193276 datasets were downloaded. The WGCNA and CIBERSORT algorithms were performed to identify the most correlated gene module with CD4+ T cells. Matascape was used for GO enrichment and KEGG pathway analysis of the key gene module. LASSO regression analysis was used to identify the key genes and construct the prediction model. The correlation between the key genes and CD4+ T-cell infiltration was investigated. GSEA was performed to research the underlying signaling pathways of the key genes. The key gene-correlated transcription factors were identified through the RcisTarget and Gene-motif rankings databases. The miRcode and DIANA-LncBase databases were used to build the lncRNA-miRNA-mRNA network. Results In the brown module, 5 key genes (chromosome 1 open reading frame 106 (C1orf106), component of oligomeric Golgi complex 8 (COG8), envoplakin (EVPL), GTPases of immunity-associated protein family member 6 (GIMAP6), and interferon-alpha inducible protein 6 (IFI6)) highly associated with CD4+ T-cell infiltration were identified. The prediction model was constructed and showed better predictive performance in the training set, and this satisfactory model performance was validated in another skin biopsy dataset and a muscle biopsy dataset. The expression levels of the key genes promoted the CD4+ T-cell infiltration. GSEA results revealed that the key genes were remarkably enriched in many immunity-associated pathways, such as JAK/STAT signaling pathway. The cisbp_M2205, transcription factor-binding site, was enriched in C1orf106, EVPL, and IF16. Finally, 3,835 lncRNAs and 52 miRNAs significantly correlated with key genes were used to build a ceRNA network. Conclusion The C1orf106, COG8, EVPL, GIMAP6, and IFI6 genes are associated with CD4+ T-cell infiltration. The prediction model constructed based on the 5 key genes may better predict the level of CD4+ T-cell infiltration in damaged muscle and lesional skin of DM. These key genes could be recognized as potential biomarkers and immunotherapeutic targets of DM.
Collapse
Affiliation(s)
- Peng Huang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
- Children’s Brain Development and Brain injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Li Tang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
- Children’s Brain Development and Brain injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Lu Zhang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
- Children’s Brain Development and Brain injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yi Ren
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
- Children’s Brain Development and Brain injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Hong Peng
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
- Children’s Brain Development and Brain injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yangyang Xiao
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
- Children’s Brain Development and Brain injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jie Xu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
- Children’s Brain Development and Brain injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Dingan Mao
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
- Children’s Brain Development and Brain injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Lingjuan Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
- Children’s Brain Development and Brain injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Liqun Liu, ; Lingjuan Liu,
| | - Liqun Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, China
- Children’s Brain Development and Brain injury Research Office, The Second Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Liqun Liu, ; Lingjuan Liu,
| |
Collapse
|
9
|
Pathophysiological Mechanisms and Treatment of Dermatomyositis and Immune Mediated Necrotizing Myopathies: A Focused Review. Int J Mol Sci 2022; 23:ijms23084301. [PMID: 35457124 PMCID: PMC9030619 DOI: 10.3390/ijms23084301] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/31/2022] [Accepted: 04/05/2022] [Indexed: 12/15/2022] Open
Abstract
Idiopathic inflammatory myopathies (IIM), collectively known as myositis, are a composite group of rare autoimmune diseases affecting mostly skeletal muscle, although other organs or tissues may also be involved. The main clinical feature of myositis is subacute, progressive, symmetrical muscle weakness in the proximal arms and legs, whereas subtypes of myositis may also present with extramuscular features, such as skin involvement, arthritis or interstitial lung disease (ILD). Established subgroups of IIM include dermatomyositis (DM), immune-mediated necrotizing myopathy (IMNM), anti-synthetase syndrome (ASyS), overlap myositis (OM) and inclusion body myositis (IBM). Although these subgroups have overlapping clinical features, the widespread variation in the clinical manifestations of IIM suggests different pathophysiological mechanisms. Various components of the immune system are known to be important immunopathogenic pathways in IIM, although the exact pathophysiological mechanisms causing the muscle damage remain unknown. Current treatment, which consists of glucocorticoids and other immunosuppressive or immunomodulating agents, often fails to achieve a sustained beneficial response and is associated with various adverse effects. New therapeutic targets have been identified that may improve outcomes in patients with IIM. A better understanding of the overlapping and diverging pathophysiological mechanisms of the major subgroups of myositis is needed to optimize treatment. The aim of this review is to report on recent advancements regarding DM and IMNM.
Collapse
|
10
|
Pinal-Fernandez I, Greenberg SA. Type I Interferons in Dermatomyositis Myoblasts: Toxic Effect and a Potential Autocrine Loop. Neurology 2022; 98:869-870. [DOI: 10.1212/wnl.0000000000200679] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
11
|
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
|
12
|
Yuzawa R, Koike H, Manabe I, Oishi Y. VDR regulates simulated microgravity-induced atrophy in C2C12 myotubes. Sci Rep 2022; 12:1377. [PMID: 35082348 PMCID: PMC8791983 DOI: 10.1038/s41598-022-05354-0] [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: 10/02/2021] [Accepted: 01/10/2022] [Indexed: 11/30/2022] Open
Abstract
Muscle wasting is a major problem leading to reduced quality of life and higher risks of mortality and various diseases. Muscle atrophy is caused by multiple conditions in which protein degradation exceeds its synthesis, including disuse, malnutrition, and microgravity. While Vitamin D receptor (VDR) is well known to regulate calcium and phosphate metabolism to maintain bone, recent studies have shown that VDR also plays roles in skeletal muscle development and homeostasis. Moreover, its expression is upregulated in muscle undergoing atrophy as well as after muscle injury. Here we show that VDR regulates simulated microgravity-induced atrophy in C2C12 myotubes in vitro. After 8 h of microgravity simulated using 3D-clinorotation, the VDR-binding motif was associated with chromatin regions closed by the simulated microgravity and enhancer regions inactivated by it, which suggests VDR mediates repression of enhancers. In addition, VDR was induced and translocated into the nuclei in response to simulated microgravity. VDR-deficient C2C12 myotubes showed resistance to simulated microgravity-induced atrophy and reduced induction of FBXO32, an atrophy-associated ubiquitin ligase. These results demonstrate that VDR contributes to the regulation of simulated microgravity-induced atrophy at least in part by controlling expression of atrophy-related genes.
Collapse
|
13
|
Abstract
Zika virus (ZIKV) infection became a worldwide concern due to its correlation with the development of microcephaly and other neurological disorders. ZIKV neurotropism is well characterized, but the role of peripheral viral amplification to brain infection remains unknown. Here, we found that ZIKV replicates in human primary skeletal muscle myoblasts, impairing its differentiation into myotubes but not interfering with the integrity of the already-formed muscle fibers. Using mouse models, we showed ZIKV tropism to muscle tissue either during embryogenesis after maternal transmission or when infection occurred after birth. Interestingly, ZIKV replication in the mouse skeletal muscle started immediately after ZIKV inoculation, preceding viral RNA detection in the brain and causing no disruption to the integrity of the blood brain barrier, and remained active for more than 2 weeks, whereas replication in the spleen and liver were not sustained over time. In addition, ZIKV infection of the skeletal muscle induces necrotic lesions, inflammation, and fiber atrophy. We also found a reduction in the expression of regulatory myogenic factors that are essential for muscle repair after injury. Taken together, our results indicate that the skeletal muscle is an early site of viral amplification and lesion that may result in late consequences in muscle development after ZIKV infection. IMPORTANCE Zika Virus (ZIKV) neurotropism and its deleterious effects on central nervous system have been well characterized. However, investigations of the initial replication sites for the establishment of infection and viral spread to neural tissues remain underexplored. A complete description of the range of ZIKV-induced lesions and others factors that can influence the severity of the disease is necessary to prevent ZIKV’s deleterious effects. ZIKV has been shown to access the central nervous system without significantly affecting blood-brain barrier permeability. Here, we demonstrated that skeletal muscle is an earlier site of ZIKV replication, contributing to the increase of peripheral ZIKV load. ZIKV replication in muscle promotes necrotic lesions and inflammation and also impairs myogenesis. Overall, our findings showed that skeletal muscle is involved in pathogenesis and opens new fields in the investigation of the long-term consequences of early infection.
Collapse
|
14
|
Kurosaka M, Ogura Y, Sato S, Kohda K, Funabashi T. Transcription factor signal transducer and activator of transcription 6 (STAT6) is an inhibitory factor for adult myogenesis. Skelet Muscle 2021; 11:14. [PMID: 34051858 PMCID: PMC8164270 DOI: 10.1186/s13395-021-00271-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 05/18/2021] [Indexed: 01/25/2023] Open
Abstract
Background The signal transducer and activator of transcription 6 (STAT6) transcription factor plays a vitally important role in immune cells, where it is activated mainly by interleukin-4 (IL-4). Because IL-4 is an essential cytokine for myotube formation, STAT6 might also be involved in myogenesis as part of IL-4 signaling. This study was conducted to elucidate the role of STAT6 in adult myogenesis in vitro and in vivo. Methods Myoblasts were isolated from male mice and were differentiated on a culture dish to evaluate the change in STAT6 during myotube formation. Then, the effects of STAT6 overexpression and inhibition on proliferation, differentiation, and fusion in those cells were studied. Additionally, to elucidate the myogenic role of STAT6 in vivo, muscle regeneration after injury was evaluated in STAT6 knockout mice. Results IL-4 can increase STAT6 phosphorylation, but STAT6 phosphorylation decreased during myotube formation in culture. STAT6 overexpression decreased, but STAT6 knockdown increased the differentiation index and the fusion index. Results indicate that STAT6 inhibited myogenin protein expression. Results of in vivo experiments show that STAT6 knockout mice exhibited better regeneration than wild-type mice 5 days after cardiotoxin-induced injury. It is particularly interesting that results obtained using cells from STAT6 knockout mice suggest that this STAT6 inhibitory action for myogenesis was not mediated by IL-4 but might instead be associated with p38 mitogen-activated protein kinase phosphorylation. However, STAT6 was not involved in the proliferation of myogenic cells in vitro and in vivo. Conclusion Results suggest that STAT6 functions as an inhibitor of adult myogenesis. Moreover, results suggest that the IL-4-STAT6 signaling axis is unlikely to be responsible for myotube formation. Supplementary Information The online version contains supplementary material available at 10.1186/s13395-021-00271-8.
Collapse
Affiliation(s)
- Mitsutoshi Kurosaka
- Department of Physiology, St. Marianna University School of Medicine, Kawasaki, Kanagawa, 216-8511, Japan
| | - Yuji Ogura
- Department of Physiology, St. Marianna University School of Medicine, Kawasaki, Kanagawa, 216-8511, Japan.
| | - Shuichi Sato
- School of Kinesiology, The University of Louisiana at Lafayette, Lafayette, LA, USA.,New Iberia Research Center, The University of Louisiana at Lafayette, New Iberia, LA, USA
| | - Kazuhisa Kohda
- Department of Physiology, St. Marianna University School of Medicine, Kawasaki, Kanagawa, 216-8511, Japan
| | - Toshiya Funabashi
- Department of Physiology, St. Marianna University School of Medicine, Kawasaki, Kanagawa, 216-8511, Japan
| |
Collapse
|
15
|
Yin R, Wang G, Zhang L, Li T, Liu S. Dermatomyositis: immunological landscape, biomarkers, and potential candidate drugs. Clin Rheumatol 2021; 40:2301-2310. [PMID: 33389314 DOI: 10.1007/s10067-020-05568-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/14/2020] [Accepted: 12/22/2020] [Indexed: 10/22/2022]
Abstract
INTRODUCTION Dermatomyositis (DM) is a rare inflammatory disease characterized by the invasion of the skin and muscles. Environmental, genetic, and immunological factors contribute to disease pathology. To date, no bioinformatics studies have been conducted on the potential pathogenic genes and immune cell infiltration in DM. Therefore, we aimed to identify differentially expressed genes (DEGs) and immune cells, as well as potential pathogenic genes and immune characteristics, which may be useful for the diagnosis and treatment of DM. METHOD GSE1551, GSE5370, GSE39454, and GSE48280 from Gene Expression Omnibus were included in our study. Limma, ClusterProfiler, and Kyoto Encyclopedia of Genes and Genomes were used to identify DEGs, Gene Ontology (GO), and perform pathway analyses, respectively. Cytoscape was used to construct the protein-protein interaction (PPI) network. Small-molecule drugs were identified using a connectivity map (CMap), and the TIMER database was used to identify infiltrating cells. RESULTS DEG analysis identified 12 downregulated and 163 upregulated genes. GO analysis showed that DEGs were enriched in immune-related pathways. Ten hub genes were identified from the PPI network. Additionally, CMap analysis showed that caffeic acid, sulfaphenazole, molindone, tiabendazole, and bacitracin were potential small-molecule drugs with therapeutic significance. We identified eight immune cells with differential infiltration in patients with DM and controls. Finally, we constructed a powerful diagnostic model based on memory B cells, M1, and M2 macrophages. CONCLUSIONS This study explored the potential molecular mechanism and immunological landscape of DM and may guide future research and treatment of DM. KEY POINTS • We explored the molecular mechanism and immunological landscape of dermatomyositis. • GO analysis showed that DEGs were enriched in immune-related pathways. • We predicted small-molecular drugs with potential therapeutic significance based on bioanalytical techniques. • We identified six immune cells with differential infiltration in patients with DM and controls.
Collapse
Affiliation(s)
- Ruxue Yin
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450052, Henan, China
| | - Gangjian Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450052, China
| | - Lei Zhang
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450052, Henan, China
| | - Tianfang Li
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450052, Henan, China.
| | - Shengyun Liu
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
16
|
Tsoi LC, Gharaee-Kermani M, Berthier CC, Nault T, Hile GA, Estadt SN, Patrick MT, Wasikowski R, Billi AC, Lowe L, Reed TJ, Gudjonsson JE, Kahlenberg JM. IL18-containing 5-gene signature distinguishes histologically identical dermatomyositis and lupus erythematosus skin lesions. JCI Insight 2020; 5:139558. [PMID: 32644977 PMCID: PMC7455118 DOI: 10.1172/jci.insight.139558] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/02/2020] [Indexed: 12/26/2022] Open
Abstract
Skin lesions in dermatomyositis (DM) are common, are frequently refractory, and have prognostic significance. Histologically, DM lesions appear similar to cutaneous lupus erythematosus (CLE) lesions and frequently cannot be differentiated. We thus compared the transcriptional profile of DM biopsies with CLE lesions to identify unique features. Type I IFN signaling, including IFN-κ upregulation, was a common pathway in both DM and CLE; however, CLE also exhibited other inflammatory pathways. Notably, DM lesions could be distinguished from CLE by a 5-gene biomarker panel that included IL18 upregulation. Using single-cell RNA-sequencing, we further identified keratinocytes as the main source of increased IL-18 in DM skin. This study identifies a potentially novel molecular signature, with significant clinical implications for differentiating DM from CLE lesions, and highlights the potential role for IL-18 in the pathophysiology of DM skin disease. IL-18 distinguishes dermatomyositis skin inflammation from cutaneous lupus erythematosus lesions.
Collapse
Affiliation(s)
- Lam C Tsoi
- Department of Dermatology and.,Department of Computational Medicine & Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | - Tori Nault
- Division of General Medicine, Department of Internal Medicine
| | | | - Shannon N Estadt
- Division of Rheumatology, Department of Internal Medicine.,Program in Immunology, and
| | | | | | | | - Lori Lowe
- Department of Dermatology and.,Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Tamra J Reed
- Division of Rheumatology, Department of Internal Medicine
| | | | | |
Collapse
|
17
|
Khoo T, Limaye V. Biologic therapy in the idiopathic inflammatory myopathies. Rheumatol Int 2019; 40:191-205. [PMID: 31680207 DOI: 10.1007/s00296-019-04467-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 10/24/2019] [Indexed: 11/29/2022]
Abstract
The idiopathic inflammatory myopathies (IIM) are a group of autoimmune diseases resulting from inflammation of muscle and manifesting as weakness, though a range of extra-muscular manifestations are observed. These are often correlated closely with disease subtype and the presence of myositis-specific/myositis-associated antibodies. IIM are notoriously difficult to treat and often refractory to glucocorticoid therapy and synthetic immunosuppressants. Both the innate and adaptive immune systems are implicated in the pathogenesis of IIM. A growing understanding of the key cytokines as well as the cell-mediated and antibody effectors of disease has identified multiple potential targets for biologic therapy. The most widely used of these is B-cell depletion via rituximab though the tumour necrosis factor inhibitors and other biologic therapies used in diseases such as rheumatoid arthritis, systemic lupus erythematosus and multiple sclerosis have also been trialled. This review summarises the literature thus far on biologic therapy in IIM, highlighting both the significant trials that influence current treatment regimens and also the continuing need for further research to inform more effective therapies.
Collapse
Affiliation(s)
- Thomas Khoo
- Central Adelaide Local Health Network, Adelaide, Australia
| | - Vidya Limaye
- Rheumatology Unit, Royal Adelaide Hospital, Adelaide, Australia. .,Discipline of Medicine, School of Medicine, University of Adelaide, Adelaide, Australia.
| |
Collapse
|
18
|
Mammen AL, Allenbach Y, Stenzel W, Benveniste O. 239th ENMC International Workshop: Classification of dermatomyositis, Amsterdam, the Netherlands, 14-16 December 2018. Neuromuscul Disord 2019; 30:70-92. [PMID: 31791867 DOI: 10.1016/j.nmd.2019.10.005] [Citation(s) in RCA: 156] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 10/21/2019] [Indexed: 12/29/2022]
Affiliation(s)
- Andrew L Mammen
- Muscle Disease Unit, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, 50 South Drive, Building 50, Room 1146, MD 20892, United States.
| | - Yves Allenbach
- Department of Internal Medicine and Clinical Immunology, Pitié Salpetrière Hospital, AP-HP Sorbonne University, Paris, France
| | - Werner Stenzel
- Department of Neuropathology, Charité-Universitatsmedizin, Berlin, Germany
| | - Olivier Benveniste
- Department of Internal Medicine and Clinical Immunology, Pitié Salpetrière Hospital, AP-HP Sorbonne University, Paris, France
| | | |
Collapse
|
19
|
Ladislau L, Suárez-Calvet X, Toquet S, Landon-Cardinal O, Amelin D, Depp M, Rodero MP, Hathazi D, Duffy D, Bondet V, Preusse C, Bienvenu B, Rozenberg F, Roos A, Benjamim CF, Gallardo E, Illa I, Mouly V, Stenzel W, Butler-Browne G, Benveniste O, Allenbach Y. JAK inhibitor improves type I interferon induced damage: proof of concept in dermatomyositis. Brain 2018; 141:1609-1621. [DOI: 10.1093/brain/awy105] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 02/17/2018] [Indexed: 12/17/2022] Open
Affiliation(s)
- Leandro Ladislau
- Sorbonne Université, INSERM, Association Institut de Myologie, Center of Research in Myology, UMRS 974, AP-HP, Department of Internal Medicine and Clinical Immunology, DHU I2B, Pitié-Salpêtrière Hospital, F-75013, Paris, France
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Xavier Suárez-Calvet
- Sorbonne Université, INSERM, Association Institut de Myologie, Center of Research in Myology, UMRS 974, AP-HP, Department of Internal Medicine and Clinical Immunology, DHU I2B, Pitié-Salpêtrière Hospital, F-75013, Paris, France
- Neuromuscular Diseases Unit, Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona and Institut de Recerca Sant Pau, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Ségolène Toquet
- Sorbonne Université, INSERM, Association Institut de Myologie, Center of Research in Myology, UMRS 974, AP-HP, Department of Internal Medicine and Clinical Immunology, DHU I2B, Pitié-Salpêtrière Hospital, F-75013, Paris, France
| | - Océane Landon-Cardinal
- Sorbonne Université, INSERM, Association Institut de Myologie, Center of Research in Myology, UMRS 974, AP-HP, Department of Internal Medicine and Clinical Immunology, DHU I2B, Pitié-Salpêtrière Hospital, F-75013, Paris, France
| | - Damien Amelin
- Sorbonne Université, INSERM, Association Institut de Myologie, Center of Research in Myology, UMRS 974, AP-HP, Department of Internal Medicine and Clinical Immunology, DHU I2B, Pitié-Salpêtrière Hospital, F-75013, Paris, France
| | - Marine Depp
- Laboratory of Neurogenetics and Neuroinflammation, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1163 and Université Paris Descartes, Université Sorbonne Paris Cité, Institut Imagine, 75015 Paris, France
| | - Mathieu P Rodero
- Laboratory of Neurogenetics and Neuroinflammation, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR1163 and Université Paris Descartes, Université Sorbonne Paris Cité, Institut Imagine, 75015 Paris, France
| | - Denisa Hathazi
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V, Biomedical Research Department, Tissue Omics group, Otto-Hahn-Str. 6b, 44227, Dortmund, Germany
| | - Darragh Duffy
- INSERM UMR 1223 and Laboratory of Dendritic Cell Immunobiology, Institut Pasteur, Paris, France
| | - Vincent Bondet
- INSERM UMR 1223 and Laboratory of Dendritic Cell Immunobiology, Institut Pasteur, Paris, France
| | - Corinna Preusse
- Department of Neuropathology, Charité University, Berlin, Germany
| | - Boris Bienvenu
- Department of Internal Medicine, Saint Joseph Hospital, Marseille, France
| | - Flore Rozenberg
- Departement de Virologie, Hôpital Cochin, Paris Descartes Universités, Paris, France
| | - Andreas Roos
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V, Biomedical Research Department, Tissue Omics group, Otto-Hahn-Str. 6b, 44227, Dortmund, Germany
- Institute of Genetic Medicine, John Walton Muscular Dystrophy Research Centre, International Centre for Life, Central Parkway, Newcastle upon Tyne, England, UK
| | - Claudia F Benjamim
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Eduard Gallardo
- Neuromuscular Diseases Unit, Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona and Institut de Recerca Sant Pau, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Isabel Illa
- Neuromuscular Diseases Unit, Neurology, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona and Institut de Recerca Sant Pau, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Vincent Mouly
- Sorbonne Université, INSERM, Association Institut de Myologie, Center of Research in Myology, UMRS 974, AP-HP, Department of Internal Medicine and Clinical Immunology, DHU I2B, Pitié-Salpêtrière Hospital, F-75013, Paris, France
| | - Werner Stenzel
- Department of Neuropathology, Charité University, Berlin, Germany
| | - Gillian Butler-Browne
- Sorbonne Université, INSERM, Association Institut de Myologie, Center of Research in Myology, UMRS 974, AP-HP, Department of Internal Medicine and Clinical Immunology, DHU I2B, Pitié-Salpêtrière Hospital, F-75013, Paris, France
| | - Olivier Benveniste
- Sorbonne Université, INSERM, Association Institut de Myologie, Center of Research in Myology, UMRS 974, AP-HP, Department of Internal Medicine and Clinical Immunology, DHU I2B, Pitié-Salpêtrière Hospital, F-75013, Paris, France
| | - Yves Allenbach
- Sorbonne Université, INSERM, Association Institut de Myologie, Center of Research in Myology, UMRS 974, AP-HP, Department of Internal Medicine and Clinical Immunology, DHU I2B, Pitié-Salpêtrière Hospital, F-75013, Paris, France
| |
Collapse
|
20
|
Dimitrova E, Caromile LA, Laubenbacher R, Shapiro LH. The innate immune response to ischemic injury: a multiscale modeling perspective. BMC SYSTEMS BIOLOGY 2018; 12:50. [PMID: 29631571 PMCID: PMC5891907 DOI: 10.1186/s12918-018-0580-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 03/28/2018] [Indexed: 12/13/2022]
Abstract
Background Cell death as a result of ischemic injury triggers powerful mechanisms regulated by germline-encoded Pattern Recognition Receptors (PRRs) with shared specificity that recognize invading pathogens and endogenous ligands released from dying cells, and as such are essential to human health. Alternatively, dysregulation of these mechanisms contributes to extreme inflammation, deleterious tissue damage and impaired healing in various diseases. The Toll-like receptors (TLRs) are a prototypical family of PRRs that may be powerful anti-inflammatory targets if agents can be designed that antagonize their harmful effects while preserving host defense functions. This requires an understanding of the complex interactions and consequences of targeting the TLR-mediated pathways as well as technologies to analyze and interpret these, which will then allow the simulation of perturbations targeting specific pathway components, predict potential outcomes and identify safe and effective therapeutic targets. Results We constructed a multiscale mathematical model that spans the tissue and intracellular scales, and captures the consequences of targeting various regulatory components of injury-induced TLR4 signal transduction on potential pro-inflammatory or pro-healing outcomes. We applied known interactions to simulate how inactivation of specific regulatory nodes affects dynamics in the context of injury and to predict phenotypes of potential therapeutic interventions. We propose rules to link model behavior to qualitative estimates of pro-inflammatory signal activation, macrophage infiltration, production of reactive oxygen species and resolution. We tested the validity of the model by assessing its ability to reproduce published data not used in its construction. Conclusions These studies will enable us to form a conceptual framework focusing on TLR4-mediated ischemic repair to assess potential molecular targets that can be utilized therapeutically to improve efficacy and safety in treating ischemic/inflammatory injury.
Collapse
Affiliation(s)
- Elena Dimitrova
- Department of Mathematical Sciences, Clemson University, Clemson, SC, USA
| | - Leslie A Caromile
- Center for Vascular Biology, Department of Cell Biology, University of Connecticut School of Medicine, Farmington, 06030, CT, USA
| | - Reinhard Laubenbacher
- Center for Quantitative Medicine, Department of Cell Biology, University of Connecticut School of Medicine, Farmington, CT, USA. .,Jackson Laboratory for Genomic Medicine, Farmington, CT, USA.
| | - Linda H Shapiro
- Center for Vascular Biology, Department of Cell Biology, University of Connecticut School of Medicine, Farmington, 06030, CT, USA.
| |
Collapse
|
21
|
Gitiaux C, Latroche C, Weiss‐Gayet M, Rodero MP, Duffy D, Bader‐Meunier B, Glorion C, Nusbaum P, Bodemer C, Mouchiroud G, Chelly J, Germain S, Desguerre I, Chazaud B. Myogenic Progenitor Cells Exhibit Type I Interferon–Driven Proangiogenic Properties and Molecular Signature During Juvenile Dermatomyositis. Arthritis Rheumatol 2017; 70:134-145. [DOI: 10.1002/art.40328] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 09/15/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Cyril Gitiaux
- Département de Neurophysiologie Clinique Hôpital Necker‐Enfants Malades, AP‐HP, Centre de Référence pour les Maladies Neuromusculaires, Garches‐Necker‐Mondor‐Hendaye, AP‐HP, FILNEMUS, and Institut Cochin, INSERM U1016, CNRS UMR 8104, Université Paris Descartes Paris France
| | - Claire Latroche
- Institut Cochin INSERM U1016, CNRS UMR 8104, Université Paris Descartes Paris France
| | - Michèle Weiss‐Gayet
- Institut NeuroMyoGène Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217 Villeurbanne France
| | - Mathieu P. Rodero
- Laboratory of Neurogenetics and Neuroinflammation INSERM UMR1163, Institut Imagine Paris France
| | - Darragh Duffy
- Immunobiology of Dendritic Cells INSERM U1223, Institut Pasteur Paris France
| | - Brigitte Bader‐Meunier
- Département de Rhumatologie Pédiatrique Hôpital Necker Enfants Malades, AP‐HP Paris France
| | - Christophe Glorion
- Département de Chirurgie Orthopédique Hôpital Necker Enfants Malades, AP‐HP Paris France
| | - Patrick Nusbaum
- Service de Biochimie et de Génétique Moléculaire Hôpital Cochin, AP‐HP Paris France
| | - Christine Bodemer
- Département de Dermatologie Hôpital Necker Enfants Malades, AP‐HP Paris France
| | - Guy Mouchiroud
- Institut NeuroMyoGène Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217 Villeurbanne France
| | - Jamel Chelly
- Institut Cochin INSERM U1016, CNRS UMR 8104, Université Paris Descartes Paris France
| | - Stéphane Germain
- Center for Interdisciplinary Research in Biology Collège de France, INSERM, CNRS, PSL Research University Paris France
| | - Isabelle Desguerre
- Centre de Référence pour les Maladies Neuromusculaires, Garches‐Necker‐Mondor‐Hendaye, AP‐HP, FILNEMUS Paris France
| | - Bénédicte Chazaud
- Institut NeuroMyoGène Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217 Villeurbanne France
| |
Collapse
|
22
|
Dysregulated innate immune function in the aetiopathogenesis of idiopathic inflammatory myopathies. Autoimmun Rev 2016; 16:87-95. [PMID: 27666811 DOI: 10.1016/j.autrev.2016.09.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 09/08/2016] [Indexed: 12/20/2022]
Abstract
The idiopathic inflammatory myopathies (IIMs) are a heterogeneous group of systemic muscle conditions that are believed to be autoimmune in nature. They have distinct pathological features, but the aetiopathogenesis of each subtype remains largely unknown. Recently, there has been increased interest in the complex role the innate immune system plays in initiating and perpetuating these conditions, and how this may differ between subtypes. This article summarises the traditional paradigms of IIM pathogenesis and reviews the accumulating evidence for disturbances in innate immune processes in these rare, but debilitating chronic conditions.
Collapse
|
23
|
Lahoria R, Selcen D, Engel AG. Microvascular alterations and the role of complement in dermatomyositis. Brain 2016; 139:1891-903. [DOI: 10.1093/brain/aww122] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 04/07/2016] [Indexed: 12/21/2022] Open
|
24
|
|
25
|
Lu X, Peng Q, Wang G. Discovery of new biomarkers of idiopathic inflammatory myopathy. Clin Chim Acta 2015; 444:117-25. [DOI: 10.1016/j.cca.2015.02.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 02/02/2015] [Accepted: 02/02/2015] [Indexed: 01/19/2023]
|