1
|
Huang LH. Comment on: Where are we now in biologic drugs for myositis? Rheumatology (Oxford) 2024; 63:e303-e304. [PMID: 39024051 DOI: 10.1093/rheumatology/keae363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/20/2024] [Accepted: 06/08/2024] [Indexed: 07/20/2024] Open
Affiliation(s)
- Li-Heng Huang
- Department of Allergy, Immunology and Rheumatology, National Taiwan University Hospital, Taipei City, Taiwan
| |
Collapse
|
2
|
Wu Y, Deshpande A, Geraci N, Budde P, Sellers V, Velisetty P, Sun CC, 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 2024. [PMID: 39279150 DOI: 10.1002/art.42989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [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 Serono, Billerica, Massachusetts
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Zhang G, Zhang L, Zhao D, Liu X, Li W, Yan C, Dai T. Protein kinase R is highly expressed in dermatomyositis and promotes interferon-beta-induced muscle damage. Ann Rheum Dis 2024:ard-2024-226057. [PMID: 39237131 DOI: 10.1136/ard-2024-226057] [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: 05/02/2024] [Accepted: 08/23/2024] [Indexed: 09/07/2024]
Abstract
OBJECTIVES Dermatomyositis (DM) has been consistently linked to the type I interferon (IFN-I) pathway. However, the precise pathogenesis remains incompletely elucidated. We aimed to explore potential molecular mechanisms and identify promising therapeutic targets in DM. METHODS We employed bioinformatics analysis to investigate molecular signatures, aiming to shed light on the pathogenesis of DM. The expression of protein kinase R (PKR) in DM muscle tissues was determined by real-time quantitative PCR, western blot and immunohistochemistry (IHC) analysis. We then assessed the sensitivity and specificity of sarcoplasmic PKR expression by IHC in a consecutive DM cohort and other diseases in this retrospective study. Furthermore, IFN-β was used to stimulate myoblasts and myotubes, and the relationship between PKR and IFN-β-induced pathogenic molecules was investigated in vitro. RESULTS Bioinformatics analysis indicated two primary pathological processes: viral infection and the IFN-I signalling pathway. We subsequently verified that PKR was notably expressed in the cytoplasm of myofibers in DM patients. The sensitivity and specificity of sarcoplasmic PKR expression in DM were 84.6% and 97.6%, respectively. In vitro studies revealed that IFN-β upregulates the expression of PKR, along with several molecules associated with DM muscle damage. Conversely, inhibiting PKR has been shown to downregulate IFN-β-induced pathogenic molecules in both myoblasts and myotubes. CONCLUSIONS We observed that PKR exhibits specific expression in the cytoplasm of DM muscle and inhibiting PKR ameliorates IFN-β-induced muscle damage in vitro. These findings provide insights into the diagnostic and therapeutic roles of PKR in DM.
Collapse
Affiliation(s)
- Guoyong Zhang
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
- University of Health and Rehabilitation Sciences, Qingdao, Shandong, People's Republic of China
| | - Lining Zhang
- Department of Neurology, Research Institute of Neuromuscular and Neurodegenerative Diseases,Qilu Hospital of Shandong University, Shandong Provincial Key Laboratory of Mitochondrial Medicine and Rare Diseases, Jinan, Shandong, People's Republic of China
- Department of Rheumatology, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China
| | - Dandan Zhao
- Department of Neurology, Research Institute of Neuromuscular and Neurodegenerative Diseases,Qilu Hospital of Shandong University, Shandong Provincial Key Laboratory of Mitochondrial Medicine and Rare Diseases, Jinan, Shandong, People's Republic of China
| | - Xiaoyu Liu
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People's Republic of China
| | - Wei Li
- Department of Neurology, Research Institute of Neuromuscular and Neurodegenerative Diseases,Qilu Hospital of Shandong University, Shandong Provincial Key Laboratory of Mitochondrial Medicine and Rare Diseases, Jinan, Shandong, People's Republic of China
| | - Chuanzhu Yan
- Department of Neurology, Research Institute of Neuromuscular and Neurodegenerative Diseases,Qilu Hospital of Shandong University, Shandong Provincial Key Laboratory of Mitochondrial Medicine and Rare Diseases, Jinan, Shandong, People's Republic of China
- Qingdao Municipal Key Laboratory of Mitochondria Medicine, Qingdao, Shandong, People's Republic of China
| | - Tingjun Dai
- Department of Neurology, Research Institute of Neuromuscular and Neurodegenerative Diseases,Qilu Hospital of Shandong University, Shandong Provincial Key Laboratory of Mitochondrial Medicine and Rare Diseases, Jinan, Shandong, People's Republic of China
| |
Collapse
|
4
|
Veldkamp SR, van Wijk F, van Royen-Kerkhof A, Jansen MH. Personalised medicine in juvenile dermatomyositis: From novel insights in disease mechanisms to changes in clinical practice. Best Pract Res Clin Rheumatol 2024; 38:101976. [PMID: 39174374 DOI: 10.1016/j.berh.2024.101976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/12/2024] [Accepted: 07/15/2024] [Indexed: 08/24/2024]
Abstract
Juvenile dermatomyositis is characterized by childhood-onset chronic inflammation of the muscles and skin, with potential involvement of other organs. Patients are at risk for long-term morbidity due to insufficient disease control and steroid-related toxicity. Personalised treatment is challenged by a lack of validated tools that can reliably predict treatment response and monitor ongoing (subclinical) inflammation, and by a lack of evidence regarding the best choice of medication for individual patients. A better understanding of the involved disease mechanisms could reveal potential biomarkers and novel therapeutic targets. In this review, we highlight the most relevant immune and non-immune mechanisms, elucidating the effects of interferon overexpression on tissue alongside the interplay between the interferon signature, mitochondrial function, and immune cells. We review mechanism-based biomarkers that are promising for clinical implementation, and the latest advances in targeted therapy development. Finally, we discuss key steps needed for translating these discoveries into clinical practice.
Collapse
Affiliation(s)
- Saskia R Veldkamp
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Femke van Wijk
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Annet van Royen-Kerkhof
- Department of Pediatric Immunology and Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Marc Ha Jansen
- Department of Pediatric Immunology and Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
5
|
Nishimura A, Nelke C, Huber M, Mensch A, Roth A, Oberwittler C, Zimmerlein B, Krämer HH, Neuen-Jacob E, Stenzel W, Müller-Ladner U, Ruck T, Schänzer A. Differentiating idiopathic inflammatory myopathies by automated morphometric analysis of MHC-1, MHC-2 and ICAM-1 in muscle tissue. Neuropathol Appl Neurobiol 2024; 50:e12998. [PMID: 39030945 DOI: 10.1111/nan.12998] [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: 04/26/2024] [Revised: 06/25/2024] [Accepted: 07/01/2024] [Indexed: 07/22/2024]
Abstract
AIMS Diagnosis of idiopathic inflammatory myopathies (IIM) is based on morphological characteristics and the evaluation of disease-related proteins. However, although broadly applied, substantial bias is imposed by the respective methods, observers and individual staining approaches. We aimed to quantify the protein levels of major histocompatibility complex (MHC)-1, (MHC)-2 and intercellular adhesion molecule (ICAM)-1 using an automated morphometric method to mitigate bias. METHODS Double immunofluorescence staining was performed on whole muscle sections to study differences in protein expression in myofibre and endomysial vessels. We analysed all IIM subtypes including dermatomyositis (DM), anti-synthetase syndrome (ASyS), inclusion body myositis (IBM), immune-mediated-necrotising myopathy (IMNM), dysferlinopathy (DYSF), SARS-CoV-2 infection and vaccination-associated myopathy. Biopsies with neurogenic atrophy (NA) and normal morphology served as controls. Bulk RNA-Sequencing (RNA-Seq) was performed on a subset of samples. RESULTS Our study highlights the significance of MHC-1, MHC-2 and ICAM-1 in diagnosing IIM subtypes and reveals distinct immunological profiles. RNASeq confirmed the precision of our method and identified specific gene pathways in the disease subtypes. Notably, ASyS, DM and SARS-CoV-2-associated myopathy showed increased ICAM-1 expression in the endomysial capillaries, indicating ICAM-1-associated vascular activation in these conditions. In addition, ICAM-1 showed high discrimination between different subgroups with high sensitivity and specificity. CONCLUSIONS Automated morphometric analysis provides precise quantitative data on immune-associated proteins that can be integrated into our pathophysiological understanding of IIM. Further, ICAM-1 holds diagnostic value for the detection of IIM pathology.
Collapse
Affiliation(s)
- Anna Nishimura
- Institute of Neuropathology, Justus-Liebig University Giessen, Germany
| | - Christopher Nelke
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Melanie Huber
- Department of Rheumatology and Clinical Immunology, Campus Kerckhoff, Justus-Liebig-University, Giessen, Germany
| | - Alexander Mensch
- Department of Neurology, University Medicine Halle, Halle (Saale), Germany
| | - Angela Roth
- Institute of Neuropathology, Justus-Liebig University Giessen, Germany
| | | | | | - Heidrun H Krämer
- Department of Neurology, Justus-Liebig-University, Giessen, Germany
- Translational Neuroscience Network Giessen (TNNG), Justus Liebig University Giessen, Giessen, Germany
| | - Eva Neuen-Jacob
- Institute of Neuropathology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Werner Stenzel
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ulf Müller-Ladner
- Department of Rheumatology and Clinical Immunology, Campus Kerckhoff, Justus-Liebig-University, Giessen, Germany
| | - Tobias Ruck
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Anne Schänzer
- Institute of Neuropathology, Justus-Liebig University Giessen, Germany
- Translational Neuroscience Network Giessen (TNNG), Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
6
|
Srinivasa Murthy M, Haikal A. A Novel Treatment for Skin Manifestations in Dermatomyositis: A Case Report. Cureus 2024; 16:e66704. [PMID: 39262502 PMCID: PMC11390155 DOI: 10.7759/cureus.66704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2024] [Indexed: 09/13/2024] Open
Abstract
We present a case of refractory cutaneous dermatomyositis (DM) in a 51-year-old Hispanic female which failed multiple treatments but found symptomatic relief with anifrolumab. Anifrolumab was the only treatment that was associated with significant improvement in the rash and pruritis of the patient and lowered her corticosteroid needs. To our knowledge, this is the only second case report that has shown success in treating refractory cutaneous symptoms of DM with anifrolumab after failing standard and multiple combinations of therapies. Anifrolumab is a new first-in-class human monoclonal antibody, which inhibits type 1 interferon receptor (IFN-1) and is used to treat systemic lupus erythematosus (SLE). It is FDA-approved for non-renal manifestations of SLE. This IFN pathway seems to be also active in patients with DM. The presence of IFN-1 and IFN-2 has been reported in muscle biopsies of patients with inflammatory myopathies. Moreover, the IFN activation signature is present in the muscle, blood, and skin of patients with DM. IFN-1 has been assumed to activate toll-like receptors which activate the dendritic cells leading to the secretion of cytokines and chemokines. This potential pathophysiological role of IFN in DM may explain the symptom improvement experienced by our patient after starting anifrolumab treatment. Anifrolumab has additionally been shown to have a good safety profile when used to treat patients with SLE with up to three years of treatment on background conventional disease-modifying antirheumatic drug (DMARD) therapies. In conclusion, SLE and DM share similarities in their pathophysiology and cutaneous disease involvement and can be differentiated clinically. Skin manifestations of DM can persist despite combinations of therapies even when weakness resolves. With this case report, we aim to highlight the possibility of utilizing anifrolumab for treating DM skin manifestations, especially in refractory cases. More research is needed to guide where anifrolumab stands in the therapeutic algorithm for DM. It is unknown whether it treats the myositis component, DM-related arthritis, or coexistent rheumatoid arthritis.
Collapse
Affiliation(s)
| | - Ammar Haikal
- Rheumatology, Hackensack University Medical Center, Hackensack, USA
| |
Collapse
|
7
|
Kannappan R, Kumar R, Cichelli K, Brent LH. A Review of Myositis-Associated Interstitial Lung Disease. J Clin Med 2024; 13:4055. [PMID: 39064092 PMCID: PMC11278012 DOI: 10.3390/jcm13144055] [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: 05/22/2024] [Revised: 06/27/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
There is a well-established relationship between different subsets of idiopathic inflammatory myopathies (IIMs, myositis) and interstitial lung disease (ILD), with lung complications sometimes presenting prior to myopathic manifestations. The subtypes of myositis include those that are strongly associated with ILD, such as polymyositis (PM) and dermatomyositis (DM). Research has shown that in certain patients, these can then be further divided into subtypes using myositis-specific antibodies (MSAs), which are specific for myositis, and myositis-associated antibodies (MAAs), which can be found in myositis in overlap syndromes with other connective tissue diseases (CTDs). Notably, certain MSAs and MAAs are associated with ILD in patients with myositis. The clinical presentations of ILD in patients with myositis can vary widely and can be insidious in onset and difficult to diagnose. As ILD can progress rapidly in some cases, it is essential that clinicians are able to identify and diagnose ILD in patients with myositis. For this reason, the aim of this review is to highlight the clinical features, diagnostic criteria, important histopathologic, laboratory, and radiographic features, and treatment modalities for those patients with myositis-associated ILD.
Collapse
Affiliation(s)
- Renuka Kannappan
- Department of Internal Medicine, Temple University Hospital, Philadelphia, PA 19140, USA
| | - Raagni Kumar
- Department of Internal Medicine, Temple University Hospital, Philadelphia, PA 19140, USA
| | - Kimberly Cichelli
- Section of Rheumatology, Temple University Hospital, Philadelphia, PA 19140, USA
| | - Lawrence H. Brent
- Section of Rheumatology, Temple University Hospital, Philadelphia, PA 19140, USA
| |
Collapse
|
8
|
Huang L, Li X, Zhou W, Zhu H, Lao Y, Huang X, Deng Z, Tang Y, Wang J. The Clinical Value of the Neutrophil-to-Lymphocyte Ratio, the C-Reactive Protein-to-Albumin Ratio, the Systemic Inflammatory Index, and the Systemic Inflammatory Response Index in Patients with the Anti-Synthetase Syndrome. J Inflamm Res 2024; 17:3617-3628. [PMID: 38855168 PMCID: PMC11162194 DOI: 10.2147/jir.s460610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 05/22/2024] [Indexed: 06/11/2024] Open
Abstract
Objective There are no studies examining the role of the neutrophil-to-lymphocyte ratio (NLR), the C-reactive protein-to-albumin ratio (CAR), the systemic inflammatory index (SII), and the systemic inflammatory response index (SIRI) in anti-synthetase syndrome (ASS). We aim to compare NLR, CAR, SII, and SIRI in ASS and dermatomyositis/polymyositis (DM/PM), as well as to examine potential correlations between NLR, CAR, SII, and SIRI and clinical features and laboratory parameters in ASS. Methods Retrospective collection of data from 111 patients with ASS and 175 patients with DM/PM. A Spearman rank correlation analysis was utilized to analyze the correlation between NLR, CAR, SII, and SIRI and inflammatory indexes. Receiver operating characteristic (ROC) curves were used to assess the diagnostic value. Univariate logistic regression analysis was performed to assess risk factors for interstitial lung disease (ILD). Results Compared with DM/PM, NLR, CAR, SII, and SIRI were significantly greater in ASS patients (p < 0.05). NLR, CAR, SII, and SIRI were correlated with albumin, lactic dehydrogenase (LDH), C-reactive protein (CRP), ferritin, white blood cell (WBC), platelets, and myositis disease activity assessment visual analog scales (MYOACT) score (p < 0.05). The ROC curves analysis showed that NLR, SII, and SIRI were all highly predictive of the occurrence of ASS. Comparisons based on clinical characteristics showed elevated levels of NLR, CAR, SII, and SIRI in ASS patients with ILD, fever, and infection (p < 0.05). Univariate logistic regression analysis revealed that NLR, CAR, and SII were significant risk factors for ASS-ILD (p < 0.05). Conclusion The levels of NLR, CAR, SII, and SIRI were higher in ASS than in DM/PM and correlated with disease activity and specific clinical features. NLR, CAR, SII, and SIRI may be an aid in differentiating ASS from DM/PM and maybe promising biomarkers for ASS.
Collapse
Affiliation(s)
- Liuyi Huang
- Department of Clinical Laboratory, the First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Nanning, Guangxi, 530021, People’s Republic of China
| | - Xi Li
- Department of Clinical Laboratory, the First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Nanning, Guangxi, 530021, People’s Republic of China
| | - Wei Zhou
- Department of Clinical Laboratory, the First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Nanning, Guangxi, 530021, People’s Republic of China
| | - Haiqing Zhu
- Department of Clinical Laboratory, the First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Nanning, Guangxi, 530021, People’s Republic of China
| | - Yuehong Lao
- Department of Clinical Laboratory, the First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Nanning, Guangxi, 530021, People’s Republic of China
| | - Xiaoxia Huang
- Department of Clinical Laboratory, the First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Nanning, Guangxi, 530021, People’s Republic of China
| | - Zhenjia Deng
- Department of Clinical Laboratory, the First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Nanning, Guangxi, 530021, People’s Republic of China
| | - Yuting Tang
- Department of Clinical Laboratory, the First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Nanning, Guangxi, 530021, People’s Republic of China
| | - Jian Wang
- Department of Clinical Laboratory, the First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Nanning, Guangxi, 530021, People’s Republic of China
| |
Collapse
|
9
|
Nies JF, Krusche M. [Fever in rheumatological diseases]. Z Rheumatol 2024; 83:341-353. [PMID: 38634905 DOI: 10.1007/s00393-024-01505-y] [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] [Accepted: 02/20/2024] [Indexed: 04/19/2024]
Abstract
Fever is a frequent and important symptom in patients with rheumatological diseases and can be an expression of activity of the underlying rheumatological disease. There is great variability in the incidence of fever as a symptom of the disease between individual diseases. The growing understanding of the molecular signatures of the diseases can help to explain these discrepancies: A genetic overactivation of potently pyrogenic cytokines is the reason why fever is nearly always present in autoinflammatory syndromes. In contrast, fever is less common in polyarthritis and myositis and mostly limited to severe courses of disease. In the diagnostic work-up of fever, frequent differential diagnoses, such as infections, malignancies, side effects of drugs and hypersensitivity reactions should be considered. This article provides an overview of the physiology of the development of fever, describes the relevance of fever in individual rheumatological diseases and proposes a workflow for the clinical clarification of rheumatological patients who present with fever.
Collapse
Affiliation(s)
- Jasper F Nies
- Klinik II für Innere Medizin: Nephrologie, Rheumatologie, Diabetologie und Allgemeine Innere Medizin, Uniklinik Köln, Kerpener Str. 62, 50937, Köln, Deutschland.
| | - Martin Krusche
- III. Medizinische Klinik und Poliklinik für Nephrologie, Rheumatologie und Endokrinologie, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20251, Hamburg, Deutschland
| |
Collapse
|
10
|
Mo Y, Ye Y, Peng L, Sun X, Zhong X, Wu R. The central helicase domain holds the major conformational epitopes of melanoma differentiation-associated gene 5 autoantibodies. Rheumatology (Oxford) 2024; 63:1456-1465. [PMID: 37551942 PMCID: PMC11065446 DOI: 10.1093/rheumatology/kead397] [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: 12/20/2022] [Revised: 06/23/2023] [Accepted: 07/17/2023] [Indexed: 08/09/2023] Open
Abstract
OBJECTIVE Autoantibodies against MDA5 (melanoma differentiation-associated protein 5) serve as a biomarker for DM (dermatomyositis) and indicate a risk factor for interstitial lung disease (ILD). MDA5 is a protein responsible for sensing RNA virus infection and activating signalling pathways against it. However, little is known about the antigen epitopes on MDA5 autoantibodies. We aimed to determine the interaction of the MDA5 autoantibody-antigen epitope. METHODS Cell-based assays (CBAs), immunoprecipitation-immunoblot assays, and various immunoblotting techniques were used in the study. RESULTS We demonstrated that DM patient autoantibodies recognize MDA5 epitopes in a native conformation-dependent manner. Furthermore, we identified the central helicase domain (3Hel) formed by Hel1, Hel2i, Hel2, and pincer as the major epitopes. As proof of principle, the purified 3Hel efficiently absorbed MDA5 autoantibodies from patient sera through immunoprecipitation-immunoblot assay. CONCLUSION Our study uncovered the nature of the antigen epitopes on MDA5 and can provide guidance for diagnosis and a targeted therapeutic approach development.
Collapse
Affiliation(s)
- Yongxin Mo
- Department of Biotherapy Centre, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yan Ye
- Department of Rheumatology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Lisheng Peng
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiaobo Sun
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiaofen Zhong
- Department of Biotherapy Centre, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Rui Wu
- Department of Rehabilitation, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
11
|
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
|
12
|
Beckett M, Tan J, Bonnardeaux E, Dutz J, Shojania K, To F, Obrzut A, Aviña-Zubieta J, Huang K. Tofacitinib therapy in refractory inflammatory myositis: a retrospective cohort study of 41 patients. Rheumatology (Oxford) 2024; 63:1432-1436. [PMID: 37584672 DOI: 10.1093/rheumatology/kead404] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 07/10/2023] [Accepted: 07/17/2023] [Indexed: 08/17/2023] Open
Abstract
OBJECTIVES To evaluate the efficacy and safety of tofacitinib in treatment-refractory inflammatory myositis in a real-world clinical setting. METHODS All patients with refractory inflammatory myositis treated with tofacitinib from a single urban centre in Vancouver, British Columbia, Canada, were included from June 2016 to December 2022. The medical records of these patients were retrospectively reviewed. RESULTS A total of 41 patients were included, 23 with classic DM, 12 with amyopathic DM (ADM) and six with PM phenotype. The patients failed an average of four to five non-steroidal immunosuppressants before initiation of tofacitinib. In the classic DM and ADM group, tofacitinib offered clinically and statistically significant cutaneous improvement. In all myositis patients including the PM phenotype, no meaningful muscle strength response to tofacitinib was observed. 53.7% of the patients discontinued tofacitinib due to lack of benefit or death. Of the 19 patients who remained on tofacitinib at the conclusion of this study, tofacitinib demonstrated clinically and statistically significant improvement in cutaneous disease activity. CONCLUSION Tofacitinib appears to be highly effective in targeting cutaneous manifestations in classic DM and ADM; however, minimal benefit in muscle strength in the DM or PM phenotype were observed.
Collapse
Affiliation(s)
- Madelaine Beckett
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Julia Tan
- Division of Rheumatology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Evelyne Bonnardeaux
- Department of Dermatology, Department of Internal Medicine, University of Montreal, Montreal, QC, Canada
| | - Jan Dutz
- Faculty of Medicine, Department of Dermatology, University of British Columbia, Vancouver, BC, Canada
- Arthritis Research Canada, Vancouver, BC, Canada
- Division of Rheumatology, Vancouver General Hospital, Vancouver, BC, Canada
- BC Children's Hospital Research Center, Vancouver, BC, Canada
| | - Kamran Shojania
- Division of Rheumatology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
- Arthritis Research Canada, Vancouver, BC, Canada
- Division of Rheumatology, Vancouver General Hospital, Vancouver, BC, Canada
| | - Fergus To
- Division of Rheumatology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
- Division of Rheumatology, Vancouver General Hospital, Vancouver, BC, Canada
| | | | - J Aviña-Zubieta
- Division of Rheumatology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
- Arthritis Research Canada, Vancouver, BC, Canada
- Division of Rheumatology, Vancouver General Hospital, Vancouver, BC, Canada
| | - Kun Huang
- Division of Rheumatology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
- Division of Rheumatology, Vancouver General Hospital, Vancouver, BC, Canada
| |
Collapse
|
13
|
Qin L, Lin H, Zhang G, Wang J, Feng T, Lei Y, Xie Y, Xu T, Zhang X. Screening and validation of differentially expressed genes in polymyositis. Heliyon 2024; 10:e24537. [PMID: 38317877 PMCID: PMC10839795 DOI: 10.1016/j.heliyon.2024.e24537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 01/10/2024] [Accepted: 01/10/2024] [Indexed: 02/07/2024] Open
Abstract
Background Polymyositis (PM), a prevalent inflammatory myopathy, currently lacks defined pathogenic mechanism. To illuminate its pathogenesis, we integrated bioinformatics and clinical specimens to examine potential aberrant gene expression patterns and their localization. Methods We obtained GSE128470 and GSE3112 dataset from the Gene Expression Omnibus, performed Gene Set Enrichment Analysis (GSEA) and immune infiltration analysis using CiberSort, identified differentially expressed genes with Limma, conducted functional annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, constructed a Protein-Protein Interaction network, and identified hub genes using Cytoscape. ROC analysis evaluated hub gene diagnostic accuracy for PM, validated their expression levels with clinical specimens. Results DEG analysis revealed 51 upregulated and 779 downregulated genes. Gene Ontology (GO) analysis implicated Type I interferon (IFN-Ⅰ) signaling, while KEGG pointed to cell adhesion molecule activation and oxidative phosphorylation inhibition. Protein-Protein Interaction (PPI) analysis identified 8 diagnosffftic hub genes. Clinical samples confirmed their upregulation in PM, especially IRF1 and IRF9 between muscle fibers. Different immune cell infiltrations were observed in PM patients versus controls. Conclusions Our study explores potential pathogenic factors, diagnostic markers, and immune cells in PM, with a focus on verifying IRF1 and IRF9 upregulation in the IFN-I signaling pathway. These findings bear significance for PM diagnosis and treatment.
Collapse
Affiliation(s)
- Linmang Qin
- Department of Rheumatology and Immunology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Haobo Lin
- Department of Rheumatology and Immunology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Guangfeng Zhang
- Department of Rheumatology and Immunology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jieying Wang
- Department of Rheumatology and Immunology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Tianxiao Feng
- Department of Rheumatology and Immunology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yunxia Lei
- Department of Rheumatology and Immunology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yuesheng Xie
- Department of Rheumatology and Immunology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ting Xu
- Department of Rheumatology and Immunology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xiao Zhang
- Department of Rheumatology and Immunology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
14
|
Wang S, Tang Y, Chen X, Song S, Chen X, Zhou Q, Zeng L. Mitochondrial-related hub genes in dermatomyositis: muscle and skin datasets-based identification and in vivo validation. Front Genet 2024; 15:1325035. [PMID: 38389573 PMCID: PMC10882082 DOI: 10.3389/fgene.2024.1325035] [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: 10/20/2023] [Accepted: 01/23/2024] [Indexed: 02/24/2024] Open
Abstract
Background: Mitochondrial dysfunction has been implicated in the pathogenesis of dermatomyositis (DM), a rare autoimmune disease affecting the skin and muscles. However, the genetic basis underlying dysfunctional mitochondria and the development of DM remains incomplete. Methods: The datasets of DM muscle and skin tissues were retrieved from the Gene Expression Omnibus database. The mitochondrial related genes (MRGs) were retrieved from MitoCarta. DM-related modules in muscle and skin tissues were identified with the analysis of weighted gene co-expression network (WGCNA), and then compared with the MRGs to obtain the overlapping mitochondrial related module genes (mito-MGs). Subsequently, differential expression genes (DEGs) obtained from muscle and skin datasets were overlapped with MRGs to identify mitochondrial related DEGs (mito-DEGs). Next, functional enrichment analysis was applied to analyze possible relevant biological pathways. We used the Jvenn online tool to intersect mito-MGs with mito-DEGs to identify hub genes and validate them using reverse transcription quantitative polymerase chain reaction (RT-qPCR) and immunohistochemistry staining. In addition, we evaluated immune infiltration in muscle and skin tissues of DM patients using the one-sample gene set enrichment analysis (ssGSEA) algorithm and predicted potential transcription factor (TF) -gene network by NetworkAnalyst. Results: The WGCNA analysis revealed 105 mito-MGs, while the DEG analysis identified 3 mito-DEGs. These genes showed functional enrichment for amino acid metabolism, energy metabolism and oxidative phosphorylation. Through the intersection analysis of the mito-MGs from the WGCNA analysis and the mito-DEGs from the DEG set, three DM mito-hub genes (IFI27, CMPK2, and LAP3) were identified and validated by RT-qPCR and immunohistochemistry analysis. Additionally, positive correlations were observed between hub genes and immune cell abundance. The TF-hub gene regulatory network revealed significant interactions involving ERG, VDR, and ZFX with CMPK2 and LAP3, as well as SOX2 with LAP3 and IFI27, and AR with IFI27 and CMPK2. Conclusion: The mito-hub genes (IFI27, CMPK2, and LAP3) are identified in both muscles and skin tissues from DM patients. These genes may be associated with immune infiltration in DM, providing a new entry point for the pathogenesis of DM.
Collapse
Affiliation(s)
- Shuo Wang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yiping Tang
- Department of Internal Medicine, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xixi Chen
- Department of Rheumatology and Immunology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Siyuan Song
- Baylor College of Medicine, Houston, TX, United States
| | - Xi Chen
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - Qiao Zhou
- Department of Rheumatology and Immunology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Li Zeng
- Department of Neurology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
15
|
Connolly CM, Gupta L, Fujimoto M, Machado PM, Paik JJ. Idiopathic inflammatory myopathies: current insights and future frontiers. THE LANCET. RHEUMATOLOGY 2024; 6:e115-e127. [PMID: 38267098 DOI: 10.1016/s2665-9913(23)00322-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/24/2023] [Accepted: 11/29/2023] [Indexed: 01/26/2024]
Abstract
Idiopathic inflammatory myopathies are a group of autoimmune diseases with a broad spectrum of clinical presentations, primarily characterised by immune-mediated muscle injury. Until recently, there was little insight into the pathogenesis of idiopathic inflammatory myopathies, which challenged the recognition of the breadth of heterogeneity of this group of diseases as well as the development of new therapeutics. However, the landscape of idiopathic inflammatory myopathies is evolving. In the past decade, advances in diagnostic tools have facilitated an enhanced understanding of the underlying disease mechanisms in idiopathic inflammatory myopathies, enabling the expansion of therapeutic trials. The fields of transcriptomics, prot§eomics, and machine learning offer the potential to gain greater insights into the underlying pathophysiology of idiopathic inflammatory myopathies. Harnessing insights gained from these sophisticated tools could contribute to the identification of differences at a molecular level among patients, accelerating the development of targeted, tailored therapies. Bolstered by the validation and standardisation of robust outcome measures, many promising therapies are in clinical trial development. Although challenges remain, there is great optimism in the field due to the progress in innovative diagnostics, outcome measures, and therapeutic approaches. In this Review, we discuss the expanding landscape of idiopathic inflammatory myopathies as the frontier of precision medicine becomes imminent.
Collapse
Affiliation(s)
- Caoilfhionn M Connolly
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Latika Gupta
- Department of Rheumatology, Royal Wolverhampton Hospitals NHS Trust, Wolverhampton, UK; Division of Musculoskeletal and Dermatological Sciences, Centre for Musculoskeletal Research, School of Biological Sciences, The University of Manchester, Manchester, UK; Department of Rheumatology, City Hospital, Sandwell and West Birmingham Hospitals NHS Trust, Birmingham, UK
| | - Manabu Fujimoto
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Pedro M Machado
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK; Centre for Rheumatology, University College London, London, UK; National Institute for Health and Care Research University College London Hospitals Biomedical Research Centre, University College London Hospitals NHS Foundation Trust, London, UK; Department of Rheumatology, Northwick Park Hospital, London North West University Healthcare NHS Trust, London, UK
| | - Julie J Paik
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
16
|
Kuzuya M. Drug-related sarcopenia as a secondary sarcopenia. Geriatr Gerontol Int 2024; 24:195-203. [PMID: 38158766 PMCID: PMC11503558 DOI: 10.1111/ggi.14770] [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: 11/06/2023] [Accepted: 11/24/2023] [Indexed: 01/03/2024]
Abstract
Sarcopenia has a significant impact on falls, physical function, activities of daily living, and quality of life in older adults, and its prevention and treatment are becoming increasingly important as the global population ages. In addition to primary age-related sarcopenia, activity-related sarcopenia, disease-related sarcopenia, and nutrition-related sarcopenia have been proposed as secondary sarcopenia. Polypharmacy and potentially inappropriate medication based on multiple diseases cause health problems in older patients. In some cases, drugs used for therapeutic or preventive purposes act on skeletal muscle as adverse drug reactions and induce sarcopenia. Although sarcopenia caused by these adverse drug reactions may be more common in older patients, in particular those taking many medications, drug-related sarcopenia has not yet received much attention. This review summarizes drugs that may induce sarcopenia and emphasizes the importance of drug-related sarcopenia as a secondary sarcopenia. Geriatr Gerontol Int 2024; 24: 195-203.
Collapse
Affiliation(s)
- Masafumi Kuzuya
- Meitetsu HospitalNagoyaJapan
- Professor Emeritus Nagoya UniversityNagoyaJapan
| |
Collapse
|
17
|
Essouma M. Autoimmune inflammatory myopathy biomarkers. Clin Chim Acta 2024; 553:117742. [PMID: 38176522 DOI: 10.1016/j.cca.2023.117742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 12/21/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024]
Abstract
The autoimmune inflammatory myopathy disease spectrum, commonly known as myositis, is a group of systemic diseases that mainly affect the muscles, skin and lungs. Biomarker assessment helps in understanding disease mechanisms, allowing for the implementation of precise strategies in the classification, diagnosis, and management of these diseases. This review examines the pathogenic mechanisms and highlights current data on blood and tissue biomarkers of autoimmune inflammatory myopathies.
Collapse
Affiliation(s)
- Mickael Essouma
- Network of Immunity in Infections, Malignancy and Autoimmunity, Universal Scientific Education and Research Network, Cameroon
| |
Collapse
|
18
|
Shih Y, Chen S, Huang J, Chen Y, Zhu Z, Zhao Q, Zhao X, Xue F, Xiang J, Chen X, Zhu X, Pan M, Wu J, Zheng J, Li H, Cao H. Serum level of galectin-9 as a potential biomarker for high risk of malignancy in dermatomyositis. Rheumatology (Oxford) 2024; 63:251-258. [PMID: 37184873 DOI: 10.1093/rheumatology/kead222] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 03/28/2023] [Accepted: 04/14/2023] [Indexed: 05/16/2023] Open
Abstract
OBJECTIVES Galectin-9, as immune checkpoint protein, plays a role in regulating autoimmunity and tumour immunity. Therefore, we explored the pathophysiological link between galectin-9 and malignancy in cancer-related DM (CRDM). METHODS Serum galectin-9 were quantified via enzyme-linked immunosorbent assay, and its association with serological indices was evaluated using Spearman analysis. Receiver operating characteristic (ROC) analysis was utilized to determine the cut-off value of galectin-9. RESULTS Serum levels of galectin-9 were significantly higher in DM patients [23.38 (13.85-32.57) ng/ml] than those in healthy controls (HCs) [6.81 (5.42-7.89) ng/ml, P < 0.0001], and were positively correlated with the cutaneous dermatomyositis disease area severity index activity (CDASI-A) scores (rs=0.3065, P = 0.0172). DM patients with new-onset and untreated cancer (new-CRDM) [31.58 (23.85-38.84) ng/ml] had higher levels of galectin-9 than those with stable and treated cancer (stable-CRDM) [17.49 (10.23-27.91) ng/ml, P = 0.0288], non-cancer-related DM (non-CRDM) [21.05 (11.97-28.02) ng/ml, P = 0.0258], and tumour patients without DM [7.46 (4.90-8.51) ng/ml, P < 0.0001]. Serum galectin-9 levels significantly decreased [27.79 (17.04-41.43) ng/ml vs 13.88 (5.15-20.37) ng/ml, P = 0.002] after anti-cancer treatment in CRDM patients. The combination of serum galectin-9 and anti-transcriptional intermediary factor 1-γ (anti-TIF1-γ) antibody (AUC = 0.889, 95% CI 0.803-0.977) showed the highest predictive value for the presence of cancer in DM. CONCLUSION Increased galectin-9 levels were related to tumor progression in CRDM, and galectin-9 was downregulated upon cancer treatment. Monitoring serum galectin-9 levels and anti-TIF1-γ antibodies might be an attractive strategy to achieve tumour diagnosis and predict CRDM outcome.
Collapse
Affiliation(s)
- Yanting Shih
- Department of Dermatology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shile Chen
- Department of Dermatology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Huang
- Department of Dermatology, Wuxi Branch of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongheng Chen
- Department of Dermatology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zicong Zhu
- Department of Dermatology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Zhao
- Department of Dermatology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoqing Zhao
- Department of Dermatology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Xue
- Department of Dermatology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Xiang
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaosong Chen
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuemei Zhu
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meng Pan
- Department of Dermatology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Wu
- Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Jie Zheng
- Department of Dermatology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Li
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hua Cao
- Department of Dermatology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
19
|
Corman VM, Preusse C, Melchert J, Benveniste O, Koll R, Goebel HH, Jones TC, Drosten C, Schara-Schmidt U, Leonard-Louis S, Stenzel W, Radke J. Deep RNA sequencing of muscle tissue reveals absence of viral signatures in dermatomyositis. FREE NEUROPATHOLOGY 2024; 5:5-1. [PMID: 38205217 PMCID: PMC10774810 DOI: 10.17879/freeneuropathology-2024-5149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024]
Abstract
Objective: To explore a possible connection between active viral infections and manifestation of dermatomyositis (DM). Methods: Skeletal muscle biopsies were analyzed from patients diagnosed with juvenile (n=10) and adult (n=12) DM. Adult DM patients harbored autoantibodies against either TIF-1γ (n=7) or MDA5 (n=5). Additionally, we investigated skeletal muscle biopsies from non-diseased controls (NDC, n=5). We used an unbiased high-throughput RNA sequencing (HTS) approach to detect viral sequences. To further increase sequencing depth, a host depletion approach was applied. Results: In this observational study, no relevant viral sequences were detected either by native sequencing or after host depletion. The absence of detectable viral sequences makes an active viral infection of the muscle tissue unlikely to be the cause of DM in our cohorts. Discussion: Type I interferons (IFN) play a major role in the pathogenesis of both juvenile and adult DM. The IFN response is remarkably conserved between DM subtypes classified by specific autoantibodies. Certain acute viral infections are accompanied by a prominent type I IFN response involving similar downstream mechanisms as in DM. Aiming to elucidate the pathogenesis of DM in skeletal muscle tissue, we used deep RNA sequencing and a host depletion approach to detect possible causative viruses.
Collapse
Affiliation(s)
- Victor M. Corman
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Virology, 10117 Berlin, Germany and German Centre for Infection Research (DZIF), Partner Site Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Corinna Preusse
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Neuropathology, 10117 Berlin, Germany
| | - Julia Melchert
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Virology, 10117 Berlin, Germany and German Centre for Infection Research (DZIF), Partner Site Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Olivier Benveniste
- Department of Internal Medicine and Clinical Immunology, Pitié-Salpêtrière University Hospital, 75013 Paris, France
| | - Randi Koll
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Neuropathology, 10117 Berlin, Germany
| | - Hans-Hilmar Goebel
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Neuropathology, 10117 Berlin, Germany
| | - Terry C. Jones
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Virology, 10117 Berlin, Germany and German Centre for Infection Research (DZIF), Partner Site Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Centre for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge, United Kingdom
| | - Christian Drosten
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Virology, 10117 Berlin, Germany and German Centre for Infection Research (DZIF), Partner Site Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Ulrike Schara-Schmidt
- Department of Pediatric Neurology and Centre for Neuromuscular Disorders in children and adolescents, Center for Translational Neuro- and Behavioral Sciences, University Duisburg-Essen, Essen, Germany
| | - Sarah Leonard-Louis
- Department of Neuropathology, Sorbonne Université, Assistance Publique-Hôpitaux de Paris, INSERM, Hôpital Pitié-Salpêtrière, France
| | - Werner Stenzel
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Neuropathology, 10117 Berlin, Germany
| | - Josefine Radke
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Neuropathology, 10117 Berlin, Germany
- Institute of Pathology, Universitätsmedizin Greifswald, Greifswald, Germany
| |
Collapse
|
20
|
Schutz PW, Cheung S, Yi L, Rossi FMV. Cellular activation patterns of CD10+ fibro-adipogenic progenitors across acquired disease states in human skeletal muscle biopsies. FREE NEUROPATHOLOGY 2024; 5:5-3. [PMID: 38357523 PMCID: PMC10865694 DOI: 10.17879/freeneuropathology-2024-5162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 01/27/2023] [Indexed: 02/16/2024]
Abstract
Background: Fibro-adipogenic progenitors (FAP) are muscle resident mesenchymal stem cells pivotal for regulation of myofiber repair. Experimental results show in addition involvement in a range of other pathological conditions and potential for pharmacological intervention. FAP histopathology in human muscle biopsies is largely unknown, but has potential to inform translational research. Methods: CD10+ FAPs in 32 archival muscle biopsies from 8 groups (normal, dermatomyositis, inclusion body myositis (IBM), anti-synthetase syndrome, immune-mediated necrotizing myopathy (IMNM), denervation, type 2 atrophy, rhabdomyolysis) were visualized by CD10 immunohistochemistry and their histology compared. Groups are compared by semi-quantitative scoring. Results: Histological activation of endomysial CD10+ FAPs includes prominent expansion of a network of cell processes surrounding muscle fibers, as well as endomysial cell clusters evidencing proliferation. Prominence of periarteriolar processes is a notable feature in some pathologies. FAP activation is often associated with fiber degeneration/regeneration, foci of inflammation, and denervation in keeping with experimental results. Type 2 atrophy shows no evidence of FAP activation. Dermatomyositis and anti-synthetase syndrome associated myositis demonstrate diffuse activation. Conclusion: Assessment of CD10+ FAP activation is routinely possible using CD10 immunohistochemistry and demonstrates several patterns in keeping with preclinical results. Prominent expansion of FAP processes surrounding myofibers suggests enhanced interaction between myofiber/basement membranes and FAPs during activation. The presence of diffuse FAP activation in dermatomyositis biopsies unrelated to fiber repair raises the possibility of FAP activation as part of the autoimmune process. Future diagnostic applications, clinical significance and therapeutic potential remain to be elucidated.
Collapse
Affiliation(s)
- Peter W. Schutz
- Department of Pathology, University of British Columbia, Vancouver, Canada
| | - Simon Cheung
- Department of Pathology, Vancouver General Hospital, Vancouver, Canada
| | - Lin Yi
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| | - Fabio M. V. Rossi
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| |
Collapse
|
21
|
Honda M, Shimizu F, Sato R, Nakamori M. Contribution of Complement, Microangiopathy and Inflammation in Idiopathic Inflammatory Myopathies. J Neuromuscul Dis 2024; 11:5-16. [PMID: 38143369 PMCID: PMC10789353 DOI: 10.3233/jnd-230168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/20/2023] [Indexed: 12/26/2023]
Abstract
PURPOSE OF REVIEW Idiopathic inflammatory myopathies (IIMs) are a heterogeneous group characterized by muscle weakness and skin symptoms and are categorized into six subtypes: dermatomyositis (DM), polymyositis (PM), anti-synthetase syndrome (ASS), immune-mediated myopathy (IMNM), inclusion body myopathy (IBM), and overlap myositis. Myositis-specific autoantibodies were detected for the diagnosis and classification of IIM. This review highlights the pathogenic contributions of the complement system, microangiopathy, and inflammation in IIM. RECENT FINDINGS Deposition of complement around capillaries and/or the sarcolemma was observed in muscle biopsy specimens from patients with DM, ASS, and IMNM, suggesting the pathomechanism of complement-dependent muscle and endothelial cell injury. A recent study using human muscle microvascular endothelial cells showed that Jo-1 antibodies from ASS induce complement-dependent cellular cytotoxicity in vitro. Based on both clinical and pathological observations, antibody- and complement-mediated microangiopathy may contribute to the development of DM and anti-Jo-1 ASS. Juvenile DM is characterized by the loss of capillaries, perivascular inflammation, and small-vessel angiopathies, which may be related to microinfarction and perifascicular atrophy. Several serum biomarkers that reflect the IFN1 signature and microangiopathy are elevated in patients with DM. The pathological observation of myxovirus resistance protein A (MxA), which suggests a type 1 interferon (IFN1) signature in DM, supports the diagnosis and further understanding of the pathomechanism of IIM. A recent report showed that an increase in triggering receptor expressed on myeloid cells (TREM-1) around perimysial blood vessels and muscles in patients with IIM plays a role in triggering inflammation and promoting the migration of inflammatory cells by secreting proinflammatory cytokines, such as tumor necrosis factor α. SUMMARY The deposition of complement in muscles and capillaries is a characteristic feature of DM, ASS, and IMNM. Microangiopathy plays a pathogenic role in DM, possibly resulting in perifascicular atrophy. Further understanding of the detailed pathomechanism regarding complement, microangiopathy, and inflammation may lead to novel therapeutic approaches for IIM.
Collapse
Affiliation(s)
- Masaya Honda
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Fumitaka Shimizu
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Ryota Sato
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Masayuki Nakamori
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| |
Collapse
|
22
|
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
|
23
|
Yang Y, Song J, Zhao H, Zhang H, Guo M. Patients with dermatomyositis shared partially similar transcriptome signature with COVID-19 infection. Autoimmunity 2023; 56:2220984. [PMID: 37353938 DOI: 10.1080/08916934.2023.2220984] [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: 02/08/2023] [Accepted: 05/28/2023] [Indexed: 06/25/2023]
Abstract
Dermatomyositis (DM) is an autoimmune disease that primarily affects the skin and skeletal muscle. Virus infection and type I interferon-related signaling pathways play an important role in the pathogenesis of dermatomyositis. In this study, we found that the skin of patients with DM and the skin of patients with COVID-19 have similar transcriptional profiles, and identified key genes involved in dermatomyositis based on bioinformatics analysis. These hub-genes might be served as potential biomarkers for the early diagnosis and therapy of DM, including MX1, ISG15, IFIT3, IFIT1, RSAD2, IFIT2, IFI6, XAF1, IRF9, MX2.
Collapse
Affiliation(s)
- Yiying Yang
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, Hunan, China
| | - Jie Song
- Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, Hunan, China
| | - Hongjun Zhao
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| | - Huali Zhang
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
- Sepsis Translational Medicine Key Lab of Hunan Province, Changsha, Hunan, China
| | - Muyao Guo
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
| |
Collapse
|
24
|
Chronopoulou S, Tsochantaridis I, Tokamani M, Kokkinopliti KD, Tsomakidis P, Giannakakis A, Galanis A, Pappa A, Sandaltzopoulos R. Expression and purification of human interferon alpha 2a (IFNα2a) in the methylotrophic yeast Pichia pastoris. Protein Expr Purif 2023; 211:106339. [PMID: 37467825 DOI: 10.1016/j.pep.2023.106339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 07/15/2023] [Indexed: 07/21/2023]
Abstract
Human interferon alpha 2a (IFNα2a) is a secreted glycoprotein that exerts a wide spectrum of biological effects, such as triggering of antiviral, antitumor and immunosuppressive responses. IFNα2a is used as pharmaceutical polypeptide in chronic hepatitis C virus (HCV) infection, chronic myelogenous leukemia, advanced renal cell carcinoma, and metastatic malignant melanoma. So far, the pharmaceutical polypeptide of this cytokine is produced in prokaryotic expression systems (E. coli). Here we report the expression and purification of recombinant human IFNα2a in the methylotrophic yeast Pichia pastoris. The cDNA encoding for human IFNα2a, modified to bear the P. pastoris codon bias, was cloned into the pPinkα-HC vector in order to be expressed as a secreted protein upon induction. Proper expression and secretion of recombinant human IFNα2a (approximately 19 kDa) was confirmed by PCR-sequencing, SDS-PAGE and Western blot analysis following methanol-induced expression in a number of individual transformed strains. Purification of the recombinant protein was performed by affinity chromatography, achieving a robust yield of purified active form. The purified recombinant protein showed an impressive stability to thermal denaturation as observed by Differential Scanning Fluorimetry. The biological activity of the P. pastoris-produced IFNα2a was confirmed in A549 and HT29 cells by monitoring transcriptional up-regulation of a panel of known interferon-stimulated genes (ISGs). Our results document that the P. pastoris expression system is a suitable system for producing biologically functional IFNα2a in a secreted form.
Collapse
Affiliation(s)
- Sofia Chronopoulou
- Department of Molecular Biology & Genetics, Democritus University of Thrace, Alexandroupolis, Greece.
| | - Ilias Tsochantaridis
- Department of Molecular Biology & Genetics, Democritus University of Thrace, Alexandroupolis, Greece.
| | - Maria Tokamani
- Department of Molecular Biology & Genetics, Democritus University of Thrace, Alexandroupolis, Greece.
| | | | - Petros Tsomakidis
- Department of Molecular Biology & Genetics, Democritus University of Thrace, Alexandroupolis, Greece.
| | - Antonis Giannakakis
- Department of Molecular Biology & Genetics, Democritus University of Thrace, Alexandroupolis, Greece.
| | - Alex Galanis
- Department of Molecular Biology & Genetics, Democritus University of Thrace, Alexandroupolis, Greece.
| | - Aglaia Pappa
- Department of Molecular Biology & Genetics, Democritus University of Thrace, Alexandroupolis, Greece.
| | - Raphael Sandaltzopoulos
- Department of Molecular Biology & Genetics, Democritus University of Thrace, Alexandroupolis, Greece.
| |
Collapse
|
25
|
Kim H. Insights into juvenile myositis via engineered muscle. Nat Rev Rheumatol 2023; 19:543-544. [PMID: 37495869 PMCID: PMC11259152 DOI: 10.1038/s41584-023-01003-6] [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] [Indexed: 07/28/2023]
Abstract
The pathogenesis of juvenile dermatomyositis (JDM) is complex and various evidence implicate a role for type I interferons. Could the use of a bioengineered paediatric skeletal muscle model provide insight into this disease and have potential for high throughput testing of therapeutic agents?
Collapse
Affiliation(s)
- Hanna Kim
- Juvenile Myositis Pathogenesis and Therapeutics Unit, National Institute of Arthritis and Musculoskeletal and Skin Disease, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
26
|
Doskaliuk B, Ravichandran N, Sen P, Day J, Joshi M, Nune A, Nikiphorou E, Saha S, Tan AL, Shinjo SK, Ziade N, Velikova T, Milchert M, Jagtap K, Parodis I, Gracia-Ramos AE, Cavagna L, Kuwana M, Knitza J, Chen YM, Makol A, Agarwal V, Patel A, Pauling JD, Wincup C, Barman B, Tehozol EAZ, Serrano JR, La Torre IGD, Colunga-Pedraza IJ, Merayo-Chalico J, Chibuzo OC, Katchamart W, Goo PA, Shumnalieva R, Hoff LS, Kibbi LE, Halabi H, Vaidya B, Shaharir SS, Hasan ATMT, Dey D, Gutiérrez CET, Caballero-Uribe CV, Lilleker JB, Salim B, Gheita T, Chatterjee T, Distler O, Saavedra MA, Chinoy H, Agarwal V, Aggarwal R, Gupta L. Long-term safety of COVID vaccination in individuals with idiopathic inflammatory myopathies: results from the COVAD study. Rheumatol Int 2023; 43:1651-1664. [PMID: 37351634 PMCID: PMC10348925 DOI: 10.1007/s00296-023-05345-y] [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: 04/18/2023] [Accepted: 05/10/2023] [Indexed: 06/24/2023]
Abstract
Limited evidence on long-term COVID-19 vaccine safety in patients with idiopathic inflammatory myopathies (IIMs) continues to contribute to vaccine hesitancy. We studied delayed-onset vaccine adverse events (AEs) in patients with IIMs, other systemic autoimmune and inflammatory disorders (SAIDs), and healthy controls (HCs), using data from the second COVID-19 Vaccination in Autoimmune Diseases (COVAD) study. A validated self-reporting e-survey was circulated by the COVAD study group (157 collaborators, 106 countries) from Feb-June 2022. We collected data on demographics, comorbidities, IIM/SAID details, COVID-19 history, and vaccination details. Delayed-onset (> 7 day) AEs were analyzed using regression models. A total of 15165 respondents undertook the survey, of whom 8759 responses from vaccinated individuals [median age 46 (35-58) years, 74.4% females, 45.4% Caucasians] were analyzed. Of these, 1390 (15.9%) had IIMs, 50.6% other SAIDs, and 33.5% HCs. Among IIMs, 16.3% and 10.2% patients reported minor and major AEs, respectively, and 0.72% (n = 10) required hospitalization. Notably patients with IIMs experienced fewer minor AEs than other SAIDs, though rashes were expectedly more than HCs [OR 4.0; 95% CI 2.2-7.0, p < 0.001]. IIM patients with active disease, overlap myositis, autoimmune comorbidities, and ChadOx1 nCOV-19 (Oxford/AstraZeneca) recipients reported AEs more often, while those with inclusion body myositis, and BNT162b2 (Pfizer) recipients reported fewer AEs. Vaccination is reassuringly safe in individuals with IIMs, with AEs, hospitalizations comparable to SAIDs, and largely limited to those with autoimmune multimorbidity and active disease. These observations may inform guidelines to identify high-risk patients warranting close monitoring in the post-vaccination period.
Collapse
Affiliation(s)
- Bohdana Doskaliuk
- Department of Pathophysiology, Ivano-Frankivsk National Medical University, Ivano-Frankivsk, Ukraine
| | - Naveen Ravichandran
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Parikshit Sen
- Maulana Azad Medical College, 2-Bahadurshah Zafar Marg, New Delhi, Delhi, 110002, India
| | - Jessica Day
- Department of Rheumatology, Royal Melbourne Hospital, Parkville, VIC, 3050, Australia
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Mrudula Joshi
- Byramjee Jeejeebhoy Government Medical College and Sassoon General Hospitals, Pune, India
| | - Arvind Nune
- Southport and Ormskirk Hospital NHS Trust, Southport, PR8 6PN, UK
| | - Elena Nikiphorou
- Centre for Rheumatic Diseases, King's College London, London, UK
- Rheumatology Department, King's College Hospital, London, UK
| | - Sreoshy Saha
- Mymensingh Medical College, Mymensingh, Bangladesh
| | - Ai Lyn Tan
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals Trust, Leeds, UK
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - Samuel Katsuyuki Shinjo
- Division of RheumatologyFaculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Nelly Ziade
- Rheumatology Department, Saint-Joseph University, Beirut, Lebanon
- Rheumatology Department, Hotel-Dieu de France Hospital, Beirut, Lebanon
| | - Tsvetelina Velikova
- Medical Faculty, Sofia University St. Kliment Ohridski, 1 Kozyak Str, 1407, Sofia, Bulgaria
| | - Marcin Milchert
- Department of Internal Medicine, Rheumatology, Diabetology, Geriatrics and Clinical Immunology, Pomeranian Medical University in Szczecin, Ul Unii Lubelskiej 1, 71-252, Szczecin, Poland
| | - Kshitij Jagtap
- Seth Gordhandhas Sunderdas Medical College and King Edwards Memorial Hospital, Mumbai, Maharashtra, India
| | - Ioannis Parodis
- Division of RheumatologyDepartment of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Department of RheumatologyFaculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Abraham Edgar Gracia-Ramos
- Department of Internal Medicine, General Hospital, National Medical Center "La Raza", Instituto Mexicano del Seguro Social, Av. Jacaranda S/N, Col. La Raza, Del. Azcapotzalco, C.P. 02990, Mexico City, Mexico
| | - Lorenzo Cavagna
- Rheumatology UnitDipartimento Di Medicine Interna E Terapia Medica, Università Degli Studi Di Pavia, Pavia, Lombardy, Italy
| | - Masataka Kuwana
- Department of Allergy and Rheumatology, Nippon Medical School Graduate School of Medicine, 1-1-5 Sendagi, Bunkyo-Ku, Tokyo, 113-8602, Japan
| | - Johannes Knitza
- Medizinische Klinik 3-Rheumatologie und Immunologie, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Ulmenweg 18, 91054, Erlangen, Deutschland
| | - Yi Ming Chen
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung City, Taiwan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Ashima Makol
- Division of Rheumatology, Mayo Clinic, Rochester, MN, USA
| | - Vishwesh Agarwal
- Mahatma Gandhi Mission Medical College, Navi Mumbai, Maharashtra, India
| | - Aarat Patel
- Bon Secours Rheumatology Center and Division of Pediatric RheumatologyDepartment of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - John D Pauling
- Bristol Medical School Translational Health Sciences, University of Bristol, Bristol, UK
- Department of Rheumatology, North Bristol NHS Trust, Bristol, UK
| | - Chris Wincup
- Division of Medicine, Rayne InstituteDepartment of Rheumatology, University College London, London, UK
- Centre for Adolescent Rheumatology Versus Arthritis at UCL, UCLH, GOSH, London, UK
| | - Bhupen Barman
- Department of General Medicine, All India Institute of Medical Sciences (AIIMS), Guwahati, India
| | - Erick Adrian Zamora Tehozol
- Rheumatology, Medical Care & Research, Centro Medico Pensiones Hospital, Instituto Mexicano del Seguro Social Delegación Yucatán, Yucatán, Mexico
| | - Jorge Rojas Serrano
- Rheumatologist and Clinical InvestigatorInterstitial Lung Disease and Rheumatology Unit, Instituto Nacional de Enfermedades Respiratorias, Mexico City, Mexico
| | - Ignacio García-De La Torre
- Departamento de Inmunología Y Reumatología, Hospital General de Occidente and Universidad de Guadalajara, Guadalajara, Jalisco, Mexico
| | | | - Javier Merayo-Chalico
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas Y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Okwara Celestine Chibuzo
- Department of Medicine, University of Nigeria Teaching Hospital, Ituku-Ozalla/University of Nigeria, Enugu Campus, Enugu, Nigeria
| | - Wanruchada Katchamart
- Division of RheumatologyDepartment of MedicineFaculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | | | - Russka Shumnalieva
- Department of RheumatologyClinic of Rheumatology, University Hospital "St. Ivan Rilski", Medical University-Sofia, Sofia, Bulgaria
| | | | - Lina El Kibbi
- Internal Medicine Department, Rheumatology Unit, Specialized Medical Center, Riyadh, Saudi Arabia
| | - Hussein Halabi
- Department of Internal MedicineSection of Rheumatology, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
| | - Binit Vaidya
- National Center for Rheumatic Diseases (NCRD), Ratopul, Kathmandu, Nepal
| | | | - A T M Tanveer Hasan
- Department of Rheumatology, Enam Medical College & Hospital, Dhaka, Bangladesh
| | - Dzifa Dey
- Department of Medicine and Therapeutics, Rheumatology Unit, University of Ghana Medical SchoolCollege of Health Sciences, Korle-Bu, Accra, Ghana
| | - Carlos Enrique Toro Gutiérrez
- Reference Center for Osteoporosis, Rheumatology and Dermatology, Pontifica Universidad Javeriana Cali, Cali, Colombia
| | | | - James B Lilleker
- Division of Musculoskeletal and Dermatological Sciences, Centre for Musculoskeletal Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
- Manchester Centre for Clinical Neurosciences, Salford Royal NHS Foundation Trust, Salford, UK
| | - Babur Salim
- Rheumatology Department, Fauji Foundation Hospital, Rawalpindi, Pakistan
| | - Tamer Gheita
- Rheumatology Department, Kasr Al Ainy School of Medicine, Cairo University, Cairo, Egypt
| | - Tulika Chatterjee
- Department of Internal Medicine, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
| | - Oliver Distler
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Miguel A Saavedra
- Departamento de Reumatología Hospital de Especialidades Dr. Antonio Fraga Mouret, Centro Médico Nacional La Raza, IMSS, Mexico City, Mexico
| | - Hector Chinoy
- Division of Musculoskeletal and Dermatological Sciences, Centre for Musculoskeletal Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
- National Institute for Health Research Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, The University of Manchester, Manchester, UK
- Department of Rheumatology, Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Salford, UK
| | - Vikas Agarwal
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Rohit Aggarwal
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Latika Gupta
- Division of Musculoskeletal and Dermatological Sciences, Centre for Musculoskeletal Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK.
- Department of Rheumatology, Royal Wolverhampton Hospitals NHS Trust, Wolverhampton, UK.
- City Hospital, Sandwell and West Birmingham Hospitals NHS Trust, Birmingham, UK.
| |
Collapse
|
27
|
Morales M, Alayi TD, Tawalbeh SM, Sydenstricker AV, Spathis R, Kim H, Nagaraju K, Hathout Y, Rider LG. Urine proteomics by mass spectrometry identifies proteins involved in key pathogenic pathways in patients with juvenile dermatomyositis. Rheumatology (Oxford) 2023; 62:3161-3168. [PMID: 36661295 PMCID: PMC10473190 DOI: 10.1093/rheumatology/kead033] [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: 09/27/2022] [Revised: 12/20/2022] [Accepted: 01/10/2023] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVES To identify and validate biomarkers in JDM patients using a multiplexing tandem mass tag urine proteome profiling approach. METHODS First morning void urine samples were collected from JDM patients (n = 20) and healthy control subjects (n = 21) and processed for analysis using a standardized liquid chromatography-tandem mass spectrometry approach. Biomarkers with significantly altered levels were correlated with clinical measures of myositis disease activity and damage. A subset of candidate biomarkers was validated using commercially available ELISA kits. RESULTS In total, 2348 proteins were detected in the samples, with 275 proteins quantified across all samples. Among the differentially altered proteins, cathepsin D and galectin-3 binding protein were significantly increased in the urine of JDM patients (adjusted P < 0.05), supporting previous findings in myositis patients. These two candidate biomarkers were confirmed with ELISAs. Cathepsin D positively correlated with Myositis Damage Index (r = 0.57, P < 0.05) and negatively correlated with the Childhood Myositis Assessment Scale (r = -0.54, P < 0.05). We also identified novel JDM candidate biomarkers involved with key features of myositis, including extracellular matrix remodelling proteins. CONCLUSION This study confirmed the presence of several proteins in the urine of JDM patients that were previously found to be elevated in the blood of myositis patients and identified novel candidate biomarkers that require validation. These results support the use of urine as a source for biomarker development in JDM.
Collapse
Affiliation(s)
- Melissa Morales
- Department of Pharmaceutical Sciences, Binghamton University-State University of New York, Binghamton, NY, USA
| | - Tchilabalo D Alayi
- Department of Pharmaceutical Sciences, Binghamton University-State University of New York, Binghamton, NY, USA
| | - Shefa M Tawalbeh
- Department of Biomedical Systems and Informatics Engineering, Yarmouk University, Irbid, Jordan
| | - Agnes V Sydenstricker
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Rita Spathis
- Department of Pharmaceutical Sciences, Binghamton University-State University of New York, Binghamton, NY, USA
| | - Hanna Kim
- Juvenile Myositis Pathogenesis and Therapeutics Unit, National Institute of Arthritis Musculoskeletal and Skin Diseases, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Kanneboyina Nagaraju
- Department of Pharmaceutical Sciences, Binghamton University-State University of New York, Binghamton, NY, USA
| | - Yetrib Hathout
- Department of Pharmaceutical Sciences, Binghamton University-State University of New York, Binghamton, NY, USA
| | - Lisa G Rider
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
28
|
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: 3.0] [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
|
29
|
Zhang Z, Tao W, Cheng D, Qin M, Fu J, Liu D. Deciphering the crosstalk of immune dysregulation between COVID-19 and idiopathic inflammatory myopathy. Front Immunol 2023; 14:1197493. [PMID: 37638007 PMCID: PMC10449257 DOI: 10.3389/fimmu.2023.1197493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023] Open
Abstract
Background The coronavirus disease (COVID-19) pandemic is a serious threat to public health worldwide. Growing evidence reveals that there are certain links between COVID-19 and autoimmune diseases; in particular, COVID-19 and idiopathic inflammatory myopathies (IIM) have been observed to be clinically comorbid. Hence, this study aimed to elucidate the molecular mechanisms of COVID-19 and IIM from a genomic perspective. Methods We obtained transcriptome data of patients with COVID-19 and IIM separately from the GEO database and identified common differentially expressed genes (DEGs) by intersection. We then performed functional enrichment, PPI, machine learning, gene expression regulatory network, and immune infiltration analyses of co-expressed genes. Results A total of 91 common genes were identified between COVID-19 and IIM. Functional enrichment analysis revealed that these genes were mainly involved in immune dysregulation, response to external stimuli, and MAPK signaling pathways. The MCODE algorithm recognized two densely linked clusters in the common genes, which were related to inflammatory factors and interferon signaling. Subsequently, three key genes (CDKN1A, IFI27, and STAB1) were screened using machine learning to predict the occurrence of COVID-19 related IIM. These key genes exhibited excellent diagnostic performance in both training and validation cohorts. Moreover, we created TF-gene and miRNA-gene networks to reveal the regulation of key genes. Finally, we estimated the relationship between key genes and immune cell infiltration, of which IFI27 was positively associated with M1 macrophages. Conclusion Our work revealed common molecular mechanisms, core genes, potential targets, and therapeutic approaches for COVID-19 and IIM from a genomic perspective. This provides new ideas for the diagnosis and treatment of COVID-19 related IIM in the future.
Collapse
Affiliation(s)
- Zhao Zhang
- Department of Orthopaedics, Xi-Jing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Weidong Tao
- Department of Orthopaedics, Xi-Jing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Debin Cheng
- Department of Orthopaedics, Xi-Jing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Marong Qin
- School of Chemistry, Cardiff University, Cardiff, United Kingdom
| | - Jun Fu
- Department of Orthopaedics, Xi-Jing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Dong Liu
- Department of Orthopaedics, Xi-Jing Hospital, The Fourth Military Medical University, Xi’an, China
| |
Collapse
|
30
|
Skarlis C, Michalakeas N, Gerochristou M, Raftopoulou S, Marketos N, Boki K, Vassilopoulos D, Stratigos AP, Boumpas D, Mavragani C. The Role of Myositis-Specific and Myositis-Associated Autoantibodies and the Activation of Type I Interferon Pathway in the Generation of Clinical Phenotypes of Inflammatory Myopathies. Mediterr J Rheumatol 2023; 34:275-278. [PMID: 37654645 PMCID: PMC10466366 DOI: 10.31138/mjr.34.2.275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 10/14/2022] [Indexed: 09/02/2023] Open
Abstract
Idiopathic inflammatory myopathies (IIMs) are a group of heterogeneous autoimmune diseases with a prevalence of 20 cases per 100000 of population. Despite their diversity, IMMs are characterised by several common clinical features such as muscle inflammation, proximal muscle weakness, abnormal electromyography and/or muscle biopsy. Over the last years, it has been increasingly recognised that an array of autoantibodies known as myositis-specific antibodies (MSAs) and myositis-associated antibodies (MAAs) are associated with distinct clinical phenotypes and diverse prognosis. Although the exact underlying mechanism of IIMs is not fully understood, accumulating data suggest that the activation of type I interferon pathway plays a central role in disease development. Previous studies have reported the upregulation of type I interferon (IFN) induced genes in peripheral blood and muscle biopsies derived from myositis patients. Given the heterogeneity of inflammatory myopathies along with the central role of type I IFN pathway in disease pathogenesis, the aim of the current study is to elucidate the link between distinct clinical phenotypes of inflammatory myopathies with the presence of serum MSAs or MAAs, as well as with type I IFN activation.
Collapse
Affiliation(s)
- Charalampos Skarlis
- Department of Physiology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Michalakeas
- Department of Physiology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Gerochristou
- Department of Physiology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- First Department of Dermatology and Venereology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Sylvia Raftopoulou
- Department of Physiology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Marketos
- Department of Physiology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Dimitrios Vassilopoulos
- Clinical Immunology-Rheumatology, Hippokration General Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Joint Academic Rheumatology Program, Athens, Greece
| | | | - Dimitrios Boumpas
- Joint Academic Rheumatology Program, Athens, Greece
- Fourth Department of Internal Medicine, University Hospital Attikon, National and Kapodistrian University of Athens, Athens, Greece
| | - Clio Mavragani
- Department of Physiology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Joint Academic Rheumatology Program, Athens, Greece
- Fourth Department of Internal Medicine, University Hospital Attikon, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
31
|
Moon SJ, Jung SM, Baek IW, Park KS, Kim KJ. Molecular signature of neutrophil extracellular trap mediating disease module in idiopathic inflammatory myopathy. J Autoimmun 2023; 138:103063. [PMID: 37220716 DOI: 10.1016/j.jaut.2023.103063] [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: 03/20/2023] [Revised: 05/07/2023] [Accepted: 05/09/2023] [Indexed: 05/25/2023]
Abstract
The rarity and heterogeneity of idiopathic inflammatory myopathy (IIM) pose challenges for researching IIM in affected individuals. We analyzed integrated transcriptomic datasets obtained using muscle tissues from patients with five distinct IIM subtypes to investigate the shared and distinctive cellular and molecular characteristics. A transcriptomic dataset of muscle tissues from normal controls (n = 105) and patients with dermatomyositis (n = 89), polymyositis (n = 33), inclusion body myositis (n = 121), immune-mediated necrotizing myositis (n = 75), and anti-synthetase syndrome (n = 18) was used for differential gene-expression analysis, functional-enrichment analysis, gene set-enrichment analysis, disease-module identification, and kernel-based diffusion scoring. Damage-associated molecular pattern-associated pathways and neutrophil-mediated immunity were significantly enriched across different IIM subtypes, although their activities varied. Interferons-signaling pathways were differentially activated across all five IIM subtypes. In particular, neutrophil extracellular trap (NET) formation was significantly activated and correlated with Fcγ R-mediated signaling pathways. NET formation-associated genes were key for establishing disease modules, and FCGRs, C1QA, and SERPINE1 markedly perturbed the disease modules. Integrated transcriptomic analysis of muscle tissues identified NETs as key components of neutrophil-mediated immunity involved in the pathogenesis of IIM subtypes and, thus, has therapeutically targetable value.
Collapse
Affiliation(s)
- Su-Jin Moon
- Division of Rheumatology, Department of Internal Medicine, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung Min Jung
- Division of Rheumatology, Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - In-Woon Baek
- Division of Rheumatology, Department of Internal Medicine, Ewha Womans University College of Medicine, Seoul, Republic of Korea
| | - Kyung-Su Park
- Division of Rheumatology, Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ki-Jo Kim
- Division of Rheumatology, Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
32
|
Li M, Zhang Y, Zhang W, Sun J, Liu R, Pan Z, Zhang P, Liu S. Type 1 interferon signature in peripheral blood mononuclear cells and monocytes of idiopathic inflammatory myopathy patients with different myositis-specific autoantibodies. Front Immunol 2023; 14:1169057. [PMID: 37228600 PMCID: PMC10203462 DOI: 10.3389/fimmu.2023.1169057] [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: 02/18/2023] [Accepted: 04/21/2023] [Indexed: 05/27/2023] Open
Abstract
Background Myositis-specific autoantibodies (MSAs) are clinically used to diagnose and define idiopathic inflammatory myopathy (IIM) subsets. However, the underlying pathogenic mechanisms of patients with different MSAs remain unclear. Methods A total of 158 Chinese patients with IIM and 167 gender- and age-matched healthy controls (HCs) were enrolled. Transcriptome sequencing (RNA-Seq) was performed with peripheral blood mononuclear cells (PBMCs), followed by the identification of differentially expressed genes (DEGs) and analysis of gene set enrichment analysis, immune cell infiltration, and WGCNA. Monocyte subsets and related cytokines/chemokines were quantified. The expressions of interferon (IFN)-related genes were validated using qRT-PCR and Western blot in both PBMCs and monocytes. We also performed correlation analysis and ROC analysis to explore the potential clinical significance of the IFN-related genes. Results There were 1,364 genes altered in patients with IIM, including 952 upregulated and 412 downregulated genes. The type I interferon (IFN-I) pathway was remarkably activated in patients with IIM. Compared with patients with other MSAs, IFN-I signatures were significantly activated in patients with anti-melanoma differentiation-associated gene 5 (MDA5) antibodies. In total, 1,288 hub genes associated with IIM onset were identified using WGCNA, including 29 key DEGs associated with IFN signaling. The patients had more CD14brightCD16- classical, CD14brightCD16+ intermediate, and fewer CD14dimCD16+ non-classical monocyte subsets. Plasma cytokines like IL-6 and TNF and chemokines including CCL3 and MCPs increased. The validation of IFN-I-related gene expressions was consistent with the findings from RNA-Seq. The IFN-related genes were correlated with laboratory parameters and helpful for IIM diagnosis. Conclusion Gene expressions were remarkably altered in the PBMCs of IIM patients. Anti-MDA5+ IIM patients had a more pronounced activated IFN signature than others. Monocytes exhibited a proinflammatory feature and contributed to the IFN signature of IIM patients.
Collapse
Affiliation(s)
- Mengdi Li
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yusheng Zhang
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenzhe Zhang
- Department of Radiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jinlei Sun
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rui Liu
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhou Pan
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Panpan Zhang
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shengyun Liu
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
33
|
Zhang P, Li M, Zhang Y, Lian C, Sun J, He Y, Hu W, Wang L, Li T, Liu S, Zhang Y. Plasma proteomic profiling reveals KRT19 could be a potential biomarker in patients with anti-MDA5+ dermatomyositis. Clin Rheumatol 2023:10.1007/s10067-023-06624-6. [PMID: 37160775 DOI: 10.1007/s10067-023-06624-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/29/2023] [Accepted: 05/03/2023] [Indexed: 05/11/2023]
Abstract
OBJECTIVE To investigate the immune response-related protein profiling in plasma of patients with idiopathic inflammatory myopathies (IIMs), especially in anti-MDA5+ dermatomyositis (DM). METHODS A total of 166 IIM patients and 107 healthy controls (HCs) were enrolled in our study. Ninety-two plasma immune response-related proteins were detected by Olink proteomics in 36 IIM patients and 25 HCs. The expression of plasma KRT19 was validated in another 130 IIM patients, 82 HCs, and 55 other rheumatic diseases. RESULTS A total of 46 differentially expressed proteins were detected, including 12 upregulated proteins and 34 downregulated proteins in IIM patients compared with HCs. Pathway analysis revealed lactoferrin danger signal response pathway, TLR4 signaling and tolerance, infection, and IL-10 signaling pathway were activated. The immune response-related protein profiling significantly altered in anti-MDA5+ DM patients, with LAMP3, HSD11B1, and KRT19 significantly increased, while SH2D1A, ITGA11, TRIM21, CD28, ITGB6, and HEXIM1 tremendously decreased. In addition, KRT19 was significantly increased in IIM patients, especially in anti-MDA5+ DM patients with the diagnostic value of a significant area under the ROC curve of 0.881. CONCLUSION Immune response-related proteins are significantly altered in patients with anti-MDA5+ DM patients. KRT19 could be a potential biomarker for anti-MDA5+ DM patients. Key Points • What is already known on this topic? Anti-MDA5+ DM is a distinctive subtype of IIM. Plasma immune response-related proteins panel needs to be investigated. • What this study adds? Plasma protein profiling of immune response-related proteins significantly altered in patients with idiopathic inflammatory myopathies (IIM), especially in anti-MDA5+ DM patients. • How this study might affect research, practice, or policy? KRT19 could be a potential biomarker in patients with anti-MDA5+ dermatomyositis.
Collapse
Affiliation(s)
- Panpan Zhang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450000, Henan Province, China
| | - Mengdi Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450000, Henan Province, China
| | - Yuqi Zhang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450000, Henan Province, China
| | - Chaofeng Lian
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450000, Henan Province, China
| | - Jinlei Sun
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450000, Henan Province, China
| | - Yujie He
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450000, Henan Province, China
| | - Wenlu Hu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450000, Henan Province, China
| | - Limei Wang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450000, Henan Province, China
| | - Tianfang Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450000, Henan Province, China
| | - Shengyun Liu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450000, Henan Province, China.
| | - Yusheng Zhang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450000, Henan Province, China.
| |
Collapse
|
34
|
Markin AM, Markina YV, Bogatyreva AI, Tolstik TV, Chakal DA, Breshenkov DG, Charchyan ER. The Role of Cytokines in Cholesterol Accumulation in Cells and Atherosclerosis Progression. Int J Mol Sci 2023; 24:ijms24076426. [PMID: 37047399 PMCID: PMC10094347 DOI: 10.3390/ijms24076426] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/21/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023] Open
Abstract
Atherosclerosis is the most common cardiovascular disease and is the number one cause of death worldwide. Today, atherosclerosis is a multifactorial chronic inflammatory disease with an autoimmune component, accompanied by the accumulation of cholesterol in the vessel wall and the formation of atherosclerotic plaques, endothelial dysfunction, and chronic inflammation. In the process of accumulation of atherogenic lipids, cells of the immune system, such as monocytes, macrophages, dendritic cells, etc., play an important role, producing and/or activating the production of various cytokines—interferons, interleukins, chemokines. In this review, we have tried to summarize the most important cytokines involved in the processes of atherogenesis.
Collapse
|
35
|
La Rocca G, Ferro F, Baldini C, Libra A, Sambataro D, Colaci M, Malatino L, Palmucci S, Vancheri C, Sambataro G. Targeting intracellular pathways in idiopathic inflammatory myopathies: A narrative review. Front Med (Lausanne) 2023; 10:1158768. [PMID: 36993798 PMCID: PMC10040547 DOI: 10.3389/fmed.2023.1158768] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 02/27/2023] [Indexed: 03/16/2023] Open
Abstract
In recent decades, several pieces of evidence have drawn greater attention to the topic of innate immunity, in particular, interferon (IFN) and Interleukin 6 in the pathogenesis of idiopathic inflammatory myopathies (IIM). Both of these molecules transduce their signal through a receptor coupled with Janus kinases (JAK)/signal transducer and activator of transcription proteins (STAT). In this review, we discuss the role of the JAK/STAT pathway in IIM, evaluate a possible therapeutic role for JAK inhibitors in this group of diseases, focusing on those with the strongest IFN signature (dermatomyositis and antisynthetase syndrome).
Collapse
Affiliation(s)
- Gaetano La Rocca
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Francesco Ferro
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Chiara Baldini
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Alessandro Libra
- Regional Referral Centre for Rare Lung Disease, Azienda Ospedaliero Universitaria Policlinico “G. Rodolico-San Marco”, University of Catania, Catania, Italy
| | | | - Michele Colaci
- Internal Medicine Unit, Rheumatology Clinic, Azienda Ospedaliera per l’Emergenza Cannizzaro, University of Catania, Catania, Italy
| | - Lorenzo Malatino
- Internal Medicine Unit, Rheumatology Clinic, Azienda Ospedaliera per l’Emergenza Cannizzaro, University of Catania, Catania, Italy
| | - Stefano Palmucci
- Department of Medical Surgical Sciences and Advanced Technologies “GF Ingrassia”, University Hospital Policlinico “G. Rodolico-San Marco”, Catania, Italy
| | - Carlo Vancheri
- Regional Referral Centre for Rare Lung Disease, Azienda Ospedaliero Universitaria Policlinico “G. Rodolico-San Marco”, University of Catania, Catania, Italy
| | - Gianluca Sambataro
- Regional Referral Centre for Rare Lung Disease, Azienda Ospedaliero Universitaria Policlinico “G. Rodolico-San Marco”, University of Catania, Catania, Italy
- Artroreuma S.R.L., Rheumatology Outpatient Clinic, Catania, Italy
- *Correspondence: Gianluca Sambataro,
| |
Collapse
|
36
|
Zhao X, Si S. Five genes as diagnostic biomarkers of dermatomyositis and their correlation with immune cell infiltration. Front Immunol 2023; 14:1053099. [PMID: 36742332 PMCID: PMC9889851 DOI: 10.3389/fimmu.2023.1053099] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 01/03/2023] [Indexed: 01/19/2023] Open
Abstract
Background Dermatomyositis (DM) is a rare autoimmune disease characterized by severe muscle dysfunction, and the immune response of the muscles plays an important role in the development of DM. Currently, the diagnosis of DM relies on symptoms, physical examination, and biopsy techniques. Therefore, we used machine learning algorithm to screen key genes, and constructed and verified a diagnostic model composed of 5 key genes. In terms of immunity, The relationship between 5 genes and immune cell infiltration in muscle samples was analyzed. These diagnostic and immune-cell-related genes may contribute to the diagnosis and treatment of DM. Methods GSE5370 and GSE128470 datasets were utilised from the Gene Expression Omnibus database as DM test sets. And we also used R software to merge two datasets and to analyze the results of differentially expressed genes (DEGs) and functional correlation analysis. Then, we could detect diagnostic genes adopting least absolute shrinkage and selection operator (LASSO) logistic regression and support vector machine recursive feature elimination (SVM-RFE) analyses. The validity of putative biomarkers was assessed using the GSE1551 dataset, and we confirmed the area under the receiver operating characteristic curve (AUC) values. Finally, CIBERSORT was used to evaluate immune cell infiltration in DM muscles and the correlations between disease-related biomarkers and immune cells. Results In this study, a total of 414 DEGs were screened. ISG15, TNFRSF1A, GUSBP11, SERPINB1 and PTMA were identified as potential DM diagnostic biomarkers(AUC > 0.85),and the expressions of 5 genes in DM group were higher than that in healthy group (p < 0.05). Immune cell infiltration analyses indicated that identified DM diagnostic biomarkers may be associated with M1 macrophages, activated NK cells, Tfh cells, resting NK cells and Treg cells. Conclusion The study identified that ISG15, TNFRSF1A, GUSBP11, SERPINB1 and PTMA as potential diagnostic biomarkers of DM and these genes were closely correlated with immune cell infiltration.This will contribute to future studies in diagnosis and treatment of DM.
Collapse
|
37
|
Predictors of progression in idiopathic inflammatory myopathies with interstitial lung disease. J Transl Int Med 2022. [PMID: 37533847 PMCID: PMC10393057 DOI: 10.2478/jtim-2022-0029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Abstract
The idiopathic inflammatory myopathies (IIMs) are a group of connective tissue diseases that afect multiple organ systems, including the lungs. Interstitial lung disease (ILD) is the most common and heterogeneous complication of IIMs, with its degree ranging from mild to fatal. Thus, it is critical to identify clinical features and validated biomarkers for predicting disease progression and prognosis, which could be beneficial for therapy adjustment. In this review, we discuss predictors for rapid progression of IIM-ILD and propose guidance for disease monitoring and implications of therapy. Systematic screening of myositis-specific antibodies, measuring serum biomarker levels, pulmonary function tests, and chest high-resolution computer tomography will be beneficial for the evaluation of disease progression and prognosis.
Collapse
|
38
|
Li M, Yin F, He Y, Zhang X, Yu H, Yang Q, Xie X, Wei G, Chen H, He C, He Y, Chen J. Bibliometric and altmetric analyses of publication activity in the field of idiopathic inflammatory myopathy in 2011-2021 based on web of science core collection databases. Rheumatol Int 2022; 43:1121-1133. [PMID: 36167928 DOI: 10.1007/s00296-022-05211-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 09/10/2022] [Indexed: 11/28/2022]
Abstract
Idiopathic inflammatory myopathy is a multisystem autoimmune condition characterized by muscle inflammation (myositis) and interstitial lung disease (ILD). Bibliometric analysis and altmetric analysis are effective methods of evaluating academic productivity and measuring the influence of publications. The purpose of this study is to analyze the research productivity and influence of idiopathic inflammatory myopathy articles using bibliometric analysis and altmetric analysis. Data from articles published between 2011 and 2021 were obtained from the Web of Science. Altmetric attention scores and Mendeley reader counts of the articles were obtained from altmetric evaluations. In the study, 2060 articles related to idiopathic inflammatory myopathy were screened out showing an increasing trend in general. In terms of the origin countries of articles, the United States (n = 467, 22.67%) ranked first. Johns Hopkins University has the largest number of institutions (n = 113). The journals regarding idiopathic inflammatory myopathy and Rheumatology (n = 87) published the most articles. The most cited article was published by Mammen et al. and was related to autoantibodies. Dermatomyositis, polymyositis and other research hotspots were represented by keywords. The results of the altmetric analysis showed that citations, impact factors and h-index were significantly correlated with Altmetric Attention Scores and Mendeley Readers Count (P < 0.05). In summary, bibliometric analysis summarizes the current status of idiopathic inflammatory myopathy research and helps to understand the development of idiopathic inflammatory myopathy in the field between 2011 and 2021. Altmetric analysis was used to evaluate the academic and social influence of articles from the novel perspective of internet attention.
Collapse
Affiliation(s)
- Mengxiang Li
- Department of Rheumatology and Immunology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Feng Yin
- Department of Rheumatology and Immunology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Youxian He
- Department of Rheumatology and Immunology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Xue Zhang
- Department of Rheumatology and Immunology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Hao Yu
- Department of Rheumatology and Immunology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Qiuyu Yang
- Department of Rheumatology and Immunology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Xintong Xie
- Department of Rheumatology and Immunology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Guangliang Wei
- Department of Rheumatology and Immunology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Huidong Chen
- Department of Rheumatology and Immunology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Chengsong He
- Department of Rheumatology and Immunology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Yue He
- Department of Ophthalmology, The Affiliated Hospital of Southwest Medical University, Luzhou, People's Republic of China
| | - Jie Chen
- Department of Rheumatology and Immunology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China.
| |
Collapse
|
39
|
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: 2.0] [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
|
40
|
Skeletal muscle provides the immunological micro-milieu for specific plasma cells in anti-synthetase syndrome-associated myositis. Acta Neuropathol 2022; 144:353-372. [PMID: 35612662 PMCID: PMC9288384 DOI: 10.1007/s00401-022-02438-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 04/08/2022] [Accepted: 05/13/2022] [Indexed: 11/17/2022]
Abstract
Anti-synthetase syndrome (ASyS)-associated myositis is a major subgroup of the idiopathic inflammatory myopathies (IIM) and is characterized by disease chronicity with musculoskeletal, dermatological and pulmonary manifestations. One of eight autoantibodies against the aminoacyl-transferase RNA synthetases (ARS) is detectable in the serum of affected patients. However, disease-specific therapeutic approaches have not yet been established.To obtain a deeper understanding of the underlying pathogenesis and to identify putative therapeutic targets, we comparatively investigated the most common forms of ASyS associated with anti-PL-7, anti-PL-12 and anti-Jo-1. Our cohort consisted of 80 ASyS patients as well as healthy controls (n = 40), diseased controls (n = 40) and non-diseased controls (n = 20). We detected a reduced extent of necrosis and regeneration in muscle biopsies from PL-12+ patients compared to Jo-1+ patients, while PL-7+ patients had higher capillary dropout in biopsies of skeletal muscle. Aside from these subtle alterations, no significant differences between ASyS subgroups were observed. Interestingly, a tissue-specific subpopulation of CD138+ plasma cells and CXCL12+/CXCL13+CD20+ B cells common to ASyS myositis were identified. These cells were localized in the endomysium associated with alkaline phosphatase+ activated mesenchymal fibroblasts and CD68+MHC-II+CD169+ macrophages. An MHC-I+ and MHC-II+ MxA negative type II interferon-driven milieu of myofiber activation, topographically restricted to the perifascicular area and the adjacent perimysium, as well as perimysial clusters of T follicular helper cells defined an extra-medullary immunological niche for plasma cells and activated B cells. Consistent with this, proteomic analyses of muscle tissues from ASyS patients demonstrated alterations in antigen processing and presentation. In-depth immunological analyses of peripheral blood supported a B-cell/plasma-cell-driven pathology with a shift towards immature B cells, an increase of B-cell-related cytokines and chemokines, and activation of the complement system. We hypothesize that a B-cell-driven pathology with the presence and persistence of a specific subtype of plasma cells in the skeletal muscle is crucially involved in the self-perpetuating chronicity of ASyS myositis. This work provides the conceptual framework for the application of plasma-cell-targeting therapies in ASyS myositis.
Collapse
|
41
|
Damian L, Login CC, Solomon C, Belizna C, Encica S, Urian L, Jurcut C, Stancu B, Vulturar R. Inclusion Body Myositis and Neoplasia: A Narrative Review. Int J Mol Sci 2022; 23:ijms23137358. [PMID: 35806366 PMCID: PMC9266341 DOI: 10.3390/ijms23137358] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/28/2022] [Accepted: 06/28/2022] [Indexed: 02/04/2023] Open
Abstract
Inclusion body myositis (IBM) is an acquired, late-onset inflammatory myopathy, with both inflammatory and degenerative pathogenesis. Although idiopathic inflammatory myopathies may be associated with malignancies, IBM is generally not considered paraneoplastic. Many studies of malignancy in inflammatory myopathies did not include IBM patients. Indeed, IBM is often diagnosed only after around 5 years from onset, while paraneoplastic myositis is generally defined as the co-occurrence of malignancy and myopathy within 1 to 3 years of each other. Nevertheless, a significant association with large granular lymphocyte leukemia has been recently described in IBM, and there are reports of cancer-associated IBM. We review the pathogenic mechanisms supposed to be involved in IBM and outline the common mechanisms in IBM and malignancy, as well as the therapeutic perspectives. The terminally differentiated, CD8+ highly cytotoxic T cells expressing NK features are central in the pathogenesis of IBM and, paradoxically, play a role in some cancers as well. Interferon gamma plays a central role, mostly during the early stages of the disease. The secondary mitochondrial dysfunction, the autophagy and cell cycle dysregulation, and the crosstalk between metabolic and mitogenic pathways could be shared by IBM and cancer. There are intermingled subcellular mechanisms in IBM and neoplasia, and probably their co-existence is underestimated. The link between IBM and cancers deserves further interest, in order to search for efficient therapies in IBM and to improve muscle function, life quality, and survival in both diseases.
Collapse
Affiliation(s)
- Laura Damian
- Centre for Rare Autoimmune and Autoinflammatory Diseases (ERN-ReCONNET), Department of Rheumatology, Emergency Clinical County Hospital Cluj, 400347 Cluj-Napoca, Romania;
- CMI Reumatologie Dr. Damian, 6-8 Petru Maior St., 400002 Cluj-Napoca, Romania
| | - Cristian Cezar Login
- Department of Physiology, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania
- Correspondence:
| | - Carolina Solomon
- Radiology Department, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania;
- Radiology Department, Emergency Clinical County Hospital Cluj, 400006 Cluj-Napoca, Romania
| | - Cristina Belizna
- UMR CNRS 6015—INSERM U1083, University of Angers, 49100 Angers, France;
- Internal Medicine Department Clinique de l’Anjou, Angers and Vascular and Coagulation Department, University Hospital Angers, 49100 Angers, France
| | - Svetlana Encica
- Department of Pathology, “Niculae Stancioiu” Heart Institute Cluj-Napoca, 19-21 Calea Moților St., 400001 Cluj-Napoca, Romania;
| | - Laura Urian
- Department of Hematology, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400004 Cluj-Napoca, Romania;
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, 400014 Cluj-Napoca, Romania
| | - Ciprian Jurcut
- Department of Internal Medicine, “Carol Davila” Central Military Emergency University Hospital, Calea Plevnei No 134, 010825 Bucharest, Romania;
| | - Bogdan Stancu
- 2nd Surgical Department, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Romana Vulturar
- Department of Molecular Sciences, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
- Cognitive Neuroscience Laboratory, University “Babes-Bolyai” Cluj-Napoca, 400294 Cluj-Napoca, Romania
| |
Collapse
|
42
|
Merve A, Schneider U, Kara E, Papadopoulou C, Stenzel W. Muscle biopsy in myositis: What the rheumatologist needs to know. Best Pract Res Clin Rheumatol 2022; 36:101763. [PMID: 35773136 DOI: 10.1016/j.berh.2022.101763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The appropriate analysis of skeletal muscle tissues is a key element in many diagnostic procedures and can deliver valuable information about the organ that is affected. Although arguably the frequency of muscle biopsy may be declining in certain domains where genetic analysis is now the first line of diagnostic evaluation, it still has an important role in assessment of patients with neuromuscular disorders such as congenital myopathies, muscular dystrophies, metabolic and inflammatory diseases. Here, we have comprehensively discussed the aspects of a modern and fruitful approach to muscle biopsy histopathological studies in rheumatological disorders. We have focussed on the neuromuscular involvement in myositis and its differential diagnoses in both adult and paediatric settings. We have also covered the clinical indications for the biopsy, technical aspects and practical points relevant for the rheumatologists. Finally, we have critically discussed the current and future opportunities that a muscle biopsy may offer and its limitations.
Collapse
Affiliation(s)
- Ashirwad Merve
- Department of Neuropathology, UCL Institute of Neurology, National Hospital for Neurology and Neurosurgery, University College London Hospitals, London, UK; Department of Histopathology, Great Ormond Street Hospital for Children, London, UK
| | - Udo Schneider
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology, Charitéplatz 1, 10117 Berlin, Germany
| | - Eleanna Kara
- Department of Neuropathology, UCL Institute of Neurology, National Hospital for Neurology and Neurosurgery, University College London Hospitals, London, UK
| | | | - Werner Stenzel
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Neurology, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
43
|
Vaughn LS, Frederick K, Burnett SB, Sharma N, Bragg DC, Camargos S, Cardoso F, Patel RC. DYT- PRKRA Mutation P222L Enhances PACT's Stimulatory Activity on Type I Interferon Induction. Biomolecules 2022; 12:713. [PMID: 35625640 PMCID: PMC9138762 DOI: 10.3390/biom12050713] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 12/10/2022] Open
Abstract
DYT-PRKRA (dystonia 16 or DYT-PRKRA) is caused by mutations in the PRKRA gene that encodes PACT, the protein activator of interferon (IFN)-induced double-stranded (ds) RNA-activated protein kinase (PKR). PACT participates in several cellular pathways, of which its role as a PKR activator protein during integrated stress response (ISR) is the best characterized. Previously, we have established that the DYT-PRKRA mutations cause enhanced activation of PKR during ISR to sensitize DYT-PRKRA cells to apoptosis. In this study, we evaluate if the most prevalent substitution mutation reported in DYT-PRKRA patients alters PACT's functional role in induction of type I IFNs via the retinoic acid-inducible gene I (RIG-I) signaling. Our results indicate that the P222L mutation augments PACT's ability to induce IFN β in response to dsRNA and the basal expression of IFN β and IFN-stimulated genes (ISGs) is higher in DYT-PRKRA patient cells compared to cells from the unaffected controls. Additionally, IFN β and ISGs are also induced at higher levels in DYT-PRKRA cells in response to dsRNA. These results offer a new avenue for investigations directed towards understanding the underlying molecular pathomechanisms in DYT-PRKRA.
Collapse
Affiliation(s)
- Lauren S. Vaughn
- Department of Biological Sciences, University of South Carolina, 700 Sumter Street, Columbia, SC 29208, USA; (L.S.V.); (K.F.); (S.B.B.)
| | - Kenneth Frederick
- Department of Biological Sciences, University of South Carolina, 700 Sumter Street, Columbia, SC 29208, USA; (L.S.V.); (K.F.); (S.B.B.)
| | - Samuel B. Burnett
- Department of Biological Sciences, University of South Carolina, 700 Sumter Street, Columbia, SC 29208, USA; (L.S.V.); (K.F.); (S.B.B.)
| | - Nutan Sharma
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA; (N.S.); (D.C.B.)
| | - D. Cristopher Bragg
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA; (N.S.); (D.C.B.)
| | - Sarah Camargos
- Department of Internal Medicine, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil; (S.C.); (F.C.)
| | - Francisco Cardoso
- Department of Internal Medicine, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil; (S.C.); (F.C.)
| | - Rekha C. Patel
- Department of Biological Sciences, University of South Carolina, 700 Sumter Street, Columbia, SC 29208, USA; (L.S.V.); (K.F.); (S.B.B.)
| |
Collapse
|
44
|
Zeng R, Glaubitz S, Schmidt J. Antibody Therapies in Autoimmune Inflammatory Myopathies: Promising Treatment Options. Neurotherapeutics 2022; 19:911-921. [PMID: 35394612 PMCID: PMC9294121 DOI: 10.1007/s13311-022-01220-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2022] [Indexed: 12/29/2022] Open
Abstract
Inflammatory myopathies, including polymyositis (PM), dermatomyositis (DM), inclusion body myositis (IBM), necrotizing myopathy (NM), antisynthetase syndrome (ASS) and overlap myositis (OM), in short myositis, are rare diseases. All forms of myositis have progressive muscle weakness in common, with each subtype characterized by different autoantibody profiles, histological findings and extramuscular manifestations. Due to better understanding of the pathogenesis of the muscle inflammation in myositis, new molecular pathways for targeted therapy have been discovered. Current therapies aim at different components of the innate or the adaptive immune response. Additionally, non-inflammatory mechanisms in myositis have come into focus as possible treatment targets. The use of therapeutical antibodies in myositis has been examined in various clinical studies, several of them randomized controlled ones: Depletion of B-cells by rituximab has been established as treatment of refractory myositis. IVIG, an antibody therapy in the wider sense, has now been licensed for DM following a recent positive clinical trial. Negative study results were reported in randomized trials with infliximab, sifalimumab and bimagrumab. Studies on basiliximab and eculizumab are currently underway, and are expected to yield results in a couple of years. Despite some promising results of clinical studies with antibody therapy in myositis, further research is crucial to optimize the treatment for this debilitating disease and to find treatment alternatives for treatment-refractory patients.
Collapse
Affiliation(s)
- Rachel Zeng
- Muscle Immunobiology Group, Neuromuscular Center, Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Stefanie Glaubitz
- Muscle Immunobiology Group, Neuromuscular Center, Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Jens Schmidt
- Muscle Immunobiology Group, Neuromuscular Center, Department of Neurology, University Medical Center Göttingen, Göttingen, Germany.
- Department of Neurology and Pain Treatment, Immanuel Klinik Rüdersdorf, University Hospital of the Brandenburg Medical School Theodor Fontane, Rüdersdorf bei Berlin, Germany.
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, Rüdersdorf bei Berlin, Germany.
| |
Collapse
|
45
|
Chen D, Huang W, Zhongjie W, Feifeng R, Luo L, Jun Z, Dongmei H, Tian M, Tang L. OUP accepted manuscript. Rheumatology (Oxford) 2022; 61:e221-e223. [PMID: 35148369 DOI: 10.1093/rheumatology/keac084] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/15/2022] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Dandan Chen
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wenhan Huang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wang Zhongjie
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ren Feifeng
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lei Luo
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhou Jun
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Huang Dongmei
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mengxue Tian
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lin Tang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
46
|
Morphological Characteristics of Idiopathic Inflammatory Myopathies in Juvenile Patients. Cells 2021; 11:cells11010109. [PMID: 35011672 PMCID: PMC8750180 DOI: 10.3390/cells11010109] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/20/2021] [Accepted: 12/25/2021] [Indexed: 12/16/2022] Open
Abstract
Background: In juvenile idiopathic inflammatory myopathies (IIMs), morphological characteristic features of distinct subgroups are not well defined. New treatment strategies require a precise diagnosis of the subgroups in IIM, and, therefore, knowledge about the pathomorphology of juvenile IIMs is warranted. Methods: Muscle biopsies from 15 patients (median age 8 (range 3–17) years, 73% female) with IIM and seven controls were analyzed by standard methods, immunohistochemistry, and transmission electron microscopy (TEM). Detailed clinical and laboratory data were accessed retrospectively. Results: Proximal muscle weakness and skin symptoms were the main clinical symptoms. Dermatomyositis (DM) was diagnosed in 9/15, antisynthetase syndrome (ASyS) in 4/15, and overlap myositis (OM) in 2/15. Analysis of skeletal muscle tissues showed inflammatory cells and diffuse upregulation of MHC class I in all subtypes. Morphological key findings were COX-deficient fibers as a striking pathology in DM and perimysial alkaline phosphatase positivity in anti-Jo-1-ASyS. Vascular staining of the type 1 IFN-surrogate marker, MxA, correlated with endothelial tubuloreticular inclusions in both groups. None of these specific morphological findings were present in anti-PL7-ASyS or OM patients. Conclusions: Morphological characteristics discriminate IIM subtypes in juvenile patients, emphasizing differences in aetiopathogenesis and supporting the notion of individual and targeted therapeutic strategies.
Collapse
|
47
|
Grochowska J, Czerwinska J, Borowski LS, Szczesny RJ. Mitochondrial RNA, a new trigger of the innate immune system. WILEY INTERDISCIPLINARY REVIEWS-RNA 2021; 13:e1690. [PMID: 34498404 DOI: 10.1002/wrna.1690] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 02/06/2023]
Abstract
Mitochondria play a pivotal role in numerous cellular processes. One of them is regulation of the innate immune pathway. In this instance, mitochondria function in two different aspects of regulatory mechanisms. First, mitochondria are part of the antiviral signaling cascade that is triggered in the cytoplasm and transmitted to effector proteins through mitochondria-localized proteins. Second, mitochondria can become an endogenous source of innate immune stimuli. Under some pathophysiological conditions, mitochondria release to the cytoplasm immunogenic factors, such as mitochondrial nucleic acids. Here, we focus on immunogenic mitochondrial double-stranded RNA (mt-dsRNA) and its origin and metabolism. We discuss factors that are responsible for regulating mt-dsRNA and its escape from mitochondria, emphasizing the contribution of polynucleotide phosphorylase (PNPase, PNPT1). Finally, we review current knowledge of the role of PNPase in human health and disease. This article is categorized under: RNA in Disease and Development > RNA in Disease.
Collapse
Affiliation(s)
- Joanna Grochowska
- Institute of Biochemistry and Biophysics Polish Academy of Sciences, Warsaw, Poland
| | - Jolanta Czerwinska
- Institute of Biochemistry and Biophysics Polish Academy of Sciences, Warsaw, Poland
| | - Lukasz S Borowski
- Faculty of Biology, Institute of Genetics and Biotechnology, University of Warsaw, Warsaw, Poland
| | - Roman J Szczesny
- Institute of Biochemistry and Biophysics Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
48
|
Stenzel W, Goebel HH. Recent data and developments in myositis. Brain Pathol 2021; 31:e12966. [PMID: 34043261 PMCID: PMC8412128 DOI: 10.1111/bpa.12966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 04/01/2021] [Indexed: 10/25/2022] Open
Affiliation(s)
- Werner Stenzel
- Department of Neuropathology, Charite Universitätsmedizin, Berlin, Germany
| | - Hans H Goebel
- Department of Neuropathology, Charite Universitätsmedizin, Berlin, Germany
| |
Collapse
|