Skip to main content

EDITORIAL article

Front. Neurol., 08 August 2023
Sec. Neuromuscular Disorders and Peripheral Neuropathies
This article is part of the Research Topic Inflammatory Muscle Diseases: An Update View all 5 articles

Editorial: Inflammatory muscle diseases: an update

  • 1Department of Pathology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
  • 2University of Notre Dame Australia, Fremantle, WA, Australia
  • 3Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, WA, Australia
  • 4Department of Neurology, Fiona Stanley Hospital, Murdoch, WA, Australia
  • 5Department of Neurology, School of Medicine, University of California, Irvine, Irvine, CA, United States
  • 6Pathology and Laboratory Medicine, School of Medicine, University of California, Irvine, Irvine, CA, United States
  • 7School of Medicine, The Institute for Immunology, University of California, Irvine, Irvine, CA, United States
  • 8Department of Neuropathology, Berlin Institute of Health (BIH), Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
  • 9Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
  • 10Department of Genome Medicine Development, Medical Genome Center, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
  • 11Department of Clinical Genome Analysis, Medical Genome Center, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan

Editorial on the Research Topic
Inflammatory muscle diseases: an update

The evolution of classification of the idiopathic inflammatory myopathies (IIM) is fueled by myositis-specific antibodies (MSA), clinicopathological features, and discoveries in the “-omics” fields such as proteomics and transcriptomics. The major IIM in the current classification include dermatomyositis (DM), immune-mediated necrotizing myopathy (IMNM), antisynthetase syndrome (ASS), and inclusion body myositis (IBM) (1, 2). SARS-CoV-2 (COVID-19) infection-associated myopathy and immune checkpoint inhibitors-related myositis (iR-myositis) attract attention as emerging entities. The existence of polymyositis (PM) among the IIM has become controversial because most “cases” can be re-classified into other IIM, predominantly IMNM and ASS, using serological-clinical-pathological variables (3). Interestingly, PM with mitochondrial pathology (PM-Mito), which contains features resembling IBM except for rimmed vacuoles (i.e., endomysial inflammation, mitochondrial pathology, highly differentiated cytotoxic T cells, and type 2 interferon (IFN2, IFN-γ) pathway upregulation) has been re-recognized and proposed as early IBM (eIBM) in IBM-spectrum disease (4, 5). Many of the IIM are paradigmatically associated with certain MSA, i.e., DM with DM specific antibodies (DMSAs) including anti-TIF1-γ, anti-Mi-2, anti-MDA5, anti-NXP-2, and anti-SAE antibodies; IMNM with anti-SRP and anti-HMGCR antibodies; ASS with anti-tRNA synthetase antibodies including anti-Jo-1, anti-OJ, anti-PL-7, anti-PL-12, anti-EJ, anti-KS, anti-Zo, anti-Ha, and anti-Ly antibodies; and IBM with anti-cN1A antibody. The clinicopathological features associated with the same MSA in adult patients could be different from juvenile patients (age < 18 years old); this is particularly true for jDM with TIF1-γ where cancer is never associated with the disease in contrast to adults where cancer association accounts for approximately 50% of patients. The serological-clinical-pathological features in association with human leukocyte antigen (HLA) haplotypes in different IIM subtypes have been summarized elsewhere (1, 2, 613).

Muscle imaging can assist and complement IIM diagnosis and long-term management. The most common modalities, including magnetic resonance imaging (MRI), ultrasonography, and positron emission tomography (PET) can identify typical patterns of individual skeletal muscle involvement and disease activity (1416). Patterns of fat replacement and tissue edema recognized by T1-weighted and T2-weighted fat suppressed/short tau inversion recovery (STIR) MRI sequences are helpful for diagnosis. The Dixon MRI sequence measures muscle fat fraction, which quantitatively reflects disease activity and progression. Ultrasonography is more affordable and more practical for bedside evaluation, but its performance largely depends on the user's skills. Furthermore, it can only evaluate the superficial muscle layers. PET using different tracers specific for corresponding cellular mechanisms can demonstrate disease activity for each IIM. Fluorine-18 (18F)-labeled fluorodeoxyglucose PET (18FDG-PET) can identify patterns of muscle involvement, and detect concurrent interstitial lung disease and malignancy in IIM via glucose metabolism (17). 11C-labeled Pittsburgh compound B-PET (PIB-PET) (18, 19) and 18F-Flobetapir PET identify amyloid-beta in IBM (20). The use of PET imaging that utilizes a CD8 T lymphocyte-based ligand shows tremendous potential in preliminary studies in IBM (21). Electrical impedance myography (EIM), although mainly present in research settings, is a promising portable device that can operate in bedside settings. EIM quantitatively assesses disturbances in the electrical properties of diseased muscle using electrical current (22). The article by Zubair et al. summarizes the current state and future direction of these modalities in IIM.

The most prevalent IIM in juvenile patients is juvenile DM (jDM, 75% of juvenile IIM) followed by IMNM (2.9–21% of juvenile IIM) and rarely ASS; IBM is not present in this age group. Without serological information, juvenile IMNM risks being misdiagnosed as jDM due to its association with skin lesions or as inherited myopathy due to insidious muscle weakness and refractoriness to steroid treatment. Misdiagnosis and delayed proper treatment in IMNM likely lead to unreversible muscle damage. A recent article by Wang and Liang raises awareness of and discusses current information on juvenile IMNM.

Characteristic molecular pathway activations and their surrogate immunohistochemical markers have been identified in all major subtypes of IIM except for IMNM. The IFN1 signaling pathway is highly upregulated in DM and associated with sarcoplasmic myxovirus resistance protein A (MxA) expression; the finding is currently recognized as one of the diagnostic criteria for DM (23). The IFN2 signaling pathway is highly upregulated in IBM and to a lesser degree in PM-Mito and ASS; the upregulation can be demonstrated by surrogate markers e.g., HLA-DR (major histocompatibility complex, MHC class II) and guanylate-binding protein (GBP6) (5, 11, 24). Notably, GBP6 IHC positivity in a muscle biopsy containing highly differentiated cytotoxic T cells (highlighted by KLRG1, CD57, and PD1 expression) differentiates IBM from PM-Mito (5). The myopathological features of IMNM are less specific than the other IIM and pose diagnostic challenges in the absence of serology results. Although the upregulation of genes associated with the autophagy-lysosome pathway and diffuse p62 sarcoplasmic staining suggest autophagy pathway alteration in IMNM, the pattern can also present in non-immune mediated rhabdomyolysis (25, 26). In IMNM, muscle RING-finger protein-1 (MuRF1, a muscle-specific E3 ubiquitin ligase-associated muscle atrophy gene) is upregulated in atrophic myotubes induced by in vitro anti-SRP and anti-HMGCR antibodies treatment (27). Although MuRF1 is upregulated in multiple muscle atrophy models and MuRF1 knockout mice were not affected by glucocorticoids-induced atrophy (28, 29), Yang et al. recently demonstrated sarcoplasmic MuRF1 expression in regenerating myofibers of IMNM and DM but not in atrophic fiber. Whether MuRF1 upregulation in IMNM and DM is associated with mechanisms different from in vitro muscle atrophy models and whether the expression can be useful in IMNM diagnosis needs further exploration.

COVID-19-associated myopathy is likely caused by an immune-mediated mechanism rather than direct SAR-CoV-2 infection. This speculation is supported by autopsy studies in critically ill COVID-19 patients with myositis symptoms showing prominent myopathology without viral protein expression (30, 31) and an absence of angiotensin-converting enzyme 2 (ACE2, the protein essential for viral entry) expression on skeletal muscle (32). DM, IMNM, and ASS developing during or after SAR-CoV-2 infection has been observed. A recent case report by Niedzielska et al.. expands the spectrum of concurrent anti-Mi-2 DM in patients with SARS-CoV-2 infection whose condition improved after corticosteroid therapy. Whether SARS-CoV-2 infection directly induced myositis or triggered pre-existing asymptomatic conditions in these single case reports are debatable.

iR-myositis (in the context of immune checkpoint inhibition; ICI) is often associated with oculobulbar weakness, myocarditis, anti-acetylcholine receptor (anti-AChR) antibody, anti-striational antibodies, and MSA (3335). This entity is frequently mistaken for myasthenia gravis. A transcriptomic study has classified ICIs-myositis into ICI-MYO1 associated with myocarditis and prominent inflammation in muscle biopsy; ICI-MYO2 with mild inflammation in muscle biopsy; and ICI-DM with DM myopathology, anti-TIF1-γ antibody, and IFN1 pathway upregulation (35). IFN2 and interleukin 6 (IL6) pathways are highly upregulated in ICI-MYO1 and ICI-DM. Since targeting the IL6 pathway is reported to improve other ICI-triggered immune adverse events, it could be useful for ICIs-myositis treatment.

Conclusion

IIM consists of evolving heterogeneous entities which require multimodality approaches for diagnosis and appropriate management.

Author contributions

JT: Conceptualization, Writing—original draft. MN: Writing—review and editing. TM: Writing—review and editing. WS: Writing—review and editing. IN: Writing—review and editing.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher's note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Tanboon J, Uruha A, Stenzel W, Nishino I. Where are we moving in the classification of idiopathic inflammatory myopathies? Curr Opin Neurol. (2020) 33:590–603. doi: 10.1097/WCO.0000000000000855

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Lundberg IE, Fujimoto M, Vencovsky J, Aggarwal R, Holmqvist M, Christopher-Stine L, et al. Idiopathic inflammatory myopathies. Nat Rev Dis Primers. (2021) 7:86. doi: 10.1038/s41572-021-00321-x

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Mariampillai K, Granger B, Amelin D, Guiguet M, Hachulla E, Maurier F, et al. Development of a New Classification System for Idiopathic Inflammatory Myopathies Based on Clinical Manifestations and Myositis-Specific Autoantibodies. JAMA Neurol. (2018) 75:1528–37. doi: 10.1001/jamaneurol.2018.2598

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Blume G, Pestronk A, Frank B, Johns DR. Polymyositis with cytochrome oxidase negative muscle fibres. Early quadriceps weakness and poor response to immunosuppressive therapy. Brain. (1997) 120:39–45. doi: 10.1093/brain/120.1.39

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Kleefeld F, Uruha A, Schänzer A, Nishimura A, Roos A, Schneider U, et al. Morphologic and molecular patterns of polymyositis with mitochondrial pathology and inclusion body myositis. Neurology. (2022) 99:e2212–22. doi: 10.1212/WNL.0000000000201103

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Greenberg SA. Inclusion body myositis: clinical features and pathogenesis. Nat Rev Rheumatol. (2019) 15:257–72. doi: 10.1038/s41584-019-0186-x

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Greenberg SA, Pinkus JL, Kong SW, Baecher-Allan C, Amato AA, Dorfman DM. Highly differentiated cytotoxic T cells in inclusion body myositis. Brain. (2019) 142:2590–604. doi: 10.1093/brain/awz207

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Tanboon J, Inoue M, Saito Y, Tachimori H, Hayashi S, Noguchi S, et al. Dermatomyositis: Muscle Pathology According to Antibody Subtypes. Neurology. (2022) 98:e739–49. doi: 10.1212/WNL.0000000000013176

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Preuße C, Paesler B, Nelke C, Cengiz D, Müntefering T, Roos A, et al. Skeletal muscle provides the immunological micro-milieu for specific plasma cells in anti-synthetase syndrome-associated myositis. Acta Neuropathol. (2022) 144:353–72. doi: 10.1007/s00401-022-02438-z

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Vulsteke JB, Derua R, Dubucquoi S, Coutant F, Sanges S, Goncalves D, et al. Mass spectrometry-based identification of new anti-Ly and known antisynthetase autoantibodies. Ann Rheum Dis. (2022). 2022:222686. doi: 10.1136/ard-2022-222686

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Tanboon J, Inoue M, Hirakawa S, Tachimori H, Hayashi S, Noguchi S, et al. Muscle pathology of antisynthetase syndrome according to antibody subtypes. Brain Pathol. (2023) 33:e13155. doi: 10.1111/bpa.13155

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Papadopoulou C, Chew C, Wilkinson MGL, McCann L, Wedderburn LR. Juvenile idiopathic inflammatory myositis: an update on pathophysiology and clinical care. Nat Rev Rheumatol. (2023) 19:343–62. doi: 10.1038/s41584-023-00967-9

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Rothwell S, Chinoy H, Lamb JA. Genetics of idiopathic inflammatory myopathies: insights into disease pathogenesis. Curr Opin Rheumatol. (2019) 31:611–6. doi: 10.1097/BOR.0000000000000652

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Walter AW, Lim J, Raaphorst J, Smithuis FF, den Harder JM, Eftimov F, et al. Ultrasound and MR muscle imaging in new onset idiopathic inflammatory myopathies at diagnosis and after treatment: a comparative pilot study. Rheumatology (Oxford). (2022) 62:300–9. doi: 10.1093/rheumatology/keac263

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Paramalingam S, Needham M, Bulsara M, Mastaglia FL, Keen HI. The longitudinal study of muscle changes with ultrasound: Differential changes in idiopathic inflammatory myopathy subgroups. Rheumatology. (2023). 24:kead239. doi: 10.1093/rheumatology/kead239

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Bentick G, Fairley J, Nadesapillai S, Wicks I, Day J. Defining the clinical utility of PET or PET-CT in idiopathic inflammatory myopathies: A systematic literature review. Semin Arthritis Rheum. (2022) 57:152107. doi: 10.1016/j.semarthrit.2022.152107

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Li Y, Zhou Y, Wang Q. Multiple values of 18F-FDG PET/CT in idiopathic inflammatory myopathy. Clin Rheumatol. (2017) 36:2297–305. doi: 10.1007/s10067-017-3794-3

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Maetzler W, Reimold M, Schittenhelm J, Vorgerd M, Bornemann A, Kötter I, et al. Increased [11C]PIB-PET levels in inclusion body myositis are indicative of amyloid beta deposition. J Neurol Neurosurg Psychiatry. (2011) 82:1060–2. doi: 10.1136/jnnp.2009.197640

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Noto YI, Kondo M, Tsuji Y, Matsushima S, Mizuno T, Tokuda T, et al. Diagnostic value of muscle [11C] PIB-PET in inclusion body myositis. Front Neurol. (2020) 10:386. doi: 10.3389/fneur.2019.01386

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Lilleker JB, Hodgson R, Roberts M, Herholz K, Howard J, Hinz R, et al. [18F]Florbetapir positron emission tomography: identification of muscle amyloid in inclusion body myositis and differentiation from polymyositis. Ann Rheum Dis. (2019) 78:657–62. doi: 10.1136/annrheumdis-2018-214644

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Quinn C, Moulton K, Korn R, Le W, Goel N, Farwell M, et al. CD8 Positron Emission Tomography (PET/CT) Imaging with 89Zr-Df-IAB22M2C in Patients with Inclusion Body Myositis. Arthritis Rheumatol. (2021) 73:720–726. doi: 10.2967/jnumed.121.262485

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Tarulli A, Esper GJ, Lee KS, Aaron R, Shiffman CA, Rutkove SB. Electrical impedance myography in the bedside assessment of inflammatory myopathy. Neurology. (2005) 65:451–2. doi: 10.1212/01.wnl.0000172338.95064.cb

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Mammen AL, Allenbach Y, Stenzel W, Benveniste O, ENMC 239th Workshop Study Group. 239th ENMC International Workshop: Classification of dermatomyositis, Amsterdam, the Netherlands, 14-16 December 2018. Neuromuscul Disord. (2020) 30:70–92 doi: 10.1016/j.nmd.2019.10.005

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Pinal-Fernandez I, Casal-Dominguez M, Derfoul A, Pak K, Plotz P, Miller FW, et al. Identification of distinctive interferon gene signatures in different types of myositis. Neurology. (2019) 93:e1193–204. doi: 10.1212/WNL.0000000000008128

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Fischer N, Preuße C, Radke J, Pehl D, Allenbach Y, Schneider U, et al. Sequestosome-1 (p62) expression reveals chaperone-assisted selective autophagy in immune-mediated necrotizing myopathies. Brain Pathol. (2020) 30:261–71. doi: 10.1111/bpa.12772

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Acosta IJ, Stenzel W, Hofer M, Brady S. Autophagy in non-immune-mediated rhabdomyolysis: Assessment of p62 immunohistochemistry. Muscle Nerve. (2023) 67:73–7. doi: 10.1002/mus.27739

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Arouche-Delaperche L, Allenbach Y, Amelin D, Preusse C, Mouly V, Mauhin W, et al. Pathogenic role of anti-signal recognition protein and anti-3-Hydroxy-3-methylglutaryl-CoA reductase antibodies in necrotizing myopathies: Myofiber atrophy and impairment of muscle regeneration in necrotizing autoimmune myopathies. Ann Neurol. (2017) 81:538–48. doi: 10.1002/ana.24902

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Bodine SC, Latres E, Baumhueter S, Lai VK, Nunez L, Clarke BA, et al. Identification of ubiquitin ligases required for skeletal muscle atrophy. Science. (2001) 294:1704–8. doi: 10.1126/science.1065874

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Baehr LM, Furlow JD, Bodine SC. Muscle sparing in muscle RING finger 1 null mice: response to synthetic glucocorticoids. J Physiol. (2011) 589:4759–4776. doi: 10.1113/jphysiol.2011.212845

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Aschman T, Schneider J, Greuel S, Meinhardt J, Streit S, Goebel HH, et al. Association between SARS-CoV-2 Infection and immune-mediated myopathy in patients who have died. JAMA Neurol. (2021) 78:948–60. doi: 10.1001/jamaneurol.2021.2004

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Suh J, Mukerji SS, Collens SI, Padera RF, Pinkus GS, Amato AA, et al. Skeletal muscle and peripheral nerve histopathology in COVID-19. Neurology. (2021) 97:e849–58. doi: 10.1212/WNL.0000000000012344

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Hikmet F, Méar L, Edvinsson Å, Micke P, Uhlén M, Lindskog C. The protein expression profile of ACE2 in human tissues. Mol Syst Biol. (2020) 16:e9610. doi: 10.15252/msb.20209610

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Allenbach Y, Anquetil C, Manouchehri A, Benveniste O, Lambotte O, Lebrun-Vignes B, et al. Immune checkpoint inhibitor-induced myositis, the earliest and most lethal complication among rheumatic and musculoskeletal toxicities. Autoimmun Rev. (2020) 19:102586. doi: 10.1016/j.autrev.2020.102586

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Müller-Jensen L, Knauss S, Ginesta Roque L, Schinke C, Maierhof SK, Bartels F, et al. Autoantibody profiles in patients with immune checkpoint inhibitor-induced neurological immune related adverse events. Front Immunol. (2023) 14:1108116. doi: 10.3389/fimmu.2023.1108116

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Pinal-Fernandez I, Quintana A, Milisenda JC, Casal-Dominguez M, Muñoz-Braceras S, Derfoul A, et al. Transcriptomic profiling reveals distinct subsets of immune checkpoint inhibitor induced myositis. Ann Rheum Dis. (2023) 82:829–36. doi: 10.1136/ard-2022-223792

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: myositis, dermatomyositis, inclusion body myositis, immune mediated necrotizing myopathy, antisynthetase syndrome, imaging, immune checkpoint inhibitor, COVID-19

Citation: Tanboon J, Needham M, Mozaffar T, Stenzel W and Nishino I (2023) Editorial: Inflammatory muscle diseases: an update. Front. Neurol. 14:1259275. doi: 10.3389/fneur.2023.1259275

Received: 15 July 2023; Accepted: 24 July 2023;
Published: 08 August 2023.

Edited and reviewed by: Chiara Terracciano, Gugliemo da Saliceto Hospital, Italy

Copyright © 2023 Tanboon, Needham, Mozaffar, Stenzel and Nishino. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Jantima Tanboon, jantima.tan@mahidol.ac.th

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.