Expression of interferon-regulated genes in juvenile dermatomyositis versus Mendelian autoinflammatory interferonopathies

Hanna Kim, Fatima Gunter-Rahman, John A McGrath, Esther Lee, Adriana A de Jesus, Ira N Targoff, Yan Huang, Terrance P O'Hanlon, Wanxia L Tsai, Massimo Gadina, Frederick W Miller, Raphaela Goldbach-Mansky, Lisa G Rider, Hanna Kim, Fatima Gunter-Rahman, John A McGrath, Esther Lee, Adriana A de Jesus, Ira N Targoff, Yan Huang, Terrance P O'Hanlon, Wanxia L Tsai, Massimo Gadina, Frederick W Miller, Raphaela Goldbach-Mansky, Lisa G Rider

Abstract

Background: Juvenile dermatomyositis (JDM) is a systemic autoimmune disease with a prominent interferon (IFN) signature, but the pathogenesis of JDM and the etiology of its IFN signature remain unknown. The Mendelian autoinflammatory interferonopathies, Chronic Atypical Neutrophilic Dermatosis with Lipodystrophy and Elevated temperature (CANDLE) and STING-Associated Vasculopathy with onset in Infancy (SAVI), are caused by genetic mutations and have extremely elevated IFN signatures thought to drive pathology. The phenotypic overlap of some clinical features of CANDLE and SAVI with JDM led to the comparison of a standardized interferon-regulated gene score (IRG-S) in JDM and myositis-specific autoantibody (MSA) JDM subgroups, with CANDLE and SAVI.

Methods: A peripheral 28-component IRG-S assessed by NanoString™ in 57 JDM patients subtyped by MSA was compared with IRG-S in healthy controls (HC) and CANDLE/SAVI patients. Principal component analysis (PCA) was performed, and individual genes were evaluated for their contribution to the score. IRG-S were correlated with disease assessments and patient characteristics.

Results: IRG-S in JDM patients were significantly higher than in HC but lower than in CANDLE or SAVI. JDM IRG-S overlapped more with SAVI than CANDLE by PCA. Among MSA groups, anti-MDA5 autoantibody-positive patients' IRG-S overlapped most with SAVI. The IFI27 proportion was significantly higher in SAVI and CANDLE than JDM, but IFIT1 contributed more to IRG-S in JDM. Overall, the contribution of individual interferon-regulated genes (IRG) in JDM was more similar to SAVI. IRG-S correlated moderately with JDM disease activity measures (rs = 0.33-0.47) and more strongly with skin activity (rs = 0.58-0.79) in anti-TIF1 autoantibody-positive patients. Weakness and joint disease activity (multinomial OR 0.91 and 3.3) were the best predictors of high IRG-S.

Conclusions: Our findings demonstrate peripheral IRG expression in JDM overlaps with monogenic interferonopathies, particularly SAVI, and correlates with disease activity. Anti-MDA5 autoantibody-positive JDM IRG-S were notably more similar to SAVI. This may reflect both a shared IFN signature, which is driven by IFN-β and STING pathways in SAVI, as well as the shared phenotype of vasculopathy in SAVI and JDM, particularly in anti-MDA5 autoantibody-positive JDM, and indicate potential therapeutic targets for JDM.

Trial registration: ClinicalTrials.gov NCT02974595.

Keywords: (3–10): juvenile dermatomyositis; Biomarkers; Interferon; Interferonopathy; Myositis; Myositis-specific autoantibodies; Pediatric rheumatology.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
Peripheral blood interferon-regulated gene (IRG) scores in juvenile dermatomyositis (JDM) compared to monogenic autoinflammatory disease patients and healthy controls. Median and interquartile ranges are shown. Dotted horizontal line represents 95th percentile of healthy controls [22]. CANDLE (n = 11), SAVI (n = 7), NOMID (n = 18), HC (n = 26). JDM overall (n = 57):*q < 0.05, **q < 0.01. JDM-HQ (n = 14): **p < 0.01. JDM overall and JDM-HQ are higher than NOMID and HC; JDM overall is lower than CANDLE and SAVI. Abbreviations: IRG, interferon-regulated gene; JDM, juvenile dermatomyositis; JDM-HQ, highest-quartile scores in JDM; CANDLE, Chronic Atypical Neutrophilic Dermatosis with Lipodystrophy and Elevated temperature; SAVI, STING-Associated Vasculopathy with onset during Infancy; NOMID, neonatal-onset multisystem inflammatory disease; HC, healthy controls; IQR, interquartile range
Fig. 2
Fig. 2
Principal component 2 (PC2) and 3 (PC3) scores from principal component analysis (PCA) of interferon-regulated gene (IRG) scores from JDM patients with other conditions. a PCA-A with JDM (including JDM highest quartile IRG score, JDM-HQ), CANDLE, SAVI, NOMID, and HC. b PCA-B with anti-MDA5 autoantibody-positive subgroup of JDM, CANDLE, SAVI, NOMID, and HC. c PCA-C with anti-NXP2 autoantibody-positive subgroup of JDM, CANDLE, SAVI, NOMID, and HC. d PCA-D with anti-TIF1 autoantibody-positive subgroup of JDM, CANDLE, SAVI, NOMID, and HC. Dotted circles represent each group, including CANDLE (pink), SAVI (red), and JDM-HQ or JDM myositis-specific autoantibody (MSA) group (anti-TIF1, anti-NXP2, anti-MDA5) (blue). JDM, as opposed to JDM-HQ, in a represents the lower 3 quartiles of JDM IRG scores. Abbreviations: IRG, interferon-regulated gene; JDM, juvenile dermatomyositis; JDM-HQ, highest-quartile scores in JDM; TIF1, subgroup of JDM with anti-TIF1 myositis-specific autoantibodies; NXP2, subgroup of JDM with anti-NXP2 myositis-specific autoantibodies; MDA5, subgroup of JDM with anti-MDA5 myositis-specific autoantibodies; neg, subgroup of JDM that was negative on testing for myositis-specific autoantibodies; CANDLE, Chronic Atypical Neutrophilic Dermatosis with Lipodystrophy and Elevated temperature; SAVI, STING-Associated Vasculopathy with onset during Infancy; NOMID, neonatal-onset multisystem inflammatory disease; HC, healthy controls
Fig. 3
Fig. 3
Proportion of individual genes from 28 normalized IRG-S for JDM highest quartile (JDM-HQ), CANDLE, SAVI. *JDM-HQ gene proportion significantly different from SAVI. †JDM-HQ gene proportion significantly different from CANDLE. ‡JDM-HQ gene proportion significantly different from CANDLE and SAVI. Heat map of median proportion of the respective gene by normalized Z-score with red for highest proportion and dark blue for lowest proportion. Bold indicates values in a diagnosis group(s) significantly higher compared to values in the italicized/underlined group(s). Italicized/underlined indicates values in a diagnosis group(s) significantly lower compared to values in bold group(s). a All 28 IRGs are included. b IFI27 has been removed. Individual gene percentages (%) of the total IRG score, by normalized Z-scores (see the “Methods” section) are shown in alphabetical order by gene as median and interquartile ranges for JDM highest-quartile, CANDLE, and SAVI patients. Medians percentages of each gene by diagnosis may not sum to 100 due to skewing in the group, but they do in individual patients. JDM-HQ (n = 14) included 4 with anti-TIF1 Ab, 4 with anti-MDA4 Ab, 3 with anti-NXP2 Ab, and 3 MSA-neg. The JDM Physician Global Activity (PGA) median is 4.1/10 with an interquartile range of 2.4 to 4.7. Abbreviations: IRG, interferon-regulated gene; JDM-HQ, juvenile dermatomyositis patients with the highest quartile of IRG scores; CANDLE, Chronic Atypical Neutrophilic Dermatosis with Lipodystrophy and Elevated temperature; SAVI, STING associated Vasculopathy with onset during Infancy; anti-TIF1 Ab, includes subgroup of JDM patients with anti-TIF1 autoantibodies; anti-NXP2 Ab, subgroup of JDM patients with anti-NXP2 autoantibodies; anti-MDA5 Ab, includes subgroup of JDM patients with anti-MDA5 autoantibodies; MSA-neg, includes subgroup of JDM that is negative for myositis-specific autoantibodies; PGA. Physician Global Activity on 0–10 visual analog scale

References

    1. Pachman LM, Khojah AM. Advances in juvenile dermatomyositis: myositis specific antibodies aid in understanding disease heterogeneity. J Pediatr. 2018;195:16–27. doi: 10.1016/j.jpeds.2017.12.053.
    1. Tansley SL, Simou S, Shaddick G, Betteridge ZE, Almeida B, Gunawardena H, Thomson W, Beresford MW, Midgley A, Muntoni F, et al. Autoantibodies in juvenile-onset myositis: their diagnostic value and associated clinical phenotype in a large UK cohort. J Autoimmun. 2017;84:55–64. doi: 10.1016/j.jaut.2017.06.007.
    1. Rider LG, Nistala K. The juvenile idiopathic inflammatory myopathies: pathogenesis, clinical and autoantibody phenotypes, and outcomes. J Intern Med. 2016;280(1):24–38. doi: 10.1111/joim.12444.
    1. Habers GE, Huber AM, Mamyrova G, Targoff IN, O'Hanlon TP, Adams S, Pandey JP, Boonacker C, van Brussel M, Miller FW, et al. Brief report: association of myositis autoantibodies, clinical features, and environmental exposures at illness onset with disease course in juvenile myositis. Arthritis Rheumatol. 2016;68(3):761–768. doi: 10.1002/art.39466.
    1. Miller FW, Lamb JA, Schmidt J, Nagaraju K. Risk factors and disease mechanisms in myositis. Nat Rev Rheumatol. 2018;14(5):255–268. doi: 10.1038/nrrheum.2018.48.
    1. Bilgic H, Ytterberg SR, Amin S, McNallan KT, Wilson JC, Koeuth T, Ellingson S, Newman B, Bauer JW, Peterson EJ, et al. Interleukin-6 and type I interferon-regulated genes and chemokines mark disease activity in dermatomyositis. Arthritis Rheum. 2009;60(11):3436–3446. doi: 10.1002/art.24936.
    1. Greenberg SA, Higgs BW, Morehouse C, Walsh RJ, Kong SW, Brohawn P, Zhu W, Amato A, Salajegheh M, White B, et al. Relationship between disease activity and type 1 interferon- and other cytokine-inducible gene expression in blood in dermatomyositis and polymyositis. Genes Immun. 2012;13(3):207–213. doi: 10.1038/gene.2011.61.
    1. Moneta GM, Pires Marafon D, Marasco E, Rosina S, Verardo M, Fiorillo C, Minetti C, Bracci-Laudiero L, Ravelli A, De Benedetti F, et al. Muscle expression of type I and type II interferons is increased in juvenile dermatomyositis and related to clinical and histological features. Arthritis Rheumatol. 2019;71(6):1011–21.
    1. Wong D, Kea B, Pesich R, Higgs BW, Zhu W, Brown P, Yao Y, Fiorentino D. Interferon and biologic signatures in dermatomyositis skin: specificity and heterogeneity across diseases. PLoS One. 2012;7(1):e29161. doi: 10.1371/journal.pone.0029161.
    1. Huard C, Gulla SV, Bennett DV, Coyle AJ, Vleugels RA, Greenberg SA. Correlation of cutaneous disease activity with type 1 interferon gene signature and interferon beta in dermatomyositis. Br J Dermatol. 2017;176(5):1224–1230. doi: 10.1111/bjd.15006.
    1. Baechler EC, Bauer JW, Slattery CA, Ortmann WA, Espe KJ, Novitzke J, Ytterberg SR, Gregersen PK, Behrens TW, Reed AM. An interferon signature in the peripheral blood of dermatomyositis patients is associated with disease activity. Mol Med. 2007;13(1–2):59–68. doi: 10.2119/2006-00085.Baechler.
    1. Greenberg SA, Pinkus JL, Pinkus GS, Burleson T, Sanoudou D, Tawil R, Barohn RJ, Saperstein DS, Briemberg HR, Ericsson M, et al. Interferon-alpha/beta-mediated innate immune mechanisms in dermatomyositis. Ann Neurol. 2005;57(5):664–678. doi: 10.1002/ana.20464.
    1. Fall N, Bove KE, Stringer K, Lovell DJ, Brunner HI, Weiss J, Higgins GC, Bowyer SL, Graham TB, Thornton S, et al. Association between lack of angiogenic response in muscle tissue and high expression of angiostatic ELR-negative CXC chemokines in patients with juvenile dermatomyositis: possible link to vasculopathy. Arthritis Rheum. 2005;52(10):3175–3180. doi: 10.1002/art.21303.
    1. Brehm A, Liu Y, Sheikh A, Marrero B, Omoyinmi E, Zhou Q, Montealegre G, Biancotto A, Reinhardt A, Almeida de Jesus A, et al. Additive loss-of-function proteasome subunit mutations in CANDLE/PRAAS patients promote type I IFN production. J Clin Invest. 2015;125(11):4196–4211. doi: 10.1172/JCI81260.
    1. Liu Y, Ramot Y, Torrelo A, Paller AS, Si N, Babay S, Kim PW, Sheikh A, Lee CC, Chen Y, et al. Mutations in proteasome subunit beta type 8 cause chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature with evidence of genetic and phenotypic heterogeneity. Arthritis Rheum. 2012;64(3):895–907. doi: 10.1002/art.33368.
    1. Kim H, Sanchez GA, Goldbach-Mansky R. Insights from Mendelian interferonopathies: comparison of CANDLE, SAVI with AGS, monogenic lupus. J Mol Med (Berl) 2016;94(10):1111–1127. doi: 10.1007/s00109-016-1465-5.
    1. Liu Y, Jesus AA, Marrero B, Yang D, Ramsey SE, Montealegre Sanchez GA, Tenbrock K, Wittkowski H, Jones OY, Kuehn HS, et al. Activated STING in a vascular and pulmonary syndrome. N Engl J Med. 2014;371(6):507–518. doi: 10.1056/NEJMoa1312625.
    1. Rodero MP, Crow YJ. Type I interferon-mediated monogenic autoinflammation: the type I interferonopathies, a conceptual overview. J Exp Med. 2016;213(12):2527–2538. doi: 10.1084/jem.20161596.
    1. de Jesus AA, Canna SW, Liu Y, Goldbach-Mansky R. Molecular mechanisms in genetically defined autoinflammatory diseases: disorders of amplified danger signaling. Annu Rev Immunol. 2015;33:823–874. doi: 10.1146/annurev-immunol-032414-112227.
    1. Sanchez GAM, Reinhardt A, Ramsey S, Wittkowski H, Hashkes PJ, Berkun Y, Schalm S, Murias S, Dare JA, Brown D, et al. JAK1/2 inhibition with baricitinib in the treatment of autoinflammatory interferonopathies. J Clin Invest. 2018;128(7):3041–3052. doi: 10.1172/JCI98814.
    1. Torrelo A. CANDLE syndrome as a paradigm of proteasome-related autoinflammation. Front Immunol. 2017;8:927. doi: 10.3389/fimmu.2017.00927.
    1. Kim H, de Jesus AA, Brooks SR, Liu Y, Huang Y, VanTries R, Montealegre Sanchez GA, Rotman Y, Gadina M, Goldbach-Mansky R. Development of a validated interferon score using NanoString technology. J Interf Cytokine Res. 2018;38(4):171–185. doi: 10.1089/jir.2017.0127.
    1. Bohan A, Peter JB. Polymyositis and dermatomyositis (first of two parts) N Engl J Med. 1975;292(7):344–347. doi: 10.1056/NEJM197502132920706.
    1. Trieu EP, Targoff IN. Immunoprecipitation: Western blot for proteins of low abundance. Methods Mol Biol. 2015;1312:327–342. doi: 10.1007/978-1-4939-2694-7_34.
    1. Benjamini Y, Krieger AM, Yekutieli D. Adaptive linear step-up procedures that control the false discovery rate. Biometrika. 2006;93(3):491–507. doi: 10.1093/biomet/93.3.491.
    1. de Jesus AA, Hou Y, Brooks S, Malle L, Biancotto A, Huang Y, Calvo KR, Marrero B, Moir S, Oler AJ, et al. Distinct interferon signatures and cytokine patterns define additional systemic autoinflammatory diseases. J Clin Invest. 2020. 10.1172/JCI129301.
    1. Rider LG, Aggarwal R, Machado PM, Hogrel JY, Reed AM, Christopher-Stine L, Ruperto N. Update on outcome assessment in myositis. Nat Rev Rheumatol. 2018;14(5):303–318. doi: 10.1038/nrrheum.2018.33.
    1. Rice GI, Melki I, Fremond ML, Briggs TA, Rodero MP, Kitabayashi N, Oojageer A, Bader-Meunier B, Belot A, Bodemer C, et al. Assessment of type I interferon signaling in pediatric inflammatory disease. J Clin Immunol. 2017;37(2):123–132. doi: 10.1007/s10875-016-0359-1.
    1. Li Y, Li C, Xue P, Zhong B, Mao AP, Ran Y, Chen H, Wang YY, Yang F, Shu HB. ISG56 is a negative-feedback regulator of virus-triggered signaling and cellular antiviral response. Proc Natl Acad Sci U S A. 2009;106(19):7945–7950. doi: 10.1073/pnas.0900818106.
    1. Crowe WE, Bove KE, Levinson JE, Hilton PK. Clinical and pathogenetic implications of histopathology in childhood polydermatomyositis. Arthritis Rheum. 1982;25(2):126–139. doi: 10.1002/art.1780250203.
    1. Gitiaux C, De Antonio M, Aouizerate J, Gherardi RK, Guilbert T, Barnerias C, Bodemer C, Brochard-Payet K, Quartier P, Musset L, et al. Vasculopathy-related clinical and pathological features are associated with severe juvenile dermatomyositis. Rheumatology. 2016;55(3):470–479.
    1. Gadina M, Johnson C, Schwartz D, Bonelli M, Hasni S, Kanno Y, Changelian P, Laurence A, O’Shea JJ. Translational and clinical advances in JAK-STAT biology: the present and future of jakinibs. J Leukoc Biol. 2018;104(3):499–514. doi: 10.1002/JLB.5RI0218-084R.
    1. Fremond ML, Rodero MP, Jeremiah N, Belot A, Jeziorski E, Duffy D, Bessis D, Cros G, Rice GI, Charbit B, et al. Efficacy of the Janus kinase 1/2 inhibitor ruxolitinib in the treatment of vasculopathy associated with TMEM173-activating mutations in 3 children. J Allergy Clin Immunol. 2016;138(6):1752–1755. doi: 10.1016/j.jaci.2016.07.015.
    1. Aeschlimann FA, Fremond ML, Duffy D, Rice GI, Charuel JL, Bondet V, Saire E, Neven B, Bodemer C, Balu L, et al. A child with severe juvenile dermatomyositis treated with ruxolitinib. Brain. 2018;141(11):e80. doi: 10.1093/brain/awy255.
    1. Papadopoulou C, Hong Y, Omoyinmi E, Brogan PA, Eleftheriou D. Janus kinase 1/2 inhibition with baricitinib in the treatment of juvenile dermatomyositis. Brain. 2019;142(3):e8.
    1. Kato M, Ikeda K, Kageyama T, Kasuya T, Kumagai T, Furuya H, Furuta S, Tamachi T, Suto A, Suzuki K, et al. Successful treatment for refractory interstitial lung disease and pneumomediastinum with multidisciplinary therapy including tofacitinib in a patient with anti-MDA5 antibody-positive dermatomyositis. J Clin Rheumatol. 2019. 10.1097/RHU.0000000000000984.
    1. Kurasawa K, Arai S, Namiki Y, Tanaka A, Takamura Y, Owada T, Arima M, Maezawa R. Tofacitinib for refractory interstitial lung diseases in anti-melanoma differentiation-associated 5 gene antibody-positive dermatomyositis. Rheumatology. 2018;57(12):2114–2119. doi: 10.1093/rheumatology/key188.
    1. Sabbagh S, Almeida de Jesus A, Hwang S, Kuehn HS, Kim H, Jung L, Carrasco R, Rosenzweig S, Goldbach-Mansky R, Rider LG. Treatment of anti-MDA5 autoantibody-positive juvenile dermatomyositis using tofacitinib. Brain. 2019;142(11):e59. doi: 10.1093/brain/awz293.
    1. Reed AM, Crowson CS, Hein M, de Padilla CL, Olazagasti JM, Aggarwal R, Ascherman DP, Levesque MC, Oddis CV, Group RIMS Biologic predictors of clinical improvement in rituximab-treated refractory myositis. BMC Musculoskelet Disord. 2015;16:257. doi: 10.1186/s12891-015-0710-3.
    1. Nagaraju K, Ghimbovschi S, Rayavarapu S, Phadke A, Rider LG, Hoffman EP, Miller FW. Muscle myeloid type I interferon gene expression may predict therapeutic responses to rituximab in myositis patients. Rheumatology. 2016;55(9):1673–1680. doi: 10.1093/rheumatology/kew213.
    1. Lopez De Padilla CM, Crowson CS, Hein MS, Strausbauch MA, Aggarwal R, Levesque MC, Ascherman DP, Oddis CV, Reed AM. Interferon-regulated chemokine score associated with improvement in disease activity in refractory myositis patients treated with rituximab. Clin Exp Rheumatol. 2015;33(5):655–663.

Source: PubMed

3
Abonnieren