Skip to main content

Bench to bedside review of myositis autoantibodies

Abstract

Idiopathic inflammatory myopathies represent a heterogeneous group of autoimmune diseases with systemic involvement. Even though numerous specific autoantibodies have been recognized, they have not been included, with the only exception of anti-Jo-1, into the 2017 Classification Criteria, thus perpetuating a clinical-serologic gap. The lack of homogeneous grouping based on the antibody profile deeply impacts the diagnostic approach, therapeutic choices and prognostic stratification of these patients. This review is intended to highlight the comprehensive scenario regarding myositis-related autoantibodies, from the molecular characterization and biological significance to target antigens, from the detection tools, with a special focus on immunofluorescence patterns on HEp-2 cells, to their relative prevalence and ethnic diversity, from the clinical presentation to prognosis. If, on the one hand, a notable body of literature is present, on the other data are fragmented, retrospectively based and collected from small case series, so that they do not sufficiently support the decision-making process (i.e. therapeutic approach) into the clinics.

Background

The detection of autoantibodies in autoimmune diseases, either systemic or organ specific, can have both diagnostic and prognostic importance. Some autoantibodies have a clear pathogenic role, such as anti-erythrocyte membrane proteins antibodies in autoimmune hemolytic anemia and anti-dsDNA in Systemic Lupus Erythematosus (SLE). However, the presence of autoantibodies is more frequently considered to be an epiphenomenon, even though their detection plays a critical role for the diagnosis of some connective tissue diseases (CTDs), [i.e. anti-SSA/Ro in Sjögren syndrome (SjS) and anti-Sm in SLE], as included into the classification criteria of CTDs [1, 2]. Repeated serum sampling might be informative about the clinical course of the disease and response to immunosuppressive therapy, as in the case of anti-dsDNA antibodies in SLE. Furthermore, the presence of some autoantibodies could help to discriminate specific clinical patterns within the same disease, as with diffuse and limited cutaneous systemic sclerosis (SSc) and their relationship with anti-Scl-70 and anti-centromere, respectively.

In idiopathic inflammatory myopathies (IIMs), albeit anti-Jo-1 autoantibody was discovered more than thirty years ago, the percentage of patients in whom an autoantibody could not be recognized (the so called “serologic gap”) was still high until recently. IIMs have been historically divided in polymyositis (PM) and dermatomyositis (DM) on a purely clinical basis despite phenotypic variability.

Due to this heterogeneity, the numerous autoantibodies and unavailability of reliable assays in all the laboratories, the clinical use of serology lagged behind and autoantibodies are not part of the most recent IIM Classification Criteria [3].

A comprehensive review regarding the clinical features, diagnostic work-up and relationship to some peculiar autoantibodies has been recently published by Milone [4].

Myositis-specific and –associated autoantibodies: definitions

Autoantibodies found in IIM patients have been classified into two main categories: myositis-specific autoantibodies (MSAs), which can be found in IIMs exclusively, and myositis-associated autoantibodies (MAAs), which can also be found in other CTDs [5, 6]. MSAs and MAAs are summarized in Table 1.

Table 1 Summary of the main features of MSAs and MAAs

There is no agreement about the attribution of rare and newly discovered autoantibodies to either MSAs or MAAs group [7]. Anti-synthetase autoantibodies (ARS) themselves, especially anti-PL-7, PL-12 and KS, often detected in interstitial pneumonia with autoimmune features (IPAF) patients, independently from muscular involvement, are still discussed as MSAs [8].

The MAAs group contains Anti-Pm-Scl, U1/U2RNP and Ku, which are associated with overlap syndromes with muscular involvement [9]. Anti-fibrillarin and anti-U1-snRNP are sometimes considered as MAAs, even though they are more specific for the diagnosis of SSc and mixed connective tissue disease (MCTD), respectively [10, 11]. Anti-Ro52 are usually considered a MAA, even though they are more frequently found in association with other MSA (ARS, anti-MDA5 and anti-SRP, in particular) [12], and define a peculiar clinical spectrum in which the lung involvement is more common than the muscular one [13].

Detection methods

There are several methods to test for MSAs and MAAs, with variable sensibility, specificity, costs, complexity and feasibility in clinical and research settings. Indirect immuno-fluorescence (IIF) on HEp-2 cells, counter-immuno-electrophoresis (CIE), immuno-diffusion (ID) and immuno-enzymatic assays such as enzyme-linked immunosorbent assay (ELISA), fluorescent enzyme-linked assay (FEIA) and chemiluminescent immuno-assay (CLIA) are the most commonly adopted systems in diagnostic laboratories. However, immuno-precipitation (IP) of RNAs with silver staining and/or protein IP of cellular lysates (usually K562 cells) radiolabeled with 35S-methionin, is the gold standard for most antibodies. In order to streamline the detection of many autoantibodies at the same time in a cost/effective manner, recent multiplex assays, like immunoblots (IB) and Addressable Laser Beads Immuno Assay (ALBIA) have been developed [14].

Anti-nuclear autoantibodies (ANA) determination by IIF is virtually universally available and it can be considered an accessible screening method for many MSAs and MAAs [15]. Furthermore, the recognition of particular IIF patterns can hint to some specific autoantibodies. However, using ANA IIF as the sole screening method for MSAs/MAAs, is not recommended because of low sensitivity, very low specificity and/or lack of antigen expression by HEp-2 cells [16]. In addition to this, ANA IIF is burdened by reproducibility issues due to the operator-dependent recognition of rare patterns and variation among different commercial HEp-2 substrates [17].

CIE and ID have historically been the first methods to detect specific MSAs/MAAs. Even though they identify numerous specificities within a single assay, they are semi-quantitative, work-intensive and scarcely sensitive [14]. For all those reasons, they have been largely substituted by immunoenzymatic tests.

The main advantages of ELISAs are standardization, large-scale reproducibility and quantitative results. The disadvantage of the conjugation of antigens to a substrate resides on the possible loss of conformational epitopes and/or the formation of neo-epitopes, which may in turn impact the test performance [14].

Immunoblot (IB) assays can simultaneously test for many autoantibodies, albeit the denaturation of proteins during gel preparation implicates the recognition of linear epitopes only [14].

Commercial multiplex IBs, like dot blots or line blot assays (LIA), based on recombinant or synthetic peptides have been increasingly available, benefitting from pure antigens not requiring a gel passage [18, 19].

IP is the gold standard as it evaluates the binding of the autoantibodies to the RNA and protein complexes in their native conformation, yielding the best sensitivity and specificity [20]. The major limitations of IP are due to technical difficulties, costs and use of radioactive reagents [20]. In addition, interpretation may be complex because the target antigen can co-precipitate with non-target complexed proteins, with the consequence of multiple IP bands [20]. Thus, a comparison with reference sera or further purification and characterization with other methods (i.e. mass spectrometry) is necessary [14]. Quantitative PCR of reverse transcribed RNA components extracted from standard IP can be also used as a detection method for autoantibodies binding ribonucleoproteic complexes [21].

Myositis-specific autoantibodies

Anti-synthetases autoantibodies

ARS are a group of autoantibodies directed against the aminoacyl transfer RNA (tRNA) synthetases, which are amino acid-charging enzymes. Autoantibodies to eight tRNA synthetases have been discovered so far: histidyl (Jo-1), threonyl (PL-7), alanyl (PL-12), glycyl (EJ), isoleucyl (OJ), asparaginyl (KS), phenylalanyl (ZO), and tyrosyl (YRS/HA) tRNA synthetases [22].

Anti-Jo-1 antibodies were identified by double immune-diffusion (DID) of calf thymus extract in 1980 and were the first MSAs described [23]. IP represents the gold standard for their identification with the following protein bands: Jo-1 50 kDa, PL-7 80 kDa, PL-12 110 kDa, EJ 75 kDa, OJ 150 kDa and a multi-enzyme complex of 170, 130, and 75 kDa, KS 65 kDa, ZO 60/70 kDa, YRS/HA 59 kDa [22].

IIF on HEp-2 cells usually demonstrates a cytoplasmic pattern, ranging from fine (Jo-1) to dense fine speckled or homogeneous (PL-7, PL-12) whereas the nucleoplasm is usually negative (Fig. 1a–e) [24]. In these cases, also patients without muscular involvement should be assessed for ARS especially when interstitial lung disease (ILD), arthritis or scleroderma features are present [25].

Fig. 1
figure 1

IIF on HEp-2 ANA slides of myositis-specific autoantibodies from patients with IIMs (EUROIMMUN, Lübeck, Germany). a Anti-Jo-1 and b anti-Jo-1 and anti-Ro52, c anti-PL-7, d anti-PL-12, e anti-KS, f anti-Mi-2, g anti-SAE-1, h anti-MDA5, i anti-NXP-2 and j anti-NXP-2 with coiled bodies, k anti-TIF1γ, l anti-SRP

Anti-Jo-1 is the only autoantibody routinely tested as widely available in most commercial ENA screening assays. An ELISA screening test has been recently developed to identify ARS, with high sensibility and specificity if compared to IP [26] and some commercially available IBs can identify some non-Jo-1 anti-synthetase antibodies [20, 25].

Anti-Jo-1 was first discovered in the ‘80ies in patients with PM [23]. Larger cohorts later demonstrated that its presence was associated with the classical triad of arthritis, myositis and ILD in the majority of patients, in addition to Raynaud’s phenomenon, mechanic’s hands and fever. This clinical presentation together with anti-Jo-1 autoantibodies, led to the description of the antisynthetase syndrome (ASSD), the first attempt to phenotype IIMs in clinical-serologic syndromes [27]. Anti-Jo-1 is detected in 15-25% of patients with polymyositis/dermatomyositis (PM/DM), whereas the other ARS are rarer (anti-PL-7 4-12%, PL-12 < 5%, EJ < 5%, OJ < 5% and only few cases reported with anti-KS, ZO e HA/YRS) [22]. In two-third of the cases, high titers of anti-Ro52 antibodies can be also detected and have been associated with an higher risk of ILD [28].

Clinically, anti-PL-7 patients more frequently present hypo-myopathic features [29, 30], whereas in anti-PL-12 and anti-KS patients the disease can be limited to the lung [3133]. In a quarter of anti-Jo-1 patients, a symmetrical polyarthritis mimicking rheumatoid arthritis is the main presenting finding and, in some of them, anti-cyclic citrullinated peptide antibodies and rheumatoid factors can be also detected [34].

Regardless of clinical characteristics at presentation (arthritis, myositis and ILD) every patient tends to develop the other features of ASSD when not properly treated [34]. Of note, the lower esophageal sphincter is involved more frequently if compared to the other IIMs [35]. ASSD is histopathologically classified within perimysial immune-myopathies in which perimysium fragmentation and muscle fiber necrosis are the main feature, differently from other DM biopsically characterized by atrophy and vasculopathy [36]. Type I interferon-signature in ASSD is responsible for MHC class I upregulation [37] and MHC class II perifascicular expression [38].

Anti-Mi-2

Anti-Mi-2 antibodies were the first autoantibodies specific for DM recognized by DID using calf thymus extract [39]. Mi-2 is a helicase of the Nucleosome Remodeling Deacetylase (NuRD) multi-protein complex with nucleosome remodeling and histone deacetylase/demethylase activities [40]. Anti-Mi-2 autoantibodies immunoprecipitate a major protein of 240 kDa, composed by two proteins, Mi-2α and Mi-2β of 220 and 218 kDa, respectively. Other NuRD complex proteins co-precipitate at 200, 150, 75, 65, 63, 50 and 34 kDa [41, 42]. IIF on HEp-2 cells reveals a characteristic fine speckled ANA pattern; during metaphase, chromatin mass is not stained but the nucleoplasm presents the same fine tiny speckles (Fig. 1f). Commercial ELISA and immunoblot kits identify anti-Mi-2 autoantibodies. Anti-Mi-2 are commonly detected in DM patients, either in adults (11–59%) or in children (4–10%), with a great variability among the studies. Their presence in PM and sporadic inclusion body myositis (sIBM) is rarer [40].

The Mi-2 protein is over-regulated during muscle regeneration in DM patients and thought to be related to UV rays exposition, sex and HLA (DRB1*0302 and DRB1*0701) [4345]. Anti-Mi-2 positive DM patients usually exhibit mild myopathy despite high creatine kinase (CK) levels, without lung involvement and/or cancer [43]. Overall, anti-Mi2 positive is associated with a positive prognosis and a good response to corticosteroids [43].

Anti-SAE

Small Ubiquitin-like Modifiers (SUMOs) have a key role in post-transcriptional modification of specific proteins in a ubiquitin-like fashion. This process is controlled by the SUMO-Activating Enzyme (SAE), a heterodimer composed of two subunits, SAE-1 and SAE-2 [46], representing the targets of anti-SAE autoantibodies. IP characteristically shows two bands of 40 and 90 kDa, respectively [46, 47]. The IIF ANA pattern is coarse or fine speckled and nucleoli are typically not stained (Fig. 1g) [47].

Anti-SAE are associated with a typical DM phenotype with different prevalence in European (4-10%) and Asian (1–3%) cohorts [4850], probably due to the strict association with HLADRB1*04-DQA1*03-DQB1*03 haplotypes [51].

The cutaneous involvement is usually severe and typically precedes the muscular involvement. Other clinical relationships cannot be excluded because of the few described cases. However, ILD seems to be rare, whereas severe dysphagia and systemic symptoms have been reported [47]. Only one case series claimed an association with cancer [52].

Anti-MDA5

Melanoma Differentiation-Associated gene 5 (MDA5) or Interferon-induced helicase C domain-containing protein 1 (IFIH1), is an innate cytosolic sensor, member of the retinoic acid-inducible gene I (RIG-I)-like receptors family (RLRs). MDA5 is able to recognize double-stranded RNA and to initiate signaling events leading to type I interferons production [53].

Anti-MDA5 autoantibodies were firstly detected in IP as a 140 kDa band in a Japanese case series of patients with clinically amyopathic dermatomyositis (CADM) and rapidly progressive interstitial lung disease (RP-ILD). For this reason, they were initially called anti-CADM-140 autoantibodies [54]. Nowadays, ELISA and IB tests are commercially available.

IIF on HEp-2 cells is usually negative. In our experience, a faint fine speckled cytoplasmic fluorescence may be detected in scattered cells (unpublished data) (Fig. 1h).

Clinically, DM anti-MDA5 positive patients present low grade/absent muscle inflammation and acute or subacute RP-ILD [55, 56], which is considered the major negative prognostic factor of this subgroup [57].

MDA5 represents the most frequent target antigen in DM patients of Asian ancestry (10–48% of cases) [58] whereas its prevalence in Europe and USA ranges from 0 to 13%, with great variability among the studies [5961] and a different clinical presentation. A forthcoming European case series is going to be presented at the European League Against Rheumatism 2018 Congress by Cavagna et al. (unpublished data). A seasonal pattern of CADM has been proposed by Muro et al. [62], suggesting the influence of environmental factors and HLA-DRB1*04:01 and *12:02 have been proposed as further predisposing factors [63].

In addition to classic DM-related cutaneous manifestations, skin involvement is usually severe and characterized by the so called “inverse Gottron papules”, which are tender palmar papules that tend to evolve towards ulcerated-necrotic lesions, with or without digital pulp ulcers [64, 65]. In addition, polyarthritis, recurrent oral aphtosis and diffuse alopecia have been described [66]. A juvenile DM with anti-MDA5 autoantibodies has been also described [67]. No association with malignancies has been demonstrated so far. Macrophage activation syndrome have been described in CADM associated RP-ILD patients. Particularly, a ferritin level of above 1500 ng/mL has been claimed as a predictor of death [68, 69]. Anti-MDA5 autoantibodies titer seems to correlate with disease activity and response to therapy [69].

Anti-TIF-1

The transcription intermediary factors-1 (TIF-1) family belongs to the tripartite motif-containing proteins (TRIM) superfamily and is involved in multiple biological processes, such as cycle regulation, mitosis and innate immunity [70].

Targoff et al. and Kaji et al. [71, 72] independently described two antibodies directed against a 155 and 140 kDa, rapidly identified as TIF-1γ (TRIM33) and TIF-1α (TRIM24), respectively. Subsequently, a third 120 kDa band, partially overlapping with anti-PL-12, was identified as TIF1β (TRIM28) [73].

IIF on HEp-2 cells demonstrates a fine speckled nuclear pattern (Fig. 1i). ELISA and IB, compared to IP, are reliable test for the detection of anti-TIF-1γ autoantibodies [74]. Two-thirds of the patients present anti-TIF-1γ and anti-TIF-1α autoantibodies, whereas the remaining one-third is positive for anti-TIF-1γ autoantibodies exclusively [70]. Albeit MSAs are claimed to be mutually exclusive, double-positive patients for anti-TIF-1α/Mi-2 autoantibodies have been described [75].

Hyper-expression of TIF-1γ has been found in tumors [76] and regenerating myofibres of DM patients [77]. A meta-analysis demonstrated that anti-TIF-1γ has a 78% sensitivity and 89% specificity for the diagnosis of cancer-associated myositis, with a 58% positive and 95% negative predictive value [78]. The risk of malignancy is higher in patients with anti-TIF-1γ/α than in those with anti-TIF-1γ alone [70].

Clinically, anti-TIF-1 positive patients can be classified in two age groups: (1) younger than 40-year-old patients, with a classical DM at presentation and (2) older than 40-year-old patients, with cancer-associated myositis [70]. Solid tumors, like ovary, lung and breast cancer are the most commonly associated neoplasia, but hematologic disorders and malignancies have been described as well [79]. In general anti-TIF-1γ patients exhibit a hypo-myopathic DM with reduced prevalence of systemic involvement, namely ILD, Raynaud’s phenomenon and arthritis [80]. Conversely, nutcracker esophagus is three times more frequent in anti-TIF-1γ patients than other IIMs [35]. Widespread cutaneous involvement is associated with unique features, such as palmar hyper-keratotic papules, psoriatic-like dermatitis and atrophic hypo-pigmented patches with telangiectasias [80]. An ovoid palatal patch may be present in about one half of patients, more frequently females with cancer-associated amyopathic disease [81].

Anti-NXP-2

Nuclear matrix protein 2 (NXP-2), encoded by the microrchidia 3 gene, is a 140 kDa protein involved in epigenetic regulation, RNA metabolism and preservation of nuclear chromatin architecture [82]. Anti-NXP-2 autoantibodies were found in a cohort of juvenile DM patients as a 140 kDa protein firstly named anti-MJ [83].

IIF on HEp-2 cells reveals a fine speckled nuclear pattern (Fig. 1k). Moreover, a nuclear dots pattern is detectable in 60% of sera [84], due to co-localization of NXP-2 with pro-myelocitic leukemia (PML) bodies [85] (Fig. 1j).

Anti-NXP-2 antibodies have been initially associated with a severe juvenile DM complicated by calcinosis, polyarthritis and intestinal vasculitis [86]. More recently, they have been also found in adult patients, with variable prevalence from 1.6 to 17% [8789]. Anti-NXP-2 autoantibodies show a bimodal spectrum of clinical association, with calcinosis being more frequent in younger patients and cancer more common in the elderly [90], especially in male gender [88], even though with a lower prevalence when compared to anti-TIF-1 [87, 88].

Anti-SRP

The signal recognition particle (SRP) is a complex of six proteins (9, 14, 19, 54, 68 and 72 kDa) and a 300 nucleotides long RNA (7SL RNA) involved in the recognition and transportation of proteins to the endoplasmic reticulum [91]. Anti-SRP autoantibodies are more frequently directed against the SRP-54 fragment, albeit anti-SRP-68, anti-SRP-72 and anti-7SL RNA autoantibodies have been also described [91].

A dense fine speckled cytoplasmic pattern has been associated with the presence of anti-SRP (Fig. 2a); moreover, IIF on stomach–liver–kidney rat sections demonstrates a cytoplasmic staining of gastric chief cells (Fig. 2b) and hepatocytes (Fig. 2c) [92].

Fig. 2
figure 2

Immunofluorescence patterns from patients with IMNMs with anti-SRP and anti-HMGCR antibodies on HEp-2 ANA slides (EUROIMMUN, Lübeck, Germany) and rat liver and stomach slides (DiaSorin, Italy), compared to AMA-M2 antibodies. a Anti-SRP cytoplasmic dense fine speckled pattern on HEp-2, b chief cells on stomach and c fine granular liver staining; d AMA-M2 cytoplasmic reticular on HEp-2, e granular staining of parietal stomach cells and f diffused fluorescence of hepatocytes; g Anti-HMGCR faint cytoplasmic fluorescence on few numbers of HEp-2 cells, h negative stomach and i fine cytoplasmic fluorescence of scattered hepatocytes around the biliary ducts

Anti-SRP-54 autoantibodies ELISA tests are commercially available, but they are less sensitive than IP [93]. Anti-SRP antibodies can also be tested on LIA assays, however careful temperature control is necessary in order to avoid false positive results [19].

These autoantibodies are strongly associated with immune-mediated necrotizing myositis (IMNM) where they may play a pathogenic role [94]. Histopathologically, they are characterized by scarce inflammatory CD8+ endomysial infiltrate, class I MHC upregulation, necrosis and myofiber regeneration [36] with poor response to therapy, mimicking muscular dystrophy [95]. Being strongly associated to HLA-DR5, the prevalence of anti-SRP autoantibodies is higher in Asian (8–13%) than in European patients [96]. Prevalence of anti-SRP in IMNM is highly variable among studies, ranging from 0 to 54%, because of differences regarding the type of assay (IB vs IP), low number of patients and genetic background [97100]. In addition, esophageal involvement is common [101], whereas the possibility of a prominent cardiac involvement has not been yet confirmed [99, 102]. Lung involvement has been reported in a few cases [103], as well as an overlap with anti-synthetase antibodies like PL-12 and anti-Jo-1 [104, 105]. Intriguingly, autoantibodies level correlates to disease activity, CK levels and response to therapy [106].

Anti-HMGCR

The 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR) is the rate-controlling enzyme of the mevalonate pathway, bringing to the production of cholesterol. Of note, HMGCR is the same enzyme targeted by statins. Autoantibodies towards a complex 200/100 kDa band were first described in patients with IMNM, and only after identified as anti-HMGCR autoantibodies [107].

IIF pattern is difficult to recognize. In a minority of cases finely granular cytoplasmic staining with a perinuclear reinforcement is visible on a small number of scattered cells (3% of the total cellularity) (Fig. 2g). On rat liver, a scattered cytoplasmic staining of hepatocytes around the liver lobules, namely anti-HMGCR Antibody Associated Liver Immunofluorescence Pattern (HALIP), can be noted (Fig. 2i) [108, 109]. Anti-HMGCR antibodies can be identified with different immunoenzymatic technologies, such as ELISA, CLIA, IB or ALBIA [110].

A history of statin exposure is not mandatory to develop anti-HMGCR positive IMNM, being of some relevance only in patients older than 50 [111, 112]. Anti-HMGCR antibodies are not found in self-limiting statin associated myopathy [113], albeit they may be associated with an increased risk of cancer [114]. An association with the DRB1*11:01 haplotype has been demonstrated, whereas DQA1 and DQB1 seem to have a protective role [115].

Anti-HMGCR positive patients present with a typical IMNM, responds well to immunosuppressive therapy and intravenous immunoglobulins [116], but tend to relapse after tapering [111]. Younger patients experience more severe disease with worse prognosis [117]. Autoantibody titers seem to correlate with CK levels, muscular weakness and response to therapy [111].

Myositis-associated autoantibodies

Numerous MAAs have been described so far. Characteristically, they can be found in IIMs, albeit not specific as found in other CTDs [118].

Anti-PM-Scl

Anti-PM-Scl autoantibodies are directed against the exosome, a macromolecular nucleolar complex composed by 11–16 proteins (from 20 to 110 kDa) that degrades mRNA. The two pivotal proteins of the complex are PM-Scl-75 and PM-Scl-100. IP represents the gold standard for their determination. Historically, an ID test after positive nucleolar staining in IIF was used to confirm anti-PM-Scl reactivity.

PM-Scl-100 and PM-Scl-75 were identified in 1992 and, in the following years, the immuno-dominant epitope PM1α was cloned and employed to develop reliable and specific ELISA tests [119]. PM1α ELISA and PM-Scl-100 LIA tests show concordance with IP at high level (> 90 and 98.3%, respectively), whereas PM-Scl-75 LIA has a lower specificity, especially when considering PM/SSc overlap syndromes [120]. Single positivity against PM-Scl-75 or -100 can be detected and associates with different disease phenotypes. HEp-2 IIF typically shows a mixed homogeneous nucleolar and fine speckled nuclear pattern when anti-PM-Scl-100 are present, whilst anti-PM1-α and PM-Scl-75 may show both nucleolar and non-nucleolar patterns (Fig. 3a, b).

Fig. 3
figure 3

Myositis-associated autoantibodies and some peculiar IIF pattern on HEp-2 ANA slides (EUROIMMUN, Lübeck, Germany). a anti-PM-Scl-100, b anti-PM-Scl-75, c anti-U1-snRNP, d anti-Ku, e anti-fibrillarin, f cytoplasmic discrete dots as seen in anti-Su/Ago2, g nuclear few nuclear dots as seen in anti-SMN, h punctate nuclear envelope as seen in anti-NUP

PM-Scl autoantibodies are found in 4–12% of adult patients with myositis [121, 122] with low prevalence in Asiatic and paediatric cohorts [123]. Their presence has been associated with HLA-DQA1*0501, DQB1*02 and DRB1*0301 alleles [122].

Despite their presence in many connective tissue diseases, these autoantibodies are typically present in PM/SSc overlap syndromes with an increased risk of Raynaud’s phenomenon, arthritis, mechanic’s hands and ILD [124]. In detail, isolated anti-PM-Scl-75 have been more frequently found in patients with joint contractures and SSc, higher CK levels associate with anti-PM-Scl-100, whereas the simultaneous presence of anti-PM-Scl-75 and -100 are linked to muscle involvement, digital ulcers and ILD but lower prevalence of lung hypertension [125].

Autoantibodies directed against C1D, an exosome associated protein, were detected by ELISA and Western blot analysis in 23% of a PM/SSc overlap syndrome cohort, with frequencies comparable to anti-PM/Scl antibodies [126].

Anti-RNP

The RNP/Sm complex comprises several proteins (70 kD, A, A′, B, B′, B″, C, D, E, F, G) and five RNA (U1, U2, U4, U5 and U6). U1 RNA interacts with 70 kD, A and C to create the U1-snRNP [127]. High-titre anti-U1-snRNP and in particular when targeting the 70kD protein are considered specific markers of mixed connective tissue disease (MTCD), whereas low titres can be found in other CTDs [127].

Many home-made or commercial assays can detect anti-RNP autoantibodies, with differences among immunoassays depending on the immobilized antigen [127]. Usually, anti-U1-snRNP (more often the 70k subunit) and the anti-Sm (typically the D subunit) are the only autoantibodies tested in clinical practice. Large speckled and large coarse speckled are the most frequent HEp-2 IIF patterns observed (Fig. 3c).

Patients with myositis may exhibit anti-U1-snRNP positivity, especially those with a mild disease [128, 129]. They are usually steroid-responsive, even though ILD and/or neurological involvement may be part of the clinical presentation [128, 129]. Whether the only presence of anti-U1-snRNP and myositis has to be considered an incomplete form of MTCD or a true myositis, is still a matter of debate [128, 129]. In addition, anti-U2-RNP [130], U5-RNP [131] and anti-U4/U6-RNP [132] have been described in patients with PM/SSc overlap syndrome.

Anti-fibrillarin

Fibrillarin, a highly conserved nucleolar 34 kDa protein involved in the processing of ribosomal RNA, is part of the U3-small nucleolar (sno)-RNP complex together with other proteins and U3 RNA. Fibrillarin is the primary target of anti-U3-snoRNP autoantibodies [133]. IP is the gold standard for its detection showing good concordance with IB assays that use the recombinant protein [134].

HEp-2 IIF demonstrates a typical “clumpy” nucleolar pattern, with jagged staining of the nucleoli, coiled bodies and peri-chromosomal staining at the metaphase plates [135] (Fig. 3e).

Anti-fibrillarin antibodies are detected in a small percentage of SSc patients and rarely in SLE, primary Raynaud’s phenomenon and myositis [136]. In detail, they identify a subset of SSc patients more often of African origin, with serious cutaneous and visceral involvement and a higher prevalence of myositis [137, 138].

Anti-Ku

The Ku protein, involved in the canonical non-homologous end-joining pathway of the DNA repair, is a heterodimer consisting of the two subunits, 70 and 80 kDa [139]. Anti-Ku can be identified with numerous assays, such as ELISA, CIE or IB. IIF demonstrates a fine speckled nuclear pattern with a peculiar ring beam surrounding the metaphase on HEp-2 cells and a clumpy speckled pattern on primate’s liver [140] (Fig. 3d).

Anti-Ku autoantibodies have been identified in 9-19% of the patients with PM/SSc overlap syndromes and SLE, being associated with arthralgia, Raynaud’s phenomenon and ILD [141, 142]. Of note, whilst muscular involvement seems to be steroid-sensitive, ILD is more frequently progressive, severe and steroid-resistant [143].

Anti-Ro

Antibodies directed against the ribonucleoproteic complex SSA/Ro and SSB/La have been originally identified in SjS and SLE. Actually, antigen Ro is made by two separate complexes of 52 and 60 kDa called Ro52/TRIM21 and SSA/Ro60, respectively. Antigen SSB/La has a molecular weight of 48 kDa [144].

ANA may result falsely negative on traditional HEp-2 cells when isolate anti-Ro are present, because Ro52 is a cytoplasmic antigen and Ro60 may be lost during the preparation. For this reason, human SSA/Ro60-transfected HEp-2 cells (HEp-2000) are sometimes used [145, 146]. Otherwise, a characteristic pattern defined as “myriad discrete fine speckled” may be observed [147]. Anti-SSB autoantibodies show a similar pattern [148].

Anti-Ro52 can be found in IIMs [149] and are frequently associated with other MSAs, in particular anti-synthetase [28], anti-MDA5 [61] and anti-SRP autoantibodies [12].

They are known to be a negative prognostic factor regarding systemic involvement such as ILD, whereas their role in the severity of muscular involvement has not been identified [13]. Anti-Ro52 autoantibodies are known to be associated with atrioventricular congenital heart block [150].

Anti-cN-1A

Cytosolic 5′nucleotidase 1A (cN-1A o NT5C1A) is a protein involved in the hydrolysis of adenosine monophosphate, controlling energy and metabolic cell balance [151]. Anti-cN-1A autoantibodies, first called anti-Mup44, were simultaneously described by Salajegheh et al. and Pluk et al. [152, 153] as targeting a 44 kDa protein in patients with sIBM.

These autoantibodies were initially detected by immunoblotting from purified skeletal muscle extracts [153]. A novel standardized IgG ELISA is now available [154]. In addition to IgG, circulating IgA and IgM anti-cN-1A autoantibodies have been recognized [155]. IIF ANA pattern is still undefined.

Anti-cN-1A autoantibodies are demonstrated in one-third of the patients with sIBM and in less than 5% with other IIMs or neuromuscular diseases [151]. A recent study demonstrated that positive anti-cN-1A sIBM patients are included in a more severe sIBM subtype and represent a homogeneous group as exhibiting higher mortality risk, less proximal upper limb weakness (not typical of sIBMs) and a cytochrome oxidase deficiency in muscular fibers, when compared to negative patients [156].

It is not known whether they have to be considered as MSAs or MAAs as also demonstrated in other autoimmune diseases, such as SjS (30%) and SLE (20%) [157]. Furthermore, they have been recently demonstrated in a cohort of severe juvenile myositis with lung involvement, juvenile idiopathic arthritis, but also in 12% of healthy children [158].

Despite low sensitivity, anti-cN-1A autoantibodies are high specific and highly predictive of sIBM [159] thus being of particular importance when bioptic specimens are not diagnostic.

Miscellaneous autoantibodies in IIM

Several other autoantibodies have been identified as associated with IIMs, but little is known about their clinical relevance. In fact, they are not routinely determined because easy-to-perform routine specific immunoassays still lack and they are rarely found.

Anti-RuvBL1/2

RuvBL1 (49kD) and RuvBL2 (48kD) constitute a nuclear complex involved into DNA repair and transcription. Two distinct bands of approximately 50 kDa are found in IP [160]. By means of ELISA and/or IB techniques, anti-RuvBL1/2 have been found in several CTDs, but those involved in SSc and myositis recognize different conformational epitopes identified by IP exclusively [160]. On HEp-2 cells, a fine speckled pattern is associated with these antibodies, with increased fluorescence in prophase and decreased in metaphase. Additionally, a fine speckled pattern can be found in the cytoplasm of about 40% positive sera [160]. Anti-RuvBL1/2 antibodies are highly specific for SSc, are associated with PM/SSc overlaps with diffuse cutaneous sclerosis and more frequently found in older patients of male sex [160162] or, less frequently, in necrotizing polymyositis with morphea [162].

Anti-Su/Ago2

Anti-Su/Argonaute-2 (anti-Su/Ago2) autoantibodies have been originally identified in SLE patients by means of immunodiffusion technique in the late ‘80ies [163]. Although their high prevalence in CTDs, few studies are available.

By IP, two distinct 100 and 102 kDa adjacent bands can be seen in addition to a further 200 kDa band [164]. Argonaute-2 protein constitutes the 100 kDa band and plays a key role in miRNA and interference RNA maturation and metabolism [163]. Argonaute-2 colocalized with GW bodies, a cytoplasmic organelle associated with RNA metabolism [164]. Its location and function is responsible for the particular cytoplasmic pattern of these autoantibodies also known as “GW-bodies-like” or “cytoplasmic discrete dots” (Fig. 3f) [164].

Anti-Su/Ago2 autoantibodies are frequently associated with other MSA or MAA antibodies, in particular ARS, anti-TIF-1γ and anti-MDA5 [165]; anti-Ro52 antibodies are found in almost one half of the patients [166]. It has been reported that anti-Su/Ago2 antibodies can be demonstrated in about 7.5% of the patients of Japanese origin. Apparently, there is no statistical difference between anti-Su/Ago2 positive and negative patients; however, a correlation seems to exist with ILD and absence of cancers [165].

Anti-SMN

The Survival of Motor Neuron (SMN) is a multi-ribonucleoproteic complex able to interact with the RNP-complex related D–E–F-G proteins. The SMN complex is involved into the assembly of snRNPs and co-localizes with Cajal bodies. These autoantibodies have been first described in a small number of PM patients negative for anti-U1-snRNP and/or anti-Sm but positive for RNP D–E–F-G bands by IP. This observation was indeed responsible for the identification of other SMN-complex components, namely Gemin 2 (33 kDa), Gemin 3 (130 kDa), Gemin 4 (120 kDa) and SMN itself (38 kDa) [167].

Anti-SMN antibodies typically exhibit a few nuclear dots pattern on HEp-2 cells with well distinguished 2–7 nuclear dots, similarly to anti-p80-coilin, anti-NXP2 and anti-PML pattern, seldom associated to cytoplasmic or nuclear speckled patterns (Fig. 3g).

It is not clear whether positive patients exhibit distinct clinical features. Anyhow, in the original small group of positive patients [167] and a small Italian cohort [20], a PM/SSc overlap syndrome was present. It is of note that SMN-complex genetic mutations are frequently found in neuromuscular degenerative diseases such as spinal-muscle atrophy. That is why anti-SMN autoantibodies are of relevance in basic research [168].

Anti-NPC

Nuclear pore complex (NPC) regulates protein and RNA trafficking to the nucleus. It is constituted by a complex of several proteins including Nup358/RanBP2, Nup90, Nup62 and gp210 [169]. Anti-NPC autoantibodies, and, in particular, anti gp210 are typically associated to Primary Biliary Cholangitis (PBC) and Autoimmune Hepatitis (AIH) [170]. In a cohort study from Canada, anti-NPC antibodies were found in a PM/SSc overlap syndrome and called anti-NUP Syndrome, which was found to be associated with HLA-DQ1*0501. In this case, a typical nuclear speckled laminar pattern on HEp-2 cells was observed [171] (Fig. 3h).

AMA-M2

Among the ten different anti-mitochondrial antibodies (AMA), called M1–M10, anti-M2 antibodies (AMA-M2) are the hallmark of PBC [172]. However, they can be also found in 7–12% of IIM patients without PBC [173]. AMA antibodies are readily detectable on HEp-2 cells as they display a pathognomonic cytoplasmic reticular pattern, and in triple tissue slides (Fig. 2d–f). In a Japanese study, the presence of AMA-M2 in the course of IIM was associated with muscle atrophy, granuloma formation [173] and heart involvement with high risk of supraventricular arrhythmias [174]. A distinct inflammatory phenotype associated with chronic skeletal muscle disease and severe cardiac involvement was also found in a North American cohort [175]. These associations have not been confirmed in an European series [176].

Other antibodies

Several cytoplasmic autoantibodies are described in IIM patients such as anti-KJ towards a 30/43 kDa translocation factor [177], anti-Fer directed against the elongation factor 1 and anti-Wa recognizing a 48 kDa cytoplasmic protein with still unknown function [132]. All these antibodies are typically found in anti-synthetase-like syndromes. Anti-Mas antibodies are directed against a selenocysteine-containing tRNA complex lacking any tRNA-synthetase activity but involved in protein translocation. The band of precipitation is found at 48 kDa [178]. These antibodies have been described in AIH and in a single patient with non-immune mediated rhabdomyolysis [178].

DNA-repairing complexes, especially mismatch-repair complexes such as PMS1, PMS2 and MLH1, are frequently recognized as target antigens in IIMs [179]. Initially defined as MSAs, they do indeed frequently associate with other MSAs, in particular anti-Mi-2, but they can also be found in other non-muscular diseases, such as SLE. They generally mark mild disease [180].

Anti-cortactin antibodies have been initially found in IIM patients characterized by the simultaneous presence of anti-MDA5 or anti-HMGCR antibodies by ELISA [181]. As blot confirming assay identified an unexpected 68 kDa band, it was then found that MDA5 and HMGCR extracts used in the ELISA tests were contaminated by cortactin [181]. Anti-cortactin antibodies were originally found in myasthenia gravis [182] and later in IIM patients (about 20%) and other systemic connective tissue diseases [181].

Anti-Four-and-a-Half LIM domain 1 (FHL1) antibodies were identified in about 25% of IIM patients. These antibodies associated with a severe prognosis, muscle atrophy, vasculitis, dysphagia and advanced muscular damage. Curiously, FHL1 mutations cause hereditary X-linked congenital myopathies [183].

Conclusions

Although autoantibodies are considered to be epiphenomenon in autoimmunity, their presence frequently plays a pivotal role for the diagnosis of these diseases. Indeed, several of them exhibit a pathogenethic role in IIMs. Despite this, there is still a gap between bench and bedside because the intense basic research efforts have not been translated in clinical practice, as already futuristically underlined more than 20 years ago [96, 184]. As a fact, only anti-Jo-1 have been included into the 2017 Classification Criteria for Adult and Juvenile IIMs [3].

Remarkably, in the context of heterogeneously grouped diseases such as myositis, they should be even more appreciated as able to clinically stratify patients in terms of diagnostic work-up, histological patterns, peculiar organ involvement, severity, and, therefore, treatment intensity and prognosis. This process could be accomplished by a laboratory auto-immunologist [185] well-trained in recognition of IIF ANA nuclear and, also, cytoplasmic patterns, in strict collaboration with the clinical doctor, as a decision-maker for running in-depth analysis towards the identification of the culprit autoantibody.

In addition, multicentric studies with a multidisciplinary approach may help bridging the divide of the selection bias depending on the setting where patients are initially screened (i.e. pneumologic vs. dermatologic vs. immuno-rheumatologic vs. neurologic outpatient clinics).

Abbreviations

SLE:

systemic lupus erythematosus

CTD:

connective tissue disease

SjS:

Sjögren syndrome

SSc:

systemic sclerosis

IIM:

idiopathic inflammatory myopathies

PM:

polymyositis

DM:

dermatomyositis

MSA:

myositis-specific autoantibodies

MAA:

myositis-associated autoantibodies

ARS:

anti-synthetase autoantibodies

IPAF:

interstitial pneumonia with autoimmune features

IIF:

indirect immuno-fluorescence

CIE:

counter-immuno-electrophoresis

ID:

immuno-diffusion

ELISA:

enzyme-linked immunosorbent assay

FEIA:

fluorescent enzyme-linked assay

CLIA:

chemiluminescent immuno-assay

IP:

immuno-precipitation

ALBIA:

addressable laser beads immunoassay

ANA:

anti-nuclear autoantibodies

IB:

immunoblot

LIA:

line blot assays

tRNA:

transfer RNA

DID:

double immune-diffusion

ILD:

interstitial lung disease

ASSD:

antisynthetase syndrome

PM/DM:

polymyositis/dermatomyositis

NuRD:

nucleosome remodelling deacetylase

sIBM:

sporadic inclusion body myositis

CK:

creatine kinase

SUMOs:

small ubiquitin-like modifiers

SAE:

SUMO-activating enzyme

MDA5:

Melanoma Differentiation-Associated gene 5

RIG-I:

retinoic acid-inducible gene I

RLR:

RIG-I-like receptors family

RP-ILD:

rapidly progressive interstitial lung disease

CADM:

clinically amyopathic dermatomyositis

TIF1:

transcription intermediary factors-1

TRIM:

tripartite motif-containing proteins

NXP-2:

nuclear matrix protein 2

PML:

pro-myelocitic leukaemia bodies

SRP:

signal recognition particle

IMNM:

immune-mediated necrotizing myositis

HMGCR:

3-hydroxy-3-methylglutaryl-coenzime A reductase

HALIP:

anti-HMGCR antibody associated liver immunofluorescence pattern

MTCD:

mixed connective tissue disease

sno:

small nucleolar

cN-1A:

cytosolic 5′nucleotidase

SMN:

survival of motor neuron

NPC:

nuclear pore complex

PBC:

primary biliary cholangitis

AIH:

autoimmune hepatitis

FHL1:

anti-four-and-a-half LIM domain 1

AMA:

anti-mitochondrial antibodies

References

  1. Petri M, Orbai A-M, Alarcón GS, Gordon C, Merrill JT, Fortin PR, et al. Derivation and validation of the Systemic Lupus International Collaborating Clinics classification criteria for systemic lupus erythematosus. Arthritis Rheum. 2012;64:2677–86.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Shiboski CH, Shiboski SC, Seror R, Criswell LA, Labetoulle M, Lietman TM, et al. 2016 American College of Rheumatology/European League Against Rheumatism classification criteria for primary Sjögren’s syndrome. Ann Rheum Dis. 2017;76:9–16.

    Article  PubMed  Google Scholar 

  3. Lundberg IE, Tjärnlund A, Bottai M, Werth VP, Pilkington C, de Visser M, et al. 2017 European League Against Rheumatism/American College of Rheumatology classification criteria for adult and juvenile idiopathic inflammatory myopathies and their major subgroups. Ann Rheum Dis. 2017;76:1955–64.

    Article  PubMed  Google Scholar 

  4. Milone M. Diagnosis and Management of Immune-Mediated Myopathies. Mayo Clin Proc. 2017;92:826–37.

    Article  PubMed  Google Scholar 

  5. Targoff IN. Autoantibodies in polymyositis. Rheum Dis Clin North Am. 1992;18:455–82.

    CAS  PubMed  Google Scholar 

  6. Targoff IN. Idiopathic inflammatory myopathy: autoantibody update. Curr Rheumatol Rep. 2002;4:434–41.

    Article  PubMed  Google Scholar 

  7. Satoh M, Tanaka S, Ceribelli A, Calise SJ, Chan EKL. A comprehensive overview on myositis-specific antibodies: new and old biomarkers in idiopathic inflammatory myopathy. Clin Rev Allergy Immunol. 2015;52:1–9.

    Article  CAS  Google Scholar 

  8. Nakashima R, Imura Y, Hosono Y, Seto M, Murakami A, Watanabe K, et al. The multicenter study of a new assay for simultaneous detection of multiple anti-aminoacyl-tRNA synthetases in myositis and interstitial pneumonia. PLoS ONE. 2014;9:e85062.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Colafrancesco S, Priori R, Valesini G. Inflammatory myopathies and overlap syndromes: update on histological and serological profile. Best Pract Res Clin Rheumatol. 2015;29:810–25.

    Article  PubMed  Google Scholar 

  10. Gunawardena H, Betteridge ZE, McHugh NJ. Myositis-specific autoantibodies: their clinical and pathogenic significance in disease expression. Rheumatology (Oxford). 2009;48:607–12.

    Article  CAS  Google Scholar 

  11. Chinoy H, Fertig N, Oddis CV, Ollier WER, Cooper RG. The diagnostic utility of myositis autoantibody testing for predicting the risk of cancer-associated myositis. Ann Rheum Dis. 2007;66:1345–9.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Frank MB, McCubbin V, Trieu E, Wu Y, Isenberg DA, Targoff IN. The association of anti-Ro52 autoantibodies with myositis and scleroderma autoantibodies. J Autoimmun. 1999;12:137–42.

    Article  CAS  PubMed  Google Scholar 

  13. Ferreira JP, Almeida I, Marinho A, Cerveira C, Vasconcelos C. Anti-ro52 antibodies and interstitial lung disease in connective tissue diseases excluding scleroderma. ISRN Rheumatol. 2012;2012:415272.

    Article  PubMed  PubMed Central  Google Scholar 

  14. van Dooren SHJ, van Venrooij WJ, Pruijn GJM. Myositis-specific autoantibodies: detection and clinical associations. Autoimmun Highlights. 2011;2:5–20.

    Article  CAS  Google Scholar 

  15. Chan EKL, Damoiseaux J, Carballo OG, Conrad K, de Melo Cruvinel W, Francescantonio PLC, et al. Report of the first international consensus on standardized nomenclature of antinuclear antibody HEp-2 cell patterns 2014–2015. Front Immunol. 2015;6:412.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. García-DeLaTorre I. Clinical usefulness of autoantibodies in idiopathic inflammatory myositis. Front Immunol. 2015;6:331.

    Google Scholar 

  17. Damoiseaux J, von Mühlen CA, Garcia-De La Torre I, Carballo OG, de Melo Cruvinel W, Francescantonio PLC, et al. International consensus on ANA patterns (ICAP): the bumpy road towards a consensus on reporting ANA results. Autoimmun Highlights. 2016;7:1–8.

    Article  CAS  Google Scholar 

  18. Ghirardello A, Bendo R, Rampudda ME, Bassi N, Zampieri S, Doria A. Commercial blot assays in the diagnosis of systemic rheumatic diseases. Autoimmun Rev. 2009;8:645–9.

    Article  CAS  PubMed  Google Scholar 

  19. Rönnelid J, Barbasso Helmers S, Storfors H, Grip K, Rönnblom L, Franck-Larsson K, et al. Use of a commercial line blot assay as a screening test for autoantibodies in inflammatory myopathies. Autoimmun Rev. 2009;9:58–61.

    Article  PubMed  CAS  Google Scholar 

  20. Cavazzana I, Fredi M, Ceribelli A, Mordenti C, Ferrari F, Carabellese N, et al. Testing for myositis specific autoantibodies: comparison between line blot and immunoprecipitation assays in 57 myositis sera. J Immunol Methods. 2016;433:1–5.

    Article  CAS  PubMed  Google Scholar 

  21. Ceribelli A, Satoh M, Chan EK. A new immunoprecipitation-real time quantitative PCR assay for anti-Th/To and anti-U3RNP antibody detection in systemic sclerosis. Arthritis Res Ther. 2012;14:R128.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Mahler M, Miller FW, Fritzler MJ. Idiopathic inflammatory myopathies and the anti-synthetase syndrome: a comprehensive review. Autoimmun Rev. 2014;13:367–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Nishikai M, Reichlin M. Heterogeneity of precipitating antibodies in polymyositis and dermatomyositis. Characterization of the Jo-1 antibody system. Arthritis Rheum. 1980;23:881–8.

    Article  CAS  PubMed  Google Scholar 

  24. Agmon-Levin N, Damoiseaux J, Kallenberg C, Sack U, Witte T, Herold M, et al. International recommendations for the assessment of autoantibodies to cellular antigens referred to as anti-nuclear antibodies. Ann Rheum Dis. 2014;73:17–23.

    Article  CAS  PubMed  Google Scholar 

  25. Infantino M, Palterer B, Biagiotti R, et al. Reflex testing of speckled cytoplasmic patterns observed in routine ANA HEp-2 indirect immunofluorescence with a multiplex anti-synthetase dot-blot assay: a multicentric pilot study. Immunol Res. 2018;66(1):74–8.

    Article  CAS  PubMed  Google Scholar 

  26. Abe T, Tsunoda S, Nishioka A, Azuma K, Tsuboi K, Ogita C, et al. Reliability and clinical utility of enzyme-linked immunosorbent assay for detection of anti-aminoacyl-tRNA synthetase antibody. Nihon Rinsho Meneki Gakkai Kaishi. 2016;39:140–4.

    Article  PubMed  Google Scholar 

  27. Yoshida S, Akizuki M, Mimori T, Yamagata H, Inada S, Homma M. The precipitating antibody to an acidic nuclear protein antigen, the Jo-1, in connective tissue diseases. A marker for a subset of polymyositis with interstitial pulmonary fibrosis. Arthritis Rheum. 1983;26:604–11.

    Article  CAS  PubMed  Google Scholar 

  28. Yamasaki Y, Satoh M, Mizushima M, Okazaki T, Nagafuchi H, Ooka S, et al. Clinical subsets associated with different anti-aminoacyl transfer RNA synthetase antibodies and their association with coexisting anti-Ro52. Mod Rheumatol. 2016;26:403–9.

    Article  CAS  PubMed  Google Scholar 

  29. Yamasaki Y, Yamada H, Nozaki T, Akaogi J, Nichols C, Lyons R, et al. Unusually high frequency of autoantibodies to PL-7 associated with milder muscle disease in Japanese patients with polymyositis/dermatomyositis. Arthritis Rheum. 2006;54:2004–9.

    Article  CAS  PubMed  Google Scholar 

  30. Marie I, Josse S, Decaux O, Diot E, Landron C, Roblot P, et al. Clinical manifestations and outcome of anti-PL7 positive patients with antisynthetase syndrome. Eur J Intern Med. 2013;24:474–9.

    Article  CAS  PubMed  Google Scholar 

  31. Targoff IN, Arnett FC. Clinical manifestations in patients with antibody to PL-12 antigen (alanyl-tRNA synthetase). Am J Med. 1990;88:241–51.

    Article  CAS  PubMed  Google Scholar 

  32. Hirakata M, Suwa A, Nagai S, Kron MA, Trieu EP, Mimori T, et al. Anti-KS: identification of autoantibodies to asparaginyl-transfer RNA synthetase associated with interstitial lung disease. J Immunol. 1999;162:2315–20.

    CAS  PubMed  Google Scholar 

  33. Scirè CA, Gonzalez-Gay MA, Selva-O’Callaghan A, Cavagna L, Selva-O’Callaghan A, Cavagna L. Clinical spectrum time course of interstitial pneumonia with autoimmune features in patients positive for antisynthetase antibodies. Respir Med. 2017;132:265–6.

    Article  PubMed  Google Scholar 

  34. Cavagna L, Nuño L, Scirè CA, Govoni M, Longo FJL, Franceschini F, et al. Clinical spectrum time course in anti Jo-1 positive antisynthetase syndrome: results from an international retrospective multicenter study. Medicine (Baltimore). 2015;94:e1144.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Casal-Dominguez M, Pinal-Fernandez I, Mego M, Accarino A, Jubany L, Azpiroz F, et al. High-resolution manometry in patients with idiopathic inflammatory myopathy: Elevated prevalence of esophageal involvement and differences according to autoantibody status and clinical subset. Muscle Nerve. 2016;45(suppl_4):iv18–21.

    Google Scholar 

  36. Pestronk A. Acquired immune and inflammatory myopathies. Curr Opin Rheumatol. 2011;23:595–604.

    Article  CAS  PubMed  Google Scholar 

  37. Mescam-Mancini L, Allenbach Y, Hervier B, Devilliers H, Mariampillay K, Dubourg O, et al. Anti-Jo-1 antibody-positive patients show a characteristic necrotizing perifascicular myositis. Brain. 2015;138(Pt 9):2485–92.

    Article  PubMed  Google Scholar 

  38. Aouizerate J, De Antonio M, Bassez G, Gherardi RK, Berenbaum F, Guillevin L, et al. Myofiber HLA-DR expression is a distinctive biomarker for antisynthetase-associated myopathy. Acta Neuropathol Commun. 2014;2:154.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Targoff IN, Reichlin M. The association between Mi-2 antibodies and dermatomyositis. Arthritis Rheum. 1985;28:796–803.

    Article  CAS  PubMed  Google Scholar 

  40. Ghirardello A, Zampieri S, Iaccarino L, Tarricone E, Bendo R, Gambari PF, et al. Anti-Mi-2 antibodies. Autoimmunity. 2005;38:79–83.

    Article  CAS  PubMed  Google Scholar 

  41. Nilasena DS, Trieu EP, Targoff IN. Analysis of the Mi-2 autoantigen of dermatomyositis. Arthritis Rheum. 1995;38:123–8.

    Article  CAS  PubMed  Google Scholar 

  42. Zhang Y, LeRoy G, Seelig HP, Lane WS, Reinberg D. The dermatomyositis-specific autoantigen Mi2 is a component of a complex containing histone deacetylase and nucleosome remodeling activities. Cell. 1998;95:279–89.

    Article  CAS  PubMed  Google Scholar 

  43. Petri MH, Satoh M, Martin-Marquez BT, Vargas-Ramírez R, Jara LJ, Saavedra MA, et al. Implications in the difference of anti-Mi-2 and -p155/140 autoantibody prevalence in two dermatomyositis cohorts from Mexico City and Guadalajara. Arthritis Res Ther. 2013;15:R48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Love LA, Weinberg CR, McConnaughey DR, Oddis CV, Medsger TA, Reveille JD, et al. Ultraviolet radiation intensity predicts the relative distribution of dermatomyositis and anti-Mi-2 autoantibodies in women. Arthritis Rheum. 2009;60:2499–504.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Prieto S, Grau JM. The geoepidemiology of autoimmune muscle disease. Autoimmun Rev. 2010;9:A330–4.

    Article  PubMed  Google Scholar 

  46. Tarricone E, Ghirardello A, Rampudda M, Bassi N, Punzi L, Doria A. Anti-SAE antibodies in autoimmune myositis: identification by unlabelled protein immunoprecipitation in an Italian patient cohort. J Immunol Methods. 2012;384:128–34.

    Article  CAS  PubMed  Google Scholar 

  47. Betteridge Z, Gunawardena H, North J, Slinn J, McHugh N. Identification of a novel autoantibody directed against small ubiquitin-like modifier activating enzyme in dermatomyositis. Arthritis Rheum. 2007;56:3132–7.

    Article  CAS  PubMed  Google Scholar 

  48. Ge Y, Lu X, Shu X, Peng Q, Wang G. Clinical characteristics of anti-SAE antibodies in Chinese patients with dermatomyositis in comparison with different patient cohorts. Sci Rep. 2017;7:188.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Muro Y, Sugiura K, Akiyama M. Low prevalence of anti-small ubiquitin-like modifier activating enzyme antibodies in dermatomyositis patients. Autoimmunity. 2013;46:279–84.

    Article  CAS  PubMed  Google Scholar 

  50. Fujimoto M, Matsushita T, Hamaguchi Y, Kaji K, Asano Y, Ogawa F, et al. Autoantibodies to small ubiquitin-like modifier activating enzymes in Japanese patients with dermatomyositis: comparison with a UK Caucasian cohort. Ann Rheum Dis. 2013;72:151–3.

    Article  PubMed  Google Scholar 

  51. Betteridge ZE, Gunawardena H, Chinoy H, North J, Ollier WER, Cooper RG, et al. Clinical and human leucocyte antigen class II haplotype associations of autoantibodies to small ubiquitin-like modifier enzyme, a dermatomyositis-specific autoantigen target, in UK Caucasian adult-onset myositis. Ann Rheum Dis. 2009;68:1621–5.

    Article  CAS  PubMed  Google Scholar 

  52. Muro Y, Sugiura K, Nara M, Sakamoto I, Suzuki N, Akiyama M. High incidence of cancer in anti-small ubiquitin-like modifier activating enzyme antibody-positive dermatomyositis. Rheumatology (Oxford). 2015;54:1745–7.

    Article  Google Scholar 

  53. Nakashima R, Imura Y, Kobayashi S, Yukawa N, Yoshifuji H, Nojima T, et al. The RIG-I-like receptor IFIH1/MDA5 is a dermatomyositis-specific autoantigen identified by the anti-CADM-140 antibody. Rheumatology (Oxford). 2010;49:433–40.

    Article  CAS  Google Scholar 

  54. Sato S, Hirakata M, Kuwana M, Suwa A, Inada S, Mimori T, et al. Autoantibodies to a 140-kd polypeptide, CADM-140, in Japanese patients with clinically amyopathic dermatomyositis. Arthritis Rheum. 2005;52:1571–6.

    Article  CAS  PubMed  Google Scholar 

  55. Sato S, Hoshino K, Satoh T, Fujita T, Kawakami Y, Fujita T, et al. RNA helicase encoded by melanoma differentiation-associated gene 5 is a major autoantigen in patients with clinically amyopathic dermatomyositis: association with rapidly progressive interstitial lung disease. Arthritis Rheum. 2009;60:2193–200.

    Article  CAS  PubMed  Google Scholar 

  56. Parronchi P, Radice A, Palterer B, Liotta F, Scaletti C. MDA5-positive dermatomyositis: an uncommon entity in Europe with variable clinical presentations. Clin Mol Allergy. 2015;13:22.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Zhang L, Wu G, Gao D, Liu G, Pan L, Ni L, et al. Factors Associated with interstitial lung disease in patients with polymyositis and dermatomyositis: a systematic review and meta-analysis. PLoS ONE. 2016;11:e0155381.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Chen Z, Hu W, Wang Y, Guo Z, Sun L, Kuwana M. Distinct profiles of myositis-specific autoantibodies in Chinese and Japanese patients with polymyositis/dermatomyositis. Clin Rheumatol. 2015;34:1627–31.

    Article  PubMed  Google Scholar 

  59. Ceribelli A, Fredi M, Taraborelli M, Cavazzana I, Tincani A, Selmi C, et al. Prevalence and clinical significance of anti-MDA5 antibodies in European patients with polymyositis/dermatomyositis. Clin Exp Rheumatol. 2014;32:891–7.

    PubMed  Google Scholar 

  60. Labrador-Horrillo M, Martinez MA, Selva-O’Callaghan A, Trallero-Araguas E, Balada E, Vilardell-Tarres M, et al. Anti-MDA5 antibodies in a large Mediterranean population of adults with dermatomyositis. J Immunol Res. 2014;2014:290797.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Hall JC, Casciola-Rosen L, Samedy L-A, Werner J, Owoyemi K, Danoff SK, et al. Anti-melanoma differentiation-associated protein 5-associated dermatomyositis: expanding the clinical spectrum. Arthritis Care Res (Hoboken). 2013;65:1307–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Muro Y, Sugiura K, Hoshino K, Akiyama M, Tamakoshi K. Epidemiologic study of clinically amyopathic dermatomyositis and anti-melanoma differentiation-associated gene 5 antibodies in central Japan. Arthritis Res Ther. 2011;13:R214.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Chen Z, Wang Y, Kuwana M, Xu X, Hu W, Feng X, et al. HLA-DRB1 alleles as genetic risk factors for the development of anti-MDA5 antibodies in patients with dermatomyositis. J Rheumatol. 2017;44:1389–93.

    Article  PubMed  Google Scholar 

  64. Ward I, Hiles P, Arroyo R, Downs W, Bell D. Digital pulp ulcerations and inverse gottron papules in melanoma differentiation-associated gene 5-related dermatomyositis. J Clin Rheumatol. 2016;22:274–5.

    Article  PubMed  Google Scholar 

  65. Cao H, Xia Q, Pan M, Zhao X, Li X, Shi R, et al. Gottron papules and gottron sign with ulceration: a distinctive cutaneous feature in a subset of patients with classic dermatomyositis and clinically amyopathic dermatomyositis. J Rheumatol. 2016;43:1735–42.

    Article  CAS  PubMed  Google Scholar 

  66. Fiorentino D, Chung L, Zwerner J, Rosen A, Casciola-Rosen L. The mucocutaneous and systemic phenotype of dermatomyositis patients with antibodies to MDA5 (CADM-140): a retrospective study. J Am Acad Dermatol. 2011;65:25–34.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Tansley SL, Simou S, Shaddick G, Betteridge ZE, Almeida B, Gunawardena H, 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Gono T, Kawaguchi Y, Hara M, Masuda I, Katsumata Y, Shinozaki M, et al. Increased ferritin predicts development and severity of acute interstitial lung disease as a complication of dermatomyositis. Rheumatology (Oxford). 2010;49:1354–60.

    Article  CAS  Google Scholar 

  69. Muro Y, Sugiura K, Akiyama M. Limitations of a single-point evaluation of anti-MDA5 antibody, ferritin, and IL-18 in predicting the prognosis of interstitial lung disease with anti-MDA5 antibody-positive dermatomyositis. Clin Rheumatol. 2013;32:395–8.

    Article  PubMed  Google Scholar 

  70. Fujimoto M, Hamaguchi Y, Kaji K, Matsushita T, Ichimura Y, Kodera M, et al. Myositis-specific anti-155/140 autoantibodies target transcription intermediary factor 1 family proteins. Arthritis Rheum. 2012;64:513–22.

    Article  CAS  PubMed  Google Scholar 

  71. Targoff IN, Mamyrova G, Trieu EP, Perurena O, Koneru B, O’Hanlon TP, et al. A novel autoantibody to a 155-kd protein is associated with dermatomyositis. Arthritis Rheum. 2006;54:3682–9.

    Article  CAS  PubMed  Google Scholar 

  72. Kaji K, Fujimoto M, Hasegawa M, Kondo M, Saito Y, Komura K, et al. Identification of a novel autoantibody reactive with 155 and 140 kDa nuclear proteins in patients with dermatomyositis: an association with malignancy. Rheumatology (Oxford). 2007;46:25–8.

    Article  CAS  Google Scholar 

  73. Satoh M, Chan JYF, Ross SJ, Li Y, Yamasaki Y, Yamada H, et al. Autoantibodies to transcription intermediary factor TIF1β associated with dermatomyositis. Arthritis Res Ther. 2012;14:R79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Labrador-Horrillo M, Martínez MA, Selva-O’Callaghan A, Trallero-Araguás E, Balada E, Vilardell-Tarrés M, et al. Anti-TIF1γ antibodies (anti-p155) in adult patients with dermatomyositis: comparison of different diagnostic assays. Ann Rheum Dis. 2012;71:993–6.

    Article  CAS  PubMed  Google Scholar 

  75. Muro Y, Ishikawa A, Sugiura K, Akiyama M. Clinical features of anti-TIF1-α antibody-positive dermatomyositis patients are closely associated with coexistent dermatomyositis-specific autoantibodies and anti-TIF1-γ or anti-Mi-2 autoantibodies. Rheumatology (Oxford). 2012;51:1508–13.

    Article  CAS  Google Scholar 

  76. Kasuya A, Hamaguchi Y, Fujimoto M, Tokura Y. TIF1γ-overexpressing, highly progressive endometrial carcinoma in a patient with dermato-myositis positive for malignancy-associated anti-p155/140 autoantibody. Acta Derm Venereol. 2013;93:715–6.

    Article  PubMed  Google Scholar 

  77. Mohassel P, Rosen P, Casciola-Rosen L, Pak K, Mammen AL. Expression of the dermatomyositis autoantigen transcription intermediary factor 1γ in regenerating muscle. Arthritis Rheumatol. 2015;67:266–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Trallero-Araguás E, Rodrigo-Pendás JÁ, Selva-O’Callaghan A, Martínez-Gõmez X, Bosch X, Labrador-Horrillo M, et al. Usefulness of anti-p155 autoantibody for diagnosing cancer-associated dermatomyositis: a systematic review and meta-analysis. Arthritis Rheum. 2012;64:523–32.

    Article  PubMed  CAS  Google Scholar 

  79. Palterer B, Vitiello G, Cammelli D. First report of anti-TIF1γ dermatomyositis in a patient with myelodysplastic syndrome. Reumatismo. 2017;69:75–7.

    Article  CAS  PubMed  Google Scholar 

  80. Fiorentino DF, Kuo K, Chung L, Zaba L, Li S, Casciola-Rosen L. Distinctive cutaneous and systemic features associated with antitranscriptional intermediary factor-1γ antibodies in adults with dermatomyositis. J Am Acad Dermatol. 2015;72:449–55.

    Article  PubMed  PubMed Central  Google Scholar 

  81. Bernet LL, Lewis MA, Rieger KE, Casciola-Rosen L, Fiorentino DF. Ovoid palatal patch in dermatomyositis: a novel finding associated with anti-TIF1γ (p155) antibodies. JAMA Dermatol. 2016;152:1049–51.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Kimura Y, Sakai F, Nakano O, Kisaki O, Sugimoto H, Sawamura T, et al. The newly identified human nuclear protein NXP-2 possesses three distinct domains, the nuclear matrix-binding, RNA-binding, and coiled-coil domains. J Biol Chem. 2002;277:20611–7.

    Article  CAS  PubMed  Google Scholar 

  83. Targoff IN, Trieu EP, Levy-Neto M. Sera with autoantibodies to the MJ antigen react with NXP2. Arthritis Rheum. 2007;56:S787.

    Google Scholar 

  84. Fredi M, Bartoli F, Cavazzana I, Ceribelli A, Carabellese N, Tincani A, et al. Calcinosis in poly-dermatomyositis: clinical and laboratory predictors and treatment options. Clin Exp Rheumatol. 2017;35:303–8.

    PubMed  Google Scholar 

  85. Mimura Y, Takahashi K, Kawata K, Akazawa T, Inoue N. Two-step colocalization of MORC3 with PML nuclear bodies. J Cell Sci. 2010;123(Pt 12):2014–24.

    Article  CAS  PubMed  Google Scholar 

  86. Espada G, Maldonado Cocco JA, Fertig N, Oddis CV. Clinical and serologic characterization of an Argentine pediatric myositis cohort: identification of a novel autoantibody (anti-MJ) to a 142-kDa protein. J Rheumatol. 2009;36:2547–51.

    Article  CAS  PubMed  Google Scholar 

  87. Ichimura Y, Matsushita T, Hamaguchi Y, Kaji K, Hasegawa M, Tanino Y, et al. Anti-NXP2 autoantibodies in adult patients with idiopathic inflammatory myopathies: possible association with malignancy. Ann Rheum Dis. 2012;71:710–3.

    Article  CAS  PubMed  Google Scholar 

  88. Fiorentino DF, Chung LS, Christopher-Stine L, Zaba L, Li S, Mammen AL, et al. Most patients with cancer-associated dermatomyositis have antibodies to nuclear matrix protein NXP-2 or transcription intermediary factor 1γ. Arthritis Rheum. 2013;65:2954–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Ceribelli A, Fredi M, Taraborelli M, Cavazzana I, Franceschini F, Quinzanini M, et al. Anti-MJ/NXP-2 autoantibody specificity in a cohort of adult Italian patients with polymyositis/dermatomyositis. Arthritis Res Ther. 2012;14:R97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Tansley SL, Betteridge ZE, Shaddick G, Gunawardena H, Arnold K, Wedderburn LR, et al. Calcinosis in juvenile dermatomyositis is influenced by both anti-NXP2 autoantibody status and age at disease onset. Rheumatology (Oxford). 2014;53:2204–8.

    Article  Google Scholar 

  91. Reeves WH, Nigam SK, Blobel G. Human autoantibodies reactive with the signal-recognition particle. Proc Natl Acad Sci USA. 1986;83:9507–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Picard C, Vincent T, Lega J-CC, Hue S, Fortenfant F, Lakomy D, et al. Heterogeneous clinical spectrum of anti-SRP myositis and importance of the methods of detection of anti-SRP autoantibodies: a multicentric study. Immunol Res. 2016;64:677–86.

    Article  CAS  PubMed  Google Scholar 

  93. Suzuki S, Nishikawa A, Kuwana M, Nishimura H, Watanabe Y, Nakahara J, et al. Inflammatory myopathy with anti-signal recognition particle antibodies: case series of 100 patients. Orphanet J Rare Dis. 2015;10:61.

    Article  PubMed  PubMed Central  Google Scholar 

  94. Allenbach Y, Arouche-Delaperche L, Preusse C, et al. Necrosis in anti-SRP+ and anti-HMGCR+myopathies: role of autoantibodies and complement. Neurology. 2018;90(6):e507–17.

    Article  CAS  PubMed  Google Scholar 

  95. Ikeda K, Mori-Yoshimura M, Yamamoto T, Sonoo M, Suzuki S, Kondo Y, et al. Chronic myopathy associated with anti-signal recognition particle antibodies can be misdiagnosed as facioscapulohumeral muscular dystrophy. J Clin Neuromuscul Dis. 2016;17:197–206.

    Article  PubMed  Google Scholar 

  96. Love LA, Leff RL, Fraser DD, Targoff IN, Dalakas M, Plotz PH, et al. A new approach to the classification of idiopathic inflammatory myopathy: myositis-specific autoantibodies define useful homogeneous patient groups. Medicine (Baltimore). 1991;70:360–74.

    Article  CAS  PubMed  Google Scholar 

  97. Aggarwal R, Oddis CV, Goudeau D, Fertig N, Metes I, Stephens C, et al. Anti-signal recognition particle autoantibody ELISA validation and clinical associations. Rheumatology. 2015;54:1194–9.

    Article  CAS  PubMed  Google Scholar 

  98. Suzuki S, Yonekawa T, Kuwana M, Hayashi YK, Okazaki Y, Kawaguchi Y, et al. Clinical and histological findings associated with autoantibodies detected by RNA immunoprecipitation in inflammatory myopathies. J Neuroimmunol. 2014;274:202–8.

    Article  CAS  PubMed  Google Scholar 

  99. Wang L, Liu L, Hao H, Gao F, Liu X, Wang Z, et al. Myopathy with anti-signal recognition particle antibodies: clinical and histopathological features in Chinese patients. Neuromuscul Disord. 2014;24:335–41.

    Article  CAS  PubMed  Google Scholar 

  100. Ellis E, Ann Tan J, Lester S, Tucker G, Blumbergs P, Roberts-Thomson P, et al. Necrotizing myopathy: clinicoserologic associations. Muscle Nerve. 2012;45:189–94.

    Article  PubMed  Google Scholar 

  101. Kao AH, Lacomis D, Lucas M, Fertig N, Oddis CV. Anti-signal recognition particle autoantibody in patients with and patients without idiopathic inflammatory myopathy. Arthritis Rheum. 2004;50:209–15.

    Article  CAS  PubMed  Google Scholar 

  102. Takada T, Hirakata M, Suwa A, Kaneko Y, Kuwana M, Ishihara T, et al. Clinical and histopathological features of myopathies in Japanese patients with anti-SRP autoantibodies. Mod Rheumatol. 2009;19:156–64.

    Article  PubMed  Google Scholar 

  103. Togawa R, Tanino Y, Nikaido T, Fukuhara N, Uematsu M, Misa K, et al. Three cases of interstitial pneumonia with anti-signal recognition particle antibody. Allergol Int. 2017;66:485–7.

    Article  PubMed  Google Scholar 

  104. Sugie K, Tonomura Y, Ueno S. Characterization of dermatomyositis with coexistence of anti-Jo-1 and anti-SRP antibodies. Intern Med. 2012;51:799–802.

    Article  PubMed  Google Scholar 

  105. Malkan A, Cappelen-Smith C, Beran R, Griffith N, Toong C, Wang M-X, et al. Anti-synthetase syndrome associated with anti PL-12 and anti-Signal recognition particle antibodies and a necrotizing auto-immune myositis. J Clin Neurosci. 2015;22:396–8.

    Article  CAS  PubMed  Google Scholar 

  106. Benveniste O, Drouot L, Jouen F, Charuel J-L, Bloch-Queyrat C, Behin A, et al. Correlation of anti-signal recognition particle autoantibody levels with creatine kinase activity in patients with necrotizing myopathy. Arthritis Rheum. 2011;63:1961–71.

    Article  CAS  PubMed  Google Scholar 

  107. Christopher-Stine L, Casciola-Rosen LA, Hong G, Chung T, Corse AM, Mammen AL. A novel autoantibody recognizing 200-kd and 100-kd proteins is associated with an immune-mediated necrotizing myopathy. Arthritis Rheum. 2010;62:2757–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Alvarado-Cardenas M, Marin-Sánchez A, Martínez MA, Martínez-Martínez L, Pinal-Fernandez I, Labrador-Horrillo M, et al. Statin-associated autoimmune myopathy: a distinct new IFL pattern can increase the rate of HMGCR antibody detection by clinical laboratories. Autoimmun Rev. 2016;15:1161–6.

    Article  CAS  PubMed  Google Scholar 

  109. Palterer B, Cammelli D, Vitiello G, Giudizi MGMG, Cammelli D, Giudizi MGMG, et al. Anti-HMGCR and anti-DFS70 antibodies immunofluorescence patterns. Autoimmune Dis. 2017;16:321–2.

    CAS  Google Scholar 

  110. Musset L, Miyara M, Benveniste O, Charuel J-L, Shikhman A, Boyer O, et al. Analysis of autoantibodies to 3-hydroxy-3-methylglutaryl-coenzyme A reductase using different technologies. J Immunol Res. 2014;2014:1–8.

    Article  CAS  Google Scholar 

  111. Musset L, Allenbach Y, Benveniste O, Boyer O, Bossuyt X, Bentow C, et al. Anti-HMGCR antibodies as a biomarker for immune-mediated necrotizing myopathies: a history of statins and experience from a large international multi-center study. Autoimmun Rev. 2016;15:983–93.

    Article  CAS  PubMed  Google Scholar 

  112. Alshehri A, Choksi R, Bucelli R, Pestronk A. Myopathy with anti-HMGCR antibodies. Neurol Neuroimmunol Neuroinflamm. 2015;2:e124.

    Article  PubMed  PubMed Central  Google Scholar 

  113. Keating P, Young J, George P, Florkowski C, Spellerberg M, Kennedy N. Anti-HMGCR autoantibodies in self-limiting statin-induced myopathy. Int J Rheum Dis. 2017;20:2179–81.

    Article  PubMed  Google Scholar 

  114. Kadoya M, Hida A, Hashimoto Maeda M, Taira K, Ikenaga C, Uchio N, et al. Cancer association as a risk factor for anti-HMGCR antibody-positive myopathy. Neurol Neuroimmunol Neuroinflamm. 2016;3:e290.

    Article  PubMed  PubMed Central  Google Scholar 

  115. Mammen AL, Gaudet D, Brisson D, Christopher-Stine L, Lloyd TE, Leffell MS, et al. Increased frequency of DRB1*11:01 in anti-HMG-CoA reductase-associated autoimmune myopathy. Arthritis Care Res (Hoboken). 2012;64:1233–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Giudizi M, Cammelli D, Vivarelli E, Biagiotti R, Ferraro A, Bentow C, et al. Anti-HMGCR antibody-associated necrotizing myopathy: diagnosis and treatment illustrated using a case report. Scand J Rheumatol. 2016;45:427–9.

    Article  CAS  PubMed  Google Scholar 

  117. Tiniakou E, Pinal-Fernandez I, Lloyd TE, Albayda J, Paik J, Werner JL, et al. More severe disease and slower recovery in younger patients with anti-3-hydroxy-3-methylglutaryl-coenzyme A reductase-associated autoimmune myopathy. Rheumatology (Oxford). 2017;56:787–94.

    Google Scholar 

  118. Rider LG, Shah M, Mamyrova G, Huber AM, Rice MM, Targoff IN, et al. The myositis autoantibody phenotypes of the juvenile idiopathic inflammatory myopathies. Medicine (Baltimore). 2013;92:223–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Mahler M, Fritzler MJ. PM1-Alpha ELISA: the assay of choice for the detection of anti-PM/Scl autoantibodies? Autoimmun Rev. 2009;8:373–8.

    Article  CAS  PubMed  Google Scholar 

  120. Jaskowski TD, Wilson A, Hill HR, Tebo AE. Diagnostic assays for Anti-PM/Scl IgG antibodies: heterogeneity in antibody response or lack of standardization? Clin Chim Acta. 2011;412:1100–5.

    Article  CAS  PubMed  Google Scholar 

  121. Lega J-CC, Fabien N, Reynaud Q, Durieu I, Durupt SS, Dutertre M, et al. The clinical phenotype associated with myositis-specific and associated autoantibodies: a meta-analysis revisiting the so-called antisynthetase syndrome. Autoimmun Rev. 2014;13:883–91.

    Article  CAS  PubMed  Google Scholar 

  122. Mahler M, Raijmakers R. Novel aspects of autoantibodies to the PM/Scl complex: clinical, genetic and diagnostic insights. Autoimmun Rev. 2007;6:432–7.

    Article  CAS  PubMed  Google Scholar 

  123. Muro Y, Hosono Y, Sugiura K, Ogawa Y, Mimori T, Akiyama M. Anti-PM/Scl antibodies are found in Japanese patients with various systemic autoimmune conditions besides myositis and scleroderma. Arthritis Res Ther. 2015;17:57.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  124. Aguila LA, Lopes MRU, Pretti FZ, Sampaio-Barros PD, Carlos De Souza FH, Borba EF, et al. Clinical and laboratory features of overlap syndromes of idiopathic inflammatory myopathies associated with systemic lupus erythematosus, systemic sclerosis, or rheumatoid arthritis. Clin Rheumatol. 2014;33:1093–8.

    Article  PubMed  Google Scholar 

  125. Hanke K, Brückner CS, Dähnrich C, Huscher D, Komorowski L, Meyer W, et al. Antibodies against PM/Scl-75 and PM/Scl-100 are independent markers for different subsets of systemic sclerosis patients. Arthritis Res Ther. 2009;11:R22.

    Article  PubMed  PubMed Central  Google Scholar 

  126. Schilders G, Egberts WV, Raijmakers R, Pruijn GJM. C1D is a major autoantibody target in patients with the polymyositis-scleroderma overlap syndrome. Arthritis Rheum. 2007;56:2449–54.

    Article  CAS  PubMed  Google Scholar 

  127. Migliorini P, Baldini C, Rocchi V, Bombardieri S. Anti-Sm and anti-RNP antibodies. Autoimmunity. 2005;38:47–54.

    Article  CAS  PubMed  Google Scholar 

  128. Lundberg I, Nennesmo I, Hedfors E. A clinical, serological, and histopathological study of myositis patients with and without anti-RNP antibodies. Semin Arthritis Rheum. 1992;22:127–38.

    Article  CAS  PubMed  Google Scholar 

  129. Coppo P, Clauvel JP, Bengoufa D, Oksenhendler E, Lacroix C, Lassoued K. Inflammatory myositis associated with anti-U1-small nuclear ribonucleoprotein antibodies: a subset of myositis associated with a favourable outcome. Rheumatology (Oxford). 2002;41:1040–6.

    Article  CAS  Google Scholar 

  130. Craft J, Mimori T, Olsen TL, Hardin JA. The U2 small nuclear ribonucleoprotein particle as an autoantigen. Analysis with sera from patients with overlap syndromes. J Clin Invest. 1988;81:1716–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Kubo M, Ihn H, Kuwana M, Asano Y, Tamaki T, Yamane K, et al. Anti-U5 snRNP antibody as a possible serological marker for scleroderma-polymyositis overlap. Rheumatology (Oxford). 2002;41:531–4.

    Article  CAS  Google Scholar 

  132. Sibilia J, Chatelus E, Meyer A, Gottenberg J-E, Sordet C, Goetz J. How can we diagnose and better understand inflammatory myopathies? The usefulness of auto-antibodies. Presse Med. 2010;39:1010–25.

    Article  PubMed  Google Scholar 

  133. Van Eenennaam H, Vogelzangs JHP, Bisschops L, Te Boome LCJ, Seelig HP, Renz M, et al. Autoantibodies against small nucleolar ribonucleoprotein complexes and their clinical associations. Clin Exp Immunol. 2002;130:532–40.

    Article  PubMed  Google Scholar 

  134. Peterson LK, Jaskowski TD, Mayes MD, Tebo AE. Detection of anti-U3-RNP/fibrillarin IgG antibodies by line immunoblot assay has comparable clinical significance to immunoprecipitation testing in systemic sclerosis. Immunol Res. 2016;64:483–8.

    Article  CAS  PubMed  Google Scholar 

  135. Reimer G, Steen VD, Penning CA, Medsger TA, Tan EM. Correlates between autoantibodies to nucleolar antigens and clinical features in patients with systemic sclerosis (scleroderma). Arthritis Rheum. 1988;31:525–32.

    Article  CAS  PubMed  Google Scholar 

  136. Murata K-Y, Nakatani K, Yananeki M, Nakanishi I, Ito H. Anti-U3 ribonucleoprotein antibody-positive inflammatory myopathy: a case report. J Med Case Rep. 2016;10:169.

    Article  PubMed  PubMed Central  Google Scholar 

  137. Tall F, Dechomet M, Riviere S, Cottin V, Ballot E, Tiev KP, et al. The clinical relevance of antifibrillarin (anti-U3-RNP) autoantibodies in systemic sclerosis. Scand J Immunol. 2017;85:73–9.

    Article  CAS  PubMed  Google Scholar 

  138. Tormey VJ, Bunn CC, Denton CP, Black CM. Anti-fibrillarin antibodies in systemic sclerosis. Rheumatology (Oxford). 2001;40:1157–62.

    Article  CAS  Google Scholar 

  139. Belizna C, Henrion D, Beucher A, Lavigne C, Ghaali A, Lévesque H. Anti-Ku antibodies: clinical, genetic and diagnostic insights. Autoimmun Rev. 2010;9:691–4.

    Article  CAS  PubMed  Google Scholar 

  140. Cooley HM, Melny BJ, Gleeson R, Greco T, Kay TW. Clinical and serological associations of anti-Ku antibody. J Rheumatol. 1999;26:563–7.

    CAS  PubMed  Google Scholar 

  141. Mimori T, Hardin JA, Steitz JA. Characterization of the DNA-binding protein antigen Ku recognized by autoantibodies from patients with rheumatic disorders. J Biol Chem. 1986;261:2274–8.

    CAS  PubMed  Google Scholar 

  142. Cavazzana I, Ceribelli A, Quinzanini M, Scarsi M, Airò P, Cattaneo R, et al. Prevalence and clinical associations of anti-Ku antibodies in systemic autoimmune diseases. Lupus. 2008;17:727–32.

    Article  CAS  PubMed  Google Scholar 

  143. Rigolet A, Musset L, Dubourg O, Maisonobe T, Grenier P, Charuel J-L, et al. Inflammatory myopathies with anti-Ku antibodies: a prognosis dependent on associated lung disease. Medicine (Baltimore). 2012;91:95–102.

    Article  CAS  PubMed  Google Scholar 

  144. Franceschini F, Cavazzana I. Anti-Ro/SSA and La/SSB antibodies. Autoimmunity. 2005;38:55–63.

    Article  CAS  PubMed  Google Scholar 

  145. Pollock W, Toh BH. Routine immunofluorescence detection of Ro/SS-A autoantibody using HEp-2 cells transfected with human 60 kDa Ro/SS-A. J Clin Pathol. 1999;52:684–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Fritzler MJ, Hanson C, Miller J, Eystathioy T. Specificity of autoantibodies to SS-A/Ro on a transfected and overexpressed human 60 kDa Ro autoantigen substrate. J Clin Lab Anal. 2002;16:103–8.

    Article  CAS  PubMed  Google Scholar 

  147. Dellavance A, Alvarenga RR, Rodrigues SSH, Barbosa SH, Camilo ACP, Shiguedomi HSO, et al. Autoantibodies to 60 kDa SS-A/Ro yield a specific nuclear myriad discrete fine speckled immunofluorescence pattern. J Immunol Methods. 2013;390:35–40.

    Article  CAS  PubMed  Google Scholar 

  148. Defendenti C, Atzeni F, Spina MF, Grosso S, Cereda A, Guercilena G, et al. Clinical and laboratory aspects of Ro/SSA-52 autoantibodies. Autoimmun Rev. 2011;10:150–4.

    Article  CAS  PubMed  Google Scholar 

  149. Ghillani P, André C, Toly C, Rouquette AM, Bengoufa D, Nicaise P, et al. Clinical significance of anti-Ro52 (TRIM21) antibodies non-associated with anti-SSA 60 kDa antibodies: results of a multicentric study. Autoimmun Rev. 2011;10:509–13.

    Article  CAS  PubMed  Google Scholar 

  150. Sonesson S-E, Hedlund M, Ambrosi A, Wahren-Herlenius M. Factors influencing fetal cardiac conduction in anti-Ro/SSA-positive pregnancies. Rheumatology (Oxford). 2017;56:1755–62.

    Article  Google Scholar 

  151. Lloyd TE, Christopher-Stine L, Pinal-Fernandez I, Tiniakou E, Petri M, Baer A, et al. Cytosolic 5′-nucleotidase 1A As a target of circulating autoantibodies in autoimmune diseases. Arthritis Care Res (Hoboken). 2016;68:66–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Salajegheh M, Lam T, Greenberg SA. Autoantibodies against a 43 KDa muscle protein in inclusion body myositis. PLoS ONE. 2011;6:e20266.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Pluk H, van Hoeve BJA, van Dooren SHJ, Stammen-Vogelzangs J, van der Heijden A, Schelhaas HJ, et al. Autoantibodies to cytosolic 5′-nucleotidase 1A in inclusion body myositis. Ann Neurol. 2013;73:397–407.

    Article  CAS  PubMed  Google Scholar 

  154. Kramp SL, Karayev D, Shen G, Metzger AL, Morris RI, Karayev E, et al. Development and evaluation of a standardized ELISA for the determination of autoantibodies against cN-1A (Mup44, NT5C1A) in sporadic inclusion body myositis. Auto Immun highlights. 2016;7:16.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  155. Greenberg SA. Cytoplasmic 5′-nucleotidase autoantibodies in inclusion body myositis: isotypes and diagnostic utility. Muscle Nerve. 2014;50:488–92.

    Article  CAS  PubMed  Google Scholar 

  156. Lilleker JB, Rietveld A, Pye SR, Mariampillai K, Benveniste O, Peeters MTJ, et al. Cytosolic 5′-nucleotidase 1A autoantibody profile and clinical characteristics in inclusion body myositis. Ann Rheum Dis. 2017;76:862–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Herbert MK, Stammen-Vogelzangs J, Verbeek MM, Rietveld A, Lundberg IE, Chinoy H, et al. Disease specificity of autoantibodies to cytosolic 5′-nucleotidase 1A in sporadic inclusion body myositis versus known autoimmune diseases. Ann Rheum Dis. 2016;75:696–701.

    Article  CAS  PubMed  Google Scholar 

  158. Yeker RM, Pinal-Fernandez I, Kishi T, Pak K, Targoff IN, Miller FW, et al. Anti-NT5C1A autoantibodies are associated with more severe disease in patients with juvenile myositis. Ann Rheum Dis. 2018. https://doi.org/10.1136/annrheumdis-2017-212436.

    PubMed  Google Scholar 

  159. Herbert MK, Pruijn GJM. Novel serology testing for sporadic inclusion body myositis: disease-specificity and diagnostic utility. Curr Opin Rheumatol. 2015;27:595–600.

    Article  CAS  PubMed  Google Scholar 

  160. Kaji K, Fertig N, Medsger TA, Satoh T, Hoshino K, Hamaguchi Y, et al. Autoantibodies to RuvBL1 and RuvBL2: a novel systemic sclerosis-related antibody associated with diffuse cutaneous and skeletal muscle involvement. Arthritis Care Res. 2014;66:575–84.

    Article  CAS  Google Scholar 

  161. Pauling JD, Salazar G, Lu H, Betteridge ZE, Assassi S, Mayes MD, et al. Presence of anti-eukaryotic initiation factor-2B, anti-RuvBL1/2 and anti-synthetase antibodies in patients with anti-nuclear antibody negative systemic sclerosis. Rheumatology. 2017. https://doi.org/10.1093/rheumatology/kex458.

    Google Scholar 

  162. Takahashi T, Nakanishi T, Hamaguchi Y, Tanaka T, Fujimoto N. Case of anti-RuvBL1/2 antibody-positive morphea and polymyositis. J Dermatol. 2017:44:1188–90.

    Article  PubMed  Google Scholar 

  163. Satoh M, Langdon JJ, Chou C-H, McCauliffe DP, Treadwell EL, Ogasawara T, et al. Characterization of the Su antigen, a macromolecular complex of 100/102 and 200-kDa proteins recognized by autoantibodies in systemic rheumatic diseases. Clin Immunol Immunopathol. 1994;73:132–41.

    Article  CAS  PubMed  Google Scholar 

  164. Satoh M, Chan JYF, Ceribelli A, Vazquez del-Mercado M, EKL A. Autoantibodies to argonaute 2 (Su antigen). Adv Exp Med Biol. 2013;768:45–59.

    Article  CAS  PubMed  Google Scholar 

  165. Ogawa-Momohara M, Muro Y, Satoh M, Akiyama M. Autoantibodies to Su/Argonaute 2 in Japanese patients with inflammatory myopathy. Clin Chim Acta. 2017;471:304–7.

    Article  CAS  PubMed  Google Scholar 

  166. Bhanji RA, Eystathioy T, Chan EKL, Bloch DB, Fritzler MJ. Clinical and serological features of patients with autoantibodies to GW/P bodies. Clin Immunol. 2007;125:247–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Satoh M, Chan JYF, Ross SJ, Ceribelli A, Cavazzana I, Franceschini F, et al. Autoantibodies to survival of motor neuron complex in patients with polymyositis: immunoprecipitation of D, E, F, and G proteins without other components of small nuclear ribonucleoproteins. Arthritis Rheum. 2011;63:1972–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Cobben JM, de Visser M, Scheffer H. From gene to disease; “survival” motor neuron protein and hereditary proximal spinal muscle atrophy. Ned Tijdschr Geneeskd. 2001;145:2525–7.

    CAS  PubMed  Google Scholar 

  169. Enarson P, Rattner JB, Ou Y, Miyachi K, Horigome T, Fritzler MJ. Autoantigens of the nuclear pore complex. J Mol Med (Berl). 2004;82:423–33.

    Article  CAS  PubMed  Google Scholar 

  170. Nishio A, Water J, Leung PS, Joplin R, Neuberger JM, Lake J, et al. Comparative studies of antimitochondrial autoantibodies in sera and bile in primary biliary cirrhosis. Hepatology. 1997;25:1085–9.

    Article  CAS  PubMed  Google Scholar 

  171. Senécal J-L, Isabelle C, Fritzler MJ, Targoff IN, Goldstein R, Gagné M, et al. An autoimmune myositis-overlap syndrome associated with autoantibodies to nuclear pore complexes. Medicine (Baltimore). 2014;93:383–94.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  172. Lleo A, Marzorati S, Anaya J-M, Gershwin ME. Primary biliary cholangitis: a comprehensive overview. Hepatol Int. 2017;11:485–99.

    Article  PubMed  Google Scholar 

  173. Maeda MH, Tsuji S, Shimizu J. Inflammatory myopathies associated with anti-mitochondrial antibodies. Brain. 2012;135(Pt 6):1767–77.

    Article  PubMed  Google Scholar 

  174. Konishi H, Fukuzawa K, Mori S, Satomi-Kobayashi S, Kiuchi K, Suzuki A, et al. Anti-mitochondrial M2 antibodies enhance the risk of supraventricular arrhythmias in patients with elevated hepatobiliary enzyme levels. Intern Med. 2017;56:1771–9.

    Article  PubMed  PubMed Central  Google Scholar 

  175. Albayda J, Khan A, Casciola-Rosen L, Corse AM, Paik JJ, Christopher-Stine L. Inflammatory myopathy associated with anti-mitochondrial antibodies: a distinct phenotype with cardiac involvement. Semin Arthritis Rheum. 2018;47:552–6.

    Article  CAS  PubMed  Google Scholar 

  176. Mauhin W, Mariampillai K, Allenbach Y, Charuel J-L, Musset L, Benveniste O. Anti-mitochondrial antibodies are not a hallmark of severity in idiopathic inflammatory myopathies. Joint Bone Spine. 2017. https://doi.org/10.1016/j.jbspin.2017.04.004.

    PubMed  Google Scholar 

  177. Targoff IN, Arnett FC, Berman L, O’Brien C, Reichlin M. Anti-KJ: a new antibody associated with the syndrome of polymyositis and interstitial lung disease. J Clin Invest. 1989;84:162–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Gelpi C, Sontheimer EJ, Rodriguez-Sanchez JL. Autoantibodies against a serine tRNA-protein complex implicated in cotranslational selenocysteine insertion. Proc Natl Acad Sci USA. 1992;89:9739–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Casciola-Rosen LA, Pluta AF, Plotz PH, Cox AE, Morris S, Wigley FM, et al. The DNA mismatch repair enzyme PMS1 is a myositis-specific autoantigen. Arthritis Rheum. 2001;44:389–96.

    Article  CAS  PubMed  Google Scholar 

  180. Muro Y, Nakashima R, Hosono Y, Sugiura K, Mimori T, Akiyama M. Autoantibodies to DNA mismatch repair enzymes in polymyositis/dermatomyositis and other autoimmune diseases: a possible marker of favorable prognosis. Arthritis Rheumatol (Hoboken, NJ). 2014;66:3457–62.

    Article  CAS  Google Scholar 

  181. Labrador-Horrillo M, Martínez MA, Selva-O’Callaghan A, Trallero-Araguás E, Grau-Junyent JM, Vilardell-Tarrés M, et al. Identification of a novel myositis-associated antibody directed against cortactin. Autoimmun Rev. 2014;13:1008–12.

    Article  CAS  PubMed  Google Scholar 

  182. Berrih-Aknin S. Cortactin: a new target in autoimmune myositis and myasthenia gravis. Autoimmun Rev. 2014;13:1001–2.

    Article  CAS  PubMed  Google Scholar 

  183. Albrecht I, Wick C, Hallgren Å, Tjärnlund A, Nagaraju K, Andrade F, et al. Development of autoantibodies against muscle-specific FHL1 in severe inflammatory myopathies. J Clin Invest. 2015;125:4612–24.

    Article  PubMed  PubMed Central  Google Scholar 

  184. Amato AA, Griggs RC. Unicorns, dragons, polymyositis, and other mythological beasts. Neurology. 2003;61:288–9.

    Article  PubMed  Google Scholar 

  185. Tozzoli R, Villalta D, Bizzaro N. Challenges in the standardization of autoantibody testing: a comprehensive review. Clin Rev Allergy Immunol. 2017;53:68–77.

    Article  CAS  PubMed  Google Scholar 

Download references

Authors’ contributions

BP and GV drafted the manuscript. All authors read and approved the final manuscript.

Acknowledgements

We thank the team of Immunoallergology Laboratory, AOU-Careggi, Florence for the great deal of teamwork and constant readiness to our requests.

Competing interests

The authors declare that they have no competing interests.

Availability of data and materials

Not applicable.

Consent for publication

Not applicable.

Ethics approval and consent to participate

Not applicable.

Funding

This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Boaz Palterer.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Palterer, B., Vitiello, G., Carraresi, A. et al. Bench to bedside review of myositis autoantibodies. Clin Mol Allergy 16, 5 (2018). https://doi.org/10.1186/s12948-018-0084-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12948-018-0084-9

Keywords