Disease specificity of autoantibodies to cytosolic 5'-nucleotidase 1A in sporadic inclusion body...

11
ORIGINAL ARTICLE Autoantibodies to Cytosolic 5 0 - Nucleotidase 1A in Inclusion Body Myositis Helma Pluk, PhD, 1 * Bas J. A. van Hoeve, MD, 2 * Sander H. J. van Dooren, PhD, 1 * Judith Stammen-Vogelzangs, 1 Annemarie van der Heijden, 1 Helenius J. Schelhaas, MD, PhD, 2 Marcel M. Verbeek, PhD, 2 Umesh A. Badrising, MD, PhD, 3 Snjolaug Arnardottir, MD, PhD, 4 Karina Gheorghe, 5 Ingrid E. Lundberg, PhD, 5 Wilbert C. Boelens, PhD, 1 Baziel G. van Engelen, MD, PhD, 2 and Ger J. M. Pruijn, PhD 1 Objective: Sporadic inclusion body myositis (sIBM) is an inflammatory myopathy characterized by both degenerative and autoimmune features. In contrast to other inflammatory myopathies, myositis-specific autoantibodies had not been found in sIBM patients until recently. We used human skeletal muscle extracts as a source of antigens to detect autoantibodies in sIBM and to characterize the corresponding antigen. Methods: Autoantibodies to skeletal muscle antigens were detected by immunoblotting. The target antigen was immunoaffinity-purified from skeletal muscle extracts and characterized by mass spectrometry. A cDNA encoding this protein was cloned and expressed in vitro, and its recognition by patient sera was analyzed in an immunoprecipitation assay. Epitopes were mapped using microarrays of overlapping peptides. Results: An M r 44,000 polypeptide (Mup44) was frequently targeted by sIBM autoantibodies. The target protein was purified, and subsequent mass spectrometry analysis revealed that Mup44 is the cytosolic 5 0 -nucleotidase 1A (cN1A). By immunoprecipitation of recombinant cN1A, high concentrations of anti-Mup44 autoantibodies were detected in 33% of sIBM patient sera, whereas their prevalence in dermatomyositis, polymyositis, and other neuromuscular disorders appeared to be rare (4.2%, 4.5%, and 3.2%, respectively). Low concentrations of anti-Mup44 antibodies were found in myositis as well as other neuromuscular disorders, but not in healthy controls. Three major autoepitope regions of cN1A were mapped by using microarrays containing a set of overlapping peptides covering the complete cN1A amino acid sequence. Interpretation: Anti-Mup44 autoantibodies, which are targeted to cN1A, represent the first serological biomarker for sIBM and may facilitate the diagnosis of this type of myositis. ANN NEUROL 2013;73:397–407 S poradic inclusion body myositis (sIBM), the most commonly acquired muscle disease in people aged >45 years, is clinically characterized by insidious onset of muscle weakness, without evidence of extramuscular manifestations, finally causing serious disability. 1–3 sIBM, together with dermatomyositis (DM) and polymyositis (PM), belongs to the group of idiopathic inflammatory myopathies, which are defined by an acquired muscle weakness and the presence of inflammatory infiltrates in skeletal muscle tissue in the absence of a known cause. 4–8 Histopathologically, sIBM is not only characterized by inflammatory aspects, but also has degenerative features. View this article online at wileyonlinelibrary.com. DOI: 10.1002/ana.23822 Received Aug 16, 2012, and in revised form Nov 20, 2012. Accepted for publication Nov 21, 2012. Address correspondence to Dr Pruijn, 271 Department of Biomolecular Chemistry, Radboud University Nijmegen, PO Box 9101, NL-6500 HB Nijmegen, the Netherlands. E-mail: [email protected] Current address for Dr Pluk: Department of Biochemistry, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands. Current address for Dr van Hoeve: Department of Neurology, VieCuri Medical Center, Venlo, the Netherlands. Current address for Dr van Dooren: Institute of Pharmacology and Toxicology, Biomedical Center, University of Bonn, Bonn, Germany. From the 1 Department of Biomolecular Chemistry, Institute for Molecules and Materials and Nijmegen Center for Molecular Life Sciences, Radboud University Nijmegen, Nijmegen, the Netherlands; 2 Department of Neurology, Center for Neuroscience, Donders Institute for Brain, Cognition, and Behavior, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; 3 Department of Neurology, Leiden University Medical Center, Leiden, the Netherlands; 4 Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden; and 5 Rheumatology Unit, Department of Medicine, Karolinska University Hospital, Solna, Karolinska Institute, Stockholm, Sweden. Additional supporting information can be found in the online version of this article. V C 2013 American Neurological Association 397

Transcript of Disease specificity of autoantibodies to cytosolic 5'-nucleotidase 1A in sporadic inclusion body...

ORIGINAL ARTICLE

Autoantibodies to Cytosolic 50-Nucleotidase 1A in Inclusion Body Myositis

Helma Pluk, PhD,1* Bas J. A. van Hoeve, MD,2* Sander H. J. van Dooren, PhD,1*

Judith Stammen-Vogelzangs,1 Annemarie van der Heijden,1

Helenius J. Schelhaas, MD, PhD,2 Marcel M. Verbeek, PhD,2 Umesh A. Badrising, MD, PhD,3

Snjolaug Arnardottir, MD, PhD,4 Karina Gheorghe,5 Ingrid E. Lundberg, PhD,5

Wilbert C. Boelens, PhD,1 Baziel G. van Engelen, MD, PhD,2 and Ger J. M. Pruijn, PhD1

Objective: Sporadic inclusion body myositis (sIBM) is an inflammatory myopathy characterized by both degenerativeand autoimmune features. In contrast to other inflammatory myopathies, myositis-specific autoantibodies had notbeen found in sIBM patients until recently. We used human skeletal muscle extracts as a source of antigens to detectautoantibodies in sIBM and to characterize the corresponding antigen.Methods: Autoantibodies to skeletal muscle antigens were detected by immunoblotting. The target antigen wasimmunoaffinity-purified from skeletal muscle extracts and characterized by mass spectrometry. A cDNA encodingthis protein was cloned and expressed in vitro, and its recognition by patient sera was analyzed in animmunoprecipitation assay. Epitopes were mapped using microarrays of overlapping peptides.Results: An Mr 44,000 polypeptide (Mup44) was frequently targeted by sIBM autoantibodies. The target protein waspurified, and subsequent mass spectrometry analysis revealed that Mup44 is the cytosolic 50-nucleotidase 1A (cN1A).By immunoprecipitation of recombinant cN1A, high concentrations of anti-Mup44 autoantibodies were detected in33% of sIBM patient sera, whereas their prevalence in dermatomyositis, polymyositis, and other neuromusculardisorders appeared to be rare (4.2%, 4.5%, and 3.2%, respectively). Low concentrations of anti-Mup44 antibodieswere found in myositis as well as other neuromuscular disorders, but not in healthy controls. Three majorautoepitope regions of cN1A were mapped by using microarrays containing a set of overlapping peptides coveringthe complete cN1A amino acid sequence.Interpretation: Anti-Mup44 autoantibodies, which are targeted to cN1A, represent the first serological biomarker forsIBM and may facilitate the diagnosis of this type of myositis.

ANN NEUROL 2013;73:397–407

Sporadic inclusion body myositis (sIBM), the most

commonly acquired muscle disease in people aged

>45 years, is clinically characterized by insidious onset

of muscle weakness, without evidence of extramuscular

manifestations, finally causing serious disability.1–3 sIBM,

together with dermatomyositis (DM) and polymyositis

(PM), belongs to the group of idiopathic inflammatory

myopathies, which are defined by an acquired muscle

weakness and the presence of inflammatory infiltrates in

skeletal muscle tissue in the absence of a known cause.4–8

Histopathologically, sIBM is not only characterized by

inflammatory aspects, but also has degenerative features.

View this article online at wileyonlinelibrary.com. DOI: 10.1002/ana.23822

Received Aug 16, 2012, and in revised form Nov 20, 2012. Accepted for publication Nov 21, 2012.

Address correspondence to Dr Pruijn, 271 Department of Biomolecular Chemistry, Radboud University Nijmegen, PO Box 9101, NL-6500 HB Nijmegen,

the Netherlands. E-mail: [email protected]

Current address for Dr Pluk: Department of Biochemistry, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands. Current address

for Dr van Hoeve: Department of Neurology, VieCuri Medical Center, Venlo, the Netherlands. Current address for Dr van Dooren: Institute of

Pharmacology and Toxicology, Biomedical Center, University of Bonn, Bonn, Germany.

From the 1Department of Biomolecular Chemistry, Institute for Molecules and Materials and Nijmegen Center for Molecular Life Sciences, Radboud

University Nijmegen, Nijmegen, the Netherlands; 2Department of Neurology, Center for Neuroscience, Donders Institute for Brain, Cognition, and

Behavior, Radboud University Nijmegen Medical Center, Nijmegen, the Netherlands; 3Department of Neurology, Leiden University Medical Center, Leiden,

the Netherlands; 4Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden; and 5Rheumatology Unit, Department of Medicine,

Karolinska University Hospital, Solna, Karolinska Institute, Stockholm, Sweden.

Additional supporting information can be found in the online version of this article.

VC 2013 American Neurological Association 397

The presence of sarcoplasmic and myonuclear protein

aggregates resembles the histopathological features of neu-

rodegenerative diseases. Examples are aggregates containing

the TAR DNA-binding protein 43 (TDP-43) as in fronto-

temporal dementia and b-amyloid and tau as seen in the

brain of patients with Alzheimer disease. The exact patho-

genic mechanisms are unknown, and it has been debated

whether sIBM is a primarily degenerative or a primarily

immune-mediated disease.1,9 Recent studies on sIBM have

focused on degenerative aspects with multiple proteins

that accumulate in sIBM muscle tissue.10–13

The presence of high-titer autoantibodies in the

sera of 60 to 80% of PM and DM patients supports the

view that autoimmunity plays an important pathogenic

role in these disorders.7,8,14 Some of the autoantibodies,

such as anti–Jo-1, anti–Mi-2, and anti-SRP antibodies,

are highly specific for myositis and are not found or are

only sporadically found in other rheumatic diseases. The

production of autoantibodies may precede the onset of

clinical symptoms, and is often correlated with specific

clinical phenotypes (reviewed in Lundberg and Grundt-

man7). These autoantibodies target ubiquitous antigens,

and their role in the pathogenesis of inflammatory

myopathies is still unclear.

The general view for sIBM is that the degenerative

component plays a more prominent role in the pathogene-

sis than the immune component.1,11 Endoplasmic reticu-

lum stress, proteasome inhibition, mitochondrial abnor-

malities, and oxidative stress are factors that individually

or in concordance contribute to the sarcoplasmic and

myonuclear accumulation of several aggregate-prone pro-

teins (eg, b-amyloid, aB-crystallin, TDP-43), including

some that are typically associated with neurodegenerative

diseases such as Alzheimer disease.1,9–12,15–17 The specific-

ity and sensitivity of cytoplasmic aggregates is a matter of

debate, although some studies suggest that TDP-43 accu-

mulation may have clinical utility in distinguishing sIBM

from other inflammatory myopathies.10,13,16

Although sIBM is far less responsive to immunomo-

dulatory therapies as compared to DM and PM, the pres-

ence of cytotoxic T cells surrounding or infiltrating muscle

fibers and the clonal expansion of lymphocytes support the

hypothesis that the immune system plays an important

role in the pathogenesis of sIBM as well. Clonal expansion

of T and B cells has been extensively docu-

mented.1,2,9,14,18,19 In addition, plasma cells have been

found within the affected muscles,20 and immunoglobulin

mRNAs have been reported to be among the most abun-

dant transcripts in sIBM muscle.21 In addition, the associa-

tion with HLA-DR3 further supports a role of the

immune system in the pathogenesis of sIBM,22,23 further

supported by the presence of autoantibodies such as anti-

Ro52 and anti-La being detected in a substantial number

of patients.7,24–28 Recently, a highly specific autoantibody

targeting a 43kDa protein was found in 13 of 25 sIBM

sera, but the target of this autoantibody was not defined.29

Here we report the identification of sIBM-associ-

ated autoantibodies targeting a muscle-specific Mr

44,000 polypeptide, Mup44, and show that anti-Mup44

antibodies are frequently found in sIBM patients,

whereas they occur only at low frequencies in other dis-

ease groups, including DM and PM. Furthermore, the

characterization of the molecular identity of Mup44 will

be described, as well as the mapping of the major epi-

topes recognized by sIBM sera.

Subjects and Methods

Serum SamplesSerum samples from a group of well-characterized myositis

patients described by Abdo et al30 (30 sIBM, 24 DM, and 23

PM patients) were used in this study. All sIBM patients (cohort

sIBM-N) fulfilled the European Neuromuscular Center crite-

ria31 for sIBM. PM and DM were diagnosed according to the

diagnostic criteria of Tanimoto et al.32 Sera from 2 other

cohorts of sIBM patients, 1 from Leiden (sIBM-L)33 and 1

from Stockholm (sIBM-S),34 as well as sera from patients suf-

fering from muscular dystrophies (myotonic muscular dystro-

phy, oculopharyngeal muscular dystrophy, limb-girdle muscular

dystrophy, facioscapulohumeral muscular dystrophy, Miyoshi

muscular dystrophy), metabolic myopathies (mitochondrial my-

opathy, McArdle disease, lipid myopathy), motor neuron dis-

ease (amyotrophic lateral sclerosis, progressive spinal muscular

atrophy, postpolio syndrome), poly(radiculo)neuropathy (hered-

itary motor and sensory polyneuropathy type I or II, chronic

inflammatory demyelinating polyradiculoneuropathy), congeni-

tal myopathy (nemaline rod myopathy, multicore disease), and

miscellaneous neuromuscular disorders (myasthenia gravis, toxic

myopathy, ocular myositis, sarcoid myopathy, hypokalemic peri-

odic paralysis)35 were analyzed in parallel. As controls, sera

from healthy individuals obtained from the Sanquin Blood

Supply Foundation (Nijmegen, the Netherlands) were analyzed.

Local medical ethical committee approval and written informed

consent from all patients were obtained.

Based upon the observation of the anti-Mup44 antibody

detected in 2 patients diagnosed with PM, these patients were

reassessed clinically as well as histologically (using a new muscle

biopsy from 1 of these patients) as described.24

Muscle Extract and Cell LysatesHuman healthy hamstring muscle obtained from reconstructive

surgery (a kind gift of Dr Snoeck, Canisius-Wilhelmina Hospital,

Nijmegen, the Netherlands) and mouse calf complex muscle (a

kind gift of Dr Wieringa, Department of Cell Biology, Radboud

University Nijmegen Medical Center, Nijmegen, the Nether-

lands) were frozen in liquid nitrogen and stored at �80�C.

ANNALS of Neurology

398 Volume 73, No. 3

Small pieces of frozen muscle tissue were pulverized with

a Microdismembrator (Braun Biotech International, Melsungen,

Germany), resuspended in 10 volumes of radioimmunoprecipi-

tation assay (RIPA) buffer (50mM Tris-HCl, pH 7.4, 150mM

NaCl, 1% Nonidet P-40 [NP-40], 0.5% sodium deoxycholate,

0.1% sodium dodecyl sulfate [SDS], 10mM dithiothreitol,

0.5mM phenylmethylsulfonyl fluoride [PMSF], and complete

protease inhibitor cocktail [Roche, Mannheim, Germany]),

sonicated at 4�C, and centrifuged for 15 minutes at 21,000 �g. The supernatants were used for SDS–polyacrylamide gel elec-

trophoresis (PAGE) followed by Western blotting or for immu-

noaffinity purification.

Human Jurkat T-lymphocyte and mouse C2C12 myo-

blast cells were cultured according to standard procedures. The

myoblasts were differentiated into myotubes by incubation with

2% horse serum for 5 to 7 days. Cells were lysed by sonication

in lysis buffer (50mM Hepes-KOH, pH 7.4, 100mM KCl,

10mM MgCl2, 0.05% NP-40, 0.5mM PMSF, and complete

protease inhibitor cocktail) followed by centrifugation at 21,000

� g. The supernatants were used for SDS-PAGE followed by

Western blotting.

ImmunoblottingHuman or mouse muscle extracts, and Jurkat or C2C12 myo-

tube cell lysates were separated by SDS-PAGE using the full

width of the gels, and blotted to nitrocellulose membranes.

Membranes were cut into strips 3 to 4mm wide, blocked with

5% nonfat dry milk in phosphate-buffered saline– 0.1%

Tween-20 (PBS-T) and incubated with human patient sera,

1,000-fold diluted in 5% milk–PBS-T. Bound antibodies were

visualized by incubation with goat–antihuman IRDye800-

labeled secondary antibody (Rockland, Gilbertsville, PA) and

scanning with the Odyssey system (LI-COR Biosciences,

Lincoln, NE).

Immunoaffinity Purification of Mup44Immunoglobulin G (IgG) from 250ll serum (from 2 sIBM

patients and from a pool of healthy individuals) was isolated by

incubation with 250ll protein A-agarose beads (Kem-en-Tec,

Taastrup, Denmark) and subsequently covalently coupled to the

beads using dimethyl pimelimidate dihydrochloride (Sigma

Aldrich, St Louis, MO) in 0.2M sodium borate buffer, pH 9.0.

Antibody-coupled beads were washed with ethanolamine and

PBS and stored at 4�C until further use.

Human muscle lysate (10mg total protein) was incubated

with 250ll protein A-agarose beads in RIPA buffer to remove

endogenous IgG from the muscle lysate. Subsequently, 50ll

antibody-coupled beads were added to the precleared muscle

lysate. After an incubation for 16 hours at 4�C, the beads were

washed 5� with RIPA buffer, and bound proteins were released

by heating (100�C) in SDS sample buffer (2% SDS, 5% b-

mercaptoethanol, 10% glycerol, 0.01% bromophenol bleu,

125mM Tris-HCl, pH 6.8) for 5 minutes. The released pro-

teins were separated by SDS-PAGE and visualized by colloidal

Coomassie brilliant blue staining. The band migrating at the

position of Mup44 was excised from the gel, in-gel digested

with trypsin, and analyzed by mass spectrometry.

Mass SpectrometryThe samples, which were digested with trypsin after reduction

and alkylation, were loaded on stage tips and eluted in a final

volume of 20ll, of which 5ll was used for the analysis by liq-

uid chromatography–mass spectrometry (LC-MS/MS). In brief,

the samples were resolved by reverse phase nano–high-perform-

ance liquid chromatography prior to nanoelectrospray ioniza-

tion. The peptide ions were analyzed by the LTQ FT Ultra MS

mass spectrometer (Thermo Scientific, Waltham, MA). Peptide

and protein identities were extracted from the data by means of

the Mascot program using the RefSeq33 database (Homo sapiens

taxonomy) with added sequence tags. Likely contaminants were

added to this database (eg, human keratins, trypsin). The fol-

lowing modifications were allowed in the search: carbamidome-

thylation of cysteines (fixed), oxidation of methionine (vari-

able), and acetylation of the N-terminus (variable). Protein

identification validation was performed by a script developed

in-house. Briefly, the software classifies protein identifications

based on the number of uniquely identified peptide sequences,

clusters proteins sharing the same set of peptides, and validates

the proteins with the following criteria: proteins with 1 peptide

must have a peptide score >49; proteins with >1 peptide must

have a peptide score >29.

cDNA Cloning and Expression of Mup44RNA was isolated from a human muscle (hamstring) lysate and

converted into cDNA by reverse transcriptase using an oli-

go(dT) primer. Cytosolic 50-nucleotidase 1A (cN1A) cDNAs

were generated by polymerase chain reaction (PCR) using total

muscle cDNA and the cN1A–specific primers 50-CACCATG-

GAACCTGGGCAGCC-30 and 50-CTGAATTCCTACTGTG-

CAGATGGGGCC-30. The resulting amplicon of 1,119 base

pairs was subsequently cloned into the pENTR/TEV/D-TOPO

vector according to the manufacturer’s guidelines (Invitrogen,

Carlsbad, CA). The sequence of the selected clones was con-

firmed by DNA sequencing. In all cases, nucleotides 729 and

823 of the coding sequence appeared to be different from the

corresponding nucleotides in cN1A REFSEQ mRNA

(NM_032526.1): 729G!A; 823A!G. Because the complete

cloning procedure was performed twice, these changes are most

likely genetic polymorphisms. The alteration of nucleotide 729

does not lead to an alteration of the amino acid sequence,

whereas the A-to-G substitution at position 823 leads to an

altered codon: Ile!Val.

The cN1A cDNA was transferred to the pDEST17 vector

by recombination cloning using LR Clonase. Radiolabeled (35S)

recombinant cN1A was expressed using a TNT Quick coupled

in vitro transcription/translation system (Promega, Madison,

WI).

Immunoprecipitation of Mup44Antibodies from patient sera (10ll per serum) were coupled to

protein A-agarose beads (Kem-en-Tec) in IPP500 (500mM

Pluk et al: Anti–cN1A Autoantibodies

March 2013 399

NaCl, 10mM Tris-HCl, pH 8.0, 0.05% NP-40) overnight at

4�C. Beads were washed 3� with IPP500 and once with

IPP150 (150mM NaCl, 10mM Tris-HCl, pH 8.0, 0.05% NP-

40). In vitro translated 35S-labeled recombinant cN1A was incu-

bated with the antibody-coupled beads for 2 hours at 4�C. After

washing the beads 4� with IPP150, bound proteins were eluted

with SDS sample buffer and subjected to SDS-PAGE. Immuno-

precipitated cN1A was quantified by phosphorimaging.

Microarray-Based Epitope MappingMicroarrays containing 90 synthetic peptides (15 amino acids

each) derived from the amino acid sequence of cN1A (11

amino acids overlap between consecutive peptides) were pur-

chased from JPT Peptide Technologies (Berlin, Germany). Each

slide contained 3 identical arrays of peptides. The slides were

blocked by incubation with MTBST (5% non-fat dried milk in

Tris-buffered saline, 0.05% Tween-20) for 1 hour at room tem-

perature. Subsequently, the slides were incubated with 300ll

100-fold diluted patient sera in MTBST for 2 hours at 37�C

in a humid chamber, and after washing 5� with MTBST, the

arrays were incubated with Alexa Fluor-568–labeled goat–anti-

human secondary antibodies (A21090; Molecular Probes,

Eugene, OR) for 1 hour at 30�C under agitation. After washing

5� with MTBST and 5� with water, the slides were dried, and

bound antibodies were visualized by a ProScanArray microarray

scanner (PerkinElmer, Waltham, MA). Signals were quantified

using Quantity One software (Bio-Rad, Hercules, CA).

Results

Identification of Anti-Mup44 AutoantibodiesUsing Human Skeletal Muscle ExtractTo investigate the presence of autoantibodies in sIBM

patient sera targeted against skeletal muscle components,

healthy human hamstring extracts were analyzed by im-

munoblotting. Several polypeptides were recognized by

the antibodies in patient sera. Although different sera

were reactive with distinct muscle antigens, 1 prominent,

Mr 44,000 polypeptide appeared to be commonly

detected by several sIBM sera (Fig 1A). Unlike other anti-

gens that were reactive with antibodies from sIBM patient

sera, such as for example an Mr 60,000 polypeptide recog-

nized by Patient C (see Fig 1A, lane 3), the Mr 44,000

polypeptide appeared to be skeletal muscle specific. Using

the same set of sIBM patient sera, this polypeptide was not

detectable in extracts from cultured cell lines such as

human Jurkat and HeLa cells, nor in differentiated mouse

C2C12 myotubes (see Fig 1B, C and data not shown).

Despite the lack of detection of the antigen in the C2C12

myotubes, the reactive sera did stain a band of approxi-

mately the same molecular weight when mouse skeletal

muscle extracts were used (data not shown).

Immunoblotting using adjacent blot strips con-

firmed that the Mr 44,000 antigen detected by different

sIBM patient sera showed exactly the same electropho-

retic mobility in SDS-PAGE gels (data not shown),

strongly suggesting that a common antigenic protein,

here designated Mup44 (Muscle protein with a molecular

weight of 44,000), is recognized by these sera. The initial

immunoblot screening with 31 sIBM sera revealed 9

(29%) anti-Mup44–positive sera.

Molecular Identity of Mup44To reveal the molecular identity of Mup44, the protein

was isolated from human skeletal muscle extracts by

immunoaffinity chromatography using antibodies isolated

FIGURE 1: Reactivity of sporadic inclusion body myositis (sIBM) sera with skeletal muscle polypeptides. (A) Human skeletalmuscle lysate was separated by sodium dodecyl sulfate–polyacrylamide gel electrophoresis and blotted to nitrocellulose mem-branes. Blot strips were incubated with sIBM patient sera (A–I) and a normal healthy serum (NHS), followed by detection ofthe bound antibodies by incubation with an IRDye800-labeled secondary antibody. The arrow marks the position of theMup44 antigen. (B, C) Human Jurkat cell extract (B) and mouse C2C12 myotube cell extract (C) were analyzed as described inA. Asterisks and arrowheads in A–C indicate putatively identical antigens. The positions of molecular weight markers (31,000)are indicated on the right in each panel. Data are representative of at least 2 independent experiments.

ANNALS of Neurology

400 Volume 73, No. 3

from 2 anti-Mup44–positive patient sera. As a control,

serum antibodies from a healthy individual were used in

parallel. Separation of the isolated proteins by SDS-PAGE

and staining with Coomassie brilliant blue revealed a poly-

peptide of the expected molecular weight only in the

material obtained with antibodies from the anti-Mup44–

positive patients (Fig 2A). A polypeptide migrating slightly

faster than the putative Mup44 was observed in all 3

samples. The Mr 44,000 band was excised from the gel,

subjected to in-gel digestion with trypsin, and analyzed by

LC-MS/MS. Database searching with the LC-MS/MS

results strongly suggested that Mup44 corresponds to

cN1A (NP_115915.1). The Exponentially Modified Pro-

tein Abundance Index (emPAI) scores of cN1A, which is

the highest scoring protein in the LC-MS/MS results list-

ing, were 5.4, 9.0, and 0.9 for the material excised from

lanes 1, 2, and 3 of Figure 2A, respectively (Supplemen-

tary Fig). The peptide amino acid sequences obtained by

the mass spectrometry data covered 65% of cN1A (see Fig

2B). Both the calculated molecular mass of cN1A

(41,021kDa) and cN1A being expressed at relatively high

levels in (skeletal) muscle are consistent with the observa-

tions made for Mup44. The second highest scoring pro-

tein, alpha-actin (NP_001091.1; NP_00150.1), with rela-

tively high emPAI scores in all 3 samples (6.7, 5.8, and

3.0, respectively), most probably results from contamination

of the excised Mup44 band with alpha-actin, which is

among the most abundant proteins in skeletal muscle extracts

and probably corresponds to the band migrating slightly

faster than Mup44 in the immunoaffinity-purified material.

To confirm that Mup44 is indeed identical to

cN1A, a cDNA encoding cN1A was generated by RT-

PCR using cN1A-specific primers and RNA isolated from

human muscle extracts as starting material. Subsequently35S-labeled cN1A was expressed by in vitro transcription-

translation in a reticulocyte lysate. Immunoprecipitation of

the recombinant protein with anti-Mup44–positive patient

sera showed that the antibodies in these sera are directed to

cN1A (Fig 3). The efficiency by which cN1A was precipi-

tated appeared to correlate with the intensity of the bands

observed by immunoblotting with skeletal muscle extracts.

The weak signal of immunoprecipitated cN1A obtained

with a sIBM serum that did not contain detectable levels

of anti-Mup44 antibodies by immunoblotting and the lack

of cN1A immunoprecipitation by sera from healthy con-

trols suggested that the immunoprecipitation approach is

more sensitive than immunoblotting for the detection of

anti-Mup44–positive sera; 57% of the sIBM sera used in

immunoblotting appeared to precipitate >1% of recombi-

nant cN1A (see below).

Association of Anti-Mup44 Autoantibodieswith sIBMTo investigate the disease-specificity of anti-Mup44 auto-

antibodies, sera from 3 cohorts of sIBM patients, as well

as sera from DM and PM patients, from patients suffer-

ing from other neuromuscular diseases (NMDs; nonmyo-

sitis NMD; see Subjects and Methods for specifications),

and from healthy individuals were analyzed. To detect

the presence of anti-Mup44 autoantibodies, immunopre-

cipitation assays were performed with the radiolabeled

recombinant cN1A protein. Precipitated cN1A was ana-

lyzed by SDS-PAGE and quantified by phosphorimaging.

Sera from 2 cohorts of sIBM patients were collected in

FIGURE 2: Molecular identity of Mup44. (A) Mup44 was immunoaffinity purified from human muscle extracts using antibodiesisolated from 2 sporadic inclusion body myositis patients (IBM1 and IBM2). Antibodies from healthy individuals (normal healthyserum [NHS]) were used in parallel. As an additional control (C), the immobilized IBM1 antibodies were used without muscleextract. Precipitated proteins were separated by sodium dodecyl sulfate–polyacrylamide gel electrophoresis and visualized bycolloidal Coomassie brilliant blue staining. The arrow marks the position of the Mup44 polypeptide. The positions of molecularweight markers are indicated on the left. (B) Sequence coverage of cytosolic 50-nucleotidase 1A based upon mass spectrome-try data. Peptides found in the material isolated with antibodies from patients IBM1 and IBM2 are marked by lines below andabove the amino acid sequence, respectively.

Pluk et al: Anti–cN1A Autoantibodies

March 2013 401

the Netherlands (Nijmegen30 and Leiden,33 respectively),

and the third cohort was from Sweden (Stockholm34).

The results showed great variations in the efficiencies by

which cN1A was precipitated by distinct sera. High reac-

tivities were almost exclusively found in sIBM sera,

whereas low reactivities were also observed with DM,

PM, and nonmyositis NMD sera (Fig 4). To differentiate

between high and low cN1A immunoprecipitation effi-

ciencies, cutoff levels of 5% and 1% (fraction of input

cN1A precipitated) were arbitrarily chosen. High anti–

cN1A reactivities were frequently observed in all 3 sIBM

cohorts (40%, 25%, and 34%, respectively; Table).

Although some of the DM, PM, and nonmyositis NMD

samples precipitated >5% of the input cN1A protein,

their reactivities tended to be lower than those of the

sIBM sera. When the weak reactivities was also taken

into account, the prevalence of anti-Mup44 in sIBM sera

was still much higher than that in the other diseases. Al-

together, anti-Mup44 was detected in 60% of the sIBM

sera, in 11% of the other diseases, and in none of the

healthy controls (see Table).

Major Epitope Regions of cN1A Targeted bysIBM SeraTo get more insight into the regions of cN1A that are

preferentially targeted by sIBM sera, microarrays contain-

ing a set of overlapping synthetic peptides covering the

complete amino acid sequence of cN1A were generated

and probed with anti-Mup44–positive sera. In total,

ninety 15-mer peptides with 11 amino acid overlap were

synthesized and spotted in triplicate on the surface of

glass slides. Nine slides were incubated with anti-Mup44

autoantibody–containing sIBM sera and 2 with normal

healthy control sera. After a subsequent incubation with

a fluorescently labeled secondary antibody, bound anti-

bodies were visualized by a high-resolution fluorescence

scanner. Three major epitope regions were identified (Fig

5). One of these, located close to the N-terminus, com-

prises amino acid residues 25 to 50 and was recognized

by 6 of the 9 sera, although within this region some het-

erogeneity among the reactive sera was observed with

regard to the peptides that were most strongly reactive. A

second autoreactive region is located close to the C-ter-

minus (aa341–368) and was recognized by 5 of the 9

anti-Mup44–positive sera, albeit to different extents, and

here also some heterogeneity was observed. The third

epitope region is more centrally located in the primary

sequence of cN1A (aa221–243), and peptides in this

region were recognized by all anti-Mup44–positive sera

analyzed. Except for a single peptide that showed a weak

reactivity with 1 of the control sera, the control sera were

not reactive with any of the cN1A peptides on the arrays.

These data strongly suggest that cN1A contains at least 3

nondiscontinuous autoepitopes.

Discussion

This is the first study to reveal the presence of the anti-

Mup44 autoantibody in sIBM sera and the identity of its

FIGURE 3: Immunoprecipitation of cytosolic 50-nucleotidase 1A (cN1A) by patient sera. In vitro translated, 35S-labeled cN1A(lane 1) was incubated with antibodies from sporadic inclusion body myositis (sIBM) and healthy control (normal healthy serum[NHS]) sera coupled to protein A-agarose beads. The immunoprecipitated proteins were separated by sodium dodecyl sulfate–polyacrylamide gel electrophoresis and visualized by phosphorimaging. As a control, an immunoprecipitation with beads lack-ing antibodies was performed in parallel (lane 10). Pluses and minuses refer to the level of anti-Mup44 reactivity in immuno-blotting with skeletal muscle lysates. Data are representative of at least 2 independent experiments.

ANNALS of Neurology

402 Volume 73, No. 3

target, cN1A. Because previous studies with routinely used

sources of autoantigens, such as cultured cell lysates, failed

to detect myositis-specific autoantibodies associated with

sIBM, we hypothesized that such antibodies might be

directed to antigens specifically occurring in the affected

tissues (ie, skeletal muscle). This led to the identification of

an autoantibody frequently occurring in sIBM. Using

human skeletal muscle proteins as antigens in immunoblot-

ting, we were able to identify an Mr 44,000 muscle auto-

antigen, designated Mup44, which in immunoblotting

experiments was recognized by 29% of the sIBM sera,

whereas it was hardly reactive or not reactive with other

myositis and healthy control sera (data not shown). A

more sensitive and more antigen-specific assay developed

after determining the molecular identity of Mup44 (cN1A)

allowed the detection of anti-Mup44 autoantibodies in up

to 60% of sIBM patients.

It has been common practice to use cultured cell

lines such as HeLa cells for the identification of autoanti-

gens in autoimmune diseases. The reason anti-Mup44

autoantibodies have escaped detection until recently may

at least in part be the apparent absence of Mup44 expres-

sion in such cell lines. Interestingly, Dalakas and

coworkers noted in 1997 the recognition of various

human muscle antigens ranging in size from 35 to

145kDa by sIBM patient sera, an observation that, how-

ever, was not explored in more detail.36 While our stud-

ies on Mup44 were in progress, Salajegheh and co-

workers reported the presence of autoantibodies to an Mr

43,000 muscle antigen in sIBM.29 Also in this study, the

use of normal human muscle lysates in immunoblotting

allowed the detection of these autoantibodies, but these

investigators did not demonstrate that the antigen was

not detectable in lysates from other cells or tissues. The

FIGURE 4: Anti-Mup44 autoantibodies in sera from various patient groups. In vitro translated, 35S-labeled cytosolic 50-nucleo-tidase 1A (cN1A) was incubated with antibodies from 3 sporadic inclusion body myositis cohorts (sIBM-N 5 patients seen atthe Radboud University Nijmegen Medical Center; sIBM-L 5 patients seen at the Leiden University Medical Center; sIBM-S 5

patients seen at the Karolinska University Hospital in Stockholm), dermatomyositis (DM) and polymyositis (PM) patients,patients with other neuromuscular diseases (Neuromusc.), and healthy controls (normal healthy serum [NHS]), coupled to pro-tein A-agarose beads. The immunoprecipitated proteins were separated by sodium dodecyl sulfate–polyacrylamide gel electro-phoresis and visualized by phosphorimaging. For each of the samples, the percentage of immunoprecipitated (IP) cN1A isdepicted in the graph.

Pluk et al: Anti–cN1A Autoantibodies

March 2013 403

reported prevalence of the anti-43k autoantibodies is

higher than what we observed for anti-Mup44 in immu-

noblotting (52 vs 29%). Nevertheless, it is likely that

these autoantibodies are targeting the same antigen. The

difference in frequency might be due to the relatively

low number of samples analyzed (Salajegheh and co-workers analyzed only 25 sIBM patients), differencesbetween distinct cohorts, and/or different sensitivities ofthe assays. The slight difference in molecular weights, Mr

44,000 versus Mr 43,000, might well be due to the inac-curacy of molecular weight determination by SDS-

PAGE. Also the specificity of the anti-43k antibodies wasreported to be high, because these antibodies were not

detected in samples from 10 DM, 10 PM, 5 myastheniagravis patients, and 15 healthy volunteers.29

The frequency of anti-Mup44 reactivity in sIBM

sera is remarkably high when compared with other auto-

antibodies in myositis. The most frequently detected my-

ositis-specific autoantibody anti–Jo-1, for example, is tar-

geted by only 20 to 30% of PM and DM

patients.7,8,25,27 The analysis of sera from other neuro-

muscular disorders, including DM and PM, by a

FIGURE 5: Major cytosolic 50-nucleotidase 1A (cN1A) epitopes recognized by anti-Mup44 autoantibodies. A panel of 90 over-lapping peptides from cN1A (15aa; 11aa overlap) immobilized on glass slides was incubated with 9 anti-Mup44–positive spo-radic inclusion body myositis (sIBM) sera and 2 sera from healthy controls (normal healthy serum [NHS]; first 2 rows). Antibodybinding to the peptides on the arrays was visualized by a fluorescently labeled secondary antibody and quantified by a fluores-cence scanner. The reactivity with each of the peptides (1–90 from N- to C-terminus of cN1A) is depicted in arbitrary units(AU). Data were obtained by analyzing microarrays in triplicate. Average values are shown in the graph.

TABLE : Prevalence of Anti-Mup44 Autoantibodies in Myositis and Other Neuromuscular Disorders

Sera No. High Anti-Mup44a Total Anti-Mup44b

No. % No. %

sIBM

sIBM-N 30 12 40 17 57

sIBM-L 32 8 25 20 63

sIBM-S 32 11 34 19 59

Total sIBM 94 31 33 56 60

DM 24 1 4 5 21

PM 22 1 5 3 14

Neuromuscular disorders 94 3 3 7 7

NHSc 32 0 0 0 0aEfficiency of immunoprecipitation >5% of input cN1A.bEfficiency of immunoprecipitation >1% of input cN1A.cSera from healthy individuals.cN1A ¼ cytosolic 50-nucleotidase 1A; DM ¼ dermatomyositis; NHS ¼ normal healthy serum; PM ¼ polymyositis; sIBM ¼ spo-radic inclusion body myositis; sIBM-L ¼ patients from the Leiden University Medical Center; sIBM-N ¼ patients from the Rad-boud University Nijmegen Medical Center; sIBM-S ¼ patients from the Karolinska University Hospital, Stockholm.

ANNALS of Neurology

404 Volume 73, No. 3

recombinant cN1A immunoprecipitation assay, demon-

strated that anti-Mup44 autoantibodies can be detected

in these sera as well, although both their concentration

and their prevalence are much lower than that in sIBM.

Relatively high concentrations of anti-Mup44 autoanti-

bodies were found in 2 PM patients. It is well known

that sIBM patients can be easily misdiagnosed as having

PM, and therefore we re-evaluated these patients clini-

cally and histologically. This showed that 1 of these

patients was misdiagnosed with PM, although this

patient did not show any indication for sIBM, neither

clinically nor histologically. A renewed muscle biopsy

lacked sIBM-specific histological abnormalities, such as

rimmed vacuoles or TDP-43 positive inclusions (data

not shown). Therefore, the results for this patient have

been omitted from the data. Recently, we observed that

anti-Mup44 autoantibodies may also occur in the sera of

systemic lupus erythematosus and Sj€ogren syndrome

patients, although in these cases also, the prevalence is

much lower than that in sIBM (unpublished data). In

this respect, it is important to note that no correlation

was observed between sIBM patients with symptoms of

Sj€ogren or sicca syndrome and the presence of anti-

Mup44 autoantibodies. Moreover, a comparison of the

presence of anti-Mup44, anti-Ro, and anti-La autoanti-

bodies in the sera of the sIBM patients revealed that

there is no correlation. For 92 sIBM patients, anti-Ro

and anti-La data were available. In 22 of these anti-Ro

and/or anti-La was detected; 16% of the anti-Mup44–

positive and 27% of the anti-Mup44–negative sera

(cN1A immunoprecipitation data; cutoff level 5% of

input) contained anti-Ro and/or anti-La antibodies.

These observations are in agreement with those of Salaje-

gheh and coworkers, who did not observe a relationship

between the anti-43k autoantibodies and antinuclear

antibodies (ANA) in 16 sIBM patients tested for ANA.29

We conclude that anti-Mup44 autoantibodies display a

high degree of specificity for sIBM. It will be interesting

to investigate whether the cN1A epitope(s) recognized by

other neuromuscular and autoimmune diseases are differ-

ent from those targeted by sIBM sera. The possible iden-

tification of disease-specific epitopes may facilitate the

development of anti-Mup44 tests with even higher

specificities.

Recent studies have focused on the degenerativeaspects of sIBM and have discussed the use of accumu-lated and aggregated proteins as biomarkers.10,16,37 Thespecificity and sensitivity of these proteins is debated,and no single protein appears to aggregate specifically inall cases of sIBM. Small alterations in protein homeosta-sis may result in the accumulation of assorted pro-teins.10–13,16 Therefore, anti-Mup44, representing an

antibody-based sIBM biomarker that may distinguishsIBM from other inflammatory myopathies, is of addi-tional diagnostic value. This is particularly relevant inview of the abovementioned difficulty of differentiatingPM and sIBM based upon the histological analyses ofmuscle biopsies.1,5 Moreover, anti-Mup44 detection is aless invasive and less laborious procedure compared tomuscle biopsy-based investigations. The presence of anti-Mup44 could be of help in providing a diagnosis beforeovert clinical or histopathological features become appa-rent. Further research will focus on the possible presenceof distinctive clinical features, prognosis, and possibletreatment responses of anti-Mup44–positive sIBMpatients.

We identified the target of anti-Mup44 as the cyto-

solic 50-nucleotidase IA, a muscle-specific enzyme belong-

ing to the family of 50- nucleotidases. cN1A catalyzes the

hydrolysis of adenosine monophosphate to adenosine and

inorganic phosphate and, with other members of this

enzyme family, is involved in the physiological control of

energy balance, metabolic regulation, and cell replication.

The human 50 nucleotidases are of clinical interest due

to their ability to inhibit the activation of nucleoside ana-

log drugs that are used in anticancer and antiviral drug

therapy. Interestingly, almost 3 decades ago an increase in

interstitial staining of skeletal muscle of inflammatory

myopathy patients was observed with a lead salt-based

staining method for 50-nucleotidases.38 It remains to be

investigated whether the expression of cN1A is altered in

sIBM and, if so, whether this correlates with the produc-

tion of anti-Mup44 autoantibodies.

The 3 major autoepitope regions of cN1A identi-

fied by the analysis of a set of overlapping peptides from

cN1A suggest that relatively small fragments of this pro-

tein can be used to detect the presence of anti-Mup44

autoantibodies in patient sera. One of these regions

seems to be particularly suitable for this purpose, because

peptides in this region were recognized by all of the anti-

Mup44–positive sIBM sera analyzed, but not by the sera

from healthy controls. The most N-terminal and central

epitope regions are rich in charged amino acids, and all

3 regions belong to the most hydrophilic parts of the

protein, as revealed by Kyte and Doolittle hydrophobicity

plots (data not shown), which suggests that these epi-

topes are exposed at the surface of the folded protein.

Despite the frequent recognition of linear 15-mer cN1A

peptides by patient sera, our data do not exclude the pos-

sibility that discontinuous or conformational epitopes are

equally or even more important for the recognition of

cN1A by autoantibodies. Because no data on the 3-

dimensional structure of native cN1A are available, more

detailed insight into the major B-cell epitopes of cN1A

Pluk et al: Anti–cN1A Autoantibodies

March 2013 405

has to await ultrastructural analyses of this or a structur-

ally related protein.

In conclusion, our study has identified a new bio-

marker for sIBM, the presence of which can be easily

assessed by a serological test. The availability of the

recombinant target protein of the anti-Mup44 antibodies

and insight into the regions of this protein that contain

the major continuous epitopes facilitate the rapid genera-

tion of such serological tests.

Acknowledgment

This work was supported in part by the European Union

Sixth Framework Program (project AutoCure; LSH-

018661; GP), European Science Foundation (EuMyoNet;

IL), Swedish Research Council (IL), Swedish Rheumatism

Association (IL), and King Gustaf V 80 Year Foundation

(IL), and through the regional agreement on medical train-

ing and clinical research (ALF) between Stockholm County

Council and Karolinska Institutet (IL).

We thank Dr M. Snoeck for human skeletal muscle

material, Dr B. Wieringa for mouse skeletal muscle sam-

ples, and E. Jemseby for handling the serum biobank.

Potential Conflicts of Interest

H.P.: patent and royalties pending, method of detecting

autoantibodies from patients with sporadic inclusion-

body myositis. U.A.B.: consultancy, Novartis. B.G.v.E.:

employment, Research Director of the European Neuro-

muscular Center; grants/grants pending, Global FSH,

Netherlands Organization for Scientific Research, Prinses

Beatrix Fonds, Dutch FSHD Foundation; patent and

royalties pending, method of detecting autoantibodies

from patients with sporadic inclusion-body myositis.

G.J.M.P.: consultancy, Euro-Diagnostica, Axis-Shield

Diagnostics; patent and royalties pending, method of

detecting autoantibodies from patients with sporadic

inclusion-body myositis.

References1. Amato AA, Barohn RJ. Inclusion body myositis: old and new con-

cepts. J Neurol Neurosurg Psychiatry 2009;80:1186–1193.

2. Dalakas MC. Sporadic inclusion body myositis-diagnosis, pathogene-sis and therapeutic strategies. Nat Clin Pract Neurol 200;2:437–447.

3. Machado P, Miller A, Holton J, Hanna M. Sporadic inclusion bodymyositis: an unsolved mystery. Acta Reumatol Port 2009;34:161–182.

4. Dalakas MC, Hohlfeld R. Polymyositis and dermatomyositis. Lan-cet 2003;362:971–982.

5. Dalakas MC. Immunotherapy of myositis: issues, concerns andfuture prospects. Nat Rev Rheumatol 2010;6:129–137.

6. Hengstman GJ, van den Hoogen FH, van Engelen BG. Treatmentof the inflammatory myopathies: update and practical recommen-dations. Expert Opin Pharmacother 2009;10:1183–1190.

7. Lundberg IE, Grundtman C. Developments in the scientific andclinical understanding of inflammatory myopathies. Arthritis ResTher 2008;10:220.

8. Mammen AL. Dermatomyositis and polymyositis: clinical presenta-tion, autoantibodies, and pathogenesis. Ann N Y Acad Sci 2010;1184:134–153.

9. Needham M, Mastaglia FL. Sporadic inclusion body myositis: acontinuing puzzle. Neuromuscul Disord 2008;18:6–16.

10. Weihl CC, Pestronk A. Sporadic inclusion body myositis: possiblepathogenesis inferred from biomarkers. Curr Opin Neurol 2010;23:482–488.

11. Askanas V, Engel WK, Nogalska A. Inclusion body myositis: a de-generative muscle disease associated with intra-muscle fibermulti-protein aggregates, proteasome inhibition, endoplasmicreticulum stress and decreased lysosomal degradation. BrainPathol 2009;19:493–506.

12. Nogalska A, D’Agostino C, Engel WK, et al. Novel demonstrationof amyloid-beta oligomers in sporadic inclusion-body myositismuscle fibers. Acta Neuropathol 2010;120:661–666.

13. Kusters B, van Hoeve BJ, Schelhaas HJ, et al. TDP-43 accumula-tion is common in myopathies with rimmed vacuoles. Acta Neuro-pathol 2009;117:209–211.

14. Grundtman C, Malmstrom V, Lundberg IE. Immune mechanismsin the pathogenesis of idiopathic inflammatory myopathies. Arthri-tis Res Ther 2007;9:208.

15. Askanas V, Engel WK. Inclusion-body myositis: a myodegenerativeconformational disorder associated with Abeta, protein misfold-ing, and proteasome inhibition. Neurology 2006;66:S39–S48.

16. Greenberg SA. Theories of the pathogenesis of inclusion bodymyositis. Curr Rheumatol Rep 2010;12:221–228.

17. Henriques-Pons A, Nagaraju K. Nonimmune mechanisms of mus-cle damage in myositis: role of the endoplasmic reticulum stressresponse and autophagy in the disease pathogenesis. Curr OpinRheumatol 2009;21:581–587.

18. Dalakas MC. Inflammatory, immune, and viral aspects of inclusion-body myositis. Neurology 2006;66:S33–S38.

19. Bradshaw EM, Orihuela A, McArdel SL, et al. A local antigen-driven humoral response is present in the inflammatory myopa-thies. J Immunol 2007;178:547–556.

20. Greenberg SA, Bradshaw EM, Pinkus JL, et al. Plasma cells inmuscle in inclusion body myositis and polymyositis. Neurology2005;65:1782–1787.

21. Greenberg SA, Sanoudou D, Haslett JN, et al. Molecular profilesof inflammatory myopathies. Neurology 2002;59:1170–1182.

22. Badrising UA, Schreuder GM, Giphart MJ, et al. Associations withautoimmune disorders and HLA class I and II antigens in inclusionbody myositis. Neurology 2004;63:2396–2398.

23. Rojana-Udomsart A, James I, Castley A, et al. High-resolutionHLA-DRB1 genotyping in an Australian inclusion body myositis (s-IBM) cohort: an analysis of disease-associated alleles and diplo-types. J Neuroimmunol 2012;250:77–82.

24. Hengstman GJ, Brouwer R, Egberts WT, et al. Clinical and sero-logical characteristics of 125 Dutch myositis patients. Myositis spe-cific autoantibodies aid in the differential diagnosis of theidiopathic inflammatory myopathies. J Neurol 2002;249:69–75.

25. Brouwer R, Hengstman GJ, Vree Egberts W, et al. Autoantibodyprofiles in the sera of European patients with myositis. Ann RheumDis 2001;60:116–123.

26. Chahin N, Engel AG. Correlation of muscle biopsy, clinicalcourse, and outcome in PM and sporadic IBM. Neurology 2008;70:418–424.

27. Ronnelid J, Barbasso Helmers S, Storfors H, et al. Use of a com-mercial line blot assay as a screening test for autoantibodies ininflammatory myopathies. Autoimmun Rev 2009;9:58–61.

ANNALS of Neurology

406 Volume 73, No. 3

28. Hengstman GJ, van Engelen BG, Badrising UA, et al. Presence ofthe anti-Jo-1 autoantibody excludes inclusion body myositis. AnnNeurol 1998;44:423.

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

30. Abdo WF, van Mierlo T, Hengstman GJ, et al. Increased plasmaamyloid-beta42 protein in sporadic inclusion body myositis. ActaNeuropathol 2009;118:429–431.

31. Verschuuren JJ, Badrising UA, van Engelen B, et al. Inclusionbody myositis. In: Emery AEH, ed. Diagnostic criteria for neuro-muscular disorders. London, UK: Royal Society of Medicine Press,1997:81–84.

32. Tanimoto K, Nakano K, Kano S, et al. Classification criteriafor polymyositis and dermatomyositis. J Rheumatol 1995;22:668–674.

33. Badrising UA, Maat-Schieman ML, Ferrari MD, et al. Comparison ofweakness progression in inclusion body myositis during treatmentwith methotrexate or placebo. Ann Neurol 2002;51:369–372.

34. Pandya JM, Fasth AE, Zong M, et al. Expanded T cell receptorVbeta-restricted T cells from patients with sporadic inclusion bodymyositis are proinflammatory and cytotoxic CD28null T cells. Ar-thritis Rheum 2010;62:3457–3466.

35. Hengstman GJ, van Brenk L, Vree Egberts WT, et al. High speci-ficity of myositis specific autoantibodies for myositiscompared with other neuromuscular disorders. J Neurol 2005;252:534–537.

36. Dalakas MC, Illa I, Gallardo E, Juarez C. Inclusion body myositisand paraproteinemia: incidence and immunopathologic correla-tions. Ann Neurol 1997;41:100–104.

37. Dalakas MC. Inflammatory muscle diseases: a critical review onpathogenesis and therapies. Curr Opin Pharmacol 2010;10:346–352.

38. El-Shammaa NA, Fishbein WN, Armbrustmacher VW. Interstitial50-nucleotidase stain for frozen biopsy specimens of skeletal mus-cle. A useful adjunct in the diagnosis of polymyositis. Arch PatholLab Med 1984;108:251–256.

Pluk et al: Anti–cN1A Autoantibodies

March 2013 407