Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Arthritis & Rheumatology

Vol. 72, No. 1, January 2020, pp 179–191


DOI 10.1002/art.41075
© 2019, American College of Rheumatology

Proinflammatory Histidyl–Transfer RNA


Synthetase–Specific CD4+ T Cells in the Blood and Lungs of
Patients With Idiopathic Inflammatory Myopathies
Angeles S. Galindo-Feria,1 Inka Albrecht,1 Cátia Fernandes-Cerqueira,1 Antonella Notarnicola,1 Eddie A. James,2
Jessica Herrath, Maryam Dastmalchi,1 Tatyana Sandalova,3 Lars Rönnblom,4 Per-Johan Jakobsson,1
1

Maryam Fathi,5 Adnane Achour,3 Johan Grunewald,1 Vivianne Malmström,1 and Ingrid E. Lundberg1

Objective. Autoantibodies targeting histidyl–transfer RNA synthetase (HisRS; anti–Jo-­1) are common in the
i­diopathic inflammatory myopathies (IIMs) and antisynthetase syndrome. This study was undertaken to investigate
immunity against HisRS in the blood and lungs of patients with IIM/antisynthetase syndrome.
Methods. Bronchoalveolar lavage (BAL) fluid, BAL fluid cells, and peripheral blood mononuclear cells (PBMCs)
from patients with IIM/antisynthetase syndrome (n = 24) were stimulated with full-­length HisRS protein or a HisRS-­
derived peptide (HisRS11–23). BAL fluid and PBMCs from patients with sarcoidosis (n = 7) and healthy subjects (n = 12)
were included as controls. The CD4+ T cell response was determined according to levels of CD40L up-­regulation
and cytokine expression using flow cytometry. Anti–Jo-­1 autoantibody responses in the serum and BAL fluid were
assessed by enzyme-­linked immunosorbent assay. Lung biopsy samples from patients with IIM/antisynthetase syn-
drome (n = 14) were investigated by immunohistochemistry.
Results. In BAL fluid, CD4+ T cells from 3 of 4 patients with IIM/antisynthetase syndrome responded to stimula-
tion with HisRS protein, as measured by the median fold change in CD40L expresssion in stimulated cells compared
to unstimulated cells (median fold change 3.6, interquartile range [IQR] 2.7–14.7), and 2 of 3 patients with IIM/antisyn-
thetase syndrome had the highest responses to HisRS11–23 (median fold change 88, IQR 27–149). In PBMCs, CD4+ T
cells from 14 of 18 patients with IIM/antisynthetase syndrome responded to HisRS protein (median fold change 7.38,
IQR 2.69–31.86; P < 0.001), whereas a HisRS11–23 response was present in 11 of 14 patients with IIM/antisynthetase
syndrome (median fold change 3.4, IQR 1.87–10.9; P < 0.001). In the control group, there was a HisRS11–23 response in
3 of 7 patients with sarcoidosis (median fold change 2.09, IQR 1.45–3.29) and in 5 of 12 healthy controls (median fold
change 2, IQR 1.89–2.42). CD4+ T cells from patients with IIM/antisynthetase syndrome displayed a pronounced Th1
phenotype in the BAL fluid when compared to the PBMCs (P < 0.001), producing high amounts of interferon-­γ and
interleukin-­2 following stimulation. Anti–Jo-­1 autoantibodies were detected in BAL fluid and germinal center (GC)–like
structures were seen in the lung biopsy samples from patients with IIM/antisynthetase syndrome.
Conclusion. The results of this study demonstrate a pronounced presence of HisRS-­reactive CD4+ T cells in
PBMCs and BAL fluid cells from patients with IIM/antisynthetase syndrome as compared to patients with sarcoido-
sis and healthy controls. These findings, combined with the presence of anti–Jo-­1 autoantibodies in BAL fluid and
GC-­like structures in the lungs, suggest that immune activation against HisRS might take place within the lungs of
patients with IIM/antisynthetase syndrome.

Supported by L’Association française contre les myopathies (AFM- Molecular Medicine, Stockholm, Sweden; 2Eddie A. James, PhD: Benaroya
Téléthon), Mexican National Council for Science and Technology (Conacyt), Research Institute at Virginia Mason Hospital, Seattle, Washington;
3
Karolinska Institutet (KID funding), King Gustaf V 80 Year Foundation, Konung Tatyana Sandalova, PhD, Adnane Achour, PhD: Science for Life Laboratory,
Gustaf V:s och Drottning Victorias Frimurarestiftelse, Reumatikerförbundet, Department of Medicine Solna, Karolinska Institutet, and Department
Stockholm County Council (ALF project), Myositis Association, Swedish Heart- of Infectious Diseases, Karolinska University Hospital, Solna, Stockholm,
Lung Foundation, Swedish Research Council (grant K2014-52X-14045-14-3), Sweden; 4Lars Rönnblom, MD, PhD: Science for Life Laboratory, Stockholm,
and Swedish Rheumatism Association. Sweden, and Uppsala University, Uppsala, Sweden; 5Maryam Fathi, MD,
1
Angeles S. Galindo-Feria, MD, Inka Albrecht, PhD, Cátia Fernandes- PhD: Karolinska University Hospital, Stockholm, Sweden.
Cerqueira, PhD, Antonella Notarnicola, MD, Jessica Herrath, PhD, Maryam Drs. Galindo-Feria and Albrecht contributed equally to this work.
Dastmalchi, MD, PhD, Per-Johan Jakobsson, MD, PhD, Johan Grunewald, Dr. Albrecht owns stock in Sanofi Genzyme. Drs. Albrecht and Lundberg
MD, PhD, Vivianne Malmström, PhD, Ingrid E. Lundberg, MD, PhD: have filed a patent application on a diagnostic test to identify patients with
Karolinska Institutet, Karolinska University Hospital, and Center for anti-FHL1 autoantibodies. Dr. Rönnblom has received research support from

179
180 | GALINDO-­FERIA ET AL

INTRODUCTION of the HisRS protein, makes this region an attractive part of the
protein to search for T cell–reactive peptides.
Idiopathic inflammatory myopathies (IIMs) represent a group An additional question is, in which body compartment might
of chronic autoimmune disorders that are characterized by mus- HisRS-­reactive T cells be triggered? An interesting possibility is
cle weakness and systemic extramuscular manifestations in the that such immune activation may take place in the lungs, as indi-
joints, skin, heart, and, particularly, the lungs (1). The identification cated by the detection of expanded T cell clones in bronchoal-
of autoantibodies unique to IIMs, also known as myositis-­specific veolar lavage (BAL) fluid and peripheral blood from patients with
autoantibodies (MSA), has allowed recognition of correlations IIM-­
associated interstitial pneumonitis (24), and the finding of
with distinct clinical phenotypes (2,3). Among the MSA, anti–Jo-­1 expansions in BAL fluid CD4+ and CD8+ T cells and correspond-
autoantibodies targeting histidyl-­transfer RNA synthetase (HisRS) ing biased T cell receptor V-­gene usage in the muscle of patients
are the most common (found in 25–35% of patients with IIM) (4–6) with IIM (25). These observations suggest the involvement of a
and are associated with interstitial lung disease (ILD) and antisyn- shared antigen–driven immune response in the inflammatory pro-
thetase syndrome (3,7). cess between the lungs and muscles of these patients. However,
Insights from studies of several autoimmune diseases have antigen-­specific T cell responses in cells derived from the lungs of
placed the presence and presentation of the autoantigen in the lung, patients with IIM have hitherto not been demonstrated.
where innate immunity and genetics play important roles in the ini- The present study aimed to address T cell specificity and trig-
tiation of the autoimmune response, as seen in rheumatoid arthritis gering in anti–Jo-­1+ patients with IIM/antisynthetase syndrome.
(RA) (8–10), sarcoidosis (11), and multiple sclerosis (MS) (12). Sev- Specifically, we aimed to investigate 1) T cell reactivity and phe-
eral genetic and experimental findings support the role of adaptive notype upon stimulation with full-­ length HisRS protein and an
immunity in anti–Jo-­1 autoantibody formation. Thus, HLA–DRB1*03 HLA–DRB1*03 binding HisRS-­ derived peptide in PBMCs from
has been found to be strongly associated with anti–Jo-­1 autoan- anti–Jo-­1+ patients with IIM/antisynthetase syndrome, 2) whether
tibodies (13–15). Furthermore, observations of gene–environment such antigen-­ specific T cells are present in the lungs of anti–­
interactions suggest an additional risk of anti–Jo-­1 autoantibodies Jo-­1+ patients with IIM/antisynthetase syndrome, and 3) whether
in HLA–DRB1*03–positive patients who are ever smokers (14), indi- immune activation and the presence of anti–Jo-­1 autoantibodies
cating the importance of environmental exposures in the develop- can be assessed in the lungs of anti–Jo-­1+ patients with IIM/anti-
ment of anti–Jo-­1 autoantibodies. In addition, the presence of class synthetase syndrome.
switching, epitope spreading, and affinity maturation of anti–Jo-­1
autoantibodies points toward a significant contribution of antigen-­
PATIENTS AND METHODS
specific T cells in the development of these antibodies (16–19).
A question that has been difficult to elucidate is, how might Patients and selection criteria. Samples were collected
HisRS, a ubiquitous cytoplasmic protein, induce an autoimmune between November 2010 and June 2013 at the Rheumatology
reaction? The high expression of HisRS in specific microenviron- Unit and Respiratory Medicine Unit of Karolinska University Hospital
ments, such as in normal lung tissue (20), combined with altered (Stockholm, Sweden). Patients with IIM fulfilled the criteria for definite,
expression of HisRS and/or an enriched conformation of highly sen- probable, or possible polymyositis or dermatomyositis according to
sitive HisRS in response to granzyme B cleavage in the lung (21), the Bohan and Peter criteria (26,27). The diagnosis of ILD, meeting
supports the role of the lung as a potential site where immune toler- the American Thoracic society criteria (28), was based on findings
ance could be broken. A previous study demonstrated the presence from chest radiography/high-­resolution computed tomography and/
of T cells reactive to both intact HisRS protein and HisRS fragments, or pulmonary function tests. The diagnosis of antisynthetase syn-
most of which were ~150 amino acids in length, in peripheral blood drome was based on the presence of anti–Jo-­ 1 autoantibodies
mononuclear cells (PBMCs) from patients with IIM as well as PBMCs in addition to one of the following features: ILD, myositis, arthritis,
from healthy controls (22). Other observations revealed that both Raynaud’s phenomenon, fever, or mechanic’s hands (7).
intact and granzyme B–cleaved HisRS, corresponding to residues
1–48, can drive specific immune responses, including recruitment Sample collection. PBMCs were obtained from anti–Jo-­1+
of CD4+ and CD8+ T cells, as well as immature dendritic cells and patients with IIM/antisynthetase syndrome (n = 15; 6 male and 9
activated monocytes (23). This finding, together with identification of female) and anti–Jo-­1− patients (n = 3; all female) (corresponding
a B cell epitope among the 60 amino acids of the N-­terminal region to patients P1–P18 in Table 1).

AstraZeneca. Dr. Lundberg has received consulting fees, speaking fees, and/or Address correspondence to Angeles S. Galindo-Feria, MD, Karolinska
honoraria from Corbus (less than $10,000 each), consulting fees and honoraria University Hospital, Karolinska Institutet, Department of Medicine, Division
from Bristol-Myers Squibb (less than $10,000), consulting fees from aTyr (less of Rheumatology, Center for Molecular Medicine, L8:04 SE-171 76 Stockholm,
than $10.000), honoraria from MedImmune (less than $10,000), research Sweden. E-mail: angeles.galindo.feria@ki.se.
support from Bristol-Myers Squibb and AstraZeneca, and owns stock or stock Submitted for publication November 23, 2018; accepted in revised form
options in Roche. No other disclosures relevant to this article were reported. August 6, 2019.
Table 1. Clinical and laboratory characteristics of the 24 patients with idiopathic inflammatory myopathy/antisynthetase syndrome according to type of experiment*
T cell reactivity†
HLA–DRB1
PBMCs BAL fluid
Muscle Anti–Jo-­1 status genotype
Full-­length Full-­length inflammatory
Patient Diagnosis protein HisRS11–23 protein HisRS11–23 infiltrates Serum BAL fluid Allele 1 Allele 2
P1 PM/antisynthetase syndrome + Not tested Nonviable cells Nonviable cells Present Positive‡§¶ Negative‡ 04 15
P2 Antisynthetase syndrome/ILD ++ ++ ++ +++ Absent Positive‡ Positive‡ 01 13
P3 Antisynthetase syndrome/ILD ++ +++ + +++ Absent Positive‡§¶ Positive‡ NA NA
P4 DM ++ +++ −# +# Present Negative‡§ Negative‡ 09 13
P5 PM +++ Not tested +++ Not tested Present Negative‡ Negative‡ 03 10
ANTIGEN-­SPECIFIC T CELLS IN INFLAMMATORY MYOPATHIES

P6 DM/ILD − + Nonviable cells Nonviable cells Absent Negative‡ NA 03 03


P7 Antisynthetase syndrome/ILD +++ + NA NA Absent Positive‡ NA 14 15
P8 Antisynthetase syndrome/ILD ++ ++ NA NA Absent Positive‡§¶ Positive‡ 03 13
P9 Antisynthetase syndrome/ILD +++ + NA NA Absent Positive‡ Positive‡ 03 04
P10 Antisynthetase syndrome/ILD ++ ++ NA NA Present Positiveद NA 03 15
P11 Antisynthetase syndrome/ILD ++ − NA NA Present Positive‡§¶ NA 03 13
P12 Antisynthetase syndrome/ILD +++ ++ NA NA Present Positive‡ NA 03 04
P13 Antisynthetase syndrome/ILD +++ + NA NA Present Positiveद NA 03 04
P14 Antisynthetase syndrome/ILD +++ − NA NA Present Positive‡§¶ NA 03 04
P15 Antisynthetase syndrome + Not tested NA NA Absent PositiveঠNA 01 03
P16 Antisynthetase syndrome/ILD − Not tested NA NA Present Positive§ NA 03 15
P17 Antisynthetase syndrome/ILD − − NA NA Absent Positive‡¶ NA 03 15
P18 DM − − NA NA Present Positive‡ NA 03 13
P19 Antisynthetase syndrome/ILD NA NA NA NA Present Positive‡§¶ Positive‡ 01 11
P20 DM/ILD NA NA NA NA Present Negative‡§¶ Negative‡ 03 15
P21 DM NA NA NA NA Absent Negative‡§¶ Negative‡ 04 07
P22 PM NA NA NA NA Present Negative‡§¶ Negative‡ 01 16
P23 DM/ILD NA NA NA NA Absent Negative‡ Negative‡ 03 04
P24 PM/antisynthetase syndrome NA NA NA NA Present Positive‡¶ Positive‡ 03 03
* HisRS11–23 = histidyl–transferase RNA synthetase peptide 11–23; PM = polymyositis; ILD = interstitial lung disease; NA = not available; DM = dermatomyositis.
† In peripheral blood mononuclear cells (PBMCs) and bronchoalveolar lavage (BAL) fluid cells, + = low reactivity, ++ = medium reactivity, +++ = high reactivity, and − = nonreactive.
‡ Detected by enzyme-­linked immunosorbent assay.
§ Detected by immunoprecipitation.
¶ Detected by line blotting.
# Analysis of BAL fluid cells with short-­term stimulation (1 day), and in low-­viability cells with long-­term stimulation (5 days).
|
181
182 | GALINDO-­FERIA ET AL

From 6 of these patients, paired BAL fluid and BAL fluid able paired serum samples and cell-­depleted BAL fluid were
cell pellets were obtained by bronchoscopy (from 3 anti–Jo-­1+ collected (n = 9; 4 male and 5 female). All samples were pro-
patients and 3 anti–Jo-­1− patients [patients P1–P6 in Table 1]) vided by the Respiratory Medicine Unit, according to a published
(29). Additional cell-­depleted BAL fluid samples were obtained protocol (29). Descriptions of the lung and lymph node biopsy
from 4 anti–Jo-­ 1+ patients with IIM/antisynthetase syndrome samples from patients with sarcoidosis, as detailed in pathology
(1 male and 3 female) and 4 anti–Jo-­1− patients (1 male and 3 reports, are presented in Supplementary Table 4 (available at
female) (patients P8 and P9 and P19–P24 in Table 1). All samples http://onlin​elibr​ary.wiley.com/doi/10.1002/art.41075/​abstract).
were stored at −80°C at the Human Laboratory Area of the Center For the healthy control T cell experiments, fresh PBMCs were
for Molecular Medicine (Solna, Stockholm, Sweden). In addition, isolated from buffy coats and were obtained from healthy donors
serum samples were obtained from anti–Jo-­1+ patients with IIM/ who expressed at least one copy of the HLA–DRB1*03 allele
antisynthetase syndrome (n = 17; 6 male and 11 female) and anti– (n = 12; 3 male and 9 female). Blood samples from healthy sub-
Jo-­1− patients (n = 7; 1 male and 6 female) (patients P1–P24 in jects were provided by Uppsala Bioresource at Uppsala University
Table 1). Hospital in Sweden. For the autoantibody analysis, additional paired
Patients included in the experiments were clustered in groups serum and cell-­depleted BAL fluid from healthy controls (n = 18; 8
according to the availability of samples. Group 1 included patients male and 10 female) were provided by the Respiratory Medicine
with available PBMCs, BAL fluid, BAL fluid cell pellets, and paired Unit.
serum samples (patients P1–P6). Group 2 included patients with
available PBMCs and paired serum samples (patients P7–P18). Ethics. The study was approved by the Regional Ethics
Group 3 included patients with serum samples and cell-­depleted Committee of Stockholm at Karolinska Institutet and Uppsala
BAL fluid samples (patients P8 and P9 and P19–P24). T cell University, and performed in accordance with the Declara-
experiments included patients from group 1 and group 2. Anti– tion of Helsinki guidelines on studies with human subjects. All
Jo-­1 antibody experiments included patients from group 1 and ­subjects provided written informed consent prior to any study
group 3. For technical reasons (low cell numbers, low cell viability procedure.
of BAL fluid cells, or low volume of BAL fluid), some samples were
excluded. Details regarding the distribution of patients accord- Functional T cell assay. Fresh samples were used for all
ing to the type of analysis, degree of T cell reactivity, presence or analyses. PBMCs were obtained from heparinized blood and
absence of muscle inflammatory infiltrates, antibody status, and were prepared by centrifugation over Ficoll-­Hypaque gradi-
HLA genotyping are provided in Table 1. ents. PBMCs were resuspended in 96-­well plates in RPMI 1640
Lung tissue specimens from patients with IIM/antisynthetase medium supplemented with 10% heat-­inactivated fetal bovine
syndrome (n = 14; 4 anti–Jo-­1+ and 10 anti–Jo-­1−) and from serum (1 × 106 cells/well) according to an adapted protocol
comparator cases (patients with chronic obstructive lung disease (30). Cells were incubated for 5 days with recombinant human
[COPD]; n = 10) were retrieved from regional biobanks. full-­length HisRS (20 μg/ml; Bio Supply), with the candidate
Age, sex, smoking status, diagnosis, disease duration from peptide HisRS11–23 (20 μg/ml, sequence VKLQGERVRGLKQ;
onset of symptoms, clinical manifestations, anti–Jo-­1 antibody GenScript) or with RPMI medium as a negative control.
status, and treatment at the time of blood/BAL fluid sampling Cells were restimulated on day 5 for 8 hours, and brefeldin
are summarized in Table 1 and Supplementary Table 1 (available A (10 μg/ml; Sigma-­Aldrich) was added for the last 6 hours.
on the Arthritis & Rheumatology web site at http://onlin​elibr​ary. Stimulation with staphylococcal enterotoxin B (10 μg/ml,
wiley.com/doi/10.1002/art.41075/​abstract). Features of the mus- Sigma-­Aldrich) was used as a positive control.
cle biopsy specimens obtained at the time of disease onset from Following stimulation, cells were stained with Live/Dead
patients with IIM/antisynthetase syndrome, as detailed in pathol- Fixable Green Dead Cell Stain as well as with antibodies toward
ogy reports, are summarized in Supplementary Table 2 (http:// CD14, CD3, CD4, CD40L, interferon-­γ (IFNγ), interleukin-­2 ­(IL-­2),
onlin​elibr​ary.wiley.com/doi/10.1002/art.41075/​abstract). and IL-­17A. In some experiments, blocking antibodies for HLA–
DR or HLA–DQ or staining with anti-­ CXCR3, anti-­ CCR5, and
Control groups. For the disease control T cell experi- anti-­CCR6 antibodies were used. Details on the antibodies used
ments, paired PBMCs and BAL fluid cell pellets were retrieved are specified in Supplementary Materials and Methods and Sup-
from patients with sarcoidosis (n = 7; 6 male and 1 female) plementary Table 5 (available at http://onlin​ elibr​
ary.wiley.com/
(patients Sarc1–Sarc7 in Supplementary Table 3, avail­able at doi/10.1002/art.41075/​abstract).
http://onlin​elibr​ary.wiley.com/doi/10.1002/art.41075/​abstract),
since this disease is characterized by the presence of auto- Flow cytometry. Cells were run on a Gallios Analyzer
immune features along with ILD, and is associated with HLA– (Beckman Coulter), and data were analyzed using FlowJo soft-
DRB1*03:01. For detection of anti–Jo-­ 1 autoantibodies, ware, version 7.5.1 (Tree Star). The gating strategy is specified
additional control patients with sarcoidosis and with avail­ in Supplementary Materials and Methods and Supplementary
ANTIGEN-­SPECIFIC T CELLS IN INFLAMMATORY MYOPATHIES | 183

Figure 1 (available at http://onlin​elibr​ary.wiley.com/doi/10.1002/ ditions, Wilcoxon’s signed-­rank test was used. Two-­tailed P values
art.41075/​abstract). less than 0.05 were considered significant. Up-­regulated expression
of CD40L in T cells was normalized to the value in unstimulated cells,
Identification of the HisRS T cell epitope. The predic- and presented as the median fold change according to the following
tion of a CD4+ T cell epitope was based on the crystal struc- categories: low (<3-­fold), intermediate (3–10-­fold), or high (>10-­fold).
ture of the HLA–DRB1*03:01 molecule in complex with class The analyses were performed using Prism 5 software (Graph Pad)
II–­associated invariant chain peptide, which reveals the binding and IBM SPSS Statistics, version 25.
pockets of HLA and how the anchor residues are docking into
HLA (31). Prior motif studies established that HLA–DRB1*03:01
RESULTS
epitopes most commonly contain an aliphatic residue in position
1, an acidic or polar residue in position 4, and either a basic Clinical characteristics of the patients and controls.
residue at position 6 paired with a small residue at position 9 A total of 17 anti–Jo-­1+ and 7 anti–Jo-­1− patients with IIM/anti-
(submotif 1) or a nonbasic residue at position 6 paired with a synthetase syndrome were included in our analyses (Table 1). The
large residue at position 9 (submotif 2) (32). In this way, a theo- anti–Jo-­1+ patients (median age 55 years [IQR 53–55 years]) had a
retical prediction of CD4+ T cell epitopes can be made from any longer disease duration and higher frequency of ILD compared to
antigen. In our case, the sequence of the first 60 amino acids of the anti–Jo-­1− patients (median age 57 years [IQR 54–57 years])
HisRS (UniProtKB identifier P12081) was examined to predict (P = 0.033 and P = 0.021, respectively). There was no difference
peptides that can bind DRB1*03:01; residues 11–23 (HisRS11–23) in sex, age, or smoking status between the 2 groups of patients
contained 2 such motifs and therefore had a high probability of (P = 0.625, P = 0.425, and P = 0.724, respectively). A compari-
containing a bona fide DRB1*03:01 epitope. Thus, HisRS11–23 son of the characteristics of the 2 groups of patients is presented
peptide was selected for further functional assays (Figure 1A). in Supplementary Table 7 (available at http://onlin​elibr​ary.wiley.com/
doi/10.1002/art.41075/​abstract).
BAL fluid and serum samples for detection of anti–Jo-­1 Group 1 (T cell group, patients P1–P6) had a median dis-
autoantibodies. Anti–Jo-­1 IgG and total IgG were measured by ease duration of 5 months (IQR 1–48 months). Five patients in
enzyme-­linked immunosorbent assay (ELISA) in BAL fluid and serum this group were untreated and 1 patient was receiving immu-
from patients with IIM/antisynthetase syndrome, control patients nosuppressive treatment. Group 2 (T cell group, patients P7–
with sarcoidosis, and healthy controls. Details on the protocol P18) had a median disease duration of 64 months (IQR 1–193
used are presented in Supplementary Materials and Methods months), and 9 of 12 patients were receiving immunosuppres-
(http://onlin​e​libr​ary.wiley.com/doi/10.1002/art.41075/​abstract). sive treatment. Group 3 (antibody group, patients P19–P24) had
a median disease duration of 13 months (IQR 4–60 months),
Immunohistochemistry and histopathology. Paraffin-­ and 4 of 6 patients were receiving immunosuppressive treat-
embedded lung tissue from patients with IIM/antisynthetase ment (see Supplementary Table 1 [http://onlin​elibr​ary.wiley.com/
syndrome and patients with COPD were analyzed by immuno- doi/10.1002/art.41075/​abstract]).
histochemistry according to a previously published protocol (33). The control patients with sarcoidosis and healthy controls
The primary antibodies used were anti-­ CD3, anti-­
CD138, anti-­ assessed in T cell and antibody analyses had a median age of
CXCR3, and anti-­CCR5 or the isotype controls IgG, IgG2b, and 55 years (IQR 43–62 years) and 33 years (IQR 28–49 years),
IgG1. Details on the protocol and antibodies used for ELISAs and respectively. Detailed clinical information on the patients with
immunohistochemistry are specified in Supplementary Materials sarcoidosis is presented in Supplementary Table 3 (http://onlin​e​
and Methods, Supplementary Table 6, and Supplementary Figure libr​ary.wiley.com/doi/10.1002/art.41075/​abstract).
2 (available at http://onlin​elibr​ary.wiley.com/doi/10.1002/art.41075/​
abstract). Whole sections were evaluated independently by 2 inves- Presence of HisRS-­T cell reactivity in the peripheral
tigators (AGF and CFC) on coded slides by semiquantitative scoring blood of patients with IIM/antisynthetase syndrome. To
for the extent of immunohistochemical staining on a 4-­point scale, identify the potential presence of antigen-­specific T cells against
where 0 = none, 1 = low, 2 = intermediate, and 3 = high amount HisRS in the blood of patients with IIM/antisynthetase syndrome, we
of staining. Results are expressed as the mean of 2 observations. assessed the up-­regulation of CD40L in CD4+ T cells following stim-
ulation with HisRS protein or HisRS11–23 in patients from group 1 and
Statistical analysis. Continuous variables are expressed as group 2. After stimulation of PBMCs with HisRS protein, we iden-
the median with interquartile range (IQR; 25th–75th percentiles) or tified up-­regulation of CD40L on CD4+ T cells in 14 of 18 patients
minimum and maximum values. Categorical variables are presented with IIM/antisynthetase syndrome, with a median fold change of
as frequencies and percentages. Differences between groups were 7.38 (IQR 2.69–31.86) relative to unstimulated cells (P = 0.0002).
analyzed using Mann-­Whitney U test. For comparison of the per- High (>10-­fold) up-­regulation of CD40L in CD4+ T cells fol-
centage of CD40L up-­regulation between different stimulation con- lowing stimulation with the HisRS protein was observed in 6 of 14
184 | GALINDO-­FERIA ET AL

Figure 1. Anti–histidyl–transfer RNA synthetase (HisRS)–specific T cells are enriched in the blood of patients with idiopathic inflammatory
myopathy (IIM)/antisynthetase syndrome. A, A 13-­mer peptide within the first 60 amino acids of HisRS was predicted to represent a potential
T cell epitope, HisRS11–23. B, Representative plots from fluorescence-­activated cell sorter analysis (gated on lymphocytes and CD4+CD3+ T
cells, with exclusion of dead cells and monocytes) show up-­regulation of CD40L in CD4+ T cells as well as production of interferon-­γ (IFNγ),
interleukin-­2 (IL-­2), and IL-­17 on peripheral blood mononuclear cells (PBMCs) from an anti–Jo-­1+ patient with IIM/antisynthetase syndrome,
in unstimulated (un-­stim) conditions or after stimulation with full-­length HisRS protein or HisRS11–23. C–E, Up-­regulation of CD40L expression
was examined on CD4+ T cells in PBMCs from anti–Jo-­1+ patients with IIM/antisynthetase syndrome (n = 18) (patients P1–P18 in Table 1)
(C), patients with sarcoidosis (n = 6) (D), and HLA–DRB1*03–positive healthy controls (HCs) (n = 12) (E). Solid symbols in C–E represent the
median fold change in PBMCs from these groups, while gray-­shaded symbols represent the median fold change in PBMCs from anti–Jo-­
1− patients (C), patients with sardoidosis who showed reactivity toward HisRS protein and HisRS11–23 (D), and a healthy control subject who
showed reactivity to HisRS protein (E). Categories of fold change were low (<3-­fold), intermediate (3–10-­fold), or high (>10-­fold). Cells were
left unstimulated or stimulated with full-­length HisRS protein, HisRS11–23, or staphylococcal enterotoxin B (SEB) as a positive control. F, IFNγ
production was assessed on CD40L+CD4+ T cells in PBMCs from patients with IIM/antisynthetase syndrome. Solid symbols represent anti–
Jo-­1+ patients and gray-­shaded symbols represent anti–Jo-­1− patients; horizontal lines show the median. Cells were left unstimulated or
stimulated with full-­length HisRS recombinant protein, HisRS11–23, or SEB. * = P < 0.05 by 2-­tailed Wilcoxon’s matched-­pairs signed rank test.

patients with IIM/antisynthetase syndrome (5 anti–­Jo-­1+ and 1 After stimulation of PBMCs with HisRS11–23, we detected
anti–Jo-­1−), with a median fold change of 35 (IQR 22.76–48.53) ­up-­regulation of CD40L on CD4+ T cells in 11 of 14 patients with
relative to unstimulated cells (Figures 1B and C). The highest IIM/antisynthetase syndrome, with a median fold change of 3.4
responses were present in the PBMCs from patients with IIM/ (IQR 1.87–10.9) relative to unstimulated cells (P = 0.0012). The
antisynthetase syndrome, specifically those who expressed at highest response toward HisRS11–23 (>10-­ fold change versus
least one copy of the HLA–DRB1*03 allele and were anti–Jo-­1+ unstimulated cells) was observed in 3 of 11 patients with IIM/
as compared to anti–Jo-­1+ patients without the HLA–DRB1*03 antisynthetase syndrome (2 anti–Jo-­1+ and 1 anti–Jo-­1−), with
allele (mean fold change in CD40L expression relative to unstim- a median fold change of 12.75 (IQR 10.9–37) (Figures 1B and C).
ulated cells 16.05 versus 7.57; P not significant) (see Supple- In the sarcoidosis group, up-­ regulation of CD40L was
mentary Figures 3A and C, available at http://onlin​elibr​ary.wiley. detected in PBMCs from 2 of 7 patients after stimulation
com/doi/10.1002/art.41075/​abstract). with HisRS protein, with a median fold change of 5.08 (IQR
ANTIGEN-­SPECIFIC T CELLS IN INFLAMMATORY MYOPATHIES | 185

A B Anti DR/DQ block


100

% CD40L upregulation
50

0
HisRS anti DQ anti DR
[11-23]

Figure 2. HisRS reactivity is restricted to HLA–DR. A, Representative plots from fluorescence-­activated cell sorter analysis show up-­regulation
of CD40L on CD4+ T cells in PBMCs from anti–Jo-­1+ patients with IIM/antisynthetase syndrome after stimulation with HisRS11–23 compared
to unstimulated cells. This up-­regulation was abolished in the presence of anti–HLA–DR but not in the presence of anti–HLA–DQ blocking
antibodies. B, Effects of anti–HLA–DQ and anti–HLA–DR blocking antibodies on CD40L up-­regulation by HisRS11–23 were quantified in PBMCs
from anti–Jo-­1+ patients with IIM/antisynthetase syndrome (n = 2; patients P2 and P12 in Table 1). Bars show the median percentage change
(with upper limit of maximum range) relative to HisRS11–23 (set at 100%). See Figure 1 for definitions.

3.7–6.47) relative to unstimulated cells, and detected in 3 of 7 selected BAL fluid cells and corresponding PBMCs from anti–­
patients following stimulation with HisRS11–23, with a median fold Jo-­ 1+ patients with IIM/antisynthetase syndrome (n = 2) and
change of 2.09 (IQR 1.45–3.29) relative to unstimulated cells anti–Jo-­1− patients (n = 2) (group 1, patients P2–P5 [Table 1])
(Figure 1D). The HisRS-­reactive patients with sarcoidosis were and patients with sarcoidosis (n = 7) (see Supplementary Table
positive for either HLA–DRB1*04/*15 or HLA–DRB1*01/*03 and 3 [http://onlin​elibr​ary.wiley.com/doi/10.1002/art.41075/​abstract]),
were former smokers (see Supplementary Table 3 [http://onlin​e​ and stimulated the cells with either HisRS protein or HisRS11–23.
libr​ary.wiley.com/doi/10.1002/art.41075/​abstract]). A CD40L response of BAL fluid cells stimulated with HisRS full-­
In the healthy control group who were positive for HLA– length protein was present in 3 of 4 patients with IIM/antisynthetase
DRB1*03, up-­regulation of CD40L was observed in PBMCs stim- syndrome (patients P2, P3, and P5), with a median fold change in
ulated with HisRS protein in 1 of 12 subjects (5-­fold change relative CD40L expression of 3.6 (IQR 2.68–14.7) relative to unstimulated
to unstimulated cells). After stimulation of PBMCs with HisRS11–23, cells. The corresponding CD4+ T cells from PBMCs had a similar
up-­regulation of CD40L was observed in 5 of 12 healthy controls, up-­regulation of CD40L in response to the HisRS protein in all 4
with a median fold change of 2 (IQR 1.89–2.42) relative to unstim- patients tested (patients P2–P5), with a median fold change of 4.58
ulated cells (P = 0.06) (Figure 1E). (IQR 3.25–25.36) relative to unstimulated cells (Figures 3A and B).
We next evaluated the proinflammatory effector functions of When the response to HisRS11–23 stimulation was analyzed
HisRS-­specific T cells by examining cytokine production within the in the lung compartment of patients with IIM/antisynthetase syn-
CD4+CD40L+ T cell population. The HisRS-­specific CD4+CD40L+ drome, we identified a high CD40L up-­regulation in BAL fluid
T cells in peripheral blood mainly produced IL-­2 and IFNγ. In the cells from 2 of 3 patients (patients P2 and P3) when compared
patients with IIM/antisynthetase syndrome, a median of 12% (IQR to unstimulated conditions, with a median fold change of 88 (IQR
5.39–25.6%) of CD4+CD40L+ T cells produced IFNγ after antigenic 27–149). Furthermore, the CD40L responses in PBMCs were also
stimulation with HisRS protein, and a median of 4% (IQR 2–18%) high in all 3 patients evaluated (patients P2–P4), with a median
produced IFNγ after stimulation with HisRS11–23 (P = 0.0084 and fold change of 12.7 (IQR 4.18–37) relative to unstimuated cells.
P = 0.02 respectively, compared to unstimulated cells) (Figure 1F). Moreover, the highest frequencies of CD4+CD40L+ T cells
To confirm an HLA–DR dependency of the observed T cell were found in BAL fluid after stimulation with HisRS11–23, with a
responses, PBMCs from 2 anti–Jo-­1+ patients were stimulated median fold change of 7.83% (IQR 1.37–29.9%), whereas in
with HisRS11–23 in the presence of anti–HLA–DR or anti–HLA–DQ PBMCs, the median fold change was 0.38% (IQR 0.02–5.89%),
blocking antibodies. Anti–HLA–DR blocking efficiently abrogated suggesting an enrichment of antigen-­specific T cells in the lung
the up-­regulation of CD40L upon stimulation with HisRS11–23, while compartment (see Supplementary Table 8, available at http://onlin​e​
anti–HLA–DQ blocking did not (Figures 2A and B), thereby indicat- libr​ary.wiley.com/doi/10.1002/art.41075/​abstract). In addition, the
ing that the HisRS11–23 epitope is presented by HLA–DR. highest reactivity to HisRS11–23 was seen in the 2 anti–Jo-­ 1+
patients with IIM/antisynthetase syndrome, whereas BAL fluid
Presence of HisRS-­specific T cells in the lungs of cells from 1 anti–Jo-­1− patient (patient P4) showed no response
patients with IIM/antisynthetase syndrome display a upon HisRS11–23 stimulation.
­pronounced Th1 phenotype as compared to PBMCs. To In order to characterize the phenotype of CD4+ T cells in
investigate whether the presence and functionality of antigen-­ BAL fluid and PBMCs, we selected unstimulated, paired CD4+
specific CD4+ T cells may vary in different compartments, we T cells from both compartments, obtained from 3 anti–Jo-­ 1+
186 | GALINDO-­FERIA ET AL

Figure 3. Anti–HisRS–specific T cells are detected in the lungs of patients with IIM/antisynthetase syndrome. A, Representative plots from
fluorescence-­activated cell sorter analysis (gated on lymphocytes and CD4+CD3+ T cells, with exclusion of dead cells and monocytes) show
up-­regulation of CD40L as well as production of IFNγ, IL-­2, and IL-­17 on CD4+ T cells obtained from bronchoalveolar lavage (BAL) fluid
(BALF) from an anti–Jo-­1+ patient with IIM/antisynthetase syndrome. BAL fluid T cells were left unstimulated or stimulated with full-­length
HisRS recombinant protein or HisRS11–23. B and C, CD40L up-­regulation on CD4+ T cells, relative to unstimulated cells, was compared
between BAL fluid cells and corresponding PBMCs from patients with IIM/antisynthetase syndrome (n = 4; patients P2–P5 in Table 1) (B) and
in BAL fluid CD4+ T cells between patients with IIM/antisynthetase syndrome (n = 4) and patients with sarcoidosis (n = 7) (C). Cells were left
unstimulated or stimulated with full-­length HisRS protein, HisRS11–23 peptide, or SEB for 5 days. Solid symbols represent the median fold change
in individual anti–Jo-­1+ patients with IIM/antisynthetase syndrome or patients with sarcoidosis; gray-­shaded symbols represent the median fold
change in anti–Jo-­1− patients. Categories of fold change were low (<3-­fold), intermediate (3–10-­fold), or high (>10-­fold). D, IFNγ production
on CD40L+CD4+ T cells was compared between BAL fluid cells and corresponding PBMCs from anti–Jo-­1+ patients with IIM/antisynthetase
syndrome after stimulation with full-­length HisRS recombinant protein or HisRS11–23. Solid symbols represent anti–Jo-­1+ patients and gray-­
shaded symbols represent anti–Jo-­1− patients; horizontal lines show the median. * = P < 0.05; *** = P < 0.001, by 2-­tailed Mann-Whitney
U test. See Figure 1 for definitions.

patients with IIM/antisynthetase syndrome (patients P1–P3) and 2 CD4+CD40L+ T cells from BAL fluid produced IFNγ in response to
anti–Jo-­1− patients (patients P4 and P5). Almost all of the CD4+ T antigenic stimulation with either HisRS full-­length protein or HisRS11–23,
cells in BAL fluid were positive for the Th1-­associated chemokine compared to 10% of the CD4+CD40L+ T cells in the corresponding
receptors CXCR3 (97%) (Figure 4A) and CCR5 (93%) (Figure 4B), blood samples (P < 0.001 and P < 0.05, respectively) (Figure 3D).
compared to 46% and 50% of PBMCs, respectively (P < 0.01 and In the disease control group of patients with sarcoidosis, BAL
P < 0.05, respectively). Moreover, ~60% of the BAL fluid T cells fluid cells from 2 of 7 patients with sarcoidosis (patients Sarc1 and
expressed CCR6, compared to 20% of PBMCs (P < 0.01) (Fig- Sarc4) displayed a high (>10-­fold) CD40L response to the HisRS
ure 4C). In BAL fluid, >90% of CCR6+CD4+ T cells were positive protein, with a median fold change of 20 (IQR 15.75–25.5) relative
for CXCR3, indicating that CD4+ T cells in BAL fluid had a Th1/ to unstimulated cells. Two other control patients with sarcoidosis (of
Th17 phenotype in anti–Jo-­1+ and anti–Jo-­1− patients. the 7 tested) presented high CD40L up-­regulation in response to
Furthermore, HisRS-­specific BAL fluid CD4+CD40L+ T cells HisRS11–23 (patients Sarc3 and Sarc7), with a median fold change
presented a pronounced proinflammatory Th1 phenotype that of 59 (IQR 22.7–95) relative to unstimulated conditions (Figure 3C).
was characterized by an increased production of IFNγ, when com- CD40L up-­regulation in BAL fluid cells from the group of patients with
pared to the corresponding CD4+CD40L+ T cells from PBMCs, in sarcoidosis who responded to HisRS full-­length protein was 4 times
both anti–Jo-­1+ and anti–Jo-­1− patients. On average, 60% of the higher compared to that in the corresponding PBMCs (Figure 1D
ANTIGEN-­SPECIFIC T CELLS IN INFLAMMATORY MYOPATHIES | 187

Figure 4. Th1 cells and anti–Jo-­1 antibodies are present in the lungs of patients with IIM/antisynthetase syndrome. A–C, Representative plots
from fluorescence-­activated cell sorter analysis (bottom) and quantification of the results (top) show the percentage of CD4+CD3+CXCR3+ T cells
(A), CD4+CD3+CCR5+ T cells (B), and CD4+CD3+CXCR3+CCR6+ T cells (C) in PBMCs and bronchoalveolar lavage (BAL) fluid (BALF) cells from
patients with IIM/antisynthetase syndrome (n = 5). D and E, Presence of anti–Jo-­1 autoantibodies was analyzed by enzyme-­linked immunosorbent
assay (ELISA) in the BAL fluid (D) and corresponding serum (E) of patients with IIM/antisynthetase syndrome, patients with sarcoidosis, and healthy
controls. The broken horizontal line indicates the cutoff value for positivity, calculated using the values in healthy controls (mean absorbance in healthy
controls + 3SD). Results are expressed as the normalized (Norm.) values for the absorbance at 405 nm (optical density ratio of anti–Jo-­1 IgG to total
IgG in the BAL fluid and serum). Symbols in A–E show individual subjects; horizontal lines show the mean. F, Presence of anti–Jo-­1 IgG autoantibodies
was compared between the BAL fluid and serum of anti–Jo-­1+ (n = 6) and anti–Jo-­1− (n = 7) patients with IIM/antisynthetase syndrome, assessed by
ELISA with values normalized to the total IgG levels in corresponding compartments. Patients were classified as anti–Jo-­1+ if they were seropositive
by at least one method (ELISA, immunoprecipitation, or line blotting, as described in Table 1). The broken horizontal line indicates the cutoff value for
seropositivity. * = P < 0.05; ** = P < 0.01, by 2-­tailed Mann-Whitney U test. See Figure 1 for definitions.

and Figure 3C). The frequencies of CD4+CD40L+ T cells in patients tibodies in BAL fluid from 6 of 7 anti–Jo-­1+ patients and in
with IIM/antisynthetase syndrome, control patients with sarcoidosis, 2 of 9 patients with sarcoidosis, but not in healthy controls or in
and healthy controls are presented in Supplementary Table 8 (http:// anti–Jo-­1− individuals (Figure 4D). After normalizing the amount
onlin​elibr​ary.wiley.com/doi/10.1002/art.41075/​abstract). of anti–Jo-­1 autoantibodies to the values for total IgG, all of the
anti–Jo-­1+ patients who showed reactivity in the BAL fluid were
Anti–Jo-­1 autoantibodies in BAL fluid. To further dis- also reactive in the serum (Figures 4E and F).
sect the HisRS-­specific immune response in the lung compart-
ment, cell-­depleted BAL fluid samples from patients with IIM/ Infiltration of lung tissue by T cells and plasma cells
antisynthetase syndrome (n = 13), corresponding to group 1 in patients with IIM/antisynthetase syndrome. Germinal
and group 3 (patients P1–P5 in group 1, and patients P8 and center (GC)–like structures characterized by T cells and sur-
P9 and P19–P24 in group 3), patients with sarcoidosis (n = 9), rounding plasma cells were observed in the lung tissue from 2
and healthy controls (n = 18) were analyzed for the presence of of 4 anti–Jo-­1+ patients with IIM/antisynthetase s­ yndrome but
anti–Jo-­1 autoantibodies. We detected anti–Jo-­1 IgG autoan- in none of the lung tissue samples from anti–Jo-­1− patients
188 | GALINDO-­FERIA ET AL

Figure 5. Histologic examination of the lungs of a representative anti–Jo-­1+ patient with idiopathic inflammatory myopathy/antisynthetase
syndrome and interstitial lung disease. A, Immunohistochemical analysis of the lung tissue sections reveals T cell infiltrates (CD3 staining) (upper
panels) and infiltration of plasmablasts (CD138 staining) (lower panels). Boxed areas in the left panels are shown at higher magnification on
the right. Original magnification × 10 on left; × 25 on right. B, Immunohistochemical staining of the lung tissue shows several germinal center–
like formations on CD3 staining (left and upper right panels) and CD138 staining (lower right panel). Boxed area in the left panel is shown at
higher magnification on the upper right. Original magnification × 5 on left; × 10 on right. Isotype controls for CD3, CCR5, and CXCR3 with
semiquantitative analysis are presented in Supplementary Figure 2 (http://onlinelibrary.wiley.com/doi/10.1002/art.41075/abstract).

or patients with COPD (Figures 5A and B). In addition, tary Figures 2A and C, available at http://onlinelibrary.wiley.com/
we observed infiltrating CD3+ T cells in all patient groups doi/10.1002/art.41075/abstract). There was no difference in the
(patients with IIM/antisynthetase syndrome and patients with expression of CXCR3 and CCR5 between anti–Jo-­1+ and anti–
COPD), with a higher number of infiltrating CD3+ T cells in Jo-­1− patients (staining not performed in lung biopsy samples
patients with COPD than in patients with IIM/antisynthetase syn- from patients with COPD). Transbronchial lung biopsy samples
drome (P = 0.009). No differences between groups were seen from patients with sarcoidosis did not show the presence of GC
for CD138+ plasma cell expression (P = 0.37) (see Supplemen- formation (see Supplementary Table 4).
ANTIGEN-­SPECIFIC T CELLS IN INFLAMMATORY MYOPATHIES | 189

DISCUSSION gene–environment interaction. In this context, the previously


described higher expression of HisRS in lung tissue compared
In this study, we describe activation of antigen-­specific CD4+ to other types of tissue is interesting (20). Also, a granzyme
T cells in peripheral blood and BAL fluid cells from patients with B–sensitive form of HisRS has been identified exclusively in the
IIM/antisynthetase syndrome by an HLA–DRB1*03-­associated lung (21). Therefore, increased amounts of granzyme B triggered
T cell epitope corresponding to the stretch of residues 11–23 by proinflammatory environmental stimuli, such as infections or
within the HisRS protein. The stimulation of CD4+ T cells with the smoke exposure (35), may contribute to the release of N-­terminal
full-­length HisRS protein and the HisRS11–23 peptide induced up-­ HisRS fragments and the formation and enhanced processing of
regulation of CD40L in CD4+ T cells in both the lung and blood neoepitopes that can be recognized by HisRS-reactive T cells in
compartments, indicating the presence of HisRS-­specific T cells patients with IIM/antisynthetase syndrome (21,36).
in patients with IIM/antisynthetase syndrome. The N-­ terminal HisRS fragments have chemotactic
To our knowledge, the phenotype of T cells involved in properties that can induce migration of CCR5-­ expressing
the immune reactivity toward a specific peptide in IIM/antisyn- lymphocytes, activated monocytes, and immature dendritic
thetase syndrome has not been previously addressed. The cells (23). The aforementioned mechanism suggests that
present description of T cell reactivity is directed against a the HisRS protein or fragments released from damaged
candidate HisRS-­derived peptide (HisRS11–23) that we selected lungs may be involved in the recruitment of activated T cells,
based on binding predictions to HLA–DRB1*03:01. The high including HisRS-­activated cells to inflammatory sites in the
reactivity observed in patients with IIM/antisynthetase syn- lungs, thereby providing costimulatory signals to B cells and
drome compared to patients with sarcoidosis and healthy antigen-­presenting cells. In this hypothesis, the muscle might
controls indicates that the HisRS11–23 epitope is an important become targeted in later stages of the disease (20).
target for autoreactive T cells in IIM/antisynthetase syndrome. Consistent with previous reports (37–40), the present study
Furthermore, our results demonstrate that the activation of demonstrated high expression of the chemokine receptors CCR5
HisRS-­specific T cells was abrogated by blocking HLA–DR and CXCR3 (markers of Th1 cells) and CCR6 (Th17 marker) in
but not by blocking HLA–DQ. We thus demonstrate a T cell nearly 100% of CD4+ T cells isolated from the BAL fluid of both
activation bias in HLA–DRB1*03:01–positive patients toward anti–Jo-­1+ and anti–Jo-­ 1− patients. The enriched expression
HisRS11–23. These findings confirm previous results demon- of these chemokine receptors in BAL fluid T cells compared to
strating T cell proliferation following stimulation with substan- peripheral blood T cells was not unexpected, as several studies
tially larger fragments of HisRS in T cell cultures (22). have shown that these receptors are known to regulate antigen-­
Nevertheless, the observed T cell activation was not entirely induced T cell homing in lung diseases, such as asthma (41,42),
limited to the anti–Jo-­ 1+ subset of patients with IIM/antisyn- COPD (43), and sarcoidosis (44), and can even be found in
thetase syndrome, but occurred in some anti–Jo-­1− patients, healthy smokers (45). Moreover, the identification of a Th1/Th17
although to a substantially lower degree. While the genetic asso- phenotype (Th17.1 cells) in BAL fluid from patients with IIM/anti-
ciation between IIM with anti–Jo-­1 antibody positivity and HLA– synthetase syndrome, characterized by an excessive secretion of
DRB1*0301 is well established, we find it interesting that patients IFNγ, has also been reported in patients with chronic sarcoidosis
with other class II major histocompatibility complex alleles could (38,46,47) and in other autoimmune diseases such as Crohn’s
also be implicated in the presentation of HisRS-­derived peptides. disease (48) and arthritis (49).
Our findings of an increased reactivity against the HisRS The design of the present study has a number of limitations.
protein, and in particular the HisRS-­derived peptide HisRS11–23 in One limitation was the low availability of paired samples from
lung-­derived T cells compared to blood-­derived cells, are, in our BAL fluid and PBMCs from untreated patients with inflammatory
opinion, striking, and despite the limited number of cases with BAL active IIM/antisynthetase syndrome. Acquiring BAL fluid sam-
fluid–derived T cells, this observation may point to the lung as a ples requires an invasive procedure, and it may be unethical to
potential site of primary activation of T cells against HisRS, similar delay the start of treatment until the bronchoscopy can be per-
to what has been proposed in other autoimmune diseases such formed. Nevertheless, the results obtained from this small num-
as RA (8,9,34) and MS (12). We also identified the presence of ber of patients were consistent, contributing to the significance
anti–Jo-­1 autoantibodies in BAL fluid as well as GC-­like structures of our observations.
in lung tissue from anti–Jo-­1+ patients, supporting the hypothesis In conclusion, the results of the present study indicate that
of the lungs as a potential site for immune activation and anti–Jo-­1 specific HisRS-­derived peptides in HLA–DRB1*03–positive indi-
autoantibody production. As some patients displayed reactivity to viduals, including the HisRS11–23 peptide investigated herein, may
the HisRS full-­length protein but not to the HisRS11–23 epitope, be presented to locally available T cells, which become activated
other HisRS-­derived peptides are likely to be implicated. and subsequently adopt a proinflammatory phenotype. These
Interestingly, 2 patients with sarcoidosis with the highest T cells may promote B cell maturation and activation, GC-­like
HisRS T cell reactivity were former smokers, indicating a possible structure formation, and subsequent production of anti–Jo-­
­ 1
190 | GALINDO-­FERIA ET AL

autoantibodies. One possible site for the initial activation of the 5. Aggarwal R, Cassidy E, Fertig N, Koontz DC, Lucas M, Ascherman
immune system could be in the lungs, where environmental DP, et al. Patients with non-­Jo-­1 anti-­tRNA-­synthetase autoantibod-
ies have worse survival than Jo-­1 positive patients. Ann Rheum Dis
factors such as smoking or infections may cause an unspecific 2014;73:227–32.
inflammation in the lung tissue, leading to granzyme B release 6. Hervier B, Devilliers H, Stanciu R, Meyer A, Uzunhan Y, Meau A,
and HisRS cleavage that may then lead to the recruitment of T et al. Hierarchical cluster and survival analyses of antisynthetase
cells. Further studies on other relevant peptides with a broader syndrome: phenotype and outcome are correlated with anti-­tRNA
synthetase antibody specificity. Autoimmun Rev 2012;12:210–7.
coverage of the antigenic sites of HisRS, as well as studies on
7. Connors GR, Christopher-Stine L, Oddis CV, Danoff SK. Interstitial
the phenotypes of locally infiltrating T cells, in patients with lung disease associated with the idiopathic inflammatory myopa-
IIM/antisynthetase syndrome remain a priority. thies: what progress has been made in the past 35 years? Chest
2010;138:1464–74.
8. Reynisdottir G, Karimi R, Joshua V, Olsen H, Hensvold AH, Harju A,
ACKNOWLEDGMENTS et al. Structural changes and antibody enrichment in the lungs are
early features of anti–citrullinated protein antibody–positive rheuma-
We thank Eva Lindroos and Yvonne Sundström for their toid arthritis. Arthritis Rheumatol 2014;66:31–9.
excellent technical assistance, as well as nurse Christina 9. Catrina AI, Ytterberg AJ, Reynisdottir G, Malmström V, Klareskog L.
Lungs, joints and immunity against citrullinated proteins in rheuma-
Ottosson for collecting blood samples, Julia Norkko and Gloria
toid arthritis. Nat Rev Rheumatol 2014;10:645–53.
Rostvall for handling serum samples, and Benita Dahlberg for 10. Willis VC, Demoruelle MK, Derber LA, Chartier-Logan CJ, Parish
handling BAL fluid samples. We also thank Barbro Larsson and MC, Pedraza IF, et al. Sputum autoantibodies in patients with es-
Leonid Padyukov for the genotyping of the samples, as well tablished rheumatoid arthritis and subjects at risk of future clinically
apparent disease. Arthritis Rheum 2013;65:2545–54.
as Karine Chemin for assistance with the T cell assays, and
11. Kinloch AJ, Kaiser Y, Wolfgeher D, Ai J, Eklund A, Clark MR, et al.
Karolina Tandre for providing blood samples from healthy In situ humoral immunity to vimentin in HLA-­DRB1*03+ patients with
controls. Finally, we thank Associate Professor Inger Nennesmo pulmonary sarcoidosis. Front Immunol 2018;9:1516.
for providing the muscle pathology reports, and Karine 12. Hedström AK, Sundqvist E, Bäärnhielm M, Nordin N, Hillert J, ­Kockum
Chemin, Valérie Leclair, and Lars Klareskog for constructive I, et al. Smoking and two human leukocyte antigen genes interact to
increase the risk for multiple sclerosis. Brain 2011;134:653–64.
criticism of the manuscript.
13. O’Hanlon TP, Carrick DM, Targoff IN, Arnett FC, Reveille J, Carrington
M, et al. Immunogenetic risk and protective factors for the idiopathic
AUTHOR CONTRIBUTIONS inflammatory myopathies: distinct HLA-­A, -­B, -­Cw, -­DRB1, and -­DQA1
allelic profiles distinguish European American patients with different
All authors were involved in drafting the article or revising it criti-
myositis autoantibodies. Medicine (Baltimore) 2006;85:111–27.
cally for important intellectual content, and all authors approved the final
­version to be published. Dr. Galindo-­Feria had full access to all of the 14. Chinoy H, Adimulam S, Marriage F, New P, Vincze M, Zilahi E, et al.
data in the study and takes responsibility for the integrity of the data and Interaction of HLA-­DRB1*03 and smoking for the development of
the accuracy of the data analysis. anti-­Jo-­1 antibodies in adult idiopathic inflammatory myopathies: a
Study conception and design. Galindo-­ Feria, Albrecht, Fernandes-­ European-­wide case study. Ann Rheum Dis 2012;71:961–5.
Cerqueira, Notarnicola, James, Dastmalchi, Rönnblom, Jakobsson, Fathi, 15. Rothwell S, Chinoy H, Lamb JA, Miller FW, Rider LG, Wedderburn
Grunewald, Malmström, Lundberg. LR, et al. Focused HLA analysis in Caucasians with myositis iden-
Acquisition of data. Galindo-­ Feria, Albrecht, Fernandes-­ Cerqueira, tifies significant associations with autoantibody subgroups. Ann
Notarnicola, Herrath, Dastmalchi, Fathi, Lundberg. Rheum Dis 2019;78:996–1002.
Analysis and interpretation of data. Galindo-­Feria, Albrecht, Fernandes-­ 16. Goldstein R, Duvic M, Targoff IN, Reichlin M, McMenemy AM,
Cerqueira, Notarnicola, James, Dastmalchi, Sandalova, Fathi, Achour, ­Reveille JD, et al. HLA–D region genes associated with autoanti-
Grunewald, Malmström, Lundberg. body responses to histidyl–transfer RNA synthetase (Jo-­1) and o ­ ther
translation-­related factors in myositis. Arthritis Rheum 1990;33:
ADDITIONAL DISCLOSURES 1240–8.
17. Miller FW, Waite KA, Biswas T, Plotz PH. The role of an autoantigen,
Author Albrecht is currently an employee of Sanofi Genzyme. Author histidyl-­tRNA-­synthetase, in the induction and maintenance of auto-
Herrath is currently an employee of Pfizer. immunity. Proc Natl Acad Sci U S A 1990;87:9933–7.
18. Ascherman DP. Role of Jo-­1 in the immunopathogenesis of the anti-­
REFERENCES synthetase syndrome. Curr Rheumatol Rep 2015;17:56.
19. Miller FW, Twitty SA, Biswas T, Plotz PH. Origin and regulation of a
1. Marie I. Morbidity and mortality in adult polymyositis and dermato- disease-­specific autoantibody response: antigenic epitopes, spec-
myositis. Curr Rheumatol Rep 2012;14:275–85. trotype stability, and isotype restriction of anti-­Jo-­1 autoantibodies.
2. McHugh NJ, Tansley SL. Autoantibodies in myositis. Nat Rev Rheu- J Clin Invest 1990;85:468–75.
matol 2018;14:290–302. 20. Casciola-Rosen L, Nagaraju K, Plotz P, Wang K, Levine S,
3. Betteridge Z, Tansley S, Shaddick G, Chinoy H, Cooper RG, New ­Gabrielson E, et al. Enhanced autoantigen expression in regener-
RP, et al. Frequency, mutual exclusivity and clinical associations of ating muscle cells in idiopathic inflammatory myopathy. J Exp Med
myositis autoantibodies in a combined European cohort of idiopathic 2005;201:591–601.
inflammatory myopathy patients. J Autoimmun 2019;101:48–55. 21. Levine SM, Raben N, Xie D, Askin FB, Tuder R, Mullins M, et al.
4. Brouwer R, Hengstman GJ, Vree Egberts W, Ehrfeld H, Bozic B, Novel conformation of histidyl–transfer RNA synthetase in the lung:
Ghirardello A, et al. Autoantibody profiles in the sera of European the target tissue in Jo-­1 autoantibody–associated myositis. Arthritis
patients with myositis. Ann Rheum Dis 2001;60:116–23. Rheum 2007;56:2729–39.
ANTIGEN-­SPECIFIC T CELLS IN INFLAMMATORY MYOPATHIES | 191

22. Ascherman DP, Oriss TB, Oddis CV, Wright TM. Critical requirement 36. Casciola-Rosen L, Andrade F, Ulanet D, Wong WB, Rosen A.
for professional APCs in eliciting T cell responses to novel fragments Cleavage by granzyme B is strongly predictive of autoantigen
of histidyl-­tRNA synthetase (Jo-­1) in Jo-­1 antibody-­positive poly­ status: implications for initiation of autoimmunity. J Exp Med
myositis. J Immunol 2002;169:7127–34. 1999;190:815–26.
23. Howard OM, Dong HF, Yang D, Raben N, Nagaraju K, Rosen A, et al. 37. Shimizu S, Yoshinouchi T, Niimi T, Ohtsuki Y, Fujita J, Maeda H, et al.
Histidyl-­tRNA synthetase and asparaginyl-­tRNA synthetase, autoan- Differing distributions of CXCR37-­and CCR37-­positive cells among
tigens in myositis, activate chemokine receptors on T lymphocytes types of interstitial pneumonia associated with collagen vascular dis-
and immature dendritic cells. J Exp Med 2002;196:781–91. eases. Virchows Arch 2007;450:51–8.
24. Chino Y, Murata H, Goto D, Matsumoto I, Tsutsumi A, Sakamoto T, 38. Ramstein J, Broos CE, Simpson LJ, Ansel KM, Sun SA, Ho ME,
et al. T cell receptor BV gene repertoire of lymphocytes in broncho- et al. IFN-­γ-­producing T-­helper 17.1 cells are increased in s­ arcoidosis
alveolar lavage fluid of polymyositis/dermatomyositis patients with and are more prevalent than T-­helper type 1 cells. Am J Respir Crit
interstitial pneumonitis. Int J Mol Med 2006;17:101–9. Care Med 2016;193:1281–91.
25. Englund P, Wahlström J, Fathi M, Rasmussen E, Grunewald J, 39. Acosta-Rodriguez EV, Rivino L, Geginat J, Jarrossay D, Gattorno M,
­Tornling G, et al. Restricted T cell receptor BV gene usage in the Lanzavecchia A, et al. Surface phenotype and antigenic specificity of
lungs and muscles of patients with idiopathic inflammatory myopa- human interleukin 17-­producing T helper memory cells. Nat Immunol
thies. Arthritis Rheum 2007;56:372–83. 2007;8:639–46.
26. Bohan A, Peter JB. Polymyositis and dermatomyositis (first of two 40. Sallusto F, Lanzavecchia A, Mackay CR. Chemokines and chemo­
parts). N Engl J Med 1975;292:344–7. kine receptors in T-­cell priming and Th1/Th2-­mediated responses.
27. Bohan A, Peter JB. Polymyositis and dermatomyositis (second of Immunol Today 1998;19:568–74.
two parts). N Engl J Med 1975;292:403–7. 41. Campbell JJ, Brightling CE, Symon FA, Qin S, Murphy KE, Hodge M,
28. American Thoracic Society, European Respiratory Society. Amer- et al. Expression of chemokine receptors by lung T cells from normal
ican Thoracic Society/European Respiratory Society International and asthmatic subjects. J Immunol 2001;166:2842–8.
multidisciplinary consensus classification of the idiopathic interstitial 42. Thomas SY, Banerji A, Medoff BD, Lilly CM, Luster AD. Multiple
pneumonias. Am J Respir Crit Care Med 2002;165:277–304. chemokine receptors, including CCR42 and CXCR42, regulate
29. Karimi R, Tornling G, Grunewald J, Eklund A, Skold CM. Cell re- antigen-­induced T cell homing to the human asthmatic airway. J Im-
covery in bronchoalveolar lavage fluid in smokers is dependent on munol 2007;179:1901–12.
cumulative smoking history. PLoS One 2012;7:e34232. 43. Forsslund H, Yang M, Mikko M, Karimi R, Nyrén S, Engvall B, et al.
30. Snir O, Bäcklund J, Boström J, Andersson I, Kihlberg J, Buckner JH, Gender differences in the T-­cell profiles of the airways in COPD
et al. Multifunctional T cell reactivity with native and glycosylated type patients associated with clinical phenotypes. Int J Chron Obstruct
II collagen in rheumatoid arthritis. Arthritis Rheum 2012;64:2482–8. Pulmon Dis 2016;12:35–48.
31. Ghosh P, Amaya M, Mellins E, Wiley DC. The structure of an interme- 44. Kaiser Y, Lakshmikanth T, Chen Y, Mikes J, Eklund A, Brodin P, et al.
diate in class II MHC maturation: CLIP bound to HLA-­DR3. Nature Mass cytometry identifies distinct lung CD4+ T cell patterns in Löf-
1995;378:457–62. gren’s syndrome and non-­ Löfgren’s syndrome sarcoidosis. Front
32. James EA, Moustakas AK, Bui J, Nouv R, Papadopoulos GK, Immunol 2017;8:1130.
Kwok WW. The binding of antigenic peptides to HLA-­DR is influ- 45. Brozyna S, Ahern J, Hodge G, Nairn J, Holmes M, Reynolds PN,
enced by interactions between pocket 6 and pocket 9. J Immunol et al. Chemotactic mediators of Th1 T-­cell trafficking in smokers and
2009;183:3249–58. COPD patients. COPD 2009;6:4–16.
33. Zickert A, Oke V, Parodis I, Svenungsson E, Sundström Y, 46. Broos CE, Koth LL, van Nimwegen M, In ’t Veen JC, Paulissen SM,
­Gunnarsson I. Interferon (IFN)-­λ is a potential mediator in lupus ne- van Hamburg JP, et al. Increased T-­helper 17.1 cells in sarcoidosis
phritis. Lupus Sci Med 2016;3:e000170. mediastinal lymph nodes. Eur Respir J 2018;51:1701124.
34. Klareskog L, Stolt P, Lundberg K, Källberg H, Bengtsson C, 47. Kaiser Y, Eklund A, Grunewald J. Moving target: shifting the focus
Grunewald J, et al, and the Epidemiological Investigation of Rheuma- to pulmonary sarcoidosis as an autoimmune spectrum disorder. Eur
toid Arthritis Study Group. A new model for an etiology of rheumatoid Respir J 2019;54:1802153.
arthritis: smoking may trigger HLA–DR (shared epitope)–restricted 48. Annunziato F, Cosmi L, Santarlasci V, Maggi L, Liotta F, Mazzinghi B,
immune reactions to autoantigens modified by citrullination. Arthritis et al. Phenotypic and functional features of human Th17 cells. J Exp
Rheum 2006;54:38–46. Med 2007;204:1849–61.
35. Hodge S, Hodge G, Nairn J, Holmes M, Reynolds PN. Increased 49. Nistala K, Adams S, Cambrook H, Ursu S, Olivito B, de Jager W, et al.
airway granzyme B and perforin in current and ex-­smoking COPD Th17 plasticity in human autoimmune arthritis is driven by the inflam-
subjects. COPD 2006;3:179–87. matory environment. Proc Natl Acad Sci U S A 2010;107:14751–6.

You might also like