• Aucun résultat trouvé

Idiopathic inflammatory myopathies: from immunopathogenesis to new therapeutic targets

N/A
N/A
Protected

Academic year: 2021

Partager "Idiopathic inflammatory myopathies: from immunopathogenesis to new therapeutic targets"

Copied!
8
0
0

Texte intégral

(1)

INVITED REVIEW

Idiopathic inflammatory myopathies: from

immunopathogenesis to new therapeutic targets

Syed A. HAQ

1

and Anne TOURNADRE

2

1BSM Medical University, Dhaka, Bangladesh, and2Rheumatology department CHU Clermont-Ferrand and UMR 1019 INRA/

University of Auvergne, Clermont-Ferrand, France

Abstract

Pathogenesis of idiopathic inflammatory myositis (IIM) involves strong interactions between dendritic cells (DCs), activated Th1 and Th17 cells, B cells, muscle cells, genes and environment. Local maturation of DCs per-mit the activation and polarization of CD4+ T cells into TH1 and TH17 that play a key role in maintaining

chronic muscle inflammation. T-cell mediated myocytotoxicity promotes the liberation of specific muscle autoantigens from regenerating muscle cells with production of myositis-specific autoantibodies. Type I inter-feron signature is a key characteristic of IIM. Type I IFN that can be induced by immune complexes containing myositis-specific autoantibodies is produced by scattered plasmacytoid DCs but also by muscle cells particularly regenerating muscle cells. These immature muscle precursors appear to be critical in the pathogenesis of IIM as they up-regulate muscle autoantigens, type I IFN, HLA class I antigens and TLR3-7, all together involved in maintaining chronic muscle inflammation. In addition to the role of immune and muscle cells, genome-wide association studies have confirmed the importance of several MHC and non-MHC genes in IIM. Environmental factors can contribute to the pathogenesis of IIM. In sIBM, distinct features suggest both degenerative and inflammatory processes. In addition to our better understanding of the pathogenesis, identify molecular path-way leads to consider new targeted therapies including cytokine inhibition, B-cell and T-cell costimulation blockade, type I IFN neutralization or inhibition of the ubiquitin proteasome pathway.

Key words: dermatomyositis, idiopathic inflammatory myositis, inclusion body myositis, pathogenesis, polymyositis, treatment.

INTRODUCTION

Idiopathic inflammatory myositis (IIM) is a heteroge-neous group of autoimmune myopathies. It comprises at least five conditions, namely, polymyositis (PM), der-matomyositis (DM), juvenile derder-matomyositis (JDM), sporadic inclusion body myositis (sIBM) and immune-mediated necrotizing myopathy (IMNM). In addition, new classifications distinguish overlap myositis and cancer-associated myositis.1,2However, these classifica-tions are limited by clinical, histological and serological

overlap. Not much was known about the etiopathogen-esis of this condition. The evidence for an immune-me-diated disease comes from the presence of cellular infiltrates in muscle biopsies, a T cell-mediated myocy-totoxicity, autoantibodies in the peripheral blood and association with class I major histocompatibility (MHC) overexpression. On the basis of these under-standings, so far, IIMs have been treated with glucocor-ticoids and other conventional immunosuppressive agents, including methotrexate and azathioprine. The treatment is unsuccessful in some cases and is often fraught with the consequences of non-specific immuno-suppression. Therapies targeting specific cytokines, receptors and intracellular molecules have shown pro-mise in the treatment of other rheumatic conditions, like rheumatoid arthritis (RA), ankylosing spondylitis

Correspondence: Dr Anne Tournadre, Rheumatology

Department, Gabriel Montpied Hospital, 58 rue Montalembert BP69 63003 Clermont-Ferrand cedex 1, France

(2)

(AS) and systemic lupus erythematosus (SLE). It has become imperative that the pathogenesis of IIMs be explored in detail so that more specific and targeted therapies that are likely to be more effective and safe may be developed. In this communication, keeping the sequence of events in consideration, the roles of den-dritic cells, lymphocytes and cytokines will be presented with transcriptional regulation. Critical contribution of regenerating muscle cells in pathophysiology will be discussed as well as the role of genetics and environ-ment. Finally, we will address potential therapeutic tar-gets.

DENDRITIC CELLS, LYMPHOCYTES AND

CYTOKINES

Dendritic cells (DCs) are professional antigen-present-ing cells that support adaptive and innate immune responses. Their activation in the lymphoid and periph-eral tissues leads to their migration, maturation and to the activation of CD4+ and CD8+T cells as well as B cells. It appears from histological studies that CD4+ T cells and B cells play a predominant role in the patho-genesis of DM, whereas this CD8+T cells in the patho-genesis of PM. Immature CD1a+ and mature LAMP3+ (lysosome-associated membrane glycoprotein 3) den-dritic cells are found in the inflammatory infiltrates from endomysial areas in PM and perimysial areas in DM.3DCs are of two types: myeloid (or conventional) DCs (mDCs or cDCs) supporting specific adaptive immune response and plasmacytoid DCs (pDCs) which produce type I IFN (interferon) and have a key role in innate immunity. pDCs are found mainly in DM.4

mDCs predominate in PM and sIBM.5The local matu-ration of DCs is suggested by the presence of CCL20 involved in the migration of immature DCs with the lack of the C–C motif chemokines CCL19 and CCL21 involved in the migration of mature DCs.6Activation of toll-like receptors (TLR) and C-type lectin receptors (CLR) on the surface of immature DCs by pathogen-as-sociated molecular patterns (PAMPs) and muscle auto-antigens could favor their maturation.7,8Once mature, mDCs can activate CD4+T cells. An inverse correlation of peripheral blood T cell count with disease activity and limited pattern expression of T cell receptors (TCR) in muscle tissue hint at homing of T cells during active disease with local proliferation and possibly presence of a pathogenic antigen in the muscle tissue.9,10

Activated CD4+T cells develop into distinct types of T helper (TH) cells that can be identified according to

the types of cytokines T cells produce when they are

stimulated to differentiate. TH1 cells produce large

amount of IFN-c, induce delayed hypersensitivity reac-tions, target macrophages, and are essential for the defense against intracellular pathogens. TH2 cells

mainly produce interleukin 4 (IL4) and contribute to immunoglobulin E (IgE) production and eosinophil recruitment to clear parasites. TH17 cells preferentially

produce IL17, but not IFN-c as TH1 cells, or IL-4 as TH2

cells. The TH17 pathway plays a critical role in inducing

and maintaining chronic inflammation and autoimmu-nity. TH1 cells develop in presence of IL12 and IL18.

Differentiation of human TH17 cells depends on key

cytokines, namely IL-6, IL-23 and transforming growth factor (TGF)-b. All these cytokines are locally produced by macrophages, monocytes, endothelial cells and DCs. IL18 is over-expressed in endomysial areas in PM and in the perivascular areas in DM.11TH1 cells secrete

IFN-c. In addition to enhancing natural killer (NK) cell activity, antigen processing and presentation by antigen presenting cells (APCs), IFN-c increases expression of MHC class I and TLR3 on myoblasts.7,8 The role of muscle cell, human leukocyte antigen (HLA) class I and TLR3 is elaborated in the next section. TH17 cells

elabo-rate IL17. In IIMs, IL17 is detected by immunohisto-chemistry in muscle lymphocytic infiltrates.3 In combination with IL1, it induces production of IL6 and CCL20 by myoblasts.12 The IL-23–TH17 pathway is

important for maintaining chronic tissue inflammation. Locally IL17 amplifies the initial response by acting in synergy with pro-inflammatory cytokines produced by macrophages and DCs to promote the production of the b chemokine CCL20, adhesion molecules and IL6 that facilitate in turn migration of mononuclear cells to the endomysial and perimysial spaces, maturation of DCs and differentiation of T cells.13

Mature mDCs also activate CD8+cells by presenting antigen in the groove of HLA class I antigens. It leads to activation, proliferation and differentiation of cytotoxic T cells. In PM, endomysial CD8+T cells surround and invade muscle fibers that express MHC class I anti-gens.14IL15 and IL15 receptors upregulated in muscle tissue of IIMs may be responsible for local T cell activa-tion and expansion.15It is assumed that an interaction between myocytotoxic CD8+ T cells and regenerating muscle cells expressing MHC class I antigen leads to lib-eration of cytotoxic effector molecules, including gran-zyme B. Granzyme B generates myositis-specific autoantigens after cleavage of autoantigenic peptides such as histidyl-tRNA (transfer RNA) synthetases and exposition of new dominant epitopes.16 In addition to antibody response initiation, autoantigenic tRNA

(3)

synthetases are chemotactic for lymphocytes and imma-ture DCs17 leading to a positive feedback loop for maintaining the inflammation.

In PM and DM, a large proportion of T cells are of CD28nullphenotype.18 They are thought to arise from repeated antigenic stimulation, and are terminally dif-ferentiated and apoptosis resistant. Both CD4+CD28null and CD8+CD28null T cells contain granzyme and perforin, having NK/cytotoxic properties and are easily stimulated to produce cytokines.19 In autologous muscle T cell co-culture both types of CD28null cells were found to be highly toxic to mytubes.20

mDCs play a key role in humoral activity with activa-tion of B cells and producactiva-tion of autoantibodies. B cells and plasma cells in muscle tissue, large numbers of B cells and auto-antibodies in peripheral blood, high serum B cell activating factor (BAFF) also known as B lymphocyte stimulator (BLyS), evidences of in situ dif-ferentiation of B cells, including somatic mutation, iso-type switching, clonal expansion and intraclonal variation, hint at the role of these cells in the pathogen-esis.21The majority of patients with IIMs have at least one myositis specific autoantibody (MSA), provided sensitive techniques are used.22

Scattered pDCs are found in the infiltrates in IIMs.23 There are evidences that immune complexes containing anti–Jo-1 or anti–Ro 52/anti–Ro 60 autoantibodies and RNA may activate IFNa production by pDCs.23 IFNa

activates specific transcription activators including STATA. It stimulates transcription of genes responsible for synthesis of a series of pro-inflammatory cytokines, particularly those driving TH1 and TH17 cell lines. It

also enhances expression of HLA class I and class II molecules. The expression of type I IFN inducible genes and their protein products are upregulated in blood and in muscle tissue in IIMs (type 1 IFN signature).24 Beside the endogenous IFNa production by pDCs, immature muscle precursors may act as a local source of IFNb.7

TRANSCRIPTIONAL REGULATION

Micro-RNAs (miRNAs) are key components for post-transcriptional regulation. They function via base-pair-ing with complementary sequences within mRNA molecules. As a result, these mRNA strands are silenced because they can no longer be translated into proteins by ribosomes, and such complexes are often actively disassembled by the cell (“target degradation”).25It is plausible that certain miRNAs may modulate

inflamma-tion through silencing mRNAs coding for pro-inflam-matory cytokines. In IIM, a decreased expression of such miRNAs (miR-1, miR-133a, miR-133b) has been reported and miRNAs and could influence the NFjB pathway.26,27

The signal transducer and activator of transcription 4 (STAT4) is activated by IL-1, IL-23, IFN-a and IFN-b. It induces transcription of genes that drive the TH1 and

TH17 responses. Polymorphisms within the STAT4 gene

have been associated with IIMs in a Japanese popula-tion.28

REGENERATING MUSCLE CELLS

Chronic muscle inflammation leading to muscle wast-ing is a typical feature of IIMs and contrast with the presence in muscle tissue of immature muscle precur-sors involved in regeneration. Unlike differentiated muscle fibers, the regenerating muscle cells in IIMs over-express class I HLA antigens, TLR3, TLR7 and myositis-specific antigens such ashistidyl-tRNA synthetase, Mi-2 or 3-hydroxy-3-methylglutaryl-coenzyme A (HMGCR) associated with statin-induced IMNM.8,29–32

In addition to making the cells vulnerable to cyto-toxic T cell injury, overexpression of HLA class I anti-gens leads to protein accumulation and misfolding in the endoplasmic reticulum (ER). Such misfolding acti-vates the ER stress pathway leading to activation of the NFjB pathway with enhanced synthesis of pro-inflam-matory cytokines and endogenous class I MHC up-regu-lation, and ER death pathway through caspase 12, leading to muscle degeneration.33,34 In presence of necrotic cell materials, muscle autoantigens released from regenerating muscles may activate IFN.35Further studies suggest that PAMPs or alarmins, notably high mobility group box 1 (HMB1), released from degener-ated and necrotic muscle cells, activate myoblasts through the TLR3 pathway.8,35 Activation of TLR3 on myoblasts leads to release of IL6 and CCL20.7 This is further enhanced in the presence of IL17. IL6 and CCL20 contribute to recruitment and differentiation of DCs and T cells, particularly TH17 cells. The

chemotac-tic effect of muscle autoantigens on DCs and lympho-cytes have been mentioned earlier. Moreover, TLR3 activation leads to expression of IFNb by the muscle cells.7 Contrary to the expectation from above-men-tioned activation of pDCs, IFNb, and not IFNa induci-ble transcripts, are upregulated in IIMS.30 It may

indicate that the major source of type I IFN in IIMs may be immature muscle cells rather than plasmacytoid DCs. IFNb in turn activates nuclear mechanisms leading

(4)

to inflammation and also further up-regulates class I HLA expression on myocytes.7

GENETICS AND ENVIRONMENT

A genome-wide association study in DM has confirmed the importance of the MHC complex but also suggested non-MHC genetic features overlapped with other autoimmune diseases.36 Further studies support the link between HLA genotype and IIM serological or clini-cal phenotype. The 8.1 ancestral haplotype (HLA-A1-B8-Cw7-DRB1*0301-DQA1*0501-C4A*Q0), which is carried by most Caucasians with HLA-B8 is associated with various immune-mediated disorders, including IIM.37Alleles of the 8.1 ancestral haplotype are impor-tant risk markers for the development of IIM with dis-tinct allelic profiles associated with different myositis antibodies.38,39 In a study in the Han population in

China, specific HLA class II alleles were found to be associated with DM and its pulmonary complications and dysphagia.40 An association between HLA-DRB1 alleles and anti-melanoma differentiation-associated gene 5 antibodies has been reported in a Japanese pop-ulation.41HLA-DR11 allele has been reported to confer risk for anti-HMGCR positive statin-induced myopa-thy.42In sporadic IBM, strongest association has been reported with HLA-DRB1*03:01/*01:01 genotype.43

There are evidences that environmental triggers play an important role in the pathogenesis of IIMs. A large proportion of T cells in the blood and muscles of IIMs bears CD28nullphenotype.18Circulating CD28nullwere significantly more common in cytomegalovirus (CMV) seropositive IIM patients, suggesting that viral infec-tions may be important triggers in IIMs.18 In a recent report, the causation of PM has been linked with recur-rent infection in bronchiectasis.44 The frequency of smoking was higher in anti-Jo-1 positive IIM patients compared to controls. In these smokers, the frequency of HLA-DRB1*03 was increased.45

It not only indicates an association between smoking and pathogenesis of IIMs, but also provides a testimony to interaction between environment and gene. The association between statins and IIMs has already been referred to.32,42Low serum vitamin D level is found to be asso-ciated with IIMs.46

SPECIAL CASE OF SPORADIC INCLUSION

BODY MYOSITIS

It appears from a recently reported study that the prevalence of sIBM has increased in recent decades.47

sIBM has certain distinctive features for which its pathogenesis merits a separate discussion. There are evidences of both degenerative and inflammatory pro-cesses in its pathogenesis. Evidences of adaptive immunity include presence of mDCs, CD4+ T cells, cytotoxic CD8+ T cells and plasma cells in muscle tis-sue,48,49 clonally restricted maturation of intramuscu-lar T and B cells,21 and a sIBM-specific autoantibody directed against cytosolic 50-nucleotidase 1A (cN1A).50 Evidences of degeneration include appear-ance of vacuoles and deposits of abnormal proteins (b-amyloid, tau and a-synuclein) in the cells and in filamentous inclusions. It has been postulated that in genetically predisposed individuals, effects of ageing (maybe acting through oxidative stress-related muta-tion in mitochondrial DNA)51 and environmental factors, including viral infection52 may reduce the sir-tuin 1 (SIRT1) activity.53It leads to increase in

mem-brane-bound amyloid b precursor protein (AbPP).53 AbPP has three-fold effect: (i) it inhibits proteasome and thus leads to accumulation of misfolded pro-teins; (ii) it leads to increase in myostatin leading to muscle fiber atrophy; and (iii) abnormal processing of AbPP generates a cleavage product Ab.54 Ab

mole-cules can aggregate to form flexible soluble oligomers which can induce other Ab molecules to also take the misfolded oligomeric form, leading to a chain reaction. These oligomer aggregates may precipitate as congophilic materials. Ab may cause damage to the cells by generating reactive oxygen species, lipid per-oxidation and so on. Misfolded Ab can induce tau to misfold. Misfolded proteins accumulated as a result of overproduction of AbPP and Ab as well as inhibi-tion of proteasome result in ER stress.54 As men-tioned earlier, ER stress may lead to activation of the NFjB pathway and enhanced synthesis of pro-inflam-matory cytokines, and to apoptosis through the cas-pase pathway.33 ER stress-related activation of NFjB may be one of the linkages between degenerative and inflammatory mechanisms found in sIBM. Myonu-clear breakdown is evidenced by lining of the rimmed vacuoles by nuclear membrane proteins like laminin A/C, 12 emerin and histone 120.55 Accumu-lation of nuclear nucleic acid binding protein trans-activation response DNA protein 43 in the extra nuclear sarcoplasm may represent myonuclear break-down or increased shuttling of nuclear proteins to sarcoplasm.56 Looked from the perspective of

innu-merable sketchy and mutually contradictory research observations, the above scheme of pathogenesis of sIBM sounds an oversimplified one. There is much

(5)

controversy around most of the steps described in this scheme and the pathogenesis of this disease still remains a mystery.57

EMERGING THERAPEUTIC TARGETS

Cytokine inhibition through TNFa inhibitors broadly used with efficacy in RA and spondyloarthritis is still controversial in IIM.58,59 It should not be recom-mended in regard to the possibility of muscle disease exacerbation. IL1 blockade by anakinra, an IL1 recep-tor antagonist, has been used in 15 patients for 12 months with clinical response in six patients. IL1RA could act on TH1/TH17 balance, reducing TH17

pathway activation.60 No improvement with anakinra was noted in four patients with sIBM.61The therapeu-tic effect of intravenous immunoglobulins may be mediated by the TH17 lymphocyte pathway.3 In

addi-tion, development of monoclonal antibodies targeting IL12/IL23 (ustekinumab) or IL17 pathway (secuk-inumab and brodalumab) tested in RA, spondy-loarthritis and psoriasis, may be beneficial in IIM as well. Tocilizumab is a monoclonal antibody that acts as an IL6 receptor antagonist developed to treat RA. In addition to inhibiting IL-6 activity, it blocks its autocrine growth activity on B cells. Its efficacy for refractory PM was reported in two patients.62 Treat-ment with tocilizumab requires further investigations in IIM and a randomized controlled study is ongoing (Clinical Trials NCT02043548).

Rituximab (RTX), an CD20 (B cell) anti-body, is found to be effective in IIMs.63–65The national French registry, the Auto-immunity and Rituximab (AIR) registry, which contains data from patients with autoimmune diseases treated with RTX since 2005, ana-lyzed the efficacy and tolerance of RTX in 30 patients with refractory IIMs. RTX was well tolerated, effective in 16 patients, time of improvement after the first infusion was 3.2 months (2–5 months).64

Unfortunately, RTX has not shown efficacy in the only randomized pla-cebo-phase trial performed including 200 patients.63 Seventy-six PM, 76 DM and 48 JDM refractory to ster-oids and immunosuppressive therapy, were random-ized to receive either rituximab at the beginning of the study or rituximab 8 weeks later. Although there were no significant differences between the arms arms at the endpoint (week 8), 83% of patients improved at the end of the follow-up (week 44). While the trial itself was negative, the overall response rate in a group of patients with refractory myositis suggest that RTX was effective but the study design made identification of

such an effect difficult. The long time to improvement after RTX and the assessment of the response after only 8 weeks of placebo, made it difficult to distinguish the response in the two treatment arms. Presence of antisynthetase and anti-Mi-2 autoantibodies, juvenile DM subset, and lower disease damage were predictors of good response, indicating a subset of patients in whom B cells might play a more pivotal role.66

Targeting T cells might be an effective option for the treatment of refractory myositis. Alemtuzumab, a mon-oclonal antibody (MAb) targeting CD52, a protein pre-sent on CD28nullT cells, is reported to be effective in IBM67 and polymyositis.68 T cell costimulation block-ade with abatacept has been discussed in one case report.69

Sifalimumab, an anti-IFN-a monoclonal antibody, suppressed the type I IFN gene signature and appeared to be promising in a phase 1b clinical trial in IIMs.70

Fifty-one patients were included in a randomized, dou-ble-blind placebo-controlled study. Neutralization of the type I IFN gene signature correlated with improve-ment in muscle testing.

Inhibition of the ubiquitin proteasome pathway with bortezomib improved muscle function in a mouse model of myositis.34

From the pathogenesis that has been discussed above, targeting BAFF (BlyS), IL15, IL18 or miRNA offers potential therapeutic alternatives. The recently approved belimumab, a monoclonal antibody, effectively targets the soluble form of BlyS and has shown efficacy in sys-temic lupus erythematosus. One trial with belimumab started in January 2015 in IIMs (ClinicalTrials.gov: NCT02347891). Other trials with experimental treat-ments are ongoing, such as inhibition of complement activity by humanized anti-C5 antibody in DM, (Clini-calTrials.gov: NCT00005571), siponimod as elective sphingosine 1-phosphate (S1P)-1 and -5 receptor that regulates lymphocyte migration in PM and DM (Clini-calTrials.gov: NCT01148810), bimagrumab that modu-lates myostatin and activin signaling (ClinicalTrials.gov: NCT01925209) or arimoclomol that could enhance heat shock protein expression with neuroprotective effects in IBM (ClinicalTrials.gov: NCT00769860).

CONCLUSIONS

An interaction between dendritic cell-lymphocyte path-way and regenerating myocytes may be responsible for the pathogenesis of the IIMs. An appropriate genetic predisposition and a chance encounter with some environmental trigger also appear to provide the

(6)

background. Currently identified molecular targets are ushering an advent of more specific therapeutic options. While no biologic agent is currently consis-tently effective in IIMs, it appears that we are heading toward discovering mechanisms responsible for hom-ing of autoimmunity specifically to the muscles. How-ever, a highly specific, effective and individualized therapy remains beyond our dreams.

ACKNOWLEDGMENTS

The authors wish to acknowledge Syed Jamil Abdal and Tanveer Hasan for their help in preparation of the manuscript.

REFERENCES

1 Hoogendijk J, Amato AA, Lecky BR et al. (2004) 119th ENMC international workshop: trial design in adult idio-pathic inflammatory myopathies, with the exception of inclusion-body myositis. Neuromuscul Disord 14, 337–45. 2 Troyanov Y, Targoff IN, Tremblay JL et al. (2005) Novel

classification of idiopathic inflammatory myopathies based on overlap syndrome features and autoantibodies: analysis of 100 French Canadian patients. Medicine 84, 231–49.

3 Tournadre A, Porcherot M, Cherin P, Marie I, Hachulla E, Miossec P (2009) TH1 and TH17 balance in inflammatory myopathies: interaction withdendritic cells and possible link with response to high-dose immunoglobulins. Cytokine 46, 297–301.

4 Lopez de Padilla CM, Vallejo AN, McNallan KT et al. (2007) Plasmacytoid dendritic cells in inflamed muscle of patients with juvenile dermatomyositis. Arthritis Rheum 56, 1658–68.

5 Greenberg SA, Pinkus GS, Amato AA, Pinkus JL (2007) Myeloid dendritic cells in inclusion-body myositis and polymyositis. Muscle Nerve 35, 17–23.

6 Page G, Chevrel G, Miossec P (2004) Anatomic localiza-tion of immature and mature dendritic cell subsets in der-matomyositis and polymyositis: interaction with chemokines and TH1 cytokine-producing cells. Arthritis Rheum 50, 199–208.

7 Tournadre A, Lenief V, Eljaafari A, Miossec P (2012) Im-mature muscle precursors are a source of interferon-b in myositis: role of Toll-like receptor 3 activation and contri-bution to HLA class I up-regulation. Arthritis Rheum 64, 533–41.

8 Tournadre A, Lenief V, Miossec P (2010) Expression of Toll-like receptor 3 and Toll-like receptor 7 in muscle is characteristic of inflammatory myopathy and is differen-tially regulated by TH1 and TH17 cytokines. Arthritis Rheum 62, 2144–51.

9 Wang DX, Lu X, Zu N et al. (2012) Clinical significance of peripheral blood lymphocyte subsets in patients with polymyositis and dermatomyositis. Clin Rheumatol 31, 1691–7.

10 Lindberg C, Oldfors A, Tarkowski A (1994) Restricted use of T cell receptor V genes in endomysial infiltrates of patients with inflammatory myopathies. Eur J Immunol 24, 2659–63.

11 Tucci M, Quatraro C, Dammacco F, Silvestris F (2006) In-terleukin-18 overexpression as a hallmark of the activity of autoimmune inflammatory myopathies. Clin Exp Immunol 146, 21–31.

12 Chevrel G, Page G, Granet C, Streichenberger N, Varennes A, Miossec P (2003) Interleukin-17 increases the effects of IL-1b on muscle cells: arguments for the role of T cells in the pathogenesis of myositis. J Neuroimmunol 137, 125– 33.

13 Tournadre A, Miossec P (2009) Chemokines and dendritic cells in inflammatory myopathies. Ann Rheum Dis 68, 300–4.

14 Hohlfeld R, Engel AG (1991) Coculture with autologous myotubes of cytotoxic T cells isolated from muscle in inflammatory myopathies. Ann Neurol 29, 498–507. 15 Zong M, Loell I, Lindroos E et al. (2012) Effects of

immunosuppressive treatment on interleukin-15 and interleukin-15 receptor a expression in muscle tissue of patients with polymyositis and dermatomyositis. Ann Rheum Dis 71, 1055–63.

16 Levine SM, Raben N, Xie D et al. (2007) Novel conforma-tion of histidyl-transfer RNA synthetase in the lung: the target tissue in Jo-1 autoantibody-associated myositis. Arthritis Rheum 56, 2729–39.

17 Howard OM, Dong HF, Yang D et al. (2002) Histidyl-tRNAsynthetase and asparaginyl-Histidyl-tRNAsynthetase, autoanti-gens in myositis, activate chemokine receptors on T lym-phocytes and immature dendritic cells. J Exp Med 196, 781–91.

18 Fasth AE, Dastmalchi M, Rahbar A et al. (2009) T cell infil-trates in the muscles of patients with dermatomyositis and polymyositis are dominated by CD28nullT cells. J Immunol 183, 4792–9.

19 Fasth AE, Bjorkstrom NK, Anthoni M et al. (2010) Activat-ing NK-cell receptors co-stimulate CD4(+)CD28nullT cells in patients with rheumatoid arthritis. Eur J Immunol 40, 378–87.

20 Loell IM, Pandya J, Raghavan S et al. (2011) Persisting CD28(null) T cells, but not regulatory T cells, in muscle tissue of myositis patients after immunosuppressive ther-apy (Abstract). Arthritis Rheum 63, S86.

21 Bradshaw EM, Orihuela A, McArdel SL et al. (2007) A local antigen-driven humoral response is present in the inflammatory myopathies. J Immunol 178, 547–56. 22 Betteridge ZE, Gunawardena H, McHugh NJ (2011) Novel

autoantibodies and clinical phenotypes in adult and juve-nile myositis. Arthritis Res Ther 13, 209.

(7)

23 Eloranta ML, Helmers SB, Ulfgren AK, R€onnblom L, Alm GV, Lundberg IE (2007) A possible mechanism for endogenous activation of the type I interferon system in myositis patients with anti-Jo-1 or anti-Ro 52/anti-Ro 60 autoantibodies. Arthritis Rheum 56, 3112–24.

24 Walsh RJ, Kong SW, Yao Y et al. (2007) Type I interferon-inducible gene expression in blood is present and reflects disease activity in dermatomyositis and polymyositis. Arthritis Rheum 56, 3784–92.

25 Bartel DP (2009) MicroRNAs: target recognition and regu-latory functions. Cell 136 (2), 215–33.

26 Ceribelli A, Yao B, Dominguez-Gutierrez PR, Nahid MA, Satoh M, Chan EK (2011) MicroRNAs in systemic rheu-matic diseases. Arthritis Res Ther 13, 229.

27 Georgantas RW, Streicher K, Greenberg SA et al. (2014) In-hibition of myogenic microRNAs 1, 133, and 206 by inflammatory cytokines links inflammation and muscle degeneration in adult inflammatory myopathies. Arthritis Rheumatol 66, 1022–33.

28 Sugiura T, Kawaguchi Y, Goto K et al. (2014) Positive association between STAT4 polymorphisms and polymyositis/dermatomyositis in a Japanese population. Ann Rheum Dis 71, 1646–50.

29 Emslie-Smith AM, Arahata K, Engel AG (1989) Major his-tocompatibility complex class I antigen expression, immunolocalization of interferon subtypes, and T cell-me-diated cytotoxicity in myopathies. Hum Pathol 20, 224–31. 30 Cappelletti C, Baggi F, Zolezzi F (2011) Type I interferon and Toll-like receptor expression characterizes inflamma-tory myopathies. Neurology 76, 2079–88.

31 Casciola-Rosen L, Nagaraju K, Plotz P et al. (2005) En-hanced autoantigen expression in regenerating muscle cells in idiopathic inflammatory myopathy. J Exp Med 201, 591–601.

32 Mammen AL, Chung T, Christopher-Stine L et al. (2011) Autoantibodies against 3-hydroxy-3-methylglutaryl-coen-zyme A reductase in patients with statin-associated autoimmune myopathy. Arthritis Rheum 63, 713–21. 33 Nagaraju K, Casciola-Rosen L, Lundberg I et al. (2005)

Ac-tivation of the endoplasmic reticulum stress response in autoimmune myositis: potential role in muscle fiber dam-age and dysfunction. Arthritis Rheum 52, 1824–35. 34 Rayavarapu S, Coley W, Van der Meulen JH et al. (2013)

Activation of the ubiquitin proteasome pathway in a mouse model of inflammatory myopathy: a potential ther-apeutic target. Arthritis Rheum 65 (12), 3248–58.

35 Harris HE, Andersson U, Pisetsky DS (2012) HMGB1: a multifunctional alarmin driving autoimmune and inflam-matory disease. Nat Rev Rheumatol 8, 195–202.

36 Miller FW, Cooper RG, Vencovsky J et al.; Myositis Genet-ics Consortium (2013) Genome-wide association study of dermatomyositis reveals genetic overlap with other autoimmune disorders. Arthritis Rheum 12, 3239–47. 37 Price P, Witt C, Allcock R et al. (1999) The genetic basis

for the association of the 8.1 ancestral haplotype (A1, B8,

DR3) with multiple immunopathological diseases. Im-munol Rev 167, 257–74.

38 O’Hanlon TP, Carrick DM, Arnett FC et al. (2005) Im-munogenetic risk and protectivefactors for the idiopathic inflammatory myopathies: distinct HLAA, B, Cw, -DRB1and -DQA1 allelic profiles and motifs define clinico-pathologic groups in caucasians. Medicine (Baltimore) 84, 338–49.

39 O’Hanlon TP1, Carrick DM, Targoff IN et al. (2006) Im-munogenetic risk and protective factors for the idiopathic inflammatory myopathies: distinct HLAA, B, Cw, -DRB1, and -DQA1 allelic profiles distinguish European American patients with different myositis autoantibodies. Medicine (Baltimore) 85, 111–27.

40 Gao X, Han L, Yuan L et al. (2014) HLA class II alleles may influence susceptibility to adult dermatomyositis and polymyositis in a Han Chinese population. BMC Dermatol 14 (9), 1–7.

41 Gono T, Kawaguchi Y, Kuwana M et al. (2012) Brief report: association of HLA-DRB1* 0101/*0405 with sus-ceptibility to antimelanoma differentiation associated gene 5 antibody-positive dermatomyositis in the Japanese population. Arthritis Rheum 64, 3736–40.

42 Mammen AL, Gaudet D, Brisson D et al. (2012) Increased frequency of HLA DRB1*11:01 in anti-hydroxymethylglu-taryl-coenzyme A reductaseassociated autoimmune myopathy. Arthritis Care Res (Hoboken) 64, 1233–7. 43 Rojana-udomsart A, James I, Castley A et al. (2012)

High-resolution HLA-DRB1 genotyping in an Australian inclu-sion body myositis (s-IBM) cohort: an analysis of disease-associated alleles and diplotypes. J Neuroimmunol250, 77– 82.

44 Ku BD, Shin HY (2012) Recurrentpolymyositis triggered by bronchiectasis-associated infections. Int J Rheum Dis 15 (5), e109–12.

45 Chinoy H, Adimulam S, Marriage F et al. (2012) Interac-tion of HLA-DRB1*03 and smoking for the development of anti-Jo-1 antibodies in adult idiopathic inflammatory myopathies: a European-wide case study. Ann Rheum Dis 71, 961–5.

46 Azali P, BarbassoHelmers S, Kockum I et al. (2013) Low serum levels of vitamin D in idiopathic inflammatory myopathies. Ann Rheum Dis 72 (4), 512–6.

47 Tan JA, Roberts-Thomson PJ, Blumbergs P, Hakendorf P, Cox SR, Limaye V (2013) Incidence and prevalence of idiopathic inflammatory myopathies in South Australia: a 30-year epidemiologic study of histology-proven cases. Int J Rheum Dis 16 (3), 331–8.

48 Engel AG, Arahata K (1984) Monoclonal antibody analy-sis of mononuclear cells in myopathies. II: Phenotypes of autoinvasive cells in polymyositis and inclusion body myositis. Ann Neurol 16, 209–15.

49 Greenberg SA, Bradshaw EM, Pinkus JL et al. (2005) Plasma cells in muscle in inclusion body myositis and polymyositis. Neurology 65, 1782–7.

(8)

50 Larman HB, Salajegheh M, Nazareno R et al. (2013) Cy-tosolic 50-nucleotidase 1A autoimmunity in sporadic inclusion body myositis. Ann Neurol 73 (3), 408–18. 51 Pak JW, Herbst A, Bua E, Gokey N, McKenzie D, Aiken JM

(2003) Mitochondrial DNA mutations as a fundamental mechanism in physiological declines associated with aging. Aging Cell 2, 1–7.

52 Dalakas MC, Rakocevic G, Shatunov A, Goldfarb L, Raju R, Salajegheh M (2007) Inclusion body myositis with human immunodeficiency virus infection: four cases with clonal expansion of viral-specific T cells. Ann Neurol 61, 466–75.

53 Nogalska A, D’Agostino C, Engel WK, Davies KJ, Askanas V (2007) Decreased SIRT1 deacetylase activity in sporadic inclusion-body myositis muscle fibers. Neurobiol Aging 31, 1637–48.

54 Askanas V, Engel WK, Nogalska A (2012) Pathogenic con-siderations in sporadic inclusion-body myositis, a degen-erative muscle disease associated with aging and abnormalities of myoproteostasis. J Neuropathol Exp Neurol 71 (8), 680–93.

55 Greenberg SA, Pinkus JL, Amato AA (2006) Nuclear mem-brane proteins are present within rimmed vacuoles in inclusion-body myositis. Muscle Nerve 34, 406–16. 56 Salajegheh M, Pinkus JL, Taylor JP et al. (2009)

Sarcoplas-mic redistribution of nuclear TDP-43 in inclusion body myositis. Muscle Nerve 40 (1), 19–31.

57 Greenberg SA (2010) Theories of the pathogenesis of inclusion body myositis. Curr Rheumatol Rep 12 (3), 221– 8.

58 Tournadre A, Dubost JJ, Soubrier M (2010) Treatment of inflammatory muscle disease in adults. Joint Bone Spine 77, 390–4.

59 The Muscle Study Group (2011) A randomized pilot trial of etanercept in dermatomyositis. Ann Neurol 70, 427–36.

60 Zong MI, Dorph C, Dastmalchi M et al. (2014) Anakinra treatment in patients with refractory inflammatory myopathies and possible predictive response biomarkers:

a mechanistic study with 12 months follow-up. Ann Rheum Dis 73, 913–20.

61 Kosmidis ML, Alexopoulos H, Tzioufas AG, Dalakas MC (2013) The effect of anakinra, an IL1 receptor antagonist, in patients with sporadic inclusion body myositis (sIBM): a small pilot study. J Neurol Sci 334, 123–5.

62 Narazaki M, Hagihara K, Shima Y, Ogata A, Kishimoto T, Tanaka T (2011) Therapeutic effect of tocilizumab on two patients with polymyositis. Rheumatology (Oxford) 50, 1344–6.

63 Oddis CV, Reed AM, Aggarwal R et al. (2013) Rituximab in the treatment of refractory adult and juvenile dermato-myositisand adult polymyositis. Arthritis Rheum 65, 314–24. 64 Couderc M, Gottenberg JE, Mariette X et al. (2011) Efficay and safety of rituximab in the treatment of refractory inflammatorymyopathies in adults: results from the AIR registry. Rheumatology (Oxford) 50, 2283–9.

65 Unger L, Kampf S, L€uthke K, Aringer M (2014) Rituximab therapy in patients with refractory dermatomyositis or polymyositis: differential effects in a real-life population. Rheumatology (Oxford) 53, 1630–8.

66 Aggarwal R, Bandos A, Reed AM et al. (2014) Predictors of clinical improvement in rituximab-treated refractory adult and juvenile dermatomyositis and adult polymyositis. Arthritis Rheumatol 66, 740–9.

67 Dalakas MC, Rakocevic G, Schmidt J et al. (2009) Effect of Alemtuzumab (CAMPATH 1-H) in patients with inclu-sion-body myositis. Brain 132, 1536–44.

68 Ruck T, Bittner S, Kuhlmann T, Wiendl H, Meuth SG (2015) Long-term efficacy of alemtuzumab in polymyosi-tis. Rheumatology (Oxford) 54, 560–2.

69 Kerola AM, Kauppi MJ (2015) Abatacept as a successful therapy for myositis– a case-based review. Clin Rheumatol 34, 609–12.

70 Higgs BW, Zhu W, Morehouse C et al. (2014) A phase 1b clinical trial evaluating sifalimumab, an anti-IFN-a monoclonal antibody, shows target neutralisation of a type I IFN signature in blood of dermatomyositis and polymyositis patients. Ann Rheum Dis 73, 256–62.

Références

Documents relatifs

être marcher / être faire avoir / être avoir / aller venir penser / aller / dire. Ecris si les groupes nominaux sont au singulier ou au pluriel et au masculin ou au

le nombre de personnes en recherche d’emploi entrées en formation au cours de l’année au nombre de personnes ayant déclaré avoir connu au moins un mois principalement au chômage

Chaque mois, l’ensemble des demandeurs d’emploi inscrits doivent actualiser leur situation auprès de Pôle emploi. Ils disposent pour cela d’une plage d’un peu plus de 15

BAF, barrier-to-autointegration factor; ECM, Extracellular matrix; EDMD, Emery-Dreifuss muscular dystrophy; ERK, extracellular signal-regulated kinase; F-actin, actin filament;

De plus, nous ne voulons pas seulement être en mesure d’indiquer les services qui permettent de résoudre un problème fonctionnel, mais nous voulons aussi donner les va- leurs

dermatomyositis, polymyositis and CADM can also be associated with other presentations of respiratory involvement, namely pulmonary arterial hypertension (PAH) [5], pleural disease

Among the high number of genes specifically over-expressed in the G1 subgroup (HBV infection with a low number of viral DNA, AXIN1 mutations, a younger age, a high sera level of

nutrient balances and soil fertility. The Upper Oueme catchment in the West African subhumid zone is a region in Northern Benin, which actually experiences major changes in land