You are on page 1of 7

Immunological Medicine

ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/timm20

Murine models of idiopathic inflammatory


myopathy

Risa Konishi, Yuki Ichimura & Naoko Okiyama

To cite this article: Risa Konishi, Yuki Ichimura & Naoko Okiyama (2023) Murine
models of idiopathic inflammatory myopathy, Immunological Medicine, 46:1, 9-14, DOI:
10.1080/25785826.2022.2137968

To link to this article: https://doi.org/10.1080/25785826.2022.2137968

© 2022 The Author(s). Published by Informa


UK Limited, trading as Taylor & Francis
Group on behalf of the Japanese Society of
Clinical Immunology.

Published online: 25 Oct 2022.

Submit your article to this journal

Article views: 1250

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=timm20
IMMUNOLOGICAL MEDICINE
2023, VOL. 46, NO. 1, 9–14
https://doi.org/10.1080/25785826.2022.2137968

REVIEW ARTICLE

Murine models of idiopathic inflammatory myopathy


Risa Konishi , Yuki Ichimura and Naoko Okiyama
Department of Dermatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University,
Tokyo, Japan

ABSTRACT ARTICLE HISTORY


Idiopathic inflammatory myopathies (IIMs) are characterized by inflammation of muscles and Received 19 August 2022
other organs. Several myositis-specific autoantibodies (MSAs) have been identified in IIMs Accepted 16 October 2022
and were found to be associated with distinct clinical features. Although MSAs are valuable
KEYWORDS
for the diagnosis of IIMs, the pathogenic roles of these antibodies remain unknown. To
Murine model; idiopathic
investigate the pathogenesis of IIMs, several animal models of experimental myositis have inflammatory myopathy;
been established. Classical murine models of autoimmune myositis, experimental auto- dermatomyositis; myositis-
immune myositis, and C protein-induced myositis are established by immunization with specific autoantibody; TIF1c
muscle-specific antigens, myosin, and skeletal C protein, respectively. Furthermore, a murine
model of experimental myositis was generated by immunization with a murine recombinant
histidyl-tRNA synthetase, Jo-1, in which muscle and lung inflammation reflecting anti-synthe-
tase syndrome are induced depending on acquired immunity. Recently, the transfer of
human IgGs from patients with immune-mediated necrotizing myopathy, comprising anti-
signal recognition particles and anti-3-hydroxy-3-methylglutaryl coenzyme A reductase anti-
bodies, was found to induce complement-mediated myositis in recipient mice. CD8þ T cell-
mediated myositis can be established depending on autoimmunity against transcriptional
intermediary factor 1c (TIF1c), an autoantigen for MSAs induced by recombinant human
TIF1c immunization. These new murine models reflecting MSA-related IIMs are useful tools
for accurately understanding the pathological mechanisms underlying IIMs.

1. Introduction IIMs and are reportedly associated with distinct


clinical features [4–6]. Almost all MSAs have been
Idiopathic inflammatory myopathies (IIMs), includ-
identified in patients with DM: anti-Mi-2, anti-mel-
ing dermatomyositis (DM), polymyositis (PM),
anoma differentiation-associated gene 5 (MDA5),
immune-mediated necrotizing myopathy (IMNM),
anti-transcriptional intermediary factor 1c (TIF1c),
inclusion body myositis (IBM), and anti-synthetase
anti-nuclear matrix protein 2 (NXP2), and anti-
syndrome (ASS), are characterized by inflammation
small ubiquitin-like modifier activating enzyme
of the proximal arm and leg muscles, and other
(SAE) antibodies, each of which was found to be
organs, such as the skin, lung, and joints are fre- associated with characteristic clinical features [6].
quently affected [1]. Anti-MDA5 antibody exhibits high specificity for
DM is distinguished from other IIMs based on clinically amyopathic DM, presenting with rapidly
its specific cutaneous manifestations, heliotrope progressive interstitial lung disease (ILD) [7]. Anti-
rash, and Gottron papules/signs. Perifascicular atro- TIF1c antibody has been detected in juvenile and
phy is the hallmark histopathological feature of DM, adult patients with DM and is closely correlated
along with the sarcoplasmic expression of myxovirus with internal malignancies, especially in elderly
resistance protein A (MxA) and sarcolemmal mem- patients [8]. Previously, we reported that the anti-
brane attack complex deposition [2]. Conversely, NXP2 antibody, also detected in juvenile and adult
IMNM is characterized by relatively severe proximal patients with IIMs, is characterized by extensive
weakness and myofiber necrosis with minimal muscular involvement but occasionally lacks a typ-
inflammatory cell infiltration on muscle biopsy [3], ical rash [9]. In contrast, anti-SAE antibody-positive
the hallmark histological feature of IBM. patients with DM frequently display extensive
DM, IMNM, and ASS are characterized by the rash [10].
presence of myositis-specific autoantibodies (MSAs). ASS is a subgroup of IIMs presenting a DM/PM
Several MSAs have been identified in patients with phenotype defined by the presence of autoantibodies

CONTACT Naoko Okiyama okiy.derm@tmd.ac.jp Department of Dermatology, Graduate School of Medical and Dental Sciences, Tokyo Medical
and Dental University, Tokyo, Japan
ß 2022 The Author(s). Published by Informa UK Limited, trading as Taylor & Francis Group on behalf of the Japanese Society of Clinical Immunology.
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0/), which permits
unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
10 R. KONISHI ET AL.

Table 1. Murine models of inflammatory myopathies.


Experimental Upregulation of Anti-SRP¶/HMGCR§
autoimmune MHC class I C protein-induced Jo-1- TIF1c-induced antibodies-
Model name myositis (EAM) in muscle myositis (CIM) immunized mice myositis (TIM) transferred mice
Related None None None Anti- Dermatomyositis Immune-mediated
human disease synthetase necrotizing
syndrome myopathy
Myositis-specific None None None Anti- Anti- Anti-SRP¶/
autoantibody (Some mice ARS† antibody TIF1c HMGCR§
have anti- antibody antibodies
ARS† antibody)
Clinical features of Not applicable Not applicable Not applicable Myositis, specific Myositis, specific Myositis
related rash, ILD‡ rash,
human disease ILD‡, cancer
Autoantigen in Myosin None Skeletal C protein Jo-1 TIF1c SRP¶, HMGCR§
murine model
Features in a Myositis Myositis Myositis Myositis, ILD‡ Myositis Myositis
murine model
Pathogenesis in a CD4þ T cell Endoplasmic CD8þ T cell Innate immunity CD8þ T cells Antibody
murine model reticulum stress
TIF1c: transcriptional intermediary factor 1c; ¶SRP: signal recognition particle; §HMGCR: 3-hydroxy-3-methylglutaryl coenzyme A reductase; †ARS:
aminoacyl transfer RNA synthetases; ‡ILD: interstitial lung disease.

against aminoacyl transfer RNA synthetases (ARS), 2. Murine models of myositis


accompanied by clinical features such as chronic
2.1. Experimental autoimmune myositis (EAM)
ILD, myositis, arthralgia, fever, and mechanic’s
hands [11]. Anti-ARS antibodies include autoanti- The classical murine model of autoimmune myositis
bodies against histidyl (Jo-1), threonyl (PL-7), alanyl is known as EAM [16,19]. Using SJL/J mice, EAM
(PL-12), glycyl (EJ), isoleucyl (OJ), asparaginyl (KS), can be induced by repeated immunization with
tyrosyl (Ha), and phenylalanyl (Zo); among these, muscle homogenate or partially purified myosin of
anti-Jo-1 antibody is most commonly detected [12]. the skeletal muscle with complete Freund’s adjuvant
IMNM is categorized into three groups: anti-3- (CFA) containing mycobacteria on four occasions,
hydroxy-3-methylglutaryl coenzyme A reductase performed at weekly intervals. Myositis is induced
(HMGCR) antibody-positive IMNM, anti-signal rec- in the quadriceps femoris muscles of immunized
ognition particle (SRP) antibody-positive IMNM, mice, in which CD4þ T cells predominantly infil-
and seronegative IMNM [13]. Anti-SRP antibodies trate the muscle tissue [20]. Immunoblot analysis of
are substantially associated with severe neurological immunoglobulin G (IgG) from EAM mice revealed
symptoms when compared with anti-HMGCR anti- the presence of antibodies against a wide variety of
bodies [3]. Seronegative IMNM has been associated muscle proteins. The transfer of T cells or IgGs col-
with cancer [14]. IBM is also characterized by myo- lected from EAM mice was shown to cause myositis
pathology; however, no specific MSA has in naïve mice [16].
been identified. However, SJL/J mice exhibit a defect in the dys-
Although MSAs are useful for diagnosing and ferlin gene and develop spontaneous muscle fiber
prognostic estimation of IIMs, their pathogenic role degeneration and inflammatory cell infiltration inde-
remains unknown, and it remains to be determined pendent of EAM immunization [21]. Another subse-
whether autoantibodies are simple disease bio- quent study has shown that severe EAM can be
markers or true pathogenic effectors. induced also in BALB/c mice by a twice immuniza-
To comprehensively clarify the pathogenesis of tion with increased administration doses of rat
IIMs and develop novel therapeutic strategies, myosin and pertussis toxin (PT) [22].
numerous animal models of IIMs have been estab-
lished, including infectious, genetic, and antigen-
2.2. Genetically modified mice with upregulation
induced models [15]. Although previously estab-
of major histocompatibility complex (MHC) classI
lished murine models of experimental autoimmune
in the skeletal muscles develop myositis
myositis depend on immune responses against
muscle tissue-specific antigens [16,17], we recently In the human IIMs, upregulation of MHC class I on
established a murine model of myositis by immun- the surface of muscle cells is observed in an early
ization with TIF1c, an autoantigen for a DM-spe- phase. Nagaraju et al. [23] have reported that trans-
cific MSA [18]. Herein, we review the history and genic mice with conditional upregulation of MHC
evolution of murine models of myositis and sum- class I expression in the skeletal muscles develop
marize these models in Table 1. clinically muscle weakness depending on muscle cell
IMMUNOLOGICAL MEDICINE 11

damage and muscle inflammation. The mice often 2.4. Jo-1-induced ASS model
produce autoantibodies including anti-histidyl-tRNA
Another murine model of experimental myositis
synthetase antibodies. The mechanism of the myo-
was generated by immunization with purified epit-
sitis has been described to be depending on activa-
opic peptides derived from histidyl-tRNA synthetase
tion of the endoplasmic reticulum stress response
(HisRS, Jo-1) with CFA [38]. Subcutaneous injec-
[24]. Even though the transgenic mice are not
tions of murine HisRS induced local muscle and
widely available, this murine model well reflects
lung inflammation in congenic B6 mice (B6.G7).
human IIMs.
Histological studies of muscle tissues revealed
diverse infiltration patterns, with perimysial/epimy-
sial inflammation in a perivascular distribution,
2.3. C Protein-induced myositis (CIM) endomysial inflammation, and muscle fiber inva-
Subsequently, biochemical studies have suggested sion/degeneration. The immunized mice showed a
HisRS-specific CD4þ T cell response and antibody
that skeletal C protein, a myosin-binding protein, is
production. However, DO11.10/Rag-2-KO mice with
the major immunogen in the myosin fraction that
can induce EAM in rats [25]. Sugihara et al. [17] abrogated HisRS-specific B and T cell responses
established modified and improved EAM, i.e., CIM, demonstrated significant muscle inflammation after
in C57BL/6 (B6) mice by administering a single sub- immunization with HisRS [39]. Moreover, C3H/HeJ
cutaneous injection of CFA-conjugated human or (TLR4-KO) mice lacking significant anti-HisRS anti-
murine C protein fragments, along with an intraper- body levels exhibited muscle inflammation induced
itoneal injection of pertussis toxin (PT), another by immunization with HisRS. These findings sup-
port a key role for innate immune responses, but
adjuvant. CIM is an improved murine model of
not HisRS-specific autoimmunity, in a HisRS-
autoimmune myositis when compared with classic
EAM, given that CIM can be induced even in B6 induced model of myositis. However, analysis of
mice, the most commonly used mouse strain for bronchoalveolar lavage fluid samples collected from
gene modification. anti-Jo-1 antibody-positive patients with IIM
The C protein-immunized mice develop myositis revealed that HisRS-specific CD4þ T cells might be
2–3 weeks after immunization. Immunohistochemistry present within the lungs of patients [40].
studies revealed that CD8þ T cells localize to the
endomysial site of muscle tissues of CIM mice when
compared with perimysial and perivascular sites. In
addition, Sugihara et al. [26] found that major histo-
2.5. Transfer of human IgGs from patients with
compatibility complex (MHC) class I knockout (KO)
IMNM induces muscle deficiency
mice failed to develop CIM, and perforin deficiency, a
cytolytic protein in cytotoxic CD8þ T cell granules, Anti-SRP and anti-HMGCR antibodies are often
inhibited muscle fiber injury in CIM. Moreover, the detected as MSAs specific to IMNM. Bergua et al.
adoptive transfer of CD8þ T cells collected from CIM [41] found that transferring human IgGs from
mice could induce more severe muscle fiber injury in patients with IMNM, comprising anti-SRP or
recipient B6 mice than in CD4þ T cells. Transferring HMGCR antibodies, provoked muscle deficiency
bone marrow-derived dendritic cells presenting a and myofiber necrosis in recipient B6 wild-type or
CD8 epitope peptide from the C protein could induce Rag-2-KO mice. Muscle deficiency tends to be less
myositis in recipient mice [27]. These results indicate severe in mice receiving IgGs from anti-HMGCR
that CIM is characterized by CD8þ T cell-mediated antibody-positive patients than in those receiving
muscle fiber injury. IgGs from anti-SRP antibody-positive patients.
Moreover, along with autoaggressive CD8þ T These phenomena may reflect the features of IMNM
cells, adjuvant-activated muscle tissue conditions are [3]. The myopathy severity in IMNM IgG-trans-
essential for established CIM, like ’seeds and soil’ ferred complement C3-KO mice was reduced,
[28,29]. Given that innate/acquired immunity medi- whereas supplementation with human complement
ates myositis, previous studies using the CIM model could increase severity. This model suggested that
have identified several therapeutic targets, including patient-derived anti-SRP and anti-HMGCR antibod-
interleukin (IL)-6, IL-1, tumor necrosis factor-a, L- ies are pathogenic toward muscles in vivo through a
selectin, CXCL10, CD80/86, IL-23, and high mobil- complement-mediated mechanism. Accordingly,
ity group box 1 (HMGB1) [30–36]. In contrast, anti-SRP and anti-HMGCR autoantibodies are not
interferon (IFN)-c, but not IL-4, might be a sup- only diagnostic biomarkers of IMNM but also direct
pressive factor in CIM [37]. pathogenic factors.
12 R. KONISHI ET AL.

Figure 1. A new murine model of dermatomyositis, transcriptional intermediary factor 1c (TIF1c)-induced myositis (TIM), can
be induced by weekly subcutaneous injections of recombinant human TIF1c protein emulsified in complete Freund’s adjuvant
(CFA), four times. Adoptive transfer of CD8þ T cells, but not CD4þ T cells and IgGs, from TIF1c-immunized mice induces myo-
sitis in recipient mice.

2.6. Tif1c-induced myositis model (TIM) identified as pathogenic in TIM. In contrast, lMT
mice, which completely lack B-cell lineages, devel-
Anti-TIF1c antibody-positive DM is known to be
oped myositis, and the adoptive transfer of IgGs col-
associated with malignancies, particularly in elderly
lected from TIM mice failed to induce myositis in
patients [8], and may also be associated with preg-
nancy [42]. TIF1c is frequently mutated or overex- recipient mice. These results indicate that B cells
pressed in tumors [43,44] and is overexpressed in and autoantibodies are not essential for developing
the embryo and mammary epithelial cells during TIM. In other words, the anti-TIF1c antibody
pregnancy [45,46]. Therefore, it can be speculated detected in patients with DM may be a diagnostic
that cancer and pregnancy trigger autoimmunity biomarker but not a direct pathogenic factor, unlike
against TIF1c and autoimmunity to TIF1c can anti-SRP/HMGCR antibodies in patients with
induce the establishment of myositis. However, elu- IMNM. Accordingly, TIM, which is dependent on
cidating the suggested mechanisms can be autoimmunity against TIF1c, was mediated by
challenging. TIF1c-specific CD8þ T cells but not TIF1c-specific
We established TIM in B6 mice by administering CD4þ T cells, B cells, and autoantibodies (Figure 1).
weekly subcutaneous injections of recombinant Type I IFNs are suspected to be involved in the
human TIF1cprotein emulsified in CFA four times, pathogenesis of DM [47,48]. We have reported that
along with an intraperitoneal injection of PT [18]. IFN-a/b receptor deficiency partially inhibited the
The immunized mice developed TIF1c-specific T development of TIM. In addition, treatment with
cells and anti-human and murine TIF1c antibodies, tofacitinib, a Janus kinase (JAK) inhibitor for vari-
resulting in myositis in the hamstrings and quadri- ous cytokine receptor signaling, including type I
ceps two weeks after the last immunization. IFN signaling, could treat established TIM without
Histological studies revealed atrophy and necrosis of suppressing TIF1c-specific T cell response and auto-
muscle fibers, accompanied by infiltrating mono- antibody production. Therefore, JAK inhibitors
nuclear cells in the perifascicular and endomysial could afford a good treatment strategy for DM that
sites of muscle tissues. does not involve acquired immunity, which is
Immunohistochemistry studies showed that T important for protecting against pathogen-
cells, particularly CD8þ T cells, predominantly infil- induced infection.
trated and adhered to the muscle fibers, which upre- While EAM and CIM completely depend on
gulated the expression of MHC class I and type I immune responses specific to muscular antigens,
interferon (IFN)-responsive molecule Mx1. Beta 2 myosin, and C protein [16,17], TIM induction relies
microglobulin-KO mice lacking MHC class I expres- on autoimmunity against a ubiquitous intracellular
sion, perforin-KO mice, and anti-CD8 depleting molecule, TIF1c, an autoantigen for MSAs. This
antibody-treated mice rarely developed TIM. new murine model of experimental myositis, which
Furthermore, the adoptive transfer of CD8þ T cells closely reflects the pathological mechanisms of DM,
from TIM mice could induce myositis in recipient could be a useful tool for developing novel treat-
B6 mice, whereas the transfer of CD4þ T cells failed ment strategies against anti-TIF1c antibody-posi-
to exhibit this effect. Collectively, CD8þ T cells were tive DM.
IMMUNOLOGICAL MEDICINE 13

3. Conclusion syndrome: a comprehensive review. Autoimmun


Rev. 2014;13(4–5):367–371.
In recent years, murine models of myositis for each [12] Galindo-Feria AS, Notarnicola A, Lundberg IE,
MSA, such as TIM, have been developed (Table 1). et al. Aminoacyl-tRNA synthetases: on anti-synthe-
These models suggest that each MSA-related IIM tase syndrome and beyond. Front Immunol. 2022;
13(866087):866087.
exhibits distinct pathogenesis; thus, the establish-
[13] Allenbach Y, Benveniste O, Stenzel W, et al.
ment of myositis models for other MSAs can Immune-mediated necrotizing myopathy: clinical
be expected. features and pathogenesis. Nat Rev Rheumatol.
2020;16(12):689–701.
[14] Allenbach Y, Keraen J, Bouvier AM, et al. High
Disclosure statement risk of cancer in autoimmune necrotizing myopa-
No potential conflict of interest was reported by thies: usefulness of myositis specific antibody.
the author(s). Brain. 2016;139(Pt 8):2131–2135.
[15] Ascherman DP. Animal models of inflammatory
myopathy. Curr Rheumatol Rep. 2012;14(3):
ORCID 257–263.
[16] Matsubara S, Okumura S. Experimental auto-
Risa Konishi http://orcid.org/0000-0003-3681-2441 immune myositis in SJL/J mice produced by
Yuki Ichimura http://orcid.org/0000-0003-0375-2243 immunization with syngeneic myosin B fraction.
Naoko Okiyama http://orcid.org/0000-0002-5398-0773 Transfer by both immunoglobulin G and T cells. J
Neurol Sci. 1996;144(1–2):171–175.
[17] Sugihara T, Sekine C, Nakae T, et al. A new mur-
References ine model to define the critical pathologic and
therapeutic mediators of polymyositis. Arthritis
[1] Selva-O’Callaghan A, Pinal-Fernandez I, Trallero-
Rheum. 2007;56(4):1304–1314.
Araguas E, et al. Classification and management of
[18] Okiyama N, Ichimura Y, Shobo M, et al. Immune
adult inflammatory myopathies. Lancet Neurol. response to dermatomyositis-specific autoantigen,
2018;17(9):816–828. transcriptional intermediary factor 1gamma can
[2] Tanboon J, Inoue M, Saito Y, et al. result in experimental myositis. Ann Rheum Dis.
Dermatomyositis: muscle pathology according to 2021;80(9):1201–1208.
antibody subtypes. Neurology. 2022;98(7): [19] Rosenberg NL, Ringel SP, Kotzin BL. Experimental
e739–e749. autoimmune myositis in SJL/J mice. Clin Exp
[3] Watanabe Y, Uruha A, Suzuki S, et al. Clinical fea- Immunol. 1987;68(1):117–129.
tures and prognosis in anti-SRP and anti-HMGCR [20] Rosenberg NL, Kotzin BL. Aberrant expression of
necrotising myopathy. J Neurol Neurosurg class II MHC antigens by skeletal muscle endothe-
Psychiatry. 2016;87(10):1038–1044. lial cells in experimental autoimmune myositis. J
[4] McHugh NJ, Tansley SL. Autoantibodies in myo- Immunol. 1989;142(12):4289–4294.
sitis. Nat Rev Rheumatol. 2018;14(5):290–302. [21] Weller AH, Magliato SA, Bell KP, et al.
[5] Lundberg IE, de Visser M, Werth VP. Spontaneous myopathy in the SJL/J mouse: path-
Classification of myositis. Nat Rev Rheumatol. ology and strength loss. Muscle Nerve. 1997;20(1):
2018;14(5):269–278. 72–82.
[6] Okiyama N. Clinical features and cutaneous mani- [22] Kang J, Zhang HY, Feng GD, et al. Development
festations of juvenile and adult patients of derm- of an improved animal model of experimental
atomyositis associated with Myositis-specific autoimmune myositis. Int J Clin Exp Pathol. 2015;
autoantibodies. J Clin Med. 2021;10(8):1725. 8(11):14457–14464.
[7] Fujimoto M, Watanabe R, Ishitsuka Y, et al. [23] Nagaraju K, Raben N, Loeffler L, et al. Conditional
Recent advances in dermatomyositis-specific auto- up-regulation of MHC class I in skeletal muscle
antibodies. Curr Opin Rheumatol. 2016;28(6): leads to self-sustaining autoimmune myositis and
636–644. myositis-specific autoantibodies. Proc Natl Acad
[8] Fujimoto M, Hamaguchi Y, Kaji K, et al. Myositis- Sci USA. 2000;97(16):9209–9214.
[24] Nagaraju K, Casciola-Rosen L, Lundberg I, et al.
specific anti-155/140 autoantibodies target tran-
Activation of the endoplasmic reticulum stress
scription intermediary factor 1 family proteins.
response in autoimmune myositis: potential role in
Arthritis Rheum. 2012;64(2):513–522.
muscle fiber damage and dysfunction. Arthritis
[9] Ichimura Y, Konishi R, Shobo M, et al. Anti-
Rheum. 2005;52(6):1824–1835.
nuclear matrix protein 2 antibody-positive inflam- [25] Kohyama K, Matsumoto Y. C-protein in the skel-
matory myopathies represent extensive myositis etal muscle induces severe autoimmune polymyosi-
without dermatomyositis-specific rash. tis in lewis rats. J Neuroimmunol. 1999;98(2):
Rheumatology. 2022;61(3):1222–1227. 130–135.
[10] Inoue S, Okiyama N, Shobo M, et al. Diffuse ery- [26] Sugihara T, Okiyama N, Suzuki M, et al.
thema with ’angel wings’ sign in Japanese patients Definitive engagement of cytotoxic CD8 T cells in
with anti-small ubiquitin-like modifier activating C protein-induced myositis, a murine model of
enzyme antibody-associated dermatomyositis. Br J polymyositis. Arthritis Rheum. 2010;62(10):
Dermatol. 2018;179(6):1414–1415. 3088–3092.
[11] Mahler M, Miller FW, Fritzler MJ. Idiopathic [27] Okiyama N, Hasegawa H, Oida T, et al.
inflammatory myopathies and the anti-synthetase Experimental myositis inducible with transfer of
14 R. KONISHI ET AL.

dendritic cells presenting a skeletal muscle C pro- associated with muscle and lung inflammation. J
tein-derived CD8 epitope peptide. Int Immunol. Autoimmun. 2007;29(2–3):174–186.
2015;27(7):327–332. [39] Soejima M, Kang EH, Gu X, et al. Role of innate
[28] Okiyama N, Sugihara T, Oida T, et al. T lympho- immunity in a murine model of histidyl-transfer
cytes and muscle condition act like seeds and soil RNA synthetase (Jo-1)-mediated myositis. Arthritis
in a murine polymyositis model. Arthritis Rheum. Rheum. 2011;63(2):479–487.
2012;64(11):3741–3749. [40] Galindo-Feria AS, Albrecht I, Fernandes-Cerqueira
[29] Kimura N, Hirata S, Miyasaka N, et al. Injury and C, et al. Proinflammatory Histidyl-transfer RNA
subsequent regeneration of muscles for activation synthetase-specific CD4þ T cells in the blood and
of local innate immunity to facilitate the develop- lungs of patients with idiopathic inflammatory
ment and relapse of autoimmune myositis in myopathies. Arthritis Rheumatol. 2020;72(1):
C57BL/6 mice. Arthritis Rheumatol. 2015;67(4): 179–191.
1107–1116. [41] Bergua C, Chiavelli H, Allenbach Y, et al. In vivo
[30] Okiyama N, Sugihara T, Iwakura Y, et al. pathogenicity of IgG from patients with anti-SRP
Therapeutic effects of interleukin-6 blockade in a or anti-HMGCR autoantibodies in immune-medi-
murine model of polymyositis that does not ated necrotising myopathy. Ann Rheum Dis. 2019;
require interleukin-17A. Arthritis Rheum. 2009; 78(1):131–139.
60(8):2505–2512. [42] Oya K, Inoue S, Saito A, et al. Pregnancy triggers
[31] Sugihara T, Okiyama N, Watanabe N, et al. the onset of anti-transcriptional intermediary fac-
Interleukin-1 and tumor necrosis factor a blockade tor 1c antibody-positive dermatomyositis: a case
treatment of experimental polymyositis in mice. series. Rheumatology. 2020;59(6):1450–1451.
Arthritis Rheum. 2012;64(8):2655–2662. [43] Pinal-Fernandez I, Ferrer-Fabregas B, Trallero-
[32] Oishi K, Hamaguchi Y, Matsushita T, et al. A cru- Araguas E, et al. Tumour TIF1 mutations and loss
cial role of L-selectin in C protein-induced experi- of heterozygosity related to cancer-associated myo-
mental polymyositis in mice. Arthritis Rheumatol. sitis. Rheumatology. 2018;57(2):388–396.
2014;66(7):1864–1871. [44] Kasuya A, Hamaguchi Y, Fujimoto M, et al.
[33] Kim J, Choi JY, Park SH, et al. Therapeutic effect TIF1c-overexpressing, highly progressive endomet-
of anti-C-X-C motif chemokine 10 (CXCL10) anti- rial carcinoma in a patient with dermato-myositis
body on C protein-induced myositis mouse. positive for malignancy-associated anti-p155/140
Arthritis Res Ther. 2014;16(3):R126. autoantibody. Acta Derm Venereol. 2013;93(6):
[34] Hasegawa H, Kawahata K, Mizoguchi F, et al. 715–716.
Direct suppression of autoaggressive CD8þ T cells [45] Yan KP, Dolle P, Mark M, et al. Molecular clon-
with CD80/86 blockade in CD8þ T cell-mediated ing, genomic structure, and expression analysis of
polymyositis models of mice. Clin Exp Rheumatol. the mouse transcriptional intermediary factor 1
2017;35(4):593–597. gamma gene. Gene. 2004;334:3–13.
[35] Umezawa N, Kawahata K, Mizoguchi F, et al. [46] Hesling C, Lopez J, Fattet L, et al. Tif1c is essential
Interleukin-23 as a therapeutic target for inflam- for the terminal differentiation of mammary alveo-
matory myopathy. Sci Rep. 2018;8(1):5498. lar epithelial cells and for lactation through
[36] Kamiya M, Mizoguchi F, Kawahata K, et al. SMAD4 inhibition. Development. 2013;140(1):
Targeting necroptosis in muscle fibers ameliorates 167–175.
inflammatory myopathies. Nat Commun. 2022; [47] Uruha A, Nishikawa A, Tsuburaya RS, et al.
13(1):166. Sarcoplasmic MxA expression: a valuable marker
[37] Yoshihashi-Nakazato Y, Kawahata K, Kimura N, of dermatomyositis. Neurology. 2017;88(5):
et al. Interferon-c, but not interleukin-4, sup- 493–500.
presses experimental polymyositis. Arthritis [48] Okiyama N, Yamaguchi Y, Kodera M, et al.
Rheumatol. 2016;68(6):1505–1510. Distinct histopathologic patterns of finger erup-
[38] Katsumata Y, Ridgway WM, Oriss T, et al. tions in dermatomyositis based on Myositis-spe-
Species-specific immune responses generated by cific autoantibody profiles. JAMA Dermatol. 2019;
histidyl-tRNA synthetase immunization are 155(9):1080–1082.

You might also like