Download as pdf or txt
Download as pdf or txt
You are on page 1of 25

Clinical Reviews in Allergy & Immunology

https://doi.org/10.1007/s12016-020-08798-2

Type I Interferons in the Pathogenesis and Treatment


of Autoimmune Diseases
Jiao Jiang 1,2 & Ming Zhao 1,2 & Christopher Chang 3,4 & Haijing Wu 1,2 & Qianjin Lu 1,2

# Springer Science+Business Media, LLC, part of Springer Nature 2020

Abstract
Type I interferons (IFN-Is) are a very important group of cytokines that are produced by innate immune cells but also act on
adaptive immune cells. IFN-Is possess antiviral, antitumor, and anti-proliferative effects, as well are associated with the initiation
and maintenance of autoimmune disorders. Studies have shown that aberrantly expressed IFN-Is and/or type I IFN-inducible
gene signatures in the serum or tissues of patients with autoimmune disorders are linked to their pathogenesis, clinical manifes-
tations, and disease activity. Type I interferonopathies with mutations in genes impacting the type I IFN signaling pathway have
shown symptoms and characteristics similar to those of systemic lupus erythematosus (SLE). Furthermore, both interventions in
animal models and clinical trials of therapies targeting the type I IFN signaling pathway have shown efficacy in the treatment of
autoimmune diseases. Our review aims to summarize the functions and targeted therapies (as well as clinical trials) of IFN-Is in
both adult and pediatric autoimmune diseases, such as SLE, pediatric SLE (pSLE), rheumatoid arthritis (RA), juvenile idiopathic
arthritis (JIA), juvenile dermatomyositis (JDM), Sjögren syndrome (SjS), and systemic sclerosis (SSc), discussing the potential
abnormal regulation of transcription factors and epigenetic modifications and providing a potential mechanism for pathogenesis
and therapeutic strategies for future clinical use.

Keywords Autoimmunedisease . TypeI interferonsignaling pathway . Epigenetic modifications . Systemiclupus erythematosus .


Juvenile idiopathic arthritis . Sjogren’s syndrome . Interferonopathies

Abbreviations IRF Interferon regulatory family


AGS Aicardi-Goutières syndrome ISG Interferon-stimulated gene
BTK Bruton’s tyrosine kinase JAK Janus kinase
cGAS Cyclic GMP-AMP synthase JDM Juvenile dermatomyositis
IFN-I Type I interferon JIA Juvenile idiopathic arthritis
IRAK Interleukin-1 receptor-associated kinase MAVS Mitochondrial antiviral signaling protein
MDA Melanoma differentiation-associated gene
MyD Myeloid differentiation primary response gene
pDC Plasmacytoid dendritic cell
* Haijing Wu
PRR Pattern recognition receptor
chriswu1010@csu.edu.cn
pSjS primary Sjögren syndrome
* Qianjin Lu
pSLE Pediatric systemic lupus erythematosus
qianlu5860@csu.edu.cn
RA Rheumatoid arthritis
1
Department of Dermatology, Second Xiangya Hospital, Central RIG-I Retinoic acid-inducible gene-I
South University, Changsha 410008, Hunan, People’s Republic of SSc Systemic sclerosis
China STAT Signal transducers and activators of transcription
2
Hunan Key Laboratory of Medical Epigenomics, Second Xiangya STING Stimulator of interferon
Hospital, Central South University, Changsha 410008, Hunan, Syk Spleen tyrosine kinase
People’s Republic of China
TBK TANK-binding kinase
3
Division of Pediatric Immunology and Allergy, Joe DiMaggio TLR Toll-like receptor
Children’s Hospital, Hollywood, FL 33021, USA
TRAF Tumor necrosis factor receptor-associated factor
4
Division of Rheumatology, Allergy and Clinical Immunology, TREX Three prime repair exonuclease
University of California, Davis, CA 95616, USA
Clinic Rev Allerg Immunol

TRIF Toll-IL-1 receptor domain-containing adaptor in- antibodies (mAbs) and small molecule inhibitors are designed
ducing IFN-β to target the type I IFN signaling pathway for the treatment of
TYK Tyrosine kinase autoimmune disorders, such as therapies targeting IFN-α or
IFNARs, pDCs, Toll-like receptors (TLRs), and their down-
stream pathways. Clinical trials using these treatments have
Introduction already produced positive results. The JAK inhibitors
tofacitinib and baricitinib have been approved by the United
Autoimmune diseases include a wide range of diseases, in- States Food and Drug Administration (FDA) and the
cluding systemic lupus erythematosus (SLE), rheumatoid ar- European Medicines Agency (EMA) for the treatment of RA
thritis (RA), Sjögren syndrome (SjS), and systemic sclerosis [19]. A mAb against IFNAR, anifrolumab (formerly MEDI-
(SSc) [1]. They occur when the immune system loses the 546), has just completed phase III clinical trials in patients
ability to maintain self-tolerance and produces overactive with SLE and has shown excellent efficacy and safety profiles
immunocytes and an abnormal balance of cytokines [3]. [20]. The JAK1/JAK2 inhibitor baricitinib has also entered
These lead to abnormally increased autoantibodies against phase III trials for the treatment of SLE [21]. Furthermore,
self-tissues, resulting in inflammation and damage to multiple there are also many ongoing clinical trials and experiments
organs, such as the kidneys, joints, skin, and central nervous about treatments blocking the type I IFN pathway in both
system. The conventional treatments of autoimmune disorders humans and animals with SLE, pediatric SLE (pSLE), RA,
include anti-inflammatory and immunosuppressive agents, JIA, SjS, and other autoimmune diseases. However, there are
which often provide only partial amelioration of symptoms also some challenges that will be discussed in more detail
[4–8]. In addition, these agents possess numerous side effects, below. We focus on type I IFNs (mainly IFN-α and IFN-β)
including infection, obesity, abnormal glucose metabolism, and summarize their roles in the pathogenesis and treatment of
osteoporosis, and even avascular necrosis [4, 5]. Due to the autoimmune diseases. Our purpose is to find new potential
complex and unclear mechanisms of the pathogenesis of au- pathogenesis and therapeutic strategies for type I IFNs for
toimmune diseases, the effective therapies are lacking [6–8]. further studies.
In 1979, IFNs were found to be increased in autoimmune
disorders, and there is a positive correlation between the
amount of IFNs in the sera and disease activity as well as The Family and Origin of Type I Interferons
the level of antibodies to DNA in patients with SLE [9]. In
addition, after IFN-α therapy, autoimmune disorders such as The IFN family contains three groups: type I, type II, and type
autoimmune thyroid disease and SLE-like syndrome can oc- III IFNs. IFN-Is include IFN-α (13 distinct subtypes of IFN-α
cur in patients with hepatitis C virus or malignant carcinoid in humans and 14 in mice), IFN-β, IFN-δ, IFN-ε, IFN-κ,
tumors [10, 11]. Similarly, type I IFN signatures are also in- IFN-ω, and IFN-ζ [22]. Humans can express IFN-α, IFN-β,
creased in the patients with RA and dermatomyositis (DM), IFN-ε, IFN-κ, and IFN-ω [22]. The production of type I IFNs
demonstrating a role of IFNs in promoting the development of is induced when different stimuli are detected by pattern rec-
autoimmune diseases [12, 13]. ognition receptors (PRRs) on the plasma membrane in
Type I IFNs are widely expressed cytokines that are in- endosomes and the cytoplasm of aforementioned type I IFN-
volved in innate immunity and adaptive immune system and producing cells [17]. Different stimuli consist of exogenous
in particular play an important role in antiviral activity and pathogens, apoptotic cells, apoptosis-derived membrane vesi-
cancer immunosurveillance [14–16]. IFN-Is are mainly pro- cles (AdMVs), NETotic neutrophils, endogenous self-nucleic
duced by innate immune cells in mammals, such as macro- acids, and related immune complexes (ICs) [23–26]. The
phages and dendritic cells (DCs) [17]. IFN-β can be induced PRRs, such as TLRs, retinoic acid-inducible gene-I (RIG-I)-
by most types of cells, whereas IFN-α is largely produced by like receptors (RLRs), and nod-like receptors (NLRs), can be
plasmacytoid DC (pDCs) [17]. Once produced, type I IFNs divided into DNA or RNA sensors during the productions of
interact with the interferon-α/β receptor (IFNAR, including type I IFNs [2].
subunits IFNAR1 and IFNAR2) expressed in most nucleated TLR9 is an endosomal membrane sensor for DNA stimuli
cells, and ultimately initiate the transcription of interferon- and is mainly expressed in pDCs, macrophages, B cells [24,
stimulated genes (ISGs) to produce type I IFN gene signature 25, 27, 28]. Extracellular stimuli containing nucleic acids can
through JAK/signal transducers and activators of transcription be internalized by FcγRIIa on the surface of pDCs to form
(STAT) pathway [14, 17, 18]. Many molecules encoded by endosomes [23–26]. TLR9 in the endoplasmic reticulum (ER)
ISGs induce antiviral and antitumor activity but may also play can traffic into the endosome and deliver extracellular DNA
a role in autoimmunity [12]. signals. Upon recognizing unmethylated CpG DNA released
With more attention on the significance of type I IFN sig- by the above complexes, TLR9 interacts with myeloid differ-
naling pathway in autoimmune diseases, many monoclonal entiation primary response gene 88 (MyD88). MyD88 then
Clinic Rev Allerg Immunol

recruits and activates interleukin-1 receptor-associated kinase IFNARs, and its subunits IFNAR1 and IFNAR2 separately
4 (IRAK4), triggering IRAK1, tumor necrosis factor receptor- bind to and activate different types of Janus kinase (JAK)
associated factor 6 (TRAF6), and TRAF3 pathway to activate protein families: IFNAR1 activates tyrosine kinase 2
interferon regulatory family 7 (IRF7), which can translocate to (TYK2) and IFNAR2 activates JAK1 [15]. Later, cytoplasmic
the nucleus and initiate transcription of IFN-Is [23–26]. transcription factors STAT1 and STAT2 can be phosphory-
Cyclic GMP-AMP synthase (cGAS) is expressed in most lated by JAKs, and interferon regulatory family 9 (IRF9) can
cells and is the most important intracellular DNA sensor dur- bind to STAT1/STAT2 heterodimers to form the
ing the production of IFN-Is [25, 29, 30]. When activated by heterotrimeric complex IFN-stimulated gene factor 3
double-stranded (ds) DNA, cGAS initiates the synthesis of (ISGF3), which can initiate transcription of ISGs by combin-
cyclic dinucleotide second messenger (cGAMP) and then ac- ing with IFN-stimulated response elements (ISREs, consensus
tivates stimulator of interferon genes (STING) in the ER [29]. sequence TTTCNNTTTC) [15, 17]. ISGs include a series of
STING then recruits TANK-binding kinase 1 (TBK1) to genes encoding antiviral and antitumor molecules, and tran-
phosphorylate IRF3, which can translocate to the nucleus to scription factors such as IRFs [12] (Fig. 1). Notably, IRF7
promote IFN-β production [29]. Other intracellular receptors promotes the expression of ISGs that also include IRF7,
for DNA stimuli include interferon-induced protein 16 forming a positive feedback loop within the type I IFN signal-
(IFI16), Z-DNA-binding protein 1 (ZBP1, also known as ing pathway [38].
DAI), and DNA-dependent RNA polymerase III [25, 30, 31].
TLR3, TLR7, and TLR8 are endosomal membrane recep-
tors expressed in many innate immune cells, and TLR7 is The Regulation of the Type I Interferon
particularly highly expressed in pDCs [24, 27, 28, 32, 33]. Signaling Pathway
TLR3 can detect dsRNA in endosomes internalized by
FcγRIIa, and TLR7 and TLR8 can detect single-stranded The Interferon Regulatory Family
(ss) RNA in endosomes [24, 33]. After activation, TLR3 can
bind to Toll-IL-1 receptor domain-containing adaptor induc- Interferon regulatory family (IRF) family is the regulator of
ing IFN-β (TRIF) to activate TRAF3 and then recruit and IFN signaling pathways and includes 9 members in human:
activate TBK1 to phosphorylate IRF3 for IFN-β production IRF1, IRF2, IRF3, IRF4 (PIP/LSIRF/ICSAT), IRF5, IRF6,
[34]. Currently, cyclin-dependent kinases (CDKs) have been IRF7, IRF8 (ICSBP), and IRF9 (ISGF3γ) [14]. IRF proteins
found to be critical in the translation of IFN-β, and CDK contain conserved amino-terminal DNA-binding domains
inhibition dramatically suppresses the transcription of ISGs (DBDs), carboxy-terminal IRF-association domain 1 (IAD1)
[35]. Activated TLR7 and TLR8 induce the transcription of and IAD2 [14]. The DBDs can detect and regulate ISRE, and
IFN-Is in the same manner as that of TLR9 [34]. the IADs can mediate homodimers and heterodimers with
RLRs include RIG-I, melanoma differentiation-associated other IRFs and transcription factors [14].
gene 5 (MDA5), and laboratory of genetics and physiology 2 The type I IFN signaling pathway begins with IRF3 being
(LGP2) [33]. RIG-I and MDA5 are expressed in cytoplasm of phosphorylated via signaling from PRRs, which leads to ex-
fibroblasts, cDCs, macrophages [33, 35]. RIG-I detects short pression of IFN-β and IFN-α4 through dimerization, nuclear
ds-5′ tri-phosphorylated RNA and ss-5′ tri-phosphorylated translocation, and formation of a multiprotein complex with
RNA, whereas MDA5 can recognize long dsRNA and coactivators CREB binding protein (CBP) and p300 [39].
ssRNA [33, 35, 36]. After activation, RIG-I and MDA5 bind IRF7 is poorly expressed in multiple cells but can be strongly
to the adaptor mitochondrial antiviral signaling protein induced by the type I IFN signaling pathway [38]. Type I IFNs
(MAVS) on the outer membrane of mitochondria and then induced by IRF3 then initiate the transcription and expression
activate IRF3 through TRAF3 and TBK1 [37]. TLR4 can of ISGs that include IRF7, initiating a positive feedback loop
sense extracellular lipopolysaccharide (LPS) on the cell mem- of IRF7 [12]. At the later phase, both IRF3 and IRF7 can
brane. Activated TLR4 recruits TRAM and then activates promote the expression of IFN-Is [39].
TRIF, TBK1, and IRF3 to initiate the transcription of IFN-Is IRF1, IRF5, IRF8, and IRF9 also participate in the regula-
[34]. tion of type I IFN signaling pathways [38]. IRF9 is a part of
ISGF3 that is necessary for the transcription of ISGs [15, 17].
IRF8 has been proved to cooperatively promote the rapid pro-
Type I Interferon Signaling Pathway duction of IFN-β induced by IRF3 in human monocytes [40].
IRF1 and MyD88 are required for the expression of IFN-β in
IFN-α and IFN-β exert their immune functions by ligating cDCs but not pDCs [41]. IRF5 promotes the transcriptions of
with their specific IFNARs that are expressed on the plasma specific ISREs that are mainly for encoding proinflammatory
membrane of most nucleated cells [18]. The interactions be- cytokines [38, 39]. Additionally, IRF4 and IRF8 are essential
tween IFNAR and IFN-α/β drive the dimerization of for DC subset development [39].
Clinic Rev Allerg Immunol

Post-Translational Modifications of the Type I control non-derepressible 5 (GCN5) [82]. The GCN5/PCAF
Interferon Signaling Pathway complex is recruited to acetylate nucleosomes to accelerate the
transcription of IFN-β through recruiting SWItch/sucrose
Post-translational modifications (PTMs), including phosphor- non-fermentable (SWI/SNF) to bind to transcription initiation
ylation, polyubiquitylation, methylation, acetylation, factor IID (TFIID) [83].
ISGylation, and SUMOylation, are emerging mechanisms Histone deacetylase 1 (HDAC1) and HDAC8 can in-
for regulating the type I IFN signaling pathway to maintain hibit the production of IFN-β by inducing histone H3/H4
the balance of the production of IFN-Is and transcription of deacetylation of the Ifnb1 promoter [84]. However,
ISGs [17, 42]. PTMs of type I IFN signaling pathway are HDAC has also been shown to promote the initiation of
summarized in Fig. 2 and Table 1. transcription or transcription elongation of ISGs by
recruiting P-TEFb to ISG promoters through Brd4 [85].
Epigenetic Modifications in the Regulation of the Furthermore, small ubiquitin-like modifier (SUMO) can
Type I Interferon Signaling Pathway silence Ifnb1 at its promoter through sumoylation [86].
In addition, PARP9-DTX3L is also an E3 ubiquitin ligase
Epigenetic modifications generate hereditary alterations in and can bind to STAT1 to act on host histone H2BJ to
gene expression without changing the DNA coding sequence promote the expression of ISGs [42, 87]. The human
[71]. Epigenetic modifications, including DNA methylation, Bre1/RNF20 complex is necessary to promote global in-
histone modifications, microRNAs (miRNAs), and long non- creased monoubiquitination of histone 2B at lysine 120 to
coding RNAs (lncRNAs), are important in regulating the type activate the expression of ISGs [88].
I IFN pathway [71]. miRNA and lncRNAs are also important in regulating
DNA methylation is involved in heritable DNA modifica- type I IFN signaling pathways. miR-466l can directly sup-
tion at the 5′-position of cytosine rings residing in the dinu- press multiple IFN-α species via their 3′ untranslated re-
cleotide sequence CpG [72]. Decreased DNA methylation in gion (3′UTR) [89]. miR-130b negatively regulates IFN-α
IFN-induced genes have been found in patients with SLE, in renal cells in a mouse model of lupus nephritis, whereas
compared to healthy controls [73]. Several different methyla- miR-618 can increase IFN-α production in patients with
tion positions and genes have been identified in patients with SSc [90, 91]. miR-146a can suppress IFN-β production
RA, including MX dynamin-like GTPase 1 (MX1), IFN- and miRNA-302d can suppress ISG expression by
induced protein 44-like (IFI44L), Deltex E3 Ubiquitin targeting IRF9 [92, 93]. miR-279 can inhibit type I IFN
Ligase 3L (DTX3L), and polymerase family member 9 signaling directly via inhibiting STAT [94]. miR-155 can
(PARP9), forming the IFN-inducible gene interaction network inhibit suppressors of cytokine signaling 1 (SOCS1) and
[74]. In addition, hypomethylation of IFN-regulated genes, promote the phosphorylation of STAT1 and STAT3 in
including MX1, IFI44L, and PARP9, have also been identified hepatocarcinoma cells [95]. Controversially, type I IFN
in patients with prim ary SjS (pSjS) [75]. Many signaling was found to be enhanced in miR-155-deficient
hypomethylations of type I IFN-associated genes have been CD8+ T cells [96]. In addition, miR-221 and miR-203 can
identified in CD4+ and CD8+ methylated regions in patients activate JAK–STAT pathway by inhibiting SOCS1 and
with SSc [76]. Furthermore, inhibition of DNA methylation SOCS3 [97]. The miR-21 negatively regulates IFN-α sig-
can induce type I IFN responses in cancer, demonstrating naling through targeting MyD88 and IRAK1 in patients
negative regulatory functions of DNA methylation in type I with HCV infection [98]. Furthermore, lncRNA#32 has
IFN pathway [77]. been shown to upregulate ISG expression and the antiviral
Histone modifications include methylation, acetylation, functions of type I IFN [99]. lncLrrc55-AS can promote
ubiquitination, and phosphorylation [78]. The dimethylation the production of IRF3 and IFN-I [100]. However,
of histone H3 at lysine 9 (H3K9me2) has an inhibitory effect lncRNA-CMPK2 has been identified as a negative regula-
on IFN and ISG expression [79]. Methyltransferase SET tor of the type I IFN signaling pathway [101].
domain-containing protein 2 (SETD2) can catalyze the
trimethylation of H3K36 at certain ISG promoters, such as
ISG15, to activate them, and SETD2 can also amplify the Susceptibility Genes in the Type I Interferon
functions of IFN-Is by inducing STAT1 methylation [80]. Signaling Pathway in Autoimmune Diseases
Disruption of telomeric silencing-1 like (DOT1L) protein is
a methyltransferase that can mediate the modification of Several susceptibility genes have been found to be related to
H3K79me2/3 at Ifnb1 promoters, promoting the transcription the production of type I IFN or type I IFN signaling pathway
and expression of IFN-β [81]. The transcription of IFN-β can in autoimmune diseases. Susceptibility genes of SLE associ-
also be regulated by the histone acetyltransferase proteins ated with type I IFNs include IRF5, IRF7, IRF8, IRAK1, tu-
CBP and p300/CBP-associated factors (PCAF)/general mor necrosis factor, alpha-induced protein 3 (TNFAIP3),
Clinic Rev Allerg Immunol

LPS
AdMV TLR4
Apoptotic cell
FcγRIIa
Virus TRAM TRIF

IFN-α
dsDNA
Endosome
cGAS IFN-β

RIG-1

ssRNA P STAT2
TLR3
STAT1 P
cGAMP ISGF3
TRIF TLR7 TLR8 TLR9 IRF9
MAVS STING

TRAF3 Endoplasmic TRAF3 MyD88


reticulum
IRAK4 IRAK1 TRAF6

TBK1 TRAF3

IRF7 ISGs
Ribosome
CDK transcription
IRF3

IRF7
IRF3
IFN-Is
transcription

Fig. 1 The production of type I interferons and type I interferon TYK2 and JAK1 bind to IFNAR1 and IFNAR2, respectively. Type I
signaling pathway. Extracellular stimuli containing DNA can be IFN signals then pass to the inner side of cells via the JAK–STAT sig-
internalized to form endosomes by FcγRIIa on the surface of pDCs, naling pathway. Later, IFNAR1 can be phosphorylated by JAKs, leading
and then recognized by TLR9 to activate MyD88. MyD88 recruits to repositioning and binding of STAT2. Phosphorylated STAT2 can re-
IRAK4 and leads, through a pathway involving IRAK1, TRAF6, and cruit and phosphorylate STAT1 to form STAT1/STAT2 heterodimers
TRAF3, to activation of IRF7, which can translocate into the nucleus and separate from IFNAR. The heterotrimeric complex ISGF3 comprises
and initiate transcription of IFN-Is. cGAS can be activated by IRF9 and STAT1/STAT2 heterodimers to initiate transcription of ISGs
intracellular dsDNA. cGAS phosphorylates IRF3 to translocate into the by combining with ISREs. AdMV: apoptosis-derived membrane vesicle;
nucleus and induce IFN-β production through the cGAMP/STING/ RIG-I: retinoic acid-inducible gene-I; MAVS: mitochondrial antiviral
TBK1 pathway. CDKs are very critical in the translation of IFN-β. signaling protein; MDA5: melanoma differentiation-associated gene 5;
TLR3 can detect dsRNA and then activate IRF3 to induce IFN-β produc- TRAF3: tumor necrosis factor receptor-associated factor 3; TBK1:
tion by activating TRIF, TRAF3, and TBK1. TLR7 and 8 can recognize TANK-binding kinase 1; IRF3: interferon regulatory family 3; cGAS:
ssRNA in endosomes and then bind to MyD88 and interact with IRAK4, cyclic GMP-AMP synthase; cGAMP: cyclic dinucleotide second mes-
IRAK1, TRAF6, and TRAF3 to activate IRF7 to induce the transcription senger; STING: stimulator of IFN genes; LPS: lipopolysaccharide; TLR:
of IFN-Is. RIG-I and MDA5 mainly recognize dsRNA and then bind to Toll-like receptor; TRAM: TRIF-related adaptor molecule; TRIF: TIR-
the adaptor MAVS on the outer membrane of mitochondria, and MAVS domain-containing adaptor protein-inducing IFN-β; MyD88: myeloid
then activates IRF3 through TRAF3 and TBK1. TLR4 can sense LPS on differentiation primary response gene 88; IRAK1: Interleukin-1 recep-
the cell membrane. Activated TLR4 recruits TRAM and then activates tor-associated kinase 1; IFN: interferon; CDK: cyclin-dependent kinase;
TRIF, TBK1, and IRF3 to initiate the transcription of IFN-Is. The inter- IFNAR1: interferon-α/β receptor 1; JAK: Janus kinase 1; TYK2: tyro-
action between IFN-α/β and IFNARs drives the dimerization of IFNARs, sine kinase 2; STAT1: signal transducers and activators of transcription 1;
leading to the autophosphorylation and transphosphorylation of JAKs. ISGF3: IFN-stimulated gene factor 3; ISG: interferon-stimulated gene

TNFAIP3 interacting protein 1 (TNIP1), interferon induced the MHC region [103, 104]. A variant of STAT4 (rs7574865)
with helicase C domain 1 (IFIH1), TYK2, STAT4, protein is related to elevated sensitivity to IFN-α signaling [103].
tyrosine phosphatase non-receptor type 22 (PTPN22), three STAT4 protein can be phosphorylated and activated by
prime repair exonuclease 1(TREX1), E26 transformation- interacting with IFNAR2 cytoplasmic domain, and then trans-
specific 1 (ETS1), and ubiquitin-conjugating enzyme E2 L3 locate into nucleus to initiate the expression of IFN-γ and
(UBE2L3) [102–107]. other proinflammatory molecules [108–110]. PTPN22 is the
Large-scale association studies have shown a relationship susceptibility gene for both SLE and RA in the European
between polymorphisms of IRF5, IRF7, IRF8, and SLE risk population [103, 105]. High IFN-α and low TNF-α in serum
[102]. Several variants of IRF5, IRF7, and IRF8 can induce occur in patients with SLE with the rs2476601 risk allele of
transcription and expression of proteins in regulating the type PTPN22 [103, 105]. PTPN22 protein can directly interact
I IFN signaling pathway [102]. Notably, IRF5 and STAT4 are with and promote the ubiquitination of TRAF3 [111].
considered the most strongly SLE-associated loci except for TNFAIP3 and TNIP1, which are proven to be susceptibility
Clinic Rev Allerg Immunol

Fig. 2 The post-translational modifications (PTMs) of type I interferon IFN genes; TLR: Toll-like receptor; TRAM: TRIF-related adaptor mole-
pathway. The gray boxes represent the negative regulators of type I IFN cule; TRIF: TIR-domain-containing adaptor protein-inducing IFN-β;
signaling pathway, and the orange arrows shows the path of type I IFN MyD88: myeloid differentiation primary response gene 88; IRAK1:
pathway and the blue one represents the negative webs. IFI35, RNF125, Interleukin-1 receptor-associated kinase 1; IFNAR1: interact with inter-
NLRC5, PCBP2, CYLD, TRIM13, and TRIM40 can negatively regulate feron-α/β receptor 1; JAK: Janus kinase 1; TYK2: tyrosine kinase 2;
the type I IFN pathway through sensing of RNA via RIG-I or MDA5 and STAT1: signal transducers and activators of transcription 1; ISGF3:
signaling MAVS. A20, TRIP, TRIM8, TRIM11, TRIM26, TRIM27, IFN-stimulated gene factor 3; ISG: interferon-stimulated gene; IFI35:
TRIM56, and SHP-2 can suppress TBK1, and NLRC5, TRIM26, interferon-induced protein 35; RNF125: ring-finger protein 125;
TRIM27, TRIM56, PLASy, ISG56, ASCC3, RAUL, SMAD2, and NLRC5: NLR family CARD domain-containing 5; PCBP2: poly(rC)-
SMAD3 can inhibit IRF3. TRIM38 can induce SUMOylation of cGAS binding protein 2; CYLD: cylindromatosis lysine 63 deubiquitinase;
and STING to inhibit type I IFN response, and TRIM32 and TRIM56 can TRIP: TNF receptor-associated factor (TRAF)–interacting protein;
induce polyubiquitination of STING. The TRIF can be negatively regu- TRIM: tripartite motif-containing; GBP4: guanylate binding protein 4;
lated by TRIM32, TRIM38, SHP-2, and TRAF3 and TRAF6 through OTUB1: OTU domain-containing ubiquitin aldehyde binding protein 1;
TLRs signaling can be inhibited by TRIM38, OTUB1, OTUB2, and DUBA: deubiquitinating enzyme A; PLAS: protein inhibitor of activated
DUBA. GBP4 can disrupt the interactions between TRAF6 and IRF7, STAT; ISG56: IFN-stimulated gene 56; IFIT1: interferon-induced protein
and IRF7 can be directly suppressed by PLASy, ISG56, Nmi, OASL1, with tetratricopeptide repeats 1; Nmi: N-Myc and STATs interactor;
DDX24, ASCC3, and RAUL. The production of IFN-β can be directly OASL1: oligoadenylate synthetase-like 1; DDX24: DEAD-box helicase
inhibited via TRIP, ISG56, SMAD2, and SMAD3. PKD2 can regulate 24; ASCC3: activating signal cointegrator complex 3; RAUL: RTA-
ubiquitination and degradation of IFNAR1, and USP18 can compete with associated ubiquitin ligase; SMAD2: SMAD family member 2; PKD2:
JAK1 for IFNAR2. SHP-1, SHP-2, PTPN1, TC-PTP, SOCS1, SOCS3, protein kinase D2; USP18: ubiquitin-specific peptidase 18; SHP-1: SH2-
PLAS1, PLASy, C1S, and DCST1 can suppress the JAK–STAT pathway containing protein tyrosine phosphatase 1; PTPN1: protein tyrosine phos-
by inhibiting JAKs or STATs. RIG-I: retinoic acid-inducible gene-I; phatase, non-receptor type 1; PTP1B: protein tyrosine phosphatase 1B;
MAVS: mitochondrial antiviral signaling protein; MDA5: melanoma TC-PTP: T cell-protein tyrosine phosphatase; PTPN2: protein tyrosine
differentiation-associated gene 5; TRAF3: tumor necrosis factor phosphatase, non-receptor type 2; SOCS: suppressors of cytokine signal-
receptor–associated factor 3; TBK1: TANK-binding kinase 1; IRF3: in- ing; CIS: cytokine-induced SRC-homology 2 (SH2) protein; DCST1:
terferon regulatory family 3; cGAS: cyclic GMP-AMP synthase; DC-STAMP domain-containing 1
cGAMP: cyclic dinucleotide second messenger; STING: stimulator of
Clinic Rev Allerg Immunol

Table 1 Negative regulators in the type I IFN signaling pathway

Negative regulator Mechanism Ref

IFI35 (also called IFP35) Inhibiting dephosphorylation, promoting K48-linked ubiquitination and [43]
proteasomal degradation of RIG-I
RNF125 Conjugating ubiquitin to and suppressing MDA5/MAVS/RIG-I [44]
NLRC5 Blocking the interactions between MAVS and RIG-I and IRF3 phosphorylation [45]
A20 Blocking IRF signaling induced by RIG-I and the IFN response by removing [46]
K63-linked ubiquitin chains from TBK1/IKKi
PCBP2 Interacting with and promoting the proteasomal degradation of MAVS to inhibit [47]
the RIG-I/MAVS pathway
CYLD Removing K63-linked polyubiquitin chains from RIG-I [48]
TRIP Promoting K48-linked polyubiquitination and proteasomal degradation of [49]
TBK1;
Inhibiting IFN-β production promoted by TLR3, TLR4 and RIG-I
TRIM family TRIM 8 Disrupting the TRIF-TBK1 interaction [50]
TRIM 11 Interacting with TBK1 to inhibit the functions of IRF3 and IFN-β production [50]
TRIM 13 Suppressing MDA5-induced IFN-Is; upregulating RIG-I [50]
TRIM 26 Inhibiting RNA virus infection by TBK1; [50]
Promoting K48-linked polyubiquitination and proteasome-mediated degradation
of phosphorylated IRF3
TRIM 27 Targeting TBK1 and inhibiting the expression of IRF3 [50, 51]
TRIM 32 Inducing K63-linked polyubiquitination of STING; [50]
Synthesizing unanchored polyubiquitin chains to activate NEMO, IKKβ, and
TBK1 to induce IRF3 and NF-κB activation;
Promoting the degradation of TRIF to suppress TLR3/4 responses
TRIM 38 Inducing SUMOylation of cGAS and STING to prevent K48-linked [50]
polyubiquitination and proteasomal degradation;
Negatively affecting TLR signaling by destabilizing TRAF6, TRIF, TAB2/3 and
NAP1
TRIM 40 Promoting K27- and K48-linked polyubiquitin to RIG-I and MDA5 and [50]
proteasomal degradation
TRIM 56 Inducing K63-linked polyubiquitination of STING; [50]
Synthesizing unanchored polyubiquitin chains to activate NEMO, IKKβ and
TBK1 to induce IRF3 and NF-κB activation
GBP4 Disrupting the interaction between TRAF6 and IRF7 [52]
OTUB1 and OTUB2 Deubiquitinating TRAF3 and TRAF6 [48]
DUBA (also called OTUD5) Suppressing K63-specific autoubiquitination of TRAF3 [48]
PIAS PIASy targets IRF3 and IRF7 to negatively regulate IFN-Is expression; [53, 54]
PIAS1 and PIASy inhibit STAT1
ISG56 (also called IFIT1) Inhibiting Sendai virus (SeV)-activated IRF3, IFN-β promoter; [55, 56]
Facilitating interaction of SAP25-HDAC2-Sin3A at LPS-induced gene loci, to
inhibit activation at promoters of IFNB1, IRF7, IRF8
Nmi Promoting the K48-linked ubiquitination and proteasomal degradation of IRF7 [57]
OASL1 Inhibiting translation of IRF7 mRNA [58]
DDX24 Disrupting the IRF7/RIP1 interaction to suppress RLR activity; [59]
possibly competing with RIG-I for binding of RNA
ASCC3 Dampening ISG expression by interacting with IRF-3 and IRF-7 [60]
RAUL Ubiquitinating IRF7 and IRF3 to suppress the type I IFNs signature [61]
SMAD2 and SMAD3 Inhibiting IRF3 phosphorylation and transcription by directly interacting to [62]
reduce IFN-β expression
PKD2 Regulating ubiquitination and degradation of IFNAR1 [63]
USP18 Competing with JAK1 for binding IFNAR2 [64]
SHP-1 Dephosphorylating JAKs; [65, 66]
Inhibiting activation of the kinase IRAK1 to promote type I IFN signatures
SHP-2 Inhibiting signal transduction of TBK1 and TRIF to suppress TLR4- and [67]
TLR3-activated IFN-β;
Inhibiting JAK–STAT signaling
PTPN1 (also called PTP1B) Dephosphorylating JAK2 and TYK2 [68]
TC-PTP (also called PTPN2) Modulating JAK1 and JAK3 [68]
Clinic Rev Allerg Immunol

Table 1 (continued)

Negative regulator Mechanism Ref

SOCS SOCS1 Inhibiting the catalytic activity of JAKs by interacting with the JAK activation [69]
loop
SOCS3 Inhibiting JAKs through receptor binding and JAK activation loop; Competing
for receptor phosphotyrosine-docking sites
CIS Blocking the recruitment and activation of STATs
DCST1 Promoting ubiquitination-mediated degradation of STAT2 [70]

IFI35 interferon-induced protein 35, RNF125 ring-finger protein 125, NLRC5 NLR family CARD domain-containing 5, PCBP2 poly(rC)-binding
protein 2, CYLD cylindromatosis lysine 63 deubiquitinase, TRIP TNF receptor-associated factor (TRAF)–interacting protein, TRIM tripartite motif-
containing, GBP4 guanylate binding protein 4, OTUB1 OTU domain-containing ubiquitin aldehyde binding protein 1, DUBA deubiquitinating enzyme
A, PLAS protein inhibitor of activated STAT, ISG56 IFN-stimulated gene 56, IFIT1 interferon-induced protein with tetratricopeptide repeats 1, Nmi N-
Myc and STATs interactor, OASL1 oligoadenylate synthetase-like 1, DDX24 DEAD-box helicase 24, ASCC3 activating signal cointegrator complex 3,
RAUL RTA-associated ubiquitin ligase, SMAD2 SMAD family member 2, PKD2 protein kinase D2, USP18 ubiquitin-specific peptidase 18, SHP-1
SH2-containing protein tyrosine phosphatase 1, PTPN1 protein tyrosine phosphatase, non-receptor type 1, PTP1B protein tyrosine phosphatase 1B, TC-
PTP T cell-protein tyrosine phosphatase, PTPN2 protein tyrosine phosphatase, non-receptor type 2, SOCS suppressors of cytokine signaling, CIS
cytokine-induced SRC-homology 2 (SH2) protein, DCST1 DC-STAMP domain-containing 1

genes of SLE through GWAS, encode A20 and A20-binding Type I Interferon Signaling Pathway
protein, respectively, and are regulators in type I IFN pathway, and Autoimmune Diseases
NF-κB activation, and TNF-mediated apoptosis [102–104].
TREX1 encodes 3′-5′ DNA exonuclease TREX1 to clear Type I Interferonopathies
aberrant DNA and is a susceptibility gene of SLE [103, 105].
IFIH1 encodes the dsRNA sensor MDA5 [102]. A missense Type I interferonopathy is a type of Mendelian genetic disor-
allele of IFIH1 (rs1990760) is identified to be related to in- der with diverse molecular defects and clinical features but
creased transcription and expression of IFIH1 and is involved with the same overproduction of type I IFNs [118, 119].
in elevated expression of IFN-induced genes in anti-dsDNA- Type I interferonopathies include a wide range of genetic dis-
positive patients with SLE [102, 104]. TYK2 encodes TYK2, a eases, such as Aicardi-Goutières syndrome (AGS), Sting-
member of the JAK–STAT pathway [105]. UBE2L3 has associated vasculopathy with onset in infancy (SAVI),
genome-wide significance in relation to SLE in Asian but spondyloenchrondro-dysplasia (SPENCD), Singleton-
not European populations [103]. UBE2L3 encodes an E2 Merten syndrome, and ISG15 deficiency [118, 119]. Type I
ubiquitin-conjugating enzyme that can interact with Triad3A interferonopathies usually occur in childhood and some of
(also called RNF216) to regulate the degradation of TLR4 and them possess lupus-like features, supporting the association
TLR9 [112]. IRAK1 acts on the delivery of TLR signaling, between lupus and high level of IFN-Is [108]. Type I
and IRAK1 gene polymorphisms were identified to be related interferonopathies can be caused by mutations in multiple
to the pathogenesis of SLE [106, 107]. ETS1 is a negative genes associated with type I IFN signaling pathway, such as
regulator in expression of IFN-induced genes by interacting TREX1, ribonuclease H2 subunit A (RNASEH2A),
with ISGs [102]. ETS1 is an SLE susceptibility locus in Asia, RNASEH2B, RNASEH2C, SAM, and HD domain-
and the SNP rs6590330 is related to decreased expression of containing deoxynucleoside triphosphate
ETS1 [102, 105]. triphosphohydrolase 1 (SAMHD1), adenosine deaminase
Many SLE risk susceptibility genes involved in the type I RNA specific (ADAR1), and IFIH1 [119]. Type I
IFN pathway also participate in the pathogenesis of other au- interferonopathies are summarized in Table 2.
toimmune diseases, such as RA, JIA, SSc, and SjS. Due to the poor response of biologic disease–modifying
Susceptibility genes in RA related to the type I IFN pathway antirheumatic drugs (DMARDs) in type I interferonopathies,
include PTPN22, STAT4, TNFAIP3, IRF5, UBE2L3, ETS1, there is increasing interest in targeted therapies related to the
IRF8, TYK2, IRAK1, and TRAF6 [105, 113]. IRF5, type I IFN signaling pathway [137]. JAK inhibition has shown
PTPN22, STAT4, TYK2, and UBE2L3 are associated with an promising results in type I interferonopathies [137–140]. Case
increased risk of JIA [114, 115]. Susceptibility genes in SSc reports of JAK inhibitors, such as baricitinib, ruxolitinib to
include IRF5, IRF7, IRF8, PTPN22, STAT4, TNFAIP3, treat ubiquitin-specific peptidase 18 (USP18) deficiency,
TNIP1, and TYK2, while IRF5, STAT4 TNFAIP3, and SAVI, AGS, and TREX1 chilblain lupus, have been published
TNIP1 are involved in SjS [116, 117]. The association be- [136–142]. Cases of chilblain lupus have also shown a good
tween susceptibility genes and type I IFN signaling pathway response to tofacitinib therapy [127]. Furthermore, in the
is described in Fig. 3. NCT01724580 and NCT02974595 programs, 18 patients
Clinic Rev Allerg Immunol

with CANDLE or SAVI treated with baricitinib also had good suppressed after MyD88 and TRIF inhibitors in PBMC of
results [137]. The side effects of JAK inhibition seem to be JSLE patients [158]. However, more clinical research on in-
mild and acceptable, with slight and reversible decreases in hibitors involved in type I IFN pathway is urgently needed
lymphocytes and platelets, elevated transaminases and serum [157, 158].
creatinine, and herpes zoster infection [143, 144]. However, With better understanding of the role of IFN-I, many prom-
due to the extremely low incidence of type I ising treatments targeting type I IFN signaling pathway are
interferonopathies, conducting clinical trials is difficult. undergoing clinical studies in SLE [159]. Rontalizumab, a
In vitro experiments or the use of animal models may help humanized IgG1 anti-IFN-α mAb, has an acceptable safety
identify additional JAK–STAT candidates. profile and induces decreased expression of IRGs in phase I
studies of patients with SLE [160, 161]. However, due to the
Systemic Lupus Erythematosus and Pediatric unmet endpoints in phase II trials, the development of
Systemic Lupus Erythematosus rontalizumab was terminated [160]. Sifalimumab (formerly
MEDI-545) is a fully humanized IgG1 anti-IFN-α mAb
In 1999, genes induced by IFN-Is were found to be upregu- [162]. In a phase IIb trial in 2016, SLE patients treated with
lated in peripheral blood mononuclear cells (PBMCs) of ap- sifalimumab met the primary endpoint and their other ob-
proximately two-thirds of adult patients with SLE through served indexes and symptoms were also significantly im-
microarray gene expression analysis [145]. The level of type proved [162]. AGS-009 is a humanized IgG4 anti-IFN-α
I IFNs is also positively related to the disease activities and mAb that was shown to be generally tolerated by patients with
severity of symptoms (such as skin rash, fever, and leucope- SLE [163]. However, there were no subsequent trials.
nia) in patients with SLE [146]. In addition, ANA and anti- Interferon-α-kinoid (IFN-K) is a therapeutic vaccine targeting
dsDNA antibodies appear in patients after treatment with IFN- IFN-α [164, 165]. In a phase IIb trial, IFN-K significantly
Is in patients with malignant carcinoid tumors [11]. Moreover, decreased IFN gene signatures and achieved the biological
TLRs have been identified to be crucial in the maintaining coprimary endpoint but not the clinical coprimary endpoint,
SLE [147]. revealing a need for further evaluation [165].
IFN-α is a powerful inducer for the differentiation or acti- An anti-IFNAR1 mAb, anifrolumab, showed significant
vation of DCs [148]. The activation of T cells and the sup- effects in decreasing the disease activity of SLE in phase II
pression of Treg cells can be induced by IFN-Is in lupus-prone trials, and another phase IIb study in patients with moderate to
mice after UVB exposure [149]. IFN-Is participate in early severe SLE was considered very successful [166, 167]. Two
transitional B cells and promote the expansion of phase III trials (known as TULIP–1 and TULIP–2) in patients
autoantibody-secreting cells [150]. In patients with SLE, with SLE were completed in 2019. One of the endpoints,
IFN-Is can lead to a strong inflammatory response by sup- British Isles Lupus Disease Activity Group (BILAG)–based
pressing the maturation of miR-146a [151]. IFN-β is in- Composite Lupus Assessment (BICLA), but not SLE re-
creased in circulating B cells in SLE patients, especially those sponse index (SRI), was met in the first phase III trial,
with nephritis and autoantibodies [152]. Moreover, increased TULIP-1, and both were achieved in TULIP-2 [20, 168].
IFN-κ promotes the development of CLE after ultraviolet light Regardless of the seemingly conflicting results, anifrolumab
exposure [146, 153, 154]. is the most successful and promising IFN pathway-targeted
pSLE or childhood-onset SLE (cSLE) occurs during child- therapy for SLE and awaits use in clinical practice [20, 168].
hood with more severe damage to internal organs (especially BIIB059 is a mAb that binds to blood DC antigen 2
to kidney) than adult SLE, revealing its stronger association (BDCA2), an inhibitory receptor on pDCs, and induces its
with genetics [155]. The lupus-like phenotypes in children, rapid internalization to inhibit the production of IFN-Is
also called “monogenic lupus,” is one of type I [169]. BIIB059 was seemingly beneficial to patients with
interferonopathies and can be caused by mutation of genes SLE with cutaneous manifestations [169]. Venetoclax (also
involved in type I IFN signaling pathway, such as called ABT-199) is a Bcl-2 antagonist that can kill pDCs in
DNase1L3, TREX1, SAMHD1, RNASEH2ABC, ADAR1, lupus-prone mice and has shown a good clinical response in
IFIH1, ISG15 [130, 155]. In a study assessing type I IFN patients with SLE in a phase I trial [170–172].
signaling in pediatric autoimmune diseases, 82% (64 of 78) Hydroxychloroquine, a TLR9, TLR7, and TLR8 antago-
of samples from patients with juvenile SLE (JSLE) and 75% nist, is a conventional antimalarial drug for SLE [6]. The pre-
(76 of 101) of samples from children with juvenile DM (JDM) clinical data of CPG-52364, an inhibitor of TLR7, TLR8, and
had positive IFN scores [156]. Upregulated expression of TLR9, showed that it can enhance the effectiveness of
ISGs was also observed in cSLE, and it can be suppressed hydroxychloroquine [173]. Dynavax posted a news article
with TBK1 inhibitors BX795 [157]. The expression of about a phase 1b/2a trial of DV1179, a TLR7 and TLR9
TLRs in JSLE is also increased [158]. TLRs can be activated inhibitor, in patients with SLE, but it did not meet the phar-
by apoptotic neutrophils and the IFN-α expression can be macodynamic endpoints. A small molecular TLR9 inhibitor
Clinic Rev Allerg Immunol

Fig. 3 The susceptibility genes involved in type I interferon pathway in signaling and elevated expression of IFN-γ. TNFAIP3 and TNIP1 encode
autoimmune diseases. The red words represent the susceptibility genes A20 and A20-binding protein separately that are regulatory factors in type
involved in type I IFN pathway and some of them also represent their I IFN pathway, NF-κB activation. ETS1 can negatively regulate the ex-
encoded proteins. TREX1 encodes 3′-5’ DNA exonuclease TREX1, and pression of IFN-induced genes by interacting with ISGs. The SNP
TREX1 is related to the susceptibility of SLE by impacting the rs6590330 of ETS1 is related to the decreased expression of ETS1.
degradations and increasing the expression of IFN-Is. IFIH1 encodes MDA5: melanoma differentiation-associated gene 5; MAVS: mitochon-
dsRNA sensor MDA5 and is involved in increased transcription of drial antiviral signaling protein; TRAF3: tumor necrosis factor receptor–
IFIH1 and elevated expression of IFN-induced genes. UBE2L3 encodes associated factor 3; TBK1: TANK-binding kinase 1; IRF3: interferon
an E2 ubiquitin-conjugating enzyme that involves in the degradation of regulatory family 3; TLR: Toll-like receptor; TRAM: TRIF-related adap-
TLR4 and TLR9. IRAK1 and TRAF6 are related to the production of tor molecule; TRIF: TIR-domain-containing adaptor protein-inducing
IFN-Is and NF-κB, and the multiple SNPs with IRAK1 and TRAF6 are IFN-β; MyD88: myeloid differentiation primary response gene 88;
associated with onset and development of autoimmune diseases. PTPN22 IRAK1: Interleukin-1 receptor-associated kinase 1; IFNAR1: interact
can regulate type I IFN signatures by interacting with TRAF3, and the with interferon-α/β receptor 1; JAK: Janus kinase 1; TYK2: tyrosine
polymorphism in PTPN22 (rs2476601) is related to high serum IFN-α kinase 2; STAT1: signal transducers and activators of transcription 1;
activity. IRF5, IRF7, and IRF8 play an important role in upregulating the ISGF3: IFN-stimulated gene factor 3; ISG: interferon-stimulated gene;
type I IFN responses. Several polymorphisms of IRF5, IRF7, and IRF8 TREX1: three prime repair exonuclease 1; IFIH1: interferon induced with
are related to their expression of proteins and increased IFN-Is. TYK2 helicase C domain 1; UBE2L3: ubiquitin-conjugating enzyme E2 L3;
encodes TYK2 in JAK–STAT pathway, and a variant (rs280519) of PTPN22: protein tyrosine phosphatase non-receptor type 22; TNFAIP3:
TYK2 is related to increased production of IFN-I. STAT4 encodes tumor necrosis factor, alpha-induced protein 3; TNIP1: TNFAIP3
STAT4 protein that is associated with increased sensitivity to IFN-α interacting protein 1

ST2825 has demonstrated great potential, with promising significantly improve lesion activity in Phase II trials [177].
safety and efficacy profiles in patients with SLE [173]. For nucleic acid-targeted therapies, rhDNase seemed to be
The IRAK4 inhibitor BMS-986126, and Bruton’s tyrosine unsuccessful, whereas RSLV-132 showed good results in a
kinase (BTK) inhibitor evobrutinib (also called M2591, phase I trial and has entered a phase II trial for SLE (Table 3)
MSC2364447C) have been shown to suppress the type 1 [176].
IFN signaling pathway in preclinical experiments of SLE,
but the results of clinical trials are either lacking or unpub- Rheumatoid Arthritis
lished [174, 175]. Baricitinib (also called LY3009104) is a
selective inhibitor of JAK1/JAK2 and has shown significant Case reports show that IFN-β1, a therapy used in multiple
efficacy in patients with SLE in phase trials [21]. Tofacitinib, sclerosis (MS), can lead to the development of RA [179].
an inhibitor of JAK1/JAK3, is approved by the FDA for the Type I IFN signatures can be detected in the peripheral blood
treatment of RA, psoriatic arthritis, and ulcerative colitis of patients with RA, even in the preclinical phase [12]. Data
[176]. However, clinical trials of tofacitinib for SLE and from clinical trials show that high IFN-I signatures can predict
CLE are still in progress [176]. Filgotinib, a highly selective RA as a biomarker in at-risk individuals [180]. In addition,
JAK1 inhibitor, has just finished phase II clinical trials. R333, synovial DCs have been shown to promote the inflammatory
a topical JAK/SYK inhibitor for DLE treatment, failed to response and differentiation of Th1 and Th17 cells [7].
Table 2 Introduction of type I interferonopathies

Disease Clinical feature Gene involved Function of involved gene Ref

AGS · Autosomal recessive disorder TREX1 (OMIM 225750) Encoding 3′-5′ DNA exonuclease TREX1 [120, 121]
· Systemic inflammation occurs in infancy to clear aberrant DNA
· Leukoencephalopathy RNASEH2A (OMIM 610333) Encoding RNASEH2 subunits A, B and C, [120, 121]
· Basal ganglia calcifications RNASEH2B (OMIM 610181) respectively;
Clinic Rev Allerg Immunol

· Cerebral atrophy RNASEH2C (OMIM 610329) Complex formed by RNASEH2A, B and C


· Developmental retardation and functions as RNase H2 and can degrade
microcephaly RNA/DNA hybrids
· Lymphocytosis SAMHD1 (OMIM 612952) SAMHD1 encodes SAMHD1 protein: [120–123]
· Dystonia, seizures, and fever preventing the synthesis of cDNA by
depleting the dNTPs pool;
repairing DNA damage;
possessing nuclease activity to ssRNA
ADAR1 (OMIM 615010) Encoding ADAR (adenosine deaminase [120, 121, 124]
acting on RNA), which can edit long
dsRNA
IFIH1 (OMIM 615846) Encoding MDA5, a RNA sensor in the [120, 121, 125]
type I IFN pathway
RVCL · Autosomal dominant disorder occurring TREX1 (OMIM 192315) Encoding 3′-5′ DNA exonuclease TREX1 [120, 126]
in early adulthood to clear aberrant DNA
· Retinal vasculopathy
· Cerebral leukoencephalopathy
· Dementia
Familial chilblain lupus · Autosomal dominant disorder occurring TREX1 (OMIM 610448) Encoding 3′-5′ DNA exonuclease TREX1 [120, 127]
in early adulthood to clear aberrant DNA
· Cold-induced painful erythematous infil- SAMHD1 (OMIM 614415) Preventing the synthesis of cDNA by [123, 127]
trates in acral locations depleting the dNTPs pool;
· Similar histological findings to cutaneous Repairing DNA damage and DSB;
lupus erythematosus Possessing nuclease activity to ssRNA
SAVI · Autoinflammatory vasculopathy in lung TMEM173 (OMIM 615934) Encoding STING, the adaptor of type I IFN [128]
and skin signaling
· Necrotizing lesions on face, ears, nose,
digits
· Interstitial lung disease
SPENCD · Specific metaphyseal and vertebral ACP5 (OMIM 271550) Encoding TRAP: [129, 130]
lesions in radiology negative regulator of osteopontin and IRF7
· Immune disorders like thrombocytopenia,
thyroiditis
· Neurological symptoms like
baryencephalia, spasticity
Singleton-Merten syndrome · Early and extreme major vessels and IFIH1 (OMIM 182250) Encoding MDA5, an RNA sensor in the [125, 131]
valvular calcification type I IFN pathway
· Dental anomalies (early-onset DDX58 (OMIM 616298) Encoding RIG-I, an RNA sensor in the [131, 132]
periodontitis and root resorption) type I IFN pathway
· Osteopenia
· Acro-osteolysis
ISG15 deficiency · Basal ganglia calcification and seizures ISG15 (OMIM 61626) [133]
Clinic Rev Allerg Immunol

AGS Aicardi-Goutières syndrome, SAMHD1 SAM And HD domain-containing deoxynucleoside triphosphate triphosphohydrolase 1, DBS dried blood spots, ADAR adenosine deaminase RNA specific,

family 7, RIG-I retinoic acid-inducible gene-I, ISG15 interferon-stimulated gene 15, CANDLE chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature, PSMB8 proteasome
infancy, TMEM173 transmembrane protein 173, STING stimulator of interferon genes, SPENCD spondyloenchrondro-dysplasia, TRAP tartrate-resistant acid phosphatase, IRF7 interferon regulatory
TREX1 three prime repair exonuclease 1, MDA5 melanoma differentiation-associated gene 5, RVCL retinal vasculopathy with cerebral leukodystrophy, SAVI Sting-associated vasculopathy with onset in
Furthermore, some susceptibility genes in RA are related to
the type I IFN signaling pathway, such as IRF5, STAT4, and

[134, 135]
PTPN22 [105, 113].

[136]
Ref

Due to the unsatisfactory response to DMARDs, inhibitors


targeting the type I IFN pathway have entered clinical trials of
patients with RA. The selective JAK1/JAK3 inhibitor
tofacitinib and the selective JAK1/JAK2 inhibitor baricitinib

and IFNAR2, then downregulating type


are effective and are approved by the FDA and EMA for the
Encoding ISG15, a negative regulator of

blocking the interactions between JAK1 treatment of RA, especially for those intolerant or unrespon-
Encoding PSMB8 involved in the

sive to DMARDs [19]. Upadacitinib and filgotinib are selec-


tive JAK1 inhibitors, and both have shown good results in
Function of involved gene

the type I IFN response

patients with RA in phase II and III studies [19, 181, 182].


I interferon signatures
induction of IFN-Is

The JAK1/JAK3 inhibitor, peficitinib, has also been shown to


Encoding USP18:

be effective in patients with RA, but the data from phase III
trials are still pending [183, 184]. On the other hand,
decernotinib, a JAK3 inhibitor, did not show clear dose–
response curves and had a higher risk of toxic effects at higher
doses [185]. The JAK1/JAK2 inhibitor ruxolitinib is also a
promising candidate and is entering clinical trials [178]. JAK
inhibitors are generally tolerated in patients with RA, with a
risk of herpes zoster but not serious infections or malignancies
[186]. Currently, Syk and BTK inhibitors have also entered
PSMB8 (OMIM 256040)

clinical trials for RA [187, 188].


USP18 (OMIM 607057)

subunit β type 8, USP18 ubiquitin-specific peptidase 18, JAK1 janus kinase 1, IFNAR2 interferon-α/β receptor 2

Although INF-Is have been implicated in the pathogenesis


of RA, intra-articular injections with IFN-α and intraperitone-
Gene involved

al injection with IFN-β can prevent the occurrence or devel-


opment of RA in wild-type mice or RA mouse models [189].
IFN-β injected daily can also relieve the symptoms of RA in
rhesus monkeys [189]. However, in clinical trials, treatment
with subcutaneous recombinant IFN-β showed no improve-
ment in active patients in a phase II clinical trial (Table 4)
[190].
· Perinatal-onset intracranial Hemorrhage;

Juvenile Idiopathic Arthritis


· Atypical neutrophilic dermatosis

· Septic-shock–like presentation;
· Occurrence in early childhood

Juvenile idiopathic arthritis (JIA) is the most common pediat-


ric rheumatic disease with persistent arthritis in children or
· Mycobacterial disease

· Brain malformations;

teenagers under 16 years of age [191]. Both IFN-γ and IFN-


· Thrombocytopenia
· Liver dysfunction;

Is contribute to the pathogenesis of JIA [192, 193]. Type I


Clinical feature

· Lipodystrophy

· Calcifications;
· Hyperthermia

IFN-producing cells were identified to be enriched in synovial


fluid and the IFN-α producing cells were found in the lymph-
follicular-like structures, but the expression of IFN-α was not
upregulated in the serum and synovial fluid [193]. In
enthesitis-associated JIA, the expression of TLR2 and TLR4
was elevated in the peripheral blood and synovial fluid mono-
cytes [194]. IRF5 (rs2004640) and PTPN22 1858C > T have
been reported to be involved in an increased risk of JIA [113,
Table 2 (continued)

195].
Deficiency of USP18
CANDLE syndrome

More and more clinical studies involving inhibitors sup-


pressing the type I IFN pathway have been conducted in pa-
tients with JIA. Selective JAK inhibitors, such as tofacitinib,
Disease

have shown promising results in the treatment of JIA. In a


phase I, open-label, and multicenter study (NCT01513902)
Clinic Rev Allerg Immunol

in 2017, tofacitinib was well tolerated in patients with JIA patients with pSjS [210, 211]. In addition, a positive correla-
[196]. Tofacitinib has been shown to be effective in the treat- tion has been identified between genes induced by IFN-Is and
ment of refractory JIA in case reports [197]. Pfizer posted that titers of anti-Ro/SSA, anti-La/SSB autoantibodies, EULAR
the phase III clinical trial (NCT02592434) for tofacitinib was SjS disease activity index (ESSDAI), biological markers of
completed in 2019 with good results, supporting the use of activity, and BAFF mRNA in monocytes of patients with
tofacitinib in patients with JIA. Clinical studies involving SjS, demonstrating a key role of IFN-Is in pSjS [210, 212].
baricitinib (NCT4088396; NCT03773965; NCT03773978) pDCs, the main source of IFN-Is, have been found in the
and upadacitinib (NCT03725007) in patients with JIA are still salivary gland of patients with pSjS, whereas they are de-
in the recruiting stage, and there are no results yet. creased in the peripheral blood [210, 212–214]. This phenom-
enon also exists in SLE, and both may be caused by the re-
Juvenile Dermatomyositis translocation of pDCs from peripheral blood to diseased tissue
[210, 212].
Juvenile dermatomyositis (JDM) is a rare pediatric autoim- A study (NCT03100942) involving the JAK1 inhibitor
mune disease with an average age at disease onset of 7.5 years. filgotinib, Syk inhibitor lanraplenib, and BTK inhibitor
JDM is characterized by proximal muscle weakness and pa- tirabrutinib has been completed, but results are pending [8].
thognomonic skin rashes [198]. Overexpressed type I IFN- The JAK inhibitor filgotinib-treated mouse models with SjS
inducible transcripts and activated type I IFN signatures are showed significantly increased salivary flow rates and remark-
observed in patients with DM [13]. Similarly, serum IFN-α able reductions in lymphocytic infiltration of the salivary
activity is increased for a short time in newly diagnosed pa- gland and IFN-I signatures [215]. The JAK1/2 inhibitor
tients with JDM and is associated with high serum muscle– AG490 and ruxolitinib and the TBK1 inhibitor BX795 also
derived enzymes [199]. The expression of IFN-α/β-inducible exhibited good responses in animal models with SjS [216,
genes in muscle biopsy and proteins induced by type I IFNs 217]. On the other hand, hydroxychloroquine, an inhibitor of
such as myxovirus resistance protein A (MxA) in PBMC, TLRs, decreased IFN-I signatures but did not improve the
affected muscle and skin, are elevated in patients with JDM symptoms of SjS in a clinical trial (NCT00632866) [218].
[200–202]. TLR3 was also found to be elevated in vascular Moreover, IFN-α administered via the oromucosal route in
endothelial cells, myeloid DCs (mDCs), and regenerating patients with pSjS was demonstrated to be effective and safe
myofibers in patients with JDM [203]. Type I IFN signatures in the improvement of salivary output in a phase II clinical
are increased in B cells of JDM, and the activation of TLR7 trial [219]. However, another trial of 497 patients with pSjS
and IFN-α may lead to the expanded immature transitional B with the treatment of either IFN-α or placebo failed to meet
cell population [204]. In addition, in vivo and in vitro studies the primary endpoints [219]. Therefore, more evidence is
have shown that myogenic precursor cells are also a source of needed to further understand the role of IFN-Is in the patho-
IFN-Is [205]. genesis and treatment of SjS.
The JAK inhibitors, ruxolitinib and tofacitinib, have been
reported to be effective in the treatment of adult DM [206,
207]. In addition, a case report showed that refractory patients Systemic Sclerosis
with JDM responded to the JAK1/JAK2 inhibitor, baricitinib
[208]. A phase 1b clinical study (NCT00533091) observed Systemic sclerosis (SSc), also called scleroderma, is charac-
that the anti-IFN-α mAb, sifalimumab inhibited type I IFN terized by fibrosis and dysfunction of the skin, internal organs,
signatures in patients with DM, but there is as yet no pediatric and a vasculopathy [8]. Case reports have shown that SSc
data [209]. A clinical trial (NCT02612857) of the TLR 7, 8, 9 occurs in patients after using IFN-α to treat chronic myelog-
inhibitor, IMO-8400, has been completed in patients with enous leukemia or IFN-β to treat hepatitis C [220]. IFN sig-
DM, but again, no pediatric data is available. Clinical trials natures can also be found in peripheral blood and affected skin
on the treatment of JDM by inhibiting type I IFN signatures of patients with SSc, even during the early phase of disease
are sorely needed. [221]. In addition, higher IFN signatures are positively corre-
lated with anti-topoisomerase antibodies or anti-U1-RNP an-
Sjögren Syndrome tibodies but negatively correlated with anti-RNA polymerase
III antibodies in patients with SSc [220, 222]. The level of
Primary Sjögren syndrome (pSjS) is an inflammatory autoim- type I IFNs is also related to severe symptoms in the skin,
mune disorder with the loss of secretory function in exocrine lung, and skeletal muscle of patients with SSc [223].
glands, causing dryness of the mucosal surfaces, especially in Moreover, disease activity can be suppressed after treatment
the mouth (salivary) and eyes (lacrimal glands) [210]. IFN-α with the anti-IFNAR mAb anifrolumab in patients with SSc,
expression levels at the transcript or protein levels have been demonstrating an important role of type I IFNs in the patho-
found to be increased in the labial salivary glands and blood of genesis of SSc [224].
Table 3 Clinical trials of molecules targeting the type I interferon pathway in patients with SLE

Mechanism Type of inhibitor Molecule Current developmental phase Ref

IFN-α Anti-IFN-α mAb Rontalizumab Phase II (completed) NCT00962832 [160]


Development terminated
Sifalimumab (formerly MEDI-545) Phase IIb (completed) NCT01283139 [162]
AGS-009 (NNC-0152-0000-0001) Phase I (completed) NCT00960362 [163]
No following trials yet
Therapeutic vaccine: IFN-α Interferon-α-kinoid (IFN-K) Phase I/II (completed) NCT01058343 [164]
Phase II (active, not recruiting) NCT02665364a
IFN-I Inhibitor of IFN-Is JNJ-55920839 Phase I (completed but no results) NCT02609789a
IFNAR1 Anti-IFNAR1 mAb Anifrolumab (formerly MEDI-546) Phase III (completed) NCT02446912 [20, 168]
Phase III (completed) NCT02446899
pDC Anti-BDCA2 mAb BIIB059 Phase II (completed) NCT02847598 [169]
Anti-CD123 Talacotuzumab (JNJ-56022473) Phase I (withdrawn, based on benefit risk assessment) NCT02920424 [176]
Bcl-2 inhibitor Venetoclax (ABT-199) Phase I (completed) NCT01686555 [171, 172]
TLRs Inhibitor of TLR7, 8, 9 CpG-52,364 Phase I (completed) NCT00547014 [173]
Oligonucleotide inhibitor of TLR7, 9 DV1179 Phase Ib/IIa (completed)b
MyD88 Inhibitor of MyD88 ST2825 Preclinical [173]
Syk Syk kinase inhibitors Lanraplenib (GS 9876) Phase II (completed, for CLE) NCT03134222a
GSK 2646264 Phase I (completed, for CLE) NCT02927457 [173]
Fostamatinib (R935788, R788) Phase II (withdrawn for business reasons) NCT00752999 [173]
IRAK4 IRAK4 inhibitor BMS-986126 Preclinical [174]
BTK BTK inhibitor Evobrutinib (M2591, MSC2364447C) Phase II (active, not recruiting) NCT02975336 [175]
ABBV-105 phase II (recruiting) NCT03978520a
GDC-0853 phase II (active, not recruiting) NCT03407482a
JAK JAK1/JAK2 inhibitor Baricitinib (LY3009104) Phase III (recruiting) NCT03843125a [21]
Phase III (recruiting) NCT03616912a
Phase III (recruiting) NCT03616964
Ruxolitinib (INCB018424) Preclinical in cutaneous lupus [178]
JAK1/JAK3 inhibitor Tofacitinib (CP-690550) Phase I (completed) NCT02535689a
Phase I (recruiting for CLE) NCT03159936a
Phase I/II (recruiting for CLE) NCT03288324a
JAK1/JAK2/JAK3 inhibitor Tanzisertib (CC-930) Phase II (terminated for benefit/risk profile) NCT01466725a
JAK1 selective inhibitor Solcitinib (GSK2586184) Phase II (terminated for meeting futility criterion) NCT01777256a
Filgotinib (GLPG0634) Phase II (completed, for CLE) NCT03134222a
topical JAK/SYK inhibitor R333 Phase II (completed) NCT01597050 [177]
Tyk2 Tyk2 inhibitor BMS-986165 Phase II (recruiting) NCT03920267a
Phase II (recruiting) NCT03252587a
Nucleic acid human RNase RSLV-132 Phase II (active, not recruiting) NCT02660944 [176]
Recombinant human Dnase I rhDNase Development terminated

IFN interferon, mAB monoclonal antibody, IFNAR1 interferon-α/β receptor 1, pDCs plasmacytoid dendritic cells, TLR Toll-like receptor, MyD88 myeloid differentiation primary response gene 88, Syk
spleen tyrosine kinase, IRAK4 interleukin-1 receptor-associated kinase 4, BTK Bruton’s tyrosine kinase, JAK Janus kinase, TYK2 tyrosine kinase 2
a
Deriving from the ClinicalTrials.gov website
b
Deriving from http://investors.dynavax.com
Clinic Rev Allerg Immunol
Clinic Rev Allerg Immunol

A phase 1, multicenter, open-label study (NCT00930683) but the treatments are often not much better, with many
in which patients with SSc were treated with the anti-IFN-α associated with significant side effects [4–7]. The type I
mAb anifrolumab has shown good results and supported its IFN signaling pathway is of great significance in antiviral
further clinical development [225]. pDCs can directly promote and antitumor immunity [15, 17]. In addition, as men-
the development of skin fibrosis, and depleting pDCs can tioned above, the type I IFN signaling pathway is essen-
prevent the progression of the fibrotic process in mouse tial for the initiation and development of autoimmune dis-
models of SSc [226]. Clinical trials (NCT03817424) of eases [15, 17]. Several inhibitors blocking specific mole-
VIB7734, an inhibitor of pDCs, in patients with SSc are still cules in the type I IFN signaling pathway, such as
recruiting, and another clinical study (NCT04206644) of JAK tofacitinib and baricitinib for the treatment of RA, are
inhibitors is also ongoing. licensed by the FDA/EMA, and more are currently under-
going clinical trials [19]. There are encouraging signs that
many targeted inhibitors have completed or entered phase
Perspectives III clinical trials and seem to be very promising, including
the anti-IFNAR1 mAb anifrolumab for SLE [20, 168];
Autoimmune diseases consist of a wide range of systemic JAK1/JAK2 inhibitor baricitinib for SLE [21]; and the
diseases that affect up to 10% of the world’s population JAK1 inhibitors upadacitinib and filgotinib for RA [181,
[227]. Not only are the symptoms potentially disabling, 182]. Tofacitinib has completed a phase III clinical trial in

Table 4 Clinical trials of molecules targeting the type I interferon pathway in patients with RA

Mechanism Type of inhibitor Inhibitor Phase Ref

JAKs JAK1/JAK3 inhibitor Tofacitinib (Xeljanz; tasocitinib; CP-690550) FDA and EMA approved [19]
JAK1/JAK2 inhibitor Baricitinib (LY 3009104; INCB28050) FDA and EMA approved [19]
JAK1 inhibitor Upadacitinib (ABT-494) Phase III (active, not recruiting) NCT02675426a
Phase II/III (active, not recruiting) NCT02720523a
Phase III (active, not recruiting) NCT02706847a
Phase III (active, not recruiting) NCT02706951a
Phase III (active, not recruiting) NCT02706873a
Phase III (active, not recruiting) NCT02629159a
Phase III (active, not recruiting) NCT02955212a
Phase III (active, not recruiting) NCT03086343a
Filgotinib (GLPG0634) Phase III (active, not recruiting) NCT03025308a [182]
Phase III (completed) NCT02873936
Phase III (completed) NCT02886728a
Phase III (completed) NCT02886728a
Itacitinib (INCB039110) Phase II (completed) NCT01626573a
JAK1/JAK3 inhibitor Peficitinib (ASP015K) Phase III (completed) NCT02305849a
Phase III (completed) NCT02308163a
Phase III (recruiting) NCT03660059a
JAK1/JAK2 inhibitor Ruxolitinib (INCB18424) Phase II (completed) NCT00550043a
JAK3 inhibitor Decernotinib (VX-509) Phase II (completed) NCT01052194a
Phase II/III (completed) NCT01830985a
Development terminated
PF-06651600 Phase II (completed) NCT02969044a
Syk Syk inhibitor Fostamatinib Phase III (terminated) NCT01242514a [188]
Phase III (completed) NCT01197521
Phase III (completed) NCT01197534a
Development terminated
Lanraplenib (GS-9876) Phase II (completed) NCT02885181a
BTK BTK inhibitor Spebrutinib (CC-292) Phase II (completed) NCT01975610 [188]
ABBV-105 Phase II (active, not recruiting) NCT03823378a
Phase II (active, not recruiting) NCT03682705a
HM71224 Phase I (completed) NCT01765478a
TK-020 Phase I (completed) NCT02413255a

IFN interferon, JAK Janus kinase, TYK2 tyrosine kinase 2, Syk spleen tyrosine kinase, BTK Bruton’s tyrosine kinase
a
Deriving from the ClinicalTrials.gov website
Clinic Rev Allerg Immunol

patients with JIA and shown positive results [198]. Other References
studies are still not completed so results are unavailable.
More data and evidence are needed to support targeted 1. Wahren-Herlenius M, Dorner T (2013) Immunopathogenic mech-
anisms of systemic autoimmune disease. Lancet (London,
inhibitors that have not entered phase III clinical trials
England) 382(9894):819–831. https://doi.org/10.1016/S0140-
but which have shown good responses in case reports or 6736(13)60954-X
animal models. 2. Yan N, Chen ZJ (2012) Intrinsic antiviral immunity. Nat Immunol
The role of IFN-Is in the pathogenesis of autoimmune dis- 13(3):214–222. https://doi.org/10.1038/ni.2229
eases is still unclear. Under most circumstances, IFN-Is are 3. Abramson J, Husebye ES (2016) Autoimmune regulator and self-
tolerance - molecular and clinical aspects. Immunol Rev 271(1):
proinflammatory, but type I IFNs also seem to have a protec- 127–140. https://doi.org/10.1111/imr.12419
tive function in RA and MS based on some studies [189, 228]. 4. Seibel MJ, Cooper MS, Zhou H (2013) Glucocorticoid-induced
Some have proposed that conflicting results may be a result of osteoporosis: mechanisms, management, and future perspectives.
the varying immunomodulatory and proinflammatory func- Lancet Diabetes Endocrinol 1(1):59–70. https://doi.org/10.1016/
tions of interacting cytokines in autoimmune disorders that S2213-8587(13)70045-7
5. Bakshi J, Segura BT, Wincup C, Rahman A (2018) Unmet needs
are predominately driven by Th1 cells or Th17 cells [229]. in the pathogenesis and treatment of systemic lupus erythemato-
Furthermore, the results of anifrolumab in the phase III sus. Clin Rev Allergy Immunol 55(3):352–367. https://doi.org/10.
TULIP-1 and TULIP-2 trials are partially inconsistent [20, 1007/s12016-017-8640-5
168]. The end point BICLA, but not SRI, was met in 6. Davis LS, Reimold AM (2017) Research and therapeutics-
traditional and emerging therapies in systemic lupus erythemato-
TULIP-1, but both endpoints were met in TULIP-2 [20,
sus. Rheumatology (Oxford, England) 56(suppl_1):i100–i113.
168]. Heterogeneity of the disease state and genetics may be https://doi.org/10.1093/rheumatology/kew417
one of the reasons that different types of patients with SLE 7. Cheung TT, McInnes IB (2017) Future therapeutic targets in rheu-
may have varying responses to specific therapies. The estab- matoid arthritis? Semin Immunopathol 39(4):487–500. https://doi.
lishment of subtypes and personalized approaches based on org/10.1007/s00281-017-0623-3
8. Mavragani CP, Moutsopoulos HM (2019) Sjogren’s syndrome:
immune genotypes is needed for solving disease heterogene-
old and new therapeutic targets J autoimmun:102364. https://doi.
ity and efficiently achieving better targeted therapies [6]. In org/10.1016/j.jaut.2019.102364
addition, the anti-IFN-α mAbs rontalizumab and AGS-009 9. Hooks JJ, Moutsopoulos HM, Geis SA, Stahl NI, Decker JL,
did not achieve the expected results as anti-IFNAR mAbs, Notkins AL (1979) Immune interferon in the circulation of pa-
which may be caused by the 13 different subtypes of IFN-α tients with autoimmune disease. N Engl J Med 301(1):5–8.
https://doi.org/10.1056/NEJM197907053010102
in humans, making it difficult to block all types of IFN-α 10. Prummel MF, Laurberg P (2003) Interferon-alpha and autoim-
[160]. Besides, IFN-β may continue to play a role in disease mune thyroid disease. Thyroid 13(6):547–551. https://doi.org/10.
progression even in the presence of anti-IFN-α therapies. 1089/105072503322238809
Overall, type I IFN signaling pathway is of great signifi- 11. Ronnblom LE, Alm GV, Oberg KE (1990) Possible induction of
cance in the pathogenesis and treatment of both adult and systemic lupus erythematosus by interferon-alpha treatment in a
patient with a malignant carcinoid tumour. J Intern Med 227(3):
pediatric autoimmune diseases. Many inhibitors targeting the 207–210. https://doi.org/10.1111/j.1365-2796.1990.tb00144.x
type I IFN signaling pathway have shown positive results in 12. Barrat FJ, Crow MK, Ivashkiv LB (2019) Interferon target-gene
clinical trials of autoimmune diseases. Thus, it is very impor- expression and epigenomic signatures in health and disease. Nat
tant to further explore the role of the type I IFN signaling Immunol 20(12):1574–1583. https://doi.org/10.1038/s41590-
019-0466-2
pathway in autoimmune diseases to develop more effective
13. Higgs BW, Liu Z, White B, Zhu W, White WI, Morehouse C,
and safe therapies. Brohawn P, Kiener PA, Richman L, Fiorentino D, Greenberg SA,
Jallal B, Yao Y (2011) Patients with systemic lupus erythemato-
Acknowledgments We thank Xing Wang, Meixi Liu, Zhenwei Tang, sus, myositis, rheumatoid arthritis and scleroderma share activa-
and Bowen Li for their guidance in revising the manuscript. tion of a common type I interferon pathway. Ann Rheum Dis
70(11):2029–2036. https://doi.org/10.1136/ard.2011.150326
Author Contributions Concept and design: Qianjin Lu, Haijing Wu, 14. Negishi H, Taniguchi T, Yanai H (2018) The interferon (IFN)
Ming Zhao, Jiao Jiang; Drafting of the manuscript: Jiao Jiang; Revising class of cytokines and the IFN regulatory factor (IRF) transcription
of the manuscript: Qianjin Lu, Haijing Wu, Christopher Chang, Ming factor family. Cold Spring Harb Perspect Biol 10(11). https://doi.
Zhao, Jiao Jiang. org/10.1101/cshperspect.a028423
Funding information 15. Lazear HM, Schoggins JW, Diamond MS (2019) Shared and dis-
This work was supported by the National Natural Science Foundation tinct functions of type I and type III interferons. Immunity 50(4):
of China (No. 81972943 and No. 81830097) and the Hunan Talent 907–923. https://doi.org/10.1016/j.immuni.2019.03.025
Young Investigator (No. 2019RS2012). 16. Hillion S, Arleevskaya MI, Blanco P, Bordron A, Brooks WH,
Cesbron JY, Kaveri S, Vivier E, Renaudineau Y (2020) The innate
part of the adaptive immune system. Clin Rev Allergy Immunol
Compliance with Ethical Standards 58(2):151–154. https://doi.org/10.1007/s12016-019-08740-1
17. Ivashkiv LB, Donlin LT (2014) Regulation of type I interferon
Conflict of Interest The authors declare that they have no conflicts of responses. Nat Rev Immunol 14(1):36–49. https://doi.org/10.
interest. 1038/nri3581
Clinic Rev Allerg Immunol

18. Schreiber G (2017) The molecular basis for differential type I 35. Cingoz O, Goff SP (2018) Cyclin-dependent kinase activity is
interferon signaling. J Biol Chem 292(18):7285–7294. https:// required for type I interferon production. Proc Natl Acad Sci U
doi.org/10.1074/jbc.R116.774562 S A 115(13):E2950–E2959. https://doi.org/10.1073/pnas.
19. Taylor PC (2019) Clinical efficacy of launched JAK inhibitors in 1720431115
rheumatoid arthritis. Rheumatology (Oxford, England) 58(Suppl 36. Dias Junior AG, Sampaio NG, Rehwinkel J (2019) A balancing
1):i17–i26. https://doi.org/10.1093/rheumatology/key225 act: MDA5 in antiviral immunity and autoinflammation. Trends
20. Morand EF, Furie R, Tanaka Y, Bruce IN, Askanase AD, Richez Microbiol 27(1):75–85. https://doi.org/10.1016/j.tim.2018.08.007
C, Bae SC, Brohawn PZ, Pineda L, Berglind A, Tummala R, 37. Yoneyama M, Fujita T (2009) RNA recognition and signal trans-
Investigators T-T (2020) Trial of anifrolumab in active systemic duction by RIG-I-like receptors. Immunol Rev 227(1):54–65.
lupus erythematosus. N Engl J Med 382(3):211–221. https://doi. https://doi.org/10.1111/j.1600-065X.2008.00727.x
org/10.1056/NEJMoa1912196 38. Taniguchi T, Ogasawara K, Takaoka A, Tanaka N (2001) IRF
21. Kubo S, Nakayamada S, Tanaka Y (2019) Baricitinib for the treat- family of transcription factors as regulators of host defense.
ment of rheumatoid arthritis and systemic lupus erythematosus: a Annu Rev Immunol 19:623–655. https://doi.org/10.1146/
2019 update. Expert Rev Clin Immunol 15(7):693–700. https:// annurev.immunol.19.1.623
doi.org/10.1080/1744666X.2019.1608821 39. Tailor P, Tamura T, Ozato K (2006) IRF family proteins and type I
22. Pestka S, Krause CD, Walter MR (2004) Interferons, interferon- interferon induction in dendritic cells. Cell Res 16(2):134–140.
like cytokines, and their receptors. Immunol Rev 202:8–32. https://doi.org/10.1038/sj.cr.7310018
https://doi.org/10.1111/j.0105-2896.2004.00204.x 40. Li P, Wong JJ, Sum C, Sin WX, Ng KQ, Koh MB, Chin KC
23. Kato Y, Park J, Takamatsu H, Konaka H, Aoki W, Aburaya S, (2011) IRF8 and IRF3 cooperatively regulate rapid interferon-
Ueda M, Nishide M, Koyama S, Hayama Y, Kinehara Y, Hirano beta induction in human blood monocytes. Blood 117(10):2847–
T, Shima Y, Narazaki M, Kumanogoh A (2018) Apoptosis- 2854. https://doi.org/10.1182/blood-2010-07-294272
derived membrane vesicles drive the cGAS-STING pathway and 41. Mancuso G, Gambuzza M, Midiri A, Biondo C, Papasergi S,
enhance type I IFN production in systemic lupus erythematosus. Akira S, Teti G, Beninati C (2009) Bacterial recognition by
Ann Rheum Dis 77(10):1507–1515. https://doi.org/10.1136/ TLR7 in the lysosomes of conventional dendritic cells. Nat
annrheumdis-2018-212988 Immunol 10(6):587–594. https://doi.org/10.1038/ni.1733
24. Baccala R, Hoebe K, Kono DH, Beutler B, Theofilopoulos AN 42. Chen K, Liu J, Cao X (2017) Regulation of type I interferon
(2007) TLR-dependent and TLR-independent pathways of type I signaling in immunity and inflammation: a comprehensive re-
interferon induction in systemic autoimmunity. Nat Med 13(5): view. J Autoimmun 83:1–11. https://doi.org/10.1016/j.jaut.2017.
543–551. https://doi.org/10.1038/nm1590 03.008
25. Lou H, Pickering MC (2018) Extracellular DNA and autoimmune 43. Das A, Dinh PX, Panda D, Pattnaik AK (2014) Interferon-
diseases. Cell Mol Immunol 15(8):746–755. https://doi.org/10. inducible protein IFI35 negatively regulates RIG-I antiviral sig-
1038/cmi.2017.136 naling and supports vesicular stomatitis virus replication. J Virol
26. Raftery N, Stevenson NJ (2017) Advances in anti-viral immune 88(6):3103–3113. https://doi.org/10.1128/JVI.03202-13
defence: revealing the importance of the IFN JAK/STAT path- 44. Arimoto K, Takahashi H, Hishiki T, Konishi H, Fujita T,
way. Cell Mol Life Sci 74(14):2525–2535. https://doi.org/10. Shimotohno K (2007) Negative regulation of the RIG-I signaling
1007/s00018-017-2520-2 by the ubiquitin ligase RNF125. Proc Natl Acad Sci U S A
27. Lester SN, Li K (2014) Toll-like receptors in antiviral innate im- 104(18):7500–7505. https://doi.org/10.1073/pnas.0611551104
munity. J Mol Biol 426(6):1246–1264. https://doi.org/10.1016/j. 45. Cui J, Zhu L, Xia X, Wang HY, Legras X, Hong J, Ji J, Shen P,
jmb.2013.11.024 Zheng S, Chen ZJ, Wang RF (2010) NLRC5 negatively regulates
28. Lee CC, Avalos AM, Ploegh HL (2012) Accessory molecules for the NF-kappaB and type I interferon signaling pathways. Cell
toll-like receptors and their function. Nat Rev Immunol 12(3): 141(3):483–496. https://doi.org/10.1016/j.cell.2010.03.040
168–179. https://doi.org/10.1038/nri3151 46. Catrysse L, Vereecke L, Beyaert R, van Loo G (2014) A20 in
29. Zevini A, Olagnier D, Hiscott J (2017) Crosstalk between cyto- inflammation and autoimmunity. Trends Immunol 35(1):22–31.
plasmic RIG-I and STING sensing pathways. Trends Immunol https://doi.org/10.1016/j.it.2013.10.005
38(3):194–205. https://doi.org/10.1016/j.it.2016.12.004 47. Li X, Fu Z, Liang H, Wang Y, Qi X, Ding M, Sun X, Zhou Z,
30. Briard B, Place DE, Kanneganti TD (2020) DNA sensing in the Huang Y, Gu H, Li L, Chen X, Li D, Zhao Q, Liu F, Wang H,
innate immune response. Physiology (Bethesda) 35(2):112–124. Wang J, Zen K, Zhang CY (2018) H5N1 influenza virus-specific
https://doi.org/10.1152/physiol.00022.2019 miRNA-like small RNA increases cytokine production and mouse
31. Jonsson KL, Laustsen A, Krapp C, Skipper KA, Thavachelvam K, mortality via targeting poly(rC)-binding protein 2. Cell Res 28(2):
Hotter D, Egedal JH, Kjolby M, Mohammadi P, Prabakaran T, 157–171. https://doi.org/10.1038/cr.2018.3
Sorensen LK, Sun C, Jensen SB, Holm CK, Lebbink RJ, 48. Maelfait J, Beyaert R (2012) Emerging role of ubiquitination in
Johannsen M, Nyegaard M, Mikkelsen JG, Kirchhoff F, Paludan antiviral RIG-I signaling. Microbiol Mol Biol Rev 76(1):33–45.
SR, Jakobsen MR (2017) IFI16 is required for DNA sensing in https://doi.org/10.1128/MMBR.05012-11
human macrophages by promoting production and function of 49. Zhang M, Wang L, Zhao X, Zhao K, Meng H, Zhao W, Gao C
cGAMP. Nat Commun 8:14391. https://doi.org/10.1038/ (2012) TRAF-interacting protein (TRIP) negatively regulates
ncomms14391 IFN-beta production and antiviral response by promoting
32. Arleevskaya MI, Larionova RV, Brooks WH, Bettacchioli E, proteasomal degradation of TANK-binding kinase 1. J Exp Med
Renaudineau Y (2020) Toll-like receptors, infections, and rheu- 209(10):1703–1711. https://doi.org/10.1084/jem.20120024
matoid arthritis. Clin Rev Allergy Immunol 58(2):172–181. 50. van Gent M, Sparrer KMJ, Gack MU (2018) TRIM proteins and
https://doi.org/10.1007/s12016-019-08742-z their roles in antiviral host defenses. Annu Rev Virol 5(1):385–
33. Brisse M, Ly H (2019) Comparative structure and function anal- 405. https://doi.org/10.1146/annurev-virology-092917-043323
ysis of the RIG-I-like receptors: RIG-I and MDA5. Front Immunol 51. Ozato K, Shin DM, Chang TH, Morse HC 3rd (2008) TRIM
10:1586. https://doi.org/10.3389/fimmu.2019.01586 family proteins and their emerging roles in innate immunity. Nat
34. O'Neill LA, Bowie AG (2007) The family of five: TIR-domain- Rev Immunol 8(11):849–860. https://doi.org/10.1038/nri2413
containing adaptors in toll-like receptor signalling. Nat Rev 52. Hu Y, Wang J, Yang B, Zheng N, Qin M, Ji Y, Lin G, Tian L, Wu
Immunol 7(5):353–364. https://doi.org/10.1038/nri2079 X, Wu L, Sun B (2011) Guanylate binding protein 4 negatively
Clinic Rev Allerg Immunol

regulates virus-induced type I IFN and antiviral response by 67. An H, Zhao W, Hou J, Zhang Y, Xie Y, Zheng Y, Xu H, Qian C,
targeting IFN regulatory factor 7. J Immunol 187(12):6456– Zhou J, Yu Y, Liu S, Feng G, Cao X (2006) SHP-2 phosphatase
6462. https://doi.org/10.4049/jimmunol.1003691 negatively regulates the TRIF adaptor protein-dependent type I
53. Liu B, Mink S, Wong KA, Stein N, Getman C, Dempsey PW, Wu interferon and proinflammatory cytokine production. Immunity
H, Shuai K (2004) PIAS1 selectively inhibits interferon-inducible 25(6):919–928. https://doi.org/10.1016/j.immuni.2006.10.014
genes and is important in innate immunity. Nat Immunol 5(9): 68. Bourdeau A, Dube N, Tremblay ML (2005) Cytoplasmic protein
891–898. https://doi.org/10.1038/ni1104 tyrosine phosphatases, regulation and function: the roles of
54. Kubota T, Matsuoka M, Xu S, Otsuki N, Takeda M, Kato A, Ozato PTP1B and TC-PTP. Curr Opin Cell Biol 17(2):203–209.
K (2011) PIASy inhibits virus-induced and interferon-stimulated https://doi.org/10.1016/j.ceb.2005.02.001
transcription through distinct mechanisms. J Biol Chem 286(10): 69. Alexander WS (2002) Suppressors of cytokine signalling (SOCS)
8165–8175. https://doi.org/10.1074/jbc.M110.195255 in the immune system. Nat Rev Immunol 2(6):410–416. https://
55. Li Y, Li C, Xue P, Zhong B, Mao AP, Ran Y, Chen H, Wang YY, doi.org/10.1038/nri818
Yang F, Shu HB (2009) ISG56 is a negative-feedback regulator of 70. Nair S, Bist P, Dikshit N, Krishnan MN (2016) Global functional
virus-triggered signaling and cellular antiviral response. Proc Natl profiling of human ubiquitome identifies E3 ubiquitin ligase
Acad Sci U S A 106(19):7945–7950. https://doi.org/10.1073/ DCST1 as a novel negative regulator of type-I interferon signal-
pnas.0900818106 ing. Sci Rep 6:36179. https://doi.org/10.1038/srep36179
56. John SP, Sun J, Carlson RJ, Cao B, Bradfield CJ, Song J, 71. Jeronimo C, Bastian PJ, Bjartell A, Carbone GM, Catto JW, Clark
Smelkinson M, Fraser IDC (2018) IFIT1 exerts opposing regula- SJ, Henrique R, Nelson WG, Shariat SF (2011) Epigenetics in
tory effects on the inflammatory and interferon gene programs in prostate cancer: biologic and clinical relevance. Eur Urol 60(4):
LPS-activated human macrophages. Cell Rep 25(1):95–106 e106. 753–766. https://doi.org/10.1016/j.eururo.2011.06.035
https://doi.org/10.1016/j.celrep.2018.09.002 72. Szyf M (2003) DNA methylation and cancer therapy. Drug Resist
57. Wang J, Yang B, Hu Y, Zheng Y, Zhou H, Wang Y, Ma Y, Mao Updat 6(6):341–353. https://doi.org/10.1016/j.drup.2003.10.002
K, Yang L, Lin G, Ji Y, Wu X, Sun B (2013) Negative regulation 73. Imgenberg-Kreuz J, Carlsson Almlof J, Leonard D, Alexsson A,
of Nmi on virus-triggered type I IFN production by targeting Nordmark G, Eloranta ML, Rantapaa-Dahlqvist S, Bengtsson AA,
IRF7. J Immunol 191(6):3393–3399. https://doi.org/10.4049/ Jonsen A, Padyukov L, Gunnarsson I, Svenungsson E, Sjowall C,
jimmunol.1300740 Ronnblom L, Syvanen AC, Sandling JK (2018) DNA methylation
58. Lee MS, Kim B, Oh GT, Kim YJ (2013) OASL1 inhibits transla- mapping identifies gene regulatory effects in patients with system-
tion of the type I interferon-regulating transcription factor IRF7. ic lupus erythematosus. Ann Rheum Dis 77(5):736–743. https://
Nat Immunol 14(4):346–355. https://doi.org/10.1038/ni.2535 doi.org/10.1136/annrheumdis-2017-212379
59. Ma Z, Moore R, Xu X, Barber GN (2013) DDX24 negatively 74. Zhu H, Wu LF, Mo XB, Lu X, Tang H, Zhu XW, Xia W, Guo YF,
regulates cytosolic RNA-mediated innate immune signaling. Wang MJ, Zeng KQ, Wu J, Qiu YH, Lin X, Zhang YH, Liu YZ,
PLoS Pathog 9(10):e1003721. https://doi.org/10.1371/journal. Yi NJ, Deng FY, Lei SF (2019) Rheumatoid arthritis-associated
ppat.1003721 DNA methylation sites in peripheral blood mononuclear cells.
60. Li J, Ding SC, Cho H, Chung BC, Gale M Jr, Chanda SK, Ann Rheum Dis 78(1):36–42. https://doi.org/10.1136/
Diamond MS (2013) A short hairpin RNA screen of interferon- annrheumdis-2018-213970
stimulated genes identifies a novel negative regulator of the cel- 75. Imgenberg-Kreuz J, Sandling JK, Almlof JC, Nordlund J, Signer L,
lular antiviral response. mBio 4(3):e00385-00313. https://doi.org/ Norheim KB, Omdal R, Ronnblom L, Eloranta ML, Syvanen AC,
10.1128/mBio.00385-13 Nordmark G (2016) Genome-wide DNA methylation analysis in
61. Yu Y, Hayward GS (2010) The ubiquitin E3 ligase RAUL nega- multiple tissues in primary Sjogren’s syndrome reveals regulatory
tively regulates type i interferon through ubiquitination of the effects at interferon-induced genes. Ann Rheum Dis 75(11):2029–
transcription factors IRF7 and IRF3. Immunity 33(6):863–877. 2036. https://doi.org/10.1136/annrheumdis-2015-208659
https://doi.org/10.1016/j.immuni.2010.11.027 76. Ding W, Pu W, Wang L, Jiang S, Zhou X, Tu W, Yu L, Zhang J,
62. Sugiyama Y, Kakoi K, Kimura A, Takada I, Kashiwagi I, Guo S, Liu Q, Ma Y, Chen S, Wu W, Reveille J, Zou H, Jin L,
Wakabayashi Y, Morita R, Nomura M, Yoshimura A (2012) Wang J (2018) Genome-wide DNA methylation analysis in sys-
Smad2 and Smad3 are redundantly essential for the suppression temic sclerosis reveals hypomethylation of IFN-associated genes
of iNOS synthesis in macrophages by regulating IRF3 and in CD4(+) and CD8(+) T cells. J Investig Dermatol 138(5):1069–
STAT1 pathways. Int Immunol 24(4):253–265. https://doi.org/ 1077. https://doi.org/10.1016/j.jid.2017.12.003
10.1093/intimm/dxr126 77. Chiappinelli KB, Strissel PL, Desrichard A, Li H, Henke C,
63. Zheng H, Qian J, Varghese B, Baker DP, Fuchs S (2011) Ligand- Akman B, Hein A, Rote NS, Cope LM, Snyder A, Makarov V,
stimulated downregulation of the alpha interferon receptor: role of Budhu S, Slamon DJ, Wolchok JD, Pardoll DM, Beckmann MW,
protein kinase D2. Mol Cell Biol 31(4):710–720. https://doi.org/ Zahnow CA, Merghoub T, Chan TA, Baylin SB, Strick R (2015)
10.1128/MCB.01154-10 Inhibiting DNA methylation causes an interferon response in can-
64. Arimoto KI, Lochte S, Stoner SA, Burkart C, Zhang Y, Miyauchi cer via dsRNA including endogenous retroviruses. Cell 162(5):
S, Wilmes S, Fan JB, Heinisch JJ, Li Z, Yan M, Pellegrini S, 974–986. https://doi.org/10.1016/j.cell.2015.07.011
Colland F, Piehler J, Zhang DE (2017) STAT2 is an essential 78. Shilatifard A (2008) Molecular implementation and physiological
adaptor in USP18-mediated suppression of type I interferon sig- roles for histone H3 lysine 4 (H3K4) methylation. Curr Opin Cell
naling. Nat Struct Mol Biol 24(3):279–289. https://doi.org/10. Biol 20(3):341–348. https://doi.org/10.1016/j.ceb.2008.03.019
1038/nsmb.3378 79. Fang TC, Schaefer U, Mecklenbrauker I, Stienen A, Dewell S,
65. Yasukawa H, Sasaki A, Yoshimura A (2000) Negative regulation Chen MS, Rioja I, Parravicini V, Prinjha RK, Chandwani R,
of cytokine signaling pathways. Annu Rev Immunol 18:143–164. MacDonald MR, Lee K, Rice CM, Tarakhovsky A (2012)
https://doi.org/10.1146/annurev.immunol.18.1.143 Histone H3 lysine 9 di-methylation as an epigenetic signature of
66. An H, Hou J, Zhou J, Zhao W, Xu H, Zheng Y, Yu Y, Liu S, Cao the interferon response. J Exp Med 209(4):661–669. https://doi.
X (2008) Phosphatase SHP-1 promotes TLR- and RIG-I-activated org/10.1084/jem.20112343
production of type I interferon by inhibiting the kinase IRAK1. 80. Chen K, Liu J, Liu S, Xia M, Zhang X, Han D, Jiang Y, Wang C,
Nat Immunol 9(5):542–550. https://doi.org/10.1038/ni.1604 Cao X (2017) Methyltransferase SETD2-mediated methylation of
Clinic Rev Allerg Immunol

STAT1 is critical for interferon antiviral activity. Cell 170(3):492– 93. Smith S, Fernando T, Wu PW, Seo J, Ni Gabhann J, Piskareva O,
506 e414. https://doi.org/10.1016/j.cell.2017.06.042 McCarthy E, Howard D, O'Connell P, Conway R, Gallagher P,
81. Chen X, Liu X, Zhang Y, Huai W, Zhou Q, Xu S, Chen X, Li N, Molloy E, Stallings RL, Kearns G, Forbess L, Ishimori M,
Cao X (2020) Methyltransferase Dot1l preferentially promotes Venuturupalli S, Wallace D, Weisman M, Jefferies CA (2017)
innate IL-6 and IFN-beta production by mediating H3K79me2/3 MicroRNA-302d targets IRF9 to regulate the IFN-induced gene
methylation in macrophages. Cell Mol Immunol 17(1):76–84. expression in SLE. J Autoimmun 79:105–111. https://doi.org/10.
https://doi.org/10.1038/s41423-018-0170-4 1016/j.jaut.2017.03.003
82. Munshi N, Agalioti T, Lomvardas S, Merika M, Chen G, Thanos 94. Yoon WH, Meinhardt H, Montell DJ (2011) miRNA-mediated
D (2001) Coordination of a transcriptional switch by HMGI(Y) feedback inhibition of JAK/STAT morphogen signalling estab-
acetylation. Science 293(5532):1133–1136. https://doi.org/10. lishes a cell fate threshold. Nat Cell Biol 13(9):1062–1069.
1126/science.293.5532.1133 https://doi.org/10.1038/ncb2316
83. Agalioti T, Lomvardas S, Parekh B, Yie J, Maniatis T, Thanos D 95. Kohanbash G, Okada H (2012) MicroRNAs and STAT interplay.
(2000) Ordered recruitment of chromatin modifying and general Semin Cancer Biol 22(1):70–75. https://doi.org/10.1016/j.
transcription factors to the IFN-beta promoter. Cell 103(4):667– semcancer.2011.12.010
678. https://doi.org/10.1016/s0092-8674(00)00169-0 96. Gracias DT, Stelekati E, Hope JL, Boesteanu AC, Doering TA,
84. Meng J, Liu X, Zhang P, Li D, Xu S, Zhou Q, Guo M, Huai W, Norton J, Mueller YM, Fraietta JA, Wherry EJ, Turner M,
Chen X, Wang Q, Li N, Cao X (2016) Rb selectively inhibits Katsikis PD (2013) The microRNA miR-155 controls CD8(+) T
innate IFN-beta production by enhancing deacetylation of IFN- cell responses by regulating interferon signaling. Nat Immunol
beta promoter through HDAC1 and HDAC8. J Autoimmun 73: 14(6):593–602. https://doi.org/10.1038/ni.2576
42–53. https://doi.org/10.1016/j.jaut.2016.05.012 97. Navarro A, Pairet S, Alvarez-Larran A, Pons A, Ferrer G,
85. Marie IJ, Chang HM, Levy DE (2018) HDAC stimulates gene Longaron R, Fernandez-Rodriguez C, Camacho L, Monzo M,
expression through BRD4 availability in response to IFN and in Besses C, Bellosillo B (2016) miR-203 and miR-221 regulate
interferonopathies. J Exp Med 215(12):3194–3212. https://doi. SOCS1 and SOCS3 in essential thrombocythemia. Blood.
org/10.1084/jem.20180520 Cancer J 6:e406. https://doi.org/10.1038/bcj.2016.10
86. Decque A, Joffre O, Magalhaes JG, Cossec JC, Blecher-Gonen R, 98. Chen Y, Chen J, Wang H, Shi J, Wu K, Liu S, Liu Y, Wu J (2013)
Lapaquette P, Silvin A, Manel N, Joubert PE, Seeler JS, Albert HCV-induced miR-21 contributes to evasion of host immune sys-
ML, Amit I, Amigorena S, Dejean A (2016) Sumoylation coordi- tem by targeting MyD88 and IRAK1. PLoS Pathog 9(4):
nates the repression of inflammatory and anti-viral gene-expres- e1003248. https://doi.org/10.1371/journal.ppat.1003248
sion programs during innate sensing. Nat Immunol 17(2):140– 99. Nishitsuji H, Ujino S, Yoshio S, Sugiyama M, Mizokami M,
149. https://doi.org/10.1038/ni.3342 Kanto T, Shimotohno K (2016) Long noncoding RNA #32 con-
87. Zhang Y, Mao D, Roswit WT, Jin X, Patel AC, Patel DA, Agapov tributes to antiviral responses by controlling interferon-stimulated
E, Wang Z, Tidwell RM, Atkinson JJ, Huang G, McCarthy R, Yu gene expression. Proc Natl Acad Sci U S A 113(37):10388–
J, Yun NE, Paessler S, Lawson TG, Omattage NS, Brett TJ, 10393. https://doi.org/10.1073/pnas.1525022113
Holtzman MJ (2015) PARP9-DTX3L ubiquitin ligase targets host 100. Zhou Y, Li M, Xue Y, Li Z, Wen W, Liu X, Ma Y, Zhang L, Shen
histone H2BJ and viral 3C protease to enhance interferon signal- Z, Cao X (2019) Interferon-inducible cytoplasmic lncLrrc55-AS
ing and control viral infection. Nat Immunol 16(12):1215–1227. promotes antiviral innate responses by strengthening IRF3 phos-
https://doi.org/10.1038/ni.3279 phorylation. Cell Res 29(8):641–654. https://doi.org/10.1038/
88. Fonseca GJ, Thillainadesan G, Yousef AF, Ablack JN, Mossman s41422-019-0193-0
KL, Torchia J, Mymryk JS (2012) Adenovirus evasion of 101. Kambara H, Niazi F, Kostadinova L, Moonka DK, Siegel CT,
interferon-mediated innate immunity by direct antagonism of a cel- Post AB, Carnero E, Barriocanal M, Fortes P, Anthony DD,
lular histone posttranslational modification. Cell Host Microbe Valadkhan S (2014) Negative regulation of the interferon response
11(6):597–606. https://doi.org/10.1016/j.chom.2012.05.005 by an interferon-induced long non-coding RNA. Nucleic Acids
89. Li Y, Fan X, He X, Sun H, Zou Z, Yuan H, Xu H, Wang C, Shi X Res 42(16):10668–10680. https://doi.org/10.1093/nar/gku713
(2012) MicroRNA-466l inhibits antiviral innate immune response 102. Cui Y, Sheng Y, Zhang X (2013) Genetic susceptibility to SLE:
by targeting interferon-alpha. Cell Mol Immunol 9(6):497–502. recent progress from GWAS. J Autoimmun 41:25–33. https://doi.
https://doi.org/10.1038/cmi.2012.35 org/10.1016/j.jaut.2013.01.008
90. Han X, Wang Y, Zhang X, Qin Y, Qu B, Wu L, Ma J, Zhou Z, 103. Deng Y, Tsao BP (2010) Genetic susceptibility to systemic lupus
Qian J, Dai M, Tang Y, Chan EK, Harley JB, Zhou S, Shen N erythematosus in the genomic era. Nat Rev Rheumatol 6(12):683–
(2016) MicroRNA-130b ameliorates murine lupus nephritis 692. https://doi.org/10.1038/nrrheum.2010.176
through targeting the type I interferon pathway on renal mesangial 104. Ghodke-Puranik Y, Niewold TB (2015) Immunogenetics of systemic
cells. Arthritis Rheumatol (Hoboken, NJ) 68(9):2232–2243. lupus erythematosus: a comprehensive review. J Autoimmun 64:125–
https://doi.org/10.1002/art.39725 136. https://doi.org/10.1016/j.jaut.2015.08.004
91. Rossato M, Affandi AJ, Thordardottir S, Wichers CGK, Cossu M, 105. Delgado-Vega A, Sanchez E, Lofgren S, Castillejo-Lopez C,
Broen JCA, Moret FM, Bossini-Castillo L, Chouri E, van Bon L, Alarcon-Riquelme ME (2010) Recent findings on genetics of sys-
Wolters F, Marut W, van der Kroef M, Silva-Cardoso S, Bekker temic autoimmune diseases. Curr Opin Immunol 22(6):698–705.
CPJ, Dolstra H, van Laar JM, Martin J, van Roon JAG, Reedquist https://doi.org/10.1016/j.coi.2010.09.002
KA, Beretta L, Radstake T (2017) Association of MicroRNA-618 106. Zhai Y, Xu K, Leng RX, Cen H, Wang W, Zhu Y, Zhou M, Feng
expression with altered frequency and activation of plasmacytoid CC, Ye DQ (2013) Association of interleukin-1 receptor-associated
dendritic cells in patients with systemic sclerosis. Arthritis kinase (IRAK1) gene polymorphisms (rs3027898, rs1059702) with
Rheumatol (Hoboken, NJ) 69(9):1891–1902. https://doi.org/10. systemic lupus erythematosus in a Chinese Han population. Inflamm
1002/art.40163 Res 62(6):555–560. https://doi.org/10.1007/s00011-013-0607-2
92. Ho BC, Yu IS, Lu LF, Rudensky A, Chen HY, Tsai CW, Chang 107. Jacob CO, Zhu J, Armstrong DL, Yan M, Han J, Zhou XJ,
YL, Wu CT, Chang LY, Shih SR, Lin SW, Lee CN, Yang PC, Yu Thomas JA, Reiff A, Myones BL, Ojwang JO, Kaufman KM,
SL (2014) Inhibition of miR-146a prevents enterovirus-induced Klein-Gitelman M, McCurdy D, Wagner-Weiner L, Silverman
death by restoring the production of type I interferon. Nat E, Ziegler J, Kelly JA, Merrill JT, Harley JB, Ramsey-Goldman
Commun 5:3344. https://doi.org/10.1038/ncomms4344 R, Vila LM, Bae SC, Vyse TJ, Gilkeson GS, Gaffney PM, Moser
Clinic Rev Allerg Immunol

KL, Langefeld CD, Zidovetzki R, Mohan C (2009) Identification 119. Lee-Kirsch MA, Wolf C, Kretschmer S, Roers A (2015) Type I
of IRAK1 as a risk gene with critical role in the pathogenesis of interferonopathies–an expanding disease spectrum of
systemic lupus erythematosus. Proc Natl Acad Sci U S A 106(15): immunodysregulation. Semin Immunopathol 37(4):349–357.
6256–6261. https://doi.org/10.1073/pnas.0901181106 https://doi.org/10.1007/s00281-015-0500-x
108. Tyler DR, Persky ME, Matthews LA, Chan S, Farrar JD (2007) 120. Crow YJ, Hayward BE, Parmar R, Robins P, Leitch A, Ali M,
Pre-assembly of STAT4 with the human IFN-alpha/beta receptor- Black DN, van Bokhoven H, Brunner HG, Hamel BC, Corry PC,
2 subunit is mediated by the STAT4 N-domain. Mol Immunol Cowan FM, Frints SG, Klepper J, Livingston JH, Lynch SA,
44(8):1864–1872. https://doi.org/10.1016/j.molimm.2006.10.006 Massey RF, Meritet JF, Michaud JL, Ponsot G, Voit T, Lebon
109. Hagberg N, Joelsson M, Leonard D, Reid S, Eloranta ML, Mo J, P, Bonthron DT, Jackson AP, Barnes DE, Lindahl T (2006)
Nilsson MK, Syvanen AC, Bryceson YT, Ronnblom L (2018) Mutations in the gene encoding the 3′-5' DNA exonuclease
The STAT4 SLE risk allele rs7574865[T] is associated with in- TREX1 cause Aicardi-Goutieres syndrome at the AGS1 locus.
creased IL-12-induced IFN-gamma production in T cells from Nat Genet 38(8):917–920. https://doi.org/10.1038/ng1845
patients with SLE. Ann Rheum Dis 77(7):1070–1077. https:// 121. Crow YJ, Leitch A, Hayward BE, Garner A, Parmar R, Griffith E,
doi.org/10.1136/annrheumdis-2017-212794 Ali M, Semple C, Aicardi J, Babul-Hirji R, Baumann C, Baxter P,
110. Hagberg N, Ronnblom L (2019) Interferon-alpha enhances the IL- Bertini E, Chandler KE, Chitayat D, Cau D, Dery C, Fazzi E,
12-induced STAT4 activation selectively in carriers of the STAT4 Goizet C, King MD, Klepper J, Lacombe D, Lanzi G, Lyall H,
SLE risk allele rs7574865[T]. Ann Rheum Dis 78(3):429–431. Martinez-Frias ML, Mathieu M, McKeown C, Monier A, Oade Y,
https://doi.org/10.1136/annrheumdis-2018-213836 Quarrell OW, Rittey CD, Rogers RC, Sanchis A, Stephenson JB,
111. Wang Y, Shaked I, Stanford SM, Zhou W, Curtsinger JM, Tacke U, Till M, Tolmie JL, Tomlin P, Voit T, Weschke B,
Mikulski Z, Shaheen ZR, Cheng G, Sawatzke K, Campbell AM, Woods CG, Lebon P, Bonthron DT, Ponting CP, Jackson AP
Auger JL, Bilgic H, Shoyama FM, Schmeling DO, Balfour HH Jr, (2006) Mutations in genes encoding ribonuclease H2 subunits
Hasegawa K, Chan AC, Corbett JA, Binstadt BA, Mescher MF, cause Aicardi-Goutieres syndrome and mimic congenital viral
Ley K, Bottini N, Peterson EJ (2013) The autoimmunity- brain infection. Nat Genet 38(8):910–916. https://doi.org/10.
associated gene PTPN22 potentiates toll-like receptor-driven, type 1038/ng1842
1 interferon-dependent immunity. Immunity 39(1):111–122. 122. Rice GI, Bond J, Asipu A, Brunette RL, Manfield IW, Carr IM,
https://doi.org/10.1016/j.immuni.2013.06.013 Fuller JC, Jackson RM, Lamb T, Briggs TA, Ali M, Gornall H,
112. Chuang TH, Ulevitch RJ (2004) Triad3A, an E3 ubiquitin-protein Couthard LR, Aeby A, Attard-Montalto SP, Bertini E, Bodemer
ligase regulating toll-like receptors. Nat Immunol 5(5):495–502. C, Brockmann K, Brueton LA, Corry PC, Desguerre I, Fazzi E,
https://doi.org/10.1038/ni1066 Cazorla AG, Gener B, Hamel BC, Heiberg A, Hunter M, van der
113. Yarwood A, Huizinga TW, Worthington J (2016) The genetics of Knaap MS, Kumar R, Lagae L, Landrieu PG, Lourenco CM,
rheumatoid arthritis: risk and protection in different stages of the Marom D, McDermott MF, van der Merwe W, Orcesi S,
evolution of RA. Rheumatology (Oxford, England) 55(2):199– Prendiville JS, Rasmussen M, Shalev SA, Soler DM, Shinawi
209. https://doi.org/10.1093/rheumatology/keu323 M, Spiegel R, Tan TY, Vanderver A, Wakeling EL, Wassmer E,
114. Hinks A, Cobb J, Marion MC, Prahalad S, Sudman M, Bowes J, Whittaker E, Lebon P, Stetson DB, Bonthron DT, Crow YJ (2009)
Martin P, Comeau ME, Sajuthi S, Andrews R, Brown M, Chen Mutations involved in Aicardi-Goutieres syndrome implicate
WM, Concannon P, Deloukas P, Edkins S, Eyre S, Gaffney PM, SAMHD1 as regulator of the innate immune response. Nat
Guthery SL, Guthridge JM, Hunt SE, James JA, Keddache M, Genet 41(7):829–832. https://doi.org/10.1038/ng.373
Moser KL, Nigrovic PA, Onengut-Gumuscu S, Onslow ML, 123. Coquel F, Neumayer C, Lin YL, Pasero P (2019) SAMHD1 and
Rose CD, Rich SS, Steel KJ, Wakeland EK, Wallace CA, the innate immune response to cytosolic DNA during DNA repli-
Wedderburn LR, Woo P, Boston Children's JIAR, British cation. Curr Opin Immunol 56:24–30. https://doi.org/10.1016/j.
Society of P, Adolescent Rheumatology Study G, Childhood coi.2018.09.017
Arthritis Prospective S, Childhood Arthritis Response to 124. Rice GI, Kasher PR, Forte GM, Mannion NM, Greenwood SM,
Medication S, German Society for Pediatric R, Study JIAGE, Szynkiewicz M, Dickerson JE, Bhaskar SS, Zampini M, Briggs
Registry NJG, Study T, United Kingdom Juvenile Idiopathic TA, Jenkinson EM, Bacino CA, Battini R, Bertini E, Brogan PA,
Arthritis Genetics C, Bohnsack JF, Haas JP, Glass DN, Brueton LA, Carpanelli M, De Laet C, de Lonlay P, del Toro M,
Langefeld CD, Thomson W, Thompson SD (2013) Dense Desguerre I, Fazzi E, Garcia-Cazorla A, Heiberg A, Kawaguchi
genotyping of immune-related disease regions identifies 14 new M, Kumar R, Lin JP, Lourenco CM, Male AM, Marques W Jr,
susceptibility loci for juvenile idiopathic arthritis. Nat Genet 45(6): Mignot C, Olivieri I, Orcesi S, Prabhakar P, Rasmussen M,
664–669. https://doi.org/10.1038/ng.2614 Robinson RA, Rozenberg F, Schmidt JL, Steindl K, Tan TY,
115. Nordang GB, Viken MK, Amundsen SS, Sanchez ES, Flato B, van der Merwe WG, Vanderver A, Vassallo G, Wakeling EL,
Forre OT, Martin J, Kvien TK, Lie BA (2012) Interferon regula- Wassmer E, Whittaker E, Livingston JH, Lebon P, Suzuki T,
tory factor 5 gene polymorphism confers risk to several rheumatic McLaughlin PJ, Keegan LP, O'Connell MA, Lovell SC, Crow
diseases and correlates with expression of alternative thymic tran- YJ (2012) Mutations in ADAR1 cause Aicardi-Goutieres syn-
scripts. Rheumatology (Oxford, England) 51(4):619–626. https:// drome associated with a type I interferon signature. Nat Genet
doi.org/10.1093/rheumatology/ker364 44(11):1243–1248. https://doi.org/10.1038/ng.2414
116. Harris VM, Scofield RH, Sivils KL (2019) Genetics in Sjogren’s 125. Oda H, Nakagawa K, Abe J, Awaya T, Funabiki M, Hijikata A,
syndrome: where we are and where we go. Clin Exp Rheumatol Nishikomori R, Funatsuka M, Ohshima Y, Sugawara Y, Yasumi
37 Suppl 118(3):234–239 T, Kato H, Shirai T, Ohara O, Fujita T, Heike T (2014) Aicardi-
117. Burbelo PD, Ambatipudi K, Alevizos I (2014) Genome-wide as- Goutieres syndrome is caused by IFIH1 mutations. Am J Hum
sociation studies in Sjogren’s syndrome: what do the genes tell us Genet 95(1):121–125. https://doi.org/10.1016/j.ajhg.2014.06.007
about disease pathogenesis? Autoimmun Rev 13(7):756–761. 126. Pelzer N, Hoogeveen ES, Haan J, Bunnik R, Poot CC, van Zwet
https://doi.org/10.1016/j.autrev.2014.02.002 EW, Inderson A, Fogteloo AJ, Reinders MEJ, Middelkoop HAM,
118. Crow YJ (2015) Type I interferonopathies: mendelian type I in- Kruit MC, van den Maagdenberg A, Ferrari MD, Terwindt GM
terferon up-regulation. Curr Opin Immunol 32:7–12. https://doi. (2019) Systemic features of retinal vasculopathy with cerebral
org/10.1016/j.coi.2014.10.005 leukoencephalopathy and systemic manifestations: a monogenic
Clinic Rev Allerg Immunol

small vessel disease. J Intern Med 285(3):317–332. https://doi. JAK inhibitor therapy in a child with inherited USP18 deficiency.
org/10.1111/joim.12848 N Engl J Med 382(3):256–265. https://doi.org/10.1056/
127. Konig N, Fiehn C, Wolf C, Schuster M, Cura Costa E, Tungler V, NEJMoa1905633
Alvarez HA, Chara O, Engel K, Goldbach-Mansky R, Gunther C, 137. Sanchez GAM, Reinhardt A, Ramsey S, Wittkowski H, Hashkes
Lee-Kirsch MA (2017) Familial chilblain lupus due to a gain-of- PJ, Berkun Y, Schalm S, Murias S, Dare JA, Brown D, Stone DL,
function mutation in STING. Ann Rheum Dis 76(2):468–472. Gao L, Klausmeier T, Foell D, de Jesus AA, Chapelle DC, Kim H,
https://doi.org/10.1136/annrheumdis-2016-209841 Dill S, Colbert RA, Failla L, Kost B, O'Brien M, Reynolds JC,
128. Liu Y, Jesus AA, Marrero B, Yang D, Ramsey SE, Sanchez Folio LR, Calvo KR, Paul SM, Weir N, Brofferio A, Soldatos A,
GAM, Tenbrock K, Wittkowski H, Jones OY, Kuehn HS, Lee Biancotto A, Cowen EW, Digiovanna JJ, Gadina M, Lipton AJ,
CR, DiMattia MA, Cowen EW, Gonzalez B, Palmer I, Hadigan C, Holland SM, Fontana J, Alawad AS, Brown RJ,
DiGiovanna JJ, Biancotto A, Kim H, Tsai WL, Trier AM, Rother KI, Heller T, Brooks KM, Kumar P, Brooks SR,
Huang Y, Stone DL, Hill S, Kim HJ, St Hilaire C, Gurprasad S, Waldman M, Singh HK, Nickeleit V, Silk M, Prakash A, Janes
Plass N, Chapelle D, Horkayne-Szakaly I, Foell D, Barysenka A, JM, Ozen S, Wakim PG, Brogan PA, Macias WL, Goldbach-
Candotti F, Holland SM, Hughes JD, Mehmet H, Issekutz AC, Mansky R (2018) JAK1/2 inhibition with baricitinib in the treat-
Raffeld M, McElwee J, Fontana JR, Minniti CP, Moir S, Kastner ment of autoinflammatory interferonopathies. J Clin Invest
DL, Gadina M, Steven AC, Wingfield PT, Brooks SR, 128(7):3041–3052. https://doi.org/10.1172/JCI98814
Rosenzweig SD, Fleisher TA, Deng Z, Boehm M, Paller AS, 138. Fremond ML, Rodero MP, Jeremiah N, Belot A, Jeziorski E,
Goldbach-Mansky R (2014) Activated STING in a vascular and Duffy D, Bessis D, Cros G, Rice GI, Charbit B, Hulin A,
pulmonary syndrome. N Engl J Med 371(6):507–518. https://doi. Khoudour N, Caballero CM, Bodemer C, Fabre M, Berteloot L,
org/10.1056/NEJMoa1312625 Le Bourgeois M, Reix P, Walzer T, Moshous D, Blanche S,
129. Navarro V, Scott C, Briggs TA, Barete S, Frances C, Lebon P, Fischer A, Bader-Meunier B, Rieux-Laucat F, Crow YJ, Neven
Maisonobe T, Rice GI, Wouters CH, Crow YJ (2008) Two further B (2016) Efficacy of the Janus kinase 1/2 inhibitor ruxolitinib in
cases of spondyloenchondrodysplasia (SPENCD) with immune the treatment of vasculopathy associated with TMEM173-
dysregulation. Am J Med Genet A 146A(21):2810–2815. https:// activating mutations in 3 children. J Allergy Clin Immunol
doi.org/10.1002/ajmg.a.32518 138(6):1752–1755. https://doi.org/10.1016/j.jaci.2016.07.015
130. Costa-Reis P, Sullivan KE (2017) Monogenic lupus: it’s all new! 139. Tungler V, Konig N, Gunther C, Engel K, Fiehn C, Smitka M, von
Curr Opin Immunol 49:87–95. https://doi.org/10.1016/j.coi.2017. der Hagen M, Berner R, Lee-Kirsch MA (2016) Response to:
10.008 'JAK inhibition in STING-associated interferonopathy' by Crow
131. Rutsch F, MacDougall M, Lu C, Buers I, Mamaeva O, Nitschke et al. cJ. Clin Invest 128(7):2760–2762. https://doi.org/10.1172/
Y, Rice GI, Erlandsen H, Kehl HG, Thiele H, Nurnberg P, Hohne JCI121526
W, Crow YJ, Feigenbaum A, Hennekam RC (2015) A specific 140. Meesilpavikkai K, Dik WA, Schrijver B, van Helden-Meeuwsen
IFIH1 gain-of-function mutation causes Singleton-Merten syn- CG, Versnel MA, van Hagen PM, Bijlsma EK, Ruivenkamp
drome. Am J Hum Genet 96(2):275–282. https://doi.org/10. CAL, Oele MJ, Dalm V (2019) Efficacy of baricitinib in the treat-
1016/j.ajhg.2014.12.014 ment of chilblains associated with Aicardi-Goutieres syndrome, a
132. Jang MA, Kim EK, Now H, Nguyen NT, Kim WJ, Yoo JY, Lee J, type I interferonopathy. Arthritis Rheumatol (Hoboken, NJ) 71(5):
Jeong YM, Kim CH, Kim OH, Sohn S, Nam SH, Hong Y, Lee 829–831. https://doi.org/10.1002/art.40805
YS, Chang SA, Jang SY, Kim JW, Lee MS, Lim SY, Sung KS,
141. Briand C, Fremond ML, Bessis D, Carbasse A, Rice GI, Bondet
Park KT, Kim BJ, Lee JH, Kim DK, Kee C, Ki CS (2015)
V, Duffy D, Chatenoud L, Blanche S, Crow YJ, Neven B (2019)
Mutations in DDX58, which encodes RIG-I, cause atypical
Efficacy of JAK1/2 inhibition in the treatment of chilblain lupus
Singleton-Merten syndrome. Am J Hum Genet 96(2):266–274.
due to TREX1 deficiency. Ann Rheum Dis 78(3):431–433.
https://doi.org/10.1016/j.ajhg.2014.11.019
https://doi.org/10.1136/annrheumdis-2018-214037
133. Shipman L (2014) Cytokines: ISG15–human deficiency reveals
142. Zimmermann N, Wolf C, Schwenke R, Luth A, Schmidt F, Engel
new proficiency. Nat Rev Immunol 14(12):780–781. https://doi.
K, Lee-Kirsch MA, Gunther C (2019) Assessment of clinical re-
org/10.1038/nri3767
sponse to Janus kinase inhibition in patients with familial chilblain
134. Cavalcante MP, Brunelli JB, Miranda CC, Novak GV, Malle L,
lupus and TREX1 mutation. JAMA Dermatol 155(3):342–346.
Aikawa NE, Jesus AA, Silva CA (2016) CANDLE syndrome:
https://doi.org/10.1001/jamadermatol.2018.5077
chronic atypical neutrophilic dermatosis with lipodystrophy and
elevated temperature-a rare case with a novel mutation. Eur J 143. Fragoulis GE, McInnes IB, Siebert S (2019) JAK-inhibitors. New
Pediatr 175(5):735–740. https://doi.org/10.1007/s00431-015- players in the field of immune-mediated diseases, beyond rheuma-
2668-4 toid arthritis. Rheumatology (Oxford, England) 58(Suppl 1):i43–
135. Brehm A, Liu Y, Sheikh A, Marrero B, Omoyinmi E, Zhou Q, i54. https://doi.org/10.1093/rheumatology/key276
Montealegre G, Biancotto A, Reinhardt A, Almeida de Jesus A, 144. Winthrop KL (2017) The emerging safety profile of JAK inhibi-
Pelletier M, Tsai WL, Remmers EF, Kardava L, Hill S, Kim H, tors in rheumatic disease. Nat Rev Rheumatol 13(4):234–243.
Lachmann HJ, Megarbane A, Chae JJ, Brady J, Castillo RD, https://doi.org/10.1038/nrrheum.2017.23
Brown D, Casano AV, Gao L, Chapelle D, Huang Y, Stone D, 145. Bronson PG, Chaivorapol C, Ortmann W, Behrens TW, Graham
Chen Y, Sotzny F, Lee CC, Kastner DL, Torrelo A, Zlotogorski A, RR (2012) The genetics of type I interferon in systemic lupus
Moir S, Gadina M, McCoy P, Wesley R, Rother KI, Hildebrand erythematosus. Curr Opin Immunol 24(5):530–537. https://doi.
PW, Brogan P, Kruger E, Aksentijevich I, Goldbach-Mansky R org/10.1016/j.coi.2012.07.008
(2015) Additive loss-of-function proteasome subunit mutations in 146. Psarras A, Emery P, Vital EM (2017) Type I interferon-mediated
CANDLE/PRAAS patients promote type I IFN production. J Clin autoimmune diseases: pathogenesis, diagnosis and targeted thera-
Invest 125(11):4196–4211. https://doi.org/10.1172/JCI81260 py. Rheumatology (Oxford, England) 56(10):1662–1675. https://
136. Alsohime F, Martin-Fernandez M, Temsah MH, Alabdulhafid M, doi.org/10.1093/rheumatology/kew431
Le Voyer T, Alghamdi M, Qiu X, Alotaibi N, Alkahtani A, Buta 147. Guiducci C, Gong M, Xu Z, Gill M, Chaussabel D, Meeker T,
S, Jouanguy E, Al-Eyadhy A, Gruber C, Hasan GM, Bashiri FA, Chan JH, Wright T, Punaro M, Bolland S, Soumelis V,
Halwani R, Hassan HH, Al-Muhsen S, Alkhamis N, Alsum Z, Banchereau J, Coffman RL, Pascual V, Barrat FJ (2010) TLR
Casanova JL, Bustamante J, Bogunovic D, Alangari AA (2020) recognition of self nucleic acids hampers glucocorticoid activity
Clinic Rev Allerg Immunol

in lupus. Nature 465(7300):937–941. https://doi.org/10.1038/ 161. McBride JM, Jiang J, Abbas AR, Morimoto A, Li J, Maciuca R,
nature09102 Townsend M, Wallace DJ, Kennedy WP, Drappa J (2012) Safety
148. Rizza P, Moretti F, Capone I, Belardelli F (2015) Role of type I and pharmacodynamics of rontalizumab in patients with systemic
interferon in inducing a protective immune response: perspectives lupus erythematosus: results of a phase I, placebo-controlled, dou-
for clinical applications. Cytokine Growth Factor Rev 26(2):195– ble-blind, dose-escalation study. Arthritis Rheum 64(11):3666–
201. https://doi.org/10.1016/j.cytogfr.2014.10.002 3676. https://doi.org/10.1002/art.34632
149. Wolf SJ, Estadt SN, Theros J, Moore T, Ellis J, Liu J, Reed TJ, 162. Khamashta M, Merrill JT, Werth VP, Furie R, Kalunian K, Illei
Jacob CO, Gudjonsson JE, Kahlenberg JM (2019) Ultraviolet light GG, Drappa J, Wang L, Greth W, investigators CDs (2016)
induces increased T cell activation in lupus-prone mice via type I Sifalimumab, an anti-interferon-alpha monoclonal antibody, in
IFN-dependent inhibition of T regulatory cells. J Autoimmun 103: moderate to severe systemic lupus erythematosus: a randomised,
102291. https://doi.org/10.1016/j.jaut.2019.06.002 double-blind, placebo-controlled study. Ann Rheum Dis 75 (11):
150. Liu M, Guo Q, Wu C, Sterlin D, Goswami S, Zhang Y, Li T, Bao 1909–1916. https://doi.org/10.1136/annrheumdis-2015-208562
C, Shen N, Fu Q, Zhang X (2019) Type I interferons promote the 163. Chasset F, Arnaud L (2018) Targeting interferons and their path-
survival and proinflammatory properties of transitional B cells in ways in systemic lupus erythematosus. Autoimmun Rev 17(1):
systemic lupus erythematosus patients. Cell Mol Immunol 16(4): 44–52. https://doi.org/10.1016/j.autrev.2017.11.009
367–379. https://doi.org/10.1038/s41423-018-0010-6 164. Ducreux J, Houssiau FA, Vandepapeliere P, Jorgensen C, Lazaro
151. Dieudonne Y, Gies V, Guffroy A, Keime C, Bird AK, Liesveld J, E, Spertini F, Colaone F, Roucairol C, Laborie M, Croughs T,
Barnas JL, Poindron V, Douiri N, Soulas-Sprauel P, Martin T, Meffre Grouard-Vogel G, Lauwerys BR (2016) Interferon alpha kinoid
E, Anolik JH, Korganow AS (2019) Transitional B cells in quiescent induces neutralizing anti-interferon alpha antibodies that decrease
SLE: An early checkpoint imprinted by IFN. J Autoimmun 102:150– the expression of interferon-induced and B cell activation associ-
158. https://doi.org/10.1016/j.jaut.2019.05.002 ated transcripts: analysis of extended follow-up data from the in-
152. Hamilton JA, Hsu HC, Mountz JD (2019) Autoreactive B cells in terferon alpha kinoid phase I/II study. Rheumatology (Oxford,
SLE, villains or innocent bystanders? Immunol Rev 292(1):120– England) 55(10):1901–1905. https://doi.org/10.1093/
138. https://doi.org/10.1111/imr.12815 rheumatology/kew262
165. Houssiau FA, Thanou A, Mazur M, Ramiterre E, Gomez Mora
153. Chyuan IT, Tzeng HT, Chen JY (2019) Signaling pathways of
DA, Misterska-Skora M, Perich-Campos RA, Smakotina SA,
type I and type III interferons and targeted therapies in systemic
Cerpa Cruz S, Louzir B, Croughs T, Tee ML (2020) IFN-alpha
lupus erythematosus. Cells 8(9). https://doi.org/10.3390/
kinoid in systemic lupus erythematosus: results from a phase IIb,
cells8090963
randomised, placebo-controlled study. Ann Rheum Dis 79(3):
154. Sarkar MK, Hile GA, Tsoi LC, Xing X, Liu J, Liang Y, Berthier
347–355. https://doi.org/10.1136/annrheumdis-2019-216379
CC, Swindell WR, Patrick MT, Shao S, Tsou PS, Uppala R,
166. Furie R, Khamashta M, Merrill JT, Werth VP, Kalunian K,
Beamer MA, Srivastava A, Bielas SL, Harms PW, Getsios S,
Brohawn P, Illei GG, Drappa J, Wang L, Yoo S, Investigators
Elder JT, Voorhees JJ, Gudjonsson JE, Kahlenberg JM (2018)
CDS (2017) Anifrolumab, an anti-interferon-alpha receptor
Photosensitivity and type I IFN responses in cutaneous lupus are
monoclonal antibody, in moderate-to-severe systemic lupus ery-
driven by epidermal-derived interferon kappa. Ann Rheum Dis
thematosus. Arthritis & rheumatology (Hoboken, NJ) 69(2):376–
77(11):1653–1664. https://doi.org/10.1136/annrheumdis-2018-
386. https://doi.org/10.1002/art.39962
213197
167. Morand EF, Trasieva T, Berglind A, Illei GG, Tummala R (2018)
155. Lo MS (2018) Insights gained from the study of pediatric systemic Lupus low disease activity state (LLDAS) attainment discrimi-
lupus erythematosus. Front Immunol 9:1278. https://doi.org/10. nates responders in a systemic lupus erythematosus trial: post-
3389/fimmu.2018.01278 hoc analysis of the phase IIb MUSE trial of anifrolumab. Ann
156. Rice GI, Melki I, Fremond ML, Briggs TA, Rodero MP, Rheum Dis 77(5):706–713. https://doi.org/10.1136/
Kitabayashi N, Oojageer A, Bader-Meunier B, Belot A, annrheumdis-2017-212504
Bodemer C, Quartier P, Crow YJ (2017) Assessment of type I 168. Salmon JE, Niewold TB (2020) A successful trial for lupus - how
interferon signaling in pediatric inflammatory disease. J Clin good is good enough? N Engl J Med 382(3):287–288. https://doi.
Immunol 37(2):123–132. https://doi.org/10.1007/s10875-016- org/10.1056/NEJMe1915490
0359-1 169. Furie R, Werth VP, Merola JF, Stevenson L, Reynolds TL, Naik
157. Wahadat MJ, Bodewes ILA, Maria NI, van Helden-Meeuwsen H, Wang W, Christmann R, Gardet A, Pellerin A, Hamann S,
CG, van Dijk-Hummelman A, Steenwijk EC, Kamphuis S, Auluck P, Barbey C, Gulati P, Rabah D, Franchimont N (2019)
Versnel MA (2018) Type I IFN signature in childhood-onset sys- Monoclonal antibody targeting BDCA2 ameliorates skin lesions
temic lupus erythematosus: a conspiracy of DNA- and RNA- in systemic lupus erythematosus. J Clin Invest 129(3):1359–1371.
sensing receptors? Arthritis Res Therapy 20(1):4. https://doi.org/ https://doi.org/10.1172/JCI124466
10.1186/s13075-017-1501-z 170. Zhan Y, Carrington EM, Ko HJ, Vikstrom IB, Oon S, Zhang JG,
158. Midgley A, Thorbinson C, Beresford MW (2012) Expression of Vremec D, Brady JL, Bouillet P, Wu L, Huang DC, Wicks IP,
toll-like receptors and their detection of nuclear self-antigen lead- Morand EF, Strasser A, Lew AM (2015) Bcl-2 antagonists kill
ing to immune activation in JSLE. Rheumatology (Oxford, plasmacytoid dendritic cells from lupus-prone mice and dampen
England) 51(5):824–832. https://doi.org/10.1093/rheumatology/ interferon-alpha production. Arthritis & rheumatology (Hoboken,
ker400 NJ) 67(3):797–808. https://doi.org/10.1002/art.38966
159. Fava A, Petri M (2019) Systemic lupus erythematosus: diagnosis 171. Lu P, Fleischmann R, Curtis C, Ignatenko S, Clarke SH, Desai M,
and clinical management. J Autoimmun 96:1–13. https://doi.org/ Wong SL, Grebe KM, Black K, Zeng J, Stolzenbach J, Medema
10.1016/j.jaut.2018.11.001 JK (2018) Safety and pharmacodynamics of venetoclax (ABT-
160. Kalunian KC, Merrill JT, Maciuca R, McBride JM, Townsend 199) in a randomized single and multiple ascending dose study
MJ, Wei X, Davis JC Jr, Kennedy WP (2016) A phase II study in women with systemic lupus erythematosus. Lupus 27(2):290–
of the efficacy and safety of rontalizumab (rhuMAb interferon- 302. https://doi.org/10.1177/0961203317719334
alpha) in patients with systemic lupus erythematosus (ROSE). 172. Nader A, Minocha M, Othman AA (2020) Exposure-response
Ann Rheum Dis 75(1):196–202. https://doi.org/10.1136/ analyses of the effects of venetoclax, a selective BCL-2 inhibitor,
annrheumdis-2014-206090 on B-lymphocyte and total lymphocyte counts in women with
Clinic Rev Allerg Immunol

systemic lupus erythematosus. Clin Pharmacokinet 59(3):335– arthritis. Arthritis & rheumatology (Hoboken, NJ) 69(5):932–
347. https://doi.org/10.1007/s40262-019-00818-5 942. https://doi.org/10.1002/art.40054
173. Li J, Wang X, Zhang F, Yin H (2013) Toll-like receptors as ther- 185. Westhovens R (2019) Clinical efficacy of new JAK inhibitors
apeutic targets for autoimmune connective tissue diseases. under development. Just more of the same? Rheumatology
Pharmacol Ther 138(3):441–451. https://doi.org/10.1016/j. (Oxford, England) 58(Suppl 1):i27–i33. https://doi.org/10.1093/
pharmthera.2013.03.003 rheumatology/key256
174. Dudhgaonkar S, Ranade S, Nagar J, Subramani S, Prasad DS, 186. Harigai M (2019) Growing evidence of the safety of JAK inhibi-
Karunanithi P, Srivastava R, Venkatesh K, Selvam S, tors in patients with rheumatoid arthritis. Rheumatology (Oxford,
Krishnamurthy P, Mariappan TT, Saxena A, Fan L, Stetsko DK, England) 58(Suppl 1):i34–i42. https://doi.org/10.1093/
Holloway DA, Li X, Zhu J, Yang WP, Ruepp S, Nair S, Santella J, rheumatology/key287
Duncia J, Hynes J, McIntyre KW, Carman JA (2017) Selective 187. Kjelgaard-Petersen CF, Platt A, Braddock M, Jenkins MA, Musa
IRAK4 inhibition attenuates disease in murine lupus models and K, Graham E, Gantzel T, Slynn G, Weinblatt ME, Karsdal MA,
demonstrates steroid sparing activity. J Immunol 198(3):1308– Thudium CS, Bay-Jensen AC (2018) Translational biomarkers
1319. https://doi.org/10.4049/jimmunol.1600583 and ex vivo models of joint tissues as a tool for drug development
175. Haselmayer P, Camps M, Liu-Bujalski L, Nguyen N, Morandi F, in rheumatoid arthritis. Arthritis & rheumatology (Hoboken, NJ)
Head J, O'Mahony A, Zimmerli SC, Bruns L, Bender AT, 70(9):1419–1428. https://doi.org/10.1002/art.40527
Schroeder P, Grenningloh R (2019) Efficacy and pharmacody- 188. Schafer PH, Kivitz AJ, Ma J, Korish S, Sutherland D, Li L,
namic modeling of the BTK inhibitor evobrutinib in autoimmune Azaryan A, Kosek J, Adams M, Capone L, Hur EM, Hough
disease models. J Immunol 202(10):2888–2906. https://doi.org/ DR, Ringheim GE (2020) Spebrutinib (CC-292) affects markers
10.4049/jimmunol.1800583 of B cell activation, chemotaxis, and osteoclasts in patients with
176. Klavdianou K, Lazarini A, Fanouriakis A (2020) Targeted biolog- rheumatoid arthritis: results from a mechanistic study.
ic therapy for systemic lupus erythematosus: emerging pathways Rheumatology and therapy 7(1):101–119. https://doi.org/10.
and drug pipeline. BioDrugs. https://doi.org/10.1007/s40259-020- 1007/s40744-019-00182-7
00405-2 189. Nehmar R, Mariotte A, de Cauwer A, Sibilia J, Bahram S, Georgel
177. You H, Xu D, Zhao J, Li J, Wang Q, Tian X, Li M, Zeng X (2020) P (2018) Therapeutic perspectives for interferons and plasmacytoid
JAK inhibitors: prospects in connective tissue diseases. Clin Rev dendritic cells in rheumatoid arthritis. Trends Mol Med 24(4):338–
Allergy Immunol. https://doi.org/10.1007/s12016-020-08786-6 347. https://doi.org/10.1016/j.molmed.2018.02.001
178. Malemud CJ (2018) The role of the JAK/STAT signal pathway in 190. van Holten J, Pavelka K, Vencovsky J, Stahl H, Rozman B,
rheumatoid arthritis. Ther Adv Musculoskelet Dis 10(5–6):117– Genovese M, Kivitz AJ, Alvaro J, Nuki G, Furst DE, Herrero-
127. https://doi.org/10.1177/1759720X18776224 Beaumont G, McInnes IB, Musikic P, Tak PP (2005) A
179. Hojjati SM, Heidari B, Babaei M (2016) Development of rheuma- multicentre, randomised, double blind, placebo controlled phase
toid arthritis during treatment of multiple sclerosis with interferon II study of subcutaneous interferon beta-1a in the treatment of
beta 1-a. coincidence of two conditions or a complication of treat- patients with active rheumatoid arthritis. Ann Rheum Dis 64(1):
ment: a case report. J Adv Res 7(5):611–613. https://doi.org/10. 64–69. https://doi.org/10.1136/ard.2003.020347
1016/j.jare.2016.06.004 191. Mauro A, Rigante D, Cimaz R (2017) Investigational drugs for
180. Lubbers J, Brink M, van de Stadt LA, Vosslamber S, Wesseling treatment of juvenile idiopathic arthritis. Expert Opin Investig
JG, van Schaardenburg D, Rantapaa-Dahlqvist S, Verweij CL Drugs 26(4):381–387. https://doi.org/10.1080/13543784.2017.
(2013) The type I IFN signature as a biomarker of preclinical 1301929
rheumatoid arthritis. Ann Rheum Dis 72(5):776–780. https://doi. 192. Bracaglia C, de Graaf K, Pires Marafon D, Guilhot F, Ferlin W,
org/10.1136/annrheumdis-2012-202753 Prencipe G, Caiello I, Davi S, Schulert G, Ravelli A, Grom AA, de
181. Nader A, Stodtmann S, Friedel A, Mohamed MF, Othman AA Min C, De Benedetti F (2017) Elevated circulating levels of
(2020) Pharmacokinetics of Upadacitinib in healthy subjects and interferon-gamma and interferon-gamma-induced chemokines
subjects with rheumatoid arthritis, Crohn’s disease, ulcerative co- characterise patients with macrophage activation syndrome com-
litis, or atopic dermatitis: population analyses of phase 1 and 2 plicating systemic juvenile idiopathic arthritis. Ann Rheum Dis
clinical trials. J Clin Pharmacol 60(4):528–539. https://doi.org/ 76(1):166–172. https://doi.org/10.1136/annrheumdis-2015-
10.1002/jcph.1550 209020
182. Genovese MC, Kalunian K, Gottenberg JE, Mozaffarian N, 193. Gattorno M, Chicha L, Gregorio A, Ferlito F, Rossi F, Jarrossay
Bartok B, Matzkies F, Gao J, Guo Y, Tasset C, Sundy JS, de D, Lanzavecchia A, Martini A, Manz MG (2007) Distinct expres-
Vlam K, Walker D, Takeuchi T (2019) Effect of filgotinib vs sion pattern of IFN-alpha and TNF-alpha in juvenile idiopathic
placebo on clinical response in patients with moderate to severe arthritis synovial tissue. Rheumatology (Oxford, England) 46(4):
rheumatoid arthritis refractory to disease-modifying antirheumatic 657–665. https://doi.org/10.1093/rheumatology/kel346
drug therapy: the FINCH 2 randomized clinical trial. JAMA 194. Myles A, Aggarwal A (2011) Expression of toll-like receptors 2
322(4):315–325. https://doi.org/10.1001/jama.2019.9055 and 4 is increased in peripheral blood and synovial fluid mono-
183. Takeuchi T, Tanaka Y, Tanaka S, Kawakami A, Iwasaki M, cytes of patients with enthesitis-related arthritis subtype of juvenile
Katayama K, Rokuda M, Izutsu H, Ushijima S, Kaneko Y, idiopathic arthritis. Rheumatology (Oxford, England) 50(3):481–
Shiomi T, Yamada E, van der Heijde D (2019) Efficacy and safety 488. https://doi.org/10.1093/rheumatology/keq362
of peficitinib (ASP015K) in patients with rheumatoid arthritis and 195. Stanford SM, Bottini N (2014) PTPN22: the archetypal non-HLA
an inadequate response to methotrexate: results of a phase III autoimmunity gene. Nat Rev Rheumatol 10(10):602–611. https://
randomised, double-blind, placebo-controlled trial (RAJ4) in doi.org/10.1038/nrrheum.2014.109
Japan. Ann Rheum Dis 78(10):1305–1319. https://doi.org/10. 196. Ruperto N, Brunner HI, Zuber Z, Tzaribachev N, Kingsbury DJ,
1136/annrheumdis-2019-215164 Foeldvari I, Horneff G, Smolewska E, Vehe RK, Hazra A, Wang
184. Genovese MC, Greenwald M, Codding C, Zubrzycka- R, Mebus CA, Alvey C, Lamba M, Krishnaswami S, Stock TC,
Sienkiewicz A, Kivitz AJ, Wang A, Shay K, Wang X, Garg JP, Wang M, Suehiro R, Martini A, Lovell DJ, Pediatric
Cardiel MH (2017) Peficitinib, a JAK inhibitor, in combination Rheumatology International Trials O, Pediatric Rheumatology
with limited conventional synthetic disease-modifying antirheu- Collaborative Study G (2017) Pharmacokinetic and safety profile
matic drugs in the treatment of moderate-to-severe rheumatoid of tofacitinib in children with polyarticular course juvenile
Clinic Rev Allerg Immunol

idiopathic arthritis: results of a phase 1, open-label, multicenter 209. Higgs BW, Zhu W, Morehouse C, White WI, Brohawn P, Guo X,
study. Pediatr Rheumatol Online J 15(1):86. https://doi.org/10. Rebelatto M, Le C, Amato A, Fiorentino D, Greenberg SA,
1186/s12969-017-0212-y Drappa J, Richman L, Greth W, Jallal B, Yao Y (2014) A phase
197. Huang Z, Lee PY, Yao X, Zheng S, Li T (2019) Tofacitinib treat- 1b clinical trial evaluating sifalimumab, an anti-IFN-alpha mono-
ment of refractory systemic juvenile idiopathic arthritis. Pediatrics clonal antibody, shows target neutralisation of a type I IFN signa-
143(5). https://doi.org/10.1542/peds.2018-2845 ture in blood of dermatomyositis and polymyositis patients. Ann
198. Feldman BM, Rider LG, Reed AM, Pachman LM (2008) Juvenile Rheum Dis 73(1):256–262. https://doi.org/10.1136/annrheumdis-
dermatomyositis and other idiopathic inflammatory myopathies of 2012-202794
childhood. Lancet (London, England) 371(9631):2201–2212. 210. Yao Y, Liu Z, Jallal B, Shen N, Ronnblom L (2013) Type I
https://doi.org/10.1016/S0140-6736(08)60955-1 interferons in Sjogren’s syndrome. Autoimmun Rev 12(5):558–
199. Niewold TB, Kariuki SN, Morgan GA, Shrestha S, Pachman LM 566. https://doi.org/10.1016/j.autrev.2012.10.006
(2009) Elevated serum interferon-alpha activity in juvenile derma- 211. Zheng L, Zhang Z, Yu C, Tu L, Zhong L, Yang C (2009)
tomyositis: associations with disease activity at diagnosis and after Association between IFN-alpha and primary Sjogren’s syndrome.
thirty-six months of therapy. Arthritis Rheum 60(6):1815–1824. Oral Surg Oral Med Oral Pathol Oral Radiol Endod 107(1):e12–
https://doi.org/10.1002/art.24555 e18. https://doi.org/10.1016/j.tripleo.2008.09.015
200. Moneta GM, Pires Marafon D, Marasco E, Rosina S, Verardo M, 212. Brkic Z, Maria NI, van Helden-Meeuwsen CG, van de Merwe JP,
Fiorillo C, Minetti C, Bracci-Laudiero L, Ravelli A, De Benedetti van Daele PL, Dalm VA, Wildenberg ME, Beumer W, Drexhage
F, Nicolai R (2019) Muscle expression of type I and type II inter- HA, Versnel MA (2013) Prevalence of interferon type I signature
ferons is increased in juvenile dermatomyositis and related to clin- in CD14 monocytes of patients with Sjogren’s syndrome and as-
ical and histologic features. Arthritis & rheumatology (Hoboken, sociation with disease activity and BAFF gene expression. Ann
NJ) 71(6):1011–1021. https://doi.org/10.1002/art.40800 Rheum Dis 72(5):728–735. https://doi.org/10.1136/annrheumdis-
201. O'Connor KA, Abbott KA, Sabin B, Kuroda M, Pachman LM 2012-201381
(2006) MxA gene expression in juvenile dermatomyositis periph- 213. Gottenberg JE, Cagnard N, Lucchesi C, Letourneur F, Mistou S,
eral blood mononuclear cells: association with muscle involve- Lazure T, Jacques S, Ba N, Ittah M, Lepajolec C, Labetoulle M,
ment. Clin Immunol 120(3):319–325. https://doi.org/10.1016/j. Ardizzone M, Sibilia J, Fournier C, Chiocchia G, Mariette X
clim.2006.05.011 (2006) Activation of IFN pathways and plasmacytoid dendritic
202. Baechler EC, Bilgic H, Reed AM (2011) Type I interferon path- cell recruitment in target organs of primary Sjogren’s syndrome.
way in adult and juvenile dermatomyositis. Arthritis research & Proc Natl Acad Sci U S A 103(8):2770–2775. https://doi.org/10.
therapy 13(6):249. https://doi.org/10.1186/ar3531 1073/pnas.0510837103
203. Cappelletti C, Baggi F, Zolezzi F, Biancolini D, Beretta O, Severa M, 214. Bave U, Nordmark G, Lovgren T, Ronnelid J, Cajander S,
Coccia EM, Confalonieri P, Morandi L, Mora M, Mantegazza R, Eloranta ML, Alm GV, Ronnblom L (2005) Activation of the type
Bernasconi P (2011) Type I interferon and toll-like receptor expres- I interferon system in primary Sjogren’s syndrome: a possible
sion characterizes inflammatory myopathies. Neurology 76(24): etiopathogenic mechanism. Arthritis Rheum 52(4):1185–1195.
2079–2088. https://doi.org/10.1212/WNL.0b013e31821f440a https://doi.org/10.1002/art.20998
204. Piper CJM, Wilkinson MGL, Deakin CT, Otto GW, Dowle S, 215. Lee J, Lee J, Kwok SK, Baek S, Jang SG, Hong SM, Min JW,
Duurland CL, Adams S, Marasco E, Rosser EC, Radziszewska Choi SS, Lee J, Cho ML, Park SH (2018) JAK-1 inhibition sup-
A, Carsetti R, Ioannou Y, Beales PL, Kelberman D, Isenberg DA, presses interferon-induced BAFF production in human salivary
Mauri C, Nistala K, Wedderburn LR (2018) CD19(+ gland: potential therapeutic strategy for primary Sjogren’s syn-
)CD24(hi)CD38(hi) B cells are expanded in juvenile dermatomy- drome. Arthritis & rheumatology (Hoboken, NJ) 70(12):2057–
ositis and exhibit a pro-inflammatory phenotype after activation 2066. https://doi.org/10.1002/art.40589
through toll-like receptor 7 and interferon-alpha. Front Immunol 216. Charras A, Arvaniti P, Le Dantec C, Arleevskaya MI, Zachou K,
9:1372. https://doi.org/10.3389/fimmu.2018.01372 Dalekos GN, Bordon A, Renaudineau Y (2020) JAK inhibitors
205. Gitiaux C, Latroche C, Weiss-Gayet M, Rodero MP, Duffy D, suppress innate epigenetic reprogramming: a promise for patients
Bader-Meunier B, Glorion C, Nusbaum P, Bodemer C, with Sjogren’s syndrome. Clin Rev Allergy Immunol 58(2):182–
Mouchiroud G, Chelly J, Germain S, Desguerre I, Chazaud B 193. https://doi.org/10.1007/s12016-019-08743-y
(2018) Myogenic progenitor cells exhibit type I interferon-driven 217. Bodewes ILA, Huijser E, van Helden-Meeuwsen CG, Tas L,
proangiogenic properties and molecular signature during juvenile Huizinga R, Dalm V, van Hagen PM, Groot N, Kamphuis S, van
dermatomyositis. Arthritis & rheumatology (Hoboken, NJ) 70(1): Daele PLA, Versnel MA (2018) TBK1: a key regulator and poten-
134–145. https://doi.org/10.1002/art.40328 tial treatment target for interferon positive Sjogren’s syndrome, sys-
206. Ladislau L, Suarez-Calvet X, Toquet S, Landon-Cardinal O, temic lupus erythematosus and systemic sclerosis. J Autoimmun 91:
Amelin D, Depp M, Rodero MP, Hathazi D, Duffy D, Bondet 97–102. https://doi.org/10.1016/j.jaut.2018.02.001
V, Preusse C, Bienvenu B, Rozenberg F, Roos A, Benjamim 218. Bodewes ILA, Gottenberg JE, van Helden-Meeuwsen CG,
CF, Gallardo E, Illa I, Mouly V, Stenzel W, Butler-Browne G, Mariette X, Versnel MA (2020) Hydroxychloroquine treatment
Benveniste O, Allenbach Y (2018) JAK inhibitor improves type I downregulates systemic interferon activation in primary Sjogren's
interferon induced damage: proof of concept in dermatomyositis. syndrome in the JOQUER randomized trial. Rheumatology
Brain 141(6):1609–1621. https://doi.org/10.1093/brain/awy105 (Oxford, England) 59(1):107–111. https://doi.org/10.1093/
207. Moghadam-Kia S, Charlton D, Aggarwal R, Oddis CV (2019) rheumatology/kez242
Management of refractory cutaneous dermatomyositis: potential 219. Meijer JM, Pijpe J, Bootsma H, Vissink A, Kallenberg CG (2007)
role of Janus kinase inhibition with tofacitinib. Rheumatology The future of biologic agents in the treatment of Sjogren’s syn-
(Oxford, England) 58(6):1011–1015. https://doi.org/10.1093/ drome. Clin Rev Allergy Immunol 32(3):292–297. https://doi.org/
rheumatology/key366 10.1007/s12016-007-8005-6
208. Papadopoulou C, Hong Y, Omoyinmi E, Brogan PA, Eleftheriou 220. Skaug B, Assassi S (2019) Type I interferon dysregulation in
D (2019) Janus kinase 1/2 inhibition with baricitinib in the treat- systemic sclerosis Cytokine:154635. https://doi.org/10.1016/j.
ment of juvenile dermatomyositis. Brain 142(3):e8. https://doi. cyto.2018.12.018
org/10.1093/brain/awz005
Clinic Rev Allerg Immunol

221. Wu M, Assassi S (2013) The role of type 1 interferon in systemic monoclonal antibody MEDI-546 in subjects with systemic sclero-
sclerosis. Front Immunol 4:266. https://doi.org/10.3389/fimmu. sis: a phase 1, multicenter, open label study. Arthritis research &
2013.00266 therapy 16(1):R57. https://doi.org/10.1186/ar4492
222. Assassi S, Mayes MD, Arnett FC, Gourh P, Agarwal SK, 226. Ah Kioon MD, Tripodo C, Fernandez D, Kirou KA, Spiera RF,
McNearney TA, Chaussabel D, Oommen N, Fischbach M, Shah Crow MK, Gordon JK, Barrat FJ (2018) Plasmacytoid dendritic
KR, Charles J, Pascual V, Reveille JD, Tan FK (2010) Systemic cells promote systemic sclerosis with a key role for TLR8. Sci
sclerosis and lupus: points in an interferon-mediated continuum. Transl Med 10(423). https://doi.org/10.1126/scitranslmed.
Arthritis Rheum 62(2):589–598. https://doi.org/10.1002/art. aam8458
27224 227. Cattalini M, Soliani M, Caparello MC, Cimaz R (2019) Sex dif-
223. Liu X, Mayes MD, Tan FK, Wu M, Reveille JD, Harper BE, ferences in pediatric rheumatology. Clin Rev Allergy Immunol
Draeger HT, Gonzalez EB, Assassi S (2013) Correlation of 56(3):293–307. https://doi.org/10.1007/s12016-017-8642-3
interferon-inducible chemokine plasma levels with disease sever- 228. Axtell RC, Raman C (2012) Janus-like effects of type I interferon
ity in systemic sclerosis. Arthritis Rheum 65(1):226–235. https:// in autoimmune diseases. Immunol Rev 248(1):23–35. https://doi.
doi.org/10.1002/art.37742 org/10.1111/j.1600-065X.2012.01131.x
224. Guo X, Higgs BW, Bay-Jensen AC, Karsdal MA, Yao Y, Roskos 229. Axtell RC, Raman C, Steinman L (2013) Type I interferons: ben-
LK, White WI (2015) Suppression of T cell activation and colla- eficial in Th1 and detrimental in Th17 autoimmunity. Clin Rev
gen accumulation by an anti-IFNAR1 mAb, anifrolumab, in adult Allergy Immunol 44(2):114–120. https://doi.org/10.1007/s12016-
patients with systemic sclerosis. The Journal of investigative der- 011-8296-5
matology 135(10):2402–2409. https://doi.org/10.1038/jid.2015.
188
225. Goldberg A, Geppert T, Schiopu E, Frech T, Hsu V, Simms RW, Publisher’s Note Springer Nature remains neutral with regard to jurisdic-
Peng SL, Yao Y, Elgeioushi N, Chang L, Wang B, Yoo S (2014) tional claims in published maps and institutional affiliations.
Dose-escalation of human anti-interferon-alpha receptor

You might also like