You are on page 1of 16

Immunol Res (2012) 53:25–40

DOI 10.1007/s12026-012-8293-7

SINGAPORE IMMUNOLOGY NETWORK

Activation and regulation of interferon-b


in immune responses
Wei-Xiang Sin • Peng Li • Joe Poh-Sheng Yeong •

Keh-Chuang Chin

Keh-Chuang Chin

Published online: 13 March 2012


Ó Springer Science+Business Media, LLC 2012

Abstract Interferons (IFNs) were discovered more than half a century ago, and extensive research has since identified
multifarious roles for type I IFN in human immune responses. Here, we review the functions of IFN-b in innate and
adaptive immunity. We also discuss the activation and influence of IFN-b on myeloid cell types, including monocytes and
dendritic cells, as well as address the effects of IFN-b on T cells and B cells. Findings from our own laboratory, which
explores the molecular mechanisms of IFN-b activation by LPS and viruses, as well as from other groups investigating the
regulation of IFN-b by viral proteins and endogenous factors are described. The effects of post-translational modifications
of the interferon regulatory factor (IRF)-3 on IFN-b induction are also highlighted. Many unanswered questions remain
concerning the regulation of the type I IFN response in inflammation, especially the role of transcription factors in the
modulation of inflammatory gene expression, and these questions will form the basis for exciting avenues of future
research.

Keywords Interferon-b  Interferon regulatory factors  Monocytes  Transcriptional regulation 


Post-translational modification

Introduction purified and characterized in the late 1970s and early 1980s
[3–5].
More than 50 years ago, Isaac and Lindenmann identified IFN-b has been implicated in many human diseases
an inhibitory glycoprotein that ‘‘interfered’’ with influenza related to inflammation, including autoimmune diseases
virus replication and dissemination [1]. This novel glyco- and various cancers. In 1993, IFN-b1a and IFN-b1b were
protein was named interferon (IFN), and since then, there approved by the United States Food and Drug Adminis-
has been significant progress in our understanding of IFN tration for the treatment of relapsing–remitting multiple
biology. To date, 10 major mammalian IFNs have been sclerosis (MS) [6], although the mechanisms of action and
described: IFN-a, IFN-b, IFN-c, IFN-d, IFN-e, IFN-f, IFN- efficacy of this treatment are still hotly debated and
j, IFN-m, IFN-s, and IFN-x [2]. Among the seven IFNs intensely controversial [7, 8]. Recently, a 21-year follow-
that have been identified in humans, this review focuses on up study provided important new evidence that IFN-b1b
the regulation and functions of IFN-b, which was first reduced all-cause mortality in MS patients [9, 10].
Recent clinical and laboratory findings have shed new
light on the roles of IFN-b in human health and disease.
IFN-b exerts a wide range of biological activities in the
W.-X. Sin  P. Li  J. P.-S. Yeong  K.-C. Chin (&) human immune system, including the initiation of anti-
Laboratory of Gene Regulation and Inflammation, Singapore viral protein synthesis, promotion of cytotoxic activity, and
Immunology Network (SIgN), Agency for Science, Technology driving the differentiation and maturation of certain leu-
and Research (A*STAR), 8A Biomedical Grove, #04 Immunos,
Biopolis, Singapore 138648, Singapore
kocytes [11–16]. The Laboratory of Gene Regulation and
e-mail: kehchuang_chin@immunol.a-star.edu.sg Inflammation based within the Singapore Immunology

123
26 Singapore Immunology Network: SIgN (2012) 53:25–40

Network (SIgN)—a research institute under the auspices of detect intracellular pathogen-associated molecular patterns
Singapore’s Agency for Science, Technology and Research (PAMPs) via internalization into endosomal compartments
(A*STAR)—aims to improve our current knowledge of the [31] followed by degradation in phago-lysosomes. Recent
regulation and functions of IFN-b in inflammatory cells, infection studies using viruses and L. monocytogenes have
including human monocytes and dendritic cells (DCs), unveiled several novel cytosolic PRRs in macrophages,
which sit at the junction of innate and adaptive immunity. namely NOD2 [30, 32], DAI [33], Aim2 [34, 35], and
The first part of this article reviews the roles of IFN-b in LRRFIP1 [36], which can activate IFN-b with comparable
the development and function of innate and adaptive efficiency to more established TLRs, retinoic acid-induc-
immune cells, focusing on key developments in the field ible gene-I (RIG-I), and melanoma differentiation-asso-
since 2007—the 50th anniversary of the ‘‘birth’’ of IFNs. ciated gene-5 (Mda-5) (Table 1).
The second part describes our group’s research and related Nucleotide-binding oligomerization domain (NOD) pro-
investigations into the regulation of IFN-b production, teins comprise a family of NOD-like receptors (NLRs)—
from transcription through to cellular signaling. intracellular PRRs that recognize peptidoglycan (PGN)
degradation products such as muramyl dipeptides (MDP)
and muramyl tripeptides (MTP). Herskovits and colleagues
IFN-b in innate immune cells showed that NOD2 is required for optimal induction of IFN-
b in macrophages after phagocytosis and degradation of
Monocytes and macrophages L. monocytogenes [30]. Recently, two other groups discov-
ered that Mycobacterium tuberculosis (Mtb) and respiratory
Although typically elicited by viruses, type I IFN can also syncytial virus (RSV) stimulate the cytosolic NOD2 path-
be induced by bacterial infections [17–19]. Regardless of way to initiate type I IFN expression, either by a TANK-
the type of pathogen encountered, macrophages are early binding kinase 1 (TBK1)-IRF5-dependent mechanism in the
producers of IFN-b that primes other macrophages and case of bacterial PGN [32] or via a mitochondrial anti-viral
nearby immune cells to secrete a repertoire of pro-inflam- signaling protein (MAVS)-IRF3-mediated pathway for sin-
matory cytokines [20–22]. IFN-b plays a pivotal role in gle-stranded RNA (ssRNA) [37].
macrophage function by synergizing with signaling path- DNA-dependent activator of IRFs (DAI, also known as
ways downstream of pattern recognition receptors (PRRs), DLM-1 or ZBP1) is a cytosolic DNA sensor and activator
including Toll-like receptors (TLRs), to induce optimal of the innate immune response [33]. Intracellular DAI
production of pro-inflammatory cytokines. IFN-b synergy facilitates B-form DNA-mediated induction of IFN-b in a
with PRR signaling can be either beneficial or deleterious TBK1-IRF3-dependent manner [33]. While Wang et al.
to the host depending on the setting [23]. Previous reports [38] reported that silencing of DAI mRNA expression
have identified starkly contrasting effects of IFN-b synergy inhibits IFN-b activation, a separate group claimed that
with PRR under different conditions, either increasing DAI deficiency does not affect IFN-b production [39]. This
resistance to infection and improving survival [24], or discrepancy may be due to the different cell types used in
alternatively, decreasing resistance to infection [25] and these experiments.
impairing survival [26]. For example, in infection with Another new DNA sensor recently discovered in mac-
Listeria monocytogenes [25, 27, 28] or Tropheryma rophages and DCs is ‘‘absent in melanoma 2’’ (Aim2),
whipplei [29], type I IFN production may be detrimental to which was detected by screening for proteins that interact
the host. Host defense critically depends on the anti- with double-stranded DNA (dsDNA) and that can be
microbial activities of macrophages [20, 30], which can induced by IFN-b [40]. Aim2 localizes to the cell

Table 1 Novel intracellular PRRs that induce IFN-b in monocytes and macrophages
PRR Ligand Pathogen studied Signaling adaptor/ Reference
mediator

NOD2 MDP, MTP L. monocytogenes TBK1, IRF5 [30, 32, 37]


50 PPP RNA M. tuberculosis MAVS, IRF3
DAI/DLM-1/ZBP1 Cytosolic DNA: poly(dA:dT) ND TBK1, IRF3 [33, 38]
AIM2-like receptors Cytosolic DNA: poly(dA:dT) HSV-1, VACV DNA ASC, Caspase-1, [34, 42]
(Aim2, IFI16) STING, TBK1
LRRFIP1 dsRNA, dsDNA VSV, L. monocytogenes b-catenin, IRF3 [36]
ND not done, HSV herpes simplex virus, VACV vaccinia virus, VSV vesicular stomatitis virus, ASC apoptosis-associated speck-like protein
containing a CARD, MAVS mitochondrial antiviral signaling protein, STING stimulator of interferon genes, TBK TANK-binding kinase

123
Singapore Immunology Network: SIgN (2012) 53:25–40 27

cytoplasm and binds dsDNA via its HIN200 domain well as express high levels of MHC class I and II that
(hematopoietic IFN-inducible proteins with a 200 amino permit the activation of T-cell responses [48]. As described
acid repeat) [41], thus sensing DNA and inducing inflam- below, IFN-b production by DCs helps to establish a crit-
masome-mediated production of pro-inflammatory cyto- ical link between innate and adaptive immunity, which is
kines, especially IL-1b [34]. This study also revealed that required for the effective clearance of pathogens.
knocking down Aim2 resulted in a significant increase in
poly(dA:dT)-induced IFN-b, which may indicate that Dendritic cells (DCs)
Aim2 plays a redundant role or acts as a negative regulator
of the IFN-b response. Further investigations will be nec- DCs are archetypal sentinel cells that detect pathogens and
essary to address this question. IFI16 is another pyrin and elicit immune responses [23]. DCs have long been known to
HIN domain (PYHIN)-containing protein that functions as produce type I IFN in response to viruses, purified bacterial
an innate sensor of intracellular DNA, and it has been DNA, and synthetic prokaryotic oligodeoxynucleotides
proposed that PYHIN proteins may represent a new family (ODN) [49], but until comparatively recently, only macro-
of innate DNA sensors, named AIM2-like receptors phages were thought to be capable of responding to whole
(ALRs) [42]. bacteria [50]. However, a novel role for DCs in innate
In addition, Yang et al. [36] identified a novel PRR that immunity has recently emerged with the report that con-
can detect both dsDNA and dsRNA to trigger type I IFN ventional DCs (cDCs), but not plasmacytoid DCs (pDCs) or
production in macrophages challenged with vesicular sto- macrophages, were able to produce large amounts of IFN-b
matitis virus (VSV) and L. monocytogenes. Leucine-rich following bacterial degradation in phago-lysosomes and
repeat flightless-interacting protein 1 (LRRFIP1) promoted activation of TLR7, MyD88, and IRF1 [50].
the activation of interaction partner b-catenin, which The intriguing realization that APCs are the main
enhanced IFN-b expression by binding to IRF3 and sources of IFN-b during infection [21, 51, 52] underscores
recruiting histone acetyltransferase p300 to the IFNB the pivotal role of IFN-b in adaptive immunity [53]. Using
enhanceosome [36]. a single-cell visualization method, Scheu et al. [51] char-
Interestingly, NOD2 [43], Aim2 [34, 40], and RIG-I [44] acterized the relative contributions of each cell type to in
are each induced by IFN-b itself, suggesting that they are vivo IFN-b production in response to different stimuli.
engaged in a positive feedback loop with this cytokine. IFN-b induces DC up-regulation of MHC class I molecules
Upon pathogen stimulation, these PRRs activate IFN-b [54], enhances DC maturation and activation [55, 56], and
secretion into the extracellular milieu where it binds to type ultimately promotes Th1 responses [57, 58]. Type I IFN is
I IFN receptors (IFNARs) on the secreting cell, as well as a major driver of DC maturation into immuno-stimulatory
on neighboring cells, thus driving an autocrine/paracrine cells and is required for the optimal generation of Th1
loop that up-regulates a series of genes including NOD2 responses [14]. The high levels of type I IFNs secreted by
[43], Aim2 [34, 40], and RIG-I [44]. This sequence of pDCs can activate CD8? T cells and support cross-pre-
events presumably heightens immune surveillance in the sentation, while also enhancing CD4? Th1 development
local micro-environment and increases cellular resistance and restricting differentiation toward Th2 and Th17 [59]
to infection. (Fig. 1).
A number of cytosolic PRRs have been implicated The immuno-modulatory effects of IFN-b on DC-med-
in inflammasome complex formation and inflammatory iated T-cell responses provide possible targets for the
caspase-1 activation. Intriguingly, type I IFN signaling development of novel therapies. DC vaccination strategies
downstream of the IFNAR was reported to inhibit NLRP1 involve activating DCs with immunogenic antigens in vitro
and NLRP3 inflammasome activation, which reduced to boost T-cell responses, followed by re-infusion of the
mature IL-1b production in mouse and human monocytes/ activated cells to generate protective immunity to cancers
macrophages pre-treated with IFN-b and then challenged or viral and bacterial infections. However, optimizing the
with lipopolysaccharide (LPS) and alum adjuvant [45]. delivery and presentation of DCs remains a difficult chal-
This study also demonstrated that type I IFN signaling lenge. Surprisingly, several recent studies have announced
induced IL-10 and reduced pro-IL-1b synthesis [45], unprecedented efficacy of DC-based vaccines in the pres-
indicating that IFN-b treatment can suppress both pro-IL- ence of type I IFN, which may accentuate DC activation
1b expression and processing to bioactive IL-1b [45]. [14]. TLR7 activation enhances the ability of migratory
Macrophage-derived cytokines including IFN-b play a DCs to acquire antigens provided that type I IFN is
key role in the differentiation of immune effector cells, available [60]. In a separate investigation, the TLR agonists
thus helping to bridge the innate and adaptive immune poly(I:C) and LPS together with type I and II IFNs gen-
responses [46, 47]. Antigen-presenting cells (APCs) like erated mature DCs with high migratory capacity and IL-12
macrophages and DCs produce both type I and II IFNs, as production [61]. As a result, type I IFN has now been

123
28 Singapore Immunology Network: SIgN (2012) 53:25–40

Fig. 1 Roles of IFN-b in


Novel
different immune cells. IFN-b
intracellular
enhances DC maturation and
PRRs that
activation. Macrophage- and
induce IFN-β
DC-derived IFN-b promotes
Th1 responses and activates
CD8? T cells, as well as
enhances CD4? Th1
development while restricting
differentiation toward Th2 and
Th17. It also induces antibody IFNβ MΦ IFNβ
production and isotype class
switching by enhancing B-cell
development and differentiation
into plasma cells
Immune effector cells
IFNβ

DC T cell B cell

Maturation; ↑ Th1 development; ↑ Plasma cell


Activation; ↓ Th2 and Th17 differentiation;
DC-based differentiation; Activates plasma cells;
vaccine adjuvant Promotes Th1 Antibody production;
responses; Isotype class switching;
Activates CD8+ T cells; Auto-antibodies in
Autoimmune diseases autoimmune diseases

recognized as a vaccine adjuvant, and it may even be capacity of this population [66, 67]. In addition, IL-10/
possible to predict vaccine efficacy by assessing the level IL-27-producing CD4? T cells are augmented after IFN-b
of type I IFN induced [62]. therapy, which may suppress effector T-cell responses and
constrain the expansion of Th17 cells [67, 68]. However,
a conflicting report instead indicated that IL-10 production
IFN-b in adaptive immune cells is unaltered and that IL-17 levels are decreased upon IFN-
b treatment in MS, leading the authors to conclude that
T cells IFN-b is pro-inflammatory in Th17-induced experimental
autoimmune encephalomyelitis. These data indicate that
The ability of IFN-b to shape the adaptive immune care must be exercised when administering IFN-b therapy
response by activating and priming T cells is well recog- in the clinic before determining the disease subtype (Th1
nized. Numerous studies have confirmed that the timing of vs. Th17) [69].
IFN-b exposure determines the positive or negative effect The therapeutic role of IFN-a in graft-versus-host dis-
on Th1 polarization [63], which is influenced by distinct ease (GVHD) and graft-versus-leukemia (GVL) has been
signal transducers and activators of transcription (STAT) known for decades [70, 71], especially in the context of
factors that are recruited upon type I IFN activation in DCs relapsing chronic myeloid leukemia following bone mar-
[64]. Type I IFN can, therefore, influence T-cell activation row transplantation [72]. However, the mechanisms of
and differentiation directly, but can also modulate T-cell action remain obscure, and the application of type I IFN in
response through effects on DCs (Fig. 1). this setting remains controversial [73, 74]. A study by
Although the mechanisms of action of IFN-b therapy Robb et al. [75] suggested that the efficacy of type I IFN in
for relapsing–remitting MS remain unclear at this time, protecting patients from GVHD and GVL can be attributed
effects on T-cell homeostasis seem likely to contribute to to the suppression of donor CD4? T-cell proliferation and
the efficacy of this intervention. IFN-b ameliorates Th1 differentiation. In contrast, CD8? T cell–dependent GVHD
cell pathologies in MS patients by abrogating the pro- and GVL responses were enhanced by the pleiotropic
inflammatory properties of IFN-c and IL-12 [65]. IFN-b effects of type I IFN. Further investigations will be nec-
also restores the function of regulatory T cells in MS essary to address this apparent dichotomy and to clarify the
patients by increasing the frequency and inhibitory complex role of type I IFN in this context.

123
Singapore Immunology Network: SIgN (2012) 53:25–40 29

B cells in the cytoplasm. The kinase TBK1 phosphorylates tran-


scription factor IRF3 [87], which then dimerizes and
Type I IFN is able to induce long-lived antibody produc- translocates into the nucleus to induce IFN-b expression
tion and isotype class switching by inducing activation and [88]. IFN-b is secreted into the extracellular milieu, where
differentiation of plasma cells and memory B cells [53, 76]. it interacts with IFNARs on the host cell membrane to
Type I IFN enhances proliferation and prevents apoptosis activate downstream JAK-STAT signaling [89]. The
of primary B cells, even in the absence of mitogenic stimuli phosphorylation and activation of the kinases JAK1 and
[77], and generally enhances B cell development and Tyk2 lead to the phosphorylation and activation of the
function [78] (Fig. 1). transcription factors STAT1 and STAT2, which results in
A classical study published in 2002 demonstrated that the formation of transcriptional complexes that bind to
IFN-a/b increased survival and decreased apoptosis of B specific promoter regions of downstream cytokine genes
cells, as well as amplified B-cell activation and antibody (known as IFN-stimulated response elements; ISREs).
responses upon B-cell receptor stimulation [79]. IFN-a/b STAT1/STAT2 binding to ISREs in the presence of path-
secretion by virus-activated pDCs was then shown to ogenic stimuli such as LPS and Sendai virus (SeV) cul-
induce the differentiation of CD40-activated B cells into minates in the induction of pro-inflammatory cytokines
antibody-secreting plasma cells [80]. In line with these including type I IFNs [90]. This signaling cascade, thus,
findings, increasing evidence now links type I IFN with B constitutes a positive feedback loop that amplifies the
cell production of auto-antibodies and increased risk of production of type I IFNs.
diseases such as systemic lupus erythematosus (SLE) TLR4 on the host cell membrane is a well-known
[81–83]. sensor of Gram-negative bacterial infections. However,
there are emerging reports that the Gram-negative bacteria
Francisella tularensis and Salmonella typhimurium acti-
Activation of IFN-b vate cytosolic PRRs instead of TLR4 [91–94]. It was
reported that F. tularensis can activate the caspase-1 in-
IFN-b can directly and indirectly influence the biological flammasome via the intracellular sensor Aim2 and can
functions of a plethora of innate and adaptive immune induce type I IFN and other pro-inflammatory cytokines
cells, including monocytes, macrophages, DCs, T cells and via an as yet unidentified sensor. Aim2 is now acknowl-
B cells. Type I IFN production can also be stimulated by edged to function as a sensor of undigested viral DNA,
pathogen ligands from diverse microorganisms [84], synthetic DNA, and DNA derived from killed bacteria
among which bacterial LPS and viruses are two of the most (presumably released from the phago-lysosome), ulti-
well studied. Type I IFN has been shown to be critical for mately leading to inflammasome activation [34]. In vitro
the control of bacterial, viral, and even parasitic protozoan and in vivo studies have now shown that this inflamma-
infections [23]. The classical IFN-b signaling pathway some activation is Aim2- and IRF3-dependent [95].
consists of membrane-associated or cytosolic receptors, Although inflammasome activation depended on an intact
adaptor proteins in the cytosol, and transcription factors in type I IFN response, F. tularensis-induced type I IFN
the nucleus. production required IRF3, but was independent of a
number of TLRs, RLRs (RIG-I like receptors), and NLRs
Activation of IFN-b by LPS (including Aim2) [95]. More research will be needed to
unravel this complex relationship.
PAMPs activate various membrane PRRs on the host cell,
among which TLRs are the most extensively studied. TLRs Activation of IFN-b by viruses
can sense a variety of bacterial and viral ligands, which
include LPS (TLR4), PGN (TLR2), Pam3CSK4 (TLR1/2), During viral infection, secreted IFN-b binds to IFNARs on
dsRNA and poly(I:C) (TLR3), ssRNA (TLR7/8), and CpG the surface of cells to trigger the expression of a large
ODN (TLR9) [85]. LPS is known to interact with TLR4 in number of anti-viral genes [96]. dsRNA is a PAMP derived
conjunction with CD14 and MD2 receptors on the host cell from viruses that can be detected by TLR3 to induce
membrane [86], which activates downstream MyD88- activation of TRIF, which is also a key mediator of the LPS
dependent and MyD88-independent (TRIF-dependent) response pathway. TRIF has been shown to bind to TLR3
signaling pathways. The MyD88-independent pathway by co-immunoprecipitation [97], and the same methodol-
leads to type I IFN production, although there is also sig- ogy was employed to prove that the N-terminal domain of
nificant cross-talk with the MyD88-dependent pathway TRIF interacts with TBK1 and TRAF6, while the C-ter-
through NF-jB [86]. TRIF is a critical mediator in this minal domain of TRIF binds to the kinase RIP1 [98].
pathway, and TRAM and TBK1 are key adaptor molecules Furthermore, it was shown that TBK1 physically interacts

123
30 Singapore Immunology Network: SIgN (2012) 53:25–40

with IRF3 and that macrophages from TBK1 knockout of how differential IFN-b induction is achieved at the
mice exhibit defective IRF3 activation and IFN-b tran- molecular level. Other groups have previously reported the
scription [87]. In this virus infection model, TBK1 and initiation of IFN-b transcription at 6 h post-virus infection
IKKi phosphorylated IRF3, which either homodimerizes or in HeLa cells [104], while our group has observed early
heterodimerizes with IRF7, translocates into the nucleus, expression of IFN-b in human blood monocytes within
and binds to the IFN-b promoter region to up-regulate IFN- only 1–2 h of exposure to SeV [105]. This disparity raised
b expression [87]. IRF7 is another member of the IRF the possibility that distinct myeloid-specific transcription
family of transcription factors that can be activated upon factor(s) may be involved in the rapid induction of IFN-b
virus infection [99]. It was previously reported that the in monocytes compared with non-myeloid cell types [105].
response to myxoma virus infection in mouse pDCs is We determined that this monocyte-restricted phenomenon
dependent on IRF7 but not on IRF3 [100]. Upon virus can be attributed to the constitutive binding of IRF8 and
infection, latent IRF7 in the cytoplasm is phosphorylated PU.1 to the ETS/IRF composite element (EICE) motif of
by TBK1/IKKi and forms a homodimer, followed by the IFN-b promoter, thus creating a preformed activation
translocation into the nucleus to establish the IFNB en- complex on the IFN-b promoter, which facilitates rapid
hanceosome together with IRF3 and other transcription recruitment of IRF3 via direct interactions with IRF8
factors (Fig. 2). (Fig. 2) [105]. In contrast to other ubiquitously expressed
Extensive studies on the chromatin structure and pro- IRF family members, IRF8 is expressed only in myeloid
moter architecture of the IFNB gene have revealed that cells. IRF8 (also known as interferon consensus sequence
IFN-b transcriptional activation requires the enhancer binding protein; ICSBP) is a transcription factor protein
region located immediately upstream of the core promoter originally isolated based on recognition of the promoter
[101]. Virus infection results in the coordinate activation of region of the H-2LD MHC class I gene [106]. siRNA-
transcription factors that assemble on this IFN-b enhancer mediated knockdown of IRF8 in monocytes dramatically
region. NF-jB, ATF-2/c-Jun, PU.1, and IRFs have been blocked SeV- and LPS-induced IFN-b transcription, while
identified as possessing cognate binding sites on the IFN-b ectopic expression of IRF8 in mouse 32D (IRF8-/-) cells
promoter region, which regulate IFN-b mRNA transcrip- successfully rescued SeV-induced IFN-b transcription
tion: the binding of these regulators is concentrated on [105]. Through these and other experiments, we have been
*(-100)bp of the IFN-b genomic locus [102, 103] able to demonstrate that IRF8 directly synergizes with
(Fig. 2). IRF3 in monocytes to facilitate faster IFN-b transcription
Type I IFN production differs in magnitude and kinetics after pathogenic stimulation.
between different cell types. One of the research interests Different patterns of type I IFN production can be
of our laboratory has been to build a better understanding induced by different PRRs in response to viral infections.

a Classical model of the IFN -β enhanceosome in non-myeloid cells


C-Jun
ATF-2

IRF3 IRF7 IRF3 IRF7 p50 p65


-100 -50
5’..AATGACATAGGAAAACTGAAAGGGAGAAGTGAAAGTGGGAAATTCCTCTGA..3’
3’..TTACTGTATCCTTTTGACTTTCCCTCTTCACTTTCACCCTTTAAGGAGACT..5’
PRDIV PRDIII PRDI PRDII

b Hypothetical model of the IFN -β enhanceosome in myeloid cells

? ? IRF3 IRF8 PU.1


-100 -50
5’..AATGACATAGGAAAACTGAAAGGGAGAAGTGAAAGTGGGAAATTCCTCTGA..3’
3’..TTACTGTATCCTTTTGACTTTCCCTCTTCACTTTCACCCTTTAAGGAGACT..5’
EIRE EICE

Fig. 2 Models of the IFN-b enhanceosome in non-myeloid and Our laboratory determined that rapid induction of IFN-b in monocytes
myeloid cells. a Assembly of transcription factors on the IFN-b can be attributed to the constitutive binding of IRF8 and PU.1 to the
enhanceosome as determined from crystal structures. ATF-2/c-Jun, ETS/IRF composite element (EICE) motif of the IFN-b promoter.
IRF3/IRF7, and NF-jB bind cooperatively to the IFN-b enhancer: Putative IRF3 and IRF8 binding sites are underlined. The potential
ATF-2/c-Jun in complex with two IRF molecules on positive involvement of other transcription factors at the ETS/IRF response
regulatory domain (PRD) IV-PRDIII, and NF-jB in complex with element (EIRE) motif within the PRDIII region as well as the PRDIV
two IRF molecules on the PRDI and PRDII regions of the enhancer. b region in monocytes remains to be confirmed

123
Singapore Immunology Network: SIgN (2012) 53:25–40 31

For example, West Nile virus activates TLR3 [107], production upon MDP stimulation is impaired in TBK1-
whereas VSV interacts with TLR7, and herpes simplex and IRF5-deficient macrophages, but not in IRF3-deficient
virus (HSV)-1 and HSV-2 use TLR9 to activate type I IFN macrophages [32]. There are also reports that IRF1 is
signaling [108–110]. Besides membrane-associated TLRs, involved in Mtb infection, although only demonstrated in
members of the cytosolic RLR family can also be stimu- THP-1 cell lines [118]. However, for other strains of bac-
lated by viruses. For instance, RIG-I is employed by teria like L. monocytogenes, TBK1 has been shown to
Newcastle disease virus (NDV) [111], while Mda-5 is activate type I IFN through IRF3 [119] (Table 2).
activated by Dengue virus [112]. This may be due to the The protozoan parasite Plasmodium falciparum is the
different infection strategies used: while some viruses bind etiological agent of malaria [120]. P. falciparum elicits
to cell membranes, other viruses gain entry into the cytosol significant amounts of type I IFN from pDCs [121]. Plas-
through fusion of the virus envelope with the plasma modial DNA has been shown to act as a PAMP that
membrane [113]. stimulates type I IFN production, since DNase treatment
ablates any stimulatory effect for this molecule. TLR9 is
Activation of IFN-b by non-viral pathogens believed to be the receptor activated by parasite DNA [122,
123], which contains a specific AT-rich DNA motif
Mtb is the causative agent of human tuberculosis [114], and responsible for that stimulation [124]. Stimulator of IFN
like many other pathogens, Mtb can induce production of genes (STING), an ER-associated protein, forms a complex
type I IFN. Previous studies have found that Mtb-induced with TBK1 to activate IRF3 and IRF7, leading to type I
type I IFN is IRF3-independent, since IFN-b induction was IFN production. This STING-TBK1-IRF3/IRF7 pathway
not defective in IRF3-deficient bone marrow-derived contributes to DNA sensing in plasmodial infection [124]
macrophages (BMDMs) stimulated with mycobacterial (Table 2).
ligands [32]. The signal transduction pathway leading to
type I IFN induction is therefore likely to be different in
Mtb compared with other Gram-negative bacteria. Indeed, Regulation of IFN-b
while LPS interacts with TLR4 on the cell membrane, Mtb
instead binds to intracellular NOD2 [115]. NOD2 is located The production of type I IFN by host cells in response to
in the cytoplasm and recognizes bacterial cell wall ligands pathogen exposure is critical in innate and adaptive
such as MDP [116]. The mycobacterial ESX1 system immunity (reviewed in [19]). However, dysregulated
causes phago-lysosomal escape and entry into the cytosol, expression of type I IFN can be detrimental to the host, and
which is required for effective receptor-interacting protein- systemic overproduction of type I IFN can lead to septic
2 (Rip2) activation downstream of NOD2 [117]. Conse- shock syndrome in Gram-negative sepsis [23]. Further-
quently, TBK1 becomes activated and phosphorylates more, type I IFN and members of the IRF family have also
IRF5, which in turn translocates into the nucleus to induce been implicated in the induction of autoimmune responses
type I IFN. Experimental data have shown that type I IFN and in the pathogenesis of diseases such as SLE and

Table 2 Pathogen-induced signaling pathways leading to IFN-b production


PAMP Type Transcription Knockout Phenotype Signaling Ref.
factor studied of knockout pathway
involved involved

dsRNA/Sendai Virus IRF3 IRF3-/- Inhibited downstream TLR3 [180]


virus dendritic cells genes
Encephalomyocarditis Virus IRF3, IRF7 IRF3-/-, Increased lethality TLR7 [181]
virus IRF7-/-mice
Mycobacterium Gram-negative IRF5 IRF5-/-BMDMs Impaired interferon NOD2 [32]
tuberculosis bacteria production
Plasmodium Malaria IRF3, IRF7 IRF3-/-, Resistant to malaria STING [124]
falciparum IRF7-/-BMDMs disease
Group B Gram-positive IRF1 IRF1-/- mice Susceptible to TLR7 [50]
Streptococcus bacteria infection
LPS Gram-negative IRF3 IRF3-/- mice Decreased downstream TLR4 [182]
bacteria cytokines
Listeria Gram-positive IRF3 IRF3-/- mice Increased lethality TLR-independent; [25]
monocytogenes bacteria TBK1-dependent

123
32 Singapore Immunology Network: SIgN (2012) 53:25–40

Sjogren’s syndrome [84, 125, 126]. Levels of type I IFN, and nuclear translocation by the EBV oncoprotein latent
thus, need to be carefully regulated during the course of membrane protein-1 (LMP1) [141]; polyubiquitination of
infection. One of the research interests of our laboratory IRF7 and subsequent proteasomal degradation by TRIM21
has been to better understand how inflammatory processes [142]; SUMO modification of IRF7 transcriptional activity
are regulated at the molecular level. These studies on mediated by PIAS1 [143, 144]; and lysine acetylation
mechanisms that limit the expression of type I IFN may effects on IRF7 DNA binding as mediated by p300/CREB-
lead to the design of better therapeutic interventions in binding protein-associated factor (PCAF) and GCN5 [145].
inflammatory and autoimmune diseases [127]. Recently, TRIM28 was found to act as a IRF7-specific
SUMO E3 ligase (analogous to PIAS1) that attenuates
Regulation of IFN-b induction by viral proteins transcriptional activity and abrogates IFNA1, IFNB, and
ISRE promoter activation and gene expression [146]. MafB
Viruses have evolved a plethora of countermeasures to is another molecule that was found to inhibit IRF7-medi-
subvert the innate anti-viral host response. For example, ated transcription by disrupting the binding of IRF7 to
Kaposi’s sarcoma-associated herpesvirus (KSHV) can target gene promoters [147]. Interested readers are directed
potently attenuate the production of type I IFN (reviewed to the comprehensive review of Ning et al. [99]. We shall
in [128, 129]). Several KSHV viral proteins have been discuss in greater detail the modulation of IRF3 activity in
identified that suppress type I IFN induction at various the following sections.
levels. For instance, at the post-translational level, ORF45
blocks IRF7 phosphorylation and nuclear translocation Regulation of IRF3-mediated IFN-b induction
[130, 131], and the ubiquitin E3 ligase replication tran- by endogenous factors
scription activator (RTA) targets IRF7 for proteasomal
degradation [132]. At the transcriptional level, the viral Many groups have studied IRF3-mediated IFN-b activation
IRF homologues vIRF1 and vIRF3 interfere with the DNA following TLR3 ligation. IRF3 can be regulated by myriad
binding of IRF3 and IRF7, respectively [133, 134], and K- post-translational modifications, including phosphor-
bZIP, ORF36, and latency-associated nuclear antigen ylation, ubiquitination, SUMOylation, ISGylation, and
(LANA-1) interfere with IFN-b promoter activation [135– S-glutathionylation (Fig. 3). The growing list of proteins
137] (Fig. 3). that modulate IRF3 activation, though by no means
exhaustive, currently includes A20, Atg5-Atg12, CYLD,
Regulation of IRF7-mediated IFN-b induction DAK, DUBA, gC1qR, ISG15, ISG56, LGP2, NLRX1,
optineurin, OTUB1/2, Pin1, RBCK1, RNF5, RNF125,
IRF7 is subject to multiple layers of regulation at the SARM, SHIP-1, SIKE, SINTBAD, TAG, TRIAD3A, and
transcriptional, translational, and post-translational levels TRIM21 [148–152].
(Fig. 3). At the transcriptional level, the Epstein–Barr virus
(EBV) BRLF1 protein was shown to repress the tran- Phosphorylation of IRF3
scription of IRF7 and IRF3, leading to decreased IRF7 and
IRF3 mRNA and protein levels and reduced IFN-b The cytoplasmic ubiquitin-editing protein A20 (also known
production [138]. ATF4 is a component of the cellular as TNFAIP3) is induced by TLR ligands and can down-
integrated stress response that can also inhibit IRF7 tran- regulate IRF3-dependent gene expression to reduce
scription to reduce type I IFN induction [139]. Interest- inflammation (reviewed in [153]). A20 is induced upon
ingly, IRF7 can in turn regulate ATF4 levels and activity, SeV infection [154] and functions as a negative regulator
suggesting a cross-talk between the IFN response and of TLR3-induced IRF3 activation and IFN-b transcription.
cellular integrated stress response [139]. A20 was initially shown to interact with TRIF and TBK1-
IRF7 translation was recently found to be inhibited by IKKi to inhibit activation of ISRE and the IFNB promoter
the translational repressors 4E-BP1 and 4E-BP2 [140]. [154, 155]. Recently, the adaptor protein Tax1-binding
Accordingly, 4E-BP1 and 4E-BP2 knockout mice show protein 1 (TAX1BP1) was found to be required for A20
enhanced type I IFN production in pDCs and increased inhibition of TBK1-IKKi by limiting Lys[63]-linked
resistance to VSV [140]. Moreover, 4E-BP1- and 4E-BP2- polyubiquitination of TBK1-IKKi [149]. Finally, A20-
deficient mouse embryonic fibroblasts exhibited enhanced binding inhibitor of NF-jB 1 (ABIN1) has been identified
IRF7 translation and type I IFN responses, resulting in as a novel player that cooperates with TAX1BP1 and
increased resistance to encephalomyocarditis virus and TBK1-IKKi in recruiting A20 proteins to form a complex
VSV [140]. with TBK1-IKKi [156]. While additional novel mediators
Numerous post-translational modifications can regulate may yet be identified, the prevailing hypothesis is that the
IRF7 activity: modification of IRF7 phosphorylation status trimolecular complex comprising ABIN1-TAX1BP1-A20

123
Singapore Immunology Network: SIgN (2012) 53:25–40 33

Cell membrane
dsRNA dsRNA
Endosome

TLR3
RIG-I /
MDA-5

TRIF MAVS

A20, TAX1BP1, ABIN1


(TBK1-IKKi K63-linked EBV
polyubiquitination) TBK1 IKKi
LMP1 (IRF7 K63-linked
polyubiquitination)
IRF3 post-translational modifications

Pin1, TRIM21/Ro52, RAUL, IRF3 EBV


Caspase-8, RBCK1, Cullin-1 BRLF1 (IRF7 transcription)
(IRF3 polyubiquitination) IRF7
KSHV
SUMO1, SUMO2, SUMO3 RTA (IRF7 polyubiquitination)
(IRF3 SUMOylation);
SENP2 (IRF3 de-SUMOylation) ATF4 (IRF7 transcription);
4E-BP1, 4E-BP2 (IRF7 translation);
Herc5 (IRF3 ISGylation) TRIM21, RAUL (IRF7 polyubiquitination);
SUMO1, SUMO2, SUMO3,
GRX-1 (IRF3 de-glutathionylation) PIAS1, TRIM28 (IRF7 SUMOylation)

A20, TAX1BP1, ABIN1 P P P P P P EBV


(IRF3 phosphorylation) LMP1
IRF3
IRF3

IRF3

IRF7

IRF7
IRF7

(IRF7 phosphorylation)
KSHV
ORF45
(IRF7 phosphorylation)
IRF3-CL (IRF3 nuclear translocation)
Nuclear membrane

KSHV
vIRF1 (IRF3 DNA binding); KSHV
IFN-β
K-bZIP, ORF36, LANA-1 vIRF3
(promoter activation) (IRF7 DNA binding)

E2F1, PPAR-γ, MafB PCAF, GCN5, MafB


(promoter activation) (IRF7 DNA binding)

Fig. 3 Positive and negative regulators of IFN-b induction. Viral proteasomal degradation, while their phosphorylation status affects
proteins and endogenous factors regulate IFN-b induction through their nuclear translocation and DNA binding. IFN-b induction is also
different post-translational modifications of IRF3 (left; shaded boxes) regulated at the level of promoter activation by various transcription
and IRF7 (right). Polyubiquitination of the IRFs leads to their factors

is sufficient to disrupt TBK1-IKKi Lys[63]-linked poly- phosphorylation, homodimerization, and DNA binding via
ubiquitination, decrease IRF3 phosphorylation, and depress TRIF protein degradation and can block MAVS/VISA/IPS-
virus or poly(I:C)-induced IFN-b promoter activation. 1/Cardif-mediated ISRE promoter activation in reporter
Lin et al. probed the signaling pathway upstream of IRF3 gene assays [157]. The authors, therefore, contend that A20
and concluded that the virus-inducible, NF-jB-dependent inhibition acts upstream of TBK1-IKKi and impacts more
E3 ligase activity of A20 completely blocked RIG-I-medi- greatly on RIG-I signaling than on TLR3-TRIF signaling
ated IFNB, ISRE, and IFNA4 promoter activation, but only [157].
partially inhibited TLR3-TRIF- and TBK1-IKKi-mediated It was recently discovered that A20 is also a negative
ISRE promoter activation. A20 blocks RIG-I-induced IRF3 regulator of IRF7. The EBV oncoprotein LMP1 induces

123
34 Singapore Immunology Network: SIgN (2012) 53:25–40

Lys[63]-linked polyubiquitination of IRF7 [158], but can expression of pro-inflammatory cytokine genes such as IL-
simultaneously induce A20, which decreases the poly- 12p40 following IFN-c/CpG stimulation of macrophages
ubiquitination of IRF7 to reduce transcriptional activation [161]. TRIM21 mediates Lys[63]-ubiquitination of IRF8 to
of the IFN-a promoter [158]. In this way, A20 seems to enhance the expression of IL-12p40 in myeloid cells [161].
function as a negative regulator that balances the positive In contrast, Cbl-mediated ubiquitination of IRF8 can lead
signaling of LMP1-IRF7 activation. to the destabilization and degradation of IRF8, in a manner
analogous to Pin1-mediated ubiquitination of IRF3 [162].
Ubiquitination of IRF3 RAUL is another HECT-type ubiquitin E3 ligase that has
been implicated in the proteasomal degradation of IRF3.
Ye et al. [127] showed that the degradation of IRF3 is RAUL inhibits type I IFN production by catalyzing the
responsible for switching off IFN-b production after acute polyubuquitination and proteasomal degradation of IRF3
viral infection, with no significant contribution from tran- and IRF7 [163]. RAUL is positively regulated by a deubiq-
scriptional repressors. The peptidyl-prolyl isomerase Pin1 uitinating enzyme ubiquitin-specific processing protease 7
interacts with phosphorylated IRF3 to promote polyubiq- (Usp7) [163]. Interestingly, the KSHV protein RTA medi-
uitination and proteasomal degradation of IRF3, thereby ates RAUL deubiquitination via Usp7, thereby enhancing
suppressing TLR3- and RIG-I-mediated IFN-b production RAUL degradation of IRF3 and IRF7 [163]. RIG-I-induced
following poly(I:C) stimulation or NDV infection [159]. caspase-8 cleaves IRF3 into an intermediary that is amena-
At this point, it is interesting to highlight two conflicting ble to polyubiquitination and proteasomal degradation
reports that describe the role of the E3 ubiquitin ligase [164]. Other ubiquitin ligases implicated in the proteasomal
TRIM21 (also known as Ro52) in IRF3-mediated IFN-b degradation of IRF3 include RBCK1 and Cullin-1.
induction. Higgs et al. [160] demonstrated a negative reg-
ulatory role for TRIM21, which promoted polyubiquitina- SUMOylation of IRF3
tion and proteasomal degradation of IRF3 to inhibit
poly(I:C)-induced activation of the IFN-b promoter Ubiquitin is structurally and functionally related to another
downstream of TLR3 stimulation. However, Yang et al. post-translational modification tag, small ubiquitin-related
[148] instead demonstrated a positive regulatory role for modifier (SUMO). SUMOylation has been shown to serve
TRIM21, which interfered with Pin1-IRF3 interactions to dual functions by antagonizing polyubiquitination and
prevent IRF3 polyubiquitination and proteasomal degra- proteasomal degradation on the one hand, but also syner-
dation, thereby enhancing IFN-b production in response to gizing with E3 ubiquitin ligase recruitment to enhance this
SeV infection. The authors attributed this discrepancy to pathway of degradation in other contexts (reviewed in [165,
differing experimental conditions yielding different out- 166]). Kubota et al. [143] found that TLR and RIG-I/MDA5
comes [148]. In Higgs’ study, TRIM21 inhibited IFN-b activation upon VSV infection leads to TRIF- and MAVS/
promoter activation after 48-h SeV infection of 293T cells VISA/IPS-1/Cardif-mediated SUMO1, SUMO2, and
[160], while in Yang’s study, TRIM21 increased IFN-b SUMO3 modification of mouse IRF3 and IRF7 and that this
promoter activation after 9-h SeV infection of HEK293 SUMOylation negatively regulates type I IFN gene
cells [148]. Small hairpin (sh)RNA knockdown of TRIM21 expression. On the other hand, Ran et al. [167] showed that
led to an increase in IFN-b transcript levels after 48-h the deSUMOylating enzyme Sentrin/SUMO-specific pro-
poly(I:C) stimulation of 293T cells in Higgs’ article [160], tease 2 (SENP2) caused the deSUMOylation, polyubiqui-
while siRNA knockdown of TRIM21 resulted in reduced tination, and proteasomal degradation of IRF3 to impair
IFN-b transcript levels after 9-h SeV infection of HEK293 virus-induced IFN-b transcription. These variable findings
cells in Yang’s report [148]. Although cell type–specific could perhaps be attributed to species-specific differences,
responses to different stimuli may account for some of but they may also indicate cross-talk between the SU-
these observations, it is interesting to speculate that MOylation and ubiquitination pathways acting on IRF3.
unknown time critical factors may also contribute to the
disparate actions of TRIM21 after infection. ISGylation of IRF3
Another recent report from Higgs et al. [142] revealed
that TRIM21 negatively regulates the stability and activity In 2006, our laboratory and a separate group made the
of IRF7 protein in a similar fashion to IRF3. TRIM21 also almost simultaneous discovery that interferon-induced
promotes the polyubiquitination and subsequent proteaso- HECT-type E3 protein ligase Herc5 is involved in the
mal degradation of IRF7, resulting in decreased IRF7 conjugation of ubiquitin-like protein ISG15 to protein tar-
protein expression and decreased transactivation of the gets in human cells [168, 169]. One such protein target has
IFNA4 promoter downstream of TLR7/9 stimulation [142]. now been identified as IRF3 [170]. Lu et al. [170] illus-
TRIM21 has been also found to enhance IRF8-dependent trated that ISGylation of IRF3 in response to IFN treatment

123
Singapore Immunology Network: SIgN (2012) 53:25–40 35

and NDV infection prevents proteasomal degradation and IFN-b mRNA transcription and protein secretion in primary
augments nuclear translocation of phosphorylated IRF3 to peritoneal macrophages. PPAR-c acts as a transcriptional
support IFN-b promoter activation. Shi et al. [171] further repressor by inhibiting TLR4- and TLR3-induced IRF3
demonstrated that Herc5-mediated IRF3 ISGylation dis- transcriptional activation—preventing IRF3 binding to the
rupts the Pin1-IRF3 interaction to inhibit IRF3 poly- IFN-b promoter and impairing IFN-b signaling [156].
ubiquitination and degradation by proteasomes. Herc5 is, Transcription factor MafB is involved in the establish-
therefore, a positive regulator of IRF3-mediated IFN-b ment and maintenance of the myeloid lineage and has been
promoter activation and IFN-b mRNA expression. identified as a negative regulator of IFN-b induction at the
IFNB gene promoter [147]. MafB blocks the type I IFN
S-glutathionylation of IRF3 amplification loop by interacting with IRF3 and interfering
with the recruitment of co-activators to reduce IRF3 tran-
S-glutathionylation is another crucial post-translational scriptional activity [147].
modification of IRF3 that can regulate IFN-b activation. Finally, a fascinating function of the IFN-b enhancer as
IRF3 is S-glutathionylated in non-infected cells, and a ‘‘signal amplifier’’ has been described. It was demon-
deglutathionylation of IRF3 catalyzed by glutaredoxin-1 strated that virus infection leads to IFN-b enhancer asso-
(GRX-1) following viral infection is required for the ciation with NF-jB at a different chromosomal genomic
recruitment of transcription co-activator CBP-p300 [172]. region to trigger mono-allelic IFN-b gene expression [104].
However, phosphorylation, dimerization, and nuclear Thereafter, an autocrine/paracrine loop of IFN-b signaling
translocation of IRF3 do not appear to depend on GRX-1- through the IFNAR induced IRF7 expression, promoted
mediated deglutathionylation of IRF3 [172]. IFNB enhanceosome assembly on the remaining alleles,
and resulted in multi-allelic IFN-b expression [104]. In a
Other regulators of IRF3: E2F1 and IRF3-CL separate study, LPS stimulation of TLR4 drove activated
NF-jB RelA recruitment from the cytosol to the IFN-b
Xu et al. [173] characterized the IRF3 promoter and found locus at a cluster of novel NF-jB sites downstream of the
that it contains E2F transcription factor binding sites human IFN-b gene, remote from the IFNB enhanceosome
(TFBS). Mutation of the E2F TFBS increased IRF3 pro- region, which by itself was insufficient for maximal IFN-b
moter activation, while over-expression of E2F1 decreased induction upon LPS stimulation [179].
IRF3 promoter activation, thereby implicating E2F1 in the
negative regulation of IRF3 gene expression [174]. In
human monocytic cells stimulated with LPS, our laboratory Concluding remarks
previously found that TLR4-induced NF-jB recruits E2F1,
both of which then cooperatively bind to RelA and E2F1 More than half a century of research has spawned a huge
binding sites in NF-jB target genes, such as TNF and IL1B body of data describing the multifarious roles of type I IFN
[175]. It is therefore interesting to speculate that E2F1 may in human health and disease. As much as we now know
positively regulate NF-jB target gene expression, while about the beneficial and detrimental effects of IFN-b,
also negatively regulating IRF3-mediated gene expression. an in-depth understanding of the underlying molecular
Very recently, an alternative splicing isoform of IRF3 mechanisms of IFN-b responses is still lacking. Scientific
was cloned that shares a common N-terminus with IRF3, efforts in this area are complicated by the complex inter-
but exhibits a unique C-terminus of 125 amino acids [176]. play between multiple signaling pathways and numerous
The mRNA transcript and protein of this isoform, termed regulatory factors that modulate the actions of IFN-b. From
IRF3-CL, was detected in many cell lines [176]. IRF3-CL a survey of the current literature, it is clear that many
was also found to inhibit SeV-induced IRF3 nuclear determinants influence the magnitude and kinetics of the
translocation and IFN-b induction [176]. Together with IFN-b response in different physiological and pathological
other alternative splicing isoforms such as IRF-3a [177] settings. Such context-specific factors include: the cell type
and IRF3-nirs3 [178], IRF3-CL may represent a general in which IFN-b is induced; the species of pathogen
mechanism of reducing IRF3 activity [176]. invading the host cell; the duration of infection; and the
local microenvironment where the infection occurs. It is
Novel transcriptional regulators at the IFN-b promoter: fascinating that the strength and longevity of IFN-b syn-
PPAR-c and MafB thesis are so finely attuned to every nuance of pathogenic
or inflammatory stimuli received by the host cell. Out-
Zhao et al. [156] recently reported that the peroxisome standing questions include: the timing and cell types in
proliferator-activated receptor (PPAR)-c agonist troglit- which IFN-b is induced; how signaling pathways are
azone negatively regulates LPS- and poly(I:C)-induced activated and repressed to fine-tune IFN-b expression;

123
36 Singapore Immunology Network: SIgN (2012) 53:25–40

which molecules are involved in the spatio-temporal reg- 15. Nguyen KB, et al. Coordinated and distinct roles for IFN-alpha
ulation of IFN-b; and why indeed are such tight controls beta, IL-12, and IL-15 regulation of NK cell responses to viral
infection. J Immunol. 2002;169:4279–87.
required for the regulation of IFN-b production. Many 16. Su L, David M. Inhibition of B cell receptor-mediated apoptosis
unanswered questions remain regarding the role of tran- by IFN. J Immunol. 1999;162:6317–21.
scription factors in regulating the type I IFN response in 17. Sing A, et al. Bacterial induction of beta interferon in mice is a
inflammation, and these quandaries will provide the plat- function of the lipopolysaccharide component. Infect Immun.
2000;68:1600–7.
form for exciting avenues of future research. 18. Parker D, Prince A. Type I interferon response to extracellular
bacteria in the airway epithelium. Trends Immunol. 2011;
Acknowledgments The authors thank Dr Neil McCarthy for criti- 32:582–8.
cally reviewing the manuscript. W.-X.S. and J.P.-S.Y. are supported 19. Perry AK, Chen G, Zheng D, Tang H, Cheng G. The host type I
by the Agency for Science, Technology and Research (A*STAR) interferon response to viral and bacterial infections. Cell Res.
graduate scholarships. Work in the Laboratory of Gene Regulation 2005;15:407–22.
and Inflammation is supported by the Singapore Immunology 20. Mancuso G, et al. Type I IFN signaling is crucial for host
Network (SIgN), Agency for Science, Technology and Research resistance against different species of pathogenic bacteria. J
(A*STAR) (Grant 06-001; K.-C.C.). Immunol. 2007;178:3126–33.
21. Utaisincharoen P, Anuntagool N, Limposuwan K, Chaisuriya P,
Sirisinha S. Involvement of beta interferon in enhancing
inducible nitric oxide synthase production and antimicrobial
References activity of Burkholderia pseudomallei-infected macrophages.
Infect Immun. 2003;71:3053–7.
1. Isaacs A, Lindenmann J. Virus interference. 1. The interferon. P R 22. Gautier G, et al. A type I interferon autocrine-paracrine loop is
Soc Lond B Biol. 1957;147:258–267. involved in Toll-like receptor-induced interleukin-12p70 secre-
2. Pestka S. The interferons: 50 years after their discovery, there is tion by dendritic cells. J Exp Med. 2005;201:1435–46.
much more to learn. J Biol Chem. 2007;282:20047–51. 23. Decker T, Muller M, Stockinger S. The yin and yang of type I
3. Friesen HJ, et al. Purification and molecular characterization of interferon activity in bacterial infection. Nat Rev Immunol.
human fibroblast interferon. Arch Biochem Biophys. 1981;206: 2005;5:675–87.
432–50. 24. Mahieu T, Libert C. Should we inhibit type I interferons in
4. Rubinstein M, Rubinstein S, Familletti PC, Miller RS, Waldman sepsis? Infect Immun. 2007;75:22–9.
AA, Pestka S. Human leukocyte interferon: production, purifi- 25. O’Connell RM, et al. Type I interferon production enhances
cation to homogeneity, and initial characterization. Proc Natl susceptibility to Listeria monocytogenes infection. J Exp Med.
Acad Sci U S A. 1979;76:640–4. 2004;200:437–45.
5. Stein S, Kenny C, Friesen HJ, Shively J, Del Valle U, Pestka S. 26. Thomas KE, Galligan CL, Newman RD, Fish EN, Vogel SN.
NH2-terminal amino acid sequence of human fibroblast inter- Contribution of interferon-beta to the murine macrophage
feron. Proc Natl Acad Sci U S A. 1980;77:5716–9. response to the toll-like receptor 4 agonist, lipopolysaccharide. J
6. Borden EC, et al. Interferons at age 50: past, current and future Biol Chem. 2006;281:31119–30.
impact on biomedicine. Nat Rev Drug Discov. 2007;6:975–90. 27. Zwaferink H, Stockinger S, Hazemi P, Lemmens-Gruber R,
7. de Andres C, et al. Interferon beta-1a therapy enhances CD4? Decker T. IFN-b increases listeriolysin O-induced membrane
regulatory T-cell function: an ex vivo and in vitro longitudinal permeabilization and death of macrophages. J Immunol. 2008;
study in relapsing-remitting multiple sclerosis. J Neuroimmunol. 180:4116–23.
2007;182:204–11. 28. Carrero JA, Calderon B, Unanue ER. Type I interferon sensi-
8. Ebers GC, et al. Long-term follow-up of the original interferon- tizes lymphocytes to apoptosis and reduces resistance to listeria
beta1b trial in multiple sclerosis: design and lessons from a 16- infection. J Exp Med. 2004;200:535–40.
year observational study. Clin Ther. 2009;31:1724–36. 29. Al Moussawi K, Ghigo E, Kalinke U, Alexopoulou L, Mege J-L,
9. Goodin DS, et al. Predictive value of baseline and short-term Desnues B. Type I interferon induction is detrimental during
outcomes on mortality in multiple sclerosis: data from the infection with the Whipple’s disease bacterium, Tropheryma
interferon beta-1b 21-year long-term follow-up study. 21st whipplei. Plos Pathog. 2010;6:e1000722.
Meeting of the European Neurological Society (ENS) 30. Herskovits AA, Auerbuch V, Portnoy DA. Bacterial ligands
2011;Abstract P585. generated in a phagosome are targets of the cytosolic innate
10. Ebers GC, et al. Cause of death in patients with multiple scle- immune system. Plos Pathog. 2007;3:e51.
rosis: the 21-year long-term follow-up study. 21st Meeting of 31. Gruenberg J, van der Goot FG. Mechanisms of pathogen entry
the European Neurological Society (ENS) 2011;Abstracts P429. through the endosomal compartments. Nat Rev Mol Cell Biol.
11. Chiang EY, et al. Targeted depletion of lymphotoxin-alpha- 2006;7:495–504.
expressing TH1 and TH17 cells inhibits autoimmune disease. 32. Pandey AK, et al. NOD2, RIP2 and IRF5 play a critical role in
Nat Med. 2009;15:766–73. the type I interferon response to Mycobacterium tuberculosis.
12. Ito T, Amakawa R, Inaba M, Ikehara S, Inaba K, Fukuhara S. PLoS Pathog. 2009;5:e1000500.
Differential regulation of human blood dendritic cell subsets by 33. Takaoka A, et al. DAI (DLM-1/ZBP1) is a cytosolic DNA
IFNs. J Immunol. 2001;166:2961–9. sensor and an activator of innate immune response. Nature.
13. Kito T, Kuroda E, Yokota A, Yamashita U. Enhancement of 2007;448:501–5.
macrophage cytotoxicity against murine gliomas by interferon 34. Hornung V, et al. AIM2 recognizes cytosolic dsDNA and forms
beta: increase in nitric oxide production in response to glioma- a caspase-1-activating inflammasome with ASC. Nature. 2009;
derived soluble factors. J Neurosurg. 2002;97:619–26. 458:514–8.
14. Longhi MP, et al. Dendritic cells require a systemic type I 35. Roberts TL, et al. HIN-200 proteins regulate caspase activation
interferon response to mature and induce CD4? Th1 immunity in response to foreign cytoplasmic DNA. Science. 2009;323:
with poly IC as adjuvant. J Exp Med. 2009;206:1589–602. 1057–60.

123
Singapore Immunology Network: SIgN (2012) 53:25–40 37

36. Yang P, et al. The cytosolic nucleic acid sensor LRRFIP1 59. Huber JP, Farrar JD. Regulation of effector and memory T-cell
mediates the production of type I interferon via a beta-catenin- functions by type I interferon. Immunology. 2011;132:466–74.
dependent pathway. Nat Immunol. 2010;11:487–94. 60. Kastenmuller K, et al. Protective T cell immunity in mice fol-
37. Sabbah A, et al. Activation of innate immune antiviral responses lowing protein-TLR7/8 agonist-conjugate immunization
by Nod2. Nat Immunol. 2009;10:1073–80. requires aggregation, type I IFN, and multiple DC subsets. J Clin
38. Wang ZC, et al. Regulation of innate immune responses by DAI Invest. 2011;121:1782–96.
(DLM-1/ZBP1) and other DNA-sensing molecules. Proc Natl 61. Nguyen-Pham TN, et al. Type I and II interferons enhance
Acad Sci U S A. 2008;105:5477–82. dendritic cell maturation and migration capacity by regulating
39. Lippmann J, et al. IFNb responses induced by intracellular CD38 and CD74 that have synergistic effects with TLR ago-
bacteria or cytosolic DNA in different human cells do not nists. Cell Mol Immunol. 2011;8:341–7.
require ZBP1 (DLM-1/DAI). Cell Microbiol. 2008;10:2579–88. 62. Rajagopal D, Paturel C, Morel Y, Uematsu S, Akira S, Diebold
40. Burckstummer T, et al. An orthogonal proteomic-genomic SS. Plasmacytoid dendritic cell-derived type I interferon is
screen identifies AIM2 as a cytoplasmic DNA sensor for the crucial for the adjuvant activity of Toll-like receptor 7 agonists.
inflammasome. Nat Immunol. 2009;10:266–72. Blood. 2010;115:1949–57.
41. Ludlow LEA, Johnstone RW, Clarke CJP. The HIN-200 family: 63. Nagai T, Devergne O, Mueller TF, Perkins DL, van Seventer
more than interferon-inducible genes? Exp Cell Res. 2005;308: JM, van Seventer GA. Timing of IFN-beta exposure during
1–17. human dendritic cell maturation and naive Th cell stimulation
42. Unterholzner L, et al. IFI16 is an innate immune sensor for has contrasting effects on Th1 subset generation: a role for IFN-
intracellular DNA. Nat Immunol. 2010;11:997–1004. beta-mediated regulation of IL-12 family cytokines and IL-18 in
43. Kim YG, et al. Viral infection augments Nod1/2 signaling to naive Th cell differentiation. J Immunol. 2003;171:5233–43.
potentiate lethality associated with secondary bacterial infec- 64. Longman RS, Braun D, Pellegrini S, Rice CM, Darnell RB,
tions. Cell Host Microbe. 2011;9:496–507. Albert ML. Dendritic-cell maturation alters intracellular sig-
44. Hu JZ, et al. Role of cell-to-cell variability in activating a naling networks, enabling differential effects of IFN-alpha/beta
positive feedback antiviral response in human dendritic cells. on antigen cross-presentation. Blood. 2007;109:1113–22.
Plos One 2011;6:e16614. 65. Nagai T, Devergne O, van Seventer GA, van Seventer JM.
45. Guarda G, et al. Type I interferon inhibits interleukin-1 production Interferon-beta mediates opposing effects on interferon-gamma-
and inflammasome activation. Immunity. 2011;34:213–23. dependent Interleukin-12 p70 secretion by human monocyte-
46. O’Riordan M, Yi CH, Gonzales R, Lee KD, Portnoy DA. Innate derived dendritic cells. Scand J Immunol. 2007;65:107–17.
recognition of bacteria by a macrophage cytosolic surveillance 66. Korporal M, et al. Interferon beta-induced restoration of regu-
pathway. Proc Natl Acad Sci U S A. 2002;99:13861–6. latory T-cell function in multiple sclerosis is prompted by an
47. Mocci S, Dalrymple SA, Nishinakamura R, Murray R. The increase in newly generated naive regulatory T cells. Arch
cytokine stew and innate resistance to L-monocytogenes. Neurol-Chicago 2008;65:1434–1439.
Immunol Rev. 1997;158:107–14. 67. Martin-Saavedra FM, Gonzalez-Garcia C, Bravo B, Ballester S.
48. Sen GC. Viruses and interferons. Annu Rev Microbiol. 2001; Beta interferon restricts the inflammatory potential of CD4?
55:255–81. cells through the boost of the Th2 phenotype, the inhibition of
49. Tailor P, Tamura T, Ozato K. IRF family proteins and type I Th17 response and the prevalence of naturally occurring T
interferon induction in dendritic cells. Cell Res. 2006;16:134– regulatory cells. Mol Immunol. 2008;45:4008–19.
40. 68. Zhang X, Markovic-Plese S. Interferon beta inhibits the Th17
50. Mancuso G, et al. Bacterial recognition by TLR7 in the lyso- cell-mediated autoimmune response in patients with relapsing-
somes of conventional dendritic cells. Nat Immunol. 2009;10: remitting multiple sclerosis. Clin Neurol Neurosurg. 2010;112:
587–94. 641–5.
51. Scheu S, Dresing P, Locksley RM. Visualization of IFNbeta 69. Axtell RC, et al. T helper type 1 and 17 cells determine efficacy
production by plasmacytoid versus conventional dendritic cells of interferon-beta in multiple sclerosis and experimental
under specific stimulation conditions in vivo. Proc Natl Acad Sci encephalomyelitis. Nat Med. 2010;16:406–12.
U S A. 2008;105:20416–21. 70. Steegmann JL, et al. Interferon alpha for chronic myeloid leu-
52. Cervantes-Barragan L, et al. Control of coronavirus infection kemia relapsing after allogeneic bone marrow transplantation.
through plasmacytoid dendritic-cell-derived type I interferon. Bone Marrow Transplant. 1999;23:483–8.
Blood. 2007;109:1131–7. 71. Streetly M, Kazmi M, Radia D, Hoyle C, Schey SA. Second
53. Le Bon A, Schiavoni G, D’Agostino G, Gresser I, Belardelli F, autologous transplant with cyclosporin/interferon alpha-induced
Tough DF. Type I interferons potently enhance humoral graft versus host disease for patients who have failed first-line
immunity and can promote isotype switching by stimulating consolidation. Bone Marrow Transplant. 2004;33:1131–5.
dendritic cells in vivo. Immunity. 2001;14:461–70. 72. Porter DL, Roth MS, Mcgarigle C, Ferrara JLM, Antin JH.
54. Lindahl P, Gresser I, Leary P, Tovey M. Interferon treatment of Induction of graft-versus-host disease as immunotherapy for
mice: enhanced expression of histocompatibility antigens on relapsed chronic myeloid-leukemia. N Engl J Med. 1994;
lymphoid cells. Proc Natl Acad Sci U S A. 1976;73:1284–7. 330:100–6.
55. Santini SM, et al. Type I interferon as a powerful adjuvant for 73. Thornley TB, et al. Type 1 IFN mediates cross-talk between
monocyte-derived dendritic cell development and activity in vitro innate and adaptive immunity that abrogates transplantation
and in Hu-PBL-SCID mice. J Exp Med. 2000;191:1777–88. tolerance. J Immunol. 2007;179:6620–9.
56. Luft T, et al. Type I IFNs enhance the terminal differentiation of 74. Serrano J, Prieto E, Mazarbeitia F, Roman A, Llamas P, Tomas
dendritic cells. J Immunol. 1998;161:1947–53. JF. Atypical chronic graft-versus-host disease following inter-
57. Wenner CA, Guler ML, Macatonia SE, O’Garra A, Murphy feron therapy for chronic myeloid leukaemia relapsing after
KM. Roles of IFN-gamma and IFN-alpha in IL-12-induced T allogeneic BMT. Bone Marrow Transplant. 2001;27:85–7.
helper cell-1 development. J Immunol. 1996;156:1442–7. 75. Robb RJ, et al. Type I-IFNs control GVHD and GVL responses
58. Ito T, et al. Plasmacytoid dendritic cells regulate Th cell after transplantation. Blood. 2011;118:3399–409.
responses through OX40 ligand and type I IFNs. J Immunol. 76. Slifka MK, Antia R, Whitmire JK, Ahmed R. Humoral immu-
2004;172:4253–9. nity due to long-lived plasma cells. Immunity. 1998;8:363–72.

123
38 Singapore Immunology Network: SIgN (2012) 53:25–40

77. Ruuth K, Carlsson L, Hallberg B, Lundgren E. Interferon-alpha NF-kappaB activation and apoptosis pathways. J Biol Chem.
promotes survival of human primary B-lymphocytes via phos- 2004;279:15652–61.
phatidylinositol 3-kinase. Biochem Biophys Res Commun. 99. Ning S, Pagano JS, Barber GN. IRF7: activation, regulation,
2001;284:583–6. modification and function. Genes Immun. 2011;12:399–414.
78. Oka H. Regulation of human B-cell responsiveness by inter- 100. Dai P, et al. Myxoma virus induces type I interferon production in
feron-alpha—interferon-alpha-mediated suppression of B-cell murine plasmacytoid dendritic cells via a TLR9/MyD88-, IRF5/
function is reversed through direct interactions between mono- IRF7-, and IFNAR-dependent pathway. J Virol. 2011;85:10814–25.
cytes and B-cells. Cell Immunol. 1993;146:238–48. 101. Smale ST, Fisher AG. Chromatin structure and gene regulation
79. Braun D, Caramalho I, Demengeot J. IFN-a/b enhances BCR- in the immune system. Annu Rev Immunol. 2002;20:427–62.
dependent B cell responses. Int Immunol. 2002;14:411–9. 102. Panne D, Maniatis T, Harrison SC. An atomic model of the
80. Jego G, Palucka AK, Blanck J-P, Chalouni C, Pascual V, Ban- interferon-beta enhanceosome. Cell. 2007;129:1111–23.
chereau J. Plasmacytoid dendritic cells induce plasma cell dif- 103. Ford E, Thanos D. The transcriptional code of human IFN-b
ferentiation through Type I interferon and interleukin 6. gene expression. Biochimica et Biophysica Acta (BBA)—Gene
Immunity. 2003;19:225–34. Regul Mech. 2010;1799:328–336.
81. Niewold TB, Clark DN, Salloum R, Poole BD. Interferon alpha 104. Apostolou E, Thanos D. Virus infection induces NF-kappaB-
in systemic lupus erythematosus. J Biomed Biotechnol. 2010; dependent interchromosomal associations mediating monoall-
2010:948364. elic IFN-beta gene expression. Cell. 2008;134:85–96.
82. Thibault DL, Graham KL, Lee LY, Balboni I, Hertzog PJ, Utz 105. Li P, et al. IRF8 and IRF3 cooperatively regulate rapid inter-
PJ. Type I interferon receptor controls B-cell expression of feron-beta induction in human blood monocytes. Blood.
nucleic acid-sensing Toll-like receptors and autoantibody pro- 2011;117:2847–54.
duction in a murine model of lupus. Arthritis Res Ther. 2009; 106. Driggers PH, et al. An interferon gamma-regulated protein that
11:R112. binds the interferon-inducible enhancer element of major his-
83. de Goer de Herve MG, et al. Interferon-alpha triggers B cell tocompatibility complex class I genes. Proc Natl Acad Sci U S
effector 1 (Be1) commitment. Plos One 2011;6:e19366. A. 1990;87:3743–7.
84. Theofilopoulos AN, Baccala R, Beutler B, Kono DH. Type I 107. Wang T, Town T, Alexopoulou L, Anderson JF, Fikrig E, Flavell
interferons (alpha/beta) in immunity and autoimmunity. Annu RA. Toll-like receptor 3 mediates West Nile virus entry into the
Rev Immunol. 2005;23:307–36. brain causing lethal encephalitis. Nat Med. 2004;10:1366–73.
85. Kang JY, Lee JO. Structural biology of the Toll-like receptor 108. Hemmi H, et al. The roles of two IkappaB kinase-related kinases
family. Annu Rev Biochem. 2011;80:917–41. in lipopolysaccharide and double stranded RNA signaling and
86. Akira S, Takeda K. Toll-like receptor signalling. Nat Rev viral infection. J Exp Med. 2004;199:1641–50.
Immunol. 2004;4:499–511. 109. Krug A, Luker GD, Barchet W, Leib DA, Akira S, Colonna M.
87. Fitzgerald KA, et al. IKKepsilon and TBK1 are essential com- Herpes simplex virus type 1 activates murine natural interferon-
ponents of the IRF3 signaling pathway. Nat Immunol. 2003;4: producing cells through toll-like receptor 9. Blood. 2004;103:
491–6. 1433–7.
88. Sharma S, tenOever BR, Grandvaux N, Zhou GP, Lin R, Hiscott 110. Lund J, Sato A, Akira S, Medzhitov R, Iwasaki A. Toll-like
J. Triggering the interferon antiviral response through an IKK- receptor 9-mediated recognition of Herpes simplex virus-2 by
related pathway. Science. 2003;300:1148–51. plasmacytoid dendritic cells. J Exp Med. 2003;198:513–20.
89. Uze G, Schreiber G, Piehler J, Pellegrini S. The receptor of the 111. Yoneyama M, et al. The RNA helicase RIG-I has an essential
type I interferon family. Curr Top Microbiol Immunol. 2007; function in double-stranded RNA-induced innate antiviral
316:71–95. responses. Nat Immunol. 2004;5:730–7.
90. Schindler C, Levy DE, Decker T. JAK-STAT signaling: from 112. Nasirudeen AM, Wong HH, Thien P, Xu S, Lam KP, Liu DX.
interferons to cytokines. J Biol Chem. 2007;282:20059–63. RIG-I, MDA5 and TLR3 synergistically play an important role
91. Mariathasan S, Weiss DS, Dixit VM, Monack DM. Innate in restriction of dengue virus infection. PLoS Negl Trop Dis.
immunity against Francisella tularensis is dependent on the 2011;5:e926.
ASC/caspase-1 axis. J Exp Med. 2005;202:1043–9. 113. Hogle JM. Poliovirus cell entry: common structural themes in
92. Henry T, Brotcke A, Weiss DS, Thompson LJ, Monack DM. viral cell entry pathways. Annu Rev Microbiol. 2002;56:677–
Type I interferon signaling is required for activation of the in- 702.
flammasome during Francisella infection. J Exp Med. 2007; 114. Rohde K, Yates RM, Purdy GE, Russell DG. Mycobacterium
204:987–94. tuberculosis and the environment within the phagosome.
93. Mariathasan S, et al. Cryopyrin activates the inflammasome in Immunol Rev. 2007;219:37–54.
response to toxins and ATP. Nature. 2006;440:228–32. 115. Sirard JC, Vignal C, Dessein R, Chamaillard M. Nod-like
94. Lara-Tejero M, et al. Role of the caspase-1 inflammasome in receptors: cytosolic watchdogs for immunity against pathogens.
Salmonella typhimurium pathogenesis. J Exp Med. 2006;203: PLoS Pathog. 2007;3:e152.
1407–12. 116. Girardin SE, et al. Peptidoglycan molecular requirements
95. Fernandes-Alnemri T, et al. The AIM2 inflammasome is critical allowing detection by Nod1 and Nod2. J Biol Chem. 2003;
for innate immunity to Francisella tularensis. Nat Immunol. 278:41702–8.
2010;11:385–93. 117. van der Wel N, et al. M. tuberculosis and M. leprae translocate
96. Stetson DB, Medzhitov R. Type I interferons in host defense. from the phagolysosome to the cytosol in myeloid cells. Cell.
Immunity. 2006;25:373–81. 2007;129:1287–98.
97. Jiang Z, Mak TW, Sen G, Li X. Toll-like receptor 3-mediated 118. Qiao Y, et al. Host defense responses to infection by Myco-
activation of NF-kappaB and IRF3 diverges at Toll-IL-1 bacterium tuberculosis. Induction of IRF-1 and a serine protease
receptor domain-containing adapter inducing IFN-beta. Proc inhibitor. J Biol Chem. 2002;277:22377–85.
Natl Acad Sci U S A. 2004;101:3533–8. 119. Stetson DB, Medzhitov R. Recognition of cytosolic DNA acti-
98. Han KJ, Su X, Xu LG, Bin LH, Zhang J, Shu HB. Mechanisms vates an IRF3-dependent innate immune response. Immunity.
of the TRIF-induced interferon-stimulated response element and 2006;24:93–103.

123
Singapore Immunology Network: SIgN (2012) 53:25–40 39

120. Clark IA, Alleva LM, Mills AC, Cowden WB. Pathogenesis of the cellular integrated stress responses. J Immunol. 2011;186:
malaria and clinically similar conditions. Clin Microbiol Rev. 1001–10.
2004;17:509–39. 140. Colina R, et al. Translational control of the innate immune
121. Voisine C, Mastelic B, Sponaas AM, Langhorne J. Classical response through IRF-7. Nature. 2008;452:323–8.
CD11c? dendritic cells, not plasmacytoid dendritic cells, induce 141. Zhang L, Pagano JS. Interferon regulatory factor 7 mediates
T cell responses to Plasmodium chabaudi malaria. Int J Parasi- activation of Tap-2 by Epstein-Barr virus latent membrane
tol. 2010;40:711–9. protein 1. J Virol. 2001;75:341–50.
122. Khor CC, et al. A Mal functional variant is associated with 142. Higgs R, et al. Self protection from anti-viral responses—Ro52
protection against invasive pneumococcal disease, bacteremia, promotes degradation of the transcription factor IRF7 down-
malaria and tuberculosis. Nat Genet. 2007;39:523–8. stream of the viral toll-like receptors. PLoS ONE. 2010;5:e11776.
123. Parroche P, et al. Malaria hemozoin is immunologically inert but 143. Kubota T, et al. Virus infection triggers SUMOylation of IRF3
radically enhances innate responses by presenting malaria DNA and IRF7, leading to the negative regulation of type I interferon
to Toll-like receptor 9. Proc Natl Acad Sci U S A. 2007; gene expression. J Biol Chem. 2008;283:25660–70.
104:1919–24. 144. Chang T-H, et al. Ebola Zaire virus blocks type I interferon
124. Sharma S, et al. Innate immune recognition of an AT-rich stem- production by exploiting the host SUMO modification machin-
loop DNA motif in the Plasmodium falciparum genome. ery. PLoS Pathog. 2009;5:e1000493.
Immunity. 2011;35:194–207. 145. Caillaud A, et al. Acetylation of interferon regulatory factor-7
125. Sozzani S, Type I. Interferons in systemic autoimmunity. by p300/CREB-binding protein (CBP)-associated factor (PCAF)
Autoimmunity. 2010;43:196–203. impairs its DNA binding. J Biol Chem. 2002;277:49417–21.
126. Salloum R, Niewold TB. Interferon regulatory factors in human 146. Liang Q, et al. Tripartite motif-containing protein 28 is a small
lupus pathogenesis. Transl Res. 2011;157:326–31. ubiquitin-related modifier E3 ligase and negative regulator of
127. Ye J, Maniatis T. Negative regulation of interferon-b gene IFN regulatory factor 7. J Immunol. 2011;187:4754–63.
expression during acute and persistent virus infections. PLoS 147. Kim H, Seed B. The transcription factor MafB antagonizes
ONE. 2011;6:e20681. antiviral responses by blocking recruitment of coactivators to
128. Sathish N, Yuan Y. Evasion and subversion of interferon-med- the transcription factor IRF3. Nat Immunol. 2010;11:743–50.
iated antiviral immunity by Kaposi’s sarcoma-associated her- 148. Yang K, et al. TRIM21 is essential to sustain IFN regulatory
pesvirus: an overview. J Virol. 2011;85:10934–44. factor 3 activation during antiviral response. J Immunol. 2009;
129. Rezaee SAR, Cunningham C, Davison AJ, Blackbourn DJ. 182:3782–92.
Kaposi’s sarcoma-associated herpesvirus immune modulation: 149. Parvatiyar K, Barber GN, Harhaj EW. TAX1BP1 and A20
an overview. J Gen Virol. 2006;87:1781–804. inhibit antiviral signaling by targeting TBK1-IKKi kinases. J
130. Sathish N, Zhu FX, Golub EE, Liang Q, Yuan Y. Mechanisms Biol Chem. 2010;285:14999–5009.
of autoinhibition of IRF-7 and a probable model for inactivation 150. Yang Y-K, et al. ARF-like protein 16 (ARL16) inhibits RIG-I by
of IRF-7 by Kaposi’s sarcoma-associated herpesvirus protein binding with its C-terminal domain in a GTP-dependent manner.
ORF45. J Biol Chem. 2011;286:746–56. J Biol Chem. 2011;286:10568–80.
131. Zhu FX, King SM, Smith EJ, Levy DE, Yuan Y. A Kaposi’s sar- 151. Gabhann JN, et al. Absence of SHIP-1 results in constitutive
coma-associated herpesviral protein inhibits virus-mediated induc- phosphorylation of tank-binding kinase 1 and enhanced TLR3-
tion of type I interferon by blocking IRF-7 phosphorylation and dependent IFN-b production. J Immunol. 2010;184:2314–20.
nuclear accumulation. Proc Natl Acad Sci U S A. 2002;99:5573–8. 152. Siednienko J, Gajanayake T, Fitzgerald KA, Moynagh P, Mig-
132. Yu Y, Wang SE, Hayward GS. The KSHV immediate-early gin SM. Absence of MyD88 results in enhanced TLR3-depen-
transcription factor RTA encodes ubiquitin E3 ligase activity dent phosphorylation of IRF3 and increased IFN-b and
that targets IRF7 for proteosome-mediated degradation. Immu- RANTES production. J Immunol. 2011;186:2514–22.
nity. 2005;22:59–70. 153. Verstrepen L, Verhelst K, van Loo G, Carpentier I, Ley SC, Beyaert
133. Lin R, et al. HHV-8 encoded vIRF-1 represses the interferon R. Expression, biological activities and mechanisms of action of
antiviral response by blocking IRF-3 recruitment of the CBP/ A20 (TNFAIP3). Biochem Pharmacol. 2010;80:2009–20.
p300 coactivators. Oncogene. 2001;20:800–11. 154. Wang Y-Y, Li L, Han K-J, Zhai Z, Shu H-B. A20 is a potent
134. Joo CH, Shin YC, Gack M, Wu L, Levy D, Jung JU. Inhibition inhibitor of TLR3- and Sendai virus-induced activation of NF-
of interferon regulatory factor 7 (IRF7)-mediated interferon jB and ISRE and IFN-b promoter. FEBS Lett. 2004;576:86–90.
signal transduction by the Kaposi’s sarcoma-associated herpes- 155. Saitoh T, et al. A20 is a negative regulator of IFN regulatory
virus viral IRF homolog vIRF3. J Virol. 2007;81:8282–92. factor 3 signaling. J Immunol. 2005;174:1507–12.
135. Lefort S, Soucy-Faulkner A, Grandvaux N, Flamand L. Binding 156. Zhao W, et al. Peroxisome proliferator-activated receptor c
of Kaposi’s sarcoma-associated herpesvirus K-bZIP to inter- negatively regulates IFN-b production in toll-like receptor
feron-responsive factor 3 elements modulates antiviral gene (TLR) 3- and TLR4-stimulated macrophages by preventing
expression. J Virol. 2007;81:10950–60. interferon regulatory factor 3 binding to the IFN-b promoter. J
136. Hwang S, et al. Conserved herpesviral kinase promotes viral Biol Chem. 2011;286:5519–28.
persistence by inhibiting the IRF-3-mediated type I interferon 157. Lin R, et al. Negative regulation of the retinoic acid-inducible
response. Cell Host Microbe. 2009;5:166–78. gene I-induced antiviral state by the ubiquitin-editing protein
137. Cloutier N, Flamand L. Kaposi sarcoma-associated herpesvirus A20. J Biol Chem. 2006;281:2095–103.
latency-associated nuclear antigen inhibits interferon (IFN) b 158. Ning S, Pagano JS. The A20 deubiquitinase activity negatively
expression by competing with IFN regulatory factor-3 for regulates LMP1 activation of IRF7. J Virol. 2010;84:6130–8.
binding to IFNB promoter. J Biol Chem. 2010;285:7208–21. 159. Saitoh T, et al. Negative regulation of interferon-regulatory
138. Bentz GL, Liu R, Hahn AM, Shackelford J, Pagano JS. Epstein– factor 3-dependent innate antiviral response by the prolyl
Barr virus BRLF1 inhibits transcription of IRF3 and IRF7 and isomerase Pin1. Nat Immunol. 2006;7:598–605.
suppresses induction of interferon-b. Virology. 2010;402:121–8. 160. Higgs R, Gabhann JN, Larbi NB, Breen EP, Fitzgerald KA,
139. Liang Q, Deng H, Sun C-W, Townes TM, Zhu F. Negative Jefferies CA. The E3 ubiquitin ligase Ro52 negatively regulates
regulation of IRF7 activation by activating transcription factor 4 IFN-b production post-pathogen recognition by polyubiquitin-
suggests a cross-regulation between the IFN responses and mediated degradation of IRF3. J Immunol. 2008;181:1780–6.

123
40 Singapore Immunology Network: SIgN (2012) 53:25–40

161. Kong HJ, et al. Cutting edge: autoantigen Ro52 is an interferon 173. Xu H-G, Ren W, Lu C, Zhou G-P. Characterization of the
inducible E3 ligase that ubiquitinates IRF-8 and enhances cyto- human IRF-3 promoter and its regulation by the transcription
kine expression in macrophages. J Immunol. 2007;179:26–30. factor E2F1. Mol Biol Rep. 2010;37:3073–80.
162. Xiong H, et al. Ubiquitin-dependent degradation of interferon 174. Xu H-G, Ren W, Zou L, Wang Y, Jin R, Zhou G-P. Direct
regulatory factor-8 mediated by Cbl down-regulates interleukin- repression of the human IRF-3 promoter by E2F1. Immunoge-
12 expression. J Biol Chem. 2005;280:23531–9. netics. 2011;63:189–96.
163. Yu Y, Hayward GS. The ubiquitin E3 ligase RAUL negatively 175. Lim C-A, et al. Genome-wide mapping of RELA(p65) binding
regulates type I interferon through ubiquitination of the tran- identifies E2F1 as a transcriptional activator recruited by NF-jB
scription factors IRF7 and IRF3. Immunity. 2010;33:863–77. upon TLR4 activation. Mol Cell. 2007;27:622–35.
164. Sears N, Sen GC, Stark GR, Chattopadhyay S. Caspase-8- 176. Li C, Ma L, Chen X. Interferon regulatory factor 3-CL, an
mediated cleavage inhibits IRF-3 protein by facilitating its isoform of IRF3, antagonizes activity of IRF3. Cell Mol
proteasome-mediated degradation. J Biol Chem. 2011;286: Immunol. 2011;8:67–74.
33037–44. 177. Karpova AY, Ronco LV, Howley PM. Functional character-
165. Praefcke GJK, Hofmann K, Dohmen RJ. SUMO playing tag ization of interferon regulatory factor 3a (IRF-3a), an alternative
with ubiquitin. Trends Biochem Sci. 2011;37:23–31. splice isoform of IRF-3. Mol Cell Biol. 2001;21:4169–76.
166. Denuc A, Marfany G. SUMO and ubiquitin paths converge. 178. Marozin S, Altomonte J, Stadler F, Thasler WE, Schmid RM,
Biochem Soc Trans. 2010;38:34–9. Ebert O. Inhibition of the IFN-beta response in hepatocellular
167. Ran Y, et al. SENP2 negatively regulates cellular antiviral carcinoma by alternative spliced isoform of IFN regulatory
response by deSUMOylating IRF3 and conditioning it for factor-3. Mol Ther. 2008;16:1789–97.
ubiquitination and degradation. J Mol Cell Biol. 2011;3:283–92. 179. Goh FG, Thomson SJP, Krausgruber T, Lanfrancotti A, Copley
168. Dastur A, Beaudenon S, Kelley M, Krug RM, Huibregtse JM. RR, Udalova IA. Beyond the enhanceosome: cluster of novel jB
Herc5, an interferon-induced HECT E3 enzyme, is required for sites downstream of the human IFN-b gene is essential for
conjugation of ISG15 in human cells. J Biol Chem. 2006; lipopolysaccharide-induced gene activation. Blood. 2010;116:
281:4334–8. 5580–8.
169. Wong JJY, Pung YF, Sze NS-K, Chin K-C. HERC5 is an IFN- 180. Yount JS, Gitlin L, Moran TM, Lopez CB. MDA5 participates
induced HECT-type E3 protein ligase that mediates type I IFN- in the detection of paramyxovirus infection and is essential for
induced ISGylation of protein targets. Proc Natl Acad Sci. the early activation of dendritic cells in response to Sendai virus
2006;103:10735–40. defective interfering particles. J Immunol. 2008;180:4910–8.
170. Lu G, et al. ISG15 enhances the innate antiviral response by 181. Honda K, et al. IRF-7 is the master regulator of type-I inter-
inhibition of IRF-3 degradation. Cell Mol Biol (Noisy-le-grand) feron-dependent immune responses. Nature. 2005;434:772–7.
2006;52:29–41. 182. Carrigan SO, et al. IFN regulatory factor 3 contributes to the
171. Shi H-X, et al. Positive regulation of interferon regulatory factor host response during Pseudomonas aeruginosa lung infection in
3 activation by Herc5 via ISG15 modification. Mol Cell Biol. mice. J Immunol. 2010;185:3602–9.
2010;30:2424–36.
172. Prinarakis E, Chantzoura E, Thanos D, Spyrou G. S-glutath-
ionylation of IRF3 regulates IRF3-CBP interaction and activa-
tion of the IFN[beta] pathway. EMBO J. 2008;27:865–75.

123

You might also like