Toll-Like Receptors and Innate Immunity

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

J Mol Med (2006) 84:712–725

DOI 10.1007/s00109-006-0084-y

REVIEW

Toll-like receptors and innate immunity


Satoshi Uematsu & Shizuo Akira

Received: 10 April 2006 / Accepted: 8 June 2006 / Published online: 10 August 2006
# Springer-Verlag 2006

Abstract The innate immune system is an evolutionally


conserved host defense mechanism against pathogens.
Innate immune responses are initiated by pattern recogni-
tion receptors (PRRs), which recognize specific structures
of microorganisms. Among them, Toll-like receptors
(TLRs) are capable of sensing organisms ranging from
bacteria to fungi, protozoa, and viruses, and play a major
role in innate immunity. However, TLRs recognize patho-
gens either on the cell surface or in the lysosome/endosome
compartment. Recently, cytoplasmic PRRs have been
identified to detect pathogens that have invaded cytosols.
In this review, we focus on the functions of PRRs in innate
immunity and their downstream signaling cascades. Dr. SATOSHI UEMATSU Dr. SHIZUO AKIRA
received his M.D. in 1997 from received his M.D. and Ph.D.
Keywords TLR . NLR . RIG-I Osaka University medical degree from the Osaka
school, Osaka City, Japan. He University, Japan. He is
obtained his Ph.D. in 2004 at currently a professor in the
Osaka University. He is Department of Host Defense at
Introduction currently an assistant professor the Research Institute for
of the Department of Host Microbial Diseases, Osaka
Defense at the Research Institute University. His research interests
The innate immune response is the first line of defense for Microbial Diseases, Osaka focus on pathogen recognition
against microbial infections. Since the discovery of the University, Japan. His research and signaling pathways of
Drosophila protein Toll, which induces effective immune focuses on innate immunity, Toll-like receptors and
responses to Aspergillus fumigatus [1], accumulating especially the defense TLR-independent pathogen
mechanism of macrophages and recognition in innate immunity.
evidence showed that the innate immune system specifically dendritic cells in intracellular
recognizes invading microorganisms. The targets of innate bacterial infection.

S. Uematsu : S. Akira immune recognition are the conserved molecular patterns


Department of Host Defense, (pathogen-associated molecular patterns, PAMPs) of micro-
Research Institute for Microbial Diseases, Osaka University,
organisms. Receptors in innate immunity are therefore called
3-1 Yamada-oka,
Suita, Osaka 565-0851, Japan pattern recognition receptors (PRRs) [2]. PAMPs are
generated by microbes and not by the host, suggesting that
S. Akira (*) PAMPs are good targets for innate immunity to discriminate
ERATO, Japan Science and Technology Corporation,
between self- and nonself. Furthermore, PAMPs are essential
3-1 Yamada-oka,
Suita, Osaka 565-0871, Japan for microbial survival and are conserved structures among
e-mail: sakira@biken.osaka-u.ac.jp many pathogens, which allows innate immunity to recognize
J Mol Med (2006) 84:712–725 713

microorganisms with limited numbers of PRRs. Among various cells such as macrophages and DCs [7]. LPS forms a
PRRs, Toll-like receptors (TLRs), mammalian homologs of complex with an accessory protein, LPS-binding protein in
Toll, were highlighted as the key recognition structures of serum, which converts oligomeric micelles of LPS to a
the innate immune system in the past few years [3]. TLRs monomer for delivery to CD14, a glycosylphosphatidylino-
are capable of sensing organisms ranging from bacteria to sitol (GPI)-anchored, high-affinity membrane protein that
fungi, protozoa, and viruses. However, host recognition and can also circulate in a soluble form. CD14 concentrates LPS
response is not restricted to TLRs. Some members of the for binding to the TLR4/MD2 complex [8].
NACHT (domain present in NAIP, CIITA, HET-E, and TLR2 recognizes various microbial components, such as
TP1)-LRR (leucine-rich repeat) (NLR) family, which lipoproteins/lipopeptides and peptidoglycans from gram-pos-
includes both nucleotide-binding oligomerization domain itive and gram-negative bacteria, and lipoteichoic acid from
(NOD) proteins and NACHT-LRR- and pyrin-domain- gram-positive bacteria, a phenol-soluble modulin from Staph-
containing proteins (NALPs), recognize microbial compo- ylococcus aureus, and glycolipids from Treponema maltophi-
nents in the cytosol [4]. Furthermore, the RNA helicase, lum [3, 9]. TLR2 is also reported to be involved in the
retinoic acid inducible gene-I (RIG-I), was identified as a recognition of atypical LPS from nonenterobacteria, whose
cytosolic receptor for intracellular double-stranded (dsRNA) structures are different from typical LPS of gram-negative
[5] and induces type I interferons (IFNs) in response to bacteria [3, 10]. However, a recent report has indicated that
dsRNA viruses in a TLR-independent manner [6]. The lipoproteins contaminated in a LPS preparation from Por-
recent understanding of these PRRs in innate immunity and phyromonas gingivalis stimulated TLR2 and that LPS from P.
their signaling pathways is the focus of this review. gingivalis itself had poor activity for TLR4 stimulation [11].
There are also controversial reports regarding peptidoglycan
recognition by TLR2. More careful analyses will be needed to
TLR exclude any possibility of contaminates remaining.
TLR1 and TLR6 are structurally related to TLR2 [12].
A mammalian homolog of a Toll receptor (now termed TLR2 and TLR1 or TLR6 form a heterodimer, which is
TLR4) was identified through database searches. The TLR involved in the discrimination between the molecular
family now consists of 13 mammalian members: with each structure of diacyl and triacyl lipopeptides [13–15]. CD36,
TLR having its intrinsic signaling pathway and inducing a member of the class II scavenger family of proteins was
specific biological responses against microorganisms. Rec- shown to serve as a facilitator or coreceptor for diacyl
ognition of microbial components by TLRs triggers the lipopeptide recognition through the TLR2/6 complex [16].
activation of signal transduction pathways, which then TLR5 is responsible for the recognition of flagellin, the
induces dendritic cell (DC) maturation and cytokine produc- major portion of bacterial flagella [17]. Gewirtz et al. [18]
tion, resulting in development of adaptive immunity [3]. reported that TLR5 is expressed on the basolateral surface,
but not the apical side of intestinal epithelial cells,
Structure suggesting that TLR5 serves as the sensor for the
pathogenic bacteria that invade across the epithelium. A
The cytoplasmic portion of a TLR is similar to that of the common stop codon polymorphism in the ligand-binding
interleukin (IL)-1 receptor family. It is therefore called the domain of TLR5 (TLR5 392STOP SNP) is unable to
Toll/IL-1 receptor (TIR) domain. A TIR domain is required mediate flagellin signaling and is associated with suscep-
for initiating intracellular signaling. The extracellular region tibility to pneumonia caused by Legionella pneumophila
of TLRs and IL-1R are markedly different. Whereas IL-1R [19]. However, researchers in Vietnam have reported that
possesses an Ig-like domain, TLRs contain LRRs in the the TLR5 392STOP SNP is not associated with suscepti-
extracellular domain. LRRs are responsible for the recog- bility to typhoid fever [20]. A recent work has reported that
nition of PAMPs [3] (Fig. 1). some commensal bacteria, such as a and e Proteobacteria,
change the TLR5 recognition site of flagellin without losing
Pathogen recognition flagellar motility [21]. This modification may contribute to
the persistence of these bacteria on mucosal surfaces.
Bacteria Bacterial DNA, which contains unmethylated CpG
motifs, is a potent stimulator of the host immune response.
Lipopolysaccharide (LPS) is a cell wall component of gram- In vertebrates, the frequency of CpG motifs is severely
negative bacteria, a strong immunostimulant (Table 1). LPS reduced and the cysteine residues of CpG motifs are highly
is composed of lipid A (endotoxin), core oligosaccharide, methylated, which leads to abrogation of the immunostim-
and O-antigen. TLR4 recognizes lipid A of LPS; this occurs ulatory activity. Analysis of TLR9-deficient mice showed
through the TLR4/MD2/CD14 complex, which is present on that TLR9 is a receptor for CpG DNA [22].
714 J Mol Med (2006) 84:712–725

Fig. 1 TLR signaling pathways. TLR signaling pathways originate TLR4-mediated IRF3 activation. In addition, TLR4 also requires
from the cytoplasmic TIR domain. A TIR domain-containing adaptor, another adaptor, TRAM, for IRF3 activation. TRIF interacts with
MyD88, associates with the cytoplasmic TIR domain of TLRs and TBK1, IKKi, RIP1, and TRAF6. IRF3 is activated by TBK1 and
recruits IRAKs to the receptor upon ligand binding. IRAKs then IKKi, which catalyze IRF3 phosphorylation and stimulate its nuclear
activates TRAF6, leading to the activation of TAK1. TAK1 activates translocation and DNA binding. pDCs express TLR7 (also TLR8 in
the IKK complex consisting of IKKα, IKKβ, and NEMO/IKKγ. The human) and TLR9 and secrete large amounts of IFN-αs and IFN-β in
IKK complex phosphorylates IκB, resulting in nuclear translocation of response to ssRNA and CpG DNA. This induction by TLR7 and
NF-kB, which induces expression of inflammatory cytokines. Among TLR9 depends entirely on MyD88. After stimulation, IRF7 trans-
the TLR family members, TLR4 on cell surfaces and TLR3, 7, 8, and locates into the nucleus to induce type I IFNs. IRF7 is activated
9 on endosomal membrane can induce type I IFNs. Stimulation of through forming a signaling complex with MyD88, IRAKs, and
macrophages and DCs with LPS and dsRNA results in the activation TRAF6 in the cytoplasm. IRAK-1 but not IRAK-4 is an essential
of IRF3 and subsequent induction of IFN-β and IFN-inducible genes. enzyme for phosphorylation and activation of IRF7 in TLR7- and
A TIR domain-containing adapter, TRIF is responsible for TLR3- and TLR9-mediated type I IFNs induction

Mouse TLR11, a relative of TLR5, is expressed abun- Several components located in the cell wall or cell surface
dantly in the kidney and bladder. TLR11-deficient mice are of fungi were identified as potential ligands for TLRs. Yeast
susceptible to uropathogenic bacterial infections, indicating zymosan activates TLR2/TLR6 heterodimers, whereas
that TLR11 senses the component of uropathogenic bacteria. mannan, derived from Saccharomyces cerevisiae and C.
However, TLR11 is thought to be nonfunctional in human albicans is detected by TLR4. Phospholipomannan,
because of the presence of stop codon [23]. present on the cell surface of C. albicans, is recognized
by TLR2, while TLR4 mainly interacts with glucuronox-
Fungi ylomannan, the major capsular polysaccharide of C.
neoformans [24].
TLRs are involved in the recognition of fungal pathogens Dectin-1 is a lectin family receptor for the fungal cell
such as Candida albicans, A. fumigatus, Cryptococcus wall component, b-glucan [25]. Dectin-1 functionally
neoformans, and Pneumocystis carinii [3, 24] (Table 1). collaborates with TLR2 and induces a strong immune
J Mol Med (2006) 84:712–725 715

Table 1 Pathogen recognition of TLRs TLR2 is also involved in the recognition of viral
components such as measles virus, human cytomegalovirus,
TLRs Ligands
and herpes simplex virus type 1 (HSV-1) [35–37].
TLR1 Triacyl lipopeptides (bacteria) During viral replication, dsRNA is generated, and TLR3
TLR2 Peptidoglycan, lipoprotein, lipopeptides, atypical is involved in the recognition of a synthetic analog of
LPS (bacteria) dsRNA, polyinosine–deoxycytidylic acid (poly I:C), a
Zymosan, phospholipomannan (fungi) potent inducer of type I IFNs [38, 39]. TLR3-deficient
GPI anchor (protozoa)
mice are susceptible to mouse cytomegalovirus [40]. On the
Envelope protein (virus)
TLR3 Poly (I:C), dsRNA (virus) other hand, TLR3-deficient mice show more resistance to
TLR4 LPS (bacteria) West Nile virus (WNV) infection. This single-stranded
Mannan, glucuronoxylomannan (fungi) RNA (ssRNA) flavivirus triggers inflammatory responses
Glycoinositolphospholipids (protozoa) via TLR3, which leads a disruption of the blood brain
RSV fusion protein (virus) barrier, followed by enhanced brain infection [41]. These
TLR5 Flagellin (bacteria) findings suggested that WNV utilizes TLR3 to efficiently
TLR6 Diacyl lipopeptides (bacteria)
enter the brain.
TLR7/TLR8 Synthetic imidazoquinoline-like molecules,
Mouse splenic DCs are divided into CD11c high B220−
ssRNA (virus)
TLR9 CpG DNA (bacteria, protozoa, virus) and CD11c dull B220+ cells. The latter contain plasmacytoid
Hemozoin (protozoa) DCs (pDCs), which induce large amounts of IFN-α during
TLR11 Component of uropathogenic bacteria (bacteria) viral infection. CpG DNA motifs are also found in genomes
Profilin like molecule (protozoa) of DNA viruses, such as HSV-1, HSV-2, and murine
cytomegalovirus (MCMV). Mouse pDCs recognize CpG
TLRs recognize molecular patterns associated with a broad range of
pathogens including bacteria, fungi, protozoa, and viruses DNA of HSV-2 via TLR9 and produce IFN-α [42]. TLR9-
deficient mice are susceptible to MCMV infection, suggest-
ing that TLR9 induces antiviral responses by sensing CpG
response to yeast via recruitment of the tyrosine kinase, Syk DNA of DNA virus [40, 43, 44].
[26–28]. TLR2 recognizes a variety of microbial products TLR7 and TLR8 are structurally highly conserved
through functional cooperation with several proteins, some proteins [12]. The synthetic imidazoquinoline-like mole-
of which are structurally related to TLR2 whereas others cules imiquimod (R-837) and resiquimod (R848) have
are not. potent antiviral activities and are used for clinical treatment
of viral infections. Analysis of TLR7-deficient mice
Protozoa showed that TLR7 recognizes these synthetic compounds
[45]. Human TLR7 and TLR8, but not murine TLR8,
TLRs also sense the components of protozoa including recognize imidazoquinoline compounds [46]. Furthermore,
Trypanosoma cruzi, Trypanosoma brucei, Toxoplasma murine TLR7 recognizes guanosine analogs such as
gondii, Leishmania major, and Plasmodium falciparum loxoribine, which has antiviral and antitumor activities
(Table 1). GPI anchors, glycoinositolphospholipids, and [47]. All these compounds are structurally similar to
genomic DNA-derived from T. cruzi are recognized by ribonucleic acids. After a short while, TLR7 and human
TLR2, TLR4, and TLR9, respectively [29]. Murine TLR11 TLR8 also were shown to recognize guanosine- or uridine-
senses the profilin-like molecule of T. gondii [30]. Malaria rich ssRNA from viruses such as human immunodeficiency
parasites digest host hemoglobin into a hydrophobic heme virus, vesicular stomatitis virus (VSV), and influenza virus
polymer known as hemozoin. TLR9 recognizes hemozoin [48, 49].
from P. falciparum [31]. However, the precise mechanism
of how TLR9 recognizes both DNA and non-DNA crystals Signaling pathways
remains unclear.
The activation of TLR signaling pathways originates
Virus from the cytoplasmic TIR domains. Four TIR domain-
containing adaptors (MyD88, TIRAP/MAL, TRIF, and
TLR4 recognizes not only bacterial components but also TRAM) were shown to play an important role in
viral envelope proteins (Table 1). The fusion protein from TLR signaling pathway. These adaptors are associated
respiratory syncytial virus (RSV) is recognized by TLR4 with TLRs through homophilic interaction of TIR
[32]. TLR4-mutated C3H/HeJ mice are sensitive to RSV domains. Each TLR mediates distinctive responses in
infection [33]. The envelope protein of mouse mammary association with a different combination of these
tumor virus directly activates B cells via TLR4 [34]. adapters (Fig. 1).
716 J Mol Med (2006) 84:712–725

MyD88-dependent pathway tion, TRIF interacts with TBK1 and IKKi, which phos-
phorylates IRF3, allowing IRF3 to translocate into the
TLR family members use many of the same signaling nucleus and activate the IFN-β promoter.
components as IL-1R because both have a conserved TRAF6 is reported to bind TRIF and cooperatively
cytoplasmic domain, the TIR domain. The TIR domain- activate NF-κB [64]. However, cells doubly deficient in
containing adapter MyD88 is utilized by all TLRs with the TRAF6 and MyD88 still partially activated NF-κB in
exception of TLR3. MyD88 possesses a TIR domain in its response to LPS, suggesting that TRIF activates NF-κB
C terminus and a death domain in its N terminus. Upon through both TRAF6-dependent and TRAF6-independent
stimulation, MyD88 recruits IL-1 receptor kinase (IRAK) to pathways in TLR4 signaling [65, 66].
TLRs through the interaction of both molecules’ death Receptor-interacting protein-1 (RIP1) was implicated in
domains. Two members of the IRAK family, IL-1 receptor- the TLR3-mediated NF-κB response to dsRNA. RIP1 binds
associated kinase (IRAK-1) and IRAK-4 are activated by the C terminus of TRIF via a RIP homotypic interaction
phosphorylation, and associate with TRAF6. TRAF6 forms motif (RHIM). In cells lacking RIP1, TLR3-mediated NF-
a complex with ubiquitin-conjugating enzymes to activate κB activation and the subsequent induction of intercellular
TAK1, leading to the activation of nuclear factor (NF)-κB adhesion molecule 1 is severely impaired, whereas IFN-β
(IκB) and activator protein-1 through the canonical IκB induction is intact. By contrast, responses to LPS were
kinase (IKK) complex and the mitogen-activated protein normal in the absence of RIP1. However, LPS fails to
kinase pathway, respectively. The IKK complex phosphor- stimulate NF-κB activation in rip−/−MyD88−/− cells,
ylates the inhibitor of IκB, which sequesters the transcrip- revealing that RIP1 is also required for the TRIF-dependent
tion factor NF-κB to induce target genes. This signaling TLR4-induced NF-κB pathway. In the TNF-α pathway,
pathway is called the “MyD88-dependent pathway” and is RIP1 interacts with the E3 ubiquitin ligase TRAF2 and is
essential for the expression of inflammatory cytokine modified by a polyubiquitin chain. Upon TLR3 activation,
genes, including TNF-α, IL-6, IL-12, and IL-1β, and RIP1 is also modified by polyubiquitin chains and is
costimulatory molecules [3]. In the case of the TLR2 and recruited to TLR3 along with TRAF6 and TAK1, suggest-
TLR4 signaling pathways, TIRAP/MAL is also required for ing that RIP1 uses a similar ubiquitin-dependent mecha-
the activation of the MyD88-dependent pathway [50, 51]. nism to activate IKK-β in response to TNF-α and TLR3
ligands [67].
MyD88-independent pathway
Specific pathway in pDCs
In addition to proinflammatory signals, some members of
the TLR family trigger the induction of type I IFNs. In pDCs are specialized for producing large amounts of type I
particular, TLR3 and TLR4 have the ability to induce IFN- IFNs during viral infection [68]. pDCs highly express
β and IFN-inducible genes in a MyD88-independent TLR7 and TLR9 and produce high levels of IFN-α in
manner [52]. Thus, this signaling pathway is called the response to TLR7 and TLR9 ligands [47]. It appears that
MyD88-independent pathway. A third adapter, TRIF [53, IFN-α induction by TLR7 and TLR9 depends entirely on
54], is essential for the TLR3- and TLR4-mediated MyD88 [69]. IRF7 is a transcriptional factor structurally
MyD88-independent pathway [39, 55]. TRIF is also related to IRF3, which is expressed constitutively in pDCs.
required for the induction of inflammatory cytokines in Overexpression of IRF7 activates IFN-α- and IFN-β-
the TLR4 signaling pathway. dependent promoters. IRF7 forms a signaling complex with
A fourth adapter, TRAM is essential for the TLR4- MyD88 and TRAF6 in the cytoplasm [70, 71]. After ligand
mediated MyD88-independent pathway [56–58]. The N stimulation, IRF7 translocates into the nucleus to induce
terminus of TRAM has a myristoylation site, the mutation IFN-αs [70, 71]. Similar to IRF3, IRF7 is activated by its
of which alters its normal membrane localization and phosphorylation. However, in TBK1-deficient cells, TLR9-
abolishes the TLR4 signaling. TRAM acts as a bridging mediated IFN-α production is still observed [70]. Mouse
adapter between TLR4 and TRIF [58]. pDCs lacking IRAK-4 fails to produce both inflammatory
TLR3- and TLR4-mediated signaling activates a member cytokines and IFN-αs [71]. Human TLR7-, TLR8-, and
of the IRF family of transcription factors, IRF3 to induce TLR9-mediated induction of IFN-α/β and -λ was also
IFN-β and IFN-inducible genes. Two noncanonical IKKs, IRAK-4 dependent [72]. Recently, IRAK-1 was shown to
inducible IKK (IKKi)/IKKɛ and TRAF family member- serve as an IRF7 kinase. In IRAK-1-deficient mice, TLR7-
associated NF-κB activator-binding kinase 1 (TBK1)/NF- and TLR9-induced IFN-α production was completely
κB-activating kinase (NAK)/TRAF2-associated kinase abolished, although inflammatory cytokines are normally
(T2K), were shown to be responsible for TLR3- and produced. Furthermore, IRF7 activation by TLR9 ligand is
TLR4-mediated IRF-3 activation [59–63]. Upon stimula- impaired in IRAK-1-deficient mice, in spite of normal
J Mol Med (2006) 84:712–725 717

NF-κB activation. IRAK-1 but not IRAK-4 can directly to physically interact with NOD1 and RICK (but not with
bind and phosphorylate IRF7; thus, IRAK-1 specifically NOD2) and suppress NOD1/RICK-mediated NF-κB activa-
mediates IFN-α induction downstream of MyD88 and tion [86]. In addition to NF-κB activation and NOD1 and
IRAK-4 [73]. NOD2 signaling give rise to the activation of the mitogen-
IRF8 is involved in TLR9-mediated responses because activated protein kinase (MAPK) pathway although the
pDCs lacking IRF8 fails to produce proinflammatory precise mechanism remains unknown [4] (Fig. 2).
cytokines and IFN-α in response to TLR9 ligand; in Inflammasome is a multiprotein complex and is respon-
IRF8-deficient cells, TLR9-stimulated NF-κB activation is sible for the activation of caspase 1 and 5, leading to the
unexpectedly impaired, suggesting that IRF8 mediates processing and secretion of the proinflammatory cytokines
NF-κB activation in TLR9 signaling [74]. IL-1β and IL-18 [87]. Two types of the inflammasome
were reported to date: NALP1 inflammasome and NALP2/
3 inflammasome (Fig. 3). Whereas NALP1 inflammasome
NLR is composed of NALP1, ASC, caspase 1, and caspase 5,
NALP2/3 inflammasome contains NALP2/3, ASC, caspase
Structure 1, and Cardinal. NALPs are the chief platforms for
inflammasome [88]. ASC is an essential adapter molecule
Mammalian NLR family members are also thought to that connects the NALPs to caspase 1. Because NALP2/3
recognize PAMPs. NLR proteins contain three characteristic do not have FIND domain and CARD domains in the C
domains. The first is a LRR domain, which is involved in terminus, the NALP2/3 inflammasome contains Cardinal,
ligand recognition. The second is a centrally located NOD which has the same structural organizations as the C-
(also known as a NACHT domain), which acts as an ATP- terminal region of NALP1 [88]. All of the inflammasome
dependent dimerization domain. The last N-teminal domain is components reside in the cytoplasm. Activation of inflam-
composed of protein–protein interaction cassettes. NLRs are masome is thought to occur through the recognition of
divided into subgroups according to the N-terminal domain: PAMPS by NALPs. However, the precise activation
caspase-recruitment domain (CARD), pyrin domain (PYD), mechanism remains unknown. After activation of inflam-
baculoviral inhibitory repeat (BIR)-like, and unclassified. masome, activated caspases cleave the proinflammatory
NODs, NALPs, and IPAFs contain CARDs, PYDs, and BIR, cytokines, IL-1β and IL-18. Processing leads to the
respectively. All these domains are involved in the regulation secretion of mature IL-1β and IL-18 and induces inflam-
of proapoptotic and proinflammatory signaling [75]. matory responses. IL-1β production is tightly controlled to
avoid excessive inflammation. Caspase-1 activation is
Pathogen recognition and signaling pathway inhibited by CARD containing ICEBERG, COP, and
DASC (POP1) and possibly by human caspase-12. Acti-
Recent reports showed that some members of the NLR vated caspase-1 is blocked by PI-9 and IL-1β activity is
family are involved in intracellular recognition of microbial inhibited by IL1Ra [89].
products. Although initial studies identified LPS as a The NALP3/cryopyrin/CIAS1 inflammasome is activated
NOD2 ligand [76], it is well established now that NOD1 by MDP. Recent reports showed that NALP3/cryopyrin/
and NOD2 recognize peptidoglycan-derived peptides, γ-D- CIAS1 is involved in the recognition of bacterial RNA, ATP,
glutamyl-meso-diaminopimelic acid (iE-DAP) [77] and and uric acid crystals, and is essential for caspase-1
muramyl dipeptide (MDP) [78, 79], respectively. Recogni- activation and IL-1β and IL-18 production in response to
tion of ligands through the LRR domains activates NOD these ligands [90–92] (Fig. 3).
proteins, which then recruit the serine–threonine kinase Another CARD-containing NOD-LRR protein, Ipaf is
RICK (also known as RIP2 or CARDIACK) through a responsible for Salmonella typhimurium-induced, but not
CARD–CARD interaction [80]. In the NOD2 signaling TLR-induced, caspase-1 activation. ASC but not RICK/
pathway, activation of RICK leads to K63 (Lys63)-linked RIP2 is also essential for S. typhimurium-induced caspase-1
polyubiquitylation of NEMO/IKKγ, which is followed by activation [85] (Fig. 3).
phosphorylation of IKKβ and the phosphorylation of IκB, The Lgn1 locus was found to control the intracellular
leading to the translocation of NF-κB to the nucleus [81, replication of L. pneumophila in murine macrophages.
82]. CARD12 (also known as IPAF or CLAN) binds both NAIP5/Birc1e, a NLR protein with BIR domains, was
NOD1 and NOD2 through NOD–NOD interactions [83] identified to be responsible for the Lgn1 effect [93].
and inhibits NOD1- and NOD2-mediated activation of NAIP5/Birc1e is assumed to function as a cytoplasmic
NF-κB in vitro [84]. However, recent reports show that sensor of L.pneumophila and is responsible for L.pneumo-
invasive bacteria lead to the activation of CARD12 and the phila-mediated caspase-1 activation. The pathway used for
induction of expression of IL-1β [85]. CARD6 was shown caspase-1-dependent processing and secretion of IL-1 was
718 J Mol Med (2006) 84:712–725

Fig. 2 Signaling pathways of


NOD1 and NOD2. NOD1 and
NOD2 recognize iE-DAP and
MDP, respectively, and activate
NF-κB via RIP2/RICK. NOD1
and NOD2 signaling gives rise
to the activation of MAPKs by
unknown mechanism. CARD12
negatively regulates RICK/
RIP2-mediated NF-κB
activation by both NOD1 and
NOD2. CARD6 also negatively
regulates RICK/RIP2-mediated
NF-κB activation by NOD1

distinct from the pathway used for caspase-1-dependent Diseases


inhibition of L. pneumophila growth. It is interesting to note
that the adapter molecule ASC is essential for the secretion The importance of NLRs was highlighted by findings that
of IL-1β after L. pneumophila infection but not for the mutations affecting the function of these proteins are
control of L. pneumophila replication. These data suggest associated with the occurrence of autoimmune diseases.
that Ipaf and Asc have separate and distinct functions in NOD2 is associated with two autoinflammatory disorders.
controlling innate immune responses to L. pneumophila [94] A missense point mutation in the human NOD2 gene is
(Fig. 3). correlated with susceptibility to Crohn’s disease [4]. Blau
syndrome, an inheritable disorder characterized by granu-

Fig. 3 Bacterial recognition of


NLRs and their signaling. NLRs
recognize bacterial proteins in
the cytoplasm and trigger signal-
ing pathways. MDP, RNA, ATP,
and uric acid crystal are recog-
nized by NALP3, which forms
an inflammasome comprised of
ASC, CARDINAL, and caspase-
1. NALP1 also forms an inflam-
masome composed of ASC,
caspase-1, and caspase-5. Little
is known about the natural
stimuli that lead to the activation
of NALP1 inflammasome.
Activated caspase-1 cleaves
pro-IL-1β for the maturation of
IL-1β. Another NOD-LRR
protein, IPAF, is activated by S.
typhimurium and induces
maturation of IL-1β associating
with ASC. Naip5/Birc1e is acti-
vated by L. pneumophila and
induces caspase-1 activation in
an IPAF/ASC-dependent manner
J Mol Med (2006) 84:712–725 719

lomatous polyarthritis, panuveitis, and exanthema, was (EMCV), and RNAi of RIG-I type I inhibited induction of
shown to share a mutated form of CARD15 (which encodes type I IFNs in response to Newcastle disease virus (NDV),
NOD2) [95]. Sendai virus, and EMCV [5]. Most RIG-I-deficient mice
NALP3/cryopyrin/CIAS1 is associated with three auto- are embryonic lethal because of severe liver degeneration.
inflammatory syndromes: familial-cold autoinflammatory Conventional DCs and fibroblasts derived from RIG-I-
syndrome, Muckle–Wells syndrome, and neonatal-onset deficient mice are almost completely unable to produce
multisystem inflammatory disease [96, 97]. These diseases type I IFN and inflammatory cytokines after infection with
are now referred as cryopyrin-associated periodic syndrome NDV, Sendai virus, or VSV. However, RIG-I-deficient
(CAPS). CAPS patients have systemic inflammation with pDCs produced type I IFNs normally in response to NDV.
increased levels of C-reactive protein, serum amyloid A, In pDCs, TLR systems are responsible for RNA virus-
and inflammatory cytokines. However, it is not well mediated type I IFN induction. Thus, RIG-I and the TLR
understood how the mutated NALP3/cryopyrin/CIAS1 system exert antiviral responses in a cell-type-specific
contributes to the induction of systemic infection [75]. manner [6].
CIITA is a master transcription regulator of MHC class
II genes [98]. Mutations in CIITA lead to MHC class II Other molecules
deficiency or a rare form of autosomal recessive hereditary
immunodeficiency, group A bare lymphocyte syndrome Melanoma differentiation-associated gene 5 (MDA5, also
[99, 100]. called Helicard) was identified as the DExD/H box RNA
Familial Mediterranean fever (FMF) is an autosomal helicase and contains two CARD-like domains and a single
recessive inherited disease that is characterized by recurrent helicase domain [102–105]. Like RIG-I, overexpression of
episodes of fever accompanied with topical signs of MDA5 enhanced NDV-induced type I IFN production and
inflammation. The gene, Mediterranean fever, which antiviral responses in response to VSV or EMCV. RNAi of
encodes the pyrin (also known as marenostrin) protein is MDA5 blocks NDV-induced activation of type I IFN
mutated in patients with FMF [75]. promoters [105]. MDA5 was identified as a binding target
for V proteins of paramyxoviruses [104]. Although the
precise mechanism is unclear, these V proteins inhibit the
Antiviral RNA helicases dsRNA-induced activation of the IFN-β gene through
MDA5. Virus-encoded proteins may specifically target
Whereas TLR2 and TLR4 recognize viral components at these RNA helicases to escape from antiviral detection by
the cell surface, TLR3, TLR7, TLR8, and TLR9 are the host cells.
exclusively expressed in endosomal compartments. After An RNA helicase Lgp2 is also related to RIG-I and
phagocytes internalize viruses or virus-infected apoptotic MDA5, but lacks the CARD-like domains. Overexpression
cells, viral nucleic acids are released in phagolysosomes of Lgp2 inhibits Sendai virus-induced activation of IFN-β
and are recognized by these TLRs [3]. Recent reports promoter, suggesting that Lgp2 is a negative regulator of
showed that hosts also have a mechanism to detect RIG-I and MDA5 [105, 106] (Fig. 4).
replicating viruses in the cytoplasm in a TLR-independent
manner [101]. Signaling pathway of antiviral RNA helicases

RIG-I The signaling pathway of RIG-I and MDA5 ultimately


engages the IKK complex (IKKα/β/γ) and TBK1/IKKi,
RIG-I was identified as a cytoplasmic receptor for viral which leads to phosphorylation and activation of NF-κB
dsRNA. RIG-I is a DExD/H box RNA helicase, which and IRF3 and IRF7, respectively. IFN-β promoter stimula-
possesses C-terminal helicase domain and two tandem tor 1 (IPS-1) was identified as a molecule that acts
caspase-recruiting domains at the N terminus (Fig. 4). downstream of RIG-I and MDA5. IPS-1 consists of two
RIG-I binds to RNA with its C-terminal helicase domain domains characterized as an N-terminal CARD domain and
and unwinds dsRNA in an ATPase dependent manner. The a C-terminal effector domain. IPS-1 associates with RIG-I
binding of the helicase domain and RNA is likely to induce and MDA5 with a CARD domain. Overexpression of IPS-1
conformational changes that exposes the N-terminal activates IFN-β, IFN-α4, IFN-α6, and NF-κB promoters
CARDs to recruit downstream signaling proteins, leading and inhibits VSV replication. RNAi of IPS-1 blocks type I
to the activation of IRF3 and NF-κB [5]. IFN production in response to intracellular administration
The functional significance of RIG-I was shown in vitro of dsRNA or infection with VSV or NDV. TBK1 and IKKi
and in vivo [5, 6]. Overexpression of RIG-I blocks are required for IPS-1-mediated activation of the type I IFN
replication of VSV and encephalomyocarditis virus promoters, although it does not directly bind to these
720 J Mol Med (2006) 84:712–725

Fig. 4 Signaling pathways of


antiviral RNA helicases. Viruses
produce dsRNA during
replication in cytoplasm. RIG-I
and Mda5 recognize dsRNA
to initiate antiviral signaling.
IPS-1 interacts with RIG-I and
Mda5 via the CARD-like
domain, followed by the
activation of IRF3 and IRF7 via
TBK1- and IKKi-dependent
phosphorylation. IPS-1 also
activates NF-κB via FADD/
RIP1-dependent pathways.
Synthetic dsDNA also activates
type I IFN promoters, although
a receptor responsible for
DNA recognition was not
identified

protein kinases. IPS-1 interacts with FADD and RIP1 via its hepatitis C virus that has serine protease activity essential
effector domain to facilitate NF-κB activation. Thus, IPS-1 for the production of mature viral proteins, was recently
is an adaptor protein that mediates RIG-I- and MDA5- shown to block the activation of IRF3. NS3-4A counteracts
dependent antiviral responses [107]. not only TLR3-dependent pathways by targeting Trif for
IPS-1 was independently identified by three different proteolytic cleavage, but also counteracts TLR3-indepen-
groups: mitochondrial antiviral signaling protein (MAVS) dent pathways by targeting an undefined protein of the
[108], virus-induced signaling adaptor [109], and CARD- RIG-I-dependent pathway [114, 115]. Meylan et al. [110]
adaptor-inducing IFN-β (Cardif) [110]. The MAVS study showed that Cardif is cleaved by wild-type but not by a
shows that MAVS localizes to the outer mitochondrial catalytically inactive form of NS3-4A, which cleaves the
membrane. The mitochondrial-targeting transmembrane C-terminal region of Cardif, causing disruption of IRF3 and
domain is essential for MAVS signaling, implying the NF-κB activation, probably by causing mislocalization of
importance of mitochondria in innate immunity [108] cleaved Cardif from mitochondria. Also, the V proteins of
(Fig. 4). paramyxoviruses, such as simian virus 5, human para-
influenza virus 2, mumps virus, Sendai virus, and Hendra
Viral modification of the host immune system virus, associate with MDA5 and block the downstream
signaling of MDA5 [104, 105]. Thus, for their survival
Viruses have various mechanisms to escape from host many viruses antagonize important components of the host
immunity. Vaccinia virus (VV) protein A52R was shown to antiviral defense.
block the activation of NF-κB by multiple TLRs, in
particular, TLR3 [111, 112]. A52R associates with both DNA recognition in cytoplasm
IRAK2 and TRAF6, and disrupts signaling complexes
containing these proteins. Furthermore, deletion of the DNA, including host DNA, can be recognized indepen-
A52R gene from VV reduced virus virulence [112]. The dently of TLR9. DNase II, a chief DNase present in the
other VV protein, A46R, has a TIR domain, and as such is phagosome of macrophages, clears DNA from phagocy-
the only viral member of the IL-1R/TLR family identified tosed apoptotic cells or debris [116]. In DNase II-deficient
to date [111]. Recent studies revealed that A46R inhibited mice, macrophages begin to produce IFN-β, which lead to
IL-1R/TLR, but not TNF-induced NF-κB activation, by subsequent TLR9-independent inflammatory events [117].
associating with TIR-domain containing adaptor molecules Furthermore, dsDNA but not ssDNA derived from either
[111, 113]. A46R interacts with multiple TIR-containing pathogens or the host activates both immune and nonim-
adaptors and thereby inhibits the activation of IRF3 and mune cells when introduced into the cytoplasm by
NF-κB [113]. NS3-4A, a multifunctional protein of transfection [118, 119]. Double-stranded breaks form of
J Mol Med (2006) 84:712–725 721

DNA (B-DNA) but not Z-form DNA stimulated mouse and 5. Yoneyama M, Kikuchi M, Natsukawa T, Shinobu N, Imaizumi T,
human stromal cells and DCs, resulting in the production of Miyagishi M, Taira K, Akira S, Fujita T (2004) The RNA helicase
RIG-I has an essential function in double-stranded RNA-induced
type I IFN and chemokines. B-DNA activated IRF3 and the innate antiviral responses. Nat Immunol 5:730–737
IFN-β promoter via both TBK1 and IKKi, whereas it 6. Kato H, Sato S, Yoneyama M, Yamamoto M, Uematsu S, Matsui
activated transcription factor NF-κB independently of both K, Tsujimura T, Takeda K, Fujita T, Takeuchi O, Akira S (2005)
TBK1 and IKKi. Both of these pathways required the Cell type-specific involvement of RIG-I in antiviral response.
Immunity 23:19–28
adaptor molecule IPS-1 in vitro but not TLRs or RIG-I and 7. Shimazu R, Akashi S, Ogata H, Nagai Y, Fukudome K, Miyake
MDA5. In addition, B-DNA activation of fibroblasts K, Kimoto M (1999) MD-2, a molecule that confers lipopoly-
conferred resistance to viral infection, suggesting that saccharide responsiveness on Toll-like receptor 4. J Exp Med
B-DNA-induced IFN genes might be important in antiviral 189:1777–1782
8. Takeda K, Kaisho T, Akira S (2003) Toll-like receptors. Annu
innate immune responses. Furthermore, B-DNA activation Rev Immunol 21:335–376
of the signaling pathways may be involved in the pathogen- 9. Takeuchi O, Hoshino K, Kawai T, Sanjo H, Takada H, Ogawa T,
esis of nucleic acid-dependent autoimmune diseases [120] Takeda K, Akira S (1999) Differential roles of TLR2 and TLR4
(Fig. 4). It is thus necessary to identify a cytoplasmic in recognition of gram-negative and gram-positive bacterial cell
wall components. Immunity 11:443–451
detector of DNA. 10. Netea MG, van Deuren M, Kullberg BJ, Cavaillon JM, Van der
Meer JW (2002) Does the shape of lipid A determine the
interaction of LPS with Toll-like receptors? Trends Immunol
Conclusion 23:135–139
11. Hashimoto M, Asai Y, Ogawa T (2004) Separation and structural
analysis of lipoprotein in a lipopolysaccharide preparation from
Since the discovery of mammalian TLRs, there was a rapid Porphyromonas gingivalis. Int Immunol 16:1431–1437
clarification of the molecular mechanisms of pathogen 12. Akira S (2004) Toll receptor families: structure and function.
recognition. Research on TLRs has also revealed that Semin Immunol 16:1–2
13. Takeuchi O, Kawai T, Muhlradt PF, Morr M, Radolf JD,
recognition of intracellular pathogens is also mediated via Zychlinsky A, Takeda K, Akira S (2001) Discrimination of
a TLR-independent mechanism. Newly described NLR bacterial lipoproteins by Toll-like receptor 6. Int Immunol
family members, which recognize pathogens in the cyto- 13:933–940
plasm, are genetically linked to several human immuno- 14. Takeuchi O, Sato S, Horiuchi T, Hoshino K, Takeda K, Dong Z,
Modlin RL, Akira S (2002) Cutting edge: role of Toll-like receptor
logical disorders. Innate immune responses via RIG-I are 1 in mediating immune response to microbial lipoproteins.
essential for the control of viral replication in the J Immunol 169:10–14
cytoplasm. Furthermore, TLR system and RIG-I system 15. Alexopoulou L, Thomas V, Schnare M, Lobet Y, Anguita J,
were tactfully utilized against viral infection in a cell-type- Schoen RT, Medzhitov R, Fikrig E, Flavell RA (2002)
Hyporesponsiveness to vaccination with Borrelia burgdorferi
specific manner. Thus, hosts have developed the skillful OspA in humans and in TLR1- and TLR2-deficient mice. Nat
and complicated innate immune system to counter the Med 8:878–884
various pathogens with various modes of infection. Future 16. Hoebe K, Georgel P, Rutschmann S, Du X, Mudd S, Crozat K,
studies on PRRs should provide a more comprehensive Sovath S, Shamel L, Hartung T, Zahringer U, Beutler B (2005)
CD36 is a sensor of diacylglycerides. Nature 433:523–527
understanding of innate immune responses. 17. Hayashi F, Smith KD, Ozinsky A, Hawn TR, Yi EC, Goodlett
DR, Eng JK, Akira S, Underhill DM, Aderem A (2001) The
Acknowledgements We thank M. Hashimoto for secretarial assis- innate immune response to bacterial flagellin is mediated by
tance. This work was supported by grants from Special Coordination Toll-like receptor 5. Nature 410:1099–1103
Funds, the Ministry of Education, Culture, Sports, Science and 18. Gewirtz AT, Navas TA, Lyons S, Godowski PJ, Madara JL
Technology, and the Japan Research Foundation for Clinical (2001) Cutting edge: bacterial flagellin activates basolaterally
Pharmacology. expressed TLR5 to induce epithelial proinflammatory gene
expression. J Immunol 167:1882–1885
19. Hawn TR, Verbon A, Lettinga KD, Zhao LP, Li SS, Laws RJ,
Skerrett SJ, Beutler B, Schroeder L, Nachman A, Ozinsky A,
References
Smith KD, Aderem A (2003) A common dominant TLR5 stop
codon polymorphism abolishes flagellin signaling and is
1. Lemaitre B, Nicolas E, Michaut L, Reichhart JM, Hoffmann JA associated with susceptibility to legionnaires’ disease. J Exp
(1996) The dorsoventral regulatory gene cassette spatzle/Toll/ Med 198: 1563–1572
cactus controls the potent antifungal response in Drosophila 20. Dunstan SJ, Hawn TR, Hue NT, Parry CP, Ho VA, Vinh H, Diep
adults. Cell 86:973–983 TS, House D, Wain J, Aderem A, Hien TT, Farrar JJ (2005) Host
2. Medzhitov R, Janeway CJ (1997) Innate immunity: the virtues of susceptibility and clinical outcomes in toll-like receptor
a nonclonal system of recognition. Cell 91:295–298 5-deficient patients with typhoid fever in Vietnam. J Infect Dis
3. Akira S, Uematsu S, Takeuchi O (2006) Pathogen recognition 191:1068–1071
and innate immunity. Cell 124:783–801 21. Andersen-Nissen E, Smith KD, Strobe KL, Barrett SL,
4. Strober W, Murray PJ, Kitani A, Watanabe T (2006) Signalling Cookson BT, Logan SM, Aderem A (2005) Evasion of Toll-
pathways and molecular interactions of NOD1 and NOD2. Nat like receptor 5 by flagellated bacteria. Proc Natl Acad Sci
Rev Immunol 6:9–20 USA 102:9247–9252
722 J Mol Med (2006) 84:712–725

22. Hemmi H, Takeuchi O, Kawai T, Kaisho T, Sato S, Sanjo H, 39. Yamamoto M, Sato S, Hemmi H, Hoshino K, Kaisho T, Sanjo
Matsumoto M, Hoshino K, Wagner H, Takeda K, Akira S (2000) H, Takeuchi O, Sugiyama M, Okabe M, Takeda K, Akira S
A Toll-like receptor recognizes bacterial DNA. Nature 408: (2003) Role of adaptor TRIF in the MyD88-independent toll-
740–745 like receptor signaling pathway. Science 301:640–643
23. Zhang D, Zhang G, Hayden MS, Greenblatt MB, Bussey C, 40. Tabeta K, Georgel P, Janssen E, Du X, Hoebe K, Crozat K,
Flavell RA, Ghosh S (2004) A toll-like receptor that prevents Mudd S, Shamel L, Sovath S, Goode J, Alexopoulou L, Flavell
infection by uropathogenic bacteria. Science 303:1522–1526 RA, Beutler B (2004) Toll-like receptors 9 and 3 as essential
24. Netea MG, Van der Graaf C, Van der Meer JW, Kullberg BJ components of innate immune defense against mouse cytomeg-
(2004) Recognition of fungal pathogens by Toll-like receptors. alovirus infection. Proc Natl Acad Sci USA 101:3516–3521
Eur J Clin Microbiol Infect Dis 23:672–676 41. Wang T, Town T, Alexopoulou L, Anderson JF, Fikrig E,
25. Brown GD, Taylor PR, Reid DM, Willment JA, Williams DL, Flavell RA (2004) Toll-like receptor 3 mediates West Nile
Martinez-Pomares L, Wong SY, Gordon S (2002) Dectin-1 is virus entry into the brain causing lethal encephalitis. Nat
a major beta-glucan receptor on macrophages. J Exp Med Med 10: 1366–1373
196:407–412 42. Lund J, Sato A, Akira S, Medzhitov R, Iwasaki A (2003) Toll-
26. Gantner BN, Simmons RM, Canavera SJ, Akira S, Underhill like receptor 9-mediated recognition of herpes simplex virus-2
DM (2003) Collaborative induction of inflammatory responses by plasmacytoid dendritic cells. J Exp Med 198:513–520
by dectin-1 and Toll-like receptor 2. J Exp Med 197:1107–1117 43. Krug A, French AR, Barchet W, Fischer JA, Dzionek A, Pingel
27. Rogers NC, Slack EC, Edwards AD, Nolte MA, Schulz O, JT, Orihuela MM, Akira S, Yokoyama WM, Colonna M (2004)
Schweighoffer E, Williams DL, Gordon S, Tybulewicz VL, TLR9-dependent recognition of MCMV by IPC and DC
Brown GD, Reis ESC (2005) Syk-dependent cytokine induction generates coordinated cytokine responses that activate antiviral
by Dectin-1 reveals a novel pattern recognition pathway for C NK cell function. Immunity 21:107–119
type lectins. Immunity 22:507–517 44. Krug A, Luker GD, Barchet W, Leib DA, Akira S, Colonna M
28. Underhill DM, Rossnagle E, Lowell CA, Simmons RM (2004) herpes simplex virus type 1 activates murine natural
(2005) Dectin-1 activates Syk tyrosine kinase in a dynamic interferon-producing cells through toll-like receptor 9. Blood
subset of macrophages for reactive oxygen production. 103:1433–1437
Blood 106:2543–2550 45. Hemmi H, Kaisho T, Takeuchi O, Sato S, Sanjo H, Hoshino K,
29. Gazzinelli RT, Ropert C, Campos MA (2004) Role of the Toll/ Horiuchi T, Tomizawa H, Takeda K, Akira S (2002) Small anti-
interleukin-1 receptor signaling pathway in host resistance and viral compounds activate immune cells via the TLR7 MyD88-
pathogenesis during infection with protozoan parasites. Immunol dependent signaling pathway. Nat Immunol 3:196–200
Rev 201:9–25 46. Ito T, Amakawa R, Kaisho T, Hemmi H, Tajima K, Uehira K,
30. Yarovinsky F, Zhang D, Andersen JF, Bannenberg GL, Serhan Ozaki Y, Tomizawa H, Akira S, Fukuhara S (2002) Interferon-
CN, Hayden MS, Hieny S, Sutterwala FS, Flavell RA, Ghosh S, alpha and interleukin-12 are induced differentially by Toll-like
Sher A (2005) TLR11 activation of dendritic cells by a protozoan receptor 7 ligands in human blood dendritic cell subsets. J Exp
profilin-like protein. Science 308(5728):1626–1629 Med 195:1507–1512
31. Coban C, Ishii KJ, Kawai T, Hemmi H, Sato S, Uematsu S,
47. Akira S, Hemmi H (2003) Recognition of pathogen-associated
Yamamoto M, Takeuchi O, Itagaki S, Kumar N, Horii T, Akira S
molecular patterns by TLR family. Immunol Lett 85:85–95
(2005) Toll-like receptor 9 mediates innate immune activation by
48. Diebold SS, Kaisho T, Hemmi H, Akira S, Reis E, Sousa C
the malaria pigment hemozoin. J Exp Med 201:19–25
(2004) Innate antiviral responses by means of TLR7-mediated
32. Kurt-Jones EA, Popova L, Kwinn L, Haynes LM, Jones LP,
recognition of single-stranded RNA. Science 303:1529–1531
Tripp RA, Walsh EE, Freeman MW, Golenbock DT, Anderson
49. Heil F, Hemmi H, Hochrein H, Ampenberger F, Kirschning C,
LJ, Finberg RW (2000) Pattern recognition receptors TLR4 and
Akira S, Lipford G, Wagner H, Bauer S (2004) Species-specific
CD14 mediate response to respiratory syncytial virus. Nat
recognition of single-stranded RNA via toll-like receptor 7 and 8.
Immunol 1:398–401
Science 303:1526–1529
33. Haynes LM, Moore DD, Kurt-Jones EA, Finberg RW,
Anderson LJ, Tripp RA (2001) Involvement of toll-like 50. Horng T, Barton GM, Flavell RA, Medzhitov R (2002) The
receptor 4 in innate immunity to respiratory syncytial virus. adaptor molecule TIRAP provides signalling specificity for
J Virol 75:10730–10737 Toll-like receptors. Nature 420:329–333
34. Rassa JC, Meyers JL, Zhang Y, Kudaravalli R, Ross SR (2002) 51. Yamamoto M, Sato S, Hemmi H, Sanjo H, Uematsu S,
Murine retroviruses activate B cells via interaction with toll-like Kaisho T, Hoshino K, Takeuchi O, Kobayashi M, Fujita T,
receptor 4. Proc Natl Acad Sci USA 99:2281–2286 Takeda K, Akira S (2002) Essential role for TIRAP in
35. Bieback K, Lien E, Klagge IM, Avota E, Schneider-Schaulies activation of the signalling cascade shared by TLR2 and
J, Duprex WP, Wagner H, Kirschning CJ, Ter Meulen V, TLR4. Nature 420:324–329
Schneider-Schaulies S (2002) Hemagglutinin protein of wild- 52. Kawai T, Adachi O, Ogawa T, Takeda K, Akira S (1999)
type measles virus activates toll-like receptor 2 signaling. J Unresponsiveness of MyD88-deficient mice to endotoxin.
Virol 76:8729–8736 Immunity 11:115–122
36. Compton T, Kurt-Jones EA, Boehme KW, Belko J, Latz E, 53. Yamamoto M, Sato S, Mori K, Hoshino K, Takeuchi O, Takeda
Golenbock DT, Finberg RW (2003) Human cytomegalovirus K, Akira S (2002) Cutting edge: a novel Toll/IL-1 receptor
activates inflammatory cytokine responses via CD14 and domain-containing adapter that preferentially activates the IFN-
Toll-like receptor 2. J Virol 77:4588–4596 beta promoter in the Toll-like receptor signaling. J Immunol
37. Kurt-Jones EA, Chan M, Zhou S, Wang J, Reed G, Bronson R, 169:6668–6672
Arnold MM, Knipe DM, Finberg RW (2004) Herpes simplex 54. Oshiumi H, Matsumoto M, Funami K, Akazawa T, Seya T
virus 1 interaction with Toll-like receptor 2 contributes to lethal (2003) TICAM-1, an adaptor molecule that participates in Toll-
encephalitis. Proc Natl Acad Sci USA 101:1315–1320 like receptor 3-mediated interferon-beta induction. Nat Immunol
38. Alexopoulou L, Holt AC, Medzhitov R, Flavell RA (2001) 4:161–167
Recognition of double-stranded RNA and activation of 55. Hoebe K, Du X, Georgel P, Janssen E, Tabeta K, Kim SO,
NF-kappaB by Toll-like receptor 3. Nature 413:732–738 Goode J, Lin P, Mann N, Mudd S, Crozat K, Sovath S, Han J,
J Mol Med (2006) 84:712–725 723

Beutler B (2003) Identification of Lps2 as a key transducer of 71. Honda K, Yanai H, Mizutani T, Negishi H, Shimada N, Suzuki
MyD88-independent TIR signalling. Nature 424:743–748 N, Ohba Y, Takaoka A, Yeh WC, Taniguchi T (2004) Role of a
56. Yamamoto M, Sato S, Hemmi H, Uematsu S, Hoshino K, Kaisho transductional-transcriptional processor complex involving
T, Takeuchi O, Takeda K, Akira S (2003) TRAM is specifically MyD88 and IRF-7 in Toll-like receptor signaling. Proc Natl
involved in the Toll-like receptor 4-mediated MyD88-indepen- Acad Sci USA 101:15416–15421
dent signaling pathway. Nat Immunol 4:1144–1150 72. Yang K, Puel A, Zhang S, Eidenschenk C, Ku CL, Casrouge A,
57. Bin LH, Xu LG, Shu HB (2003) TIRP, a novel Toll/interleukin-1 Picard C, von Bernuth H, Senechal B, Plancoulaine S, Al-Hajjar
receptor (TIR) domain-containing adapter protein involved in S, Al-Ghonaium A, Marodi L, Davidson D, Speert D, Roifman
TIR signaling. J Biol Chem 278:24526–24532 C, Garty BZ, Ozinsky A, Barrat FJ, Coffman RL, Miller RL, Li
58. Oshiumi H, Sasai M, Shida K, Fujita T, Matsumoto M, Seya T X, Lebon P, Rodriguez-Gallego C, Chapel H, Geissmann F,
(2003) TIR-containing adapter molecule (TICAM)-2, a bridging Jouanguy E, Casanova JL (2005) Human TLR-7-, -8-, and -9-
adapter recruiting to toll-like receptor 4 TICAM-1 that induces mediated induction of IFN-alpha/beta and -lambda Is IRAK-4
interferon-beta. J Biol Chem 278:49751–49762 dependent and redundant for protective immunity to viruses.
59. Sharma S, tenOever BR, Grandvaux N, Zhou GP, Lin R, Hiscott Immunity 23:465–478
J (2003) Triggering the interferon antiviral response through an 73. Uematsu S, Sato S, Yamamoto M, Hirotani T, Kato H, Takeshita
IKK-related pathway. Science 300:1148–1151 F, Matsuda M, Coban C, Ishii KJ, Kawai T, Takeuchi O, Akira S
60. Fitzgerald KA, McWhirter SM, Faia KL, Rowe DC, Latz E, (2005) Interleukin-1 receptor-associated kinase-1 plays an
Golenbock DT, Coyle AJ, Liao SM, Maniatis T (2003) essential role for Toll-like receptor (TLR)7- and TLR9-mediated
IKKepsilon and TBK1 are essential components of the IRF3 interferon-{alpha} induction. J Exp Med 201:915–923
signaling pathway. Nat Immunol 4:491–496 74. Tsujimura H, Tamura T, Kong HJ, Nishiyama A, Ishii KJ,
61. McWhirter SM, Fitzgerald KA, Rosains J, Rowe DC, Golenbock Klinman, DM, Ozato K (2004) Toll-like receptor 9 signaling
DT, Maniatis T (2004) IFN-regulatory factor 3-dependent gene activates NF-kappaB through IFN regulatory factor-8/IFN
expression is defective in Tbk1-deficient mouse embryonic consensus sequence binding protein in dendritic cells. J Immunol
fibroblasts. Proc Natl Acad Sci USA 101:233–238 172:6820–6827
62. Perry AK, Chow EK, Goodnough JB, Yeh WC, Cheng G (2004) 75. Ting JP, Kastner DL, Hoffman HM (2006) CATERPILLERs,
Differential requirement for TANK-binding kinase-1 in type I pyrin and hereditary immunological disorders. Nat Rev Immunol
interferon responses to toll-like receptor activation and viral 6:183–195
infection. J Exp Med 199:1651–1658 76. Ogura Y, Bonen DK, Inohara N, Nicolae DL, Chen FF, Ramos
63. Hemmi H, Takeuchi O, Sato S, Yamamoto M, Kaisho T, Sanjo R, Britton H, Moran T, Karaliuskas R, Duerr RH, Achkar JP,
H, Kawai T, Hoshino K, Takeda K, Akira S (2004) The roles of Brant SR, Bayless TM, Kirschner BS, Hanauer SB, Nunez G,
two IkappaB Kinase-related kinases in lipopolysaccharide and Cho JH (2001) A frameshift mutation in NOD2 associated with
double stranded RNA signaling and viral infection. J Exp Med susceptibility to Crohn’s disease. Nature 411:603–606
199:1641–1650 77. Chamaillard M, Hashimoto M, Horie Y, Masumoto J, Qiu S,
64. Sato S, Sugiyama M, Yamamoto M, Watanabe Y, Kawai T, Saab L, Ogura Y, Kawasaki A, Fukase K, Kusumoto S, Valvano
Takeda K, Akira S (2003) Toll/IL-1 receptor domain-containing MA, Foster SJ, Mak TW, Nunez G, Inohara N (2003) An
adaptor inducing IFN-beta (TRIF) associates with TNF receptor- essential role for NOD1 in host recognition of bacterial
associated factor 6 and TANK-binding kinase 1, and activates peptidoglycan containing diaminopimelic acid. Nat Immunol
two distinct transcription factors, NF-kappa B and IFN-regula- 4:702–707
tory factor-3, in the Toll-like receptor signaling. J Immunol 78. Inohara N, Ogura Y, Fontalba A, Gutierrez O, Pons F, Crespo
171:4304–4310 J, Fukase K, Inamura S, Kusumoto S, Hashimoto M, Foster
65. Kawai T, Takeuchi O, Fujita T, Inoue J, Muhlradt PF, Sato S, SJ, Moran AP, Fernandez-Luna JL, Nunez G (2003) Host
Hoshino K, Akira S (2001) Lipopolysaccharide stimulates the recognition of bacterial muramyl dipeptide mediated through
MyD88-independent pathway and results in activation of IFN- NOD2. Implications for Crohn’s disease. J Biol Chem 278:
regulatory factor 3 and the expression of a subset of lipopoly- 5509–5512
saccharide-inducible genes. J Immunol 167:5887–5894 79. Girardin SE, Boneca IG, Viala J, Chamaillard M, Labigne A,
66. Gohda J, Matsumura T, Inoue J (2004) Cutting edge: TNFR- Thomas G, Philpott DJ, Sansonetti PJ (2003) Nod2 is a general
associated factor (TRAF) 6 is essential for MyD88-dependent sensor of peptidoglycan through muramyl dipeptide (MDP)
pathway but not toll/IL-1 receptor domain-containing adaptor- detection. J Biol Chem 278:8869–8872
inducing IFN-beta (TRIF)-dependent pathway in TLR signaling. 80. Kobayashi K, Inohara, N, Hernandez LD, Galan JE, Nunez G,
J Immunol 173:2913–2917 Janeway CA, Medzhitov R, Flavell RA (2002) RICK/Rip2/
67. Meylan E, Burns K, Hofmann K, Blancheteau V, Martinon F, CARDIAK mediates signalling for receptors of the innate and
Kelliher M, Tschopp J (2004) RIP1 is an essential mediator of adaptive immune systems. Nature 416:194–199
Toll-like receptor 3-induced NF-kappaB activation. Nat Immunol 81. Abbott DW, Wilkins A, Asara JM, Cantley LC (2004) The
5:503–507 Crohn’s disease protein, NOD2, requires RIP2 in order to induce
68. Liu YJ (2005) IPC: professional type 1 interferon-producing ubiquitinylation of a novel site on NEMO. Curr Biol 14:
cells and plasmacytoid dendritic cell precursors. Annu Rev 2217–2227
Immunol 23:275–306 82. Zhou H, Wertz I, O’Rourke K, Ultsch M, Seshagiri S, Eby M,
69. Hemmi H, Kaisho T, Takeda K, Akira S (2003) The roles of Xiao W, Dixit VM (2004) Bcl10 activates the NF-kappaB
Toll-like receptor 9, MyD88, and DNA-dependent protein kinase pathway through ubiquitination of NEMO. Nature 427:167–171
catalytic subunit in the effects of two distinct CpG DNAs on 83. Damiano JS, Stehlik C, Pio F, Godzik A, Reed JC (2001) CLAN,
dendritic cell subsets. J Immunol 170:3059–3064 a novel human CED-4-like gene. Genomics 75:77–83
70. Kawai T, Sato S, Ishii KJ, Coban C, Hemmi H, Yamamoto M, 84. Damiano JS, Oliveira V, Welsh K, Reed JC (2004) Heterotypic
Terai K, Matsuda M, Inoue J, Uematsu S, Takeuchi O, Akira S interactions among NACHT domains: implications for regulation
(2004) Interferon-alpha induction through Toll-like receptors of innate immune responses. Biochem J 381:213–219
involves a direct interaction of IRF7 with MyD88 and TRAF6. 85. Mariathasan S, Newton K, Monack DM, Vucic D, French DM,
Nat Immunol 5:1061–1068 Lee WP, Roose-Girma M, Erickson S, Dixit VM (2004)
724 J Mol Med (2006) 84:712–725

Differential activation of the inflammasome by caspase-1 hereditary MHC class II deficiency (or bare lymphocyte
adaptors ASC and Ipaf. Nature 430:213–218 syndrome). Cell 75:135–146
86. Stehlik C, Hayashi H, Pio F, Godzik A, Reed JC (2003) CARD6 101. Kawai T, Akira S (2006) Innate immune recognition of viral
is a modulator of NF-kappa B activation by Nod1- and Cardiak- infection. Nat Immunol 7:131–137
mediated pathways. J Biol Chem 278:31941–31949 102. Kang DC, Gopalkrishnan RV, Wu Q, Jankowsky E, Pyle AM,
87. Martinon F, Burns K, Tschopp J (2002) The inflammasome: Fisher PB (2002) mda-5: An interferon-inducible putative RNA
a molecular platform triggering activation of inflammatory helicase with double-stranded RNA-dependent ATPase activity
caspases and processing of proIL-beta. Mol Cell 10:417–426 and melanoma growth-suppressive properties. Proc Natl Acad
88. Petrilli V, Papin S, Tschopp J (2005) The inflammasome. Curr Sci USA 99:637–642
Biol 15:R581 103. Kovacsovics M, Martinon F, Micheau O, Bodmer JL, Hofmann
89. Martinon F, Tschopp J (2004) Inflammatory caspases: linking an K, Tschopp J (2002) Overexpression of Helicard, a CARD-
intracellular innate immune system to autoinflammatory dis- containing helicase cleaved during apoptosis, accelerates DNA
eases. Cell 117:561–574 degradation. Curr Biol 12:838–843
90. Kanneganti TD, Ozoren N, Body-Malapel M, Amer A, Park 104. Andrejeva J, Childs KS, Young DF, Carlos TS, Stock N,
JH, Franchi L, Whitfield J, Barchet W, Colonna M, Goodbourn S, Randall RE (2004) The V proteins of
Vandenabeele P, Bertin J, Coyle A, Grant EP, Akira S, Nunez paramyxoviruses bind the IFN-inducible RNA helicase,
G (2006) Bacterial RNA and small antiviral compounds mda-5, and inhibit its activation of the IFN-beta promoter.
activate caspase-1 through cryopyrin/Nalp3. Nature 440:233– Proc Natl Acad Sci USA 101:17264–17269
236 105. Yoneyama M, Kikuchi M, Matsumoto K, Imaizumi T, Miyagishi
91. Mariathasan S, Weiss DS, Newton K, McBride J, O’Rourke K, M, Taira K, Foy E, Loo YM, Gale M Jr, Akira S, Yonehara S,
Roose-Girma M, Lee WP, Weinrauch Y, Monack DM, Dixit VM Kato A, Fujita T (2005) Shared and unique functions of the
(2006) Cryopyrin activates the inflammasome in response to DExD/H-box helicases RIG-I, MDA5, and LGP2 in antiviral
toxins and ATP. Nature 440:228–232 innate immunity. J Immunol 175:2851–2858
92. Martinon F, Petrilli V, Mayor A, Tardivel A, Tschopp J 106. Rothenfusser S, Goutagny N, DiPerna G, Gong M, Monks BG,
(2006) Gout-associated uric acid crystals activate the NALP3 Schoenemeyer A, Yamamoto M, Akira S, Fitzgerald KA (2005)
inflammasome. Nature 440:237–241 The RNA helicase Lgp2 inhibits TLR-independent sensing of
93. Diez E, Lee SH, Gauthier S, Yaraghi Z, Tremblay M, Vidal S, viral replication by retinoic acid-inducible gene-I. J Immunol
Gros P (2003) Birc1e is the gene within the Lgn1 locus 175:5260–5268
associated with resistance to Legionella pneumophila. Nat Genet 107. Kawai T, Takahashi K, Sato S, Coban C, Kumar H, Kato H, Ishii
33:55–60 KJ, Takeuchi O, Akira S (2005) IPS-1, an adaptor triggering
94. Zamboni DS, Kobayashi KS, Kohlsdorf T, Ogura Y, Long RIG-I- and Mda5-mediated type I interferon induction. Nat
EM, Vance RE, Kuida K, Mariathasan S, Dixit VM, Flavell Immunol 6:981–988
RA, Dietrich WF, Roy CR (2006) The Birc1e cytosolic 108. Seth RB, Sun L, Ea CK, Chen ZJ (2005) Identification and
pattern-recognition receptor contributes to the detection and characterization of MAVS, a mitochondrial antiviral signaling
control of Legionella pneumophila infection. Nat Immunol protein that activates NF-kappaB and IRF 3. Cell 122:669–682
7:318–325 109. Xu LG, Wang YY, Han KJ, Li LY, Zhai Z, Shu HB (2005) VISA
95. Miceli-Richard C, Lesage S, Rybojad M, Prieur AM, Manouvrier- is an adapter protein required for virus-triggered IFN-beta
Hanu S, Hafner R, Chamaillard M, Zouali H, Thomas G, Hugot signaling. Mol Cell 19:727–740
JP (2001) CARD15 mutations in Blau syndrome. Nat Genet 110. Meylan E, Curran J, Hofmann K, Moradpour D, Binder M,
29:19–20 Bartenschlager R, Tschopp J (2005) Cardif is an adaptor protein
96. Hoffman HM, Mueller JL, Broide DH, Wanderer AA, Kolodner in the RIG-I antiviral pathway and is targeted by hepatitis C
RD (2001) Mutation of a new gene encoding a putative virus. Nature 437:1167–1172
pyrin-like protein causes familial cold autoinflammatory 111. Bowie A, Kiss-Toth E, Symons JA, Smith GL, Dower SK,
syndrome and Muckle–Wells syndrome. Nat Genet 29: O’Neill LA (2000) A46R and A52R from vaccinia virus are
301–305 antagonists of host IL-1 and toll-like receptor signaling. Proc
97. Aksentijevich I, Nowak M, Mallah M, Chae JJ, Watford WT, Natl Acad Sci USA 97:10162–10167
Hofmann SR, Stein L, Russo R, Goldsmith D, Dent P, 112. Harte MT, Haga IR, Maloney G, Gray P, Reading PC, Bartlett
Rosenberg HF, Austin F, Remmers EF, Balow JE Jr, Rosenzweig NW, Smith GL, Bowie A, O’Neill LA (2003) The poxvirus
S, Komarow H, Shoham NG, Wood G, Jones J, Mangra N, protein A52R targets Toll-like receptor signaling complexes to
Carrero H, Adams BS, Moore TL, Schikler K, Hoffman H, suppress host defense. J Exp Med 197:343–351
Lovell DJ, Lipnick R, Barron K, O’Shea JJ, Kastner DL, 113. Stack J, Haga IR, Schroder M, Bartlett NW, Maloney G,
Goldbach-Mansky R (2002) De novo CIAS1 mutations, cytokine Reading PC, Fitzgerald KA, Smith GL, Bowie AG (2005)
activation, and evidence for genetic heterogeneity in patients Vaccinia virus protein A46R targets multiple Toll-like-interleu-
with neonatal-onset multisystem inflammatory disease kin-1 receptor adaptors and contributes to virulence. J Exp Med
(NOMID): a new member of the expanding family of 201:1007–1018
pyrin-associated autoinflammatory diseases. Arthritis Rheum 114. Foy E, Li K, Sumpter R Jr, Loo YM, Johnson CL, Wang C, Fish
46:3340–3348 PM, Yoneyama M, Fujita T, Lemon SM, Gale M Jr (2005)
98. Harton JA, Linhoff MW, Zhang J, Ting JP (2002) Cutting edge: Control of antiviral defenses through hepatitis C virus disruption
CATERPILLER: a large family of mammalian genes containing of retinoic acid-inducible gene-I signaling. Proc Natl Acad Sci
CARD, pyrin, nucleotide-binding, and leucine-rich repeat USA 102:2986–2991
domains. J Immunol 169:4088–4093 115. Li XD, Sun L, Seth RB, Pineda G, Chen ZJ (2005) Hepatitis C
99. Ting JP, Davis BK (2005) CATERPILLER: a novel gene family virus protease NS3/4A cleaves mitochondrial antiviral signaling
important in immunity, cell death, and diseases. Annu Rev protein off the mitochondria to evade innate immunity. Proc Natl
Immunol 23:387–414 Acad Sci USA 102:17717–17722
100. Steimle V, Otten LA, Zufferey M, Mach B (1993) Complemen- 116. Kawane K, Fukuyama H, Yoshida H, Nagase H, Ohsawa Y,
tation cloning of an MHC class II transactivator mutated in Uchiyama Y, Okada K, Iida T, Nagata S (2003) Impaired thymic
J Mol Med (2006) 84:712–725 725

development in mouse embryos deficient in apoptotic DNA 119. Ishii KJ, Suzuki K, Coban C, Takeshita F, Itoh Y, Matoba H,
degradation. Nat Immunol 4:138–144 Kohn LD, Klinman DM (2001) Genomic DNA released by
117. Yoshida H, Okabe Y, Kawane K, Fukuyama H, Nagata S (2005) dying cells induces the maturation of APCs. J Immunol
Lethal anemia caused by interferon-beta produced in mouse 167:2602–2607
embryos carrying undigested DNA. Nat Immunol 6:49–56 120. Ishii KJ, Coban C, Kato H, Takahashi K, Torii Y, Takeshita F,
118. Suzuki K, Mori A, Ishii KJ, Saito J, Singer DS, Klinman DM, Ludwig H, Sutter G, Suzuki K, Hemmi H, Sato S, Yamamoto M,
Krause PR, Kohn LD (1999) Activation of target-tissue immune- Uematsu S, Kawai T, Takeuchi O, Akira S (2006) A Toll-like
recognition molecules by double-stranded polynucleotides. Proc receptor-independent antiviral response induced by double-
Natl Acad Sci USA 96:2285–2290 stranded B-form DNA. Nat Immunol 7:40–48

You might also like