16
Biochem. J. (2009) 420, 1–16 (Printed in Great Britain) doi:10.1042/BJ20090272 1 REVIEW ARTICLE Pathogen recognition in the innate immune response Himanshu KUMAR*, Taro KAWAI*and Shizuo AKIRA*1 *Laboratory of Host Defense, WPI Immunology Frontier Research Center, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan, and Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan Immunity against microbial pathogens primarily depends on the recognition of pathogen components by innate receptors expressed on immune and non-immune cells. Innate receptors are evolutionarily conserved germ-line-encoded proteins and include TLRs (Toll-like receptors), RLRs [RIG-I (retinoic acid-inducible gene-I)-like receptors] and NLRs (Nod-like receptors). These re- ceptors recognize pathogens or pathogen-derived products in different cellular compartments, such as the plasma membrane, the endosomes or the cytoplasm, and induce the expression of cytokines, chemokines and co-stimulatory molecules to eliminate pathogens and instruct pathogen-specific adaptive immune responses. In the present review, we will discuss the recent progress in the study of pathogen recognition by TLRs, RLRs and NLRs and their signalling pathways. Key words: innate immune response, Nod-like receptor (NLR), pathogen recognition, retinoic acid-inducible gene-I receptor (RIG-I-like receptor, RLR), Toll-like receptor (TLR). INTRODUCTION The innate immune system is the first line of the defence system against microbial pathogens such as Gram-positive and Gram- negative bacteria, fungi and viruses. Innate immune cells such as macrophages and DCs (dendritic cells) directly kill the pathogenic micro-organism through phagocytosis or induce the produc- tion of cytokines, which aid elimination of the pathogens [1– 4]. The responses of the innate immune system instruct the development of long-lasting pathogen-specific adaptive immune responses. The adaptive immune system consists of B- and T-cells, which provide pathogen specific immunity to the host through somatic rearrangement of antigen receptor genes. B- cells produce pathogen-specific antibodies to neutralize toxins produced by pathogens, whereas T-cells provide the cytokine milieu to clear pathogen-infected cells through their cytotoxic effects or via signals to B-cells [5]. The mechanisms for innate immune recognition of pathogens and signalling have received increasing research attention. These studies were initiated after the discovery of Toll protein, which plays an important role in the defence against fungal infection in Drosophila (fruitfly) [6]. Further studies led to the discovery Abbreviations used: AIM2, absent in melanoma 2; alum, aluminium salts; AP-1, activator protein-1; ASC, apoptosis-associated speck-like protein containing a CARD (caspase recruitment domain); Atg, autophagy-related; Atg16L1, autophagy-related 16-like 1; Bcl, B-cell lymphoma; BIR, baculovirus inhibitor of apoptosis protein repeat; Birc1, BIR-containing 1; CARD, caspase recruitment domain; CARDIAK, CARD-containing IL (interleukin)-1β converting enzyme-associated kinase; Cardif, CARD adaptor inducing IFN-β; CARDINAL, CARD i nhibitor of nuclear factor-κB-activating ligands; cDC, conventional dendritic cell; CIAS, cold-induced autoinflammatory syndrome 1; CLAN, CARD LRR- and NACHT-domain-containing protein; CLR, C-type lectin receptor; CTD, C-terminal domain; DAI, DNA-dependent activator of IRFs; DAK, dihydroacetone kinase; DC, dendritic cell; DED, death effector domain; ds, double-stranded; DUBA, de-ubiquitinating enzyme A; DV, dengue virus; EDA, extracellular domain A; EMCV, encephalomyocarditis virus; ER, endoplasmic reticulum; ERK, extracellular-signal-regulated kinase; FADD, Fas-associated death-domain; GPI, glycosylphosphatidylinositol; HCV, hepatitis C virus; HIN, haematopoietic interferon-inducible; HSE, herpes simplex virus encephalitis; HSV, herpes simplex virus; iE-DAP, γ-D-glutamyl-m- diaminopimelic acid; IFI, interferon-inducible, IFN, interferon; IκB, inhibitor of κB; IKK, IκB kinase; IL, interleukin; iNOS, inducible NO synthase; IPAF, IL-1β converting enzyme protease activating factor; IPS-1, IFN-β promoter stimulator-1; IRAK, IL-1 receptor-associated kinase; IRF, IFN regulatory factor; ISRE, IFN-stimulated response element; IV, influenza virus; JEV, Japanese-encephalitis virus; JNK, c-jun N-terminal kinase; LBP, LPS-binding protein; LCMV, lymphocytic choriomeningitis virus; Lgp2, Laboratory of Genetics and Physiology 2; LPDC, lamina propria DC; LPS, lipopolysaccharide; LRR, leucine-rich repeat; LT, lethal toxin; LTA, lipoteichoic acid; MAL, MyD88-adaptor-like; MALP-2, macrophage-activating lipopeptide; MAPK, mitogen- activated protein kinase; MAVS, mitochondrial antiviral signalling; MCMV, murine cytomegalovirus; MD2, myeloid differentiation protein-2; Mda5, melanoma differentiation-associated gene 5; MDP, muramyl dipeptide; MITA, mediator of IRF3 activation; MMTV, mouse mammary tumour virus; MSU, monosodium urate; mTOR, mammalian target of rapamycin; MyD88, myeloid differentiation primary response gene 88; NACHT, NTPase-domain named after NAIP, CIITA, HET-E and TP1; NAIP, NLR family; apoptosis inhibitory protein; NAIP5, neuronal apoptosis inhibitor protein 5; NAK, NF-κB activating kinase; NALP, NACHT/LRR/PYD-containing protein; NAP, NAK-associated protein; NDV, Newcastle-disease virus; NEMO, NF-κB essential modifier; NF-κB, nuclear factor κB; NK, natural killer; NLR, Nod-like receptor; NLRC, NLR family CARD-domain-containing; NLRP, NLR family pyrin-domain-containing; NOD, nucleotide- binding oligomerization domain; ODN, oligodeoxynucleotide; PAMP, pathogen-associated molecular pattern; pDCs, plasmacytoid dendritic cells; PGN, peptidoglycan; Phox, phagocyte oxidase; PKR, protein kinase receptor; PRR, pattern-recognition receptor; PYCARD, PYD- and CARD-domain-containing; PYD, pyrin domain; PYHIN, PYD and HIN domain family member 1; PYPAF, PYD-containing APAF-1 (apoptotic peptidase activating factor 1)-like protein; RD, repressor domain; RICK, RIP-like interacting caspase-like apoptosis-regulatory protein kinase; RIG-I, retinoic acid-inducible gene-I; RIP, receptor- interacting protein; RLR, RIG-I-like receptor; RNF, ring finger; ROS, reactive oxygen species; RSV, respiratory syncytial virus; SeV, Sendai virus; SINTBAD, similar to NAP1 TBK1 adaptor; SNP, single-nucleotide polymorphism; ss, single-stranded; STING, stimulator of interferon genes; T2K, TRAF2-associated kinase; TAB, TAK1-binding protein; TAK1, transforming-growth-factor-β-activated kinase 1; TANK, TRAF family member associated NF-κB activator; TBK1, TANK-binding kinase 1; Th, T-helper; TICAM, TIR-domain-containing adaptor molecule; TIR, Toll/IL-1 receptor; TIRAP, TIR-containing adaptor protein; TLR, Toll-like receptor; TNF, tumour necrosis factor; TNFR, TNF receptor; TRADD, TNF-receptor-associated DD; TRAF, TNF-receptor-associated factor; TRAM, TRIF-related adaptor molecule; TRIF, TIR-containing adaptor inducing IFN-β; TRIM, tripartite motif; UBC, ubiquitin C; UEV, ubiquitin-conjugating enzyme variant; VISA, virus-induced signalling adaptor; VSV, vesicular-stomatitis virus; WNV, West Nile virus; WT, wild-type. 1 To whom correspondence should be addressed (email [email protected]). c The Authors Journal compilation c 2009 Biochemical Society

REVIEW ARTICLE Pathogen recognition in the innate immune ... · Key words: innate immune response, Nod-like receptor (NLR), pathogen recognition, retinoic acid-inducible gene-I receptor

  • Upload
    others

  • View
    5

  • Download
    0

Embed Size (px)

Citation preview

Page 1: REVIEW ARTICLE Pathogen recognition in the innate immune ... · Key words: innate immune response, Nod-like receptor (NLR), pathogen recognition, retinoic acid-inducible gene-I receptor

Biochem. J. (2009) 420, 1–16 (Printed in Great Britain) doi:10.1042/BJ20090272 1

REVIEW ARTICLEPathogen recognition in the innate immune responseHimanshu KUMAR*†, Taro KAWAI*† and Shizuo AKIRA*†1

*Laboratory of Host Defense, WPI Immunology Frontier Research Center, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan, and †Department of Host Defense,Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan

Immunity against microbial pathogens primarily depends onthe recognition of pathogen components by innate receptorsexpressed on immune and non-immune cells. Innate receptors areevolutionarily conserved germ-line-encoded proteins and includeTLRs (Toll-like receptors), RLRs [RIG-I (retinoic acid-induciblegene-I)-like receptors] and NLRs (Nod-like receptors). These re-ceptors recognize pathogens or pathogen-derived products indifferent cellular compartments, such as the plasma membrane,the endosomes or the cytoplasm, and induce the expression

of cytokines, chemokines and co-stimulatory molecules toeliminate pathogens and instruct pathogen-specific adaptiveimmune responses. In the present review, we will discuss therecent progress in the study of pathogen recognition by TLRs,RLRs and NLRs and their signalling pathways.

Key words: innate immune response, Nod-like receptor (NLR),pathogen recognition, retinoic acid-inducible gene-I receptor(RIG-I-like receptor, RLR), Toll-like receptor (TLR).

INTRODUCTION

The innate immune system is the first line of the defence systemagainst microbial pathogens such as Gram-positive and Gram-negative bacteria, fungi and viruses. Innate immune cells such asmacrophages and DCs (dendritic cells) directly kill the pathogenicmicro-organism through phagocytosis or induce the produc-tion of cytokines, which aid elimination of the pathogens [1–4]. The responses of the innate immune system instruct thedevelopment of long-lasting pathogen-specific adaptive immuneresponses. The adaptive immune system consists of B- and

T-cells, which provide pathogen specific immunity to the hostthrough somatic rearrangement of antigen receptor genes. B-cells produce pathogen-specific antibodies to neutralize toxinsproduced by pathogens, whereas T-cells provide the cytokinemilieu to clear pathogen-infected cells through their cytotoxiceffects or via signals to B-cells [5].

The mechanisms for innate immune recognition of pathogensand signalling have received increasing research attention. Thesestudies were initiated after the discovery of Toll protein, whichplays an important role in the defence against fungal infectionin Drosophila (fruitfly) [6]. Further studies led to the discovery

Abbreviations used: AIM2, absent in melanoma 2; alum, aluminium salts; AP-1, activator protein-1; ASC, apoptosis-associated speck-like proteincontaining a CARD (caspase recruitment domain); Atg, autophagy-related; Atg16L1, autophagy-related 16-like 1; Bcl, B-cell lymphoma; BIR, baculovirusinhibitor of apoptosis protein repeat; Birc1, BIR-containing 1; CARD, caspase recruitment domain; CARDIAK, CARD-containing IL (interleukin)-1β

converting enzyme-associated kinase; Cardif, CARD adaptor inducing IFN-β; CARDINAL, CARD inhibitor of nuclear factor-κB-activating ligands; cDC,conventional dendritic cell; CIAS, cold-induced autoinflammatory syndrome 1; CLAN, CARD LRR- and NACHT-domain-containing protein; CLR, C-typelectin receptor; CTD, C-terminal domain; DAI, DNA-dependent activator of IRFs; DAK, dihydroacetone kinase; DC, dendritic cell; DED, death effectordomain; ds, double-stranded; DUBA, de-ubiquitinating enzyme A; DV, dengue virus; EDA, extracellular domain A; EMCV, encephalomyocarditis virus;ER, endoplasmic reticulum; ERK, extracellular-signal-regulated kinase; FADD, Fas-associated death-domain; GPI, glycosylphosphatidylinositol; HCV,hepatitis C virus; HIN, haematopoietic interferon-inducible; HSE, herpes simplex virus encephalitis; HSV, herpes simplex virus; iE-DAP, γ-D-glutamyl-m-diaminopimelic acid; IFI, interferon-inducible, IFN, interferon; IκB, inhibitor of κB; IKK, IκB kinase; IL, interleukin; iNOS, inducible NO synthase; IPAF,IL-1β converting enzyme protease activating factor; IPS-1, IFN-β promoter stimulator-1; IRAK, IL-1 receptor-associated kinase; IRF, IFN regulatoryfactor; ISRE, IFN-stimulated response element; IV, influenza virus; JEV, Japanese-encephalitis virus; JNK, c-jun N-terminal kinase; LBP, LPS-bindingprotein; LCMV, lymphocytic choriomeningitis virus; Lgp2, Laboratory of Genetics and Physiology 2; LPDC, lamina propria DC; LPS, lipopolysaccharide;LRR, leucine-rich repeat; LT, lethal toxin; LTA, lipoteichoic acid; MAL, MyD88-adaptor-like; MALP-2, macrophage-activating lipopeptide; MAPK, mitogen-activated protein kinase; MAVS, mitochondrial antiviral signalling; MCMV, murine cytomegalovirus; MD2, myeloid differentiation protein-2; Mda5, melanomadifferentiation-associated gene 5; MDP, muramyl dipeptide; MITA, mediator of IRF3 activation; MMTV, mouse mammary tumour virus; MSU, monosodiumurate; mTOR, mammalian target of rapamycin; MyD88, myeloid differentiation primary response gene 88; NACHT, NTPase-domain named after NAIP,CIITA, HET-E and TP1; NAIP, NLR family; apoptosis inhibitory protein; NAIP5, neuronal apoptosis inhibitor protein 5; NAK, NF-κB activating kinase; NALP,NACHT/LRR/PYD-containing protein; NAP, NAK-associated protein; NDV, Newcastle-disease virus; NEMO, NF-κB essential modifier; NF-κB, nuclear factorκB; NK, natural killer; NLR, Nod-like receptor; NLRC, NLR family CARD-domain-containing; NLRP, NLR family pyrin-domain-containing; NOD, nucleotide-binding oligomerization domain; ODN, oligodeoxynucleotide; PAMP, pathogen-associated molecular pattern; pDCs, plasmacytoid dendritic cells; PGN,peptidoglycan; Phox, phagocyte oxidase; PKR, protein kinase receptor; PRR, pattern-recognition receptor; PYCARD, PYD- and CARD-domain-containing;PYD, pyrin domain; PYHIN, PYD and HIN domain family member 1; PYPAF, PYD-containing APAF-1 (apoptotic peptidase activating factor 1)-like protein;RD, repressor domain; RICK, RIP-like interacting caspase-like apoptosis-regulatory protein kinase; RIG-I, retinoic acid-inducible gene-I; RIP, receptor-interacting protein; RLR, RIG-I-like receptor; RNF, ring finger; ROS, reactive oxygen species; RSV, respiratory syncytial virus; SeV, Sendai virus; SINTBAD,similar to NAP1 TBK1 adaptor; SNP, single-nucleotide polymorphism; ss, single-stranded; STING, stimulator of interferon genes; T2K, TRAF2-associatedkinase; TAB, TAK1-binding protein; TAK1, transforming-growth-factor-β-activated kinase 1; TANK, TRAF family member associated NF-κB activator; TBK1,TANK-binding kinase 1; Th, T-helper; TICAM, TIR-domain-containing adaptor molecule; TIR, Toll/IL-1 receptor; TIRAP, TIR-containing adaptor protein; TLR,Toll-like receptor; TNF, tumour necrosis factor; TNFR, TNF receptor; TRADD, TNF-receptor-associated DD; TRAF, TNF-receptor-associated factor; TRAM,TRIF-related adaptor molecule; TRIF, TIR-containing adaptor inducing IFN-β; TRIM, tripartite motif; UBC, ubiquitin C; UEV, ubiquitin-conjugating enzymevariant; VISA, virus-induced signalling adaptor; VSV, vesicular-stomatitis virus; WNV, West Nile virus; WT, wild-type.

1 To whom correspondence should be addressed (email [email protected]).

c© The Authors Journal compilation c© 2009 Biochemical Society

Page 2: REVIEW ARTICLE Pathogen recognition in the innate immune ... · Key words: innate immune response, Nod-like receptor (NLR), pathogen recognition, retinoic acid-inducible gene-I receptor

2 H. Kumar, T. Kawai and S. Akira

of the TLR (Toll-like receptor) family of proteins in mammals[7–9]. Recently, other proteins families such as RLRs [RIG-I(retinoic acid-inducible gene-I)-like receptors] [1,10] and NLRs(Nod-like receptors) [1,11–13] were discovered. These familiesof receptors are collectively known as PRRs (pattern-recognitionreceptors) [14], which recognize the specific molecular structuresof pathogens known as PAMPs (pathogen-associated molecularpatterns) in various compartments of cells, such as the plasmamembrane, the endolysosome and the cytoplasm.

In the present review we will focus on recent advances in thestudy of recognition and signalling mediated by TLRs, RLRs andNLRs.

TOLL-LIKE RECEPTORS

The Toll protein was originally identified in fruitflies (Drosophila)and is involved in dorsoventral polarity during embryonicdevelopment [6]. Further studies have shown that the Toll proteinplays an essential role in mounting an effective immune responseagainst the fungus Aspergillus fumigatus [6]. These studies ledto the identification of homologues of Toll proteins in humansand mice through database searches and which are referred toas TLRs [7]. To date, 10 and 13 TLR members have beenidentified in humans and mice respectively [1]. The TLRs aretype I membrane glycoproteins and consist of an extracellularLRR (leucine-rich repeat) domain, a transmembrane domain anda cytoplasmic TIR ]Toll/IL (interleukin)-1 receptor] domain [15].The LRR domain of TLRs consists of 16–28 tandem repeatsof the LRR motif [16] and is involved in the recognition ofligands such as protein (e.g. flagellin and porin from bacteria),sugar (e.g. zymosan from fungi), lipid [LPS (lipopolysaccharide),lipid A and LTA (lipoteichoic acid) from bacteria], nucleic acid(CpG-containing DNA from bacteria and viruses and viral RNA),derivatives of protein or peptide (lipoprotein and lipopeptidesfrom various pathogens), derivatives of lipid (lipoarabinomannan)from mycobacteria) and a complex derivative of protein orpeptides, sugar and lipid (diacyl lipopeptides from mycoplasma)[1]. The TIR domain of TLRs consists of approx. 150 aminoacids and shows homology with the cytoplasmic region of theIL-1 receptor. Therefore, it is termed the TIR domain [15,17].The TIR domain interacts with TIR-domain-containing adaptorssuch as MyD88 (myeloid differentiation primary response gene88), TIRAP [TIR-containing adaptor protein, also known as MAL(MyD88-adaptor-like], TRIF [TIR-containing adaptor-inducingIFN (interferon)-β, also known as TICAM1 (TIR-domain-containing adaptor molecule 1] and TRAM (TRIF-related adaptormolecule, also known as TICAM2). In turn, the downstreamsignalling pathways activate MAPKs (mitogen-activated proteinkinases) and transcription factors such as NF-κB (nuclear factorκB) and IRFs (IFN regulatory factors) to induce production ofinflammatory cytokines and type I IFNs.

The TLR family members are expressed on various immune andnon-immune cells such as B-cells, NK (natural killer) cells, DCs,macrophages, fibroblast cells, epithelial cells and endothelialcells. However, TLRs are differentially localized within the cells.TLR1, TLR2, TLR4, TLR5 and TLR6 are expressed on the cellsurface, whereas TLR3, TLR7, TLR8 and TLR9 are expressed inthe endosomes (Figure 1).

Recently, co-crystallization of TLR1–TLR2, TLR3 and TLR4and their ligand-recognition properties has been reported. Theheterodimer or homodimer of the LRR domain of TLR1–TLR2,TLR4 and TLR3 show a horseshoe-like structure or m-shapedframework and consists of both a concave and a convex surface.These surfaces are responsible for ligand binding and ligand-induced TLR dimerization [18–21].

Pathogen recognition by the extracellular cell-surface TLRs

TLR1, TLR2 and TLR6

TLR2 recognizes a structurally diverse range of PAMPs thatinclude proteins such as V-antigen (LcrV) from Yersinia,haemagglutinin protein from measles virus, glycolipids, LTAfrom Staphylococcus aureus and Streptococcus pneumoniae[22], lipopeptides or lipoproteins such as MALP (macrophage-activating lipopeptide)-2 and R-MALP from Mycoplasma species[23–25], lipoproteins from Escherichia coli [26], Borreliaburgdorferi [27], Mycoplasma species [28] and Mycobacteriumtuberculosis [29], peptidoglycan from Staph. aureus, Strep.pneumoniae and Strep. pyogenes [30–32] and polysaccharidesknown as zymosan from Saccharomyces cerevisiae [33,34]. Theligands for TLR2 have been described in detail in a review article[35]. In addition, TLR2 also recognizes complete pathogens,including the species of the bacterium Chlamydia [36], virusessuch as HSV (herpes simplex virus) [37] and varicella-zoster virus[38] (Figure 1). The diversity of ligand recognition by TLR2 ispossible because TLR2 can recognize the ligands in associationwith structurally related TLRs such as TLR1 and TLR6. TheTLR2–TLR1 and TLR2–TLR6 heterodimers recognize triacyllipopeptide derived from Gram-negative bacteria and diacyl lipo-peptide derived from mycoplasma respectively [39,40]. TLR2also recognizes zymosan (β-1,3-glucan and β-1,6-glucan) inassociation with the structurally unrelated C-type lectin familyknown as dectin-1 [41]. Furthermore, the class II scavengerreceptor CD36 has been shown to be involved in phagocytosisand cytokine production in response to Staph. aureus and its cell-wall components such as LTA and MALP-2, suggesting that CD36functions as a co-receptor of TLR2/6 [25].

TLR4

TLR4 recognizes LPS from Gram-negative bacteria [8,9],glycoinositolphospholipids from Trypanosoma [42], the fusionprotein from RSV (respiratory syncytial virus) [43] and theenvelope protein from MMTV (mouse mammary tumour virus)[44]. TLR4 also recognizes diterpene (taxol) purified from thebark of Taxus brevifolia (the Pacific yew) [45,46] (Figure 1). Inaddition, TLR4 directly or indirectly recognizes endogenous mo-lecules such as heat-shock proteins, fibrinogen, hyaluronic acid,β-defensin and extracellular domain A in fibronectin [47]. Therecognition of LPS is triggered by a complex that contains TLR4,a recognition subunit MD2 (myeloid differentiation protein-2) andmembrane-bound GPI (glycosylphosphatidylinositol)-anchoredCD14. The activation of TLR4 is further supported by anotherprotein known as LBP (LPS-binding protein) [48]. The studiesshow that the lipid A (an active component of LPS) binds toMD2 and forms a complex. The lipid A–MD2 complex interactswith TLR4 and activates signalling, suggesting that MD2 is moreimportant in the recognition of lipid A [18].

TLR5

TLR5 recognizes a monomer of flagellin [49], an importantstructural protein of pathogenic and non-pathogenic motilebacteria. It is also important for adhesion and invasion at theluminal surface of the epithelial cells covering the mucosal tissuesduring infection [50]. Flagellin from Salmonella typhimuriumcontains 494 amino acids and consists of two functional domains.The 140 amino acids of the N-terminal domain and 90 amino acidsof the C-terminal domain of this protein are highly conserved andessential for the polymerization and motility of flagellin [51]. Thecentral domain of the protein is highly variable among Salmonella

c© The Authors Journal compilation c© 2009 Biochemical Society

Page 3: REVIEW ARTICLE Pathogen recognition in the innate immune ... · Key words: innate immune response, Nod-like receptor (NLR), pathogen recognition, retinoic acid-inducible gene-I receptor

Pathogen recognition in the innate immune response 3

Figure 1 PAMPs recognized by TLRs and their adaptors

Plasma-membrane-localized TLRs (TLR2, TLR4, TLR5, TLR11 alone and TLR2 in association with TLR1 or TLR6) and endosomally localized TLRs (TLR3, TLR7 and TLR9) recognize the indicatedligands. TLR1, TLR2, TLR4 and TLR6 recruit TIRAP and MyD88. MyD88 also contains the DD. In addition to TIRAP and MyD88, TLR4 recruits TRAM and TRIF. TLR5, TLR7, TLR9 and TLR11 recruitMyD88, whereas TLR3 recruits TRIF.

serovars and bacterial species, and this region is exposed at theouter surface of the flagellum. Amino acids 89–96 are essentialfor TLR5 activation, but this region is located deep inside thetertiary structure of the flagellin protein and becomes accessibleonly when flagellin is present in monomer form [52,53]. However,it is not clear how flagellated bacteria deliver monomeric flagellinunder physiological conditions. TLR5 is mainly expressed on theluminal surface of the epithelial cells covering the mucosal tissuesand trachea, and the bronchi and the alveoli of the respiratory tract[54–58] (Figure 1). Upon activation with flagellin, epithelial cellsinduce cytokines and chemokines, and neutrophil recruitment.Recently, we have shown that TLR5+ small-intestinal LPDCs(lamina propria DCs) are important for the induction of humoraland cellular immunity in the intestine. These LPDCs can induceretinoic acid and are involved in the generation of IgA+ plasmacells, as well as the differentiation of both Th1 (T-helper 17) andTh1 cells in the intestine in a TLR5-dependent manner [59].

TLR11

TLR11 recognizes profilins from Toxoplasma gondii, anobligate intracellular protozoan parasite [60]. It also recognizesuropathogenic E. coli (Figure 1). Mice deficient in TLR11 showincreased susceptibility to these pathogens [61]. TLR11 has been

shown to be expressed on epithelial cells of the bladder in mouse.However, TLR11 is not expressed in humans, as the predictedmRNA has at least one stop codon [62].

Pathogen recognition by intracellular TLRs

TLR3

TLR3 recognizes viral ds (double-stranded) RNA originatingfrom dsRNA viruses such as reovirus [63]. TLR3 also recognizesdsRNA produced during replication of ss (single-stranded) RNAviruses, such as WNV (West Nile virus) [64], RSV [65] andEMCV (encephalomyocarditis virus) [66]. In addition, TLR3recognizes a synthetic analogue of dsRNA known as poly(I-C)(Figure 1). TLR3 is expressed in the endosomes of immune cells,including cDCs (conventional DCs – a type of antigen-presentingcell that induces various cytokines after stimulation with ligands),macrophages, B-cells, NK cells and non-immune cells, includingepithelial cells. However, TLR3 is not expressed on pDCs(plasmacytoid DCs – a type of DC that produces high amounts oftype I IFNs). In addition, TLR3 is highly expressed in the brain[67]. TLR3-deficient mice infected with various RNA viruses suchas MCMV (murine cytomegalovirus), VSV (vesicular-stomatitisvirus), LCMV (lymphocytic choriomeningitis virus), RSV or

c© The Authors Journal compilation c© 2009 Biochemical Society

Page 4: REVIEW ARTICLE Pathogen recognition in the innate immune ... · Key words: innate immune response, Nod-like receptor (NLR), pathogen recognition, retinoic acid-inducible gene-I receptor

4 H. Kumar, T. Kawai and S. Akira

reovirus show comparable susceptibility with WT (wild-type)mice, suggesting that TLR3 is dispensable for protection againstthese viruses [68]. However, TLR3-deficient mice infected withlethal doses of WNV show resistance to the WNV infection,suggesting that TLR3-mediated inflammatory responses inducedeath of the mice [69]. Therefore, the role of TLR3 in viralinfection is unclear.

TLR7, TLR8 and TLR9

TLR7, TLR8 and TLR9 are located in the intracellular endosomalcompartment, where they sense microbial nucleic acids suchas RNA and DNA. TLR7 and TLR8 (but not mouse TLR8)respond to synthetic antiviral imidazoquinoline compounds suchas R848, loxoribine and imiquimod and ssRNAs rich in guanosineor uridine derived from viruses [70–72] (Figure 1). Generally,these viruses gain entry into cells through receptor-mediatedendocytosis and reach the phagolysosome, where the virus-coatprotein is hydrolysed to expose the viral RNA to the TLRs. Incontrast, the host ssRNA does not reach the endocytic vesiclesbecause it is degraded by RNase.

TLR9 recognizes unmethylated CpG motifs of ssDNA andinduces inflammatory cytokines and type I IFNs. These sequencesare commonly present in the genomes of bacteria and viruses[73–77]. However, in the host, these sequences are highlymethylated at the cytosine base and, therefore, the host CpG motifsstimulate poorly. Synthetic ssDNA-containing CpG dinucleotidesmotifs can also induce the production of inflammatory cytokinesand type I IFNs through TLR9. There are two structurally distincttypes of CpG DNAs known, namely the A-type (D-type) andthe B-type (K-type). A-type CpG ODNs (oligodeoxynucleotides)stimulate pDCs to induce a robust amount of IFN-α and a littleIL-12 [78]. By contrast, B-type CpG ODN is a potent inducer ofinflammatory cytokines such as IL-6, IL-12 and TNF-α (tumournecrosis factor-α) and up-regulates co-stimulatory molecules suchas CD80, CD86 and MHC class II in pDCs and, to lesser extent, inB-cells [79]. DNA viruses such as MCMV, HSV (herpes simplexvirus)-1 and HSV-2 induce inflammatory cytokines and type IIFNs through TLR9 [73–77]. It has been shown that TLR9-deficient mice are susceptible to MCMV infection [76]. HSVrecognition by pDCs does not require viral replication, becauseUV-inactivated virus still induces IFN-α [75]. In addition to theseligands, TLR9 also recognizes the malarial pigment known ashaemozoin [80]. Recently, it was shown that cleavage of TLR9by lysosomal cathepsins is involved in the activation of signalling[81–83].

Signalling through TLR

Upon recognition, TLRs recruit various TIR-domain-containingadaptors to the TIR domain of TLRs. TLR5, TLR7, TLR9 andTLR11 only use MyD88. TLR1, TLR2, TLR4 and TLR6 useTIRAP in addition to MyD88, which links TLR to MyD88. TLR3only uses TRIF. TLR4 uses TRIF and TRAM, and TRAM linksTLR4 with TRIF. Taken together, TLR signalling can be broadlydivided into two signalling pathways: the MyD88-dependent andTRIF-dependent pathways [1,17,47,84,85] (Figures 1 and 2).

In the MyD88-dependent signalling pathway, the IRAK (IL-1receptor-associated kinase) family members such as IRAK4,IRAK1 and IRAK2 are recruited to the MyD88. IRAK4 is initiallyactivated, and IRAK1 and IRAK2 are sequentially activated [86].The activated IRAK family proteins associate with TRAF6 (TNFreceptor-associated factor 6), an E3 ubiquitin ligase, which formsa complex with E2 ubiquitin-conjugating enzymes such as UBC13(ubiquitin C 13) and UEV1A (ubiquitin-conjugating enzyme vari-ant 1A). This complex polyubiquitinates TRAF6 itself and IKKγ

[IκB kinase γ , also known as NEMO (NF-κB essential modifier)]through K63 (Lys63) linkage [87,88]. The polyubiquitinatedTRAF6 activates the protein kinase TAK (transforming growthfactor-β-activated kinase 1) and TABs (TAK1-binding proteins)such as TAB1, TAB2 and TAB3, which subsequently activatetranscription factors such as NF-κB and AP-1 (activator protein-1) through the canonical IKK complex and the MAPK [ERK(extracellular-signal-regulated kinase), JNK (c-jun N-terminalkinase) and p38] pathway respectively for the transcription ofinflammatory cytokine genes [87]. TAK1-deficient cells showreduced inflammatory cytokine levels and impaired NFκB andMAPK activation after stimulation with various TLR ligands[89,90]. TLR4 also activates NF-κB via TRIF by two distinctsignalling pathways [91]. The N-terminus of TRIF interacts withTRAF6 through the TRAF6-binding motifs [92] and the C-ter-minus of TRIF interacts with RIP1 (receptor-interacting protein1), both of which co-operate to activate NF-κB [93] (Figure 2).Recently, we showed that the TRIF-dependent signalling pathwayis negatively regulated by Atg16L1 (autophagy-related 16-like 1).Mice lacking Atg16L1 show enhanced production of cytokine,suggesting an essential role for autophagy in innate immuneregulation [94].

Stimulation with TLR4 and TLR3 ligands activates theTRIF-dependent signalling pathway and induces inflammatorycytokines in addition to type I IFNs and IFN-inducible genesin DCs and macrophages, which depend on IRF3 and IRF7.The production of type I IFNs is absent in TRIF-deficient cells[95]. IRF3 and IRF7 are activated by IKK-related kinase TBK1[TANK binding kinase 1, also known as T2K (TRAF2-associatedkinase) or NAK (NF-κB activating kinase)] and IKKi (alsoknown as IKKε) [96,97]. TBK1 and IKKi interact with TANK(TRAF family member-associated NF-κB activator), NAP1(NAK-associated protein 1) and, similar to NAP1, SINTBAD(TBK1 adaptor), which then phosphorylates IRF3 and IRF7.Phosphorylated IRF3 and IRF7 form a homodimer, whichsubsequently translocates into the nucleus and binds to the ISREs(IFN-stimulated response elements) to induce type I IFNs andIFN-inducible genes [98]. TRAF3 has been proposed to linkTRIF to TBK1, because TRAF3 interacts with these proteins andthe production of IFN-β is abrogated in TRAF3-deficient cells[99,100] (Figure 2).

In pDCs, TLR7 and TLR9 are highly expressed and inducea huge amount of type I IFNs, particularly IFN-α, after virusinfection. Upon stimulation, MyD88 forms a complex with IRF7[101,102] (which is highly expressed in pDCs) and TRAF6 toinduce the production of type I IFNs [100]. IRAK1 interactswith MyD88 and can phosphorylate IRF7 [103]. IRAK1-deficientpDCs consistently have defects in type I IFN production, butshow intact inflammatory cytokine production. Similarly, theproduction of type I IFNs is decreased in IKKα-deficient mice,and IKKα can bind to, and phosphorylate, IRF7 [104]. Thesefindings suggest that IRAK1 and IKKα act as IRF7 kinases.By contrast, pDCs lacking MyD88 or IRAK4 do not inducetype I IFNs and inflammatory cytokines. Taken together, theseobservations suggest that the TLR7– or TLR9–MyD88–TRAF6–IRAK4–IRAK1–IKKα–IRF7 signalling pathway is active inpDCs for the robust production of type I IFNs after virus infection(Figure 3). Recently, the involvement of the serine/threonineprotein kinase mTOR (mammalian target of rapamycin) and itsdownstream signalling kinases, such as the p70 ribosomal S6protein kinases p70S6K1 and p70S6K2, has been shown to playroles in pDC for the production of type I IFNs. Inhibition ofmTOR and its downstream kinases blocks the interaction betweenTLR9 and MyD88 and inhibits further activation of IRF7[105].

c© The Authors Journal compilation c© 2009 Biochemical Society

Page 5: REVIEW ARTICLE Pathogen recognition in the innate immune ... · Key words: innate immune response, Nod-like receptor (NLR), pathogen recognition, retinoic acid-inducible gene-I receptor

Pathogen recognition in the innate immune response 5

Figure 2 TLR signalling in conventional DCs or macrophages

The engagement of TLRs with their respective ligands initiates signalling. MyD88 recruits the IRAK family of proteins and TRAF6. TRAF6 activates TAK1 which, in turn, activates the IKK complexconsisting of IKKα, IKKβ and NEMO/IKKγ , and phosphorylates IκBs. Phosphorylated IκBs are ubiquitinated and undergo proteasome-mediated degradation, and NF-κB subunits, which consist ofp50 and p65, translocate to the nucleus. TAK1 also activates the MAPK signalling pathway. The activated NF-κB and MAPK initiate the transcription of inflammatory cytokine genes. TRIF recruits RIP1and TRAF6. Activated TRAF6 and RIP1 activate NF-κB and MAPK to induce transcription of inflammatory cytokine genes. TRIF interacts with TRAF3 and activates TBK1/IKKi, which phosphorylateIRF3 and IRF7. The phosphorylated IRF3 and IRF7 are translocated to the nucleus for the transcription of type I IFNs.

The ER (endoplasmic reticulum)-localized transmembraneprotein known as UNC93B1 was shown to be essential forthe production of inflammatory cytokines after stimulation withTLR3, TLR7 and TLR9 ligands [106]. In addition, UNC93B1plays a crucial role in the cross-presentation of an exogenousantigen via MHC Class I and Class II. Moreover, UNC93B1 isrequired for the translocation of TLR7 and TLR9 from the ER tothe endolysosome [107–109].

TLR and human diseases

Genetic variations or a deficiency of genes encoding TLR andTLR signalling proteins have been implicated in the predispositionto innate immune diseases [110]. Recently, autosomal recessiveMyD88-deficient pediatric patients have been reported. Thesepatients show susceptibility to pyrogenic bacterial infections suchas Strep. pneumoniae and Staph. aureus, and severe complicationshave been reported in several patients in early childhood.Otherwise, these patients are healthy and have normal immunityto other microbes, and their clinical severity improves with age.These observations suggest that MyD88-dependent signalling isessential for protective immunity against a few types of pyrogenicbacteria, but is dispensable for the host defence to the majority ofother infections in humans [111].

IRAK4 deficiency has also been reported in humans. TheIRAK4 gene is located on an autosomal chromosome and 28individuals with a recessive gene have been reported so far.These patients show increased susceptibility to infection by Gram-positive and Gram-negative bacteria in early childhood [112].Similar to MyD88 deficiency or mutants, IRAK4-deficient peoplealso show an improvement in symptoms with advancing age.These patients also show impaired induction of type I IFNs, butdo not show any susceptibility to viral infection or HSE (herpessimplex virus encephalitis). Furthermore, these individuals donot show susceptibility to any parasitic or fungal diseases. Pat-ients with TLR3 deficiency have also been reported. Thesepatients show increased susceptibility to HSV-1 [113]. These ob-servations suggest that TLRs play an important role in thepathogenesis of some diseases. UNC93B deficiency has alsobeen documented in some patients. Cells from these patientsdo not respond to TLR3, TLR7, TLR8 or TLR9. However,similar to IRAK4-deficient patients, UNC93B patients do notshow susceptibility to viral diseases such as HSE [114].

RIG-I-LIKE RECEPTORS

Much progress has been made in TLR-independent recognition ofviral nucleic acids, particularly RNA recognition by intracellular

c© The Authors Journal compilation c© 2009 Biochemical Society

Page 6: REVIEW ARTICLE Pathogen recognition in the innate immune ... · Key words: innate immune response, Nod-like receptor (NLR), pathogen recognition, retinoic acid-inducible gene-I receptor

6 H. Kumar, T. Kawai and S. Akira

Figure 3 TLR signalling in pDCs

TLR7 and TLR9, which are associated with UNC-93B, are transported to the endolysosome. In the endolysosome, these TLRs interact with their respective ligands and recruit MyD88. MyD88interacts with TRAF6 through IRAK4 and activates TAK1. TAK1 activates NF-κB through the IKK complex. MyD88 also interacts with IRAK4 and IRAK1. The IRAK1 and IKKα phosphorylate IRF7.IRAK1 interacts with TRAF3 and phosphorylates IRF7. The phosphorylated IRF7 is translocated to the nucleus for the transcription of type I IFNs.

sensors [1,115–118]. The intracellular sensors are important forthe detection of RNA derived from RNA viruses and replicatingDNA viruses. The recognition of RNA by intracellular sensorssubsequently activates innate antiviral responses, mainly throughthe induction of type I IFNs and inflammatory cytokines in mostcell types.

In this regard, DExD/H-box RNA helicase, known RIG-I, wasfound to have a role in the cytoplasmic recognition of dsRNAand activation of IRFs and NF-κB [119]. Two members ofthis family known as Mda5 (melanoma differentiation-associatedgene 5) and Lgp2 (Laboratory of Genetics and Physiology 2)were subsequently identified [120]. These proteins are collectivelyknown as RLRs. RIG-I and Mda5 contain two tandem repeatsof the CARD (caspase recruitment domain) at their N-terminus,which are important for activating downstream signalling. Theintermediate portion of these proteins contains the helicasedomain, which is similar to other members of the DExD/H-boxRNA helicase family. For RIG-I, this domain contains an ATP-binding region, which is essential for RIG-I function. However,an ATP-binding region was not found in Mda5. In addition, RIG-

I contains an RD (repressor domain) at the C-terminus, whichrepresses the activity of RIG-I [121,122]. Repression of Mda5 inthe steady state is not known, but it has been postulated that MdA5might be negatively regulated by other proteins such as DAK(dihydroacetone kinase) [123]. Lgp2 contains the RNA helicasedomain, but is devoid of the CARD. This protein was consideredto be a negative regulator for RIG-I and Mda5. However, a studyof Lgp2-deficient mice revealed that Lgp2 acts as both a negativeand positive regulator depending on the virus [124]. Recently, ahomologue of RIG-I helicase (DExD/H-box helicase), Dicer-2,was identified in Drosophila and which controls the expressionof an antiviral protein known as Vago after virus infection,suggesting an evolutionarily conserved role of RLR in antiviralresponses [125].

Pathogen recognition by RLRs

Studies of RIG-I- and Mda5-deficient mice revealed that thesesensors recognize different classes of RNA viruses. RIG-I-deficient cells infected with NDV (Newcastle-disease virus),

c© The Authors Journal compilation c© 2009 Biochemical Society

Page 7: REVIEW ARTICLE Pathogen recognition in the innate immune ... · Key words: innate immune response, Nod-like receptor (NLR), pathogen recognition, retinoic acid-inducible gene-I receptor

Pathogen recognition in the innate immune response 7

VSV, SeV (Sendai virus) and IV (influenza virus) show impairedtype I IFN and inflammatory cytokine production [126,127].Furthermore, members of the flavivirus family, such as JEV(Japanese encephalitis virus) and HCV (hepatitis C virus) arealso recognized by RIG-I [128,129]. However, dengue virus andWNV, which belong to the flavivirus family, do not require RIG-I.By contrast, Mda5-deficient mice show normal production oftype I IFNs and inflammatory cytokines against NDV, VSV, SeV,IV and JEV. However, they show impaired ability to produce type IIFNs and inflammatory cytokines against picornaviruses such asEMCV, Theiler’s virus and Mengo virus [128]. These observationssuggest that these two sensors induce antiviral responses against awide spectrum of RNA viruses with different specificity. However,RLRs are not sufficient for the protection against other RNAviruses such as IV, RSV and LCMV in vivo. In other words, in vivoinfection of LCMV induces the production of type I IFNs and thepromotion of virus-specific CD8 T-cells through the TLR7 ratherthan the RLR signalling pathway [130]. For IV infection, the RLRsignalling pathway is essential for the induction of cytokines infibroblasts, alveolar macrophages, and cDCs, whereas TLR7 actsin pDCs to induce cytokines [131,132]. However, the productionof virus-specific antibodies is dependent on TLR7 rather thanRLR. Similar observations were reported for RSV infection[133], suggesting that TLR and RLR signalling pathways togetherinduce the host defence against these viruses. Moreover, co-operative activation of TLR and RLR is also required for theadjuvant effects of poly(I-C) [134].

RIG-I and Mda5 recognize in-vitro-synthesized dsRNA anda synthetic analogue of dsRNA poly(I-C) respectively [128].Further studies revealed that the 5′-triphosphate moiety of RNAis essential for RIG-I recognition [135] and it is independent ofthe strand property (single or double) of RNA [136]. A recentbiochemical study revealed that the 5′-triphosphate moiety ofRNA is recognized by the CTD (C-terminal domain) of RIG-I[122]. Host-cell RNA is not recognized by RIG-I because, duringsynthesis of cellular RNA, the 5′-ends are either modified bythe addition of a 7-methylguanosine cap or the 5′-triphosphateis removed before transportation to the cytoplasm. Thus RIG-Ican discriminate between viral and host RNA. RIG-I can bind toa 25-bp dsRNA to efficiently induce type I IFNs and the RNAend structures (blunt end, 5′-overhang and 3′-overhang), and thenucleotide sequences are not critical for binding to RIG-I [137].Furthermore, it has been shown, using NMR of the CTD, thatRIG-I recognizes two distinct viral RNA patterns, including dsand 5′-triphosphate ssRNA, and gel-filtration analysis revealedthat a dimer of RIG-I CTD mediates the recognition of 5′-triphosphate RNA [118]. The length of dsRNA is critical fordifferential recognition by RIG-I and Mda5; RNA viruses have ashorter RNA length (approx. 1.2–1.4 kbp) and are recognized byRIG-I, whereas viruses with longer dsRNA (longer than 3.4 kbp)are recognized by Mda5 [138].

SIGNALLING THROUGH RLRs FOR ANTIVIRAL RESPONSES

In response to viral infection, the CARDs of RIG-I and Mda5associate with the CARD-containing adaptor protein knownas IPS-1 {IFN promoter stimulator-1, also known as MAVS(mitochondrial antiviral signalling), Cardif (CARD adaptor in-ducing IFN-β) and VISA (virus-induced signalling adaptor [139–142]} to induce inflammatory cytokines and type I IFNs. Ectopicexpression of IPS-1 in cells activates NF-κB and IFN promoters.In addition, IPS-1-deficient cells show a complete abrogation ofinflammatory cytokines and type I IFNs after virus infection.Furthermore, IPS-1-deficient mice infected with various RNAviruses recognized by RIG-I and Mda5 show enhanced motility

compared with that in WT mice [143,144]. These observationscollectively suggest that IPS-1 is the sole adaptor for RIG-I/Mda5 and plays an essential role in host defence againstvarious RNA viruses. It has been shown that IPS-1 is localizedin the outer membrane of mitochondria [140]. NS3/4 (non-structural 3/4) protease from HCV cleaves and dislodges IPS-1from the mitochondria to block IFN production, suggesting thatmitochondrial localization is essential for IPS-1 function withrespect to antiviral responses [141]. The NLR family proteinknown as NLRX1 was recently reported to be localized in the outermembrane of mitochondria and to inhibit IPS-1-mediated type IIFN induction in response to virus infection [145]. Signallingthrough RIG-I is further regulated by ubiquitination. It was shownthat TRIM25 (tripartite motif 25), a ubiquitin E3 ligase whichcontains an RNF (RING-finger) domain, a B-box/coiled-coildomain and a SPRY (SPla/RYanodine) domain, interacts with theN-terminal CARD of RIG-I. This interaction leads to the Lys63-linked ubiquitination of RIG-I. Furthermore, TRIM25-deficientcells show abrogation of type I IFNs production, suggesting thatubiquitination of RIG-I is essential for activation of the signallingpathway[146]. By contrast, RIG-I is inhibited by another ubiquitinligase, RNF125, which induces ubiquitination and proteasomaldegradation of RIG-I. These observations suggest that theubiquitination is an additional regulatory mechanism for the RIG-I-mediated signalling pathway [147] (Figure 4).

TRAF3, an E3 ubiquitin ligase that polyubiquitinates throughits C-terminal TRAF domain, was shown to interact withIPS-1 and activates TBK1 and IKKi [99,100,148]. Recently,a de-ubiquitinase enzymatic protein known as DUBA (de-ubiquitinating enzyme A) was reported to deubiquitinate TRAF3and inhibit the RLR-mediated signalling pathway [149].

RLR-mediated NF-κB activation is achieved via the FADD(Fas-associated death domain) protein, which interacts withcaspase 8 and caspase 10 and forms a complex with IPS-1[139,150]. The TNFR-I (TNF receptor-I) signalling adaptorTRADD (TNF-receptor-associated DD), an adaptor in theTNFR-I signalling pathway, has been suggested to be involvedin RLR signalling. The engagement of RLRs by viruses leads tothe formation of a molecular complex that consists of TRADD,FADD and RIP1. This complex interacts with IPS-1 and activatesIRF3 and NF-κB [151].

The ER-localized STING [stimulator of interferon genes, alsoknown as MITA (mediator of IRF3 activation)] protein wasrecently identified and shown to be involved in host defenceagainst RNA virus. Knockdown of STING impaired IFN-βproduction by poly(I-C) transfection. The replication of RNAviruses is higher in STING-deficient fibroblast cells than in WTcells. In addition, the production of IFN-β was also reducedin STING-deficient fibroblast cells. However, STING-deficientbone-marrow-derived macrophages and DCs show replication ofvirus comparable with that shown by WT cells, suggesting a cell-type-specific role of STING [152,153].

Autophagy is an essential biological process that maintainscellular homoeostasis, development, differentiation and tissueremodelling. Recent studies have highlighted the importance ofautophagy in innate immunity. Recently it was shown that IPS-1 and RIG-I were associated with the Atg5–Atg12 complex,which is an essential component for autophagy. Mouse embryonicfibroblasts deficient in Atg5 and Atg12 show increased levelsof type I IFN production in response to VSV infection [154].This observation suggests that autophagy is important in thenegative regulation of the RLR signalling pathway. However, ithas been shown that autophagy plays an opposite role in pDCs.In pDCs, TLR7 recognizes ssRNA in the endolysosome, butit also recognizes replicating VSV. Atg5-deficient pDCs show

c© The Authors Journal compilation c© 2009 Biochemical Society

Page 8: REVIEW ARTICLE Pathogen recognition in the innate immune ... · Key words: innate immune response, Nod-like receptor (NLR), pathogen recognition, retinoic acid-inducible gene-I receptor

8 H. Kumar, T. Kawai and S. Akira

Figure 4 Recognition of RNA and RNA viruses by RLRs

Viral RNA, ssRNA, dsRNA and dsRNA analogue poly(I-C) are recognized in the cytoplasm of cDCs, macrophages and fibroblast cells by RIG-I and Mda5. Upon recognition, the CARD of Mda5 andubiquitinated CARD of RIG-I bind to IPS-1 (located on the outer membrane of mitochondria). The CARD of RIG-I undergoes ubiquitination by TRIM25. IPS-1 then interacts with TRADD and formsa complex with FADD and caspase 8/10 to activate NF-κB through the IKK complex. TRADD also interacts with TRAF3 and activates TBK1/IKKi, which phosphorylates IRF3 and IRF7 to induce thetranscription of type I IFNs. The mitochondrial NLRX1 inhibits IPS-1-mediated signalling. Mitochondrially localized STING interacts with RIG-I and IPS-1 and activates NF-κB and IRFs.

reduced IFN-α production after VSV infection, suggesting that theautophagosome is formed after VSV infection and contains viralRNA, and this autophagosome traffics to the lysosome, where itforms a complex with the autophagolysosome after fusion withthe lysosome. In the autophagolysosome, TLR7 recognizes viralRNA for the production of IFN-α [155].

RECOGNITION OF DNA BY CYTOSOLIC RECEPTOR

Bacterial and viral dsDNA are immunostimulatory componentsthat activate various cell types to induce type I IFNsand inflammatory cytokines through cytosolic DNA sensors.Cytosolic recognition of dsDNA induces TBK1/IKKi-dependenttype I IFNs and NF-κB-dependent inflammatory cytokines[156,157] (Figure 5). DAI (DNA-dependent activator of IRFs) hasbeen reported to be a cytosolic sensor for DNA [158]. However,DAI-deficient cells do not show impaired cytokine production,suggesting that DAI is not essential for the recognition of DNA[159]. Recently, we have shown that a DNA vaccine that consistsof plasmid DNA and has inherited properties of adjuvant inducesinnate and adaptive immunity, such as antigen-specific antibodyproduction and CD4 and CD8 T-cell responses, through TBK1 butnot TLR9, MyD88 and DAI. This suggests that the DNA-inducedadjuvant effects are TLR- and DAI-independent phenomena and

these effects may depend on an unknown DNA sensor that signalsthrough TBK1 [159].

Recently, STING has been shown to be involved in therecognition of dsDNA. STING was shown to play a pivotal rolein type I IFN production by dsDNA when introduced into thecytosol. Furthermore, infection with a DNA virus such as HSV-1, and Listeria monocytogenes, organisms which are known tointroduce DNA into the cytosol during infection, shows a reducedtype I IFN production in STING-deficient cells. This suggeststhat STING plays an important role in DNA sensor signallingpathways [152,153].

NOD-LIKE RECEPTORS

The NLR family of proteins are cytosolic, intracellular PRRsthat recognize PAMPs and endogenous ligands. The recognitionof ligands induces a signalling cascade leading to activation ofNF-κB, or a cytoplasmic multiprotein complex known asthe inflammasome, to produce inflammatory cytokines [1,11–13,160,161]. In addition, NLRs are also involved in the signallingfor cell death after microbial infection [162]. The NLR familycomprises 23 proteins in humans and 34 proteins in mice[163]. However, the function of most of the NLR proteins ispoorly understood. These proteins have a trimodular structure

c© The Authors Journal compilation c© 2009 Biochemical Society

Page 9: REVIEW ARTICLE Pathogen recognition in the innate immune ... · Key words: innate immune response, Nod-like receptor (NLR), pathogen recognition, retinoic acid-inducible gene-I receptor

Pathogen recognition in the innate immune response 9

Figure 5 Recognition of DNA and DNA viruses by DNA sensors

An unknown cytosolic DNA sensor or DAI activates NF-κB through activation of the IKK complex and IRF3 and IRF7 through TBK1/IKKi. ER-localized STING induces the activation of NF-κB andIRFs. DNA virus infection and introduction of dsDNA activates the inflammasome consisting of AIM2/ASC and NALP3/ASC respectively. The inflammasome converts inactive pro-caspase 1 intoactive caspase 1 for the maturation and production of IL-1β . Adenovirus infection directly or indirectly activates the NALP3 inflammasome.

and consist of the following domains. The C-terminal domainconsists of tandem repeats of LRR, which are essential forsensing or recognition of the microbial components. A centrallylocated nucleotide binding NOD domain is essential for self-oligomerization and formation of a complex for the activationand recruitment of downstream signalling proteins. The variableN-terminal domains are defined by CARD, DED (death effectordomain), PYD (pyrin domain) or BIR (baculovirus inhibitorof apoptosis protein repeat) domain [163]. These domains areessential for downstream signal transduction through homotypicprotein–protein interactions. Mutations in these proteins werereported to be associated with chronic inflammatory diseasessuch as familial cold autoinflammatory syndromes (familialcold urticaria) [164,165], Muckle-well (or urticaria–deafness–amyloidosis) syndrome and Crohn’s disease [166].

NOD1 AND NOD2

NOD1 (also known as CARD4) recognizes a distinct substructureof PGN (peptidoglycan), namely iE-DAP (γ -D-glutamyl-m-

diaminopimelic acid, a dipeptide), which is present in Gram-negative and Gram-positive bacteria such as Bacillus subtilis andL. monocytogenes [167,168]. NOD2 (CARD15) recognizes MDP(muramyl dipeptide), the largest component of PGN motif, whichis also present in Gram-negative and Gram-positive bacteria [169].

Upon stimulation with these ligands, NOD1 and NOD2 interactwith a CARD domain-containing serine/threonine kinase knownas CARDIAK [CARD-containing IL-1β-converting-enzyme-associated kinase, also known as RICK (RIP-like interactingcaspase-like apoptosis-regulatory protein kinase) and RIP2][170,171] and induces antimicrobial peptides [172] and in-flammatory cytokines through activation of MAPKs [173] andNF-κB. MAPKs are activated through another CARD-containingprotein known as CARD9 [174] (Figure 6). Ex vivo studiesshowed that various pathogenic bacteria, such as E. coli[175], Shigella flexneri [168], Pseudomonas aeruginosa [176],Chlamydia species [177–179], Campylobacter jejuni [180] andHaemophilus influenza [181] are sensed by NOD1 and Strep.pneumonia [182] and M. tuberculosis [183] are sensed by NOD2.L. monocytogenes has been reported to be sensed by both NOD1and NOD2 [184,185]. In vivo, it has been shown that NOD1 and

c© The Authors Journal compilation c© 2009 Biochemical Society

Page 10: REVIEW ARTICLE Pathogen recognition in the innate immune ... · Key words: innate immune response, Nod-like receptor (NLR), pathogen recognition, retinoic acid-inducible gene-I receptor

10 H. Kumar, T. Kawai and S. Akira

Figure 6 NLR signalling pathway

NOD1 and NOD2 sense various PAMPs of pathogenic bacteria in the cytoplasm or PAMPs thatare transported through the type III or type IV secretion systems. Upon recognition of PAMPs,these sensors undergo for self-oligomerization and formation of a complex that triggers theassociation of CARDIAK, which in turn activates NF-κB. NOD1 and NOD2 also activatethe MAPK signalling pathway through CARD9.

NOD2-deficient mice are susceptible to L. monocytogenes and H.pylori respectively [185,186].

The inflammasome

Pathogen infection into the host results in the production ofvarious inflammatory cytokines. The IL-1 family of cytokines,including IL-1β, IL-18 and IL-33, are key cytokines that regulatevarious components of innate and adaptive immunity. Theproduction of these cytokines is regulated by two signals inthe innate immune cells (e.g. macrophages). The first signal istranscriptional and translational up-regulation of pro-forms of thecytokine (pro-IL-1β, pro-IL-18) in response to various TLR, NLRand RLR agonists. The second signal is the processing of the pro-form of these cytokines to the mature, secretary form of the cyto-kines by the inflammasome. Depending on the NLR proteins, theinflammasome is categorized into three types such as NALP3[NACHT (NTPase-domain named after NAIP, CIITA, HET-E and TP1)–LRR-PYD–containing protein 3, also known asNLRP3, CIAS1 (cold-induced autoinflammatory syndrome 1) andcryopyrin] inflammasome, the IPAF [IL-1β-converting-enzymeprotease-activating factor, also known as the NLRC4 (NLRfamily, CARD domain containing 4)] and CLAN (CARD LRR-and NACHT-domain-containing protein) inflammasome and theNALP1 (also known as NLRP1) inflammasome (Figure 7).

NALP3 inflammasome

The NALP3 inflammasome is activated by various exogenous andhost endogenous ligands. Exogenous ligands include microbialligands such as MDP, bacterial and viral RNA, toxins such asnigericin, maitotoxin, environmental pollutants such as asbestosand silica [187] and vaccine adjuvant alum (aluminium salts)[188,189]. Host endogenous ligands include MSU (monosodiumurate), calcium pyrophosphate dehydrate, amyloid-β fibrillarpeptide and ATP. In addition, the NALP3 inflammasome is alsoactivated by UV light and skin irritants such as picryl chloride and2,4-dinitrofluorobenzene. The NALP3 inflammasome consistsof NALP3, CARDINAL (CARD inhibitor of NF-κB-activatingligands), ASC (apoptosis-associated speck-like protein containinga CARD) and caspase 1, which process cytosolic pro-IL-1β tobioactive secretory IL-1β [187].

Activation of the inflammasome leads to the production of IL-1β, which may play an important role in clearing pathogensfrom the host. A recent study has shown that the NALP3inflammasome is essential for alum-induced adjuvant effectssuch as antibody production to antigens and Th2-mediatedinflammation [189]. However, there is another study showing thatthe NALP3 inflammasome is dispensable for the adjuvant effects,but indispensible for IL-1β production [190]. There are severalpossible differences among these studies, such as preparationof mixture of antigen and alum before immunization, the dose ofantigen or adjuvant, the purity of antigen (some antigen hasimpurity of immunomodulatory substances), and the route andprotocol of immunization. Therefore it is almost impossible toevaluate the data from two different laboratories. Furthermore,the differences in results could be due to multiple mechanismsof action of alum. Taken together, further studies are needed toconclude the involvement of NALP3 inflammasome in adjuvanteffects of alum. It has been suggested that activation of theinflammasome by the host ligand plays an important role inthe pathogenesis of arthritic diseases such as gout, pseudogout[187] and Alzheimer’s disease [191]. The exogenous ligand silicainduces NALP3-inflammasome-dependent silicosis [192,193].These observations suggest that the NALP3 inflammasome playsa pivotal role in the activation of innate immune responses for hostdefence and is a mediator for the pathogenesis of various diseases.

Recently, the introduction of bacterial, viral and host dsDNAinto the cytosol of cells has been shown to induce the productionof IL-1β, which requires the inflammasome components ASCand caspase 1, but not NALP3. However, the introduction ofadenoviral DNA showed that IL-1β production depends onNALP3, ASC and caspase 1. AIM2 (absent in melanoma 2)has been shown to recognize dsDNA. AIM2 is a member of theIFI20X (interferon-inducible protein 20X)/IFI16 or PYHIN [PYDand HIN (haematopoietic interferon-inducible) nuclear proteindomain family member 1]. It has been shown that the HIN200domain of AIM2 binds to dsDNA, whereas PYD associates withASC [194–197] (Figure 5).

The NALP3 inflammasome is activated by numerous ligands;however, it is unknown whether these ligands bind directly toNALP3. Pore-forming toxins such as staphylococcal α-toxin andpneumolysin, K+ ionophores and a low concentration of detergentinduce an efflux of K+ to activate the NALP3 inflammasome.In addition, ATP, maitotoxin and nigericin activate hemichannelpannexin-1 and the purinergic receptor P2X7 to induce effluxof K+ and activate the NALP3 inflammasome. Furthermore,ROS (reactive oxygen species) are also reported to activate theNALP3 inflammasome [187]. Two different mechanisms for ROSproduction have been proposed, including efflux of K+-inducedstress to the mitochondria and NADPH oxidase-mediated

c© The Authors Journal compilation c© 2009 Biochemical Society

Page 11: REVIEW ARTICLE Pathogen recognition in the innate immune ... · Key words: innate immune response, Nod-like receptor (NLR), pathogen recognition, retinoic acid-inducible gene-I receptor

Pathogen recognition in the innate immune response 11

Figure 7 Activation of the inflammasome

The inflammasome is activated in two steps. The first step is transcriptional and translational up-regulation of pro-forms of the cytokine (pro-IL-1β , pro-IL-18) in response to various TLR, NLR andRLR agonists through activation of NF-κB. In the second step, the inactive form of the cytokines is converted into a biologically active form by the inflammasome. The ligands (described in the text)directly and indirectly (through an unknown sensor) induce the formation of various inflammasomes such as the NALP3 inflammasome, the IPAF inflammasome or the NALP1 inflammasome. Inaddition to ASC, the NALP3 inflammasome contains CARDINAL protein (the domain arrangement of each component is shown in the box labelled ‘Components of inflammasome’. The inflammasomeconverts inactive pro-caspase 1 into active caspase 1, which induces cell death (in the case of NALP1 inflammasome) and/or converts pro-cytokines (inactive) into bioactive secretory cytokines. TheNALP1 protein interacts with anti-apoptotic proteins such as Bcl-2 and Bcl-XL and inhibits caspase-1-dependent cell death and production of IL-1β and IL-18.

induction in the phagolysosome. Other mechanisms have alsobeen proposed recently. Phagocytosis of silica crystal and fibrousparticles of amyloid-β induce lysosomal destabilization andpermeabilization which leads to the release of cathepsin B into thecytosol [191,198]. The released cathepsin B activates the NALP3inflammasome.

IPAF inflammasome

The IPAF inflammasome is activated by intracellular pathogenssuch as Salmonella typhimurium [199] and Legionellapneumophila [200]. It was suggested that S. typhimurium andL. pneumophila deliver flagellin to the cytosol via the type IIIand type IV secretion systems respectively [199,200]. The intro-duction of recombinant purified flagellin protein into the cyto-sol also promotes IPAF-dependent caspase 1 activation [201].Flagellin of Ps. aeruginosa also induces IPAF- and caspase-1-dependent production of IL-1β [202]. In addition to IPAF,NAIP5 (apoptosis inhibitory protein 5, also known as Birc1 (BIR-

containing 1)] was reported to be involved in the recognition ofL. pneumophila [203,204]. NAIP5 consists of three consecutiverepeats of the BIR domain at the N-terminus. Previous studieshave indicated that NAIP5 regulates the replication of L.pneumophila via caspase-1-dependent cell death. It has beenreported that 35 amino acids from the C-terminus of flagellinand infection with L. pneumophila is sufficient to activate theinflammasome, but is not activated in the absence of NAIP5. Inaddition, full-length flagellin activates NAIP5-independent, butIPAF-dependent, cell death [205]. These findings suggest thatthe IPAF and NAIP5 proteins are essential for the flagellin-induced immune response.

NALP1 inflammasome

Anthrax LT (lethal toxin) is a potent toxin of Bacillus anthracisthat induces cell death [206]. LT is composed of a protectiveantigen and a lethal factor. The protective antigen is a receptor-binding protein that creates a pore to deliver the lethal factor

c© The Authors Journal compilation c© 2009 Biochemical Society

Page 12: REVIEW ARTICLE Pathogen recognition in the innate immune ... · Key words: innate immune response, Nod-like receptor (NLR), pathogen recognition, retinoic acid-inducible gene-I receptor

12 H. Kumar, T. Kawai and S. Akira

into the cytosol of infected cells and induce cell death. It hasbeen shown that the polymorphic gene known as Nlrp1b isresponsible for susceptibility to LT [207]. It was also shownthat LT-induced macrophage death requires caspase 1, which isactivated in susceptible macrophages [207]. Furthermore, the LTof B. anthracis was shown to induce IL-1β, which depends onthe NALP1 inflammasome. In addition, it was shown that NOD2is also required for IL-1β production and NOD2–NALP1 formsa molecular complex during infection [208].

NALP1 is implicated in the association with anti-apoptoticproteins Bcl2 (B-cell lymphoma-2) and Bcl-X(L), which suppresscaspase-1 activation, indicating a role of Bcl-2 proteins in theinhibition of LT-induced cytotoxicity [209].

CONCLUSIONS AND FUTURE DIRECTIONS

Although numerous studies have greatly enhanced ourunderstanding of innate immune recognition by TLRs, RLRs,NLRs and unknown PRRs, we still know relatively little aboutthe recognition of the vast array of micro-organisms and thecross-talk among these receptors. Although the cytosolic receptorfor DNA, which induces IL-1β production, has been reported,we have not yet characterized the sensors for DNA or DNAviruses that activate IRFs. The function of most of the membersof the NLR family in host defence and their ligands are alsounidentified. Characterization of these PRRs in innate immunitywill further improve our understanding of the complexities ofinnate immune regulation. In addition, structural studies of PRR–ligand complexes will improve our understanding of innateimmunity and facilitate the design of novel drugs that targetPRRs.

ACKNOWLEDGEMENTS

We thank the members of our laboratory for insightful discussions and apologizeto those authors whose work could not be included in this review owing to spacelimitations.

FUNDING

This work was supported by the Japan Society for the Promotion of Science [grant numberPO8123 (postdoctoral fellowship to H. K.)].

REFERENCES

1 Akira, S., Uematsu, S. and Takeuchi, O. (2006) Pathogen recognition and innateimmunity. Cell 124, 783–801

2 Medzhitov, R. (2007) Recognition of microorganisms and activation of the immuneresponse. Nature 449, 819–826

3 Beutler, B. (2004) Inferences, questions and possibilities in Toll-like receptor signalling.Nature 430, 257–263

4 Janeway, Jr., C. A. and Medzhitov, R. (2002) Innate immune recognition. Annu Rev.Immunol. 20, 197–216

5 Hoebe, K., Janssen, E. and Beutler, B. (2004) The interface between innate and adaptiveimmunity. Nat. Immunol. 5, 971–974

6 Lemaitre, B., Nicolas, E., Michaut, L., Reichhart, J. M. and Hoffmann, J. A. (1996) Thedorsoventral regulatory gene cassette spatzle/Toll/cactus controls the potent antifungalresponse in Drosophila adults. Cell 86, 973–983

7 Medzhitov, R., Preston-Hurlburt, P. and Janeway, Jr., C. A. (1997) A human homologueof the Drosophila Toll protein signals activation of adaptive immunity. Nature 388,394–397

8 Poltorak, A., He, X., Smirnova, I., Liu, M. Y., Van Huffel, C., Du, X., Birdwell, D., Alejos,E., Silva, M., Galanos, C. et al. (1998) Defective LPS signalling in C3H/HeJ andC57BL/10ScCr mice: mutations in Tlr4 gene. Science 282, 2085–2088

9 Hoshino, K., Takeuchi, O., Kawai, T., Sanjo, H., Ogawa, T., Takeda, Y., Takeda, K. andAkira, S. (1999) Cutting edge: Toll-like receptor 4 (TLR4)-deficient mice arehyporesponsive to lipopolysaccharide: evidence for TLR4 as the Lps gene product.J Immunol. 162, 3749–3752

10 Yoneyama, M. and Fujita, T. (2007) Function of RIG-I-like receptors in antiviral innateimmunity. J. Biol. Chem. 282, 15315–15318

11 Chen, G., Shaw, M. H., Kim, Y. G. and Nunez, G. (2008) Nod-like receptors: role ininnate immunity and inflammatory disease. Annu. Rev. Pathol. 4, 365–398

12 Ye, Z. and Ting, J. P. (2008) NLR, the nucleotide-binding domain leucine-rich repeatcontaining gene family. Curr. Opin. Immunol. 20, 3–9

13 Dostert, C., Meylan, E. and Tschopp, J. (2008) Intracellular pattern-recognitionreceptors. Adv. Drug Deliv. Rev. 60, 830–840

14 Janeway, C. A., Jr. (1989) Approaching the asymptote? Evolution and revolution inimmunology. Cold Spring Harbor Symp. Quant. Biol. 54, 1–13

15 Akira, S. and Takeda, K. (2004) Toll-like receptor signalling. Nat. Rev. Immunol. 4,499–511

16 Matsushima, N., Tanaka, T., Enkhbayar, P., Mikami, T., Taga, M., Yamada, K. and Kuroki,Y. (2007) Comparative sequence analysis of leucine-rich repeats (LRRs) withinvertebrate toll-like receptors. BMC Genom. 8, 124

17 O’Neill, L. A. and Bowie, A. G. (2007) The family of five: TIR-domain-containingadaptors in Toll-like receptor signalling. Nat. Rev. Immunol. 7, 353–364

18 Jin, M. S., Kim, S. E., Heo, J. Y., Lee, M. E., Kim, H. M., Paik, S. G., Lee, H. and Lee,J. O. (2007) Crystal structure of the TLR1–TLR2 heterodimer induced by binding of atri-acylated lipopeptide. Cell 130, 1071–1082

19 Liu, L., Botos, I., Wang, Y., Leonard, J. N., Shiloach, J., Segal, D. M. and Davies, D. R.(2008) Structural basis of toll-like receptor 3 signalling with double-stranded RNA.Science 320, 379–381

20 Kim, H. M., Park, B. S., Kim, J. I., Kim, S. E., Lee, J., Oh, S. C., Enkhbayar, P.,Matsushima, N., Lee, H., Yoo, O. J. and Lee, J. O. (2007) Crystal structure of theTLR4-D-2 complex with bound endotoxin antagonist Eritoran. Cell 130, 906–917

21 Jin, M. S. and Lee, J. O. (2008) Structures of the Toll-like receptor family and its ligandcomplexes. Immunity 29, 182–191

22 Schroder, N. W., Morath, S., Alexander, C., Hamann, L., Hartung, T., Zahringer, U.,Gobel, U. B., Weber, J. R. and Schumann, R. R. (2003) Lipoteichoic acid (LTA) ofStreptococcus pneumoniae and Staphylococcus aureus activates immune cells viaToll-like receptor (TLR)-2, lipopolysaccharide-binding protein (LBP), and CD14,whereas TLR-4 and MD-2 are not involved. J. Biol. Chem. 278, 15587–15594

23 Takeuchi, O., Kaufmann, A., Grote, K., Kawai, T., Hoshino, K., Morr, M., Muhlradt, P. F.and Akira, S. (2000) Cutting edge: preferentially the R-stereoisomer of the mycoplasmallipopeptide macrophage-activating lipopeptide-2 activates immune cells through atoll-like receptor 2- and MyD88-dependent signalling pathway. J. Immunol. 164,554–557

24 Lien, E., Sellati, T. J., Yoshimura, A., Flo, T. H., Rawadi, G., Finberg, R. W., Carroll, J. D.,Espevik, T., Ingalls, R. R., Radolf, J. D. and Golenbock, D. T. (1999) Toll-like receptor 2functions as a pattern recognition receptor for diverse bacterial products. J. Biol. Chem.274, 33419–33425

25 Hoebe, K., Georgel, P., Rutschmann, S., Du, X., Mudd, S., Crozat, K., Sovath, S.,Shamel, L., Hartung, T., Zahringer, U. and Beutler, B. (2005) CD36 is a sensor ofdiacylglycerides. Nature 433, 523–527

26 Aliprantis, A. O., Yang, R. B., Mark, M. R., Suggett, S., Devaux, B., Radolf, J. D., Klimpel,G. R., Godowski, P. and Zychlinsky, A. (1999) Cell activation and apoptosis by bacteriallipoproteins through toll-like receptor-2. Science 285, 736–739

27 Hirschfeld, M., Kirschning, C. J., Schwandner, R., Wesche, H., Weis, J. H., Wooten,R. M. and Weis, J. J. (1999) Cutting edge: inflammatory signalling by Borreliaburgdorferi lipoproteins is mediated by toll-like receptor 2. J. Immunol. 163,2382–2386

28 Hasebe, A., Mu, H. H., Washburn, L. R., Chan, F. V., Pennock, N. D., Taylor, M. L. andCole, B. C. (2007) Inflammatory lipoproteins purified from a toxigenic and arthritogenicstrain of Mycoplasma arthritidis are dependent on Toll-like receptor 2 and CD14. InfectImmun. 75, 1820–1826

29 Lopez, M., Sly, L. M., Luu, Y., Young, D., Cooper, H. and Reiner, N. E. (2003) The 19-kDaMycobacterium tuberculosis protein induces macrophage apoptosis through Toll-likereceptor-2. J. Immunol. 170, 2409–2416

30 Dziarski, R. and Gupta, D. (2005) Staphylococcus aureus peptidoglycan is a Toll-likereceptor 2 activator: a reevaluation. Infect. Immun. 73, 5212–5216

31 Ozinsky, A., Underhill, D. M., Fontenot, J. D., Hajjar, A. M., Smith, K. D., Wilson, C. B.,Schroeder, L. and Aderem, A. (2000) The repertoire for pattern recognition of pathogensby the innate immune system is defined by cooperation between toll-like receptors.Proc. Natl. Acad. Sci. U.S.A. 97, 13766–13771

32 Schwandner, R., Dziarski, R., Wesche, H., Rothe, M. and Kirschning, C. J. (1999)Peptidoglycan- and lipoteichoic acid-induced cell activation is mediated by Toll-likereceptor 2. J. Biol. Chem. 274, 17406–17409

33 Levitz, S. M. (2004) Interactions of Toll-like receptors with fungi. Microbes Infect. 6,1351–1355

c© The Authors Journal compilation c© 2009 Biochemical Society

Page 13: REVIEW ARTICLE Pathogen recognition in the innate immune ... · Key words: innate immune response, Nod-like receptor (NLR), pathogen recognition, retinoic acid-inducible gene-I receptor

Pathogen recognition in the innate immune response 13

34 Frasnelli, M. E., Tarussio, D., Chobaz-Peclat, V., Busso, N. and So, A. (2005) TLR2modulates inflammation in zymosan-induced arthritis in mice. Arthritis Res. Ther. 7,R370–R379

35 Zahringer, U., Lindner, B., Inamura, S., Heine, H. and Alexander, C. (2008) TLR2 –promiscuous or specific? A critical re-evaluation of a receptor expressing apparentbroad specificity. Immunobiology 213, 205–224

36 Netea, M. G., Kullberg, B. J., Galama, J. M., Stalenhoef, A. F., Dinarello, C. A. and Vander Meer, J. W. (2002) Non-LPS components of Chlamydia pneumoniae stimulatecytokine production through Toll-like receptor 2-dependent pathways. Eur. J. Immunol.32, 1188–1195

37 Kurt-Jones, E. A., Chan, M., Zhou, S., Wang, J., Reed, G., Bronson, R., Arnold, M. M.,Knipe, D. M. and Finberg, R. W. (2004) Herpes simplex virus 1 interaction with Toll-likereceptor 2 contributes to lethal encephalitis. Proc. Natl. Acad. Sci. U.S.A. 101,1315–1320

38 Wang, J. P., Kurt-Jones, E. A., Shin, O. S., Manchak, M. D., Levin, M. J. and Finberg,R. W. (2005) Varicella-zoster virus activates inflammatory cytokines in humanmonocytes and macrophages via Toll-like receptor 2. J. Virol. 79, 12658- 12666

39 Takeuchi, O., Sato, S., Horiuchi, T., Hoshino, K., Takeda, K., Dong, Z., Modlin, R. L. andAkira, S. (2002) Cutting edge: role of Toll-like receptor 1 in mediating immune responseto microbial lipoproteins. J. Immunol. 169, 10–14

40 Takeuchi, O., Kawai, T., Muhlradt, P. F., Morr, M., Radolf, J. D., Zychlinsky, A., Takeda, K.and Akira, S. (2001) Discrimination of bacterial lipoproteins by Toll-like receptor 6. Int.Immunol. 13, 933–940

41 Gantner, B. N., Simmons, R. M., Canavera, S. J., Akira, S. and Underhill, D. M. (2003)Collaborative induction of inflammatory responses by dectin-1 and Toll-like receptor 2.J. Exp. Med. 197, 1107–1117

42 Gazzinelli, R. T., Ropert, C. and Campos, M. A. (2004) Role of the Toll/interleukin-1receptor signalling pathway in host resistance and pathogenesis during infection withprotozoan parasites. Immunol. Rev. 201, 9–25

43 Kurt-Jones, E. A., Popova, L., Kwinn, L., Haynes, L. M., Jones, L. P., Tripp, R. A., Walsh,E. E., Freeman, M. W., Golenbock, D. T., Anderson, L. J. and Finberg, R. W. (2000)Pattern recognition receptors TLR4 and CD14 mediate response to respiratory syncytialvirus. Nat. Immunol. 1, 398–401

44 Burzyn, D., Rassa, J. C., Kim, D., Nepomnaschy, I., Ross, S. R. and Piazzon, I. (2004)Toll-like receptor 4-dependent activation of dendritic cells by a retrovirus. J. Virol. 78,576–584

45 Byrd-Leifer, C. A., Block, E. F., Takeda, K., Akira, S. and Ding, A. (2001) The role ofMyD88 and TLR4 in the LPS-mimetic activity of taxol. Eur. J. Immunol. 31,2448–2457

46 Kawasaki, K., Akashi, S., Shimazu, R., Yoshida, T., Miyake, K. and Nishijima, M. (2000)Mouse toll-like receptor 4·MD-2 complex mediates lipopolysaccharide-mimetic signaltransduction by taxol. J. Biol. Chem. 275, 2251–2254

47 Takeda, K. and Akira, S. (2005) Toll-like receptors in innate immunity. Int. Immunol. 17,1–14

48 Lu, Y. C., Yeh, W. C. and Ohashi, P. S. (2008) LPS/TLR4 signal transduction pathway.Cytokine 42, 145–151

49 Hayashi, F., Smith, K. D., Ozinsky, A., Hawn, T. R., Yi, E. C., Goodlett, D. R., Eng, J. K.,Akira, S., Underhill, D. M. and Aderem, A. (2001) The innate immune response tobacterial flagellin is mediated by Toll-like receptor 5. Nature 410, 1099–1103

50 Butler, S. M. and Camilli, A. (2005) Going against the grain: chemotaxis and infection inVibrio cholerae. Nat. Rev. Microbiol. 3, 611–620

51 Beatson, S. A., Minamino, T. and Pallen, M. J. (2006) Variation in bacterial flagellins:from sequence to structure. Trends Microbiol. 14, 151–155

52 Jacchieri, S. G., Torquato, R. and Brentani, R. R. (2003) Structural study of binding offlagellin by Toll-like receptor 5. J. Bacteriol. 185, 4243–4247

53 Andersen-Nissen, E., Smith, K. D., Strobe, K. L., Barrett, S. L., Cookson, B. T., Logan,S. M. and Aderem, A. (2005) Evasion of Toll-like receptor 5 by flagellated bacteria. Proc.Natl. Acad. Sci. U.S.A. 102, 9247–9252

54 Tseng, J., Do, J., Widdicombe, J. H. and Machen, T. E. (2006) Innate immune responsesof human tracheal epithelium to Pseudomonas aeruginosa flagellin, TNF-α, and IL-1β .Am. J. Physiol. Cell Physiol. 290, C678–C690

55 Zhang, Z., Louboutin, J. P., Weiner, D. J., Goldberg, J. B. and Wilson, J. M. (2005)Human airway epithelial cells sense Pseudomonas aeruginosa infection via recognitionof flagellin by Toll-like receptor 5. Infect. Immun. 73, 7151–7160

56 Liaudet, L., Szabo, C., Evgenov, O. V., Murthy, K. G., Pacher, P., Virag, L., Mabley, J. G.,Marton, A., Soriano, F. G., Kirov, M. Y. et al. (2003) Flagellin from Gram-negativebacteria is a potent mediator of acute pulmonary inflammation in sepsis. Shock 19,131–137

57 Lopez-Boado, Y. S., Wilson, C. L. and Parks, W. C. (2001) Regulation of matrilysinexpression in airway epithelial cells by Pseudomonas aeruginosa flagellin. J. Biol.Chem. 276, 41417–41423

58 Muir, A., Soong, G., Sokol, S., Reddy, B., Gomez, M. I., Van Heeckeren, A. and Prince, A.(2004) Toll-like receptors in normal and cystic fibrosis airway epithelial cells. Am. J.Respir. Cell Mol. Biol. 30, 777–783

59 Uematsu, S., Fujimoto, K., Jang, M. H., Yang, B. G., Jung, Y. J., Nishiyama, M., Sato, S.,Tsujimura, T., Yamamoto, M., Yokota, Y. et al. (2008) Regulation of humoral and cellulargut immunity by lamina propria dendritic cells expressing Toll-like receptor 5. Nat.Immunol. 9, 769–776

60 Yarovinsky, F., Zhang, D., Andersen, J. F., Bannenberg, G. L., Serhan, C. N., Hayden,M. S., Hieny, S., Sutterwala, F. S., Flavell, R. A., Ghosh, S. and Sher, A. (2005) TLR11activation of dendritic cells by a protozoan profilin-like protein. Science 308,1626–1629

61 Zhang, D., Zhang, G., Hayden, M. S., Greenblatt, M. B., Bussey, C., Flavell, R. A. andGhosh, S. (2004) A toll-like receptor that prevents infection by uropathogenic bacteria.Science 303, 1522–1526

62 Lauw, F. N., Caffrey, D. R. and Golenbock, D. T. (2005) Of mice and man: TLR11 (finally)finds profilin. Trends Immunol. 26, 509–511

63 Alexopoulou, L., Holt, A. C., Medzhitov, R. and Flavell, R. A. (2001) Recognition ofdouble-stranded RNA and activation of NF-κB by Toll-like receptor 3. Nature 413,732–738

64 Wang, T., Town, T., Alexopoulou, L., Anderson, J. F., Fikrig, E. and Flavell, R. A. (2004)Toll-like receptor 3 mediates West Nile virus entry into the brain causing lethalencephalitis. Nat. Med. 10, 1366–1373

65 Groskreutz, D. J., Monick, M. M., Powers, L. S., Yarovinsky, T. O., Look, D. C. andHunninghake, G. W. (2006) Respiratory syncytial virus induces TLR3 protein and proteinkinase R, leading to increased double-stranded RNA responsiveness in airway epithelialcells. J. Immunol. 176, 1733–1740

66 Hardarson, H. S., Baker, J. S., Yang, Z., Purevjav, E., Huang, C. H., Alexopoulou, L., Li,N., Flavell, R. A., Bowles, N. E. and Vallejo, J. G. (2007) Toll-like receptor 3 is anessential component of the innate stress response in virus-induced cardiac injury. Am.J. Physiol. Heart Circ. Physiol. 292, H251–H258

67 Lafon, M., Megret, F., Lafage, M. and Prehaud, C. (2006) The innate immune facet ofbrain: human neurons express TLR-3 and sense viral dsRNA. J. Mol. Neurosci. 29,185–194

68 Edelmann, K. H., Richardson-Burns, S., Alexopoulou, L., Tyler, K. L., Flavell, R. A. andOldstone, M. B. (2004) Does Toll-like receptor 3 play a biological role in virusinfections? Virology 322, 231–238

69 Daffis, S., Samuel, M. A., Suthar, M. S., Gale, Jr., M. and Diamond, M. S. (2008)Toll-like receptor 3 has a protective role against West Nile virus infection. J. Virol. 82,10349–10358

70 Hemmi, H., Kaisho, T., Takeuchi, O., Sato, S., Sanjo, H., Hoshino, K., Horiuchi, T.,Tomizawa, H., Takeda, K. and Akira, S. (2002) Small anti-viral compounds activateimmune cells via the TLR7/MyD88-dependent signalling pathway. Nat. Immunol. 3,196–200

71 Heil, F., Hemmi, H., Hochrein, H., Ampenberger, F., Kirschning, C., Akira, S., Lipford, G.,Wagner, H. and Bauer, S. (2004) Species-specific recognition of single- stranded RNAvia Toll-like receptor 7 and 8. Science 303, 1526–1529

72 Diebold, S. S., Kaisho, T., Hemmi, H., Akira, S. and Reis e Sousa, C. (2004) Innateantiviral responses by means of TLR7-mediated recognition of single-stranded RNA.Science 303, 1529–1531

73 Hemmi, H., Takeuchi, O., Kawai, T., Kaisho, T., Sato, S., Sanjo, H., Matsumoto, M.,Hoshino, K., Wagner, H., Takeda, K. and Akira, S. (2000) A Toll-like receptor recognizesbacterial DNA. Nature 408, 740–745

74 Lund, J., Sato, A., Akira, S., Medzhitov, R. and Iwasaki, A. (2003) Toll-like receptor9-mediated recognition of Herpes simplex virus-2 by plasmacytoid dendritic cells.J. Exp. Med. 198, 513–520

75 Hochrein, H., Schlatter, B., O’Keeffe, M., Wagner, C., Schmitz, F., Schiemann, M., Bauer,S., Suter, M. and Wagner, H. (2004) Herpes simplex virus type-1 induces IFN-αproduction via Toll-like receptor 9-dependent and -independent pathways. Proc. Natl.Acad. Sci. U.S.A. 101, 11416–11421

76 Tabeta, K., Georgel, P., Janssen, E., Du, X., Hoebe, K., Crozat, K., Mudd, S., Shamel, L.,Sovath, S., Goode, J. et al. (2004) Toll-like receptors 9 and 3 as essential components ofinnate immune defense against mouse cytomegalovirus infection. Proc. Natl. Acad. Sci.U.S.A. 101, 3516–3521

77 Krug, A., Luker, G. D., Barchet, W., Leib, D. A., Akira, S. and Colonna, M. (2004) Herpessimplex virus type 1 activates murine natural interferon-producing cells through toll-likereceptor 9. Blood 103, 1433–1437

78 Krug, A., Rothenfusser, S., Hornung, V., Jahrsdorfer, B., Blackwell, S., Ballas, Z. K.,Endres, S., Krieg, A. M. and Hartmann, G. (2001) Identification of CpG oligonucleotidesequences with high induction of IFN-β/β in plasmacytoid dendritic cells. Eur. J.Immunol. 31, 2154–2163

79 Verthelyi, D., Ishii, K. J., Gursel, M., Takeshita, F. and Klinman, D. M. (2001) Humanperipheral blood cells differentially recognize and respond to two distinct CPG motifs.J. Immunol. 166, 2372–2377

c© The Authors Journal compilation c© 2009 Biochemical Society

Page 14: REVIEW ARTICLE Pathogen recognition in the innate immune ... · Key words: innate immune response, Nod-like receptor (NLR), pathogen recognition, retinoic acid-inducible gene-I receptor

14 H. Kumar, T. Kawai and S. Akira

80 Coban, C., Ishii, K. J., Kawai, T., Hemmi, H., Sato, S., Uematsu, S., Yamamoto, M.,Takeuchi, O., Itagaki, S., Kumar, N. et al. (2005) Toll-like receptor 9 mediates innateimmune activation by the malaria pigment hemozoin. J. Exp. Med. 201, 19–25

81 Asagiri, M., Hirai, T., Kunigami, T., Kamano, S., Gober, H. J., Okamoto, K., Nishikawa,K., Latz, E., Golenbock, D. T., Aoki, K. et al. (2008) Cathepsin K-dependent Toll-likereceptor 9 signalling revealed in experimental arthritis. Science 319, 624–627

82 Matsumoto, F., Saitoh, S., Fukui, R., Kobayashi, T., Tanimura, N., Konno, K., Kusumoto,Y., Akashi-Takamura, S. and Miyake, K. (2008) Cathepsins are required for Toll-likereceptor 9 responses. Biochem. Biophys. Res. Commun. 367, 693–699

83 Park, B., Brinkmann, M. M., Spooner, E., Lee, C. C., Kim, Y. M. and Ploegh, H. L. (2008)Proteolytic cleavage in an endolysosomal compartment is required for activation ofToll-like receptor 9. Nat. Immunol. 9, 1407–1414

84 West, A. P., Koblansky, A. A. and Ghosh, S. (2006) Recognition and signalling byToll-like receptors. Annu. Rev. Cell Dev. Biol. 22, 409–437

85 Kawai, T. and Akira, S. (2007) Signalling to NF-κB by Toll-like receptors. Trends Mol.Med. 13, 460–469

86 Kawagoe, T., Sato, S., Matsushita, K., Kato, H., Matsui, K., Kumagai, Y., Saitoh, T.,Kawai, T., Takeuchi, O. and Akira, S. (2008) Sequential control of Toll-likereceptor-dependent responses by IRAK1 and IRAK2. Nat. Immunol. 9, 684–691

87 Adhikari, A., Xu, M. and Chen, Z. J. (2007) Ubiquitin-mediated activation of TAK1 andIKK. Oncogene 26, 3214–3226

88 Chen, F., Bhatia, D., Chang, Q. and Castranova, V. (2006) Finding NEMO by K63-linkedpolyubiquitin chain. Cell Death Differ. 13, 1835–1838

89 Shim, J. H., Xiao, C., Paschal, A. E., Bailey, S. T., Rao, P., Hayden, M. S., Lee, K. Y.,Bussey, C., Steckel, M., Tanaka, N. et al. (2005) TAK1, but not TAB1 or TAB2, plays anessential role in multiple signalling pathways in vivo. Genes Dev. 19, 2668–2681

90 Sato, S., Sanjo, H., Takeda, K., Ninomiya-Tsuji, J., Yamamoto, M., Kawai, T., Matsumoto,K., Takeuchi, O. and Akira, S. (2005) Essential function for the kinase TAK1 in innate andadaptive immune responses. Nat. Immunol. 6, 1087–1095

91 Imler, J. L. and Hoffmann, J. A. (2003) Toll signalling: the TIReless quest for specificity.Nat. Immunol. 4, 105–106

92 Sato, S., Sugiyama, M., Yamamoto, M., Watanabe, Y., Kawai, T., Takeda, K. and Akira, S.(2003) Toll/IL-1 receptor domain-containing adaptor inducing IFN-β (TRIF) associateswith TNF receptor-associated factor 6 and TANK-binding kinase 1, and activates twodistinct transcription factors, NF-κB and IFN-regulatory factor-3, in the Toll-like receptorsignalling. J. Immunol. 171, 4304–4310

93 Meylan, E., Burns, K., Hofmann, K., Blancheteau, V., Martinon, F., Kelliher, M. andTschopp, J. (2004) RIP1 is an essential mediator of Toll-like receptor 3-induced NF-κBactivation. Nat. Immunol. 5, 503–507

94 Saitoh, T., Fujita, N., Jang, M. H., Uematsu, S., Yang, B. G., Satoh, T., Omori, H., Noda,T., Yamamoto, N., Komatsu, M. et al. (2008) Loss of the autophagy protein Atg16L1enhances endotoxin-induced IL-1β production. Nature 456, 264–268

95 Yamamoto, M., Sato, S., Hemmi, H., Hoshino, K., Kaisho, T., Sanjo, H., Takeuchi, O.,Sugiyama, M., Okabe, M., Takeda, K. and Akira, S. (2003) Role of adaptor TRIF in theMyD88-independent toll-like receptor signalling pathway. Science 301, 640–643

96 Sharma, S., tenOever, B. R., Grandvaux, N., Zhou, G. P., Lin, R. and Hiscott, J. (2003)Triggering the interferon antiviral response through an IKK-related pathway. Science300, 1148–1151

97 Fitzgerald, K. A., McWhirter, S. M., Faia, K. L., Rowe, D. C., Latz, E., Golenbock, D. T.,Coyle, A. J., Liao, S. M. and Maniatis, T. (2003) IKKε and TBK1 are essentialcomponents of the IRF3 signalling pathway. Nat. Immunol. 4, 491–496

98 Chau, T. L., Gioia, R., Gatot, J. S., Patrascu, F., Carpentier, I., Chapelle, J. P., O’Neill, L.,Beyaert, R., Piette, J. and Chariot, A. (2008) Are the IKKs and IKK-related kinases TBK1and IKK-ε similarly activated? Trends Biochem. Sci. 33, 171–180

99 Oganesyan, G., Saha, S. K., Guo, B., He, J. Q., Shahangian, A., Zarnegar, B., Perry, A.and Cheng, G. (2006) Critical role of TRAF3 in the Toll-like receptor-dependent and-independent antiviral response. Nature 439, 208–211

100 Hacker, H., Redecke, V., Blagoev, B., Kratchmarova, I., Hsu, L. C., Wang, G. G., Kamps,M. P., Raz, E., Wagner, H., Hacker, G. et al. (2006) Specificity in Toll-like receptorsignalling through distinct effector functions of TRAF3 and TRAF6. Nature 439, 204–207

101 Kawai, T., Sato, S., Ishii, K. J., Coban, C., Hemmi, H., Yamamoto, M., Terai, K., Matsuda,M., Inoue, J., Uematsu, S. et al. (2004) Interferon-α induction through Toll-likereceptors involves a direct interaction of IRF7 with MyD88 and TRAF6. Nat. Immunol. 5,1061–1068

102 Honda, K., Yanai, H., Mizutani, T., Negishi, H., Shimada, N., Suzuki, N., Ohba, Y.,Takaoka, A., Yeh, W. C. and Taniguchi, T. (2004) Role of a transductional–transcriptionalprocessor complex involving MyD88 and IRF-7 in Toll-like receptor signalling. Proc.Natl. Acad. Sci. U.S.A. 101, 15416–15421

103 Uematsu, S., Sato, S., Yamamoto, M., Hirotani, T., Kato, H., Takeshita, F., Matsuda, M.,Coban, C., Ishii, K. J., Kawai, T. et al. (2005) Interleukin-1 receptor-associated kinase-1plays an essential role for Toll-like receptor (TLR)7- and TLR9-mediated interferon-αinduction. J. Exp. Med. 201, 915–923

104 Hoshino, K., Sugiyama, T., Matsumoto, M., Tanaka, T., Saito, M., Hemmi, H., Ohara, O.,Akira, S. and Kaisho, T. (2006) IκB kinase-α is critical for interferon-α productioninduced by Toll-like receptors 7 and 9. Nature 440, 949–953

105 Cao, W., Manicassamy, S., Tang, H., Kasturi, S. P., Pirani, A., Murthy, N. and Pulendran,B. (2008) Toll-like receptor-mediated induction of type I interferon in plasmacytoiddendritic cells requires the rapamycin-sensitive PI(3)K–mTOR–p70S6K pathway. Nat.Immunol. 9, 1157–1164

106 Tabeta, K., Hoebe, K., Janssen, E. M., Du, X., Georgel, P., Crozat, K., Mudd, S., Mann,N., Sovath, S., Goode, J. et al. (2006) The Unc93b1 mutation 3d disrupts exogenousantigen presentation and signalling via Toll-like receptors 3, 7 and 9. Nat. Immunol. 7,156–164

107 Kim, Y. M., Brinkmann, M. M., Paquet, M. E. and Ploegh, H. L. (2008) UNC93B1delivers nucleotide-sensing Toll-like receptors to endolysosomes. Nature 452,234–238

108 Ewald, S. E., Lee, B. L., Lau, L., Wickliffe, K. E., Shi, G. P., Chapman, H. A. and Barton,G. M. (2008) The ectodomain of Toll-like receptor 9 is cleaved to generate a functionalreceptor. Nature 456, 658–662

109 Brinkmann, M. M., Spooner, E., Hoebe, K., Beutler, B., Ploegh, H. L. and Kim, Y. M.(2007) The interaction between the ER membrane protein UNC93B and TLR3, 7, and 9 iscrucial for TLR signalling. J. Cell Biol. 177, 265–275

110 Bustamante, J., Boisson-Dupuis, S., Jouanguy, E., Picard, C., Puel, A., Abel, L. andCasanova, J. L. (2008) Novel primary immunodeficiencies revealed by the investigationof paediatric infectious diseases. Curr. Opin. Immunol. 20, 39–48

111 von Bernuth, H., Picard, C., Jin, Z., Pankla, R., Xiao, H., Ku, C. L., Chrabieh, M.,Mustapha, I. B., Ghandil, P., Camcioglu, Y. et al. (2008) Pyogenic bacterial infections inhumans with MyD88 deficiency. Science 321, 691–696

112 Picard, C., Puel, A., Bonnet, M., Ku, C. L., Bustamante, J., Yang, K., Soudais, C.,Dupuis, S., Feinberg, J., Fieschi, C. et al. (2003) Pyogenic bacterial infections inhumans with IRAK-4 deficiency. Science 299, 2076–2079

113 Zhang, S. Y., Jouanguy, E., Ugolini, S., Smahi, A., Elain, G., Romero, P., Segal, D.,Sancho-Shimizu, V., Lorenzo, L., Puel, A. et al. (2007) TLR3 deficiency in patients withherpes simplex encephalitis. Science 317, 1522–1527

114 Casrouge, A., Zhang, S. Y., Eidenschenk, C., Jouanguy, E., Puel, A., Yang, K., Alcais, A.,Picard, C., Mahfoufi, N., Nicolas, N. et al. (2006) Herpes simplex virus encephalitis inhuman UNC-93B deficiency. Science 314, 308–312

115 Kawai, T. and Akira, S. (2006) Innate immune recognition of viral infection. Nat.Immunol. 7, 131–137

116 Ishii, K. J., Koyama, S., Nakagawa, A., Coban, C. and Akira, S. (2008) Host innateimmune receptors and beyond: making sense of microbial infections. Cell Host Microbe3, 352–363

117 Takeuchi, O. and Akira, S. (2008) MDA5/RIG-I and virus recognition. Curr. Opin.Immunol. 20, 17–22

118 Yoneyama, M. and Fujita, T. (2008) Structural mechanism of RNA recognition by theRIG-I-like receptors. Immunity 29, 178–181

119 Yoneyama, M., Kikuchi, M., Natsukawa, T., Shinobu, N., Imaizumi, T., Miyagishi, M.,Taira, K., Akira, S. and Fujita, T. (2004) The RNA helicase RIG-I has an essential functionin double-stranded RNA-induced innate antiviral responses. Nat. Immunol. 5, 730–737

120 Eisenacher, K., Steinberg, C., Reindl, W. and Krug, A. (2007) The role of viral nucleicacid recognition in dendritic cells for innate and adaptive antiviral immunity.Immunobiology 212, 701–714

121 Cui, S., Eisenacher, K., Kirchhofer, A., Brzozka, K., Lammens, A., Lammens, K., Fujita, T.,Conzelmann, K. K., Krug, A. and Hopfner, K. P. (2008) The C-terminal regulatory domainis the RNA 5′-triphosphate sensor of RIG-I. Mol. Cell 29, 169–179

122 Takahasi, K., Yoneyama, M., Nishihori, T., Hirai, R., Kumeta, H., Narita, R., Gale, Jr., M.,Inagaki, F. and Fujita, T. (2008) Nonself RNA-sensing mechanism of RIG-I helicase andactivation of antiviral immune responses. Mol. Cell. 29, 428–440

123 Diao, F., Li, S., Tian, Y., Zhang, M., Xu, L. G., Zhang, Y., Wang, R. P., Chen, D., Zhai, Z.,Zhong, B., Tien, P. and Shu, H. B. (2007) Negative regulation of MDA5- but notRIG-I-mediated innate antiviral signalling by the dihydroxyacetone kinase. Proc. Natl.Acad. Sci. U.S.A. 104, 11706–11711

124 Venkataraman, T., Valdes, M., Elsby, R., Kakuta, S., Caceres, G., Saijo, S., Iwakura, Y.and Barber, G. N. (2007) Loss of DExD/H box RNA helicase LGP2 manifests disparateantiviral responses. J. Immunol. 178, 6444–6455

125 Deddouche, S., Matt, N., Budd, A., Mueller, S., Kemp, C., Galiana-Arnoux, D., Dostert,C., Antoniewski, C., Hoffmann, J. A. and Imler, J. L. (2008) The DExD/H-box helicaseDicer-2 mediates the induction of antiviral activity in Drosophila. Nat. Immunol. 9,1425–1432

126 Kato, H., Sato, S., Yoneyama, M., Yamamoto, M., Uematsu, S., Matsui, K., Tsujimura, T.,Takeda, K., Fujita, T., Takeuchi, O. and Akira, S. (2005) Cell type-specific involvement ofRIG-I in antiviral response. Immunity 23, 19–28

127 Loo, Y. M., Fornek, J., Crochet, N., Bajwa, G., Perwitasari, O., Martinez-Sobrido, L.,Akira, S., Gill, M. A., Garcia-Sastre, A., Katze, M. G. and Gale, Jr., M. (2008) DistinctRIG-I and MDA5 signalling by RNA viruses in innate immunity. J. Virol. 82, 335–345

c© The Authors Journal compilation c© 2009 Biochemical Society

Page 15: REVIEW ARTICLE Pathogen recognition in the innate immune ... · Key words: innate immune response, Nod-like receptor (NLR), pathogen recognition, retinoic acid-inducible gene-I receptor

Pathogen recognition in the innate immune response 15

128 Kato, H., Takeuchi, O., Sato, S., Yoneyama, M., Yamamoto, M., Matsui, K., Uematsu, S.,Jung, A., Kawai, T., Ishii, K. J. et al. (2006) Differential roles of MDA5 and RIG-Ihelicases in the recognition of RNA viruses. Nature 441, 101–105

129 Saito, T., Owen, D. M., Jiang, F., Marcotrigiano, J. and Gale, Jr., M. (2008) Innateimmunity induced by composition-dependent RIG-I recognition of hepatitis C virusRNA. Nature 454, 523–527

130 Jung, A., Kato, H., Kumagai, Y., Kumar, H., Kawai, T., Takeuchi, O. and Akira, S. (2008)Lymphocytoid choriomeningitis virus activates plasmacytoid dendritic cells and inducesa cytotoxic T-cell response via MyD88. J. Virol. 82, 196–206

131 Koyama, S., Ishii, K. J., Kumar, H., Tanimoto, T., Coban, C., Uematsu, S., Kawai, T. andAkira, S. (2007) Differential role of TLR- and RLR-signalling in the immune responses toinfluenza A virus infection and vaccination. J. Immunol. 179, 4711–4720

132 Kumagai, Y., Takeuchi, O., Kato, H., Kumar, H., Matsui, K., Morii, E., Aozasa, K., Kawai,T. and Akira, S. (2007) Alveolar macrophages are the primary interferon-α producer inpulmonary infection with RNA viruses. Immunity 27, 240–252

133 Bhoj, V. G., Sun, Q., Bhoj, E. J., Somers, C., Chen, X., Torres, J. P., Mejias, A., Gomez,A. M., Jafri, H., Ramilo, O. and Chen, Z. J. (2008) MAVS and MyD88 are essential forinnate immunity but not cytotoxic T lymphocyte response against respiratory syncytialvirus. Proc. Natl. Acad. Sci. U.S.A. 105, 14046–14051

134 Kumar, H., Koyama, S., Ishii, K. J., Kawai, T. and Akira, S. (2008) Cutting edge:cooperation of IPS-1- and TRIF-dependent pathways in polyIC-enhanced antibodyproduction and cytotoxic T cell responses. J. Immunol. 180, 683–687

135 Hornung, V., Ellegast, J., Kim, S., Brzozka, K., Jung, A., Kato, H., Poeck, H., Akira, S.,Conzelmann, K. K., Schlee, M. et al. (2006) 5′-Triphosphate RNA is the ligand for RIG-I.Science 314, 994–997

136 Pichlmair, A., Schulz, O., Tan, C. P., Naslund, T. I., Liljestrom, P., Weber, F. and Reis eSousa, C. (2006) RIG-I-mediated antiviral responses to single-stranded RNA bearing5′-phosphates. Science 314, 997–1001

137 Yoneyama, M., Onomoto, K. and Fujita, T. (2008) Cytoplasmic recognition of RNA. Adv.Drug Deliv. Rev. 60, 841–846

138 Kato, H., Takeuchi, O., Mikamo-Satoh, E., Hirai, R., Kawai, T., Matsushita, K., Hiiragi, A.,Dermody, T. S., Fujita, T. and Akira, S. (2008) Length-dependent recognition ofdouble-stranded ribonucleic acids by retinoic acid-inducible gene-I and melanomadifferentiation-associated gene 5. J. Exp. Med. 205, 1601–1610

139 Kawai, T., Takahashi, K., Sato, S., Coban, C., Kumar, H., Kato, H., Ishii, K. J., Takeuchi,O. and Akira, S. (2005) IPS-1, an adaptor triggering RIG-I- and Mda5-mediated type Iinterferon induction. Nat. Immunol. 6, 981–988

140 Seth, R. B., Sun, L., Ea, C. K. and Chen, Z. J. (2005) Identification and characterization ofMAVS, a mitochondrial antiviral signalling protein that activates NF-κB and IRF 3. Cell122, 669–682

141 Meylan, E., Curran, J., Hofmann, K., Moradpour, D., Binder, M., Bartenschlager, R. andTschopp, J. (2005) Cardif is an adaptor protein in the RIG-I antiviral pathway and istargeted by hepatitis C virus. Nature 437, 1167–1172

142 Xu, L. G., Wang, Y. Y., Han, K. J., Li, L. Y., Zhai, Z. and Shu, H. B. (2005) VISA is anadapter protein required for virus-triggered IFN-β signalling. Mol. Cell. 19,727–740

143 Kumar, H., Kawai, T., Kato, H., Sato, S., Takahashi, K., Coban, C., Yamamoto, M.,Uematsu, S., Ishii, K. J., Takeuchi, O. and Akira, S. (2006) Essential role of IPS-1 ininnate immune responses against RNA viruses. J. Exp. Med. 203, 1795–1803

144 Sun, Q., Sun, L., Liu, H. H., Chen, X., Seth, R. B., Forman, J. and Chen, Z. J. (2006) Thespecific and essential role of MAVS in antiviral innate immune responses. Immunity 24,633–642

145 Moore, C. B., Bergstralh, D. T., Duncan, J. A., Lei, Y., Morrison, T. E., Zimmermann,A. G., Accavitti-Loper, M. A., Madden, V. J., Sun, L., Ye, Z. et al. (2008) NLRX1 is aregulator of mitochondrial antiviral immunity. Nature. 451, 573–577

146 Gack, M. U., Shin, Y. C., Joo, C. H., Urano, T., Liang, C., Sun, L., Takeuchi, O., Akira, S.,Chen, Z., Inoue, S. and Jung, J. U. (2007) TRIM25 RING-finger E3 ubiquitin ligase isessential for RIG-I-mediated antiviral activity. Nature 446, 916–920

147 Arimoto, K., Takahashi, H., Hishiki, T., Konishi, H., Fujita, T. and Shimotohno, K. (2007)Negative regulation of the RIG-I signalling by the ubiquitin ligase RNF125. Proc. Natl.Acad. Sci. U.S.A. 104, 7500–7505

148 Saha, S. K., Pietras, E. M., He, J. Q., Kang, J. R., Liu, S. Y., Oganesyan, G., Shahangian,A., Zarnegar, B., Shiba, T. L., Wang, Y. and Cheng, G. (2006) Regulation of antiviralresponses by a direct and specific interaction between TRAF3 and Cardif. EMBO J. 25,3257–3263

149 Kayagaki, N., Phung, Q., Chan, S., Chaudhari, R., Quan, C., O’Rourke, K. M., Eby, M.,Pietras, E., Cheng, G., Bazan, J. F. et al. (2007) DUBA: a deubiquitinase that regulatestype I interferon production. Science 318, 1628–1632

150 Takahashi, K., Kawai, T., Kumar, H., Sato, S., Yonehara, S. and Akira, S. (2006) Roles ofcaspase-8 and caspase-10 in innate immune responses to double-stranded RNA.J. Immunol. 176, 4520–4524

151 Michallet, M. C., Meylan, E., Ermolaeva, M. A., Vazquez, J., Rebsamen, M., Curran, J.,Poeck, H., Bscheider, M., Hartmann, G., Konig, M. et al. (2008) TRADD protein is anessential component of the RIG-like helicase antiviral pathway. Immunity 28, 651–661

152 Ishikawa, H. and Barber, G. N. (2008) STING is an endoplasmic reticulum adaptor thatfacilitates innate immune signalling. Nature 455, 674–678

153 Zhong, B., Yang, Y., Li, S., Wang, Y. Y., Li, Y., Diao, F., Lei, C., He, X., Zhang, L., Tien, P.and Shu, H. B. (2008) The adaptor protein MITA links virus-sensing receptors to IRF3transcription factor activation. Immunity 29, 538–550

154 Jounai, N., Takeshita, F., Kobiyama, K., Sawano, A., Miyawaki, A., Xin, K. Q., Ishii, K. J.,Kawai, T., Akira, S., Suzuki, K. and Okuda, K. (2007) The Atg5–Atg12 conjugateassociates with innate antiviral immune responses. Proc. Natl. Acad. Sci. U.S.A. 104,14050–14055

155 Lee, H. K., Lund, J. M., Ramanathan, B., Mizushima, N. and Iwasaki, A. (2007)Autophagy-dependent viral recognition by plasmacytoid dendritic cells. Science 315,1398–1401

156 Ishii, K. J., Coban, C., Kato, H., Takahashi, K., Torii, Y., Takeshita, F., Ludwig, H., Sutter,G., Suzuki, K. and Hemmi, H. (2006) A Toll-like receptor-independent antiviral responseinduced by double-stranded B-form DNA. Nat. Immunol. 7, 40–48

157 Stetson, D. B. and Medzhitov, R. (2006) Recognition of cytosolic DNA activates anIRF3-dependent innate immune response. Immunity 24, 93–103

158 Takaoka, A., Wang, Z., Choi, M. K., Yanai, H., Negishi, H., Ban, T., Lu, Y., Miyagishi, M.,Kodama, T., Honda, K. et al. (2007) DAI (DLM-1/ZBP1) is a cytosolic DNA sensor and anactivator of innate immune response. Nature 448, 501–505

159 Ishii, K. J., Kawagoe, T., Koyama, S., Matsui, K., Kumar, H., Kawai, T., Uematsu, S.,Takeuchi, O., Takeshita, F., Coban, C. and Akira, S. (2008) TANK-binding kinase-1delineates innate and adaptive immune responses to DNA vaccines. Nature 451,725–729

160 Ting, J. P. and Davis, B. K. (2005) CATERPILLER: a novel gene family important inimmunity, cell death, and diseases. Annu. Rev. Immunol. 23, 387–414

161 Shaw, M. H., Reimer, T., Kim, Y. G. and Nunez, G. (2008) NOD-like receptors (NLRs):bona fide intracellular microbial sensors. Curr. Opin. Immunol. 20, 377–382

162 Ting, J. P., Willingham, S. B. and Bergstralh, D. T. (2008) NLRs at the intersection of celldeath and immunity. Nat. Rev. Immunol. 8, 372–379

163 Kanneganti, T. D., Lamkanfi, M. and Nunez, G. (2007) Intracellular NOD-like receptors inhost defense and disease. Immunity 27, 549–559

164 Stojanov, S. and Kastner, D. L. (2005) Familial autoinflammatory diseases: genetics,pathogenesis and treatment. Curr. Opin. Rheumatol. 17, 586–599

165 Hoffman, H. M., Wanderer, A. A. and Broide, D. H. (2001) Familial coldautoinflammatory syndrome: phenotype and genotype of an autosomal dominantperiodic fever. J. Allergy Clin. Immunol. 108, 615–620

166 Girardin, S. E., Hugot, J. P. and Sansonetti, P. J. (2003) Lessons from Nod2 studies:towards a link between Crohn’s disease and bacterial sensing. Trends Immunol. 24,652–658

167 Chamaillard, M., Hashimoto, M., Horie, Y., Masumoto, J., Qiu, S., Saab, L., Ogura, Y.,Kawasaki, A., Fukase, K., Kusumoto, S. et al. (2003) An essential role for NOD1 in hostrecognition of bacterial peptidoglycan containing diaminopimelic acid. Nat. Immunol. 4,702–707

168 Girardin, S. E., Boneca, I. G., Carneiro, L. A., Antignac, A., Jehanno, M., Viala, J., Tedin,K., Taha, M. K., Labigne, A., Zahringer, U. et al. (2003) Nod1 detects a uniquemuropeptide from Gram-negative bacterial peptidoglycan. Science. 300, 1584–1587

169 Girardin, S. E., Boneca, I. G., Viala, J., Chamaillard, M., Labigne, A., Thomas, G.,Philpott, D. J. and Sansonetti, P. J. (2003) Nod2 is a general sensor of peptidoglycanthrough muramyl dipeptide (MDP) detection. J. Biol. Chem. 278, 8869–8872

170 Inohara, N., Koseki, T., del Peso, L., Hu, Y., Yee, C., Chen, S., Carrio, R., Merino, J., Liu,D., Ni, J. and Nunez, G. (1999) Nod1, an Apaf-1-like activator of caspase-9 and nuclearfactor-κB. J. Biol. Chem. 274, 14560–14567

171 Ogura, Y., Inohara, N., Benito, A., Chen, F. F., Yamaoka, S. and Nunez, G. (2001) Nod2, aNod1/Apaf-1 family member that is restricted to monocytes and activates NF-κB. J. Biol.Chem. 276, 4812–4818

172 Peyrin-Biroulet, L., Vignal, C., Dessein, R., Simonet, M., Desreumaux, P. andChamaillard, M. (2006) NODs in defence: from vulnerable antimicrobial peptides tochronic inflammation. Trends Microbiol. 14, 432–438

173 Park, J. H., Kim, Y. G., Shaw, M., Kanneganti, T. D., Fujimoto, Y., Fukase, K., Inohara, N.and Nunez, G. (2007) Nod1/RICK and TLR signalling regulate chemokine andantimicrobial innate immune responses in mesothelial cells. J. Immunol. 179, 514–521

174 Hsu, Y. M., Zhang, Y., You, Y., Wang, D., Li, H., Duramad, O., Qin, X. F., Dong, C. andLin, X. (2007) The adaptor protein CARD9 is required for innate immune responses tointracellular pathogens. Nat. Immunol. 8, 198–205

175 Kim, J. G., Lee, S. J. and Kagnoff, M. F. (2004) Nod1 is an essential signal transducer inintestinal epithelial cells infected with bacteria that avoid recognition by toll-likereceptors. Infect. Immun. 72, 1487–1495

c© The Authors Journal compilation c© 2009 Biochemical Society

Page 16: REVIEW ARTICLE Pathogen recognition in the innate immune ... · Key words: innate immune response, Nod-like receptor (NLR), pathogen recognition, retinoic acid-inducible gene-I receptor

16 H. Kumar, T. Kawai and S. Akira

176 Travassos, L. H., Carneiro, L. A., Girardin, S. E., Boneca, I. G., Lemos, R., Bozza, M. T.,Domingues, R. C., Coyle, A. J., Bertin, J., Philpott, D. J. and Plotkowski, M. C. (2005)Nod1 participates in the innate immune response to Pseudomonas aeruginosa. J. Biol.Chem. 280, 36714–36718

177 Opitz, B., Forster, S., Hocke, A. C., Maass, M., Schmeck, B., Hippenstiel, S., Suttorp, N.and Krull, M. (2005) Nod1-mediated endothelial cell activation by Chlamydophilapneumoniae. Circ. Res. 96, 319–326

178 Buchholz, K. R. and Stephens, R. S. (2008) The cytosolic pattern recognition receptorNOD1 induces inflammatory interleukin-8 during Chlamydia trachomatis infection.Infect Immun. 76, 3150–3155

179 Welter-Stahl, L., Ojcius, D. M., Viala, J., Girardin, S., Liu, W., Delarbre, C., Philpott, D.,Kelly, K. A. and Darville, T. (2006) Stimulation of the cytosolic receptor forpeptidoglycan, Nod1, by infection with Chlamydia trachomatis or Chlamydiamuridarum. Cell Microbiol. 8, 1047–1057

180 Zilbauer, M., Dorrell, N., Elmi, A., Lindley, K. J., Schuller, S., Jones, H. E., Klein, N. J.,Nunez, G., Wren, B. W. and Bajaj-Elliott, M. (2007) A major role for intestinal epithelialnucleotide oligomerization domain 1 (NOD1) in eliciting host bactericidal immuneresponses to Campylobacter jejuni. Cell Microbiol. 9, 2404–2416

181 Ratner, A. J., Aguilar, J. L., Shchepetov, M., Lysenko, E. S. and Weiser, J. N. (2007)Nod1mediates cytoplasmic sensing of combinations of extracellular bacteria. CellMicrobiol. 9, 1343–1351

182 Opitz, B., Puschel, A., Schmeck, B., Hocke, A. C., Rosseau, S., Hammerschmidt, S.,Schumann, R. R., Suttorp, N. and Hippenstiel, S. (2004) Nucleotide-bindingoligomerization domain proteins are innate immune receptors for internalizedStreptococcus pneumoniae. J. Biol. Chem. 279, 36426–36432

183 Ferwerda, G., Girardin, S. E., Kullberg, B. J., Le Bourhis, L., de Jong, D. J., Langenberg,D. M., van Crevel, R., Adema, G. J., Ottenhoff, T. H., Van der Meer, J. W. and Netea,M. G. (2005) NOD2 and toll-like receptors are nonredundant recognition systems ofMycobacterium tuberculosis. PLoS Pathog. 1, 279–285

184 Hasegawa, M., Yang, K., Hashimoto, M., Park, J. H., Kim, Y. G., Fujimoto, Y., Nunez, G.,Fukase, K. and Inohara, N. (2006) Differential release and distribution of Nod1 and Nod2immunostimulatory molecules among bacterial species and environments. J. Biol.Chem. 281, 29054–29063

185 Kobayashi, K. S., Chamaillard, M., Ogura, Y., Henegariu, O., Inohara, N., Nunez, G. andFlavell, R. A. (2005) Nod2-dependent regulation of innate and adaptive immunity in theintestinal tract. Science 307, 731–734

186 Viala, J., Chaput, C., Boneca, I. G., Cardona, A., Girardin, S. E., Moran, A. P., Athman,R., Memet, S., Huerre, M. R., Coyle, A. J. et al. (2004) Nod1 responds to peptidoglycandelivered by the Helicobacter pylori cag pathogenicity island. Nat. Immunol. 5,1166–1174

187 Petrilli, V., Dostert, C., Muruve, D. A. and Tschopp, J. (2007) The inflammasome: adanger sensing complex triggering innate immunity. Curr. Opin. Immunol. 19,615–622

188 Li, H., Nookala, S. and Re, F. (2007) Aluminum hydroxide adjuvants activate caspase-1and induce IL-1β and IL-18 release. J. Immunol. 178, 5271–5276

189 Eisenbarth, S. C., Colegio, O. R., O’Connor, W., Sutterwala, F. S. and Flavell, R. A. (2008)Crucial role for the Nalp3 inflammasome in the immunostimulatory properties ofaluminium adjuvants. Nature 453, 1122–1126

190 Franchi, L. and Nunez, G. (2008) The Nlrp3 inflammasome is critical for aluminiumhydroxide-mediated IL-1β secretion but dispensable for adjuvant activity. Eur. J.Immunol. 38, 2085–2089

191 Halle, A., Hornung, V., Petzold, G. C., Stewart, C. R., Monks, B. G., Reinheckel, T.,Fitzgerald, K. A., Latz, E., Moore, K. J. and Golenbock, D. T. (2008) The NALP3inflammasome is involved in the innate immune response to amyloid-β . Nat. Immunol.9, 857–865

192 Dostert, C., Petrilli, V., Van Bruggen, R., Steele, C., Mossman, B. T. and Tschopp, J.(2008) Innate immune activation through Nalp3 inflammasome sensing of asbestos andsilica. Science 320, 674–677

193 Cassel, S. L., Eisenbarth, S. C., Iyer, S. S., Sadler, J. J., Colegio, O. R., Tephly, L. A.,Carter, A. B., Rothman, P. B., Flavell, R. A. and Sutterwala, F. S. (2008) The Nalp3inflammasome is essential for the development of silicosis. Proc. Natl. Acad. Sci. U.S.A.105, 9035–9040

194 Roberts, T. L., Idris, A., Dunn, J. A., Kelly, G. M., Burnton, C. M., Hodgson, S., Hardy,L. L., Garceau, V., Sweet, M. J., Ross, I. L. et al. (2009) HIN-200 proteins regulatecaspase activation in response to foreign cytoplasmic DNA, Science 323, 1057–1060

195 Hornung, V., Ablasser, A., Charrel-Dennis, M., Bauernfeind, F., Horvath, G., Caffrey,D. R., Latz, E. and Fitzgerald, K. A. (2009) AIM2 recognizes cytosolic dsDNA and forms acaspase-1-activating inflammasome with ASC, Nature 458, 514–518

196 Fernandes-Alnemri, T., Yu, J. W., Datta, P., Wu, J. and Alnemri, E. S. (2009) AIM2activates the inflammasome and cell death in response to cytoplasmic DNA, Nature 458,509–513

197 Burckstummer, T., Baumann, C., Bluml, S., Dixit, E., Durnberger, G., Jahn, H.,Planyavsky, M., Bilban, M., Colinge, J., Bennett, K. L. and Superti-Furga, G. (2009) Anorthogonal proteomic-genomic screen identifies AIM2 as a cytoplasmic DNA sensor forthe inflammasome. Nat, Immunol. 10, 266–272

198 Hornung, V., Bauernfeind, F., Halle, A., Samstad, E. O., Kono, H., Rock, K. L.,Fitzgerald, K. A. and Latz, E. (2008) Silica crystals and aluminum salts activate theNALP3 inflammasome through phagosomal destabilization. Nat. Immunol. 9,847–856

199 Franchi, L., Amer, A., Body-Malapel, M., Kanneganti, T. D., Ozoren, N., Jagirdar, R.,Inohara, N., Vandenabeele, P., Bertin, J., Coyle, A. et al. (2006) Cytosolic flagellinrequires Ipaf for activation of caspase-1 and interleukin 1β in Salmonella-infectedmacrophages. Nat. Immunol. 7, 576–582

200 Amer, A., Franchi, L., Kanneganti, T. D., Body-Malapel, M., Ozoren, N., Brady, G.,Meshinchi, S., Jagirdar, R., Gewirtz, A., Akira, S. and Nunez, G. (2006) Regulation ofLegionella phagosome maturation and infection through flagellin and host Ipaf. J. Biol.Chem. 281, 35217–35223

201 Miao, E. A., Alpuche-Aranda, C. M., Dors, M., Clark, A. E., Bader, M. W., Miller, S. I. andAderem, A. (2006) Cytoplasmic flagellin activates caspase-1 and secretion of interleukin1β via Ipaf. Nat. Immunol. 7, 569–575

202 Miao, E. A., Ernst, R. K., Dors, M., Mao, D. P. and Aderem, A. (2008) Pseudomonasaeruginosa activates caspase 1 through Ipaf. Proc. Natl. Acad. Sci. U.S.A. 105,2562–2567

203 Wright, E. K., Goodart, S. A., Growney, J. D., Hadinoto, V., Endrizzi, M. G., Long, E. M.,Sadigh, K., Abney, A. L., Bernstein-Hanley, I. and Dietrich, W. F. (2003) Naip5 affectshost susceptibility to the intracellular pathogen Legionella pneumophila. Curr. Biol. 13,27–36

204 Diez, E., Lee, S. H., Gauthier, S., Yaraghi, Z., Tremblay, M., Vidal, S. and Gros, P. (2003)Birc1e is the gene within the Lgn1 locus associated with resistance to Legionellapneumophila. Nat. Genet. 33, 55–60

205 Lightfield, K. L., Persson, J., Brubaker, S. W., Witte, C. E., von Moltke, J., Dunipace,E. A., Henry, T., Sun, Y. H., Cado, D., Dietrich, W. F. et al. (2008) Critical function forNaip5 in inflammasome activation by a conserved carboxy-terminal domain of flagellin.Nat. Immunol. 9, 1171–1178

206 Ezzell, J. W., Ivins, B. E. and Leppla, S. H. (1984) Immunoelectrophoretic analysis,toxicity, and kinetics of in vitro production of the protective antigen and lethal factorcomponents of Bacillus anthracis toxin. Infect. Immun. 45, 761–767

207 Boyden, E. D. and Dietrich, W. F. (2006) Nalp1b controls mouse macrophagesusceptibility to anthrax lethal toxin. Nat. Genet. 38, 240–244

208 Hsu, L. C., Ali, S. R., McGillivray, S., Tseng, P. H., Mariathasan, S., Humke, E. W.,Eckmann, L., Powell, J. J., Nizet, V., Dixit, V. M. and Karin, M. (2008) ANOD2–NALP1complex mediates caspase-1-dependent IL-1β secretion in response toBacillus anthracis infection and muramyl dipeptide. Proc. Natl. Acad. Sci. U.S.A. 105,7803–7808

209 Bruey, J. M., Bruey-Sedano, N., Luciano, F., Zhai, D., Balpai, R., Xu, C., Kress, C. L.,Bailly-Maitre, B., Li, X., Osterman, A., Matsuzawa, S. et al. (2007) Bcl-2 and Bcl-XLregulate proinflammatory caspase-1 activation by interaction with NALP1. Cell 129,45–56

Received 16 February 2009/5 March 2009; accepted 6 March 2009Published on the Internet 28 April 2009, doi:10.1042/BJ20090272

c© The Authors Journal compilation c© 2009 Biochemical Society