Annurev.pathol NOD

download Annurev.pathol NOD

of 36

Transcript of Annurev.pathol NOD

  • 8/6/2019 Annurev.pathol NOD

    1/36

    NOD-Like Receptors:Role in Innate Immunityand Inflammatory Disease

    Grace Chen, Michael H. Shaw, Yun-Gi Kim,and Gabriel Nunez

    Departments of Pathology and Internal Medicine and the Comprehensive Cancer CenterUniversity of Michigan Medical School, Ann Arbor, Michigan 48109;email: [email protected]; [email protected]; [email protected]; [email protected]

    Annu. Rev. Pathol. Mech. Dis. 2009. 4:36598

    First published online as a Review in Advance onOctober 17, 2008

    The Annual Review of Pathology: Mechanisms ofDisease is online at pathmechdis.annualreviews.org

    This articles doi:10.1146/annurev.pathol.4.110807.092239

    Copyright c 2009 by Annual Reviews.All rights reserved

    1553-4006/09/0228-0365$20.00

    Key Words

    caspase-1, Crohns disease, IL-1, NOD2, NLRP3

    Abstract

    The NOD-like receptors (NLRs)are a specialized group of intracellulareceptorsthatrepresentakeycomponentofthehostinnateimmunesys

    tem. Since the discovery of the first NLR almost 10 years ago, the studof this special class of microbial sensors has burgeoned; consequently,

    better understanding of the mechanism by which these receptors recog

    nize microbes and other danger signals and of how they activate inflammatory signaling pathways has emerged. Moreover, in addition to theiprimary role in host defense against invading pathogens, their ability t

    regulate nuclear factorkappa B (NF-B) signaling, interleukin-1-bet(IL-1) production, and cell death indicates that they are crucial to th

    pathogenesis of a variety of inflammatory human diseases.

    365

  • 8/6/2019 Annurev.pathol NOD

    2/36

    PAMPs: pathogen-associated molecularpatterns

    PRRs: pathogenrecognition receptors

    LPS:lipopolysaccharide

    PGN: peptidoglycan

    TLRs: Toll-likereceptors

    INTRODUCTION

    Essential to human survival is the ability toeradicate pathogenic microorganisms. To en-

    sure the efficient detection and removal ofharmful microbes, two effector mechanisms

    have evolved: the innate and adaptive im-mune systems. The adaptive immune response

    is characterized by a delayed response involv-ing gene rearrangements for the clonal se-lection and expansion of T cell and B cell

    lymphocytes with antigen-specific receptors.This process results in the generation of a di-

    verse, yet specific repertoire of immune effec-tors that also contribute to immunologic mem-

    ory. The innate immune system, however, israpidly activated and does not require the so-

    matic gene rearrangements that form the cor-nerstone of adaptive immunity. It represents

    one of the first lines of defense against microor-ganisms, whereby conserved microbial struc-

    tures known as pathogen-associated molecularpatterns (PAMPs) are recognized by germline-

    encoded innate immune receptors, also known

    as pathogen recognition receptors (PRRs). Ex-amples of PAMPs include lipopolysaccharides

    (LPS), peptidoglycan (PGN), flagellin, and mi-crobial nucleic acids. Recognition of these

    PAMPs by PRRs results in the activation ofsignaling pathways, which promotes an inflam-

    matory, antimicrobial response. Emerging datahave also demonstrated a link between the in-

    nate PRRs and the activation of the adaptiveimmune response that can act in concert in de-

    fense against invasive organisms (1). Moreover,it has also become apparent that these recep-

    tors are involved in sensing not only invading

    pathogens, but also endogenous nonmicrobialdanger or stress signals, both of which re-

    sult in the activation of inflammatory signal-ing pathways such as nuclear factorkappa B

    (NF-B) and mitogen-activatedproteinkinases(MAPKs). This upstream link to complexeffec-

    tor pathways highlights the importance of theseimmune receptors in both microbial defense

    and the pathogenesis of noninfectious, inflam-matory diseases when signaling becomes dys-

    regulated.

    Three major classes of PRRs have b

    identified: (a) the Toll-like receptors (TLR which are transmembrane proteins with

    extramembranous domain involved in land recognition on either the extracellu

    surface or within endosomes and a cytopl

    mic domain involved in signal transducti(b) the NOD-like receptors (NLRs), which

    intracellular, cytoplasmic sensors; and (c) tretinoid acidinducible gene1 (RIG-1)-l

    receptors (RLRs), which are cytosolic helicathat primarily sense viruses. Historically,

    TLRs were first recognized for their rolehost defense, but the importance of NLRs

    complementing the functions of the TLRs become increasingly clear. The high evoluti

    ary conservation of the NLRs attests to theirficacy and vitality in host defense. Homologs

    the NLRs (e.g., R genes) have been discove

    throughout the plant and animal kingdomincluding phylogenetically primitive organis

    such as the zebrafish (2) and the sea urchwhich has at least 203 identified putative NL

    (3).

    DEFINING FEATURES OF THENOD-LIKE RECEPTOR FAMILY

    In humans, the NLR family is composed of

    proteins ( Table 1), and there are at least NLR genes in mice. Although primarily

    pressed in immune cells, including both lyphocytes and antigen-presenting cells (APC

    such as macrophages and dendritic cells, NL

    can also be expressed in nonimmune cells, cluding epithelial and mesothelial cells. T

    family of proteins is defined by a triptite structure consisting of (a) a variable

    terminal protein-protein interaction domadefined by the caspase recruitment dom

    (CARD), pyrin domain (PYD), acidic transtivating domain, or baculovirus inhibitor

    peat (BIR); (b) a central nucleotide-bindoligomerization (NOD) domain, which me

    ates self-oligomerization that occurs during tivation (4); and (c) a C-terminal leucine-r

    repeat (LRR) that detects PAMPs.

    366 Chen et al.

  • 8/6/2019 Annurev.pathol NOD

    3/36

    Table 1 NOD-like receptor (NLR) family members

    HGNC-approved symbol

    Human Mouse Other names and aliases Domain organization NLR family

    CIITA Cllta NLRA; MHC2TA; C2TA

    Nlra; MHC2TA; C2TA NLRA

    NAIP NLRB1; BIRC1; CLR5.1

    Naip1 Birc1a

    Naip2 Birc1b Naip3 Birc1c

    NLRB Naip4 Birc1d

    Naip5 Birc1e

    Naip6 Birc1f

    Naip7 Birc1g

    NOD1 NLRC1; CARD4; CLR7.1NLRC

    Nod1 Nlrc1; Card4

    NOD2 NLRC2; CARD15; CD;

    BLAU; IBD1; PSORAS1;

    CLR16.3

    NLRC

    Nod2 Nlrc2; Card15

    NLRC3 NOD3; CLR16.2

    NLRC Nlrc3 CLR16.2

    NLRC4 CARD12; CLAN; CLR2.1;

    IPAF NLRC

    Nlrc4 Card12; CLAN; Ipaf

    NLRC5 NOD27; CLR16.1NLRC

    NIrc5

    NLRP1 NALP1; DEFCAP; NAC;

    CARD7; CLR17.1

    Nlrp1a NALP1

    NLRPNlrp1b

    Nlrp1c NLRP2 NALP2; PYPAF2; NBS1;

    PAN1; CLR19.9 NLRP

    Nlrp2 Pypaf2; Nbs1; Pan1

    NLRP3 CIAS1; PYPAF1; Cryopyrin;

    NALP3; CLR1.1 NLRP

    Nlrp3 Cias1; Pypaf1; Cryopyrin;

    Nalp3; Mmig1

    NLRP4 NALP4; PYPAF4; PAN2;

    RNH2; CLR19.5

    Nlrp4a Nalp4a; Nalp-eta; Nalp9D

    Nlrp4b Nalp4b; Nalp-gamma; Nalp9ENLRP Nlrp4c Nalp4c; Nalp-alpha; Rnh2

    Nlrp4d Nalp4d; Nalp-beta

    Nlrp4e Nalp4e; Nalp-epsilon

    Nlrp4f Nalp4f; Nalp-kappa; Nalp9F

    Nlrp4g Nalp4g

    (Continued

    www.annualreviews.org NLRs: Role in Immunity and Disease 367

  • 8/6/2019 Annurev.pathol NOD

    4/36

    Table 1 (Continued)

    HGNC-approved symbol

    Human Mouse Other names and aliases Domain organization NLR famil

    NLRP5 NALP5; PYPAF8; MATER;

    PAN11; CLR19.8 NLRP

    Nlrp5 Mater; Op1

    NLRP6 NALP6; PYPAF5; PANS;

    CLR11.4NLRP

    Nlrp6 NLRP7 NALP7; PYPAF3; NOD12;

    PAN7; CLR19.4 NLRP

    NLRP8 NALP8; PAN4; NOD16;

    CLR19.2 NLRP

    NLRP9 NALP9; NOD6; PAN12;

    CLR19.1

    Nlrp9a Nalp9a; Nalp-thetaNLRP Nlrp9b Nalp9b; Nalp-delta

    Nlrp9c Nalp9c; Nalp-zeta

    NLRP10 NALP10; PAN5; NOD8;

    PYNOD; CLR11.1 NLRP

    Nlrp10 Nalp10; Pynod NLRP11 NALP11; PYPAF6; NOD17;

    PAN10; CLR19.6 NLRP

    NLRP12 NALP12; PYPAF7; Monarch1;

    RNO2; PAN6; CLR19.3 NLRP

    Nlrp12 Nalp12

    NLRP13 NALP13; NOD14; PAN13;

    CLR19.7 NLRP

    NLRP14 NALP14; NOD5; PAN8;

    CLR11.2 NLRP

    Nlrp14 Nalp14; Nalp-iota; GC-LRR

    NLRX1 NOD9; CLR11.3

    NLRX

    The N-terminal domain of the NLRsis critical for downstream signaling. CARDdomains were originally associated with pro-

    teins involved in apoptosis and inflammationsuch as many of the caspases, including caspase-

    1; however, CARDs have also been shownto mediate caspase-independent interactions.

    The structure of PYD is homologous to tof CARD and promotes homophilic intactions with other PYD-containing prote

    that are important for downstream signalevents. Both CARD and PYD are members

    the death domainfold superfamily, membof which are involved in both apoptosis a

    368 Chen et al.

  • 8/6/2019 Annurev.pathol NOD

    5/36

    inflammation. Finally, the BIR-containing pro-

    teins can be classified into two major groups,

    inhibitor of apoptosis proteins (IAPs) and neu-ronal apoptosis inhibitor proteins (NAIPs),

    both of which are NLR family members. Be-cause of the variety in domain structures for

    each NLR, the nomenclature of the differentNLRs has been inconsistent and confusing. Re-

    cently, in order to promote uniformity in thenomenclature of the various NLRs, a new sys-

    tem approved by the Human Genome Orga-nization Gene Nomenclature Committee was

    established (5). This new annotation systemrecognizes the evolutionarily conserved NOD

    and LRR domains that are the central defin-

    ing features of these PRRs, but it further sub-divides the NLR family into four subfamilies

    (indicated by letters A through C and P) basedon the type of N-terminal effector domain

    (Table 1). An additional subfamily, NLRX, hasno strong homology to the N-terminal domain

    of any of the other four subsets and currentlyconsists of only one member, NLRX1 (Nod9),

    which appears to be unique in its localization tothe mitochondria (6, 7).

    NOD-LIKE RECEPTORSIGNALING: DIVERSE ANDCOMPLEX PATHWAYS WITH

    MULTIPLE LEVELSOF REGULATION

    In general, the primary function of PRRs is theactivation of inflammatory signaling pathways.

    In some respects, NLR signaling is very sim-ilar to that of the TLRs, with shared down-

    stream targets (Figure 1). For example, uponrecognition of their respective PAMPs, the pri-

    marily membrane-bound TLRs recruit adaptorproteins such as MyD88 and TRIF, which acti-

    vate the MAPK and NF-B signaling pathways,

    resulting in the induction of proinflammatoryand antimicrobial mediators such as interleukin

    (IL)-6, tumor necrosis factor alpha (TNF-),and IL-1 (for a review, see Reference 8). Sim-

    ilarly, stimulation of the intracellular NLRs ac-tivates downstream signaling pathways for the

    production of proinflammatory mediators to

    defend the host against infection. However, the

    end targets of NLR signaling are not the samefor all NLRs. Three major activation targets of

    NLR signaling after PAMP recognition have

    been identified: (a) NF-B, (b) MAPKs, and(c) caspase-1 (Figure 1).

    Nuclear FactorKappaB/Mitogen-ActivatedProtein Kinase Signaling

    The earliest-identified and best-characterized

    NLRs are NOD1 and NOD2, which are pro-totypical of NLR activation of both the NF-

    B and MAPK pathways (912). Upon recog-

    nition of their respective agonists, both NOD1and NOD2 self-oligomerize to recruit and ac-

    tivate the adaptor protein RICK (also knownas RIP2), which is essential for the activation

    of both NF-B and the MAPKs (4, 10, 1316). RICK is a serine-threonine kinase that be-

    comes polyubiquitinated upon interaction withNOD1 or NOD2 through homotypic CARD-

    CARD interactions (17). This RICK K63linked ubiquitination step is essential for re-

    cruitment of the kinase TAK1, which activatesthe NF-B-activating complex and is inhibited

    by the deubiquitinase A20 (17). RICK itself

    recruits and promotes the K63-linked polyu-biquitination of the I-kappa-B kinase gamma

    (IK) regulator subunit of the IK complex,or NF-B essential modulator (NEMO), which

    can also facilitate recruitment of TAK1 in aubiquitin-dependent manner (18). The colo-

    calization of NEMO and TAK1 promotes thesubsequent phosphorylation of the IK sub-

    unit of IK by TAK1 and results in the phos-phorylation and degradation of IB, a criti-

    cal step that allows the cytoplasmic release andnuclear translocation of NF-B (19). NF-B

    subsequently activates transcription of inflam-

    matory cytokines and chemokines such asTNF-, IL-6, IL-8, and membrane cofactor

    protein 1, which are important for stimulationand recruitment of additional effector cells dur-

    ing host defense.As has been demonstrated for both NOD1

    and NOD2 signaling, ubiquitination plays a

    www.annualreviews.org NLRs: Role in Immunity and Disease 369

  • 8/6/2019 Annurev.pathol NOD

    6/36

    a

    b

    c

    Lipoprotein

    Flagellin

    Viral

    dsRNAT

    RIF

    Endosome

    TLR1

    or

    TLR6

    TLR5

    TLRs

    TLR2

    TLR

    4

    LPS

    MyD88

    Imiquimod

    IRA

    K1

    IRAK4

    TRAF6

    TAK1T

    AB1

    TAB2/3

    NEMO

    TLR3

    TLR7

    TLR9

    CpGDNA

    MKKs

    Nucleus

    ERK

    JNK

    IKK

    IKK

    NF-B

    NF-B

    R1

    r R6

    TLR5

    7

    TLR9

    C

    TLRs NNN

    FNF-FBBB

    NF-B

    TAK1

    TAK1

    iE-DAP

    MDP

    NOD1

    N

    OD2

    RICK

    CA

    RD9

    RICK

    CARD9

    NLR

    inflammasome

    Ub

    Ub

    TAB1 T

    AB2/3

    TAB1

    TAB2/3

    NEMO

    MKKs

    MKKs

    p38

    JNK

    ERK

    p3

    8

    JNK

    ERK

    IKK

    IKK

    Pro-IL-1

    IL-1

    Figure1

    Toll-likereceptor(TLR)andNod-likereceptor(NLR)signalingpathways.(a)Extracellularpathogen-associatedmolecularpatterns(PAMPs)

    arerecognizedbyTLRsat

    theplasmamembranean

    dendosomes,whichsignalthroughtheadaptorsMyD88andToll/interleukin-1receptordomain-containingadapter-inducinginterferonbeta

    (TRIF),aswellasthroughinterleukin-1receptorassociatedkin

    ase(IRAK)proteinsandtumornecrosi

    sfactorreceptorassociatedfactor6(TRAF6).(b)TheNLR

    proteinsNOD1andNO

    D2senseintracellularD--glutyamyl-meso-DAP(iE-DAP)andmuramyldipeptide(MDP),respectively,

    leadingtor

    ecruitmentoftheadaptor

    proteinsRICKandcaspaserecruitmentdomain9(CARD9).Su

    bsequently,bothTLRsandNOD1/NO

    D2signalingpathwaysrecruitTAK1,whichmediatesthe

    activationofnuclearfactorkappaB(NF-B)andmitogen-activ

    atedproteinkinases(MAPKs),resultinginthetranscriptionalupregulationof

    proinflammatorygenes.

    (c)ActivationofNLRsb

    ymicrobialorendogenousmoleculesinthecytosolresultsintheformationof

    caspase-1-activatinginflammasomes.Activationofcaspase-1

    inducesprocessingoftheinterleukin-1-beta(IL-1)precursorandsecretionofthematurecytokine.Abbreviations:ERK,extracellularsignal

    regulatedproteinkinase;

    IKK,

    I-kappa-Bkinase;JNK,c-JunN-terminalkinase;MKK,M

    APkinasekinase;NEMO,

    NF-Bessentialmodulator.

    370 Chen et al.

  • 8/6/2019 Annurev.pathol NOD

    7/36

    key role in activation of downstream signaling.

    For NOD2-mediated activation of NF-B in

    particular, the E3 ligase tumor necrosis factorreceptorassociated factor 6 (TRAF6) was sug-

    gested to be importantbut dispensableforNOD1 signaling, whereas TRAF2 and TRAF5

    were essential (17, 20). The significance of thedifferential utilization of E3 ligases for ubiqui-

    tination is not clear, but this ligase may provideadditional points of regulation during the in-

    duction of inflammatory cytokine production.NOD2 is also additionally regulated by Erbin,

    which interacts with NOD2 and is capable ofdownregulating NOD2-dependent activation

    of NF-B. However, whether Erbin also reg-

    ulates signaling of other NLRs remains to bedetermined (21).

    Stimulation of NOD1 and NOD2 also re-sults in the activation of MAPKs, including

    the p38, extracellular signalregulated proteinkinase (ERK), and c-Jun N-terminal kinase

    ( JNK) pathways (Figure 1) (15). In contrastto NF-B, the molecular events that occur for

    activation of these pathways are not as well de-fined, but they involve similar upstream signal-

    ing molecules such as RICK and TAK1 (15,22, 23). Recent evidence also suggests that the

    adaptor protein CARD9 may be particularly

    important in NOD2- and RICK-mediated ac-tivation of MAPK pathways and less so for

    NF-B (24).Importantly, there are NLR members that,

    instead of promoting NF-B activation, mayhave a primarily negative regulatory role. These

    members include NLRP12 (25, 26), NLRC3 inT cells (27), and NLRP2 in macrophages (28).

    As the studies identifying these members wereall performed in vitro and typically involved

    overexpression of proteins or the use of tumorcell lines, the physiologic roles of these NLRs

    in vivo remain to be determined.

    Biological Responses to NOD-1and NOD-2 Signaling

    A consequence of NOD1- and NOD2-mediated activation of NF-B and MAPK is

    the upregulatedtranscription and productionof

    MDP: muramyldipeptide

    inflammatory mediators, including cytokines,

    chemoattractants, adhesive molecules, and in-

    ducible molecules (e.g., iNOS, Cox-2). ForNOD1 specifically, which is expressed in ep-

    ithelial and mesothelial cells, its stimulation caninduce chemokine production and recruitment

    of effector immune cells, including neutrophilsin vivo (29, 30). Equally important is the upreg-

    ulation of antimicrobial peptide productionespecially in epithelial cells, which may be NF-B dependent, by both NOD1 and NOD2 sig-naling. Disruption of this function may con-

    tribute to the pathogenesis of inflammatorydiseases in the bowel (3136). NOD1 signal-

    ing is also important in the coordination of an

    adaptive immune response by both T and Bcells through synergistic antigen presentation

    with TLRs (1). Similarly, a role for NOD2 inmounting adaptive responses is implicated by

    the observation that the NOD2 agonist, mu-ramyl dipeptide (MDP), can act as an effective

    adjuvant for antigen-specific T cell responsesand antibody production (37, 38).

    Aside from the induction of proinflamma-tory mediators, NOD1 and NOD2 have also

    been shown to induce apoptosisin in vitro over-expression systems; in fact, these proteins were

    originally identified by their structural homol-

    ogy to Apaf-1 and CED-4, known regulatorsof apoptosis (13, 14). The specific pathways in-

    volved in the induction of apoptosis are not en-tirely clear, but for NOD1 this process involves

    both caspase-8 and caspase-9 and requiresRICK. Note, however, that these studies were

    all performed in vitro and that functional rele-vanceremainstobeinvestigatedinvivo(13,23).

    Interleukin-1 Productionand the Inflammasome

    In addition to NF-B and MAPKs, the third

    important pathway activated by NLR signalinginvolves ASC (adaptor protein apoptosis speck

    protein with caspase recruitment) to activatecaspase-1 (Figure 1). NLRs that participate in

    caspase-1 activation include NLRP1, NLRP3,and NLRC4. Caspase-1 activation is required

    for the cleavage of pro-IL-1 and pro-IL-18

    www.annualreviews.org NLRs: Role in Immunity and Disease 371

  • 8/6/2019 Annurev.pathol NOD

    8/36

    into their mature, biologically active forms. Re-

    cruitment of ASC by these NLRs is believed to

    occur through homophilic PYD-PYD interac-tions. Via its CARD domain, ASC, in turn, in-

    teracts with caspase-1. These protein-proteininteraction domains promote oligomerization,

    recruitment,andapproximationofeffectorpro-teins essential for activationof caspase-1 (4, 39).

    Indeed, a hallmark of this caspase-1-dependentpathway is the assembly of large macromolec-

    ular complexes through CARD-CARD andPYD-PYD protein-protein interactions that

    function to form a scaffold for procaspase-1 recruitment and activation. This molecu-

    lar platform, of which NLR family members

    are the cornerstone, has been termed the in-flammasome (40) as an analogy to the apopto-

    some, an activator of caspase-9 duringapoptosis(41).

    Currently there are three well-described in-flammasomes named after the NLR involved:

    NLRP1, NLRP3, and NLRC4. Common tothese inflammasomes is the role of ASC as the

    adaptor protein that bridges these NLRs tocaspase-1 (40, 42). The NLRP1 inflammasome

    was the first to be characterized and is the onlyinflammasome that has been reconstituted in

    vitro with purified proteins (40, 43). The ex-

    istence of the NLRP3 and NLRC4 inflamma-somes, the activities of which are dictated by

    the specific PRRs recognized by the NLRs, hasbeen implicated primarily by the ability of both

    NLRP3 and NLRC4 to activate caspase-1 in anASC-dependent manner (4450).

    OtherNLRmembersthatcanassociatewithprocaspase-1 and promote IL-1 production

    such as NLRP2have only been demonstratedin vitro, but whether the latter is a bona fide in-

    flammasome that functions to activate caspase-1 during host defense in vivo remains to be

    determined (28). NAIP with its BIR domain,rather than its CARD or PYD domain, has also

    been shown to be capable of activating caspase-

    1 at least for the murine isoform Naip5; how-ever, the relevance of its ability to interact with

    caspase-1 and whether it acts alone or togetherwith NLRC4 remain controversial. We discuss

    this issue in more detail below.

    Another important consequence of caspa

    1 activation is the induction of a newly rognized process of programmed cell de

    termed pyroptosis, which is distinct from aptosis and necrosis. Pyroptosis is typically

    duced in macrophages infected with cert

    intracellular bacteria such as Salmonella andassociated with a proinflammatory response

    volving caspase-1-mediated IL-1 and IL-production (51). This response causes rapid f

    mation of plasma membrane pores, cellular sis, and release of intracellular inflammato

    contents to fuel additional inflammatory snaling pathways (52). Caspase-1 activation a

    the release of IL-1 and IL-18 have bshown to be particularly important in h

    defense against such pathogens as Shig(53), Legionella (48), Francisella (54), Liria (55), Yersinia (56), and Bacillus anthr

    (52). How the different inflammasomes enlisted to defend against specific pathoge

    or stimuli is described in the followsections.

    NOD-LIKE RECEPTORSRECOGNIZE SPECIFICBACTERIAL ANDENDOGENOUS MOLECULES

    Although numerous studies have delineated

    importance of individual NLRs against specpathogens, a direct interaction between a p

    tative ligand and its corresponding NLR hnot been demonstrated for most of the NL

    Therefore, the possibility that the interactibetween NLR and PRR is indirect and invol

    an intermediary host factor or activity cannbe precluded. In this section, we describe w

    is known about the sensing of pathogens by various NLRs (Table 2).

    NOD1

    Originally, LPS preparations were demostrated to activate both NOD1 and NO

    through RICK, but this was recently shownbe due to contamination of LPS preparatio

    with PGN moieties (10, 15, 57). Both NO

    372 Chen et al.

  • 8/6/2019 Annurev.pathol NOD

    9/36

    Table 2 NOD-like receptor (NLR) agonists and downstream signaling pathwaysa

    Receptor Agonist Bacteria Signaling pathway

    Nod1 GM-tripeptide Helicobacter pylori NF-B

    meso-lanthionine, meso-DAP Shigella flexneri MAPK

    -d-Glu-DAP(iEDAP) Listeria monocytogenes

    Campylobacter jejuni

    d-lactyl-l-Ala--Glu-meso-DAP-Gly (FK156) Enteropathogenic Escherichia coli

    heptanolyl--Glu-meso-DAP-Ala (FK565) Chlamydia pneumoniae

    Pseudomonas aeruginosa

    Bacillusspp.

    Nod2 MDP Streptococcus pneumoniae NF-B

    MurNAc-l-Ala-g-d-Glu-l-Lys (M-TRILys) Listeria monocytogenes MAPK

    Mycobacterium tuberculosis

    Salmonella Typhimurium

    Staphylococcus aureus

    Shigella flexneri

    Nlrc4 flagellin (Salmonella, Legionella, Pseudomonas) Salmonella Typhimurium caspase-1

    unknown (Shigella) Legionella pneumoniae

    Pseudomonas aeruginosa

    Shigella flexneri

    Listeria monocytogenesb

    Naip unknown Legionella pneumophila caspase-1

    Pseudomonas aeruginosa

    Salmonella

    Listeria

    Nlrp1b anthrax lethal toxin Bacillus anthracis caspase-1

    Nlrp3 bacterial RNA Staphylococcus aureus caspase-1

    viral RNA and DNA Listeria monocytogenesb

    uric acid crystals

    LPS

    LTA

    MDPsilica

    asbestos

    aAbbreviations: LPS, lipopolysaccharide; MAPK, mitogen-activated protein kinase; MDP, muramyl dipeptide; NF-B, nuclear factorkappa B.bBoth Nlrc4 and Nlrp3 may play redundant roles in caspase-1 activation in response to this pathogen, as both have been shown to contribute to IL-1

    production in vitro.

    and NOD2 recognize PGN moieties found

    in the bacterial cell that are secreted bybacteria (5861). PGN provides structure and

    rigidity to bacteria and is found in virtually

    all bacteria, although the amount, location,and specific composition may vary (62).PGN consists of sugar chains of alternating

    N-acetylglucosamine (GlcNAc) and N-acetyl

    muramic acid (MurNAc), which are cross-linked by short peptide chains (Figure 2).

    These peptide chains contain unique aminoacids that are differentially found in gram-

    negative and gram-positive bacteria and that

    are also differentially recognized by NOD1 andNOD2. Specifically, NOD1 can sense PGN

    moieties containing meso-diaminopimelic

    acid, an amino acid that is found predomi-nantly in gram-negative bacteria but also insome gram-positive bacteria such as Listeriamonocytogenes and Bacillus spp. The minimalstructure recognized by NOD1 is the dipeptide

    D--glutyamyl-meso-diaminolimelic acid (iE-

    DAP) (59, 60). Studies defining synthetic iE-DAP derivatives that stimulate NOD1 revealed

    www.annualreviews.org NLRs: Role in Immunity and Disease 373

  • 8/6/2019 Annurev.pathol NOD

    10/36

    Plasma membrane

    PGN

    Lipoteichoic acid

    Polysaccharide

    Cytosol

    Cytosol

    LPS Porin

    Lipopeptide

    Plasma membrane

    PGN

    Outer membrane

    L-alanine

    D-glutamic acid

    Mesodiaminopimelic acid

    D-alanine

    Tetra-peptidechain (amino acids)

    MDP(Nod2 ligand)

    iE-DAP

    (Nod1 ligand)Pep

    tidoglycan(PGN)

    Glycan chain

    Gram-negativ

    ebacteria

    Gram-positivebacteria

    NAGNAM NAG NAM

    NAG NAMNAG NAM

    374 Chen et al.

  • 8/6/2019 Annurev.pathol NOD

    11/36

    that the attachment of hydrophobic acyl

    residues enhanced stimulation of NOD1 up to

    severalhundredfold(63).Astheseresiduescon-tained fatty acid chains similar to those found

    in the phospholipids that make up the hostcell membrane, it is possible that the increased

    NOD1-stimulatory activity is due to an in-creased interaction with the cellmembrane that

    facilitates translocation into the intracellularcompartment of the host cell (63). The

    lipophilicity of the NOD1 ligand appears im-portant for recognition by NOD1, butprecisely

    how NOD1 interacts with these lipophilicmolecules and whether this actively contributes

    to its transfer and recognition into the host cell

    remain to be determined.In vitro studies have demonstrated that

    many bacteria have NOD1-stimulatory activ-ity, with the strongest activity associated with

    the genus Bacillus (58). Moreover, the pres-ence of bacteria has not always been necessary

    for NOD1 stimulation, as water-soluble ex-tracts from food and soil as well as supernatant

    from overnight bacterial cultures are capable ofstimulating NOD1 (58). This suggests that

    physical contact with live bacteria is not nec-essary and that NOD1 agonists can be pro-

    duced and released by bacteria. Consistent

    with this observation, Shigella mutants that in-crease release of PGN fragments upregulated

    NOD1-dependent NF-B activation in vitro(64). In addition, where and how NOD1 in-

    teracts with bacteria are not clear. Localiza-tion of bacteria to the intracellular compart-

    ment has been shown to not necessarily berequired for NOD1 stimulation, as the Liste-ria streptolysin O mutant, which is requiredfor Listeria to escape from the phagosome into

    the cytosol, is still capable of activating NF-B

    in a NOD1-dependent manner (58). Localiza-tion studies of NOD1 through overexpression

    have shown that NOD1 becomes membrane

    bound, at least during infection with Shigellaflexneri at sites of bacterial entry. However,

    whether this occurs in vivo remains to be de-termined, as endogenous NOD1 localization

    during infection has been difficult to visualize(65).

    Despite the identification of the PGN moi-ety sensed by NOD1, the role of NOD1 dur-

    ing in vivo infection remains unclear, with thepartial exception ofHelicobacter pylori, a bacte-

    rial organism associated with the developmentof gastritis and duodenal ulcers. Specifically,

    Nod1-deficient mice have shown increased sus-ceptibility to H. pylori infection, with an as-

    sociated impairment of -defensin-4 produc-

    tion (34, 66). In vitro studies, on the otherhand, have shownNOD1-dependent activation

    by other pathogenic bacteria such as L. mono-cytogenes (15), S. flexneri (9), Campylobacter je-

    juni(36, 67), enteroinvasive Escherichia coli(68),

    Chlamydophila pneumonia (69), and Pseudomonasaeruginosa (70). However, these findings werebased on in vitro studies only and may not nec-

    essarily reflect requirements in vivo. For exam-ple, NOD1 was originally implicated in pro-

    tection against Chlamydia trachomatis infectionin vitro, where Nod1-deficient mouse embry-

    onic fibroblasts (MEFs) exhibited decreased cy-

    tokine production after C. trachomatisinfection;however, Nod1-deficient mice did not have

    increased bacterial load or clinical symptoms,suggesting either that NOD1 is not directly in-

    volved or that other redundant signaling path-ways can compensate (71).

    Figure 2

    Generation of NOD1 and NOD2 ligands from bacterial peptidoglycan (PGN). In this simplified diagram,the bacterial cell wall and PGN structure from Escherichia coliare depicted. Parallel PGN strands composedof the alternating amino sugars N-acetylglucosamine (NAG) and N-acetyl muramic acid (NAM) are cross-linked to each other by stem peptides. Notice that E. coliPGN lacks bridging amino acids linking stempeptides and that cross-linking occurs via a direct link between a meso-diaminolimelic acid (meso-DAP)residue and the d-alanine residue in position four from a peptide anchored on the parallel glycan strand.Minimal motifs required for NOD1 and NOD2 (dashed boxes) recognition are also shown. Abbreviations:iE-DAP, d--glutyamyl-meso-DAP; LPS, lipopolysaccharide; MDP, muramyl dipeptide.

    www.annualreviews.org NLRs: Role in Immunity and Disease 375

  • 8/6/2019 Annurev.pathol NOD

    12/36

    NOD2

    Separate studies have also demonstrated thatNOD2 can detect pathogenic bacteria. In

    contrast to NOD1, which appears to beprimarily involved in sensing gram-negative

    bacterial pathogens, NOD2 senses the spe-cific MDP motif that is found in a broader

    range of bacteria, with some overlap withthose recognized by NOD1 (Figure 1). These

    bacteria include Streptococcus pneumoniae (72),

    L. monocytogenes(33, 73), Mycobacterium tuber-culosis (74), Salmonella (75), and Staphylococcus

    aureus (76). Again, most of these studies wereperformed in vitro; therefore, the in vivo rel-

    evance of these findings remains to be deter-mined. One exception, however, is L. monocyto-

    genes, in which increased bacterial burdens wereobserved in Nod2-deficient mice when infected

    orally rather than intravenously or intraperi-toneally (33). This immunity is associated with

    decreased production of-defensins in Panethcells (33). In addition, both Nod1 and Nod2

    have been shown to be important for bacterialrecognition and host defense against L. mono-cytogenesafter exposure of macrophages or an-

    imals to LPS or E. coliin vivo (77). These ob-servations suggest that the intracellular sensors

    NOD1 and NOD2 play a critical role in hostdefense when TLRsignaling is reduced, such as

    within the intestine due to low expression levelsof TLRs (78, 79) or after induction of toleriza-

    tion by exposure to TLR ligands.

    NLRC4

    As mentioned above, a subset of NLRs partic-ipate in the formation of inflammasomes that

    ultimately leads to activation of caspase-1 and

    maturation of IL-1. To date, these NLRs in-clude NLRC4, NLRP1, and NLRP3. Studies

    have shown that the NLRC4 inflammasomecan activate caspase-1 in response to infection

    by S. Typhimurium, an activity not shared bythe NLRP3 inflammasome (49, 50). Salmonella

    infection can lead to gastroenteritis as well as totyphoid fever, which canbe associated with high

    morbidity and mortality. What is recognized

    by NLRC4 is flagellin, as flagellin-defici

    Salmonella mutants elicit substantially redulevels of caspase-1 activation (49). Similarly,

    tosolic detection ofL. monocytogenesby Ipaf

    quires flagellin (80). A direct interaction btween flagellin and NLRC4 has not yet be

    demonstrated, however, and therefore recogtion is likely indirect. TLR5, a member of t

    TLR family, also senses flagellin; howeverhas been shown that the cytoplasmic delivery

    flagellin requires NLRC4 but not TLR5, cosistent withits cellular location.As an additio

    consequence of caspase-1 activation, caspa1 can lead to early macrophage cell death,

    NLRC4-dependent process in the host defeagainst Salmonella (47).

    In vivo, NLRC4 has also demonstrated

    tivity against Legionella pneumophila, an intcellular pathogen that causes Legionnaires d

    ease, which can be deadly. The pathogenesisLegionella requires successful entry and repli

    tion inside host macrophages and is dependon the formation of a specialized vacuole th

    blocks the fusion of the phagosome to the lysome, a process dependent on an intact fun

    tional type IV secretion system (TFSS). Athecasewith Salmonella, Legionella also conta

    flagellin, and NLRC4-dependent caspase-1 tivation requires the presence of intact flage

    and its intracellular delivery (48, 81). As a

    sult of NLRC4-induced caspase-1 activatiLegionella growth is restricted by promoti

    the fusion of the Legionella-containing phagsome to thelysosome fordegradation(48).H

    caspase-1 activation controls phagosome muration in response to Legionella infection

    not known, but it may involve interactioncaspase-1 with host proteins involved in pha

    some formation and transport or direct targing ofLegionella virulence factors required

    lysosomal evasion.In vitro, NLRC4 is important in respond

    to Shigella, an intestinal pathogen that cau

    dysentery and, as mentioned above, can aelicit NOD1-dependent MAPK activation a

    secretion of IL-8 (9). Like Legionella, Shigcan escape from within membrane vacuo

    and enter the cytosol. Interestingly, Shig

    376 Chen et al.

  • 8/6/2019 Annurev.pathol NOD

    13/36

    does not express flagellin genes, yet it can ac-

    tivate caspase-1 macrophages and induce py-

    ropoptosis in an NLRC4-dependent process(82). This suggests that NLRC4 also rec-

    ognizes an as-yet-unidentified PAMP or sig-nal associated with Shigella. Shigella also in-

    duces autophagy, which is facilitatedin NLRC4but not in ASC-deficient macrophages, sug-

    gesting that NLRC4 provides an additionalhost-defense measure independent of ASC and

    IL-1 production by inhibiting autophagy (amembrane-trafficking system in which cyto-

    plasmic components are sequestered for de-livery to lysosomes, where cytoplasmic ma-

    terial is degraded). This downregulation of

    autophagy is specific to Shigella-induced au-tophagy, as autophagy from serum starvation or

    rapamycin treatment does not require NLRC4or caspase-1 (82). The reason for the inhibi-

    tion of autophagy by NLRC4 is not clear, but ithas been proposed that this inhibition allows

    the induction of pyroptosis, which may bet-ter promote an effective inflammatory response

    (82).

    NAIP

    One of the mouse homologs of human NAIP,

    Naip5, is also important in the host defenseagainst Legionella. This finding arose from

    observations that different mouse strainsshow varying permissiveness to intracellular

    replication of Legionella and that this permis-siveness is genetically controlled (83, 84). The

    genetic locus responsible for the increasedsusceptibility to Legionella infection in the A/J

    mouse strain in particular has been determinedto be Naip5 (85). Naip5 promotes the fusion

    of the Legionella-containing phagosomes tothe lysosome, thereby preventing intracellu-

    lar replication (86). Studies in vitro have also

    suggested that Naip5 activation requires the in-tracellular delivery of flagellin, which results in

    caspase-1 activation and in promotion of earlycell death, thereby controlling intracellular

    replication ofLegionella (81, 87, 88). However,these studies used large numbers of Legionellabacteria, which can lead to nonspecific early

    cell death of macrophages; they also used

    an indirect, nonspecific method of detectingcaspase-1 activation. In a separate study using

    low bacterial load and caspase-1-specific anti-

    bodies, Legionella-induced caspase-1 activationand IL-1 production occurred independently

    of Naip5 but required Nlrc4 as well as flagellin(89). Naip5, however, was still required for in-

    hibition ofLegionella replication intracellularly,irrespective of Nlrc4 signaling; this suggests

    a caspase-1-independent pathway regulatedby Naip5 that restricts Legionella replication

    and acts in concert with Nlrc4 signaling(89). The PAMP involved in this process has

    yet to be determined, as it does not requireflagellin. In addition, Naip5like Nlrc4has

    been suggested to regulate autophagy: A/J

    mice that harbor mutant Naip5 exhibitedslower rates of autophagy induction after

    Legionella infection of macrophages comparedwith Legionella-resistant C57BL/6 that have

    intact Naip5 (90). This observation furtherdemonstrates the complexity of intracellular

    Legionella replication by different NLRs, and itremains an active area of investigation.

    Based on in vitro studies using A/J mice thatare considered Naip5 deficient compared with

    C57BL/6, which have intact Naip5 and are re-sistant toLegionella replication, Naip5 also con-

    tributes to IL-1 production by bone marrow

    derived macrophages after infection withP. aeruginosa, an opportunistic bacteria that can

    cause pneumonia and sepsis (91). The decreasein IL-1 production in A/J mice, although

    statistically significant, may reflect inherentlydifferent genetic susceptibility differences be-

    tween the two mouse strains that are indepen-dent of Naip5; therefore, this decrease may

    be physiologically irrelevant in vivo. There-fore, additional studies examining a true

    Naip5-knockout strain should provide addi-tional insight into and confirmation of these

    findings.

    NLRP1

    Recently discovered mutations in the Nlrp1

    (Nalp1b) gene in various mouse strains have

    www.annualreviews.org NLRs: Role in Immunity and Disease 377

  • 8/6/2019 Annurev.pathol NOD

    14/36

    been found to influence the susceptibility of

    mice to Bacillus anthracislethal toxin (LT) (92),

    the major virulence factor in the pathogen-esis of anthrax. This was the first demon-

    stration of an NLR that responds to avirulence factor, as opposed to a specific bacte-

    rial component. LT results in the formation ofpores allowing lethal factor, a protease, to enter

    cells, resulting in proteolytic cleavage of hostsubstrates and subsequent cell death. Suscepti-

    bility to LT-induced macrophage lysis/necrosisand death requires caspase-1 activation and is

    associated with Nalp1b polymorphisms. Five ofthese polymorphisms have been identified, ex-

    plaining the variability in susceptibility to LT-

    induced cell death in different mouse strains(92). Caspase-1 activation by LT results in

    pyroptosis similar to that seen in NLRC4-mediated cell death induced by Salmonella

    infection (52).The human NLRP1 inflammasome, recon-

    stituted with purified proteins including ASC,NLRP1, andprocaspase-1, hasalso beenshown

    to respond to MDP and to induce the matura-tion of IL-1. Because only purified proteins

    were used in this cell-free system (40), it is pos-sible that, in this case, a direct interaction be-

    tween the inflammasome and MDP occurred

    under physiological conditions.

    NLRP3

    NLRP3 is an NLR that also participates ininflammasome formation through the recruit-

    ment of ASC and subsequent activation ofcaspase-1 and secretion of IL-1 and IL-18.

    NLRP3 detects PAMPs such as LPS, MDP, andbacterial and viral RNA, including the double-

    stranded RNA analog poly I:C (4446) as wellas the imidazoquinoline antiviral compounds

    R837 and R848 (93). Consistently, NLRP3

    has been found to respond to infection withthe Sendai and influenza viruses, resulting in

    caspase-1 activation (44). Subsequently, it wasshown that NLRP3 can detect viral DNA (94).

    In vitro, NLRP3 has also been demonstrated tobe required for caspase-1 activation/IL-1 se-

    cretion in response to the bacterial pathogens

    S. aureusand L. monocytogenes(46). However

    the case ofL. monocytogenes, the role of NLRhas been controversial, as there are oppos

    data to suggest that caspase-1 activation byL

    teria can occur independently of NLRP3 (102). This discrepancy may be explained

    redundant contributions by both NLRP3 aNLRC4 in caspase-1 activation in response

    Listeria (80).Characteristic of NLRP3, however, is its

    pability to respond to a broad repertoire of onists that are not necessarily microbial in o

    gin, but rather are endogenous signals. NLRhas been suggested to respond to changes

    cellular ion concentrations, particularly potsium, that are produced, for example, by

    marine toxin maitotoxin and the K+/H+

    tiport ionophore nigericin (46). NLRP3 actition is also observed with uric acid crystals (

    and reactive oxygen species that are generain response to, for example, asbestos and sil

    (7).Significant insight into the mechanism

    which NLRP3 recognizes its various ligacame from the observation that the addition

    ATP to macrophages prestimulated with bterial molecules such as LPS can significan

    enhance caspase-1 activation and IL-1 p

    duction. Identifying the role of ATP in NLRactivation has been controversial, as high no

    physiological concentrations of ATP are tycally required for enhanced IL-1 producti

    An important function of ATP is the stimlation of the P2X7 receptor, which, in tu

    results in the opening of a pore mediatedthe hemichannel protein pannexin-1 (96, 9

    Therefore, a consequence of ATP additionthe ability of bacterial products to enter the

    tosol through the pore and to subsequently tivate NLRP3 and caspase-1. Indeed, with

    addition of ATP, several bacterial product

    including LPS, PGN, and lipoteichoic acas well as heat-killed bacteriacan activ

    caspase-1inanNLRP3-dependent(butaTLand RICK-independent) manner (44, 45,

    98). Moreover, the requirement for ATP canbypassed when other methods of cytosolic

    livery are implemented; such methods inclu

    378 Chen et al.

  • 8/6/2019 Annurev.pathol NOD

    15/36

    the use of pore-forming bacterial toxins (e.g.,

    streptolysin O) or the lipophilic DOTAP deliv-

    ery system (98, 99). However, not all bacterialpathogens that can invade the cytosol require

    NLRP3 for caspase-1 activation as Salmonellaand Francisellawhich are capable of deliver-

    ing bacterial products into the cytosol throughtype III secretion systems (TTSS) or through

    pore-forming molecules without ATPcanactivate caspase-1 independently of NLRP3

    (98). Thus, NLRP3 recognition of PAMPs ap-pears to be linked to pannexin-1-dependent

    pore formation. MDP, a NOD2 agonist, hasalso been shown to activate NLRP3 (100) so

    as to activate caspase-1. This activation is in-

    dependent of NOD2 and involves the internal-ization of MDP into acidified vesicles that are

    released into the cytosol upon addition of ATPin a pannexin-1-dependent manner (101).

    An increasingly recognized consequence ofactivation of the P2X7 receptor and pannexin-

    1 by ATP is the alteration in potassiumconcentrations in the cell (i.e., intracellular

    potassium depletion), which has been sug-gested to be important for NLRP3 but not

    for NLRC4 signaling (102, 103). Treatmentof macrophages with the pore-forming tox-

    ins nigericin and maitotoxin is also associ-

    ated with intracellular potassium depletionand results in NLRP3-induced activation

    of caspase-1 (46). Therefore, in additionto facilitating entry of NRLP3-responsive

    PAMPs, pore-forming toxins also promotechanges in intracellular potassium concen-

    trations that may be required for NLRP3activation. However, the concentration of

    potassium itself may not regulate caspase-1 ac-tivation, as changes in potassium concentration

    in the cell are associated with other ionic fluxesor cellular events that may be critically involved

    in NLRP3 signaling. Moreover, in the absence

    of PAMP prestimulation, ATP does not acti-vate caspase-1, even though it alone can trigger

    potent efflux of intracellular potassium (102).Thus, the true significance of potassium con-

    centration changes associated with NLR sig-naling remains to be fully elucidated. However,

    one can interpret changes in cellular potassium

    concentrations associated with pore formation,

    uric acid release, and ATP generation as surro-gate indicators of cellular stress or injury; there-

    fore, NLRP3 andperhaps other NLRs mayalso

    function to recognize and respond to endoge-nous danger signals besides microbial infection

    (95, 104).

    MECHANISMS OFINTRACELLULAR DELIVERYOF MICROBIAL PRODUCTSFOR DETECTION BY

    NOD-LIKE RECEPTORS

    A defining feature of the NLRs is their in-

    tracellular localization. With the exception ofNLXR1, which has been shown to be im-

    portant within the mitochondria (6, 7), andCIITA, which resides in part in the nucleus,

    all NLRs characterized to date are locatedwithin the cytoplasm. This raises the question

    of how these receptors recognize pathogens,or how pathogens are delivered into the intra-

    cellular compartment. Based on their cytoso-lic localization, NLRs may respond primarily

    to (a) bacteria that escape extracellular detec-tion by the TLRs and invade directly into the

    cell, (b) bacterial components that are deliv-

    ered into the cytosol through secretion sys-tems or through pore-forming molecules, or

    (c)bacterialproductsthatareuptakenbytheim-mune cell through phagocytosis or pinocytosis

    (Figure 3). Consistent with the first possibil-ity, NOD1 activation depends upon the intra-

    cellular localization of directly invasive bacteriasuch as S. flexneri(9, 64, 105) and S. pneumoniae(72). Also, only live and not paraformaldehyde-fixed C. jejunielicit NOD1-dependent NF-B

    activation (36).Bacteria with active secretion systems pro-

    vide an additional mechanism for intracellu-

    lar entry that is important for NLR recog-nition. Two secretion systems, type III and

    type IV, are involved in the injection of viru-lence proteins into the host cell to stimulate

    NLR signaling. These secretion systems arefound in gram-negative bacteria and are en-

    coded within regions of the bacterial genome

    www.annualreviews.org NLRs: Role in Immunity and Disease 379

  • 8/6/2019 Annurev.pathol NOD

    16/36

    P2X7R

    ATPPannexin-1a

    -BNF

    NLRinflammasome

    Pore-formingtoxin

    Pro-IL-1

    Nucleus

    IL-1

    Caspase-1

    Phagosome

    Bacteria type III or IVsecretion system

    P2X7R

    PAMPs

    Figure 3

    Modes of intracellular entry of microbes for Nod-like receptor (NLR)activation. Microbes and microbial molecules enter the cytosol viapore-forming toxins, type III or IV secretion systems, or ATP-mediatedactivation of the pannexin-1 pore. Sensing of microbial molecules in the cytosolby several NLRs results in the formation of inflammasomes. The mechanismwhereby NLRs recognize microbial molecules appears indirect and remainspoorly understood. Activation of caspase-1 induces processing of the

    interleukin-1-beta (IL-1) precursor and secretion of the mature cytokine.Abbreviation: PAMPs, pathogen-associated molecular patterns.

    known as pathogenicity islands. These secre-tion systems allow NLR detection by non-

    phagocytosing cells such as epithelial cells. ForTTSS, the mechanism by which gram-negative

    bacteria invade the cell is well illustrated by

    Salmonella (106). Two important features of

    Salmonella pathogenesis are (a) the bacterias

    ability to invade nonphagocytic cells such asintestinal epithelial cells and (b) their ability

    to survive as well as replicate within phago-cytes. Both of these features depend upon

    intact TTSS. TTSS consists of a complexassembly of proteins that form transporter ap-

    paratuses; these contain approximately 20 pro-tein subunits for the trafficking of bacterial

    proteins involved in bacterial virulence. As

    scribed above, the delivery of flagellin to tcytosol for NLRC4 inflammasome activat

    has been proposed to occur via TTSS, a

    Salmonella mutants with nonfunctional TTare unable to activate NLRC4 in macropha

    (50).InthecaseofLegionella, TFSSis essential

    the survival of the pathogen within the phagsome. In a not-fully-understood mechanis

    Legionellaonce phagocytosed by the APsuch as the macrophagereplicates within

    phagosome and avoids fusion with the lysome through secretion of virulence prote

    by the TFSS, which causes pore formati

    Legionella is recognized by NLRC4 most lik

    as a result of translocation of bacterial liga

    (flagellin) through the TFSS into the cyto(107). As with Legionella, delivery of PG

    derived molecules into the cytosol by H. pyforNOD1signalingrequiresanintactTFSS

    sociated with the cag pathogenicity island, a

    H. pyloristrains that lackcaghave reduced lev

    of NF-B activation (66).Bacterial secretion systems also ena

    pathogens to form pores to escape phasomes after phagocytosis, as well as

    transport bacterial products. For example, pore-forming toxin listeriolysin produced

    Listeria permits the delivery of PGN tha

    recognizable by NOD1 and NOD2 (12, 108). Other bacteria that secrete pore-form

    toxins, such as S. pneumoniae, which produpneumolysin, and B. anthraciswith anthroly

    O, can also allow internalization of PGN frments for recognition by NLRs, especially

    nonphagocytosing cells such as epithelial c(109).

    With certain bacteria, activation of NLcan occur with heat-killed bacteria alone, su

    gesting that neither an active secretion stem nor direct invasion is required for reco

    nition. In these cases, dead bacteria are lik

    directly uptaken into the cell by phagocyto(73). For example, inhibition of macropha

    phagocytosis of heat-killed S. aureusresulteddecreased inflammatory cytokine producti

    which was partly dependent on both NO

    380 Chen et al.

  • 8/6/2019 Annurev.pathol NOD

    17/36

    and NOD2 (76). The entry of MDP has also

    been proposed to occur in intestinal epithe-

    lial cells through a host-dependent rather thanbacterial-dependent mechanism, in which the

    active peptide transporter hPepT1 transportsMDP in vitro (110).

    NOD-LIKE RECEPTORSIGNALING SYNERGIZES ANDCOMPLEMENTS TOLL-LIKERECEPTOR SIGNALING

    There is great overlap and an apparent redun-dancy in the natureof thepathogensrecognized

    by the various NLRs and TLRs, and many of

    the receptors converge to common pathways(e.g., NF-B and IL-1 activation). An obvi-

    ous reason for the existence of multiple recep-tors channeling into common end targets is

    the potentiation of the inflammatory responseto infection. This is consistent with the syn-

    ergy observed with TLR and NLR signalingin response to PGN (74, 111, 112) and be-

    tween TLR and NOD2 agonists (33). In addi-tion, a segregation of function between NLRs

    and TLRs is naturally imposed by their cellu-lar localization, such that NLRs may acquire

    greater importance in the detection of intracel-

    lularly located bacteria. Regardless, NLRs andTLRs also play nonredundant roles, as is re-

    flected in the observations of TLR- and NLR-specific transcriptomes associated with Listeria

    infection andof synergistic activation of NF-B(113).

    Additional evidence for complementaryroles of NLRs and TLRs in the inflamma-

    tory response has been demonstrated with IL-1 secretion (see Figure 4a). The release

    of IL-1 involves two distinct steps: (a) in-duction of pro-IL-1, which is NF-B de-

    pendent, and (b) cleavage of pro-IL-1 by

    caspase-1 to the mature, active IL-1. The firststep requires TLR activation of NF-B (98);

    however, activation of caspase-1 occurs inde-pendently of TLRs and requires the inflam-

    masome (98). This two-part pathway, differen-tially regulated by TLRs and NLRs, may serve

    as a safeguard against excessive production of

    IL-1, which can cause pathology (see In-flammatory Diseases Associated with Excessive

    Interleukin-1 Production, below). Similarly,

    multiple NLRs or TLRs may recognize simi-lar stimuli but regulate different aspects of the

    same process. Using the same example of reg-ulation of IL-1 production, caspase-1 activa-

    tion by MDP stimulation requires NLRP3 andATP but not NOD2; however, the induction

    of IL-1 messenger RNA, on the other hand,requires NOD2 but not NLRP3 (101). There-

    fore, similar to therequirement for TLRsignal-ing to promote pro-IL-1 production through

    NF-B,inthecaseofIL-1 induction by MDP,NOD2 mediates the transcription of pro-IL-

    1byNF-B, whereas NLRP3 mediates IL-1

    processing through caspase-1 activation. Also,as described above, studies have suggested that

    Legionella recognition by both NLRC4 andNaip5 can result in alternate activities to re-

    strict intracellular replication ofLegionella andthat recognition of flagellin in either Legionella

    or Salmonella by TLR5 and NLRC4 can resultin the activation of separate pathways, NF-B

    and caspase-1 activation, respectively (4850,89, 114). Thus, the multiplicity of receptors in-

    volved in the recognition of the same ligandallows not only multiple levels of control to

    prevent excessive production of cytokines (e.g.,IL-1) that can harm the host, but it also pro-

    motes a concerted response against infection

    through the activation of separate, but coop-erative pathways.

    In addition to having a synergistic effect with TLR signaling, NLRs may provide the

    host with a backup defense mechanism thatmay be required under conditions when TLR

    signaling has been tolerized (Figure 4b). Awell-recognized phenomenon associated with

    TLR signaling is the induction of tolerance: After an initial LPS exposure to, for exam-

    ple, macrophages, cytokine responses are sup-pressed upon secondary exposure to LPS.

    Possible physiologic explanations for this phe-

    nomenon are (a) to prevent excessive cytokineproduction by bacterial stimuli, which can lead

    www.annualreviews.org NLRs: Role in Immunity and Disease 381

  • 8/6/2019 Annurev.pathol NOD

    18/36

    TLRs

    NF-B

    Pro-IL-1 IL-1

    Caspase-1

    NLRinflammasome

    TLRs

    Invasion ofintracellular bacteria

    TLR activation

    Reduced TLR signaling (tolerance)(prevent overproduction of proinflammatory cytokine)

    +Enhanced NLR signaling

    Prepare to fight invasive pathogen

    a b

    Figure 4

    The cooperative interplay between Toll-like receptors (TLRs) and Nod-like receptors (NLRs) in response to intracellular bacteria.(a) Stimulation of TLRs or NOD1/NOD2 induces the production of pro-interleukin-1-beta (pro-IL-1) through the activation ofnuclear factorkappa B (NF-B) signaling, leading to transcription of the IL-1 gene. In contrast, the presence of microbial molecuin the cytosol is sensed by the NLRs, resulting in inflammasome formation and caspase-1 activation. Activated caspase-1 cleavespro-IL-1 and results in secretion of mature IL-1. (b) Intracellular bacteria such as Listeria are recognized by TLRs, leading to TLactivation and production of proinflammatory and antimicrobial molecules. Persistent TLR stimulation leads to TLR tolerization anto reduced TLR signaling. Cytosolic invasion of the bacteria activates NLRs (e.g., NOD1/NOD2 and inflammasome-inducing NLR

    resulting in enhanced NLR signaling and in production of antimicrobial molecules.

    to the clinical syndrome of sepsis and to mul-tiorgan failure in the host, and (b) to stymie

    an excessive immune response against com-mensal bacteria, which can lead to patholog-

    ical inflammation. Although theoretically this

    is a good safeguard mechanism, a potenrisk in the development of hyporesponsiven

    with serial LPS challenge in APCs is an creased susceptibility to bacterial superinf

    tion (115). Because TLR-induced tolerizat

    382 Chen et al.

  • 8/6/2019 Annurev.pathol NOD

    19/36

    is limited to TLR agonists that act in the

    same pathway, NLRs may play a protective role

    against bacterial infection in the TLR-tolerizedstate (Figure 4b) (77). Indeed, it has recently

    been shown that cross-tolerization does not oc-cur with the combination of the NLR ago-

    nist MDP and the TLR agonist LPS, as pres-timulation with either LPS or MDP does not

    abrogate cytokine responses to MDP or LPS,respectively (77). Rather, enhancement in NF-B and MAPK activation as well as in cytokineproduction occurred after stimulation of MDP

    in macrophages made refractory to TLR lig-ands, and vice versa (77).

    A functional consequence of this phe-

    nomenon is the heightened importance ofNOD1 and NOD2 response to intracellular

    bacterial infection in macrophages tolerizedto previous exposure with TLR agonists (77).

    Consistently, it was demonstrated that afterprevious exposure to E. colior LPS, response to

    subsequent infection with Listeria was greatlycompromised in the absence of NOD1 and

    NOD2 signaling both in vitro and in vivo (77).Similarly, in the case of Salmonella infection,

    TLR-tolerant macrophages were still capableofIL-1productionthrough NLRC4signaling

    (49). Thus, NLR signaling can potentiate TLR

    signaling and can provide additional protectionto the host against bacterial invasion. One im-

    plicationfor this model is that immune cells canremain sensitive to infection by pathogenic bac-

    teria through NLR signaling in cells that havebecome tolerized to TLR ligands, such as com-

    mensal bacteria in the gut.Note, however, that several groups (116

    118) have also shownboth in humans andin micethat cross-tolerization still does oc-

    cur with stimulation of immune cells by NLRagonists followed by TLR agonists. This dis-

    crepancy may be due in part to differences inexperimental methods, including the timing of

    pretreatment (early versus late second expo-

    sure) or cell type (macrophages versus den-dritic cells), and it suggests that the regulation

    of tolerization and synergy between NLR andTLRs may be specific to both context and cell

    type.

    IBD: inflammatorybowel disease

    UC: ulcerative colit

    CD: Crohns diseas

    DYSREGULATED NOD-LIKERECEPTOR SIGNALING ISINVOLVED IN THEPATHOGENESIS OF MULTIPLEHUMAN DISEASES

    The physiologic significance of the variousNLRs lies primarily in their roles in host de-

    fense against microbial infection. However, itis becoming increasingly clear that NLRs alsofunction in organ homeostasis, which is un-

    derscored by the fact that many inflammatoryand noninflammatory disease processes can

    be attributed to dysregulated NLR signaling(Figure 5). The following subsections de-

    scribe certain defined NLR roles in humandiseases.

    Inflammatory Bowel Disease

    The role of NLRs in intestinal homeostasis was

    highlighted by the observations of mutationsin NOD2 that are associated with the devel-

    opment of inflammatory bowel disease (IBD).IBD encompasses two different diseases, ulcer-

    ative colitis (UC) and Crohns disease (CD);these diseases cause inflammation of the intes-

    tine, leading to the common clinical presenta-

    tion of abdominal pain, bloody diarrhea, andweight loss. Despite their similar clinical symp-

    toms, UC and CD have distinguishing clinicaland histologic features. In UC, inflammation is

    limited to the mucosal layer of the colon, mostcommonly therectum. In CD,however, inflam-

    mation is transmural and can involve any part ofthe gastrointestinal tract. Because of the trans-

    mural inflammation in CD, complications nottypically seen in UC can occur; these include

    bowel-wall perforation and fistula formation.CD also differs from UC with regard to the af-

    fected areas of the bowel: CD most commonly

    involves the lower part of the small intestine,the terminal ileum.

    The underlying pathogenesis of IBD is un-clear, but studies suggest that both environ-

    mental and genetic factors contribute to theetiology of CD. Genetic linkage analyses of

    affected families have identified eight genetic

    www.annualreviews.org NLRs: Role in Immunity and Disease 383

  • 8/6/2019 Annurev.pathol NOD

    20/36

    Blau syndrome/early-onsetsarcoidosis

    NOD2(skin, eyes, joints)

    Cryopyrinopathies

    (MWS, FCAS, NOMID)NLRP3

    (skin, eyes, joints)

    Asthma

    NOD1

    (lungs)

    Barelymphocyte

    syndrome

    CTIIA(lymphocytes)

    Inflammatory bowel disease

    NOD2, NOD1?(intestine)

    Sarcoidosis

    NOD1(lungs)

    Vitiligo

    NLRP1(skin)

    Gain-of-functionmutations

    Loss-of-functionmutations

    Figure 5

    Genetic variation in Nod-like receptors (NLRs) is associated with development of human disease.Gain-of-function point mutations within the NOD domains ofNLRP3 and NOD2 cause autoinflammatodisorders: The former cause Muckle-Wells syndrome (MWS), familial cold autoinflammatory syndrome

    (FCAS), and neonatal-onset multisystem inflammatory disease (NOMID), and the latter cause Blausyndrome/early-onset sarcoidosis. Consistently, the latter diseases are inherited in a Mendelian-dominantfashion. In contrast, loss-of-function mutations ofNOD1, NOD2, and CTIIA are associated with thedevelopment of asthma, adult sarcoidosis, bare lymphocyte syndrome, and Crohns disease. Genetic variatin NLRP1 is associated with vitiligo. The type of functional alteration of the disease-associated NLRP1alleles remains to be determined.

    loci(IBD1IBD8) containingCD-susceptibility

    genes (119). The risk allele for IBD1 has beenidentified as NOD2 by genetic and functional

    studies (120, 121). Although multiple variantsof NOD2 have been found to be associated

    with CD, approximately 40% of CD patientsof North American or Western European de-

    scent carry at least one of the three major

    disease-associated variants: G908R, R702W,and a frame-shift deletion mutation at L1007

    (L1007fsinsC). Patients homozygous for thesemutations have a 20- to 40-fold increased risk

    for disease development, whereas heterozyg

    subjects have only a two- to fourfold increarisk (120, 121). These CD-associated NO

    variants exhibit reduced capacity to activNF-B following MDP stimulation (61), co

    sistent with the finding that these mutationsnear or within the LRR domain of NOD2. T

    impaired sensing of PGN and/or MDP sugests that these CD-associated mutations res

    in a loss-of-function phenotype. Furthermo

    monocytes isolated from CD patients harbing the L1007fsins mutation exhibit reduc

    384 Chen et al.

  • 8/6/2019 Annurev.pathol NOD

    21/36

    3

    34

    4

    Reduced NOD2-dependentexpression of bactericidal-defensins in Paneth cells

    Impaired recognition andclearance of bacteria byintestinal phagocytes

    Dysregulation of TLR2-mediatedinflammation in intestinalmacrophages and/or dendritic cells

    Enhanced production of IL-1

    Macrophage

    Nod2

    IL-1

    Nod2

    Proinflammatorycytokines

    TLR2

    4

    innflflaaammmmmmaammmmaaaccrrooopphphh

    EEnEnnnhhhaaanncncced

    11LL

    -defensins

    Paneth cellsNod2

    4 ILIh

    1-1L-L4 LIL

    222R2R2TLRRRTLR

    Dendritic cell

    1

    1

    2

    2

    Figure 6

    Proposed models for the role of NOD2 in Crohns disease. Shown are four nonexclusive models of the contribution of mutant NODalleles to Crohns disease. In the first two models, impaired NOD2 function is associated with either increased invasion of intestinalbacteria by deficient production of-defensins (1) or reduced clearance of bacteria by intestinal phagocytes, leading to inappropriateactivation of NOD2-independent pathogen-recognition receptor (PRR)-signaling pathways (2). In the third model, NOD2 acts as abrake of commensal bacteriadriven inflammation, and the presence of deficient NOD2 alleles results in enhanced Toll-like receptor(TLR)-induced activation (3). In the fourth model, which is based on a mouse Nod2-knockin model, Crohns diseaseassociated NODmutations function as gain-of-function alleles, resulting in inappropriate interleukin-1-beta (IL-1) production (4).

    production of proinflammatory cytokines such

    as TNF-, IL-6, and IL-8, as well as the anti-inflammatory cytokine IL-10 (122, 123). How-

    ever, the loss of NF-B inducibility by the CD-associated NOD2 variants is inconsistent with

    the occurrence of increased NF-B-dependentinflammation observed in clinical samples iso-

    lated from CD patients. In order to reconcilethese observations and provide a mechanistic

    explanation, two broad hypotheses regardingthe role of NOD2 in the pathogenesis of CD

    have been advanced (Figure 6). The first hy-pothesisisthatmutationsinNOD2resultinde-

    ficiencies in epithelial-barrier function and/orin immune cells required for limiting bacterial

    invasion or clearance, which subsequently leads

    to increased inflammation at intestinal sites.The second contends that primary dysregula-

    tion of the mucosal immune system leads to ex-

    cessive activation of proinflammatory signalingpathways.

    Evidence for a role for NOD2 in regulatingepithelial-barrier function comes from trans-

    fection experiments demonstrating that wild-type (but not mutant) NOD2 can restrict pro-

    liferation of Salmonella in intestinal epithelialcells (75). The ability of NOD2 to restrict in-

    vasive bacterial growth may be related to itsability to activate NF-B-dependent produc-

    tion of the -defensins by Paneth cells, whichare specialized epithelial cells located in the

    crypts of the ileal mucosa. Consistent with this

    www.annualreviews.org NLRs: Role in Immunity and Disease 385

  • 8/6/2019 Annurev.pathol NOD

    22/36

    hypothesis is the observation that NOD2 is ex-

    pressedconstitutively in Paneth cells (124, 125).

    In addition, CD patients with mutant NOD2have reduced expression of two -defensins

    in the ileal mucosa, human -defensin-5 and-defensin-6 (31). Furthermore, mice genet-

    ically deficient for Nod2 displayed impairedprotection against oral, but not intravenous or

    intraperitoneal, administration of L. monocyto-

    genes. The increased susceptibility of Nod2-

    deficient mice was correlated with diminishedexpression of Panethcellderived antimicrobial

    peptides, Defcr4 and Defcr-rs10 (33). Thus,impaired production of-defensins in Paneth

    cells due to NOD2 mutations, which results

    in a compromise in barrier function, might bea plausible link between NOD2 and suscepti-

    bility to CD. However, the observation of de-creased -defensins should be interpreted with

    caution, as this may merely reflect Paneth cellloss from inflamed, damaged epithelium (126).

    Another possibility is that NOD2 mutationsresult in impaired clearance of locally inva-

    sive bacteria due to defective recognition byintestinal phagocytes. This alternative loss-of-

    function hypothesis is consistent with observa-tions of impaired production of antimicrobial

    molecules and of IL-1, induced by MDP in

    monocytes, expressing CD-associated NOD2mutations (61, 123). The defective removal of

    intestinal bacteria may drive inflammation viaactivation of NOD2-independent PRRs, in-

    cluding NOD1 and TLRs.The second hypothesis regarding the patho-

    genesis of CD contends that the disease re-sults from inappropriate hyperresponsiveness

    to commensal bacteria in the normal intestinemicroflora (127). This hyperreactive hypoth-

    esis is supported by evidence demonstratingthat (a) NOD2 functions as a negative regu-

    lator of IL-12 production mediated by PGNthroughTLR2and(b)intheabsenceofNOD2-

    mediated regulation, PGN elicits a heightened

    NF-B-dependent IL-12 or IL-23 responseby intestinal APCs, thereby promoting an ex-

    uberant adaptive Th1 or Th17 type of im-mune response often observed in CD patients

    (128, 129). In this model, it is assumed that in

    normal intestine, APCs are constantly expo

    to commensal bacteriaderived PGN withany overt immune response. It is then pos

    lated that in normal individuals, NOD2 futions as a brake, dampening any potential

    nate immune response to PGN. In contr

    PGN-mediated immune responses in indivuals with NOD2 mutations are refractory

    NOD2 modulation, thus resulting in high lels of IL-12/IL-23 andcreating an environm

    capable of generating a pathological Th1/Thimmune response. Consistent with the ideat

    dysregulated IL-12 promotes colitis, treatmof CD patients with antibody against IL-12

    been shown to be effective (130). Alternativedata derived from the analysis of knockin m

    expressing Nod2, which mimics L1007fsinsuggest that macrophages from these knoc

    mice had increased levels of IL-1 product

    (131). In addition to increased cytokine prodtion, theseknockin mice were shown to be m

    susceptible to dextran sodium sulfateinduccolitis, suggesting that the frame-shiftmutat

    associated with CD is a gain-of-function mtation. However, unlike the published knoc

    Nod2 mouse model, monocytes from healtor CD patients homozygous for NOD2 mu

    tions exhibit loss-of-function phenotypes (123). Thus, the relevance of the knoc

    Nod2 mouse model remains uncertain.The hpotheses presented here may not be mutua

    exclusive; further, they suggest that NOD2 m

    contribute to CD development via multimechanisms.

    Due to the similarity in signaling and struture between NOD1 and NOD2, research

    are interested in determining whether NOis also associated with susceptibility to IB

    Studies have been conflicting and may be poulation dependent; however, two studies h

    demonstrated an association of NOD1 wIBD (132, 133). The first study to identify an

    sociation between NOD1 polymorphisms aIBD involved two independent cohorts co

    prising over 1000 IBD patients in England

    these patients the common deletion allelea complex polymorphism (ND1+326561) w

    significantly associated with IBD and wit

    386 Chen et al.

  • 8/6/2019 Annurev.pathol NOD

    23/36

  • 8/6/2019 Annurev.pathol NOD

    24/36

  • 8/6/2019 Annurev.pathol NOD

    25/36

    such as BS, researchers have attempted

    to determine whether NOD2 variants are

    also associated with disease susceptibility forsarcoid. However, unlike with NOD1, no

    correlation between NOD2 mutations and sar-coid has been observed (162164). Interest-

    ingly, however, NOD2 has been associated with juvenile-onset sarcoid (EOS). This disease is

    distinct from adult sarcoidosis in that it usuallyis seen in individuals younger than four years

    with the clinical triad of skin, joint, and eyeinvolvementsimilar to that seen in BS, but

    lacking pulmonary findings. In a small studyof 10 Japanese patients diagnosed with EOS,

    nine had heterozygous missense mutations in

    the NOD domain of NOD2, totaling six dif-ferent variants (165). Only one variant was

    identical to that reported with Blau syndrome,while the remaining five were novel. Regard-

    less, EOS-associated NOD2 variants, like theirBS-associated NOD2 counterparts, exhibited

    increased basal NF-B activity compared towild type that was further enhanced with the

    addition of MDP in a reporter assay in vitro(165).

    Allergic diseases. Allergies are associated

    with hyperreactivity to antigens; they are char-acterized by increased immunoglobin E re-

    sponse and are therefore inflammatory innature. Certain NOD1 genetic variants are

    associated with an increased risk of develop-ing asthma and atopic eczema, and therefore

    NOD1 may play a role in the modulation of amucosal allergic response (166, 167). Because

    asthma results from gene-environment inter-actions, its association with NOD1 may impli-

    cate bacterial signaling through NOD1 in thepathogenesis of asthma. This would be consis-

    tent with the prevailing hygiene hypothesis

    of asthma risk: Studies have demonstrated thatindividuals with reduced exposure to bacteria

    during childhood have an increased risk of de-veloping asthma (168170). Moreover, NOD1

    polymorphisms can also modify the protectiveeffect of early exposure to allergens (171), pro-

    vidingadditional evidence for a possible role for

    NOD1 in modulating the response to environ-

    mental bacteria and development of asthma.

    Cancer. Recent studies have suggested thatTLR signaling plays an important role in car-

    cinogenesis, especially within the gastrointesti-

    nal tract (172, 173). A role for NLRs has not yetbeen investigated; however, a tumor-suppressor

    function in NOD1 has been suggested in breastcancer xenograft models (174). This phenotype

    was attributed to a functional role for NOD1in regulating apoptosis, in which retrovirally

    infected MCF-7 cells with defective NOD1expression were more sensitive to TNF--

    induced apoptosis in vitro in the presence ofcyclohexamide, which facilitates apoptosis by

    inhibiting expression of antiapoptotic factors.In vivo, this mutant cell line also showed in-

    creased growth potential as xenografts; this po-

    tential was not associated with decreased apop-tosis, but rather with a lack of responsiveness

    to estrogen-induced proliferation, suggestingan additional role for NOD1 in regulating es-

    trogen receptor expression. However, sponta-neous growth of tumors has not been observed

    in Nod1-deficient mice, and whether NOD1influences tumor growth in other organs that

    are not necessarilyhormone responsive remainsto be determined.

    CONCLUSIONS ANDPERSPECTIVES

    The identification and initial characterization

    of NLR proteins have yielded new insights intothe host recognition system involved in micro-

    bial detection and host-defense mechanisms.The involvement of NLRs in the pathogenesis

    of several genetic diseases indicates that theseproteins play an important role in the regula-

    tion of immune and inflammatory responses.

    There is conclusive evidence that several NLRssense conserved microbial molecules to activate

    discrete signaling pathways including NF-B,MAPK, and caspase-1 activation. There is also

    clear evidence for interplay between NLRs andTLRs in the regulation of the inflammatory re-

    sponse against microbes.

    www.annualreviews.org NLRs: Role in Immunity and Disease 389

  • 8/6/2019 Annurev.pathol NOD

    26/36

  • 8/6/2019 Annurev.pathol NOD

    27/36

  • 8/6/2019 Annurev.pathol NOD

    28/36

  • 8/6/2019 Annurev.pathol NOD

    29/36

    44. Kanneganti TD, Body-Malapel M, Amer A, Park JH, Whitfield J, et al. 2006. Critical role for

    Cryopyrin/Nalp3 in activation of caspase-1 in response to viral infection and double-stranded RNA.

    J. Biol. Chem. 281:3656068

    45. Sutterwala FS, Ogura Y, Szczepanik M, Lara-Tejero M, Lichtenberger GS, et al. 2006. Critical role

    for NALP3/CIAS1/Cryopyrin in innate and adaptive immunity through its regulation of caspase-1.

    Immunity 24:31727

    46. Mariathasan S, Weiss DS, Newton K, McBride J, ORourke K, et al. 2006. Cryopyrin activates the

    inflammasome in response to toxins and ATP. Nature 440:22832

    47. Mariathasan S, Newton K, Monack DM, Vucic D, French DM, et al. 2004. Differential activation of theinflammasome by caspase-1 adaptors ASC and Ipaf. Nature 430:21318

    48. Amer A, Franchi L, Kanneganti TD, Body-Malapel M, Ozoren N, et al. 2006. Regulation ofLegionella

    phagosome maturation and infection through flagellin and host Ipaf. J. Biol. Chem. 281:3521723

    49. Franchi L, Amer A, Body-MalapelM, Kanneganti TD,Ozoren N, et al.2006. Cytosolic flagellin requires

    Ipaf for activation of caspase-1 and interleukin 1 in salmonella-infected macrophages. Nat. Immunol.

    7:57682

    50. Miao EA, Alpuche-Aranda CM, Dors M, Clark AE, Bader MW, et al. 2006. Cytoplasmic flagellin

    activates caspase-1 and secretion of interleukin 1 via Ipaf. Nat. Immunol. 7:56975

    51. Fink SL, Cookson BT. 2007. Pyroptosis and host cell death responses during Salmonella infection. Cell

    Microbiol. 9:256270

    52. Fink SL, Bergsbaken T, Cookson BT. 2008. Anthrax lethal toxin and Salmonella elicit the common cell

    death pathway of caspase-1-dependent pyroptosis via distinct mechanisms. Proc. Natl. Acad. Sci. USA105:431217

    53. Sansonetti PJ, Phalipon A, Arondel J, Thirumalai K, Banerjee S, et al. 2000. Caspase-1 activation of

    IL-1 and IL-18 are essential for Shigella flexneriinduced inflammation. Immunity 12:58190

    54. Mariathasan S, Weiss DS, Dixit VM, Monack DM. 2005. Innate immunity against Francisella tularensis

    is dependent on the ASC/caspase-1 axis. J. Exp. Med. 202:104349

    55. Tsuji NM, Tsutsui H, Seki E, Kuida K, Okamura H, et al. 2004. Roles of caspase-1 in Listeria infection

    in mice. Int. Immunol. 16:33543

    56. Bergsbaken T, Cookson BT. 2007. Macrophage activation redirects Yersinia-infected host cell death

    from apoptosis to caspase-1-dependent pyroptosis. PLoS Pathog. 3:e161

    57. Inohara N, Ogura Y, Chen FF, Muto A, Nunez G. 2001. Human Nod1 confers responsiveness to

    bacterial lipopolysaccharides. J. Biol. Chem. 276:255154

    58. Hasegawa M, Yang K, Hashimoto M, Park JH, Kim YG, et al. 2006. Differential release and distributionof Nod1 and Nod2 immunostimulatory molecules among bacterial species and environments. J. Biol.

    Chem. 281:2905463

    59. Girardin SE, Boneca IG, Carneiro LA, Antignac A, Jehanno M, et al. 2003. Nod1 detects a unique

    muropeptide from gram-negative bacterial peptidoglycan. Science 300:158487

    60. Chamaillard M, Hashimoto M, Horie Y, Masumoto J, Qiu S, et al. 2003. An essential role for NOD1 in

    host recognition of bacterial peptidoglycan containing diaminopimelic acid. Nat. Immunol. 4:7027

    61. Inohara N, Ogura Y, Fontalba A, Gutierrez O, Pons F, et al. 2003. Host recognition of bacterial muramyl

    dipeptide mediated through NOD2. Implications for Crohns disease. J. Biol. Chem. 278:550912

    62. McDonald C, Inohara N, Nunez G. 2005. Peptidoglycan signaling in innateimmunityand inflammatory

    disease. J. Biol. Chem. 280:2017780

    63. Hasegawa M, Kawasaki A, Yang K, Fujimoto Y, Masumoto J, et al. 2007. A role of lipophilic

    peptidoglycanrelated molecules in induction of Nod1-mediated immune responses. J. Biol. Chem.282:1175764

    64. Nigro G, Fazio LL, Martino MC, Rossi G, Tattoli I, et al. 2008. Muramylpeptide shedding modulates

    cell sensing ofShigella flexneri. Cell Microbiol. 10:68295

    65. Kufer TA, Kremmer E, Adam AC, Philpott DJ, Sansonetti PJ. 2008. The pattern-recognition molecule

    Nod1 is localized at the plasma membrane at sites of bacterial interaction. Cell Microbiol. 10:47786

    66. Viala J, Chaput C, Boneca IG, Cardona A, Girardin SE, et al. 2004. Nod1 responds to peptidoglycan

    delivered by the Helicobacter pyloricag pathogenicity island. Nat. Immunol. 5:116674

    www.annualreviews.org NLRs: Role in Immunity and Disease 393

  • 8/6/2019 Annurev.pathol NOD

    30/36

    67. Zilbauer M, Dorrell N, Boughan PK, Harris A, Wren BW, et al. 2005. Intestinal innate immun

    to Campylobacter jejuni results in induction of bactericidal human -defensins 2 and 3. Infect. Imm

    73:728189

    68. Kim JG, Lee SJ, Kagnoff MF. 2004. Nod1 is an essential signal transducer in intestinal epithelial c

    infected with bacteria that avoid recognition by toll-like receptors. Infect. Immun. 72:148795

    69. Opitz B, Forster S, Hocke AC, Maass M, Schmeck B, et al. 2005. Nod1-mediated endothelial c

    activation byChlamydophila pneumoniae. Circ. Res. 96:31926

    70. Travassos LH, Carneiro LA, Girardin SE, Boneca IG, Lemos R, et al. 2005. Nod1 participates in

    innate immune response to Pseudomonas aeruginosa. J. Biol. Chem. 280:367141871. Welter-Stahl L, Ojcius DM, Viala J, Girardin S, Liu W, et al. 2006. Stimulation of the cytosolic recep

    for peptidoglycan, Nod1, by infection with Chlamydia trachomatisor Chlamydia muridarum. Cell Micro

    8:104757

    72. Opitz B, Puschel A, Schmeck B, Hocke AC, Rosseau S, et al. 2004. Nucleotide-binding oligomerizat

    domain proteins are innate immune receptors for internalized Streptococcus pneumoniae. J. Biol. Ch

    279:3642632

    73. Herskovits AA, Auerbuch V, Portnoy DA. 2007. Bacterial ligands generated in a phagosome are targ

    of the cytosolic innate immune system. PLoS Pathog. 3:e51

    74. Ferwerda G, Girardin SE, Kullberg BJ, Le Bourhis L, de Jong DJ, et al. 2005. NOD2 and toll-l

    receptors are nonredundant recognition systems ofMycobacterium tuberculosis. PLoS Pathog. 1:2798

    75. Hisamatsu T, Suzuki M, Reinecker HC, Nadeau WJ, McCormick BA, et al. 2003. CARD15/NO

    functions as an antibacterial factor in human intestinal epithelial cells. Gastroenterology 124:99310076. Kapetanovic R, Nahori MA, Balloy V, Fitting C, Philpott DJ, et al. 2007. Contribution of phagocyt

    and intracellular sensing for cytokine production byStaphylococcus aureusactivated macrophages. Inf

    Immun. 75:83037

    77. Kim YG, Park JH, Shaw MH, Franchi L, Inohara N, et al. 2008. The cytosolic sensors Nod1 a

    Nod2 are critical for bacterial recognition and host defense after exposure to Toll-like receptor ligan

    Immunity 28:24657

    78. Abreu MT, Thomas LS, Arnold ET, Lukasek K, Michelsen KS, et al. 2003. TLR signaling at t

    intestinal epithelial interface. J. Endotoxin Res. 9:32230

    79. Melmed G, Thomas LS, Lee N, Tesfay SY, Lukasek K, et al. 2003. Human intestinal epithelial cells

    broadly unresponsive to Toll-like receptor 2dependent bacterial ligands: implications for host-micro

    interactions in the gut. J. Immunol. 170:140615

    80. Warren SE, Mao DP, Rodriguez AE, Miao EA, Aderem A. 2008. Multiple Nod-like receptors activcaspase-1 during Listeria monocytogenesinfection. J. Immunol. 180:755864

    81. Molofsky AB, Byrne BG, Whitfield NN, Madigan CA, Fuse ET, et al. 2006. Cytosolic recognition

    flagellin by mouse macrophages restricts Legionella pneumophila infection. J. Exp. Med. 203:109310

    82. Suzuki T, Franchi L, Toma C, Ashida H, Ogawa M, et al. 2007. Differential regulation of caspas

    activation, pyroptosis, and autophagy via Ipaf and ASC in Shigella-infected macrophages. PLoS Pat

    3:e111

    83. Yamamoto Y, Klein TW, Friedman H. 1991. Legionella pneumophila growth in macrophages from s

    ceptible mice is genetically controlled. Proc. Soc. Exp. Biol. Med. 196:4059

    84. Yamamoto Y, Klein TW, Friedman H. 1992. Genetic control of macrophage susceptibility to infect

    byLegionella pneumophila. FEMS Microbiol. Immunol. 4:13745

    85. Fortier A, Min-Oo G, Forbes J, Lam-Yuk-Tseung S, Gros P. 2005. Single gene effects in mouse mod

    of host/pathogen interactions. J. Leukoc. Biol. 77:8687786. Fortier A, de Chastellier C, Balor S, Gros P. 2007. Birc1e/Naip5 rapidly antagonizes modulation

    phagosome maturation byLegionella pneumophila. Cell Microbiol. 9:91023

    87. Zamboni DS,Kobayashi KS,Kohlsdorf T, Ogura Y, Long EM, et al. 2006. TheBirc1ecytosolic patte

    recognition receptor contributes to the detection and control of Legionella pneumophila infection. N

    Immunol. 7:31825

    88. Ren T, Zamboni DS, Roy CR, Dietrich WF, Vance RE. 2006. Flagellin-deficient Legionella muta

    evade caspase-1- and Naip5-mediated macrophage immunity. PLoS Pathog. 2:e18

    394 Chen et al.

  • 8/6/2019 Annurev.pathol NOD

    31/36

    89. Lamkanfi M, Amer A, Kanneganti TD, Munoz-Planillo R, Chen G, et al. 2007. The Nod-like receptor

    family member Naip5/Birc1e restricts Legionella pneumophila growth independently of caspase-1 activa-

    tion. J. Immunol. 178:802227

    90. Amer AO, Swanson MS. 2005. Autophagy is an immediate macrophage response to Legionella pneu-

    mophila. Cell Microbiol. 7:76578

    91. Miao EA, Ernst RK, Dors M, Mao DP, Aderem A. 2008. Pseudomonas aeruginosa activates caspase 1

    through Ipaf. Proc. Natl. Acad. Sci. USA 105:256267

    92. Boyden ED, Dietrich WF. 2006. Nalp1b controls mouse macrophage susceptibility to anthrax lethal

    toxin. Nat. Genet. 38:2404493. Kanneganti TD, Ozoren N, Body-Malapel M, Amer A, Park JH, et al. 2006. Bacterial RNA and small

    antiviral compounds activate caspase-1 through cryopyrin/Nalp3. Nature 440:23336

    94. Muruve DA, Petrilli V, Zaiss AK, White LR, Clark SA, et al. 2008. The inflammasome recognizes

    cytosolic microbial and host DNA and triggers an innate immune response. Nature 452:1037

    95. Martinon F, Petrilli V, Mayor A, Tardivel A, Tschopp J. 2006. Gout-associated uric acid crystals activate

    the NALP3 inflammasome. Nature 440:23741

    96. Locovei S, Wang J, Dahl G. 2006. Activation of pannexin 1 channels by ATP through P2Y receptors

    and by cytoplasmic calcium. FEBS Lett. 580:23944

    97. Pelegrin P, Surprenant A. 2006. Pannexin-1 mediates large pore formation and interleukin-1 release

    by the ATP-gated P2X7 receptor. EMBO J. 25:507182