ebook img

Recognition of damage-associated molecular patterns related to nucleic acids during inflammation and vaccination. PDF

1 MB·English
by  JounaiNao
Save to my drive
Quick download
Download
Most books are stored in the elastic cloud where traffic is expensive. For this reason, we have a limit on daily download.

Preview Recognition of damage-associated molecular patterns related to nucleic acids during inflammation and vaccination.

REVIEWARTICLE published:08January2013 CELLULAR AND INFECTION MICROBIOLOGY doi:10.3389/fcimb.2012.00168 Recognition of damage-associated molecular patterns related to nucleic acids during inflammation and vaccination NaoJounai1,2,KoujiKobiyama1,2,FumihikoTakeshita1,2 andKenJ.Ishii1,2* 1LaboratoryofAdjuvantInnovation,NationalInstituteofBiomedicalInnovation,Osaka,Japan 2LaboratoryofVaccineScience,WPIImmunologyFrontierResearchCenter,OsakaUniversity,Osaka,Japan Editedby: All mammalian cells are equipped with large numbers of sensors for protection NelsonGekara,UmeaUniversity, from various sorts of invaders, who, in turn, are equipped with molecules containing Sweden pathogen-associatedmolecularpatterns(PAMPs).Oncethesesensorsrecognizenon-self Reviewedby: antigens containing PAMPs, various physiological responses including inflammation are DarioS.Zamboni,Universidadede induced to eliminate the pathogens. However, the host sometimes suffers from chronic SãoPaulo,Brazil WillemVanEden,Utrecht infection or continuous injuries, resulting in production of self-molecules containing University,Netherlands damage-associated molecular patterns (DAMPs). DAMPs are also responsible for the YanShi,UniversityofCalgary, eliminationofpathogens,butpromiscuousrecognitionofDAMPsthroughsensorsagainst Canada PAMPs has been reported. Accumulation of DAMPs leads to massive inflammation and *Correspondence: continuous production of DAMPs; that is, a vicious circle leading to the development of KenJ.Ishii,LaboratoryofAdjuvant Innovation,NationalInstitute autoimmunedisease.Fromavaccinologicalpointofview,theaccuraterecognitionofboth ofBiomedicalInnovation, PAMPsand DAMPsis important for vaccine immunogenicity, because vaccine adjuvants 7-6-8Saito-Asagi,Ibaraki, are composed of several PAMPs and/or DAMPs, which are also associated with severe Osaka567-0085,Japan. adverseeventsaftervaccination.Here,wereviewastherolesofPAMPsandDAMPsupon e-mail:[email protected] infection with pathogens or inflammation, and the sensors responsible for recognizing them, as well as their relationship with the development of autoimmune disease or the immunogenicityofvaccines. Keywords: PAMPs (pathogen-associatedmolecular patterns), DAMPs (damage-associatedmolecularpatterns), nucleicacids,metabolites,innateimmunity,DNAsensors,uricacid,vaccineadjuvant INTRODUCTION whereas continuous inflammatory responses owing to impaired Host cells are equipped with numerous types of receptors to regulationofinflammatorysignalingresultsinchronicinflamma- discriminateselffromnon-self.Whencellsareattackedbyinfec- torydiseaseorautoimmunedisease.Therefore,“bipolarsensors” tiouspathogens,hostcellularreceptorssuchasToll-likereceptors for both PAMPs andDAMPs appear to be the mostly responsi- (TLRs), nucleotide oligomerization domain (NOD)-like recep- blefordysregulatedinflammation.Here,wedescribethevarious tors(NLRs),retinoicacid-induciblegene-I(RIG-I)-likereceptors types of DAMPs and their receptors, with a special focus on (RLRs), C-type lectin receptors, andother non-classifiedrecep- nucleicacidsasDAMPs. torsrecognizepathogen-associatedmolecularpatterns(PAMPs), small molecular motifs conserved amongst microbes. Through LIPID-RELATEDDAMPs the recognition of PAMP molecules, innate immune responses LIPOPOLYSACCHARIDE(LPS) are induced, and inflammatory cytokines areproduced that aid A representative lipid for the induction of inflammatory in the elimination of the pathogens. However, in some circum- responses is LPS, a PAMP present in gram-negative bacte- stances host inflammatory responses can cause host cell death ria. Upon recognition by TLR4, LPS promotes the production leading to tissue injury, and the release of host cellular com- of various inflammatory cytokines following bacterial infection ponents to the extracellular environment. These cellular com- (Table1). However, Shi et al. reported that, TLR4 also rec- ponents could be considered “messengers” for danger; they are ognizes endogenous fatty acids and can activate inflammatory alsoknownas“damage-associatedmolecularpatterns”(DAMPs). responses in adipocytes and macrophages (Shi et al., 2006). DAMPs include lipids, sugars, metabolites, and nucleic acids In addition, TLR4-deficient mice developed reduced inflamma- such as RNA and DNA species. DAMPs are important for the tory cytokine production in response to a high fat diet (Shi elimination of pathogens, but are also implicated in the devel- et al., 2006). Previous studies have revealed that saturated fatty opment of autoimmune disease and chronic inflammatory dis- acids are released from hypertrophied adipocytes in the pres- ease, and are used as adjuvants for vaccines. Interestingly, high enceofmacrophages,andthatreleasedfattyacidsaresensedby numbersofPAMPreceptorsalsorecognizeendogenousDAMPs macrophages in a TLR4-dependent manner, following excessive and can augment inflammatory responses against pathogens, production of inflammatory cytokines such as tumor necrosis FrontiersinCellularandInfectionMicrobiology www.frontiersin.org January2013|Volume2|Article168|1 Jounaietal. Immunesensorsfornucleicacids Table1|AssociationofPAMPorDAMPsensorswithautoimmunediseases. Receptor PAMP DAMP Autoimmunedisease TLR1/TLR2 Lipopeptide SerumamyloidAprotein Atherosclerosis,rheumatoidarthritis,Crohn’sdisease TLR4 LPS Fattyacid Obesity Hyaluronicacid Rheumatoidarthritis,sarcoidosis,systemicsclerosis, pancreaticcancer NLRP3 Uricacid Uricacid Hyperuricemia,gout ATP Unknown RIG-I,MDA5,TLR7/8 VirusRNA ImmunocomplexofsnRNPs SLE TLR9 BacterialDNA Self-DNA-containingimmune SLE complexes,histone RAGE −/? HMGB1 SLE DAI,IFI16,AIM2,H2B, BacterialDNA,VirusDNA Self-DNA? SLE? RNApolIII factor(TNF)-α(Suganamietal.,2007).Becausetheproduction SUGAR-RELATEDDAMPs of pro-inflammatory or inflammatory cytokines is dysregulated Hyaluronic acid (HA) is a non-sulfated linear polysaccharide, in obese adipose tissues, obesity can be thought of as a chronic andamajorcomponentoftheextracellularmatrix.Weigeletal. inflammatory disease caused by fatty acids acting as DAMP revealedthatHAisinducedanddegradedduringinflammatory molecules(BergandScherer,2005). responsesandthatitfunctionsinimmunecellactivationornew bloodvessel formation (Weigel et al.,1986). Interestingly, small SERUMAMYLOIDAPROTEIN(SAA) molecular weight HA (sHA), produced by the degradation of Some lipoproteins can also act as DAMP molecules. In 1982, HA during inflammation, can induce the maturation of den- Hoffman and Benditt revealed that the treatment of mice with driticcells(DCs)forpathogenelimination(Termeeretal.,2002). LPS of Salmonella typhosa increased SAA levels (Hoffman and Bonemarrow-derivedDCsfrommiceexpressingnon-functional Benditt, 1982). According to several studies, SAA functions in TLR4 could not be activated by sHA, while DCs from TLR2- cholesterol transport as well as inthe production of proinflam- deficient mice retained the ability for sHA-mediated activation. matory cytokines, suggesting that SAA is a DAMP molecule This suggests that sHA can act as a DAMP molecule signaling that responds to bacterial endotoxins (Banka et al., 1995; He throughTLR4toinduceDCmaturationuponpathogeninfection et al., 2003). In support of this, increased levels of SAA may (Termeeretal.,2002).Consistentwiththis,excessive sHAlevels be closely related to various diseases such as atherosclerosis, appearedtobecloselyassociatedwithinflammatoryautoimmune rheumatoidarthritis,andCrohn’sdisease(Chambersetal.,1983, diseasessuchasrheumatoidarthritis,sarcoidosis,systemicsclero- 1987; Malle and De Beer, 1996). SAA binds to two recep- sis,andpancreaticcancer(Hallgrenetal.,1985;Witteretal.,1987; tors, TLR4 and TLR2, which also recognize bacterial PAMP Sugaharaetal.,2006;Yoshizakietal.,2008)(Table1). molecules such as triacyl lipopeptides (in cooperation with TLR1), diacyl lipopeptides or lipoteichoic acids (together with METABOLITE-RELATEDDAMPs TLR6) (Schwandner et al., 1999; Takeuchi et al., 2001, 2002; URICACID Cheng et al., 2008; Hiratsuka et al., 2008) (Table1). Recently, Uricacidisametabolite ofpurinenucleotidesandfreebasesin Loser et al. showed direct evidence for the local production humans and other primates, and it functions as an antioxidant of the SAA molecules myeloid-related protein-8 (Mrp8) and toprotecterythrocytemembranesfromlipidoxidation(Kellogg + Mrp14, which induced autoreactive CD8 T cells and sys- andFridovich,1977).However,itwaspreviouslyshownthatsol- temic autoimmunity through TLR4 signaling in mice (Loser ubleuricacid-inducedinflammatorycytokinessuchasmonocyte et al., 2010). Taken together, these findings suggest that TLR4 chemoattractant protein-1 in rat vascular smooth muscle cells may be a key receptor in the discrimination of lipid PAMPs (Kanellis et al., 2003). Shi et al. also reported that uric acid is from lipid DAMPs molecules, because promiscuous recogni- producedinultraviolet-irradiatedBALB/c3T3cells,andactivates tion of lipids via TLR4 unfortunately causes inflammatory dis- DCs(Shietal.,2003).Inaddition,highlevelsofuricacidinthe ease. Although a consensus recognition structure for TLR4 has bloodareassociatedwiththedevelopmentofhyperuricemiaand not yet been identified, antagonists of TLR4 signaling by lipid- gout(Johnsonetal.,2005),suggestingthatitactsasaDAMPdur- DAMPs might be candidate drugs for the treatment of chronic ing cell injury and can induce inflammatory responses that are inflammatorydisease. relatedtoautoinflammatorydiseasessuchasgout(Table1). FrontiersinCellularandInfectionMicrobiology www.frontiersin.org January2013|Volume2|Article168|2 Jounaietal. Immunesensorsfornucleicacids Receptors that recognize uric acid have been reported and purinemetabolites.Nucleicacidsexistinallorganismsincluding Liu-Bryan et al. revealed that TLR2, TLR4, and their adaptor pathogens, and function as a store of genetic information for moleculeMyD88areimportantforuricacid-mediatedinflamma- protein translation and synthesis. Bacterial genomic DNA can tion(Liu-Bryanetal.,2005).Incontrast,theuricacid-mediated be recognized as a PAMP, as it contains unmethylated CpG activation of DCs was shown to be TLR4-independent, sug- motifs whose frequency is higher in genomic DNA derived gesting the possible existence of other receptors that recognize from pathogens compared with that of vertebrates. The earli- uric acid in addition to TLR2 and TLR4 (Shi et al., 2003). est research related to bacterial genomic DNA as PAMPs was To solve this question, Martinon et al. demonstrated that uric reported more than hundred years ago. Bruns et al. inves- acid could be sensed by another receptor, NOD-like recep- tigated heat-killed gram-negative or gram-positive bacteria as tor family, pyrin domain-containing 3 (NLRP3), and induced an immunotherapeutic agent termed Coley’s toxin, for cancer to produce interleukin (IL)-1β through caspase-1 activation (Swain, 1895). Although LPS is a major factor in mediating (Martinon et al., 2006). NLRP3 is a member of the NLR fam- anti-tumoreffects,otherfactorsmaybeconnectedwithitsphysi- ily, and a component of the inflammasome, a platform that ological function,as gram-positive bacteria donotexpress LPS. inducesIL-1βandIL-18production.NLRP3sensesvarioustypes A hundred years on from the discovery of Coley’s toxin, sev- of pathogen infections or irritants such as Candida albicans, eral studies have shown that bacterial DNA can activate nat- Legionella pneumophila, Listeria monocytogenes, Malaria hemo- ural killer (NK) cells or B cells, suggesting that the bacterial zoin, alum, silica, and asbestos as well as uric acid (Kanneganti genomic DNA in Coley’s toxin could contribute to its anti- et al., 2006; Martinon et al., 2006; Dostert et al., 2008, tumor activity by stimulating NK cells (Shimada et al., 1986; 2009; Eisenbarth et al., 2008; Gross et al., 2009). Collectively, Messina et al., 1991). Krieg et al. further revealed that bac- these results revealed that NLRP3 is a promiscuous receptor terial genomic DNA contains unmethylated CpG motifs that that senses PAMPs and DAMPs and can induce inflammatory can stimulate B cells and NK cells, and induce inflammatory responses. cytokine production. Interestingly, methylated bacterial DNA failed to stimulate immune cells, indicating that unmethylated ADENOSINETRIPHOSPHATE(ATP) CpG motifs may act as PAMP molecules (Krieg et al., 1995; ATP is an essential purine base required for almost all physi- Klinman et al., 1996). However, whether genomic DNA con- cal responses such as glucose metabolism, muscle contraction, taining methylated CpG motifs is incapable of innate immune biosynthesis, and molecular transfer. However, extracellular activationremainscontroversial.In1962,Glasgowetal.reported ATP from injured cells or non-apoptotic cells also serves thatultraviolet-inactivatedvacciniavirus,aDNAvirus,resulted as a danger signal through the activation of NLRP3 and in IFN production in mouse cells (Glasgow and Habel, 1962). caspase-1 (Communi et al., 2000). Previous detailed research In addition, Suzuki et al. showed that viral DNA, vertebrate has shown the importance of other ion channel molecules, DNA and bacterial DNA induced the upregulation of major namely, P2X7 and pannexin-1, in inducing extracellular ATP- histocompatibility complex (MHC) class I expression and the mediated caspase-1 activation following IL-1β maturation type I IFN-related activation of transcription factors such as (Ferrari et al., 2006; Kanneganti et al., 2007). The formation STAT3 in rat thyroid cells, suggesting that genomic DNA also of the NLRP3 inflammasome requires an adaptor molecule, activates innateimmunesignalingin aCpG-motif-independent apoptosis-associated speck-like protein containing a carboxy- manner (Suzuki et al., 1999). Interestingly, the structure of terminalcaspaserecruitmentdomain(ASC).ASC-deficientmice DNAstronglyaffects DNA-mediatedinnateimmuneactivation. cannot activate caspase-1 and thus do not produce mature Double-stranded, right-handed B-form DNA, but not the left- IL-1β following exposure to large amounts of ATP, suggest- handed Z-form DNA, strongly induced type I IFN production. ing that ATP-mediated IL-1β production is dependent on the Genomic DNA has a high content of B-form DNA, indicating NLRP3 inflammasome (Mariathasan et al., 2004). However, that it may also function as a PAMP or DAMP (Ishii et al., althoughextracellularATPhasbeensuggestedtoactasaDAMP 2006). Mitochondrial DNA has been also reported to function molecule, there is no correlation between high amounts of as a DAMP molecule. Zhang et al. reported that cellular injury extracellular ATP acting as DAMPs in vitro and physiological causedthereleaseofmitochondrialDNA,andinducedsystemic conditions in vivo. Eckle et al. suggested that most extracel- inflammatory responses via p38 MAPK activation in a TLR9- lular ATP might be immediately hydrolyzed by ectonucleoti- dependent manner. In addition, trauma patients had higher dases (Eckle et al., 2007). Taken together, investigation into amountsofmitochondrialDNAthandidhealthyvolunteers,sug- the roles of extracellular ATP in inducing pathological and gesting that mitochondrial DNA could be considered a marker immuneresponses invivo may provideimportant cluesregard- of inflammatory disease (Zhang et al., 2010). When the clear- ingthemechanismunderlyinginflammationinductionbyDAMP ance of mitochondrial DNA by autophagy was inhibited, IL-1β molecule recognition or in the development of inflammatory productionwasaugmentedviatheNLRP3inflammasometoacti- diseases. vatecaspase-1,indicatingthattheamountofmitochondrialDNA DAMPactivity isregulated byautophagyto suppresserroneous NUCLEICACID-RELATEDDAMPs activationofinnateimmunity(Nakahiraetal.,2011).Indeed,it UNMETHYLATEDCpGMOTIFANDGENOMICDNA was revealedthatautophagynegatively regulates RNA-mediated As described above, uric acid and ATP are products of purine typeIIFNproduction,possiblytomaintaincellularhomeostasis metabolism. Nucleic acids such as adenine or guanine are also (Jounaietal.,2007). FrontiersinCellularandInfectionMicrobiology www.frontiersin.org January2013|Volume2|Article168|3 Jounaietal. Immunesensorsfornucleicacids CORRELATIONBETWEENAUTOIMMUNEDISEASEANDDNADAMPs TLRs BothDNAandRNAcanfunctionasPAMPsandDAMPs,andare AlargebodyofresearchexistsdemonstratingtheTLR-mediated closelyconnectedwithinflammatoryresponsesandthedevelop- sensing of nucleic acids. TLR3 preferentially senses double- ment of inflammatory disease. Direct evidence for DNA acting strandedRNA(dsRNA)species,whichcanoriginatefromsome as a DAMP was shown using DNase-deficient mice. DNase I is viruses,andTLR3isassociatedwithinductionofinnateimmu- presentinextracellularcompartmentssuchastheseraandurine, nityinresponsetoinfectionwithWestNilevirus,respiratorysyn- andfunctionstodegradesingle-strandedDNA(ssDNA),double- cytialvirus, andencephalomyocarditis virus (Wang et al.,2004; strandedDNA(dsDNA),orchromatin,whicharereleasedfrom Groskreutz et al., 2006; Hardarson et al., 2007) (Figure2). In damaged or necrotic cells. Napirei et al. constructed DNase I- addition,artificialdsRNA,poly(I:C),hasbeenwell-characterized deficientmice,andreportedthattheypresentedwiththeclassical asaligandfor TLR3.Althoughpathogen-relateddsRNAsact as symptomsofsystemiclupuserythematosus(SLE)andglomeru- PAMPs,Karikoetal.reportedthathostmessengerRNAcouldbe lonephritis(Napireietal.,2000).Inaddition,DNaseIIdeficient sensedbyTLR3toinduceinflammatoryresponses(Karikoetal., mice showed a similar phenotype to DNase I knockout mice. 2004).RNAreleasedfromnecroticcellscanalsoelicittypeIIFN DNaseII inthelysosomes of macrophages degrades DNAfrom production,suggestingthathostRNAmightfunctionasaDAMP apoptotic cells or nuclear genome DNA from liver erythrob- uponcellularinjury(Karikoetal.,2004). lasts.Interestingly,DNaseII-deficientmicepresentedwithlethal TLR7 and TLR8 recognize single-stranded RNA (ssRNA), anemia owing to high levels of type I IFN production, caused andinduceanti-viralinnateimmuneresponsesagainstinfluenza by the accumulation of non-degraded genomic DNA in liver virusorvesicularstomatitisvirus(Lundetal.,2004)(Figure2). macrophages(Yoshidaetal.,2005). Insupportofthis,DNaseII Regardlessoftheircommonligands,thecellularandtissuedistri- andIFNRa/bdoubleknockoutmiceshowedanon-lethalpheno- butionofTLR7expressionisincontrasttothatofTLR8.Human type,butdevelopedrheumatoidarthritis-likesymptoms(Kawane TLR7ishighlyexpressedinplasmacytoidDCsthatpreferentially et al., 2006), which could be attenuated by anti-TNF-α anti- inducetypeIIFNproduction,andisexpressedatlowerlevelsin bodytreatment.Thissuggestedthattheaccumulationofgenomic myeloidcells.Conversely,thelevelofTLR8expressionishigherin DNA in macrophages induced inflammatory cytokines, includ- monocytes andinmonocyte-derived DCsthanin plasmacytoid ing type I IFNs and TNF-α, and the synergistic action of these DCs(Hornungetal.,2002).Furthermore,mouseTLR8didnot inflammatorycytokinesresultedinlethalsystemicinflammation respond to ssRNA,but human TLR8 did, suggesting that TLR8 (Kawane et al., 2006). Furthermore, studies on DNase III, also might be inactivated in mice, although several papers have also known as TREX1, also revealed that DNA could function as a linkedmouseTLR8withneuronalapoptosisandautoimmunity DAMP.TREX1isthemajor3(cid:2) →5(cid:2) DNAexonucleaseforDNA (Heiletal.,2004;Gordenetal.,2006;Maetal.,2006). editinginDNAreplicationorDNArepair.Moritaetal.showed In addition to the recognition of PAMPs, Vollmer et al. that trex1-deficient mice had a reduced survival rate owing to revealed that promiscuous recognition through TLR7 or TLR8 high susceptibility to inflammatory myocarditis, although null causes the development of SLE with high levels of type I mice showednospontaneous mutationsortumor development IFNs and TNF-α production (Vollmer et al., 2005). Because (Moritaetal.,2004).Toexplainwhytrex1-deficientmicedevelop the sera from SLE patients contains high levels of autoantibod- inflammatory myocarditis, Crow et al. demonstrated that the ies against self-antigens, such as small nuclear ribonucleopro- mutationinthetrex1genethatabolishedTREX1enzymeactiv- tein particles (snRNPs) includingssRNA,TLR7, or TLR8 could ity was responsible for the development of Aicardi-Goutieres recognize the immunocomplex of snRNPs with autoantibodies syndrome (AGS), a severe neurological brain disease with high thorough Fc receptor-mediated internalization (Vollmer et al., levels of IFN-α in cerebrospinal fluid or serum, suggesting that 2005). Interestingly, TLR7 appears to be a specific sensor for TREX1 is a suppressor of DNA DAMP-mediated inflammatory theinductionoftypeIIFNproductionfromplasmacytoidDCs, responses (Crow et al., 2006). Furthermore, it was previously whereasTLR8isspecificforTNF-αproductionfrommonocytes shown that the abolishment of interferon regulatory factor 3 inSLEpatients,suggestingthatplasmacytoidDCsandmonocytes (IRF3) or IFN-α receptor 1 ameliorated the AGS symptoms in collaboratetodevelopinflammatoryresponsesinSLEviadistinct trex1-deficientmice(Stetsonetal.,2008).Collectively,thesefind- sensors. ings suggestthatthedysregulation ofself-DNAresults insevere TLR9 senses ssDNA containing unmethylated CpG motifs. inflammatoryresponsessuchashighlevelsoftypeIIFNsleading Previous studies have revealed that TLR9 recognizes genomic toautoinflammatorydisease. DNA from pathogens such as murine cytomegalovirus and Herpes simplexvirustype1ortype2asPAMPs(Hemmi etal., 2000; Lund et al., 2003; Krug et al., 2004a,b) (Figure2). With NUCLEICACIDSENSORS regard to the development of autoinflammatory disease, TLR9 Host cells are equipped with numerous types of receptors to hasbeenalsobeenreportedtorecognizeself-antigenscomplexed recognize nucleic acids as PAMPs or DAMPs. These recep- with autoantibodies. Leadbetter et al. revealed that autoreactive tors function to protect the host from pathogen infection, but Bcellswereactivatedbyachromatin-autoantibodycomplexina may also cause autoimmune disorders by inducing the con- TLR9-andMyD88-dependentmanner(Leadbetter etal.,2002). stitutive activation of inflammatory responses (Figure1). In Inaddition,self-DNA-containingimmunecomplexes,whichare this section, we introduce the well-characterized nucleic acid a well-characterized marker for SLE, were recognized by TLR9 sensors. throughFcγRIIA-mediatedinternalizationinplasmacytoidDCs FrontiersinCellularandInfectionMicrobiology www.frontiersin.org January2013|Volume2|Article168|4 Jounaietal. Immunesensorsfornucleicacids FIGURE1|Autoimmunedisordersmaybeinducedbypromiscuoussensingofnucleicacids. (Means et al., 2005). Thus, immune complexes containingself- mice developed attenuated lupus symptoms (Christensen et al., DNAmaysignalasDAMPsthroughTLR9,althoughextracellular 2006).Inaddition,arecentstudyrevealedthatTLR9suppressed receptors such as FcγRIIA may be required for the delivery of the progression of autoinflammatory disease by antagonizing autoimmunecomplexestotheTLR9-localizingcompartment. TLR7, suggesting that TLR9 counteracts TLR7 upon the recog- As described previously, the subcellular localization of TLRs nition of self-immunocomplexes containing ssRNA or ssDNA is important for the recognition of DNA, because TLR3, 7, 8 (Nickerson et al., 2010). To support the interaction between and9localizeto theendosomal compartment. Previous studies TLR7andTLR9uponthedevelopmentofautoimmunedisease, identifiedthreeadaptormolecules,Unc93B1,PRAT4A,andgp96, Fukuietal.generatedUnc93B1D34A/D34Aknock-inmicetoshow whichareimportantforthetraffickingofTLRstositesforsens- that TLR9 competes with TLR7 for binding to Unc93B1 in the ingtheirligands.Unc93B1functionstocontrolthetraffickingof healthystate,whileTLR7isconstitutivelyactivateduponautoin- TLRs 3, 7, and 9 from the endoplasmic reticulum (ER) to the flammatory responses becauseTLR9hasalower affinityfor the endosome. PRAT4A is localized in the ER and acts as a regula- Unc93B1-likeUnc93B1D34A/D34Amutant(Fukuietal.,2011). torofthesubcellulardistributionofmostTLRsexceptforTLR3. Gp96isamemberoftheheatshockprotein(HSP)90family,and RIG-I-LIKERECEPTORS(RLRs) residesintheERwhereitcontrolsthematurationofTLRs2,4,5, Although TLRs can sense both non-self and self nucleic acids, 7,and9(SaitohandMiyake,2009).BecauseTLR7andTLR9are fibroblasts, and endothelial cells that do not express TLRs also regulatedbythesamemolecularmachinery,thecrosstalkbetween producetypeIIFNsinresponsetoinfectionwithpathogens,indi- TLR7 and TLR9 may affect the sensing of auto-nucleic acids cating the existence of other receptors that sense nucleic acids. and the development of autoinflammatory disease. Christensen Yoneyamaetal.determinedthatacytoplasmicDExD/HboxRNA et al. showed that a deficiency of TLR9 results in malignant helicase, RIG-I, senses infection by RNA viruses as well as arti- symptomsinamousemodeloflupus,despitethelevelsofanti- ficial dsRNA, and induces innate antiviral immune responses body production specific for DNA and chromatin being down- mediated by type I IFNs (Yoneyama et al., 2004) (Figure2). In regulated (Christensen et al.,2005). In contrast, TLR7-deficient addition to RIG-I, melanoma differentiation factor-5 (MDA5) FrontiersinCellularandInfectionMicrobiology www.frontiersin.org January2013|Volume2|Article168|5 Jounaietal. Immunesensorsfornucleicacids FIGURE2|Intracellularsensorsfornucleicacids. and laboratory of genetics and physiology-2 (LGP2) were also (Satoetal.,2009;Nakashimaetal.,2010). Accompanying these identified; these receptors were classified as RLRs because their observations, loss of function single nucleotide polymorphisms protein structures were similar to that of RIG-I (Yoneyama have been found in RIG-I and IPS-1 that are closely related et al., 2005). To induce an anti-pathogen immune response, a to the development of autoimmune disease (Pothlichet et al., CARDdomaininRIG-IandMDA5transmitsdown-streamsig- 2011),suggestingthatinhibitionofRLRsignalingmaybeimpor- nals through homophilic interactions with the CARD adaptor tant in the progression of autoimmune disease. However, as molecule, IFN-β promoter stimulator-1 (IPS-1, also known as describedearlier,excessiveproductionofinflammatorycytokines MAVS,Cardif,orVISA)(Kawaietal.,2005;Meylanetal.,2005; includingtypeIIFNsappearstoresultinautoinflammatorydis- Seth etal.,2005; Xuet al.,2005). Thefunction of LGP2 iscon- ease. In contrast, the dysfunction of RLRs induces poor type I troversial. Some in vitro studies showed that LGP2 negatively IFN production, but leads to autoimmune disease (Nakashima regulatesRIG-I-orMDA5-mediatedinnateimmuneresponsesby et al., 2010; Pothlichet et al., 2011). One possibility to explain competingforbindingwiththeirRNAligands(Yoneyamaetal., this phenomenon is that non-functional RLRs result in an 2005; Bamming and Horvath, 2009). However, in vivo studies increased susceptibility against various types of virus infec- usinglgp2-deficientmicerevealedthatLGP2isacofactorofRLR- tions, and the subsequent virus-mediated cell death may cause mediated innate immune signaling (Venkataraman et al., 2007; the release of DAMPs and signaling through DAMP receptors. Satohetal.,2010). Support this possibility, the loss of MDA5 function increased RLRssensepathogen-derivedRNAspeciesasPAMPstoinduce the susceptibility of beta cells to viral infection with picor- type I IFN production, while MDA5 has been detected as an navirus or encephalomyocarditis virus-D, and resulted in type autoantigen in clinically amyopathic dermatomyositis patients 1 diabetes, whose types of diabetes are often caused by virus (Sato et al., 2009; Nakashima et al., 2010). Although it is infection or autoimmunity (Colli et al.,2010; McCartney et al., not clear how extracellular MDA5 is produced, the accumula- 2011). Further analyses are required to elucidate the cross-talk tion of immunocomplexes containing MDA5 is a marker for between RLR signaling and the development of autoimmune the frequency of rapidly progressive interstitial lung disease disease. FrontiersinCellularandInfectionMicrobiology www.frontiersin.org January2013|Volume2|Article168|6 Jounaietal. Immunesensorsfornucleicacids ABSENTINMELANOMA2(AIM2)-LIKERECEPTORS(ALRs) (Rozzoetal.,2001). Interestingly, p202 levels are varied among AlthoughvariousNLRfamilymembersthatcaninducetheacti- mousespecies,whileAIM2isexpressedatthesamelevel,indicat- vation of caspase-1 and maturation of IL-1β, IL-18, and IL-33 ingthatp202expressionistightlycorrelatedtoSLEdevelopment. in response to a wide range of PAMP and DAMP molecules Furthermore, Ravichandran et al. revealed that ablation of the havebeenidentified,nosensorofintracellulardsDNAforIL-1β aim2geneleadstohigherexpressionofp202andtypeIIFNsin maturation has been identified. However, four research groups mice, andaim2-deficientmice areproneto SLE (Panchanathan concurrently reported a role for the novel intracellular DNA et al., 2010). Taken together, these findings suggest that mouse sensor,AIM2,intheactivationofcaspase-1followingIL-1βpro- p202mightbehomologous tohumanIFI16. Insupportofthis, duction (Burckstummer et al., 2009; Fernandes-Alnemri et al., expression levels of IFI16 and anti-IFI16 autoantibodies were 2009; Hornungetal.,2009; Roberts etal.,2009). AIM2belongs dramaticallyincreasedinSLEpatients,indicatingthatIFI16has to a family of hematopoietic interferon-inducible nuclear pro- similarfeaturestop202(Mondinietal.,2006). teins with a 200-amino acid repeat (HIN-200), known as the A recent article described a correlation between psoriasis p200orPYHINfamily.Currently,fourHIN-200familymolecules symptoms and AIM2 activation. Psoriasis is a chronic autoin- havebeenidentifiedinhumans,andsixinmice.HIN-200fam- flammatorydiseasecausedbyincreasedIL-1βproductionleading ilymolecules sharesimilarstructural features,includingapyrin to Th17 cell maturation (Ghoreschi et al., 2010). Dombrowski domain at the NH terminus, and a HIN-200 domain at the et al. observed increased levels of cytosolic DNA fragments in 2 COOHterminus.SimilartotheroleofNLRP3inIL-1βproduc- skin lesions from psoriatic patients, which could be sensed by tion, AIM2 causes oligomerization of the inflammasome upon AIM2(Dombrowskietal.,2011).Interestingly,thoseDNAfrag- DNAbinding.TheAIM2inflammasomerecruitsASC,anessen- ments, which might be released from skin lesions in psoriatic tialadaptormolecule,andinducesNLRP3inflammasomeforma- patients,wereinternalizedthroughbindingtotheantimicrobial tionthroughhomophilicinteractionsbetweenthepyrindomain peptide LL-37(Dombrowski etal.,2011). Previous studies have inAIM2andthatinASC(Figure2).TheimportanceoftheAIM2 shownthatthecomplexofself-DNAwithLL-37canactivateplas- inflammasome upon PAMP recognition has been confirmed by macytoid DCs to produce type I IFNs, and complex-mediated infectionexperimentsusingaim2-deficientmacrophagesinfected type I IFN production is closely related with skin lesion devel- withFrancisella tularensis,L.monocytogenes,vacciniavirus,her- opmentinpsoriasis(Nestleetal.,2005;Landeetal.,2007).AIM2 pes simplex virus-1 and mouse cytomegalovirus (Fernandes- isaninterferon-induciblegene,suggestingthatLL-37complexes Alnemrietal.,2010;Rathinametal.,2010). withself-DNAactivateplasmacytoidDCstoproducetypeIIFNs, A second ALR, interferon-inducible protein 16 (IFI16) in and that the subsequent upregulation of AIM2 leads to IL-1β humans (a homologue of p204 in mice), has been also investi- production, and finally, psoriatic skin lesions occur because of gated as an intracellular dsDNA sensor. However, while AIM2 the increased levels of type I IFN production as well as IL-1β induces IL-1β production in response to intracellular dsDNA production. binding, IFI16 is a sensor for type I IFN production upon recognition of intracellular dsDNA (Unterholzner et al., 2010). HIGHMOBILITYGROUPBOX1(HMGB1) Although IFI16 also contains a pyrin domain, the pyrin in HMGB1 has been reported to be a major DAMP molecule. IFI16 is quite distinct from that in AIM2 as it has a lower Goodwin et al. first identified HMGB1 from calf thymus chro- affinity for ASC. Consistent with these different features of matin as a non-histone DNA-binding protein (Goodwin et al., pyrin,IFI16-mediated typeIIFNproductionuponintracellular 1973).However,Wangetal.showedthatamousemacrophagecell dsDNA stimulation was not affected by ASC deficiency, sug- linereleasedHMGB1inresponsetoLPSstimulation.Inaddition, gesting that the two HIN-200 family molecules regulate both LPS-treated mice developed increased serum levels of HMGB1, IL-1βandtypeIIFNproductionupontherecognitionofintra- similartohumanpatientswithsepsis,suggestingthatHMGB1isa cellular dsDNA (Unterholzner et al., 2010). Although AIM2- DAMPmoleculeinregardtosepsissymptoms(Wangetal.,1999). mediated signaling appears to be distinct from IFI16-mediated Accumulating evidence suggests that cellular injury results in type I IFN production, recent research has revealed that IFI16 thereleaseofHMGB1leadingtoinflammation(Abrahametal., negatively regulates the AIM2-mediated activation of caspase-1 2000; Scaffidi et al., 2002). Consistent with these observations, (Veerankietal.,2011).Asincreasedinflammatorycytokinepro- numerous studies have showed a correlation between HMGB1 duction is closely related to the development of autoinflam- and autoimmune/inflammatory diseases such as atherosclero- matory disease, the regulation between AIM2-mediated innate sis, diabetes, SLE, rheumatoid arthritis and Sjögren syndrome immune signaling and IFI16 might be deregulated in patients (Taniguchi et al., 2003; Porto et al., 2006; Urbonaviciute et al., withautoimmunedisease. 2008;Devarajetal.,2009). Roberts et al. identified p202 and AIM2 as cytosolic DNA Asdescribedpreviously,higherserumlevelsofimmunocom- bindingproteinsinmice.p202isanotherALRmoleculewithout plexes of self-DNA with autoantibodies is a hallmark of SLE. apyrindomain,indicatinganinabilitytobindASCforinflam- Previous research has shown that HMGB1 is also contained in masome formation (Roberts et al., 2009). p202 appears to be a immunocomplexesandcanelicitinflammatorycytokineproduc- negativeregulatorforAIM2-mediatedsignaling,asthereduction tion, suggesting thatHMGB1may bea carrier of DNADAMPs ofp202resultsinhigherAIM2-mediatedactivationofcaspase-1 (Tian et al., 2007; Urbonaviciute et al., 2008). Furthermore, in response to intracellular DNA. However, elevated levels of HMGB1 appears to promiscuously bind numerous molecules p202 have been reported to induce SLE-like symptoms in mice such as LPS, IFN-γ, IL-1β, and CXCL12 to induce synergistic FrontiersinCellularandInfectionMicrobiology www.frontiersin.org January2013|Volume2|Article168|7 Jounaietal. Immunesensorsfornucleicacids physiological responses (Sha et al., 2008; Youn et al., 2008; dsDNA.PreviousreportsshowedthathistonesactasDAMPs,and Campanaet al.,2009). Moreover, HMGB1 can sensepathogen- that excessive intracellular dsDNA induces type I IFNs through derivednucleicacids,whichinducetypeIIFNproduction(Yanai H2B(Kobiyamaetal.,2010).Inconfirmationofthis,H1orH2 et al., 2009). Collectively, HMGB1 might be a promiscuous arereleasedfromthenucleusafterDNAdamage,andaretranslo- carrier that enhances innate immune responses against PAMPs cated to mitochondria following the induction of apoptosis. In andDAMPs. addition, H1, H2A, and H2B may act as antimicrobial proteins The receptors for HMGB1 have been investigated, but are incertainanimals,suggestingthatH2BisanintracellulardsDNA still controversial. A well-studied receptor for HMGB1 is the sensor thatrecognizes dsDNAPAMPs andDAMPs (Kawashima receptor for advanced glycation end products (RAGE). Similar et al., 2011). Histones may be related to autoimmune diseases to HMGB1, RAGE is a promiscuous receptor that can bind to as anti-histone antibodies were detected in patients with such various ligands including DNA, RNA, SAA protein, HSPs and diseases.Furtheranalysesarerequiredtoclarifytherelationship prionprotein,suggestingthatRAGEmaysenseavarietyofDAMP betweenhistonesandautoimmunedisease. molecules in an HMGB1-dependent or -independent manner (Simsetal.,2010).Experimentswithrage-deficientmicerevealed Ku70 that HMGB1-mediated DNA sensing requires RAGE for inter- Ku70 functions in DNA repair, V(D)J recombination and in nalization of DNA complexes to produce type I IFNs via TLR9 retaining the telomere. Zhang et al. showed that various DNA (Tian et al., 2007). Interestingly, RAGE could associate with species-induced the production of type III interferon, IFN-λ1, TLR9uponrecognitionoftheAtypeofCpG-HMGB1complex, andidentified Ku70 as a novel DNAsensor by pull-down assay indicating a possible function for RAGE as a bridge molecule from the nucleus compartment (Zhang et al., 2011a). While between theextracellular HMGB1-DNAcomplex andtheTLR9 other DNA sensors are important for the production of type I compartment (Tian et al., 2007). In contrast to this observa- IFNs,Ku70appearstobeimportantfortypeIIIIFNproduction tion,nucleosomescouldsenseHMGB1complexesindependently through IRF1 and IRF7. Furthermore, Ku70-mediated type III of RAGE. Instead of RAGE, TLR2 appears to be important for IFNproductionisrestrictedwhenthelengthofintracellularDNA the recognition of HMGB1-nucleosome complexes, suggesting stimuliisgreaterthan500basepairs. that the sensing machinery of the HMGB1-nucleosome com- plex mightbedistinct fromthatof theHMGB1-DNAcomplex, RNAPOLYMERASEIII as the HMGB1-nucleosome complex could not elicit produc- As described above, RIG-I senses intracellularRNAspecies, but tion of type I IFNs even though TNF-α or IL-10 were induced may also recognize intracellular dsDNA. siRNA treatment of a (Urbonaviciuteetal.,2008).Furthermore,recentresearchidenti- human hepatoma cell line, Huh7, suppressed dsDNA-mediated fiedanovelligandforRAGE,complementC3a,thatbindshuman type I IFN production. Subsequently, Chiu et al. showed that stimulatory CpG DNA to induce type I IFNs in an HMGB1- RIG-IsensesthetranscribedRNAbyproductsofDNAtemplates independent manner. This suggests that RAGE-mediated DNA that are generated by RNA polymerase III (as is the case for sensing may involve numerous ligands (Ruan et al., 2010). poly(dA·dT)·poly(dT·dA)andEBVgenomicDNA)andinduces Although there are many varieties of HMGB1- or RAGE- production of type I IFNs (Chiu et al., 2009). An inhibitor of mediated DNArecognition, both molecules arestrongly associ- RNApolymeraseIII suppressedDNA-mediatedtypeIIFNpro- atedwiththeinductionofinflammationandthedevelopmentof duction, suggesting that RNA polymerase III is a distinct DNA chronicinflammatorydisease. sensor.However, RNApolymeraseIII-mediated dsDNAsensing isrestricted tosequences ofDNAstimulicontainingless dA·dT DNA-DEPENDENTACTIVATOROFIFN-REGULATORYFACTORS(DAI) thandG·dC. DAI has been identified as a molecule that recognizes intra- cellular DNA. Previous studies have revealed that DAI senses DHX9ANDDHX36 Z-type DNA; however, it may also bind to B-type DNA and AlthoughtheDExD/Hbox RNAhelicasefamilycontainsRIG-I induce type I IFN production through associations with TBK1 andMDA5,whichfunctionasRNAsensors,recentreportshave andIRF3(Takaokaetal.,2007).Interestingly,DAI-deficientmice revealedasimilarRNAhelicasefamilyofmolecules(DExDcfam- respondednormallytocytosolicdsDNAstimulation,suggesting ily) that contain DHX9 and DHX36, which function as ssDNA thatDAImayfunctionasoneofanumberofDNAsensorsina sensors in plasmacytoid DCs (Kim et al., 2010). Interestingly, celltype-specificfashion(Ishiietal.,2008).Currently,thefunc- whileDHX36sensesCpG-A,DHX9sensesCpG-BinaMyD88- tionofDAIiscontroversial,althoughthegeneticadjuvanticityof dependent manner. This may suggest that ssDNA PAMPs or DAIhasbeenshowntoinducestrongcytotoxicTcellresponses DAMPs are recognized by either DHX9 or DHX36, but recent (Lladser et al., 2011). Although the ability of DAI to recognize researchhasshownthatDHX9collaborateswithIPS-1torecog- DNADAMPshasnotbeendeterminedyet,DAImightbealink nizedsRNAinmyeloidDCs,indicatingthepromiscuoussensing betweenthedevelopmentofautoimmunediseaseandhostDNA ofDHX9(Zhangetal.,2011b). immunecomplexes. LEUCINE-RICHREPEATFLIGHTLESS-INTERACTINGPROTEIN1(Lrrfip1) HISTONES Some sensor molecules such as TLRs or NLRs share common HistoneH2B(H2B)isacomponentofchromatin,andKobiyama molecularpatterns,suchasleucinerichrepeats(LRRs),whichare et al. identified that H2B also functions to sense intracellular importantforligandrecognitionorprotein–proteininteractions. FrontiersinCellularandInfectionMicrobiology www.frontiersin.org January2013|Volume2|Article168|8 Jounaietal. Immunesensorsfornucleicacids AnLRR-containingmolecule,Lrrfip1,hasbeenreportedtosense antigen-presentingcells aswell asantigen-specificantibody and intracellular DNA or RNA (Yang et al., 2010). Interestingly, cytotoxic T cell responses. This suggests that self-DNA DAMPs whereas other DNA sensors often regulate type I IFN-related can activate innate immune responses that induce acquired transcriptionfactorssuchasIRF3/7orcaspase-1toinducemat- immunoresponses. Recently, Marichal et al. demonstrated that uration of IL-1β, Lrrfip1 stimulates β-cateninand CBP/p300 to the adjuvanticity of alum was dependent on self-DNA released enhanceifnb1transcription,indicatinganovelpathwayinvolving from cells at the alum inoculation site (Marichal et al., 2011). β-cateninfortypeIIFNproductionuponcytosolicDNAsensing. NLRP3 appears to be a key sensor in the induction of alum- Because Wnt/β-catenin signaling is also linked to tumor devel- mediatedinnateimmunity,althoughitsfunctionisonlypartially opment,furtheranalysesmayidentifythemachineryinvolvedin dependent upon alum adjuvanticity. Intraperitoneal inocula- the regulation of type I IFN signaling by Lrrfip1 under tumor tion ofmice with aluminducedtherecruitment of neutrophils, development. and the resulting alum deposits contained high amounts of genomicDNA.BecausetreatmentwithDNaseIattenuatedalum STING(STIMULATOROFINTERFERONGENESPROTEIN) adjuvanticity, the alum-mediated release of genomic DNA may The major function of MHC class II is antigen presentation, account for its potent adjuvanticity. In addition, the alum- whilemonoclonalantibodiesagainstMHCclassIIcancausecell mediated induction of antibody production is dependent on activation or apoptotic cell death. Jin et al. identified a novel TBK1andIRF3asdemonstratedusingknockoutmice, suggest- tetraspanin family molecule, MPYS, associated with MHC-II- ingthatalum-mediatedgenomicDNAinduceshighadjuvantic- mediated cell death (Jin et al., 2008). Three research groups ity of alum via the TBK1/IRF3 pathway, while alum-mediated performing cDNA library screening to identify molecules asso- uric acid production is less related to alum adjuvanticity via ciated with activation of the typeI IFN promoter identifiedthe NLRP3 (Marichal et al., 2011). Furthermore, self-DNAs from samemolecule,STING(alsoknownasMITA,andERIS).STING alum inoculation can activate inflammatory monocytes, and isanoveladaptormoleculethatactivatesinnateimmunesignal- homodimers of IL-12p40 are more important than type I IFN ingmediated byintracellularnucleicacidstimuli (Ishikawaand productionuponalumadjuvanticity.Takentogether,thesefind- Barber,2008; Zhonget al.,2008; Sunetal.,2009). Surprisingly, ings suggest that self-DNA DAMPs are important for pathogen theBarberresearchgroupfurtherrevealedthatSTINGisessen- elimination, the development of autoimmune disease and the tialfortheinductionoftypeIIFNproductionfollowingsensing adjuvanticity of alum. Further analyses are required to elu- ofcytosolicdsDNA,usingSTING-deficientmice.Basedontheir cidate which types of cells produce self-genomic DNA after imaginganalysis,STINGappearstolocalizetotheERduringthe adjuvant inoculation, and which sensors recognize extracellular steadystate,buttranslocatestotheGolgiapparatusuponintra- genomicDNAs. cellular dsDNA stimulation to activate down-stream molecules In addition to alum adjuvant, there are many licensed adju- such as TBK1.This suggests thatSTING is an essential adaptor vants such as MF59®, AS03®, and AS04®. Both MF59® and molecule for cytosolic dsDNA-mediated type I IFN production AS03®areemulsionsofoil/watercontainingsqualene.Although inmice. bothadjuvantselicitantibodyresponsesaswellascell-mediated Cyclic-di-GMPandc-di-AMParesmallmoleculesthatfunc- immuneresponsesspecificforantigens,theirmodeofactionhas tionassecondmessengersandareimportantforcellsurvival,dif- notbeenidentified.Informationonthereceptorsforandsignal- ferentiation,colonization,andbiofilmformation.Recentresearch ing induced by these adjuvants is needed, because unfortunate has revealed that the cytosolic delivery of c-di-GMP or c-di- sideeffectscanbeexpectedmoreeasily. AMP-induced type I interferon (IFN) production from bone marrowmacrophages,suggesting thatc-di-GMP andc-di-AMP CONCLUDINGREMARKS arebacterialPAMPmolecules(McWhirteretal.,2009;Woodward Many sorts of nucleic acid species exist in the environ- et al., 2010). As type I IFN production by c-di-GMP or c-di- ment. These species affect all organisms such as the evolu- AMPrequirestheirinternalization,liveinvasivebacteriapossibly tion of organisms, the inflammatory response, and the advent produce these second messenger molecules after internalization of drug-resistant microorganisms. To prevent pathogen infec- intocells. tion, mammalian cells have equipped themselves with many Recentreports haverevealedthatSTINGisadirectsensorof sorts of sensors to recognize exogenous nucleic acid species as bacterialsecondmessengermolecules,suchasc-di-GMPorc-di- PAMPs, while those sensors are also stimulated by endogenous AMP (Burdette et al., 2011; Jin et al., 2011). This indicates the nucleic acids species as DAMPs. Dysfunction of the machiner- novelpossibilitythatcytosolicdsDNAstimulationmightproduce ies sensing both PAMPs and DAMPs is strongly associated c-di-GMP/c-di-AMPorrelatedmoleculesthatcanbesensedby with chronic inflammatory disease or autoimmunity. In addi- STINGandinducetypeIIFNproduction. tion, both PAMPs and DAMPs underlie the action of vaccines, because most modern vaccines contain adjuvants, which are ADJUVANTICITYTHROUGHDNADAMPs composed of both PAMP- and DAMP-associated molecules. Although DNA DAMPs are closely associated with the devel- Therefore, the machinery responsible for sensing nucleic acids opment of autoimmune disease, DNA DAMPs also contribute species should be further elucidated to help us understand to the activation of acquired immune responses following vac- machineryofchronicinfection,autoimmunedevelopment,iden- cination with alum adjuvant. Previous studies have shown that tifying the side effects of vaccines, and developing safe vaccine genomic DNA from dying cells induces the maturation of adjuvants. FrontiersinCellularandInfectionMicrobiology www.frontiersin.org January2013|Volume2|Article168|9 Jounaietal. Immunesensorsfornucleicacids REFERENCES Akira, S., and Shlomchik, M. J. fortheNalp3inflammasomeinthe bronchoalveolar lavage fluid: a Abraham,E.,Arcaroli,J.,Carmody,A., (2005).Toll-likereceptor9controls immunostimulatory properties of newmarkerinsarcoidosisreflecting Wang,H.,andTracey,K.J.(2000). anti-DNAautoantibodyproduction aluminium adjuvants. Nature 453, pulmonary disease. Br. Med. J. HMG-1 as a mediator of acute inmurinelupus.J.Exp.Med.202, 1122–1126. (Clin.Res.Ed.)290,1778–1781. lunginflammation.J.Immunol.165, 321–331. Fernandes-Alnemri,T.,Yu,J.W.,Datta, Hardarson, H. S., Baker, J. S., Yang, 2950–2954. Christensen,S.R.,Shupe,J.,Nickerson, P.,Wu,J.,andAlnemri,E.S.(2009). Z., Purevjav, E., Huang, C. H., Bamming, D., and Horvath, C. M. K., Kashgarian, M., Flavell, R. A., AIM2 activates the inflammasome Alexopoulou,L.,etal.(2007).Toll- (2009).Regulation of signal trans- andShlomchik,M.J.(2006).Toll- andcelldeathinresponsetocyto- likereceptor3isanessentialcom- duction by enzymatically inactive like receptor 7 and TLR9 dictate plasmicDNA.Nature458,509–513. ponentoftheinnatestressresponse antiviral RNA helicase proteins autoantibody specificity and have Fernandes-Alnemri, T., Yu, J. W., invirus-inducedcardiacinjury.Am. MDA5, RIG-I, and LGP2. J. Biol. opposing inflammatory and regu- Juliana, C., Solorzano, L., Kang, J.Physiol. HeartCirc. Physiol.292, Chem.284,9700–9712. latory roles in a murine model of S.,Wu,J.,etal.(2010).TheAIM2 H251–H258. Banka,C.L.,Yuan,T.,deBeer,M.C., lupus.Immunity25,417–428. inflammasomeiscriticalforinnate He, R., Sang, H., and Ye, R. D. Kindy, M., Curtiss, L. K., and de Colli, M. L., Moore, F., Gurzov, E. immunity to Francisella tularensis. (2003). Serum amyloid A induces Beer,F.C.(1995).SerumamyloidA N., Ortis, F., and Eizirik, D. L. Nat.Immunol.11,385–393. IL-8secretionthroughaGprotein- (SAA):influenceonHDL-mediated (2010). MDA5 and PTPN2, two Ferrari,D., Pizzirani, C., Adinolfi, E., coupled receptor, FPRL1/LXA4R. cellular cholesterol efflux. J. Lipid candidate genes for type 1 dia- Lemoli,R.M.,Curti,A.,Idzko,M., Blood101,1572–1581. Res.36,1058–1065. betes, modify pancreatic beta-cell etal.(2006).TheP2X7receptor:a Heil, F., Hemmi, H., Hochrein, H., Berg,A.H.,andScherer,P.E.(2005). responses to the viral by-product key player in IL-1 processing and Ampenberger, F., Kirschning, Adipose tissue, inflammation, and double-stranded RNA. Hum. Mol. release.J.Immunol.176,3877–3883. C., Akira, S., et al. (2004). cardiovasculardisease.Circ.Res.96, Genet.19,135–146. Fukui,R.,Saitoh,S.,Kanno,A.,Onji, Species-specific recognition of 939–949. Communi, D., Janssens, R., Suarez- M.,Shibata,T.,Ito,A.,etal.(2011). single-stranded RNA via toll-like Burckstummer, T., Baumann, C., Huerta, N., Robaye, B., and Unc93B1 restricts systemic lethal receptor 7 and 8. Science 303, Bluml,S.,Dixit,E.,Durnberger,G., Boeynaems,J.M.(2000).Advances inflammationbyorchestratingToll- 1526–1529. Jahn,H.,etal.(2009).Anorthog- in signalling by extracellular like receptor 7 and 9 trafficking. Hemmi, H., Takeuchi, O., Kawai, T., onal proteomic-genomic screen nucleotides.theroleandtransduc- Immunity35,69–81. Kaisho,T.,Sato,S.,Sanjo,H.,etal. identifies AIM2 as a cytoplasmic tionmechanismsofP2Yreceptors. Ghoreschi, K., Laurence, A., Yang, X. (2000). A Toll-like receptor recog- DNAsensorfortheinflammasome. Cell.Signal.12,351–360. P., Tato, C. M., McGeachy, M. nizes bacterial DNA. Nature 408, Nat.Immunol.10,266–272. Crow, Y. J., Hayward, B. E., Parmar, J., Konkel, J. E., et al. (2010). 740–745. Burdette,D.L.,Monroe,K.M.,Sotelo- R., Robins, P., Leitch, A., Ali, M., Generation of pathogenic T(H)17 Hiratsuka, S., Watanabe, A., Sakurai, Troha, K., Iwig, J. S., Eckert, B., etal.(2006).Mutationsinthegene cellsintheabsenceofTGF-betasig- Y., Akashi-Takamura,S., Ishibashi, Hyodo,M.,etal.(2011).STINGis encodingthe3(cid:2)−5(cid:2)DNAexonucle- nalling.Nature467,967–971. S., Miyake, K., et al. (2008). The a direct innate immune sensor of aseTREX1causeAicardi-Goutieres Glasgow,L.A.,andHabel,K.(1962). S100A8-serum amyloid A3-TLR4 cyclicdi-GMP.Nature478,515–518. syndrome at the AGS1 locus. Nat. The role of interferon in vaccinia paracrinecascadeestablishesapre- Campana,L.,Bosurgi,L.,Bianchi,M. Genet.38,917–920. virus infection of mouse embryo metastaticphase.Nat.CellBiol.10, E., Manfredi, A. A., and Rovere- Devaraj,S., Dasu, M.R., Park,S.H., tissue culture. J. Exp. Med. 115, 1349–1355. Querini,P.(2009).Requirementof andJialal, I.(2009).Increasedlev- 503–512. Hoffman, J. S., and Benditt, E. P. HMGB1 for stromal cell-derived els of ligands of Toll-like recep- Goodwin, G. H., Sanders, C., and (1982). Changes in high density factor-1/CXCL12-dependent tors 2 and 4 in type 1 diabetes. Johns,E.W.(1973).Anewgroupof lipoproteincontentfollowingendo- migration of macrophages and Diabetologia52,1665–1668. chromatin-associatedproteinswith toxinadministrationinthemouse. dendritic cells. J. Leukoc. Biol. 86, Dombrowski, Y., Peric, M., Koglin, a high content of acidic and basic Formation of serum amyloid 609–615. S., Kammerbauer, C., Goss, C., amino acids. Eur. J. Biochem. 38, protein-rich subfractions. J. Biol. Chambers, R. E., MacFarlane, D. G., Anz, D., et al. (2011). Cytosolic 14–19. Chem.257,10510–10517. Whicher, J. T., and Dieppe, P. A. DNA triggers inflammasome acti- Gorden,K.K.,Qiu,X.X.,Binsfeld,C. Hornung, V., Ablasser, A., Charrel- (1983). Serum amyloid-A protein vationinkeratinocytesinpsoriatic C., Vasilakos, J. P., and Alkan, S. Dennis, M., Bauernfeind, F., concentrationinrheumatoidarthri- lesions. Sci. Transl. Med. 3:82ra38. S. (2006). Cutting edge: activation Horvath, G., Caffrey, D. R., et al. tis and its role in monitoring dis- doi:10.1126/scitranslmed.3002001 of murine TLR8 by a combina- (2009). AIM2 recognizes cytosolic ease activity.Ann.Rheum.Dis.42, Dostert,C.,Guarda,G.,Romero,J.F., tion of imidazoquinoline immune dsDNA and forms a caspase-1- 665–667. Menu, P., Gross, O., Tardivel, A., response modifiers and polyT activatinginflammasomewithASC. Chambers, R. E., Stross, P., Barry, etal.(2009).Malarialhemozoinis oligodeoxynucleotides. J.Immunol. Nature458,514–518. R. E., and Whicher, J. T. (1987). a Nalp3 inflammasome activating 177,6584–6587. Hornung,V.,Rothenfusser,S.,Britsch, SerumamyloidAproteincompared danger signal. PLoS ONE 4:e6510. Groskreutz, D. J., Monick, M. M., S.,Krug,A.,Jahrsdorfer,B.,Giese, with C-reactive protein, alpha 1- doi:10.1371/journal.pone.0006510 Powers, L. S., Yarovinsky, T. O., T., et al. (2002). Quantitative antichymotrypsinandalpha1-acid Dostert, C., Petrilli, V., Van Bruggen, Look, D. C., and Hunninghake, expression of toll-like receptor glycoproteinasamonitorofinflam- R.,Steele,C.,Mossman,B.T.,and G. W. (2006). Respiratory syn- 1-10 mRNA in cellular subsets of matory boweldisease. Eur.J.Clin. Tschopp,J.(2008).Innateimmune cytial virus induces TLR3 pro- humanperipheralblood mononu- Invest.17,460–467. activation through Nalp3 inflam- tein and protein kinase R, leading clear cells and sensitivity to CpG Cheng,N.,He,R.,Tian,J.,Ye,P.P.,and masomesensingofasbestosandsil- to increased double-stranded RNA oligodeoxynucleotides. J.Immunol. Ye,R.D.(2008).Cuttingedge:TLR2 ica.Science320,674–677. responsiveness in airway epithelial 168,4531–4537. is a functional receptor for acute- Eckle, T., Fullbier, L., Wehrmann, cells.J.Immunol.176,1733–1740. Ishii, K. J., Coban, C., Kato, H., phaseserumamyloidA.J.Immunol. M., Khoury, J., Mittelbronn, M., Gross, O., Poeck, H., Bscheider, M., Takahashi, K., Torii, Y., Takeshita, 181,22–26. Ibla,J.,etal.(2007).Identification Dostert, C., Hannesschlager, N., F.,etal.(2006).AToll-likereceptor- Chiu,Y.H.,Macmillan,J.B.,andChen, of ectonucleotidases CD39 and Endres,S.,etal.(2009).Sykkinase independent antiviral response Z. J. (2009). RNA polymerase III CD73 in innate protection during signalling couples to the Nlrp3 inducedbydouble-strandedB-form detectscytosolicDNAandinduces acutelunginjury.J.Immunol.178, inflammasomeforanti-fungalhost DNA.Nat.Immunol.7,40–48. typeIinterferonsthroughtheRIG-I 8127–8137. defence.Nature459,433–436. Ishii, K. J., Kawagoe, T., Koyama, S., pathway.Cell138,576–591. Eisenbarth, S. C., Colegio, O. R., Hallgren, R., Eklund, A., Engstrom- Matsui, K., Kumar, H., Kawai, T., Christensen, S. R., Kashgarian, M., O’Connor,W.,Sutterwala,F.S.,and Laurent, A., and Schmekel, etal.(2008).TANK-bindingkinase- Alexopoulou, L., Flavell, R. A., Flavell, R. A. (2008). Crucial role B. (1985). Hyaluronate in 1 delineates innate and adaptive FrontiersinCellularandInfectionMicrobiology www.frontiersin.org January2013|Volume2|Article168|10

See more

The list of books you might like

Most books are stored in the elastic cloud where traffic is expensive. For this reason, we have a limit on daily download.