• Nenhum resultado encontrado

Nasal vaccination with attenuated Salmonella expressing VapA: TLR2 activation is not essential for protection against R. equi infection

N/A
N/A
Protected

Academic year: 2021

Share "Nasal vaccination with attenuated Salmonella expressing VapA: TLR2 activation is not essential for protection against R. equi infection"

Copied!
8
0
0

Texto

(1)

ContentslistsavailableatScienceDirect

Vaccine

jo u rn a l h o m e pa ge : w w w . e l s e v i e r . c o m / l o c a t e / v a c c i n e

Nasal

vaccination

with

attenuated

Salmonella

expressing

VapA:

TLR2

activation

is

not

essential

for

protection

against

R.

equi

infection

Silvia

Almeida

Cardoso

a,b

,

Aline

Ferreira

Oliveira

c

,

Luciana

Pereira

Ruas

a,b

,

Marcel

Montels

Trevisani

a

,

Leandro

Licursi

De

Oliveira

d

,

Ebert

Seixas

Hanna

a,b

,

Maria

Cristina

Roque-Barreira

a

,

Sandro

Gomes

Soares

a,b,∗

aDepartamentodeBiologiaCelulareMoleculareBioagentesPatogênicos,FaculdadedeMedicinadeRibeirãoPreto,USP,14049-900,Brazil bInventBiotecnologia,14040-900,Brazil

cDepartamentodeMicrobiologiaeImunologia,InstitutodeBiologia,UniversidadeEstadualdeCampinas(UNICAMP),Campinas13083-862,SP,Brazil dDepartamentodeBiologiaGeral,UniversidadeFederaldeVic¸osa(UFV),Vic¸osa36570-000,MG,Brazil

a

r

t

i

c

l

e

i

n

f

o

Articlehistory: Received6May2013

Receivedinrevisedform15July2013 Accepted25July2013

Available online 7 August 2013 Keywords: Rhodococcusequi Vectoredvaccine VapA AttenuatedSalmonella Toll-likereceptors

a

b

s

t

r

a

c

t

VirulentstrainsofRhodococcusequihavealargeplasmidof80–90kb,whichencodesseveral virulence-associated proteins (Vap), including VapA, alipoprotein highly associated withdisease. We have previouslydemonstratedthatoralimmunisationwithattenuatedSalmonellaentericaTyphimuriumstrain expressingtheantigenVapA(STMVapA+)inducesspecificandlong-termhumoralandcellular immu-nityagainstR.equi.ItwasshownthatVapAactivatesToll-likereceptor2(TLR2)onmacrophagesby establishinganinteractionthatultimatelyfavoursimmunityagainstR.equiinfection.Thepurposeof thisstudywastoevaluatetheimmuneresponsetriggeredbynasalimmunisationwithSTMVapA+and todeterminewhetherTLR2supportsthevaccineeffect.Wedevelopedanoptimisedprotocolforasingle nasalimmunisationthatconferredprotectionagainstR.equiinfectioninmice,whichwasmanifested byefficientR.equiclearanceinchallengedanimals.NasalvaccinationwithSTMVapA+hasalsoinduced protectioninTlr2−/−miceandmicewithnon-functionalTLR4.Moreover,spleencellsofvaccinatedmice augmentedT-betexpression,aswellastheproductionofIL-12,IFN-␥,nitricoxideandhydrogen perox-ide.Notably,thepopulationofCD4+Tcellswithmemoryphenotypesignificantlyincreasedinthespleens

ofvaccinatedmicechallenged1or5monthsafterimmunisation.Intheseanimals,thespleenbacterial burdenwasalsoreduced.WhensimilarexperimentalprocedureswereperformedinTLR2knockout mice,anincreaseinCD4+Tcellswithmemoryphenotypewasnotobserved.Consequently,weconclude

thatnasalvaccinationwithattenuatedSalmonellaexpressingtheR.equivirulencefactorVapAconfers long-lastingprotectionagainstexperimentalrhodoccocosisandthatTLR2engagementwasnotcrucial toinducethisprotectionbutmayberequiredforalong-termimmuneresponse.

© 2013 Elsevier Ltd. All rights reserved.

1. Introduction

Rhodococcusequiisasoilorganismdistributedworldwidethat infectsfoalsaged1–6monthsandcausesseveregranulomatous pneumonia[1].AsR.equisurvivesandreproducesinsidehostcells, pneumoniacausedbythispathogenmustbetreatedwithlipophilic drugsoveraprolongedperiodoftime[2].Currently,novaccines areavailable,althoughseveralimmunisationstrategieshavebeen testedtopreventrhodococcosis[3].

∗ Correspondingauthorat:InventBiotecnologia,IncubadoraSupera,USP,Av. Bandeirantes,3900,14040-900RibeirãoPreto,SP,Brazil.Tel.:+551681370617.

E-mailaddresses:soares@inventbiotech.com.br,soares@fmrp.usp.br

(S.G.Soares).

As R. equi harbours an 80–90-kb plasmid, proteins (Vaps) encodedbythisplasmidareconsideredgoodcandidatesfor vacci-nalantigens.VapAisthemostinvestigatedimmunogenencoded by this pathogenicity island and hasbeen reported toaccount for R. equi virulence[4,5] and elicit specific immune responses

[6,7]. In this context, our group has previously demonstrated that oral immunisation with live attenuated Salmonella enter-icaTyphimuriumexpressingVapA conferslong-termprotection againstR.equi.Protectionisduetotheinductionofstrongmucosal, humoralandcell-mediatedTh1-immunity,whichisappropriately regulated,asverifiedbytheprofileofcytokinesexpressedinthe organs of vaccinated miceafter bacterialchallenge [8,9].These promising resultshavemotivated ustoadvancestudies onthe vaccinalconceptionofVapA.

StudiesontheinnateimmuneresponsetoR.equihaveshown that infected macrophages with viable bacteria undergo NFkB

0264-410X/$–seefrontmatter © 2013 Elsevier Ltd. All rights reserved.

(2)

S.A.Cardosoetal./Vaccine31 (2013) 4528–4535 4529

translocationandproduceavarietyofpro-inflammatory media-tors,includingTNF,IL-12,andnitricoxide(NO).Thisresponsewas attributedtoa pathwaythatis stronglydependentonthe acti-vationofToll-likereceptor2(TLR2)butnotofTLR4.Aspurified VapAinducesresponsesthataresimilartothosetriggeredbythe wholebacteria,TLR2activationwasattributedtoitsinteraction withVapA.Theseobservationspromptedustoinvestigatewhether TLR2accountsforprotectionconferredbyvaccinationwithVapA.

Inthepresentstudy,westandardisedanewrouteand regi-menofimmunisationwiththeVapAantigeninthecontextofa Salmonella-vectoredvaccineandinvestigatedtherole thatTLR2 playsinthevaccineeffect.Wefoundthatnasalvaccinationofmice withasingledoseofVapA-expressingSalmonellaconfers protec-tionagainstR.equiinfectionviaamechanismthatdoesnotdepend onTLR2activation.Moreover,thevaccinationprocedure stimu-latesmemoryTCD4+cellsandpromotesspecificimmuneresponse

recall.

2. Materialsandmethods 2.1. Experimentalanimals

Groupsof femalemice, 6–8weeksof age,includingBALB/c, C3H/HeJ,C3H/HePAS,Tlr2−/− andwildtype (WT)C57BL/6mice (fivemicepergroup),werehousedunderspecificpathogen-free conditionsintheAnimalResearchFacilitiesoftheMedicalSchool ofRibeirãoPreto-USP.Thestudieswereconductedasrequiredby theEthicsCommitteeonAnimalResearchoftheUniversityofSão Paulo.

2.2. Bacterialstrains

TheattenuatedS.entericaTyphimurium␹3987strains,carrying eithertheVapAantigen(STMVapA+)ortheemptyvector(control VapA−),weregrownandpreparedfortheimmunisationof ani-malsasdescribedbyOliveiraetal.[9].ThevirulentstrainofR.equi (ATCC33701)wasgrownandthetitreofthechallengeinoculum wascalculatedasdescribedbyOliveiraetal.[8].

2.3. Immunisationandchallengeprotocol

Groupsofanimalswereimmunisedwithasingledoseortwo dosesofattenuatedSalmonellaharbouringtheVapAantigenvia intranasalroute(1×109 CFU,5␮L/animal).Asacontrol,groups

wereinoculatedwithattenuatedSalmonellacarryinganempty vec-toraccordingtothesameschedule.

ThechallengeswereconductedusinganATCCvirulent,R.equi strain3370130daysafterthefirstimmunisation. IntheBalb/c recallexperiments,theanimalswerechallenged5monthsafter immunisation.Incontrast,intheC57Bl/6recallexperiments,WT andTlr2−/−animalswerechallenged120daysafterimmunisation. Organswerecollected5daysafterchallengeforR.equiclearance evaluationandotheranalysesasdescribedbelow.

2.4. Estimationofbacterialclearanceintheorgansofinfected mice

QuantificationofviableR.equirecoveredfromthespleenand liverofchallengedmicewasperformedonday5post-infectionas reportedelsewhere[8].Briefly,30daysafterthefirst immunisa-tionprocedurewithSTMVapA+,micewereinfectedwith4×106

CFUs(ColonyForming Units)ofvirulent R.equi viaintravenous route.Fivedaysafterthechallenge,spleenandliverwerecollected andasepticallyhomogenised.TodeterminethenumberofCFUs,

aliquotsof100␮LofthehomogenateswereseriallydilutedinPBS, platedontoBHIagarinduplicate,andincubatedat37◦Cfor36h.

2.5. Detectionofcytokinesinspleenhomogenates

Spleen homogenatesampleswereanalysed viaELISAfor IL-12p40,INF-␥,andTNF-␣usingcommerciallyavailablekits(OptEIA set;Pharmingen,SanDiego,CA,USA).Theassayswereperformed accordingtothemanufacturer’srecommendations.

2.6. RealtimequantitativePCRanalysis

TotalcellularRNAwasextractedfromspleencellsamplesusing TRIzolReagent(InvitrogenLifeTechnologies,Carlsbad,CA,USA) accordingtothemanufacturer’srecommendations.RealtimePCR wasperformedaspreviouslydescribedbyOliveiraetal.[9]using primersforGATA-3,T-bet,TLR2,and␤-actin.

2.7. Flowcytometryanalysis

Stainingforflowcytometrywasperformedonspleencells iso-latedfromimmunisedand controlmice30days post-infection. Afterharvestingthespleencells,1×106 cellswerewashedwith

ice-coldPBSandincubatedfor30minat4◦Cwith0.5␮gof anti-CD16/CD32mAb(Fcblock,clone2.4G2,BDPharmingen,SanDiego, CA).Next,thecellswereincubatedforover30minuteswith5␮gof phycoerythrin-conjugatedanti-CD62L,fluorescein isothiocyanate-conjugatedanti-CD44,anti-TLR2,andPECy5-conjugatedanti-CD3 (BDPharmingen,SanDiego,CA).Washingstepswereperformed withPBS0.5%BSAandthecellswerethensubmittedfor analy-sisonaGuavaflowcytometer(Millipore,Billerica,MA,USA)using GuavaCytoSoftversion4.2.1(Millipore,Billerica,MA,USA). 2.8. Statisticalanalysis

Statistical analysis (one-way analysis of variance) was per-formedin conjunctionwiththeTukeymultiplecomparisontest usingGraphPadPrismversion5.00(GraphPadSoftware,SanDiego, CA,USA).TheresultsarepresentedasthemeanandSE.Differences betweengroupswereconsideredsignificantatthelevelofp<0.05. 3. Results

3.1. NasalimmunisationwithSTMVapA+protectsmiceagainst R.equiinfection

Ourpreviousstudieshavedemonstratedthatoral immunisa-tionofBalb/cmicewithliveattenuatedS.entericaTyphimurium (STM)expressingVapAantigen(STMVapA+)isaneffectivestrategy toinduceprotectionagainstR.equiinfection[8,9].Inthepresent study,weperformednasalimmunisationofBalb/cmicewitha sin-gledoseortwodosesoftheSTMVapA+.Bothproceduresresulted inasignificantreductionintheR.equiburdeninthespleenandliver ofmicechallenged30daysafterthefirstimmunisationcompared withtheorgansofanimalsinjectedwithcontrolVapA−(negative control)(Fig.1AandB).Becausebothimmunisationregimens pro-motedcomparableR.equi clearance,theprotocolusingasingle nasalinoculationofSTMVapA+waselectedforsubsequent exper-iments.

Tooptimisethequantityof thevaccineinoculum,groups of Balb/cmicewereimmunisedwithdifferentdosesofSTMVapA+ (102–109)and challengedwithR.equi 30days after

immunisa-tion.AsignificantreductioninsplenicR.equiburdenwasobserved whentheinoculumoftheVapA+vaccinestrainwasatleast107

(3)

Fig.1. NasalimmunisationwithSTMVapA+reducesR.equiburdenininfectedmice.Balb/cmicewereimmunisedwithasingledoseortwodosesofattenuatedSalmonella entericaTyphimuriumcarryingVapAantigen(STMVapA+).Allanimalswereeuthanisedonday5afterinfection,andR.equiburdenwasevaluatedinthespleen(A)and liver(B).PanelCshowstherecoveringofR.equifromthespleenoftheanimalsthatwereimmunisedwithasingledoseofvariousvaccineinoculum.AttenuatedSalmonella carryingemptyvectorwasusedasanegativecontrol(Control).Datarepresentthemean±SE,n=5animals.#p<0.05.

3.2. NasalimmunisationwithSTMVapA+inducesTh1immunity Toevaluatewhethertheprotectionconferredwiththenovel vaccinationprocedurewasassociatedwiththedevelopmentofTh1 immunityaspreviouslyverifiedfororalvaccination,weassessed thelevelofsomemediatorsinthespleensofmiceimmunisedwith theSTMVapA+strainandchallengedtheanimalswithR.equi.IL-12 (Fig.2A),nitricoxide(Fig.2B)andhydrogenperoxidelevels(Fig.2C) weresignificantlyhigherinthespleentissueofVapA+-immunised mice,whilethelevelsofTNF-␣remainedlower(Fig.2D).

Tobettercharacterisethe immune responseinduced bythe vaccinationprocedure, wedetermined themRNAlevelsforthe transcriptionfactorsGATA-3andT-bet30daysafter immunisa-tion.Comparedwiththenegativecontrols,i.e.,miceinjectedwith controlVapA−orPBS,miceimmunisedwithSTMVapA+displayed higherlevelsofT-betmRNA(Fig.2E)andlowerlevelsofGATA-3 mRNA(Fig.2F).Similaranalyseswereperformed5daysafter chal-lenging,andnodifferenceinthelevelsofT-betmRNAwasobserved amongallgroups(Fig.2E).GATA-3mRNAlevelsremainedlowerin vaccinated/challengedanimals(Fig.2F).

3.3. NasalimmunisationwithSTMVapA+inducesmemoryTcells andimmuneresponserecall

ToevaluatetheinductionofTcellswithmemoryphenotype vianasalvaccinationwithSTMVapA+,wehaveassessedthe fre-quencyofcellsco-expressingCD3,CD62LandCD44(high)markers, whichcharacteriseTcellswithmemoryphenotype,30daysafter immunisation.Thispopulation,accordingtotheflowcytometric analysis,waslargerinthespleensofvaccinatedmicecompared withthecontrolgroups(Fig.3A).Wealsoinvestigatedwhetherthe

expansionofthepopulationofcellswasassociatedwithimmune response recall. Accordingly, Balb/c mice were immunised and challenged5monthslater.Next,spleenandliverwerecollected foranalysis5daysafterchallenge.Ourresultsdemonstratea sig-nificantdecreaseinR.equiburdeninthegroupimmunisedwith STMVapA+(Fig.3BandC).

3.4. TLR-2expressionisupregulatedafterimmunisationand downregulatedafterchallenge

It is wellestablishedthat VapA canactivate Toll-like recep-tor2;thisinteractionisconsideredamajormechanisminvolved intriggeringspecificimmuneresponsestoR.equi.Therefore,the expressionlevelsofTLR2inthespleen cellsofmicevaccinated withSTMVapA+wereevaluatedviarealtimePCR(Fig.4A)and flowcytometry(Fig.4B),whichwascompared withthe expres-sionlevelsinthecellsofmiceinjectedwiththecontrolsVapA− orPBS.Thirtydaysafterimmunisation,micespleencellsdisplayed anincreaseinTLR2expressionlevels,asdemonstratedbyelevated mRNA(Fig.4A)andproteinlevels(Fig.4B).Fivedaysafterchallenge withR.equi,murinespleencellsdisplayedreducedTLR2expression levelssimilartothatdetectedinthenegativecontrolgroup. 3.5. ProtectionagainstR.equiconferredbySTMVapA+isnot dependentonTLR2

WefurtherinvestigatedtherolethatTLR2playsintheVapA+ vaccine-inducedprotectionagainstR.equiinfection.Accordingly, groupsofTLR2knockoutmice(C57Bl/6Tlr2−/−)orwildtypemice (WT)weretreatedwiththestandardvaccinationand challenge protocols. Asexpected, among all WT vaccinated mice, R. equi

(4)

S.A.Cardosoetal./Vaccine31 (2013) 4528–4535 4531

Fig.2. NasalimmunisationwithSTMVapA+inducesTh1-immunity.Balb/cmicewereimmunisedwithasingledoseofSTMVapA+andthelevelsofIL-12(A),nitricoxide (B),hydrogenperoxide(C)andTNF-␣(D)inthespleenwereevaluated.TherelativemRNAexpressionlevelsofT-bet(E)andGATA-3(F)werealsoanalysed.Attenuated Salmonellacarryingemptyvectorwasusedasnegativecontrol.Datarepresentthemean±SE,n=5animals.#p<0.05.

burdeninthespleen(Fig.5A)andliver(Fig.5B)waslessthanthat detectedintheorgansofnegativecontrolmiceinjectedwiththe controlsVapA−orPBS.Nonetheless,similarresultswereobtained withtheTlr2−/−group,therebyindicatingthatTLR2isnotessential fortheprotectionconferredbyourVapA+methodofvaccination

usingattenuatedSalmonella.Thisindicationwasreinforcedbythe factthatsimilarlevelsofIL-12(Fig.5C),IFN-␥(Fig.5D)andTNF␣ (Fig.5E)weredetectedinthespleentissueoftheTlr2−/−andWT animals.Additionally,theseresultsshowthatSTMVapA+ vaccina-tionofC57BL/6andBalb/cmiceisabletoconferprotectionagainst R.equiinfectionandinducesTh1immuneresponseasdenotedby increasedproductionofIL-12andIFN-␥.

Similarimmunisationandchallengeswereperformedinmice withnon-functionalTLR4(C3H/HeJ).Intheseanimals,R.equi clear-ance was comparable to that observed in the spleen (Fig. 5F) andliver(Fig.5G)ofvaccinatedmiceexpressingfunctionalTLR4 (C3H/HePAS).Thisresultindicatesthattheprotectionconferredby vaccinationwiththeSTMVapA+strainisalsoindependentofTLR4.

3.6. HighfrequencyofmemoryphenotypeCD4Tcellsis maintainedintheabsenceofTLR2

TounderstandthefunctionofTLR2inthenasal immunisation-mediated response, we investigated whethertheemergence of memoryTcellsinducedbyvaccinationcouldbeaffectedbythe absenceofTLR2.ThefrequencyofCD4+orCD8+Tcells

express-ingCD44high/CD62LwasmeasuredinthespleenofWTandTlr2−/−

mice30daysafterimmunisation.AsshowninFig.6A,vaccination ledtoanincreasedcellpopulationwithaCD4+CD62L+CD44+high

phenotype.Notably,thisincreasewassimilarforWTandTlr2−/− mice.Incontrast,thepopulationofCD8+CD62L+CD44+highcellswas

elevatedonlyinthespleensofvaccinatedWTmice,thereby indi-catingthatvaccinationhadnoeffectonthefrequencyofthesecells inTlr2−/−animals(Fig.6B).

Finally,weassessedthefrequencyofTcellswithmemory phe-notypeinchallengedanimals(120dayspost-immunisation).Inthis particular case, the CD4+CD62L+CD44+high populationremained

(5)

Fig.3. NasalimmunisationwithSTMVapA+increasesthesplenicpopulationofTcellswithmemoryphenotypeandtherecallofimmuneresponse.VaccinatedBalb/cmice withSTMVapA+werechallengedwithR.equi30daysafterimmunisation,andthefrequencyofCD3+/CD62L+/CD44highcells(A)wasanalysed.AsecondgroupofBalb/cmice

waschallenged5monthsafterimmunisationandevaluatedintermsofspleen(B)andliver(C)R.equiburden.AttenuatedSalmonellacarryingemptyvectorwasusedasa negativecontrol.Datarepresentthemean±SE,n=5animals.#p<0.05.

elevatedin WT,but notinTlr2−/− mice(Fig.6C).Regardingthe CD8+CD62L+CD44+highcells,nonoticeabledifferencewasdetected

amongallgroupsofanimals(Fig.6D).

4. Discussion

Variousstrategieshavebeenproposedforthedevelopmentofa safeandeffectivevaccineagainstrhodococcosis.Forinstance,the passiveimmunisationwithhyperimmuneplasma[10],vaccination procedureswithinactivatedR.equistrains[11],VapADNAvaccine

[12]and,morerecently,attenuatedR.equivaccine[13]havebeen reported.However,noneoftheseapproacheshaveprovidedan effi-cientmethodtocontrolthedisease.Amongthedifferentstrategies ofvaccinationforintracellularpathogens,a promisingapproach maybetheuseofliveoralvaccines.Inthiscontext,ourgrouphas

demonstratedthatoralimmunisationwithanattenuatedS.enterica TyphimuriumstrainexpressingtheVapAproteininduces protec-tionagainstexperimentalrhodococcosisinmice[8].Morerecently, wehavedemonstratedthattheoralimmunisationprotocolinduces a strongandspecifichumoral andcell-mediated Th1-immunity againsttheheterologousantigen,withanappropriateregulatory responseandalong-termprotectionagainstR.equiinfection[9].In thepresentwork,wereportthatnasaldeliveryisaneffectiveroute forimmunisation,whichconfersprotectionwithasingledoseof thevaccine.Wehavealsoverifiedthatnasalvaccinationinduces aTh1immuneresponse,whichisasprotectiveandspecificasthe oralimmunisation[9].

Nasalroutehasbeenproposedasanalternativeroutefor vacci-nationduetoeaseoftheprocessofimmunisationandthepresence of nasopharynx-associatedlymphoidtissue (NALT).Aswiththe Peyer’s patches,NALTaccomplishestherequirementstoinduce

Fig.4.NasalvaccinationwithSTMVapA+enhancesTLR2+expressionlevelsinspleencells,whichisrevertedbyR.equichallenge.TherelativeexpressionlevelsofTLR2 mRNAinthespleencellsofvaccinatedBalb/cmiceeitherunchallengedorchallengedorwithR.equiisshowninpanelA.PanelBshowstherelativefrequencyofspleen cellspositiveforTLR2stainingasdetectedviaflowcytometry.AttenuatedSalmonellaharbouringemptyvectorwasusedasanegativecontrol.In(A),cDNAcontentwas normalisedonthebasisofpredetermined␤-actinlevels.Datarepresentthemean±SEM(n=5animals).#p<0.01.In(B),datarepresentthemean±SE.#p<0.05.

(6)

S.A.Cardosoetal./Vaccine31 (2013) 4528–4535 4533

Fig.5.ProtectioninducedbynasalvaccinationwithSTMVapA+isindependentofTLR2activation.WTandTlr2−/−C57BL/6micewerechallengedwithR.equi30daysafter

vaccinationwithSTMVapA+.Fivedaysafterchallenge,R.equiburdeninthespleen(A)andliver(B)wasevaluated.Thespleentissuewasalsoanalysedforthelevelsof IL12p40(C),IFN-␥(D),andTNF-␣(E).SimilarexperimentalprocedureswereperformedwithC3H/HePASandC3H/HeJmice;R.equiburdenwasassessedinthespleen(F) andliver(G)samples.AttenuatedSalmonellacarryingemptyvectorwasusedasanegativecontrol.Thebarsrepresentthemean±SEM(n=5animals).#p<0.05.

andregulateaprotectiveimmuneresponse,whichinvolvesMcells, APCs,TcellsandBcells[14].Nonetheless,NALTischaracterisedas aTh0environment, whichcanberevertedtoeitherTh1orTh2 proneambiance[15].Althoughtheoraladministrationisa nat-uralroutefor Salmonella-basedvaccines,theintranasaldelivery

allowsfor theimmunogentoreachthesame organs,including thelungsandPeyer’spatchesand,asaconsequence,caninduce serologicandproliferativeresponses[16].Currently,Kimetal.[17]

havedemonstratedthatattenuatedSalmonellaTyphimuriumisan efficientcarrierofheterologousantigens,whichisabletoinvolve

(7)

Fig.6. NasalimmunisationwithSTMVapA+inducesincreasedfrequencyandrecallofCD4+TcellswithmemoryphenotypeviaaTLR2-dependentmechanism.Immunised

WTandTlr2−/−C57BL/6micewerechallengedwithR.equi30days(panelsAandB)or120days(panelsCandD)aftervaccination.ThefrequenciesofCD4+/CD62L+/CD44high

(AandC)andCD8+/CD62L+/CD44highcells(BandD)wereassessedviaflowcytometryinspleensamples.AttenuatedSalmonellacarryingemptyvectorwasusedasanegative

control.Thebarsrepresentthemean±SEM(n=5animals).#p<0.05.

Mcellsinnasopharynxandtracheobronchiallymphoidtissueand inducelocalandsystemic-specificimmuneresponses.Such find-ingshavemotivatedseveralgroupstodevelopSalmonella-vectored vaccinesforadministeredvianasalrouteinmice[18,19]andmares

[20].

Darrah and co-workers [21] have demonstrated that the lipoproteinVapAactivatesmacrophageTLR2,areceptorthatisa majormediatoroftheinnateresponsetoR.equi.Therefore,along withtheknowledgethatTLR2activationfavoursthe differentia-tionofTh1cells,it islikely thatthis receptorcouldplaya role intheprotectionconferredbySalmonella-expressingVapAused herein.Thisideawasreinforcedwiththefactthatvaccination ele-vatedTLR2expressionlevelsonmurinemacrophages.However, thehighTLR2expressionlevelswerenotmaintainedwhenthe immunisedmicewerechallenged withR. equi.Withregardsto thisfindings,theymightberelatedto(i)asupposedTLR2 inter-nalisation,whichaccompaniestheinternalisationofR.equior(ii) triggeringasignalviaTLR2activation,therebyinducinganegative feedbackmechanismabletocontrolinflammationafterchallenge. ThelatterassumptionissupportedbytheobservationthatTNF-␣ levelsalsodecreasedafterR.equichallenge.

ActivationofTLRsdirectlymodulatesTcellbiology[22–24].In thiscontext,alterationofTLR2expressionlevelsbythesecellscould affectvaccineefficiencyandthefrequencyofTcellpopulations. Surprisingly,theresultsofprotectionwiththeSTMVapA+vaccine weresomewhatsimilarforboththeWTandtheTlr2−/−animals, asshownbyasimilarreductioninR.equiburdeninthespleenand liver.Furthermore,thepopulationofCD4+ Tcellswithmemory

phenotypewassimilarlyexpandedinTlr2−/−andWTvaccinated mice.ThefrequencyofCD8+Tcellswithmemoryphenotype;

how-ever,wasonlygreaterinWTmiceaftervaccination.Theseresults are supportedby thestudy of Cottalorda and co-workers [25], whichdemonstratedthatTLR2playsacriticalroleinthe gener-ationofmemoryCD8+Tcells.OurresultsindicatethatCD4+Tcells

playamajorroleinR.equiimmunity.Thisisinagreementwith thefactthatactivatedCD4+TcellsexpressTLR3andTLR9butnot

TLR2andTLR4[24].Followingvaccination,activationofCD4+T

cellsoccursindependentoftheirdirectinteractionwithVapAor Salmonellacomponents.

OurresultsdonotexcludethepossibilitythatTLR2might par-ticipateintheprotectiveresponseagainstR.equiinducedbySTM VapA+.Acloserexaminationofourresultsrevealsthatvaccinated WTmicedisplayedareducedbacterialburdenofalmost3logs, whilethevaccinatedTlr2−/− micedisplayedareducedburdenof approximately1.5logs.Thus,theVapAantigen,inthecontextof theSalmonellacell,isabletoinduceaprotectiveimmuneresponse viaapathwaythatisindependentofTLR2activation. Neverthe-less,TLR2seemstoactsynergisticallytomountastrongprotective responseagainstR.equi.

TheexactmechanismbywhichtheVapA+antigenispresented bytheattenuatedSalmonellaremainsunclear.S.entericaserovars survive inside the hostcells. Although attenuated strains were shown tobeunable tocausedisease,theyretain theability to colonisethehosttissueandtriggercertainsignalsinthecontextof thehostcells.Likewise,R.equiisanintracellularpathogenand nat-uralinfectionshouldbemimickedviavaccinationwithSTMVapA+, whichhastheadvantageofprovidingbetterconditionsfor presen-tationoftheVapAantigen.Suchpresentationseemstobecrucial, as VapA ishighly associated withR. equi virulenceand thus is extensivelydescribedasagoodantigenforvaccination.Currently, manystrategieshavebeenproposedbasedontheimmunity devel-opedagainstVapA,butonlyfewofstrategieswereconsideredto havepotentialforvaccineapplication.Theresultspresentedherein suggestthatthe␹3987attenuatedstrainofSalmonellaisan effi-cientcarrierfororalornasalimmunisationwithVapA.Thevaccine stimulatesantigen-specificCD4+Tcellswithouttheparticipation

ofTLR2and leadstoa profileof immuneresponsethat confers resistancetotheR.equiinfection.Mostimportantly,theefficient

(8)

S.A.Cardosoetal./Vaccine31 (2013) 4528–4535 4535

clearanceofR.equifromtheorgansofvaccinatedmiceandthe long-termrecallofimmunityindicatethatthestrategyusedhereinmay beapplicableforvaccinationagainstrhodococcosis.

Acknowledgments

WewouldliketothankSandraO.Thomazand Patricia Ven-druscolofortechnicalsupport.Thisstudywasinpartsupported bygrantsfromFAPESP(05/50460-1)andCNPq(154963/2006-2). S.A.C.wasaPhDfellowofCAPES,andMCRBisseniorfellowofCNPq. Contributors:S.A.C.andS.G.S.designedexperiments,performed assays,interpreteddataandwrotethemanuscript.A.F.O.designed experimentsandperformedassayswithTLR2KOmice.M.M.T. per-formedassayswithTLR4-deficientanimals.L.P.R.performedPCR andflowcytometryanalysis.L.L.O.,M.C.R.B.andE.S.H.interpreted dataandcontributedtothemanuscript.Competinginterests:The technologydescribedinthispaperissubjecttopendingpatents. Thedata andmaterials described hereinadheretothe Vaccine journalpoliciesforacademic,non-commercialresearch.

References

[1]Cornish N,Washington JA. Rhodococcus equiinfections: clinical features and laboratory diagnosis. Current Clinical Topics in Infectious Diseases 1999;19:198–215.

[2]PrescottJF.Rhodococcusequi:ananimalandhumanpathogen.Clinical Micro-biologyReviews1991;4:20–34.

[3]GiguèreS,CohenND,ChaffinMK,HinesSA,HondalusMK,PrescottJF,etal. Rhodococcusequi:clinicalmanifestations,virulence,andimmunity.Journalof VeterinaryInternalMedicine2011;25(6):1221–30.

[4]Takai S,KoikeK,Ohbushi S,IzumiC, Tsubaki S.Identificationof 15-to 17-kilodaltonantigensassociatedwithvirulentRhodococcusequi.Journalof ClinicalMicrobiology1991;29:439–43.

[5]JainS,BloomBR,HondalusMK.DeletionofvapAencodingvirulence asso-ciatedproteinAattenuatestheintracellularactinomyceteRhodococcusequi. MolecularMicrobiology2003;50:115–28.

[6]Hooper-McGrevyKE,GiguereS,WilkieBN,PrescottJF.Evaluationofequine immunoglobulinspecificRhodococcusequivirulence-associatedproteinsAand CforuseinprotectingfoalsagainstRhodococcusequi-inducedpneumonia. AmericanJournalofVeterinaryResearch2001;62:1307–13.

[7]Taouji S,Nomura I, Giguere S,Tomomitsu S,Kakuda T,Ganne V, etal. ImmunogenicityofsyntheticpeptidesrepresentingBcellepitopesofVapA ofRhodococcusequi.Vaccine2004;22:1114–22.

[8]OliveiraAF,FerrazLC,BrocchiM,Roque-BarreiraMC.Oraladministration ofaliveattenuatedSalmonellavaccinestrainexpressingtheVapAprotein inducesprotectionagainstinfectionbyRhodococcusequi.Microbesand Infec-tion2007;9:382–90.

[9]OliveiraAF,RuasLP,CardosoAS,SoaresSG,Roque-BarreiraMC.Vaccinationof micewithSalmonellaexpressingVapA:mucosalandsystemicTh1responses provideprotectionagainstRhodococcusequiinfection.PLoSONE2010;5:1–12.

[10]PrescottJF,NicholsonVM,PattersonMC,ZandonaMeleiroMC,Caterinode AraujoA,YagerJA,etal.UseofRhodococcusequivirulence-associated pro-teinforimmunizationoffoalsagainstR.equipneumonia.AmericanJournalof VeterinaryResearch1997;58:356–9.

[11]VargaJ,FodorL,RusvalM,SoosI,MakraiL.PreventionofRhodococcusequi pneumoniaoffoalsusingtwodifferentinactivatedvaccines.Veterinary Micro-biology1997;56:205–12.

[12]HaghighiHR,PrescottJF.AssessmentinmiceofVapA-DNAvaccinationagainst Rhodococcusequiinfection.VeterinaryImmunologyandImmunopathology 2005;104:215–25.

[13]vanderGeizeR,GrommenAWF,HesselsGI,JacobsAAC,DijkhuizenL.The steroid catabolicpathway ofthe intracellularpathogen Rhodococcusequi is importantforpathogenesisandatargetforvaccinedevelopment.PLoS Pathogens2011;7(8):e1002181.

[14]KiyonoH,FukuyamaS.NALT-versusPeyer’s-patch-mediatedmucosal immu-nity.NatureReviewsImmunology2004;4:699–710.

[15]HiroiT,IwataniK,IijimaH,KodamaS,YanagitaM,KiyonoH.Nasalimmune system:distinctiveTh0andTh1/Th2typeenvironmentsinmurine nasal-associatedlymphoidtissuesandnasalpassage,respectively.EuropeanJournal ofImmunology1998;28(10):3346–53.

[16]PasettiMF,PickettTE,LevineMM,SzteinMB.Acomparisonofimmunogenicity andinvivodistributionofSalmonellaentéricaserovarTyphiandTyphimurium livevectorvaccinesdeliveredbymucosalroutesinthemurinemodel.Vaccine 2000;18(28):3208–13.

[17]KimDY,SatoA,FukuyamaS,SagaraH,NagatakeT,KongIG,etal.Theairway antigensamplingsystem:respiratoryMcellsasanalternativegatewayfor inhaledantigens.JournalofImmunology2011;186(7):4253–62.

[18]RocheJK,RojoAL,CostaLB,SmeltzR,ManqueP,WoehlbierU,etal.Intranasal vaccinationinmicewithanattenuatedSalmonellaentericaSerovar908htrA expressingCp15ofCryptosporidium:impactofmalnutritionwithpreservation ofcytokinesecretion.Vaccine2013;31(6):912–8.

[19]Scavone P,UmpiérrezA,MaskellDJ,ZuninoP. Nasalimmunizationwith attenuatedSalmonellaTyphimuriumexpressinganMrpA-TetCfusionprotein significantlyreducesProteusmirabiliscolonizationinthemouseurinarytract. JournalofMedicalMicrobiology2011;60(Pt.7):899–904.

[20]CauseyRC,ArtiushinSC,CrowleyIF,WeberJA,HomolaAD,KelleyA,etal. Immunisation ofthe equine uterus againstStreptococcus equisubspecies zooepidemicususinganintranasalattenuatedSalmonellavector.Veterinary Journal2010;184(2):156–61.

[21]Darrah PA, Monaco MCG, Jain S, Hondalus MK, Golenbock DT, Mosser DM.InnateimmuneresponsestoRhodococcusequi.JournalofImmunology 2004;173:1914–24.

[22]CottalordaA,VerscheldeC,MarcaisA,TomkowiakM,MusetteP,Uematsu S,etal.TLR2engagementonCD8Tcellslowersthethresholdforoptimal antigen-inducedTcellactivation.EuropeanJournalofImmunology2006;36: 1684–93.

[23]Komai-Koma M, Jones L, Ogg GS, Xu D, Liew FY. TLR2 is expressed on activated T cells as a costimulatory receptor. Proceedings of the NationalAcademy ofSciencesoftheUnitedStatesofAmerica2004;101: 3029–34.

[24]GelmanAE, ZhangJ, ChoiY,Turka LA.Toll-likereceptorligandsdirectly promote activatedCD4+Tcellsurvival.JournalofImmunology2004;172:

6065–73.

[25]CottalordaA,MercierBC,Mbitikon-KoboFM,ArpinC,TeohDY,McMichael A,etal.TLR2engagementonmemoryCD8+Tcellsimprovestheir cytokine-mediated proliferation and IFN-gamma secretion in the absence of Ag. EuropeanJournalofImmunology2009;39:2673–81.

Referências

Documentos relacionados

measurements of oxygen consumption, feed intake and/or plasma cortisol levels under different conditions: fasting fish, fed fish and fish subjected to a 3 min emersi

After 6 weeks, vaccinated mice and sham-treated controls were inoculated with the non-lethal murine malaria parasite Plasmo- dium yoelii subspecies yoelii 17XNL ( P. yoelii )

cpx R and lon genes, could also protect chickens against fowl typhoid, and only slight anorexia and depression were tem- porarily observed by the authors, while in the unvaccinated

A panel latent class model was used to determine girls’ preferences for vaccination based on five characteristics: degree of protection against cervical cancer; duration of

Here, parental cell line (AGS Par) or cells transfected with the empty vector (MDA-435 Mock) were used as control in parallel with the respective WT E-Cadherin expressing cell

Level of protection of chickens against highly pathogenic H5 avian influenza virus with Newcastle disease virus based live attenuated vector vaccine depends on homology of

In the present study, it was demonstrated that the 265gE - mutant virus is attenuated for calves, induces protection upon challenge and reactivation with a large infectious dose

Passively transferable protection against Schistosoma japonicum induced in the mouse by multiple vaccination with attenuated larvae: the development of immunity, antibody