ebook img

MicroRNA and cellular targets profiling reveal miR-217 and miR-576-3p as proviral factors during PDF

25 Pages·2017·6.3 MB·English
by  
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 MicroRNA and cellular targets profiling reveal miR-217 and miR-576-3p as proviral factors during

RESEARCHARTICLE MicroRNA and cellular targets profiling reveal miR-217 and miR-576-3p as proviral factors during Oropouche infection VictorEmmanuelVianaGeddes1,AnibalSilvadeOliveira2,AmilcarTanuri1, EuricoArruda2,MarceloRibeiro-Alves3,RenatoSantanaAguiar1* 1 DepartamentodeGene´tica,InstitutodeBiologia,UniversidadeFederaldoRiodeJaneiro,RiodeJaneiro, RiodeJaneiro,Brazil,2 DepartamentodeBiologiaCelulareMolecular,CentrodePesquisaemVirologia, FaculdadedeMedicinadeRibeirãoPreto,UniversidadedeSãoPaulo,RibeiraoPreto,SãoPaulo,Brazil, a1111111111 3 InstitutoNacionaldeInfectologiaEvandroChagas,FIOCRUZ,RiodeJaneiro,RiodeJaneiro,Brazil a1111111111 a1111111111 *[email protected] a1111111111 a1111111111 Abstract OropoucheVirusistheetiologicalagentofanarbovirusfebrilediseasethataffectsthousands ofpeopleandiswidespreadthroughoutCentralandSouthAmericancountries.Althoughiso- OPENACCESS latedin1950’s,stillthereisscarceinformationregardingthevirusbiologyanditsprevalence Citation:GeddesVEV,deOliveiraAS,TanuriA, islikelyunderestimated.Inordertoidentifyandelucidateinteractionswithhostcellsfactors ArrudaE,Ribeiro-AlvesM,AguiarRS(2018) MicroRNAandcellulartargetsprofilingrevealmiR- andincreasetheunderstandingabouttheOropoucheVirusbiology,weperformedmicroRNA 217andmiR-576-3pasproviralfactorsduring (miRNA)andtargetgenesscreeninginhumanhepatocarcinomacelllineHuH-7.Cellular Oropoucheinfection.PLoSNeglTropDis12(5): miRNAsareshortnon-codingRNAsthatregulatesgeneexpressionpost-transcriptionally e0006508.https://doi.org/10.1371/journal. andplaykeyrolesinseveralstepsofviralinfections.ThelargescaleRT-qPCRbased pntd.0006508 screeningfound13differentiallyexpressedmiRNAsinOropoucheinfectedcells.Furthervali- Editor:PatriciaV.Aguilar,UniversityofTexas dationconfirmedthatmiR-217andmiR-576-3pwere5.5foldup-regulatedatearlystagesof MedicalBranch,UNITEDSTATES virusinfection(6hourspost-infection).Usingbioinformaticsandpathwayenrichmentanaly- Received:September22,2017 sis,wepredictedthecellulartargetsgenesformiR-217andmiR-576-3p.Differentialexpres- Accepted:May8,2018 sionanalysisofRNAfrom95selectedtargetsrevealedgenesinvolvedininnateimmunity Published:May29,2018 modulation,viralreleaseandneurologicaldisorderoutcomes.Furtheranalysisrevealedthe Copyright:©2018Geddesetal.Thisisanopen geneofdecappingprotein2(DCP2),apreviousknownrestrictionfactorforbunyaviruses accessarticledistributedunderthetermsofthe transcription,asamiR-217candidatetargetthatisprogressivelydown-regulatedduringOro- CreativeCommonsAttributionLicense,which poucheinfection.Ouranalysisalsoshowedthatactivatorsgenesinvolvedininnateimmune permitsunrestricteduse,distribution,and responsethroughIFN-βpathway,asSTING(StimulatorofInterferonGenes)andTRAF3 reproductioninanymedium,providedtheoriginal authorandsourcearecredited. (TNF-ReceptorAssociatedFactor3),weredown-regulatedastheinfectionprogress.Inhibi- tionofmiR-217ormiR-576-3prestrictsOROVreplication,decreasingviralRNA(upto8.3 DataAvailabilityStatement:Allrelevantdataare withinthepaperanditsSupportingInformation fold)andvirustiter(3fold).Finally,weshowedthatvirusescapeIFN-βmediatedimmune files. responseincreasingthelevelsofcellularmiR-576-3presultinginadecreasingofitspartners Funding:Thisworkwassupportedbyfunding STINGandTRAF3.Weconcludedstatingthatthepresentstudy,thefirstforaPeribunyaviri- fromFundac¸ãodeAmparoàPesquisadoEstado daemember,givesinsightsinitsprospectivepathwaysthatcouldhelptounderstandvirus doRiodeJaneiro-FAPERJ(www.faperj.br)under biology,interactionswithhostcellsandpathogenesis,suggestingthatthevirusescapesthe thegrantnumber:E-26/010.001562/2014,Ref.: 210.022/2014.RSAandATwererecipientofthat antiviralcellularpathwaysincreasingtheexpressionofcognatesmiRNAs. funding.VEVGwasrecipientofafellowship grantedbyCoordenac¸ãodeAperfeic¸oamentode PLOSNeglectedTropicalDiseases|https://doi.org/10.1371/journal.pntd.0006508 May29,2018 1/25 miR-217andmiR-576-3pareproviralfactorsforOropouchereplication PessoaldeN´ıvelSuperior-CAPES(www.capes. gov.br)underthegrantnumber:1545985.The Authorsummary fundershadnoroleinstudydesign,datacollection andanalysis,decisiontopublish,orpreparationof OropoucheViruscausestypicalarboviralfebrileillnessandiswidelydistributedintropi- themanuscript. calregionofAmericas,mainlyAmazonregion,associatedwithcasesofencephalitis. Competinginterests:Theauthorshavedeclared 500,000peopleareestimatedtobeinfectedwithOropoucheworldwideandsomestatesin thatnocompetinginterestsexist. BrazildetectedhighernumberofcasesamongotherarbovirusessuchasDengueandChi- kungunya.Asmuchasclimatechange,humanmigrationandvectorandhostavailability mightincreasetheriskofvirustransmission.Despiteitsestimatedhighprevalencein CentralandSouthAmericapopulations,theliteratureconcerningthemainaspectsof viralbiologyremainscarceandbegantobeinvestigatedonlyinthelasttwodecades. Nonetheless,littleisknownaboutvirus-hostcellinteractionsandpathogenesis.Virus infectionregulatescellularpathwayseitherpromotingitsreplicationorescapingfrom immuneresponsethroughmicroRNAs.KnowingwhichmicroRNAsandtargetgenesare modulatedininfectioncouldgiveusnewinsightstounderstandmultipleaspectsofinfec- tion.Here,wedepictedcandidatemiRNAs,genesandpathwaysaffectedbyOropouche Virusinfectioninhepatocytecells.Wehopethisworkserveasguidelineforprospective studiesinordertoassessthecomplexityregardingtheorthobunyavirusesinfections. Introduction OropoucheVirus(OROV)istheetiologicalagentofOropouchefever,anarthropod-borne viraldiseasecharacterizedbysymptomslikefever,headache,myalgia,arthralgia,malaise,pho- tophobia,nausea,vomiting,dizziness,skinrash,andinfewcasesencephalitisandmeningitis [1–7].ItwasfirstdescribedinTrinidadin1955[8]andthefirstBrazilianstrainwasisolated fromadeadpale-throatedthree-toedsloth(Bradypustridactylus)nearahighwayconstruction campsiteinBele´m,Para´state,northernBrazil[9].Itisestimatethatmorethan500,000people wereinfectedinatleast30outbreaksinSouthandCentralAmericabetween1961and2009[8, 10,11,12],placingOropouchefeverasoneofthemostprevalentarboviraldiseaseinsome statesofBrazil,afterDengue,ChikungunyaandYellowFever.However,theviruspathogenesis isstillobscure,andOropouchefeverisstillconsideredaneglecteddisease.Duringurbanout- breaks,thevirusismainlytransmittedbyitsmajortransmissionvector,themidgeCulicoides paraensis[3,9,13].Otherinsectspecies,likemosquitoesofthegenusAedesandCulex,might alsobepotentialvectors[9].OROVisclassifiedintheorderBunyavirales,Peribunyaviridae family,Orthobunyavirusgenus,asBunyamweraVirus,LaCrosseVirusandtherecentlydis- coveredSchmallenbergVirus[14].TheorderBunyaviralesisthelargestvirusorder,contain- ingseveralvirusesimplicatedintheetiologyofrelevanthumandiseases,suchasLaCrosse Virus(LACV)andOropoucheVirus(Orthobunyavirus),RiftValleyFeverVirus(RVFV) (Phlebovirus),Crimean-CongoFeverVirus(CCFV)(Orthonairovirus)andtherodent-borne HantaanVirus(HTNV),AndesVirus(ANDV)andSinNombreVirus(SNV)(Orthohanta- virus).OROVhasatri-segmentednegativestrandRNAgenomewithasmallsegment(S)that encodesthenucleocapsidproteinNandanon-structuralproteinNSs;amedium(M)segment thatencodestheglycoproteinsGcandGnandanothernon-structuralprotein,NSm,anda large(L)segmentthatencodestheviralRNA-dependentRNApolymerase(RdRP)[15]. DespiteitsrelevanceasahumanpathogenanditshighprevalenceinSouthAmerica,littleis knownaboutOROVreplicativecycle,pathogenesisandvirus-hostinteractions.Arecent studydemonstratedthattheOROVentryinHeLacellsisdependentonclathrin-coatedpits [16].AnotherreportshowedtherelevanceofMAVS,IRF-3andIRF-7,componentsofthe PLOSNeglectedTropicalDiseases|https://doi.org/10.1371/journal.pntd.0006508 May29,2018 2/25 miR-217andmiR-576-3pareproviralfactorsforOropouchereplication innateimmuneresponse,inrestrictingOROVinfectioninknockoutmicemodelsandnon- myeloidcells[17].Despitethat,theviruspathogenesisandthecellularpathwaysregulatedby OROVinfectionarenotknownindetail.Geneexpressionandpost-transcriptionalregulation ismediatedbyshortnon-codingRNAs(microRNAs,miRNAsormiR)thatplaysimportant rolesduringvirusreplication. MicroRNAsspanbetween19–22nucleotidesinlengthandtheirfirstdescriptionwasmade innematodes[18,19],thoughnowtheyhavebeenidentifiedinseveralphylaofplantsandani- mals[20],andeveninviralgenomes[21].Inmammals,theycanbegeneratedfromintronic andexonicregionsofprotein-codinggenesorintergenicregions[22].Theycanbefoundas singlemiRNAgenesorinclustersthatencodeslongprecursormolecules,thepri-miRNA, rangingfromhundredtothousandnucleotidesinlength[23,24].Pri-miRNAsbeginstobe editedinthenucleusbytheenzymeDroshaintopre-miRNAs,shorter70nucleotideslong moleculeswithhairpinstructures[25,26].Thosepre-miRNAsareexportedfromthenucleus intothecytoplasmbyproteinssuchasexportin5andRAN-GTP[27],andarefurtherpro- cessedbyDicerintoa22nucleotideslongdouble-strandedRNA(commonlyreferredas miRNA:miRNA(cid:3))[28,29].Thedouble-strandedRNAisloadedintoanArgonaute-driven RNAinducedsilencingcomplex(RISC),whichselectsonestrandandbindstoatargetmRNA (commonlyinthe3’-untranslatedregion,or3’-UTRregion)[30,31]bybasecomplementarity. ThemiRNAinteractionwithitstargetmRNAinducesgenesilencingbydegradation(when fullcomplementaritybetweenthemiRNAandthetargetsequenceoccurs)[32],ortransla- tionalinhibition(incaseofpartialcomplementarity)[33,34].Sincetheseedsequence(the minimalcomplementaritysitebetweenmiRNAandmRNA)isusually7–8nucleotideslong,a singlemiRNAcouldregulatesexpressionofseveralgenes,aswellasasinglegenecouldbereg- ulatedbymanymiRNAs[35,36]. MiRNAshavealreadybeendescribedinfluencingdiseaseprogression,pathogenicityand replicativecycleofseveralviruses,beingeitherinhibitoryorstimulatoryoftheinfection[37, 38].Theliver-specificmiRNA-122stimulatesHCVtranslation,stabilizingandprotectingthe 5’-UTRofviralRNAsfromdegradation,leadingtoanaccumulationofthesameinthecyto- plasm[39–42].InrestingCD4+Tlymphocytes,HIV-1viralproductionisimpairedbycellular miRNAsthatcontributetoestablishthevirallatency[43].AnothermiRNA,miR-29a,targets HIV-1RNAtoaccumulateinRNAprocessingbodies(P-bodies),inhibitingvirusinfection throughtranslationsuppression[44].Evendifferentstrainsofthesameviruscanelicitdiffer- entmiRNAregulationresponses,asdemonstratedforthehighly-pathogenicavian-derived InfluenzaAH7N7strainandthelow-pathogenicswine-derivedInfluenzaAH1N1strain[45], suggestingthatmiRNAsignatureprofilescouldraisecluesaboutpathogenicityvariation. ConcerningmiRNAregulationbybunyaviruses,astudywithpathogenicandnon-patho- genicstrainsofhantavirusesdemonstratedthevariationonmiRNAprofileamongthediffer- entspecie-specificvirusesandcelltypes[46].AnotherstudywiththeHantavirusRespiratory Syndrome(HPS)-causingagent,AndesVirus(ANDV),identifieddown-regulationofmiR- 126expression,amiRNAthatactsasregulatorofSPRED1[47].Increasedexpressionof SPRED1wassuggestedtobeoneofthemechanismsthataugmentendothelialcellspermeabil- ity,leadingtoHPS.ArecentstudywithPBMCofpatientspresentingacutehemorrhagicfever causedbytheCrimean-CongoHemorrhagicFeverVirus(CCHFV)showedthederegulation ofseveralmiRNAs,someofthemassociatedwithinnateimmunityandviralescapemecha- nisms[48].TheonlystudywithphlebovirusesdescribedtheassociationbetweenmiR-142-3p andtheendocyticvesicleproteinVAMP3,suggestingacontrolmechanismfortheintracellu- lartraffickingofUukuniemiVirus(UUKV)[49]. DuetothescarcityofinformationregardingtheregulationofbunyavirusesbymiRNAand theincreasingnecessityofbetterunderstandingofvirus-hostinteractionsofrelevantemerging PLOSNeglectedTropicalDiseases|https://doi.org/10.1371/journal.pntd.0006508 May29,2018 3/25 miR-217andmiR-576-3pareproviralfactorsforOropouchereplication pathogens,weaimedtoevaluateandidentifythecellularmiRNAprofileandtargetgenes inducedbyOROVinfectioninvitro.WedemonstratedthatmiRNAsmiR-217andmiR-576- 3p,differentiallyexpressedduringinfection,couldberegulatingcrucialpathways,likeinnate immunityresponse,mainlyinupstreamproteinsofinterferon-βinductionpathway(adaptor andkinaseproteins,aswellastranscriptionfactors),proteinshutoffandapoptosis. Methods Celllines,virusandinfection CelllinesVero(ATCC,CCL-81),U87-MG(ATCC,HBT-14)andHeLa(ATCC,CCL-2)were maintainedinDMEM(Gibco)supplementedwith10%v/vFetalBovineSerum(FBS)(Gibco) and1%v/vofpenicillin-streptomycin(10.000U/ml-10.000μg/ml)(Gibco)at37˚Cand5% CO .HuH-7cellsweremaintainedinDMEMwithoutsodiumpyruvate(Gibco)supplemented 2 with10%v/vHyCloneserum(GELifeSciences),1%v/vantibiotics,1%200mML-Glutamine (Gibco)and1%v/vnon-essentialaminoacids(Gibco)at37˚Cand5%CO .Jurkat(ATCC, 2 TIB-152)andTHP-1(ATCC,TIB-202)weremaintainedinRPMI-1640medium(Gibco)sup- plementedwith10%v/vFBS,1%v/vantibioticsand1%v/vsodiumbicarbonate(Gibco)at 37˚Cand5%CO .OROVstrainBeAn19991wasoriginallyobtainedfromtheEvandroChagas 2 InstituteandpropagatedbyserialpassagesinVerocellsbyroutinemethodsusingDMEM. TheOROVstockusedinthepresentexperimentswaspropagatedinHeLacellsandtitratedto 2x106PFU/ml.InfectionswereperformedatMOI1during1hat37˚Cand5%CO in 2 mediumwithoutFBS,underbiosafetylevel3conditionsataBSL-3laboratoryatUniversidade FederaldoRiodeJaneiro. Virustitration VirustitrationwasperformedbyplaqueassayinVerocellsplatedat3x105cells/wellin12 wellplates1daypriortoinfection.After1hincubationwiththevirus,cellswerereplenished byDMEMsupplementedwith1%v/vFBS,1%v/vantibioticsand1%v/vcarboxymethylcellu- lose(CMC)(Sigma-Aldrich),andincubatedat37˚Cand5%CO during4days.Cellswere 2 fixedwith4%formaldehydefor20minatroomtemperature,washedinPhosphateBuffered Saline(PBS)(Gibco)andstainedwith20%v/vethanol-violetcrystalsolutionfor15min. THP-1PMAtreatment Inordertoinducemonocyte-to-macrophagedifferentiation,THP-1cellswerestimulatedwith 100nMphorbol12-myristate13-acetate(PMA)(Sigma-Aldrich)instandardRPMImedium for24hor3daysfollowedby5daysincubationatRPMImediumwithoutPMA.FreshRPMI mediumwasprovidedtocellsaftertreatmentandbeforeinfections.THP-1derivedmacro- phagescellswereinfectedasdescribedabove,at24hor8dayspostPMAtreatment. Flowcytometryandimmunofluorescencestaining Cells(105cells/sample)werefixedwith4%paraformaldehydefor20minandpermeabilizedin 1%v/vTritonX-100PBSsolution.Blockingwasperformedin5%v/vDonkeySerum(Sigma- Aldrich)PBSsolutionfor1hat37˚C.OROVinfectedanduninfectedcellswereincubatedwith mousepolyclonalanti-OROVantibodyat1:300dilutioninblockingsolutionat37˚Cfor30 min.CellswerethenwashedthriceinPBSandincubatedwith2μg/mlDonkeyanti-mouse AlexaFluor488secondaryantibody(ThermoFisherScientific)at37˚Cfor30min.Afterincuba- tionwiththesecondaryantibody,cellswerewashedandresuspendedinPBS.Flowcytometry PLOSNeglectedTropicalDiseases|https://doi.org/10.1371/journal.pntd.0006508 May29,2018 4/25 miR-217andmiR-576-3pareproviralfactorsforOropouchereplication wasperformedinAccuriC6cytometer(BDBiosciences).Atleast10,000gatedeventswere countedperexperimentalreplicaatFITCchannel. Cellviabilityassay HuH-7wereplatedat2x104cells/welldensityon96-wellplateandincubatedat37˚Cand5% CO for12h.Afterthat,cellswereinfectedasdescribedabove.Cellviabilitywasevaluatedby 2 CellTiter-Blue(Promega)accordingtomanufacturer’sinstructions.Thefluorescencewasmea- suredatSpectraMaxParadigmMulti-ModeDetectionPlatform(MolecularDevices). RNAisolationandqualityassessment TotalcellularRNAformicroarrayandtargetmRNART-qPCRanalysiswasisolatedusingMir- Vana kit(ThermoFisherScientific)accordingtomanufacturer’sinstructions.RNAquantification andqualitywasassessedby2100BioanalyzerusingRNA6000Nanokit(AgilentTechnologies). OnlysampleswithaRNAIntegrityNumber(RIN)(cid:21)9.0wereusedformicroarray.Extraction ofRNAformiRNAvalidationwithspecificprimerswasperformedusingPureLinkRNAMini Kit(ThermoFisherScientific)andquantificationandintegritywereassessedinNanoVueSpec- trophotometer(GELifeSciences).AllRNAsweretreatedwithDNase(TURBODNA-freeKit, ThermoFisherScientific)beforeRT-qPCRexperimentstoavoidDNAcontamination. MiRNAscreening InordertoevaluatetheexpressionprofileofmiRNAs,anarrayusingTaqmanchemistrywas performedasfollows:12hafterinfection,sixindependentreplicasofmock-infectedorOro- poucheinfected(4x106cells/replicaatMOI1)HuH-7cellsweretrypsinized(Trypsin0.25%, Gibco)andthetotalcellularRNAwasextractedandquantifiedasdescribedabove.cDNAwas generatedusingTaqManMicroRNAReverseTranscriptionKit(ThermoFisherScientific)with 100ngofRNApersampleaccordingtomanufacturer’sinstructions.ThecDNAwaspreampli- fiedusingMegaplexPreAmpPrimers(ThermoFisherScientific)andTaqmanPreAmpMaster Mix(ThermoFisherScientific)asinstructedbymanufacturer.TheqPCRreactionwasperformed usingTaqmanOpenArrayHumanMicroRNAPanels(ThermoFisherScientific),TaqmanOpen- ArrayReal-TimeMasterMixandtheOpenArrayAccufillsystemOpenArrayreal-timerobotics (ThermoFisherScientific).Thisplatformisabletoquantify754humaninventoriedmiRNAs. MiRNAarraystatisticalanalysis Rstatisticallanguage[50]wasusedforbackgroundcorrectionanddataexploratoryanalysis(Rn intensitycumulativecurveandHighResolutionMelting—HRMgraphs)foreachRT-qPCR reaction.Forrelativeexpressionquantification,afourparameterssigmoidalcurveadjustment wasdoneusingtheqpcRfunctionsinRlanguage[51].Quantificationcycle(Cq)wasdetermined astherelativecycletosecondderivativemaximumpointofadjustedsigmoidalcurve(cpD2). Theamplificationefficiencywasdeterminedattheexponentialamplificationregion,atthe meanpointbetweenrelativecyclestothefirstderivativemaximumpointandsecondderivative maximumpointofadjustedsigmoidalcurve[expR=cpD2-(cpD1-cpD2)],andcalculatedasthe ratiobetweentheexpRcorrespondingcyclefluorescenceandthepriorcyclefluorescence.For eachmiRNA,theamplificationefficiencywasdeterminedasthemeanofefficienciescalculated forthecorrespondingmiRNA.Endogenoussmall-nucleolarRNAsRNU44,RNU48andU6 RNAwerecandidatesfornormalizationcontrolsselectedbythegeNormmethod[52].Asan alternativenormalizationmethod,thenormalizationfactorwascalculatedbythegeometric meanofallmiRNAexpressedineachsample[53].Fornormalizedexpressioncomparison PLOSNeglectedTropicalDiseases|https://doi.org/10.1371/journal.pntd.0006508 May29,2018 5/25 miR-217andmiR-576-3pareproviralfactorsforOropouchereplication betweentwosamplegroups,weperformedanon-parametricT-testwith1,000permutations [54].Forthreeormoregroupscomparisonweusedaone-waynon-parametricANOVAwith unrestrictedpermutation(n=1,000)followedbyanon-parametricpairwiseT-testmeancom- parisonwithpermutation(n=1,000)followedbyBonferronicorrection[54].Resultswere presentedasmean±S.E.M(standarderrormean).Two-tailedp-valuesinsamplegroups’com- parisonlowerorequalto0.01,0.05or0.1wereconsideredashighlysignificant,significantand suggestive,respectively.TherelationshipbetweensampleprofileswasinvestigatedbyBayesian InfiniteMixturesModelclusteranalysis[55]andrepresentedby2Dheatmapwithdendrograms (bi-cluster).Forthepurposeofdisplayintheheatmap,k-nearestneighborsmethod(k=5)was performedtopredictthemissingvaluesinuninfectedcellsformiR-217,miR-26a-2-3pand miR-92a-5p.Afterimputationofthemissingvalues,ascaled(Z-score)normalizationwasper- formed(subtractedmiRNAmeandividedbymiRNAstandarddeviation). ValidationofmiRNAthroughprimerdesignedRT-qPCR ReversetranscriptionwasperformedusingmiRNA1st-StrandcDNASynthesisKit(Agilent Technologies) andqPCRreactionsweremadewithHigh-SpecificitymiRNAQPCRCoreKit (AgilentTechnologies) andforwardspecificprimersforeachmiRNAinvestigated.HumanU6 RNAforwardprimer(AgilentTechnologies) wasusedasnormalizationcontrol.Alltheexperi- mentsweredoneinfourindependentreplicasforeachtimepointandsamplegroup.The qPCRreactionwasperformedin7500Real-TimePCRSystem(AppliedBiosystems).The cyclingparametersweresetforstandardSYBRGreenmethodaccordingtomanufacturer’s instructionsasfollow:95˚C–10minand95˚C–10sec,60˚C–15sec,72˚C–20secfor40 cycles.ThemiRNAforwardprimerssequencesaredepictedinS1Table.Statisticalanalysis wasperformedusingnon-parametricalMann-Whitneytests. Targetgenesbioinformaticsprediction WeonlyconsideraputativetargetfordifferentiallyexpressedmiRNA(miRNA:mRNAinter- action)theonespredictedinatleast3outof6publicdatabasesasfollows:TargetScan, (avail- ableathttp://www.targetscan.org/index.html)miRTarget2 (availableathttp://mirdb.org/ miRDB/),PicTar (availableathttps://pictar.mdc-berlin.de/),miRBase(availableathttp://www. mirbase.org/),TarBase (availableathttp://carolina.imis.athena-innovation.gr/diana_tools/ web/index.php?r=tarbasev8%2Findex)andmiRandaV3.3a.Interactionnetworktreewas designedusingCytoscapev3.2.1software(CytoscapeConsortium). Genesetenrichmentanalysis(GSEA) Ontologyenrichmentanalysis[56]wasperformedforthepredictedtargetsofthedifferentially expressedmiR-217andmiR-576-3p.Theontologieswereenrichedmainlytobiologicalpro- cesses,molecularfunction,cellularcomponents,andgeneinteraction/regulationpathways. Onlygenespredictedinatleast3outof6databaseswereconsideredcandidatetargets.Gene EntrezidforthepredictontologieswereusedintheGeneOntologyDatabase(GO,availableat http://www.geneontology.org/),KEGG(availableathttp://www.genome.jp/kegg/)andREAC- TOME(availableathttp://www.reactome.org/PathwayBrowser)forthispurpose.Onlygenes overrepresentedinhypergeometrictestswithp-value(cid:20)0.001wereconsidered. MicroRNAinhibitorstransfection HuH-7cellswereseeded(105cells/replica)intriplicateinto24wellsplateovernight.Negative controlinhibitor,miR-217inhibitorandmiR-576-3pinhibitor(Integrated DNATechnologies) PLOSNeglectedTropicalDiseases|https://doi.org/10.1371/journal.pntd.0006508 May29,2018 6/25 miR-217andmiR-576-3pareproviralfactorsforOropouchereplication weretransfectedatafinalconcentrationof75nMusing2μlofLipofectamine2000(Thermo FisherScientific)perreplica.GreenfluorescentshortRNAsiGLO(Dharmacon,GELifeSci- ences)wasusedtoassesstransfectionefficiencyandestablishthemiRNAinhibitorconcentra- tionforinhibitionexperiments.3hpost-transfection,cellswereinfectedwithOROVatMOI 1andRNAwereextractedformiRNAquantification(6hpost-infection)ortargetgeneand OROVRNAquantification(18hpost-infection)byRT-qPCR.OROVsegmentSRNAwas quantifiedusingprimersandprobe[57]withTaqman2xUniversalPCRMasterMix(Thermo FisherScientific)andnormalizedbyGAPDHusingPrimeTimeprimersandprobemix(Inte- gratedDNATechnologies). ValidationoftargetmRNAthroughRT-qPCR Cellswereseeded(106cells/sample)andinfectedwithOROVatMOI1.TheRNAwas extractedat12hpostinfectionandreversetranscriptionwasperformedusingHigh-Capacity cDNAReverseTranscriptionKit(ThermoFisherScientific)and1μgofRNA.Quantitative PCRwasdoneinsixreplicaperconditionusing50ng/wellofcDNAonCustomTaqman ArrayFastplates(96well)(ThermoFisherScientific)usingspecificprimersandprobesand TaqmanFastUniversalPCRMasterMix(ThermoFisherScientific)accordingtomanufactur- er’sinstructionson7500FastReal-TimePCRSystem(ThermoFisherScientific).Statistical analysiswasdoneasdescribedformicroarrayusingendogenous18S,GAPDH,HPRT1and GUSBasnormalizationgenes.FortargetkineticsSYBRGreenPCRMasterMix(AppliedBio- systems)andpre-designedPrimeTimeprimers(Integrated DNATechnologies) wereused accordingtomanufacturer’sinstruction(forprimerssequencesseeS1Table). Results OROVinfectshepatocytesandbloodcelllinesinvitro InordertoexpandtheknowledgeontherangeofOROV-permissivecells,bloodandhepato- cytecelllineswereusedtoevaluateinvitroinfection(Fig1A).TCD4+lymphocytes(Jurkat), monocytes(THP-1)andhepatocytes(HuH-7)celllineageswereinfectedwithOROVatMOI1 and,at12hpostinfection,infectivitywasassessedbyimmunofluorescenceusingspecificanti- bodiesagainstOROVproteinsandvirus-positivecellswerecountedbyflowcytometry.Atindi- catedtimepoints,21%ofJurkatcellswereinfected,whileTHP-1presentednosusceptibilityto theOROVinfection.THP-1cellscanbeinducedtodifferentiateintomacrophagebyPMA treatment,becomingpermissivetosomeviralinfections,asdescribedelsewhere[58–61].In ordertoassessifTHP-1cellsdifferentiatedintomacrophageswerepermissivetoOROVinfec- tion,THP-1cellsweretreatedwithPMAfor24horfor3days,followedbyincubationin mediumwithoutPMAfor5moredays.DifferentiationofTHP-1intomacrophage-likepheno- typewasaccompaniedbymicroscopyandattachment.At12hpostinfection,31%and50%of THP-1treatedwithPMAfor24hor8days,respectively,wereinfectedwithOROV,suggesting anincreasingpermissivenesstoOROVinfectionasthecellsshiftfrommonocytetomacro- phage-likephenotypes(Fig1A).AtthesameMOI,HuH-7cellsshowedtobemorepermissive toOROVinfection,presenting90%infectedcellsat12hpost-infection(Fig1A).Basedonthis resultwithHuH-7cells,andconsideringpreviousdemonstrationsthattheliverisanimportant replicationsiteduringexperimentalOROVinfectioninhamster[62,63]andmouse[64],we chosethehepatocytecelllineHuH-7asourinvitromodelforfurtherexperiments. Toassessthemostsuitableconditionstoensurethatmostcellswouldbeinfectedatindi- catedtimepoints,HuH-7cellswereinfectedwithdifferentMOIsandtheinfectivitywasmea- suredbyflowcytometry(Fig1B).Wereached30%ofinfectivityatMOI0.1withaplateauof 90%inhigherconcentrationsofvirus(MOIs1,5and10),withnofurtherincreaseof PLOSNeglectedTropicalDiseases|https://doi.org/10.1371/journal.pntd.0006508 May29,2018 7/25 miR-217andmiR-576-3pareproviralfactorsforOropouchereplication Fig1.Oropoucheinfectioninbloodandhepatocytecelllines.Jurkat,THP-1andHuH-7cellswereinfectedwithOROVatMOI1.At12hpostinfection,infectivity wasassessedbyflowcytometryusingpolyclonalanti-OROVantibody.(A)InfectioninTlymphocyte(Jurkat),monocyte(THP-1)andhepatocyte(HuH-7)celllines (x-axis)wasperformedasdescribedinmaterialsandmethodssection.Errorbarsrepresentsstandarddeviation(SD)offourindependentexperiments.THP-1 NA=THP-1notactivated;THP-1(24)=activatedfor24hinPMA;THP-1(8d)=activatedfor3daysinPMAfollowedby5dayswithoutPMA.(B)OROVinfection inHuh-7cellsatdifferentMOIs(x-axis).Infectivitywasassessedbyflowcytometry.ErrorbarsrepresentsSD.(C)RelativecellviabilityofHuH-7cellsinfectedwith OROV(MOI1)atdifferenttimepoints.Cellviabilitywasassessedbyfluorimetricassay.Absolutevaluesofuninfectedcellsweresetas1(lefty-axis).Blackcolumns representuninfectedcellsandgraycolumnsrepresentOROVinfectedcells(MOI1).ErrorbarsrepresentsSDforfivereplicatesoftwoindependentexperiments.Gray linerepresentsmeanvirustiterinsupernatantmeasuredbyplaqueassayatindicatedtimepoints(righty-axis).(cid:3)(cid:3)=p(cid:20)0.01. https://doi.org/10.1371/journal.pntd.0006508.g001 infectivitylevels(Fig1B).Toassurethatcellswerestillviableforfurtherexperiments,we assessedthecytopathiceffectat6,12,18and24hpostinfectionwithMOIof1usingCell Titer-Blue(Fig1C).WedidnotdetectcelldeathassociatedtotheOROVinfectionatleast18 hpostinfection.However,only44%ofcellswereviableat24hpostinfection.Wealsoquanti- fiedthevirustitergeneratedinthosecellsbyplaqueassayandat6hpost-infection,thetiterin thesupernatantwas6x103PFU/ml(Fig1C,grayline).Astheinfectionprogressed,apeakof 8.6x105PFU/mlcouldbedetectedat18hpostinfection,reachingaplateauwithnofurther increaseinviraltiterat24hpostinfection(Fig1C,grayline).Basedonthesedata,wepro- ceededusingHuH-7cellsinfectedwithMOI1insubsequentexperimentstoevaluatethe virus-hostinteractions. CellularmiRNAsignatureinHuH-7cellsinfectedwithOROV MiRNAscanbeinformativeofcellulartargetsmodulatedbyvirusinfection.Inordertoiden- tifycandidatecellularpathwaysdifferentiallyexpressedinOROVinfectedcells,weperformed anexploratoryscreeningof754humanmiRNAsthroughprobe-basedRT-qPCR.MiRNAs expressionwasevaluatedinfouruninfected(control)andfiveOROVinfectedbiologicalrepli- casat12hpostinfection.WefoundthirteenmiRNAsdifferentiallyexpresseduponOROV PLOSNeglectedTropicalDiseases|https://doi.org/10.1371/journal.pntd.0006508 May29,2018 8/25 miR-217andmiR-576-3pareproviralfactorsforOropouchereplication infectioninHuH-7cellswithstatisticalsignificance:twelveup-regulatedafterinfectionand onlyonedown-regulated(miR-450b-5p)(Fig2andTable1).Thereproducibilityofeffectsin miRNAswasindicatedbysmallvariancenotedamongbiologicalreplicas,asdepictedinthe heatmaphierarchicaldendrogram(Fig2). ThedifferentialexpressionofthemiRNAsinOROVinfectedcellswasclassifiedintothree groups:up-regulated,down-regulatedandinfection-dependentmiRNAs(selectivelyexpressed miRNAs).MiRNAsmiR-324-3p(1.73x),miR-1227(1.95x),miR-362-3p(1.85x),miR-99b-3p (2.21x),miR-19b-1-5p(4.11x),miR-628-3p(2.77x),miR-26a-1-3p(42.47x),miR-576-3p (2.49x)andmiR-27a-5p(108x)wereup-regulated,inOROV-infectedcellsrelativetounin- fectedcells.MiR-450b-5pwasdown-regulated4.65timesininfectedcellscomparedtounin- fectedcells.TheinductionofmiR-26a-2-3pandmiR-217wereinconsistentandobservedonly inthreeoutoffiveinfectedreplicas.FromthethirteenselectedmiRNAsfromthescreening, onlymiR-576-3pandmiR-26a-1-3psustainedsignificance(p(cid:20)0.05,p(cid:20)0.01,respectively) afterBonferronicorrectionaccordingtothemethodusedinthisstudy.Nonetheless,some miRNAspresentedborderlinelimitsofsignificance(p=0.0595),namely,miR-1227,miR-19b- 1-5pandmiR-450b-5p(Table1). InordertovalidatethemiRNAsthatweresignificantlyderegulatedinthearray(miR-26a- 1-3pandmiR-576-3p)andtoverifytheexpressionofthemiRNAsonlydetectedininfected cellsintheexpressionprofilearray(miR-217,miR-26a-2-3pandmiR-92a-1-5p),wedesigned specificprimersforeachmiRNAandcheckeditsexpressionbyRT-qPCR(Fig3).Ourvalida- tionexperimentsshowedthesametendencyofthemiRNAspanelwithanincreasingexpres- sionofbothmiR-217(Fig3A)andmiR-576-3p(Fig3B)duringinfection,reachingapeakof expressionat6hpost-infection(about5.5foldincreaseforbothmiRNAs).Thekineticsof expressionofmiR-217suggestsanearlyinductionduringinfectioncomparedtomiR-576-3p, sincemiR-217wasup-regulated2.26timesasearlyas3hpost-infectionwhilemiR-576-3pwas onlyup-regulated1.44atthesamepoint.However,atlaterstagesofinfection,miR-217expres- sionwasalreadyclosertouninfectedexpressionlevels(up-regulatedonly1.7at12hpost- infection),whereasmiR-576-3pwasstillup-regulated2.83timesininfectedcells,indicatinga slightlydifferentkineticsforthosemiRNAs. Toconfirmtherobustnessofouranalysis,wefurthervalidatedtheexpressionofthree otherlessstablestarmiRNAs:thehighlysignificantmiRNAmiR-26a-1-3pandtwomiRNAs detectedonlyuponinfection,miR-26a-2-3pandmiR-92a-1-5p(Fig3C).ThosethreemiRNAs wereup-regulated5.3,4.5and6.3fold,respectively,at12hpost-infectionincomparisonwith uninfectedcells(p(cid:20)0.01).Altogether,theseresultswithspecificprimerstoeachmiRNAcor- roboratewithourlarge-scalepaneldata,identifyingmiRNAsthataremodulatedduring OROVinfectionshowingthesametendencywithdifferentapproaches.StarmiRNAnomen- claturecorrespondstopassengerstrandslessfavorabletoprocessingbyRISCwithlowerlikeli- hoodtoregulategeneexpression[65,66].AsmostofthosemiRNAsarepreviouslyannotated asstarmiRNAs(asexampleofmiR-26a-1-3ppreviouslyannotatedasmiR-26a-1(cid:3))andsome predictiondatabasealgorithmsuseprovedinteractionascriteriaforprediction,weonly selectedmiR-217andmiR-576-3p,bothmaturestrandmiRNAs,forfurthertargetprediction analysis(onedetectedonlyininfectedcellsandtheotheroneup-regulatedsignificantlyupon infectioninthearray,respectively,andbothvalidated). CellulartargetgenesregulatedbymiR-217andmiR-576-3p ToinvestigatepossiblepathwaysregulatedbymiR-217andmiR-576-3pduringOROVinfec- tion,weperformedtargetpredictionusingTargetScan,miRTarget2,PicTar,miRBase,TarBase andmiRandadatabases.Targetgenespredictedbyatleast3outof6ofthosedatabaseswere PLOSNeglectedTropicalDiseases|https://doi.org/10.1371/journal.pntd.0006508 May29,2018 9/25 miR-217andmiR-576-3pareproviralfactorsforOropouchereplication Fig2.HeatmapofdifferentiallyexpressedmiRNAsinOROVinfectedHuH-7cells.Depictionofthe13miRNAs differentiallyexpressedinOROVinfectedcellsrelativetouninfectedcells.LinesrepresentindividualmiRNAsand columnsrepresentindependentreplicates(fourmockinfectedandfiveOROVinfected).Colorscalerepresents normalizedexpressionlevelsofmiRNAsinthetwoconditionsinlog scale;reddenotesup-regulationandgreen 2 denotesdown-regulation,respectively.Dendrogramrepresentingthe1Dclusterizationofsamplesandthe2Dmap correspondingtothelevelsofstandardizedgeneexpressionprofiles(z-score).Reddottedlinesinthedendrogram indicateweakunions,discouragedbytheBayesianclusteringanalysis.Valuesrepresentedinthedendrogrambranches correspondtolog-oddsoftheunionofcorrespondingbranches.Forthepurposeofdisplay,missingvaluesof uninfectedcellsformiR-217,miR-26a-2-3pandmiR-92a-5pwerepredictedbyk-nearestneighborsmethodand imputedafternormalizationasdescribedinMethodssection. https://doi.org/10.1371/journal.pntd.0006508.g002 PLOSNeglectedTropicalDiseases|https://doi.org/10.1371/journal.pntd.0006508 May29,2018 10/25

Description:
sis, we predicted the cellular targets genes for miR-217 and miR-576-3p. Differential from Fundação de Amparo à Pesquisa do Estado do Rio de
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.