187
Universidade de Lisboa Faculdade de Medicina Instituto de Medicina Molecular INTERACTIONS OF PLASMODIUM BLOOD AND LIVER STAGES WITHIN A SINGLE HOST Sílvia Vilar Portugal A dissertation for the degree of Doctor of Philosophy in Biomedical Sciences Specialization in Biopathological Sciences Supervised by Maria Manuel Mota, M.Sc, Ph.D Principal Investigator of Unidade de Malária in Instituto de Medicina Molecular Auxiliary Professor at Faculdade de Medicina da Universidade de Lisboa 2010

INTERACTIONS OF PLASMODIUM BLOOD AND LIVER …repositorio.ul.pt/bitstream/10451/2226/1/ulsd058613_td_Silvia... · e ao Pedro obrigada por tratarem do meu computador. À Cati ... um

Embed Size (px)

Citation preview

  • UniversidadedeLisboa

    FaculdadedeMedicinaInstitutodeMedicinaMolecular

    INTERACTIONSOFPLASMODIUMBLOODANDLIVERSTAGESWITHINASINGLEHOST

    SlviaVilarPortugal

    AdissertationforthedegreeofDoctorofPhilosophyinBiomedicalSciences

    SpecializationinBiopathologicalSciences

    SupervisedbyMariaManuelMota,M.Sc,Ph.D

    PrincipalInvestigatorofUnidadedeMalriainInstitutodeMedicinaMolecular

    AuxiliaryProfessoratFaculdadedeMedicinadaUniversidadedeLisboa

    2010

  • II

    A impresso desta dissertao foi aprovada pela ComissoCoordenadora do Conselho Cientifico da Faculdade de Medicina daUniversidadedeLisboaemreuniode13deAbrilde2010.As opinies expressas nesta publicao so da responsabilidade doseuautor.

  • III

  • IV

    TheresearchdescribedinthisthesiswasperformedattheInstitutode Medicina Molecular, Lisboa, Portugal, and was financiallysupported by Fundao para a Cincia e Tecnologia, Portugal(SFRH/BD/31523/2006).O trabalho de investigao descrito nesta tese foi realizado noInstituto de Medicina Molecular, Lisboa, Portugal, e foi financiadopela Fundao para a Cincia e Tecnologia, Portugal(SFRH/BD/31523/2006).

  • V

    Preface

    This dissertation assembles data obtained duringmyPh.D researchproject, developed at Faculdade de Medicina da Universidade deLisboa, Instituto deMedicinaMolecular, Unidade deMalaria, underthe supervision of Prof.MariaM.Mota, fromOctober 2006 toApril2010.Thisthesisisstructuresin5chapters,precededbyasummarybothinPortugueseandEnglish,outlining theaims, resultsandoutcomesofthisproject.Thefirstchapterprovidesaninsightonpreviousmalariaknowledge,andtheaimsofthiswork.The second chapter contains the description of the methods andmaterialsemployedtocarryoutthepresentwork.Thethirdchaptercontainstheoriginaldataregardingthisproject.Chapter four encloses an overall discussion and conclusions of thestudiesperformed,togetherwithasectionwherefutureperspectivesoftheworkdevelopedaredescribed.In the Appendix is included a table with the information obtainedwithamicroarrayanalysisperformedduringthecourseofthiswork.

    Thedatadescribedinthisdissertationistheresultofmyownwork.Thisworkhasneverbeenpreviouslysubmittedforanydegreeatthisoranyotheruniversity

  • VI

  • acknowledgements

    VII

    Acknowledgements

    The oneswho got to knowme duringmytime in Lisbon, know that I like a lot towork,alwaysasmuchasIcan,andhaveitdone as soon as possible. Butmost of thetimes, in order to be able to doalmost asmuchaswhatIplannedfor,Icountedwithmany important people without whom itwouldhavebeen impossible toachieveallthis. ThankyouJeremieforthephoto.

    Maria,poracharquevaliaapena termeno laboratriodesdeodiaemque me propus vir. Pela responsabilidade que me foi gradualmenteatribuindoequemepermitiucrescernolaboratrioathoje.Epelapressoboa, que sempre me fez sentir, para estar altura do que precisava otrabalho.

    Obrigada pelo projecto que deu origem a esta tese, e obrigada por tantasvezes teresacreditadoquea soluoestava jalinaprximaexperincia, eobrigada ainda pormuitas emuitas vezes teresminimizado o facto de quenotnhamosencontradosoluonenhuma...

    Mar, porque me ensinou a mexer na bancada, me deixou participaractivamente no seu trabalho, e no final me delegou responsabilidadesefectivasnafinalizaodeprojectosseus.Eporterfeitoistotudocomumcarinhosemprepresenteesempremuitovisvel.

    AnaGomes,Mosqeao Johny,por tantasvezesseremcorreiodecaixascomgelosecoeamostras,oupormelevaremetrazeremaolabshoraquetenhoquelestar,poraceitaremosmeushorrioseaindameouviremfalardasexperinciasduranteo jantarsempreatrasado.EaAnaGomes fez istovezessemconta!!

    Sem vocs seria efectivamente muito mais difcil ou incrivelmente maisaborrecido.

    To Unidade de Malria, to everyone i met in the several UMAs I crossedsince the summer of 2005. All of you, Im sure, at a certain momentcontributedtotheworkpresentedhere,andtothefunIhadproducingit.

  • ackowledgements

    VIII

    In particular I thank Cline for the contribution with the analysis of themicroarray results and for helping me interpret them with so muchpatience.

    Depois emparticular querodizermuito obrigada Vanessapelo trabalhoquederamasminhasamostrasemMuniqueepelaimensadisponibilidadeparadiscutirtrabalhoouajudarcomumcasacodeEltonJohnouumgorrode Nikita; Carina pelas discusses cientficas ou as conversas da vidaquandoficmosatrabalharatsmil;aoMiguelpelascrticaseficienteseporexemplo,poratravessarmeiacidadecomigodemota,parareporantesdas10hotremosquepartidaUBD;S,PamplonaeMargaridaVigriopor ajudarem nas minhas dvidas de imunologia; S por me ensinar aesplectomizarratinhos,Fernandapormepermitirpedidosdemosquitosforadetempoeporencurtarasminhasausnciasdacasadosanimaiscomumainjecoaquioualienquantoviajo;eElianapelaprontidocomquemeaceitounasuacasaquemepermitiuexperinciasrelmpagoemParis.

    To Mario Recker for the great help in producing the model that allowedfitting the results produced in the lab during this thesis, with fieldobservationinmalariaendemicsettings.

    ToChrisNewbold,HalDrakesmithandAndyArmitagefortheinputonthehepcidinexperiments,andforthehelppreparingthefinalarticle.

    Lgia pela contribuio com os hepatcitos primrios, e por tornaragradveisdiasaproduzirmuitomenosclulasdoqueasquegostaramos.

    AoCludioMarinhoporgostardefalarcomigosobreoprojectoquelevouaestatese,epelastantasperguntasquefezsempre.

    AoJooFerreiraeaoBrunoSilvaSantosqueenquantoComitdestatesemequestionaram e me incentivaram a perseguir os objectivos. E porperguntaremsemprecomoqueest?!omistrio.

    AtodoostaffdascasasdosanimaisdoIMMedoIGC,eAlina,Doloreseao Manel em particular por me deixarem abusar, atender os pedidos deratinhos em cima da hora, usar a cmara de fluxo sem marcao, poracreditaremqueeraimportanteeupodertrabalhartudooquequisesse.

  • acknowledgements

    IX

    RosaMaria pela prontido com que produziu os vrios anticorpos queuseiaolongodestetrabalho.

    To all of you that read parts of this thesis_ Miguel, Pat, Teresa, Bruno,Margarida,Vanessa,Pamplona,andAndy

    Thankyouforyourtime,yourcomments,corrections,andsuggestions.IhopeIdiditright!

    AoRuben,aoDanieleaoPedroobrigadaportrataremdomeucomputador.

    CatieaoseuamigoManelquemeajudaramaidentificaronomedoqueescolhiparaacapadestatese.EaoCsarqueaceitouimprimir.

    primaPaulaquenoinciomedeuasuacasapormaisdeumano.

    DepoisAnapornoseimportardepartilharcasacomumaquasesempreausente,porsepreocuparquedurma,quecoma,quedescanse.

    Aos meus pais por participarem activamente agora como antes, desde otempoemqueomeupaivendiaoscalendriosdosescuteirosquedeveriavendereu,atatemposmaisrecentesemqueaminhameforraasgaiolasdosmosquitoscomredeenastroquenosservematodosno lab.EQuelquetantassegundasfeiramelevouaocomboioahorasindecentesparaquepudesse comear a trabalhar a horas decentes. E Di porme perdoar ostantosratinhossacrificados.

    Obrigadapormedeixaremcontinuarasercuidadaporvocs.

    EaoRicardopelompetoquemedoseuamor,emtudoemaisnotrabalho.Obrigadapormeouviresjdemasiadotardeeaindasobretrabalho,porvirescomigoao lab emnoites oumadrugadas emquedevamos snamorar, porleres as coisas que escrevo ainda que tarde, e por perguntares e se fosseassim...?porajudaresnonomedateseeporarranjaresosartigosacabadosdesairdoprelooudaarcadavelha,aosquaisnotenhoacesso.

  • X

  • resumo

    XI

    Resumo A infeco pelo agente causador da malria, o parasita Plasmodium,

    encontrase ainda hoje disseminada pelas populaes de 108 pases no

    mundo.Amalriaproduzsintomasquevariamentrefebres ligeirasatao

    coma,anemiasevera, sndromerespiratriaagudaoumalriacerebral.S

    em 2008 esta infeco foi responsvel por mais de 800000 mortes, das

    quais a maioria se ficou a dever ao parasita Plasmodium falciparum.

    Adicionalmenteamalriaaindaresponsvelporumareduode1.3%no

    crescimentoeconmicodospasesdemaiorendemicidade.

    O parasita Plasmodium, pertence ao filo Apicomplexa e partilha com o

    Homem a presena na Terra desde o prprio aparecimento da espcie

    humana,tendoaadaptaoparasitahospedeirovindoaevoluiraolongodo

    tempo, procurando um balano entre a transmisso do parasita e a

    sobrevivnciadohospedeiro.

    comapicadadeumafmeademosquitoAnophelesqueoparasitachega

    ao hospedeiro mamfero. Depois de uma passagem pela pele, os

    esporozotosmigramvia corrente sanguneaatao fgadoondedepoisde

    atravessar vrios hepatcitos, invademumltimo coma formaode um

    vacolo parasitrio. No hepatcito dse um processo de crescimento e

    replicao que no homem demora vrios dias at formao de vrios

    milhares de merozotos que sero libertados de novo na corrente

    sangunea. Uma vez no sangue osmerozotos entram numa nova fase de

    reproduo assexuada, com sucessivos ciclos de invaso de eritrcitos,

    replicao e libertao para a corrente sangunea e nova invaso de

    eritrcitos. nesta fase da infeco que todos os sintomas associados

    malriasurgem.

  • resumo

    XII

    Ocasionalmente, o ciclo de reproduo assexuada dentro dos eritrcitos

    para formao de novos merozotos substitudo pela formao de

    gametcitos femininos oumasculinos que podero ser aspirados durante

    umanovapicadademosquito.nohemocliodestevectorquesedafase

    sexualdoparasitadamalriacomaformaodeumzigotoesubsequente

    oocinetoquemigraparaalminabasalondesetransformanumoocisto.A

    partir da esquizogonia dos oocistos surgem novos esporozotos que uma

    vezchegadossglndulassalivaresdomosquitosestoprontosparaquese

    inicieumanovainfeco.

    Em zonas de alta transmisso de Plasmodium provvel a ocorrncia de

    sobreposiodas faseshepticaesanguneanumshospedeiro,bastando

    para isso que uma picada infecciosa ocorra num indivduo que alberga j

    uma infeco circulante proveniente de uma picada anterior. Apesar de

    potencialmente importantes, as possveis interaces entre as diferentes

    fasesdedesenvolvimentodoparasitaeohospedeiroqueosacolhenunca

    foramobjectodeestudo.

    O objectivo desta tese prendese precisamente com o estudo das

    interacesquesurgirodestasituaoaquedamosonomedereinfeco.

    Quo efectivos sero os esporozotos na infeco do fgado de indivduos

    comPlasmodiumnosangue,quandocomparadoscoma infecodo fgado

    deindivduossemqualquerparasitademalriapresente?

    Fazendo uso de modelos animais previamente estabelecidos e diferentes

    clonesdeparasitas, quenospermitiramdistinguir as infecesheptica e

    sangunea nos animais reinfectados, verificmos uma reduo fortssima

    na infeco no fgado de animais reinfectados. A capacidade dos

    esporozotosparainfectarhepatcitosderatinhoscominfecosanguneaa

    decorrer apareceu altamente limitada quando comparada com a mesma

    capacidadeparainfectarhepatcitosderatinhosnaive.

  • resumo

    XIII

    O estudodetalhadodeste fenmenopermitiunos relacionar a reduoda

    infeco heptica com a diminuio tanto do nmero como do

    desenvolvimento das formas exoeritrocticas (EEF no original) no fgado.

    Estareduoverificouseindependentedonveldaparasitmiadainfeco

    primria,desdequeestaseencontrasseacimadeumvalorquesemostrou

    ser baixo e rapidamente atingido. Mais ainda, verificouse que a

    administraodeumtratamentoantimalricoaosratinhosinfectadoscom

    Plasmodium no sangue antes da reinfeco, resulta na perda deste efeito

    protector.

    Vriashipteses,baseadasemtrabalhosanterioresaesteenumestudode

    expressogenticaquerealizmosparamelhorcompreenderasalteraes

    hepticas em resposta presena de Plasmodium no sangue, foram

    colocadas na tentativa de entender o mecanismo pelo qual se observa

    tamanhareduona infecode fgadoderatinhosreinfectados.Diversas

    molculas associadasao sistema imunitrio, inflamaoe apoptoseno

    fgado foram testadas pormeio de ratinhos transgnicos, ou pelo uso de

    anticorpos depletantes ou drogas bloqueantes sem que nenhum dos

    factorestestadosindicasseserrelevante.

    A infeco de hepatcitos cocultivados com eritrcitos infectados com

    Plasmodium, no mostrou ser eficiente em produzir o mesmo tipo de

    reduo,afastandoahiptesedeumfactorsolvellibertadopeloeritrcito

    infectado.

    Ao avaliar alteraes no fgado relacionas com a disponibilidade de

    nutrientes verificmos que o gene codificante da hormona reguladora do

    ferro, hepcidina se encontrava sobreexpresso no fgado de animais com

    parasitasnosangue.Sabendoqueadisponibilidadedeferropodelimitaro

    crescimentodevriospatogniosincluindooPlasmodium,perguntmonos

    seseriaesseocernedareduonainfecoverificada.

  • resumo

    XIV

    A expresso do gene codificante da hepcidina provou estar intimamente

    ligadapresenadeeritrcitos infectadosnosanguederatinhos, subindo

    rapidamente com poucos ciclos replicativos de Plasmodium no sangue, e

    voltando rapidamente a valores basais com o tratamento antimalrico, o

    que rapidamente reduz a parasitemia para zero permitindo ento que a

    infeco heptica se processe como em ratinhos naive. Alm disso, pde

    verificarse uma redistribuio de ferro no fgado, perdendose parte do

    contedo nos hepatcitos para incrementar o contedo em macrfagos

    residenteseinfiltradosnofgado.

    Adicionalmente,mostrousequeahepcidinapersepodereduzirainfeco

    heptica.Aadministraoaratinhosdeumadenovrusexpressandoogene

    codificante desta hormona reguladora de ferro confirmou que este

    componente promovido pelo parasita no sangue,mesmona sua ausncia,

    podeactuarlimitandoainfecoporesporozotos.

    Ao tentar perceber a implicao destes resultados na malria humana,

    deparmonos com dados epidemiolgicos h muito conhecidos mas

    parcamente explicados. Em reas altamente endmicas conhecido

    consistentementequeaincidnciadainfecoaumentainicialmentecoma

    idade das crianas para depois decrescer, possivelmente pela aco da

    imunidade adquirida. Ao mesmo tempo, a complexidade da infeco, em

    nmerodeclonesdeparasitasdiferentesnosanguedeindivduos,aumenta

    medidaqueascrianascrescememidade.

    Divisando um modelo baseado unicamente na existncia de um valor

    mnimodeparasitmiaqueinibisseoestabelecimentodainfecoheptica,

    tal como foi observado no decurso desta tese, e assumindo o h muito

    estabelecido,queadensidadedeparasitasnosanguedecrescecomaidade

    dos indivduos. Instituise ento que a probabilidade de um picada

    infecciosaproduzir infecodependedonveldeparasitmianomomento

  • resumo

    XV

    dainfecoedahistriaclnicadohospedeironoqueconcerneaepisdios

    prvios. Sob estas assumpes mnimas o modelo criado prev

    correctamente um aumento inicial de infeces nas crianas seguindose

    um declnio medida que os indivduos adquirem imunidade devido a

    repetidasinfeces.Queristodizerqueumadensidademnimadeparasitas

    nosangue,daqualdependaainibiodenovasinfecesdePlasmodiumno

    fgado, pode por si justificar o aumento do risco de infeco e crescente

    complexidadedasmesmasemcrianasnovas.

    A aplicao do modelo com os resultados esperados mais

    proeminentementeobservadaquandotestadosegundoreascomtaxasde

    transmisso moderadas ou elevadas, e permite explicar ainda algumas

    diferenasdeincidnciadadoenaaolongodaidadedascrianasemreas

    comdiferentestaxasdetransmisso.

    O ciclo de vida do Plasmodium tem vindo a desenvolverse ao longo de

    milhes de anos de coevoluo das interaces hospedeiroparasita, com

    implicaes importantes para a sade humana. A infeco de eritrcitos

    acimadeumadensidademnimaelevaaproduodahormonareguladora

    doferro,hepcidina,queredistribuindooferroprotegeonichodoparasita

    existente, inibindo o estabelecimento de uma infeco secundria,

    prevenindo assim a superinfeco. Este fenmeno actua

    independentementeedeformacomplementarimunidadeadquiridaevem

    aclarar observaes epidemiolgicas prvias, podendo ainda ter

    implicaesemfuturasintervenesnalutacontraamalria.

    Palavraschave:malria,reinfeco,superinfeco,hepcidina,ferro

  • XVI

  • summary

    XVII

    Summary

    Inregionsofhighmalariatransmission,infectedindividualsareconstantly

    exposed to potential reinfection. Mosquito bites transmit livertropic

    sporozoitesintosubjectswhoalreadyhavebloodstageparasitaemia.How

    these two stages of thePlasmodium life cycle interactwithin their host is

    unknown. Here, using a rodent model, we show ongoing blood stage

    infections impair the growth of subsequently inoculated sporozoites.

    Secondary infectionsarearrested in liverhepatocytesand fail to compete

    forcolonizationofredbloodcells.Thisprotectionoftheerythrocyteniche

    onlyoccursbeyondacertainthresholdofbloodparasitedensity,andsois

    phenotypicallyakintoquorumsensing.WeeliminatePlasmodiumsecreted

    factors,hostcellsurvivalandinnateoradaptiveimmunityasexplanations

    for this observation. Instead, we find parasitized erythrocytes induce

    expression of the host iron regulatory hormone hepcidin, which by

    divertingironfromhepatocytestomacrophages,limitsPlasmodiumgrowth

    intheliver.Presumingasimilarinteractionbetweenmalariaandthehuman

    host we demonstrate how parasite thresholddensity dependent growth

    inhibitionalonecanexplaintheepidemiologicalpatternsofagerelatedrisk

    and complexity of infections in young children. Our findings thus have

    broad implications for malaria and have general relevance for

    understandinghostpathogeninteractions.

  • XVIII

  • XIX

    Top 10 Abreviations

    CQ ChloroquineEEF ExoErythocyticFormsEIR EntomologicalInoculationRategfp GreenFluorescentProteinhamp Hepcidinhprt HypoxanthineGuaninePhosphoribosyltransferaseiRBC InfectedRedBloodCellPbA P.bergheiANKAPbNK P.bergheiNK65iRBCsRBC RedBloodCell

  • XX

  • XXI

    Table of contents

    Preface VAcknowledgements VIIResumo XISummary XVIITop10Abreviations XIXTableofcontents XXIIntroduction 1Malaria 3Plasmodiumphylogenyanditscoevolutionwithman 4ThePlasmodiumlifecycleinmammals 8Malariatransmissionandnaturallyacquiredimmunity 22Aims 28

    MaterialsandMethods 31Results 47BloodstagePlasmodiumparasitessuppresscoinfectionintheliver 49AdditionalResults 87

    Discussion 99Generaldiscussionandconclusions 101Thenextinline 123

    Appendix 127Bibliography 153

  • XXII

  • Introduction

  • 2

  • introduction

    3

    Fromthebeginning

    Malaria

    Descriptions of malaria appear in history prior to its name in several

    ancestralcivilizations.AncientChinesemanuscriptsdatingasearlyas2700

    BChavedescriptionsofpatientswithfeveraccompaniedbyenlargedspleen

    (reviewed in 1). Later, Indian and Egyptian manuscripts also recorded

    indications of malaria infection. Furthermore, DNA of one of the species

    causing malaria, Plasmodium falciparum was recently isolated from an

    approximately 4000 year old Egyptian mummy 2 and Plasmodium

    falciparum DNA was also found in the mummy of Egyptian King

    Tutankhamunandthreemembersofhisfamily3.ReportsbyHippocratesin

    400BCdescribed for the first time thevariousperiodicmalaria fevers. In

    theCorpusHippocraticum,hedistinguishedthe intermittentmalarial fever

    from the continuous feverofother infectiousdiseases, andalsonoted the

    daily, everyotherday, and everythirdday increase in body temperature

    (reviewedin4).

    Fig. I1.LaMal'aria (18501851), oil over canvas by the French artist AntoineAuguste Ernest Hbert (18171908). Reproduction rights requested to Muse dOrsay andMuseHbert.

  • introduction

    4

    ThenameMalaria appears later from the Italianmal'aria, translating into

    badair,asinRome,wherethediseaseragedforcenturies,itwascommonly

    believed until the 20th century that swamp and marshes produced the

    illness(reviewedin5).

    Inworldhistory, thedisentanglementof theGreekEmpire isattributedto

    malariaandthediseaseisalsobelievedtohavestoppedarmiesofEuropean

    andAsianempiresondifferentoccasions1.Nowasthen,malariastilllimits

    civilizations. Indeed, in the secondhalf of the20th century, countrieswith

    intensive malaria showed an average increase of gross domestic product

    (GDP)5timeslowerthanthatobservedfornonmalarialcountries6.

    The clinical features of malaria infection, which made it so recognisable

    throughout centuries, occur only during the asexual cycles ofPlasmodium

    parasite inside red blood cells (RBCs). In humans, symptoms range from

    mildfevertocoma,severeanaemia,respiratorydistressorcerebralmalaria7.The2009WorldMalariaReportoftheWorldHealthOrganization(WHO)

    states that 108 countries were endemic for malaria in 2009, and that

    863000deathswereattributabletomalariain2008,themajorityofwhich

    were African children under 5 years of age 8. Moreover, the disease is

    estimatedtoberesponsibleforanannualaveragereductionof1.3%inthe

    economicgrowthforthosecountrieswiththehighestburden9.

    Fromtheverybeginning

    Plasmodiumphylogenyanditscoevolutionwithman

    Plasmodium is the causative agent of malaria and belongs to the

    Apicomplexaphylum.Apicomplexanshaveprobablybeenpresentonearth

    since the earliest animals appeared and modern Plasmodium species

    descend from apicomplexans that interacted and adapted to increasingly

    sophisticated hosts over hundreds of millions of years 10. Species of the

    genus Plasmodium have coexisted with humankind throughout its

  • introduction

    5

    evolution.P.falciparum,P.vivax,P.ovaleandP.malariaearethespeciesthat

    systematically infecthumans. Inaddition,a fifthone,P.knowlesi,anatural

    parasite ofmacaquemonkeys in southeast Asia,was recently reported to

    infect considerable numbers of humans 11, besides several reports on

    isolated cases in severalAsian regions 12,13. Likewise, recentwork reports

    that P. falciparum infections are not rare in gorillas 14. These species

    represent only a glimpse of the systematic and ecological diversity of

    PlasmodiumandrelatedgeneraofApicomplexaparasites.Severalhundreds

    of malaria parasite species use squamat reptiles, birds, and mammals as

    vertebrate hosts along with many genera of dipteran vectors 15, but the

    evolutionary and ecological events that led to this diversification and

    successremainpoorlyresolved.Recentgeneticdatachallengedtheclassical

    systematic classification ofmalaria parasites, bothwithin thePlasmodium

    genus, and in its relations among the currently recognized genera of the

    orderHaemosporidiaeofApicomplexaparasites.Astudyrecoveringmulti

    gene phylogeny ofmalaria parasites, including the analysis of sequencing

    datafrom4genes,indicatedthatparasitesidentifiedasPlasmodiumspecies

    fall in twomajor clades, each cladebeingassociatedwithauniquevector

    family,onecladecontainingparasitesofmammals,andotheroflizardsand

    birds. Within the mammalian parasite clade, there are 4 monophyletic

    lineages: the human parasite, P. falciparum, a primate lineage that

    containsP. vivax andP. knowlesi, a lineage of parasites that infectAfrican

    rodents,andthethreesamplesofHepatocystisisolatedfrombats16.

    Evolutionarily,Plasmodiumemergencewasaccompaniedbyamajorchange

    in parasites life cycle that differed from other genera. In addition to an

    initial roundofasexual replication in fixed tissues thatwasgeneral forall

    parasites, thePlasmodium life cycle includes additional rounds of asexual

    reproduction in blood cells, revealing increasing complexity when

    compared to ancestral parasites. There was also a shift to the use of

  • introduction

    6

    mosquitovectors,whichseems tohavemade theexploitationofagreater

    varietyofvertebratetaxaashostspossible16.

    Fig.I2.PhylogenetictreeofmalariacausingparasitesAdaptedfromamultygene analysis study performed by Ellen S.Martinsen and collaborators, using 11mammalian parasite species, 7 species that infect lizards, and 39 lineages fromavianhosts.

    It has been established that Plasmodium entered mammals only once,

    coincident with a switch from Culicine to Anopheline mosquitoes 17. All

    knownvectorsofmammalianinfectingPlasmodiabelong to theAnopheles

    genus 18, indicating that the shift of Plasmodium into mammals was

    associatedwithspecializationtoanophelinevectors.Itiswellacceptedthat

    the closest identified sister taxon of P. falciparum is P. reichenowi whose

    hostisthechimpanzee19.ItwasrecentlyproposedthatextantP.falciparum

    populationsoriginated fromP. reichenowi as late as1.5millionyears ago,

  • introduction

    7

    likelybyasinglehost transfer 20,anevent thatappears tohavehappened

    frequentlybetweenprimates, includinghumans 19.Othersbelieve that the

    appearance of P. falciparum occurred even later during the initiation of

    agriculture, ten thousand years ago 21. Still, there is some controversy

    around theseproposalsand the relationshipbetweenP. falciparumandP.

    reichenowiisnotyetcompletelyclear.

    Nevertheless, it isclear thatPlasmodium (or itsancestors)and theirhosts

    had millions of years to coevolve and coadapt, leading to a balance

    between parasite replication and host survival enabling transmission to

    otherhosts.ApossibleexplanationforP.falciparumbeingthemostvirulent

    speciesinfectinghumansisthefactthat,beingmuchmorerecentthanother

    Plasmodium species, it hasnot yet had thenecessary time to finetune its

    effectsonthehostpopulation.

    Despite the significant burden of malariarelated mortality only a small

    proportion of infections lead to severe disease or death. Plasmodium

    infections in malaria endemic regions most often go unnoticed even in

    children, who are themost affected sector of the population. In children,

    around half of all infectious bites produce no infection, about a quarter

    leads to asymptomatic infections, roughly the other quarter gives rise to

    uncomplicated malaria with mild symptoms, and only a very small

    proportion of the infective bites will induce severe malaria with

    manifestations such as coma, cerebral malaria, respiratory distress, or

    severeanaemia22thatcanultimatelyleadtodeath.Still,theprevalenceofP.

    falciparum is high enough to kill more than half amillion children every

    yearinsubSaharanAfrica.

    EvidencefortheadaptationofPlasmodiumtoman,andmansstrategiesto

    circumvent malaria infection can be seen in how the infection has

    modulated certain human genes in affected populations, selecting for

    resistance.AsHaldanehypothesisedandAllisondemonstrated23,malariais

  • introduction

    8

    knowntobeastrongevolutionaryforceofselectionintherecenthistoryof

    the human genome. Sicklesell disease, thalassemia, and glucose6

    phosphatase deficiency, among other erythrocyte defects that together

    comprise the most common Mendelian human diseases, are under

    Plasmodiuminfectionselectionforce,andcanaidunderstandingthegenetic

    basis of resistance shownby some exposed populations. Indeed, different

    resistant alleles have been identified for sickle cell trait in different areas

    andatdifferenttimes,suggestingconvergentevolution(reviewedin24).

    Plasmodiummodusoperandi

    ThePlasmodiumlifecycleinmammals

    Alfonse Laveran identified the exflagelating malaria parasite in human

    blood in 1880 25. Eight years later, using an avian model, Ronald Ross

    attributed the transmissionofmalariadisease to theanophelinemosquito26.Thefirstobservationofthepreerythrocyticstageparasitewasdescribed

    laterin1948byH.E.ShortandP.C.Garnhamintheliverofasimianmodel27.Thethreestagesofmalariainfectionwerethendescribed,andfromthere

    onresearchtriestofillinthegaps,andunderstandtheexactdetailsofthe

    entire life cycle, what the parasites needs are, and how host and vector

    moleculescontributetoorfightinfection.

    Duringitslifecyclethemalariaparasitetakesmanyforms,shiftingbetween

    invasive and replicative stages both in the vertebrate host and in the

    anophelinevector.

  • introduction

    9

    Fig.I3.Plasmodiumlifecycle(A)Duringabloodmealananophelinemosquitoinjects Plasmodium sporozoites into the host dermis. (B) After reaching a bloodvessel, sporozoites will travel to the liver where after traversing severalhepatocytes, sporozoites invade a final one. (C) After asexual replication anddevelopment inside a hepatocyte,merozoites are released into the blood stream.(D) Merozoites infect red blood cells during cycles of asexual replication. (E)Occasionally replication cycles will originate female and male gametocytes. (F)Throughanotherbloodmeal, amosquito ingestsgametocytes into itsmidgut. (G)Fertilization of gametes occurs in the mosquito midgut with the formation ofookinetesand latertheoocysts. (H)Sporozoitesreleasedfromtheoocystmigratetothesalivaryglandofthemosquitoawaitingthenextbloodmeal.Depositionintheskin.Allnaturalhumanmalariainfectionsstartwiththe

    depositionofPlasmodium sporozoitesbya femaleanophelinemosquito in

    thehostsdermis,duringabloodmeal.Eachsporozoiteisapproximately10

    mlongand1mwide,andonceinthedermis,oneoffourpossiblefates

    willdetermineitsfuture:(i)itcaninvadeabloodvesselandreachtheliver27,(ii)itcaninvadealymphaticvesselandaccumulateinthedraininglymph

    node28,(iii)itcanremaininthedermis29,or(iv)itcanbereingestedinto

    the mosquito gut during the blood meal 30. Only the first of these fates

    allowsinfectiontoproceed.

  • introduction

    10

    Intheavasculartissuesofthedermis,sporozoitesactivelyglideforwardin

    random directions. This movement however, seems to be specific to its

    substrate, as it differs from the one observed in salivary gland ducts and

    from that exhibited in vitro 28. Like other apicomplexan parasites,

    Plasmodium sporozoites display a unique kind of locomotion, known as

    glidingmotility,definedby the lackofobviousmodification in themoving

    cell shape, and theneed for a supporting substrate 31. Theparasite glides

    both to achievemotility and to invade cells, and the twomechanisms are

    madepossiblebythesameactomyosinmotorlocatedinthecorticalspace

    inbetweentheplasmamembraneandtheinnermembranecomplex(IMC),

    a continuous layerof flattenedvesiclesunderneath theplasmamembrane3134. In this space, transmembrane proteins displaying adhesive

    extracellular domains and anchored to the motor trigger either forward

    locomotionor penetration into thehost cells 31,35,36. One suchmolecule of

    particular importance is the thrombospondinrelated anonymous protein

    (TRAP),whoseexpressionisrestrictedtoPlasmodiumsporozoites37,38.

    Mostsporozoitesremaininthedermisforatleast5minutes,astheremoval

    of the biting site 5 minutes after the bite greatly reduces the number of

    infections in mice. However, a similar removal 15 minutes after the

    mosquito bite does not cause any significant alteration in the outcome of

    liverinfection39,showingthatduringthistimeframefollowinginoculationa

    significantandsufficientproportionofthesporozoitesleavethebitingsite

    to find a blood vessel, which will later lead to infection of hepatocytes.

    Nevertheless, it takes one hour for half of the inoculated sporozoites to

    leave the dermis, either into blood or lymphatic vessels, and sporozoites

    havebeenfoundinthedermisupto7hoursafter injection40.Duringthis

    period in the skin, sporozoites might trigger the first line of immune

    response.

  • introduction

    11

    To leave thedermis sporozoitesmaymakeuseof their ability to traverse

    cells41,disruptingmembranesofendothelialcellsandpenetratingvessels.

    Severalproteinshavebeenidentifiedasessentialforcelltraversal,suchas

    the sporozoite microneme protein essential for cell traversal (SPECT) 42,

    SPECT243, cell traversalprotein forookinetesandsporozoites (CelTOS) 44

    orphospholipase (PL) 45. Indeed, sporozoites lackingSPECTareunable to

    reachbloodvessels41.

    Closetoonethirdofsporozoitesinoculatedintomicethroughmosquitobite

    are later found in lymphaticvessels 28.Fromthere,onlya fewsporozoites

    canberescuedbacktobloodcirculation,whiletherestaredrainedtothe

    closest lymph node. Once there, some sporozoites have been observed to

    developintoyoungandsmallExoErythocyticForms(EEFs)duringthefirst

    hours. This initial development is not continued and, instead, these small

    EEFs are then cleared, which seems to contribute to the initiation of an

    immuneresponse46.

    Themechanismofinvasionofthebloodvesselsisstillnotfullyunderstood.

    Ithasbeendescribedthatglidingspeedisdecreasedwhensporozoitesare

    incontactwiththevesselsanditisknownthattheentryprocesslastsless

    thanaminute47.

    Theliverstage.Onceinsidethevessel,sporozoitesarecarriedintheblood

    flowatthesamespeedaserythrocytes40,andwill laterbearrestedinthe

    liverbynotcompletelyunderstoodmechanisms.Circumsporozoiteprotein

    (CSP),aproteinencodedbyasinglecopygene,andthemajorcoatprotein

    ofPlasmodiumsporozoites48,hasbeenshowntobeinvolvedinthisprocess.

    IndeedrecombinantCSPbindsspecificallytosulfatedglycoconjugates49.It

    has beenproposed that heparan sulfate proteoglycans (HSPGs) of stellate

    cells extending through endothelial fenestrations to the sinusoidal lumen,

    canattractandarrestsporozoites.Thesecellssynthesizeproteoglycansthat

    areeight timesmoresulphatedthanthoseofhepatocytesand incorporate

  • introduction

    12

    twice the amount sulphate into heparan sulphate, known to be

    preferentially attractive to sporozoites 50. Being unable to pass through

    fenestrationsof the liversinusoid,sporozoitesmustmigratethroughcells,

    mostly Kupffer cells, using their capacity to traverse barriers and finally

    access the liver parenchyma 51. Still, this is a matter of controversy and

    someauthorshaveproposedthatthepassagethroughKupffercellsoccurs

    byamechanismakintophagocytosis52.Oncehavinggainingfreeaccessto

    the liverparenchyma,sporozoitestraverseseveralhepatocytes,disrupting

    theirplasmamembranes toget in andoutof cells 53, until theyultimately

    invadeafinalhepatocytewithformationofaparasitophorousvacuole(PV).

    Differentstudieshaveshownhepatocytetraversalmakessporozoitesprone

    forfinalhepatocyteinfection,eitherbyactivatingsporozoitestoformaPV54,55orbyleadingtothereleaseofhepatocytegrowthfactor,whichsustains

    hostcellviability56.Importantly,althoughdemonstratedtobebeneficialfor

    infection,traversalabilityofPlasmodiumsporozoiteshasbeenshownnotto

    be essential for hepatocyte invasion in vitro, since parasites lacking

    essential proteins for cell migration as SPECT, CelTOS or PL still invade

    hepatocytes and complete liver stage development similarly to wild type

    parasites 4244. Notmuch is known about the signal to stopmigration and

    invade,butmousestudiesindicatethatsporozoitesareactivatedtoinitiate

    the invasion process in the presence of cells expressing high levels of

    sulfatedHSPGs50,54.

    To invade hepatocytes, Plasmodium sporozoites, like other apicomplexan

    parasites, release proteins from apical organelles termed rhoptries,

    micronemesanddensegranules,andattachtohostcellsusingcelladhesive

    domains of some of these proteins. TRAP seems to be essential to this

    process57.Althoughadhesiontothehostcell is initiallyreversible, it later

    becomesirreversible,formingtightjunctionsthatwillallowtheparasiteto

    moveforwardpropelledby itsactomyosinmotor,allowing it toenter the

  • introduction

    13

    cell.With the completionof the tight junctions, the sporozoite is included

    insidethehostcellsurroundedbythePVmembrane58.

    Insidehepatocytes,sporozoitesdevelopandreplicateoriginatingthousands

    of merozoites that ultimately will proceed to blood infection. The

    astonishingreplicationrateobservedmostcertainlyimpliesstrongcellular

    andmolecularhostparasiteinteractions.Althoughlittleisknownaboutthe

    intrahepatic development of Plasmodium parasites, recent studies have

    providedanewandmorecomprehensiveinsightintogeneexpressionand

    protein abundance profiles of Plasmodium liver stage 59. Plasmodium

    sporozoite and liver stage asparaginerich protein (SLARP) has been

    observed to be essential for initiation of EEF development, as parasites

    mutant for this protein are arrested in very early development 60. Other

    parasiteproteinshavebeenshown tobeessential fordevelopment inside

    hepatocytes. Parasites deficient for the microneme protein P36p present

    reduced invasion and impaired development 61, and upregulated in

    infective sporozoites genes 3 an 4 (UIS3) or (UIS4)deficient parasites

    present impaired development 62. Plasmodium Fatty Acid Synthesis II

    pathwayenzymeswere found tobemodulatedduring infection, and later

    functionalstudiesrevealedthattype2fattyacidsynthesisisimportantfor

    liverstagedevelopment63and,morespecifically,essentialforthelatesteps

    of liver stage development 64. Another recent study showed that cGMP

    dependent protein kinase (PbPKG) disruption in sporozoites leads to an

    arrestofliverstages65.

    Hostmoleculeshavealsoproventobeimportantforcompletedevelopment

    of the Plasmodium parasite inside hepatocytes. The expression of several

    host genes is modulated during liver infection, as shown recently in a

    microarray study comparing infected and noninfected cells patterns of

    geneexpressionover the timecourseof infection 66. In functional studies,

    tetraspaninCD81 67 and scavanger receptorB1 68 havebeen shown to be

  • introduction

    14

    importantforliverstagedevelopment.Theirabsencereducesliverinfection

    toverylowlevelsinrodentmodels.Yetanotherfunctionalscreen,aimedat

    the host kinome, has linked an efficient parasite infection with several

    humankinases69.

    During the liver stage, as will later be shown for the blood stage,

    PlasmodiumparasitesseemtoexportproteinsbeyondthePVtothecytosol

    of the hepatocyte or even to the hepatocyte nucleus. Up to now this has

    been shown for CSP, and mutant parasites incapable of this export

    movementshowimpaireddevelopment70.

    By theendofexoerythrocyticstage,merozoitesarereleased in theblood

    stream within large vesicles known as merosomes 71. These budding

    structures full of merozoites squeeze through endothelial cells, and are

    initially hidden from the host's innate immune system by being covered

    withahepatocytederivedmembrane.

    The blood stage. After reaching circulation, merosomes release the

    merozoites into the bloodstream. This seems to occur only when

    merosomesreachthelungmicrovasculature.Althoughthereasonbehindis

    not known, it has been speculated that low macrophage density and

    reduced blood velocity with reduced shear forces within the lung

    microvasculatureenhancestheabilityofmerozoitestoinvadeerythrocytes,

    sinceoncemerozoitesarefreeincirculationtheyquicklyhavetofindared

    bloodcelltoinfect.72.

    MerozoitesinvadeRBCs,highlydifferentiatedcells,whosemainpurposeis

    oxygen distribution throughout the body. However, RBCs lack several

    cellular functions as they are anucleated, transcriptionally and

    translationally inactive, lack any secretory apparatus, have only a limited

    repertoire of solute and iron transporters, and are readily removed from

    circulationwhendamaged(reviewedin73).

  • introduction

    15

    Invasion of RBCs follows sequential steps: cell recognition, merozoite

    binding,reorientationanddeformationoftheRBC, junctionformationand

    parasiteentry.IninP.falciparumalone,morethan50surfaceandsecreted

    proteins have been identified as playing a role in these events 74. Initial

    hostparasite contact is believed to be randombut, once it has happened,

    merozoitesurfaceproteins(MSP)immediatelymediateadhesiontothehost

    cell 75. Themerozoites surface is covered in glycosylphosphatidylinositol

    (GPI)anchoredproteinsandtheirpartners76.Manyoftheseproteinsseem

    toactasligandsforRBCs.BesidessharingGPIanchors,surfaceproteinsof

    the merozoite also share with sporozoites and gametes the cysteinerich

    domains that have been shown to potentiate adherence 77 ofPlasmodium

    species.Assuch, it isbelievedtheyplaysimilarrolesinthecytoadherance

    during the blood stage of infection. In order to invade RBCs, merozoites

    makeuseofcomplexmechanisms,evidencinggreatadaptationtotheirhost.

    Someparasitestrainsthatdependonsialicacidreceptors,forexample,can

    evenshifttheirinvasionpathwaytoasialicacidindependententryprocess78.

    Afterthisinitialbinding,thereisareorientationofthemerozoiteinorderto

    position its apical and secretory ornanelles (rhoptries, micronemes and

    densegranules)incontactwiththeRBCmembrane.Anindentationisthen

    formed in the contact zone.Anumberof proteins of the apical organelles

    bindtospecificerythrocytereceptors.Apicalmembraneantigen1(AMA1)

    is known to establish the apical interaction through parasite adhesins

    initially located at the neck of the rhoptries and in the micronemes 79.

    Recently, parasites with green fluorescent protein (GFP)tagged AMA1

    were used in live imaging studies that revealed this proteins crucial

    function80.

    Theactual invasionprocess involves twomajorprotein families, theDuffy

    bindinglike (DBL) protein family 81, and the P. falciparum reticulocyte

  • introduction

    16

    binding protein homolog (PfRh) family 82. DBL and PfRh proteins are

    importantformerozoiteinvasionofRBCs,butarenotconsideredessential,

    asgenedisruptionforeachofthemindifferentP.falciparumlinesstillshow

    normal bloodstage growth rates 8387. This might be due to

    complementationbetweendifferent familymembersor to a great level of

    adaptation of Plasmodium to its host cell. When the apical interaction is

    formed,themerozoiteestablishesatightjunction,whichisaccompaniedby

    the releaseofmoreproteins from themicronemeand rhoptry organelles.

    When attachment is finally achieved, the merozoite is hypothesised to

    discharge mediators into the RBC, and although these have not yet been

    identified, visualization of merozoite invasion by electron migrographs

    suggests material transfer 88. Entry is thenmediated by activation of the

    actomyosinmotorinthepellicleoftheinvadingmerozoite89.Attheendof

    theinvasionprocess,theRBCbilayerenvelopstheinvadingmerozoiteina

    newvacuole90,whichlacksnormalproteinsoftheRBCmembrane.Proteins

    likeBand3arecleavedbyproteolyticenzymessecretedbytheparasite 91

    and are replaced by recruitment of detergentresistantmembrane (DRM)

    raftproteins,makingthePVricherinlipidraftsanditsproteins90.

    Inside the normally quiescent environment of a RBC, the merozoite

    undergoes some rounds of nuclear division, transforming sequentially in

    ring,trophozoiteandschizontstages.Iteventuallyegressesandreleases16

    to 32 newmerozoites thatwill follow the same sequence of events. InP.

    falciparum,theringstagelastsapproximately24h,accountingforhalfofthe

    entireerythrocyticcycle92.

    Following entry, and throughout all the developmental and replication

    processes, theparasitemodifies thepermeability and adhesiveproperties

    ofitshostcell,possiblytopromoteitsownsurvival.

    The ring stage is followedby the trophozoite stage, duringwhichmost of

    the cytoplasm of the RBC is consumed, a process that leads to the

  • introduction

    17

    degradationof60to80%ofthehaemoglobinpresentintheRBC93.Thisis

    followed by the schizont stage, during which 4 to 5 rounds of binary

    replication take place, originating merozoites that will later invade other

    RBCs. The most common and concentrated molecule inside RBCs is

    haemoglobin that is degraded by Plasmodium. Haemoglobin proteolysis

    yieldshaembesidesaminoacidsasendproducts.Haemeisnotmetabolized

    orrecycledbytheparasite.Instead,itisstoredashemozoin,apolymerthat

    confersthecharacteristicpigmentationtotheorgansofinfectedindividuals94,95.Theaminoacidsofthehaemoglobinpolypeptidechainareusedinthe

    synthesisofparasitesproteins96andappeartobeusedasanenergysource95. The recovery of the amino acids is essential for the parasite as it has

    limited capacity of de novo synthesis 94. However, the recycling of amino

    acids originating from haemoglobin degradation is not sufficient for

    parasite maintenance as haeomglobin lacks some essential amino acids,

    whichtheparasiteisabletowithdrawfromthehostplasma97.Thefeeding

    of theparasite in theRBCoccurs through a cytostome, an invaginationof

    thePVmembraneandtheparasiteplasmamembrane,whichingestssmall

    packets of haemoglobin from the host cytosol. Budding vesicles full of

    haemoglobinaretransportedandfusedtoanacidicdigestivevacuole(DV)

    where haemoglobin is degraded and haeme is detoxified by the action of

    several proteases 98 acting like haemoglobinases. Data suggest that this

    degradation process is ordered and requires, among other reactions, an

    initialasparticproteasemediatedcleavage,followedbysecondaryaspartic

    proteaseandcysteineproteasecleavages.Theasparticproteaseshavebeen

    shown to be Plasmepsin I and II and the cysteine protease has been

    identified has Falcipain 99,100. Different patterns of gene expression are

    observedduring theerythrocytic cycle, suggesting that their functionsare

    not exactly the same. Several other Plasmepsin proteins were later

    identifiedbothinsideandoutsidetheDV.Theirfunctionalredundancywas

  • introduction

    18

    shown when independent disruption of Plasmepsin I, II, IV or histidine

    asparticprotease(HAP)wasobtainedandverylittleeffectontheparasite

    growthwasobserved101,102.

    Plasmodiumhastheabilitytoexporthundredsof itsownproteinsbeyond

    itsplasmamembraneandthePVtothecytosoloftheerythrocyte103.This

    represents5%ofitswholegenome.Althoughthefunctionofmanyofthese

    proteins remains unknown, some of them have been associated with

    virulence,promotingcelladhesionand/orrigidityoftheerythrocyte104,105.

    Tobeexported,proteinsmustfirstenterthesecretorypathway106showing

    a recessed aminoterminal hydrophobic endoplasmatic reticulum (ER)

    signal sequence that allows transport across theplasmamembraneof the

    parasite but not the PVmembrane.Most proteins exported across the PV

    membrane require an additional sequence element knownasPlasmodium

    export element (PEXEL), or a vacuolar transport sign (VTS) that is found

    downstream of the ER signal sequence 107. This export movement has

    recently been proposed to be performed by a proteinaceous translocon

    withinthePVmembrane,anATPpoweredcomplexcontainingheatshock

    protein 101, a novel protein PTEX150 and a known parasite protein

    identified as exported protein 2 (EXP2) that potentially works as the

    commonportalthroughwhichmostorallexportedproteinsmustpass108.

    MoreoverPlasmepsinVwasrecentlyshowntobetheproteaseresponsible

    forcleavageofthePEXELmotifofproteinstobeexported,therebyallowing

    theirtraversalofthePVandtraffickingtotheRBCsurface109,110.Following

    thisexportviathistransloconatthePVmembrane,exportedproteinshave

    toreachdifferentdestinationsinthehostRBC,suchasthecytoplasmorthe

    plasmamembrane. Because RBCs have no secretory system, the parasite

    buildsitsowntoallowitsproteinstoreachthehostcellplasmamembrane.

    Maurer's cleft is a central structural component of this extracellular

    proteinexportsystem111.Thesestacksofflattenedlamellaeoflongslender

  • introduction

    19

    membraneswithatranslucent lumenwere identifiedmorethanacentury

    agoandarelocatedbelowtheerythrocyteplasmamembrane112,113.Several

    parasiteproteinsaresynthesizedintheparasitesERandthentransferred

    tothecleft114,115.ItisstillnotcompletelyclearwhethertheMaurer'scleftis

    part of a continuousnetwork that connects thePV to the erythrocyte cell

    surfacelikesomeimagingreconstructionsofthinsectionsseemtosuggest116, or if the cleft is a welldefined structure continuously supplied with

    vesiclesbudding from themembrane lining thePV.Subsequently, vesicles

    bud out from the cleft and migrate towards the erythrocyte cell surface,

    withsecretionoftheircontentsintothemediumandincorporationofsome

    proteinsintheerythrocyteplasmamembrane74.

    The very strong remodelling of the host RBC eventually leads to its

    distortion,withpositivelychargedknobbyprotrusions.Usingatomic force

    microscopy, theknobwasshown toconsistof twosubunits thatmightbe

    central to thephenomenonof cytoadherence inP. falciparummalaria 117.

    Indeed, inP. falciparum infections,schizontsareknowntobarelycirculate118. Instead, they are sequestered in different organs by adhering to the

    endothelial cells of the vessels. Knobs are created by the deposition of

    parasite proteins, such as knobassociated histidinerich protein (KAHRP)

    andtheadhesionproteinP.falciparumerythrocytemembrane1(PfEMP1),

    whichareinsertedintheRBCmembrane.

    PfEMP1 is restricted to P. falciparum, and undergoes clonal antigenic

    variation switching, changing its antigen type at high frequencies during

    intraerythrocyticcycles119,120.PfEMP1belongstoalargepolymorphicgene

    familycalledvar,inwhicheachindividualgeneencodesadifferentformof

    the protein, and only one is expressed at a time through a mutually

    exclusive mechanism. It comprises three regions: the cytoplasmatic or

    acidic terminal segment (ATS) that is anchored to the knobs; the

    transmembrane regions that are inside the RBC membrane; and the

  • introduction

    20

    ectodomain orNterminal segment followed by theDBL domains and the

    Cysrich interdomain regions (CIDR) (reviewed in 74) that interact with

    endothelial cell receptors, leading to sequestration and thus preventing

    destruction of infected RBC (iRBC) in the spleen 121. PfEMP1 is highly

    relatedtovirulenceasitisbelievedthattheadherenceitpromotestriggers

    much of the associated pathology, including cerebral malaria end

    preganancyassociatedmalaria(reviewedin122).

    Following this period (whose length varies betweendifferentPlasmodium

    species) where Plasmodium replicates with the consumption of the RBC

    cytoplasm,parasitesleavetheRBCbyaprocesscalledegressthathappens

    veryfastandmustbetightlyregulated.Severalhypotheticalmodelsexplain

    egress.AlthoughthesevarybydisagreeingonwhetherthePVmembraneor

    theRBCmembranedisintegratesfirst,allthemodelsacceptthatproteases

    play a critical role. Protease inhibitors have been shown to prevent P.

    falciparumiRBCrupture,andtopromoteaccumulationofmatureschizonts

    invitroduetoegressblocking123,whichwaslaterattributedtoprevention

    of the proteolytic effect of serine repeat antigen (SERA)5 124. Further

    studies showed thatdifferentprotease inhibitorshavedifferent effectson

    egress 125. Still, the mechanism by which egress is regulated is only

    beginningtobeunderstood.Ithasbeenrecentlyreportedthatjustpriorto

    egress essential serine protease PfSUB1 is discharged from Plasmodium

    organelles called exonemes, from the merozoite into the PV 126, where it

    mediates theproteolyticmaturationofmembersofSERA family thathave

    been previously implicated in egress 127. Furthermore PfSUB1 has been

    shown to directlymediate primary proteolytic processing ofMSP1,MSP6

    andMSP7.PfSUB1seemstopreparenotonlythemerozoiteforreleasefrom

    theerythrocytebutalsotoensurethat,once incirculation,merozoitesget

    insideanewcellwithoutdelay128,avoidingexposuretothehostresponse.

  • introduction

    21

    Shifting tosexualstages.Differentiationandsexual commitmenthappen

    prior to schizogeny during asexual replication cycles inside RBCs. All

    siblings of a schizont are either asexual, female gametocytes or male

    gametocytes 129, but the mechanism that determines which of these

    possibilitiesoccursremainsunknown.TranslationalrepressionandmRNA

    turnoverhavebeenshowntobekeyplayers indeterminingstagespecific

    gene expression in Plasmodium. More specifically, development of zygote

    inhibitedRNAhelicase(DOZI)wasidentifiedinthefemalegametocyteand

    shown to have a central role in the silencing andmaintenance of steady

    statelevelsofapopulationofgametocytespecifictranscripts,allowingthe

    coordinated production of essential proteins for the further development

    andestablishmentofinfectioninsidethemosquito130.

    Inside the mosquito. Mature gametocytes in the blood can later be

    engorgedduringamosquitobloodmeal.Theabruptenvironmentalchange

    insidethemosquitotriggersgametocytestoroundupandemergefromthe

    RBCwithinminutes 131.Thepresenceof xanthurenicacidallows themale

    gametocytetotransformintoeightmotilemicrogametesafterthreerounds

    of exceedingly fast genome replication followed by nuclear division and

    axoneme assembly, and the female emerges from the erythrocyte as a

    roundshaped nonmotile gamete 132. Exflagelation of themale gametocyte

    and the vibratory movements in waves of its microgametes allow it to

    penetrateintothefemalegamete,andfertilizationtooccur133.Theresulting

    zygotewillbe theonlyparasitesurvivor in theaggressiveenvironmentof

    themosquitogut,andfurtherdevelopsintoatetraploid134motileookinete

    that is the only invasive formof thewhole cycle that is not originatedby

    replication.Thereisagreatlossofookinetesduringtraversalofthemidgut

    epithelialcellstoreachthebasallamina,duetohostprotectivemechanisms135.Onceinthebasallamina,ookinetesbecomesessileand,aftermeiosis136,

    transformintooocysts thatare theonlyextracellulardevelopmentalstage

  • introduction

    22

    of thewhole cycle. Oocystsmake use of their capsule to recruit nutrients

    from the hemolymph 137 to grow, 50 to 60m in diameter, and originate

    sporozoites.CSP, theprotein thatwillcoverallofsporozoites,starts tobe

    expressed and accumulate in the oocyst plasmamembrane even prior to

    sporozoite formation 138. With the retraction on the oocyst plasma

    membrane, several lobes called sporoblastsappear in this form 139.CSP is

    essential for this formation 140, and also seems to be essential for the

    organization of the microtubule organizing centres (MTOCs) 138 that will

    later lead to the formationof theapical complexandnucleipositioning in

    thedaughtersporozoites.

    Sporozoitereleaseintothehemolymphoccursasynchronously141andwas

    shown to be dependent on proteinase activity, including that of egress

    cysteineprotease1(ECP1)142.Oncereleasedinthehemocoel,sporozoites

    canbespreadinthewholemosquitobodybutonlyrecognizespecifichost

    receptorsinthebasallaminaofthesalivaryglands139.Invasionoccursonly

    inthisorganandseveralparasite ligands, likeCSP143,TRAP144orMAEBL145, have been identified as being important for recognition and invasion.

    Afterattaching,sporozoitesbreachthebasallaminaandinvadethroughthe

    basalmembraneofsalivarysecretoryacinarcellswiththebrief formation

    of a vacuole thatwill allow them to emerge from the apical side of these

    samecellsintothesalivaryglandduct146.

    Sporozoites are now ready to infect a new mammalian host when the

    infectedmosquitotakesanotherbloodmeal.

    Malariaoutcome

    Malariatransmissionandnaturallyacquiredimmunity

    MeasuringmalariaendemicityorPlasmodiumprevalenceinageographical

    area has been the subject of an active debate for decades. Surveys of

  • introduction

    23

    splenomegaly across apopulation 147, examinationofperipheral blood for

    asexual malaria parasites 148, stability/instability of transmission,

    entomologicalinoculationrates(EIR)149orhaemoglobinmeasurements150

    were, and are, some of the metrics to establish classes of endemicity inwhichagivenpopulationshouldfall.ConsideringAfricaalone,theexisting

    endemicpopulationsshowratesoftransmissionthatcandifferinintensity

    by100fold.

    Fig.I4.Global distribution of malaria transmission risk. Adapdted fromWHOWorldMalariaReport2005.

    Clinical presentations ofP. falciparummalaria vary according to different

    transmission intensities, but the biological interactions promoting these

    differentoutcomesarenot easilyunderstood. Studies across fifty yearsof

    field research in areas with different rates of transmission, in several

    communitieswithdifferentaccesstohealthsystemsortreatment,andwith

    variations in the prevalence of infection occurring from year to year in a

    givenplace,allowallsortsofinterpretationsconcerninghowmortalityand

    morbidityvarywithtransmissionsrates.Thereis,however,aconsensusin

  • introduction

    24

    accepting that the mean age of severe malaria disease decreases as P.

    falciparum transmission increases. The frequency of cerebral malaria

    declineswith increasing transmission rateswhile anaemia takes over the

    clinical burden (reviewed in 151). It is generally accepted that in low to

    moderatetransmissionsettings,theincidenceofseverediseasegrowsafter

    the first year of age, while in high transmission areas the peak in severe

    disease happens between the fifth and the seventh month of age, after

    which it declines significantly before the first birthday (reviewed 152).

    Common to all settings is the fact that for the first 3 months of age the

    incidenceofseveredisease isvery low,most likelydue to thepresenceof

    maternalantibodies 153. Inaddition, themuch lowerriskofseveredisease

    after5yearsofageindicatesthatsomedegreeofimmunityisacquiredearly

    in life. However, under very low transmission regions the risk of clinical

    malariaisextendeduntiladulthood154.

    Anextensivestudyinanareawithdecreasingparasiteprevalenceover16

    years showed that although transmissions decreased significantly in the

    first ten years, therewere nomajor alterations in clinical cases reported.

    Onlylater,whenparasiteprevalencefellbelowacertainthreshold,coulda

    drop in hospital admissions be observed. At the same time, as parasite

    prevalencedropped, themean age of slidepositive children increased 155.

    Moreover in crosssectional studieswith increasingmalaria transmission,

    there isan initial increase in therateofhospitaladmissionswithmalaria,

    but thereafter, the risk of hospitalization rises either more slowly or

    plateausatintermediaterangesoftransmissionintensityandmaydecrease

    slightlyinareasofveryhightransmission156.

    Very strong evidence that protection against malaria increases with age

    more thanwith any other factors comes from a study performed inMali

    where itwas observed that older children treatedwith chloroquinewere

    better able to clear chloroquineresistant P. falciparum parasites than

  • introduction

    25

    younger children treated in a similar way 157, again pointing towards an

    effectiveacquiredimmuneresponse.

    However, although in endemic settings older children and adults are

    resistant to severe morbidity and death, they are still susceptible to

    infection158,showingthatonlyacertainlevelofimmunitycanbedeveloped

    againstmalaria.Thisnatural immunity isacquiredat thecostofveryhigh

    early mortality and is still defective, as multiple infections are required

    before clinical protection is achieved, and persistent infection is typical.

    Indeed,backin1900,RobertKochcomparedareasofdifferentendemicities

    anddeducedthattherewastheneedforheavyandcontinuedexposureto

    theparasite inorder toacquireprotectionagainstmalaria.Later, in1920,

    the essential features of naturally acquired immunity againstPlasmodium

    weredescribed: itwasaccepted thatnatural immunitywas(i)effective in

    adultsafteruninterruptedlifelongheavyexposure,(ii) lostuponcessation

    of exposure, (iii) species specific, (iv) somewhat stage specific, and (v)

    acquired at a rate which was dependent upon the degree of exposure

    (reviewedin159).

    In spite of these premises, very little is known about the mechanisms

    through which immunity against malaria is acquired. Early studies with

    humans, showed that antibodies can protect againstmalaria infection 160,

    andseveralantigenshave laterbeenshown tobeassociatedwithmalaria

    protection.Antibodies againstMSP1 161,MSP2 162 orMSP3 163were found

    significantly elevated in the sera of protected individuals in different

    endemic areas and are thought to correlate with protection. These

    associations vary between studies, and antigens encountered until now

    show considerable polymorphisms through the existence of alternative

    allelic forms 164 and through antigenic variation 165, limiting their use as

    possibletargetsforvaccinationandtreatment.Also,theantibodyresponse

    to themost commonmerozoite antigens is believed to be shortlived 166,

  • introduction

    26

    peaking oneweek after themalaria episode and rapidly decaying to very

    lowlevelswithin6to8weeks.Thereasonforsuchashorthalflife isstill

    notunderstood.Itmightbethatthecatabolichalflifeoftheseantibodiesis

    justshorter,butotherhypothesishavebeenraisedsuchasthepredilection

    towardsIgG3whichisknowntobeshortlived167,orthepoordevelopment

    of memory or longlived plasma cells 168. Independent of the mechanism

    behind it, thisshorthalflifeofantimalariaantibodiescontinues tocreate

    problems when planning an intervention strategy toward an effective

    vaccineagainstmalaria.

    Liverstagenaturallyacquiredimmunityisnotbelievedtobestrongenough

    or acquired fast enough to provide any kind of protection in endemic

    populations. Still, it cannot be said that natural immunity against

    Plasmodium preerythrocytic stage is not involved in the final outcome

    observed. Indeed, infection of both humans and mice with Plasmodium

    sporozoites followed by chloroquine treatment that abrogates the

    establishment of blood stages protects individuals from subsequent

    infections169,170.Itistrue,however,thatearlystudiesperformedinhumans,

    bypassing the hepatic phase of the infection through direct blood stage

    infection, showed that immunity could be maintained (reviewed in 162).

    However,vaccinedevelopmenteffortshave investeda significanteffort in

    preerythrocyticimmunizationandprovenefficientinrenderingprotection

    forvariableperiodsoftime,probablybecauseoftheveryhighnumbersof

    attenuatedsporozoitesused171,whichareverydifferentfromtheverylow

    numbersofsporozoitesthatmosquitoesinjectduringabloodmeal172.Both

    attenuatedformsoftheparasite173andlivesporozoites170cancompletely

    protect humans, making them resistant to infection. However, so far the

    mostadvancedvaccinecandidateagainstPlasmodiumfalciparumisRTS,S.It

    is a recombinant, yeastexpressed subunit vaccine using hepatitis B virus

  • introduction

    27

    surfaceantigenscarryingepitopesfromP.falciparumCSP174themajorcoat

    proteinofsporozoites.

    InareaswithveryhighEIRs,aschildrengetoldermalariaattacksbecome

    lessfrequentafteraninitialpeakatyoungage,andparasitelevelsbetween

    manifestationssteadilydecrease.Althoughthechildseemstobeacquiring

    immunity, they remain parasitized, often with higher parasitaemias than

    otherchildrenatgreaterriskofclinicaldiseasefromareaswithlowerEIRs175. This protection against clinical disease is the product of acquired

    immunity that is able to control parasitaemia but does not fully abrogate

    infection, producing only nonsterilizing immunity 176. Several reports of

    Plasmodium superinfection describe single individuals hosting more than

    one Plasmodium species, or different genotypes of the same Plasmodium

    speciesinfectingRBCs177179.However,theseobservationshavebeenmuch

    more frequentamongasymptomaticcarriers than inclinicalcases 180,and

    havebeenshowntodependontheageofindividualsinapopulation175,181

    183.

    Premunition or concomitant immunity is theprotection against infections

    andclinicaldisease thatassociateswithpersistenceofmultiple infections,

    providingprotectionagainstnewinfectionsbymaintainingalowgradeand

    generally asymptomatic parasitaemia and high levels of antibodies

    (reviewed in 159,184). A better understanding of mechanisms behind

    premunition is clearly central to the comprehensionof naturally acquired

    immunity tomalaria. Periodic bloodstage infection presumably serves to

    boost a preexisting immune response and maintain high frequencies of

    effectorcellsinareasofhightransmission.Exposuretoagreatervarietyor

    antigensof familieswithassociatedvariance increases the repertoire that

    eachindividualrecognises185.

    Antigenic variation creates distinct waves of parasitemia that must be

    chased by different antibody responses, on the one hand promoting long

  • introduction

    28

    infections thatmightendanger thehost,whileon theotherhandallowing

    eventualacquisitionofdiseasecontrollingimmunity.Effectiveantiparasitic

    immunityis,ifever,achievedonlyafterverymanyandfrequentinfections149,186.

    So, thedoubt remains,whether reducing the risk ofPlasmodium infection

    willalwaysleadtoadecreaseinthethreatofseveremalariathroughoutlife.

    Thisisaconcernthatwasraised50yearsago,whenwonderingifaltering

    thenaturalriskofexposuretoparasite,byreducingvectorortransmission

    itself,would change the epidemiology of severe disease 187. Knowing that

    increasing exposure to Plasmodium parasites is reflected in increasingly

    rapidacquisitionofimmuneresponsesthatlimitslifethreateningeffectsof

    malaria, the fullunderstandingof the transmission/immunityassociations

    isofmajorimportanceindefiningmalariacontrolinterventions.

    Aims

    Plasmodium passage through the skin followed by infection of liver

    hepatocytes and, later, of blood erythrocytes, are natural and sequential

    steps of Plasmodium life cycle in themammalian host and should not be

    seen as independent entities. The sophistication and complexity of

    Plasmodium life cycle are evidence for coevolution with man, and are

    reflected in the complicatednatureof relationshipsbetween transmission

    rates, immunity and disease severity in endemic areas. Importantly, liver

    and blood stages of Plasmodium infection may frequently occur

    simultaneouslyinthesameindividualwheretransmissionsratesallowit.

    Indeed, after infecting the hepatocyte for approximately a week, P.

    falciparumcaninfectRBCsforweeksorevenmonthsafterthat188.Thus,a

    new infection initiated by amosquito bitemay occurwhile the parasites

    from a previous one are still replicating inside erythrocytes. Very little is

    knownabouttheinteractionsbetweenthetwodifferentstagesofamalaria

  • introduction

    29

    infection. So far it has been shown that malaria bloodstage can be

    immunosuppressive 189, but it is clear that very close interactions are

    establishedbetweenparasiteandhostthroughouttheparasiteslifecycle.

    The objective of thework presented here arises from intending to clarify

    effectsofonestageonanotherinindividualsduallyinfectedwithbloodand

    posterior liver stage malaria, and try to understand how reinfection fits

    what is known so far regardingdifferentpatterns of infection indifferent

    EIRsettings.

    ToaccessPlasmodiumbloodstage/liverstage interactionsnewtoolswere

    usedtooutwitpastlimitations,wehavemadeuseofwildtypePlasmodium

    parasitestostartbloodstageinfectionsandofgfpexpressingorluciferase

    expressing Plasmodium parasites to initiate liver stage infections. Using

    transgenic parasites we were able to distinguish the two infections

    circumventing the lackof specific liverstagemarkers.Because theGFPor

    luciferasegenesareunderthecontrolofaPlasmodiumhousekeepinggene

    promoter,theirexpressioncorrelateswithPlasmodiumliverload.

    ThestudyofthetemporalcoincidenceofPlasmodiumbloodandliverstages

    inasinglehostatagiventimewasmostlyperformedinrodentmodelsof

    infectiontoanswer4majorquestions:

    What would be the impact of an ongoing blood stage infection on the

    establishmentofasecondarysporozoiteinfectionintheliver?

    Bywhatmolecularmechanisms do any interactive effects between blood

    andliverstagesoccur?

    What, if any are the consequences of such interaction for the reinfected

    individual?

    Howsuch interactions adjust to established relationsbetweenpatternsof

    infectionandtransmissionratesindifferentendemicsettings?

  • 30

  • Materials and Methods

  • 32

  • materials & methods

    33

    Mice.C57BL/6,BALB/c,BalbSCIDaswellasmicedeficientinRagII,IL10,

    caspase 3, MyD88 or Kit Wsh/Wsh deficient were bred in the specific

    pathogenfree facilities of the Instituto de Gulbenkian de Cincia (Oeiras,

    Portugal).RAGII/cdeficientmicewerekindlyprovidedbyJamesDiSanto

    (CytokinesandLymphoidDevelopmentunit,InstitutPasteur,Paris,France).

    NOS2 (B6.129P2Nos2tm1Lau/J), TCRdelta (B6.129P2Tcrdtm1Mom/J),

    and C5a (B10.D2Hc0.H2d.H2T18c/oSnJ) deficientmice were purchased

    atTheJacksonLaboratoryalongwiththeirrespectivewildtypelittermates.

    AllmicewerehousedintheInstitutodeMedicinaMolecular(IMM)facilities

    andtheIMMAnimalCareCommitteeapprovedallprotocols.IFNdeficient

    mice were kindly provided by Rui Appelberg (Microbiology and

    Immunology of Infection laboratory, Instituto de Biologia Molecular e

    CelularIBMC,Porto,Portugal).ExperimentswithIFNdeficientmicewere

    performed at IBMC, and IBMC Animal Care Committee approved all

    protocols.AlltransgenicmiceweregenotypedbytailgenomicDNAPCRto

    confirmtheirrespectivemutations.

    Plasmodium blood infection. Primary blood stage infection ofmicewas

    achieved by 30 minutes exposure of mice to 15 Anopheles mosquitoes

    infected with P. berghei ANKA (parasite line GFPcon259cl2), or by intra

    peritonealinoculationofthedesignatedquantityofredbloodcellsinfected

    with P. berghei ANKA (1.49L), P. berghei NK65, P. yoelii 17X NL or P.

    chabaudi chabaudi AS. Peripheral blood parasitaemia was determined by

    GiemsastainingfollowedbymicroscopiccountingofiRBCsandresultsare

    expressed as percentage ofRBCs.On Fig. 1 C parasitemiawas verified by

    realtimeinvivoimagingusingtheinvivoIVISLuminaImagingSystem190

    asdescribedbellow.AndonFig. 1Eparasitemiawasdeterminedby flow

    cytometry, measuring red blood cells infected with gfpexpressing P.

    bergheiANKA.Theresultsareexpressedaspercentageofinfectedredblood

    cells,aspreviouslydescribed191.

  • materials & methods

    34

    Plasmodiumliverinfection.Greenfluorescentprotein(gfp)expressingP.

    berghei ANKA (parasite line GFPcon259cl2) or gfpexpressing P. yoelii

    sporozoites 191,192 were obtained by dissection of Anopheles stephensi

    infectedmosquitoesbredintheinsectariumofIMM.Micewereinfectedby

    intravenous inoculationof thedesignatedquantityofGFPsporozoites,or

    by30minutesexposureofmiceto15AnophelesmosquitoesinfectedwithP.

    berghei ANKA. Parasite liver load was quantified 40, 48 or 72h post

    infection.

    RealtimeinvivoimagingluminescentPlasmodium.Navemice(control)

    andbloodstage infected(PbAinFig.1AandPbNKinFig.1BandFig.1C),

    were intraperitoneally injected with 200l of anaesthesia mixture (80

    mg/kgKetamineand10mg/kgXylazine)diluted inPBS inorder toallow

    eachmouseexposure to15Anophelesmosquitoes infectedwith luciferase

    expressingP.berghei (parasite line354cl4).After30minutesofmosquito

    bites,micewereshaved intheabdomen.FortyhpostinfectionforFig.1A

    and 1B and 6 days postinfection for Fig. 1C, Dluciferin dissolved in PBS

    (150mg/kg;CaliperLifeSciences,USA)wasinjectedsubcutaneouslyinthe

    neck. Animals were anesthetized again as described above for the whole

    duration of measurements (performed within 5 to 10 minutes after the

    injectionofDluciferin).Bioluminescenceimagingwasacquiredwitha12.5

    cmfieldofview(FOV),mediumbinningfactorandanexposuretimeof180

    seconds. Luciferase activity in animalswas visualized through imaging of

    wholebodiesusingtheinvivoIVISLuminaImagingSystem190.

    qRealTimePCRquantificationof liver infection.Liverswere collected

    andhomogenizedindenaturingsolution(4Mguanidinethiocyanate;25mM

    sodiumcitratepH7,0.5%NLauroylsarcosineand,0.7%Mercaptoethanol

    in DEPCtreated water). Total RNA was extracted using RNeasy Mini kit

    (Qiagen),and thenreverse transcribed intocDNAusingTranscriptorFirst

  • materials & methods

    35

    Strand cDNA Synthesis kit (Roche), according to the manufacturers

    protocols. Infection load in the liver was determined as previously

    describedbyqRTPCRusingclassicPbA18SrRNA193specificprimersorgfp

    specificprimers(gfpexpressioncorrelateswithPbA18SrRNA,fig.S1).qRT

    PCR reactions used Power SYBR Green PCR Master Mix (Applied

    Biosystems) and were performed according to the manufacturers

    instructions on anABI Prism7000 system (AppliedBiosystems). Relative

    amounts of PbA 18S rRNA and gfp mRNA were calculated against the

    Hypoxanthine Guanine Phosphoribosyltransferase (hprt) housekeeping

    gene,followingaprimedenaturationof10minutesat95C,then50cycles

    of95Cfor15secondsand60Cfor1minute.PbA18SrRNA,gfpandhprt

    specific primer sequenceswere: 5CGG CTT AAT TTG ACT CAA CAC G3

    and5TTAGCATGCCAGAGTCTCGTTC3forPbA18SrRNA,5GTCAGT

    GGAGAGGGTGAAGG3and5ACTTCAGCACGTGTCTTGTAGTTC3

    forgfpand5TGCTCGAGATGTGATGAAGG3and5TCCCCTGTT

    GAC TGG TCA TT 3 for mouse hprt. External standardization was

    performed using plasmids encoding the fulllength genes cDNA cloned in

    TOPOTA(Invitrogen).

    Chloroquine treatment. Mice infected 4 days previously with P. berghei

    NK65 and control mice received 0.8 mg of chloroquine (CQ) by intra

    peritonealinjectionfor1or2daysbeforereinfection,aprotocolleadingto

    decreaseinperipheralbloodparasitaemiauntilzerobyday2oftreatment.

    Liver slice histopathology, morphometric analysis and

    immunofluorescence. Liver tissues were harvested from control or re

    infected mice 40 h after sporozoite infection. Tissues were fixed in 4%

    paraformaldehyde for 15 minutes, washed three times in PBS and then

    sliced into 50 m sections using a vibratome (VT1000S, Leica). Sections

    were later permeabilized and blocked overnight in 0.3% Triton X100

  • materials & methods

    36

    (Calbiochem)and1%BovineSerumAlbumin(Sigma)toavoidnonspecific

    reactivity. Sections were then incubated overnight at 4C in the same

    solution containing antiGFP IgG Alexa flour 488 congugate antibody

    (Invitrogen), Alexa 594 phaloidin (Invitrogen) and 4',6diamidino2

    phenylindole(DAPI,Sigma).Aftermounting15to20sectionsonslides,the

    areasofthesectionsweremeasuredusingascope,theEEFswerecounted

    usingtheLeicaDM5000BWidefieldFluorescenceMicroscope,andthesizes

    of 20 randomly chosenEEFs of eachmouseweremeasured using a Zeiss

    LSM 510 META Point Scanning Confocal Microscope. All images of

    immunofluorescencestainedsectionswereanalysedusingtheImageJ1.42b

    software. Areas and numbers of EEF were normalized to the total area

    observed.

    Cells. Huh7 cells, a human hepatoma cell line, were cultured in RPMI

    (Gibco/Invitrogen)mediumsupplementedwith10%fetalcalfserum(FCS,

    Gibco/Invitrogen), 1% penicillin/streptomycin (pen/strep,

    Gibco/Invitrogen), 1% glutamine (Gibco/Invitrogen) at pH 7 and

    maintainedat37Cwith5%CO2.

    Isolation of murine primary hepatocytes.Mouse primary hepatocytes

    were isolated as previously described 194. Briefly, cells were initially

    obtainedbyperfusionofmouse liver lobuleswith liverperfusionmedium

    and liver digest medium (Gibco/Invitrogen) at 37C using a peristaltic

    pump. Hepatocytes were then purified using a 1.12 g/ml, 1.08 g/ml and

    1.06g/ml Percoll gradient. Cells were cultured in Williams E medium

    containing 4% FCS, 1% pen/strep in 24well plates coated with 0.2%

    GelatineinPBS.Cellsweremaintainedincultureat37Cand5%CO2.

    Sporozoite infectionanddevelopment incontactwith infectedblood.

    Mouse primary hepatocytes and Huh7 cells, a human hepatoma cell line,

    were cultured as described above in complete Williams E or RPMI

  • materials & methods

    37

    (Gibco/Invitrogen) in transwell system plates (COSTAR/Corning). In the

    lower chamber, liverderived cells were infected with 20,000 P. berghei

    ANKA sporozoites, and allowed to share medium with upper chambers

    containing serum(CTRL),noninfectedblood (NI)orblood containing6x

    105P.bergheiNK65iRBCs(PbNK).After36hofcoculture,hepatomacells

    werecollectedandtreatedforFACSanalysisandprimaryhepatocyteswere

    fixed in4%paraformaldehydefor10minutes,washedthreetimes inPBS,

    andlaterpermeabilizedandblocked1hin0.3%TritonX100(Calbiochem)

    and 1% Bovine Serum Albumin (Sigma) to avoid nonspecific reactivity.

    Coverslipswere then incubated1hat4C in the same solution containing

    antiGFP IgG Alexa flour 488 congugate antibody (Invitrogen), Alexa 594

    phaloidin (Invitrogen) and DAPI (Sigma). After mounting coverslips on

    slides,EEFswerecountedusingtheLeicaDM5000BWidefieldFluorescence

    Microscope, and the sizes of 20 randomly chosen EEFs of each coverslip

    were measured using a Zeiss LSM 510 META Point Scanning Confocal

    Microscope. All immunofluorescencestained images were analysed using

    theImageJ1.42bsoftware.

    Fluorescence activated cell sorting (FACS) analysis of sporozoite

    infection.FACSanalysisofsporozoiteinfectedHuh7hepatomacellcultures

    at36hpostsporozoiteadditionwasperformedtodeterminethepercentage

    ofparasitecontainingcellsandparasiteGFPintensitywithininfectedcells.

    CellsamplesforFACSanalysiswereprocessedaspreviouslydescribed195.

    Transcription profiling. Total RNA from livers of nave mice (control

    group), mice infected with 106 P. berghei NK65 iRBCs for 7 days (Blood

    stagegroup,BS),miceinfectedwith5x104P.bergheiANKAGFPsporozoites

    for40h(Liverstagegroup,LS),andmiceinfectedwith106P.bergheiNK65

    iRBCs for 7 days then reinfected with 5x104 P. berghei ANKAGFP

    sporozoitesfor40h(Reinfectiongroup,Reinf)wasextractedusingRNeasy

  • materials & methods

    38

    Mini kit (Qiagen), following the manufacturers instructions. Three

    biological replicates were analysed for each group. RNA quality and

    quantitywasassessedonaBioanalysernanochip(Agilent).TotalRNAwas

    reverse transcribed and endlabelled as cRNA using the GeneChip WT

    Amplified DoubleStranded cDNA Synthesis Kit and the GeneChip WT

    TerminalLabelingKitasrecommendedbyAffymetrix.Hybridisation toan

    AffymetrixMouseGene1.0STArraywascarriedoutat45Cfor16hunder

    rotation (60 rpm). Arrays were washed on an Affymetrix FS450 and

    scanned using an Affymetrix Genechip Scanner 3000 7G. Fluorescence

    intensities were background adjusted, quantile normalised and median

    polished into expression values using the robust multiarray averaging

    programRMA196inR/Bioconductorsuite197(softwarepackage:affy198)To

    assess significance and differential expression (DE), each infected group

    wascomparedtothenoninfectedgroup,andoutof28815mainprobesets

    represented on the array, 6273 transcript clusters were classified as

    significant (Ftest, p

  • materials & methods

    39

    (ii)IFN.MiceinfectedwithP.bergheiNK65andcontrolmicereceived0.4

    mgofIFNspecificmonoclonalantibody(cloneR46A2,RatIgG1)1,2and3

    days, after inoculation with infected blood, a protocol leading IFN

    depletion at the timeof sporozoite infection, 5 dayspostP. bergheiNK65

    infection.

    (iii)TNF.MiceinfectedwithP.bergheiNK65andcontrolmicereceived0.4

    mgofTNFspecificmonoclonal antibody (cloneXT3.11,Rat IgG1)4and6

    days after inoculation of infected blood, a protocol leading to TNF

    depletion at the timeof sporozoite infection, 5 dayspostP. bergheiNK65

    infection.

    NNitroLarginine methyl ester hydrochloride treatment. Mice

    infectedwithP. bergheiNK65 and controlmice received50mg/kgofN

    NitroLargininemethylesterhydrochloride(LNAME,Sigma)3,4,5and6

    daysafterinoculationofinfectedblood,aprotocolleadingtoNOSinhibition

    atthetimeofsporozoiteinfection,7dayspostP.bergheiNK65infection.

    Smethylisothioureatreatment.Mice infectedwithP.bergheiNK65and

    controlmicereceived100gofSmethylisothiourea(SMT,Sigma)3,4,5

    and 6 days after inoculation of infected blood, a protocol leading to iNOS

    inhibitionatthetimeofsporozoite infection,7dayspostP.bergheiNK65

    infection.

    C3a receptor antagonist treatment. C5a deficient (B10.D2Hc0.H2d.H2

    T18c/oSnJ)naveorP.bergheiNK65infectedmicereceived0.5mgofC3a

    receptor antagonist (SB290157, Calbiochem) 4h before sporozoite re

    infection,5dayspostP.bergheiNK65infection.

    Kupffer cell depletion using clodronate liposomes 201,202.Kupffer cells

    wereeliminatedfromnaveandP.bergheiNK65infectedmicebyinjection

    of200lclodronateliposomes(liposomeencapsulateddichloromethylene

  • materials & methods

    40

    diphosphonate;clodLip)daily,fromdayoneafterP.bergheiNK65infection

    untilthedayofsporozoiteinfection(4dayspostbloodstageinfection).As

    controls, the same volume of PBS containing liposomeswas injected into

    naveorP. bergheiNK65 infectedmice. Clodronatewas a gift fromRoche

    Diagnostics GmbH (Mannheim, Germany). It was encapsulated into

    liposomes,aspreviouslydescribed203.Kupffercelldepletionwasverifiedby

    immunofluorescencemicroscopy.Liverswerecollectedand50msections

    obtained and fixed, as described above. Sections were then incubated

    overnight at 4C in a solution containing rat antimouse pan tissue fixed

    macrophages antibody (Fitzgerald) and Alexa 660 phaloidin (Invitrogen).

    After washing twice with PBS, sections were incubated for 6h with

    secondary Alexa fluor 595 donkey antirat IgG (Invitrogen) and DAPI

    (Sigma).Aftermountingsectionsonslides,sectionswereimagedonaZeiss

    LSM 510 META Point Scanning Confocal Microscope. Images of

    immunofluorescencestainedsectionswereanalysedusingtheImageJ1.42b

    software.

    Splenectomy. Intraperitoneal injection of 250 l of anaesthesia mixture

    (80mg/kgKetamineand10mg/kgXylazine)wasadministeredtomice10

    to 15 min before surgery. After observing the animals for response to

    anaesthesia,theanimalswerelaidontheirrightside;andthefurremoved

    with a razor. A 1.5 to 2 cm long skin incision was made in the left

    hypochondrium, the connective tissue under the skin was loosened and

    another incision in the peritoneal wall was made. Gently the spleen was

    pulledontotheexteriorsurfaceoftheperitoneum.Theartery,attachedto

    thehilumofthespleen,closetothestomachwastiedoffwithasingleknot

    using 30 plain CATGUT suture. The efferent venule attached at the other

    end of the spleen was tied off similarly by a single knot using 30 plain

    CATGUT.Cuttingawaythemesenteryandconnectivetissue,thespleenwas

    removed. Peritoneal wall and underlying muscles were closed with plain

  • materials & methods

    41

    CATGUT (30) and the skin incisions closed with wound clips. Mice with

    accessorial spleen were excluded from the study. The control group

    underwent a sham surgery and they were maintained in the same

    conditions.

    Liver hepcidin (hamp1) quantification.After RNA extraction and cDNA

    conversionasdescribedabove,hepcidin(hamp1)mRNAexpression in the

    liverwasdeterminedbyTaqManqRTPCRusingTaqManGeneExpression

    MasterMix(AppliedBiosystems)andAppliedBiosystemsinventoriedgene

    expression assays for hamp1 (ABI TaqMan Gene Expression Assay

    probe/primer set Mm00519025_m1) and gapdh (Glyceraldehyde3

    phosphate dehydrogenase) (ABI TaqMan Gene Expression Assay

    probe/primer set Mm99999915_g1), according to the manufacturers

    instructions. qRTPCR was carried out using an ABI Prism 7500 FAST

    system (Applied Biosystems). Changes in hepcidin mRNA expression

    betweencontrolandreinfectedmouse liverswerecalculatedusing the2

    DDCtmethodwithgapdhastheendogenouscontrolgene.

    Ferritinstaining.Liverswerecollectedand50msectionswereobtained

    andfixedasdescribedabove.Sectionswerethenincubatedovernightat4C

    in a solution containing rabbit antimouse heavy and light ferritin chain

    antibody(akindgiftbyPaoloArosio,FacultyofMedicine,Brescia,Italy)and

    Alexa 660 phaloidin (Invitrogen). After washing twice with PBS, sections

    were incubatedfor6hwithsecondaryAlexafluor595donkeyantirat IgG

    (Invitrogen) andDAPI (Sigma). Sectionswere imagedonaZeissLSM510

    METAPointScanningConfocalMicroscope.Imagesofimmunofluorescence

    stainedsectionswereanalysedusingtheImageJ1.42bsoftware.

    Totalnonhemeironquantification.Totalnonhemeironwasmeasured

    as described previously 204, in brief after drying samples (spleens and

    primaryhepatocytes) for48hat45C, tissueswereweighed,anddigested

  • materials & methods

    42

    for 48h in 10% trichloroacetic acid/10% HCl at 65C. Two hundred

    microlitersof the extractwere thenadded to1mlof chromogen solution

    (0.01%bathophenanthrolinedisulfonic acid, 0.1% thioglycolic acid, 2.5M

    sodium acetate) and after 15 minutes of incubation, absorbance was

    measured at 535 nm. A standard curve performed for known iron

    concentrationsbetween10and500g/mlrevealedalinearityofresponse

    with a slope of ~1. Sampleswere diluted appropriately to fallwithin the

    linearrange.Valueswereexpressedas%ofcontrol,inmicrogramsofiron

    pergramofdryweight.

    HAMPadenovirus in vivo infection.Micewere infectedwith adenoviral

    5x1011 pu of pAd.CMV.GFP (control) or pAd.CMV.Hamp.ires.GFP.Wpre

    (HAMP.adV) kindlyprovidedby StefanoRivella for (Weill CornellMedical

    College,USA)48hprior tosporozoite infection,hepcidinexpressionwas2

    fold upregulated by the Hamp.adv compared to uninfected and GFP.adv

    control groups. All mice were then infected with 2x104 P. berghei ANKA

    (1.49L)sporozoites,and40h later liverswerecollectedand liver infection

    wasdeterminedbyexpressionofPbA18SrRNA.

    Iron treatments. (i) In vitro: Ferric Ammonium Citrate (FAC, Sigma)

    bathophenanthrolinedisulfonate (BPS, Acros) and Desferrioxamine (DFO,

    Sigma)solutionswerepreparedinat thedesignatedconcentrations.Huh7

    cells were incubated with different concentrations of FAC, BPS or DFO,

    using water as control for FAC and PBS as control for BPS and DFO

    respectively, 24h prior to sporozoite infection and for 36h postinfection,

    afterwhichcellswerecollectedforFACSanalysis.

    (ii) In vivo:DFO (Sigma) solutionwas prepared in PBS at the designated

    concentration.Micewere inoculated intraperitonealywith 250mg/Kg of

    DFOusingPBSascontrol24hpriortosporozoiteinfection.

  • materials & methods

    43

    Tuneldetectionofapoptosis.Livertissueswereharvestedfromwildtype

    andcaspase3deficientmiceinfectedwithP.bergheiNK65for8daysalong

    with noninfected wild type mice. Tissues were frozen in OCT freezing

    medium and then sliced into 16msections using a cryostat. Analysis of

    D