• Nenhum resultado encontrado

Inhibitory receptor MHC I molecule Activating ligand Healthy cell

N/A
N/A
Protected

Academic year: 2019

Share "Inhibitory receptor MHC I molecule Activating ligand Healthy cell"

Copied!
9
0
0

Texto

(1)

ww w . e l s e v i e r . c o m / l o c a t e / b j i d

The

Brazilian

Journal

of

INFECTIOUS

DISEASES

Review

article

Natural

killer

cells

in

hepatitis

B

virus

infection

Shao-fei

Wu

a,1

,

Wen-jing

Wang

b,1

,

Yue-qiu

Gao

a,∗

aDepartmentofHepatopathy,ShuguangHospitalAffiliatedtoShanghaiUniversityofTraditionalChineseMedicine,Shanghai,China bDepartmentofGynecology,ShuguangHospitalAffiliatedtoShanghaiUniversityofTraditionalChineseMedicine,Shanghai,China

a

r

t

i

c

l

e

i

n

f

o

Articlehistory:

Received2February2015

Accepted5May2015

Availableonline25June2015

Keywords:

NKcells

HBV Review

a

b

s

t

r

a

c

t

Naturalkillercellsareauniquetypeoflymphocyteswithcytotoxiccapacity,andplay

impor-tantrolesagainsttumorsandinfections.Recently,naturalkillercellshavebeenincreasingly

valuedintheireffectsinhepatitisBvirusinfection.SincehepatitisBvirusisnotcytopathic,

thesubsequentantiviralimmuneresponsesofthehostareresponsibleforsustainingthe

liverinjury,whichmayresultincirrhosisandevenhepatocellularcarcinoma.Many

stud-ieshaveconfirmedthatnaturalkillercellsparticipateinanti-hepatitisBvirusresponses

bothintheearlyphaseafterinfectionandinthechronicphaseviacytolysis,degranulation,

andcytokinesecretion.However,naturalkillercellsplaydichotomicroles:theyexert

antivi-ralandimmunoregulatoryfunctionswhilstcontributetothepathogenesisofliverinjury.

Here,wereviewtherolesofnaturalkillercellsinhepatitisBvirusinfection,introducing

noveltherapeuticstrategiesforcontrollinghepatitisBvirusinfectionviathemodulationof

naturalkillercells.

©2015ElsevierEditoraLtda.Allrightsreserved.

Introduction

HepatitisBvirus(HBV)infectionisoneofthemostcommon

chronicviralinfectionsinhumans.HBVisnotcytopathic,i.e.

whencellularimmuneresponsesaredeficientor

pharmaco-logicallysuppressed,HBVcanreplicateathighlevelsinvivo

withoutdetectablepathologicalconsequences.1,2Ithasbeen

shownthatliverinjuryisinfactduetotheantiviralimmune

response ofthe host. In chronic HBV infection, prolonged

progressmaycausepersistentliverinflammation,eventually

resultingincirrhosisandhepatocellularcarcinoma(HCC).3

Itiswellknownthatvirus-specificCD8+ cytotoxicT

lym-phocytesandCD4+T-helpercellsplayeffectiveandregulatory

Correspondingauthor.

E-mailaddress:gaoyueqiu@hotmail.com(YQ.Gao).

1 Theseauthorscontributedequallytothisstudyandsharethefirstauthorship.

roles inanti-HBV immunity.4 However, the roles ofinnate

immunityhavetriggeredextensivedebates.Innateimmunity

isconsideredtocomposethefirst-linehostdefenseagainst

pathogens,includingsensingdangersignals,controllingviral

replication and disseminationvery early afterinfection,as

well as for timely orchestration of virus-specific adaptive

responses.5However,ithasbeenreportedthatHBVdoesnot

modulatehostcellulargenetranscription,andwould

there-foreinduceneitherinnateantiviralresponseinhepatocytes

nor intrahepatic innate immune responses.6 Furthermore,

a portion of chronically infected patients undergo a

long-lasting immunetolerance phasethat is characterizedbya

lack ofclinical symptoms and high HBV load.7 Hence, the

innateimmunityisdeemedtoaccountforthe“absence”of

http://dx.doi.org/10.1016/j.bjid.2015.05.006

(2)

anti-HBVresponse.Nevertheless,somefindingsare

inconsis-tentwiththeseconclusions,suggestinganactiveroleofinnate

immunityinHBVclearance.Natural killer(NK)cellsare an

essentialpartoftheinnateimmunesystem,andparticipate

intheantiviralresponsesdespitethedichotomic

character-isticstheydisplay.8 Here,wewillreviewtherecentstudies

regardingalteredNKcellsphenotypesandfunctionsinHBV

infection.

General

features

of

NK

cells

HumanNKcellsaregenerallydefinedasagroupof

lympho-cytesthatexpressCD56butlacktheTcellreceptor(TCR)–CD3

complex.They representabout 15% ofall peripheralblood

lymphocytesandthisproportioncanrisetomorethan30%

in the liver.9 NK cells exert their effects mainly through

therecognitionandkillingoftargetcells andthe secretion

ofcytokinessuch asinterferon-␥(IFN-␥)and tumor

necro-sisfactor-␣ (TNF-␣), whichcanmodulate antiviral immune

responses.Byproducingantiviralcytokinesandchemokines,

NKcellsalsoplayanimportantroleinbridgingthe innate

andadaptive immuneresponses.10 NKcellscan befurther

divided into two subsets, the CD56bright and the CD56dim

subsets, according to the cell membrane density of CD56.

CD56dim NKcells,expressinghighlevels ofthe low-affinity

Fc␥-receptorCD16andthekillerimmunoglobulin-like

recep-tor(KIR),representsabout 90%ofallbloodNKcells,whilst

CD56bright NK cells constitute less than 10% of all blood

NK cells and do not express CD16and KIR.11 Referring to

functions,CD56dimNKcellsefficientlykilltargetcellsby

degra-nulationbutsecretelowlevelsofcytokines;ontheotherhand,

CD56brightNKcellsproducealargeamountofcytokinesupon

stimulationbut areless cytotoxic.However,CD56bright cells

exhibitsimilarorenhancedcytotoxicityagainsttargetcells

afterprolongedactivationcomparedwithCD56dimcells.12In

addition,CD56brightNKcellsconstitutivelyexpressthe

high-andintermediate-affinityinterleukin-2receptors(IL-2R)and

expandinvitroandinvivoinresponsetolowdosesofIL-2.9,13,14

Onthe contrary,restingCD56dim NKcellsonlyexpressthe

intermediate-affinityIL-2Randproliferateweaklyinresponse

tohighdosesofIL-2invitro,evenafterinductionofthe

high-affinityIL-2R.9,11,13

NKcellsexpressanarrayofreceptorsincludinginhibitory

andactivatingreceptors,inducingnegativeandpositive

sig-nalswhencombinedwiththeirligands,whichcandetermine

whether or notNK cells become activated, enabling them

todetect infectedor neoplastic cells whilesparing normal

cells.15MHCclassImolecules,encodedbyhighlyconserved

genesinheritedindependently andexpressedbymost

nor-malcells,arenaturalligandsforNKcellinhibitoryreceptors.

Thus,thestrengthoftheactivatingsignalsonencountering

normalcellsisdampenedbyinhibitorysignals,andNKcells

areleftquiescent.However,MHCclassImoleculesareoften

down-regulatedoncancerousorvirus-infectedcells,

deliver-ing insufficientinhibitory signals, thus activatingNK cells.

Thisisknown asthe “missing-self” mechanism ofNKcell

activation.Inanothercondition,NKcellsareselectively

acti-vatedby‘stressed’cells,whichexpressupregulatedactivating

ligands for NK cells and thereby overcome the inhibitory

signalingdeliveredbyMHCclassImolecules.Thisisknown

asthe“stress-inducedself”triggeringofNKcellactivation16,17

(Fig.1).Inbothconditions,NKcellactivationleadsto

degranu-lationand/orcytokinesecretion.Inaddition,NKcellscanbe

activateduponstimulationbyIL-12,IL-15,IL-18andIFN-␣/␤.14

NK

cells

in

the

early

immune

responses

to

HBV

infection

Innateimmunityisthefirstbarrieragainstinfectionand

neo-plasticgrowth,andisdistinguishedbytherapidparticipation

in the earlyresponse toantigenswithout presensitization.

NKcells,asanimportantelementoftheinnateimmunity,

display at least three effector functions that contribute to

infectioncontrol:theycandirectlykillinfectedcellsby

releas-ingcytolyticgranules,induceapoptosisoftargetcellsthrough

crosslinkingdeathreceptorsandligands,andproducea

vari-ety of immunomodulative cytokines (Fig. 2). Studies have

confirmedthatNKcellstakepartintheantiviralresponses

tocertainRNAviruses(e.g. HIV,HCV)andDNAviruses(e.g.

CMV).18–20However,HBViscurrentlyconsideredasa“stealth”

virus anditsinfection ischaracterizedbyalackofobvious

symptomsandliverdamagethatevenlastsforseveralyears.

Onthisaccount,patientsattheveryearlystageofinfection

are almost unrecognizable.On the other hand,only

chim-panzeeandtupaiacanbeinfectednaturallybyHBV,providing

insufficientanimalmodelsforitsstudy.Althoughwoodchuck

infectedwiththewoodchuckhepatitisvirus (WHV)offersa

favorablemodel,21whetheritisanentirelyrepresentativeof

thehumansettingisyettobediscussed.Mousemodelsof

per-sistentHBVinfectionsthatareanalogoustothoseofhuman

chronicHBV infectionshavebeen established,22,23 butwith

the sameproblemsaswiththe woodchuckmodel.Todate,

takingintoconsiderationthesefactors,ourcognitionofthe

anti-HBVinnateimmuneresponsesattheearliest

presymp-tomaticstagesisstilllimited.

SomestudiessuggestthatNKcellsandeveninnate

immu-nity donotsignificantly contributetothe initialcontrolof

HBVinfection.Studiesinchimpanzeesandwoodchuckshave

shownthatHBVdoesnotinducesignificantchangesin

intra-hepaticgeneexpressionduringentryandreproductioninthe

liver,andtheviralloaddoesnotdecreaseuntiltheonsetofthe

adaptiveimmuneresponse,severalweekslater.24,25Withthe

developmentofinfection,theclearanceofHBV-infected

hepa-tocytesbyadaptiveimmunitycorrelateswithelevatedlevels

ofIFN-␥andTNF-␣intheliver,2whichcouldbeproducedby

activatedNKcells.However,follow-upexperimentsindicate

thatratherthanNKcells,CD8+cells,whicharealsosources

ofIFN-␥andTNF-␣,arethemaineffectorcellsresponsiblefor

viralclearance.26

However, some other experiments showed

contradic-tory results. NK cells express mRNAs coding for toll-like

receptors(TLR),TLR1toTLR9,whichrecognizecertain

non-self molecules and initiate immune responses.27 In vitro,

HBV-plasmid DNA promotes NK cell activation including

cytotoxicity and IFN-␥ production in the liver through

TLR/IFN-␣-mediatedsignalingpathways.28Inastudycarried

out ina high-titer HBV replication mouse model, NK cells

wererequiredtoeliminateHBVinfection,presumablythrough

(3)

NK cell

Inhibitory receptor

MHC I molecule

Activating ligand

Healthy cell

a

b

c

Unactivated Missing self Stress-induced self Stressed cell Stressed cell Activating

receptor

+

+

+

Fig.1–RecognitionofhealthyandstressedcellsbyNKcells.(a)MHCImoleculesareexpressedbymostnormalcells,which transmitinhibitorysignals.Thestrengthofinhibitorysignalsovercomesactivatingsignals,andNKcellsareleft

unactivated.(b)MHCImoleculesareoftendown-regulatedonstressedcells,offeringinsufficientinhibitorysignals,thus activatingNKcells.Thisisthe“missing-self”mechanismofNKcellactivation.(c)Somestressedcellsup-regulateactivating ligandsratherthandown-regulateMHCImolecules,whichdeliveroverwhelmingactivatingsignalsandactivatetheNK cells.Thisisthe“stress-inducedself”mechanismofNKcellactivation.

afterinfectionwithhigh titersofWHV,NKcellswere

acti-vated and virus load was reduced significantly, suggesting

thattheinnateresponseisactivatedintheliversoonafter

exposuretoacertaindoseofWHV.29 Owingtothedelayed

appearanceofsymptomsorthetotallyasymptomaticnature

ofHBVinfection,studiesoftheearlyimmuneeventsin

HBV-infectedpeopleare rare. Astudy oftwoblood donorswho

wereidentified byaccidenttohaveseroconvertedwiththe

appearanceofhepatitisBsurfaceantigen(HBsAg)and

sub-sequentriseofHBVviremiawithoutalanineaminotransferse

(ALT)elevation,haveshownthatNKcellsareabletomount

anearlyand efficientresponsetoHBV,andthatthe innate

immunesystemisabletorecognizeHBVfromthebeginningof

theinfection,whichprobablycontributestothetimely

induc-tionofadaptiveresponses.30InacutehepatitisB,activation

receptor-expressingNKcellsarepreferentiallyenriched,while

the inhibitory receptor-expressing NK cells are reduced.31

Moreover, according to two independent studies, NK cells

displayedincreasedordecreasedcytolyticactivityandIFN-␥

production.31,32TheseresultsrevealthatNKcellsparticipate

intheearly-stageresponseafterHBVinfection.

However,mostofthesestudieswerecarriedoutinanimal

models,whichmaynotfullyrepresenttherealityinhumans.

Ontheotherhand,datafromhumanbeingsareinsufficient.

Hence,theroleofNKcellsintheearlyphaseofHBVinfection

isstilltobedemonstratedinhumans.

Contribution

of

NK

cells

to

HBV-induced

liver

injury

InHBVinfection,NKcellsalwaysplaycontradictoryroles,as

theyexertantiviralandimmunoregulatoryfunctionswhilst

contributetothepathogenesisofliverinjury.Theapoptosis

ofHBV-infectedhepatocytesisinfavorofself-protectionasit

helpstoviralclearance.Butinthecaseofsustainedinfection,

thecontinuousinjurytotheliveristhepreconditionforliver

fibrosis andHCC.Inthe immune-activatedstageofchronic

hepatitisB(CHB),NKcellsskewtowardacytolyticactivity,

whichcorrelatespositivelywiththeseverityofliverdamage.33

EnhancedcytolyticactivityofNKcellscanbepartlyattributed

toalteredlevelsofcertaincytokines33andtotherecognition

betweenNKG2Danditsligands.34

Other than cytotoxic effectors-mediated liver damage,

apoptotic signal transmission is also an important factor

(4)

Signaling receptors/ ligands

Perforin/ granzyme

Cytokine receptors

Cytokine secretiors

Cytokines

NK cell

Cytolysis Intercellular

signaling

Fig.2–NKcellsmediatetheirfunctionsthroughatleastthreemechanisms:releasingperforin/granzymeforcytolysis, deliveringintercellularsignalingthroughreceptor–ligandcrosslinking,andsecretingcytokines.

littleornoTNF-relatedapoptosis-inducingligand(TRAIL)on

theirsurface35andhepatocytesexpressminimalTRAIL

death-inducing receptors.36 However, in patients with HBV and

liverinflammation,TRAIL-expressingNK cellsare enriched

in the liver and the hepatocytes express upregulated

lev-elsoftheTRAILdeath-inducingreceptor,indicatingthatNK

cellsmaycontributetoliverinflammationbyTRAIL-mediated

death of hepatocytes.37 Besides, after being activated, NK

cells may induce massive HBV-infected hepatocyte

degen-eration through the Fas/Fas ligand interaction,38 and are

even involved in the disease progression of HBV-related

acute-on-chronicliverfailure(ACLF).39Intrahepatic

PD-1/PD-L1up-regulationiscloselyassociatedwiththeviralloadand

inflammatory responses, which decreases simultaneously

withinflammationremission,implyingthatthePD-1/PD-L1

systemparticipatesinliverinjury.40,41AlthoughNKcell

dys-functionisreportedtocorrelate withPD-1up-regulation,42

directevidencesarelackingtocertifythatPD-1altersNKcell

functioninHBVinfection.

NK

cells

in

chronic

HBV

infection

ThecontroversialroleofNKcellsintheacutephaseofHBV

infectionhasbeenunderlinedabove.Inchronicinfection,NK

cellsdisplayvaryingchangesinproportion,phenotypeand/or

function,whichdifferbetweenstudies.ThedefectsinNKcells

arereflectedinmanyaspects:(1)thepercentagesofhepatic

and peripheral NK cells are reduced in immune-activated

CHBpatients,withorwithoutchangesintheirsubsets33,43,44;

(2) altered expression of activating or inhibitory receptors

onNKcells33,43–46;(3)up-regulationofsomemoleculeswith

inhibitoryeffectssuchasTcellimmunoglobulinandmucin

domain containing molecule-3 (Tim-3);47 (4) maintained or

evenenhancedcytolyticactivity,whichiscorrelated tothe

severity of liver injury33,43,46; and (5) impaired capacity to

produce cytokines such as IFN-␥ and TNF-␣.43,44,46,47 Viral

loadreductionthroughanti-viraltherapypartlyrestoresNK

cells numbers, cytokine production and inhibitory

recep-tor expression.43,45,48,49 Although these studies are mostly

consistent, there are discordancesin somedetails suchas

thechanges intheexpressionofsomereceptors33,43–46 and

changesincytolyticactivity.Thereasonsforthesedifferences

are notclearyet,but maybeascribed toethnicvariations,

samplesize,diseaseprocessandseverity.Despiteinconsistent

conclusionsbetweenstudies,NKcellsdisplayintactoreven

increasedcytotoxicitybutimpairedanti-viralcytokines

secre-tionduringchronicHBVinfection,andthesechangesmaybe

partlyrectifiedafterviralloadreduction.

The factors contributing to NK cell dysfunction were

demonstratedbymanystudies.NKcellsexpressmRNAsforall

(5)

HBV-specific CD8+ T cell

TRAIL receptor TRAIL Perforin

NKG2D/2B4

mDC

pDC

IFN-α↓ OX40L ↑

Activ ating cytokine

↓ Inhibitor

y

cytokine

NKG2D/2B4 ligand

NK cell

Dysfunction

Macrophage/

Kupffer

Fig.3–mDCs,pDCsandmacrophagescontributetoNKcelldysfunctionthroughalteredcytokinesecretionandsignaling receptorexpression.DisorderedNKcellssubsequentlymediateCD8+Tcellsdeath.

stimulatedtoinducetheproductionofIFN-␥.27Arecentstudy

revealedthat NKcells canbeactivated byHBV-based DNA

plasmidsinaTLR-dependentmanner,displaying enhanced

NKcelleffectorfunctionsincludingcellularcytotoxicityand

IFN-␥production.26However,inthepersistentHBVinfection

phase,NKcellssufferfunctionaldisordersthatcanberestored

withviralreplicationsuppression.Itcanbededucedthat

dur-inglong-lastingvirusreplication,NKcellsareinfluencedby

pathologicalcircumstancesinducedbytheHBVinfection.

NKcellsinteractwithsomeotherimmunecellsthrough

receptor–ligandcombinationand/orregionalcytokine

secre-tion.Dendriticcells (DCs)are the mostimportant

antigen-presentingcells,andaredividedintomyeloiddendriticcells

(mDC) and plasmacytoid dendritic cell (pDC). DCs bridge

theinnateandadaptiveimmunities,andconsequentlyplay

pivotal roles in eliminating external antigens. During HBV

infection,bothmDCsandpDCsdisplayfunctionaldefects,50–52

whichinfluencethecrosstalkwithNKcells,making

ineffi-cienttheNKcellsactivation.mDCsaresubstantiallyimpaired

intheirabilitytoactivateNKcellsviadecreasedmDC-derived

cytokine secretion, which in turn fail to secrete adequate

amountsofIFN-␥.52pDCsarepoorNKactivators,potentially

throughanOX40L/IFN-␥-dependentpathway53,54(Fig.3).

BesidesDCs,intrahepaticKupffercellsalsoparticipatein

the anti-HBVimmune response, partlythrough interfering

withthe NKcells.Kupffercellsare themainsourceofthe

immune-suppressive cytokine interleukin-10 (IL-10), which

maintainsthehumoralimmunetoleranceduringpersistent

HBV infection55 (Fig.3).IL-10actsasaninhibitorycytokine

blunting NK activation, which restrains IFN-␥ secretion.56

However,anotherstudy demonstratedthat IL-10had a

sig-nificant effect on NK cytotoxicity rather than on IFN-␥ or

TNF-␣productionorNKcellactivatoryreceptorexpression.57

BlockingtheIL-10pathwaymaycorrectdefectivecapacityof

NK cells toproduceIFN-␥.58 Furthermore,plasma levelsof

transforminggrowthfactor-␤1(TGF-␤1)isalsoelevatedduring

HBV infection,59 which mayimpair NKcell-mediated

cyto-toxiccapacityandIFN-␥productionbydown-regulatingthe

expressionoftheactivatingreceptorsNKG2Dand2B4.

More-over,theirintracellularadaptorproteins,respectivelyDAP10

andSAP,arealsodecreased.60

Since the immunesystem is areticular and syntrophic

entirety,NKcelldysfunctionalsoinfluencesthefunctionof

the other parts of the system, especially effector T cells.

CD8+ T cells are the main effector cells responsible for

(6)

infection.25However,inthechronicinfectionstage,theyare

markedlydiminishedinnumberandexhaustiveinfunctions

inpatients who failed tocontrol virus replication.61,62 The

decreaseinCD8+ effectorTcellscanbepartlyattributedto

NKcells,becauseNKcellscannotonlymediateCD8+Tcells

deaththrough up-regulationof TRAILreceptors,63 but also

limitCD8+ TcellsimmunitybyNKG2D-and2B4-dependent

perforin-mediatedlysis64,65(Fig.3).

Unfortunately,therestorationofHBV-specificCD8+Tcells

isdifficulttoachieve.Evenifafavorablevirologicalresponseis

obtainedafteranti-viraltreatment,HBV-specificCD8+Tcells

remainatlowlevelsandexpressanexhaustedphenotype.66,67

This“permanentsignature”mayresultfromchronicexposure

toHBV-relatedantigens,whichinturnchangesthe

differen-tiationprogramofCD8+ Tcells.Unlikewhatisobservedin

HBVinfection,typeIinterferoncanprotectanti-viralCD8+T

cellsfromeliminationbyNKcell-mediatedperforin

expres-sioninlymphocyticchoriomeningitisvirusinfection.68These

diverse mechanisms may partly explain whyCHB is

clini-callydifficulttomanage,withanunsatisfyingresponserate

toIFNtreatment.Furthermore,althoughnotyetobservedin

HBV infection,NK cells may preventactivatedCD4+Tcells

frombeingkilledbytheinteractionbetweenNKG2Aandits

ligand.69

NKcellsmaynotbeasimportantasDCsinbridgingthe

innateandadaptive immunities,buttheirrolesareunique,

andcorrelationalstudiesareattractingmoreandmore

atten-tioninHBVinfection.

NK

cells

as

potential

targets

for

immunotherapy

Intheclinicalsetting,currentlyavailableantiviraltreatment

forCHB infection can bedivided into two classes of

ther-apeutic agents:nucleos(t)ide analogs (NAs)and IFN-␣. NAs

includenucleoside(lamivudine,telbivudineandentecavir)or

nucleotide(adefovirandtenofovir)analogs.Themajor

advan-tagesofNAsaregoodtoleranceandpotentantiviralactivity

associatedwithhighratesofon-treatmentresponseto

ther-apy.TheadvantagesofIFNincludeafinitecourseoftreatment,

absenceofdrugresistance,andanopportunitytoachievea

durableresponsetotherapy.Obviously,theadvantagesofone

agentare the disadvantages ofthe other. Besides, the two

agentshavebeenconfirmedtocontributelittletothe

restora-tionofHBV-specificCD8+Tcells.Sincecurrenttherapiesare

farfromsufficientfromeconomicalandeffectivenesspoints

ofview,noveltherapeuticstrategiesare ingreatneed.Itis

unclearwhyindividualsresponddifferentlytoIFN,70butitis

supposedtobedue,atleastinpart,toNKcell

characteris-ticsincludingcellnumbers,receptorexpressionandfunction

alteration.67,71Similarresultswerealsoobtainedwiththeuse

ofNAs,43,45indicatingthatthemodulationofNKcellfunction

mayhelptosuppressHBVreplication.AberrantDNA

methyla-tionisanearlyandubiquitouseventduringHCCdevelopment,

andcanbedetectedeveninprecancerouslivertissuessuch

aschronichepatitis,livercirrhosis, ordysplasticnodules.72

Arecentstudyconfirmedthatthisprocesswasclosely

asso-ciatedwith NKcell activity.73 Hence,regulation ofNK cell

functionisapotentialwaytotreatCHBandtopreventthe

occurrenceofcirrhosisandHCC.

IthasbeendiscussedabovethatNKcellfunctional

alter-ations include at least three aspects: decreased cytokine

secretion, increased cytotoxicity and apoptosis-mediating

capacity(Fig.3).Therefore,weshouldturntoincreasing

anti-viralcytokineproductionandprotectothercellsfrombeing

killed,targetingontheinteractionbetweenNKcells’

recep-torstotheirligandsandcytokinesinthemicroenvironment.

Astheimmunesystemisacomplexentity,theinfluenceon

everypartofthissystemshouldbeconsideredtoformulate

newstrategiesinordertoavoidimmuneoverreaction.

StrategiesbasedontheregulationofNKcell functionto

treatCHBwereonlycarriedoutinanimalexperimentssofar.

NKcellsmayachieveenhancedanti-viralviabilityor

allevi-atedhepatocytelysisbyblockadeofsomeoftheiractivating

orinhibitoryreceptorsandligandinteractions.45,74Increased

expressionofPD-1 inthe liverresultsinimmunedisorder,

which istothedisadvantageofHBV clearance.PD-1/PD-L1

blockadecouldreverseimmunedysfunctionbyaugmenting

IFN-␥ secretionand accelerating HBV elimination invivo.75

IL-10andTGF-␤1havebeenshowntorestrainNKcells

func-tion,andblockadeofIL-10and/orTGF-␤1restoredthecapacity

ofNKcellstoproduceIFN-␥,thereby enhancingtheir

non-cytolyticantiviralcapacity.58,60ThecontributionofIFN-and

TNF-␣inHBVclearanceisundisputed,especiallyintheacute

phase of infection.2 However, in the immunotolerant and

immunoreactivephases,elevatedlevelsofIFN-␥andTNF-␣

arenotenoughtohelpeliminatingthevirus,butallows

sus-tainingtheliverinjury.76

Concluding

remarks

ThereasonwhyHBVinfection isdifficulttomanageinthe

clinicalsettingisbecauseofthepropertiesofthevirusitself,

butalsooftheimmuneresponsesofthehosts.Asdiscussed

above,theroleofNKcellsinHBVinfectionisstillcontroversial:

whetherNKcellsisinvolvedintheearlyphaseofHBV

infec-tion,andwhatalternations havebeen occurredtoNKcells

duringchronicinfectionarenotknownyet.Whereas,itcannot

bedeniedthatthepersistenceofHBVinfectionandongoing

liverinjurycouldbepartlyduetothedysfunctionofNKcells

andsubsequentmediationofdisordersoftheimmunesystem

anddeathofhepatocytes.Onaccountofourlimited

knowl-edge,newimmunotherapiesbasingonNKcellsare onlyin

theinfancystage.AnewsubsetofhumanNKcells,

denom-inatedasNK-22cells,wasrecentlydiscovered.77Theylocate

inmucosa-associatedlymphoidtissues,andarehard-wiredto

secreteIL-22,whichprovideprotectiontohepatocytes78,79and

promoteproliferationofliverstem/progenitorcells,80

provid-inganoveltherapeuticcandidateforchronicHBVinfection.

Insummary,onthebasisofexistingachievements,more

researchesarestillneededtodefinetheexactrolesofNKcells

inHBVinfection.Newstrategiesshouldbeaimingon

melio-rating the functionof NKcells, inhibiting viral replication,

alleviatingliverinjuryandavoidingcirrhosisandHCC.

Conflicts

of

interest

(7)

Acknowledgements

This work was supported by Science Research Project of

TwelfthFive-year-Plan“AIDSandViralHepatitisMajor

Infec-tiousDiseasesPreventionandControl”,2012ZX10005004-002

and2012ZX10005010-002-003,and NationalNatural Science

FoundationofChina,81102570and81202662.

r

e

f

e

r

e

n

c

e

s

1. GuidottiLG,ChisariFV.Immunobiologyandpathogenesisof viralhepatitis.AnnuRevPathol.2006;1:23–61.

2. GuidottiLG,RochfordR,ChungJ,ShapiroM,PurcellR,Chisari FV.Viralclearancewithoutdestructionofinfectedcells duringacuteHBVinfection.Science.1999;284:825–9.

3. FerrariC,MissaleG,BoniC,UrbaniS.Immunopathogenesis ofhepatitisB.JHepatol.2003;39Suppl1:S36–42.

4. BertolettiA,FerrariC.Innateandadaptiveimmuneresponses inchronichepatitisBvirusinfections:towardsrestorationof immunecontrolofviralinfection.Gut.2012;61:1754–64.

5. MedzhitovR,JanewayCJr.Innateimmunity.NEnglJMed. 2000;343:338–44.

6. WielandS,ThimmeR,PurcellRH,ChisariFV.Genomic analysisofthehostresponsetohepatitisBvirusinfection. ProcNatlAcadSciUSA.2004;101:6669–74.

7. FattovichG,BortolottiF,DonatoF.Naturalhistoryofchronic hepatitisB:specialemphasisondiseaseprogressionand prognosticfactors.JHepatol.2008;48:335–52.

8. HanQ,ZhangC,ZhangJ,TianZ.Theroleofinnateimmunity inHBVinfection.SeminImmunopathol.2013;35:23–38.

9. DohertyDG,NorrisS,Madrigal-EstebasL,etal.Thehuman livercontainsmultiplepopulationsofNKcells,Tcells,and CD3+CD56+naturalTcellswithdistinctcytotoxicactivities andTh1,Th2,andTh0cytokinesecretionpatterns.J Immunol.1999;163:2314–21.

10.CooperMA,FehnigerTA,TurnerSC,etal.Humannatural killercells:auniqueinnateimmunoregulatoryroleforthe CD56(bright)subset.Blood.2001;97:3146–51.

11.CooperMA,FehnigerTA,CaligiuriMA.Thebiologyofhuman naturalkiller-cellsubsets.TrendsImmunol.2001;22:633–40.

12.StrowigT,BrilotF,MunzC.NoncytotoxicfunctionsofNK cells:directpathogenrestrictionandassistancetoadaptive immunity.JImmunol.2008;180:7785–91.

13.JostS,AltfeldM.Controlofhumanviralinfectionsbynatural killercells.AnnuRevImmunol.2013;31:163–94.

14.VivierE,TomaselloE,BaratinM,WalzerT,UgoliniS. Functionsofnaturalkillercells.NatImmunol.2008;9:503–10.

15.YamagiwaS,KamimuraH,IchidaT.Naturalkillercell receptorsandtheirligandsinliverdiseases.MedMol Morphol.2009;42:1–8.

16.VivierE,UgoliniS,BlaiseD,ChabannonC,BrossayL. TargetingnaturalkillercellsandnaturalkillerTcellsin cancer.NatRevImmunol.2012;12:239–52.

17.CooperMA,ColonnaM,YokoyamaWM.Hiddentalentsof naturalkillers:NKcellsininnateandadaptiveimmunity. EMBORep.2009;10:1103–10.

18.MartinMP,GaoX,LeeJH,etal.Epistaticinteractionbetween KIR3DS1andHLA-BdelaystheprogressiontoAIDS.Nat Genet.2002;31:429–34.

19.KhakooSI,ThioCL,MartinMP,etal.HLAandNKcell inhibitoryreceptorgenesinresolvinghepatitisCvirus infection.Science.2004;305:872–4.

20.OrangeJS,FassettMS,KoopmanLA,BoysonJE,StromingerJL. Viralevasionofnaturalkillercells.NatImmunol.2002;3: 1006–12.

21.FletcherSP,ChinDJ,JiY,etal.Transcriptomicanalysisofthe woodchuckmodelofchronichepatitisB.Hepatology. 2012;56:820–30.

22.YangD,LiuL,ZhuD,etal.AmousemodelforHBV immunotoleranceandimmunotherapy.CellMolImmunol. 2014;11:71–8.

23.HuangLR,WuHL,ChenPJ,ChenDS.Animmunocompetent mousemodelforthetoleranceofhumanchronichepatitisB virusinfection.ProcNatlAcadSciUSA.2006;103:17862–7.

24.FletcherSP,ChinDJ,ChengDT,etal.Identificationofan intrahepatictranscriptionalsignatureassociatedwith self-limitinginfectioninthewoodchuckmodelofhepatitisB. Hepatology.2013;57:13–22.

25.ThimmeR,WielandS,SteigerC,etal.CD8(+)Tcellsmediate viralclearanceanddiseasepathogenesisduringacute hepatitisBvirusinfection.JVirol.2003;77:68–76.

26.ZhuR,Mancini-BourgineM,ZhangXM,BayardF,DengQ, MichelML.Plasmidvector-linkedmaturationofnaturalkiller (NK)cellsiscoupledtoantigen-dependentNKcellactivation duringDNA-basedimmunizationinmice.JVirol.

2011;85:10201–12.

27.LauzonNM,MianF,MacKenzieR,AshkarAA.Thedirect effectsofToll-likereceptorligandsonhumanNKcellcytokine productionandcytotoxicity.CellImmunol.2006;241:102–12.

28.YangPL,AlthageA,ChungJ,etal.Immuneeffectorsrequired forhepatitisBvirusclearance.ProcNatlAcadSciUSA. 2010;107:798–802.

29.GuyCS,Mulrooney-CousinsPM,ChurchillND,MichalakTI. Intrahepaticexpressionofgenesaffiliatedwithinnateand adaptiveimmuneresponsesimmediatelyafterinvasionand duringacuteinfectionwithwoodchuckhepadnavirus.JVirol. 2008;82:8579–91.

30.FisicaroP,ValdattaC,BoniC,etal.Earlykineticsofinnateand adaptiveimmuneresponsesduringhepatitisBvirus

infection.Gut.2009;58:974–82.

31.ZhaoJ,LiY,JinL,etal.Naturalkillercellsarecharacterizedby theconcomitantlyincreasedinterferon-gammaand

cytotoxicityinacuteresolvedhepatitisBpatients.PLoSONE. 2012;7:e49135.

32.LunemannS,MaloneDF,HengstJ,etal.Compromised functionofnaturalkillercellsinacuteandchronicviral hepatitis.JInfectDis.2014;209:1362–73.

33.ZhangZ,ZhangS,ZouZ,etal.Hypercytolyticactivityof hepaticnaturalkillercellscorrelateswithliverinjuryin chronichepatitisBpatients.Hepatology.2011;53:73–85.

34.ChenY,WeiH,SunR,DongZ,ZhangJ,TianZ.Increased susceptibilitytoliverinjuryinhepatitisBvirustransgenic miceinvolvesNKG2D-ligandinteractionandnaturalkiller cells.Hepatology.2007;46:706–15.

35.MirandolaP,PontiC,GobbiG,etal.ActivatedhumanNKand CD8+TcellsexpressbothTNF-relatedapoptosis-inducing ligand(TRAIL)andTRAILreceptorsbutareresistantto TRAIL-mediatedcytotoxicity.Blood.2004;104:2418–24.

36.IchikawaK,LiuW,ZhaoL,etal.Tumoricidalactivityofa novelanti-humanDR5monoclonalantibodywithout hepatocytecytotoxicity.NatMed.2001;7:954–60.

37.DunnC,BrunettoM,ReynoldsG,etal.Cytokinesinduced duringchronichepatitisBvirusinfectionpromoteapathway forNKcell-mediatedliverdamage.JExpMed.

2007;204:667–80.

38.OkazakiA,HiragaN,ImamuraM,etal.Severe necroinflammatoryreactioncausedbynaturalkiller

cell-mediatedFas/Fasligandinteractionanddendriticcellsin humanhepatocytechimericmouse.Hepatology.

2012;56:555–66.

(8)

40.XieZ,ChenY,ZhaoS,etal.IntrahepaticPD-1/PD-L1 up-regulationcloselycorrelateswithinflammationandvirus replicationinpatientswithchronicHBVinfection.Immunol Invest.2009;38:624–38.

41.GermanidisG,ArgentouN,HytiroglouP,etal.LiverFOXP3 andPD1/PDL1expressionisdown-regulatedinchronicHBV hepatitisonmaintainedremissionrelatedtothedegreeof inflammation.FrontImmunol.2013;4:207.

42.WiesmayrS,WebberSA,MacedoC,etal.DecreasedNKp46 andNKG2DandelevatedPD-1areassociatedwithaltered NK-cellfunctioninpediatrictransplantpatientswithPTLD. EurJImmunol.2012;42:541–50.

43.TjwaET,vanOordGW,HegmansJP,JanssenHL,WoltmanAM. Viralloadreductionimprovesactivationandfunctionof naturalkillercellsinpatientswithchronichepatitisB.J Hepatol.2011;54:209–18.

44.LiY,WangJJ,GaoS,etal.Decreasedperipheralnaturalkiller cellsactivityintheimmuneactivatedstageofchronic hepatitisB.PLOSONE.2014;9:e86927.

45.LiF,WeiH,WeiH,etal.Blockingthenaturalkillercell inhibitoryreceptorNKG2Aincreasesactivityofhuman naturalkillercellsandclearshepatitisBvirusinfectionin mice.Gastroenterology.2013;144:392–401.

46.OlivieroB,VarchettaS,PaudiceE,etal.Naturalkillercell functionaldichotomyinchronichepatitisBandchronic hepatitisCvirusinfections.Gastroenterology.

2009;137:1151–60,e1151–7.

47.JuY,HouN,MengJ,etal.Tcellimmunoglobulin-and mucin-domain-containingmolecule-3(Tim-3)mediates naturalkillercellsuppressioninchronichepatitisB.J Hepatol.2010;52:322–9.

48.LvJ,JinQ,SunH,etal.Antiviraltreatmentaltersthe frequencyofactivatingandinhibitoryreceptor-expressing naturalkillercellsinchronichepatitisBvirusinfected patients.MediatorsInflamm.2012;2012:804043.

49.ZhangL,WangQ,ZhaoP,HuX,JiangY.Effectsofentecaviron peripheralbloodlymphocyteprofilesinchronichepatitisB patientswithsuboptimalresponsestoadefovir.ClinExp PharmacolPhysiol.2014;41:514–23.

50.vanderMolenRG,SprengersD,BindaRS,etal.Functional impairmentofmyeloidandplasmacytoiddendriticcellsof patientswithchronichepatitisB.Hepatology.2004;40: 738–46.

51.WoltmanAM,BoonstraA,JanssenHL.Dendriticcellsin chronicviralhepatitisBandC:victimsorguardianangels? Gut.2010;59:115–25.

52.TjwaET,vanOordGW,BiestaPJ,BoonstraA,JanssenHL, WoltmanAM.RestorationofTLR3-activatedmyeloid dendriticcellactivityleadstoimprovednaturalkillercell functioninchronichepatitisBvirusinfection.JVirol. 2012;86:4102–9.

53.ShiCC,TjwaET,BiestaPJ,etal.HepatitisBvirussuppresses thefunctionalinteractionbetweennaturalkillercellsand plasmacytoiddendriticcells.JViralHepat.2012;19:e26–33.

54.MartinetJ,Dufeu-DuchesneT,BruderCostaJ,etal.Altered functionsofplasmacytoiddendriticcellsandreduced cytolyticactivityofnaturalkillercellsinpatientswithchronic HBVinfection.Gastroenterology.2012;143:1586–96,e1588.

55.XuL,YinW,SunR,WeiH,TianZ.Kupffercell-derivedIL-10 playsakeyroleinmaintaininghumoralimmunetolerancein hepatitisBvirus-persistentmice.Hepatology.2014;59:443–52.

56.TuZ,BozorgzadehA,PierceRH,KurtisJ,CrispeIN,OrloffMS. TLR-dependentcrosstalkbetweenhumanKupffercellsand NKcells.JExpMed.2008;205:233–44.

57.ParkJY,LeeSH,YoonSR,etal.IL-15-inducedIL-10increases thecytolyticactivityofhumannaturalkillercells.MolCells. 2011;32:265–72.

58.PeppaD,MiccoL,JavaidA,etal.Blockadeof

immunosuppressivecytokinesrestoresNKcellantiviral

functioninchronichepatitisBvirusinfection.PLoSPathog. 2010;6:e1001227.

59.MurawakiY,NishimuraY,IkutaY,IdobeY,KitamuraY, KawasakiH.Plasmatransforminggrowthfactor-beta1 concentrationsinpatientswithchronicviralhepatitis.J GastroenterolHepatol.1998;13:680–4.

60.SunC,FuB,GaoY,etal.TGF-beta1down-regulationof NKG2D/DAP10and2B4/SAPexpressiononhumanNKcells contributestoHBVpersistence.PLoSPathog.2012; 8:e1002594.

61.MainiMK,BoniC,LeeCK,etal.Theroleofvirus-specific CD8(+)cellsinliverdamageandviralcontrolduring persistenthepatitisBvirusinfection.JExpMed. 2000;191:1269–80.

62.BoniC,FisicaroP,ValdattaC,etal.Characterizationof hepatitisBvirus(HBV)-specificT-celldysfunctioninchronic HBVinfection.JVirol.2007;81:4215–25.

63.PeppaD,GillUS,ReynoldsG,etal.Up-regulationofadeath receptorrendersantiviralTcellssusceptibletoNK cell-mediateddeletion.JExpMed.2013;210:99–114.

64.WaggonerSN,TaniguchiRT,MathewPA,KumarV,WelshRM. Absenceofmouse2B4promotesNKcell-mediatedkillingof activatedCD8+Tcells,leadingtoprolongedviralpersistence andalteredpathogenesis.JClinInvest.2010;120:1925–38.

65.LangPA,LangKS,XuHC,etal.Naturalkillercellactivation enhancesimmunepathologyandpromoteschronicinfection bylimitingCD8+T-cellimmunity.ProcNatlAcadSciUSA. 2012;109:1210–5.

66.WherryEJ,BarberDL,KaechSM,BlattmanJN,AhmedR. Antigen-independentmemoryCD8Tcellsdonotdevelop duringchronicviralinfection.ProcNatlAcadSciUSA. 2004;101:16004–9.

67.MiccoL,PeppaD,LoggiE,etal.Differentialboostingofinnate andadaptiveantiviralresponsesduring

pegylated-interferon-alphatherapyofchronichepatitisB.J Hepatol.2013;58:225–33.

68.XuHC,GrusdatM,PandyraAA,etal.TypeIinterferon protectsantiviralCD8+TcellsfromNKcellcytotoxicity. Immunity.2014;40:949–60.

69.LuL,IkizawaK,HuD,WerneckMB,WucherpfennigKW, CantorH.RegulationofactivatedCD4+TcellsbyNKcellsvia theQa-1-NKG2Ainhibitorypathway.Immunity.

2007;26:593–604.

70.BusterEH,HansenBE,LauGK,etal.Factorsthatpredict responseofpatientswithhepatitisBeantigen-positive chronichepatitisBtopeginterferon-alfa.Gastroenterology. 2009;137:2002–9.

71.MahdaviM,AmirrasouliH,AlavianSM,etal.Impactof pegylatedinterferon-alfa-2aonperforinlevelinpatientswith chronichepatitisB;preliminarystudy.HepatitisMonthly. 2013;13:e11903.

72.GaoW,KondoY,ShenL,etal.VariableDNAmethylation patternsassociatedwithprogressionofdiseasein hepatocellularcarcinomas.Carcinogenesis.2008;29: 1901–10.

73.OkamotoY,ShinjoK,ShimizuY,etal.Hepatitisvirus infectionaffectsDNAmethylationinmicewithhumanized livers.Gastroenterology.2014;146:562–72.

74.HuangM,SunR,WeiH,TianZ.Simultaneousknockdownof multipleligandsofinnatereceptorNKG2Dpreventsnatural killercell-mediatedfulminanthepatitisinmice.Hepatology. 2013;57:277–88.

75.TzengHT,TsaiHF,LiaoHJ,etal.PD-1blockagereverses immunedysfunctionandhepatitisBviralpersistenceina mouseanimalmodel.PLoSONE.2012;7:e39179.

(9)

77.CellaM,FuchsA,VermiW,etal.Ahumannaturalkillercell subsetprovidesaninnatesourceofIL-22formucosal immunity.Nature.2009;457:722–5.

78.ZenewiczLA,YancopoulosGD,ValenzuelaDM,MurphyAJ, KarowM,FlavellRA.Interleukin-22butnotinterleukin-17 providesprotectiontohepatocytesduringacuteliver inflammation.Immunity.2007;27:647–59.

79.ZhangY,CobleighMA,LianJQ,etal.Aproinflammatoryrole forinterleukin-22intheimmuneresponsetohepatitisB virus.Gastroenterology.2011;141:1897–906.

Imagem

Fig. 1 – Recognition of healthy and stressed cells by NK cells. (a) MHC I molecules are expressed by most normal cells, which transmit inhibitory signals
Fig. 2 – NK cells mediate their functions through at least three mechanisms: releasing perforin/granzyme for cytolysis, delivering intercellular signaling through receptor–ligand crosslinking, and secreting cytokines.
Fig. 3 – mDCs, pDCs and macrophages contribute to NK cell dysfunction through altered cytokine secretion and signaling receptor expression

Referências

Documentos relacionados

The needle is passed through the uterine cavity until its posterior side, where the thread is passed over the fundus until the anterior wall; there, the needle is inserted below the

its own. Thus, there are two possibilities: a) the activating receptor recognizes its ligand (not generating a signal) and the inhibitory receptor does not recognize the ligand (not

In the present study, the activation status of B cells from peripheral blood of healthy controls (N = 20) and patients with acute hepatitis B (AHB, N = 15) or chronic hepatitis

The objective of this study was to examine the relationship between the expression of B cell activating factor (BAFF) and BAFF receptor in patients with disease activity of

The inclusion criteria applied were the following: (i) study design–the RCT was performed to compare the therapeu- tic effects of ADV and ETV; (ii) study duration–patients were

Effects of eicosanoid biosynthesis inhibitors on the prophenoloxidase- activating system and microaggregation reactions in the hemolymph of Rhodnius prolixus infected with

Absolute counts of monocytes, of natural killer T-cell (NKT) and of natural killer (NK) cells. Results refer to 10 healthy volunteers, 18 patients with flu-like syndrome and 31

Webb (1976), terem acentuado acima de tudo a vertente economicista e funcional dos sindicatos – o chamado “sindicalismo de mercado” –, que efectivamente deu lugar às modalidades