PreprintPDF Available

Unique Immunological Profile In Patients With COVID-19

Authors:

Abstract and Figures

The relationship between SARS-CoV-2 and host immunity is unknown. We show here that patients with COVID-19 had an altered immune phenotype, with an expansion of adaptive FceRIg neg NK cells, and inflammatory CD14 ⁺ CD16 ⁺ monocytes. T cells were reduced and overexpressed the Tim-3 exhaustion molecule. Low frequencies of CD8 T cells and NKG2A ⁺ NK cells, and expansion of mature CD57 ⁺ NK cells were associated with poor prognosis. These findings unveil a unique immunological profile in COVID-19 patients.
Content may be subject to copyright.
1
Preprint:Pleasenotethatthisarticlehasnotcompletedpeerreview.
UniqueImmunologicalProfileInPatientsWithCOVID-
19
CURRENTSTATUS:UND ERREVI EW
StefaniaVarchetta
FondazioneIRCCSPoliclinicoSanMatteo
DalilaMele
FondazioneIRCCSPoliclinicoSanMatteo
BarbaraOliviero
FondazioneIRCCSPoliclinicoSanMatteo
StefaniaMantovani
FondazioneIRCCSPoliclinicoSanMatteo
SerenaLudovisi
FondazioneIRCCSPoliclinicoSanMatteo
AntonellaCerino
FondazioneIRCCSPoliclinicoSanMatteo
MarcoVecchia
FondazioneIRCCSPoliclinicoSanMatteo
SilviaRoda
FondazioneIRCCSPoliclinicoSanMatteo
MicheleSachs
FondazioneIRCCSPoliclinicoSanMatteo
RaffaeleBruno
FondazioneIRCCSPoliclinicoSanMatteo
MarioU.Mondelli
FondazioneIRCCSPoliclinicosanMatteo
mario.mondelli@unipv.itCorrespondingAuthor
2
DOI:
10.21203/rs.3.rs-23953/v1
SUBJECTAREAS
Immunology InfectiousDiseases
KEYWORDS
SARS-CoV-2,hostimmunity,alteredimmunephenotypeinpatients
3
Abstract
TherelationshipbetweenSARS-CoV-2andhostimmunityisunknown.Weshowherethatpatients
withCOVID-19hadanalteredimmunephenotype,withanexpansionofadaptiveFceRIgnegNKcells,
andinflammatoryCD14+CD16+monocytes.TcellswerereducedandoverexpressedtheTim-3
exhaustionmolecule.LowfrequenciesofCD8TcellsandNKG2A+NKcells,andexpansionofmature
CD57+NKcellswereassociatedwithpoorprognosis.Thesefindingsunveilauniqueimmunological
profileinCOVID-19patients.
Introduction,ResultsAndDiscussion
SevereAcuteRespiratorySyndromeCoronavirus-2(SARS-CoV-2)isresponsibleforapandemicthus
farresponsiblefornearly1millioncasesofCoronavirusDisease-19(COVID-19)withacase/fatality
rateof4.5%[1].Theinfectionusuallycausesmildsymptoms,butmayberesponsibleforsevere
interstitialpneumonia,myocarditis,acutekidneyinjury,acuterespiratorydistresssyndrome(ARDS),
multiorganfailureanddeath[2].Laboratorytestsindicatethatpatientswithsevereprogressionof
COVID-19showsignsofsecondaryhaemophagocyticlymphohistiocytosis(sHLH),a
hyperinflammatorysyndromecharacterisedbyapotentiallyfatalcytokinestormwithmultiorgan
failure,whichmaybetriggeredbyviralinfections[3].AkintosHLH,COVID-19ischaracterizedby
lymphopenia,andincreasedserumferritin,D-dimer,C-reactiveprotein(CRP),andlactic-
dehydrogenase(LDH),whicharealsoconsideredpredictorsofpooroutcome[4].
Moreover,severalserumcytokineconcentrationsareincreasedduringCOVID-19,supportingthe
hypothesisthatvirallydrivenhyperinflammationplaysakeypathogeneticrole[2].
Despiteclearevidenceofongoingoverexuberantinflammation,therearenosystematicstudies
addressingphenotypicandfunctionalalterationsofinnateandadaptiveimmunecells,thatarelikely
exposedtoavarietyofstimuliinCOVID-19patientsatpresentation.Thelackofacomprehensive
immunologicalanalysispromptedustoassessthephenotypicandfunctionalstatusofNKcells,γδT
cells,monocytesandCD4andCD8Tcellsinpatientspresentingwithclinicallymoderatetosevere
interstitialpneumoniaemerginginthesettingofCOVID-19.Patientclinicaldetailsandlaboratory
findings,aswellasperipheralbloodmononuclearcells(PBMC)flowcytometricanalysisarereported
4
inSupplementaryInformation,PatientsandMethods.
ThefrequencyofNKcellswassignificantlyhigherinCOVID-19patientscomparedtohealthycontrols,
beingsignificantlyenrichedinmature(CD56dimCD57+)NKcells(Fig.1a).Interestingly,therewasa
relativeexpansionofCD57+/FcεRnegadaptiveNKcellscomparedwithnon-COVID-19disease
controlsandhealthycontrols(Fig.1a)suggestingaSARS-CoV-2-relatedexpansionofthispopulation,
whereastheproportionofCD56brightNKcellswasreduced.AnincreaseinCD16+NKcellswasalso
evidentcomparedwithhealthycontrols(Fig1a).Notably,thefrequencyofCXCR6-expressingNKcells
waslowinCOVID-19patients(Fig.1a),mostlikelysincethesecellshometothelungswherethey
concentrate,theirligandCXCL16beingproducedinlargeamountsbyalveolarmacrophages[5].
AdditionalchangesinNKcellsincludedsignificantreductionsinthefrequenciesofSiglec-7,DNAM-1,
NKG2D,NKp30(Fig.1a),thelatterbeingparticularlyevidentintheadaptivesubset(Fig.1c).
Importantly,thefrequencyofPD-1positiveNKcellswassignificantlyhigherintheadaptivecompared
withconventionalNKcellsinpatientswithCOVID-19(Fig.1c).NochangeswerenotedinbulkNKcell
expressionofNKG2C,NKG2A,GITR,TRAIL,CD69,PD-1,TIGIT.ThetrendnotedforTIM-3wasnot
statisticallysignificant(Suppl.Fig.1a).Ofnote,althoughnosignificantchangesindegranulation
activityorIFNγproductionwereobservedusingK562astargetcells(Suppl.Fig.1b),therewasan
increasedabilityofNKcellstoexertantibody-dependentcell-mediatedcytotoxicity(ADCC),a
functionexquisitelyperformedbyadaptiveNKcells[6](Fig.1d).TheproportionsofCD56bright,
NKG2AandNKp46positiveNKcellsweresignificantlylowerandtheproportionofmatureCD57+cells
significantlyhigherinpatientswhosuccumbedcomparedwiththosewhosurvived(Fig.2a).The
relativefrequenciesoftotalCD3+,CD4+andCD8+Tcellsweresignificantlylowerthanhealthy
controls,althoughnoapparentdifferenceswerenotedwithdiseasecontrols(Fig.2b).Patientswho
diedshowedasignificantlylowerfrequencyofCD8Tcellscomparedwiththosewhosurvived(Fig.2a).
Moreover,bothCD4andCD8TcellsfromCOVID-19patientsoverexpressedTim-3comparedwith
healthycontrols,suggestingapanT-cellexhaustionprofile(Fig.2c).Nodifferenceswerefoundin
CD45RO,HLA-DR,GITRexpressionorTregpopulationfrequency(Suppl.Fig.2).Importantly,therewas
5
aclearrelativeexpansionofCD14/CD16doublepositivemonocytes,aphenotypeassociatedwithan
inflammatoryprofile(Fig.2d)[7].Therewerenostatisticallysignificantchangesinthefrequencyof
γδTcells(Fig.2e).Negativecorrelationswerefoundbetweenlaboratoryindicatorsofsevereor
progressivedisease.Thus,NKcellsexpressingtheactivatingreceptorsNKp30andNKp46,aswellas
CD45RO+andTim-3+CD4Tcells,correlatednegativelywithLDH(Fig.2f).Anegativecorrelationwas
alsopresentbetweenNKp46+NKcellsandCRP(Fig.2g).
InformationonPBMCphenotypeandfunctionarevirtuallylackinginpatientswithCOVID-19.Herewe
hadtheopportunitytoevaluatepatientsadmittedtohospitalbecauseofmoderatetosevereCOVID-
19interstitialpneumoniaandcomparedthemtoasmallgroupofSARS-CoV-2negativepneumonia
andhealthyWeshowedthatpatientswithCOVID-19hadarelativeexpansionofmatureadaptiveNK
cellsendowedwithADCCfunction,whichwasincreasedinthissettinginlinewithfindingsinother
viralinfections,particularlycytomegalovirus[6].Otherphenotypicfeatureswerecompatiblewitha
dysfunctionalNKcellphenotype,namelythereducedfrequenciesofSiglec-7-,NKG2D-andNKp30-
expressingcells[8,9].Arecentstudyaddressedthekineticsandbreadthofimmuneresponses
associatedwithclinicalresolutionofCOVID-19inasinglepatientwithrelativelymilddisease[10].
Antibody-secretingcellsappearedatthetimeofviralclearancetogetherwithfollicularhelperTcells
andactivatedCD4andCD8Tcells.Incontrast,inourpatientswithmoderatetosevereinterstitial
pneumonia,someofwhomsadlysuccumbed,Tim-3positiveexhaustedCD4andCD8Tcellslargely
prevailedatpresentationandlowerfrequenciesofCD8+Tcellswerelinkedtopoorprognosis.A
recentstudyfoundlowerfrequenciesofCD8TcellsandNKcellswitharelativeenrichmentofNKG2A-
expressingcellswhichreturnedtonormalafterclinicalrecovery,suggestingrescueofimpairedTand
NKcellfunction[11].Interestingly,althoughnodifferenceinthefrequencyofNKG2A-expressingNK
cellswasfoundbetweenpatientsandcontrolsinthepresentstudy,NKG2A+NKcellswerelowerin
patientswhodidnotsurvive,suggestingthatlossofthisinhibitoryreceptorsomehowunleashedNK
cellsinpatientswithfataloutcome.
OurstudyprovidesimportantnovelinsightsintothepathogeneticmechanismsofCOVID-19,
6
characterizedbyarapidexpansionofphenotypicallymatureNKcellspersistingathighfrequencyin
patientwithpoorprognosis.ThesimultaneouslyreducedfrequencyofCD4+andCD8+Tcells
expressingtheTim-3exhaustionmarkerunveilsamultifacetedbehaviorofthetwoarmsofimmunity
inthisclinicalsetting.Therelativeenrichmentofinflammatorymonocyteslendssupporttothe
hypothesisthatCOVID-19resemblesinparttothemacrophage-activationsyndromewhichisthought
tobecloselyrelatedtohemophagocyticlymphohistiocytosis(HLH)[12],anuncommonlife-
threateningdisorderofseverehyperinflammationcausedbyuncontrolledproliferationofactivated
lymphocytesandmacrophagesthatsecretehighlevelsofinflammatorycytokines.Ofnotepatterns
similartocytokinestormsyndromeshavebeendescribedforCOVID-19andSARS[2].
ItisdifficultatthisearlystagetopreciselyframeCOVID-19withinanimmunologicallycoherent
clinicalentity.Indeed,severalpeculiaritieshaveemergedthatcontributetotheuniquenessofits
immuneprofile.UnderstandingthedynamicsandthequalityofimmuneresponsestoSARS-CoV-2will
provideinvaluabletranslationalinformationtodesigneffectivetreatmentsforthispotentiallydeadly
disease.
Methods
Providedassupplementalfile.
Declarations
AuthorContributions:SVandDLdesignedandperformedexperimentsandcriticallycontributedto
draftingthemanuscript;BO,SM,ACperformedexperimentsandcriticallyreadthemanuscript;SL,
MV,SR,MS,RBrecruitedpatients,preparedthedatabaseandcriticallyreadthemanuscript,MUM
designedanddiscussedtheexperimentsandwrotethemanuscript.
CompetingInterests:none.
Ethics:Thestudyprotocolconformedtotheethicalguidelinesofthe1975DeclarationofHelsinkiand
wasapprovedbytheInstitutionalReviewBoardandEthicalCommitteeofFondazioneIRCCS
PoliclinicoSanMatteo(Protocolnumber20200033215).Allpatientsprovidedwrittenor,incasethey
wereunabletosign,verballywitnessedinformedconsentaspertheabovestudyprotocol.
References
1.  h t t p s://worldometers.in f o / c o r o n a v i r u s/
7
2.  H uang,etal.ClinicalFea t u r e s o f P a t i e ntsInfectedWith201 9 N o v e l C o r onavirusin
Wuhan,China.Lan c e t 395,497-506(2020).
3.  R a mos-Casals,etal.Ad u l t H a e m o p hagocyticSyndrom e . L a n c e t 383,1503–16(2014).
4.  R u an,etal.ClinicalPredic t o r s o f M o r t a lit yDuetoCOVID-19B a s e d o n a n A nalysisof
Dataof150Patien t s F r o m W u han,China.Intensive C a r e M e d .
https://doi.org/10.1 0 0 7 / s 0 0 1 3 4-020-05991-x(2 0 2 0 ) .
5.  M organ,J.etal.Expre s s i o n o f C X C R 6anditsligandCXCL 1 6 i n t h e l u n ginhealthand
disease.Clin.Exp.Alle r g y 3 5 , 1572–1580(2005)
6.  S c hlums,H.etal.Cytom e g a l o v i r u s In fectionDrivesAdaptiv e E p i g e n e t i c
DiversificationofNKC e l l s w i t h A l t e r e dSignalingandEffe c t o r I m m u n it y 4 2,443–456
(2015).
7.  Z i e gler-Heitbrock,TheC D 1 4 + C D 1 6+BloodMonocy t e s : T h e i r R o l e in Infectionand
Inflammation.J.Leu k o c . B i o l . 8 1 ,584-92.
8.  V a rchetta,etal.Lacko f S i g l e c - 7 E x p ressionIdentifiesaDy s f u n c t i o n a l N aturalKiller
CellSubsetAssociate d W i t h L i v e r I n flammationandFib r o s i s i n C h r o n ic H CVInfection.
Gut65,1998-2006(2016).
9.  V i t a le ,etal.AnHistoricalO v e r v i e w : T h e D iscoveryofHowN K C e l l s C a n K i l l E nemies,
RecruitDefenseTro o p s , a n d M ore.Front.Immuno l . 10,1415(2019).
10.  T h e v arajan,etal.Breadth o f c o n c o m it a ntimmunerespon s e s p r i o r t o p atient
recovery:acaser e p o r t o f n o n - severeCOVID-19.Na t . M e d .
https://doi.org/10.1 0 3 8 / s 4 1 5 9 1-020-0819-2(20 2 0 ) .
11.  Z h e n g,M.etal.Functiona l e x h a u s t i o n o fantivirallymphocyte s i n C O V I D - 1 9 patients.
Mol.Immunol.https : / / d o i . o r g / 1 0 .1 038/s41423-02 0 - 0 4 0 2 - 2 ( 2 020).
12.  F i s m an,N.Hemophago c y t i c s y n d r o mesandinfection.E m e r g i n g I n f e ct.Dis.6,601–8
(2000).
8
Covid19IrccsSanMatteoPaviaTaskForce
IDStaff
RaffaeleBruno,MarioUMondelli,EnricoBrunetti,AngelaDiMatteo,ElenaSeminari,LauraMaiocchi,
ValentinaZuccaro,LaylaPagnucco,BiancaMariani,SerenaLudovisi,RaffaellaLissandrin,AldoParisi,
PaoloSacchi,SavinoFAPatruno,GiuseppeMichelone,RobertoGulminetti,DomenicoZanaboni,
StefanoNovati,RenatoMaserati,PaoloOrsolini,MarcoVecchia.
IDResidents
MarcoSciarra,ErikaAsperges,MartaColaneri,AlessandroDiFilippo,MargheritaSambo,Simona
Biscarini,MatteoLupi,SilviaRoda,TeresaChiaraPieri,IlariaGallazzi,MicheleSachs,PietroValsecchi.
EmergencyCareUnit
ECUStaff:StefanoPerlini,ClaudiaAlfano,MarcoBonzano,FedericaBriganti,GiuseppeCrescenzi,
AnnaGiuliaFalchi,RobertaGuarnone,BarbaraGuglielmana,ElenaMaggi,IlariaMartino,Pietro
Pettenazza,SerenaPiolidiMarco,FedericaQuaglia,AnnaSabena,FrancescoSalinaro,Francesco
Speciale,IlariaZunino.
ECUResidents:MarziaDeLorenzo,GianmarcoSecco,LorenzoDimitry,GiovanniCappa,IgorMaisak,
BenedettaChiodi,MassimilianoSciarrini,BrunoBarcella,FlaviaResta,LucaMoroni,GiuliaVezzoni,
LorenzoScattaglia,ElisaBoscolo,CaterinaZattera,TassiMicheleFidel,CapozzaVincenzo,Damiano
Vignaroli,MarcoBazzini.
IntensiveCareUnit
GiorgioIotti,FrancescoMojoli,MirkoBelliato,LucianoPerotti,SilviaMongodi,GuidoTavazzi
PaediatricUnit
GianluigiMarseglia,AmeliaLicari,IlariaBrambilla
VirologyStaff
BarbariniDaniela,BrunoAntonella,CambieriPatrizia,CampaniniGiulia,ComolliGiuditta,Corbella
Marta,DaturiRossana,FurioneMilena,MarianiBianca,MaseratiRoberta,MonzilloEnza,Paolucci
Stefania,PareaMaurizio,PercivalleElena,PirallaAntonio,RovidaFrancesca,SarasiniAntonella,
ZavattoniMaurizio.
9
VirologyTechnicalstaff
AdzasehounGuy,BellottiLaura,CabanoErmanna,CasaliGiuliana,DossenaLuca,FriscoGabriella,
GarbagnoliGabriella,GirelloAlessia,LandiniViviana,LucchelliClaudia,MaliardiValentina,Pezzaia
Simona,PremoliMarta.
VirologyResidents
BonettiAlice,CanevaGiacomo,CassanitiIrene,CorcioneAlfonso,DiMartinoRaffella,DiNapoli
Annapia,FerrariAlessandro,FerrariGuglielmo,FiorinaLoretta,GiardinaFederica,MercatoAlessandra,
NovazziFederica,RatanoGiacomo,RossiBeatrice,SciabicaIreneMaria,TallaritaMonica,Vecchio
NepitaEdoardo.
ResearchLaboratories,DivisionofInfectiousDiseasesandImmunology
AntonellaCerino,StefaniaVarchetta,BarbaraOliviero,StefaniaMantovani,DalilaMele.
PharmacyUnit
MonicaCalvi,MichelaTizzoni
HospitalManagement
CarloNicora,AntonioTriarico,VincenzoPetronella,CarloMarena,AlbaMuzzi,PaoloLago
DataUnit
FrancescoComandatore,GherardBatistiBissignandi,StefanoGaiarsa,MarcoRettani,ClaudioBandi
Figures
10
11
Figure1
NKcellcharacterizationinSARS-CoV-2infection.a)FrequencyofNKcellsandexpansionof
matureCD57+andadaptive(FcεRIγneg)NKcellsinCOVID-19patients.Reducedfrequency
ofCXCR6,Siglec-7,NKG2DandNKp30,andincreasedproportionofCD16+cells.b)Dotplot
showinggatingonCD57+FcεRIγnegadaptiveandCD57+FcεRIγposconventionalNKcells.c)
RepresentativedotplotsandgraphsshowingNKp30reductionandPD1increaseinadaptive
comparedwithconventionalNKcellsinCOVID-19patients.Representativedotplotsare
gatedontotalCD57+NKcells.CirclesindicateadaptiveNK;squares,conventionalNK.d)
IncreasedNKdegranulationandIFNγexpressioninCOVID-19patients.e)Representative
IFNγandCD107adotplotsinpatientsandcontrols.Fullredsymbolsindicatepatientswho
subsequentlydied.Middlebarsrepresentmedians.TheOneWayAnovatestwasusedto
comparethreegroups.*p<0.05,**p<0.01,***p<0.001.
12
Figure2
13
a)ExpansionofmatureCD57+NKcellsandreductionofCD56brightNKcells,NKG2A+and
NKp46+NKcellsandCD8Tcellsinpatientswhosurvivedandinthosewhosuccumbed.b)
FrequenciesoftotalCD3+,CD4+andCD8+TcellswerereducedinCOVID-19patients
comparedtoHD.c)Tim-3expressingCD4andCD8TcellswereincreasedinCOVID-19
patients.d)ExpansionofCD14+CD16+doublepositivemonocytesinCOVID-19patients
andrepresentativedotplots.e)NodifferenceswereobservedintheproportionofγδTcells.
f&g)CorrelationsofNKandCD4TreceptormoleculeswithLDHandCRP.Middlebars
representmedianvalues.TheMann-Whitneytestwasusedtocomparesurvivorsversus
deadpatients.TheOneWayAnovatestwasusedtocomparethreegroups.ThePearson
testwasusedtoexaminecorrelations.*p<0.05,**p<0.01.
SupplementaryFiles
Thisisalistofsupplementaryfilesassociatedwiththispreprint.Clicktodownload.
supinfo.pdf
... Some studies also reported that NK cells can expand to adaptive NK cells, which represent a terminally differentiated subset of NK cells defined by increased expression of NKG2C and CD57 (26,27). Regarding the cytotoxic activity of NK cells in COVID-19, functional impairment, which is founded by either indirect observations such as decline of perforin levels, increase in exhaustion markers, or decreased degranulation, as measured by CD107a expression when PBMCs from COVID-19 patients are cocultured with tumour target cells (19, [28][29][30][31]. Some studies have used an in vitro cytotoxic killing assay for the assessment of cytotoxicity. ...
... Consistent with other reports, we found that SARS-CoV-2 infection triggers maturation of NK cells leading to increased frequencies of CD56 dim NK cells (26,29,32,44). The increased levels of terminally differentiated inflammatory NK cells in severe COVID-19 patients were also confirmed in two additional scRNA-seq datasets (33). ...
... Adaptive NK cells are another CD56 dim subset of terminally differentiated NK cells, were non-significantly increased in our COVID-19 cohort subjected to transcriptome analysis. Using flow cytometry, two studies found an increased frequency of these adaptive NK cells, phenotyped by either CD56 dim NKG2C + CD57 + or FceRig -CD56 +CD57+ cells, in severe but not in moderate COVID-19 patients (26,29). In contrast, Leem et al. and Witkowski et al., reported no significant numerical changes of adaptive NK cells (32,33). ...
Article
Full-text available
COVID-19 is characterised by a broad spectrum of clinical and pathological features. Natural killer (NK) cells play an important role in innate immune responses to viral infections. Here, we analysed the phenotype and activity of NK cells in the blood of COVID-19 patients using flow cytometry, single-cell RNA-sequencing (scRNA-seq), and a cytotoxic killing assay. In the plasma of patients, we quantified the main cytokines and chemokines. Our cohort comprises COVID-19 patients hospitalised in a low-care ward unit (WARD), patients with severe COVID-19 disease symptoms hospitalised in intensive care units (ICU), and post-COVID-19 patients, who were discharged from hospital six weeks earlier. NK cells from hospitalised COVID-19 patients displayed an activated phenotype with substantial differences between WARD and ICU patients and the timing when samples were taken post-onset of symptoms. While NK cells from COVID-19 patients at an early stage of infection showed increased expression of the cytotoxic molecules perforin and granzyme A and B, NK cells from patients at later stages of COVID-19 presented enhanced levels of IFN-γ and TNF-α which were measured ex vivo in the absence of usual in vitro stimulation. These activated NK cells were phenotyped as CD49a⁺CD69a⁺CD107a⁺ cells, and their emergence in patients correlated to the number of neutrophils, and plasma IL-15, a key cytokine in NK cell activation. Despite lower amounts of cytotoxic molecules in NK cells of patients with severe symptoms, majority of COVID-19 patients displayed a normal cytotoxic killing of Raji tumour target cells. In vitro stimulation of patients blood cells by IL-12+IL-18 revealed a defective IFN-γ production in NK cells of ICU patients only, indicative of an exhausted phenotype. ScRNA-seq revealed, predominantly in patients with severe COVID-19 disease symptoms, the emergence of an NK cell subset with a platelet gene signature that we identified by flow and imaging cytometry as aggregates of NK cells with CD42a⁺CD62P⁺ activated platelets. Post-COVID-19 patients show slow recovery of NK cell frequencies and phenotype. Our study points to substantial changes in NK cell phenotype during COVID-19 disease and forms a basis to explore the contribution of platelet-NK cell aggregates to antiviral immunity against SARS-CoV-2 and disease pathology.
... While a drop in total lymphocyte counts has been widely reported, some groups have found no or minimal change in the blood of total NK cell levels (Varchetta et al., 2020). ...
... This study found that the rare subset of adaptive NK cells, which are NKG2C + CD57 + , were increased in severe cases, as were CD-56 bright cells with a cytotoxic profile (perforin, Ksp37, MIP-1β, CD98, Tim-3 and granzyme B positive) (Maucourant et al., 2020). Another study found the related CD57+/FcεRIγ-adaptive NK cells to be increased in patients and associated with poor prognosis as well (Varchetta et al., 2020). One report found that activated CD56-CD16+ NK cells, highly cytotoxic cells, which, along with CD56 dim NKs, mediate ADCC, were increased (Marcos-Jimenez et al., 2020). ...
... Peripheral blood T and NK cells have been found to express the checkpoint molecule PD-1 in a number of studies (Diao et al., 2020;Varchetta et al., 2020;. In adaptive NK cells, which -as mentioned in one study above-were one of the few lymphocyte populations to increase with disease severity, PD-1 was found to be increased (Maucourant et al., 2020). ...
Article
Full-text available
The outbreak of the novel coronavirus SARS-CoV-2 and the attendant physiological symptoms associated with the COVID-19 disease have led to an explosion of interest studying different aspects of the immune response. As of yet, the particular roles of natural killer cells are not well understood in this disease. NK cells are critical first-response cytotoxic cells of the innate immune system. NK cells are traditionally considered important for their roles in innate immunity against tumors and viral infected cells, as well as their ability to produce cytokines, particularly interferon-?, and participate in antibody dependent cell cytotoxicity (ADCC). Here, we describe the role of NK cells in peripheral blood and in the lungs with respect to the pathology caused by SARS-CoV-2 and discuss the implications of proposed different types of therapies on NK cells. Evidence is accumulating that NK cells play an important role in initial surveillance as part of innate immunity. With the progression of the disease and rising inflammation, these cells, when in circulation, appear to become exhausted and ineffective. In the COVID lung, however, a complex interplay between inflammatory cells, chemokines, cytokines and aberrantly activated migratory NK cells occurs, potentiating local inflammation and the critical situation in the lungs.
... In this study, admission levels of 17 biomarkers (increased levels of CRP and 14 cytokines and reduced levels of MIP-1β and lymphocytes) were associated significantly with progression to severe disease. Our biomarker findings are similar to previously reported associations of the levels of several cytokines (e.g., IL-1ra, IL-2, IL-6, IL-8, IL-10, and IP-10) with COVID-19 severity and mortality (25,27,(39)(40)(41). The association of the IL-10 level with COVID-19 progression to severity has been reported in a considerable number of publications (40)(41)(42). ...
... Our biomarker findings are similar to previously reported associations of the levels of several cytokines (e.g., IL-1ra, IL-2, IL-6, IL-8, IL-10, and IP-10) with COVID-19 severity and mortality (25,27,(39)(40)(41). The association of the IL-10 level with COVID-19 progression to severity has been reported in a considerable number of publications (40)(41)(42). IL-10 is an immunoregulatory cytokine with the main functions of limiting inflammatory responses and regulating immune cell differentiation and proliferation (43). Information about the role of IL-12 (p70) in COVID-19 is more limited. ...
Article
Full-text available
Background: Cardiovascular comorbidities such as hypertension and inflammatory response dysregulation are associated with worse COVID-19 prognoses. Different cytokines have been proposed to play vital pathophysiological roles in COVID-19 progression, but appropriate prognostic biomarkers remain lacking. We hypothesized that the combination of immunological and clinical variables at admission could predict the clinical progression of COVID-19 in hypertensive patients. Methods: The levels of biomarkers, including C-reactive protein, lymphocytes, monocytes, and a panel of 29 cytokines, were measured in blood samples from 167 hypertensive patients included in the BRACE-CORONA trial. The primary outcome was the highest score during hospitalization on the modified WHO Ordinal Scale for Clinical Improvement. The probability of progression to severe disease was estimated using a logistic regression model that included clinical variables and biomarkers associated significantly with the primary outcome. Results: During hospitalization, 13 (7.8%) patients showed progression to more severe forms of COVID-19, including three deaths. Obesity, diabetes, oxygen saturation, lung involvement on computed tomography examination, the C-reactive protein level, levels of 15 cytokines, and lymphopenia on admission were associated with progression to severe COVID-19. Elevated levels of interleukin-10 and interleukin-12 (p70) combined with two or three of the abovementioned clinical comorbidities were associated strongly with progression to severe COVID-19. The risk of progression to severe disease reached 97.5% in the presence of the five variables included in our model. Conclusions: This study demonstrated that interleukin-10 and interleukin-12 (p70) levels, in combination with clinical variables, at hospital admission are key biomarkers associated with an increased risk of disease progression in hypertensive patients with COVID-19.
... Functionally, NK cells exhibit reduced cytotoxicity in COVID-19, previously explained by an over-expression of the inhibitory NKG2A receptor (Zheng et al., 2020), but not confirmed by other groups. In fact, depending on the studies, other markers were found to be responsible, such as NKG2D, DNAM-1 or NKp30 (Varchetta et al., 2020), or even the presence of adaptive NKG2C + NK cells which correlate with disease severity (Maucourant et al., 2020). Alternatively, it was also reported that granzyme A-expressing NK cells were negatively correlated with serum IL-6 levels in COVID-19 patients, suggesting a link to inflammation (20 Mazzoni et al., 2020). ...
Article
Full-text available
Introduction Increasing evidence has shown that coronavirus disease 19 (COVID-19) severity is driven by a dysregulated immunological response. Previous studies have demonstrated that natural killer (NK) cell dysfunction underpins severe illness in COVID-19 patients, but have lacked an in-depth analysis of NK cell markers as a driver of death in the most critically ill patients. Methods We enrolled 50 non-vaccinated hospitalized patients infected with the initial virus or the alpha variant of SARS-CoV-2 with moderate or severe illness, to evaluate phenotypic and functional features of NK cells. Results Here, we show that, consistent with previous studies, evolution NK cells from COVID-19 patients are more activated, with the decreased activation of natural cytotoxicity receptors and impaired cytotoxicity and IFN-γ production, in association with disease regardless of the SARS-CoV-2 strain. Fatality was observed in 6 of 17 patients with severe disease; NK cells from all of these patients displayed a peculiar phenotype of an activated memory-like phenotype associated with massive TNF-α production. Discussion These data suggest that fatal COVID-19 infection is driven by an uncoordinated inflammatory response in part mediated by a specific subset of activated NK cells.
... This difference is particularly obvious in patients with fatal outcomes. Moreover, the frequency of adaptive/memory NK cells in deceased patients increases statistically (83). Other work showed that adaptive NK cells had signs of expansion in patients with COVID-19, but this did not rely on the HCMV activation secondary to COVID-19. ...
Article
Full-text available
Natural killer (NK) cells are an important component of the innate immune system due to their strong ability to kill virally infected or transformed cells without prior exposure to the antigen (Ag). However, the biology of human NK (hNK) cells has largely remained elusive. Recent advances have characterized several novel hNK subsets. Among them, adaptive NK cells demonstrate an intriguing specialized antibody (Ab)-dependent response and several adaptive immune features. Most adaptive NK cells express a higher level of NKG2C but lack an intracellular signaling adaptor, FcϵRIγ (hereafter abbreviated as FcRγ). The specific expression pattern of these genes, with other signature genes, is the result of a specific epigenetic modification. The expansion of adaptive NK cells in vivo has been documented in various viral infections, while the frequency of adaptive NK cells among peripheral blood mononuclear cells correlates with improved prognosis of monoclonal Ab treatment against leukemia. This review summarizes the discovery and signature phenotype of adaptive NK cells. We also discuss the reported association between adaptive NK cells and pathological conditions. Finally, we briefly highlight the application of adaptive NK cells in adoptive cell therapy against cancer.
... The disease caused by SARS-CoV-2 (i.e., presents severe symptoms including pneumonia, acute respiratory distress syndrome (5), neurological symptoms, organ failure, and death. More importantly, severe COVID-19 patients may experience dysregulation of an appropriate immune response, characterized by lymphopenia (6), high neutrophil levels in peripheral blood (7), and increased pro-inflammatory cytokines and chemokines (8). Repurposed therapeutics such as monoclonal antibody cocktails, convalescent plasma, and dexamethasone have shown promising results in treating COVID-19 (9). ...
Article
Full-text available
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is highly contagious and presents a significant public health issue. Current therapies used to treat coronavirus disease 2019 (COVID-19) include monoclonal antibody cocktail, convalescent plasma, antivirals, immunomodulators, and anticoagulants. The vaccines from Pfizer and Moderna have recently been authorized for emergency use, which are invaluable for the prevention of SARS-CoV-2 infection. However, their long-term side effects are not yet documented, and populations with immunocompromised conditions (e.g., organ-transplantation and immunodeficient patients) may not be able to mount an effective immune response. In addition, there are concerns that wide-scale immunity to SARS-CoV-2 may introduce immune pressure that could select for escape mutants to the existing vaccines and monoclonal antibody therapies. Emerging evidence has shown that chimeric antigen receptor (CAR)- natural killer (NK) immunotherapy has potent antitumor response in hematologic cancers with minimal adverse effects in recent studies, however, the potentials of CAR-NK cells in treating COVID-19 has not yet been fully exploited. Here, we improve upon a novel approach for the generation of CAR-NK cells for targeting SARS-CoV-2 and its various mutants. CAR-NK cells were generated using the scFv domain of S309 (henceforward, S309-CAR-NK), a SARS-CoV and SARS-CoV-2 neutralizing antibody (NAbs) that targets the highly conserved region of SARS-CoV-2 spike (S) glycoprotein and is therefore more likely to recognize different variants of SARS-CoV-2 isolates. S309-CAR-NK cells can specifically bind to pseudotyped SARS-CoV-2 virus and its D614G, N501Y, and E484K mutants. Furthermore, S309-CAR-NK cells can specifically kill target cells expressing SARS-CoV-2 S protein in vitro and show superior killing activity and cytokine production, compared to that of the recently reported CR3022-CAR-NK cells. Thus, these results pave the way for generating ‘off-the-shelf’ S309-CAR-NK cells for treatment in high-risk individuals as well as provide an alternative strategy for patients unresponsive to current vaccines.
... A significantly higher number of memory NK cells in deceased patients (66). ...
Article
Full-text available
SARS-CoV-2 is the cause of a recent pandemic that has led to more than 3 million deaths worldwide. Most individuals are asymptomatic or display mild symptoms, which raises an inherent question as to how does the immune response differs from patients manifesting severe disease? During the initial phase of infection, dysregulated effector immune cells such as neutrophils, macrophages, monocytes, megakaryocytes, basophils, eosinophils, erythroid progenitor cells, and Th17 cells can alter the trajectory of an infected patient to severe disease. On the other hand, properly functioning CD4+, CD8+ cells, NK cells, and DCs reduce the disease severity. Detailed understanding of the immune response of convalescent individuals transitioning from the effector phase to the immunogenic memory phase can provide vital clues to understanding essential variables to assess vaccine-induced protection. Although neutralizing antibodies can wane over time, long-lasting B and T memory cells can persist in recovered individuals. The natural immunological memory captures the diverse repertoire of SARS-CoV-2 epitopes after natural infection whereas, currently approved vaccines are based on a single epitope, spike protein. It is essential to understand the nature of the immune response to natural infection to better identify ‘correlates of protection’ against this disease. This article discusses recent findings regarding immune response against natural infection to SARS-CoV-2 and the nature of immunogenic memory. More precise knowledge of the acute phase of immune response and its transition to immunological memory will contribute to the future design of vaccines and the identification of variables essential to maintain immune protection across diverse populations.
Article
Background Severe cases of coronavirus disease 2019 (COVID-19) have increased risk for acute kidney injury (AKI). The exacerbation of the immune response seems to contribute to AKI development, but the immunopathological process is not completely understood. Objectives To analyze levels of circulant immune mediators in COVID-19 patients evolving with or without AKI. We have also investigated the possible associations of these mediators with viral load and clinical outcomes. Methods This is a longitudinal study performed with hospitalized patients with moderate to severe COVID-19. Serum levels of 27 immune mediators were measured by a multiplex immunoassay. Data were analyzed at two timepoints during the follow-up: within the first 13 days of the disease onset (early sample) and from the 14th day to death or hospital discharge (follow-up sample). Results We studied 82 COVID-19 patients (59.5 ± 17.5 years, 54.9% male). Of these, 34 (41.5%) developed AKI. These patients presented higher SARS-CoV-2 viral load (P=0.03), higher frequency of diabetes (P=0.01) and death (P=0.0004). Overall, AKI patients presented significantly higher and sustained levels (P<0.05) of CCL-2, CCL-3, CCL-4, CXCL-8, CXCL-10, IFN-γ, IL-2, IL-6, TNF-α, IL-1Ra, IL-10 and VEGF. Importantly, higher levels of CCL-2, CXCL-10, IL-2, TNF-α, IL-10, FGFb, and VEGF were observed in AKI patients independently of death. ROC curves demonstrated that early alterations in CCL-2, CXCL-8, CXCL-10, IFN-γ, IL-6, IL-1Ra and IL-10 show a good predictive value regarding AKI development. Lastly, immune mediators were significantly associated with each other and with SARS-CoV-2 viral load in AKI patients. Conclusions COVID-19 associated AKI is accompanied by substantial alterations in circulant levels of immune mediators, which could significantly contribute to the establishment of kidney injury.
Chapter
Full-text available
Coronavirus disease 2019 (COVID-19) and its etiological agent severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) have caused considerable illness and death worldwide. The innate immune system seems to play a principal in the disease, as a hallmark of severe COVID-19 is excessive inflammation. Monocytes and macrophages are important innate immune cells that become pro-inflammatory and promote adaptive immune responses during viral infection. In this chapter we present evidence linking these cells to severity of COVID-19. Namely, monocytes and macrophages infiltrate the infected tissue during the early stages of infection and show pro-inflammatory responses that appear to be linked to those predicting tissue pathology during disease. Additionally, studies in isolated cells demonstrate that monocytes and macrophages respond by producing pro-inflammatory cytokines when directly stimulated by SARS-CoV-2. While most anti-inflammatory pharmaceutical treatments for COVID-19 have focused on systemic infiltration, some of the most promising have known or suspected effects on monocyte and macrophage inflammatory responses. Therefore, targeting these cells to treat severe COVID-19 is a promising strategy for this important disease.
Article
Introduction: COVID-19 pandemic has caused huge loss of human lives and extensive socio-economic damages. The immuno-pathology of this disease is neither clearly understood nor there are effective drugs for severe cases of COVID-19. Repurposing of available drugs for the treatment of COVID-19 is imperative. Areas Covered: This review has gathered the evidence from PubMed, Google Scholar, WHO, and other reliable websites on COVID-19 and summarized the existing knowledge of the immuno-pathology of COVID-19. We elucidated how vitamin D through its diverse actions on immune effector cells, epithelial cells, or renin-angiotensin-aldosterone system could have a modulatory role on the pathogenic mechanisms of COVID-19. The epidemiological evidence associating vitamin D deficiency with the severity and incidence of COVID-19 is also presented. However, the evidence of clinical benefit to patients of COVID-19 from randomized controlled trials with vitamin D has not come as yet. Expert opinion: It is now established that fatality of COVID-19 is primarily determined by hyperactivation of the host’s innate immune system in response to SARS-CoV-2 invasion, and thus the research on the immuno-modulatory and other roles of vitamin D against viral infections should be pursued vigorously. This would be also useful for future pandemics caused by other novel viruses.
Article
Full-text available
Background: A recent cluster of pneumonia cases in Wuhan, China, was caused by a novel betacoronavirus, the 2019 novel coronavirus (2019-nCoV). We report the epidemiological, clinical, laboratory, and radiological characteristics and treatment and clinical outcomes of these patients. Methods: All patients with suspected 2019-nCoV were admitted to a designated hospital in Wuhan. We prospectively collected and analysed data on patients with laboratory-confirmed 2019-nCoV infection by real-time RT-PCR and next-generation sequencing. Data were obtained with standardised data collection forms shared by the International Severe Acute Respiratory and Emerging Infection Consortium from electronic medical records. Researchers also directly communicated with patients or their families to ascertain epidemiological and symptom data. Outcomes were also compared between patients who had been admitted to the intensive care unit (ICU) and those who had not. Findings: By Jan 2, 2020, 41 admitted hospital patients had been identified as having laboratory-confirmed 2019-nCoV infection. Most of the infected patients were men (30 [73%] of 41); less than half had underlying diseases (13 [32%]), including diabetes (eight [20%]), hypertension (six [15%]), and cardiovascular disease (six [15%]). Median age was 49·0 years (IQR 41·0-58·0). 27 (66%) of 41 patients had been exposed to Huanan seafood market. One family cluster was found. Common symptoms at onset of illness were fever (40 [98%] of 41 patients), cough (31 [76%]), and myalgia or fatigue (18 [44%]); less common symptoms were sputum production (11 [28%] of 39), headache (three [8%] of 38), haemoptysis (two [5%] of 39), and diarrhoea (one [3%] of 38). Dyspnoea developed in 22 (55%) of 40 patients (median time from illness onset to dyspnoea 8·0 days [IQR 5·0-13·0]). 26 (63%) of 41 patients had lymphopenia. All 41 patients had pneumonia with abnormal findings on chest CT. Complications included acute respiratory distress syndrome (12 [29%]), RNAaemia (six [15%]), acute cardiac injury (five [12%]) and secondary infection (four [10%]). 13 (32%) patients were admitted to an ICU and six (15%) died. Compared with non-ICU patients, ICU patients had higher plasma levels of IL2, IL7, IL10, GSCF, IP10, MCP1, MIP1A, and TNFα. Interpretation: The 2019-nCoV infection caused clusters of severe respiratory illness similar to severe acute respiratory syndrome coronavirus and was associated with ICU admission and high mortality. Major gaps in our knowledge of the origin, epidemiology, duration of human transmission, and clinical spectrum of disease need fulfilment by future studies. Funding: Ministry of Science and Technology, Chinese Academy of Medical Sciences, National Natural Science Foundation of China, and Beijing Municipal Science and Technology Commission.
Article
The mechanisms underlying human natural killer (NK) cell phenotypic and functional heterogeneity are unknown. Here, we describe the emergence of diverse subsets of human NK cells selectively lacking expression of signaling proteins after human cytomegalovirus (HCMV) infection. The absence of B and myeloid cell-related signaling protein expression in these NK cell subsets correlated with promoter DNA hypermethylation. Genome-wide DNA methylation patterns were strikingly similar between HCMV-associated adaptive NK cells and cytotoxic effector T cells but differed from those of canonical NK cells. Functional interrogation demonstrated altered cytokine responsiveness in adaptive NK cells that was linked to reduced expression of the transcription factor PLZF. Furthermore, subsets of adaptive NK cells demonstrated significantly reduced functional responses to activated autologous T cells. The present results uncover a spectrum of epigenetically unique adaptive NK cell subsets that diversify in response to viral infection and have distinct functional capabilities compared to canonical NK cell subsets. Copyright © 2015 Elsevier Inc. All rights reserved.
Article
Haemophagocytic syndromes (haemophagocytic lymphohistiocytosis) have a wide range of causes, symptoms, and outcomes, but all lead to a hyperinflammatory response and organ damage-mainly reported in paediatric patients, but reports of adult presentation are increasing. Analysis of the genetic and molecular pathophysiology of these syndromes have improved the understanding of the crosstalk between lymphocytes and histiocytes and their regulatoty mechanisms. Clinical presentations with a broad differential diagnosis, and often life-threatening outcome, complicate the management, which might include supportive intensive care, immunosuppressive and biological treatments, or haemopoietic stem cell transplantation. Insufficient knowledge of these syndromes could contribute to poor prognosis. Early diagnosis is essential to initiate appropriate treatment and improve the quality of life and survival of patients with this challenging disorder.
Article
Chemokine receptors (CR) play an important role in T cell migration, but their contribution to lung trafficking is unclear. We hypothesized that if a particular CR was involved in T cell homing its expression would be enriched on lung T cells compared with peripheral blood T cells (PBT). We have measured the CR expression on BAL T cells from patients with sarcoid, other interstitial lung diseases (ILD), asthma and healthy volunteers. Of 14 CR studied in sarcoid, CXCR6 expression was the most markedly increased in the lung compared with the blood, a finding that was also seen in ILD patients. A striking although lesser increase was also seen in asthmatics and healthy controls. Analysis of expression of the CXCR6 ligand, CXCL16, by immunohistochemistry suggested that alveolar macrophages (AM) were the major source of CXCL16 in the lung. AM expressed mRNA for CXCL16 and released nanogram quantities after adhesion to plastic as shown by RT-PCR, Western blotting and ELISA. Bronchoalveolar lavage (BAL) fluid from all subjects contained large amounts of CXCL16. The full-length CXCL16 was the predominant isoform in AM lysates, supernatants and BAL. This data suggests that CXCR6 and CXCL16 may play a role in T cell recruitment to the lung.