coronavirus systems biological assessment of immunity to … · arunachalam et al., science 369,...

12
RESEARCH ARTICLE CORONAVIRUS Systems biological assessment of immunity to mild versus severe COVID-19 infection in humans Prabhu S. Arunachalam 1 * , Florian Wimmers 1 * , Chris Ka Pun Mok 2 * , Ranawaka A. P. M. Perera 3 * , Madeleine Scott 1,4 , Thomas Hagan 1 , Natalia Sigal 1 , Yupeng Feng 1 , Laurel Bristow 5 , Owen Tak-Yin Tsang 6 , Dhananjay Wagh 7 , John Coller 7 , Kathryn L. Pellegrini 8 , Dmitri Kazmin 1 , Ghina Alaaeddine 5 , Wai Shing Leung 6 , Jacky Man Chun Chan 6 , Thomas Shiu Hong Chik 6 , Chris Yau Chung Choi 6 , Christopher Huerta 5 , Michele Paine McCullough 5 , Huibin Lv 2 , Evan Anderson 9 , Srilatha Edupuganti 5 , Amit A. Upadhyay 8 , Steve E. Bosinger 8,10 , Holden Terry Maecker 1 , Purvesh Khatri 1,4 , Nadine Rouphael 5 , Malik Peiris 2,3 , Bali Pulendran 1,11,12 Coronavirus disease 2019 (COVID-19) represents a global crisis, yet major knowledge gaps remain about human immunity to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We analyzed immune responses in 76 COVID-19 patients and 69 healthy individuals from Hong Kong and Atlanta, Georgia, United States. In the peripheral blood mononuclear cells (PBMCs) of COVID-19 patients, we observed reduced expression of human leukocyte antigen class DR (HLA-DR) and proinflammatory cytokines by myeloid cells as well as impaired mammalian target of rapamycin (mTOR) signaling and interferon-a (IFN-a) production by plasmacytoid dendritic cells. By contrast, we detected enhanced plasma levels of inflammatory mediatorsincluding EN-RAGE, TNFSF14, and oncostatin Mwhich correlated with disease severity and increased bacterial products in plasma. Single-cell transcriptomics revealed a lack of type I IFNs, reduced HLA-DR in the myeloid cells of patients with severe COVID-19, and transient expression of IFN-stimulated genes. This was consistent with bulk PBMC transcriptomics and transient, low IFN-a levels in plasma during infection. These results reveal mechanisms and potential therapeutic targets for COVID-19. T he recent emergence of the severe acute respiratory syndrome coronavirus 2 (SARS- CoV-2) in Wuhan, China, in December 2019 and its rapid international spread caused a global pandemic. Research has moved rapidly in isolating, sequencing, and cloning the virus; developing diagnostic kits; and test- ing candidate vaccines. However, key ques- tions remain about the dynamic interaction between the human immune system and the SARS-CoV-2 virus. Coronavirus disease 2019 (COVID-19) presents with a spectrum of clinical phenotypes, with most patients exhibiting mild to moderate symptoms and 15% of patients progressing, typically within a week, to severe or critical dis- ease that requires hospitalization (1). A mi- nority of those who are hospitalized develop acute respiratory disease syndrome (ARDS) and require mechanical ventilation. Epidemi- ological data so far suggest that COVID-19 has a case fatality rate several times greater than that of seasonal influenza (1). The elderly and individuals with underlying medical comor- bidities such as cardiovascular disease, dia- betes mellitus, chronic lung disease, chronic kidney disease, obesity, hypertension, or can- cer have a much higher mortality rate than healthy young adults (2). The underlying causes of this difference are unknown, but they may be due to an impaired interferon (IFN) response and dysregulated inflammatory responses, as have been observed with other zoonotic corona- virus infections such as severe acute respiratory syndrome (SARS) and Middle East respira- tory syndrome (MERS) (3). Current research is uncovering how the adaptive immune re- sponse to SARS-CoV-2 is induced with optimal functional capacities to clear SARS-CoV-2 viral infection (46). Understanding the immunological mecha- nisms underlying the diverse clinical presen- tations of COVID-19 is a crucial step in the design of rational therapeutic strategies. Re- cent studies have suggested that COVID-19 patients are characterized by lymphopenia and increased numbers of neutrophils (79). Most patients with severe COVID-19 exhibit enhanced levels of proinflammatory cytokines including interleukin-6 (IL-6) and IL-1b as well as MCP-1, IP-10, and granulocyte colony- stimulating factor (G-CSF) in the plasma (10). It has been proposed that high levels of pro- inflammatory cytokines might lead to shock as well as respiratory failure or multiple organ failure, and several trials to assess inflamma- tory mediators are under way (11). However, little is known about the immunological mech- anisms underlying COVID-19 severity and the extent to which they differ from the immune responses to other respiratory viruses. Fur- thermore, the question of whether individuals in different parts of the world respond differ- ently to SARS-CoV-2 remains unknown. In this study, we used a systems biological approach [mass cytometry and single-cell transcriptomics of leukocytes, transcriptomics of bulk periph- eral blood mononuclear cells (PBMCs), and multiplex analysis of cytokines in plasma] to analyze the immune response in 76 COVID-19 patients and 69 age- and sex-matched controls from two geographically distant cohorts. Analysis of peripheral blood leukocytes from COVID-19 patients by mass cytometry COVID-19infected patient samples and sam- ples from age- and sex-matched healthy con- trols were obtained from two independent cohorts: (i) the Princess Margaret Hospital at Hong Kong University and (ii) the Hope Clinic at Emory University in Atlanta, Georgia, United States. Patient characteristics and the different assays performed are shown in Table 1. We used mass cytometry to assess immune responses to SARS-CoV-2 infection in 52 COVID-19 patients, who were confirmed positive for viral RNA by polymerase chain reaction (PCR), and 62 age- and gender-matched healthy controls distrib- uted between the two cohorts. To characterize immune cell phenotypes in PBMCs, we used a phospho-CyTOF panel that includes 22 cell surface markers and 12 intracellular markers against an assortment of kinases and phospho- specific epitopes of signaling molecules and H3K27aca marker of histone modification that drives epigenetic remodeling (12, 13) (table S1). The experimental strategy is described in Fig. 1A. The phospho-CyTOF identified 12 main subtypes of innate and adaptive immune cells in both cohorts, as represented in the t-distributed stochastic neighbor embedding (t-SNE) plots (Fig. 1B). There was a notable increase in the frequency of plasmablast and effector CD8 T cells in all infected individuals (Fig. 1B) in both cohorts, as has been described recently in other studies (6, 8, 14). Of note, the kinetics of the CD8 effector T cell response were prolonged RESEARCH Arunachalam et al., Science 369, 12101220 (2020) 4 September 2020 1 of 11 1 Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA. 2 HKU- Pasteur Research Pole, School of Public Health, HKU Li Ka Shing Faculty of Medicine, The University of Hong Kong (HKU), Hong Kong. 3 Centre of Influenza Research, School of Public Health, HKU Li Ka Shing Faculty of Medicine, HKU, Hong Kong. 4 Center for Biomedical Informatics, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA. 5 Hope Clinic of the Emory Vaccine Center, Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Decatur, GA 30030, USA. 6 Infectious Diseases Centre, Princess Margaret Hospital, Hospital Authority of Hong Kong, Hong Kong. 7 Stanford Functional Genomics Facility, Stanford University School of Medicine, Stanford, CA 94305, USA. 8 Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA 30329, USA. 9 Department of Pediatrics, Division of Infectious Disease, Emory University School of Medicine, Atlanta, GA 30322, USA. 10 Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30329, USA. 11 Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA. 12 Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA. *These authors contributed equally to this work. These authors contributed equally to this work. Corresponding author. Email: [email protected] on June 29, 2021 http://science.sciencemag.org/ Downloaded from

Upload: others

Post on 08-Feb-2021

2 views

Category:

Documents


0 download

TRANSCRIPT

  • RESEARCH ARTICLE◥

    CORONAVIRUS

    Systems biological assessment of immunity to mildversus severe COVID-19 infection in humansPrabhu S. Arunachalam1*, Florian Wimmers1*, Chris Ka Pun Mok2*, Ranawaka A. P. M. Perera3*,Madeleine Scott1,4†, Thomas Hagan1†, Natalia Sigal1†, Yupeng Feng1†, Laurel Bristow5,Owen Tak-Yin Tsang6, Dhananjay Wagh7, John Coller7, Kathryn L. Pellegrini8, Dmitri Kazmin1,Ghina Alaaeddine5, Wai Shing Leung6, Jacky Man Chun Chan6, Thomas Shiu Hong Chik6,Chris Yau Chung Choi6, Christopher Huerta5, Michele Paine McCullough5, Huibin Lv2, Evan Anderson9,Srilatha Edupuganti5, Amit A. Upadhyay8, Steve E. Bosinger8,10, Holden Terry Maecker1,Purvesh Khatri1,4, Nadine Rouphael5, Malik Peiris2,3, Bali Pulendran1,11,12‡

    Coronavirus disease 2019 (COVID-19) represents a global crisis, yet major knowledge gaps remain abouthuman immunity to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We analyzedimmune responses in 76 COVID-19 patients and 69 healthy individuals from Hong Kong and Atlanta,Georgia, United States. In the peripheral blood mononuclear cells (PBMCs) of COVID-19 patients, weobserved reduced expression of human leukocyte antigen class DR (HLA-DR) and proinflammatorycytokines by myeloid cells as well as impaired mammalian target of rapamycin (mTOR) signaling andinterferon-a (IFN-a) production by plasmacytoid dendritic cells. By contrast, we detected enhancedplasma levels of inflammatory mediators—including EN-RAGE, TNFSF14, and oncostatin M—whichcorrelated with disease severity and increased bacterial products in plasma. Single-cell transcriptomicsrevealed a lack of type I IFNs, reduced HLA-DR in the myeloid cells of patients with severe COVID-19,and transient expression of IFN-stimulated genes. This was consistent with bulk PBMC transcriptomics andtransient, low IFN-a levels in plasma during infection. These results reveal mechanisms and potentialtherapeutic targets for COVID-19.

    The recent emergence of the severe acuterespiratory syndrome coronavirus 2 (SARS-CoV-2) inWuhan,China, inDecember2019and its rapid international spread causeda global pandemic. Research has moved

    rapidly in isolating, sequencing, and cloningthe virus; developing diagnostic kits; and test-ing candidate vaccines. However, key ques-tions remain about the dynamic interactionbetween the human immune system and theSARS-CoV-2 virus.

    Coronavirus disease 2019 (COVID-19) presentswith a spectrum of clinical phenotypes, withmost patients exhibiting mild to moderatesymptoms and 15% of patients progressing,typically within a week, to severe or critical dis-ease that requires hospitalization (1). A mi-nority of those who are hospitalized developacute respiratory disease syndrome (ARDS)and require mechanical ventilation. Epidemi-ological data so far suggest that COVID-19 hasa case fatality rate several times greater thanthat of seasonal influenza (1). The elderly andindividuals with underlying medical comor-bidities such as cardiovascular disease, dia-betes mellitus, chronic lung disease, chronickidney disease, obesity, hypertension, or can-cer have a much higher mortality rate thanhealthy young adults (2). The underlying causesof this difference are unknown, but they maybe due to an impaired interferon (IFN) responseand dysregulated inflammatory responses, ashave been observed with other zoonotic corona-virus infections such as severe acute respiratorysyndrome (SARS) and Middle East respira-tory syndrome (MERS) (3). Current research isuncovering how the adaptive immune re-sponse to SARS-CoV-2 is inducedwith optimalfunctional capacities to clear SARS-CoV-2 viralinfection (4–6).Understanding the immunological mecha-

    nisms underlying the diverse clinical presen-tations of COVID-19 is a crucial step in the

    design of rational therapeutic strategies. Re-cent studies have suggested that COVID-19patients are characterized by lymphopeniaand increased numbers of neutrophils (7–9).Most patients with severe COVID-19 exhibitenhanced levels of proinflammatory cytokinesincluding interleukin-6 (IL-6) and IL-1b aswell as MCP-1, IP-10, and granulocyte colony-stimulating factor (G-CSF) in the plasma (10).It has been proposed that high levels of pro-inflammatory cytokines might lead to shockas well as respiratory failure or multiple organfailure, and several trials to assess inflamma-tory mediators are under way (11). However,little is known about the immunological mech-anisms underlying COVID-19 severity and theextent to which they differ from the immuneresponses to other respiratory viruses. Fur-thermore, the question of whether individualsin different parts of the world respond differ-ently to SARS-CoV-2 remains unknown. In thisstudy, we used a systems biological approach[mass cytometry and single-cell transcriptomicsof leukocytes, transcriptomics of bulk periph-eral blood mononuclear cells (PBMCs), andmultiplex analysis of cytokines in plasma] toanalyze the immune response in 76 COVID-19patients and 69 age- and sex-matched controlsfrom two geographically distant cohorts.

    Analysis of peripheral blood leukocytes fromCOVID-19 patients by mass cytometry

    COVID-19–infected patient samples and sam-ples from age- and sex-matched healthy con-trols were obtained from two independentcohorts: (i) the Princess Margaret Hospital atHong Kong University and (ii) the Hope Clinicat EmoryUniversity inAtlanta, Georgia, UnitedStates. Patient characteristics and the differentassays performed are shown in Table 1.We usedmass cytometry to assess immune responses toSARS-CoV-2 infection in 52 COVID-19 patients,who were confirmed positive for viral RNA bypolymerase chain reaction (PCR), and 62 age-and gender-matched healthy controls distrib-uted between the two cohorts. To characterizeimmune cell phenotypes in PBMCs, we used aphospho-CyTOF panel that includes 22 cellsurface markers and 12 intracellular markersagainst an assortment of kinases and phospho-specific epitopes of signaling molecules andH3K27ac—a marker of histone modificationthat drives epigenetic remodeling (12, 13) (tableS1). The experimental strategy is described inFig. 1A. The phospho-CyTOF identified 12mainsubtypes of innate and adaptive immune cells inboth cohorts, as represented in the t-distributedstochastic neighbor embedding (t-SNE) plots(Fig. 1B). There was a notable increase in thefrequency of plasmablast and effector CD8T cells in all infected individuals (Fig. 1B) in bothcohorts, as has been described recently in otherstudies (6, 8, 14). Of note, the kinetics of theCD8 effector T cell response were prolonged

    RESEARCH

    Arunachalam et al., Science 369, 1210–1220 (2020) 4 September 2020 1 of 11

    1Institute for Immunity, Transplantation and Infection, StanfordUniversity School of Medicine, Stanford, CA 94305, USA. 2HKU-Pasteur Research Pole, School of Public Health, HKU Li KaShing Faculty of Medicine, The University of Hong Kong (HKU),Hong Kong. 3Centre of Influenza Research, School of PublicHealth, HKU Li Ka Shing Faculty of Medicine, HKU, Hong Kong.4Center for Biomedical Informatics, Department of Medicine,Stanford University School of Medicine, Stanford, CA 94305,USA. 5Hope Clinic of the Emory Vaccine Center, Department ofMedicine, Division of Infectious Diseases, Emory UniversitySchool of Medicine, Decatur, GA 30030, USA. 6InfectiousDiseases Centre, Princess Margaret Hospital, Hospital Authorityof Hong Kong, Hong Kong. 7Stanford Functional GenomicsFacility, Stanford University School of Medicine, Stanford, CA94305, USA. 8Emory Vaccine Center, Yerkes National PrimateResearch Center, Atlanta, GA 30329, USA. 9Department ofPediatrics, Division of Infectious Disease, Emory UniversitySchool of Medicine, Atlanta, GA 30322, USA. 10Department ofPathology and Laboratory Medicine, Emory University, Atlanta,GA 30329, USA. 11Department of Pathology, Stanford UniversitySchool of Medicine, Stanford, CA 94305, USA. 12Department ofMicrobiology and Immunology, Stanford University School ofMedicine, Stanford, CA 94305, USA.*These authors contributed equally to this work.†These authors contributed equally to this work.‡Corresponding author. Email: [email protected]

    on June 29, 2021

    http://science.sciencemag.org/

    Dow

    nloaded from

    http://science.sciencemag.org/

  • and continued to increase up to day 40 afteronset of the symptoms (fig. S1).We next used manual gating to identify 25

    immune cell subsets (fig. S2) and determinedwhether there were changes in the frequency

    or signaling molecules of innate immune cellpopulations consistent between the two co-horts. There were several differences, but no-tably the frequency of plasmacytoid dendriticcells (pDCs) was significantly reduced in the

    PBMCs of SARS-CoV-2–infected individualsin both cohorts (Fig. 1C). The kinetics of pDCresponse did not show an association with thetime since symptom onset (fig. S1C). Neitherdid the observed changes correlate with the

    Arunachalam et al., Science 369, 1210–1220 (2020) 4 September 2020 2 of 11

    A

    C

    D

    Wilcoxon, p = 0.01Wilcoxon, p = 0.0015

    Atlanta Hong Kong

    Healthy Infected Healthy Infected

    0.03

    0.10

    0.30

    1.00

    Fre

    quen

    cy (

    % o

    f CD

    45 c

    ells

    )

    pDC frequency

    pDC

    pS6 (Ser 235/236)

    Cou

    nt

    Healthy

    Infected

    E

    IKBɑ

    Cou

    nt

    mDC

    Healthy

    Infected

    B

    HealthyMild/moderateSevereICUConvalescent

    HealthyModerateSevereICU

    HealthyModerateSevereICU

    Wilcoxon, p = 0.01

    0.0

    0.5

    1.0

    1.5

    Healthy Infected

    pS6

    (FC

    rel

    ativ

    e to

    hea

    lthy

    cont

    rols

    ) Wilcoxon, p = 4.5e−10

    0

    1

    2

    Healthy Infected

    pS6 (mTOR) levels in pDCs

    Atlanta Hong Kong

    Wilcoxon, p = 0.00021

    0

    1

    2

    Healthy Infected

    IKBα

    (F

    C r

    elat

    ive

    to h

    ealth

    y co

    ntro

    ls) Wilcoxon, p = 2.9e−06

    0

    1

    2

    Healthy Infected

    Atlanta Hong Kong

    IKBα levels in mDCs

    −10

    −5

    0

    5

    10

    NK

    cel

    ls−

    Ikba

    Pla

    smab

    last

    spD

    Cs−

    pCR

    EB

    Effe

    ctor

    CD

    8 T

    cel

    lsE

    ffect

    or C

    D4

    T c

    ells

    NK

    cel

    ls−

    pSTA

    T1

    CD

    4 T

    cel

    ls−

    pSTA

    T1

    NK

    T c

    ells

    −pC

    RE

    BN

    K c

    ells

    −pC

    RE

    BN

    oncl

    assi

    cal m

    onoc

    ytes

    −pE

    RK

    Treg

    s−pS

    TAT

    1In

    term

    edia

    te m

    onoc

    ytes

    −pS

    TAT

    3N

    K c

    ells

    −pS

    TAT

    3C

    D8

    T c

    ells

    −pp

    38Tr

    egs−

    pSTA

    T3

    B c

    ells

    −pS

    TAT

    1In

    term

    edia

    te m

    onoc

    ytes

    −A

    ktC

    D8

    T c

    ells

    −pS

    TAT

    1N

    K c

    ells

    −H

    3K27

    acIn

    term

    edia

    te m

    onoc

    ytes

    −pE

    RK

    mD

    Cs−

    pSTA

    T3

    CD

    8 T

    cel

    ls−

    pCR

    EB

    CD

    4 T

    cel

    ls−

    pSTA

    T3

    NK

    cel

    lsTr

    egs−

    H3K

    27ac

    Pla

    smab

    last

    s−pS

    6pD

    Cs−

    pp38

    mD

    Cs−

    H3K

    27ac

    Cla

    ssic

    al m

    onoc

    ytes

    −H

    3K27

    acC

    D4

    T c

    ells

    −pp

    38Tr

    egs−

    pSTA

    T5

    pDC

    s−Ik

    baN

    oncl

    assi

    cal m

    onoc

    ytes

    −pp

    38P

    lasm

    abla

    sts−

    pCR

    EB

    Non

    clas

    sica

    l mon

    ocyt

    es−

    pSTA

    T3

    CD

    8 T

    cel

    ls−

    pSTA

    T3

    Inte

    rmed

    iate

    mon

    ocyt

    es−

    pp38

    B c

    ells

    −pC

    RE

    BC

    D4

    T c

    ells

    −H

    3K27

    acN

    oncl

    assi

    cal m

    onoc

    ytes

    −pC

    RE

    BIn

    term

    edia

    te m

    onoc

    ytes

    −pS

    TAT

    1C

    D8

    T c

    ells

    −H

    3K27

    acC

    D4

    T c

    ells

    −pS

    TAT

    5N

    K c

    ells

    −pE

    RK

    Pla

    smab

    last

    s−pS

    TAT

    3N

    K c

    ells

    −pp

    38N

    K T

    cel

    ls−

    pp38

    NK

    T c

    ells

    −Ik

    baN

    K T

    cel

    ls−

    pSTA

    T1

    Pla

    smab

    last

    s−H

    3K27

    acIn

    term

    edia

    te m

    onoc

    ytes

    NK

    T c

    ells

    −pS

    TAT

    3C

    D4

    T c

    ells

    −pC

    RE

    BE

    ffect

    or C

    D8

    T c

    ells

    −pp

    38C

    D8

    T c

    ells

    −pS

    TAT

    5P

    lasm

    abla

    sts−

    pp38

    Effe

    ctor

    CD

    8 T

    cel

    ls−

    H3K

    27ac

    Treg

    s−pC

    RE

    BN

    K T

    cel

    ls−

    pSTA

    T5

    Effe

    ctor

    CD

    8 T

    cel

    ls−

    pCR

    EB

    Non

    clas

    sica

    l mon

    ocyt

    espD

    Cs−

    pSTA

    T3

    Cla

    ssic

    al m

    onoc

    ytes

    −pS

    TAT

    1pD

    Cs

    B c

    ells

    −pE

    RK

    CD

    8 T

    cel

    ls−

    Ikba

    Inte

    rmed

    iate

    mon

    ocyt

    es−

    Ikba

    Cla

    ssic

    al m

    onoc

    ytes

    −pP

    LCg2

    CD

    4 T

    cel

    ls−

    Ikba

    pDC

    s−pS

    6E

    ffect

    or C

    D4

    T c

    ells

    −pS

    TAT

    5m

    DC

    s−pC

    RE

    BB

    cel

    lsm

    DC

    s−pP

    LCg2

    Non

    clas

    sica

    l mon

    ocyt

    es−

    pPLC

    g2m

    DC

    s−Ik

    baP

    lasm

    abla

    sts−

    Ikba

    B c

    ells

    −Ik

    baE

    ffect

    or C

    D8

    T c

    ells

    −Ik

    baC

    lass

    ical

    mon

    ocyt

    es−

    Ikba

    Effe

    ctor

    CD

    4 T

    cel

    ls−

    Ikba

    t sta

    tistic

    40 samples from Atlanta cohort100 samples from Hong Kong cohort

    F

    Fig. 1. Mass cytometry analysis of human peripheral blood leukocytesfrom COVID-19 patients. (A) A schematic representation of the experimentalstrategy. PFA, paraformaldehyde. (B) Representation of mass cytometry–identified cell clusters visualized by t-SNE in two-dimensional space. The boxplots on the bottom show frequency of plasmablasts (CD3−, CD20−, CD56−,HLA-DR+, CD14−, CD16−, CD11c−, CD123−, CD19lo, CD27hi, and CD38hi) and effectorCD8 T cells (CD3+, CD8+, CD38hi, and HLA-DRhi) in both cohorts. (C) Frequenciesof pDCs (CD3−, CD20−, CD56−, HLA-DR+, CD14−, CD16−, CD11c−, and CD123+)in healthy and COVID-19–infected individuals in both cohorts. (D and E) Boxplots showing fold change (FC) of pS6 staining in pDCs (D) and IkBa stainingin mDCs (E) relative to the medians of healthy controls. The histograms on

    the right depict representative staining of the same. (F) Distinguishing features[false discovery rate (FDR) < 0.01] through linear modeling analysis of themass cytometry data between healthy and infected subjects. In all box plots, theboxes show median, upper, and lower quartiles. The whiskers show 5th to95th percentiles. Each dot represents an individual sample (healthy: n = 17 and45; infected: n = 19 and 54, for Atlanta and Hong Kong cohorts, respectively).For the t-SNE analysis, n = 34 and 60 for Atlanta and Hong Kong cohorts,respectively. The colors of the dots indicate the severity of clinical disease, asshown in the legends. The differences between the groups were measured byMann-Whitney rank sum test (Wilcoxon, paired = FALSE). The P values depictingsignificance are shown within the box plots.

    RESEARCH | RESEARCH ARTICLEon June 29, 2021

    http://science.sciencemag.org/

    Dow

    nloaded from

    http://science.sciencemag.org/

  • clinical severity of infection (fig. S1). Addi-tionally, there was reduced expression of pS6[(phosphorylated ribosomal protein S6), acanonical target of mammalian target of rapa-mycin (mTOR) activation (15)] in pDCs anddecreased IkBa—an inhibitor of the signalingof the NF-kb transcription factor—in myeloiddendritic cells (mDCs) (Fig. 1, D and E). mTORsignaling is known to mediate the productionof interferon-a (IFN-a) in pDCs (16), whichsuggests that pDCs may be impaired in theircapacity to produce IFN-a in COVID-19 patients.Finally, we used a linear modeling approachto detect features that distinguish healthy frominfected individuals and those that discrimi-nate individuals on the basis of the clinical se-verity of COVID-19. This analysis was performedwith the cohort (Hong Kong or Atlanta) as acovariate to identify only features that wereconsistent across both cohorts. The distinguish-

    ing features between healthy and infectedindividuals are shown in Fig. 1F. These includefrequencies of plasmablast and effector T cellsand the changes in innate immune cells de-scribed above in addition to STAT1 (signaltransducer and activator of transcription 1)and other signaling events in T cells and nat-ural killer (NK) cells. Of note, no features weresignificantly different between clinical sever-ity groups.We further examined the effect of various

    therapeutic interventions on the immune re-sponses using samples from the Hong Kongcohort, in which some patients were treatedwith IFN-b1, corticosteroids, or antivirals. Theinfected individuals, irrespective of the inter-vention, showed an increased plasmablast andeffector CD8 T cell frequency compared withhealthy controls (fig. S3). However, there wasan increased frequency of effector CD8 T cells

    (fig. S3, bottom panel, right column) and de-creased pS6 signal in the pDCs of antiviral-treated individuals (fig. S4).

    COVID-19 results in functional impairmentof blood myeloid cells and pDCs

    Given the earlier findings that mTOR signal-ing in pDCs mediates the production of IFN-ain response to Toll-like receptor (TLR) stimu-lation (16), the reduced expression of pS6 inpDCs suggests that such cells may be impairedin their capacity to produce IFN-a. To test this,we performed ex vivo stimulation of PBMCsfrom healthy or COVID-19–infected individ-uals, using a mixture of synthetic TLR7 andTLR 8 (TLR7/8) and TLR3 ligands, which areknown to be expressed by viruses, and we per-formed an intracellular staining assay to detectcytokine responses. The TLR ligands includedTLR3 and TLR7/8 ligands, polyIC and R848.Consistent with our hypothesis, there was re-duced production of IFN-a in response to theTLR stimuli in the pDCs of infected individu-als compared with those of healthy controls(Fig. 2A). The TNF-a response was also signif-icantly reduced in the pDCs of infected indi-viduals, which demonstrates that the pDCs arefunctionally impaired in COVID-19 infection.We also determined the ability of mDCs andCD14+ monocytes to respond to TLR stimuli.Notably, the response in mDCs as well as thatin monocytes were also significantly lower inresponse to stimulationwith a bacterial ligandcocktail (composed of TLR2, TLR4, and TLR5ligands) or with the viral TLR cocktail (Fig. 2Band fig. S5). Furthermore, the reduced IkBalevels did not translate into enhanced NF-kbsubunit p65 phosphorylation as measuredby p65 (Ser529) in the same cells (Fig. 2C). Theseresults suggest that the innate immune cells inthe periphery of COVID-19–infected individualsare suppressed in their response to TLR stim-ulation, irrespective of the clinical severity.

    Enhanced concentrations of cytokinesand inflammatory mediators in plasmafrom COVID-19 patients

    The impaired cytokine response of myeloidcells and pDCs in response to TLR stimulationwas unexpected and seemingly at odds withthe literature describing an enhanced inflam-matory response in COVID-19–infected indi-viduals. Several studies have described higherplasma levels of cytokines, including but notlimited to IL-6, TNF-a, and CXCL10 (10, 17–19).Therefore, we evaluated cytokines and chemo-kines in plasma samples from the Atlanta co-hort using the Olink multiplex inflammationpanel that measures 92 different cytokinesand chemokines. Of the 92 analytesmeasured,71 proteins were detected within the dynamicrange of the assay. Of these 71 proteins, 43 cy-tokines, including IL-6, MCP-3, and CXCL10,were significantly up-regulated in COVID-19

    Arunachalam et al., Science 369, 1210–1220 (2020) 4 September 2020 3 of 11

    Table 1. Patient characteristics and number of samples used in different assays. NA, notapplicable.

    Characteristics Hong Kong cohort Atlanta cohort

    Number of subjects.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    COVID-19 36 patients* 40 patients*.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    Flu/RSV NA 16 patients.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    Healthy 45 individuals 24 individuals.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    Age.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    COVID-19 [median (range)] 55 (18–80) 56 (25–94).. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    Flu/RSV NA 66 (51–86).. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    Healthy 53 (21–69) 52 (23–91).. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    Gender.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    COVID-19 (male, %) 58% 55%.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    Flu/RSV (male, %) NA 31%.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    Healthy (male, %) 58% 42%.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    Clinical severity of COVID-19 patients.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    Mild/moderate 75% 18%.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    Severe (no ICU) 14% 60%.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    ICU 11% 18%.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    Clinical severity of flu/RSV patients.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    Mild/moderate NA 37.5%.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    Severe (no ICU) NA 37.5%.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    ICU NA 31%.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    Intervention.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    IFN-b1 20% NA.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    Corticosteroids 19% NA.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    Antivirals 61% NA.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    Assays using COVID-19 samples.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    Phospho-CyTOF 54 PBMC samples (36 patients) 19 PBMC samples (16 patients).. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    In vitro stimulation NA 17 PBMC samples (15 patients).. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    Olink proteomics NA 36 plasma samples (29 patients).. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    CITE-seq NA 7 PBMC samples (7 patients).. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    Bulk RNA-seq NA 17 PBMC samples (15 patients).. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    Bacterial products NA 51 plasma samples (40 patients).. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    Assays using flu/RSV samples.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    Phospho-CyTOF NA 4 PBMC samples (4 patients).. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    Olink proteomics NA 19 plasma samples (16 patients).. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    *Some patients have blood from multiple time points.

    RESEARCH | RESEARCH ARTICLEon June 29, 2021

    http://science.sciencemag.org/

    Dow

    nloaded from

    http://science.sciencemag.org/

  • infection (Fig. 3, top row, and fig. S6). Theseresults demonstrate that plasma levels of in-flammatory molecules were significantly up-regulated,despite the impairedcytokine responsein blood myeloid cells and pDCs, which sug-gests a tissue origin of the plasma cytokines.

    In addition to IL-6 and other cytokines de-scribed previously (10), we identified threeproteins that were significantly enhanced inCOVID-19 infection and strongly correlatedwith clinical severity (Fig. 3, bottom row). Thesewere TNFSF14 [LIGHT, a ligand of lympho-

    toxin B receptor that is highly expressed inhuman lung fibroblasts and implicated in lungtissue fibrosis and remodeling and inflam-mation (20)], EN-RAGE [S100A12, a biomarkerof pulmonary injury that is implicated in path-ogenesis of sepsis-induced ARDS (21)], and

    Arunachalam et al., Science 369, 1210–1220 (2020) 4 September 2020 4 of 11

    Fig. 2. Flow cytometry analysis of exvivo stimulated human peripheral bloodleukocytes from COVID-19 patients.(A) Box plots showing the fraction of pDCsin PBMCs of healthy or infected donors(CD3−, CD20−, CD56−, HLA-DR+, CD14−,CD16−, CD11c−, and CD123+) producingIFN-a, TNF-a, or IFN-a + TNF-a inresponse to stimulation with the viralcocktail (polyIC + R848). The contour plotson the right show IFN-a, TNF-a, or IFN-a +TNF-a staining in pDCs. (B) Box plotsshowing the fraction of mDCs in PBMCs ofhealthy or infected donors (CD3−, CD20−,CD56−, HLA-DR+, CD14−, CD16−, CD123+,and CD11c−) producing IL-6, TNF-a, orIL-6 + TNF-a in response to no stimulation(top), the bacterial cocktail (middle;Pam3CSK4, LPS, and Flagellin), or the viralcocktail (bottom; polyIC + R848). Theflow cytometry plots on the right arerepresentative plots gated on mDCsshowing IL-6, TNF-a, or IL-6 + TNF-aresponse. (C) Fold change of NF-kb p65(Ser529) staining in PBMCs stimulatedwith bacterial cocktail relative to nostimulation in healthy and infected donorsto show the reduced induction of p65phosphorylation in infected individuals. Thehistograms show representative flowcytometry plots of p65 staining in mDCs.GeoMFI, geometric mean fluorescenceintensity. In all box plots, the boxes showmedian, upper, and lower quartiles.The whiskers show 5th to 95th percentiles.Each dot represents an Atlanta cohortpatient (n = 14 and 17 for healthy andinfected, respectively). Colors of thedots indicate the severity of clinical dis-ease, as shown in the legends. Thedifferences between the groups weremeasured by Mann-Whitney rank sum test.The P values depicting significance areshown within the box plots.

    Hea

    lthy

    Infe

    cted

    No stimulation Viral cocktail

    0.26% 9.97%

    0% 31.0%

    Wilcoxon, p = 0.003 Wilcoxon, p = 0.0017 Wilcoxon, p = 0.00071

    IFN− TNF− IFN− + TNF−

    Healthy Infected Healthy Infected Healthy Infected

    1

    10

    100

    % o

    f pD

    Cs

    Severity Healthy Moderate Severe ICU

    A Cytokine responses in pDCs

    B Cytokine responses in myeloid DCs

    Wilcoxon, p = 0.018 Wilcoxon, p = 0.00061 Wilcoxon, p = 0.049

    IL−6 TNF− IL−6 + TNF−

    Healthy Infected Healthy Infected Healthy Infected

    0.1

    1.0

    10.0

    % o

    f mD

    Cs

    No stimulation

    Wilcoxon, p = 4.5e−06 Wilcoxon, p = 9.6e−06 Wilcoxon, p = 9.1e−08IL−6 TNF− IL−6 + TNF−

    Healthy Infected Healthy Infected Healthy Infected

    0.1

    1.0

    10.0

    % o

    f mD

    Cs

    Bacterial cocktail

    Viral cocktail

    Wilcoxon, p = 8e−04 Wilcoxon, p = 0.00019 Wilcoxon, p = 0.00069

    IL−6 TNF− IL−6 + TNF−

    Healthy Infected Healthy Infected Healthy Infected

    0.1

    1.0

    10.0

    % o

    f mD

    Cs

    Viral cocktail

    IFN-

    TN

    F-

    Healthy Infected

    No

    stim

    ulat

    ion

    Bac

    teria

    lV

    iral

    IL-6

    TN

    F-

    0.50%

    13.6%

    11.5%

    0.15%

    1.99%

    5.14%

    p65 (Ser529) in mDCs

    Wilcoxon, p = 2.8e−06

    0.0

    0.5

    1.0

    1.5

    2.0

    Healthy Infected

    Fol

    d ch

    ange

    in G

    eoM

    FI o

    f p65

    rel

    ativ

    e to

    n

    o st

    imul

    atio

    n

    Bacterial cocktail

    Hea

    lthy

    No stimulation

    Infe

    cted

    Bacterial

    p65 (Ser529)

    C

    RESEARCH | RESEARCH ARTICLEon June 29, 2021

    http://science.sciencemag.org/

    Dow

    nloaded from

    http://science.sciencemag.org/

  • oncostatin M [(OSM), a regulator of IL-6].Of note, the TNFSF14 is distinctively enhancedin the plasma of COVID-19–infected individualsbut not in cases of other related pulmonaryinfections such as influenza (flu) virus andrespiratory syncytial virus (RSV) (Fig. 3). Giventhe pronounced and unappreciated obser-vations of the enhanced plasma concentra-tions of TNFSF14, EN-RAGE, and OSM andtheir correlation to disease severity, we used anenzyme-linked immunosorbent assay (ELISA)to independently validate these results. Con-sistent with the multiplex Olink analysis, wefound a significant increase of these inflam-matory mediators in the plasma of severe andintensive care unit (ICU) COVID-19 patients.

    Furthermore, we found a correlation betweenmultiplex analysis by Olink and the ELISAresults (fig. S7). These results suggest thatCOVID-19 infection induces a distinctive in-flammatory program characterized by cyto-kines released from tissues (most likely thelungs) but suppression of the innate immunesystem in the periphery. These observationsmay also represent previously unexplored ther-apeutic strategies for intervention against se-vere COVID-19.

    Single-cell transcriptional responseto COVID-19 infection

    To investigate the molecular and cellular pro-cesses that lead to the distinctive inflamma-

    tory program, we used cellular indexing oftranscriptomes and epitopes by sequencing(CITE-seq) and profiled the gene and pro-tein expression in PBMC samples of COVID-19–infected individuals. Cryopreserved PBMC sam-ples from a total of 12 age-matched subjectsin the Atlanta cohort (five healthy controls andseven COVID-19 patients; Table 2) were en-riched for DCs, stained using a cocktail of 36DNA-labeled antibodies (table S2), and analyzedusing droplet-based single-cell gene expres-sion profiling approaches (Fig. 4A). We per-formed the experiment in two batches andobtained transcriptomes for more than 63,000cells after initial preprocessing. Next, we gen-erated a cell-by-gene matrix and conducted

    Arunachalam et al., Science 369, 1210–1220 (2020) 4 September 2020 5 of 11

    Fig. 3. Multiplex analysis of cytokines in theplasma of COVID-19 patients. Cytokine levels inthe plasma of healthy or infected individuals.The infected individuals are further classified onthe basis of the severity of their clinical COVID-19disease. The normalized protein expression valuesplotted on the y axes are arbitrary units defined byOlink Proteomics to represent Olink data. In allbox plots, the boxes show median, upper, and lowerquartiles. The whiskers show 5th to 95th percent-iles. Each dot represents an Atlanta cohortsample (n = 18 healthy, 4 moderate, 18 severe,12 ICU, 2 convalescent, 8 flu, and 11 RSV).The colors of the dots indicate the severity of clinicaldisease, as shown in the legends. The differencesbetween the groups were measured by Mann-Whitneyrank sum test (Wilcoxon, paired = FALSE; *P <0.05; **P < 0.01; ***P < 0.001; ns, not significant).

    ***

    ns

    ****

    **

    ***

    ****

    **

    ns

    ns

    ns

    *

    *

    **ns

    ns

    ns

    *ns

    ns

    ***

    **ns

    OSM TNFSF14 EN−RAGE

    IL−6 TNF−α MCP−3

    Healt

    hy

    Mode

    rate

    Seve

    reIC

    U

    Conv

    alesc

    ent

    Flu RSV

    Healt

    hy

    Mode

    rate

    Seve

    reIC

    U

    Conv

    alesc

    ent

    Flu RSV

    Healt

    hy

    Mode

    rate

    Seve

    reIC

    U

    Conv

    alesc

    ent

    Flu RSV

    Healt

    hy

    Mode

    rate

    Seve

    reIC

    U

    Conv

    alesc

    ent

    Flu RSV

    Healt

    hy

    Mode

    rate

    Seve

    reIC

    U

    Conv

    alesc

    ent

    Flu RSV

    Healt

    hy

    Mode

    rate

    Seve

    reIC

    U

    Conv

    alesc

    ent

    Flu RSV

    0

    4

    8

    12

    0

    5

    10

    0

    2

    4

    0

    3

    6

    9

    0

    5

    10

    0

    5

    10

    15

    Infection severity

    Nor

    mal

    ized

    pro

    tein

    exp

    ress

    ion

    (log2

    )

    Fig. 4. Early, transient ISG expression in COVID-19 infection. (A) A sche-matic representation of DC enrichment strategy used in CITE-seq analysis.scRNA-seq, single-cell RNA-seq. (B) UMAP representation of PBMCs from allanalyzed samples (n = 12), colored by manually annotated cell type. (C) Pairwisecomparison of genes from healthy individuals (n = 5) and COVID-19–infectedpatients (n = 7) was conducted for each cluster. DEGs were analyzed foroverrepresentation of BTMs. The ringplot shows overrepresented pathways inup- and down-regulated genes of each cluster. The heatmap on the right showsthe average expression levels of 33 ISGs derived from the enriched BTMsin different cell clusters of healthy (n = 5) and COVID-19 subjects (n = 7).(D) UMAP representation of PBMCs from all analyzed samples showing the

    expression levels of selected IFNs and ISGs. (E) Kinetics of circulating IFN-alevels (picograms per milliliter) in plasma measured using SIMoA technology(n = 18 healthy and 40 COVID-19–infected patients). (F) Correlation betweencirculating IFN-a levels in plasma and the average expression of ISGs measuredby CITE-seq analysis. (G) Hierarchically clustered heatmap of the expression ofthe CITE-seq ISG signature (C) in the bulk RNA-seq dataset, performed usingan extended group of subjects (n = 17 healthy and 17 COVID-19–infectedsamples). Colors represent gene-wise z scores. (H) Bar chart representingthe proportion of variance in CITE-seq ISG signature expression explained bythe covariates in the x axis through principal variance component analysis(PVCA). resid, xxxxxxx.

    RESEARCH | RESEARCH ARTICLEon June 29, 2021

    http://science.sciencemag.org/

    Dow

    nloaded from

    http://science.sciencemag.org/

  • Arunachalam et al., Science 369, 1210–1220 (2020) 4 September 2020 6 of 11

    Top BTMs enriched in DEGs COVID-19 (n=7) vs healthy (n=5) for each cluster

    A

    C

    D

    F G H

    E

    B

    1

    10

    100

    1000

    10000

    0 20 40 60Days post symptom onset

    Con

    cent

    ratio

    n (f

    g/m

    l)

    SeverityHealthyModerateSevereICU

    Pro

    port

    ion

    varia

    nce

    expl

    aine

    d

    0.0

    0.2

    0.4

    0.6

    0.8

    1.0

    Age

    Sev

    erity

    Tim

    e

    Gen

    der

    resi

    d

    TAP

    1IF

    ITM

    1IF

    IT2

    STAT

    1IF

    IT1

    IFIT

    3D

    DX

    60IF

    IH1

    DD

    X58

    OA

    SL

    PM

    LR

    SA

    D2

    SP

    100H

    ES

    X1

    HE

    RC

    5U

    SP

    18E

    IF2A

    K2

    PLS

    CR

    1O

    AS

    1IR

    F7

    SE

    RP

    ING

    1PA

    RP

    9M

    X2

    IFI27

    AP

    OB

    EC

    3AO

    AS

    3S

    IGLE

    C1

    ATF

    3C

    1QB

    BC

    L2A1

    IFN

    GR

    2M

    GS

    T1

    HLX

    Severity

    Days sym

    pto

    ms

    Days postsymptom onset

    20

    5

    SeverityModerateSevereICU

    −3

    −2

    −1

    0

    1

    2

    3z-score

    antigen processing and presentation (M200)enriched in antigen presentation (I) (M71)

    reg. of antigen presentation and immune response (M5.0)Monocyte surface signature (S4)

    TLR and inflammatory signaling (M16)immune activ ric cluster (M37.0)enriched in antigen presentation (II) (M95.0)

    enriched in T cells (I) (M7.0)cell cycle and transcription (M4.0)

    antiviral IFN signature (M75)enriched in activated dendritic cells (II) (M165)

    type I interferon response (M127)innate antiviral response (M150)

    0

    10

    20

    Log of 1/P val

    % Path is DEG

    0.3

    0.6

    0.9

    A

    IFN signaling

    Antigen presentation

    Cor: 0.85

    5

    10

    15

    20

    0.0

    2.5

    5.0

    7.5

    10.0

    Log of Concentration, fg/ml

    33 g

    ene

    ISG

    Sco

    re new_idcov1cov2cov3cov4cov5cov6cov7hd2hd3hd4hd5

    IFN-α ELISA of plasma

    CITE-seq vs IFN-α ELISA Bulk RNA-seq of total PBMCs Bulk RNA-seq of total PBMCs

    TAP

    1IR

    F7

    US

    P18

    HE

    SX

    1O

    AS

    1E

    IF2A

    K2

    HE

    RC

    5IF

    IT2

    SE

    RP

    ING

    1B

    CL2A

    1IF

    NG

    R2

    MG

    ST

    1A

    PO

    BE

    C3A

    STA

    T1

    IFI27

    DD

    X60

    SP

    100R

    SA

    D2

    IFIT

    3IF

    IT1

    C1Q

    BO

    AS

    LS

    IGLE

    C1

    OA

    S3

    HLX

    DD

    X58

    IFIH

    1M

    X2

    PM

    LP

    LSC

    R1

    PA

    RP

    9IF

    ITM

    1AT

    F3

    gro

    up

    celltype

    0.51.0

    average

    exp p

    ersu

    bject

    Expr. z−score

    −2−1012

    groupCOVID−19Healthy

    celltype

    C10−PDCC11−C MONOC16−C MONO

    C3−C MONOC5−CDC2

    CITE-seq of COVID-19 (n=7) and healthy (n=5) subjects

    CITE-seq of COVID-19 (n=7) and healthy (n=5) subjects

    CITE-seq of COVID-19 (n=7) and healthy (n=5) subjectsExperiment layout

    Mixing ~1:2 and CITE-seq

    Total PBMCs

    Frozen PBMCs

    Magnetic enrichment of dendritic cellsSubjects

    7 COVID-19 infected5 healthy control

    RESEARCH | RESEARCH ARTICLEon June 29, 2021

    http://science.sciencemag.org/

    Dow

    nloaded from

    http://science.sciencemag.org/

  • dimensionality reduction through uniformmanifold approximation and projection (UMAP)and graph-based clustering. Analysis of celldistribution within the UMAP between exper-iments revealed no major differences, and weanalyzed the datasets from the two experimentstogetherwithout batch correction (fig. S8).Next,we calculated the per-cell quality control (QC)metrics (fig. S9), differentially expressed genes(DEGs) in each cluster compared with all othercells (fig. S10 and table S4), and the abundanceof DNA-labeled antibodies in each cell (fig. S11).Using this information, we filtered low-qualitycells andmanually annotated the clusters. AfterQC and cluster annotation, we retained a finaldataset with 57,669 high-quality transcriptomesand a median of ~4781 cells per sample and1803 individual genes per cell that we used toconstruct the single-cell immune cell landscapeof COVID-19 (Fig. 4B).We observed several clusters that were pri-

    marily identified in COVID-19–infected indi-viduals, including a population of plasmablasts,platelets, and red blood cells and several pop-ulations of granulocytes. Notably, we detectedclusters of T cells and monocytes that werecharacterized by the expression of interferon-stimulated genes (ISGs) such as IFI27, IFITM3,or ISG15 (see C11-C MONO_IFN and C18-T_IFNin fig. S10). These IFN response–enriched clus-ters emerged only in samples from COVID-19patients (fig. S12).To describe the specific transcriptional state

    of single cells from COVID-19–infected indi-viduals, we determined the DEGs for cells fromall COVID-19–infected samples in a given clus-ter compared with the cells from all healthyindividuals in the same cluster. We then an-alyzed these DEGs with overrepresentationanalysis using blood transcriptional modules(BTMs) (22) to better understand which im-mune pathways are differentially regulated inpatientswithCOVID-19 comparedwith healthy

    individuals (Fig. 4C and fig. S13). The analysisindicated amarked induction of antiviral BTMs,especially in cell types belonging to the mye-loid and dendritic cell lineage. Detailed anal-ysis of the expression pattern of the distinctunion of genes driving the enrichment of theseantiviral pathways inmonocytes and dendriticcells revealed thatmany ISGswereup-regulatedin these cell types (Fig. 4C, heatmap). Givenour observations of muted IFN-a productionin pDCs (Fig. 2A), we investigated the expres-sion of genes encoding various type I and typeII IFNs across cell types (Fig. 4D and fig. S14).Notably, with the exception of modest levelsof IFN-g expression in T and NK cells, we couldnot detect any expression of IFN-a and -b genes,which is consistent with the functional datademonstrating impaired type I IFN produc-tion by pDCs and myeloid cells (Fig. 2). How-ever, there was an enhanced expression of ISGsin patients with COVID-19 (Fig. 4D) in spite ofan impaired capacity of the innate cells in theblood compartment to produce these cytokines.Despite the lack of type I IFN gene expres-

    sion, the presence of an ISG signature in thePBMCsof COVID-19–infected individuals raisedthe possibility that low quantities of type I IFNsproduced in the lung by SARS-CoV-2 infection(17) might circulate in the plasma and inducethe expression of ISGs in PBMCs.We thusmea-sured the concentration of IFN-a in plasmausing a highly sensitive ELISA enabled by singlemolecule array (SIMoA) technology. We ob-served a marked increase in the concentra-tion of IFN-a, which peaked around day 8 afteronset of symptoms and regressed to baselinelevels byday 20 (Fig. 4E).Notably,we observed astrong correlation between the average ex-pression levels of the ISG signature in PBMCsidentified by CITE-seq analysis and the IFN-aconcentration in plasma (Fig. 4F). Addition-ally, we noticed a strong temporal dependenceof the IFN-a response.

    To investigate this further and to indepen-dently validate the observations in the CITE-seqanalysis, we performed bulk RNA sequenc-ing (RNA-seq) analysis of PBMCs in an ex-tended group of subjects (17 COVID-19 patientsand 17 healthy controls) from the same cohort.We first evaluated whether the ISG signaturecontaining 33 genes identified in the CITE-seqdata was also observed in the bulk RNA-seqdataset. We observed a strong induction of theISGs in COVID-19 subjects compared withhealthy donors in this dataset as well (Fig. 4G).Of note, we did not detect expression of genesencoding IFN-a or IFN-b, consistent with theCITE-seq and flow cytometry experiments (Fig.4D and Fig. 2, respectively). We also performedan unbiased analysis of an extended set of genesin the IFN transcriptional network (23) andfound that these were induced in COVID-19subjects relative to healthy controls, as observedfor the limited ISG signature (fig. S15A). Sim-ilar to the observation with CITE-seq data (Fig.4F), there was a strong correlation between cir-culating IFN-a and the ISG response measuredby the bulk transcriptomics (fig. S15B). Addi-tionally, we analyzed the individual impact ofmajor covariates—time, disease severity, sex, andage—on the observed ISG signature. Althoughtime emerged as the primary driver of ISG sig-nature, COVID-19 clinical severity also had aneffect (Fig. 4H and fig. S15C). Taken together,these data demonstrate that, early during SARS-CoV-2 infection, there are low levels of circulat-ing IFN-a that induce ISGs in the peripherywhile the innate immune cells in the periph-ery are impaired in their capacity to produceinflammatory cytokines.In addition to an enhanced ISG signature,

    the CITE-seq analysis revealed a significantdecrease in the expression of genes involved inthe antigen-presentation pathways in myeloidcells (Fig. 4C and fig. S13). Consistentwith this,we observed a reduction in the expression ofthe proteins CD86 and human leukocyte anti-gen class DR (HLA-DR) on monocytes andmDCs of COVID-19 patients, which wasmostpronounced in subjects with severe COVID-19infection (Fig. 5A and fig. S16A). HLA-DR is animportant mediator of antigen presentationand is crucial for the induction of T helper cellresponses. Using the phospho-CyTOF data fromboth the Atlanta and Hong Kong cohorts, weconfirmed the reduced expression of HLA-DRonmonocytes andmDCs in patientswith severeCOVID-19 disease (Fig. 5B). By contrast, S100A12,the gene encoding EN-RAGE, was substantiallyincreased in the PBMCs of COVID-19 patients,whereas the expression of genes encoding otherproinflammatory cytokines was either absent orunchanged compared with healthy controls(Fig. 5C and fig. S16B). Notably, the S100A12expression was highly restricted to monocyteclusters (Fig. 5D) and showed a significant cor-relation with EN-RAGE protein levels in plasma

    Arunachalam et al., Science 369, 1210–1220 (2020) 4 September 2020 7 of 11

    Table 2. Detailed characteristics of patient samples used in the CITE-seq analysis. Dashesindicate that the information is not applicable. dec., deceased; F, female; M, male; B, Black; W, white.

    ID Infection Response ICU Day Age Sex Ethnicity

    cov1 COVID-19 Severe, dec. Y 15 60 F B.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    cov2 COVID-19 Severe N 15 75 F W.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    cov3 COVID-19 Severe N 16 59 M B.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    cov4 COVID-19 Severe N 8 48 M B.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    cov5 COVID-19 Moderate N 9 53 F B.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    cov6 COVID-19 Moderate N 2 75 F W.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    cov7 COVID-19 Moderate N 9 47 F B.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    hd1 Healthy – – – 84 F W.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    hd2 Healthy – – – 68 F W.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    hd3 Healthy – – – 38 M W.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    hd4 Healthy – – – 90 M W.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    hd5 Healthy – – – 70 F W.. .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... .. ... ... .. ... ... .. ... ... .. .

    RESEARCH | RESEARCH ARTICLEon June 29, 2021

    http://science.sciencemag.org/

    Dow

    nloaded from

    http://science.sciencemag.org/

  • measured by Olink (Fig. 5E). Finally, we exam-ined whether there is an association betweenHLA-DR and S100A12 expression in our data-set, and we found a strong inverse correlationbetween S100A12 gene expression and thegenes encoding the antigen presentation ma-chinery (HLA-DPA1, HLA-DPB1, HLA-DR, andCD74) (Fig. 5F and fig. S17). Notably, the re-ceptor for S100A12, AGER (RAGE), was ex-pressed sparsely in PBMCs (fig. S18), whichsuggests that the target of EN-RAGE actionwas likely to be elsewhere—perhaps the lung,where RAGE is known to be expressed in type

    I alveolar epithelial cells and mediate inflam-mation (24).Taken together, CITE-seq analysis of PBMCs

    in COVID-19 patients revealed the followingmechanistic insights: (i) a lack of expression ofgenes encoding type I IFN and proinflamma-tory cytokines in PBMCs, which was consistentwith the mass cytometry (Fig. 1C) and func-tional data (Fig. 2); (ii) an early but transientwave of ISG expression, which was entirelyconsistent with analysis of RNA-seq from bulkPBMCs (Fig. 4G and fig. S15A) and stronglycorrelated with an early burst of plasma IFN-a

    (Fig. 4F), likely of lung origin (17); and (iii) theimpaired expression ofHLA-DR and CD86 butenhanced expression of S100A12 in myeloidcells, which was consistent with the mass cy-tometry (Fig. 5B), Olink (Fig. 3), and ELISA(fig. S7) data, and is a phenotype reminiscentof myeloid-derived suppressor cells describedpreviously (25).

    Severe COVID-19 infection is associated withthe systemic release of bacterial products

    The increased levels of proinflammatory me-diators in the plasma—including IL-6, TNF,

    Arunachalam et al., Science 369, 1210–1220 (2020) 4 September 2020 8 of 11

    mDC C mono

    Healt

    hy

    Mod

    erat

    e

    Seve

    re

    Healt

    hy

    Mod

    erat

    e

    Seve

    re

    2.5

    3.0

    3.5

    4.0

    4.5

    5.0M

    FI H

    LA−

    DR

    (lo

    g10)

    Atlanta HK

    Healt

    hy

    Mod

    erat

    e

    Seve

    re

    Healt

    hy

    Mod

    erat

    e

    Seve

    re

    1.5

    2.0

    2.5

    3.0

    MF

    I HLA

    −D

    R (

    log1

    0)

    Atlanta HK

    Healt

    hy

    Mod

    erat

    e

    Seve

    re

    Healt

    hy

    Mod

    erat

    e

    Seve

    re

    1.5

    2.0

    2.5

    3.0

    3.5

    MF

    I HLA

    −D

    R (

    log1

    0)

    A B CYTOF: mDC HLA-DR levels CYTOF: C mono HLA-DR levelsFlow: HLA-DR levels (CITE-seq samples)

    ******

    *

    ******

    0.20***

    0.16*

    D F

    Exp lvl

    E S100A12 CITE-seq vs EN-RAGE plasma levels

    4

    6

    8

    Rel

    . Exp

    ress

    ion

    IL6

    8

    10

    12

    Rel

    . Exp

    ress

    ion

    TNF

    10.0

    12.5

    15.0

    17.5

    Rel

    . Exp

    ress

    ion

    S100A12

    6

    7

    8

    9

    Rel

    . Exp

    ress

    ion

    TNFSF14

    8

    9

    10

    11

    12

    Rel

    . Exp

    ress

    ion

    SeverityHealthyModerateSevereICU

    OSMC Bulk RNA-seq of total PBMCs

    Cor = 0.7pval = 0.016

    2.5

    5.0

    7.5

    10.0

    12.5

    1 2 3 4S100A12

    EN

    −R

    AG

    E

    diseaseCOVID−19

    Healthy

    CITE-seq CITE-seq, myeloid cells

    Fig. 5. Attenuated inflammatory response in peripheral innate immunecells from COVID-19 patients. (A) Flow cytometry analysis of PBMCs analyzedin parallel to the CITE-seq experiment. The log10 median fluorescence intensity(MFI) of HLA-DR expression is shown. (B) Median intensity of HLA-DR expressionin the phospho-CyTOF experiment from Fig. 1. Squares represent individualsamples [Hong Kong (HK): healthy = 30, moderate = 15, and severe = 10; andAtlanta: healthy = 17, moderate = 4, and severe = 13]. The boxes indicate median,upper, and lower quartiles. The whisker length equals 1.5 times the interquartilerange. (C) Relative (Rel.) expression of genes encoding different cytokines in the

    bulk RNA-seq dataset. The boxes show median, upper, and lower quartiles,and the whiskers show 5th to 95th percentiles. (D) UMAP representation ofS100A12 expression in PBMCs from all samples analyzed by CITE-seq.(E and F) Correlation (Cor) analysis of S100A12 expression in cells from myeloidand dendritic cell clusters (C MONO_1, NC MONO, CDC2, PDC, C MONO_IFN,C MONO_2, and C MONO_3) with EN-RAGE levels in plasma (E) or HLA-DPA1expression in the same clusters (F) (n = 5 healthy and 7 COVID-19 subjects). Thestatistical significance between the groups in (B) and (C) was determined bytwo-sided Mann-Whitney rank-sum test; *P < 0.05; **P < 0.01; ***P < 0.001.

    RESEARCH | RESEARCH ARTICLEon June 29, 2021

    http://science.sciencemag.org/

    Dow

    nloaded from

    http://science.sciencemag.org/

  • TNFSF14, EN-RAGE, andOSM(Fig. 3)—coupledwith suppressed innate immune responsesin blood monocytes and DCs (Fig. 2 and fig.S5) suggested a sepsis-like clinical condition(26, 27). In this context, it has been previouslysuggested that proinflammatory cytokinesand bacterial products in the plasmamay playpathogenic roles in sepsis, and the combina-tion of these factors could be important indetermining patient survival (28, 29). There-fore, to determine whether a similar mech-anism could be at play in patients with severeCOVID-19, we measured bacterial DNA andlipopolysaccharide (LPS) in the plasma. Nota-bly, the plasma of severe and ICU patients hadsignificantly higher levels of bacterial DNA,as measured by PCR quantitation of bacterial16S ribosomal RNA (rRNA) gene product, andof LPS, as measured by a TLR4-based reporterassay (Fig. 6, A and B). Furthermore, there wasa significant correlation between bacterial DNAor LPS and the plasma levels of the inflamma-tory mediators IL-6, TNF, MCP-3, EN-RAGE,TNFSF14, andOSM (Fig. 6C and fig. S19). Theseresults suggest that the enhanced cytokine re-

    lease may in part be caused by increased bacte-rial products in the lung or in other tissues.

    Discussion

    We used a systems biology approach to deter-mine host immune responses to COVID-19.Mass cytometry analysis of peripheral bloodleukocytes from two independent cohorts re-vealed several common features of immuneresponses induced upon SARS-CoV-2 infec-tion. There was a notable and protracted in-crease in the frequencies of plasmablasts andeffector CD8 T cells in the peripheral blood,consistent with recent studies (6, 8, 14). Nota-bly, the effector T cells continued to increaseup to day 40 after symptom onset. Studieshave shown that SARS-CoV-2 infection indu-ces exhaustion and apoptosis in T cells (30, 31).Whether the continuing effector CD8 T cellresponse reflects continuous exposure to anti-gen and whether the cells are exhausted willrequire further investigation.In contrast to robust activation of B and T

    cells, we observed a significant decrease in thefrequency of pDCs. Furthermore, mTOR sig-

    naling in pDCs was reduced significantly inCOVID-19–infected individuals, as measuredby decreased pS6 signaling by mass cytome-try. These results suggest that pDCs, the pri-mary producers of type I IFNs, are impaired inCOVID-19 infection, which is consistent withstudies in SARS-CoV infection (32). To deter-mine whether the reduced mTOR signaling inpDCs resulted in impairment of type I IFN pro-duction, we stimulated cells in vitro with TLRligands. Our results demonstrate that pDCsfrom COVID-19–infected patients are func-tionally impaired in their capacity to produceIFN-a in response to TLR stimulation. Takentogether, these data suggest that COVID-19causes an impaired type I IFN response inthe periphery. Administration of type I IFNhas been proposed as a strategy for COVID-19intervention (33); however, it must be notedthat type I IFN signaling has been shown toelevate angiotensin-converting enzyme 2 (ACE2)expression (34) in lung cells, which can po-tentially lead to enhanced infection.In addition to the impaired IFN-a produc-

    tion by pDCs, there was a marked diminution

    Arunachalam et al., Science 369, 1210–1220 (2020) 4 September 2020 9 of 11

    R = 0.39 , p = 0.0046

    R = 0.49 , p = 0.00027

    R = 0.28 , p = 0.044

    R = 0.45 , p = 0.00083

    R = 0.45 , p = 0.00076

    R = 0.45 , p = 0.00087

    EN−RAGE OSM TNFSF14

    IL−6 TNF MCP−3

    1000 2000 1000 2000 1000 2000

    1000 2000 1000 2000 1000 2000

    2.5

    5.0

    7.5

    2

    4

    6

    1

    2

    3

    4

    2.5

    5.0

    7.5

    10.0

    2.5

    5.0

    7.5

    10.0

    2.5

    5.0

    7.5

    10.0

    Bacterial DNA (copies/ml plasma)

    Cyt

    okin

    e re

    spon

    se (

    NP

    X)

    ns

    ***

    ns

    1000

    2000

    3000

    Healt

    hy

    Mod

    erat

    e

    Seve

    re/IC

    U

    Cop

    ies/

    ml p

    lasm

    a

    qPCR (Bacterial 16S rRNA gene)

    7500

    ns

    ****

    ns

    0

    5

    10

    15

    20

    Healt

    hy

    Mod

    erat

    e

    Seve

    re/IC

    U

    ng/m

    l

    LPS in plasma

    SeverityHealthyModerate

    SevereICU

    B

    CASeverity Healthy Moderate Severe ICU

    Fig. 6. Systemic release of microbial products in severe COVID-19 infection. (A and B) Box plots showing bacterial16S rRNA gene (A) and LPS (B) measured in the plasma of healthy or infected individuals. qPCR, quantitative PCR.(C) Spearman’s correlation between cytokines and bacterial DNA measured in plasma. Each dot represents a sample(n = 18 and 51 for healthy and infected, respectively). The colors of the dots indicate the severity of clinical disease, asshown in the legends. The boxes show median, upper, and lower quartiles in the box plots. The whiskers show 5th to95th percentiles. The differences between the groups were measured by Mann-Whitney rank sum test; ***P < 0.001;****P < 0.0001. NPX, normalized protein expression units; R, correlation coefficient.

    RESEARCH | RESEARCH ARTICLEon June 29, 2021

    http://science.sciencemag.org/

    Dow

    nloaded from

    http://science.sciencemag.org/

  • of the proinflammatory cytokines IL-6, TNF-a,and IL-1b produced by monocytes and mDCsupon TLR stimulation (Fig. 2B). This was con-sistent with the lack of or diminished expres-sion of the genes encoding IL-6 and TNF inthe CITE-seq analysis (Fig. 5C). These resultssuggest an impaired innate response in bloodleukocytes of patients with COVID-19. Thisconcept was further supported by the CyTOFand flow cytometry data that showed decreasedHLA-DR and CD86 expression, respectively,inmyeloid cells (Fig. 5, D and E, and fig. S16).To obtain deeper insight into the mechanismsof host immunity to SARS-CoV-2, we performedCITE-seq single-cell RNA-seq and bulkRNA-seqanalysis in COVID-19 patients at various stagesof clinical severity. Our data demonstrate thatSARS-CoV-2 infection results in an early waveof IFN-a in the circulation that induces an ISGsignature. Although the ISG signature showsa strong temporal dependence in our data-sets, we also find that the ISG signature isstrongly induced in patients with moderateCOVID-19 infection (Fig. 4G). Consistent withthis, Hadjadj et al. (5) have reported an en-hanced expression of ISGs in patients withmoderate disease compared with those withsevere or critical disease. Taken together, thesedata suggest that SARS-CoV-2 infection inducesan early, transient type I IFN production in thelungs that induces ISGs in the peripheral blood,primarily in patients with mild or moderatedisease. Additionally, we observed reducedexpression of genes encoding proinflamma-tory cytokines, as well as HLA-DR expressioninmyeloid cells, whichwas consistentwith theCyTOF and flow cytometry data showing re-duced HLA-DR and CD86 expression, respec-tively, in myeloid cells.Our multiplex analysis of plasma cytokines

    revealed enhanced levels of several proin-flammatory cytokines, as has been observedpreviously (35), and revealed a strong associ-ation of the inflammatory mediators EN-RAGE,TNFSF14, and OSM with the clinical severityof the disease. Notably, the expression of genesencoding both TNFSF14 and OSM were down-regulated in the PBMCs from COVID-19 pa-tients with severe disease in the analysis ofCITE-seq data (Fig. 5C), which suggests a tis-sue origin for these cytokines. The gene en-coding EN-RAGE, however, was expressed athigh levels in blood myeloid cells in patientswith severe COVID-19 (Fig. 5, C to F) (althoughit is also possible that EN-RAGE is expressedin the lungs too). Of note, these three cytokineshave been associated with lung inflamma-tory diseases. In particular, EN-RAGE has beenshown to be expressed by CD14+ HLA-DRlo

    cells, the myeloid-derived suppressor cells, andit is a marker of inflammation in severe sepsis(21, 25, 36). Additionally, its receptor, RAGE,is highly expressed in type I alveolar cells inthe lung (24). Notably, we observed that the

    classical monocytes and myeloid cells fromsevere COVID-19 patients in the single-cellRNA-seq data expressed high levels of S100A12,the gene encoding EN-RAGE, but not the typ-ical inflammatory molecules IL-6 and TNF-a.These data suggest that the proinflammatorycytokines observed in plasma likely originatefrom the cells in lung tissue rather than fromperipheral blood cells. Taken together withthe mass cytometry data, the plasma cytokinedata may be utilized to construct an immu-nological profile that discriminates betweensevere versus moderate COVID-19 disease(fig. S20).These results suggest that SARS-CoV-2 infec-

    tion results in a spatial dichotomy in the innateimmune response, characterized by suppres-sion of peripheral innate immunity in the faceof proinflammatory responses that have beenreported in the lungs (37). Furthermore, thereis a temporal shift in the cytokine responsefrom an early but transient type I IFN responseto a proinflammatory response during the laterandmore severe stages, which is similar to thatobserved with other diseases such as influenza(38). Notably, there were enhanced levels ofbacterial DNA and LPS in the plasma, whichwere positively correlated with the plasmalevels of EN-RAGE, TNFSF14, OSM, and IL-6,which suggests a role for bacterial products—perhaps of lung origin—in augmenting the pro-duction of inflammatory cytokines in severeCOVID-19. The biological consequence of theimpaired innate response in peripheral bloodis unknown but may reflect a homeostaticmechanism to prevent rampant systemic hyper-activation, in the face of tissue inflammation.Finally, these results highlight molecules suchas EN-RAGE or TNFSF14, and their receptors,which could represent attractive therapeutictargets against COVID-19.

    REFERENCES AND NOTES

    1. Z. Wu, J. M. McGoogan, JAMA 323, 1239–1242 (2020).2. CDC COVID-19 Response Team, MMWR Morb. Mortal. Wkly. Rep.

    69, 382–386 (2020).3. R. Channappanavar, S. Perlman, Semin. Immunopathol. 39,

    529–539 (2017).4. A. Grifoni et al., Cell 181, 1489–1501.e15 (2020).5. J. Hadjadj et al., Science 369, 718–724 (2020).6. D. Mathew et al., Science 369, eabc8511 (2020).7. C. Wu et al., JAMA Intern. Med. 180, 934–943 (2020).8. L. Kuri-Cervantes et al., Sci. Immunol. 5, eabd7114 (2020).9. L. Tan et al., Signal Transduct. Target. Ther. 5, 33 (2020).10. C. Huang et al., Lancet 395, 497–506 (2020).11. E. J. Giamarellos-Bourboulis et al., Cell Host Microbe 27,

    992–1000.e3 (2020).12. P. Cheung et al., Cell 173, 1385–1397.e14 (2018).13. R. Fernandez, H. Maecker, Biol. Protoc. 5, e1496

    (2015).14. A. J. Wilk et al., Nat. Med. 26, 1070–1076 (2020).15. A. Arif, J. Jia, B. Willard, X. Li, P. L. Fox, Mol. Cell 73, 446–457.e6

    (2019).16. W. Cao et al., Nat. Immunol. 9, 1157–1164 (2008).17. D. Blanco-Melo et al., Cell 181, 1036–1045.e9 (2020).18. P. A. Mudd et al., medRxiv 2020.05.28.20115667

    [Preprint]. 30 May 2020. https://doi.org/10.1101/2020.05.28.20115667.

    19. C. Lucas et al., Nature 10.1038/s41586-020-2588-y(2020).

    20. R. da Silva Antunes, A. K. Mehta, L. Madge, J. Tocker, M. Croft,Front. Immunol. 9, 576 (2018).

    21. Z. Zhang, N. Han, Y. Shen, Mol. Immunol. 122, 38–48(2020).

    22. S. Li et al., Nat. Immunol. 15, 195–204 (2014).23. S. Mostafavi et al., Cell 164, 564–578 (2016).24. E. A. Oczypok, T. N. Perkins, T. D. Oury, Paediatr. Respir. Rev.

    23, 40–49 (2017).25. F. Zhao et al., Immunology 136, 176–183 (2012).26. T. van der Poll, F. L. van de Veerdonk, B. P. Scicluna,

    M. G. Netea, Nat. Rev. Immunol. 17, 407–420 (2017).27. N. Watanabe et al., PLOS ONE 13, e0202049 (2018).28. L. C. Casey, R. A. Balk, R. C. Bone, Ann. Intern. Med. 119,

    771–778 (1993).29. S. M. Opal et al., J. Infect. Dis. 180, 1584–1589 (1999).30. M. Zheng et al., Cell. Mol. Immunol. 17, 533–535 (2020).31. B. Diao et al., Front. Immunol. 11, 827 (2020).32. R. Channappanavar et al., Cell Host Microbe 19, 181–193

    (2016).33. E. Sallard, F. X. Lescure, Y. Yazdanpanah, F. Mentre,

    N. Peiffer-Smadja, Antiviral Res. 178, 104791 (2020).34. C. G. K. Ziegler et al., Cell 181, 1016–1035.e19 (2020).35. G. Chen et al., J. Clin. Invest. 130, 2620–2629 (2020).36. O. M. Pena et al., EBioMedicine 1, 64–71 (2014).37. M. Liao et al., Nat. Med. 26, 842–844 (2020).38. J. Dunning et al., Nat. Immunol. 19, 625–635 (2018).

    ACKNOWLEDGMENTS

    We thank all participants as well as the Hope Clinic and EmoryChildren Center staff and faculty. We particularly acknowledgeK. Hellmeister, A. Kay, A. Cheng, J. Traenkner, A. M. Drobeniuc,H. Macenczak, N. McNair, Y. Saklawi, A. Mehta, M. Bower,T. Girmay, E. Butler, T. Sirajud-Deen, H. Huston, D. Kleinhenz,L. Hussaini, E. Scherer, B. Johnson, J. Kleinhenz, J. Morales,V.Karmali, Y. Xu, and D. Wang. We are grateful for the support ofthe Emory Department of Medicine and Pediatrics and theGeorgia Research Alliance. We are thankful to the Human ImmuneMonitoring Center (HIMC) for assisting with sample shipments.We thank G. Kim and M. Blanco from the Stanford FunctionalGenomics Facility at Stanford University for assistance with single-cell RNA-seq and the Yerkes Nonhuman Primate (NHP) GenomicsCore [supported in part by National Institutes of Health (NIH) grantP51 OD011132]. We thank the HIMC and the Parker Institute forCancer Immunotherapy (PICI) for maintenance and access to theflow cytometer. We acknowledge the support of the clinicianswho facilitated this study, including J. Y. H. Chan, D. P.-L. Lau, andY. M. Ho, and the dedicated clinical team at the InfectiousDiseases Centre, Princess Margaret Hospital, Hospital Authorityof Hong Kong. We used equipment purchased with NIH grants(S10OD018220 and 1S10OD021763) to generate the data.Funding: This work was supported by NIH grants HIPC U19AI090023(to B.P.), U19AI057266 (to B.P. and principal investigatorR. Ahmed from Emory University), and U24AI120134 (to S.E.B.);the Sean Parker Cancer Institute; the Soffer endowment(to B.P.); the Violetta Horton endowment (to B.P.); a Calmetteand Yersin scholarship from the Pasteur International NetworkAssociation (to H.L.); the National Natural Science Foundation ofChina (NSFC)–Research Grants Council (RGC) Joint ResearchScheme (N_HKU737/18) (to C.K.P.M. and M.P.); the GuangzhouMedical University High-level University Innovation TeamTraining Program [Guangzhou Medical University released (2017)no. 159] (to C.K.P.M. and M.P.); the U.S. NIH (contract no.HHSN272201400006C) (to M.P.); and the RGC of theHong Kong Special Administrative Region, China (project no.T11-712/19-N) (to M.P.). Next-generation sequencing serviceswere provided by the Yerkes NHP Genomics Core, which issupported in part by NIH P51 OD 011132, and the data wereacquired on a NovaSeq 6000 funded by NIH S10 OD 026799.Author contributions: Conceptualization: B.P., P.S.A., and F.W.;Investigation: P.S.A., F.W., C.K.P.M., M.P., N.S., Y.F., L.B., D.W.,J.C., K.L.P., G.A., C.H., and M.P.M.; Data curation and analysis:P.S.A., F.W., M.S., T.H., N.S., Y.F., D.K., A.A.U., H.T.M., and B.P.;Patient recruitment and clinical data curation: C.K.P.M., M.P.,L.B., O.T.-Y.T., G.A., W.S.L., J.M.C.C., T.S.H.C., C.Y.C.C., C.H., M.P.M.,H.L., E.A., S.E., N.R., and M.P.; Supervision: B.P., M.P., N.R., P.K.,H.T.M., S.E.B., and E.A.; Data visualization: P.S.A., F.W., M.S., and T.H.;Writing: P.S.A., F.W., M.S., T.H., and B.P.; Funding acquisition:B.P. All the authors read and accepted the manuscript. Competinginterests: B.P. and P.S.A. are inventors on a provisional patentapplication (no. 63/026,577) submitted by the Board of Trusteesof the Leland Stanford Junior University, Stanford, CA, thatcovers the use of “Therapeutic Methods for Treating COVID-19Infections.” Data and materials availability: The CITE-seq data and

    Arunachalam et al., Science 369, 1210–1220 (2020) 4 September 2020 10 of 11

    RESEARCH | RESEARCH ARTICLEon June 29, 2021

    http://science.sciencemag.org/

    Dow

    nloaded from

    https://doi.org/10.1101/2020.05.28.20115667https://doi.org/10.1101/2020.05.28.20115667http://science.sciencemag.org/

  • bulk transcriptomics data are publicly available in the GeneExpression Omnibus (GEO) under accession numbers GSE155673and GSE152418, respectively. This work is licensed under aCreative Commons Attribution 4.0 International (CC BY 4.0)license, which permits unrestricted use, distribution, andreproduction in any medium, provided the original work is properlycited. To view a copy of this license, visit https://creativecommons.org/licenses/by/4.0/. This license does not apply to figures/photos/artwork or other content included in the article that is

    credited to a third party; obtain authorization from the rightsholder before using such material.

    SUPPLEMENTARY MATERIALS

    science.sciencemag.org/content/369/6508/1210/suppl/DC1Materials and MethodsFigs. S1 to S21Tables S1 to S4

    References (39–43)MDAR Reproducibility Checklist

    View/request a protocol for this paper from Bio-protocol.

    5 May 2020; resubmitted 10 July 2020Accepted 4 August 2020Published online 11 August 202010.1126/science.abc6261

    Arunachalam et al., Science 369, 1210–1220 (2020) 4 September 2020 11 of 11

    Fig. 4. Early, transient ISG expression in COVID-19 infection. (A) A schematic representation of the DCenrichment strategy used in CITE-seq analysis. (B) UMAP representation of PBMCs from all analyzedsamples (n = 12),colored by manually annotated cell type. (C) Pairwise comparison of genesfrom healthy individuals (n = 5) and COVID-19–infected patients (n = 7) was conducted for each cluster.DEGs were analyzed for overrepresentation ofBTMs. The ringplot shows overrepresented pathways in up- and down-regulated genes of each cluster. Theheatmap on the right shows the average expression levels of 33 ISGs derived from the enriched BTMs indifferent cell clusters of healthy (n = 5) and COVID-19 subjects (n = 7). (D) UMAP representation of PBMCsfrom all analyzed samples showing the expression levels of selected IFNs and ISGs. (E) Kinetics ofcirculating IFN-a levels (femtograms per milliliter) in plasma measured using SIMoA technology (n = 18healthy and40 COVID-19–infected patients). (F) Correlation between circulating IFN-a levels in plasma and theaverage expression of ISGs measured by CITE-seq analysis. (G) Hierarchically clustered heatmap of theexpression of theCITE-seq ISG signature (C) in the bulk RNA-seq dataset, performed using an extended group ofsubjects (n = 17 healthy and 17 COVID-19–infected samples). Colors represent gene-wise z scores. (H)Bar chart representing the proportion of variance in CITE-seq ISG signature expression explained bythe covariates in the x axis through principal variance component analysis (PVCA). resid, residual.(figure on next page)

    RESEARCH | RESEARCH ARTICLEon June 29, 2021

    http://science.sciencemag.org/

    Dow

    nloaded from

    https://creativecommons.org/licenses/by/4.0/https://creativecommons.org/licenses/by/4.0/https://science.sciencemag.org/content/369/6508/1210/suppl/DC1https://en.bio-protocol.org/cjrap.aspx?eid=10.1126/science.abc6261http://science.sciencemag.org/

  • humansSystems biological assessment of immunity to mild versus severe COVID-19 infection in

    Bosinger, Holden Terry Maecker, Purvesh Khatri, Nadine Rouphael, Malik Peiris and Bali PulendranChristopher Huerta, Michele Paine McCullough, Huibin Lv, Evan Anderson, Srilatha Edupuganti, Amit A. Upadhyay, Steve E. Kazmin, Ghina Alaaeddine, Wai Shing Leung, Jacky Man Chun Chan, Thomas Shiu Hong Chik, Chris Yau Chung Choi,Natalia Sigal, Yupeng Feng, Laurel Bristow, Owen Tak-Yin Tsang, Dhananjay Wagh, John Coller, Kathryn L. Pellegrini, Dmitri Prabhu S. Arunachalam, Florian Wimmers, Chris Ka Pun Mok, Ranawaka A. P. M. Perera, Madeleine Scott, Thomas Hagan,

    originally published online August 11, 2020DOI: 10.1126/science.abc6261 (6508), 1210-1220.369Science

    , this issue p. eabc8511, p. 1210Sciencetherapeutic interventions.

    potentialwith mild-to-severe disease. These studies provide a compendium of immune cell information and roadmaps for report a systems biology approach to assess the immune system of COVID-19 patientset al.parameters. Arunachalam

    COVID-19 patients and found three prominent and distinct immunotypes that are related to disease severity and clinicalimmune modulation associated with COVID-19. They performed high-dimensional flow cytometry of hospitalized

    present a comprehensive atlas ofet al.system responds to and influences COVID-19 severity remains unclear. Mathew Coronavirus disease 2019 (COVID-19) has affected millions of people globally, yet how the human immune

    Immune profiling of COVID-19 patients

    ARTICLE TOOLS http://science.sciencemag.org/content/369/6508/1210

    MATERIALSSUPPLEMENTARY http://science.sciencemag.org/content/suppl/2020/08/11/science.abc6261.DC1

    CONTENTRELATED

    http://stm.sciencemag.org/content/scitransmed/12/555/eabc9396.fullhttp://stm.sciencemag.org/content/scitransmed/12/557/eabc5332.fullhttp://stm.sciencemag.org/content/scitransmed/12/550/eabc3539.fullhttp://stm.sciencemag.org/content/scitransmed/12/554/eabc1126.full

    REFERENCES

    http://science.sciencemag.org/content/369/6508/1210#BIBLThis article cites 42 articles, 4 of which you can access for free

    PERMISSIONS http://www.sciencemag.org/help/reprints-and-permissions

    Terms of ServiceUse of this article is subject to the

    is a registered trademark of AAAS.ScienceScience, 1200 New York Avenue NW, Washington, DC 20005. The title (print ISSN 0036-8075; online ISSN 1095-9203) is published by the American Association for the Advancement ofScience

    Science. No claim to original U.S. Government WorksCopyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of

    on June 29, 2021

    http://science.sciencemag.org/

    Dow

    nloaded from

    http://science.sciencemag.org/content/369/6508/1210http://science.sciencemag.org/content/suppl/2020/08/11/science.abc6261.DC1http://stm.sciencemag.org/content/scitransmed/12/554/eabc1126.fullhttp://stm.sciencemag.org/content/scitransmed/12/550/eabc3539.fullhttp://stm.sciencemag.org/content/scitransmed/12/557/eabc5332.fullhttp://stm.sciencemag.org/content/scitransmed/12/555/eabc9396.fullhttp://science.sciencemag.org/content/369/6508/1210#BIBLhttp://www.sciencemag.org/help/reprints-and-permissionshttp://www.sciencemag.org/about/terms-servicehttp://science.sciencemag.org/