coronavirus neuropilin-1 facilitates sars-cov-2 cell ......1 2 3 relative infection a ctrl ace2 nrp1...

7
CORONAVIRUS Neuropilin-1 facilitates SARS-CoV-2 cell entry and infectivity Ludovico Cantuti-Castelvetri 1,2 * , Ravi Ojha 3 * , Liliana D. Pedro 1,2 * , Minou Djannatian 1,2 * , Jonas Franz 4,5,6 * , Suvi Kuivanen 7 * , Franziska van der Meer 4 , Katri Kallio 3 , Tug ˘berk Kaya 1,2,8 , Maria Anastasina 3,9 , Teemu Smura 7 , Lev Levanov 7 , Leonora Szirovicza 7 , Allan Tobi 10 , Hannimari Kallio-Kokko 11 , Pamela Österlund 12 , Merja Joensuu 13 , Frédéric A. Meunier 13 , Sarah J. Butcher 3,9 , Martin Sebastian Winkler 14 , Brit Mollenhauer 15,16 , Ari Helenius 17 , Ozgun Gokce 8 , Tambet Teesalu 3,19,20 , Jussi Hepojoki 5,21 , Olli Vapalahti 7,11,22 , Christine Stadelmann 4 , Giuseppe Balistreri 3,18 , Mikael Simons 1,2,23 The causative agent of coronavirus disease 2019 (COVID-19) is the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). For many viruses, tissue tropism is determined by the availability of virus receptors and entry cofactors on the surface of host cells. In this study, we found that neuropilin-1 (NRP1), known to bind furin-cleaved substrates, significantly potentiates SARS-CoV-2 infectivity, an effect blocked by a monoclonal blocking antibody against NRP1. A SARS-CoV-2 mutant with an altered furin cleavage site did not depend on NRP1 for infectivity. Pathological analysis of olfactory epithelium obtained from human COVID-19 autopsies revealed that SARS-CoV-2 infected NRP1-positive cells facing the nasal cavity. Our data provide insight into SARS-CoV-2 cell infectivity and define a potential target for antiviral intervention. A n outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections has caused a pandemic asso- ciated with a severe acute pulmonary disease named COVID-19 (coronavirus disease 2019) (1). A related coronavirus, SARS- CoV, led to a much smaller outbreak in 2003, possibly due to infection occurring predomi- nantly in the lower respiratory system, where- as SARS-CoV-2 spreads rapidly through active pharyngeal viral shedding (2). Despite these differences, uptake of both viruses is mediated by the same cellular receptor: angiotensin- converting enzyme 2 (ACE2) (35). One hy- pothesis to explain the enhanced spreading of SARS-CoV-2 is the presence of a polybasic furin-type cleavage site, RRAR^S, at the S1-S2 junction in the SARS-CoV-2 spike (S) protein that is absent in SARS-CoV ( 6). Similar sequences are found in the S proteins of many other path- ogenic human viruses, including Ebola, HIV-1, and highly virulent strains of avian influenza (6, 7). The presence of the polybasic cleavage site in SARS-CoV-2 results in enhanced path- ogenicity by priming the fusion activity (8) and could potentially create additional cell surface receptor binding sites. Proteolytic cleavage of RRAR^S by furin exposes a conserved C- terminal motif, RXXR OH [where R is arginine and X is any amino acid; R can be substituted by lysine (K)], in the S protein. Such C-terminal sequences that conform to the C-end rule (CendR) are known to bind to and activate neuropilin (NRP1 and NRP2) receptors at the cell surface (9). Recent cryoelectron microscopy struc- tures of the SARS-CoV-2 S protein demon- strated that the S1-S2 junction is part of a solvent-exposed loop and is therefore acces- sible for receptor interactions (10, 11). To determine whether SARS-CoV-2 can use NRP1 for virus entry and infectivity, we gen- erated lentiviral particles pseudotyped with the SARS-CoV-2 S protein. Pseudoviruses are well suited for virus entry assays, as they allow viral entry to be distinguished from other vi- rus life-cycle steps. Human embryonic kidney 293 T (HEK-293T) cells, which have almost no detectable ACE2 and NRP1 transcripts (fig. S1), were transfected with plasmids that encode the two established host factors (4), human ACE2 and the transmembrane protease serine 2 (TMPRSS2), or NRP1. When expressed alone, ACE2 rendered cells susceptible to infec- tion (Fig. 1A). Although NRP1 did not promote infection in HEK-293T cells, its coexpres- sion with ACE2 and TMPRSS2 markedly enhanced infection (Fig. 1, A and B). NRP1 ex- pression increased infection in Caco-2 cells, which endogenously express ACE2 ( 12) (Fig. 1C and fig. S1D), showing that NRP1 can potentiate infection in the presence of other host factors. To test the specificity of NRP1-dependent virus entry, we developed monoclonal antibodies (mAbs) that were designed to functionally block the extracellular b1b2 domain of NRP1, which is known to mediate binding to CendR peptides ( 13). The mAb3 was observed to bind to the re- combinant b1b2 domain of wild-type (WT) NRP1 but not to the triple-mutant b1b2 do- main (S346A, E348A, and T349A in the CendR binding pocket) (fig. S2A). The potency of the mAbs in preventing cellular binding and in- ternalization of NRP ligands was tested using 80-nm silver nanoparticles (AgNP) coated with the prototypic NRP1-binding CendR peptide RPARPAR OH (9) (fig. S2B). mAb3 efficiently blocked AgNP-CendR binding (fig. S2C) and internalization (fig. S2, D and E), whereas an- other monoclonal antibody, mAb2, had no effect and was used as a control in further experiments. Treatment of HEK-293T with mAb3 significantly reduced infection by SARS-CoV-2 pseudoviruses in cells expressing ACE2, TMPRSS2, and NRP1 (Fig. 1D), but not in cells expressing ACE2 and TMPRSS2 only (fig. S2F). When SARS-CoV-2 pseudovirus was preincubated with recom- binant, soluble extracellular b1b2 domain of NRP1, the wild type significantly reduced in- fection compared with the triple mutant (Fig. 1E and fig. S2G). Next, we explored the role of NRP1 using SARS-CoV-2 isolated from COVID-19 patients from the Helsinki University Hospital. We used WT SARS-CoV-2 and a cleavage-impaired SARS- CoV-2 mutant that was isolated from Vero-E6 cells, which rapidly accumulate mutations at the furin cleavage site of the S protein dur- ing passaging (Fig. 2, A and B) (14). First, we confirmed that furin cleaved the WT, but not the mutant, SARS-CoV-2 S protein by analyz- ing S protein processing in Chinese hamster ovary cells with functional (parental) or deficient (FD11) furin enzyme (fig. S3) (15). Next, we RESEARCH Cantuti-Castelvetri et al., Science 370, 856860 (2020) 13 November 2020 1 of 5 1 Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany. 2 German Center for Neurodegenerative Diseases (DZNE), Munich, Germany. 3 Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland. 4 Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany. 5 Campus Institute for Dynamics of Biological Networks, University of Göttingen, Göttingen, Germany. 6 Max Planck Institute for Experimental Medicine, Göttingen, Germany. 7 Department of Virology, Medicum, University of Helsinki, Helsinki, Finland. 8 Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany. 9 Helsinki Institute of Life SciencesInstitute of Biotechnology, University of Helsinki, Helsinki, Finland. 10 Laboratory of Cancer Biology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia. 11 Department of Virology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland. 12 Department of Health Security, Finnish Institute for Health and Welfare (THL), Helsinki, Finland. 13 Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia. 14 Department of Anesthesiology and Intensive Care Medicine, University Medical Center Göttingen, Göttingen, Germany. 15 Department of Neurology, University Medical Center Göttingen, Göttingen, Germany. 16 Paracelsus-Elena-Klinik Kassel, Kassel, Germany. 17 Institute of Biochemistry, ETH Zürich, Zürich, Switzerland. 18 The Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia. 19 Cancer Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA. 20 Center for Nanomedicine and Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, USA. 21 Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland. 22 Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland. 23 Munich Cluster of Systems Neurology (SyNergy), Munich, Germany. *These authors contributed equally to this work. Corresponding author. Email: [email protected] (G.B.); [email protected] (M.S.) Corrected 12 November 2020. See full text. on June 8, 2021 http://science.sciencemag.org/ Downloaded from

Upload: others

Post on 28-Jan-2021

4 views

Category:

Documents


0 download

TRANSCRIPT

  • CORONAVIRUS

    Neuropilin-1 facilitates SARS-CoV-2 cell entryand infectivityLudovico Cantuti-Castelvetri1,2*, Ravi Ojha3*, Liliana D. Pedro1,2*, Minou Djannatian1,2*, Jonas Franz4,5,6*,Suvi Kuivanen7*, Franziska van der Meer4, Katri Kallio3, Tuğberk Kaya1,2,8, Maria Anastasina3,9,Teemu Smura7, Lev Levanov7, Leonora Szirovicza7, Allan Tobi10, Hannimari Kallio-Kokko11,Pamela Österlund12, Merja Joensuu13, Frédéric A. Meunier13, Sarah J. Butcher3,9,Martin Sebastian Winkler14, Brit Mollenhauer15,16, Ari Helenius17, Ozgun Gokce8,Tambet Teesalu3,19,20, Jussi Hepojoki5,21, Olli Vapalahti7,11,22, Christine Stadelmann4,Giuseppe Balistreri3,18†, Mikael Simons1,2,23†

    The causative agent of coronavirus disease 2019 (COVID-19) is the severe acute respiratory syndromecoronavirus 2 (SARS-CoV-2). For many viruses, tissue tropism is determined by the availability of virusreceptors and entry cofactors on the surface of host cells. In this study, we found that neuropilin-1(NRP1), known to bind furin-cleaved substrates, significantly potentiates SARS-CoV-2 infectivity, aneffect blocked by a monoclonal blocking antibody against NRP1. A SARS-CoV-2 mutant with an alteredfurin cleavage site did not depend on NRP1 for infectivity. Pathological analysis of olfactory epitheliumobtained from human COVID-19 autopsies revealed that SARS-CoV-2 infected NRP1-positive cellsfacing the nasal cavity. Our data provide insight into SARS-CoV-2 cell infectivity and define a potentialtarget for antiviral intervention.

    An outbreak of severe acute respiratorysyndrome coronavirus 2 (SARS-CoV-2)infections has caused a pandemic asso-ciated with a severe acute pulmonarydisease named COVID-19 (coronavirus

    disease 2019) (1). A related coronavirus, SARS-CoV, led to a much smaller outbreak in 2003,possibly due to infection occurring predomi-nantly in the lower respiratory system, where-as SARS-CoV-2 spreads rapidly through activepharyngeal viral shedding (2). Despite thesedifferences, uptake of both viruses is mediatedby the same cellular receptor: angiotensin-converting enzyme 2 (ACE2) (3–5). One hy-pothesis to explain the enhanced spreadingof SARS-CoV-2 is the presence of a polybasicfurin-type cleavage site, RRAR^S, at the S1-S2junction in the SARS-CoV-2 spike (S) proteinthat is absent in SARS-CoV (6). Similar sequencesare found in the S proteins of many other path-ogenic human viruses, including Ebola, HIV-1,and highly virulent strains of avian influenza(6, 7). The presence of the polybasic cleavagesite in SARS-CoV-2 results in enhanced path-ogenicity by priming the fusion activity (8) andcould potentially create additional cell surface

    receptor binding sites. Proteolytic cleavageof RRAR^S by furin exposes a conserved C-terminal motif, RXXROH [where R is arginineand X is any amino acid; R can be substitutedby lysine (K)], in the S protein. Such C-terminalsequences that conformto theC-end rule (CendR)are known to bind to and activate neuropilin(NRP1 and NRP2) receptors at the cell surface(9). Recent cryo–electron microscopy struc-tures of the SARS-CoV-2 S protein demon-strated that the S1-S2 junction is part of asolvent-exposed loop and is therefore acces-sible for receptor interactions (10, 11).To determine whether SARS-CoV-2 can use

    NRP1 for virus entry and infectivity, we gen-erated lentiviral particles pseudotyped withthe SARS-CoV-2 S protein. Pseudoviruses arewell suited for virus entry assays, as they allowviral entry to be distinguished from other vi-rus life-cycle steps. Human embryonic kidney293 T (HEK-293T) cells, which have almost nodetectableACE2 andNRP1 transcripts (fig. S1),were transfected with plasmids that encodethe two established host factors (4), humanACE2 and the transmembrane protease serine2 (TMPRSS2), orNRP1.When expressed alone,

    ACE2 rendered cells susceptible to infec-tion (Fig. 1A). AlthoughNRP1 did not promoteinfection in HEK-293T cells, its coexpres-sion with ACE2 and TMPRSS2 markedlyenhanced infection (Fig. 1, A and B). NRP1 ex-pression increased infection in Caco-2 cells,which endogenously express ACE2 (12) (Fig. 1Cand fig. S1D), showing thatNRP1 can potentiateinfection in the presence of other host factors.To test the specificity of NRP1-dependent virusentry, we developed monoclonal antibodies(mAbs) that were designed to functionally blockthe extracellular b1b2 domain of NRP1, whichis known tomediate binding to CendR peptides(13). The mAb3 was observed to bind to the re-combinant b1b2 domain of wild-type (WT)NRP1 but not to the triple-mutant b1b2 do-main (S346A, E348A, and T349A in the CendRbinding pocket) (fig. S2A). The potency of themAbs in preventing cellular binding and in-ternalization of NRP ligands was tested using80-nm silver nanoparticles (AgNP) coated withthe prototypic NRP1-binding CendR peptideRPARPAROH (9) (fig. S2B). mAb3 efficientlyblocked AgNP-CendR binding (fig. S2C) andinternalization (fig. S2, D and E), whereas an-othermonoclonal antibody,mAb2, hadno effectandwasused as a control in further experiments.Treatment ofHEK-293TwithmAb3 significantlyreduced infection by SARS-CoV-2 pseudovirusesin cells expressing ACE2, TMPRSS2, and NRP1(Fig. 1D), but not in cells expressing ACE2 andTMPRSS2 only (fig. S2F). When SARS-CoV-2pseudovirus was preincubated with recom-binant, soluble extracellular b1b2 domain ofNRP1, the wild type significantly reduced in-fection compared with the triple mutant (Fig.1E and fig. S2G).Next, we explored the role of NRP1 using

    SARS-CoV-2 isolated from COVID-19 patientsfrom theHelsinki University Hospital. We usedWT SARS-CoV-2 and a cleavage-impaired SARS-CoV-2 mutant that was isolated from Vero-E6cells, which rapidly accumulate mutations atthe furin cleavage site of the S protein dur-ing passaging (Fig. 2, A and B) (14). First, weconfirmed that furin cleaved the WT, but notthemutant, SARS-CoV-2 S protein by analyz-ing S protein processing in Chinese hamsterovary cells with functional (parental) or deficient(FD11) furin enzyme (fig. S3) (15). Next, we

    RESEARCH

    Cantuti-Castelvetri et al., Science 370, 856–860 (2020) 13 November 2020 1 of 5

    1Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany. 2German Center for Neurodegenerative Diseases (DZNE), Munich, Germany. 3Faculty of Biological andEnvironmental Sciences, Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki, Finland. 4Department of Neuropathology, University Medical Center Göttingen,Göttingen, Germany. 5Campus Institute for Dynamics of Biological Networks, University of Göttingen, Göttingen, Germany. 6Max Planck Institute for Experimental Medicine, Göttingen, Germany.7Department of Virology, Medicum, University of Helsinki, Helsinki, Finland. 8Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany. 9HelsinkiInstitute of Life Sciences–Institute of Biotechnology, University of Helsinki, Helsinki, Finland. 10Laboratory of Cancer Biology, Institute of Biomedicine and Translational Medicine, University ofTartu, Tartu, Estonia. 11Department of Virology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland. 12Department of Health Security, Finnish Institute for Health and Welfare(THL), Helsinki, Finland. 13Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia. 14Department ofAnesthesiology and Intensive Care Medicine, University Medical Center Göttingen, Göttingen, Germany. 15Department of Neurology, University Medical Center Göttingen, Göttingen, Germany.16Paracelsus-Elena-Klinik Kassel, Kassel, Germany. 17Institute of Biochemistry, ETH Zürich, Zürich, Switzerland. 18The Queensland Brain Institute, The University of Queensland, Brisbane,Queensland, Australia. 19Cancer Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA. 20Center for Nanomedicine and Department of Molecular, Cellular, andDevelopmental Biology, University of California, Santa Barbara, Santa Barbara, CA, USA. 21Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland.22Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland. 23Munich Cluster of Systems Neurology (SyNergy), Munich, Germany.*These authors contributed equally to this work.†Corresponding author. Email: [email protected] (G.B.); [email protected] (M.S.)

    Corrected 12 November 2020. See full text. on June 8, 2021

    http://science.sciencemag.org/

    Dow

    nloaded from

    https://science.sciencemag.org/content/370/6518/856http://science.sciencemag.org/

  • validated that exogenous ACE2 expression ren-dered HEK-293T cells susceptible to infectionwith SARS-CoV-2 (Fig. 2, C and D). NRP1 ex-pression alone caused lower levels of infection,which were detectable only with increasingvirus titer (Fig. 2, C and D). We then comparedthe ability of WT and mutant SARS-CoV-2 toinfect HEK-293T that stably express ACE2;ACE2 andTMPRSS2; or ACE2, TMPRSS2, andNRP1. Infection of these cell lines by the WT,but not the mutant, virus increased in the pres-ence of NRP1, providing further evidence thatNRP1 requires a furin-cleaved substrate for itseffects (Fig. 2, E and F). We studied the effect ofthe NRP1-blocking antibody, mAb3, on infec-tion of Caco-2 cells by WT and mutant SARS-CoV-2 and found that preincubation withNRP1-blocking antibody reduced WT virusinfection by ~40%, whereas the control mAb2had no effect (Fig. 2, G and H). NRP1-blockingantibody had no effect on the infection by themutated virus (Fig. 2, G and H).Cleavage of SARS-CoV-2 S protein at the

    S1-S2 site generates the C-terminal end se-quence TQTNSPRRAROH. To determinewheth-er this specific sequence can function as asubstrate for NRP1, we used AgNPs coated

    with TQTNSPRRAROH peptide or differentcontrol peptides, including one with a ter-minal amide group (TQTNSPRRARNH2), whichreduces NRP1 binding (9) (Fig. 3A). We foundthat AgNP-TQTNSPRRAROH, but not controlAgNPs, were efficiently taken up byHEK-293Tcells expressingNRP1 (Fig. 3, B andC). Next, wedetermined whether AgNP-TQTNSPRRAROHparticles were also internalized into cells in vivo.We chose to study nanoparticle entry in themouse olfactory epithelium, owing to the knownexpression of NRP1 in the olfactory system (16),including olfactory neuronal cells of the epithe-lium (fig. S4). AgNPs-TQTNSPRRAROHand con-trol AgNP-TQTNSPRRARNH2were administeredinto the nose of anesthetized adult mice. Sixhours after administration, we observed a sig-nificantly largeruptakeofAgNP-TQTNSPRRAROHthan of AgNP-TQTNSPRRARNH2 into the ol-factory epithelium (Fig. 3, D and E) and, un-expectedly, into neurons and blood vessels ofthe cortex (Fig. 3, F and G). Similar resultswere obtained for AgNPs coated with the pro-totypic NRP1-binding CendR peptide RPAR-PAROH (fig. S5).Having obtained evidence for a role of NRP1

    in cell entry of SARS-CoV-2, we examined whether

    NRP1 expression correlated with the detectionof virus RNA in single-cell transcriptomes. Forthese analyses, we used published single-cellRNA sequencing (scRNA-seq) datasets of cul-tured experimentally infected human bron-chial epithelial cells and cells isolated frombronchoalveolar lavage fluid (BALF) of severelyaffected COVID-19 patients (17). Among theproposed entry and amplification factors, NRP1,FURIN, and TMPRSS11Awere enriched in SARS-CoV-2–infected cells compared with noninfectedcells (fig. S6). We also detected increased ex-pression of these proteins after infection (fig.S6). In addition, RNA expression of NRP1 andits homolog NRP2 was elevated in SARS-CoV-2–positive cells compared with adjacent cellsin the BALF of severely affected COVID-19 pa-tients (fig. S7).Because the availability of virus receptors

    and entry cofactors on the surface of host cellsdetermines infectivity, we compared the ex-pression patterns of ACE2 and NRP1 in pub-lished scRNA-seq datasets of human lung tissue(18) and human olfactory epithelium (19).Whereas ACE2 was detected at very low lev-els, bothNRP1 andNRP2were abundantly ex-pressed in almost all pulmonary and olfactory

    Cantuti-Castelvetri et al., Science 370, 856–860 (2020) 13 November 2020 2 of 5

    ctrl NRP10

    1

    2

    3

    rela

    tive

    infe

    ctio

    n

    A

    ctrl ACE2 NRP1 TSS2

    0.0

    0.5

    1.0

    1.5

    rela

    tive

    infe

    ctio

    n

    SA

    RS

    -2V

    SV

    -Gcontrol ACE2 NRP1 TMPRSS2

    D

    0

    5

    10

    15

    20

    infe

    cted

    cells

    [%]

    cont

    rol A

    bm

    Ab3

    GFP merge

    ctrl mAb3

    *

    0.0

    0.2

    0.4

    0.6

    0.8

    1.0

    1.2

    rela

    tive

    infe

    ctio

    n

    E

    mut

    b1b

    2w

    t b1b

    2

    GFP merge

    mut b1b2

    wt b1b2

    **

    C

    NR

    P1

    SARS-2

    cont

    rol

    *

    **** ****

    ****

    0

    2

    4

    6

    rela

    tive

    infe

    ctio

    n

    ACE2TSS2NRP1

    - + +- + +

    - - +

    B

    AC

    E2+

    TSS

    2N

    RP

    1+A

    CE

    2+TS

    S2

    GFP merge*

    **

    Fig. 1. NRP1 facilitates the cellular entry of SARS-CoV-2 pseudotyped particles.(A) Representative images and quantification of SARS-CoV-2 S protein (SARS-2)(blue bars) and vesicular stomatitis virus glycoprotein (VSV-G) pseudotype (graybars) infectivity in HEK-293T cells transiently expressing control (ctrl) vector,ACE2, NRP1, or TMPRSS2 (TSS2). Data are normalized to the respectiveinfectivity of SARS-2 and VSV-G pseudotype in ACE2-expressing cells. Two-wayanalysis of variance (ANOVA) was carried out with Tukey’s correction for multiplecomparisons. (B) HEK-293T cells transiently expressing ACE2 and TMPRSS2or NRP1, ACE2, and TMPRSS2 were inoculated with SARS-2 pseudotype. Data arenormalized to SARS-2 infectivity in cells expressing ACE2 and TMPRSS2. One-way ANOVA was performed with Tukey’s correction for multiple comparisons.(C) SARS-2 pseudotype infectivity in Caco-2 cells expressing NRP1 or control

    vector. Data are normalized to the respective infectivity of SARS-2 and VSV-Gpseudotype in control cells. Two-way ANOVA was carried out with Sidak’scorrection for multiple comparisons. (D and E) HEK-293T cells transientlyexpressing NRP1, ACE2, and TMPRSS2 were inoculated with SARS-2 pseudotypein the presence of mAb3 antibody against NRP1 [(D), mAb3] or control mAb2[(D), ctrl Ab] and in the presence of soluble NRP1 wild-type b1b2 domain[(E), wt b1b2] or NRP1 mutant b1b2 domain [(E), mut b1b2]. Data in (E) arenormalized to untreated cells expressing NRP1, ACE2, and TMPRSS2. Two-tailedunpaired Student’s t test was performed. All data are represented as means ± SDsfrom three independent experiments [(A) to (C)] or three biological replicates[(D) and (E)]. *P < 0.05, **P < 0.01, ****P < 0.0001. All images show GFP-positive,infected cells (magenta) and Hoechst stain (cyan). Scale bars, 100 mm.

    RESEARCH | REPORT

    Corrected 12 November 2020. See full text. on June 8, 2021

    http://science.sciencemag.org/

    Dow

    nloaded from

    http://science.sciencemag.org/

  • cells, with the highest levels of expression inendothelial cells (figs. S8 and S9). We con-firmed these results by examining NRP1 im-munoreactivity in human autopsy tissue anddetected NRP1 in the epithelial surface layer

    of the human respiratory and olfactory epi-thelium (fig. S10A). ACE2 was hardly de-tectable in these tissues (fig. S10B). Withinthe olfactory epithelium, NRP1 was also ob-served in cells positive for oligodendrocyte

    transcription factor 2 (OLIG2), which is mostlyexpressed by olfactory neuronal progenitors(fig. S10C).In light of the widely reported disturbance

    of olfaction in a large fraction of COVID-19

    Cantuti-Castelvetri et al., Science 370, 856–860 (2020) 13 November 2020 3 of 5

    Fig. 2. A blocking antibody againstthe b1b2 domain of NRP1 reducesinfection by wild-type SARS-CoV-2(SARS-2-wt) but not a mutant with adeletion at the furin-cleavage site(SARS-2-mut). (A) Sequence analysis ofviruses isolated at different passages(P) from different cell types. The firstsequence is the reference from theWuhan isolate (NC_045512.2). Thesequence abundance in each virus pop-ulation is indicated as a percentage.ND, not determined. §, SARS-2-wt; §§,SARS-2-mut. A, Ala; S, Ser; Y, Tyr; Q,Gln; T, Thr; N, Asn; P, Pro; R, Arg; V, Val.(B) A deletion adjacent to the furin-cleavage site abrogates the enzymaticcleavage of the S protein. Immunoblotanalysis was carried out on cell lysatesfrom Vero-E6 cells infected for 16 hourswith two viral populations (§ and §§).Numbers indicate protein size (in kilo-daltons). (C and D) Representativeimages and quantification of SARS-2-wtinfectivity in HEK-293T cells stablyexpressing ACE2 (blue bars) or NRP1(orange bars) compared with nontrans-fected cells (gray bars). Different virustiters were used. Data are normalized tothe infectivity in ACE2-expressing cellsat multiplicity of infection (MOI) = 5.Two-way ANOVA was done with Tukey’scorrection for multiple comparisons.(E and F) Representative images (E) andquantification (F) of HEK-293T cellsstably expressing the indicated combi-nations of ACE2, TMPRSS2 (TSS2), andNRP1 after inoculation with SARS-2-wt(wt; blue bars) or SARS-2-mut (mut;gray bars). Data are normalized to therespective infectivity in ACE2-expressingcells. Two-way ANOVA was performedwith Tukey’s correction for multiplecomparisons. (G and H) Caco-2 cellinfection in the presence of controlmAb2 (ctrl. Ab) or mAb3 blocking anti-bodies against NRP1 after SARS-2-wt(wt; blue bars) or SARS-2-mut (mut;gray bars) inoculation. Data are normal-ized to the respective vehicle control(phosphate-buffered saline) sample.Two-way ANOVA was performed withTukey’s correction for multiple compar-isons. Data are means ± SDs from threeindependent experiments. *P < 0.05,**P < 0.01, ***P < 0.001, ****P < 0.0001. Magenta, SARS-2-wt– and SARS-2-mut–infected cells; Hoechst stain, cyan. Scale bars, 50 mm.

    Sequence Furin cleavage site %

    NC_045512.2_Wuhan-Hu-1WT

    FIN-UH25M-Nasopharynx

    Finland/1/2020-Nasopharynx

    FIN-UH25M-Caco-2 P3

    Finland/1/2020-VeroE6 P1

    Finland/1/2020-VeroE6 P3

    Finland/1/2020-VeroE6 P7

    ASYQTQTNSPRRAR^SVA

    ASYQTQTNSPRRAR^SVA

    ASYQTQTNSPRRAR^SVA

    ASYQTQTNSPRRAR^SVA

    ASY - - - - - SPRRAR^SVA

    ASY - - - - - SPRRAR^SVA

    ASY - - - - - SPRRAR^SVA

    100

    100

    ND

    100

    53

    78

    100

    A

    wt S

    ARS-

    2

    mut S

    ARS-

    2

    contr

    olB

    §

    §§

    §§§

    0

    1

    2

    3

    rela

    tive

    infe

    ctio

    n

    F

    wt S

    ARS-

    2m

    utan

    t SAR

    S-2

    ACE2 TSS2+ACE2 NRP1+TSS2+ACE2 E

    mutmutmut

    TSS2

    wtACE2 ACE2 ACE2

    TSS2

    wt wt

    NRP1

    §§§

    **

    *

    *** ***

    S

    S1

    tubulin

    180

    130

    100

    70

    55

    mock 0.313 0.625 1.250 2.500 5.000

    0.0

    0.2

    0.4

    0.6

    0.8

    1.0

    1.2

    SARS-CoV-2 titer [MOI]re

    lativ

    ein

    fect

    ion

    C D

    ********

    ****

    *********

    NR

    P1AC

    E2MOI 2.5 MOI 5

    wt S

    ARS-

    2m

    utan

    t SAR

    S-2

    PBS control Ab mAb3G H

    §§§

    mutmutmutmut

    ctrl. Ab

    wtcontrol SARS-2 SARS-2 SARS-2

    mAb3PBS

    wt wtwt

    0.0

    0.5

    1.0

    1.5

    rela

    tive

    infe

    ctio

    n

    ****

    RESEARCH | REPORT

    Corrected 12 November 2020. See full text. on June 8, 2021

    http://science.sciencemag.org/

    Dow

    nloaded from

    http://science.sciencemag.org/

  • patients (20, 21) and the enrichment of NRPsin the olfactory epithelium,we analyzed a seriesof autopsies from six COVID-19 patients andeight noninfected control patients to determinewhether SARS-CoV-2 could infect NRP1-positivecells (Fig. 4 and table S1). Using antibodiesagainst the S protein, we detected infection ofthe olfactory epithelium in five of six COVID-19patients. The infected olfactory epithelial cellsshowed high expression of NRP1 (Fig. 4, Aand B). Additional costaining indicated in-fection of cells positive for OLIG2 (Fig. 4Band fig. S11).There is limited knowledge about the virus–

    host interactions that determine cellular entryof SARS-CoV-2. Viruses display considerableredundancy and flexibility because they canexploit weak multivalent interactions to en-hance affinity. To date, studies of SARS-CoV-2entry have focused almost entirely on ACE2,which is expressed at very low protein levelsin respiratory and olfactory epithelial cells(22). This raises the possibility that cofactorsare required to facilitate virus–host cell in-teractions in cells with low ACE2 expression.NRP1 could represent such an ACE2 poten-tiating factor by promoting the interactionof the virus with ACE2. The reason a numberof viruses (23–26) use NRPs as entry factorsmay be their high expression on epithelia facingthe external environment and their function in

    Cantuti-Castelvetri et al., Science 370, 856–860 (2020) 13 November 2020 4 of 5

    Fig. 4. SARS-CoV-2infects the olfactoryepithelium. (A) Costain-ing of S protein (brown)and NRP1 (purple) in theapical olfactory epithe-lium (OE) in a nonin-fected control (left) andin the apical OE (middle)and adjacent mucosa(right) in a COVID-19patient. LP, laminapropria; HB, horizontalbasal cells. (B) Costain-ing of NRP1 (magenta) orOLIG-2 (magenta) withS protein (yellow) inOE cells. Nuclei areshown in blue. Scalebars, 10 mm.

    0.0

    0.2

    0.4

    0.6

    0.8

    AgN

    P-p

    ositi

    vear

    ea[%

    ]

    0

    20

    40

    60

    80

    100

    AgN

    P-po

    sitiv

    ece

    lls[%

    ]

    ***

    ACoV2-pre

    CoV2-post CoV2-post amide

    CoV2-Ala

    biotin -Ala -pre -post -post-amide

    SARS-2

    ***

    0.0

    0.5

    1.0

    1.5

    2.0

    2.5

    AgN

    P-p

    ositi

    vear

    ea[%

    ] *

    cont

    rol

    SAR

    S2 p

    eptid

    e

    AgNP NeuN + AQP4 merge

    ...TQTNSPRRAR SVA...S1 S2

    SARS-2-AlaSARS-2-preSARS-2-postSARS-2-post amide

    SARS-2 S protein

    nano

    parti

    cles biot-X-TQTNSPRRAA-OH

    biot-X-TQTNSPRRAR^SVA-OHbiot-X-TQTNSPRRAR-OHbiot-X-TQTNSPRRAR-NH2

    B C

    F G

    cont

    rol

    SAR

    S2 p

    eptid

    e

    AgNP NeuN + AQP4 mergeD E***

    -post-post-amide

    SARS-2

    -post-post-amide

    SARS-2

    Fig. 3. NRP mediates entry of nanoparticles coated with SARS-CoV-2(SARS-2) S–derived CendR peptides into cultured cells, olfactory epithe-lium, and the central nervous system of mice. (A) Peptide sequences usedfor AgNP coating. The peptides mimic SARS-2 S protein after furin cleavage(post) and as controls; S protein before cleavage (pre), in which the terminalamino acid is replaced by alanine (Ala); or with an amide terminus (postamide). X, any amino acid. The arrow indicates the cleavage site. (B and C)Representative images and quantification of the internalization of peptide-coatedAgNPs in HEK-293T cells expressing NRP1. Merged images show AgNP-positive

    cells (magenta) and Hoechst stain (cyan). One-way ANOVA was carried out withTukey’s correction for multiple comparisons. (D to G) Representative images andquantification of the main olfactory epithelium [(D) and (E), respectively] andcortex [(F) and (G), respectively] 6 hours after intranasal administration ofAgNPs coated with SARS2-post and SARS2-post amide peptides. n = 4 replicatesfor (C); n = 5 (E) and n = 4 (G) mice per condition. Data are means ± SDs.Two-tailed unpaired Student’s t test; *P < 0.05, ***P < 0.001. Magenta,AgNPs; cyan, Hoechst stain; green, NeuN (neuronal marker); yellow, AQP4.Scale bars, 100 mm (B), 20 mm [(D) and (F)].

    RESEARCH | REPORT

    Corrected 12 November 2020. See full text. on June 8, 2021

    http://science.sciencemag.org/

    Dow

    nloaded from

    http://science.sciencemag.org/

  • enabling cell, vascular, and tissue penetration(9, 13).

    REFERENCES AND NOTES

    1. N. Zhu et al., N. Engl. J. Med. 382, 727–733 (2020).2. R. Wölfel et al., Nature 581, 465–469 (2020).3. P. Zhou et al., Nature 579, 270–273 (2020).4. M. Hoffmann et al., Cell 181, 271–280.e8 (2020).5. N. J. Matheson, P. J. Lehner, Science 369, 510–511 (2020).6. B. Coutard et al., Antiviral Res. 176, 104742 (2020).7. L. V. Tse, A. M. Hamilton, T. Friling, G. R. Whittaker, J. Virol. 88,

    1673–1683 (2014).8. M. Hoffmann, H. Kleine-Weber, S. Pöhlmann, Mol. Cell 78,

    779–784.e5 (2020).9. T. Teesalu, K. N. Sugahara, V. R. Kotamraju, E. Ruoslahti,

    Proc. Natl. Acad. Sci. U.S.A. 106, 16157–16162 (2009).10. D. Wrapp et al., Science 367, 1260–1263 (2020).11. A. C. Walls et al., Cell 181, 281–292.e6 (2020).12. K. Liao, D. Sikkema, C. Wang, T. N. Lee, J. Immunol. Methods

    389, 52–60 (2013).13. A. Plein, A. Fantin, C. Ruhrberg, Microcirculation 21, 315–323

    (2014).14. A. D. Davidson et al., Genome Med. 12, 68 (2020).15. N. G. Ravindra et al., bioRxiv 2020.05.06.081695 [Preprint].

    13 July 2020.16. A. Kawakami, T. Kitsukawa, S. Takagi, H. Fujisawa, J. Neurobiol.

    29, 1–17 (1996).17. M. Liao et al., Nat. Med. 26, 842–844 (2020).18. X. Han et al., Nature 581, 303–309 (2020).19. M. A. Durante et al., Nat. Neurosci. 23, 323–326 (2020).20. L. Mao et al., JAMA Neurol. 77, 683–690 (2020).21. K. W. Cooper et al., Neuron 107, 219–233 (2020).22. F. Hikmet et al., Mol. Syst. Biol. 16, e9610 (2020).23. D. Ghez et al., J. Virol. 80, 6844–6854 (2006).24. N. Martinez-Martin et al., Cell 174, 1158–1171.e19 (2018).25. H. B. Wang et al., Nat. Commun. 6, 6240 (2015).

    26. M. Raaben et al., Cell Host Microbe 22, 688–696.e5 (2017).

    ACKNOWLEDGMENTS

    We thank R. Müller, K. Schulz, and U. Scheidt for experttechnical assistance; S. Osborne for proofreading themanuscript, and the DNA Dream Lab facility and K. Kogan fordesign and cloning of plasmids. Funding: The work in Munichand Göttingen was supported by grants from the GermanResearch Foundation (SPP2191, TRR128-2, TRR274-1,SyNergy Excellence Cluster, EXC2145, Projekt ID390857198,EXC 2067/1- 390729940, and STA 1389/5-1), the ERC(Consolidator Grant to M.S.), and the Dr. Miriam andSheldon G. Adelson Medical Research Foundation. Thework at the University of Helsinki was supported by theUniversity of Helsinki and by donations from Finnishcolleagues to whom we are very grateful. The Academy ofFinland supported G.B. (318434), O.V. (336490), S.J.B.(315950 and 336471), and J.H. (1308613 and 1314119).O.V. was supported by Jane and Aatos Erkko Foundation,EU Horizon 2020 program VEO (874735), and HelsinkiUniversity Hospital Funds (TYH2018322). S.J.B. wassupported by the Swedish Research Foundation and M.A.by the Marie Sklodowska-Curie Actions (799929).M.J. is supported by The Australian Research Council’sDiscovery Early Career Researcher Award (DE190100565).F.A.M. is supported by an Australian National Health andMedical Research Council Senior Research Fellowship(GNT1155794). T.T. and A.T. are supported by the EuropeanRegional Development Fund (project 2014-2020.4.01.15-0012),Wellcome Trust International Fellowship WT095077MA,European Research Council grant GLIOGUIDE, and theEstonian Research Council (grants PRG230 and EAG79 to T.T.).Author contributions: G.B., M.S., and A.H. conceived theproject. L.C.C., R.O., L.D.P., M.D., J.F., S.K., F.v.d.M., K.K.,M.A., and L.S. designed and carried out experiments. A.T.,T.T., L.L., O.V., J.H., O.G., H.K.K., P.O., and M.J., developed

    and provided tools. L.C.C., R.O., L.D.P., M.D., J.F., S.K.,T.K., C.S., T.S., M.J., F.A.M., S.J.B., J.H., and O.V. analyzedthe data or supervised data acquisition. L.C.C., R.O.,L.D.P., M.D., J.F., S.K., T.K., and O.G. visualized the data.T.K. and O.G. performed the scRNA-seq data analysis.M.S.W., B.M., C.S., and H.K.K. provided human samples.G.B. and M.S. wrote the manuscript. G.B. and M.S.supervised the project. Competing interests: T.T., O.V.,and G.B. have a pending patent on the monoclonalantibody 3 (mAb3) against the NRP1 b1 domain forSARS-CoV-2 inhibition. Data and materials availability: Alldata needed to evaluate the conclusions in the paper arepresent in the paper or the supplementary materials.This work is licensed under a Creative Commons Attribution4.0 International (CC BY 4.0) license, which permitsunrestricted use, distribution, and reproduction in any medium,provided the original work is properly cited. To view acopy 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 articlethat is credited to a third party; obtain authorization from therights holder before using such material.

    SUPPLEMENTARY MATERIALS

    science.sciencemag.org/content/370/6518/856/suppl/DC1Materials and MethodsFigs. S1 to S11Table S1References (27–37)MDAR Reproducibility Checklist

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

    12 June 2020; accepted 12 October 2020Published online 20 October 202010.1126/science.abd2985

    Cantuti-Castelvetri et al., Science 370, 856–860 (2020) 13 November 2020 5 of 5

    RESEARCH | REPORT

    Corrected 12 November 2020. See full text. on June 8, 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/370/6518/856/suppl/DC1https://en.bio-protocol.org/cjrap.aspx?eid=10.1126/science.abd2985http://science.sciencemag.org/

  • Neuropilin-1 facilitates SARS-CoV-2 cell entry and infectivity

    Balistreri and Mikael SimonsMollenhauer, Ari Helenius, Ozgun Gokce, Tambet Teesalu, Jussi Hepojoki, Olli Vapalahti, Christine Stadelmann, Giuseppe Kallio-Kokko, Pamela Österlund, Merja Joensuu, Frédéric A. Meunier, Sarah J. Butcher, Martin Sebastian Winkler, BritMeer, Katri Kallio, Tugberk Kaya, Maria Anastasina, Teemu Smura, Lev Levanov, Leonora Szirovicza, Allan Tobi, Hannimari Ludovico Cantuti-Castelvetri, Ravi Ojha, Liliana D. Pedro, Minou Djannatian, Jonas Franz, Suvi Kuivanen, Franziska van der

    originally published online October 20, 2020DOI: 10.1126/science.abd2985 (6518), 856-860.370Science

    , this issue p. 856, p. 861; see also p. 765Sciencepotential targets for future antiviral therapeutics.antibodies reduced viral infection in cell culture. Understanding the role of NRP1 in SARS-CoV-2 infection may suggestprotein binds directly to cell surface NRP1 and blocking this interaction with a small-molecule inhibitor or monoclonal

    found that the furin-cleaved S1 fragment of the spikeet al.highest expression in endothelial and epithelial cells. Daly potentiates SARS-CoV-2 infectivity. NRP1 is abundantly expressed in the respiratory and olfactory epithelium, with

    now show that neuropilin-1 (NRP1), which is known to bind furin-cleaved substrates,et al.Kielian). Cantuti-Castelvetri SARS-CoV-2 contains a cleavage site for the protease furin that is absent from SARS-CoV (see the Perspective by however, their tissue tropism differs, raising the possibility that additional host factors are involved. The spike protein ofcoronavirus 2 (SARS-CoV-2) and the earlier SARS-CoV use angiotensin-converting enzyme 2 (ACE2) as a receptor;

    Virus-host interactions determine cellular entry and spreading in tissues. Severe acute respiratory syndromeAnother host factor for SARS-CoV-2

    ARTICLE TOOLS http://science.sciencemag.org/content/370/6518/856

    MATERIALSSUPPLEMENTARY http://science.sciencemag.org/content/suppl/2020/10/19/science.abd2985.DC1

    CONTENTRELATED

    http://science.sciencemag.org/content/sci/370/6518/861.fullhttp://science.sciencemag.org/content/sci/370/6518/765.fullhttp://stm.sciencemag.org/content/scitransmed/12/564/eabd5487.fullhttp://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.full

    REFERENCES

    http://science.sciencemag.org/content/370/6518/856#BIBLThis article cites 37 articles, 6 of which you can access for free

    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 8, 2021

    http://science.sciencemag.org/

    Dow

    nloaded from

    http://science.sciencemag.org/content/370/6518/856http://science.sciencemag.org/content/suppl/2020/10/19/science.abd2985.DC1http://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://stm.sciencemag.org/content/scitransmed/12/564/eabd5487.fullhttp://science.sciencemag.org/content/sci/370/6518/765.fullhttp://science.sciencemag.org/content/sci/370/6518/861.fullhttp://science.sciencemag.org/content/370/6518/856#BIBLhttp://www.sciencemag.org/about/terms-servicehttp://science.sciencemag.org/

  • 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 8, 2021

    http://science.sciencemag.org/

    Dow

    nloaded from

    http://www.sciencemag.org/help/reprints-and-permissionshttp://www.sciencemag.org/about/terms-servicehttp://science.sciencemag.org/