INTRODUCTION
Bats are natural hosts of numerous coronaviruses, including members of the
Betacoronavirus genus, which comprises viruses belonging to the severe acute respiratory syndrome coronavirus (SARS-CoV) 1 and 2 lineages (
1). RNAs from SARS-CoV-2 related coronaviruses (SC2r-CoVs) have been recently detected in different species of
Rhinolophus living in Northern Laos. One of these viruses, named BANAL-52, was found in
R. malayanus and is, to date, the closest relative to SARS-CoV-2, with an overall sequence identity of 96.8% (
2). Other viruses belonging to this lineage have been identified in
Rhinolophus bats sampled in China (
3,
4) Thailand (
5), Cambodia (
6), and Japan (
7). SC2r-CoVs are thus probably widely distributed in South-East Asia. In addition, numerous other bat species worldwide are infected with betacoronaviruses, including species of the
Myotis,
Nyctalus,
Tadarida, and
Eptesicus genera (
8–14).
The risk of spillback transmission of SARS-CoV-2 from humans to domestic animals or wildlife remains a major concern, since this reverse zoonotic transmission has been already documented in pet animals and farmed minks, as well as in tigers and gorillas in zoos (
15,
16). Infection and transmission of SARS-CoV-2 has also been reported in white-tailed deer (
17). Given the likely bat origin of SARS-CoV-2, bats could be putatively at risk of spillback transmission (
18). The establishment of novel bat reservoirs would have a severe impact on wildlife conservation and public health measures.
SARS-CoV, SARS-CoV-2, and one of the BANAL viruses use the surface receptor angiotensin-converting enzyme 2 (ACE2) to enter human cells (
2,
4,
19–21). Viral binding to ACE2 is followed by the proteolytic cleavage of the viral spike (S) proteins by either the plasma-membrane resident transmembrane protease serine 2 (TMPRSS2) or the endosomal cathepsin L (CTSL) (
22). This cleavage is mandatory for the fusion between the viral and cellular membranes. Thus, localization and expression of TMPRSS2 and CTSL dictate whether the virus enters cells by fusing at the cell surface or in endosomes (
22,
23).
Several approaches have been used to predict the ability of ACE2 from phylogenetically diverse bat species to promote SARS-CoV-2 entry. First, comparison of ACE2 protein sequences from 37 bat species, including species of the genus
Rhinolophus, predicted a low or very low ability to interact with viral S proteins (
24). Second, ectopic expression of ACE2 from several bat species in nonpermissive mammalian cells, followed by infection with genuine viruses or pseudoviruses carrying SARS-CoV-2 S proteins, revealed that ACE2 from
Rhinolophus,
Myotis, and
Eptesicus species allowed viral entry, albeit often less efficiently than human ACE2. However, these approaches using
in silico analysis or ectopic expression of bat ACE2 in human or hamster cells do not allow to draw conclusions as to which bat species might actually support SARS-CoV-2 replication. Other factors unique to bat cells may potentially modulate viral entry and replication. Indeed, experiments performed with cells derived from lung tissue of
Rhinolophus alcyone and
Myotis daubentonii showed that they were not susceptible to infection with vesicular stomatitis viruses (VSVs) bearing SARS-CoV-2 S proteins (
20). Cells originating from lung and kidney tissue of
Rhinolophus sinicus and
Eptesicus fuscus were not permissive to SARS-CoV-2 either (
25,
26). These studies underline the limitation of predicting the ability of S proteins to interact with ACE2 orthologs based on computational models or ectopic expression. These findings also highlight the limitation of predicting viral host tropism based on cellular studies.
Only a few models are available to study the replication of betacoronaviruses in bat cells. Viral replication was detected in
Rhinolophus sinicus lung and brain cells, as well as in
Pipistrellus abramus kidney cells (
27), but viral titers were very low. In contrast, SARS-CoV-2 replicated efficiently in
R. sinicus intestinal organoids (
28), confirming further the ability of
Rhinolophus cells to support viral replication. Intranasal inoculation of SARS-CoV-2 in
Rousettus aegypticus resulted in transient infection of their respiratory tract and oral shedding of the virus (
29), indicating that bats unrelated to the
Rhinolophus genus can be productively infected with the virus. Since the manipulation of bat organoid and animal models remains challenging, there is a need to develop cell lines from various organs and species to gain deeper insights into bat-virus coevolution (
30). Here, we developed novel cellular models derived from understudied bat species widely circulating in Europe and Asia. The various susceptibilities and permissivities of the cells to SARS-CoV-2 infection offered the opportunity to uncover species-specific molecular restrictions to viral replication.
DISCUSSION
The development of novel bat cellular models is essential to understand the molecular mechanisms underlying the ability of bats to serve as reservoirs for numerous viruses, including alpha- and betacoronaviruses. We first produced
R. ferrumequinum,
M. myotis,
M. nattereri, and
M. brandtii primary cells to evaluate their permissivity to infection with SARS-CoV-2. None of them supported viral replication, not even
R. ferrumequinum cells, which have been isolated from bats belonging to the same genus as the bat host of BANAL-52, a potential ancestor of SARS-CoV-2 (
2). These primary cells, which were generated from patagium biopsy specimens of living bats, exhibited a dermal-fibroblast phenotype. A single-cell transcriptomic analysis showed that
R. sinicus skin cells express moderate levels of ACE2 and very little TMPRSS2 (
53). The virus may thus be able to enter the skin primary cells, but the fusion between viral and membrane may not take place as the S protein is not cleaved. Further experiments will be required to identify at which step of the replication cycle the virus is stopped in these primary cells.
We established the first three immortalized
Nyctalus noctula cell lines using liver, kidney, and lung tissues from a single bat. These organs may be physiologically relevant for bat infection since bat coronaviruses, such as MERS-CoV, infect the lungs and livers of fruit bats (
1). In addition to coronaviruses,
N. noctula carry other viruses with zoonotic potential such as paramyxoviruses and hantaviruses (
54,
55). The Nn cells that we have developed represent thus novel opportunities to study bat-borne viruses. We found that Nn kidney cells expressed higher levels of ACE2 than Nn cells derived from the lung or liver. Likewise, ACE2 is expressed at high levels in
R. sinicus kidney, as revealed by comparative single-cell transcriptomic (
53) and
in silico analysis of ACE2 expression pattern in various tissues. ACE2 is also highly expressed in human kidney (
56). Thus, kidney cells appear relevant to study betacoronavirus replication.
Myotis myotis,
Tadarida brasiliensis,
Eptesicus serotinus, and
Nyctalus noctula cells were resistant to infection. ACE2 from
M. myotis and
T. brasiliensis, as well as from a species of the
Eptesicus genus, permitted S-mediated entry of pseudotyped VSV when ectopically expressed in human cells refractory to SARS-CoV-2 infection (
57). This means that when expressed at high levels, ACE2 from these three species interacts with the viral S protein. As in human A549 cells, ACE2 may be expressed at a level which is too low to allow viral entry in our bat cell lines. The potential ability of
N. noctula ACE2 to bind S protein has not been reported and the genome of this bat genus is yet to be sequenced. Hence, low affinity between S protein and ACE2 and/or low level of ACE2 expression may hamper viral replication in these cells. Our results highlight the importance of performing experiments in the context of genuine infection of bat cells to predict their ability to support viral replication.
SARS-CoV-2 RNA was detected in nontransduced A549, FLG-ID, and NnKi cells at early times postinfection. Since the virus could not be removed by trypsin treatment of the cells, it likely represents input virus that entered the cells via an ACE2-independent manner, as described previously for Vero E6 cells (
22) and human H522 lung adenocarcinoma cells (
58). The VSV-based entry assay clearly showed that A549, FLG-ID, and NnKi cells are not susceptible to S-mediated infection. On the contrary, the same assay demonstrated that a subset of MmBr cells is susceptible to S-mediated infection, suggesting that these cells express all the factors necessary for binding, entry, and viral fusion. However, since they were not permissive to SARS-CoV-2, they probably lack factors that are essential for viral replication. Alternatively, they may express potent viral restriction factors that remain to be identified.
To bypass entry-mediated restriction(s), we generated eight chimeric bat cell lines stably expressing hACE2. Four cell lines (MmNep-ACE2, NnLi-ACE2, NnLu-ACE2, and NnKi-ACE2) expressed too little hACE2 to draw conclusion about a potential rescue of infection. Four cell lines (FLG-ID-ACE2, MmBr-ACE2, MmNep-ACE2
+, and NnLi-ACE2
+) expressed more hACE2 than A549-ACE2, which were competent for viral replication. The virus completed its replication cycle in FLG-ID-ACE2 cells. MmBr-ACE2 cells were competent for production of viral RNA and proteins but not for infectious particles release. The virus replicated poorly in MmNep-ACE2
+ and NnLi-ACE2
+ cells, even when pretreated with trypsin. A protease-independent restriction may thus exist in these cells. The various permissivities of these four cell lines to SARS-CoV-2 infection offer opportunities to decipher species-specific and tissue-specific antiviral mechanisms that have evolved in bats. Efficient viral RNA and protein production in A549-ACE2, FLG-ID-ACE2, and MmBr-ACE2 cells suggest that they express proteases that cleave S proteins. It also shows that ACE2 alone was responsible for the lack of viral replication in nontransduced A549, FLG-ID and MmBr cells. This ACE2-mediated entry block might be rather due to a low or absent ACE2 expression than to an incompatibility between ACE2 and S protein since ectopic expression of
Myotis sp. and
Eptesicus sp. ACE2 facilitated S-mediated entry of pseudoviruses (
43,
57). Infectious particles were produced in MmBr cells but were retained at the cell surface. This block may be mediated by the restriction factor tetherin, which retains numerous enveloped viruses, including SARS-CoV-2, at the surface of human cells (
47). An analysis of the tetherin gene of 27 species of bats indicates that bats have undergone tetherin gene expansion and diversification relative to other mammals (
59). Bats belonging to the genus
Myotis possess five unique tetherin variants that may potently restrict SARS-CoV-2 release (
59). Alternatively, infectious virion retention in MmBr cells could be due to an overexpression of hACE2 and may thus not be bat specific.
Since infection induced S-mediated syncytium formation in MmBr cells, viruses might spread from cell-to-cell via syncytia, as do other syncytium-forming viruses such as respiratory syncytial virus, parainfluenza viruses and measles virus (
60). Syncytium-mediated intercellular spreading allows viruses to escape virus-neutralizing antibodies. Such mode of transport has been previously proposed in human cells infected with the Middle East respiratory syndrome coronavirus (MERS-CoV) (
61), another betacoronavirus. Analysis of
postmortem samples of patients that succumb of COVID-19 revealed the presence of syncytial pneumocytes positives for viral RNAs (
62). However, the significance of syncytium formation for virus pathogenicity remains to be investigated.
SARS-CoV-2 has evolved numerous synergetic mechanisms to evade the IFN response in human cells (
63), resulting in an absence of IFN and ISG expression in some cells, including A549 cells (
52,
64). The virus was unable to counteract
OAS1 and
IFIH1 induction in
Eptesicus serotinus kidney cells and in
Myotis myotis nasal epithelial cells. This is especially intriguing in
E. serotinus cells since the virus replicates to high levels in these cells and thus produce viral proteins with described IFN antagonist activities. Similarly, MERS-CoV suppresses the antiviral IFN response in human cells but not in
E. fuscus cells (
65). One can envisage that escape of IFN-mediated restriction by betacoronaviruses is species specific. For instance, SARS-CoV-2 Nsp14 targets human IFNAR1 for lysosomal degradation (
63) but may be unable to degrade bat IFNAR1. This inability to evade the expression of two ISGs in
E. serotinus kidney cells and in
Myotis myotis nasal epithelial cells may contribute to the cellular control of infection in these cells, as in experimentally infected
E. fuscus (
66). Alternatively, the basal level of IFN may be high in these two cell lines, as reported in several other bat species (
67–69). Expression of a mutated form of IRF3, which is a key transcription factor involved in the induction of the IFN signaling cascade, contributes to enhanced IFN responses in bat species, including
E. fuscus, compared to humans (
70). Investigation of IRF7, another transcription factor that mediates IFN expression, in
Pteropus alecto cells revealed a more widespread tissue distribution in bats than in humans (
71,
72). Bats may thus launch IFN-dependent measures against viruses in a faster and broader manner than humans (
73). Expression of atypical ISGs has been reported for different bat species, including RNA-degrading RNase L (RNaseL) in
P. alecto cells and RNA-binding Microrchidia 3 (MORC3) in
Pteropus vampyrus and
Eidolon helvum cells (
74,
75). Pursuing the characterization of bat innate immunity in relevant
in vitro models is essential to understand the mechanisms by which they control the replication of numerous unrelated viruses.
An obvious need to develop additional bat cell lines still remains (
30). Particularly valuable cells would be cells derived from bat intestine, a tissue that expresses high level of proteins known to mediate or facilitate cellular entry of bat-borne betacoronaviruses, such as ACE2 and TMPRSS2, at least in
R. sinicus (
53). This tissue is relevant for coronavirus infection, as demonstrated by the detection of viral genomes in duodenum tissue of
Rousettus aegypticus experimentally infected with SARS-CoV-2 (
29) and in anal swabs of
Rhinolophus bats infected with SC2r-CoVs (
76).
MATERIALS AND METHODS
Bat primary cells.
M. myotis samples were collected in July 2020 from two bat colonies in Inca and Llucmajor on Mallorca (Balearic Islands, Spain) (agreement CEP 31/2020 delivered by the Ministry of the Environment and Territory, government of the Balearic Islands).
R. ferrumequinum biopsy specimens were collected in France in 2020. Authorization for bat capture was delivered by the French Ministry of Ecology, Environment, and Sustainable development (approval C692660703 from the Departmental Direction of Population Protection, Rhone, France). All methods were approved by the Société Française pour l’Étude et la Protection des Mammifères. Patagium biopsy specimens were shipped in freezing medium Cryo-SFM (PromoCell), on dry ice or at 4°C with ice packs. Primary cells were obtained as previously described (
77,
78). Briefly, skin biopsy specimens were washed twice with sterile phosphate-buffered saline (PBS), excised in small pieces and enzymatically digested, either with 500 μL of collagenase D (1 mg/mL) (Roche) and overnight incubation at 37°C without agitation, or with 100 to 200 μL of TrypLE Express Enzyme (Gibco) and incubation 10 min at 37°C under gentle agitation. Dissociated cells and remaining pieces of tissue were placed in a single well of a 6-well plate containing 2 mL of Dulbecco modified Eagle medium (DMEM; Gibco) containing 20% heat-inactivated fetal bovine serum (FBS; Eurobio), 1% penicillin/streptomycin (P/S; Gibco), and 50 μg/mL gentamicin (Gibco) and then incubated at 37°C under 5% CO
2. Cell cultures were regularly checked to determine the need for medium refreshment or splitting. After 5 to 10 passages, cells were grown in DMEM supplemented with 10% FBS.
Cell lines.
FLG-ID, FLG-R, FLN-ID, FLN-R, and Tb1Lu cell lines (
Table 1) were maintained in equal volumes of Ham F-12 and Iscove modified Dulbecco medium (Gibco), supplemented with 10% FBS and 1% P/S (Gibco) in nonvented flasks. Mm cells, which were obtained from a single common serotine bat (
Eptesicus serotinus), were previously described (
35). Nn kidney-, liver-, and lung-derived cell cultures were obtained from a common noctule bat (
Nyctalus noctula) euthanized because of poor chance of survival associated with traumatic injuries sustained while a dead tree sheltering bat hibernaculum was cut. The decision to euthanize the specimen was made by a veterinarian following inspection of a group of noctule bats presented for examination and therapy in the rescue center at the University of Veterinary and Pharmaceutical Sciences Brno, Czech Republic, in November 2015 (
79). The bat was anesthetized with isofluranum (Piramal Enterprises, Ltd.) and euthanized by quick decapitation. The cadaver was immersed into 96% ethanol for a few seconds and then subjected to necropsy under aseptic conditions to collect organs, which were loosened mechanically with scalpel blades, minced into small pieces, suspended in DMEM (Biosera) containing 1 mg/mL collagenase (Thermo Fisher Scientific) and 1 mg/mL trypsin (Sigma-Aldrich), and incubated at 37°C on a shaking Thermoblock for 45 min. The cells were then separated through a 100 μm nylon filter and washed twice in a medium supplemented with 10% FBS to stop enzymatic digestion. The cells yielded in this way were cultured in DMEM supplemented with 10% FBS and 1% P/S (Sigma). Primary cells were immortalized by transfection of pRSVAg1 plasmid expressing simian vacuolating virus 40 large T antigen (SV40T) with lipofectamine 2000 (Invitrogen) according to the manufacturer’s protocol, expanded and cryopreserved. Mm and Nn cell lines (
Table 1), as well as African green monkey Vero E6 cells (ATCC CRL-1586), human lung epithelial A549 cells (a gift from Frédéric Tangy, Institut Pasteur, Paris, France) and human colorectal adenocarcinoma Caco-TC7 cells (ATCC HTB-37), were maintained in DMEM (Gibco), supplemented with 10% FBS and 1% P/S in vented flasks. All cells were maintained at 37°C in a humidified atmosphere with 5% CO
2. Bat and A549 cells were modified to stably express hACE2 using the pLenti6-hACE2 lentiviral transduction as described previously (
37). Briefly, 2 × 10
5 cells were resuspended in 150 μL of culture medium containing 15 μL of ultracentrifuged lentiviral vectors supplemented with 2 mM HEPES (Gibco) and 4 μg/mL Polybrene (Sigma). Cells were agitated for 30 s every 5 min for 2.5 h at 37°C in a Thermomixer and then plated. At 48 h after transduction, blasticidin (concentrations ranging from 7 to 15 μg/mL depending on cell lines) was added in the culture media.
Viruses and infections.
The SARS-CoV-2 strain BetaCoV/France/IDF0372/2020 (historical) and hCoV-19/France/PDL-IPP01065/2021 (20H/501Y.V2 or SA) were supplied by the French National Reference Centre for Respiratory Viruses hosted by Institut Pasteur (Paris, France) and headed by S. van der Werf. The human samples from which the historical and South African strains were isolated were provided by X. Lescure and Y. Yazdanpanah from the Bichat Hospital, Paris, France, and Vincent Foissaud, HIA Percy, Clamart, France, respectively. These strains were supplied through the European Virus Archive goes Global (EVAG) platform, a project that has received funding from the European Union’s Horizon 2020 research and innovation program under grant agreement 653316. The hCoV-19/Japan/TY7-501/2021 strain (20J/501Y.V3 or Brazil) was kindly provided by Jessica Vanhomwegen (Environment and Infectious Risks Research and Expertise Unit, Institut Pasteur). Viral stocks were produced by amplification on Vero E6 cells, for 72 h in DMEM supplemented with 2% FBS and 1% P/S. The cleared supernatant was stored at −80°C and titrated on Vero E6 cells by using standard plaque assays to measure PFU/mL. Cells were infected at the indicated MOIs in DMEM without FBS. Virus inoculum was either removed after 6 h and replaced or topped up with FBS containing culture medium to a final concentration of 2% FBS and 1% P/S. For infections with proteolytically activated SARS-CoV-2, cell monolayers were washed twice with PBS before adding virus inoculum in DMEM supplemented with 2 or 4 μg/mL of trypsin TPCK (Sigma) and no FBS. After 4h, DMEM containing FBS was added to a final concentration of 2%.
TCID50 assays.
Supernatants of infected cells were first cleared of cell debris by centrifugation at 3,500 rpm for 10 min at 4°C. They were 10-fold serially diluted in DMEM supplemented with 2% FBS and 1% P/S. For the “ultracentrifugation” condition in MmBr-ACE2 50% tissue culture infective dose (TCID
50) assays, cleared supernatants were ultracentrifuged for 1 h at 45,000 rpm at 4°C. to remove cytokines and other proteins. Virus-containing pellets were resuspended in DMEM with 2% FBS and 1% P/S after 4 h of incubation at 4°C. For the “lysate” condition, infected MmBr-ACE2 or A459-ACE2 cells were lysed and scraped in ddH
2O. After one freeze-thaw cycle, whole-cell lysates were cleared by centrifugation, supplemented with 10× PBS to a physiological condition and used for serial dilutions. Around 9 × 10
3 Vero E6 cells and 50-μL potions of serially diluted virus suspensions were deposited in 96-well plate in quintuplicate wells. Cells were fixed with 4% paraformaldehyde (PFA) for 30 min at room temperature and revealed with crystal violet 5 days later. CPEs were assessed by calculating the TCID
50 values using the Spearman-Karber method (
80).
Flow cytometry.
Cells were detached with trypsin or versene for hACE2 staining. Cells were then fixed in 4% PFA for 30 min at 4°C and staining was performed in PBS, 2% bovine serum albumin (BSA), 2 mM EDTA, and 0.1% saponin (FACS buffer). Cells were incubated with goat pAB anti-hACE2-647 (1:100, FAB933R; R&D Systems) and/or with antibodies recognizing the spike protein of SARS-CoV (anti-S, 1:1,000, GTX632604; Genetex) or anti-S mAb10 (1 μg/mL; kindly provided by Hugo Mouquet, Institut Pasteur, Paris, France) and subsequently with secondary antibodies anti-human Alexa Fluor-647 (1:1,000, A21455; Thermo), anti-mouse Alexa Fluor-488 (1:1000, A28175; Thermo) or Dylight488 (1:100, SA5-10166; Thermo) for 30 min at 4°C. Cells were acquired on an Attune NxT flow cytometer (Thermo Fisher), and data were analyzed using FlowJo software v10 (TriStar).
RNA extraction and RT-qPCR assays.
Total RNA was extracted from cells with the NucleoSpin RNA II kit (Macherey-Nagel) according to the manufacturer’s instructions. First-strand complementary DNA (cDNA) synthesis was performed with the RevertAid H Minus M-MuLV reverse transcriptase (Thermo Fisher Scientific) using random primers. For batACE2 determination, total RNA was treated with DNase I (DNase-free kit; Thermo Fisher Scientific) for 30 min at 37°C before cDNA synthesis with SuperScript IV reverse transcriptase. Quantitative real-time PCR was performed on a real-time PCR system (QuantStudio 6 Flex; Applied Biosystems) with Power SYBR green RNA-to-CT 1-Step kit (Thermo Fisher Scientific). Data were analyzed using the 2
–ΔΔCT method, with all samples normalized to GAPDH. Genome equivalent concentrations were determined by extrapolation from a standard curve generated from serial dilutions of the pcDNA3.1-hACE2 plasmid (Addgene, 145033) or plasmids encoding a fragment of the RNA-dependent RNA polymerase/IP4 of SARS-CoV-2 or a fragment of the ACE2 genome of each bat species. Primers used for RT-qPCR analysis are listed in
Table 2.
Cloning of qPCR amplicon.
To quantify the amounts of bat ACE2 in each cell line, plasmids containing the qPCR amplicon obtained with the primers described in
Table 2 were generated via TOPO cloning. Briefly, total RNA was extracted from a cadaver of
Myotis myotis stored at the University of Veterinary and Pharmaceutical Sciences in Brno. For the remaining two bat species, total RNA extracted from NnKi and FLG-R cells were used. RNA was treated for 30 min at 37°C with DNase I and cDNA synthesized with SuperScript IV reverse transcriptase. These cDNAs were then used as the template for PCR amplification of the qPCR bACE2 amplicon using the primers in
Table 2 and Phusion high-fidelity DNA polymerase (Thermo). PCR products were gel-purified (NucleoSpin gel and PCR cleanup kit; Macherey-Nagel) and cloned into pCR-Blunt II-TOPO vectors using a Zero Blunt TOPO PCR cloning kit (Thermo). Inserts were verified via Sanger sequencing. Plasmids were then used as quantitative qPCR standards.
Western blot analysis.
Proteins extracted from cell lysates were resolved by SDS-PAGE on 4 to 12% NuPAGE Bis-Tris gel (Life Technologies) with MOPS running buffer and semidry transferred to a nitrocellulose membrane with Trans-Blot Turbo system (Bio-Rad). After blocking with 0.05% Tween 20 in PBS containing 5% dry milk powder for 1 h at room temperature, the membrane was incubated with goat pAB anti-hACE2-700 (1:200, FAB933N R&D Systems) and mouse monoclonal antibody anti-β-actin (1:5,000, A5316; Sigma) diluted in blocking buffer overnight at 4°C. The membranes were then incubated with DyLight800 secondary antibody (1:5,000, 46421; Thermo) diluted in blocking buffer for 1 h. Finally, the membranes were revealed using an Odyssey CLx infrared imaging system (LI-COR Bioscience).
Immunofluorescence microscopy and live cell imaging.
Cells grown on glass coverslips were fixed in 4% PFA for 30 min at room temperature and permeabilized with 0.2% Triton X-100 (Sigma/Merck) in PBS for 10 min at room temperature. After blocking with 3% BSA (Sigma) in PBS for 1 h at room temperature, cells were incubated with goat pAB anti-hACE2 (1:50, AF933; R&D Systems) and monoclonal antibody anti-SARS-CoV-2-spike (1:1,000, GTX632604; Genetex) in 1% BSA in PBS (AB buffer) for 1 h at room temperature or overnight at 4°C. Subsequently, cells were incubated with anti-goat Alexa 488 (A-11055; Thermo Fisher Scientific) and anti-mouse Alexa 555 (A21427; Thermo Fisher Scientific) secondary antibodies diluted 1:500 in antibody buffer for 30 min at room temperature. Finally, cells were stained with NucBlue Fixed Cell ReadyProbes reagent (Thermo) in PBS for 5 min at room temperature. Coverslips were washed with ultrapure water (Gibco) and mounted in ProLong Gold antifade (Life Technologies). Sample were visualized with a Leica TCS SP8 confocal microscope (Leica Microsystems), and a white light excitation laser and a 405-nm diode laser were used for excitation. Confocal images were taken with an automatically optimized pixel format, a 4× frame averaging, and a scan speed of 400 Hz through an HC PL APO CS2 63× NA 1.4 oil immersion objective. Overlay pictures of single channel images were digitally processed in Leica LAS X lite software. For live imaging, 5.4 × 104 to 105 cells were plated per quadrant in a μ‐Dish 35 mm Quad dish (80416; Ibidi). Cells were infected the next day with SARS‐CoV‐2 at an MOI of 1 in culture media supplemented with 2.5% FBS and 1% P/S containing propidium iodide. Transmission and fluorescence images were taken at 37°C every 15 min, for up to 48 h, using a Nikon BioStation IMQ, with three fields for each condition.
Attachment and entry assays.
Cells plated in monolayers were prechilled on ice for 10 min and washed once with cold PBS. Cells were then incubated with SARS-CoV-2 at an MOI of 1 for 1 h on ice. After three washes with cold PBS, half of the cells was lysed in RA1 lysis buffer (Macherey-Nagel) (“on ice”). The second half of the cells was trypsinized for 15 min on ice and 15 min at 37°C after washing the virus inoculum and then washed with PBS and lysed (“on ice + trypsin”). The remaining cells were directly transferred to 37°C after washing of the virus inoculum and incubated for 2 or 6 h in warm culture media supplemented with 2% FBS and 1% P/S. After this incubation period, these cells were trypsinized for 30 min at 37°C, washed with PBS, and lysed in RA1 buffer (“2 h” and “6 h”). Finally, the total RNA was extracted from all cell lysates using a NucleoSpin RNA II kit (Macherey-Nagel).
VSV-based entry assays.
VSV encoding GFP has been previously described as VSV* (
81). The chimeric virus VSV*ΔG-SARS-CoV-2-S
Δ21 (VSV*ΔG-S), which lacks the homotypic glycoprotein G but rather encodes the spike protein of SARS-CoV-2 (Wuhan-Hu-1 strain), along with GFP, has recently been described (
42). Cells were seeded at 1 × 10
5 cells per well in 24-well plates in DMEM with 1% FBS. The next day, cells were infected with VSV* (at the indicated MOI) or VSV*ΔG-S (MOI of 7) in DMEM without FBS. The virus suspension was removed after 2 h and replaced with DMEM with 1% FBS. At 16 hpi, the cells were washed once with PBS, trypsinized, and subsequently fixed in 4% PFA. Fixed cells were washed once with PBS and analyzed by flow cytometry. The percentage of infected cells was identified based on GFP expression.
Transmission electron microscopy.
Infected and mock-infected MmBr-ACE2 cells were fixed at 16 hpi by incubation for 24 h in 1% glutaraldehyde/4% paraformaldehyde (Sigma, St. Louis, MO) in 0.1 M phosphate buffer (pH 7.2). Samples were then washed in PBS and postfixed by incubation for 1 h with 2% osmium tetroxide (Agar Scientific, Stansted, UK). The cells were then fully dehydrated in a graded series of ethanol solutions and propylene oxide. They were impregnated with a 1:1 mixture of propylene oxide/Epon resin (Sigma) and left overnight in pure resin. Samples were then embedded in Epon resin (Sigma), which was allowed to polymerize for 48 h at 60°C. Ultrathin sections (90 nm) of these blocks were obtained with a Leica EM UC7 ultramicrotome (Wetzlar, Germany). Sections were stained with 2% uranyl acetate (Agar Scientific) and 5% lead citrate (Sigma), and observations were made with a transmission electron microscope (JEM-1011; JEOL, Tokyo, Japan).
Poly(I·C) stimulation.
Cells were plated in monolayers in 24-well culture plates. The next day, they were transfected with 250 ng low-molecular-weight poly(I·C) (InvivoGen) or PBS, respectively, using INTERFERin (Polyplus Transfection) transfection reagent. Cells were lysed 16 h after transfection, and the total RNA was extracted using a NucleoSpin RNA II kit (Macherey-Nagel).
Statistical analysis.
Graphical representation and statistical analyses were performed using Prism version 9.0.2 software (GraphPad). Unless otherwise stated, results are shown as means ± the standard deviations (SD) from three independent experiments. Significance was calculated using either Dunnett’s multiple-comparison test with two-way analysis of variance (ANOVA) or Tukey’s multiple-comparison test with two-way ANOVA, as indicated. Statistically significant differences are indicated by asterisks (*, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001; ns, not significant).
ACKNOWLEDGMENTS
We thank Noémie Aurine (SBRI, U1208 INSERM, France) for help designing RT-qPCR primers for bat samples; Ondine Filippi-Codaccioni and Marc López-Roig (Université Lyon 1, France) for help in bat sampling; the French National Reference Centre for Respiratory Viruses hosted by Institut Pasteur (France) and headed by S. van der Werf for providing the historical viral strains; Hugo Mouquet and Cyril Planchais (Institut Pasteur) for providing anti-S antibodies; Françoise Porrot (Institut Pasteur) for lentiviral production; Florence Guivel-Benhassine (Institut Pasteur) for help in titration assays; and Matthias Lenk (Friedrich-Loeffler-Institut, Germany) for providing the E. serotinus and Tadarida brasiliensis cell lines. We are grateful to the members of our laboratories for helpful discussions. We acknowledge the UTechS Photonic BioImaging (Imagopole), C2RT, Institut Pasteur, supported by the French National Research Agency (France BioImaging; ANR-10-INBS-04; Investments for the Future) for the use of the confocal microscope.
This study was funded by the CNRS (N.J. and O.S.), Institut Pasteur (N.J., L.D., and O.S.), the “Urgence COVID-19” fundraising campaign of Institut Pasteur (N.J., L.D., and O.S.), the University of Veterinary Sciences Brno (FVHE/Pikula/ITA2021) (J.P.), the FINOVI Fondation (AO13) and Covid IDEX Université Lyon 1 (B.P.), LabEx Ecofect (ANR-11-LABX-0048) (Do.P.), Labex IBEID (ANR-10-LABX-62-IBEID) (O.S.), ANR/FRM Flash Covid PROTEO-SARS-CoV-2 (O.S.), and IDISCOVR (O.S.). S.-M.A. and De.P. are supported by the Pasteur-Paris University International Ph.D. Program and the Vaccine Research Institute (ANR-10-LABX-77), respectively. D.L. was funded by the Chinese Scholarship Council and Institut Pasteur. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
We declare that no competing interests exist.