Study design and participants
We recruited patients between February 11th, 2020 and March 1st, 2020 at Yongjia People’s Hospital and Yongjia Center for Disease Control and Prevention in Zhejiang Province, China. All the patients with COVID–19 were clinically diagnosed and laboratory-confirmed. A laboratory-confirmed case of COVID–19 was defined as a positive result on high-throughput sequencing or real-time reverse-transcriptase polymerase-chain-reaction (RT-PCR) assay of nasal and pharyngeal swab specimens on the basis of the WHO interim guidance (WHO, 2020). The criteria of recovery were normal temperature for at least 3 days, obvious improvement in clinical symptoms, significant absorption of pulmonary inflammation on computer tomography scan and negative tests for SARS-CoV–2 two times in a row with a test interval for at least one day. Patients tested positive for HIV, HBV, HAV, HCV or syphilis were excluded. All the clinical data were all reviewed by a trained team of physicians from department of respiratory, intensive care and infectious diseases clinicians. Patients without the baseline assessment test were excluded. 23 patients and two healthy volunteers were included in the samples.The sample size is sufficient to find blood with high antibody titer against the SARS-CoV–2 S protein for the subsequent antibody isolation.
Blood samples were collected and peripheral blood mononuclear cells (PBMCs) and plasma were isolated and frozen for subsequent analysis between 3–4 days after patients recovered from COVID–19. The plasma was titered against the SARS-CoV–2 antigens and two samples with the highest titer were chosen for further analysis. The study was approved by the Ethics Committee of Xinhua Hospital affiliated to Shanghai Jiao Tong university, Yongjia People’s Hospital and Yongjia Center for Disease Control and Prevention in Zhejiang Province. A written informed consent was achieved from patients or their legal guardian.
PBMC and serum sample preparation
Blood samples were collected 3–4 days after donors were discharged from the hospital and separated into plasma and peripheral blood mononuclear cells (PBMCs) by Ficoll-Hypaque gradient centrifugation. Plasmas and PBMCs were maintained in freezing media and stored at –80oC. The plasma was heat-inactivated at 56oC for 1h before use.
Titer measurements by ELISA
The ELISA protocol was adapted from previously established protocols. 384 well plates were coated overnight at 4°C with PBS containing 1 μg/mL of the respective protein S1 RBD-mFc tag (SinoBiological, catalog#40592-V05H) orFull length S-his (SinoBiological, catalog#40589-V08B1) orS1-mFc (Sinobiological, catalog#40591-V05H1 or S1-his (Kactus, catalog# COV-VM4S1). The next day the plate was washed 4 times with washing buffer (PBS and 0.05% Tween) and then incubated 1 hour at 37°C in blocking buffer (PBS with 2% BSA). After two washes the plate was incubated for 1 hour at 37 °C with the serum or the positive control ACE2 protein (Sinobiological,catalog#10108-H08H). The human serum samples were diluted to 1:100 in PBS + 2% BSA followed by 5-fold serial dilutions. The plates were then washed 4 times and incubated for 1 hour in blocking buffer (PBS with 0.05% Tween and 1% BSA) containing diluted (1:5000) secondary antibody (HRP labeled mouse anti-human IgG Fc antibody,Thermo Fisher, catalog#05–4220, RRID: AB_ 2532922, clone name: HP6017, Lot number# UC282110) for 1 hour at room temperature. Following this the plate was washed again 4 times and developed in TMB substrate (Biolegend, catalog#421101) for 5 min before stopping the reaction with the stop solution (Solarbio,catalog# C1058).
Flow cytometry for B cell immune profiling
PBMC was thawed at 37°C and then centrifuged at 450 g for 8 min. The supernatant was discarded, and the cells resuspended in 200 μL of DMEM (Gibco, catalog#11995–065). Following the addition of 1μL of Dnase I (Qiagen, catalog#79254), cells were incubated for 3 min and spun down again. The pellet was resuspended in 20μL of FcR Blocking Reagent (Miltenyi Biotec, catalog#130–059–901), incubated for 10 mins and centrifuged. The cells were suspended in 200μL PBS. 3μL (1:70 dilution) of anti-CD19 (FITC labeled, eBioscience, catalog#11–0199–42, RRID: AB_ AB_10669461, clone name: HIB19, Lot number#1995396), anti-CD27 (APC labeled, eBiosciences, catalog#17–0279–42, RRID: AB_10671130, clone name: O323, Lot number#2087761), anti-CD38 (PE labeled mouse IgG1 isotype control, eBioscience, catalog#12–0388–42, RRID: 1989502, clone name: HB7, Lot number#1989502) or its isotype PE Mouse isotype control (BioLegend, catalog#400114, clone name: MOPC–21, Lot number#4307319) and FITC labeled mouse IgG1 isotype control (ebioscience, catalog#11–4714–41, RRID: AB_10598647, clone name: P3.6.2.8.1, Lot number#4307319) and APC labeled mouse IgG1 isotype control ( BD Biosciences, catalog#550854, clone name: MOPC–21, Lot number#7215834) was then added, and incubated for 30 min at room temperature. Following centrifugation, cells were resuspended in a 100μL of 4% PFA (Beyotime, catalog#P0099–500ml). After 10 min the cells were washed twice by centrifugation and finally resuspended in PBS and ready for flow cytometry analysis using a Cytoflex(Beckman Coulter). The median fluorescence intensity (MFI) was calculated with FlowJo (version 10.6.0).
Patient sample information for FACS
|
labeling
|
Cell Number
|
Viability (%)
|
Gender
|
PBMC
|
Patient 4
|
2.54×105/mL, 200μL
|
84
|
Female
|
PBMC
|
Patient 20
|
7.1×105/mL, 200μL
|
90
|
Male
|
PBMC
|
Healthy donor
|
7.0×105/mL, 200μL
|
92
|
Male
|
Isolation of S protein specific B cells
Avitag and His tag SARS-CoV–2 S protein was expressed in human embryonic kidney cell 293-F (ThermoFisher, catalog# R79007. The cell line was not authenticated.) which was tested negative for mycoplasma contamination. The S protein was purified using HISTRAP HP column(GE, catalog#17–5248–01) and biotinylated using Biotin-Protein Ligase (GeneCopoeia, catalog#BI001). The B cells were stained with biotinylated S protein and incubated at 4℃ for 1 hour. After incubation, the cells were washed three times with PBS. Subsequently, the cells were labeled with Streptavidin microbeads (Miltenyi Biotec, catalog# 130–048–101) at 4℃ for 1 hour. After the incubation, the cell suspension was loaded onto a MACS column which is placed on a magnetic field of a MACS separator. The column was washed three times so the magnetically labeled B cells was retained in the column and unlabeled cells passed through. After removing from the MACS separator, the magnetically labeled B cells were eluted. The isolated B cells were counted by using 0.4% (w/v) Trypan blue stain.
Single-cell BCR sequencing
Enriched S protein specific B cells were individually co-compartmentalized in droplets with single barcoded hydrogel beads and lysis and reverse transcription reagents using a microfluidic device as described15. Droplets of ~1 nL volume were formed at 250 s–1. The droplets were collected in a 1.5mL tube containing HFE–7500 (3MTM, catalog#Novec 7500) and 0.1% surfactant (RanBio, catalog#008-FluoroSurfactant), UV photo-cleaved for 90 seconds (OmniCure ac475–365) and incubated at 50°C for cell lysis and cDNA synthesis. Reverse transcription of VH and VL mRNAs from single B cells took place in droplet using barcoded primers carrying the T7-SBS12 sequences followed by barcode and gene-specific primer sequences complementary to heavy chain J genes and light chain constant region sequences.
The emulsion containing the barcoded cDNA was broken by adding one volume of 1H,1H,2H,2H-Perfluoro–1-octanol (Sigma-Aldrich, catalog#370533–25G) after the droplet RT reaction finished. The pooled, barcoded cDNAs were purified with Agencourt RNA CleanUp beads (Beckman, catalog#A63987) at a 1:1 ratio (vol/vol) twice and eluted in 60 μL DNase- and RNase-free H2O. The sequencing library was generated by two-step nested PCR using GoTaq Polymerase (Promega, catalog#M7406). In the first PCR, forward primers were priming on the T7 and with reverse primers priming on the VH, Vλ and Vκ leader and framework 1 sequenced of the V genes. In the second PCR, the forward primer appends the Illumina P7 and Illumina index sequences by priming on the SBS12 sequence and the reverse primer appends the Illumina P5 and SBS3 sequence. The approximately 550 bp final PCR products were extracted by agarose gel electrophoresis (Qiagen, catalog#28606). The constructed NGS libraries were sent out for sequencing using Illumina MiSeq PE300 which allows sequencing of the entire VH and VL domain as well as the barcode sequence (GeneScript sequencing service supplier) with data varying from 6–12 million reads per samples. The resulting FASTQ data was analyzed by a bioinformatics pipeline enable trimming, merging, barcode extraction and clustering as described16. Briefly, paired-end reads were first trimmed at the 3’ end to remove low quality score bases then merged using the program FLASH requiring at least 10bp overlap. The barcodes were extracted form merged reads followed by clustering requiring the DNA sharing at least 93% identify. The consensus sequence was created from clusters by aligning up to 200 sequencing using ClustalO(version1.2), and each antibody sequence was characterized for immunoglobulin content using VDJFasta(version 2.0). We also applied a minimum number of reads 10 for VH and VL. VH-VL pairing was carried out by identifying the most abundant VH and VL consensus sequence (by number of reads that contributed to that in each barcode cluster).
Production of recombinant antibody
The DNA of variable regions of the heavy and light chains were synthesized by Genewiz and cloned into expression plasmids containing the human IgG1 heavy chain and kappa light chain constant regions. The antibodies were expressed in 293F cell for 5 days after the co-transfection of both heavy and light chain expression plasmids. Antibodies were purified from cell culture supernatants using Protein-A affinity chromatography.
Antibody binding and competition with receptor ACE2
The binding affinity of antibodies to S protein was analyzed by ELISA. 384 well plate (Corning, catalog#3700), was coated overnight at 4°C with PBS containing 30μL 20nM of the SARS-CoV–2 Spike S1+S2 ECD, his Tag protein (SinoBiological, catalog#40589-V08B1). The next day the plate was washed 5 times with washing buffer (PBS and 0.05% Tween) and then incubated 1 hour at room temperature in blocking buffer (PBS with 2% BSA). After 5 washes the plate was incubated with serial dilution of purified antibodies for 1 hour at room temperature. The plates were then washed 5 times and incubated for 1 hour in blocking buffer(PBS with 0.05% Tween and 1% BSA) containing Mouse anti-Human IgG Fc HRP labeled (Thermo Fisher, catalog#05–4220, RRID: AB_ 2532922, clone name: HP6017, Lot number# UC282110, dilution 1:5000) for 1 hour at room temperature. Following this the plate was washed again 5 times and developed in TMB substrate (Biolegend, catalog#421101) for 5 min before stopping the reaction with the stop solution (Solarbio, catalog#C1058). The OD values were determined using Thermo MultiSkan or MD SpectraMax i3X at 450nm wavelength, data was analyzed with GraphPad Prism (version 8.0.1).
The blocking with receptor ACE2 was performed using cell surface expressed ACE2. 10nM SARS-CoV–2 spike S1, mFc tag protein (SinoBiological, catalog#40591-V05H1) was incubated with serial dilution of purified antibodies at room temperature for 1 hour and then added to Vero E6 cells (ATCC,CRL–1587. The cell line was validated by CoBIOER (http://www.co-bioer.com/) and tested negative for mycoplasma contamination.These cells exhibit cytopathic effects when infected with multiple virus. Plaques are also produced. 105 cellsper well) in duplicate. Then detection reagent rabbit anti mouse IgG Fc-AF647 (Jackson ImmunoResearch, catalog#315–606–046, RRID: AB_2340251, 1:800 dilution) was used. Half-maximal inhibitory concentration (IC50) of the evaluated antibodies was determined with flow cytometry (Beckman, Cytoflex) and FlowJo software (version 10.6.0) analysis.
Antibody neutralization activity against pseudovirus
Murine leukemia virus-based SARS-CoV–2 S pseudotyped virus were prepared by GenScript. Human embryonic kidney cell HEK293T cells (ATCC,catalog# CRL–3216, the cell line was tested negative for mycoplasma contamination. The cell line was not authenticated.) were co-transfected with envelop-expressing plasmid for SARS-CoV–2 Spike protein and HIV–1 backbone plasmids expressing packaging proteins and luciferase reporter at a mass ratio of 1:3 using Polyethylenimine (PEI) (Polyscience, catalog#23966–1) according to the manufacture’s instruction. Forty-eight hours after transfection, the supernatant containing pseudovirus was harvested and filtered with micropore filter (0.45 μm) followed by ultracentrifugation at 83,000 × g, 4℃ for 2 h. The pseudovirus pellet was resuspended in DPBS and stored at –196°C. The titer of pseudovirus was determined by p24 ELISA kit (Takara, catalog# 632200) following the manufacture’s instruction.
As for target cell establishment, HeLa cells (human cervix carcinoma cell, ATCC,catalog# CCL–2, the cell line was tested negative for mycoplasma contamination. The cell line was not authenticated.) were transduced with lentivirus containing codon optimized human ACE2 CDS sequence (Uniprot ID: Q9BYF1) followed by single clone selection. The expression of ACE2 was validated with Spike-ECD (GenScript, catalog#Z03481).
Neutralization assay were performed by incubating pseudovirus with serial dilution of purified antibodies at room temperature for 1hour (starting from 100ug/ml, 3 folds dilution for 8 points). ACE2 overexpression HeLa cells (approximately 8×104 per well) were cultured in DMEM containing 10% FBS, 1μg/mL puromycin were added in triplicate into virus-antibody mixture. Following infection at 37oC 5%CO2 for 48 hours, luciferase activity was determined using Promega Bio-Glo luciferase assay (Promega, catalog#G7491) system. The dose-response curves were plotted with the relative luminescence unit against the sample concentration. The assay results were processed by Microsoft Office Excel 2016 and GraphPad Prism 6. GenScript has applied this peudovirus neutralization assay to many other SARS-COV–2 neutralizing antibodies.
Live virus assay
Microneutralization assays towards live virus were performed as previously described1 with slight modifications. This Vero E6 cell line (ATCC, CRL–1586, the clone is different from the one used for in vitro blocking. The cell line was not authenticated) have been tested negative for mycoplasma contamination using a commercial EZ-PCRTM Mycoplasma Test kit (Biological Industries, Beit-Haemek, Israel; 20–700–20; Lot#: 1251719) (data not shown) and have been used for culture of the live virus1. Briefly, Vero E6 cells seeded in 96-well plates were incubated with SARS-CoV–2 (100 TCID50 per well) (BetaCoV/Wuhan/WIV04/2019; National Virus Resource Center, Wuhan, China; IVCAS 6.7512) in absence or presence of diluted antibodies for 1 hour, fixed with 4% paraformaldehyde diluted in PBS for 15 minutes at room temperature followed by penetrating solution containing 0.25% triton-X 100 for 10–15 minutes. After three washes, cells were blocked at 37 ℃ for 1 hour using PBS containing 5% BSA, then incubated with in-house prepared anti-SARS-CoV–2 NP rabbit serum (1:1000 dilution) as primary antibody and Goat Anti-Rabbit IgG H&L (Alexa Fluor® 488) (1:500 dilution, Abcam, Cambridge, UK; ab150077; Lot#: GR3244688–2) as the secondary antibody. Specificity of the primary antibody to SAR-CoV–2 NP was validated by Western Blot with pCAGGS-SARS-CoV–2 NP transfected cells (data not shown). Cell nuclei were strained using Hoechst 33258 (Beyotime, Shanghai, China; C1018) at room temperature for 10 minutes. Images were taken using an Operetta CLSTM system (PerkinElmer, Waltham, USA), which counts numbers of nuclei and cells infected with viruses, respectively. Inhibition was calculated by the number of non-infected cells over the number of total nuclei. IC50 and IC90 were calculated with GraphPad Prism 8.0.
Surface plasmon resonance analysis.
SPR experiments were performed using Biacore T200 system (GE Healthcare). In brief, experiments were performed at 25°C in HBS-EP+ buffer. Antibody was immobilized onto a protein A sensor chip (GE healthcare, catalog#29139131-AA). Serially diluted SARS-CoV–2 RBD (WT, AcroBiosystems, catalog#SPD-C52H3), RBD (N354D/D364Y, AcroBiosystems, catalog#SPD-S52H3), RBD (R408I, AcroBiosystems, catalog#SPD-S52H8), RBD (W436R, AcroBiosystems, catalog#SPD-S52H7), RBD (V367F, AcroBiosystems, catalog#SPD-S52H4)or SARS-CoV–2 spike S1 domain (D614G, Sino Biological, catalog#40591-V08H3) were injected through flow cells for 60s of association followed by a 150s dissociation phase at a flow rate of 30 μL min−1. Prior to next cycle, the sensor surface was regenerated with Glycine-HCl (pH 1.5) for 30s at a flow rate of 30 μL min−1.KD values were calculated using the 1:1 binding kinetics model.
For all the experiments described above, the researchers were not blinded to group allocation during data collection and analysis. The experiments of Figures 4, 5 and 6 were successfully replicated twice. The experiment of immune profiling of B cells of COVID–19 patients by flow cytometry was not replicated because the very limited amount of blood samples. All the detailed information,validation statements and relevant citations of primary antibodies can be found on the manufacturer’s website of the supplier and Certificates of the antibodies can be searched by the lot numbers.