Cell lines
Human lung adenocarcinoma epithelial Calu-3 cells (ATCC-HTB-55) were cultured in Eagle’s Minimum Essential Medium (EMEM). African green monkey kidney Vero E6 cells (ATCC-CRL-1586) and human kidney HEK29T cells (ATCC-CRL-3216) were cultured in Dulbecco’s Modified Eagle's medium (DMEM). All media were supplemented with 10% FBS and 1% penicillin/streptomycin. Vero E6 cells stably expressing TMPRSS2 (Vero E6-TMPRSS2) were established and cultured in DMEM in the presence of puromycin (1µg/ml).
All cells had been previously tested for mycoplasma contamination and incubated at 37°C in a humidified atmosphere with 5% CO2.
Primary airway epithelial cells (AECs)
Primary AECs were obtained and cultured as previously described28. Briefly, Human unused donor tracheobronchial tissue was obtained at the time of lung transplant. The tissue was washed and placed in DMEM with 0.1% protease and antibiotics overnight at 4ºC. The next day, the solution was agitated, and the remaining tissue was removed. Cell pellets were treated with 0.05% trypsin-EDTA, then filtered through a cell strainer. Cells were plated onto 6mm/0.4mm Transwell ALI insert after treatment with FNC coating mixture. 10% FBS in DMEM and ALI media were added in equal volumes to each basal compartment and cultures were incubated at 37ºC with 5% CO2. The next day, the media was removed and both compartments were washed with PBS and antibiotics. ALI media was then added to each basal compartment and changed every three days for at least 28 days until differentiated airways were ready for use .
Viruses
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) strains USA-WA1/2022, lineage P.1, lineage B.1.617.2, and lineage B.1.1.529 were obtained from BEI Resources of the National Institute of Allergy and Infectious Diseases (NIAID) and propagated in Vero E6-TMPRSS2 cells. Virus titer was measured in Vero E6 cells by TCID50 assay. All the studies involving live viruses were conducted in the Vitalant Research Institute BSL-3 under approved safety protocols.
Ethics Statement
The studies involving human participants were reviewed and approved by the Human Research Protection Program, University of California, San Francisco. The patients/participants provided their written informed consent to participate in this study.
Preparation of PAV-104
The synthetic method of PAV-104 was illustrated in Fig. 1. To a solution of aldehyde1 (10 g, 65.79 mmol, 1.0 eq) in toluene was added 2,4-dimethoxybenzyl amine 2 (10.99 g, 65.79 mmol, 1.0 eq) and the reaction mixture was heated at 80°C for 24 h. Solvent was removed and the residue was taken in MeOH and cooled using an ice bath. Then sodium borohydride (4.97g, 131.58 mmol, 2.0 eq) was added slowly and the reaction mixture was stirred at room temperature for 12 h. Solvent was removed and residue was taken in ethyl acetate and then sat. NaHCO3 was added and stirred for 1 h. Organic layer was separated, dried (MgSO4) and solvent was removed to give amine 3, which was used in the next step without further purification.
To a solution of the crude amine 3 (5.0 g, 19.1 mmol, 1.0 eq) in DMF (25 mL) were added acid 4 (3.17 g, 19.1 mmol, 1.0 eq), HATU (8.7 g, 22.92 mmol, 1.2 eq,), and DIEA (12.32 g, 95.5 mmol, 5.0 eq) and the reaction mixture was stirred at room temperature for 12 h. The reaction mixture was then diluted with ethyl acetate (EtOAc) and washed with 10% aqueous HCl (1X), sat. NaHCO3 (1X) and water (3X). Organic layer was collected, dried (MgSO4) and evaporated to give a residue, which was taken in MeOH and then K2CO3 (2.64 g. 19.1 mmol, 1.0 eq) was added and stirred at room temperature for 12 h. Solvent was removed and the residue was taken in Ethyl acetate and washed with 10% HCl (1X). Organic layer was separated, dried and solvent was removed to give a residue, which was purified by column chromatography (EtOAc/ Hexane) to give compound 5.
To a stirred solution of compound 5 (1.0 g, 2.22 mmol, 1.0 eq) and cesium carbonate (1.08 g, 3.33 mmol, 1.5 eq) in DMF (15 mL) was added methyl 4-(chloromethyl) benzoate 6 (450 mg, 2.44 mmol, 1.2 eq) and the reaction mixture was stirred at room temperature for 18 h. The reaction mixture was diluted with ethyl acetate and washed with water (3x). Organic layer was dried and concentrated to give crude product 7. The crude compound 7 was stirred in a 1:1 mixture of TFA: DCM for 12 h. Concentration followed by chromatography purification (Hexane/ EtOAc) provided compound 8.
To a stirred solution of compound 8 (0.84 mmol, 1.0 eq) in 3:1 mixture of THF: H2O (12 mL) was added LiOH (40 mg, 1.68 mmol, 2.0 eq) and the reaction mixture was stirred at 65°C for 12 h. The reaction mixture was evaporated under vacuum to give a residue, which was stirred in a mixture of 10% aqueous HCl and ethyl acetate for 30 min. Organic layer was collected, washed (H2O, 1X), dried and concentrated to give crude acid 9.
To a solution of the amine 10 (68 mg, 0.552 mmol, 1.2 eq) in DMF (25 mL) were added acid 9 (200 mg, 0.46 mmol, 1.0 eq), HATU (210 mg, 0.552 mmol, 1.2 eq,) and DIEA (0.300 mg, 2.3 mmol, 5.0 eq). The reaction mixture was stirred at room temperature for 12 h. The reaction mixture was then diluted with EtOAc and washed with 10% aqueous HCl (1X), sat. NaHCO3 (1X) and water (3X). Organic layer was collected, dried (MgSO4) and evaporated to give a residue, which was purified by column chromatography (EtOAc/ Hexane) to give PAV-104.
Drug cytotoxicity assay
The cytotoxic effect of PAV-104 on Calu-3 cells was measured using an MTT assay kit (Abcam, ab211091) following the manufacturer’s instructions. In brief, Calu-3 cells were seeded in 96-well cell culture plates. Appropriate concentrations of PAV-104 were added to the medium (0-5000 nM). After 48 hours, the media was removed and 100 µl MTT reagent (1:1 dilution in DMEM medium (serum free)) was added to each well and incubated for 3 h at 37ºC. Then the medium was removed, and 150 µl MTT solvent was added into each well. Quantification was performed by reading absorbance at OD = 590 nm. The data from three independent experiments was used to calculate the CC50 by nonlinear regression using GraphPad Prism 8.0 software.
SARS-CoV-2 infection and drug administration
Calu-3 cells were seeded at 0.5 x 106 cells per well in 0.5 ml volumes using a 24-well plate, or were seeded at 1 x 105 cells per well in 0.1 ml volumes using a 96-well plate. The following day, cells were pretreated with or without PAV-104 or remdesivir for one hour. Then viral inoculum (MOI of 0.01; 500 µl/well or 100µl/well) was prepared using EMEM containing indicated concentrations of PAV-104 or remdesivir and added to the wells. The inoculated plates were incubated at 37ºC with 5% CO2. At indicated infection time points, supernatants were collected and stored at -80ºC. Cells were lysed with TRizol (Thermo Fisher Scientific, 15596026) for RNA extraction.
For infection of primary AECs in ALI culture, cells were pretreated with PAV-104 in the basal compartment for one hour. SARS-CoV-2 (diluted in ALI-culture medium, MOI = 0.1) was added to the apical chamber of inserts (250 µl) and the basal compartment (500 µl). Then the cultures were incubated for 2 hours at 37ºC (5% CO2) to enable virus entry. Subsequently, the cells were washed and fresh ALI medium (500 µl) containing PAV-104 was added into the basal compartment. Cells were incubated at 37ºC (5% CO2) and harvested for analysis at 36 hours post-infection.
Viral titer by TCID assay
Virus production in the supernatant was measured by quantifying TCID50. Vero E6 cells were plated in 96-well plates at 5 X 104 cells per well. The next day, supernatants collected from Calu-3 cells were subjected to 10-fold serial dilutions (101 to 1011) and inoculated onto Vero E6 cells. The cells were incubated at 37ºC with 5% CO2. Three to five days post infection, each inoculated well was evaluated for presence or absence of viral CPE. TCID50 was calculated based on the method of Reed and Muench.
RT-qPCR
Total RNA was extracted using TRIzol reagent according to the manufacturer's instructions. Reverse transcription was performed using RevertAid First Strand cDNA Synthesis Kit (Thermo Fisher Scientific, K1622) in accordance with the manufacturer's instructions. RT-qPCR was performed for each sample using Taqman Universal Master mix II, with UNG (Thermo Fisher Scientific, 4440038) on a ViiA7 Real time PCR system. Primers and probes for detection of the RNaseP gene and SARS-CoV-2 nucleocapsid (N) gene were obtained from IDT (2019-nCoV RUO Kit (Integrated DNA Technologies, 10006713)). The expression level of the N gene was determined relative to the endogenous control of the cellular RNaseP gene.
RNA-sequencing analysis
RNA concentration and quality was measured using High Sensitivity RNA ScreenTape Analysis (Agilent, 5067 − 1500). cDNA libraries were constructed and sequencing was performed by Novogene using their mRNA sequencing protocol. The raw RNA sequencing data were aligned to the human genome (GRCh38) using STAR (version 2.7.3a). Analysis of differential expression was performed using DESeq2 according to a standard protocol. Genes with adjusted P-value < 0.05 were considered as significantly differentially expressed. Gene set enrichment analysis was performed using the fgsea package (version 1.22.0) in R. The Reactome database (version 7.5.1) was downloaded from MSigDB (https://www.gsea-msigdb.org).
Immunofluorescence microscopy and image analysis
Cells were fixed and permeabilized with cold methanol : acetone (1:1) for 10 min at 4℃ according to our previous method28. In brief, cells were then incubated with blocking buffer (5% goat serum (Seracare Life Sciences Inc, 55600007)), a primary antibody (monoclonal rabbit anti-SARS-CoV-2 N antibody (GeneTex, GTX135357)), a secondary antibody (Goat anti-Rabbit IgG (H + L) secondary antibody, FITC (Thermo Fisher, 65-6111)), and DAPI (Thermo Fisher Scientific, D1306). Images were acquired using a fluorescence microscope.
To measure the frequency of infected cells, randomly-selected areas were imaged. Each treatment had three replicates. The FITC-positive cells and DAPI-positive cells were quantified using CellProfiler software as previously described. The same threshold value was applied to the images of each area.
Quantification of the western blots was carried out with Image J software.
Production of SARS-CoV-2 virus-like particles (VLPs)
HEK293T cells were seeded in T75 cell culture flasks. The next day, cells were transfected with empty pcDNA3.1 plasmid or pcDNA3.1 plasmid encoding the SARS-CoV-2 M (Addgene-158078), E (Addgene-158080), N (Addgene-158079), and S proteins (Addgene-158074), as indicated. 1 µg of each plasmid was used, with 5 µg of total plasmid in each transfection, normalized using empty vectors, in 400 µl Opti-MEM and 18 µl of PEI. The transfection mixture was incubated at room temperature for 15 min and dropped into the HEK293T cells. Six hours post transfection, the media was removed and supplemented with fresh medium containing PAV-104 at indicated concentrations. The supernatant and cell lysate were collected after 60 hours. For the purification of SARS-CoV-2 VLPs, the supernatant was passed through a 0.45 µm syringe filter, then loaded on top of a 20% sucrose cushion in PBS, and ultracentrifuged at 30,000 rpm in an SW41 rotor for two hours. VLP-containing pellets were washed with ice cold PBS and resuspended in SDS loading buffer, followed by sonication in an ice-water bath. Or VLP-containing pellets were resuspended in PBS (passed through 0.22 µm syringe filter) for quantification by NTA. Cells were lysed in RIPA buffer (Thermo Fisher Scientific, 89900) and sonicated in an ice-water bath.
Immunoblots of SARS-CoV-2 VLPs
Total protein in pellet and cell lysate samples were separated by SDS-PAGE, and subsequently electro-transferred onto a supported PVDF membrane. Membranes were cut and probed for M with rabbit anti-SARS-CoV-2 M (Thermo Fisher Scientific, PA1-41160), N with rabbit anti-SARS-CoV-2 N (Rockland Immunochemicals, 200-401-A50), E with rabbit anti-SARS-CoV-2 E (Thermo Fisher Scientific, PA5-112047), and S with rabbit anti-SARS-CoV-2 S1/S2 (Thermo Fisher Scientific, PA5-112048) or with mouse anti-SARS-CoV-2 spike (Genetex Inc, GTX632604). Goat anti-rabbit IgG HRP and goat anti-mouse IgG HRP secondary antibodies were used as appropriate. β-actin was used as a cell lysate and pellet loading control by probing membranes with rabbit anti-human β-actin, conjugated with HRP (Cell Signaling Technology, 12620). All antibodies were diluted in 5% milk and membranes were washed with Tween 20 washing buffer (Thermo Fisher Scientific, J60304.K3). Chemiluminescent signal was visualized using SuperSignal West Femo Substrate (Thermo Fisher Scientific, PI34094) or using ECL Blotting Reagents (SIGMA, GERPN2109), and imaged using ImageQuant LAS 4000.
Quantification of VLPs by nanoparticle tracking analysis (NTA)
VLP-containing pellets were diluted in PBS (passed through 0.22 µm syringe filter) to a concentration in the range of 107-109/ml and examined using a NanoSight NS300 (NanoSight Ltd) equipped with a 405 nm laser. Five 60 s-long videos were taken for each sample with camera level 16 and the detection threshold set at 5. Raw data of particle movement and laser scattering were analyzed using NTA software (version 3.3, NanoSight Ltd). The output data were presented as nanoparticle concentration and size.
Drug Resin Affinity Chromatography (DRAC)
Drug Resin Affinity Chromatography experiments were performed where 30 µl of extract prepared from Calu3 cells under different infection and treatment conditions were adjusted to a protein concentration of approximately 2.3 mg/ml in column buffer and supplemented with an “energy cocktail” (to a final concentration of 1mM rATP, 1mM rGTP, 1mM rCTP, 1mM UTP, 4mM creatine phosphate, pH 7.6) and 5 µg/mL creatine kinase) and incubated on a column containing 30 µl of affi-gel resin coupled to either PAV-104 or a 4% agarose matrix (control) for one hour at room temperature. The PAV-104 resin conditions were run side-by-side in triplicate, while the control resin conditions were done in single point. The flow-through material was collected, and the resin was washed with 1.5 mL column buffer then eluted with 100 µl PAV-104 plus the energy cocktail at room temperature for two hours then stripped with 100 µl 1% SDS. The eluate and SDS-stripped material run on agarose gels and are analyzed by western blot for SARS-CoV-2 N protein (Rockland Immunochemicals, 200-401-A50).
Glycerol gradient sedimentation and ELISA-based assessment of SARS-CoV-2 N
Cell extracts from SARS-CoV-2 N-transfected HEK-293T cells in the presence or absence of PAV-104 were centrifuged at 15000 g for 10 mins at 4℃ to obtain the supernatant. 200 µl of supernatant were loaded on the top of a 5 ml continuous 10–40% glycerol gradient in lysis buffer (v/v, Pierce IP Lysis Buffer (Thermo Fisher, 87787)) prepared using the Gradient Master machine (Biocomp, Gradient Station). After concentration at 135000 g for 20 h at 4℃ in a SW55 rotor (Beckman Coulter), 22 fractions of 250 µl were collected from the top to the bottom of the gradient.
Proteins were assessed by the commercial SARS-CoV-2 N protein sandwich ELISA kit (GeneTex, GTX535824) following manufacturer's instructions. In brief, each fraction was diluted to 1:1000 using assay dilute reagent. 50 µl of each standard and samples were added into the appropriate wells, then incubated at room temperature for 2 hours. The solutions in the wells were aspirated and wells were washed with a washing buffer six times. Then the conjugate solution was added and incubated at room temperature for 1 hour. The solutions in the wells were aspirated and wells were washed with a washing buffer six times once again. TMB solution was added to the wells and incubated in darkness for 15 mins at room temperature. Stop solution was added to each well. Finally, optical density at 450 nm was read within 15 mins.
Statistical analysis
Statistical analysis was performed using GraphPad Prism version 8 software. Data were presented as means ± SEM or median. Data were analyzed for statistical significance using an unpaired or paired Student’s t test to compare two groups, or using a paired t test. Only p values of 0.05 or lower were considered statistically significant (p > 0.05 [ns], p ≤ 0.05 [*], p ≤ 0.01 [**], p ≤ 0.001 [***], p ≤ 0.0001 [****]).