Rapid cloning-free mutagenesis of new SARS-CoV-2 variants using a novel reverse genetics platform

  1. Enja Tatjana Kipfer
  2. David Hauser
  3. Martin J Lett
  4. Fabian Otte
  5. Lorena Urda
  6. Yuepeng Zhang
  7. Christopher MR Lang
  8. Mohamed Chami
  9. Christian Mittelholzer
  10. Thomas Klimkait  Is a corresponding author
  1. Molecular Virology, Department of Biomedicine, University of Basel, Switzerland
  2. BioEM Lab, Biozentrum, University of Basel, Mattenstrasse, Switzerland
7 figures, 1 table and 8 additional files

Figures

Figure 1 with 1 supplement
Rescue and characterization of recombinant SARS-CoV-2.

(A) Schematic representation of the SARS-CoV-2 genome and the infectious subgenomic amplicons (ISA)-based method for virus recovery. Eight respectively four overlapping fragments covering the whole SARS-CoV-2 genome were PCR amplified. The heterologous CMV promoter was cloned upstream of the 5′ untranslated region (UTR) and a poly(A) tail, HDV ribozyme, and SV40 termination signal downstream of the 3′ UTR. (B) Infectious virus reconstituted from four fragments (rCoV2-4fr) assessed by cytopathic effect (CPE, top) on susceptible Vero E6 cells by supernatant transfer. Plaque size was compared by standard plaque assay 2 d after inoculation on Vero E6 cells (bottom). (C) Workflow for the rescue of recombinant SARS-CoV-2. Four fragments were PCR amplified, mixed in equimolar ratios, transfected into HEK293T cells, and infectious virus was recovered 7 d post-transfection. Commercially available SARS-CoV-2 rapid antigen tests can be used for a quick non-quantitative analysis. (D) Detection of intracellular SARS-CoV-2 nucleocapsid protein (N, green) and nuclei (Hoechst, blue) in Vero E6 cells infected with parental wild-type or recombinant virus by immunocytochemistry. (E) Growth kinetics of recombinant virus and its parental wild-type virus. Vero E6 cells were infected in triplicates at a multiplicity of infection (MOI) of 0.01, supernatant was collected 12, 24, 48, and 72 hr post-infection and analyzed by plaque assay. Cell layers were washed 2 hr post-infection. Data represents mean ± SEM, analyzed with multiple t-tests and Benjamini, Krieger, and Yekutieli correction (N = 3 individual biological replicates, n = 3 technical replicates). (F) Cryo-transmission electron microscope pictures of parental wild-type virus and recombinant virus in glutaraldehyde-fixed samples. Scale bar is 100 µm (top) and 2 mm (bottom) in (B), 20 µm in (D), and 100 nm in (F).

Figure 1—figure supplement 1
Clonal virus populations and reconstitution efficiency.

(A) Schematic representation of the workflow to generate clonal virus populations. Right after transfection, cells were diluted to less than 0.5 virus-producing cells/well in 96-well plates. Then, 7 d post-transfection, the supernatant was transferred onto Vero E6-TMPRSS2 cells. Plates were observed until a cytopathic effect (CPE) became apparent. (B) Clonal virus populations arising from a single virus-producing cell were identified after supernatant transfer onto Vero E6-TMPRSS2 cells. CPE of infectious virus was assessed by microscopy and virus was collected for further analysis. Thereafter, plates were fixed and stained for the fast enumeration of positive wells (in light blue).

Sequence integrity using CLEVER.

(A) Schematic representation of the sequence alignment of recombinant viruses sequenced by next-generation sequencing (NGS), mutations with a relative abundance of >10% are indicated with a star. A total of eight bulk (gray) and five clonal (green) populations were analyzed. (B) Details on substitution and position in the genome. For a more detailed analysis see Supplementary file 2.

Creating chimeric virus by fragment exchange.

(A) Schematic representation of the exchange of individual fragments. Shown is the replacement of the Wuhan S sequence by the sequence encoding for the Omicron BA.1 or BA.5 S gene. The genetic background (outside of S) is kept in the original Wuhan sequence. All fragments needed to reconstitute the virus were transfected and chimeric virus was rescued. (B) Successful rescue of infectious chimeric virus was assessed by cytopathic effect (CPE) formation on Vero E6 cells. Scale bar represents 100 μm. (C) Titers of neutralizing antibodies against different SARS-CoV-2 S gene variants were validated in sera from vaccinated individuals. Sera were incubated with parental wild-type virus (Wuhan), Omicron BA.1 or BA.5 clinical isolates (BA.1, BA.5), as well as chimeric viruses having the Wuhan background combined with either the Omicron BA.1 S gene (WuhanBA.1 S) or Omicron BA.5 S gene (WuhanBA.5 S). Neutralizing titers were determined with a neutralization assay and TCID50 read-out. Data represents mean ± SEM, analyzed with one-way ANOVA followed by Bonferroni’s test (N = 5).

Figure 4 with 1 supplement
Direct mutagenesis using the CLEVER primer design.

(A) Schematic representation of the CLEVER primer design for direct mutagenesis. Shown is the (i) introduction of small nucleotide changes, (ii) the deletion of larger sequences, here shown for ORF3a, and (iii) the insertion of nucleotide stretches such as 3xFLAG as well as a timeline showing the expected work-flow needed from in silico design to virus rescue. (B) Details on the G614D and N501Y substitution within the S gene. Shown is position, primer design, and the integration into the viral genome confirmed by Sanger sequencing. (C) Validation of mutations by immunoblot. Shown is the validation of the ∆ORF3a (left) and ORF8-3xFLAG virus (right). Vero E6 cells were assessed with α-β-actin (α-β-ACT) and viral infection was detected using α-NSP2. ORF3a expression or ORF8/FLAG expression, respectively, was compared to wild-type infected cells and uninfected controls. (D) Validation of ∆ORF3a by immunocytochemistry. ∆ORF3a virus created by direct mutagenesis was compared to its parental wild-type virus. Shown is the expression of ORF3a (magenta) in both viruses. Nucleocapsid (N, green) expression was used to assess viral infection, nuclei were stained with Hoechst (blue). (E) Validation of ORF8-3xFLAG by immunocytochemistry. C-terminal tagging of ORF8 with 3xFLAG was achieved with direct mutagenesis. Shown is the expression of ORF8 (magenta) and FLAG (green) in both viruses. Nuclei were stained with Hoechst (blue). Scale bar is 20 µm in (D, E).

Figure 4—figure supplement 1
CLEVER primer design for direct mutagenesis.

Shown are the different approaches of primer design to guarantee a 100 bp overlap between the neighboring fragments. Primers can either be separated in distance to ensure homology regions or additional nucleotides must be added to the 5′ end of the primer. Small nucleotide changes, deletions, or insertions can directly be introduced by adjusting the primer annealing sites and the homology regions, as depicted.

Figure 5 with 2 supplements
Direct rescue and mutagenesis of clinical isolates.

(A) Schematic representation of the circular assembly within the eukaryotic cell with the linker fragment. The heterologous elements needed downstream of the 3′ untranslated region (UTR) (pA, HDVr, SV40) and upstream of the 5′ UTR (CMV) are assembled in one fragment, separated by a spacer sequence and flanked by homologous regions needed for intracellular recombination. (B) Representative agarose gel pictures from PCR fragments amplified by one-step RT-PCR from viral RNA and the linker fragment (L). Recombinant virus was rescued from five (top) or eight fragments (bottom), plus the linker fragment. Asterisks mark fragments harboring the introduced changes within their homology region. (C, D) Validation of (C) Omicron BA.5 ∆ORF3a and (D) XBB.1.5 ∆ORF3a by immunocytochemistry. The expression of ORF3a (magenta) in Omicron BA.5 and XBB.1.5 clinical isolates and recombinant ∆ORF3a viruses is shown. Nucleocapsid (N, green) expression was used to assess viral infection, nuclei were stained with Hoechst (blue). Scale bar represents 20 μm.

Figure 5—figure supplement 1
Cloning-free rescue of chimeric virus.

(A) All fragments were directly amplified by one-step RT-PCR from viral RNA of Wuhan, Omicron BA.1, and Omicron BA.5. Eight fragments were amplified to prove high flexibility in exchanging fragments. (B) In addition to the chimeric viruses described in Figure 3, the genetic background (region outside of S) was replaced by the sequence of Omicron BA.1 or Omicron BA.5 and combined with a heterologous S sequence of Wuhan, Omicron BA.1 or BA.5, respectively. (C) Infectious chimeric virus was rescued and assessed via cytopathic effect (CPE) formation on Vero E6 cells. Scale bar represents 100 μm. (D) Scheme for the rapid distinguishment between Wuhan, Omicron BA.1, BA.5, and XBB.1.5 variants. Indicated regions (S or M) were Sanger sequenced to discriminate variants or confirm chimeric viruses. Amino acids are highlighted in yellow for the clear identification of the S gene variant and/or the background (within M).

Figure 5—figure supplement 2
Cloning-free rescue of Chikungunya virus (CHIKV) and Dengue virus (DENV).

(A) Schematic representation of the CHIKV genome and the design for the cloning-free rescue. The genome was divided into three overlapping fragments, and a silent SNP was introduced by PCR (red asterisk). (B, C) Successful recombination of the four PCR products (B) within the eukaryotic cell leads to a circular product and virus production (C), cytopathic effect (CPE) on BHK-21 cells for rCHIKV and negative control. (D) Schematic representation of the DENV genome and the design for the cloning-free rescue. The genome has been divided into two overlapping fragments and a silent SNP has been introduced by PCR (red asterisk). The protocol has been tested on two different clinical isolates (DENV1 and DENV3), whereas two different recombination sites were tested for DENV1 (rDENV1-A and rDENV1-B, only one schematically represented). (E, F) Overlapping PCR products (E) were transfected into BHK-21 cells, and CPE (F) was assessed on VeroE6-TMPRSS2 cells (rDENV1-A, rDENV1-B, rDENV3, and negative control). Scale bar is 100 µm in (C) and (F).

Overview of reverse genetics methods for SARS-CoV-2.

The most commonly used reverse genetics systems for the rescue of recombinant SARS-CoV-2 are listed and the prominent intermediate steps are depicted. Note that this is a schematic summary and additional steps (such as purification, linearization before transcription, etc.) or small aberrations of the protocol (e.g., different starting material) can apply. The first groups reporting the successful adaptation to SARS-CoV-2 are mentioned. For the CLEVER method, additionally the direct mutagenesis within the initial RT-PCR step is depicted (mutated sites marked with red asterisks). Repeated icons are only labeled once. DNA fragments are represented as blue lines. T7, T7 RNA polymerase; BAC, bacterial artificial chromosome; YAC, yeast artificial chromosome; CPER, circular polymerase extension reaction; ISA, infectious subgenomic amplicons; CLEVER, CLoning-free and Exchangeable system for Virus Engineering and Rescue.

Author response image 1
rCoV2∆ORF3678.

Detection of intracellular SARS-CoV-2 nucleocapsid protein (N, green) and nuclei (Hoechst, blue) in Vero E6TMPRSS2 cells infected with rCoV2∆ORF3678 by immunocytochemistry. Scalebar is 200 µm in overview and 50 µm in ROI images.

Tables

Appendix 1—key resources table
Reagent type (species) or resourceDesignationSource or referenceIdentifiersAdditional information
Strain, strain background (Escherichia coli)DH5α E. coliNEBCat# C2987Competent cells
Strain, strain background (E. coli)One Shot Stbl3 Chemically Competent E. coliInvitrogenCat# C737303Competent cells
Strain, strain background (SARS-CoV-2)SARS-CoV-2 Wuhan (SARS-CoV_FR-3)OtherGenBank OR018857Provided by G. Kochs, Freiburg, Germany
Strain, strain background (SARS-CoV-2)SARS-CoV-2 Wuhan (SARS-CoV_Muc)OtherGenBank OR018856Provided by G. Kochs, Freiburg, Germany
Strain, strain background (SARS-CoV-2)SARS-CoV-2 Omicron variant BA.1Human donorsGenBank OR018858Approved by Ethikkommission Nordwest- und Zentralschweiz #2022-00303
Strain, strain background (SARS-CoV-2)SARS-CoV-2 Omicron variant BA.5Human donorsGenBank OR018859Approved by Ethikkommission Nordwest- und Zentralschweiz #2022-00303
Strain, strain background (SARS-CoV-2)SARS-CoV-2 Omicron variant XBB.1.5Human donorsGenBank OX393614Approved by Ethikkommission Nordwest- und Zentralschweiz #2022-00303
Strain, strain background (CHIKV)Chikungunya virus (CHIKV)OtherProvided by K. Leuzinger, Basel, Switzerland
Strain, strain background (DENV1)Dengue virus serotype 1 (DENV1)OtherProvided by K. Leuzinger, Basel, Switzerland
Strain, strain background (DENV3)Dengue virus serotype 3 (DENV3)OtherProvided by K. Leuzinger, Basel, Switzerland
Cell line (Chlorocebus sabaeus)African green monkey kidney cells (Vero E6)OtherProvided by V. Thiel, Bern, Switzerland
Cell line (Homo sapiens)Adenocarcinomic human alveolar basal epithelial cells (A549)NIBSCCat# 101006
Cell line (Mesocricetus auratus)Baby hamster kidney cells (BHK-21)Thermo ScientificATCC CCL-10
Cell line (H. sapiens)Human embryonic kidney cells (HEK293T)OtherProvided by D. Pinschewer, Basel, Switzerland
Cell line (H. sapiens)Human embryonic kidney cells (HEK293)BataviaCat# 110-025; Lot# 19D006
Cell line (Cricetulus griseus)Chinese hamster ovary cells (CHO-K1)Thermo ScientificATCC CCL-61
Biological sample (H. sapiens)Serum samplesHuman donorsApproved by Ethikkommission Nordwest- und Zentralschweiz #2022-00303
AntibodyAnti-β-actin (mouse monoclonal)Cell Signaling TechnologyCat# 3700; RRID:AB_2242334; Lot# 20WB (1:1000)
AntibodyAnti-FLAG (rabbit polyclonal)Cell Signaling TechnologyCat# 14793; RRID:AB_2572291; Lot# 5WB (1:1000)
AntibodyAnti-FLAG (rat monoclonal)BioLegendCat# 637301; RRID:AB_1134266; Lot# B318853ICC (1:1000)
AntibodyAnti-SARS-CoV-2 NSP2 (rabbit polyclonal)GeneTexCat# GTX135717; RRID:AB_2909866; Lot# B318853WB (1:5000)
AntibodyAnti-SARS-CoV-2 Nucleocapsid protein (mouse monoclonal, 4F3C4)Sven Reiche (doi: 10.1016/j.virusres.2006.07.005)ICC (1:500)
AntibodyAnti-SARS-CoV-2 ORF3a (sheep polyclonal)MRC PPU reagents (doi:10.1371/journal.pbio.3001091)WB (1:1000)
ICC (1:500)
AntibodyAnti-SARS-CoV-2 ORF8 (rabbit polyclonal)Novus BiologicalsCat# NBP3-07972; Lot# 25966-2102WB (1:1000)
ICC (1:1000)
Recombinant DNA reagentpLEX307-TMPRSS2-blast (plasmid)AddgeneCat# 158458
Recombinant DNA reagentpUC19_CoV-2_LinkerThis studyAddgene plasmid #211731Plasmid expressing 5’ and 3’ regions for direct rescue
Recombinant DNA reagentpUC19_CoV-2_frA (plasmid)This studyPlasmid encoding SARS-CoV-2 ‘fragment A,’ see Supplementary file 7
Recombinant DNA reagentpUC19_CoV-2_frB (plasmid)This studyPlasmid encoding SARS-CoV-2 ‘fragment B,’ see Supplementary file 7
Recombinant DNA reagentpUC19_CoV-2_frC (plasmid)This studyPlasmid encoding SARS-CoV-2 ‘fragment C,’ see Supplementary file 7
Recombinant DNA reagentpUC19_CoV-2_frD (plasmid)This studyPlasmid encoding SARS-CoV-2 ‘fragment D’ Wuhan isolate, see Supplementary file 7
Recombinant DNA reagentpUC19_CoV-2_frD_S Omicron BA.1 (plasmid)This studyPlasmid encoding SARS-CoV-2 ‘fragment D’ Omicron BA.1 isolate, see Supplementary file 7
Recombinant DNA reagentpUC19_CoV-2_frD_S Omicron BA.5 (plasmid)This studyPlasmid encoding SARS-CoV-2 ‘fragment D’ Omicron BA.5 isolate, see Supplementary file 7
Recombinant DNA reagentpUC57_CoV-2_frA (plasmid)GenScriptHigh-quality DNA ordered from GenScript
Recombinant DNA reagentpUC57_CoV-2_frB (plasmid)GenScriptHigh-quality DNA ordered from GenScript
Recombinant DNA reagentpUC57_CoV-2_frC (plasmid)GenScriptHigh-quality DNA ordered from GenScript
Recombinant DNA reagentSARS-CoV-2 Omicron Strain S gene original_pcDNA3.1(+)GenScriptCat# MC_0101273
Sequence-based reagentOligonucleotides (primers)This studyFor all primers, see Supplementary file 5
Commercial assay or kitMaxwell RSC Viral Total Nucleic Acid Purification KitPromegaCat# AS1330
Commercial assay or kitMaxwell RSC miRNA from Plasma or SerumPromegaCat# AS1680
Commercial assay or kitSuperScript IV One-Step RT-PCR SystemInvitrogenCat# 12594100
Commercial assay or kitSF Cell Line 4D-Nucleofector-X KitLonzaCat# V4XC-2012
Commercial assay or kitEasySeq SARS-CoV-2 WGS Library Prep KitNimaGenSKU: RC-COV096
Chemical compound, drugBlasticidinSigma-AldrichCat# SBR00022
Chemical compound, drugHoechst 33342 dyeSigma-AldrichCat# B2261
Chemical compound, drugRNase AThermo ScientificCat# EN0531
Software, algorithmImageJ v2.9.0/1.53tNHIhttps://imagej.nih.gov/ij/download.html; RRID:SCR_003070
Software, algorithmPrism v9GraphPadhttps://www.graphpad.com/scientific-software/prism/; RRID:SCR_002798
Software, algorithmOmeroOpen Microscopy Environmenthttp://www.openmicroscopy.org/site/products/omero; RRID:SCR_002629
Software, algorithmAdobe Illustrator CCAdobehttp://www.adobe.com/products/illustrator.html; RRID:SCR_010279
Software, algorithm7500 Real-Time PCR Software Version 2.0.6Applied BiosystemsRRID:SCR_014596
Software, algorithmImmunoSpot Software Version 7.0.26.0ImmunoSpotRRID:SCR_011082
Software, algorithmNikon NIS-Elements AR Versions 5.30.07NikonRRID:SCR_014329
Software, algorithmLI-COR Image Studio Version 2.0LI-COR
OtherSARS-CoV-2 Rapid Antigen TestRocheCat# 9901-NCOV-01GSARS-CoV-2 Antigen Test to check for viral rescue, see Figure 1

Additional files

Supplementary file 1

Infectivity assessment of recombinant virus on Vero E6 cells.

Vero E6 cells were infected with the indicated recombinant virus and pictures were taken 3 d post-infection. Of note, the virus was not titrated and the development of CPE is not quantitative. Scale bar represents 100 μm.

https://cdn.elifesciences.org/articles/89035/elife-89035-supp1-v1.pdf
Supplementary file 2

Genomic characterization of recombinant SARS-CoV-2 virus based on NGS data.

Mutations with a relative abundance of >10% in the entire virus population are listed. Ambiguities or low coverage are highlighted in italics.

https://cdn.elifesciences.org/articles/89035/elife-89035-supp2-v1.xlsx
Supplementary file 3

Homology regions successfully used for recombinant SARS-CoV-2, CHIKV, and DENV1/DENV3.

Length, GC content, and the hypothetical annealing temperature (according to OligoCalc, salt-adjusted) are listed. Note that homology regions were chosen independently from GC content or annealing temperature and values are only listed for completion.

https://cdn.elifesciences.org/articles/89035/elife-89035-supp3-v1.xlsx
Supplementary file 4

Sanger sequencing data of the region of interest for the generated mutant rSARS-CoV-2, rCHIKV, and rDENV1/rDENV3.

https://cdn.elifesciences.org/articles/89035/elife-89035-supp4-v1.xlsx
Supplementary file 5

Oligonucleotide list.

https://cdn.elifesciences.org/articles/89035/elife-89035-supp5-v1.xlsx
Supplementary file 6

PCR settings for individual fragments.

https://cdn.elifesciences.org/articles/89035/elife-89035-supp6-v1.xlsx
Supplementary file 7

Plasmids.

https://cdn.elifesciences.org/articles/89035/elife-89035-supp7-v1.xlsx
MDAR checklist
https://cdn.elifesciences.org/articles/89035/elife-89035-mdarchecklist1-v1.pdf

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Enja Tatjana Kipfer
  2. David Hauser
  3. Martin J Lett
  4. Fabian Otte
  5. Lorena Urda
  6. Yuepeng Zhang
  7. Christopher MR Lang
  8. Mohamed Chami
  9. Christian Mittelholzer
  10. Thomas Klimkait
(2023)
Rapid cloning-free mutagenesis of new SARS-CoV-2 variants using a novel reverse genetics platform
eLife 12:RP89035.
https://doi.org/10.7554/eLife.89035.3