Virus strains
TBFV were represented by TBEV strain Hypr (of the European subtype), LIV (strain LI/31) and LGTV (strain TP-21), which were all provided by the Collection of Arboviruses, Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice. MBFV used in this study included ZIKV (Brazilian strain Paraiba_01), kindly provided by Prof. Paolo M. de A. Zanotto, University of São Paulo, Brazil; USUV (Eu1 lineage strain 200/TM/10), provided by the Collection of Arboviruses, Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice55; and WNV (strain 13–104, representative of genomic lineage 2, isolated in the Czech Republic), kindly provided by Prof. Zdenek Hubalek and Dr. Ivo Rudolf, Institute of Vertebrate Biology of the Czech Academy of Sciences56.
Mammalian and tick cell lines
Porcine kidney stable PS cells (National Reference Laboratory for Cell Cultures, National Institute of Public Health, Prague) were grown in Leibovitz’ L-15 medium (L-15). Golden hamster BHK-21 (ATTC CCL-10) and African green monkey Vero cells (ATCC CCL-81) were grown in Dulbecco’s modified Eagle’s medium (DMEM). Furin-deficient human LoVo cells (human colon carcinoma, ATCC CCL-229) were grown in Ham’s F12 medium. Cells were cultured in media supplemented with foetal bovine serum (3% in L15, 10% in DMEM, or 20% in Ham's F12), 100 U ml− 1 penicillin, 100 µg ml− 1 streptomycin, and 2 mM glutamine (Sigma-Aldrich, Prague, Czech Republic), at 37°C in 5% CO2 (except for PS cells, which were cultured without extra CO2). The tick cell line IRE/CTVM1957 (derived from Ixodes ricinus, Tick Cell Biobank, University of Liverpool, UK) was maintained at 28°C without extra CO2, in L15 medium supplemented with 10% foetal bovine serum and 5% tryptose phosphate broth, as previously described58.
Production of prM-containing immature particles
Immature prM-containing virus particles were produced using LoVo cells, which are deficient in furin protease59, and thus provide an easy-to-use platform for immature virus production without affecting the pH within the maturation pathway. For propagation, cells were seeded in 96-well plates (approximately 3 × 104 cells per well), and incubated at 37°C under 5% CO2 for 24 hours, to form a confluent monolayer. Then these cells were infected with viruses at a multiplicity of infection (MOI) of 2. After 12 h, the virus suspension was removed, the cells were washed three times with phosphate-buffered saline (PBS), and fresh medium was added. At 48 h post-infection (hpi), the virus was harvested, and viral titre was determined by plaque assays, or viral RNA copies were quantified by quantitative PCR with reverse transcription (RT-qPCR), as described below.
Plaque assay
Plaque assays were performed as previously described60, with slight modifications. Briefly, viruses were diluted 10-fold in 24-well tissue culture plates, and 1–1.5 × 105 PS or Vero cells were added to each well. After 4 h of incubation, the wells were covered with 1.5% (w/v) carboxymethylcellulose (CMC) in L-15 (PS cells for TBEV, LGTV, LIV, and USUV) or DMEM (Vero cells for ZIKV and WNV) medium. After five days of incubation at 37°C, infected plates were washed with PBS, and cell monolayers were stained with naphthol blue-black. Viral titre was expressed as plaque-forming units (PFU) per ml.
RNA isolation, RT-qPCR, RT-PCR, and sequencing
Viral RNA was isolated from culture supernatants using the QIAmpViral RNA mini kit (Qiagen, Germantown, MD, USA) following the manufacturer’s instructions. RT-qPCR was performed on a LightCycler 480 II in a 96-well plate block (Roche, Basel Switzerland), using the Advanced Kits for Tick-borne Encephalitis, Zika Virus, or West Nile Virus (Genesig, Germantown, MD, USA) or the Usutu Virus Kit (#FR439; Genekam Biotechnology AG, Duisburg, Germany). Lyophilized OneStep qRT-PCR master mix (Oasig) was used, following the manufacturer’s instructions. For TBEV, ZIKV, and WNV, the copy numbers per µl were calculated from calibration curves based on standards provided by the manufacturer. For USUV, the calibration curve was developed using serial dilutions of RNA isolated from a virus stock with a known titre in PFU ml− 1; therefore, the qRT-PCR results for USUV were presented in relative PFU per ml (rPFU ml− 1). For sequencing, the isolated RNA was reverse-transcribed using a QIAGEN OneStep Ahead RT-PCR Kit (Qiagen, Germantown, MD, USA). Amplified products were separated in 1% agarose gel, and purified using the Wizard SV Gel and PCR Clean-Up System (Promega, USA). Sanger sequencing was performed by the commercial provider Eurofins Genomics.
Plaque-reduction assay
PS cells were seeded in 6-well tissue culture plates (0.8–1 × 106 cells per well), and incubated for 24 h. Confluent cells were pretreated for 2 h with the furin inhibitor Decanoyl-RVKR-CMK (Sigma Aldrich), and then infection was performed using 50 PFU per well, at 37°C for 1 h. Next, the unbound virus was removed, the cells were washed three times with PBS, and fresh medium with a CMC overlay was added. After incubation at 37°C for five days, infected plates were washed with PBS, and cell monolayers were stained with naphthol blue-black as described for the plaque assay.
Virus in vitro furin cleavage assay
The prM-TBEV sample was concentrated using Amicon ultra-centrifugal 100-kDa filters, and medium-buffer exchange was performed by centrifugation (4,500 rpm, 4°C, 7 min; TX-400 rotor, 75003629; Thermofisher Scientific). Next, the medium was changed to Tris-maleate buffer20 (TMB, 50 mM) containing CaCl2 (10 mM), with the pH adjusted to 5.5 or 7.5. For furin in vitro cleavage, 1 µl of recombinant human furin (r-furin, #SRP6274; Sigma Aldrich) was added to the 100-µl reaction, followed by incubation at 37°C for 2 h. Next, LoVo cells in TMB were incubated with the virus (MOI 0.1) for 4 h. Then the unbound virus was removed, the cells were washed three times with PBS, and fresh medium was added. Finally, at 48 hpi, the viral progeny was harvested for further analysis. Infection with prM-USUV was performed using PS cells, with the same in vitro cleavage procedure as described above.
Fluorescent infectious assay
Recombinant mCherry-expressing TBEV was used to visualize viral infection61. PS cells were cultured in 96-well plates, and infected as described above. At 48 hpi, nuclei were stained with Hoechst 33342 (Invitrogen), following the manufacturer’s recommended protocol. Immunofluorescence of prM-TBEV and prM-USUV infection was visualised after fixation in ice-cold methanol-acetone mixture (1:1) with primary anti-flavivirus protein E antibodies (1:250; Sigma Aldrich) and then with secondary antibodies AlexaFluor 488 (1:1000; Thermofisher Scientific) and DAPI (1:2000; Sigma Aldrich) to stain cell nuclei. Images were acquired using an Olympus IX81 epifluorescence microscope, equipped with Olympus 10× UPLFLN lenses, and a Hamamatsu OrcaR2 camera controlled by Olympus Xcellence software. Raw images were processed using ImageJ/Fiji software.
Western blot analysis
To investigate prM cleavage by furin, we performed western blot analysis. Virus samples used for western blot analysis were produced in medium mixture DMEM:HAM F12 (1:1) supplemented with a reduced FBS concentration (2%), and concentrated using Amicon ultra-centrifugal 100-kDa filters. Laemmli sample buffer was mixed 1:3 with the virus sample, and incubated for 5 min at 95°C. After cooling on ice, samples were ready for downstream analysis. Proteins were separated by standard SDS-PAGE, and then transferred to an Immobilon®-P PVDF Membrane (Millipore). Blots were blocked overnight at 4°C in blocking buffer comprising PBS, 0.05% Tween 20, and 2% Amersham ECL Prime Blocking Reagent (Cytiva). Next, the blots were stained with primary antibodies against TBEV E protein (mouse monoclonal antibody 149362 diluted 1:2000) or M protein (in-house rabbit polyclonal serum63 diluted 1:500), overnight at 4°C, followed by the secondary antibodies anti-mouse IgG-HRP (Invitrogen AB_228307) or anti-rabbit IgG-HRP (Invitrogen AB_228341) for 1 h at room temperature. Finally, blots were visualized on an Amersham™ Imager 680 (GE Healthcare).
Mouse infection
We evaluated the infectivity of prM-containing viruses in 6-week-old female BALB/c mice (ENVIGO RMS). Six groups of mice were inoculated s.c. with TBEV or prM-TBEV strain Hypr as follows: group 1 (n = 5), TBEV 103 genomic equivalents (gen. eq.) per mouse; group 2 (n = 10), TBEV 102 gen. eq. per mouse; and group 3 (n = 5), TBEV 101 gen. eq. per mouse, respectively. Groups 4–6 were infected with the equivalent doses of the prM-TBEV variant. For comparison with MBFV, WNV strain 13–104 was used and the experimental set-up and mouse groups were the same as in the TBEV experiment. Over a 28-day experimental period, the survival rates and symptoms were monitored daily. Symptoms were evaluated using the following clinical scores 1 = healthy, 2 = piloerection, 3 = hunched back, 4 = paralysis, and 5 = death. Changes in body weight upon infection were monitored for 14 days. All mice exhibiting a clinical score of 4 were humanely terminated (by cervical dislocation) immediately upon detection of symptoms.
Blood was taken 3 dpi from the tail vein to evaluate the virus titre in the serum. After dissection of the brain on day 8, tissue samples were homogenized in DMEM medium (+ 10% FBS) to obtain a 20% suspension. After centrifugation (5000 rpm, 4°C, 10 min; TX-400 rotor, 75003629; Thermofisher Scientific), the supernatant from suspension was used for the plaque assay.
Protease fluorescent assay
Four internally quenched fluorescent substrates—Dabcyl-EGSRSRRSVL-Edans, Dabcyl-EGSRTRRSVL-Edans, Dabcyl-HSRRSRRSLT-Edans, and Dabcyl-HSKRSRRSIA-Edans—were synthesized without C-terminal Edans, using a standard solid phase peptide synthesis protocol64, with 2-chlorotrityl chloride resin support. The N-Dabcylated side chain protected peptides were released from the resin using a mixture of acetic acid/2,2,2-trifluoroethanol/dichloromethane (1:1:3) for 2 h, and then were evaporated and dried. Next, the Edans was introduced via Edans acid sodium salt (1.5 eq. to peptide) and PyBOP (2.0 eq. to peptide) in the presence of DIPEA (3 eq. to peptide) in DMF as a solvent, overnight. After solvent evaporation, side chains were deprotected using a mixture of TFA/triisopropylsilane/water (95:2.5:2.5) for 1 h, and then liquids were evaporated. The residues were purified by preparative RP HPLC, and characterised by LC/MS-ESI: Dabcyl-EGSRSRRSVL-Edans [M + H]+ 1645.8, Dabcyl-EGSRTRRSVL-Edans [M + H]+ 1659.8, Dabcyl-HSRRSRRSLT-Edans [M + H]+ 1754.8, and Dabcyl-HSKRSRRSIA-Edans [M + H]+ 1696.9.
Lyophilized human recombinant furin protease produced in Hi-5 insect cells (PeproTech) was diluted to 1 mg ml− 1 using water, according to the manufacturer’s instructions. The assay buffer of an appropriate pH contained 25 mM acetic acid, 25 mM 2-[morpholino]ethanesulfonic acid (MES), 25 mM glycine, and 1 mM CaCl265. Fluorescence measurements were performed in 96-well Greiner chimney black plates. All assays were started by adding furin to a final concentration of 10 ng mL− 1 in the 100-µl reaction volume. The time-dependent increase of fluorescence upon enzyme addition was monitored for 30 min, with a kinetic cycle of 1 min, using a Tecan Spark plate reader with an excitation wavelength of 336 nm and emission wavelength of 490 nm. All measurements were made in triplicate.
Quaternary structure analysis
For both TBEV and USUV, models of the prM-E complexes (containing the complete protein sequences of the prM and E proteins) were made using AlphaFold266. These were used to generate the quaternary structure model of a dimer of heterodimers, based on the PDB code templates 7Z51, 5O6A, 7LCH, and 7QRE18, 38, 63, 67. Structural analysis of the models was performed in PyMOL 2.5.4. The quaternary models were used to predict mutations that would potentially affect the infectivity of viruses, which were modelled with AlphaFold2 to check the effects on the quaternary structure of TBEV and USUV, particularly the proximity of the furin cleavage site.
Site-directed mutagenesis and transfection
The reverse genetics system used was based on the generation of three infectious subgenomic overlapping DNA fragments encompassing the whole TBEV genome (strain Hypr), as previously described68. Fragments I and III were flanked by the human cytomegalovirus promoter (pCMV) and hepatitis delta ribozyme, followed by the simian virus 40 polyadenylation signal sequence (HDR/SV40pA). All three fragments were synthesized and individually cloned into pUC57 or pC11 vectors (GenScript, Piscataway, NJ, USA).
To produce genomic fragments with mutations in prM and E proteins, fragment I was used as a template for amplification with modified primers (Supplementary Fig. 8, Supplementary Table 1). This enabled production of two sub-fragments, Ia and Ib, each with overlapping homology arms. PCR and fragment purification were performed as previously described61.
For transfection, BHK-21 cells were seeded in a 24-well plate at a density of 3 × 105 cells (400 µl) per well, and incubated overnight. Cell transfection was performed using an equimolar mixture of the four DNA fragments. A DNA-lipid complex was prepared using X-tremeGENE Transfection Reagent and 200 µl Opti-MEM (Thermofisher Scientific), followed by a 15-min incubation at room temperature. The entire mixture was added drop-wise to the well with the cell monolayer, and then incubated for 4 days. Then the supernatant was collected, and viral titre was determined by plaque assay. Finally, mutagenesis was confirmed by sequencing.
Statistics, reproducibility, and graphics
GraphPad software Prism v.8 was used for statistical analysis. In all figures, data were plotted as means with error bars denoting the SD. All statistical tests were performed using the Mann-Whitney nonparametric test. In animal experiments, the survival rates were statistically evaluated using the log-rank Mantel-Cox test. The significance threshold was set at P < 0.05. Figures 1a, 2h, 5h, and Supplementary Fig. 8 were created with BioRender.com.