Virus adaptation to heparan sulfate comes with capsid stability tradeoff

  1. Han Kang Tee  Is a corresponding author
  2. Simon Crouzet
  3. Arunima Muliyil
  4. Gregory Mathez
  5. Valeria Cagno
  6. Matteo Dal Peraro
  7. Aleksandar Antanasijevic
  8. Sophie Clément
  9. Caroline Tapparel  Is a corresponding author
  1. Department of Microbiology and Molecular Medicine, University of Geneva, Switzerland
  2. Interschool Institute of Bioengineering (SV), School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Switzerland
  3. Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), Switzerland
11 figures, 1 table and 2 additional files

Figures

Figure 1 with 2 supplements
Lysosomotropic drugs inhibit infection by MP4 but not by MP4-97R/167G.

(A) Schematic illustration of the virus inhibitory assay workflow. Cells were pre-treated with lysosomotropic drugs and infected (MOI 0.1) in presence of the drug. After inoculum removal, infected cells were cultured in drug-free media and infected cells were stained by immunofluorescence (IF) with anti-VP2 Ab. Figure 1A was created with BioRender.com. (B) Hydroxychloquine (HCQ) and Bafilomycin A1 (BAF-A1) dose response assay in infected Vero cells. (C) HCQ effect in Vero cells pre-treated or not with heparinase III (hepIII) or sodium chlorate (NaClO3) as in A. Mean and S.E.M of biological triplicates are shown. Results are shown as % of virus-positive cells relative to nontreated control. In B, statistical significance (one-way ANOVA) between treated and untreated virus or between treated MP4 and MP4-97R/167G was calculated based on the area under curve (AUC). In C, statistical significance (two-way ANOVA) was calculated for each virus between each condition. *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001.

Figure 1—figure supplement 1
Toxicity and efficacy of lysosomotropic drugs.

(A) The cytotoxic effect of lysosomotropic drugs was evaluated with LDH and MTT assays. Vero cells were treated with a range of different concentrations of hydroxychloroquine (HCQ) or bafilomycin A1 (BAF-A1) for 2 hr. At 24 hr post-treatment, cell supernatants and lysates were collected for LDH assay and MTT assay, respectively, to determine cytotoxicity effect (n=2 biological replicates). (B) Inhibition of endosomal acidification confirmed with LysoTracker staining (red). Lysosomes are stained in green with an anti-LAMP-1 Ab and nuclei in blue (DAPI). Representative immunofluorescence (IF) images are shown (scale bar, 10 µm). (C) Quantified LysoTracker signal intensities calculated from nontreated, HCQ and BAF-A1-treated conditions are shown relative to lysosomal-associated membrane protein 1 (LAMP-1) intensity (n=3 biological replicates). Statistical significance was calculated with one-way ANOVA. (D) Representative IF staining of EV-A71 (anti-VP2 in green) 24 hpi of Vero cells in presence of 25 µM HCQ or 250 nM BAF-A1 (scale bar, 300 µm). (E) Viral RNA load quantification by real-time RT-qPCR of infected Vero cells with and without drug treatment at 24 hpi. (F) Cytotoxic effect of HCQ and BAF-A1 were evaluated using LDH and MTT assays in Rhabdomyosarcoma (RD) cells. (G) Dose response assay with HCQ and BAF-A1 on RD cells were performed exactly like in Vero cells (Figure 1). Infected cells (stained with anti-VP2 Ab) were quantitated at 24 hpi after treatment with increasing drug concentrations. Results are shown as % of virus-positive cells relative to nontreated control. Area under curve (AUC) was calculated and statistical significance (one-way ANOVA) between treated and untreated virus or between treated MP4 and MP4-97R/167 G are shown. Mean and S.E.M of biological duplicates are shown. **p<0.01, ***p<0.001, ****p<0.0001.

Figure 1—figure supplement 2
Virus infection in SCARB2-KO cells.

Virus infection was performed on Rhabdomyosarcoma (RD) WT and RD ΔSCARB2 cells. Cells were lysed, and viral RNA copy numbers were quantitated at 24 hpi using RT-qPCR. Results are expressed as % Virus RNA copy number relative to RD WT cells (set to 100%). Mean and S.E.M of biological triplicates are shown.

****p<0.0001.

Hydroxychloroquine (HCQ) targets viral entry.

(A) Virus binding assay in Vero cells in presence of 25 µM HCQ. (B) Single-cycle replication kinetic in nontreated and HCQ-treated Vero cells. At each timepoint, cell lysates were collected, and viral RNA copy numbers were quantitated using RT-qPCR (C) Time-of-addition assay in Vero cells treated with HCQ starting at different timepoints. Infected cells (MOI 0.1) were quantitated 24 hpi by immunofluorescence (IF). (D) Schematic diagram of Vero cells pre-treated with HCQ and subsequently subjected to transfection of in vitro RNA transcript or infection with EV-A71 nanoluciferase (Nluc) reporter viruses. At the indicated timepoints, cell supernatants were collected, and luciferase activity was measured. Figure 2D was created with BioRender.com.(E & F) Results are expressed in % relative light unit (RLU) of treated versus nontreated virus at indicated timepoints. The mean and S.E.M from biological triplicates are shown. Statistical significance was calculated using two-way ANOVA, comparing treated and untreated control. *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001.

Hydroxychloroquine (HCQ) delays the uncoating of MP4.

(A) Schematic illustration of the neutral red assay workflow. Vero cells were pre-treated with or without HCQ for 1 hr. Neutral red-labeled viruses were allowed for cell infection at 37 °C for 1 hr (MOI 0.1). The inoculum was then removed and replaced with fresh media. Infected cells were exposed to light for 30 min at different timepoints and further incubated up to 24 hpi for immunofluorescence (IF) staining. Figure 3A was created with BioRender.com. (B) Effect of light inactivation on replication of neutral red-labeled MP4 (left panel) or MP4-97R/167G (right panel). Results are plotted as % of virus-positive cells relative to non-treated dark control. Mean and S.E.M of biological triplicates are shown. Statistical significances (two-way ANOVA) were calculated between treated and nontreated conditions. (C) Schematic illustration of virus uncoating monitored with the combinational use of RNA-FISH to detect EV-A71 RNA (red) and IF with anti-VP2 Ab to detect the viral capsid (green). Co-staining highlights intact viruses in yellow while empty capsids and free RNA are in green and red, respectively. (C, right panel) Representative images (scale, 20 µm) of MP4 and MP4-97R/167G binding after 1 hr at 4 °C with virus genomic RNA (vRNA) (red) and capsids (green). Figure 3C was created with BioRender.com. (D) Co-localization of capsid and vRNA in individual cells at 2 hpi and 4 hpi analyzed using Mander’s overlap coefficient (n=32 individual cells from two independent experiments). Statistical comparison (unpaired t-test) of untreated and treated groups. *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001. (E) Representative images of the 4 hr time point. Arrows: empty capsid.

Figure 4 with 1 supplement
MP4-97R/167G uncoats from early endosomes.

(A) Nontreated and hydroxychloroquine (HCQ)-treated Vero cells were stained with anti-EEA-1 antibody (green) to label early endosomes, and DAPI (blue) to label cell nuclei. (B) Schematic representation of endosomal route upon overexpression of Rab5a WT or constitutively active (CA) mutant. In C and D, Vero cells transiently expressing Rab5a-eGFP WT or CA were fluorescence-activated flow cytometry (FACS)-sorted and infected with the two viral variants. Infections were compared at different time post-infection. Figure 4B was created with BioRender.com. In (C) viral capsids (anti-VP2 Ab, in red) localize in early endosomes at 2 hpi in cells expressing Rab5a WT or CA. In (D) the proportion of cells containing replicating viruses (stained with the anti-dsRNA J2 Ab, see Fig.S3B for representative images) is calculated at 7 hpi. Results and statistical significance (two-way ANOVA) are expressed relative to cells with Rab5a WT. Mean and S.E.M from triplicates are shown. ***p<0.001. In B and C, white boxes are enlarged in the right panel. Scale bar: 20 µm.

Figure 4—figure supplement 1
Localization of viral capsids and double-stranded RNA at respectively 0.5 hpi and 7 hpi in fluorescence-activated flow cytometry (FACS)-sorted GFP-positive Vero cells transiently expressing Rab5a-eGFP WT or constitutively active (CA) and infected with the two viral variants.

(A) Vero cells infected with MP4 and MP4-97R/167G were fixed at 0.5 hpi. Viral capsid localization was imaged with an anti-VP2 Ab (in red). Magnified areas are highlighted in white box and displayed at left bottom of merged image. Scale bar: 20 µm. (B) Vero cells infected with MP4 and MP4-97R/167G were fixed at 7 hpi and stained with anti-dsRNA J2 Ab to highlight viral replication. The images shown are representative examples of those used for the quantifications presented in Figure 4D. Scale bar: 400 µm.

Figure 5 with 4 supplements
MP4 displayed stronger capsid stability and reduced sensitivity to acidification and high temperatures.

(A) Negative staining electron microscopy (nsEM) analysis of MP4 and MP4-97R/167G incubated at pH 7 and pH 5. Representative raw micrographs are shown in each case. (B) Representative 2D class averages generated from datasets shown in panel A (box size = 54 nm; left) and the overlay of the corresponding 3D maps (right). Gray and orange shade indicates virus particle reconstructions at pH 7 and pH 5, respectively. (C) Temperature sensitivity assay. Infected Vero cells (MOI 0.5) were quantitated by immunostaining with an anti-VP2 Ab at 24 hpi after 1 hr incubation at increasing temperatures. Results are shown as % of virus-positive cells relative to 4 °C treated control. Error bars indicate mean and S.E.M from biological triplicates. (D) For sSCARB2 inhibition assay, viruses (MOI 0.5) were incubated 1 hr at 37 °C with 1 µg of soluble scavenger receptor class B member 2 (SCARB2) (sSCARB2) before infection of Vero cells. Infected Vero cells were quantitated by immunostaining with an anti-VP2 Ab at 24 hpi. Results are shown as % of virus-positive cells relative to nontreated controls. Statistically significance was calculated with two-way ANOVA. ***p<0.001, ****p<0.0001.

Figure 5—figure supplement 1
Visual presentation of prediction of changes induced by the L97R/E167G mutations on electrostatic surface potential.

Electrostatic surface potential calculated by Adaptive Poisson–Boltzmann Solver (APBS) from −5 kT/e (red) to +5 kT/e (blue).

Figure 5—figure supplement 2
Prediction of changes in amino acid interactions and capsid stability performed using crystal structure of full assembled capsid on DynaMut server.

Interatomic interactions displayed and compared between WT and mutant capsid structures. VP1-97 and VP1-167 residues are labeled in light green and represented as sticks together with the surrounding interaction residues. Changes in interactions are highlighted on both WT and mutant structures with red asterisks (*).

Figure 5—figure supplement 3
VP1-L97R and VP1-E167G mutations decrease capsid stability.

Computation of the vibrational entropy change (ΔΔSVib) between WT and mutants. Amino acids in red indicate an increase in molecule flexibility.

Figure 5—figure supplement 4
Raw micrographs and 2D classes of viruses incubated with soluble scavenger receptor class B member 2 (SCARB2).

Raw micrographs and 2D classes of MP4 and MP4-97R/167G incubated at neutral pH with and without sSCARB2. Representative 2D classes are shown in each case to recapitulate overall diversity in electron microscopy (EM) images (box size = 61 nm). Red squares indicate 2D class averages with strong staining in the middle of the particle indicating partially open capsids with low density of protein and genome components, which is consistent with empty capsids.

Figure 6 with 1 supplement
Heparan-sulfate-binding VP1-145Q variant exhibits resistance to hydroxychloroquine (HCQ) and higher sensitivity to soluble scavenger receptor class B member 2 (sSCARB2) inhibition and thermal stress.

(A) Virus inhibitory assay with VP1-145 variants were performed with 25 µg HCQ on Vero cells (MOI 0.1). (B) For temperature sensitivity assays, VP1-145 variants were incubated at increasing temperature for 1 hr before inoculated onto Vero cells (MOI 0.5). (C) For sSCARB2 inhibition assay, VP1-145 variants were incubated 1 hr at 37 °C with 1 µg of soluble SCARB2 (sSCARB2) before infection of Vero cells (MOI 0.5). Infected cells were quantitated by immunostaining with anti-VP2 Ab at 24 hpi. Results are shown as % of virus-positive cells relative to nontreated control (A & C) or 4 °C treated control (B). Mean and S.E.M of biological triplicates are shown. Statistically significant differences (two-way ANOVA) are shown. **p<0.01, ***p<0.001, ****p<0.0001.

Figure 6—figure supplement 1
Human clinical strain variants exhibited similar hydroxychloroquine (HCQ) effects compared to MP4 variants.

Virus inhibitory assay with human clinical strain variant carries VP1-97L residue (EV71-97L) and VP1-97R/167G mutations (EV71-97R/167G) were treated with 25 µM HCQ on Vero cells (MOI 0.1). Infected cells were quantitated by immunostaining with anti-VP2 Ab at 24 hpi. Results are shown as % of virus-positive cells relative to nontreated control. Mean and S.E.M of biological triplicates are shown. Statistically significant differences (two-way ANOVA) are shown. *p<0.05, ****p<0.0001.

Seesaw model depicting the interplay between capsid mutations, heparan sulfate-binding, capsid stability as well as the resulting fitness changes in both in vitro and in vivo settings.

Viruses undergo continuous mutations to optimize fitness across diverse environments. In cell culture, they adapt to attain an ‘in vitro advantage’ by decreasing capsid stability while acquiring HS-binding capacity, consequently enhancing their infectivity. Conversely, during human infection, viruses adapt to secure an ‘in vivo advantage’ by bolstering capsid stability, relinquishing heparan sulfate (HS)-binding capacity, and thereby evading viral trapping and resisting environmental stresses. Figure 7 was created with BioRender.com.

Author response image 1
Molecular dynamic simulations of a subpart (corresponding to a capsid pentamer) of the wild type VP1 capsid (a) and the L97R-E167G mutated version (b), each at pH 5 and 7 and at 37°C (310K) and 50°C (323K).

The extraction of the subpart and the lack of physical constraints around the external-facing regions are inducing artificial moves during the simulation, and preventing its convergence, as witnessed by the high variability of root mean square deviations (RMSDs).

Author response image 2
Binding of (a) EV71-VP1-97R167G and (b) EV71-VP-97L167E in RD and Caco-2 cells with and without SCARB2 (Cas9 CRISPR KO using two different guide RNAs, g1 and g2) and/or HS expression (heparinase treatment).

Viral loads were measured by RT-qPCR in whole cell extracts and are expressed as the mean ± SEM.

Author response image 3
PaSTRy assay was performed by incubating virus in citrate phosphate buffer adjusted to pH7 and pH5.

Viral RNA release from capsid was detected using SYBR green II dye when the virus capsid was heating gradually with temperature increase of 1°C from 25°C to 95 °C.

Author response image 4
Raw micrographs and 2D classes of MP4 incubated with soluble SCARB2 for 30 minutes at neutral or acidic pH.

Red squares indicate 2D class averages with strong staining in the middle of the particle indicating partially open capsids with low density of protein and genome components, which is consistent with empty capsids.

Tables

Key resources table
Reagent type (species) or resourceDesignationSource or referenceIdentifiersAdditional information
AntibodyAnti-enterovirus 71 Ab
(mouse monoclonal)
Merck MilliporeMAB979;
RRID:AB_95300
IF (1:1000)
RNAscope (1:100)
AntibodyAnti-dsRNA monoclonal antibody J2
(mouse monoclonal)
SciconsRRID:AB_2651015IF (1:500)
antibodyAnti-LAMP1 antibody
(rabbit monoclonal)
Cell SignalingD2D11, RRID:AB_2687579IF (1:100)
AntibodyAnti-EEA1 antibody
(goat polyclonal)
Santa Cruzsc-6414, RRID:AB_640035IF (1:100)
AntibodyGoat anti-Mouse IgG (H+L) Highly Cross-Adsorbed Secondary Antibody, Alexa Fluor 488 (goat polyclonal)Thermo Fisher ScientificA-11029; RRID:A-11029IF (1:2000)
RNAscope (1:200)
AntibodyGoat anti-Mouse IgG (H+L) Highly Cross-Adsorbed Secondary Antibody, Alexa Fluor 594 (goat polyclonal)Thermo Fisher ScientificA-11032; RRID:AB_2534091IF (1:2000)
Strain, strain background (virus)MP4Cagno et al., 2019GenBank: JN544419Mouse-adapted virus
Strain, strain background (virus)HU-97LTee et al., 2021GenBank: EU414331Clinical strain
Strain, strain background (virus)IEQKobayashi and Koike, 2020GenBank: AF316321
Strain, strain background (virus)IEEKobayashi and Koike, 2020GenBank: AF316321
Chemical compound, drugHydroxychloroquineTocris747-36-4
Chemical compound, drugBafilomycin A1InvivoGen88899-55-2
Chemical compound, drugSodium chlorateSigma7775-09-9
Chemical compound, drugNeutral redSigma Aldrich553-24-2
Chemical compound, drugHeparinase IIIAmsbio37290-86-1
Chemical compound, drugPuromycinInvivoGen58-58-2
Chemical compound, drugLysoTracker Deep RedThermo Fisher ScientificLysoTracker Deep Red
Chemical compound, drugSYBR green II RNA gel stainThermo Fisher ScientificSYBR green II RNA gel stain
Recombinant proteinRecombinant Human LIMPII/SR-B2 Fc Chimera Protein, CFBio-Techne1966-LM
Commercial kit or assayRNAscope V-EV71-C1 probeBiotechne1087481-C1
Commercial kit or assayRNase P housekeeping geneThermo Fisher Scientific4316861
Commercial kit or assayLipofectamine 2000Thermo Fisher Scientific11668019
Commercial kit or assaySuperscript II reverse transcriptaseThermo Fisher Scientific18064022
Commercial kit or assayRandom hexamer primersRoche11034731001
Commercial kit or assayPlatinum Taq DNA Polymerase, DNA-freeThermo Fisher Scientific15966025
Commercial kit or assayTSA Vivid 570 kitTocris7526
Commercial kit or assayE.Z.N.A. Viral RNA kitOmega Bio-TekR6874-02
Commercial kit or assayRNAscope Multiplex Fluorescent V2Biotechne323270
Commercial kit or assayRNA-Protein Co-detection Ancillary KitBiotechne323180
Commercial kit or assayKAPA SYBR FAST One-Step qRT-PCR KitsKapa BiosystemsKK4650
Commercial kit or assayNano-Glo Luciferase Assay SystemPromegaN1110
Commercial kit or assayThiazolyl Blue Tetrazolium Bromide (MTT)MerckM5655
Commercial kit or assayCyQUANT LDH Cytotoxicity AssayThermo Fisher ScientificC20300
Cell line (Cercopithecus aethiops)Vero cellsATCC, USARRID:CVCL_0059
Cell line (Homo sapiens)Rhabdomyosarcoma (RD) cellsATCC, USARRID:CVCL_1649
Cell line (Homo sapiens)RD-SCARB2-KOCaroline Tapparel Yamayoshi et al., 2013
Cell line (Homo sapiens)RD-ΔEXT1+hSCARB2Satoshi Koike Kuronita et al., 2002
Sequence-based reagentRT-qPCR assay primer/for (Entero/Ge/08 assay)Nishimura et al., 2024PCR primers5’-GCTGCGYTGGCGGCC-3’
Sequence-based reagentRT-qPCR assay primer/Rev (Entero/Ge/08 assay)Nishimura et al., 2024PCR primers5’-GAAACACGGACACCCAAAGTAGT-3’
Sequence-based reagentRT-qPCR assay primer/probe (Entero/Ge/08 assay)Nishimura et al., 2024PCR primers5’-CTCCGGCCCCTGAATGYGGCTAA-3’
Recombinant DNA reagentEV-A71/MP4 (Genbank accession number: JN544419; subgenogroup C2)Jen-Reng Wang Guo et al., 2022
Recombinant DNA reagentIEQ (Genbank accession number: JN544419: AF316321; subgenogroup B4)Jen-Reng Wang Kobayashi and Koike, 2020
Recombinant DNA reagentIEE (Genbank accession number: JN544419: AF316321; subgenogroup B4)Jen-Reng Wang Kobayashi and Koike, 2020
Recombinant DNA reagenteGFP-Rab5a WTPierre-Yves Lozach Dang et al., 2014
Recombinant DNA reagenteGFP-Rab5a S34NPierre-Yves Lozach Dang et al., 2014
Recombinant DNA reagenteGFP-Rab5a Q79LPierre-Yves Lozach Dang et al., 2014
SoftwareGeneious 10.2.16https://www.geneious.comhttps://www.geneious.com
SoftwareImageXpress Micro XLMolecular DevicesMolecular Devices
SoftwareGraphPad Prism 9https://www.graphpad.com/scientific-software/prism/https://www.graphpad.com/scientific-software/prism/
SoftwareUCSF Chimera (version 1.13.1)https://www.cgl.ucsf.edu/chimera/https://www.cgl.ucsf.edu/chimera/

Additional files

Supplementary file 1

Predicted Gibbs free energy change value (ΔΔG) was computed using I-mutant 2 server with calculation formula and an indication of protein structure stabilization as shown: Predicted Gibbs free energy change value (ΔΔG): ΔG (mutated protein) - ΔG (WT) in kcal/mol.

ΔΔG<0: Destabilizing mutation. ΔΔG>0: Stabilizing mutation.

https://cdn.elifesciences.org/articles/98441/elife-98441-supp1-v3.docx
MDAR checklist
https://cdn.elifesciences.org/articles/98441/elife-98441-mdarchecklist1-v3.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. Han Kang Tee
  2. Simon Crouzet
  3. Arunima Muliyil
  4. Gregory Mathez
  5. Valeria Cagno
  6. Matteo Dal Peraro
  7. Aleksandar Antanasijevic
  8. Sophie Clément
  9. Caroline Tapparel
(2024)
Virus adaptation to heparan sulfate comes with capsid stability tradeoff
eLife 13:e98441.
https://doi.org/10.7554/eLife.98441