T cell deficiency precipitates antibody evasion and emergence of neurovirulent polyomavirus

  1. Matthew D Lauver
  2. Ge Jin
  3. Katelyn N Ayers
  4. Sarah N Carey
  5. Charles S Specht
  6. Catherine S Abendroth
  7. Aron E Lukacher  Is a corresponding author
  1. Department of Microbiology and Immunology, Pennsylvania State University, United States
  2. Department of Pathology and Laboratory Medicine, Penn State Milton S. Hershey Medical Center, United States
7 figures, 1 table and 4 additional files

Figures

Figure 1 with 3 supplements
Increased MuPyV replication in the kidney following T cell depletion in VP1 mAb-treated μMT mice.

(A) Experimental approach for VP1 mAb treatment and T cell depletion. (B) Viral DNA levels in the kidney 10 days post T cell depletion with combined αCD4 and αCD8β. Viral DNA was quantified by qPCR and compared to a standard curve (n=16–18). (C) Viral DNA levels in the kidney 10 days post depletion with αCD4 or αCD8β. Viral DNA was quantified by qPCR and compared to a standard curve (n=13–21). (D) (Left) Foci of virus replication the kidney cortex 10 days post T cell depletion. Kidneys were stained for CD13 (white), THP (green) and VP1 (red). (Right) Quantification of virus foci in the kidney. Data are the average of two kidney sections per mouse (n=12–13). Error bars are mean ± SD. Data are from at least two independent experiments. Data were analyzed by Mann-Whitney U test (B, D) or Kruskal-Wallis test with Dunn’s multiple comparisons test (C). *p<0.05, **p<0.01, ***p<0.001.

Figure 1—figure supplement 1
Passive immunization with a VP1 mAb in B cell-deficient mice neutralizes WT virus but not a VP1 mutant virus.

(A) VP1-specifc IgG 30 dpi from WT or μMT mice detected by ELISA with VP1 pentamers (n=10–12). (B) Viremia at 30 dpi in WT, μMT, or μMT mice treated with VP1 mAb starting 4 dpi. LOD: Limit of detection (n=8). (C–D) Neutralization of A2 (C) or A2.Δ295 (D) by sera from WT or VP1-mAb treated μMT mice 20 dpi. LT mRNA fold change is relative to infection by each virus in the absence of serum (n=8–9). (E) Neutralizing titers against A2 and A2.Δ295 from WT or VP1-mAb treated μMT mice 20 dpi. Values are the inverse log10 of the 50% neutralizing dilution of serum. ND: Not determined (n=8–9). Error bars are mean ± SD. Data are from at least two independent experiments.

Figure 1—figure supplement 2
Kidney T cell infiltrates in WT and VP1-mAb treated μMT mice.

(A–B) Number of kidney-infiltrating CD4 T cells (A) and CD8 T cells (B) in the kidneys of WT and VP1-mAb-treated μMT mice 30 dpi. Kidney infiltrating cells were identified by i.v. injection of anti-CD45 prior to euthanasia (n=7–8). Error bars are mean ± SD. Data are from at least two independent experiments. Data were analyzed by Mann-Whitney U test (A–B).

Figure 1—figure supplement 3
Confirmation of T cell depletion.

(A) Representative plot of CD4+ and CD8+ cells in the blood of mice treated with IgG or αCD4 and αCD8β at euthanasia 30 days post infection (10 days after the start of antibody treatment). Plots gated on CD45+ cells. (B–C) Frequency of CD4+ (B) and CD8+ (C) cells of total CD45+ cells in the blood of mice treated with IgG or αCD4 and αCD8β (n=11–13). (D) Representative plot of CD4+ and CD8+ cells in the blood of mice treated with IgG, αCD4, or αCD8β at euthanasia 30 days post infection (10 days after the start of antibody treatment). Plots gated on CD45+ cells. (E–F) Frequency of CD4+ (E) and CD8+ (F) cells of total CD45+ cells in the blood of mice treated with IgG, αCD4, or αCD8β (n=13–21). Error bars are mean ± SD. Data are from at least two independent experiments.

Figure 2 with 1 supplement
T cell loss enables viremia.

(A) Experimental scheme for VP1 mAb treatment, T cell depletion, and detection of viremia. (B) (Left) Viral titers in the blood of control and T cell depleted mice over time. Viremia was measured by plaque assay from whole blood. (Center) Peak levels of viremia detected in control and T cell depleted mice. (Right) Time to development of viremia in control and T-cell-depleted mice. Indicated significances are with comparison to the IgG group. LOD: Limit of detection (n=10–27). (C–E) Viral DNA levels in the kidney (C), spleen (D), and brain (E) at the time of euthanasia in control and T-cell-depleted mice. Viral DNA was quantified by qPCR and compared to a standard curve (n=9–27). Error bars are mean ± SD. Data are from at least two independent experiments. Data were analyzed by Kruskal-Wallis test with Dunn’s multiple comparisons test (B Center), (C–E) or Mantel-Cox test with Bonferroni’s correction for multiple comparisons (B Right). *p<0.05, ***p<0.001, ****p<0.0001.

Figure 2—figure supplement 1
Confirmation of T cell depletion.

(A–B) Frequency of CD4+ (A) and CD8+ (B) cells of total CD45+ cells from the blood of mice treated with IgG, αCD4 and αCD8β, αCD4, or αCD8β. Blood was collected at euthanasia (n=9–27). Error bars are mean ± SD. Data are from at least two independent experiments.

Viremia is mediated by VP1 Ab escape mutations.

(A) Location of VP1 mutations (blue) in the HI loop of one copy of VP1 relative to the location of VP1 mAb (cyan/purple) in the Cryo-EM structure of WT VP1 and the VP1 mAb. PDB ID: 7K22 (Lauver et al., 2020). (B) VP1 mAb neutralization of VP1 mutant viruses. Viruses were preincubated with 10 μg VP1 mAb or control IgG for 30 min prior to addition to 1x105 NMuMG epithelial cells. A2 was diluted to an MOI of 0.1 PFU/cell, mutant viruses were diluted to match A2 by genomic equivalents (g.e.). Viral LT mRNA levels were quantified 24 hpi and normalized for each virus to infection with control IgG (n=6). (C–D) Viral mRNA levels in the kidney (C) and spleen (D) 4 dpi with VP1 mutant viruses compared to WT. WT mice were infected i.v. with 1x106 PFU of A2 or mutant viruses matched by g.e. Viral LT mRNA levels were quantified by qPCR and compared to a standard curve (n=6–7). Error bars are mean ± SD. Data are from at least two independent experiments. Data were analyzed by Mann-Whitney U test with Holm-Šídák correction for multiple comparisons (B) or Kruskal-Wallis test with Dunn’s multiple comparisons test (C–D). *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001.

Figure 4 with 1 supplement
T cells prevent the outgrowth of Ab escape mutant virus.

(A) Experimental scheme for VP1 mAb treatment, T cell depletion, A2.Δ295 challenge, and detection of viremia in μMT mice. (B) (Left) Viral titers in the blood of control and T-cell-depleted mice over time. Viremia was measured by plaque assay from whole blood. (Center) Peak levels of viremia detected in control and T-cell-depleted mice. (Right) Time to development of viremia in control and T-cell-depleted mice in B. LOD: Limit of detection (n=8–9). (C–E) Viral DNA levels in the kidney (C), spleen (D), and brain (E) 28 days post challenge (n=8–9). Error bars are mean ± SD. Data are from two independent experiments. Data were analyzed by Mantel-Cox test (B Right) or Mann-Whitney U test (B center), (C–E). ***p<0.001, ****p<0.0001.

Figure 4—figure supplement 1
Confirmation of T cell depletion.

(A–B) Frequency of CD4+ (A) and CD8+ (B) cells out of total CD45+ cells the blood of mice treated with IgG or αCD4 and αCD8β. Blood was collected 2 weeks after the start of antibody treatment (n=8–9). Error bars are mean ± SD. Data are from at least two independent experiments.

Figure 5 with 1 supplement
A compensatory mutation in VP1 arises to rescue defects in receptor binding caused by an Ab escape mutation.

(A) VP1 mAb neutralization assay with D295N and Δ297 VP1 mutant viruses. Viruses were preincubated with 10 μg VP1 mAb or control IgG for 30 min prior to addition to 1x105 NMuMG epithelial cells. A2 was diluted to an MOI of 0.1 PFU/cell, mutant viruses were diluted to match A2 by g.e. Viral LT mRNA levels were quantified 24 hpi and normalized for each virus to infection with control IgG (n=6). (B) Binding of VP1 mAb to WT or VP1 mutant viruses. Wells were coated with 1x109 g.e. of WT or VP1 mutant virus and incubated with VP1 mAb. VP1 mAb binding was quantified using an anti-rat secondary antibody and values were normalized to binding to WT virus (n=4). (C) Binding avidity of VP1 mAb for A2 and A2.D295N. VP1 mAb binding to A2 and A2.D295N was performed as in B. Prior to detection of mAb binding, virus/mAb complexes were treated with varying concentrations of NH4SCN for 15 min. Binding at each concentration was normalized to binding at 0 M NH4SCN for each virus (n=4). (D) Quantification of virus production in a single round of replication by plaque assay. Virus was added to 1x105 A31 cells at an MOI of 0.1 PFU/cell. Cells were lysed at 60 hpi and infectious virus was quantified by plaque assay and divided by the input virus quantity. A2.Δ297 was not included due to inability of this mutant to form plaques (n=8). (E) Frequency of T-antigen-positive cells 24 hpi with WT or mutant viruses. 1x105 A31 cells were infected with A2 at an MOI of 1 PFU/cell or mutant viruses matched by g.e. Cells were collected at 24 hpi, permeabilized, stained for T ag protein, and quantified by flow cytometry. (F) Frequency of T-antigen-positive cells at 24, 48, 72, and 96 hpi with WT or mutant viruses. 1x105 A31 cells were infected with A2 at an MOI of 0.1 PFU/cell or mutant viruses matched by g.e. Cells were collected at each time point, permeabilized, stained for T ag protein, and quantified by flow cytometry (n=6–13). (G) Detection of virus binding in kidney sections. PFA-fixed frozen kidney sections were treated with neuraminidase or buffer alone prior to incubation with WT or VP1 mutant virus. Sections were then stained for VP1 (red) and kidney markers [CD13 (white), THP (green)]. Neu: Neuraminidase. Representative of three independent experiments. (H) LT mRNA levels in the kidney 4 dpi with WT or mutant viruses. WT mice were infected i.v. with 1x106 PFU of A2 or mutant viruses matched by g.e. Viral LT mRNA levels were quantified by qPCR and compared to a standard curve (n=9–10). (I) Detection of D295 and H297 mutations in the kidney. VP1 sequences were PCR amplified from kidney DNA samples of the mice that developed the D295N/Δ297 and D295A/Δ297 double mutant viruses. VP1 clones were sequenced and screened for the presence of mutations at D295 and H297; the frequency of each mutation in 50 clones is shown. Error bars are mean ± SD. Data are from at least two independent experiments. Data were analyzed by Mann-Whitney U test with Holm-Šídák correction for multiple comparisons (A), one-way ANOVA with Dunnett’s multiple comparisons test (B and D–E), two-way ANOVA with Dunnett’s multiple comparisons test (F), or Kruskal-Wallis test with Dunn’s multiple comparisons test (H). **p<0.01, ***p<0.001, ****p<0.0001.

Figure 5—figure supplement 1
Defects in mutant virus spread and receptor binding.

(A) (Left) Plaque formation by WT and mutant viruses in A31 fibroblasts 6 dpi. (Right) Area of individual plaques formed by A2, A2.D295N, and A2.D295N/Δ297. Plaque area was measured using ImageJ (n=80). (B) Encapsidated genomes produced by WT and mutant viral DNA transfection. Data is relative to WT (n=9). (C) pH-dependent hemagglutination curves for WT and mutant viruses. The reported HA titer was the inverse of the highest dilution showing hemagglutination (n=3). (D) Percent of virus binding lost with neuraminidase pre-treatment. Bound virus was detected with polyclonal VP1 Ab and quantified by flow cytometry. For each virus, the percent of binding lost to neuraminidase treatment was calculated by comparing virus binding with and without neuraminidase treatment (n=6). (E) Quantification of T antigen positive cells 24 hpi with WT or mutant viruses with neuraminidase pretreatment. Cells were treated and infected as in (E). At 24 hpi, cells were collected, permeabilized, stained for T ag protein, and quantified by flow cytometry (n=6). (F) Quantification of LT mRNA levels with neuraminidase pretreatment. A31 cells were treated with neuraminidase and then infected with A2 at an MOI of 0.1 PFU/cell or mutant viruses matched by g.e. Viral LT mRNA levels were quantified by qPCR and compared to a standard curve (n=6). Error bars are mean ± SD. Data are from at least two independent experiments. Data were analyzed by Kruskal-Wallis test with Dunn’s multiple comparisons test (A), one-way ANOVA with Dunnett’s multiple comparisons test (B and D), or Mann-Whitney U test with Holm-Šídák correction for multiple comparisons (E–F). **p<0.01, ***p<0.001, ****p<0.0001.

Reduced kidney tropism and heightened neurovirulence by a VP1 double mutant virus.

(A) Viremia in μMT mice 30 dpi with A2 or A2.D295N/Δ297 i.v. Viremia was quantified by plaque assay from whole blood (n=9). (B) H&E stained sagittal sections of kidneys from μMT mice 30 dpi after i.v. inoculation with A2 or A2.D295N/Δ297. Left: ×50 magnification. Right: ×500 magnification. (C) Foci of VP1+ cells in kidneys from μMT mice 30 dpi with A2 or A2.D295N/Δ297 i.v. Kidneys were stained for CD13 (white), THP (green) and VP1 (red). (D) Quantification of the number (Left, n=9) and size (Right, n=42–45) of VP1+ foci in kidneys from μMT mice 30 dpi with A2 or A2.D295N/Δ297 i.v. Foci number is the average of two sagittal kidney sections per mouse. For quantifying foci area, five random foci per mouse or the maximum number of foci found were imaged and the area of each VP1+ focus was calculated using ImageJ. (E) VP1+ cells in the lateral (top) and third (bottom) ventricles of WT mice 4 dpi with A2 or A2.D295N/Δ297 inoculated i.c. and quantification of VP1+ cells in the third ventricle (n=16–18). (F) H&E stained coronal sections from brains of sham (top left), A2 (top right), or A2.D295N/Δ297 (bottom left) i.c.-inoculated mice 30 dpi (×200 magnification). Arrows indicate sites of ependymal inflammation. (G) VP1 staining in the ventricles of WT mice 30 dpi with A2 or A2.D295N/Δ297 i.c. VP1+ cells are indicated with white markers. (H) Quantification of hydrocephalus 30 dpi after i.c. inoculation with vehicle, A2, or A2.D295N/Δ297. Coronal head height was measured with a Vernier caliper in line with the ear canal to the nearest 0.5 mm (n=10–13). Error bars are mean ± SD. Data are from at least two independent experiments. Data were analyzed by Mann-Whitney U test (A, D, and E) or one-way ANOVA with Dunnett’s multiple comparisons test (H). ***p<0.001, ****p<0.0001.

T cell deficiency and a narrow antiviral Ab response enable emergence of Ab-escape, neurovirulent PyV variants.

Model for the evolution of Ab escape, neurovirulent PyV VP1 variants in the setting of T cell deficiency and a neutralizing Ab response with limited coverage of VP1 epitopes on the virus capsid. Figure was created with Biorender.com.

Tables

Key resources table
Reagent type (species) or resourceDesignationSource or referenceIdentifiersAdditional information
AntibodyAnti-VP1 (Rat Clone 8A7H5)Swimm et al., 2010Clone 8A7H5250 µg/week
AntibodyChromPure Rat IgGJackson ImmunoResearchCat#012-000-003
RRID: AB_2337136
250 µg/week
AntibodyAnti-CD8β (H35-17.2)Golstein et al., 1982N/A250 µg/week
AntibodyAnti-CD4 (GK1.5)Dialynas et al., 1983N/A250 µg/week
AntibodyAnti-VP1 (Rabbit polyclonal)Provided by Robert GarceaN/AIF (1:1,000)
FC (1:10,000)
AntibodyAnti-Vimentin (Rat Clone 280618)R&D SystemsCat#MAB2105
RRID: AB_2241653
IF (1:500)
AntibodyAnti-GFAP (Goat polyclonal)AbcamCat#ab53554
RRID: AB_880202
IF (1:1,000)
AntibodyAnti-THP (Rat monoclonal)R&D SystemsCat#MAB5175
RRID: AB_2890000
IF (1:1,000)
AntibodyAnti-CD13 (Goat polyclonal)R&D SystemsCat#AF2335
RRID: AB_2227288
IF (1:500)
AntibodyAnti-Goat IgG AF488 (Bovine polyclonal)Jackson ImmunoResearchCat#805-545-180
RRID: AB_2340883
IF (1:500)
AntibodyAnti-Rat IgG AF555 (Donkey polyclonal)AbcamCat#ab150154
RRID: AB_2813834
IF (1:500)
AntibodyAnti-Rabbit IgG AF647 (Donkey polyclonal)Jackson ImmunoResearchCat#711-605-152
RRID: AB_2492288
IF (1:500)
AntibodyAnti-CD8α-AF700 (Clone 53–6.7)BiolegendCat#100730
RRID:AB_493703
FC (1:200)
AntibodyAnti-CD45-PerCP/Cy5.5 (Clone 30-F11)BiolegendCat#103132
RRID:AB_893340
FC (1:200)
AntibodyAnti-CD45-FITC (Clone 30-F11)BDCat#553080
RRID:AB_394610
FC (3 µg/mouse)
AntibodyAnti-CD4-PE (Clone RM4-5)BiolegendCat#100512
RRID:AB_312715
FC (1:200)
AntibodyAnti-CD4-BV421
(clone GK1.5)
BDCat#562891
RRID: AB_2737870
FC (1:200)
AntibodyAnti-Rat IgG-APC (Goat polyclonal)BDCat#551019
RRID:AB_398484
FC (1:200)
AntibodyAnti-Mouse IgG-HRP (Goat polyclonal)Bethyl Laboratories INCCat#A90-116P
RRID:AB_67183
ELISA (1:7,000)
OtherFixable Viability Dye eFluor 780ThermoFisherRef#65-0865-14FC (1:1,000)
OtherFlow Cytometry Absolute Count StandardBangs LaboratoriesCat#580
OtherMuPyV (Strain A2)N/AN/A
OtherSheep Red Blood CellsInnovative ResearchCat#ISHRBC10P15ML
Peptide, recombinant proteinVP1 pentamersProvided by Robert GarceaN/A
Peptide, recombinant proteinBenzonase NucleaseSigmaCat#E1014
Peptide, recombinant proteinNeuraminidase from Vibrio choleraeSigmaCat#N6514IF (1:100)
FC (1:200)
Peptide, recombinant proteinCollagenase (Type I)WorthingtonCat#LS004197
Chemical compound, drugTRIzol ReagantThermoFisherRef#15596018
Chemical compound, drugRevertAid H Minus Reverse TranscriptaseThermoFisherCat#EP0451
Chemical compound, drugLipofectamine 2000 Transfection ReagentThermoFisherCat#11668030
Chemical compound, drugLipofectamine 3000 Transfection ReagentThermoFisherCat# L3000008
Commercial assay, kitPFHM-II Protein-Free Hybridoma MediumThermoFisherRef#12040–077
Commercial assay, kitTOPO TA Cloning KitThermoFisherRef#45–0641
Commercial assay, kitTBP PrimeTime XL qPCR AssayIDTMm.PT.39a.22214839
Commercial assay, kit1-Step Ultra TMB-ELISAThermoFisherRef#34028
Commercial assay, kit96 Well EIA/RIA Polystyrene High Bind MicroplateFisher ScientificCat#3590
Commercial assay, kitPerfectCTa FastMix II ROXQuantabioP/N 84210
Commercial assay, kitPureLink Viral RNA/DNA mini KitThermoFisherRef#12280–050
Commercial assay, kitWizard Genomic DNA Purification KitPromegaRef#A1120
Commercial assay, kitQuikChange II Site-Directed Mutagenesis KitAgilentCat#200523
Commercial assay, kitCELLine Disposable BioreactorFisher ScientificCat#353137
Cell Line (M. musculus)BALB/3T3 Clone A31ATCCCCL-163; RRID:CVCL_0184
Cell Line (M. musculus)NMuMGATCCCRL-1636; RRID:CVCL_0075
Strain, strain background (M. musculus)C57BL/6 MiceNational Cancer InstituteCat#OIC55
Genetic reagent (M. musculus)μMT MiceJackson LaboratoryCat#002288; RRID:IMSR_JAX:002288
Software, algorithmPrismGraphpadv 9.3.1; RRID:SCR_002798
Software, algorithmFlowJoBDv 10.6.1; RRID:SCR_008520
Software, algorithmImageJNIHv 1.8.0; RRID:SCR_003070
Software, algorithmLeica LAS XLeicav 3.7.2; RRID:SCR_013673

Additional files

Supplementary file 1

VP1 mutations.

Identity, location, and frequency of detected VP1 mutations. Superscripted numbers indicate the VP1 loop in which the mutations are located (1: BC, 2: DE, 3: EF, 4: HI). Deletions are indicated by a “Δ” followed by the deleted residues; the identity of the deleted amino acids is indicated in “()”. The duplication of a residue is indicated with “dup.” The presence of two mutations in a virus is indicated with “+”. Sets of mutations separated by “and” indicate that both of the listed mutant viruses were isolated from the same mouse.

https://cdn.elifesciences.org/articles/83030/elife-83030-supp1-v2.docx
Supplementary file 2

Oligonucleotide sequences.

Sequences of oligonucleotides used for site-directed mutagenesis, cloning, qPCR, and sequencing.

https://cdn.elifesciences.org/articles/83030/elife-83030-supp2-v2.docx
Supplementary file 3

Statistical information.

Statistical tests used and exact p values for the comparisons of data presented in each figure as indicated.

https://cdn.elifesciences.org/articles/83030/elife-83030-supp3-v2.docx
MDAR checklist
https://cdn.elifesciences.org/articles/83030/elife-83030-mdarchecklist1-v2.docx

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. Matthew D Lauver
  2. Ge Jin
  3. Katelyn N Ayers
  4. Sarah N Carey
  5. Charles S Specht
  6. Catherine S Abendroth
  7. Aron E Lukacher
(2022)
T cell deficiency precipitates antibody evasion and emergence of neurovirulent polyomavirus
eLife 11:e83030.
https://doi.org/10.7554/eLife.83030