Delineating the transcriptional landscape and clonal diversity of virus-specific CD4+ T cells during chronic viral infection

  1. Ryan Zander  Is a corresponding author
  2. Achia Khatun
  3. Moujtaba Y Kasmani
  4. Yao Chen
  5. Weiguo Cui  Is a corresponding author
  1. Blood Research Institute, Versiti Wisconsin, United States
  2. Department of Microbiology and Immunology, Medical College of Wisconsin, United States
4 figures, 1 table and 2 additional files

Figures

Figure 1 with 1 supplement
Clonal distribution and transcriptional landscape of GP66-specific CD4+ T cells during chronic lymphocytic choriomeningitis virus (LCMV) infection.

(A–I) GP66-specific splenic CD4 T cells were isolated and sort-purified from two individual LCMV Clone 13 (Cl13)-infected mice on day 10 post-infection, and the 10× Genomics pipeline was used to generate a combined single cell RNA-sequencing (scRNA-seq) + T cell receptor (TCR) seq library. A downstream computational analysis was then performed in R. (A) Summary data showing the size of and clonal distribution of GP66-specific CD4+ T cells (with at least 2 cells per clone) recovered from the two separate mice. (B) Pie charts showing the shared number of public clones when assessing the sequence of either TCR alpha chains, TCR beta chains, or both TCR alpha and beta chains. (C) Summary data showing the relative frequency of each T cell clone recovered from the two mice. (D) Uniform manifold approximation and projection (UMAP) plot showing the presence of eight transcriptionally distinct clusters of GP66-specific CD4 T cells. (E) Dot plot showing the relative expression of differentially expressed genes between the eight clusters. (F–H) UMAP plots depicting the expression of either T helper type 1 (Th1) cell-associated genes (F), T follicular helper (Tfh) cell-associated genes (G), or memory-associated genes (H). (I) Module score analyses were performed to compare the relative Th1, Tfh, and memory T cell signatures among the distinct clusters of GP66-specific CD4+ T cells.

Figure 1—figure supplement 1
CD4+ T cells responding to chronic viral infection are composed of several transcriptionally and functionally distinct populations.

(A) Heatmap showing the top differentially expressed genes in all eight clusters. (B) Violin plots displaying the relative expression of indicated genes in Ly6c-hi and Lag3-hi T helper type 1 (Th1) cell subsets. (C) Module score analysis showing the average expression levels of the previously identified T-bet T follicular helper type 1 (Tfh1) program in the eight clusters identified in Figure 1D. (D) Dot plot summary showing the average expression of indicated cytokines and chemokines, and the percent of CD4+ T cell subsets expressing each molecule.

Validation of heterogeneity in CD4 T cells responding to chronic lymphocytic choriomeningitis virus (LCMV) infection using spectral flow cytometry.

(A) MX1-GFP reporter mice were infected with LCMV Clone 13 (Cl13), and spectral flow cytometry was used to assess the phenotype of splenic CD4 T cells on day 25 p.i. Using a panel of 18 parameters, t-distributed stochastic neighbor embedding (t-SNE) visualization was performed on total PD-1hi CD4 T cells (which include both GP66-specific and other LCMV epitope-reactive CD4 T cells). Representative t-SNE flow plots in A are displayed for one mouse, with data being representative of three individual mice. (B) Representative flow plots showing potential gating strategy to identify CX3CR1 ± CXCR6+T helper type 1 (Th1) cell subsets. (C) Summary data showing the relative expression of indicated surface molecules in CX3CR1+ and CX3CR1− Th1 cell subsets. (D) Representative flow plots showing potential gating strategies to identify Slamf7+Eomes + (far left), and IL-1R2+CD4 T cell subsets (which may be composed of pre-T follicular helper [Tfh] and Tfh1 subsets). Data (mean ± SEM) in C are from three individual mice, and were analyzed with unpaired student’s t-tests. Data are representative of at least two independent experiments. * p < 0.05, ** p < 0.01.

Figure 3 with 1 supplement
Trajectory analysis and clonal lineage tracing highlights that most GP66-specific CD4 T cells display developmental plasticity during chronic lymphocytic choriomeningitis virus (LCMV) infection, although a small fraction of clones exhibit a biased cell fate decision.

(A and B) Monocle tree trajectory plots showing predicted cellular differentiation of GP66-specific CD4 T cell subsets based on trajectory analysis. (C) Summary chart displaying clonal overlap among GP66+ CD4 T cell subsets. (D) Uniform manifold approximation and projection (UMAP) plot as in Figure 1D, but colored by clone membership. (E) Heatmap showing subset distribution and frequency of 329 clones (n=168 clones from mouse 1 [M1] and n=159 clones from mouse 2 [M2]) among indicated CD4 T cell lineage (T helper type 1 [Th1], T follicular helper [Tfh], and Slamf6hi memory) as defined by Monocle state.

Figure 3—figure supplement 1
Trajectory analysis and clonal lineage tracing indicate that a small fraction of GP66-specific CD4 T cells display display a biased cell fate decision.

(A) Heatmap showing subset distribution and frequency of 176 clones (with four or more cells per clone) among indicated CD4 T cell lineage (T helper type 1 [Th1], T follicular helper [Tfh], and Slamf6hi memory) as defined by Monocle state. (B–C) Logistic regression analysis comparing Th1 (B) or Tfh (C) single cell-fate biased clones vs subset-specific (Th1, (B); Tfh (C)) module scores calculated at the clonal level. p Values were calculated using Wald test.

Figure 4 with 1 supplement
CD4 T cells responding to chronic viral infection display altered subset distribution patterns and core gene expression modules compared to their acute infection counterparts.

(A) A canonical correlation analysis (CCA) was performed on single cell RNA-sequencing (scRNA-seq) libraries obtained from GP66-specific CD4+ T cells during chronic lymphocytic choriomeningitis virus (LCMV) Clone 13 (Cl13) infection (this study) and acute LCMV Armstrong infection (Khatun et al., 2021) that were integrated together using the integration function in Seurat (Butler et al., 2018). (B) Summary graph showing the relative subset distribution of GP66 + CD4 T cells subsets during acute and chronic LCMV infection on day 10 p.i. (C–H) Module score analyses showing the average expression levels of the T helper type 1 (Th1), T follicular helper (Tfh), memory T cell, T cell receptor (TCR) signaling, and T cell dysfunction programs among the CD4 T cell subsets identified in A. (I–J) Summary dot plots showing the average gene expression and the proportion of each cell subset expressing indicated gene. All genes listed in (I) were among the top 50 differentially expressed genes that were upregulated across multiple distinct subsets in response to chronic LCMV infection.

Figure 4—figure supplement 1
CD4 T cells responding to chronic lymphocytic choriomeningitis virus (LCMV) infection display altered subset distribution patterns and changes in gene expression programs compared to their acute infection counterparts.

(A) Representative flow plots (left) and summary data (right) showing the proportion of the Slamf7+Eomes+ subset among T cell receptor (TCR) Tg Smarta CD4+ T cells on day 14 p.i. (B–E) Module score analyses showing the average expression levels of genes upregulated in memory cells vs effector cells (B), T cell memory precursor (Tcmp) or progenitor CD8 T cell signatures (C and D), or an effector CD8 T cell gene signature in each of the eight clusters identified in our single cell RNA-sequencing (scRNA-seq) analysis. (F) Summary dot plot showing the relative expression of each indicated gene (that were selected from gene sets used for module score analyses) and the proportion of each CD4+ T cell subset expressing the respective gene. Data (mean ± SEM) in (A) are from three to four mice per group, were analyzed using an unpaired student’s t-test, and are representative of at least two independent experiments. ** p < 0.01.

Tables

Key resources table
Reagent type (species) or resourceDesignationSource or referenceIdentifiersAdditional information
Strain and strain backgroundLymphocytic choriomeningitis virus (LCMV) Clone 13 (Cl13) strainRafi Ahmed LaboratoryGrew up in house
Strain and strain backgroundLCMV Armstrong strainRafi Ahmed LaboratoryGrew up in house
AntibodyAPC/fire rat monoclonal anti-mouse CD4 (GK1.5)BiolegendCat #100460 RRID:AB_25721111:300
AntibodyAF647 rat monoclonal anti-mouse CD4 (GK1.5)BiolegendCat #100460 RRID:AB_25721111:300
AntibodyPe/Cyanine7 rat monoclonal anti-mouse PD.1 (RMP1-30)BiolegendCat #109110 RRID:AB_5720171:200
AntibodyAPC/Cyanine7 rat monoclonal anti-mouse PD.1 (29F.1A12)BiolegendCat #135224 RRID:AB_ 25635231:200
AntibodyAPC/Cyanine7 rat monoclonal anti-mouse/human CD44 (IM-7)BiolegendCat #103028 RRID:AB_8307851:1000
AntibodyAPC rat monoclonal anti-mouse CXCR5 (L138D7)BiolegendCat #145506
RRID: AB_2561970
1:100
AntibodyPE/Dazzle 594 rat monoclonal anti-mouse CXCR6 (SA051D1)BiolegendCat #151117 RRID:AB_27217001:200
AntibodyPerCPeFluor710 rat monoclonal anti-mouse CXCR6 (Danid2)ThermofisherCat # 46-9186-82
RRID:AB_ AB_2734904
1:200
AntibodyBV785 rat monoclonal anti-mouse Lag3 (C9B7W)BiolegendCat # 125219
RRID:AB_256657
1:100
AntibodyPE/Cyanine7 rat monoclonal anti-mouse CD127 (A7R34)BiolegendCat #135014 RRID:AB_19372651:100
AntibodyBrilliant Blue mouse monoclonal anti-mouse 700 Tim3 (5D12)BD BiosciencesCat #747619
RRID:AB_2744185
1:100
AntibodySpark NIR 685 Armenian hamster monoclonal anti-mouse CD69 (H1.2F3)BiolegendCat #104557
RRID:AB_2860607
1:100
AntibodyBUV395 rat monoclonal anti-mouse CD44 (IM7)BD BiosciencesCat #740215
RRID: AB_2739963
1:500
AntibodyBV711 rat monoclonal anti-mouse PSGL1 (2PH1)BD BiosciencesCat #740746
RRID: AB_2740414
1:100
AntibodyBV650 rat monoclonal anti-mouse CD83 (Michel-19)BD BiosciencesCat #740619
RRID: AB_2740317
1:100
AntibodyBV605 rat monoclonal anti-mouse PD-1 (29 F.1A12)BiolegendCat #135220
RRID: AB_2562616
1:200
AntibodyBV510 mouse monoclonal anti-mouse Ly108 (13G3)BD BiosciencesCat #745073
RRID: AB_2742691
1:200
AntibodyPercp cy5.5 mouse monoclonal anti-mouse CX3CR1 (SA011F11)BiolegendCat #149010
RRID: AB_2564494
1:100
AntibodyPerCP rat monoclonal anti-mouse Ly-6c (HK1.4)BiolegendCat #128028
RRID: AB_10897805
1:100
AntibodyPacific Blue rat anti-mouse Bst2BiolegendCat #127108
RRID: AB_2028455
1:100
AntibodyBV421 rat monoclonal anti-mouse Slamf7 (129C1)BD BiosciencesCat #747997
RRID: AB_2872458
1:100
AntibodyGoat polyclonal Anti-mouse IL1R2ThermofisherCat # PA5-47759
RRID:AB_11152697
1:400
AntibodyPe/Cyanine5 syrian hamster monoclonal anti-mouse ICOS (15F9)BiolegendCat # 107708
RRID: AB_313337
1:100
AntibodyAPC-Fire750 rat monoclonal anti-mouse OX40 (OX-86)BiolegendCat #119423
RRID: AB_2715993
1:100
OtherPE LCMV I-A(b) GP66-77 tetramerNIH Tetramer Core Facilityhttps://tetramer.yerkes.emory.edu1:500
Commercial assay or kitChromium Single Cell 3’ Library & Gel Bead Kit v210× GenomicsCat#PN-120267
Commercial assay or kitChromium Single Cell A Chip Kit10× GenomicsCat#PN-1000009
Commercial assay or kitChromium i7 Multiplex Kit10× GenomicsCat# PN-120262
Commercial assay or kitDynabeads MyOne SilaneThermofisherCat#37002D
Commercial assay or kitSPRIselect Reagent KitBeckman CoulterCat#B23318
Commercial assay or kitKappa NGS quantification kitKAPABiosystemsCat#KK4824
Commercial assay or kitNextSeq 500/550 High Output Kit v2.5 (150 cycles)IlluminaCat#20024907
Software and algorithmCell Ranger10× Genomicshttps://support.10xgenomics.com/single-cell-gene-expression/software/pipelines/latest/installation
Software and algorithmSeuratSatija et al., 2015https://satijalab.org/seurat/
Software and algorithmMonocle-2Qiu et al., 2017;
Trapnell et al., 2014
http://cole-trapnell-lab.github.io/monocle-release/docs/
Software and algorithmFlowjo Version 10.5.3Tree StarN/A
Software and algorithmPrism 8Graphpad SoftwareN/A

Additional files

MDAR checklist
https://cdn.elifesciences.org/articles/80079/elife-80079-mdarchecklist1-v2.docx
Supplementary file 1

Clonal Distribution and cellular fate of GP66-specific CD4 T cell clones (with 4 or more cells per clone) during LCMV Cl13 infection.

https://cdn.elifesciences.org/articles/80079/elife-80079-supp1-v2.xls

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. Ryan Zander
  2. Achia Khatun
  3. Moujtaba Y Kasmani
  4. Yao Chen
  5. Weiguo Cui
(2022)
Delineating the transcriptional landscape and clonal diversity of virus-specific CD4+ T cells during chronic viral infection
eLife 11:e80079.
https://doi.org/10.7554/eLife.80079