Abstract
T cell antigen receptor (TCR) recognition followed by clonal expansion is a fundamental feature of adaptive immune responses. Here, we present a mass cytometric (CyTOF) approach to track T cell responses by combining antibodies for specific TCR Vα and Vβ chains with antibodies against T cell activation and differentiation proteins in mice. This strategy identifies expansions of CD8+ and CD4+ T cells expressing specific Vβ and Vα chains with varying differentiation states in response to Listeria monocytogenes, tumors and respiratory influenza infection. Expanded T cell populations expressing Vβ chains could be directly linked to the recognition of specific antigens from Listeria, tumor cells or influenza. In the setting of influenza infection, we found that common therapeutic approaches of intramuscular vaccination or convalescent serum transfer altered the TCR diversity and differentiation state of responding T cells. Thus, we present a method to monitor broad changes in TCR use paired with T cell phenotyping during adaptive immune responses.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$29.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$209.00 per year
only $17.42 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout
Similar content being viewed by others
Data availability
Preprocessed and debarcoded CyTOF data of gated populations used in this study have been uploaded to Mendeley Data under https://doi.org/10.17632/dpm5rm7cxj.1. Variable chain usage information derived from TCR-seq data used in this study have been uploaded to Mendeley Data under https://doi.org/10.17632/dpm5rm7cxj.1.
Code availability
Automated Vα-Vβ assignments script is available at GitHub via https://github.com/SpitzerLab/Semi-supervised-Clonotyping-Assignments/.
References
Abdelbary, M., Hobbs, S. J., Gibbs, J. S., Yewdell, J. W. & Nolz, J. C. T cell receptor signaling strength establishes the chemotactic properties of effector CD8+ T cells that control tissue-residency. Nat. Commun. 14, 3928 (2023).
Huppa, J. B., Gleimer, M., Sumen, C. & Davis, M. M. Continuous T cell receptor signaling required for synapse maintenance and full effector potential. Nat. Immunol. 4, 749–755 (2003).
Zikherman, J. & Au-Yeung, B. The role of T cell receptor signaling thresholds in guiding T cell fate decisions. Curr. Opin. Immunol. 33, 43–48 (2015).
Moon, J. J. et al. Naive CD4+ T cell frequency varies for different epitopes and predicts repertoire diversity and response magnitude. Immunity 27, 203–213 (2007).
Tan, T. C. J. et al. Suboptimal T-cell receptor signaling compromises protein translation, ribosome biogenesis, and proliferation of mouse CD8 T cells. Proc. Natl Acad. Sci. USA 114, E6117–E6126 (2017).
Soerens, A. G. et al. Functional T cells are capable of supernumerary cell division and longevity. Nature 614, 762–766 (2023).
Bousso, P. et al. Diversity, functionality, and stability of the T cell repertoire derived in vivo from a single human T cell precursor. Proc. Natl Acad. Sci. USA 97, 274–278 (2000).
DuPage, M. & Bluestone, J. A. Harnessing the plasticity of CD4+ T cells to treat immune-mediated disease. Nat. Rev. Immunol. 16, 149–163 (2016).
Akondy, R. S. et al. Origin and differentiation of human memory CD8 T cells after vaccination. Nature 552, 362–367 (2017).
Sarkar, S. et al. Strength of stimulus and clonal competition impact the rate of memory CD8 T cell differentiation12. J. Immunol. 179, 6704–6714 (2007).
Joshi, N. S. & Kaech, S. M. Effector CD8 T cell development: a balancing act between memory cell potential and terminal differentiation. J. Immunol. 180, 1309–1315 (2008).
Wei, S. C. et al. Distinct cellular mechanisms underlie anti-CTLA-4 and anti-PD-1 checkpoint blockade. Cell 170, 1120–1133 (2017).
Cha, E. et al. Improved survival with T cell clonotype stability after anti-CTLA-4 treatment in cancer patients. Sci. Transl. Med. 6, 238ra70 (2014).
Sheikh, N. et al. Clonotypic diversification of intratumoral T cells following Sipuleucel-T treatment in prostate cancer subjects. Cancer Res. 76, 3711–3718 (2016).
Reuben, A. et al. Comprehensive T cell repertoire characterization of non-small cell lung cancer. Nat. Commun. 11, 603 (2020).
Anderton, S. M. & Wraith, D. C. Selection and fine-tuning of the autoimmune T-cell repertoire. Nat. Rev. Immunol. 2, 487–498 (2002).
Peri, A. et al. The landscape of T cell antigens for cancer immunotherapy. Nat. Cancer 4, 937–954 (2023).
De Simone, M., Rossetti, G. & Pagani, M. Single cell T cell receptor sequencing: techniques and future challenges. Front. Immunol. 9, 384005 (2018).
Chiffelle, J. et al. T-cell repertoire analysis and metrics of diversity and clonality. Curr. Opin. Biotechnol. 65, 284–295 (2020).
Yost, K. E. et al. Clonal replacement of tumor-specific T cells following PD-1 blockade. Nat. Med. 25, 1251–1259 (2019).
Jiang, N., Schonnesen, A. A. & Ma, K.-Y. Ushering in integrated T cell repertoire profiling in cancer. Trends Cancer Res. 5, 85–94 (2019).
Carter, J. A. et al. Single T cell sequencing demonstrates the functional role of αβ TCR pairing in cell lineage and antigen specificity. Front. Immunol. 10, 464451 (2019).
Anthony, D. D. & Lehmann, P. V. T-cell epitope mapping using the ELISPOT approach. Methods 29, 260–269 (2003).
Fehlings, M. et al. Checkpoint blockade immunotherapy reshapes the high-dimensional phenotypic heterogeneity of murine intratumoural neoantigen-specific CD8+ T cells. Nat. Commun. 8, 562 (2017).
Hombrink, P. et al. High-throughput identification of potential minor histocompatibility antigens by MHC tetramer-based screening: feasibility and limitations. PLoS ONE 6, e22523 (2011).
Spitzer, M. H. et al. An interactive reference framework for modeling a dynamic immune system. Science 349, 1259425 (2015).
Pilch, H. et al. Improved assessment of T-cell receptor (TCR) VB repertoire in clinical specimens: combination of TCR-CDR3 spectratyping with flow cytometry-based TCR VB frequency analysis. Clin. Diagn. Lab. Immunol. 9, 257–266 (2002).
Cukalac, T. et al. Paired TCRαβ analysis of virus-specific CD8+ T cells exposes diversity in a previously defined ‘narrow’ repertoire. Immunol. Cell Biol. 93, 804–814 (2015).
Yang, Y. et al. Focused specificity of intestinal TH17 cells towards commensal bacterial antigens. Nature 510, 152–156 (2014).
Good, Z. et al. Proliferation tracing with single-cell mass cytometry optimizes generation of stem cell memory-like T cells. Nat. Biotechnol. 37, 259–266 (2019).
Brockstedt, D. G. et al. Listeria-based cancer vaccines that segregate immunogenicity from toxicity. Proc. Natl Acad. Sci. USA 101, 13832–13837 (2004).
Levine, L. S. et al. Single-cell analysis by mass cytometry reveals metabolic states of early-activated CD8+ T cells during the primary immune response. Immunity 54, 829–844 (2021).
Kelly, J. M. et al. Identification of conserved T cell receptor CDR3 residues contacting known exposed peptide side chains from a major histocompatibility complex class I-bound determinant. Eur. J. Immunol. 23, 3318–3326 (1993).
Hogquist, K. A. et al. T cell receptor antagonist peptides induce positive selection. Cell 76, 17–27 (1994).
Safley, S. A., Cluff, C. W., Marshall, N. E. & Ziegler, H. K. Role of listeriolysin-O (LLO) in the T lymphocyte response to infection with Listeria monocytogenes. Identification of T cell epitopes of LLO. J. Immunol. 146, 3604–3616 (1991).
Geginat, G., Schenk, S., Skoberne, M., Goebel, W. & Hof, H. A novel approach of direct ex vivo epitope mapping identifies dominant and subdominant CD4 and CD8 T cell epitopes from Listeria monocytogenes. J. Immunol. 166, 1877–1884 (2001).
Straub, A. et al. Recruitment of epitope-specific T cell clones with a low-avidity threshold supports efficacy against mutational escape upon re-infection. Immunity 56, 1269–1284 (2023).
Sockolosky, J. T. et al. Selective targeting of engineered T cells using orthogonal IL-2 cytokine-receptor complexes. Science 359, 1037–1042 (2018).
Hufford, M. M., Kim, T. S., Sun, J. & Braciale, T. J. The effector T cell response to influenza infection. Curr. Top Microbiol. Immunol. 386, 423–455 (2014).
Fehlings, M. et al. Multiplex peptide-MHC tetramer staining using mass cytometry for deep analysis of the influenza-specific T-cell response in mice. J. Immunol. Methods 453, 30–36 (2018).
Vitiello, A. et al. Immunodominance analysis of CTL responses to influenza PR8 virus reveals two new dominant and subdominant Kb-restricted epitopes. J. Immunol. 157, 5555–5562 (1996).
Belz, G. T., Xie, W., Altman, J. D. & Doherty, P. C. A previously unrecognized H-2Db-restricted peptide prominent in the primary influenza a virus-specific CD8+ T-cell response is much less apparent following secondary challenge. J. Virol. https://doi.org/10.1128/jvi.74.8.3486-3493.2000 (2000).
Arpaia, N. et al. A distinct function of regulatory T cells in tissue protection. Cell 162, 1078–1089 (2015).
Kok, L., Masopust, D. & Schumacher, T. N. The precursors of CD8+ tissue resident memory T cells: from lymphoid organs to infected tissues. Nat. Rev. Immunol. 22, 283–293 (2021).
Mercado, R. et al. Early programming of T cell populations responding to bacterial infection. J. Immunol. 165, 6833–6839 (2000).
McGill, J. & Legge, K. L. Cutting edge: contribution of lung-resident T cell proliferation to the overall magnitude of the antigen-specific CD8 T cell response in the lungs following murine influenza virus infection. J. Immunol. 183, 4177–4181 (2009).
Pai, J. A. & Satpathy, A. T. High-throughput and single-cell T cell receptor sequencing technologies. Nat. Methods 18, 881–892 (2021).
Sakaguchi, S., Yamaguchi, T., Nomura, T. & Ono, M. Regulatory T cells and immune tolerance. Cell 133, 775–787 (2008).
Rotrosen, E. & Kupper, T. S. Assessing the generation of tissue resident memory T cells by vaccines. Nat. Rev. Immunol. 23, 655–665 (2023).
Chang, J. T., Wherry, E. J. & Goldrath, A. W. Molecular regulation of effector and memory T cell differentiation. Nat. Immunol. 15, 1104–1115 (2014).
Montelongo-Jauregui, D., Vila, T., Sultan, A. S. & Jabra-Rizk, M. A. Convalescent serum therapy for COVID-19: a 19th century remedy for a 21st century disease. PLoS Pathog. 16, e1008735 (2020).
Lavoie, P. M., Dumont, A. R., McGrath, H., Kernaleguen, A.-E. & Sékaly, R.-P. Delayed expansion of a restricted T cell repertoire by low-density TCR ligands. Int. Immunol. 17, 931–941 (2005).
Liu, B. et al. Temporal single-cell tracing reveals clonal revival and expansion of precursor exhausted T cells during anti-PD-1 therapy in lung cancer. Nat. Cancer 3, 108–121 (2021).
Davis, M. M. & Boyd, S. D. Recent progress in the analysis of αβ T cell and B cell receptor repertoires. Curr. Opin. Immunol. 59, 109–114 (2019).
Sinnathamby, G. et al. Priming and activation of human ovarian and breast cancer-specific CD8+ T cells by polyvalent Listeria monocytogenes-based vaccines. J. Immunother. 32, 856–869 (2009).
Hartmann, F. J. et al. Scalable conjugation and characterization of immunoglobulins with stable mass isotope reporters for single-cell mass cytometry analysis. in Mass Cytometry: Methods and Protocols (eds McGuire, H. M. & Ashhurst, T. M.) 55–81 (Springer New York, 2019).
Zunder, E. R. et al. Palladium-based mass tag cell barcoding with a doublet-filtering scheme and single-cell deconvolution algorithm. Nat. Protoc. 10, 316–333 (2015).
Finck, R. et al. Normalization of mass cytometry data with bead standards. Cytometry A 83, 483–494 (2013).
Utsunomiya, Y. et al. Analysis of a monoclonal rat antibody directed to the alpha-chain variable region (V alpha 3) of the mouse T cell antigen receptor. J. Immunol. 143, 2602–2608 (1989).
Acknowledgements
We acknowledge the UCSF Parnassus Flow Cytometry CoLab (RRID: SCR_018206) for assisting in generating mass cytometry data and the UC Berkeley Cancer Research Laboratory Flow Cytometry Facility. We also thank D. Portnoy (University of California, Berkeley) for providing attenuated Listeria strains for this study. Research reported here was supported in part by grants from the NIH 1DP2CA247830-01, P30 DK063720, DP5 OD023056, R01 DE032033, DoD award BC220499, the Parker Institute for Cancer Immunotherapy, and the NIH S10 Instrumentation Grant S10 1S10OD018040-01. J.G.C. is an HHMI Gilliam Fellow. M.D. is a Pew-Stewart Scholar and a St. Baldrick’s Scholar with generous support from Hope with Hazel.
Author information
Authors and Affiliations
Contributions
J.G.C., R.D., M.H.S. and M.D. conceived, designed and directed the study. J.G.C., R.D., A.M., I.T. and D.M.M. performed experiments and generated data. R.D., A.S. and M.H.S generated computational methodology for single-cell data analysis. Writing, review and editing of primary manuscript were done by J.G.C., R.D., A.S., D.H.B., M.H.S. and M.D. Funding acquisition for study was done by M.H.S. and M.D.
Corresponding authors
Ethics declarations
Competing interests
R.D. is a shareholder in BioNTech. M.H.S. is founder and shareholder of Teiko.bio and Prox Biosciences, has received a speaking honorarium from Fluidigm, Arsenal and Kumquat, has been a paid consultant for Five Prime, Ono, January, Earli, Astellas and Indaptus, and has received research funding from Roche/Genentech, Pfizer, Valitor and Bristol Myers Squibb. The other authors declare no competing interests.
Peer review
Peer review information
Nature Immunology thanks Sean Bendall, Alexandre Harari and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Nick Bernard, in collaboration with the Nature Immunology team.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Extended data
Extended Data Fig. 1 Tracking a Listeria-specific CD8+ T cell response against LADD by TCR use and phenotyping with CyTOF.
(a) Gating strategy for all CyTOF analyses preceding input into semi-supervised Vα-Vβ assignment script. (b) Representative flow plots and quantification of roliferating CD8+ T cells from spleens 5 days after LADD, LADD-OVA, or PBS injection IV. P values (from top to bottom) 0.0286, 0.0159. (c) Bar plot of Vα2, Vβ14, and Vα2_Vβ14 percentages in each cluster for conditions LAAD and LAAD-OVA. (d) UMAP visualization of pooled CD8+ T cells colored by the expression of Vβ12+ in LADD, LADD-OVA, or PBS infected mice. Results from n=5 for PBS, n=4 for LADD, n=4 for LADD-OVA, P values were calculated by unpaired two-tailed student’s T-test, mean ± s.e.m.
Extended Data Fig. 2 Tracking Listeria-specific CD4+ T cells against LADD by TCR use and phenotyping with CyTOF.
(a) Representative flow plots and quantification of proliferating (Ki-67+) CD4+ T cells from spleen 5 days after LADD, LADD-OVA, or PBS injection. P values (from top to bottom) 0.0159, 0.0286. (b) UMAP visualization of CD4+ T conventional (non-Treg) cell clusters based on expression of non-TCR proteins. (c) Heatmap of non-TCR protein expression annotated by cluster and fraction of cells falling into each cluster. (d) UMAP visualization of CD4+ T cells colored by the expression Ki-67 in LADD-infected mice. (e) Frequency of Ki-67+ and Ki-67- CD4+ T cells using specific TCR Vβ (left) or Vα chains (right) in response to PBS, LADD, or LADD-OVA. (f-h) Frequency of Vβ13+ (f), Vβ14+ (g), or Vβ13+Vα2+ among Ki-67+(h) versus Ki-67- CD4+ T cells in LADD or LADD-OVA infected mice. P values (from left to right) 0.0286, 0.0079, 0.0286, 0.0159, 0.0159, 0.0286, 0.0079, 0.0159, 0.0286. (i-k) UMAP visualization of pooled CD4+ T cells colored by the expression of Vβ13+ (i), Vβ13+Vα2+ (j), and Vβ14 (k) in LADD, LADD-OVA, or PBS injected mice. (m) Bar plot of Vβ13+, Vβ13+Vα2+, and Vβ14+ percentages in Teff clusters. Results from n=5 for PBS, n=4 for LADD, n=4 for LADD-OVA, P values were calculated by unpaired two-tailed Student’s t-test, mean ± s.e.m.
Extended Data Fig. 3 Comparative analysis of Vβ and Vα frequencies and Vβ14+Vα2+ CD8 expansion captured by CyTOF versus TCR sequencing during LADD-OVA infection.
(a) Schematic of experimental approach to compare CyTOF to mRNA-based TCR-seq (iRepertoire) from spleens of LADD-OVA-infected mice. (b) Two-tailed spearman correlation analysis of Vβ and Vα frequencies in CD8+ T cells (left) and CD4+ T cells (right). (c-d) Comparative quantification of the TRBV/Vβ and TRAV/Vα chains captured by CyTOF of all CD8+ T (c) and CD4+ T cells (d). Note: Anti-Vα3.2 RR3- 16 clone binds specifically to a Vα3.2 allele preferentially expressed in CD8+ T cells over CD4+ T cells59. (e) Quantification of the TRBV/Vβ chain (top) and TRAV/Vα chain (bottom) frequencies in SIINFEKL-loaded H-2Kbtetramer+ CD8+ T cells sorted from spleens. (f) Fold change calculation of TRBV/Vβ chain and TRAV/Vα chain frequencies of SIINFEKL-loaded H-2Kb tetramer+ CD8+ T cells over frequencies of bulk CD8+ T cells. (g) Schematic comparison of approach used in this study compared to Straub et al.37. (h) Re-annotated clusters identified by Straub et al. plotted in UMAP space. (i) Scaled gene expression within the scRNA-seq dataset of genes encoding the proteins measured in Fig. 1, stratified by cluster. (j) UMAP visualization of all clustered cells colored by Ki67 or PD-1 gene expression. (k) UMAP visualization of all clustered cells colored by TRBV31 assignment or TRAV14D-2 assignment (left), and UMAP visualization of TRBV31 assigned cells colored by TRAV14D-2 assignment (right). (l) UMAP visualization of all clustered cells colored by identified specificity. (m) Quantification of TRBV gene usage among SIINFEKL-reactive cells (right) or of cells of unidentified reactivity (left). (n) Quantification of TRBV gene usage among SIINFEKLreactive cells expressing TRAV14D-2. (o) Quantification of TRBV/Vβ chain assignment of all CD8 T cells as measured by CyTOF or scRNA-seq. (p) Quantification of the TRBV/Vβ chains measured in the CyTOF assay of all CD8 T cells that were assigned a TRBV/Vβ chain by either CyTOF or scRNA-seq. (q) Two-tailed spearman correlation analysis of data shown in (j). Results from n=3 LADD-OVA infected mice for bulk TCRseq (a-f), data are presented as mean values ± s.e.m.
Extended Data Fig. 4 Expansion and activation of T cell subsets during influenza infection across multiple tissues.
(a) Gating strategy for all CyTOF analyses preceding input into semi-supervised Vα-Vβ assignment script. (b) Representative flow plots and quantification of proliferating (PD-1+Ki67+) CD8+ T cells. P values (from top to bottom) <0.0001, <0.0001, 0.0008. P values (from top to bottom) <0.0001, <0.0001, 0.0008. (c) Representative flow plots and quantification of proliferating (Ki67+) Foxp3- CD4+ Teff cells. P values (from top to bottom) <0.0001, <0.0001, 0.0008. P values (from top to bottom) <0.0001, <0.0001, 0.0004. (d) Representative flow plots and quantification of proliferating (Ki67+) Foxp3+ Tregs. P values (from top to bottom) <0.0001, <0.0001, 0.0008. P values (from top to bottom) <0.0001. Results from n=7 for PBS, n=7 for PR8, P values were calculated by unpaired two-tailed Student’s t-test, mean ± s.e.m.
Extended Data Fig. 5 Identification of influenza NP-specific Vβ14+ CD4+ T cells and Vβ9+ eTregs after influenza infection.
(a) UMAP visualization of CD4+ T conventional (non- Treg) cell clusters based on expression of non-TCR proteins. (b) Heatmap of non-TCR protein expression annotated by cluster and fraction of cells falling into each cluster. (c) UMAP visualization of CD4+ T cells colored by the expression Ki-67 in influenza-infected mice. (d) Frequencies of Vβ14+ CD4 T cells in non-proliferating (Ki-67-) vs. proliferating (Ki-67+) cells across indicated tissues. P values (from left to right) 0.0003, 0.0002, 0.0002. (e) UMAP visualization of pooled CD4+ T cells colored by Vβ14 expression. (f) Quantification of fold-enrichment in Vβ-chain usage by CD4+ T cells stimulated with the PR8 NP peptide (LILRGSVAHKSCLPACV). P value is <0.0001. (g) Schematic summarizing that Vβ14+CD4+ T cells are enriched to recognize PR8 NP antigen from influenza. (h) UMAP of Treg populations based on expression of non-TCR proteins. (i) Heatmap of non-TCR protein expression annotated by cluster and fraction of cells falling into each cluster. (j) UMAP visualization of Tregs colored by the expression Ki-67 in influenza-infected mice. (k) Frequencies of Vβ9+ Tregs in non-proliferating (Ki-67-) vs. proliferating (Ki-67+) cells across indicated tissues. P values (from left to right) 0.0207, 0.0002. (l) UMAP visualization of pooled Tregs from PBS or influenza-infected mice colored by Vβ9 expression. Results from n=7 for PBS, n=7 for PR8, P values were calculated by unpaired two-tailed Student’s t-test (d,k) or two-way ANOVA (f), mean ± s.e.m.
Extended Data Fig. 6 Influenza-specific conventional CD4+ T cell differentiation and Vβ9+ Treg expansion is altered by vaccination.
(a) UMAP visualization of CD4+ T conventional (non-Treg) cell clusters based on expression of non-TCR proteins. (b) Heatmap of non-TCR protein expression annotated by cluster and fraction of cells falling into each cluster. (c) UMAP visualization of CD4+ T cells colored by the expression Ki-67 from all cohorts of mice. (d) Frequencies of Vβ14+ CD4 T cells in nonproliferating (Ki-67-) vs. proliferating (Ki-67+) cells in lungs of primary, vaccinated, or rechallenged mice. P values (from left to right) 0.0387, 0.0037, 0.0145. (e) UMAP visualization of pooled CD4+ T cells from primary or rechallenged mice colored by Vβ14 expression. (f) UMAP of Treg populations based on the expression of non-TCR proteins. (g) Heatmap of non-TCR protein expression annotated by cluster and fraction of cells falling into each cluster. (h) UMAP visualization of Tregs colored by the expression Ki-67 from all cohorts of mice. (i) Frequencies of Vβ9+ Tregs in nonproliferating (Ki-67-) vs. proliferating (Ki-67+) cells in lungs of primary, vaccinated, or rechallenged mice. P values (from left to right) <0.0001, <0.0001, <0.0001. (j) UMAP visualization of pooled Tregs from primary or rechallenged mice colored by Vβ9 expression. Results from n=7 for PBS, n=7 for PR8, P values were calculated by oneway ANOVA, mean ± s.e.m.
Extended Data Fig. 7 Convalescent therapy has a limited effect on the CD4+ Teff and Treg repertoire during influenza infection.
(a) UMAP visualization of CD4+ T conventional (non-Treg) cell clusters based on expression of non-TCR proteins. (b) Heatmap of non-TCR protein expression annotated by cluster and fraction of cells falling into each cluster. (c) UMAP visualization of CD4+ T cells colored by the expression Ki-67 from all cohorts of mice. (d) Frequencies of Vβ14+ and Vβ10b+ CD4 T cells in non-proliferating (Ki-67-) vs. proliferating (Ki-67+) cells from the lungs of influenza infected mice treated with naive or convalescent serum at specified times after infection. (e) UMAP visualization of pooled CD4+ T cells from early vs. late convalescent serum treated mice colored by Vβ14 expression. (f) UMAP of Tregs based on the expression of non-TCR proteins. (g) Heatmap of non-TCR protein expression annotated by cluster and fraction of cells falling into each cluster. (h) UMAP visualization of Tregs colored by the expression Ki-67 from all cohorts of mice. (i) Frequencies of Vβ9+ Tregs in nonproliferating (Ki-67-) vs. proliferating (Ki-67+) cells from the lungs of infected mice treated with naive or convalescent serum at specified times after infection. P values (from top to bottom) 0.0299, 0.0087. (j) UMAP visualization of pooled Tregs from early vs. late convalescent serum treated mice colored by Vβ9 expression. Results from n=7 for PBS, n=14 for naïve serum-treated infected mice, n=7 for convalescent serum treated infected mice at 4 hours, n=7 for convalescent serum-treated infected mice at 4 days, P values were calculated by one-way ANOVA, mean ± s.e.m.
Supplementary information
Supplementary Information
Supplementary Figs. 1–3 and Tables 1 and 2.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Garcia Castillo, J., DeBarge, R., Mende, A. et al. A mass cytometry method pairing T cell receptor and differentiation state analysis. Nat Immunol 25, 1754–1763 (2024). https://doi.org/10.1038/s41590-024-01937-3
Received:
Accepted:
Published:
Issue Date:
DOI: https://doi.org/10.1038/s41590-024-01937-3