Abstract
The evolution of T cell molecular signatures in the distal lung of patients with severe pneumonia is understudied. Here, we analyzed T cell subsets in longitudinal bronchoalveolar lavage fluid samples from 273 patients with severe pneumonia, including unvaccinated patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or with respiratory failure not linked to pneumonia. In patients with SARS-CoV-2 pneumonia, activation of interferon signaling pathways, low activation of the NF-κB pathway and preferential targeting of spike and nucleocapsid proteins early after intubation were associated with favorable outcomes, whereas loss of interferon signaling, activation of NF-κB-driven programs and specificity for the ORF1ab complex late in disease were associated with mortality. These results suggest that in patients with severe SARS-CoV-2 pneumonia, alveolar T cell interferon responses targeting structural SARS-CoV-2 proteins characterize individuals who recover, whereas responses against nonstructural proteins and activation of NF-κB are associated with poor outcomes.
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
27,99 € / 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
Patient-specific clinical metadata are available in Supplementary Data 2. Deidentified clinical information from this cohort is available on AccessClinicalData@NIAID (https://accessclinicaldata.niaid.nih.gov/study-viewer/clinical_trials/SCRIPT). Fluorescence-activated cell sorting cell-type proportions are available in Supplementary Data 1. Bulk RNA-sequencing data, including raw counts tables for CD4+ T cells, CD8+ T cells and Treg cells, are available in Supplementary Data 3, 4 and 5, respectively. Bulk TCR-sequencing data, including MiXCR-processed raw sequencing files for CD4+ T cells and CD8+ T cells, are available in Supplementary Data 9 and 10, respectively. Raw data for both bulk RNA sequencing and TCR sequencing are available in the dbGaP repository for the SCRIPT project (phs002300.v1.p1).
This study used reference genome GRCh38 for RNA-sequencing experiment data processing (available from iGenomes reference, originally donwloaded from NCBI Homo sapiens Annotation Release 106 (https://s3.amazonaws.com/igenomes.illumina.com/Homo_sapiens/NCBI/GRCh38/Homo_sapiens_NCBI_GRCh38.tar.gz), single-cell object from Grant et al. for bulk RNA-sequencing deconvolution (GSE155249, GSE155249_main.h5ad file), MIRA (https://adaptivepublic.blob.core.windows.net/publishedproject-supplements/covid-2020/ImmuneCODE-MIRA-Release002.1.zip) and IEDB (http://tools.iedb.org/mhci/download/ and http://tools.iedb.org/mhcii/download/) databases for TCR binding prediction.
Code availability
All code used for processing, analysis and figure generation is available at https://github.com/NUPulmonary/2024_Markov_Ren_Senkow.
References
Budinger, G. R. S., Misharin, A. V., Ridge, K. M., Singer, B. D. & Wunderink, R. G. Distinctive features of severe SARS-CoV-2 pneumonia. J. Clin. Invest. 131, e149412 (2021).
Gao, C. A. et al. Machine learning links unresolving secondary pneumonia to mortality in patients with severe pneumonia, including COVID-19. J. Clin. Invest. 133, e170682 (2023).
Grant, R. A. et al. Circuits between infected macrophages and T cells in SARS-CoV-2 pneumonia. Nature 590, 635–641 (2021).
Rydyznski Moderbacher, C. et al. Antigen-specific adaptive immunity to SARS-CoV-2 in acute COVID-19 and associations with age and disease severity. Cell 183, 996–1012 (2020).
Markov, N. S. et al. SCRIPT CarpeDiem dataset: demographics, outcomes, and per-day clinical parameters for critically ill patients with suspected pneumonia (version 1.1.0). PhysioNet https://doi.org/10.13026/5phr-4r89 (2023).
Pickens, C. O. et al. Bacterial superinfection pneumonia in patients mechanically ventilated for COVID-19 pneumonia. Am. J. Respir. Crit. Care Med. 204, 921–932 (2021).
Grant, R. A. et al. Prolonged exposure to lung-derived cytokines is associated with activation of microglia in patients with COVID-19. JCI Insight 9, e178859 (2024).
Pickens, C. I. et al. An adjudication protocol for severe pneumonia. Open Forum Infect. Dis. 10, ofad336 (2023).
Recovery Collaborative Group et al. Dexamethasone in hospitalized patients with COVID-19. N. Engl. J. Med. 384, 693–704 (2021).
Wu, H., Harder, C. & Culley, C. The 2016 clinical practice guidelines for management of hospital-acquired and ventilator-associated pneumonia. Can. J. Hosp. Pharm. 70, 251–252 (2017).
Szabo, P. A. et al. Longitudinal profiling of respiratory and systemic immune responses reveals myeloid cell-driven lung inflammation in severe COVID-19. Immunity 54, 797–814 (2021).
Bergamaschi, L. et al. Longitudinal analysis reveals that delayed bystander CD8+ T cell activation and early immune pathology distinguish severe COVID-19 from mild disease. Immunity 54, 1257–1275 (2021).
Neidleman, J. et al. Distinctive features of SARS-CoV-2-specific T cells predict recovery from severe COVID-19. Cell Rep. 36, 109414 (2021).
Mathew, D. et al. Deep immune profiling of COVID-19 patients reveals distinct immunotypes with therapeutic implications. Science 369, eabc8511 (2020).
Jovisic, M., Mambetsariev, N., Singer, B. D. & Morales-Nebreda, L. Differential roles of regulatory T cells in acute respiratory infections. J. Clin. Invest. 133, e170505 (2023).
Zhao, J. et al. Airway memory CD4+ T cells mediate protective immunity against emerging respiratory coronaviruses. Immunity 44, 1379–1391 (2016).
Woodland, D. L. & Blackman, M. A. Immunity and age: living in the past? Trends Immunol. 27, 303–307 (2006).
Goronzy, J. J., Fang, F., Cavanagh, M. M., Qi, Q. & Weyand, C. M. Naive T cell maintenance and function in human aging. J. Immunol. 194, 4073–4080 (2015).
Goronzy, J. J. & Weyand, C. M. Successful and maladaptive T cell aging. Immunity 46, 364–378 (2017).
Huang, H., Wang, C., Rubelt, F., Scriba, T. J. & Davis, M. M. Analyzing the Mycobacterium tuberculosis immune response by T-cell receptor clustering with GLIPH2 and genome-wide antigen screening. Nat. Biotechnol. 38, 1194–1202 (2020).
Lineburg, K. E. et al. CD8+ T cells specific for an immunodominant SARS-CoV-2 nucleocapsid epitope cross-react with selective seasonal coronaviruses. Immunity 54, 1055–1065 (2021).
Pogorelyy, M. V. et al. Resolving SARS-CoV-2 CD4+ T cell specificity via reverse epitope discovery. Cell Rep. Med. 3, 100697 (2022).
Nolan, S. et al. A large-scale database of T-cell receptor β (TCRβ) sequences and binding associations from natural and synthetic exposure to SARS-CoV-2. Preprint at Res. Sq. https://doi.org/10.21203/rs.3.rs-51964/v1 (2020).
Moutaftsi, M. et al. Uncovering the interplay between CD8, CD4 and antibody responses to complex pathogens. Future Microbiol. 5, 221–239 (2010).
Orenbuch, R. et al. arcasHLA: high-resolution HLA typing from RNAseq. Bioinformatics 36, 33–40 (2020).
Reynisson, B., Alvarez, B., Paul, S., Peters, B. & Nielsen, M. NetMHCpan-4.1 and NetMHCIIpan-4.0: improved predictions of MHC antigen presentation by concurrent motif deconvolution and integration of MS MHC eluted ligand data. Nucleic Acids Res. 48, W449–W454 (2020).
Gonzalez-Galarza, F. F. et al. Allele frequency net database (AFND) 2020 update: gold-standard data classification, open access genotype data and new query tools. Nucleic Acids Res. 48, D783–D788 (2020).
Chi, H., Pepper, M. & Thomas, P. G. Principles and therapeutic applications of adaptive immunity. Cell 187, 2052–2078 (2024).
Schultheiss, C. et al. Next-generation sequencing of T and B cell receptor repertoires from COVID-19 patients showed signatures associated with severity of disease. Immunity 53, 442–455 (2020).
Nguyen, T. H. O. et al. CD8+ T cells specific for an immunodominant SARS-CoV-2 nucleocapsid epitope display high naive precursor frequency and TCR promiscuity. Immunity 54, 1066–1082 (2021).
Le Bert, N. et al. SARS-CoV-2-specific T cell immunity in cases of COVID-19 and SARS, and uninfected controls. Nature 584, 457–462 (2020).
Grifoni, A. et al. Targets of T cell responses to SARS-CoV-2 coronavirus in humans with COVID-19 disease and unexposed individuals. Cell 181, 1489–1501 (2020).
Sekine, T. et al. Robust T cell immunity in convalescent individuals with asymptomatic or mild COVID-19. Cell 183, 158–168 (2020).
Bacher, P. et al. Low-avidity CD4+ T cell responses to SARS-CoV-2 in unexposed individuals and humans with severe COVID-19. Immunity 53, 1258–1271 (2020).
Dykema, A. G. et al. Functional characterization of CD4+ T cell receptors crossreactive for SARS-CoV-2 and endemic coronaviruses. J. Clin. Invest. 131, e146922 (2021).
Mateus, J. et al. Selective and cross-reactive SARS-CoV-2 T cell epitopes in unexposed humans. Science 370, 89–94 (2020).
Schulien, I. et al. Characterization of pre-existing and induced SARS-CoV-2-specific CD8+ T cells. Nat. Med. 27, 78–85 (2021).
Channappanavar, R. et al. Dysregulated type I interferon and inflammatory monocyte–macrophage responses cause lethal pneumonia in SARS-CoV-infected mice. Cell Host Microbe 19, 181–193 (2016).
Zhao, J., Zhao, J. & Perlman, S. T cell responses are required for protection from clinical disease and for virus clearance in severe acute respiratory syndrome coronavirus-infected mice. J. Virol. 84, 9318–9325 (2010).
Hadjadj, J. et al. Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients. Science 369, 718–724 (2020).
Tan, A. T. et al. Early induction of functional SARS-CoV-2-specific T cells associates with rapid viral clearance and mild disease in COVID-19 patients. Cell Rep. 34, 108728 (2021).
Swadling, L. et al. Pre-existing polymerase-specific T cells expand in abortive seronegative SARS-CoV-2. Nature 601, 110–117 (2022).
Ng, K. W. et al. Preexisting and de novo humoral immunity to SARS-CoV-2 in humans. Science 370, 1339–1343 (2020).
Braun, J. et al. SARS-CoV-2-reactive T cells in healthy donors and patients with COVID-19. Nature 587, 270–274 (2020).
Starren, J. B., Winter, A. Q. & Lloyd-Jones, D. M. Enabling a learning health system through a unified enterprise data warehouse: the experience of the Northwestern University Clinical and Translational Sciences (NUCATS) Institute. Clin. Transl. Sci. 8, 269–271 (2015).
Walter, J. M., Helmin, K. A., Abdala-Valencia, H., Wunderink, R. G. & Singer, B. D. Multidimensional assessment of alveolar T cells in critically ill patients. JCI Insight 3, e123287 (2018).
Qadir, N. et al. An update on management of adult patients with acute respiratory distress syndrome: an official American Thoracic Society Clinical Practice Guideline. Am. J. Respir. Crit. Care Med. 209, 24–36 (2024).
Beigel, J. H. et al. Remdesivir for the treatment of COVID-19—final report. N. Engl. J. Med. 383, 1813–1826 (2020).
Ewels, P. A. et al. The nf-core framework for community-curated bioinformatics pipelines. Nat. Biotechnol. 38, 276–278 (2020).
Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).
Patro, R., Duggal, G., Love, M. I., Irizarry, R. A. & Kingsford, C. Salmon provides fast and bias-aware quantification of transcript expression. Nat. Methods 14, 417–419 (2017).
Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).
Gu, Z., Eils, R. & Schlesner, M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics 32, 2847–2849 (2016).
Rahnenfuhrer, A. A. topGO: enrichment analysis for Gene Ontology. https://bioconductor.org/packages/release/bioc/vignettes/topGO/inst/doc/topGO.pdf (2023).
Durinck, S., Spellman, P. T., Birney, E. & Huber, W. Mapping identifiers for the integration of genomic datasets with the R/Bioconductor package biomaRt. Nat. Protoc. 4, 1184–1191 (2009).
Sayols, S. rrvgo: a bioconductor package for interpreting lists of Gene Ontology terms. MicroPubl. Biol. 2023, 000811 (2023).
Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).
Korotkevich, G., Sukhov, V. & Sergushichev, A. Fast gene set enrichment analysis. Preprint at bioRxiv https://doi.org/10.1101/060012 (2019).
Aliee, H. & Theis, F. J. AutoGeneS: automatic gene selection using multi-objective optimization for RNA-seq deconvolution. Cell Syst. 12, 706–715 (2021).
Bolotin, D. A. et al. MiXCR: software for comprehensive adaptive immunity profiling. Nat. Methods 12, 380–381 (2015).
ImmunoMind Team. immunarch: an R package for painless bioinformatics analysis of T-cell and B-cell immune repertoires. Zenodo https://doi.org/10.5281/zenodo.3367200 (2019).
Butts, C. network: classes for relational data. The Statnet Project. R package version 1.13.0.1 https://CRAN.R-project.org/package=network (2015).
Pedersen, T. tidygraph: a tidy API for graph manipulation. https://tidygraph.data-imaginist.com/ (2023).
Virtanen, P. et al. SciPy 1.0: fundamental algorithms for scientific computing in Python. Nat. Methods 17, 261–272 (2020).
Dash, P. et al. Quantifiable predictive features define epitope-specific T cell receptor repertoires. Nature 547, 89–93 (2017).
Constantin, A. & Patil, I. ggsignif: R package for displaying significance brackets for ‘ggplot2’. Preprint at PsyArxiv https://doi.org/10.31234/osf.io/7awm6 (2021).
Kassambara, A. ggpubr: ‘ggplot2’ based publication ready plots. https://rpkgs.datanovia.com/ggpubr/ (2023).
Wickham, H. ggplot2: Elegant Graphics for Data Analysis (Springer-Verlag: 2016). .
Acknowledgements
We thank the Robert H. Lurie Comprehensive Cancer Center of Northwestern University in Chicago for the use of the Flow Cytometry Core Facility. The Lurie Cancer Center is supported, in part, by an NCI Cancer Center Support Grant P30 CA060553. This research was supported, in part, through the computational resources and staff contributions provided for the Quest High-Performance Computing Facility at Northwestern University, which is jointly supported by the Office of the Provost, the Office for Research and Northwestern University Information Technology. We acknowledge the support of The Simpson Querrey Lung Institute for Translational Sciences at Northwestern University, the Dixon Translational Research Grants Initiative at Northwestern Medicine and the Northwestern University Clinical and Translational Sciences Institute (UL1TR001422). This work was also supported by the Chicago Biomedical Consortium with support from the Searle Funds at The Chicago Community Trust. N.S.M. was supported by AHA 24PRE1196998. C.A.G. was supported by T32HL076139 and F32HL162377. R.A.G. was funded by NIH grants T32AG020506 and F31AG071225 and Schmidt Science Fellows. A.V.M. was supported by NIH grants U19AI135964, U19AI181102, P01AG049665, P01HL154998, R01HL153312, R01HL158139, R01ES034350 and R21AG075423. R.G.W. was supported by NIH grants U19AI135964, U01TR003528, P01HL154998, R01HL149883 and R01LM013337. G.R.S.B. was supported by a Chicago Biomedical Consortium grant, Northwestern University Dixon Translational Science Award and Simpson Querrey Lung Institute for Translational Science and NIH grants AG049665, HL154998, HL14575, HL158139, HL147290, AG075423 and AI135964 and The Veterans Administration award I01CX001777. B.D.S. was supported by NIH awards R01HL149883, R01HL153122, P01HL154998, P01AG049665, U19AI135964 and U19AI181102. L.M.-N. was supported by the Parker B. Francis Opportunity Award and NIH awards K08HL15935 and U19AI135964.
Author information
Authors and Affiliations
Consortia
Contributions
Conception: A.V.M., R.G.W., G.R.S.B., B.D.S. and L.M.-N. Design: N.S.M., A.V.M., R.G.W., G.R.S.B., B.D.S. and L.M.-N. Implementation: N.S.M., Z.R., K.J.S., R.A.G., C.A.G., E.S.M., L.S., H.K., K.A.H., M.J., J.M.A., X.G.P.-L., H.A.-V., S.S., J.N., M.K., E.A.O., R.L.-R., J.F.H., L.M.S., E.R.-G., A.V.M., R.G.W., B.D.S. and L.M.-N. Data analysis/interpretation: N.S.M., Z.R., K.J.S., E.S.M., A.V.M., R.G.W., G.R.S.B., B.D.S. and L.M.-N. Drafting the article: Z.R., K.J.S., B.D.S. and L.M.-N. Revising/reviewing the article: N.S.M., G.R.S.B., B.D.S. and L.M.-N. Public responsibility: L.R., R.G.W., B.D.S. and L.M.-N.
Corresponding author
Ethics declarations
Competing interests
B.D.S. holds US patent number US 10,905,706 B2, ‘Compositions and methods to accelerate resolution of acute lung inflammation’, and serves on the Scientific Advisory Board of Zoe Biosciences, outside of the submitted work. The other authors have no competing interests to declare.
Peer review
Peer review information
Nature Immunology thanks Sophie Valkenburg and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Ioana Staicu, 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 Graphical abstract and the study cohort.
(a) Graphical abstract. (b) CONSORT diagram of patients included in this study. (c) Schematic depicting multi-step analysis of BAL fluid samples with flow cytometry, bulk RNA-sequencing, and bulk TCR-sequencing by diagnosis (NPC, OP, SARS-CoV-2-P, OVP) and T cell subset (CD4+, CD8+, Treg).
Extended Data Fig. 2 SARS-CoV-2-P is characterized by a lymphomonocytic alveolar infiltrate early following intubation.
(a) Flow cytometry analysis of BAL immune cell subset composition in CD3ε+CD4+ T cells, CD3ε+CD8+ T cells, Treg cells (CD3ε+CD4+CD25+CD127−), monocytes (HLA-DR+CD4+CD206−), macrophages (CD206+), and neutrophils (CD15+) indicating whether a given patient was discharged or deceased, the duration of mechanical ventilation (range 0–110 days; blanks indicate chronically ventilated patients), and presence (superinfection) or absence of bacterial infection with columns ordered by diagnosis (n = 432 samples (NPC [n = 36], OP [n = 187], SARS-CoV-2-P [n = 165], and OVP [n = 44]) and then by days from intubation when the sample was obtained. VAP (ventilator-associated pneumonia) designates samples from NPC or patients with SARS-CoV-2-P or OVP who cleared the virus and then developed bacterial pneumonia. Each column represents a BAL sample. (b) Box plot of percentage of BAL CD4+ T cells detected in BAL fluid samples from diagnosis groups (q < 0.05, pairwise Wilcoxon rank-sum tests with FDR correction). (c) As in b, for CD8+ T cells. (d) As in b, for Treg cells. (e) As in b, for monocytes. (f) As in b, for macrophages. (g) As in b, for neutrophils. (h) Box plot of BAL CD4+ T cell percentages between early (≤48 hours following intubation) and late (>48 hours following intubation) samples by diagnosis (q < 0.05, pairwise Wilcoxon rank-sum tests with FDR correction). (i) As in h, for CD8+ T cells. (j) As in h, for Treg cells. (k) Box plot of BAL CD3+ T cell percentage grouped by the presence or absence of bacterial superinfection in early (≤48 hours following intubation) and late (>48 hours following intubation) SARS-CoV-2-P samples (q < 0.05, pairwise Wilcoxon rank-sum tests with FDR correction). (l) As in i, for monocytes. (m) Spearman correlation analysis between the percentage of BAL monocytes and the duration of mechanical ventilation by diagnosis. (n) As in i, for neutrophils. (o) As in m, for neutrophils. (p) As in k, for neutrophils. (q) As in i, for macrophages. (r) As in m, for macrophages.
Extended Data Fig. 3 Persistent BAL T cell enrichment is associated with discharged status.
(a) Box plot of BAL CD4+ T cell percentage by discharged versus deceased status. n = 432 samples (NPC [n = 36], OP [n = 187], SARS-CoV-2-P [n = 165], and OVP [n = 44]). q < 0.05, pairwise Wilcoxon rank-sum tests with FDR correction. (b) As in a, for CD8+ T cells. (c) As in a, for Treg cells. (d) As in a, for macrophages. (e) As in a, for monocytes. (f) As in a, grouped by discharged and deceased status and by early (≤48 hours following intubation) and late (>48 hours following intubation) timing of BAL sampling relative to intubation. (g) As in f, for CD8+ T cells. (h) As in f, for Treg cells. (i) As in f, for monocytes. (j) As in f, for macrophages. (k) As in a, for neutrophils. (l) As in f, for neutrophils.
Extended Data Fig. 4 BAL immune cell subset abundance and phenotype correlate with clinical features.
(a) Correlation analysis between the percentage of BAL immune cell subsets and clinical, physiological, and laboratory variables in NPC. No significant values after calculating Spearman rank correlation coefficient with FDR correction. (b) As in a, for OP. (c) As in a, for OVP. (d) Proportion of SARS-CoV-2-P BAL fluid samples, comparing presence or absence of bacterial superinfection with discharged versus deceased status. Nonsignificant by Fisher exact test. (e) Proportion of SARS-CoV-2-P BAL fluid samples, comparing pneumonia episode outcome status (cured, indeterminate, not cured) with discharged versus deceased status (q < 0.05, Fisher exact test with FDR correction). (f) Correlation analysis between BAL CD4+ and CD8+ T cell surface expression of CD127 and HLA-DR and clinical, laboratory, and physiological variables in SARS-CoV-2-P samples. Spearman rank correlation coefficient with FDR correction (q < 0.05 [*]). (g) Left: Correlation plot of CD4+ T cell surface expression of CD127 against the duration of mechanical ventilation. Spearman rank correlation coefficient shown. Shaded area represents 95% CI. Second from left: As in left, for CD4+ T cell surface expression of HLA-DR. Second from right: As in left, for CD8+ T cell surface expression of CD127. Right: As in left, for CD8+ T cell surface expression of HLA-DR.
Extended Data Fig. 5 BAL T cells in SARS-CoV-2-P are transcriptionally enriched for immune processes compared with NPC, OP, and OVP.
(a) Number of patients contributing BAL samples for RNA-seq analysis (n = 336 samples; NPC [n = 38], OP [n = 51], SARS-CoV-2-P [n = 200], and OVP [n = 47]). (b) Proportion of samples grouped by T cell subset and diagnosis. (c) Number of samples grouped by early (≤48 hours following intubation) and late (>48 hours following intubation) timing of BAL sampling relative to intubation or obtained in patients receiving chronic ventilation. (d) Box plot of mean Sequential Organ Failure Assessment (SOFA) scores over the day of the BAL. Nonsignificant after pairwise Wilcoxon rank-sum tests with FDR correction). (e) Proportion of discharged versus deceased patients. Nonsignificant after pairwise χ2 tests for homogeneity of proportions with FDR correction. (f) Proportion of sex (pairwise χ2 tests for homogeneity of proportions with FDR correction). (g) Top: K-means clustering of 975 differentially expressed genes (q < 0.05, likelihood-ratio test with FDR correction) in CD8+ T cell samples. Columns represent unique samples and column headers are color-coded by diagnosis, discharged versus deceased status, duration of mechanical ventilation (range 0–90 days, blanks indicate chronically ventilated patients), and superinfection status with columns ordered by NPC (n = 15), OP (n = 15), SARS-CoV-2-P (n = 72), and OVP (n = 23) and then by days from intubation when the sample was obtained. VAP (ventilator-associated pneumonia) designates samples from NPC or patients with SARS-CoV-2-P or OVP who cleared the virus and then developed bacterial pneumonia. Representative genes are shown for each cluster. Bottom: As in top, for CD4+ T cell samples (866 differentially expressed genes) in NPC (n = 14), OP (n = 23), SARS-CoV-2-P (n = 81), and OVP (n = 20). (h) Top: GSEA of Hallmark gene sets for the pairwise comparison between SARS-CoV-2-P samples and OVP samples in CD8+ T cells from g, top. Count denotes pathway size after removing genes not detected in the expression dataset. Enrichment denotes significant (q < 0.25 with FDR correction) upregulated (red) and downregulated (blue) pathways by normalized enrichment score. Bottom: As in h, top for CD4+ T cells from g, bottom. (i) Left: Gene ontology (GO) parent term annotation (q < 0.05 with FDR correction) from cluster 2_CD8. Points are color-coded by unique terms and size denotes the number of genes within each GO term. Right: As in left, for cluster 1_CD8. (j) As in I for cluster 1_CD4.
Extended Data Fig. 6 BAL T cells have an activated memory phenotype during severe pneumonia.
(a) K-means clustering of 80 differentially expressed genes (q < 0.05, likelihood-ratio test with FDR correction) between NPC (n = 9), OP (n = 13), SARS-CoV-2-P (n = 47), and OVP (n = 4) in Treg cell samples. Columns represent unique samples and column headers are color-coded by diagnosis, discharged versus deceased status, duration of mechanical ventilation (range 0–90, blanks indicate chronically ventilated patients), and superinfection status. VAP (ventilator-associated pneumonia) designates samples from NPC or patients with SARS-CoV-2-P or OVP who cleared the virus and then developed bacterial pneumonia. Samples were clustered using Euclidean distance and Ward’s minimum variance linkage method. Representative genes are shown for each cluster. (b) GSEA of Hallmark gene sets for the pairwise comparison between SARS-CoV-2-P samples and NPC, OP, and OVP samples in Treg samples from a. Count denotes pathway size after removing genes not detected in the expression dataset. Enrichment denotes significant (q < 0.25 with FDR correction) upregulated (red) and downregulated (blue) pathways by normalized enrichment score. (c) As in b, for the pairwise comparison between SARS-CoV-2-P samples and OVP samples. (d) Deconvolution analysis showing inferred proportion of BAL CD8+ T cell subsets. Column data labels are as in a. (e) Proportion of inferred BAL CD8+ T cell subsets by diagnosis. (q < 0.05, pairwise Wilcoxon rank-sum tests with FDR correction). (f) Proportion of inferred BAL CD8+ T cell subsets by discharged versus deceased status and early (≤48 hours following intubation) and late (>48 hours following intubation) timing of BAL sampling relative to intubation in SARS-CoV-2-P samples (q < 0.05, pairwise Wilcoxon rank-sum tests with FDR correction). (g) As in d, for CD4+ T cell subsets. (h) As in e, for CD4+ T cell subsets. (i) As in f, for CD4+ T cell subsets.
Extended Data Fig. 7 TCR repertoire diversity is lower in SARS-CoV-2-P samples complicated by superinfection or VAP.
(a) Number of patients contributing BAL samples for TCR-seq analysis (n = 130 samples; NPC [n = 13], OP [n = 16], SARS-CoV-2-P [n = 73], and OVP [n = 28]). (b) Proportion of TCR-seq samples grouped by T cell subset and diagnosis. (c) Number of TCR-seq samples grouped by early (≤48 hours following intubation) and late (>48 hours following intubation) timing of BAL sampling relative to intubation. (d) Box plot of the mean Sequential Organ Failure Assessment (SOFA) scores on the day of BAL sample. Nonsignificant after pairwise Wilcoxon rank-sum tests with FDR correction. (e) Proportion of discharged versus deceased status patients. Nonsignificant after pairwise χ2 tests for homogeneity of proportions with FDR correction. (f) Proportion of sex (pairwise χ2 tests for homogeneity of proportions with FDR correction. (g) TCR richness analysis showing Chao 1 values in combined BAL CD4+ and CD8+ T cells by diagnosis. (q < 0.05, pairwise Wilcoxon rank-sum tests with FDR correction). (h) As in g, grouped by early (≤48 hours following intubation) and late (>48 hours following intubation) timing of BAL sampling relative to intubation. (i) As in g, grouped by discharged versus deceased status. (j) Pearson correlation analysis between TCR richness (Chao 1 values) and patient age in combined BAL CD4+ and CD8+ T cells in SARS-CoV-2-P samples (left) and NPC, OP, and OVP samples (right). Shaded area represents 95% CI. (k) As in j, correlated with the duration of mechanical ventilation. (l) Richness analysis showing Chao 1 values in SARS-CoV-2-P samples grouped by discharged versus deceased status in primary SARS-CoV-2 infection only (top), bacterial superinfection (middle), and VAP (bottom). Wilcoxon rank sum tests.
Extended Data Fig. 8 BAL T cells in SARS-CoV-2-P are virus specific in HLA context.
(a) Proportion of HLA-A molecules identified in CD8+ T cells in SARS-CoV-2-P samples (n of patients = 14). (b) As in a, for HLA-B. (c) Left: Proportion of TCRβ (V) gene usage in CD8+ TCR sequences in SARS-CoV-2-P samples (n of patients = 14). Middle: Absolute number of TCRβ sequences for each V region. Right: Representative epitopes from TCR chains exhibiting a count > 5 per dominant gene (TRBV12-3 and TRBV27). (d) As in c, for CD4+ TCR sequences. Right: Dominant genes (TCR count > 30) and representative epitopes are annotated for TRBV6-6 and TRBV20-1. (e) Proportion of HLA-A allele representation for detected CD8+ TCRβ (V) segments. (f) As in e, for HLA-B. (g) Heatmap of association measure of HLA alleles distribution within distinct V genes (1 – normalized Shannon entropy). (h) Proportion of TCRs detected by patient with distinct V gene segments.
Extended Data Fig. 9 Generation probabilities (Pgen) of BAL CD4+ TCRs is lower in SARS-CoV-2-P than in OVP, OP, and NPC.
(a) Logarithmic distribution of Pgen for post-GLIPH2-enriched CDR3β amino acid sequences across diagnoses (q < 0.05, pairwise Wilcoxon rank-sum tests with FDR correction). Each dot corresponds to a TCRβ chain. (b) Probability density of the data in a. (c) Logarithmic distribution of Pgen for post-GLIPH2-enriched and cross-referenced CDR3β amino acid sequences to MIRA MHCII dataset in SARS-CoV-2-P and OP. Each dot corresponds to a TCRβ chain. Nonsignificant after Wilcoxon rank-sum test. (d) Probability density of the data in c. (e) Similarity network analysis of BAL CD4+ TCR sequences cross-referenced to MIRA MHCII dataset epitope pools. Nodes represent unique TCR (CDR3β) sequences and are color-coded by diagnosis status (non-SARS-CoV-2-P [NPC, OP, and OVP], SARS-CoV-2, or shared). Edges connect TCR sequences belonging to the same patterns or specificity groups identified through the GLIPH2 algorithm. Dot size represents calculated generation probability (Pgen) of individual TCRβ sequences. Left: Representative TCR sequences from prominent clusters are annotated.
Supplementary information
Supplementary Information
Supplementary Tables 1–3.
Supplementary Data 1
Fluorescence-activated cell sorting cell-type proportions in BAL samples.
Supplementary Data 2
Anonymized metadata by patient and samples for bulk RNA sequencing.
Supplementary Data 3
Raw counts for CD4+ T cell bulk RNA-sequencing samples.
Supplementary Data 4
Raw counts for CD8+ T cell bulk RNA-sequencing samples.
Supplementary Data 5
Raw counts for Treg cell bulk RNA-sequencing samples.
Supplementary Data 6
a, Differentially expressed genes in CD8+ T cell bulk RNA-sequencing samples from k-means clustering analysis (cluster 1_CD8). b, Differentially expressed genes in CD8+ T cell bulk RNA-sequencing samples from k-means clustering analysis (cluster 2_CD8). c, Differentially expressed genes in CD4+ T cell bulk RNA-sequencing samples from k-means clustering analysis (cluster 1_CD4). d, Differentially expressed genes in CD4+ T cell bulk RNA-sequencing samples from k-means clustering analysis (cluster 2_CD4).
Supplementary Data 7
a, Differential expression analysis in CD8+ T cells of pairwise comparisons between SARS-CoV-2-P samples and combined NPC, OP and OVP samples. b, Differential expression analysis in CD4+ T cells of pairwise comparisons between SARS-CoV-2-P samples and combined NPC, OP and OVP samples. c, Differential expression analysis in CD8+ T cells of pairwise comparisons between SARS-CoV-2-P samples and OVP samples. d, Differential expression analysis in CD4+ T cells of pairwise comparisons between SARS-CoV-2-P samples and OVP samples. e, Differential expression analysis in Treg cells of pairwise comparisons between SARS-CoV-2-P samples and combined NPC, OP and OVP samples. f, Differential expression analysis in Treg cells of pairwise comparisons between SARS-CoV-2-P samples and OVP samples. g, Differential expression analysis in CD4+ T cells of pairwise comparisons between early (≤48 h after intubation) and late (>48 h after intubation) SARS-CoV-2-P samples. h, Differential expression analysis in CD8+ T cells of pairwise comparisons between early (≤48 h after intubation) and late (>48 h after intubation) SARS-CoV-2-P samples.
Supplementary Data 8
a, Significant GO processes identified in CD8+ T cell bulk RNA-sequencing samples from k-means clustering analysis (cluster 1_CD8). b, Significant GO processes identified in CD8+ T cell bulk RNA-sequencing samples from k-means clustering analysis (cluster 2_CD8). c, Significant GO parent terms and terms identified in CD8+ T cell bulk RNA-sequencing samples from k-means clustering analysis (cluster 1_CD8). d, Significant GO parent terms and terms identified in CD8+ T cell bulk RNA-sequencing samples from k-means clustering analysis (cluster 2_CD8). e, Significant GO processes identified in CD4+ T cell bulk RNA-sequencing samples from k-means clustering analysis (cluster 1_CD4). f, Significant GO parent terms and terms identified in CD4+ T cell bulk RNA-sequencing samples from k-means clustering analysis (cluster 1_CD4).
Supplementary Data 9
MiXCR-processed raw sequencing files with TCR repertoire data for CD4+ T cell bulk TCR-sequencing samples.
Supplementary Data 10
MiXCR-processed raw sequencing files with TCR repertoire data for CD8+ T cell bulk TCR-sequencing samples.
Supplementary Data 11
a, Anonymized metadata by patient and samples for bulk TCR sequencing. b, GLIPH2 analysis establishing specificity groups from CD4+ T cell bulk TCR-sequencing samples. c, GLIPH2 analysis establishing specificity groups from CD8+ T cell bulk TCR-sequencing samples.
Supplementary Data 12
a, Cross-reference of CD4+ T cell repertoire sequences with the MIRA dataset. b, Cross-reference of CD8+ T cell repertoire sequences with the MIRA dataset. c, MIRA MHC class I and MHC class II peptide pools identified in a patient cohort. d, Hierarchical distribution of MHC class I immunodominant epitopes by patient from the bulk TCR-sequencing subset. e, Pairwise similarity and average conservation score estimation between SARS-CoV-2 and HCoV epitopes.
Supplementary Data 13
a, Distribution of Pgen values for CD8+ T cells metadata. b, Distribution of Pgen values for CD4+ T cells metadata. c, Similarity network analysis of CD8+ T cells metadata. d, Similarity network analysis of CD4+ T cells metadata.
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
Markov, N.S., Ren, Z., Senkow, K.J. et al. Distinctive evolution of alveolar T cell responses is associated with clinical outcomes in unvaccinated patients with SARS-CoV-2 pneumonia. Nat Immunol 25, 1607–1622 (2024). https://doi.org/10.1038/s41590-024-01914-w
Received:
Accepted:
Published:
Issue Date:
DOI: https://doi.org/10.1038/s41590-024-01914-w