Abstract
Viral variant and host vaccination status impact infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), yet how these factors shift cellular responses in the human nasal mucosa remains uncharacterized. We performed single-cell RNA sequencing (scRNA-seq) on nasopharyngeal swabs from vaccinated and unvaccinated adults with acute Delta and Omicron SARS-CoV-2 infections and integrated with data from acute infections with ancestral SARS-CoV-2. Patients with Delta and Omicron exhibited greater similarity in nasal cell composition driven by myeloid, T cell and SARS-CoV-2hi cell subsets, which was distinct from that of ancestral cases. Delta-infected samples had a marked increase in viral RNA, and a subset of PER2+EGR1+GDF15+ epithelial cells was enriched in SARS-CoV-2 RNA+ cells in all variants. Prior vaccination was associated with increased frequency and activation of nasal macrophages. Expression of interferon-stimulated genes negatively correlated with coronavirus disease 2019 (COVID-19) severity in patients with ancestral and Delta but not Omicron variants. Our study defines nasal cell responses and signatures of disease severity across SARS-CoV-2 variants and vaccination.
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
scRNA-seq data are publicly available for download and visualization via the Broad Institute Single Cell Portal (accession code SCP2593). Custom reference FASTA and GTF for SARS-CoV-2 are available for download: https://github.com/ShalekLab/SARSCoV2-genome-reference.
Code availability
Code was written using R version 4.2.3 and publicly available packages. No new algorithms or functions were created, and code used in-built functions in listed packages available on CRAN. All code generated and used to analyze the data reported in this paper is available on GitHub in the jo-m-lab repository: https://github.com/jo-m-lab/SARSCoV2-Variants-Vaccines-Paper.
References
Gallo, O., Locatello, L. G., Mazzoni, A., Novelli, L. & Annunziato, F. The central role of the nasal microenvironment in the transmission, modulation, and clinical progression of SARS-CoV-2 infection. Mucosal Immunol. 14, 305–316 (2021).
Sungnak, W. et al. SARS-CoV-2 entry factors are highly expressed in nasal epithelial cells together with innate immune genes. Nat. Med. 26, 681–687 (2020).
Ahn, J. H. et al. Nasal ciliated cells are primary targets for SARS-CoV-2 replication in the early stage of COVID-19. J. Clin. Invest. 131, e148517 (2021).
Mlcochova, P. et al. SARS-CoV-2 B.1.617.2 Delta variant replication and immune evasion. Nature 599, 114–119 (2021).
Puhach, O., Meyer, B. & Eckerle, I. SARS-CoV-2 viral load and shedding kinetics. Nat. Rev. Microbiol. 21, 147–161 (2023).
Shuai, H. et al. Attenuated replication and pathogenicity of SARS-CoV-2 B.1.1.529 Omicron. Nature 603, 693–699 (2022).
Suzuki, R. et al. Attenuated fusogenicity and pathogenicity of SARS-CoV-2 Omicron variant. Nature 603, 700–705 (2022).
Willett, B. J. et al. SARS-CoV-2 Omicron is an immune escape variant with an altered cell entry pathway. Nat. Microbiol. 7, 1161–1179 (2022).
Carabelli, A. M. et al. SARS-CoV-2 variant biology: immune escape, transmission and fitness. Nat. Rev. Microbiol. 21, 162–177 (2023).
Minkoff, J. M. & tenOever, B. Innate immune evasion strategies of SARS-CoV-2. Nat. Rev. Microbiol. 21, 178–194 (2023).
Sette, A. & Crotty, S. Immunological memory to SARS-CoV-2 infection and COVID-19 vaccines. Immunol. Rev. 310, 27–46 (2022).
Ssemaganda, A. et al. Expansion of cytotoxic tissue-resident CD8+ T cells and CCR6+CD161+ CD4+ T cells in the nasal mucosa following mRNA COVID-19 vaccination. Nat. Commun. 13, 3357 (2022).
Puhach, O. et al. SARS-CoV-2 convalescence and hybrid immunity elicits mucosal immune responses. EBioMedicine 98, 104893 (2023).
Pieren, D. K. J. et al. Limited induction of polyfunctional lung-resident memory T cells against SARS-CoV-2 by mRNA vaccination compared to infection. Nat. Commun. 14, 1887 (2023).
Mitsi, E. et al. Respiratory mucosal immune memory to SARS-CoV-2 after infection and vaccination. Nat. Commun. 14, 6815 (2023).
Morens, D. M., Taubenberger, J. K. & Fauci, A. S. Rethinking next-generation vaccines for coronaviruses, influenzaviruses, and other respiratory viruses. Cell Host Microbe 31, 146–157 (2023).
Meyerowitz, E. A., Scott, J., Richterman, A., Male, V. & Cevik, M. Clinical course and management of COVID-19 in the era of widespread population immunity. Nat. Rev. Microbiol. 22, 75–88 (2024).
Lucas, C. et al. Longitudinal analyses reveal immunological misfiring in severe COVID-19. Nature 584, 463–469 (2020).
Arunachalam, P. S. et al. Systems biological assessment of immunity to mild versus severe COVID-19 infection in humans. Science 369, 1210–1220 (2020).
Unterman, A. et al. Single-cell multi-omics reveals dyssynchrony of the innate and adaptive immune system in progressive COVID-19. Nat. Commun. 13, 440 (2022).
Diray-Arce, J. et al. Multi-omic longitudinal study reveals immune correlates of clinical course among hospitalized COVID-19 patients. Cell Rep. Med. 4, 101079 (2023).
Bastard, P. et al. Autoantibodies against type I IFNs in patients with life-threatening COVID-19. Science 370, eabd4585 (2020).
van der Wijst, M. G. P. et al. Type I interferon autoantibodies are associated with systemic immune alterations in patients with COVID-19. Sci. Transl. Med. 13, eabh2624 (2021).
Zhang, Q. et al. Inborn errors of type I IFN immunity in patients with life-threatening COVID-19. Science 370, eabd4570 (2020).
Ziegler, C. G. K. et al. Impaired local intrinsic immunity to SARS-CoV-2 infection in severe COVID-19. Cell 184, 4713–4733 (2021).
Sposito, B. et al. The interferon landscape along the respiratory tract impacts the severity of COVID-19. Cell 184, 4953–4968 (2021).
Chua, R. L. et al. COVID-19 severity correlates with airway epithelium–immune cell interactions identified by single-cell analysis. Nat. Biotechnol. 38, 970–979 (2020).
Loske, J. et al. Pre-activated antiviral innate immunity in the upper airways controls early SARS-CoV-2 infection in children. Nat. Biotechnol. 40, 319–324 (2021).
Yoshida, M. et al. Local and systemic responses to SARS-CoV-2 infection in children and adults. Nature 602, 321–327 (2022).
Lindeboom, R. G. H. et al. Human SARS-CoV-2 challenge uncovers local and systemic response dynamics. Nature 631, 189–198 (2024).
Hewitt, R. J. & Lloyd, C. M. Regulation of immune responses by the airway epithelial cell landscape. Nat. Rev. Immunol. 21, 347–362 (2021).
Montoro, D. T. et al. A revised airway epithelial hierarchy includes CFTR-expressing ionocytes. Nature 560, 319–324 (2018).
Plasschaert, L. W. et al. A single-cell atlas of the airway epithelium reveals the CFTR-rich pulmonary ionocyte. Nature 560, 377–381 (2018).
Ruiz García, S. et al. Novel dynamics of human mucociliary differentiation revealed by single-cell RNA sequencing of nasal epithelial cultures. Development 146, dev177428 (2019).
Revinski, D. R. et al. CDC20B is required for deuterosome-mediated centriole production in multiciliated cells. Nat. Commun. 9, 4668 (2018).
Jackson, C. B., Farzan, M., Chen, B. & Choe, H. Mechanisms of SARS-CoV-2 entry into cells. Nat. Rev. Mol. Cell Biol. 23, 3–20 (2022).
Schulte-Schrepping, J. et al. Severe COVID-19 is marked by a dysregulated myeloid cell compartment. Cell 182, 1419–1440 (2020).
Cumplido-Laso, G., Benitez, D. A., Mulero-Navarro, S. & Carvajal-Gonzalez, J. M. Transcriptional regulation of airway epithelial cell differentiation: insights into the Notch pathway and beyond. Int. J. Mol. Sci. 24, 14789 (2023).
Zhou, Y. et al. Airway basal cells show regionally distinct potential to undergo metaplastic differentiation. eLife 11, e80083 (2022).
Silver, A. C., Arjona, A., Walker, W. E. & Fikrig, E. The circadian clock controls Toll-like receptor 9-mediated innate and adaptive immunity. Immunity 36, 251–261 (2012).
Katoku-Kikyo, N. et al. The circadian regulator PER1 promotes cell reprogramming by inhibiting inflammatory signaling from macrophages. PLoS Biol. 21, e3002419 (2023).
Wang, T. et al. PER1 prevents excessive innate immune response during endotoxin-induced liver injury through regulation of macrophage recruitment in mice. Cell Death Dis. 7, e2176 (2016).
Gibbs, J. et al. An epithelial circadian clock controls pulmonary inflammation and glucocorticoid action. Nat. Med. 20, 919–926 (2014).
Luan, H. H. et al. GDF15 is an inflammation-induced central mediator of tissue tolerance. Cell 178, 1231–1244 (2019).
Liew, F. et al. Large-scale phenotyping of patients with long COVID post-hospitalization reveals mechanistic subtypes of disease. Nat. Immunol. 25, 607–621 (2024).
Zhao, Y. et al. EGR1 functions as a new host restriction factor for SARS-CoV-2 to inhibit virus replication through the E3 ubiquitin ligase MARCH8. J. Virol. 97, e0102823 (2023).
Arunachalam, P. S. et al. Systems vaccinology of the BNT162b2 mRNA vaccine in humans. Nature 596, 410–416 (2021).
Tahtinen, S. et al. IL-1 and IL-1ra are key regulators of the inflammatory response to RNA vaccines. Nat. Immunol. 23, 532–542 (2022).
Yamaguchi, Y. et al. Consecutive BNT162b2 mRNA vaccination induces short-term epigenetic memory in innate immune cells. JCI Insight 7, e163347 (2022).
Cheong, J.-G. et al. Epigenetic memory of coronavirus infection in innate immune cells and their progenitors. Cell 186, 3882–3902 (2023).
WHO R&D Blueprint Novel Coronavirus COVID-19 Therapeutic Trial Synopsis (World Health Organization, 2020); https://www.who.int/publications/i/item/covid-19-therapeutic-trial-synopsis
Tang, Y. et al. Human nasopharyngeal swab processing for viable single-cell suspension. protocols.io; https://doi.org/10.17504/protocols.io.bjhkkj4w (2020).
Ordovas-Montanes, J. et al. Allergic inflammatory memory in human respiratory epithelial progenitor cells. Nature 560, 649–654 (2018).
Aicher, T. P. et al. Seq-Well: a sample-efficient, portable picowell platform for massively parallel single-cell RNA sequencing. In Single Cell Methods: Sequencing and Proteomics (ed. Proserpio, V.) 111–132 (Springer, 2019).
Gierahn, T. M. et al. Seq-Well: portable, low-cost RNA sequencing of single cells at high throughput. Nat. Methods 14, 395–398 (2017).
Drake, R. S. et al. in Single Cell Transcriptomics: Methods and Protocols (eds Calogero, R. A. & Benes, V.) 57–104 (Springer, 2023).
Hughes, T. K. et al. Second-strand synthesis-based massively parallel scRNA-seq reveals cellular states and molecular features of human inflammatory skin pathologies. Immunity 53, 878–894 (2020).
Macosko, E. Z. et al. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell 161, 1202–1214 (2015).
Kim, D. et al. The architecture of SARS-CoV-2 transcriptome. Cell 181, 914–921 (2020).
Hafemeister, C. & Satija, R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 20, 296 (2019).
Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).
Deprez, M. et al. A single-cell atlas of the human healthy airways. Am. J. Respir. Crit. Care Med. 202, 1636–1645 (2020).
Zheng, H. B. et al. Concerted changes in the pediatric single-cell intestinal ecosystem before and after anti-TNF blockade. eLife 12, RP91792 (2023).
Kazer, S. W. et al. Primary nasal influenza infection rewires tissue-scale memory response dynamics. Immunity 57, 1955–1974 (2024).
Quinn, T. P., Erb, I., Richardson, M. F. & Crowley, T. M. Understanding sequencing data as compositions: an outlook and review. Bioinformatics 34, 2870–2878 (2018).
Kotliar, D. et al. Identifying gene expression programs of cell-type identity and cellular activity with single-cell RNA-seq. eLife 8, e43803 (2019).
DeBruine, Z. J., Andrew Pospisilik, J. & Triche, T. J. Fast and interpretable non-negative matrix factorization for atlas-scale single cell data. Preprint at bioRxiv https://doi.org/10.1101/2021.09.01.458620 (2024).
Huang, D. W., Sherman, B. T. & Lempicki, R. A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57 (2009).
Quinn, T. P., Richardson, M. F., Lovell, D. & Crowley, T. M. propr: an R-package for identifying proportionally abundant features using compositional data analysis. Sci. Rep. 7, 16252 (2017).
Ziegler, C. G. K. et al. SARS-CoV-2 receptor ACE2 is an interferon-stimulated gene in human airway epithelial cells and is detected in specific cell subsets across tissues. Cell 181, 1016–1035 (2020).
Acknowledgements
We thank the study participants and their families for enabling this research, the clinical support staff at the UMMC for assistance with sample collection and members of the Ordovas-Montanes laboratory (Boston Children’s Hospital, Harvard Medical School, Broad Institute, Ragon Institute), the Shalek laboratory (MIT, Broad Institute, Ragon Institute), the Horwitz laboratory (Boston Children’s Hospital) and the Glover laboratory (UMMC, Tulane University) for thoughtful discussion and feedback. We sincerely thank A. Sharpe and all members of the Sharpe laboratory (Harvard Medical School) for insightful discussion and feedback as well. We thank J. Gould, K. Siddle, B. Li, S. Fleming and the Broad Institute viral-ngs and Cumulus teams for assistance with computational pipelines. This project was made possible in part by grant number 2020-216949 from the Chan Zuckerberg Initiative DAF, an advised fund of the Silicon Valley Community Foundation to A.K.S. and J.O.-M. Work performed through the UMMC Molecular and Genomics Facility is supported, in part, by funds from the NIGMS, including the Molecular Center of Health and Disease (P20GM144041, M.R.G.), the Mississippi INBRE (P20GM103476, A. Flynt) and Obesity, Cardiorenal and Metabolic Diseases—COBRE (P30GM149404, J. Hall). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript. J.O-.M. is a New York Stem Cell Foundation—Robertson Investigator. J.O.-M. was supported by the AbbVie–Harvard Medical School Alliance, the Richard and Susan Smith Family Foundation, the AGA Research Foundation’s AGA–Takeda Pharmaceuticals Research Scholar Award in IBD (AGA2020-13-01), the HDDC Pilot and Feasibility (P30 DK034854), the Leona M. and Harry B. Helmsley Charitable Trust, the Pew Charitable Trusts Biomedical Scholars, the Broad Next Generation Award, the Chan Zuckerberg Initiative Pediatric Networks, the Mathers Foundation, the New York Stem Cell Foundation, NIH R01 HL162642, NIH R01 DE031928 and the Cell Discovery Network, a collaborative funded by the Manton Foundation and the Warren Alpert Foundation at Boston Children’s Hospital. J.M.L.W. was supported by NIH Training Grant 5TL1TR002543, S.W.K. was supported by the Cancer Research Institute’s Irvington Postdoctoral Fellowship, and A.K.S. was supported by the Bill and Melinda Gates Foundation, a Sloan Fellowship in Chemistry, the NIH (5U24AI118672) and the Ragon Institute of MGH, MIT and Harvard.
Author information
Authors and Affiliations
Contributions
Conceptualization, J.O.-M., A.K.S., S.C.G., B.H.H.; methodology, J.M.L.W., V.N.M., A.H.O., Y.T., J.D.B., S.W.K., K.K., C.A., C.G.K.Z., A.W.N., S.C.G., J.O.-M., A.K.S., B.H.H.; software, K.K., S.W.K.; formal analysis, J.M.L.W., V.N.M., A.H.O., Y.T., J.D.B., S.W.K., K.K; investigation, J.M.L.W., V.N.M., A.H.O., Y.T., J.D.B., C.G.K.Z., A.W.N., S.I., T.J., M.G., R.S.D., J.T.B., B.C.B., D.A.R.; resources, S.C.G., A.H.O.; data curation, A.H.O, Y.T., N.S.D., T.O.R., J.M.L.W.; writing (original draft), J.M.L.W., J.O.-M., A.K.S., S.C.G., B.H.H.; writing (review and editing), J.M.L.W., V.N.M., A.H.O., Y.T., J.D.B., S.W.K., K.K., C.A., C.G.K.Z., S.I., T.J., M.G., A.W.N., R.S.D., A.P., B.C.B., P.D., S.T., S.K.K., H.L., T.G.W., Y.T.D., N.S.D., Y.P., Y.G., M.S., J.H., J.T.B., G.D., M.R.G., D.A.R., I.J.F., J.J.L., T.O.R., J.O.-M., A.K.S., S.C.G., B.H.H.; visualization, J.M.L.W., Y.T.; supervision, J.O.-M., A.K.S., S.C.G., B.H.H., T.O.R.; project administration, Y.P., T.O.R.; funding acquisition, J.O.-M., A.K.S., S.C.G., B.H.H.
Corresponding author
Ethics declarations
Competing interests
V.N.M. reports compensation from MPM Capital and RA Capital Management unrelated to this work. S.W.K. reports compensation for consulting services with Monopteros Therapeutics, Flagship Pioneering and Radera Biosciences. M.S. reports compensation for consulting services with GE Precision Healthcare. J.J.L. reports compensation for consulting services with Blueprint Medicines and Human Immunology Biosciences. J.O.-M. reports compensation for consulting services with Cellarity, Tessel Biosciences and Radera Biotherapeutics. A.K.S. reports compensation for consulting and/or SAB membership from Merck, Honeycomb Biotechnologies, Cellarity, Hovione, Ochre Bio, Third Rock Ventures, Relation Therapeutics, Dahlia Biosciences, Bio-Rad Laboratories, IntrECate Biotherapeutics, FogPharma and Passkey Therapeutics. C.G.K.Z., V.N.M., A.H.O., A.W.N., Y.T., J.D.B., A.K.S., S.C.G., B.H.H. and J.O.-M. are co-inventors on a provisional patent application relating to methods of stratifying and treating viral infections. The other authors declare no competing interests.
Peer review
Peer review information
Nature Immunology thanks Daniel Altmann and Martijn Nawijn for their contribution to the peer review of this work. Peer reviewer reports are available. 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 Participant metadata across variant and vaccination groups.
a-b) Stacked bar charts showing fraction of Delta or Omicron cases in SARS-CoV-2 variant (a) and lineage (b) groups as identified by targeted sequencing of viral RNA from supernatant collected during nasopharyngeal swab processing (restricted to participants with SARS-CoV-2 S CT value < 30, Delta: n = 22, Omicron: n = 12). ‘Undetermined’: CT values > 30, sample not sequenced (Delta: n = 8, Omicron: n = 9). SARS-CoV-2 variant (a) and lineages (b) were determined using DRAGEN COVIDSeq Test software (see Methods, ‘Viral Variant Sequencing’). c) Comparison of COVID-19 severity score (WHO score of respiratory support) at time of nasopharyngeal swab collection (‘Swab’) and peak disease severity (‘Peak’) in all ancestral (n = 32), Delta (n = 33), and Omicron (n = 21) cases. d) Length of time in days from positive SARS-CoV-2 PCR test to nasopharyngeal swab collection (all cases), from reported symptom onset to nasopharyngeal swab collection (Ancestral: n = 30, Delta: n = 31, Omicron: n = 18, Delta vs. Ancestral: p = 0.031), and from reported symptom onset to peak COVID-19 severity score (Ancestral: n = 30, Delta: n = 31, Omicron: n = 18). e) SARS-CoV-2 plasma serology across variant groups. Plasma samples were taken from a subset of participants (Ancestral: n = 24, Delta: n = 30, Omicron: n = 17) on the same day as the nasopharyngeal swab used for scRNA-seq. Dotted lines indicate limits of detection. RBD-W: original Washington strain receptor binding domain; RBD-D: Delta strain receptor binding domain; NP: nucleoprotein. f) Comparison of COVID-19 severity score as in (c) across vaccination groups (Delta unvaccinated (‘Unvax’): n = 19, Delta vaccinated (‘Vax’): n = 14, Omicron unvaccinated: n = 10, Omicron vaccinated: n = 11). g) Length of time in days between first vaccine dose or most recent vaccine dose and positive SARS-CoV-2 PCR test in vaccinated Delta (n = 14) and Omicron (n = 10) cases. h) Disease course timing as in (d) across vaccination groups (All cases for PCR test to sample collection, for symptom onset data Delta unvaccinated: n = 19, Delta vaccinated: n = 12, Omicron unvaccinated: n = 7, Omicron vaccinated: n = 10. i) SARS-CoV-2 plasma serology as in (e) across vaccination groups (Delta unvaccinated: n = 18, Delta vaccinated: n = 12, Omicron unvaccinated: n = 7, Omicron vaccinated: n = 10, Delta vaccinated vs Delta unvaccinated NP IgG: p = 0.049). d,e,h,i, statistical test represents two-sided Kruskal-Wallis test results with Benjamini-Hochberg correction for multiple comparison. *Dunn post hoc test, p < 0.05.
Extended Data Fig. 2 scRNA-seq Quality Metrics and Cell Type Distributions.
a) Single cell quality metrics (total number of cells, number of unique UMI (unique molecular identifiers) per cell, number of unique genes per cell, and percent mitochondrial transcripts) for each participant (bars or violins) after filtering for low quality cells. b) UMAP of 48,730 cells from all participants (n = 112) colored by variant group (left), UMAP of 23,987 cells from Delta and Omicron participants (n = 54) colored by vaccination status (center), and UMAP of 48,730 cells from all participants (n = 112) colored by WHO peak respiratory support score (right). c) Stacked bar chart showing frequency of major cell types across variant (control: n = 26, ancestral, n = 32, Delta, n = 33, Omicron, n = 21) and vaccination (Delta unvaccinated (‘Unvax’): n = 19, Delta vaccinated (‘Vax’): n = 14, Omicron unvaccinated: n = 10, Omicron vaccinated: n = 11) groups. d) Frequency of secretory cells (ancestral vs. control: p = 4.8*10−4), deuterosomal cells (ancestral vs. control: p = 5.6*10−6, Delta vs. control: p = 0.054), ciliated cells (Delta vs. control: p = 0.011, Omicron vs. control: p = 0.031), SARS-CoV-2hi cells (ancestral vs. control: p = 0.013, Delta vs. control: p = 0.03, Omicron vs. control: p = 5*10−3), T cells, and macrophages as a fraction of all cells in that lineage (epithelial or immune) in that sample from all controls (n = 26) and all ancestral (n = 32), Delta (n = 33), and Omicron (n = 21) cases. Box plots represent median (center line), upper and lower quartiles (box limits); and 1.5x the inter-quartile range (whiskers). Compare to Fig. 1f. Statistical test represents two-sided Kruskal-Wallis test with Benjamini-Hochberg correction for multiple comparisons. *Dunn’s post hoc test, p < 0.05.
Extended Data Fig. 3 Epithelial diversity in the nasal mucosa during SARS-CoV-2 infection.
a) UMAP of 43,322 epithelial cells from all participants (n = 112) colored by major epithelial cell type as identified in Fig. 1d. b-c) UMAPs of detailed subsets within (b) basal/secretory cells (n = 112 participants, 9,106 cells) and (c) squamous cells (n = 112 participants, 3,913 cells). Cells from all participants belonging to that cell type were sub-clustered with louvain clustering. See Supplementary Tables 3, 4 for full lists of subset marker genes. d) Violin plot of marker genes for basal, secretory, goblet, squamous, and ionocyte subsets. e–g) UMAPs of detailed subsets within (e) ciliated cells (n = 112 participants, 20,683 cells), (f) MThi cells (n = 112 participants, 5,930 cells) and (g) SARS-CoV-2 RNAhi cells (n = 112 participants, 2,551 cells). h) Violin plot of marker genes for ciliated, deuterosomal, MThi, and SARS-CoV-2 RNAhi subsets.
Extended Data Fig. 4 Immune cell diversity in the nasal mucosa during SARS-CoV-2 infection.
a) UMAP of 5,408 immune cells from all participants colored by major immune cell type as identified in Fig. 1d. b-c) UMAPs of detailed subclusters within (b) T cells (n = 112 participants, 2,705 cells) and (c) myeloid cells (n = 112 participants, 2,535 cells). Cells from all participants belonging to that cell type were sub-clustered with louvain clustering. See Supplementary Table 5 for full lists of subset marker genes. d) Violin plot of marker genes for each immune subset.
Extended Data Fig. 5 Non-negative matrix factorization (NMF) Analysis.
a) Gene loadings of ciliated factors 8, 10, 13, and 21 identified by running NMF on all ciliated cells (n = 20,683) from all participants (n = 112). Complete list of genes contributing to all 25 factors is provided in Supplementary Table 7. b) Average score of ciliated factor 8 (Delta vs. control: p = 0.012, Omicron vs. control: p = 8.46*10−3), ciliated factor 10 (ancestral vs. control: p = 7.54*10−6, Delta vs. control, p = 3.32*10−3, Omicron vs. control: p = 0.011), ciliated factor 13 (ancestral vs. control: p = 3.89*10−3, Delta vs. control: p = 1.98*10−5, Omicron vs. control: p = 3*10−3), and ciliated factor 21 (ancestral vs. control: p = 1.08*10−4, Delta vs. control, p = 4.51*10−4, Omicron vs. control: p = 4.35*10−3) within pseudobulk ciliated cells for all controls (n = 26) and ancestral (n = 32), Delta (n = 33), and Omicron (n = 21) cases. c) Gene loadings of secretory factors 6, 11, 5, and 20 identified by running NMF on all basal and secretory cells (n = 9,106) from all participants (n = 112). Complete list of genes contributing to all 23 factors is provided in Supplementary Table 7. d) Average factor score of secretory factor 6 (ancestral vs. control: p = 0.033, Delta vs. control, p = 5.1*10−4, Omicron vs. control: p = 0.035) and secretory factor 11 (ancestral vs. control: p = 2.72*10−3) within pseudobulk secretory cells for all controls (n = 26) and ancestral (n = 32), Delta (n = 33), and Omicron (n = 21) cases. e) Violin plot of scores for secretory factors 6, 11, 5, and 20 across all basal, goblet, and secretory subsets in all participants (n = 112). b,d Box plots represent median (center line), upper and lower quartiles (box limits), and 1.5x the inter-quartile range (whiskers). Statistical tests represent two-sided Kruskal-Wallis test with Benjamini-Hochberg correction for multiple comparisons. *Dunn’s post hoc test, p < 0.05.
Extended Data Fig. 6 Detection and assignment of SARS-CoV-2 RNA+ cells.
a) Schematic of method for SARS-CoV-2 RNA+ assignment. scRNA-seq data was aligned to a joint human and SARS-CoV-2 genome. The amount of ambient RNA in each cell was estimated using Cellbender. Cells were assigned SARS-CoV-2 RNA+ if the amount of viral RNA was greater than the expected ambient amount (see Methods). b) Total SARS-CoV-2 transcripts per cell in a subset of ancestral (n = 30), Delta (n = 31), and Omicron (n = 18) cases, divided by time from symptom onset. c) Spearman correlation (two-tailed test) between the total number of transcripts (UMI) per cell and the total number of SARS-CoV-2 transcripts per cell in all ancestral (n = 32), Delta (n = 33), and Omicron (n = 21) cases. Schematic in a created with BioRender.com.
Extended Data Fig. 7 Frequency of Major Cell Types Across Vaccination Groups.
a) Frequency of B cells, dendritic cells, macrophages, and T cells as a percentage of all cells in the sample in control participants. Control (‘Ctrl’) unvaccinated (‘Unvax’) (n = 13) samples were collected during the Ancestral wave, and control vaccinated (‘Vax’) (n = 13) samples were collected during the Delta and Omicron waves. b) Frequency of epithelial cell types (basal cells, ciliated cells, deuterosomal cells, goblet cells, ionocytes, MThi cells, SARS-CoV-2hi cells (Ctrl Vax vs Ctrl Unvax: p = 0.041), secretory cells, and squamous cells) as a percentage of all cells in the sample in unvaccinated (‘Unvax’) controls (n = 13), vaccinated (‘Vax’) controls (n = 13), unvaccinated Delta (n = 19), vaccinated Delta (n = 14), unvaccinated Omicron (n = 10), and vaccinated Omicron (n = 11) cases. c) Spearman correlation (two-tailed test) between time from most recent SARS-CoV-2 vaccine dose and frequency of immune cell types (B cells, dendritic cells, macrophages, and T cells) in all vaccinated Delta (n = 14) and Omicron (n = 11) cases. Error bands represent 95% confidence interval. a,b Box plots represent median (center line), upper and lower quartiles (box limits); and 1.5x the inter-quartile range (whiskers). Statistical tests represent two-sided Kruskal-Wallis test with Benjamini-Hochberg correction for multiple comparisons. *Dunn’s post hoc test, p < 0.05.
Extended Data Fig. 8 Expression of ISG Modules Across COVID-19 Severity.
a-b) Violin plots of IFN alpha (a) and IFN gamma (b) response gene module scores in ciliated (n = 15,180 cells), secretory (n = 6,656), and T cells (n = 2,181) from all ancestral (n = 32), Delta (n = 33), and Omicron (n = 21) cases. Cells are separated by COVID-19 severity score (WHO range 0–8). Genes uses for module score were derived from previously published results of stimulating human nasal basal cells with IFN alpha or IFN gamma (Supplementary Table 10)70. Lines represent local regression using locally estimated scatterplot smoothing. Error bands represent normal confidence interval.
Extended Data Fig. 9 Association of clinical metadata and nasal cell type frequencies.
Heatmap of spearman correlation (two-tailed test) between clinical parameters and demographic information (rows) and frequency of major cell types (as identified in Fig. 1) and SARS-CoV-2 RNA+ cells (as identified in Fig. 3) (columns) in all participants (n = 112). Color indicates spearman R, *FDR < 0.05.
Supplementary information
Supplementary Table 1
Demographic and clinical metadata of all participants.
Supplementary Table 2
Clustering and removal of low-quality cells: marker genes and annotations.
Supplementary Table 3
Epithelial clustering round 1 marker genes and annotations.
Supplementary Table 4
Epithelial clustering round 2–3 marker genes and annotations.
Supplementary Table 5
Immune clustering marker genes and annotations.
Supplementary Table 6
Abundance of cell subsets for each participant (normalized to 500 cells per participant).
Supplementary Table 7
Gene loadings for NMF analysis within ciliated and secretory cells.
Supplementary Table 8
DE genes in macrophages and T cells between vaccinated and unvaccinated Delta-infected participants.
Supplementary Table 9
GO terms in genes upregulated in macrophages from vaccinated Delta cases compared to macrophages from unvaccinated Delta cases.
Supplementary Table 10
Human nasal basal cell cytokine stimulation response genes (Ziegler et al.70).
Supplementary Table 11
Significant correlations between cell type frequencies and participant 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
Walsh, J.M.L., Miao, V.N., Owings, A.H. et al. Variants and vaccines impact nasal immunity over three waves of SARS-CoV-2. Nat Immunol 26, 294–307 (2025). https://doi.org/10.1038/s41590-024-02052-z
Received:
Accepted:
Published:
Issue Date:
DOI: https://doi.org/10.1038/s41590-024-02052-z
This article is cited by
-
Variant-specific local tissue response to SARS-CoV-2 in the nasal mucosa
Nature Immunology (2025)