Circuits between infected macrophages and T cells in SARS-CoV-2 pneumonia.

Rogan A Grant, Luisa Morales-Nebreda, Nikolay S Markov, Suchitra Swaminathan, Melissa Querrey, Estefany R Guzman, Darryl A Abbott, Helen K Donnelly, Alvaro Donayre, Isaac A Goldberg, Zasu M Klug, Nicole Borkowski, Ziyan Lu, Hermon Kihshen, Yuliya Politanska, Lango Sichizya, Mengjia Kang, Ali Shilatifard, Chao Qi, Jon W Lomasney, A Christine Argento, Jacqueline M Kruser, Elizabeth S Malsin, Chiagozie O Pickens, Sean B Smith, James M Walter, Anna E Pawlowski, Daniel Schneider, Prasanth Nannapaneni, Hiam Abdala-Valencia, Ankit Bharat, Cara J Gottardi, G R Scott Budinger, Alexander V Misharin, Benjamin D Singer, Richard G Wunderink, NU SCRIPT Study Investigators
Author Information
  1. Rogan A Grant: Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA. ORCID
  2. Luisa Morales-Nebreda: Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
  3. Nikolay S Markov: Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA. ORCID
  4. Suchitra Swaminathan: Division of Rheumatology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
  5. Melissa Querrey: Division of Thoracic Surgery, Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
  6. Estefany R Guzman: Robert H. Lurie Comprehensive Cancer Research Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
  7. Darryl A Abbott: Robert H. Lurie Comprehensive Cancer Research Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
  8. Helen K Donnelly: Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
  9. Alvaro Donayre: Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
  10. Isaac A Goldberg: Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA. ORCID
  11. Zasu M Klug: Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
  12. Nicole Borkowski: Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
  13. Ziyan Lu: Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
  14. Hermon Kihshen: Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
  15. Yuliya Politanska: Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
  16. Lango Sichizya: Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
  17. Mengjia Kang: Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
  18. Ali Shilatifard: Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA. ORCID
  19. Chao Qi: Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
  20. Jon W Lomasney: Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA. ORCID
  21. A Christine Argento: Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
  22. Jacqueline M Kruser: Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
  23. Elizabeth S Malsin: Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
  24. Chiagozie O Pickens: Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
  25. Sean B Smith: Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
  26. James M Walter: Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
  27. Anna E Pawlowski: Clinical and Translational Sciences Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
  28. Daniel Schneider: Clinical and Translational Sciences Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA. ORCID
  29. Prasanth Nannapaneni: Clinical and Translational Sciences Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
  30. Hiam Abdala-Valencia: Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
  31. Ankit Bharat: Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
  32. Cara J Gottardi: Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
  33. G R Scott Budinger: Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA. s-buding@northwestern.edu. ORCID
  34. Alexander V Misharin: Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA. a-misharin@northwestern.edu. ORCID
  35. Benjamin D Singer: Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA. benjamin-singer@northwestern.edu. ORCID
  36. Richard G Wunderink: Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA. r-wunderink@northwestern.edu. ORCID

Abstract

Some patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) develop severe pneumonia and acute respiratory distress syndrome (ARDS). Distinct clinical features in these patients have led to speculation that the immune response to virus in the SARS-CoV-2-infected alveolus differs from that in other types of pneumonia. Here we investigate SARS-CoV-2 pathobiology by characterizing the immune response in the alveoli of patients infected with the virus. We collected bronchoalveolar lavage fluid samples from 88 patients with SARS-CoV-2-induced respiratory failure and 211 patients with known or suspected pneumonia from other pathogens, and analysed them using flow cytometry and bulk transcriptomic profiling. We performed single-cell RNA sequencing on 10 bronchoalveolar lavage fluid samples collected from patients with severe coronavirus disease 2019 (COVID-19) within 48 h of intubation. In the majority of patients with SARS-CoV-2 infection, the alveolar space was persistently enriched in T cells and monocytes. Bulk and single-cell transcriptomic profiling suggested that SARS-CoV-2 infects alveolar macrophages, which in turn respond by producing T cell chemoattractants. These T cells produce interferon-γ to induce inflammatory cytokine release from alveolar macrophages and further promote T cell activation. Collectively, our results suggest that SARS-CoV-2 causes a slowly unfolding, spatially limited alveolitis in which alveolar macrophages containing SARS-CoV-2 and T cells form a positive feedback loop that drives persistent alveolar inflammation.

References

  1. Yang, X. et al. Clinical course and outcomes of critically ill patients with SARS-CoV-2 pneumonia in Wuhan, China: a single-centered, retrospective, observational study. Lancet Respir. Med. 8, 475–481 (2020). [PMID: 32105632]
  2. Gattinoni, L. et al. COVID-19 does not lead to a “typical” acute respiratory distress syndrome. Am. J. Respir. Crit. Care Med. 201, 1299–1300 (2020). [PMID: 32228035]
  3. Baud, D. et al. Real estimates of mortality following COVID-19 infection. Lancet Infect. Dis. 20, 773 (2020). [PMID: 32171390]
  4. Zhou, F. et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet 395, 1054–1062 (2020). [PMID: 32171076]
  5. RECOVERY Collaborative Group. Dexamethasone in hospitalized patients with Covid-19—preliminary report. N. Engl. J. Med. NEJMoa2021436 (2020).
  6. National Heart, Lung, and Blood Institute PETAL Clinical Trials Network. Early neuromuscular blockade in the acute respiratory distress syndrome. N. Engl. J. Med. 380, 1997–2008 (2019). [DOI: 10.1056/NEJMoa1901686]
  7. Watanabe, S., Alexander, M., Misharin, A. V. & Budinger, G. R. S. The role of macrophages in the resolution of inflammation. J. Clin. Invest. 129, 2619–2628 (2019). [PMID: 31107246]
  8. Kim, D. et al. The architecture of SARS-CoV-2 transcriptome. Cell 181, 914–921 (2020). [PMID: 32330414]
  9. Liao, M. et al. Single-cell landscape of bronchoalveolar immune cells in patients with COVID-19. Nat. Med. 26, 842–844 (2020). [PMID: 32398875]
  10. 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). [PMID: 32591762]
  11. Sola, I., Almazán, F., Zúñiga, S. & Enjuanes, L. Continuous and discontinuous RNA synthesis in coronaviruses. Annu. Rev. Virol. 2, 265–288 (2015). [PMID: 26958916]
  12. Zhou, J. et al. Active replication of Middle East respiratory syndrome coronavirus and aberrant induction of inflammatory cytokines and chemokines in human macrophages: implications for pathogenesis. J. Infect. Dis. 209, 1331–1342 (2014). [PMID: 24065148]
  13. Yip, M. S. et al. Antibody-dependent infection of human macrophages by severe acute respiratory syndrome coronavirus. Virol. J. 11, 82 (2014). [PMID: 24885320]
  14. Cheung, C. Y. et al. Cytokine responses in severe acute respiratory syndrome coronavirus-infected macrophages in vitro: possible relevance to pathogenesis. J. Virol. 79, 7819–7826 (2005). [PMID: 15919935]
  15. Soberanes, S. et al. Metformin targets mitochondrial electron transport to reduce air-pollution-induced thrombosis. Cell Metab. 29, 503 (2019). [PMID: 30726761]
  16. Price, L. C., McCabe, C., Garfield, B. & Wort, S. J. Thrombosis and COVID-19 pneumonia: the clot thickens! Eur. Respir. J. 56, 2001608 (2020). [PMID: 32554532]
  17. Vabret, N. et al. Immunology of COVID-19: current state of the science. Immunity 52, 910–941 (2020). [PMID: 32505227]
  18. Wilk, A. J. et al. A single-cell atlas of the peripheral immune response in patients with severe COVID-19. Nat. Med. 26, 1070–1076 (2020). [PMID: 32514174]
  19. Schulte-Schrepping, J. et al. Severe COVID-19 is marked by a dysregulated myeloid cell compartment. Cell 182, 1419–1440 (2020). [PMID: 32810438]
  20. Habermann, A. C. et al. Single-cell RNA sequencing reveals profibrotic roles of distinct epithelial and mesenchymal lineages in pulmonary fibrosis. Sci. Adv. 6, eaba1972 (2020). [PMID: 32832598]
  21. Califano, D., Furuya, Y. & Metzger, D. W. Effects of influenza on alveolar macrophage viability are dependent on mouse genetic strain. J. Immunol. 201, 134–144 (2018). [PMID: 29760191]
  22. 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). [PMID: 32327758]
  23. Hou, Y. J. et al. SARS-CoV-2 reverse genetics reveals a variable infection gradient in the respiratory tract. Cell 182, 429–446 (2020). [PMID: 32526206]
  24. Chu, H. et al. Comparative replication and immune activation profiles of SARS-CoV-2 and SARS-CoV in human lungs: an ex vivo study with implications for the pathogenesis of COVID-19. Clin. Infect. Dis. 71, 1400–1409 (2020). [PMID: 32270184]
  25. Blanco-Melo, D. et al. Imbalanced host response to SARS-CoV-2 drives development of COVID-19. Cell 181, 1036–1045 (2020). [PMID: 32416070]
  26. Konno, Y. et al. SARS-CoV-2 ORF3b is a potent interferon antagonist whose activity is increased by a naturally occurring elongation variant. Cell Rep. 32, 108185 (2020). [PMID: 32941788]
  27. Hadjadj, J. et al. Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients. Science 369, 718–724 (2020). [PMID: 32661059]
  28. Jaume, M. et al. Anti-severe acute respiratory syndrome coronavirus spike antibodies trigger infection of human immune cells via a pH- and cysteine protease-independent FcγR pathway. J. Virol. 85, 10582–10597 (2011). [PMID: 21775467]
  29. Martines, R. B. et al. Pathology and pathogenesis of SARS-CoV-2 associated with fatal coronavirus disease, United States. Emerg. Infect. Dis. 26, 2005–2015 (2020). [PMID: 32437316]
  30. Sariol, A. & Perlman, S. Lessons for COVID-19 immunity from other coronavirus infections. Immunity 53, 248–263 (2020). [PMID: 32717182]
  31. Liu, L. et al. Anti-spike IgG causes severe acute lung injury by skewing macrophage responses during acute SARS-CoV infection. JCI Insight 4, e123158 (2019). [>PMCID: ]
  32. Guilliams, M., Lambrecht, B. N. & Hammad, H. Division of labor between lung dendritic cells and macrophages in the defense against pulmonary infections. Mucosal Immunol. 6, 464–473 (2013). [PMID: 23549447]
  33. Gioia, C. et al. T-Cell response profiling to biological threat agents including the SARS coronavirus. Int. J. Immunopathol. Pharmacol. 18, 525–530 (2005). [PMID: 16164833]
  34. 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). [PMID: 32473127]
  35. 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). [PMID: 32668444]
  36. Mateus, J. et al. Selective and cross-reactive SARS-CoV-2 T cell epitopes in unexposed humans. Science 370, 89–94 (2020). [PMID: 32753554]
  37. Braun, J. et al. SARS-CoV-2-reactive T cells in healthy donors and patients with COVID-19. Nature 587, 270–274 (2020). [PMID: 32726801]
  38. 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). [PMID: 33296686]
  39. Oran, D. P. & Topol, E. J. Prevalence of asymptomatic SARS-CoV-2 infection: a narrative review. Ann. Intern. Med. 173, 362–367 (2020). [PMID: 32491919]
  40. Neupane, A. S. et al. Patrolling alveolar macrophages conceal bacteria from the immune system to maintain homeostasis. Cell 183, 110–125 (2020). [PMID: 32888431]
  41. Louie, J. K. et al. Factors associated with death or hospitalization due to pandemic 2009 influenza A(H1N1) infection in California. J. Am. Med. Assoc. 302, 1896–1902 (2009). [DOI: 10.1001/jama.2009.1583]
  42. Leisman, D. E. et al. Cytokine elevation in severe and critical COVID-19: a rapid systematic review, meta-analysis, and comparison with other inflammatory syndromes. Lancet Respir. Med. 8, 1233–1244 (2020). [PMID: 33075298]
  43. Kox, M., Waalders, N. J. B., Kooistra, E. J., Gerretsen, J. & Pickkers, P. Cytokine levels in critically ill patients with COVID-19 and other conditions. J. Am. Med. Assoc. (2020).
  44. 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). [>PMCID: ]
  45. Acute Respiratory Distress Syndrome Network. Ventilation with lower tidal volumes as compared with traditional tidal volumes for acute lung injury and the acute respiratory distress syndrome. N. Engl. J. Med. 342, 1301–1308 (2000). [DOI: 10.1056/NEJM200005043421801]
  46. Brower, R. G. et al. Higher versus lower positive end-expiratory pressures in patients with the acute respiratory distress syndrome. N. Engl. J. Med. 351, 327–336 (2004). [PMID: 15269312]
  47. Guérin, C. et al. Prone positioning in severe acute respiratory distress syndrome. N. Engl. J. Med. 368, 2159–2168 (2013). [PMID: 23688302]
  48. Gao, C. A. et al. Bronchoscopy on intubated COVID-19 patients is associated with low infectious risk to operators. Ann. Am. Thorac. Soc. https://doi.org/10.1513/AnnalsATS.202009-1225RL (2020).
  49. Walter, J. M. et al. Multidimensional assessment of the host response in mechanically ventilated patients with suspected pneumonia. Am. J. Respir. Crit. Care Med. 199, 1225–1237 (2019). [PMID: 30398927]
  50. Yu, Y.-R. A. et al. Flow cytometric analysis of myeloid cells in human blood, bronchoalveolar lavage, and lung tissues. Am. J. Respir. Cell Mol. Biol. 54, 13–24 (2016). [PMID: 26267148]
  51. Bharat, A. et al. Flow cytometry reveals similarities between lung macrophages in humans and mice. Am. J. Respir. Cell Mol. Biol. 54, 147–149 (2016). [PMID: 26274047]
  52. Charbonneau, D. H. & James, L. N. FluView and FluNet: tools for influenza activity and surveillance. Med. Ref. Serv. Q. 38, 358–368 (2019). [PMID: 31687905]
  53. Di Tommaso, P. et al. Nextflow enables reproducible computational workflows. Nat. Biotechnol. 35, 316–319 (2017). [PMID: 28398311]
  54. Ewels, P. A. et al. The nf-core framework for community-curated bioinformatics pipelines. Nat. Biotechnol. 38, 276–278 (2020). [PMID: 32055031]
  55. Kurtzer, G. M., Sochat, V. & Bauer, M. W. Singularity: scientific containers for mobility of compute. PLoS ONE 12, e0177459 (2017). [PMID: 28494014]
  56. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013). [PMID: 23104886]
  57. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014). [PMID: 24227677]
  58. Lee, S. et al. NGSCheckMate: software for validating sample identity in next-generation sequencing studies within and across data types. Nucleic Acids Res. 45, e103 (2017). [PMID: 28369524]
  59. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014). [PMID: 25516281]
  60. Langfelder, P. & Horvath, S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics 9, 559 (2008). [PMID: 19114008]
  61. McInnes, L., Healy, J. & Melville, J. UMAP: uniform manifold approximation and projection for dimension reduction. Preprint at https://arxiv.org/abs/1802.03426 (2018).
  62. Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018). [PMID: 29409532]
  63. Wolock, S. L., Lopez, R. & Klein, A. M. Scrublet: computational identification of cell doublets in single-cell transcriptomic data. Cell Syst. 8, 281–291 (2019). [PMID: 30954476]
  64. Polański, K. et al. BBKNN: fast batch alignment of single cell transcriptomes. Bioinformatics 36, 964–965 (2020). [PMID: 31400197]
  65. 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). [PMID: 16199517]
  66. Köster, J. & Rahmann, S. Snakemake-a scalable bioinformatics workflow engine. Bioinformatics 28, 2520–2522 (2012). [PMID: 22908215]
  67. Heaton, H. et al. Souporcell: robust clustering of single-cell RNA-seq data by genotype without reference genotypes. Nat. Methods 17, 615–620 (2020). [PMID: 32366989]
  68. Aliee, H. & Theis, F. AutoGeneS: Automatic gene selection using multi-objective optimization for RNA-seq deconvolution. Preprint at https://doi.org/10.1101/2020.02.21.940650 (2020).
  69. Wickham, H. et al. Welcome to the tidyverse. J. Open Source Softw. 4, 1686 (2019). [DOI: 10.21105/joss.01686]
  70. Virtanen, P. et al. SciPy 1.0: fundamental algorithms for scientific computing in Python. Nat. Methods 17, 261–272 (2020). [PMID: 32015543]
  71. Seabold, S. & Perktold, J. Statsmodels: econometric and statistical modeling with Python. in Proc. 9th Python in Science Conference Vol. 57 (eds van der Walt, S. & Millman, J.) 92–96 (2010).
  72. Brunson, J. ggalluvial: layered grammar for alluvial plots. J. Open Source Softw. 5, 2017 (2020). [DOI: 10.21105/joss.02017]
  73. Hunter, J. D. Matplotlib: a 2D graphics environment. Comput. Sci. Eng. 9, 90–95 (2007). [DOI: 10.1109/MCSE.2007.55]

Grants

  1. I01 CX001777/CSRD VA
  2. HL147290/NIH HHS
  3. R01 HL131745/NHLBI NIH HHS
  4. S10 OD011996/NIH HHS
  5. P01 AG049665/NIA NIH HHS
  6. U19 AI135964/NIAID NIH HHS
  7. P30 CA060553/NCI NIH HHS
  8. HL14578/NIH HHS
  9. R01 HL145478/NHLBI NIH HHS
  10. R01 HL147575/NHLBI NIH HHS
  11. R56 HL135124/NHLBI NIH HHS
  12. R01 HL134800/NHLBI NIH HHS
  13. GM129312/NIH HHS
  14. HL134800/NIH HHS
  15. K08 HL159356/NHLBI NIH HHS
  16. L30 CA153420/NCI NIH HHS
  17. K08 HL128867/NHLBI NIH HHS
  18. 1S10OD011996-01/NIH HHS
  19. R01 HL153312/NHLBI NIH HHS
  20. UL1 TR001422/NCATS NIH HHS
  21. T32 AG020506/NIA NIH HHS
  22. K99 AG068544/NIA NIH HHS
  23. HL147575/NIH HHS
  24. R01 HL147290/NHLBI NIH HHS
  25. F32 HL151127/NHLBI NIH HHS
  26. R01 HL154686/NHLBI NIH HHS
  27. R01 GM129312/NIGMS NIH HHS
  28. F31 AG071225/NIA NIH HHS
  29. R01 HL149883/NHLBI NIH HHS
  30. K07 CA216330/NCI NIH HHS
  31. T32 HL076139/NHLBI NIH HHS

MeSH Term

Bronchoalveolar Lavage Fluid
COVID-19
Cohort Studies
Humans
Interferon-gamma
Interferons
Macrophages, Alveolar
Pneumonia, Viral
RNA-Seq
SARS-CoV-2
Signal Transduction
Single-Cell Analysis
T-Lymphocytes
Time Factors

Chemicals

Interferon-gamma
Interferons