Dysregulated transcriptional responses to SARS-CoV-2 in the periphery.

Micah T McClain, Florica J Constantine, Ricardo Henao, Yiling Liu, Ephraim L Tsalik, Thomas W Burke, Julie M Steinbrink, Elizabeth Petzold, Bradly P Nicholson, Robert Rolfe, Bryan D Kraft, Matthew S Kelly, Daniel R Saban, Chen Yu, Xiling Shen, Emily M Ko, Gregory D Sempowski, Thomas N Denny, Geoffrey S Ginsburg, Christopher W Woods
Author Information
  1. Micah T McClain: Durham Veterans Affairs Medical Center, Durham, NC, USA. micah.mcclain@duke.edu. ORCID
  2. Florica J Constantine: Center for Applied Genomics and Precision Medicine, Duke University, Durham, NC, USA.
  3. Ricardo Henao: Center for Applied Genomics and Precision Medicine, Duke University, Durham, NC, USA.
  4. Yiling Liu: Center for Applied Genomics and Precision Medicine, Duke University, Durham, NC, USA.
  5. Ephraim L Tsalik: Durham Veterans Affairs Medical Center, Durham, NC, USA.
  6. Thomas W Burke: Center for Applied Genomics and Precision Medicine, Duke University, Durham, NC, USA.
  7. Julie M Steinbrink: Center for Applied Genomics and Precision Medicine, Duke University, Durham, NC, USA.
  8. Elizabeth Petzold: Center for Applied Genomics and Precision Medicine, Duke University, Durham, NC, USA.
  9. Bradly P Nicholson: Institute for Medical Research, Durham, NC, USA.
  10. Robert Rolfe: Division of Infectious Diseases, Duke University Medical Center, Durham, NC, USA.
  11. Bryan D Kraft: Durham Veterans Affairs Medical Center, Durham, NC, USA.
  12. Matthew S Kelly: Duke University Medical Center, Durham, NC, USA.
  13. Daniel R Saban: Department of Opthalmology, Duke University School of Medicine, Durham, NC, USA.
  14. Chen Yu: Department of Opthalmology, Duke University School of Medicine, Durham, NC, USA.
  15. Xiling Shen: Center for Genomics and Computational Biology, Duke University, Durham, NC, USA.
  16. Emily M Ko: Center for Applied Genomics and Precision Medicine, Duke University, Durham, NC, USA.
  17. Gregory D Sempowski: Duke Human Vaccine Institute, Durham, NC, USA.
  18. Thomas N Denny: Duke Human Vaccine Institute, Durham, NC, USA.
  19. Geoffrey S Ginsburg: Center for Applied Genomics and Precision Medicine, Duke University, Durham, NC, USA.
  20. Christopher W Woods: Durham Veterans Affairs Medical Center, Durham, NC, USA.

Abstract

SARS-CoV-2 infection has been shown to trigger a wide spectrum of immune responses and clinical manifestations in human hosts. Here, we sought to elucidate novel aspects of the host response to SARS-CoV-2 infection through RNA sequencing of peripheral blood samples from 46 subjects with COVID-19 and directly comparing them to subjects with seasonal coronavirus, influenza, bacterial pneumonia, and healthy controls. Early SARS-CoV-2 infection triggers a powerful transcriptomic response in peripheral blood with conserved components that are heavily interferon-driven but also marked by indicators of early B-cell activation and antibody production. Interferon responses during SARS-CoV-2 infection demonstrate unique patterns of dysregulated expression compared to other infectious and healthy states. Heterogeneous activation of coagulation and fibrinolytic pathways are present in early COVID-19, as are IL1 and JAK/STAT signaling pathways, which persist into late disease. Classifiers based on differentially expressed genes accurately distinguished SARS-CoV-2 infection from other acute illnesses (auROC 0.95 [95% CI 0.92-0.98]). The transcriptome in peripheral blood reveals both diverse and conserved components of the immune response in COVID-19 and provides for potential biomarker-based approaches to diagnosis.

References

  1. Tay, M. Z., Poh, C. M., Rénia, L., MacAry, P. A. & Ng, L. F. P. The trinity of COVID-19: immunity, inflammation and intervention. Nat. Rev. Immunol. 20, 363–374 (2020). [PMID: 32346093]
  2. Ong, E. Z. et al. A dynamic immune response shapes COVID-19 progression. Cell Host Microbe 27, 879–882.e2 (2020). [PMID: 32359396]
  3. Ouyang, Y. et al. Down-regulated gene expression spectrum and immune responses changed during the disease progression in COVID-19 patients. Clin. Infect. Dis. 71, 2052–2060 (2020). [PMID: 32307550]
  4. Wen, W. et al. Immune cell profiling of COVID-19 patients in the recovery stage by single-cell sequencing. Cell Discov. 6, 31 (2020). [PMID: 32377375]
  5. Liao, M. et al. Single-cell landscape of bronchoalveolar immune cells in patients with COVID-19. Nat. Med. 26, 842–844 (2020). [PMID: 32398875]
  6. Blanco-Melo, D. et al. Imbalanced host response to SARS-CoV-2 drives development of COVID-19. Cell 181, 1036–1045.e1039 (2020). [PMID: 32416070]
  7. Zhou, Z. et al. Heightened innate immune responses in the respiratory tract of COVID-19 patients. Cell Host Microbe 27, 883–890.e882 (2020). [PMID: 32407669]
  8. Mick, E. et al. Upper airway gene expression reveals suppressed immune responses to SARS-CoV-2 compared with other respiratory viruses. Nat. Commun. 11, 5854 (2020). [PMID: 33203890]
  9. 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]
  10. Huang, L. et al. Blood single cell immune profiling reveals the interferon-MAPK pathway mediated adaptive immune response for COVID-19. Preprint at medRxiv https://doi.org/10.1101/2020.03.15.20033472 (2020).
  11. Zhang, J.-Y. et al. Single-cell landscape of immunological responses in patients with COVID-19. Nat. Immunol. 21, 1107–1118 (2020). [PMID: 32788748]
  12. Arunachalam, P. S. et al. Systems biological assessment of immunity to mild versus severe COVID-19 infection in humans. Science 369, 1210–1220 (2020). [PMID: 32788292]
  13. Lee, J. S. et al. Immunophenotyping of COVID-19 and influenza highlights the role of type I interferons in development of severe COVID-19. Sci. Immunol. 5, eabd1554 (2020). [PMID: 32651212]
  14. Hadjadj, J. et al. Impaired type I interferon activity and inflammatory responses in severe COVID-19 patients. Science 369, 718–724 (2020). [PMID: 32661059]
  15. Kuri-Cervantes, L. et al. Comprehensive mapping of immune perturbations associated with severe COVID-19. Sci. Immunol. 5, eabd7114 (2020). [PMID: 32669287]
  16. Nakaya, H. I. et al. Systems biology of vaccination for seasonal influenza in humans. Nat. Immunol. 12, 786–795 (2011). [PMID: 21743478]
  17. Herberg, J. A. et al. Diagnostic test accuracy of a 2-transcript host RNA signature for discriminating bacterial vs viral infection in febrile children. JAMA 316, 835–845 (2016). [PMID: 27552617]
  18. Sweeney, T. E., Wong, H. R. & Khatri, P. Robust classification of bacterial and viral infections via integrated host gene expression diagnostics. Sci. Transl. Med. 8, 346ra391 (2016). [DOI: 10.1126/scitranslmed.aaf7165]
  19. Tsalik, E. L. et al. Host gene expression classifiers diagnose acute respiratory illness etiology. Sci. Transl. Med. 8, 322ra311 (2016). [DOI: 10.1126/scitranslmed.aad6873]
  20. McClain, M. T. et al. A genomic signature of influenza infection shows potential for presymptomatic detection, guiding early therapy, and monitoring clinical responses. Open Forum Infect. Dis. 3, ofw007 (2016). [PMID: 26933666]
  21. Zaas, A. K. et al. A host-based RT-PCR gene expression signature to identify acute respiratory viral infection. Sci. Transl. Med. 5, 203ra126 (2013). [PMID: 24048524]
  22. Woods, C. W. et al. A host transcriptional signature for presymptomatic detection of infection in humans exposed to influenza H1N1 or H3N2. PLoS One 8, e52198 (2013). [PMID: 23326326]
  23. Zaas, A. K. et al. Gene expression signatures diagnose influenza and other symptomatic respiratory viral infections in humans. Cell Host Microbe 6, 207–217 (2009). [PMID: 19664979]
  24. Mejias, A. et al. Whole blood gene expression profiles to assess pathogenesis and disease severity in infants with respiratory syncytial virus infection. PLoS Med. 10, e1001549 (2013). [PMID: 24265599]
  25. Spiegel, M. et al. Inhibition of beta interferon induction by severe acute respiratory syndrome coronavirus suggests a two-step model for activation of interferon regulatory factor 3. J. Virol. 79, 2079–2086 (2005). [PMID: 15681410]
  26. Magro, C. et al. Complement associated microvascular injury and thrombosis in the pathogenesis of severe COVID-19 infection: a report of five cases. Transl. Res.: J. Lab. Clin. Med. 220, 1–13 (2020). [DOI: 10.1016/j.trsl.2020.04.007]
  27. Poissy, J. et al. Pulmonary embolism in COVID-19 patients: awareness of an increased prevalence. Circulation 142, 184–186 (2020).
  28. Lewis, D. A. et al. Whole blood gene expression profiles distinguish clinical phenotypes of venous thromboembolism. Thrombosis Res. 135, 659–665 (2015). [DOI: 10.1016/j.thromres.2015.02.003]
  29. Kalil, A. C. et al. Baricitinib plus Remdesivir for hospitalized adults with Covid-19. N. Engl. J. Med.  https://doi.org/10.1056/NEJMoa2031994 (2020).
  30. Stone, J. H. et al. Efficacy of Tocilizumab in patients hospitalized with Covid-19. N. Engl. J. Med. 383, 2333–2344 (2020). [PMID: 33085857]
  31. Takahashi, T., Luzum, J. A., Nicol, M. R. & Jacobson, P. A. Pharmacogenomics of COVID-19 therapies. npj Genom. Med. 5, 35 (2020). [PMID: 32864162]
  32. Zhao, R. et al. Early detection of SARS-CoV-2 antibodies in COVID-19 patients as a serologic marker of infection. Clin. Infect. Dis. 71, 2066–2072 (2020).
  33. Zhao, J. et al. Antibody responses to SARS-CoV-2 in patients with novel coronavirus disease 2019. Clin. Infect. Dis. 71, 2027–2034 (2020). [PMID: 32221519]
  34. Chen, W. et al. SARS-CoV-2 neutralizing antibody levels are correlated with severity of COVID-19 pneumonia. Biomed. Pharmacother. 130, 110629–110629 (2020). [PMID: 33406577]
  35. Liu, S. T. H. et al. Convalescent plasma treatment of severe COVID-19: a propensity score–matched control study. Nat. Med. 26, 1708–1713 (2020). [PMID: 32934372]
  36. McClain, M. T. et al. A blood-based host gene expression assay for early detection of respiratory viral infection: an index-cluster prospective cohort study. Lancet Infect. Dis. S1473-3099, 30486–2 (2020).
  37. Xiong, Y. et al. Transcriptomic characteristics of bronchoalveolar lavage fluid and peripheral blood mononuclear cells in COVID-19 patients. Emerg. Microbes Infect. 9, 761–770 (2020). [PMID: 32228226]
  38. Yu, K. et al. Dysregulated adaptive immune response contributes to severe COVID-19. Cell Res. 30, 814–816 (2020). [PMID: 32759967]
  39. He, X. et al. Temporal dynamics in viral shedding and transmissibility of COVID-19. Nat. Med. 26, 672–675 (2020). [PMID: 32296168]
  40. Gupta, R. K. et al. Concise whole blood transcriptional signatures for incipient tuberculosis: a systematic review and patient-level pooled meta-analysis. Lancet Respiratory Med. 8, 395–406 (2020). [DOI: 10.1016/S2213-2600(19)30282-6]
  41. Mayhew, M. B. et al. A generalizable 29-mRNA neural-network classifier for acute bacterial and viral infections. Nat. Commun. 11, 1177 (2020). [PMID: 32132525]
  42. McCall, M. N., Bolstad, B. M. & Irizarry, R. A. Frozen robust multiarray analysis (fRMA). Biostatistics 11, 242–253 (2010). [PMID: 20097884]
  43. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013). [PMID: 23104886]
  44. Robinson, M. D. & Oshlack, A. A scaling normalization method for differential expression analysis of RNA-seq data. Genome Biol. 11, R25 (2010). [PMID: 20196867]
  45. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015). [PMID: 25605792]
  46. Friedman, J., Hastie, T. & Tibshirani, R. Regularization paths for generalized linear models via coordinate descent. J. Stat. Softw. 33, 1–22 (2010). [PMID: 20808728]
  47. Newman, A. M. et al. Robust enumeration of cell subsets from tissue expression profiles. Nat. Methods 12, 453–457 (2015). [PMID: 25822800]

Grants

  1. UM1 AI104681/NIAID NIH HHS
  2. UC6 AI058607/NIAID NIH HHS
  3. K08 HL130557/NHLBI NIH HHS
  4. T32 AI100851/NIAID NIH HHS
  5. U01 AI066569/NIAID NIH HHS
  6. 75N93019C00015/NIAID NIH HHS

MeSH Term

COVID-19
Cytokines
Gene Expression Profiling
Host-Pathogen Interactions
Humans
Influenza, Human
Leukocytes, Mononuclear
Pneumonia, Bacterial
SARS-CoV-2
Sequence Analysis, RNA
Signal Transduction
Transcriptome

Chemicals

Cytokines