Impaired antibacterial immune signaling and changes in the lung microbiome precede secondary bacterial pneumonia in COVID-19

Secondary bacterial infections, including ventilator associated pneumonia (VAP), lead to worse clinical outcomes and increased mortality following viral respiratory infections. Critically ill patients with coronavirus disease 2019 (COVID-19) face an elevated risk of VAP, although susceptibility varies widely. Because mechanisms underlying VAP predisposition remained unknown, we assessed lower respiratory tract host immune responses and microbiome dynamics in 36 patients, including 28 COVID-19 patients, 15 of whom developed VAP, and eight critically ill controls. We employed a combination of tracheal aspirate bulk and single cell RNA sequencing (scRNA-seq). Two days before VAP onset, a lower respiratory transcriptional signature of bacterial infection was observed, characterized by increased expression of neutrophil degranulation, toll-like receptor and cytokine signaling pathways. When assessed at an earlier time point following endotracheal intubation, more than two weeks prior to VAP onset, we observed a striking early impairment in antibacterial innate and adaptive immune signaling that markedly differed from COVID-19 patients who did not develop VAP. scRNA-seq further demonstrated suppressed immune signaling across monocytes/macrophages, neutrophils and T cells. While viral load did not differ at an early post-intubation timepoint, impaired SARS-CoV-2 clearance and persistent interferon signaling characterized the patients who later developed VAP. Longitudinal metatranscriptomic analysis revealed disruption of lung microbiome community composition in patients who developed VAP, providing a connection between dysregulated immune signaling and outgrowth of opportunistic pathogens. Together, these findings demonstrate that COVID-19 patients who develop VAP have impaired antibacterial immune defense weeks before secondary infection onset.

[1]  Gavin J. D. Smith,et al.  Early induction of functional SARS-CoV-2-specific T cells associates with rapid viral clearance and mild disease in COVID-19 patients , 2021, Cell Reports.

[2]  S. Marsch,et al.  Community-acquired and hospital-acquired respiratory tract infection and bloodstream infection in patients hospitalized with COVID-19 pneumonia , 2021, Journal of Intensive Care.

[3]  G. Dougan,et al.  Ventilator-associated pneumonia in critically ill patients with COVID-19 , 2021, Critical Care.

[4]  D. Pestaña,et al.  Nosocomial infections associated to COVID-19 in the intensive care unit: clinical characteristics and outcome , 2020, European journal of clinical microbiology & infectious diseases : official publication of the European Society of Clinical Microbiology.

[5]  N. Neff,et al.  Upper airway gene expression reveals suppressed immune responses to SARS-CoV-2 compared with other respiratory viruses , 2020, Nature Communications.

[6]  R. Schwartz,et al.  Imbalanced Host Response to SARS-CoV-2 Drives Development of COVID-19 , 2020, Cell.

[7]  Ryan King,et al.  IDseq—An open source cloud-based pipeline and analysis service for metagenomic pathogen detection and monitoring , 2020, bioRxiv.

[8]  Kouji Matsushima,et al.  CXCR6 regulates localization of tissue-resident memory CD8 T cells to the airways , 2019, The Journal of experimental medicine.

[9]  Yun-Fang Juan,et al.  Metagenomic next-generation sequencing of samples from pediatric febrile illness in Tororo, Uganda , 2019, PloS one.

[10]  Fan Zhang,et al.  Fast, sensitive, and accurate integration of single cell data with Harmony , 2018, bioRxiv.

[11]  Katherine S. Pollard,et al.  Integrating host response and unbiased microbe detection for lower respiratory tract infection diagnosis in critically ill adults , 2018, Proceedings of the National Academy of Sciences.

[12]  M. Shankar-Hari,et al.  Cell-surface signatures of immune dysfunction risk-stratify critically ill patients: INFECT study , 2018, Intensive Care Medicine.

[13]  A. Mathieu,et al.  Antigen-Induced but Not Innate Memory CD8 T Cells Express NKG2D and Are Recruited to the Lung Parenchyma upon Viral Infection , 2017, The Journal of Immunology.

[14]  Fabian J Theis,et al.  SCANPY: large-scale single-cell gene expression data analysis , 2018, Genome Biology.

[15]  Wouter A. A. de Steenhuijsen Piters,et al.  The microbiota of the respiratory tract: gatekeeper to respiratory health , 2017, Nature Reviews Microbiology.

[16]  Lior Pachter,et al.  Near-optimal probabilistic RNA-seq quantification , 2016, Nature Biotechnology.

[17]  Marc Salit,et al.  Evaluation of the External RNA Controls Consortium (ERCC) reference material using a modified Latin square design , 2016, bioRxiv.

[18]  M. Robinson,et al.  Differential analyses for RNA-seq: transcript-level estimates improve gene-level inferences , 2015, F1000Research.

[19]  M. Robinson,et al.  Differential analyses for RNA-seq: transcript-level estimates improve gene-level inferences. , 2015, F1000Research.

[20]  P. Linsley,et al.  MAST: a flexible statistical framework for assessing transcriptional changes and characterizing heterogeneity in single-cell RNA sequencing data , 2015, Genome Biology.

[21]  W. Huber,et al.  Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2 , 2014, Genome Biology.

[22]  Andreas Krämer,et al.  Causal analysis approaches in Ingenuity Pathway Analysis , 2013, Bioinform..

[23]  S. Johnston,et al.  Outgrowth of the bacterial airway microbiome after rhinovirus exacerbation of chronic obstructive pulmonary disease. , 2013, American journal of respiratory and critical care medicine.

[24]  D. Metzger,et al.  Immune Dysfunction and Bacterial Coinfections following Influenza , 2013, The Journal of Immunology.

[25]  E. Walsh,et al.  Bacterial Complications of Respiratory Tract Viral Illness: A Comprehensive Evaluation , 2013, The Journal of infectious diseases.

[26]  Thomas R. Gingeras,et al.  STAR: ultrafast universal RNA-seq aligner , 2013, Bioinform..

[27]  S. Johnston,et al.  Rhinovirus infection induces degradation of antimicrobial peptides and secondary bacterial infection in chronic obstructive pulmonary disease. , 2012, American journal of respiratory and critical care medicine.

[28]  J. Alcorn,et al.  Influenza A Inhibits Th17-Mediated Host Defense against Bacterial Pneumonia in Mice , 2011, The Journal of Immunology.

[29]  A. Shahangian,et al.  Type I IFNs mediate development of postinfluenza bacterial pneumonia in mice. , 2009, The Journal of clinical investigation.

[30]  Anthony S Fauci,et al.  Predominant role of bacterial pneumonia as a cause of death in pandemic influenza: implications for pandemic influenza preparedness. , 2008, The Journal of infectious diseases.

[31]  B. Lambrecht,et al.  Sustained desensitization to bacterial Toll-like receptor ligands after resolutionof respiratory influenza infection , 2008, The Journal of experimental medicine.

[32]  Eoin L. Brodie,et al.  Loss of Bacterial Diversity during Antibiotic Treatment of Intubated Patients Colonized with Pseudomonas aeruginosa , 2006, Journal of Clinical Microbiology.

[33]  B. Kramer Ventilator-Associated Pneumonia in Critically Ill Patients , 1999, Annals of Internal Medicine.

[34]  K. Takase,et al.  [T cell activation]. , 1995, Ryumachi. [Rheumatism].