Evidence for vagal sensory neural involvement in influenza pathogenesis and disease

PLoS Pathog. 2024 Apr 16;20(4):e1011635. doi: 10.1371/journal.ppat.1011635. eCollection 2024 Apr.

Abstract

Influenza A virus (IAV) is a common respiratory pathogen and a global cause of significant and often severe morbidity. Although inflammatory immune responses to IAV infections are well described, little is known about how neuroimmune processes contribute to IAV pathogenesis. In the present study, we employed surgical, genetic, and pharmacological approaches to manipulate pulmonary vagal sensory neuron innervation and activity in the lungs to explore potential crosstalk between pulmonary sensory neurons and immune processes. Intranasal inoculation of mice with H1N1 strains of IAV resulted in stereotypical antiviral lung inflammation and tissue pathology, changes in breathing, loss of body weight and other clinical signs of severe IAV disease. Unilateral cervical vagotomy and genetic ablation of pulmonary vagal sensory neurons had a moderate effect on the pulmonary inflammation induced by IAV infection, but significantly worsened clinical disease presentation. Inhibition of pulmonary vagal sensory neuron activity via inhalation of the charged sodium channel blocker, QX-314, resulted in a moderate decrease in lung pathology, but again this was accompanied by a paradoxical worsening of clinical signs. Notably, vagal sensory ganglia neuroinflammation was induced by IAV infection and this was significantly potentiated by QX-314 administration. This vagal ganglia hyperinflammation was characterized by alterations in IAV-induced host defense gene expression, increased neuropeptide gene and protein expression, and an increase in the number of inflammatory cells present within the ganglia. These data suggest that pulmonary vagal sensory neurons play a role in the regulation of the inflammatory process during IAV infection and suggest that vagal neuroinflammation may be an important contributor to IAV pathogenesis and clinical presentation. Targeting these pathways could offer therapeutic opportunities to treat IAV-induced morbidity and mortality.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Female
  • Influenza A Virus, H1N1 Subtype*
  • Influenza, Human / virology
  • Lung / pathology
  • Lung / virology
  • Male
  • Mice
  • Mice, Inbred C57BL
  • Orthomyxoviridae Infections* / immunology
  • Orthomyxoviridae Infections* / pathology
  • Orthomyxoviridae Infections* / virology
  • Sensory Receptor Cells* / pathology
  • Sensory Receptor Cells* / virology
  • Vagus Nerve* / pathology
  • Vagus Nerve* / virology

Grants and funding

This study was funded, in part, by grants obtained by A.E.M and S.B.M (2020/GNT2002765) from the National Health and Medical Research Council of Australia, and B.Y.C and A.E.M from the University of Melbourne, Australia. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.