Abstract

Virus-induced inflammation plays a critical role in determining the clinical outcome of an acute respiratory virus infection. We have shown previously that the administration of immunobiotic Lactobacillus plantarum (Lp) directly to the respiratory tract prevents lethal inflammatory responses to subsequent infection with a mouse respiratory virus pathogen. While Lp-mediated protective responses involve non-redundant contributions of both Toll-like receptor 2 (TLR2) and NOD2, the cellular basis of these findings remains unclear. Here, we address the impact of Lp and its capacity to suppress inflammation in virus-infected respiratory epithelial cells in two cell culture models. We found that both MLE-12 cells and polarized mouse tracheal epithelial cells (mTECs) were susceptible to infection with Influenza A and released proinflammatory cytokines, including CCL2, CCL5, CXCL1, and CXCL10, in response to replicating virus. MLE-12 cells express NOD2 (81 ± 6.3%) and TLR2 (19 ± 4%), respond to Lp, and are TLR2-specific, but not NOD2-specific, biochemical agonists. By contrast, we found that mTECs express NOD2 (81 ± 17%) but minimal TLR2 (0.93 ± 0.58%); nonetheless, mTECs respond to Lp and the TLR2 agonist, Pam2CSK4, but not NOD2 agonists or the bifunctional TLR2-NOD2 agonist, CL-429. Although MLE-12 cells and mTECS were both activated by Lp, little to no cytokine suppression was observed in response to Lp followed by virus infection via a protocol that replicated experimental conditions that were effective in vivo. Further study and a more complex approach may be required to reveal critical factors that suppress virus-induced inflammatory responses.

Highlights

  • Influenza (Inf) and Respiratory Syncytial Virus (RSV) together with Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) are currently the most important respiratory virus pathogens currently circulating worldwide

  • As neither MLE-12 cells nor mouse tracheal epithelial cells (mTECs) were susceptible to infection with pneumonia virus of mice (PVM), we focused this study on inflammatory responses to infection with Influenza A (Inf A)

  • We have focused on outcomes associated with these specific cytokines as they have been featured in reports documenting proinflammatory responses to Inf A, RSV, PVM, and SARS-CoV-2 [8,9,10,11,12,13,14,15,44]

Read more

Summary

Introduction

Influenza (Inf) and Respiratory Syncytial Virus (RSV) together with Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) are currently the most important respiratory virus pathogens currently circulating worldwide. Virus-induced inflammation plays a critical role in determining the clinical outcome of acute respiratory virus infection [7,8,9,10,11,12]. PVM is a rodent-specific virus of the same virus family (Pneumoviridae) and genus (Pneumovirus) as RSV; our acute infection model with PVM phenocopies the severe outcomes that have been described in RSV-infected human infants and children [1,16]. Among the findings that suggest a crucial role for the inflammatory response in the pathogenesis of PVM disease, Walsh and colleagues [17] reported that administration of a sphingosine-1-phosphate (S1P) receptor 1 (S1P1R) agonist blunted the cytokine storm and provided protection against the lethal sequelae of PVM infection. Bonville and colleagues [18,19] documented a similar role for the chemokine CCL3, and Bondue and colleagues [20] identified the chemerin receptor ChemR23 as a critical factor underlying inflammatory pathogenesis

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call