Abstract

Endothelial cells have been considered the central regulators of cytokine storm in the respiratory system during influenza virus infection. Studies have found that elevated autophagy could be an essential component of viral pathogenesis in influenza infection. However, few studies have been performed to examine whether autophagy occurs in human pulmonary endothelial cells (HPMECs). In addition, specific mechanisms about how inflammatory responses are regulated in the endothelial cells remain unclear. We hypothesized that infection of influenza A viruses subtypes H1N1 and H9N2 triggered autophagy, which played an important role in the induction of proinflammatory cytokines, both in human lung epithelial A549 cells and in HPMECs. In this report, we showed our evidence that blockage of autophagy significantly inhibited influenza virus-induced proinflammatory responses and suppressed viral replication. Our data indicated that the inhibition of the cytokine response and viral replication was affected by increasing the expression of endothelial sphingosine 1-phosphate receptor 1 (S1PR1), which might be through the regulation of NF-κB signaling. Overexpression of S1PR1 decreased p65 phosphorylation and translocation into the nucleus. Furthermore, we demonstrated that S1PR1 stimulation inhibited Akt-mTOR signaling, which might contribute to activation of autophagy in HPMECs. Thus, our study provides knowledge crucial to better understanding novel mechanisms underlying the S1PR1-mediated attenuation of cytokine amplification in the pulmonary system during influenza virus infection.

Highlights

  • Emerging and re-emerging infections of influenza A viruses (IAV) have posed considerable threats to public health, in particular the ones of highly pathogenic avian influenza with early exacerbation and dysregulation of innate cellular and cytokine responses, or cytokine storm [1,2,3]

  • We have demonstrated that influenza A virus was able to trigger autophagy in human pulmonary endothelial cells (HPMECs)

  • We showed that activation of the endothelial S1P1 signaling led to suppression of autophagy, which could result in the decreased induction of proinflammatory cytokines and chemokines and reduced virus replication

Read more

Summary

Introduction

Emerging and re-emerging infections of influenza A viruses (IAV) have posed considerable threats to public health, in particular the ones of highly pathogenic avian influenza with early exacerbation and dysregulation of innate cellular and cytokine responses, or cytokine storm [1,2,3]. Mounting evidence has identified pulmonary endothelial cells as central regulators of the cytokine storm, which challenges the long-standing assumption that alveolar epithelial cells are the main target cell type in viral pathogenesis in influenza [4]. Accumulating data have revealed that elevated autophagy induced by IAV mediates alveolar epithelial cell death and is important for replication of IAV [11,12,13]. We provide evidence that IAV stimulated proinflammatory cytokines and induced autophagy both in human lung epithelial A549 cells and in HPMECs. We demonstrated that over-expressed S1PR1 in pulmonary endothelial cells suppressed autophagy, inhibited the inflammatory responses and virus replication, which might be regulated by suppressing NF-κB signaling. Autophagic pathway affected by S1PR1 signaling in the pulmonary endothelial cells could provide a novel therapeutic target for attenuation of mortality and morbidity in influenza infection

Objectives
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