Partial Attenuation of Respiratory Syncytial Virus with a Deletion of a Small Hydrophobic Gene Is Associated with Elevated Interleukin-1β Responses

J Virol. 2015 Sep;89(17):8974-81. doi: 10.1128/JVI.01070-15. Epub 2015 Jun 17.

Abstract

The small hydrophobic (SH) gene of respiratory syncytial virus (RSV), a major cause of infant hospitalization, encodes a viroporin of unknown function. SH gene knockout virus (RSV ΔSH) is partially attenuated in vivo, but not in vitro, suggesting that the SH protein may have an immunomodulatory role. RSV ΔSH has been tested as a live attenuated vaccine in humans and cattle, and here we demonstrate that it protected against viral rechallenge in mice. We compared the immune response to infection with RSV wild type and RSV ΔSH in vivo using BALB/c mice and in vitro using epithelial cells, neutrophils, and macrophages. Strikingly, the interleukin-1β (IL-1β) response to RSV ΔSH infection was greater than to wild-type RSV, in spite of a decreased viral load, and when IL-1β was blocked in vivo, the viral load returned to wild-type levels. A significantly greater IL-1β response to RSV ΔSH was also detected in vitro, with higher-magnitude responses in neutrophils and macrophages than in epithelial cells. Depleting macrophages (with clodronate liposome) and neutrophils (with anti-Ly6G/1A8) demonstrated the contribution of these cells to the IL-1β response in vivo, the first demonstration of neutrophilic IL-1β production in response to viral lung infection. In this study, we describe an increased IL-1β response to RSV ΔSH, which may explain the attenuation in vivo and supports targeting the SH gene in live attenuated vaccines.

Importance: There is a pressing need for a vaccine for respiratory syncytial virus (RSV). A number of live attenuated RSV vaccine strains have been developed in which the small hydrophobic (SH) gene has been deleted, even though the function of the SH protein is unknown. The structure of the SH protein has recently been solved, showing it is a pore-forming protein (viroporin). Here, we demonstrate that the IL-1β response to RSV ΔSH is greater in spite of a lower viral load, which contributes to the attenuation in vivo. This potentially suggests a novel method by which viruses can evade the host response. As all Pneumovirinae and some Paramyxovirinae carry similar SH genes, this new understanding may also enable the development of live attenuated vaccines for both RSV and other members of the Paramyxoviridae.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Cell Line
  • Epithelial Cells / immunology
  • Epithelial Cells / virology
  • Female
  • Gene Deletion
  • Gene Knockout Techniques
  • Humans
  • Interleukin-1beta / biosynthesis
  • Interleukin-1beta / immunology*
  • Macrophages / immunology
  • Macrophages / virology
  • Mice
  • Mice, Inbred BALB C
  • Neutrophils / immunology
  • Neutrophils / virology
  • Respiratory Syncytial Virus Infections / immunology*
  • Respiratory Syncytial Virus Infections / virology
  • Respiratory Syncytial Virus Vaccines / immunology
  • Respiratory Syncytial Viruses / genetics*
  • Respiratory Syncytial Viruses / growth & development
  • Respiratory Syncytial Viruses / immunology*
  • Retroviridae Proteins, Oncogenic / genetics*
  • Vaccination
  • Vaccines, Attenuated / immunology
  • Viral Load / immunology

Substances

  • IL1B protein, mouse
  • Interleukin-1beta
  • Respiratory Syncytial Virus Vaccines
  • Retroviridae Proteins, Oncogenic
  • Vaccines, Attenuated
  • small hydrophobic protein, virus