Streptolysin S promotes programmed cell death and enhances inflammatory signaling in epithelial keratinocytes during group A Streptococcus infection

RA Flaherty, JM Puricelli, DL Higashi… - Infection and …, 2015 - Am Soc Microbiol
RA Flaherty, JM Puricelli, DL Higashi, CJ Park, SW Lee
Infection and immunity, 2015Am Soc Microbiol
Streptococcus pyogenes, or group A Streptococcus (GAS), is a pathogen that causes a
multitude of human diseases from pharyngitis to severe infections such as toxic shock
syndrome and necrotizing fasciitis. One of the primary virulence factors produced by GAS is
the peptide toxin streptolysin S (SLS). In addition to its well-recognized role as a cytolysin,
recent evidence has indicated that SLS may influence host cell signaling pathways at
sublytic concentrations during infection. We employed an antibody array-based approach to …
Abstract
Streptococcus pyogenes, or group A Streptococcus (GAS), is a pathogen that causes a multitude of human diseases from pharyngitis to severe infections such as toxic shock syndrome and necrotizing fasciitis. One of the primary virulence factors produced by GAS is the peptide toxin streptolysin S (SLS). In addition to its well-recognized role as a cytolysin, recent evidence has indicated that SLS may influence host cell signaling pathways at sublytic concentrations during infection. We employed an antibody array-based approach to comprehensively identify global host cell changes in human epithelial keratinocytes in response to the SLS toxin. We identified key SLS-dependent host responses, including the initiation of specific programmed cell death and inflammatory cascades with concomitant downregulation of Akt-mediated cytoprotection. Significant signaling responses identified by our array analysis were confirmed using biochemical and protein identification methods. To further demonstrate that the observed SLS-dependent host signaling changes were mediated primarily by the secreted toxin, we designed a Transwell infection system in which direct bacterial attachment to host cells was prevented, while secreted factors were allowed access to host cells. The results using this approach were consistent with our direct infection studies and reveal that SLS is a bacterial toxin that does not require bacterial attachment to host cells for activity. In light of these findings, we propose that the production of SLS by GAS during skin infection promotes invasive outcomes by triggering programmed cell death and inflammatory cascades in host cells to breach the keratinocyte barrier for dissemination into deeper tissues.
American Society for Microbiology