The virological synapse facilitates herpes simplex virus entry into T cells

M Aubert, M Yoon, DD Sloan, PG Spear… - Journal of …, 2009 - Am Soc Microbiol
M Aubert, M Yoon, DD Sloan, PG Spear, KR Jerome
Journal of virology, 2009Am Soc Microbiol
The virological synapse (VS) is a specialized molecular structure that facilitates the transfer
of certain lymphotropic viruses into uninfected T cells. However, the role of the VS in the
transfer of nonlymphotropic viruses into T cells is unknown. Herpes simplex virus (HSV) has
been shown in vitro to infect T cells and modulate T-cell receptor function, thereby
suppressing T-cell antiviral function. However, whether such infection of T cells occurs in
vivo is unknown. Here, we examined whether T-cell infection could be observed in human …
Abstract
The virological synapse (VS) is a specialized molecular structure that facilitates the transfer of certain lymphotropic viruses into uninfected T cells. However, the role of the VS in the transfer of nonlymphotropic viruses into T cells is unknown. Herpes simplex virus (HSV) has been shown in vitro to infect T cells and modulate T-cell receptor function, thereby suppressing T-cell antiviral function. However, whether such infection of T cells occurs in vivo is unknown. Here, we examined whether T-cell infection could be observed in human HSV disease and investigated the mechanism of HSV entry into T cells. We found that HSV-infected T cells were readily detectable during human disease, suggesting that infection and modulation of T-cell function plays a role in human immunopathology. HSV infection of both CD4+ and CD8+ T cells occurred much more efficiently via direct cell-to-cell spread from infected fibroblasts than by cell-free virus. Activation of T cells increased their permissivity to HSV infection. Cell-to-cell spread to T cells did not require HSV glycoproteins E and I (gE and gI), which are critical for cell-to-cell spread between epithelial cells. Transfer of HSV to T cells required gD, and the four known entry receptors appear to be contributing to viral entry, with a dominant role for the herpesvirus entry mediator and nectin-1. VS-like structures enriched in activated lymphocyte function-associated antigen 1 (LFA-1) were observed at the point of contact between HSV-infected fibroblasts and T cells. Consistent with spread occurring via the VS, transfer of HSV was increased by activation of LFA-1, and cell-to-cell spread could be inhibited by antibodies to LFA-1 or gD. Taken together, these results constitute the first demonstration of VS-dependent cell-to-cell spread for a predominantly nonlymphotropic virus. Furthermore, they support an important role for infection and immunomodulation of T cells in clinical human disease. Targeting of the VS might allow selective immunopotentiation during infections with HSV or other nonlymphotropic viruses.
American Society for Microbiology