[HTML][HTML] The HSV-1 mechanisms of cell-to-cell spread and fusion are critically dependent on host PTP1B

JC Carmichael, H Yokota, RC Craven, A Schmitt… - PLoS …, 2018 - journals.plos.org
JC Carmichael, H Yokota, RC Craven, A Schmitt, JW Wills
PLoS pathogens, 2018journals.plos.org
All herpesviruses have mechanisms for passing through cell junctions, which exclude
neutralizing antibodies and offer a clear path to neighboring, uninfected cells. In the case of
herpes simplex virus type 1 (HSV-1), direct cell-to-cell transmission takes place between
epithelial cells and sensory neurons, where latency is established. The spreading
mechanism is poorly understood, but mutations in four different HSV-1 genes can
dysregulate it, causing neighboring cells to fuse to produce syncytia. Because the host …
All herpesviruses have mechanisms for passing through cell junctions, which exclude neutralizing antibodies and offer a clear path to neighboring, uninfected cells. In the case of herpes simplex virus type 1 (HSV-1), direct cell-to-cell transmission takes place between epithelial cells and sensory neurons, where latency is established. The spreading mechanism is poorly understood, but mutations in four different HSV-1 genes can dysregulate it, causing neighboring cells to fuse to produce syncytia. Because the host proteins involved are largely unknown (other than the virus entry receptor), we were intrigued by an earlier discovery that cells infected with wild-type HSV-1 will form syncytia when treated with salubrinal. A biotinylated derivative of this drug was used to pull down cellular complexes, which were analyzed by mass spectrometry. One candidate was a protein tyrosine phosphatase (PTP1B), and although it ultimately proved not to be the target of salubrinal, it was found to be critical for the mechanism of cell-to-cell spread. In particular, a highly specific inhibitor of PTP1B (CAS 765317-72-4) blocked salubrinal-induced fusion, and by itself resulted in a dramatic reduction in the ability of HSV-1 to spread in the presence of neutralizing antibodies. The importance of this phosphatase was confirmed in the absence of drugs by using PTP1B-/- cells. Importantly, replication assays showed that virus titers were unaffected when PTP1B was inhibited or absent. Only cell-to-cell spread was altered. We also examined the effects of salubrinal and the PTP1B inhibitor on the four Syn mutants of HSV-1, and strikingly different responses were found. That is, both drugs individually enhanced fusion for some mutants and reduced fusion for others. PTP1B is the first host factor identified to be specifically required for cell-to-cell spread, and it may be a therapeutic target for preventing HSV-1 reactivation disease.
PLOS