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

dc.contributor.authorCarmichael, Jillian C.
dc.contributor.authorYokota, Hiroki
dc.contributor.authorCraven, Rebecca C.
dc.contributor.authorSchmitt, Anthony
dc.contributor.authorWills, John W.
dc.contributor.departmentBiomedical Engineering, School of Engineering and Technologyen_US
dc.date.accessioned2019-02-14T17:36:15Z
dc.date.available2019-02-14T17:36:15Z
dc.date.issued2018-05-09
dc.description.abstractAll 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.en_US
dc.eprint.versionFinal published versionen_US
dc.identifier.citationCarmichael, J. C., Yokota, H., Craven, R. C., Schmitt, A., & Wills, J. W. (2018). The HSV-1 mechanisms of cell-to-cell spread and fusion are critically dependent on host PTP1B. PLoS pathogens, 14(5), e1007054. doi:10.1371/journal.ppat.1007054en_US
dc.identifier.urihttps://hdl.handle.net/1805/18374
dc.language.isoen_USen_US
dc.publisherPublic Library of Scienceen_US
dc.relation.isversionof10.1371/journal.ppat.1007054en_US
dc.relation.journalPLoS pathogensen_US
dc.rightsAttribution 3.0 United States
dc.rights.urihttp://creativecommons.org/licenses/by/3.0/us/
dc.sourcePMCen_US
dc.subjectCell Lineen_US
dc.subjectCercopithecus aethiopsen_US
dc.subjectCinnamatesen_US
dc.subjectGiant Cellsen_US
dc.subjectHerpesvirus 1, Humanen_US
dc.subjectIntercellular Junctionsen_US
dc.subjectMass Spectrometryen_US
dc.subjectProtein Tyrosine Phosphatase, Non-Receptor Type 1en_US
dc.subjectReceptors, Virusen_US
dc.subjectThioureaen_US
dc.subjectVero Cellsen_US
dc.subjectViral Envelope Proteinsen_US
dc.subjectVirus Internalizationen_US
dc.subjectVirus Replicationen_US
dc.titleThe HSV-1 mechanisms of cell-to-cell spread and fusion are critically dependent on host PTP1Ben_US
dc.typeArticleen_US
Files
Original bundle
Now showing 1 - 1 of 1
Loading...
Thumbnail Image
Name:
ppat.1007054.pdf
Size:
9.37 MB
Format:
Adobe Portable Document Format
Description:
Main article
License bundle
Now showing 1 - 1 of 1
No Thumbnail Available
Name:
license.txt
Size:
1.99 KB
Format:
Item-specific license agreed upon to submission
Description: