The Merkel cell polyomavirus minor capsid protein.

The surface of polyomavirus virions is composed of pentameric knobs of the major capsid protein, VP1. In previously studied polyomavirus species, such as SV40, two interior capsid proteins, VP2 and VP3, emerge from the virion to play important roles during the infectious entry process. Translation o...

Full description

Bibliographic Details
Main Authors: Rachel M Schowalter, Christopher B Buck
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2013-01-01
Series:PLoS Pathogens
Online Access:https://www.ncbi.nlm.nih.gov/pmc/articles/pmid/23990782/?tool=EBI
id doaj-de0d2acb4a8d419eb85da9eaa010620d
record_format Article
spelling doaj-de0d2acb4a8d419eb85da9eaa010620d2021-04-21T17:25:00ZengPublic Library of Science (PLoS)PLoS Pathogens1553-73661553-73742013-01-0198e100355810.1371/journal.ppat.1003558The Merkel cell polyomavirus minor capsid protein.Rachel M SchowalterChristopher B BuckThe surface of polyomavirus virions is composed of pentameric knobs of the major capsid protein, VP1. In previously studied polyomavirus species, such as SV40, two interior capsid proteins, VP2 and VP3, emerge from the virion to play important roles during the infectious entry process. Translation of the VP3 protein initiates at a highly conserved Met-Ala-Leu motif within the VP2 open reading frame. Phylogenetic analyses indicate that Merkel cell polyomavirus (MCV or MCPyV) is a member of a divergent clade of polyomaviruses that lack the conserved VP3 N-terminal motif. Consistent with this observation, we show that VP3 is not detectable in MCV-infected cells, VP3 is not found in native MCV virions, and mutation of possible alternative VP3-initiating methionine codons did not significantly affect MCV infectivity in culture. In contrast, VP2 knockout resulted in a >100-fold decrease in native MCV infectivity, despite normal virion assembly, viral DNA packaging, and cell attachment. Although pseudovirus-based experiments confirmed that VP2 plays an essential role for infection of some cell lines, other cell lines were readily transduced by pseudovirions lacking VP2. In cell lines where VP2 was needed for efficient infectious entry, the presence of a conserved myristoyl modification on the N-terminus of VP2 was important for its function. The results show that a single minor capsid protein, VP2, facilitates a post-attachment stage of MCV infectious entry into some, but not all, cell types.https://www.ncbi.nlm.nih.gov/pmc/articles/pmid/23990782/?tool=EBI
collection DOAJ
language English
format Article
sources DOAJ
author Rachel M Schowalter
Christopher B Buck
spellingShingle Rachel M Schowalter
Christopher B Buck
The Merkel cell polyomavirus minor capsid protein.
PLoS Pathogens
author_facet Rachel M Schowalter
Christopher B Buck
author_sort Rachel M Schowalter
title The Merkel cell polyomavirus minor capsid protein.
title_short The Merkel cell polyomavirus minor capsid protein.
title_full The Merkel cell polyomavirus minor capsid protein.
title_fullStr The Merkel cell polyomavirus minor capsid protein.
title_full_unstemmed The Merkel cell polyomavirus minor capsid protein.
title_sort merkel cell polyomavirus minor capsid protein.
publisher Public Library of Science (PLoS)
series PLoS Pathogens
issn 1553-7366
1553-7374
publishDate 2013-01-01
description The surface of polyomavirus virions is composed of pentameric knobs of the major capsid protein, VP1. In previously studied polyomavirus species, such as SV40, two interior capsid proteins, VP2 and VP3, emerge from the virion to play important roles during the infectious entry process. Translation of the VP3 protein initiates at a highly conserved Met-Ala-Leu motif within the VP2 open reading frame. Phylogenetic analyses indicate that Merkel cell polyomavirus (MCV or MCPyV) is a member of a divergent clade of polyomaviruses that lack the conserved VP3 N-terminal motif. Consistent with this observation, we show that VP3 is not detectable in MCV-infected cells, VP3 is not found in native MCV virions, and mutation of possible alternative VP3-initiating methionine codons did not significantly affect MCV infectivity in culture. In contrast, VP2 knockout resulted in a >100-fold decrease in native MCV infectivity, despite normal virion assembly, viral DNA packaging, and cell attachment. Although pseudovirus-based experiments confirmed that VP2 plays an essential role for infection of some cell lines, other cell lines were readily transduced by pseudovirions lacking VP2. In cell lines where VP2 was needed for efficient infectious entry, the presence of a conserved myristoyl modification on the N-terminus of VP2 was important for its function. The results show that a single minor capsid protein, VP2, facilitates a post-attachment stage of MCV infectious entry into some, but not all, cell types.
url https://www.ncbi.nlm.nih.gov/pmc/articles/pmid/23990782/?tool=EBI
work_keys_str_mv AT rachelmschowalter themerkelcellpolyomavirusminorcapsidprotein
AT christopherbbuck themerkelcellpolyomavirusminorcapsidprotein
AT rachelmschowalter merkelcellpolyomavirusminorcapsidprotein
AT christopherbbuck merkelcellpolyomavirusminorcapsidprotein
_version_ 1714666211414900736