author_facet Riolobos, Laura
Valle, Noelia
Hernando, Eva
Maroto, Beatriz
Kann, Michael
Almendral, José M.
Riolobos, Laura
Valle, Noelia
Hernando, Eva
Maroto, Beatriz
Kann, Michael
Almendral, José M.
author Riolobos, Laura
Valle, Noelia
Hernando, Eva
Maroto, Beatriz
Kann, Michael
Almendral, José M.
spellingShingle Riolobos, Laura
Valle, Noelia
Hernando, Eva
Maroto, Beatriz
Kann, Michael
Almendral, José M.
Journal of Virology
Viral Oncolysis That Targets Raf-1 Signaling Control of Nuclear Transport
Virology
Insect Science
Immunology
Microbiology
author_sort riolobos, laura
spelling Riolobos, Laura Valle, Noelia Hernando, Eva Maroto, Beatriz Kann, Michael Almendral, José M. 0022-538X 1098-5514 American Society for Microbiology Virology Insect Science Immunology Microbiology http://dx.doi.org/10.1128/jvi.01550-09 <jats:title>ABSTRACT</jats:title> <jats:p> The central role of Raf protein kinase isoforms in human cancer demands specific anti-Raf therapeutic inhibitors. Parvoviruses are currently used in experimental cancer therapy due to their natural oncotropism and lytic life cycle. In searching for mechanisms underlying parvovirus oncolysis, we found that trimers of the major structural protein (VP) of the parvovirus minute virus of mice (MVM), which have to be imported into the nucleus for capsid assembly, undergo phosphorylation by the Raf-1 kinase. Purified Raf-1 phosphorylated the capsid subunits <jats:italic>in vitro</jats:italic> to the two-dimensional pattern found in natural MVM infections. VP trimers isolated from mammalian cells translocated into the nucleus of digitonin-permeabilized human cells. In contrast, VP trimers isolated from insect cells, which are devoid of Raf-1, were neither phosphorylated nor imported into the mammalian nucleus. However, the coexpression of a constitutively active Raf-1 kinase in insect cells restored VP trimer phosphorylation and nuclear transport competence. In MVM-infected normal and transformed cells, Raf-1 inhibition resulted in cytoplasmic retention of capsid proteins, preventing their nuclear assembly and progeny virus maturation. The level of Raf-1 activity in cancer cells was consistent with the extent of VP specific phosphorylation and with the permissiveness to MVM infection. Thus, Raf-1 control of nuclear translocation of MVM capsid assembly intermediates provides a novel target for viral oncolysis. MVM may reinforce specific therapies against frequent human cancers with deregulated Raf signaling. </jats:p> Viral Oncolysis That Targets Raf-1 Signaling Control of Nuclear Transport Journal of Virology
doi_str_mv 10.1128/jvi.01550-09
facet_avail Online
Free
finc_class_facet Medizin
Biologie
format ElectronicArticle
fullrecord blob:ai-49-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTEyOC9qdmkuMDE1NTAtMDk
id ai-49-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTEyOC9qdmkuMDE1NTAtMDk
institution DE-D275
DE-Bn3
DE-Brt1
DE-Zwi2
DE-D161
DE-Gla1
DE-Zi4
DE-15
DE-Pl11
DE-Rs1
DE-105
DE-14
DE-Ch1
DE-L229
imprint American Society for Microbiology, 2010
imprint_str_mv American Society for Microbiology, 2010
issn 0022-538X
1098-5514
issn_str_mv 0022-538X
1098-5514
language English
mega_collection American Society for Microbiology (CrossRef)
match_str riolobos2010viraloncolysisthattargetsraf1signalingcontrolofnucleartransport
publishDateSort 2010
publisher American Society for Microbiology
recordtype ai
record_format ai
series Journal of Virology
source_id 49
title Viral Oncolysis That Targets Raf-1 Signaling Control of Nuclear Transport
title_unstemmed Viral Oncolysis That Targets Raf-1 Signaling Control of Nuclear Transport
title_full Viral Oncolysis That Targets Raf-1 Signaling Control of Nuclear Transport
title_fullStr Viral Oncolysis That Targets Raf-1 Signaling Control of Nuclear Transport
title_full_unstemmed Viral Oncolysis That Targets Raf-1 Signaling Control of Nuclear Transport
title_short Viral Oncolysis That Targets Raf-1 Signaling Control of Nuclear Transport
title_sort viral oncolysis that targets raf-1 signaling control of nuclear transport
topic Virology
Insect Science
Immunology
Microbiology
url http://dx.doi.org/10.1128/jvi.01550-09
publishDate 2010
physical 2090-2099
description <jats:title>ABSTRACT</jats:title> <jats:p> The central role of Raf protein kinase isoforms in human cancer demands specific anti-Raf therapeutic inhibitors. Parvoviruses are currently used in experimental cancer therapy due to their natural oncotropism and lytic life cycle. In searching for mechanisms underlying parvovirus oncolysis, we found that trimers of the major structural protein (VP) of the parvovirus minute virus of mice (MVM), which have to be imported into the nucleus for capsid assembly, undergo phosphorylation by the Raf-1 kinase. Purified Raf-1 phosphorylated the capsid subunits <jats:italic>in vitro</jats:italic> to the two-dimensional pattern found in natural MVM infections. VP trimers isolated from mammalian cells translocated into the nucleus of digitonin-permeabilized human cells. In contrast, VP trimers isolated from insect cells, which are devoid of Raf-1, were neither phosphorylated nor imported into the mammalian nucleus. However, the coexpression of a constitutively active Raf-1 kinase in insect cells restored VP trimer phosphorylation and nuclear transport competence. In MVM-infected normal and transformed cells, Raf-1 inhibition resulted in cytoplasmic retention of capsid proteins, preventing their nuclear assembly and progeny virus maturation. The level of Raf-1 activity in cancer cells was consistent with the extent of VP specific phosphorylation and with the permissiveness to MVM infection. Thus, Raf-1 control of nuclear translocation of MVM capsid assembly intermediates provides a novel target for viral oncolysis. MVM may reinforce specific therapies against frequent human cancers with deregulated Raf signaling. </jats:p>
container_issue 4
container_start_page 2090
container_title Journal of Virology
container_volume 84
format_de105 Article, E-Article
format_de14 Article, E-Article
format_de15 Article, E-Article
format_de520 Article, E-Article
format_de540 Article, E-Article
format_dech1 Article, E-Article
format_ded117 Article, E-Article
format_degla1 E-Article
format_del152 Buch
format_del189 Article, E-Article
format_dezi4 Article
format_dezwi2 Article, E-Article
format_finc Article, E-Article
format_nrw Article, E-Article
_version_ 1792334182864650240
geogr_code not assigned
last_indexed 2024-03-01T14:23:49.236Z
geogr_code_person not assigned
openURL url_ver=Z39.88-2004&ctx_ver=Z39.88-2004&ctx_enc=info%3Aofi%2Fenc%3AUTF-8&rfr_id=info%3Asid%2Fvufind.svn.sourceforge.net%3Agenerator&rft.title=Viral+Oncolysis+That+Targets+Raf-1+Signaling+Control+of+Nuclear+Transport&rft.date=2010-02-15&genre=article&issn=1098-5514&volume=84&issue=4&spage=2090&epage=2099&pages=2090-2099&jtitle=Journal+of+Virology&atitle=Viral+Oncolysis+That+Targets+Raf-1+Signaling+Control+of+Nuclear+Transport&aulast=Almendral&aufirst=Jose%CC%81+M.&rft_id=info%3Adoi%2F10.1128%2Fjvi.01550-09&rft.language%5B0%5D=eng
SOLR
_version_ 1792334182864650240
author Riolobos, Laura, Valle, Noelia, Hernando, Eva, Maroto, Beatriz, Kann, Michael, Almendral, José M.
author_facet Riolobos, Laura, Valle, Noelia, Hernando, Eva, Maroto, Beatriz, Kann, Michael, Almendral, José M., Riolobos, Laura, Valle, Noelia, Hernando, Eva, Maroto, Beatriz, Kann, Michael, Almendral, José M.
author_sort riolobos, laura
container_issue 4
container_start_page 2090
container_title Journal of Virology
container_volume 84
description <jats:title>ABSTRACT</jats:title> <jats:p> The central role of Raf protein kinase isoforms in human cancer demands specific anti-Raf therapeutic inhibitors. Parvoviruses are currently used in experimental cancer therapy due to their natural oncotropism and lytic life cycle. In searching for mechanisms underlying parvovirus oncolysis, we found that trimers of the major structural protein (VP) of the parvovirus minute virus of mice (MVM), which have to be imported into the nucleus for capsid assembly, undergo phosphorylation by the Raf-1 kinase. Purified Raf-1 phosphorylated the capsid subunits <jats:italic>in vitro</jats:italic> to the two-dimensional pattern found in natural MVM infections. VP trimers isolated from mammalian cells translocated into the nucleus of digitonin-permeabilized human cells. In contrast, VP trimers isolated from insect cells, which are devoid of Raf-1, were neither phosphorylated nor imported into the mammalian nucleus. However, the coexpression of a constitutively active Raf-1 kinase in insect cells restored VP trimer phosphorylation and nuclear transport competence. In MVM-infected normal and transformed cells, Raf-1 inhibition resulted in cytoplasmic retention of capsid proteins, preventing their nuclear assembly and progeny virus maturation. The level of Raf-1 activity in cancer cells was consistent with the extent of VP specific phosphorylation and with the permissiveness to MVM infection. Thus, Raf-1 control of nuclear translocation of MVM capsid assembly intermediates provides a novel target for viral oncolysis. MVM may reinforce specific therapies against frequent human cancers with deregulated Raf signaling. </jats:p>
doi_str_mv 10.1128/jvi.01550-09
facet_avail Online, Free
finc_class_facet Medizin, Biologie
format ElectronicArticle
format_de105 Article, E-Article
format_de14 Article, E-Article
format_de15 Article, E-Article
format_de520 Article, E-Article
format_de540 Article, E-Article
format_dech1 Article, E-Article
format_ded117 Article, E-Article
format_degla1 E-Article
format_del152 Buch
format_del189 Article, E-Article
format_dezi4 Article
format_dezwi2 Article, E-Article
format_finc Article, E-Article
format_nrw Article, E-Article
geogr_code not assigned
geogr_code_person not assigned
id ai-49-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTEyOC9qdmkuMDE1NTAtMDk
imprint American Society for Microbiology, 2010
imprint_str_mv American Society for Microbiology, 2010
institution DE-D275, DE-Bn3, DE-Brt1, DE-Zwi2, DE-D161, DE-Gla1, DE-Zi4, DE-15, DE-Pl11, DE-Rs1, DE-105, DE-14, DE-Ch1, DE-L229
issn 0022-538X, 1098-5514
issn_str_mv 0022-538X, 1098-5514
language English
last_indexed 2024-03-01T14:23:49.236Z
match_str riolobos2010viraloncolysisthattargetsraf1signalingcontrolofnucleartransport
mega_collection American Society for Microbiology (CrossRef)
physical 2090-2099
publishDate 2010
publishDateSort 2010
publisher American Society for Microbiology
record_format ai
recordtype ai
series Journal of Virology
source_id 49
spelling Riolobos, Laura Valle, Noelia Hernando, Eva Maroto, Beatriz Kann, Michael Almendral, José M. 0022-538X 1098-5514 American Society for Microbiology Virology Insect Science Immunology Microbiology http://dx.doi.org/10.1128/jvi.01550-09 <jats:title>ABSTRACT</jats:title> <jats:p> The central role of Raf protein kinase isoforms in human cancer demands specific anti-Raf therapeutic inhibitors. Parvoviruses are currently used in experimental cancer therapy due to their natural oncotropism and lytic life cycle. In searching for mechanisms underlying parvovirus oncolysis, we found that trimers of the major structural protein (VP) of the parvovirus minute virus of mice (MVM), which have to be imported into the nucleus for capsid assembly, undergo phosphorylation by the Raf-1 kinase. Purified Raf-1 phosphorylated the capsid subunits <jats:italic>in vitro</jats:italic> to the two-dimensional pattern found in natural MVM infections. VP trimers isolated from mammalian cells translocated into the nucleus of digitonin-permeabilized human cells. In contrast, VP trimers isolated from insect cells, which are devoid of Raf-1, were neither phosphorylated nor imported into the mammalian nucleus. However, the coexpression of a constitutively active Raf-1 kinase in insect cells restored VP trimer phosphorylation and nuclear transport competence. In MVM-infected normal and transformed cells, Raf-1 inhibition resulted in cytoplasmic retention of capsid proteins, preventing their nuclear assembly and progeny virus maturation. The level of Raf-1 activity in cancer cells was consistent with the extent of VP specific phosphorylation and with the permissiveness to MVM infection. Thus, Raf-1 control of nuclear translocation of MVM capsid assembly intermediates provides a novel target for viral oncolysis. MVM may reinforce specific therapies against frequent human cancers with deregulated Raf signaling. </jats:p> Viral Oncolysis That Targets Raf-1 Signaling Control of Nuclear Transport Journal of Virology
spellingShingle Riolobos, Laura, Valle, Noelia, Hernando, Eva, Maroto, Beatriz, Kann, Michael, Almendral, José M., Journal of Virology, Viral Oncolysis That Targets Raf-1 Signaling Control of Nuclear Transport, Virology, Insect Science, Immunology, Microbiology
title Viral Oncolysis That Targets Raf-1 Signaling Control of Nuclear Transport
title_full Viral Oncolysis That Targets Raf-1 Signaling Control of Nuclear Transport
title_fullStr Viral Oncolysis That Targets Raf-1 Signaling Control of Nuclear Transport
title_full_unstemmed Viral Oncolysis That Targets Raf-1 Signaling Control of Nuclear Transport
title_short Viral Oncolysis That Targets Raf-1 Signaling Control of Nuclear Transport
title_sort viral oncolysis that targets raf-1 signaling control of nuclear transport
title_unstemmed Viral Oncolysis That Targets Raf-1 Signaling Control of Nuclear Transport
topic Virology, Insect Science, Immunology, Microbiology
url http://dx.doi.org/10.1128/jvi.01550-09