A Novel Selective LSD1/KDM1A Inhibitor Epigenetically Blocks Herpes Simplex Virus Lytic Replication and Reactivation from Latency

ABSTRACT Cellular processes requiring access to the DNA genome are regulated by an overlay of epigenetic modifications, including histone modification and chromatin remodeling. Similar to the cellular host, many nuclear DNA viruses that depend upon the host cell’s transcriptional machinery are also...

Descripción completa

Guardado en:
Detalles Bibliográficos
Autores principales: Yu Liang, Debra Quenelle, Jodi L. Vogel, Cristina Mascaro, Alberto Ortega, Thomas M. Kristie
Formato: article
Lenguaje:EN
Publicado: American Society for Microbiology 2013
Materias:
Acceso en línea:https://doaj.org/article/6b6c8ab959fb42f28121b92a4680f7e4
Etiquetas: Agregar Etiqueta
Sin Etiquetas, Sea el primero en etiquetar este registro!
id oai:doaj.org-article:6b6c8ab959fb42f28121b92a4680f7e4
record_format dspace
spelling oai:doaj.org-article:6b6c8ab959fb42f28121b92a4680f7e42021-11-15T15:40:24ZA Novel Selective LSD1/KDM1A Inhibitor Epigenetically Blocks Herpes Simplex Virus Lytic Replication and Reactivation from Latency10.1128/mBio.00558-122150-7511https://doaj.org/article/6b6c8ab959fb42f28121b92a4680f7e42013-03-01T00:00:00Zhttps://journals.asm.org/doi/10.1128/mBio.00558-12https://doaj.org/toc/2150-7511ABSTRACT Cellular processes requiring access to the DNA genome are regulated by an overlay of epigenetic modifications, including histone modification and chromatin remodeling. Similar to the cellular host, many nuclear DNA viruses that depend upon the host cell’s transcriptional machinery are also subject to the regulatory impact of chromatin assembly and modification. Infection of cells with alphaherpesviruses (herpes simplex virus [HSV] and varicella-zoster virus [VZV]) results in the deposition of nucleosomes bearing repressive histone H3K9 methylation on the viral genome. This repressive state is modulated by the recruitment of a cellular coactivator complex containing the histone H3K9 demethylase LSD1 to the viral immediate-early (IE) gene promoters. Inhibition of the activity of this enzyme results in increased repressive chromatin assembly and suppression of viral gene expression during lytic infection as well as reactivation from latency in a mouse ganglion explant model. However, available small-molecule LSD1 inhibitors are not originally designed to inhibit LSD1, but rather monoamine oxidases (MAO) in general. Thus, their specificity for and potency to LSD1 is low. In this study, a novel specific LSD1 inhibitor was identified that potently repressed HSV IE gene expression, genome replication, and reactivation from latency. Importantly, the inhibitor also suppressed primary infection of HSV in vivo in a mouse model. Based on common control of a number of DNA viruses by epigenetic modulation, it was also demonstrated that this LSD1 inhibitor blocks initial gene expression of the human cytomegalovirus and adenovirus type 5. IMPORTANCE Epigenetic mechanisms, including histone modification and chromatin remodeling, play important regulatory roles in all cellular processes requiring access to the genome. These mechanisms are often altered in disease conditions, including various cancers, and thus represent novel targets for drugs. Similarly, many viral pathogens are regulated by an epigenetic overlay that determines the outcome of infection. Therefore, these epigenetic targets also represent novel antiviral targets. Here, a novel inhibitor was identified with high specificity and potency for the histone demethylase LSD1, a critical component of the herpes simplex virus (HSV) gene expression paradigm. This inhibitor was demonstrated to have potent antiviral potential in both cultured cells and animal models. Thus, in addition to clearly demonstrating the critical role of LSD1 in regulation of HSV infection, as well as other DNA viruses, the data extends the therapeutic potential of chromatin modulation inhibitors from the focused field of oncology to the arena of antiviral agents.Yu LiangDebra QuenelleJodi L. VogelCristina MascaroAlberto OrtegaThomas M. KristieAmerican Society for MicrobiologyarticleMicrobiologyQR1-502ENmBio, Vol 4, Iss 1 (2013)
institution DOAJ
collection DOAJ
language EN
topic Microbiology
QR1-502
spellingShingle Microbiology
QR1-502
Yu Liang
Debra Quenelle
Jodi L. Vogel
Cristina Mascaro
Alberto Ortega
Thomas M. Kristie
A Novel Selective LSD1/KDM1A Inhibitor Epigenetically Blocks Herpes Simplex Virus Lytic Replication and Reactivation from Latency
description ABSTRACT Cellular processes requiring access to the DNA genome are regulated by an overlay of epigenetic modifications, including histone modification and chromatin remodeling. Similar to the cellular host, many nuclear DNA viruses that depend upon the host cell’s transcriptional machinery are also subject to the regulatory impact of chromatin assembly and modification. Infection of cells with alphaherpesviruses (herpes simplex virus [HSV] and varicella-zoster virus [VZV]) results in the deposition of nucleosomes bearing repressive histone H3K9 methylation on the viral genome. This repressive state is modulated by the recruitment of a cellular coactivator complex containing the histone H3K9 demethylase LSD1 to the viral immediate-early (IE) gene promoters. Inhibition of the activity of this enzyme results in increased repressive chromatin assembly and suppression of viral gene expression during lytic infection as well as reactivation from latency in a mouse ganglion explant model. However, available small-molecule LSD1 inhibitors are not originally designed to inhibit LSD1, but rather monoamine oxidases (MAO) in general. Thus, their specificity for and potency to LSD1 is low. In this study, a novel specific LSD1 inhibitor was identified that potently repressed HSV IE gene expression, genome replication, and reactivation from latency. Importantly, the inhibitor also suppressed primary infection of HSV in vivo in a mouse model. Based on common control of a number of DNA viruses by epigenetic modulation, it was also demonstrated that this LSD1 inhibitor blocks initial gene expression of the human cytomegalovirus and adenovirus type 5. IMPORTANCE Epigenetic mechanisms, including histone modification and chromatin remodeling, play important regulatory roles in all cellular processes requiring access to the genome. These mechanisms are often altered in disease conditions, including various cancers, and thus represent novel targets for drugs. Similarly, many viral pathogens are regulated by an epigenetic overlay that determines the outcome of infection. Therefore, these epigenetic targets also represent novel antiviral targets. Here, a novel inhibitor was identified with high specificity and potency for the histone demethylase LSD1, a critical component of the herpes simplex virus (HSV) gene expression paradigm. This inhibitor was demonstrated to have potent antiviral potential in both cultured cells and animal models. Thus, in addition to clearly demonstrating the critical role of LSD1 in regulation of HSV infection, as well as other DNA viruses, the data extends the therapeutic potential of chromatin modulation inhibitors from the focused field of oncology to the arena of antiviral agents.
format article
author Yu Liang
Debra Quenelle
Jodi L. Vogel
Cristina Mascaro
Alberto Ortega
Thomas M. Kristie
author_facet Yu Liang
Debra Quenelle
Jodi L. Vogel
Cristina Mascaro
Alberto Ortega
Thomas M. Kristie
author_sort Yu Liang
title A Novel Selective LSD1/KDM1A Inhibitor Epigenetically Blocks Herpes Simplex Virus Lytic Replication and Reactivation from Latency
title_short A Novel Selective LSD1/KDM1A Inhibitor Epigenetically Blocks Herpes Simplex Virus Lytic Replication and Reactivation from Latency
title_full A Novel Selective LSD1/KDM1A Inhibitor Epigenetically Blocks Herpes Simplex Virus Lytic Replication and Reactivation from Latency
title_fullStr A Novel Selective LSD1/KDM1A Inhibitor Epigenetically Blocks Herpes Simplex Virus Lytic Replication and Reactivation from Latency
title_full_unstemmed A Novel Selective LSD1/KDM1A Inhibitor Epigenetically Blocks Herpes Simplex Virus Lytic Replication and Reactivation from Latency
title_sort novel selective lsd1/kdm1a inhibitor epigenetically blocks herpes simplex virus lytic replication and reactivation from latency
publisher American Society for Microbiology
publishDate 2013
url https://doaj.org/article/6b6c8ab959fb42f28121b92a4680f7e4
work_keys_str_mv AT yuliang anovelselectivelsd1kdm1ainhibitorepigeneticallyblocksherpessimplexviruslyticreplicationandreactivationfromlatency
AT debraquenelle anovelselectivelsd1kdm1ainhibitorepigeneticallyblocksherpessimplexviruslyticreplicationandreactivationfromlatency
AT jodilvogel anovelselectivelsd1kdm1ainhibitorepigeneticallyblocksherpessimplexviruslyticreplicationandreactivationfromlatency
AT cristinamascaro anovelselectivelsd1kdm1ainhibitorepigeneticallyblocksherpessimplexviruslyticreplicationandreactivationfromlatency
AT albertoortega anovelselectivelsd1kdm1ainhibitorepigeneticallyblocksherpessimplexviruslyticreplicationandreactivationfromlatency
AT thomasmkristie anovelselectivelsd1kdm1ainhibitorepigeneticallyblocksherpessimplexviruslyticreplicationandreactivationfromlatency
AT yuliang novelselectivelsd1kdm1ainhibitorepigeneticallyblocksherpessimplexviruslyticreplicationandreactivationfromlatency
AT debraquenelle novelselectivelsd1kdm1ainhibitorepigeneticallyblocksherpessimplexviruslyticreplicationandreactivationfromlatency
AT jodilvogel novelselectivelsd1kdm1ainhibitorepigeneticallyblocksherpessimplexviruslyticreplicationandreactivationfromlatency
AT cristinamascaro novelselectivelsd1kdm1ainhibitorepigeneticallyblocksherpessimplexviruslyticreplicationandreactivationfromlatency
AT albertoortega novelselectivelsd1kdm1ainhibitorepigeneticallyblocksherpessimplexviruslyticreplicationandreactivationfromlatency
AT thomasmkristie novelselectivelsd1kdm1ainhibitorepigeneticallyblocksherpessimplexviruslyticreplicationandreactivationfromlatency
_version_ 1718427770034323456