Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 623, 2022 02 02.
Article in English | MEDLINE | ID: mdl-35110532

ABSTRACT

RNA polymerase III (Pol III) transcribes noncoding RNA, including transfer RNA (tRNA), and is commonly targeted during cancer and viral infection. We find that Herpes Simplex Virus-1 (HSV-1) stimulates tRNA expression 10-fold. Perturbation of host tRNA synthesis requires nuclear viral entry, but not synthesis of specific viral transcripts. tRNA with a specific codon bias were not targeted-rather increased transcription was observed from euchromatic, actively transcribed loci. tRNA upregulation is linked to unique crosstalk between the Pol II and III transcriptional machinery. While viral infection results in depletion of Pol II on host mRNA promoters, we find that Pol II binding to tRNA loci increases. Finally, we report Pol III and associated factors bind the viral genome, which suggests a previously unrecognized role in HSV-1 gene expression. These findings provide insight into mechanisms by which HSV-1 alters the host nuclear environment, shifting key processes in favor of the pathogen.


Subject(s)
Herpesvirus 1, Human/physiology , RNA Polymerase III/metabolism , Transcription Factors , Genome, Viral , Herpesvirus 1, Human/genetics , Humans , Promoter Regions, Genetic , RNA Polymerase II/metabolism , RNA Polymerase III/genetics , RNA, Messenger/metabolism , RNA, Transfer , RNA, Untranslated , Transcription, Genetic , Transcriptional Activation , Virus Replication
2.
EMBO Rep ; 22(9): e53496, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34313382

ABSTRACT

The dynamic nature of interactions between invading viral pathogens and their hosts has fascinated scientists for several decades. The well-known capacity of herpes simplex virus (HSV) to establish life-long infections in humans reflects a dynamic balance between maintaining a latent state in which viral genomes are silenced and re-entry into the lytic phase during reactivation. Silencing of the viral genome has been shown to be a function of innate immune signalling, intrinsic cellular antiviral mechanisms and epigenetic repression. Thus, although many important observations have been made identifying cellular processes that contribute to the repression of the viral genome and latency, the field has lacked an understanding of how these factors work together. In this issue of EMBO Reports, Suzich et al (2021) present convincing evidence that brings together individual observations into a cohesive model that explains many of these outstanding mysteries. Here, we will review the background data that lead to this outstanding piece of work.


Subject(s)
Herpesvirus 1, Human , Epigenetic Repression , Genome, Viral , Herpesvirus 1, Human/genetics , Humans , Virus Latency/genetics
3.
J Virol ; 94(19)2020 09 15.
Article in English | MEDLINE | ID: mdl-32699090

ABSTRACT

Lysine-specific demethylase 1 (LSD1) targets cellular proteins, including histone H3, p53, E2F, and Dnmt1, and is involved in the regulation of gene expression, DNA replication, the cell cycle, and the DNA damage response. LSD1 catalyzes demethylation of histone H3K9 associated with herpes simplex virus 1 (HSV-1) immediate early (IE) promoters and is necessary for IE gene expression, viral DNA replication, and reactivation from latency. We previously found that LSD1 associates with HSV-1 replication forks and replicating viral DNA, suggesting that it may play a direct role in viral replication or coupled processes. We investigated the effects of the LSD1 inhibitor SP-2509 on the HSV-1 life cycle. Unlike previously investigated LSD1 inhibitors tranylcypromine (TCP) and OG-L002, which covalently attach to the LSD1 cofactor flavin adenine dinucleotide (FAD) to inhibit demethylase activity, SP-2509 has previously been shown to inhibit LSD1 protein-protein interactions. We found that SP-2509 does not inhibit HSV-1 IE gene expression or transcription factor and RNA polymerase II (Pol II) association with viral DNA prior to the onset of replication. However, SP-2509 does inhibit viral DNA replication, late gene expression, and virus production. We used EdC labeling of nascent viral DNA to image aberrant viral replication compartments that form in the presence of SP-2509. Treatment resulted in the formation of small replication foci that colocalize with replication proteins but are defective for Pol II recruitment. Taken together, these data highlight a potential new role for LSD1 in the regulation of HSV-1 DNA replication and gene expression after the onset of DNA replication.IMPORTANCE Treatment of HSV-1-infected cells with SP-2509 blocked viral DNA replication, gene expression after the onset of DNA replication, and virus production. These data support a potential new role for LSD1 in the regulation of viral DNA replication and successive steps in the virus life cycle, and further highlight the promising potential to utilize LSD1 inhibition as an antiviral approach.


Subject(s)
Antiviral Agents/pharmacology , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/physiology , Histone Demethylases/drug effects , Hydrazines/pharmacology , Sulfonamides/pharmacology , Virus Replication/drug effects , Animals , Cell Line , Chlorocebus aethiops , DNA Replication/drug effects , DNA, Viral , Gene Expression Regulation, Viral/drug effects , Genes, Immediate-Early , Herpes Simplex/drug therapy , Histones/metabolism , Humans , Promoter Regions, Genetic , Vero Cells
4.
Elife ; 82019 10 22.
Article in English | MEDLINE | ID: mdl-31638576

ABSTRACT

Herpes simplex virus-1 (HSV-1) replicates within the nucleus coopting the host's RNA Polymerase II (Pol II) machinery for production of viral mRNAs culminating in host transcriptional shut off. The mechanism behind this rapid reprogramming of the host transcriptional environment is largely unknown. We identified ICP4 as responsible for preferential recruitment of the Pol II machinery to the viral genome. ICP4 is a viral nucleoprotein which binds double-stranded DNA. We determined ICP4 discriminately binds the viral genome due to the absence of cellular nucleosomes and high density of cognate binding sites. We posit that ICP4's ability to recruit not just Pol II, but also more limiting essential components, such as TBP and Mediator, create a competitive transcriptional environment. These distinguishing characteristics ultimately result in a rapid and efficient reprogramming of the host's transcriptional machinery, which does not occur in the absence of ICP4.


Subject(s)
Herpesvirus 1, Human/metabolism , Immediate-Early Proteins/metabolism , Nucleoproteins/metabolism , Transcription Factors/metabolism , Virus Replication/physiology , Base Sequence , Binding Sites , Carrier Proteins , Cell Line , Environment , Genome, Viral , Herpesvirus 1, Human/genetics , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Humans , Immediate-Early Proteins/genetics , Promoter Regions, Genetic , RNA Polymerase II/metabolism
5.
Proc Natl Acad Sci U S A ; 116(9): 3734-3739, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30808759

ABSTRACT

In herpes simplex virus type 1 (HSV-1) infection, the coupling of genome replication and transcription regulation has been known for many years; however, the underlying mechanism has not been elucidated. We performed a comprehensive transcriptomic assessment and factor-binding analysis for Pol II, TBP, TAF1, and Sp1 to assess the effect genome replication has on viral transcription initiation and elongation. The onset of genome replication resulted in the binding of TBP, TAF1, and Pol II to previously silent late promoters. The viral transcription factor, ICP4, was continuously needed in addition to DNA replication for activation of late gene transcription initiation. Furthermore, late promoters contain a motif that closely matches the consensus initiator element (Inr), which robustly bound TAF1 postreplication. Continued DNA replication resulted in reduced binding of Sp1, TBP, and Pol II to early promoters. Therefore, the initiation of early gene transcription is attenuated following DNA replication. Herein, we propose a model for how viral DNA replication results in the differential utilization of cellular factors that function in transcription initiation, leading to the delineation of kinetic class in HSV-productive infection.


Subject(s)
Immediate-Early Proteins/genetics , RNA Polymerase II/genetics , Simplexvirus/genetics , Transcription, Genetic , Animals , Chlorocebus aethiops , DNA Replication/genetics , Genome, Viral/genetics , Humans , Promoter Regions, Genetic , Protein Binding , TATA Box/genetics , Transcription Factors/genetics , Vero Cells , Virus Replication/genetics
6.
Immunity ; 50(1): 51-63.e5, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30635239

ABSTRACT

Interferon-inducible human oligoadenylate synthetase-like (OASL) and its mouse ortholog, Oasl2, enhance RNA-sensor RIG-I-mediated type I interferon (IFN) induction and inhibit RNA virus replication. Here, we show that OASL and Oasl2 have the opposite effect in the context of DNA virus infection. In Oasl2-/- mice and OASL-deficient human cells, DNA viruses such as vaccinia, herpes simplex, and adenovirus induced increased IFN production, which resulted in reduced virus replication and pathology. Correspondingly, ectopic expression of OASL in human cells inhibited IFN induction through the cGAS-STING DNA-sensing pathway. cGAS was necessary for the reduced DNA virus replication observed in OASL-deficient cells. OASL directly and specifically bound to cGAS independently of double-stranded DNA, resulting in a non-competitive inhibition of the second messenger cyclic GMP-AMP production. Our findings define distinct mechanisms by which OASL differentially regulates host IFN responses during RNA and DNA virus infection and identify OASL as a negative-feedback regulator of cGAS.


Subject(s)
2',5'-Oligoadenylate Synthetase/metabolism , DNA Virus Infections/immunology , DNA Viruses/physiology , RNA Virus Infections/immunology , RNA Viruses/immunology , 2',5'-Oligoadenylate Synthetase/genetics , Animals , Cyclic AMP/metabolism , Humans , Interferon Type I/genetics , Interferon Type I/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nucleotidyltransferases/metabolism , RNA, Small Interfering/genetics , Signal Transduction , THP-1 Cells , Virus Replication
7.
mBio ; 9(4)2018 07 17.
Article in English | MEDLINE | ID: mdl-30018111

ABSTRACT

Herpesviruses utilize multiple mechanisms to redirect host proteins for use in viral processes and to avoid recognition and repression by the host. To investigate dynamic interactions between herpes simplex virus type 1 (HSV-1) DNA and viral and host proteins throughout infection, we developed an approach to identify proteins that associate with the infecting viral genome from nuclear entry through packaging. To accomplish this, virus stocks were prepared in the presence of ethynyl-modified nucleotides to enable covalent tagging of viral genomes after infection for analysis of viral genome-protein interactions by imaging or affinity purification. Affinity purification was combined with stable isotope labeling of amino acids in cell culture (SILAC) mass spectrometry to enable the distinction between proteins that were brought into the cell by the virus or expressed within the infected cell before or during infection. We found that input viral DNA progressed within 6 h through four temporal stages where the genomes sequentially (i) interacted with intrinsic antiviral and DNA damage response proteins, (ii) underwent a robust transcriptional switch mediated largely by ICP4, (iii) engaged in replication, repair, and continued transcription, and then (iv) transitioned to a more transcriptionally inert state engaging de novo-synthesized viral structural components while maintaining interactions with replication proteins. Using a combination of genetic, imaging, and proteomic approaches, we provide a new and temporally compressed view of the HSV-1 life cycle based on input genome-proteome dynamics.IMPORTANCE Herpesviruses are highly prevalent and ubiquitous human pathogens. Studies of herpesviruses and other viruses have previously been limited by the ability to directly study events that occur on the viral DNA throughout infection. We present a new powerful approach, which allows for the temporal investigation of viral genome-protein interactions at all phases of infection. This work has integrated many results from previous studies with the discovery of novel factors potentially involved in viral infection that may represent new antiviral targets. In addition, the study provides a new view of the HSV-1 life cycle based on genome-proteome dynamics.


Subject(s)
Cell Nucleus/virology , Genome, Viral/physiology , Herpes Simplex/pathology , Herpesvirus 1, Human/physiology , Nuclear Proteins/metabolism , Viral Proteins/metabolism , Virus Replication , Cell Line , Cell Nucleus/metabolism , DNA Damage , DNA Repair , DNA Replication , DNA, Viral/genetics , DNA, Viral/metabolism , Genome, Viral/genetics , Host-Pathogen Interactions , Humans , Immediate-Early Proteins/metabolism , Nuclear Proteins/genetics , Transcription Factors/metabolism , Transcription, Genetic , Viral Proteins/biosynthesis , Viral Proteins/genetics , Virus Assembly , Virus Internalization
9.
J Vis Exp ; (126)2017 08 31.
Article in English | MEDLINE | ID: mdl-28892026

ABSTRACT

The goal of this protocol is to isolate herpes simplex virus type 1 (HSV-1) DNA from infected cells for the identification of associated viral and cellular proteins by mass spectrometry. Although proteins that interact with viral genomes play major roles in determining the outcome of infection, a comprehensive analysis of viral genome associated proteins was not previously feasible. Here we demonstrate a method that enables the direct purification of HSV-1 genomes from infected cells. Replicating viral DNA is selectively labeled with modified nucleotides that contain an alkyne functional group. Labeled DNA is then specifically and irreversibly tagged via the covalent attachment of biotin azide via a copper(I)-catalyzed azide-alkyne cycloaddition or click reaction. Biotin-tagged DNA is purified on streptavidin-coated beads and associated proteins are eluted and identified by mass spectrometry. This method enables the selective targeting and isolation of HSV-1 replication forks or whole genomes from complex biological environments. Furthermore, adaptation of this approach will allow for the investigation of various aspects of herpesviral infection, as well as the examination of the genomes of other DNA viruses.


Subject(s)
DNA, Viral/genetics , Viral Proteins/genetics , Humans
10.
mBio ; 8(3)2017 06 13.
Article in English | MEDLINE | ID: mdl-28611249

ABSTRACT

Herpes simplex virus 1 (HSV-1) genes are transcribed by cellular RNA polymerase II (RNA Pol II). While four viral immediate early proteins (ICP4, ICP0, ICP27, and ICP22) function in some capacity in viral transcription, the mechanism by which ICP22 functions remains unclear. We observed that the FACT complex (comprised of SSRP1 and Spt16) was relocalized in infected cells as a function of ICP22. ICP22 was also required for the association of FACT and the transcription elongation factors SPT5 and SPT6 with viral genomes. We further demonstrated that the FACT complex interacts with ICP22 throughout infection. We therefore hypothesized that ICP22 recruits cellular transcription elongation factors to viral genomes for efficient transcription elongation of viral genes. We reevaluated the phenotype of an ICP22 mutant virus by determining the abundance of all viral mRNAs throughout infection by transcriptome sequencing (RNA-seq). The accumulation of almost all viral mRNAs late in infection was reduced compared to the wild type, regardless of kinetic class. Using chromatin immunoprecipitation sequencing (ChIP-seq), we mapped the location of RNA Pol II on viral genes and found that RNA Pol II levels on the bodies of viral genes were reduced in the ICP22 mutant compared to wild-type virus. In contrast, the association of RNA Pol II with transcription start sites in the mutant was not reduced. Taken together, our results indicate that ICP22 plays a role in recruiting elongation factors like the FACT complex to the HSV-1 genome to allow for efficient viral transcription elongation late in viral infection and ultimately infectious virion production.IMPORTANCE HSV-1 interacts with many cellular proteins throughout productive infection. Here, we demonstrate the interaction of a viral protein, ICP22, with a subset of cellular proteins known to be involved in transcription elongation. We determined that ICP22 is required to recruit the FACT complex and other transcription elongation factors to viral genomes and that in the absence of ICP22 viral transcription is globally reduced late in productive infection, due to an elongation defect. This insight defines a fundamental role of ICP22 in HSV-1 infection and elucidates the involvement of cellular factors in HSV-1 transcription.


Subject(s)
Gene Expression Regulation, Viral , Herpesvirus 1, Human/genetics , Immediate-Early Proteins/genetics , Transcription Elongation, Genetic , Animals , Cell Line , Chlorocebus aethiops , Genes, Viral , Genome, Viral , High-Throughput Nucleotide Sequencing , Immediate-Early Proteins/physiology , Mutation , Phosphorylation , RNA Polymerase II/metabolism , Vero Cells , Viral Proteins/genetics , Viral Proteins/metabolism , Virion/genetics
11.
PLoS Pathog ; 13(1): e1006166, 2017 01.
Article in English | MEDLINE | ID: mdl-28095497

ABSTRACT

Herpes simplex virus type 1 (HSV-1) infects over half the human population. Much of the infectious cycle occurs in the nucleus of cells where the virus has evolved mechanisms to manipulate host processes for the production of virus. The genome of HSV-1 is coordinately expressed, maintained, and replicated such that progeny virions are produced within 4-6 hours post infection. In this study, we selectively purify HSV-1 replication forks and associated proteins from virus-infected cells and identify select viral and cellular replication, repair, and transcription factors that associate with viral replication forks. Pulse chase analyses and imaging studies reveal temporal and spatial dynamics between viral replication forks and associated proteins and demonstrate that several DNA repair complexes and key transcription factors are recruited to or near replication forks. Consistent with these observations we show that the initiation of viral DNA replication is sufficient to license late gene transcription. These data provide insight into mechanisms that couple HSV-1 DNA replication with transcription and repair for the coordinated expression and maintenance of the viral genome.


Subject(s)
DNA Replication/genetics , Herpes Simplex/genetics , Herpesvirus 1, Human/growth & development , Host-Pathogen Interactions/genetics , Virus Replication/genetics , Animals , Cell Line , Chlorocebus aethiops , DNA, Viral/analysis , DNA, Viral/genetics , Fluorescent Antibody Technique , Genes, Viral/genetics , Genome, Viral/genetics , Herpesvirus 1, Human/genetics , Host-Pathogen Interactions/physiology , Humans , Mass Spectrometry , Vero Cells , Virus Replication/physiology
12.
Virology ; 496: 186-193, 2016 09.
Article in English | MEDLINE | ID: mdl-27336950

ABSTRACT

West Nile virus (WNV) is a flavivirus that swept rapidly across North America in 1999, declined in prevalence, and then resurged in 2012. To date, no vaccine is available to prevent infection in the human population. Herpes simplex virus (HSV) replication-defective vaccine vectors induce a durable immunity characterized by strong antibody and CD8(+) T cell responses even in HSV-immune animals. In this study, a WNV protein expression cassette was optimized for virus-like particle (VLP) production in transfection studies, and the cassette was recombined into an HSV-1 d106-WNV virus vector, which produced extracellular VLPs, as confirmed by immunoelectron microscopy. Immunization of mice with the d106-WNV recombinant vector elicited a specific anti-WNV IgG response. This study highlights the flavivirus coding sequences needed for efficient assembly of virus-like particles. This information will facilitate generation of additional vaccine vectors against other flaviviruses including the recently emerged Zika virus.


Subject(s)
Genetic Vectors/genetics , Herpesvirus 1, Human/genetics , Vaccines, Virus-Like Particle/genetics , Viral Structural Proteins/genetics , West Nile virus/genetics , Amino Acid Sequence , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Capsid/immunology , Capsid/ultrastructure , Cell Line , Gene Order , Humans , Immunization , Mice , Vaccines, Virus-Like Particle/administration & dosage , Vaccines, Virus-Like Particle/immunology , Vaccines, Virus-Like Particle/ultrastructure , Viral Structural Proteins/chemistry , Viral Structural Proteins/immunology , West Nile virus/immunology
13.
Virology ; 487: 215-21, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26547038

ABSTRACT

A collection of genomic DNA sequences of herpes simplex virus (HSV) strains has been defined and analyzed, and some information is available about genomic stability upon limited passage of viruses in culture. The nature of genomic change upon extensive laboratory passage remains to be determined. In this report we review the history of the HSV-1 KOS laboratory strain and the related KOS1.1 laboratory sub-strain, also called KOS (M), and determine the complete genomic sequence of an early passage stock of the KOS laboratory sub-strain and a laboratory stock of the KOS1.1 sub-strain. The genomes of the two sub-strains are highly similar with only five coding changes, 20 non-coding changes, and about twenty non-ORF sequence changes. The coding changes could potentially explain the KOS1.1 phenotypic properties of increased replication at high temperature and reduced neuroinvasiveness. The study also provides sequence markers to define the provenance of specific laboratory KOS virus stocks.


Subject(s)
DNA, Viral/genetics , Genome, Viral/genetics , Herpesvirus 1, Human/genetics , Amino Acid Sequence , Animals , Base Sequence , Chlorocebus aethiops , Genetic Markers/genetics , Herpesvirus 1, Human/classification , Male , Mice , Molecular Sequence Data , Sequence Analysis, DNA , Vero Cells
14.
PLoS Pathog ; 11(5): e1004939, 2015 May.
Article in English | MEDLINE | ID: mdl-26018390

ABSTRACT

Much of the HSV-1 life cycle is carried out in the cell nucleus, including the expression, replication, repair, and packaging of viral genomes. Viral proteins, as well as cellular factors, play essential roles in these processes. Isolation of proteins on nascent DNA (iPOND) was developed to label and purify cellular replication forks. We adapted aspects of this method to label viral genomes to both image, and purify replicating HSV-1 genomes for the identification of associated proteins. Many viral and cellular factors were enriched on viral genomes, including factors that mediate DNA replication, repair, chromatin remodeling, transcription, and RNA processing. As infection proceeded, packaging and structural components were enriched to a greater extent. Among the more abundant proteins that copurified with genomes were the viral transcription factor ICP4 and the replication protein ICP8. Furthermore, all seven viral replication proteins were enriched on viral genomes, along with cellular PCNA and topoisomerases, while other cellular replication proteins were not detected. The chromatin-remodeling complexes present on viral genomes included the INO80, SWI/SNF, NURD, and FACT complexes, which may prevent chromatinization of the genome. Consistent with this conclusion, histones were not readily recovered with purified viral genomes, and imaging studies revealed an underrepresentation of histones on viral genomes. RNA polymerase II, the mediator complex, TFIID, TFIIH, and several other transcriptional activators and repressors were also affinity purified with viral DNA. The presence of INO80, NURD, SWI/SNF, mediator, TFIID, and TFIIH components is consistent with previous studies in which these complexes copurified with ICP4. Therefore, ICP4 is likely involved in the recruitment of these key cellular chromatin remodeling and transcription factors to viral genomes. Taken together, iPOND is a valuable method for the study of viral genome dynamics during infection and provides a comprehensive view of how HSV-1 selectively utilizes cellular resources.


Subject(s)
Gene Expression Regulation, Viral , Genome, Viral , Herpes Simplex/metabolism , Herpesvirus 1, Human/physiology , Lung/metabolism , Nuclear Proteins/metabolism , Viral Proteins/metabolism , Animals , Blotting, Western , Cell Nucleus/genetics , Cells, Cultured , Chlorocebus aethiops , Chromatin Assembly and Disassembly , DNA Replication , Fluorescent Antibody Technique , Herpes Simplex/genetics , Herpes Simplex/virology , Humans , Lung/cytology , Lung/embryology , Nuclear Proteins/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Vero Cells , Viral Proteins/genetics , Virus Replication
15.
J Virol ; 88(12): 6847-61, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24719411

ABSTRACT

UNLABELLED: Herpes simplex virus 1 (HSV-1) can undergo a productive infection in nonneuronal and neuronal cells such that the genes of the virus are transcribed in an ordered cascade. HSV-1 can also establish a more quiescent or latent infection in peripheral neurons, where gene expression is substantially reduced relative to that in productive infection. HSV mutants defective in multiple immediate early (IE) gene functions are highly defective for later gene expression and model some aspects of latency in vivo. We compared the expression of wild-type (wt) virus and IE gene mutants in nonneuronal cells (MRC5) and adult murine trigeminal ganglion (TG) neurons using the Illumina platform for cDNA sequencing (RNA-seq). RNA-seq analysis of wild-type virus revealed that expression of the genome mostly followed the previously established kinetics, validating the method, while highlighting variations in gene expression within individual kinetic classes. The accumulation of immediate early transcripts differed between MRC5 cells and neurons, with a greater abundance in neurons. Analysis of a mutant defective in all five IE genes (d109) showed dysregulated genome-wide low-level transcription that was more highly attenuated in MRC5 cells than in TG neurons. Furthermore, a subset of genes in d109 was more abundantly expressed over time in neurons. While the majority of the viral genome became relatively quiescent, the latency-associated transcript was specifically upregulated. Unexpectedly, other genes within repeat regions of the genome, as well as the unique genes just adjacent the repeat regions, also remained relatively active in neurons. The relative permissiveness of TG neurons to viral gene expression near the joint region is likely significant during the establishment and reactivation of latency. IMPORTANCE: During productive infection, the genes of HSV-1 are transcribed in an ordered cascade. HSV can also establish a more quiescent or latent infection in peripheral neurons. HSV mutants defective in multiple immediate early (IE) genes establish a quiescent infection that models aspects of latency in vivo. We simultaneously quantified the expression of all the HSV genes in nonneuronal and neuronal cells by RNA-seq analysis. The results for productive infection shed further light on the nature of genes and promoters of different kinetic classes. In quiescent infection, there was greater transcription across the genome in neurons than in nonneuronal cells. In particular, the transcription of the latency-associated transcript (LAT), IE genes, and genes in the unique regions adjacent to the repeats persisted in neurons. The relative activity of this region of the genome in the absence of viral activators suggests a more dynamic state for quiescent genomes persisting in neurons.


Subject(s)
Fibroblasts/virology , Gene Expression Regulation, Viral , Genome, Viral , Herpes Simplex/virology , Herpesvirus 1, Human/genetics , Neurons/virology , Trigeminal Ganglion/virology , Virus Replication , Animals , Herpesvirus 1, Human/physiology , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Mice , Virus Activation
16.
PLoS One ; 8(10): e78242, 2013.
Article in English | MEDLINE | ID: mdl-24147125

ABSTRACT

The herpes simplex virus type 1 (HSV-1) immediate early protein, ICP4, participates in the regulation of viral gene expression by both activating and repressing RNA polII transcription. We used affinity purification of ICP4 expressed in infected cells followed by mass spectrometry and western blot analysis to determine the composition of cellular complexes associated with ICP4 throughout infection. ICP4 was associated with TFIID complexes containing a distinct set of TAFs. These complexes were most abundant early, but were detected throughout infection, whereas Mediator was found in ICP4 containing complexes later in infection, indicating a temporal pattern for the utilization of these complexes for the transcription of the viral genome. The form of Mediator copurifying with ICP4 was enriched for the kinase domain and also lacked the activator-specific component, Med26, suggesting that Mediator-ICP4 interactions may be involved in repression of viral transcription. The N-terminal 774 amino acids of ICP4, which retains partial function, were sufficient to form complexes with TFIID and Mediator, although these interactions were not as strong as with full-length ICP4. Additionally, components involved in transcription elongation, chromatin remodeling, and mRNA processing were isolated with ICP4. Together our data indicate that ICP4 plays a more integrated role in mediating HSV transcription, possibly affecting multiple steps in transcription and gene expression.


Subject(s)
Immediate-Early Proteins/metabolism , Simplexvirus/metabolism , Simplexvirus/physiology , Animals , Chlorocebus aethiops , Gene Expression Regulation, Viral/genetics , Gene Expression Regulation, Viral/physiology , Immediate-Early Proteins/genetics , Protein Binding , Simplexvirus/genetics , Transcription Factor TFIID/genetics , Transcription Factor TFIID/metabolism , Vero Cells
17.
J Virol ; 87(2): 1010-8, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23135715

ABSTRACT

ICP4 is the major activator of herpes simplex virus (HSV) transcription. Previous studies have defined several regions of ICP4 that are important for viral gene expression, including a DNA binding domain and transactivation domains that are contained in the C-terminal and N-terminal 520 and 274 amino acids, respectively. Here we show that the N-terminal 210 amino acids of ICP4 are required for interactions with components of TFIID and mediator and, as a consequence, are necessary for the activation of viral genes. A mutant of ICP4 deleted for amino acids 30 to 210, d3-10, was unable to complement an ICP4 null virus at the level of viral replication. This was the result of a severe deficiency in viral gene and protein expression. The absence of viral gene expression coincided with a defect in the recruitment of RNA polymerase II to a representative early promoter (thymidine kinase [TK]). Affinity purification experiments demonstrated that d3-10 ICP4 was not found in complexes with components of TFIID and mediator, suggesting that the defect in RNA polymerase II (Pol II) recruitment was the result of ablated interactions between d3-10 and TFIID and mediator. Complementation assays suggested that the N-terminal and C-terminal regions of ICP4 cooperate to mediate gene expression. The complementation was the result of the formation of more functional heterodimers, which restored the ability of the d3-10-containing molecules to interact with TFIID. Together, these studies suggest that the N terminus contains a true activation domain, mediating interactions with TFIID, mediator, and perhaps other transcription factors, and that the C terminus of the molecule contains activities that augment the functions of the activation domain.


Subject(s)
Gene Expression Regulation, Viral , Immediate-Early Proteins/metabolism , Simplexvirus/genetics , Animals , Cell Line , DNA Mutational Analysis , Gene Deletion , Genetic Complementation Test , Immediate-Early Proteins/genetics , Protein Interaction Mapping , Sequence Deletion , Transcription Factor TFIID/metabolism , Transcription, Genetic
18.
Proc Natl Acad Sci U S A ; 109(44): E3008-17, 2012 Oct 30.
Article in English | MEDLINE | ID: mdl-23027953

ABSTRACT

Innate sensing of microbial components is well documented to occur at many cellular sites, including at the cell surface, in the cytosol, and in intracellular vesicles, but there is limited evidence of nuclear innate signaling. In this study we have defined the mechanisms of interferon regulatory factor-3 (IRF-3) signaling in primary human foreskin fibroblasts (HFF) infected with herpes simplex virus 1 (HSV-1) in the absence of viral gene expression. We found that the interferon inducible protein 16 (IFI16) DNA sensor, which is required for induction of IRF-3 signaling in these cells, is nuclear, and its localization does not change detectably upon HSV-1 d109 infection and induction of IRF-3 signaling. Consistent with the IFI16 sensor being nuclear, conditions that block viral DNA release from incoming capsids inhibit IRF-3 signaling. An unknown factor must be exported from the nucleus to activate IRF-3 through cytoplasmic STING, which is required for IRF-3 activation and signaling. However, when the viral ICP0 protein is expressed in the nucleus, it causes the nuclear relocalization and degradation of IFI16, inhibiting IRF-3 signaling. Therefore, HSV-1 infection is sensed in HFF by nuclear IFI16 upon release of encapsidated viral DNA into the nucleus, and the viral nuclear ICP0 protein can inhibit the process by targeting IFI16 for degradation. Together these results define a pathway for nuclear innate sensing of HSV DNA by IFI16 in infected HFF and document a mechanism by which a virus can block this nuclear innate response.


Subject(s)
Herpes Labialis/metabolism , Immediate-Early Proteins/physiology , Interferon Regulatory Factor-3/metabolism , Nuclear Proteins/physiology , Phosphoproteins/physiology , Signal Transduction/physiology , Ubiquitin-Protein Ligases/physiology , Cells, Cultured , DNA, Viral/metabolism , Flow Cytometry , Fluorescent Antibody Technique, Indirect , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/metabolism , Humans , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Polymerase Chain Reaction
19.
J Virol ; 86(12): 6862-74, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22496239

ABSTRACT

Infected cell polypeptide 4 (ICP4) activates transcription from most viral promoters. Two transactivation domains, one N-terminal and one C terminal, are largely responsible for the activation functions of ICP4. A mutant ICP4 molecule lacking the C-terminal activation domain (n208) efficiently activates many early genes, whereas late genes are poorly activated, and virus growth is severely impaired. The regions within the N terminus of ICP4 (amino acids 1 to 210) that contribute to activation were investigated by analysis of deletion mutants in the presence or absence of the C-terminal activation domain. The mutants were assessed for their abilities to support viral replication and to regulate gene expression. Several deletions in regions conserved in other alphaherpesviruses resulted in impaired activation and viral growth, without affecting DNA binding. The single small deletion that had the greatest effect on activation in the absence of the C terminus corresponded to a highly conserved stretch of amino acids between 81 and 96, rendering the molecule nonfunctional. However, when the C terminus was present, the same deletion had a minimal effect on activity. The amino terminus of ICP4 was predicted to be relatively disordered compared to the DNA-binding domain and the C-terminal 500 amino acids. Moreover, the amino terminus appears to be in a relatively extended conformation as determined by the hydrodynamic properties of several mutants. The data support a model where the amino terminus is an extended and possibly flexible region of the protein, allowing it to efficiently interact with multiple transcription factors at a distance from where it is bound to DNA, thereby enabling ICP4 to function as a general activator of polymerase II transcription. The C terminus of ICP4 can compensate for some of the mutations in the N terminus, suggesting that it either specifies redundant interactions or enables the amino terminus to function more efficiently.


Subject(s)
Gene Expression Regulation, Viral , Herpes Simplex/virology , Herpesvirus 1, Human/metabolism , Immediate-Early Proteins/chemistry , Immediate-Early Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Cell Line , Chlorocebus aethiops , Herpesvirus 1, Human/chemistry , Herpesvirus 1, Human/genetics , Humans , Immediate-Early Proteins/genetics , Molecular Sequence Data , Promoter Regions, Genetic , Sequence Alignment , Transcriptional Activation , Vero Cells
20.
J Virol ; 85(12): 5733-44, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21450820

ABSTRACT

The infected cell polypeptide 4 (ICP4) of herpes simplex virus 1 (HSV-1) is a regulator of viral transcription that is required for productive infection. Since viral genes are transcribed by cellular RNA polymerase II (RNA pol II), ICP4 must interact with components of the pol II machinery to regulate viral gene expression. It has been shown previously that ICP4 interacts with TATA box-binding protein (TBP), TFIIB, and the TBP-associated factor 1 (TAF1) in vitro. In this study, ICP4-containing complexes were isolated from infected cells by tandem affinity purification (TAP). Forty-six proteins that copurified with ICP4 were identified by mass spectrometry. Additional copurifying proteins were identified by Western blot analysis. These included 11 components of TFIID and 4 components of the Mediator complex. The significance of the ICP4-Mediator interaction was further investigated using immunofluorescence and chromatin immunoprecipitation. Mediator was found to colocalize with ICP4 starting at early and continuing into late times of infection. In addition, Mediator was recruited to viral promoters in an ICP4-dependent manner. Taken together, the data suggest that ICP4 interacts with components of TFIID and Mediator in the context of viral infection, and this may explain the broad transactivation properties of ICP4.


Subject(s)
Herpesvirus 1, Human/pathogenicity , Immediate-Early Proteins/metabolism , Mediator Complex/metabolism , Transcription Factor TFIID/metabolism , Animals , Blotting, Western , Cell Line , Chlorocebus aethiops , Chromatin Immunoprecipitation , Chromatography, Affinity/methods , Fluorescent Antibody Technique , HeLa Cells , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/metabolism , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/isolation & purification , Mass Spectrometry , Mediator Complex/genetics , Transcription Factor TFIID/genetics , Transcription Factor TFIID/isolation & purification , Transcriptional Activation , Vero Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...