Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
mBio ; 14(1): e0354222, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36692302

ABSTRACT

Transcription of herpes simplex virus 1 (HSV-1) immediate early (IE) genes is controlled at multiple levels by the cellular transcriptional coactivator, HCF-1. HCF-1 is complexed with epigenetic factors that prevent silencing of the viral genome upon infection, transcription factors that drive initiation of IE gene expression, and transcription elongation factors required to circumvent RNAPII pausing at IE genes and promote productive IE mRNA synthesis. Significantly, the coactivator is also implicated in the control of viral reactivation from latency in sensory neurons based on studies that demonstrate that HCF-1-associated epigenetic and transcriptional elongation complexes are critical to initiate IE expression and viral reactivation. Here, an HCF-1 conditional knockout mouse model (HCF-1cKO) was derived to probe the role and significance of HCF-1 in the regulation of HSV-1 latency/reactivation in vivo. Upon deletion of HCF-1 in sensory neurons, there is a striking reduction in the number of latently infected neurons that initiate viral reactivation. Importantly, this correlated with a defect in the removal of repressive chromatin associated with latent viral genomes. These data demonstrate that HCF-1 is a critical regulatory factor that governs the initiation of HSV reactivation, in part, by promoting the transition of latent viral genomes from a repressed heterochromatic state. IMPORTANCE Herpes simplex virus is responsible for a substantial worldwide disease burden. An initial infection leads to the establishment of a lifelong persistent infection in sensory neurons. Periodic reactivation can result in recurrent oral and genital lesions to more significant ocular disease. Despite the significance of this pathogen, many of the regulatory factors and molecular mechanisms that govern the viral latency-reactivation cycles have yet to be elucidated. Initiation of both lytic infection and reactivation are dependent on the expression of the viral immediate early genes. In vivo deletion of a central component of the IE regulatory paradigm, the cellular transcriptional coactivator HCF-1, reduces the epigenetic transition of latent viral genomes, thus suppressing HSV reactivation. These observations define HCF-1 as a critical regulator that controls the initiation of HSV reactivation from latency in vivo and contribute to understanding of the molecular mechanisms that govern viral reactivation.


Subject(s)
Herpes Simplex , Herpesvirus 1, Human , Animals , Mice , Gene Expression Regulation, Viral , Herpesvirus 1, Human/physiology , Heterochromatin , Transcription Factors/metabolism , Transcription, Genetic , Virus Latency/physiology
2.
J Virol ; 93(24)2019 12 15.
Article in English | MEDLINE | ID: mdl-31554680

ABSTRACT

Ocular herpes simplex keratitis (HSK) is a consequence of viral reactivations from trigeminal ganglia (TG) and occurs almost exclusively in the same eye in humans. In our murine oro-ocular (OO) model, herpes simplex virus 1 (HSV-1) inoculation in one side of the lip propagates virus to infect the ipsilateral TG. Replication here allows infection of the brainstem and infection of the contralateral TG. Interestingly, HSK was observed in our OO model only from the eye ipsilateral to the site of lip infection. Thus, unilateral restriction of HSV-1 may be due to differential kinetics of virus arrival in the ipsilateral versus contralateral TG. We inoculated mice with HSV-1 reporter viruses and then superinfected them to monitor changes in acute- and latent-phase gene expression in TG after superinfection compared to the control (single inoculation). Delaying superinfection by 4 days after initial right lip inoculation elicited failed superinfecting-virus gene expression and eliminated clinical signs of disease. Initial inoculation with thymidine kinase-deficient HSV-1 (TKdel) completely abolished reactivation of wild-type (WT) superinfecting virus from TG during the latent stage. In light of these seemingly failed infections, viral genome was detected in both TG. Our data demonstrate that inoculation of HSV-1 in the lip propagates virus to both TG, but with delay in reaching the TG contralateral to the side of lip infection. This delay is responsible for restricting viral replication to the ipsilateral TG, which abrogates ocular disease and viral reactivations from the contralateral side. These observations may help to understand why HSK is observed unilaterally in humans, and they provide insight into vaccine strategies to protect against HSK.IMPORTANCE Herpetic keratitis (HK) is the leading cause of blindness by an infectious agent in the developed world. This disease can occur after reactivation of herpes simplex virus 1 in the trigeminal ganglia, leading to dissemination of virus to, and infection of, the cornea. A clinical paradox is evidenced by the bilateral presence of latent viral genomes in both trigeminal ganglia, while for any given patient the disease is unilateral with recurrences in a single eye. Our study links the kinetics of early infection to unilateral disease phenomenon and demonstrates protection against viral reactivation when kinetics are exploited. Our results have direct implications in the understanding of human disease pathogenesis and immunotherapeutic strategies for the treatment of HK and viral reactivations.


Subject(s)
Herpesvirus 1, Human/physiology , Keratitis, Herpetic/virology , Lip/virology , Virus Latency/physiology , Virus Replication/physiology , Animals , Cornea/virology , Female , Gene Expression Regulation, Viral , Genes, Viral/genetics , Herpesvirus 1, Human/genetics , Mice , Mice, Inbred BALB C , MicroRNAs/genetics , MicroRNAs/metabolism , Trigeminal Ganglion/virology , Virus Latency/genetics
3.
Proc Natl Acad Sci U S A ; 113(36): 10085-90, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27528682

ABSTRACT

Viruses are intracellular pathogens that hijack host cell machinery and resources to replicate. Rather than being constant, host physiology is rhythmic, undergoing circadian (∼24 h) oscillations in many virus-relevant pathways, but whether daily rhythms impact on viral replication is unknown. We find that the time of day of host infection regulates virus progression in live mice and individual cells. Furthermore, we demonstrate that herpes and influenza A virus infections are enhanced when host circadian rhythms are abolished by disrupting the key clock gene transcription factor Bmal1. Intracellular trafficking, biosynthetic processes, protein synthesis, and chromatin assembly all contribute to circadian regulation of virus infection. Moreover, herpesviruses differentially target components of the molecular circadian clockwork. Our work demonstrates that viruses exploit the clockwork for their own gain and that the clock represents a novel target for modulating viral replication that extends beyond any single family of these ubiquitous pathogens.


Subject(s)
ARNTL Transcription Factors/genetics , Circadian Clocks/genetics , Herpes Simplex/virology , Herpesviridae Infections/virology , Host-Pathogen Interactions , Orthomyxoviridae Infections/virology , Tumor Virus Infections/virology , ARNTL Transcription Factors/deficiency , Animals , Biological Transport , CLOCK Proteins/genetics , CLOCK Proteins/metabolism , Cell Line , Chromatin Assembly and Disassembly , Circadian Rhythm/genetics , Cricetinae , Epithelial Cells/metabolism , Epithelial Cells/virology , Female , Gene Expression Regulation , Genes, Reporter , Herpes Simplex/genetics , Herpes Simplex/metabolism , Herpesviridae Infections/genetics , Herpesviridae Infections/metabolism , Herpesvirus 1, Human/pathogenicity , Herpesvirus 1, Human/physiology , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H1N1 Subtype/physiology , Luciferases/genetics , Luciferases/metabolism , Mice , Mice, Knockout , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/metabolism , Rhadinovirus/pathogenicity , Rhadinovirus/physiology , Tumor Virus Infections/genetics , Tumor Virus Infections/metabolism , Virus Replication
4.
PLoS Pathog ; 12(4): e1005539, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27055281

ABSTRACT

Herpes simplex virus 1 (HSV-1) establishes life-long latent infection within sensory neurons, during which viral lytic gene expression is silenced. The only highly expressed viral gene product during latent infection is the latency-associated transcript (LAT), a non-protein coding RNA that has been strongly implicated in the epigenetic regulation of HSV-1 gene expression. We have investigated LAT-mediated control of latent gene expression using chromatin immunoprecipitation analyses and LAT-negative viruses engineered to express firefly luciferase or ß-galactosidase from a heterologous lytic promoter. Whilst we were unable to determine a significant effect of LAT expression upon heterochromatin enrichment on latent HSV-1 genomes, we show that reporter gene expression from latent HSV-1 genomes occurs at a greater frequency in the absence of LAT. Furthermore, using luciferase reporter viruses we have observed that HSV-1 gene expression decreases during long-term latent infection, with a most marked effect during LAT-negative virus infection. Finally, using a fluorescent mouse model of infection to isolate and culture single latently infected neurons, we also show that reactivation occurs at a greater frequency from cultures harbouring LAT-negative HSV-1. Together, our data suggest that the HSV-1 LAT RNA represses HSV-1 gene expression in small populations of neurons within the mouse TG, a phenomenon that directly impacts upon the frequency of reactivation and the maintenance of the transcriptionally active latent reservoir.


Subject(s)
Gene Expression Regulation, Viral , Herpesvirus 1, Human/genetics , Neurons/metabolism , Transcription, Genetic , Viral Proteins/genetics , Virus Latency/genetics , Virus Physiological Phenomena/genetics , Animals , Cells, Cultured , Epigenesis, Genetic/genetics , Gene Expression/genetics , RNA, Viral/genetics , RNA, Viral/metabolism
5.
PLoS One ; 10(11): e0142751, 2015.
Article in English | MEDLINE | ID: mdl-26562415

ABSTRACT

The 2013-present Ebola virus outbreak in Western Africa has prompted the production of many diagnostic assays, mostly based on nucleic acid amplification technologies (NAT). The calibration and performance assessment of established assays and those under evaluation requires reference materials that can be used in parallel with the clinical sample to standardise or control for every step of the procedure, from extraction to the final qualitative/quantitative result. We have developed safe and stable Ebola virus RNA reference materials by encapsidating anti sense viral RNA into HIV-1-like particles. The lentiviral particles are replication-deficient and non-infectious due to the lack of HIV-1 genes and Envelope protein. Ebola virus genes were subcloned for encapsidation into two lentiviral preparations, one containing NP-VP35-GP and the other VP40 and L RNA. Each reference material was formulated as a high-titre standard for use as a calibrator for secondary or internal standards, and a 10,000-fold lower titre preparation to serve as an in-run control. The preparations have been freeze-dried to maximise stability. These HIV-Ebola virus RNA reference materials were suitable for use with in-house and commercial quantitative RT-PCR assays and with digital RT-PCR. The HIV-Ebola virus RNA reference materials are stable at up to 37°C for two weeks, allowing the shipment of the material worldwide at ambient temperature. These results support further evaluation of the HIV-Ebola virus RNA reference materials as part of an International collaborative study for the establishment of the 1st International Standard for Ebola virus RNA.


Subject(s)
Ebolavirus/genetics , Ebolavirus/isolation & purification , Hemorrhagic Fever, Ebola/diagnosis , Lentivirus/genetics , Nucleic Acid Amplification Techniques/methods , RNA, Viral/genetics , Virion/genetics , Africa, Western/epidemiology , Calibration , HEK293 Cells , HIV-1/genetics , Hemorrhagic Fever, Ebola/epidemiology , Hemorrhagic Fever, Ebola/virology , Humans , Nucleic Acid Amplification Techniques/standards , RNA, Viral/isolation & purification , Reference Standards
7.
Nat Commun ; 6: 7126, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25989971

ABSTRACT

Herpes simplex virus 1 (HSV-1) is an important human pathogen and a paradigm for virus-induced host shut-off. Here we show that global changes in transcription and RNA processing and their impact on translation can be analysed in a single experimental setting by applying 4sU-tagging of newly transcribed RNA and ribosome profiling to lytic HSV-1 infection. Unexpectedly, we find that HSV-1 triggers the disruption of transcription termination of cellular, but not viral, genes. This results in extensive transcription for tens of thousands of nucleotides beyond poly(A) sites and into downstream genes, leading to novel intergenic splicing between exons of neighbouring cellular genes. As a consequence, hundreds of cellular genes seem to be transcriptionally induced but are not translated. In contrast to previous reports, we show that HSV-1 does not inhibit co-transcriptional splicing. Our approach thus substantially advances our understanding of HSV-1 biology and establishes HSV-1 as a model system for studying transcription termination.


Subject(s)
Herpes Simplex/virology , Herpesvirus 1, Human/genetics , Transcription, Genetic , Alternative Splicing , Codon, Terminator , Culture Media , Exons , Fibroblasts/metabolism , Fibroblasts/virology , Gene Expression Regulation, Viral , Genes, Viral , Genome, Viral , Humans , Introns , Open Reading Frames , Poly A , Protein Biosynthesis , RNA/biosynthesis , RNA, Messenger/metabolism , Ribosomes/ultrastructure , Time Factors , Virus Replication/genetics
8.
J Virol ; 88(8): 3965-75, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24501409

ABSTRACT

UNLABELLED: Lymphocyte colonization by gammaherpesviruses (γHVs) is an important target for cancer prevention. However, how it works is not clear. Epstein-Barr virus drives autonomous B cell proliferation in vitro but in vivo may more subtly exploit the proliferative pathways provided by lymphoid germinal centers (GCs). Murid herpesvirus 4 (MuHV-4), which realistically infects inbred mice, provides a useful tool with which to understand further how a γHV colonizes B cells in vivo. Not all γHVs necessarily behave the same, but common events can with MuHV-4 be assigned an importance for host colonization and so a potential as therapeutic targets. MuHV-4-driven B cell proliferation depends quantitatively on CD4(+) T cell help. Here we show that it also depends on T cell-independent survival signals provided by the B cell-activating factor (BAFF) receptor (BAFF-R). B cells could be infected in BAFF-R(-/-) mice, but virus loads remained low. This corresponded to a BAFF-R-dependent defect in GC colonization. The close parallels between normal, antigen-driven B cell responses and virus-infected B cell proliferation argue that in vivo, γHVs mostly induce infected B cells into normal GC reactions rather than generating large numbers of autonomously proliferating blasts. IMPORTANCE: γHVs cause cancers by driving the proliferation of infected cells. B cells are a particular target. Thus, we need to know how virus-driven B cell proliferation works. Controversy exists as to whether viral genes drive it directly or less directly orchestrate the engagement of normal, host-driven pathways. Here we show that the B cell proliferation driven by a murid γHV requires BAFF-R. This supports the idea that γHVs exploit host proliferation pathways and suggests that interfering with BAFF-R could more generally reduce γHV-associated B cell proliferation.


Subject(s)
B-Cell Activating Factor/metabolism , B-Cell Activation Factor Receptor/deficiency , Herpesviridae Infections/metabolism , Herpesviridae Infections/virology , Rhadinovirus/physiology , Animals , B-Cell Activation Factor Receptor/genetics , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Female , Gammaherpesvirinae/genetics , Gammaherpesvirinae/physiology , Herpesviridae Infections/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Rhadinovirus/genetics
9.
PLoS Pathog ; 9(8): e1003514, 2013.
Article in English | MEDLINE | ID: mdl-23950709

ABSTRACT

Herpes simplex virus type 1 (HSV-1) is a neurotropic virus causing vesicular oral or genital skin lesions, meningitis and other diseases particularly harmful in immunocompromised individuals. To comprehensively investigate the complex interaction between HSV-1 and its host we combined two genome-scale screens for host factors (HFs) involved in virus replication. A yeast two-hybrid screen for protein interactions and a RNA interference (RNAi) screen with a druggable genome small interfering RNA (siRNA) library confirmed existing and identified novel HFs which functionally influence HSV-1 infection. Bioinformatic analyses found the 358 HFs were enriched for several pathways and multi-protein complexes. Of particular interest was the identification of Med23 as a strongly anti-viral component of the largely pro-viral Mediator complex, which links specific transcription factors to RNA polymerase II. The anti-viral effect of Med23 on HSV-1 replication was confirmed in gain-of-function gene overexpression experiments, and this inhibitory effect was specific to HSV-1, as a range of other viruses including Vaccinia virus and Semliki Forest virus were unaffected by Med23 depletion. We found Med23 significantly upregulated expression of the type III interferon family (IFN-λ) at the mRNA and protein level by directly interacting with the transcription factor IRF7. The synergistic effect of Med23 and IRF7 on IFN-λ induction suggests this is the major transcription factor for IFN-λ expression. Genotypic analysis of patients suffering recurrent orofacial HSV-1 outbreaks, previously shown to be deficient in IFN-λ secretion, found a significant correlation with a single nucleotide polymorphism in the IFN-λ3 (IL28b) promoter strongly linked to Hepatitis C disease and treatment outcome. This paper describes a link between Med23 and IFN-λ, provides evidence for the crucial role of IFN-λ in HSV-1 immune control, and highlights the power of integrative genome-scale approaches to identify HFs critical for disease progression and outcome.


Subject(s)
Genome, Human , Herpesvirus 1, Human/physiology , Interleukins/biosynthesis , Mediator Complex/biosynthesis , Up-Regulation , Virus Replication/physiology , Gene Deletion , HeLa Cells , Herpes Simplex/genetics , Herpes Simplex/immunology , Herpes Simplex/metabolism , Humans , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/immunology , Interferon Regulatory Factor-7/metabolism , Interferons , Interleukins/genetics , Interleukins/immunology , Mediator Complex/genetics , Mediator Complex/immunology , Polymorphism, Single Nucleotide , RNA Polymerase II/genetics , RNA Polymerase II/immunology , RNA Polymerase II/metabolism
10.
J Virol ; 87(19): 10477-88, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23903843

ABSTRACT

Herpes simplex virus 1 (HSV-1) is a ubiquitous and important human pathogen. It is known to persist in trigeminal ganglia (TG), but how it reaches this site has been difficult to determine, as viral transmission is sporadic, pathogenesis is complicated, and early infection is largely asymptomatic. We used mice to compare the most likely natural HSV-1 host entry routes: oral and nasal. Intranasal infection was 100-fold more efficient than oral and targeted predominantly the olfactory neuroepithelium. Live imaging of HSV-1-expressed luciferase showed infection progressing from the nose to the TG and then reemerging in the facial skin. The brain remained largely luciferase negative throughout. Infected cell tagging by viral Cre recombinase expression in floxed reporter gene mice showed nasal virus routinely reaching the TG and only rarely reaching the olfactory bulbs. Thus, HSV-1 spread from the olfactory neuroepithelium to the TG and reemerged peripherally without causing significant neurological disease. This recapitulation of typical clinical infection suggests that HSV-1 might sometimes also enter humans via the respiratory tract.


Subject(s)
Herpes Simplex/virology , Herpesvirus 1, Human/pathogenicity , Neuroepithelial Cells/virology , Olfactory Bulb/virology , Trigeminal Ganglion/virology , Virus Internalization , Animals , Blotting, Western , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Herpes Simplex/genetics , Herpes Simplex/pathology , Humans , Immunoenzyme Techniques , Kidney/metabolism , Kidney/pathology , Kidney/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neuroepithelial Cells/metabolism , Neuroepithelial Cells/pathology , Olfactory Bulb/metabolism , Olfactory Bulb/pathology , Trigeminal Ganglion/metabolism , Trigeminal Ganglion/pathology , Virus Replication
11.
PLoS Pathog ; 8(11): e1002986, 2012.
Article in English | MEDLINE | ID: mdl-23133384

ABSTRACT

Herpesviruses are ubiquitous pathogens that cause much disease. The difficulty of clearing their established infections makes host entry an important target for control. However, while herpesviruses have been studied extensively in vitro, how they cross differentiated mucus-covered epithelia in vivo is unclear. To establish general principles we tracked host entry by Murid Herpesvirus-4 (MuHV-4), a lymphotropic rhadinovirus related to the Kaposi's Sarcoma-associated Herpesvirus. Spontaneously acquired virions targeted the olfactory neuroepithelium. Like many herpesviruses, MuHV-4 binds to heparan sulfate (HS), and virions unable to bind HS showed poor host entry. While the respiratory epithelium expressed only basolateral HS and was bound poorly by incoming virions, the neuroepithelium also displayed HS on its apical neuronal cilia and was bound strongly. Incoming virions tracked down the neuronal cilia, and either infected neurons or reached the underlying microvilli of the adjacent glial (sustentacular) cells and infected them. Thus the olfactory neuroepithelium provides an important and complex site of HS-dependent herpesvirus uptake.


Subject(s)
Heparitin Sulfate/metabolism , Herpesviridae Infections/metabolism , Neuroepithelial Cells/metabolism , Olfactory Bulb/metabolism , Rhadinovirus/metabolism , Virus Internalization , Animals , Cell Line , Cricetinae , Herpesviridae Infections/pathology , Mice , Mice, Inbred BALB C , NIH 3T3 Cells , Neuroepithelial Cells/pathology , Neuroepithelial Cells/virology , Olfactory Bulb/pathology , Olfactory Bulb/virology , Rhadinovirus/pathogenicity
12.
J Gen Virol ; 93(Pt 10): 2118-2130, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22815272

ABSTRACT

Recent RNA interference (RNAi) studies have identified many host proteins that modulate virus infection, but small interfering RNA 'off-target' effects and the use of transformed cell lines limit their conclusiveness. As murine embryonic stem (mES) cells can be genetically modified and resources exist where many and eventually all known mouse genes are insertionally inactivated, it was reasoned that mES cells would provide a useful alternative to RNAi screens. Beyond allowing investigation of host-pathogen interactions in vitro, mES cells have the potential to differentiate into other primary cell types, as well as being used to generate knockout mice for in vivo studies. However, mES cells are poorly characterized for virus infection. To investigate whether ES cells can be used to explore host-virus interactions, this study characterized the responses of mES cells following infection by herpes simplex virus type 1 (HSV-1) and influenza A virus. HSV-1 replicated lytically in mES cells, although mES cells were less permissive than most other cell types tested. Influenza virus was able to enter mES cells and express some viral proteins, but the replication cycle was incomplete and no infectious virus was produced. Knockdown of the host protein AHCYL1 in mES cells reduced HSV-1 replication, showing the potential for using mES cells to study host-virus interactions. Transcriptional profiling, however, indicated the lack of an efficient innate immune response in these cells. mES cells may thus be useful to identify host proteins that play a role in virus replication, but they are not suitable to determine factors that are involved in innate host defence.


Subject(s)
Embryonic Stem Cells/physiology , Embryonic Stem Cells/virology , Influenza A virus/physiology , Orthomyxoviridae Infections/genetics , Virus Replication/genetics , Animals , Cell Line , Cricetinae , DNA Replication/genetics , Dogs , Embryonic Stem Cells/metabolism , HeLa Cells , Herpes Simplex/genetics , Herpes Simplex/metabolism , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/metabolism , Herpesvirus 1, Human/physiology , Host-Pathogen Interactions , Humans , Influenza A virus/genetics , Influenza A virus/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , RNA, Small Interfering/genetics , Transcription, Genetic , Viral Proteins/genetics , Viral Proteins/metabolism
13.
FEMS Microbiol Rev ; 36(3): 684-705, 2012 May.
Article in English | MEDLINE | ID: mdl-22150699

ABSTRACT

Herpes simplex virus type 1 is a neurotropic herpesvirus that establishes latency within sensory neurones. Following primary infection, the virus replicates productively within mucosal epithelial cells and enters sensory neurones via nerve termini. The virus is then transported to neuronal cell bodies where latency can be established. Periodically, the virus can reactivate to resume its normal lytic cycle gene expression programme and result in the generation of new virus progeny that are transported axonally back to the periphery. The ability to establish lifelong latency within the host and to periodically reactivate to facilitate dissemination is central to the survival strategy of this virus. Although incompletely understood, this review will focus on the mechanisms involved in the regulation of latency that centre on the functions of the virus-encoded latency-associated transcripts (LATs), epigenetic regulation of the latent virus genome and the molecular events that precipitate reactivation.


Subject(s)
Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/pathogenicity , Host-Pathogen Interactions , Neurons/immunology , Neurons/virology , Virus Latency , Gene Expression Regulation, Viral , Humans , Transcription, Genetic , Virus Activation
14.
PLoS Pathog ; 7(10): e1002278, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21998580

ABSTRACT

Herpes simplex virus-1 (HSV-1) causes lifelong infection affecting between 50 and 90% of the global population. In addition to causing dermal lesions, HSV-1 is a leading cause of blindness resulting from recurrent corneal infection. Corneal disease is characterized by loss of corneal immunologic privilege and extensive neovascularization driven by vascular endothelial growth factor-A (VEGF-A). In the current study, we identify HSV-1 infected cells as the dominant source of VEGF-A during acute infection, and VEGF-A transcription did not require TLR signaling or MAP kinase activation. Rather than being an innate response to the pathogen, VEGF-A transcription was directly activated by the HSV-1 encoded immediate early transcription factor, ICP4. ICP4 bound the proximal human VEGF-A promoter and was sufficient to promote transcription. Transcriptional activation also required cis GC-box elements common to the VEGF-A promoter and HSV-1 early genes. Our results suggest that the neovascularization characteristic of ocular HSV-1 disease is a direct result of HSV-1's major transcriptional regulator, ICP4, and similarities between the VEGF-A promoter and those of HSV-1 early genes.


Subject(s)
Immediate-Early Proteins/metabolism , Keratitis, Herpetic/pathology , Trans-Activators/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Line , Eye/pathology , Eye/virology , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/metabolism , Herpesvirus 1, Human/pathogenicity , Humans , Immediate-Early Proteins/genetics , Keratitis, Herpetic/virology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence/methods , Neovascularization, Pathologic/genetics , Plasmids , Promoter Regions, Genetic , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA , Trans-Activators/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation , Vascular Endothelial Growth Factor A/genetics
15.
J Gen Virol ; 92(Pt 11): 2575-2585, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21752961

ABSTRACT

Herpes simplex virus (HSV) type-1 establishes lifelong latency in sensory neurones and it is widely assumed that latency is the consequence of a failure to initiate virus immediate-early (IE) gene expression. However, using a Cre reporter mouse system in conjunction with Cre-expressing HSV-1 recombinants we have previously shown that activation of the IE ICP0 promoter can precede latency establishment in at least 30% of latently infected cells. During productive infection of non-neuronal cells, IE promoter activation is largely dependent on the transactivator VP16 a late structural component of the virion. Of significance, VP16 has recently been shown to exhibit altered regulation in neurones; where its de novo synthesis is necessary for IE gene expression during both lytic infection and reactivation from latency. In the current study, we utilized the Cre reporter mouse model system to characterize the full extent of viral promoter activity compatible with cell survival and latency establishment. In contrast to the high frequency activation of representative IE promoters prior to latency establishment, cell marking using a virus recombinant expressing Cre under VP16 promoter control was very inefficient. Furthermore, infection of neuronal cultures with VP16 mutants reveals a strong VP16 requirement for IE promoter activity in non-neuronal cells, but not sensory neurones. We conclude that only IE promoter activation can efficiently precede latency establishment and that this activation is likely to occur through a VP16-independent mechanism.


Subject(s)
Gene Expression Regulation, Viral , Genes, Immediate-Early , Herpes Simplex Virus Protein Vmw65/metabolism , Herpesvirus 1, Human/physiology , Promoter Regions, Genetic , Sensory Receptor Cells/virology , Virus Latency , Animals , Female , Herpesvirus 1, Human/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
16.
PLoS One ; 5(9)2010 Sep 29.
Article in English | MEDLINE | ID: mdl-20927329

ABSTRACT

BACKGROUND: Several alphaherpesviruses, including herpes simplex virus 1 (HSV-1) and pseudorabies virus (PRV), establish lifelong latency in neurons of the trigeminal ganglion (TG). Although it is thought that efficient establishment of alphaherpesvirus latency is based on a subtle interplay between virus, neurons and the immune system, it is not clear which immune components are of major importance for the establishment of latency. METHODOLOGY/PRINCIPAL FINDINGS: Here, using an in vitro model that enables a natural route of infection, we show that interferon alpha (IFNalpha) has the previously uncharacterized capacity to induce a quiescent HSV-1 and PRV infection in porcine TG neurons that shows strong similarity to in vivo latency. IFNalpha induced a stably suppressed HSV-1 and PRV infection in TG neurons in vitro. Subsequent treatment of neurons containing stably suppressed virus with forskolin resulted in reactivation of both viruses. HSV and PRV latency in vivo is often accompanied by the expression of latency associated transcripts (LATs). Infection of TG neurons with an HSV-1 mutant expressing LacZ under control of the LAT promoter showed activation of the LAT promoter and RT-PCR analysis confirmed that both HSV-1 and PRV express LATs during latency in vitro. CONCLUSIONS/SIGNIFICANCE: These data represent a unique in vitro model of alphaherpesvirus latency and indicate that IFNalpha may be a driving force in promoting efficient latency establishment.


Subject(s)
Herpes Simplex/metabolism , Herpesvirus 1, Human/physiology , Herpesvirus 1, Suid/physiology , Interferon-alpha/metabolism , Pseudorabies/metabolism , Sensory Receptor Cells/virology , Trigeminal Ganglion/virology , Virus Latency , Animals , Cell Line , Cells, Cultured , Gene Expression Regulation, Viral , Herpes Simplex/virology , Herpesvirus 1, Human/genetics , Herpesvirus 1, Suid/genetics , Mice , Pseudorabies/virology , Sensory Receptor Cells/metabolism
17.
J Gen Virol ; 91(Pt 9): 2176-85, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20573854

ABSTRACT

Cancers with viral aetiologies can potentially be prevented by antiviral vaccines. Therefore, it is important to understand how viral infections and cancers might be linked. Some cancers frequently carry gammaherpesvirus genomes. However, they generally express the same viral genes as non-transformed cells, and differ mainly in also carrying oncogenic host mutations. Infection, therefore, seems to play a triggering or accessory role in disease. The hit-and-run hypothesis proposes that cumulative host mutations can allow viral genomes to be lost entirely, such that cancers remaining virus-positive represent only a fraction of those to which infection contributes. This would have considerable implications for disease control. However, the hit-and-run hypothesis has so far lacked experimental support. Here, we tested it by using Cre-lox recombination to trigger transforming mutations in virus-infected cells. Thus, 'floxed' oncogene mice were infected with Cre recombinase-positive murid herpesvirus-4 (MuHV-4). The emerging cancers showed the expected genetic changes but, by the time of presentation, almost all lacked viral genomes. Vaccination with a non-persistent MuHV-4 mutant nonetheless conferred complete protection. Equivalent human gammaherpesvirus vaccines could therefore potentially prevent not only viral genome-positive cancers, but possibly also some cancers less suspected of a viral origin because of viral genome loss.


Subject(s)
Cancer Vaccines/pharmacology , Herpesviridae Infections/immunology , Herpesviridae Infections/prevention & control , Rhadinovirus/immunology , Rhadinovirus/pathogenicity , Tumor Virus Infections/immunology , Tumor Virus Infections/prevention & control , Viral Vaccines/pharmacology , Animals , Base Sequence , DNA Primers/genetics , Genes, p53 , Genes, ras , Genome, Viral , Herpesviridae Infections/genetics , Herpesviridae Infections/virology , Humans , Mice , Mice, Mutant Strains , Mice, Transgenic , Models, Biological , Mutagenesis, Insertional , Mutation , Rhadinovirus/genetics , Sarcoma, Experimental/genetics , Sarcoma, Experimental/immunology , Sarcoma, Experimental/prevention & control , Sarcoma, Experimental/virology , Tumor Virus Infections/genetics , Tumor Virus Infections/virology , Vaccination/methods
18.
J Gen Virol ; 91(Pt 4): 867-79, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19940063

ABSTRACT

Two novel gammaherpesviruses were isolated, one from a field vole (Microtus agrestis) and the other from wood mice (Apodemus sylvaticus). The genome of the latter, designated wood mouse herpesvirus (WMHV), was completely sequenced. WMHV had the same genome structure and predicted gene content as murid herpesvirus 4 (MuHV4; murine gammaherpesvirus 68). Overall nucleotide sequence identity between WMHV and MuHV4 was 85 % and most of the 10 kb region at the left end of the unique region was particularly highly conserved, especially the viral tRNA-like sequences and the coding regions of genes M1 and M4. The partial sequence (71 913 bp) of another gammaherpesvirus, Brest herpesvirus (BRHV), which was isolated ostensibly from a white-toothed shrew (Crocidura russula), was also determined. The BRHV sequence was 99.2 % identical to the corresponding portion of the WMHV genome. Thus, WMHV and BRHV appeared to be strains of a new virus species. Biological characterization of WMHV indicated that it grew with similar kinetics to MuHV4 in cell culture. The pathogenesis of WMHV in wood mice was also extremely similar to that of MuHV4, except for the absence of inducible bronchus-associated lymphoid tissue at day 14 post-infection and a higher load of latently infected cells at 21 days post-infection.


Subject(s)
Arvicolinae/virology , Gammaherpesvirinae/classification , Murinae/virology , Rhadinovirus/classification , Amino Acid Sequence , Animals , Base Sequence , DNA, Viral/chemistry , Gammaherpesvirinae/genetics , Gammaherpesvirinae/growth & development , Genome, Viral , Molecular Sequence Data , Rhadinovirus/genetics , Rhadinovirus/growth & development , Viral Matrix Proteins/analysis , Viral Matrix Proteins/genetics
19.
J Gen Virol ; 90(Pt 1): 21-32, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19088269

ABSTRACT

Luciferase-based imaging allows a global view of microbial pathogenesis. We applied this technique to gammaherpesvirus infection by inserting a luciferase expression cassette into the genome of murine herpesvirus-4 (MuHV-4). The recombinant virus strongly expressed luciferase in lytically infected cells without significant attenuation. We used it to compare different routes of virus inoculation. After intranasal infection of anaesthetized mice, luciferase was expressed in the nose and lungs for 7-10 days and in lymphoid tissue, most consistently the superficial cervical lymph nodes, for up to 30 days. Gastrointestinal infection was not observed. Intraperitoneal infection was very different to intranasal, with strong luciferase expression in the liver, kidneys, intestines, reproductive tract and spleen, but none in the nose or lungs. The nose has not previously been identified as a site of MuHV-4 infection. After intranasal infection of non-anaesthetized mice, it was the only site of non-lymphoid luciferase expression. Nevertheless, lymphoid colonization and persistence were still established, even at low inoculation doses. In contrast, virus delivered orally was very poorly infectious. Inoculation route therefore had a major impact on pathogenesis. Low dose intranasal infection without anaesthesia seems most likely to mimic natural transmission, and may therefore be particularly informative about normal viral gene functions.


Subject(s)
Herpesviridae Infections/pathology , Herpesviridae Infections/virology , Rhadinovirus/growth & development , Tumor Virus Infections/pathology , Tumor Virus Infections/virology , Whole Body Imaging , Animal Structures/virology , Animals , Female , Genes, Reporter , Luciferases/genetics , Luciferases/metabolism , Mice , Mice, Inbred BALB C
20.
J Gen Virol ; 89(Pt 12): 2965-2974, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19008381

ABSTRACT

Herpes simplex virus type 1 (HSV-1) has the capacity to establish a life-long latent infection in sensory neurones and also to periodically reactivate from these cells. Since mutant viruses defective for immediate-early (IE) expression retain the capacity for latency establishment it is widely assumed that latency is the consequence of a block in IE gene expression. However, it is not clear whether viral gene expression can precede latency establishment following wild-type virus infection. In order to address this question we have utilized a reporter mouse model system to facilitate a historical analysis of viral promoter activation in vivo. This system utilizes recombinant viruses expressing Cre recombinase under the control of different viral promoters and the Cre reporter mouse strain ROSA26R. In this model, viral promoter-driven Cre recombinase mediates a permanent genetic change, resulting in reporter gene activation and permanent marking of latently infected cells. The analyses of HSV-1 recombinants containing human cytomegalovirus major immediate-early, ICP0, gC or latency-associated transcript promoters linked to Cre recombinase in this system have revealed the existence of a population of neurones that have experienced IE promoter activation prior to the establishment of latency.


Subject(s)
Gene Expression Regulation, Viral , Herpesvirus 1, Human/physiology , Herpesvirus 1, Human/pathogenicity , Neurons/virology , Promoter Regions, Genetic/physiology , Virus Activation , Virus Latency , Animals , Cytomegalovirus/genetics , Cytomegalovirus/metabolism , Ganglia, Spinal/cytology , Ganglia, Spinal/virology , Genes, Reporter , Herpes Simplex/virology , Herpesvirus 1, Human/enzymology , Herpesvirus 1, Human/genetics , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Integrases/genetics , Integrases/metabolism , Mice , Promoter Regions, Genetic/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...