Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Virol Sin ; 38(1): 108-118, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36436797

ABSTRACT

Herpes simplex virus-1 (HSV-1) is a widespread neurotropic virus that can reach the brain and cause a rare but acute herpes simplex encephalitis (HSE) with a high mortality rate. Most patients present with changes in neurological and behavioral status, and survivors suffer long-term neurological sequelae. To date, the pathogenesis leading to brain damage is still not well understood. HSV-1 induced encephalitis in the central nervous system (CNS) in animals are usually very diffuse and progressing rapidly, and mostly fatal, making the analysis difficult. Here, we established a mouse model of HSE via intracerebral inoculation of modified version of neural-attenuated strains of HSV-1 (deletion of ICP34.5 and inserting a strong promoter into the latency-associated transcript region), in which the LMR-αΔpA strain initiated moderate productive infection, leading to strong host immune and inflammatory response characterized by persistent microglia activation. This viral replication activity and prolonged inflammatory response activated signaling pathways in neuronal damage, amyloidosis, Alzheimer's disease, and neurodegeneration, eventually leading to neuronal loss and behavioral changes characterized by hypokinesia. Our study reveals detailed pathogenic processes and persistent inflammatory responses in the CNS and provides a controlled, mild and non-lethal HSE model for studying long-term neuronal injury and increased risk of neurodegenerative diseases due to HSV-1 infection.


Subject(s)
Encephalitis, Herpes Simplex , Herpes Simplex , Herpesvirus 1, Human , Mice , Animals , Herpesvirus 1, Human/physiology , Encephalitis, Herpes Simplex/complications , Encephalitis, Herpes Simplex/pathology , Brain/pathology , Inflammation
3.
Virol J ; 18(1): 26, 2021 01 23.
Article in English | MEDLINE | ID: mdl-33485391

ABSTRACT

BACKGROUND: Herpes Simplex Virus type I (HSV-1) is a large double-stranded DNA virus that enters productive infection in epithelial cells and reorganizes the host nucleus. Cohesin, a major constituent of interphase and mitotic chromosomes comprised of SMC1, SMC3, and SCC1 (Mcd1/Rad21), SCC3 (SA1/SA2), have diverse functions, including sister chromatid cohesion, DNA double-stranded breaks repair, and transcriptional control. Little is known about the role of cohesin in HSV-1 lytic infection. METHODS: We measured the effect on HSV-1 transcription, genome copy number, and viral titer by depleting cohesin components SMC1 or Rad21 using RNAi, followed by immunofluorescence, qPCR, and ChIP experiments to gain insight into cohesin's function in HSV-1 transcription and replication. RESULTS: Here, we report that cohesion subunits SMC1 and Rad21 are recruited to the lytic HSV-1 replication compartment. The knockdown results in decreased viral transcription, protein expression, and maturation of viral replication compartments. SMC1 and Rad21 knockdown leads to the reduced overall RNA pol II occupancy level but increased RNA pol II ser5 phosphorylation binding on viral genes. Consistent with this, the knockdown increased H3K27me3 modification on these genes. CONCLUSIONS: These results suggest that cohesin facilitates HSV-1 lytic transcription by promoting RNA Pol II transcription activity and preventing chromatin's silencing on the viral genome.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Fibroblasts/virology , Herpesvirus 1, Human/genetics , RNA Polymerase II/metabolism , Transcription, Genetic , Cell Cycle Proteins/classification , Cell Cycle Proteins/genetics , Cell Line , Chromosomal Proteins, Non-Histone/classification , Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins/metabolism , Genome, Viral , HeLa Cells , Host Microbial Interactions , Humans , Nuclear Proteins/metabolism , Protein Binding , Cohesins
5.
Virol J ; 17(1): 95, 2020 07 08.
Article in English | MEDLINE | ID: mdl-32641145

ABSTRACT

BACKGROUND: Following acute infection, Herpes Simplex virus-1 (HSV-1) establishes lifelong latency and recurrent reactivation in the sensory neurons of trigeminal ganglia (TG). Infected tree shrew differs from mouse and show characteristics similar to human infection. A detailed transcriptomic analysis of the tree shrew model could provide mechanistic insights into HSV-1 infection in humans. METHODS: We sequenced the transcriptome of infected TGs from tree shrews and mice, and 4 human donors, then examined viral genes expression up to 58 days in infected TGs from mouse and tree shrew, and compare the latency data with that in human TGs. RESULTS: Here, we found that all HSV-1 genes could be detected in mouse TGs during acute infection, but 22 viral genes necessary for viral transcription, replication and viral maturation were not expressed in tree shrew TGs during this stage. Importantly, during latency, we found that LAT could be detected both in mouse and tree shrew, but the latter also has an ICP0 transcript signal absent in mouse but present in human samples. Importantly, we observed that infected human and tree shrew TGs have a more similar LAT region transcription peak. More importantly, we observed that HSV-1 spontaneously reactivates from latently infected tree shrews with relatively high efficiency. CONCLUSIONS: These results represent the first longitudinal transcriptomic characterization of HSV-1 infection in during acute, latency and recurrent phases, and revealed that tree shrew infection has important similar features with human infection.


Subject(s)
Genes, Viral , Herpes Simplex/veterinary , Herpesvirus 1, Human/genetics , Transcriptome , Trigeminal Ganglion/virology , Tupaiidae/virology , Acute Disease , Adult , Animals , Female , Gene Expression , Gene Expression Profiling , Herpes Simplex/virology , Herpesvirus 1, Human/physiology , Humans , Longitudinal Studies , Male , Mice , Mice, Inbred BALB C , RNA-Seq , Viral Proteins/genetics , Virus Latency , Virus Replication
6.
J Neurovirol ; 26(3): 391-403, 2020 06.
Article in English | MEDLINE | ID: mdl-32301037

ABSTRACT

Herpes simplex virus type I (HSV-1) infection causes inflammation in the cornea known as herpes simplex virus keratitis (HSK), a common but serious corneal disease. It is not entirely clear whether the virus during recurring infection comes from the trigeminal ganglia or the eye tissue, including the retina and ciliary ganglion. Because the tree shrew is closely related to primates and tree shrew eye anatomic structures are similar to humans, we studied HSV-1 corneal infection in the tree shrew. We found that HSK symptoms closely mimic those found in human HSK showing typical punctiform and dendritic viral keratitis during the acute infection period. Following the HSV-specific lesions, complications such as stromal scarring, corneal thickening (primary infection), opacity, and neovascularization were observed. In the tree shrew model, following ocular inoculation, the cornea becomes infected, and viral protein can be detected using anti-HSV-1 antibodies in the epithelial layer and retina neuronal ganglion cells. The HSV-1 transcripts, ICP0, ICP4, and LAT can be detected at 3 days post-infection (dpi), peaking at 5 dpi. After 2 weeks, ICP4 and ICP0 transcripts are reduced to a basal level, but the Latency Associated Transcripts (LATs) continue to accumulate. Interestingly, after the acute infection, we still detected abundant active HSV-1 in tree shrew eyes. Further, we found HSV-1 persistent in the ciliary ganglion and cornea. These findings are discussed in support of the tree shrew as a non-human primate HSK model, which could be useful for mechanistic studies of HSK.


Subject(s)
Cornea/virology , Gene Expression Regulation, Viral , Herpes Simplex/virology , Herpesvirus 1, Human/genetics , Keratitis, Herpetic/virology , Neovascularization, Pathologic/virology , Animals , Cornea/pathology , Disease Models, Animal , Female , Ganglia, Parasympathetic/pathology , Ganglia, Parasympathetic/virology , Herpes Simplex/pathology , Herpesvirus 1, Human/growth & development , Herpesvirus 1, Human/metabolism , Herpesvirus 1, Human/pathogenicity , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Keratitis, Herpetic/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Neovascularization, Pathologic/pathology , Neurons/pathology , Neurons/virology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Trigeminal Ganglion/pathology , Trigeminal Ganglion/virology , Tupaia , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Virus Latency
7.
J Virol ; 92(16)2018 08 15.
Article in English | MEDLINE | ID: mdl-29899087

ABSTRACT

Herpes simplex virus 1 (HSV-1) is a prevalent human pathogen that infects the cornea, causing potentially blinding herpetic disease. A clinical herpes vaccine is still lacking. In the present study, a novel prime/pull vaccine was tested in a human leukocyte antigen (HLA) transgenic rabbit model of ocular herpes (HLA Tg rabbits). Three peptide epitopes were selected, from the HSV-1 membrane glycoprotein C (UL44400-408), the DNA replication binding helicase (UL9196-204), and the tegument protein (UL25572-580), all preferentially recognized by CD8+ T cells from "naturally protected" HSV-1-seropositive healthy asymptomatic (ASYMP) individuals (who never had recurrent corneal herpetic disease). HLA Tg rabbits were immunized with a mixture of these three ASYMP CD8+ T cell peptide epitopes (UL44400-408, UL9196-204, and UL25572-580), which were delivered subcutaneously with CpG2007 adjuvant (prime). Fifteen days later, half of the rabbits received a topical ocular treatment with a recombinant neurotropic adeno-associated virus type 8 (AAV8) vector expressing the T cell-attracting CXCL10 chemokine (pull). The frequency and function of HSV-specific CD8+ T cells induced by the prime/pull vaccine were assessed in the peripheral blood, cornea, and trigeminal ganglion (TG). Compared to the cells generated in response to peptide immunization alone, the peptide/CXCL10 prime/pull vaccine generated frequent polyfunctional gamma interferon-positive (IFN-γ+) CD107+ CD8+ T cells that infiltrated both the cornea and TG. CD8+ T cell mobilization into the cornea and TG of prime/pull-vaccinated rabbits was associated with a significant reduction in corneal herpesvirus infection and disease following an ocular HSV-1 (strain McKrae) challenge. These findings draw attention to the novel prime/pull vaccine strategy for mobilizing antiviral CD8+ T cells into tissues to protect against herpesvirus infection and disease.IMPORTANCE There is an urgent need for a vaccine against widespread herpes simplex virus infections. The present study demonstrates that immunization of HLA transgenic rabbits with a peptide/CXCL10 prime/pull vaccine triggered mobilization of HSV-specific CD8+ T cells locally into the cornea and TG, the sites of acute and latent herpesvirus infections, respectively. Mobilization of antiviral CD8+ T cells into the cornea and TG of rabbits that received the prime/pull vaccine was associated with protection against ocular herpesvirus infection and disease following an ocular HSV-1 challenge. These results highlight the importance of the prime/pull vaccine strategy to bolster the number and function of protective CD8+ T cells within infected tissues.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chemokine CXCL10/metabolism , Cornea/immunology , Herpes Simplex Virus Vaccines/immunology , Keratitis, Herpetic/prevention & control , T-Lymphocyte Subsets/immunology , Trigeminal Ganglion/immunology , Animals , Animals, Genetically Modified , Chemokine CXCL10/administration & dosage , Disease Models, Animal , Epitopes/immunology , HLA Antigens/genetics , HLA Antigens/metabolism , Herpes Simplex Virus Vaccines/administration & dosage , Humans , Interferon-gamma/analysis , Keratitis, Herpetic/pathology , Keratitis, Herpetic/virology , Lysosomal-Associated Membrane Protein 1/analysis , Rabbits , Simplexvirus/immunology , Simplexvirus/isolation & purification , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Viral Load
8.
Viruses ; 10(5)2018 05 02.
Article in English | MEDLINE | ID: mdl-29724057

ABSTRACT

Herpes simplex virus type 1(HSV-1) presents a conundrum to public health worldwide because of its specific pathogenicity and clinical features. Some experimental vaccines, such as the recombinant viral glycoproteins, exhibit the viral immunogenicity of a host-specific immune response, but none of these has achieved a valid epidemiological protective efficacy in the human population. In the present study, we constructed an attenuated HSV-1 strain M3 through the partial deletion of UL7, UL41, and the latency-associated transcript (LAT) using the CRISPR/Cas9 system. The mutant strain exhibited lowered infectivity and virulence in macaques. Neutralization testing and ELISpot detection of the specific T-cell responses confirmed the specific immunity induced by M3 immunization and this immunity defended against the challenges of the wild-type strain and restricted the entry of the wild-type strain into the trigeminal ganglion. These results in rhesus macaques demonstrated the potential of the attenuated vaccine for the prevention of HSV-1 in humans.


Subject(s)
Antigens, Viral/immunology , Herpes Simplex/prevention & control , Herpesvirus 1, Human/genetics , Herpesvirus Vaccines/immunology , Animals , Antigens, Viral/genetics , CRISPR-Cas Systems , Herpes Simplex/immunology , Herpesvirus Vaccines/genetics , Immunogenicity, Vaccine , Macaca mulatta/virology , Mutation , Neutralization Tests , Phenotype , Trigeminal Ganglion/virology , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Viral Matrix Proteins/genetics , Viral Matrix Proteins/immunology , Viral Proteins/genetics , Viral Proteins/immunology , Virus Replication
9.
Sci Rep ; 7: 39861, 2017 01 03.
Article in English | MEDLINE | ID: mdl-28045091

ABSTRACT

CTCF is an essential chromatin regulator implicated in important nuclear processes including in nuclear organization and transcription. Herpes Simplex Virus-1 (HSV-1) is a ubiquitous human pathogen, which enters productive infection in human epithelial and many other cell types. CTCF is known to bind several sites in the HSV-1 genome during latency and reactivation, but its function has not been defined. Here, we report that CTCF interacts extensively with the HSV-1 DNA during lytic infection by ChIP-seq, and its knockdown results in the reduction of viral transcription, viral genome copy number and virus yield. CTCF knockdown led to increased H3K9me3 and H3K27me3, and a reduction of RNA pol II occupancy on viral genes. Importantly, ChIP-seq analysis revealed that there is a higher level of CTD Ser2P modified RNA Pol II near CTCF peaks relative to the Ser5P form in the viral genome. Consistent with this, CTCF knockdown reduced the Ser2P but increased Ser5P modified forms of RNA Pol II on viral genes. These results suggest that CTCF promotes HSV-1 lytic transcription by facilitating the elongation of RNA Pol II and preventing silenced chromatin on the viral genome.


Subject(s)
CCCTC-Binding Factor/metabolism , Genome, Viral , Herpesvirus 1, Human/genetics , RNA, Viral/genetics , Virus Replication , Animals , Chlorocebus aethiops , Chromatin Assembly and Disassembly , HEK293 Cells , HeLa Cells , Herpesvirus 1, Human/metabolism , Herpesvirus 1, Human/pathogenicity , Histones/genetics , Histones/metabolism , Humans , Protein Binding , RNA Polymerase II/metabolism , RNA, Viral/metabolism , Transcription, Genetic , Vero Cells
11.
Sci Rep ; 6: 28075, 2016 06 29.
Article in English | MEDLINE | ID: mdl-27354008

ABSTRACT

Pathogen invasion triggers a number of cellular responses and alters the host transcriptome. Here we report that the type of changes to cellular transcriptome is related to the type of cellular functions affected by lytic infection of Herpes Simplex Virus type I in Human primary fibroblasts. Specifically, genes involved in stress responses and nuclear transport exhibited mostly changes in alternative polyadenylation (APA), cell cycle genes showed mostly alternative splicing (AS) changes, while genes in neurogenesis, rarely underwent these changes. Transcriptome wide, the infection resulted in 1,032 cases of AS, 161 incidences of APA, 1,827 events of isoform changes, and up regulation of 596 genes and down regulations of 61 genes compared to uninfected cells. Thus, these findings provided important and specific links between cellular responses to HSV-1 infection and the type of alterations to the host transcriptome, highlighting important roles of RNA processing in virus-host interactions.


Subject(s)
Fibroblasts/metabolism , Gene Expression Regulation , Herpes Simplex/metabolism , Herpesvirus 1, Human/metabolism , Transcriptome , Cell Line , Fibroblasts/virology , Herpes Simplex/genetics , Herpes Simplex/pathology , Herpesvirus 1, Human/genetics , Humans
12.
J Neurovirol ; 22(3): 293-306, 2016 06.
Article in English | MEDLINE | ID: mdl-26501779

ABSTRACT

Herpes Simplex Virus type I (HSV-1) latently infects peripheral nervous system (PNS) sensory neurons, and its reactivation leads to recurring cold sores. The reactivated HSV-1 can travel retrograde from the PNS into the central nervous system (CNS) and is known to be causative of Herpes Simplex viral encephalitis. HSV-1 infection in the PNS is well documented, but little is known on the fate of HSV-1 once it enters the CNS. In the murine model, HSV-1 genome persists in the CNS once infected through an ocular route. To gain more details of HSV-1 infection in the CNS, we characterized HSV-1 infection of the tree shrew (Tupaia belangeri chinensis) brain following ocular inoculation. Here, we report that HSV-1 enters the tree shrew brain following ocular inoculation and HSV-1 transcripts, ICP0, ICP4, and LAT can be detected at 5 days post-infection (p.i.), peaking at 10 days p.i. After 2 weeks, ICP4 and ICP0 transcripts are reduced to a basal level, but the LAT intron region continues to be expressed. Live virus could be recovered from the olfactory bulb and brain stem tissue. Viral proteins could be detected using anti-HSV-1 antibodies and anti-ICP4 antibody, during the acute stage but not beyond. In situ hybridization could detect LAT during acute infection in most brain regions and in olfactory bulb and brain stem tissue well beyond the acute stage. Using a homogenate from these tissues' post-acute infection, we did not recover live HSV-1 virus, supporting a latent infection, but using a modified explant cocultivation technique, we were able to recover reactivated virus from these tissues, suggesting that the HSV-1 virus latently infects the tree shrew CNS. Compared to mouse, the CNS acute infection of the tree shrew is delayed and the olfactory bulb contains most latent virus. During the acute stage, a portion of the infected tree shrews exhibit symptoms similar to human viral encephalitis. These findings, together with the fact that tree shrews are closely related to primates, provided a valuable alternative model to study HSV-1 infection and pathogenesis in the CNS.


Subject(s)
Encephalitis, Herpes Simplex/virology , Gene Expression Regulation, Viral , Herpesvirus 1, Human/genetics , Trigeminal Ganglion/virology , Virus Activation , Virus Latency , Animals , Brain Stem/metabolism , Brain Stem/pathology , Brain Stem/virology , Disease Models, Animal , Encephalitis, Herpes Simplex/genetics , Encephalitis, Herpes Simplex/metabolism , Encephalitis, Herpes Simplex/pathology , Female , Herpesvirus 1, Human/metabolism , Herpesvirus 1, Human/pathogenicity , Host Specificity , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Mice , Mice, Inbred BALB C , MicroRNAs/genetics , MicroRNAs/metabolism , Olfactory Bulb/metabolism , Olfactory Bulb/pathology , Olfactory Bulb/virology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tissue Culture Techniques , Trigeminal Ganglion/metabolism , Trigeminal Ganglion/pathology , Tupaiidae , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
13.
J Virol ; 90(2): 790-804, 2016 01 15.
Article in English | MEDLINE | ID: mdl-26512084

ABSTRACT

UNLABELLED: Studies of herpes simplex virus (HSV) infections of humans are limited by the use of rodent models such as mice, rabbits, and guinea pigs. Tree shrews (Tupaia belangeri chinensis) are small mammals indigenous to southwest Asia. At behavioral, anatomical, genomic, and evolutionary levels, tree shrews are much closer to primates than rodents are, and tree shrews are susceptible to HSV infection. Thus, we have studied herpes simplex virus 1 (HSV-1) infection in the tree shrew trigeminal ganglion (TG) following ocular inoculation. In situ hybridization, PCR, and quantitative reverse transcription-PCR (qRT-PCR) analyses confirm that HSV-1 latently infects neurons of the TG. When explant cocultivation of trigeminal ganglia was performed, the virus was recovered after 5 days of cocultivation with high efficiency. Swabbing the corneas of latently infected tree shrews revealed that tree shrews shed virus spontaneously at low frequencies. However, tree shrews differ significantly from mice in the expression of key HSV-1 genes, including ICP0, ICP4, and latency-associated transcript (LAT). In acutely infected tree shrew TGs, no level of ICP4 was observed, suggesting the absence of infection or a very weak, acute infection compared to that of the mouse. Immunofluorescence staining with ICP4 monoclonal antibody, and immunohistochemistry detection by HSV-1 polyclonal antibodies, showed a lack of viral proteins in tree shrew TGs during both acute and latent phases of infection. Cultivation of supernatant from homogenized, acutely infected TGs with RS1 cells also exhibited an absence of infectious HSV-1 from tree shrew TGs. We conclude that the tree shrew has an undetectable, or a much weaker, acute infection in the TGs. Interestingly, compared to mice, tree shrew TGs express high levels of ICP0 transcript in addition to LAT during latency. However, the ICP0 transcript remained nuclear, and no ICP0 protein could be seen during the course of mouse and tree shrew TG infections. Taken together, these observations suggest that the tree shrew TG infection differs significantly from the existing rodent models. IMPORTANCE: Herpes simplex viruses (HSVs) establish lifelong infection in more than 80% of the human population, and their reactivation leads to oral and genital herpes. Currently, rodent models are the preferred models for latency studies. Rodents are distant from primates and may not fully represent human latency. The tree shrew is a small mammal, a prosimian primate, indigenous to southwest Asia. In an attempt to further develop the tree shrew as a useful model to study herpesvirus infection, we studied the establishment of latency and reactivation of HSV-1 in tree shrews following ocular inoculation. We found that the latent virus, which resides in the sensory neurons of the trigeminal ganglion, could be stress reactivated to produce infectious virus, following explant cocultivation and that spontaneous reactivation could be detected by cell culture of tears. Interestingly, the tree shrew model is quite different from the mouse model of HSV infection, in that the virus exhibited only a mild acute infection following inoculation with no detectable infectious virus from the sensory neurons. The mild infection may be more similar to human infection in that the sensory neurons continue to function after herpes reactivation and the affected skin tissue does not lose sensation. Our findings suggest that the tree shrew is a viable model to study HSV latency.


Subject(s)
Herpesvirus 1, Human/physiology , Transcription, Genetic , Trigeminal Ganglion/virology , Tupaiidae/virology , Virus Latency , Virus Replication , Animals , Disease Models, Animal , Female , Gene Expression Profiling , Herpesviridae , Mice, Inbred BALB C , Viral Proteins/biosynthesis , Virus Shedding
14.
Mol Ther Methods Clin Dev ; 2: 14059, 2015.
Article in English | MEDLINE | ID: mdl-26052529

ABSTRACT

Genetic diseases of the brain usually have pathologic lesions distributed throughout, thus requiring global correction. Herpes simplex virus-1 (HSV-1) vectors may be especially useful for gene delivery in these disorders since they can spread trans-synaptically along neuronal pathways to distal sites from a localized injection. We have previously shown that a nonpathogenic HSV-1 (strain 1716), which is deleted in the ICP34.5 gene, and expressing the lysosomal enzyme ß-glucuronidase (GUSB) from the latency-associated transcript (LAT) promoter, spreads within the brains of GUSB-deficient mucopolysaccharidosis VII mice to reverse the pathognomonic storage lesions throughout the diseased brain. In this study, we tested the ability of the 1716 LAT-GUSB vector to improve behavioral deficits. The treatment significantly decreased anxiogenic behaviors associated with the mutation, as indicated by open-field behavior and decreased neophobia in a novel object-recognition task. The treated mice also exhibited an improvement in cognitive function associated with the cerebral cortex in a familiar object test. The results indicate the functional therapeutic potential of the 1716 LAT-GUSB vector.

15.
PLoS One ; 10(2): e0117471, 2015.
Article in English | MEDLINE | ID: mdl-25710170

ABSTRACT

HSV is a large double stranded DNA virus, capable of causing a variety of diseases from the common cold sore to devastating encephalitis. Although DNA within the HSV virion does not contain any histone protein, within 1 h of infecting a cell and entering its nucleus the viral genome acquires some histone protein (nucleosomes). During lytic infection, partial micrococcal nuclease (MNase) digestion does not give the classic ladder band pattern, seen on digestion of cell DNA or latent viral DNA. However, complete digestion does give a mono-nucleosome band, strongly suggesting that there are some nucleosomes present on the viral genome during the lytic infection, but that they are not evenly positioned, with a 200 bp repeat pattern, like cell DNA. Where then are the nucleosomes positioned? Here we perform HSV-1 genome wide nucleosome mapping, at a time when viral replication is in full swing (6 hr PI), using a microarray consisting of 50mer oligonucleotides, covering the whole viral genome (152 kb). Arrays were probed with MNase-protected fragments of DNA from infected cells. Cells were not treated with crosslinking agents, thus we are only mapping tightly bound nucleosomes. The data show that nucleosome deposition is not random. The distribution of signal on the arrays suggest that nucleosomes are located at preferred positions on the genome, and that there are some positions that are not occupied (nucleosome free regions -NFR or Nucleosome depleted regions -NDR), or occupied at frequency below our limit of detection in the population of genomes. Occupancy of only a fraction of the possible sites may explain the lack of a typical MNase partial digestion band ladder pattern for HSV DNA during lytic infection. On average, DNA encoding Immediate Early (IE), Early (E) and Late (L) genes appear to have a similar density of nucleosomes.


Subject(s)
Genome, Viral , Herpesvirus 1, Human/genetics , Nucleosomes/metabolism , Carbocyanines/chemistry , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Nucleus/virology , Cluster Analysis , Comparative Genomic Hybridization , DNA Probes/metabolism , DNA, Viral/metabolism , Genes, Immediate-Early , Herpesvirus 1, Human/physiology , Humans , Micrococcal Nuclease/metabolism , Nucleosomes/chemistry , Virus Replication/genetics
16.
J Neurovirol ; 21(4): 358-69, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25672886

ABSTRACT

Herpes simplex virus (HSV) is a double-stranded DNA virus that can cause lytic infections in epithelial cells of the skin and latent infections in neuronal cells of the peripheral nervous system. After virion attachment to the cell membrane, the capsid enters the cytoplasm and is transported to the nucleus. Following docking at the nuclear pore, the HSV DNA, and contents of the virion, are injected into the nucleus. The viral DNA that enters the nucleus is devoid of histones, but begins to be covered with them soon after entry. The covering of histones, in the form of nucleosomes, reaches a maximum during the early stages of infection and drops off during late infection (after DNA replication). However, during latency, the genome is saturated with nucleosomes. In this study, we examine the role of proliferating cell nuclear antigen (PCNA), a cellular DNA polymerase accessory protein (processivity factor), and cell DNA polymerases in histone deposition during the early stages of HSV infection. Using SiRNA knockdown, and a cytosine arabinoside (araC) chemical inhibitor, we conclude that PCNA is important for viral replication and histone deposition. However, cell DNA polymerases that bind PCNA do not appear to be required for these processes and PCNA does not appear to bind to the viral DNA polymerase (which has its own viral processivity factor).


Subject(s)
Herpesvirus 1, Human/physiology , Host-Parasite Interactions/physiology , Proliferating Cell Nuclear Antigen/metabolism , Virus Replication/physiology , Animals , Blotting, Southern , Chlorocebus aethiops , Chromatin Immunoprecipitation , DNA, Viral , Herpes Simplex/virology , Nucleosomes , Reverse Transcriptase Polymerase Chain Reaction , Vero Cells
17.
Cell Rep ; 9(6): 2263-78, 2014 Dec 24.
Article in English | MEDLINE | ID: mdl-25497088

ABSTRACT

Telomeres protect the ends of cellular chromosomes. We show here that infection with herpes simplex virus 1 (HSV-1) results in chromosomal structural aberrations at telomeres and the accumulation of telomere dysfunction-induced DNA damage foci (TIFs). At the molecular level, HSV-1 induces transcription of telomere repeat-containing RNA (TERRA), followed by the proteolytic degradation of the telomere protein TPP1 and loss of the telomere repeat DNA signal. The HSV-1-encoded E3 ubiquitin ligase ICP0 is required for TERRA transcription and facilitates TPP1 degradation. Small hairpin RNA (shRNA) depletion of TPP1 increases viral replication, indicating that TPP1 inhibits viral replication. Viral replication protein ICP8 forms foci that coincide with telomeric proteins, and ICP8-null virus failed to degrade telomere DNA signal. These findings suggest that HSV-1 reorganizes telomeres to form ICP8-associated prereplication foci and to promote viral genomic replication.


Subject(s)
Herpesvirus 1, Human/physiology , Telomere/virology , Virus Replication , Cell Line , Chromosome Aberrations , DNA Damage , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Herpesvirus 1, Human/metabolism , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Proteolysis , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Repetitive Sequences, Nucleic Acid , Serine Proteases/genetics , Serine Proteases/metabolism , Shelterin Complex/metabolism , Telomere/chemistry , Telomere/genetics , Telomere-Binding Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
18.
J Am Assoc Lab Anim Sci ; 52(3): 277-85, 2013.
Article in English | MEDLINE | ID: mdl-23849410

ABSTRACT

Herpes simplex virus (HSV) has been studied in well-established mouse models to generate latently infected animals for investigations into viral pathogenesis, latency mechanisms, and reactivation. Mice exhibit clinical signs of debilitating infection, during which time they may become severely ill before recovery or die spontaneously. Because the cohort of mice that does survive provides valuable data on latency, there is keen interest in developing methodologies for earlier detection and treatment of severe disease to ultimately increase survival rates. Here, BALB/c mice were inoculated ocularly with either a wildtype (LAT(+)) or mutant (LAT(-)) strain of HSV1. Mice were monitored daily through day 30 after infection; trigeminal ganglia were harvested at day 60 to assess viral DNA load. Cages were provided with nesting material, and fluid supplementation was administered to mice with body temperatures of 35 °C or lower, as measured by subcutaneous microchip thermometry. The results showed that infected mice with temperatures less than 34.5 °C did not recover to normothermia and were euthanized or spontaneously died, regardless of infective viral strain. By using a combination of criteria including body temperature (less than 34.5 °C) and weight loss (more than 0.05 g daily) for removal of animals from the study, approximately 98% of mice that died spontaneously could have been euthanized prior to death, without concern of potential recovery to the experimental endpoint (100% specificity). Frequent monitoring of alterations to general wellbeing, body temperature, and weight was crucial for establishing humane endpoints in this ocular HSV model.


Subject(s)
Animal Welfare , Disease Models, Animal , Herpesviridae Infections/pathology , Herpesvirus 1, Human/physiology , Mice, Inbred BALB C , Animals , Body Temperature , DNA, Viral/analysis , Disease Progression , Eye Infections/pathology , Eye Infections/virology , Female , Guidelines as Topic , Herpesviridae Infections/virology , Mice , Trigeminal Ganglion/virology , Virus Latency , Weight Loss
19.
J Neurovirol ; 19(1): 57-64, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23354549

ABSTRACT

Although the induction of the cellular DNA damage response by herpes simplex virus-1 (HSV-1) infection of epithelial cells in tissue culture promotes productive infection, there has been no experimental observation of the effect of the cellular DNA damage response on HSV-1 infection in vivo or in neuronal derived cell lines in tissue culture. Thus, it has been speculated that the lack of cellular DNA damage induction during infection of neurons may promote latency in these cells. This work examines the profile of HSV-1 promoter induction and protein expression, in the absence or presence of infection; using cellular DNA damage inducing topoisomerase inhibitors (Camptothecin and Etoposide) on a neuroblastoma cell line (C1300) in which HSV-1 infection fails to induce the DNA damage response. In the absence of infection, a plasmid expressing the immediate early ICP0 promoter was the most induced by the DNA damage drug treatments compared to the early (RR) and late (VP16) gene promoters. Similarly, drug treatment of C1300 cells infected with HSV-1 virus showed enhanced protein expression for ICP0, but not ICP4 and VP16 proteins. However, when the cells were infected with a HSV-1 virus defective in the immediate early gene trans-activator VP16 (in814) and treated with the DNA damaging drugs, there was enhanced expression of immediate early and late HSV-1 proteins. Although, viral infection of the neuroblastoma cell alone did not induce DNA damage, cellular DNA damage induced by drug treatments facilitated viral promoter induction and viral protein expression. This implicates a mechanism by which HSV-1 viral genes in a quiescent or latent state may become induced by cellular DNA damage in neuronal cells to facilitate productive infection.


Subject(s)
DNA Damage/genetics , Gene Expression Regulation, Viral/genetics , Herpesvirus 1, Human/physiology , Neurons/virology , Virus Activation/genetics , Virus Latency/genetics , Blotting, Western , Cell Line, Tumor , Genes, Viral/genetics , Herpes Simplex/genetics , Humans , Neuroblastoma , Promoter Regions, Genetic , Transfection , Viral Proteins/genetics
20.
J Neurovirol ; 19(1): 102-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23335177

ABSTRACT

Herpes simplex virus type 1 establishes a latent infection in the sensory neurons of the peripheral nervous system of humans. Although about 80 genes are expressed during the lytic cycle of the virus infection, essentially only one gene is expressed during the latent cycle. This gene is known as the latency-associated transcript (LAT), and it appears to play a role in the latency cycle through an anti-apoptotic function in the 5' end of the gene and miRNA encoded along the length of the transcript which downregulate some of the viral immediate-early gene products. The LAT gene is about 8.3 kb long and consists of two exons separated by an unusual intron. The intron between the exons consists of two nested introns. This arrangement of introns has been called a twintron. Furthermore, the larger (2 kb) intron has been shown to be very stable. In this study, we measure the stability of the shorter 1.5-kb nested intron and find its half-life is similar to the longer intron. This was achieved by deleting the 0.5-kb overlapping intron from a plasmid construct designed to express the LAT transcript from a tet-inducible promoter and measuring the half-life of the 1.5-kb intron in tissue culture cells. This finding supports the hypothesis that it is the common branch-point region of these nested introns that is responsible for their stability.


Subject(s)
Herpesvirus 1, Human/genetics , Introns/genetics , MicroRNAs/genetics , RNA Stability/genetics , Blotting, Northern , Half-Life , RNA/analysis , RNA/genetics
SELECTION OF CITATIONS
SEARCH DETAIL