Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
Nucleic Acids Res ; 49(6): 3217-3241, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33675667

ABSTRACT

Epstein-Barr virus (EBV), a herpes virus also termed HHV 4 and the first identified human tumor virus, establishes a stable, long-term latent infection in human B cells, its preferred host. Upon induction of EBV's lytic phase, the latently infected cells turn into a virus factory, a process that is governed by EBV. In the lytic, productive phase, all herpes viruses ensure the efficient induction of all lytic viral genes to produce progeny, but certain of these genes also repress the ensuing antiviral responses of the virally infected host cells, regulate their apoptotic death or control the cellular transcriptome. We now find that EBV causes previously unknown massive and global alterations in the chromatin of its host cell upon induction of the viral lytic phase and prior to the onset of viral DNA replication. The viral initiator protein of the lytic cycle, BZLF1, binds to >105 binding sites with different sequence motifs in cellular chromatin in a concentration dependent manner implementing a binary molar switch probably to prevent noise-induced erroneous induction of EBV's lytic phase. Concomitant with DNA binding of BZLF1, silent chromatin opens locally as shown by ATAC-seq experiments, while previously wide-open cellular chromatin becomes inaccessible on a global scale within hours. While viral transcripts increase drastically, the induction of the lytic phase results in a massive reduction of cellular transcripts and a loss of chromatin-chromatin interactions of cellular promoters with their distal regulatory elements as shown in Capture-C experiments. Our data document that EBV's lytic cycle induces discrete early processes that disrupt the architecture of host cellular chromatin and repress the cellular epigenome and transcriptome likely supporting the efficient de novo synthesis of this herpes virus.


Subject(s)
Chromatin/virology , Gene Expression Regulation , Herpesvirus 4, Human/physiology , Trans-Activators/metabolism , Transcriptome , Binding Sites , Cell Line , Chromatin/chemistry , Chromatin/metabolism , DNA/metabolism , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/metabolism , Humans
2.
Proc Natl Acad Sci U S A ; 117(18): 10003-10014, 2020 05 05.
Article in English | MEDLINE | ID: mdl-32300008

ABSTRACT

Transcription factors (TFs) enact precise regulation of gene expression through site-specific, genome-wide binding. Common methods for TF-occupancy profiling, such as chromatin immunoprecipitation, are limited by requirement of TF-specific antibodies and provide only end-point snapshots of TF binding. Alternatively, TF-tagging techniques, in which a TF is fused to a DNA-modifying enzyme that marks TF-binding events across the genome as they occur, do not require TF-specific antibodies and offer the potential for unique applications, such as recording of TF occupancy over time and cell type specificity through conditional expression of the TF-enzyme fusion. Here, we create a viral toolkit for one such method, calling cards, and demonstrate that these reagents can be delivered to the live mouse brain and used to report TF occupancy. Further, we establish a Cre-dependent calling cards system and, in proof-of-principle experiments, show utility in defining cell type-specific TF profiles and recording and integrating TF-binding events across time. This versatile approach will enable unique studies of TF-mediated gene regulation in live animal models.


Subject(s)
Chromatin/genetics , DNA Transposable Elements/genetics , DNA-Binding Proteins/genetics , Epigenomics/methods , Transcription Factors/genetics , Algorithms , Animals , Antibodies/genetics , Binding Sites/genetics , Chromatin/virology , Dependovirus/genetics , Gene Expression Regulation/genetics , Genome/genetics , Humans , Integrases/genetics , Mice , Tissue Distribution/genetics
3.
PLoS Pathog ; 16(1): e1008264, 2020 01.
Article in English | MEDLINE | ID: mdl-31999790

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) infection is a chronic condition, where viral DNA integrates into the genome. Latently infected cells form a persistent, heterogeneous reservoir that at any time can reactivate the integrated HIV-1. Here we confirmed that latently infected cells from HIV-1 positive study participants exhibited active HIV-1 transcription but without production of mature spliced mRNAs. To elucidate the mechanisms behind this we employed primary HIV-1 latency models to study latency establishment and maintenance. We characterized proviral transcription and chromatin development in cultures of resting primary CD4+ T-cells for four months after ex vivo HIV-1 infection. As heterochromatin (marked with H3K9me3 or H3K27me3) gradually stabilized, the provirus became less accessible with reduced activation potential. In a subset of infected cells, active marks (e.g. H3K27ac) and elongating RNAPII remained detectable at the latent provirus, despite prolonged proviral silencing. In many aspects, latent HIV-1 resembled an active enhancer in a subset of resting cells. The enhancer chromatin actively promoted latency and the enhancer-specific CBP/P300-inhibitor GNE049 was identified as a new latency reversal agent. The division of the latent reservoir according to distinct chromatin compositions with different reactivation potential enforces the notion that even though a relatively large set of cells contains the HIV-1 provirus, only a discrete subset is readily able to reactivate the provirus and spread the infection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Chromatin/virology , HIV Infections/virology , HIV-1/physiology , Proviruses/physiology , CD4-Positive T-Lymphocytes/virology , HIV Infections/immunology , HIV-1/genetics , Humans , Proviruses/genetics , Virus Activation , Virus Assembly , Virus Latency
4.
Virus Genes ; 57(5): 459-463, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34185196

ABSTRACT

Baculovirus infection modulates the chromatin states and gene expression of host insect cells. Here we performed chromatin immunoprecipitation followed by deep sequencing (ChIP-seq) of H3 trimethylated at Lys4 (H3K4me3) histone modification in Bombyx mori nucleopolyhedrovirus-infected Bombyx mori cells. The ChIP-seq data revealed the changes of the genome-wide distribution and accumulation of euchromatic histone marks in host insect cells during the progression of baculovirus infection.


Subject(s)
Bombyx/genetics , Chromatin/genetics , Histones/genetics , Nucleopolyhedroviruses/genetics , Animals , Baculoviridae/genetics , Baculoviridae/pathogenicity , Bombyx/virology , Chromatin/virology , Gene Expression Regulation/genetics , High-Throughput Nucleotide Sequencing , Histone Code/genetics , Nucleopolyhedroviruses/pathogenicity , Protein Processing, Post-Translational/genetics
5.
Nucleic Acids Res ; 47(7): 3607-3618, 2019 04 23.
Article in English | MEDLINE | ID: mdl-30767014

ABSTRACT

The integration of the retroviral genome into the chromatin of the infected cell is catalysed by the integrase (IN)•viral DNA complex (intasome). This process requires functional association between the integration complex and the nucleosomes. Direct intasome/histone contacts have been reported to modulate the interaction between the integration complex and the target DNA (tDNA). Both prototype foamy virus (PFV) and HIV-1 integrases can directly bind histone amino-terminal tails. We have further investigated this final association by studying the effect of isolated histone tails on HIV-1 integration. We show here that the binding of HIV-1 IN to a peptide derived from the H4 tail strongly stimulates integration catalysis in vitro. This stimulation was not observed with peptide tails from other variants or with alpha-retroviral (RAV) and spuma-retroviral PFV integrases. Biochemical analyses show that the peptide tail induces both an increase in the IN oligomerization state and affinity for the target DNA, which are associated with substantial structural rearrangements in the IN carboxy-terminal domain (CTD) observed by NMR. Our data indicate that the H4 peptide tail promotes the formation of active strand transfer complexes (STCs) and support an activation step of the incoming intasome at the contact of the histone tail.


Subject(s)
HIV Integrase/genetics , HIV-1/genetics , Histones/genetics , Virus Integration/genetics , Catalysis , Chromatin/genetics , Chromatin/virology , Genome, Viral/genetics , HIV-1/pathogenicity , Host-Pathogen Interactions/genetics , Humans , Nucleosomes/genetics , Nucleosomes/virology , Spumavirus/genetics
6.
J Virol ; 93(10)2019 05 15.
Article in English | MEDLINE | ID: mdl-30842325

ABSTRACT

Human adenovirus expresses several early proteins that control various aspects of the viral replication program, including an orchestrated expression of viral genes. Two of the earliest viral transcriptional units activated after viral genome entry into the host cell nucleus are the E1 and E4 units, which each express a variety of proteins. Chief among these are the E1A proteins that function to reprogram the host cell and activate transcription of all other viral genes. The E4 gene encodes multiple proteins, including E4orf3, which functions to disrupt cellular antiviral defenses, including the DNA damage response pathway and activation of antiviral genes. Here we report that E1A directly interacts with E4orf3 via the conserved N terminus of E1A to regulate the expression of viral genes. We show that E4orf3 indiscriminately drives high nucleosomal density of viral genomes, which is restrictive to viral gene expression and which E1A overcomes via a direct interaction with E4orf3. We also show that during infection E1A colocalizes with E4orf3 to nuclear tracks that are associated with heterochromatin formation. The inability of E1A to interact with E4orf3 has a significant negative impact on overall viral replication, the ability of the virus to reprogram the host cell, and the levels of viral gene expression. Together these results show that E1A and E4orf3 work together to fine-tune the viral replication program during the course of infection and highlight a novel mechanism that regulates viral gene expression.IMPORTANCE To successfully replicate, human adenovirus needs to carry out a rapid yet ordered transcriptional program that executes and drives viral replication. Early in infection, the viral E1A proteins are the key activators and regulators of viral transcription. Here we report, for the first time, that E1A works together with E4orf3 to perfect the viral transcriptional program and identify a novel mechanism by which the virus can adjust viral gene expression by modifying its genome's nucleosomal organization via cooperation between E1A and E4orf3.


Subject(s)
Adenovirus E1A Proteins/metabolism , Adenovirus E4 Proteins/metabolism , Chromatin/metabolism , A549 Cells , Adenoviridae/genetics , Adenoviridae Infections/virology , Adenovirus E1A Proteins/physiology , Adenovirus E4 Proteins/physiology , Adenoviruses, Human/physiology , Cell Line , Cell Nucleus/virology , Chromatin/virology , Cytoplasm/metabolism , Gene Expression Regulation, Viral/genetics , Gene Expression Regulation, Viral/physiology , Genes, Viral , Humans , Nuclear Proteins/metabolism , Protein Binding , Transcription Factors/metabolism , Virus Replication
7.
J Virol ; 93(4)2019 02 15.
Article in English | MEDLINE | ID: mdl-30518654

ABSTRACT

Simian virus 40 (SV40) exists as chromatin throughout its life cycle and undergoes typical epigenetic regulation mediated by changes in nucleosome location and associated histone modifications. In order to investigate the role of epigenetic regulation during the encapsidation of late-stage minichromosomes into virions, we mapped the locations of nucleosomes containing acetylated or methylated lysines in the histone tails of H3 and H4 present in the chromatin from 48-h-postinfection minichromosomes and disrupted virions. In minichromosomes obtained late in infection, nucleosomes were found carrying various histone modifications primarily in the regulatory region, with a major nucleosome located within the enhancer and other nucleosomes at the early and late transcriptional start sites. The nucleosome found in the enhancer would be expected to repress early transcription by blocking access to part of the SP1 binding sites and the left side of the enhancer in late-stage minichromosomes while also allowing late transcription. In chromatin from virions, the principal nucleosome located in the enhancer was shifted ∼70 bases in the late direction from what was found in minichromosomes, and the level of modified histones was increased throughout the genome. The shifting of the enhancer-associated nucleosome to the late side would effectively serve as a switch to relieve the repression of early transcription found in late minichromosomes while likely also repressing late transcription by blocking access to necessary regulatory sequences. This epigenetic switch appeared to occur during the final stage of virion formation.IMPORTANCE For a virus to complete infection, it must produce a new virus particle in which the genome is able to support a new infection. This is particularly important for viruses like simian virus 40 (SV40), which exist as chromatin throughout their life cycles, since chromatin structure plays a major role in the regulation of the life cycle. In order to determine the role of SV40 chromatin structure late in infection, we mapped the locations of nucleosomes and their histone tail modifications in SV40 minichromosomes and in the SV40 chromatin found in virions using chromatin immunoprecipitation-DNA sequencing (ChIP-Seq). We have identified a novel viral transcriptional control mechanism in which a nucleosome found in the regulatory region of the SV40 minichromosome is directed to slide during the formation of the virus particle, exposing transcription factor binding sites required for early transcription that were previously blocked by the presence of the nucleosome.


Subject(s)
Nucleosomes/genetics , Simian virus 40/genetics , Transcription, Genetic/genetics , Acetylation , Animals , Base Sequence/genetics , Binding Sites , Cell Line , Chlorocebus aethiops , Chromatin/metabolism , Chromatin/virology , Chromatin Assembly and Disassembly , DNA, Viral/genetics , Epigenesis, Genetic/genetics , Histones/metabolism , Nucleosomes/metabolism , Protein Processing, Post-Translational , Simian virus 40/metabolism , Virion/genetics , Virus Replication/genetics
8.
PLoS Pathog ; 14(6): e1007117, 2018 06.
Article in English | MEDLINE | ID: mdl-29906285

ABSTRACT

The murine leukaemia virus (MLV) Gag cleavage product, p12, is essential for both early and late steps in viral replication. The N-terminal domain of p12 binds directly to capsid (CA) and stabilises the mature viral core, whereas defects in the C-terminal domain (CTD) of p12 can be rescued by addition of heterologous chromatin binding sequences (CBSs). We and others hypothesised that p12 tethers the pre-integration complex (PIC) to host chromatin ready for integration. Using confocal microscopy, we have observed for the first time that CA localises to mitotic chromatin in infected cells in a p12-dependent manner. GST-tagged p12 alone, however, did not localise to chromatin and mass-spectrometry analysis of its interactions identified only proteins known to bind the p12 region of Gag. Surprisingly, the ability to interact with chromatin was conferred by a single amino acid change, M63I, in the p12 CTD. Interestingly, GST-p12_M63I showed increased phosphorylation in mitosis relative to interphase, which correlated with an increased interaction with mitotic chromatin. Mass-spectrometry analysis of GST-p12_M63I revealed nucleosomal histones as primary interactants. Direct binding of MLV p12_M63I peptides to histones was confirmed by biolayer-interferometry (BLI) assays using highly-avid recombinant poly-nucleosomal arrays. Excitingly, using this method, we also observed binding between MLV p12_WT and nucleosomes. Nucleosome binding was additionally detected with p12 orthologs from feline and gibbon ape leukemia viruses using both pull-down and BLI assays, indicating that this a common feature of gammaretroviral p12 proteins. Importantly, p12 peptides were able to block the binding of the prototypic foamy virus CBS to nucleosomes and vice versa, implying that their docking sites overlap and suggesting a conserved mode of chromatin tethering for different retroviral genera. We propose that p12 is acting in a similar capacity to CPSF6 in HIV-1 infection by facilitating initial chromatin targeting of CA-containing PICs prior to integration.


Subject(s)
Capsid/metabolism , Chromatin/metabolism , Gene Products, gag/genetics , Mitosis , Nucleosomes/metabolism , Virion/genetics , Virus Integration/physiology , Animals , Chromatin/chemistry , Chromatin/virology , Gene Expression Regulation , Gene Products, gag/chemistry , Gene Products, gag/metabolism , HeLa Cells , Histones/genetics , Histones/metabolism , Humans , Mice , Mutation , Protein Binding , Virion/growth & development , Virion/metabolism , Virus Assembly , Virus Replication
9.
Arch Virol ; 165(2): 321-330, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31828511

ABSTRACT

Persistence of human immunodeficiency virus 1 (HIV-1) latency and residual immune activation remain major barriers to treatment in patients receiving highly active antiretroviral therapy (HAART). In the present study, we investigated the molecular mechanisms of persistent HIV infection and residual immune activation in HAART-treated patients. We showed that the expression level of B-cell CLL/lymphoma 11B (BCL11B) was significantly increased in CD4+T cells from HIV-infected patients undergoing HAART, and this was accompanied by increased expression of BCL11B-associated chromatin modifiers and inflammatory factors in comparison to healthy controls and untreated patients with HIV. In vitro assays showed that BCL11B significantly inhibited HIV-1 long terminal repeat (LTR)-mediated transcription. Knockdown of BCL11B resulted in the activation of HIV latent cells, and dissociation of BCL11B and its related chromatin remodeling factors from the HIV LTR. Our findings suggested that increased expression of BCL11B and its related chromatin modifiers contribute to HIV-1 transcriptional silencing, and alteration of BCL11B levels might lead to abnormal transcription and inflammation.


Subject(s)
Chromatin Assembly and Disassembly/genetics , HIV Infections/genetics , HIV-1/genetics , Repressor Proteins/genetics , Repressor Proteins/immunology , Transcription, Genetic/immunology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/immunology , Adult , Antiretroviral Therapy, Highly Active/methods , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Chromatin/genetics , Chromatin/virology , Chromatin Assembly and Disassembly/drug effects , Chromatin Assembly and Disassembly/immunology , Female , HIV Infections/drug therapy , HIV Infections/immunology , HIV Infections/virology , HIV Long Terminal Repeat/genetics , HIV Long Terminal Repeat/immunology , HIV-1/drug effects , HIV-1/immunology , Humans , Male , Transcription, Genetic/drug effects , Virus Latency/genetics , Virus Latency/immunology
10.
Nature ; 507(7492): 315-22, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24476821

ABSTRACT

Urothelial carcinoma of the bladder is a common malignancy that causes approximately 150,000 deaths per year worldwide. So far, no molecularly targeted agents have been approved for treatment of the disease. As part of The Cancer Genome Atlas project, we report here an integrated analysis of 131 urothelial carcinomas to provide a comprehensive landscape of molecular alterations. There were statistically significant recurrent mutations in 32 genes, including multiple genes involved in cell-cycle regulation, chromatin regulation, and kinase signalling pathways, as well as 9 genes not previously reported as significantly mutated in any cancer. RNA sequencing revealed four expression subtypes, two of which (papillary-like and basal/squamous-like) were also evident in microRNA sequencing and protein data. Whole-genome and RNA sequencing identified recurrent in-frame activating FGFR3-TACC3 fusions and expression or integration of several viruses (including HPV16) that are associated with gene inactivation. Our analyses identified potential therapeutic targets in 69% of the tumours, including 42% with targets in the phosphatidylinositol-3-OH kinase/AKT/mTOR pathway and 45% with targets (including ERBB2) in the RTK/MAPK pathway. Chromatin regulatory genes were more frequently mutated in urothelial carcinoma than in any other common cancer studied so far, indicating the future possibility of targeted therapy for chromatin abnormalities.


Subject(s)
Urinary Bladder Neoplasms/genetics , Cell Cycle/genetics , Chromatin/genetics , Chromatin/virology , Down-Regulation/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Molecular Targeted Therapy , Oxidative Stress/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/genetics , TOR Serine-Threonine Kinases/metabolism , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/virology , Virus Integration/genetics
11.
J Virol ; 92(3)2018 02 01.
Article in English | MEDLINE | ID: mdl-29142137

ABSTRACT

The human genome is structurally organized in three-dimensional space to facilitate functional partitioning of transcription. We learned that the latent episome of the human Epstein-Barr virus (EBV) preferentially associates with gene-poor chromosomes and avoids gene-rich chromosomes. Kaposi's sarcoma-associated herpesvirus behaves similarly, but human papillomavirus does not. Contacts on the EBV side localize to OriP, the latent origin of replication. This genetic element and the EBNA1 protein that binds there are sufficient to reconstitute chromosome association preferences of the entire episome. Contacts on the human side localize to gene-poor and AT-rich regions of chromatin distant from transcription start sites. Upon reactivation from latency, however, the episome moves away from repressive heterochromatin and toward active euchromatin. Our work adds three-dimensional relocalization to the molecular events that occur during reactivation. Involvement of myriad interchromosomal associations also suggests a role for this type of long-range association in gene regulation.IMPORTANCE The human genome is structurally organized in three-dimensional space, and this structure functionally affects transcriptional activity. We set out to investigate whether a double-stranded DNA virus, Epstein-Barr virus (EBV), uses mechanisms similar to those of the human genome to regulate transcription. We found that the EBV genome associates with repressive compartments of the nucleus during latency and with active compartments during reactivation. This study advances our knowledge of the EBV life cycle, adding three-dimensional relocalization as a novel component to the molecular events that occur during reactivation. Furthermore, the data add to our understanding of nuclear compartments, showing that disperse interchromosomal interactions may be important for regulating transcription.


Subject(s)
Chromatin/genetics , Epstein-Barr Virus Nuclear Antigens/metabolism , Herpesvirus 4, Human/physiology , Plasmids/genetics , Cell Line , Cell Nucleus/genetics , Cell Nucleus/virology , Chromatin/virology , Chromosomes, Human/genetics , Chromosomes, Human/virology , Humans , K562 Cells , Replication Origin
12.
J Virol ; 92(18)2018 09 15.
Article in English | MEDLINE | ID: mdl-29997215

ABSTRACT

Adenoviruses are DNA viruses with a lytic infection cycle. Following the fate of incoming as well as recently replicated genomes during infections is a challenge. In this study, we used the ANCHOR3 technology based on a bacterial partitioning system to establish a versatile in vivo imaging system for adenoviral genomes. The system allows the visualization of both individual incoming and newly replicated genomes in real time in living cells. We demonstrate that incoming adenoviral genomes are attached to condensed cellular chromatin during mitosis, facilitating the equal distribution of viral genomes in daughter cells after cell division. We show that the formation of replication centers occurs in conjunction with in vivo genome replication and determine replication rates. Visualization of adenoviral DNA revealed that adenoviruses exhibit two kinetically distinct phases of genome replication. Low-level replication occurred during early replication, while high-level replication was associated with late replication phases. The transition between these phases occurred concomitantly with morphological changes of viral replication compartments and with the appearance of virus-induced postreplication (ViPR) bodies, identified by the nucleolar protein Mybbp1A. Taken together, our real-time genome imaging system revealed hitherto uncharacterized features of adenoviral genomes in vivo The system is able to identify novel spatiotemporal aspects of the adenovirus life cycle and is potentially transferable to other viral systems with a double-stranded DNA phase.IMPORTANCE Viruses must deliver their genomes to host cells to ensure replication and propagation. Characterizing the fate of viral genomes is crucial to understand the viral life cycle and the fate of virus-derived vector tools. Here, we integrated the ANCHOR3 system, an in vivo DNA-tagging technology, into the adenoviral genome for real-time genome detection. ANCHOR3 tagging permitted the in vivo visualization of incoming genomes at the onset of infection and of replicated genomes at late phases of infection. Using this system, we show viral genome attachment to condensed host chromosomes during mitosis, identifying this mechanism as a mode of cell-to-cell transfer. We characterize the spatiotemporal organization of adenovirus replication and identify two kinetically distinct phases of viral genome replication. The ANCHOR3 system is the first technique that allows the continuous visualization of adenoviral genomes during the entire virus life cycle, opening the way for further in-depth study.


Subject(s)
Adenoviridae/physiology , Chromatin/virology , DNA, Viral/metabolism , Virus Replication , Adenoviridae/genetics , Cell Line , Chromatin/genetics , DNA-Binding Proteins , Genome, Viral , HEK293 Cells , Humans , Kinetics , Life Cycle Stages , Nuclear Proteins/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , RNA-Binding Proteins , Staining and Labeling , Transcription Factors , Virus Attachment
13.
J Virol ; 92(6)2018 03 15.
Article in English | MEDLINE | ID: mdl-29321313

ABSTRACT

Hepatitis B virus (HBV) is a global major health problem, with over one million deaths annually caused by chronic liver damage. Understanding host factors that modulate HBV replication may aid the development of anti-HBV therapies. Our recent genome-wide small interfering RNA screen using recombinant HBV demonstrated that TIP60 inhibited HBV infection. Here, we show that TIP60 complex contributes to anti-HBV defense. The TIP60 complex bound to the HBV promoter and suppressed HBV transcription driven by the precore/core promoter. The silencing of EP400, TRRAP, BAF53a, RUVBL1, and RUVBL2, which form the TIP60 complex, also resulted in increased HBV transcription. These results contribute to our enhanced understanding of the molecular mechanism of HBV transcription associated with the chromatin structure of HBV covalently closed circular DNA (cccDNA). Exploiting these intrinsic cellular defenses might help develop new anti-HBV agents.IMPORTANCE Investigating the molecular mechanism of HBV replication is important to understand the persistent nature of HBV infection and to aid the development of new HBV agents, which are currently limited to HBV polymerase inhibitors. Previously, we developed a new reporter HBV. By screening host factors using this recombinant virus, we identified several gene products that regulate HBV infection, including TIP60. Here, we showed that TIP60, a catalytic subunit of the NuA4 complex, inhibited HBV replication. Depletion of TIP60 increased the level of HBV mRNA. Moreover, TIP60 localized in the HBV cccDNA chromatin complex catalyzed the acetylation of histone H4 to recruit Brd4. These results suggest that TIP60, in concert with other cellular factors, plays an important role in the regulation of the HBV chromatin structure by acting as a critical component of the intrinsic antiviral defense, which sheds new light on the regulation of HBV replication.


Subject(s)
Hepatitis B virus/physiology , Hepatitis B/metabolism , Lysine Acetyltransferase 5/metabolism , Multiprotein Complexes/metabolism , Transcription, Genetic , Virus Replication/physiology , Acetylation , Cell Cycle Proteins , Chromatin/genetics , Chromatin/metabolism , Chromatin/pathology , Chromatin/virology , Hep G2 Cells , Hepatitis B/genetics , Hepatitis B/pathology , Histones/genetics , Histones/metabolism , Humans , Lysine Acetyltransferase 5/genetics , Multiprotein Complexes/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
14.
Mol Cell Proteomics ; 16(4 suppl 1): S92-S107, 2017 04.
Article in English | MEDLINE | ID: mdl-28179408

ABSTRACT

Herpes simplex virus (HSV-1) lytic infection results in global changes to the host cell proteome and the proteins associated with host chromatin. We present a system level characterization of proteome dynamics during infection by performing a multi-dimensional analysis during HSV-1 lytic infection of human foreskin fibroblast (HFF) cells. Our study includes identification and quantification of the host and viral proteomes, phosphoproteomes, chromatin bound proteomes and post-translational modifications (PTMs) on cellular histones during infection. We analyzed proteomes across six time points of virus infection (0, 3, 6, 9, 12 and 15 h post-infection) and clustered trends in abundance using fuzzy c-means. Globally, we accurately quantified more than 4000 proteins, 200 differently modified histone peptides and 9000 phosphorylation sites on cellular proteins. In addition, we identified 67 viral proteins and quantified 571 phosphorylation events (465 with high confidence site localization) on viral proteins, which is currently the most comprehensive map of HSV-1 phosphoproteome. We investigated chromatin bound proteins by proteomic analysis of the high-salt chromatin fraction and identified 510 proteins that were significantly different in abundance during infection. We found 53 histone marks significantly regulated during virus infection, including a steady increase of histone H3 acetylation (H3K9ac and H3K14ac). Our data provide a resource of unprecedented depth for human and viral proteome dynamics during infection. Collectively, our results indicate that the proteome composition of the chromatin of HFF cells is highly affected during HSV-1 infection, and that phosphorylation events are abundant on viral proteins. We propose that our epi-proteomics approach will prove to be important in the characterization of other model infectious systems that involve changes to chromatin composition.


Subject(s)
Chromatin/virology , Foreskin/virology , Herpes Simplex/metabolism , Herpesvirus 1, Human/metabolism , Proteomics/methods , Viral Proteins/metabolism , Cells, Cultured , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/virology , Foreskin/cytology , Foreskin/metabolism , Fuzzy Logic , Gene Expression Regulation, Viral , HeLa Cells , Histones/metabolism , Humans , Male , Phosphorylation , Protein Processing, Post-Translational
15.
Nucleic Acids Res ; 45(20): 11673-11683, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-28981850

ABSTRACT

The HIRA histone chaperone complex deposits histone H3.3 into nucleosomes in a DNA replication- and sequence-independent manner. As herpesvirus genomes enter the nucleus as naked DNA, we asked whether the HIRA chaperone complex affects herpesvirus infection. After infection of primary cells with HSV or CMV, or transient transfection with naked plasmid DNA, HIRA re-localizes to PML bodies, sites of cellular anti-viral activity. HIRA co-localizes with viral genomes, binds to incoming viral and plasmid DNAs and deposits histone H3.3 onto these. Anti-viral interferons (IFN) specifically induce HIRA/PML co-localization at PML nuclear bodies and HIRA recruitment to IFN target genes, although HIRA is not required for IFN-inducible expression of these genes. HIRA is, however, required for suppression of viral gene expression, virus replication and lytic infection and restricts murine CMV replication in vivo. We propose that the HIRA chaperone complex represses incoming naked viral DNAs through chromatinization as part of intrinsic cellular immunity.


Subject(s)
Cell Cycle Proteins/metabolism , DNA, Viral/metabolism , Herpesvirus 1, Human/metabolism , Histone Chaperones/metabolism , Histones/metabolism , Transcription Factors/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/immunology , Cell Line , Cell Line, Tumor , Chromatin/genetics , Chromatin/metabolism , Chromatin/virology , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/metabolism , Cytomegalovirus Infections/virology , DNA, Viral/genetics , HEK293 Cells , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/immunology , Histone Chaperones/genetics , Histone Chaperones/immunology , Humans , Inclusion Bodies/immunology , Inclusion Bodies/metabolism , Inclusion Bodies/virology , Mice, Inbred C57BL , Muromegalovirus/genetics , Muromegalovirus/physiology , Promyelocytic Leukemia Protein/metabolism , Protein Binding , Transcription Factors/genetics , Transcription Factors/immunology
16.
Proc Natl Acad Sci U S A ; 113(8): E1054-63, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26858452

ABSTRACT

Integration is vital to retroviral replication and influences the establishment of the latent HIV reservoir. HIV-1 integration favors active genes, which is in part determined by the interaction between integrase and lens epithelium-derived growth factor (LEDGF)/p75. Because gene targeting remains significantly enriched, relative to random in LEDGF/p75 deficient cells, other host factors likely contribute to gene-tropic integration. Nucleoporins 153 and 358, which bind HIV-1 capsid, play comparatively minor roles in integration targeting, but the influence of another capsid binding protein, cleavage and polyadenylation specificity factor 6 (CPSF6), has not been reported. In this study we knocked down or knocked out CPSF6 in parallel or in tandem with LEDGF/p75. CPSF6 knockout changed viral infectivity kinetics, decreased proviral formation, and preferentially decreased integration into transcriptionally active genes, spliced genes, and regions of chromatin enriched in genes and activating histone modifications. LEDGF/p75 depletion by contrast preferentially altered positional integration targeting within gene bodies. Dual factor knockout reduced integration into genes to below the levels observed with either single knockout and revealed that CPSF6 played a more dominant role than LEDGF/p75 in directing integration to euchromatin. CPSF6 complementation rescued HIV-1 integration site distribution in CPSF6 knockout cells, but complementation with a capsid binding mutant of CPSF6 did not. We conclude that integration targeting proceeds via two distinct mechanisms: capsid-CPSF6 binding directs HIV-1 to actively transcribed euchromatin, where the integrase-LEDGF/p75 interaction drives integration into gene bodies.


Subject(s)
Capsid/metabolism , Chromatin/metabolism , HIV-1/physiology , Virus Integration/physiology , mRNA Cleavage and Polyadenylation Factors/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Chromatin/genetics , Chromatin/virology , Gene Knockdown Techniques , HEK293 Cells , Humans , Mutation , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic , mRNA Cleavage and Polyadenylation Factors/genetics
17.
Traffic ; 17(4): 327-40, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26787361

ABSTRACT

Viruses are small infectious agents that replicate in cells of a host organism and that evolved to use cellular machineries for all stages of the viral life cycle. Here, we critically assess current knowledge on a particular mechanism of persisting viruses, namely, how they tether their genomes to host chromatin, and what consequences arise from this process. A group of persisting DNA viruses, i.e. gamma-herpesviruses and papillomaviruses (PV), uses this tethering strategy to maintain their genomes in the nuclei during cell division. Thus, these viruses face the challenge of viral genome loss during mitosis, as they are transported with the host chromosomes to the nascent daughter nuclei. Incidentally, another group of viruses, certain retroviruses and PV, have adopted this tethering strategy to deliver their genomes into the nuclei of dividing cells during cell entry. By exploiting a phase in the cell cycle when the nuclear envelope is disassembled, viruses bypass the need to engage with the nuclear import machinery. Recent reports suggest that tethering may induce severe cellular consequences that involve activation of mitotic checkpoints, causing missegregation of host chromosomes and genomic instability, which may contribute to cancer.


Subject(s)
Chromatin/virology , Genome, Viral , Host-Pathogen Interactions , Animals , Chromatin/genetics , Chromatin/metabolism , Humans , Mitosis
18.
Retrovirology ; 14(1): 54, 2017 Nov 28.
Article in English | MEDLINE | ID: mdl-29179726

ABSTRACT

BACKGROUND: Stable insertion of the retroviral DNA genome into host chromatin requires the functional association between the intasome (integrase·viral DNA complex) and the nucleosome. The data from the literature suggest that direct protein-protein contacts between integrase and histones may be involved in anchoring the intasome to the nucleosome. Since histone tails are candidates for interactions with the incoming intasomes we have investigated whether they could participate in modulating the nucleosomal integration process. RESULTS: We show here that histone tails are required for an optimal association between HIV-1 integrase (IN) and the nucleosome for efficient integration. We also demonstrate direct interactions between IN and the amino-terminal tail of human histone H4 in vitro. Structure/function studies enabled us to identify amino acids in the carboxy-terminal domain of IN that are important for this interaction. Analysis of the nucleosome-binding properties of catalytically active mutated INs confirmed that their ability to engage the nucleosome for integration in vitro was affected. Pseudovirus particles bearing mutations that affect the IN/H4 association also showed impaired replication capacity due to altered integration and re-targeting of their insertion sites toward dynamic regions of the chromatin with lower nucleosome occupancy. CONCLUSIONS: Collectively, our data support a functional association between HIV-1 IN and histone tails that promotes anchoring of the intasome to nucleosomes and optimal integration into chromatin.


Subject(s)
HIV Integrase/metabolism , HIV-1/metabolism , Histones/metabolism , Nucleosomes/metabolism , Virus Integration , Cell Line, Transformed , Chromatin/virology , DNA, Viral/metabolism , HEK293 Cells , HIV-1/genetics , Histones/chemistry , Host-Parasite Interactions/physiology , Humans , Protein Binding
19.
J Virol ; 90(8): 4059-4066, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26842481

ABSTRACT

UNLABELLED: The association of host histones with parvoviral DNA is poorly understood. We analyzed the chromatinization and histone acetylation of canine parvovirus DNA during infection by confocal imaging andin situproximity ligation assay combined with chromatin immunoprecipitation and high-throughput sequencing. We found that during late infection, parvovirus replication bodies were rich in histones bearing modifications characteristic of transcriptionally active chromatin, i.e., histone H3 lysine 27 acetylation (H3K27ac). H3K27ac, in particular, was located in close proximity to the viral DNA-binding protein NS1. Importantly, our results show for the first time that in the chromatinized parvoviral genome, the two viral promoters in particular were rich in H3K27ac. Histone acetyltransferase (HAT) inhibitors efficiently interfered with the expression of viral proteins and infection progress. Altogether, our data suggest that the acetylation of histones on parvoviral DNA is essential for viral gene expression and the completion of the viral life cycle. IMPORTANCE: Viral DNA introduced into cell nuclei is exposed to cellular responses to foreign DNA, including chromatinization and epigenetic silencing, both of which determine the outcome of infection. How the incoming parvovirus resists cellular epigenetic downregulation of its genes is not understood. Here, the critical role of epigenetic modifications in the regulation of parvovirus infection was demonstrated. We showed for the first time that a successful parvovirus infection is characterized by the deposition of nucleosomes with active histone acetylation on the viral promoter areas. The results provide new insights into the regulation of parvoviral gene expression, which is an important aspect of the development of parvovirus-based virotherapy.


Subject(s)
Chromatin/virology , Genome, Viral , Histones/metabolism , Parvoviridae Infections/virology , Parvovirus, Canine/genetics , Promoter Regions, Genetic , Acetylation , Animals , Cats , Cell Line , DNA, Viral/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Viral , Lysine/metabolism , Microscopy, Confocal , Parvovirus, Canine/metabolism , Virus Integration
20.
J Virol ; 90(24): 11197-11207, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27707926

ABSTRACT

Murine leukemia virus (MLV) p12, encoded within Gag, binds the viral preintegration complex (PIC) to the mitotic chromatin. This acts to anchor the viral PIC in the nucleus as the nuclear envelope re-forms postmitosis. Mutations within the p12 C terminus (p12 PM13 to PM15) block early stages in viral replication. Within the p12 PM13 region (p12 60PSPMA65), our studies indicated that chromatin tethering was not detected when the wild-type (WT) p12 protein (M63) was expressed as a green fluorescent protein (GFP) fusion; however, constructs bearing p12-I63 were tethered. N-terminal truncations of the activated p12-I63-GFP indicated that tethering increased further upon deletion of p12 25DLLTEDPPPY34, which includes the late domain required for viral assembly. The p12 PM15 sequence (p12 70RREPP74) is critical for wild-type viral viability; however, virions bearing the PM15 mutation (p12 70AAAAA74) with a second M63I mutant were viable, with a titer 18-fold lower than that of the WT. The p12 M63I mutation amplified chromatin tethering and compensated for the loss of chromatin binding of p12 PM15. Rescue of the p12-M63-PM15 nonviable mutant with prototype foamy virus (PFV) and Kaposi's sarcoma herpesvirus (KSHV) tethering sequences confirmed the function of p1270-74 in chromatin binding. Minimally, full-strength tethering was seen with only p12 61SPIASRLRGRR71 fused to GFP. These results indicate that the p12 C terminus alone is sufficient for chromatin binding and that the presence of the p12 25DLLTEDPPPY34 motif in the N terminus suppresses the ability to tether. IMPORTANCE: This study defines a regulatory mechanism controlling the differential roles of the MLV p12 protein in early and late replication. During viral assembly and egress, the late domain within the p12 N terminus functions to bind host vesicle release factors. During viral entry, the C terminus of p12 is required for tethering to host mitotic chromosomes. Our studies indicate that the p12 domain including the PPPY late sequence temporally represses the p12 chromatin tethering motif. Maximal p12 tethering was identified with only an 11-amino-acid minimal chromatin tethering motif encoded at p1261-71 Within this region, the p12-M63I substitution switches p12 into a tethering-competent state, partially rescuing the p12-PM15 tethering mutant. A model for how this conformational change regulates early versus late functions is presented.


Subject(s)
Chromatin/metabolism , Gene Products, gag/genetics , Host-Pathogen Interactions , Moloney murine leukemia virus/genetics , Virion/genetics , Amino Acid Sequence , Animals , Binding Sites , Chromatin/chemistry , Chromatin/virology , Gene Expression Regulation , Gene Products, gag/chemistry , Gene Products, gag/metabolism , HEK293 Cells , Humans , Mice , Mitosis , Moloney murine leukemia virus/growth & development , Moloney murine leukemia virus/metabolism , Mutation , Protein Binding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , Virion/growth & development , Virion/metabolism , Virus Assembly , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL