Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Front Cell Infect Microbiol ; 10: 577428, 2020.
Article in English | MEDLINE | ID: mdl-33117732

ABSTRACT

Tethering of viral genomes to host chromosomes has been recognized in a variety of DNA and RNA viruses. It can occur during both the productive cycle and latent infection and may impact viral genomes in manifold ways including their protection, localization, transcription, replication, integration, and segregation. Tethering is typically accomplished by dedicated viral proteins that simultaneously associate with both the viral genome and cellular chromatin via nucleic acid, histone and/or non-histone protein interactions. Some of the most prominent tethering proteins have been identified in DNA viruses establishing sustained latent infections, including members of the papillomaviruses and herpesviruses. Herpesvirus particles have linear genomes that circularize in infected cell nuclei and usually persist as extrachromosomal episomes. In several γ-herpesviruses, tethering facilitates the nuclear retention and faithful segregation of viral episomes during cell division, thus contributing to persistence of these viruses in the absence of infectious particle production. However, it has not been studied whether the genomes of human Cytomegalovirus (hCMV), the prototypical ß-herpesvirus, are tethered to host chromosomes. Here we provide evidence by fluorescence in situ hybridization that hCMV genomes associate with the surface of human mitotic chromosomes following infection of both non-permissive myeloid and permissive fibroblast cells. This chromosome association occurs at lower frequency in the absence of the immediate-early 1 (IE1) proteins, which bind to histones and have been implicated in the maintenance of hCMV episomes. Our findings point to a mechanism of hCMV genome maintenance through mitosis and suggest a supporting but non-essential role of IE1 in this process.


Subject(s)
Cytomegalovirus , Immediate-Early Proteins , Chromosomes , Cytomegalovirus/genetics , Humans , Immediate-Early Proteins/genetics , In Situ Hybridization, Fluorescence , Viral Proteins
2.
mBio ; 11(5)2020 09 29.
Article in English | MEDLINE | ID: mdl-32994332

ABSTRACT

The genomes of DNA tumor viruses regain nuclear localization after nuclear envelope breakdown during mitosis through the action of a viral protein with a chromatin-tethering domain (CTD). Here, we report that the human cytomegalovirus (HCMV) genome is maintained during mitosis by the CTD of the viral IE19 protein. Deletion of the IE19 CTD or disruption of the IE19 splice acceptor site reduced viral genome maintenance and progeny virion formation during infection of dividing fibroblasts, both of which were rescued by IE19 ectopic expression. The discovery of a viral genome maintenance factor during productive infection provides new insight into the mode of HCMV infection implicated in birth defects, organ transplant failure, and cancer.IMPORTANCE Human cytomegalovirus (HCMV) is the leading infectious cause of birth defects, represents a serious complication for immunocompromised HIV/AIDS and organ transplant patients, and contributes to both immunosenescence and cardiovascular diseases. HCMV is also implicated in cancers such as glioblastoma multiforme (GBM) and infects ex vivo-cultured GBM tumor cells. In dividing tumor cells, the genomes of DNA tumor viruses regain nuclear localization after nuclear envelope breakdown during mitosis. This mitotic survival is mediated by a viral protein with a chromatin-tethering domain (CTD). Here, we report that the HCMV genome is maintained in dividing fibroblasts by the CTD of the viral IE19 protein. The discovery of a viral genome maintenance factor during productive infection could help explain viral genome dynamics within HCMV-positive tumors as well as during latency.


Subject(s)
Chromatin/metabolism , Cytomegalovirus/genetics , Cytomegalovirus/physiology , Genome, Viral , Immediate-Early Proteins/genetics , Mitosis/genetics , Cell Line , Cells, Cultured , Chromatin/genetics , Fibroblasts/virology , HEK293 Cells , Host-Pathogen Interactions , Humans
3.
PLoS Pathog ; 16(5): e1008537, 2020 05.
Article in English | MEDLINE | ID: mdl-32365141

ABSTRACT

Promyelocytic leukemia (PML) bodies are nuclear organelles implicated in intrinsic and innate antiviral defense. The eponymous PML proteins, central to the self-organization of PML bodies, and other restriction factors found in these organelles are common targets of viral antagonism. The 72-kDa immediate-early protein 1 (IE1) is the principal antagonist of PML bodies encoded by the human cytomegalovirus (hCMV). IE1 is believed to disrupt PML bodies by inhibiting PML SUMOylation, while PML was proposed to act as an E3 ligase for IE1 SUMOylation. PML targeting by IE1 is considered to be crucial for hCMV replication at low multiplicities of infection, in part via counteracting antiviral gene induction linked to the cellular interferon (IFN) response. However, current concepts of IE1-PML interaction are largely derived from mutant IE1 proteins known or predicted to be metabolically unstable and globally misfolded. We performed systematic clustered charge-to-alanine scanning mutagenesis and identified a stable IE1 mutant protein (IE1cc172-176) with wild-type characteristics except for neither interacting with PML proteins nor inhibiting PML SUMOylation. Consequently, IE1cc172-176 does not associate with PML bodies and is selectively impaired for disrupting these organelles. Surprisingly, functional analysis of IE1cc172-176 revealed that the protein is hypermodified by mixed SUMO chains and that IE1 SUMOylation depends on nucleosome rather than PML binding. Furthermore, a mutant hCMV expressing IE1cc172-176 was only slightly attenuated compared to an IE1-null virus even at low multiplicities of infection. Finally, hCMV-induced expression of cytokine and IFN-stimulated genes turned out to be reduced rather than increased in the presence of IE1cc172-176 relative to wild-type IE1. Our findings challenge present views on the relationship of IE1 with PML and the role of PML in hCMV replication. This study also provides initial evidence for the idea that disruption of PML bodies upon viral infection is linked to activation rather than inhibition of innate immunity.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus/physiology , Immediate-Early Proteins , Immunity, Innate , Promyelocytic Leukemia Protein , Virus Replication , Cell Line , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/pathology , Gene Expression Regulation, Viral/immunology , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/immunology , Mutation , Promyelocytic Leukemia Protein/genetics , Promyelocytic Leukemia Protein/immunology , Sumoylation/immunology , Virus Replication/genetics , Virus Replication/immunology
4.
J Immunol ; 205(1): 261-271, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32423918

ABSTRACT

IFNs, produced during viral infections, induce the expression of hundreds of IFN-stimulated genes (ISGs). Some ISGs have specific antiviral activity, whereas others regulate the cellular response. Besides functioning as an antiviral effector, ISG15 is a negative regulator of IFN signaling, and inherited ISG15 deficiency leads to autoinflammatory IFNopathies, in which individuals exhibit elevated ISG expression in the absence of pathogenic infection. We have recapitulated these effects in cultured human A549-ISG15-/- cells and (using A549-UBA7-/- cells) confirmed that posttranslational modification by ISG15 (ISGylation) is not required for regulation of the type I IFN response. ISG15-deficient cells pretreated with IFN-α were resistant to paramyxovirus infection. We also showed that IFN-α treatment of ISG15-deficient cells led to significant inhibition of global protein synthesis, leading us to ask whether resistance was due to the direct antiviral activity of ISGs or whether cells were nonpermissive because of translation defects. We took advantage of the knowledge that IFN-induced protein with tetratricopeptide repeats 1 (IFIT1) is the principal antiviral ISG for parainfluenza virus 5. Knockdown of IFIT1 restored parainfluenza virus 5 infection in IFN-α-pretreated, ISG15-deficient cells, confirming that resistance was due to the direct antiviral activity of the IFN response. However, resistance could be induced if cells were pretreated with IFN-α for longer times, presumably because of inhibition of protein synthesis. These data show that the cause of virus resistance is 2-fold; ISG15 deficiency leads to the early overexpression of specific antiviral ISGs, but the later response is dominated by an unanticipated, ISG15-dependent loss of translational control.


Subject(s)
Cytokines/deficiency , Disease Resistance/genetics , Interferon-alpha/metabolism , Paramyxoviridae Infections/immunology , Signal Transduction/immunology , Ubiquitins/deficiency , A549 Cells , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Chlorocebus aethiops , Gene Knockdown Techniques , Gene Knockout Techniques , Humans , Parainfluenza Virus 2, Human/immunology , Parainfluenza Virus 3, Human/immunology , Parainfluenza Virus 5/immunology , Paramyxoviridae Infections/virology , Protein Processing, Post-Translational/immunology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Signal Transduction/genetics , Ubiquitin-Activating Enzymes/genetics , Vero Cells
5.
mBio ; 11(2)2020 03 17.
Article in English | MEDLINE | ID: mdl-32184235

ABSTRACT

Human adenoviruses (HAdVs) have developed mechanisms to manipulate cellular antiviral measures to ensure proper DNA replication, with detailed processes far from being understood. Host cells repress incoming viral genomes through a network of transcriptional regulators that normally control cellular homeostasis. The nuclear domains involved are promyelocytic leukemia protein nuclear bodies (PML-NBs), interferon-inducible, dot-like nuclear structures and hot spots of SUMO posttranslational modification (PTM). In HAdV-infected cells, such SUMO factories are found in close proximity to newly established viral replication centers (RCs) marked by the adenoviral DNA binding protein (DBP) E2A. Here, we show that E2A is a novel target of host SUMOylation, leading to PTMs supporting E2A function in promoting productive infection. Our data show that SUMOylated E2A interacts with PML. Decreasing SUMO-E2A protein levels by generating HAdV variants mutated in the three main SUMO conjugation motifs (SCMs) led to lower numbers of viral RCs and PML-NBs, and these two structures were no longer next to each other. Our data further indicate that SUMOylated E2A binds the host transcription factor Sp100A, promoting HAdV gene expression, and represents the molecular bridge between PML tracks and adjacent viral RCs. Consequently, E2A SCM mutations repressed late viral gene expression and progeny production. These data highlight a novel mechanism used by the virus to benefit from host antiviral responses by exploiting the cellular SUMO conjugation machinery.IMPORTANCE PML nuclear bodies (PML-NBs) are implicated in general antiviral defense based on recruiting host restriction factors; however, it is not understood so far why viruses would establish viral replication centers (RCs) juxtaposed to such "antiviral" compartments. To understand this enigma, we investigate the cross talk between PML-NB components and viral RCs to find the missing link connecting both compartments to promote efficient viral replication and gene expression. Taken together, the current concept is more intricate than originally believed, since viruses apparently take advantage of several specific PML-NB-associated proteins to promote productive infection. Simultaneously, they efficiently inhibit antiviral measures to maintain the viral infectious program. Our data provide evidence that SUMOylation of the viral RC marker protein E2A represents the basis of this virus-host interface and regulates various downstream events to support HAdV productive infection. These results are the basis of our current attempts to generate and screen for specific E2A SUMOylation inhibitors to constitute novel therapeutic approaches to limit and prevent HAdV-mediated diseases and mortality of immunosuppressed patients.


Subject(s)
Adenovirus E2 Proteins/metabolism , Host-Pathogen Interactions , Promyelocytic Leukemia Protein/metabolism , Sumoylation , Viral Proteins/metabolism , Virus Replication , Adenovirus E2 Proteins/genetics , Adenoviruses, Human/physiology , Cell Line , Humans , Mutation , Promyelocytic Leukemia Protein/genetics , Protein Processing, Post-Translational , Viral Proteins/genetics
6.
Mol Cell ; 71(5): 745-760.e5, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30193098

ABSTRACT

DNA damage can be sensed as a danger-associated molecular pattern by the innate immune system. Here we find that keratinocytes and other human cells mount an innate immune response within hours of etoposide-induced DNA damage, which involves the DNA sensing adaptor STING but is independent of the cytosolic DNA receptor cGAS. This non-canonical activation of STING is mediated by the DNA binding protein IFI16, together with the DNA damage response factors ATM and PARP-1, resulting in the assembly of an alternative STING signaling complex that includes the tumor suppressor p53 and the E3 ubiquitin ligase TRAF6. TRAF6 catalyzes the formation of K63-linked ubiquitin chains on STING, leading to the activation of the transcription factor NF-κB and the induction of an alternative STING-dependent gene expression program. We propose that STING acts as a signaling hub that coordinates a transcriptional response depending on its mode of activation.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , Cell Nucleus/genetics , DNA Damage/genetics , Membrane Proteins/genetics , NF-kappa B/genetics , Nuclear Proteins/genetics , Phosphoproteins/genetics , Signal Transduction/genetics , Cell Line , Cytosol/metabolism , DNA/genetics , HEK293 Cells , Humans , Immunity, Innate/genetics , Keratinocytes/physiology , Poly (ADP-Ribose) Polymerase-1/genetics , Tumor Suppressor Protein p53/genetics , Ubiquitin/genetics , Ubiquitin-Protein Ligases/genetics
7.
J Gen Virol ; 99(9): 1274-1285, 2018 09.
Article in English | MEDLINE | ID: mdl-30045780

ABSTRACT

The relationship between human cytomegalovirus (HCMV) and tumours has been extensively investigated, mainly in glioblastoma multiforme (GBM), a malignant tumour of the central nervous system with low overall survival rates. Several reports have demonstrated the presence of HCMV in GBM, although typically restricted to a low number of cells, and studies have indicated that viral proteins have the ability to dysregulate cellular processes and increase tumour malignancy. Treatment of GBM involves the use of the chemotherapeutic agents temozolomide (TMZ) and carmustine (bis-chloroethylnitrosourea, BCNU), which lead to the attachment of adducts to the DNA backbone, causing errors during replication and consequent cell death. It is known that HCMV infection can modulate DNA repair pathways, but what effects the virus may exhibit during chemotherapy are unknown. Here we approach this question by analysing HCMV infection and viral protein accumulation in GBM cell lines with different genotypes and their response to TMZ and BCNU in the presence of the virus. We demonstrate that A172, TP365MG and U251MG GBM cells are efficiently infected by both low-passage (TB40E) and high-passage (AD169) HCMV strains. However, the GBM cell lines vary widely in their permissiveness to viral gene expression and exhibit very different patterns of immediate early, early and late protein accumulation. HCMV reduces the viability of permissive GBM cells in a multiplicity-dependent manner in both the absence and presence of TMZ or BNCU. In sum, we demonstrate that GBM cell lines are equally susceptible but differentially permissive to infection by both low- and high-passage strains of HCMV. This observation not only indicates that viral replication is largely controlled by cellular factors in this system, but also provides a possible explanation for why viral gene products are only found in a subset of cells in GBM tumours. Furthermore, we conclude that the virus does not confer increased resistance to chemotherapeutic drugs in various GBM cell lines, but instead reduces tumour cell viability. These results highlight that the oncomodulatory potential of HCMV is not limited to cancer-promoting activities, but also includes adverse effects on tumour cell proliferation or survival.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Cytomegalovirus , Glioblastoma/drug therapy , Antineoplastic Agents/administration & dosage , Carmustine/administration & dosage , Carmustine/pharmacology , Cell Line, Tumor , Gene Expression Regulation, Viral , Glioblastoma/virology , Humans , Temozolomide/administration & dosage , Temozolomide/pharmacology , Viral Proteins/genetics , Viral Proteins/metabolism
8.
J Virol ; 92(17)2018 09 01.
Article in English | MEDLINE | ID: mdl-29950413

ABSTRACT

The mechanisms underlying neurodevelopmental damage caused by virus infections remain poorly defined. Congenital human cytomegalovirus (HCMV) infection is the leading cause of fetal brain development disorders. Previous work has linked HCMV infection to perturbations of neural cell fate, including premature differentiation of neural progenitor cells (NPCs). Here, we show that HCMV infection of NPCs results in loss of the SOX2 protein, a key pluripotency-associated transcription factor. SOX2 depletion maps to the HCMV major immediate early (IE) transcription unit and is individually mediated by the IE1 and IE2 proteins. IE1 causes SOX2 downregulation by promoting the nuclear accumulation and inhibiting the phosphorylation of STAT3, a transcriptional activator of SOX2 expression. Deranged signaling resulting in depletion of a critical stem cell protein is an unanticipated mechanism by which the viral major IE proteins may contribute to brain development disorders caused by congenital HCMV infection.IMPORTANCE Human cytomegalovirus (HCMV) infections are a leading cause of brain damage, hearing loss, and other neurological disabilities in children. We report that the HCMV proteins known as IE1 and IE2 target expression of human SOX2, a central pluripotency-associated transcription factor that governs neural progenitor cell (NPC) fate and is required for normal brain development. Both during HCMV infection and when expressed alone, IE1 causes the loss of SOX2 from NPCs. IE1 mediates SOX2 depletion by targeting STAT3, a critical upstream regulator of SOX2 expression. Our findings reveal an unanticipated mechanism by which a common virus may cause damage to the developing nervous system and suggest novel targets for medical intervention.


Subject(s)
Cytomegalovirus/growth & development , Host-Pathogen Interactions , Immediate-Early Proteins/metabolism , Neural Stem Cells/pathology , Neural Stem Cells/virology , SOXB1 Transcription Factors/metabolism , STAT3 Transcription Factor/metabolism , Cells, Cultured , Humans
9.
Antiviral Res ; 150: 79-92, 2018 02.
Article in English | MEDLINE | ID: mdl-29037975

ABSTRACT

Viral interferon (IFN) antagonists are a diverse class of viral proteins that counteract the host IFN response, which is important for controlling viral infections. Viral IFN antagonists are often multifunctional proteins that perform vital roles in virus replication beyond IFN antagonism. The critical importance of viral IFN antagonists is highlighted by the fact that almost all viruses encode one of these proteins. Inhibition of viral IFN antagonists has the potential to exert pleiotropic antiviral effects and thus this important protein class represents a diverse plethora of novel therapeutic targets. To exploit this, we have successfully developed and executed a novel modular cell-based platform that facilitates the safe and rapid screening for inhibitors of a viral IFN antagonist of choice. The platform is based on two reporter cell-lines that provide a simple method to detect activation of IFN induction or signaling via an eGFP gene placed under the control of the IFNß or an ISRE-containing promoter, respectively. Expression of a target IFN antagonist in the appropriate reporter cell-line will block the IFN response and hence eGFP expression. We hypothesized that addition of a compound that inhibits IFN antagonist function will release the block imposed on the IFN response and hence restore eGFP expression, providing a measurable parameter for high throughput screening (HTS). We demonstrate assay proof-of-concept by (i) exploiting hepatitis C virus (HCV) protease inhibitors to inhibit NS3-4A's capacity to block IFN induction and (ii) successfully executing two HTS targeting viral IFN antagonists that block IFN signaling; NS2 and IE1 from human respiratory syncytial virus (RSV) and cytomegalovirus (CMV) respectively, two clinically important viruses for which vaccine development has thus far been unsuccessful and new antivirals are required. Both screens performed robustly and Z' Factor scores of >0.6 were achieved. We identified (i) four hit compounds that specifically inhibit RSV NS2's ability to block IFN signaling by mediating STAT2 degradation and exhibit modest antiviral activity and (ii) two hit compounds that interfere with IE1 transcription and significantly impair CMV replication. Overall, we demonstrate assay proof-of-concept as we target viral IFN antagonists from unrelated viruses and demonstrate its suitability for HTS.


Subject(s)
Antiviral Agents/pharmacology , Drug Discovery , Drug Evaluation, Preclinical , High-Throughput Screening Assays , Interferons/antagonists & inhibitors , Interferons/pharmacology , Viral Proteins/metabolism , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Discovery/methods , Drug Evaluation, Preclinical/methods , Gene Expression Regulation/drug effects , Gene Expression Regulation, Viral/drug effects , Genes, Reporter , Humans , Protein Binding , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Virus, Human/physiology , Signal Transduction , Virus Replication/drug effects
10.
J Virol ; 90(20): 9543-55, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27512077

ABSTRACT

UNLABELLED: Kaposi's sarcoma-associated herpesvirus (KSHV) is the causative agent of commonly fatal malignancies of immunocompromised individuals, including primary effusion lymphoma (PEL) and Kaposi's sarcoma (KS). A hallmark of all herpesviruses is their biphasic life cycle-viral latency and the productive lytic cycle-and it is well established that reactivation of the KSHV lytic cycle is associated with KS pathogenesis. Therefore, a thorough appreciation of the mechanisms that govern reactivation is required to better understand disease progression. The viral protein replication and transcription activator (RTA) is the KSHV lytic switch protein due to its ability to drive the expression of various lytic genes, leading to reactivation of the entire lytic cycle. While the mechanisms for activating lytic gene expression have received much attention, how RTA impacts cellular function is less well understood. To address this, we developed a cell line with doxycycline-inducible RTA expression and applied stable isotope labeling of amino acids in cell culture (SILAC)-based quantitative proteomics. Using this methodology, we have identified a novel cellular protein (AT-rich interacting domain containing 3B [ARID3B]) whose expression was enhanced by RTA and that relocalized to replication compartments upon lytic reactivation. We also show that small interfering RNA (siRNA) knockdown or overexpression of ARID3B led to an enhancement or inhibition of lytic reactivation, respectively. Furthermore, DNA affinity and chromatin immunoprecipitation assays demonstrated that ARID3B specifically interacts with A/T-rich elements in the KSHV origin of lytic replication (oriLyt), and this was dependent on lytic cycle reactivation. Therefore, we have identified a novel cellular protein whose expression is enhanced by KSHV RTA with the ability to inhibit KSHV reactivation. IMPORTANCE: Kaposi's sarcoma-associated herpesvirus (KSHV) is the causative agent of fatal malignancies of immunocompromised individuals, including Kaposi's sarcoma (KS). Herpesviruses are able to establish a latent infection, in which they escape immune detection by restricting viral gene expression. Importantly, however, reactivation of productive viral replication (the lytic cycle) is necessary for the pathogenesis of KS. Therefore, it is important that we comprehensively understand the mechanisms that govern lytic reactivation, to better understand disease progression. In this study, we have identified a novel cellular protein (AT-rich interacting domain protein 3B [ARID3B]) that we show is able to temper lytic reactivation. We showed that the master lytic switch protein, RTA, enhanced ARID3B levels, which then interacted with viral DNA in a lytic cycle-dependent manner. Therefore, we have added a new factor to the list of cellular proteins that regulate the KSHV lytic cycle, which has implications for our understanding of KSHV biology.


Subject(s)
DNA-Binding Proteins/genetics , Herpesvirus 8, Human/genetics , Sarcoma, Kaposi/virology , Viral Proteins/genetics , Cell Line, Tumor , Chromatin Immunoprecipitation/methods , DNA Replication/genetics , DNA, Viral/genetics , Gene Expression Regulation, Viral/genetics , Humans , Immediate-Early Proteins/genetics , Lymphoma, Primary Effusion/genetics , Lymphoma, Primary Effusion/virology , RNA, Small Interfering/genetics , Trans-Activators/genetics , Virus Activation/genetics , Virus Latency/genetics , Virus Replication/genetics
11.
PLoS Pathog ; 12(7): e1005748, 2016 07.
Article in English | MEDLINE | ID: mdl-27387064

ABSTRACT

The human cytomegalovirus (hCMV) major immediate-early 1 protein (IE1) is best known for activating transcription to facilitate viral replication. Here we present transcriptome data indicating that IE1 is as significant a repressor as it is an activator of host gene expression. Human cells induced to express IE1 exhibit global repression of IL6- and oncostatin M-responsive STAT3 target genes. This repression is followed by STAT1 phosphorylation and activation of STAT1 target genes normally induced by IFNγ. The observed repression and subsequent activation are both mediated through the same region (amino acids 410 to 445) in the C-terminal domain of IE1, and this region serves as a binding site for STAT3. Depletion of STAT3 phenocopies the STAT1-dependent IFNγ-like response to IE1. In contrast, depletion of the IL6 receptor (IL6ST) or the STAT kinase JAK1 prevents this response. Accordingly, treatment with IL6 leads to prolonged STAT1 instead of STAT3 activation in wild-type IE1 expressing cells, but not in cells expressing a mutant protein (IE1dl410-420) deficient for STAT3 binding. A very similar STAT1-directed response to IL6 is also present in cells infected with a wild-type or revertant hCMV, but not an IE1dl410-420 mutant virus, and this response results in restricted viral replication. We conclude that IE1 is sufficient and necessary to rewire upstream IL6-type to downstream IFNγ-like signaling, two pathways linked to opposing actions, resulting in repressed STAT3- and activated STAT1-responsive genes. These findings relate transcriptional repressor and activator functions of IE1 and suggest unexpected outcomes relevant to viral pathogenesis in response to cytokines or growth factors that signal through the IL6ST-JAK1-STAT3 axis in hCMV-infected cells. Our results also reveal that IE1, a protein considered to be a key activator of the hCMV productive cycle, has an unanticipated role in tempering viral replication.


Subject(s)
Cytomegalovirus Infections/metabolism , Gene Expression Regulation/physiology , Immediate-Early Proteins/metabolism , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Cell Line , Cytomegalovirus/immunology , Cytomegalovirus/metabolism , Cytomegalovirus/pathogenicity , Cytomegalovirus Infections/immunology , Fibroblasts/virology , Humans , Immunoblotting , Immunoprecipitation , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-6/biosynthesis , Interleukin-6/immunology , Mutagenesis, Site-Directed , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Virus Replication/physiology
12.
Med Microbiol Immunol ; 204(3): 285-93, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25732096

ABSTRACT

The morphogenesis of human cytomegalovirus (HCMV) particles is incompletely understood. Analysis of the protein composition of HCMV virions and subviral dense bodies (DBs) by mass spectrometry provides valuable information to increase our knowledge about viral morphogenesis. Here we addressed the viral proteome of virions and DBs from two fibroblast-passaged isolates and the widely used endotheliotropic TB4-BAC40 strain of HCMV. The results show a striking concordance of the particle proteomes of different strains. One surprising finding was that only low levels of gpUL128-131A were found in TB40-BAC4 virions. These three proteins, together with gH and gL, form a protein complex that is critical for the endothelial cell tropism of that strain. This indicates that either few molecules of that complex per virion or a small fraction of pentamer-positive virions suffice to retain the tropism. Furthermore, using a pp65-deficient variant of TB40-BAC4, we confirm our previous finding that the major tegument protein serves as a scaffold to support the upload of a fraction of the outer tegument proteins into particles. The results demonstrate that HCMV particle morphogenesis is an orchestrated process that leads to the formation of particles with a largely strain-independent protein composition.


Subject(s)
Cytomegalovirus/classification , Cytomegalovirus/physiology , Proteome , Proteomics , Viral Proteins/metabolism , Virion , Cell Line , Cells, Cultured , Cytomegalovirus/isolation & purification , Endothelial Cells/virology , Humans , Mass Spectrometry , Open Reading Frames , Phosphoproteins/metabolism , Proteomics/methods , Viral Matrix Proteins/metabolism , Viral Tropism , Virus Assembly
13.
J Virol ; 88(2): 1228-48, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24227840

ABSTRACT

The 72-kDa immediate early 1 (IE1) protein encoded by human cytomegalovirus (hCMV) is a nuclearly localized promiscuous regulator of viral and cellular transcription. IE1 has long been known to associate with host mitotic chromatin, yet the mechanisms underlying this interaction have not been specified. In this study, we identify the cellular chromosome receptor for IE1. We demonstrate that the viral protein targets human nucleosomes by directly binding to core histones in a nucleic acid-independent manner. IE1 exhibits two separable histone-interacting regions with differential binding specificities for H2A-H2B and H3-H4. The H2A-H2B binding region was mapped to an evolutionarily conserved 10-amino-acid motif within the chromatin-tethering domain (CTD) of IE1. Results from experimental approaches combined with molecular modeling indicate that the IE1 CTD adopts a ß-hairpin structure, docking with the acidic pocket formed by H2A-H2B on the nucleosome surface. IE1 binds to the acidic pocket in a way similar to that of the latency-associated nuclear antigen (LANA) of the Kaposi's sarcoma-associated herpesvirus. Consequently, the IE1 and LANA CTDs compete for binding to nucleosome cores and chromatin. Our work elucidates in detail how a key viral regulator is anchored to human chromosomes and identifies the nucleosomal acidic pocket as a joint target of proteins from distantly related viruses. Based on the striking similarities between the IE1 and LANA CTDs and the fact that nucleosome targeting by IE1 is dispensable for productive replication even in "clinical" strains of hCMV, we speculate that the two viral proteins may serve analogous functions during latency of their respective viruses.


Subject(s)
Chromosomes, Human/virology , Cytomegalovirus Infections/virology , Cytomegalovirus/metabolism , Immediate-Early Proteins/metabolism , Nucleosomes/metabolism , Amino Acid Sequence , Binding Sites , Chromosomes, Human/genetics , Chromosomes, Human/metabolism , Cytomegalovirus/genetics , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/metabolism , Histones/genetics , Histones/metabolism , Humans , Immediate-Early Proteins/chemistry , Immediate-Early Proteins/genetics , Models, Molecular , Molecular Sequence Data , Nucleosomes/chemistry , Nucleosomes/virology , Protein Binding , Protein Structure, Tertiary
14.
J Virol ; 87(19): 10763-76, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23903834

ABSTRACT

In the canonical STAT3 signaling pathway, binding of agonist to receptors activates Janus kinases that phosphorylate cytoplasmic STAT3 at tyrosine 705 (Y705). Phosphorylated STAT3 dimers accumulate in the nucleus and drive the expression of genes involved in inflammation, angiogenesis, invasion, and proliferation. Here, we demonstrate that human cytomegalovirus (HCMV) infection rapidly promotes nuclear localization of STAT3 in the absence of robust phosphorylation at Y705. Furthermore, infection disrupts interleukin-6 (IL-6)-induced phosphorylation of STAT3 and expression of a subset of IL-6-induced STAT3-regulated genes, including SOCS3. We show that the HCMV 72-kDa immediate-early 1 (IE1) protein associates with STAT3 and is necessary to localize STAT3 to the nucleus during infection. Furthermore, expression of IE1 is sufficient to disrupt IL-6-induced phosphorylation of STAT3, binding of STAT3 to the SOCS3 promoter, and SOCS3 gene expression. Finally, inhibition of STAT3 nuclear localization or STAT3 expression during infection is linked to diminished HCMV genome replication. Viral gene expression is also disrupted, with the greatest impact seen following viral DNA synthesis. Our study identifies IE1 as a new regulator of STAT3 intracellular localization and IL-6 signaling and points to an unanticipated role of STAT3 in HCMV infection.


Subject(s)
Cell Nucleus/metabolism , Cytomegalovirus Infections/virology , Cytomegalovirus/physiology , Immediate-Early Proteins/metabolism , Interleukin-6/metabolism , STAT3 Transcription Factor/metabolism , Animals , Astrocytoma/metabolism , Astrocytoma/pathology , Astrocytoma/virology , Blotting, Western , Cell Nucleus/genetics , Cells, Cultured , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/virology , Fluorescent Antibody Technique , Humans , Immediate-Early Proteins/genetics , Interleukin-6/genetics , Mice , Phosphorylation , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/genetics , Signal Transduction , Trans-Activators , Virus Replication
15.
Proc Natl Acad Sci U S A ; 110(32): 13126-31, 2013 Aug 06.
Article in English | MEDLINE | ID: mdl-23878222

ABSTRACT

Human CMV (hCMV) establishes lifelong infections in most of us, causing developmental defects in human embryos and life-threatening disease in immunocompromised individuals. During productive infection, the viral >230,000-bp dsDNA genome is expressed widely and in a temporal cascade. The hCMV genome does not carry histones when encapsidated but has been proposed to form nucleosomes after release into the host cell nucleus. Here, we present hCMV genome-wide nucleosome occupancy and nascent transcript maps during infection of permissive human primary cells. We show that nucleosomes occupy nuclear viral DNA in a nonrandom and highly predictable fashion. At early times of infection, nucleosomes associate with the hCMV genome largely according to their intrinsic DNA sequence preferences, indicating that initial nucleosome formation is genetically encoded in the virus. However, as infection proceeds to the late phase, nucleosomes redistribute extensively to establish patterns mostly determined by nongenetic factors. We propose that these factors include key regulators of viral gene expression encoded at the hCMV major immediate-early (IE) locus. Indeed, mutant virus genomes deficient for IE1 expression exhibit globally increased nucleosome loads and reduced nucleosome dynamics compared with WT genomes. The temporal nucleosome occupancy differences between IE1-deficient and WT viruses correlate inversely with changes in the pattern of viral nascent and total transcript accumulation. These results provide a framework of spatial and temporal nucleosome organization across the genome of a major human pathogen and suggest that an hCMV major IE protein governs overall viral chromatin structure and function.


Subject(s)
Chromatin/genetics , Cytomegalovirus/genetics , Genome, Viral/genetics , Immediate-Early Proteins/genetics , Nucleosomes/genetics , Cell Line , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Nucleus/virology , Chromatin/metabolism , Cytomegalovirus/metabolism , Cytomegalovirus/physiology , DNA, Viral/genetics , DNA, Viral/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/virology , Host-Pathogen Interactions , Humans , Immediate-Early Proteins/metabolism , Immunoblotting , Mutation , Nucleosomes/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
16.
J Virol ; 86(18): 9817-27, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22761369

ABSTRACT

In the nuclei of permissive cells, human cytomegalovirus genomes form nucleosomal structures initially resembling heterochromatin but gradually switching to a euchromatin-like state. This switch is characterized by a decrease in histone H3 K9 methylation and a marked increase in H3 tail acetylation and H3 K4 methylation across the viral genome. We used ganciclovir and a mutant virus encoding a reversibly destabilized DNA polymerase to examine the impact of DNA replication on histone modification dynamics at the viral chromatin. The changes in H3 tail acetylation and H3 K9 methylation proceeded in a DNA replication-independent fashion. In contrast, the increase in H3 K4 methylation proved to depend widely on viral DNA synthesis. Consistently, labeling of nascent DNA using "click chemistry" revealed preferential incorporation of methylated H3 K4 into viral (but not cellular) chromatin during or following DNA replication. This study demonstrates largely selective epigenetic tagging of postreplicative human cytomegalovirus chromatin.


Subject(s)
Chromatin/genetics , Chromatin/metabolism , Cytomegalovirus/genetics , Cytomegalovirus/physiology , Histones/metabolism , Base Sequence , Cell Line , Chromatin/chemistry , Cytomegalovirus/pathogenicity , DNA Replication , DNA, Viral/biosynthesis , DNA, Viral/genetics , Epigenesis, Genetic , Euchromatin/chemistry , Euchromatin/genetics , Euchromatin/metabolism , Genome, Viral , Heterochromatin/chemistry , Heterochromatin/genetics , Heterochromatin/metabolism , Histones/chemistry , Humans , Lysine/chemistry , Methylation , Virus Replication/genetics , Virus Replication/physiology
17.
PLoS Pathog ; 7(4): e1002016, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21533215

ABSTRACT

Human cytomegalovirus (hCMV) is a highly prevalent pathogen that, upon primary infection, establishes life-long persistence in all infected individuals. Acute hCMV infections cause a variety of diseases in humans with developmental or acquired immune deficits. In addition, persistent hCMV infection may contribute to various chronic disease conditions even in immunologically normal people. The pathogenesis of hCMV disease has been frequently linked to inflammatory host immune responses triggered by virus-infected cells. Moreover, hCMV infection activates numerous host genes many of which encode pro-inflammatory proteins. However, little is known about the relative contributions of individual viral gene products to these changes in cellular transcription. We systematically analyzed the effects of the hCMV 72-kDa immediate-early 1 (IE1) protein, a major transcriptional activator and antagonist of type I interferon (IFN) signaling, on the human transcriptome. Following expression under conditions closely mimicking the situation during productive infection, IE1 elicits a global type II IFN-like host cell response. This response is dominated by the selective up-regulation of immune stimulatory genes normally controlled by IFN-γ and includes the synthesis and secretion of pro-inflammatory chemokines. IE1-mediated induction of IFN-stimulated genes strictly depends on tyrosine-phosphorylated signal transducer and activator of transcription 1 (STAT1) and correlates with the nuclear accumulation and sequence-specific binding of STAT1 to IFN-γ-responsive promoters. However, neither synthesis nor secretion of IFN-γ or other IFNs seems to be required for the IE1-dependent effects on cellular gene expression. Our results demonstrate that a single hCMV protein can trigger a pro-inflammatory host transcriptional response via an unexpected STAT1-dependent but IFN-independent mechanism and identify IE1 as a candidate determinant of hCMV pathogenicity.


Subject(s)
Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Immediate-Early Proteins/immunology , Interferon-gamma/immunology , STAT1 Transcription Factor/immunology , Transcription, Genetic/immunology , Acute Disease , Cell Line , Cytomegalovirus/genetics , Cytomegalovirus Infections/genetics , Humans , Immediate-Early Proteins/genetics , Inflammation/genetics , Inflammation/immunology , Interferon-gamma/genetics , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/immunology , STAT1 Transcription Factor/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Transcription, Genetic/genetics
18.
Rev Med Virol ; 21(3): 154-80, 2011 May.
Article in English | MEDLINE | ID: mdl-21538665

ABSTRACT

Herpesvirus infections of humans can cause a broad variety of symptoms ranging from mild afflictions to life-threatening disease. During infection, the large double-stranded DNA genomes of all herpesviruses are transcribed, replicated and encapsidated in the host cell nucleus, where DNA is typically structured and manoeuvred through nucleosomes. Nucleosomes individually assemble DNA around core histone octamers to form 'beads-on-a-string' chromatin fibres. Herpesviruses have responded to the advantages and challenges of chromatin formation in biologically unique ways. Although herpesvirus DNA is devoid of histones within nucleocapsids, nuclear viral genomes most likely form irregularly arranged or unstable nucleosomes during productive infection, and regular nucleosomal arrays resembling host cell chromatin in latently infected cells. Besides variations in nucleosome density, herpesvirus chromatin 'bead strings' undergo dynamic changes in histone composition and modification during the different stages of productive replication, latent infection and reactivation from latency, raising the likely possibility that epigenetic processes may dictate, at least in part, the outcome of infection and ensuing pathogenesis. Here, we summarise and discuss several new and important aspects regarding the nucleosome-based mechanisms that regulate herpesvirus chromatin structure and function in infected cells. Special emphasis is given to processes of histone deposition, histone variant exchange and covalent histone modification in relation to the transcription from the viral genome during productive and latent infections by human cytomegalovirus and herpes simplex virus type 1. We also present an overview on emerging histone-directed antiviral strategies that may be developed into 'epigenetic therapies' to improve current prevention and treatment options targeting herpesvirus infection and disease.


Subject(s)
Chromatin/metabolism , Cytomegalovirus/physiology , DNA, Viral/metabolism , Herpesvirus 1, Human/physiology , Histones/metabolism , Nucleosomes/metabolism , Virus Replication , Cytomegalovirus/growth & development , Herpesvirus 1, Human/growth & development , Humans , Models, Biological , Virus Activation , Virus Latency
19.
Cell Mol Life Sci ; 68(6): 1079-90, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20803231

ABSTRACT

Human cytomegalovirus (HCMV) is a major pathogen in immunocompromised individuals. Here, non-toxic concentrations of the anti-cancer kinase inhibitor sorafenib were shown to inhibit replication of different HCMV strains (including a ganciclovir-resistant strain) in different cell types. In contrast to established anti-HCMV drugs, sorafenib inhibited HCMV major immediate early promoter activity and HCMV immediate early antigen (IEA) expression. Sorafenib is known to inhibit Raf. Comparison of sorafenib with the MEK inhibitor U0126 suggested that sorafenib inhibits HCMV IEA expression through inhibition of Raf but independently of signaling through the Raf downstream kinase MEK 1/2. In concordance, siRNA-mediated depletion of Raf but not of MEK-reduced IEA expression. In conclusion, sorafenib diminished HCMV replication in clinically relevant concentrations and inhibited HCMV IEA expression, a pathophysiologically relevant event that is not affected by established anti-HCMV drugs. Moreover, we demonstrated for the first time that Raf activation is involved in HCMV IEA expression.


Subject(s)
Benzenesulfonates/pharmacology , Cytomegalovirus/drug effects , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Virus Replication/drug effects , DNA Primers/genetics , Genes, Immediate-Early/genetics , Humans , Immunoblotting , Luciferases , Niacinamide/analogs & derivatives , Phenylurea Compounds , Promoter Regions, Genetic/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Sorafenib , raf Kinases/antagonists & inhibitors
20.
Rev Med Virol ; 20(1): 34-50, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19890944

ABSTRACT

The double-stranded DNA genomes of herpesviruses exist in at least three alternative global chromatin states characterised by distinct nucleosome content. When encapsidated in virus particles, the viral DNA is devoid of any nucleosomes. In contrast, within latently infected nuclei herpesvirus genomes are believed to form regular nucleosomal structures resembling cellular chromatin. Finally, during productive infection nuclear viral DNA appears to adopt a state of intermediate chromatin formation with irregularly spaced nucleosomes. Nucleosome occupancy coupled with posttranslational histone modifications and other epigenetic marks may contribute significantly to the extent and timing of transcription from the viral genome and, consequently, to the outcome of infection. Recent research has provided first insights into the viral and cellular mechanisms that either maintain individual herpesvirus chromatin states or mediate transition between them. Here, we summarise and discuss both early work and new developments pointing towards common principles pertinent to the dynamic structure and epigenetic regulation of herpesvirus chromatin. Special emphasis is given to the emerging similarities in nucleosome assembly and disassembly processes on herpes simplex virus type 1 and human cytomegalovirus genomes over the course of the viral productive replication cycle and during the switch between latent and lytic infectious stages.


Subject(s)
Chromatin Assembly and Disassembly , Cytomegalovirus/physiology , Genome, Viral , Herpesviridae Infections/virology , Herpesvirus 1, Human/physiology , DNA, Viral/genetics , Herpesviridae Infections/genetics , Humans , Virus Latency/genetics , Virus Replication/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...