Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS Pathog ; 14(9): e1007313, 2018 09.
Article in English | MEDLINE | ID: mdl-30235352

ABSTRACT

Herpes simplex virus 1 (HSV-1) latency establishment is tightly controlled by promyelocytic leukemia (PML) nuclear bodies (NBs) (or ND10), although their exact contribution is still elusive. A hallmark of HSV-1 latency is the interaction between latent viral genomes and PML NBs, leading to the formation of viral DNA-containing PML NBs (vDCP NBs), and the complete silencing of HSV-1. Using a replication-defective HSV-1-infected human primary fibroblast model reproducing the formation of vDCP NBs, combined with an immuno-FISH approach developed to detect latent/quiescent HSV-1, we show that vDCP NBs contain both histone H3.3 and its chaperone complexes, i.e., DAXX/ATRX and HIRA complex (HIRA, UBN1, CABIN1, and ASF1a). HIRA also co-localizes with vDCP NBs present in trigeminal ganglia (TG) neurons from HSV-1-infected wild type mice. ChIP and Re-ChIP show that vDCP NBs-associated latent/quiescent viral genomes are chromatinized almost exclusively with H3.3 modified on its lysine (K) 9 by trimethylation, consistent with an interaction of the H3.3 chaperones with multiple viral loci and with the transcriptional silencing of HSV-1. Only simultaneous inactivation of both H3.3 chaperone complexes has a significant impact on the deposition of H3.3 on viral genomes, suggesting a compensation mechanism. In contrast, the sole depletion of PML significantly impacts the chromatinization of the latent/quiescent viral genomes with H3.3 without any overall replacement with H3.1. vDCP NBs-associated HSV-1 genomes are not definitively silenced since the destabilization of vDCP NBs by ICP0, which is essential for HSV-1 reactivation in vivo, allows the recovery of a transcriptional lytic program and the replication of viral genomes. Consequently, the present study demonstrates a specific chromatin regulation of vDCP NBs-associated latent/quiescent HSV-1 through an H3.3-dependent HSV-1 chromatinization involving the two H3.3 chaperones DAXX/ATRX and HIRA complexes. Additionally, the study reveals that PML NBs are major actors in latent/quiescent HSV-1 H3.3 chromatinization through a PML NB/histone H3.3/H3.3 chaperone axis.


Subject(s)
Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/metabolism , Promyelocytic Leukemia Protein/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins/metabolism , Cell Nucleus Structures/metabolism , Cell Nucleus Structures/virology , Cells, Cultured , Co-Repressor Proteins , DNA, Viral/genetics , DNA, Viral/metabolism , Female , Genome, Viral , Herpesvirus 1, Human/pathogenicity , Histone Chaperones/metabolism , Histones/metabolism , Host-Pathogen Interactions , Humans , Mice , Mice, Inbred BALB C , Molecular Chaperones , Nuclear Proteins/metabolism , Promyelocytic Leukemia Protein/deficiency , Promyelocytic Leukemia Protein/genetics , Transcription Factors/metabolism , Virus Latency/genetics , Virus Latency/physiology , X-linked Nuclear Protein/metabolism
2.
Proc Natl Acad Sci U S A ; 114(19): E3823-E3829, 2017 05 09.
Article in English | MEDLINE | ID: mdl-28439026

ABSTRACT

Nuclear domain 10 (ND10) bodies are small (0.1-1 µM) nuclear structures containing both constant [e.g., promyelocytic leukemia protein (PML), SP100, death domain-associated protein (Daxx)] and variable proteins, depending on the function of the cells or the stress to which they are exposed. In herpes simplex virus (HSV)-infected cells, ND10 bodies assemble at the sites of DNA entering the nucleus after infection. In sequence, the ND10 bodies become viral replication compartments, and ICP0, a viral E3 ligase, degrades both PML and SP100. The amounts of PML and SP100 and the number of ND10 structures increase in cells exposed to IFN-ß. Earlier studies have shown that PML has three key functions. Thus, (i) the interaction of PML with viral components facilitates the initiation of replication compartments, (ii) viral replication is significantly less affected by IFN-ß in PML-/- cells than in parental PML+/+ cells, and (iii) viral yields are significantly lower in PML-/- cells exposed to low ratios of virus per cell compared with parental PML+/+ cells. This report focuses on the function of SP100. In contrast to PML-/- cells, SP100-/- cells retain the sensitivity of parental SP100+/+ cells to IFN-ß and support replication of the ΔICP0 virus. At low multiplicities of infection, wild-type virus yields are higher in SP100-/- cells than in parental HEp-2 cells. In addition, the number of viral replication compartments is significantly higher in SP100-/- cells than in parental SP100+/+ cells or in PML-/- cells.


Subject(s)
Antigens, Nuclear/metabolism , Autoantigens/metabolism , Cell Nucleus Structures/metabolism , Promyelocytic Leukemia Protein/metabolism , Simplexvirus/physiology , Virus Assembly/physiology , Animals , Antigens, Nuclear/genetics , Autoantigens/genetics , Cell Nucleus Structures/genetics , Cell Nucleus Structures/virology , Chlorocebus aethiops , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promyelocytic Leukemia Protein/genetics , Vero Cells
3.
Virology ; 366(2): 316-29, 2007 Sep 30.
Article in English | MEDLINE | ID: mdl-17543368

ABSTRACT

Promyelocytic leukemia oncogenic domains (PODs), also called nuclear domain 10 (ND10), are subnuclear structures that have been implicated in a variety of cellular processes as well as the life cycle of DNA viruses including papillomaviruses. In order to investigate the interplay between papillomaviruses and PODs, we analyzed the status of PODs in organotypic raft cultures of human keratinocytes harboring HPV genome that support the differentiation-dependent HPV life cycle. The number of PODs per nucleus was increased in the presence of HPV genomes selectively within the poorly differentiated layers but was absent in the terminally differentiated layers of the stratified epithelium. This increase in PODs was correlated with an increase in abundance of post-translationally modified PML protein. Neither the E2-dependent transcription nor viral DNA replication was reliant upon the presence of PML. Implications of these findings in terms of HPV's interaction with its host are discussed.


Subject(s)
Cell Nucleus Structures/virology , Keratinocytes/virology , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Papillomaviridae/physiology , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Cells, Cultured , Humans , Neoplasm Proteins/chemistry , Nuclear Proteins/chemistry , Promyelocytic Leukemia Protein , Protein Structure, Tertiary , Transcription Factors/chemistry , Tumor Suppressor Proteins/chemistry , Viral Proteins/biosynthesis , Virus Replication/physiology
4.
J Virol ; 81(9): 4744-52, 2007 May.
Article in English | MEDLINE | ID: mdl-17301128

ABSTRACT

The PML oncogenic domain (POD/ND10/PML body) is a common target of DNA viruses, which replicate their genomes in proximity to this nuclear structure. The adenovirus early protein E4 ORF3 is both necessary and sufficient to rearrange PODs from punctate bodies into track-like structures. Although multiple hypotheses exist, the precise reason for this activity has not yet been elucidated. PML, the protein responsible for nucleating PODs, is an interferon (IFN)-stimulated gene, implicating the participation of this nuclear body in an innate antiviral response. Here, we demonstrate that E4 ORF3 is critical to the replicative success of adenovirus during the IFN-induced antiviral state. When cells are pretreated with either IFN-alpha or IFN-gamma, a mutant virus that does not express E4 ORF3 is severely compromised for replication. This result suggests the functional significance of ORF3 track formation is the inhibition of a POD-mediated, antiviral mechanism. Replication of the E4 ORF3 mutant virus can be rescued following the introduction of E4 ORF3 from evolutionarily divergent adenoviruses, suggesting a conserved function for E4 ORF3 inhibition of the IFN-induced antiviral state. Furthermore, E4 ORF3 inhibition of an IFN-induced response is unrelated to the inhibition of adenovirus replication by the Mre11-Rad50-Nbs1 DNA repair complex. We propose that the evolutionarily conserved function of the adenovirus E4 ORF3 protein is the inhibition of a host interferon response to viral infection via disruption of the PML oncogenic domain.


Subject(s)
Adenovirus E4 Proteins/metabolism , Cell Nucleus Structures/virology , Interferons/metabolism , Open Reading Frames/genetics , Virus Replication , Adenovirus E4 Proteins/genetics , Animals , Cell Nucleus Structures/metabolism , Chlorocebus aethiops , DNA Replication/physiology , Immunoblotting , Microscopy, Fluorescence , Nuclear Proteins/metabolism , Vero Cells
5.
J Med Virol ; 78(12): 1542-53, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17063514

ABSTRACT

The ultrastructural replication cycle of human herpesvirus 6A and 6B, both T-lymphotropic viruses, with tropism for the central nervous system, was compared by electron microscopy in the same cells, that is, in the T-lymphoblastoid cell line SupT-1 and in human astrocytes. Both HHV-6A and HHV-6B replicated efficiently in SupT-1 and formed viral particles. The tegument is the least characterized structure of the herpesviral particle and both variants were able to form intranuclear membrane compartments called tegusomes in SupT-1 where tegumentation occurred. Also, tegumentation occurred in HHV-6A infected cells in the nucleoplasm without the presence of a tegusome. This suggests that there is more than one possible route of tegumentation. Differences in the replication cycles between HHV-6A and HHV-6B were also observed in the cytoplasm. One such difference was that prominent annulate lamellae were only found in the cytoplasm of HHV-6A infected cells. In astrocytes a successful formation of viral particles was only seen with the HHV-6A variant. The HHV-6A virus life cycle in astrocytes resembled the life cycle in the T-cell line SupT-1, except that no annulate lamellae were found. Complete viral particles were found extracellularly around the astrocytes and the supernatant of infected astrocytes were able to re-infect SupT-1 cells. This suggests that HHV-6A infection in astrocytes can generate complete, viable, and infectious viral particles. The HHV-6 variants behave differently in the same type of cells and have different tropisms for astrocytes, supporting the notion that the variants might induce different diseases.


Subject(s)
Astrocytes/virology , Cell Nucleus Structures/virology , Herpesvirus 6, Human/growth & development , T-Lymphocytes/virology , Virus Replication , Astrocytes/ultrastructure , Cell Nucleus Structures/physiology , Cell Nucleus Structures/ultrastructure , Cells, Cultured , Cytopathogenic Effect, Viral , Herpesvirus 6, Human/pathogenicity , Herpesvirus 6, Human/ultrastructure , Humans , Microscopy, Electron, Transmission , T-Lymphocytes/ultrastructure , Virion/growth & development , Virion/ultrastructure
6.
Science ; 312(5775): 875-8, 2006 May 12.
Article in English | MEDLINE | ID: mdl-16690857

ABSTRACT

The replication of many viruses is associated with specific intracellular compartments called virus factories or virioplasm. These are thought to provide a physical scaffold to concentrate viral components and thereby increase the efficiency of replication. The formation of virus replication sites often results in rearrangement of cellular membranes and reorganization of the cytoskeleton. Similar rearrangements are seen in cells in response to protein aggregation, where aggresomes and autophagosomes are produced to facilitate protein degradation. Here I review the evidence that some viruses induce aggresomes and autophagosomes to generate sites of replication.


Subject(s)
Autophagy , Cytoplasmic Vesicles/virology , DNA Viruses/physiology , Phagosomes/virology , RNA Viruses/physiology , Virus Replication , Cell Membrane Structures/ultrastructure , Cell Membrane Structures/virology , Cell Nucleus/ultrastructure , Cell Nucleus/virology , Cell Nucleus Structures/ultrastructure , Cell Nucleus Structures/virology , Cytoplasmic Vesicles/physiology , Cytoplasmic Vesicles/ultrastructure , Models, Biological , Phagosomes/physiology , Proteins/metabolism
7.
J Submicrosc Cytol Pathol ; 34(2): 177-86, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12117278

ABSTRACT

Intranuclear virus-like particles (VLPs) have been observed in different cell lines and adult tissues of Drosophila. In the present study, intranuclear VLPs have been found in larval tissues (salivary glands, midgut, fat body) as well as in adult tissues (midgut, genitals, fat body) of a rare interspecific hybrid (D. mauritiana x D. melanogaster) called 'mame'. The intranuclear VLPs were round or slightly elliptical with a diameter of 30 nm, and they were found mainly in highly organised clusters, forming large crystalline arrays, near the nucleolus and the polycene chromosomes. These particles were never observed in the cytoplasm of any mame's tissue. A few VLPs were also seen in the corresponding tissues of D. melanogaster, but they were never observed in any tissue of D. mauritiana. There is the intriguing possibility that these VLPs are related to transposable elements and probably contribute to the speciation process, in an unknown, so far, manner.


Subject(s)
Cell Nucleus Structures/virology , Drosophila melanogaster , Inclusion Bodies, Viral , Animals , Cell Nucleus Structures/ultrastructure , Chimera , Fat Body/ultrastructure , Fat Body/virology , Female , Inclusion Bodies, Viral/ultrastructure , Intestines/ultrastructure , Intestines/virology , Larva/physiology , Larva/ultrastructure , Larva/virology , Male , Ovary/ultrastructure , Ovary/virology , Salivary Glands/ultrastructure , Salivary Glands/virology , Testis/ultrastructure , Testis/virology
8.
Virus Genes ; 24(1): 5-10, 2002.
Article in English | MEDLINE | ID: mdl-11928989

ABSTRACT

The sequencing and deciphering of the human genome provided an insight into the gene complement of the human chromosomes as well as information on the nongenic sequences that constitute the chromosomal DNA molecules. The analyses of the genes and nongenic sequences in the human genome also provided important information on the presence of endogenous retroviruses, retroposons, retrotransposition of genes in the human genome as well as retroduplication of genes and distribution of the duplicated genes in different chromosomes. These issues were discussed in the first Special Issue of Virus Genes on Molecular Evolution of Viruses-Past and Present. In that issue, the discovery of the reverse transcriptase gene in archeabacteria, the retrovirus in drosophilae and endogenous retroviruses in the human genome were discussed. The aim of the present special issue on Molecular Evolution of Viruses is to consider the strategies developed by RNA and DNA viruses to control the nucleus and the nuclear functions in the infected cells.


Subject(s)
Biological Evolution , Genes, Immediate-Early/physiology , Genome, Human , Genome, Viral , Herpesviridae/genetics , Cell Nucleus Structures/virology , DNA, Viral/genetics , Evolution, Molecular , Humans
9.
J Virol ; 75(8): 3819-31, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11264371

ABSTRACT

Herpes simplex virus type 1 (HSV-1) immediate-early regulatory protein ICP0 stimulates the initiation of lytic infection and reactivation from quiescence in human fibroblast cells. These functions correlate with its ability to localize to and disrupt centromeres and specific subnuclear structures known as ND10, PML nuclear bodies, or promyelocytic oncogenic domains. Since the natural site of herpesvirus latency is in neurons, we investigated the status of ND10 and centromeres in uninfected and infected human cells with neuronal characteristics. We found that NT2 cells, a neuronally committed human teratocarcinoma cell line, have abnormal ND10 characterized by low expression of the major ND10 component PML and no detectable expression of another major ND10 antigen, Sp100. In addition, PML is less extensively modified by the ubiquitin-like protein SUMO-1 in NT2 cells compared to fibroblasts. After treatment with retinoic acid, NT2 cells differentiate into neuron-like hNT cells which express very high levels of both PML and Sp100. Infection of both NT2 and hNT cells by HSV-1 was poor compared to human fibroblasts, and after low-multiplicity infection yields of virus were reduced by 2 to 3 orders of magnitude. ICP0-deficient mutants were also disabled in the neuron-related cell lines, and cells quiescently infected with an ICP0-null virus could be established. These results correlated with less-efficient disruption of ND10 and centromeres induced by ICP0 in NT2 and hNT cells. Furthermore, the ability of ICP0 to activate gene expression in transfection assays in NT2 cells was poor compared to Vero cells. These results suggest that a contributory factor in the reduced HSV-1 replication in the neuron-related cells is inefficient ICP0 function; it is possible that this is pertinent to the establishment of latent infection in neurons in vivo.


Subject(s)
Cell Nucleus Structures/pathology , Herpesvirus 1, Human/physiology , Neurons/virology , Teratocarcinoma/pathology , Teratocarcinoma/virology , Cell Differentiation/drug effects , Cell Nucleus Structures/chemistry , Cell Nucleus Structures/virology , Centromere/metabolism , Chromosomal Proteins, Non-Histone/analysis , Fibroblasts/cytology , Fibroblasts/virology , Fluorescent Antibody Technique , Gene Deletion , Gene Expression Regulation, Viral , Herpesvirus 1, Human/genetics , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Kinetics , Mutation/genetics , Neurons/cytology , Neurons/drug effects , Superinfection/metabolism , Superinfection/virology , Transcriptional Activation , Tretinoin/pharmacology , Tumor Cells, Cultured , Ubiquitin-Protein Ligases , Virus Activation , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...