Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Proc Natl Acad Sci U S A ; 121(16): e2314426121, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38574017

ABSTRACT

Epstein-Barr Virus (EBV) infects more than 90% of the adult population worldwide. EBV infection is associated with Burkitt lymphoma (BL) though alone is not sufficient to induce carcinogenesis implying the involvement of co-factors. BL is endemic in African regions faced with mycotoxins exposure. Exposure to mycotoxins and oncogenic viruses has been shown to increase cancer risks partly through the deregulation of the immune response. A recent transcriptome profiling of B cells exposed to aflatoxin B1 (AFB1) revealed an upregulation of the Chemokine ligand 22 (CCL22) expression although the underlying mechanisms were not investigated. Here, we tested whether mycotoxins and EBV exposure may together contribute to endemic BL (eBL) carcinogenesis via immunomodulatory mechanisms involving CCL22. Our results revealed that B cells exposure to AFB1 and EBV synergistically stimulated CCL22 secretion via the activation of Nuclear Factor-kappa B pathway. By expressing EBV latent genes in B cells, we revealed that elevated levels of CCL22 result not only from the expression of the latent membrane protein LMP1 as previously reported but also from the expression of other viral latent genes. Importantly, CCL22 overexpression resulting from AFB1-exposure in vitro increased EBV infection through the activation of phosphoinositide-3-kinase pathway. Moreover, inhibiting CCL22 in vitro and in humanized mice in vivo limited EBV infection and decreased viral genes expression, supporting the notion that CCL22 overexpression plays an important role in B cell infection. These findings unravel new mechanisms that may underpin eBL development and identify novel pathways that can be targeted in drug development.


Subject(s)
Burkitt Lymphoma , Epstein-Barr Virus Infections , Animals , Mice , Herpesvirus 4, Human/genetics , Epstein-Barr Virus Infections/complications , Aflatoxin B1/toxicity , Ligands , Burkitt Lymphoma/metabolism , Chemokines , Carcinogenesis
2.
Proc Natl Acad Sci U S A ; 121(5): e2308776121, 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38252831

ABSTRACT

We present a drug design strategy based on structural knowledge of protein-protein interfaces selected through virus-host coevolution and translated into highly potential small molecules. This approach is grounded on Vinland, the most comprehensive atlas of virus-human protein-protein interactions with annotation of interacting domains. From this inspiration, we identified small viral protein domains responsible for interaction with human proteins. These peptides form a library of new chemical entities used to screen for replication modulators of several pathogens. As a proof of concept, a peptide from a KSHV protein, identified as an inhibitor of influenza virus replication, was translated into a small molecule series with low nanomolar antiviral activity. By targeting the NEET proteins, these molecules turn out to be of therapeutic interest in a nonalcoholic steatohepatitis mouse model with kidney lesions. This study provides a biomimetic framework to design original chemistries targeting cellular proteins, with indications going far beyond infectious diseases.


Subject(s)
Influenza, Human , Viruses , Animals , Mice , Humans , Proteome , Peptides/pharmacology , Drug Discovery
4.
Nucleic Acids Res ; 49(18): 10657-10676, 2021 10 11.
Article in English | MEDLINE | ID: mdl-34530456

ABSTRACT

Epstein-Barr virus (EBV) is a human herpesvirus associated with human cancers worldwide. Ex vivo, the virus efficiently infects resting human B lymphocytes and induces their continuous proliferation. This process is accompanied by a global reprogramming of cellular gene transcription. However, very little is known on the impact of EBV infection on the regulation of alternative splicing, a pivotal mechanism that plays an essential role in cell fate determination and is often deregulated in cancer. In this study, we have developed a systematic time-resolved analysis of cellular mRNA splice variant expression during EBV infection of resting B lymphocytes. Our results reveal that major modifications of alternative splice variant expression appear as early as day 1 post-infection and suggest that splicing regulation provides-besides transcription-an additional mechanism of gene expression regulation at the onset of B cell activation and proliferation. We also report a role for the viral proteins, EBNA2 and EBNA-LP, in the modulation of specific alternative splicing events and reveal a previously unknown function for EBNA-LP-together with the RBM4 splicing factor-in the alternative splicing regulation of two important modulators of cell proliferation and apoptosis respectively, NUMB and BCL-X.


Subject(s)
Alternative Splicing , B-Lymphocytes/virology , Herpesvirus 4, Human/physiology , Viral Proteins/metabolism , Cells, Cultured , Exons , Humans , Membrane Proteins/genetics , RNA Splice Sites , RNA-Binding Proteins/metabolism , Viral Proteins/physiology
5.
Nucleic Acids Res ; 48(22): 12502-12522, 2020 12 16.
Article in English | MEDLINE | ID: mdl-33264393

ABSTRACT

Coronaviruses represent a large family of enveloped RNA viruses that infect a large spectrum of animals. In humans, the severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) is responsible for the current COVID-19 pandemic and is genetically related to SARS-CoV and Middle East respiratory syndrome-related coronavirus (MERS-CoV), which caused outbreaks in 2002 and 2012, respectively. All viruses described to date entirely rely on the protein synthesis machinery of the host cells to produce proteins required for their replication and spread. As such, virus often need to control the cellular translational apparatus to avoid the first line of the cellular defense intended to limit the viral propagation. Thus, coronaviruses have developed remarkable strategies to hijack the host translational machinery in order to favor viral protein production. In this review, we will describe some of these strategies and will highlight the role of viral proteins and RNAs in this process.


Subject(s)
COVID-19/prevention & control , Genome, Viral/genetics , Protein Biosynthesis/genetics , RNA, Viral/genetics , SARS-CoV-2/genetics , Animals , COVID-19/epidemiology , COVID-19/virology , Gene Expression Regulation, Viral , Humans , Pandemics , SARS-CoV-2/physiology , Virus Replication
6.
PLoS Pathog ; 15(10): e1008093, 2019 10.
Article in English | MEDLINE | ID: mdl-31600344

ABSTRACT

ISG20 is a broad spectrum antiviral protein thought to directly degrade viral RNA. However, this mechanism of inhibition remains controversial. Using the Vesicular Stomatitis Virus (VSV) as a model RNA virus, we show here that ISG20 interferes with viral replication by decreasing protein synthesis in the absence of RNA degradation. Importantly, we demonstrate that ISG20 exerts a translational control over a large panel of non-self RNA substrates including those originating from transfected DNA, while sparing endogenous transcripts. This activity correlates with the protein's ability to localize in cytoplasmic processing bodies. Finally, these functions are conserved in the ISG20 murine ortholog, whose genetic ablation results in mice with increased susceptibility to viral infection. Overall, our results posit ISG20 as an important defense factor able to discriminate the self/non-self origins of the RNA through translation modulation.


Subject(s)
Antiviral Agents/pharmacology , Exoribonucleases/pharmacology , Protein Biosynthesis , RNA, Viral/metabolism , Vesicular Stomatitis/immunology , Vesiculovirus/immunology , Virus Replication/drug effects , Animals , Exoribonucleases/physiology , HeLa Cells , Humans , Mice , Mice, Knockout , RNA Stability , RNA, Viral/genetics , Vesicular Stomatitis/drug therapy , Vesicular Stomatitis/virology , Vesiculovirus/drug effects
7.
J Virol ; 93(13)2019 07 01.
Article in English | MEDLINE | ID: mdl-30996097

ABSTRACT

The histone modifier lysine (K)-specific demethylase 2B (KDM2B) plays a role in the differentiation of hematopoietic cells, and its expression appears to be deregulated in certain cancers of hematological and lymphoid origins. We have previously found that the KDM2B gene is differentially methylated in cell lines derived from Epstein-Barr virus (EBV)-associated endemic Burkitt lymphoma (eBL) compared with that in EBV-negative sporadic Burkitt lymphoma-derived cells. However, whether KDM2B plays a role in eBL development has not been previously investigated. Oncogenic viruses have been shown to hijack the host cell epigenome to complete their life cycle and to promote the transformation process by perturbing cell chromatin organization. Here, we investigated whether EBV alters KDM2B levels to enable its life cycle and promote B-cell transformation. We show that infection of B cells with EBV leads to downregulation of KDM2B levels. We also show that LMP1, one of the main EBV transforming proteins, induces increased DNMT1 recruitment to the KDM2B gene and augments its methylation. By altering KDM2B levels and performing chromatin immunoprecipitation in EBV-infected B cells, we show that KDM2B is recruited to the EBV gene promoters and inhibits their expression. Furthermore, forced KDM2B expression in immortalized B cells led to altered mRNA levels of some differentiation-related genes. Our data show that EBV deregulates KDM2B levels through an epigenetic mechanism and provide evidence for a role of KDM2B in regulating virus and host cell gene expression, warranting further investigations to assess the role of KDM2B in the process of EBV-mediated lymphomagenesis.IMPORTANCE In Africa, Epstein-Barr virus infection is associated with endemic Burkitt lymphoma, a pediatric cancer. The molecular events leading to its development are poorly understood compared with those leading to sporadic Burkitt lymphoma. In a previous study, by analyzing the DNA methylation changes in endemic compared with sporadic Burkitt lymphoma cell lines, we identified several differential methylated genomic positions in the proximity of genes with a potential role in cancer, and among them was the KDM2B gene. KDM2B encodes a histone H3 demethylase already shown to be involved in some hematological disorders. However, whether KDM2B plays a role in the development of Epstein-Barr virus-mediated lymphoma has not been investigated before. In this study, we show that Epstein-Barr virus deregulates KDM2B expression and describe the underlying mechanisms. We also reveal a role of the demethylase in controlling viral and B-cell gene expression, thus highlighting a novel interaction between the virus and the cellular epigenome.


Subject(s)
Epigenesis, Genetic , Epstein-Barr Virus Infections/metabolism , F-Box Proteins/genetics , F-Box Proteins/metabolism , Herpesvirus 4, Human/physiology , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Adolescent , Adult , B-Lymphocytes/virology , Burkitt Lymphoma/metabolism , Cell Line , Child , Child, Preschool , Chromatin/metabolism , Chromatin Immunoprecipitation , DNA Methylation , Down-Regulation , Epstein-Barr Virus Infections/genetics , Female , Gene Expression Regulation , Humans , Male , Middle Aged , Young Adult
8.
J Virol ; 92(3)2018 02 01.
Article in English | MEDLINE | ID: mdl-29142127

ABSTRACT

Epstein-Barr virus (EBV) expresses several mRNAs produced from intronless genes that could potentially be unfavorably translated compared to cellular spliced mRNAs. To overcome this situation, the virus encodes an RNA-binding protein (RBP) called EB2, which was previously found to both facilitate the export of nuclear mRNAs and increase their translational yield. Here, we show that EB2 binds both nuclear and cytoplasmic cap-binding complexes (CBC and eukaryotic initiation factor 4F [eIF4F], respectively) as well as the poly(A)-binding protein (PABP) to enhance translation initiation of a given messenger ribonucleoparticle (mRNP). Interestingly, such an effect can be obtained only if EB2 is initially bound to the native mRNPs in the nucleus. We also demonstrate that the EB2-eIF4F-PABP association renders translation of these mRNPs less sensitive to translation initiation inhibitors. Taken together, our data suggest that EB2 binds and stabilizes cap-binding complexes in order to increase mRNP translation and furthermore demonstrate the importance of the mRNP assembly process in the nucleus to promote protein synthesis in the cytoplasm.IMPORTANCE Most herpesvirus early and late genes are devoid of introns. However, it is now well documented that mRNA splicing facilitates recruitment on the mRNAs of cellular factors involved in nuclear mRNA export and translation efficiency. To overcome the absence of splicing of herpesvirus mRNAs, a viral protein, EB2 in the case of Epstein-Barr virus, is produced to facilitate the cytoplasmic accumulation of viral mRNAs. Although we previously showed that EB2 also specifically enhances translation of its target mRNAs, the mechanism was unknown. Here, we show that EB2 first is recruited to the mRNA cap structure in the nucleus and then interacts with the proteins eIF4G and PABP to enhance the initiation step of translation.


Subject(s)
Eukaryotic Initiation Factor-4G/metabolism , Peptide Chain Initiation, Translational , Phosphoproteins/metabolism , Poly(A)-Binding Proteins/metabolism , Trans-Activators/metabolism , Active Transport, Cell Nucleus , Cytoplasm/virology , HEK293 Cells , HeLa Cells , Herpesvirus 4, Human , Humans , Phosphoproteins/genetics , RNA Splicing , RNA Transport , RNA, Messenger/genetics , Trans-Activators/genetics
9.
PLoS Pathog ; 13(9): e1006610, 2017 09.
Article in English | MEDLINE | ID: mdl-28957419

ABSTRACT

IFITMs are broad antiviral factors that block incoming virions in endosomal vesicles, protecting target cells from infection. In the case of HIV-1, we and others reported the existence of an additional antiviral mechanism through which IFITMs lead to the production of virions of reduced infectivity. However, whether this second mechanism of inhibition is unique to HIV or extends to other viruses is currently unknown. To address this question, we have analyzed the susceptibility of a broad spectrum of viruses to the negative imprinting of the virion particles infectivity by IFITMs. The results we have gathered indicate that this second antiviral property of IFITMs extends well beyond HIV and we were able to identify viruses susceptible to the three IFITMs altogether (HIV-1, SIV, MLV, MPMV, VSV, MeV, EBOV, WNV), as well as viruses that displayed a member-specific susceptibility (EBV, DUGV), or were resistant to all IFITMs (HCV, RVFV, MOPV, AAV). The swapping of genetic elements between resistant and susceptible viruses allowed us to point to specificities in the viral mode of assembly, rather than glycoproteins as dominant factors of susceptibility. However, we also show that, contrarily to X4-, R5-tropic HIV-1 envelopes confer resistance against IFITM3, suggesting that viral receptors add an additional layer of complexity in the IFITMs-HIV interplay. Lastly, we show that the overall antiviral effects ascribed to IFITMs during spreading infections, are the result of a bimodal inhibition in which IFITMs act both by protecting target cells from incoming viruses and in driving the production of virions of reduced infectivity. Overall, our study reports for the first time that the negative imprinting of the virion particles infectivity is a conserved antiviral property of IFITMs and establishes IFITMs as a paradigm of restriction factor capable of interfering with two distinct phases of a virus life cycle.


Subject(s)
Antigens, Differentiation/metabolism , Virion , Virus Replication , Cell Line , HIV-1/physiology , Host-Pathogen Interactions , Humans , Virus Internalization
10.
Virologie (Montrouge) ; 23(5): 16-31, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31826846

ABSTRACT

Currently, more than 10% of human cancers are associated with viral infection. Studies on oncoviruses led to the development of clinical intervention strategies and elucidated fundamental cellular events altered upon cell transformation. Cancer cells exhibit several hallmarks including genomic instability, defined as a high frequency of mutations including gain or loss of chromosomes. The centrosome is an organelle that governs mitotic chromosome segregation and that functions as a signaling platform downstream of the DNA damage response. Here, we review the current literature to highlight how oncoviruses induce genomic instability via the deregulation of the centrosome. Viral interference with the centrosome duplication cycle, leading to centrosome amplification, is illustrated, with a special emphasis on mechanisms shared by several viral families. In addition, we discuss how oncoviruses could alter the signaling functions of the centrosome, and we comment on the bibliographic gaps that could be addressed by future research.


Subject(s)
Aneuploidy , Genomic Instability , Mitosis , Cell Transformation, Neoplastic/genetics , Centrosome , Genomic Instability/genetics , Humans , Mitosis/genetics
11.
Virologie (Montrouge) ; 23(5): 304-320, 2019 10 01.
Article in French | MEDLINE | ID: mdl-31826851

ABSTRACT

Currently, more than 10% of human cancers are associated with viral infection. Studies on oncoviruses led to the development of clinical intervention strategies and elucidated fundamental cellular events altered upon cell transformation. Cancer cells exhibit several hallmarks including genomic instability, defined as a high frequency of mutations including gain or loss of chromosomes. The centrosome is an organelle that governs mitotic chromosome segregation and that functions as a signaling platform downstream of the DNA damage response. Here, we review the current literature to highlight how oncoviruses induce genomic instability via the deregulation of the centrosome. Viral interference with the centrosome duplication cycle, leading to centrosome amplification, is illustrated, with a special emphasis on mechanisms shared by several viral families. In addition, we discuss how oncoviruses could alter the signaling functions of the centrosome, and we comment on the bibliographic gaps that could be addressed by future research.


Subject(s)
Aneuploidy , Genomic Instability , Mitosis , Cell Transformation, Neoplastic/genetics , Centrosome , Genomic Instability/genetics , Humans , Mitosis/genetics
12.
J Gen Virol ; 98(2): 251-265, 2017 02.
Article in English | MEDLINE | ID: mdl-28284242

ABSTRACT

The Epstein-Barr virus (EBV) nuclear antigen 1 (EBNA1) is a sequence-specific DNA-binding protein that plays an essential role in viral episome replication and segregation, by recruiting the cellular complex of DNA replication onto the origin (oriP) and by tethering the viral DNA onto the mitotic chromosomes. Whereas the mechanisms of viral DNA replication are well documented, those involved in tethering EBNA1 to the cellular chromatin are far from being understood. Here, we have identified regulator of chromosome condensation 1 (RCC1) as a novel cellular partner for EBNA1. RCC1 is the major nuclear guanine nucleotide exchange factor for the small GTPase Ran enzyme. RCC1, associated with chromatin, is involved in the formation of RanGTP gradients critical for nucleo-cytoplasmic transport, mitotic spindle formation and nuclear envelope reassembly following mitosis. Using several approaches, we have demonstrated a direct interaction between these two proteins and found that the EBNA1 domains responsible for EBNA1 tethering to the mitotic chromosomes are also involved in the interaction with RCC1. The use of an EBNA1 peptide array confirmed the interaction of RCC1 with these regions and also the importance of the N-terminal region of RCC1 in this interaction. Finally, using confocal microscopy and Förster resonance energy transfer analysis to follow the dynamics of interaction between the two proteins throughout the cell cycle, we have demonstrated that EBNA1 and RCC1 closely associate on the chromosomes during metaphase, suggesting an essential role for the interaction during this phase, perhaps in tethering EBNA1 to mitotic chromosomes.


Subject(s)
Cell Cycle Proteins/metabolism , Epstein-Barr Virus Nuclear Antigens/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Mitosis , Nuclear Proteins/metabolism , Protein Interaction Domains and Motifs , Amino Acid Motifs , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Chromatin/metabolism , Chromosomes, Human/metabolism , Epstein-Barr Virus Nuclear Antigens/chemistry , Epstein-Barr Virus Nuclear Antigens/genetics , Fluorescence Resonance Energy Transfer , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/genetics , HEK293 Cells , HeLa Cells , Humans , Metaphase , Microscopy, Confocal , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Protein Array Analysis , Protein Interaction Mapping , Spindle Apparatus/metabolism
13.
PLoS Pathog ; 10(5): e1004125, 2014 May.
Article in English | MEDLINE | ID: mdl-24809689

ABSTRACT

The DOK1 tumor suppressor gene encodes an adapter protein that acts as a negative regulator of several signaling pathways. We have previously reported that DOK1 expression is up-regulated upon cellular stress, via the transcription factor E2F1, and down-regulated in a variety of human malignancies due to aberrant hypermethylation of its promoter. Here we show that Epstein Barr virus (EBV) infection of primary human B-cells leads to the down-regulation of DOK1 gene expression via the viral oncoprotein LMP1. LMP1 alone induces recruitment to the DOK1 promoter of at least two independent inhibitory complexes, one containing E2F1/pRB/DNMT1 and another containing at least EZH2. These events result in tri-methylation of histone H3 at lysine 27 (H3K27me3) of the DOK1 promoter and gene expression silencing. We also present evidence that the presence of additional EBV proteins leads to further repression of DOK1 expression with an additional mechanism. Indeed, EBV infection of B-cells induces DNA methylation at the DOK1 promoter region including the E2F1 responsive elements that, in turn, lose the ability to interact with E2F complexes. Treatment of EBV-infected B-cell-lines with the methyl-transferase inhibitor 5-aza-2'-deoxycytidine rescues DOK1 expression. In summary, our data show the deregulation of DOK1 gene expression by EBV and provide novel insights into the regulation of the DOK1 tumor suppressor in viral-related carcinogenesis.


Subject(s)
DNA-Binding Proteins/genetics , Epstein-Barr Virus Infections/genetics , Herpesvirus 4, Human/physiology , Phosphoproteins/genetics , RNA-Binding Proteins/genetics , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Cell Transformation, Viral/genetics , Cells, Cultured , DNA Methylation , DNA-Binding Proteins/metabolism , Down-Regulation/genetics , Epstein-Barr Virus Infections/immunology , Gene Expression Regulation , Gene Silencing , Genes, Tumor Suppressor , Humans , Phosphoproteins/metabolism , Primary Cell Culture , RNA-Binding Proteins/metabolism , Viral Matrix Proteins/physiology
14.
J Immunol ; 193(7): 3398-408, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25194054

ABSTRACT

The stimulation of TLRs by pathogen-derived molecules leads to the production of proinflammatory cytokines. Because uncontrolled inflammation can be life threatening, TLR regulation is important; however, few studies have identified the signaling pathways that contribute to the modulation of TLR expression. In this study, we examined the relationship between activation and the transcriptional regulation of TLR9. We demonstrate that infection of primary human epithelial cells, B cells, and plasmacytoid dendritic cells with dsDNA viruses induces a regulatory temporary negative-feedback loop that blocks TLR9 transcription and function. TLR9 transcriptional downregulation was dependent on TLR9 signaling and was not induced by TLR5 or other NF-κB activators, such as TNF-α. Engagement of the TLR9 receptor induced the recruitment of a suppressive complex, consisting of NF-κBp65 and HDAC3, to an NF-κB cis element on the TLR9 promoter. Knockdown of HDAC3 blocked the transient suppression in which TLR9 function was restored. These results provide a framework for understanding the complex pathways involved in transcriptional regulation of TLR9, immune induction, and inflammation against viruses.


Subject(s)
DNA Virus Infections/immunology , DNA Viruses/immunology , Promoter Regions, Genetic/immunology , Toll-Like Receptor 9/immunology , Transcription, Genetic/immunology , Animals , DNA Virus Infections/genetics , DNA Virus Infections/pathology , Dendritic Cells/immunology , Dendritic Cells/pathology , Epithelial Cells/immunology , Epithelial Cells/pathology , Female , Gene Knockdown Techniques , HEK293 Cells , Histone Deacetylases/genetics , Histone Deacetylases/immunology , Humans , Male , Mice , NIH 3T3 Cells , Plasma Cells/immunology , Plasma Cells/pathology , Toll-Like Receptor 9/genetics , Transcription Factor RelA/genetics , Transcription Factor RelA/immunology , Transcription, Genetic/genetics
15.
Biochem J ; 472(1): 111-9, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26349537

ABSTRACT

mRNA is bound to a complex network of hundreds of RNA-binding proteins (RBPs) which constitute the mature ribonucleoprotein (mRNP). Such a complex particle is initially scaffolded in the nucleus and stays associated throughout mRNA's journey to the cytoplasm, where it participates in translation. However, due to the size, complexity and variability of the mRNP, it remains technically challenging to assess its impact on translation. By designing a novel in vitro translational assay, we have been able to compare the translational efficiency of reporter mRNAs that are, or are not, associated with their cognate RBPs. This showed the strong impact of these RBPs on translational efficiency, and revealed intrinsic variations according to the structure of both the mRNA and its nuclear history, e.g. the use of intron-containing mRNA constructs showed that splicing strongly enhanced translation. The present study shows that nuclear and cytoplasmic gene expression steps in vitro are coupled in eukaryotes and this is determined from the very birth of the mRNA in the nucleus by a network of hundreds of RBPs.


Subject(s)
Protein Biosynthesis , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Ribonucleoproteins/genetics , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cytoplasm/genetics , Cytoplasm/metabolism , Gene Expression , HeLa Cells , Humans , Internal Ribosome Entry Sites/genetics , Luciferases/genetics , Luciferases/metabolism , Models, Genetic , Protein Binding , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Ribonucleoproteins/metabolism , Ribosomes/genetics , Ribosomes/metabolism
16.
Nucleic Acids Res ; 42(15): 9700-16, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25092922

ABSTRACT

The Epstein-Barr virus (EBV) nuclear antigen 3 family of protein is critical for the EBV-induced primary B-cell growth transformation process. Using a yeast two-hybrid screen we identified 22 novel cellular partners of the EBNA3s. Most importantly, among the newly identified partners, five are known to play direct and important roles in transcriptional regulation. Of these, the Myc-interacting zinc finger protein-1 (MIZ-1) is a transcription factor initially characterized as a binding partner of MYC. MIZ-1 activates the transcription of a number of target genes including the cell cycle inhibitor CDKN2B. Focusing on the EBNA3A/MIZ-1 interaction we demonstrate that binding occurs in EBV-infected cells expressing both proteins at endogenous physiological levels and that in the presence of EBNA3A, a significant fraction of MIZ-1 translocates from the cytoplasm to the nucleus. Moreover, we show that a trimeric complex composed of a MIZ-1 recognition DNA element, MIZ-1 and EBNA3A can be formed, and that interaction of MIZ-1 with nucleophosmin (NPM), one of its coactivator, is prevented by EBNA3A. Finally, we show that, in the presence of EBNA3A, expression of the MIZ-1 target gene, CDKN2B, is downregulated and repressive H3K27 marks are established on its promoter region suggesting that EBNA3A directly counteracts the growth inhibitory action of MIZ-1.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p15/genetics , Epstein-Barr Virus Nuclear Antigens/metabolism , Gene Expression Regulation , Kruppel-Like Transcription Factors/metabolism , Repressor Proteins/metabolism , Transcription, Genetic , Alcohol Oxidoreductases/metabolism , Cell Nucleus/metabolism , Cyclin-Dependent Kinase Inhibitor p15/biosynthesis , DNA-Binding Proteins/metabolism , Down-Regulation , Epstein-Barr Virus Nuclear Antigens/chemistry , HEK293 Cells , HeLa Cells , Histones/metabolism , Humans , Kruppel-Like Transcription Factors/chemistry , Nuclear Proteins/metabolism , Nucleophosmin , Promoter Regions, Genetic , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins c-myc/metabolism , Repressor Proteins/chemistry
17.
PLoS Genet ; 9(2): e1003285, 2013.
Article in English | MEDLINE | ID: mdl-23408912

ABSTRACT

The differentiation of post-meiotic spermatids in animals is characterized by a unique reorganization of their nuclear architecture and chromatin composition. In many species, the formation of sperm nuclei involves the massive replacement of nucleosomes with protamines, followed by a phase of extreme nuclear compaction. At fertilization, the reconstitution of a nucleosome-based paternal chromatin after the removal of protamines requires the deposition of maternally provided histones before the first round of DNA replication. This process exclusively uses the histone H3 variant H3.3 and constitutes a unique case of genome-wide replication-independent (RI) de novo chromatin assembly. We had previously shown that the histone H3.3 chaperone HIRA plays a central role for paternal chromatin assembly in Drosophila. Although several conserved HIRA-interacting proteins have been identified from yeast to human, their conservation in Drosophila, as well as their actual implication in this highly peculiar RI nucleosome assembly process, is an open question. Here, we show that Yemanuclein (YEM), the Drosophila member of the Hpc2/Ubinuclein family, is essential for histone deposition in the male pronucleus. yem loss of function alleles affect male pronucleus formation in a way remarkably similar to Hira mutants and abolish RI paternal chromatin assembly. In addition, we demonstrate that HIRA and YEM proteins interact and are mutually dependent for their targeting to the decondensing male pronucleus. Finally, we show that the alternative ATRX/XNP-dependent H3.3 deposition pathway is not involved in paternal chromatin assembly, thus underlining the specific implication of the HIRA/YEM complex for this essential step of zygote formation.


Subject(s)
Cell Cycle Proteins , DNA-Binding Proteins , Drosophila Proteins , Histone Chaperones , Nuclear Proteins , Nucleosomes , Transcription Factors , Zygote , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chromatin/metabolism , Chromatin/ultrastructure , Chromatin Assembly and Disassembly , DNA Replication/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Fertilization/genetics , Histone Chaperones/genetics , Histone Chaperones/metabolism , Histones/genetics , Histones/metabolism , Male , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleosomes/genetics , Nucleosomes/metabolism , Spermatozoa/cytology , Spermatozoa/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Zygote/growth & development , Zygote/metabolism
18.
Carcinogenesis ; 36(11): 1440-51, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26424750

ABSTRACT

Although Epstein-Barr virus (EBV) infection is widely distributed, certain EBV-driven malignancies are geographically restricted. EBV-associated Burkitt's lymphoma (eBL) is endemic in children living in sub-Saharan Africa. This population is heavily exposed to food contaminated with the mycotoxin aflatoxin B1 (AFB1). Here, we show that exposure to AFB1 in in vitro and in vivo models induces activation of the EBV lytic cycle and increases EBV load, two events that are associated with an increased risk of eBL in vivo. AFB1 treatment leads to the alteration of cellular gene expression, with consequent activations of signaling pathways, e.g. PI3K, that in turn mediate reactivation of the EBV life cycle. Finally, we show that AFB1 triggers EBV-driven cellular transformation both in primary human B cells and in a humanized animal model. In summary, our data provide evidence for a role of AFB1 as a cofactor in EBV-mediated carcinogenesis.


Subject(s)
Aflatoxin B1/toxicity , B-Lymphocytes/virology , Burkitt Lymphoma/virology , Environmental Exposure , Epstein-Barr Virus Infections/pathology , Herpesvirus 4, Human/drug effects , Animals , B-Lymphocytes/pathology , Burkitt Lymphoma/chemically induced , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Cell Transformation, Neoplastic/drug effects , Cells, Cultured , Female , Herpesvirus 4, Human/physiology , Humans , Male , Mice, Inbred NOD , Mice, SCID , Signal Transduction , Virus Activation , Virus Replication/drug effects
19.
J Virol ; 88(21): 12825-38, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25165108

ABSTRACT

UNLABELLED: During their productive cycle, herpesviruses exhibit a strictly regulated temporal cascade of gene expression that has three general stages: immediate early (IE), early (E), and late (L). Promoter complexity differs strikingly between IE/E genes and L genes. IE and E promoters contain cis-regulating sequences upstream of a TATA box, whereas L promoters comprise a unique cis element. In the case of the gammaherpesviruses, this element is usually a TATT motif found in the position where the consensus TATA box of eukaryotic promoters is typically found. Epstein-Barr virus (EBV) encodes a protein, called BcRF1, which has structural homology with the TATA-binding protein and interacts specifically with the TATT box. However, although necessary for the expression of the L genes, BcRF1 is not sufficient, suggesting that other viral proteins are also required. Here, we present the identification and characterization of a viral protein complex necessary and sufficient for the expression of the late viral genes. This viral complex is composed of five different proteins in addition to BcRF1 and interacts with cellular RNA polymerase II. During the viral productive cycle, this complex, which we call the vPIC (for viral preinitiation complex), works in concert with the viral DNA replication machinery to activate expression of the late viral genes. The EBV vPIC components have homologs in beta- and gammaherpesviruses but not in alphaherpesviruses. Our results not only reveal that beta- and gammaherpesviruses encode their own transcription preinitiation complex responsible for the expression of the late viral genes but also indicate the close evolutionary history of these viruses. IMPORTANCE: Control of late gene transcription in DNA viruses is a major unsolved question in virology. In eukaryotes, the first step in transcriptional activation is the formation of a permissive chromatin, which allows assembly of the preinitiation complex (PIC) at the core promoter. Fixation of the TATA box-binding protein (TBP) is a key rate-limiting step in this process. This study provides evidence that EBV encodes a complex composed of six proteins necessary for the expression of the late viral genes. This complex is formed around a viral TBP-like protein and interacts with cellular RNA polymerase II, suggesting that it is directly involved in the assembly of a virus-specific PIC (vPIC).


Subject(s)
Herpesvirus 4, Human/physiology , Host-Pathogen Interactions , RNA Polymerase II/metabolism , Transcription Initiation, Genetic , Transcription, Genetic , Viral Proteins/metabolism , Cell Line , Herpesvirus 4, Human/genetics , Humans , Protein Binding
20.
PLoS Pathog ; 9(3): e1003186, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23516355

ABSTRACT

Many studies have proved that oncogenic viruses develop redundant mechanisms to alter the functions of the tumor suppressor p53. Here we show that Epstein-Barr virus (EBV), via the oncoprotein LMP-1, induces the expression of ΔNp73α, a strong antagonist of p53. This phenomenon is mediated by the LMP-1 dependent activation of c-Jun NH2-terminal kinase 1 (JNK-1) which in turn favours the recruitment of p73 to ΔNp73α promoter. A specific chemical inhibitor of JNK-1 or silencing JNK-1 expression strongly down-regulated ΔNp73α mRNA levels in LMP-1-containing cells. Accordingly, LMP-1 mutants deficient to activate JNK-1 did not induce ΔNp73α accumulation. The recruitment of p73 to the ΔNp73α promoter correlated with the displacement of the histone-lysine N-methyltransferase EZH2 which is part of the transcriptional repressive polycomb 2 complex. Inhibition of ΔNp73α expression in lymphoblastoid cells (LCLs) led to the stimulation of apoptosis and up-regulation of a large number of cellular genes as determined by whole transcriptome shotgun sequencing (RNA-seq). In particular, the expression of genes encoding products known to play anti-proliferative/pro-apoptotic functions, as well as genes known to be deregulated in different B cells malignancy, was altered by ΔNp73α down-regulation. Together, these findings reveal a novel EBV mechanism that appears to play an important role in the transformation of primary B cells.


Subject(s)
B-Lymphocytes/metabolism , DNA-Binding Proteins/genetics , Gene Expression Regulation, Viral , Herpesvirus 4, Human/genetics , Nuclear Proteins/genetics , Tumor Suppressor Proteins/genetics , Viral Matrix Proteins/genetics , Apoptosis , B-Lymphocytes/virology , Cell Transformation, Viral/genetics , Cell Transformation, Viral/physiology , DNA-Binding Proteins/metabolism , Down-Regulation , Epigenesis, Genetic , Herpesvirus 4, Human/physiology , Host-Pathogen Interactions , Humans , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , Sequence Analysis, RNA , Transcription, Genetic , Transcriptional Activation , Tumor Protein p73 , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Proteins/metabolism , Up-Regulation , Viral Matrix Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL