Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 5 de 5
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
J Virol ; 94(17)2020 08 17.
Article de Anglais | MEDLINE | ID: mdl-32581094

RÉSUMÉ

The Epstein-Barr virus (EBV) BHLF1 gene encodes an abundant linear and several circular RNAs believed to perform noncoding functions during virus replication, although an open reading frame (ORF) is retained among an unknown percentage of EBV isolates. Evidence suggests that BHLF1 is also transcribed during latent infection, which prompted us to investigate the contribution of this locus to latency. Analysis of transcripts transiting BHLF1 revealed that its transcription is widespread among B-cell lines supporting the latency I or III program of EBV protein expression and is more complex than originally presumed. EBV-negative Burkitt lymphoma cell lines infected with either wild-type or two different BHLF1 mutant EBVs were initially indistinguishable in supporting latency III. However, cells infected with BHLF1- virus ultimately transitioned to the more restrictive latency I program, whereas cells infected with wild-type virus either sustained latency III or transitioned more slowly to latency I. Upon infection of primary B cells, which require latency III for growth in vitro, both BHLF1- viruses exhibited variably reduced immortalization potential relative to the wild-type virus. Finally, in transfection experiments, efficient protein expression from an intact BHLF1 ORF required the EBV posttranscriptional regulator protein SM, whose expression is limited to the replicative cycle. Thus, one way in which BHLF1 may contribute to latency is through a mechanism, possibly mediated or regulated by a long noncoding RNA, that supports latency III critical for the establishment of EBV latency and lifelong persistence within its host, whereas any retained protein-dependent function of BHLF1 may be restricted to the replication cycle.IMPORTANCE Epstein-Barr virus (EBV) has significant oncogenic potential that is linked to its latent infection of B lymphocytes, during which virus replication is not supported. The establishment of latent infection, which is lifelong and can precede tumor development by years, requires the concerted actions of nearly a dozen EBV proteins and numerous small non-protein-coding RNAs. Elucidating how these EBV products contribute to latency is crucial for understanding EBV's role in specific malignancies and, ultimately, for clinical intervention. Historically, EBV genes that contribute to virus replication have been excluded from consideration of a role in latency, primarily because of the general incompatibility between virus production and cell survival. However, here, we provide evidence that the genetic locus containing one such gene, BHLF1, indeed contributes to key aspects of EBV latency, including its ability to promote the continuous growth of B lymphocytes, thus providing significant new insight into EBV biology and oncogenic potential.


Sujet(s)
Lymphocytes B/virologie , Herpèsvirus humain de type 4/génétique , Herpèsvirus humain de type 4/métabolisme , Protéines virales/génétique , Protéines virales/métabolisme , Latence virale/physiologie , Lymphome de Burkitt , Lignée cellulaire , Infections à virus Epstein-Barr/virologie , Antigènes nucléaires du virus d'Epstein-Barr/génétique , Antigènes nucléaires du virus d'Epstein-Barr/métabolisme , Régulation de l'expression des gènes viraux , Cellules HEK293 , Herpèsvirus humain de type 4/croissance et développement , Humains , ARN long non codant/génétique , Transcriptome , Réplication virale
2.
J Virol ; 92(18)2018 09 15.
Article de Anglais | MEDLINE | ID: mdl-29976663

RÉSUMÉ

Epstein Barr virus (EBV) is a potentially oncogenic gammaherpesvirus that establishes a chronic, latent infection in memory B cells. The EBV genome persists in infected host cells as a chromatinized episome and is subject to chromatin-mediated regulation. Binding of the host insulator protein CTCF to the EBV genome has an established role in maintaining viral latency type. CTCF is posttranslationally modified by the host enzyme PARP1. PARP1, or poly(ADP-ribose) polymerase 1, catalyzes the transfer of a poly(ADP-ribose) (PAR) moiety from NAD+ onto acceptor proteins, including itself, histone proteins, and CTCF. PARylation of CTCF by PARP1 can affect CTCF's insulator activity, DNA binding capacity, and ability to form chromatin loops. Both PARP1 and CTCF have been implicated in the regulation of EBV latency and lytic reactivation. Thus, we predicted that pharmacological inhibition with PARP1 inhibitors would affect EBV latency type through a chromatin-specific mechanism. Here, we show that PARP1 and CTCF colocalize at specific sites throughout the EBV genome and provide evidence to suggest that PARP1 acts to stabilize CTCF binding and maintain the open chromatin landscape at the active Cp promoter during type III latency. Further, PARP1 activity is important in maintaining latency type-specific viral gene expression. The data presented here provide a rationale for the use of PARP inhibitors in the treatment of EBV-associated cancers exhibiting type III latency and ultimately could contribute to an EBV-specific treatment strategy for AIDS-related or posttransplant lymphomas.IMPORTANCE EBV is a human gammaherpesvirus that infects more than 95% of individuals worldwide. Upon infection, EBV circularizes as an episome and establishes a chronic, latent infection in B cells. In doing so, the virus utilizes host cell machinery to regulate and maintain the viral genome. In otherwise healthy individuals, EBV infection is typically nonpathological; however, latent infection is potentially oncogenic and is responsible for 1% of human cancers. During latent infection, EBV expresses specific sets of proteins according to the given latency type, each of which is associated with specific types of cancers. For example, type III latency, in which the virus expresses its full repertoire of latent proteins, is characteristic of AIDS-associated and posttransplant lymphomas associated with EBV infection. Understanding how viral latency type is regulated at the chromatin level may reveal potential targets for EBV-specific pharmacological intervention in EBV-associated cancers.


Sujet(s)
Facteur de liaison à la séquence CCCTC/génétique , Herpèsvirus humain de type 4/physiologie , Poly (ADP-Ribose) polymerase-1/génétique , Latence virale/génétique , Facteur de liaison à la séquence CCCTC/métabolisme , Lignée cellulaire tumorale , Chromatine/génétique , Chromatine/métabolisme , Épigénomique , Régulation de l'expression des gènes viraux , Génome viral , Herpèsvirus humain de type 4/composition chimique , Herpèsvirus humain de type 4/génétique , Humains , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Poly (ADP-Ribose) polymerase-1/effets des médicaments et des substances chimiques , Leucémie-lymphome lymphoblastique à précurseurs B et T/virologie , Régions promotrices (génétique) , Transcription génétique , Latence virale/effets des médicaments et des substances chimiques
3.
Oncotarget ; 9(12): 10585-10605, 2018 Feb 13.
Article de Anglais | MEDLINE | ID: mdl-29535829

RÉSUMÉ

The enzyme Poly(ADP-ribose) polymerase 1 (PARP1) plays a very important role in the DNA damage response, but its role in numerous aspects is not fully understood. We recently showed that in the absence of DNA damage, PARP1 regulates the expression of the chromatin-modifying enzyme EZH2. Work from other groups has shown that EZH2 participates in the DNA damage response. These combined data suggest that EZH2 could be a target of PARP1 in both untreated and genotoxic agent-treated conditions. In this work we tested the hypothesis that, in response to DNA damage, PARP1 regulates EZH2 activity. Here we report that PARP1 regulates EZH2 activity after DNA damage. In particular, we find that EZH2 is a direct target of PARP1 upon induction of alkylating and UV-induced DNA damage in cells and in vitro. PARylation of EZH2 inhibits EZH2 histone methyltransferase (H3K27me) enzymatic activity. We observed in cells that the induction of PARP1 activity by DNA alkylating agents decreases the association of EZH2 with chromatin, and PARylation of histone H3 reduces EZH2 affinity for its target histone H3. Our findings establish that PARP1 and PARylation are important regulators of EZH2 function and link EZH2-mediated heterochromatin formation, DNA damage and PARylation. These findings may also have clinical implications, as they suggest that inhibitors of EZH2 can improve anti-tumor effects of PARP1 inhibitors in BRCA1/2-deficient cancers.

4.
Mol Cancer Res ; 15(8): 967-972, 2017 08.
Article de Anglais | MEDLINE | ID: mdl-28634224

RÉSUMÉ

Burkitt lymphoma/leukemia cells carry t(8;14)(q24;q32) chromosomal translocation encoding IGH/MYC, which results in the constitutive expression of the MYC oncogene. Here, it is demonstrated that untreated and cytarabine (AraC)-treated IGH/MYC-positive Burkitt lymphoma cells accumulate a high number of potentially lethal DNA double-strand breaks (DSB) and display low levels of the BRCA2 tumor suppressor protein, which is a key element of homologous recombination (HR)-mediated DSB repair. BRCA2 deficiency in IGH/MYC-positive cells was associated with diminished HR activity and hypersensitivity to PARP1 inhibitors (olaparib, talazoparib) used alone or in combination with cytarabine in vitro Moreover, talazoparib exerted a therapeutic effect in NGS mice bearing primary Burkitt lymphoma xenografts. In conclusion, IGH/MYC-positive Burkitt lymphoma/leukemia cells have decreased BRCA2 and are sensitive to PARP1 inhibition alone or in combination with other chemotherapies.Implications: This study postulates that IGH/MYC-induced BRCA2 deficiency may predispose Burkitt lymphoma cells to synthetic lethality triggered by PARP1 inhibitors.Visual Overview: http://mcr.aacrjournals.org/content/molcanres/15/8/967/F1.large.jpgMol Cancer Res; 15(8); 967-72. ©2017 AACR.


Sujet(s)
Protéine BRCA2/génétique , Lymphome de Burkitt/traitement médicamenteux , Cassures double-brin de l'ADN/effets des médicaments et des substances chimiques , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Animaux , Protéine BRCA2/déficit , Lymphome de Burkitt/génétique , Cytarabine/administration et posologie , Réparation de l'ADN/effets des médicaments et des substances chimiques , Gènes myc/génétique , Recombinaison homologue/effets des médicaments et des substances chimiques , Humains , Souris , Phtalazines/administration et posologie , Pipérazines/administration et posologie , Poly (ADP-Ribose) polymerase-1/génétique , Mutations synthétiques létales/génétique , Translocation génétique/génétique , Tests d'activité antitumorale sur modèle de xénogreffe
5.
Virology ; 507: 220-230, 2017 07.
Article de Anglais | MEDLINE | ID: mdl-28456021

RÉSUMÉ

The Epstein Barr virus (EBV) genome persists in infected host cells as a chromatinized episome and is subject to chromatin-mediated regulation. Binding of the host insulator protein CTCF to the EBV genome has an established role in maintaining viral latency type, and in other herpesviruses, loss of CTCF binding at specific regions correlates with viral reactivation. Here, we demonstrate that binding of PARP1, an important cofactor of CTCF, at the BZLF1 lytic switch promoter restricts EBV reactivation. Knockdown of PARP1 in the Akata-EBV cell line significantly increases viral copy number and lytic protein expression. Interestingly, CTCF knockdown has no effect on viral reactivation, and CTCF binding across the EBV genome is largely unchanged following reactivation. Moreover, EBV reactivation attenuates PARP activity, and Zta expression alone is sufficient to decrease PARP activity. Here we demonstrate a restrictive function of PARP1 in EBV lytic reactivation.


Sujet(s)
Infections à virus Epstein-Barr/enzymologie , Infections à virus Epstein-Barr/virologie , Herpèsvirus humain de type 4/physiologie , Poly (ADP-Ribose) polymerase-1/métabolisme , Régions promotrices (génétique) , Transactivateurs/génétique , Activation virale , Lignée cellulaire , Infections à virus Epstein-Barr/génétique , Régulation de l'expression des gènes viraux , Herpèsvirus humain de type 4/génétique , Interactions hôte-pathogène , Humains , Poly (ADP-Ribose) polymerase-1/génétique , Liaison aux protéines , Transactivateurs/métabolisme , Latence virale
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...