scholarly journals Epigenetic control of Epstein–Barr virus transcription – relevance to viral life cycle?

2013 ◽  
Vol 4 ◽  
Author(s):  
Alison J. Sinclair
2019 ◽  
Vol 9 (1) ◽  
Author(s):  
Hyojin Song ◽  
Yoojoo Lim ◽  
Hogune Im ◽  
Jeong Mo Bae ◽  
Gyeong Hoon Kang ◽  
...  

2013 ◽  
Vol 9 (9) ◽  
pp. e1003636 ◽  
Author(s):  
Michael J. McClellan ◽  
C. David Wood ◽  
Opeoluwa Ojeniyi ◽  
Tim J. Cooper ◽  
Aditi Kanhere ◽  
...  

2013 ◽  
Vol 94 (12) ◽  
pp. 2750-2758 ◽  
Author(s):  
Yi-Ru Liu ◽  
Sheng-Yen Huang ◽  
Jen-Yang Chen ◽  
Lily Hui-Ching Wang

Elevated levels of antibodies against Epstein–Barr virus (EBV) and the presence of viral DNA in plasma are reliable biomarkers for the diagnosis of nasopharyngeal carcinoma (NPC) in high-prevalence areas, such as South-East Asia. The presence of these viral markers in the circulation suggests that a minimal level of virus reactivation may have occurred in an infected individual, although the underlying mechanism of reactivation remains to be elucidated. Here, we showed that treatment with nocodazole, which provokes the depolymerization of microtubules, induces the expression of two EBV lytic cycle proteins, Zta and EA-D, in EBV-positive NPC cells. This effect was independent of mitotic arrest, as viral reactivation was not abolished in cells synchronized at interphase. Notably, the induction of Zta by nocodazole was mediated by transcriptional upregulation via protein kinase C (PKC). Pre-treatment with inhibitors for PKC or its downstream signalling partners p38 mitogen-activated protein kinase (MAPK) and c-Jun N-terminal kinase (JNK) abolished the nocodazole-mediated induction of Zta and EA-D. Interestingly, the effect of nocodazole, as well as colchicine and vinblastine, on lytic gene expression occurred only in NPC epithelial cells but not in cells derived from lymphocytes. These results establish a novel role of microtubule integrity in controlling the EBV life cycle through PKC and its downstream pathways, which represents a tissue-specific mechanism for controlling the life-cycle switch of EBV.


2013 ◽  
Vol 87 (9) ◽  
pp. 5311-5315 ◽  
Author(s):  
C. B. Whitehurst ◽  
M. K. Sanders ◽  
M. Law ◽  
F.-Z. Wang ◽  
J. Xiong ◽  
...  

1986 ◽  
Vol 14 (6) ◽  
pp. 2611-2620 ◽  
Author(s):  
Myriam Bodescot ◽  
Olivier Brison ◽  
Michel Perricaudet

2012 ◽  
Vol 86 (20) ◽  
pp. 11322-11332 ◽  
Author(s):  
E. K. Hoebe ◽  
C. Wille ◽  
E. S. Hopmans ◽  
A. R. Robinson ◽  
J. M. Middeldorp ◽  
...  

2018 ◽  
Author(s):  
Romina C. Vargas-Ayala ◽  
Antonin Jay ◽  
Hector Hernandez-Vargas ◽  
Audrey Diederichs ◽  
Alexis Robitaille ◽  
...  

AbstractHistone modifier lysine (K)-specific demethylase 2B(KDM2B) plays a role in hematopoietic cells differentiation and its expression appears to be deregulated in certain cancers of hematological and lymphoid origins. We have previously found that KDM2B gene is differentially methylated in cell lines derived from the Epstein-Barr virus (EBV) associated endemic Burkitt’s lymphomas (eBL) compared to EBV negative sporadic BL cells. However, whether KDM2B plays a role in eBL development has never been previously demonstrated. 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 would alter KDM2B levels to enable its life cycle and promote B-cells transformation. We show that infection of B-cells with EBV leads to down-regulation of KDM2B levels. We also show that LMP1, one of the main EBV transforming proteins, induces increased DNMT1 recruitment to KDM2B gene and augments its methylation. By altering KDM2B levels and performing chromatin immunoprecipitation in EBV infected B-cells, we were able to 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 to the sporadic Burkitt lymphoma. In a previous study, by analyzing the DNA methylation changes in endemic compared to sporadic Burkitt lymphomas cell lines, we identified several differential methylated genomic positions in proximity of genes with a potential role in cancer, among them 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 never 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-cells genes expression, thus highlighting a novel interaction between the virus and the cellular epigenome.


2017 ◽  
Vol 98 (3) ◽  
pp. 435-446 ◽  
Author(s):  
Caleb Cornaby ◽  
Jillian L Jafek ◽  
Cameron Birrell ◽  
Vera Mayhew ◽  
Lauren Syndergaard ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document