Cellular differentiation regulator BLIMP1 induces Epstein-Barr virus lytic reactivation in epithelial and B cells by activating transcription from both the R and Z …

JA Reusch, DM Nawandar, KL Wright… - Journal of …, 2015 - Am Soc Microbiol
JA Reusch, DM Nawandar, KL Wright, SC Kenney, JE Mertz
Journal of virology, 2015Am Soc Microbiol
Epstein-Barr virus (EBV) maintains a lifelong latent infection within a subset of its host's
memory B cells, while lytic EBV replication takes place in plasma cells and differentiated
epithelial cells. Therefore, cellular transcription factors, such as BLIMP1, that are key
mediators of differentiation likely contribute to the EBV latent-to-lytic switch. Previous reports
showed that ectopic BLIMP1 expression induces reactivation in some EBV-positive (EBV+)
B-cell lines and transcription from Zp, with all Z+ cells in oral hairy leukoplakia being …
Abstract
Epstein-Barr virus (EBV) maintains a lifelong latent infection within a subset of its host's memory B cells, while lytic EBV replication takes place in plasma cells and differentiated epithelial cells. Therefore, cellular transcription factors, such as BLIMP1, that are key mediators of differentiation likely contribute to the EBV latent-to-lytic switch. Previous reports showed that ectopic BLIMP1 expression induces reactivation in some EBV-positive (EBV+) B-cell lines and transcription from Zp, with all Z+ cells in oral hairy leukoplakia being BLIMP1+. Here, we examined BLIMP1's role in inducing EBV lytic gene expression in numerous EBV+ epithelial and B-cell lines and activating transcription from Rp. BLIMP1 addition was sufficient to induce reactivation in latently infected epithelial cells derived from gastric cancers, nasopharyngeal carcinomas, and normal oral keratinocytes (NOK) as well as some, but not all B-cell lines. BLIMP1 strongly induced transcription from Rp as well as Zp, with there being three or more synergistically acting BLIMP1-responsive elements (BRE) within Rp. BLIMP1's DNA-binding domain was required for reactivation, but BLIMP1 did not directly bind the nucleotide (nt) −660 Rp BRE. siRNA knockdown of BLIMP1 inhibited 12-O-tetradecanoyl-phorbol-13-acetate (TPA)-induced lytic reactivation in NOK-Akata cells, cells that can be reactivated by R, but not Z. Thus, we conclude that BLIMP1 expression is both necessary and sufficient to induce EBV lytic replication in many (possibly all) EBV+ epithelial-cell types, but in only a subset of EBV+ B-cell types; it does so, at least in part, by strongly activating expression of both EBV immediately early genes, BZLF1 and BRLF1.
IMPORTANCE This study is the first one to show that the cellular transcription factor BLIMP1, a key player in both epithelial and B-cell differentiation, induces reactivation of the oncogenic herpesvirus Epstein-Barr virus (EBV) out of latency into lytic replication in a variety of cancerous epithelial cell types as well as in some, but not all, B-cell types that contain this virus in a dormant state. The mechanism by which BLIMP1 does so involves strongly turning on expression of both of the immediate early genes of the virus, probably by directly acting upon the promoters as part of protein complexes or indirectly by altering the expression or activities of some cellular transcription factors and signaling pathways. The fact that EBV+ cancers usually contain mostly undifferentiated cells may be due in part to these cells dying from lytic EBV infection when they differentiate and express wild-type BLIMP1.
American Society for Microbiology