[HTML][HTML] The EBV latent antigen 3C inhibits apoptosis through targeted regulation of interferon regulatory factors 4 and 8

S Banerjee, J Lu, Q Cai, A Saha, HC Jha… - PLoS …, 2013 - journals.plos.org
PLoS pathogens, 2013journals.plos.org
Epstein-Barr virus (EBV) is linked to a broad spectrum of B-cell malignancies. EBV nuclear
antigen 3C (EBNA3C) is an encoded latent antigen required for growth transformation of
primary human B-lymphocytes. Interferon regulatory factor 4 (IRF4) and 8 (IRF8) are
transcription factors of the IRF family that regulate diverse functions in B cell development.
IRF4 is an oncoprotein with anti-apoptotic properties and IRF8 functions as a regulator of
apoptosis and tumor suppressor in many hematopoietic malignancies. We now demonstrate …
Epstein-Barr virus (EBV) is linked to a broad spectrum of B-cell malignancies. EBV nuclear antigen 3C (EBNA3C) is an encoded latent antigen required for growth transformation of primary human B-lymphocytes. Interferon regulatory factor 4 (IRF4) and 8 (IRF8) are transcription factors of the IRF family that regulate diverse functions in B cell development. IRF4 is an oncoprotein with anti-apoptotic properties and IRF8 functions as a regulator of apoptosis and tumor suppressor in many hematopoietic malignancies. We now demonstrate that EBNA3C can contribute to B-cell transformation by modulating the molecular interplay between cellular IRF4 and IRF8. We show that EBNA3C physically interacts with IRF4 and IRF8 with its N-terminal domain in vitro and forms a molecular complex in cells. We identified the Spi-1/B motif of IRF4 as critical for EBNA3C interaction. We also demonstrated that EBNA3C can stabilize IRF4, which leads to downregulation of IRF8 by enhancing its proteasome-mediated degradation. Further, si-RNA mediated knock-down of endogenous IRF4 results in a substantial reduction in proliferation of EBV-transformed lymphoblastoid cell lines (LCLs), as well as augmentation of DNA damage-induced apoptosis. IRF4 knockdown also showed reduced expression of its targeted downstream signalling proteins which include CDK6, Cyclin B1 and c-Myc all critical for cell proliferation. These studies provide novel insights into the contribution of EBNA3C to EBV-mediated B-cell transformation through regulation of IRF4 and IRF8 and add another molecular link to the mechanisms by which EBV dysregulates cellular activities, increasing the potential for therapeutic intervention against EBV-associated cancers.
PLOS