Abstract

Human gammaherpesviruses Epstein-Barr virus (EBV) and Kaposi’s sarcoma-associated herpesvirus (KSHV) persist as life-long infections alternating between latency and lytic replication. Human endogenous retroviruses (HERVs), via integration into the host genome, represent genetic remnants of ancient retroviral infections. Both show similar epigenetic silencing while dormant, but can reactivate in response to cell signaling cues or triggers that, for gammaherpesviruses, result in productive lytic replication. Given their co-existence with humans and shared epigenetic silencing, we asked if HERV expression might be linked to lytic activation of human gammaherpesviruses. We found ERVW-1 mRNA, encoding the functional HERV-W envelope protein Syncytin-1, along with other repeat class elements, to be elevated upon lytic activation of EBV. Knockdown/knockout of ERVW-1 reduced lytic activation of EBV and KSHV in response to various lytic cycle triggers. In this regard, reduced expression of immediate early proteins ZEBRA and RTA for EBV and KSHV, respectively, places Syncytin-1’s influence on lytic activation mechanistically upstream of the latent-to-lytic switch. Conversely, overexpression of Syncytin-1 enhanced lytic activation of EBV and KSHV in response to lytic triggers, though this was not sufficient to induce lytic activation in the absence of such triggers. Syncytin-1 is expressed in replicating B cell blasts and lymphoma-derived B cell lines where it appears to contribute to cell cycle progression. Together, human gammaherpesviruses and B cells appear to have adapted a dependency on Syncytin-1 that facilitates the ability of EBV and KSHV to activate lytic replication from latency, while promoting viral persistence during latency by contributing to B cell proliferation.

Highlights

  • Human gammaherpesviruses Epstein-Barr virus (EBV; Human herpesvirus 4) and Kaposi’s sarcoma-associated herpesvirus (KSHV; Human herpesvirus 8) cause persistent, life-long infections (Knipe and Howley, 2013)

  • Induction of the lytic phase of EBV was accomplished by exposure of cells to the histone deacetylase inhibitor (HDACi) sodium butyrate (NaB) for 3, 24, or 48 h, prior to incorporation of 5-Bromouridine into nascent RNA followed by immune-capture and sequencing of nascent transcripts (Bru-Seq) (Frey et al, 2020)

  • Because we observed Syncytin-1 in lymphoblastoid cell lines (LCL) and Burkitt lymphoma (BL)-derived cell lines, and found that it promotes the lytic phase of human gammaherpesviruses, we further explored its role in B cell biology

Read more

Summary

Introduction

Human gammaherpesviruses Epstein-Barr virus (EBV; Human herpesvirus 4) and Kaposi’s sarcoma-associated herpesvirus (KSHV; Human herpesvirus 8) cause persistent, life-long infections (Knipe and Howley, 2013). Regulation of these phases is central to their immune evasion, transmission, and pathogenesis in acute and chronic diseases as well as in associated lymphoproliferative diseases (LPD), lymphomas, and epithelial cell cancers (ThorleyLawson and Gross, 2004; Knipe and Howley, 2013; ThorleyLawson, 2015; Aneja and Yuan, 2017; Broussard and Damania, 2020) Both viruses are known to persist latently in B lymphocytes with periodic lytic (re)activation producing virus particles that can infect new B cells, expanding the pool of latently infected B cells or epithelial cells, supporting transmission to new hosts (Hong et al, 2005; Kalla et al, 2010; Knipe and Howley, 2013; Broussard and Damania, 2020). This contributes to high prevalence rates of 1–60% for KSHV among distinct populations and a near ubiquitous presence of EBV with greater than 90% prevalence overall (Henke-Gendo and Schulz, 2004; Knipe and Howley, 2013)

Methods
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call