Hepatitis B e Antigen Inhibits NF-κB Activity by Interrupting K63-Linked Ubiquitination of NEMO

J Virol. 2019 Jan 4;93(2):e00667-18. doi: 10.1128/JVI.00667-18. Print 2019 Jan 15.

Abstract

Viruses have adopted diverse strategies to suppress antiviral responses. Hepatitis B virus (HBV), a virus that is prevalent worldwide, manipulates the host's innate immune system to evade scavenging. It is reported that the hepatitis B e antigen (HBeAg) can interfere with NF-κB activity, which then leads to high viral loads, while HBV with the G1896A mutation remains infectious without the production of HBeAg but can induce more severe proinflammatory response and liver damage. The aim of current work was to study the molecular mechanism by which HBeAg suppresses interleukin-1β (IL-1β)-stimulated NF-κB activity, which leads to the suppression of the innate immune responses to HBV infection. Our study revealed that HBeAg could interact with NEMO, a regulatory subunit associated with IκB kinase, which regulates the activation of NF-κB. HBeAg suppressed the IL-1β-induced tumor necrosis factor (TNF)-associated factor 6 (TRAF6)-dependent K63-linked ubiquitination of NEMO, thereby downregulating NF-κB activity and promoting virus replication. We further demonstrated the inhibitory effect of HBeAg on the NF-κB signaling pathway using primary human hepatocytes, HBV-infected HepG2-NTCP cells, and clinical liver samples. Our study reveals a molecular mechanism whereby HBeAg suppresses IL-1β-induced NF-κB activation by decreasing the TRAF6-dependent K63-linked ubiquitination of NEMO, which may thereby enhance HBV replication and promote a persistent infection.IMPORTANCE The role of HBeAg in inflammatory responses during the infection of hepatitis B virus (HBV) is not fully understood, and several previous reports with regard to the NF-κB pathway are controversial. In this study, we showed that HBeAg could suppress both Toll-like receptor 2 (TLR2)- and IL-1β-induced activation of NF-κB in cells and clinical samples, and we further revealed novel molecular mechanisms. We found that HBeAg can associate with NEMO, the regulatory subunit for IκB kinase (IKK) that controls the NF-κB signaling pathway, and thereby inhibits TRAF6-mediated K63-linked ubiquitination of NEMO, resulting in downregulation of NF-κB activity and promotion of virus replication. In contrast, the HBeAg-negative HBV mutant can induce higher levels of NF-κB activity. These results are important for understanding the HBV-induced pathogenesis of chronic hepatitis and indicate that different clinical measures should be considered to treat HBeAg-positive and HBeAg-negative infections. Our findings represent a conceptual advance in HBV-related suppression of NF-κB signaling.

Keywords: HBeAg; NEMO; NF-κB; hepatitis B virus; innate immunity; ubiquitination.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Adult
  • Female
  • HEK293 Cells
  • HeLa Cells
  • Hep G2 Cells
  • Hepatitis B / immunology*
  • Hepatitis B / virology
  • Hepatitis B e Antigens / metabolism*
  • Hepatitis B virus / immunology
  • Hepatitis B virus / metabolism*
  • Host-Pathogen Interactions
  • Humans
  • I-kappa B Kinase / chemistry
  • I-kappa B Kinase / metabolism*
  • Immunity, Innate
  • Interleukin-1beta / metabolism
  • Intracellular Signaling Peptides and Proteins
  • Lysine / metabolism
  • Male
  • Middle Aged
  • NF-kappa B / metabolism*
  • Signal Transduction
  • TNF Receptor-Associated Factor 6 / metabolism
  • Two-Hybrid System Techniques
  • Ubiquitination

Substances

  • Hepatitis B e Antigens
  • IKBKG protein, human
  • IL1B protein, human
  • Interleukin-1beta
  • Intracellular Signaling Peptides and Proteins
  • NF-kappa B
  • TNF Receptor-Associated Factor 6
  • Tifab protein, human
  • I-kappa B Kinase
  • Lysine