Human Papillomavirus E7 Oncoprotein Subverts Host Innate Immunity via SUV39H1-Mediated Epigenetic Silencing of Immune Sensor Genes

J Virol. 2020 Jan 31;94(4):e01812-19. doi: 10.1128/JVI.01812-19. Print 2020 Jan 31.

Abstract

Subversion of innate immunity by oncoviruses, such as human papillomavirus (HPV), favors carcinogenesis because the mechanism(s) of viral immune evasion can also hamper cancer immunosurveillance. Previously, we demonstrated that high-risk (hr) HPVs trigger simultaneous epigenetic silencing of multiple effectors of innate immunity to promote viral persistence. Here, we expand on those observations and show that the HPV E7 oncoprotein upregulates the H3K9-specific methyltransferase, whose action shuts down the host innate immune response. Specifically, we demonstrate that SUV39H1 contributes to chromatin repression at the promoter regions of the viral nucleic acid sensors RIG-I and cGAS and the adaptor molecule STING in HPV-transformed cells. Inhibition of SUV39H1 leads to transcriptional activation of these genes, especially RIG-I, followed by increased beta interferon (IFN-β) and IFN-λ1 production after poly(dA·dT) or RIG-I agonist M8 transfection. Collectively, our findings provide new evidence that the E7 oncoprotein plays a central role in dampening host innate immunity and raise the possibility that targeting the downstream effector SUV39H1 or the RIG-I pathway is a viable strategy to treat viral and neoplastic disease.IMPORTANCE High-risk HPVs are major viral human carcinogens responsible for approximately 5% of all human cancers. The growth of HPV-transformed cells depends on the ability of viral oncoproteins to manipulate a variety of cellular circuits, including those involved in innate immunity. Here, we show that one of these strategies relies on E7-mediated transcriptional activation of the chromatin repressor SUV39H1, which then promotes epigenetic silencing of RIG-I, cGAS, and STING genes, thereby shutting down interferon secretion in HPV-transformed cells. Pharmacological or genetic inhibition of SUV39H1 restored the innate response in HPV-transformed cells, mostly through activation of RIG-I signaling. We also show that IFN production upon transfection of poly(dA·dT) or the RIG-I agonist M8 predominantly occurs through RIG-I signaling. Altogether, the reversible nature of the modifications associated with E7-mediated SUV39H1 upregulation provides a rationale for the design of novel anticancer and antiviral therapies targeting these molecules.

Keywords: HPV; RIG-I; SUV39H1; epigenetics; innate immunity.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Cell Line
  • DEAD Box Protein 58 / metabolism
  • Epigenesis, Genetic / genetics
  • HEK293 Cells
  • HeLa Cells
  • Host-Pathogen Interactions / genetics
  • Host-Pathogen Interactions / immunology
  • Humans
  • Immune Evasion / genetics
  • Immune Evasion / immunology
  • Immunity, Innate / genetics
  • Immunity, Innate / immunology
  • Interferon-beta / metabolism
  • Keratinocytes / virology
  • Membrane Proteins / metabolism
  • Methyltransferases / genetics
  • Methyltransferases / metabolism*
  • Nucleotidyltransferases / metabolism
  • Oncogene Proteins, Viral / metabolism
  • Papillomaviridae / metabolism*
  • Papillomaviridae / pathogenicity
  • Papillomavirus E7 Proteins / metabolism*
  • Papillomavirus E7 Proteins / physiology
  • Papillomavirus Infections / virology
  • Receptors, Immunologic
  • Repressor Proteins / genetics
  • Repressor Proteins / metabolism*
  • Signal Transduction / genetics
  • Transcriptional Activation / genetics

Substances

  • Membrane Proteins
  • Oncogene Proteins, Viral
  • Papillomavirus E7 Proteins
  • Receptors, Immunologic
  • Repressor Proteins
  • STING1 protein, human
  • Interferon-beta
  • SUV39H1 protein, human
  • Methyltransferases
  • Nucleotidyltransferases
  • cGAS protein, human
  • RIGI protein, human
  • DEAD Box Protein 58