Human cytomegalovirus microRNAs miR-US5-1 and miR-UL112-3p block proinflammatory cytokine production in response to NF-κB-activating factors through direct downregulation of IKKα and IKKβ

Meaghan H. Hancock, Lauren M. Hook, Jennifer Mitchell, Jay A. Nelson

Research output: Contribution to journalArticlepeer-review

43 Scopus citations

Abstract

Emerging evidence indicates that human cytomegalovirus (HCMV) manipulates host cell signaling pathways using both proteins and noncoding RNAs. Several studies have shown that HCMV induces NF-κB signaling early in infection, resulting in the induction of antiviral proinflammatory cytokines with a subsequent reduction of these cytokines late in infection. The mechanism for late cytokine reduction is unknown. In this study, we show that HCMV micro RNAs (mi RNAs) mi R-US5-1 and mi R-UL112-3p target the IκB kinase (IKK) complex components IKKα and IKKβ to limit production of proinflammatory cytokines in response to interleukin 1β (IL-1β) and tumor necrosis factor alpha (TNF-α). Transfection of mi R-UL112-3p and mi R-US5-1 mimics reduced endogenous IKKα and IKKβ protein levels, and site-directed mutagenesis of the 3= untranslated regions (UTRs) identified the binding sites for each mi RNA. Infection with mutant viruses lacking these mi RNAs resulted in increased levels of IKKα and IKKβ proteins, an impaired ability to control NF-κB signaling at late times of lytic infection, and increased production of proinflammatory cytokines compared to wild-type virus in cell types relevant to HCMV infection in vivo. These phenotypes were rescued by preexpression of mi R-US5-1 and mi R-UL112-3p in infected cells or by a mi R-US5-1/mi R-UL112-3p double mutant virus that expresses short hairpin RNAs (sh RNAs) targeting IKKα and IKKβ, demonstrating the gene specificity of the mi RNAs. These observations describe a mechanism through which HCMV mi RNAs expressed late in the infectious cycle downregulate proinflammatory cytokine production to create a cellular proviral environment. IMPORTANCE Human cytomegalovirus (HCMV) is a significant cause of morbidity and mortality in transplant recipients and causes hearing loss and mental retardation when acquired congenitally. Initial events during HCMV infection result in the activation of NF-κB signaling, which culminates in the production of IL-6, CCL5, and TNF-α. Several viruses have developed mechanisms to block the antiviral effects of these cytokines. We show here that two HCMV mi RNAs, mi R-US5-1 and mi R-UL112-3p, specifically downregulate IKKα and IKKβ signaling factors necessary to propagate NF-κB signaling and subsequent IL-6, CCL5, and TNF-α production. Regulation of these proinflammatory cytokines during lytic infection and during latency is critical to viral survival in the host. IMPORTANCE Human cytomegalovirus (HCMV) is a significant cause of morbidity and mortality in transplant recipients and causes hearing loss and mental retardation when acquired congenitally. Initial events during HCMV infection result in the activation of NF-кB signaling, which culminates in the production of IL-6, CCL5, and TNF-α. Several viruses have developed mechanisms to block the antiviral effects of these cytokines. We show here that two HCMV miRNAs, miR-US5-1 and miR-UL112-3p, specifically downregulate IKKα and IKKβ signaling factors necessary to propagate NF-кB signaling and subsequent IL-6, CCL5, and TNF-α production. Regulation of these proinflammatory cytokines during lytic infection and during latency is critical to viral survival in the host.

Original languageEnglish (US)
Article numbere00109-17
JournalmBio
Volume8
Issue number2
DOIs
StatePublished - Mar 1 2017

ASJC Scopus subject areas

  • Microbiology
  • Virology

Fingerprint

Dive into the research topics of 'Human cytomegalovirus microRNAs miR-US5-1 and miR-UL112-3p block proinflammatory cytokine production in response to NF-κB-activating factors through direct downregulation of IKKα and IKKβ'. Together they form a unique fingerprint.

Cite this