Abstract

Plasmacytoid dendritic cells (pDCs) play important roles in antiviral innate immunity by producing type I interferon (IFN). In this study, we assess the immune responses of primary human pDCs to two poxviruses, vaccinia and myxoma virus. Vaccinia, an orthopoxvirus, was used for immunization against smallpox, a contagious human disease with high mortality. Myxoma virus, a Leporipoxvirus, causes lethal disease in rabbits, but is non-pathogenic in humans. We report that myxoma virus infection of human pDCs induces IFN-α and TNF production, whereas vaccinia infection does not. Co-infection of pDCs with myxoma virus plus vaccinia blocks myxoma induction effects. We find that heat-inactivated vaccinia (Heat-VAC; by incubating the virus at 55°C for 1 h) gains the ability to induce IFN-α and TNF in primary human pDCs. Induction of IFN-α in pDCs by myxoma virus or Heat-VAC is blocked by chloroquine, which inhibits endosomal acidification required for TLR7/9 signaling, and by inhibitors of cellular kinases PI3K and Akt. Using purified pDCs from genetic knockout mice, we demonstrate that Heat-VAC-induced type I IFN production in pDCs requires the endosomal RNA sensor TLR7 and its adaptor MyD88, transcription factor IRF7 and the type I IFN feedback loop mediated by IFNAR1. These results indicate that (i) vaccinia virus, but not myxoma virus, expresses inhibitor(s) of the poxvirus sensing pathway(s) in pDCs; and (ii) Heat-VAC infection fails to produce inhibitor(s) but rather produces novel activator(s), likely viral RNA transcripts that are sensed by the TLR7/MyD88 pathway. Using vaccinia gene deletion mutants, we show that the Z-DNA/RNA binding domain at the N-terminus of the vaccinia immunomodulatory E3 protein is an antagonist of the innate immune response of human pDCs to poxvirus infection and TLR agonists. The myxoma virus ortholog of vaccinia E3 (M029) lacks the N-terminal Z-DNA/RNA binding domain, which might contribute to the immunostimulating properties of myxoma virus.

Highlights

  • Induction of antiviral effectors like type I interferon (IFN) in a nonpermissive host underlies one mechanism that restricts poxvirus host tropism [1]

  • To test whether primary human plasmacytoid dendritic cells (pDCs) respond differently to vaccinia and myxoma virus, we purified pDCs from human peripheral blood mononuclear cells using anti-BDCA-4 antibody-coated magnetic beads

  • To better understand vaccinia inhibition of poxvirus sensing in pDCs, we focused our attention on the vaccinia E3 protein, a 190aa polypeptide composed of two distinct domains: an N-terminal Z-DNA/RNA binding domain (ZBD) and a C-terminal dsRNA

Read more

Summary

Introduction

Induction of antiviral effectors like type I interferon (IFN) in a nonpermissive host underlies one mechanism that restricts poxvirus host tropism [1]. The interactions of poxviruses with the sentinel cells of the host immune system, with plasmacytoid dendritic cells (pDCs), are of significance because: (i) pDCs are potent producers of type I IFN during virus infections [2]; (ii) through the production of type I IFN, pDCs activate NK cells, conventional DCs, Bcells, and T cells to augment antiviral innate and adaptive immunity [3]; and (iii) type I IFN signaling is crucial for protection of mice against infection by vaccinia virus [4] or myxoma virus [5]. We hypothesize that myxoma virus and vaccinia are sensed differently and trigger different immune responses in infected innate sentinel cells, such as pDCs, that might contribute to their recognition by early immune response pathways, and affect their pathogenesis and immunogenicity in humans

Methods
Results
Conclusion
Full Text
Published version (Free)

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