Abstract

Complement receptor 3 (CR3, CD11b/CD18) is a major macrophage phagocytic receptor. The biochemical pathways through which CR3 regulates immunologic responses have not been fully characterized. Francisella tularensis is a remarkably infectious, facultative intracellular pathogen of macrophages that causes tularemia. Early evasion of the host immune response contributes to the virulence of F. tularensis and CR3 is an important receptor for its phagocytosis. Here we confirm that efficient attachment and uptake of the highly virulent Type A F. tularensis spp. tularensis strain Schu S4 by human monocyte-derived macrophages (hMDMs) requires complement C3 opsonization and CR3. However, despite a>40-fold increase in uptake following C3 opsonization, Schu S4 induces limited pro-inflammatory cytokine production compared with non-opsonized Schu S4 and the low virulent F. novicida. This suggests that engagement of CR3 by opsonized Schu S4 contributes specifically to the immune suppression during and shortly following phagocytosis which we demonstrate by CD11b siRNA knockdown in hMDMs. This immune suppression is concomitant with early inhibition of ERK1/2, p38 MAPK and NF-κB activation. Furthermore, TLR2 siRNA knockdown shows that pro-inflammatory cytokine production and MAPK activation in response to non-opsonized Schu S4 depends on TLR2 signaling providing evidence that CR3-TLR2 crosstalk mediates immune suppression for opsonized Schu S4. Deletion of the CD11b cytoplasmic tail reverses the CR3-mediated decrease in ERK and p38 activation during opsonized Schu-S4 infection. The CR3-mediated signaling pathway involved in this immune suppression includes Lyn kinase and Akt activation, and increased MKP-1, which limits TLR2-mediated pro-inflammatory responses. These data indicate that while the highly virulent F. tularensis uses CR3 for efficient uptake, optimal engagement of this receptor down-regulates TLR2-dependent pro-inflammatory responses by inhibiting MAPK activation through outside-in signaling. CR3-linked immune suppression is an important mechanism involved in the pathogenesis of F. tularensis infection.

Highlights

  • Francisella tularensis is a remarkably infectious facultative intracellular pathogen that causes the zoonotic disease tularemia [1,2]

  • We first set out to confirm the role of C3 and C3 receptors in Schu S4 phagocytosis by human monocyte-derived macrophages

  • We provide evidence for a pathway that links complement receptor 3 (CR3)-mediated phagocytosis for F. tularensis with immune suppression that involves crosstalk with TLR2

Read more

Summary

Introduction

Francisella tularensis is a remarkably infectious facultative intracellular pathogen that causes the zoonotic disease tularemia [1,2]. As is the case for many other intracellular pathogens, F. tularensis is able to avoid and/or suppress macrophage host defense mechanisms to enable survival and intracellular replication following phagocytosis. Macrophages combat F. tularensis infection by generating TLR2dependent pro-inflammatory cytokines such as TNF-a and IL-1b [7,8,9,10,11,12]. This cytokine response is largely muted at the early stage of F. tularensis infection. Several studies have indicated that F. tularensis infection leads to broad immune suppression in Author Summary

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