Fine tuning inflammation at the front door: macrophage complement receptor 3-mediates phagocytosis and immune suppression for Francisella tularensis.

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 c...

Full description

Bibliographic Details
Main Authors: Shipan Dai, Murugesan V S Rajaram, Heather M Curry, Rachel Leander, Larry S Schlesinger
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2013-01-01
Series:PLoS Pathogens
Online Access:http://europepmc.org/articles/PMC3554622?pdf=render
id doaj-5670708cfebb4193953b981b03821933
record_format Article
spelling doaj-5670708cfebb4193953b981b038219332020-11-25T01:58:12ZengPublic Library of Science (PLoS)PLoS Pathogens1553-73661553-73742013-01-0191e100311410.1371/journal.ppat.1003114Fine tuning inflammation at the front door: macrophage complement receptor 3-mediates phagocytosis and immune suppression for Francisella tularensis.Shipan DaiMurugesan V S RajaramHeather M CurryRachel LeanderLarry S SchlesingerComplement 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.http://europepmc.org/articles/PMC3554622?pdf=render
collection DOAJ
language English
format Article
sources DOAJ
author Shipan Dai
Murugesan V S Rajaram
Heather M Curry
Rachel Leander
Larry S Schlesinger
spellingShingle Shipan Dai
Murugesan V S Rajaram
Heather M Curry
Rachel Leander
Larry S Schlesinger
Fine tuning inflammation at the front door: macrophage complement receptor 3-mediates phagocytosis and immune suppression for Francisella tularensis.
PLoS Pathogens
author_facet Shipan Dai
Murugesan V S Rajaram
Heather M Curry
Rachel Leander
Larry S Schlesinger
author_sort Shipan Dai
title Fine tuning inflammation at the front door: macrophage complement receptor 3-mediates phagocytosis and immune suppression for Francisella tularensis.
title_short Fine tuning inflammation at the front door: macrophage complement receptor 3-mediates phagocytosis and immune suppression for Francisella tularensis.
title_full Fine tuning inflammation at the front door: macrophage complement receptor 3-mediates phagocytosis and immune suppression for Francisella tularensis.
title_fullStr Fine tuning inflammation at the front door: macrophage complement receptor 3-mediates phagocytosis and immune suppression for Francisella tularensis.
title_full_unstemmed Fine tuning inflammation at the front door: macrophage complement receptor 3-mediates phagocytosis and immune suppression for Francisella tularensis.
title_sort fine tuning inflammation at the front door: macrophage complement receptor 3-mediates phagocytosis and immune suppression for francisella tularensis.
publisher Public Library of Science (PLoS)
series PLoS Pathogens
issn 1553-7366
1553-7374
publishDate 2013-01-01
description 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.
url http://europepmc.org/articles/PMC3554622?pdf=render
work_keys_str_mv AT shipandai finetuninginflammationatthefrontdoormacrophagecomplementreceptor3mediatesphagocytosisandimmunesuppressionforfrancisellatularensis
AT murugesanvsrajaram finetuninginflammationatthefrontdoormacrophagecomplementreceptor3mediatesphagocytosisandimmunesuppressionforfrancisellatularensis
AT heathermcurry finetuninginflammationatthefrontdoormacrophagecomplementreceptor3mediatesphagocytosisandimmunesuppressionforfrancisellatularensis
AT rachelleander finetuninginflammationatthefrontdoormacrophagecomplementreceptor3mediatesphagocytosisandimmunesuppressionforfrancisellatularensis
AT larrysschlesinger finetuninginflammationatthefrontdoormacrophagecomplementreceptor3mediatesphagocytosisandimmunesuppressionforfrancisellatularensis
_version_ 1724970925033521152