TCRβ-expressing macrophages induced by a pathogenic murine malaria correlate with parasite burden and enhanced phagocytic activity.

Macrophages express a wide array of invariant receptors that facilitate host defense and mediate pathogenesis during pathogen invasion. We report on a novel population of CD11bhighCD14+F4/80+ macrophages that express TCRβ. This population expands dramatically during a Plasmodium berghei ANKA infecti...

Full description

Bibliographic Details
Main Authors: Miranda S Oakley, Joanna K Chorazeczewski, Maya Aleshnick, Vivek Anantharaman, Victoria Majam, Bhavna Chawla, Timothy G Myers, Qin Su, Winter A Okoth, Kazuyo Takeda, Adovi Akue, Mark KuKuruga, L Aravind, Sanjai Kumar
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2018-01-01
Series:PLoS ONE
Online Access:http://europepmc.org/articles/PMC6059462?pdf=render
id doaj-d11403876389432dbbf5a9e4209c03f2
record_format Article
spelling doaj-d11403876389432dbbf5a9e4209c03f22020-11-25T02:10:39ZengPublic Library of Science (PLoS)PLoS ONE1932-62032018-01-01137e020104310.1371/journal.pone.0201043TCRβ-expressing macrophages induced by a pathogenic murine malaria correlate with parasite burden and enhanced phagocytic activity.Miranda S OakleyJoanna K ChorazeczewskiMaya AleshnickVivek AnantharamanVictoria MajamBhavna ChawlaTimothy G MyersQin SuWinter A OkothKazuyo TakedaAdovi AkueMark KuKurugaL AravindSanjai KumarMacrophages express a wide array of invariant receptors that facilitate host defense and mediate pathogenesis during pathogen invasion. We report on a novel population of CD11bhighCD14+F4/80+ macrophages that express TCRβ. This population expands dramatically during a Plasmodium berghei ANKA infection and sequesters in the brain during experimental cerebral malaria. Importantly, measurement of TCRβ transcript and protein levels in macrophages in wildtype versus nude and Rag1 knockout mice establishes that the observed expression is not a consequence of passive receptor expression due to phagocytosis or trogocytosis of peripheral T cells or nonspecific antibody staining to an Fc receptor or cross reactive epitope. We also demonstrate that TCRβ on brain sequestered macrophages undergoes productive gene rearrangements and shows preferential Vβ usage. Remarkably, there is a significant correlation in the proportion of macrophages that express TCRβ and peripheral parasitemia. In addition, presence of TCRβ on the macrophage also correlates with a significant increase (1.9 fold) in the phagocytosis of parasitized erythrocytes. By transcriptional profiling, we identify a novel set of genes and pathways that associate with TCRβ expression by the macrophage. Expansion of TCRβ-expressing macrophages points towards a convergence of the innate and adaptive immune responses where both arms of the immune system cooperate to modulate the host response to malaria and possibly other infections.http://europepmc.org/articles/PMC6059462?pdf=render
collection DOAJ
language English
format Article
sources DOAJ
author Miranda S Oakley
Joanna K Chorazeczewski
Maya Aleshnick
Vivek Anantharaman
Victoria Majam
Bhavna Chawla
Timothy G Myers
Qin Su
Winter A Okoth
Kazuyo Takeda
Adovi Akue
Mark KuKuruga
L Aravind
Sanjai Kumar
spellingShingle Miranda S Oakley
Joanna K Chorazeczewski
Maya Aleshnick
Vivek Anantharaman
Victoria Majam
Bhavna Chawla
Timothy G Myers
Qin Su
Winter A Okoth
Kazuyo Takeda
Adovi Akue
Mark KuKuruga
L Aravind
Sanjai Kumar
TCRβ-expressing macrophages induced by a pathogenic murine malaria correlate with parasite burden and enhanced phagocytic activity.
PLoS ONE
author_facet Miranda S Oakley
Joanna K Chorazeczewski
Maya Aleshnick
Vivek Anantharaman
Victoria Majam
Bhavna Chawla
Timothy G Myers
Qin Su
Winter A Okoth
Kazuyo Takeda
Adovi Akue
Mark KuKuruga
L Aravind
Sanjai Kumar
author_sort Miranda S Oakley
title TCRβ-expressing macrophages induced by a pathogenic murine malaria correlate with parasite burden and enhanced phagocytic activity.
title_short TCRβ-expressing macrophages induced by a pathogenic murine malaria correlate with parasite burden and enhanced phagocytic activity.
title_full TCRβ-expressing macrophages induced by a pathogenic murine malaria correlate with parasite burden and enhanced phagocytic activity.
title_fullStr TCRβ-expressing macrophages induced by a pathogenic murine malaria correlate with parasite burden and enhanced phagocytic activity.
title_full_unstemmed TCRβ-expressing macrophages induced by a pathogenic murine malaria correlate with parasite burden and enhanced phagocytic activity.
title_sort tcrβ-expressing macrophages induced by a pathogenic murine malaria correlate with parasite burden and enhanced phagocytic activity.
publisher Public Library of Science (PLoS)
series PLoS ONE
issn 1932-6203
publishDate 2018-01-01
description Macrophages express a wide array of invariant receptors that facilitate host defense and mediate pathogenesis during pathogen invasion. We report on a novel population of CD11bhighCD14+F4/80+ macrophages that express TCRβ. This population expands dramatically during a Plasmodium berghei ANKA infection and sequesters in the brain during experimental cerebral malaria. Importantly, measurement of TCRβ transcript and protein levels in macrophages in wildtype versus nude and Rag1 knockout mice establishes that the observed expression is not a consequence of passive receptor expression due to phagocytosis or trogocytosis of peripheral T cells or nonspecific antibody staining to an Fc receptor or cross reactive epitope. We also demonstrate that TCRβ on brain sequestered macrophages undergoes productive gene rearrangements and shows preferential Vβ usage. Remarkably, there is a significant correlation in the proportion of macrophages that express TCRβ and peripheral parasitemia. In addition, presence of TCRβ on the macrophage also correlates with a significant increase (1.9 fold) in the phagocytosis of parasitized erythrocytes. By transcriptional profiling, we identify a novel set of genes and pathways that associate with TCRβ expression by the macrophage. Expansion of TCRβ-expressing macrophages points towards a convergence of the innate and adaptive immune responses where both arms of the immune system cooperate to modulate the host response to malaria and possibly other infections.
url http://europepmc.org/articles/PMC6059462?pdf=render
work_keys_str_mv AT mirandasoakley tcrbexpressingmacrophagesinducedbyapathogenicmurinemalariacorrelatewithparasiteburdenandenhancedphagocyticactivity
AT joannakchorazeczewski tcrbexpressingmacrophagesinducedbyapathogenicmurinemalariacorrelatewithparasiteburdenandenhancedphagocyticactivity
AT mayaaleshnick tcrbexpressingmacrophagesinducedbyapathogenicmurinemalariacorrelatewithparasiteburdenandenhancedphagocyticactivity
AT vivekanantharaman tcrbexpressingmacrophagesinducedbyapathogenicmurinemalariacorrelatewithparasiteburdenandenhancedphagocyticactivity
AT victoriamajam tcrbexpressingmacrophagesinducedbyapathogenicmurinemalariacorrelatewithparasiteburdenandenhancedphagocyticactivity
AT bhavnachawla tcrbexpressingmacrophagesinducedbyapathogenicmurinemalariacorrelatewithparasiteburdenandenhancedphagocyticactivity
AT timothygmyers tcrbexpressingmacrophagesinducedbyapathogenicmurinemalariacorrelatewithparasiteburdenandenhancedphagocyticactivity
AT qinsu tcrbexpressingmacrophagesinducedbyapathogenicmurinemalariacorrelatewithparasiteburdenandenhancedphagocyticactivity
AT winteraokoth tcrbexpressingmacrophagesinducedbyapathogenicmurinemalariacorrelatewithparasiteburdenandenhancedphagocyticactivity
AT kazuyotakeda tcrbexpressingmacrophagesinducedbyapathogenicmurinemalariacorrelatewithparasiteburdenandenhancedphagocyticactivity
AT adoviakue tcrbexpressingmacrophagesinducedbyapathogenicmurinemalariacorrelatewithparasiteburdenandenhancedphagocyticactivity
AT markkukuruga tcrbexpressingmacrophagesinducedbyapathogenicmurinemalariacorrelatewithparasiteburdenandenhancedphagocyticactivity
AT laravind tcrbexpressingmacrophagesinducedbyapathogenicmurinemalariacorrelatewithparasiteburdenandenhancedphagocyticactivity
AT sanjaikumar tcrbexpressingmacrophagesinducedbyapathogenicmurinemalariacorrelatewithparasiteburdenandenhancedphagocyticactivity
_version_ 1724918442903994368