Leishmania amazonensis Subverts the Transcription Factor Landscape in Dendritic Cells to Avoid Inflammasome Activation and Stall Maturation

Leishmania parasites are the causative agents of human leishmaniases. They infect professional phagocytes of their mammalian hosts, including dendritic cells (DCs) that are essential for the initiation of adaptive immune responses. These immune functions strictly depend on the DC's capacity to...

Full description

Bibliographic Details
Main Authors: Hervé Lecoeur, Thibault Rosazza, Kossiwa Kokou, Hugo Varet, Jean-Yves Coppée, Arezou Lari, Pierre-Henri Commère, Robert Weil, Guangxun Meng, Genevieve Milon, Gerald F. Späth, Eric Prina
Format: Article
Language:English
Published: Frontiers Media S.A. 2020-06-01
Series:Frontiers in Immunology
Subjects:
Online Access:https://www.frontiersin.org/article/10.3389/fimmu.2020.01098/full
id doaj-c15b5a0f1de84692ba3a78623bdbd5b2
record_format Article
collection DOAJ
language English
format Article
sources DOAJ
author Hervé Lecoeur
Hervé Lecoeur
Hervé Lecoeur
Thibault Rosazza
Thibault Rosazza
Kossiwa Kokou
Kossiwa Kokou
Kossiwa Kokou
Hugo Varet
Jean-Yves Coppée
Arezou Lari
Pierre-Henri Commère
Robert Weil
Guangxun Meng
Guangxun Meng
Genevieve Milon
Gerald F. Späth
Gerald F. Späth
Eric Prina
Eric Prina
spellingShingle Hervé Lecoeur
Hervé Lecoeur
Hervé Lecoeur
Thibault Rosazza
Thibault Rosazza
Kossiwa Kokou
Kossiwa Kokou
Kossiwa Kokou
Hugo Varet
Jean-Yves Coppée
Arezou Lari
Pierre-Henri Commère
Robert Weil
Guangxun Meng
Guangxun Meng
Genevieve Milon
Gerald F. Späth
Gerald F. Späth
Eric Prina
Eric Prina
Leishmania amazonensis Subverts the Transcription Factor Landscape in Dendritic Cells to Avoid Inflammasome Activation and Stall Maturation
Frontiers in Immunology
dendritic cell
Leishmania amazonensis
amastigote
transcription factor
NF-κB
NLRP3
author_facet Hervé Lecoeur
Hervé Lecoeur
Hervé Lecoeur
Thibault Rosazza
Thibault Rosazza
Kossiwa Kokou
Kossiwa Kokou
Kossiwa Kokou
Hugo Varet
Jean-Yves Coppée
Arezou Lari
Pierre-Henri Commère
Robert Weil
Guangxun Meng
Guangxun Meng
Genevieve Milon
Gerald F. Späth
Gerald F. Späth
Eric Prina
Eric Prina
author_sort Hervé Lecoeur
title Leishmania amazonensis Subverts the Transcription Factor Landscape in Dendritic Cells to Avoid Inflammasome Activation and Stall Maturation
title_short Leishmania amazonensis Subverts the Transcription Factor Landscape in Dendritic Cells to Avoid Inflammasome Activation and Stall Maturation
title_full Leishmania amazonensis Subverts the Transcription Factor Landscape in Dendritic Cells to Avoid Inflammasome Activation and Stall Maturation
title_fullStr Leishmania amazonensis Subverts the Transcription Factor Landscape in Dendritic Cells to Avoid Inflammasome Activation and Stall Maturation
title_full_unstemmed Leishmania amazonensis Subverts the Transcription Factor Landscape in Dendritic Cells to Avoid Inflammasome Activation and Stall Maturation
title_sort leishmania amazonensis subverts the transcription factor landscape in dendritic cells to avoid inflammasome activation and stall maturation
publisher Frontiers Media S.A.
series Frontiers in Immunology
issn 1664-3224
publishDate 2020-06-01
description Leishmania parasites are the causative agents of human leishmaniases. They infect professional phagocytes of their mammalian hosts, including dendritic cells (DCs) that are essential for the initiation of adaptive immune responses. These immune functions strictly depend on the DC's capacity to differentiate from immature, antigen-capturing cells to mature, antigen-presenting cells—a process accompanied by profound changes in cellular phenotype and expression profile. Only little is known on how intracellular Leishmania affects this important process and DC transcriptional regulation. Here, we investigate these important open questions analyzing phenotypic, cytokine profile and transcriptomic changes in murine, immature bone marrow-derived DCs (iBMDCs) infected with antibody-opsonized and non-opsonized Leishmania amazonensis (L.am) amastigotes. DCs infected by non-opsonized amastigotes remained phenotypically immature whereas those infected by opsonized parasites displayed a semi-mature phenotype. The low frequency of infected DCs in culture led us to use DsRed2-transgenic parasites allowing for the enrichment of infected BMDCs by FACS. Sorted infected DCs were then subjected to transcriptomic analyses using Affymetrix GeneChip technology. Independent of parasite opsonization, Leishmania infection induced expression of genes related to key DC processes involved in MHC Class I-restricted antigen presentation and alternative NF-κB activation. DCs infected by non-opsonized parasites maintained an immature phenotype and showed a small but significant down-regulation of gene expression related to pro-inflammatory TLR signaling, the canonical NF-kB pathway and the NLRP3 inflammasome. This transcriptomic profile was further enhanced in DCs infected with opsonized parasites that displayed a semi-mature phenotype despite absence of inflammasome activation. This paradoxical DC phenotype represents a Leishmania-specific signature, which to our knowledge has not been observed with other opsonized infectious agents. In conclusion, systems-analyses of our transcriptomics data uncovered important and previously unappreciated changes in the DC transcription factor landscape, thus revealing a novel Leishmania immune subversion strategy directly acting on transcriptional control of gene expression. Our data raise important questions on the dynamic and reciprocal interplay between trans-acting and epigenetic regulators in establishing permissive conditions for intracellular Leishmania infection and polarization of the immune response.
topic dendritic cell
Leishmania amazonensis
amastigote
transcription factor
NF-κB
NLRP3
url https://www.frontiersin.org/article/10.3389/fimmu.2020.01098/full
work_keys_str_mv AT hervelecoeur leishmaniaamazonensissubvertsthetranscriptionfactorlandscapeindendriticcellstoavoidinflammasomeactivationandstallmaturation
AT hervelecoeur leishmaniaamazonensissubvertsthetranscriptionfactorlandscapeindendriticcellstoavoidinflammasomeactivationandstallmaturation
AT hervelecoeur leishmaniaamazonensissubvertsthetranscriptionfactorlandscapeindendriticcellstoavoidinflammasomeactivationandstallmaturation
AT thibaultrosazza leishmaniaamazonensissubvertsthetranscriptionfactorlandscapeindendriticcellstoavoidinflammasomeactivationandstallmaturation
AT thibaultrosazza leishmaniaamazonensissubvertsthetranscriptionfactorlandscapeindendriticcellstoavoidinflammasomeactivationandstallmaturation
AT kossiwakokou leishmaniaamazonensissubvertsthetranscriptionfactorlandscapeindendriticcellstoavoidinflammasomeactivationandstallmaturation
AT kossiwakokou leishmaniaamazonensissubvertsthetranscriptionfactorlandscapeindendriticcellstoavoidinflammasomeactivationandstallmaturation
AT kossiwakokou leishmaniaamazonensissubvertsthetranscriptionfactorlandscapeindendriticcellstoavoidinflammasomeactivationandstallmaturation
AT hugovaret leishmaniaamazonensissubvertsthetranscriptionfactorlandscapeindendriticcellstoavoidinflammasomeactivationandstallmaturation
AT jeanyvescoppee leishmaniaamazonensissubvertsthetranscriptionfactorlandscapeindendriticcellstoavoidinflammasomeactivationandstallmaturation
AT arezoulari leishmaniaamazonensissubvertsthetranscriptionfactorlandscapeindendriticcellstoavoidinflammasomeactivationandstallmaturation
AT pierrehenricommere leishmaniaamazonensissubvertsthetranscriptionfactorlandscapeindendriticcellstoavoidinflammasomeactivationandstallmaturation
AT robertweil leishmaniaamazonensissubvertsthetranscriptionfactorlandscapeindendriticcellstoavoidinflammasomeactivationandstallmaturation
AT guangxunmeng leishmaniaamazonensissubvertsthetranscriptionfactorlandscapeindendriticcellstoavoidinflammasomeactivationandstallmaturation
AT guangxunmeng leishmaniaamazonensissubvertsthetranscriptionfactorlandscapeindendriticcellstoavoidinflammasomeactivationandstallmaturation
AT genevievemilon leishmaniaamazonensissubvertsthetranscriptionfactorlandscapeindendriticcellstoavoidinflammasomeactivationandstallmaturation
AT geraldfspath leishmaniaamazonensissubvertsthetranscriptionfactorlandscapeindendriticcellstoavoidinflammasomeactivationandstallmaturation
AT geraldfspath leishmaniaamazonensissubvertsthetranscriptionfactorlandscapeindendriticcellstoavoidinflammasomeactivationandstallmaturation
AT ericprina leishmaniaamazonensissubvertsthetranscriptionfactorlandscapeindendriticcellstoavoidinflammasomeactivationandstallmaturation
AT ericprina leishmaniaamazonensissubvertsthetranscriptionfactorlandscapeindendriticcellstoavoidinflammasomeactivationandstallmaturation
_version_ 1724734234786004992
spelling doaj-c15b5a0f1de84692ba3a78623bdbd5b22020-11-25T02:51:30ZengFrontiers Media S.A.Frontiers in Immunology1664-32242020-06-011110.3389/fimmu.2020.01098538541Leishmania amazonensis Subverts the Transcription Factor Landscape in Dendritic Cells to Avoid Inflammasome Activation and Stall MaturationHervé Lecoeur0Hervé Lecoeur1Hervé Lecoeur2Thibault Rosazza3Thibault Rosazza4Kossiwa Kokou5Kossiwa Kokou6Kossiwa Kokou7Hugo Varet8Jean-Yves Coppée9Arezou Lari10Pierre-Henri Commère11Robert Weil12Guangxun Meng13Guangxun Meng14Genevieve Milon15Gerald F. Späth16Gerald F. Späth17Eric Prina18Eric Prina19Institut Pasteur, INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Département des Parasites et Insectes Vecteurs, Paris, FrancePasteur Institute of Shanghai, Innate Immunity Unit, Key Laboratory of Molecular Virology and Immunology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, ChinaPasteur International Unit “Inflammation and Leishmania Infection”, Paris, FranceInstitut Pasteur, INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Département des Parasites et Insectes Vecteurs, Paris, FrancePasteur International Unit “Inflammation and Leishmania Infection”, Paris, FranceInstitut Pasteur, INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Département des Parasites et Insectes Vecteurs, Paris, FrancePasteur Institute of Shanghai, Innate Immunity Unit, Key Laboratory of Molecular Virology and Immunology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, ChinaPasteur International Unit “Inflammation and Leishmania Infection”, Paris, FranceHub de Bioinformatique et Biostatistique – Département Biologie Computationnelle, Institut Pasteur, USR 3756 CNRS, Paris, FranceInstitut Pasteur - Transcriptome and Epigenome Platform - Biomics Pole - C2RT, Paris, FranceSystems Biomedicine Unit, Institut Pasteur of Iran, Teheran, IranInstitut Pasteur, Plate-Forme de Cytométrie, Paris, FranceSorbonne Universités, Institut National de la Santé et de la Recherche Médicale (Inserm, UMR1135), Centre National de la Recherche Scientifique (CNRS, ERL8255), Centre d'Immunologie et des Maladies Infectieuses CIMI, Paris, FrancePasteur Institute of Shanghai, Innate Immunity Unit, Key Laboratory of Molecular Virology and Immunology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, ChinaPasteur International Unit “Inflammation and Leishmania Infection”, Paris, FranceInstitut Pasteur, Laboratoire Immunophysiologie et Parasitisme, Département des Parasites et Insectes Vecteurs, Paris, FranceInstitut Pasteur, INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Département des Parasites et Insectes Vecteurs, Paris, FrancePasteur International Unit “Inflammation and Leishmania Infection”, Paris, FranceInstitut Pasteur, INSERM U1201, Unité de Parasitologie Moléculaire et Signalisation, Département des Parasites et Insectes Vecteurs, Paris, FrancePasteur International Unit “Inflammation and Leishmania Infection”, Paris, FranceLeishmania parasites are the causative agents of human leishmaniases. They infect professional phagocytes of their mammalian hosts, including dendritic cells (DCs) that are essential for the initiation of adaptive immune responses. These immune functions strictly depend on the DC's capacity to differentiate from immature, antigen-capturing cells to mature, antigen-presenting cells—a process accompanied by profound changes in cellular phenotype and expression profile. Only little is known on how intracellular Leishmania affects this important process and DC transcriptional regulation. Here, we investigate these important open questions analyzing phenotypic, cytokine profile and transcriptomic changes in murine, immature bone marrow-derived DCs (iBMDCs) infected with antibody-opsonized and non-opsonized Leishmania amazonensis (L.am) amastigotes. DCs infected by non-opsonized amastigotes remained phenotypically immature whereas those infected by opsonized parasites displayed a semi-mature phenotype. The low frequency of infected DCs in culture led us to use DsRed2-transgenic parasites allowing for the enrichment of infected BMDCs by FACS. Sorted infected DCs were then subjected to transcriptomic analyses using Affymetrix GeneChip technology. Independent of parasite opsonization, Leishmania infection induced expression of genes related to key DC processes involved in MHC Class I-restricted antigen presentation and alternative NF-κB activation. DCs infected by non-opsonized parasites maintained an immature phenotype and showed a small but significant down-regulation of gene expression related to pro-inflammatory TLR signaling, the canonical NF-kB pathway and the NLRP3 inflammasome. This transcriptomic profile was further enhanced in DCs infected with opsonized parasites that displayed a semi-mature phenotype despite absence of inflammasome activation. This paradoxical DC phenotype represents a Leishmania-specific signature, which to our knowledge has not been observed with other opsonized infectious agents. In conclusion, systems-analyses of our transcriptomics data uncovered important and previously unappreciated changes in the DC transcription factor landscape, thus revealing a novel Leishmania immune subversion strategy directly acting on transcriptional control of gene expression. Our data raise important questions on the dynamic and reciprocal interplay between trans-acting and epigenetic regulators in establishing permissive conditions for intracellular Leishmania infection and polarization of the immune response.https://www.frontiersin.org/article/10.3389/fimmu.2020.01098/fulldendritic cellLeishmania amazonensisamastigotetranscription factorNF-κBNLRP3