Autophagic digestion of Leishmania major by host macrophages is associated with differential expression of BNIP3, CTSE, and the miRNAs miR-101c, miR-129, and miR-210

Abstract Background Autophagy participates in innate immunity by eliminating intracellular pathogens. Consequently, numerous microorganisms have developed strategies to impair the autophagic machinery in phagocytes. In the current study, interactions between Leishmania major (L. m.) and the autophag...

Full description

Bibliographic Details
Main Authors: Benjamin Frank, Ana Marcu, Antonio Luis de Oliveira Almeida Petersen, Heike Weber, Christian Stigloher, Jeremy C. Mottram, Claus Juergen Scholz, Uta Schurigt
Format: Article
Language:English
Published: BMC 2015-07-01
Series:Parasites & Vectors
Subjects:
Online Access:https://doi.org/10.1186/s13071-015-0974-3
_version_ 1797811835704967168
author Benjamin Frank
Ana Marcu
Antonio Luis de Oliveira Almeida Petersen
Heike Weber
Christian Stigloher
Jeremy C. Mottram
Claus Juergen Scholz
Uta Schurigt
author_facet Benjamin Frank
Ana Marcu
Antonio Luis de Oliveira Almeida Petersen
Heike Weber
Christian Stigloher
Jeremy C. Mottram
Claus Juergen Scholz
Uta Schurigt
author_sort Benjamin Frank
collection DOAJ
description Abstract Background Autophagy participates in innate immunity by eliminating intracellular pathogens. Consequently, numerous microorganisms have developed strategies to impair the autophagic machinery in phagocytes. In the current study, interactions between Leishmania major (L. m.) and the autophagic machinery of bone marrow-derived macrophages (BMDM) were analyzed. Methods BMDM were generated from BALB/c mice, and the cells were infected with L. m. promastigotes. Transmission electron microscopy (TEM) and electron tomography were used to investigate the ultrastructure of BMDM and the intracellular parasites. Affymetrix® chip analyses were conducted to identify autophagy-related messenger RNAs (mRNAs) and microRNAs (miRNAs). The protein expression levels of autophagy related 5 (ATG5), BCL2/adenovirus E1B 19 kDa protein-interacting protein 3 (BNIP3), cathepsin E (CTSE), mechanistic target of rapamycin (MTOR), microtubule-associated proteins 1A/1B light chain 3B (LC3B), and ubiquitin (UB) were investigated through western blot analyses. BMDM were transfected with specific small interfering RNAs (siRNAs) against autophagy-related genes and with mimics or inhibitors of autophagy-associated miRNAs. The infection rates of BMDM were determined by light microscopy after a parasite-specific staining. Results The experiments demonstrated autophagy induction in BMDM after in vitro infection with L. m.. The results suggested a putative MTOR phosphorylation-dependent counteracting mechanism in the early infection phase and indicated that intracellular amastigotes were cleared by autophagy in BMDM in the late infection phase. Transcriptomic analyses and specific downregulation of protein expression with siRNAs suggested there is an association between the infection-specific over expression of BNIP3, as well as CTSE, and the autophagic activity of BMDM. Transfection with mimics of mmu-miR-101c and mmu-miR-129-5p, as well as with an inhibitor of mmu-miR-210-5p, demonstrated direct effects of the respective miRNAs on parasite clearance in L. m.-infected BMDM. Furthermore, Affymetrix® chip analyses revealed a complex autophagy-related RNA network consisting of differentially expressed mRNAs and miRNAs in BMDM, which indicates high glycolytic and inflammatory activity in the host macrophages. Conclusions Autophagy in L. m.-infected host macrophages is a highly regulated cellular process at both the RNA level and the protein level. Autophagy has the potential to clear parasites from the host. The results obtained from experiments with murine host macrophages could be translated in the future to develop innovative and therapeutic antileishmanial strategies for human patients.
first_indexed 2024-03-13T07:29:40Z
format Article
id doaj.art-a6a6490b278f427c83cfd4c1e143bda7
institution Directory Open Access Journal
issn 1756-3305
language English
last_indexed 2024-03-13T07:29:40Z
publishDate 2015-07-01
publisher BMC
record_format Article
series Parasites & Vectors
spelling doaj.art-a6a6490b278f427c83cfd4c1e143bda72023-06-04T11:11:39ZengBMCParasites & Vectors1756-33052015-07-018113110.1186/s13071-015-0974-3Autophagic digestion of Leishmania major by host macrophages is associated with differential expression of BNIP3, CTSE, and the miRNAs miR-101c, miR-129, and miR-210Benjamin Frank0Ana Marcu1Antonio Luis de Oliveira Almeida Petersen2Heike Weber3Christian Stigloher4Jeremy C. Mottram5Claus Juergen Scholz6Uta Schurigt7Institute for Molecular Infection Biology, University of WuerzburgInstitute for Molecular Infection Biology, University of WuerzburgWellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of GlasgowInterdisciplinary Center for Clinical Research (IZKF), University of WuerzburgDivision of Electron Microscopy, Biocenter of the University of WuerzburgWellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of GlasgowInterdisciplinary Center for Clinical Research (IZKF), University of WuerzburgInstitute for Molecular Infection Biology, University of WuerzburgAbstract Background Autophagy participates in innate immunity by eliminating intracellular pathogens. Consequently, numerous microorganisms have developed strategies to impair the autophagic machinery in phagocytes. In the current study, interactions between Leishmania major (L. m.) and the autophagic machinery of bone marrow-derived macrophages (BMDM) were analyzed. Methods BMDM were generated from BALB/c mice, and the cells were infected with L. m. promastigotes. Transmission electron microscopy (TEM) and electron tomography were used to investigate the ultrastructure of BMDM and the intracellular parasites. Affymetrix® chip analyses were conducted to identify autophagy-related messenger RNAs (mRNAs) and microRNAs (miRNAs). The protein expression levels of autophagy related 5 (ATG5), BCL2/adenovirus E1B 19 kDa protein-interacting protein 3 (BNIP3), cathepsin E (CTSE), mechanistic target of rapamycin (MTOR), microtubule-associated proteins 1A/1B light chain 3B (LC3B), and ubiquitin (UB) were investigated through western blot analyses. BMDM were transfected with specific small interfering RNAs (siRNAs) against autophagy-related genes and with mimics or inhibitors of autophagy-associated miRNAs. The infection rates of BMDM were determined by light microscopy after a parasite-specific staining. Results The experiments demonstrated autophagy induction in BMDM after in vitro infection with L. m.. The results suggested a putative MTOR phosphorylation-dependent counteracting mechanism in the early infection phase and indicated that intracellular amastigotes were cleared by autophagy in BMDM in the late infection phase. Transcriptomic analyses and specific downregulation of protein expression with siRNAs suggested there is an association between the infection-specific over expression of BNIP3, as well as CTSE, and the autophagic activity of BMDM. Transfection with mimics of mmu-miR-101c and mmu-miR-129-5p, as well as with an inhibitor of mmu-miR-210-5p, demonstrated direct effects of the respective miRNAs on parasite clearance in L. m.-infected BMDM. Furthermore, Affymetrix® chip analyses revealed a complex autophagy-related RNA network consisting of differentially expressed mRNAs and miRNAs in BMDM, which indicates high glycolytic and inflammatory activity in the host macrophages. Conclusions Autophagy in L. m.-infected host macrophages is a highly regulated cellular process at both the RNA level and the protein level. Autophagy has the potential to clear parasites from the host. The results obtained from experiments with murine host macrophages could be translated in the future to develop innovative and therapeutic antileishmanial strategies for human patients.https://doi.org/10.1186/s13071-015-0974-3AutophagyBNIP3CTSEElectron tomographyLeishmania majorMacrophages
spellingShingle Benjamin Frank
Ana Marcu
Antonio Luis de Oliveira Almeida Petersen
Heike Weber
Christian Stigloher
Jeremy C. Mottram
Claus Juergen Scholz
Uta Schurigt
Autophagic digestion of Leishmania major by host macrophages is associated with differential expression of BNIP3, CTSE, and the miRNAs miR-101c, miR-129, and miR-210
Parasites & Vectors
Autophagy
BNIP3
CTSE
Electron tomography
Leishmania major
Macrophages
title Autophagic digestion of Leishmania major by host macrophages is associated with differential expression of BNIP3, CTSE, and the miRNAs miR-101c, miR-129, and miR-210
title_full Autophagic digestion of Leishmania major by host macrophages is associated with differential expression of BNIP3, CTSE, and the miRNAs miR-101c, miR-129, and miR-210
title_fullStr Autophagic digestion of Leishmania major by host macrophages is associated with differential expression of BNIP3, CTSE, and the miRNAs miR-101c, miR-129, and miR-210
title_full_unstemmed Autophagic digestion of Leishmania major by host macrophages is associated with differential expression of BNIP3, CTSE, and the miRNAs miR-101c, miR-129, and miR-210
title_short Autophagic digestion of Leishmania major by host macrophages is associated with differential expression of BNIP3, CTSE, and the miRNAs miR-101c, miR-129, and miR-210
title_sort autophagic digestion of leishmania major by host macrophages is associated with differential expression of bnip3 ctse and the mirnas mir 101c mir 129 and mir 210
topic Autophagy
BNIP3
CTSE
Electron tomography
Leishmania major
Macrophages
url https://doi.org/10.1186/s13071-015-0974-3
work_keys_str_mv AT benjaminfrank autophagicdigestionofleishmaniamajorbyhostmacrophagesisassociatedwithdifferentialexpressionofbnip3ctseandthemirnasmir101cmir129andmir210
AT anamarcu autophagicdigestionofleishmaniamajorbyhostmacrophagesisassociatedwithdifferentialexpressionofbnip3ctseandthemirnasmir101cmir129andmir210
AT antonioluisdeoliveiraalmeidapetersen autophagicdigestionofleishmaniamajorbyhostmacrophagesisassociatedwithdifferentialexpressionofbnip3ctseandthemirnasmir101cmir129andmir210
AT heikeweber autophagicdigestionofleishmaniamajorbyhostmacrophagesisassociatedwithdifferentialexpressionofbnip3ctseandthemirnasmir101cmir129andmir210
AT christianstigloher autophagicdigestionofleishmaniamajorbyhostmacrophagesisassociatedwithdifferentialexpressionofbnip3ctseandthemirnasmir101cmir129andmir210
AT jeremycmottram autophagicdigestionofleishmaniamajorbyhostmacrophagesisassociatedwithdifferentialexpressionofbnip3ctseandthemirnasmir101cmir129andmir210
AT clausjuergenscholz autophagicdigestionofleishmaniamajorbyhostmacrophagesisassociatedwithdifferentialexpressionofbnip3ctseandthemirnasmir101cmir129andmir210
AT utaschurigt autophagicdigestionofleishmaniamajorbyhostmacrophagesisassociatedwithdifferentialexpressionofbnip3ctseandthemirnasmir101cmir129andmir210