Cargando…

Dimethyl fumarate reduces TNF and Plasmodium falciparum induced brain endothelium activation in vitro

BACKGROUND: Cerebral malaria (CM) is associated with morbidity and mortality despite the use of potent anti-malarial agents. Brain endothelial cell activation and dysfunction from oxidative and inflammatory host responses and products released by Plasmodium falciparum-infected erythrocytes (IE), are...

Descripción completa

Detalles Bibliográficos
Autores principales: Mita-Mendoza, Neida K., Magallon-Tejada, Ariel, Parmar, Priyanka, Furtado, Raquel, Aldrich, Margaret, Saidi, Alex, Taylor, Terrie, Smith, Joe, Seydel, Karl, Daily, Johanna P.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7579885/
https://www.ncbi.nlm.nih.gov/pubmed/33087130
http://dx.doi.org/10.1186/s12936-020-03447-7
_version_ 1783598683656290304
author Mita-Mendoza, Neida K.
Magallon-Tejada, Ariel
Parmar, Priyanka
Furtado, Raquel
Aldrich, Margaret
Saidi, Alex
Taylor, Terrie
Smith, Joe
Seydel, Karl
Daily, Johanna P.
author_facet Mita-Mendoza, Neida K.
Magallon-Tejada, Ariel
Parmar, Priyanka
Furtado, Raquel
Aldrich, Margaret
Saidi, Alex
Taylor, Terrie
Smith, Joe
Seydel, Karl
Daily, Johanna P.
author_sort Mita-Mendoza, Neida K.
collection PubMed
description BACKGROUND: Cerebral malaria (CM) is associated with morbidity and mortality despite the use of potent anti-malarial agents. Brain endothelial cell activation and dysfunction from oxidative and inflammatory host responses and products released by Plasmodium falciparum-infected erythrocytes (IE), are likely the major contributors to the encephalopathy, seizures, and brain swelling that are associated with CM. The development of adjunctive therapy to reduce the pathological consequences of host response pathways could improve outcomes. A potentially protective role of the nuclear factor E2-related factor 2 (NRF2) pathway, which serves as a therapeutic target in brain microvascular diseases and central nervous system (CNS) inflammatory diseases such as multiple sclerosis was tested to protect endothelial cells in an in vitro culture system subjected to tumour necrosis factor (TNF) or infected red blood cell exposure. NRF2 is a transcription factor that mediates anti-oxidant and anti-inflammatory responses. METHODS: To accurately reflect clinically relevant parasite biology a unique panel of parasite isolates derived from patients with stringently defined CM was developed. The effect of TNF and these parasite lines on primary human brain microvascular endothelial cell (HBMVEC) activation in an in vitro co-culture model was tested. HBMVEC activation was measured by cellular release of IL6 and nuclear translocation of NFκB. The transcriptional and functional effects of dimethyl fumarate (DMF), an FDA approved drug which induces the NRF2 pathway, on host and parasite induced HBMVEC activation was characterized. In addition, the effect of DMF on parasite binding to TNF stimulated HBMVEC in a semi-static binding assay was examined. RESULTS: Transcriptional profiling demonstrates that DMF upregulates the NRF2-Mediated Oxidative Stress Response, ErbB4 Signaling Pathway, Peroxisome Proliferator-activated Receptor (PPAR) Signaling and downregulates iNOS Signaling and the Neuroinflammation Signaling Pathway on TNF activated HBMVEC. The parasite lines derived from eight paediatric CM patients demonstrated increased binding to TNF activated HBMVEC and varied in their binding and activation of HBMVEC. Overall DMF reduced both TNF and CM derived parasite activation of HBMVEC. CONCLUSIONS: These findings provide evidence that targeting the NRF2 pathway in TNF and parasite activated HBMVEC mediates multiple protective pathways and may represent a novel adjunctive therapy to improve infection outcomes in CM.
format Online
Article
Text
id pubmed-7579885
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-75798852020-10-22 Dimethyl fumarate reduces TNF and Plasmodium falciparum induced brain endothelium activation in vitro Mita-Mendoza, Neida K. Magallon-Tejada, Ariel Parmar, Priyanka Furtado, Raquel Aldrich, Margaret Saidi, Alex Taylor, Terrie Smith, Joe Seydel, Karl Daily, Johanna P. Malar J Research BACKGROUND: Cerebral malaria (CM) is associated with morbidity and mortality despite the use of potent anti-malarial agents. Brain endothelial cell activation and dysfunction from oxidative and inflammatory host responses and products released by Plasmodium falciparum-infected erythrocytes (IE), are likely the major contributors to the encephalopathy, seizures, and brain swelling that are associated with CM. The development of adjunctive therapy to reduce the pathological consequences of host response pathways could improve outcomes. A potentially protective role of the nuclear factor E2-related factor 2 (NRF2) pathway, which serves as a therapeutic target in brain microvascular diseases and central nervous system (CNS) inflammatory diseases such as multiple sclerosis was tested to protect endothelial cells in an in vitro culture system subjected to tumour necrosis factor (TNF) or infected red blood cell exposure. NRF2 is a transcription factor that mediates anti-oxidant and anti-inflammatory responses. METHODS: To accurately reflect clinically relevant parasite biology a unique panel of parasite isolates derived from patients with stringently defined CM was developed. The effect of TNF and these parasite lines on primary human brain microvascular endothelial cell (HBMVEC) activation in an in vitro co-culture model was tested. HBMVEC activation was measured by cellular release of IL6 and nuclear translocation of NFκB. The transcriptional and functional effects of dimethyl fumarate (DMF), an FDA approved drug which induces the NRF2 pathway, on host and parasite induced HBMVEC activation was characterized. In addition, the effect of DMF on parasite binding to TNF stimulated HBMVEC in a semi-static binding assay was examined. RESULTS: Transcriptional profiling demonstrates that DMF upregulates the NRF2-Mediated Oxidative Stress Response, ErbB4 Signaling Pathway, Peroxisome Proliferator-activated Receptor (PPAR) Signaling and downregulates iNOS Signaling and the Neuroinflammation Signaling Pathway on TNF activated HBMVEC. The parasite lines derived from eight paediatric CM patients demonstrated increased binding to TNF activated HBMVEC and varied in their binding and activation of HBMVEC. Overall DMF reduced both TNF and CM derived parasite activation of HBMVEC. CONCLUSIONS: These findings provide evidence that targeting the NRF2 pathway in TNF and parasite activated HBMVEC mediates multiple protective pathways and may represent a novel adjunctive therapy to improve infection outcomes in CM. BioMed Central 2020-10-21 /pmc/articles/PMC7579885/ /pubmed/33087130 http://dx.doi.org/10.1186/s12936-020-03447-7 Text en © The Author(s) 2020 Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Mita-Mendoza, Neida K.
Magallon-Tejada, Ariel
Parmar, Priyanka
Furtado, Raquel
Aldrich, Margaret
Saidi, Alex
Taylor, Terrie
Smith, Joe
Seydel, Karl
Daily, Johanna P.
Dimethyl fumarate reduces TNF and Plasmodium falciparum induced brain endothelium activation in vitro
title Dimethyl fumarate reduces TNF and Plasmodium falciparum induced brain endothelium activation in vitro
title_full Dimethyl fumarate reduces TNF and Plasmodium falciparum induced brain endothelium activation in vitro
title_fullStr Dimethyl fumarate reduces TNF and Plasmodium falciparum induced brain endothelium activation in vitro
title_full_unstemmed Dimethyl fumarate reduces TNF and Plasmodium falciparum induced brain endothelium activation in vitro
title_short Dimethyl fumarate reduces TNF and Plasmodium falciparum induced brain endothelium activation in vitro
title_sort dimethyl fumarate reduces tnf and plasmodium falciparum induced brain endothelium activation in vitro
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7579885/
https://www.ncbi.nlm.nih.gov/pubmed/33087130
http://dx.doi.org/10.1186/s12936-020-03447-7
work_keys_str_mv AT mitamendozaneidak dimethylfumaratereducestnfandplasmodiumfalciparuminducedbrainendotheliumactivationinvitro
AT magallontejadaariel dimethylfumaratereducestnfandplasmodiumfalciparuminducedbrainendotheliumactivationinvitro
AT parmarpriyanka dimethylfumaratereducestnfandplasmodiumfalciparuminducedbrainendotheliumactivationinvitro
AT furtadoraquel dimethylfumaratereducestnfandplasmodiumfalciparuminducedbrainendotheliumactivationinvitro
AT aldrichmargaret dimethylfumaratereducestnfandplasmodiumfalciparuminducedbrainendotheliumactivationinvitro
AT saidialex dimethylfumaratereducestnfandplasmodiumfalciparuminducedbrainendotheliumactivationinvitro
AT taylorterrie dimethylfumaratereducestnfandplasmodiumfalciparuminducedbrainendotheliumactivationinvitro
AT smithjoe dimethylfumaratereducestnfandplasmodiumfalciparuminducedbrainendotheliumactivationinvitro
AT seydelkarl dimethylfumaratereducestnfandplasmodiumfalciparuminducedbrainendotheliumactivationinvitro
AT dailyjohannap dimethylfumaratereducestnfandplasmodiumfalciparuminducedbrainendotheliumactivationinvitro