Cargando…

MyD88-TLR4-dependent choroid plexus activation precedes perilesional inflammation and secondary brain edema in a mouse model of intracerebral hemorrhage

BACKGROUND: The functional neurological outcome of patients with intracerebral hemorrhage (ICH) strongly relates to the degree of secondary brain injury (ICH-SBI) evolving within days after the initial bleeding. Different mechanisms including the incitement of inflammatory pathways, dysfunction of t...

Descripción completa

Detalles Bibliográficos
Autores principales: Akeret, Kevin, Buzzi, Raphael M., Thomson, Bart R., Schwendinger, Nina, Klohs, Jan, Schulthess-Lutz, Nadja, Baselgia, Livio, Hansen, Kerstin, Regli, Luca, Vallelian, Florence, Hugelshofer, Michael, Schaer, Dominik J.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9730653/
https://www.ncbi.nlm.nih.gov/pubmed/36482445
http://dx.doi.org/10.1186/s12974-022-02641-5
_version_ 1784845725279977472
author Akeret, Kevin
Buzzi, Raphael M.
Thomson, Bart R.
Schwendinger, Nina
Klohs, Jan
Schulthess-Lutz, Nadja
Baselgia, Livio
Hansen, Kerstin
Regli, Luca
Vallelian, Florence
Hugelshofer, Michael
Schaer, Dominik J.
author_facet Akeret, Kevin
Buzzi, Raphael M.
Thomson, Bart R.
Schwendinger, Nina
Klohs, Jan
Schulthess-Lutz, Nadja
Baselgia, Livio
Hansen, Kerstin
Regli, Luca
Vallelian, Florence
Hugelshofer, Michael
Schaer, Dominik J.
author_sort Akeret, Kevin
collection PubMed
description BACKGROUND: The functional neurological outcome of patients with intracerebral hemorrhage (ICH) strongly relates to the degree of secondary brain injury (ICH-SBI) evolving within days after the initial bleeding. Different mechanisms including the incitement of inflammatory pathways, dysfunction of the blood–brain barrier (BBB), activation of resident microglia, and an influx of blood-borne immune cells, have been hypothesized to contribute to ICH-SBI. Yet, the spatiotemporal interplay of specific inflammatory processes within different brain compartments has not been sufficiently characterized, limiting potential therapeutic interventions to prevent and treat ICH-SBI. METHODS: We used a whole-blood injection model in mice, to systematically characterized the spatial and temporal dynamics of inflammatory processes after ICH using 7-Tesla magnetic resonance imaging (MRI), spatial RNA sequencing (spRNAseq), functional BBB assessment, and immunofluorescence average-intensity-mapping. RESULTS: We identified a pronounced early response of the choroid plexus (CP) peaking at 12–24 h that was characterized by inflammatory cytokine expression, epithelial and endothelial expression of leukocyte adhesion molecules, and the accumulation of leukocytes. In contrast, we observed a delayed secondary reaction pattern at the injection site (striatum) peaking at 96 h, defined by gene expression corresponding to perilesional leukocyte infiltration and correlating to the delayed signal alteration seen on MRI. Pathway analysis revealed a dependence of the early inflammatory reaction in the CP on toll-like receptor 4 (TLR4) signaling via myeloid differentiation factor 88 (MyD88). TLR4 and MyD88 knockout mice corroborated this observation, lacking the early upregulation of adhesion molecules and leukocyte infiltration within the CP 24 h after whole-blood injection. CONCLUSIONS: We report a biphasic brain reaction pattern after ICH with a MyD88-TLR4-dependent early inflammatory response of the CP, preceding inflammation, edema and leukocyte infiltration at the lesion site. Pharmacological targeting of the early CP activation might harbor the potential to modulate the development of ICH-SBI. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12974-022-02641-5.
format Online
Article
Text
id pubmed-9730653
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-97306532022-12-09 MyD88-TLR4-dependent choroid plexus activation precedes perilesional inflammation and secondary brain edema in a mouse model of intracerebral hemorrhage Akeret, Kevin Buzzi, Raphael M. Thomson, Bart R. Schwendinger, Nina Klohs, Jan Schulthess-Lutz, Nadja Baselgia, Livio Hansen, Kerstin Regli, Luca Vallelian, Florence Hugelshofer, Michael Schaer, Dominik J. J Neuroinflammation Research BACKGROUND: The functional neurological outcome of patients with intracerebral hemorrhage (ICH) strongly relates to the degree of secondary brain injury (ICH-SBI) evolving within days after the initial bleeding. Different mechanisms including the incitement of inflammatory pathways, dysfunction of the blood–brain barrier (BBB), activation of resident microglia, and an influx of blood-borne immune cells, have been hypothesized to contribute to ICH-SBI. Yet, the spatiotemporal interplay of specific inflammatory processes within different brain compartments has not been sufficiently characterized, limiting potential therapeutic interventions to prevent and treat ICH-SBI. METHODS: We used a whole-blood injection model in mice, to systematically characterized the spatial and temporal dynamics of inflammatory processes after ICH using 7-Tesla magnetic resonance imaging (MRI), spatial RNA sequencing (spRNAseq), functional BBB assessment, and immunofluorescence average-intensity-mapping. RESULTS: We identified a pronounced early response of the choroid plexus (CP) peaking at 12–24 h that was characterized by inflammatory cytokine expression, epithelial and endothelial expression of leukocyte adhesion molecules, and the accumulation of leukocytes. In contrast, we observed a delayed secondary reaction pattern at the injection site (striatum) peaking at 96 h, defined by gene expression corresponding to perilesional leukocyte infiltration and correlating to the delayed signal alteration seen on MRI. Pathway analysis revealed a dependence of the early inflammatory reaction in the CP on toll-like receptor 4 (TLR4) signaling via myeloid differentiation factor 88 (MyD88). TLR4 and MyD88 knockout mice corroborated this observation, lacking the early upregulation of adhesion molecules and leukocyte infiltration within the CP 24 h after whole-blood injection. CONCLUSIONS: We report a biphasic brain reaction pattern after ICH with a MyD88-TLR4-dependent early inflammatory response of the CP, preceding inflammation, edema and leukocyte infiltration at the lesion site. Pharmacological targeting of the early CP activation might harbor the potential to modulate the development of ICH-SBI. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12974-022-02641-5. BioMed Central 2022-12-08 /pmc/articles/PMC9730653/ /pubmed/36482445 http://dx.doi.org/10.1186/s12974-022-02641-5 Text en © The Author(s) 2022 https://creativecommons.org/licenses/by/4.0/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/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://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
Akeret, Kevin
Buzzi, Raphael M.
Thomson, Bart R.
Schwendinger, Nina
Klohs, Jan
Schulthess-Lutz, Nadja
Baselgia, Livio
Hansen, Kerstin
Regli, Luca
Vallelian, Florence
Hugelshofer, Michael
Schaer, Dominik J.
MyD88-TLR4-dependent choroid plexus activation precedes perilesional inflammation and secondary brain edema in a mouse model of intracerebral hemorrhage
title MyD88-TLR4-dependent choroid plexus activation precedes perilesional inflammation and secondary brain edema in a mouse model of intracerebral hemorrhage
title_full MyD88-TLR4-dependent choroid plexus activation precedes perilesional inflammation and secondary brain edema in a mouse model of intracerebral hemorrhage
title_fullStr MyD88-TLR4-dependent choroid plexus activation precedes perilesional inflammation and secondary brain edema in a mouse model of intracerebral hemorrhage
title_full_unstemmed MyD88-TLR4-dependent choroid plexus activation precedes perilesional inflammation and secondary brain edema in a mouse model of intracerebral hemorrhage
title_short MyD88-TLR4-dependent choroid plexus activation precedes perilesional inflammation and secondary brain edema in a mouse model of intracerebral hemorrhage
title_sort myd88-tlr4-dependent choroid plexus activation precedes perilesional inflammation and secondary brain edema in a mouse model of intracerebral hemorrhage
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9730653/
https://www.ncbi.nlm.nih.gov/pubmed/36482445
http://dx.doi.org/10.1186/s12974-022-02641-5
work_keys_str_mv AT akeretkevin myd88tlr4dependentchoroidplexusactivationprecedesperilesionalinflammationandsecondarybrainedemainamousemodelofintracerebralhemorrhage
AT buzziraphaelm myd88tlr4dependentchoroidplexusactivationprecedesperilesionalinflammationandsecondarybrainedemainamousemodelofintracerebralhemorrhage
AT thomsonbartr myd88tlr4dependentchoroidplexusactivationprecedesperilesionalinflammationandsecondarybrainedemainamousemodelofintracerebralhemorrhage
AT schwendingernina myd88tlr4dependentchoroidplexusactivationprecedesperilesionalinflammationandsecondarybrainedemainamousemodelofintracerebralhemorrhage
AT klohsjan myd88tlr4dependentchoroidplexusactivationprecedesperilesionalinflammationandsecondarybrainedemainamousemodelofintracerebralhemorrhage
AT schulthesslutznadja myd88tlr4dependentchoroidplexusactivationprecedesperilesionalinflammationandsecondarybrainedemainamousemodelofintracerebralhemorrhage
AT baselgialivio myd88tlr4dependentchoroidplexusactivationprecedesperilesionalinflammationandsecondarybrainedemainamousemodelofintracerebralhemorrhage
AT hansenkerstin myd88tlr4dependentchoroidplexusactivationprecedesperilesionalinflammationandsecondarybrainedemainamousemodelofintracerebralhemorrhage
AT regliluca myd88tlr4dependentchoroidplexusactivationprecedesperilesionalinflammationandsecondarybrainedemainamousemodelofintracerebralhemorrhage
AT vallelianflorence myd88tlr4dependentchoroidplexusactivationprecedesperilesionalinflammationandsecondarybrainedemainamousemodelofintracerebralhemorrhage
AT hugelshofermichael myd88tlr4dependentchoroidplexusactivationprecedesperilesionalinflammationandsecondarybrainedemainamousemodelofintracerebralhemorrhage
AT schaerdominikj myd88tlr4dependentchoroidplexusactivationprecedesperilesionalinflammationandsecondarybrainedemainamousemodelofintracerebralhemorrhage