Cargando…

Early effects of LPS-induced neuroinflammation on the rat hippocampal glycolytic pathway

Neuroinflammation is a common feature during the development of neurological disorders and neurodegenerative diseases, where glial cells, such as microglia and astrocytes, play key roles in the activation and maintenance of inflammatory responses in the central nervous system. Neuroinflammation is n...

Descripción completa

Detalles Bibliográficos
Autores principales: Vizuete, Adriana Fernanda K., Fróes, Fernanda, Seady, Marina, Zanotto, Caroline, Bobermin, Larissa Daniele, Roginski, Ana Cristina, Wajner, Moacir, Quincozes-Santos, André, Gonçalves, Carlos Alberto
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9552490/
https://www.ncbi.nlm.nih.gov/pubmed/36221097
http://dx.doi.org/10.1186/s12974-022-02612-w
_version_ 1784806262168354816
author Vizuete, Adriana Fernanda K.
Fróes, Fernanda
Seady, Marina
Zanotto, Caroline
Bobermin, Larissa Daniele
Roginski, Ana Cristina
Wajner, Moacir
Quincozes-Santos, André
Gonçalves, Carlos Alberto
author_facet Vizuete, Adriana Fernanda K.
Fróes, Fernanda
Seady, Marina
Zanotto, Caroline
Bobermin, Larissa Daniele
Roginski, Ana Cristina
Wajner, Moacir
Quincozes-Santos, André
Gonçalves, Carlos Alberto
author_sort Vizuete, Adriana Fernanda K.
collection PubMed
description Neuroinflammation is a common feature during the development of neurological disorders and neurodegenerative diseases, where glial cells, such as microglia and astrocytes, play key roles in the activation and maintenance of inflammatory responses in the central nervous system. Neuroinflammation is now known to involve a neurometabolic shift, in addition to an increase in energy consumption. We used two approaches (in vivo and ex vivo) to evaluate the effects of lipopolysaccharide (LPS)-induced neuroinflammation on neurometabolic reprogramming, and on the modulation of the glycolytic pathway during the neuroinflammatory response. For this, we investigated inflammatory cytokines and receptors in the rat hippocampus, as well as markers of glial reactivity. Mitochondrial respirometry and the glycolytic pathway were evaluated by multiple parameters, including enzymatic activity, gene expression and regulation by protein kinases. Metabolic (e.g., metformin, 3PO, oxamic acid, fluorocitrate) and inflammatory (e.g., minocycline, MCC950, arundic acid) inhibitors were used in ex vivo hippocampal slices. The induction of early inflammatory changes by LPS (both in vivo and ex vivo) enhanced glycolytic parameters, such as glucose uptake, PFK1 activity and lactate release. This increased glucose consumption was independent of the energy expenditure for glutamate uptake, which was in fact diverted for the maintenance of the immune response. Accordingly, inhibitors of the glycolytic pathway and Krebs cycle reverted neuroinflammation (reducing IL-1β and S100B) and the changes in glycolytic parameters induced by LPS in acute hippocampal slices. Moreover, the inhibition of S100B, a protein predominantly synthesized and secreted by astrocytes, inhibition of microglia activation and abrogation of NLRP3 inflammasome assembly confirmed the role of neuroinflammation in the upregulation of glycolysis in the hippocampus. Our data indicate a neurometabolic glycolytic shift, induced by inflammatory activation, as well as a central and integrative role of astrocytes, and suggest that interference in the control of neurometabolism may be a promising strategy for downregulating neuroinflammation and consequently for diminishing negative neurological outcomes. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12974-022-02612-w.
format Online
Article
Text
id pubmed-9552490
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-95524902022-10-12 Early effects of LPS-induced neuroinflammation on the rat hippocampal glycolytic pathway Vizuete, Adriana Fernanda K. Fróes, Fernanda Seady, Marina Zanotto, Caroline Bobermin, Larissa Daniele Roginski, Ana Cristina Wajner, Moacir Quincozes-Santos, André Gonçalves, Carlos Alberto J Neuroinflammation Research Neuroinflammation is a common feature during the development of neurological disorders and neurodegenerative diseases, where glial cells, such as microglia and astrocytes, play key roles in the activation and maintenance of inflammatory responses in the central nervous system. Neuroinflammation is now known to involve a neurometabolic shift, in addition to an increase in energy consumption. We used two approaches (in vivo and ex vivo) to evaluate the effects of lipopolysaccharide (LPS)-induced neuroinflammation on neurometabolic reprogramming, and on the modulation of the glycolytic pathway during the neuroinflammatory response. For this, we investigated inflammatory cytokines and receptors in the rat hippocampus, as well as markers of glial reactivity. Mitochondrial respirometry and the glycolytic pathway were evaluated by multiple parameters, including enzymatic activity, gene expression and regulation by protein kinases. Metabolic (e.g., metformin, 3PO, oxamic acid, fluorocitrate) and inflammatory (e.g., minocycline, MCC950, arundic acid) inhibitors were used in ex vivo hippocampal slices. The induction of early inflammatory changes by LPS (both in vivo and ex vivo) enhanced glycolytic parameters, such as glucose uptake, PFK1 activity and lactate release. This increased glucose consumption was independent of the energy expenditure for glutamate uptake, which was in fact diverted for the maintenance of the immune response. Accordingly, inhibitors of the glycolytic pathway and Krebs cycle reverted neuroinflammation (reducing IL-1β and S100B) and the changes in glycolytic parameters induced by LPS in acute hippocampal slices. Moreover, the inhibition of S100B, a protein predominantly synthesized and secreted by astrocytes, inhibition of microglia activation and abrogation of NLRP3 inflammasome assembly confirmed the role of neuroinflammation in the upregulation of glycolysis in the hippocampus. Our data indicate a neurometabolic glycolytic shift, induced by inflammatory activation, as well as a central and integrative role of astrocytes, and suggest that interference in the control of neurometabolism may be a promising strategy for downregulating neuroinflammation and consequently for diminishing negative neurological outcomes. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12974-022-02612-w. BioMed Central 2022-10-11 /pmc/articles/PMC9552490/ /pubmed/36221097 http://dx.doi.org/10.1186/s12974-022-02612-w 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
Vizuete, Adriana Fernanda K.
Fróes, Fernanda
Seady, Marina
Zanotto, Caroline
Bobermin, Larissa Daniele
Roginski, Ana Cristina
Wajner, Moacir
Quincozes-Santos, André
Gonçalves, Carlos Alberto
Early effects of LPS-induced neuroinflammation on the rat hippocampal glycolytic pathway
title Early effects of LPS-induced neuroinflammation on the rat hippocampal glycolytic pathway
title_full Early effects of LPS-induced neuroinflammation on the rat hippocampal glycolytic pathway
title_fullStr Early effects of LPS-induced neuroinflammation on the rat hippocampal glycolytic pathway
title_full_unstemmed Early effects of LPS-induced neuroinflammation on the rat hippocampal glycolytic pathway
title_short Early effects of LPS-induced neuroinflammation on the rat hippocampal glycolytic pathway
title_sort early effects of lps-induced neuroinflammation on the rat hippocampal glycolytic pathway
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9552490/
https://www.ncbi.nlm.nih.gov/pubmed/36221097
http://dx.doi.org/10.1186/s12974-022-02612-w
work_keys_str_mv AT vizueteadrianafernandak earlyeffectsoflpsinducedneuroinflammationontherathippocampalglycolyticpathway
AT froesfernanda earlyeffectsoflpsinducedneuroinflammationontherathippocampalglycolyticpathway
AT seadymarina earlyeffectsoflpsinducedneuroinflammationontherathippocampalglycolyticpathway
AT zanottocaroline earlyeffectsoflpsinducedneuroinflammationontherathippocampalglycolyticpathway
AT boberminlarissadaniele earlyeffectsoflpsinducedneuroinflammationontherathippocampalglycolyticpathway
AT roginskianacristina earlyeffectsoflpsinducedneuroinflammationontherathippocampalglycolyticpathway
AT wajnermoacir earlyeffectsoflpsinducedneuroinflammationontherathippocampalglycolyticpathway
AT quincozessantosandre earlyeffectsoflpsinducedneuroinflammationontherathippocampalglycolyticpathway
AT goncalvescarlosalberto earlyeffectsoflpsinducedneuroinflammationontherathippocampalglycolyticpathway