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

Detalhes bibliográficos
Autor(a) principal: Vizuete, Adriana Fernanda Kuckartz
Data de Publicação: 2022
Outros Autores: Fróes, Fernanda Carolina Telles da Silva, Seady, Marina Pedra, Boeckel, Caroline Zanotto de, Bobermin, Larissa Daniele, Roginski, Ana Cristina, Wajner, Moacir, Quincozes-Santos, André, Goncalves, Carlos Alberto Saraiva
Tipo de documento: Artigo
Idioma: eng
Título da fonte: Repositório Institucional da UFRGS
Texto Completo: http://hdl.handle.net/10183/254322
Resumo: 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.
id UFRGS-2_f0c7035386382ecaf1d359ef825a28bc
oai_identifier_str oai:www.lume.ufrgs.br:10183/254322
network_acronym_str UFRGS-2
network_name_str Repositório Institucional da UFRGS
repository_id_str
spelling Vizuete, Adriana Fernanda KuckartzFróes, Fernanda Carolina Telles da SilvaSeady, Marina PedraBoeckel, Caroline Zanotto deBobermin, Larissa DanieleRoginski, Ana CristinaWajner, MoacirQuincozes-Santos, AndréGoncalves, Carlos Alberto Saraiva2023-02-07T05:02:48Z20221742-2094http://hdl.handle.net/10183/254322001158312Neuroinflammation 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.application/pdfengJournal of neuroinflammation. London. Vol. 19 (Oct. 2022), 255, 23 p.HipocampoDoenças neuroinflamatóriasGlicóliseGlucoseLipopolissacarídeosMetabolismoDoenças do sistema nervosoNeuroinflammationGlycolysisGlucose uptakePFK1S100BIL-1βEarly effects of LPS-induced neuroinflammation on the rat hippocampal glycolytic pathwayEstrangeiroinfo:eu-repo/semantics/articleinfo:eu-repo/semantics/publishedVersioninfo:eu-repo/semantics/openAccessreponame:Repositório Institucional da UFRGSinstname:Universidade Federal do Rio Grande do Sul (UFRGS)instacron:UFRGSTEXT001158312.pdf.txt001158312.pdf.txtExtracted Texttext/plain84185http://www.lume.ufrgs.br/bitstream/10183/254322/2/001158312.pdf.txte3a2e5b49be8c64a28743e9153d42c35MD52ORIGINAL001158312.pdfTexto completo (inglês)application/pdf3598326http://www.lume.ufrgs.br/bitstream/10183/254322/1/001158312.pdf6e6fc4563141720fdbb717e5dcce91aaMD5110183/2543222023-02-08 06:03:49.137731oai:www.lume.ufrgs.br:10183/254322Repositório de PublicaçõesPUBhttps://lume.ufrgs.br/oai/requestopendoar:2023-02-08T08:03:49Repositório Institucional da UFRGS - Universidade Federal do Rio Grande do Sul (UFRGS)false
dc.title.pt_BR.fl_str_mv 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
spellingShingle Early effects of LPS-induced neuroinflammation on the rat hippocampal glycolytic pathway
Vizuete, Adriana Fernanda Kuckartz
Hipocampo
Doenças neuroinflamatórias
Glicólise
Glucose
Lipopolissacarídeos
Metabolismo
Doenças do sistema nervoso
Neuroinflammation
Glycolysis
Glucose uptake
PFK1
S100B
IL-1β
title_short 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_sort Early effects of LPS-induced neuroinflammation on the rat hippocampal glycolytic pathway
author Vizuete, Adriana Fernanda Kuckartz
author_facet Vizuete, Adriana Fernanda Kuckartz
Fróes, Fernanda Carolina Telles da Silva
Seady, Marina Pedra
Boeckel, Caroline Zanotto de
Bobermin, Larissa Daniele
Roginski, Ana Cristina
Wajner, Moacir
Quincozes-Santos, André
Goncalves, Carlos Alberto Saraiva
author_role author
author2 Fróes, Fernanda Carolina Telles da Silva
Seady, Marina Pedra
Boeckel, Caroline Zanotto de
Bobermin, Larissa Daniele
Roginski, Ana Cristina
Wajner, Moacir
Quincozes-Santos, André
Goncalves, Carlos Alberto Saraiva
author2_role author
author
author
author
author
author
author
author
dc.contributor.author.fl_str_mv Vizuete, Adriana Fernanda Kuckartz
Fróes, Fernanda Carolina Telles da Silva
Seady, Marina Pedra
Boeckel, Caroline Zanotto de
Bobermin, Larissa Daniele
Roginski, Ana Cristina
Wajner, Moacir
Quincozes-Santos, André
Goncalves, Carlos Alberto Saraiva
dc.subject.por.fl_str_mv Hipocampo
Doenças neuroinflamatórias
Glicólise
Glucose
Lipopolissacarídeos
Metabolismo
Doenças do sistema nervoso
topic Hipocampo
Doenças neuroinflamatórias
Glicólise
Glucose
Lipopolissacarídeos
Metabolismo
Doenças do sistema nervoso
Neuroinflammation
Glycolysis
Glucose uptake
PFK1
S100B
IL-1β
dc.subject.eng.fl_str_mv Neuroinflammation
Glycolysis
Glucose uptake
PFK1
S100B
IL-1β
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.
publishDate 2022
dc.date.issued.fl_str_mv 2022
dc.date.accessioned.fl_str_mv 2023-02-07T05:02:48Z
dc.type.driver.fl_str_mv Estrangeiro
info:eu-repo/semantics/article
dc.type.status.fl_str_mv info:eu-repo/semantics/publishedVersion
format article
status_str publishedVersion
dc.identifier.uri.fl_str_mv http://hdl.handle.net/10183/254322
dc.identifier.issn.pt_BR.fl_str_mv 1742-2094
dc.identifier.nrb.pt_BR.fl_str_mv 001158312
identifier_str_mv 1742-2094
001158312
url http://hdl.handle.net/10183/254322
dc.language.iso.fl_str_mv eng
language eng
dc.relation.ispartof.pt_BR.fl_str_mv Journal of neuroinflammation. London. Vol. 19 (Oct. 2022), 255, 23 p.
dc.rights.driver.fl_str_mv info:eu-repo/semantics/openAccess
eu_rights_str_mv openAccess
dc.format.none.fl_str_mv application/pdf
dc.source.none.fl_str_mv reponame:Repositório Institucional da UFRGS
instname:Universidade Federal do Rio Grande do Sul (UFRGS)
instacron:UFRGS
instname_str Universidade Federal do Rio Grande do Sul (UFRGS)
instacron_str UFRGS
institution UFRGS
reponame_str Repositório Institucional da UFRGS
collection Repositório Institucional da UFRGS
bitstream.url.fl_str_mv http://www.lume.ufrgs.br/bitstream/10183/254322/2/001158312.pdf.txt
http://www.lume.ufrgs.br/bitstream/10183/254322/1/001158312.pdf
bitstream.checksum.fl_str_mv e3a2e5b49be8c64a28743e9153d42c35
6e6fc4563141720fdbb717e5dcce91aa
bitstream.checksumAlgorithm.fl_str_mv MD5
MD5
repository.name.fl_str_mv Repositório Institucional da UFRGS - Universidade Federal do Rio Grande do Sul (UFRGS)
repository.mail.fl_str_mv
_version_ 1801225080160649216