Cystathionine γ-lyase and hydrogen sulfide modulates glucose transporter Glut1 expression via NF-κB and PI3k/Akt in macrophages during inflammation.

Macrophages play a crucial role in inflammation, a defense mechanism of the innate immune system. Metabolic function powered by glucose transporter isoform 1 (Glut1) is necessary for macrophage activity during inflammation. The present study investigated the roles of cystathionine-γ-lyase (CSE) and...

Full description

Bibliographic Details
Main Authors: Alex Cornwell, Samantha Fedotova, Sara Cowan, Alireza Badiei
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2022-01-01
Series:PLoS ONE
Online Access:https://doi.org/10.1371/journal.pone.0278910
_version_ 1797837936692035584
author Alex Cornwell
Samantha Fedotova
Sara Cowan
Alireza Badiei
author_facet Alex Cornwell
Samantha Fedotova
Sara Cowan
Alireza Badiei
author_sort Alex Cornwell
collection DOAJ
description Macrophages play a crucial role in inflammation, a defense mechanism of the innate immune system. Metabolic function powered by glucose transporter isoform 1 (Glut1) is necessary for macrophage activity during inflammation. The present study investigated the roles of cystathionine-γ-lyase (CSE) and its byproduct, hydrogen sulfide (H2S), in macrophage glucose metabolism to explore the mechanism by which H2S acts as an inflammatory regulator in lipopolysaccharide- (LPS) induced macrophages. Our results demonstrated that LPS-treated macrophages increased Glut1 expression. LPS-induced Glut1 expression is regulated via nuclear factor (NF)-κB activation and is associated with phosphatidylinositol-3-kinase PI3k activation. Small interfering (si) RNA-mediated silencing of CSE decreased the LPS-induced NF-κB activation and Glut1 expression, suggesting a role for H2S in metabolic function in macrophages during pro-inflammatory response. Confoundingly, treatment with GYY4137, an H2S-donor molecule, also displayed inhibitory effects upon LPS-induced NF-κB activation and Glut1 expression. Moreover, GYY4137 treatment increased Akt activation, suggesting a role in promoting resolution of inflammation. Our study provides evidence that the source of H2S, either endogenous (via CSE) or exogenous (via GYY4137), supports or inhibits the LPS-induced NF-κB activity and Glut1 expression, respectively. Therefore, H2S may influence metabolic programming in immune cells to alter glucose substrate availability that impacts the immune response.
first_indexed 2024-04-09T15:32:42Z
format Article
id doaj.art-d89832833f07416bbbe2f454cc68e219
institution Directory Open Access Journal
issn 1932-6203
language English
last_indexed 2024-04-09T15:32:42Z
publishDate 2022-01-01
publisher Public Library of Science (PLoS)
record_format Article
series PLoS ONE
spelling doaj.art-d89832833f07416bbbe2f454cc68e2192023-04-28T05:32:00ZengPublic Library of Science (PLoS)PLoS ONE1932-62032022-01-011712e027891010.1371/journal.pone.0278910Cystathionine γ-lyase and hydrogen sulfide modulates glucose transporter Glut1 expression via NF-κB and PI3k/Akt in macrophages during inflammation.Alex CornwellSamantha FedotovaSara CowanAlireza BadieiMacrophages play a crucial role in inflammation, a defense mechanism of the innate immune system. Metabolic function powered by glucose transporter isoform 1 (Glut1) is necessary for macrophage activity during inflammation. The present study investigated the roles of cystathionine-γ-lyase (CSE) and its byproduct, hydrogen sulfide (H2S), in macrophage glucose metabolism to explore the mechanism by which H2S acts as an inflammatory regulator in lipopolysaccharide- (LPS) induced macrophages. Our results demonstrated that LPS-treated macrophages increased Glut1 expression. LPS-induced Glut1 expression is regulated via nuclear factor (NF)-κB activation and is associated with phosphatidylinositol-3-kinase PI3k activation. Small interfering (si) RNA-mediated silencing of CSE decreased the LPS-induced NF-κB activation and Glut1 expression, suggesting a role for H2S in metabolic function in macrophages during pro-inflammatory response. Confoundingly, treatment with GYY4137, an H2S-donor molecule, also displayed inhibitory effects upon LPS-induced NF-κB activation and Glut1 expression. Moreover, GYY4137 treatment increased Akt activation, suggesting a role in promoting resolution of inflammation. Our study provides evidence that the source of H2S, either endogenous (via CSE) or exogenous (via GYY4137), supports or inhibits the LPS-induced NF-κB activity and Glut1 expression, respectively. Therefore, H2S may influence metabolic programming in immune cells to alter glucose substrate availability that impacts the immune response.https://doi.org/10.1371/journal.pone.0278910
spellingShingle Alex Cornwell
Samantha Fedotova
Sara Cowan
Alireza Badiei
Cystathionine γ-lyase and hydrogen sulfide modulates glucose transporter Glut1 expression via NF-κB and PI3k/Akt in macrophages during inflammation.
PLoS ONE
title Cystathionine γ-lyase and hydrogen sulfide modulates glucose transporter Glut1 expression via NF-κB and PI3k/Akt in macrophages during inflammation.
title_full Cystathionine γ-lyase and hydrogen sulfide modulates glucose transporter Glut1 expression via NF-κB and PI3k/Akt in macrophages during inflammation.
title_fullStr Cystathionine γ-lyase and hydrogen sulfide modulates glucose transporter Glut1 expression via NF-κB and PI3k/Akt in macrophages during inflammation.
title_full_unstemmed Cystathionine γ-lyase and hydrogen sulfide modulates glucose transporter Glut1 expression via NF-κB and PI3k/Akt in macrophages during inflammation.
title_short Cystathionine γ-lyase and hydrogen sulfide modulates glucose transporter Glut1 expression via NF-κB and PI3k/Akt in macrophages during inflammation.
title_sort cystathionine γ lyase and hydrogen sulfide modulates glucose transporter glut1 expression via nf κb and pi3k akt in macrophages during inflammation
url https://doi.org/10.1371/journal.pone.0278910
work_keys_str_mv AT alexcornwell cystathionineglyaseandhydrogensulfidemodulatesglucosetransporterglut1expressionvianfkbandpi3kaktinmacrophagesduringinflammation
AT samanthafedotova cystathionineglyaseandhydrogensulfidemodulatesglucosetransporterglut1expressionvianfkbandpi3kaktinmacrophagesduringinflammation
AT saracowan cystathionineglyaseandhydrogensulfidemodulatesglucosetransporterglut1expressionvianfkbandpi3kaktinmacrophagesduringinflammation
AT alirezabadiei cystathionineglyaseandhydrogensulfidemodulatesglucosetransporterglut1expressionvianfkbandpi3kaktinmacrophagesduringinflammation