α-Aminobutyric Acid Constrains Macrophage-Associated Inflammatory Diseases through Metabolic Reprogramming and Epigenetic Modification

Metabolites play critical roles in macrophage polarization and in their function in response to infection and inflammation. α-aminobutyric acid (AABA), a non-proteinogenic amino acid which can be generated from methionine, threonine, serine, and glycine, has not been studied extensively in relation...

Full description

Bibliographic Details
Main Authors: Fei Li, Yuting Xia, Shijie Yuan, Xiaorong Xie, Lin Li, Yuan Luo, Qiuyang Du, Yuqi Yuan, Ran He
Format: Article
Language:English
Published: MDPI AG 2023-06-01
Series:International Journal of Molecular Sciences
Subjects:
Online Access:https://www.mdpi.com/1422-0067/24/13/10444
_version_ 1797591623346946048
author Fei Li
Yuting Xia
Shijie Yuan
Xiaorong Xie
Lin Li
Yuan Luo
Qiuyang Du
Yuqi Yuan
Ran He
author_facet Fei Li
Yuting Xia
Shijie Yuan
Xiaorong Xie
Lin Li
Yuan Luo
Qiuyang Du
Yuqi Yuan
Ran He
author_sort Fei Li
collection DOAJ
description Metabolites play critical roles in macrophage polarization and in their function in response to infection and inflammation. α-aminobutyric acid (AABA), a non-proteinogenic amino acid which can be generated from methionine, threonine, serine, and glycine, has not been studied extensively in relation to macrophage polarization and function. In this study, we aimed to investigate the immunomodulatory function of AABA in regulating M1 macrophage polarization and function in vitro and in vivo. We stimulated bone-marrow-derived macrophages with lipopolysaccharide (LPS) to generate M1 macrophages. Subsequently, we induced sepsis and colitis in mice, followed by treatment with AABA. We then analyzed the samples using ELISA, real-time PCR, Western blotting, flow cytometry, and histopathological analysis to evaluate cytokine secretion, inflammatory gene expression, macrophage activation, disease progression, and inflammation severity. Additionally, metabolomic and chromatin immunoprecipitation-qPCR were conducted to investigate the function of AABA on metabolic reprogramming and epigenetic modifications of M1 macrophages. Our results revealed that AABA inhibited M1 macrophage polarization and function, which led to prolonged survival in septic mice and reduced disease severity in colitis mice. Mechanically, AABA promoted oxidative phosphorylation (OXPHOS) and glutamine and arginine metabolism while inhibiting glycolysis. Moreover, AABA could increase the occupancy of trimethylation of histone H3K27 at the promoter regions of M1 macrophage-associated inflammatory genes, which contributed to the inhibition of M1 macrophage polarization. These findings suggest that AABA may have therapeutic potential for inflammatory diseases by regulating macrophage polarization and function through metabolic and epigenetic pathways.
first_indexed 2024-03-11T01:39:56Z
format Article
id doaj.art-520679895232449fada3cea47a0f7e5a
institution Directory Open Access Journal
issn 1661-6596
1422-0067
language English
last_indexed 2024-03-11T01:39:56Z
publishDate 2023-06-01
publisher MDPI AG
record_format Article
series International Journal of Molecular Sciences
spelling doaj.art-520679895232449fada3cea47a0f7e5a2023-11-18T16:38:20ZengMDPI AGInternational Journal of Molecular Sciences1661-65961422-00672023-06-0124131044410.3390/ijms241310444α-Aminobutyric Acid Constrains Macrophage-Associated Inflammatory Diseases through Metabolic Reprogramming and Epigenetic ModificationFei Li0Yuting Xia1Shijie Yuan2Xiaorong Xie3Lin Li4Yuan Luo5Qiuyang Du6Yuqi Yuan7Ran He8Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430032, ChinaDepartment of Dermatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430032, ChinaDepartment of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430032, ChinaDepartment of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430032, ChinaState Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, ChinaDepartment of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430032, ChinaDepartment of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430032, ChinaDepartment of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430032, ChinaDepartment of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430032, ChinaMetabolites play critical roles in macrophage polarization and in their function in response to infection and inflammation. α-aminobutyric acid (AABA), a non-proteinogenic amino acid which can be generated from methionine, threonine, serine, and glycine, has not been studied extensively in relation to macrophage polarization and function. In this study, we aimed to investigate the immunomodulatory function of AABA in regulating M1 macrophage polarization and function in vitro and in vivo. We stimulated bone-marrow-derived macrophages with lipopolysaccharide (LPS) to generate M1 macrophages. Subsequently, we induced sepsis and colitis in mice, followed by treatment with AABA. We then analyzed the samples using ELISA, real-time PCR, Western blotting, flow cytometry, and histopathological analysis to evaluate cytokine secretion, inflammatory gene expression, macrophage activation, disease progression, and inflammation severity. Additionally, metabolomic and chromatin immunoprecipitation-qPCR were conducted to investigate the function of AABA on metabolic reprogramming and epigenetic modifications of M1 macrophages. Our results revealed that AABA inhibited M1 macrophage polarization and function, which led to prolonged survival in septic mice and reduced disease severity in colitis mice. Mechanically, AABA promoted oxidative phosphorylation (OXPHOS) and glutamine and arginine metabolism while inhibiting glycolysis. Moreover, AABA could increase the occupancy of trimethylation of histone H3K27 at the promoter regions of M1 macrophage-associated inflammatory genes, which contributed to the inhibition of M1 macrophage polarization. These findings suggest that AABA may have therapeutic potential for inflammatory diseases by regulating macrophage polarization and function through metabolic and epigenetic pathways.https://www.mdpi.com/1422-0067/24/13/10444macrophageinflammationα-aminobutyric acidmetabolic reprogrammingEZH2H3K27me3
spellingShingle Fei Li
Yuting Xia
Shijie Yuan
Xiaorong Xie
Lin Li
Yuan Luo
Qiuyang Du
Yuqi Yuan
Ran He
α-Aminobutyric Acid Constrains Macrophage-Associated Inflammatory Diseases through Metabolic Reprogramming and Epigenetic Modification
International Journal of Molecular Sciences
macrophage
inflammation
α-aminobutyric acid
metabolic reprogramming
EZH2
H3K27me3
title α-Aminobutyric Acid Constrains Macrophage-Associated Inflammatory Diseases through Metabolic Reprogramming and Epigenetic Modification
title_full α-Aminobutyric Acid Constrains Macrophage-Associated Inflammatory Diseases through Metabolic Reprogramming and Epigenetic Modification
title_fullStr α-Aminobutyric Acid Constrains Macrophage-Associated Inflammatory Diseases through Metabolic Reprogramming and Epigenetic Modification
title_full_unstemmed α-Aminobutyric Acid Constrains Macrophage-Associated Inflammatory Diseases through Metabolic Reprogramming and Epigenetic Modification
title_short α-Aminobutyric Acid Constrains Macrophage-Associated Inflammatory Diseases through Metabolic Reprogramming and Epigenetic Modification
title_sort α aminobutyric acid constrains macrophage associated inflammatory diseases through metabolic reprogramming and epigenetic modification
topic macrophage
inflammation
α-aminobutyric acid
metabolic reprogramming
EZH2
H3K27me3
url https://www.mdpi.com/1422-0067/24/13/10444
work_keys_str_mv AT feili aaminobutyricacidconstrainsmacrophageassociatedinflammatorydiseasesthroughmetabolicreprogrammingandepigeneticmodification
AT yutingxia aaminobutyricacidconstrainsmacrophageassociatedinflammatorydiseasesthroughmetabolicreprogrammingandepigeneticmodification
AT shijieyuan aaminobutyricacidconstrainsmacrophageassociatedinflammatorydiseasesthroughmetabolicreprogrammingandepigeneticmodification
AT xiaorongxie aaminobutyricacidconstrainsmacrophageassociatedinflammatorydiseasesthroughmetabolicreprogrammingandepigeneticmodification
AT linli aaminobutyricacidconstrainsmacrophageassociatedinflammatorydiseasesthroughmetabolicreprogrammingandepigeneticmodification
AT yuanluo aaminobutyricacidconstrainsmacrophageassociatedinflammatorydiseasesthroughmetabolicreprogrammingandepigeneticmodification
AT qiuyangdu aaminobutyricacidconstrainsmacrophageassociatedinflammatorydiseasesthroughmetabolicreprogrammingandepigeneticmodification
AT yuqiyuan aaminobutyricacidconstrainsmacrophageassociatedinflammatorydiseasesthroughmetabolicreprogrammingandepigeneticmodification
AT ranhe aaminobutyricacidconstrainsmacrophageassociatedinflammatorydiseasesthroughmetabolicreprogrammingandepigeneticmodification