Cargando…

An Fgr kinase inhibitor attenuates sepsis-associated encephalopathy by ameliorating mitochondrial dysfunction, oxidative stress, and neuroinflammation via the SIRT1/PGC-1α signaling pathway

BACKGROUND: Sepsis-associated encephalopathy (SAE) is characterized by diffuse brain dysfunction, long-term cognitive impairment, and increased morbidity and mortality. The current treatment for SAE is mainly symptomatic; the lack of specific treatment options and a poor understanding of the underly...

Descripción completa

Detalles Bibliográficos
Autores principales: Liu, Yuqiang, Yang, Han, Luo, Nanbo, Fu, Yifei, Qiu, Fang, Pan, Zhenglong, Li, Xiongjuan, Jian, Wenling, Yang, Xinping, Xue, Qingsheng, Luo, Yan, Yu, Buwei, Liu, Zhiheng
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10360347/
https://www.ncbi.nlm.nih.gov/pubmed/37475042
http://dx.doi.org/10.1186/s12967-023-04345-7
_version_ 1785076085856141312
author Liu, Yuqiang
Yang, Han
Luo, Nanbo
Fu, Yifei
Qiu, Fang
Pan, Zhenglong
Li, Xiongjuan
Jian, Wenling
Yang, Xinping
Xue, Qingsheng
Luo, Yan
Yu, Buwei
Liu, Zhiheng
author_facet Liu, Yuqiang
Yang, Han
Luo, Nanbo
Fu, Yifei
Qiu, Fang
Pan, Zhenglong
Li, Xiongjuan
Jian, Wenling
Yang, Xinping
Xue, Qingsheng
Luo, Yan
Yu, Buwei
Liu, Zhiheng
author_sort Liu, Yuqiang
collection PubMed
description BACKGROUND: Sepsis-associated encephalopathy (SAE) is characterized by diffuse brain dysfunction, long-term cognitive impairment, and increased morbidity and mortality. The current treatment for SAE is mainly symptomatic; the lack of specific treatment options and a poor understanding of the underlying mechanism of disease are responsible for poor patient outcomes. Fgr is a member of the Src family of tyrosine kinases and is involved in the innate immune response, hematologic cancer, diet-induced obesity, and hemorrhage-induced thalamic pain. This study investigated the protection provided by an Fgr kinase inhibitor in SAE and the underlying mechanism(s) of action. METHODS: A cecal ligation and puncture (CLP)-induced mouse sepsis model was established. Mice were treated with or without an Fgr inhibitor and a PGC-1α inhibitor/activator. An open field test, a novel object recognition test, and an elevated plus maze were used to assess neurobehavioral changes in the mice. Western blotting and immunofluorescence were used to measure protein expression, and mRNA levels were measured using quantitative PCR (qPCR). An enzyme-linked immunosorbent assay was performed to quantify inflammatory cytokines. Mitochondrial membrane potential and morphology were measured by JC-1, electron microscopy, and the MitoTracker Deep Red probe. Oxidative stress and mitochondrial dysfunction were analyzed. In addition, the regulatory effect of Fgr on sirtuin 1 (SIRT1) was assessed. RESULTS: CLP-induced sepsis increased the expression of Fgr in the hippocampal neurons. Pharmacological inhibition of Fgr attenuated CLP-induced neuroinflammation, the survival rate, cognitive and emotional dysfunction, oxidative stress, and mitochondrial dysfunction. Moreover, Fgr interacted with SIRT1 and reduced its activity and expression. In addition, activation of SIRT1/PGC-1α promoted the protective effects of the Fgr inhibitor on CLP-induced brain dysfunction, while inactivation of SIRT1/PGC-1α counteracted the benefits of the Fgr inhibitor. CONCLUSIONS: To our knowledge, this is the first report of Fgr kinase inhibition markedly ameliorating SAE through activation of the SIRT1/PGC-1α pathway, and this may be a promising therapeutic target for SAE. GRAPHICAL ABSTRACT: [Image: see text]
format Online
Article
Text
id pubmed-10360347
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-103603472023-07-22 An Fgr kinase inhibitor attenuates sepsis-associated encephalopathy by ameliorating mitochondrial dysfunction, oxidative stress, and neuroinflammation via the SIRT1/PGC-1α signaling pathway Liu, Yuqiang Yang, Han Luo, Nanbo Fu, Yifei Qiu, Fang Pan, Zhenglong Li, Xiongjuan Jian, Wenling Yang, Xinping Xue, Qingsheng Luo, Yan Yu, Buwei Liu, Zhiheng J Transl Med Research BACKGROUND: Sepsis-associated encephalopathy (SAE) is characterized by diffuse brain dysfunction, long-term cognitive impairment, and increased morbidity and mortality. The current treatment for SAE is mainly symptomatic; the lack of specific treatment options and a poor understanding of the underlying mechanism of disease are responsible for poor patient outcomes. Fgr is a member of the Src family of tyrosine kinases and is involved in the innate immune response, hematologic cancer, diet-induced obesity, and hemorrhage-induced thalamic pain. This study investigated the protection provided by an Fgr kinase inhibitor in SAE and the underlying mechanism(s) of action. METHODS: A cecal ligation and puncture (CLP)-induced mouse sepsis model was established. Mice were treated with or without an Fgr inhibitor and a PGC-1α inhibitor/activator. An open field test, a novel object recognition test, and an elevated plus maze were used to assess neurobehavioral changes in the mice. Western blotting and immunofluorescence were used to measure protein expression, and mRNA levels were measured using quantitative PCR (qPCR). An enzyme-linked immunosorbent assay was performed to quantify inflammatory cytokines. Mitochondrial membrane potential and morphology were measured by JC-1, electron microscopy, and the MitoTracker Deep Red probe. Oxidative stress and mitochondrial dysfunction were analyzed. In addition, the regulatory effect of Fgr on sirtuin 1 (SIRT1) was assessed. RESULTS: CLP-induced sepsis increased the expression of Fgr in the hippocampal neurons. Pharmacological inhibition of Fgr attenuated CLP-induced neuroinflammation, the survival rate, cognitive and emotional dysfunction, oxidative stress, and mitochondrial dysfunction. Moreover, Fgr interacted with SIRT1 and reduced its activity and expression. In addition, activation of SIRT1/PGC-1α promoted the protective effects of the Fgr inhibitor on CLP-induced brain dysfunction, while inactivation of SIRT1/PGC-1α counteracted the benefits of the Fgr inhibitor. CONCLUSIONS: To our knowledge, this is the first report of Fgr kinase inhibition markedly ameliorating SAE through activation of the SIRT1/PGC-1α pathway, and this may be a promising therapeutic target for SAE. GRAPHICAL ABSTRACT: [Image: see text] BioMed Central 2023-07-20 /pmc/articles/PMC10360347/ /pubmed/37475042 http://dx.doi.org/10.1186/s12967-023-04345-7 Text en © The Author(s) 2023 https://creativecommons.org/licenses/by/4.0/Open Access This 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
Liu, Yuqiang
Yang, Han
Luo, Nanbo
Fu, Yifei
Qiu, Fang
Pan, Zhenglong
Li, Xiongjuan
Jian, Wenling
Yang, Xinping
Xue, Qingsheng
Luo, Yan
Yu, Buwei
Liu, Zhiheng
An Fgr kinase inhibitor attenuates sepsis-associated encephalopathy by ameliorating mitochondrial dysfunction, oxidative stress, and neuroinflammation via the SIRT1/PGC-1α signaling pathway
title An Fgr kinase inhibitor attenuates sepsis-associated encephalopathy by ameliorating mitochondrial dysfunction, oxidative stress, and neuroinflammation via the SIRT1/PGC-1α signaling pathway
title_full An Fgr kinase inhibitor attenuates sepsis-associated encephalopathy by ameliorating mitochondrial dysfunction, oxidative stress, and neuroinflammation via the SIRT1/PGC-1α signaling pathway
title_fullStr An Fgr kinase inhibitor attenuates sepsis-associated encephalopathy by ameliorating mitochondrial dysfunction, oxidative stress, and neuroinflammation via the SIRT1/PGC-1α signaling pathway
title_full_unstemmed An Fgr kinase inhibitor attenuates sepsis-associated encephalopathy by ameliorating mitochondrial dysfunction, oxidative stress, and neuroinflammation via the SIRT1/PGC-1α signaling pathway
title_short An Fgr kinase inhibitor attenuates sepsis-associated encephalopathy by ameliorating mitochondrial dysfunction, oxidative stress, and neuroinflammation via the SIRT1/PGC-1α signaling pathway
title_sort fgr kinase inhibitor attenuates sepsis-associated encephalopathy by ameliorating mitochondrial dysfunction, oxidative stress, and neuroinflammation via the sirt1/pgc-1α signaling pathway
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10360347/
https://www.ncbi.nlm.nih.gov/pubmed/37475042
http://dx.doi.org/10.1186/s12967-023-04345-7
work_keys_str_mv AT liuyuqiang anfgrkinaseinhibitorattenuatessepsisassociatedencephalopathybyamelioratingmitochondrialdysfunctionoxidativestressandneuroinflammationviathesirt1pgc1asignalingpathway
AT yanghan anfgrkinaseinhibitorattenuatessepsisassociatedencephalopathybyamelioratingmitochondrialdysfunctionoxidativestressandneuroinflammationviathesirt1pgc1asignalingpathway
AT luonanbo anfgrkinaseinhibitorattenuatessepsisassociatedencephalopathybyamelioratingmitochondrialdysfunctionoxidativestressandneuroinflammationviathesirt1pgc1asignalingpathway
AT fuyifei anfgrkinaseinhibitorattenuatessepsisassociatedencephalopathybyamelioratingmitochondrialdysfunctionoxidativestressandneuroinflammationviathesirt1pgc1asignalingpathway
AT qiufang anfgrkinaseinhibitorattenuatessepsisassociatedencephalopathybyamelioratingmitochondrialdysfunctionoxidativestressandneuroinflammationviathesirt1pgc1asignalingpathway
AT panzhenglong anfgrkinaseinhibitorattenuatessepsisassociatedencephalopathybyamelioratingmitochondrialdysfunctionoxidativestressandneuroinflammationviathesirt1pgc1asignalingpathway
AT lixiongjuan anfgrkinaseinhibitorattenuatessepsisassociatedencephalopathybyamelioratingmitochondrialdysfunctionoxidativestressandneuroinflammationviathesirt1pgc1asignalingpathway
AT jianwenling anfgrkinaseinhibitorattenuatessepsisassociatedencephalopathybyamelioratingmitochondrialdysfunctionoxidativestressandneuroinflammationviathesirt1pgc1asignalingpathway
AT yangxinping anfgrkinaseinhibitorattenuatessepsisassociatedencephalopathybyamelioratingmitochondrialdysfunctionoxidativestressandneuroinflammationviathesirt1pgc1asignalingpathway
AT xueqingsheng anfgrkinaseinhibitorattenuatessepsisassociatedencephalopathybyamelioratingmitochondrialdysfunctionoxidativestressandneuroinflammationviathesirt1pgc1asignalingpathway
AT luoyan anfgrkinaseinhibitorattenuatessepsisassociatedencephalopathybyamelioratingmitochondrialdysfunctionoxidativestressandneuroinflammationviathesirt1pgc1asignalingpathway
AT yubuwei anfgrkinaseinhibitorattenuatessepsisassociatedencephalopathybyamelioratingmitochondrialdysfunctionoxidativestressandneuroinflammationviathesirt1pgc1asignalingpathway
AT liuzhiheng anfgrkinaseinhibitorattenuatessepsisassociatedencephalopathybyamelioratingmitochondrialdysfunctionoxidativestressandneuroinflammationviathesirt1pgc1asignalingpathway
AT liuyuqiang fgrkinaseinhibitorattenuatessepsisassociatedencephalopathybyamelioratingmitochondrialdysfunctionoxidativestressandneuroinflammationviathesirt1pgc1asignalingpathway
AT yanghan fgrkinaseinhibitorattenuatessepsisassociatedencephalopathybyamelioratingmitochondrialdysfunctionoxidativestressandneuroinflammationviathesirt1pgc1asignalingpathway
AT luonanbo fgrkinaseinhibitorattenuatessepsisassociatedencephalopathybyamelioratingmitochondrialdysfunctionoxidativestressandneuroinflammationviathesirt1pgc1asignalingpathway
AT fuyifei fgrkinaseinhibitorattenuatessepsisassociatedencephalopathybyamelioratingmitochondrialdysfunctionoxidativestressandneuroinflammationviathesirt1pgc1asignalingpathway
AT qiufang fgrkinaseinhibitorattenuatessepsisassociatedencephalopathybyamelioratingmitochondrialdysfunctionoxidativestressandneuroinflammationviathesirt1pgc1asignalingpathway
AT panzhenglong fgrkinaseinhibitorattenuatessepsisassociatedencephalopathybyamelioratingmitochondrialdysfunctionoxidativestressandneuroinflammationviathesirt1pgc1asignalingpathway
AT lixiongjuan fgrkinaseinhibitorattenuatessepsisassociatedencephalopathybyamelioratingmitochondrialdysfunctionoxidativestressandneuroinflammationviathesirt1pgc1asignalingpathway
AT jianwenling fgrkinaseinhibitorattenuatessepsisassociatedencephalopathybyamelioratingmitochondrialdysfunctionoxidativestressandneuroinflammationviathesirt1pgc1asignalingpathway
AT yangxinping fgrkinaseinhibitorattenuatessepsisassociatedencephalopathybyamelioratingmitochondrialdysfunctionoxidativestressandneuroinflammationviathesirt1pgc1asignalingpathway
AT xueqingsheng fgrkinaseinhibitorattenuatessepsisassociatedencephalopathybyamelioratingmitochondrialdysfunctionoxidativestressandneuroinflammationviathesirt1pgc1asignalingpathway
AT luoyan fgrkinaseinhibitorattenuatessepsisassociatedencephalopathybyamelioratingmitochondrialdysfunctionoxidativestressandneuroinflammationviathesirt1pgc1asignalingpathway
AT yubuwei fgrkinaseinhibitorattenuatessepsisassociatedencephalopathybyamelioratingmitochondrialdysfunctionoxidativestressandneuroinflammationviathesirt1pgc1asignalingpathway
AT liuzhiheng fgrkinaseinhibitorattenuatessepsisassociatedencephalopathybyamelioratingmitochondrialdysfunctionoxidativestressandneuroinflammationviathesirt1pgc1asignalingpathway