Deletion of TP signaling in macrophages delays liver repair following APAP-induced liver injury by reducing accumulation of reparative macrophage and production of HGF.

Mina Tanabe, Kanako Hosono, Atsushi Yamashita, Yoshiya Ito, Masataka Majima, Shuh Narumiya, Chika Kusano, Hideki Amano
{"title":"Deletion of TP signaling in macrophages delays liver repair following APAP-induced liver injury by reducing accumulation of reparative macrophage and production of HGF.","authors":"Mina Tanabe, Kanako Hosono, Atsushi Yamashita, Yoshiya Ito, Masataka Majima, Shuh Narumiya, Chika Kusano, Hideki Amano","doi":"10.1186/s41232-024-00356-z","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Acetaminophen (APAP)-induced liver injury is the most common cause of acute liver failure. Macrophages are key players in liver restoration following APAP-induced liver injury. Thromboxane A<sub>2</sub> (TXA<sub>2</sub>) and its receptor, thromboxane prostanoid (TP) receptor, have been shown to be involved in tissue repair. However, whether TP signaling plays a role in liver repair after APAP hepatotoxicity by affecting macrophage function remains unclear.</p><p><strong>Methods: </strong>Male TP knockout (TP<sup>-/-</sup>) and C57BL/6 wild-type (WT) mice were treated with APAP (300 mg/kg). In addition, macrophage-specific TP-knockout (TP<sup>△mac</sup>) and control WT mice were treated with APAP. We explored changes in liver inflammation, liver repair, and macrophage accumulation in mice treated with APAP.</p><p><strong>Results: </strong>Compared with WT mice, TP<sup>-/-</sup> mice showed aggravated liver injury as indicated by increased levels of alanine transaminase (ALT) and necrotic area as well as delayed liver repair as indicated by decreased expression of proliferating cell nuclear antigen (PCNA). Macrophage deletion exacerbated APAP-induced liver injury and impaired liver repair. Transplantation of TP-deficient bone marrow (BM) cells to WT or TP<sup>-/-</sup> mice aggravated APAP hepatotoxicity with suppressed accumulation of macrophages, while transplantation of WT-BM cells to WT or TP<sup>-/-</sup> mice attenuated APAP-induced liver injury with accumulation of macrophages in the injured regions. Macrophage-specific TP<sup>-/-</sup> mice exacerbated liver injury and delayed liver repair, which was associated with increased pro-inflammatory macrophages and decreased reparative macrophages and hepatocyte growth factor (HGF) expression. In vitro, TP signaling facilitated macrophage polarization to a reparative phenotype. Transfer of cultured BM-derived macrophages from control mice to macrophage-specific TP<sup>-/-</sup> mice attenuated APAP-induced liver injury and promoted liver repair. HGF treatment mitigated APAP-induced inflammation and promoted liver repair after APAP-induced liver injury.</p><p><strong>Conclusions: </strong>Deletion of TP signaling in macrophages delays liver repair following APAP-induced liver injury, which is associated with reduced accumulation of reparative macrophages and the hepatotrophic factor HGF. Specific activation of TP signaling in macrophages may be a potential therapeutic target for liver repair and regeneration after APAP hepatotoxicity.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2024-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11451145/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Inflammation and regeneration","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1186/s41232-024-00356-z","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

Abstract

Background: Acetaminophen (APAP)-induced liver injury is the most common cause of acute liver failure. Macrophages are key players in liver restoration following APAP-induced liver injury. Thromboxane A2 (TXA2) and its receptor, thromboxane prostanoid (TP) receptor, have been shown to be involved in tissue repair. However, whether TP signaling plays a role in liver repair after APAP hepatotoxicity by affecting macrophage function remains unclear.

Methods: Male TP knockout (TP-/-) and C57BL/6 wild-type (WT) mice were treated with APAP (300 mg/kg). In addition, macrophage-specific TP-knockout (TP△mac) and control WT mice were treated with APAP. We explored changes in liver inflammation, liver repair, and macrophage accumulation in mice treated with APAP.

Results: Compared with WT mice, TP-/- mice showed aggravated liver injury as indicated by increased levels of alanine transaminase (ALT) and necrotic area as well as delayed liver repair as indicated by decreased expression of proliferating cell nuclear antigen (PCNA). Macrophage deletion exacerbated APAP-induced liver injury and impaired liver repair. Transplantation of TP-deficient bone marrow (BM) cells to WT or TP-/- mice aggravated APAP hepatotoxicity with suppressed accumulation of macrophages, while transplantation of WT-BM cells to WT or TP-/- mice attenuated APAP-induced liver injury with accumulation of macrophages in the injured regions. Macrophage-specific TP-/- mice exacerbated liver injury and delayed liver repair, which was associated with increased pro-inflammatory macrophages and decreased reparative macrophages and hepatocyte growth factor (HGF) expression. In vitro, TP signaling facilitated macrophage polarization to a reparative phenotype. Transfer of cultured BM-derived macrophages from control mice to macrophage-specific TP-/- mice attenuated APAP-induced liver injury and promoted liver repair. HGF treatment mitigated APAP-induced inflammation and promoted liver repair after APAP-induced liver injury.

Conclusions: Deletion of TP signaling in macrophages delays liver repair following APAP-induced liver injury, which is associated with reduced accumulation of reparative macrophages and the hepatotrophic factor HGF. Specific activation of TP signaling in macrophages may be a potential therapeutic target for liver repair and regeneration after APAP hepatotoxicity.

巨噬细胞中 TP 信号的缺失会减少修复性巨噬细胞的聚集和 HGF 的产生,从而延迟 APAP 诱导的肝损伤后的肝修复。
背景:对乙酰氨基酚(APAP)引起的肝损伤是急性肝衰竭最常见的原因。巨噬细胞是 APAP 诱导肝损伤后肝脏恢复的关键角色。血栓素 A2(TXA2)及其受体--血栓素类前列腺素(TP)受体已被证明参与组织修复。然而,TP 信号是否通过影响巨噬细胞功能在 APAP 肝毒性后的肝脏修复中发挥作用仍不清楚:方法:雄性 TP 基因敲除(TP-/-)小鼠和 C57BL/6 野生型(WT)小鼠均接受 APAP(300 mg/kg)治疗。此外,巨噬细胞特异性TP基因敲除(TP△mac)小鼠和对照WT小鼠也接受了APAP治疗。我们探讨了使用 APAP 治疗的小鼠肝脏炎症、肝脏修复和巨噬细胞积累的变化:结果:与 WT 小鼠相比,TP-/- 小鼠的丙氨酸转氨酶(ALT)水平和坏死面积增加,表明肝损伤加重;增殖细胞核抗原(PCNA)表达减少,表明肝修复延迟。巨噬细胞的缺失加剧了 APAP 诱导的肝损伤并损害了肝修复。将 TP 缺失的骨髓(BM)细胞移植给 WT 或 TP-/- 小鼠会加重 APAP 的肝毒性,同时抑制巨噬细胞的聚集;而将 WT-BM 细胞移植给 WT 或 TP-/- 小鼠会减轻 APAP 诱导的肝损伤,同时在损伤区域聚集巨噬细胞。巨噬细胞特异性 TP-/- 小鼠加剧了肝损伤并延迟了肝修复,这与促炎性巨噬细胞增加、修复性巨噬细胞和肝细胞生长因子(HGF)表达减少有关。在体外,TP 信号促进巨噬细胞极化为修复表型。将培养的巨噬细胞从对照组小鼠转移到巨噬细胞特异性 TP-/- 小鼠体内,可减轻 APAP 引起的肝损伤并促进肝修复。HGF治疗减轻了APAP诱导的炎症,促进了APAP诱导的肝损伤后的肝修复:结论:巨噬细胞中 TP 信号的缺失会延迟 APAP 诱导的肝损伤后的肝修复,这与修复性巨噬细胞和肝营养因子 HGF 的积累减少有关。特异性激活巨噬细胞中的 TP 信号可能是 APAP 肝毒性后肝脏修复和再生的潜在治疗靶点。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
CiteScore
11.00
自引率
0.00%
发文量
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信