Acidic preconditioning induced intracellular acid adaptation to protect renal injury via dynamic phosphorylation of focal adhesion kinase-dependent activation of sodium hydrogen exchanger 1.

IF 8.2 2区 生物学 Q1 CELL BIOLOGY Cell Communication and Signaling Pub Date : 2024-08-08 DOI:10.1186/s12964-024-01773-w
Annan Chen, Jian Zhang, Zhixin Yan, Yufei Lu, Weize Chen, Yingxue Sun, Qiuyu Gu, Fang Li, Yan Yang, Shanfang Qiu, Xueping Lin, Dong Zhang, Jie Teng, Yi Fang, Bo Shen, Nana Song, Xiaoqiang Ding
{"title":"Acidic preconditioning induced intracellular acid adaptation to protect renal injury via dynamic phosphorylation of focal adhesion kinase-dependent activation of sodium hydrogen exchanger 1.","authors":"Annan Chen, Jian Zhang, Zhixin Yan, Yufei Lu, Weize Chen, Yingxue Sun, Qiuyu Gu, Fang Li, Yan Yang, Shanfang Qiu, Xueping Lin, Dong Zhang, Jie Teng, Yi Fang, Bo Shen, Nana Song, Xiaoqiang Ding","doi":"10.1186/s12964-024-01773-w","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Disruptions in intracellular pH (pH<sub>i</sub>) homeostasis, causing deviations from the physiological range, can damage renal epithelial cells. However, the existence of an adaptive mechanism to restore pH<sub>i</sub> to normalcy remains unclear. Early research identified H<sup>+</sup> as a critical mediator of ischemic preconditioning (IPC), leading to the concept of acidic preconditioning (AP). This concept proposes that short-term, repetitive acidic stimulation can enhance a cell's capacity to withstand subsequent adverse stress. While AP has demonstrated protective effects in various ischemia-reperfusion (I/R) injury models, its application in kidney injury remains largely unexplored.</p><p><strong>Methods: </strong>An AP model was established in human kidney (HK2) cells by treating them with an acidic medium for 12 h, followed by a recovery period with a normal medium for 6 h. To induce hypoxia/reoxygenation (H/R) injury, HK2 cells were subjected to hypoxia for 24 h and reoxygenation for 1 h. In vivo, a mouse model of IPC was established by clamping the bilateral renal pedicles for 15 min, followed by reperfusion for 4 days. Conversely, the I/R model involved clamping the bilateral renal pedicles for 35 min and reperfusion for 24 h. Western blotting was employed to evaluate the expression levels of cleaved caspase 3, cleaved caspase 9, NHE1, KIM1, FAK, and NOX4. A pH-sensitive fluorescent probe was used to measure pH<sub>i</sub>, while a Hemin/CNF microelectrode monitored kidney tissue pH. Immunofluorescence staining was performed to visualize the localization of NHE1, NOX4, and FAK, along with the actin cytoskeleton structure in HK2 cells. Cell adhesion and scratch assays were conducted to assess cell motility.</p><p><strong>Results: </strong>Our findings demonstrated that AP could effectively mitigate H/R injury in HK2 cells. This protective effect and the maintenance of pH<sub>i</sub> homeostasis by AP involved the upregulation of Na<sup>+</sup>/H<sup>+</sup> exchanger 1 (NHE1) expression and activity. The activity of NHE1 was regulated by dynamic changes in pH<sub>i</sub>-dependent phosphorylation of Focal Adhesion Kinase (FAK) at Y397. This process was associated with NOX4-mediated reactive oxygen species (ROS) production. Furthermore, AP induced the co-localization of FAK, NOX4, and NHE1 in focal adhesions, promoting cytoskeletal remodeling and enhancing cell adhesion and migration capabilities.</p><p><strong>Conclusions: </strong>This study provides compelling evidence that AP maintains pH<sub>i</sub> homeostasis and promotes cytoskeletal remodeling through FAK/NOX4/NHE1 signaling. This signaling pathway ultimately contributes to alleviated H/R injury in HK2 cells.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":null,"pages":null},"PeriodicalIF":8.2000,"publicationDate":"2024-08-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11308338/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cell Communication and Signaling","FirstCategoryId":"99","ListUrlMain":"https://doi.org/10.1186/s12964-024-01773-w","RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"CELL BIOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Background: Disruptions in intracellular pH (pHi) homeostasis, causing deviations from the physiological range, can damage renal epithelial cells. However, the existence of an adaptive mechanism to restore pHi to normalcy remains unclear. Early research identified H+ as a critical mediator of ischemic preconditioning (IPC), leading to the concept of acidic preconditioning (AP). This concept proposes that short-term, repetitive acidic stimulation can enhance a cell's capacity to withstand subsequent adverse stress. While AP has demonstrated protective effects in various ischemia-reperfusion (I/R) injury models, its application in kidney injury remains largely unexplored.

Methods: An AP model was established in human kidney (HK2) cells by treating them with an acidic medium for 12 h, followed by a recovery period with a normal medium for 6 h. To induce hypoxia/reoxygenation (H/R) injury, HK2 cells were subjected to hypoxia for 24 h and reoxygenation for 1 h. In vivo, a mouse model of IPC was established by clamping the bilateral renal pedicles for 15 min, followed by reperfusion for 4 days. Conversely, the I/R model involved clamping the bilateral renal pedicles for 35 min and reperfusion for 24 h. Western blotting was employed to evaluate the expression levels of cleaved caspase 3, cleaved caspase 9, NHE1, KIM1, FAK, and NOX4. A pH-sensitive fluorescent probe was used to measure pHi, while a Hemin/CNF microelectrode monitored kidney tissue pH. Immunofluorescence staining was performed to visualize the localization of NHE1, NOX4, and FAK, along with the actin cytoskeleton structure in HK2 cells. Cell adhesion and scratch assays were conducted to assess cell motility.

Results: Our findings demonstrated that AP could effectively mitigate H/R injury in HK2 cells. This protective effect and the maintenance of pHi homeostasis by AP involved the upregulation of Na+/H+ exchanger 1 (NHE1) expression and activity. The activity of NHE1 was regulated by dynamic changes in pHi-dependent phosphorylation of Focal Adhesion Kinase (FAK) at Y397. This process was associated with NOX4-mediated reactive oxygen species (ROS) production. Furthermore, AP induced the co-localization of FAK, NOX4, and NHE1 in focal adhesions, promoting cytoskeletal remodeling and enhancing cell adhesion and migration capabilities.

Conclusions: This study provides compelling evidence that AP maintains pHi homeostasis and promotes cytoskeletal remodeling through FAK/NOX4/NHE1 signaling. This signaling pathway ultimately contributes to alleviated H/R injury in HK2 cells.

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
酸性预处理通过局灶粘附激酶依赖性激活钠氢交换子1的动态磷酸化诱导细胞内酸适应以保护肾损伤。
背景:细胞内 pH 值(pHi)平衡的破坏会导致偏离生理范围,从而损害肾上皮细胞。然而,将 pHi 恢复正常的适应机制是否存在仍不清楚。早期的研究发现 H+ 是缺血预处理(IPC)的关键介质,从而提出了酸性预处理(AP)的概念。这一概念认为,短期、重复的酸性刺激可增强细胞抵御后续不利应激的能力。虽然酸性预处理在各种缺血再灌注(I/R)损伤模型中显示出保护作用,但其在肾损伤中的应用在很大程度上仍未得到探索:为了诱导缺氧/再氧合(H/R)损伤,HK2细胞要经历24小时的缺氧和1小时的再氧合。在体内,通过夹闭双侧肾蒂15分钟,然后再灌注4天,建立了小鼠IPC模型。Western印迹技术用于评估裂解的caspase 3、裂解的caspase 9、NHE1、KIM1、FAK和NOX4的表达水平。pH 敏感荧光探针用于测量 pHi,而 Hemin/CNF 微电极则用于监测肾组织 pH。免疫荧光染色可观察到 HK2 细胞中 NHE1、NOX4 和 FAK 的定位以及肌动蛋白细胞骨架结构。还进行了细胞粘附和划痕试验以评估细胞的运动性:结果:我们的研究结果表明,AP 能有效减轻 HK2 细胞的 H/R 损伤。结果:我们的研究结果表明,AP 能有效缓解 HK2 细胞的 H/R 损伤,AP 的这种保护作用和维持 pHi 平衡的作用涉及 Na+/H+ 交换子 1(NHE1)表达和活性的上调。NHE1的活性受pHi依赖的焦点粘附激酶(FAK)Y397磷酸化的动态变化调控。这一过程与 NOX4 介导的活性氧(ROS)产生有关。此外,AP诱导FAK、NOX4和NHE1在病灶粘附处共定位,促进细胞骨架重塑,增强细胞粘附和迁移能力:本研究提供了令人信服的证据,证明 AP 可通过 FAK/NOX4/NHE1 信号维持 pHi 平衡并促进细胞骨架重塑。该信号通路最终有助于减轻 HK2 细胞的 H/R 损伤。
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 去求助
来源期刊
CiteScore
11.00
自引率
0.00%
发文量
180
期刊介绍: Cell Communication and Signaling (CCS) is a peer-reviewed, open-access scientific journal that focuses on cellular signaling pathways in both normal and pathological conditions. It publishes original research, reviews, and commentaries, welcoming studies that utilize molecular, morphological, biochemical, structural, and cell biology approaches. CCS also encourages interdisciplinary work and innovative models, including in silico, in vitro, and in vivo approaches, to facilitate investigations of cell signaling pathways, networks, and behavior. Starting from January 2019, CCS is proud to announce its affiliation with the International Cell Death Society. The journal now encourages submissions covering all aspects of cell death, including apoptotic and non-apoptotic mechanisms, cell death in model systems, autophagy, clearance of dying cells, and the immunological and pathological consequences of dying cells in the tissue microenvironment.
期刊最新文献
ONC212, alone or in synergistic conjunction with Navitoclax (ABT-263), promotes cancer cell apoptosis via unconventional mitochondrial-independent caspase-3 activation Bivalent chromatin accommodates survivin and BRG1/SWI complex to activate DNA damage response in CD4+ cells Correction: Acidic preconditioning induced intracellular acid adaptation to protect renal injury via dynamic phosphorylation of focal adhesion kinase-dependent activation of sodium hydrogen exchanger 1 A novel amphiphilic squalene-based compound with open-chain polyethers reduces malignant melanoma metastasis in-vitro and in-vivo TMEM100 acts as a TAK1 receptor that prevents pathological cardiac hypertrophy progression
×
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
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 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 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1