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

Disruptions in intracellular pH (pH ) homeostasis, causing deviations from the physiological range, can damage renal epithelial cells. However, the existence of an adaptive mechanism to restore pH to normalcy remains unclear. Early research identified H as a critical mediator of ischemic preconditio...

Full description

Saved in:
Bibliographic Details
Published inCell communication and signaling Vol. 22; no. 1; pp. 393 - 21
Main Authors Chen, Annan, Zhang, Jian, Yan, Zhixin, Lu, Yufei, Chen, Weize, Sun, Yingxue, Gu, Qiuyu, Li, Fang, Yang, Yan, Qiu, Shanfang, Lin, Xueping, Zhang, Dong, Teng, Jie, Fang, Yi, Shen, Bo, Song, Nana, Ding, Xiaoqiang
Format Journal Article
LanguageEnglish
Published England BioMed Central Ltd 08.08.2024
BioMed Central
BMC
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Disruptions in intracellular pH (pH ) homeostasis, causing deviations from the physiological range, can damage renal epithelial cells. However, the existence of an adaptive mechanism to restore pH 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. 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 , 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. Our findings demonstrated that AP could effectively mitigate H/R injury in HK2 cells. This protective effect and the maintenance of pH 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 pH -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. This study provides compelling evidence that AP maintains pH homeostasis and promotes cytoskeletal remodeling through FAK/NOX4/NHE1 signaling. This signaling pathway ultimately contributes to alleviated H/R injury in HK2 cells.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
ISSN:1478-811X
1478-811X
DOI:10.1186/s12964-024-01773-w