Role of the CDKL1-SOX11 signaling axis in acute kidney injury.

Josie A Silvaroli, Gabriela V Martinez, Thitinee Vanichapol, Alan J Davidson, Diana Zepeda-Orozco, Navjot S Pabla, Ji Young Kim
{"title":"Role of the CDKL1-SOX11 signaling axis in acute kidney injury.","authors":"Josie A Silvaroli, Gabriela V Martinez, Thitinee Vanichapol, Alan J Davidson, Diana Zepeda-Orozco, Navjot S Pabla, Ji Young Kim","doi":"10.1152/ajprenal.00147.2024","DOIUrl":null,"url":null,"abstract":"<p><p>The biology of the cyclin-dependent kinase-like (CDKL) kinase family remains enigmatic. Contrary to their nomenclature, CDKLs do not rely on cyclins for activation and are not involved in cell cycle regulation. Instead, they share structural similarities with mitogen-activated protein kinases and glycogen synthase kinase-3, although their specific functions and associated signaling pathways are still unknown. Previous studies have shown that the activation of CDKL5 kinase contributes to the development of acute kidney injury (AKI) by suppressing the protective SOX9-dependent transcriptional program in tubular epithelial cells. In the current study, we measured the functional activity of all five CDKL kinases and discovered that, in addition to CDKL5, CDKL1 is also activated in tubular epithelial cells during AKI. To explore the role of CDKL1, we generated a germline knockout mouse that exhibited no abnormalities under normal conditions. Notably, when these mice were challenged with bilateral ischemia-reperfusion and rhabdomyolysis, they were found to be protected from AKI. Further mechanistic investigations revealed that CDKL1 phosphorylates and destabilizes SOX11, contributing to tubular dysfunction. In summary, this study has unveiled a previously unknown CDKL1-SOX11 axis that drives tubular dysfunction during AKI.<b>NEW & NOTEWORTHY</b> Identifying and targeting pathogenic protein kinases holds potential for drug discovery in treating acute kidney injury. Our study, using novel germline knockout mice, revealed that Cdkl1 kinase deficiency does not affect mouse viability but provides protection against acute kidney injury. This underscores the importance of Cdkl1 kinase in kidney injury and supports the development of targeted small-molecule inhibitors as potential therapeutics.</p>","PeriodicalId":93867,"journal":{"name":"American journal of physiology. Renal physiology","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2024-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"American journal of physiology. Renal physiology","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1152/ajprenal.00147.2024","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2024/7/11 0:00:00","PubModel":"Epub","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

Abstract

The biology of the cyclin-dependent kinase-like (CDKL) kinase family remains enigmatic. Contrary to their nomenclature, CDKLs do not rely on cyclins for activation and are not involved in cell cycle regulation. Instead, they share structural similarities with mitogen-activated protein kinases and glycogen synthase kinase-3, although their specific functions and associated signaling pathways are still unknown. Previous studies have shown that the activation of CDKL5 kinase contributes to the development of acute kidney injury (AKI) by suppressing the protective SOX9-dependent transcriptional program in tubular epithelial cells. In the current study, we measured the functional activity of all five CDKL kinases and discovered that, in addition to CDKL5, CDKL1 is also activated in tubular epithelial cells during AKI. To explore the role of CDKL1, we generated a germline knockout mouse that exhibited no abnormalities under normal conditions. Notably, when these mice were challenged with bilateral ischemia-reperfusion and rhabdomyolysis, they were found to be protected from AKI. Further mechanistic investigations revealed that CDKL1 phosphorylates and destabilizes SOX11, contributing to tubular dysfunction. In summary, this study has unveiled a previously unknown CDKL1-SOX11 axis that drives tubular dysfunction during AKI.NEW & NOTEWORTHY Identifying and targeting pathogenic protein kinases holds potential for drug discovery in treating acute kidney injury. Our study, using novel germline knockout mice, revealed that Cdkl1 kinase deficiency does not affect mouse viability but provides protection against acute kidney injury. This underscores the importance of Cdkl1 kinase in kidney injury and supports the development of targeted small-molecule inhibitors as potential therapeutics.

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
CDKL1-SOX11 信号轴在急性肾损伤中的作用
CDKL(类似细胞周期蛋白依赖性激酶)激酶家族的生物学特性仍是一个谜。与它们的命名相反,CDKL 并不依赖于细胞周期蛋白进行激活,也不参与细胞周期调控。相反,它们在结构上与 MAPKs(丝裂原活化蛋白激酶)和 GSK3(糖原合酶激酶 3)相似,但其具体功能和相关信号通路仍然未知。先前的研究表明,CDKL5 激酶的激活会抑制肾小管上皮细胞中依赖于 SOX9 的保护性转录程序,从而导致急性肾损伤(AKI)的发生。在本研究中,我们测量了所有五种 CDKL 激酶的功能活性,发现除了 CDKL5 外,CDKL1 也会在 AKI 期间激活肾小管上皮细胞。为了探索 CDKL1 的作用,我们产生了一种种系基因敲除小鼠,这种小鼠在正常情况下没有任何异常。值得注意的是,当这些小鼠受到双侧缺血再灌注和横纹肌溶解的挑战时,发现它们对 AKI 具有保护作用。进一步的机理研究发现,CDKL1 磷酸化并破坏 SOX11 的稳定性,导致肾小管功能障碍。总之,这些研究揭示了之前未知的 CDKL1-SOX11 轴,它在 AKI 期间驱动肾小管功能障碍。
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
自引率
0.00%
发文量
0
期刊最新文献
Chronic central nervous system leptin administration attenuates kidney dysfunction and injury in a model of ischemia/reperfusion-induced acute kidney injury. Deletion of AT1a receptors selectively in the proximal tubules alters the hypotensive and natriuretic response to ANP via NPRA/cGMP/NO Signaling. Matrix metalloproteinases in kidney homeostasis and diseases: an update. Optimized protocol for the multi-omics processing of cryopreserved human kidney tissue. Bayesian mapping of protein kinases to vasopressin-regulated phosphorylation sites in renal collecting duct.
×
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