已入深夜,您辛苦了!由于当前在线用户较少,发布求助请尽量完整的填写文献信息,科研通机器人24小时在线,伴您度过漫漫科研夜!祝你早点完成任务,早点休息,好梦!

Persistent DNA damage underlies tubular cell polyploidization and progression to chronic kidney disease in kidneys deficient in the DNA repair protein FAN1

DNA损伤 生物 DNA修复 细胞生物学 肾脏疾病 癌症研究 DNA 遗传学 内分泌学
作者
Merlin Airik,Yu Leng Phua,Amy B. Huynh,Blake T. McCourt,Brittney M. Rush,Roderick J. Tan,Jerry Vockley,Susan Murray,A. Dorman,Peter J. Conlon,Rannar Airik
出处
期刊:Kidney International [Elsevier]
卷期号:102 (5): 1042-1056 被引量:12
标识
DOI:10.1016/j.kint.2022.07.003
摘要

Defective DNA repair pathways contribute to the development of chronic kidney disease (CKD) in humans. However, the molecular mechanisms underlying DNA damage–induced CKD pathogenesis are not well understood. Here, we investigated the role of tubular cell DNA damage in the pathogenesis of CKD using mice in which the DNA repair protein Fan1 was knocked out. The phenotype of these mice is orthologous to the human DNA damage syndrome, karyomegalic interstitial nephritis (KIN). Inactivation of Fan1 in kidney proximal tubule cells sensitized the kidneys to genotoxic and obstructive injury characterized by replication stress and persistent DNA damage response activity. Accumulation of DNA damage in Fan1 tubular cells induced epithelial dedifferentiation and tubular injury. Characteristic to KIN, cells with chronic DNA damage failed to complete mitosis and underwent polyploidization. In vitro and in vivo studies showed that polyploidization was caused by the overexpression of DNA replication factors CDT1 and CDC6 in FAN1 deficient cells. Mechanistically, inhibiting DNA replication with Roscovitine reduced tubular injury, blocked the development of KIN and mitigated kidney function in these Fan1 knockout mice. Thus, our data delineate a mechanistic pathway by which persistent DNA damage in the kidney tubular cells leads to kidney injury and development of CKD. Furthermore, therapeutic modulation of cell cycle activity may provide an opportunity to mitigate the DNA damage response induced CKD progression. Defective DNA repair pathways contribute to the development of chronic kidney disease (CKD) in humans. However, the molecular mechanisms underlying DNA damage–induced CKD pathogenesis are not well understood. Here, we investigated the role of tubular cell DNA damage in the pathogenesis of CKD using mice in which the DNA repair protein Fan1 was knocked out. The phenotype of these mice is orthologous to the human DNA damage syndrome, karyomegalic interstitial nephritis (KIN). Inactivation of Fan1 in kidney proximal tubule cells sensitized the kidneys to genotoxic and obstructive injury characterized by replication stress and persistent DNA damage response activity. Accumulation of DNA damage in Fan1 tubular cells induced epithelial dedifferentiation and tubular injury. Characteristic to KIN, cells with chronic DNA damage failed to complete mitosis and underwent polyploidization. In vitro and in vivo studies showed that polyploidization was caused by the overexpression of DNA replication factors CDT1 and CDC6 in FAN1 deficient cells. Mechanistically, inhibiting DNA replication with Roscovitine reduced tubular injury, blocked the development of KIN and mitigated kidney function in these Fan1 knockout mice. Thus, our data delineate a mechanistic pathway by which persistent DNA damage in the kidney tubular cells leads to kidney injury and development of CKD. Furthermore, therapeutic modulation of cell cycle activity may provide an opportunity to mitigate the DNA damage response induced CKD progression. Polyploid tubular cells and chronic kidney diseaseKidney InternationalVol. 102Issue 5PreviewDefective DNA repair drives chronic kidney disease (CKD), but mechanisms are unclear. Airik and colleagues use a genetic model of defective DNA repair mimicking karyomegalic nephritis, a form of CKD characterized by tubular epithelial cells (TEC) with large nuclei and tubulointerstitial nephritis. They show that DNA damage in TEC triggers endoreplication leading to polyploid TEC and CKD. Blocking endoreplication preserved kidney function, suggesting that DNA damage triggers CKD via TEC polyploidization, questioning the concept of G2/M-arrest. Full-Text PDF
最长约 10秒,即可获得该文献文件

科研通智能强力驱动
Strongly Powered by AbleSci AI
更新
大幅提高文件上传限制,最高150M (2024-4-1)

科研通是完全免费的文献互助平台,具备全网最快的应助速度,最高的求助完成率。 对每一个文献求助,科研通都将尽心尽力,给求助人一个满意的交代。
实时播报
呆呆完成签到,获得积分10
刚刚
Dlan完成签到,获得积分10
2秒前
呆呆发布了新的文献求助10
4秒前
6秒前
wanci应助释棱采纳,获得10
12秒前
liuyiduo发布了新的文献求助10
13秒前
高高的笑柳完成签到 ,获得积分10
13秒前
冷静的莞完成签到 ,获得积分10
14秒前
17秒前
细腻的雅山完成签到 ,获得积分10
18秒前
shuhaha完成签到,获得积分10
21秒前
2023完成签到,获得积分10
23秒前
24秒前
顺心孤云发布了新的文献求助10
31秒前
赘婿应助阿橘采纳,获得10
33秒前
soar完成签到 ,获得积分10
42秒前
抄手爱吃皮完成签到 ,获得积分10
47秒前
49秒前
52秒前
Lilycat完成签到 ,获得积分10
53秒前
清森完成签到 ,获得积分10
54秒前
shhs发布了新的文献求助10
54秒前
callmekar发布了新的文献求助10
55秒前
SKKK发布了新的文献求助10
56秒前
1分钟前
wbs13521完成签到,获得积分10
1分钟前
上官若男应助顺心孤云采纳,获得10
1分钟前
1分钟前
Lucas应助Forizix采纳,获得10
1分钟前
veefoo发布了新的文献求助10
1分钟前
苗条丹南发布了新的文献求助10
1分钟前
领导范儿应助callmekar采纳,获得10
1分钟前
Anthonywll完成签到 ,获得积分10
1分钟前
1分钟前
Forizix完成签到,获得积分10
1分钟前
SKKK关注了科研通微信公众号
1分钟前
Forizix发布了新的文献求助10
1分钟前
1分钟前
1分钟前
乐观尔容完成签到,获得积分10
1分钟前
高分求助中
Manual of Clinical Microbiology, 4 Volume Set (ASM Books) 13th Edition 1000
Teaching Social and Emotional Learning in Physical Education 900
The three stars each : the Astrolabes and related texts 550
Boris Pesce - Gli impiegati della Fiat dal 1955 al 1999 un percorso nella memoria 500
Chinese-English Translation Lexicon Version 3.0 500
[Lambert-Eaton syndrome without calcium channel autoantibodies] 500
少脉山油柑叶的化学成分研究 500
热门求助领域 (近24小时)
化学 材料科学 医学 生物 有机化学 工程类 生物化学 纳米技术 物理 内科学 计算机科学 化学工程 复合材料 遗传学 基因 物理化学 催化作用 电极 光电子学 量子力学
热门帖子
关注 科研通微信公众号,转发送积分 2400261
求助须知:如何正确求助?哪些是违规求助? 2100893
关于积分的说明 5296576
捐赠科研通 1828543
什么是DOI,文献DOI怎么找? 911352
版权声明 560198
科研通“疑难数据库(出版商)”最低求助积分说明 487129