Progressive cellular senescence mediates renal dysfunction in ischemic nephropathy

Seo Rin Kim, Amrutesh S. Puranik, Kai Jiang, Xiaojun Chen, Xiang Yang Zhu, Ian Taylor, Alireza Khodadadi-Jamayran, Amir Lerman, La Tonya J. Hickson, Bennett G. Childs, Stephen C. Textor, Tamara Tchkonia, Timothy B. Niewold, James L. Kirkland, Lilach O. Lerman

Research output: Contribution to journalArticlepeer-review

Abstract

Background Peripheral vascular diseases may induce chronic ischemia and cellular injury distal to the arterial obstruction. Cellular senescence involves proliferation arrest in response to stress, which can damage neighboring cells. Renal artery stenosis (RAS) induces stenotic-kidney dysfunction and injury, but whether these arise from cellular senescenceand their temporal pattern remain unknown. Methods Chronic renal ischemia was induced in transgenic INK-ATTAC and wild type C57BL/6 mice by unilateral RAS, and kidney function (in vivo micro-MRI) and tissue damage were assessed. Mouse healthy and stenotic kidneys were analyzed using unbiased single-cell RNA-sequencing. To demonstrate translational relevance, cellular senescence was studied in human stenotic kidneys. Results Using intraperitoneal AP20187 injections starting 1, 2, or 4 weeks after RAS, selective clearance of cells highly expressing p16Ink4a attenuated cellular senescence and improved stenotic-kidney function; however, starting treatment immediately after RAS induction was unsuccessful. Broader clearance of senescent cells, using the oral senolytic combination dasatinib and quercetin, in C57BL/6 RAS mice was more effective in clearing cells positive for p21 (Cdkn1a) and alleviating renal dysfunction and damage. Unbiased, single-cell RNA sequencing in freshly dissociated cells from healthy and stenotic mouse kidneys identified stenotic-kidney epithelial cells undergoing both mesenchymal transition and senescence. As in mice, injured human stenotic kidneys exhibited cellular senescence, suggesting this process is conserved. Conclusions Maladaptive tubular cell senescence, involving upregulated p16 (Cdkn2a), p19 (Cdkn2d), and p21 (Cdkn1a) expression, is associated with renal dysfunction and injury in chronic ischemia. These findings support development of senolytic strategies to delay chronic ischemic renal injury.

Original languageEnglish (US)
Pages (from-to)1987-2004
Number of pages18
JournalJournal of the American Society of Nephrology
Volume32
Issue number8
DOIs
StatePublished - Aug 2021

ASJC Scopus subject areas

  • General Medicine

Fingerprint

Dive into the research topics of 'Progressive cellular senescence mediates renal dysfunction in ischemic nephropathy'. Together they form a unique fingerprint.

Cite this