Reticulon-1A mediates diabetic kidney disease progression through endoplasmic reticulum-mitochondrial contacts in tubular epithelial cells

Recent epidemiological studies suggest that some patients with diabetes progress to kidney failure without significant albuminuria and glomerular injury, suggesting a critical role of kidney tubular epithelial cell (TEC) injury in diabetic kidney disease (DKD) progression. However, the major risk fa...

Full description

Saved in:
Bibliographic Details
Published inKidney international Vol. 102; no. 2; pp. 293 - 306
Main Authors Xie, Yifan, E, Jing, Cai, Hong, Zhong, Fang, Xiao, Wenzhen, Gordon, Ronald E., Wang, Lois, Zheng, Ya-Li, Zhang, Aihua, Lee, Kyung, He, John Cijiang
Format Journal Article
LanguageEnglish
Published United States Elsevier Inc 01.08.2022
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Recent epidemiological studies suggest that some patients with diabetes progress to kidney failure without significant albuminuria and glomerular injury, suggesting a critical role of kidney tubular epithelial cell (TEC) injury in diabetic kidney disease (DKD) progression. However, the major risk factors contributing to TEC injury and progression in DKD remain unclear. We previously showed that expression of endoplasmic reticulum-resident protein Reticulon-1A (RTN1A) increased in human DKD, and the increased RTN1A expression promoted TEC injury through endoplasmic reticulum (ER) stress response. Here, we show that TEC-specific RTN1A overexpression worsened DKD in mice, evidenced by enhanced tubular injury, tubulointerstitial fibrosis, and kidney function decline. But RTN1A overexpression did not exacerbate diabetes-induced glomerular injury or albuminuria. Notably, RTN1A overexpression worsened both ER stress and mitochondrial dysfunction in TECs under diabetic conditions by regulation of ER-mitochondria contacts. Mechanistically, ER-bound RTN1A interacted with mitochondrial hexokinase-1 and the voltage-dependent anion channel-1 (VDAC1), interfering with their association. This disengagement of VDAC1 from hexokinase-1 resulted in activation of apoptotic and inflammasome pathways, leading to TEC injury and loss. Thus, our observations highlight the importance of ER-mitochondrial crosstalk in TEC injury and the salient role of RTN1A-mediated ER-mitochondrial contact regulation in DKD progression. [Display omitted]
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
JCH and KL conceived and designed the study; YLZ and AZ assisted in the supervision of the study; YX, JE, HC, ZF, WX, and LW conducted the experiments and acquired the data; REG performed the TEM images acquisition and analysis; YX, JE, HC, ZF, WX, KL, JCH analyzed the data; JCH and KL drafted and revised the manuscript; All authors approved the final version of the manuscript.
AUTHOR CONTRIBUTIONS
These authors contributed equally to the work.
ISSN:0085-2538
1523-1755
1523-1755
DOI:10.1016/j.kint.2022.02.038