NAT10-mediated ac4C acetylation of TFRC promotes sepsis-induced pulmonary injury through regulating ferroptosis

Abstract Background Sepsis-induced pulmonary injury (SPI) is a common complication of sepsis with a high rate of mortality. N4-acetylcytidine (ac 4 C) is mediated by the ac 4 C “writer”, N-acetyltransferase (NAT)10, to regulate the stabilization of mRNA. This study aimed to investigate the role of N...

Full description

Saved in:
Bibliographic Details
Published inMolecular medicine (Cambridge, Mass.) Vol. 30; no. 1; pp. 140 - 11
Main Authors Xing, Pengcheng, Zhou, Minjie, Sun, Jian, Wang, Donglian, Huang, Weipeng, An, Peng
Format Journal Article
LanguageEnglish
Published London BioMed Central 09.09.2024
BMC
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Abstract Background Sepsis-induced pulmonary injury (SPI) is a common complication of sepsis with a high rate of mortality. N4-acetylcytidine (ac 4 C) is mediated by the ac 4 C “writer”, N-acetyltransferase (NAT)10, to regulate the stabilization of mRNA. This study aimed to investigate the role of NAT10 in SPI and the underlying mechanism. Methods Twenty-three acute respiratory distress syndrome (ARDS) patients and 27 non-ARDS volunteers were recruited. A sepsis rat model was established. Reverse transcription-quantitative polymerase chain reaction was used to detect the expression of NAT10 and transferrin receptor (TFRC). Cell viability was detected by cell counting kit-8. The levels of Fe 2+ , glutathione, and malondialdehyde were assessed by commercial kits. Lipid reactive oxygen species production was measured by flow cytometric analysis. Western blot was used to detect ferroptosis-related protein levels. Haematoxylin & eosin staining was performed to observe the pulmonary pathological symptoms. Results The results showed that NAT10 was increased in ARDS patients and lipopolysaccharide-treated human lung microvascular endothelial cell line-5a (HULEC-5a) cells. NAT10 inhibition increased cell viability and decreased ferroptosis in HULEC-5a cells. TFRC was a downstream regulatory target of NAT10-mediated ac 4 C acetylation. Overexpression of TFRC decreased cell viability and promoted ferroptosis. In in vivo study, NAT10 inhibition alleviated SPI. Conclusion NAT10-mediated ac 4 C acetylation of TFRC aggravated SPI through promoting ferroptosis. Graphical Abstract
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
ISSN:1528-3658
1076-1551
1528-3658
DOI:10.1186/s10020-024-00912-w