LncRNA NEAT1 aggravates human microvascular endothelial cell injury by inhibiting the Apelin/Nrf2/HO-1 signalling pathway in type 2 diabetes mellitus with obstructive sleep apnoea

Epigenetics. 2024 Dec;19(1):2293409. doi: 10.1080/15592294.2023.2293409. Epub 2024 Jan 17.

Abstract

Long noncoding RNAs (lncRNAs) regulate the progression of type 2 diabetes mellitus complicated with obstructive sleep apnoea (T2DM-OSA). However, the role of the lncRNA nuclear paraspeckle assembly transcript 1 (NEAT1) in T2DM-OSA remains unknown. This study aimed to reveal the function of NEAT1 in T2DM-OSA and the underlying mechanism. KKAy mice were exposed to intermittent hypoxia (IH) or intermittent normoxia to generate a T2DM-OSA mouse model. HMEC-1 cells were treated with high glucose (HG) and IH to construct a T2DM-OSA cell model. RNA expression was detected by qRT-PCR. The protein expression of Apelin, NF-E2-related factor 2 (Nrf2), haem oxygenase-1 (HO-1), and up-frameshift suppressor 1 (UPF1) was assessed using western blot. Cell injury was evaluated using flow cytometry, enzyme-linked immunosorbent assay, and oxidative stress kit assays. RIP, RNA pull-down, and actinomycin D assays were performed to determine the associations between NEAT1, UPF1, and Apelin. NEAT1 expression was upregulated in the aortic vascular tissues of mice with T2DM exposed to IH and HMEC-1 cells stimulated with HG and IH, whereas Apelin expression was downregulated. The absence of NEAT1 protected HMEC-1 cells from HG- and IH-induced damage. Furthermore, NEAT1 destabilized Apelin mRNA by recruiting UPF1. Apelin overexpression decreased HG- and IH-induced injury to HMEC-1 cells by activating the Nrf2/HO-1 pathway. Moreover, NEAT1 knockdown reduced HG- and IH-induced injury to HMEC-1 cells through Apelin. NEAT1 silencing reduced HMEC-1 cell injury through the Apelin/Nrf2/HO-1 signalling pathway in T2DM-OSA.Abbreviations: LncRNAs, long non-coding RNAs; T2DM, type 2 diabetes mellitus; OSA, obstructive sleep apnoea; NEAT1, nuclear paraspeckle assembly transcript 1; IH, intermittent hypoxia; HMEC-1, human microvascular endothelial cells; HG, high glucose; Nrf2, NF-E2-related factor 2; UPF1, up-frameshift suppressor 1; HO-1, haem oxygenase-1; qRT-PCR, quantitative real-time polymerase chain reaction; ELISA, enzyme-linked immunosorbent assay; GAPDH, glyceraldehyde 3-phosphate dehydrogenase; TNF-α, tumour necrosis factor-α; CCK-8, Cell Counting Kit-8; IL-1β, interleukin-1β; ROS, reactive oxygen species; MDA, malondialdehyde; SOD, superoxide dismutase; RIP, RNA immunoprecipitation; SD, standard deviations; GSH, glutathione; AIS, acute ischaemic stroke; HMGB1, high mobility group box-1 protein; TLR4, toll-like receptor 4.

Keywords: Apelin; T2DM with OSA; inflammation; lncRNA NEAT1; oxidative stress.

MeSH terms

  • Animals
  • Apelin / genetics
  • Apelin / metabolism
  • Brain Ischemia* / complications
  • DNA Methylation
  • Diabetes Mellitus, Type 2* / complications
  • Diabetes Mellitus, Type 2* / genetics
  • Endothelial Cells / metabolism
  • Glucose
  • Heme Oxygenase (Decyclizing) / metabolism
  • Humans
  • Hypoxia
  • Mice
  • NF-E2-Related Factor 2 / genetics
  • NF-E2-Related Factor 2 / metabolism
  • RNA Helicases*
  • RNA, Long Noncoding* / genetics
  • RNA, Long Noncoding* / metabolism
  • Sleep Apnea, Obstructive* / complications
  • Sleep Apnea, Obstructive* / genetics
  • Sleep Apnea, Obstructive* / metabolism
  • Stroke* / complications
  • Trans-Activators / metabolism
  • Tumor Necrosis Factor-alpha / metabolism

Substances

  • Apelin
  • Glucose
  • Heme Oxygenase (Decyclizing)
  • NF-E2-Related Factor 2
  • RNA Helicases
  • RNA, Long Noncoding
  • Trans-Activators
  • Tumor Necrosis Factor-alpha
  • UPF1 protein, human
  • NEAT1 long non-coding RNA, human

Grants and funding

This work was supported by Regulation of long-chain noncoding RNA NEAT1 on the pathogenesis of type 2 diabetes mellitus complicated with OSA(C202303066059).