关键词: Sentrin/SUMO- specific protease 1 hypoxia-reoxygenation injury hypoxia‑inducible factor‑1α liver sinusoidal endothelial cells

Mesh : Animals Mice Capillaries / metabolism Cell Hypoxia Endothelial Cells / metabolism Hypoxia / metabolism Liver / metabolism Peptide Hydrolases Signal Transduction Vascular Endothelial Growth Factor A / metabolism

来  源:   DOI:10.3892/mmr.2024.13188   PDF(Pubmed)

Abstract:
Liver sinusoidal endothelial cells (LSECs) have an important role in hepatic ischemia‑reperfusion injury (I/R), but the specific molecular mechanism of action is unknown. LSEC proliferation is regulated and fenestration is maintained via the Sentrin/SUMO‑specific protease 1 (SENP1)/hypoxia‑inducible factor‑1α (HIF‑1α) signaling axis under hypoxic conditions. In the present study, a hypoxia‑reoxygenation (H‑R) injury model was established using mouse LSECs to explore the relationship between SENP1 and H‑R injury in vitro, and the specific underlying mechanism was identified, revealing new targets for the clinical attenuation of hepatic I/R injury. Following the culture of LSECs under H‑R conditions, it was demonstrated that the expression of SENP1 was upregulated by reverse transcription‑quantitative polymerase chain reaction and western blotting (WB). In addition, scanning electron microscopy indicated that fenestrae damage was increased, a Cell Counting Kit‑8 assay demonstrated that the proliferation of cells was impaired and flow cytometry showed that apoptosis was increased. After silencing SENP1 expression with short interfering RNA, the proliferation activity of LSECs decreased, the fenestrae damage increased, the apoptosis rate increased and the expression levels of SENP1, HIF‑1α, heme oxygenase and Bcl‑2 were downregulated (as demonstrated by WB), while the expression levels of apoptosis‑related proteins, cleaved‑caspase‑3 and Bax, were upregulated. Enzyme‑linked immunosorbent assay detection showed that the level of vascular endothelial growth factor in the supernatant decreased and the level of IL‑6 and TNF‑α increased. Following the administration of an HIF‑1α signaling pathway agonist, the situation was reversed. These results therefore suggested that SENP1 attenuated the reduction in proliferation, apoptosis and fenestration of LSECs observed following H‑R injury through the HIF‑1α signaling pathway. In conclusion, SENP1 may attenuate H‑R injury in LSECs in a HIF‑1α signaling pathway‑dependent manner.
摘要:
肝窦内皮细胞(LSEC)在肝脏缺血再灌注损伤(I/R)中具有重要作用,但具体的分子作用机制尚不清楚。在低氧条件下,通过Sentrin/SUMO特异性蛋白酶1(SENP1)/缺氧诱导因子-1α(HIF-1α)信号轴调节LSEC增殖并维持开窗。在本研究中,使用小鼠LSEC建立缺氧-复氧(H-R)损伤模型,以探索SENP1与H-R损伤之间的关系。并确定了具体的潜在机制,揭示了临床减轻肝I/R损伤的新目标。在H‑R条件下的LSEC培养之后,通过逆转录-定量聚合酶链反应和蛋白质印迹(WB)证明SENP1的表达上调。此外,扫描电镜显示窗孔损伤增加,细胞计数试剂盒-8分析显示细胞增殖受损,流式细胞术显示细胞凋亡增加。用短干扰RNA沉默SENP1表达后,LSECs的增殖活性下降,窗孔损伤增加,细胞凋亡率增加,SENP1,HIF-1α的表达水平,血红素加氧酶和Bcl-2下调(如WB所示),而凋亡相关蛋白的表达水平,cleaved‑caspase‑3和Bax,被上调了。酶联免疫吸附法检测显示,上清液中血管内皮生长因子的水平降低,IL-6和TNF-α的水平升高。在给予HIF‑1α信号通路激动剂后,形势逆转了。因此,这些结果表明SENP1减弱了增殖的减少,通过HIF‑1α信号通路观察到的H‑R损伤后LSECs的凋亡和开窗。总之,SENP1可能以HIF‑1α信号通路依赖性方式减轻LSEC中的H‑R损伤。
公众号