skip to main content

Neuritin promotes autophagic flux by inhibiting the cGAS-STING pathway to alleviate brain injury after subarachnoid haemorrhage

Zhang, Hao ; Ren, Kunhao ; Hu, Youjie ; Liu, Bin ; He, Yaowen ; Xu, Hui ; Ma, Ketao ; Tian, Weidong ; Dai, Linzhi ; Zhao, Dong

Brain research, 2024-08, Vol.1836, p.148909-148909, Article 148909 [Periódico revisado por pares]

Netherlands: Elsevier B.V

Texto completo disponível

Citações Citado por
  • Título:
    Neuritin promotes autophagic flux by inhibiting the cGAS-STING pathway to alleviate brain injury after subarachnoid haemorrhage
  • Autor: Zhang, Hao ; Ren, Kunhao ; Hu, Youjie ; Liu, Bin ; He, Yaowen ; Xu, Hui ; Ma, Ketao ; Tian, Weidong ; Dai, Linzhi ; Zhao, Dong
  • Assuntos: Animals ; Autophagic flux ; Autophagy - drug effects ; Autophagy - physiology ; Brain Injuries - metabolism ; cGAS-STING pathway ; Disease Models, Animal ; Early brain injury ; GPI-Linked Proteins - metabolism ; Male ; Membrane Proteins - metabolism ; Neuritin ; Neuropeptides - metabolism ; Rats ; Rats, Sprague-Dawley ; Signal Transduction - drug effects ; Signal Transduction - physiology ; Subarachnoid haemorrhage ; Subarachnoid Hemorrhage - complications ; Subarachnoid Hemorrhage - metabolism
  • É parte de: Brain research, 2024-08, Vol.1836, p.148909-148909, Article 148909
  • Notas: ObjectType-Article-1
    SourceType-Scholarly Journals-1
    ObjectType-Feature-2
    content type line 23
  • Descrição: [Display omitted] •The expression of endogenous cGAS-STING-related proteins in the brain tissue of rat was significantly increased after SAH.•Autophagic flux impairment is associated with the cGAS-STING pathway after SAH.•Neuritin restores autophagic flux by downregulating the cGAS-STING pathway. Early brain injury (EBI) is closely associated with poor prognosis in patients with subarachnoid haemorrhage (SAH), with autophagy playing a pivotal role in EBI. However, research has shown that the stimulator of interferon genes (STING) pathway impacts autophagic flux. While the regulatory impact of neuritin on EBI and autophagic flux has been established previously, the underlying mechanism remains unclear. This study aimed to determine the role of the cGAS-STING pathway in neuritin-mediated regulation of autophagic flux following SAH. A SAH model was established in male Sprague-Dawley rats via intravascular perforation. Neuritin overexpressions using adeno-associated virus, the STING antagonist “C-176,” and the activator, “CMA,” were determined to investigate the cGAS-STING pathway’s influence on autophagic flux and brain injury post-SAH, along with the neuritin's regulatory effect on STING. In this study, SAH grade, neurological score, haematoxylin and eosin (H&E) staining, brain water content (BWC), sandwich enzyme-linked immunosorbent assay, Evans blue staining, immunofluorescence staining, western blot analysis, and transmission electron microscopy (TEM) were examined. Neuritin overexpression significantly ameliorated neurobehavioural scores, blood–brain barrier injury, brain oedema, and impaired autophagic flux in SAH-induced rats. STING expression remarkably increased post-SAH. C-176 and CMA mitigated and aggravated autophagic flux injury and brain injury, respectively, while inhibiting and enhancing STING, respectively. Particularly, CMA treatment nullified the protective effects of neuritin against autophagic flux and mitigated brain injury. Neuritin alleviated EBI by restoring impaired autophagic flux after SAH through the regulation of the cGAS-STING pathway.
  • Editor: Netherlands: Elsevier B.V
  • Idioma: Inglês

Buscando em bases de dados remotas. Favor aguardar.