1. Academic Validation
  2. Iron overload contributes to general anaesthesia-induced neurotoxicity and cognitive deficits

Iron overload contributes to general anaesthesia-induced neurotoxicity and cognitive deficits

  • J Neuroinflammation. 2020 Apr 11;17(1):110. doi: 10.1186/s12974-020-01777-6.
Jing Wu 1 Jian-Jun Yang 2 Yan Cao 1 Huihui Li 1 Hongting Zhao 1 Shuofei Yang 3 Kuanyu Li 4
Affiliations

Affiliations

  • 1 Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, 22 Hankou Road, Nanjing, 210093, China.
  • 2 Department of Anesthesiology, The first Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China.
  • 3 Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China. [email protected].
  • 4 Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, 22 Hankou Road, Nanjing, 210093, China. [email protected].
Abstract

Background: Increasing evidence suggests that multiple or long-time exposure to general anaesthesia (GA) could be detrimental to cognitive development in young subjects and might also contribute to accelerated neurodegeneration in the elderly. Iron is essential for normal neuronal function, and excess iron in the brain is implicated in several neurodegenerative diseases. However, the role of iron in GA-induced neurotoxicity and cognitive deficits remains elusive.

Methods: We used the primary hippocampal neurons and rodents including young rats and aged mice to examine whether GA impacted iron metabolism and whether the impact contributed to neuronal outcomes. In addition, a pharmacological suppression of iron metabolism was performed to explore the molecular mechanism underlying GA-mediated iron overload in the brain.

Results: Our results demonstrated that GA, induced by intravenous ketamine or inhalational sevoflurane, disturbed iron homeostasis and caused iron overload in both in vitro hippocampal neuron culture and in vivo hippocampus. Interestingly, ketamine- or sevoflurane-induced cognitive deficits, very likely, resulted from a novel iron-dependent regulated cell death, Ferroptosis. Notably, iron chelator deferiprone attenuated the GA-induced mitochondrial dysfunction, Ferroptosis, and further cognitive deficits. Moreover, we found that GA-induced iron overload was activated by NMDAR-RASD1 signalling via DMT1 action in the brain.

Conclusion: We conclude that disturbed iron metabolism may be involved in the pathogenesis of GA-induced neurotoxicity and cognitive deficits. Our study provides new vision for consideration in GA-associated neurological disorders.

Keywords

Cognition; Ferroptosis; General anaesthesia; Iron; Neurotoxicity.

Figures
Products