1. Academic Validation
  2. IL-33 Provides Neuroprotection through Suppressing Apoptotic, Autophagic and NF-κB-Mediated Inflammatory Pathways in a Rat Model of Recurrent Neonatal Seizure

IL-33 Provides Neuroprotection through Suppressing Apoptotic, Autophagic and NF-κB-Mediated Inflammatory Pathways in a Rat Model of Recurrent Neonatal Seizure

  • Front Mol Neurosci. 2017 Dec 19;10:423. doi: 10.3389/fnmol.2017.00423.
Yuan Gao 1 2 Cheng-Liang Luo 2 Li-Li Li 3 Guang-Hua Ye 1 Cheng Gao 2 Hao-Chen Wang 2 Wen-Wen Huang 2 Tao Wang 2 Zu-Feng Wang 2 Hong Ni 3 Xi-Ping Chen 2 Lu-Yang Tao 2
Affiliations

Affiliations

  • 1 Department of Forensic Medicine, Wenzhou Medical University, Wenzhou, China.
  • 2 Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China.
  • 3 Department of Neurology Laboratory, Children's Hospital of Soochow University, Suzhou, China.
Abstract

Interleukin-33 (IL-33) is a novel identified chromatin-associated cytokine of IL-1 family cytokines. It signals through a heterodimer comprised of ST2L and IL-1RAcp, and plays a crucial role in many diseases. However, very little is known about the role and underlying intricate mechanisms of IL-33 in recurrent neonatal seizure (RNS). To determine whether IL-33 plays an important regulatory role, we established a neonatal seizure model in this study. Rats were subjected to recurrent seizures induced by inhaling volatile flurothyl. Recombinant IL-33 or PBS were also administered by intraperitoneally (IP) before surgery, respectively. Here, our current results indicated that RNS contributed to a significant reduction in IL-33 and its specific receptor (ST2L) expressions in cortex. While, in hippocampus, RNS induced an increase in IL-33 and ST2L evidently, compared with Sham group. After injection with IL-33, however, a remarkable increase in total IL-33 was detected both in brain cortex and hippocampus. In addition, IL-33 was mainly co-localized in the nuclear of GFAP+ astrocytes and the cytoplasm of the Iba-1+ microglia and IL-33+/NeuN+ merged cells. In parallel, ST2L was expressed mainly in the membrane of GFAP+ astrocytes, Iba-1+ microglia and NeuN+ neurons, respectively. Furthermore, administration of IL-33 improved RNS-induced behavioral deficits, promoted bodyweight gain, and ameliorated spatial learning and memory ability. Moreover, IL-33 pretreatment blocked the activation of NF-κB, resisted inflammatory cytokines IL-1β and TNF-α increase, as well as suppressed Apoptosis and Autophagy activation after RNS. Collectively, IL-33 provides potential neuroprotection through suppressing Apoptosis, Autophagy and at least in part by NF-κB-mediated inflammatory pathways after RNS.

Keywords

IL-33; ST2L; apoptosis; autophagy; recurrent neonatal seizure.

Figures
Products