1. Academic Validation
  2. METTL14-mediated m6A epitranscriptomic modification contributes to chemotherapy-induced neuropathic pain by stabilizing GluN2A expression via IGF2BP2

METTL14-mediated m6A epitranscriptomic modification contributes to chemotherapy-induced neuropathic pain by stabilizing GluN2A expression via IGF2BP2

  • J Clin Invest. 2024 Feb 6:e174847. doi: 10.1172/JCI174847.
Weicheng Lu 1 Xiaohua Yang 1 Weiqiang Zhong 1 Guojun Chen 1 Xinqi Guo 2 Qingqing Ye 1 Yixin Xu 1 Zhenhua Qi 1 Yaqi Ye 1 Jingyun Zhang 3 Yuge Wang 3 Xintong Wang 1 Shu Wang 1 Qiyue Zhao 2 Weian Zeng 1 Junting Huang 3 Huijie Ma 2 Jingdun Xie 1
Affiliations

Affiliations

  • 1 Department of Anesthesiology, State Key Laboratory of Oncology in South Chi, Sun Yat-sen University Cancer Center, Guangzhou, China.
  • 2 Department of Physiology, Hebei Medical University, Shijiazhuang, China.
  • 3 Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
Abstract

Epigenetics is a biological process that modifies and regulates gene expression, affects neuronal function, and contributes to pain. However, the mechanism by which Epigenetics facilitates and maintains chronic pain is poorly understood. We aimed to determine whether N6-methyladenosine (m6A) specifically modified by methyltransferase 14 (METTL14) alters neuronal activity and governs pain by sensitizing the GluN2A subunit of the N-methyl-D-aspartate receptor (NMDAR) in the dorsal root ganglion (DRG) neurons in a model of chemotherapy-induced neuropathic pain (CINP). Using dot blotting, immunofluorescence, gain/loss-of-function, and behavioral assays, we found that m6A levels were upregulated in L4-L6 DRG neurons in the CINP in a DBP/METT14-dependent manner, which was also confirmed in human DRGs. Blocking METTL14 reduced m6A methylation and attenuated pain hypersensitivity. Mechanistically, METTL14-mediated m6A modification facilitated the synaptic plasticity of DRG neurons by enhancing the GluN2A subunit of NMDAR, and inhibiting METTL14 blocked this effect. In contrast, overexpression of METTL14 upregulated m6A modifications, enhanced presynaptic NMDAR activity in DRG neurons, and facilitated pain sensation. Our findings reveal a previously unrecognized mechanism of METTL14-mediated m6A modification in DRG neurons to maintain neuropathic pain. Targeting these molecules may provide a new strategy for pain treatment.

Keywords

Epigenetics; Neuroscience; Pain; Synapses.

Figures
Products