RT Journal Article SR Electronic T1 Selective Activation of TASK-3-containing K+ Channels Reveals Their Therapeutic Potentials in Analgesia JF bioRxiv FD Cold Spring Harbor Laboratory SP 657387 DO 10.1101/657387 A1 Ping Liao A1 Yunguang Qiu A1 Yiqing Mo A1 Jie Fu A1 Zhenpeng Song A1 Lu Huang A1 Suwen Bai A1 Yang Wang A1 Jia-Jie Zhu A1 Fuyun Tian A1 Zhuo Chen A1 Nanfang Pan A1 Er-Yi Sun A1 Linghui Yang A1 Xi Lan A1 Yinbin Chen A1 Dongping Huang A1 Peihua Sun A1 Lifen Zhao A1 Dehua Yang A1 Weiqiang Lu A1 Tingting Yang A1 Junjie Xiao A1 Wei-Guang Li A1 Zhaobing Gao A1 Bing Shen A1 Qiansen Zhang A1 Jin Liu A1 Hualiang Jiang A1 Ruotian Jiang A1 Huaiyu Yang YR 2019 UL http://biorxiv.org/content/early/2019/06/05/657387.abstract AB The paucity of selective agonists for TASK-3, a member of two-pore domain K+ (K2P) channels, limited our understanding of its biological functions. Targeting a novel druggable transmembrane cavity using structure-based drug design approach, we discovered a biguanide compound CHET3 as a highly selective allosteric activator for TASK-3-containing K2P channels including TASK-3 homomer and TASK-3/TASK-1 heteromer. CHET3 displayed unexpectedly potent analgesia in vivo in a variety of acute and chronic pain models in rodents that could be abolished by pharmacology or genetic ablation of TASK-3. We further found that TASK-3-containing channels anatomically define a unique subset population of small-sized, TRPM8, TRPV1 or Tyrosine Hydroxylase positive nociceptive sensory neurons and functionally regulate their membrane excitability, supporting the CHET3 analgesia in thermal hyperalgesia and mechanical allodynia under chronic pain. Overall, our proof-of-concept study reveals TASK-3-containing K2P channels as a novel druggable target for treating pain.One Sentence Summary Identification of a novel drug target and its new hit compounds for developing new-generation non-opioid analgesics.