4.7 Article

The Role of Prostaglandin E1 as a Pain Mediator through Facilitation of Hyperpolarization-Activated Cyclic Nucleotide-Gated Channel 2 via the EP2 Receptor in Trigeminal Ganglion Neurons of Mice

期刊

出版社

MDPI
DOI: 10.3390/ijms222413534

关键词

EP2; HCN2 channel; pain; PGE1; trigeminal ganglion neuron

资金

  1. National Research Foundation (NRF) of Korea [NRF-2021R1A2C2011021]
  2. Hanyang University [HY-202100000003298]

向作者/读者索取更多资源

The study confirms that PGE1 can induce pain in mice and enhances the excitability of trigeminal ganglion neurons through HCN channel facilitation. The research also identifies the target mechanism of PGE1 in nociceptive neurons, suggesting its potential as a therapeutic target for pain management under inflammatory conditions.
Cyclooxygenase metabolizes dihomo-gamma-linolenic acid and arachidonic acid to form prostaglandin (PG) E, including PGE1 and PGE2, respectively. Although PGE2 is well known to play an important role in the development and maintenance of hyperalgesia and allodynia, the role of PGE1 in pain is unknown. We confirm whether PGE1 induced pain using orofacial pain behavioral test in mice and determine the target molecule of PGE1 in TG neurons with whole-cell patch-clamp and immunohistochemistry. Intradermal injection of PGE1 to the whisker pads of mice induced a reduced threshold, enhancing the excitability of HCN channel-expressing trigeminal ganglion (TG) neurons. The HCN channel-generated inward current (I-h) was increased by 135.3 +/- 4.8% at 100 nM of PGE1 in small- or medium-sized TG, and the action of PGE1 on I-h showed a concentration-dependent effect, with a median effective dose (ED50) of 29.3 nM. Adenylyl cyclase inhibitor (MDL12330A), 8-bromo-cAMP, and the EP2 receptor antagonist AH6809 inhibited PGE1-induced I-h. Additionally, PGE1-induced mechanical allodynia was blocked by CsCl and AH6809. PGE1 plays a role in mechanical allodynia through HCN2 channel facilitation via the EP2 receptor in nociceptive neurons, suggesting a potential therapeutic target in that PGE1 could be involved in pain as endogenous substances under inflammatory conditions.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.7
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据