单胺氧化酶抑制剂沙芬胺能调节钠离子电流的大小、门控和滞后。

IF 2.8 3区 医学 Q2 PHARMACOLOGY & PHARMACY BMC Pharmacology & Toxicology Pub Date : 2024-02-08 DOI:10.1186/s40360-024-00739-5
Te-Yu Hung, Sheng-Nan Wu, Chin-Wei Huang
{"title":"单胺氧化酶抑制剂沙芬胺能调节钠离子电流的大小、门控和滞后。","authors":"Te-Yu Hung, Sheng-Nan Wu, Chin-Wei Huang","doi":"10.1186/s40360-024-00739-5","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Safinamide (SAF), an α-aminoamide derivative and a selective, reversible monoamine oxidase (MAO)-B inhibitor, has both dopaminergic and nondopaminergic (glutamatergic) properties. Several studies have explored the potential of SAF against various neurological disorders; however, to what extent SAF modulates the magnitude, gating, and voltage-dependent hysteresis [Hys<sub>(V)</sub>] of ionic currents remains unknown.</p><p><strong>Methods: </strong>With the aid of patch-clamp technology, we investigated the effects of SAF on voltage-gated sodium ion (Na<sub>V</sub>) channels in pituitary GH3 cells.</p><p><strong>Results: </strong>SAF concentration-dependently stimulated the transient (peak) and late (sustained) components of voltage-gated sodium ion current (I<sub>Na</sub>) in pituitary GH<sub>3</sub> cells. The conductance-voltage relationship of transient I<sub>Na</sub> [I<sub>Na(T)</sub>] was shifted to more negative potentials with the SAF presence; however, the steady-state inactivation curve of I<sub>Na(T)</sub> was shifted in a rightward direction in its existence. SAF increased the decaying time constant of I<sub>Na(T)</sub> induced by a train of depolarizing stimuli. Notably, subsequent addition of ranolazine or mirogabalin reversed the SAF-induced increase in the decaying time constant. SAF also increased the magnitude of window I<sub>Na</sub> induced by an ascending ramp voltage V<sub>ramp</sub>. Furthermore, SAF enhanced the Hys<sub>(V)</sub> behavior of persistent I<sub>Na</sub> induced by an upright isosceles-triangular V<sub>ramp</sub>. Single-channel cell-attached recordings indicated SAF effectively increased the open-state probability of Na<sub>V</sub> channels. Molecular docking revealed SAF interacts with both MAO and Na<sub>V</sub> channels.</p><p><strong>Conclusion: </strong>SAF may interact directly with Na<sub>V</sub> channels in pituitary neuroendocrine cells, modulating membrane excitability.</p>","PeriodicalId":9023,"journal":{"name":"BMC Pharmacology & Toxicology","volume":null,"pages":null},"PeriodicalIF":2.8000,"publicationDate":"2024-02-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10851555/pdf/","citationCount":"0","resultStr":"{\"title\":\"Safinamide, an inhibitor of monoamine oxidase, modulates the magnitude, gating, and hysteresis of sodium ion current.\",\"authors\":\"Te-Yu Hung, Sheng-Nan Wu, Chin-Wei Huang\",\"doi\":\"10.1186/s40360-024-00739-5\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><strong>Background: </strong>Safinamide (SAF), an α-aminoamide derivative and a selective, reversible monoamine oxidase (MAO)-B inhibitor, has both dopaminergic and nondopaminergic (glutamatergic) properties. Several studies have explored the potential of SAF against various neurological disorders; however, to what extent SAF modulates the magnitude, gating, and voltage-dependent hysteresis [Hys<sub>(V)</sub>] of ionic currents remains unknown.</p><p><strong>Methods: </strong>With the aid of patch-clamp technology, we investigated the effects of SAF on voltage-gated sodium ion (Na<sub>V</sub>) channels in pituitary GH3 cells.</p><p><strong>Results: </strong>SAF concentration-dependently stimulated the transient (peak) and late (sustained) components of voltage-gated sodium ion current (I<sub>Na</sub>) in pituitary GH<sub>3</sub> cells. The conductance-voltage relationship of transient I<sub>Na</sub> [I<sub>Na(T)</sub>] was shifted to more negative potentials with the SAF presence; however, the steady-state inactivation curve of I<sub>Na(T)</sub> was shifted in a rightward direction in its existence. SAF increased the decaying time constant of I<sub>Na(T)</sub> induced by a train of depolarizing stimuli. Notably, subsequent addition of ranolazine or mirogabalin reversed the SAF-induced increase in the decaying time constant. SAF also increased the magnitude of window I<sub>Na</sub> induced by an ascending ramp voltage V<sub>ramp</sub>. Furthermore, SAF enhanced the Hys<sub>(V)</sub> behavior of persistent I<sub>Na</sub> induced by an upright isosceles-triangular V<sub>ramp</sub>. Single-channel cell-attached recordings indicated SAF effectively increased the open-state probability of Na<sub>V</sub> channels. Molecular docking revealed SAF interacts with both MAO and Na<sub>V</sub> channels.</p><p><strong>Conclusion: </strong>SAF may interact directly with Na<sub>V</sub> channels in pituitary neuroendocrine cells, modulating membrane excitability.</p>\",\"PeriodicalId\":9023,\"journal\":{\"name\":\"BMC Pharmacology & Toxicology\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":2.8000,\"publicationDate\":\"2024-02-08\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10851555/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"BMC Pharmacology & Toxicology\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.1186/s40360-024-00739-5\",\"RegionNum\":3,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q2\",\"JCRName\":\"PHARMACOLOGY & PHARMACY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"BMC Pharmacology & Toxicology","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1186/s40360-024-00739-5","RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q2","JCRName":"PHARMACOLOGY & PHARMACY","Score":null,"Total":0}
引用次数: 0

摘要

背景:萨非那胺(SAF)是一种α-氨基酰胺衍生物,也是一种选择性、可逆的单胺氧化酶(MAO)-B 抑制剂,具有多巴胺能和非多巴胺能(谷氨酸能)特性。有几项研究探讨了 SAF 治疗各种神经系统疾病的潜力;然而,SAF 在多大程度上调节离子电流的大小、门控和电压依赖性滞后 [Hys(V)]仍是未知数:方法:借助贴片钳技术,我们研究了 SAF 对垂体 GH3 细胞中电压门控钠离子(NaV)通道的影响:结果:SAF浓度依赖性地刺激了垂体GH3细胞中电压门控钠离子电流(INa)的瞬时(峰值)和晚期(持续)成分。瞬时INa[INa(T)]的电导-电压关系随SAF的存在而向更负的电位移动;然而,INa(T)的稳态失活曲线在其存在时向右移动。SAF 增加了一连串去极化刺激诱导的 INa(T)衰减时间常数。值得注意的是,随后加入雷诺嗪或米格巴林可逆转 SAF 诱导的衰减时间常数的增加。SAF 还增加了上升斜坡电压 Vramp 诱导的窗口 INa 的幅度。此外,SAF 还增强了直立等腰三角形 Vramp 诱导的持续 INa 的 Hys(V) 行为。单通道细胞连接记录表明,SAF 有效地增加了 NaV 通道的开放状态概率。分子对接显示 SAF 可与 MAO 和 NaV 通道相互作用:结论:SAF 可直接与垂体神经内分泌细胞中的 NaV 通道相互作用,从而调节膜的兴奋性。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
Safinamide, an inhibitor of monoamine oxidase, modulates the magnitude, gating, and hysteresis of sodium ion current.

Background: Safinamide (SAF), an α-aminoamide derivative and a selective, reversible monoamine oxidase (MAO)-B inhibitor, has both dopaminergic and nondopaminergic (glutamatergic) properties. Several studies have explored the potential of SAF against various neurological disorders; however, to what extent SAF modulates the magnitude, gating, and voltage-dependent hysteresis [Hys(V)] of ionic currents remains unknown.

Methods: With the aid of patch-clamp technology, we investigated the effects of SAF on voltage-gated sodium ion (NaV) channels in pituitary GH3 cells.

Results: SAF concentration-dependently stimulated the transient (peak) and late (sustained) components of voltage-gated sodium ion current (INa) in pituitary GH3 cells. The conductance-voltage relationship of transient INa [INa(T)] was shifted to more negative potentials with the SAF presence; however, the steady-state inactivation curve of INa(T) was shifted in a rightward direction in its existence. SAF increased the decaying time constant of INa(T) induced by a train of depolarizing stimuli. Notably, subsequent addition of ranolazine or mirogabalin reversed the SAF-induced increase in the decaying time constant. SAF also increased the magnitude of window INa induced by an ascending ramp voltage Vramp. Furthermore, SAF enhanced the Hys(V) behavior of persistent INa induced by an upright isosceles-triangular Vramp. Single-channel cell-attached recordings indicated SAF effectively increased the open-state probability of NaV channels. Molecular docking revealed SAF interacts with both MAO and NaV channels.

Conclusion: SAF may interact directly with NaV channels in pituitary neuroendocrine cells, modulating membrane excitability.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
BMC Pharmacology & Toxicology
BMC Pharmacology & Toxicology PHARMACOLOGY & PHARMACYTOXICOLOGY&nb-TOXICOLOGY
CiteScore
4.80
自引率
0.00%
发文量
87
审稿时长
12 weeks
期刊介绍: BMC Pharmacology and Toxicology is an open access, peer-reviewed journal that considers articles on all aspects of chemically defined therapeutic and toxic agents. The journal welcomes submissions from all fields of experimental and clinical pharmacology including clinical trials and toxicology.
期刊最新文献
Sodium Houttuyniae attenuates ferroptosis by regulating TRAF6-c-Myc signaling pathways in lipopolysaccharide-induced acute lung injury (ALI). The subchronic toxicity of higher olefins in Han Wistar rats. Appropriate use of triazolam in elderly patients considering a quantitative benefit-risk assessment based on the pharmacokinetic-pharmacodynamic modeling and simulation approach supported by real-world data. Comparison of the efficacy and adverse effects of oral ferrous succinate tablets and intravenous iron sucrose: a retrospective study. Drug-induced liver injury associated with atypical generation antipsychotics from the FDA Adverse Event Reporting System (FAERS).
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1