Diversely evolved xibalbin variants from remipede venom inhibit potassium channels and activate PKA-II and Erk1/2 signaling.

IF 4.4 1区 生物学 Q1 BIOLOGY BMC Biology Pub Date : 2024-07-29 DOI:10.1186/s12915-024-01955-5
Ernesto Lopes Pinheiro-Junior, Ehsan Alirahimi, Steve Peigneur, Jörg Isensee, Susanne Schiffmann, Pelin Erkoc, Robert Fürst, Andreas Vilcinskas, Tobias Sennoner, Ivan Koludarov, Benjamin-Florian Hempel, Jan Tytgat, Tim Hucho, Björn M von Reumont
{"title":"Diversely evolved xibalbin variants from remipede venom inhibit potassium channels and activate PKA-II and Erk1/2 signaling.","authors":"Ernesto Lopes Pinheiro-Junior, Ehsan Alirahimi, Steve Peigneur, Jörg Isensee, Susanne Schiffmann, Pelin Erkoc, Robert Fürst, Andreas Vilcinskas, Tobias Sennoner, Ivan Koludarov, Benjamin-Florian Hempel, Jan Tytgat, Tim Hucho, Björn M von Reumont","doi":"10.1186/s12915-024-01955-5","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>The identification of novel toxins from overlooked and taxonomically exceptional species bears potential for various pharmacological applications. The remipede Xibalbanus tulumensis, an underwater cave-dwelling crustacean, is the only crustacean for which a venom system has been described. Its venom contains several xibalbin peptides that have an inhibitor cysteine knot (ICK) scaffold.</p><p><strong>Results: </strong>Our screenings revealed that all tested xibalbin variants particularly inhibit potassium channels. Xib<sub>1</sub> and xib<sub>13</sub> with their eight-cysteine domain similar to spider knottins also inhibit voltage-gated sodium channels. No activity was noted on calcium channels. Expanding the functional testing, we demonstrate that xib<sub>1</sub> and xib<sub>13</sub> increase PKA-II and Erk1/2 sensitization signaling in nociceptive neurons, which may initiate pain sensitization. Our phylogenetic analysis suggests that xib<sub>13</sub> either originates from the common ancestor of pancrustaceans or earlier while xib<sub>1</sub> is more restricted to remipedes. The ten-cysteine scaffolded xib<sub>2</sub> emerged from xib<sub>1</sub>, a result that is supported by our phylogenetic and machine learning-based analyses.</p><p><strong>Conclusions: </strong>Our functional characterization of synthesized variants of xib<sub>1</sub>, xib<sub>2</sub>, and xib<sub>13</sub> elucidates their potential as inhibitors of potassium channels in mammalian systems. The specific interaction of xib<sub>2</sub> with Kv1.6 channels, which are relevant to treating variants of epilepsy, shows potential for further studies. At higher concentrations, xib<sub>1</sub> and xib<sub>13</sub> activate the kinases PKA-II and ERK1/2 in mammalian sensory neurons, suggesting pain sensitization and potential applications related to pain research and therapy. While tested insect channels suggest that all probably act as neurotoxins, the biological function of xib<sub>1</sub>, xib<sub>2,</sub> and xib<sub>13</sub> requires further elucidation. A novel finding on their evolutionary origin is the apparent emergence of X. tulumensis-specific xib<sub>2</sub> from xib<sub>1</sub>. Our study is an important cornerstone for future studies to untangle the origin and function of these enigmatic proteins as important components of remipede but also other pancrustacean and arthropod venoms.</p>","PeriodicalId":9339,"journal":{"name":"BMC Biology","volume":null,"pages":null},"PeriodicalIF":4.4000,"publicationDate":"2024-07-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11288129/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"BMC Biology","FirstCategoryId":"99","ListUrlMain":"https://doi.org/10.1186/s12915-024-01955-5","RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"BIOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Background: The identification of novel toxins from overlooked and taxonomically exceptional species bears potential for various pharmacological applications. The remipede Xibalbanus tulumensis, an underwater cave-dwelling crustacean, is the only crustacean for which a venom system has been described. Its venom contains several xibalbin peptides that have an inhibitor cysteine knot (ICK) scaffold.

Results: Our screenings revealed that all tested xibalbin variants particularly inhibit potassium channels. Xib1 and xib13 with their eight-cysteine domain similar to spider knottins also inhibit voltage-gated sodium channels. No activity was noted on calcium channels. Expanding the functional testing, we demonstrate that xib1 and xib13 increase PKA-II and Erk1/2 sensitization signaling in nociceptive neurons, which may initiate pain sensitization. Our phylogenetic analysis suggests that xib13 either originates from the common ancestor of pancrustaceans or earlier while xib1 is more restricted to remipedes. The ten-cysteine scaffolded xib2 emerged from xib1, a result that is supported by our phylogenetic and machine learning-based analyses.

Conclusions: Our functional characterization of synthesized variants of xib1, xib2, and xib13 elucidates their potential as inhibitors of potassium channels in mammalian systems. The specific interaction of xib2 with Kv1.6 channels, which are relevant to treating variants of epilepsy, shows potential for further studies. At higher concentrations, xib1 and xib13 activate the kinases PKA-II and ERK1/2 in mammalian sensory neurons, suggesting pain sensitization and potential applications related to pain research and therapy. While tested insect channels suggest that all probably act as neurotoxins, the biological function of xib1, xib2, and xib13 requires further elucidation. A novel finding on their evolutionary origin is the apparent emergence of X. tulumensis-specific xib2 from xib1. Our study is an important cornerstone for future studies to untangle the origin and function of these enigmatic proteins as important components of remipede but also other pancrustacean and arthropod venoms.

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
从remipede毒液中演化出的不同xibalbin变体可抑制钾通道并激活PKA-II和Erk1/2信号传导。
背景:从被忽视和分类学上特殊的物种中鉴定新型毒素具有各种药理应用的潜力。Xibalbanus tulumensis是一种生活在水下洞穴中的甲壳类动物,也是唯一一种被描述有毒液系统的甲壳类动物。它的毒液中含有几种具有抑制半胱氨酸结(ICK)支架的 xibalbin 肽:结果:我们的筛选结果表明,所有测试的 xibalbin 变体都特别抑制钾通道。Xib1 和 xib13 的 8 个半胱氨酸结构域与蜘蛛结蛋白相似,也能抑制电压门控钠通道。对钙通道没有抑制作用。通过扩大功能测试,我们证明 xib1 和 xib13 增加了痛觉神经元中的 PKA-II 和 Erk1/2 敏化信号,这可能会启动痛觉敏化。我们的系统发育分析表明,xib13起源于泛壳类动物的共同祖先或更早,而xib1则更多地局限于 remipedes。十半胱氨酸支架的 xib2 来自 xib1,这一结果得到了我们的系统发育和基于机器学习的分析的支持:我们对合成的 xib1、xib2 和 xib13 变体进行的功能表征阐明了它们在哺乳动物系统中作为钾通道抑制剂的潜力。xib2与Kv1.6通道的特异性相互作用显示了进一步研究的潜力,而Kv1.6通道与癫痫变体的治疗相关。在较高浓度下,xib1 和 xib13 能激活哺乳动物感觉神经元中的激酶 PKA-II 和 ERK1/2,这表明它们具有痛觉敏感性,具有与疼痛研究和治疗相关的潜在应用价值。虽然经过测试的昆虫通道表明它们都可能具有神经毒素的作用,但 xib1、xib2 和 xib13 的生物功能还需要进一步阐明。关于它们进化起源的一个新发现是,X. tulumensis 特异性 xib2 显然是从 xib1 中产生的。我们的研究为今后的研究奠定了重要的基石,有助于解开这些神秘蛋白的起源和功能,它们不仅是雷米佩德毒液的重要成分,也是其他泛甲壳动物和节肢动物毒液的重要成分。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 去求助
来源期刊
BMC Biology
BMC Biology 生物-生物学
CiteScore
7.80
自引率
1.90%
发文量
260
审稿时长
3 months
期刊介绍: BMC Biology is a broad scope journal covering all areas of biology. Our content includes research articles, new methods and tools. BMC Biology also publishes reviews, Q&A, and commentaries.
期刊最新文献
Genome-wide analysis reveals transcriptional and translational changes during diapause of the Asian corn borer (Ostrinia furnacalis) The subcellular distribution of miRNA isoforms, tRNA-derived fragments, and rRNA-derived fragments depends on nucleotide sequence and cell type Genomes of diverse Actinidia species provide insights into cis-regulatory motifs and genes associated with critical traits The mrp-3 gene is involved in haem efflux and detoxification in a blood-feeding nematode Single-cell RNA-seq analysis of rat molars reveals cell identity and driver genes associated with dental mesenchymal cell differentiation
×
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