Sendai virus-mediated RNA delivery restores fertility to congenital and chemotherapy-induced infertile female mice

Mito Kanatsu-Shinohara, Hiroko Morimoto, Tianjiao Liu, Masaru Tamura, Takashi Shinohara
{"title":"Sendai virus-mediated RNA delivery restores fertility to congenital and chemotherapy-induced infertile female mice","authors":"Mito Kanatsu-Shinohara, Hiroko Morimoto, Tianjiao Liu, Masaru Tamura, Takashi Shinohara","doi":"10.1093/pnasnexus/pgae375","DOIUrl":null,"url":null,"abstract":"Current infertility treatment strategies focus on mature gametes, leaving a significant proportion of cases with gamete progenitors that stopped complete differentiation. On the other hand, recent advancements in next generation sequencing have identified many candidate genes that may promote maturation of germ cells. Although gene therapy has shown success in mice, concerns about the integration of DNA vectors into oocytes hinder clinical applications. Here, we present the restoration of fertility in female mice through Sendai virus (SeV)-mediated RNA delivery. Ovaries lacking Kitl expression exhibit only primordial follicles due to impaired signaling to oocytes expressing the KIT tyrosine kinase. Despite SeVs being immunogenic and larger than the blood-follicle barrier, the administration of Kitl-expressing SeVs reinitiated oogenesis in genetically infertile mice that have only primordial follicles, resulting in the birth of normal offspring through natural mating. This virus also effectively addressed iatrogenic infertility induced by busulfan, a widely used cancer chemotherapy agent. Offspring born through SeV administration and natural mating displayed normal genomic imprinting patterns and fertility. Since SeVs pose no genotoxicity risk, the successful restoration of fertility by SeVs represents a promising approach for treating congenital infertility with somatic cell defects and protecting fertility of cancer patients who may become infertile due to loss of oocytes during cancer therapy.","PeriodicalId":516525,"journal":{"name":"PNAS Nexus","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2024-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"PNAS Nexus","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1093/pnasnexus/pgae375","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

Abstract

Current infertility treatment strategies focus on mature gametes, leaving a significant proportion of cases with gamete progenitors that stopped complete differentiation. On the other hand, recent advancements in next generation sequencing have identified many candidate genes that may promote maturation of germ cells. Although gene therapy has shown success in mice, concerns about the integration of DNA vectors into oocytes hinder clinical applications. Here, we present the restoration of fertility in female mice through Sendai virus (SeV)-mediated RNA delivery. Ovaries lacking Kitl expression exhibit only primordial follicles due to impaired signaling to oocytes expressing the KIT tyrosine kinase. Despite SeVs being immunogenic and larger than the blood-follicle barrier, the administration of Kitl-expressing SeVs reinitiated oogenesis in genetically infertile mice that have only primordial follicles, resulting in the birth of normal offspring through natural mating. This virus also effectively addressed iatrogenic infertility induced by busulfan, a widely used cancer chemotherapy agent. Offspring born through SeV administration and natural mating displayed normal genomic imprinting patterns and fertility. Since SeVs pose no genotoxicity risk, the successful restoration of fertility by SeVs represents a promising approach for treating congenital infertility with somatic cell defects and protecting fertility of cancer patients who may become infertile due to loss of oocytes during cancer therapy.
仙台病毒介导的核糖核酸递送可恢复先天性和化疗引起的不育雌性小鼠的生育能力
目前的不孕症治疗策略主要针对成熟的配子,因此有相当一部分病例的配子祖细胞停止了完全分化。另一方面,下一代测序技术的最新进展发现了许多可能促进生殖细胞成熟的候选基因。虽然基因治疗在小鼠身上取得了成功,但 DNA 载体整合到卵母细胞中的问题阻碍了临床应用。在这里,我们介绍了通过仙台病毒(SeV)介导的 RNA 递送恢复雌性小鼠生育能力的方法。由于向表达 KIT 酪氨酸激酶的卵母细胞发出的信号受损,缺乏 Kitl 表达的卵巢只表现出原始卵泡。尽管 SeVs 具有免疫原性,且体积大于血-卵泡屏障,但给只有原始卵泡的遗传性不育小鼠注射表达 Kitl 的 SeVs 后,小鼠的卵子生成得以恢复,并通过自然交配产下了正常的后代。这种病毒还有效地解决了广泛使用的癌症化疗药物--丁硫克百威(busulfan)诱发的先天性不孕症。通过 SeV 给药和自然交配出生的后代显示出正常的基因组印记模式和生育能力。由于 SeV 没有遗传毒性风险,因此 SeV 成功恢复生育能力是治疗体细胞缺陷先天性不孕症和保护癌症患者生育能力的一种有前途的方法。
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 求助全文
来源期刊
自引率
0.00%
发文量
0
×
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
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信