TRMT1 的双叶致病变体会破坏 tRNA 的修饰并诱发综合神经发育障碍

Stephanie Efthymiou, Cailyn Leo, Chenghong Deng, Kejia Zhang, Sheng-Jia Lin, Reza Maroofian, Rauan Kaiyrzhanov, Renee Qing Lin, Irem Karagoz, Annarita Scardamaglia, Daniel Owrang, Valentina Turchetti, Friederike Jahnke, Cassidy Petree, Anna V Derrick, Mark I Rees, Javeria Raza Alvi, Tipu Sultan, Chumei Li, Marie-Line Jacquemont, Frederic Tran-Mau-Them, Maria Irene Valenzuela, Rich Sidlow, Grace Yoon, Michelle Morrow, Alexis Carere, Mary O'Connor, Julie Fleischer, Erica H. Gerkes, Chanika Phornphutkul, Bertrand Isidor, Clotilde Rivier-Ringenbach, Christophe Philippe, Semra Hiz Kurul, Didem Soydemir, Bulent Kara, Deniz Sunnetci-Akkoyunlu, Viktoria Bothe, Konrad Platzer, Dagmar Wieczorek, Margarete Koch-Hogrebe, Nils Rahner, Ann-Charlotte Thuresson, Hans Matsson, Carina Frykholm, Sevcan Tug Bozdogan, Atil Bisgin, Nicolas Chatron, Gaetan Lesca, Sara Cabet, Zeynep Tumer, Tina Duelund Hjortshoj, Gitte Ronde, Thorsten Marquardt, Janine Reunert, Erum Afzal, Mina Zamani, Reza Azizimalamiri, Hamid Galehdari, Pardis Nourbakhshd, Niloofar Chamanrou, Seo-Kyung Chung, Mohnish Suri, Paul J Benke, Maha S Zaki, Joseph G Gleeson, Daniel G Calame, Davut Pehlivan, Halil Ibrahim Yilmaz, Alper Gezdirici, Aboulfazl Rad, Iman Sabri Abumansour, Gabriela Oprea, Jai Sidpra, Kshitij Mankad, Barbara Vona, Andrew Fry, Gaurav K Varshney, Henry Houlden, Dragony Fu
{"title":"TRMT1 的双叶致病变体会破坏 tRNA 的修饰并诱发综合神经发育障碍","authors":"Stephanie Efthymiou, Cailyn Leo, Chenghong Deng, Kejia Zhang, Sheng-Jia Lin, Reza Maroofian, Rauan Kaiyrzhanov, Renee Qing Lin, Irem Karagoz, Annarita Scardamaglia, Daniel Owrang, Valentina Turchetti, Friederike Jahnke, Cassidy Petree, Anna V Derrick, Mark I Rees, Javeria Raza Alvi, Tipu Sultan, Chumei Li, Marie-Line Jacquemont, Frederic Tran-Mau-Them, Maria Irene Valenzuela, Rich Sidlow, Grace Yoon, Michelle Morrow, Alexis Carere, Mary O'Connor, Julie Fleischer, Erica H. Gerkes, Chanika Phornphutkul, Bertrand Isidor, Clotilde Rivier-Ringenbach, Christophe Philippe, Semra Hiz Kurul, Didem Soydemir, Bulent Kara, Deniz Sunnetci-Akkoyunlu, Viktoria Bothe, Konrad Platzer, Dagmar Wieczorek, Margarete Koch-Hogrebe, Nils Rahner, Ann-Charlotte Thuresson, Hans Matsson, Carina Frykholm, Sevcan Tug Bozdogan, Atil Bisgin, Nicolas Chatron, Gaetan Lesca, Sara Cabet, Zeynep Tumer, Tina Duelund Hjortshoj, Gitte Ronde, Thorsten Marquardt, Janine Reunert, Erum Afzal, Mina Zamani, Reza Azizimalamiri, Hamid Galehdari, Pardis Nourbakhshd, Niloofar Chamanrou, Seo-Kyung Chung, Mohnish Suri, Paul J Benke, Maha S Zaki, Joseph G Gleeson, Daniel G Calame, Davut Pehlivan, Halil Ibrahim Yilmaz, Alper Gezdirici, Aboulfazl Rad, Iman Sabri Abumansour, Gabriela Oprea, Jai Sidpra, Kshitij Mankad, Barbara Vona, Andrew Fry, Gaurav K Varshney, Henry Houlden, Dragony Fu","doi":"10.1101/2024.07.18.24310581","DOIUrl":null,"url":null,"abstract":"The post-transcriptional modification of tRNAs plays a key role in tRNA folding and function to ensure proper levels of protein synthesis during growth and development. Pathogenic variants in tRNA modification enzymes have been implicated in diverse human neurodevelopmental and neurological disorders. However, the molecular basis for many of these disorders remains unknown, thereby limiting our understanding and potential treatment of pathologies linked to tRNA modification. Here, we describe an extensive cohort of 31 individuals from 24 unrelated families with bi-allelic variants in the <em>tRNA methyltransferase 1</em> (<em>TRMT1</em>) gene who present with a syndromic neurodevelopmental disorder universally characterized by intellectual disability in affected patients. Developmental delay, behavioral abnormalities and facial dysmorphisms represent additional core phenotypes of this syndrome. The variants include novel and ultra-rare TRMT1 variants that segregate with clinical pathology. We found that a subset of variants causes mis-splicing and loss of TRMT1 protein expression. Notably, patient cells with TRMT1 variants exhibit a deficiency in tRNA modifications catalyzed by TRMT1. Molecular analysis of TRMT1 variants reveal distinct regions of the TRMT1 protein required for tRNA modification activity and binding, including a TRMT1 subdomain critical for tRNA interaction. Importantly, depletion of TRMT1 in zebrafish is sufficient to induce developmental and behavioral phenotypes that recapitulate those observed in human patients with pathogenic TRMT1 variants. Altogether, these findings demonstrate that loss of TRMT1-catalyzed tRNA modifications leads to a syndromic form of intellectual disability and elucidate the molecular underpinnings of tRNA modification deficiency caused by pathogenic TRMT1 variants.","PeriodicalId":501375,"journal":{"name":"medRxiv - Genetic and Genomic Medicine","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2024-07-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":"{\"title\":\"Biallelic pathogenic variants in TRMT1 disrupt tRNA modification and induce a syndromic neurodevelopmental disorder\",\"authors\":\"Stephanie Efthymiou, Cailyn Leo, Chenghong Deng, Kejia Zhang, Sheng-Jia Lin, Reza Maroofian, Rauan Kaiyrzhanov, Renee Qing Lin, Irem Karagoz, Annarita Scardamaglia, Daniel Owrang, Valentina Turchetti, Friederike Jahnke, Cassidy Petree, Anna V Derrick, Mark I Rees, Javeria Raza Alvi, Tipu Sultan, Chumei Li, Marie-Line Jacquemont, Frederic Tran-Mau-Them, Maria Irene Valenzuela, Rich Sidlow, Grace Yoon, Michelle Morrow, Alexis Carere, Mary O'Connor, Julie Fleischer, Erica H. Gerkes, Chanika Phornphutkul, Bertrand Isidor, Clotilde Rivier-Ringenbach, Christophe Philippe, Semra Hiz Kurul, Didem Soydemir, Bulent Kara, Deniz Sunnetci-Akkoyunlu, Viktoria Bothe, Konrad Platzer, Dagmar Wieczorek, Margarete Koch-Hogrebe, Nils Rahner, Ann-Charlotte Thuresson, Hans Matsson, Carina Frykholm, Sevcan Tug Bozdogan, Atil Bisgin, Nicolas Chatron, Gaetan Lesca, Sara Cabet, Zeynep Tumer, Tina Duelund Hjortshoj, Gitte Ronde, Thorsten Marquardt, Janine Reunert, Erum Afzal, Mina Zamani, Reza Azizimalamiri, Hamid Galehdari, Pardis Nourbakhshd, Niloofar Chamanrou, Seo-Kyung Chung, Mohnish Suri, Paul J Benke, Maha S Zaki, Joseph G Gleeson, Daniel G Calame, Davut Pehlivan, Halil Ibrahim Yilmaz, Alper Gezdirici, Aboulfazl Rad, Iman Sabri Abumansour, Gabriela Oprea, Jai Sidpra, Kshitij Mankad, Barbara Vona, Andrew Fry, Gaurav K Varshney, Henry Houlden, Dragony Fu\",\"doi\":\"10.1101/2024.07.18.24310581\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"The post-transcriptional modification of tRNAs plays a key role in tRNA folding and function to ensure proper levels of protein synthesis during growth and development. Pathogenic variants in tRNA modification enzymes have been implicated in diverse human neurodevelopmental and neurological disorders. However, the molecular basis for many of these disorders remains unknown, thereby limiting our understanding and potential treatment of pathologies linked to tRNA modification. Here, we describe an extensive cohort of 31 individuals from 24 unrelated families with bi-allelic variants in the <em>tRNA methyltransferase 1</em> (<em>TRMT1</em>) gene who present with a syndromic neurodevelopmental disorder universally characterized by intellectual disability in affected patients. Developmental delay, behavioral abnormalities and facial dysmorphisms represent additional core phenotypes of this syndrome. The variants include novel and ultra-rare TRMT1 variants that segregate with clinical pathology. We found that a subset of variants causes mis-splicing and loss of TRMT1 protein expression. Notably, patient cells with TRMT1 variants exhibit a deficiency in tRNA modifications catalyzed by TRMT1. Molecular analysis of TRMT1 variants reveal distinct regions of the TRMT1 protein required for tRNA modification activity and binding, including a TRMT1 subdomain critical for tRNA interaction. Importantly, depletion of TRMT1 in zebrafish is sufficient to induce developmental and behavioral phenotypes that recapitulate those observed in human patients with pathogenic TRMT1 variants. Altogether, these findings demonstrate that loss of TRMT1-catalyzed tRNA modifications leads to a syndromic form of intellectual disability and elucidate the molecular underpinnings of tRNA modification deficiency caused by pathogenic TRMT1 variants.\",\"PeriodicalId\":501375,\"journal\":{\"name\":\"medRxiv - Genetic and Genomic Medicine\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":0.0000,\"publicationDate\":\"2024-07-22\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"medRxiv - Genetic and Genomic Medicine\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://doi.org/10.1101/2024.07.18.24310581\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"\",\"JCRName\":\"\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"medRxiv - Genetic and Genomic Medicine","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1101/2024.07.18.24310581","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

摘要

tRNA 的转录后修饰在 tRNA 的折叠和功能中发挥着关键作用,以确保在生长和发育过程中蛋白质合成的适当水平。tRNA 修饰酶的致病变异与多种人类神经发育和神经系统疾病有关。然而,许多这类疾病的分子基础仍然未知,从而限制了我们对与 tRNA 修饰有关的病理的理解和潜在治疗。在这里,我们描述了一个由来自 24 个非亲缘家庭的 31 人组成的庞大队列,这些人的 tRNA 甲基转移酶 1(TRMT1)基因存在双等位基因变异。发育迟缓、行为异常和面部畸形是该综合征的其他核心表型。这些变异包括与临床病理分离的新型和超罕见的 TRMT1 变异。我们发现,变体的一个子集会导致错误剪接和 TRMT1 蛋白表达的缺失。值得注意的是,带有 TRMT1 变体的患者细胞缺乏由 TRMT1 催化的 tRNA 修饰。对TRMT1变体的分子分析表明,TRMT1蛋白的不同区域需要tRNA修饰活性和结合,包括对tRNA相互作用至关重要的TRMT1亚域。重要的是,斑马鱼体内 TRMT1 的缺失足以诱发发育和行为表型,这些表型再现了在患有致病性 TRMT1 变体的人类患者身上观察到的表型。总之,这些研究结果表明,TRMT1催化的tRNA修饰缺失会导致一种综合症形式的智力残疾,并阐明了致病性TRMT1变体导致的tRNA修饰缺失的分子基础。
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
Biallelic pathogenic variants in TRMT1 disrupt tRNA modification and induce a syndromic neurodevelopmental disorder
The post-transcriptional modification of tRNAs plays a key role in tRNA folding and function to ensure proper levels of protein synthesis during growth and development. Pathogenic variants in tRNA modification enzymes have been implicated in diverse human neurodevelopmental and neurological disorders. However, the molecular basis for many of these disorders remains unknown, thereby limiting our understanding and potential treatment of pathologies linked to tRNA modification. Here, we describe an extensive cohort of 31 individuals from 24 unrelated families with bi-allelic variants in the tRNA methyltransferase 1 (TRMT1) gene who present with a syndromic neurodevelopmental disorder universally characterized by intellectual disability in affected patients. Developmental delay, behavioral abnormalities and facial dysmorphisms represent additional core phenotypes of this syndrome. The variants include novel and ultra-rare TRMT1 variants that segregate with clinical pathology. We found that a subset of variants causes mis-splicing and loss of TRMT1 protein expression. Notably, patient cells with TRMT1 variants exhibit a deficiency in tRNA modifications catalyzed by TRMT1. Molecular analysis of TRMT1 variants reveal distinct regions of the TRMT1 protein required for tRNA modification activity and binding, including a TRMT1 subdomain critical for tRNA interaction. Importantly, depletion of TRMT1 in zebrafish is sufficient to induce developmental and behavioral phenotypes that recapitulate those observed in human patients with pathogenic TRMT1 variants. Altogether, these findings demonstrate that loss of TRMT1-catalyzed tRNA modifications leads to a syndromic form of intellectual disability and elucidate the molecular underpinnings of tRNA modification deficiency caused by pathogenic TRMT1 variants.
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
自引率
0.00%
发文量
0
期刊最新文献
Identifying individuals at risk for surgical supravalvar aortic stenosis by polygenic risk score with graded phenotyping Exome wide association study for blood lipids in 1,158,017 individuals from diverse populations Allelic effects on KLHL17 expression likely mediated by JunB/D underlie a PDAC GWAS signal at chr1p36.33 Genetic associations between SGLT2 inhibition, DPP4 inhibition or GLP1R agonism and prostate cancer risk: a two-sample Mendelian randomisation study A Genome-wide Association Study Identifies Novel Genetic Variants Associated with Knee Pain in the UK Biobank (N = 441,757)
×
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