在分化的 LUHMES 中整合 CTCF 循环、甲基组和转录组,作为人类神经元中 15q11-q13 位点印记动态的模型。

IF 3.1 2区 生物学 Q3 BIOCHEMISTRY & MOLECULAR BIOLOGY Human molecular genetics Pub Date : 2024-09-19 DOI:10.1093/hmg/ddae111
Orangel J Gutierrez Fugón, Osman Sharifi, Nicholas Heath, Daniela C Soto, J Antonio Gomez, Dag H Yasui, Aron Judd P Mendiola, Henriette O'Geen, Ulrika Beitnere, Marketa Tomkova, Viktoria Haghani, Greg Dillon, David J Segal, Janine M LaSalle
{"title":"在分化的 LUHMES 中整合 CTCF 循环、甲基组和转录组,作为人类神经元中 15q11-q13 位点印记动态的模型。","authors":"Orangel J Gutierrez Fugón, Osman Sharifi, Nicholas Heath, Daniela C Soto, J Antonio Gomez, Dag H Yasui, Aron Judd P Mendiola, Henriette O'Geen, Ulrika Beitnere, Marketa Tomkova, Viktoria Haghani, Greg Dillon, David J Segal, Janine M LaSalle","doi":"10.1093/hmg/ddae111","DOIUrl":null,"url":null,"abstract":"<p><p>Human cell line models, including the neuronal precursor line LUHMES, are important for investigating developmental transcriptional dynamics within imprinted regions, particularly the 15q11-q13 Angelman (AS) and Prader-Willi (PWS) syndrome locus. AS results from loss of maternal UBE3A in neurons, where the paternal allele is silenced by a convergent antisense transcript UBE3A-ATS, a lncRNA that terminates at PWAR1 in non-neurons. qRT-PCR analysis confirmed the exclusive and progressive increase in UBE3A-ATS in differentiating LUHMES neurons, validating their use for studying UBE3A silencing. Genome-wide transcriptome analyses revealed changes to 11 834 genes during neuronal differentiation, including the upregulation of most genes within the 15q11-q13 locus. To identify dynamic changes in chromatin loops linked to transcriptional activity, we performed a HiChIP validated by 4C, which identified two neuron-specific CTCF loops between MAGEL2-SNRPN and PWAR1-UBE3A. To determine if allele-specific differentially methylated regions (DMR) may be associated with CTCF loop anchors, whole genome long-read nanopore sequencing was performed. We identified a paternally hypomethylated DMR near the SNRPN upstream loop anchor exclusive to neurons and a paternally hypermethylated DMR near the PWAR1 CTCF anchor exclusive to undifferentiated cells, consistent with increases in neuronal transcription. Additionally, DMRs near CTCF loop anchors were observed in both cell types, indicative of allele-specific differences in chromatin loops regulating imprinted transcription. These results provide an integrated view of the 15q11-q13 epigenetic landscape during LUHMES neuronal differentiation, underscoring the complex interplay of transcription, chromatin looping, and DNA methylation. They also provide insights for future therapeutic approaches for AS and PWS.</p>","PeriodicalId":13070,"journal":{"name":"Human molecular genetics","volume":null,"pages":null},"PeriodicalIF":3.1000,"publicationDate":"2024-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11413648/pdf/","citationCount":"0","resultStr":"{\"title\":\"Integration of CTCF loops, methylome, and transcriptome in differentiating LUHMES as a model for imprinting dynamics of the 15q11-q13 locus in human neurons.\",\"authors\":\"Orangel J Gutierrez Fugón, Osman Sharifi, Nicholas Heath, Daniela C Soto, J Antonio Gomez, Dag H Yasui, Aron Judd P Mendiola, Henriette O'Geen, Ulrika Beitnere, Marketa Tomkova, Viktoria Haghani, Greg Dillon, David J Segal, Janine M LaSalle\",\"doi\":\"10.1093/hmg/ddae111\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><p>Human cell line models, including the neuronal precursor line LUHMES, are important for investigating developmental transcriptional dynamics within imprinted regions, particularly the 15q11-q13 Angelman (AS) and Prader-Willi (PWS) syndrome locus. AS results from loss of maternal UBE3A in neurons, where the paternal allele is silenced by a convergent antisense transcript UBE3A-ATS, a lncRNA that terminates at PWAR1 in non-neurons. qRT-PCR analysis confirmed the exclusive and progressive increase in UBE3A-ATS in differentiating LUHMES neurons, validating their use for studying UBE3A silencing. Genome-wide transcriptome analyses revealed changes to 11 834 genes during neuronal differentiation, including the upregulation of most genes within the 15q11-q13 locus. To identify dynamic changes in chromatin loops linked to transcriptional activity, we performed a HiChIP validated by 4C, which identified two neuron-specific CTCF loops between MAGEL2-SNRPN and PWAR1-UBE3A. To determine if allele-specific differentially methylated regions (DMR) may be associated with CTCF loop anchors, whole genome long-read nanopore sequencing was performed. We identified a paternally hypomethylated DMR near the SNRPN upstream loop anchor exclusive to neurons and a paternally hypermethylated DMR near the PWAR1 CTCF anchor exclusive to undifferentiated cells, consistent with increases in neuronal transcription. Additionally, DMRs near CTCF loop anchors were observed in both cell types, indicative of allele-specific differences in chromatin loops regulating imprinted transcription. These results provide an integrated view of the 15q11-q13 epigenetic landscape during LUHMES neuronal differentiation, underscoring the complex interplay of transcription, chromatin looping, and DNA methylation. They also provide insights for future therapeutic approaches for AS and PWS.</p>\",\"PeriodicalId\":13070,\"journal\":{\"name\":\"Human molecular genetics\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":3.1000,\"publicationDate\":\"2024-09-19\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11413648/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Human molecular genetics\",\"FirstCategoryId\":\"99\",\"ListUrlMain\":\"https://doi.org/10.1093/hmg/ddae111\",\"RegionNum\":2,\"RegionCategory\":\"生物学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q3\",\"JCRName\":\"BIOCHEMISTRY & MOLECULAR BIOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Human molecular genetics","FirstCategoryId":"99","ListUrlMain":"https://doi.org/10.1093/hmg/ddae111","RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q3","JCRName":"BIOCHEMISTRY & MOLECULAR BIOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

包括神经元前体系 LUHMES 在内的人类细胞系模型对于研究印记区内的发育转录动态非常重要,尤其是 15q11-q13 Angelman(AS)和 Prader-Willi(PWS)综合征基因座。AS是神经元中母系UBE3A缺失的结果,父系等位基因被收敛的反义转录本UBE3A-ATS沉默,UBE3A-ATS是一种lncRNA,在非神经元中终止于PWAR1。全基因组转录组分析揭示了神经元分化过程中11 834个基因的变化,包括15q11-q13位点内大多数基因的上调。为了确定与转录活性相关的染色质环路的动态变化,我们进行了经 4C 验证的 HiChIP,在 MAGEL2-SNRPN 和 PWAR1-UBE3A 之间发现了两个神经元特异性 CTCF 环路。为了确定等位基因特异性差异甲基化区域(DMR)是否可能与 CTCF 环锚相关,我们进行了全基因组长读程纳米孔测序。我们在 SNRPN 上游环路锚附近发现了神经元独有的父系低甲基化 DMR,在 PWAR1 CTCF 锚附近发现了未分化细胞独有的父系高甲基化 DMR,这与神经元转录的增加是一致的。此外,在这两种细胞类型中都观察到了 CTCF 环锚附近的 DMRs,这表明调节印记转录的染色质环存在等位基因特异性差异。这些结果提供了 LUHMES 神经元分化过程中 15q11-q13 表观遗传景观的综合视图,强调了转录、染色质环和 DNA 甲基化之间复杂的相互作用。这些结果还为未来的AS和PWS治疗方法提供了启示。
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
Integration of CTCF loops, methylome, and transcriptome in differentiating LUHMES as a model for imprinting dynamics of the 15q11-q13 locus in human neurons.

Human cell line models, including the neuronal precursor line LUHMES, are important for investigating developmental transcriptional dynamics within imprinted regions, particularly the 15q11-q13 Angelman (AS) and Prader-Willi (PWS) syndrome locus. AS results from loss of maternal UBE3A in neurons, where the paternal allele is silenced by a convergent antisense transcript UBE3A-ATS, a lncRNA that terminates at PWAR1 in non-neurons. qRT-PCR analysis confirmed the exclusive and progressive increase in UBE3A-ATS in differentiating LUHMES neurons, validating their use for studying UBE3A silencing. Genome-wide transcriptome analyses revealed changes to 11 834 genes during neuronal differentiation, including the upregulation of most genes within the 15q11-q13 locus. To identify dynamic changes in chromatin loops linked to transcriptional activity, we performed a HiChIP validated by 4C, which identified two neuron-specific CTCF loops between MAGEL2-SNRPN and PWAR1-UBE3A. To determine if allele-specific differentially methylated regions (DMR) may be associated with CTCF loop anchors, whole genome long-read nanopore sequencing was performed. We identified a paternally hypomethylated DMR near the SNRPN upstream loop anchor exclusive to neurons and a paternally hypermethylated DMR near the PWAR1 CTCF anchor exclusive to undifferentiated cells, consistent with increases in neuronal transcription. Additionally, DMRs near CTCF loop anchors were observed in both cell types, indicative of allele-specific differences in chromatin loops regulating imprinted transcription. These results provide an integrated view of the 15q11-q13 epigenetic landscape during LUHMES neuronal differentiation, underscoring the complex interplay of transcription, chromatin looping, and DNA methylation. They also provide insights for future therapeutic approaches for AS and PWS.

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
Human molecular genetics
Human molecular genetics 生物-生化与分子生物学
CiteScore
6.90
自引率
2.90%
发文量
294
审稿时长
2-4 weeks
期刊介绍: Human Molecular Genetics concentrates on full-length research papers covering a wide range of topics in all aspects of human molecular genetics. These include: the molecular basis of human genetic disease developmental genetics cancer genetics neurogenetics chromosome and genome structure and function therapy of genetic disease stem cells in human genetic disease and therapy, including the application of iPS cells genome-wide association studies mouse and other models of human diseases functional genomics computational genomics In addition, the journal also publishes research on other model systems for the analysis of genes, especially when there is an obvious relevance to human genetics.
期刊最新文献
Investigating MATN3 and ASPN as novel drivers of gastric cancer progression via EMT pathways. Exploring the complexity of systemic sclerosis etiology by trio whole genome sequencing. A novel copy number variant in the murine Cdh23 gene gives rise to profound deafness and vestibular dysfunction. Joint models reveal genetic architecture of pubertal stage transitions and their association with BMI in admixed Chilean population. Neurodevelopmental disorder-associated CYFIP2 regulates membraneless organelles and eIF2α phosphorylation via protein interactors and actin cytoskeleton.
×
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