首页 > 最新文献

Nucleic Acids Research最新文献

英文 中文
A deep learning-based method enables the automatic and accurate assembly of chromosome-level genomes 基于深度学习的方法可自动准确地组装染色体级基因组
IF 14.9 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-09-17 DOI: 10.1093/nar/gkae789
Zijie Jiang, Zhixiang Peng, Zhaoyuan Wei, Jiahe Sun, Yongjiang Luo, Lingzi Bie, Guoqing Zhang, Yi Wang
The application of high-throughput chromosome conformation capture (Hi-C) technology enables the construction of chromosome-level assemblies. However, the correction of errors and the anchoring of sequences to chromosomes in the assembly remain significant challenges. In this study, we developed a deep learning-based method, AutoHiC, to address the challenges in chromosome-level genome assembly by enhancing contiguity and accuracy. Conventional Hi-C-aided scaffolding often requires manual refinement, but AutoHiC instead utilizes Hi-C data for automated workflows and iterative error correction. When trained on data from 300+ species, AutoHiC demonstrated a robust average error detection accuracy exceeding 90%. The benchmarking results confirmed its significant impact on genome contiguity and error correction. The innovative approach and comprehensive results of AutoHiC constitute a breakthrough in automated error detection, promising more accurate genome assemblies for advancing genomics research.
高通量染色体构象捕获(Hi-C)技术的应用使染色体级组装的构建成为可能。然而,装配中的错误纠正和序列与染色体的锚定仍然是重大挑战。在这项研究中,我们开发了一种基于深度学习的方法 AutoHiC,通过提高连续性和准确性来应对染色体级基因组组装中的挑战。传统的Hi-C辅助脚手架通常需要人工完善,而AutoHiC则利用Hi-C数据进行自动工作流和迭代纠错。在对来自 300 多个物种的数据进行训练后,AutoHiC 显示出超过 90% 的强大平均错误检测准确率。基准测试结果证实了它对基因组连续性和纠错的重大影响。AutoHiC 的创新方法和全面结果是自动错误检测领域的一个突破,有望为推进基因组学研究提供更准确的基因组组装。
{"title":"A deep learning-based method enables the automatic and accurate assembly of chromosome-level genomes","authors":"Zijie Jiang, Zhixiang Peng, Zhaoyuan Wei, Jiahe Sun, Yongjiang Luo, Lingzi Bie, Guoqing Zhang, Yi Wang","doi":"10.1093/nar/gkae789","DOIUrl":"https://doi.org/10.1093/nar/gkae789","url":null,"abstract":"The application of high-throughput chromosome conformation capture (Hi-C) technology enables the construction of chromosome-level assemblies. However, the correction of errors and the anchoring of sequences to chromosomes in the assembly remain significant challenges. In this study, we developed a deep learning-based method, AutoHiC, to address the challenges in chromosome-level genome assembly by enhancing contiguity and accuracy. Conventional Hi-C-aided scaffolding often requires manual refinement, but AutoHiC instead utilizes Hi-C data for automated workflows and iterative error correction. When trained on data from 300+ species, AutoHiC demonstrated a robust average error detection accuracy exceeding 90%. The benchmarking results confirmed its significant impact on genome contiguity and error correction. The innovative approach and comprehensive results of AutoHiC constitute a breakthrough in automated error detection, promising more accurate genome assemblies for advancing genomics research.","PeriodicalId":19471,"journal":{"name":"Nucleic Acids Research","volume":null,"pages":null},"PeriodicalIF":14.9,"publicationDate":"2024-09-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142236671","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
How to scan naked DNA using promiscuous recognition and no clamping: a model for pioneer transcription factors 如何利用杂乱识别和无钳位扫描裸 DNA:先驱转录因子模型
IF 14.9 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-09-17 DOI: 10.1093/nar/gkae790
Rama Reddy Goluguri, Catherine Ghosh, Joshua Quintong, Mourad Sadqi, Victor Muñoz
Most DNA scanning proteins uniquely recognize their cognate sequence motif and slide on DNA assisted by some sort of clamping interface. The pioneer transcription factors that control cell fate in eukaryotes must forgo both elements to gain access to DNA in naked and chromatin forms; thus, whether or how these factors scan naked DNA is unknown. Here, we use single-molecule techniques to investigate naked DNA scanning by the Engrailed homeodomain (enHD) as paradigm of highly promiscuous recognition and open DNA binding interface. We find that enHD scans naked DNA quite effectively, and about 200000-fold faster than expected for a continuous promiscuous slide. To do so, enHD scans about 675 bp of DNA in 100 ms and then redeploys stochastically to another location 530 bp afar in just 10 ms. During the scanning phase enHD alternates between slow- and medium-paced modes every 3 and 40 ms, respectively. We also find that enHD binds nucleosomes and does so with enhanced affinity relative to naked DNA. Our results demonstrate that pioneer-like transcription factors can in principle do both, target nucleosomes and scan active DNA efficiently. The hybrid scanning mechanism used by enHD appears particularly well suited for the highly complex genomic signals of eukaryotic cells.
大多数 DNA 扫描蛋白都能独特地识别它们的同源序列图案,并在某种夹持界面的辅助下在 DNA 上滑动。在真核生物中,控制细胞命运的先驱转录因子必须放弃这两种元素,才能获得裸DNA和染色质形式的DNA;因此,这些因子是否或如何扫描裸DNA尚不清楚。在这里,我们利用单分子技术研究了裸DNA扫描的啮合同源结构域(enHD),它是高度杂乱识别和开放式DNA结合界面的典范。我们发现,enHD 扫描裸 DNA 的效率相当高,比预期的连续杂交滑动快约 20 万倍。为此,enHD 在 100 毫秒内扫描约 675 bp 的 DNA,然后在 10 毫秒内随机重新部署到 530 bp 之外的另一个位置。在扫描阶段,enHD 分别每 3 毫秒和 40 毫秒交替使用慢速和中速模式。我们还发现,enHD 能与核小体结合,而且与裸 DNA 相比亲和力更强。我们的研究结果表明,类先驱转录因子原则上可以同时针对核小体和有效扫描活性 DNA。enHD使用的混合扫描机制似乎特别适合真核细胞高度复杂的基因组信号。
{"title":"How to scan naked DNA using promiscuous recognition and no clamping: a model for pioneer transcription factors","authors":"Rama Reddy Goluguri, Catherine Ghosh, Joshua Quintong, Mourad Sadqi, Victor Muñoz","doi":"10.1093/nar/gkae790","DOIUrl":"https://doi.org/10.1093/nar/gkae790","url":null,"abstract":"Most DNA scanning proteins uniquely recognize their cognate sequence motif and slide on DNA assisted by some sort of clamping interface. The pioneer transcription factors that control cell fate in eukaryotes must forgo both elements to gain access to DNA in naked and chromatin forms; thus, whether or how these factors scan naked DNA is unknown. Here, we use single-molecule techniques to investigate naked DNA scanning by the Engrailed homeodomain (enHD) as paradigm of highly promiscuous recognition and open DNA binding interface. We find that enHD scans naked DNA quite effectively, and about 200000-fold faster than expected for a continuous promiscuous slide. To do so, enHD scans about 675 bp of DNA in 100 ms and then redeploys stochastically to another location 530 bp afar in just 10 ms. During the scanning phase enHD alternates between slow- and medium-paced modes every 3 and 40 ms, respectively. We also find that enHD binds nucleosomes and does so with enhanced affinity relative to naked DNA. Our results demonstrate that pioneer-like transcription factors can in principle do both, target nucleosomes and scan active DNA efficiently. The hybrid scanning mechanism used by enHD appears particularly well suited for the highly complex genomic signals of eukaryotic cells.","PeriodicalId":19471,"journal":{"name":"Nucleic Acids Research","volume":null,"pages":null},"PeriodicalIF":14.9,"publicationDate":"2024-09-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142236664","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
4CAC: 4-class classifier of metagenome contigs using machine learning and assembly graphs 4CAC:利用机器学习和装配图对元基因组等位基因进行四级分类
IF 14.9 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-09-17 DOI: 10.1093/nar/gkae799
Lianrong Pu, Ron Shamir
Microbial communities usually harbor a mix of bacteria, archaea, plasmids, viruses and microeukaryotes. Within these communities, viruses, plasmids, and microeukaryotes coexist in relatively low abundance, yet they engage in intricate interactions with bacteria. Moreover, viruses and plasmids, as mobile genetic elements, play important roles in horizontal gene transfer and the development of antibiotic resistance within microbial populations. However, due to the difficulty of identifying viruses, plasmids, and microeukaryotes in microbial communities, our understanding of these minor classes lags behind that of bacteria and archaea. Recently, several classifiers have been developed to separate one or more minor classes from bacteria and archaea in metagenome assemblies. However, these classifiers often overlook the issue of class imbalance, leading to low precision in identifying the minor classes. Here, we developed a classifier called 4CAC that is able to identify viruses, plasmids, microeukaryotes, and prokaryotes simultaneously from metagenome assemblies. 4CAC generates an initial four-way classification using several sequence length-adjusted XGBoost models and further improves the classification using the assembly graph. Evaluation on simulated and real metagenome datasets demonstrates that 4CAC substantially outperforms existing classifiers and combinations thereof on short reads. On long reads, it also shows an advantage unless the abundance of the minor classes is very low. 4CAC runs 1–2 orders of magnitude faster than the other classifiers. The 4CAC software is available at https://github.com/Shamir-Lab/4CAC.
微生物群落通常混合了细菌、古菌、质粒、病毒和微真核细胞。在这些群落中,病毒、质粒和微真核细胞以相对较低的丰度共存,但它们与细菌之间存在着错综复杂的相互作用。此外,病毒和质粒作为可移动的遗传因子,在微生物种群内的水平基因转移和抗生素耐药性的产生中发挥着重要作用。然而,由于难以识别微生物群落中的病毒、质粒和微真核细胞,我们对这些小类的了解落后于对细菌和古细菌的了解。最近,人们开发了几种分类器,将元基因组组装中的一个或多个小类从细菌和古细菌中分离出来。然而,这些分类器往往忽略了类不平衡的问题,导致识别小类的精度很低。在这里,我们开发了一种名为 4CAC 的分类器,它能同时从元基因组集合中识别病毒、质粒、微真核细胞和原核细胞。4CAC 利用几个序列长度调整的 XGBoost 模型生成初始的四向分类,并利用组装图进一步改进分类。在模拟和真实元基因组数据集上进行的评估表明,4CAC 在短读数上的表现大大优于现有的分类器及其组合。在长读数上,除非小类的丰度非常低,否则它也显示出优势。4CAC 的运行速度比其他分类器快 1-2 个数量级。4CAC 软件可在 https://github.com/Shamir-Lab/4CAC 上获取。
{"title":"4CAC: 4-class classifier of metagenome contigs using machine learning and assembly graphs","authors":"Lianrong Pu, Ron Shamir","doi":"10.1093/nar/gkae799","DOIUrl":"https://doi.org/10.1093/nar/gkae799","url":null,"abstract":"Microbial communities usually harbor a mix of bacteria, archaea, plasmids, viruses and microeukaryotes. Within these communities, viruses, plasmids, and microeukaryotes coexist in relatively low abundance, yet they engage in intricate interactions with bacteria. Moreover, viruses and plasmids, as mobile genetic elements, play important roles in horizontal gene transfer and the development of antibiotic resistance within microbial populations. However, due to the difficulty of identifying viruses, plasmids, and microeukaryotes in microbial communities, our understanding of these minor classes lags behind that of bacteria and archaea. Recently, several classifiers have been developed to separate one or more minor classes from bacteria and archaea in metagenome assemblies. However, these classifiers often overlook the issue of class imbalance, leading to low precision in identifying the minor classes. Here, we developed a classifier called 4CAC that is able to identify viruses, plasmids, microeukaryotes, and prokaryotes simultaneously from metagenome assemblies. 4CAC generates an initial four-way classification using several sequence length-adjusted XGBoost models and further improves the classification using the assembly graph. Evaluation on simulated and real metagenome datasets demonstrates that 4CAC substantially outperforms existing classifiers and combinations thereof on short reads. On long reads, it also shows an advantage unless the abundance of the minor classes is very low. 4CAC runs 1–2 orders of magnitude faster than the other classifiers. The 4CAC software is available at https://github.com/Shamir-Lab/4CAC.","PeriodicalId":19471,"journal":{"name":"Nucleic Acids Research","volume":null,"pages":null},"PeriodicalIF":14.9,"publicationDate":"2024-09-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142236667","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Crystal structure and nucleic acid binding mode of CPV NSP9: implications for viroplasm in Reovirales CPV NSP9 的晶体结构和核酸结合模式:对 Reovirales 中病毒质的影响
IF 14.9 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-09-17 DOI: 10.1093/nar/gkae803
Yeda Wang, Hangtian Guo, Yuhao Lu, Wanbin Yang, Tinghan Li, Xiaoyun Ji
Cytoplasmic polyhedrosis viruses (CPVs), like other members of the order Reovirales, produce viroplasms, hubs of viral assembly that shield them from host immunity. Our study investigates the potential role of NSP9, a nucleic acid-binding non-structural protein encoded by CPVs, in viroplasm biogenesis. We determined the crystal structure of the NSP9 core (NSP9ΔC), which shows a dimeric organization topologically similar to the P9-1 homodimers of plant reoviruses. The disordered C-terminal region of NSP9 facilitates oligomerization but is dispensable for nucleic acid binding. NSP9 robustly binds to single- and double-stranded nucleic acids, regardless of RNA or DNA origin. Mutagenesis studies further confirmed that the dimeric form of NSP9 is critical for nucleic acid binding due to positively charged residues that form a tunnel during homodimerization. Gel migration assays reveal a unique nucleic acid binding pattern, with the sequential appearance of two distinct complexes dependent on protein concentration. The similar gel migration pattern shared by NSP9 and rotavirus NSP3, coupled with its structural resemblance to P9-1, hints at a potential role in translational regulation or viral genome packaging, which may be linked to viroplasm. This study advances our understanding of viroplasm biogenesis and Reovirales replication, providing insights into potential antiviral drug targets.
细胞质多角体病毒(CPVs)与Reovirales目其他成员一样,会产生病毒体,病毒体是病毒组装的枢纽,能使病毒免受宿主免疫的影响。我们的研究调查了 CPV 编码的核酸结合非结构蛋白 NSP9 在病毒体生物发生过程中的潜在作用。我们测定了 NSP9 核心(NSP9ΔC)的晶体结构,它显示出与植物再病毒的 P9-1 同源二聚体拓扑结构相似的二聚体组织。NSP9 的无序 C 端区域有利于寡聚化,但对核酸结合来说是不可或缺的。NSP9 可与单链和双链核酸牢固结合,无论其来源是 RNA 还是 DNA。突变研究进一步证实,NSP9 的二聚体形式对核酸结合至关重要,因为带正电荷的残基在同源二聚体化过程中形成了一条隧道。凝胶迁移试验揭示了一种独特的核酸结合模式,两种不同的复合物依次出现,取决于蛋白质的浓度。NSP9 和轮状病毒 NSP3 有着相似的凝胶迁移模式,再加上其结构与 P9-1 相似,这暗示了它在翻译调控或病毒基因组包装中的潜在作用,而这可能与病毒质体有关。这项研究增进了我们对病毒质生物发生和轮状病毒复制的了解,为潜在的抗病毒药物靶点提供了启示。
{"title":"Crystal structure and nucleic acid binding mode of CPV NSP9: implications for viroplasm in Reovirales","authors":"Yeda Wang, Hangtian Guo, Yuhao Lu, Wanbin Yang, Tinghan Li, Xiaoyun Ji","doi":"10.1093/nar/gkae803","DOIUrl":"https://doi.org/10.1093/nar/gkae803","url":null,"abstract":"Cytoplasmic polyhedrosis viruses (CPVs), like other members of the order Reovirales, produce viroplasms, hubs of viral assembly that shield them from host immunity. Our study investigates the potential role of NSP9, a nucleic acid-binding non-structural protein encoded by CPVs, in viroplasm biogenesis. We determined the crystal structure of the NSP9 core (NSP9ΔC), which shows a dimeric organization topologically similar to the P9-1 homodimers of plant reoviruses. The disordered C-terminal region of NSP9 facilitates oligomerization but is dispensable for nucleic acid binding. NSP9 robustly binds to single- and double-stranded nucleic acids, regardless of RNA or DNA origin. Mutagenesis studies further confirmed that the dimeric form of NSP9 is critical for nucleic acid binding due to positively charged residues that form a tunnel during homodimerization. Gel migration assays reveal a unique nucleic acid binding pattern, with the sequential appearance of two distinct complexes dependent on protein concentration. The similar gel migration pattern shared by NSP9 and rotavirus NSP3, coupled with its structural resemblance to P9-1, hints at a potential role in translational regulation or viral genome packaging, which may be linked to viroplasm. This study advances our understanding of viroplasm biogenesis and Reovirales replication, providing insights into potential antiviral drug targets.","PeriodicalId":19471,"journal":{"name":"Nucleic Acids Research","volume":null,"pages":null},"PeriodicalIF":14.9,"publicationDate":"2024-09-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142236668","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Transcription start site scanning requires the fungi-specific hydrophobic loop of Tfb3 转录起始位点扫描需要 Tfb3 真菌特有的疏水环
IF 14.9 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-09-17 DOI: 10.1093/nar/gkae805
Chun Yang, Pratik Basnet, Samah Sharmin, Hui Shen, Craig D Kaplan, Kenji Murakami
RNA polymerase II (pol II) initiates transcription from transcription start sites (TSSs) located ∼30–35 bp downstream of the TATA box in metazoans, whereas in the yeast Saccharomyces cerevisiae, pol II scans further downstream TSSs located ∼40–120 bp downstream of the TATA box. Previously, we found that removal of the kinase module TFIIK (Kin28–Ccl1–Tfb3) from TFIIH shifts the TSS in a yeast in vitro system upstream to the location observed in metazoans and that addition of recombinant Tfb3 back to TFIIH-ΔTFIIK restores the downstream TSS usage. Here, we report that this biochemical activity of yeast TFIIK in TSS scanning is attributable to the Tfb3 RING domain at the interface with pol II in the pre-initiation complex (PIC): especially, swapping Tfb3 Pro51—a residue conserved among all fungi—with Ala or Ser as in MAT1, the metazoan homolog of Tfb3, confers an upstream TSS shift in vitro in a similar manner to the removal of TFIIK. Yeast genetic analysis suggests that both Pro51 and Arg64 of Tfb3 are required to maintain the stability of the Tfb3–pol II interface in the PIC. Cryo-electron microscopy analysis of a yeast PIC lacking TFIIK reveals considerable variability in the orientation of TFIIH, which impairs TSS scanning after promoter opening.
在类人猿中,RNA聚合酶II(pol II)从位于TATA框下游30-35 bp的转录起始位点(TSS)启动转录,而在酵母菌中,pol II扫描位于TATA框下游40-120 bp的更下游的TSS。此前,我们发现从 TFIIH 中移除激酶模块 TFIIK(Kin28-Ccl1-Tfb3)后,酵母体外系统中的 TSS 会向上游移动,移至元古动物体内观察到的位置,而将重组 Tfb3 添加回 TFIIH-ΔTFIIK 后,下游 TSS 的使用会恢复。在这里,我们报告了酵母 TFIIK 在 TSS 扫描中的这种生化活性可归因于预启动复合体(PIC)中与 pol II 接口的 Tfb3 RING 结构域:特别是,将 Tfb3 的 Pro51(所有真菌中都保留的残基)与 Ala 或 Ser(如在 MAT1(Tfb3 的后生同源物)中)互换,可在体外以类似于移除 TFIIK 的方式产生上游 TSS 偏移。酵母遗传分析表明,Tfb3的Pro51和Arg64都是维持PIC中Tfb3-pol II界面稳定性所必需的。对缺乏 TFIIK 的酵母 PIC 进行的冷冻电镜分析表明,TFIIH 的取向存在很大差异,这影响了启动子开启后的 TSS 扫描。
{"title":"Transcription start site scanning requires the fungi-specific hydrophobic loop of Tfb3","authors":"Chun Yang, Pratik Basnet, Samah Sharmin, Hui Shen, Craig D Kaplan, Kenji Murakami","doi":"10.1093/nar/gkae805","DOIUrl":"https://doi.org/10.1093/nar/gkae805","url":null,"abstract":"RNA polymerase II (pol II) initiates transcription from transcription start sites (TSSs) located ∼30–35 bp downstream of the TATA box in metazoans, whereas in the yeast Saccharomyces cerevisiae, pol II scans further downstream TSSs located ∼40–120 bp downstream of the TATA box. Previously, we found that removal of the kinase module TFIIK (Kin28–Ccl1–Tfb3) from TFIIH shifts the TSS in a yeast in vitro system upstream to the location observed in metazoans and that addition of recombinant Tfb3 back to TFIIH-ΔTFIIK restores the downstream TSS usage. Here, we report that this biochemical activity of yeast TFIIK in TSS scanning is attributable to the Tfb3 RING domain at the interface with pol II in the pre-initiation complex (PIC): especially, swapping Tfb3 Pro51—a residue conserved among all fungi—with Ala or Ser as in MAT1, the metazoan homolog of Tfb3, confers an upstream TSS shift in vitro in a similar manner to the removal of TFIIK. Yeast genetic analysis suggests that both Pro51 and Arg64 of Tfb3 are required to maintain the stability of the Tfb3–pol II interface in the PIC. Cryo-electron microscopy analysis of a yeast PIC lacking TFIIK reveals considerable variability in the orientation of TFIIH, which impairs TSS scanning after promoter opening.","PeriodicalId":19471,"journal":{"name":"Nucleic Acids Research","volume":null,"pages":null},"PeriodicalIF":14.9,"publicationDate":"2024-09-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142236665","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
G-quadruplexes in an SVA retrotransposon cause aberrant TAF1 gene expression in X-linked dystonia parkinsonism SVA逆转录质子中的G-四叠体导致X连锁肌张力障碍性帕金森病中TAF1基因的异常表达
IF 14.9 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-09-17 DOI: 10.1093/nar/gkae797
Giulia Nicoletto, Marianna Terreri, Ilaria Maurizio, Emanuela Ruggiero, Filippo M Cernilogar, Christine A Vaine, Maria Vittoria Cottini, Irina Shcherbakova, Ellen B Penney, Irene Gallina, David Monchaud, D Cristopher Bragg, Gunnar Schotta, Sara N Richter
G-quadruplexes (G4s) are non-canonical nucleic acid structures that form in guanine (G)-rich genomic regions. X-linked dystonia parkinsonism (XDP) is an inherited neurodegenerative disease in which a SINE–VNTR–Alu (SVA) retrotransposon, characterised by amplification of a G-rich repeat, is inserted into the coding sequence of TAF1, a key partner of RNA polymerase II. XDP SVA alters TAF1 expression, but the cause of this outcome in XDP remains unknown. To assess whether G4s form in XDP SVA and affect TAF1 expression, we first characterised bioinformatically predicted XDP SVA G4s in vitro. We next showed that highly stable G4s can form and stop polymerase amplification at the SVA region from patient-derived fibroblasts and neural progenitor cells. Using chromatin immunoprecipitazion (ChIP) with an anti-G4 antibody coupled to sequencing or quantitative PCR, we showed that XDP SVA G4s are folded even when embedded in a chromatin context in patient-derived cells. Using the G4 ligands BRACO-19 and quarfloxin and total RNA-sequencing analysis, we showed that stabilisation of the XDP SVA G4s reduces TAF1 transcripts downstream and around the SVA, and increases upstream transcripts, while destabilisation using the G4 unfolder PhpC increases TAF1 transcripts. Our data indicate that G4 formation in the XDP SVA is a major cause of aberrant TAF1 expression, opening the way for the development of strategies to unfold G4s and potentially target the disease.
G-quadruplexes (G4s) 是在富含鸟嘌呤 (G) 的基因组区域形成的非经典核酸结构。X-连锁肌张力障碍性帕金森病(XDP)是一种遗传性神经退行性疾病,在这种疾病中,一个SINE-VNTR-Alu(SVA)反转座子插入到RNA聚合酶II的关键伙伴TAF1的编码序列中,该反转座子的特点是富含G的重复扩增。XDP SVA改变了TAF1的表达,但在XDP中造成这种结果的原因仍不清楚。为了评估 G4 是否在 XDP SVA 中形成并影响 TAF1 的表达,我们首先对生物信息学预测的 XDP SVA G4 进行了体外鉴定。接下来,我们从患者来源的成纤维细胞和神经祖细胞中发现,高度稳定的 G4s 可以在 SVA 区域形成并阻止聚合酶扩增。通过使用抗 G4 抗体进行染色质免疫沉淀(ChIP),并结合测序或定量 PCR,我们发现 XDP SVA G4 即使嵌入患者来源细胞的染色质中也会折叠。通过使用 G4 配体 BRACO-19 和 quarfloxin 以及总 RNA 序列分析,我们发现稳定 XDP SVA G4s 会减少 SVA 下游和周围的 TAF1 转录本,并增加上游转录本,而使用 G4 解除older PhpC 破坏稳定则会增加 TAF1 转录本。我们的数据表明,XDP SVA 中 G4 的形成是 TAF1 表达异常的主要原因,这为开发 G4 的解稳策略和潜在的靶向疾病开辟了道路。
{"title":"G-quadruplexes in an SVA retrotransposon cause aberrant TAF1 gene expression in X-linked dystonia parkinsonism","authors":"Giulia Nicoletto, Marianna Terreri, Ilaria Maurizio, Emanuela Ruggiero, Filippo M Cernilogar, Christine A Vaine, Maria Vittoria Cottini, Irina Shcherbakova, Ellen B Penney, Irene Gallina, David Monchaud, D Cristopher Bragg, Gunnar Schotta, Sara N Richter","doi":"10.1093/nar/gkae797","DOIUrl":"https://doi.org/10.1093/nar/gkae797","url":null,"abstract":"G-quadruplexes (G4s) are non-canonical nucleic acid structures that form in guanine (G)-rich genomic regions. X-linked dystonia parkinsonism (XDP) is an inherited neurodegenerative disease in which a SINE–VNTR–Alu (SVA) retrotransposon, characterised by amplification of a G-rich repeat, is inserted into the coding sequence of TAF1, a key partner of RNA polymerase II. XDP SVA alters TAF1 expression, but the cause of this outcome in XDP remains unknown. To assess whether G4s form in XDP SVA and affect TAF1 expression, we first characterised bioinformatically predicted XDP SVA G4s in vitro. We next showed that highly stable G4s can form and stop polymerase amplification at the SVA region from patient-derived fibroblasts and neural progenitor cells. Using chromatin immunoprecipitazion (ChIP) with an anti-G4 antibody coupled to sequencing or quantitative PCR, we showed that XDP SVA G4s are folded even when embedded in a chromatin context in patient-derived cells. Using the G4 ligands BRACO-19 and quarfloxin and total RNA-sequencing analysis, we showed that stabilisation of the XDP SVA G4s reduces TAF1 transcripts downstream and around the SVA, and increases upstream transcripts, while destabilisation using the G4 unfolder PhpC increases TAF1 transcripts. Our data indicate that G4 formation in the XDP SVA is a major cause of aberrant TAF1 expression, opening the way for the development of strategies to unfold G4s and potentially target the disease.","PeriodicalId":19471,"journal":{"name":"Nucleic Acids Research","volume":null,"pages":null},"PeriodicalIF":14.9,"publicationDate":"2024-09-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142236669","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Using nucleolytic toxins as restriction enzymes enables new RNA applications 使用核酸溶解毒素作为限制酶可实现新的 RNA 应用
IF 14.9 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-09-14 DOI: 10.1093/nar/gkae779
Ulli Rothweiler, Sigurd Eidem Gundesø, Emma Wu Mikalsen, Steingrim Svenning, Mahavir Singh, Francis Combes, Frida J Pettersson, Antonia Mangold, Yvonne Piotrowski, Felix Schwab, Olav Lanes, Bernd Ketelsen Striberny
Over the past five decades, DNA restriction enzymes have revolutionized biotechnology. While these enzymes are widely used in DNA research and DNA engineering, the emerging field of RNA and mRNA therapeutics requires sequence-specific RNA endoribonucleases. Here, we describe EcoToxN1, a member of the type III toxin-antitoxin family of sequence-specific RNA endoribonucleases, and its use in RNA and mRNA analysis. This enzyme recognizes a specific pentamer in a single-stranded RNA and cleaves the RNA within this sequence. The enzyme is neither dependent on annealing of guide RNA or DNA oligos to the template nor does it require magnesium. Furthermore, it performs over a wide range of temperatures. With its unique functions and characteristics, EcoToxN1 can be classified as an RNA restriction enzyme. EcoToxN1 enables new workflows in RNA analysis and biomanufacturing, meeting the demand for faster, cheaper, and more robust analysis methods.
过去五十年来,DNA 限制酶彻底改变了生物技术。这些酶被广泛应用于 DNA 研究和 DNA 工程,而新兴的 RNA 和 mRNA 治疗领域则需要序列特异的 RNA 内切核酸酶。在这里,我们介绍了序列特异性 RNA 内切核酸酶 III 型毒素-抗毒素家族的成员 EcoToxN1 及其在 RNA 和 mRNA 分析中的应用。这种酶能识别单链 RNA 中的特定五聚体,并在此序列内切割 RNA。这种酶既不依赖于引导 RNA 或 DNA 寡聚物与模板的退火,也不需要镁。此外,它的工作温度范围很广。凭借其独特的功能和特性,EcoToxN1 可被归类为一种 RNA 限制酶。EcoToxN1 为 RNA 分析和生物制造带来了新的工作流程,满足了对更快、更便宜、更可靠的分析方法的需求。
{"title":"Using nucleolytic toxins as restriction enzymes enables new RNA applications","authors":"Ulli Rothweiler, Sigurd Eidem Gundesø, Emma Wu Mikalsen, Steingrim Svenning, Mahavir Singh, Francis Combes, Frida J Pettersson, Antonia Mangold, Yvonne Piotrowski, Felix Schwab, Olav Lanes, Bernd Ketelsen Striberny","doi":"10.1093/nar/gkae779","DOIUrl":"https://doi.org/10.1093/nar/gkae779","url":null,"abstract":"Over the past five decades, DNA restriction enzymes have revolutionized biotechnology. While these enzymes are widely used in DNA research and DNA engineering, the emerging field of RNA and mRNA therapeutics requires sequence-specific RNA endoribonucleases. Here, we describe EcoToxN1, a member of the type III toxin-antitoxin family of sequence-specific RNA endoribonucleases, and its use in RNA and mRNA analysis. This enzyme recognizes a specific pentamer in a single-stranded RNA and cleaves the RNA within this sequence. The enzyme is neither dependent on annealing of guide RNA or DNA oligos to the template nor does it require magnesium. Furthermore, it performs over a wide range of temperatures. With its unique functions and characteristics, EcoToxN1 can be classified as an RNA restriction enzyme. EcoToxN1 enables new workflows in RNA analysis and biomanufacturing, meeting the demand for faster, cheaper, and more robust analysis methods.","PeriodicalId":19471,"journal":{"name":"Nucleic Acids Research","volume":null,"pages":null},"PeriodicalIF":14.9,"publicationDate":"2024-09-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142233309","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Advancing microRNA target site prediction with transformer and base-pairing patterns 利用转换器和碱基配对模式推进 microRNA 目标位点预测
IF 14.9 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-09-14 DOI: 10.1093/nar/gkae782
Yue Bi, Fuyi Li, Cong Wang, Tong Pan, Chen Davidovich, Geoffrey I Webb, Jiangning Song
MicroRNAs (miRNAs) are short non-coding RNAs involved in various cellular processes, playing a crucial role in gene regulation. Identifying miRNA targets remains a central challenge and is pivotal for elucidating the complex gene regulatory networks. Traditional computational approaches have predominantly focused on identifying miRNA targets through perfect Watson–Crick base pairings within the seed region, referred to as canonical sites. However, emerging evidence suggests that perfect seed matches are not a prerequisite for miRNA-mediated regulation, underscoring the importance of also recognizing imperfect, or non-canonical, sites. To address this challenge, we propose Mimosa, a new computational approach that employs the Transformer framework to enhance the prediction of miRNA targets. Mimosa distinguishes itself by integrating contextual, positional and base-pairing information to capture in-depth attributes, thereby improving its predictive capabilities. Its unique ability to identify non-canonical base-pairing patterns makes Mimosa a standout model, reducing the reliance on pre-selecting candidate targets. Mimosa achieves superior performance in gene-level predictions and also shows impressive performance in site-level predictions across various non-human species through extensive benchmarking tests. To facilitate research efforts in miRNA targeting, we have developed an easy-to-use web server for comprehensive end-to-end predictions, which is publicly available at http://monash.bioweb.cloud.edu.au/Mimosa.
微RNA(miRNA)是参与各种细胞过程的非编码短RNA,在基因调控中发挥着至关重要的作用。鉴定 miRNA 靶标仍然是一项核心挑战,对于阐明复杂的基因调控网络至关重要。传统的计算方法主要侧重于通过种子区内完美的沃森-克里克碱基配对(称为典型位点)来鉴定 miRNA 靶点。然而,新出现的证据表明,完美的种子配对并不是 miRNA 介导调控的先决条件,这凸显了识别不完美或非规范位点的重要性。为了应对这一挑战,我们提出了一种新的计算方法--Mimosa,它采用 Transformer 框架来加强 miRNA 靶点的预测。Mimosa 通过整合上下文、位置和碱基配对信息来捕捉深度属性,从而提高其预测能力。Mimosa 独有的识别非经典碱基配对模式的能力使其成为一个杰出的模型,减少了对预选候选靶标的依赖。通过广泛的基准测试,Mimosa 在基因水平预测方面表现出色,在各种非人类物种的位点水平预测方面也表现不俗。为了促进 miRNA 靶向的研究工作,我们开发了一个易于使用的网络服务器,用于进行全面的端到端预测,该服务器可在 http://monash.bioweb.cloud.edu.au/Mimosa 上公开获取。
{"title":"Advancing microRNA target site prediction with transformer and base-pairing patterns","authors":"Yue Bi, Fuyi Li, Cong Wang, Tong Pan, Chen Davidovich, Geoffrey I Webb, Jiangning Song","doi":"10.1093/nar/gkae782","DOIUrl":"https://doi.org/10.1093/nar/gkae782","url":null,"abstract":"MicroRNAs (miRNAs) are short non-coding RNAs involved in various cellular processes, playing a crucial role in gene regulation. Identifying miRNA targets remains a central challenge and is pivotal for elucidating the complex gene regulatory networks. Traditional computational approaches have predominantly focused on identifying miRNA targets through perfect Watson–Crick base pairings within the seed region, referred to as canonical sites. However, emerging evidence suggests that perfect seed matches are not a prerequisite for miRNA-mediated regulation, underscoring the importance of also recognizing imperfect, or non-canonical, sites. To address this challenge, we propose Mimosa, a new computational approach that employs the Transformer framework to enhance the prediction of miRNA targets. Mimosa distinguishes itself by integrating contextual, positional and base-pairing information to capture in-depth attributes, thereby improving its predictive capabilities. Its unique ability to identify non-canonical base-pairing patterns makes Mimosa a standout model, reducing the reliance on pre-selecting candidate targets. Mimosa achieves superior performance in gene-level predictions and also shows impressive performance in site-level predictions across various non-human species through extensive benchmarking tests. To facilitate research efforts in miRNA targeting, we have developed an easy-to-use web server for comprehensive end-to-end predictions, which is publicly available at http://monash.bioweb.cloud.edu.au/Mimosa.","PeriodicalId":19471,"journal":{"name":"Nucleic Acids Research","volume":null,"pages":null},"PeriodicalIF":14.9,"publicationDate":"2024-09-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142233311","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Accelerated discovery and miniaturization of novel single-stranded cytidine deaminases 加速发现新型单链胞苷脱氨酶并使其微型化
IF 14.9 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-09-14 DOI: 10.1093/nar/gkae800
Jiacheng Deng, Xueyuan Li, Hao Yu, Lin Yang, Ziru Wang, Wenfeng Yi, Ying Liu, Wenyu Xiao, Hongyong Xiang, Zicong Xie, Dongmei Lv, Hongsheng Ouyang, Daxin Pang, Hongming Yuan
Cytidine base editors (CBEs) hold significant potential in genetic disease treatment and in breeding superior traits into animals. However, their large protein sizes limit their delivery by adeno-associated virus (AAV), given its packing capacity of <4.7 kb. To overcome this, we employed a web-based fast generic discovery (WFG) strategy, identifying several small ssDNA deaminases (Sdds) and constructing multiple Sdd-CBE 1.0 versions. SflSdd-CBE 1.0 demonstrated high C-to-T editing efficiency, comparable to AncBE4max, while SviSdd-CBE 1.0 exhibited moderate C-to-T editing efficiency with a narrow editing window (C3 to C5). Utilizing AlphaFold2, we devised a one-step miniaturization strategy, reducing the size of Sdds while preserving their efficiency. Notably, we administered AAV8 expressing PCSK9 targeted sgRNA and SflSdd-CBEs (nSaCas9) 2.0 into mice, leading to gene-editing events (with editing efficiency up to 15%) and reduced serum cholesterol levels, underscoring the potential of Sdds in gene therapy. These findings offer new single-stranded editing tools for the treatment of rare genetic diseases.
胞嘧啶碱基编辑器(CBEs)在遗传疾病治疗和动物优良性状培育方面具有巨大潜力。然而,由于腺相关病毒(AAV)的包装容量为 4.7 kb,其巨大的蛋白质尺寸限制了其通过腺相关病毒(AAV)的传输。为了克服这一问题,我们采用了基于网络的快速通用发现(WFG)策略,确定了几种小型 ssDNA 去氨酶(Sdds),并构建了多个 Sdd-CBE 1.0 版本。SflSdd-CBE 1.0 表现出很高的 C-T 编辑效率,与 AncBE4max 相当,而 SviSdd-CBE 1.0 表现出中等的 C-T 编辑效率,编辑窗口较窄(C3 到 C5)。利用 AlphaFold2,我们设计了一种一步到位的微型化策略,在保持其效率的同时缩小了 Sdds 的尺寸。值得注意的是,我们给小鼠注射了表达 PCSK9 靶向 sgRNA 和 SflSdd-CBEs (nSaCas9)2.0 的 AAV8,导致了基因编辑事件(编辑效率高达 15%)和血清胆固醇水平的降低,突出了 Sdds 在基因治疗中的潜力。这些发现为治疗罕见遗传病提供了新的单链编辑工具。
{"title":"Accelerated discovery and miniaturization of novel single-stranded cytidine deaminases","authors":"Jiacheng Deng, Xueyuan Li, Hao Yu, Lin Yang, Ziru Wang, Wenfeng Yi, Ying Liu, Wenyu Xiao, Hongyong Xiang, Zicong Xie, Dongmei Lv, Hongsheng Ouyang, Daxin Pang, Hongming Yuan","doi":"10.1093/nar/gkae800","DOIUrl":"https://doi.org/10.1093/nar/gkae800","url":null,"abstract":"Cytidine base editors (CBEs) hold significant potential in genetic disease treatment and in breeding superior traits into animals. However, their large protein sizes limit their delivery by adeno-associated virus (AAV), given its packing capacity of <4.7 kb. To overcome this, we employed a web-based fast generic discovery (WFG) strategy, identifying several small ssDNA deaminases (Sdds) and constructing multiple Sdd-CBE 1.0 versions. SflSdd-CBE 1.0 demonstrated high C-to-T editing efficiency, comparable to AncBE4max, while SviSdd-CBE 1.0 exhibited moderate C-to-T editing efficiency with a narrow editing window (C3 to C5). Utilizing AlphaFold2, we devised a one-step miniaturization strategy, reducing the size of Sdds while preserving their efficiency. Notably, we administered AAV8 expressing PCSK9 targeted sgRNA and SflSdd-CBEs (nSaCas9) 2.0 into mice, leading to gene-editing events (with editing efficiency up to 15%) and reduced serum cholesterol levels, underscoring the potential of Sdds in gene therapy. These findings offer new single-stranded editing tools for the treatment of rare genetic diseases.","PeriodicalId":19471,"journal":{"name":"Nucleic Acids Research","volume":null,"pages":null},"PeriodicalIF":14.9,"publicationDate":"2024-09-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142233312","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Striving for clarity in language about gene expression 努力提高基因表达语言的清晰度
IF 14.9 2区 生物学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-09-14 DOI: 10.1093/nar/gkae764
Ana S G Cunningham, Myriam Gorospe
What do we mean when we say ‘gene expression’? In the decades following Crick's 1958 central dogma of molecular biology, whereby genetic information flows from DNA (genes) to RNA (transcripts) to protein (products), we have learned a great deal about DNA, RNA, proteins, and the ensuing phenotypic changes. With the advent of high-throughput technologies (1990s), molecular biologists and computer scientists forged critical collaborations to understand the vast amount of data being generated, rapidly escalating gene expression research to the ‘omics’ level: entire sets of genes (genomes), transcribed RNAs (transcriptomes), and synthesized proteins (proteomes). However, some concessions came to be made for molecular biologists and computer scientists to understand each other—one of the most prevalent being the increasingly widespread use of ‘gene’ to mean ‘RNAs originating from a DNA segment’. This loosening of terminology, we will argue, creates ambiguity and confusion. We propose guidelines to increase precision and clarity when communicating about gene expression, most notably to reserve ‘gene’ for the DNA template and ‘transcript’ for the RNA transcribed from that gene. Striving to use perspicuous terminology will promote rigorous gene expression science and accelerate discovery in this highly promising area of biology.
我们说 "基因表达 "是什么意思?克里克于 1958 年提出了分子生物学的核心教条,即遗传信息从 DNA(基因)流向 RNA(转录本)再流向蛋白质(产物),在此之后的几十年里,我们已经对 DNA、RNA、蛋白质以及随之而来的表型变化有了大量了解。随着高通量技术的出现(20 世纪 90 年代),分子生物学家和计算机科学家建立了重要的合作关系,以了解正在产生的大量数据,从而将基因表达研究迅速提升到 "全微观 "水平:整套基因(基因组)、转录 RNA(转录组)和合成蛋白质(蛋白质组)。然而,为了让分子生物学家和计算机科学家相互理解,他们不得不做出一些让步--其中最普遍的让步就是越来越广泛地使用 "基因 "来指代 "源自 DNA 片段的 RNA"。我们认为,这种术语上的松动造成了歧义和混淆。我们提出了在交流基因表达时提高准确性和清晰度的指导原则,其中最值得注意的是将 "基因 "保留给 DNA 模板,而将 "转录本 "保留给从该基因转录的 RNA。努力使用清晰的术语将促进严谨的基因表达科学,并加速这一极具潜力的生物学领域的发现。
{"title":"Striving for clarity in language about gene expression","authors":"Ana S G Cunningham, Myriam Gorospe","doi":"10.1093/nar/gkae764","DOIUrl":"https://doi.org/10.1093/nar/gkae764","url":null,"abstract":"What do we mean when we say ‘gene expression’? In the decades following Crick's 1958 central dogma of molecular biology, whereby genetic information flows from DNA (genes) to RNA (transcripts) to protein (products), we have learned a great deal about DNA, RNA, proteins, and the ensuing phenotypic changes. With the advent of high-throughput technologies (1990s), molecular biologists and computer scientists forged critical collaborations to understand the vast amount of data being generated, rapidly escalating gene expression research to the ‘omics’ level: entire sets of genes (genomes), transcribed RNAs (transcriptomes), and synthesized proteins (proteomes). However, some concessions came to be made for molecular biologists and computer scientists to understand each other—one of the most prevalent being the increasingly widespread use of ‘gene’ to mean ‘RNAs originating from a DNA segment’. This loosening of terminology, we will argue, creates ambiguity and confusion. We propose guidelines to increase precision and clarity when communicating about gene expression, most notably to reserve ‘gene’ for the DNA template and ‘transcript’ for the RNA transcribed from that gene. Striving to use perspicuous terminology will promote rigorous gene expression science and accelerate discovery in this highly promising area of biology.","PeriodicalId":19471,"journal":{"name":"Nucleic Acids Research","volume":null,"pages":null},"PeriodicalIF":14.9,"publicationDate":"2024-09-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142233313","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Nucleic Acids Research
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
×
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
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