Enhancer

增强子
  • 文章类型: Journal Article
    Self-transcribing active regulatory region sequencing (STARR-seq) is a high-throughput sequencing method capable of simultaneously discovering and validating all enhancers within the genome. In this method, candidate sequences are inserted into plasmid vectors and electroporated into cells. Acting as both enhancers and target genes, the self-transcription of these sequences will also be enhanced by themselves. By sequencing the transcriptome and comparing the results with the non-inserted control, the locations and activity of enhancers can be determined. In traditional enhancer discovery strategies, the chromatin open regions and transcription active regions were sequenced and predicted as enhancers. However, the activity of these putative enhancers could only be validated one by one without a high-throughput method. STARR-seq solved this limitation, allowing simultaneous enhancers discovery and activity validation in a high-throughput manner. Since the introduction of STARR-seq, it has been widely used to discover enhancers and validate enhancer activity in a number of organisms and cells. In this review, we present the traditional enhancer prediction methods and the basic principles, development history, specific applications of STARR-seq, and its future prospects, aiming to provide a reference for researchers in related fields conducting enhancer studies.
    自转录活性调节区测序(self-transcribing active regulatory region sequencing,STARR-seq)是一种可发现并同时验证全基因组增强子活性的高通量测序方法。其原理为:将待验证序列插入质粒载体并电转入细胞中,该序列在作为增强子提高靶基因转录的同时,其本身也作为靶基因被增强转录。通过对转录组进行测序,并对比未插入片段的测序结果,可获得增强子在基因组位置及活性的信息。在传统增强子研究方法中,通过对染色质开放区域和转录活性区域进行测序以预测增强子,但只能逐一验证预测结果,无法高通量验证增强子活性。STARR-seq技术解决了上述缺陷,可在对全基因组增强子高通量挖掘的同时,对其活性进行可靠的验证。自STARR-seq技术发明以来,已被广泛运用于不同物种与细胞中的增强子发现及活性验证研究。本文对传统增强子预测方法以及STARR-seq技术的基本原理、发展历史和具体运用进行了介绍,并对其发展前景进行展望,以期为后续增强子相关领域研究人员提供参考。.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    小鼠FOXA1和GATA4是先驱因子的原型,通过与ALB1基因增强子中的N1核小体结合来启动肝细胞发育。使用低温电子显微镜(cryo-EM),我们分别和组合确定了游离N1核小体及其与FOXA1和GATA4的复合物的结构。我们发现FOXA1和GATA4的DNA结合域主要识别接头DNA和核小体的内部位点,分别,而它们的内在无序区域与组蛋白H2A-H2B上的酸性斑块相互作用。FOXA1通过重新定位N1核小体有效增强GATA4结合。体内DNA编辑和生物信息学分析表明,FOXA1和GATA4的共结合模式在调节涉及肝细胞功能的基因中起重要作用。我们的结果揭示了FOXA1和GATA4通过核小体重新定位与核小体合作结合的机制,通过弯曲接头DNA和阻碍核小体包装来打开染色质。
    Mouse FOXA1 and GATA4 are prototypes of pioneer factors, initiating liver cell development by binding to the N1 nucleosome in the enhancer of the ALB1 gene. Using cryoelectron microscopy (cryo-EM), we determined the structures of the free N1 nucleosome and its complexes with FOXA1 and GATA4, both individually and in combination. We found that the DNA-binding domains of FOXA1 and GATA4 mainly recognize the linker DNA and an internal site in the nucleosome, respectively, whereas their intrinsically disordered regions interact with the acidic patch on histone H2A-H2B. FOXA1 efficiently enhances GATA4 binding by repositioning the N1 nucleosome. In vivo DNA editing and bioinformatics analyses suggest that the co-binding mode of FOXA1 and GATA4 plays important roles in regulating genes involved in liver cell functions. Our results reveal the mechanism whereby FOXA1 and GATA4 cooperatively bind to the nucleosome through nucleosome repositioning, opening chromatin by bending linker DNA and obstructing nucleosome packing.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    增强子在基因表达调控中至关重要,决定转录活性的特异性和时间,这突出了他们的鉴定对于解开基因调控的复杂性的重要性。因此,确定增强剂及其优势至关重要。基因组中的重复序列是相同或对称片段的重复序列。有大量证据表明,重复序列包含大量的遗传信息。因此,我们介绍W2V重复指数,设计用于鉴定增强子序列片段并通过分析增强子区域中的重复K聚体序列来评估其强度。利用word2vector算法进行数值转换,利用Manta射线觅食优化进行特征选择,该方法有效地捕获了K-mer序列的频率和分布。通过专注于重复的K-mer序列,它最大限度地降低了计算复杂性,并有助于分析较大的K值。实验表明,我们的方法在几乎所有指标上都优于所有其他高级方法。
    Enhancers are crucial in gene expression regulation, dictating the specificity and timing of transcriptional activity, which highlights the importance of their identification for unravelling the intricacies of genetic regulation. Therefore, it is critical to identify enhancers and their strengths. Repeated sequences in the genome are repeats of the same or symmetrical fragments. There has been a great deal of evidence that repetitive sequences contain enormous amounts of genetic information. Thus, We introduce the W2V-Repeated Index, designed to identify enhancer sequence fragments and evaluates their strength through the analysis of repeated K-mer sequences in enhancer regions. Utilizing the word2vector algorithm for numerical conversion and Manta Ray Foraging Optimization for feature selection, this method effectively captures the frequency and distribution of K-mer sequences. By concentrating on repeated K-mer sequences, it minimizes computational complexity and facilitates the analysis of larger K values. Experiments indicate that our method performs better than all other advanced methods on almost all indicators.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    组蛋白修改,被称为组蛋白标记,是调节细胞内基因表达的关键。组蛋白标记的大量潜在组合在仅通过生物学实验方法解码调控机制方面提出了相当大的挑战。为了克服这一挑战,我们开发了一种叫做CatLearning的方法。它利用具有专门适应残差网络的改进的卷积神经网络架构来定量解释组蛋白标记并预测基因表达。该架构集成了高达500Kb的远程组蛋白信息,并在没有3D信息的情况下学习染色质相互作用特征。通过只使用一个组蛋白标记,CatLearning实现了高水平的准确性。此外,CatLearning通过模拟增强子和整个基因组的组蛋白修饰变化来预测基因表达。这些发现有助于理解组蛋白标记的结构,并为具有表观遗传变化的疾病开发诊断和治疗靶标。
    Histone modifications, known as histone marks, are pivotal in regulating gene expression within cells. The vast array of potential combinations of histone marks presents a considerable challenge in decoding the regulatory mechanisms solely through biological experimental approaches. To overcome this challenge, we have developed a method called CatLearning. It utilizes a modified convolutional neural network architecture with a specialized adaptation Residual Network to quantitatively interpret histone marks and predict gene expression. This architecture integrates long-range histone information up to 500Kb and learns chromatin interaction features without 3D information. By using only one histone mark, CatLearning achieves a high level of accuracy. Furthermore, CatLearning predicts gene expression by simulating changes in histone modifications at enhancers and throughout the genome. These findings help comprehend the architecture of histone marks and develop diagnostic and therapeutic targets for diseases with epigenetic changes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    基因打靶(GT)允许精确操作基因组序列,如敲入和序列替换,但是种子植物中的GT仍然是一项具有挑战性的任务。已知工程化的序列特异性核酸酶(SSN)在生物体中通过同源定向修复(HDR)促进GT。这里,我们证明了Cas12a和耐温Cas12a变体(ttCas12a)可以通过顺序转化策略在拟南芥(拟南芥)的两个基因座上有效地建立精确和可遗传的GT。因此,ttCas12a显示出比未修饰的Cas12a更高的GT效率。此外,还研究了通过顺序转化策略对GT的转录和翻译增强子的效率。这些增强剂及其组合有望在顺序转化策略中显示GT效率的增加,类似于以前的一体化战略报告,但只观察到最大两倍的增加。这些结果表明,靶位点的双链断裂(DSB)频率是决定植物中GT遗传效率的最重要因素之一。另一方面,更高的DSB频率并不总是导致更高的GT效率,这表明GT通过HDR需要一些额外的因素。因此,不再期望DSB的增加能提高GT效率,未来需要建立新的战略。这项研究为植物中精确和可遗传的GT技术开辟了广泛的应用。
    Gene targeting (GT) allows precise manipulation of genome sequences, such as knock-ins and sequence substitutions, but GT in seed plants remains a challenging task. Engineered sequence-specific nucleases (SSNs) are known to facilitate GT via homology-directed repair (HDR) in organisms. Here, we demonstrate that Cas12a and a temperature-tolerant Cas12a variant (ttCas12a) can efficiently establish precise and heritable GT at two loci in Arabidopsis thaliana (Arabidopsis) through a sequential transformation strategy. As a result, ttCas12a showed higher GT efficiency than unmodified Cas12a. In addition, the efficiency of transcriptional and translational enhancers for GT via sequential transformation strategy was also investigated. These enhancers and their combinations were expected to show an increase in GT efficiency in the sequential transformation strategy, similar to previous reports of all-in-one strategies, but only a maximum twofold increase was observed. These results indicate that the frequency of double strand breaks (DSBs) at the target site is one of the most important factors determining the efficiency of genetic GT in plants. On the other hand, a higher frequency of DSBs does not always lead to higher efficiency of GT, suggesting that some additional factors are required for GT via HDR. Therefore, the increase in DSB can no longer be expected to improve GT efficiency, and a new strategy needs to be established in the future. This research opens up a wide range of applications for precise and heritable GT technology in plants.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    MDS1和EVI1复合基因座(MECOM),编码几种变体的转录因子,与卵巢癌的进展有关。调节区在卵巢癌中调节MECOM表达的功能尚不完全清楚。在这项研究中,在用溴结构域和外端(BET)抑制剂JQ-1处理的卵巢癌细胞系中评估MECOM表达。使用CCK-8、集落形成、伤口愈合和Transwell。在稳定的sgRNA转染的OVCAR3细胞系中估计致癌表型。通过皮下注射增强子缺失的OVCAR3细胞系来测定异种移植小鼠模型。结果显示MECOM的表达在用JQ-1处理的细胞系中下调。来自3种卵巢癌细胞系中公开的ChIP测序(H3K27Ac)的数据在第一外显子周围显示出潜在的增强子。MECOM增强子缺失后,OVCAR3细胞的mRNA和蛋白表达下调。同样,细胞和异种移植小鼠模型中的致癌表型均显着减弱。本研究表明JQ-1可以抑制MECOM的表达和肿瘤发生。MECOM增强子活性的缺失在抑制卵巢癌进展中具有不可或缺的作用,通过应用这种非编码DNA缺失,这为卵巢癌治疗提供了有希望的机会。
    MDS1 and EVI1 complex locus (MECOM), a transcription factor encoding several variants, has been implicated in progression of ovarian cancer. The function of regulatory regions in regulating MECOM expression in ovarian cancer is not fully understood. In this study, MECOM expression was evaluated in ovarian cancer cell lines treated with bromodomain and extraterminal (BET) inhibitor JQ-1. Oncogenic phenotypes were assayed using assays of CCK-8, colony formation, wound-healing and transwell. Oncogenic phenotypes were estimated in stable sgRNA-transfected OVCAR3 cell lines. Xenograft mouse model was assayed via subcutaneous injection of enhancer-deleted OVCAR3 cell lines. The results displayed that expression of MECOM is downregulated in cell lines treated with JQ-1. Data from published ChIP-sequencing (H3K27Ac) in 3 ovarian cancer cell lines displayed a potential enhancer around the first exon. mRNA and protein expression were downregulated in OVCAR3 cells after deletion of the MECOM enhancer. Similarly, oncogenic phenotypes both in cells and in the xenograft mouse model were significantly attenuated. This study demonstrates that JQ-1 can inhibit the expression of MECOM and tumorigenesis. Deletion of the enhancer activity of MECOM has an indispensable role in inhibiting ovarian cancer progress, which sheds light on a promising opportunity for ovarian cancer treatment through the application of this non-coding DNA deletion.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    背景:识别增强子-启动子相互作用(EPI)对于人类发育至关重要。基因组中的EPI在调节转录中起关键作用。然而,分类EPI的实验方法在努力方面过于昂贵,时间,和资源。因此,越来越多的研究正在开发计算技术,特别是使用深度学习和其他机器学习技术,来解决这些问题。不幸的是,当前的大多数计算方法基于卷积神经网络,递归神经网络,或者它们的组合,它们不考虑上下文细节以及增强子和启动子序列之间的长程相互作用。为了克服上述限制,本研究提出了一种称为EPI-Trans的基于变压器的新模型。变换器模型中的多头注意力机制自动学习代表增强子和启动子序列之间长相互关系的特征。此外,创建具有可转移性的通用模型,该模型可用作各种细胞系的预训练模型。此外,使用特定细胞系数据集对通用模型的参数进行微调以提高性能。
    结果:根据从六个基准细胞系获得的结果,特定的平均AUROC,泛型,最好的型号是94.2%,95%,和95.7%,而平均AUPR为80.5%,66.1%,分别为79.6%。
    结论:本研究提出了一种基于变压器的EPI预测深度学习模型。某些细胞系的比较结果表明,EPI-Trans优于其他尖端技术,可以在识别EPI的挑战中提供卓越的性能。
    BACKGROUND: Recognition of enhancer-promoter Interactions (EPIs) is crucial for human development. EPIs in the genome play a key role in regulating transcription. However, experimental approaches for classifying EPIs are too expensive in terms of effort, time, and resources. Therefore, more and more studies are being done on developing computational techniques, particularly using deep learning and other machine learning techniques, to address such problems. Unfortunately, the majority of current computational methods are based on convolutional neural networks, recurrent neural networks, or a combination of them, which don\'t take into consideration contextual details and the long-range interactions between the enhancer and promoter sequences. A new transformer-based model called EPI-Trans is presented in this study to overcome the aforementioned limitations. The multi-head attention mechanism in the transformer model automatically learns features that represent the long interrelationships between enhancer and promoter sequences. Furthermore, a generic model is created with transferability that can be utilized as a pre-trained model for various cell lines. Moreover, the parameters of the generic model are fine-tuned using a particular cell line dataset to improve performance.
    RESULTS: Based on the results obtained from six benchmark cell lines, the average AUROC for the specific, generic, and best models is 94.2%, 95%, and 95.7%, while the average AUPR is 80.5%, 66.1%, and 79.6% respectively.
    CONCLUSIONS: This study proposed a transformer-based deep learning model for EPI prediction. The comparative results on certain cell lines show that EPI-Trans outperforms other cutting-edge techniques and can provide superior performance on the challenge of recognizing EPI.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:增强子是重要的基因调控元件,可促进发育和疾病中关键基因的表达。异常增强子可以调节癌症风险并激活导致各种癌症发生的癌基因。然而,大多数增强剂在癌症中的潜在机制仍不清楚。这里,我们的目的是探讨黑素瘤中一种关键增强剂的功能和机制。
    方法:应用多组学数据来鉴定与黑素瘤进展有关的增强子(enh17)。为了评估enh17的功能,将CRISPR/Cas9技术应用于黑素瘤细胞系A375中的敲除enh17。RNA-seq,进行与荧光素酶报告基因测定整合的ChIP-seq和Hi-C数据分析以鉴定enh17的潜在靶基因。进行功能实验以进一步验证靶基因ETV4的功能。进行与CUT&Tag测序整合的多组学数据以验证推断的转录因子STAT3的结合谱。
    结果:增强剂,这里叫enh17,被发现异常激活并参与黑色素瘤的进展。CRISPR/Cas9介导的enh17缺失抑制细胞增殖,迁移,和黑色素瘤在体外和体内的肿瘤生长。机械上,我们将ETV4鉴定为受enh17调控的靶基因,功能实验进一步支持ETV4为参与癌症相关表型的靶基因.此外,STAT3作为转录因子与enh17结合以调节ETV4的转录。
    结论:我们的研究结果表明,在黑色素瘤中,enh17具有致癌作用并促进肿瘤进展,其转录调控机制得到了充分阐明,这可能为黑色素瘤的预防和治疗打开一个有希望的窗口。
    BACKGROUND: Enhancers are important gene regulatory elements that promote the expression of critical genes in development and disease. Aberrant enhancer can modulate cancer risk and activate oncogenes that lead to the occurrence of various cancers. However, the underlying mechanism of most enhancers in cancer remains unclear. Here, we aim to explore the function and mechanism of a crucial enhancer in melanoma.
    METHODS: Multi-omics data were applied to identify an enhancer (enh17) involved in melanoma progression. To evaluate the function of enh17, CRISPR/Cas9 technology were applied to knockout enh17 in melanoma cell line A375. RNA-seq, ChIP-seq and Hi-C data analysis integrated with luciferase reporter assay were performed to identify the potential target gene of enh17. Functional experiments were conducted to further validate the function of the target gene ETV4. Multi-omics data integrated with CUT&Tag sequencing were performed to validate the binding profile of the inferred transcription factor STAT3.
    RESULTS: An enhancer, named enh17 here, was found to be aberrantly activated and involved in melanoma progression. CRISPR/Cas9-mediated deletion of enh17 inhibited cell proliferation, migration, and tumor growth of melanoma both in vitro and in vivo. Mechanistically, we identified ETV4 as a target gene regulated by enh17, and functional experiments further support ETV4 as a target gene that is involved in cancer-associated phenotypes. In addition, STAT3 acts as a transcription factor binding with enh17 to regulate the transcription of ETV4.
    CONCLUSIONS: Our findings revealed that enh17 plays an oncogenic role and promotes tumor progression in melanoma, and its transcriptional regulatory mechanisms were fully elucidated, which may open a promising window for melanoma prevention and treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    p53肿瘤抑制因子的异常在肺癌的发生发展中至关重要,p53可以调节靶基因启动子来对抗癌症。最近的研究表明p53与增强子元件广泛结合。然而,p53是否通过塑造增强子景观发挥肿瘤抑制作用仍知之甚少。在目前的研究中,基于我们建立的TP53敲除人支气管上皮细胞(BEAS-2B),我们采用了几种功能基因组学方法来评估整个基因组中p53结合位点的增强子活性.在p53缺失后,共有943种活性常规增强子和370种超增强子(SE)消失了,表明p53调节了数百个增强子元件的活性。我们发现一种p53依赖性SE,位于9号染色体上,命名为KLF4-SE,调节Krüppel样因子4(KLF4)基因的表达。此外,p53缺失显著降低KLF4-SE增强子活性和KLF4表达,但在亚硝胺4-(甲基亚硝胺)-1-(3-吡啶基)-1-丁酮诱导的细胞转化模型中,集落形成能力增加。随后,在TP53敲除细胞中,KLF4的过度表达部分逆转了p53缺乏引起的克隆形成能力的增加。始终如一,KLF4在肺癌组织和细胞系中的表达也降低。KLF4过表达显著抑制肺癌细胞增殖和迁移。总的来说,我们的研究结果表明,p53对增强子形成和活性的调节是p53肿瘤抑制功能的组成部分。因此,我们的发现为p53在肺癌发生中的调控机制提供了新的见解,并为筛选治疗靶点提供了新的策略。
    The abnormality of the p53 tumor suppressor is crucial in lung cancer development, because p53 regulates target gene promoters to combat cancer. Recent studies have shown extensive p53 binding to enhancer elements. However, whether p53 exerts a tumor suppressor role by shaping the enhancer landscape remains poorly understood. In the current study, we employed several functional genomics approaches to assess the enhancer activity at p53 binding sites throughout the genome based on our established TP53 knockout (KO) human bronchial epithelial cells (BEAS-2B). A total of 943 active regular enhancers and 370 super-enhancers (SEs) disappeared upon the deletion of p53, indicating that p53 modulates the activity of hundreds of enhancer elements. We found that one p53-dependent SE, located on chromosome 9 and designated as KLF4-SE, regulated the expression of the Krüppel-like factor 4 ( KLF4) gene. Furthermore, the deletion of p53 significantly decreased the KLF4-SE enhancer activity and the KLF4 expression, but increased colony formation ability in the nitrosamines 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone-induced cell transformation model. Subsequently, in TP53 KO cells, the overexpression of KLF4 partially reversed the increased clonogenic capacity caused by p53 deficiency. Consistently, KLF4 expression also decreased in lung cancer tissues and cell lines. It appeared that overexpression of KLF4 significantly suppressed the proliferation and migration of lung cancer cells. Collectively, our results suggest that the regulation of enhancer formation and activity by p53 is an integral component of the p53 tumor suppressor function. Therefore, our findings offer some novel insights into the regulation mechanism of p53 in lung oncogenesis and introduce a new strategy for screening therapeutic targets.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    基因转录失调是肿瘤的标志,可以作为癌症药物开发的目标。然而,开发针对异常表达转录因子(TFs)的小分子抑制剂极具挑战性,除了TFs的核受体家族.关于胃食管腺癌(GEA)中TF与转录辅因子之间的相互作用或靶向TF和转录辅因子复合物的治疗作用知之甚少。在这项研究中,我们发现,ETS同源因子(EHF)的表达是由核心转录调节电路(CRC),特别是ELF3-KLF5-GATA6,干扰其表达抑制了GEA细胞的恶性生物学行为。重要的是,我们将AjubaLIM蛋白(AJUBA)鉴定为一种新的EHF共激活因子,可协同协调GEA中的转录网络活性.此外,我们确定KRAS信号是EHF和AJUBA下游的共同通路.适用,脂质纳米粒对EHF和AJUBA的双重靶向作用在体外和体内减轻了GEA的恶性生物学行为。总之,EHF被CRC上调,并通过KRAS途径与AJUBA相互作用促进GEA恶性肿瘤。通过脂质纳米粒靶向EHF及其共激活剂AJUBA是一种新的潜在治疗策略。
    Transcriptional dysregulation of genes is a hallmark of tumors and can serve as targets for cancer drug development. However, it is extremely challenging to develop small-molecule inhibitors to target abnormally expressed transcription factors (TFs) except for the nuclear receptor family of TFs. Little is known about the interaction between TFs and transcription cofactors in gastroesophageal adenocarcinoma (GEA) or the therapeutic effects of targeting TF and transcription cofactor complexes. In this study, we found that ETS homologous factor (EHF) expression is promoted by a core transcriptional regulatory circuitry (CRC), specifically ELF3-KLF5-GATA6, and interference with its expression suppressed the malignant biological behavior of GEA cells. Importantly, we identified Ajuba LIM protein (AJUBA) as a new coactivator of EHF that cooperatively orchestrates transcriptional network activity in GEA. Furthermore, we identified KRAS signaling as a common pathway downstream of EHF and AJUBA. Applicably, dual targeting of EHF and AJUBA by lipid nanoparticles cooperatively attenuated the malignant biological behaviors of GEA in vitro and in vivo. In conclusion, EHF is upregulated by the CRC and promotes GEA malignancy by interacting with AJUBA through the KRAS pathway. Targeting of both EHF and its coactivator AJUBA through lipid nanoparticles is a novel potential therapeutic strategy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号