Transcriptional Activation

转录激活
  • 文章类型: Journal Article
    人T细胞白血病病毒1型(HTLV-1)是成人T细胞白血病/淋巴瘤的病原体。HTLV-I的癌基因产品税被认为通过激活生长促进基因来促进病毒感染细胞的增殖,从而在白血病发生中起关键作用。这些基因编码生长因子及其受体,细胞因子,细胞粘附分子,生长信号传感器,转录因子和细胞周期调节因子。我们在这里显示Tax激活编码共激活相关精氨酸甲基转移酶1(CARM1)的基因,通过组蛋白甲基化表观遗传学增强基因表达。Tax激活了Carm1基因并增加了蛋白质表达,不仅在人T细胞系中,而且在正常外周血淋巴细胞(PHA-PBLs)中。税收增加了靶基因IL-2Rα上的R17甲基化组蛋白H3,伴随着CARM1的表达增加。短发夹RNA(shRNA)介导的CARM1敲除降低了Tax介导的IL-2Rα和CyclinD2基因表达的诱导,降低E2F活化并抑制细胞周期进程。税收通过Carm1基因内含子1中的反应元件起作用,通过NF-κB途径。这些结果表明,Tax介导的Carm1基因激活有助于白血病靶基因表达和细胞周期进程,确定Tax介导的反式激活促进细胞生长的第一个表观遗传靶基因。
    Human T-cell leukemia virus type 1 (HTLV-1) is the causative agent of adult T-cell leukemia/lymphoma. The oncogene product Tax of HTLV-I is thought to play crucial roles in leukemogenesis by promoting proliferation of the virus-infected cells through activation of growth-promoting genes. These genes code for growth factors and their receptors, cytokines, cell adhesion molecules, growth signal transducers, transcription factors and cell cycle regulators. We show here that Tax activates the gene coding for coactivator-associated arginine methyltransferase 1 (CARM1), which epigenetically enhances gene expression through methylation of histones. Tax activated the Carm1 gene and increased protein expression, not only in human T-cell lines but also in normal peripheral blood lymphocytes (PHA-PBLs). Tax increased R17-methylated histone H3 on the target gene IL-2Rα, concomitant with increased expression of CARM1. Short hairpin RNA (shRNA)-mediated knockdown of CARM1 decreased Tax-mediated induction of IL-2Rα and Cyclin D2 gene expression, reduced E2F activation and inhibited cell cycle progression. Tax acted via response elements in intron 1 of the Carm1 gene, through the NF-κB pathway. These results suggest that Tax-mediated activation of the Carm1 gene contributes to leukemogenic target-gene expression and cell cycle progression, identifying the first epigenetic target gene for Tax-mediated trans-activation in cell growth promotion.
    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
    合子基因组激活(ZGA)是哺乳动物胚胎发生过程中的关键事件。标志着从母体到合子控制发育的过渡。在以基因表达的复杂级联为特征的ZGA过程中,谁以精心安排的顺序向第一张多米诺骨牌倾斜是最令人感兴趣的主题。最近,Dux,Obox和Nr5a2被鉴定为位于转录层次顶部的先驱转录因子。通过逆转录转座子元件作为转录激活的枢纽的共同选择,这些先驱转录因子重新连接基因调控网络,从而启动ZGA。在这次审查中,我们提供了这些先驱转录因子功能的潜在机制的快照。我们认为ZGA是胚胎自身基因组开始影响发育轨迹的起点,因此,在ZGA期间深入解剖先驱转录因子的功能将成为我们对早期胚胎发育的理解的基石,这将为推进我们对哺乳动物发育生物学的掌握和优化体外生产(IVP)技术铺平道路。
    Zygotic genome activation (ZGA) is a pivotal event in mammalian embryogenesis, marking the transition from maternal to zygotic control of development. During the ZGA process that is characterized by the intricate cascade of gene expression, who tipped the first domino in a meticulously arranged sequence is a subject of paramount interest. Recently, Dux, Obox and Nr5a2 were identified as pioneer transcription factors that reside at the top of transcriptional hierarchy. Through co-option of retrotransposon elements as hubs for transcriptional activation, these pioneer transcription factors rewire the gene regulatory network, thus initiating ZGA. In this review, we provide a snapshot of the mechanisms underlying the functions of these pioneer transcription factors. We propose that ZGA is the starting point where the embryo\'s own genome begins to influence development trajectory, therefore in-depth dissecting the functions of pioneer transcription factors during ZGA will form a cornerstone of our understanding for early embryonic development, which will pave the way for advancing our grasp of mammalian developmental biology and optimizing in vitro production (IVP) techniques.
    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
    帕妥珠单抗(Perjeta®),与HER2(人表皮生长因子受体2)的二聚化臂结合的人源化抗体,作为HER2过表达的恶性肿瘤的单一治疗剂失败。由于HER2与配体结合的EGFR(表皮生长因子受体)的分子相互作用已暗示有丝分裂信号传导和恶性增殖,我们假设这种相互作用,而不是单独的HER2表达和寡聚化,可能是帕妥珠单抗治疗疗效的潜在分子靶标和预测因子。因此,我们使用Förster共振能量转移(FRET)显微镜和荧光相关和交叉相关光谱(FCS/FCCS)研究了HER2+EGFR+SK-BR-3乳腺肿瘤细胞中存在和不存在帕妥珠单抗时,EGF刺激下HER2和EGFR分子之间的静态和动态相互作用.通过蛋白质印迹和MTT测定测量信号传导的相应激活和细胞增殖的变化。HER2扩散的自相关函数最好通过校正三重态形成的三分量模型拟合,在这些成分中,缓慢扩散的膜成分揭示了EGFR配体结合诱导的聚集,如光子计数直方图和共扩散分数所证明。帕妥珠单抗治疗有效地预防了这种聚集,这也抑制了刺激后EGFR和HER2的相互作用,如通过FRET效率的变化监测。总的来说,数据表明,帕妥珠单抗,通过阻碍EGFR和HER2之间的刺激后相互作用,抑制EGFR诱发的HER2聚集和磷酸化,并导致细胞增殖的剂量依赖性降低,特别是当存在较高量的EGF时。因此,我们提出,在预测帕妥珠单抗治疗结果时,可以将HER2阳性肿瘤上的EGFR表达作为一个潜在的生物标志物加以考虑.
    Pertuzumab (Perjeta®), a humanized antibody binding to the dimerization arm of HER2 (Human epidermal growth factor receptor-2), has failed as a monotherapy agent in HER2 overexpressing malignancies. Since the molecular interaction of HER2 with ligand-bound EGFR (epidermal growth factor receptor) has been implied in mitogenic signaling and malignant proliferation, we hypothesized that this interaction, rather than HER2 expression and oligomerization alone, could be a potential molecular target and predictor of the efficacy of pertuzumab treatment. Therefore, we investigated static and dynamic interactions between HER2 and EGFR molecules upon EGF stimulus in the presence and absence of pertuzumab in HER2+ EGFR+ SK-BR-3 breast tumor cells using Förster resonance energy transfer (FRET) microscopy and fluorescence correlation and cross-correlation spectroscopy (FCS/FCCS). The consequential activation of signaling and changes in cell proliferation were measured by Western blotting and MTT assay. The autocorrelation functions of HER2 diffusion were best fitted by a three-component model corrected for triplet formation, and among these components the slowly diffusing membrane component revealed aggregation induced by EGFR ligand binding, as evidenced by photon-counting histograms and co-diffusing fractions. This aggregation has efficiently been prevented by pertuzumab treatment, which also inhibited the post-stimulus interaction of EGFR and HER2, as monitored by changes in FRET efficiency. Overall, the data demonstrated that pertuzumab, by hindering post-stimulus interaction between EGFR and HER2, inhibits EGFR-evoked HER2 aggregation and phosphorylation and leads to a dose-dependent decrease in cell proliferation, particularly when higher amounts of EGF are present. Consequently, we propose that EGFR expression on HER2-positive tumors could be taken into consideration as a potential biomarker when predicting the outcome of pertuzumab 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
    家族性血小板疾病伴髓样恶性肿瘤(FPDMM)是一种由RUNX1中的杂合种系突变引起的常染色体显性疾病。它的特点是血小板减少症,血小板功能障碍,和血液恶性肿瘤的易感性。尽管FPDMM是涉及异常DNA甲基化的疾病的前兆,FPDMM中的DNA甲基化状态仍然未知,主要是由于缺乏动物模型和在获得患者来源的样本方面的挑战。这里,使用基因组编辑技术,我们建立了两系具有不同FPDMM模拟杂合RUNX1突变的人诱导多能干细胞(iPSCs).这些iPSCs显示造血祖细胞(HPCs)和巨核细胞(Mks)的分化缺陷,与FPDMM一致。模拟FPDMM的HPCs显示出不同于野生型HPCs的DNA甲基化模式,高甲基化区域显示ETS转录因子(TF)基序的富集。我们发现,ETS家族成员FLI1的表达,在具有RUNX1反式激活域(TAD)突变的FPDMM模拟HPCs中显著下调。我们证明了FLI1促进了结合位点定向的DNA去甲基化,并且FLI1的过表达恢复了它们的巨核细胞分化效率和高甲基化状态。这些发现表明,FLI1在调节DNA甲基化和纠正具有RUNX1TAD突变的FPDMM模拟HPCs的巨核细胞分化缺陷中起着至关重要的作用。
    Familial platelet disorder with associated myeloid malignancies (FPDMM) is an autosomal dominant disease caused by heterozygous germline mutations in RUNX1. It is characterized by thrombocytopenia, platelet dysfunction, and a predisposition to hematological malignancies. Although FPDMM is a precursor for diseases involving abnormal DNA methylation, the DNA methylation status in FPDMM remains unknown, largely due to a lack of animal models and challenges in obtaining patient-derived samples. Here, using genome editing techniques, we established two lines of human induced pluripotent stem cells (iPSCs) with different FPDMM-mimicking heterozygous RUNX1 mutations. These iPSCs showed defective differentiation of hematopoietic progenitor cells (HPCs) and megakaryocytes (Mks), consistent with FPDMM. The FPDMM-mimicking HPCs showed DNA methylation patterns distinct from those of wild-type HPCs, with hypermethylated regions showing the enrichment of ETS transcription factor (TF) motifs. We found that the expression of FLI1, an ETS family member, was significantly downregulated in FPDMM-mimicking HPCs with a RUNX1 transactivation domain (TAD) mutation. We demonstrated that FLI1 promoted binding-site-directed DNA demethylation, and that overexpression of FLI1 restored their megakaryocytic differentiation efficiency and hypermethylation status. These findings suggest that FLI1 plays a crucial role in regulating DNA methylation and correcting defective megakaryocytic differentiation in FPDMM-mimicking HPCs with a RUNX1 TAD mutation.
    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
    共激活剂相关的精氨酸甲基转移酶1(CARM1)是一种蛋白质精氨酸甲基转移酶,可在组蛋白和非组蛋白底物上沉积不对称的二甲基化标记。CARM1在转录中的调节作用首先在雌激素受体阳性(ER)乳腺癌中得到鉴定。从那以后,对CARM1激活ER靶基因的机制进行了进一步研究.CARM1在ER阴性(ER-)乳腺癌中表达最高,高表达与不良预后相关。提示CARM1的致癌作用。的确,在ER-乳腺癌中,CARM1可以至少部分地通过蛋白质的甲基化和癌基因的激活来促进增殖和转移。在这次审查中,我们总结了CARM1在乳腺癌中的转录激活机制。CARM1的甲基转移酶活性对其许多功能很重要,在这里,我们还强调了CARM1的非酶作用.我们还涵盖了通常在癌症中失调的CARM1调节的生物过程,以及在癌症治疗中利用CARM1的方法。
    Coactivator-associated arginine methyltransferase 1 (CARM1) is a protein arginine methyltransferase that deposits asymmetrical dimethylation marks on both histone and nonhistone substrates. The regulatory role of CARM1 in transcription was first identified in estrogen receptor positive (ER+) breast cancer. Since then, the mechanism of CARM1 in activating ER-target genes has been further interrogated. CARM1 is expressed at the highest level in ER negative (ER-) breast cancer and higher expression correlates with poor prognosis, suggesting an oncogenic role of CARM1. Indeed, in ER- breast cancer, CARM1 can promote proliferation and metastasis at least partly through methylation of proteins and activation of oncogenes. In this review, we summarize the mechanisms of transcriptional activation by CARM1 in breast cancer. The methyltransferase activity of CARM1 is important for many of its functions; here, we also highlight the nonenzymatic roles of CARM1. We also cover the biological processes regulated by CARM1 that are often deregulated in cancer and the ways to harness CARM1 in cancer 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
    基因转移剂(GTA)是不能自我繁殖和具有传染性的噬菌体样颗粒。Caulobactercrescentus,一种被称为模型生物的细菌,用于研究细菌细胞生物学和细胞周期调控,最近已被证明产生真正的GTA颗粒(CcGTA)。由于C.crescentus最终死于释放GTA颗粒,GTA颗粒的产生必须严格调节并与宿主生理学整合以防止细胞群崩溃。CcGTA生物合成基因簇的两个直接激活剂,加菲和加夫,已经被确认,然而,尚不清楚GafYZ如何控制转录或它们如何协调CcGTA基因簇的基因表达与基因组其他地方的其他辅助基因,以实现完整的CcGTA生产。这里,我们表明,CcGTA基因簇被GafY转录共激活,整合宿主因子(IHF),并通过GafZ介导的转录抗终止。我们提供证据表明GafZ是一种转录抗终止子,可能与RNA聚合酶形成抗终止复合物,Nusa,NusG,和NusE绕过14kbCcGTA簇内的转录终止子。总的来说,我们揭示了协调C.crescentus中GTA颗粒合成的两层调控。
    Gene Transfer Agents (GTAs) are phage-like particles that cannot self-multiply and be infectious. Caulobacter crescentus, a bacterium best known as a model organism to study bacterial cell biology and cell cycle regulation, has recently been demonstrated to produce bona fide GTA particles (CcGTA). Since C. crescentus ultimately die to release GTA particles, the production of GTA particles must be tightly regulated and integrated with the host physiology to prevent a collapse in cell population. Two direct activators of the CcGTA biosynthetic gene cluster, GafY and GafZ, have been identified, however, it is unknown how GafYZ controls transcription or how they coordinate gene expression of the CcGTA gene cluster with other accessory genes elsewhere on the genome for complete CcGTA production. Here, we show that the CcGTA gene cluster is transcriptionally co-activated by GafY, integration host factor (IHF), and by GafZ-mediated transcription anti-termination. We present evidence that GafZ is a transcription anti-terminator that likely forms an anti-termination complex with RNA polymerase, NusA, NusG, and NusE to bypass transcription terminators within the 14 kb CcGTA cluster. Overall, we reveal a two-tier regulation that coordinates the synthesis of GTA particles in C. crescentus.
    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
    与其他亚型乳腺癌相比,三阴性乳腺癌(TNBC)更具侵袭性,并且具有更高的转移率。由于缺乏药物靶向受体,化疗是目前TNBC唯一可用的全身治疗方法.然而,部分患者仍可能出现耐药性,预后不良。因此,TNBC患者迫切需要新的分子生物标志物和新的治疗靶点.为了提供对TNBC进展的分子见解,我们研究了含有5个cullinneddylation1结构域(DCUN1D5)在TNBC调控中的功能和潜在机制。通过TCGA数据集和手术标本用免疫组织化学(IHC)染色方法,DCUN1D5在TNBC肿瘤组织中被鉴定为显著上调并且与预后负相关。进行了一系列体外和体内实验以证实DCUN1D5在TNBC中的致癌作用。FN1或PI3K/AKT激活剂IGF-1的过表达可以恢复DCUN1D5敲低诱导的增殖和侵袭能力,DCUN1D5可以作为转录因子阴阳1(YY1)的新转录靶标。总之,YY1增强的DCUN1D5表达可通过FN1/PI3K/AKT途径促进TNBC进展,DCUN1D5可能是TNBC治疗的潜在预后生物标志物和治疗靶点。
    Triple-negative breast cancer (TNBC) is more aggressive and has a higher metastasis rate compared with other subtypes of breast cancer. Due to the lack of drug-targetable receptors, chemotherapy is now the only available systemic treatment for TNBC. However, some patients might still develop drug resistance and have poor prognosis. Therefore, novel molecular biomarkers and new treatment targets are urgently needed for patients with TNBC. To provide molecular insights into TNBC progression, we investigated the function and the underlying mechanism of Defective in cullin neddylation 1 domain containing 5 (DCUN1D5) in the regulation of TNBC. By TCGA dataset and surgical specimens with immunohistochemical (IHC) staining method, DCUN1D5 was identified to be significantly upregulated in TNBC tumor tissues and negatively associated with prognosis. A series of in vitro and in vivo experiments were performed to confirm the oncogenic role of DCUN1D5 in TNBC. Overexpression of FN1 or PI3K/AKT activator IGF-1 could restore the proliferative and invasive ability induced by DCUN1D5 knockdown and DCUN1D5 could act as a novel transcriptional target of transcription factor Yin Yang 1 (YY1). In conclusion, YY1-enhanced DCUN1D5 expression could promote TNBC progression by FN1/PI3K/AKT pathway and DCUN1D5 might be a potential prognostic biomarker and therapeutic target for TNBC 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
    哺乳动物SWI/SNF样BAF复合物在动物发育和病理状况中起关键作用。先前的基因缺失研究和人基因突变的表征暗示复合物既抑制又激活大量基因。然而,由于基因缺失或自然突变的相对长期性质,复合物在细胞中的直接功能仍不清楚。在这里,我们通过将生长素诱导的Degron标签(AID)敲入Smarca4基因来产生小鼠系,其编码BAF复合物的必需ATP酶亚基BRG1。我们表明,在CD4+T细胞中,通过osTIR1表达和生长素处理6至10小时,标记的BRG1可以有效地耗尽。肝细胞,和从敲入小鼠中分离的成纤维细胞。BRG1的急性耗尽导致新生RNA和RNA聚合酶II在大量基因上的结合减少,与BRG1的丢失呈正相关。Further,这些变化与DNaseI超敏位点(DHSs)和p300结合的可及性降低相关。急性BRG1耗竭导致核小体移位的三种主要模式,导致转录因子基序周围以及增强子和转录起始位点(TSS)处的核小体间距较窄。这与BRG1的丢失,染色质可及性降低和新生RNA减少有关。BRG1的急性消耗严重损害了曲古抑菌素A(TSA)诱导的组蛋白乙酰化,表明BRG1的染色质重塑活性和组蛋白乙酰化之间存在实质性的相互作用。我们的数据表明,BRG1主要在染色质可及性方面发挥直接的积极作用,RNAPII结合,通过调节核小体定位并促进转录因子与其靶位点的结合来产生新生RNA。
    The mammalian SWI/SNF-like BAF complexes play critical roles during animal development and pathological conditions. Previous gene deletion studies and characterization of human gene mutations implicate that the complexes both repress and activate a large number of genes. However, the direct function of the complexes in cells remains largely unclear due to the relatively long-term nature of gene deletion or natural mutation. Here we generate a mouse line by knocking in the auxin-inducible degron tag (AID) to the Smarca4 gene, which encodes BRG1, the essential ATPase subunit of the BAF complexes. We show that the tagged BRG1 can be efficiently depleted by osTIR1 expression and auxin treatment for 6 to 10 h in CD4 + T cells, hepatocytes, and fibroblasts isolated from the knock-in mice. The acute depletion of BRG1 leads to decreases in nascent RNAs and RNA polymerase II binding at a large number of genes, which are positively correlated with the loss of BRG1. Further, these changes are correlated with diminished accessibility at DNase I Hypersensitive Sites (DHSs) and p300 binding. The acute BRG1 depletion results in three major patterns of nucleosome shifts leading to narrower nucleosome spacing surrounding transcription factor motifs and at enhancers and transcription start sites (TSSs), which are correlated with loss of BRG1, decreased chromatin accessibility and decreased nascent RNAs. Acute depletion of BRG1 severely compromises the Trichostatin A (TSA) -induced histone acetylation, suggesting a substantial interplay between the chromatin remodeling activity of BRG1 and histone acetylation. Our data suggest BRG1 mainly plays a direct positive role in chromatin accessibility, RNAPII binding, and nascent RNA production by regulating nucleosome positioning and facilitating transcription factor binding to their target sites.
    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
    基于CRISPR的基因治疗提供疾病相关基因序列的精确靶向和特异性编辑。可能产生持久的治疗效果。然而,高效交付仍然是其广泛应用的重大挑战。在这项研究中,我们设计了一种名为TSPscp的新型短肽偶联生物可还原聚合物作为CRISPR系统安全有效的递送载体。我们的结果表明,TSPscp显着增强了多个CRISPR系统的转录激活和基因组编辑活性,如分解-seq和Deep-seq所证实的。这是由于其通过促进细胞摄取和溶酶体逃逸来促进质粒DNA递送的能力。此外,TSPscp通过递送微环DNA进一步增强CRISPR的基因组编辑,常规质粒DNA的浓缩形式。更重要的是,TSPscp显著改善体内CRISPR系统的递送和基因组编辑。总之,我们的研究强调了TSPscp作为CRISPR体内应用的一种有前景的递送工具.
    CRISPR-based gene therapy offers precise targeting and specific editing of disease-related gene sequences, potentially yielding long-lasting treatment effects. However, efficient delivery remains a significant challenge for its widespread application. In this study, we design a novel short peptide-conjugated bioreducible polymer named TSPscp as a safe and effective delivery vector for the CRISPR system. Our results show that TSPscp markedly boosts transcriptional activation and genome editing activities of multiple CRISPR systems as confirmed by decomposition-seq and Deep-seq, which is resulted from its capability in facilitating delivery of plasmid DNA by promoting cellular uptake and lysosomal escape. Additionally, TSPscp further enhances genome editing of CRISPR by delivery of minicircle DNA, a condensed form of regular plasmid DNA. More importantly, TSPscp significantly improves delivery and genome editing of CRISPR system in vivo. In summary, our study highlights TSPscp as a promising delivery tool for CRISPR applications in vivo.
    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
    增强子如何在3D染色质组织的背景下调节其靶基因已被广泛研究,并且最近出现了不需要增强子-启动子直接接触的模型。这里,我们使用乳腺癌细胞系中雌激素受体依赖性增强子的激活来研究两个位点的增强子-启动子通讯。这允许从激素刺激到有效基因激活的分子事件的高时间分辨率跟踪。我们研究了如何通过DNA荧光原位杂交测定两个增强子-启动子空间接近度,以及染色质原位片段化和邻近连接产生的接触频率,在增强子驱动的基因激活过程中动态变化。这些正交方法产生看似矛盾的结果:在增强子激活后,增强子-启动子接触频率增加,而空间接近度降低。我们使用不同的雌激素受体配体和转录抑制剂来探索这种明显的差异。我们的数据表明,增强子-启动子接触频率与转录无关,而增强子-启动子接近度的改变取决于转录。我们的结果强调了接触频率和细胞核中物理距离之间的关系,特别是在短基因组距离上,并不总是简单的。
    How enhancers regulate their target genes in the context of 3D chromatin organization is extensively studied and models which do not require direct enhancer-promoter contact have recently emerged. Here, we use the activation of estrogen receptor-dependent enhancers in a breast cancer cell line to study enhancer-promoter communication at two loci. This allows high temporal resolution tracking of molecular events from hormone stimulation to efficient gene activation. We examine how both enhancer-promoter spatial proximity assayed by DNA fluorescence in situ hybridization, and contact frequencies resulting from chromatin in situ fragmentation and proximity ligation, change dynamically during enhancer-driven gene activation. These orthogonal methods produce seemingly paradoxical results: upon enhancer activation enhancer-promoter contact frequencies increase while spatial proximity decreases. We explore this apparent discrepancy using different estrogen receptor ligands and transcription inhibitors. Our data demonstrate that enhancer-promoter contact frequencies are transcription independent whereas altered enhancer-promoter proximity depends on transcription. Our results emphasize that the relationship between contact frequencies and physical distance in the nucleus, especially over short genomic distances, is not always a simple one.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号