TFIIIC

TFIIIC
  • 文章类型: Journal Article
    RNA聚合酶III(RNAPIII)合成小的必需非编码RNA分子,例如tRNA和5SrRNA。在酵母和脊椎动物中,RNAPIII需要一般转录因子TFIIIA,TFIIIB,和TFIIC启动转录。TFIIC,由六个亚基组成,与RNAPIII依赖性基因中的内部启动子元件结合。关于锥虫原生动物锥虫和主要利什曼原虫RNAPIII转录的信息有限,早期与真核细胞谱系不同。对首次发布的锥虫基因组序列草案的分析未能识别任何TFIIIC亚基的直向同源物,表明这种转录因子在这些寄生虫中不存在。然而,最近在数据库中注释了一个假定的TFIIIC亚基。在这里,我们描述了该亚基在T.brucei和L.major中的特征,并证明它对应于Tau95。计算机模拟分析表明,两种蛋白质都具有典型的Tau95序列:DNA结合区和二聚化结构域。正如预期的转录因子,Tau95以两种寄生虫的昆虫形式定位于细胞核。染色质免疫沉淀(ChIP)试验证明Tau95与布鲁氏菌中的tRNA和U2snRNA基因结合。值得注意的是,通过进行串联亲和纯化,我们在布鲁氏菌和主要品系中鉴定了TFIIIC亚基Tau55,Tau131和Tau138的直向同源物。因此,与假设相反,锥虫寄生虫确实具有TFIIIC复合物。Tau95的其他推定的相互作用伴侣在布鲁氏菌和L.major中鉴定出。关键点:•四亚基TFIIIC复合物存在于T.brucei和L.major•TbTau95与tRNA和U2snRNA基因相关•Tau95的推定相互作用伴侣可能包括一些RNAPII调节因子。
    RNA polymerase III (RNAP III) synthetizes small essential non-coding RNA molecules such as tRNAs and 5S rRNA. In yeast and vertebrates, RNAP III needs general transcription factors TFIIIA, TFIIIB, and TFIIIC to initiate transcription. TFIIIC, composed of six subunits, binds to internal promoter elements in RNAP III-dependent genes. Limited information is available about RNAP III transcription in the trypanosomatid protozoa Trypanosoma brucei and Leishmania major, which diverged early from the eukaryotic lineage. Analyses of the first published draft of the trypanosomatid genome sequences failed to recognize orthologs of any of the TFIIIC subunits, suggesting that this transcription factor is absent in these parasites. However, a putative TFIIIC subunit was recently annotated in the databases. Here we characterize this subunit in T. brucei and L. major and demonstrate that it corresponds to Tau95. In silico analyses showed that both proteins possess the typical Tau95 sequences: the DNA binding region and the dimerization domain. As anticipated for a transcription factor, Tau95 localized to the nucleus in insect forms of both parasites. Chromatin immunoprecipitation (ChIP) assays demonstrated that Tau95 binds to tRNA and U2 snRNA genes in T. brucei. Remarkably, by performing tandem affinity purifications we identified orthologs of TFIIIC subunits Tau55, Tau131, and Tau138 in T. brucei and L. major. Thus, contrary to what was assumed, trypanosomatid parasites do possess a TFIIIC complex. Other putative interacting partners of Tau95 were identified in T. brucei and L. major. KEY POINTS: • A four-subunit TFIIIC complex is present in T. brucei and L. major • TbTau95 associates with tRNA and U2 snRNA genes • Putative interacting partners of Tau95 might include some RNAP II regulators.
    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
    在脊椎动物中,SMARCAD1参与转录调控,异染色质维持,DNA修复,和复制。其参与这些过程的分子基础尚不清楚。我们使用内源性共免疫沉淀将RNA聚合酶III通用转录因子TFIIIC鉴定为小鼠和人类模型中天然SMARCAD1的相互作用伴侣。TFIIIC具有双重功能,作为一般转录因子和基因组组织者分离染色质结构域。我们发现它与SMARCAD1的伙伴关系在不同的哺乳动物细胞类型中是保守的,从体细胞到多能细胞。使用纯化的蛋白质,我们确认他们的互动是直接的。基因表达分析表明,SMARCAD1对于TFIIIC功能作为小鼠ESC中的RNA聚合酶III转录因子是不必要的。TFIIC和SMARCAD1在ESC基因组中的分布是不同的,与酵母不同,SMARCAD1在活性tRNA基因上不富集。对多能和分化的哺乳动物细胞中SMARCAD1结合配偶体的进一步分析显示,SMARCAD1与在染色质组织中具有关键调节作用的几个因素相关,比如cohesin,laminB,DDX5一起,我们的工作首次表明SMARCAD1酶通过与结构蛋白的相互作用参与哺乳动物细胞核的基因组组织.
    In vertebrates, SMARCAD1 participates in transcriptional regulation, heterochromatin maintenance, DNA repair, and replication. The molecular basis underlying its involvement in these processes is not well understood. We identified the RNA polymerase III general transcription factor TFIIIC as an interaction partner of native SMARCAD1 in mouse and human models using endogenous co-immunoprecipitations. TFIIIC has dual functionality, acting as a general transcription factor and as a genome organizer separating chromatin domains. We found that its partnership with SMARCAD1 is conserved across different mammalian cell types, from somatic to pluripotent cells. Using purified proteins, we confirmed that their interaction is direct. A gene expression analysis suggested that SMARCAD1 is dispensable for TFIIIC function as an RNA polymerase III transcription factor in mouse ESCs. The distribution of TFIIIC and SMARCAD1 in the ESC genome is distinct, and unlike in yeast, SMARCAD1 is not enriched at active tRNA genes. Further analysis of SMARCAD1-binding partners in pluripotent and differentiated mammalian cells reveals that SMARCAD1 associates with several factors that have key regulatory roles in chromatin organization, such as cohesin, laminB, and DDX5. Together, our work suggests for the first time that the SMARCAD1 enzyme participates in genome organization in mammalian nuclei through interactions with architectural proteins.
    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
    RNA聚合酶III(PolIII)负责转录5S核糖体RNA(5SrRNA),tRNAs,和其他短的非编码RNA。它募集到5SrRNA启动子需要转录因子TFIIIA,TFIIC,和TFIIIB。这里,我们使用低温电子显微镜(cryo-EM)观察与启动子结合的TFIIIA和TFIIIC的酿酒酵母复合物。基因特异性因子TFIIIA与DNA相互作用并充当TFIIIC-启动子相互作用的衔接子。我们还可视化TFIIIB亚基的DNA结合,Brf1和TBP(TATA盒结合蛋白),这导致全长5SrRNA基因包裹在复合物周围。我们的smFRET研究表明,复合物中的DNA在缓慢的时间尺度上经历了急剧的弯曲和部分解离,与我们的低温EM结果预测的模型一致。我们的发现为5SrRNA启动子上的转录起始复合物组装提供了新的见解,并使我们能够直接比较PolIII和PolII转录适应。
    RNA polymerase III (Pol III) is responsible for transcribing 5S ribosomal RNA (5S rRNA), tRNAs, and other short non-coding RNAs. Its recruitment to the 5S rRNA promoter requires transcription factors TFIIIA, TFIIIC, and TFIIIB. Here, we use cryoelectron microscopy (cryo-EM) to visualize the S. cerevisiae complex of TFIIIA and TFIIIC bound to the promoter. Gene-specific factor TFIIIA interacts with DNA and acts as an adaptor for TFIIIC-promoter interactions. We also visualize DNA binding of TFIIIB subunits, Brf1 and TBP (TATA-box binding protein), which results in the full-length 5S rRNA gene wrapping around the complex. Our smFRET study reveals that the DNA within the complex undergoes both sharp bending and partial dissociation on a slow timescale, consistent with the model predicted from our cryo-EM results. Our findings provide new insights into the transcription initiation complex assembly on the 5S rRNA promoter and allow us to directly compare Pol III and Pol II transcription adaptations.
    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
    RNA聚合酶(pol)III的TFIIF样Rpc53/Rpc37异二聚体参与转录的各个阶段。Rpc53的C末端区域与Rpc37二聚化,以锚定在polIII裂隙的叶域上。然而,Rpc53N端区域的结构和功能特征先前尚未表征。这里,我们在Rpc53N端进行了定点丙氨酸置换诱变,产生表现出冷敏感生长缺陷和严重损害的polIII转录活性的酵母菌株。圆二色性和NMR光谱显示Rpc53N末端高度无序的57个氨基酸的多肽。该多肽是一种通用的蛋白质结合模块,显示出对Rpc37和转录起始因子TFIIIC的Tfc4亚基的纳摩尔水平结合亲和力。因此,我们将该Rpc53N末端多肽表示为TFIIIC结合区或CBR。CBR中的丙氨酸置换显着降低了其对Tfc4的结合亲和力,突出了其对体外细胞生长和转录的功能重要性。我们的研究揭示了Rpc53的CBR在polIII转录起始复合物组装中的功能基础。
    The TFIIF-like Rpc53/Rpc37 heterodimer of RNA polymerase (pol) III is involved in various stages of transcription. The C-terminal region of Rpc53 dimerizes with Rpc37 to anchor on the lobe domain of the pol III cleft. However, structural and functional features of the Rpc53 N-terminal region had not been characterized previously. Here, we conducted site-directed alanine replacement mutagenesis on the Rpc53 N-terminus, generating yeast strains that exhibited a cold-sensitive growth defect and severely compromised pol III transcriptional activity. Circular dichroism and NMR spectroscopy revealed a highly disordered 57-amino acid polypeptide in the Rpc53 N-terminus. This polypeptide is a versatile protein-binding module displaying nanomolar-level binding affinities for Rpc37 and the Tfc4 subunit of the transcription initiation factor TFIIIC. Accordingly, we denote this Rpc53 N-terminus polypeptide as the TFIIIC-binding region or CBR. Alanine replacements in the CBR significantly reduced its binding affinity for Tfc4, highlighting its functional importance to cell growth and transcription in vitro. Our study reveals the functional basis for Rpc53\'s CBR in assembly of the pol III transcription initiation complex.
    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
    组蛋白乙酰化的调节决定了基因表达的模式,从而决定了细胞的同一性。由于它们在癌症生物学中的临床相关性,了解人类胚胎干细胞(hESCs)如何调节其组蛋白乙酰化的基因组模式至关重要,但在很大程度上仍有待调查。这里,我们提供的证据表明,组蛋白H3赖氨酸-18(H3K18ac)和赖氨酸-27(H3K27ac)的乙酰化仅部分通过p300在干细胞中建立,而它代表了体细胞中这些标记的主要组蛋白乙酰转移酶(HAT)。我们的分析表明,尽管p300在hESC中与H3K18ac和H3K27ac略有相关,它在分化时与这些组蛋白标记大部分重叠。有趣的是,我们显示H3K18ac在hESCs中的RNA聚合酶III转录因子C(TFIIIC)富集的“干性”基因中发现,缺少P300此外,在涉及神经元生物学的基因附近也发现了TFIIC,虽然没有H3K18ac。我们的数据表明,与以前认为的相比,在hESC中负责组蛋白乙酰化的HAT模式更为复杂,提示H3K18ac和TFIIIC在调节“干性”基因以及与hESCs神经元分化相关的基因中的推定作用。结果为hESC基因组乙酰化的可能新范例开辟了基础,这可能导致癌症和发育疾病的治疗干预的新途径。
    Regulation of histone acetylation dictates patterns of gene expression and hence cell identity. Due to their clinical relevance in cancer biology, understanding how human embryonic stem cells (hESCs) regulate their genomic patterns of histone acetylation is critical, but it remains largely to be investigated. Here, we provide evidence that acetylation of histone H3 lysine-18 (H3K18ac) and lysine-27 (H3K27ac) is only partially established by p300 in stem cells, while it represents the main histone acetyltransferase (HAT) for these marks in somatic cells. Our analysis reveals that whereas p300 marginally associated with H3K18ac and H3K27ac in hESCs, it largely overlapped with these histone marks upon differentiation. Interestingly, we show that H3K18ac is found at \"stemness\" genes enriched in RNA polymerase III transcription factor C (TFIIIC) in hESCs, whilst lacking p300. Moreover, TFIIIC was also found in the vicinity of genes involved in neuronal biology, although devoid of H3K18ac. Our data suggest a more complex pattern of HATs responsible for histone acetylations in hESCs than previously considered, suggesting a putative role for H3K18ac and TFIIIC in regulating \"stemness\" genes as well as genes associated with neuronal differentiation of hESCs. The results break ground for possible new paradigms for genome acetylation in hESCs that could lead to new avenues for therapeutic intervention in cancer and developmental diseases.
    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
    真核染色体分为具有不同结构和功能特性的结构域,例如不同水平的染色质压缩和基因转录。相对紧凑的染色质和最小转录的域被称为异色,而常染色质更开放且转录活跃。绝缘体将这些域分开并保持其独特的特征。绝缘体的破坏会导致癌症等疾病。许多绝缘子含有tRNA基因(tDNA),其中的例子已被证明可以阻止激活或沉默活动的传播。特定tDNA的这种特征通过进化是保守的,这样人类tDNA可以作为裂变酵母沉默传播的屏障。在这里,我们证明了来自甲基营养真菌巴斯德毕赤酵母的tDNA可以有效地充当远缘相关的出芽酵母的绝缘体。tDNA作为绝缘体功能的关键是TFIIIC,转录因子,也是其表达所必需的。TFIIIC结合除tDNA之外的其他基因座,其中一些有绝缘体活动。尽管已经在酵母中广泛研究了基于TFIIIC的绝缘的机理基础,它在后生中基本上没有特征。利用公开可用的全基因组ChIP-seq数据,我们考虑了从酵母到人的保守机制可能足以在后生动物更具挑战性的染色质环境中通过TFIIIC进行有效隔离的程度,并提出了在进化过程中可能获得的特征以应对新的挑战。我们证明了USF1在人类tDNA中的广泛存在,USF1是一种在脊椎动物中具有良好屏障活性的转录因子。我们预测,高等生物中基于tDNA的绝缘体通过模块的掺入而进化,例如酵母中不存在的USF1和CTCF等因子的结合位点,从而加强功能并为细胞类型之间的调节提供机会。
    Eukaryotic chromosomes are divided into domains with distinct structural and functional properties, such as differing levels of chromatin compaction and gene transcription. Domains of relatively compact chromatin and minimal transcription are termed heterochromatic, whereas euchromatin is more open and actively transcribed. Insulators separate these domains and maintain their distinct features. Disruption of insulators can cause diseases such as cancer. Many insulators contain tRNA genes (tDNAs), examples of which have been shown to block the spread of activating or silencing activities. This characteristic of specific tDNAs is conserved through evolution, such that human tDNAs can serve as barriers to the spread of silencing in fission yeast. Here we demonstrate that tDNAs from the methylotrophic fungus Pichia pastoris can function effectively as insulators in distantly-related budding yeast. Key to the function of tDNAs as insulators is TFIIIC, a transcription factor that is also required for their expression. TFIIIC binds additional loci besides tDNAs, some of which have insulator activity. Although the mechanistic basis of TFIIIC-based insulation has been studied extensively in yeast, it is largely uncharacterized in metazoa. Utilising publicly-available genome-wide ChIP-seq data, we consider the extent to which mechanisms conserved from yeast to man may suffice to allow efficient insulation by TFIIIC in the more challenging chromatin environments of metazoa and suggest features that may have been acquired during evolution to cope with new challenges. We demonstrate the widespread presence at human tDNAs of USF1, a transcription factor with well-established barrier activity in vertebrates. We predict that tDNA-based insulators in higher organisms have evolved through incorporation of modules, such as binding sites for factors like USF1 and CTCF that are absent from yeasts, thereby strengthening function and providing opportunities for regulation between cell types.
    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
    转录因子(TF)以序列特异性方式结合DNA,并且通常是细胞类型特异性因子和/或发育主调节因子。相比之下,一般TFs(GTFs)是非常大的蛋白质复合物的一部分,用于RNA聚合酶募集到启动子序列,通常以细胞类型独立的方式。然而,几个TF已经被证明可以作为3D基因组组织的锚,GTFs在基因组结构中的作用尚未得到仔细研究。这里,我们使用ChIP-seq和Hi-C数据来描述TFIIC的作用,RNA聚合酶IIIGTF之一,在3D基因组组织中。我们发现TFIIIC基因组占用主要发生在特定区域,很大程度上对应于Alu元素;重复元素(RE)的其他特征类,如MIR,根据细胞的发育起源,还发现了FLAM-C和ALR/alpha。分析还表明,TFIIIC富集区域参与细胞类型特异性DNA循环,这不依赖于与主结构蛋白CTCF的共定位。这项工作扩展了关于TFIIIC作为真正的基因组组织者的作用的先前知识,其作用通过与RE结合参与细胞类型依赖性3D基因组循环。
    Transcription factors (TFs) bind DNA in a sequence-specific manner and are generally cell type-specific factors and/or developmental master regulators. In contrast, general TFs (GTFs) are part of very large protein complexes and serve for RNA polymerases\' recruitment to promoter sequences, generally in a cell type-independent manner. Whereas, several TFs have been proven to serve as anchors for the 3D genome organization, the role of GTFs in genome architecture have not been carefully explored. Here, we used ChIP-seq and Hi-C data to depict the role of TFIIIC, one of the RNA polymerase III GTFs, in 3D genome organization. We find that TFIIIC genome occupancy mainly occurs at specific regions, which largely correspond to Alu elements; other characteristic classes of repetitive elements (REs) such as MIR, FLAM-C and ALR/alpha are also found depending on the cell\'s developmental origin. The analysis also shows that TFIIIC-enriched regions are involved in cell type-specific DNA looping, which does not depend on colocalization with the master architectural protein CTCF. This work extends previous knowledge on the role of TFIIIC as a bona fide genome organizer whose action participates in cell type-dependent 3D genome looping via binding to REs.
    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
    如何重复元素,表观遗传修饰,和建筑蛋白相互作用,确保正确的基因组表达仍然知之甚少。这里,我们报告的调节机制揭示了Alu元件(AEs)和RNA聚合酶III转录因子C(TFIIIC)在通过染色质循环和组蛋白乙酰化进行基因组结构和功能调节中的重要作用.血清剥夺后,由活性依赖性神经保护者同源异型盒蛋白(ADNP)预先标记并位于细胞周期基因附近的AE子集招募TFIIIC,通过组蛋白H3赖氨酸-18(H3K18)的直接乙酰化改变其染色质可及性。这促进了AEs与其他细胞周期基因启动子附近的远处CTCF位点的接触,在H3K18也被高度乙酰化。这些变化确保了细胞周期基因的基础转录,并且对于血清再暴露后的再激活至关重要。我们的研究揭示了一般转录因子对AE的表观遗传状态的直接操纵如何调节3D基因组折叠和表达。
    How repetitive elements, epigenetic modifications, and architectural proteins interact ensuring proper genome expression remains poorly understood. Here, we report regulatory mechanisms unveiling a central role of Alu elements (AEs) and RNA polymerase III transcription factor C (TFIIIC) in structurally and functionally modulating the genome via chromatin looping and histone acetylation. Upon serum deprivation, a subset of AEs pre-marked by the activity-dependent neuroprotector homeobox Protein (ADNP) and located near cell-cycle genes recruits TFIIIC, which alters their chromatin accessibility by direct acetylation of histone H3 lysine-18 (H3K18). This facilitates the contacts of AEs with distant CTCF sites near promoter of other cell-cycle genes, which also become hyperacetylated at H3K18. These changes ensure basal transcription of cell-cycle genes and are critical for their re-activation upon serum re-exposure. Our study reveals how direct manipulation of the epigenetic state of AEs by a general transcription factor regulates 3D genome folding and expression.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Interphase chromosomes are organized into topologically associated domains in order to establish and maintain integrity of transcriptional programs that remain poorly understood. Here, we show that condensin II and TFIIIC are recruited to bidirectionally transcribed promoters by a mechanism that is dependent on the retinoblastoma (RB) protein. Long-range chromosome contacts are disrupted by loss of condensin II loading, which leads to altered expression at bidirectional gene pairs. This study demonstrates that mammalian condensin II functions to organize long-range chromosome contacts and regulate transcription at specific genes. In addition, RB dependence of condensin II suggests that widespread misregulation of chromosome contacts and transcriptional alterations are a consequence of RB mutation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Eukaryotic transcription is a highly regulated fundamental life process. A large number of regulatory proteins and complexes, many of them with sequence-specific DNA-binding activity are known to influence transcription by RNA polymerase (pol) II with a fine precision. In comparison, only a few regulatory proteins are known for pol III, which transcribes genes encoding small, stable, non-translated RNAs. The pol III transcription is precisely regulated under various stress conditions. We used pol III transcription complex (TC) components TFIIIC (Tfc6), pol III (Rpc128) and TFIIIB (Brf1) as baits and mass spectrometry to identify their potential interactors in vivo. A large interactome constituting chromatin modifiers, regulators and factors of transcription by pol I and pol II supports the possibility of a crosstalk between the three transcription machineries. The association of proteins and complexes involved in various basic life processes like ribogenesis, RNA processing, protein folding and degradation, DNA damage response, replication and transcription underscores the possibility of the pol III TC serving as a signaling hub for communication between the transcription and other cellular physiological activities under normal growth conditions. We also found an equally large number of proteins and complexes interacting with the TC under nutrient starvation condition, of which at least 25% were non-identical under the two conditions. The data reveal the possibility of a large number of signaling cues for pol III transcription against adverse conditions, necessary for an efficient co-ordination of various cellular functions.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号