CCCTC-Binding Factor

CCCTC - 结合因子
  • 文章类型: Journal Article
    染色体重排可能会扭曲3D染色质结构,从而改变基因调控,然而,3D染色质结构如何在昆虫中进化在很大程度上是未知的。这里,我们获得了四种蝴蝶的染色体水平基因组,葡萄cloanthus,葡萄,葡萄,分别为2n=30、40和60,和Papiliobianor,2n=60。加上大规模的Hi-C数据,我们发现,染色体间重排很少破坏祖先染色体预先存在的3D染色质结构。然而,与祖先构型相比,一些染色体内重排改变了3D染色质结构。我们发现,在重排位点上出现了新的TAD和subTAD,它们的相邻区室表现出均匀的类型。两个染色体内重排改变了Rel和lft调节,可能有助于机翼模式分化和寄主植物选择。值得注意的是,蝴蝶在Hox基因簇ANT-C和BX-C之间表现出染色质环,不像果蝇.我们在蝴蝶中的CRISPR-Cas9实验证实,敲除BX-C中环的CTCF结合位点会影响ANT-C中Antp调节的表型,导致无腿幼虫。我们的结果揭示了昆虫3D染色质结构的进化模式,并提供了3D染色质结构变化在性状进化中起重要作用的证据。
    Chromosome rearrangements may distort 3D chromatin architectures and thus change gene regulation, yet how 3D chromatin structures evolve in insects is largely unknown. Here, we obtain chromosome-level genomes for four butterfly species, Graphium cloanthus, Graphium sarpedon, Graphium eurypylus with 2n = 30, 40, and 60, respectively, and Papilio bianor with 2n = 60. Together with large-scale Hi-C data, we find that inter-chromosome rearrangements very rarely disrupted the pre-existing 3D chromatin structure of ancestral chromosomes. However, some intra-chromosome rearrangements changed 3D chromatin structures compared to the ancestral configuration. We find that new TADs and subTADs have emerged across the rearrangement sites where their adjacent compartments exhibit uniform types. Two intra-chromosome rearrangements altered Rel and lft regulation, potentially contributing to wing patterning differentiation and host plant choice. Notably, butterflies exhibited chromatin loops between Hox gene cluster ANT-C and BX-C, unlike Drosophila. Our CRISPR-Cas9 experiments in butterflies confirm that knocking out the CTCF binding site of the loops in BX-C affected the phenotypes regulated by Antp in ANT-C, resulting in legless larva. Our results reveal evolutionary patterns of insect 3D chromatin structures and provide evidence that 3D chromatin structure changes can play important roles in the evolution of traits.
    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
    自然杀伤细胞(NK)是肿瘤的“职业杀手”,在抗肿瘤免疫治疗中起着至关重要的作用。NK细胞脱敏是肿瘤免疫逃逸的一个症结机制。失调的NKG2D-NKG2DL信号传导是这种脱敏过程的主要驱动因素。然而,调节NK细胞脱敏的因素在很大程度上仍未表征。这里,我们首次报道了环状RNAcircARAP2(hsa_circ_0069396)参与NK细胞脱敏模型中可溶性MICA(sMICA)诱导的NKG2D内吞。在NK细胞脱敏过程中CircARAP2上调,CircARAP2的丢失减轻了NKG2D内吞和NK细胞脱敏。使用通过RNA纯化(ChIRP)和RNA下拉方法分离染色质,我们发现RAB5A,早期内体的分子标记,是它的下游目标。值得注意的是,转录因子CTCF是circARAP2的中间功能伴侣.机械上,我们发现circARAP2与CTCF相互作用并抑制CTCF-Polycomb阻遏复合物2(PRC2)向RAB5A启动子区的募集,从而消除组蛋白H3K27和H3K9甲基化抑制以增强RAB5A转录。这些数据表明,circARAP2的抑制有效缓解sMICA诱导的NKG2D内吞和NK细胞脱敏,为肿瘤免疫逃避的治疗干预提供了新的靶点。
    Natural killer cells (NK) are the \"professional killer\" of tumors and play a crucial role in anti-tumor immunotherapy. NK cell desensitization is a key mechanism of tumor immune escape. Dysregulated NKG2D-NKG2DL signaling is a primary driver of this desensitization process. However, the factors that regulate NK cell desensitization remain largely uncharacterized. Here, we present the first report that circular RNA circARAP2 (hsa_circ_0069396) is involved in the soluble MICA (sMICA)-induced NKG2D endocytosis in the NK cell desensitization model. CircARAP2 was upregulated during NK cell desensitization and the loss of circARAP2 alleviated NKG2D endocytosis and NK cell desensitization. Using Chromatin isolation by RNA purification (ChIRP) and RNA pull-down approaches, we identified that RAB5A, a molecular marker of early endosomes, was its downstream target. Notably, transcription factor CTCF was an intermediate functional partner of circARAP2. Mechanistically, we discovered that circARAP2 interacted with CTCF and inhibited the recruitment of CTCF-Polycomb Repressive Complex 2 (PRC2) to the promoter region of RAB5A, thereby erasing histone H3K27 and H3K9 methylation suppression to enhance RAB5A transcription. These data demonstrate that inhibition of circARAP2 effectively alleviates sMICA-induced NKG2D endocytosis and NK cell desensitization, providing a novel target for therapeutic intervention in tumor immune evasion.
    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:转座因子在维持神经发育过程中的基因组结构中起着关键作用。短散布核元素(SINE),转座因子的主要亚型,已知具有CCCTC结合因子(CTCF)的结合位点,并且在协调染色质组织中至关重要。然而,在发育中的大脑中控制SINE活性的调节机制仍然难以捉摸。
    结果:在我们的研究中,我们使用ATAC-seq对小鼠神经前体细胞进行了全面的全基因组表观遗传分析,ChIP-seq,全基因组亚硫酸氢盐测序,就地Hi-C,和RNA-seq。我们的发现表明,SET结构域分叉的组蛋白赖氨酸甲基转移酶1(SETDB1)介导的H3K9me3与DNA甲基化结合,限制了神经前体细胞中选择性SINE子集的染色质可及性。机械上,Settb1的丢失会增加CTCF对这些SINE元素的访问,并有助于染色质环的重组。此外,从头环形成有助于差异基因表达,包括有丝分裂途径中富集的基因的失调。这导致在体外和体内对Setdb1进行遗传消融后胚胎脑中细胞增殖的破坏。
    结论:总之,我们的研究揭示了小鼠神经前体细胞中SINE的表观遗传调控,提示它们在维持神经发育过程中染色质组织和细胞增殖中的作用。
    BACKGROUND: Transposable elements play a critical role in maintaining genome architecture during neurodevelopment. Short Interspersed Nuclear Elements (SINEs), a major subtype of transposable elements, are known to harbor binding sites for the CCCTC-binding factor (CTCF) and pivotal in orchestrating chromatin organization. However, the regulatory mechanisms controlling the activity of SINEs in the developing brain remains elusive.
    RESULTS: In our study, we conduct a comprehensive genome-wide epigenetic analysis in mouse neural precursor cells using ATAC-seq, ChIP-seq, whole genome bisulfite sequencing, in situ Hi-C, and RNA-seq. Our findings reveal that the SET domain bifurcated histone lysine methyltransferase 1 (SETDB1)-mediated H3K9me3, in conjunction with DNA methylation, restricts chromatin accessibility on a selective subset of SINEs in neural precursor cells. Mechanistically, loss of Setdb1 increases CTCF access to these SINE elements and contributes to chromatin loop reorganization. Moreover, de novo loop formation contributes to differential gene expression, including the dysregulation of genes enriched in mitotic pathways. This leads to the disruptions of cell proliferation in the embryonic brain after genetic ablation of Setdb1 both in vitro and in vivo.
    CONCLUSIONS: In summary, our study sheds light on the epigenetic regulation of SINEs in mouse neural precursor cells, suggesting their role in maintaining chromatin organization and cell proliferation during neurodevelopment.
    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
    发育需要CTCF组织的三维基因组结构。临床鉴定的CTCF突变与不良发育结果有关。然而,潜在的机制仍然难以捉摸。在这次调查中,我们探讨了临床相关的R567W点突变的调节作用,位于CTCF的第11个锌指内,通过将这种突变引入小鼠模型和人类胚胎干细胞来源的皮质类器官模型。具有纯合CTCFR567W突变的小鼠表现出生长障碍,导致产后死亡率,和大脑的偏差,心,和病理和单细胞转录组水平的肺发育。这种突变诱导过早的干细胞样细胞衰竭,加速GABA能神经元的成熟,破坏神经发育和突触通路.此外,它特别阻碍CTCF与核心共识位点上游的外周基序结合,导致局部染色质结构和基因表达的改变,特别是在成簇的protcadherin位点。使用人类皮质类器官的比较分析反映了这种突变引起的后果。总之,这项研究阐明了CTCFR567W突变对人类神经发育障碍的影响,为潜在的治疗干预铺平道路。
    The three-dimensional genome structure organized by CTCF is required for development. Clinically identified mutations in CTCF have been linked to adverse developmental outcomes. Nevertheless, the underlying mechanism remains elusive. In this investigation, we explore the regulatory roles of a clinically relevant R567W point mutation, located within the 11th zinc finger of CTCF, by introducing this mutation into both murine models and human embryonic stem cell-derived cortical organoid models. Mice with homozygous CTCFR567W mutation exhibit growth impediments, resulting in postnatal mortality, and deviations in brain, heart, and lung development at the pathological and single-cell transcriptome levels. This mutation induces premature stem-like cell exhaustion, accelerates the maturation of GABAergic neurons, and disrupts neurodevelopmental and synaptic pathways. Additionally, it specifically hinders CTCF binding to peripheral motifs upstream to the core consensus site, causing alterations in local chromatin structure and gene expression, particularly at the clustered protocadherin locus. Comparative analysis using human cortical organoids mirrors the consequences induced by this mutation. In summary, this study elucidates the influence of the CTCFR567W mutation on human neurodevelopmental disorders, paving the way for potential therapeutic interventions.
    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
    目的:本研究旨在探讨程序性细胞死亡配体1(PD-L1)通过介导CCCTC结合因子(CTCF)表达促进人牙髓干细胞(hDPSCs)增殖和成骨分化的作用及其机制。
    方法:通过免疫共沉淀法验证PD-L1与CTCF的相互作用。用脂多糖或成骨诱导培养基处理用PD-L1过表达和CTCF敲低载体转染的hDPSC。检测炎性细胞因子和骨/牙源性分化相关基因。使用碱性磷酸酶(ALP)和茜素红S染色评估hDPSC的骨/牙源性分化。
    结果:PD-L1过表达抑制LPS诱导的促炎细胞因子上调,细胞增殖,ALP活性,和钙在hDPSC中的沉积,并提高了骨/牙源性分化相关基因的表达;然而,这种表达模式可以通过CTCF敲低逆转。免疫共沉淀结果证实了PD-L1与CTCF的结合,表明hDPSC中PD-L1过表达增加CTCF表达,从而抑制炎症反应并增加hDPSC的骨/牙源性分化。
    结论:PD-L1在hDPSC中的过表达增强了hDPSC的增殖和骨/牙源性分化,并通过上调CTCF表达来抑制炎症反应。
    OBJECTIVE: The aim of this study was to explore the effect and mechanism of programmed cell death ligand 1 (PD-L1) in promoting the proliferation and osteo/odontogenic-differentiation of human dental pulp stem cells (hDPSCs) by mediating CCCTC-binding factor (CTCF) expression.
    METHODS: The interaction between PD-L1 and CTCF was verified through co-immunoprecipitation. hDPSCs transfected with PD-L1 overexpression and CTCF knockdown vectors were treated with lipopolysaccharide or an osteogenic-inducing medium. Inflammatory cytokines and osteo/odontogenic-differentiation related genes were measured. Osteo/odontogenic-differentiation of hDPSCs was assessed using alkaline phosphatase (ALP) and alizarin red S staining.
    RESULTS: Overexpression of PD-L1 inhibited LPS-induced pro-inflammatory cytokine upregulation, cell proliferation, ALP activity, and calcium deposition in hDPSCs and elevated the expression of osteo/odontogenic-differentiation related genes; however, such expression patterns could be reversed by CTCF knockdown. Co-immunoprecipitation results confirmed the binding of PD-L1 to CTCF, indicating that PD-L1 overexpression in hDPSCs increases CTCF expression, thus inhibiting the inflammatory response and increasing osteo/odontogenic-differentiation of hDPSCs.
    CONCLUSIONS: PD-L1 overexpression in hDPSCs enhances the proliferation and osteo/odontogenic-differentiation of hDPSCs and inhibit the inflammatory response by upregulating CTCF expression.
    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
    血管内皮细胞功能影响下肢动脉硬化闭塞症(LEASO),而α-2-巨球蛋白(A2M)和CCCTC结合因子(CTCF)与此类细胞的功能密切相关。本文旨在研究CTCF通过调节A2M对LEASO血管内皮细胞的影响。建立LEASO大鼠模型,测量内膜-中膜比,血脂,和炎症因子水平。通过构建LEASO细胞模型,测定细胞活力和细胞凋亡,而自噬相关蛋白,测定股动脉组织和HUVECs中的CTCF和A2M水平。证实了CTCF对A2M的转录调控。在LEASO大鼠模型中,股动脉管腔变窄,内皮细胞紊乱;总胆固醇水平,IL-1和TNF-α增强,HDL-C下降,A2M强表达,CTCF低表达。ox-LDL处理的HUVECs的活力降低,伴随着更高的细胞凋亡,较低的LC3II/I表达,和更高的p62表达,通过sh-A2M转染逆转。CTCF过表达抑制A2M转录,促进HUVECs的活力和自噬,细胞凋亡减少。总的来说,CTCF通过抑制A2M转录改善LEASO中血管内皮细胞的功能。
    Vascular endothelial cell functions affect lower extremity arteriosclerosis obliterans (LEASO), while alpha-2-macroglobulin (A2M) and CCCTC-binding factor (CTCF) are closely related to the function of such cells. This paper aims to identify the influences of CTCF on vascular endothelial cells in LEASO by regulating A2M. A rat model of LEASO was established to measure intima-media ratio, blood lipid, and inflammatory factor levels. By constructing LEASO cell models, cell viability and apoptosis were assayed, while autophagy-related proteins, CTCF and A2M levels in femoral artery tissues and HUVECs were determined. The transcriptional regulation of CTCF on A2M was verified. In LEASO rat models, femoral artery lumen was narrowed and endothelial cells were disordered; levels of total cholesterol, IL-1, and TNF-α enhanced, and HDL-C decreased, with strong expression of A2M and low expression of CTCF. The viability of ox-LDL-treated HUVECs was decreased, together with higher apoptosis, lower LC3II/I expression, and higher p62 expression, which were reversed by sh-A2M transfection. Overexpression of CTCF inhibited A2M transcription, promoted the viability and autophagy of HUVECs, and decreased apoptosis. Collectively, CTCF improves the function of vascular endothelial cells in LEASO by inhibiting A2M transcription.
    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
    肺泡屏障的破坏可引发急性肺损伤。这项研究阐明了甲基转移酶样3(METTL3)与肺炎链球菌(SP)诱导的肺泡上皮细胞(AECs)凋亡和炎症损伤的关系。培养AECs,然后用SP感染。此外,METTL3、白细胞介素(IL)-10、IL-6、肿瘤坏死因子-α(TNF-α)的表达,单核细胞趋化蛋白-1(MCP-1),长链非编码RNA核旁斑组装转录物1(NEAT1),粘蛋白19(MUC19),N6-甲基腺苷(m6A),通过qRT-PCR检测m6A修饰后的NEAT1,蛋白质印迹,和酶联免疫吸附,m6A定量,和甲基化RNA免疫沉淀-qPCR分析,分别。此外,NEAT1的亚细胞定位通过核/胞质溶胶分馏分析进行分析,并分析了NEAT1与CCCTC结合因子(CTCF)之间的结合。这项研究的结果表明,SP诱导了AECs中的凋亡和炎症损伤,并上调了METTL3的表达。此外,METTL3的下调减轻了AECs的凋亡和炎症损伤。METTL3介导的m6A修饰增加NEAT1并促进其与CTCF的结合以促进MUC19转录。NEAT1或MUC19过表达破坏了它们在AECs中沉默METTL3的保护作用,从而增加细胞凋亡和炎症损伤。总之,这是第一项研究表明METTL3通过NEAT1/CTCF/MUC19轴加重SP诱导的细胞损伤。
    Disruption of the alveolar barrier can trigger acute lung injury. This study elucidated the association of methyltransferase-like 3 (METTL3) with Streptococcus pneumoniae (SP)-induced apoptosis and inflammatory injury of alveolar epithelial cells (AECs). AECs were cultured and then infected with SP. Furthermore, the expression of METTL3, interleukin (IL)-10, IL-6, tumor necrosis factor-alpha (TNF-α), monocyte chemoattractant protein-1 (MCP-1), long noncoding RNA nuclear paraspeckle assembly transcript 1 (NEAT1), mucin 19 (MUC19), N6-methyladenosine (m6A), and NEAT1 after m6A modification were detected by qRT-PCR, Western blot, and enzyme-linked immunosorbent, m6A quantification, and methylated RNA immunoprecipitation-qPCR analyses, respectively. Moreover, the subcellular localization of NEAT1 was analyzed by nuclear/cytosol fractionation assay, and the binding between NEAT1 and CCCTC-binding factor (CTCF) was also analyzed. The results of this investigation revealed that SP-induced apoptosis and inflammatory injury in AECs and upregulated METTL3 expression. In addition, the downregulation of METTL3 alleviated apoptosis and inflammatory injury in AECs. METTL3-mediated m6A modification increased NEAT1 and promoted its binding with CTCF to facilitate MUC19 transcription. NEAT1 or MUC19 overexpression disrupted their protective role of silencing METTL3 in AECs, thereby increasing apoptosis and inflammatory injury. In conclusion, this is the first study to suggest that METTL3 aggravates SP-induced cell damage via the NEAT1/CTCF/MUC19 axis.
    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)在染色质上的停留转化为基因组上的定量转录或结构结果。常用的甲醛交联累积地固定TF-DNA相互作用并损害测量的占有率水平。在这里,我们绘制了CTCF和MAZ等全球或单个锌指TF的占用水平,以高分辨率的脚印形式,天然染色质。通过结合增强扰动条件,我们建立了S分数,一种定量指标,用于替代天然染色质上不同基序之间CTCF或MAZ保留的连续性。保留有天然染色质的CTCF位点具有CTCF基序内的序列特征,S评分比从其他交联或天然测定获得的指标更好地解释。CTCF在天然染色质上的保留与局部SUMO化水平相关,和抗相关的转录活性。S分数成功地描绘了CTCF介导的染色质结构的其他掩蔽差异稳定性,或独立于CTCF的MAZ。总的来说,我们的研究建立了跨天然染色质结合位点的TF保留的范式连续体,解释动态基因组组织。
    Transcription factor (TF) residence on chromatin translates into quantitative transcriptional or structural outcomes on genome. Commonly used formaldehyde crosslinking fixes TF-DNA interactions cumulatively and compromises the measured occupancy level. Here we mapped the occupancy level of global or individual zinc finger TFs like CTCF and MAZ, in the form of highly resolved footprints, on native chromatin. By incorporating reinforcing perturbation conditions, we established S-score, a quantitative metric to proxy the continuum of CTCF or MAZ retention across different motifs on native chromatin. The native chromatin-retained CTCF sites harbor sequence features within CTCF motifs better explained by S-score than the metrics obtained from other crosslinking or native assays. CTCF retention on native chromatin correlates with local SUMOylation level, and anti-correlates with transcriptional activity. The S-score successfully delineates the otherwise-masked differential stability of chromatin structures mediated by CTCF, or by MAZ independent of CTCF. Overall, our study established a paradigm continuum of TF retention across binding sites on native chromatin, explaining the dynamic genome organization.
    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
    染色质环对基因转录的调控至关重要。Cohesin是一种染色质相关蛋白,通过环挤出介导染色质的相互作用。Cohesin介导的染色质相互作用具有很强的细胞类型特异性,对预测染色质环构成挑战。现有的计算方法在预测细胞类型特异性染色质环方面表现不佳。为了解决这个问题,我们提出了一个随机森林模型,根据ChromHMM鉴定的染色质状态和相关因子的占有率来预测细胞类型特异性内聚介导的染色质环。我们的结果表明,染色质状态负责环的细胞类型特异性。仅使用染色质状态作为特征,该模型在预测两种细胞类型之间的细胞类型特异性循环方面具有很高的准确性,并且可以应用于不同的细胞类型。此外,当染色质状态与CTCF的发生频率相结合时,RAD21、YY1和H3K27acChIP-seq峰,可以实现更准确的预测。我们的特征提取方法提供了预测细胞类型特异性染色质环的新见解,并揭示了染色质状态和染色质环形成之间的关系。
    Chromatin loop is of crucial importance for the regulation of gene transcription. Cohesin is a type of chromatin-associated protein that mediates the interaction of chromatin through the loop extrusion. Cohesin-mediated chromatin interactions have strong cell-type specificity, posing a challenge for predicting chromatin loops. Existing computational methods perform poorly in predicting cell-type-specific chromatin loops. To address this issue, we propose a random forest model to predict cell-type-specific cohesin-mediated chromatin loops based on chromatin states identified by ChromHMM and the occupancy of related factors. Our results show that chromatin state is responsible for cell-type-specificity of loops. Using only chromatin states as features, the model achieved high accuracy in predicting cell-type-specific loops between two cell types and can be applied to different cell types. Furthermore, when chromatin states are combined with the occurrence frequency of CTCF, RAD21, YY1, and H3K27ac ChIP-seq peaks, more accurate prediction can be achieved. Our feature extraction method provides novel insights into predicting cell-type-specific chromatin loops and reveals the relationship between chromatin state and chromatin loop formation.
    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
    CCCTC结合因子(CTCF),一种广泛表达且高度保守的蛋白质,已知在染色质结构中起关键作用。翻译后修饰(PTM)使蛋白质的功能多样化,以调节许多细胞过程。然而,PTM对CTCF全基因组结合和三维(3D)染色质结构组织的影响尚未完全了解.在这项研究中,我们发现了CTCF的PTM谱,并证明CTCF可以是O-GlcN酰化和精氨酸甲基化的。功能上,我们证明O-GlcNAcylation抑制CTCF与染色质的结合。同时,CTCFO-GlcNAcylation的缺乏导致与细胞发育相关的环结构域的破坏和染色质环的改变。此外,CTCFO-GlcNAcylation的缺乏会增加发育基因的表达,并负向调节干细胞多能性的维持和建立。总之,这些结果为PTM在3D染色质结构中的作用提供了关键见解。
    CCCTC-binding factor (CTCF), a ubiquitously expressed and highly conserved protein, is known to play a critical role in chromatin structure. Post-translational modifications (PTMs) diversify the functions of protein to regulate numerous cellular processes. However, the effects of PTMs on the genome-wide binding of CTCF and the organization of three-dimensional (3D) chromatin structure have not been fully understood. In this study, we uncovered the PTM profiling of CTCF and demonstrated that CTCF can be O-GlcNAcylated and arginine methylated. Functionally, we demonstrated that O-GlcNAcylation inhibits CTCF binding to chromatin. Meanwhile, deficiency of CTCF O-GlcNAcylation results in the disruption of loop domains and the alteration of chromatin loops associated with cellular development. Furthermore, the deficiency of CTCF O-GlcNAcylation increases the expression of developmental genes and negatively regulates maintenance and establishment of stem cell pluripotency. In conclusion, these results provide key insights into the role of PTMs for the 3D chromatin structure.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号