histone H3K4 methylation

  • 文章类型: Journal Article
    组蛋白赖氨酸脱甲基酶KDM5B在各种人类癌细胞中经常上调。然而,其在人急性髓系白血病(AML)细胞中的表达和功能作用尚不清楚。这里,我们发现KDM5B在原代人AML细胞中的表达水平很高。我们已经证明,敲低KDM5B导致原发性人AML和一些人AML细胞系中的细胞凋亡并损害增殖。我们进一步鉴定miR-140-3p为KDM5B的下游靶基因。KDM5B表达与原代人AML细胞中miR-140-3p水平呈负相关。分子研究表明,沉默KDM5B可增强miR-140-3p启动子处的H3K4三甲基化(H3K4me3),导致miR-140-3p的高表达,进而抑制B细胞CLL/淋巴瘤2(BCL2)的表达。最后,我们证明了由KDM5B敲低(KD)诱导的缺陷增殖可以用miR-140-3p抑制剂挽救或通过将KDM5BKD与BCL2抑制剂联合使用而增强.总之,我们的数据支持以下结论:KDM5B通过miR-140-3p/BCL2轴促进人AML细胞的肿瘤发生.靶向KDM5B/miR-140-3p/BCL2途径可能具有治疗人类AML的治疗前景。
    The histone lysine demethylase KDM5B is frequently up-regulated in various human cancer cells. However, its expression and functional role in human acute myeloid leukemia (AML) cells remain unclear. Here, we found that the expression level of KDM5B is high in primary human AML cells. We have demonstrated that knocking down KDM5B leads to apoptosis and impairs proliferation in primary human AML and some human AML cell lines. We further identified miR-140-3p as a downstream target gene of KDM5B. KDM5B expression was inversely correlated with the miR-140-3p level in primary human AML cells. Molecular studies showed that silencing KDM5B enhanced H3K4 trimethylation (H3K4me3) at the promoter of miR-140-3p, leading to high expression of miR-140-3p, which in turn inhibited B-cell CLL/lymphoma 2 (BCL2) expression. Finally, we demonstrate that the defective proliferation induced by KDM5B knockdown (KD) can be rescued with the miR-140-3p inhibitor or enhanced by combining KDM5B KD with a BCL2 inhibitor. Altogether, our data support the conclusion that KDM5B promotes tumorigenesis in human AML cells through the miR-140-3p/BCL2 axis. Targeting the KDM5B/miR-140-3p/BCL2 pathway may hold therapeutic promise for treating human AML.
    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
    多种信号通路通过调节恶性B细胞的迁移和对微环境壁ni的粘附来促进其存活和耐药性。NF-κB通路在套细胞淋巴瘤(MCL)中通常失调,但是确切的潜在机制还没有得到很好的理解。这里,使用共同文化模型系统,我们显示MCL细胞与基质细胞的粘附与贴壁细胞中KDM6B组蛋白脱甲基酶mRNA水平升高有关。抑制KDM6B活性,使用选择性抑制剂(GSK-J4)或siRNA介导的敲减,减少MCL对基质细胞的粘附。我们表明,KDM6B对于去除NF-κB编码基因启动子区域的抑制性染色质标记(H3K27me3)以及诱导粘附MCL细胞中NF-κB基因的表达都是必需的。GSK-J4降低RELANF-κB亚基的蛋白质水平并损害其核定位。我们进一步证明了一些粘附诱导的靶基因需要诱导的NF-κB和KDM6B活性来诱导它们(例如,IL-10细胞因子基因),而其他人则需要诱导NF-κB而不是KDM6B(例如,CCR7趋化因子基因)。总之,KDM6B在MCL中不同水平诱导NF-κB通路,从而促进MCL细胞粘附,生存,和抗药性。KDM6B代表MCL的新型潜在治疗靶标。
    Multiple signaling pathways facilitate the survival and drug resistance of malignant B-cells by regulating their migration and adhesion to microenvironmental niches. NF-κB pathways are commonly dysregulated in mantle cell lymphoma (MCL), but the exact underlying mechanisms are not well understood. Here, using a co-culture model system, we show that the adhesion of MCL cells to stromal cells is associated with elevated levels of KDM6B histone demethylase mRNA in adherent cells. The inhibition of KDM6B activity, using either a selective inhibitor (GSK-J4) or siRNA-mediated knockdown, reduces MCL adhesion to stromal cells. We showed that KDM6B is required both for the removal of repressive chromatin marks (H3K27me3) at the promoter region of NF-κB encoding genes and for inducing the expression of NF-κB genes in adherent MCL cells. GSK-J4 reduced protein levels of the RELA NF-κB subunit and impaired its nuclear localization. We further demonstrated that some adhesion-induced target genes require both induced NF-κB and KDM6B activity for their induction (e.g., IL-10 cytokine gene), while others require induction of NF-κB but not KDM6B (e.g., CCR7 chemokine gene). In conclusion, KDM6B induces the NF-κB pathway at different levels in MCL, thereby facilitating MCL cell adhesion, survival, and drug resistance. KDM6B represents a novel potential therapeutic target for MCL.
    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:多发性内分泌肿瘤1型(MEN1)基因的功能缺失突变是MEN1肿瘤综合征的原因,但它们也常见于散发性胰腺神经内分泌肿瘤和其他类型的癌症。MEN1基因产物,梅宁,参与转录和染色质调节,最重要的是作为含有COMPASS样组蛋白H3K4甲基转移酶复合物的KMT2A/MLL1和KMT2B/MLL2的组成部分。以一种相互排斥的方式,menin还与AP-1的JunD亚基和ATF/CREB转录因子相互作用。
    结果:这里,我们对253个疾病相关MEN1错义突变进行了计算机筛选,以筛选出一组9个表面暴露残基中的menin突变.通过定量质谱法评估这些突变体的蛋白质相互作用,这表明9个突变体中有7个破坏了与MLL1/MLL2和JunD复合物的相互作用。有趣的是,我们发现了三个错义突变,R52G,E255K和E359K,与JunD相比,主要减少MLL1和MLL2相互作用。R52G结合的明显丧失支持了这一观察结果。E255K和E359K突变蛋白在独特的MLL1基因组结合位点,对独特的JunD位点影响较小。
    结论:我们的结果强调了内分泌起源的家族性和散发性肿瘤中MEN1基因突变对menin与MLL1和MLL2组蛋白H3K4甲基转移酶复合物以及含有JunD的转录因子相互作用的影响。Menin结合口袋突变体R52G,E255K和E359K对MLL1/MLL2和JunD相互作用有不同的影响,转化为差异基因组结合模式。我们的发现鼓励未来研究解决MEN1疾病模型系统中menin突变体的单独MLL1/MLL2-和JunD依赖性功能的病理生理学相关性。
    BACKGROUND: Loss-of-function mutations of the multiple endocrine neoplasia type 1 (MEN1) gene are causal to the MEN1 tumor syndrome, but they are also commonly found in sporadic pancreatic neuroendocrine tumors and other types of cancers. The MEN1 gene product, menin, is involved in transcriptional and chromatin regulation, most prominently as an integral component of KMT2A/MLL1 and KMT2B/MLL2 containing COMPASS-like histone H3K4 methyltransferase complexes. In a mutually exclusive fashion, menin also interacts with the JunD subunit of the AP-1 and ATF/CREB transcription factors.
    RESULTS: Here, we applied and in silico screening approach for 253 disease-related MEN1 missense mutations in order to select a set of nine menin mutations in surface-exposed residues. The protein interactomes of these mutants were assessed by quantitative mass spectrometry, which indicated that seven of the nine mutants disrupt interactions with both MLL1/MLL2 and JunD complexes. Interestingly, we identified three missense mutations, R52G, E255K and E359K, which predominantly reduce the MLL1 and MLL2 interactions when compared with JunD. This observation was supported by a pronounced loss of binding of the R52G, E255K and E359K mutant proteins at unique MLL1 genomic binding sites with less effect on unique JunD sites.
    CONCLUSIONS: Our results underline the effects of MEN1 gene mutations in both familial and sporadic tumors of endocrine origin on the interactions of menin with the MLL1 and MLL2 histone H3K4 methyltransferase complexes and with JunD-containing transcription factors. Menin binding pocket mutants R52G, E255K and E359K have differential effects on MLL1/MLL2 and JunD interactions, which translate into differential genomic binding patterns. Our findings encourage future studies addressing the pathophysiological relevance of the separate MLL1/MLL2- and JunD-dependent functions of menin mutants in MEN1 disease model systems.
    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
    Meiosis-specific chromatin structures, guided by histone modifications, are critical mediators of a meiotic transient transcription program and progression through prophase I. Histone H3K4 can be methylated up to three times by the Set1-containing COMPASS complex and each methylation mark corresponds to a different chromatin conformation. The level of H3K4 modification is directed by the activity of additional COMPASS components. In this study, we characterized the role of the COMPASS subunits during meiosis in Saccharomyces cerevisiae. In vegetative cells, previous studies revealed a role for subunits Swd2, Sdc1, and Bre2 for H3K4me2 while Spp1 supported trimethylation. However, we found that Bre2 and Sdc1 are required for H3K4me3 as yeast prepare to enter meiosis while Spp1 is not. Interestingly, we identified distinct meiotic functions for the core COMPASS complex members that required for all H3K4me, Set1, Swd1, and Swd3. While Set1 and Swd1 are required for progression through early meiosis, Swd3 is critical for late meiosis and spore morphogenesis. Furthermore, the meiotic requirement for Set1 is independent of H3K4 methylation, suggesting the presence of nonhistone substrates. Finally, checkpoint suppression analyses indicate that Set1 and Swd1 are required for both homologous recombination and chromosome segregation. These data suggest that COMPASS has important new roles for meiosis that are independent of its well-characterized functions during mitotic divisions.
    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
    发育控制基因的Pax家族在人类疾病中经常失调。在肾脏,Pax2在发育中的肾单位中表达,但在成人近端和远端小管中不表达,而多囊肾上皮或肾细胞癌持续表达高水平。小鼠或细胞培养物中的Pax2减少可以减缓囊性上皮细胞或肾癌细胞的增殖。因此,抑制Pax活性可能是可行的,细胞类型特异性治疗。我们设计了一个无偏见的,基于细胞,高通量筛选,鉴定出削弱Pax反式激活能力的三唑并嘧啶衍生物。我们显示BG-1抑制Pax2阳性癌细胞生长和靶基因表达,但对Pax2阴性细胞影响不大。染色质免疫沉淀表明,这些抑制剂可防止Pax蛋白与肾细胞中Pax靶基因处的组蛋白H3K4甲基化复合物相互作用。因此,这些化合物可以为具有治疗潜力的肾脏特异性抑制剂提供结构支架.
    The Pax family of developmental control genes are frequently deregulated in human disease. In the kidney, Pax2 is expressed in developing nephrons but not in adult proximal and distal tubules, whereas polycystic kidney epithelia or renal cell carcinoma continues to express high levels. Pax2 reduction in mice or cell culture can slow proliferation of cystic epithelial cells or renal cancer cells. Thus, inhibition of Pax activity may be a viable, cell-type-specific therapy. We designed an unbiased, cell-based, high-throughput screen that identified triazolo pyrimidine derivatives that attenuate Pax transactivation ability. We show that BG-1 inhibits Pax2-positive cancer cell growth and target gene expression but has little effect on Pax2-negative cells. Chromatin immunoprecipitation suggests that these inhibitors prevent Pax protein interactions with the histone H3K4 methylation complex at Pax target genes in renal cells. Thus, these compounds may provide structural scaffolds for kidney-specific inhibitors with therapeutic potential.
    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
    组蛋白赖氨酸甲基转移酶2(KMT2)蛋白形成多聚体酶复合物,其在基因组中的转录调节元件处将组蛋白H3(H3K4)上的赖氨酸4甲基化。H3K4甲基化与活性转录的强烈关联导致人们努力揭示KMT2复合物在转录调控中的功能参与。许多生化和细胞研究表明,KMT2复合物通过H3K4甲基化调节靶基因的转录。然而,在许多情况下,KMT2复合物酶活性的丧失不能完全解释观察到的转录缺陷。越来越多的证据表明,在某些情况下,KMT2复合物介导的转录调控可以H3K4甲基化非依赖性方式发生。这里,我们全面回顾了KMT2复合物在基因表达中的功能,重点介绍我们目前所知道的KMT2复合物调节转录的分子机制。我们还讨论了KMT2复合物的异常转录调控如何导致不同的人类疾病,比如癌症。
    Histone lysine methyltransferase 2 (KMT2) proteins form multimeric enzymatic complexes that methylate lysine 4 on histone H3 (H3K4) at transcription regulatory elements in the genome. A strong association of H3K4 methylation with active transcription has led to intense efforts to reveal the functional involvement of KMT2 complexes in transcriptional regulation. A number of biochemical and cellular studies have shown that KMT2 complexes regulate transcription of target genes via H3K4 methylation. However, in many cases, loss of KMT2 complex enzymatic activity fails to fully account for observed transcriptional defects. Accumulating evidence indicates that, in certain contexts, KMT2 complex-mediated transcriptional regulation can occur in an H3K4 methylation-independent manner. Here, we comprehensively review functions of KMT2 complexes in gene expression, focusing on what we currently know about the molecular mechanisms by which the KMT2 complexes regulate transcription. We also discuss how aberrant transcriptional regulation by KMT2 complexes contributes to different human diseases, such as cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    一个核小体包含每个组蛋白H2A的两个拷贝,H2B,H3和H4。组蛋白H3K4me0和K36me3是哺乳动物DNA甲基转移酶催化DNA从头甲基化的两个关键染色质标志。然而,尚不清楚两个姐妹组蛋白H3s上的K4me0和K36me3标记是否独立或协同调节DNA从头甲基化。这里,利用酵母中的二价组蛋白H3系统,我们研究了K4和K36对姐妹组蛋白H3s对异位共表达的鼠Dnmt3a和Dnmt3L催化的基因组DNA甲基化的贡献。结果表明,一个姐妹H3尾巴上缺少K4me0和K36me3,或缺乏K4me0和K36me3在各自的姐妹H3导致5mC的急剧减少,揭示了两个姐妹H3在DNA甲基化调控中的协同作用。因此,破坏Dnmt3aADD结构域-H3K4me0,Dnmt3LADD结构域-H3K4me0或Dnmt3aPWWP结构域-H3K36me3相互作用的Dnmt3a或Dnmt3L突变导致DNA甲基化显著减少.这些结果支持以下模型:每个异二聚体Dnmt3a-Dnmt3L在姐妹H3s的一条尾巴上同时读取K4me0和K36me3标记,异二聚体Dnmt3a-Dnmt3L的二聚体识别姐妹组蛋白H3s的两个尾巴,以有效地执行从头DNA甲基化。
    A nucleosome contains two copies of each histone H2A, H2B, H3 and H4. Histone H3 K4me0 and K36me3 are two key chromatin marks for de novo DNA methylation catalyzed by DNA methyltransferases in mammals. However, it remains unclear whether K4me0 and K36me3 marks on both sister histone H3s regulate de novo DNA methylation independently or cooperatively. Here, taking advantage of the bivalent histone H3 system in yeast, we examined the contributions of K4 and K36 on sister histone H3s to genomic DNA methylation catalyzed by ectopically co-expressed murine Dnmt3a and Dnmt3L. The results show that lack of both K4me0 and K36me3 on one sister H3 tail, or lack of K4me0 and K36me3 on respective sister H3s results in a dramatic reduction of 5mC, revealing a synergy of two sister H3s in DNA methylation regulation. Accordingly, the Dnmt3a or Dnmt3L mutation that disrupts the interaction of Dnmt3aADD domain-H3K4me0, Dnmt3LADD domain-H3K4me0, or Dnmt3aPWWP domain-H3K36me3 causes a significant reduction of DNA methylation. These results support the model that each heterodimeric Dnmt3a-Dnmt3L reads both K4me0 and K36me3 marks on one tail of sister H3s, and the dimer of heterodimeric Dnmt3a-Dnmt3L recognizes two tails of sister histone H3s to efficiently execute de novo DNA methylation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    The histone H3 lysine 4 (H3K4) presenter WDR5 forms protein complexes with H3K4 methyltransferases MLL1-MLL4 and binding partner proteins including RBBP5, ASH2L, and DPY30, and plays a key role in histone H3K4 trimethylation, chromatin remodeling, transcriptional activation of target genes, normal biology, and diseases such as MLL-rearranged leukemia. By forming protein complexes with other proteins such as Myc, WDR5 induces transcriptional activation of key oncogenes, tumor cell cycle progression, DNA replication, cell proliferation, survival, tumor initiation, progression, invasion, and metastasis of cancer of a variety of organ origins. Several small molecule MLL/WDR5 protein-protein interaction inhibitors, such as MM-401, MM-589, WDR5-0103, Piribedil, and OICR-9429, have been confirmed to reduce H3K4 trimethylation, oncogenic gene expression, cell cycle progression, cancer cell proliferation, survival and resistance to chemotherapy without general toxicity to normal cells. Derivatives of the MLL/WDR5 interaction inhibitors with improved pharmacokinetic properties and in vivo bioavailability are expected to have the potential to be trialed in cancer patients.
    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
    BACKGROUND: Histone posttranslational modifications (PTMs) represent a focal point of chromatin regulation. The genome-wide and locus-specific distribution and the presence of distinct histone PTMs is most commonly examined with the application of histone PTM-specific antibodies. In spite of their central role in chromatin research, polyclonal antibodies suffer from disadvantages like batch-to-batch variability and insufficient documentation of their quality and specificity.
    RESULTS: To mitigate some of the pitfalls of using polyclonal antibodies against H3K4me3, we successfully validated the application of a recombinant TAF3 PHD domain as anti-H3K4me3 affinity reagent in peptide array, western blot and ChIP-like experiments coupled with qPCR and deep sequencing.
    CONCLUSIONS: The successful addition of the TAF3 PHD domain to the growing catalog of recombinant affinity reagents for histone PTMs could help to improve the reproducibility, interpretation and cross-laboratory validation of chromatin data.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    A transcriptional repressor Gfi1 promotes T helper type 2 (Th2) cell development and inhibits Th17 and inducible regulatory T-cell differentiation. However, the role of Gfi1 in regulating Th1 cell differentiation and the Th1-type immune response remains to be investigated. We herein demonstrate that Gfi1 inhibits the induction of the Th1 programme in activated CD4 T cells. The activated Gfi1-deficient CD4 T cells spontaneously develop into Th1 cells in an interleukin-12- and interferon-γ-independent manner. The increase of Th1-type immune responses was confirmed in vivo in Gfi1-deficient mice using a murine model of nickel allergy and delayed-type hypersensitivity (DTH). The expression levels of Th1-related transcription factors were found to increase in Gfi1-deficient activated CD4 T cells. Tbx21, Eomes and Runx2 were identified as possible direct targets of Gfi1. Gfi1 binds to the Tbx21, Eomes and Runx2 gene loci and reduces the histone H3K4 methylation levels in part by modulating Lsd1 recruitment. Together, these findings demonstrate a novel regulatory role of Gfi1 in the regulation of the Th1-type immune response.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号