histones

组蛋白
  • 文章类型: Journal Article
    基因表达是一个复杂的生物学过程,它弥合了基因型和表型之间的差距。典型和可遗传的表观遗传机制,如组蛋白和DNA修饰,调节DNA编码的遗传信息的释放,而不改变潜在的序列。许多其他非规范玩家,如染色质调节子和非编码RNA,也参与调节基因表达。最近,RNA修饰(表观转录组学)已被证明在塑造细胞转录组方面具有巨大的潜力。然而,它们的共转录性质和不确定的遗传力意味着它们超出了表观遗传学的当前定义,引发了该领域正在进行的辩论。在这里,我们将讨论控制基因表达的规范和非规范表观遗传机制之间的关系,并提供我们对是否(或是否)表观遗传修饰可以归类为表观遗传机制的观点。
    Gene expression is an intricate biological process that bridges gap between the genotype and the phenotype. Canonical and hereditable epigenetic mechanisms, such as histone and DNA modifications, regulate the release of genetic information encoded in DNA without altering the underlying sequence. Many other non-canonical players, such as chromatin regulators and noncoding RNAs, are also involved in regulating gene expression. Recently, RNA modifications (epitranscriptomics) have been shown to hold enormous potential in shaping cellular transcriptomes. However, their co-transcriptional nature and uncertain heritability mean that they fall outside the current definition of epigenetics, sparking an ongoing debate in the field. Here we will discuss the relationship between canonical and non-canonical epigenetic mechanisms that govern gene expression and offer our perspective on whether (or not) epitranscriptomic modifications can be classified as epigenetic mechanisms.
    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
    目的:乳腺癌的治疗方法之一是手术切除肿瘤和术后放疗预防复发。不幸的是,由于癌细胞对电离辐射的敏感性较低,因此放射治疗并不总是足够有效。本研究旨在评估白藜芦醇的放射增敏特性,piceatannol和虎杖苷对乳腺癌细胞,它们的表面上存在激素受体。
    方法:实验部分在三阴性乳腺癌细胞(HCC38)和激素依赖性细胞(MCF7)上进行。这项研究评估了细胞死亡的水平,基因表达的变化(BAX,BCL-2)和与凋亡过程相关的蛋白质(CASPASE3、8和P53),抗氧化酶表达的变化(过氧化氢酶,SOD,GPx1/2)和NRF-2。此外,RAD51蛋白和组蛋白H2AX的表达水平,参与DNA修复过程,被评估。通过双向方差分析(ANOVA)和随后的Tukey事后检验(p<0.05)评估统计学显著性。
    结果:与白藜芦醇或皮卡坦诺联合使用的电离辐射通过内部和外部途径激活细胞凋亡过程。与HCC38细胞相比,MCF7细胞对电离辐射的更高敏感性与白藜芦醇组合与细胞的抗氧化反应较弱和DNA损伤修复强度降低有关。由电离辐射诱导的DNA修复在HCC38细胞中比在MCF7细胞中更有效地发生。
    结论:白藜芦醇对两种细胞系的二苯乙烯具有最高的放射增敏潜能。与HCC38细胞相比,MCF7中电离辐射与白藜芦醇(在较小程度上与piceatannol)组合的有效性更为显着。
    OBJECTIVE: One of the treatments for breast cancer is surgical resection of the tumour and prevention of recurrence with postoperative radiotherapy. Unfortunately, radiotherapy is not always effective enough due to the low sensitivity of cancer cells to ionising radiation. This study aimed to evaluate the radiosensitising properties of resveratrol, piceatannol and polydatin on breast cancer cells, which differ in the presence of hormonal receptors on their surface.
    METHODS: The experimental part was carried out on triple-negative breast cancer cells (HCC38) and hormone-dependent cells (MCF7). The study assessed the level of cell death, changes in the expression of genes (BAX, BCL-2) and proteins related to the apoptosis process (CASPASE 3, 8 and P53), changes in the expression of antioxidant enzymes (CATALASE, SOD, GPx1/2) and NRF-2. Additionally, the expression level of RAD51 protein and histone H2AX, which are involved in DNA repair processes, was assessed. Statistical significance was evaluated by a two-way analysis of variance (ANOVA) followed by Tukey\'s post hoc test (p < 0.05).
    RESULTS: Ionising radiation in combination with resveratrol or piceatannol activates the apoptosis process by internal and external pathways. Greater sensitivity of MCF7 cells compared to HCC38 cells to ionising radiation in combination with resveratrol is associated with a weaker antioxidant response of cells and reduced intensity of DNA damage repair. DNA repair induced by ionising radiation occurs more effectively in HCC38 cells than in MCF7 cells.
    CONCLUSIONS: Resveratrol has the highest radiosensitising potential among the tested stilbene for cells of both lines. The effectiveness of ionizing radiation in combination with resveratrol (to a lesser extent with piceatannol) is more significant in MCF7 than in HCC38 cells.
    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
    变位现象描述了两个等位基因之间的相互作用,其中一个等位基因启动另一个等位基因的遗传表观遗传转换而不影响DNA序列。由于变位的表观遗传转化伴随着DNA和/或组蛋白甲基化模式的变化,影响基因表达。对植物和动物中的副突变的研究已经确定了小的非编码RNA作为启动基因座中的表观遗传变化所需的主要效应分子。由于小的非编码RNA可以跨代传播,变构效应可以在群体中遗传和维持。在这次审查中,我们将系统分析到目前为止描述的不同生命系统中的参数化例子,强调生物体之间的顺突变的共同和不同的分子和遗传方面,并考虑这种现象在进化中的作用。
    The phenomenon of paramutation describes the interaction between two alleles, in which one allele initiates inherited epigenetic conversion of another allele without affecting the DNA sequence. Epigenetic transformations due to paramutation are accompanied by the change in DNA and/or histone methylation patterns, affecting gene expression. Studies of paramutation in plants and animals have identified small non-coding RNAs as the main effector molecules required for the initiation of epigenetic changes in gene loci. Due to the fact that small non-coding RNAs can be transmitted across generations, the paramutation effect can be inherited and maintained in a population. In this review, we will systematically analyze examples of paramutation in different living systems described so far, highlighting common and different molecular and genetic aspects of paramutation between organisms, and considering the role of this phenomenon in evolution.
    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
    表观遗传改变,如染色质结构和DNA甲基化,已经在许多肿瘤类型中进行了广泛的研究。但是口腔癌,尤其是口腔腺癌,受到的关注要少得多。这里,我们将激光捕获显微切割和muti-omics小型批量测序相结合,系统地表征口腔癌的表观遗传景观,包括染色质结构,DNA甲基化,H3K27me3修改,和基因表达。在致癌作用中,肿瘤细胞表现出重组的染色质空间结构,包括受损的区室结构和改变的基因-基因相互作用网络。值得注意的是,在表型非恶性癌旁细胞中观察到一些结构改变,但在正常细胞中未观察到。我们开发了转换模型来识别个体基因组基因座的癌症倾向,从而确定每个样品的致癌状态。对癌症表观遗传景观的见解提供了证据,表明染色质重组是口腔癌进展的重要标志。这也与基因组改变和DNA甲基化重编程有关。特别是,癌细胞中拷贝数频繁变化的区域与癌症和正常样本中强烈的空间绝缘相关。口腔鳞状细胞癌中异常甲基化重编程与染色质结构和H3K27me3信号密切相关,进一步受到内在序列属性的影响。我们的发现表明,在两种不同类型的口腔癌中,结构变化既重要又保守。与转录组改变和癌症发展密切相关。值得注意的是,尽管与鳞状细胞癌相比基因组拷贝数改变的发生率相当低,甲基化改变的程度较小,但在口腔腺癌中结构改变仍然明显明显.我们期望对不同类型和亚型的原发性口腔肿瘤的表观遗传重编程的综合分析可以为口腔癌的新型检测和治疗的设计提供额外的指导。
    Epigenetic alterations, such as those in chromatin structure and DNA methylation, have been extensively studied in a number of tumor types. But oral cancer, particularly oral adenocarcinoma, has received far less attention. Here, we combined laser-capture microdissection and muti-omics mini-bulk sequencing to systematically characterize the epigenetic landscape of oral cancer, including chromatin architecture, DNA methylation, H3K27me3 modification, and gene expression. In carcinogenesis, tumor cells exhibit reorganized chromatin spatial structures, including compromised compartment structures and altered gene-gene interaction networks. Notably, some structural alterations are observed in phenotypically non-malignant paracancerous but not in normal cells. We developed transformer models to identify the cancer propensity of individual genome loci, thereby determining the carcinogenic status of each sample. Insights into cancer epigenetic landscapes provide evidence that chromatin reorganization is an important hallmark of oral cancer progression, which is also linked with genomic alterations and DNA methylation reprogramming. In particular, regions of frequent copy number alternations in cancer cells are associated with strong spatial insulation in both cancer and normal samples. Aberrant methylation reprogramming in oral squamous cell carcinomas is closely related to chromatin structure and H3K27me3 signals, which are further influenced by intrinsic sequence properties. Our findings indicate that structural changes are both significant and conserved in two distinct types of oral cancer, closely linked to transcriptomic alterations and cancer development. Notably, the structural changes remain markedly evident in oral adenocarcinoma despite the considerably lower incidence of genomic copy number alterations and lesser extent of methylation alterations compared to squamous cell carcinoma. We expect that the comprehensive analysis of epigenetic reprogramming of different types and subtypes of primary oral tumors can provide additional guidance to the design of novel detection and therapy for oral cancer.
    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
    异染色质的表观遗传遗传需要在复制过程中将亲本H3-H4四聚体转移到两个子双链体。最近的三篇论文利用酵母遗传学与遗传测定相结合,以及AlphaFold2多聚体预测与生物化学相结合,揭示了复制体成分(Mrc1/CLASPIN)是H3-H4四聚体伴侣,对于亲本组蛋白转移到女儿来说很重要。
    Epigenetic inheritance of heterochromatin requires transfer of parental H3-H4 tetramers to both daughter duplexes during replication. Three recent papers exploit yeast genetics coupled to inheritance assays and AlphaFold2-multimer predictions coupled to biochemistry to reveal that a replisome component (Mrc1/CLASPIN) is an H3-H4 tetramer chaperone important for parental histone transfer to daughters.
    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
    G9a是一种组蛋白甲基转移酶,催化组蛋白3赖氨酸9(H3K9)的甲基化,参与基因表达的调控。我们先前报道了G9a在体内和体外发育的肌腱中表达,并且G9a缺陷的肌腱细胞在体外表现出受损的增殖和分化。在这项研究中,我们通过使用G9a条件敲除(G9acKO)小鼠研究了G9a在体内肌腱发育中的功能。我们将Sox9Cre/+小鼠与G9afl/fl小鼠交叉以产生G9afl/fl;Sox9Cre/+小鼠。G9acKO小鼠在3周龄时显示出发育不良的肌腱形成。在胚胎第16.5天(E16.5)的溴脱氧尿苷标记显示G9acKO小鼠的肌腱细胞中细胞增殖减少。免疫组织化学分析显示G9a及其底物的表达水平降低,H3K9me2,在G9acKO小鼠的椎体肌腱中。G9acKO小鼠的椎骨和四肢的肌腱组织显示肌腱标记物的表达降低,腱调节素(Tnmd),和col1a1基因,提示肌腱细胞分化被抑制。G9a的过表达导致Tnmd和col1a1在体外肌腱细胞中的表达增强。这些结果表明,G9a在肌腱发育过程中调节肌腱祖细胞的增殖和分化。因此,我们的研究结果表明,G9a在肌腱发育中起着至关重要的作用。
    G9a is a histone methyltransferase that catalyzes the methylation of histone 3 lysine 9 (H3K9), which is involved in the regulation of gene expression. We had previously reported that G9a is expressed in developing tendons in vivo and in vitro and that G9a-deficient tenocytes show impaired proliferation and differentiation in vitro. In this study, we investigated the functions of G9a in tendon development in vivo by using G9a conditional knockout (G9a cKO) mice. We crossed Sox9Cre/+ mice with G9afl/fl mice to generate G9afl/fl; Sox9Cre/+ mice. The G9a cKO mice showed hypoplastic tendon formation at 3 weeks of age. Bromodeoxyuridine labeling on embryonic day 16.5 (E16.5) revealed decreased cell proliferation in the tenocytes of G9a cKO mice. Immunohistochemical analysis revealed decreased expression levels of G9a and its substrate, H3K9me2, in the vertebral tendons of G9a cKO mice. The tendon tissue of the vertebrae and limbs of G9a cKO mice showed reduced expression of a tendon marker, tenomodulin (Tnmd), and col1a1 genes, suggesting that tenocyte differentiation was suppressed. Overexpression of G9a resulted in enhancement of Tnmd and col1a1 expression in tenocytes in vitro. These results suggest that G9a regulates the proliferation and differentiation of tendon progenitor cells during tendon development. Thus, our results suggest that G9a plays an essential role in tendon development.
    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
    赖氨酸特异性组蛋白去甲基酶1(LSD1),将单甲基化或二甲基化组蛋白H3在赖氨酸4(H3K4me1/2)上,对早期胚胎发生和发育至关重要。在这里,我们表明LSD1对于小鼠胚胎干细胞(ESC)的自我更新是不必要的,但对于小鼠ESC的生长和分化是必需的。重新引入催化受损的LSD1(LSD1MUT)可以恢复小鼠ESC的增殖能力,然而,LSD1的酶活性对于确保适当的分化至关重要。的确,Lsd1敲除(KO)小鼠ESC中H3K4me1的增加不会导致与干性相关的全局基因表达程序的重大变化。然而,LSD1而非LSD1MUT的消融导致DNMT1和UHRF1蛋白减少,并伴随整体低甲基化.我们表明,LSD1和LSD1MUT都通过与HDAC1和泛素特异性肽酶7(USP7)的相互作用来控制UHRF1和DNMT1的蛋白质稳定性,因此,促进DNMT1和UHRF1的去乙酰化和去泛素化。我们的研究阐明了LSD1控制小鼠ESCDNA甲基化的机制,独立于其赖氨酸脱甲基酶活性。
    Lysine-specific histone demethylase 1 (LSD1), which demethylates mono- or di- methylated histone H3 on lysine 4 (H3K4me1/2), is essential for early embryogenesis and development. Here we show that LSD1 is dispensable for mouse embryonic stem cell (ESC) self-renewal but is required for mouse ESC growth and differentiation. Reintroduction of a catalytically-impaired LSD1 (LSD1MUT) recovers the proliferation capability of mouse ESCs, yet the enzymatic activity of LSD1 is essential to ensure proper differentiation. Indeed, increased H3K4me1 in Lsd1 knockout (KO) mouse ESCs does not lead to major changes in global gene expression programs related to stemness. However, ablation of LSD1 but not LSD1MUT results in decreased DNMT1 and UHRF1 proteins coupled to global hypomethylation. We show that both LSD1 and LSD1MUT control protein stability of UHRF1 and DNMT1 through interaction with HDAC1 and the ubiquitin-specific peptidase 7 (USP7), consequently, facilitating the deacetylation and deubiquitination of DNMT1 and UHRF1. Our studies elucidate a mechanism by which LSD1 controls DNA methylation in mouse ESCs, independently of its lysine demethylase activity.
    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H3突变神经胶质瘤是致命的脑肿瘤,其特征是表观基因组失调和分化停滞。与肿瘤细胞上可用的大量数据集相反,关于他们的肿瘤微环境(TME)的信息有限,尤其是免疫渗透。这里,我们表征了H3.3K27M和G34R/V突变神经胶质瘤的免疫TME,和多个H3.3K27M小鼠模型,使用转录组学,蛋白质组学和空间单细胞方法。免疫谱系的分辨率表明H3突变神经胶质瘤具有不同的髓系群体的高浸润,免疫检查点标志物的高水平表达,和稀缺的淋巴样细胞,结果在所有测试的H3.3K27M小鼠模型中一致再现。我们展示了这些髓样群体与H3突变细胞进行通信,介导免疫抑制和维持肿瘤的形成和维持。骨髓细胞和免疫检查点途径的双重抑制在临床前同基因小鼠模型中显示出显著的治疗益处。我们的发现提供了有价值的肿瘤组蛋白突变神经胶质瘤的TME表征,并深入了解调节骨髓浸润的方法,以造福患者。
    Histone H3-mutant gliomas are deadly brain tumors characterized by a dysregulated epigenome and stalled differentiation. In contrast to the extensive datasets available on tumor cells, limited information exists on their tumor microenvironment (TME), particularly the immune infiltrate. Here, we characterize the immune TME of H3.3K27M and G34R/V-mutant gliomas, and multiple H3.3K27M mouse models, using transcriptomic, proteomic and spatial single-cell approaches. Resolution of immune lineages indicates high infiltration of H3-mutant gliomas with diverse myeloid populations, high-level expression of immune checkpoint markers, and scarce lymphoid cells, findings uniformly reproduced in all H3.3K27M mouse models tested. We show these myeloid populations communicate with H3-mutant cells, mediating immunosuppression and sustaining tumor formation and maintenance. Dual inhibition of myeloid cells and immune checkpoint pathways show significant therapeutic benefits in pre-clinical syngeneic mouse models. Our findings provide a valuable characterization of the TME of oncohistone-mutant gliomas, and insight into the means for modulating the myeloid infiltrate for the benefit of patients.
    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
    在本期的神经元中,Torres-Berrío等人1表明,由于SUZ12VEFS域的作用,应激易感小鼠在伏隔核神经元中表现出升高的H3K27me1水平,加强特定表观遗传变化与长期压力引起的社会之间的联系,情感,和认知改变。
    In this issue of Neuron, Torres-Berrío et al.1 show that stress-susceptible mice exhibit elevated H3K27me1 levels in nucleus accumbens neurons due to the action of the SUZ12 VEFS domain, strengthening the link between specific epigenetic changes and long-lasting stress-induced social, emotional, and cognitive alterations.
    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:速效抗抑郁药(RAAD),包括解离性麻醉药,迷幻药,和移态药,通过据称调节神经可塑性来引发精神疾病的快速和持续的治疗改善,神经传递,和豁免权。这些结果可能是由,或导致,表观遗传过程的急性和/或持续夹带,重塑染色质结构并改变DNA可及性以调节基因表达。
    方法:从这个角度来看,我们概述了已知的机制,知识差距,以及围绕RAAD表观遗传效应的未来方向,专注于压力反应性DNA和大脑区域的调节,以及与常规抗抑郁药的比较。
    方法:初步相关证据表明,RAAD的给药伴随着表观遗传效应,这与常规抗抑郁药引起的效应相似。这些包括DNA甲基化的变化,组蛋白的翻译后修饰,和非编码RNA在涉及神经营养的应激反应染色质区域的差异调节,神经传递,和免疫调节,在应激反应的大脑区域。这些表观遗传变化是否对RAAD的治疗效果有因果关系,是其后果,或者是无关的,仍然未知。此外,潜在的细胞类型特异性和相关机制尚未完全阐明。候选机制包括神经元活性和5-羟色胺和原肌苷受体激酶B(TRKB)信号介导的表观遗传变化,与DNA的直接相互作用,组蛋白,或染色质重塑复合物。
    结论:相关证据表明,RAAD诱导的表观遗传变化伴随治疗和副作用,虽然因果关系,机制,和细胞类型特异性仍然很大程度上未知。解决这些研究空白可能会导致新的基于神经表观遗传学的精确疗法的发展。
    BACKGROUND: Rapid-acting antidepressants (RAADs), including dissociative anesthetics, psychedelics, and empathogens, elicit rapid and sustained therapeutic improvements in psychiatric disorders by purportedly modulating neuroplasticity, neurotransmission, and immunity. These outcomes may be mediated by, or result in, an acute and/or sustained entrainment of epigenetic processes, which remodel chromatin structure and alter DNA accessibility to regulate gene expression.
    METHODS: In this perspective, we present an overview of the known mechanisms, knowledge gaps, and future directions surrounding the epigenetic effects of RAADs, with a focus on the regulation of stress-responsive DNA and brain regions, and on the comparison with conventional antidepressants.
    METHODS: Preliminary correlative evidence indicates that administration of RAADs is accompanied by epigenetic effects which are similar to those elicited by conventional antidepressants. These include changes in DNA methylation, post-translational modifications of histones, and differential regulation of non-coding RNAs in stress-responsive chromatin areas involved in neurotrophism, neurotransmission, and immunomodulation, in stress-responsive brain regions. Whether these epigenetic changes causally contribute to the therapeutic effects of RAADs, are a consequence thereof, or are unrelated, remains unknown. Moreover, the potential cell type-specificity and mechanisms involved are yet to be fully elucidated. Candidate mechanisms include neuronal activity- and serotonin and Tropomyosine Receptor Kinase B (TRKB) signaling-mediated epigenetic changes, and direct interaction with DNA, histones, or chromatin remodeling complexes.
    CONCLUSIONS: Correlative evidence suggests that epigenetic changes induced by RAADs accompany therapeutic and side effects, although causation, mechanisms, and cell type-specificity remain largely unknown. Addressing these research gaps may lead to the development of novel neuroepigenetics-based precision therapeutics.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号