NSD3

NSD3
  • 文章类型: Journal Article
    核受体结合SET结构域(NSD)蛋白是一类组蛋白赖氨酸甲基转移酶,涉及多种癌症类型,异常表达和参与癌症相关信号通路。在这项研究中,通过基于结构的虚拟筛选,针对NSD的SET结构域鉴定了包括化合物2和3的一系列小分子化合物。我们的先导化合物3在体外对NSD2-SET和NSD3-SET表现出有效的抑制活性,IC50为0.81μM和0.84μM,分别,并有效抑制组蛋白H3赖氨酸36二甲基化,并在100nM时降低非小细胞肺癌细胞中NSD靶向基因的表达。化合物3抑制H460和H1299非小细胞肺癌细胞中的细胞增殖并降低克隆性,并诱导s期细胞周期停滞和凋亡。这些数据使我们的化合物成为研究NSD在癌症中的生物学作用的有价值的工具包。
    Nuclear receptor binding SET domain (NSD) proteins are a class of histone lysine methyltransferases and implicated in multiple cancer types with aberrant expression and involvement of cancer related signaling pathways. In this study, a series of small-molecule compounds including compound 2 and 3 are identified against the SET domain of NSDs through structure-based virtual screening. Our lead compound 3 exhibits potent inhibitory activities in vitro towards the NSD2-SET and NSD3-SET with an IC50 of 0.81 μM and 0.84 μM, respectively, and efficiently inhibits histone H3 lysine 36 dimethylation and decreases the expression of NSDs-targeted genes in non-small cell lung cancer cells at 100 nM. Compound 3 suppresses cell proliferation and reduces the clonogenicity in H460 and H1299 non-small cell lung cancer cells, and induces s-phase cell cycle arrest and apoptosis. These data establish our compounds as a valuable tool-kit for the study of the biological roles of NSDs in cancer.
    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
    NSD3(核受体结合SET结构域蛋白3)是NSD组蛋白甲基转移酶蛋白家族的成员。近年来,它已被确定为某些类型癌症的潜在癌基因。NSD3基因编码三种亚型,长版本(NSD3L),一个简短的版本(NSD3S)和WHISTLE亚型。重要的是,NSD3S同工型对应于全长蛋白的N末端区域,缺乏甲基转移酶结构域。NSD3的染色体位置经常在癌症类型中扩增,如乳房,肺,和结肠,在其他人中。最近,这种扩增与染色体增生事件相关,这可以解释在癌症中发现的不同的NSD3改变。含有NSD3的融合蛋白在白血病中也有报道(NSD3-NUP98),和NUT(睾丸核蛋白)中线癌(NSD3-NUT)。其作为癌基因的作用已被描述为通过其甲基转移酶活性调节不同的癌症途径,或者蛋白质的短同工型,通过蛋白质相互作用。具体来说,在这篇综述中,我们将重点讨论被表征为甲基转移酶依赖性的功能,以及与NSD3S同工型表达相关的那些。有证据表明,NSD3L和NSD3S同工型与癌症进展有关,建立NSD3作为治疗靶标。然而,需要进一步的功能研究来区分NSD3致癌活性是否依赖于或不依赖于蛋白质的催化结构域,以及每种同工型的贡献及其在癌症进展中的临床意义。
    NSD3 (nuclear receptor-binding SET domain protein 3) is a member of the NSD histone methyltransferase family of proteins. In recent years, it has been identified as a potential oncogene in certain types of cancer. The NSD3 gene encodes three isoforms, the long version (NSD3L), a short version (NSD3S) and the WHISTLE isoforms. Importantly, the NSD3S isoform corresponds to the N-terminal region of the full-length protein, lacking the methyltransferase domain. The chromosomal location of NSD3 is frequently amplified across cancer types, such as breast, lung, and colon, among others. Recently, this amplification has been correlated to a chromothripsis event, that could explain the different NSD3 alterations found in cancer. The fusion proteins containing NSD3 have also been reported in leukemia (NSD3-NUP98), and in NUT (nuclear protein of the testis) midline carcinoma (NSD3-NUT). Its role as an oncogene has been described by modulating different cancer pathways through its methyltransferase activity, or the short isoform of the protein, through protein interactions. Specifically, in this review we will focus on the functions that have been characterized as methyltransferase dependent, and those that have been correlated with the expression of the NSD3S isoform. There is evidence that both the NSD3L and NSD3S isoforms are relevant for cancer progression, establishing NSD3 as a therapeutic target. However, further functional studies are needed to differentiate NSD3 oncogenic activity as dependent or independent of the catalytic domain of the protein, as well as the contribution of each isoform and its clinical significance in cancer progression.
    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
    核受体结合SET结构域蛋白3(NSD3)最近被认为是对抗癌症的新表观遗传靶标。NSD3被扩增,在多种肿瘤中过度表达或突变,通过调节细胞周期促进肿瘤的发展,凋亡,DNA修复和EMT。因此,抑制,沉默或敲低NSD3是非常有前途的抗肿瘤策略。本文总结了NSD3的结构和生物学功能,重点介绍了其致癌或促癌症活性。本文还对NSD3特异性抑制剂或降解剂的发展进行了讨论和综述。
    Nuclear receptor binding SET domain protein 3 (NSD3) has recently been recognized as a new epigenetic target in the fight against cancer. NSD3, which is amplified, overexpressed or mutated in a variety of tumors, promotes tumor development by regulating the cell cycle, apoptosis, DNA repair and EMT. Therefore, the inhibition, silencing or knockdown of NSD3 are highly promising antitumor strategies. This paper summarizes the structure and biological functions of NSD3 with an emphasis on its carcinogenic or cancer-promoting activity. The development of NSD3-specific inhibitors or degraders is also discussed and reviewed in this paper.
    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
    姐妹染色单体内聚力是在整个细胞周期中实施的多步骤过程,以确保染色体正确传递到子细胞。尽管已经广泛探索了内聚力的建立和有丝分裂内聚力的溶解,对粘附素负载的调节仍然知之甚少。这里,我们报告说,在有丝分裂进入之前,甲基转移酶NSD3对于有丝分裂姐妹染色单体的凝聚力至关重要。NSD3与粘蛋白加载剂复合kollerin(由NIPBL和MAU2组成)相互作用,并促进有丝分裂出口处MAU2和粘蛋白的染色质募集。我们还表明,NSD3在后期早期与染色质相关,在招募MAU2和RAD21之前,并在前期开始时与染色质分离。在体细胞中存在的两种NSD3亚型中,长亚型负责调节kollerin和cohesin染色质负荷,有效的姐妹染色单体内聚力需要其甲基转移酶活性。基于这些观察,我们认为,NSD3依赖性甲基化通过确保适当的kollerin募集并因此确保coheresin加载有助于姐妹染色单体的内聚力。
    Sister chromatid cohesion is a multi-step process implemented throughout the cell cycle to ensure the correct transmission of chromosomes to daughter cells. Although cohesion establishment and mitotic cohesion dissolution have been extensively explored, the regulation of cohesin loading is still poorly understood. Here, we report that the methyltransferase NSD3 is essential for mitotic sister chromatid cohesion before mitosis entry. NSD3 interacts with the cohesin loader complex kollerin (composed of NIPBL and MAU2) and promotes the chromatin recruitment of MAU2 and cohesin at mitotic exit. We also show that NSD3 associates with chromatin in early anaphase, prior to the recruitment of MAU2 and RAD21, and dissociates from chromatin when prophase begins. Among the two NSD3 isoforms present in somatic cells, the long isoform is responsible for regulating kollerin and cohesin chromatin-loading, and its methyltransferase activity is required for efficient sister chromatid cohesion. Based on these observations, we propose that NSD3-dependent methylation contributes to sister chromatid cohesion by ensuring proper kollerin recruitment and thus cohesin loading.
    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
    姐妹染色单体内聚力是在整个细胞周期中实施的多步骤过程,以确保染色体正确传递到子细胞。虽然已经广泛探索了内聚建立和有丝分裂内聚溶解,对粘附素负载的调节仍然知之甚少。这里,我们报告说,在有丝分裂进入之前,甲基转移酶NSD3对于有丝分裂姐妹染色单体的凝聚力至关重要。NSD3与粘附素加载剂复合物kollerin(NIPBL/MAU2)相互作用,并促进有丝分裂出口处MAU2和粘附素的染色质募集。我们还表明,NSD3在后期早期与染色质相关,在招募MAU2和RAD21之前,并在前期开始时与染色质分离。在体细胞中存在的两种NSD3亚型中,长亚型负责调节kollerin和cohesin染色质负荷,有效的姐妹染色单体内聚力需要其甲基转移酶活性。基于这些观察,我们认为,NSD3依赖性甲基化通过确保适当的kollerin募集并因此确保coheresin加载有助于姐妹染色单体的内聚力。
    Sister chromatid cohesion is a multi-step process implemented throughout the cell cycle to ensure the correct transmission of chromosomes to daughter cells. While cohesion establishment and mitotic cohesion dissolution have been extensively explored, the regulation of cohesin loading is still poorly understood. Here, we report that the methyltransferase NSD3 is essential for mitotic sister chromatid cohesion before mitosis entry. NSD3 interacts with the cohesin loader complex kollerin (NIPBL/MAU2) and promotes the chromatin recruitment of MAU2 and cohesin at mitotic exit. We also show that NSD3 associates with chromatin in early anaphase, prior to the recruitment of MAU2 and RAD21, and dissociates from chromatin when prophase begins. Among the two NSD3 isoforms present in somatic cells, the long isoform is responsible for regulating kollerin and cohesin chromatin-loading, and its methyltransferase activity is required for efficient sister chromatid cohesion. Based on these observations, we propose that NSD3-dependent methylation contributes to sister chromatid cohesion by ensuring proper kollerin recruitment and thus cohesin loading.
    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:组蛋白H3赖氨酸36甲基转移酶的核受体结合SET域(NSD)家族成员包括NSD1,NSD2(MMSET/WHSC1),和NSD3(Wolf-Hirschhorn综合征候选1-like1,WHSC1L1)。虽然NSD基因的表达对正常生物过程和癌症至关重要,他们的表达水平对癌症预后的了解尚不清楚。
    方法:我们分析了多种癌症类型中NSD家族基因的表达模式,并检查了它们与临床特征和患者生存状况的关联。接下来,我们探索了NSD3表达与PAAD中肿瘤微环境(TME)的描述特征之间的关联,一种严重的胰腺癌.特别是,我们将NSD3启动子甲基化水平与PAAD患者预后相关.最后,我们使用一系列胰腺癌细胞实验,探索了NSD3的推定致癌作用.
    结果:我们报道NSD家族成员的表达与多种癌症的临床预后相关。此外,我们证明NSD3变异在我们分析的所有癌症中的NSD基因中最为普遍.值得注意的是,当与NSD1和NSD2相比时,我们发现NSD3被突出地表达,其表达与胰腺癌的临床预后显著相关。此外,NSD3经常被扩增,表现出低启动子甲基化,并与胰腺癌的免疫细胞浸润和增殖增强有关。最后,我们证明NSD3的敲低改变了H3K36me2甲基化,胰腺癌细胞的下游基因表达和EGFR/ERK信号传导。
    结论:我们发现表达水平,NSD家族基因的遗传变异的存在,以及它们的启动子甲基化与癌症的临床结果相关,包括胰腺癌.我们的体外实验表明,NSD3可能与胰腺癌的基因表达调控和生长因子信号传导有关。
    Members of the nuclear receptor-binding SET domain (NSD) family of histone H3 lysine 36 methyltransferases comprise NSD1, NSD2 (MMSET/WHSC1), and NSD3 (Wolf-Hirschhorn syndrome candidate 1-like 1, WHSC1L1). While the expression of NSD genes is essential to normal biological processes and cancer, knowledge of their expression levels to prognosticate in cancer remains unclear.
    We analyzed the expression patterns for NSD family genes across multiple cancer types and examined their association with clinical features and patient survival profiles. Next, we explored the association between NSD3 expression and described features of the tumor microenvironment (TME) in PAAD, a severe type of pancreatic cancer. In particular, we correlated promoter methylation levels for NSD3 with patient outcomes in PAAD. Finally, we explored the putative oncogenic roles for NSD3 using a series of experiments with pancreatic cancer cells.
    We report that the expression of NSD family members is correlated with clinical prognosis across multiple types of cancers. Also, we demonstrate that NSD3 variants are most prevalent among NSD genes across cancers we analyzed. Notably, when compared with NSD1 and NSD2, we find that NSD3 is prominently expressed, and its expression is significantly linked with clinical outcome in pancreatic cancer. Furthermore, NSD3 is frequently amplified, exhibits low promoter methylation, and is correlated with immune cell infiltration and enhanced proliferation of pancreatic cancer. Finally, we demonstrate that knockdown of NSD3 alters H3K36me2 methylation, downstream gene expression and EGFR/ERK signaling in pancreatic cancer cells.
    We find that expression levels, the presence of genetic variants of NSD family genes, as well as their promoter methylation are correlated with clinical outcomes in cancer, including pancreatic cancer. Our in vitro experiments suggest that NSD3 may be relevant to gene expression regulation and growth factor signaling in pancreatic 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
    背景:拷贝数改变是癌症中常见的遗传病变。在鳞状非小细胞肺癌中,最常见的拷贝数改变的基因座在染色体3q26-27和8p11.23。在8p11.23扩增的鳞状肺癌中可能是驱动因素的基因尚不清楚。
    方法:与拷贝数改变有关的数据,位于8p11.23扩增区域的基因的mRNA表达和蛋白质表达从各种来源中提取,包括癌症基因组图谱,人类蛋白质图谱和卡普兰迈耶绘图仪。使用cBioportal平台分析基因组数据。使用KaplanMeierPlotter平台对扩增病例与非扩增病例进行生存分析。
    结果:在11.5%至17.7%的肺鳞癌中,8p11.23位点扩增。最常扩增的基因包括NSD3、FGFR1和LETM2。只有一些扩增的基因在mRNA水平上存在伴随的过表达。这些包括NSD3,PLPP5,DDHD2,LSM1和ASH2L,而其他基因显示出低水平的相关性,而且仍然,与拷贝中性样品相比,基因座中的一些基因没有显示mRNA过表达。大多数基因座基因的蛋白质产物在鳞状肺癌中表达。在8p11.23扩增的鳞状细胞肺癌与非扩增的癌症中,总生存率没有显着差异。此外,mRNA过表达对任何扩增基因的无复发存活均无不良影响.
    结论:肺鳞状细胞癌中常见扩增位点8p11.23的一些基因是假定的致癌候选基因。基因座着丝粒部分的基因子集,比端粒部分更常见的扩增,显示高伴随的mRNA表达。
    BACKGROUND: Copy number alterations are common genetic lesions in cancer. In squamous non-small cell lung carcinomas, the most common copy-number-altered loci are at chromosomes 3q26-27 and 8p11.23. The genes that may be drivers in squamous lung cancers with 8p11.23 amplifications are unclear.
    METHODS: Data pertaining to copy number alterations, mRNA expression and protein expression of genes located in the 8p11.23 amplified region were extracted from various sources including The Cancer Genome Atlas, the Human Protein Atlas and the Kaplan Meier Plotter. Genomic data were analyzed using the cBioportal platform. Survival analysis of cases with amplifications compared to nonamplified cases was performed using the Kaplan Meier Plotter platform.
    RESULTS: The 8p11.23 locus is amplified in 11.5% to 17.7% of squamous lung carcinomas. The most frequently amplified genes include NSD3, FGFR1 and LETM2. Only some of the amplified genes present concomitant overexpression at the mRNA level. These include NSD3, PLPP5, DDHD2, LSM1 and ASH2L, while other genes display lower levels of correlation, and still, some genes in the locus show no mRNA overexpression compared with copy-neutral samples. The protein products of most locus genes are expressed in squamous lung cancers. No significant difference in overall survival in 8p11.23-amplified squamous cell lung cancers versus nonamplified cancers is observed. In addition, there is no adverse effect of mRNA overexpression for relapse-free survival of any of the amplified genes.
    CONCLUSIONS: Several genes that are part of the commonly amplified locus 8p11.23 in squamous lung carcinomas are putative oncogenic candidates. A subset of genes of the centromeric part of the locus, which is amplified more commonly than the telomeric part, show high concomitant mRNA 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
    FGFR1的邻近基因组蛋白H3赖氨酸36(H3K36)甲基转移酶NSD3已被确定为肺鳞状细胞癌(LUSC)的关键遗传驱动因素。然而,分子特征,特别是NSD3在驱动癌变中的免疫学作用,知之甚少。在这项研究中,我们系统地整合了多组学数据(例如,基因组,转录组,蛋白质组,和TMA阵列)以解剖NSD3扩增的LUSC中的免疫学谱。接下来,我们实施了药物-转录组相关性分析,以确定LUSC的分子基础和治疗脆弱性.我们发现,NSD3扩增的LUSC在多个独立的LUSC患者队列中呈现非发炎的肿瘤免疫微环境(TIME)状态。可以预见,升高的NSD3表达与较差的免疫治疗结果相关.进一步的分子表征表明,未折叠蛋白反应(UPR)信号传导的高活性可能是NSD3扩增的LUSC的非免疫原性表型的关键介质。和谐地,我们发现,与野生型组相比,NSD3扩增的LUSC对靶向UPR分支的化合物表现出更敏感的表型.简而言之,我们的多水平分析指出了NSD3以前未被重视的免疫学作用,并为NSD3扩增的鳞状肺癌提供了治疗依据.
    The histone H3 lysine 36 (H3K36) methyltransferase NSD3, a neighboring gene of FGFR1, has been identified as a critical genetic driver of lung squamous cell carcinoma (LUSC). However, the molecular characteristics, especially the immunological roles of NSD3 in driving carcinogenesis, are poorly understood. In this study, we systematically integrated multi-omics data (e.g., genome, transcriptome, proteome, and TMA array) to dissect the immunological profiles in NSD3-amplified LUSC. Next, pharmaco-transcriptomic correlation analysis was implemented to identify the molecular underpinnings and therapeutic vulnerabilities in LUSC. We revealed that NSD3-amplified LUSC presents a non-inflamed tumor immune microenvironment (TIME) state in multiple independent LUSC patient cohorts. Predictably, elevated NSD3 expression was correlated with a worse immunotherapy outcome. Further molecular characterizations revealed that the high activity of unfolded protein response (UPR) signaling might be a pivotal mediator for the non-immunogenic phenotype of NSD3-amplified LUSC. Concordantly, we showed that NSD3-amplified LUSCs exhibited a more sensitive phenotype to compounds targeting UPR branches than the wild-type group. In brief, our multi-level analyses point to a previously unappreciated immunological role for NSD3 and provide therapeutic rationales for NSD3-amplified squamous lung cancer.
    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
    核受体结合SET结构域蛋白3(NSD3)是人类癌症治疗中具有吸引力的潜在靶标。在这里,我们报道了一系列基于蛋白水解靶向嵌合体(PROTAC)策略的小分子NSD3降解物的发现.所表示的化合物8在NCI-H1703和A549肺癌细胞中诱导NSD3降解,DC50值为1.43和0.94μM,分别,并显示对其他两种NSD蛋白的选择性。图8减少组蛋白H3赖氨酸36甲基化并诱导肺癌细胞的凋亡和细胞周期停滞。此外,RNA测序和免疫组织化学检测显示,8例下调NSD3相关基因表达.重要的是,如图8所示,但不是1(报道的NSD3-PWWP拮抗剂)可以抑制NCI-H1703和A549细胞的细胞生长。8的单次施用有效地降低了肺癌异种移植模型中的NSD3蛋白水平。因此,这项研究表明,诱导NSD3降解是比阻断NSD3-PWWP结构域更有效的抑制NSD3功能的方法,这可能为肺癌提供潜在的治疗方法。
    Nuclear receptor binding SET domain protein 3 (NSD3) is an attractive potential target in the therapy for human cancers. Herein, we report the discovery of a series of small-molecule NSD3 degraders based on the proteolysis targeting chimera (PROTAC) strategy. The represented compound 8 induces NSD3 degradation with DC50 values of 1.43 and 0.94 μM in NCI-H1703 and A549 lung cancer cells, respectively, and shows selectivity over two other NSD proteins. 8 reduces histone H3 lysine 36 methylation and induces apoptosis and cell cycle arrest in lung cancer cells. Moreover, the RNA sequencing and immunohistochemistry assays showed that 8 downregulates NSD3-associated gene expression. Significantly, 8, but not 1 (a reported NSD3-PWWP antagonist) could inhibit the cell growth of NCI-H1703 and A549 cells. A single administration of 8 effectively decreases the NSD3 protein level in lung cancer xenograft models. Therefore, this study demonstrated that inducing NSD3 degradation is a more effective approach inhibiting the function of NSD3 than blocking the NSD3-PWWP domain, which may provide a potential therapeutic approach for lung cancer.
    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H3在赖氨酸36(H3K36me)的甲基化对于维持基因组稳定性至关重要。的确,这个甲基化标记对于正确的转录是必不可少的,重组,和DNA损伤反应。H3K36甲基转移酶中的功能缺失和获得突变与人类发育障碍和各种癌症密切相关。结构分析表明,核小体成分,如接头DNA和由组蛋白H2A和H3构成的疏水斑块,除了组蛋白H3尾之外,可能是H3K36甲基化的决定因素。其包括H3K36和催化SET结构域。H3K36甲基转移酶与核小体的相互作用与其自抑制性变化的调节相配合,微调了H3K36me介导NSD2和NSD3的二甲基化以及Set2/SETD2的三甲基化的精确度。鉴定与不同形式的H3K36me结合的特定结构特征和各种顺式作用因子,特别是H3K36的二(H3K36me2)和三(H3K36me3)甲基化形式,突出了H3K36me功能意义的复杂性。这里,我们巩固了这些发现,并为H3K36me2向H3K36me3转化的调节提供了结构见解。我们还讨论了H3K36me与其他染色质修饰之间合作的机制(特别是,H3K27me3,H3乙酰化,RNA中的DNA甲基化和N6-甲基腺苷)在染色质表观基因组功能的生理调节中。
    The methylation of histone H3 at lysine 36 (H3K36me) is essential for maintaining genomic stability. Indeed, this methylation mark is essential for proper transcription, recombination, and DNA damage response. Loss- and gain-of-function mutations in H3K36 methyltransferases are closely linked to human developmental disorders and various cancers. Structural analyses suggest that nucleosomal components such as the linker DNA and a hydrophobic patch constituted by histone H2A and H3 are likely determinants of H3K36 methylation in addition to the histone H3 tail, which encompasses H3K36 and the catalytic SET domain. Interaction of H3K36 methyltransferases with the nucleosome collaborates with regulation of their auto-inhibitory changes fine-tunes the precision of H3K36me in mediating dimethylation by NSD2 and NSD3 as well as trimethylation by Set2/SETD2. The identification of specific structural features and various cis-acting factors that bind to different forms of H3K36me, particularly the di-(H3K36me2) and tri-(H3K36me3) methylated forms of H3K36, have highlighted the intricacy of H3K36me functional significance. Here, we consolidate these findings and offer structural insight to the regulation of H3K36me2 to H3K36me3 conversion. We also discuss the mechanisms that underlie the cooperation between H3K36me and other chromatin modifications (in particular, H3K27me3, H3 acetylation, DNA methylation and N6-methyladenosine in RNAs) in the physiological regulation of the epigenomic functions of chromatin.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号