LSD1

LSD1
  • 文章类型: Journal Article
    转移占乳腺癌相关死亡人数的近90%,使其频繁发生恶性肿瘤,也是全球女性肿瘤死亡的主要原因。LSD1是一种组蛋白去甲基酶,在乳腺癌中起着重要作用。为了探讨LSD1对乳腺癌侵袭和迁移的影响,我们用LSD1敲除的MCF7和T47D外泌体处理乳腺癌细胞,乳腺癌细胞的侵袭和迁移能力显著增强。这一现象表明LSD1可以抑制乳腺癌细胞的侵袭和迁移。miR-1290表达在LSD1敲低MCF7外泌体中下调。通过分析miR-1290靶基因NAT1的数据库,验证miR-1290能够调控NAT1的表达。这些数据通过证明表观遗传因子LSD1如何通过控制外泌体miRNA刺激乳腺癌细胞的侵袭和迁移,为乳腺癌治疗的生物学提供了新的见解。
    Metastasis accounts for almost 90% of breast cancer-related fatalities, making it frequent malignancy and the main reason of tumor mortality globally among women. LSD1 is a histone demethylase, which plays an important role in breast cancer. In order to explore the effect of LSD1 on invasion and migration of breast cancer, we treated breast cancer cells with MCF7 and T47D exosomes knocked down by LSD1, and the invasion and migration of breast cancer cells were significantly enhanced. This phenomenon indicates that LSD1 can inhibit the invasion and migration of breast cancer cells. miR-1290 expression was downregulated in LSD1 knockdown MCF7 exosomes. By analyzing the database of miR-1290 target gene NAT1, we verified that miR-1290 could regulate the expression of NAT1. These data provide fresh insights into the biology of breast cancer therapy by demonstrating how the epigenetic factor LSD1 stimulates the breast cancer cells\' invasion and migration via controlling exosomal miRNA.
    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
    靶向HSP90和LSD1轴的药效团的合理安装已经在前列腺癌和结肠直肠癌中实现了显著的抗癌能力。在一系列的混合动力车中,抑制剂6对前列腺癌细胞系PC-3和DU145表现出明显的抗增殖活性,GI50值为0.24和0.30μM,分别。它在针对凋亡的组合攻击和细胞死亡起始中表现出显著的功效。细胞死亡过程由PARP诱导和γH2AX信号介导,并且还表征为胱天蛋白酶依赖性和Bcl-xL/Bax非依赖性。值得注意的是,与参照组(AUY922)相比,在用化合物6处理的斑马鱼中未观察到眼睛大小或形态的差异。多西他赛耐药PC-3细胞中的深刻治疗反应突出了改善多西他赛敏感性的双重抑制能力。此外,最低浓度为1.25μM,化合物6在体外有效抑制患者来源的结直肠癌(CRC)类器官的生长长达10天。一起,设计的HSP90/LSD1抑制剂为抗癌药物治疗提供了新的途径和显著的临床价值.
    The rational installation of pharmacophores targeting HSP90 and LSD1 axes has achieved significant anti-cancer capacity in prostate and colorectal cancer. Among the series of hybrids, inhibitor 6 exhibited remarkable anti-proliferative activity against prostate cancer cell lines PC-3 and DU145, with GI50 values of 0.24 and 0.30 μM, respectively. It demonstrated notable efficacy in combinatorial attack and cell death initiation towards apoptosis. The cell death process was mediated by PARP induction and γH2AX signaling, and was also characterized as caspase-dependent and Bcl-xL/Bax-independent. Notably, no difference in eye size or morphology was observed in the zebrafish treated with compound 6 compared to the reference group (AUY922). The profound treatment response in docetaxel-resistant PC-3 cells highlighted the dual inhibitory ability in improving docetaxel sensitivity. Additionally, at a minimum concentration of 1.25 μM, compound 6 effectively inhibited the growth of patient-derived colorectal cancer (CRC) organoids for up to 10 days in vitro. Together, the designed HSP90/LSD1 inhibitors present a novel route and significant clinical value for anti-cancer drug therapy.
    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
    组蛋白赖氨酸特异性脱甲基酶1(LSD1)在三阴性乳腺癌(TNBC)中经常过表达,这与TNBC患者的临床预后较差有关。然而,LSD1促进TNBC进展的潜在机制尚待确定.我们最近通过将乳腺条件性LSD1敲除小鼠与Brca1缺陷小鼠杂交,建立了基因工程小鼠模型,以探索LSD1在TNBC发病机理中的作用。Cre介导的Brca1丢失导致小鼠乳腺肿瘤形成的发生率更高,LSD1的同时耗竭阻碍了这一点,表明LSD1在促进Brca1缺陷型肿瘤中的关键作用。我们还证明了肿瘤抑制基因的沉默,组织因子途径抑制因子2(TFPI2),在功能上与LSD1介导的TNBC进展相关。与正常乳腺组织相比,小鼠Brca1缺陷型肿瘤表现出升高的LSD1表达和降低的TFPI2水平。对TCGA数据库的分析显示,在侵袭性ER阴性或基底样BC中,TFPI2的表达显着降低。通过LSD1抑制恢复TFPI2增加了TFPI2启动子处的H3K4me2富集,抑制肿瘤进展,和增强化疗药物的抗肿瘤功效。通过LSD1消融诱导TFPI2下调基质金属蛋白酶(MMPs)的活性,进而增加肿瘤环境中细胞毒性T淋巴细胞吸引趋化因子的水平,导致CD8+T细胞的肿瘤浸润增强。此外,TFPI2的诱导增强LSD1抑制剂的抗肿瘤作用和免疫检查点阻断在低免疫原性TNBC中。一起,我们的研究确定了TFPI2在LSD1介导的TNBC进展中的先前未被识别的作用,治疗反应,和免疫原性作用。
    Histone lysine-specific demethylase 1 (LSD1) is frequently overexpressed in triple negative breast cancer (TNBC), which is associated with worse clinical outcome in TNBC patients. However, the underlying mechanisms by which LSD1 promotes TNBC progression remain to be identified. We recently established a genetically engineered murine model by crossing mammary gland conditional LSD1 knockout mice with Brca1-deficient mice to explore the role of LSD1 in TNBC pathogenesis. Cre-mediated Brca1 loss led to higher incidence of tumor formation in mouse mammary glands, which was hindered by concurrent depletion of LSD1, indicating a critical role of LSD1 in promoting Brca1-deficient tumors. We also demonstrated that the silencing of a tumor suppressor gene, Tissue Factor Pathway Inhibitor 2 (TFPI2), is functionally associated with LSD1-mediated TNBC progression. Mouse Brca1-deficient tumors exhibited elevated LSD1 expression and decreased TFPI2 level compared to normal mammary tissues. Analysis of TCGA database revealed that TFPI2 expression is significantly lower in aggressive ER-negative or basal-like BC. Restoration of TFPI2 through LSD1 inhibition increased H3K4me2 enrichment at the TFPI2 promoter, suppressed tumor progression, and enhanced antitumor efficacy of chemotherapeutic agent. Induction of TFPI2 by LSD1 ablation downregulates activity of matrix metalloproteinases (MMPs) that in turn increases the level of cytotoxic T lymphocyte attracting chemokines in tumor environment, leading to enhanced tumor infiltration of CD8+ T cells. Moreover, induction of TFPI2 potentiates antitumor effect of LSD1 inhibitor and immune checkpoint blockade in poorly immunogenic TNBC. Together, our study identifies previously unrecognized roles of TFPI2 in LSD1-mediated TNBC progression, therapeutic response, and immunogenic effects.
    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
    N6-甲基腺苷(m6A)甲基化是与药物成瘾相关的重要表观遗传机制。然而,m6A修饰与羟考酮奖励之间的关系研究较少。基于开放的现场测试,本研究使用染色质免疫沉淀PCR评估羟考酮奖励,免疫荧光,和RNA测序。以剂量和时间依赖性方式观察到纹状体神经元中羟考酮丰度引起的METTL14蛋白的显着增加和PP1α蛋白的减少。羟考酮显著增加LSD1表达,纹状体中H3K4me1表达降低。在野外测试中,纹状体内注射METTL14siRNA,HOTAIRsiRNA,或LSD1shRNA阻断羟考酮诱导的运动活性增加。用METTL14/HOTAIRsiRNA和LSD1shRNA处理后,PP1α的下调也受到抑制。LSD1shRNA也逆转了由羟考酮诱导的LSD1与CoRest的增强结合以及CoRest与PP1α基因的增强结合。此外,H3K4me1去甲基化也被处理阻断。总之,研究证实,METTL14介导的HOTAIR上调导致PP1α抑制,这反过来促进了LSD1的募集,从而催化H3K4me1去甲基化并促进羟考酮成瘾。
    N6-methyladenosine (m6A) methylation is a vital epigenetic mechanism associated with drug addiction. However, the relationship between m6A modification and oxycodone rewarding is less well explored. Based on an open field test, the present study evaluated oxycodone rewarding using chromatin immunoprecipitation PCR, immunofluorescence, and RNA sequencing. A marked increase in METTL14 protein and a decrease in PP1α protein due to oxycodone abundance in the striatal neurons were observed in a dose- and time-dependent manner. Oxycodone markedly increased LSD1 expression, and decreased H3K4me1 expression in the striatum. In the open field test, intra-striatal injection of METTL14 siRNA, HOTAIR siRNA, or LSD1 shRNA blocked oxycodone-induced increase in locomotor activity. The downregulation of PP1α was also inhibited after treatment with METTL14/HOTAIR siRNA and LSD1 shRNA. Enhanced binding of LSD1 with CoRest and of CoRest with the PP1α gene induced by oxycodone was also reversed by LSD1 shRNA. In addition, H3K4me1 demethylation was also blocked by the treatment. In summary, the investigation confirmed that METTL14-mediated upregulation of HOTAIR resulted in the repression of PP1α, which in turn facilitated the recruitment of LSD1, thus catalyzing H3K4me1 demethylation and promoting oxycodone addiction.
    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
    表观遗传学研究面临着染色质修饰网络的高度复杂性和严格调控的挑战。尽管已经描述了染色质介导的基因调控的许多分离机制,作为更大的表观基因组网络的一部分,缺乏全面分析特定过程的可靠方法。为了通过一个有助于捕获和描述转录调控复杂性的系统来扩展方法的工具箱,我们在这里描述了一个强大的协议,用于生成稳定的转录活性报告系统,并总结了它们的应用。该系统允许将染色质调节剂诱导募集到荧光报告基因,然后用流式细胞术检测转录变化。报告基因整合到内源性染色质环境中,从而能够检测所研究的染色质调节剂对内源性辅因子的调节依赖性。该系统允许在单细胞水平上的简单和动态的读出以及补偿转录的细胞间差异的能力。系统的模块化设计可以简单地调整方法,以研究广泛的细胞系中的不同染色质调节剂。我们还总结了该技术在表征不同染色质效应子的沉默速度方面的应用,去除激活组蛋白修饰,表观基因组修饰的稳定性和可逆性分析,研究小分子对染色质效应子和功能效应子-共调节子关系的影响。所提出的方法允许研究活细胞中表观遗传效应蛋白的转录调控的复杂性。
    Epigenetic research faces the challenge of the high complexity and tight regulation in chromatin modification networks. Although many isolated mechanisms of chromatin-mediated gene regulation have been described, solid approaches for the comprehensive analysis of specific processes as parts of the bigger epigenome network are missing. In order to expand the toolbox of methods by a system that will help to capture and describe the complexity of transcriptional regulation, we describe here a robust protocol for the generation of stable reporter systems for transcriptional activity and summarize their applications. The system allows for the induced recruitment of a chromatin regulator to a fluorescent reporter gene, followed by the detection of transcriptional changes using flow cytometry. The reporter gene is integrated into an endogenous chromatin environment, thus enabling the detection of regulatory dependencies of the investigated chromatin regulator on endogenous cofactors. The system allows for an easy and dynamic readout at the single-cell level and the ability to compensate for cell-to-cell variances of transcription. The modular design of the system enables the simple adjustment of the method for the investigation of different chromatin regulators in a broad panel of cell lines. We also summarize applications of this technology to characterize the silencing velocity of different chromatin effectors, removal of activating histone modifications, analysis of stability and reversibility of epigenome modifications, the investigation of the effects of small molecule on chromatin effectors and of functional effector-coregulator relationships. The presented method allows to investigate the complexity of transcriptional regulation by epigenetic effector proteins in living 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
    表观遗传调节剂,如赖氨酸特异性脱甲基酶1(LSD1)和组蛋白脱乙酰酶(HDAC)是癌症的药物靶标,神经精神疾病,或者炎症,但是这些酶的抑制剂表现出相当大的副作用。对于减少全身毒性的潜在局部治疗,我们在这里介绍作为新的LSD1和HDAC抑制剂的软药物候选物。软药物是在实现其治疗功能后在体内降解为活性较低的代谢物的化合物。这已成功应用于临床皮质类固醇,但是针对表观遗传酶的软药物很少,HDAC抑制剂remetinostat是唯一的例子。我们已经开发了靶向LSD1或HDAC的新型含甲酯的抑制剂,并将这些抑制剂的生物活性与它们各自的羧酸裂解产物进行了比较。体外活性测定,细胞实验,和稳定性测定鉴定了强效的HDAC和LSD1软药物候选物,其在细胞模型中优于其相应的羧酸。
    Epigenetic modulators such as lysine-specific demethylase 1 (LSD1) and histone deacetylases (HDACs) are drug targets for cancer, neuropsychiatric disease, or inflammation, but inhibitors of these enzymes exhibit considerable side effects. For a potential local treatment with reduced systemic toxicity, we present here soft drug candidates as new LSD1 and HDAC inhibitors. A soft drug is a compound that is degraded in vivo to less active metabolites after having achieved its therapeutic function. This has been successfully applied for corticosteroids in the clinic, but soft drugs targeting epigenetic enzymes are scarce, with the HDAC inhibitor remetinostat being the only example. We have developed new methyl ester-containing inhibitors targeting LSD1 or HDACs and compared the biological activities of these to their respective carboxylic acid cleavage products. In vitro activity assays, cellular experiments, and a stability assay identified potent HDAC and LSD1 soft drug candidates that are superior to their corresponding carboxylic acids in cellular models.
    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
    卵巢癌(OCa)是所有妇科癌症中最致命的。OCa的标准治疗是铂类化疗,如卡铂或顺铂与紫杉醇联合使用。大多数患者最初对这些治疗有反应;然而,近90%的患者会复发,不可避免地会死于化疗耐药的疾病。最近的研究表明,表观遗传修饰剂赖氨酸特异性组蛋白去甲基酶1A(KDM1A/LSD1)在OCa中高度过表达。然而,KDM1A在化疗耐药中的作用以及其抑制是否能增强OCa的化疗应答仍不确定.对TCGA数据集的分析显示,对化疗反应不佳的患者中KDM1A表达较高。Westernblot分析显示化疗药物顺铂治疗,卡铂,紫杉醇增加了OCa细胞中KDM1A的表达。KDM1A敲低(KD)或用KDM1A抑制剂NCD38和SP2509治疗可使已建立的患者来源的OCa细胞对化疗药物敏感,从而降低细胞活力和克隆存活并诱导细胞凋亡。此外,KDM1A致敏的卡铂耐药A2780-CP70细胞对卡铂治疗和紫杉醇耐药SKOV3-TR细胞对紫杉醇的敲除。RNA-seq分析显示,KDM1A-KD和顺铂治疗的组合导致与上皮-间质转化(EMT)相关的基因下调。有趣的是,顺铂治疗增加了NF-κB通路基因的子集,和KDM1A-KD或KDM1A抑制逆转了这种作用。重要的是,KDM1A-KD,联合顺铂,在原位囊内OCa异种移植模型中,与单一治疗相比,肿瘤生长显着降低。总的来说,这些研究结果表明,KDM1A抑制剂联合化疗可能是治疗OCa的一种有前景的治疗方法.
    Ovarian cancer (OCa) is the deadliest of all gynecological cancers. The standard treatment for OCa is platinum-based chemotherapy, such as carboplatin or cisplatin in combination with paclitaxel. Most patients are initially responsive to these treatments; however, nearly 90% will develop recurrence and inevitably succumb to chemotherapy-resistant disease. Recent studies have revealed that the epigenetic modifier lysine-specific histone demethylase 1A (KDM1A/LSD1) is highly overexpressed in OCa. However, the role of KDM1A in chemoresistance and whether its inhibition enhances chemotherapy response in OCa remains uncertain. Analysis of TCGA datasets revealed that KDM1A expression is high in patients who poorly respond to chemotherapy. Western blot analysis show that treatment with chemotherapy drugs cisplatin, carboplatin, and paclitaxel increased KDM1A expression in OCa cells. KDM1A knockdown (KD) or treatment with KDM1A inhibitors NCD38 and SP2509 sensitized established and patient-derived OCa cells to chemotherapy drugs in reducing cell viability and clonogenic survival and inducing apoptosis. Moreover, knockdown of KDM1A sensitized carboplatin-resistant A2780-CP70 cells to carboplatin treatment and paclitaxel-resistant SKOV3-TR cells to paclitaxel. RNA-seq analysis revealed that a combination of KDM1A-KD and cisplatin treatment resulted in the downregulation of genes related to epithelial-mesenchymal transition (EMT). Interestingly, cisplatin treatment increased a subset of NF-κB pathway genes, and KDM1A-KD or KDM1A inhibition reversed this effect. Importantly, KDM1A-KD, in combination with cisplatin, significantly reduced tumor growth compared to a single treatment in an orthotopic intrabursal OCa xenograft model. Collectively, these findings suggest that combination of KDM1A inhibitors with chemotherapy could be a promising therapeutic approach for the treatment of OCa.
    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
    组蛋白是染色质的主要成分,作为维持染色体结构和调节基因表达的指导性支架。组蛋白修饰的失调与各种病理过程有关,特别是癌症的发生和发展,组蛋白甲基化起着关键作用。然而,癌症中组蛋白甲基化的具体机制和潜在治疗靶点尚未阐明.Lys特异性脱甲基酶1A(LSD1)是第一个鉴定的脱甲基酶,可从赖氨酸4或赖氨酸9处的组蛋白3中特异性去除甲基,充当基因表达的阻遏物或激活物。最近的研究表明,LSD1以多种表观遗传调控或非表观遗传方式促进癌症进展。值得注意的是,LSD1功能障碍与抑制性癌症免疫相关。已经开发了许多LSD1抑制剂,临床试验正在探索其在单一疗法中的疗效。或与其他疗法相结合。在这次审查中,本文综述了LSD1的致癌机制和LSD1抑制剂的应用现状。我们强调LSD1是癌症治疗的有希望的靶标。本综述将为进一步了解肿瘤学和表观遗传学的研究进展提供最新的理论参考。加深对癌症表观遗传学的最新认识。
    Histones are the main components of chromatin, functioning as an instructive scaffold to maintain chromosome structure and regulate gene expression. The dysregulation of histone modification is associated with various pathological processes, especially cancer initiation and development, and histone methylation plays a critical role. However, the specific mechanisms and potential therapeutic targets of histone methylation in cancer are not elucidated. Lys-specific demethylase 1A (LSD1) was the first identified demethylase that specifically removes methyl groups from histone 3 at lysine 4 or lysine 9, acting as a repressor or activator of gene expression. Recent studies have shown that LSD1 promotes cancer progression in multiple epigenetic regulation or non-epigenetic manners. Notably, LSD1 dysfunction is correlated with repressive cancer immunity. Many LSD1 inhibitors have been developed and clinical trials are exploring their efficacy in monotherapy, or combined with other therapies. In this review, we summarize the oncogenic mechanisms of LSD1 and the current applications of LSD1 inhibitors. We highlight that LSD1 is a promising target for cancer treatment. This review will provide the latest theoretical references for further understanding the research progress of oncology and epigenetics, deepening the updated appreciation of epigenetics 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
    牙周炎(PD)是一种与牙周炎细菌相关的多因素炎性疾病。赖氨酸特异性脱甲基酶1(LSD1),一种组蛋白去甲基酶,通过与miRNA靶标结合,参与了口腔疾病中炎症反应过程的调节。这项研究调查了miRNA与LSD1结合的分子机制及其对成骨分化的后续作用。首先,分离人牙周膜干细胞(hPDLSCs),培养,和特点。然后对这些细胞进行脂多糖(LPS)处理以诱导炎症,之后开始成骨分化。采用qPCR和蛋白质印迹来监测LSD1表达的变化。随后,在hPDLSCs中沉默LSD1以评估其对成骨分化的影响。通过生物信息学和双荧光素酶报告基因测定,预测并确认miR-708-3p为LSD1的靶miRNA。随后,评估miR-708-3p表达,并通过过表达评估其在PDhPDLSCs中的作用。使用染色质免疫沉淀(ChIP)和蛋白质印迹测定,我们探索了miR-708-3p和LSD1通过二甲基化H3K4(H3K4me2)对osterix(OSX)转录的潜在调节。最后,我们研究了OSX在hPDLSCs中的作用。LPS处理hPDLSCs后,LSD1的表达增加,但是这种趋势在诱导成骨分化后被逆转。沉默LSD1加强了hPDLSCs的成骨分化。miR-708-3p被发现直接结合并负调节LSD1,导致通过H3K4me2的去甲基化抑制OSX转录。此外,miR-708-3p的过表达可促进hPDLSCs在炎症微环境中成骨分化。然而,OSX的表达降低部分减弱了保护作用。我们的发现表明miR-708-3p靶向调节LSD1以通过H3K4me2甲基化增强OSX转录,最终促进hPDLSCs成骨分化。
    Periodontitis (PD) is a multifactorial inflammatory disease associated with periodontopathic bacteria. Lysine-specific demethylase 1 (LSD1), a type of histone demethylase, has been implicated in the modulation of the inflammatory response process in oral diseases by binding to miRNA targets. This study investigates the molecular mechanisms by which miRNA binds to LSD1 and its subsequent effect on osteogenic differentiation. First, human periodontal ligament stem cells (hPDLSCs) were isolated, cultured, and characterized. These cells were then subjected to lipopolysaccharide (LPS) treatment to induce inflammation, after which osteogenic differentiation was initiated. qPCR and western blot were employed to monitor changes in LSD1 expression. Subsequently, LSD1 was silenced in hPDLSCs to evaluate its impact on osteogenic differentiation. Through bioinformatics and dual luciferase reporter assay, miR-708-3p was predicted and confirmed as a target miRNA of LSD1. Subsequently, miR-708-3p expression was assessed, and its role in hPDLSCs in PD was evaluated through overexpression. Using chromatin immunoprecipitation (ChIP) and western blot assay, we explored the potential regulation of osterix (OSX) transcription by miR-708-3p and LSD1 via di-methylated H3K4 (H3K4me2). Finally, we investigated the role of OSX in hPDLSCs. Following LPS treatment of hPDLSCs, the expression of LSD1 increased, but this trend was reversed upon the induction of osteogenic differentiation. Silencing LSD1 strengthened the osteogenic differentiation of hPDLSCs. miR-708-3p was found to directly bind to and negatively regulate LSD1, leading to the repression of OSX transcription through demethylation of H3K4me2. Moreover, overexpression of miR-708-3p was found to promote hPDLSCs osteogenic differentiation in inflammatory microenvironment. However, the protective effect was partially attenuated by reduced expression of OSX. Our findings indicate that miR-708-3p targetedly regulates LSD1 to enhance OSX transcription via H3K4me2 methylation, ultimately promoting hPDLSCs osteogenic differentiation.
    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
    编码RNA结合蛋白FUS的基因,EWSR1和TAF15(FET蛋白)参与罕见肉瘤的染色体易位。FET重排肉瘤通常是侵袭性恶性肿瘤,影响所有年龄段的患者。需要新的疗法。这些易位将FET基因的5'部分与编码转录因子(TF)的3'伴侣基因融合。所得的融合蛋白是具有FET蛋白低复杂性结构域(LCD)和DNA结合结构域的致癌TF。FET融合蛋白已被证明难以直接靶向,并且有希望的策略靶向关键的共调节剂。一种候选物是赖氨酸特异性脱甲基酶1(LSD1)。LSD1由多个FET融合招募,包括EWSR1::FLI1。LSD1可促进EWSR1::FLI1活性,用非竞争性抑制剂SP-2509治疗可阻断EWSR1::FLI1转录功能。类似的分子,Seclidemstat(SP-2577),目前正在进行FET重排肉瘤的临床试验(NCT03600649)。然而,尚未证明seclidemstat是否具有抗FET融合的药理活性。这里,我们评估了seclidemstat对多种FET重排肉瘤细胞系的体外效力,包括尤因肉瘤,促纤维增生性小圆细胞瘤,透明细胞肉瘤,和粘液样脂肪肉瘤.我们还定义了seclidemstat处理的转录组效应,并评估了seclidemstat对FET融合转录调控的活性。Seclidemstat在FET重排肉瘤的细胞活力测定中显示出有效的活性,并破坏了所有测试融合体的转录功能。尽管表观遗传和靶向抑制剂不太可能在临床上作为单一药物有效,这些数据提示,对于FET重排肉瘤患者,seclidemstat仍是一种有前景的新治疗策略.
    结论:这里,我们展示了非竞争性抑制剂,Seclidemstat,具有针对多种FET融合蛋白的体外活性,这些融合蛋白会引起许多罕见且侵袭性的肉瘤。这些数据代表了对多种恶性肿瘤的FET融合活性的最大分析之一,对于研究FET重排肉瘤的人来说是宝贵的资源。
    Genes encoding the RNA-binding proteins FUS, EWSR1, and TAF15 (FET proteins) are involved in chromosomal translocations in rare sarcomas. FET-rearranged sarcomas are often aggressive malignancies affecting patients of all ages. New therapies are needed. These translocations fuse the 5\' portion of the FET gene with a 3\' partner gene encoding a transcription factor (TF). The resulting fusion proteins are oncogenic TFs with a FET protein low complexity domain (LCD) and a DNA binding domain. FET fusion proteins have proven stubbornly difficult to target directly and promising strategies target critical co-regulators. One candidate is lysine specific demethylase 1 (LSD1). LSD1 is recruited by multiple FET fusions, including EWSR1::FLI1. LSD1 promotes EWSR1::FLI1 activity and treatment with the noncompetitive inhibitor SP-2509 blocks EWSR1::FLI1 transcriptional function. A similar molecule, seclidemstat (SP-2577), is currently in clinical trials for FET-rearranged sarcomas (NCT03600649). However, whether seclidemstat has pharmacological activity against FET fusions has not been demonstrated. Here, we evaluate the in vitro potency of seclidemstat against multiple FET-rearranged sarcoma cell lines, including Ewing sarcoma, desmoplastic small round cell tumor, clear cell sarcoma, and myxoid liposarcoma. We also define the transcriptomic effects of seclidemstat treatment and evaluated the activity of seclidemstat against FET fusion transcriptional regulation. Seclidemstat showed potent activity in cell viability assays across FET-rearranged sarcomas and disrupted the transcriptional function of all tested fusions. Though epigenetic and targeted inhibitors are unlikely to be effective as a single agents in the clinic, these data suggest seclidemstat remains a promising new treatment strategy for patients with FET-rearranged sarcomas.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号