oncogene

癌基因
  • 文章类型: Journal Article
    由于在多种癌症中发现的高RNA转录水平,骨髓瘤过表达基因(MYEOV)已被认为是原癌基因。包括骨髓瘤,乳房,肺,胰腺癌和食道癌。人类和其他灵长类动物中开放阅读框(ORF)的存在表明蛋白质编码潜力。然而,我们仍然缺乏功能性MYEOV蛋白的证据。尚未确定MYEOV过表达如何影响癌组织。在这项工作中,我们表明MYEOV可能已经起源,并且仍然可以作为增强剂,调节CCND1和LTO1。首先,使用公开的ATAC-STARR-seq数据证实了人类的MYEOV3增强子活性,在B细胞来源的GM12878细胞上进行。我们在多个健康人体组织中检测到增强子组蛋白标记H3K4me1和H3K27ac重叠MYEOV,其中包括B细胞,肝和肺组织。对3D基因组数据集的分析揭示了MYEOV-3推定增强子与原癌基因CCND1之间的染色质相互作用。BLAST搜索和多序列比对结果表明,这种人类增强子元件的DNA序列与两栖动物/羊膜生物的分歧是保守的,在所有哺乳动物中也发现了273bp的保守区域,甚至在鸡身上,它始终位于相应的CCND1直系同源物附近。此外,我们观察到四个非人灵长类动物的MYEOV直向同源物中的活性增强子状态的保守性,狗,老鼠,和老鼠。当研究小鼠的这个同源区域时,没有MYEOV的ORF,我们不仅观察到了增强子染色质状态,而且使用3D基因组相互作用数据发现了小鼠增强子同源物和Ccnd1之间的相互作用.这类似于在人类中观察到的相互作用,有趣的是,与两个物种的CTCF结合位点一致。一起来看,这表明MYEOV是一种灵长类动物特异性基因,具有起源于进化上较老的增强子区域的从头ORF。这种高度保守的推定增强子元件可以调节人和小鼠的CCND1,开启了使用非灵长类动物模型研究癌症中MYEOV调节功能的可能性。
    The myeloma overexpressed gene (MYEOV) has been proposed to be a proto-oncogene due to high RNA transcript levels found in multiple cancers, including myeloma, breast, lung, pancreas and esophageal cancer. The presence of an open reading frame (ORF) in humans and other primates suggests protein-coding potential. Yet, we still lack evidence of a functional MYEOV protein. It remains undetermined how MYEOV overexpression affects cancerous tissues. In this work, we show that MYEOV has likely originated and may still function as an enhancer, regulating CCND1 and LTO1. Firstly, MYEOV 3\' enhancer activity was confirmed in humans using publicly available ATAC-STARR-seq data, performed on B-cell-derived GM12878 cells. We detected enhancer histone marks H3K4me1 and H3K27ac overlapping MYEOV in multiple healthy human tissues, which include B cells, liver and lung tissue. The analysis of 3D genome datasets revealed chromatin interactions between a MYEOV-3\'-putative enhancer and the proto-oncogene CCND1. BLAST searches and multi-sequence alignment results showed that DNA sequence from this human enhancer element is conserved from the amphibians/amniotes divergence, with a 273 bp conserved region also found in all mammals, and even in chickens, where it is consistently located near the corresponding CCND1 orthologues. Furthermore, we observed conservation of an active enhancer state in the MYEOV orthologues of four non-human primates, dogs, rats, and mice. When studying this homologous region in mice, where the ORF of MYEOV is absent, we not only observed an enhancer chromatin state but also found interactions between the mouse enhancer homolog and Ccnd1 using 3D-genome interaction data. This is similar to the interaction observed in humans and, interestingly, coincides with CTCF binding sites in both species. Taken together, this suggests that MYEOV is a primate-specific gene with a de novo ORF that originated at an evolutionarily older enhancer region. This deeply conserved putative enhancer element could regulate CCND1 in both humans and mice, opening the possibility of studying MYEOV regulatory functions in cancer using non-primate animal models.
    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:本研究的目的是检测肾横纹肌样瘤(RTK)的候选癌基因,并评估其在体外RTK中的作用。
    方法:进行信使RNA(mRNA)和微小RNA(miRNA)测序的整合分析以确定外泌体衍生的miRNA和mRNA在人RTK衍生的细胞系和人胚肾细胞系中的表达谱。进行基因本体富集分析以分析RTK细胞中差异表达的mRNA的功能特征。进行基质胶侵袭和伤口愈合测定以评估细胞侵袭和迁移能力。
    结果:40种mRNA在RTK细胞中高表达,其在RTK细胞中的表达低于对照。这些mRNA主要与细胞粘附有关。在这些mRNA中,我们选择神经纤毛蛋白1(NRP1)作为候选癌基因,因为它的上调表达与几种肿瘤的不良预后相关.敲低NRP1的RTK细胞表现出降低的侵袭和迁移能力。
    结论:我们的研究表明,NRP1通过促进RTK细胞的侵袭和迁移而充当癌基因,并且它可以作为治疗靶标。
    OBJECTIVE: The aim of this study was to detect candidate oncogenes of rhabdoid tumor of the kidney (RTK) and evaluate their roles in RTK in vitro.
    METHODS: An integrated analysis of messenger RNA (mRNA) and microRNA (miRNA) sequencing was performed to determine the expression profile of exosome-derived miRNAs and mRNAs in human RTK-derived cell lines and a human embryonic renal cell line. A Gene Ontology enrichment analysis was performed to analyze the functional characteristics of differentially expressed mRNAs in RTK cells. Matrigel invasion and wound-healing assays were performed to evaluate the cell invasion and migration abilities.
    RESULTS: Forty mRNAs were highly expressed in RTK cells targeted by exosomal miRNAs, the expression of which was lower in RTK cells than in the controls. These mRNAs were primarily related to cell adhesion. Of these mRNAs, we selected neuropilin 1 (NRP1) as a candidate oncogene because its upregulated expression is associated with a poor prognosis of several types of tumors. RTK cells in which NRP1 had been knocked down exhibited decreased invasive and migratory abilities.
    CONCLUSIONS: Our study indicates that NRP1 acts as an oncogene by promoting the invasion and migration of RTK cells and that it could serve as a therapeutic target.
    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
    长链非编码RNA(lncRNA)因其在癌症发展中的致癌或抗肿瘤作用而众所周知。AGAP2-AS1,一种新的lncRNA,已被广泛证明是各种癌症中的致癌lncRNA。大量的实验结果证明AGAP2-AS1在大量恶性肿瘤中异常高水平,比如神经胶质瘤,结直肠,肺,卵巢,前列腺,乳房,胆管癌,膀胱,结肠癌和胰腺癌。重要的是,AGAP2-AS1的生物学功能已被广泛证明。它可以促进扩散,癌细胞的迁移和侵袭。同时,还说明了AGAP2-AS1的临床意义。AGAP2-AS1在各种癌组织中异常过度表达。临床研究表明,AGAP2-AS1的异常过表达与总生存期(OS)密切相关。淋巴结转移(LNM),临床分期,肿瘤浸润,高组织学分级(HG),浆液亚型和PFI时间。然而,到目前为止,AGAP2-AS1在人类癌症中的生物学作用和临床意义尚未得到系统评价。在本次审查中,作者概述了生物学行为,根据先前的研究,AGAP2-AS1的潜在机制和临床特征。总之,AGAP2-AS1,作为一个重要的致癌基因,是一个有前途的生物标志物和癌症预后和治疗的潜在靶点。
    Long non-coding RNAs (lncRNAs) are well known for their oncogenic or anti-oncogenic roles in cancer development. AGAP2-AS1, a new lncRNA, has been extensively demonstrated as an oncogenic lncRNA in various cancers. Abundant experimental results have proved the aberrantly high level of AGAP2-AS1 in a great number of malignancies, such as glioma, colorectal, lung, ovarian, prostate, breast, cholangiocarcinoma, bladder, colon and pancreatic cancers. Importantly, the biological functions of AGAP2-AS1 have been extensively demonstrated. It could promote the proliferation, migration and invasion of cancer cells. Simultaneously, the clinical significances of AGAP2-AS1 were also illustrated. AGAP2-AS1 was exceptionally overexpressed in various cancer tissues. Clinical studies disclosed that the abnormal overexpression of AGAP2-AS1 was tightly connected with overall survival (OS), lymph nodes metastasis (LNM), clinical stage, tumor infiltration, high histological grade (HG), serous subtype and PFI times. However, to date, the biological actions and clinical significances of AGAP2-AS1 have not been systematically reviewed in human cancers. In the present review, the authors overviewed the biological actions, potential mechanisms and clinical features of AGAP2-AS1 according to the previous studies. In summary, AGAP2-AS1, as a vital oncogenic gene, is a promising biomarker and potential target for carcinoma prognosis and therapy.
    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
    弥漫性中线神经胶质瘤(DMG)是中枢神经系统的侵袭性且致命的儿科肿瘤,对治疗具有高度抵抗力。组蛋白H3(H3-K27M)上残基27的赖氨酸替换为甲硫氨酸是DMG中的驱动突变,重塑这些细胞的表观遗传景观以促进肿瘤发生。H3-K27M胶质瘤的特征是组蛋白乙酰化和甲基化途径的失调,以及致癌MYC途径。为了寻找有效的治疗方法,我们研究了组蛋白脱乙酰酶(HDACs)和MYC双重靶向治疗这些肿瘤的潜力.用Sulfopin治疗H3-K27M患者来源的细胞,一种显示在体内阻断MYC驱动的肿瘤的抑制剂,与HDAC抑制剂伏立诺他联合使用,导致细胞活力大幅下降。此外,转录组和表观基因组谱分析揭示了这种药物组合在显著的致癌途径如mTOR的下调中的协同作用。最后,患者来源的原位异种移植模型的体内研究显示,在用该药物组合治疗的小鼠中,肿瘤生长显著减少.这些结果突出了PIN1和HDAC抑制剂的联合治疗作为这些侵袭性肿瘤的有希望的治疗方法。
    弥漫性中线神经胶质瘤(DMGs)是儿童中最具侵袭性和致命性的脑癌。它们通常与组蛋白的变化有关,控制基因活性并赋予染色体结构的蛋白质。大多数患有DMGs的儿童,例如,它们的组蛋白H3蛋白具有相同的异常(称为H3-K27M突变)。这种变化影响了称为甲基和乙酰基的小化学标签如何添加到组蛋白3上,这反过来改变了蛋白质打开和关闭基因的方式。因此,肿瘤开始发展。一种针对DMGs的潜在治疗策略是使用组蛋白去乙酰化酶抑制剂(HDACi),一种有前途的药物,可以抑制从组蛋白中去除乙酰基的酶。患者可以对HDACi产生耐药性,然而,强调需要探索其他方法。一种可能性是用几种药物治疗患者,每个通常针对一个独特的生物过程,有助于癌症的出现。这种联合方法可以有多种好处;药物可能会放大彼此的作用,例如,并且细胞在当时对一种以上的化合物产生抗性的可能性也较小。此外,联合用药中的每种药物都可以较低的剂量使用,以减少副作用并使患者受益。DMG肿瘤细胞通常具有较高的称为MYC的蛋白质活性水平,这可能有助于肿瘤的生长。Algranati,Oren等人。因此,开始测试将HDACi称为Vorinostat与阻断MYC活性的药物(Sulfopin)联合使用是否可以作为这种癌症的有效治疗方法。8名DMG患者的肿瘤样本接受了单独的Sulfopin治疗,或与伏立诺他联合使用的磺福平。暴露于两种药物的细胞不太可能存活,和额外的遗传实验表明,联合治疗导致促进肿瘤发展的途径被阻断。当将磺胺嘧啶和伏立诺他都施用于生长人类DMG肿瘤的小鼠时,动物表现出肿瘤生长的更大减少。DMG的治疗选择通常有限,化疗通常无效,手术不可能。Algranati的作品,Oren等人。提示将HDACi和靶向MYC途径的药物联合使用是一种策略,应进一步研究以确定是否可能临床应用.
    Diffuse midline gliomas (DMGs) are aggressive and fatal pediatric tumors of the central nervous system that are highly resistant to treatments. Lysine to methionine substitution of residue 27 on histone H3 (H3-K27M) is a driver mutation in DMGs, reshaping the epigenetic landscape of these cells to promote tumorigenesis. H3-K27M gliomas are characterized by deregulation of histone acetylation and methylation pathways, as well as the oncogenic MYC pathway. In search of effective treatment, we examined the therapeutic potential of dual targeting of histone deacetylases (HDACs) and MYC in these tumors. Treatment of H3-K27M patient-derived cells with Sulfopin, an inhibitor shown to block MYC-driven tumors in vivo, in combination with the HDAC inhibitor Vorinostat, resulted in substantial decrease in cell viability. Moreover, transcriptome and epigenome profiling revealed synergistic effect of this drug combination in downregulation of prominent oncogenic pathways such as mTOR. Finally, in vivo studies of patient-derived orthotopic xenograft models showed significant tumor growth reduction in mice treated with the drug combination. These results highlight the combined treatment with PIN1 and HDAC inhibitors as a promising therapeutic approach for these aggressive tumors.
    Diffuse midline gliomas (DMGs) are among the most aggressive and fatal brain cancers in children. They are often associated with changes in histones, the proteins that control gene activity and give chromosomes their structure. Most children with DMGs, for example, share the same anomaly in their histone H3 protein (referred to as the H3-K27M mutation). This change affects how small chemical tags called methyl and acetyl groups can be added onto histone 3, which in turn alters the way the protein can switch genes on and off. As a result, tumours start to develop. One potential therapeutic strategy against DMGs is to use histone deacetylase inhibitors (HDACi), a promising type of drugs which inhibits the enzymes that remove acetyl groups from histones. Patients can develop resistance to HDACi, however, highlighting the need to explore other approaches. One possibility is to treat patients with several types of drugs, each usually targeting a distinct biological process that contributes to the emergence of cancer. This combined approach can have multiple benefits; the drugs potentially amplify each other’s effect, for example, and it is also less likely for cells to become resistant to more than one compound at the time. In addition, each drug in the combination can be used in a lower dose to reduce side effects and benefit patients. DMG tumour cells often feature higher activity levels of a protein known as MYC, which can contribute to the growth of the tumour. Algranati, Oren et al. therefore set out to test whether combining an HDACi known as Vorinostat with a drug that blocks MYC activity (Sulfopin) can act as an effective treatment for this cancer. Tumour samples from eight DMG patients were treated with either Sulfopin alone, or Sulfopin in association with Vorinostat. Cells exposed to both drugs were less likely to survive, and additional genetic experiments showed that the combined treatment had resulted in pathways that promote tumour development being blocked. When both Sulfopin and Vorinostat were administered to mice made to grow human DMG tumors, the animals showed a greater reduction in tumor growth. Treatment options for DMG are usually limited, with chemotherapy often being ineffective and surgery impossible. The work by Algranati, Oren et al. suggests that combining HDACi and drugs targeting the MYC pathway is a strategy that should be examined further to determine whether clinical applications are possible.
    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
    microRNAs(miRNAs)是小的非编码RNA,在细胞增殖中起关键作用,迁移,通过调节基因表达进行侵袭和凋亡。在恶性肿瘤中,miRNA-122作为肿瘤抑制因子或癌基因,通过下游基因靶向影响肿瘤进展。然而,miRNA-122在癌症中的确切作用尚不清楚。miRNA-122是放疗和化疗的潜在生物标志物和调节剂。本综述旨在总结miRNA‑122在癌症中的作用,它作为诊断和预后的生物标志物的潜力及其在癌症治疗中的意义,包括放疗和化疗,以及系统交付的策略。
    MicroRNAs (miRNAs) are small non‑coding RNAs that serve key roles in cell proliferation, migration, invasion and apoptosis by regulating gene expression. In malignant tumors, miRNA‑122 serves either as a tumor suppressor or oncogene, influencing tumor progression via downstream gene targeting. However, the precise role of miRNA‑122 in cancer remains unclear. miRNA‑122 is a potential biomarker and modulator of radiotherapy and chemotherapy. The present review aimed to summarize the roles of miRNA‑122 in cancer, its potential as a biomarker for diagnosis and prognosis and its implications in cancer therapy, including radiotherapy and chemotherapy, alongside strategies for systemic delivery.
    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
    多项研究表明,铁螯合剂通过诱导NDRG1(一种已知的肿瘤和转移抑制因子)来增强其抗癌特性。然而,NDRG1的确切作用仍然存在争议,新的研究表明,NDRG1也可以作为癌基因。我们小组最近引入了线粒体靶向铁螯合剂去铁胺(mitoDFO)和地拉罗司(mitoDFX)作为有效的抗癌剂。在这项研究中,我们评估了这些修饰的螯合剂诱导NDRG1的能力以及NDRG1在乳腺癌中的作用.我们证明了两种化合物均特异性增加NDRG1而不诱导其他NDRG家族成员。我们已经证明,线粒体靶向螯合剂的作用至少部分由GSK3α/β介导,导致NDRG1在Thr346磷酸化,而在Ser330上则较小。NDRG1的缺失增加了mitoDFX诱导的细胞死亡。值得注意的是,缺乏NDRG1的MDA-MB-231细胞表现出降低的细胞外酸化速率,并且生长速度比亲本细胞慢。而ER+MCF7细胞则相反。此外,全长NDRG1和N末端截短的同种型(59112)的过表达显着降低了ER细胞对mitoDFX的敏感性。此外,过表达全长NDRG1的细胞表现出明显加速的肿瘤形成,而其N末端截短的同工型显示出显着受损的形成肿瘤的能力。因此,在高度侵袭性的三阴性乳腺癌中,全长NDRG1的过表达促进肿瘤生长.
    Multiple studies indicate that iron chelators enhance their anti-cancer properties by inducing NDRG1, a known tumor and metastasis suppressor. However, the exact role of NDRG1 remains controversial, as newer studies have shown that NDRG1 can also act as an oncogene. Our group recently introduced mitochondrially targeted iron chelators deferoxamine (mitoDFO) and deferasirox (mitoDFX) as effective anti-cancer agents. In this study, we evaluated the ability of these modified chelators to induce NDRG1 and the role of NDRG1 in breast cancer. We demonstrated that both compounds specifically increase NDRG1 without inducing other NDRG family members. We have documented that the effect of mitochondrially targeted chelators is at least partially mediated by GSK3α/β, leading to phosphorylation of NDRG1 at Thr346 and to a lesser extent on Ser330. Loss of NDRG1 increases cell death induced by mitoDFX. Notably, MDA-MB-231 cells lacking NDRG1 exhibit reduced extracellular acidification rate and grow slower than parental cells, while the opposite is true for ER+ MCF7 cells. Moreover, overexpression of full-length NDRG1 and the N-terminally truncated isoform (59112) significantly reduced sensitivity towards mitoDFX in ER+ cells. Furthermore, cells overexpressing full-length NDRG1 exhibited a significantly accelerated tumor formation, while its N-terminally truncated isoforms showed significantly impaired capacity to form tumors. Thus, overexpression of full-length NDRG1 promotes tumor growth in highly aggressive triple-negative breast 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
    成员视黄酸受体(RAR)(α,β,和γ)和类维生素AX受体(RXRs)(α,β,和γ)属于类维生素A受体家族。它们通过核类维生素A受体调节经典类维生素A的生物学作用,由配体调节的转录因子。通过位于靶基因启动子中的特定视黄酸响应元件(RARE)的结合,RAR和RXR的成员形成异二聚体。通过与其核受体结合并触发下游靶基因的转录,维甲酸(RA)介导某些基因的表达。因此类维生素A主要控制基因表达以实现其生物学作用。RAR对许多生物过程至关重要,如发展,豁免权,繁殖,器官发生,和稳态。除了他们的生理功能,由于突变,RAR也与病理和肿瘤有关,蛋白质融合,表达水平的变化,或导致功能异常和稳态破坏的异常翻译后变化。动物组织或培养细胞的致癌发育与类视黄醇受体的表达改变有关。RAR-α在几种恶性肿瘤中过度表达。几种癌症形式的侵袭和迁移增加,包括HNSC癌,小儿低度胶质瘤,肺腺癌,和乳腺癌,已经与其上调的表达有关。已经开发了许多批准的靶向RAR-α的治疗方案,提高患者生存率。
    本研究的主要目的是鉴定新型RAR-α靶向药物并评估RAR-α在乳腺癌患者中的表达模式。
    使用UALCAN等多种生物信息学工具进行计算机内调查,TISCH,TIMER2.0,ENRICHR,和其他人被用来检查RAR-α的表达。进一步,我们评估了三法罗汀对RAR-α的计算机内抑制作用,并测试了三法罗汀在乳腺癌细胞中的细胞毒性。
    我们的研究表明RAR-α在包括乳腺癌在内的多种恶性肿瘤中上调。它调节粒细胞分化,并与视黄酸受体信号通路和细胞对雌激素刺激的反应有关。此外,通过计算机和体外研究,发现trifarotene是一种潜在的针对RAR-α的合成化合物。
    总的来说,这项研究表明RAR-α的高表达增强了乳腺癌的发病。使用trifarotene药物靶向RAR-α将显着增强乳腺癌个体对治疗的反应并延迟对药物的耐药性发展。
    UNASSIGNED: The members retinoic acid receptors (RARs) (α, β, and γ) and retinoid X receptors (RXRs) (α, β, and γ) belong to the retinoid receptor family. They regulate the biological action of classical retinoids through nuclear retinoid receptors, a transcription factor that is regulated by ligands. Through the binding of particular retinoic acid-responsive elements (RAREs) located in target gene promoters, RARs and members of the RXRs form heterodimers. By binding to its nuclear receptors and triggering the transcription of the target genes downstream, retinoic acid (RA) mediates the expression of certain genes. Retinoids so mainly control gene expression to carry out their biological actions. RARs are essential for many biological processes, such as development, immunity, reproduction, organogenesis, and homeostasis. Apart from their physiological functions, RARs are also linked to pathologies and tumors due to mutations, protein fusions, changes in expression levels, or abnormal post-translational changes that lead to aberrant functions and homeostasis breakdown. The oncogenic development of animal tissues or cultured cells is linked to altered expression of retinoid receptors. The RAR-α is over-expressed in several malignancies. Increased invasion and migration in several cancer forms, including HNSC carcinoma, pediatric low-grade gliomas, lung adenocarcinoma, and breast cancer, have been linked to its upregulated expression. Numerous approved therapeutic regimens targeting RAR-α have been developed, improving patient survival rates.
    UNASSIGNED: This study\'s main objective was to identify novel RAR-α-targeting drugs and evaluate the expression patterns of RAR-α in breast cancer patients.
    UNASSIGNED: In-silico investigation using a variety of bioinformatics tools like UALCAN, TISCH, TIMER 2.0, ENRICHR, and others were employed to examine the expression of RAR-α. Further we evaluated in-silico inhibition of RAR-α with trifarotene and also tested the cytotoxicity of trifarotene in breast cancer cells.
    UNASSIGNED: Our research indicates that RAR-α is upregulated in several malignancies including Breast Cancer. It regulates granulocyte differentiation and has an association with the retinoic acid receptor signaling pathway and cellular response to estrogen stimulus. Furthermore, trifarotene was found as a potential synthetic compound that targets RAR-α through in silico and in-vitro study.
    UNASSIGNED: Overall, this research indicates that elevated expression of RAR-α enhances the onset of breast cancer. Using trifarotene medication to target RAR-α will significantly boost the response of breast cancer individuals to treatment and delay the development of resistance to drugs.
    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
    SET域包含7(SETD7),组蛋白甲基转移酶的成员,在多种肿瘤类型中异常表达。然而,SETD7在肾透明细胞癌(ccRCC)中的生物学功能和潜在的分子机制尚不清楚。这里,我们探讨了SETD7-TAF7-CCNA2轴对ccRCC增殖和转移的生物学效应。我们确定SETD7和TAF7均上调并显着促进ccRCC细胞的增殖和迁移。同时,SETD7和TAF7的表达呈显著正相关,二者在细胞核内共定位。机械上,SETD7在K5和K300位点甲基化TAF7,导致TAF7的去泛素化和稳定。此外,再表达TAF7可以部分恢复SETD7敲低抑制ccRCC细胞的增殖和迁移。此外,TAF7转录激活以驱动细胞周期蛋白A2(CCNA2)的表达。更重要的是,TAF7在K5和K300位点的甲基化表现出更高的CCNA2转录活性,促进ccRCC的形成和进展。我们的发现揭示了SETD7介导的TAF7甲基化在ccRCC进展中调节CCNA2转录激活的独特机制,并通过靶向SETD7-TAF7-CCNA2轴的成员为开发有效的治疗策略提供了基础。
    SET domain containing 7(SETD7), a member of histone methyltransferases, is abnormally expressed in multiple tumor types. However, the biological function and underlying molecular mechanism of SETD7 in clear cell renal cell carcinoma (ccRCC) remain unclear. Here, we explored the biological effects of SETD7-TAF7-CCNA2 axis on proliferation and metastasis in ccRCC. We identified both SETD7 and TAF7 were up-regulated and significantly promoted the proliferation and migration of ccRCC cells. Concurrently, there was a significant positive correlation between the expression of SETD7 and TAF7, and the two were colocalized in the nucleus. Mechanistically, SETD7 methylates TAF7 at K5 and K300 sites, resulting in the deubiquitination and stabilization of TAF7. Furthermore, re-expression of TAF7 could partially restore SETD7 knockdown inhibited ccRCC cells proliferation and migration. In addition, TAF7 transcriptionally activated to drive the expression of cyclin A2 (CCNA2). And more importantly, the methylation of TAF7 at K5 and K300 sites exhibited higher transcriptional activity of CCNA2, which promotes formation and progression of ccRCC. Our findings reveal a unique mechanism that SETD7 mediated TAF7 methylation in regulating transcriptional activation of CCNA2 in ccRCC progression and provide a basis for developing effective therapeutic strategies by targeting members of SETD7-TAF7-CCNA2 axis.
    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
    通过DNA扩增或过表达激活癌基因是癌症发生和进展的关键驱动因素。FOXK2基因,位于染色体17q25上,编码具有叉头DNA结合域的转录因子。基因组数据集的分析表明,FOXK2在乳腺癌中经常被扩增和过表达。与患者生存率低有关。敲除FOXK2显著抑制乳腺癌细胞增殖,迁移,锚定独立生长,和在异种移植小鼠模型中延迟肿瘤生长。此外,抑制FOXK2使乳腺癌细胞对化疗敏感。共过表达FOXK2和突变型PI3KCA转化的非肿瘤性MCF-10A细胞,提示FOXK2在PI3KCA驱动的肿瘤发生中的作用。CCNE2、PDK1和ESR1被鉴定为MCF-7细胞中FOXK2的转录靶标。CCNE2/CDK2(dinaciclib)和PDK1(二氯乙酸)的小分子抑制剂与PI3KCA抑制剂(alpelisib)在体外表现出协同抗肿瘤作用。dinaciclib对FOXK2的抑制协同增强了alpelisib在异种移植小鼠模型中的抗肿瘤作用。总的来说,这些发现突出了FOXK2的致癌功能,并提示FOXK2及其下游基因代表了乳腺癌的潜在治疗靶点.
    Oncogene activation through DNA amplification or overexpression is a crucial driver of cancer initiation and progression. The FOXK2 gene, located on chromosome 17q25, encodes a transcription factor with a forkhead DNA-binding domain. Analysis of genomic datasets reveals that FOXK2 is frequently amplified and overexpressed in breast cancer, correlating with poor patient survival. Knockdown of FOXK2 significantly inhibited breast cancer cell proliferation, migration, anchorage-independent growth, and delayed tumor growth in a xenograft mouse model. Additionally, inhibiting FOXK2 sensitized breast cancer cells to chemotherapy. Co-overexpression of FOXK2 and mutant PI3KCA transformed non-tumorigenic MCF-10A cells, suggesting a role for FOXK2 in PI3KCA-driven tumorigenesis. CCNE2, PDK1, and ESR1 were identified as transcriptional targets of FOXK2 in MCF-7 cells. Small-molecule inhibitors of CCNE2/CDK2 (dinaciclib) and PDK1 (dichloroacetate) exhibited synergistic anti-tumor effects with PI3KCA inhibitor (alpelisib) in vitro. Inhibition of FOXK2 by dinaciclib synergistically enhanced the anti-tumor effects of alpelisib in a xenograft mouse model. Collectively, these findings highlight the oncogenic function of FOXK2 and suggest that FOXK2 and its downstream genes represent potential therapeutic targets in breast 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
    简介:肌动蛋白样6A(ACTL6A)表达增加与各种癌症有关,但缺乏对不同恶性肿瘤的全面调查。我们旨在利用生物信息学工具分析ACTL6A作为潜在的癌基因和治疗靶标。方法:我们全面分析了人类恶性肿瘤的ACTL6A表达谱,关注与肿瘤分级的相关性,舞台,转移,和病人的生存。检查了遗传改变,并使用严格的方法评估ACTL6A的表观遗传景观。评估了ACTL6A对肿瘤微环境中免疫细胞浸润的影响,以及分子对接研究和机器学习模型。结果:我们的分析显示ACTL6A在各种肿瘤中表达升高,与不良预后指标相关,如肿瘤分级,舞台,转移,和病人的生存。鉴定了基因突变和表观遗传修饰,以及与免疫细胞浸润和关键细胞途径的关联。机器学习模型展示了ACTL6A用于癌症检测的潜力。讨论:ACTL6A成为癌症的一个有前途的诊断和治疗靶点,对预后和治疗有影响。我们的研究提供了对其致癌作用的全面见解,强调其作为预后指标和抗癌治疗靶点的潜力。这种综合方法增强了我们对ACTL6A在癌症发病机制和治疗中的作用的理解。
    Introduction: Increased Actin-like 6A (ACTL6A) expression is associated with various cancers, but its comprehensive investigation across different malignancies is lacking. We aimed to analyze ACTL6A as a potential oncogene and therapeutic target using bioinformatics tools. Methods: We comprehensively analyzed ACTL6A expression profiles across human malignancies, focusing on correlations with tumor grade, stage, metastasis, and patient survival. Genetic alterations were examined, and the epigenetic landscape of ACTL6A was assessed using rigorous methods. The impact of ACTL6A on immune cell infiltration in the tumor microenvironment was evaluated, along with molecular docking studies and machine learning models. Results: Our analysis revealed elevated ACTL6A expression in various tumors, correlating with poor prognostic indicators such as tumor grade, stage, metastasis, and patient survival. Genetic mutations and epigenetic modifications were identified, along with associations with immune cell infiltration and key cellular pathways. Machine learning models demonstrated ACTL6A\'s potential for cancer detection. Discussion: ACTL6A emerges as a promising diagnostic and therapeutic target in cancer, with implications for prognosis and therapy. Our study provides comprehensive insights into its carcinogenic actions, highlighting its potential as both a prognostic indicator and a target for anti-cancer therapy. This integrative approach enhances our understanding of ACTL6A\'s role in cancer pathogenesis and treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号