oncogene

癌基因
  • 文章类型: Journal Article
    乳腺癌(BC)是影响全世界妇女的主要公共卫生问题。越来越多的证据强调了miRNA-206在BC发病机制中的作用。其表达的变化具有诊断和预后潜力,因为它们与临床病理参数相关,包括淋巴结转移,总生存率,肿瘤大小,转移阶段,化疗耐药,和复发。在本研究中,我们总结了,评估,并讨论了对miRNA-206在BC中的功能的最新理解。出乎意料的是,miRNA-206被发现控制致癌和肿瘤抑制途径。我们还考虑了相应的下游影响和上游监管机构。最后,我们讨论了miRNA-206的诊断和预后价值及其在开发新治疗策略方面的潜力.
    Breast cancer (BC) is a major public health problem that affects women worldwide. Growing evidence has highlighted the role of miRNA-206 in BC pathogenesis. Changes in its expression have diagnostic and prognostic potential as they are associated with clinicopathological parameters, including lymph node metastasis, overall survival, tumor size, metastatic stage, resistance to chemotherapy, and recurrence. In the present study, we summarized, assessed, and discussed the most recent understanding of the functions of miRNA-206 in BC. Unexpectedly, miRNA-206 was found to control both oncogenic and tumor-suppressive pathways. We also considered corresponding downstream effects and upstream regulators. Finally, we addressed the diagnostic and prognostic value of miRNA-206 and its potential for the development of new therapeutic strategies.
    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
    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

    求助全文

  • 文章类型: Journal Article
    RAS蛋白调节细胞分裂,通过多个下游效应子途径分化和凋亡。致癌RAS变异是癌症中最常见的驱动因素,然而,它们也驱动许多易患恶性肿瘤的良性病变,比如黑素细胞痣,甲状腺结节,结肠息肉.这些良性病变的逆转可以降低癌症发病率,然而,众所周知,下游途径抑制剂很难靶向致癌RAS的作用.在这里,我们显示了使用靶向复发性因果变异的siRNA在黑素细胞痣的原代细胞中有效抑制致癌和目前不可药物的NRASQ61K。这导致ARL6IP1的表达显着降低,ARL6IP1是一种已知的内质网应激诱导的凋亡抑制剂,以前与NRAS无关。我们继续证明,在原代细胞中单个剂量的siRNA会触发凋亡级联反应,与用MEK抑制剂治疗相反。将靶向siRNA保护性包装到脂质纳米颗粒中允许成功递送到黑素细胞痣的人源化小鼠模型中。并导致体内变异的NRAS敲低。这些数据表明,RAS诱导的细胞凋亡保护与NRAS驱动的黑素细胞痣的持久性有关,并预期靶向siRNA可以构成RAS驱动的良性肿瘤的临床试验基础。
    RAS proteins regulate cell division, differentiation and apoptosis via multiple downstream effector pathways. Oncogenic RAS variants are the commonest drivers in cancers, however they also drive many benign lesions predisposing to malignancy, such as melanocytic naevi, thyroid nodules, and colonic polyps. Reversal of these benign lesions could reduce cancer incidence, however the effects of oncogenic RAS have been notoriously difficult to target with downstream pathway inhibitors. Here we show effective suppression of oncogenic and currently undruggable NRASQ61K in primary cells from melanocytic naevi using siRNA targeted to the recurrent causal variant. This results in striking reduction in expression of ARL6IP1, a known inhibitor of endoplasmic reticulum stress-induced apoptosis not previously linked to NRAS. We go on to show that a single dose of siRNA in primary cells triggers an apoptotic cascade, in contrast to treatment with a MEK inhibitor. Protective packaging of the targeted siRNA into lipid nanoparticles permits successful delivery into a humanised mouse model of melanocytic naevi, and results in variant NRAS knockdown in vivo. These data show that RAS-induced protection from apoptosis is involved in persistence of NRAS-driven melanocytic naevi and anticipate that targeted siRNA could form the basis of clinical trials for RAS-driven benign tumours.
    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: Case Reports
    暂无摘要。
    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
    肺癌是世界范围内最普遍的人类癌症之一,具有很高的致死率。在这项研究中,我们证明GSE1(遗传抑制因子1)表达在肺腺癌中异常上调,GSE1耗竭抑制A549和H1299细胞的增殖和迁移.免疫沉淀实验表明,GSE1与细胞中的组蛋白脱乙酰酶1(HDAC1)和其他BRAF-HDAC复合物(BHC)成分相互作用。GSE1敲低A549细胞的转录组表明,基于p值<.05和倍数变化≥1.5,207个基因上调,159个基因下调。生物信息学分析表明,140个差异表达基因含有HDAC1的结合位点,包括抑癌基因KLF6(Kruppel样因子6)。的确,定量逆转录聚合酶链反应和蛋白质印迹分析显示GSE1可抑制肺癌细胞中KLF6的转录。总之,GSE1与HDAC1协同作用,通过下调KLF6的表达促进非小细胞肺癌细胞的增殖和转移。
    Lung cancer is one of the most prevalent human cancers with a high lethality rate worldwide. In this study, we demonstrated that GSE1 (genetic suppressor element 1) expression is aberrantly upregulated in lung adenocarcinoma and that GSE1 depletion inhibits the proliferation and migration of both A549 and H1299 cells. Immunoprecipitation assays demonstrated that GSE1 interacts with histone deacetylase 1 (HDAC1) and other BRAF-HDAC complex (BHC) components in cells. The transcriptome of GSE1-knockdown A549 cells indicated that 207 genes were upregulated and 159 were downregulated based on a p-value < .05 and fold change ≥ 1.5. Bioinformatics analysis suggested that 140 differentially expressed genes harbor binding sites for HDAC1, including the tumor suppressor gene KLF6 (Kruppel-like factor 6). Indeed, quantitative reverse-transcription polymerase chain reaction and western blot analysis revealed that GSE1 could inhibit the transcription of KLF6 in lung cancer cells. In conclusion, GSE1 cooperates with HDAC1 to promote the proliferation and metastasis of non-small cell lung cancer cells through the downregulation of KLF6 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
    ErbB3结合蛋白1(Ebp1)有两个亚型,p42Ebp1和p48Ebp1均能调控细胞的生长和分化。但是这些同工型通常有相反的作用,包括在不同组织和细胞中调节细胞生长的矛盾作用。P48Ebp1属于全长序列,而p42Ebp1晶体结构的构象变化表明在氨基末端缺乏α螺旋。由于这两种同工型结构的差异,它们具有不同的结合伴侣和蛋白质修饰。Ebp1既可以作为癌基因又可以作为肿瘤抑制因子。然而,这两种同工型发挥相反功能的潜在机制仍未完全理解。在这次审查中,我们总结了这两种亚型的基因和蛋白质结构,蛋白质修饰,结合伴侣和不同同工型与疾病的关联。
    ErbB3-binding protein 1(Ebp1) has two isoforms, p42 Ebp1 and p48 Ebp1, both of which can regulate cell growth and differentiation. But these isoforms often have opposite effects, including contradictory roles in regulation of cell growth in different tissues and cells. P48 Ebp1 belongs to the full-length sequence, while conformational changes in the crystal structure of p42 Ebp1 reveals a lack of an α helix at the amino terminus. Due to the differences in the structures of these two isoforms, they have different binding partners and protein modifications. Ebp1 can function as both an oncogene and a tumor suppressor factor. However, the underlying mechanisms by which these two isoforms exert opposite functions are still not fully understood. In this review, we summarize the genes and the structures of protein of these two isoforms, protein modifications, binding partners and the association of different isoforms with diseases.
    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
    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