tumor suppressor

肿瘤抑制物
  • 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
    KRIT1是一种75kDa支架蛋白,通过限制对炎性刺激的反应并维持静止和稳定的内皮屏障来调节内皮细胞表型。KRIT1功能突变的缺失导致脑海绵状畸形(CCM)的发展,一种以异常血管形成为特征的疾病,表现出屏障功能的丧失,内皮增殖增加,和改变基因表达。虽然我们在理解KRIT1以及功能相关蛋白CCM2和PDCD10如何促进血管和血管屏障的调节方面取得了许多进展,一些重要的悬而未决的问题仍然存在。此外,KRIT1广泛表达,KRIT1和其他CCM蛋白已被证明在非内皮细胞类型和组织中起重要作用,这可能与他们作为CCM致病源的作用有关,也可能与他们的作用无关。在这次审查中,我们讨论了关于KRIT1在血管生理学中的作用的一些未解决的问题,并讨论了最近的进展,这些进展表明,这种广泛表达的蛋白可能具有超出内皮细胞的作用.
    KRIT1 is a 75 kDa scaffolding protein which regulates endothelial cell phenotype by limiting the response to inflammatory stimuli and maintaining a quiescent and stable endothelial barrier. Loss of function mutations in KRIT1 lead to the development of cerebral cavernous malformations (CCM), a disease marked by the formation of abnormal blood vessels which exhibit a loss of barrier function, increased endothelial proliferation, and altered gene expression. While many advances have been made in our understanding of how KRIT1, and the functionally related proteins CCM2 and PDCD10, contribute to the regulation of blood vessels and the vascular barrier, some important open questions remain. In addition, KRIT1 is widely expressed and KRIT1 and the other CCM proteins have been shown to play important roles in non-endothelial cell types and tissues, which may or may not be related to their role as pathogenic originators of CCM. In this review, we discuss some of the unsettled questions regarding the role of KRIT1 in vascular physiology and discuss recent advances that suggest this ubiquitously expressed protein may have a role beyond the endothelial cell.
    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
    乙型肝炎感染与全球肝癌的发展密切相关,肝细胞癌(HCC)的患病率超过50%。乙型肝炎病毒(HBV)编码乙型肝炎病毒X(HBx)蛋白,HBV共价闭合环状DNA(cccDNA)微染色体转录所必需的多效性调节蛋白。在以往的研究中,HBV相关的HCC被发现在多个信号通路中受到HBx的影响,导致原癌基因和抑癌基因的基因突变和表观遗传修饰。此外,转化生长因子-β(TGF-β)在恶性肿瘤的各个阶段具有二分潜能,因为它是调节多种细胞和生理过程的关键信号通路。在早期肝癌,TGF-β具有显著的抗肿瘤作用,而在晚期肝癌中,它促进恶性进展。TGF-β与肝癌中的HBx蛋白相互作用,调节肝癌的发病机制。本文综述了HBx和TGB-β在HCC发生发展中的各自和联合作用。
    Hepatitis B infection is substantially associated with the development of liver cancer globally, with the prevalence of hepatocellular carcinoma (HCC) cases exceeding 50%. Hepatitis B virus (HBV) encodes the Hepatitis B virus X (HBx) protein, a pleiotropic regulatory protein necessary for the transcription of the HBV covalently closed circular DNA (cccDNA) microchromosome. In previous studies, HBV-associated HCC was revealed to be affected by HBx in multiple signaling pathways, resulting in genetic mutations and epigenetic modifications in proto-oncogenes and tumor suppressor genes. In addition, transforming growth factor-β (TGF-β) has dichotomous potentials at various phases of malignancy as it is a crucial signaling pathway that regulates multiple cellular and physiological processes. In early HCC, TGF-β has a significant antitumor effect, whereas in advanced HCC, it promotes malignant progression. TGF-β interacts with the HBx protein in HCC, regulating the pathogenesis of HCC. This review summarizes the respective and combined functions of HBx and TGB-β in HCC occurrence and 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
    目的:骨肉瘤的甲氨蝶呤(MTX)耐药导致预后极差。在本研究中,为了进一步了解MTX耐药在骨肉瘤中的作用,我们选择了MTXIC50增加5,500倍的骨肉瘤细胞系材料和方法:通过用逐步增加的MTX量连续培养,从MTX敏感的亲本人143B骨肉瘤细胞(143B-P)中选择超级MTX抗性的143B骨肉瘤细胞(143B-MTXSR)。为了比较143B-MTXSR和143B-P的恶性程度,将集落形成能力与塑料和软琼脂上的克隆形成测定进行了比较。此外,将肿瘤生长与骨肉瘤的原位异种移植小鼠模型进行比较。二氢叶酸还原酶(DHFR)的表达,磷酸肌醇3-激酶(PI3K)/蛋白激酶B(AKT)/哺乳动物雷帕霉素靶蛋白(mTOR),用Western免疫印迹法检测骨髓细胞瘤癌基因(MYC),并在143B-MTXSR和143B-P细胞中进行比较。
    结果:与亲本143B-P细胞相比,143B-MTXSR的MTXIC50增加了5,500倍。与143B-P相比,DHFR在143B-MTXSR中的表达增加10倍(p<0.01)。与143B-P相比,143B-MTXSR细胞在塑料(p=0.032)和软琼脂(p<0.01)上的集落形成能力降低,并且在原位异种移植小鼠模型中肿瘤生长降低(p<0.001)。这些结果表明143B-MTXSR降低了恶性肿瘤。143B-MTXSR也显示PI3K的表达增加(p<0.01),磷酸化(活化)AKT(p=0.031),磷酸化mTOR(p=0.043),与143B-P相比,c-MYC(p=0.024)。
    结论:本研究表明DHFR的表达增加,PI3K/AKT/mTOR和c-MYC似乎与超级MTX抗性有关,矛盾的是,减少恶性肿瘤。目前的结果表明,DHFR在高度扩增时可能是一种强大的肿瘤抑制因子。
    OBJECTIVE: Methotrexate (MTX) resistance in osteosarcoma leads to a very poor prognosis. In the present study, in order to further understand the basis and ramifications of MTX resistance in osteosarcoma, we selected an osteosarcoma cell line that has a 5,500-fold-increased MTX IC50 Materials and Methods: The super MTX-resistant 143B osteosarcoma cells (143B-MTXSR) were selected from MTX-sensitive parental human 143B osteosarcoma cells (143B-P) by continuous culture with step-wise increased amounts of MTX. To compare the malignancy of 143B-MTXSR and 143B-P, colony-formation capacity was compared with clonogenic assays on plastic and in soft agar. In addition, tumor growth was compared with orthotopic xenograft mouse models of osteosarcoma. Expression of dihydrofolate reductase (DHFR), phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR), and myelocytomatosis oncogene (MYC) was examined with western immunoblotting and compared in 143B-MTXSR and 143B-P cells.
    RESULTS: 143B-MTXSR had a 5,500-fold increase in the MTX IC50 compared to the parental 143B-P cells. Expression of DHFR was increased 10-fold in 143B-MTXSR compared to 143B-P (p<0.01). 143B-MTXSR cells had reduced colony-formation capacity on plastic (p=0.032) and in soft agar (p<0.01) compared to 143B-P and reduced tumor growth in orthotopic xenograft mouse models (p<0.001). These results demonstrate that 143B-MTXSR had reduced malignancy. 143B-MTXSR also showed an increased expression of PI3K (p<0.01), phosphorylated (activated) AKT (p=0.031), phosphorylated mTOR (p=0.043), and c-MYC (p=0.024) compared to 143B-P.
    CONCLUSIONS: The present study demonstrates that the increased expression of DHFR, PI3K/AKT/mTOR and c-MYC appears to be linked to super MTX resistance and, paradoxically, to reduced malignancy. The present results suggest that DHFR may be a powerful tumor suppressor when highly amplified.
    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
    驻留细胞和细胞外基质(ECM)之间的复杂相互作用深刻地影响癌症进展。在三阴性乳腺癌(TNBC)中,ECM结构的演变是由于赖氨酰氧化酶的富集,纤连蛋白,和胶原蛋白,促进远处转移。在这里,我们揭示了涉及表观遗传调节因子UBR7和组蛋白甲基转移酶EZH2在调节转化生长因子(TGF)-β/Smad信号传导中的关键转录调节机制,影响ECM基因的表达。UBR7损失导致兼性异染色质标记H3K27me3的急剧减少,从而激活ECM基因。UBR7在粘附的癌细胞和球体中的基质沉积中起着至关重要的作用,改变胶原蛋白含量和赖氨酰氧化酶活性,直接影响基体刚度和侵入性。这些发现在小鼠模型和TNBC患者体内得到进一步验证,其中降低的UBR7水平伴随着增加的ECM成分表达和活性,导致纤维化介导的基质硬度。因此,UBR7是基体硬化的主调节器,影响TNBC的转移潜力。
    The intricate interplay between resident cells and the extracellular matrix (ECM) profoundly influences cancer progression. In triple-negative breast cancer (TNBC), ECM architecture evolves due to the enrichment of lysyl oxidase, fibronectin, and collagen, promoting distant metastasis. Here we uncover a pivotal transcription regulatory mechanism involving the epigenetic regulator UBR7 and histone methyltransferase EZH2 in regulating transforming growth factor (TGF)-β/Smad signaling, affecting the expression of ECM genes. UBR7 loss leads to a dramatic reduction in facultative heterochromatin mark H3K27me3, activating ECM genes. UBR7 plays a crucial role in matrix deposition in adherent cancer cells and spheroids, altering collagen content and lysyl oxidase activity, directly affecting matrix stiffness and invasiveness. These findings are further validated in vivo in mice models and TNBC patients, where reduced UBR7 levels are accompanied by increased ECM component expression and activity, leading to fibrosis-mediated matrix stiffness. Thus, UBR7 is a master regulator of matrix stiffening, influencing the metastatic potential of TNBC.
    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
    FBXW7是特征最明确的F-box蛋白之一,充当SKP1-CUL1-F-box(SCF)E3连接酶复合物的底物受体亚基。SCFFBXW7负责降解各种致癌蛋白,如细胞周期蛋白E,c-MYC,c-JUN,NOTCH,和MCL1。因此,FBXW7主要作为主要的肿瘤抑制因子发挥作用。按照这个概念,FBXW7基因突变或下调已在许多类型的恶性肿瘤中发现并报道,如子宫内膜,结直肠,肺,和乳腺癌,这促进了扩散,入侵,迁移,和癌细胞的耐药性。因此,回顾新发现的FBXW7在生理和病理条件下的调节和肿瘤抑制功能,以开发治疗FBXW7改变的癌症的有效策略至关重要。由于越来越多的证据揭示了FBXW7的肿瘤抑制活性和作用,我们更新了FBXW7上游和下游信号,包括FBXW7泛素底物,FBXW7的多层次监管机制,和癌症中FBXW7的失调,并讨论了针对FBXW7调节因子和下游效应因子的有希望的癌症疗法,提供FBXW7的全面情况,促进该领域的研究。
    FBXW7 is one of the most well-characterized F-box proteins, serving as substrate receptor subunit of SKP1-CUL1-F-box (SCF) E3 ligase complexes. SCFFBXW7 is responsible for the degradation of various oncogenic proteins such as cyclin E, c-MYC, c-JUN, NOTCH, and MCL1. Therefore, FBXW7 functions largely as a major tumor suppressor. In keeping with this notion, FBXW7 gene mutations or downregulations have been found and reported in many types of malignant tumors, such as endometrial, colorectal, lung, and breast cancers, which facilitate the proliferation, invasion, migration, and drug resistance of cancer cells. Therefore, it is critical to review newly identified FBXW7 regulation and tumor suppressor function under physiological and pathological conditions to develop effective strategies for the treatment of FBXW7-altered cancers. Since a growing body of evidence has revealed the tumor-suppressive activity and role of FBXW7, here, we updated FBXW7 upstream and downstream signaling including FBXW7 ubiquitin substrates, the multi-level FBXW7 regulatory mechanisms, and dysregulation of FBXW7 in cancer, and discussed promising cancer therapies targeting FBXW7 regulators and downstream effectors, to provide a comprehensive picture of FBXW7 and facilitate the study in this field.
    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: Retraction of Publication
    在本文发表之后,有关读者提请编辑注意,图中某些Transwell细胞迁移和入侵测定数据。3C和D,和肿瘤图像如图所示。图4A与不同研究机构的不同作者撰写的其他文章中以不同形式出现的数据惊人地相似,已经出版了。此外,图中所示的某些数据面板。3C是重叠的,这样,来自相同原始来源的数据被选择来代表据称进行不同实验的结果。由于上述文章中的有争议的数据在提交给分子医学报告之前已经发表,编辑已经决定这篇论文应该从期刊上撤回。作者被要求解释这些担忧,但编辑部没有收到回复。编辑对读者造成的不便表示歉意。[分子医学报告15:4217-4224,2017;DOI:10.3892/mmr.2017.6493]。
    Following the publication of this paper, it was drawn to the Editor\'s attention by a concerned reader that certain of the Transwell cell migration and invasion assay data in Fig. 3C and D, and the tumour images shown in Fig. 4A were strikingly similar to data appearing in different form in other articles written by different authors at different research institutes, which had already been published. In addition, certain of the data panels shown in Fig. 3C were overlapping, such that the data from the same original source had been selected to represent the results from allegedly differently performed experiments. Owing to the fact that the contentious data in the above article had already been published prior to its submission to Molecular Medicine Reports, the Editor has decided that this paper should be retracted from the Journal. The authors were asked for an explanation to account for these concerns, but the Editorial Office did not receive a reply. The Editor apologizes to the readership for any inconvenience caused. [Molecular Medicine Reports 15: 4217‑4224, 2017; DOI: 10.3892/mmr.2017.6493].
    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
    皮肤黑素瘤(SKCM)是一种高度恶性的皮肤癌,以预后不良和死亡率升高而闻名。RARRES1,一种对视黄酸受体有反应的基因,在各种癌症类型中显示不同的功能。然而,RARRES1在SKCM中的具体作用和潜在机制尚不清楚。GSE15605用于分析RARRES1在SKCM中的表达。随后,TCGA和GEO数据库用于研究RARRES1与临床病理参数之间的关系,以及RARRES1在SKCM中的预后意义和诊断效能。GO,KEGG,进行GSEA分析以探索RARRES1的潜在功能。此外,研究了RARRES1与免疫浸润之间的关联.使用cBioPortal评估SKCM中RARRES1的基因组改变和启动子甲基化水平,UALCAN,和GEO数据库。最后,通过免疫组织化学验证SKCM中RARRES1的表达,通过体内和体外实验阐明了其在SKCM进展中的功能作用。我们发现与正常组织相比,RARRES1在SKCM中下调,这种低表达与SKCM的临床病理特征较差和预后不良有关。通过ROC分析确定RARRES1的诊断效能,是0.732。通过GO,KEGG,和GSEA富集分析,我们确定了30个主要富集在肿瘤免疫微环境中的相关基因和通路,扩散,凋亡,和自噬。此外,发现RARRES1表达与SKCM中各种免疫细胞的浸润呈正相关,特别是巨噬细胞和T辅助细胞,在其他人中。基因组改变和启动子甲基化的分析表明,RARRES1启动子的浅缺失和超甲基化可能导致RARRES1表达降低。IHC验证证实了SKCM中RARRES1的下调。此外,过表达RARRES1抑制A375细胞的增殖和迁移,促进细胞凋亡,并抑制自噬通量。在小鼠异种移植模型中,RARRES1过表达也抑制SKCM肿瘤生长。总的来说,这些研究结果表明,RARRES1可能起到抑制因子的作用,并可能作为SKCM的预后生物标志物和治疗靶点.
    Skin cutaneous melanoma (SKCM) is a highly malignant form of skin cancer, known for its unfavorable prognosis and elevated mortality rate. RARRES1, a gene responsive to retinoic acid receptors, displays varied functions in various cancer types. However, the specific role and underlying mechanisms of RARRES1 in SKCM are still unclear. GSE15605 was utilized to analyze the expression of RARRES1 in SKCM. Subsequently, the TCGA and GEO databases were employed to investigate the relationships between RARRES1 and clinicopathological parameters, as well as the prognostic implications and diagnostic efficacy of RARRES1 in SKCM. GO, KEGG, and GSEA analyses were conducted to explore the potential functions of RARRES1. Furthermore, the associations between RARRES1 and immune infiltration were examined. Genomic alterations and promoter methylation levels of RARRES1 in SKCM were assessed using cBioPortal, UALCAN, and the GEO database. Finally, RARRES1 expression in SKCM was validated through immunohistochemistry, and its functional role in SKCM progression was elucidated via in vivo and in vitro experiments. We found that RARRES1 was downregulated in SKCM compared with normal tissues, and this low expression was associated with worse clinicopathological features and poor prognosis of SKCM. The diagnostic efficacy of RARRES1, as determined by ROC analysis, was 0.732. Through GO, KEGG, and GSEA enrichment analysis, we identified 30 correlated genes and pathways that were mainly enriched in the tumor immune microenvironment, proliferation, apoptosis, and autophagy. Additionally, RARRES1 expression was found to be positively related to the infiltration of various immune cells in SKCM, particularly macrophages and T helper cells, among others. Analysis of genomic alterations and promoter methylation revealed that shallow deletion and hypermethylation of the RARRES1 promoter could lead to reduced RARRES1 expression. IHC validation confirmed the downregulation of RARRES1 in SKCM. Moreover, overexpression of RARRES1 inhibited the proliferation and migration of A375 cells, promoted apoptosis, and inhibited autophagic flux. In the mouse xenograft model, RARRES1 overexpression also suppressed SKCM tumor growth. Collectively, these findings suggest that RARRES1 may function as a suppressor and could potentially serve as a prognostic biomarker and therapeutic target for SKCM.
    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
    食管癌相关基因2(ECRG2),也称为丝氨酸肽酶抑制剂Kazal7型(SPINK7),是来自SPINK基因家族的新型肿瘤抑制基因,具有抗癌潜力。ECRG2最初是在努力发现与食管肿瘤发生有关的基因期间鉴定的。ECRG2是在原发性人类食道癌中表达缺失或降低的基因之一。此外,在几种其他类型的人类恶性肿瘤中也注意到ECRG2表达缺失或降低。在各种原发性人类癌症中鉴定了ECRG2错义突变。据报道,源自癌症的ECRG2突变体(位置30处的缬氨酸至谷氨酸)未能诱导由DNA损伤性抗癌药物触发的细胞死亡和半胱天冬酶激活。此外,ECRG2抑制了培养细胞和动物移植肿瘤中的癌细胞增殖,并抑制了癌细胞的迁移/侵袭和转移。ECRG2也被鉴定为Hu抗原R(HuR)的负调节因子,一种致癌RNA结合蛋白,已知可调节mRNA稳定性和与许多癌症相关基因相对应的转录本的表达。ECRG2功能对于调节食管中的炎症反应和维持上皮屏障完整性也是重要的。最近,ECRG2被发现是p53促凋亡转录靶标的最新成员之一。在ECRG2基因启动子的近端区域内发现了两个p53结合位点(BS-1和BS-2);癌细胞中DNA损伤剂的处理显着增加了p53与ECRG2启动子的结合,并在DNA损伤后触发了强的ECRG2启动子诱导。Further,ECRG2表达的基因缺失显著阻碍了癌细胞中DNA损伤和野生型p53诱导的凋亡细胞死亡。这些发现表明,ECRG2表达的丧失,通常在人类癌症中观察到,可能在赋予人类癌症抗癌药物抗性方面发挥重要作用。因此,ECRG2是DNA损伤诱导的细胞死亡的新型调节剂,也可能是抗癌治疗的潜在靶标。
    Esophageal Cancer-Related Gene 2 (ECRG2), also known as Serine Peptidase Inhibitor Kazal type 7 (SPINK7), is a novel tumor suppressor gene from the SPINK family of genes that exhibits anticancer potential. ECRG2 was originally identified during efforts to discover genes involved in esophageal tumorigenesis. ECRG2 was one of those genes whose expression was absent or reduced in primary human esophageal cancers. Additionally, absent or reduced ECRG2 expression was also noted in several other types of human malignancies. ECRG2 missense mutations were identified in various primary human cancers. It was reported that a cancer-derived ECRG2 mutant (valine to glutamic acid at position 30) failed to induce cell death and caspase activation triggered by DNA-damaging anticancer drugs. Furthermore, ECRG2 suppressed cancer cell proliferation in cultured cells and grafted tumors in animals and inhibited cancer cell migration/invasion and metastasis. ECRG2 also was identified as a negative regulator of Hu-antigen R (HuR), an oncogenic RNA-binding protein that is known to regulate mRNA stability and the expression of transcripts corresponding to many cancer-related genes. ECRG2 function is important also for the regulation of inflammatory responses and the maintenance of epithelial barrier integrity in the esophagus. More recently, ECRG2 was discovered as one of the newest members of the pro-apoptotic transcriptional targets of p53. Two p53-binding sites (BS-1 and BS-2) were found within the proximal region of the ECRG2 gene promoter; the treatment of DNA-damaging agents in cancer cells significantly increased p53 binding to the ECRG2 promoter and triggered a strong ECRG2 promoter induction following DNA damage. Further, the genetic depletion of ECRG2 expression significantly impeded apoptotic cell death induced by DNA damage and wild-type p53 in cancer cells. These findings suggest that the loss of ECRG2 expression, commonly observed in human cancers, could play important roles in conferring anticancer drug resistance in human cancers. Thus, ECRG2 is a novel regulator in DNA damage-induced cell death that may also be a potential target for anticancer therapeutics.
    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
    BMI1多梳环指原癌基因(BMI1)参与不同癌症的发病机制,包括急性髓系白血病(AML)。然而,BMI1(circularRNA,circBMI1)的作用尚未被研究。我们的研究旨在探讨circBMI1在AML中的作用和机制。从AML患者吸出的骨髓单个核细胞中circBMI1显着降低。受试者工作特征曲线分析显示circBMI1可以区分AML患者和对照组。通过在HL-60细胞中过度表达和敲低circBMI1,我们发现circBMI1抑制细胞增殖,促进细胞凋亡,AML的化疗药物敏感性增加。使用严重的联合免疫缺陷小鼠和circBMI1转基因小鼠的实验表明,circBMI1过表达的小鼠白细胞计数较低,这表明不太严重的AML入侵。RNA免疫沉淀和双荧光素酶报告分析揭示了circBMI1,miR-338-5p,和DNA结合的抑制剂4(ID4)。挽救实验证明,circBMI1通过结合miR-338-5p抑制AML进展,这影响了ID4的表达。通过将从circBMI1-HL-60和小干扰circBMI1-HL-60细胞中提取的外泌体与HL-60细胞共培养,我们发现circBMI1-HL-60细胞的外泌体显示出肿瘤抑制作用,即抑制HL-60增殖,促进细胞凋亡,增加化疗药物的敏感性。来自小干扰circBMI1-HL-60细胞的外泌体显示出相反的效果。circBMI1可以作为外泌体依赖性肿瘤抑制剂。circBMI1,临床诊断的潜在生物标志物,通过调节miR-338-5p/ID4在AML中充当肿瘤抑制因子,并可能通过外泌体分泌影响AML的发病机制。
    BMI1 Polycomb Ring Finger Proto-Oncogene (BMI1) is involved in the pathogenesis of different cancers, including acute myeloid leukemia (AML). However, the role of the circular RNA of BMI1 (circBMI1) has not been studied. Our study aimed to investigate the role and mechanism of circBMI1 in AML. circBMI1 was significantly decreased in bone marrow mononuclear cells aspirated from patients with AML. Receiver operating characteristic curve analysis showed that circBMI1 could distinguish patients with AML from controls. By overexpressing and knocking down circBMI1 in HL-60 cells, we found that circBMI1 inhibited cell proliferation, promoted apoptosis, and increased chemotherapeutic drug sensitivity in AML. Experiments using severe combined immune-deficient mice and circBMI1 transgenic mice showed that mice with circBMI1 overexpression had lower white blood cell counts, which suggested less severe AML invasion. RNA immunoprecipitation and dual-luciferase reporter assay revealed binding sites among circBMI1, miR-338-5p, and inhibitor of DNA binding 4 (ID4). Rescue experiments proved that circBMI1 inhibited AML progression by binding to miR-338-5p, which affected the expression of ID4. By coculturing exosomes extracted from circBMI1-HL-60 and small interfering circBMI1-HL-60 cells with HL-60 cells, we found that exosomes from circBMI1-HL-60 cells showed tumor suppressive effects, namely inhibiting HL-60 proliferation, promoting apoptosis, and increasing chemotherapeutic drug sensitivity. Exosomes from small interfering circBMI1-HL-60 cells showed the opposite effects. circBMI1 may act as an exosome-dependent tumor inhibitor. circBMI1, a potential biomarker for clinical diagnosis, acts as a tumor suppressor in AML by regulating miR-338-5p/ID4 and might affect the pathogenesis of AML by exosome secretion.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号