MCF-10A

MCF - 10A
  • 文章类型: Journal Article
    三阴性乳腺癌(TNBC)是一种侵袭性亚型,其特征是缺乏雌激素受体,孕激素受体,和人表皮生长因子受体2型表达。它以其高度恶性而闻名,侵入性,和转移倾向,由于缺乏有益的治疗靶标,导致预后不良。源自蘑菇的天然产物由于其潜在的药用特性而在肿瘤治疗中获得了极大的关注。灵芝在乳腺癌中的治疗潜力已被我们小组强调,建议将其用作辅助治疗。本研究旨在评估在波多黎各发现的两种加勒比海本地灵芝物种的潜在抗肿瘤能力,多毛灵芝(G.Multiicatum)和马丁尼克灵芝(G.马丁尼克)。抗增殖研究通过培养后的细胞活力测定进行,收获,和两个物种的分馏。获得的结果表明,大多数级分对所有细胞系都显示出一定的细胞毒性,但33%的级分(F1、F2、F7、F12)显示对癌细胞模型的选择性。我们首次证明了天然灵芝物种可以产生具有抗TNBC特性的代谢物。未来的途径将集中在这些灵芝提取物中最活跃的部分的结构阐明上。
    Triple-negative breast cancer (TNBC) is an aggressive subtype characterized by the absence of estrogen receptor, progesterone receptor, and human epidermal growth factor receptor type 2 expression. It is known for its high malignancy, invasiveness, and propensity for metastasis, resulting in a poor prognosis due to the absence of beneficial therapeutic targets. Natural products derived from mushrooms have gained significant attention in neoplastic therapy due to their potential medicinal properties. The therapeutic potential of Ganoderma lucidum in breast cancer has been highlighted by our group, suggesting its use as an adjuvant treatment. The present study aims to assess the potential antineoplastic capacity of two Caribbean native Ganoderma species found in Puerto Rico, Ganoderma multiplicatum (G. multiplicatum) and Ganoderma martinicense (G. martinicense). Antiproliferative studies were conducted via cell viability assays after cultivation, harvesting, and fractionation of both species. The obtained results indicate that most of the fractions show some cytotoxicity against all cell lines, but 33% of the fractions (F1, F2, F7, F12) display selectivity towards cancer cell models. We demonstrate for the first time that native Ganoderma species can generate metabolites with anti-TNBC properties. Future avenues will focus on structure elucidation of the most active fractions of these Ganoderma extracts.
    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
    硒是一种必需的微量元素,可共同转化为硒蛋白,具有重要的生物学功能。长期以来,补充硒对健康有益。然而,过量摄入硒时观察到细胞毒性。这项研究的目的是研究正常人乳腺上皮细胞模型中对含硒氨基酸硒代蛋氨酸和硒代半胱氨酸的反应的代谢途径。我们显示硒代蛋氨酸和硒代半胱氨酸在与癌性MCF-7和Hela细胞相同的浓度范围内抑制非癌性MCF-10A细胞的增殖,导致细胞凋亡。MCF-10A细胞中的硒酸暴露导致游离低分子量硫醇的严重消耗,这可能解释了观察到的氧化应激途径转录因子NRF2表达的上调。硒代蛋氨酸和硒代半胱氨酸均诱导未折叠蛋白反应靶基因(GRP78,ATF4,CHOP)的表达。使用针对内质网(ER)的氧化还原敏感荧光探针,我们表明,两种硒代氨基酸都将ER氧化还原平衡转向更加氧化的环境。这些结果表明,ER氧化还原状态的改变可能会破坏蛋白质折叠并引起暴露于硒氨基酸的MCF-10A细胞中ER应激诱导的凋亡。
    Selenium is an essential trace element co-translationally incorporated into selenoproteins with important biological functions. Health benefits have long been associated with selenium supplementation. However, cytotoxicity is observed upon excessive selenium intake. The aim of this study is to investigate the metabolic pathways underlying the response to the selenium-containing amino acids selenomethionine and selenocysteine in a normal human breast epithelial cell model. We show that both selenomethionine and selenocystine inhibit the proliferation of non-cancerous MCF-10A cells in the same concentration range as cancerous MCF-7 and Hela cells, which results in apoptotic cell death. Selenocystine exposure in MCF-10A cells caused a severe depletion of free low molecular weight thiols, which might explain the observed upregulation of the expression of the oxidative stress pathway transcription factor NRF2. Both selenomethionine and selenocystine induced the expression of target genes of the unfolded protein response (GRP78, ATF4, CHOP). Using a redox-sensitive fluorescent probe targeted to the endoplasmic reticulum (ER), we show that both selenoamino acids shifted the ER redox balance towards an even more oxidizing environment. These results suggest that alteration of the redox state of the ER may disrupt protein folding and cause ER stress-induced apoptosis in MCF-10A cells exposed to selenoamino acids.
    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
    在这项研究中,我们提出了一系列产生亚氨基的NNO三齿配体,酰胺基和氧代供体口袋用于Pd(II)配位。所有化合物都经过元素分析和先进的光谱技术精心表征,包括FTIR,质子和碳核磁共振。合成的化合物经过严格的评估,作为抗癌药物的潜力,利用侵袭性乳腺癌细胞系MDA-MB(ATCC)和MCF-7作为评估癌细胞生长抑制的关键模型。值得注意的是,MTT分析揭示了所有钯复合物对MDA-MB-231和MCF-7细胞的强大抗癌活性。特别是,复合物[Pd(L1)(CH3CN)]表现出优异的效力,IC50值为25.50±0.30µM(MDA-MB-231)和20.76±0.30µM(MCF-7),与顺铂各自的27.00±0.80µM和24.10±0.80µM相比,强调了其有前途的治疗潜力。此外,为了阐明抗癌作用的机理基础,酪氨酸激酶的分子对接研究,癌症研究中不可或缺的目标,进行了。这些研究的结果进一步证实了这些创新化合物固有的显著抗癌特性。这项研究为根植于配体和Pd(II)复合物之间的协同作用的有效抗癌剂的开发提供了令人信服的观点,并为未来的癌症治疗努力提供了有希望的途径。
    In this study we presented a novel series of NNO tridentate ligands generating imino, amido and oxo donor pocket for Pd(II) coordination. All the compounds were meticulously characterized by elemental analysis and advanced spectroscopic techniques, including FTIR, proton and carbon NMR. The synthesized compounds underwent rigorous evaluation for their potential as anti-cancer agents, utilizing the aggressive breast cancer cell lines MDA-MB (ATCC) and MCF-7 as a crucial model for assessing growth inhibition in cancer cells. Remarkably, the MTT assay unveiled the robust anti-cancer activity for all palladium complexes against MDA-MB-231 and MCF-7 cells. Particularly, complex [Pd(L1)(CH3CN)] exhibited exceptional potency with an IC50 value of 25.50 ± 0.30 µM (MDA-MB-231) and 20.76 ± 0.30 µM (MCF-7), compared to respective 27.00 ± 0.80 µM and 24.10 ± 0.80 µM for cisplatin, underscoring its promising therapeutic potential. Furthermore, to elucidate the mechanistic basis for the anti-cancer effects, molecular docking studies on tyrosine kinases, an integral target in cancer research, were carried out. The outcome of these investigations further substantiated the remarkable anticancer properties inherent to these innovative compounds. This research offers a compelling perspective on the development of potent anti-cancer agents rooted in the synergy between ligands and Pd(II) complexes and presenting a promising avenue for future cancer therapy endeavors.
    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:乳腺癌是女性最常见的癌症,大约10-15%的新病例被归类为三阴性乳腺癌(TNBC)。传统的化疗通常对正常细胞有毒性。因此,重要的是发现靶向TNBC同时对正常细胞造成最小损伤的新抗癌化合物。受体酪氨酸激酶样孤儿受体1(ROR1)是一种在许多人类恶性肿瘤中过表达的癌胚蛋白,包括TNBC。本研究调查了靶向ROR1的潜在小分子。方法:使用AutoDockVina和Glide,我们筛选了70,000种化学物质进行调查。我们通过协商一致投票获得了10个代表性化合物,删除结构性警报,和聚类。手动评估后,选择化合物2和4进行MD模拟和细胞活力实验。化合物4在活力测定中显示出有希望的结果,这导致我们进一步进行细胞凋亡测定和免疫印迹。结果:化合物4(CID1261330)具有-6.635和-10.8的对接评分。它适合口袋,并显示与GLU64,ASP174和PHE93的相互作用。其RMSD在0.20nm附近波动并形成两个稳定的H键,表明化合物4的稳定性。它抑制MDA-MB-231,HCC1937和HCC1395细胞系中的细胞增殖,IC50值大约为2μM至10μM,分别。化合物4不杀死非恶性上皮乳腺细胞MCF-10A(IC50>27μM)。这些结果通过MDA-MB-231细胞中的大量凋亡细胞(47.6%)而不是MCF-10A细胞中的大量凋亡细胞(7.3%)得到证实。免疫印迹分析在相同方向上提供了额外的支持。讨论:这些发现共同表明化合物4具有有效消除TNBC细胞同时对正常乳腺细胞造成最小伤害的潜力。这项研究的有希望的结果为进一步测试化合物4在其他以ROR1上调为特征的恶性肿瘤中奠定了基础。作为其更广泛适用性的概念证明。
    Introduction: Breast cancer is the most common cancer in women, with roughly 10-15% of new cases classified as triple-negative breast cancer (TNBC). Traditional chemotherapies are often toxic to normal cells. Therefore, it is important to discover new anticancer compounds that target TNBC while causing minimal damage to normal cells. Receptor tyrosine kinase-like Orphan Receptor 1 (ROR1) is an oncofetal protein overexpressed in numerous human malignancies, including TNBC. This study investigated potential small molecules targeting ROR1. Methodology: Using AutoDock Vina and Glide, we screened 70,000 chemicals for our investigation. We obtained 10 representative compounds via consensus voting, deleting structural alerts, and clustering. After manual assessment, compounds 2 and 4 were chosen for MD simulation and cell viability experiment. Compound 4 showed promising results in the viability assay, which led us to move further with the apoptosis assay and immunoblotting. Results: Compound 4 (CID1261330) had docking scores of -6.635 and -10.8. It fits into the pocket and shows interactions with GLU64, ASP174, and PHE93. Its RMSD fluctuates around 0.20 nm and forms two stable H-bonds indicating compound 4 stability. It inhibits cell proliferation in MDA-MB-231, HCC1937, and HCC1395 cell lines, with IC50 values of approximately 2 μM to 10 μM, respectively. Compound 4 did not kill non-malignant epithelial breast cells MCF-10A (IC50 > 27 μM). These results were confirmed by the significant number of apoptotic cells in MDA-MB-231 cells (47.6%) but not in MCF-10A cells (7.3%). Immunoblot analysis provided additional support in the same direction. Discussion: These findings collectively suggest that compound 4 has the potential to effectively eliminate TNBC cells while causing minimal harm to normal breast cells. The promising outcomes of this study lay the groundwork for further testing of compound 4 in other malignancies characterized by ROR1 upregulation, serving as a proof-of-concept for its broader applicability.
    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
    永生化人乳腺上皮细胞系MCF-10A的转染效率仍然是一个需要解决的问题。在这项研究中,其目的是使用磁性纳米颗粒(MNPs)和简单的磁体通过磁转染法将重组DNA(pCMV-Azu-GFP)递送至MCF-10A细胞以加速DNA递送。表面正改性二氧化硅包覆氧化铁MNPs(MSNP-NH2)的制备和表征通过TEM,FTIR,和DLS分析。通过整合密码子优化的天青蛋白以产生融合蛋白来获得重组DNA(rDNA)。然后,通过序列分析验证了在大肠杆菌细胞中克隆的rDNA。通过琼脂糖凝胶电泳研究了MSNP-NH2上与增强子聚乙烯亚胺(PEI)静电缀合的rDNA,并确定了适用于细胞的最佳条件。基于MTS测试,在处理的细胞上观察到剂量依赖性统计差异。使用激光扫描共聚焦显微镜成像和蛋白质印迹分析确定磁转染后融合蛋白的表达。观察到天青素基因可以通过磁转染转移到MCF-10A细胞中。因此,当天青蛋白基因被用作乳腺癌治疗剂时,它可以在健康细胞中表达而没有毒性作用。
    Transfection efficiency of the immortalized human breast epithelial cell line MCF-10A remains an issue that needs to be resolved. In this study, it was aimed to deliver a recombinant DNA (pCMV-Azu-GFP) to the MCF-10A cells by the magnetofection method using magnetic nanoparticles (MNPs) and a simple magnet to accelerate the DNA delivery. Surface positively modified silica-coated iron oxide MNPs (MSNP-NH2) were produced and characterized via TEM, FTIR, and DLS analyses. The recombinant DNA (rDNA) was obtained by the integration of codon-optimized azurin to produce a fusion protein. Then, rDNA cloned in Escherichia coli cells was validated by sequence analysis. The electrostatically conjugated rDNA on MSNP-NH2 with an enhancer polyethyleneimine (PEI) was studied by agarose gel electrophoresis and the optimum conditions were determined to apply to the cell. A dose-dependent statistical difference was observed on treated cells based on the MTS test. The expression of the fusion protein after magnetofection was determined using laser scanning confocal microscope imaging and western blot analysis. It was observed that the azurin gene could be transferred to MCF-10A cells by magnetofection. Thus, when the azurin gene is used as a breast cancer treatment agent, it can be expressed in healthy cells without toxic effects.
    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:乳腺癌暴露于极低频磁场(ELF-MF)会导致各种生物学反应。一些研究表明可能有增强癌症的作用,而其他人则显示出可能的治疗作用。这项研究调查了体外暴露于50HzELF-MF长达24小时对MDA-MB-231和MCF-7乳腺癌细胞系和MCF-10A乳腺癌细胞系的活力和细胞反应的影响。
    结果:将乳腺细胞系以0.1mT和1.0mT的通量密度暴露于50HzELF-MF,并在ELF-MF暴露开始后96小时进行检查。50HzELF-MF暴露的持续时间会影响肿瘤和非致瘤性乳腺细胞系的细胞活力和增殖。特别是,短期暴露(4-8小时,0.1mT和1.0mT)导致乳腺癌细胞的生存力增加,长时间和高暴露(24小时,1.0mT)导致所有细胞系的活力和增殖降低。癌症和正常乳腺细胞对ELF-MF表现出不同的反应。ELF-MF暴露后,线粒体膜电位和活性氧(ROS)的产生发生了变化,表明线粒体是乳腺细胞中ELF-MF的可能靶标。
    结论:在与普通人群和工作场所暴露的限制相一致的磁通量密度下,ELF-MF暴露会影响乳腺细胞的体外活力。96小时后效果明显,与ELF-MF暴露时间有关。
    BACKGROUND: The exposure of breast cancer to extremely low frequency magnetic fields (ELF-MFs) results in various biological responses. Some studies have suggested a possible cancer-enhancing effect, while others showed a possible therapeutic role. This study investigated the effects of in vitro exposure to 50 Hz ELF-MF for up to 24 h on the viability and cellular response of MDA-MB-231 and MCF-7 breast cancer cell lines and MCF-10A breast cell line.
    RESULTS: The breast cell lines were exposed to 50 Hz ELF-MF at flux densities of 0.1 mT and 1.0 mT and were examined 96 h after the beginning of ELF-MF exposure. The duration of 50 Hz ELF-MF exposure influenced the cell viability and proliferation of both the tumor and nontumorigenic breast cell lines. In particular, short-term exposure (4-8 h, 0.1 mT and 1.0 mT) led to an increase in viability in breast cancer cells, while long and high exposure (24 h, 1.0 mT) led to a decrease in viability and proliferation in all cell lines. Cancer and normal breast cells exhibited different responses to ELF-MF. Mitochondrial membrane potential and reactive oxygen species (ROS) production were altered after ELF-MF exposure, suggesting that the mitochondria are a probable target of ELF-MF in breast cells.
    CONCLUSIONS: The viability of breast cells in vitro is influenced by ELF-MF exposure at magnetic flux densities compatible with the limits for the general population and for workplace exposures. The effects are apparent after 96 h and are related to the ELF-MF exposure time.
    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
    三阴性乳腺癌(TNBC)由于缺乏靶向治疗和增加的复发概率而对治疗提出了重大挑战。确定TNBC中的漏洞并开发更新的治疗方法是相关的。我们先前的研究表明,转录因子EB(TFEB)通过调节DNA修复是TNBC存活所必需的,凋亡信号,和细胞周期。然而,TFEB靶向DNA修复和细胞周期通路的具体机制尚不清楚,这些影响是否决定了TNBC的生存还有待确定。这里,我们发现在MDA-MB-231TNBC细胞中,TFEB敲除降低了参与DNA复制和细胞周期进程的基因和蛋白的表达。缺乏TFEB的细胞中DNA复制减少,由EdU公司衡量。非癌性MCF10A和MDA-MB-231细胞中的TFEB沉默在G1/S同步后通过S期的进展受损;然而,这种增殖缺陷不能通过抑制因子RB1的共敲减来挽救。相反,TFEB敲低降低了G1和早期S期MDA-MB-231细胞中的起源许可。TFEB沉默与MCF10A的复制应激相关,但与TNBC细胞无关。最后,我们发现TFEB敲低使TNBC细胞对Aurora激酶A的抑制剂更敏感,一种促进有丝分裂的蛋白质。因此,抑制TFEB通过减少来源许可而损害细胞周期进程,导致延迟进入S阶段,同时使细胞对极光激酶A抑制剂敏感并降低TNBC细胞活力。相比之下,非癌细胞中的TFEB沉默与复制应激相关并导致G1/S阻滞。
    Triple-negative breast cancer (TNBC) poses significant challenges for treatment given the lack of targeted therapies and increased probability of relapse. It is pertinent to identify vulnerabilities in TNBC and develop newer treatments. Our prior research demonstrated that transcription factor EB (TFEB) is necessary for TNBC survival by regulating DNA repair, apoptosis signaling, and the cell cycle. However, specific mechanisms by which TFEB targets DNA repair and cell cycle pathways are unclear, and whether these effects dictate TNBC survival is yet to be determined. Here, we show that TFEB knockdown decreased the expression of genes and proteins involved in DNA replication and cell cycle progression in MDA-MB-231 TNBC cells. DNA replication was decreased in cells lacking TFEB, as measured by EdU incorporation. TFEB silencing in MDA-MB-231 and noncancerous MCF10A cells impaired progression through the S-phase following G1/S synchronization; however, this proliferation defect could not be rescued by co-knockdown of suppressor RB1. Instead, TFEB knockdown reduced origin licensing in G1 and early S-phase MDA-MB-231 cells. TFEB silencing was associated with replication stress in MCF10A but not in TNBC cells. Lastly, we identified that TFEB knockdown renders TNBC cells more sensitive to inhibitors of Aurora Kinase A, a protein facilitating mitosis. Thus, inhibition of TFEB impairs cell cycle progress by decreasing origin licensing, leading to delayed entry into the S-phase, while rendering TNBC cells sensitive to Aurora kinase A inhibitors and decreasing cell viability. In contrast, TFEB silencing in noncancerous cells is associated with replication stress and leads to G1/S arrest.
    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
    在乳腺癌患者的保乳治疗(BCT)和放疗后应用脂肪转移是重建患病乳房体积不足的广泛方法。然而,由于相互矛盾的数据,脂转移的安全性仍未得到澄清.这项体外研究的目的是进一步阐明脂肪转移对经照射的剩余乳腺组织的潜在影响。将乳腺上皮细胞系MCF-10A与成纤维细胞系MRC-5共培养并用2和5Gy照射。之后,细胞用来自脂肪来源的干细胞(ADSC)的条件培养基(CM)处理,分析了MCF-10A和MRC-5细胞对MCF-10A细胞功能和mRNA水平基因表达的影响。用ADSCCM处理刺激了MCF-10A细胞的迁移和侵袭并降低了存活分数。Further,细胞因子的表达,细胞外,乳腺上皮细胞间充质标志物增强。只能观察到ADSCCM对照射的成纤维细胞的作用。目前的数据表明上皮乳腺细胞系中上皮-间质转化样的变化。因此,应严格权衡BCT后脂肪转运的益处及其对受影响患者的可能风险.
    The application of lipotransfer after breast-conserving therapy (BCT) and irradiation in breast cancer patients is an already widespread procedure for reconstructing volume deficits of the diseased breast. Nevertheless, the safety of lipotransfer has still not been clarified yet due to contradictory data. The goal of this in vitro study was to further elucidate the potential effects of lipotransfer on the irradiated remaining breast tissue. The mammary epithelial cell line MCF-10A was co-cultured with the fibroblast cell line MRC-5 and irradiated with 2 and 5 Gy. Afterwards, cells were treated with conditioned medium (CM) from adipose-derived stem cells (ADSC), and the effects on the cellular functions of MCF-10A cells and on gene expression at the mRNA level in MCF-10A and MRC-5 cells were analyzed. Treatment with ADSC CM stimulated transmigration and invasion and decreased the surviving fraction of MCF-10A cells. Further, the expression of cytokines, extracellular, and mesenchymal markers was enhanced in mammary epithelial cells. Only an effect of ADSC CM on irradiated fibroblasts could be observed. The present data suggest epithelial-mesenchymal transition-like changes in the epithelial mammary breast cell line. Thus, the benefits of lipotransfer after BCT should be critically weighed against its possible risks for the affected patients.
    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
    癌症基因组的特征是小规模体细胞突变以及大规模染色体缺失的积累。扩增,和复杂的结构重组。该特征至少部分取决于癌细胞经历反复染色体断裂的能力。为了解决染色体结构重排断点与复发性DNA双链断裂(DSB)相关的程度,我们同时在雌激素受体(ER)阳性乳腺癌细胞系MCF-7和非癌对照乳腺上皮细胞系MCF-10A中定位染色体结构变异断点(使用全基因组DNA-seq)和自发DSB形成(使用Break-seq).我们在MCF-7细胞中几乎仅在染色体16q的着丝粒区域中确定了并发的DSB和结构变异断点。我们微调了16q上拷贝数变异断点的鉴定。此外,我们检测到MCF-7和MCF-10A中发生的复发DSB。我们提出了一个DSB驱动的染色体重排模型,该模型导致16q的易位,可能是10q,以及最终的16q损失,不涉及16q的着丝粒。我们从RNA-seq数据中提供了选择基因的证据,包括SHCBP1,ORC6和MYLK3,它们紧接在16q着丝粒的下游,与对照相比,在MCF-7细胞系中的表达增加。癌症基因组图谱发表的数据显示,所有这三种基因在乳腺肿瘤样品中的表达都增加。我们发现SHCBP1和ORC6在ER阳性乳腺癌队列中都是强烈的不良预后和治疗结果标志物。我们认为这些基因是乳腺癌进展的潜在癌基因。应通过鉴定在染色体重排过程中获得表达的潜在癌基因来增强对伴随16q损失的肿瘤抑制物损失的搜索。
    Cancer genomes are characterized by the accumulation of small-scale somatic mutations as well as large-scale chromosomal deletions, amplifications, and complex structural rearrangements. This characteristic is at least partially dependent on the ability of cancer cells to undergo recurrent chromosome breakage. In order to address the extent to which chromosomal structural rearrangement breakpoints correlate with recurrent DNA double-strand breaks (DSBs), we simultaneously mapped chromosome structural variation breakpoints (using whole-genome DNA-seq) and spontaneous DSB formation (using Break-seq) in the estrogen receptor (ER)-positive breast cancer cell line MCF-7 and a non-cancer control breast epithelium cell line MCF-10A. We identified concurrent DSBs and structural variation breakpoints almost exclusively in the pericentromeric region of chromosome 16q in MCF-7 cells. We fine-tuned the identification of copy number variation breakpoints on 16q. In addition, we detected recurrent DSBs that occurred in both MCF-7 and MCF-10A. We propose a model for DSB-driven chromosome rearrangements that lead to the translocation of 16q, likely with 10q, and the eventual 16q loss that does not involve the pericentromere of 16q. We present evidence from RNA-seq data that select genes, including SHCBP1, ORC6, and MYLK3, which are immediately downstream from the 16q pericentromere, show heightened expression in MCF-7 cell line compared to the control. Data published by The Cancer Genome Atlas show that all three genes have increased expression in breast tumor samples. We found that SHCBP1 and ORC6 are both strong poor prognosis and treatment outcome markers in the ER-positive breast cancer cohort. We suggest that these genes are potential oncogenes for breast cancer progression. The search for tumor suppressor loss that accompanies the 16q loss ought to be augmented by the identification of potential oncogenes that gained expression during chromosomal rearrangements.
    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
    咖啡酸功能化的Au-Fe3O4,Pt-Fe3O4和Pd-Fe3O4纳米异二聚体(NHDs)对2D和3D乳腺肿瘤(MCF-7)和健康乳腺上皮细胞(MCF-10A)的X射线增强作用通过执行细胞活力,活性氧(ROS)检测,酶活性,和克隆检测。当暴露于1Gy的单剂量时,观察到细胞内NHD会导致DNA断裂并增强MCF-7细胞中的超氧化物和羟基自由基形成。将MCF-7衍生的多细胞肿瘤球体(MCF-7MCTS)和MCF-10A衍生的多细胞球体(MCF-10AMCS)与NHD一起孵育,并用5个每日剂量的2Gy照射。Au-Fe3O4和Pt-Fe3O4NHD负载的MCF-7MCTS暴露于分馏辐照后,体积显着减少,而细胞内Au-Fe3O4和Pt-Fe3O4NHDs促进了辐照的MCF-10AMCS的生长。此外,在非癌性MCF-10A细胞中,Au-Fe3O4和Pt-Fe3O4NHD可阻止ROS形成并抑制DNA链断裂。另一方面,Pd-Fe3O4NHD增强了X辐射诱导的MCF-10A细胞损伤,这归因于ROS清除酶活性的损害。总之,咖啡酸官能化的Au-Fe3O4和Pt-Fe3O4NHD在乳腺肿瘤细胞中作为优异的X射线增强剂,同时保护健康的乳腺上皮细胞免受X射线辐射.
    The X-radiation enhancing effect of caffeic acid-functionalized Au-Fe3O4, Pt-Fe3O4, and Pd-Fe3O4 nanoheterodimers (NHDs) on 2D and 3D breast tumor (MCF-7) and healthy breast epithelial (MCF-10A) cells was comprehensively examined by performing cell viability, reactive oxygen species (ROS) detection, enzyme activity, and clonogenic assays. Intracellular NHDs were observed to cause DNA fragmentation and to enhance superoxide and hydroxyl radical formation in MCF-7 cells when exposed to a single dose of 1 Gy. MCF-7-derived multicellular tumor spheroids (MCF-7 MCTS) and MCF-10A-derived multicellular spheroids (MCF-10A MCS) were incubated with the NHDs and irradiated with five daily doses of 2 Gy. The Au-Fe3O4 and Pt-Fe3O4 NHD-loaded MCF-7 MCTS significantly decreased in volume after being exposed to fractionated irradiation, whereas intracellular Au-Fe3O4 and Pt-Fe3O4 NHDs promoted the growth of the irradiated MCF-10A MCS. Moreover, Au-Fe3O4 and Pt-Fe3O4 NHDs were shown to impede ROS formation and inhibit DNA strand breakage in the noncancerous MCF-10A cells. On the other hand, the Pd-Fe3O4 NHDs boosted X-radiation-induced damage of MCF-10 A cells, which was ascribed to impairment of the ROS scavenging enzyme activities. In summary, caffeic acid-functionalized Au-Fe3O4 and Pt-Fe3O4 NHDs performed as excellent X-radiation enhancing agents in breast tumor cells, while they protect healthy breast epithelial cells against X-radiation.
    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