Drug Resistance, Neoplasm

抗药性, 肿瘤
  • 文章类型: Journal Article
    背景:索拉非尼是晚期肝细胞癌(HCC)的标准治疗方法,但治疗过程中的获得性耐药性极大地限制了其临床疗效。脂质代谢紊乱在肝癌发生中起重要作用。然而,脂质代谢重编程是否以及如何调节HCC细胞的索拉非尼耐药仍不清楚。方法:采用连续诱导的方法建立索拉非尼耐药肝癌细胞。UHPLC-MS/MS,蛋白质组学,和流式细胞术用于评估脂质代谢。ChIP和Westernblot用于反映信号转导和转录激活因子3(STAT3)与甘油-3-磷酸酰基转移酶3(GPAT3)的相互作用。应用功能增益和功能丧失研究探讨肝癌索拉非尼耐药的驱动机制。流式细胞术和CCK8体外,和体内肿瘤大小用于评估肝癌细胞的索拉非尼敏感性。结果:我们的代谢组数据显示,在索拉非尼抗性HCC细胞中甘油三酯显著富集。使用蛋白质组学和基因组学技术的进一步分析表明,索拉非尼抗性组中GPAT3的表达显着增加,这被发现依赖于STAT3的激活。GPAT3复敏肝癌细胞对索拉非尼的恢复,而GPAT3的过表达导致索拉非尼不敏感。机械上,GPAT3上调增加甘油三酯合成,进而刺激NF-κB/Bcl2信号通路,导致索拉非尼治疗后的细胞凋亡耐受性。此外,我们的体外和体内研究表明,pan-GPAT抑制剂可有效逆转HCC细胞中的索拉非尼耐药.结论:我们的数据表明,肝癌细胞中GPAT3的升高重新编程甘油三酯代谢,这有助于对索拉非尼的获得性抗性,这表明GPAT3是增强HCC对索拉非尼敏感性的潜在靶标。
    Background: Sorafenib is the standard treatment for advanced hepatocellular carcinoma (HCC), but acquired resistance during the treatment greatly limits its clinical efficiency. Lipid metabolic disorder plays an important role in hepatocarcinogenesis. However, whether and how lipid metabolic reprogramming regulates sorafenib resistance of HCC cells remains vague. Methods: Sorafenib resistant HCC cells were established by continuous induction. UHPLC-MS/MS, proteomics, and flow cytometry were used to assess the lipid metabolism. ChIP and western blot were used to reflect the interaction of signal transducer and activator of transcription 3 (STAT3) with glycerol-3-phosphate acyltransferase 3 (GPAT3). Gain- and loss-of function studies were applied to explore the mechanism driving sorafenib resistance of HCC. Flow cytometry and CCK8 in vitro, and tumor size in vivo were used to evaluate the sorafenib sensitivity of HCC cells. Results: Our metabolome data revealed a significant enrichment of triglycerides in sorafenib-resistant HCC cells. Further analysis using proteomics and genomics techniques demonstrated a significant increase in the expression of GPAT3 in the sorafenib-resistant groups, which was found to be dependent on the activation of STAT3. The restoration of GPAT3 resensitized HCC cells to sorafenib, while overexpression of GPAT3 led to insensitivity to sorafenib. Mechanistically, GPAT3 upregulation increased triglyceride synthesis, which in turn stimulated the NF-κB/Bcl2 signaling pathway, resulting in apoptosis tolerance upon sorafenib treatment. Furthermore, our in vitro and in vivo studies revealed that pan-GPAT inhibitors effectively reversed sorafenib resistance in HCC cells. Conclusions: Our data demonstrate that GPAT3 elevation in HCC cells reprograms triglyceride metabolism which contributes to acquired resistance to sorafenib, which suggests GPAT3 as a potential target for enhancing the sensitivity of HCC to sorafenib.
    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
    乳腺癌,一个主要的全球健康问题,需要不断探索新的治疗策略。Palbociclib(PAL),一种众所周知的细胞周期蛋白依赖性激酶(CDK)抑制剂,在乳腺癌治疗中起着至关重要的作用。虽然它的功效得到认可,PAL和细胞自噬之间的相互作用,特别是在RAF/MEK/ERK信号通路的背景下,仍然没有充分探索。本研究使用体外(MCF7和MDA-MB-468细胞)和体内(荷瘤裸鼠)模型研究了PAL对乳腺癌的抑制作用。旨在阐明PAL对自噬过程的影响,并探索将其与曲美替尼(TRA)联合使用的潜力,MEK抑制剂,我们的研究旨在解决PAL诱导的耐药性的挑战.我们的发现表明,PAL显着降低MCF7和MDA-MB-468细胞的活力,并减少小鼠的肿瘤大小,同时在MCF10A细胞中显示出最小的细胞毒性。然而,PAL还诱导保护性自噬,可能通过RAF/MEK/ERK途径激活导致耐药性。引入TRA有效地中和了这种自噬,增强PAL的抗肿瘤功效。PAL和TRA的组合协同降低了细胞活力和增殖,体内研究显示肿瘤大小明显缩小。总之,PAL和TRA组合成为克服PAL诱导抗性的有希望的策略,为乳腺癌治疗提供了新的视野。
    Breast cancer, a predominant global health issue, requires ongoing exploration of new therapeutic strategies. Palbociclib (PAL), a well-known cyclin-dependent kinase (CDK) inhibitor, plays a critical role in breast cancer treatment. While its efficacy is recognized, the interplay between PAL and cellular autophagy, particularly in the context of the RAF/MEK/ERK signaling pathway, remains insufficiently explored. This study investigates PAL\'s inhibitory effects on breast cancer using both in vitro (MCF7 and MDA-MB-468 cells) and in vivo (tumor-bearing nude mice) models. Aimed at elucidating the impact of PAL on autophagic processes and exploring the potential of combining it with trametinib (TRA), an MEK inhibitor, our research seeks to address the challenge of PAL-induced drug resistance. Our findings reveal that PAL significantly decreases the viability of MCF7 and MDA-MB-468 cells and reduces tumor size in mice while showing minimal cytotoxicity in MCF10A cells. However, PAL also induces protective autophagy, potentially leading to drug resistance via the RAF/MEK/ERK pathway activation. Introducing TRA effectively neutralized this autophagy, enhancing PAL\'s anti-tumor efficacy. A combination of PAL and TRA synergistically reduced cell viability and proliferation, and in vivo studies showed notable tumor size reduction. In conclusion, the PAL and TRA combination emerges as a promising strategy for overcoming PAL-induced resistance, offering a new horizon in breast cancer treatment.
    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
    本研究旨在探讨受体酪氨酸激酶样孤儿受体2(ROR2)在三阴性乳腺癌(TNBC)中的作用。
    通过免疫组织化学染色和PCR分析原发性TNBC和转移性TNBC组织中的ROR2表达。通过PCR和Western印迹分析检测TNBC细胞系中的ROR2表达。移民,检测ROR2过表达或敲低的TNBC细胞的侵袭和化学敏感性。
    ROR2在转移性TNBC组织中高表达。ROR2击倒抑制了迁移,TNBC细胞的侵袭和化学抗性。ROR2过表达促进MDA-MB-435细胞的迁移,入侵,和化学抗性。此外,HC1599和MDA-MB-435阿霉素耐药细胞中的ROR2敲低增强了对阿霉素的化学敏感性。ROR2可以激活TNBC细胞中的PI3K/AKT/mTOR信号。
    ROR2上调并通过激活PI3K/AKT/mTOR信号促进TNBC的转移表型。
    UNASSIGNED: This study aimed to investigate the role of receptor tyrosine kinase-like orphan receptor 2 (ROR2) in triple-negative breast cancer (TNBC).
    UNASSIGNED: ROR2 expression in primary TNBC and metastatic TNBC tissues was analyzed by immunohistochemical staining and PCR. ROR2 expression in TNBC cell lines was detected by PCR and Western blot analysis. The migration, invasion and chemosensitivity of TNBC cells with overexpression or knockdown of ROR2 were examined.
    UNASSIGNED: ROR2 expression was high in metastatic TNBC tissues. ROR2 knockdown suppressed the migration, invasion and chemoresistance of TNBC cells. ROR2 overexpression in MDA-MB-435 cells promoted the migration, invasion, and chemoresistance. Moreover, ROR2 knockdown in HC1599 and MDA-MB-435 adriamycin-resistant cells enhanced chemosensitivity to adriamycin. ROR2 could activate PI3K/AKT/mTOR signaling in TNBC cells.
    UNASSIGNED: ROR2 is upregulated and promotes metastatic phenotypes of TNBC by activating PI3K/AKT/mTOR signaling.
    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
    表皮生长因子受体(EGFR)的激活突变已被确定为非小细胞肺癌(NSCLC)中EGFR酪氨酸激酶抑制剂(TKIs)定制治疗的关键预测性生物标志物,帮助提高患者的反应率和生存率。然而,耐药性挑战了这些治疗的功效,对耐药后治疗策略的理解有限。深入了解EGFR突变NSCLC的生物学和耐药机制对于开发新的治疗方法至关重要。这项研究,通过文献计量分析,总结了EGFR-TKIs耐药的研究趋势。
    关于具有EGFR抑制剂抗性的NSCLC的研究论文来自WebofScienceCoreCollection(WoSCC)。该分析使用了诸如CiteSpace之类的文献计量工具,VOSviewer,和其他平台,用于全面分析和可视化结果。
    WoSCC数据库总共包含5866个关于EGFR-TKIs治疗耐药的文件,包括4727篇文章(93.48%)和1139篇评论(6.52%),跨越81个国家和地区,4792个机构,涉及23,594名作者。自2016年以来,该领域的出版物大幅增加。中国的出版物产量最高,而美国的论文引用次数最高。哈佛大学在出版物数量方面处于领先地位。在产量最高的十大期刊中,临床癌症研究的影响因子最高,为11.5,90%的期刊在Q1或Q2分类。拉斐尔·罗塞尔是这一领域最有影响力的作家之一,在出版量中排名第二,在引文计数中排名第四。EGFR-TKIs耐药的研究主要集中在基因检测,抵抗机制,和耐药后治疗策略。
    这项研究为研究人员寻找权威参考提供了可靠的依据和指导,了解研究趋势,探索潜在的方向。
    UNASSIGNED: Activating mutations in epidermal growth factor receptor (EGFR) have been identified as key predictive biomarkers for the customized treatment with EGFR tyrosine kinase inhibitors (TKIs) in non-small cell lung cancer (NSCLC), aiding in improving patient response rates and survival. However, resistance challenges the efficacy of these treatments, with limited understanding of post-resistance therapeutic strategies. A deep understanding of the biology and resistance mechanisms of EGFR-mutant NSCLC is crucial for developing new treatment approaches. This study, through bibliometric analysis, summarizes the trends in research on resistance to EGFR-TKIs.
    UNASSIGNED: Research papers on NSCLC with EGFR inhibitor resistance were collected from the Web of Science Core Collection (WoSCC). The analysis utilized bibliometric tools like CiteSpace, VOSviewer, and other platforms for comprehensive analysis and visualization of the outcomes.
    UNASSIGNED: The WoSCC database contains a total of 5866 documents on resistance to EGFR-TKIs treatment, including 4727 articles (93.48%) and 1139 reviews (6.52%), spanning 81 countries and regions, 4792 institutions, with the involvement of 23,594 authors. Since 2016, there has been a significant increase in publications in this field. China has the highest publication output, while the United States has the highest citation count for papers. Harvard University leads in terms of the number of publications. Among the top ten journals with the highest output, Clinical Cancer Research has the highest impact factor at 11.5, with 90% of the journals classified in Q1 or Q2. Rafael Rosell is one of the most influential authors in this field, ranking second in publication volume and fourth in citation count. Research on EGFR-TKIs resistance mainly focuses on genetic testing, resistance mechanisms, and post-resistance treatment strategies.
    UNASSIGNED: This study provides researchers with a reliable basis and guidance for finding authoritative references, understanding research trends, and exploring potential directions.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Published Erratum
    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
    目的:侵袭性乳腺癌(BC)细胞高表达RhoGTP酶激活蛋白29(ARHGAP29),RhoA的负调节剂。在诱导间质转化的乳腺癌细胞中,ARHGAP29是32种GTP酶激活酶中唯一的一种,其表达显着增加。因此,我们研究了ARHGAP29的表达与BC中肿瘤进展之间是否存在相关性。由于他莫昔芬耐药的BC细胞表现出增加的间充质特性和侵袭性,我们还研究了ARHGAP29与他莫昔芬耐药性中侵袭率增加之间的关系.问题是ARHGAP29是否是BC进展的合适预后标志物。
    方法:使用组织微阵列检测ARHGAP29在BC和邻近正常乳腺组织中的表达。使用siRNA进行击倒实验以研究ARHGAP29和可能的下游参与者RhoC和pAKT1对体外他莫昔芬抗性BC球体的侵袭性生长的影响。
    结果:与邻近的正常乳腺组织相比,在BC组织中ARHGAP29的表达经常增加。此外,有证据表明ARHGAP29高表达与晚期临床肿瘤分期之间存在相关性.与它们的亲本野生型细胞相比,他莫昔芬抗性BC细胞显示显著更高的ARHGAP29表达。在他莫昔芬抗性BC细胞中敲除ARHGAP29后,RhoC的表达显著降低。Further,pAKT1的表达明显下降。敲除ARHGAP29后,三维抗他莫昔芬BC球体的侵袭性生长减少。这可以被AKT1激活剂SC79部分逆转。
    结论:ARHGAP29的表达与BC患者的临床肿瘤参数相关。此外,ARHGAP29与他莫昔芬抗性BC细胞的侵袭性增加有关。ARHGAP29单独或与其下游伴侣RhoC和pAKT1组合可能是BC进展的合适预后标志物。
    OBJECTIVE: Aggressive breast cancer (BC) cells show high expression of Rho GTPase activating protein 29 (ARHGAP29), a negative regulator of RhoA. In breast cancer cells in which mesenchymal transformation was induced, ARHGAP29 was the only one of 32 GTPase-activating enzymes whose expression increased significantly. Therefore, we investigated whether there is a correlation between expression of ARHGAP29 and tumor progression in BC. Since tamoxifen-resistant BC cells exhibit increased mesenchymal properties and invasiveness, we additionally investigated the relationship between ARHGAP29 and increased invasion rate in tamoxifen resistance. The question arises as to whether ARHGAP29 is a suitable prognostic marker for the progression of BC.
    METHODS: Tissue microarrays were used to investigate expression of ARHGAP29 in BC and adjacent normal breast tissues. Knockdown experiments using siRNA were performed to investigate the influence of ARHGAP29 and the possible downstream actors RhoC and pAKT1 on invasive growth of tamoxifen-resistant BC spheroids in vitro.
    RESULTS: Expression of ARHGAP29 was frequently increased in BC tissues compared to adjacent normal breast tissues. In addition, there was evidence of a correlation between high ARHGAP29 expression and advanced clinical tumor stage. Tamoxifen-resistant BC cells show a significantly higher expression of ARHGAP29 compared to their parental wild-type cells. After knockdown of ARHGAP29 in tamoxifen-resistant BC cells, expression of RhoC was significantly reduced. Further, expression of pAKT1 decreased significantly. Invasive growth of three-dimensional tamoxifen-resistant BC spheroids was reduced after knockdown of ARHGAP29. This could be partially reversed by AKT1 activator SC79.
    CONCLUSIONS: Expression of ARHGAP29 correlates with the clinical tumor parameters of BC patients. In addition, ARHGAP29 is involved in increased invasiveness of tamoxifen-resistant BC cells. ARHGAP29 alone or in combination with its downstream partners RhoC and pAKT1 could be suitable prognostic markers for BC progression.
    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
    西妥昔单抗耐药一直是头颈部鳞状细胞癌(HNSCC)患者接受靶向治疗的主要挑战。然而,导致西妥昔单抗耐药的机制,特别是microRNA(miRNA)调控,尚不清楚。越来越多的证据表明,miRNA可能充当“核激活miRNA”,用于靶向与靶基因相关的启动子区域或增强子。这项研究阐明了HNSCC中西妥昔单抗抗性的新机制,涉及通过miR-451a对KDM7A转录的核激活。在这里,小RNA测序,定量实时聚合酶链反应(qRT-PCR)和荧光原位杂交(FISH)的结果提供了有说服力的证据,表明在西妥昔单抗治疗后miR-451a核富集.通过RNA纯化进行染色质分离,微阵列分析,和生物信息学分析显示,miR-451a与KDM7A中的增强子区相互作用,激活其表达并进一步促进西妥昔单抗抵抗。还已经证明,核miR-451a对KDM7A的激活是由西妥昔单抗治疗诱导的,并且是AGO2依赖性的。对87个HNSCC样品的Logistic回归分析表明miR-451a和KDM7A在西妥昔单抗抗性发展中的重要性。这些发现支持miR-451a和KDM7A作为西妥昔单抗抗性的有价值的生物标志物的潜力,并强调核激活miRNA的功能。
    Cetuximab resistance has been a major challenge for head and neck squamous cell carcinoma (HNSCC) patients receiving targeted therapy. However, the mechanism that causes cetuximab resistance, especially microRNA (miRNA) regulation, remains unclear. Growing evidence suggests that miRNAs may act as \"nuclear activating miRNAs\" for targeting promoter regions or enhancers related to target genes. This study elucidates a novel mechanism underlying cetuximab resistance in HNSCC involving the nuclear activation of KDM7A transcription via miR-451a. Herein, small RNA sequencing, quantitative real-time polymerase chain reaction (qRT‒PCR) and fluorescence in situ hybridization (FISH) results provided compelling evidence of miR-451a nuclear enrichment in response to cetuximab treatment. Chromatin isolation via RNA purification, microarray analysis, and bioinformatic analysis revealed that miR-451a interacts with an enhancer region in KDM7A, activating its expression and further facilitating cetuximab resistance. It has also been demonstrated that the activation of KDM7A by nuclear miR-451a is induced by cetuximab treatment and is AGO2 dependent. Logistic regression analyses of 87 HNSCC samples indicated the significance of miR-451a and KDM7A in the development of cetuximab resistance. These discoveries support the potential of miR-451a and KDM7A as valuable biomarkers for cetuximab resistance and emphasize the function of nuclear-activating miRNAs.
    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
    Ferroptosis,一种受调节的细胞死亡形式,与铁依赖性脂质过氧化密切相关。最近的证据强烈支持铁凋亡的诱导作为治疗对常规疗法有抗性的癌症的有希望的策略。铁凋亡调节的关键参与者是铁凋亡抑制蛋白1(FSP1),其通过促进辅酶Q10的抗氧化剂形式的产生来促进癌细胞抗性。值得注意的是,FSP1独立于谷胱甘肽(GSH)和谷胱甘肽过氧化物酶-4途径赋予铁死亡抗性。因此,靶向FSP1以削弱其对铁凋亡的抑制作用可能是治疗难治性癌症的可行策略。这篇综述旨在阐明铁死亡的分子机制,FSP1抑制铁凋亡的具体途径以及FSP1抑制剂对癌细胞的影响。
    Ferroptosis, a regulated form of cell death, is intricately linked to iron‑dependent lipid peroxidation. Recent evidence strongly supports the induction of ferroptosis as a promising strategy for treating cancers resistant to conventional therapies. A key player in ferroptosis regulation is ferroptosis suppressor protein 1 (FSP1), which promotes cancer cell resistance by promoting the production of the antioxidant form of coenzyme Q10. Of note, FSP1 confers resistance to ferroptosis independently of the glutathione (GSH) and glutathione peroxidase‑4 pathway. Therefore, targeting FSP1 to weaken its inhibition of ferroptosis may be a viable strategy for treating refractory cancer. This review aims to clarify the molecular mechanisms underlying ferroptosis, the specific pathway by which FSP1 suppresses ferroptosis and the effect of FSP1 inhibitors on cancer 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
    背景:L-茶氨酸,一种来自绿茶的非蛋白质氨基酸,被认为是抗癌候选人。然而,它在肿瘤化疗耐药发展中的作用尚不清楚,其分子机制亟待探索。
    方法:通过细胞计数试剂盒-8(CCK-8)试验验证L-茶氨酸对肺癌化疗耐药的影响,transwell分析,和体外肿瘤球体形成测定;通过聚合酶链反应(PCR)和蛋白质印迹法检测蛋白质的表达。使用RNA测序(RNA-seq)和生物信息学分析来鉴定L-茶氨酸诱导的差异表达基因。通过使用慢病毒介导的转染系统构建BMAL1敲低和过表达。
    结果:L-茶氨酸提高了对顺式二氨基二氯铂(DDP)的化学抗性,并抑制了DDP抗性肺癌细胞的干性,但不抑制非抗性肺癌细胞。RNA-seq分析的结果表明,STAT3/NOTCH1途径是参与L-茶氨酸改善DDP耐药肺癌化疗耐药的潜在显性信号。机械上,L-茶氨酸通过调控STAT3/NOTCH1/BMAL1信号传导诱导的干性标记物的表达以及抑制耐药相关基因的表达水平,阻碍DDP耐药肺癌细胞的迁移和干性激活。此外,L-茶氨酸和Stat3阻断联合协同改善DDP耐药肺癌的化疗耐药.
    结论:L-茶氨酸通过调节STAT3/NOTCH1/BMAL1信号通路改善化疗耐药,减少干性,抑制DDP耐药肺癌细胞的迁移。这一发现可能为克服癌症化学耐药性的治疗选择提供一些证据,包括肺癌.
    BACKGROUND: L-Theanine, a nonproteinogenic amino acid derived from green tea, is being recognized as an anti-cancer candidate. However, it\'s roles in the development of cancer chemoresistance is still unknown and the molecular mechanism is urgently to be explored.
    METHODS: The effects of L-Theanine on lung cancer chemoresistance were validated by Cell Counting Kit-8 (CCK-8) assay, transwell assay, and in vitro tumor spheroid formation assay; the expression of proteins was detected by using polymerase chain reaction (PCR) and western blotting. RNA-sequencing (RNA-seq) and bioinformatics analysis were used to identify differentially expressed genes induced by L-Theanine. BMAL1 knockdown and overexpression were constructed by using a lentivirus-mediated transfection system.
    RESULTS: L-Theanine improved the chemoresistance to cis-diamminedichloroplatinum (DDP) and inhibited stemness of DDP-resistant lung cancer cells but not non-resistant lung cancer cells. The results from RNA-seq analysis showed that STAT3/NOTCH1 pathway was a potential dominant signaling involved in L-Theanine improving the chemoresistance in DDP-resistant lung cancer. Mechanistically, L-Theanine impeded migration and stemness activation of DDP-resistant lung cancer cells via regulating the expression of STAT3/NOTCH1/BMAL1 signaling-induced stemness markers as well as inhibiting the expression levels of drug resistance-related genes. In addition, a combination of L-Theanine and Stat3 blockade synergistically improved the chemoresistance in DDP-resistant lung cancer.
    CONCLUSIONS: L-Theanine improves the chemoresistance by regulating STAT3/NOTCH1/BMAL1 signaling, reducing stemness, and inhibiting the migration of DDP-resistant lung cancer cells. The finding might provide some evidence for therapeutic options in overcoming the chemoresistance in cancers, including lung 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
    背景:肺癌是癌症相关死亡的主要原因,是所有恶性肿瘤中发病率和死亡率最高的肿瘤之一。据报道,DEPDC1B的表达失调发生在各种肿瘤类型中。然而,这种改变在肺腺癌(LUAD)中的功能意义和潜在的分子机制尚不清楚.在这项研究中,我们研究了DEPDC1B在LUAD中的作用和临床意义。
    方法:在几个公开可用的数据集中系统评估了DEPDC1B在LUAD中的表达及其与预后的关系。DEPDC1B敲低对LUAD细胞增殖和运动的影响使用JULI阶段实时细胞历史记录,同时通过流式细胞术研究敲低对细胞周期的影响。此外,进行RNA测序(RNA-Seq)分析以鉴定由DEPDC1B调节的下游靶基因和途径。DEPDC1B的表达与免疫细胞浸润的相关性,免疫疗法抗性,还检查了化学抗性。此外,采用分子生物学方法探讨B-Myb对DEPDC1B表达的调控机制。
    结果:发现DEPDC1B在LUAD患者中上调,这与不良临床结局相关。敲除DEPDC1B抑制细胞生长,迁移和运动性,以及细胞周期进程。敲除还导致几个下游基因的下调,包括NID1、FN1和EGFR,以及多个关键途径的失活,如ERK和PI3K-AKT途径。对LUAD中肿瘤免疫环境的分析表明,高DEPDC1B表达与大量活化的CD4记忆T细胞有关,M0巨噬细胞,M1巨噬细胞,和CD8+T细胞。此外,这些肿瘤对免疫疗法的反应较差.化疗药物敏感性分析显示,DEPDC1B高表达的LUADs对长春瑞滨等一线化疗药物反应更敏感,顺铂,和依托泊苷。此外,机制研究表明,DEPDC1B是B-Myb的直接靶基因,并且其敲除减弱了B-Myb的增殖和运动作用。
    结论:总之,我们的研究结果表明,DEPDC1B是LUAD恶性进展过程中的关键调节因子.因此,DEPDC1B可能是LUAD诊断和治疗中一个有前途的预后标志物和治疗靶标。
    BACKGROUND: Lung cancer is the primary cause of cancer-related deaths, with one of the highest incidence and mortality rates of all malignant tumors. Dysregulated expression of DEPDC1B has been reported to occur in various tumor types. However, the functional implications of this alteration in lung adenocarcinoma (LUAD) and the underlying molecular mechanism remains unclear. In this study, we investigated the role and clinical significance of DEPDC1B in LUAD.
    METHODS: The expression of DEPDC1B in LUAD and its relationship with prognosis were systematically evaluated in several publically available datasets. The effects of DEPDC1B knockdown on the proliferation and motility of LUAD cells were assessed using the JULI Stage Real-time Cell History Recorder, while the effect of knockdown on the cell cycle was studied by flow cytometry. Furthermore, RNA-Sequencing (RNA-Seq) analysis was conducted to identify the downstream target genes and pathways regulated by DEPDC1B. Correlations between the expression of DEPDC1B and immune cell infiltration, immunotherapy resistance, and chemoresistance were also examined. Additionally, molecular biological methods were used to explore the regulatory mechanism of B-Myb on DEPDC1B expression.
    RESULTS: DEPDC1B was found to be upregulated in LUAD patients and this was associated with poor clinical outcomes. Knockdown of DEPDC1B inhibited cell growth, migration and motility, as well as cell cycle progression. Knockdown also resulted in the down-regulation of several downstream genes, including NID1, FN1, and EGFR, as well as the inactivation of multiple critical pathways, such as the ERK and PI3K-AKT pathways. Analysis of the tumor immuno-environment in LUAD revealed that high DEPDC1B expression was associated with an abundance of activated CD4+ memory T cells, M0 macrophages, M1 macrophages, and CD8+ T cells. Moreover, these tumors responded poorly to immunotherapy. Analysis of chemo-drug sensitivity showed that LUADs with high DEPDC1B expression were more responsive to frontline chemotherapeutic drugs such as Vinorelbine, Cisplatin, and Etoposide. Additionally, mechanistic investigations revealed that DEPDC1B is a direct target gene of B-Myb, and that its knockdown attenuated the proliferation and motility effects of B-Myb.
    CONCLUSIONS: In summary, our findings indicate that DEPDC1B is a critical regulator during the malignant progression of LUAD. DEPDC1B could therefore be a promising prognostic marker and therapeutic target in LUAD diagnosis and treatment.
    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