Chemotherapy resistance

化疗耐药
  • 文章类型: Journal Article
    目的:系统治疗在早期乳腺癌(eBC)患者的治疗中起着重要作用。然而,需要个性化的治疗概念,以避免潜在的有害过度治疗。生物标志物是个体化治疗的关键。Notch信号通路被广泛认为是eBC中合适的预后或预测标志物。这项研究主要旨在评估NOTCH1mRNA表达水平与乳腺癌肿瘤组织病理学特征之间的关系。以及相应的eBC患者的临床特征。作为次要目标,我们通过评估5年观察后NOTCH1mRNA表达与无复发间期(RFI)和总生存期之间可能的关联,研究了NOTCH1的预后和预测价值.
    方法:在414个肿瘤样本中测定相对NOTCH1mRNA表达,使用定量PCR,多中心队列(常规应用中的预后评估(PiA),2009-2011年,NCT01592825)1,270例女性eBC患者。
    结果:在三分之一的肿瘤中检测到高NOTCH1mRNA表达,并与激素受体阴性状态和高uPA/PAI-1状态相关。此外,发现高NOTCH1mRNA表达与更多的RFI相关事件相关(校正风险比2.1,95%CI1.077-4.118).接受辅助化疗并在肿瘤中具有高NOTCH1mRNA表达的患者(n=86)发生RFI事件的可能性增加了三倍(调整后的风险比3.1,95%CI1.321-7.245,p=0.009)。
    结论:在这个队列中,NOTCH1mRNA表达具有预后和预测影响。具有高NOTCH1mRNA表达的肿瘤可能对细胞毒性治疗较不敏感,并且Notch信号传导途径的下调(例如通过γ-分泌酶抑制剂)对于作为个体化治疗选择的eBC治疗可能是有价值的。
    OBJECTIVE: Systemic therapy plays a major part in the cure of patients with early breast cancer (eBC). However, personalized treatment concepts are required to avoid potentially harmful overtreatment. Biomarkers are pivotal for individualized therapy. The Notch signalling pathway is widely considered as a suitable prognostic or predictive marker in eBC. This study aimed primarily at assessing the relationship between NOTCH1 mRNA expression levels and histopathological features of breast cancer tumors, as well as clinical characteristics of the correspondent eBC patients. As a secondary aim, we investigated the prognostic and predictive value of NOTCH1 by assessing possible associations between NOTCH1 mRNA expression and recurrence-free interval (RFI) and overall survival after five years of observation.
    METHODS: The relative NOTCH1 mRNA expression was determined in 414 tumour samples, using quantitative PCR in a prospective, multicenter cohort (Prognostic Assessment in Routine Application (PiA), 2009-2011, NCT01592825) of 1,270 female eBC patients.
    RESULTS: High NOTCH1 mRNA expression was detected in one-third of the tumours and was associated with negative hormone receptor status and high uPA/PAI-1 status. In addition, high NOTCH1 mRNA expression was found to be associated with more RFI related events (adjusted hazard ratio 2.1, 95% CI 1.077-4.118). Patients who received adjuvant chemotherapy and had high NOTCH1 mRNA expression in the tumour (n = 86) were three times more likely to have an RFI event (adjusted hazard ratio 3.1, 95% CI 1.321-7.245, p = 0.009).
    CONCLUSIONS: In this cohort, NOTCH1 mRNA expression had a prognostic and predictive impact. Tumours with high NOTCH1 mRNA expression may be less sensitive to cytotoxic treatment and downregulation of the Notch signalling pathway (e.g. by γ-secretase inhibitors) may be valuable for eBC therapy as an individualised treatment option.
    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:由于治疗选择有限和预后不良,食管癌面临重大挑战,特别是在高级阶段。失调的长链非编码RNA(lncRNA)与癌症进展和治疗抗性有关。这项研究调查了失调的lncRNANONHSAT227443.1的作用,通过lncRNA-seq,及其下游靶基因MRTFB在食管鳞状细胞癌(ESCC)中的表达。
    方法:通过lncRNA-seq在具有不同化疗反应的食管癌组织中鉴定出失调的lncRNAs。使用定量实时聚合酶链反应(qRT-PCR)和蛋白质印迹评估过表达的NONHSAT227443.1的调节相互作用。功能测定,包括细胞活力,细胞增殖,和流式细胞术分析,进行了全面研究NONHSAT227443.1及其下游分子对ESCC的影响。
    结果:NONHSAT227443.1在紫杉醇加铂类化疗无反应者和食管癌细胞系中显著过表达。化疗暴露导致NONHSAT227443.1表达减少。NONHSAT227443.1负调控MRTFB表达,它们的联合失调与癌症活动增加相关,扩散,抑制细胞凋亡。MRTFB表达降低与PI3K/AKT通路激活相关。
    结论:我们的研究提供了对NONHSAT227443.1和MRTFB在食管癌中作用的见解,有助于攻击性特征和治疗抗性。NONHSAT227443.1和MRTFB可以作为潜在的治疗靶标,以增强无应答病例对紫杉醇加铂化疗的反应。
    BACKGROUND: Esophageal cancer presents significant challenges due to limited treatment options and poor prognosis, particularly in advanced stages. Dysregulated long non-coding RNAs (lncRNAs) are implicated in cancer progression and treatment resistance. This study investigated the roles of dysregulated lncRNA NONHSAT227443.1, identified through lncRNA-seq, and its downstream target gene MRTFB in esophageal squamous cell carcinoma (ESCC).
    METHODS: Dysregulated lncRNAs were identified through lncRNA-seq in esophageal cancer tissues with varying chemotherapy response. The regulatory interaction of overexpressed NONHSAT227443.1 was assessed using quantitative real-time polymerase chain reaction (qRT-PCR) and western blotting. Functional assays, including cell viability, cell proliferation, and flow cytometry analyses, were performed to comprehensively investigate the influence of NONHSAT227443.1 and its downstream molecules on ESCC.
    RESULTS: NONHSAT227443.1 was significantly overexpressed in paclitaxel plus platinum chemotherapy non-responders and esophageal cancer cell lines. Chemotherapy exposure led to diminished NONHSAT227443.1 expression. NONHSAT227443.1 negatively regulated MRTFB expression, and their combined dysregulation correlated with increased cancer activity, proliferation, and suppressed apoptosis. Diminished MRTFB expression was associated with PI3K/AKT pathway activation.
    CONCLUSIONS: Our study provides insights into NONHSAT227443.1 and MRTFB roles in esophageal cancer, contributing to aggressive traits and treatment resistance. NONHSAT227443.1 and MRTFB may serve as potential therapeutic targets to enhance the response to paclitaxel plus platinum chemotherapy in non-responsive cases.
    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
    ATR-Chk1通路在细胞对DNA损伤和复制应激的反应中是必不可少的。而长链非编码RNA(lncRNAs)在调节该途径中的作用仍然未知。在这项研究中,我们鉴定了ATR和Chk1相互作用的lncRNA(ACIL,也称为LRRC75A-AS1或SNHG29),在DNA损伤时,ATR会促进Chk1的磷酸化。高ACIL水平与对DNA损伤剂的化学抗性和乳腺癌患者的不良预后相关。ACIL敲除在体外和体内使乳腺癌细胞对DNA损伤药物敏感。ACIL通过诱导细胞周期停滞保护癌细胞免受DNA损伤,稳定复制叉并抑制计划外的源发射,从而防范复制灾难并有助于DNA损伤修复。这些发现证明了lncRNA依赖性的激活ATR-Chk1通路的机制,并强调了利用ACIL作为化疗敏感性的预测生物标志物的潜力。以及靶向ACIL逆转乳腺癌化疗耐药。
    The ATR-Chk1 pathway is essential in cellular responses to DNA damage and replication stress, whereas the role of long noncoding RNAs (lncRNAs) in regulating this pathway remains largely unknown. In this study, we identify an ATR and Chk1 interacting lncRNA (ACIL, also known as LRRC75A-AS1 or SNHG29), which promotes the phosphorylation of Chk1 by ATR upon DNA damages. High ACIL levels are associated with chemoresistance to DNA damaging agents and poor outcome of breast cancer patients. ACIL knockdown sensitizes breast cancer cells to DNA damaging drugs in vitro and in vivo. ACIL protects cancer cells against DNA damages by inducing cell cycle arrest, stabilizing replication forks and inhibiting unscheduled origin firing, thereby guarding against replication catastrophe and contributing to DNA damage repair. These findings demonstrate a lncRNA-dependent mechanism of activating the ATR-Chk1 pathway and highlight the potential of utilizing ACIL as a predictive biomarker for chemotherapy sensitivity, as well as targeting ACIL to reverse chemoresistance 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
    黑色素瘤的耐药性是癌症治疗的主要障碍。生长激素(GH)在化疗耐药中起关键作用。已经证明敲除或阻断GH受体会使肿瘤细胞对化疗敏感。广泛的研究表明,外泌体,细胞外囊泡的一个子集,通过转移关键因素使癌细胞对化疗敏感,在耐药中起重要作用。在这项研究中,我们探讨GH如何调节黑色素瘤细胞的外泌体及其在耐药性中的作用。我们用GH治疗黑色素瘤细胞,阿霉素,和GHR拮抗剂,pegvisomant,并分析了释放的外泌体。此外,我们将这些外泌体给予受体细胞。GH处理的黑色素瘤细胞释放外泌体,ABC转运蛋白(ABCC1和ABCB1)水平升高,N-钙黏着蛋白,和MMP2,增强了受体细胞的耐药性和迁移。GHR拮抗作用降低了这些外泌体水平,恢复药物敏感性和减少迁移。总的来说,我们的研究结果强调了GH在调节外泌体载体中的一个新的作用,这些载体驱动黑色素瘤的化学耐药和转移.这种理解提供了对GH在黑色素瘤化学抗性中的机制的见解,并表明GHR拮抗作用是克服黑色素瘤治疗中的化学抗性的潜在疗法。
    Drug resistance in melanoma is a major hindrance in cancer therapy. Growth hormone (GH) plays a pivotal role in contributing to the resistance to chemotherapy. Knocking down or blocking the GH receptor has been shown to sensitize the tumor cells to chemotherapy. Extensive studies have demonstrated that exosomes, a subset of extracellular vesicles, play an important role in drug resistance by transferring key factors to sensitize cancer cells to chemotherapy. In this study, we explore how GH modulates exosomal cargoes from melanoma cells and their role in drug resistance. We treated the melanoma cells with GH, doxorubicin, and the GHR antagonist, pegvisomant, and analyzed the exosomes released. Additionally, we administered these exosomes to the recipient cells. The GH-treated melanoma cells released exosomes with elevated levels of ABC transporters (ABCC1 and ABCB1), N-cadherin, and MMP2, enhancing drug resistance and migration in the recipient cells. GHR antagonism reduced these exosomal levels, restoring drug sensitivity and attenuating migration. Overall, our findings highlight a novel role of GH in modulating exosomal cargoes that drive chemoresistance and metastasis in melanoma. This understanding provides insights into the mechanisms of GH in melanoma chemoresistance and suggests GHR antagonism as a potential therapy to overcome chemoresistance in melanoma 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
    背景:随着对神经胶质瘤发展和免疫微环境的逐步认识,已经发现了许多免疫细胞。尽管人们对免疫细胞的功能和免疫疗法的临床应用越来越了解,免疫细胞亚型的确切作用和特征,神经胶质瘤如何诱导免疫细胞的亚型转化及其对神经胶质瘤进展的影响尚待了解。
    目的:在这篇综述中,我们全面集中于神经胶质瘤微环境中的四大免疫细胞,特别是中性粒细胞,巨噬细胞,淋巴细胞,骨髓来源的抑制细胞(MDSCs),和其他重要的免疫细胞。我们讨论了(1)免疫细胞亚型标志物,(2)胶质瘤诱导的免疫细胞亚型转化,(3)各亚型影响化疗耐药的机制,(4)针对免疫细胞的疗法,和(5)免疫细胞相关单细胞测序。最终,我们确定了神经胶质瘤中免疫细胞亚型的特征,全面总结了胶质瘤诱导免疫细胞亚型转化的确切机制,并总结了单细胞测序在探索胶质瘤免疫细胞亚型方面的进展。
    总而言之,我们详细分析了化疗耐药的机制,并发现了未来的免疫治疗靶点,挖掘协同结合放射治疗的新型免疫治疗方法的潜力,化疗,和手术,从而为改善针对神经胶质瘤的免疫治疗策略和增强患者预后铺平了道路。
    BACKGROUND: With the gradual understanding of glioma development and the immune microenvironment, many immune cells have been discovered. Despite the growing comprehension of immune cell functions and the clinical application of immunotherapy, the precise roles and characteristics of immune cell subtypes, how glioma induces subtype transformation of immune cells and its impact on glioma progression have yet to be understood.
    OBJECTIVE: In this review, we comprehensively center on the four major immune cells within the glioma microenvironment, particularly neutrophils, macrophages, lymphocytes, myeloid-derived suppressor cells (MDSCs), and other significant immune cells. We discuss (1) immune cell subtype markers, (2) glioma-induced immune cell subtype transformation, (3) the mechanisms of each subtype influencing chemotherapy resistance, (4) therapies targeting immune cells, and (5) immune cell-associated single-cell sequencing. Eventually, we identified the characteristics of immune cell subtypes in glioma, comprehensively summarized the exact mechanism of glioma-induced immune cell subtype transformation, and concluded the progress of single-cell sequencing in exploring immune cell subtypes in glioma.
    UNASSIGNED: In conclusion, we have analyzed the mechanism of chemotherapy resistance detailly, and have discovered prospective immunotherapy targets, excavating the potential of novel immunotherapies approach that synergistically combines radiotherapy, chemotherapy, and surgery, thereby paving the way for improved immunotherapeutic strategies against glioma and enhanced patient outcomes.
    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
    粘着斑激酶(FAK;由PTK2编码)是30多年前发现的一种位于细胞粘附位点的细胞质蛋白酪氨酸激酶。它被整合素受体与细胞外基质蛋白结合激活。FAK广泛表达,并在粘附和细胞质中作为多种蛋白质的信号传导支架发挥作用。和细胞核中的转录因子。FAK表达和内在活性对小鼠发育至关重要,与细胞运动有分子联系,细胞存活和基因表达。值得注意的是,FAK酪氨酸磷酸化升高在肿瘤中很常见,包括胰腺癌和卵巢癌,它与生存率下降有关。小分子和口服FAK抑制剂在肿瘤和基质细胞中显示出靶向抑制,对化疗耐药有影响,间质纤维化与肿瘤微环境免疫功能。在这里,我们讨论了关于FAK激活和信号传导机制的最新见解,它作为细胞质和核支架的作用,以及FAK抑制剂的肿瘤内在和外在效应。我们还讨论了针对低级别和高级别浆液性卵巢癌的FAK的正在进行和先进的临床试验的结果。FAK是耐药性的主要调节者。虽然FAK不知道是突变激活的,预防FAK活性已经揭示了多种肿瘤弱点,这些弱点支持扩大的临床组合靶向可能性。
    Focal adhesion kinase (FAK; encoded by PTK2) was discovered over 30 years ago as a cytoplasmic protein tyrosine kinase that is localized to cell adhesion sites, where it is activated by integrin receptor binding to extracellular matrix proteins. FAK is ubiquitously expressed and functions as a signaling scaffold for a variety of proteins at adhesions and in the cell cytoplasm, and with transcription factors in the nucleus. FAK expression and intrinsic activity are essential for mouse development, with molecular connections to cell motility, cell survival and gene expression. Notably, elevated FAK tyrosine phosphorylation is common in tumors, including pancreatic and ovarian cancers, where it is associated with decreased survival. Small molecule and orally available FAK inhibitors show on-target inhibition in tumor and stromal cells with effects on chemotherapy resistance, stromal fibrosis and tumor microenvironment immune function. Herein, we discuss recent insights regarding mechanisms of FAK activation and signaling, its roles as a cytoplasmic and nuclear scaffold, and the tumor-intrinsic and -extrinsic effects of FAK inhibitors. We also discuss results from ongoing and advanced clinical trials targeting FAK in low- and high-grade serous ovarian cancers, where FAK acts as a master regulator of drug resistance. Although FAK is not known to be mutationally activated, preventing FAK activity has revealed multiple tumor vulnerabilities that support expanding clinical combinatorial targeting possibilities.
    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
    胶质母细胞瘤(GBM)是一种高度侵袭性的脑肿瘤,患者生存率低。目前的标准治疗包括侵入性手术,放射治疗,和使用替莫唑胺(TMZ)的化疗。对TMZ的抵抗力是,然而,一个重大挑战。我们小组先前的工作已经确定了与TMZ抗性相关的候选基因,包括编码跨病变合成(TLS)DNA聚合酶iota(Poltry)和κ(Polκ)的基因。已知这些专门的酶可以绕过病变并耐受DNA损伤。这里,我们研究了Poly和Polk在TMZ抗性中的作用,使用缺乏MGMT的U251-MG胶质母细胞瘤细胞,敲除POLI或POLK编码Poly和Polκ的基因,分别,并评估其在随后的TMZ治疗后的生存力和遗传毒性应激反应。与亲本对应物相比,缺乏这些聚合酶中的任一种的细胞在TMZ处理后表现出活力的显著降低。缺失的聚合酶的恢复导致细胞活力的恢复。此外,敲除细胞显示细胞周期停滞增加,主要在S期后期,TMZ治疗后基因毒性应激水平较低,通过γH2AX灶的减少和流式细胞术数据评估。这意味着在不存在这些蛋白质的情况下,TMZ处理不触发显著的H2AX磷酸化反应。有趣的是,将TMZ与Mirin(双链断裂修复途径抑制剂)结合使用,进一步降低了TLSKO细胞中的细胞活力并增加了DNA损伤和γH2AX阳性细胞,但不是在亲代细胞中。这些发现强调了Poltry和Polk在赋予TMZ抗性中的关键作用,以及在不存在这些TLS聚合酶的情况下同源重组的潜在备份作用。针对这些TLS酶,随着双链断裂DNA修复抑制,可以,因此,为提高TMZ治疗GBM的有效性提供了一个有前途的策略。
    Glioblastoma (GBM) is a highly aggressive brain tumor associated with poor patient survival. The current standard treatment involves invasive surgery, radiotherapy, and chemotherapy employing temozolomide (TMZ). Resistance to TMZ is, however, a major challenge. Previous work from our group has identified candidate genes linked to TMZ resistance, including genes encoding translesion synthesis (TLS) DNA polymerases iota (Polɩ) and kappa (Polκ). These specialized enzymes are known for bypassing lesions and tolerating DNA damage. Here, we investigated the roles of Polɩ and Polκ in TMZ resistance, employing MGMT-deficient U251-MG glioblastoma cells, with knockout of either POLI or POLK genes encoding Polɩ and Polκ, respectively, and assess their viability and genotoxic stress responses upon subsequent TMZ treatment. Cells lacking either of these polymerases exhibited a significant decrease in viability following TMZ treatment compared to parental counterparts. The restoration of the missing polymerase led to a recovery of cell viability. Furthermore, knockout cells displayed increased cell cycle arrest, mainly in late S-phase, and lower levels of genotoxic stress after TMZ treatment, as assessed by a reduction of γH2AX foci and flow cytometry data. This implies that TMZ treatment does not trigger a significant H2AX phosphorylation response in the absence of these proteins. Interestingly, combining TMZ with Mirin (double-strand break repair pathway inhibitor) further reduced the cell viability and increased DNA damage and γH2AX positive cells in TLS KO cells, but not in parental cells. These findings underscore the crucial roles of Polɩ and Polκ in conferring TMZ resistance and the potential backup role of homologous recombination in the absence of these TLS polymerases. Targeting these TLS enzymes, along with double-strand break DNA repair inhibition, could, therefore, provide a promising strategy to enhance TMZ\'s effectiveness in treating GBM.
    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
    化疗耐药在结直肠癌(CRC)患者的预后和治疗失败中起着关键作用。顺铂(DDP)抗性细胞表现出固有的逃避毒性化疗药物作用的能力,其特征在于慢循环程序的激活和DNA修复。在导致DDP电阻的元素中,O6-甲基鸟嘌呤(O6-MG)-DNA-甲基转移酶(MGMT),一种DNA修复酶,扮演一个典型的角色。在这项研究中,我们澄清了MGMT在赋予CRCDDP抗性中的重要参与,阐明MGMT监管行为的潜在机制。在我们的研究中发现了对DDP耐药的癌细胞中MGMT的显着上调,和MGMT抑制增强了这些细胞在体外和体内对DDP处理的敏感性。相反,在癌细胞中,MGMT过表达消除了它们对DDP治疗的敏感性。机械上,MGMT与细胞周期蛋白依赖性激酶1(CDK1)诱导慢循环细胞之间的相互作用是通过促进CDK1的泛素化降解来实现的。同时,为了实现非同源末端连接,MGMT与XRCC6相互作用以抵抗化疗药物。我们来自88例CRC患者的转录组数据表明MGMT表达与Wnt信号通路激活相关,几种Wnt抑制剂可以抑制耐药细胞。总之,我们的研究结果指出MGMT是一个潜在的CRC治疗靶点和化疗耐药的预测指标.
    Chemotherapy resistance plays a pivotal role in the prognosis and therapeutic failure of patients with colorectal cancer (CRC). Cisplatin (DDP)-resistant cells exhibit an inherent ability to evade the toxic chemotherapeutic drug effects which are characterized by the activation of slow-cycle programs and DNA repair. Among the elements that lead to DDP resistance, O 6-methylguanine (O 6-MG)-DNA-methyltransferase (MGMT), a DNA-repair enzyme, performs a quintessential role. In this study, we clarify the significant involvement of MGMT in conferring DDP resistance in CRC, elucidating the underlying mechanism of the regulatory actions of MGMT. A notable upregulation of MGMT in DDP-resistant cancer cells was found in our study, and MGMT repression amplifies the sensitivity of these cells to DDP treatment in vitro and in vivo. Conversely, in cancer cells, MGMT overexpression abolishes their sensitivity to DDP treatment. Mechanistically, the interaction between MGMT and cyclin dependent kinase 1 (CDK1) inducing slow-cycling cells is attainted via the promotion of ubiquitination degradation of CDK1. Meanwhile, to achieve nonhomologous end joining, MGMT interacts with XRCC6 to resist chemotherapy drugs. Our transcriptome data from samples of 88 patients with CRC suggest that MGMT expression is co-related with the Wnt signaling pathway activation, and several Wnt inhibitors can repress drug-resistant cells. In summary, our results point out that MGMT is a potential therapeutic target and predictive marker of chemoresistance in CRC.
    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
    结直肠癌是世界范围内最常见的恶性肿瘤之一。高发病率和死亡率使其成为研究的焦点。化疗是结肠癌的主要治疗方式,但化疗耐药严重影响治疗效果。已经发现MIF在各种癌症中促进肿瘤进展和抗性。本研究旨在探讨MIF在结肠癌化疗耐药中的作用及其可能机制。特别是通过CXCR7表达的上调,影响结肠癌细胞的代谢和药物敏感性。通过分析TCGA和HPA数据评估结肠癌组织中MIF的表达水平及其与患者预后的关系。随后,用qRT-PCR和免疫组织化学方法检测结肠癌细胞株和耐药细胞株中MIF的表达水平,并评估了MIF对奥沙利铂敏感性的影响。使用细胞能量代谢分析仪测量MIF对结肠癌细胞代谢活性的影响。进一步的实验探索了MIF通过上调CXCR7表达影响结肠癌细胞代谢活性的机制,通过沉默CTCF验证了CTCF在调节CXCR7转录中的作用。最后,在小鼠移植瘤模型中验证了MIF对结肠癌细胞药物敏感性的影响。在这项研究中,我们发现MIF在结肠癌组织中的表达明显高于正常组织,MIF高表达与患者预后不良有关。MIF在结肠癌耐药细胞株中的表达水平显著高于亲本细胞株,MIF过表达显著增加结肠癌细胞对奥沙利铂的耐药性。相反,沉默MIF可显著降低耐药细胞的IC50值,增加细胞凋亡。MIF过表达显著增加结肠癌细胞的ECAR和OCR水平,而MIF敲低显著降低了这些代谢指标。进一步的研究表明,MIF通过上调CXCR7的表达影响结肠癌细胞的代谢活性。CTCF在CXCR7启动子区的结合峰和荧光素酶活性测定表明CTCF调控CXCR7转录,沉默CTCF可显著增强结肠癌细胞对奥沙利铂的敏感性。小鼠体内实验表明,MIF沉默联合奥沙利铂治疗可显着抑制肿瘤生长并增加肿瘤组织的坏死面积。总之,这项研究揭示了MIF通过上调CXCR7表达在结肠癌化疗耐药中的关键作用,CTCF在这一过程中发挥着重要的调节作用。我们的发现为克服结肠癌化疗耐药提供了新的理论见解和潜在的治疗靶点。未来的研究应进一步探讨MIF和CXCR7在其他类型癌症中的作用以及MIF和CXCR7作为治疗靶标的潜力。
    Colorectal cancer is one of the most common malignant tumors worldwide, with high incidence and mortality rates making it a focus of research. Chemotherapy is a primary treatment modality for colon cancer, but chemotherapy resistance severely impacts treatment efficacy. MIF has been found to promote tumor progression and resistance in various cancers. This study aims to investigate the role of MIF in chemotherapy resistance in colon cancer and its potential mechanisms, particularly through the upregulation of CXCR7 expression, affecting the metabolism and drug sensitivity of colon cancer cells. The expression levels of MIF in colon cancer tissues and its association with patient prognosis were evaluated by analyzing TCGA and HPA data. Subsequently, the expression levels of MIF in colon cancer cell lines and resistant cell lines were detected by qRT-PCR and immunohistochemistry, and the effect of MIF on oxaliplatin sensitivity was assessed. The impact of MIF on the metabolic activity of colon cancer cells was measured using a cellular energy metabolism analyzer. Further experiments explored the mechanism by which MIF affects the metabolic activity of colon cancer cells through the upregulation of CXCR7 expression, and the role of CTCF in regulating CXCR7 transcription was validated by silencing CTCF. Finally, the effect of MIF on drug sensitivity of colon cancer cells was verified in a mouse xenograft tumor model. In this study, we found that the expression of MIF in colon cancer tissues was significantly higher than in normal tissues, and high MIF expression was associated with poor prognosis in patients. The expression levels of MIF in resistant colon cancer cell lines were significantly higher than in parental cell lines, and MIF overexpression significantly increased the resistance of colon cancer cells to oxaliplatin. Conversely, silencing MIF significantly reduced the IC50 value of resistant cells and increased apoptosis. MIF overexpression significantly increased the ECAR and OCR levels of colon cancer cells, while MIF knockdown significantly reduced these metabolic indicators. Further studies indicated that MIF affects the metabolic activity of colon cancer cells by upregulating CXCR7 expression. CTCF binding peaks at the CXCR7 promoter region and luciferase activity assays indicated that CTCF regulates CXCR7 transcription, and silencing CTCF significantly enhanced the sensitivity of colon cancer cells to oxaliplatin. In vivo experiments in mice showed that MIF silencing combined with oxaliplatin treatment significantly inhibited tumor growth and increased the necrotic area of tumor tissues. In conclusion, this study reveals the crucial role of MIF in chemotherapy resistance in colon cancer through the upregulation of CXCR7 expression, with CTCF playing an important regulatory role in this process. Our findings provide new theoretical insights and potential therapeutic targets for overcoming chemotherapy resistance in colon cancer. Future research should further explore the roles of MIF and CXCR7 in other types of cancers and the potential of MIF and CXCR7 as therapeutic targets.
    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
    暂无摘要。
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号