Cell Cycle Checkpoints

细胞周期检查点
  • 文章类型: Journal Article
    眼部黑色素瘤是成人常见的原发性恶性眼部肿瘤,有效治疗方法有限。表观遗传调控在肿瘤发生发展中起着重要作用。转换/蔗糖非发酵(SWI/SNF)染色质重塑复合物和溴结构域和外结构域家族蛋白是几种癌症的表观遗传调节因子。我们旨在筛选靶向这些调节剂的候选小分子抑制剂,并研究其在眼部黑色素瘤中的作用和机制。
    我们观察到了由相应基因敲低引起的表型以及与BRD抑制剂治疗和SWI/SNF复合物敲低的协同作用。流式细胞术分析JQ-1对眼黑色素瘤细胞周期和凋亡的影响。通过RNA测序,我们还探讨了BRD4的作用机制。
    观察到BRD4抑制剂(JQ-1)的最佳肿瘤抑制作用,尽管shBRD4和shBRD9组没有统计学上明显的变化。有趣的是,在shBRD4组中JQ-1的抑制作用降低。JQ-1抑制各种细胞系和荷瘤小鼠中黑色素瘤的生长。我们发现,在用JQ-1处理的MEL270和MEL290细胞后,这28个常见的差异表达基因中有17个下调。这17个基因中有4个,TP53I11,SH2D5,SEMA5A,MDGA1与BRD4呈正相关。在TCGA数据库中,低表达TP53I11,SH2D5,SEMA5A,MDGA1提高了患者的总体生存率。此外,TP53I11,SH2D5和SEMA5A低表达组的无病生存率提高.
    JQ-1可能作用于BRD4的下游,并通过诱导G1细胞周期停滞来抑制眼部黑色素瘤的生长。
    UNASSIGNED: Ocular melanoma is a common primary malignant ocular tumor in adults with limited effective treatments. Epigenetic regulation plays an important role in tumor development. The switching/sucrose nonfermentation (SWI/SNF) chromatin remodeling complex and bromodomain and extraterminal domain family proteins are epigenetic regulators involved in several cancers. We aimed to screen a candidate small molecule inhibitor targeting these regulators and investigate its effect and mechanism in ocular melanoma.
    UNASSIGNED: We observed phenotypes caused by knockdown of the corresponding gene and synergistic effects with BRD inhibitor treatment and SWI/SNF complex knockdown. The effect of JQ-1 on ocular melanoma cell cycle and apoptosis was analyzed with flow cytometry. Via RNA sequencing, we also explored the mechanism of BRD4.
    UNASSIGNED: The best tumor inhibitory effect was observed for the BRD4 inhibitor (JQ-1), although there were no statistically obvious changes in the shBRD4 and shBRD9 groups. Interestingly, the inhibitory effect of JQ-1 was decrease in the shBRD4 group. JQ-1 inhibits the growth of melanoma in various cell lines and in tumor-bearing mice. We found 17 of these 28 common differentially expressed genes were downregulated after MEL270 and MEL290 cells treated with JQ-1. Four of these 17 genes, TP53I11, SH2D5, SEMA5A, and MDGA1, were positively correlated with BRD4. In TCGA database, low expression of TP53I11, SH2D5, SEMA5A, and MDGA1 improved the overall survival rate of patients. Furthermore, the disease-free survival rate was increased in the groups with low expression of TP53I11, SH2D5, and SEMA5A.
    UNASSIGNED: JQ-1 may act downstream of BRD4 and suppress ocular melanoma growth by inducing G1 cell cycle 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
    多发性骨髓瘤(MM)患者经历复发和耐药;因此,新的治疗方法是必不可少的。克霉唑(CTZ)是一种具有抗肿瘤活性的广谱抗真菌药物。然而,CTZ对MM的影响尚不清楚。我们研究了CTZ对MM细胞增殖和凋亡诱导机制的影响。CTZ对MM.1S的影响,使用细胞计数试剂盒-8(CCK-8)测定研究NCI-H929、KMS-11和U266细胞生长。凋亡细胞百分比用膜联蛋白V-异硫氰酸荧光素/7-氨基放线菌素D染色定量。评估线粒体膜电位(MMP)和细胞周期进程。通过荧光显微镜测量活性氧(ROS)水平。使用蛋白质印迹分析凋亡相关和核因子(NF)-κB信号蛋白的表达。CCK-8测定表明,基于剂量和暴露时间,CTZ抑制细胞增殖。流式细胞术显示CTZ降低细胞凋亡和MMP并诱导G0/G1阻滞。免疫荧光表明,CTZ在总ROS和线粒体ROS产生中均呈剂量依赖性升高。Western印迹显示,CTZ增强Bax和裂解的聚ADP-核糖聚合酶和caspase-3,同时降低Bcl-2,p-p65和p-IκBα。因此,CTZ通过促进ROS介导的线粒体凋亡抑制MM细胞增殖,诱导G0/G1阻滞,抑制NF-κB通路,并具有治疗MM的潜力。
    Patients with multiple myeloma (MM) experience relapse and drug resistance; therefore, novel treatments are essential. Clotrimazole (CTZ) is a wide-spectrum antifungal drug with antitumor activity. However, CTZ\'s effects on MM are unclear. We investigated CTZ\'s effect on MM cell proliferation and apoptosis induction mechanisms. CTZ\'s effects on MM.1S, NCI- H929, KMS-11, and U266 cell growth were investigated using Cell Counting Kit-8 (CCK-8) assay. The apoptotic cell percentage was quantified with annexin V-fluorescein isothiocyanate/7-amino actinomycin D staining. Mitochondrial membrane potential (MMP) and cell cycle progression were evaluated. Reactive oxygen species (ROS) levels were measured via fluorescence microscopy. Expression of apoptosis-related and nuclear factor (NF)-κB signaling proteins was analyzed using western blotting. The CCK-8 assay indicated that CTZ inhibited cell proliferation based on both dose and exposure time. Flow cytometry revealed that CTZ decreased apoptosis and MMP and induced G0/G1 arrest. Immunofluorescence demonstrated that CTZ dose-dependently elevated in both total and mitochondrial ROS production. Western blotting showed that CTZ enhanced Bax and cleaved poly ADP-ribose polymerase and caspase-3 while decreasing Bcl-2, p-p65, and p-IκBα. Therefore, CTZ inhibits MM cell proliferation by promoting ROS-mediated mitochondrial apoptosis, inducing G0/G1 arrest, inhibiting the NF-κB pathway, and has the potential for treating MM.
    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
    癌症治疗已经从单一药物转移到更多基于机制的靶向方法。近年来,HDAC抑制剂和其他抗癌化学物质的组合在癌症治疗方面取得了令人兴奋的进展。在这里,我们通过将二氯乙酸盐与含硒的强效HDAC抑制剂连接,开发了一种新型前药。还研讨了该化合物在前列腺癌医治中的感化和机制。方法:在温和条件下设计并合成相关前药SeSA-DCA。这种化合物的临床前研究,包括药代动力学,细胞毒性,和对前列腺癌细胞系的抗肿瘤作用,被彻底调查,并探讨了其可能的协同作用机制。结果:SeSA-DCA在生理条件下表现出良好的稳定性,在肿瘤微环境中可迅速分解为DCA和SAHA的硒类似物(SeSAHA)。CCK-8实验表明,SeSA-DCA能有效抑制多种肿瘤细胞株的增殖,尤其是前列腺癌.在进一步的研究中,我们发现SeSA-DCA还可以抑制前列腺癌细胞系的转移并促进细胞凋亡。在动物层面,口服SeSA-DCA导致明显的肿瘤消退,无明显毒性。此外,作为双分子偶联化合物,SeSA-DCA在体外和体内均表现出比具有等摩尔SeSAHA和DCA的混合物优越得多的功效。本研究结果为前列腺癌的临床治疗提供了重要的理论依据。结论:我们的体内和体外结果表明,SeSA-DCA是一种高效的PCa抗肿瘤化合物。与单药治疗相比,它能有效诱导细胞周期阻滞和生长抑制,并抑制PCa细胞系的迁移和转移。SeSA-DCA降低异种移植物生长的能力比多西他赛稍好,没有任何明显的毒性迹象。本研究结果为前列腺癌的临床治疗提供了重要的理论依据。
    Cancer therapy has moved from single agents to more mechanism-based targeted approaches. In recent years, the combination of HDAC inhibitors and other anticancer chemicals has produced exciting progress in cancer treatment. Herein, we developed a novel prodrug via the ligation of dichloroacetate to selenium-containing potent HDAC inhibitors. The effect and mechanism of this compound in the treatment of prostate cancer were also studied. Methods: The concerned prodrug SeSA-DCA was designed and synthesized under mild conditions. This compound\'s preclinical studies, including the pharmacokinetics, cell toxicity, and anti-tumor effect on prostate cancer cell lines, were thoroughly investigated, and its possible synergistic mechanism was also explored and discussed. Results: SeSA-DCA showed good stability in physiological conditions and could be rapidly decomposed into DCA and selenium analog of SAHA (SeSAHA) in the tumor microenvironment. CCK-8 experiments identified that SeSA-DCA could effectively inhibit the proliferation of a variety of tumor cell lines, especially in prostate cancer. In further studies, we found that SeSA-DCA could also inhibit the metastasis of prostate cancer cell lines and promote cell apoptosis. At the animal level, oral administration of SeSA-DCA led to significant tumor regression without obvious toxicity. Moreover, as a bimolecular coupling compound, SeSA-DCA exhibited vastly superior efficacy than the mixture with equimolar SeSAHA and DCA both in vitro and in vivo. Our findings provide an important theoretical basis for clinical prostate cancer treatment. Conclusions: Our in vivo and in vitro results showed that SeSA-DCA is a highly effective anti-tumor compound for PCa. It can effectively induce cell cycle arrest and growth suppression and inhibit the migration and metastasis of PCa cell lines compared with monotherapy. SeSA-DCA\'s ability to decrease the growth of xenografts is a little better than that of docetaxel without any apparent signs of toxicity. Our findings provide an important theoretical basis for clinical prostate 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
    癌症临床化疗期间的多药耐药性(MDR)被认为是治疗效果的主要障碍。三磷酸腺苷结合盒(ABC)转运蛋白参与MDR机制显着降低了化学疗法的功效。这项研究调查了莫林的潜力,一种膳食生物类黄酮,克服KBChR-8-5MDR细胞中的秋水仙碱抗性。通过钙黄绿素-AM药物外排测定来测量桑林的P-gp抑制活性。蛋白质印迹分析用于评估在morin处理后的P-gp信使RNA和蛋白质表达。流式细胞术分析和吖啶橙/溴化乙锭荧光染色用于研究用桑色素和紫杉醇联合处理后对细胞凋亡和细胞周期停滞的诱导。此外,进行聚合酶链反应(PCR)阵列分析以研究与MDR相关的基因表达谱,用莫林治疗期间的细胞凋亡和细胞周期停滞,紫杉醇或其组合。Morin表现出与人P-gp的强结合相互作用。药物外排试验证实了这一点,其显示P-gp外排功能随着sorin浓度的增加而降低。此外,莫林和紫杉醇的组合增强了细胞凋亡的诱导和G2/M期细胞周期停滞。Morin处理显著下调MDR细胞ABCB1基因表达和P-gp膜表达。此外,PCR阵列基因表达分析表明,用桑色素和紫杉醇的组合处理上调了促凋亡和细胞周期停滞基因,同时下调了ABCB1基因和抗凋亡基因。因此,莫林可有效逆转KBChR-8-5耐药癌细胞中的紫杉醇耐药性,并得出结论,莫林可使KBChR8-5耐药癌细胞中的紫杉醇耐药性重新敏感。
    Multidrug resistance (MDR) during clinical chemotherapy for cancer has been considered a major obstacle to treatment efficacy. The involvement of adenosine triphosphate-binding cassette (ABC) transporters in the MDR mechanism significantly reduces the efficacy of chemotherapeutics. This study investigates the potential of morin, a dietary bioflavonoid, to overcome colchicine resistance in KBChR-8-5 MDR cells. The P-gp inhibitory activity by morin was measured by calcein-AM drug efflux assay. Western blot analysis was employed to evaluate P-gp messenger RNA and protein expressions following morin treatment. Flow cytometry analysis and acridine orange/ethidium bromide fluorescence staining were utilised to investigate the induction of apoptosis and cell cycle arrest upon treatment with morin and paclitaxel in combination. Additionally, polymerase chain reaction (PCR) array analysis was conducted to study the gene expression profiles related to MDR, apoptosis and cell cycle arrest during treatment with morin, paclitaxel or their combination. Morin exhibited a strong binding interaction with human P-gp. This was corroborated by drug efflux assays, which showed a reduction in P-gp efflux function with increasing morin concentration. Furthermore, morin and paclitaxel combination potentiated the induction of apoptosis and G2/M phase cell cycle arrest. Morin treatment significantly downregulated the gene expression of ABCB1 and P-gp membrane expressions in MDR cells. Additionally, PCR array gene expression analysis revealed that the combination treatment with morin and paclitaxel upregulated proapoptotic and cell cycle arrest genes while downregulating ABCB1 gene and antiapoptotic genes. Thus, morin effectively reversed paclitaxel resistance in KBChR-8-5 drug-resistant cancer cells and concluded that morin resensitized the paclitaxel resistance in KBChR8-5 drug-resistant 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
    孟鲁司特和扎鲁司特,半胱氨酰白三烯受体拮抗剂(LTRAs),引发三阴性乳腺癌MDA-MB-231细胞凋亡并抑制细胞增殖。相比之下,只有扎鲁司特诱导G0/G1细胞周期阻滞。本研究比较了这些药物对调节细胞增殖的蛋白质的作用,凋亡,自噬,使用逆转录定量PCR,内质网(ER)和氧化应激,蛋白质印迹和流式细胞术。增殖标志物的表达,Ki-67和增殖细胞核抗原,这两种药物都减少了。扎鲁克斯特,但不是孟鲁司特,细胞周期蛋白D1和CDK4的表达降低,从而中断从G1到S期的进展。扎鲁司特还增加了细胞周期抑制剂p27的表达。两种药物均降低了抗凋亡蛋白Bcl-2和ERK1/2磷酸化的表达,自噬标记LC3-II和DNA损伤标记的水平升高,包括裂解的PARP-1,磷酸化(p)-ATM和p-组蛋白H2AX。与扎鲁司特处理的细胞相比,孟鲁司特处理的细胞中caspase3/7阳性细胞的数量更多。与扎鲁司特相比,孟鲁司特诱导更高水平的ER应激标志物CHOP。孟鲁司特激活PERK,激活转录因子6(ATF6)和需要肌醇的酶1型(IRE1)途径,而扎鲁司特仅刺激ATF6和IRE1途径。GSK2606414,一种PERK抑制剂,孟鲁司特介导的细胞凋亡减少,但不影响扎鲁司特诱导的细胞死亡。小干扰RNA对CHOP的敲除减少了孟鲁司特和扎鲁司特引发的凋亡。总之,对细胞周期调节蛋白的影响可能有助于扎鲁司特引起的细胞周期停滞。孟鲁司特的更大的凋亡效应可能是由更高水平的激活的caspase酶和三个途径的内质网应激的激活引起的:PERK,ATF6和IRE1。
    Montelukast and zafirlukast, cysteinyl leukotriene receptor antagonists (LTRAs), trigger apoptosis and inhibit cell proliferation of triple‑negative breast cancer MDA‑MB‑231 cells. By contrast, only zafirlukast induces G0/G1 cell cycle arrest. The present study compared the effects of these drugs on proteins regulating cell proliferation, apoptosis, autophagy, and endoplasmic reticulum (ER) and oxidative stress using reverse transcription‑quantitative PCR, western blotting and flow cytometry. The expression of proliferating markers, Ki‑67 and proliferating cell nuclear antigen, was decreased by both drugs. Zafirlukast, but not montelukast, decreased the expression of cyclin D1 and CDK4, disrupting progression from G1 to S phase. Zafirlukast also increased the expression of p27, a cell cycle inhibitor. Both drugs decreased the expression of anti‑apoptotic protein Bcl‑2 and ERK1/2 phosphorylation, and increased levels of the autophagy marker LC3‑II and DNA damage markers, including cleaved PARP‑1, phosphorylated (p)‑ATM and p‑histone H2AX. The number of caspase 3/7‑positive cells was greater in montelukast‑treated cells compared with zafirlukast‑treated cells. Montelukast induced higher levels of the ER stress marker CHOP compared with zafirlukast. Montelukast activated PERK, activating transcription factor 6 (ATF6) and inositol‑requiring enzyme type 1 (IRE1) pathways, while zafirlukast only stimulated ATF6 and IRE1 pathways. GSK2606414, a PERK inhibitor, decreased apoptosis mediated by montelukast, but did not affect zafirlukast‑induced cell death. The knockdown of CHOP by small interfering RNA reduced apoptosis triggered by montelukast and zafirlukast. In conclusion, the effects on cell cycle regulator proteins may contribute to cell cycle arrest caused by zafirlukast. The greater apoptotic effects of montelukast may be caused by the higher levels of activated caspase enzymes and the activation of three pathways of ER stress: PERK, ATF6, and IRE1.
    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
    非小细胞肺癌(NSCLC)由于其低的五年生存率而提出了全球性的健康挑战。强调需要新的治疗策略。我们的研究探索了Syrosingopine和UK-5099治疗NSCLC的协同机制。体外实验表明,syrosingopine和UK-5099的组合显著协同抑制NSCLC增殖。进一步的实验表明,这种组合在NSCLC细胞中诱导细胞周期停滞并促进凋亡。体内实验证明,syrosingopine和UK-5099的组合显著抑制肿瘤生长。机制研究表明,这种药物组合通过诱导乳酸积累和氧化应激促进线粒体损伤。此外,该组合通过激活血红素调节抑制剂激酶(HRI)引发整合应激反应(ISR)。重要的是,我们的研究结果表明,舒辛平和UK-5099对NSCLC的协同抑制依赖于ISR激活.总之,我们的研究提出了一种有希望的治疗方法,包括联合使用Syrosingopine和UK-5099来激活ISR,显著阻碍NSCLC生长和增殖。
    Non-small cell lung cancer (NSCLC) presents a global health challenge due to its low five-year survival rates, underscoring the need for novel therapeutic strategies. Our research explored the synergistic mechanisms of syrosingopine and UK-5099 in treating NSCLC. In vitro experiments showed that the combination of syrosingopine and UK-5099 significantly synergized to suppress NSCLC proliferation. Further experiments revealed that this combination induced cell cycle arrest and promoted apoptosis in NSCLC cells. In vivo experiments demonstrated that the combination of syrosingopine and UK-5099 markedly inhibited tumor growth. Mechanistic studies revealed that this drug combination promoted mitochondrial damage by inducing lactate accumulation and oxidative stress. Additionally, the combination triggered an integrated stress response (ISR) through the activation of heme-regulated inhibitor kinase (HRI). Importantly, our findings suggested that the synergistic suppression of NSCLC by syrosingopine and UK-5099 was dependent on ISR activation. In summary, our study proposed a promising therapeutic approach that involved the combination of Syrosingopine and UK-5099 to activate ISR, significantly hindering NSCLC growth and proliferation.
    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
    为了克服人软骨肉瘤细胞对放射治疗的抵抗力,用DNA依赖性蛋白激酶催化亚基(DNA-PKcs)抑制剂AZD7648进行碳离子(C-离子)和参考光子(X射线)照射(IR)的联合治疗来预防有效的DNA修复基因表达分析,流式细胞术,蛋白质磷酸化,端粒长度缩短。联合治疗后细胞增殖标志物和细胞周期分布显著改变,揭示了一个突出的G2/M逮捕。G2/M检查点基因细胞周期蛋白B的表达,通过单独的IR和联合治疗,CDK1和WEE1显着降低。虽然单独的IR没有显示出影响,额外的AZD7648治疗导致AKT磷酸化的剂量依赖性减少和Chk2磷酸化的增加.IR后24小时,联合治疗减少了DNA修复机制的关键基因,导致DNA修复受损和放射敏感性增加。用AZD7648和8GyX射线/C-离子IR联合处理后,在两种细胞系中均观察到端粒长度的时间依赖性缩短。我们的数据表明,抑制DNA-PKcs可能会削弱其在DNA修复机制和端粒末端保护中的功能,从而增加对X射线和C离子IR的敏感性。
    In order to overcome the resistance to radiotherapy in human chondrosarcoma cells, the prevention from efficient DNA repair with a combined treatment with the DNA-dependent protein kinase catalytic subunit (DNA-PKcs) inhibitor AZD7648 was explored for carbon ion (C-ion) as well as reference photon (X-ray) irradiation (IR) using gene expression analysis, flow cytometry, protein phosphorylation, and telomere length shortening. Proliferation markers and cell cycle distribution changed significantly after combined treatment, revealing a prominent G2/M arrest. The expression of the G2/M checkpoint genes cyclin B, CDK1, and WEE1 was significantly reduced by IR alone and the combined treatment. While IR alone showed no effects, additional AZD7648 treatment resulted in a dose-dependent reduction in AKT phosphorylation and an increase in Chk2 phosphorylation. Twenty-four hours after IR, the key genes of DNA repair mechanisms were reduced by the combined treatment, which led to impaired DNA repair and increased radiosensitivity. A time-dependent shortening of telomere length was observed in both cell lines after combined treatment with AZD7648 and 8 Gy X-ray/C-ion IR. Our data suggest that the inhibition of DNA-PKcs may increase sensitivity to X-rays and C-ion IR by impairing its functional role in DNA repair mechanisms and telomere end protection.
    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
    最近,从最近的科学进步中获得的一种名为34的化合物已经成为这项研究的重点,其目的是探讨其对实体瘤细胞系的潜在影响。结合生物信息学和生物测定,这项研究对34的影响进行了深入调查。这项研究的结果对癌症研究和治疗具有重要意义。34在抑制几种癌细胞系的生长方面显示出显著的功效,包括代表前列腺癌(PC3)和宫颈癌(HeLa)的那些。这些细胞的高灵敏度,由低IC50值表示,强调了其作为有前途的化学治疗剂的潜力。此外,34已经揭示了诱导细胞周期停滞的能力,特别是在G2/M阶段,对肿瘤发生和生长具有重要意义的现象。通过干扰癌细胞中的DNA复制,34已经显示出触发细胞死亡的能力,为癌症治疗提供了新的途径.此外,计算分析已经确定了受34种治疗影响的关键基因,提示潜在的治疗靶点。这些基因参与了关键的生物过程,包括细胞周期调节,DNA复制和微管动力学,所有这些都是癌症发展和进展的核心。总之,这项研究强调了34抑制癌细胞生长和改变细胞周期的不同机制。这些有希望的结果表明了更有效和毒性更低的抗癌疗法的潜力。进一步的体内验证和联合疗法的探索对于改善癌症治疗结果至关重要。
    Recently, a compound derived from recent scientific advances named 34 has emerged as the focus of this research, the aim of which is to explore its potential impact on solid tumor cell lines. Using a combination of bioinformatics and biological assays, this study conducted an in-depth investigation of the effects of 34. The results of this study have substantial implications for cancer research and treatment. 34 has shown remarkable efficacy in inhibiting the growth of several cancer cell lines, including those representing prostate carcinoma (PC3) and cervical carcinoma (HeLa). The high sensitivity of these cells, indicated by low IC50 values, underscores its potential as a promising chemotherapeutic agent. In addition, 34 has revealed the ability to induce cell cycle arrest, particularly in the G2/M phase, a phenomenon with critical implications for tumor initiation and growth. By interfering with DNA replication in cancer cells, 34 has shown the capacity to trigger cell death, offering a new avenue for cancer treatment. In addition, computational analyses have identified key genes affected by 34 treatment, suggesting potential therapeutic targets. These genes are involved in critical biological processes, including cell cycle regulation, DNA replication and microtubule dynamics, all of which are central to cancer development and progression. In conclusion, this study highlights the different mechanisms of 34 that inhibit cancer cell growth and alter the cell cycle. These promising results suggest the potential for more effective and less toxic anticancer therapies. Further in vivo validation and exploration of combination therapies are critical to improve cancer treatment outcomes.
    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
    最近有几篇突破性文章证实了肿瘤细胞逃避治疗诱导衰老(TIS)施加的稳定细胞周期停滞的能力。随后,接受TIS是可避免的假设应该会鼓励对衰老在癌症治疗中的基本作用进行认真的重新评估.逃避TIS的潜力破坏了成熟的衰老的肿瘤抑制功能,提出它是导致疾病复发的肿瘤休眠机制,并邀请进一步研究其对癌症治疗结果的不利贡献。此外,逃避TIS强烈表明衰老肿瘤细胞的消除,主要通过药理学手段,是提高癌症治疗疗效的合适方法,一个仍然需要进一步探索的问题。这篇评论概述了最近的证据,这些证据明确证明了治疗诱导的衰老肿瘤细胞克服终末生长停滞命运的能力,并为TIS在肿瘤生物学中的作用提供了未来的观点。
    Several breakthrough articles have recently confirmed the ability of tumor cells to escape the stable cell cycle arrest imposed by Therapy-Induced Senescence (TIS). Subsequently, accepting the hypothesis that TIS is escapable should encourage serious reassessments of the fundamental roles of senescence in cancer treatment. The potential for escape from TIS undermines the well-established tumor suppressor function of senescence, proposes it as a mechanism of tumor dormancy leading to disease recurrence and invites for further investigation of its unfavorable contribution to cancer therapy outcomes. Moreover, escaping TIS strongly indicates that the elimination of senescent tumor cells, primarily through pharmacological means, is a suitable approach for increasing the efficacy of cancer treatment, one that still requires further exploration. This commentary provides an overview of the recent evidence that unequivocally demonstrated the ability of therapy-induced senescent tumor cells in overcoming the terminal growth arrest fate and provides future perspectives on the roles of TIS in tumor biology.
    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: Letter
    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

公众号