radioresistance

抗辐射性
  • 文章类型: Journal Article
    Radiation resistance is a crucial factor influencing therapeutic outcomes in colorectal cancer (CRC). Baicalein (BE), primarily derived from Scutellaria baicalensis, has demonstrated anti-CRC properties. However, the impact of BE on the radiosensitivity of CRC remains unclear. This study aimed to evaluate the radiosensitization effects of BE and elucidate its mechanism in CRC radiotherapy. We established an in vitro radioresistant cell model (CT26-R) using parental CRC cells (CT26) subjected to ionizing radiation (IR). CT26-R cells were pretreated with or without BE, followed by transfection with pcDNA-NC and pcDNA-JAK2. The proliferation of CT26-R cells treated with BE and IR was assessed using a colony formation assay. A CRC animal model was developed in BALB/c mice via CT26-R cell transplantation. The radiosensitizing effect of BE on CRC was evaluated in vivo. TUNEL assay was conducted to detect apoptosis in tumor tissue. The expression levels of p-STAT3, JAK2, PD-L1, and SOCS3 in vitro and in vivo were measured by western blotting. Our results demonstrated that BE significantly increased radiosensitivity in vitro and in vivo and enhanced apoptosis in tumor tissues. Additionally, BE significantly downregulated the expression of p-STAT3, JAK2, and PD-L1, and significantly upregulated SOCS3 expression. These in vivo effects were reversed by pcDNA-JAK2. In summary, our data suggest that BE enhances CRC radiosensitivity by inhibiting the JAK2/STAT3 pathway.
    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
    在全身治疗不足的情况下,可以使用放射疗法(RT)减轻症状并维持非小细胞肺癌患者的开放气道。对一些病人来说,由于辐射抗性,肿瘤控制无法实现。已提出同时抑制表皮生长因子受体作为克服辐射抗性的策略,但可能会增加毒性。我们进行了一项随机试验来评估疗效,容忍度,厄洛替尼和姑息性胸部RT对晚期非小细胞肺癌患者的生活质量。
    患者以1:1的比例随机分配至单纯放疗(A组)或与厄洛替尼联合治疗(B组)。使用基线时的计算机断层扫描(CT)扫描和纳入后4-12周的计算机断层扫描来评估治疗反应。在治疗期间和随后的30天记录不良事件。患者在基线时完成与健康相关的生活质量问卷,第2、6和20周。
    共纳入114例患者。对74例患者进行CT扫描以评价治疗效果,两组之间的肿瘤大小减小无显著差异:对照组A组的中位减小14.5%,埃罗替尼组B组的中位减小17.0%(p=0.68).两个治疗组之间的总生存期没有显着差异:A组和B组的7.0和7.8个月。分别(对数秩p=0.32)。实验组的不良事件没有明显增加,除了厄洛替尼单独治疗的预期之外。总的来说,两组患者的生活质量相似.
    对于晚期非小细胞肺癌患者,合并埃罗替尼和姑息性胸部放疗的耐受性良好,但并未改善放疗的疗效。
    ClinicalTrials.gov,标识符NCT02714530。
    UNASSIGNED: Radiotherapy (RT) can be used to reduce symptoms and maintain open airways for patients with non-small cell lung cancer when systemic treatment is not sufficient. For some patients, tumor control is not achieved due to radioresistance. Concurrent inhibition of epidermal growth factor receptors has been proposed as a strategy to overcome radioresistance but may increase toxicity. We performed a randomized trial to assess the efficacy, tolerance, and quality of life of concurrent erlotinib and palliative thoracic RT for patients with advanced non-small cell lung cancer.
    UNASSIGNED: Patients were randomized 1:1 to RT alone (arm A) or in combination with erlotinib (arm B). A computed tomography (CT) scan at baseline and one at 4-12 weeks after inclusion was used to evaluate treatment response. Adverse events were registered during treatment and the subsequent 30 days. Health-related quality-of-life questionnaires were completed by the patients at baseline, weeks 2, 6, and 20.
    UNASSIGNED: A total of 114 patients were included. Of the 74 patients with CT scans available for evaluation of treatment effect, there were no significant differences in tumor size reduction between the two groups: median 14.5% reduction in the control arm A and 17.0% in the erlotinib arm B (p = 0.68). Overall survival was not significantly different between the two treatment arms: 7.0 and 7.8 months in arm A and arm B, respectively (log-rank p = 0.32). There was no significant increase in adverse events in the experimental arm, other than what is expected from erlotinib treatment alone. Overall, patients reported similar quality of life in both treatment arms.
    UNASSIGNED: Concurrent erlotinib and palliative thoracic RT for patients with advanced non-small cell lung cancer was well tolerated but did not improve the efficacy of the RT.
    UNASSIGNED: ClinicalTrials.gov, identifier NCT02714530.
    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
    MicroRNAs(miRNAs)是细胞生理学和病理生理学中强大的转录后调节因子。在这次审查中,我们关注miR-223-3p(以下简称miR-223)在各种癌症类型中的作用。MiR-223在造血中已经确立了作用,炎症,大多数癌症,它可以作为致癌或抑癌的miRNA,取决于特定的分子景观。MiR-223也与癌细胞对治疗的敏感性或抗性相关。通过这次详细的审查,我们强调,对于某些癌症(即,乳房,非小细胞肺癌,和胶质母细胞瘤),miR-223的抑癌作用在文献中一直有报道,而对于其他人(即,结直肠,卵巢,和胰腺癌,和急性淋巴细胞白血病),致癌作用盛行。在前列腺癌和其他血液恶性肿瘤中,尽管经常描述肿瘤抑制作用,没有共识。有趣的是,当miRNA充当抑癌基因或癌基因时,NLRP3和FBXW7始终被鉴定为miR-223靶标,分别,不同的癌症我们的综述还描述了miR-223在大多数分析的癌症中的生物流体或其细胞外囊泡中增加。与健康或低风险条件相比,证实了该miRNA作为临床诊断和预后生物标志物的潜在应用。
    MicroRNAs (miRNAs) are mighty post-transcriptional regulators in cell physiology and pathophysiology. In this review, we focus on the role of miR-223-3p (henceforth miR-223) in various cancer types. MiR-223 has established roles in hematopoiesis, inflammation, and most cancers, where it can act as either an oncogenic or oncosuppressive miRNA, depending on specific molecular landscapes. MiR-223 has also been linked to either the sensitivity or resistance of cancer cells to treatments in a context-dependent way. Through this detailed review, we highlight that for some cancers (i.e., breast, non-small cell lung carcinoma, and glioblastoma), the oncosuppressive role of miR-223 is consistently reported in the literature, while for others (i.e., colorectal, ovarian, and pancreatic cancers, and acute lymphocytic leukemia), an oncogenic role prevails. In prostate cancer and other hematological malignancies, although an oncosuppressive role is frequently described, there is less of a consensus. Intriguingly, NLRP3 and FBXW7 are consistently identified as miR-223 targets when the miRNA acts as an oncosuppressor or an oncogene, respectively, in different cancers. Our review also describes that miR-223 was increased in biological fluids or their extracellular vesicles in most of the cancers analyzed, as compared to healthy or lower-risk conditions, confirming the potential application of this miRNA as a diagnostic and prognostic biomarker in the clinic.
    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:放射抗性和免疫逃逸是胶质母细胞瘤(GBM)治疗效果不理想的关键原因。尽管在骨髓细胞-2(TREM2)上表达的触发受体参与形成免疫抑制微环境,但其介导癌症放射抗性的潜在机制及其作用仍不清楚,此外,靶向TREM2的药物向GBM的有效递送面临严峻挑战.因此,本研究旨在阐明靶向TREM2沉默逆转谷胱甘肽反应性仿生纳米粒子(NP)平台辅助下GBM的放射抗性和免疫逃逸的作用和机制.
    方法:首先建立抗辐射GBM细胞系和TREM2稳定敲低GBM细胞系。RNA测序,集落形成试验,westernblot,采用酶联免疫吸附法和免疫共沉淀法检测TREM2调节GBM放射抗性和免疫逃逸的分子机制。谷胱甘肽响应的仿生NP,然后构建血管肽-2(A2)-细胞膜(CM)-NP/siTREM2/spam1,以三重并靶向抑制TREM2用于体内研究。建立原位GBM小鼠模型以评估TREM2抑制的抗GBM作用。采用多重免疫荧光法检测免疫细胞的浸润情况。
    结果:TREM2通过参与DNA损伤修复并与高迁移率族蛋白盒1(HMGB1)形成正反馈回路,级联激活Toll样受体4(TLR4)/蛋白激酶B(Akt)信号,从而加速GBM的放射抗性和免疫逃逸。成功合成了A2-CM-NP/siTREM2/spam1,具有良好的被动靶向性,主动靶向和同源靶向,通过促进1型辅助性T细胞和CD8+T细胞的浸润,减少2型辅助性T细胞和调节性T细胞的浸润,将巨噬细胞重新极化为M1型,并减少促肿瘤和免疫抑制细胞因子的分泌。
    结论:靶向TREM2治疗是优化放疗和免疫治疗以改善GBM患者预后的有希望的途径。
    BACKGROUND: Radioresistance and immune escape are crucial reasons for unsatisfactory therapeutic effects of glioblastoma (GBM). Although triggering receptor expressed on myeloid cells-2 (TREM2) involved in forming immunosuppressive microenvironment, but the underlying mechanism and its roles in mediating cancer radioresistance remain unclear, moreover, the efficient delivery of drugs targeting TREM2 to GBM encounters serious challenges. Hence, this study aimed to elucidate the effect and mechanisms of targeted TREM2 silencing on reversing the radioresistance and immune escape of GBM aided by a glutathione-responsive biomimetic nanoparticle (NP) platform.
    METHODS: Radioresistant GBM cell lines and TREM2 stable knockdown GBM cell lines were firstly established. RNA sequencing, colony formation assay, western blot, enzyme-linked immunosorbent assay and co-immunoprecipitation assay were used to detect the molecular mechanisms of TREM2 in regulating the radioresistance and immune escape of GBM. The glutathione-responsive biomimetic NP, angiopep-2 (A2)- cell membrane (CM)-NP/siTREM2/spam1, was then constructed to triply and targeted inhibit TREM2 for in vivo study. Orthotopic GBM-bearing mouse models were established to evaluate the anti-GBM effect of TREM2 inhibition, multiplex immunofluorescence assay was conducted to detect the infiltration of immune cells.
    RESULTS: TREM2 was a regulator in accelerating the radioresistance and immune escape of GBM through participating in DNA damage repair and forming a positive feedback loop with high mobility group box 1 (HMGB1) to cascade the activation of Toll-like receptor 4 (TLR4)/protein kinase B (Akt) signaling. A2-CM-NP/siTREM2/spam1 was successfully synthesized with excellent passive targeting, active targeting and homologous targeting, and the in vivo results exhibited its remarkable anti-GBM therapeutic effect through promoting the infiltration of type 1 helper T cells and CD8+T cells, reducing the infiltration of type 2 helper T cells and regulatory T cells, repolarizing macrophages to M1-type, and decreasing the secretion of pro-tumor and immunosuppressive cytokines.
    CONCLUSIONS: Targeting TREM2 therapy is a promising avenue for optimizing radiotherapy and immunotherapy to improve the prognosis of GBM patients.
    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:响应电离辐射(IR)的细胞衰老通过引起永久性细胞周期停滞来限制受损细胞的复制。然而,IR可以诱导促存活信号通路,从而降低辐射诱导的细胞毒性程度并促进辐射抗性的发展。组蛋白变体H2A的差异掺入。J对高阶染色质组织和建立辐射诱导衰老的表观遗传状态具有深远的影响。然而,H2A的确切表观遗传机制和功能。仍然需要阐明响应于IR暴露的J过表达。方法:主要(无目标,NT)和过表达H2A的基因修饰成纤维细胞。J(H2A。J-OE)暴露于20Gy,并在IR后2周通过免疫组织化学和免疫荧光显微镜分析辐射诱导的衰老。在(未)照射的NT和H2A中分析转录组特征。通过RNA测序的J-OE成纤维细胞。自从H2A。J在人类皮肤的表皮稳态中起着重要作用,H2A的致癌潜力。J在皮肤鳞状细胞癌(cSCC)中进行了研究。分析cSCC的组织微阵列的H2A。通过自动图像分析的J蛋白表达模式。结果:对辐射引起的DNA损伤,H2A的过表达。J损害衰老相关异染色质灶(SAHF)的形成,从而抑制SAHF介导的增殖促进基因的沉默。细胞周期蛋白和细胞周期蛋白依赖性激酶的激活失调会干扰辐照的H2A中的细胞周期停滞。J-OE成纤维细胞,从而克服辐射诱导的衰老。比较转录组分析显示H2A中WNT16信号传导显著增加。红外暴露后的JOE成纤维细胞,促进肿瘤发展和进展的基本机制,包括上皮-间质转化的激活。cSCC的定量分析显示未分化肿瘤与高核H2A相关。J表达式,与更大的致癌潜力有关。结论:H2A。J过表达通过激活WNT16信号通路功能诱导放射抗性并促进致癌转化。H2A.J相关的特征可以通过识别具有更积极的cSCC的患者来改善风险分层,这些患者可能需要增加剂量的放疗。
    Background: Cellular senescence in response to ionizing radiation (IR) limits the replication of damaged cells by causing permanent cell cycle arrest. However, IR can induce pro-survival signaling pathways that reduce the extent of radiation-induced cytotoxicity and promote the development of radioresistance. The differential incorporation of histone variant H2A.J has profound effects on higher-order chromatin organization and on establishing the epigenetic state of radiation-induced senescence. However, the precise epigenetic mechanism and function of H2A.J overexpression in response to IR exposure still needs to be elucidated. Methods: Primary (no target, NT) and genetically modified fibroblasts overexpressing H2A.J (H2A.J-OE) were exposed to 20 Gy and analyzed 2 weeks post-IR for radiation-induced senescence by immunohistochemistry and immunofluorescence microscopy. Transcriptome signatures were analyzed in (non-)irradiated NT and H2A.J-OE fibroblasts by RNA sequencing. Since H2A.J plays an important role in the epidermal homeostasis of human skin, the oncogenic potential of H2A.J was investigated in cutaneous squamous cell carcinoma (cSCC). The tissue microarrays of cSCC were analyzed for H2A.J protein expression pattern by automated image analysis. Results: In response to radiation-induced DNA damage, the overexpression of H2A.J impairs the formation of senescence-associated heterochromatin foci (SAHF), thereby inhibiting the SAHF-mediated silencing of proliferation-promoting genes. The dysregulated activation of cyclins and cyclin-dependent kinases disturbs cell cycle arrest in irradiated H2A.J-OE fibroblasts, thereby overcoming radiation-induced senescence. Comparative transcriptome analysis revealed significantly increased WNT16 signaling in H2A.J OE fibroblasts after IR exposure, promoting the fundamental mechanisms of tumor development and progression, including the activation of the epithelial-mesenchymal transition. The quantitative analysis of cSCCs revealed that undifferentiated tumors are associated with high nuclear H2A.J expression, related with greater oncogenic potential. Conclusion: H2A.J overexpression induces radioresistance and promotes oncogenic transformation through the activation of WNT16 signaling pathway functions. H2A.J-associated signatures may improve risk stratification by identifying patients with more aggressive cSCC who may require radiotherapy with increased doses.
    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
    许多研究表明,放疗后存活的肿瘤细胞更容易转移,但潜在的机制仍不清楚。在这里,我们旨在鉴定上皮-间质转化(EMT)相关的关键基因,这与直肠癌的预后和放射敏感性有关。首先,我们通过分析从癌症基因组图谱数据库检索的直肠癌的RNA表达谱获得差异表达基因,EMT相关基因,和放疗相关数据库,分别。然后,Lasso和Cox回归分析用于基于鉴定的独立保护因子Fibulin5(FBLN5)和独立风险基因EHMT2建立EMT相关预后模型(EMTPM)。高EMTPM组的预后明显较差。然后,我们在外部临床验证队列中评估了签名.通过体内实验,我们进一步证明,EMTPM可有效区分放射敏感性直肠癌患者和放射敏感性直肠癌患者.此外,高EMTPM组的个体与相应个体相比,免疫检查点的表达增加.最后,EMTPM模型的泛癌症分析还表明其在预测接受放疗的肺鳞状细胞癌和乳腺癌患者的预后方面的潜力.总之,我们建立了基于FBLN5和EHMT2表达的直肠癌预后和放射抗性的新预测模型,并表明免疫微环境可能参与了辐射抗性的过程。该预测模型可用于选择直肠癌的管理策略。
    Many studies have shown that tumor cells that survive radiotherapy are more likely to metastasize, but the underlying mechanism remains unclear. Here we aimed to identify epithelial-mesenchymal transition (EMT)-related key genes, which associated with prognosis and radiosensitivity in rectal cancer. First, we obtained differentially expressed genes by analyzing the RNA expression profiles of rectal cancer retrieved from The Cancer Genome Atlas database, EMT-related genes, and radiotherapy-related databases, respectively. Then, Lasso and Cox regression analyses were used to establish an EMT-related prognosis model (EMTPM) based on the identified independent protective factor Fibulin5 (FBLN5) and independent risk gene EHMT2. The high-EMTPM group exhibited significantly poorer prognosis. Then, we evaluated the signature in an external clinical validation cohort. Through in vivo experiments, we further demonstrated that EMTPM effectively distinguishes radioresistant from radiosensitive patients with rectal cancer. Moreover, individuals in the high-EMTPM group showed increased expression of immune checkpoints compared to their counterparts. Finally, pan-cancer analysis of the EMTPM model also indicated its potential for predicting the prognosis of lung squamous cell carcinoma and breast cancer patients undergoing radiotherapy. In summary, we established a novel predictive model for rectal cancer prognosis and radioresistance based on FBLN5 and EHMT2 expressions, and suggested that immune microenvironment may be involved in the process of radioresistance. This predictive model could be used to select management strategies for rectal 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
    长链非编码RNA(lncRNA)与癌症进展和耐药性发展有关。此外,有证据表明lncRNAHOX转录反义基因间RNA(HOTAIR)参与结直肠癌(CRC)的进展。本研究旨在研究lncRNAHOTAIR在赋予CRC细胞放疗抗性中的功能作用,以及潜在的机制。在70对CRC肿瘤和癌旁组织中检测HOTAIR的相对表达水平,以及对辐射敏感和耐辐射样品。使用卡方检验评估HOTAIR表达水平与CRC患者临床特征之间的相关性。功能测定,如细胞增殖,进行集落形成和凋亡测定以确定在用不同剂量的辐射处理后具有HOTAIR沉默的CRC细胞中的放射敏感性。RNA下拉法和荧光原位杂交(FISH)用于确定HOTAIR与DNA损伤反应介质共济失调-毛细血管扩张突变和Rad3相关(ATR)之间的相互作用。HOTAIR在CRC肿瘤组织中显著上调,尤其是抗辐射肿瘤样本.HOTAIR的表达升高与更高级的组织学分级相关,结直肠癌患者远处转移与预后不良。沉默HOTAIR抑制CRC细胞增殖,促进凋亡和放射敏感性。在小鼠异种移植模型中,HOTAIR敲低还抑制CRC细胞的肿瘤发生并增强对放疗的敏感性。此外,数据显示,HOTAIR可以与ATR相互作用,调节DNA损伤修复信号通路。沉默HOTAIR损害了ATR-ATR相互作用蛋白(ATRIP)复合物和细胞周期进程中的信号传导。总的来说,目前的结果表明,lncRNAHOTAIR促进DNA损伤应答途径,并通过靶向ATR促进CRC细胞的放射抗性。
    Long non-coding RNAs (lncRNAs) have been implicated in cancer progression and drug resistance development. Moreover, there is evidence that lncRNA HOX transcript antisense intergenic RNA (HOTAIR) is involved in colorectal cancer (CRC) progression. The present study aimed to examine the functional role of lncRNA HOTAIR in conferring radiotherapy resistance in CRC cells, as well as the underlying mechanism. The relative expression levels of HOTAIR were examined in 70 pairs of CRC tumor and para-cancerous tissues, as well as in radiosensitive and radioresistant samples. The correlations between HOTAIR expression levels and clinical features of patients with CRC were assessed using the Chi-square test. Functional assays such as cell proliferation, colony formation and apoptosis assays were conducted to determine the radiosensitivity in CRC cells with HOTAIR silencing after treatment with different doses of radiation. RNA pull-down assay and fluorescence in situ hybridization (FISH) were used to determine the interaction between HOTAIR and DNA damage response mediator ataxia-telangiectasia mutated- and Rad3-related (ATR). HOTAIR was significantly upregulated in CRC tumor tissues, especially in radioresistant tumor samples. The elevated expression of HOTAIR was correlated with more advanced histological grades, distance metastasis and the poor prognosis in patients with CRC. Silencing HOTAIR suppressed the proliferation and promoted apoptosis and radiosensitivity in CRC cells. HOTAIR knockdown also inhibited the tumorigenesis of CRC cells and enhanced the sensitivity to radiotherapy in a mouse xenograft model. Moreover, the data showed that HOTAIR could interact with ATR to regulate the DNA damage repair signaling pathway. Silencing HOTAIR impaired the ATR-ATR interacting protein (ATRIP) complex and signaling in cell cycle progression. Collectively, the present results indicate that lncRNA HOTAIR facilitates the DNA damage response pathway and promotes radioresistance in CRC cells by targeting ATR.
    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
    骨肉瘤(OS)对电离辐射(IR)的抵抗力是有效患者治疗的障碍。脱嘌呤/无嘧啶核酸内切酶还原/氧化因子1(APE1/Ref-1)是一种具有DNA修复和还原/氧化(氧化还原)活性的多功能蛋白质。我们以前揭示了APE1在OS辐射抗性中的作用;然而,APE1的氧化还原活性是否与OS放射抗性有关尚不清楚。APE1调节共济失调-毛细血管扩张突变(ATM)的激活,在其他癌症中介导放射抗性的DNA损伤反应的起始者。APE1氧化还原活性和ATM激活在OS辐射抗性中的作用是未知的。我们的研究表明,IR增加了OS细胞中APE1的表达和ATM的激活,APE1通过其氧化还原活性直接调节ATM激活。APE1氧化还原抑制剂和ATM抑制剂的组合使用在体外和体内有效地使OS细胞对IR敏感。机械上,联合使用两种抑制剂对OS细胞的放射增敏作用是由铁细胞凋亡增加介导的.与单一抑制剂治疗相比,两种抑制剂的共同治疗显着降低了共同靶向转录因子P53的表达。总的来说,APE1氧化还原活性,ATM激活及其串扰在OS的抗辐照中起着重要作用。协同抑制APE1氧化还原活性和ATM激活通过诱导铁凋亡致敏的OS细胞对IR,这为OS放射治疗提供了一种有希望的策略。
    The resistance of osteosarcoma (OS) to ionizing radiation (IR) is an obstacle for effective patient treatment. Apurinic/apyrimidinic endonuclease-reduction/oxidation factor 1 (APE1/Ref-1) is a multifunctional protein with DNA repair and reduction/oxidation (redox) activities. We previously revealed the role of APE1 in OS radioresistance; however, whether the redox activity of APE1 is involved in OS radioresistance is unclear. APE1 regulates the activation of ataxia-telangiectasia mutated (ATM), an initiator of DNA damage response that mediates radioresistance in other cancers. The role of APE1 redox activity and ATM activation in OS radioresistance is unknown. Our study revealed that IR increased APE1 expression and ATM activation in OS cells, and APE1 directly regulated ATM activation by its redox activity. The combined use of an APE1 redox inhibitor and ATM inhibitor effectively sensitized OS cells to IR in vitro and in vivo. Mechanistically, the increased radiosensitization of OS cells by the combined use of the two inhibitors was mediated by increased ferroptosis. Co-treatment with the two inhibitors significantly decreased expression of the common targeted transcription factor P53 compared with single inhibitor treatment. Collectively, APE1 redox activity, ATM activation and their crosstalk play important roles in the resistance of OS to irradiation. Synergetic inhibition of APE1 redox activity and ATM activation sensitized OS cells to IR by inducing ferroptosis, which provides a promising strategy for OS radiotherapy.
    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
    尽管放疗在食管鳞状细胞癌(ESCC)中具有优越的疗效,肿瘤干细胞(CSCs)的放射抗性导致复发,转移,治疗失败。因此,有必要开发基于CSC的疗法来增强放射治疗。miR-339-5p(miR339)参与基于ESCC队列的干细胞分裂和DNA损伤检查点信号通路。miR339通过靶向USP8抑制ESCC细胞的干性并增强辐射诱导的DNA损伤,表明它是潜在的CSC调节剂和放射增敏剂。考虑到miRNA的循环周期有限和肿瘤靶向能力差,开发了一种基于硫化铋纳米花(Bi@PP)的多功能纳米平台,以有效地传递miR339并提高耐辐射性。有趣的是,Bi@PP由于其花形结构封装了更多的miR339,将超过1000倍的miR339输送到细胞中,优于单独的自由miR339。除了用作载体,Bi@PP有利于动态监测体内递送的miR339的分布,同时抑制肿瘤生长。此外,Bi@PP/miR339可以显着提高患者来源的异种移植模型的放疗疗效。这个多功能平台,掺入更高的miRNA负载能力,pH响应性,缺氧缓解,和CT成像,提供了另一种提高放射敏感性和优化ESCC治疗的方法。
    Despite the superior efficacy of radiotherapy in esophageal squamous cell carcinoma (ESCC), radioresistance by cancer stem cells (CSCs) leads to recurrence, metastasis, and treatment failure. Therefore, it is necessary to develop CSC-based therapies to enhance radiotherapy. miR-339-5p (miR339) is involved in stem cell division and DNA damage checkpoint signaling pathways based on ESCC cohort. miR339 inhibited ESCC cell stemness and enhanced radiation-induced DNA damage by targeting USP8, suggesting that it acts as a potential CSC regulator and radiosensitizer. Considering the limited circulating periods and poor tumor-targeting ability of miRNA, a multifunctional nanoplatform based on bismuth sulfide nanoflower (Bi@PP) is developed to efficiently deliver miR339 and improve radioresistance. Intriguingly, Bi@PP encapsulates more miR339 owing to their flower-shaped structure, delivering more than 1000-fold miR339 into cells, superior to free miR339 alone. Besides being used as a carrier, Bi@PP is advantageous for dynamically monitoring the distribution of delivered miR339 in vivo while simultaneously inhibiting tumor growth. Additionally, Bi@PP/miR339 can significantly enhance radiotherapy efficacy in patient-derived xenograft models. This multifunctional platform, incorporating higher miRNA loading capacity, pH responsiveness, hypoxia relief, and CT imaging, provides another method to promote radiosensitivity and optimize ESCC treatment.
    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
    超过一半的癌症患者在其疾病过程中接受放射治疗。数十年的放射生物学研究已经确定了影响对辐射的生物反应的6个参数,称为6“Rs”:修复,放射敏感性,重新填充,再分配,复氧,和抗肿瘤免疫反应的重新激活。细胞外囊泡(EV)是小的膜结合颗粒,其多种生物学功能被越来越多地记录。在这里,我们讨论了电动汽车在癌细胞对放射疗法反应的编排中的作用的证据。我们强调电动汽车参与DNA修复机制,调节细胞对辐射的敏感性,和促进肿瘤再增殖。此外,电动汽车影响肿瘤复氧动力学,并在促进辐射抗性方面发挥关键作用。最后,我们研究了如何将EV相关策略转化为旨在提高癌症放射治疗疗效的新策略.
    Over half of patients with cancer receive radiation therapy during the course of their disease. Decades of radiobiological research have identified 6 parameters affecting the biological response to radiation referred to as the 6 \"Rs\": Repair, Radiosensitivity, Repopulation, Redistribution, Reoxygenation, and Reactivation of the anti-tumour immune response. Extracellular Vesicles (EVs) are small membrane-bound particles whose multiple biological functions are increasingly documented. Here we discuss the evidence for a role of EVs in the orchestration of the response of cancer cells to radiotherapy. We highlight that EVs are involved in DNA repair mechanisms, modulation of cellular sensitivity to radiation, and facilitation of tumour repopulation. Moreover, EVs influence tumour reoxygenation dynamics, and play a pivotal role in fostering radioresistance. Last, we examine how EV-related strategies could be translated into novel strategies aimed at enhancing the efficacy of radiation therapy against cancer.
    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