Radiosensitization

放射增敏
  • 文章类型: Journal Article
    Activation of the stimulator of interferon genes (STING) pathway by radiotherapy (RT) has a significant effect on eliciting antitumor immune responses. The generation of hydroxyl radical (·OH) storm and the sensitization of STING-relative catalytic reactions could improve radiosensitization-mediated STING activation. Herein, multi-functional radiosensitizer with oxygen vacancies depended mimicking enzyme-like activities was fabricated to produce more dsDNA which benefits intracellular 2\', 3\'-cyclic GMP-AMP (cGAMP) generation, together with introducing exogenous cGAMP to activate immune response. MnO2@CeOx nanozymes present enhanced superoxide dismutase (SOD)-like and peroxidase (POD)-like activities due to induced oxygen vacancies accelerate the redox cycles from Ce4+ to Ce3+ via intermetallic charge transfer. CeOx shells not only serve as radiosensitizer, but also provide the conjugation site for AMP/GMP to form MnO2@CeOx-GAMP (MCG). Upon X-ray irradiation, MCG with SOD-like activity facilitates the conversion of superoxide anions generated by Ce-sensitization into H2O2 within tumor microenvironment (TME). The downstream POD-like activity catalyzes the elevated H2O2 into a profusion of ·OH for producing more damage DNA fragments. TME-responsive decomposed MCG could supply exogenous cGAMP, meanwhile the releasing Mn2+ improve the sensitivity of cyclic GMP-AMP synthase to dsDNA for producing more cGAMP, resulting in the promotion of STING pathway activation.
    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
    作为深入宫颈癌(CC)治疗的支柱的放射疗法受到其放射抗性的影响。放射动力疗法(RDT)通过产生具有深层组织渗透的活性氧(ROS)来有效地逆转放射阻力。然而,X射线刺激的光敏剂具有高毒性和能量衰减。因此,设计了X射线响应性非硒化物桥接介孔二氧化硅纳米颗粒(DMNS),装载X射线激活的光敏剂吖啶橙(AO),用于像特洛伊木马一样对放射性抗性宫颈癌(R-CC)进行点爆破RDT。DMNS可以封装大量的AO,在肿瘤微环境(TME)中,它含有高浓度的过氧化氢,X射线辐射触发了二硒烯键的裂解,导致DMSNs降解,并因此直接在肿瘤部位释放AO。一方面,它解决了快速药物清除的问题,不利分布,和简单的AO治疗引起的副作用。另一方面,它充分利用了高穿透性X线响应性RDT的优势,提高了放射治疗的敏感性。这种方法导致ROS诱导的线粒体损伤,抑制DNA损伤修复,细胞周期阻滞和促进癌细胞凋亡的R-CC。X射线响应性DMSNs@AO在克服R-CC治疗中的高级RDT障碍方面具有相当大的潜力。
    Radiotherapy as a mainstay of in-depth cervical cancer (CC) treatment suffers from its radioresistance. Radiodynamic therapy (RDT) effectively reverses radio-resistance by generating reactive oxygen species (ROS) with deep tissue penetration. However, the photosensitizers stimulated by X-ray have high toxicity and energy attenuation. Therefore, X-ray responsive diselenide-bridged mesoporous silica nanoparticles (DMSNs) are designed, loading X-ray-activated photosensitizer acridine orange (AO) for spot blasting RDT like Trojan-horse against radio-resistance cervical cancer (R-CC). DMSNs can encapsulate a large amount of AO, in the tumor microenvironment (TME), which has a high concentration of hydrogen peroxide, X-ray radiation triggers the cleavage of diselenide bonds, leading to the degradation of DMSNs and the consequent release of AO directly at the tumor site. On the one hand, it solves the problems of rapid drug clearance, adverse distribution, and side effects caused by simple AO treatment. On the other hand, it fully utilizes the advantages of highly penetrating X-ray responsive RDT to enhance radiotherapy sensitivity. This approach results in ROS-induced mitochondria damage, inhibition of DNA damage repair, cell cycle arrest and promotion of cancer cell apoptosis in R-CC. The X-ray responsive DMSNs@AO hold considerable potential in overcoming obstacles for advanced RDT in the treatment of R-CC.
    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:增殖细胞核抗原(PCNA)对于DNA复制和修复至关重要,细胞生长,和生存。PCNA还增强前列腺癌(PC)细胞中的雄激素受体(AR)信号传导。我们在AR的N端结构域鉴定了PCNA相互作用蛋白(PIP)盒,并开发了包含ARPIP盒的小肽PCNA抑制剂R9-AR-PIP。我们还鉴定了一系列与PCNA直接结合并中断PCNA功能的小分子PCNA抑制剂(PCNA-Is)。本研究调查了PCNA抑制剂对PC细胞对X射线辐射的敏感性的影响。
    方法:在4种具有不同AR表达状态的抗去势PC(CRPC)细胞中研究了靶向PCNA对PC细胞放射敏感性的影响。用单独的或组合的PCNA抑制剂和X射线辐射处理细胞。治疗对AR靶基因表达的影响,DNA损伤反应,DNA损伤,同源重组修复(HRR),和细胞毒性进行评估。
    结果:我们发现,PCNA-I1S或R9-AR-PIP结合X射线辐射,AR对DNA损伤反应基因PARP1的雄激素反应元件(ARE)占有率显着减弱,而X射线辐射本身并不能增强ARE的占有率。PCNA-I1S或R9-AR-PIP单独显著抑制PRKDC和XRCC2基因中AR占据区(AROR)的占据。R9-AR-PIP和PCNA-I1S抑制AR-Vs靶基因细胞周期蛋白A2的表达,并在AR阳性CRPC细胞中显示出辐射的累加效应。通过PCNA-I1S和R9-AR-PIP靶向PCNA下调DNA损伤反应基因EXO1,Rad54L,Rad51和/或PARP1显示了与AR阳性CRPCLNCaP-AI中各自对照相比,辐射的加性效应,22Rv1和R1-D567单元,但不在AR阴性PC-3细胞中。R9-AR-PIP和PCNA-I1S提高磷酸-DNA-PKcs(S2056)和γH2AX的水平,表明AR阳性细胞对辐射的DNA损伤。PCNA抑制剂PCNA-I1S显著减弱HRR,R9-AR-PIP,和T2AA在所有四个CRPC细胞检查,并仅在22RV1细胞中被恩扎鲁胺(Enz)抑制。在集落形成测定中,Enz和较低浓度的R9-AR-PIP增强了X射线辐射在雄激素依赖性LNCaP细胞中诱导的细胞毒性。更高浓度的R9-AR-PIP减少集落形成,并在所有AR表达细胞中与X射线辐射具有加性效应,不管AR-FL和AR-Vs,但不显著改变AR阴性PC-3细胞的集落形成。PCNA-I1S减弱集落形成,并在所有四个CRPC细胞中具有电离辐射的累加效应,无论AR表达状态如何。
    结论:这些数据为在临床前模型中使用PCNA-I1S或R9-AR-PIP联合X线辐射对CRPC肿瘤的治疗研究提供了有力的依据。
    BACKGROUND: Proliferating cell nuclear antigen (PCNA) is essential for DNA replication and repair, cell growth, and survival. PCNA also enhances androgen receptor (AR) signaling in prostate cancer (PC) cells. We identified a PCNA interaction protein (PIP) box at the N-terminal domain of AR and developed a small peptide PCNA inhibitor R9-AR-PIP containing AR PIP-box. We also identified a series of small molecule PCNA inhibitors (PCNA-Is) that bind directly to PCNA and interrupt PCNA functions. The present study investigated the effects of the PCNA inhibitors on the sensitivity of PC cells to X-ray radiation.
    METHODS: The effects of targeting PCNA on radio sensitivity of PC cells were investigated in four lines of castration-resistant PC (CRPC) cells with different AR expression statuses. The cells were treated with the PCNA inhibitors and X-ray radiation alone or in combination. The effects of the treatment on expression of AR target genes, DNA damage response, DNA damage, homologous recombination repair (HRR), and cytotoxicity were evaluated.
    RESULTS: We found that the androgen response element (ARE) occupancy of the DNA damage response gene PARP1 by AR is significantly attenuated by PCNA-I1S or R9-AR-PIP combined with X-ray radiation, while X-ray radiation alone does not enhance the ARE occupancy. PCNA-I1S or R9-AR-PIP alone significantly inhibits occupancy of the AR-occupied regions (AROR) in PRKDC and XRCC2 genes. R9-AR-PIP and PCNA-I1S inhibit expression of AR-Vs target gene cyclin A2 and show the additive effects with radiation in AR-positive CRPC cells. Targeting PCNA by PCNA-I1S and R9-AR-PIP downregulates expression of DNA damage response genes EXO1, Rad54L, Rad51, and/or PARP1 and shows the additive effects with radiation as compared with their respective controls in AR-positive CRPC LNCaP-AI, 22Rv1, and R1-D567 cells, but not in AR-negative PC-3 cells. R9-AR-PIP and PCNA-I1S elevate the levels of phospho-DNA-PKcs(S2056) and γH2AX, indicating DNA damage in response to radiation in AR-positive cells. The HRR is significantly attenuated by PCNA inhibitors PCNA-I1S, R9-AR-PIP, and T2AA in all four CRPC cells examined, and inhibited by Enzalutamide (Enz) only in 22RV1 cells. The cytotoxicity induced by X-ray radiation in androgen-dependent LNCaP cells is enhanced by Enz and a lower concentration of R9-AR-PIP in the colony formation assay. R9-AR-PIP at higher concentration reduces the colony formation and has an additive effect with X-ray radiation in all AR expressing cells, regardless of AR-FL and AR-Vs, but does not significantly alter the colony formation in AR-negative PC-3 cells. PCNA-I1S attenuates colony formation and has an additive effect with ionizing radiation in all four CRPC cells, regardless of AR expression status.
    CONCLUSIONS: These data provide a strong rationale for the therapy studies using PCNA-I1S or R9-AR-PIP in combination with X-ray radiation against CRPC tumors in preclinical models.
    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:放射治疗(RT)是头颈部鳞状细胞癌(HNSCC)患者不可或缺的治疗部分,但是抗辐射仍然是一个主要问题。这里,我们用MitoTam,他莫昔芬的线粒体靶向类似物,我们的目标是刺激铁细胞死亡,并使放射抗性细胞对RT敏感。
    方法:我们评估了生存能力,活性氧(ROS)的产生,线粒体膜电位的破坏,以及MitoTam处理后放射敏感性(UT-SCC-40)和放射抗性(UT-SCC-5)HNSCC细胞中的脂质过氧化。为了评估铁凋亡的特异性,我们使用了铁凋亡抑制剂铁抑制素-1(fer-1)。此外,评估总抗氧化能力和对叔丁基过氧化氢的敏感性以评估ROS反应。53BP1染色用于评估MitoTam治疗后的放射敏感性。
    结果:我们的数据显示ROS增加,细胞死亡,线粒体膜电位的破坏,和MitoTam处理后两种细胞系的脂质过氧化。MitoTam对细胞死亡的不利影响,fer-1阻止了膜电位和脂质过氧化,表明铁凋亡的诱导。由于固有的较高的抗氧化能力,耐辐射HNSCC细胞对MitoTam的作用不太敏感。与单独的RT或MitoTam相比,RT之前的MitoTam处理导致由53BP1病灶表达的超加性残留DNA损伤。
    结论:MitoTam诱导HNSCC细胞铁凋亡,可用于克服耐辐射细胞的抗氧化能力升高,并使此类细胞对RT敏感。因此,用MitoTam接着用RT治疗可以提供一种有希望的抗放射性癌症的有效疗法。
    结论:放射治疗应用于大多数癌症患者的治疗。通过结合ROS诱导药物MitoTam和放射疗法促进铁细胞死亡,可以克服由于抗氧化剂水平升高引起的辐射抗性。
    OBJECTIVE: Radiotherapy (RT) is an integral treatment part for patients with head and neck squamous cell carcinoma (HNSCC), but radioresistance remains a major issue. Here, we use MitoTam, a mitochondrially targeted analogue of tamoxifen, which we aim to stimulate ferroptotic cell death with, and sensitize radioresistant cells to RT.
    METHODS: We assessed viability, reactive oxygen species (ROS) production, disruption of mitochondrial membrane potential, and lipid peroxidation in radiosensitive (UT-SCC-40) and radioresistant (UT-SCC-5) HNSCC cells following MitoTam treatment. To assess ferroptosis specificity, we used the ferroptosis inhibitor ferrostatin-1 (fer-1). Also, total antioxidant capacity and sensitivity to tert-butyl hydroperoxide were evaluated to assess ROS-responses. 53BP1 staining was used to assess radiosensitivity after MitoTam treatment.
    RESULTS: Our data revealed increased ROS, cell death, disruption of mitochondrial membrane potential, and lipid peroxidation following MitoTam treatment in both cell lines. Adverse effects of MitoTam on cell death, membrane potential and lipid peroxidation were prevented by fer-1, indicating induction of ferroptosis. Radioresistant HNSCC cells were less sensitive to the effects of MitoTam due to intrinsic higher antioxidant capacity. MitoTam treatment prior to RT led to superadditive residual DNA damage expressed by 53BP1 foci compared to RT or MitoTam alone.
    CONCLUSIONS: MitoTam induced ferroptosis in HNSCC cells, which could be used to overcome the elevated antioxidant capacity of radioresistant cells and sensitize such cells to RT. Treatment with MitoTam followed by RT could therefore present a promising effective therapy of radioresistant cancers.
    CONCLUSIONS: Radiotherapy is applied in the treatment of a majority of cancer patients. Radioresistance due to elevated antioxidant levels can be overcome by promoting ferroptotic cell death combining ROS-inducing drug MitoTam with 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
    目的:本研究旨在阐明DHA如何增强BC的放射敏感性,并解释其潜在的作用机制。
    方法:通过Sanger测序确认了hsa_circ_0001610的圆形结构,RNaseR处理,使用gDNA或cDNA的RT-PCR分析。通过荧光原位杂交检测hsa_circ_0001610和microRNA-139-5p(miR-139-5p)的细胞定位。细胞计数试剂盒-8测定,伤口愈合和集落形成试验,用于评估细胞增殖,而流式细胞术用于估计细胞周期进程和细胞凋亡。进行了活性氧和丙二醛实验以验证BC细胞的铁凋亡。ncRNAs和mRNAs的表达通过qRT-PCR定量,蛋白表达采用蛋白质印迹分析。通过建立BC小鼠模型,研究了hsa_circ_0001610和DHA对BC体内放射敏感性的影响。
    结果:体内和体外实验结果表明,DHA至少部分通过抑制hsa_circ_0001610/miR-139-5p/SLC7A11通路促进BC细胞的铁凋亡,从而增强BC细胞的放射敏感性。
    结论:我们的研究结果表明,DHA可以通过下调hsa_circ_0001610诱导BC细胞的铁凋亡,从而增强放射敏感性,提出了一种有希望的增强BC放射敏感性的治疗策略,值得进一步探索。
    OBJECTIVE: This study aimed to elucidate how DHA enhances the radiosensitivity of BC and to explain its potential mechanisms of action.
    METHODS: The circular structure of hsa_circ_0001610 was confirmed by Sanger sequencing, RNase R treatment, RT-PCR analysis using gDNA or cDNA. Cellular localization of hsa_circ_0001610 and microRNA-139-5p (miR-139-5p) was detected by fluorescence in situ hybridization. Cell counting kit-8 assay, wound healing and colony formation tests for assessing cell proliferation, while flow cytometry was utilized to estimate cell cycle progression and apoptosis. Reactive oxygen species and malondialdehyde experiments were conducted to validate ferroptosis of BC cells. The expression of ncRNAs and mRNAs was quantified via qRT-PCR, and protein expression was analyzed using Western blot. The effects of hsa_circ_0001610 and DHA on radiosensitivity of BC in vivo were studied by establishing BC mice model.
    RESULTS: In vivo and in vitro experimental results indicate that DHA promotes ferroptosis of BC cells at least partly by inhibiting hsa_circ_0001610/miR-139-5p/SLC7A11 pathway, thereby enhancing the radiosensitivity of BC cells.
    CONCLUSIONS: Our findings showed that DHA can induce ferroptosis of BC cells by down-regulation of hsa_circ_0001610, thus enhancing radiosensitivity, suggesting a promising therapeutic strategy for enhancing BC radiosensitivity that is worthy of further exploration.
    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
    许多天然物质具有抗癌特性。特别是土著人使用水性植物提取物作为茶或软膏,包括薯岩花来治疗各种疾病。这项研究的目的是评估从肯尼亚收集的山药水提物的细胞毒性和放射增敏潜力,这些水提物是在三维层粘连蛋白中生长的一组HPV阴性和阳性头颈部鳞状细胞癌(HNSCC)细胞中的。富含细胞外基质(3DlrECM)。结果显示细胞毒性,山药提取物在HPV阴性和阳性HNSCC模型中以浓度和细胞模型依赖性方式进行放射增敏和增加残留双链断裂(DBS)水平。ROS清除剂的应用表明ROS诱导的DSB与通过山药Pax预处理的放射敏化之间存在关联。基于高效液相色谱-串联质谱(HPLC-MS/MS)的分析鉴定了山药提取物的主要成分包括喜树碱。总的来说,山药水提物单独和与X射线照射组合显示出有效的抗癌特性,这是值得进一步机制研究的。
    Numerous natural substances have anti-cancer properties. Especially indigenous people use aqueous plant extracts for tea or ointments including Dioscorea sansibarensis Pax to treat various diseases. The aim of this study was to evaluate the cytotoxic and radiosensitizing potential of aqueous extracts from Dioscorea sansibarensis Pax collected from Kenya in a panel of HPV-negative and -positive head and neck squamous cell carcinoma (HNSCC) cells grown in three-dimensional laminin-rich extracellular matrix (3D lrECM). The results show cytotoxicity, radiosensitization and increased levels of residual double strand breaks (DBS) by Dioscorea sansibarensis Pax extracts in HPV-negative and -positive HNSCC models in a concentration- and cell model-dependent manner. Application of ROS scavengers indicated an association between ROS-induced DSB and radiosensitization through Dioscorea sansibarensis Pax pretreatment. High-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS) based characterization of Dioscorea sansibarensis Pax identified the main components of the extract including camptothecin. Overall, Dioscorea sansibarensis Pax aqueous extracts alone and in combination with X-ray irradiation showed effective anticancer properties, which are worthy of further mechanistic investigation.
    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
    CDK4/6抑制剂联合内分泌治疗可延长激素受体(HR)阳性和HER2阴性晚期乳腺癌患者的生存期.我们研究了CDK4/6抑制剂是否增强了乳腺癌这种亚型的放射敏感性及其潜在机制。使用两种HR阳性和HER2阴性乳腺癌细胞系(MCF-7和T-47D)进行了体外和体内实验,CDK4/6抑制剂(ribociclib和palbociclib)和放疗(RT)的生物学功能和机制评估。使用克隆形成测定法评估辐射增强效果;通过免疫荧光评估γH2AX和53BP1水平以评估DNA损伤。磷酸化(p)-ERK的水平,c-Myc,和DNA双链断裂(DSB)相关分子,p-DNA-PKcs,Rad51和p-ATM,通过蛋白质印迹进行评估。我们使用NF-κBp65转录因子测定试剂盒来评估NF-κB活性。我们通过MCF-7原位异种移植模型评估了RT和ribociclib组合的抗肿瘤作用。克隆实验证明了RT与ribociclib和palbociclib预处理联合使用的协同作用。CDK4/6抑制剂协同增加RT诱导的γH2AX和53BP1的数量,下调p-DNA-PKcs的表达,Rad51和由RT激活的p-ATM,和减少RT触发p-ERK表达,NF-κB激活,和它的下游基因,c-Myc.瑞博西尼和RT联合降低MCF-7细胞异种移植瘤的生长,下调p-ERK的免疫组织化学表达,p-NF-κBp65和c-Myc与对照组比较。结合CDK4/6抑制剂至少通过减少DNA-DSB修复和减弱RT对ERK和NF-κB信号的激活来增强HR阳性和HER2阴性乳腺癌细胞的放射敏感性。
    CDK4/6 inhibitors combined with endocrine therapy prolonged survival in hormone receptor (HR)-positive and HER2-negative advanced breast cancer. We investigated whether CDK4/6 inhibitors enhance radiosensitivity and their underlying mechanisms of this subtype of breast cancer. In vitro and in vivo experiments were conducted using two HR-positive and HER2-negative breast cancer cell lines (MCF-7 and T-47D), CDK4/6 inhibitors (ribociclib and palbociclib) and radiotherapy (RT) to assess the biological functions and mechanisms. The radiation-enhancing effect was assessed using clonogenic assays; γH2AX and 53BP1 levels were assessed by immunofluorescence to evaluate DNA damage. The levels of phospho (p)-ERK, c-Myc, and DNA-double strand break (DSB)-related molecules, p-DNA-PKcs, Rad51, and p-ATM, were assessed by western blotting. We used an NF-κB p65 transcription factor assay kit to evaluate NF-κB activity. We evaluated the antitumor effect of the combination of RT and ribociclib through the MCF-7 orthotopic xenograft model. The synergistic effects of combining RT with ribociclib and palbociclib pretreatment were demonstrated by clonogenic assay. CDK4/6 inhibitors synergistically increased the numbers of RT-induced γH2AX and 53BP1, downregulated the expression of p-DNA-PKcs, Rad51 and p-ATM activated by RT, and reduced RT-triggering p-ERK expression, NF-κB activation, and its down-streaming gene, c-Myc. Combined ribociclib and RT reduced the growth of MCF-7 cell xenograft tumors, and downregulated the immunohistochemical expression of p-ERK, p-NF-κB p65, and c-Myc compared to that in the control group. Combining CDK4/6 inhibitors enhanced radiosensitivity of HR-positive and HER2-negative breast cancer cells at least by reducing DNA-DSB repair and weakening the activation of ERK and NF-κB signaling by RT.
    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
    头颈部鳞状细胞癌(HNSCC)在全球范围内普遍存在,复发率高,存活率低,以及患者生活质量差。来自PAC-1的SM-1可以激活proaspase-3并诱导癌细胞凋亡以发挥抗肿瘤作用。然而,联合放疗后SM-1对HNSCC的抑制作用尚不清楚。本研究旨在探讨SM-1对HNSCC的体内外放射增敏作用。
    MTT方法用于检测SM-1对HNSCC细胞系(HONE1,HSC-2和CAL27)活力的影响。通过集落形成实验确定SM-1联合辐射对HONE1,HSC-2和CAL27细胞系存活指数的影响。采用流式细胞术观察SM-1与放疗联合应用对细胞凋亡和细胞周期的影响,进行蛋白质印迹实验以检测细胞凋亡和细胞周期相关蛋白的表达。最后,建立CAL27异种移植瘤模型,评价SM-1联合放射的体内抗肿瘤作用。
    体外,SM-1有效抑制HNSCC细胞系HONE1、HSC-2和CAL27细胞的活性,并在联合照射过程中协同表现出抗增殖活性。同时,SM-1对HNSCC的抗肿瘤作用高于Debio1143,细胞的放射敏感性大大提高。流式细胞术和westernblot分析表明,SM-1通过抑制CyclinB1和CDC2的表达,诱导头颈部鳞状细胞癌细胞G2/M期阻滞。此外,SM-1激活caspase-3活性并上调PARP1的裂解形式以诱导细胞凋亡。在体内,SM-1联合照射显示出良好的抗肿瘤效果。
    SM-1在体外和体内增强了HNSCC细胞的辐射敏感性,支持其作为放射增敏剂的潜力与放射治疗相结合的临床试验。
    UNASSIGNED: Head and neck squamous cell carcinoma (HNSCC) is globally prevalent with high recurrence, low survival rate, and poor quality of life for patients. Derived from PAC-1, SM-1 can activate procaspase-3 and induce apoptosis in cancer cells to exert anti-tumor effects. However, the inhibitory effect of SM-1 on HNSCC after combination with radiation are unclear. This study aims to investigate the radiosensitizing effect of SM-1 on HNSCC in vitro and in vivo.
    UNASSIGNED: MTT method was used to detect the effect of SM-1 on the viability of HNSCC cell lines (HONE1, HSC-2, and CAL27). The effects of SM-1 combined with radiation on the survival index of HONE1, HSC-2, and CAL27 cell lines were determined by colony formation assay. Flow cytometry was used to investigate the effects of SM-1 and radiation combination on cell apoptosis and cell cycle, and western blot experiments were performed to detect the expression of apoptosis and cell cycle-related proteins. Finally, a xenograft tumor model of CAL27 was established to evaluate the anti-tumor effect of SM-1 combined with radiation in vivo.
    UNASSIGNED: In vitro, SM-1 effectively inhibited the activity of HNSCC cell lines HONE1, HSC-2, and CAL27 cells, and synergistically showed anti-proliferation activity during combined irradiation. Meanwhile, anti-tumor effect of SM-1 on HNSCC was higher than that of Debio1143, and the radiosensitivity of cells was greatly increased. Flow cytometry and western blot analysis showed that SM-1 induced G2/M phase arrest of head and neck squamous cell carcinoma cells via inhibiting the expression of CyclinB1 and CDC2. Moreover, SM-1 activated caspase-3 activity and up-regulated the cleaved form of PARP1 to induce cell apoptosis. In vivo, SM-1 combined irradiation showed a good anti-tumor effect.
    UNASSIGNED: SM-1 enhances HNSCC cell radiation sensitivity in vitro and in vivo, supporting its potential as a radiosensitizer for clinical trials in combination with 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
    生脉健黄散(SMJHS)是一种传统的中药复方,据报道可以抑制鼻咽癌(NPC)的进展并增强放射敏感性。然而,SMJHS抗鼻咽癌的具体活性成分和调控机制,特别是在缺氧条件下,仍然不清楚。在这项研究中,Sprague-Dawley(SD)大鼠用生脉健黄散(SMJHS)灌胃,他们的血液是从腹主动脉采集的.UHPLC-Q-ExactiveorbitrapMS/MS用于鉴定SMJHS含药血清的代谢物谱。通过网络药理学和分子对接,构建了SMJHS靶向NPC中活性成分的分子网络。HIF-1α/VEGF通路处于关键位置。SMJHS对细胞增殖的影响,迁移,通过体外实验评估缺氧NPC细胞的放射敏感性。使用慢病毒建立稳定过表达HIF-1α的NPC细胞系,以研究SMJHS对缺氧NPC细胞中HIF-1α/VEGF信号传导的调节。通过结合网络药理分析,细胞生物功能验证,分子生化实验,我们的研究发现SMJHS对缺氧条件下培养的NPC细胞具有抗增殖作用,抑制其迁移并增加其放射敏感性。此外,SMJHS抑制HIF-1α和VEGFA的表达,显示作为改善NPC治疗的有效选择的潜力。
    Shengmai Jianghuang San (SMJHS) is a traditional Chinese herbal compound reported to inhibit Nasopharyngeal Carcinoma (NPC) progression and enhance radiosensitivity. However, the specific active ingredients and regulatory mechanisms of SMJHS against NPC, particularly under hypoxic conditions, remain unclear. In this study, Sprague-Dawley (SD) rats were gavaged with Shengmai Jianghuang San (SMJHS), and their blood was collected from the abdominal aorta. UHPLC-Q-Exactive orbitrap MS/MS was used to identify the metabolite profiles of SMJHS drug-containing serum. A molecular network of the active compositions in SMJHS targeting NPC was constructed through network pharmacology and molecular docking. The HIF-1α/VEGF pathway was in key positions. The effects of SMJHS on the proliferation, migration, and radiosensitivity of hypoxic NPC cells were assessed by in vitro experiments. NPC cell lines stably overexpressing HIF-1α were established using a lentivirus to investigate the regulation of HIF-1α/VEGF signaling in hypoxic NPC cells by SMJHS. Through a combination of network pharmacological analysis, cellular biofunctional validation, and molecular biochemical experiments, our study found that SMJHS had an anti-proliferative effect on NPC cells cultured under hypoxic conditions, inhibiting their migration and increasing their radiosensitivity. Additionally, SMJHS suppressed the expression of HIF-1α and VEGFA, exhibiting potential as an effective option for improving NPC 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
    不可切除的直肠癌患者的治疗结果是复杂的,同步放化疗是主要的治疗选择。放射增敏剂可以增强局部瘤内缺氧的效果,有助于局部控制和症状缓解。本研究评估了无法切除的直肠癌患者使用过氧化氢联合放射治疗(RT)进行放射增敏的可行性和安全性。本研究共招募13例直肠癌患者。每周两次与RT组合进行放射增敏。将浸泡在3%过氧化氢溶液中的纱布插入肛门,确保与病变的牢固接触。总的来说,每周5天使用10MVX射线从四个方向以25-33个部分将45-65Gy递送至整个骨盆。在RT完成后1个月和6个月评估急性和晚期不良事件。治疗耐受性良好,没有发现急性3级或更严重的事件,没有病人出现直肠瘘,坏死,阻塞,穿孔,狭窄,溃疡或腹膜后出血。无明显的晚期不良事件,超过6个月,在分析结束时观察到。所有患者疼痛缓解,止血效果和肿瘤缩小。因此,对于无法手术的直肠肿瘤患者,在直肠中使用过氧化氢溶液浸泡的纱布可能是一个有希望的选择.本研究的局限性在于患者群体较少,观察时间相对较短。本研究在大学医院医学信息网络中心进行了回顾性注册(试验注册编号:R000061902)于2024年4月21日。
    The treatment outcomes of patients with unresectable rectal cancer are complex, and concurrent chemoradiation therapy is the main treatment option. Radiosensitizers can enhance the effect of localized intratumoral hypoxia, contributing to local control and symptomatic relief. The present study evaluated the feasibility and safety of radiosensitization using hydrogen peroxide combined with radiation therapy (RT) in patients with unresectable rectal cancer. A total of 13 patients with rectal cancer were recruited in the present study. Radiosensitization was performed twice weekly in combination with RT. Gauze soaked in 3% hydrogen peroxide solution was inserted into the anus, ensuring firm contact with the lesion. In total, 45-65 Gy was delivered in 25-33 fractions to the whole pelvis from four directions using 10 MV X-rays 5 days per week. Acute and late adverse events were evaluated 1 and 6 months after the completion of RT. Treatment was well tolerated, with no acute grade 3 or worse events noted, and no patient developed rectal fistula, necrosis, obstruction, perforation, stenosis, ulcer or retroperitoneal hemorrhage. No notable late adverse events, beyond 6 months, were observed at the end of the analysis. All patients experienced pain relief, hemostatic effects and tumor shrinkage. Therefore, the use of a hydrogen peroxide solution-soaked gauze in the rectum may be a promising option for patients with inoperable rectal tumors. The limitations of the present study are that the patient population was small and the observation time was relatively short. This study was retrospectively registered with the University Hospital Medical Information Network Center (trial registration no. R000061902) on April 21, 2024.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号