Antibiotics, Antineoplastic

抗生素,抗肿瘤
  • 文章类型: Journal Article
    背景:抗肿瘤药物,包括阿霉素,伊达比星,和表柔比星,已发现由于氧化应激-线粒体功能障碍-铁凋亡(ORMFs)而对心脏产生不利影响,这是对蒽环类抗生素诱导的心脏毒性的贡献。为了更好地理解这种现象,本研究分析了ORMFS基因的时间分辨测量值.
    方法:使用人类3D心脏微组织细胞模型研究了三种蒽环类药物对ORMFs基因的影响。以两种剂量(治疗性和毒性)在14天内收集转录组数据。WGCNA确定了关键模块相关基因,功能富集分析研究了ssGSEA量化的生物过程,如免疫细胞浸润和血管生成。从心力衰竭患者和对照受试者收集活检。收集GSE59672和GSE2965用于验证。分子对接用于鉴定蒽环类抗生素与关键基因的相互作用。
    结果:在体内或体外筛选ORMFs基因。使用WGCNA,六个共表达的基因模块被分组,MEblue成为最重要的模块。获得了8个与蓝色模块相交的关键基因与动态响应基因:CD36,CDH5,CHI3L1,HBA2,HSD11B1,OGN,RPL8和VWF。与对照样品相比,除RPL8外,所有关键基因在体外ANT处理设置中都下调,他们的表达水平随着时间的推移而变化。根据功能分析,关键模块相关基因参与血管生成和免疫系统通路.在所有ANT处理的设置中,ssGSEA显示血管生成评分和免疫细胞活性的显著下调,包括活化的CD4T细胞,未成熟B细胞,记忆B细胞,自然杀伤细胞,1型辅助T细胞,和2型T辅助细胞。分子对接显示RPL8和CHI3L1对蒽环类药物显示出显著的结合亲和力。
    结论:本研究集中于人心脏微组织和ANT治疗患者心脏活检组织中ORMFs基因的动态特征。已经强调,在蒽环类抗生素诱导的心脏毒性的情况下,ORMF基因可能有助于免疫浸润和血管生成。对这些基因的透彻了解可能会导致疾病的诊断和治疗。
    BACKGROUND: Antineoplastic medications, including doxorubicin, idarubicin, and epirubicin, have been found to adversely affect the heart due to oxidative stress - mitochondrial dysfunction - ferroptosis (ORMFs), which act as contributing attributes to anthracycline-induced cardiotoxicity. To better understand this phenomenon, the time-resolved measurements of ORMFS genes were analyzed in this study.
    METHODS: The effect of three anthracycline drugs on ORMFs genes was studied using a human 3D cardiac microtissue cell model. Transcriptome data was collected over 14 days at two doses (therapeutic and toxic). WGCNA identified key module-related genes, and functional enrichment analysis investigated the biological processes quantified by ssGSEA, such as immune cell infiltration and angiogenesis. Biopsies were collected from heart failure patients and control subjects. GSE59672 and GSE2965 were collected for validation. Molecular docking was used to identify anthracyclines\'s interaction with key genes.
    RESULTS: The ORMFs genes were screened in vivo or in vitro. Using WGCNA, six co-expressed gene modules were grouped, with MEblue emerging as the most significant module. Eight key genes intersecting the blue module with the dynamic response genes were obtained: CD36, CDH5, CHI3L1, HBA2, HSD11B1, OGN, RPL8, and VWF. Compared with control samples, all key genes except RPL8 were down-regulated in vitro ANT treatment settings, and their expression levels varied over time. According to functional analyses, the key module-related genes were engaged in angiogenesis and the immune system pathways. In all ANT-treated settings, ssGSEA demonstrated a significant down-regulation of angiogenesis score and immune cell activity, including Activated CD4 T cell, Immature B cell, Memory B cell, Natural killer cell, Type 1 T helper cell, and Type 2 T helper cell. Molecular docking revealed that RPL8 and CHI3L1 show significant binding affinity for anthracyclines.
    CONCLUSIONS: This study focuses on the dynamic characteristics of ORMFs genes in both human cardiac microtissues and cardiac biopsies from ANT-treated patients. It has been highlighted that ORMFs genes may contribute to immune infiltration and angiogenesis in cases of anthracycline-induced cardiotoxicity. A thorough understanding of these genes could potentially lead to improved diagnosis and treatment of the disease.
    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
    智能纳米药物递送系统(Cu/ZIF-8@GOx-DOX@HA,以下为CZGDH),由掺杂Cu的沸石咪唑酯骨架8(Cu/ZIF-8,以下为CZ)组成,葡萄糖氧化酶(GOx),多柔比星(DOX),透明质酸(HA)用于肿瘤的靶向给药和协同治疗。CZGDH通过HA的靶向作用特异性进入肿瘤细胞,并表现出酸度触发的生物降解作用,随后释放GOx,DOX,和肿瘤微环境(TME)中的Cu2+。GOx氧化肿瘤细胞中的葡萄糖(Glu)以产生H2O2和葡萄糖酸用于饥饿治疗(ST)。DOX进入肿瘤内细胞核进行化疗(CT)。释放的Cu2+消耗肿瘤细胞中过表达的谷胱甘肽(GSH)以产生Cu+。生成的Cu+和H2O2引发类Fenton反应生成有毒的羟基自由基(·OH),这破坏了肿瘤细胞的氧化还原平衡,并有效地杀死了肿瘤细胞进行化学动力学治疗(CDT)。因此,通过TME激活的级联反应实现了协同多峰肿瘤治疗。纳米药物递送系统具有高的载药率(48.3wt%),三模式协同治疗对肿瘤细胞有很强的杀伤作用(67.45%)。
    An intelligent nanodrug delivery system (Cu/ZIF-8@GOx-DOX@HA, hereafter CZGDH) consisting of Cu-doped zeolite imidazolate framework-8 (Cu/ZIF-8, hereafter CZ), glucose oxidase (GOx), doxorubicin (DOX), and hyaluronic acid (HA) was established for targeted drug delivery and synergistic therapy of tumors. The CZGDH specifically entered tumor cells through the targeting effect of HA and exhibited acidity-triggered biodegradation for subsequent release of GOx, DOX, and Cu2+ in the tumor microenvironment (TME). The GOx oxidized the glucose (Glu) in tumor cells to produce H2O2 and gluconic acid for starvation therapy (ST). The DOX entered the intratumoral cell nucleus for chemotherapy (CT). The released Cu2+ consumed the overexpressed glutathione (GSH) in tumor cells to produce Cu+. The generated Cu+ and H2O2 triggered the Fenton-like reaction to generate toxic hydroxyl radicals (·OH), which disrupted the redox balance of tumor cells and effectively killed tumor cells for chemodynamic therapy (CDT). Therefore, synergistic multimodal tumor treatment via TME-activated cascade reaction was achieved. The nanodrug delivery system has a high drug loading rate (48.3 wt%), and the three-mode synergistic therapy has a strong killing effect on tumor cells (67.45%).
    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
    BACKGROUND: The anthracycline chemotherapeutic antibiotic doxorubicin (DOX) can induce cumulative cardiotoxicity and lead to cardiac dysfunction. Long non-coding RNAs (lncRNAs) can function as important regulators in DOX-induced myocardial injury.
    OBJECTIVE: This study aims to investigate the functional role and molecular mechanism of lncRNA OXCT1 antisense RNA 1 (OXCT1-AS1) in DOX-induced myocardial cell injury in vitro.
    METHODS: Human cardiomyocytes (AC16) were stimulated with DOX to induce a myocardial cell injury model. OXCT1-AS1, miR-874-3p, and BDH1 expression in AC16 cells were determined by RT-qPCR. AC16 cell viability was measured by XTT assay. Flow cytometry was employed to assess the apoptosis of AC16 cells. Western blotting was used to evaluate protein levels of apoptosis-related markers. Dual-luciferase reporter assay was conducted to verify the binding ability between miR-874-3p and OXCT1-AS1 and between miR-874-3p and BDH1. The value of p<0.05 indicated statistical significance.
    RESULTS: OXCT1-AS1 expression was decreased in DOX-treated AC16 cells. Overexpression of OXCT1-AS1 reversed the reduction of cell viability and promotion of cell apoptosis caused by DOX. OXCT1-AS1 is competitively bound to miR-874-3p to upregulate BDH1. BDH1 overexpression restored AC16 cell viability and suppressed cell apoptosis under DOX stimulation. Knocking down BDH1 reversed OXCT1-AS1-mediated attenuation of AC16 cell apoptosis under DOX treatment.
    CONCLUSIONS: LncRNA OXCT1-AS1 protects human myocardial cells AC16 from DOX-induced apoptosis via the miR-874-3p/BDH1 axis.
    OBJECTIVE: O antibiótico quimioterápico antraciclina doxorrubicina (DOX) pode induzir cardiotoxicidade cumulativa e levar à disfunção cardíaca. RNAs não codificantes longos (lncRNAs) podem funcionar como importantes reguladores na lesão miocárdica induzida por DOX.
    OBJECTIVE: Este estudo tem como objetivo investigar o papel funcional e o mecanismo molecular do RNA antisense lncRNA OXCT1 1 (OXCT1-AS1) na lesão celular miocárdica induzida por DOX in vitro.
    UNASSIGNED: Cardiomiócitos humanos (AC16) foram estimulados com DOX para induzir um modelo de lesão celular miocárdica. A expressão de OXCT1-AS1, miR-874-3p e BDH1 em células AC16 foi determinada por RT-qPCR. A viabilidade das células AC16 foi medida pelo ensaio XTT. A citometria de fluxo foi empregada para avaliar a apoptose de células AC16. Western blotting foi utilizado para avaliar os níveis proteicos de marcadores relacionados à apoptose. O ensaio repórter de luciferase dupla foi conduzido para verificar a capacidade de ligação entre miR-874-3p e OXCT1-AS1 e entre miR-874-3p e BDH1. O valor de p<0,05 indicou significância estatística.
    RESULTS: A expressão de OXCT1-AS1 foi diminuída em células AC16 tratadas com DOX. A superexpressão de OXCT1-AS1 reverteu a redução da viabilidade celular e a promoção da apoptose celular causada pela DOX. OXCT1-AS1 está ligado competitivamente ao miR-874-3p para regular positivamente o BDH1. A superexpressão de BDH1 restaurou a viabilidade das células AC16 e suprimiu a apoptose celular sob estimulação com DOX. A derrubada do BDH1 reverteu a atenuação da apoptose de células AC16 mediada por OXCT1-AS1 sob tratamento com DOX.
    UNASSIGNED: LncRNA OXCT1-AS1 protege células miocárdicas humanas AC16 da apoptose induzida por DOX através do eixo miR-874-3p/BDH1.
    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
    婴儿期血管瘤是婴儿期和儿童期最常见的血管瘤。尽管证明了普萘洛尔治疗的疗效,某些患者仍然遇到抵抗或面临复发。对频繁的日常用药的需要也对患者的依从性提出了挑战。博来霉素(BLM)已证明对血管异常有效,然而,它的使用受到剂量相关并发症的限制。解决这个问题,这项研究提出了一种使用BLM负载透明质酸(HA)的微针(MN)贴片治疗血管瘤的新方法。BLM在聚乳酸(PLA)微球(MPs)的合成过程中被包封。通过扫描电子显微镜(SEM)图像证实了PLAMPs和MN贴片的成功制备。HA微针在皮肤插入后迅速溶解,释放BLM@PLA议员。这些MP在28天内逐渐降解,提供BLM的持续释放。全面的安全评估,包括细胞活力,溶血率,和兔子的皮内反应,验证MN补丁的安全性。BLM@PLA-MN在鼠血管瘤模型中表现出对血管瘤形成的有效抑制效率。非常重要的是,RNA-seq分析显示BLM@PLA-MNs通过调节P53途径发挥对血管瘤的抑制作用。总之,BLM@PLA-MN成为有效治疗血管瘤的有希望的临床候选药物。
    Hemangioma of infancy is the most common vascular tumor during infancy and childhood. Despite the proven efficacy of propranolol treatment, certain patients still encounter resistance or face recurrence. The need for frequent daily medication also poses challenges to patient adherence. Bleomycin (BLM) has demonstrated effectiveness against vascular anomalies, yet its use is limited by dose-related complications. Addressing this, this study proposes a novel approach for treating hemangiomas using BLM-loaded hyaluronic acid (HA)-based microneedle (MN) patches. BLM is encapsulated during the synthesis of polylactic acid (PLA) microspheres (MPs). The successful preparation of PLA MPs and MN patches is confirmed through scanning electron microscopy (SEM) images. The HA microneedles dissolve rapidly upon skin insertion, releasing BLM@PLA MPs. These MPs gradually degrade within 28 days, providing a sustained release of BLM. Comprehensive safety assessments, including cell viability, hemolysis ratio, and intradermal reactions in rabbits, validate the safety of MN patches. The BLM@PLA-MNs exhibit an effective inhibitory efficiency against hemangioma formation in a murine hemangioma model. Of significant importance, RNA-seq analysis reveals that BLM@PLA-MNs exert their inhibitory effect on hemangiomas by regulating the P53 pathway. In summary, BLM@PLA-MNs emerge as a promising clinical candidate for the effective treatment of hemangiomas.
    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
    前所未有的微/纳米尺度导航能力和量身定制的功能调整微/纳米马达作为新的目标药物递送系统,为生物医学应用开辟了新的视野。在这里,我们设计了一种光驱动的rGO/Cu2+1O管状纳米马达,用于主动靶向癌细胞作为药物递送系统。在真实细胞培养基(5%葡萄糖细胞等渗溶液)中,推进性能大大提高,归因于引入氧空位和还原的氧化石墨烯(rGO)层,用于分离光诱导的电子-空穴对。可以容易地调节运动速度和方向。同时,由于π-π键效应,多柔比星(DOX)可以快速加载到rGO层上。微型机器人中的Cu2+1O基质不仅可以作为光催化剂产生化学浓度梯度作为驱动力,还可以作为纳米药物杀死癌细胞。光驱动rGO/Cu2+1O纳米马达的强大推进力加上微小的尺寸赋予了它们主动的跨膜运输,协助DOX和Cu2+1O突破细胞膜屏障。与无动力纳米载体和游离DOX相比,光推进rGO/Cu2+1O纳米马达表现出更高的跨膜转运效率和显著的治疗功效。这种概念验证的纳米马达设计提出了一种针对肿瘤的创新方法,将光驱动微/纳米马达的生物医学应用范围扩大到浅表组织治疗。
    The unprecedented navigation ability in micro/nanoscale and tailored functionality tunes micro/nanomotors as new target drug delivery systems, open up new horizons for biomedical applications. Herein, we designed a light-driven rGO/Cu2 + 1O tubular nanomotor for active targeting of cancer cells as a drug delivery system. The propulsion performance is greatly enhanced in real cell media (5% glucose cells isotonic solution), attributing to the introduction of oxygen vacancy and reduced graphene oxide (rGO) layer for separating photo-induced electron-hole pairs. The motion speed and direction can be readily modulated. Meanwhile, doxorubicin (DOX) can be loaded quickly on the rGO layer because of π-π bonding effect. The Cu2 + 1O matrix in the tiny robots not only serves as a photocatalyst to generate a chemical concentration gradient as the driving force but also acts as a nanomedicine to kill cancer cells as well. The strong propulsion of light-driven rGO/Cu2 + 1O nanomotors coupled with tiny size endow them with active transmembrane transport, assisting DOX and Cu2 + 1O breaking through the barrier of the cell membrane. Compared with non-powered nanocarrier and free DOX, light-propelled rGO/Cu2 + 1O nanomotors exhibit greater transmembrane transport efficiency and significant therapeutic efficacy. This proof-of-concept nanomotor design presents an innovative approach against tumor, enlarging the list of biomedical applications of light-driven micro/nanomotors to the superficial tissue 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
    多柔比星(DOX)的临床应用主要受到其严重副作用,药物输送效率差,和有限的免疫原性死亡(ICD)效应。改善基于DOX的化疗并改善其不良反应,我们利用3LL细胞来源的细胞外囊泡包裹DOX和硝普钠(SNP)以获得DOX/SNP@CM,通过利用肿瘤细胞膜固有的同源靶向特性,可以有效地靶向肿瘤部位。DOX在化疗中发挥了作用,SNP成功地响应细胞内GSH以持续产生一氧化氮(NO)。原位产生的NO上调肿瘤细胞表面Fas的表达,从而敏化了Fas/FasL通路介导的肿瘤细胞凋亡的DOX。此外,NO还通过促进ICD对肿瘤细胞的作用来促进细胞毒性T细胞的肿瘤内浸润。重要的是,通过NO介导的Fas/FasL相互作用的操纵,抗肿瘤免疫与Fas/FasL介导的肿瘤细胞凋亡紧密合作,共同使DOX/SNP@CM与低剂量DOX发挥显著的肿瘤生长抑制作用。值得注意的是,DOX和SNP都是临床上应用广泛的药物,确保DOX/SNP@CM成为未来实际应用的潜在机会。
    The clinical application of doxorubicin (DOX) is mainly restricted by its serious side effects, poor drug delivery efficiency, and limited immunogenic death (ICD) effect. To improve DOX-based chemotherapy and ameliorate its adverse effects, we utilized 3LL cell-derived extracellular vesicles to encapsulate DOX and sodium nitroprusside (SNP) to obtain DOX/SNP@CM, which could effectively target the tumor site by harnessing the inherent homologous targeting property of tumor cell membranes. DOX performed its role on chemotherapy, and SNP successfully respond to the intracellular GSH to continuously generate nitric oxide (NO). The in situ-produced NO upregulated the Fas expression on the tumor cell surface, thereby sensitizing the Fas/FasL pathway-mediated tumor cell apoptosis of DOX. Furthermore, NO also boosted the intratumoral infiltration of cytotoxic T cells by promoted ICD effect towards tumor cells. Importantly, the anti-tumor immunity tightly cooperated with Fas/FasL mediated tumor cell apoptosis by NO-mediated manipulation on Fas/FasL interaction, collectively making DOX/SNP@CM exert significant tumor growth inhibition with low-dose DOX. Remarkably, DOX and SNP both are widely used clinical medicines, ensuring DOX/SNP@CM a potential opportunity for future practical applications.
    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
    虽然基于纳米平台的癌症治疗药物已经研究和调查了很多年,增强抗肿瘤疗效和减少毒副作用仍然是一个基本问题。
    我们利用铁(Fe2+)离子和端粒酶靶向发夹DNA结构之间的纳米粒子配位来封装阿霉素(DOX)和制造的Fe2+DNA@DOX纳米粒子(BDDFNPs)。这项工作研究了针对BDDFNPs的能力和生物分布的NIR荧光成像和药代动力学研究。体外和体内研究调查了纳米配方的毒性,成像,和协同治疗效果。
    增强的通透性和保留(EPR)效应和肿瘤靶向导致延长的血液循环时间和高的肿瘤积累。重要的是,根据正常细胞和肿瘤细胞端粒酶活性和铁依赖性的不同,BDDFNPs可以通过提高心肌细胞的抗氧化能力来降低DOX介导的心脏毒性。通过Fe2+介导的铁凋亡和β-连环蛋白/p53途径增强了协同治疗功效,并提高了肿瘤抑制率。
    基于HarpinDNA的纳米平台显示出延长的血液循环,通过端粒酶靶向的肿瘤药物积累,和协同治疗提高抗肿瘤药物疗效。我们的工作为未来的协同化疗提供了新的思路。
    UNASSIGNED: While nanoplatform-based cancer theranostics have been researched and investigated for many years, enhancing antitumor efficacy and reducing toxic side effects is still an essential problem.
    UNASSIGNED: We exploited nanoparticle coordination between ferric (Fe2+) ions and telomerase-targeting hairpin DNA structures to encapsulate doxorubicin (DOX) and fabricated Fe2+-DNA@DOX nanoparticles (BDDF NPs). This work studied the NIR fluorescence imaging and pharmacokinetic studies targeting the ability and biodistribution of BDDF NPs. In vitro and vivo studies investigated the nano formula\'s toxicity, imaging, and synergistic therapeutic effects.
    UNASSIGNED: The enhanced permeability and retention (EPR) effect and tumor targeting resulted in prolonged blood circulation times and high tumor accumulation. Significantly, BDDF NPs could reduce DOX-mediated cardiac toxicity by improving the antioxidation ability of cardiomyocytes based on the different telomerase activities and iron dependency in normal and tumor cells. The synergistic treatment efficacy is enhanced through Fe2+-mediated ferroptosis and the β-catenin/p53 pathway and improved the tumor inhibition rate.
    UNASSIGNED: Harpin DNA-based nanoplatforms demonstrated prolonged blood circulation, tumor drug accumulation via telomerase-targeting, and synergistic therapy to improve antitumor drug efficacy. Our work sheds new light on nanomaterials for future synergistic chemotherapy.
    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
    多柔比星(Dox)是一种广谱的抗肿瘤药物,而心脏毒性限制了其进一步应用。在临床环境中,脂质体递送载体用于降低Dox心脏毒性。这里,我们用细胞外囊泡(EV)代替脂质体,并深入研究了EV包裹的Dox递送机制。结果表明,EVs在体外和体内显着提高了Dox的导入效率和抗肿瘤作用,并且效率从其独特的进入模式中增加了收益。在电动汽车内在化之前,装载Dox的电动汽车重复“亲吻和奔跑”动作。一旦电动汽车接触细胞膜,Dox从电动汽车中分离出来,直接进入细胞质,导致比单一Dox更高、更快的Dox导入。这种独特的进入模式使EV和细胞膜之间的粘附而不是EV内在化的总量成为调节Dox进口的关键因素。此外,我们认为ICAM1是介导电动汽车与细胞膜粘附的分子。有趣的是,EV包裹的Dox可以通过刺激TME中IFN-γ和TNF-α的分泌来诱导ICAM1的表达,从而增加Dox负载型电动汽车的肿瘤靶向性。总之,EV和EV封装的Dox通过ICAM1协同作用,共同增强了肿瘤治疗的疗效。
    Doxorubicin (Dox) is an anti-tumor drug with a broad spectrum, whereas the cardiotoxicity limits its further application. In clinical settings, liposome delivery vehicles are used to reduce Dox cardiotoxicity. Here, we substitute extracellular vesicles (EVs) for liposomes and deeply investigate the mechanism for EV-encapsulated Dox delivery. The results demonstrate that EVs dramatically increase import efficiency and anti-tumor effects of Dox in vitro and in vivo, and the efficiency increase benefits from its unique entry pattern. Dox-loading EVs repeat a \"kiss-and-run\" motion before EVs internalization. Once EVs touch the cell membrane, Dox disassociates from EVs and directly enters the cytoplasm, leading to higher and faster Dox import than single Dox. This unique entry pattern makes the adhesion between EVs and cell membrane rather than the total amount of EV internalization the key factor for regulating the Dox import. Furthermore, we recognize ICAM1 as the molecule mediating the adhesion between EVs and cell membranes. Interestingly, EV-encapsulated Dox can induce ICAM1 expression by irritating IFN-γ and TNF-α secretion in TME, thereby increasing tumor targeting of Dox-loading EVs. Altogether, EVs and EV-encapsulated Dox synergize via ICAM1, which collectively enhances the curative effects for tumor 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
    肿瘤细胞可以表达免疫检查点蛋白程序性死亡-1(PD-1),但是癌细胞固有的PD-1如何响应细胞应激而受到调节仍在很大程度上未知.这里,我们揭示了化疗药物多柔比星(Dox)调节癌细胞固有PD-1的独特机制.Dox上调PD-1mRNA,同时降低肿瘤细胞中的PD-1蛋白水平。虽然Dox缩短了PD-1的半衰期,它不能直接诱导PD-1降解。相反,我们观察到Dox促进肽-N(4)-(N-乙酰-β-葡糖胺基)天冬酰胺酶(NGLY1)和PD-1之间的相互作用,促进NGLY1介导的PD-1去糖基化和去稳定化.PD-1的维持使肿瘤细胞对Dox介导的抗增殖作用敏感。我们的研究揭示了PD-1响应Dox的调节机制,并强调了癌细胞固有PD-1在Dox介导的抗肿瘤作用中的潜在作用。
    Tumor cells can express the immune checkpoint protein programmed death-1 (PD-1), but how cancer cell-intrinsic PD-1 is regulated in response to cellular stresses remains largely unknown. Here, we uncover a unique mechanism by which the chemotherapy drug doxorubicin (Dox) regulates cancer cell-intrinsic PD-1. Dox upregulates PD-1 mRNA while reducing PD-1 protein levels in tumor cells. Although Dox shortens the PD-1 half-life, it fails to directly induce PD-1 degradation. Instead, we observe that Dox promotes the interaction between peptide-N(4)-(N-acetyl-beta-glucosaminyl)asparagine amidase (NGLY1) and PD-1, facilitating NGLY1-mediated PD-1 deglycosylation and destabilization. The maintenance of PD-1 sensitizes tumor cells to Dox-mediated antiproliferative effects. Our study unveils a regulatory mechanism of PD-1 in response to Dox and highlights a potential role of cancer cell-intrinsic PD-1 in Dox-mediated antitumor effects.
    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
    通过使用Cd(II)和有机配体[H3L=4,4\',获得了一种新的3D金属有机骨架[Cd6(L)4(bipy)3(H2O)2·H2O](1),4'\'-(苯-1,3,5-三(氧基)三苯甲酸)苯甲酸;bipy=4,4'-联吡啶]在溶剂热条件下,通过单X射线衍射进行了全面检查,FTIR和元素分析等。以天然多糖透明质酸(HA)和羧甲基壳聚糖(CMCS)为原料,我们成功制备了HA/CMCS水凝胶,并通过扫描电子显微镜观察了其内部微观形态。使用阿霉素(Dox)作为药物模型,我们合成了一种负载阿霉素的新型金属凝胶颗粒,并使用荧光光谱法研究了它们的包封和释放效果,然后通过热重分析进一步研究它们的成分。基于此,评估了对白血病的治疗效果。最后,提出了一种从主体配体自动设计新配体结构的增强学习方法。通过生成建模和分子对接模拟,对宿主和预测的镉配合物的生物学行为进行了广泛的研究。
    A new 3D metal-organic frameworks [Cd6(L)4(bipy)3(H2O)2·H2O] (1) was gained by employing Cd(II) and organic ligand [H3L = 4,4\',4\'\'-(benzene-1,3,5-triyltris(oxy))tribenzoic acid)benzene acid; bipy = 4,4\'-bipyridine] in the solvothermal condition, which has been fully examined via single-X ray diffraction, FTIR and elemental analysis and so on. Using natural polysaccharides hyaluronic acid (HA) and carboxymethyl chitosan (CMCS) as raw materials, we successfully prepared HA/CMCS hydrogels and observed their internal micromorphology by scanning electron microscopy. Using doxorubicin (Dox) as a drug model, we synthesized a novel metal gel particle loaded with doxorubicin, and their encapsulation and release effects were studied using fluorescence spectroscopy, followed by further investigation of their components through thermogravimetric analysis. Based on this, the therapeutic effect on leukemia was evaluated. Finally, an enhanced learning method for automatically designing new ligand structures from host ligands was proposed. Through generative modeling and molecular docking simulations, the biological behavior of the host and predicted cadmium complexes was extensively studied.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号