DR5

DR5
  • 文章类型: Journal Article
    目的:急性心肌梗死(AMI)是死亡的主要原因。中性粒细胞在AMI或缺血再灌注(I/R)损伤期间穿透受损的心脏组织并产生炎症因子,趋化因子,和加剧心脏损伤的细胞外诱捕网。已经证明TRAIL-DR5途径的抑制以白细胞依赖性方式减轻心脏缺血-再灌注损伤。然而,目前尚不清楚TRAIL-DR5信号是否参与调节中性粒细胞胞外捕获网(NETs)的释放.
    方法:本研究使用各种模型来检查用可溶性小鼠TRAIL蛋白激活TRAIL-DR5通路并使用DR5敲除小鼠或mDR5-Fc融合蛋白抑制TRAIL-DR5信号通路对NETs形成和心脏损伤的影响。所使用的模型包括涉及骨髓来源的嗜中性粒细胞和原代心肌细胞的共培养模型和小鼠中的心肌I/R模型。
    结果:NETs的形成受到TRAIL-DR5信号通路抑制的抑制,这可以减轻心脏I/R损伤。这种干预减少了粘附分子和趋化因子的释放,导致中性粒细胞浸润减少,并通过下调中性粒细胞中的PAD4来抑制NETs的产生。
    结论:这项工作阐明了TRAIL-DR5信号通路如何在心肌I/R损伤期间调节中性粒细胞反应,从而为心肌梗死中靶向TRAIL-DR5信号通路的治疗干预提供科学依据。
    OBJECTIVE: Acute myocardial infarction (AMI) is a leading cause of mortality. Neutrophils penetrate injured heart tissue during AMI or ischemia-reperfusion (I/R) injury and produce inflammatory factors, chemokines, and extracellular traps that exacerbate heart injury. Inhibition of the TRAIL-DR5 pathway has been demonstrated to alleviate cardiac ischemia-reperfusion injury in a leukocyte-dependent manner. However, it remains unknown whether TRAIL-DR5 signaling is involved in regulating neutrophil extracellular traps (NETs) release.
    METHODS: This study used various models to examine the effects of activating the TRAIL-DR5 pathway with soluble mouse TRAIL protein and inhibiting the TRAIL-DR5 signaling pathway using DR5 knockout mice or mDR5-Fc fusion protein on NETs formation and cardiac injury. The models used included a co-culture model involving bone marrow-derived neutrophils and primary cardiomyocytes and a model of myocardial I/R in mice.
    RESULTS: NETs formation is suppressed by TRAIL-DR5 signaling pathway inhibition, which can lessen cardiac I/R injury. This intervention reduces the release of adhesion molecules and chemokines, resulting in decreased neutrophil infiltration and inhibiting NETs production by downregulating PAD4 in neutrophils.
    CONCLUSIONS: This work clarifies how the TRAIL-DR5 signaling pathway regulates the neutrophil response during myocardial I/R damage, thereby providing a scientific basis for therapeutic intervention targeting the TRAIL-DR5 signaling pathway in myocardial infarction.
    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:朊病毒疾病是一种传染性和致命性的神经退行性疾病,其特征是错误折叠的朊病毒蛋白亚型(PrPSc)的积累,星形细胞增多症,小胶质细胞增生,海绵状体,和神经变性。细胞膜相关PrPSc蛋白和炎性细胞因子水平的升高提示着死亡受体(DR)途径的激活。DRs的激活调节,细胞存活或凋亡,自噬和坏死基于它们相互作用的衔接子。关于朊病毒疾病中DR途径的激活知之甚少。在正常小鼠大脑中表达的DR3和DR5从未在朊病毒疾病中研究过,它们的配体和任何DR衔接子也是如此。这一研究差距是值得注意的,并在本研究中进行了调查。
    方法:C57BL/6J小鼠用落基山实验室瘙痒病小鼠朊病毒株感染。通过观察形态和行为异常来检查朊病毒病的进展。使用检测印迹和脑切片上的蛋白质的基于抗体的技术,分别测量PrP同种型和GFAP的水平作为PrPSc积累和星形细胞增生的标志物。DR的水平,它们的糖基化和胞外域脱落,和相关因素保证他们在蛋白质水平上的检查,因此,这项研究采用了蛋白质印迹分析。
    结果:感染朊病毒的小鼠出现了运动障碍和神经病理学,如PrPSc积累和星形胶质细胞增多,类似于其他朊病毒疾病。这项研究的结果表明,所有DR配体的高表达,TNFR1、Fas和p75NTR但降低DR3和DR5水平。DR衔接蛋白如TRADD和TRAF2(主要调节促存活途径)的水平降低。FADD,主要调节细胞死亡,其水平保持不变。RIPK1调节促生存,凋亡和坏死,其表达和蛋白水解(抑制坏死但激活凋亡)增加。
    结论:本研究的结果为DR3,DR5,DR6,TL1A的参与提供了证据。TRAIL,TRADD,TRAF2、FADD和RIPK1为首次在朊病毒疾病中的应用。从这项研究中获得的知识讨论了这16种差异表达的DR因子对我们对朊病毒疾病的多方面神经病理学的理解以及对朊病毒疾病特异性神经病理学的潜在靶向治疗干预的未来探索的可能影响。
    BACKGROUND: Prion diseases are transmissible and fatal neurodegenerative diseases characterized by accumulation of misfolded prion protein isoform (PrPSc), astrocytosis, microgliosis, spongiosis, and neurodegeneration. Elevated levels of cell membrane associated PrPSc protein and inflammatory cytokines hint towards the activation of death receptor (DR) pathway/s in prion diseases. Activation of DRs regulate, either cell survival or apoptosis, autophagy and necroptosis based on the adaptors they interact. Very little is known about the DR pathways activation in prion disease. DR3 and DR5 that are expressed in normal mouse brain were never studied in prion disease, so also their ligands and any DR adaptors. This research gap is notable and investigated in the present study.
    METHODS: C57BL/6J mice were infected with Rocky Mountain Laboratory scrapie mouse prion strain. The progression of prion disease was examined by observing morphological and behavioural abnormalities. The levels of PrP isoforms and GFAP were measured as the marker of PrPSc accumulation and astrocytosis respectively using antibody-based techniques that detect proteins on blot and brain section. The levels of DRs, their glycosylation and ectodomain shedding, and associated factors warrant their examination at protein level, hence western blot analysis was employed in this study.
    RESULTS: Prion-infected mice developed motor deficits and neuropathology like PrPSc accumulation and astrocytosis similar to other prion diseases. Results from this research show higher expression of all DR ligands, TNFR1, Fas and p75NTR but decreased levels DR3 and DR5. The levels of DR adaptor proteins like TRADD and TRAF2 (primarily regulate pro-survival pathways) are reduced. FADD, which primarily regulate cell death, its level remains unchanged. RIPK1, which regulate pro-survival, apoptosis and necroptosis, its expression and proteolysis (inhibits necroptosis but activates apoptosis) are increased.
    CONCLUSIONS: The findings from the present study provide evidence towards the involvement of DR3, DR5, DR6, TL1A, TRAIL, TRADD, TRAF2, FADD and RIPK1 for the first time in prion diseases. The knowledge obtained from this research discuss the possible impacts of these 16 differentially expressed DR factors on our understanding towards the multifaceted neuropathology of prion diseases and towards future explorations into potential targeted therapeutic interventions for prion disease specific neuropathology.
    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:四君子汤(SJZD),一种传统的中草药,经常用于治疗各种癌症,包括结肠癌.先前的研究表明,SJZD在调节免疫系统和增强抗肿瘤免疫力方面发挥着关键作用。然而,SJZD在对抗结肠癌中的确切作用及其在调节自然杀伤细胞中的潜在分子功能仍然难以捉摸.
    目的:通过体内和体外实验阐明SJZD与自然杀伤(NK)细胞协同抗癌作用的潜在机制。
    方法:体内实验:进行结肠癌皮下肿瘤小鼠模型和体内NK细胞耗竭实验,以观察SJZD的抗癌作用。流式细胞术评估小鼠脾脏中的免疫细胞耗竭,免疫组化(IHC)染色检测肿瘤组织中凋亡基因的表达。体外实验:使用实时聚合酶链反应(RT-PCR)研究了SJZD调节结肠癌细胞对NK细胞敏感的机制,蛋白质印迹(WB),与NK细胞共培养实验。
    结果:四君子汤(SJZD)显著阻碍小鼠肿瘤生长;NK细胞耗竭显著减弱了SJZD的肿瘤抑制作用。免疫组化(IHC)结果显示,SJZD增加了P53,死亡受体4(DR4)的表达,和肿瘤组织中的逝世亡受体5(DR5)。体外实验,24hSJZD预处理结肠癌细胞显示P53、DR4和DR5水平显著升高,与NK细胞共培养后,结肠癌细胞的活性显着降低。SJZD的这些作用随着P53抑制剂吡虫啉-α(PFT-α)的添加而逆转,导致NK细胞对结肠癌细胞的抑制作用降低。
    结论:SJZD通过调节P53表达增强DR4和DR5的水平,因此增加结肠癌细胞对NK细胞介导的杀伤的敏感性。这些发现为SJZD在结肠癌患者中的临床应用提供了理论依据。在这项研究中,我们首先使用体内试验研究了SJZD对结肠癌小鼠皮下肿瘤生长的影响,并通过NK细胞耗竭评估了NK细胞对SJZD体内抗癌作用的影响.进行了体外实验以探索SJZD在NK细胞介导的抗癌作用中的潜在作用机制。
    BACKGROUND: Sijunzi Decoction (SJZD), a traditional Chinese herbal remedy, is frequently employed in the treatment of various cancers, including colon cancer. Previous research suggests that SJZD plays a pivotal role in modulating the immune system and enhancing immunity against tumors. However, the precise role of SJZD in combating colon cancer and its potential molecular functions in regulating natural killer cells remain elusive.
    OBJECTIVE: To elucidate the potential mechanism underlying the anticolon cancer effects of SJZD in synergy with natural killer (NK) cells through both in vivo and in vitro experiments.
    METHODS: In vivo experiments: A subcutaneous tumor mouse model of colon cancer and in vivo NK cell depletion experiments were conducted to observe the anticolon cancer effects of SJZD. Flow cytometry assessed immune cell depletion in mouse spleens, while immunohistochemical (IHC) staining detected the expression of apoptotic genes in tumor tissues. In vitro experiments: The mechanism by which SJZD regulates the sensitization of colon cancer cells to NK cells was investigated using real-time polymerase chain reaction (RT-PCR), western blotting (WB), and co-culture experiments with NK cells.
    RESULTS: Sijunzi Decoction (SJZD) significantly impeded tumor growth in mice; however, NK cell depletion markedly attenuated the tumor-suppressive effect of SJZD. Immunohistochemical (IHC) results indicated that SJZD increased the expression of P53, death receptor 4 (DR4), and death receptor 5 (DR5) in tumor tissues. In vitro experiments, 24 h SJZD-pretreated colon cancer cells showed a substantial elevation in P53, DR4, and DR5 levels, and the activity of colon cancer cells significantly diminished after co-culture with NK cells. These effects of SJZD were reversed with the addition of the P53 inhibitor pifithrin-α (PFT-α), resulting in reduced inhibition of colon cancer cells by NK cells.
    CONCLUSIONS: SJZD enhances the levels of DR4 and DR5 through the modulation of P53 expression, consequently increasing the sensitivity of colon cancer cells to NK cell-mediated killing. These findings provide a theoretical foundation for the clinical application of SJZD in patients with colon cancer. In this study, we first investigated the effect of SJZD on subcutaneous tumor growth in mice with colon cancer using in vivo assays and assessed the impact of NK cells on the anticolon cancer effect of SJZD in vivo through NK cell depletion. In vitro experiments were conducted to explore the potential mechanism of action of SJZD in NK cell-mediated anticolon cancer 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
    Daurisoline(DS)是一种异喹啉生物碱,在各种癌细胞中发挥抗癌活性。然而,DS影响乳腺癌细胞存活的潜在机制仍知之甚少.因此,本研究旨在研究DS对乳腺癌细胞的潜在抗癌作用,并揭示DS增强的肿瘤坏死因子相关凋亡诱导配体(TRAIL)介导的凋亡的潜在机制。细胞计数试剂盒-8(CCK-8)和5-乙炔基-2-脱氧尿苷(EdU)测定用于评估细胞增殖能力。选择流式细胞术检查细胞周期分布。TUNEL法检测细胞凋亡。通过蛋白质印迹分析测量蛋白质表达。发现DS通过引起G1期细胞周期停滞来降低细胞活力并抑制MCF-7和MDA-MB-231细胞的增殖。DS可以通过促进caspase-8和PARP的裂解来触发细胞凋亡。ERK的磷酸化,JNK,DS治疗后p38MAPK明显上调。值得注意的是,SP600125(JNK抑制剂)预处理显著消除DS诱导的PARP裂解。DS灭活Akt/mTOR和Wnt/β-catenin信号通路并上调ER应激相关蛋白的表达。此外,DS扩增的TRAIL导致乳腺癌细胞的活力降低和凋亡。机械地,DS上调DR4和DR5的蛋白质水平,而DR5的敲低减弱了共治诱导的PARP裂解。抑制JNK可阻断DS诱导的DR5上调。这项研究为DS抑制细胞增殖的机制提供了有价值的见解,触发细胞凋亡,并增强乳腺癌细胞的TRAIL敏感性。
    Daurisoline (DS) is an isoquinoline alkaloid that exerts anticancer activities in various cancer cells. However, the underlying mechanisms through which DS affects the survival of breast cancer cells remain poorly understood. Therefore, the present study was undertaken to investigate the potential anticancer effect of DS on breast cancer cells and reveal the mechanism underlying the enhanced tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated apoptosis by DS. Cell counting kit-8 (CCK-8) and 5-ethynyl-2-deoxyuridine (EdU) assay were used to evaluate the ability of cell proliferation. Flow cytometry was selected to examine the cell cycle distribution. TUNEL assay was used to detect the cell apoptosis. The protein expression was measured by Western blot analysis. DS was found to reduce the cell viability and suppress the proliferation of MCF-7 and MDA-MB-231 cells by causing G1 phase cell cycle arrest. DS could trigger apoptosis by promoting the cleavage of caspase-8 and PARP. The phosphorylation of ERK, JNK, and p38MAPK was upregulated clearly following DS treatment. Notably, SP600125 (JNK inhibitor) pretreatment significantly abrogated DS-induced PARP cleavage. DS inactivated Akt/mTOR and Wnt/β-catenin signaling pathway and upregulated the expression of ER stress-related proteins. Additionally, DS amplified TRAIL-caused viability reduction and apoptosis in breast cancer cells. Mechanismly, DS upregulated the protein level of DR4 and DR5, and knockdown of DR5 attenuated the cotreatment-induced cleavage of PARP. Inhibition of JNK could block DS-induced upregulation of DR5. This study provides valuable insights into the mechanisms of DS inhibiting cell proliferation, triggering apoptosis, and enhancing TRAIL sensitivity of breast 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
    治疗性蛋白质开发的一种常见方法包括采用多价合成配体,实现对信号转导的复杂控制。利用液-液相分离(LLPS)的新兴概念及其将细胞表面受体组织成功能隔室的能力,我们在此设计了具有相分离模式的模块化配体,以设计可编程的受体间通信和信号通路的精确控制,从而诱导快速,强力,和肿瘤细胞的特异性凋亡。尽管简单,这些“触发器”,称为相分离肿瘤杀伤剂(以下简称psTK),足以产生死亡受体(由DR5代表)和肿瘤相关受体的受体间簇,具有显著特点:LLPS介导的稳健高阶组织,精心编排的条件激活,和广谱能力有效诱导肿瘤细胞凋亡。具有相分离模式的新型治疗性蛋白质的开发展示了空间重组信号转导的力量。这种方法促进了细胞命运的多样化,并具有针对挑战性肿瘤的靶向治疗的有希望的潜力。
    A common approach in therapeutic protein development involves employing synthetic ligands with multivalency, enabling sophisticated control of signal transduction. Leveraging the emerging concept of liquid-liquid phase separation (LLPS) and its ability to organize cell surface receptors into functional compartments, we herein have designed modular ligands with phase-separation modalities to engineer programmable interreceptor communications and precise control of signal pathways, thus inducing the rapid, potent, and specific apoptosis of tumor cells. Despite their simplicity, these \"triggers\", named phase-separated Tumor Killers (hereafter referred to as psTK), are sufficient to yield interreceptor clustering of death receptors (represented by DR5) and tumor-associated receptors, with notable features: LLPS-mediated robust high-order organization, well-choreographed conditional activation, and broad-spectrum capacity to potently induce apoptosis in tumor cells. The development of novel therapeutic proteins with phase-separation modalities showcases the power of spatially reorganizing signal transduction. This approach facilitates the diversification of cell fate and holds promising potential for targeted therapies against challenging tumors.
    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:肺癌是世界上发病率和死亡率最高的癌症,对人类健康构成严重威胁。因此,迫切需要发现新的治疗方法来改善肺癌的预后。针对泛素-蛋白酶体系统的小分子抑制剂取得了巨大的成功,其中去泛素酶抑制剂具有广泛的临床应用。据报道,去泛素酶OTUD3通过稳定癌蛋白GRP78促进肺肿瘤发生,这意味着抑制OTUD3可能是肺癌的治疗策略。
    结果:在这项研究中,我们确定了OTUD3的小分子抑制剂,Rolapitin,通过FDA批准的药物库的计算机辅助虚拟筛选和生物实验验证。Rolapitant通过抑制OTUD3的去泛素化活性来抑制肺癌细胞的增殖。定量蛋白质组分析表明,Rolapitin显着上调了死亡受体5(DR5)的表达。Rolapitin还通过上调DR5的细胞表面表达并增强TRAIL诱导的细胞凋亡来促进肺癌细胞凋亡。机械上,Rolapitant直接靶向OTUD3-GRP78轴触发内质网(ER)应激-C/EBP同源蛋白(CHOP)-DR5信号,使肺癌细胞对TRAIL诱导的细胞凋亡敏感。在体内试验中,Rolapitin在合适的剂量下抑制了免疫受损小鼠中肺癌异种移植物的生长,而没有明显的毒性。
    结论:总之,本研究确定Rolapitin为去泛素酶OTUD3的新型抑制剂,并确定OTUD3-GRP78轴是肺癌的潜在治疗靶点.
    BACKGROUND: Lung cancer is cancer with the highest morbidity and mortality in the world and poses a serious threat to human health. Therefore, discovering new treatments is urgently needed to improve lung cancer prognosis. Small molecule inhibitors targeting the ubiquitin-proteasome system have achieved great success, in which deubiquitinase inhibitors have broad clinical applications. The deubiquitylase OTUD3 was reported to promote lung tumorigenesis by stabilizing oncoprotein GRP78, implying that inhibition of OTUD3 may be a therapeutic strategy for lung cancer.
    RESULTS: In this study, we identified a small molecule inhibitor of OTUD3, Rolapitant, by computer-aided virtual screening and biological experimental verification from FDA-approved drugs library. Rolapitant inhibited the proliferation of lung cancer cells by inhibiting deubiquitinating activity of OTUD3. Quantitative proteomic profiling indicated that Rolapitant significantly upregulated the expression of death receptor 5 (DR5). Rolapitant also promoted lung cancer cell apoptosis through upregulating cell surface expression of DR5 and enhanced TRAIL-induced apoptosis. Mechanistically, Rolapitant directly targeted the OTUD3-GRP78 axis to trigger endoplasmic reticulum (ER) stress-C/EBP homologous protein (CHOP)-DR5 signaling, sensitizing lung cancer cells to TRAIL-induced apoptosis. In the vivo assays, Rolapitant suppressed the growth of lung cancer xenografts in immunocompromised mice at suitable dosages without apparent toxicity.
    CONCLUSIONS: In summary, the present study identifies Rolapitant as a novel inhibitor of deubiquitinase OTUD3 and establishes that the OTUD3-GRP78 axis is a potential therapeutic target for lung 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
    尽管免疫原性细胞死亡(ICD)诱导剂明显提高了免疫治疗的有效性,它们的潜能日益受到肿瘤细胞抗凋亡能力发展的制约,免疫原性差,和低T细胞免疫反应。在这项研究中,第一次,压电催化Mg2+掺杂羟基磷灰石(Mg-HAP)纳米粒子,它们涂有介孔二氧化硅层,并负载有ONC201作为激动剂,以特异性靶向肿瘤细胞上的死亡受体DR5,最终开发Mg-HAP@MS/ONC201纳米颗粒(MHMONP)系统,是工程。由于其优异的压电性能,MHMO促进肿瘤细胞内大量活性氧和Ca2+的释放,有效促进DR5表达上调,诱导肿瘤细胞坏死性凋亡,最终克服细胞凋亡抵抗。同时,肿瘤微环境中释放的Mg2+促进CD8+T受体激活,以响应ICD诱导的抗肿瘤免疫反应。使用RNA-seq分析,阐明了MHMO可以在压电催化下激活NF-κB通路,从而诱导M1型巨噬细胞极化。总之,设计了一种在压电催化下同时靶向肿瘤细胞和肿瘤微环境的双靶向治疗系统。该系统对于肿瘤免疫疗法的进步具有巨大的潜力。
    Although immunogenic cell death (ICD) inducers evidently enhance the effectiveness of immunotherapy, their potential is increasingly restricted by the development of apoptosis resistance in tumor cells, poor immunogenicity, and low T-cell immune responsiveness. In this study, for the first time, piezoelectrically catalyzed Mg2+-doped hydroxyapatite (Mg-HAP) nanoparticles, which are coated with a mesoporous silica layer and loaded with ONC201 as an agonist to specifically target the death receptor DR5 on tumor cells, ultimately developing an Mg-HAP@MS/ONC201 nanoparticle (MHMO NP) system, are engineered. Owing to its excellent piezoelectric properties, MHMO facilitates the release of a significant amount of reactive oxygen species and Ca2+ within tumor cells, effectively promoting the upregulation of DR5 expression and inducing tumor cell necroptosis to ultimately overcome apoptosis resistance. Concurrently, Mg2+ released in the tumor microenvironment promotes CD8+ T receptor activation in response to the antitumor immune reaction induced by ICD. Using RNA-seq analysis, it is elucidated that MHMO can activate the NF-κB pathway under piezoelectric catalysis, thus inducing M1-type macrophage polarization. In summary, a dual-targeting therapy system that targets both tumor cells and the tumor microenvironment under piezoelectric catalysis is designed. This system holds substantial potential for advancements in tumor immunotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • DOI:
    文章类型: Journal Article
    胃腺癌通常在无法手术时表现为晚期。化疗方案包括非靶向和毒性药物,导致患者5年生存结果不佳。小分子ONC201/TIC10(TRAIL诱导化合物#10)经由整合应激反应(ISR)的ClpP依赖性活化和TRAIL途径的上调诱导癌细胞死亡。我们以前在乳腺癌中发现,通过ISR依赖性上调ATF4,CHOP和TRAIL死亡受体DR5,ONC201引发肿瘤细胞TRAIL介导的细胞死亡的胰腺癌和子宫内膜癌。我们研究了ONC201与重组人TRAIL(rhTRAIL)或聚乙二醇化三聚体TRAIL(TLY012)联合诱导胃腺癌细胞凋亡的能力。AGS(半胱天冬酶8-,KRAS-,PIK3CA-突变体,HER2扩增),SNU-1(KRAS-,MLH1-突变体,微卫星不稳定),在细胞培养和体内,用ONC201和TRAIL处理SNU-5(p53突变体)和SNU-16(p53突变体)胃腺癌细胞。胃癌细胞在用ONC201和rhTRAIL/TLY012双重治疗后显示出协同作用(在对正常成纤维细胞无毒的剂量下组合指数<0.6)。用双重ONC201加TRAIL疗法观察到细胞周期的亚G1期细胞增加的协同作用。PARP增加,在ONC201加TRAIL后caspase8和caspase3裂解进一步证明了细胞凋亡。通过流式细胞术观察到使用ONC201治疗的DR5的细胞表面表达增加,免疫印迹显示ONC201上调ISR,ATF4和CHOP。我们观察到除AGS外的所有细胞中抗凋亡cIAP-1和XIAP的下调。和cFLIP在除SNU-16之外的所有单元格中。我们在人类胃癌的类器官模型中测试了该方案,以及使用AGS和SNU-1细胞的鼠皮下异种移植物。我们的结果表明,ONC201与TRAIL联合可能是通过激活ISR诱导细胞凋亡治疗胃腺癌的有效且无毒的选择。DR5的细胞表面表达增加和凋亡抑制剂的下调。我们的结果证明了ONC201加TLY012对胃癌的体内抗肿瘤作用,可以在临床试验中进一步研究。
    Gastric adenocarcinoma typically presents with advanced stage when inoperable. Chemotherapy options include non-targeted and toxic agents, leading to poor 5-year patient survival outcomes. Small molecule ONC201/TIC10 (TRAIL-Inducing Compound #10) induces cancer cell death via ClpP-dependent activation of the integrated stress response (ISR) and up-regulation of the TRAIL pathway. We previously found in breast cancer, pancreatic cancer and endometrial cancer that ONC201 primes tumor cells for TRAIL-mediated cell death through ISR-dependent upregulation of ATF4, CHOP and TRAIL death receptor DR5. We investigated the ability of ONC201 to induce apoptosis in gastric adenocarcinoma cells in combination with recombinant human TRAIL (rhTRAIL) or PEGylated trimeric TRAIL (TLY012). AGS (caspase 8-, KRAS-, PIK3CA-mutant, HER2-amplified), SNU-1 (KRAS-, MLH1-mutant, microsatellite unstable), SNU-5 (p53-mutant) and SNU-16 (p53-mutant) gastric adenocarcinoma cells were treated with ONC201 and TRAIL both in cell culture and in vivo. Gastric cancer cells showed synergy following dual therapy with ONC201 and rhTRAIL/TLY012 (combination indices < 0.6 at doses that were non-toxic towards normal fibroblasts). Synergy was observed with increased cells in the sub-G1 phase of the cell cycle with dual ONC201 plus TRAIL therapy. Increased PARP, caspase 8 and caspase 3 cleavage after ONC201 plus TRAIL further documented apoptosis. Increased cell surface expression of DR5 with ONC201 therapy was observed by flow cytometry, and immunoblotting revealed ONC201 upregulation of the ISR, ATF4, and CHOP. We observed downregulation of anti-apoptotic cIAP-1 and XIAP in all cells except AGS, and cFLIP in all cells except SNU-16. We tested the regimen in an organoid model of human gastric cancer, and in murine sub-cutaneous xenografts using AGS and SNU-1 cells. Our results suggest that ONC201 in combination with TRAIL may be an effective and non-toxic option for the treatment of gastric adenocarcinoma by inducing apoptosis via activation of the ISR, increased cell surface expression of DR5 and down-regulation of inhibitors of apoptosis. Our results demonstrate in vivo anti-tumor effects of ONC201 plus TLY012 against gastric cancer that could be further investigated in clinical trials.
    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
    肿瘤坏死因子相关的凋亡诱导配体(TRAIL)是一种有前途的抗癌药物,因为它选择性地诱导转化细胞的凋亡,而不改变健康细胞的细胞机制。不幸的是,TRAIL抗性机制在多种癌症类型中的存在代表了一个主要障碍,因此限制了TRAIL作为单一药物的使用。越来越多的研究表明,TRAIL介导的细胞凋亡可以通过与抗肿瘤药物联合治疗促进耐药肿瘤。从合成分子到天然产物。在后者中,黄酮类化合物,植物中最普遍的多酚,在抗性细胞系以及具有最小副作用的荷瘤小鼠中显示出改善TRAIL驱动的凋亡的显着能力。这里,我们总结了分子机制,例如死亡受体(DR)4和DR5的上调和关键抗凋亡蛋白的下调[例如,细胞FLICE抑制蛋白(c-FLIP),X-连锁凋亡抑制蛋白(XIAP),survivin],不同类别类黄酮的TRAIL敏化特性(例如,黄酮,黄酮醇,异黄酮,查尔酮,异戊二烯类黄酮)。最后,我们讨论局限性,主要与生物利用度问题有关,以及关于黄酮类化合物在TRAIL治疗中作为佐剂的临床应用的未来观点。
    Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) represents a promising anticancer agent, as it selectively induces apoptosis in transformed cells without altering the cellular machinery of healthy cells. Unfortunately, the presence of TRAIL resistance mechanisms in a variety of cancer types represents a major hurdle, thus limiting the use of TRAIL as a single agent. Accumulating studies have shown that TRAIL-mediated apoptosis can be facilitated in resistant tumors by combined treatment with antitumor agents, ranging from synthetic molecules to natural products. Among the latter, flavonoids, the most prevalent polyphenols in plants, have shown remarkable competence in improving TRAIL-driven apoptosis in resistant cell lines as well as tumor-bearing mice with minimal side effects. Here, we summarize the molecular mechanisms, such as the upregulation of death receptor (DR)4 and DR5 and downregulation of key anti-apoptotic proteins [e.g., cellular FLICE-inhibitory protein (c-FLIP), X-linked inhibitor of apoptosis protein (XIAP), survivin], underlying the TRAIL-sensitizing properties of different classes of flavonoids (e.g., flavones, flavonols, isoflavones, chalcones, prenylflavonoids). Finally, we discuss limitations, mainly related to bioavailability issues, and future perspectives regarding the clinical use of flavonoids as adjuvant agents in TRAIL-based therapies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: English Abstract
    Glioma cell cultures are used in basic researches of tumor processes, personalized medicine for selecting treatment regimens depending on individual characteristics of patients and pharmacology for assessing the effectiveness of chemotherapy. Suppression of glioma culture growth without reduction of malignancy grade is common. Drug cancellation may be followed by substitution of precursor cells by more malignant clones. Therefore, analysis of culture cell malignancy grade is important. In the future, intraoperative analysis of glioma cell malignancy grade can be used to select individual therapy.
    We analyzed the relationship between expression of marker genes TUBB3, CD133, CDK4, CDK6, CIRBP, DR4, DR5, EGFR, FGFR, FSHR, GDNF, GFAP, L1CAM, LEF1, MAP2, MDM2, MELK, NANOG, NOTCH2, OCT4, OLIG2, PDGFRA, PDGFA, PDGFB and SOX2 and glioma cell malignancy grade, as well as created appropriate prognostic model.
    We analyzed expression of 25 marker genes in 22 samples of human glioma cultures using quantitative real-time PCR. Statistical analysis was performed using the IBM SPSS Statistics 26.0 software. We used the Kolmogorov-Smirnov and Shapiro-Wilk tests to assess distribution normality. Nonparametric Jonckheere-Terpstra and Spearman tests were applied.
    We obtained a prognostic model for assessing the grade III and IV glioma cell malignancy based on expression of marker genes MDM2, MELK, SOX2, CDK4, DR5 and OCT4. Predictive accuracy was 83% (Akaike information criterion -55.125).
    Культуры глиом имеют широкий спектр применения: в фундаментальных исследованиях опухолевых процессов, персонализированной медицине для подбора схем лечения, учитывающих индивидуальные особенности пациента, а также в фармакологии для оценки эффективности действия лекарственных средств на опухоль. При этом часто отмечается подавление роста культуры глиомы в целом без снижения степени злокачественности. При отмене препаратов может происходить вытеснение пула предшествующих клеток более злокачественными клонами, поэтому важное значение имеет определение степени злокачественности клеток в культуре. В дальнейшем изучение степени злокачественности клеток культуры глиомы, полученной от пациента во время операции, может быть использовано для подбора персонализированной терапии, наилучшим образом подходящей данному пациенту.
    Изучение связи между экспрессией маркерных генов TUBB3, CD133, CDK4, CDK6, CIRBP, DR4, DR5, EGFR, FGFR, FSHR, GDNF, GFAP, L1CAM, LEF1, MAP2, MDM2, MELK, NANOG, NOTCH2, OCT4, OLIG2, PDGFRA, PDGFA, PDGFB и SOX2 и степенью злокачественности клеток в культурах глиом и создание прогностической модели в виде уравнения на основе регрессионного анализа.
    С помощью количественной полимеразной цепной реакции в реальном времени была проанализирована экспрессия 25 маркерных генов в 22 образцах культур глиом человека, полученных от пациентов, проходивших лечение в ФГАУ «Национальный медицинский исследовательский центр нейрохирургии им. акад. Н.Н. Бурденко» Минздрава России. При статистической обработке полученных данных, проведенной с помощью программного обеспечения IBM SPSS Statistics 26.0 (StatSoft Inc., США), для оценки нормальности распределения анализировали критерии Колмогорова—Смирнова и Шапиро—Уилка, при сравнительной статистике использовали непараметрические критерии Джонкхиера—Терпстры и Спирмена.
    Была получена прогностическая модель для определения степени злокачественности клеток в культурах глиом III и IV Grade с помощью оценки экспрессии маркерных генов MDM2, MELK, SOX2, CDK4, DR5 и OCT4 с прогностической точностью 83% (информационный критерий Акаике равен —55,125).
    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