glioma stem-like cells

胶质瘤干细胞样细胞
  • 文章类型: Journal Article
    最近的研究表明,PTPRZ1-MET(ZM)融合在胶质瘤向多形性胶质母细胞瘤(GBM)的进展中起着关键作用。因此用作区分原发性GBM和继发性GBM(sGBM)的生物标志物。然而,ZM融合影响这一进展的机制仍有待阐明。与没有ZM的GBM相比,具有ZM的GBM显示出较差的预后和更大的肿瘤相关巨噬细胞(TAM)浸润。胶质瘤干细胞样细胞(GSCs)和TAMs在胶质瘤复发中起着复杂的作用,胶质瘤进展和治疗抵抗。在这项研究中,我们分析了有或没有ZM融合的sGBM患者神经胶质瘤组织的RNA-seq数据,发现在携带ZM的sGBM患者中,干性和巨噬细胞标志物的表达高于没有ZM融合的患者。ZM增强GSCs的自我更新和增殖,从而加速神经胶质瘤的进展.此外,ZM阳性GSC促进了TAM的浸润,并将其极化朝向免疫抑制表型,这主要是通过ISG20的细胞外分泌来实现的。我们的研究确定了GSC内的MET-STAT3-ISG20轴,从而证明ZM在GBM启动和进展中的关键作用。我们的研究表明,与ZM阳性分化神经胶质瘤细胞相反,ZM阳性GSC通过MET-STAT3-ISG20轴上调ISG20表达。细胞外分泌的ISG20募集并诱导巨噬细胞M2样极化,从而促进肿瘤进展。我们的研究结果揭示了ZM阳性GBM发病机制的新机制,并确定了潜在的治疗靶点。
    Recent studies have revealed that PTPRZ1-MET (ZM) fusion plays a pivotal role in the progression of glioma to glioblastoma multiforme (GBM), thus serving as a biomarker to distinguish between primary GBM and secondary GBM (sGBM). However, the mechanisms through which ZM fusion influences this progression remain to be elucidated. GBMs with ZM showed poorer prognoses and greater infiltration of tumor-associated macrophages (TAMs) than those without ZM. Glioma stem-like cells (GSCs) and TAMs play complex roles in glioma recurrence, glioma progression and therapy resistance. In this study, we analyzed RNA-seq data from sGBM patients\' glioma tissues with or without ZM fusion, and found that stemness and macrophage markers were more highly expressed in sGBM patients harboring ZM than in those without ZM fusion. ZM enhanced the self-renewal and proliferation of GSCs, thereby accelerating glioma progression. In addition, ZM-positive GSCs facilitated the infiltration of TAMs and drove their polarization toward an immunosuppressive phenotype, which was primarily accomplished through the extracellular secretion of ISG20. Our research identified the MET-STAT3-ISG20 axis within GSCs, thus demonstrating the critical role of ZM in GBM initiation and progression. Our study demonstrated that, in contrast to ZM-positive differentiated glioma cells, ZM-positive GSCs upregulated ISG20 expression through the MET-STAT3-ISG20 axis. The extracellular secretion of ISG20 recruited and induced M2-like polarization in macrophages, thereby promoting tumor progression. Our results reveal a novel mechanism involved in ZM-positive GBM pathogenesis and identify potential therapeutic targets.
    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
    胶质母细胞瘤是一种侵袭性的,尽管采用手术的多模式治疗,但无法治愈的脑癌的五年生存率仍低于13%,DNA损伤性放化疗和最近增加的肿瘤治疗领域(TTFields)。因此,迫切需要使用更多临床和手术相关的模型来提高我们对TTField细胞反应的当前理解,这反映了胶质母细胞瘤内部深刻的空间异质性,并利用这些生物学见解,为合理设计结合TTFields的更有效的治疗策略提供信息。我们最近报道了在2D神经胶质瘤干细胞样细胞(GSC)模型中使用inovitroTM系统的临床前TTFelds,并与一系列治疗批准和临床前DNA损伤反应抑制剂(DDRi)和放化疗共同应用时,显示出显着的细胞毒性增强。在这里,我们报告了在更具临床相关性的基于3D支架的空间异质性的主要GSC模型中,临床前TTFelds交付的开发和优化。并强调替莫唑胺和临床批准的PARP抑制剂(PARPi)对TTFields效力的初步增强。这些研究,因此,代表了在临床相关的3DGSC模型中进一步评估基于TTField的治疗策略的重要平台,旨在加速临床试验的实施和提高这些患者的持续生存率的最终目标。
    Glioblastoma is an aggressive, incurable brain cancer with poor five-year survival rates of around 13% despite multimodal treatment with surgery, DNA-damaging chemoradiotherapy and the recent addition of Tumour Treating Fields (TTFields). As such, there is an urgent need to improve our current understanding of cellular responses to TTFields using more clinically and surgically relevant models, which reflect the profound spatial heterogeneity within glioblastoma, and leverage these biological insights to inform the rational design of more effective therapeutic strategies incorporating TTFields. We have recently reported the use of preclinical TTFields using the inovitroTM system within 2D glioma stem-like cell (GSC) models and demonstrated significant cytotoxicity enhancement when co-applied with a range of therapeutically approved and preclinical DNA damage response inhibitors (DDRi) and chemoradiotherapy. Here we report the development and optimisation of preclinical TTFields delivery within more clinically relevant 3D scaffold-based primary GSC models of spatial heterogeneity, and highlight some initial enhancement of TTFields potency with temozolomide and clinically approved PARP inhibitors (PARPi). These studies, therefore, represent an important platform for further preclinical assessment of TTFields-based therapeutic strategies within clinically relevant 3D GSC models, aimed towards accelerating clinical trial implementation and the ultimate goal of improving the persistently dire survival rates for these 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:缺氧与包括胶质母细胞瘤(GBM)在内的许多癌症的不良预后相关。神经胶质瘤干细胞样细胞(GSC)通常位于缺氧区域,并作为疾病进展的储库。长非编码RNA(lncRNA)已经与GBM有关。然而,对调节GSC适应缺氧的lncRNAs了解甚少。这些lncRNAs的鉴定可能为在缺氧条件下靶向GSCs提供新的治疗策略。
    方法:通过RNAseq鉴定缺氧诱导的GSCs中的lncRNAs。通过qPCR评估LUCAT1表达,RNAseq,北方印迹,GSC中的单分子FISH,在IvyGAP审讯,TCGA,CGGA数据库。LUCAT1被shRNA耗尽,CRISPR/Cas9和CRISPR/Cas13d。RNAseq,蛋白质印迹,免疫组织化学,co-IP,ChIP,ChIPseq,RNA免疫沉淀,进行了邻近连接试验以研究LUCAT1的作用机制。GSC生存能力,有限稀释试验,和原位GBM异种移植模型中的致瘤潜力进行评估,以评估消耗LUCAT1的功能后果.
    结果:HIF1α和NRF2在缺氧条件下GSCs中诱导Lucat1的新同工型。LUCATl在GBM中的低氧区域中高度表达。机械上,LUCAT1与HIF1α及其共激活剂CBP形成复合物,以调节HIF1α靶基因表达和GSC对缺氧的适应。LUCAT1的耗尽损害了GSC的自我更新。在GBM异种移植模型中沉默LUCAT1降低肿瘤生长并延长小鼠存活。
    结论:HIF1α-LUCAT1轴形成一个正反馈回路,在缺氧条件下扩增GSCs中的HIF1α信号。LUCAT1促进GSC自我更新和GBM肿瘤生长。LUCAT1是GBM的潜在治疗靶点。
    BACKGROUND: Hypoxia is associated with poor prognosis in many cancers including glioblastoma (GBM). Glioma stem-like cells (GSCs) often reside in hypoxic regions and serve as reservoirs for disease progression. Long non-coding RNAs (lncRNAs) have been implicated in GBM. However, the lncRNAs that modulate GSC adaptations to hypoxia are poorly understood. Identification of these lncRNAs may provide new therapeutic strategies to target GSCs under hypoxia.
    METHODS: lncRNAs induced by hypoxia in GSCs were identified by RNA-seq. Lung cancer-associated transcript-1 (LUCAT1) expression was assessed by qPCR, RNA-seq, Northern blot, single molecule FISH in GSCs, and interrogated in IvyGAP, The Cancer Genome Atlas, and CGGA databases. LUCAT1 was depleted by shRNA, CRISPR/Cas9, and CRISPR/Cas13d. RNA-seq, Western blot, immunohistochemistry, co-IP, ChIP, ChIP-seq, RNA immunoprecipitation, and proximity ligation assay were performed to investigate mechanisms of action of LUCAT1. GSC viability, limiting dilution assay, and tumorigenic potential in orthotopic GBM xenograft models were performed to assess the functional consequences of depleting LUCAT1.
    RESULTS: A new isoform of Lucat1 is induced by Hypoxia inducible factor 1 alpha (HIF1α) and Nuclear factor erythroid 2-related factor 2 (NRF2) in GSCs under hypoxia. LUCAT1 is highly expressed in hypoxic regions in GBM. Mechanistically, LUCAT1 formed a complex with HIF1α and its co-activator CBP to regulate HIF1α target gene expression and GSC adaptation to hypoxia. Depletion of LUCAT1 impaired GSC self-renewal. Silencing LUCAT1 decreased tumor growth and prolonged mouse survival in GBM xenograft models.
    CONCLUSIONS: A HIF1α-LUCAT1 axis forms a positive feedback loop to amplify HIF1α signaling in GSCs under hypoxia. LUCAT1 promotes GSC self-renewal and GBM tumor growth. LUCAT1 is a potential therapeutic target in GBM.
    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
    胶质母细胞瘤是成年人中最常见的原发性脑癌,是患者诊断最差的癌症之一。患有不良预后和有限的治疗选择,肿瘤复发几乎是不可避免的。此外,治疗耐药是很常见的这种疾病和恶化的预后。假设这些和其他因素主要是由于已知胶质母细胞瘤细胞能够获得干细胞样特征,从而驱动这些表型。最近,我们已经证明,用激素活性形式的维生素D3,骨化三醇(1α,25(OH)2-维生素D3)可以阻断细胞系子集的干性并减少肿瘤生长。这里,我们将细胞面板扩展到40多种不同的培养物,可以证明,虽然一半的测试细胞系是敏感的,四分之一可以归类为高反应者。利用遗传和蛋白质组学分析,我们进一步确定,治疗成功可以部分解释为维生素D3受体的特定多态性,并且高反应者显示出提示阻断干性的蛋白质组,以及迁徙潜力。
    Glioblastoma is the most common primary brain cancer in adults and represents one of the worst cancer diagnoses for patients. Suffering from a poor prognosis and limited treatment options, tumor recurrences are virtually inevitable. Additionally, treatment resistance is very common for this disease and worsens the prognosis. These and other factors are hypothesized to be largely due to the fact that glioblastoma cells are known to be able to obtain stem-like traits, thereby driving these phenotypes. Recently, we have shown that the in vitro and ex vivo treatment of glioblastoma stem-like cells with the hormonally active form of vitamin D3, calcitriol (1α,25(OH)2-vitamin D3) can block stemness in a subset of cell lines and reduce tumor growth. Here, we expanded our cell panel to over 40 different cultures and can show that, while half of the tested cell lines are sensitive, a quarter can be classified as high responders. Using genetic and proteomic analysis, we further determined that treatment success can be partially explained by specific polymorphism of the vitamin D3 receptor and that high responders display a proteome suggestive of blockade of stemness, as well as migratory potential.
    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
    胶质母细胞瘤是一种非常侵袭性的肿瘤,是最常见的原发性脑恶性肿瘤。主要特征包括对常规治疗的高抵抗力,如放疗和化疗及其弥漫性组织浸润,防止完全手术切除。对生理微环境中的迁移和侵入过程的分析允许增强对这些现象的理解,并且可以导致改进的治疗方法。这里,我们结合了两种最先进的技术,成人器官型脑组织切片培养(OTC)和光片荧光显微镜(LSFM)在称为OTCxLSFM的组合方法中清除组织。使用这种方法,我们可以证明,胶质母细胞瘤的组织浸润可以通过三氧化二砷或WP1066的治疗有效阻断,以及四跨膜蛋白的遗传耗竭,跨膜受体CD9,或信号转导和转录激活因子3(STAT3)。有了我们的分析管道,我们获得单细胞水平,三维信息,以及对肿瘤细胞形态外观的见解。
    Glioblastoma is a very aggressive tumor and represents the most common primary brain malignancy. Key characteristics include its high resistance against conventional treatments, such as radio- and chemotherapy and its diffuse tissue infiltration, preventing complete surgical resection. The analysis of migration and invasion processes in a physiological microenvironment allows for enhanced understanding of these phenomena and can lead to improved therapeutic approaches. Here, we combine two state-of-the-art techniques, adult organotypic brain tissue slice culture (OTC) and light-sheet fluorescence microscopy (LSFM) of cleared tissues in a combined method termed OTCxLSFM. Using this methodology, we can show that glioblastoma tissue infiltration can be effectively blocked through treatment with arsenic trioxide or WP1066, as well as genetic depletion of the tetraspanin, transmembrane receptor CD9, or signal transducer and activator of transcription 3 (STAT3). With our analysis pipeline, we gain single-cell level, three-dimensional information, as well as insights into the morphological appearance of the tumor cells.
    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:我们先前的研究发现XHP可以诱导GBM细胞发生凋亡。大量证据表明,神经胶质瘤干细胞样细胞(GSCs)是导致多形性胶质母细胞瘤(GBM)疾病进展和预后不良的关键因素。中医药已作为脑胶质瘤的补充和替代疗法应用于临床。
    目的:评价西黄丸对GSCs的作用及可能的分子机制。
    方法:UPLC-QTOF-MS分析用于XHP的组分分析。利用网络药理学和生物信息学方法,通过XHP中的活性成分构建靶向GSCs的分子网络。细胞活力,自我更新能力,凋亡,用CCK-8法检测GSC标志物,肿瘤球形成测定和流式细胞术,分别。使用GEPIA评估GSC标志物(CD133和SOX2)与EGFR/Akt/mTOR信号通路关键蛋白之间的相互关系,并通过蛋白质印迹进行验证。使用慢病毒构建稳定过表达Akt的GBM细胞系以评估Akt信号传导在神经胶质瘤干性调节中的作用。通过裸鼠皮下移植胶质瘤细胞模型分析XHP对胶质瘤生长的影响,苏木精-伊红染色用于检查病理变化,TUNEL染色用于检测肿瘤组织中的细胞凋亡,用免疫印迹法和免疫荧光法检测肿瘤组织中GSC标志物的表达。
    结果:生物信息学分析表明,55个匹配的靶标与XHP靶标和神经胶质瘤干细胞靶标相关。除了引起细胞凋亡,XHP可以减少GBM3D球体的数量,CD133阳性细胞的比例和体外GSC标志物(CD133和SOX2)的表达水平。此外,XHP可以减弱CD133、EGFR、p-Akt,GBM领域中的p-mTOR和SOX2。Akt的过表达显著增加SOX2的表达水平,这在XHP存在下被禁止。XHP减少了GSC标记,包括CD133和SOX2,并阻碍了体内异种移植小鼠模型中神经胶质瘤的生长。
    结论:我们首次证明XHP下调干性,通过抑制CD133/EGFR/Akt/mTOR级联的稳定性下调SOX2,抑制GSCs的自我更新并诱导细胞凋亡,并阻碍神经胶质瘤的生长。
    BACKGROUND: Our previous study found that XHP could induce GBM cells to undergo apoptosis. A lot of evidence suggests that glioma stem-like cells (GSCs) are key factors that contribute to disease progression and poor prognosis of glioblastoma multiforme (GBM). Traditional Chinese medicine has been applied in clinical practice as a complementary and alternative therapy for glioma.
    OBJECTIVE: To evaluate the effect and the potential molecular mechanism of Xihuang pill (XHP) on GSCs.
    METHODS: UPLC-QTOF-MS analysis was used for constituent analysis of XHP. Using network pharmacology and bioinformatics methods, a molecular network targeting GSCs by the active ingredients in XHP was constructed. Cell viability, self-renewal ability, apoptosis, and GSC markers were detected by CCK-8 assay, tumor sphere formation assay and flow cytometry, respectively. The interrelationship between GSC markers (CD133 and SOX2) and key proteins of the EGFR/Akt/mTOR signaling pathway was evaluated using GEPIA and verified by western blot. A GBM cell line stably overexpressing Akt was constructed using lentivirus to evaluate the role of Akt signaling in the regulation of glioma stemness. The effect of XHP on glioma growth was analyzed by a subcutaneously transplanted glioma cell model in nude mice, hematoxylin-eosin staining was used to examine pathological changes, TUNEL staining was used to detect apoptosis in tumor tissues, and the expression of GSC markers in tumor tissues was identified by western blot and immunofluorescence.
    RESULTS: Bioinformatics analysis showed that 55 matched targets were related to XHP targets and glioma stem cell targets. In addition to causing apoptosis, XHP could diminish the number of GBM 3D spheroids, the proportion of CD133-positive cells and the expression level of GSC markers (CD133 and SOX2) in vitro. Furthermore, XHP could attenuate the expression of CD133, EGFR, p-Akt, p-mTOR and SOX2 in GBM spheres. Overexpression of Akt significantly increased the expression level of SOX2, which was prohibited in the presence of XHP. XHP reduced GSC markers including CD133 and SOX2, and impeded the development of glioma growth in xenograft mouse models in vivo.
    CONCLUSIONS: We demonstrate for the first time that XHP down-regulates stemness, restrains self-renewal and induces apoptosis in GSCs and impedes glioma growth by down-regulating SOX2 through destabilizing the CD133/EGFR/Akt/mTOR cascade.
    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
    胶质母细胞瘤(GBM)仍然是大脑中最具侵袭性的肿瘤之一,尽管付出了巨大的研究努力,今天仍然无法治愈。随着许多理论围绕这种恶性肿瘤的持续复发发展,具有干细胞样表型的小细胞群的假设仍然是其浸润性质的关键驱动因素.在这篇文章中,我们研究了Chordin样1(CHRDL1),分泌的蛋白质,作为神经胶质瘤干细胞样细胞(GSC)表型的潜在关键调节因子。已经证明CHRDL1拮抗骨形态发生蛋白4(BMP4)的功能,诱导GSC分化,因此,减少致瘤性。我们,因此,使用两个先前描述的GSC球体培养物,并使用CHRDL1靶向shRNA的稳定转导来耗尽CHRDL1。我们展示了体外基于细胞的测定法(MTT,限制稀释,和球体形成测定),西方印迹,辐照程序,和定量实时PCR,分泌的BMP4拮抗剂CHRDL1的消耗显着降低了功能和分子干性性状,从而增强了辐射敏感性。因此,我们假设CHRDL1是GSCs的干性执行者,并发现更多的证据表明CHRDL1的高表达也可能作为确定BMP4易感性的标记蛋白.
    Glioblastoma (GBM) still presents as one of the most aggressive tumours in the brain, which despite enormous research efforts, remains incurable today. As many theories evolve around the persistent recurrence of this malignancy, the assumption of a small population of cells with a stem-like phenotype remains a key driver of its infiltrative nature. In this article, we research Chordin-like 1 (CHRDL1), a secreted protein, as a potential key regulator of the glioma stem-like cell (GSC) phenotype. It has been shown that CHRDL1 antagonizes the function of bone morphogenic protein 4 (BMP4), which induces GSC differentiation and, hence, reduces tumorigenicity. We, therefore, employed two previously described GSCs spheroid cultures and depleted them of CHRDL1 using the stable transduction of a CHRDL1-targeting shRNA. We show with in vitro cell-based assays (MTT, limiting dilution, and sphere formation assays), Western blots, irradiation procedures, and quantitative real-time PCR that the depletion of the secreted BMP4 antagonist CHRDL1 prominently decreases functional and molecular stemness traits resulting in enhanced radiation sensitivity. As a result, we postulate CHRDL1 as an enforcer of stemness in GSCs and find additional evidence that high CHRDL1 expression might also serve as a marker protein to determine BMP4 susceptibility.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 长链非编码RNA(lncRNA)最近被揭示为主要的调控分子,这牵涉到许多细胞功能。研究表明,lncRNA异常表达并参与神经胶质瘤的发展和肿瘤发生。本研究确定了一种与神经胶质瘤相关的新型lncRNA,神经胶质瘤干细胞样细胞(GSC),然后揭示了它们的潜在功能。在lncRNAs的筛选过程中,我们研究了与神经胶质瘤细胞相比,GSC中lncRNARP5-821D11.7(lncRNA-RP5)的过表达。构建lncRNA-RP5的慢病毒介导的shRNA并将其转染到神经胶质瘤细胞中。将转染的稳定神经胶质瘤细胞移植到裸鼠中并观察肿瘤生长。敲低lncRNA-RP5显著抑制增殖,菌落形成,迁移并通过激活Wnt/β-catenin途径减少上皮-间质转化(EMT)。此外,结果表明,lncRNARP5敲低通过内质网应激促进细胞凋亡。因此,这项研究可能提供对lncRNA-RP5的更好理解,这揭示了lncRNA-RP5在胶质瘤进展和复发的情况下可能是一个潜在的治疗靶点.
    Long noncoding RNA (lncRNA) has been recently revealed as a main regulatory molecule, which implicates many cellular functions. Studies showed that lncRNA abnormally expressed and involved in the progression and tumorigenesis of glioma. Present study identified a novel lncRNA associated with glioma, glioma stem-like cells (GSCs), and then revealed their potential functions. During the screening of lncRNAs, we investigated overexpression of lncRNA RP5-821D11.7 (lncRNA-RP5) in GSCs compared to glioma cells. Lentivirus-mediated shRNA for lncRNA-RP5 was constructed and transfected into glioma cells. Transfected stable glioma cells were transplanted into nude mice and tumor growth was observed. Knockdown of lncRNA-RP5 significantly inhibits proliferation, colony formation, migration and reduces epithelial-mesenchymal transition (EMT) by activating the Wnt/β-catenin pathway. Additionally, the results showed that lncRNA RP5 knockdown enhances cell apoptosis through endoplasmic reticulum stress. Therefore, this study may provide a better understanding about lncRNA-RP5 which revealed that it might be a potential therapeutic target in case of glioma progression and recurrence.
    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
    MEOX2转录因子的高表达与胶质瘤总体生存率低密切相关。MEOX2最近被描述为一个有趣的预后生物标志物,尤其是低级别的神经胶质瘤。MEOX2从未在神经胶质瘤干细胞样细胞(GSC)中进行过研究,负责胶质瘤复发。本研究旨在探讨MEOX2在GSC中的作用。使用siRNA的功能丧失方法用于评估MEOX2对GSC活力和干性表型的影响。MEOX2位于细胞核中,其表达在GSC之间是异质的。MEOX2表达取决于其启动子的甲基化状态,并且与IDH突变密切相关。MEOX2通过ERK/MAPK和PI3K/AKT途径参与细胞增殖和活力调节。MEOX2功能丧失与GSC分化和神经元谱系特征的获得相关。此外,MEOX2的抑制与CDH10的表达增加和pFAK的降低相关。在这项研究中,我们解开了,第一次,MEOX2通过AKT/ERK途径促进细胞活力和增殖,并可能参与GSC的表型和粘附特性。
    The high expression of MEOX2 transcription factor is closely associated with poor overall survival in glioma. MEOX2 has recently been described as an interesting prognostic biomarker, especially for lower grade glioma. MEOX2 has never been studied in glioma stem-like cells (GSC), responsible for glioma recurrence. The aim of our study was to investigate the role of MEOX2 in GSC. Loss of function approach using siRNA was used to assess the impact of MEOX2 on GSC viability and stemness phenotype. MEOX2 was localized in the nucleus and its expression was heterogeneous between GSCs. MEOX2 expression depends on the methylation state of its promoter and is strongly associated with IDH mutations. MEOX2 is involved in cell proliferation and viability regulation through ERK/MAPK and PI3K/AKT pathways. MEOX2 loss of function correlated with GSC differentiation and acquisition of neuronal lineage characteristics. Besides, inhibition of MEOX2 is correlated with increased expression of CDH10 and decreased pFAK. In this study, we unraveled, for the first time, MEOX2 contribution to cell viability and proliferation through AKT/ERK pathway and its potential involvement in phenotype and adhesion properties of GSC.
    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
    胶质母细胞瘤治疗的困难使其成为新疗法的良好候选者,如溶瘤病毒。表达拉沙病毒糖蛋白(Lassa-VSV)的水泡性口炎病毒在使用已建立的胶质母细胞瘤细胞系的动物模型中显示出重大希望。这些实验是为了确定低通道的敏感性,患者来源的细胞系对Lassa-VSV的溶胞作用。用表达绿色荧光蛋白(GFP)的Lassa-VSV感染四种患者来源的胶质母细胞瘤细胞系,并通过荧光显微镜进行分析,流式细胞术,和细胞活力测定。还将细胞分析为主要含有神经胶质瘤干细胞样细胞的肿瘤球。三个低通道,采用RNA测序(RNA-seq)进一步分析患者来源的细胞.单个细胞系的病毒基因表达水平和Lassa-VSV诱导的细胞死亡的时间过程有所不同。但每个人都容易感染Lassa-VSV.卓越脑肿瘤中心(BTCOE)4765细胞的干扰素刺激基因表达水平最高,但最容易受到Lassa-VSV诱导的细胞死亡,表明更易感的细胞不一定具有较低的干扰素途径激活。培养为肿瘤球并感染Lassa-VSV的细胞也显示出对Lassa-VSV的可变易感性,但BTCOE4765细胞最不敏感。因此,患者来源的脑肿瘤细胞对Lassa-VSV感染表现出不同的反应,但是每个品系都容易受到VSV溶瘤的影响。
    The difficulty of glioblastoma treatment makes it a good candidate for novel therapies, such as oncolytic viruses. Vesicular stomatitis virus expressing Lassa virus glycoprotein (Lassa-VSV) showed significant promise in animal models using established glioblastoma cell lines. These experiments were to determine the susceptibility of low-passage, patient-derived cell lines to Lassa-VSV oncolysis. Four patient-derived glioblastoma cell lines were infected with Lassa-VSV that expresses green fluorescent protein (GFP) and analyzed by fluorescence microscopy, flow cytometry, and cell viability assays. Cells were also analyzed as tumorspheres containing primarily glioma stem-like cells. Three low-passage, patient-derived cells were further analyzed with RNA sequencing (RNA-seq). Individual cell lines varied somewhat in their levels of viral gene expression and time course of Lassa-VSV-induced cell death, but each was susceptible to Lassa-VSV. Brain Tumor Center of Excellence (BTCOE) 4765 cells had the highest level of expression of interferon-stimulated genes but were most susceptible to Lassa-VSV-induced cell death, indicating that more susceptible cells do not necessarily have lower interferon pathway activation. Cells cultured as tumorspheres and infected with Lassa-VSV also showed variable susceptibility to Lassa-VSV, but BTCOE 4765 cells were least susceptible. Thus, patient-derived brain tumor cells show variable responses to Lassa-VSV infection, but each of the lines was susceptible to VSV oncolysis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号