Temozolomide resistance

  • 文章类型: Journal Article
    胶质母细胞瘤(GBM)由于其对替莫唑胺(TMZ)的抗性而提出了艰巨的挑战,由前脑膜(PN)向间充质(MES)表型的前脑膜向间充质转化(PMT)加剧的障碍。TAGLN2在GBM中突出表达,特别是在MES亚型中,与低度胶质瘤(LGG)和PN亚型相比。我们的研究通过一系列体外和体内实验揭示了TAGLN2参与PMT和TMZ抗性。TAGLN2敲低可以克制增殖和侵袭,引发DNA损伤和细胞凋亡,并提高GBM细胞中的TMZ灵敏度。相反,在细胞和颅内异种移植小鼠模型中,升高TAGLN2水平放大对TMZ的抗性。我们通过免疫共沉淀(Co-IP)和液相色谱-串联质谱(LC-MS/MS)分析证明了TAGLN2和ERK1/2之间的相互作用关系。TAGLN2的敲低导致p-ERK1/2的表达减少,而TAGLN2的过表达导致核内p-ERK1/2的表达增加。随后,已经证明了TAGLN2在MGMT的表达和控制中的调节作用。最后,NF-κB对TAGLN2的调节已通过染色质免疫沉淀和ChIP-PCR测定得到验证。总之,我们的结果证实TAGLN2通过与ERK/MGMT轴相互作用并受NF-κB调节而发挥其生物学功能,从而促进在胶质母细胞瘤中获得促进PMT并增加对TMZ治疗的抗性。这些结果为在临床治疗中克服TMZ抗性的靶向治疗方法的进展提供了有价值的见解。
    Glioblastoma (GBM) presents a daunting challenge due to its resistance to temozolomide (TMZ), a hurdle exacerbated by the proneural-to-mesenchymal transition (PMT) from a proneural (PN) to a mesenchymal (MES) phenotype. TAGLN2 is prominently expressed in GBM, particularly in the MES subtype compared to low-grade glioma (LGG) and the PN subtype. Our research reveals TAGLN2\'s involvement in PMT and TMZ resistance through a series of in vitro and in vivo experiments. TAGLN2 knockdown can restrain proliferation and invasion, trigger DNA damage and apoptosis, and heighten TMZ sensitivity in GBM cells. Conversely, elevating TAGLN2 levels amplifies resistance to TMZ in cellular and intracranial xenograft mouse models. We demonstrate the interaction relationship between TAGLN2 and ERK1/2 through co-immunoprecipitation (Co-IP) and liquid chromatography-tandem mass spectrometry (LC-MS/MS) spectrometry analysis. Knockdown of TAGLN2 results in a decrease in the expression of p-ERK1/2, whereas overexpression of TAGLN2 leads to an increase in p-ERK1/2 expression within the nucleus. Subsequently, the regulatory role of TAGLN2 in the expression and control of MGMT has been demonstrated. Finally, the regulation of TAGLN2 by NF-κB has been validated through chromatin immunoprecipitation and ChIP-PCR assays. In conclusion, our results confirm that TAGLN2 exerts its biological functions by interacting with the ERK/MGMT axis and being regulated by NF-κB, thereby facilitating the acquisition of promoting PMT and increased resistance to TMZ therapy in glioblastoma. These results provide valuable insights for the advancement of targeted therapeutic approaches to overcome TMZ resistance in clinical treatments.
    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
    多形性胶质母细胞瘤(GBM)是一种侵袭性脑癌,比其他脑肿瘤发生频率更高。本研究旨在通过生物信息学和湿实验室分析揭示GBM中替莫唑胺耐药的新机制。包括meta-Z分析,Kaplan-Meier生存分析,蛋白质-蛋白质相互作用(PPI)网络的建立,共表达基因网络的聚类分析,以及上调和下调基因的分层聚类。下一代测序和定量PCR分析显示下调[具有免疫球蛋白和表皮生长因子同源结构域1(TIE1)的酪氨酸激酶,钙电压门控通道辅助亚基α2Δ1(CACNA2D1),钙蛋白酶6(CAPN6)和具有血小板反应蛋白基序6(ADAMTS6)]的整合素和金属蛋白酶,并上调[血清淀粉样蛋白(SA)A1,SAA2,生长分化因子15(GDF15)和泛素特异性肽酶26(USP26)]基因。使用P值转换的Z分数为这些基因开发了不同的统计模型,和Kaplan-Meier图使用几个脑肿瘤患者队列构建。在PPI网络中观察到的节点数量最多的是ADAMTS6和TIE1。所有基因的PPI网络模型包含35个节点和241条边。使用来自患者的异柠檬酸脱氢酶(IDH)-野生型或IDH-突变型GBM样品进行免疫组织化学染色,并且与IDH-野生型GBM相比,在IDH-突变型GBM中证实了TIE1(P<0.001)和CAPN6(P<0.05)蛋白表达的显著上调。结构分析揭示了IDH-突变体模型,证明了突变体残基(R132、R140和R172)。本研究的发现将有助于脑肿瘤的新型生物标志物和治疗方法的未来发展。
    Glioblastoma multiforme (GBM) is an aggressive brain cancer that occurs more frequently than other brain tumors. The present study aimed to reveal a novel mechanism of temozolomide resistance in GBM using bioinformatics and wet lab analyses, including meta-Z analysis, Kaplan-Meier survival analysis, protein-protein interaction (PPI) network establishment, cluster analysis of co-expressed gene networks, and hierarchical clustering of upregulated and downregulated genes. Next-generation sequencing and quantitative PCR analyses revealed downregulated [tyrosine kinase with immunoglobulin and epidermal growth factor homology domains 1 (TIE1), calcium voltage-gated channel auxiliary subunit α2Δ1 (CACNA2D1), calpain 6 (CAPN6) and a disintegrin and metalloproteinase with thrombospondin motifs 6 (ADAMTS6)] and upregulated [serum amyloid (SA)A1, SAA2, growth differentiation factor 15 (GDF15) and ubiquitin specific peptidase 26 (USP26)] genes. Different statistical models were developed for these genes using the Z-score for P-value conversion, and Kaplan-Meier plots were constructed using several patient cohorts with brain tumors. The highest number of nodes was observed in the PPI network was for ADAMTS6 and TIE1. The PPI network model for all genes contained 35 nodes and 241 edges. Immunohistochemical staining was performed using isocitrate dehydrogenase (IDH)-wild-type or IDH-mutant GBM samples from patients and a significant upregulation of TIE1 (P<0.001) and CAPN6 (P<0.05) protein expression was demonstrated in IDH-mutant GBM in comparison with IDH-wild-type GBM. Structural analysis revealed an IDH-mutant model demonstrating the mutant residues (R132, R140 and R172). The findings of the present study will help the future development of novel biomarkers and therapeutics for brain tumors.
    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(GB)是成人最常见和最具侵袭性的原发性脑肿瘤,总生存期近14.6个月。最佳切除后联合替莫唑胺化疗和放疗,也被称为Stupp协议,仍然是治疗的标准;尽管如此,对替莫唑胺的抗性,可以通过许多分子途径获得,仍然是一个无法超越的障碍。影响替莫唑胺疗效的几个因素,包括其他DNA修复系统的参与,异常信号通路,自噬,表观遗传修饰,microRNAs,和细胞外囊泡的产生。血脑屏障,既是物理障碍,也是生化障碍,肿瘤微环境的前致癌和免疫抑制性质,和肿瘤特异性特征,如体积和抗原表达,是正在进行的调查的主题。在这次审查中,关于替莫唑胺耐药性获取的临床前和临床数据以及克服化疗耐药性的可能方法,或者在不恢复化学敏感性的情况下治疗神经胶质瘤,进行评估和介绍。目的是对临床上重要的分子机制及其复杂的相互关系进行彻底的检查,目的是增进了解,以更有效地打击对TMZ的抵抗。
    Glioblastoma (GB) is the most common and most aggressive primary brain tumor in adults, with an overall survival almost 14.6 months. Optimal resection followed by combined temozolomide chemotherapy and radiotherapy, also known as Stupp protocol, remains the standard of treatment; nevertheless, resistance to temozolomide, which can be obtained throughout many molecular pathways, is still an unsurpassed obstacle. Several factors influence the efficacy of temozolomide, including the involvement of other DNA repair systems, aberrant signaling pathways, autophagy, epigenetic modifications, microRNAs, and extracellular vesicle production. The blood-brain barrier, which serves as both a physical and biochemical obstacle, the tumor microenvironment\'s pro-cancerogenic and immunosuppressive nature, and tumor-specific characteristics such as volume and antigen expression, are the subject of ongoing investigation. In this review, preclinical and clinical data about temozolomide resistance acquisition and possible ways to overcome chemoresistance, or to treat gliomas without restoration of chemosensitinity, are evaluated and presented. The objective is to offer a thorough examination of the clinically significant molecular mechanisms and their intricate interrelationships, with the aim of enhancing understanding to combat resistance to TMZ more effectively.
    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
    目的:表皮生长因子受体(EGFR)及其活性突变体III型(EGFRvIII)的扩增,常见于胶质母细胞瘤(GBM),有助于GBM的化疗和放疗抵抗。阐明EGFRvIIIGBM中替莫唑胺(TMZ)耐药的潜在分子机制可以为癌症治疗提供有价值的见解。
    方法:为了阐明GBM中EGFRvIII介导的TMZ抗性的分子机制,我们使用基因表达综合和癌症基因组图谱(TCGA)数据库进行了综合分析.最初,我们确定了常见的显著差异表达基因(DEGs),并将那些与患者预后显著相关的基因作为EGFRvIII的潜在下游靶标和候选药物进行了优先排序.此外,我们分析了转录因子表达变化及其与候选基因的相关性,以阐明转录调控机制。使用估计方法和数据库,如肿瘤免疫估计资源(TIMER)和细胞标记,我们评估了TMZ抗性GBM中的免疫细胞浸润及其与候选基因表达的关系。在这项研究中,我们检查了EGFRvIII干预后GBM细胞系和TMZ抗性GBM细胞系中候选基因的表达差异。这项初步调查旨在验证EGFRvIII对候选靶标的监管影响及其可能参与GBM中的TMZ抗性。
    结果:值得注意的是,GTPase激活Rap/RanGAP结构域样3(GARNL3)作为与TMZ抗性和不良预后相关的关键DEG出现,表达降低与免疫细胞谱改变相关。转录因子分析表明,Epiregulin(EREG)是GARNL3的推定上游调节因子,将其与EGFRvIII介导的TMZ抗性联系起来。体外实验证实了EGFRvIII介导的GARNL3下调和GBM细胞系中TMZ敏感性降低,TMZ抗性GBM细胞中GARNL3水平降低进一步支持。
    结论:GARNL3在EGFRvIII阳性和TMZ抗性GBM中的下调暗示了其在TMZ抗性中的作用,提示调节EREG/GARNL3信号作为一种潜在的治疗策略。
    OBJECTIVE: Amplification of the epidermal growth factor receptor (EGFR) and its active mutant type III (EGFRvIII), frequently occurr in glioblastoma (GBM), contributing to chemotherapy and radiation resistance in GBM. Elucidating the underlying molecular mechanism of temozolomide (TMZ) resistance in EGFRvIII GBM could offer valuable insights for cancer treatment.
    METHODS: To elucidate the molecular mechanisms underlying EGFRvIII-mediated resistance to TMZ in GBM, we conducted a comprehensive analysis using Gene Expression Omnibus and The cancer genome atlas (TCGA) databases. Initially, we identified common significantly differentially expressed genes (DEGs) and prioritized those correlating significantly with patient prognosis as potential downstream targets of EGFRvIII and candidates for drug resistance. Additionally, we analyzed transcription factor expression changes and their correlation with candidate genes to elucidate transcriptional regulatory mechanisms. Using estimate method and databases such as Tumor IMmune Estimation Resource (TIMER) and CellMarker, we assessed immune cell infiltration in TMZ-resistant GBM and its relationship with candidate gene expression. In this study, we examined the expression differences of candidate genes in GBM cell lines following EGFRvIII intervention and in TMZ-resistant GBM cell lines. This preliminary investigation aimed to verify the regulatory impact of EGFRvIII on candidate targets and its potential involvement in TMZ resistance in GBM.
    RESULTS: Notably, GTPase Activating Rap/RanGAP Domain Like 3 (GARNL3) emerged as a key DEG associated with TMZ resistance and poor prognosis, with reduced expression correlating with altered immune cell profiles. Transcription factor analysis suggested Epiregulin (EREG) as a putative upstream regulator of GARNL3, linking it to EGFRvIII-mediated TMZ resistance. In vitro experiments confirmed EGFRvIII-mediated downregulation of GARNL3 and decreased TMZ sensitivity in GBM cell lines, further supported by reduced GARNL3 levels in TMZ-resistant GBM cells.
    CONCLUSIONS: GARNL3 downregulation in EGFRvIII-positive and TMZ-resistant GBM implicates its role in TMZ resistance, suggesting modulation of EREG/GARNL3 signaling as a potential therapeutic strategy.
    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
    替莫唑胺(TMZ)是临床神经胶质瘤治疗中使用的主要化疗药物,特别是对于高级别胶质母细胞瘤(GBM)。然而,TMZ耐药在GBM中的出现对其临床疗效构成重大障碍。我们的目的是阐明去泛素化酶(DUB)在GBM细胞对TMZ的抗性中的作用。我们使用广谱DUBs抑制剂G5来研究DUBs在TMZ针对GBM细胞的细胞毒性中的功能。产生具有特定DUB敲除的82个GBM细胞系,并进行CCK-8测定以评估细胞增殖和TMZ抗性。此外,GBM细胞中DUB和TMZ抗性之间的关联,随着自噬通量的调节,被检查过。pan-DUBs抑制剂G5证明了在GBM细胞中诱导细胞死亡和增强TMZ毒性的能力。随后,我们确定了参与调节GBM细胞增殖和TMZ耐药的潜在DUB.发现DUBs敲除对TMZ细胞毒性的影响与其调节TMZ诱导的自噬有关。总之,我们的研究提供了有关DUB在GBM细胞增殖和TMZ抗性中的作用的初步见解,并有助于更深入地了解GBM细胞中TMZ抗性的DUBs基因的复杂功能。
    Temozolomide (TMZ) stands as the primary chemotherapeutic drug utilized in clinical glioma treatment, particularly for high-grade glioblastoma (GBM). However, the emergence of TMZ resistance in GBM poses a significant hurdle to its clinical efficacy. Our objective was to elucidate the role of deubiquitinating enzymes (DUBs) in GBM cell resistance to TMZ. We employed the broad-spectrum DUBs inhibitor G5 to investigate the function of DUBs in TMZ cytotoxicity against GBM cells. Eighty-two GBM cell lines with specified DUBs knockout were generated and subjected to CCK-8 assays to assess cell proliferation and TMZ resistance. Furthermore, the association between DUBs and TMZ resistance in GBM cells, along with the modulation of autophagic flux, was examined. The pan-DUBs inhibitor G5 demonstrated the ability to induce cell death and enhance TMZ toxicity in GBM cells. Subsequently, we identified potential DUBs involved in regulating GBM cell proliferation and TMZ resistance. The impact of DUBs knockout on TMZ cytotoxicity was found to be associated with their regulation of TMZ-induced autophagy. In summary, our study provides primary insights into the role of DUBs in GBM cell proliferation and TMZ resistance, and contributes to a deeper understanding of the complex function of DUBs genes underlying TMZ resistance in GBM 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
    背景:替莫唑胺(TMZ)耐药性是多形性胶质母细胞瘤(GBM)治疗面临的主要障碍。Muscone,麝香(Moschus)的主要活性药理成分之一,可以穿过血脑屏障(BBB),并且正在作为抗肿瘤药物进行研究。然而,Muscone治疗GBM的研究很少,其可能的机制尚不清楚。
    目的:本研究旨在评估麝香酮对TMZ抗性GBM细胞的影响和潜在的分子机制。
    方法:使用GEO2R筛选TMZ抗性GBM细胞和TMZ敏感GBM细胞之间的差异表达基因(DEGs)。通过逐步提高TMZ浓度,建立了相对稳定的抗TMZ人GBM细胞系。通过CCK-8测定和MGMT和TOP2A表达的蛋白质印迹分析评估U251-TR细胞的耐药性特征。细胞活力,细胞增殖,细胞迁移能力,和药物协同作用通过CCK-8检测,集落形成试验,伤口愈合试验,和药物相互作用关系测试,分别。Anoikis通过钙黄绿素-AM/EthD-1染色定量,MTT测定,和流式细胞术。细胞周期停滞的测量,凋亡,线粒体膜电位(MMP),和活性氧(ROS)使用细胞周期染色进行,膜联蛋白V-FITC/PI标签,JC-1测定,和ROS测定,分别。DNA损伤通过TUNEL测定来测量,碱性彗星试验,和γ-H2AX病灶测定。GEPIA用于研究失巢凋亡标记(FAK)/耐药基因与EGFR/整合素β1信号通路中关键蛋白之间的联系。分子对接用于预测麝香酮的可能靶标。使用免疫荧光显示EGFR和FAK的细胞内共定位和表达。使用慢病毒转导构建稳定过表达EGFR的U251-TR细胞系,以评估EGFR相关信号传导在失巢凋亡抗性中的参与。WesternBlot用于检测迁移相关蛋白的表达,细胞周期蛋白,与之相关的蛋白质,DNA损伤/修复相关蛋白,和相关的途径蛋白。
    结果:DEGs分析在TMZ耐药GBM细胞中鉴定出97个化疗耐药基因和3779个上调基因。随后的实验验证了在连续低剂量TMZ诱导的U251-TR细胞中TMZ抗性和DNA修复相关基因(TOP2A和MGMT)的高表达。Muscone对U251-TR细胞迁移和增殖表现出剂量依赖性抑制,其与TMZ共同给药显示出增强治疗效果的潜力。通过下调FAK,麝香酮降低了锚定非依赖性U251-TR细胞的抗肛门凋亡性。它还通过上调p21并下调CDK1,CDK2和CyclinE1而导致G2/M期细胞周期停滞。Muscone诱导的失巢凋亡伴随线粒体膜电位崩溃,ROS生产,BAX/Bcl-2比值增加,以及细胞色素c(Cytc)水平升高,切割的caspase-9和切割的caspase-3。这些发现表明,麝香酮可能通过ROS的产生触发线粒体依赖性失巢凋亡。此外,显著的DNA损伤,DNA双链断裂(DSB),γ-H2AX灶的形成,和TOP2A表达的减少也与麝香酮诱导的失巢凋亡有关。EGFR在U251-TR细胞中的过表达促进整合素β1,FAK的表达,β-连环蛋白,TOP2A,而麝香酮抑制EGFR的表达水平,整合素β1,β-连环蛋白,FAK,TOP2AMuscone可能会影响关键DNA修复酶的表达,TOP2A,通过抑制EGFR/整合素β1/FAK途径。
    结论:我们首先证明了麝香酮通过EGFR/整合素β1/FAK途径抑制TOP2A表达,从而恢复TMZ抗性GBM细胞的失巢凋亡敏感性。这些数据表明,麝香酮可能是增强GBM治疗的有前途的联合治疗剂,特别是在TOP2A表达升高的TMZ抗性GBM的情况下。
    BACKGROUND: Temozolomide (TMZ) resistance is the main obstacle faced by glioblastoma multiforme (GBM) treatment. Muscone, one of the primary active pharmacological ingredients of Shexiang (Moschus), can cross the blood-brain barrier (BBB) and is being investigated as an antineoplastic medication. However, muscone treatment for GBM has received little research, and its possible mechanisms are still unclear.
    OBJECTIVE: This study aims to evaluate the effect and the potential molecular mechanism of muscone on TMZ-resistant GBM cells.
    METHODS: The differentially expressed genes (DEGs) between TMZ-resistant GBM cells and TMZ-sensitive GBM cells were screened using GEO2R. By progressively raising the TMZ concentration, a relatively stable TMZ-resistant human GBM cell line was established. The drug-resistance traits of U251-TR cells were assessed via the CCK-8 assay and Western Blot analysis of MGMT and TOP2A expression. Cell viability, cell proliferation, cell migration ability, and drug synergism were detected by the CCK-8 assay, colony formation assay, wound healing assay, and drug interaction relationship test, respectively. Anoikis was quantified by Calcein-AM/EthD-1 staining, MTT assay, and flow cytometry. Measurements of cell cycle arrest, apoptosis, mitochondrial membrane potential (MMP), and reactive oxygen species (ROS) were performed using cell cycle staining, Annexin V-FITC/PI labeling, JC-1 assay, and ROS assay, respectively. DNA damage was measured by TUNEL assay, alkaline comet assay, and γ-H2AX foci assay. GEPIA was used to investigate the link between the anoikis marker (FAK)/drug resistance gene and critical proteins in the EGFR/Integrin β1 signaling pathway. Molecular docking was used to anticipate the probable targets of muscone. The intracellular co-localization and expression of EGFR and FAK were shown using immunofluorescence. The U251-TR cell line stably overexpressing EGFR was constructed using lentiviral transduction to assess the involvement of EGFR-related signaling in anoikis resistance. Western Blot was employed to detect the expression of migration-related proteins, cyclins, anoikis-related proteins, DNA damage/repair-related proteins, and associated pathway proteins.
    RESULTS: DEGs analysis identified 97 deregulated chemotherapy-resistant genes and 3779 upregulated genes in TMZ-resistant GBM cells. Subsequent experiments verified TMZ resistance and the hyper-expression of DNA repair-related genes (TOP2A and MGMT) in continuously low-dose TMZ-induced U251-TR cells. Muscone exhibited dose-dependent inhibition of U251-TR cell migration and proliferation, and its co-administration with TMZ showed the potential for enhanced therapeutic efficacy. By downregulating FAK, muscone reduced anoikis resistance in anchorage-independent U251-TR cells. It also caused cell cycle arrest in the G2/M phase by upregulating p21 and downregulating CDK1, CDK2, and Cyclin E1. Muscone-induced anoikis was accompanied by mitochondrial membrane potential collapse, ROS production, an increase in the BAX/Bcl-2 ratio, as well as elevated levels of Cytochrome c (Cyt c), cleaved caspase-9, and cleaved caspase-3. These findings indicated that muscone might trigger mitochondrial-dependent anoikis via ROS generation. Moreover, significant DNA damage, DNA double-strand breaks (DSBs), the formation of γ-H2AX foci, and a reduction in TOP2A expression are also associated with muscone-induced anoikis. Overexpression of EGFR in U251-TR cells boosted the expression of Integrin β1, FAK, β-Catenin, and TOP2A, whereas muscone suppressed the expression levels of EGFR, Integrin β1, β-Catenin, FAK, and TOP2A. Muscone may influence the expression of the key DNA repair enzyme, TOP2A, by suppressing the EGFR/Integrin β1/FAK pathway.
    CONCLUSIONS: We first demonstrated that muscone suppressed TOP2A expression through the EGFR/Integrin β1/FAK pathway, hence restoring anoikis sensitivity in TMZ-resistant GBM cells. These data suggest that muscone may be a promising co-therapeutic agent for enhancing GBM treatment, particularly in cases of TMZ-resistant GBM with elevated TOP2A expression.
    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
    替莫唑胺(TMZ)耐药性仍然是胶质母细胞瘤(GBM)治疗的主要障碍。Lactylation是一种新的翻译后修饰,涉及多种肿瘤。然而,乳酸化是否在GBMTMZ耐药中起作用尚不清楚.在这里发现组蛋白H3K9乳化作用(H3K9la)通过LUC7L2介导的内含子7保留MLH1在GBM中赋予TMZ抗性。机械上,在复发性GBM组织和TMZ抗性细胞中,主要集中在组蛋白H3K9中。联合多组学分析,包括CUT和标记,SLAM-seq,和RNA-seq,揭示了H3K9的乳酸化显著富集在LUC7L2启动子中并激活LUC7L2转录以促进其表达。LUC7L2介导MLH1的内含子7保留以减少MLH1表达,从而抑制失配修复(MMR),最终导致GBMTMZ抵抗。值得注意的是,这是一种临床抗癫痫药物,stiripentol,可以穿过血脑屏障并抑制乳酸脱氢酶A/B(LDHA/B)活性,作为乳酸化抑制剂,使GBM细胞在体外和体内对TMZ更敏感。这些发现不仅阐明了GBMTMZ耐药性中的乳酸化机制,而且为临床GBM治疗提供了潜在的联合治疗策略。
    Temozolomide (TMZ) resistance remains the major obstacle in the treatment of glioblastoma (GBM). Lactylation is a novel post-translational modification that is involved in various tumors. However, whether lactylation plays a role in GBM TMZ resistance remains unclear. Here it is found that histone H3K9 lactylation (H3K9la) confers TMZ resistance in GBM via LUC7L2-mediated intron 7 retention of MLH1. Mechanistically, lactylation is upregulated in recurrent GBM tissues and TMZ-resistant cells, and is mainly concentrated in histone H3K9. Combined multi-omics analysis, including CUT&Tag, SLAM-seq, and RNA-seq, reveals that H3K9 lactylation is significantly enriched in the LUC7L2 promoter and activates LUC7L2 transcription to promote its expression. LUC7L2 mediates intron 7 retention of MLH1 to reduce MLH1 expression, and thereby inhibit mismatch repair (MMR), ultimately leading to GBM TMZ resistance. Of note, it is identified that a clinical anti-epileptic drug, stiripentol, which can cross the blood-brain barrier and inhibit lactate dehydrogenase A/B (LDHA/B) activity, acts as a lactylation inhibitor and renders GBM cells more sensitive to TMZ in vitro and in vivo. These findings not only shed light on the mechanism of lactylation in GBM TMZ resistance but also provide a potential combined therapeutic strategy for clinical GBM treatment.
    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:多形性胶质母细胞瘤(GBM)是最具侵袭性的癌症之一,目前的治疗方法疗效有限。GBM治疗中的一个重要障碍是对化疗剂替莫唑胺(TMZ)的抗性。MGMT启动子的甲基化状态已被认为是对TMZ反应的关键生物标志物。
    方法:为了探索耐药性的潜在机制,我们通过逐渐增加TMZ暴露,开发了两种抗TMZ的GBM细胞系.对TMZ抗性细胞系的转录组测序表明,组蛋白翻译后修饰的改变可能有助于赋予TMZ抗性。随后,多组学分析表明组蛋白H3赖氨酸9乙酰化(H3K9ac)水平与TMZ抗性之间存在很强的关联。
    结果:我们观察到H3K9ac和MGMT的表达之间存在显着相关性,特别是在未甲基化MGMT启动子样品中。更重要的是,我们的研究结果表明,H3K9ac可能通过促进SP1转录因子募集至MGMT转录因子结合位点来增强MGMT转录.此外,通过分析用TMZ治疗的匹配的原发性和复发性GBM肿瘤的单细胞转录组学数据,我们对肿瘤复发时发生的分子转移进行了建模.我们还注意到肿瘤干细胞特征的减少,伴随着H3K9ac的增加,SP1和MGMT级别,强调H3K9ac在TMZ治疗后肿瘤复发中的潜在作用。
    结论:H3K9ac的增加似乎增强了转录因子SP1在MGMT基因座内的结合位点的募集,因此在GBM中上调MGMT表达并驱动TMZ抗性。
    Glioblastoma multiforme (GBM) is among the most aggressive cancers, with current treatments limited in efficacy. A significant hurdle in the treatment of GBM is the resistance to the chemotherapeutic agent temozolomide (TMZ). The methylation status of the MGMT promoter has been implicated as a critical biomarker of response to TMZ.
    To explore the mechanisms underlying resistance, we developed two TMZ-resistant GBM cell lines through a gradual increase in TMZ exposure. Transcriptome sequencing of TMZ-resistant cell lines revealed that alterations in histone post-translational modifications might be instrumental in conferring TMZ resistance. Subsequently, multi-omics analysis suggests a strong association between histone H3 lysine 9 acetylation (H3K9ac) levels and TMZ resistance.
    We observed a significant correlation between the expression of H3K9ac and MGMT, particularly in the unmethylated MGMT promoter samples. More importantly, our findings suggest that H3K9ac may enhance MGMT transcription by facilitating the recruitment of the SP1 transcription factor to the MGMT transcription factor binding site. Additionally, by analyzing single-cell transcriptomics data from matched primary and recurrent GBM tumors treated with TMZ, we modeled the molecular shifts occurring upon tumor recurrence. We also noted a reduction in tumor stem cell characteristics, accompanied by an increase in H3K9ac, SP1, and MGMT levels, underscoring the potential role of H3K9ac in tumor relapse following TMZ therapy.
    The increase in H3K9ac appears to enhance the recruitment of the transcription factor SP1 to its binding sites within the MGMT locus, consequently upregulating MGMT expression and driving TMZ resistance 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(GBM)是一种高度侵袭性和耐药性的脑肿瘤,需要新的治疗策略。在这项研究中,我们通过设计和评估一系列阿比曲酮安装的异羟肟酸作为CYP17A1和HDAC6治疗GBM的潜在双重抑制剂,提出了一个机理上的突破.我们建立了GBM患者CYP17A1/HDAC6过表达与肿瘤复发和替莫唑胺耐药的相关性。化合物12,双重抑制剂,在体外表现出显著的抗GBM活性,特别是针对TMZ抗性细胞系。机械上,化合物12诱导细胞凋亡,抑制复发相关基因,诱导氧化应激并引发DNA损伤反应。此外,分子建模研究证实了其对CYP17A1和HDAC6的有效抑制活性。体内研究显示,化合物12有效抑制异种移植和原位小鼠模型中的肿瘤生长,而不诱导显著的副作用。这些发现强调了CYP17A1和HDAC6双重抑制作为克服GBM治疗耐药性的有希望的策略的潜力,并为改善治疗结果提供了新的希望。
    Glioblastoma (GBM) is a highly aggressive and treatment-resistant brain tumor, necessitating novel therapeutic strategies. In this study, we present a mechanistic breakthrough by designing and evaluating a series of abiraterone-installed hydroxamic acids as potential dual inhibitors of CYP17A1 and HDAC6 for GBM treatment. We established the correlation of CYP17A1/HDAC6 overexpression with tumor recurrence and temozolomide resistance in GBM patients. Compound 12, a dual inhibitor, demonstrated significant anti-GBM activity in vitro, particularly against TMZ-resistant cell lines. Mechanistically, compound 12 induced apoptosis, suppressed recurrence-associated genes, induced oxidative stress and initiated DNA damage response. Furthermore, molecular modeling studies confirmed its potent inhibitory activity against CYP17A1 and HDAC6. In vivo studies revealed that compound 12 effectively suppressed tumor growth in xenograft and orthotopic mouse models without inducing significant adverse effects. These findings highlight the potential of dual CYP17A1 and HDAC6 inhibition as a promising strategy for overcoming treatment resistance in GBM and offer new hope for improved therapeutic outcomes.
    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
    治疗抵抗和肿瘤复发是胶质母细胞瘤(GBM)死亡的主要原因,肿瘤内异质性发挥了重要作用。患者来源的癌症类器官已经成为能够概括肿瘤异质性的有希望的模型。我们的目标是开发患者来源的GBM类器官(PGO)以研究治疗反应和耐药性。
    GBM样品用于产生PGO,并使用全外显子组测序(WES)和单细胞核型测序进行分析。将PGO进行替莫唑胺(TMZ)以评估生存力。在TMZ之前和之后进行大量RNA测序。
    对培养3个时间点(1-3个月)的单个PGO的WES分析显示出高度的器官间相关性和遗传变异的保留(范围92.3%-97.7%)。与肿瘤相比,大多数变体保留在PGO中(范围58%-90%),并且表现出相似的拷贝数变异。单细胞核型测序显示遗传异质性的保留。单细胞多重免疫荧光显示细胞状态的维持。TMZ处理PGO显示出不同的反应,这在很大程度上与MGMT启动子甲基化相对应。TMZ前后的差异表达基因揭示了JNK激酶途径的上调。值得注意的是,JNK激酶抑制剂和TMZ的联合治疗显示出协同作用.
    总的来说,这些发现证明了PGO在保持培养中遗传和表型异质性方面的稳健性,以及在测量临床相关药物应答方面的应用.这些数据表明,PGO有可能进一步发展成为个性化适应性治疗选择和可行药物靶标发现的化身,并作为研究GBM生物学的平台。
    UNASSIGNED: Treatment resistance and tumor relapse are the primary causes of mortality in glioblastoma (GBM), with intratumoral heterogeneity playing a significant role. Patient-derived cancer organoids have emerged as a promising model capable of recapitulating tumor heterogeneity. Our objective was to develop patient-derived GBM organoids (PGO) to investigate treatment response and resistance.
    UNASSIGNED: GBM samples were used to generate PGOs and analyzed using whole-exome sequencing (WES) and single-cell karyotype sequencing. PGOs were subjected to temozolomide (TMZ) to assess viability. Bulk RNA sequencing was performed before and after TMZ.
    UNASSIGNED: WES analysis on individual PGOs cultured for 3 time points (1-3 months) showed a high inter-organoid correlation and retention of genetic variants (range 92.3%-97.7%). Most variants were retained in the PGO compared to the tumor (range 58%-90%) and exhibited similar copy number variations. Single-cell karyotype sequencing demonstrated preservation of genetic heterogeneity. Single-cell multiplex immunofluorescence showed maintenance of cellular states. TMZ treatment of PGOs showed a differential response, which largely corresponded with MGMT promoter methylation. Differentially expressed genes before and after TMZ revealed an upregulation of the JNK kinase pathway. Notably, the combination treatment of a JNK kinase inhibitor and TMZ demonstrated a synergistic effect.
    UNASSIGNED: Overall, these findings demonstrate the robustness of PGOs in retaining the genetic and phenotypic heterogeneity in culture and the application of measuring clinically relevant drug responses. These data show that PGOs have the potential to be further developed into avatars for personalized adaptive treatment selection and actionable drug target discovery and as a platform to study GBM biology.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号