MEOX2

MEOX2
  • 文章类型: Journal Article
    背景:肝星状细胞(HSC)是肝纤维化(LF)进展的关键加速因子。与HSC相比,成人来源的人肝干/祖细胞(ADHLSCs)在分化和增殖方面表现出更大的潜力,使它们在LF治疗中高度适用。这项研究的目的是通过比较ADHLSCs和HSCs之间的差异表达基因(DEGs)来确定LF的新治疗靶标。
    方法:我们使用从基因表达综合(GEO)数据库获得的GSE49995数据集研究了ADHLSC和HSC之间的DEG,旨在确定LF的新治疗靶点。随后,我们激活了HSC以深入研究间充质homeobox2(MEOX2),PH结构域富含亮氨酸的重复蛋白磷酸酶(PHLPP),和磷酸肌醇3-激酶(PI3K)/蛋白激酶B(AKT)信号通路在LF进展中,使用血小板衍生生长因子(PDGF),并且用过表达(OE)-MEOX2和shRNA-MEOX2(sh-MEOX2)慢病毒进行感染。使用细胞计数试剂盒-8(CCK-8)测定评估细胞活力,同时通过5-乙炔基-2'-脱氧尿苷(EdU)染色和流式细胞术评估细胞增殖。通过qPCR测定相对mRNA表达水平。进行蛋白质印迹分析以测量蛋白质表达水平,并使用双荧光素酶报告基因测定法研究了MEOX2的调节作用。
    结果:我们在ADHLSCs和HSCs之间鉴定了332个DEGs下调和201个DEGs上调。值得注意的是,MEOX2在ADHLSCs中的表达显著降低。这些DEGs主要参与含胶原的细胞外基质和PI3K/AKT信号通路。MEOX2可通过PI3K/AKT信号通路抑制癌细胞增殖。此外,JASRPAR2022数据库预测了MEOX2的靶基因PHLPP。我们的结果表明OE-MEOX2显着抑制HSC的细胞活力和增殖。进一步分析显示MEOX2与PHLPP启动子结合,从而上调其转录。这种作用导致p-AKT表达的抑制,因此减少HSC增殖并减缓LF的进展。
    结论:MEOX2上调PHLPP表达并抑制AKT磷酸化,从而降低HSCs的细胞活性和增殖能力,抑制LF的进展。
    BACKGROUND: Hepatic stellate cells (HSCs) serve as the crucial accelerating factor in the progression of liver fibrosis (LF). In contrast to HSCs, adult-derived human liver stem/progenitor cells (ADHLSCs) exhibit greater potency in terms of differentiation and proliferation, rendering them highly applicable in LF treatment. The objective of this study is to identify new therapeutic targets for LF by comparing differentially expressed genes (DEGs) between ADHLSCs and HSCs.
    METHODS: We investigated DEGs between ADHLSCs and HSCs using the GSE49995 dataset obtained from the Gene Expression Omnibus (GEO) database, aiming to identify new therapeutic targets for LF. Subsequently, we activated HSCs to delve deeper into the mesenchyme homeobox 2 (MEOX2), PH domain Leucine-rich repeat protein phosphatase (PHLPP), and Phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) signaling pathways in LF progression, employing platelet-derived growth factor (PDGF), and conducted infection with Overexpression (OE)-MEOX2 and shRNA-MEOX2 (sh-MEOX2) lentiviruses. Cell viability was assessed using the Cell Counting Kit-8 (CCK-8) assay, while cell proliferation was evaluated through 5-ethynyl-2\'-deoxyuridine (EdU) staining and flow cytometry. Relative mRNA expression levels were determined via qPCR. Western blot analysis was performed to measure protein expression levels, and the regulatory role of MEOX2 was investigated using dual luciferase reporter assays.
    RESULTS: We identified 332 DEGs that were down-regulated and 201 DEGs that were up-regulated between ADHLSCs and HSCs. Notably, MEOX2 expression in ADHLSCs was significantly reduced. These DEGs primarily participated in the collagen-containing extracellular matrix and the PI3K/AKT signaling pathway. MEOX2 could inhibit cancer cell proliferation via the PI3K/AKT signaling pathway. Additionally, the JASRPAR2022 database predicted the target gene PHLPP of MEOX2. Our results indicated that OE-MEOX2 significantly inhibited HSCs\' cell vitality and proliferation. Further analysis revealed that MEOX2 binds to PHLPP promoters, thereby up-regulating its transcription. This action led to the inhibition of p-AKT expression, consequently reducing HSC proliferation and slowing the progression of LF.
    CONCLUSIONS: MEOX2 up-regulates PHLPP expression and inhibits AKT phosphorylation, thereby reducing the cell activity and proliferation ability of HSCs and inhibiting the progression of LF.
    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
    小活检中分散的神经胶质瘤细胞的组织学鉴定可能具有挑战性,特别是在缺乏IDH1R132H突变或TP53改变的肿瘤中。我们假设免疫组织化学检测在神经胶质瘤中优先表达的蛋白质(EGFR,MEOX2,CD34)或胚胎发育过程中(SOX11,INSM1)可用于区分反应性神经胶质增生与神经胶质瘤。46个反应性胶质细胞的组织微阵列,81个胶质母细胞瘤,34IDH1突变型弥漫性神经胶质瘤,并分析了23例其他类型的胶质瘤。胶质肿瘤的EGFR阳性(p<0.001,χ2)明显更高(34.1%vs.0%),MEOX2(49.3%vs.2.3%),SOX11(70.5%与20.4%),和INSM1(65.4%与2.3%)。在94.3%(66/70)的胶质母细胞瘤中,观察到至少两种标志物的表达,而没有反应性神经胶质增生显示任何蛋白质的共表达。与IDH1突变肿瘤相比,胶质母细胞瘤的EGFR表达明显增高,MEOX2和CD34,并且显著降低SOX11的阳性。非弥漫性神经胶质瘤很少对所测试的五种标志物中的任何一种呈阳性。我们的结果表明,EGFR的免疫组织化学检测,MEOX2、SOX11和INSM1可用于在有限的组织学样本中检测胶质母细胞瘤细胞,特别是在组合使用时。
    Histological identification of dispersed glioma cells in small biopsies can be challenging, especially in tumours lacking the IDH1 R132H mutation or alterations in TP53. We postulated that immunohistochemical detection of proteins expressed preferentially in gliomas (EGFR, MEOX2, CD34) or during embryonal development (SOX11, INSM1) can be used to distinguish reactive gliosis from glioma. Tissue microarrays of 46 reactive glioses, 81 glioblastomas, 34 IDH1-mutant diffuse gliomas, and 23 gliomas of other types were analysed. Glial neoplasms were significantly more often (p < 0.001, χ2) positive for EGFR (34.1% vs. 0%), MEOX2 (49.3% vs. 2.3%), SOX11 (70.5% vs. 20.4%), and INSM1 (65.4% vs. 2.3%). In 94.3% (66/70) of the glioblastomas, the expression of at least two markers was observed, while no reactive gliosis showed coexpression of any of the proteins. Compared to IDH1-mutant tumours, glioblastomas showed significantly higher expression of EGFR, MEOX2, and CD34 and significantly lower positivity for SOX11. Non-diffuse gliomas were only rarely positive for any of the five markers tested. Our results indicate that immunohistochemical detection of EGFR, MEOX2, SOX11, and INSM1 can be useful for detection of glioblastoma cells in limited histological samples, especially when used in combination.
    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
    脂肪组织在能量代谢中起着至关重要的作用。多项研究表明,环状RNA(circularRNA,circRNA)参与脂肪发育和脂质代谢的调节。然而,关于它们参与绵羊基质血管部分(SVFs)的成脂分化知之甚少。这里,基于以前的测序数据和生物信息学分析,在绵羊身上发现了一部小说,它作为海绵促进miR-152抑制绵羊SVFs的成脂分化。使用生物信息学检查了circINSR和miR-152之间的相互作用,荧光素酶测定,和RNA免疫沉淀。值得注意的是,我们发现circINSR通过miR-152/间充质同源异型盒2(MEOX2)通路参与脂肪分化.MEOX2抑制绵羊SVFs的成脂分化,miR-152抑制MEOX2的表达。换句话说,circINSR直接在细胞质中分离miR-152并抑制其促进绵羊SVF成脂分化的能力.总之,这项研究揭示了circINSR在绵羊SVFs成脂分化中的作用及其调控机制,为进一步解释绵羊脂肪的发育及其调控机制提供参考。
    Adipose tissue plays a crucial role in energy metabolism. Several studies have shown that circular RNA (circRNA) is involved in the regulation of fat development and lipid metabolism. However, little is known about their involvement in the adipogenic differentiation of ovine stromal vascular fractions (SVFs). Here, based on previous sequencing data and bioinformatics analysis, a novel circINSR was identified in sheep, which acts as a sponge to promote miR-152 in inhibiting the adipogenic differentiation of ovine SVFs. The interactions between circINSR and miR-152 were examined using bioinformatics, luciferase assays, and RNA immunoprecipitation. Of note, we found that circINSR was involved in adipogenic differentiation via the miR-152/mesenchyme homeobox 2 (MEOX2) pathway. MEOX2 inhibited adipogenic differentiation of ovine SVFs and miR-152 inhibited the expression of MEOX2. In other words, circINSR directly isolates miR-152 in the cytoplasm and inhibits its ability to promote adipogenic differentiation of ovine SVFs. In summary, this study revealed the role of circINSR in the adipogenic differentiation of ovine SVFs and its regulatory mechanisms, providing a reference for further interpretation of the development of ovine fat and its regulatory mechanisms.
    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
    Pirarubicin (THP) is widely used in clinical antitumor therapy, but its cardiotoxicity seriously affects the therapeutic effect in patients. In the study, we investigated the role of ring finger protein 10 (RNF10) in cardiotoxicity induced by THP.
    A cardiac toxicity model in Sprague-Dawley (SD) rats induced by THP was established. Changes in diet, weight, electrocardiogram (ECG), and echocardiography were observed. Serum levels of brain natriuretic peptide (BNP), creatine kinase MB (CK-MB), cardiac troponin T (cTnT), and lactate dehydrogenase (LDH) were measured. The expression of RNF10 in myocardium was observed by immunohistochemistry. The expressions of RNF10, activator protein-1 (AP-1), mesenchyme homeobox 2 (Meox2), total nuclear factor (NF)-κB p65 (T-P65), phosphorylated NF-κB p65 (PP65), monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor (TNF)-α, interleukin (IL)-6, and mature IL-1β were detected by Western blot. A THP-induced H9c2 myocardial cell injury model was established. RNF10 was downregulated or overexpressed by RNF10 siRNA and a RNF10 lentiviral vector, respectively. Then, cell viability was measured. The expression of RNF10 in H9c2 cells was observed by immunofluorescence. All of the above signaling pathways were verified by Western blots.
    THP caused a series of cardiotoxic manifestations in SD rats. Our studies suggested that THP caused cardiac inflammation by inhibiting the expression of RNF10, while overexpression of RNF10 antagonized the cardiotoxicity induced by THP.
    Our study showed RNF10 improved THP-induced cardiac inflammation by regulating the AP-1/Meox2 signaling pathway. RNF10 may be a new target to treat THP-induced cardiotoxicity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    目的:探讨MEOX2表达与乳腺癌患者临床病理特征及生存的关系。材料和方法:我们使用乳腺癌组织微阵列进行免疫组织化学。使用χ平方检验分析MEOX2表达与临床病理特征之间的关联。使用Kaplan-Meier曲线测定生存分析。多变量Cox回归用于确定MEOX2表达与总生存期的相关性。结果:我们发现74.1%的患者(100/135)有不同水平的MEOX2表达。MEOX2与组织学分级相关,与Ki67表达呈负相关。较低的MEOX2表达与总生存率降低显著相关(p=0.0011)。结论:MEOX2的表达可能是乳腺癌诊断和预后的新生物标志物。
    在这项研究中,我们发现MEOX2蛋白的低表达与乳腺癌患者的低总生存率相关。MEOX2是乳腺癌患者的独立预后因素。它将成为乳腺癌诊断和预后的新生物标志物。
    Aim: To investigate associations of MEOX2 expression with clinicopathological features and survival of breast cancer patients. Materials & methods: We used a breast cancer tissue microarray for immunohistochemistry. Associations between MEOX2 expression and clinicopathological features were analyzed using the χ-square test. Survival analysis was determined using a Kaplan-Meier curve. Multivariate Cox regression was used to determine associations of MEOX2 expression with overall survival. Results: We found that 74.1% of patients (100/135) had expression of MEOX2 at varying levels. MEOX2 was associated with histological grade and negatively correlated with Ki67 expression. Lower MEOX2 expression was significantly associated with decreased overall survival (p = 0.0011). Conclusion: MEOX2 expression could be a novel diagnostic and prognostic biomarker of breast cancer.
    In this study we found that lower expression of the protein MEOX2 was associated with poor overall survival in breast cancer. MEOX2 is an independent prognostic factor for breast cancer patients. It would be a new diagnostic and prognostic biomarker for breast cancer.
    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
    未经证实:血管生成在乳腺癌的生长和转移中起关键作用,抑制血管生成已成为癌症治疗的有效策略。我们的研究旨在阐明与乳腺癌血管生成相关的关键候选基因和途径。
    UNASSIGNED:鉴定来自癌症基因组图谱(TCGA)数据库的原始乳腺癌(BRCA)基因数据集中的差异表达基因(DEGs),并进行DEGs的基因本体论分析。随后使用基因表达综合数据库确定Hub基因。通过定量实时聚合酶链反应(qRT-PCR)和免疫组织化学(IHC)评估了间充质同源盒2(MEOX2)在乳腺癌细胞和组织中的表达,分别。用Kaplan-Meier绘图仪评价MEOX2基因在乳腺癌组织中的预后价值。
    未经证实:在TCGA数据集中总共鉴定出61个血管生成相关的DEGs,其中MEOX2基因显著下调。GO功效注释和通路富集剖析显示,MEOX2在调控血管发育方面显著富集。IHC结果证实MEOX2表达在乳腺癌组织中被抑制,并且相对低水平表明该组织是密集血管化的。此外,用顺铂(DDP)和表柔比星(EPI)治疗后,乳腺癌细胞中MEOX2的表达显着升高。最后,Kaplan-Meier绘图仪证实,较高的MEOX2表达水平与较好的总生存期相关.
    UNASSIGNED:我们的研究表明,血管生成相关基因MEOX2可作为乳腺癌诊断和临床治疗的新型生物标志物。
    UNASSIGNED: Angiogenesis plays a critical role in the growth and metastasis of breast cancer and angiogenesis inhibition has become an effective strategy for cancer therapy. Our study aimed to clarify the key candidate genes and pathways related to breast cancer angiogenesis.
    UNASSIGNED: Differentially expressed genes (DEGs) in the raw breast cancer (BRCA) gene dataset from the Cancer Genome Atlas (TCGA) database were identified and gene ontology analysis of the DEGs was performed. Hub genes were subsequently determined using the Gene Expression Omnibus database. The expression of the mesenchyme homeobox 2 (MEOX2) in breast cancer cells and tissues was assessed by quantification real-time polymerase chain reaction (qRT-PCR) and immunohistochemistry (IHC), respectively. The prognostic value of the MEOX2 gene in breast cancer tissue was evaluated with the Kaplan-Meier plotter.
    UNASSIGNED: A total of 61 angiogenesis-related DEGs were identified in the TCGA dataset, among which the gene MEOX2 was significantly down-regulated. GO functional annotation and pathway enrichment analyses showed that MEOX2 was significantly enriched in the regulation of vasculature development. The IHC results confirmed that MEOX2 expression was repressed in breast cancer tissues and the relatively low level indicated the tissue was densely vascularized. Moreover, MEOX2 expression was significantly elevated in breast cancer cells after treatment with cisplatin (DDP) and epirubicin (EPI). Finally, the Kaplan-Meier plotter confirmed that higher expression levels of MEOX2 were related to better overall survival.
    UNASSIGNED: Our study revealed that the angiogenesis-associated gene MEOX2 can be used as a novel biomarker for breast cancer diagnosis and clinical therapy.
    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
    关于胶质母细胞瘤发展的最广泛接受的假设表明,胶质母细胞瘤干细胞样细胞(GSC)与肿瘤的发生和复发以及化学和放射性抗性的发生至关重要。间充质同源异型盒2(MEOX2)是胶质母细胞瘤中过度表达的转录因子,其表达与患者生存率呈负相关。从我们的观察开始,MEOX2表达在六个GSC细胞系中强烈增强,我们在两个不同的GSC细胞系中进行了shRNA介导的敲减实验,发现MEOX2耗竭导致细胞生长和球体形成能力的抑制以及凋亡性细胞死亡的增加.通过深度转录组分析,我们确定了一组响应MEOX2敲低而被调节的核心基因。在这些基因中,被抑制的基因在很大程度上富含参与缺氧反应和糖酵解途径的基因,导致高级别神经胶质瘤对治疗产生抗性的两条严格相关的途径。通过MEOX2敲低差异表达的基因的调控区的计算机研究表明,它们主要由富含Sp1和Klf4结合基序的富含GC的区域组成,胶质母细胞瘤代谢的两个主要调节因子。我们的结果显示,第一次,MEOX2参与GSC代谢基因的调节,这对这些细胞的存活和生长至关重要。
    The most widely accepted hypothesis for the development of glioblastoma suggests that glioblastoma stem-like cells (GSCs) are crucially involved in tumor initiation and recurrence as well as in the occurrence of chemo- and radio-resistance. Mesenchyme homeobox 2 (MEOX2) is a transcription factor overexpressed in glioblastoma, whose expression is negatively correlated with patient survival. Starting from our observation that MEOX2 expression is strongly enhanced in six GSC lines, we performed shRNA-mediated knock-down experiments in two different GSC lines and found that MEOX2 depletion resulted in the inhibition of cell growth and sphere-forming ability and an increase in apoptotic cell death. By a deep transcriptome analysis, we identified a core group of genes modulated in response to MEOX2 knock-down. Among these genes, the repressed ones are largely enriched in genes involved in the hypoxic response and glycolytic pathway, two strictly related pathways that contribute to the resistance of high-grade gliomas to therapies. An in silico study of the regulatory regions of genes differentially expressed by MEOX2 knock-down revealed that they mainly consisted of GC-rich regions enriched for Sp1 and Klf4 binding motifs, two main regulators of metabolism in glioblastoma. Our results show, for the first time, the involvement of MEOX2 in the regulation of genes of GSC metabolism, which is essential for the survival and growth of these 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:胶质母细胞瘤(GBM)是一种侵袭性肿瘤,通常表现为7号染色体的增加,10号染色体的丢失和异常激活的受体酪氨酸激酶信号通路。以前,我们确定了MesenchymeHomeobox2(MEOX2),位于7号染色体上的基因,作为GBM中上调的转录因子。过表达的转录因子可参与驱动GBM。这里,我们旨在解决MEOX2在GBM中的作用。
    方法:患者来源的GBM肿瘤球用于组成型敲低或过表达MEOX2,并进行包括蛋白质印迹的体外测定以评估ERK磷酸化。脑类器官模型用于研究MEOX2在生长启动中的作用。使用颅内小鼠植入模型来评估MEOX2的致瘤潜力。RNA测序,ACT-seq和CUT&Tag用于鉴定MEOX2靶基因。
    结果:MEOX2通过前馈机制增强ERK信号。我们将Ser155鉴定为MEOX2同源盒结构域上游的推定的ERK依赖性磷酸化位点。S155A取代对MEOX2蛋白水平有重要影响并改变其亚核定位。在人类恶性神经胶质瘤的脑类器官模型中,MEOX2过表达与p53和PTEN丢失协作以诱导细胞增殖。利用高通量基因组学,我们在患者来源的GBM肿瘤球体模型和新鲜冷冻的GBM肿瘤中鉴定了MEOX2的推定转录靶基因。
    结论:我们确定MEOX2是GBM中的致癌转录调节因子。MEOX2增加GBM的脑类器官模型中的增殖并进入代表GBM中的核心信号通路的ERK信号传导。
    Glioblastoma (GBM) is an aggressive tumor that frequently exhibits gain of chromosome 7, loss of chromosome 10, and aberrantly activated receptor tyrosine kinase signaling pathways. Previously, we identified Mesenchyme Homeobox 2 (MEOX2), a gene located on chromosome 7, as an upregulated transcription factor in GBM. Overexpressed transcription factors can be involved in driving GBM. Here, we aimed to address the role of MEOX2 in GBM.
    Patient-derived GBM tumorspheres were used to constitutively knockdown or overexpress MEOX2 and subjected to in vitro assays including western blot to assess ERK phosphorylation. Cerebral organoid models were used to investigate the role of MEOX2 in growth initiation. Intracranial mouse implantation models were used to assess the tumorigenic potential of MEOX2. RNA-sequencing, ACT-seq, and CUT&Tag were used to identify MEOX2 target genes.
    MEOX2 enhanced ERK signaling through a feed-forward mechanism. We identified Ser155 as a putative ERK-dependent phosphorylation site upstream of the homeobox-domain of MEOX2. S155A substitution had a major effect on MEOX2 protein levels and altered its subnuclear localization. MEOX2 overexpression cooperated with p53 and PTEN loss in cerebral organoid models of human malignant gliomas to induce cell proliferation. Using high-throughput genomics, we identified putative transcriptional target genes of MEOX2 in patient-derived GBM tumorsphere models and a fresh frozen GBM tumor.
    We identified MEOX2 as an oncogenic transcription regulator in GBM. MEOX2 increases proliferation in cerebral organoid models of GBM and feeds into ERK signaling that represents a core signaling pathway 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
    已在几种恶性肿瘤中观察到间充质同源盒2(MEOX2)的癌症抑制作用。然而,MEOX2与乳腺癌之间的关联尚未得到解决.这项研究的重点是探讨MEOX2在乳腺癌中的可能相关性。通过TCGA数据的初始表达分析揭示了乳腺癌中MEOX2的低水平。然后,我们通过实时定量PCR和免疫印迹法证实了MEOX2在乳腺癌的临床肿瘤标本中表达不良。此外,研究发现,乳腺癌患者MEOX2水平低与总生存率降低相关.一系列的细胞功能检测表明,MEOX2的强制表达具有抗癌作用,包括抑制细胞增殖,诱导G0-G1期停滞,抑制转移潜能,和增强化学敏感性。进一步分析显示MEOX2负调节磷脂酰肌醇-3激酶(PI3K)/AKT/哺乳动物雷帕霉素靶标(mTOR)和细胞外信号调节激酶(ERK1/2)途径。化学激活剂对AKT的再激活可逆转MEOX2介导的抗癌作用。体内异种移植试验验证了MEOX2在乳腺癌中的抗癌功能。一起来看,这些数据表明,MEOX2通过影响PI3K/AKT/mTOR和ERK1/2通路在乳腺癌中发挥抑癌作用.这项工作表明MEOX2是乳腺癌进展的新贡献者,这可能是抗癌治疗发展的候选靶标。
    A cancer-inhibiting role of mesenchyme homeobox 2 (MEOX2) has been observed in several malignancies. However, the association between MEOX2 and breast carcinoma has not been addressed. This research focused on investigating the possible relevance of MEOX2 in breast carcinoma. Initial expression analysis by TCGA data uncovered low levels of MEOX2 in breast carcinoma. We then confirmed that MEOX2 was poorly expressed in clinical tumor specimens of breast carcinoma by real-time quantitative PCR and immunoblotting assays. Moreover, low levels of MEOX2 in breast carcinoma patients were found to be correlated with reduced overall survival. A series of cellular function assays showed that the forced expression of MEOX2 had anticancer effects, including the inhibition of cell proliferation, the induction of G0-G1 phase arrest, the restraint of metastatic potential, and the enhancement of chemosensitivity. Further analysis revealed that MEOX2 negatively modulated the phosphatidyl-inositol-3 kinase (PI3K)/AKT/mammalian target of the rapamycin (mTOR) and extracellular signal-regulated kinase (ERK1/2) pathways. Reactivation of AKT by a chemical activator reversed MEOX2-mediated anticancer effects. An in vivo xenograft assay validated the anticancer function of MEOX2 in breast carcinoma. Taken together, these data show that MEOX2 exerts a cancer-inhibiting role in breast carcinoma by affecting the PI3K/AKT/mTOR and ERK1/2 pathways. This work suggests MEOX2 as a new contributor for breast carcinoma progression, which may be a candidate target for anticancer therapy development.
    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

公众号