MIEN1

MIEN1
  • 文章类型: Journal Article
    背景:长链非编码RNA(lncRNA)有助于胃癌(GC)的发生和发展。这项研究的目的是研究lncRNA结直肠肿瘤差异表达(CRNDE)在通过抑制GC中的miR-136-5p调节迁移和侵袭增强子1(MIEN1)表达中的潜在作用。方法:CRNDE的生物学作用,miR-136-5p,在实验室环境中和通过临床样本检查评估GC中的MIEN1。结果:发现GC组织中CRNDE明显增加,这种上调与GC患者的预后不良有关。体外实验表明,抑制细胞生长和迁移,在促进GC细胞凋亡的同时,可以通过禁用CRNDE或MIEN1,或通过增加miR-136-5p的表达来实现。MIEN1是miR-136-5p的特定受体,miR-136-5p的抗癌作用可以通过增加MIEN1的表达来抵消。通过对临床标本的检查,已经观察到MIEN1的表达与CRNDE之间存在显着正相关。相比之下,miR-136-5p在GC组织中的表达呈负相干。结论:先前未探索的GC治疗靶标涉及CRNDE/miR-136-5p/MIEN1信号转导级联。
    Background: Long noncoding RNAs (lncRNAs) contribute to the initiation and progression of gastric cancer (GC). The purpose of this study is to examine the potential role of lncRNA colorectal neoplasia differentially expressed (CRNDE) in modulating the expression of migration and invasion enhancer 1 (MIEN1) through the suppression of miR-136-5p in GC. Methods: The biological roles of CRNDE, miR-136-5p, and MIEN1 in GC were assessed both in laboratory settings and through the examination of clinical samples. Results: CRNDE was found to be significantly increased in GC tissues, and this upregulation was associated with an unfavorable prognosis of GC patients. In vitro experiments showed that inhibiting cell growth and migration, along with promoting apoptosis in GC cells, could be achieved by either disabling CRNDE or MIEN1, or by increasing the expression of miR-136-5p. MIEN1 is a specific recipient of miR-136-5p, and the anticancer effects of miR-136-5p can be counteracted by the increased expression of MIEN1. Through the examination of clinical specimens, it has been observed that there is a significant positive correlation between the expression of MIEN1 and CRNDE. In contrast, miR-136-5p expression in GC tissues shows a negative correlation. Conclusion: A previously unexplored therapeutic target for GC involves the CRNDE/miR-136-5p/MIEN1 signal transduction 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
    自噬的失调与许多疾病的发展有关。包括癌症.这里,我们揭示了E3泛素连接酶HRD1通过调节自噬在非小细胞肺癌(NSCLC)转移中的新功能。机械上,HRD1通过促进ATG3的泛素化和降解抑制自噬。此外,一种促迁徙和侵袭性因素,MIEN1(迁移和入侵增强子1),被发现在HRD1缺乏时自噬降解。重要的是,HRD1和MIEN1在肺肿瘤中表达上调且呈正相关。基于这些结果,我们提出了一种新的HRD1功能机制,即HRD1降解ATG3蛋白导致自噬抑制和MIEN1释放,从而促进NSCLC转移。因此,我们的发现为HRD1在NSCLC转移中的作用提供了新的见解,并为肺癌治疗提供了新的治疗靶点.
    Dysregulation of autophagy has been implicated in the development of many diseases, including cancer. Here, we revealed a novel function of the E3 ubiquitin ligase HRD1 in non-small cell lung carcinoma (NSCLC) metastasis by regulating autophagy. Mechanistically, HRD1 inhibits autophagy by promoting ATG3 ubiquitination and degradation. Additionally, a pro-migratory and invasive factor, MIEN1 (migration and invasion enhancer 1), was found to be autophagically degraded upon HRD1 deficiency. Importantly, expression of both HRD1 and MIEN1 are upregulated and positively correlated in lung tumors. Based on these results, we proposed a novel mechanism of HRD1 function that the degradation of ATG3 protein by HRD1 leads to autophagy inhibition and MIEN1 release, thus promoting NSCLC metastasis. Therefore, our findings provided new insights into the role of HRD1 in NSCLC metastasis and new therapeutic targets for lung cancer 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
    耐药性是一种复杂的现象,在癌症治疗期间经常发展为化疗失败。恶性细胞通过不同的机制和途径越来越多地产生对各种化疗药物的抗性。了解与耐药性有关的分子机制仍然是确定精确靶标和发现药物以改善治疗结果的重要研究领域。这篇综述强调了一些最近出现的靶标和途径的作用,这些靶标和途径在驱动耐药性中起着关键作用。
    Drug resistance is a complex phenomenon that frequently develops as a failure to chemotherapy during cancer treatment. Malignant cells increasingly generate resistance to various chemotherapeutic drugs through distinct mechanisms and pathways. Understanding the molecular mechanisms involved in drug resistance remains an important area of research for identification of precise targets and drug discovery to improve therapeutic outcomes. This review highlights the role of some recent emerging targets and pathways which play critical role in driving drug resistance.
    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
    环状RNA(circularRNAs)的异常表达与多囊卵巢综合征(PCOS)的进展有关,这通常会导致女性不孕。在这项研究中,我们确定了circ_0030018在PCOS中的作用。定量聚合酶链反应(qPCR)检测circ_0030018、microRNA(miR)-136、迁移和侵袭增强子1(MIEN1)的表达水平。进行细胞计数试剂盒-8和5-乙炔基-2'-脱氧尿苷测定以分析KGN细胞的增殖。使用荧光激活的细胞分选分析细胞凋亡。进行Transwell测定以测量细胞的迁移和侵袭能力。qPCR和Western印迹用于测量E-cadherin的水平,N-钙黏着蛋白,蜗牛,还有波形蛋白.circ_0030018或MIEN1表达与miR-136表达的相关性通过荧光素酶报告基因和RNA下拉测定来证实。结果显示circ_0030018在PCOS和KGN细胞中表达上调。circ_0030018的击倒抑制了增殖,迁移,和细胞的入侵,同时促进其凋亡。circ_0030018海绵化miR-136,其靶向MIEN1。此外,miR-136的下调消除了circ_0030018沉默的影响,而MIEN1的过表达逆转了miR-136对KGN细胞的诱导作用。总之,circ_0030018的缺失通过miR-136/MIEN1轴延迟了PCOS的进展。
    The abnormal expression of circular RNAs (circRNAs) is associated with the progression of polycystic ovary syndrome (PCOS), which commonly causes infertility in women. In this study, we identified the role of circ_0030018 in PCOS. Quantitative polymerase chain reaction (qPCR) was used to detect the expression levels of circ_0030018, microRNA (miR)-136, and migration and invasion enhancer 1 (MIEN1). Cell counting kit-8 and 5-ethynyl-2\'-deoxyuridine assays were performed to analyze the proliferation of KGN cells. Apoptosis was analyzed using fluorescence-activated cell sorting. Transwell assays were performed to measure the migration and invasion abilities of cells. qPCR and Western blotting were used to measure the levels of E-cadherin, N-cadherin, Snail, and vimentin. The correlation of circ_0030018 or MIEN1 expression with miR-136 expression was confirmed via luciferase reporter and RNA pull-down assays. Results showed that circ_0030018 expression was upregulated in patients with PCOS and KGN cells. Knockdown of circ_0030018 suppressed the proliferation, migration, and invasion of cells, while promoting their apoptosis. circ_0030018 sponged miR-136, which targeted MIEN1. Moreover, downregulation of miR-136 abrogated the effects of circ_0030018 silencing, while the overexpression of MIEN1 reversed the miR-136-induced effect on KGN cells. In summary, loss of circ_0030018 delayed the progression of PCOS via the miR-136/MIEN1 axis.
    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
    To observe the effect of miR-124-5p on progression of gastric cancer (GC) and explore the targeting mechanism.
    After collecting the specimens, we used real-time fluorescence quantitative PCR to detect the miR-124-5p level of GC tissue and corresponding adjacent tissue. Then MTT test and scratch wound-healing assay were hired to evaluate the influence of miR-124-5p in GC cell (SGC-803 and SGC7901) migration and proliferation ability. The binding of miR-124-5p to migration and invasion enhancer 1 (MIEN1) was detected through dual luciferase reporter gene experiment and western blot was utilized to assay the protein level of MIEN1.
    Compared with adjacent tissues, miR-124-5p level in GC tissues was lower significantly. MiR-124-5p mimic inhibited the metastasis and proliferation ability of SGC7901 cells and miR-124-5p inhibitor promoted the migration and proliferation ability of SGC803 cells. In addition, miR-124-5p targeted MIEN1 and negatively modulated the MIEN1 expression in SGC-803 and SGC7901 cells. Silencing MIEN1 negatively regulated the metastasis and proliferation ability of SGC7901 cells.
    MiR-124-5p inhibited the GC cell proliferation and metastasis phenotypes through MIEN1, which probably becomes a novel molecular target for clinical GC treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Previous reports have revealed that circRNA_100876 was extremely important in the progression of triple-negative breast cancer. Nevertheless, the mechanism towards the role of circRNA_100876 in Gastric cancer (GC) remains unknown. Here, we determined circRNA_100876 expression by quantitative real-time PCR (qRT-PCR) in twenty pairs of GC tissues and adjacent tissues. Our data indicated that the expression of circRNA_100876 was raised in GC tissues. In vitro, functional experiments confirmed that cell proliferation, invasion along with migration was promoted by circRNA_100876 in GC tissues. Simultaneously, relative luciferase assay uncovered that circRNA_100876 functioned as a sponge for miR-136, followed by retarding miR-136-induced inhibited effects on the corresponding target, MIEN1. Moreover, we revealed that the expression of MIEN1 was up-regulated and correlated to much worse prognosis of GC. Collectively, our data identified that the promotion of GC growth and metastasis induced by circRNA_100876 interacted with miR-136 and MIEN1, indicating an emerging announcement for uncovering the potential mechanism of GC progression.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    : Migration and invasion enhancer 1 (MIEN1) is a membrane-anchored protein and exists in various cancerous tissues. However, the roles of MIEN1 in prostate cancer have not yet been clearly addressed. We determined the expression, biological functions, and regulatory mechanisms of MIEN1 in the prostate. The results of immunohistochemical analysis indicated that MIEN1 was expressed specifically in epithelial cells and significantly higher in adenocarcinoma as compared to in normal tissues. MIEN1 enhanced in vitro cell proliferation, invasion, and in vivo tumorigenesis. Meanwhile, MIEN1 attenuated cisplatin-induced apoptosis in PC-3 cells. Overexpression of NF-ĸB-inducing kinase (NIK) enhanced MIEN1 expression, while overexpression of NF-ĸB inhibitor α (IĸBα) blocked MIEN1 expression in PC-3 cells. In prostate carcinoma cells, MIEN1 provoked Akt phosphorylation; moreover, MIEN1 downregulated N-myc downstream regulated 1 (NDRG1) but upregulated interleukin-6 (IL-6) gene expression. MK2206, an Akt inhibitor, impeded the modulation of MIEN1 on NDRG1 and IL-6 expressions. Our studies suggest that MIEN1 is an NF-ĸB downstream oncogene in the human prostate. Accordingly, the modulation of Akt signaling in the gene expressions of NDRG1 and IL-6 may account for the functions of MIEN1 in cell proliferation, invasion, and tumorigenesis in prostate carcinoma cells.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Tumor metastasis is a sequential event accounting for numerous cancer-related fatalities worldwide. The process of metastasis serially involves invasion, intravasation, extravasation, and tumor growth at the secondary site. Migration and invasion enhancer 1 (MIEN1) is a membrane associated protein overexpressed in various human cancers. Biological activity of MIEN1 is driven by geranylgeranyltransferase-I mediated prenylation at CAAX motif and methylation of the prenylated protein that anchors MIEN1 into the cellular membrane. Post-translationally modified MIEN1 interacts with Syk kinase and Annexin A2 protein; polymerizes G-actin and stabilizes F-actin filament; induces focal adhesion kinase phosphorylation and decrease cofilin phosphorylation implicated in both invasion and metastasis of different cancer types. In the present review, we discuss the structure, function, and involvement of MIEN1 in cancer progression. We also highlight the future prospects of MIEN1 as an emerging molecule and novel target in cancer cell invasion and metastasis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    MIEN1是一种新的癌基因,它涉及各种癌症类型的肿瘤进展,包括结肠癌.然而,MIEN1在结肠癌进展中的确切分子机制仍有待完全阐明。在本研究中,生物信息学预测显示miR-136可能是MIEN1的上游调节因子;荧光素酶分析和Western印迹分析显示miR-136通过直接靶向MIEN1的3'-非翻译区序列负调节MIEN1的表达。此外,使用伤口愈合和transwell侵袭的功能试验表明,过表达的miR-136抑制细胞迁移和侵袭,MIEN1的过表达部分挽救了miR-136在结肠癌细胞中的上述作用。此外,一项临床样本分析显示,miR-136在结肠癌组织中的表达普遍下调,与MIEN1表达呈负相关。此外,我们发现miR-136在结肠癌中抑制Akt/NF-κB信号通路和上皮-间质转化.这些结果表明miR-136作为肿瘤抑制因子,通过抑制结肠癌中的MIEN1表达来参与肿瘤转移,为结肠癌的治疗提供了新的靶点。
    MIEN1 is a novel oncogene, and it involves tumor progression in various cancer types, including colon cancer. However, the definite molecular mechanisms of MIEN1 in colon cancer progression remain to be completely elucidated. In the present study, bioinformatics prediction showed that miR-136 could be an upstream regulator of MIEN1; a luciferase assay and Western blot assay revealed that miR-136 negatively regulates MIEN1 expression via directly targeting its 3\'-untranslated region sequence. Moreover, a functional assay using wound healing and transwell invasion showed that overexpressed miR-136 inhibited cell migration and invasion, and overexpression of MIEN1 partly rescued the above-mentioned effects of miR-136 in colon cancer cells. Additionally, a clinical sample assay showed that miR-136 expression was generally downregulated in colon cancer tissue, which was inversely correlated with MIEN1 expression. Furthermore, we found that miR-136 suppressed the Akt/NF-κB signaling pathway and epithelial-to-mesenchymal transition in colon cancer. These results suggest that miR-136, as a tumor suppressor, acts in tumor metastasis by suppressing MIEN1 expression in colon cancer, providing a novel target for the treatment of colon cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Migration and invasion enhancer 1 (MIEN1) is a novel gene involved in prostate cancer progression by enhancing prostate cancer cell migration and invasion. DNA methylation, an important epigenetic regulation, is one of the most widely altered mechanisms in prostate cancer. This phenomenon frames the basis to study the DNA methylation patterns in the promoter region of MIEN1. Bisulfite pyrosequencing demonstrates the MIEN1 promoter contains a short interspersed nuclear Alu element (SINE Alu) repeat sequence. Validation of methylation inhibition on MIEN1 was performed using nucleoside analogs and non-nucleoside inhibitors and resulted in an increase in both MIEN1 RNA and protein in normal cells. MIEN1 mRNA and protein increases upon inhibition of individual DNA methyltransferases using RNA interference technologies. Furthermore, dual luciferase reporter assays, in silico analysis, and chromatin immunoprecipitation assays identified a sequence upstream of the transcription start site that has a site for binding of the USF transcription factors. These results suggest the MIEN1 promoter has a SINE Alu region that is hypermethylated in normal cells leading to repression of the gene. In cancer, the hypomethylation of a part of this repeat, in addition to the binding of USF, results in MIEN1 expression.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号