PTTG1

PTTG1
  • 文章类型: Journal Article
    垂体肿瘤转化基因1(PTTG1)是一个参与染色体分离的癌基因,DNA修复,凋亡,和新陈代谢。PTTG1可用于临床诊断和治疗,是口咽恶性肿瘤的潜在靶点。使用CCK-8测定评估Cal27和FaDu细胞的增殖和活力。实时PCR和蛋白质印迹,分别,分别分析PTTG1和IFIH1的mRNA和蛋白表达水平。使用RNA下拉法分析PTTG1mRNA与翻译调节蛋白IFIH1之间的相互作用,RNA免疫沉淀,和荧光素酶报告基因测定。PTTG1蛋白在口咽癌中显著过表达,而PTTG1mRNA没有。我们假设翻译调节蛋白在PTTG1中起转录后作用。IFIH1蛋白与PTTG1mRNA的42-52nt区域特异性结合,促进PTTG1的翻译,促进口咽癌细胞的增殖。给予PTTG1抑制剂PHA-848125和沉默IFIH1协同降低PTTG1的表达,抑制口咽癌细胞的增殖,预后良好。我们发现IFIH1-PTTG1轴可以调节PHA-848125反应,并在功能上介导个体间口咽癌的易感性和预后。本研究旨在确认PTTG1的上游调控基因,并进一步研究该信号通路中的特异性相互作用。这将为口咽肿瘤的治疗提供新的途径。
    The pituitary tumor-transforming gene 1 (PTTG1) is an oncogene involved in chromosomal segregation, DNA repair, apoptosis, and metabolism. PTTG1 can be used for clinical diagnosis and treatment and is a potential target for oropharyngeal carcinoma. The proliferation and viability of Cal27 and FaDu cells were assessed using the CCK-8 assay. Real-time PCR and western blotting, respectively, were used to analyze the mRNA and protein expression levels of PTTG1 and IFIH1. The interaction between PTTG1 mRNA and the translational regulatory protein IFIH1 was analyzed using RNA pull-down, RNA immunoprecipitation, and luciferase reporter assays. PTTG1 protein was significantly overexpressed in oropharyngeal carcinoma, whereas PTTG1 mRNA was not. We hypothesized that a translation regulatory protein plays a post-transcriptional role in PTTG1. The IFIH1 protein specifically bound to the 42-52 nt region of PTTG1 mRNA, promoted the translation of PTTG1, and promoted the proliferation of oropharyngeal cancer cells. Administration of the PTTG1 inhibitor PHA-848125 and silencing of IFIH1 synergistically decreased the expression of PTTG1, inhibited the proliferation of oropharyngeal cancer cells, and indicated a good prognosis. We found that the IFIH1-PTTG1 axis could regulate the PHA-848125 response and functionally mediate inter-individual oropharyngeal cancer susceptibility and prognosis. This study aimed to confirm the upstream regulatory genes of PTTG1 and further investigate the specific interactions in this signaling pathway, which will provide a new approach for the treatment of oropharyngeal carcinoma.
    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:很少有研究研究GLI1和PTTG1在接受结直肠癌(CRC)根治性手术的患者中的表达及其与淋巴结转移(LNM)的关系。因此,需要进行更多相关的研究和分析。
    目的:探讨接受CRC根治术的患者GLI1和PTTG1的表达及其与LNM的相关性。
    方法:本研究选择了2020年4月至2023年4月我院收治的103例CRC患者。收集CRC和邻近组织的样品标本以确定GLI1和PTTG1的阳性率和表达水平。这两个基因与患者临床病理数据的相关性(例如,LNM)进行了探索,并分析了LNM患者和无LNM患者之间GLI1和PTTG1表达的差异。绘制受试者工作特征(ROC)曲线以评估两种基因在CRC患者中对LNM的预测潜力。
    结果:与邻近组织相比,在CRC组织样本中观察到GLI1和PTTG1的阳性率和表达水平明显更高。在接受CRC根治性手术的患者中,GLI1和PTTG1与LNM密切相关,LNM患者的GLI1和PTTG1水平高于无LNM患者。评估CRC患者LNM时GLI1和PTTG1的ROC曲线下面积分别为0.824和0.811。
    结论:GLI1和PTTG1的表达在接受CRC根治术的患者中上调,并且在这些患者中与LNM显著相关。此外,高GLI1和PTTG1表达可提示接受根治性手术的CRC患者存在LNM.这两种基因的表达均具有一定的诊断和治疗意义。
    BACKGROUND: Few studies have investigated the expression of GLI1 and PTTG1 in patients undergoing radical surgery for colorectal carcinoma (CRC) and their association with lymph node metastasis (LNM). Therefore, more relevant studies and analyses need to be conducted.
    OBJECTIVE: To explore GLI1 and PTTG1 expression in patients undergoing radical surgery for CRC and their correlation with LNM.
    METHODS: This study selected 103 patients with CRC admitted to our hospital between April 2020 and April 2023. Sample specimens of CRC and adjacent tissues were collected to determine the positive rates and expression levels of GLI1 and PTTG1. The correlation of the two genes with patients\' clinicopathological data (e.g., LNM) was explored, and differences in GLI1 and PTTG1 expression between patients with LNM and those without were analyzed. Receiver operating characteristic (ROC) curves were plotted to evaluate the predictive potential of the two genes for LNM in patients with CRC.
    RESULTS: Significantly higher positive rates and expression levels of GLI1 and PTTG1 were observed in CRC tissue samples compared with adjacent tissues. GLI1 and PTTG1 were strongly linked to LNM in patients undergoing radical surgery for CRC, with higher GLI1 and PTTG1 levels found in patients with LNM than in those without. The areas under the ROC curve of GLI1 and PTTG1 in assessing LNM in patients with CRC were 0.824 and 0.811, respectively.
    CONCLUSIONS: GLI1 and PTTG1 expression was upregulated in patients undergoing radical surgery for CRC and are significantly related to LNM in these patients. Moreover, high GLI1 and PTTG1 expression can indicate LNM in patients with CRC undergoing radical surgery. The expression of both genes has certain diagnostic and therapeutic significance.
    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
    本研究旨在阐明垂体肿瘤转化基因1(PTTG1)在细胞增殖中的作用,迁移,入侵,和胰腺癌细胞的有氧糖酵解,并评估PTTG1作为治疗靶标的潜力。使用GEPIA数据库分析胰腺癌中的PTTG1表达。在PTTG1敲低的Panc1细胞或PTTG1过表达的Mia-PaCa2细胞中,使用细胞活力曲线和集落形成评估细胞增殖,并进行伤口愈合试验和transwell试验以评估迁移和侵袭,分别。此外,进行蛋白质印迹以评估PTTG1,增殖细胞核抗原,E-cadherin,N-钙黏着蛋白,c-myc同时,葡萄糖的摄取,细胞外酸化率(ECAR),分析了耗氧率(OCR)。我们的结果显示PTTG1在胰腺癌中表达上调,促进细胞增殖。低PTTG1有助于更高的无病生存率和总生存率。在Panc1单元格中,PTTG1敲低导致细胞活力和集落形成降低。在PTTG1敲低的Panc1中,细胞的迁移和侵袭能力也降低。相应地,PTTG1敲低降低c-myc表达,葡萄糖摄取,ECAR,和Panc1细胞中的OCR。在Mia-PaCa2细胞中,PTTG1过表达促进细胞增殖,有氧糖酵解,通过调节c-myc将β-catenin转位到细胞核。总之,PTTG1诱导增殖,迁移,和入侵,并通过调节c-myc促进胰腺癌细胞的有氧糖酵解,证明PTTG1作为治疗靶标的潜力。
    This study aimed to clarify the role of pituitary tumor-transforming gene 1 (PTTG1) in proliferation, migration, invasion, and aerobic glycolysis of pancreatic cancer cells, and evaluate the potential of PTTG1 as a therapeutic target. PTTG1 expression in pancreatic cancers was analyzed using the GEPIA databank. In the Panc1 cell with the PTTG1 knockdown or Mia-PaCa2 cells with PTTG1 overexpression, the cell proliferation was evaluated using cell viability curves and colony formation, and wound heal assay and transwell assay were performed to evaluate the migration and invasion, respectively. Furthermore, a western blot was performed to evaluate the expressions of PTTG1, proliferating cell nuclear antigen, E-cadherin, N-cadherin, and c-myc. Meanwhile, the glucose uptake, extracellular acidification rates (ECAR), and oxygen consumption rates (OCR) were analyzed. Our results showed that PTTG1 expression is upregulated in pancreatic cancer, which promoted cell proliferation. Low PTTG1 contributed to higher disease-free survival and overall survival. In Panc1 cell, PTTG1 knockdown resulted in reduced cell viability and colony formation. The migration and invasion abilities of the cells were also reduced in Panc1 with PTTG1 knockdown. Correspondingly, PTTG1 knockdown decreased c-myc expression, glucose uptake, ECAR, and OCR in Panc1 cells. In Mia-PaCa2 cells, PTTG1 overexpression promoted cell proliferation, aerobic glycolysis, and translocation of β-catenin to the nucleus by regulating c-myc. In conclusion, PTTG1 induces proliferation, migration, and invasion, and promotes aerobic glycolysis in pancreatic cancer cells via regulating c-myc, demonstrating the potential of PTTG1 as a therapeutic target.
    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
    精原细胞瘤是最常见的睾丸癌。垂体肿瘤转化基因1(PTTG1)是一种在几种肿瘤中显示致癌活性的securin。我们先前证明了核PTTG1通过其对基质金属蛋白酶2(MMP-2)和E-cadherin(CDH1)的转录活性来促进精原细胞瘤肿瘤的侵袭。我们想知道特定的相互作用物是否会影响其亚细胞分布。为了这个目标,我们研究了精原细胞瘤细胞系中的PTTG1相互作用体,这些细胞系显示出不同的PTTG1核水平与侵袭特性相关。PTTG1免疫沉淀的蛋白质组学方法揭示了新的特异性securin相互作用物。蛋白质印迹,共聚焦显微镜,细胞质/细胞核分级分离,球体形成测定,进行和Atlas数据库的询问以验证蛋白质组学结果并调查PTTG1和新发现的伴侣之间的相互作用。我们观察到血影蛋白β链(SPTBN1)和PTTG1是辅因子,用SPTBN1将securin锚定在细胞质中。SPTBN1下调决定了PTTG1核易位,提升其侵入能力。此外,缺乏SPTBN1结合的PTTG1缺失突变体强烈定位在细胞核中。Atlas数据库显示,与非精原细胞瘤相比,含有较高核PTTG1水平的精原细胞瘤显示出明显较低的SPTBN1水平。在人类精原细胞瘤标本中,我们发现PTTG1/SPTBN1强烈的共定位在PTTG1核分布区域减少。总的来说,这些结果表明,SPTBN1与PTTG1一起是一个潜在的预后因素,可用于精原细胞瘤的临床治疗.
    Seminoma is the most common testicular cancer. Pituitary tumor-transforming gene 1 (PTTG1) is a securin showing oncogenic activity in several tumors. We previously demonstrated that nuclear PTTG1 promotes seminoma tumor invasion through its transcriptional activity on matrix metalloproteinase 2 (MMP-2) and E-cadherin (CDH1). We wondered if specific interactors could affect its subcellular distribution. To this aim, we investigated the PTTG1 interactome in seminoma cell lines showing different PTTG1 nuclear levels correlated with invasive properties. A proteomic approach upon PTTG1 immunoprecipitation uncovered new specific securin interactors. Western blot, confocal microscopy, cytoplasmic/nuclear fractionation, sphere-forming assay, and Atlas database interrogation were performed to validate the proteomic results and to investigate the interplay between PTTG1 and newly uncovered partners. We observed that spectrin beta-chain (SPTBN1) and PTTG1 were cofactors, with SPTBN1 anchoring the securin in the cytoplasm. SPTBN1 downregulation determined PTTG1 nuclear translocation, promoting its invasive capability. Moreover, a PTTG1 deletion mutant lacking SPTBN1 binding was strongly localized in the nucleus. The Atlas database revealed that seminomas that contained higher nuclear PTTG1 levels showed significantly lower SPTBN1 levels in comparison to non-seminomas. In human seminoma specimens, we found a strong PTTG1/SPTBN1 colocalization that decreases in areas with nuclear PTTG1 distribution. Overall, these results suggest that SPTBN1, along with PTTG1, is a potential prognostic factor useful in the clinical management of seminoma.
    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
    肾脏的形成和肾脏发生是由精确的时空基因表达程序来控制的。受细胞周期协调调节,细胞类型特异性转录因子和表观遗传/染色质调节因子。然而,表观遗传/染色质调节因子在肾脏发育和疾病中的作用尚不清楚.在这项研究中,我们调查了删除染色质重塑因子Smarca4(Brg1)的影响,一个人类Wilms肿瘤相关基因,在表达Wnt4的细胞中。Smarca4缺乏导致严重的肾小管缺损和髓质缩短。通过无偏单细胞RNA测序分析,我们确定了多种类型的Wnt4Cre标记的间质细胞,以及与肾单位相关的细胞.Smarca4缺乏增加间质细胞,但显著减少肾小管细胞,导致细胞具有混合同一性,细胞周期调节因子和与细胞外基质和上皮间质转化/纤维化相关的基因表达升高。我们发现Smarca4丢失诱导癌基因Pttg1的显着上调和Wnt4Cre标记的细胞的过度增殖。细胞状态的这些变化可能会阻碍细胞转变为特征性的管状结构,最终导致纤维化。总之,我们的研究结果揭示了与Wnt4Cre标记细胞相关的新细胞类型和基因,并强调了Smarca4在调节肾小管细胞分化和致癌基因Pttg1表达中的关键作用.这些发现可能为SMARCA4缺乏导致的肾细胞癌的潜在治疗策略提供有价值的见解。
    Kidney formation and nephrogenesis are controlled by precise spatiotemporal gene expression programs, which are coordinately regulated by cell-cycle, cell type-specific transcription factors and epigenetic/chromatin regulators. However, the roles of epigenetic/chromatin regulators in kidney development and disease remain poorly understood. In this study, we investigated the impact of deleting the chromatin remodeling factor Smarca4 (Brg1), a human Wilms tumor-associated gene, in Wnt4-expressing cells. Smarca4 deficiency led to severe tubular defects and a shortened medulla. Through unbiased single-cell RNA sequencing analyses, we identified multiple types of Wnt4 Cre-labeled interstitial cells, along with nephron-related cells. Smarca4 deficiency increased interstitial cells but markedly reduced tubular cells, resulting in cells with mixed identity and elevated expression of cell-cycle regulators and genes associated with extracellular matrix and epithelial-to-mesenchymal transition/fibrosis. We found that Smarca4 loss induced a significant upregulation of the oncogene Pttg1 and hyperproliferation of Wnt4 Cre-labeled cells. These changes in the cellular state could hinder the cellular transition into characteristic tubular structures, eventually leading to fibrosis. In conclusion, our findings shed light on novel cell types and genes associated with Wnt4 Cre-labeled cells and highlight the critical role of Smarca4 in regulating tubular cell differentiation and the expression of the cancer-causing gene Pttg1 in the kidney. These findings may provide valuable insights into potential therapeutic strategies for renal cell carcinoma resulting from SMARCA4 deficiency.
    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
    垂体肿瘤转化基因1(PTTG1)在各种类型的肿瘤中过表达,并作为癌基因发挥作用;它也可能是肿瘤治疗的潜在靶标。同时,胰腺腺癌(PAAD)的高死亡率在很大程度上取决于治疗效果有限.基于PTTG1在癌症治疗中的潜力,本研究探讨了PTTG1对PAAD治疗的影响。癌症基因组图谱计划(TCGA)数据显示,PTTG1的高表达与胰腺癌的临床分期和预后较差有关。此外,CCK-8分析显示,在BxPC-3-PTTG1high和MIAPaCa-2-PTTG1high细胞中,吉西他滨和5-氟尿嘧啶(5-FU)的IC50增加.TIDE算法表明,PTTG1高组的免疫检查点阻断(ICB)效率较差。此外,我们发现OAd5在BxPC-3-PTTG1high和MIAPaCa-2-PTTG1high细胞中的效率增强,而在BxPC-3-PTTG1low和MIAPaCa-2-PTTG1low细胞中的效率较差。我们使用表达GFP的OAd5进行转导。因此,OAd5转导后24小时,BxPC-3-PTTG1high和MIAPaCa-2-PTTG1high细胞的荧光强度增强,而BxPC-3-PTTG1low和MIAPaCa-2-PTTG1low细胞的荧光强度降低。荧光强度表明PTTG1增加了OAd5进入。流式细胞术检测显示OAd5受体CXADR表达被PTTG1增强。在CXADR敲低的情况下,PTTG1未能进一步增强OAd5转导。总之,PTTG1通过增加细胞表面的CXADR表达来增强OAd5向胰腺癌细胞的转导。
    Pituitary tumor-transforming gene 1 (PTTG1) is overexpressed in various types of tumors and functions as an oncogene; it could also be a potential target in tumor therapy. Meanwhile, the high mortality of pancreatic adenocarcinoma (PAAD) largely depends on the limited effectiveness of therapy. Based on the promising potential of PTTG1 in cancer treatment, we explored the influence of PTTG1 on the treatment of PAAD in this study. The Cancer Genome Atlas Program (TCGA) data showed that higher expression of PTTG1 was associated with higher clinical stages and worse prognosis of pancreatic cancer. In addition, the CCK-8 assay showed that the IC50 of gemcitabine and 5-fluorouracil (5-FU) was increased in BxPC-3-PTTG1high and MIA PaCa-2-PTTG1high cells. The TIDE algorithm indicated that the immune checkpoint blockades\' (ICBs) efficiency is poor in the PTTG1 high group. Furthermore, we found that the efficiency of OAd5 was enhanced in BxPC-3-PTTG1high and MIA PaCa-2-PTTG1high cells and poor in BxPC-3-PTTG1low and MIA PaCa-2-PTTG1low cells. We used the OAd5 expressing GFP for transduction. As a result, the fluorescence intensity was enhanced in BxPC-3-PTTG1high and MIA PaCa-2-PTTG1high cells and decreased in BxPC-3-PTTG1low and MIA PaCa-2-PTTG1low cells 24 h after OAd5 transduction. The fluorescence intensity indicated that PTTG1 increased OAd5 entry. The flow cytometry assay showed that OAd5 receptor CXADR expression was enhanced by PTTG1. PTTG1 failed to further enhance OAd5 transduction in the case of CXADR knockdown. In summary, PTTG1 enhanced OAd5 transduction into pancreatic cancer cells by increasing CXADR expression on the cell surface.
    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
    未经证实:据报道,PTTG1与各种癌症的预后和进展有关,包括肾透明细胞癌(KIRC)。在这篇文章中,我们主要研究预后之间的关联,豁免权,KIRC患者的PTTG1。
    UNASSIGNED:我们从TCGA-KIRC数据库下载了转录组数据。使用PCR和免疫组织化学,分别,验证PTTG1在KIRC细胞系和蛋白水平的表达。生存分析以及单变量或多变量Cox风险回归分析用于证明PTTG1单独是否会影响KIRC的预后。最重要的是研讨PTTG1与免疫的关系。
    UNASSIGNED:论文的结果表明,与癌旁正常组织相比,KIRC中PTTG1的表达水平升高,通过PCR和免疫组织化学在细胞系和蛋白质水平上验证(P<0.05)。PTTG1高表达与KIRC患者总生存期(OS)缩短有关(P<0.05)。通过单变量或多元回归分析,PTTG1被证实是KIRCOS的独立预后因素(P<0.05)。并通过基因集富集分析(GSEA;P<0.05)获得了相关的7条通路。此外,在KIRC中发现肿瘤突变负荷(TMB)和免疫与PTTG1显着相关(P<0.05)。PTTG1与免疫治疗反应的相关性提示低PTTG1组对免疫治疗更敏感(P<0.05)。
    未经证实:PTTG1与TMB或免疫力密切相关,对KIRC患者的预后有较好的预测能力。
    UNASSIGNED: PTTG1 has been reported to be linked with the prognosis and progression of various cancers, including kidney renal clear cell carcinoma (KIRC). In this article, we mainly investigated the associations between prognosis, immunity, and PTTG1 in KIRC patients.
    UNASSIGNED: We downloaded transcriptome data from the TCGA-KIRC database. PCR and immunohistochemistry were used, respectively, to validate the expression of PTTG1 in KIRC at the cell line and the protein levels. Survival analyses as well as univariate or multivariate Cox hazard regression analyses were used to prove whether PTTG1 alone could affect the prognosis of KIRC. The most important point was to study the relationship between PTTG1 and immunity.
    UNASSIGNED: The results of the paper revealed that the expression levels of PTTG1 were elevated in KIRC compared with para-cancerous normal tissues, validated by PCR and immunohistochemistry at the cell line and the protein levels (P < 0.05). High PTTG1 expression was related to shorter overall survival (OS) in patients with KIRC (P < 0.05). Through univariate or multivariate regression analysis, PTTG1 was confirmed to be an independent prognostic factor for OS of KIRC (P < 0.05), and its related seven pathways were obtained through gene set enrichment analysis (GSEA; P < 0.05). Moreover, tumor mutational burden (TMB) and immunity were found to be significantly connected with PTTG1 in KIRC (P < 0.05). Correlations between PTTG1 and immunotherapy responses implied that the low-PTTG1 group was more sensitive to immunotherapy (P < 0.05).
    UNASSIGNED: PTTG1 was closely associated with TMB or immunity, and it had a superior ability to forecast the prognosis of KIRC 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
    肿瘤来源的细胞外囊泡(EV)通过携带microRNA(miR)促进卵巢癌(OC)转移。本研究探讨了OC细胞来源的EV携带miR-106a-5p在OC中的作用机制。测量miR-106a-5p在OC组织和细胞中的表达。从SKOV3细胞和正常细胞中提取EV。观察到EV在OC细胞中的内在化。用过表达miR-106a-5p的SKOV3-EV或SKOV3-EV处理OC细胞以检测其增殖,迁移,和入侵。miR-106a-5p的表达水平,检测KLF6和PTTG1并鉴定它们的结合关系。设计联合实验检测KLF6和PTTG1对OC细胞的影响。进行了异种移植肿瘤实验以验证EVs-miR-106a-5p和KLF6在OC转移中的作用机制。因此,miR-106a-5p在OC中增强,与OC转移相关。SKOV3-EV促进了扩散,迁移,和OC细胞的侵袭。机械上,EV将miR-106a-5p携带到其他OC细胞中,抑制KLF6,降低KLF6与PTTG1启动子的结合,并上调PTTG1转录。KLF6的过表达或PTTG1的沉默减弱了EVs-miR-106a-5p对OC细胞的促进作用。EVs-miR-106a-5p通过KLF6/PTTG1轴促进OC转移。最后,OC细胞衍生的EV通过miR-106a-5p/KLF6/PTTG1轴促进OC的进展和转移。
    Tumor-derived extracellular vesicles (EVs) promote ovarian cancer (OC) metastasis by carrying microRNAs (miRs). This study investigated the mechanism of miR-106a-5p carried by OC cell-derived EVs in OC. miR-106a-5p expression in OC tissues and cells was measured. EVs were extracted from SKOV3 cells and normal cells. The internalization of EVs in OC cells was observed. OC cells were treated with SKOV3-EVs or SKOV3-EVs overexpressing miR-106a-5p to detect the proliferation, migration, and invasion. The expression levels of miR-106a-5p, KLF6, and PTTG1 were detected and their binding relationships were identified. Combined experiments were designed to detect the effects of KLF6 and PTTG1 on OC cells. A xenograft tumor experiment was performed to verify the mechanism of EVs-miR-106a-5p and KLF6 in OC metastasis. Consequently, miR-106a-5p was enhanced in OC and correlated with OC metastasis. SKOV3-EVs promoted the proliferation, migration, and invasion of OC cells. Mechanistically, EVs carried miR-106a-5p into other OC cells, inhibited KLF6, reduced the binding of KLF6 to the PTTG1 promoter, and upregulated PTTG1 transcription. Overexpression of KLF6 or silencing of PTTG1 attenuated the promoting effect of EVs-miR-106a-5p on OC cells. EVs-miR-106a-5p facilitated OC metastasis via the KLF6/PTTG1 axis. To conclude, OC cell-derived EVs facilitated the progression and metastasis of OC via the miR-106a-5p/KLF6/PTTG1 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
    垂体肿瘤转化基因1(PTTG1)编码参与许多细胞过程的多功能蛋白。然而,PTTG1在肿瘤形成中的潜在作用及其在人类泛癌症中的预后功能尚不清楚。基因改变的分析,PTTG1表达,预后功能,和PTTG1相关的免疫分析在33种类型的肿瘤是基于各种数据库,如癌症基因组图谱数据库,基因型-组织表达数据库,和人类蛋白质图谱数据库。此外,进行PTTG1相关基因富集分析以研究PTTG1与肿瘤之间的潜在关系和可能的分子机制。PTTG1的过表达可能导致各种肿瘤的形成和不良预后。因此,PTTG1在大多数肿瘤中作为潜在的癌基因。此外,PTTG1与免疫浸润有关,免疫检查点,肿瘤突变负担,和微卫星不稳定。因此,PTTG1可能是肿瘤治疗预后和结果的潜在生物标志物,也可能是肿瘤治疗中的一个有希望的靶标。
    Pituitary tumor-transforming gene 1 (PTTG1) encodes a multifunctional protein that is involved in many cellular processes. However, the potential role of PTTG1 in tumor formation and its prognostic function in human pan-cancer is still unknown. The analysis of gene alteration, PTTG1 expression, prognostic function, and PTTG1-related immune analysis in 33 types of tumors was performed based on various databases such as The Cancer Genome Atlas database, the Genotype-Tissue Expression database, and the Human Protein Atlas database. Additionally, PTTG1-related gene enrichment analysis was performed to investigate the potential relationship and possible molecular mechanisms between PTTG1 and tumors. Overexpression of PTTG1 may lead to tumor formation and poor prognosis in various tumors. Consequently, PTTG1 acts as a potential oncogene in most tumors. Additionally, PTTG1 is related to immune infiltration, immune checkpoints, tumor mutational burden, and microsatellite instability. Thus, PTTG1 could be potential biomarker for both prognosis and outcomes of tumor treatment and it could also be a promising target in tumor 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
    已发现垂体肿瘤转化基因1(PTTG1)与细胞增殖和侵袭的过程有关,在主动脉夹层(AD)中高表达。然而,其在AD中的潜在作用和潜在机制仍不确定。本研究旨在阐明特异性蛋白1(SP1)和PTTG1在AD主动脉血管平滑肌细胞(VSMC)迁移和表型转换中的作用。收集35例AD患者的主动脉样本,通过qPCR检测组织中PTTG1的表达,免疫印迹和免疫荧光。用血小板衍生生长因子-BB(PDGF-BB)刺激人主动脉平滑肌细胞(HAVSMCs),建立AD细胞模型。还通过qPCR和蛋白质印迹检查VSMC中的PTTG1表达。通过CCK-8检测细胞活力,通过EdU染色检测细胞增殖,通过伤口愈合和transwell检测细胞迁移。然后进行Western印迹以测定迁移相关蛋白。干扰PTTG1后,通过qPCR检测平滑肌表型开关标志物平滑肌蛋白22α(SM22-α)和骨桥蛋白(OPN)的水平,免疫印迹和免疫荧光。SP1和PTTG1的结合用双荧光素酶报告基因测定和染色质免疫沉淀测定(ChIP)验证。在AD患者中发现PTTG1过表达。干扰PTTG1减弱PDGF-BB刺激的HAVSMCs的增殖和迁移,除了从收缩表型到合成表型的转换。转录因子SP1在PDGF-BB刺激的HAVSMC中上调,与PTTG1启动子序列结合并调节PTTG1的表达,其过表达逆转了PTTG1干扰对细胞增殖的影响,迁移和表型转换。SP1转录激活PTTG1激活MAPK/ERK信号通路。总之,PTTG1的SP1转录激活通过MAPK信号调节AD中HAVSMCs的迁移和表型转化。
    Pituitary tumor-transforming gene 1 (PTTG1) has been found to be associated with the process of cell proliferation and invasion, and is highly expressed in aortic dissection (AD). However, its potential role and underlying mechanism in AD remain uncertain. This study aims at elucidating the roles of specificity protein 1 (SP1) and PTTG1 in the migration and phenotypic switching of aortic vascular smooth muscle cells (VSMCs) in AD. Aortic samples were collected from 35 patients with AD for examination of PTTG1 expression in the tissues by qPCR, western blot and immunofluorescence. Human aortic vascular smooth muscle cells (HAVSMCs) were stimulated with platelet-derived growth factor-BB (PDGF-BB) to establish the cellular model of AD. PTTG1 expression in VSMCs was also examined by qPCR and western blot. Cell viability was detected by CCK-8, cell proliferation by EdU staining and cell migration by wound healing and transwell. Western blot was then performed to assay migration-related proteins. After interference with PTTG1, the levels of smooth muscle pthenotypic switch markers smooth muscle protein 22 alpha (SM22-α) and osteopontin (OPN) were detected by qPCR, western blot and immunofluorescence. The binding of SP1 and PTTG1 was verified with dual-luciferase reporter assay and chromatin immunoprecipitation assay (ChIP). PTTG1 overexpression was found in AD patients. Interference with PTTG1 attenuated the proliferation and migration of PDGF-BB-stimulated HAVSMCs, in addition to their switching from contractile phenotype to synthetic phenotype. Transcription factor SP1 was up-regulated in PDGF-BB-stimulated HAVSMCs, combined with PTTG1 promoter sequence and regulated PTTG1 expression, whose overexpression reversed the effects of PTTG1 interference on cell proliferation, migration and phenotypic switching. SP1 transcriptional activation of PTTG1 activated MAPK/ERK signaling pathway. In conclusion, SP1 transcriptional activation of PTTG1 regulates the migration and phenotypic transformation of HAVSMCs in AD by MAPK Signaling.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号