Heterogeneous-Nuclear Ribonucleoprotein Group A-B

非均匀核型核糖核蛋白 A - B 组
  • 文章类型: Journal Article
    许多研究表明,m6A在肿瘤的发展中起着重要的调节作用。HNRNPA2B1,m6ARNA甲基化阅读蛋白之一,已被证明在人类癌症中升高。
    在这项研究中,我们旨在确定HNRNPA2B1在乳腺癌中的作用。
    通过RT-qPCR和TCGA数据库研究了乳腺癌中HNRNPA2B1的表达。然后,通过CCK8检测HNRNPA2B1对癌细胞的功能,集落形成和划痕测定。此外,通过Wilcoxon符号秩检验探索BRCA中HNRNPA2B1的表达,KruskalWallis检验和逻辑回归。通过KaplanMeier和Cox回归分析考虑与HNRNPA2B1表达和存活的关联。通过基因集富集分析(GSEA)和ClusterProfilerR软件包分析HNRNPA2B1的生物学功能。
    我们发现HNRNPA2B1在乳腺癌中高表达并诱导细胞增殖和迁移。此外,我们观察到HNRNPA2B1在体内诱导肿瘤生长。此外,我们还发现HNRNPA2B1的表达与乳腺癌患者的特征和预后相关.
    我们的研究结果表明,HNRNPA2B1促进肿瘤生长,并可能作为乳腺癌新的潜在分子标志物发挥作用。
    UNASSIGNED: Numerous studies have shown that m6A plays an important regulatory role in the development of tumors. HNRNPA2B1, one of the m6A RNA methylation reading proteins, has been proven to be elevated in human cancers.
    UNASSIGNED: In this study, we aimed to identify the role of HNRNPA2B1 in breast cancer.
    UNASSIGNED: HNRNPA2B1 expression was investigated via RT-qPCR and TCGA database in breast cancer. Then, the function of HNRNPA2B1 on cancer cell was measured by CCK8 assays, colony formation and scratch assays. In addition, HNRNPA2B1 expression in BRCA was explored via the Wilcoxon signed-rank test, KruskalWallis test and logistic regression. The association with HNRNPA2B1 expression and survival were considered by KaplanMeier and Cox regression analyses. The biological function of HNRNPA2B1 was analyzed via gene set enrichment analysis (GSEA) and the cluster Profiler R software package.
    UNASSIGNED: We found that HNRNPA2B1 was highly expressed and induced cell proliferation and migration in breast cancer. Moreover, we observed HNRNPA2B1 induced tumor growth in vivo. In addition, we also found HNRNPA2B1 expression was associated with characteristics and prognosis in breast cancer patients.
    UNASSIGNED: Our findings suggested that HNRNPA2B1 promoted tumor growth and could function as a new potential molecular marker in breast cancer.
    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
    尽管RNAi和RNA剪接机制参与调节严重急性呼吸综合征冠状病毒2(SARS-CoV-2)的复制,它们在2019年冠状病毒病(COVID-19)发病机制中的确切作用仍不清楚。在这里,我们显示RNAi成分(Dicer和XPO5)和剪接因子(SRSF3和hnRNPA3)表达减少与COVID-19严重程度增加相关。SARS-CoV-2N蛋白诱导Dicer自噬降解,XPO5、SRSF3和hnRNPA3抑制miRNA生物发生和RNA剪接并触发DNA损伤,蛋白毒性应激,和肺炎。Dicer,XPO5、SRSF3和hnRNPA3敲低增加,虽然它们的过度表达减少,N蛋白诱导的肺炎的严重程度。年龄较大的小鼠表现出较低的Dicer表达,XPO5,SRSF3和hnRNPA3在其肺组织中表现出比年轻小鼠更严重的N蛋白诱导的肺炎。PJ34,一种聚(ADP-核糖)聚合酶抑制剂,或者阿那曲唑,芳香化酶抑制剂,通过恢复Dicer改善N蛋白或SARS-CoV-2引起的肺炎,XPO5、SRSF3和hnRNPA3表达。这些发现将有助于开发SARS-CoV-2相关肺炎的改进治疗方法。
    Though RNAi and RNA-splicing machineries are involved in regulating severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) replication, their precise roles in coronavirus disease 2019 (COVID-19) pathogenesis remain unclear. Herein, we show that decreased RNAi component (Dicer and XPO5) and splicing factor (SRSF3 and hnRNPA3) expression correlate with increased COVID-19 severity. SARS-CoV-2 N protein induces the autophagic degradation of Dicer, XPO5, SRSF3, and hnRNPA3, inhibiting miRNA biogenesis and RNA splicing and triggering DNA damage, proteotoxic stress, and pneumonia. Dicer, XPO5, SRSF3, and hnRNPA3 knockdown increases, while their overexpression decreases, N protein-induced pneumonia\'s severity. Older mice show lower expression of Dicer, XPO5, SRSF3, and hnRNPA3 in their lung tissues and exhibit more severe N protein-induced pneumonia than younger mice. PJ34, a poly(ADP-ribose) polymerase inhibitor, or anastrozole, an aromatase inhibitor, ameliorates N protein- or SARS-CoV-2-induced pneumonia by restoring Dicer, XPO5, SRSF3, and hnRNPA3 expression. These findings will aid in developing improved treatments for SARS-CoV-2-associated pneumonia.
    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
    喉鳞状细胞癌(LSCC)是一种常见的人类癌症,其发病机制复杂,尚未完全了解。这里,我们揭示了与LSCC肿瘤发生和进展相关的长链非编码RNA(lncRNA)。LOC730101在人LSCC组织中表现出显著的过表达,LOC730101水平升高与恶性临床病理特征相关。此外,我们证明LOC730101以hnRNPA2B1依赖性方式封装到外泌体中,作为区分LSCC患者与健康个体的有希望的血浆生物标志物(AUC=0.92,灵敏度为89.36%,特异性为86.36%).来自LSCC细胞的外泌体增强了活力,DNA合成率,正常鼻咽上皮细胞的侵袭性,在LOC730101过表达时观察到明显的效果。此外,外泌体LOC730101促进体内肿瘤生长。机械上,正常鼻咽上皮细胞的外泌体LOC730101内化通过与hnRNPA2B1的直接相互作用导致p38MAPKγ(p38γ)启动子上的H3K4me3水平升高。这种相互作用激活p38γ转录,最终推动LSCC肿瘤发生。总的来说,我们的发现揭示了一种新的外泌体lncRNA,它在LSCC癌变过程中介导正常细胞和LSCC细胞之间的通讯,提示靶向LOC730101可能是LSCC治疗的一种有希望的治疗策略.
    Laryngeal squamous cell carcinoma (LSCC) is a prevalent human cancer with a complex pathogenesis that remains incompletely understood. Here, we unveil a long non-coding RNA (lncRNA) associated with LSCC tumorigenesis and progression. LOC730101 exhibits significant overexpression in human LSCC tissues, and elevated LOC730101 levels correlate with malignant clinicopathological characteristics. Moreover, we demonstrate that LOC730101 is encapsulated into exosomes in an hnRNPA2B1-dependent manner, serving as a promising plasma biomarker for discriminating LSCC patients from healthy individuals (AUC = 0.92 with 89.36% sensitivity and 86.36% specificity). Exosomes derived from LSCC cells enhance the viability, DNA synthesis rate, and invasiveness of normal nasopharynx epithelial cells, with pronounced effects observed upon LOC730101 overexpression. Additionally, exosomal LOC730101 promotes tumor growth in vivo. Mechanistically, exosomal LOC730101 internalization by normal nasopharynx epithelial cells leads to increased H3K4me3 levels on the p38 MAPK gamma (p38γ) promoter via direct interaction with hnRNPA2B1. This interaction activates p38γ transcription, ultimately driving LSCC tumorigenesis. Collectively, our findings uncover a novel exosomal lncRNA that mediates communication between normal and LSCC cells during LSCC carcinogenesis, suggesting that targeting LOC730101 may represent a promising therapeutic strategy for LSCC treatment.
    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:骨关节炎(OA)是一种进行性疾病,影响缺乏有效治疗的关节。然而,潜在的分子机制尚未完全阐明。
    方法:通过单碘乙酸盐(MIA)关节内注射建立SD大鼠OA模型。Western印迹分析用于鉴定关节软骨中UBE2I和hnRNPA2B1的水平。构建了UBE2I的过表达和siRNA载体,并将其转染到大鼠软骨细胞中。CCK-8,TUNEL和transwell测定用于评估细胞活力,细胞凋亡和迁移能力。蛋白质印迹分析用于确定软骨形成特异性基因的水平,包括SOX9,COL2A1,Aggrecan,PRG4。然后,通过免疫沉淀证实了分子相互作用。
    结果:我们观察到在OA的关节软骨样品中UBE2I和hnRNPA2B1表达的显著上调。Pearson相关分析显示UBE2I与hnRNPA2B1水平呈正相关。功能实验表明UBE2I表达增加显著抑制细胞生长,迁移,并减少软骨形成特异性基因的表达,而降低UBE2I水平则有相反的效果。通过共定位和免疫沉淀确定UBE2I和hnRNPA2B1之间的分子相互作用。使用hnRNPA2B1抗体通过免疫沉淀富集SUMO1和SUMO3蛋白。使用SUMO化抑制剂(2-D08)和SUMO化活化剂(N106)进行拯救实验。过表达UBE2I使hnRNPA2B1在胞浆中的表达水平增高,在细胞核中的表达水平下降,通过2-D08的治疗被逆转。相反,UBE2I敲低和N106处理具有相反的效果。
    结论:UBE2I通过促进OA中的SUMO化调节hnRNPA2B1的核转位。
    BACKGROUND: Osteoarthritis (OA) is a progressive condition affecting the joints that lacking effective therapy. However, the underlying molecular mechanism has not been fully clarified.
    METHODS: A model of OA was established in Sprague-Dawley (SD) rats through intra-articularly injected with monoiodoacetate (MIA). Western blot analysis was used to identify the levels of UBE2I and hnRNPA2B1 in articular cartilage. Overexpression and siRNA vectors for UBE2I were constructed and transfected into rat chondrocytes. CCK-8, TUNEL and transwell assay were utilized to assess the cell viability, apoptosis and migration ability. Western blot analysis was used to determine the levels of chondrogenic-specific genes including SOX9, COL2A1, Aggrecan, and PRG4. Then, molecular interactions were confirmed by immunoprecipitation.
    RESULTS: We observed significant upregulation of UBE2I and hnRNPA2B1 expression in articular cartilage samples of OA. The Pearson correlation analysis revealed positive correlation between UBE2I and hnRNPA2B1 levels. Functional experiments showed that increased UBE2I expression significantly suppressed cell growth, migration, and reduced the expression of chondrogenic-specific genes, while decreasing UBE2I levels had the opposite effects. Molecular interactions between UBE2I and hnRNPA2B1were determined via co-localization and immunoprecipitation. SUMO1 and SUMO3 proteins were enriched by immunoprecipitation using hnRNPA2B1 antibodies. Rescue experiments were performed using SUMOylation inhibitor (2-D08) and SUMOylation activator (N106). Overexpression of UBE2I increased the expression of hnRNPA2B1 in the cytoplasm and decreased the level in the nucleus, which was reversed by the treatment of 2-D08. Conversely, UBE2I knockdown and N106 treatment had the opposite effect.
    CONCLUSIONS: UBE2I modulated the nuclear translocation of hnRNPA2B1 by promoting SUMOylation in OA.
    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
    幽门螺杆菌(H.pylori)感染是胃癌(GC)发病的主要危险因素。N6-甲基腺苷(m6A)在mRNA代谢中起关键作用,hnRNPA2B1作为m6A读数器显示在癌症中发挥m6A依赖性mRNA稳定作用。本研究旨在探讨hnRNPA2B1在幽门螺杆菌相关GC中的作用及其新的分子机制。多个数据集和组织微阵列用于评估hnRNPA2B1表达对幽门螺杆菌感染的反应及其在GC患者中的临床预后。hnRNPA2B1的作用是通过各种技术研究,包括葡萄糖代谢分析,m6A-表观基因组微阵列,Ribo-seq,多聚体谱分析,RIP-seq.此外,使用质谱和co-IP验证hnRNPA2B1与poly(A)结合蛋白细胞质1(PABPC1)的相互作用。这些结果表明hnRNPA2B1在GC中上调并且与不良预后相关。幽门螺杆菌感染通过向其启动子募集NF-κB诱导hnRNPA2B1上调。有趣的是,细胞质锚定的hnRNPA2B1协调PABPC1以稳定其与帽结合eIF4F复合物的关系,这促进了CIP2A的翻译,DLAT和GPX1独立于m6A修饰。总之,hnRNPA2B1通过与PABPC1-eIF4F复合物相互作用促进GC进展中表观遗传mRNA的非m6A翻译,并预测GC患者的不良预后。
    Helicobacter pylori (H. pylori) infection is the primary risk factor for the pathogenesis of gastric cancer (GC). N6-methyladenosine (m6A) plays pivotal roles in mRNA metabolism and hnRNPA2B1 as an m6A reader is shown to exert m6A-dependent mRNA stabilization in cancer. This study aims to explore the role of hnRNPA2B1 in H. pylori-associated GC and its novel molecular mechanism. Multiple datasets and tissue microarray are utilized for assessing hnRNPA2B1 expression in response to H. pylori infection and its clinical prognosis in patients with GC. The roles of hnRNPA2B1 are investigated through a variety of techniques including glucose metabolism analysis, m6A-epitranscriptomic microarray, Ribo-seq, polysome profiling, RIP-seq. In addition, hnRNPA2B1 interaction with poly(A) binding protein cytoplasmic 1 (PABPC1) is validated using mass spectrometry and co-IP. These results show that hnRNPA2B1 is upregulated in GC and correlated with poor prognosis. H. pylori infection induces hnRNPA2B1 upregulation through recruiting NF-κB to its promoter. Intriguingly, cytoplasm-anchored hnRNPA2B1 coordinated PABPC1 to stabilize its relationship with cap-binding eIF4F complex, which facilitated the translation of CIP2A, DLAT and GPX1 independent of m6A modification. In summary, hnRNPA2B1 facilitates the non-m6A translation of epigenetic mRNAs in GC progression by interacting with PABPC1-eIF4F complex and predicts poor prognosis for patients with GC.
    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
    外写组学代表了基因表达调控的另一层。具体来说,N6-甲基腺苷(m6A)调节RNA成熟,稳定性,降解,和翻译。关于microRNAs(miRNAs),虽然有报道称M6A会影响它们的生物发生,对成熟miRNAs的功能影响尚不清楚。这里,我们发现特定miRNAs上的m6A修饰减弱了它们与AGO2的偶联,损害了它们在靶mRNAs上的功能,确定它们进入细胞外囊泡(EV)的递送,并向接收细胞提供功能信息。机械上,细胞内功能损伤是由m6A介导的AGO2/miRNA相互作用的抑制引起的,通过RNA结合蛋白(RBP)hnRNPA2B1的m6A介导的识别,EV加载是有利的,并且EV-miRNA在接受细胞中的功能需要其FTO介导的去甲基化。因此,细胞表达不影响内源性转录物但为细胞间通讯提供调控信息的特定miRNA。这突出表明,当将细胞动力学与特定miRNA相关联时,应考虑进一步的复杂性水平。
    Epitranscriptomics represents a further layer of gene expression regulation. Specifically, N6-methyladenosine (m6A) regulates RNA maturation, stability, degradation, and translation. Regarding microRNAs (miRNAs), while it has been reported that m6A impacts their biogenesis, the functional effects on mature miRNAs remain unclear. Here, we show that m6A modification on specific miRNAs weakens their coupling to AGO2, impairs their function on target mRNAs, determines their delivery into extracellular vesicles (EVs), and provides functional information to receiving cells. Mechanistically, the intracellular functional impairment is caused by m6A-mediated inhibition of AGO2/miRNA interaction, the EV loading is favored by m6A-mediated recognition by the RNA-binding protein (RBP) hnRNPA2B1, and the EV-miRNA function in the receiving cell requires their FTO-mediated demethylation. Consequently, cells express specific miRNAs that do not impact endogenous transcripts but provide regulatory information for cell-to-cell communication. This highlights that a further level of complexity should be considered when relating cellular dynamics to specific miRNAs.
    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:血管生成拟态(VM)是一种神秘的生理特征,它影响胶质母细胞瘤(GBM)肿瘤的持续生长的血液供应。以前的研究确定NFATC3,FOSL1和HNRNPA2B1是VEGFR2的重要介导因子,它们的分子关系可能对GBM中的VM至关重要。
    目的:本研究的目的是了解NFATC3,FOSL1和HNRNPA2B1如何共同影响GBM中的VM。
    方法:我们在体外和体内研究了GBM细胞系U251和U373中VM的潜在基因调控机制。进行体外基于细胞的测定以探索NFATC3,FOSL1和HNRNPA2B1在GBM细胞增殖中的作用,VM和迁移,在RNA干扰(RNAi)介导的敲低以及相应的对照的背景下。使用蛋白质印迹和qRT-PCR测定来检查VEGFR2表达水平。CO-IP用于检测蛋白质-蛋白质相互作用,ChIP用于检测DNA-蛋白质复合物,RIP用于检测RNA-蛋白质复合物。组织化学染色用于检测体内VM管的形成。
    结果:关注NFATC3,FOSL1和HNRNPA2B1,我们发现它们在GBM中都显着上调,并且与U251和U373细胞系中的VM样细胞行为呈正相关。NFATC3,FOSL1或HNRNPA2B1的敲除均导致VEGFR2水平降低,VEGFR2是驱动VM的关键生长因子基因,以及抑制增殖,细胞迁移和体外VM活性。染色质免疫沉淀(ChIP)研究和荧光素酶报告基因测定显示,NFATC3与VEGFR2的启动子区结合以增强VEGFR2基因表达。值得注意的是,FOSL1作为辅因子与NFATC3相互作用,以增强NFATC3的DNA结合能力,从而增强VM样细胞行为。此外,细胞中NFATC3蛋白的水平通过HNRNPA2B1与NFATC3mRNA的结合而增强。此外,RNAi介导的GBM细胞中NFATC3,FOSL1和HNRNPA2B1的沉默降低了其体内肿瘤形成和VM样行为的能力。
    结论:综合来看,我们的研究发现NFATC3通过其与HNRNPA2B1和FOSL1的分子和上位性相互作用来影响VEGFR2的表达和VM样细胞行为,从而确定NFATC3是GBM肿瘤生长的重要介质。
    BACKGROUND: Vasculogenic mimicry (VM) is an enigmatic physiological feature that influences blood supply within glioblastoma (GBM) tumors for their sustained growth. Previous studies identify NFATC3, FOSL1 and HNRNPA2B1 as significant mediators of VEGFR2, a key player in vasculogenesis, and their molecular relationships may be crucial for VM in GBM.
    OBJECTIVE: The aim of this study was to understand how NFATC3, FOSL1 and HNRNPA2B1 collectively influence VM in GBM.
    METHODS: We have investigated the underlying gene regulatory mechanisms for VM in GBM cell lines U251 and U373 in vitro and in vivo. In vitro cell-based assays were performed to explore the role of NFATC3, FOSL1 and HNRNPA2B1 in GBM cell proliferation, VM and migration, in the context of RNA interference (RNAi)-mediated knockdown alongside corresponding controls. Western blotting and qRT-PCR assays were used to examine VEGFR2 expression levels. CO-IP was employed to detect protein-protein interactions, ChIP was used to detect DNA-protein complexes, and RIP was used to detect RNA-protein complexes. Histochemical staining was used to detect VM tube formation in vivo.
    RESULTS: Focusing on NFATC3, FOSL1 and HNRNPA2B1, we found each was significantly upregulated in GBM and positively correlated with VM-like cellular behaviors in U251 and U373 cell lines. Knockdown of NFATC3, FOSL1 or HNRNPA2B1 each resulted in decreased levels of VEGFR2, a key growth factor gene that drives VM, as well as the inhibition of proliferation, cell migration and extracorporeal VM activity. Chromatin immunoprecipitation (ChIP) studies and luciferase reporter gene assays revealed that NFATC3 binds to the promoter region of VEGFR2 to enhance VEGFR2 gene expression. Notably, FOSL1 interacts with NFATC3 as a co-factor to potentiate the DNA-binding capacity of NFATC3, resulting in enhanced VM-like cellular behaviors. Also, level of NFATC3 protein in cells was enhanced through HNRNPA2B1 binding of NFATC3 mRNA. Furthermore, RNAi-mediated silencing of NFATC3, FOSL1 and HNRNPA2B1 in GBM cells reduced their capacity for tumor formation and VM-like behaviors in vivo.
    CONCLUSIONS: Taken together, our findings identify NFATC3 as an important mediator of GBM tumor growth through its molecular and epistatic interactions with HNRNPA2B1 and FOSL1 to influence VEGFR2 expression and VM-like cellular behaviors.
    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
    转移是晚期结直肠癌(CRC)治疗失败的关键因素之一。转移性CRC经常对化学治疗剂产生抗性。这项研究旨在从涉及肿瘤转移和化学抗性的长链非编码RNA(lncRNAs)编码的“隐藏”蛋白中鉴定新的调节因子。方法:采用CRISPR/Cas9文库功能筛查来鉴定高侵袭性CRC模型中癌症转移的关键抑制剂。西方印迹,免疫荧光染色,入侵,迁移,伤口愈合,WST-1,集落形成,功能增益和丧失实验,体内实验转移模型,多重免疫组织化学染色,免疫组织化学,qRT-PCR,和RT-PCR用于评估FOXP3,PRDM16-DT,HNRNPA2B1和L-CHEK2。RNA测序,免疫共沉淀,qRT-PCR,RT-PCR,RNA亲和纯化,RNA免疫沉淀,MeRIP-定量PCR,荧光原位杂交,进行染色质免疫沉淀和荧光素酶报告基因测定以获得对PRDM16-DT在癌症转移和化学耐药性中的作用的机制见解。通过体内选择建立了耐奥沙利铂的CRC细胞系。WST-1,集落形成,入侵,迁移,Biacore技术,使用功能增益和功能丧失实验以及体内实验转移模型来确定升麻苷H-1在CRC中的功能和机制。结果:新蛋白PRDM16-DT,由LINC00982编码,被鉴定为癌症转移和化学耐药抑制因子。PRDM16-DT水平下调与CRC患者的恶性表型和不良预后呈正相关。在FOXP3的转录调控下,PRDM16-DT直接与HNRNPA2B1相互作用,并竞争性降低HNRNPA2B1与CHEK2外显子9的结合,从而形成长CHEK2(L-CHEK2),随后促进E-cadherin分泌。PRDM16-DT诱导的E-cadherin分泌抑制成纤维细胞活化,进而通过减少MMP9分泌抑制CRC转移。菊苣苷H-1,一种天然化合物,可以结合FOXP3的LEU89,HIS91和LEU92,并显着上调PRDM16-DT表达以抑制CRC转移并逆转奥沙利铂耐药。结论:lncRNALINC00982可表达一种新的蛋白PRDM16-DT,在CRC的肿瘤转移和耐药中起新的调控作用。升麻苷H-1可作用于PRDM16-DT信号通路的上游,减轻肿瘤化疗耐药。
    Metastasis is one of the key factors of treatment failure in late-stage colorectal cancer (CRC). Metastatic CRC frequently develops resistance to chemotherapeutic agents. This study aimed to identify the novel regulators from \"hidden\" proteins encoded by long noncoding RNAs (lncRNAs) involved in tumor metastasis and chemoresistance. Methods: CRISPR/Cas9 library functional screening was employed to identify the critical suppressor of cancer metastasis in highly invasive CRC models. Western blotting, immunofluorescence staining, invasion, migration, wound healing, WST-1, colony formation, gain- and loss-of-function experiments, in vivo experimental metastasis models, multiplex immunohistochemical staining, immunohistochemistry, qRT-PCR, and RT-PCR were used to assess the functional and clinical significance of FOXP3, PRDM16-DT, HNRNPA2B1, and L-CHEK2. RNA-sequencing, co-immunoprecipitation, qRT-PCR, RT-PCR, RNA affinity purification, RNA immunoprecipitation, MeRIP-quantitative PCR, fluorescence in situ hybridization, chromatin immunoprecipitation and luciferase reporter assay were performed to gain mechanistic insights into the role of PRDM16-DT in cancer metastasis and chemoresistance. An oxaliplatin-resistant CRC cell line was established by in vivo selection. WST-1, colony formation, invasion, migration, Biacore technology, gain- and loss-of-function experiments and an in vivo experimental metastasis model were used to determine the function and mechanism of cimicifugoside H-1 in CRC. Results: The novel protein PRDM16-DT, encoded by LINC00982, was identified as a cancer metastasis and chemoresistance suppressor. The down-regulated level of PRDM16-DT was positively associated with malignant phenotypes and poor prognosis of CRC patients. Transcriptionally regulated by FOXP3, PRDM16-DT directly interacted with HNRNPA2B1 and competitively decreased HNRNPA2B1 binding to exon 9 of CHEK2, resulting in the formation of long CHEK2 (L-CHEK2), subsequently promoting E-cadherin secretion. PRDM16-DT-induced E-cadherin secretion inhibited fibroblast activation, which in turn suppressed CRC metastasis by decreasing MMP9 secretion. Cimicifugoside H-1, a natural compound, can bind to LEU89, HIS91, and LEU92 of FOXP3 and significantly upregulated PRDM16-DT expression to repress CRC metastasis and reverse oxaliplatin resistance. Conclusions: lncRNA LINC00982 can express a new protein PRDM16-DT to function as a novel regulator in cancer metastasis and drug resistance of CRC. Cimicifugoside H-1 can act on the upstream of the PRDM16-DT signaling pathway to alleviate cancer chemoresistance.
    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
    1型胰岛素样生长因子受体(IGF1R)在肿瘤中发挥重要作用,然而,IGF1R的N6-甲基腺苷(m6A)等转录后调控仍不清楚。这里,我们揭示了lncRNA在癌症中下调的RNA(DRAIC)抑制透明细胞肾癌(ccRCC)中的肿瘤生长和转移的作用。机械上,DRAIC与异质核核糖核蛋白A2B1(hnRNPA2B1)物理相互作用,并通过阻断E3连接酶F-box蛋白11(FBXO11)介导的泛素化和蛋白酶体依赖性降解来增强其蛋白质稳定性。随后,hnRNPA2B1使m6A修饰的IGF1R不稳定,导致ccRCC进展的抑制。此外,四个m6A修饰位点被鉴定为负责IGF1R的mRNA降解。总的来说,我们的发现表明,DRAIC/hnRNPA2B1轴以m6A依赖性方式调节IGF1RmRNA的稳定性,并强调了IGF1R命运的重要机制。这些发现揭示了DRAIC/hnRNPA2B1/FBXO11/IGF1R轴作为ccRCC的潜在治疗靶标,并在m6A修饰的RNA和泛素修饰的蛋白质之间建立了分子命运的联系。
    Type 1 insulin-like growth factor receptor (IGF1R) plays an important role in cancer, however, posttranscriptional regulation such as N6-methyladenosine (m6A) of IGF1R remains unclear. Here, we reveal a role for a lncRNA Downregulated RNA in Cancer (DRAIC) suppress tumor growth and metastasis in clear cell Renal Carcinoma (ccRCC). Mechanistically, DRAIC physically interacts with heterogeneous nuclear ribonucleoprotein A2B1 (hnRNPA2B1) and enhances its protein stability by blocking E3 ligase F-box protein 11 (FBXO11)-mediated ubiquitination and proteasome-dependent degradation. Subsequently, hnRNPA2B1 destabilizes m6A modified-IGF1R, leading to inhibition of ccRCC progression. Moreover, four m6A modification sites are identified to be responsible for the mRNA degradation of IGF1R. Collectively, our findings reveal that DRAIC/hnRNPA2B1 axis regulates IGF1R mRNA stability in an m6A-dependent manner and highlights an important mechanism of IGF1R fate. These findings shed light on DRAIC/hnRNPA2B1/FBXO11/IGF1R axis as potential therapeutic targets in ccRCC and build a link of molecular fate between m6A-modified RNA and ubiquitin-modified protein.
    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
    RNA加工是一种必要的转录后现象,在翻译之前提供了转录物多样性的必要复杂性。这个过程中的畸变可能会导致肿瘤发生,我们以前报道过骨巨细胞瘤(GCTB)的剪接改变增加,其在编码取代色氨酸的甘氨酸34的组蛋白变体H3.3中携带突变(H3.3-G34W)。G34W与几种剪接因子相互作用,最值得注意的是反式作用剪接因子hnRNPA1L2。为了更深入地了解H3.3-G34W在GCTB和等基因HeLa细胞中的RNA加工,我们从hnRNPA1L2和H3.3-G34W相关RNA产生RNA免疫沉淀测序数据,这表明80%在基因区域重叠,并经常被注释为E2F转录因子结合位点。具有H3.3-G34W的GCTB和HeLa细胞中的剪接畸变均显着富集了已知的hnRNPA1L2结合基序(p值<0.01)。这种剪接畸变不同于hnRNPA1L2敲除,显示独立于H3.3-G34W的改变。具有功能意义,hnRNPA1L2被重新分配以紧密匹配H3.3模式,可能由G34W驱动,和正常亲本细胞中未占据的基因座。一起来看,我们的数据显示hnRNPA1L2和H3.3-G34W之间存在功能重叠,可能对GCTB发病过程中的RNA加工产生显著影响.这为未来的操作方式提供了新的治疗干预机会。
    RNA processing is an essential post-transcriptional phenomenon that provides the necessary complexity of transcript diversity prior to translation. Aberrations in this process could contribute to tumourigenesis, and we have previously reported increased splicing alterations in giant cell tumor of bone (GCTB), which carries mutations in the histone variant H3.3 encoding glycine 34 substituted for tryptophan (H3.3-G34W). G34W interacts with several splicing factors, most notably the trans-acting splicing factor hnRNPA1L2. To gain a deeper understanding of RNA processing in GCTB and isogenic HeLa cells with H3.3-G34W, we generated RNA-immunoprecipitation sequencing data from hnRNPA1L2 and H3.3-G34W associated RNAs, which showed that 80% overlapped across genic regions and were frequently annotated as E2F transcription factor binding sites. Splicing aberrations in both GCTB and HeLa cells with H3.3-G34W were significantly enriched for known hnRNPA1L2 binding motifs (p value < 0.01). This splicing aberration differed from hnRNPA1L2 knockouts, which showed alterations independent of H3.3-G34W. Of functional significance, hnRNPA1L2 was redistributed to closely match the H3.3 pattern, likely driven by G34W, and to loci not occupied in normal parental cells. Taken together, our data reveal a functional overlap between hnRNPA1L2 and H3.3-G34W with likely significant consequences for RNA processing during GCTB pathogenesis. This provides novel opportunities for therapeutic intervention in future modus operandi.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号