PTBP1

PTBP1
  • 文章类型: Journal Article
    嘧啶束结合蛋白1(PTBP1)在肿瘤进展和神经发生过程中调节多种可变剪接事件。以前,据报道,PTBP1下调可将星形胶质细胞转化为功能性神经元;然而,PTBP1如何调节星形胶质细胞生理学尚不清楚.在这项研究中,我们发现PTBP1通过ATP1a2调节谷氨酸的摄取,ATP1a2是Na+/K+-ATP酶的成员,和星形胶质细胞中的谷氨酸转运蛋白。Ptbp1敲低改变线粒体功能和能量代谢,其中PTBP1通过琥珀酸脱氢酶(SDH)/Nrf2途径调节线粒体氧化还原稳态。Ptbp1敲低后谷氨酸转运蛋白的功能障碍导致皮质中兴奋性突触传递增强。值得注意的是,我们开发了仿生阳离子三嵌段多肽系统,即,聚乙二醇44-polylysine30-polyleucine10(PEG44-PLL30-PLLeu10)与星形细胞膜涂层在体外和体内递送Ptbp1siRNA,这种方法允许Ptbp1siRNA有效地穿过血脑屏障并靶向大脑中的星形胶质细胞。总的来说,我们的研究结果提出了一个框架,PTBP1作为谷氨酸运输机制的调节剂,并表明仿生方法是体内siRNA递送的有希望的途径。
    Polypyrimidine tract-binding protein 1 (PTBP1) regulates numerous alternative splicing events during tumor progression and neurogenesis. Previously, PTBP1 downregulation was reported to convert astrocytes into functional neurons; however, how PTBP1 regulates astrocytic physiology remains unclear. In this study, we revealed that PTBP1 modulated glutamate uptake via ATP1a2, a member of Na+/K+-ATPases, and glutamate transporters in astrocytes. Ptbp1 knockdown altered mitochondrial function and energy metabolism, which involved PTBP1 regulating mitochondrial redox homeostasis via the succinate dehydrogenase (SDH)/Nrf2 pathway. The malfunction of glutamate transporters following Ptbp1 knockdown resulted in enhanced excitatory synaptic transmission in the cortex. Notably, we developed a biomimetic cationic triblock polypeptide system, i.e., polyethylene glycol44-polylysine30-polyleucine10 (PEG44-PLL30-PLLeu10) with astrocytic membrane coating to deliver Ptbp1 siRNA in vitro and in vivo, which approach allowed Ptbp1 siRNA to efficiently cross the blood-brain barrier and target astrocytes in the brain. Collectively, our findings suggest a framework whereby PTBP1 serves as a modulator in glutamate transport machinery, and indicate that biomimetic methodology is a promising route for in vivo siRNA delivery.
    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
    心肌细胞中基因的异常表达已被揭示为病理性心肌肥大的基本本质。然而,详细的机制还没有完全理解。与心脏肥大有关的基因表达的潜在调节因子仍有待进一步鉴定。这里,我们报道了RNA结合蛋白RNA结合基序蛋白4(RBM4)作为内源性保护剂,能够在体外对抗心肌细胞肥大.在血管紧张素II(AngII)的促肥大刺激下,心肌细胞和心肌中RBM4的蛋白质水平升高。敲除RBM4可进一步加重心肌细胞肥大,而RBM4的过表达抑制心肌细胞肥大。机械上,RBM4定位于细胞核中,下调聚嘧啶束结合蛋白1(PTBP1)的表达,已被证明会加重心肌细胞肥大。此外,我们认为心肌细胞肥大中RBM4的上调是由N6-甲基腺苷(m6A)引起的。AngII诱导RBM4mRNA的m6A甲基化,进一步增强含YTH域的家族蛋白1(YTHDF1)介导的RBM4翻译。因此,我们的研究结果揭示了一种由M6A组成的新通路,RBM4和PTBP1参与心肌细胞肥大。
    Aberrant gene expression in cardiomyocyte has been revealed to be the fundamental essence of pathological cardiac hypertrophy. However, the detailed mechanisms are not fully understood. The underlying regulators of gene expression involved in cardiac hypertrophy remain to be further identified. Here, we report that the RNA-binding protein RNA-binding motif protein 4 (RBM4) functions as an endogenic protector that is able to fight against cardiomyocyte hypertrophy in vitro. Under pro-hypertrophic stimulation of angiotensin II (Ang II), the protein level of RBM4 in cardiomyocyte and myocardium is elevated. Knockdown of RBM4 can further aggravate cardiomyocyte hypertrophy, while over-expression of RBM4 represses cardiomyocyte hypertrophy. Mechanistically, RBM4 is localized in the nucleus and down-regulates the expression of polypyrimidine tract-binding protein 1 (PTBP1), which has been shown to aggravate cardiomyocyte hypertrophy. In addition, we suggest that the up-regulation of RBM4 in cardiomyocyte hypertrophy is caused by N6-methyladenosine (m6A). Ang II induces m6A methylation of RBM4 mRNA, which further enhances the YTH domain-containing family protein 1 (YTHDF1)-mediated translation of RBM4. Thus, our results reveal a novel pathway consisting of m6A, RBM4 and PTBP1, which is involved in cardiomyocyte hypertrophy.
    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
    基因共表达网络可能编码迄今为止尚未充分认识到的成人神经胶质瘤的脆弱性。通过确定EGFR(EM)或PDGFRA(PM)周围的进化保守基因共表达模块,我们最近提出了EM/PM分类方案,将IDH-野生型胶质母细胞瘤(GBM)分配到神经干细胞区室中的EM亚型中,IDH突变型星形细胞瘤和少突胶质细胞瘤进入PM亚型的早期少突胶质细胞谱系。这里,我们报道了EM/PM亚型特异性基因共表达网络的鉴定以及hub基因多嘧啶束结合蛋白1(PTBP1)作为IDH野生型GBM中不依赖基因组改变的易损性的特征.由EM/PM分类方案监督,我们应用加权基因共表达网络分析来鉴定亚型特异性全局基因共表达模块.这些基因共表达模块的特征在于它们的临床相关性,脑发育过程中的细胞起源和保守表达模式。使用慢病毒载体介导的组成型或诱导型敲除,我们表征了PTBP1对IDH野生型GBM细胞存活的影响,PTBP1抑制剪接模式的分析和剪接靶神经元特异性CDC42(CDC42-N)同工型的过表达。成人神经胶质瘤的转录组可以被稳健地分配到4个大的基因共表达模块中,这些模块在预后上是相关的,并且源自EM/PM亚型的恶性细胞或肿瘤微环境。EM亚型与参与前mRNA剪接的恶性细胞固有基因模块相关,DNA复制和损伤反应,和染色体分离,以及主要参与细胞外基质组织和浸润免疫细胞的微环境衍生基因模块。PM亚型与两个主要参与转录调控和mRNA翻译的恶性细胞固有基因模块相关。分别。这些基因模块的表达水平是独立的预后因素,恶性细胞固有基因模块在脑发育过程中是保守的。专注于EM子类型,我们确定PTBP1是恶性细胞固有基因模块最重要的中心.PTBP1在大多数神经胶质瘤基因组中没有改变。PTBP1抑制CDC42-N的保守剪接。PTBP1敲低或CDC42-N过表达破坏肌动蛋白细胞骨架动力学,引起活性氧积累和细胞凋亡。PTBP1介导的CDC42-N剪接的抑制代表了一个潜在的基因组改变无关,IDH野生型GBM中发育保守的脆弱性。
    Gene co-expression networks may encode hitherto inadequately recognized vulnerabilities for adult gliomas. By identifying evolutionally conserved gene co-expression modules around EGFR (EM) or PDGFRA (PM), we recently proposed an EM/PM classification scheme, which assigns IDH-wildtype glioblastomas (GBM) into the EM subtype committed in neural stem cell compartment, IDH-mutant astrocytomas and oligodendrogliomas into the PM subtype committed in early oligodendrocyte lineage. Here, we report the identification of EM/PM subtype-specific gene co-expression networks and the characterization of hub gene polypyrimidine tract-binding protein 1 (PTBP1) as a genomic alteration-independent vulnerability in IDH-wildtype GBM. Supervised by the EM/PM classification scheme, we applied weighted gene co-expression network analysis to identify subtype-specific global gene co-expression modules. These gene co-expression modules were characterized for their clinical relevance, cellular origin and conserved expression pattern during brain development. Using lentiviral vector-mediated constitutive or inducible knockdown, we characterized the effects of PTBP1 on the survival of IDH-wildtype GBM cells, which was complemented with the analysis of PTBP1-depedent splicing pattern and overexpression of splicing target neuron-specific CDC42 (CDC42-N) isoform.  Transcriptomes of adult gliomas can be robustly assigned into 4 large gene co-expression modules that are prognostically relevant and are derived from either malignant cells of the EM/PM subtypes or tumor microenvironment. The EM subtype is associated with a malignant cell-intrinsic gene module involved in pre-mRNA splicing, DNA replication and damage response, and chromosome segregation, and a microenvironment-derived gene module predominantly involved in extracellular matrix organization and infiltrating immune cells. The PM subtype is associated with two malignant cell-intrinsic gene modules predominantly involved in transcriptional regulation and mRNA translation, respectively. Expression levels of these gene modules are independent prognostic factors and malignant cell-intrinsic gene modules are conserved during brain development. Focusing on the EM subtype, we identified PTBP1 as the most significant hub for the malignant cell-intrinsic gene module. PTBP1 is not altered in most glioma genomes. PTBP1 represses the conserved splicing of CDC42-N. PTBP1 knockdown or CDC42-N overexpression disrupts actin cytoskeleton dynamics, causing accumulation of reactive oxygen species and cell apoptosis. PTBP1-mediated repression of CDC42-N splicing represents a potential genomic alteration-independent, developmentally conserved vulnerability in IDH-wildtype GBM.
    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
    聚嘧啶束结合蛋白1(PTBP1)在剪接和转录后调控中起重要作用。此外,PTBP1已被认为是肿瘤发生的原因。然而,PTBP1参与细胞衰老,衰老和癌症抑制的关键生物学过程,还有待澄清。这里,结果表明,PTBP1与肺腺癌(LUAD)的肿瘤生长和预后有关。PTBP1在包括LUAD在内的各种癌症类型中表现出显著增加的表达,并且在多种细胞衰老模型中表现出一致降低的表达。抑制PTBP1诱导LUAD细胞中的细胞衰老。从分子机制来看,PTBP1的沉默增强了F-box蛋白5(FBXO5)外显子3的跳跃,导致产生不太稳定的RNA剪接变体,FBXO5-S,这随后降低了FBXO5的整体表达。此外,发现FBXO5的下调可诱导LUAD衰老。总的来说,这些发现说明PTBP1通过抑制衰老在LUAD中具有致癌功能,靶向PTBP1介导的异常剪接在癌症治疗中具有治疗潜力。
    Polypyrimidine tract-binding protein 1 (PTBP1) plays an essential role in splicing and post-transcriptional regulation. Moreover, PTBP1 has been implicated as a causal factor in tumorigenesis. However, the involvement of PTBP1 in cellular senescence, a key biological process in aging and cancer suppression, remains to be clarified. Here, it is shown that PTBP1 is associated with the facilitation of tumor growth and the prognosis in lung adenocarcinoma (LUAD). PTBP1 exhibited significantly increased expression in various cancer types including LUAD and showed consistently decreased expression in multiple cellular senescence models. Suppression of PTBP1 induced cellular senescence in LUAD cells. In terms of molecular mechanisms, the silencing of PTBP1 enhanced the skipping of exon 3 in F-box protein 5 (FBXO5), resulting in the generation of a less stable RNA splice variant, FBXO5-S, which subsequently reduces the overall FBXO5 expression. Additionally, downregulation of FBXO5 was found to induce senescence in LUAD. Collectively, these findings illustrate that PTBP1 possesses an oncogenic function in LUAD through inhibiting senescence, and that targeting aberrant splicing mediated by PTBP1 has therapeutic potential in 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
    目的:代谢功能障碍相关脂肪变性肝病(MASLD)与2型糖尿病的关系,胰岛素抵抗和代谢综合征是公认的。虽然锌指BED型含3(ZBED3)与2型糖尿病和代谢综合征有关,其在MASLD中的作用尚不清楚。在这项研究中,我们旨在研究ZBED3在MASLD背景下的功能。
    方法:在患有MASLD的个体中评估ZBED3的表达水平,以及MASLD的细胞和动物模型。使用NEFA诱导的MASLD细胞模型和高脂饮食(HFD)诱导的MASLD动物模型进行体外和体内分析。分别,探讨ZBED3在MASLD中的作用。ZBED3表达通过慢病毒感染或腺相关病毒的尾静脉注射而增加。采用RNA-seq和生物信息学分析来检查ZBED3调节脂质积累的途径。这些下一代转录组测序研究的发现表明,ZBED3控制SREBP1c(也称为SREBF1;参与脂肪酸从头合成的基因);因此,免疫共沉淀和LC-MS/MS用于研究ZBED3调节固醇调节元件结合蛋白1c(SREBP1c)的分子机制。
    结果:在这项研究中,我们发现ZBED3在MASLD患者和MASLD动物模型的肝脏中显著上调.ZBED3过表达促进NEFA诱导的甘油三酯在体外肝细胞中的积累。此外,肝细胞特异性Zbed3过表达促进肝脏脂肪变性.相反,肝细胞特异性敲除Zbed3导致HFD诱导的肝脂肪变性耐药.机械上,ZBED3直接与聚嘧啶束结合蛋白1(PTBP1)相互作用,并影响其与SREBP1cmRNA前体的结合,以调节SREBP1cmRNA的稳定性和可变剪接。
    结论:本研究表明ZBED3促进肝脂肪变性,并作为MASLD进展的关键调节因子。
    方法:RNA-seq数据已保存在NCBI基因表达Omnibus中(www。ncbi.nlm.nih.gov/geo/query/acc。cgi?acc=GSE231875)。MS蛋白质组学数据已通过iProX合作伙伴存储库(https://proteomecentral。proteomexchange.org/cgi/GetDataset?ID=PXD041743)。
    OBJECTIVE: The relationship between metabolic dysfunction-associated steatotic liver disease (MASLD) and type 2 diabetes mellitus, insulin resistance and the metabolic syndrome is well established. While zinc finger BED-type containing 3 (ZBED3) has been linked to type 2 diabetes mellitus and the metabolic syndrome, its role in MASLD remains unclear. In this study, we aimed to investigate the function of ZBED3 in the context of MASLD.
    METHODS: Expression levels of ZBED3 were assessed in individuals with MASLD, as well as in cellular and animal models of MASLD. In vitro and in vivo analyses were conducted using a cellular model of MASLD induced by NEFA and an animal model of MASLD induced by a high-fat diet (HFD), respectively, to investigate the role of ZBED3 in MASLD. ZBED3 expression was increased by lentiviral infection or tail-vein injection of adeno-associated virus. RNA-seq and bioinformatics analysis were employed to examine the pathways through which ZBED3 modulates lipid accumulation. Findings from these next-generation transcriptome sequencing studies indicated that ZBED3 controls SREBP1c (also known as SREBF1; a gene involved in fatty acid de novo synthesis); thus, co-immunoprecipitation and LC-MS/MS were utilised to investigate the molecular mechanisms by which ZBED3 regulates the sterol regulatory element binding protein 1c (SREBP1c).
    RESULTS: In this study, we found that ZBED3 was significantly upregulated in the liver of individuals with MASLD and in MASLD animal models. ZBED3 overexpression promoted NEFA-induced triglyceride accumulation in hepatocytes in vitro. Furthermore, the hepatocyte-specific overexpression of Zbed3 promoted hepatic steatosis. Conversely, the hepatocyte-specific knockout of Zbed3 resulted in resistance of HFD-induced hepatic steatosis. Mechanistically, ZBED3 interacts directly with polypyrimidine tract-binding protein 1 (PTBP1) and affects its binding to the SREBP1c mRNA precursor to regulate SREBP1c mRNA stability and alternative splicing.
    CONCLUSIONS: This study indicates that ZBED3 promotes hepatic steatosis and serves as a critical regulator of the progression of MASLD.
    METHODS: RNA-seq data have been deposited in the NCBI Gene Expression Omnibus ( www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE231875 ). MS proteomics data have been deposited to the ProteomeXchange Consortium via the iProX partner repository ( https://proteomecentral.proteomexchange.org/cgi/GetDataset?ID=PXD041743 ).
    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:基于顺铂(DDP)的联合化疗是治疗膀胱癌(BLca)的重要方法。顺铂化疗过程中容易发生化疗耐药,是BLca患者预后不良的重要原因之一。环状RNA(circularRNAs,circRNAs)在BLca的发育和发展中的作用被广泛认可。然而,circRNAs在BLca的DDP抗性中的确切作用尚不清楚。方法:研究circATIC的性质,Sanger测序,利用琼脂糖凝胶电泳和RNA酶R/放线菌素D处理。RT-qPCR测定用于评估circRNA的表达水平,BLca组织和细胞中的miRNA和mRNA。进行功能实验以评估circATIC在BLca进展和体外化学敏感性中的功能。各种技术,如FISH,双荧光素酶报告基因测定,陷阱,RNA消化试验,RIP和ChIRP测定用于研究PTBP1,circATIC,miR-1247-5p和RCC2。原位膀胱癌模型,进行了异种皮下移植瘤模型和异种肺转移瘤模型,以表明circATIC在BLca进展和体内化学敏感性中的功能和机制。结果:在我们的研究中,我们观察到在BLca组织和细胞以及DDP抗性细胞中circATIC表达显着增强。circATIC表达较高的患者肿瘤直径较大,术后转移发生率较高,总生存率较低。进一步的实验表明circATIC加速BLca细胞的生长和转移并诱导DDP抗性。机械上,选择性剪接酶PTBP1介导circATIC的合成。circATIC可以通过形成miR-1247-5p或构建circATIC/LIN28A/RCC2RNA-蛋白三元复合物来增强RCC2mRNA的稳定性。最后,circATIC促进RCC2表达以增强上皮-间质转化(EMT)进程并激活JNK信号通路,从而增强BLca细胞的DDP抗性。结论:我们的研究表明circATIC促进BLca进展和DDP抵抗,并可作为BLca治疗的潜在靶标。
    Background: Cisplatin (DDP) based combination chemotherapy is a vital method for the treatment of bladder cancer (BLca). Chemoresistance easily occurs in the course of cisplatin chemotherapy, which is one of the important reasons for the unfavorable prognosis of BLca patients. Circular RNAs (circRNAs) are widely recognized for their role in the development and advancement of BLca. Nevertheless, the precise role of circRNAs in DDP resistance for BLca remains unclear. Methods: To study the properties of circATIC, sanger sequencing, agarose gel electrophoresis and treatment with RNase R/Actinomycin D were utilized. RT-qPCR assay was utilized to assess the expression levels of circRNA, miRNA and mRNA in BLca tissues and cells. Functional experiments were conducted to assess the function of circATIC in BLca progression and chemosensitivity in vitro. Various techniques such as FISH, Dual-luciferase reporter assay, TRAP, RNA digestion assay, RIP and ChIRP assay were used to investigate the relationships between PTBP1, circATIC, miR-1247-5p and RCC2. Orthotopic bladder cancer model, xenograft subcutaneous tumor model and xenograft lung metastasis tumor model were performed to indicate the function and mechanism of circATIC in BLca progression and chemosensitivity in vivo. Results: In our study, we observed that circATIC expression was significantly enhanced in BLca tissues and cells and DDP resistant cells. Patients with higher circATIC expression have larger tumor diameter, higher incidence of postoperative metastasis and lower overall survival rate. Further experiments showed that circATIC accelerated BLca cell growth and metastasis and induced DDP resistance. Mechanistically, alternative splicing enzyme PTBP1 mediated the synthesis of circATIC. circATIC could enhance RCC2 mRNA stability via sponging miR-1247-5p or constructing a circATIC/LIN28A/RCC2 RNA-protein ternary complex. Finally, circATIC promotes RCC2 expression to enhance Epithelial-Mesenchymal Transition (EMT) progression and activate JNK signal pathway, thus strengthening DDP resistance in BLca cells. Conclusion: Our study demonstrated that circATIC promoted BLca progression and DDP resistance, and could serve as a potential target for BLca 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
    LncRNA在癌症进展和靶向中起着至关重要的作用,但很难确定参与结直肠癌(CRC)进展的关键lncRNAs.我们使用21对IV期CRC组织和邻近正常组织将FAM83H-AS1鉴定为肿瘤促进相关lncRNA。体外和体内实验表明,在CRC细胞中敲低FAM83H-AS1抑制肿瘤的增殖和转移,反之亦然。m6A修饰对于通过作者METTL3和读者IGF2BP2/IGFBP3的FAM83H-AS1RNA稳定性至关重要。PTBP1-一种RNA结合蛋白-负责CRC中的FAM83H-AS1功能。FAM83H-AS1的外显子4上的T4(1770-2440nt)和T5(2440-2743nt)提供了PTBP1RRM2相互作用的平台。我们的结果表明,m6A修饰通过磷酸化PTBP1对其RNA剪接作用失调FAM83H-AS1致癌作用。在患者来源的异种移植模型中,ASO-FAM83H-AS1显著抑制胃肠道(GI)肿瘤的生长,不仅是CRC,还有GC和ESCC。ASO-FAM83H-AS1和奥沙利铂/顺铂的组合与单独使用任一种药剂的治疗相比显著抑制肿瘤生长。值得注意的是,所有这三种胃肠道癌均有病理完全缓解。我们的研究结果表明,FAM83H-AS1靶向治疗将使主要接受铂类药物治疗的胃肠道肿瘤患者受益。
    LncRNA plays a crucial role in cancer progression and targeting, but it has been difficult to identify the critical lncRNAs involved in colorectal cancer (CRC) progression. We identified FAM83H-AS1 as a tumor-promoting associated lncRNA using 21 pairs of stage IV CRC tissues and adjacent normal tissues. In vitro and in vivo experiments revealed that knockdown of FAM83H-AS1 in CRC cells inhibited tumor proliferation and metastasis, and vice versa. M6A modification is critical for FAM83H-AS1 RNA stability through the writer METTL3 and the readers IGF2BP2/IGFBP3. PTBP1-an RNA binding protein-is responsible for the FAM83H-AS1 function in CRC. T4 (1770-2440 nt) and T5 (2440-2743 nt) on exon 4 of FAM83H-AS1 provide a platform for PTBP1 RRM2 interactions. Our results demonstrated that m6A modification dysregulated the FAM83H-AS1 oncogenic role by phosphorylated PTBP1 on its RNA splicing effect. In patient-derived xenograft models, ASO-FAM83H-AS1 significantly suppressed the growth of gastrointestinal (GI) tumors, not only CRC but also GC and ESCC. The combination of ASO-FAM83H-AS1 and oxaliplatin/cisplatin significantly suppressed tumor growth compared with treatment with either agent alone. Notably, there was pathological complete response in all these three GI cancers. Our findings suggest that FAM83H-AS1 targeted therapy would benefit patients primarily receiving platinum-based therapy in GI cancers.
    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
    冠状动脉钙化(CAC)是心血管疾病发病的标志性事件,涉及血管平滑肌细胞(VSMC)向成骨状态的表型转化。尽管有这样的理解,控制VSMC成骨开关的分子机制仍未完全阐明。这里,我们试图研究环状RNA(circularRNA,circRNA)在CAC中的潜在作用.通过circRNA-seq的转录组分析,我们确定circTOP1为CAC患者的潜在候选circRNA。此外,在CAC模型中,我们观察到circTOP1的过度表达加剧了血管钙化.随后的下拉测定揭示了circTOP1和PTBP1之间的相互作用,PTBP1是CAC背景下circTOP1的推定靶基因。在体内和体外实验中,我们观察到CAC模型中circTOP1和PTBP1的表达增强,并指出减少circTOP1表达可有效减少模型小鼠的钙盐沉积和矿化结节。此外,体外实验表明,PTBP1的过表达逆转了沉默circTOP1引起的信号传导减弱,从而加剧了VSMC的成骨转化和钙化。总的来说,我们的研究结果表明,circTOP1通过调节PTBP1的表达来介导VSMC转分化,从而促进CAC。
    Coronary artery calcification (CAC) is a hallmark event in the pathogenesis of cardiovascular disease, involving the phenotypic transformation of vascular smooth muscle cells (VSMC) towards an osteogenic state. Despite this understanding, the molecular mechanisms governing the VSMC osteogenic switch remain incompletely elucidated. Here, we sought to examine the potential role of circular RNA (circRNA) in the context of CAC. Through transcriptome analysis of circRNA-seq, we identified circTOP1 as a potential candidate circRNA in individuals with CAC. Furthermore, we observed that overexpression of circTOP1 exacerbated vascular calcification in a CAC model. Subsequent pull-down assays revealed an interaction between circTOP1 and PTBP1, a putative target gene of circTOP1 in the context of CAC. In both in vivo and in vitro experiments, we observed heightened expression of circTOP1 and PTBP1 in the CAC model, and noted that reducing circTOP1 expression effectively reduced calcium salt deposits and mineralized nodules in model mice. Additionally, in vitro experiments demonstrated that overexpression of PTBP1 reversed the weakening of signaling caused by silencing circTOP1, thereby exacerbating the osteogenic transition and calcification of VSMC. Collectively, our findings suggested that circTOP1 promotes CAC by modulating PTBP1 expression to mediate VSMC transdifferentiation.
    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
    SUMO特异性蛋白酶1(SENP1)与急性髓性白血病(AML)之间的遗传关联已得到验证。然而,SENP1影响AML增殖的机制,凋亡,自噬仍然未知。检测AML患者SENP1和聚嘧啶束结合蛋白1(PTBP1)水平,AML细胞系,和异种移植组织。SENP1对AML增殖的影响,凋亡,和BECN1依赖性自噬通过体外和体内功能缺失或获得实验进行评估。使用免疫沉淀(IP)的SUMO化分析,RNA下拉,RIP,和RNA稳定性分析用于探索SENP1在AML发生发展中的分子机制。AML样品中SENP1水平升高。沉默SENP1阻碍了AML的发展,如AML细胞中SENP1耗竭导致的增殖抑制和G1期停滞和凋亡的促进所证明的。此外,沉默SENP1可抑制AML细胞中BECN1-脱位自噬。此外,BECN1或PTBP1的过表达部分中和了SENP1敲低对AML细胞行为的影响。机械上,SENP1介导PTBP1去SUMO化,然后直接与BECN1mRNA相互作用并增强其稳定性。体内实验进一步证实了SENP1抑制对AML发展的抑制作用。总的来说,SENP1/PTBP1/BECN1信号轴已被确定为增强AML治疗的重要治疗靶标.
    Genetic association between SUMO-specific protease 1 (SENP1) and acute myeloid leukemia (AML) has been validated. However, the mechanism by which SENP1 affects AML proliferation, apoptosis, and autophagy remains unknown. The levels of SENP1 and polypyrimidine tract-binding protein 1 (PTBP1) were measured in AML patients, AML cell lines, and xenograft tissues. The effects of SENP1 on AML proliferation, apoptosis, and BECN1-dependent autophagy were assessed through in vitro and in vivo loss- or gain-of-function experiments. SUMOylation analysis using immunoprecipitation (IP), RNA pull-down, RIP, and RNA stability assays were used to explore the molecular mechanism of SENP1 in AML development. The SENP1 level was elevated in AML samples. Silencing SENP1 impeded the development of AML, as evidenced by the inhibition of proliferation and promotion of G1 phase arrest and apoptosis resulting from SENP1 depletion in AML cells. Moreover, silencing of SENP1 restrained BECN1-depentent autophagy in AML cells. In addition, the overexpression of BECN1 or PTBP1 partially neutralized the effect of SENP1 knockdown on AML cell behavior. Mechanistically, SENP1 mediated PTBP1 deSUMOylation, which then directly interacted with BECN1 mRNA and enhanced its stability. In vivo experiments further confirmed the repressive effects of SENP1 suppression on AML development. Collectively, the SENP1/PTBP1/BECN1 signaling axis has been identified as a significant therapeutic target for enhancing AML 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
    PTBP1是调节前体mRNA剪接的癌基因。然而,PTBP1表达与基因甲基化的关系,癌症预后,肿瘤微环境尚不清楚。PTBP1在各种癌症中的表达谱来自TCGA,以及GTEx和CGGA数据库。CGGAmRNA_325、CGGAmRNA_301和CGGAmRNA_693数据集用作验证群组。使用TIMER2.0工具对免疫细胞浸润评分进行近似。使用基因集富集分析(GSEA)对具有高和低PTBP1表达的组进行功能富集分析。甲基化数据主要来源于SMART和Mexpress数据库。采用连锁组学分析对PTBP1甲基化相关基因进行功能富集分析,以及进行蛋白质功能富集分析。使用Seurat版本4.10进行单细胞转录组分析和空间转录组分析。与正常组织相比,PTBP1在各种癌症中显著过表达和低甲基化。它与预后有关,免疫细胞浸润,免疫检查点表达,基因组变异,肿瘤新抗原负荷,以及一系列癌症中的肿瘤突变负担,在低级别神经胶质瘤中效果尤其显著。在神经胶质瘤的背景下,PTBP1表达与WHO等级和IDH1突变状态相关。PTBP1的表达和甲基化在多种癌症中起着重要作用。PTBP1可作为炎症的标志物,胶质瘤的进展和预后。
    PTBP1 is an oncogene that regulates the splicing of precursor mRNA. However, the relationship between PTBP1 expression and gene methylation, cancer prognosis, and tumor microenvironment remains unclear. The expression profiles of PTBP1 across various cancers were derived from the TCGA, as well as the GTEx and CGGA databases. The CGGA mRNA_325, CGGA mRNA_301, and CGGA mRNA_693 datasets were utilized as validation cohorts. Immune cell infiltration scores were approximated using the TIMER 2.0 tool. Functional enrichment analysis for groups with high and low PTBP1 expression was conducted using Gene Set Enrichment Analysis (GSEA). Methylation data were predominantly sourced from the SMART and Mexpress databases. Linked-omics analysis was employed to perform functional enrichment analysis of genes related to PTBP1 methylation, as well as to conduct protein functional enrichment analysis. Single-cell transcriptome analysis and spatial transcriptome analysis were carried out using Seurat version 4.10. Compared to normal tissues, PTBP1 is significantly overexpressed and hypomethylated in various cancers. It is implicated in prognosis, immune cell infiltration, immune checkpoint expression, genomic variation, tumor neoantigen load, and tumor mutational burden across a spectrum of cancers, with particularly notable effects in low-grade gliomas. In the context of gliomas, PTBP1 expression correlates with WHO grade and IDH1 mutation status. PTBP1 expression and methylation play an important role in a variety of cancers. PTBP1 can be used as a marker of inflammation, progression and prognosis in gliomas.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号