PTBP1

PTBP1
  • 文章类型: Journal Article
    尚未研究在中风后痴呆小鼠模型中通过病毒转导抑制聚嘧啶束结合蛋白1(Ptbp1)信使RNA的治疗潜力。在这项研究中,脑缺血后3天,我们通过尾静脉注射含有腺相关病毒(AAV)-pGFAP-mCherry和AAV-pGFAP-CasRx(对照载体)的病毒载体混合物或AAV-pGFAP-mCherry和AAV-pGFAP-CasRx-SgRNA-(Ptbp1)(1:5,1.0×1011个病毒基因组)的混合物。我们在脑缺血56天后在海马中观察到新的mCherry/NeuN双阳性神经元样细胞。部分mCherry/GFAP双阳性星形胶质细胞样胶质细胞可能已转化为新的mCherry/NeuN双阳性神经元样细胞,并发生形态学变化。整合到齿状回和识别记忆中的新神经元细胞显着改善。这些结果表明,通过抑制Ptbp1将海马星形胶质细胞样神经胶质细胞体内转化为功能性新神经元可能是中风后痴呆的治疗策略。
    The therapeutic potential of suppressing polypyrimidine tract-binding protein 1 (Ptbp1) messenger RNA by viral transduction in a post-stroke dementia mouse model has not yet been examined. In this study, 3 days after cerebral ischemia, we injected a viral vector cocktail containing adeno-associated virus (AAV)-pGFAP-mCherry and AAV-pGFAP-CasRx (control vector) or a cocktail of AAV-pGFAP-mCherry and AAV-pGFAP-CasRx-SgRNA-(Ptbp1) (1:5, 1.0 × 1011 viral genomes) into post-stroke mice via the tail vein. We observed new mCherry/NeuN double-positive neuron-like cells in the hippocampus 56 days after cerebral ischemia. A portion of mCherry/GFAP double-positive astrocyte-like glia could have been converted into new mCherry/NeuN double-positive neuron-like cells with morphological changes. The new neuronal cells integrated into the dentate gyrus and recognition memory was significantly ameliorated. These results demonstrated that the in vivo conversion of hippocampal astrocyte-like glia into functional new neurons by the suppression of Ptbp1 might be a therapeutic strategy for post-stroke dementia.
    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:宫颈癌是世界上最常见的妇科恶性肿瘤,严重威胁妇女的生命健康。聚嘧啶束结合蛋白1(PTBP1),作为一个重要的拼接因素,已被确定为几种癌症的原癌基因,但其在宫颈癌中的作用和机制仍知之甚少。因此,我们的目的是探索PTBP1对增殖的影响,迁移,宫颈癌细胞凋亡,及其潜在机制。
    方法:使用小干扰RNA(siRNA)测定宫颈癌细胞的生物学功能,激动剂,细胞计数套件-8(CCK-8),transwell,迁移测试,westernblot,实时PCR,免疫组织化学和免疫荧光,分别。
    结果:结果表明,与正常组相比,PTBP1在宫颈癌患者和宫颈癌细胞系中高表达。此外,PTBP1沉默显著抑制细胞增殖,以及在HeLa和SiHa细胞中的迁移。PTBP1沉默还通过上调Bax和线粒体凋亡蛋白细胞色素C诱导线粒体凋亡,和下调B细胞白血病/淋巴瘤2(Bcl2)蛋白。此外,PTBP1沉默通过下调序列体I(p62)和上调轻链3-Ⅱ/轻链3-Ⅰ(LC3-Ⅱ/LC3-Ⅰ)的比例诱导自噬。机械上,我们发现磷酸肌醇3激酶(PI3K)激动剂逆转了PTBP1沉默诱导的变化.
    结论:总体而言,PTBP1沉默主要通过PI3K/AKT通路和保护性自噬诱导宫颈癌细胞凋亡。本研究为PTBP1作为宫颈癌的治疗靶点或预后标志物提供了初步证据。
    BACKGROUND: Cervical cancer is the most common gynecological malignancy in the world and seriously threatens to women\'s lives and health. Polypyrimidine tract binding protein 1 (PTBP1), as an important splicing factor, has been identified as a proto-oncogene in several cancers, but its role and mechanism in cervical cancer remain poorly understood. Thus, our aim is to explore the impact of PTBP1 on proliferation, migration, apoptosis of cervical cancer cells, and its underlying mechanisms.
    METHODS: The biological functions in cervical cancer cells were determined using small interfering RNA (siRNA), agonist, Cell Counting Kit-8 (CCK-8), transwell, migration test, western blot, real-time-PCR, immunohistochemistry and immunofluorescence, respectively.
    RESULTS: The results indicated that PTBP1 was highly expressed in cervical cancer patients and cervical cancer cell lines compared to the normal group. Moreover, PTBP1 silencing significantly inhibited cell proliferation, and migration in both HeLa and SiHa cells. The PTBP1 silencing also induced mitochondrial apoptosis through upregulating Bax and mitochondrial apoptotic protein Cytochrome C, and downregulating B-Cell Leukemia/Lymphoma 2 (Bcl2) protein. Additionally, PTBP1 silencing induced autophagy by downregulating Sequestosome I (p62) and upregulating the ratio of Light chain 3-Ⅱ/Light chain 3-Ⅰ (LC3-Ⅱ/LC3-Ⅰ). Mechanistically, we found that the Phosphoinositide 3-kinase (PI3K) agonist reversed the changes induced by PTBP1 silencing.
    CONCLUSIONS: Overall, PTBP1 silencing can induce cervical cancer cells apoptosis mainly through PI3K/AKT pathway and protective autophagy. This study provides preliminary evidence for PTBP1 as a therapeutic target or prognostic marker for cervical 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
    嵌合RNA,不同于DNA基因融合,已成为癌症治疗中具有多种功能的有希望的治疗靶标。然而,大多数嵌合RNA的功能意义和治疗潜力尚不清楚.在这里,我们鉴定了溶质载体家族2-成员11(SLC2A11)和巨噬细胞迁移抑制因子(MIF)的新型融合转录本。在这项研究中,我们调查了癌症基因组图谱队列和孙逸仙纪念医院患者队列中SLC2A11-MIF的上调.随后,功能研究表明,SLC2A11-MIF增强了增殖,抗凋亡作用,膀胱癌细胞的体外和体内转移。机械上,SLC2A11-MIF编码的融合蛋白与聚嘧啶束结合蛋白1(PTBP1)相互作用,并调节BCa细胞中Polo样激酶1,回旋引导受体1和磷酸肌醇-3-激酶调节亚基3的mRNA半衰期。此外,PTBP1敲低消除了SLC2A11-MIF对生物学功能和mRNA稳定性的增强影响。此外,SLC2A11-MIFmRNA的表达受CCCTC结合因子调节,并通过N-乙酰转移酶10促进的RNAN4-乙酰胞苷修饰而稳定.总的来说,我们的发现揭示了一种由SLC2A11-MIF-PTBP1轴协调的显著融合蛋白,该融合蛋白在膀胱癌多步进展过程中控制mRNA稳定性.
    Chimeric RNAs, distinct from DNA gene fusions, have emerged as promising therapeutic targets with diverse functions in cancer treatment. However, the functional significance and therapeutic potential of most chimeric RNAs remain unclear. Here we identify a novel fusion transcript of solute carrier family 2-member 11 (SLC2A11) and macrophage migration inhibitory factor (MIF). In this study, we investigated the upregulation of SLC2A11-MIF in The Cancer Genome Atlas cohort and a cohort of patients from Sun Yat-Sen Memorial Hospital. Subsequently, functional investigations demonstrated that SLC2A11-MIF enhanced the proliferation, antiapoptotic effects, and metastasis of bladder cancer cells in vitro and in vivo. Mechanistically, the fusion protein encoded by SLC2A11-MIF interacted with polypyrimidine tract binding protein 1 (PTBP1) and regulated the mRNA half-lives of Polo Like Kinase 1, Roundabout guidance receptor 1, and phosphoinositide-3-kinase regulatory subunit 3 in BCa cells. Moreover, PTBP1 knockdown abolished the enhanced impact of SLC2A11-MIF on biological function and mRNA stability. Furthermore, the expression of SLC2A11-MIF mRNA is regulated by CCCTC-binding factor and stabilized through RNA N4-acetylcytidine modification facilitated by N-acetyltransferase 10. Overall, our findings revealed a significant fusion protein orchestrated by the SLC2A11-MIF-PTBP1 axis that governs mRNA stability during the multistep progression of bladder 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
    背景:胃癌(GC)是一种常见的恶性肿瘤,RNA结合蛋白聚嘧啶束结合蛋白1(PTBP1)已被确定为各种肿瘤类型的关键因素。此外,异常的自噬水平已被证明显著影响肿瘤发生和进展。尽管如此,PTBP1在GC自噬调控中的精确调控机制尚不清楚。
    方法:为了评估PTBP1在GC中的表达,我们采用了一种利用蛋白质印迹的综合方法,实时定量聚合酶链反应(RT-qPCR),和生物信息学分析。为了进一步鉴定GC细胞中与PTBP1结合的下游靶基因,我们利用RNA免疫沉淀联合测序(si-PTBP1RNA-seq).评估PTBP1对胃癌发生的影响,我们进行了CCK-8检测,集落形成试验,和GC异种移植小鼠模型测定。此外,我们用透射电子显微镜,免疫荧光,流式细胞术,westernblot,RT-qPCR,和GC异种移植小鼠模型实验,以阐明PTBP1调节GC自噬的具体机制。
    结果:我们的发现表明,与邻近的正常组织相比,PTBP1在GC组织中明显过表达。沉默PTBP1导致自噬体的异常积累,从而在体外和体内抑制GC细胞活力。机械上,干扰PTBP1促进硫氧还蛋白相互作用蛋白(TXNIP)mRNA的稳定性,导致TXNIP介导的氧化应激增加。因此,溶酶体功能受损,最终导致自噬通量的阻塞。此外,我们的结果表明,干扰PTBP1增强了氯喹的抗肿瘤作用,在体外和体内。
    结论:PTBP1敲低通过直接结合TXNIPmRNA并促进其表达而损害GC进展。基于这些结果,PTBP1成为GC的有希望的治疗靶标。
    BACKGROUND: Gastric cancer (GC) is a prevalent malignant tumor, and the RNA-binding protein polypyrimidine tract-binding protein 1 (PTBP1) has been identified as a crucial factor in various tumor types. Moreover, abnormal autophagy levels have been shown to significantly impact tumorigenesis and progression. Despite this, the precise regulatory mechanism of PTBP1 in autophagy regulation in GC remains poorly understood.
    METHODS: To assess the expression of PTBP1 in GC, we employed a comprehensive approach utilizing western blot, real-time quantitative polymerase chain reaction (RT-qPCR), and bioinformatics analysis. To further identify the downstream target genes that bind to PTBP1 in GC cells, we utilized RNA immunoprecipitation coupled with sequencing (si-PTBP1 RNA-seq). To evaluate the impact of PTBP1 on gastric carcinogenesis, we conducted CCK-8 assays, colony formation assays, and GC xenograft mouse model assays. Additionally, we utilized a transmission electron microscope, immunofluorescence, flow cytometry, western blot, RT-qPCR, and GC xenograft mouse model experiments to elucidate the specific mechanism underlying PTBP1\'s regulation of autophagy in GC.
    RESULTS: Our findings indicated that PTBP1 was significantly overexpressed in GC tissues compared with adjacent normal tissues. Silencing PTBP1 resulted in abnormal accumulation of autophagosomes, thereby inhibiting GC cell viability both in vitro and in vivo. Mechanistically, interference with PTBP1 promoted the stability of thioredoxin-interacting protein (TXNIP) mRNA, leading to increased TXNIP-mediated oxidative stress. Consequently, this impaired lysosomal function, ultimately resulting in blockage of autophagic flux. Furthermore, our results suggested that interference with PTBP1 enhanced the antitumor effects of chloroquine, both in vitro and in vivo.
    CONCLUSIONS: PTBP1 knockdown impairs GC progression by directly binding to TXNIP mRNA and promoting its expression. Based on these results, PTBP1 emerges as a promising therapeutic target for 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
    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
    本研究旨在验证急性髓系白血病(AML)患者疾病进展的新潜在指标。从27名AML患者和27名没有血液恶性肿瘤的对照中收集骨髓样品。测量骨髓样本中的嘧啶束结合蛋白1(PTBP1)表达,以及PTBP1与法国-美国-英国(FAB)分类的关联,细胞遗传学,风险分层,分析完全缓解(CR)率。最终评估AML患者PTBP1与Ki-67/p53表达之间的相关性。结果显示,AML患者的PTBP1mRNA和蛋白水平高于对照组。PTBP1表达能够区分AML患者和对照组(曲线下面积,0.8601;95%CI,0.7632-0.9570)。此外,PTBP1表达与FMS样酪氨酸激酶3(FLT3-ITD)和复杂核型(CK)内内部串联重复突变频率增加有关,而PTBP1的表达与FAB分类无关,单体核型,分离的双等位基因CCAAT/增强子结合蛋白α(CEBPA)突变,AML患者的核蛋白1(NPM1)突变。此外,根据AML患者的危险分层和较低的CR率,PTBP1表达与预后较差相关。此外,AML患者PTBP1表达与增殖标志物Ki-67表达呈正相关,与凋亡标志物p53表达呈负相关。总的来说,PTBP1是一种可行的生物标志物,有助于AML的风险预测和潜在药物靶标的确定。
    This study aimed to verify a novel potential indicator of disease progression in acute myeloid leukemia (AML) patients. Bone marrow samples were collected from 27 AML patients and 27 controls without hematological malignancies. Polypyrimidine tract-binding protein 1 (PTBP1) expression in bone marrow samples was measured, and the association of PTBP1 with the French-American-British (FAB) classification, cytogenetics, risk stratification, and complete remission (CR) rate was analyzed. The correlation between PTBP1 and Ki-67/p53 expression in AML patients was ultimately evaluated. The results showed that PTBP1 mRNA and protein levels were greater in AML patients than in controls. PTBP1 expression was able to distinguish between AML patients and controls (area under the curve, 0.8601; 95% confidence interval, 0.7632-0.9570). Furthermore, PTBP1 expression was associated with an increased frequency of internal tandem duplication mutations within FMS-like tyrosine kinase-3 (FLT3) and a complex karyotype, while PTBP1 expression was not correlated with FAB classification, monosomal karyotype, isolated biallelic CCAAT/enhancer-binding protein α (CEBPA) mutation, or nucleophosmin 1 (NPM1) mutation in patients with AML. Moreover, PTBP1 expression was associated with a poorer prognosis according to risk stratification and a lower CR rate in AML patients. In addition, PTBP1 expression was positively correlated with the expression of the proliferation marker Ki-67 and negatively correlated with the expression of the apoptosis marker p53 in AML patients. Overall, PTBP1 is a viable biomarker that contributes to the risk prediction and the determination of potential drug targets for AML.
    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

公众号