hnRNP A1

hnRNP A1
  • 文章类型: Journal Article
    目的:探讨β-arrestin2对去势抵抗性前列腺癌(CRPC)多西他赛耐药的影响及其分子机制。
    方法:PC3和DU145细胞具有稳定的β-arrestin2过表达,C4-2细胞具有稳定的β-arrestin2敲低,通过使用慢病毒和嘌呤霉素选择构建。采用MTT法和集落形成法研究β-arrestin2表达对CRPC细胞多西他赛耐药的影响。糖酵解分析用于评估由β-arrestin2调节的糖酵解能力。GO富集分析,利用GEO和TCGA的公开数据,进行了基因集富集分析和Spearman相关检验,以探讨潜在的生物学功能和机制。免疫印迹法检测PKM2、磷酸化PKM2、磷酸化ERK1/2和hnRNPA1的表达。进行了功能阻断实验,以证实PKM2和hnRNPA1在稳定的β-arrestin2过表达的细胞中通过沉默PKM2或hnRNPA1表达调节β-arrestin2的生物学功能中的作用。最后,建立裸鼠异种移植模型以证实细胞实验的实验结果。
    结果:β-Arrestin2显著降低CRPC细胞对多西他赛刺激的敏感性,通过增强PKM2的磷酸化和表达。此外,β-arrestin2通过ERK1/2信号通路增加PKM2磷酸化,并通过hnRNPA1依赖性PKM可变剪接机制以转录后方式诱导PKM2表达,而不是通过抑制其泛素化降解。
    结论:我们的研究结果表明,β-arrestin2/hnRNPA1/PKM2通路可能是治疗多西他赛耐药CRPC的一个有希望的靶点。
    OBJECTIVE: To investigate the influence of β-arrestin2 on the docetaxel resistance in castration-resistant prostate cancer (CRPC) and elucidate the underlying molecular mechanisms.
    METHODS: PC3 and DU145 cells with stable β-arrestin2 overexpression and C4-2 cells with stable β-arrestin2 knockdown, were constructed via using lentivirus and puromycin selection. MTT and colony formation assays were carried out to investigate the effect of β-arrestin2 expression on the docetaxel resistance of CRPC cells. Glycolysis analysis was used to assess the glycolytic capacity modulated by β-arrestin2. GO enrichment analysis, gene set enrichment analysis and Spearman correlation test were carried out to explore the potential biological function and mechanism via using public data from GEO and TCGA. The expressions of PKM2, Phospho-PKM2, Phospho-ERK1/2 and hnRNP A1 were detected by western blot. Functional blocking experiments were carried out to confirm the roles of PKM2 and hnRNP A1 in the regulation of β-arrestin2\'s biological functions via silencing PKM2 or hnRNP A1 expression in cells with stable β-arrestin2 overexpression. Finally, nude mice xenograft models were established to confirm the experimental results of cell experiments.
    RESULTS: β-Arrestin2 significantly decreased the sensitivity of CRPC cells to docetaxel stimulation, through enhancing the phosphorylation and expression of PKM2. Additionally, β-arrestin2 increased PKM2 phosphorylation via the ERK1/2 signaling pathway and induced PKM2 expression in a post-transcriptional manner through an hnRNP A1-dependent PKM alternative splicing mechanism, rather than by inhibiting its ubiquitination degradation.
    CONCLUSIONS: Our findings indicate that the β-arrestin2/hnRNP A1/PKM2 pathway could be a promising target for treating docetaxel-resistant CRPC.
    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
    探讨RNA结合蛋白hnRNPA1在小鼠海马神经元(HT22)糖酵解中的作用机制。
    通过HT22体外进行RIP和CLIP-qPCR,以观察hnRNPA1调节糖酵解关键蛋白表达的机制。通过VPC-80051抑制HT22中hnRNPA1蛋白的RNA结合域,观察hnRNPA1对HT22糖酵解的影响。用慢病毒过表达hnRNPA1观察过表达hnRNPA1对Aβ25-35损伤的HT22糖酵解的影响。采用Westernblot法研究了不同年龄野生型小鼠和三转基因(APP/PS1/Tau)AD小鼠脑组织中hnRNPA1的表达。
    RIP实验结果表明hnRNPA1和HK1mRNA显著结合。CLIP-qPCR结果显示hnRNPA1直接结合HK1mRNA的2605-2821区域。hnRNPA1抑制剂可以下调HT22细胞HK1mRNA和HK1蛋白的表达。hnRNPA1过表达可显著降低Aβ25-35通过hnRNPA1/HK1/丙酮酸通路对神经元的毒性作用。此外,抑制hnRNPA1与淀粉样前体蛋白(APP)RNA的结合被发现增加Aβ表达,而Aβ25-35也通过增强HT22中p38MAPK的磷酸化下调hnRNPA1的表达。它们相互作用形成双向调节,进一步下调hnRNPA1的表达,最终加重糖酵解功能障碍。蛋白免疫印迹显示小鼠脑组织hnRNPA1随年龄增长而降低,在AD小鼠中下降更大,提示hnRNPA1的降低可能是AD发病的一个易感因素。
    UNASSIGNED: To investigate the mechanism of RNA-binding protein hnRNP A1 in mouse hippocampal neurons (HT22) on glycolysis.
    UNASSIGNED: RIP and CLIP-qPCR were performed by HT22 in vitro to observe the mechanism of hnRNP A1 regulating the expression of key proteins in glycolysis. The RNA binding domain of hnRNP A1 protein in HT22 was inhibited by VPC-80051, and the effect of hnRNP A1 on glycolysis of HT22 was observed. Lentivirus overexpression of hnRNP A1 was used to observe the effect of overexpression of hnRNP A1 on glycolysis of Aβ25-35-injured HT22. The expression of hnRNP A1 in brain tissues of wild-type mice and triple-transgenic (APP/PS1/Tau) AD mice at different ages was studied by Western blot assay.
    UNASSIGNED: The results of RIP experiment showed that hnRNP A1 and HK1 mRNA were significantly bound. The results of CLIP-qPCR showed that hnRNP A1 directly bound to the 2605-2821 region of HK1 mRNA. hnRNP A1 inhibitor can down-regulate the expression of HK1 mRNA and HK1 protein in HT22 cells. Overexpression of hnRNP A1 can significantly reduce the toxic effect of Aβ25-35 on neurons via the hnRNP A1/HK1/ pyruvate pathway. In addition, inhibition of hnRNP A1 binding to amyloid precursor protein (APP) RNA was found to increase Aβ expression, while Aβ25-35 also down-regulated hnRNP A1 expression by enhancing phosphorylation of p38 MAPK in HT22. They interact to form bidirectional regulation, further down-regulating the expression of hnRNP A1, and ultimately aggravating glycolytic dysfunction. Protein immunoblotting showed that hnRNP A1 decreased with age in mouse brain tissue, and the decrease was greater in AD mice, suggesting that the decrease of hnRNP A1 may be a predisposed factor in the pathogenesis of AD.
    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
    乳腺癌是最常见的恶性肿瘤之一,由于转移而导致高死亡率。SCRIB,一种主要分布在细胞膜上的支架蛋白,是一种潜在的肿瘤抑制因子.SCRIB的错误定位和异常表达刺激EMT通路并促进肿瘤细胞转移。SCRIB具有通过可变剪接产生的两个同种型(具有或不具有外显子16)。在这项研究中,我们研究了SCRIB亚型在乳腺癌转移中的功能及其调节机制。我们表明,与全长同工型(SCRIB-L)相比,截短的SCRIB亚型(SCRIB-S)在高转移性MDA-MB-231细胞中过度表达,通过激活ERK途径促进乳腺癌转移.SCRIB-S对催化磷酸酶亚基PPP1CA的亲和力低于SCRIB-L,这种差异可能导致两种同工型在癌症转移中的不同功能。通过进行CLIP,基于RIP和MS2-GFP的实验,我们发现,异质核核糖核蛋白A1(hnRNPA1)通过与SCRIB内含子15上的富含“AG”的序列“caggauggaggcccccccgugcgag”结合来促进SCRIB外显子16的跳跃。用基于该结合序列设计的SCRIB反义寡脱氧核苷酸(ASO-SCRIB)转染MDA-MB-231细胞,不仅有效抑制hnRNPA1与SCRIB前mRNA的结合,而且抑制SCRIB-S的产生,而且还逆转了hnRNPA1对ERK通路的激活,抑制了乳腺癌的转移。本研究为乳腺癌的治疗提供了新的潜在靶点和候选药物。
    Breast cancer is one of the most common malignant tumors with high mortality due to metastases. SCRIB, a scaffold protein mainly distributed in the cell membrane, is a potential tumor suppressor. Mislocalization and aberrant expression of SCRIB stimulate the EMT pathway and promote tumor cell metastasis. SCRIB has two isoforms (with or without exon 16) produced by alternative splicing. In this study we investigated the function of SCRIB isoforms in breast cancer metastasis and their regulatory mechanisms. We showed that in contrast to the full-length isoform (SCRIB-L), the truncated SCRIB isoform (SCRIB-S) was overexpressed in highly metastatic MDA-MB-231 cells that promoted breast cancer metastasis through activation of the ERK pathway. The affinity of SCRIB-S for the catalytic phosphatase subunit PPP1CA was lower than that of SCRIB-L and such difference might contribute to the different function of the two isoforms in cancer metastasis. By conducting CLIP, RIP and MS2-GFP-based experiments, we revealed that the heterogeneous nuclear ribonucleoprotein A1 (hnRNP A1) promoted SCRIB exon 16 skipping by binding to the \"AG\"-rich sequence \"caggauggaggccccccgugccgag\" on intron 15 of SCRIB. Transfection of MDA-MB-231 cells with a SCRIB antisense oligodeoxynucleotide (ASO-SCRIB) designed on the basis of this binding sequence, not only effectively inhibited the binding of hnRNP A1 to SCRIB pre-mRNA and suppressed the production of SCRIB-S, but also reversed the activation of the ERK pathway by hnRNP A1 and inhibited the metastasis of breast cancer. This study provides a new potential target and a candidate drug for treating 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
    脊髓性肌萎缩症(SMA)是一种神经退行性疾病,由运动神经元存活1基因(SMN1)的功能拷贝缺失引起。几乎相同的模拟,SMN2,不能补偿SMN1的损失,因为外显子7被从大多数SMN2转录物中异常跳过,一个由Sam68/KHDRBS1和hnRNPA1协同活性介导的过程。这导致产生一个截断的,快速降解的非功能性蛋白质。在这里,我们介绍了Sam68RNA结合域(RBD)的几种晶体结构。Sam68-RBD通过来自两个单体的螺旋α3的反平行缔合形成稳定的对称同型二聚体。然而,域组织和二聚化界面的细节与先前表征的同源物显著不同。我们证明Sam68和hnRNPA1可以同时结合SMN2(ex7)中心区域内的近端基序。Further,我们表明,两种蛋白质的RNA结合口袋在它们的异源二聚体复合物中彼此接近,并使用交联-质谱鉴定接触残基。我们提出了三元Sam68·SMN2(ex7)·hnRNPA1复合物的模型,该模型调和了有关SMN1/2剪接的所有可用信息。我们的发现对SMA的病因具有重要意义,并为设计治疗剪接疾病的新疗法开辟了新途径。本文受版权保护。保留所有权利。
    Spinal muscular atrophy (SMA) is a neurodegenerative disease caused by the absence of a functional copy of the Survival of Motor Neuron 1 gene (SMN1). The nearly identical paralog, SMN2, cannot compensate for the loss of SMN1 because exon 7 is aberrantly skipped from most SMN2 transcripts, a process mediated by synergistic activities of Src-associated during mitosis, 68 kDa (Sam68/KHDRBS1) and heterogeneous nuclear ribonucleoprotein (hnRNP) A1. This results in the production of a truncated, nonfunctional protein that is rapidly degraded. Here, we present several crystal structures of Sam68 RNA-binding domain (RBD). Sam68-RBD forms stable symmetric homodimers by antiparallel association of helices α3 from two monomers. However, the details of domain organization and the dimerization interface differ significantly from previously characterized homologs. We demonstrate that Sam68 and hnRNP A1 can simultaneously bind proximal motifs within the central region of SMN2 (ex7). Furthermore, we show that the RNA-binding pockets of the two proteins are close to each other in their heterodimeric complex and identify contact residues using crosslinking-mass spectrometry. We present a model of the ternary Sam68·SMN2 (ex7)·hnRNP A1 complex that reconciles all available information on SMN1/2 splicing. Our findings have important implications for the etiology of SMA and open new avenues for the design of novel therapeutics to treat splicing diseases.
    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
    广泛的SLC26A4突变导致Pendred综合征和扩大的前庭水管,两者都与感音神经性听力损失(SNHL)有关。剪接位点突变,c.919-2A>G(A-2G),这在亚洲人群中很常见,损害内含子7的3'剪接位点,导致外显子8在mRNA剪接前和随后的移码过程中发生跳跃,从而在随后的外显子中产生过早的终止密码子。目前,目前尚没有有效的药物治疗方法。对于A-2G触发的SNHL,纠正突变体错误剪接的分子有望治疗这种疾病。反义寡核苷酸(ASO)可以在靶向特异性剪接沉默时促进外显子包含。这里,我们在小基因系统中系统地筛选了大量的ASO,并确定了一些显着抑制外显子8跳跃的ASO。领先的ASO,它靶向内含子8中的异质核核糖核蛋白(hnRNP)A1/A2内含子剪接沉默(ISS),促进了外显子8在来自两个纯合患者的培养外周血单核细胞中的有效包含。在部分人源化Slc26a4A-2G小鼠模型中,两次皮下注射160mg/kg的ASO可明显挽救肝脏中的外显子8剪接。我们的结果表明,ISS靶向ASO具有治疗SLC26A4中A-2G突变引起的遗传性听力损失的治疗潜力。
    A wide spectrum of SLC26A4 mutations causes Pendred syndrome and enlarged vestibular aqueduct, both associated with sensorineural hearing loss (SNHL). A splice-site mutation, c.919-2A>G (A-2G), which is common in Asian populations, impairs the 3\' splice site of intron 7, resulting in exon 8 skipping during pre-mRNA splicing and a subsequent frameshift that creates a premature termination codon in the following exon. Currently, there is no effective drug treatment for SHNL. For A-2G-triggered SNHL, molecules that correct mis-splicing of the mutant hold promise to treat the disease. Antisense oligonucleotides (ASOs) can promote exon inclusion when targeting specific splicing silencers. Here, we systematically screened a large number of ASOs in a minigene system and identified a few that markedly repressed exon 8 skipping. A lead ASO, which targets a heterogeneous nuclear ribonucleoprotein (hnRNP) A1/A2 intronic splicing silencer (ISS) in intron 8, promoted efficient exon 8 inclusion in cultured peripheral blood mononuclear cells derived from two homozygous patients. In a partially humanized Slc26a4 A-2G mouse model, two subcutaneous injections of the ASO at 160 mg/kg significantly rescued exon 8 splicing in the liver. Our results demonstrate that the ISS-targeting ASO has therapeutic potential to treat genetic hearing loss caused by the A-2G mutation in SLC26A4.
    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
    异质核核糖核蛋白(HNRNP)A1是HNRNP蛋白家族中最丰富和普遍表达的成员。近年来,越来越明显的是,HNRNPA1有助于神经退行性疾病的发展。然而,关于HNRNPA1在癌症发展中的潜在作用知之甚少。这里,我们报道了HNRNPA1在肺癌组织中的表达显著升高,并且与肺癌患者的总生存期呈负相关。此外,HNRNPA1通过直接与VRK1mRNA的3'非翻译区(UTR)结合,正向调节牛痘相关激酶1(VRK1)的翻译,从而通过VRK1介导的cAMP反应元件结合蛋白(CREB)的磷酸化增加细胞周期蛋白D1(CCND1)的表达。此外,HNRNPA1与VRK1mRNA的3'UTR的顺式作用区结合有助于增加肺癌细胞增殖。因此,我们的研究揭示了HNRNPA1通过转录后调节VRK1表达在肺癌发生中的新作用,并提示其作为肺癌患者治疗靶点的潜力.
    THeterogeneous nuclear ribonucleoprotein (HNRNP) A1 is the most abundant and ubiquitously expressed member of the HNRNP protein family. In recent years, it has become more evident that HNRNP A1 contributes to the development of neurodegenerative diseases. However, little is known about the underlying role of HNRNP A1 in cancer development. Here, we report that HNRNP A1 expression is significantly increased in lung cancer tissues and is negatively correlated with the overall survival of patients with lung cancer. Additionally, HNRNP A1 positively regulates vaccinia-related kinase 1 (VRK1) translation via binding directly to the 3\' untranslated region (UTR) of VRK1 mRNA, thus increasing cyclin D1 (CCND1) expression by VRK1-mediated phosphorylation of the cAMP response element-binding protein (CREB). Furthermore, HNRNP A1 binding to the cis-acting region of the 3\'UTR of VRK1 mRNA contributes to increased lung cancer cell proliferation. Thus, our study unveils a novel role of HNRNP A1 in lung carcinogenesis via post-transcriptional regulation of VRK1 expression and suggests its potential as a therapeutic target for patients with lung cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Heterogeneous nuclear ribonucleoprotein A1 (hnRNP A1) is a member of the hnRNP family of conserved proteins that is involved in RNA transcription, pre-mRNA splicing, mRNA transport, protein translation, microRNA processing, telomere maintenance and the regulation of transcription factor activity. HnRNP A1 is ubiquitously, yet differentially, expressed in many cell types, and due to post-translational modifications, can vary in its molecular function. While a plethora of knowledge is known about the function and dysfunction of hnRNP A1 in diseases other than neurodegenerative disease (e.g., cancer), numerous studies in amyotrophic lateral sclerosis, frontotemporal lobar degeneration, multiple sclerosis, spinal muscular atrophy, Alzheimer\'s disease, and Huntington\'s disease have found that the dysregulation of hnRNP A1 may contribute to disease pathogenesis. How hnRNP A1 mechanistically contributes to these diseases, and whether mutations and/or altered post-translational modifications contribute to pathogenesis, however, is currently under investigation. The aim of this comprehensive review is to first describe the background of hnRNP A1, including its structure, biological functions in RNA metabolism and the post-translational modifications known to modify its function. With this knowledge, the review then describes the influence of hnRNP A1 in neurodegenerative disease, and how its dysfunction may contribute the pathogenesis.
    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
    Human papillomavirus 16 (HPV16) 5\'-splice site SD226 and 3\'-splice site SA409 are required for production of the HPV16 E7 mRNAs, whereas unspliced mRNAs produce E6 mRNAs. The E6 and E7 proteins are essential in the HPV16 replication cycle but are also the major HPV16 proteins required for induction and maintenance of malignancy caused by HPV16 infection. Thus, a balanced expression of unspliced and spliced mRNAs is required for production of sufficient quantities of E6 and E7 proteins under physiological and pathophysiological conditions. If splicing becomes too efficient, the levels of unspliced E6 mRNAs will decrease below a threshold level that is no longer able to produce E6 protein quantities high enough to significantly reduce p53 protein levels. Similarly, if splicing becomes too inefficient, the levels of spliced E7 mRNAs will decrease below a threshold level that is no longer able to produce E7 protein quantities high enough to significantly reduce pRb protein levels. To determine how splicing between SD226 and SA409 is regulated, we have investigated how SA409 is controlled by the cellular proteins hnRNP A1 and hnRNP A2, two proteins that have been shown previously to control HPV16 gene expression. We found that hnRNP A1 and A2 interacted directly and specifically with a C-less RNA element located between HPV16 nucleotide positions 594 and 604 downstream of SA409. Overexpression of hnRNP A1 inhibited SA409 and promoted production of unspliced E6 mRNAs at the expense of the E7 mRNAs, whereas overexpression of hnRNP A2 inhibited SA409 to redirect splicing to SA742, a downstream 3\'-splice site that is used for generation of HPV16 E6̂E7, E1, and E4 mRNAs. Thus, high levels of either hnRNP A1 or hnRNP A2 inhibited production of the promitotic HPV16 E7 protein. We show that the hnRNP A1 and A2 proteins control the relative levels of the HPV16 unspliced and spliced HPV16 E6 and E7 mRNAs and function as inhibitors of HPV16 E7 expression.IMPORTANCE Human papillomavirus type 16 (HPV16) belongs to the high-risk-group of HPVs and is causing a variety of anogenital cancers and head and neck cancer. The two HPV16 oncoproteins E6 and E7 prevent apoptosis and promote mitosis and are essential for completion of the HPV16 life cycle and for transformation of the infected cell and maintenance of malignancy. E6 and E7 are produced from two mRNAs that are generated in a mutually exclusive manner by alternative splicing. While E6 protein is made from the unspliced mRNA, E7 is made from the spliced version of the same pre-mRNA. Since sufficient quantities of both E6 and E7 are required for malignant transformation, this intricate arrangement of gene expression renders E6 and E7 expression vulnerable to external interference. Since antiviral drugs to HPV16 are not available, a detailed knowledge of the regulation of HPV16 E6 and E7 mRNA splicing may uncover novel targets for therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    A number of studies have clearly established the oncogenic role for MAPK-interacting protein kinases (MNK) in human malignancies. Modulation of MNK activity affects translation of mRNAs involved in cancer development, progression, and resistance to therapies. As a result, there are ongoing efforts to develop and evaluate MNK inhibitors for cancer treatment. However, it is important to recognize that MNK activity also plays an important role in regulating the innate and adaptive immune systems. A better understanding of the role of MNK kinases and MNK-mediated signals in regulating the immune system could help mitigate undesired side effects while maximizing therapeutic efficacy of MNK inhibitors. Here, we provide a systematic review on the function of MNK kinases and their substrates in immune cells.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    多发性硬化症(MS)中的神经变性被认为是疾病进展和永久性残疾的基础。已经提出了MS中神经变性的许多机制,比如线粒体功能障碍,氧化应激,神经炎症,和RNA结合蛋白功能障碍。这篇综述的目的是强调MS中神经变性的机制及其模型,专注于RNA结合蛋白功能障碍。研究RNA结合蛋白功能障碍解决了我们对MS发病机理理解的空白,这将允许在不可逆的中枢神经系统损伤发生之前产生新的疗法来减轻神经变性。
    Neurodegeneration in multiple sclerosis (MS) is believed to underlie disease progression and permanent disability. Many mechanisms of neurodegeneration in MS have been proposed, such as mitochondrial dysfunction, oxidative stress, neuroinflammation, and RNA-binding protein dysfunction. The purpose of this review is to highlight mechanisms of neurodegeneration in MS and its models, with a focus on RNA-binding protein dysfunction. Studying RNA-binding protein dysfunction addresses a gap in our understanding of the pathogenesis of MS, which will allow for novel therapies to be generated to attenuate neurodegeneration before irreversible central nervous system damage occurs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号