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
    基因表达失调对癌症的进展至关重要。hnRNPA1在肝细胞癌(HCC)患者中的表达增强与其致癌功能有关。然而,hnRNPA1上调的潜在机制尚未完全阐明.在本研究中,我们鉴定了microRNA-195-5p(miR-195-5p),在HCC中下调的miRNA,作为控制hnRNPA1表达的新型调节剂。值得注意的是,我们的调查显示,hnRNPA1水平之间呈负相关,在HCC中增加了,和miR-195-5p水平,减少了。我们的发现表明,hnRNPA1通过与调节转移的mRNA相关联,显着增强了PLC/PRF/5细胞的迁移和侵袭。MiR-195-5p还通过靶向hnRNPA1干扰hnRNPA1介导的细胞迁移。我们的结果强调了miR-195-5p/hnRNPA1轴在调节癌细胞的迁移潜力及其通过协调细胞迁移过程促进HCC中的作用中的意义。
    Dysregulation of gene expression is critical for the progression of cancer. The augmented expression of hnRNP A1 in patients with hepatocellular carcinoma (HCC) has been related to its oncogenic functions. However, the underlying mechanisms responsible for upregulation of hnRNP A1 have not been fully elucidated. In the present study, we identified microRNA-195-5p (miR-195-5p), a miRNA downregulated in HCC, as a novel regulator governing hnRNP A1 expression. Notably, our investigations showed an inverse correlation between hnRNP A1 level, which was increased in HCC, and miR-195-5p level, which was decreased. Our findings demonstrated that hnRNP A1 significantly enhanced the migration and invasion of PLC/PRF/5 cells through its association with mRNAs regulating metastasis. MiR-195-5p also interfered with the hnRNP A1-mediated cell migration by targeting hnRNP A1. Our results underscore the significance of the miR-195-5p/hnRNP A1 axis in regulating the migratory potential of cancer cells and its role in promoting HCC by orchestrating cell migration processes.
    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
    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
    人端粒含有DNA序列d(TTAGGG)的多个拷贝,其可以折叠成更高阶的分子内G-四链体并调节端粒长度和染色体完整性的维持。已显示核酸结合蛋白异核核糖核蛋白A1(hnRNPA1)及其N末端蛋白水解产物UP1可有效结合并解折叠端粒DNAG-四链体。然而,对UP1结合和去折叠端粒G-四链体的分子机制的理解仍然有限。这里,我们进行了人端粒DNAG-四链体d[AGGG(TTAGGG)3]的UP1结合和解折叠的生化和生物物理表征,结合UP1中两个串联RNA识别基序(RRM)的系统位点直接诱变,揭示了RRM1负责初始结合和解折叠,而RRM2帮助RRM1完成G-四链体的解折叠。对UP1与DNAG-四链体变体之间相互作用的等温滴定量热法(ITC)和圆二色性(CD)研究表明,端粒G-四链体Loop2中的“TAG”结合基序对于UP1识别和G-四链体解折叠起始至关重要。我们共同描述了hnRNPA1(UP1)结合和人端粒DNAG-四链体展开的分子机制模型。
    The human telomere contains multiple copies of the DNA sequence d(TTAGGG) which can fold into higher order intramolecular G-quadruplexes and regulate the maintenance of telomere length and chromosomal integrity. The nucleic acid binding protein heteronuclear ribonucleoprotein A1 (hnRNP A1) and its N-terminus proteolytic product UP1 have been shown to efficiently bind and unfold telomeric DNA G-quadruplex. However, the understanding of the molecular mechanism of the UP1 binding and unfolding telomeric G-quadruplexes is still limited. Here, we performed biochemical and biophysical characterizations of UP1 binding and unfolding of human telomeric DNA G-quadruplex d[AGGG(TTAGGG)3], and in combination of systematic site-direct mutagenesis of two tandem RNA recognition motifs (RRMs) in UP1, revealed that RRM1 is responsible for initial binding and unfolding, whereas RRM2 assists RRM1 to complete the unfolding of G-quadruplex. Isothermal titration calorimetry (ITC) and circular dichroism (CD) studies of the interactions between UP1 and DNA G-quadruplex variants indicate that the \"TAG\" binding motif in Loop2 of telomeric G-quadruplex is critical for UP1 recognition and G-quadruplex unfolding initiation. Together we depict a model for molecular mechanism of hnRNP A1 (UP1) binding and unfolding of the human telomeric DNA G-quadruplex.
    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
    足细胞功能障碍已被确定为糖尿病肾病(DN)的重要病理特征。然而,长链非编码RNA(lncRNA)在这一过程中的调节作用尚未完全阐明.这里,我们对肾组织进行了无偏RNA测序(RNA-seq)分析,并确定了显着上调的长链非编码RNA,ENST00000585189.1(lncRNA585189),DN患者。此外,lncRNA585189与肾功能不全呈正相关,并且在DN患者和高糖诱导的人足细胞中均上调。功能的增益和丧失实验表明,沉默lncRNA585189降低了ROS的产生,拯救异常的线粒体形态和膜电位,减轻高糖引起的足细胞损伤。机械上,生物信息学分析预测了lncRNA585189和hnRNPA1之间的相互作用,随后被RIP证实,下拉,和EMSA检测。进一步的研究表明,lncRNA585189使hnRNPA1蛋白不稳定,导致其表达下调。相反,hnRNPA1增进了lncRNA585189的表达。此外,RIP和下拉测定均表明hnRNPA1和SIRT1之间存在直接相互作用,这增强了SIRT1mRNA的稳定性.我们的发现表明,lncRNA585189通过hnRNPA1抑制SIRT1,从而阻碍线粒体异常和足细胞损伤的恢复。总之,靶向lncRNA585189是逆转线粒体功能障碍和治疗DN的有前途的策略。
    Podocyte dysfunction has been identified as a crucial pathological characteristic of diabetic nephropathy (DN). However, the regulatory effects of long non-coding RNAs (lncRNAs) in this process have not been fully elucidated. Here, we performed an unbiased RNA-sequencing (RNA-seq) analysis of renal tissues and identified a significantly upregulated long non-coding RNA, ENST00000585189.1 (lncRNA 585189), in patients with DN. Furthermore, lncRNA 585189 was positively correlated with renal insufficiency and was upregulated in both DN patients and high-glucose-induced human podocytes. Gain- and loss-of-function experiments revealed that silencing lncRNA 585189 decreased the production of ROS, rescued aberrant mitochondrial morphology and membrane potential, and alleviated podocyte damage caused by high glucose. Mechanistically, bioinformatics analysis predicted an interaction between lncRNA 585189 and hnRNP A1, which was subsequently confirmed by RIP, pull-down, and EMSA assays. Further investigation revealed that lncRNA 585189 destabilizes the hnRNP A1 protein, leading to the downregulation of its expression. Conversely, hnRNP A1 promoted the expression of lncRNA 585189. Moreover, both RIP and pull-down assays demonstrated a direct interaction between hnRNP A1 and SIRT1, which enhanced SIRT1 mRNA stability. Our findings suggest that lncRNA 585189 suppresses SIRT1 through hnRNP A1, thereby hindering the recovery from mitochondrial abnormalities and podocyte damage. In summary, targeting lncRNA 585189 is a promising strategy for reversing mitochondrial dysfunction and treating DN.
    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
    RUNX2(Runt相关转录因子2)在人牙髓干细胞(hDPSC)的牙源性分化中起关键调节因子的作用。此外,外显子5的包含对于RUNX2功能是重要的。我们先前的研究表明Y-盒结合蛋白1(YBX1)促进RUNX2外显子5包合和hDPSC的矿化。然而,RUNX2外显子5选择性剪接的调控机制有待进一步探索。
    通过RT-PCR和Westernblot分析了hDPSCs牙源性分化过程中异质核核糖核蛋白A1(hnRNPA1)的表达水平。通过功能增益和功能丧失实验分析了hnRNPA1在RUNX2外显子5的可变剪接和牙间充质细胞的牙源性分化中的作用。
    令人惊讶的是,我们发现了一个可变的剪接因子,hnRNPA1在调节hDPSC的RUNX2外显子5包合和牙源性分化中具有与YBX1相反的作用。通过功能增益和丧失试验,我们发现hnRNPA1抑制了RUNX2外显子5的包含,从而抑制了成牙本质细胞的分化。hnRNPA1过表达可抑制碱性磷酸酶(ALP)和骨钙蛋白(OCN)的表达,以及在hDPSC和小鼠牙乳头细胞(mDPC)的牙源性分化过程中矿化结节的形成,而使用hnRNPA1敲除制剂获得了相反的结果。
    本研究表明hnRNPA1抑制RUNX2外显子5包合,降低hDPSC和mDPC的牙源性分化能力。
    RUNX2 (Runt-related transcription factor 2) acts as a key regulator in the odontogenic differentiation of human dental pulp stem cells (hDPSCs). Moreover, the inclusion of exon 5 is important for RUNX2 function. Our previous study showed that Y-Box Binding Protein 1 (YBX1) promoted RUNX2 exon 5 inclusion and mineralization of hDPSCs. However, the regulatory mechanism of RUNX2 exon 5 alternative splicing needed further exploration.
    The expression level of heterogeneous nuclear ribonucleoprotein A1 (hnRNP A1) during the odontogenic differentiation of hDPSCs was analyzed by RT-PCR and Western blot. The roles of hnRNP A1 in the alternative splicing of RUNX2 exon 5 and the odontogenic differentiation of dental mesenchymal cells were analyzed by gain- and loss-of-function experiments.
    Surprisingly, we found an alternative splicing factor, hnRNP A1, which had an opposite role to YBX1 in regulating RUNX2 exon 5 inclusion and odontogenic differentiation of hDPSCs. Through gain- and loss-of-function assay, we found that hnRNP A1 suppressed the inclusion of RUNX2 exon 5, resulting in the inhibition of odontoblastic differentiation. The overexpression of hnRNP A1 can inhibit the expression of ALP (alkaline phosphatase) and OCN (osteocalcin), and the formation of mineralized nodules during the odontogenic differentiation of both hDPSCs and mouse dental papilla cells (mDPCs), whereas the opposite results were obtained with an hnRNP A1 knockdown preparation.
    The present study indicated that hnRNP A1 suppressed RUNX2 exon 5 inclusion and reduced the odontogenic differentiation ability of hDPSCs and mDPCs.
    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
    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
    我们探索与癌细胞营养不良有关的细胞变化的研究表明,血清/葡萄糖饥饿会大大降低异源核核糖核蛋白A1(hnRNPA1)的蛋白质水平。它的损失是可逆的,血清/葡萄糖饥饿特异性,在整个细胞类型和物种中普遍存在。在此条件下,hnRNPA1mRNA水平和hnRNPA1mRNA/蛋白稳定性没有改变。CCND1mRNA,我们新确定为hnRNPA1的结合靶标,通过血清/葡萄糖饥饿降低。在类似条件下,CCND1蛋白在体内和体外均有降低,而hnRNPA1mRNA水平和CCND1mRNA水平在大多数临床样本中没有相关性。功能分析显示,CCND1mRNA的稳定性肯定取决于hnRNPA1蛋白水平,并且hnRNPA1中的RNA识别基序1(RRM1)在维持CCND1mRNA稳定性和随后的蛋白表达中起着核心作用。在小鼠异种移植模型中注射RRM1缺失的hnRNPA1表达癌细胞没有形成任何肿瘤,表达hnRNPA1的癌细胞在坏死附近的病变处保留了CCND1的表达,肿瘤体积略有增加。此外,RRM1缺失引起生长抑制,诱导细胞凋亡和自噬,而CCND1恢复完全恢复。我们的结果表明,血清/葡萄糖饥饿引发整个hnRNPA1蛋白丢失,其丢失可能在CCND1mRNA不稳定和CCND1介导的细胞事件抑制中起作用,即,促进增长,凋亡诱导,和自噬体的形成。
    Our investigation to explore cellular alterations related to undernutrition in cancer cells revealed that the protein level of heterogenous nuclear ribonucleoprotein A1 (hnRNP A1) is drastically decreased by serum/glucose starvation. Its loss was reversible, serum/glucose starvation-specific and universal throughout cell types and species. The hnRNP A1 mRNA level and hnRNP A1 mRNA/protein stability were not altered under this condition. CCND1 mRNA, which we newly identified as the binding target of hnRNP A1, was decreased by serum/glucose starvation. Under similar conditions, CCND1 protein was reduced in vitro and in vivo, whereas hnRNP A1 mRNA level and CCND1 mRNA level revealed no correlation in most clinical samples. Functional analyses revealed that CCND1 mRNA stability is certainly dependent on hnRNP A1 protein level and that RNA recognition motif-1 (RRM1) in hnRNP A1 plays a central role in maintaining CCND1 mRNA stability and subsequent protein expression. The injection of RRM1-deleted hnRNP A1-expressing cancer cells in the mouse xenograft model did not form any tumours, and that of hnRNP A1-expressing cancer cells retained CCND1 expression at the lesion adjacent to necrosis with a slight increase in tumour volume. Furthermore, RRM1 deletion caused growth suppression with the induction of apoptosis and autophagy, whereas CCND1 restoration completely recovered it. Our results indicate that serum/glucose starvation triggers entire hnRNP A1 protein loss, and its loss may play a role in CCND1 mRNA destabilization and CCND1-mediated cellular event inhibition, i.e. growth promotion, apoptosis induction and autophagosome formation.
    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
    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
    少突胶质细胞(OL)损伤和死亡是多发性硬化(MS)病理的突出特征,然而,导致OL损失的机制尚不完全清楚。功能失调的RNA结合蛋白(RBPs),以核细胞质错误定位和表达改变为标志,已被证明会导致神经系统疾病中的细胞丢失,包括在女士由于我们先前观察到RBP异质核核糖核蛋白A1(hnRNPA1)在MS的神经元中功能失调,我们假设它也可能导致MS和相关模型中的OL病理。我们发现hnRNPA1功能障碍是MS大脑中OLs的特征。这些发现在MS的实验性自身免疫性脑脊髓炎(EAE)小鼠模型中进行了概述,其中hnRNPA1功能障碍是OLs的特征,包括少突胶质前体细胞和成熟OLs,其中hnRNPA1功能障碍与脱髓鞘相关。我们还发现IFNγ诱导hnRNPA1功能障碍,表明炎症影响hnRNPA1功能。为了充分了解hnRNPA1功能障碍对OLs的影响,我们对hnRNPA1进行siRNA敲除,然后进行RNA测序.RNA测序检测到超过4000个差异表达的转录物,揭示了RNA代谢的改变,细胞形态学,和程序性细胞死亡途径。我们证实hnRNPA1敲低对OLs有害,并诱导细胞凋亡和坏死。一起,这些数据表明hnRNPA1在正常OL功能和生存中的关键作用,并提示MS中OL损伤和死亡的潜在机制,该机制涉及hnRNPA1功能障碍.
    Oligodendrocyte (OL) damage and death are prominent features of multiple sclerosis (MS) pathology, yet mechanisms contributing to OL loss are incompletely understood. Dysfunctional RNA binding proteins (RBPs), hallmarked by nucleocytoplasmic mislocalization and altered expression, have been shown to result in cell loss in neurologic diseases, including in MS. Since we previously observed that the RBP heterogeneous nuclear ribonucleoprotein A1 (hnRNP A1) was dysfunctional in neurons in MS, we hypothesized that it might also contribute to OL pathology in MS and relevant models. We discovered that hnRNP A1 dysfunction is characteristic of OLs in MS brains. These findings were recapitulated in the experimental autoimmune encephalomyelitis (EAE) mouse model of MS, where hnRNP A1 dysfunction was characteristic of OLs, including oligodendrocyte precursor cells and mature OLs in which hnRNP A1 dysfunction correlated with demyelination. We also found that hnRNP A1 dysfunction was induced by IFNγ, indicating that inflammation influences hnRNP A1 function. To fully understand the effects of hnRNP A1 dysfunction on OLs, we performed siRNA knockdown of hnRNP A1, followed by RNA sequencing. RNA sequencing detected over 4000 differentially expressed transcripts revealing alterations to RNA metabolism, cell morphology, and programmed cell death pathways. We confirmed that hnRNP A1 knockdown was detrimental to OLs and induced apoptosis and necroptosis. Together, these data demonstrate a critical role for hnRNP A1 in proper OL functioning and survival and suggest a potential mechanism of OL damage and death in MS that involves hnRNP A1 dysfunction.
    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