PRMT5

PRMT5
  • 文章类型: Journal Article
    耐紫杉醇的三阴性乳腺癌(TNBC)仍然是治疗最具挑战性的乳腺癌之一。这里,使用表观遗传化学探针筛选,我们揭示了紫杉醇耐药TNBC细胞对蛋白精氨酸甲基转移酶(PRMTs)抑制的获得性脆弱性.细胞系和内部临床样品的分析表明,抗性细胞通过稳定有丝分裂染色质组装来逃避紫杉醇的杀伤。PRMT5的遗传或药理学抑制改变RNA剪接,特别是极光激酶B(AURKB)的内含子保留,导致蛋白质表达减少,并最终导致紫杉醇耐药细胞的选择性有丝分裂灾难。此外,通过增加AURKB介导的有丝分裂信号通路的扰动,I型PRMT抑制与PRMT5抑制在抑制耐药细胞的肿瘤生长中的协同作用。这些发现在从紫杉醇耐药的TNBC患者产生的患者来源的异种移植(PDX)模型中得到了充分概括,提供了在紫杉醇耐药的TNBC中靶向PRMT的基本原理。
    Paclitaxel-resistant triple negative breast cancer (TNBC) remains one of the most challenging breast cancers to treat. Here, using an epigenetic chemical probe screen, we uncover an acquired vulnerability of paclitaxel-resistant TNBC cells to protein arginine methyltransferases (PRMTs) inhibition. Analysis of cell lines and in-house clinical samples demonstrates that resistant cells evade paclitaxel killing through stabilizing mitotic chromatin assembly. Genetic or pharmacologic inhibition of PRMT5 alters RNA splicing, particularly intron retention of aurora kinases B (AURKB), leading to a decrease in protein expression, and finally results in selective mitosis catastrophe in paclitaxel-resistant cells. In addition, type I PRMT inhibition synergies with PRMT5 inhibition in suppressing tumor growth of drug-resistant cells through augmenting perturbation of AURKB-mediated mitotic signaling pathway. These findings are fully recapitulated in a patient-derived xenograft (PDX) model generated from a paclitaxel-resistant TNBC patient, providing the rationale for targeting PRMTs in paclitaxel-resistant TNBC.
    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
    蛋白质精氨酸甲基化是由9种蛋白质精氨酸甲基转移酶(PRMT)催化的常见翻译后修饰(PTM)。作为甲基化组蛋白和非组蛋白底物的主要对称精氨酸甲基转移酶,PRMT5在许多对发育和肿瘤发生至关重要的生物过程中发挥关键作用。据报道,PRMT5在多种癌症类型中过度表达,包括前列腺癌(PCa),但对PRMT5在侵袭性PCa中的确切生物学和机制理解仍不明确.这里,我们发现PRMT5在PCa中上调,与更糟糕的患者生存率相关,促进破坏的RNA剪接,并在功能上与一系列促肿瘤发生途径合作以增强肿瘤发生。通过基因敲低或药理学抑制的PRMT5抑制降低了具有平行分化的干性并阻止了细胞周期进展而不引起明显的细胞凋亡。引人注目的是,PRMT5抑制的抗肿瘤作用的严重程度与疾病侵袭性相关,AR+PCa受影响较小。分子表征精确定位MYC,但不是(或至少在较小程度上)AR,作为PRMT5的主要伴侣,形成正反馈回路,加剧AR+和AR-PCa细胞的恶性肿瘤。受以下惊人发现的启发:PRMT5与肿瘤免疫浸润呈负相关,转录抑制免疫基因程序,我们进一步表明,尽管PRMT5抑制剂(PRMT5i)EPZ015666或单独的抗PD-1免疫疗法表现出有限的抗肿瘤作用,PRMT5i与抗PD-1的组合在体内抑制去势抗性PCa(CRPC)方面表现出优异的功效。最后,通过合成杀伤力概念扩大PRMT5i的潜在用途,我们还进行了一项全球性的CRISPR/Cas9基因敲除筛选,以揭示许多已知致癌途径的临床级药物在低剂量与PRMT5i联合使用时可重新用于靶向CRPC.总的来说,我们的研究结果为利用PRMT5i联合免疫疗法或其他靶向疗法治疗侵袭性PCa奠定了基础.
    Protein arginine methylation is a common post-translational modification (PTM) catalyzed by nine protein arginine methyltransferases (PRMTs). As the major symmetric arginine methyltransferase that methylates both histone and non-histone substrates, PRMT5 plays key roles in a number of biological processes critical for development and tumorigenesis. PRMT5 overexpression has been reported in multiple cancer types including prostate cancer (PCa), but the exact biological and mechanistic understanding of PRMT5 in aggressive PCa remains ill-defined. Here, we show that PRMT5 is upregulated in PCa, correlates with worse patient survival, promotes corrupted RNA splicing, and functionally cooperates with an array of pro-tumorigenic pathways to enhance oncogenesis. PRMT5 inhibition via either genetic knockdown or pharmacological inhibition reduces stemness with paralleled differentiation and arrests cell cycle progression without causing appreciable apoptosis. Strikingly, the severity of antitumor effect of PRMT5 inhibition correlates with disease aggressiveness, with AR+ PCa being less affected. Molecular characterization pinpoints MYC, but not (or at least to a lesser degree) AR, as the main partner of PRMT5 to form a positive feedback loop to exacerbate malignancy in both AR+ and AR- PCa cells. Inspired by the surprising finding that PRMT5 negatively correlates with tumor immune infiltration and transcriptionally suppresses an immune-gene program, we further show that although PRMT5 inhibitor (PRMT5i) EPZ015666 or anti-PD-1 immunotherapy alone exhibits limited antitumor effects, combination of PRMT5i with anti-PD-1 displays superior efficacy in inhibiting castration-resistant PCa (CRPC) in vivo. Finally, to expand the potential use of PRMT5i through a synthetic lethality concept, we also perform a global CRISPR/Cas9 knockout screen to unravel that many clinical-grade drugs of known oncogenic pathways can be repurposed to target CRPC when used in combination with PRMT5i at low doses. Collectively, our findings establish a rationale to exploit PRMT5i in combination with immunotherapy or other targeted therapies to treat aggressive PCa.
    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
    乳腺癌是全球女性中最常见的癌症。早期乳腺癌在约70-80%的患者中可以治愈,而晚期转移性乳腺癌被认为是目前疗法无法治愈的。乳腺癌是一种高度异质性的疾病,可根据关键标记分为三种主要亚型,为每种亚型制定特定的治疗策略。乳腺癌发生的复杂性通常与调节不同信号通路的表观遗传修饰有关。参与乳腺肿瘤的发生和进展,特别是精氨酸残基的甲基化。蛋白质精氨酸甲基转移酶(PRMT1-9)已经出现,通过它们甲基化组蛋白和非组蛋白底物的能力,作为癌症的重要监管者。这里,我们对PRMT家族的两个主要成员PRMT1和PRMT5的机制进行了更新概述,控制影响乳腺肿瘤发生的重要信号通路,强调它们是推定的治疗目标。
    Breast cancer is the most common cancer diagnosed in women worldwide. Early-stage breast cancer is curable in ~70-80% of patients, while advanced metastatic breast cancer is considered incurable with current therapies. Breast cancer is a highly heterogeneous disease categorized into three main subtypes based on key markers orientating specific treatment strategies for each subtype. The complexity of breast carcinogenesis is often associated with epigenetic modification regulating different signaling pathways, involved in breast tumor initiation and progression, particularly by the methylation of arginine residues. Protein arginine methyltransferases (PRMT1-9) have emerged, through their ability to methylate histones and non-histone substrates, as essential regulators of cancers. Here, we present an updated overview of the mechanisms by which PRMT1 and PRMT5, two major members of the PRMT family, control important signaling pathways impacting breast tumorigenesis, highlighting them as putative therapeutic targets.
    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
    蛋白质精氨酸甲基转移酶5(PRMT5)通过许多RNA结合蛋白中精氨酸残基的对称二甲基化(Rme2s/SDMA)调节RNA剪接和转录。然而,PRMT5将剪接与转录输出偶联的机制尚不清楚.这里,我们证明了PRMT5活性的一个主要功能是促进新型染色质逃逸,我们称之为基因组保留不完全处理的聚腺苷酸化转录本(GRIPPs)的一大类mRNA。使用新生和总转录组学,刺入受控分级细胞转录组学,和总的和分级的细胞蛋白质组学,我们显示PRMT5抑制和敲低PRMT5SNRP(Sm蛋白)接头蛋白pICln(CLNS1A)-而不是I型PRMT抑制-导致mRNA的严重滞留,SNRPB,和染色质上的SNRPD3蛋白。与大多数成绩单相比,这些染色质捕获的多聚腺苷酸化RNA转录物具有更多的内含子,拼接速度较慢,并富含被拘留的内含子。使用PRMT5抑制和诱导型等基因野生型和精氨酸突变体SNRPB的组合,我们表明,这些snRNPs的精氨酸甲基化对于介导它们的稳态染色质和RNA相互作用至关重要。总的来说,我们得出结论,PRMT5的主要作用是控制转录本加工和剪接完成,以促进染色质逃逸和随后的核输出.
    Protein Arginine Methyltransferase 5 (PRMT5) regulates RNA splicing and transcription by symmetric dimethylation of arginine residues (Rme2s/SDMA) in many RNA binding proteins. However, the mechanism by which PRMT5 couples splicing to transcriptional output is unknown. Here, we demonstrate that a major function of PRMT5 activity is to promote chromatin escape of a novel, large class of mRNAs that we term Genomically Retained Incompletely Processed Polyadenylated Transcripts (GRIPPs). Using nascent and total transcriptomics, spike-in controlled fractionated cell transcriptomics, and total and fractionated cell proteomics, we show that PRMT5 inhibition and knockdown of the PRMT5 SNRP (Sm protein) adapter protein pICln (CLNS1A) -but not type I PRMT inhibition-leads to gross detention of mRNA, SNRPB, and SNRPD3 proteins on chromatin. Compared to most transcripts, these chromatin-trapped polyadenylated RNA transcripts have more introns, are spliced slower, and are enriched in detained introns. Using a combination of PRMT5 inhibition and inducible isogenic wildtype and arginine-mutant SNRPB, we show that arginine methylation of these snRNPs is critical for mediating their homeostatic chromatin and RNA interactions. Overall, we conclude that a major role for PRMT5 is in controlling transcript processing and splicing completion to promote chromatin escape and subsequent nuclear export.
    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
    癌症是全球主要的死亡原因,由各种环境因素影响的基因的非遗传和遗传改变触发。(THIQ),特别是1,2,3,4-四氢呋喃异喹啉是各种生物碱的基本元素,在喹啉和吲哚生物碱附近普遍存在。
    在这篇评论中,研究了2016年至2024年THIQ衍生物作为抗癌剂的治疗应用.这些专利是通过对Espacenet的全面搜索收集的,谷歌专利,WIPO,和SciFinder数据库。专利中涵盖的治疗领域包括许多癌症靶标。
    THIQ类似物在药物化学中起着至关重要的作用,其中许多是药理学过程和临床试验不可或缺的。已经合成了许多THIQ化合物用于治疗目的,尤其是在癌症治疗中。他们在开发抗癌药物方面显示出巨大的希望,对各种癌症靶标表现出强亲和力和功效。多靶标配体的创建是基于THIQ的抗癌药物发现的引人注目的途径。
    UNASSIGNED: Cancer is a prominent cause of death globally, triggered by both non-genetic and genetic alterations in genes influenced by various environmental factors. The tetrahydroisoquinoline (THIQ), specifically 1,2,3,4-tetrahydroisoquinoline serves as fundamental element in various alkaloids, prevalent in proximity to quinoline and indole alkaloids.
    UNASSIGNED: In this review, the therapeutic applications of THIQ derivatives as an anticancer agent from 2016 to 2024 have been examined. The patents were gathered through comprehensive searches of the Espacenet, Google patent, WIPO, and Sci Finder databases. The therapeutic areas encompassed in the patents include numerous targets of cancer.
    UNASSIGNED: THIQ analogues play a crucial role in medicinal chemistry, with many being integral to pharmacological processes and clinical trials. Numerous THIQ compounds have been synthesized for therapeutic purposes, notably in cancer treatment. They show great promise for developing anticancer drugs, demonstrating strong affinity and efficacy against various cancer targets. The creation of multi-target ligands is a compelling avenue for THIQ-based anticancer drug discovery.
    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
    研究透明细胞肾细胞癌(ccRCC)的潜在机制,最常见的肾癌亚型,可以解决ccRCC靶向药物研究中未满足的需求。越来越多的证据表明,蛋白磷酸酶4(PP4)在癌症生物学中起着重要作用。这里,我们使用组织微阵列表征了ccRCC中PP4核心组分SMEK1的上调,并揭示其高表达与患者生存率降低密切相关.然后我们进行了细胞功能实验和动物实验来证明SMEK1的促肿瘤作用。接下来,进行RNA-seq以探索其潜在的机制,结果表明,SMEK1调控的基因广泛参与细胞运动,典型的酪氨酸激酶受体EGFR是其靶点之一。此外,通过分子实验验证了SMEK1对EGFR及其下游MAPK和AKT通路的调节作用,其中厄洛替尼,酪氨酸激酶抑制剂,可以部分阻止这一规定,证明SMEK1介导其作用依赖于EGFR的酪氨酸激酶活性。机械上,SMEK1与PRMT5结合并促进PRMT5介导的组蛋白甲基化以促进EGFR的转录。此外,我们研究了SMEK1的上游调节因子,并证明转录因子E2F1可以通过染色质免疫沉淀直接结合SMEK1启动子。功能上,E2F1还可以通过操纵SMEK1的表达来诱导ccRCC的进展。总的来说,我们的发现表明SMEK1在ccRCC中过度表达,并揭示了一种新的E2F1/SMEK1/PRMT5/EGFR-酪氨酸激酶依赖性途径用于ccRCC进展。
    Studying the mechanisms underlying clear cell renal cell carcinoma (ccRCC), the most common subtype of kidney cancer, may address an unmet need in ccRCC-targeted drug research. Growing evidences indicate that protein phosphatase 4 (PP4) plays an important role in cancer biology. Here, we characterized the upregulation of PP4 core component SMEK1 in ccRCC using tissue microarrays and revealed that its high expression is closely associated with reduced patient survival. We then conducted cell function experiments and animal experiments to prove the tumor-promoting effect of SMEK1. Next, RNA-seq was performed to explore its underlying mechanism, and the results revealed that SMEK1-regulated genes were extensively involved in cell motility, and the canonical tyrosine kinase receptor EGFR was one of its targets. Moreover, we verified the regulatory effect of SMEK1 on EGFR and its downstream MAPK and AKT pathway through molecular experiments, in which erlotinib, a tyrosine kinase inhibitor, can partially block this regulation, demonstrating that SMEK1 mediates its effects dependent on the tyrosine kinase activity of EGFR. Mechanistically, SMEK1 bond to PRMT5 and facilitated PRMT5-mediated histone methylation to promote the transcription of EGFR. Furthermore, we studied the upstream regulators of SMEK1 and demonstrated that the transcription factor E2F1 could directly bind to the SMEK1 promoter by chromatin immunoprecipitation. Functionally, E2F1 could also induce ccRCC progression by manipulating the expression of SMEK1. Collectively, our findings demonstrate the overexpression of SMEK1 in ccRCC, and reveal a novel E2F1/SMEK1/PRMT5/EGFR-tyrosine-kinase-dependent pathway for ccRCC progression.
    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
    PRMT5是一种广泛表达的精氨酸甲基转移酶,可调节涉及肿瘤细胞增殖和存活的过程。在这里描述的研究中,我们调查了PRMT5是否为肿瘤放射增敏提供了靶点.使用siRNA敲除PRMT5增强了一组对应于通常用放射疗法治疗的肿瘤类型的细胞系的放射敏感性。为了将这些研究扩展到实验性治疗环境,使用PRMT5抑制剂LLY-283.肿瘤细胞系暴露于LLY-283会降低PRMT5活性并增强其放射敏感性。通过γH2AX病灶和中性彗星分析确定,放射敏感性的增加伴随着DNA双链断裂修复的抑制。对于正常的成纤维细胞系,虽然LLY-283降低了PRMT5活性,对他们的放射敏感性没有影响.对U251细胞的转录组分析表明,LLY-283处理降低了基因的表达并改变了参与DNA损伤反应的基因的mRNA剪接模式。然后使用皮下异种移植物来评估对LLY-283和辐射的体内反应。用LLY-283治疗小鼠降低了肿瘤PRMT5的活性,并显着增强了辐射诱导的生长延迟。这些结果表明PRMT5是放射增敏的肿瘤选择性靶标。
    PRMT5 is a widely expressed arginine methyltransferase that regulates processes involved in tumor cell proliferation and survival. In the study described here, we investigated whether PRMT5 provides a target for tumor radiosensitization. Knockdown of PRMT5 using siRNA enhanced the radiosensitivity of a panel of cell lines corresponding to tumor types typically treated with radiotherapy. To extend these studies to an experimental therapeutic setting, the PRMT5 inhibitor LLY-283 was used. Exposure of the tumor cell lines to LLY-283 decreased PRMT5 activity and enhanced their radiosensitivity. This increase in radiosensitivity was accompanied by an inhibition of DNA double-strand break repair as determined by γH2AX foci and neutral comet analyses. For a normal fibroblast cell line, although LLY-283 reduced PRMT5 activity, it had no effect on their radiosensitivity. Transcriptome analysis of U251 cells showed that LLY-283 treatment reduced the expression of genes and altered the mRNA splicing pattern of genes involved in the DNA damage response. Subcutaneous xenografts were then used to evaluate the in vivo response to LLY-283 and radiation. Treatment of mice with LLY-283 decreased tumor PRMT5 activity and significantly enhanced the radiation-induced growth delay. These results suggest that PRMT5 is a tumor selective target for radiosensitization.
    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
    蛋白质精氨酸甲基转移酶5(PRMT5)在多种癌症中过表达,并被认为具有关键的致癌作用。部分通过其控制主转录调节因子E2F1来实现。我们调查了PRMT5和E2F1在神经母细胞瘤(NB)中的相关性,发现PRMT5和E2F1的表达升高发生在预后不良的高风险疾病中,并与扩增的骨髓细胞瘤病毒相关的癌基因相关。神经母细胞瘤衍生(MYCN)基因。我们的结果表明,MYCN驱动剪接因子基因的表达,与PRMT5和E2F1一起,导致一个去调节的选择性RNA剪接程序,阻止细胞凋亡。PRMT5的药理学抑制或E2F1的失活恢复正常剪接并使NB细胞对凋亡敏感。我们的发现表明,持续的癌症相关的选择性RNA剪接程序使NB细胞对凋亡脱敏,并确定PRMT5作为高危疾病的潜在治疗靶点。
    Protein arginine methyltransferase 5 (PRMT5) is over-expressed in a wide variety of cancers and is implicated as having a key oncogenic role, achieved in part through its control of the master transcription regulator E2F1. We investigated the relevance of PRMT5 and E2F1 in neuroblastoma (NB) and found that elevated expression of PRMT5 and E2F1 occurs in poor prognosis high-risk disease and correlates with an amplified Myelocytomatosis viral-related oncogene, neuroblastoma-derived (MYCN) gene. Our results show that MYCN drives the expression of splicing factor genes that, together with PRMT5 and E2F1, lead to a deregulated alternative RNA splicing programme that impedes apoptosis. Pharmacological inhibition of PRMT5 or inactivation of E2F1 restores normal splicing and renders NB cells sensitive to apoptosis. Our findings suggest that a sustained cancer-relevant alternative RNA splicing programme desensitises NB cells to apoptosis, and identify PRMT5 as a potential therapeutic target for high-risk disease.
    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
    维甲酸受体相关孤儿受体α(RORα),候选肿瘤抑制因子,在恶性乳腺癌细胞中普遍下调或丢失。然而,乳腺上皮细胞中RORα表达的调控机制尚不完全清楚。蛋白质精氨酸N-甲基转移酶5(PRMT5),一种II型甲基转移酶,催化靶蛋白中氨基酸精氨酸的对称甲基化,据报道可以调节蛋白质的稳定性。为了研究PRMT5是否以及如何调节RORα,我们通过免疫沉淀和GST下拉法检测了RORα和PRMT5之间的直接相互作用。结果表明,PRMT5直接与RORα结合,和PRMT5主要对称地二甲基化RORα的DNA结合域(DBD),而不是配体结合域(LBD)。为了研究RORα蛋白稳定性是否受PRMT5调节,我们用RORα和PRMT5表达或PRMT5沉默(shPRMT5)载体转染HEK293FT细胞,然后通过环己酰亚胺追踪测定法检查RORα蛋白稳定性。结果表明,PRMT5增加了RORα蛋白的稳定性,而沉默PRMT5加速RORα蛋白降解。在PRMT5沉默的乳腺上皮细胞中,RORα蛋白表达降低,伴随着增强的上皮-间质转化形态和细胞侵袭和迁移能力。在PRMT5过表达的乳腺上皮细胞中,RORα蛋白积累,细胞侵袭被抑制。这些发现揭示了PRMT5调节RORα蛋白稳定性的新机制。
    Retinoic acid receptor-related orphan receptor alpha (RORα), a candidate tumor suppressor, is prevalently downregulated or lost in malignant breast cancer cells. However, the mechanisms of how RORα expression is regulated in breast epithelial cells remain incompletely understood. Protein arginine N-methyltransferase 5 (PRMT5), a type II methyltransferase catalyzing the symmetric methylation of the amino acid arginine in target proteins, was reported to regulate protein stability. To study whether and how PRMT5 regulates RORα, we examined the direct interaction between RORα and PRMT5 by immunoprecipitation and GST pull-down assays. The results showed that PRMT5 directly bound to RORα, and PRMT5 mainly symmetrically dimethylated the DNA-binding domain (DBD) but not the ligand-binding domain (LBD) of RORα. To investigate whether RORα protein stability is regulated by PRMT5, we transfected HEK293FT cells with RORα and PRMT5-expressing or PRMT5-silencing (shPRMT5) vectors and then examined RORα protein stability by a cycloheximide chase assay. The results showed that PRMT5 increased RORα protein stability, while silencing PRMT5 accelerated RORα protein degradation. In PRMT5-silenced mammary epithelial cells, RORα protein expression was decreased, accompanied by an enhanced epithelial-mesenchymal transition morphology and cell invasion and migration abilities. In PRMT5-overexpressed mammary epithelial cells, RORα protein was accumulated, and cell invasion was suppressed. These findings revealed a novel mechanism by which PRMT5 regulates RORα protein stability.
    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
    Dickkopf家族蛋白(DKK)是强Wnt信号拮抗剂,在结直肠癌(CRC)的发展和进展中起重要作用。最近的工作表明,DKK,主要是DKK1与CRC中化学抗性的诱导有关,并且癌细胞中DKK1的表达与蛋白质精氨酸N-甲基转移酶5(PRMT5)的表达有关。这表明在DKK1和PRMT5之间存在调节环。在这里,我们解决了PRMT5是否有助于CRC中DKK1的表达以及CRC化学耐药的问题.使用计算机模拟和体外方法来探索PRMT5与不同DKK成员之间的关系。我们的数据表明,DKK1表达在CRC临床样本中显著上调,特别是KRAS突变的CRC,DKK1的水平与PRMT5激活呈正相关。染色质免疫沉淀(ChIP)数据表明PRMT5在调节DKK1中可能具有表观遗传作用,可能是通过H3R8的对称二甲基化。敲除DKK1或用PRMT5抑制剂CMP5与阿霉素联合治疗在KRAS突变体中产生了协同抗肿瘤作用,但不是KRAS野生型,CRC细胞。这些发现表明PRMT5调节CRC中的DKK1表达,并且PRMT5的抑制以减少CRC细胞生长的方式调节DKK1表达。
    The Dickkopf family proteins (DKKs) are strong Wnt signaling antagonists that play a significant role in colorectal cancer (CRC) development and progression. Recent work has shown that DKKs, mainly DKK1, are associated with the induction of chemoresistance in CRC and that DKK1 expression in cancer cells correlates with that of protein arginine N-methyltransferase 5 (PRMT5). This points to the presence of a regulatory loop between DKK1 and PRMT5. Herein, we addressed the question of whether PRMT5 contributes to DKK1 expression in CRC and hence CRC chemoresistance. Both in silico and in vitro approaches were used to explore the relationship between PRMT5 and different DKK members. Our data demonstrated that DKK1 expression is significantly upregulated in CRC clinical samples, KRAS-mutated CRC in particular and that the levels of DKK1 positively correlate with PRMT5 activation. Chromatin immunoprecipitation (ChIP) data indicated a possible epigenetic role of PRMT5 in regulating DKK1, possibly through the symmetric dimethylation of H3R8. Knockdown of DKK1 or treatment with the PRMT5 inhibitor CMP5 in combination with doxorubicin yielded a synergistic anti-tumor effect in KRAS mutant, but not KRAS wild-type, CRC cells. These findings suggest that PRMT5 regulates DKK1 expression in CRC and that inhibition of PRMT5 modulates DKK1 expression in such a way that reduces CRC cell growth.
    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