Tumor suppressor

肿瘤抑制物
  • 文章类型: Journal Article
    高危型人乳头瘤病毒(HPV)癌蛋白使细胞肿瘤抑制因子失活,从而重新编程宿主细胞信号通路。HPVE7蛋白结合并降解肿瘤抑制因子PTPN14,从而促进YAP1癌蛋白的核定位并抑制角质形成细胞分化。YAP1是驱动上皮细胞干性和自我更新的转录共激活因子。YAP1活性被高度保守的Hippo途径抑制,在人类癌症中经常失活。MST1/2和LATS1/2激酶形成Hippo激酶级联的核心。活性LATS1激酶在苏氨酸1079上磷酸化,并通过在包括丝氨酸127在内的氨基酸上磷酸化来抑制YAP1。这里,我们检测了高危(致癌)HPV18E7对Hippo通路活性的影响.我们发现PTPN14敲除或HPV18E7降解PTPN14可降低人角质形成细胞中LATS1T1079和YAP1S127的磷酸化并抑制角质形成细胞的分化。相反,PTPN14依赖性分化需要PTPN14中的LATS激酶和某些PPxY基序。PTPN14促进分化既不需要MST1/2激酶也不需要推定的PTPN14磷酸酶活性位点。一起,这些数据支持PTPN14被HPV18E7灭活或降解PTPN14降低LATS1活性,促进活性YAP1和抑制角质形成细胞分化。重要提示Hippo激酶级联抑制YAP1,一种癌蛋白和细胞干性和自我更新的驱动因素。越来越多的证据表明,Hippo途径被包括人乳头瘤病毒在内的肿瘤病毒所靶向。高危HPVE7癌蛋白促进YAP1核定位,高危HPVE7的致癌活性需要YAP1活性。阻断HPVE7依赖性YAP1激活可以抑制HPV介导的癌变,但HPVE7激活YAP1的机制尚未阐明.在这里,我们报道通过降解肿瘤抑制因子PTPN14,HPV18E7抑制LATS1激酶,减少对YAP1的抑制磷酸化。这些数据支持HPV癌蛋白可以抑制Hippo信号传导以激活YAP1并加强人上皮细胞中PTPN14和Hippo信号传导之间的联系。
    High-risk human papillomavirus (HPV) oncoproteins inactivate cellular tumor suppressors to reprogram host cell signaling pathways. HPV E7 proteins bind and degrade the tumor suppressor PTPN14, thereby promoting the nuclear localization of the YAP1 oncoprotein and inhibiting keratinocyte differentiation. YAP1 is a transcriptional coactivator that drives epithelial cell stemness and self-renewal. YAP1 activity is inhibited by the highly conserved Hippo pathway, which is frequently inactivated in human cancers. MST1/2 and LATS1/2 kinases form the core of the Hippo kinase cascade. Active LATS1 kinase is phosphorylated on threonine 1079 and inhibits YAP1 by phosphorylating it on amino acids including serine 127. Here, we tested the effect of high-risk (carcinogenic) HPV18 E7 on Hippo pathway activity. We found that either PTPN14 knockout or PTPN14 degradation by HPV18 E7 decreased the phosphorylation of LATS1 T1079 and YAP1 S127 in human keratinocytes and inhibited keratinocyte differentiation. Conversely, PTPN14-dependent differentiation required LATS kinases and certain PPxY motifs in PTPN14. Neither MST1/2 kinases nor the putative PTPN14 phosphatase active sites were required for PTPN14 to promote differentiation. Together, these data support that PTPN14 inactivation or degradation of PTPN14 by HPV18 E7 reduce LATS1 activity, promoting active YAP1 and inhibiting keratinocyte differentiation.IMPORTANCEThe Hippo kinase cascade inhibits YAP1, an oncoprotein and driver of cell stemness and self-renewal. There is mounting evidence that the Hippo pathway is targeted by tumor viruses including human papillomavirus. The high-risk HPV E7 oncoprotein promotes YAP1 nuclear localization and the carcinogenic activity of high-risk HPV E7 requires YAP1 activity. Blocking HPV E7-dependent YAP1 activation could inhibit HPV-mediated carcinogenesis, but the mechanism by which HPV E7 activates YAP1 has not been elucidated. Here we report that by degrading the tumor suppressor PTPN14, HPV18 E7 inhibits LATS1 kinase, reducing inhibitory phosphorylation on YAP1. These data support that an HPV oncoprotein can inhibit Hippo signaling to activate YAP1 and strengthen the link between PTPN14 and Hippo signaling in human epithelial cells.
    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
    在过去的十年中,癌症治疗方法的进步显着降低了这些患者的死亡率。然而,癌症仍然被认为是人类死亡的主要原因之一。癌症患者高逝世亡率的主要缘由之一是晚期肿瘤分期晚期诊断。因此,为了引入早期诊断标志物,有必要研究肿瘤进展的分子生物学。MicroRNAs(miRNA)在调节与肿瘤进展相关的细胞过程中具有重要作用。由于miRNA在体液中的高稳定性,它们被广泛用作早期肿瘤诊断的非侵入性标志物。因为,已经报道了miR-98在多种癌症中的失调,我们研究了miR-98在肿瘤进展过程中的分子机制.据报道,miR-98主要通过调节转录因子和信号通路来抑制肿瘤的生长。因此,miR-98可以作为肿瘤标志物和治疗靶标引入癌症患者中。
    The progress of cancer treatment methods in the last decade has significantly reduced mortality rate among these patients. Nevertheless, cancer is still recognized as one of the main causes of human deaths. One of the main reasons for the high death rate in cancer patients is the late diagnosis in the advanced tumor stages. Therefore, it is necessary to investigate the molecular biology of tumor progressions in order to introduce early diagnostic markers. MicroRNAs (miRNAs) have an important role in regulating cellular processes associated with tumor progression. Due to the high stability of miRNAs in body fluids, they are widely used as non-invasive markers in the early tumor diagnosis. Since, deregulation of miR-98 has been reported in a wide range of cancers, we investigated the molecular mechanisms of miR-98 during tumor progression. It has been reported that miR-98 mainly inhibits the tumor growth by the modulation of transcription factors and signaling pathways. Therefore, miR-98 can be introduced as a tumor marker and therapeutic target among cancer patients.
    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
    临床上,前列腺癌因其组织学和分子异质性而臭名昭著,这给精确的治疗和药物开发带来了巨大的挑战。为了克服这些困难,除了遗传改变和表观遗传调控外,研究人员还致力于调节肿瘤微环境和免疫反应。这里,我们旨在通过研究在已建立的抗癌药治疗的前列腺癌细胞中特异性改变的基因来鉴定潜在的前列腺癌生物标志物或调节剂.甘油激酶1(GK1)是在X染色体上编码的磷酸转移酶,与甘油三酯和甘油磷脂的合成有关,主要研究X连锁代谢紊乱GK缺乏症(GKD)。有趣的是,我们的DNA微阵列分析表明,几种抗癌药物高度诱导GK1的表达,特别是GK1a和GK1b亚型,在人类前列腺癌PC-3细胞中。为了阐明GK1与癌细胞死亡之间的关系,构建人GK1b特异性表达载体并转染PC-3细胞。令人惊讶的是,GK1b过表达显著降低细胞活力并显著加速凋亡性细胞死亡。这些发现表明,GK1b可能作为前列腺癌细胞死亡的一个有前途的调节剂和生物标志物。提供治疗干预的潜在途径。
    Clinically, prostate cancer is infamous for its histological and molecular heterogeneity, which causes great challenges to pinpoint therapy and pharmaceutical development. To overcome these difficulties, researchers are focusing on modulating tumor microenvironment and immune responses in addition to genetic alteration and epigenetic regulation. Here, we aimed to identify potential biomarkers or modulators of prostate cancer by investigating genes specifically altered in prostate cancer cells treated with established anti-cancer agents. Glycerol kinase 1 (GK1) is phosphotransferase encoded on the X chromosome, is associated with the synthesis of triglycerides and glycerophospholipids, and has been mainly studied for X-linked metabolic disorder GK deficiency (GKD). Interestingly, our DNA microarray analysis showed that several anti-cancer agents highly induced the expression of GK1, especially GK1a and GK1b isoforms, in human prostate cancer PC-3 cells. To elucidate the relationship between GK1 and cancer cell death, a human GK1b-specific expression vector was constructed and transfected into the PC-3 cells. Surprisingly, GK1b overexpression dramatically reduced cell viability and significantly accelerated apoptotic cell death. These findings suggest that GK1b may serve as a promising modulator and biomarker of cell death in prostate cancer, offering potential avenues for therapeutic intervention.
    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
    p53是脊椎动物中的关键肿瘤抑制因子,在人类癌症中经常发生突变。大多数突变是错义突变,使p53在抑制肿瘤的发生和进展方面无活性。开发小分子药物将突变型p53转化为活性,野生型样构象是个性化癌症治疗的重要焦点.先前的研究表明,在动物模型中,重新激活p53会抑制癌细胞增殖和肿瘤生长。具有酪氨酸220取代的选择性靶向p53突变体的化合物的早期临床证据表明在患者中再激活p53的潜在治疗益处。这项研究鉴定并检查了UCI-1001化合物系列作为几种p53突变的潜在校正剂。研究结果表明,p53突变癌细胞系中的UCI-1001处理抑制生长并恢复野生型p53活性,包括DNA结合,靶基因激活,和诱导细胞死亡。细胞热转移测定,构象特异性免疫荧光染色,和差示扫描荧光法表明UCI-1001与癌细胞中突变型p53相互作用并改变其构象。这些初步结果鉴定了UCI-1001系列的嘧啶三酮衍生物作为p53校正药物开发的候选物。
    p53 is a crucial tumor suppressor in vertebrates that is frequently mutated in human cancers. Most mutations are missense mutations that render p53 inactive in suppressing tumor initiation and progression. Developing small-molecule drugs to convert mutant p53 into an active, wild-type-like conformation is a significant focus for personalized cancer therapy. Prior research indicates that reactivating p53 suppresses cancer cell proliferation and tumor growth in animal models. Early clinical evidence with a compound selectively targeting p53 mutants with substitutions of tyrosine 220 suggests potential therapeutic benefits of reactivating p53 in patients. This study identifies and examines the UCI-1001 compound series as a potential corrector for several p53 mutations. The findings indicate that UCI-1001 treatment in p53 mutant cancer cell lines inhibits growth and reinstates wild-type p53 activities, including DNA binding, target gene activation, and induction of cell death. Cellular thermal shift assays, conformation-specific immunofluorescence staining, and differential scanning fluorometry suggest that UCI-1001 interacts with and alters the conformation of mutant p53 in cancer cells. These initial results identify pyrimidine trione derivatives of the UCI-1001 series as candidates for p53 corrector drug development.
    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
    磷酸肌醇3激酶(PI3K)负责磷酸化磷酸肌醇,以产生对调节各种细胞过程至关重要的次级信号分子,包括细胞生长,生存,和新陈代谢。PI3K是包含催化亚基(p110α,p110β,或p110δ)和调节亚基(p85)。调节亚基的结合,P85,与催化亚基,p110形成PI3K酶的组成部分。PIK3R1(磷酸肌醇-3-激酶调节亚基1)属于PI3K家族的IA类。PIK3R1由于选择性剪接而表现出结构复杂性,产生不同的同工型,突出的是p85α和p55α。虽然主要的p85α同种型包含多个结构域,包括Src同源3(SH3)域,断点簇区域同源性(BH)域,和Src同源2(SH2)域(iSH2和nSH2),较短的同工型,p55α,缺少p85α中存在的某些结构域。在这次审查中,我们将重点介绍控制PI3K信号传导的复杂调节机制,以及PIK3R1改变对细胞过程的影响。我们将进一步深入研究PIK3R1突变在各种癌症类型中的临床意义及其对预后和治疗结果的影响。此外,我们将讨论旨在调节PI3K相关通路的靶向治疗的演变前景.总的来说,这篇综述将提供对PIK3R1在癌症中的动态相互作用的见解,促进精准医学的进步和有针对性的干预措施的发展。
    Phosphoinositide 3-kinase (PI3K) is responsible for phosphorylating phosphoinositides to generate secondary signaling molecules crucial for regulating various cellular processes, including cell growth, survival, and metabolism. The PI3K is a heterodimeric enzyme complex comprising of a catalytic subunit (p110α, p110β, or p110δ) and a regulatory subunit (p85). The binding of the regulatory subunit, p85, with the catalytic subunit, p110, forms an integral component of the PI3K enzyme. PIK3R1 (phosphoinositide-3-kinase regulatory subunit 1) belongs to class IA of the PI3K family. PIK3R1 exhibits structural complexity due to alternative splicing, giving rise to distinct isoforms, prominently p85α and p55α. While the primary p85α isoform comprises multiple domains, including Src homology 3 (SH3) domains, a Breakpoint Cluster Region Homology (BH) domain, and Src homology 2 (SH2) domains (iSH2 and nSH2), the shorter isoform, p55α, lacks certain domains present in p85α. In this review, we will highlight the intricate regulatory mechanisms governing PI3K signaling along with the impact of PIK3R1 alterations on cellular processes. We will further delve into the clinical significance of PIK3R1 mutations in various cancer types and their implications for prognosis and treatment outcomes. Additionally, we will discuss the evolving landscape of targeted therapies aimed at modulating PI3K-associated pathways. Overall, this review will provide insights into the dynamic interplay of PIK3R1 in cancer, fostering advancements in precision medicine and the development of targeted interventions.
    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
    目的:探索LTBP4的表达,乳腺癌(BC)的预后意义和分子作用机制。方法:在组学数据集和实验的基础上,我们对BC中的LTBP4进行了综合分析。结果与结论:LTBP4在BC中下调,启动子甲基化高,遗传改变低。DNA甲基化与LTBP4mRNA表达呈负相关。较高的LTBP4与更好的生存率相关。LTBP4在细胞外基质受体相互作用中富集,细胞粘附分子,细胞周期和MAPK通路。LTBP4表达和甲基化与肿瘤浸润免疫细胞呈正相关和负相关,分别。总之,LTBP4是一种假定的BC肿瘤抑制因子,其表达受DNA甲基化调控,与预后有关。
    [方框:见正文]。
    Aim: To explore the LTBP4\'s expression, prognostic significance and molecular mechanism of action in breast cancer (BC).Methods: On the basis of omics datasets and experiments, we conducted a synthetical analysis of LTBP4 in BC.Results & conclusion: LTBP4 was downregulated in BC with high promoter methylation and low genetic alteration. DNA methylation was negatively associated with LTBP4 mRNA expression. Higher LTBP4 associated with better survival. LTBP4 was enrichment in extracellular matrix receptor interactions, cell adhesion molecules, cell cycle and MAPK pathway. LTBP4 expression and methylation were positively and negatively associated with tumor infiltrating immune cells, respectively. In conclusion, LTBP4 is a putative tumor suppressor in BC, which expression is regulated by DNA methylation and relates with prognosis.
    [Box: see text].
    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
    去泛素酶靶向嵌合体(DUBTAC)是一种有前途的诱导靶向蛋白稳定(TPS)的技术。尽管具有治疗潜力,到目前为止,很少有蛋白质被DUBTAC稳定。该技术的有限适用性可能是由于适度的DUBTAC诱导的蛋白质稳定作用,以及缺乏可用于DUBTAC开发的有效去泛素酶配体。这里,我们报告了MS7829和MS8588的发现,这是cGAS的一流DUBTAC,cGAS-STING途径的关键组成部分。虽然这些DUBTAC是基于cGAS抑制剂,他们有效地稳定cGAS和激活cGAS/STING/IRF3信号。为了开发这些CGASDUBTAC,我们优化了EN523,一种OTUB1共价配体,变成一种改进的配体,MS5105.我们通过生成基于MS5105的CFTRDUBTAC来验证MS5105,在稳定ΔF508-CFTR突变蛋白方面比先前报道的基于EN523的CFTRDUBTAC有效约10倍。总的来说,这项工作推进了TPS的DUBTAC技术。
    Deubiquitinase-targeting chimera (DUBTAC) is a promising technology for inducing targeted protein stabilization (TPS). Despite its therapeutic potential, very few proteins have been stabilized by DUBTACs to date. The limited applicability of this technology is likely due to the modest DUBTAC-induced protein stabilization effect, and the scarcity of effective deubiquitinase ligands that can be harnessed for DUBTAC development. Here, we report the discovery of MS7829 and MS8588, the first-in-class DUBTACs of cGAS, a key component of the cGAS-STING pathway. While these DUBTACs are based on a cGAS inhibitor, they effectively stabilized cGAS and activated the cGAS/STING/IRF3 signaling. To develop these cGAS DUBTACs, we optimized EN523, an OTUB1 covalent ligand, into an improved ligand, MS5105. We validated MS5105 by generating a MS5105-based CFTR DUBTAC, which was approximately 10-fold more effective in stabilizing the ΔF508-CFTR mutant protein than the previously reported EN523-based CFTR DUBTAC. Overall, this work advances the DUBTAC technology for TPS.
    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
    微卫星不稳定性高(MSI-H)肿瘤是恶性肿瘤,尽管有很高的突变负担,经常有完整的TP53。MSI-H肿瘤中最常见的突变之一是核糖体蛋白RPL22中的移码突变。这里,我们通过外显子6中的可变剪接开关将RPL22鉴定为MDM4剪接的调制器。RPL22缺失增加MDM4外显子6包涵体和细胞增殖,并增加对MDM抑制剂Nutlin-3a的抗性。RPL22抑制其旁系物的表达,RPL22L1,通过介导对应于截短转录物的隐蔽外显子的剪接。因此,RPL22中的破坏性突变驱动致癌MDM4诱导,并揭示MSI-H肿瘤中常见的剪接回路,这可能为MDM4-p53轴和致癌RPL22L1诱导的治疗靶向提供信息。
    Microsatellite instability-high (MSI-H) tumors are malignant tumors that, despite harboring a high mutational burden, often have intact TP53. One of the most frequent mutations in MSI-H tumors is a frameshift mutation in RPL22, a ribosomal protein. Here, we identified RPL22 as a modulator of MDM4 splicing through an alternative splicing switch in exon 6. RPL22 loss increases MDM4 exon 6 inclusion and cell proliferation and augments resistance to the MDM inhibitor Nutlin-3a. RPL22 represses the expression of its paralog, RPL22L1, by mediating the splicing of a cryptic exon corresponding to a truncated transcript. Therefore, damaging mutations in RPL22 drive oncogenic MDM4 induction and reveal a common splicing circuit in MSI-H tumors that may inform therapeutic targeting of the MDM4-p53 axis and oncogenic RPL22L1 induction.
    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:虽然黑色素瘤通常损失9p21,CDKN2A位于其上,在该位点存在其他肿瘤抑制因子的特征不完全。在这里,我们评估了microRNA-876-3p(miR-876)的表达水平和功能作用,其基因也映射到9p21。
    方法:使用定量miRNA逆转录酶聚合酶链反应(qRT-PCR)在人组织和细胞系中评估miR-876的表达。在癌症基因组图谱(TCGA)黑素瘤队列中确定MIR876拷贝数。miR-876表达的调控对黑色素瘤细胞集落形成的影响进行了评估,迁移,入侵,凋亡,细胞周期进程,和培养中的药物敏感性,以及异种移植模型中的体内肿瘤生长。使用RNA测序(RNA-Seq)测定由miR-876过表达诱导的全基因组转录组变化。
    结果:与痣相比,miR-876在原发性黑色素瘤样本中的表达显着降低,与人类黑素细胞相比,在人类黑素瘤细胞系中。对TCGA队列的分析显示MIR876在>50%的黑素瘤中缺失。miR-876过表达导致黑素瘤细胞集落形成减少,迁移,和入侵,伴随着细胞周期停滞和细胞凋亡增加。肿瘤内注射miR-876显著抑制体内黑色素瘤生长。miR-876处理的肿瘤的RNA-Seq分析揭示了几种生长促进基因的下调,随着肿瘤抑制基因的上调,通过qRT-PCR分析证实。计算分析将MAPK1(或ERK2)鉴定为miR-876作用的可能靶标。miR-876的过表达显著抑制了由MAPK1/ERK23'UTR驱动的荧光素酶表达,并导致黑色素瘤细胞中ERK蛋白表达降低。MAPK1/ERK2cDNA过表达拯救了miR-876对黑色素瘤集落形成的影响。miR-876过表达使黑色素瘤细胞对BRAF抑制剂vemurafenib治疗敏感。
    结论:这些研究确定miR-876是9p21上的一种独特的肿瘤抑制因子,在黑色素瘤中失活,并提示miR-876丢失是激活ERK和促分裂原活化蛋白激酶(MAPK)途径的另一种机制。此外,他们提出了miR-876过表达与BRAF抑制相结合作为黑色素瘤合理治疗策略的治疗潜力.
    BACKGROUND: While melanomas commonly harbor losses of 9p21, on which CDKN2A resides, the presence of additional tumor suppressor elements at this locus is incompletely characterized. Here we assess the expression levels and functional role of microRNA-876-3p (miR-876), whose gene also maps to 9p21.
    METHODS: Expression of miR-876 was assessed in human tissues and cell lines using quantitative miRNA reverse transcriptase polymerase chain reaction (qRT-PCR). MIR876 copy number was determined in The Cancer Genome Atlas (TCGA) melanoma cohort. The consequences of regulation of miR-876 expression were assessed on melanoma cell colony formation, migration, invasion, apoptosis, cell cycle progression, and drug sensitivity in culture, and on in vivo tumor growth in a xenograft model. Genome-wide transcriptomic changes induced by miR-876 overexpression were determined using RNA sequencing (RNA-Seq).
    RESULTS: miR-876 expression was significantly decreased in primary melanoma samples when compared with nevi, and in human melanoma cell lines when compared with human melanocytes. Analysis of the TCGA cohort revealed deletions in MIR876 in > 50% of melanomas. miR-876 overexpression resulted in decreased melanoma cell colony formation, migration, and invasion, which was accompanied by cell cycle arrest and increased apoptosis. Intra-tumoral injections of miR-876 significantly suppressed melanoma growth in vivo. RNA-Seq analysis of miR-876-treated tumors revealed downregulation of several growth-promoting genes, along with upregulation of tumor suppressor genes, which was confirmed by qRT-PCR analysis. Computational analyses identified MAPK1 (or ERK2) as a possible target of miR-876 action. Overexpression of miR-876 significantly suppressed luciferase expression driven by the MAPK1/ERK2 3\' UTR, and resulted in decreased ERK protein expression in melanoma cells. MAPK1/ERK2 cDNA overexpression rescued the effects of miR-876 on melanoma colony formation. miR-876 overexpression sensitized melanoma cells to treatment with the BRAF inhibitor vemurafenib.
    CONCLUSIONS: These studies identify miR-876 as a distinct tumor suppressor on 9p21 that is inactivated in melanoma and suggest miR-876 loss as an additional mechanism to activate ERK and the mitogen activated protein kinase (MAPK) pathway in melanoma. In addition, they suggest the therapeutic potential of combining miR-876 overexpression with BRAF inhibition as a rational therapeutic strategy for melanoma.
    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
    抑癌基因在正常组织稳态中起关键作用,它们的失调是包括癌症在内的人类疾病的基础。除了人类遗传学,果蝇等模型生物在发现肿瘤抑制途径方面发挥了重要作用,这些途径随后被证明与人类癌症高度相关。在这里,我们展示了增生性椎间盘(Hyd),在果蝇中遗传分离的第一个肿瘤抑制基因之一,编码具有迄今未知底物的E3泛素连接酶,和线(林),以其在胚胎分割中的作用而闻名,定义一种强制性肿瘤抑制蛋白复合物(Hyd-Lin),靶向含锌指的癌蛋白Bowl进行泛素介导的降解,Lin充当底物适配器,将Bowl招募到Hyd进行泛素化。有趣的是,Hyd-Lin复合物的活性被另一个锌指基因编码的微肽直接抑制,鼓槌(drm),通过将Bowl从Hyd-Lin建筑群中移出来充当伪底物,从而稳定碗。我们通过直接抑制微肽drm的转录,进一步确定了表观遗传调节因子Polycomb抑制复合物1(PRC1)作为Hyd-Lin-Bowl途径的关键上游调节因子。我们表明Hyd的遗传失活,林,或PRC1导致体内依赖碗的增生组织过度生长。我们还提供了Hyd的哺乳动物同源物(UBR5,已知在各种人类癌症中反复失调)的证据,Lin(LINS1),和碗(OSR1/2)构成了人类细胞中类似的蛋白质降解途径,OSR2促进前列腺癌的发生。总之,这些发现定义了一个以前未被识别的肿瘤抑制途径,该途径将表观遗传程序与组织生长控制和肿瘤发生过程中调节的蛋白质降解联系起来.
    Tumor suppressor genes play critical roles in normal tissue homeostasis, and their dysregulation underlies human diseases including cancer. Besides human genetics, model organisms such as Drosophila have been instrumental in discovering tumor suppressor pathways that were subsequently shown to be highly relevant in human cancer. Here we show that hyperplastic disc (Hyd), one of the first tumor suppressors isolated genetically in Drosophila and encoding an E3 ubiquitin ligase with hitherto unknown substrates, and Lines (Lin), best known for its role in embryonic segmentation, define an obligatory tumor suppressor protein complex (Hyd-Lin) that targets the zinc finger-containing oncoprotein Bowl for ubiquitin-mediated degradation, with Lin functioning as a substrate adaptor to recruit Bowl to Hyd for ubiquitination. Interestingly, the activity of the Hyd-Lin complex is directly inhibited by a micropeptide encoded by another zinc finger gene, drumstick (drm), which functions as a pseudosubstrate by displacing Bowl from the Hyd-Lin complex, thus stabilizing Bowl. We further identify the epigenetic regulator Polycomb repressive complex1 (PRC1) as a critical upstream regulator of the Hyd-Lin-Bowl pathway by directly repressing the transcription of the micropeptide drm Consistent with these molecular studies, we show that genetic inactivation of Hyd, Lin, or PRC1 resulted in Bowl-dependent hyperplastic tissue overgrowth in vivo. We also provide evidence that the mammalian homologs of Hyd (UBR5, known to be recurrently dysregulated in various human cancers), Lin (LINS1), and Bowl (OSR1/2) constitute an analogous protein degradation pathway in human cells, and that OSR2 promotes prostate cancer tumorigenesis. Altogether, these findings define a previously unrecognized tumor suppressor pathway that links epigenetic program to regulated protein degradation in tissue growth control and tumorigenesis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号