RUVBL2

RUVBL2
  • 文章类型: Journal Article
    雷帕霉素的机制靶点(mTOR)是细胞生长和代谢的主要调节因子,整合环境信号来调节合成代谢和分解代谢过程,调节脂质合成,生长因子诱导的细胞增殖,细胞存活,和移民。这些活动是作为两个不同复合物的一部分进行的,mTORC1和mTORC2,每个都有特定的角色。mTORC1和mTORC2是由mTOR与特定配偶体相互作用形成的精细二聚体结构。mTOR仅作为这些大型复合物的一部分,但是它们的组装和激活需要一个专用和复杂的陪伴系统。mTOR折叠和组装与TELO2-TTI1-TTI2(TTT)复合物暂时分离,协助mTOR共翻译折叠成天然构象。然后将成熟的mTOR转移至R2TP复合物以组装活性mTORC1和mTORC2复合物。R2TP与HSP90伴侣一起工作,以促进mTOR中其他亚基的掺入和二聚化。这篇综述总结了我们目前关于HSP90-R2TP-TTT伴侣系统如何促进活性mTORC1和mTORC2复合物的成熟和组装的知识,讨论互动,结构,和机制。
    The mechanistic target of rapamycin (mTOR) is a master regulator of cell growth and metabolism, integrating environmental signals to regulate anabolic and catabolic processes, regulating lipid synthesis, growth factor-induced cell proliferation, cell survival, and migration. These activities are performed as part of two distinct complexes, mTORC1 and mTORC2, each with specific roles. mTORC1 and mTORC2 are elaborated dimeric structures formed by the interaction of mTOR with specific partners. mTOR functions only as part of these large complexes, but their assembly and activation require a dedicated and sophisticated chaperone system. mTOR folding and assembly are temporarily separated with the TELO2-TTI1-TTI2 (TTT) complex assisting the cotranslational folding of mTOR into a native conformation. Matured mTOR is then transferred to the R2TP complex for assembly of active mTORC1 and mTORC2 complexes. R2TP works in concert with the HSP90 chaperone to promote the incorporation of additional subunits to mTOR and dimerization. This review summarizes our current knowledge on how the HSP90-R2TP-TTT chaperone system facilitates the maturation and assembly of active mTORC1 and mTORC2 complexes, discussing interactions, structures, and mechanisms.
    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
    人腺病毒(HAdV)是通常引起轻度疾病的小DNA病毒。某些菌株,特别是那些属于BHAdV的物种,可引起严重的肺炎,死亡率相对较高。关于这些高致病性物种如何影响受感染细胞以及它们如何抑制先天免疫的分子方面知之甚少。本研究提供了有关物种B腺病毒如何抑制干扰素信号通路的分子见解。我们的研究表明,这些病毒,与HAdV-C2不同,对I型干扰素具有抗性。这种抗性可能是由于干扰素刺激的基因表达的高效抑制而产生的。与HAdV-C2不同,HAdV-B7和B14从感染细胞中干扰素刺激的基因启动子中隔离了STAT2和RNA聚合酶II。这导致抑制的干扰素刺激的基因激活。此外,我们显示RuvBL1和RuvBL2是RNA聚合酶II募集到启动子和干扰素刺激的基因激活的重要辅因子,被重定向到细胞质形成高分子量复合物,很可能,无法与染色质联系。蛋白质组学分析还确定了这些病毒影响宿主细胞的方式的关键差异,提供对物种B相关高致病性的见解。奇怪的是,我们观察到感染细胞的蛋白质表达水平发生变化,HAdV-C2和B7比同一物种更相似,B7和B14。总的来说,我们的研究代表了高致病性HAdV-B7和B14对先天免疫抑制的首次此类研究,为未来的研究奠定了重要的基础.重要人类腺病毒形成了双链DNA病毒的大家族,已知有多种通常轻微的疾病。某些人腺病毒菌株引起严重肺炎,导致比大多数其他菌株高得多的死亡率和发病率。这种致病性增强的原因是未知的。我们的研究提供了这些高致病性菌株如何使干扰素信号通路失活的分子研究。强调这些病毒对I型干扰素缺乏敏感性,同时提供了细胞蛋白中的病毒变化如何导致更糟糕的疾病结局的全球图景。
    Human adenoviruses (HAdVs) are small DNA viruses that generally cause mild disease. Certain strains, particularly those belonging to species B HAdVs, can cause severe pneumonia and have a relatively high mortality rate. Little is known about the molecular aspects of how these highly pathogenic species affect the infected cell and how they suppress innate immunity. The present study provides molecular insights into how species B adenoviruses suppress the interferon signaling pathway. Our study shows that these viruses, unlike HAdV-C2, are resistant to type I interferon. This resistance likely arises due to the highly efficient suppression of interferon-stimulated gene expression. Unlike in HAdV-C2, HAdV-B7 and B14 sequester STAT2 and RNA polymerase II from interferon-stimulated gene promoters in infected cells. This results in suppressed interferon- stimulated gene activation. In addition, we show that RuvBL1 and RuvBL2, cofactors important for RNA polymerase II recruitment to promoters and interferon-stimulated gene activation, are redirected to the cytoplasm forming high molecular weight complexes that, likely, are unable to associate with chromatin. Proteomic analysis also identified key differences in the way these viruses affect the host cell, providing insights into species B-associated high pathogenicity. Curiously, we observed that at the level of protein expression changes to the infected cell, HAdV-C2 and B7 were more similar than those of the same species, B7 and B14. Collectively, our study represents the first such study of innate immune suppression by the highly pathogenic HAdV-B7 and B14, laying an important foundation for future investigations.IMPORTANCEHuman adenoviruses form a large family of double-stranded DNA viruses known for a variety of usually mild diseases. Certain strains of human adenovirus cause severe pneumonia leading to much higher mortality and morbidity than most other strains. The reasons for this enhanced pathogenicity are unknown. Our study provides a molecular investigation of how these highly pathogenic strains might inactivate the interferon signaling pathway, highlighting the lack of sensitivity of these viruses to type I interferon in general while providing a global picture of how viral changes in cellular proteins drive worse disease outcomes.
    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
    大分子复合物在各种细胞过程中起着重要作用。细胞内大分子组装体的组装必须克服由拥挤的细胞环境施加的障碍,其特征在于生物大分子的估计浓度为100-450g/L,其占据约5-40%的细胞质体积。大分子组装体的形成通过分子伴侣与它们的共伴侣协作来促进。R2TP蛋白复合物已成为Hsp90的共同伴侣,在大分子组装中起重要作用。R2TP复合物由RPAP3:P1H1DI的异二聚体组成,该二聚体又与与多种细胞活动(AAA)相关的ATPase成员复合。RUVBL1和RUVBL2(R1和R2)家族。使R2TP共同伴侣复合体特别重要的是,它涉及多种细胞过程,包括基因表达,翻译,协同平移复杂装配,和翻译后蛋白质复合物的形成。R2TP共同伴侣复合物的功能多功能性使其成为细胞发育的核心;因此,它与各种人类疾病有关。此外,它们在传染病病原体发展中的作用已经引起了人们的兴趣。在当前的审查中,我们讨论了这些蛋白质作为共同伴侣调节Hsp90的作用及其与Hsp70的伙伴关系。此外,我们强调了R2TP复合物中单个蛋白质的结构-功能特征,并描述了它们在各种细胞过程中的作用。
    Macromolecular complexes play essential roles in various cellular processes. The assembly of macromolecular assemblies within the cell must overcome barriers imposed by a crowded cellular environment which is characterized by an estimated concentration of biological macromolecules amounting to 100-450 g/L that take up approximately 5-40% of the cytoplasmic volume. The formation of the macromolecular assemblies is facilitated by molecular chaperones in cooperation with their co-chaperones. The R2TP protein complex has emerged as a co-chaperone of Hsp90 that plays an important role in macromolecular assembly. The R2TP complex is composed of a heterodimer of RPAP3:P1H1DI that is in turn complexed to members of the ATPase associated with diverse cellular activities (AAA +), RUVBL1 and RUVBL2 (R1 and R2) families. What makes the R2TP co-chaperone complex particularly important is that it is involved in a wide variety of cellular processes including gene expression, translation, co-translational complex assembly, and posttranslational protein complex formation. The functional versatility of the R2TP co-chaperone complex makes it central to cellular development; hence, it is implicated in various human diseases. In addition, their roles in the development of infectious disease agents has become of interest. In the current review, we discuss the roles of these proteins as co-chaperones regulating Hsp90 and its partnership with Hsp70. Furthermore, we highlight the structure-function features of the individual proteins within the R2TP complex and describe their roles in various cellular processes.
    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
    DPCD是一种可能在纤毛形成中起作用的蛋白质,其缺乏会导致原发性纤毛运动障碍(PCD)。一种由纤毛细胞受损引起的罕见疾病。除了高通量研究将DPCD确定为可能的RUVBL1(R1)和RUVBL2(R2)伴侣外,没有深入的细胞,生物化学,到目前为止,已经报道了涉及DPCD的结构调查。R1和R2蛋白是普遍存在的高度保守的AAA+家族ATP酶,它们组装和成熟大量的大分子复合物,在许多细胞过程中至关重要。特别是通过保证R2TP或R2TP类机械内的共同陪伴功能。在本研究中,我们在体内鉴定DPCD为新的R1R2伴侣。我们表明DPCD在体外和细胞中直接与R1和R2相互作用。我们表征了DPCD在溶液中的物理化学性质,并建立了DPCD的3D模型。此外,我们使用了各种正交生物物理技术,包括小角度X射线散射,结构质谱和电子显微镜来评估DPCD与R1R2相互作用的分子决定因素。有趣的是,DPCD在结合时破坏R1R2复合物的十二聚体状态,并且这种相互作用主要通过R1R2的DII结构域发生。
    DPCD is a protein that may play a role in cilia formation and whose absence leads to primary ciliary dyskinesia (PCD), a rare disease caused by impairment of ciliated cells. Except for high-throughput studies that identified DPCD as a possible RUVBL1 (R1) and RUVBL2 (R2) partner, no in-depth cellular, biochemical, and structural investigation involving DPCD have been reported so far. R1 and R2 proteins are ubiquitous highly conserved AAA + family ATPases that assemble and mature a plethora of macromolecular complexes and are pivotal in numerous cellular processes, especially by guaranteeing a co-chaperoning function within R2TP or R2TP-like machineries. In the present study, we identified DPCD as a new R1R2 partner in vivo. We show that DPCD interacts directly with R1 and R2 in vitro and in cells. We characterized the physico-chemical properties of DPCD in solution and built a 3D model of DPCD. In addition, we used a variety of orthogonal biophysical techniques including small-angle X-ray scattering, structural mass spectrometry and electron microscopy to assess the molecular determinants of DPCD interaction with R1R2. Interestingly, DPCD disrupts the dodecameric state of R1R2 complex upon binding and this interaction occurs mainly via the DII domains of R1R2.
    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
    NFAT5是唯一已知的哺乳动物张力响应转录因子,在细胞适应高渗应激中起重要作用。它还涉及各种生理和病理过程。NFAT5活性受到细胞外张力的严格调节,但是潜在的机制仍然难以捉摸。这里,我们证明NFAT5通过核孔复合物进入细胞核。我们发现NFAT5利用独特的核定位信号(NFAT5-NLS)进行核导入。siRNA筛选显示,只有核蛋白β1(KPNB1),但不是核动力蛋白α,通过与NFAT5-NLS的直接相互作用负责NFAT5的核导入。蛋白质组学分析和siRNA筛选进一步揭示,低渗下NFAT5的核输出是由exportin-T(XPOT)驱动的,其中该过程需要RuvB样AAA型ATPase2(RUVBL2)作为不可或缺的伴侣。我们的发现已经确定了NFAT5的非常规张力依赖性核质运输途径,这是协调细胞快速适应细胞外张力变化的关键步骤。这些发现为开发新的NFAT5靶向策略提供了机会,这些策略可能用于治疗与NFAT5失调相关的疾病。
    NFAT5 is the only known mammalian tonicity-responsive transcription factor with an essential role in cellular adaptation to hypertonic stress. It is also implicated in diverse physiological and pathological processes. NFAT5 activity is tightly regulated by extracellular tonicity, but the underlying mechanisms remain elusive. Here, we demonstrate that NFAT5 enters the nucleus via the nuclear pore complex. We found that NFAT5 utilizes a unique nuclear localization signal (NFAT5-NLS) for nuclear import. siRNA screening revealed that only karyopherin β1 (KPNB1), but not karyopherin α, is responsible for the nuclear import of NFAT5 via direct interaction with the NFAT5-NLS. Proteomics analysis and siRNA screening further revealed that nuclear export of NFAT5 under hypotonicity is driven by exportin-T (XPOT), where the process requires RuvB-like AAA-type ATPase 2 (RUVBL2) as an indispensable chaperone. Our findings have identified an unconventional tonicity-dependent nucleocytoplasmic trafficking pathway for NFAT5 that represents a critical step in orchestrating rapid cellular adaptation to change in extracellular tonicity. These findings offer an opportunity for the development of novel NFAT5 targeting strategies that are potentially useful for the treatment of diseases associated with NFAT5 dysregulation.
    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
    R2TP(RUVBL1-RUVBL2-RPAP3-PIH1D1)复合物,与热休克蛋白90(HSP90)合作,作为蛋白质复合物组装和稳定性的伴侣,包括RNA聚合酶,小核核糖核蛋白颗粒(snRNPs),和磷脂酰肌醇3-激酶(PI3K)样激酶(PIKK)如TOR和SMG1。PIKK稳定取决于TELO2、TTI1和TTI2(TTT)的额外复合物,对其结构和功能知之甚少。人R2TP-TTT复合物的低温电子显微镜(cryo-EM)结构,连同生化实验,揭示了TOR招募R2TP-TTT伴侣的机制。HEAT重复TTT复合物结合TOR的激酶结构域,在不阻止其活动的情况下,并将TOR传递给R2TP伴侣。此外,TTT通过抑制RUVBL1-RUVBL2ATP酶活性和通过调节R2TP的PIH1D1和RPAP3组分的构象和相互作用来调节R2TP分子伴侣。一起来看,我们的结果显示了TTT如何将TOR和R2TP的招募与该伴侣系统的调节相结合.
    The R2TP (RUVBL1-RUVBL2-RPAP3-PIH1D1) complex, in collaboration with heat shock protein 90 (HSP90), functions as a chaperone for the assembly and stability of protein complexes, including RNA polymerases, small nuclear ribonucleoprotein particles (snRNPs), and phosphatidylinositol 3-kinase (PI3K)-like kinases (PIKKs) such as TOR and SMG1. PIKK stabilization depends on an additional complex of TELO2, TTI1, and TTI2 (TTT), whose structure and function are poorly understood. The cryoelectron microscopy (cryo-EM) structure of the human R2TP-TTT complex, together with biochemical experiments, reveals the mechanism of TOR recruitment to the R2TP-TTT chaperone. The HEAT-repeat TTT complex binds the kinase domain of TOR, without blocking its activity, and delivers TOR to the R2TP chaperone. In addition, TTT regulates the R2TP chaperone by inhibiting RUVBL1-RUVBL2 ATPase activity and by modulating the conformation and interactions of the PIH1D1 and RPAP3 components of R2TP. Taken together, our results show how TTT couples the recruitment of TOR to R2TP with the regulation of this chaperone system.
    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
    甲型流感病毒(IAV)的NS1蛋白在病毒发病机制和宿主免疫应答中起重要作用。通过蛋白质组学方法,我们已经确定RuvB样蛋白1和2(RuvBL1和RuvBL2)是IAV的NS1蛋白的相互作用伴侣。用A/PR/8/34(PR8)病毒感染人肺A549细胞导致RuvBL2而不是RuvBL1的蛋白质水平降低。对RuvBL2的进一步研究表明,NS1-RuvBL2相互作用是不依赖RNA的,和RuvBL2结合NS1的RNA结合域。用野生型或delNS1PR8病毒感染干扰素(IFN)缺陷的Vero细胞可降低RuvBL2蛋白水平并诱导凋亡;与野生型病毒相比,delNS1病毒导致RuvBL2蛋白水平降低更多,并诱导更多的凋亡。通过siRNA敲除RuvBL2诱导的细胞凋亡和RuvBL2的过表达导致Vero细胞对感染诱导的细胞凋亡的抗性增加。这些结果表明,一个或多个非NS1病毒元件通过抑制RuvBL2蛋白水平诱导细胞凋亡,并且NS1通过在没有IFN影响的情况下维持感染细胞中的RuvBL2丰度来抑制非NS1病毒元件诱导的细胞凋亡。与Vero细胞相比,用PR8病毒感染IFN感受态A549细胞导致RuvBL2蛋白水平降低,但不诱导细胞凋亡.同时,用重组IFN预处理Vero细胞导致抵抗感染诱导的细胞凋亡。这些结果表明,感染引起的,受感染细胞中RuvBL2调节的凋亡被IFN存活信号抵消。我们的结果揭示了感染诱导的细胞凋亡的新机制,该机制可以由IAV感染的细胞中的NS1和I型IFN信号调节。
    The NS1 protein of influenza A virus (IAV) plays important roles in viral pathogenesis and host immune response. Through a proteomic approach, we have identified RuvB-like proteins 1 and 2 (RuvBL1 and RuvBL2) as interacting partners of the NS1 protein of IAVs. Infection of human lung A549 cells with A/PR/8/34 (PR8) virus resulted in reductions in the protein levels of RuvBL2 but not RuvBL1. Further studies with RuvBL2 demonstrated that the NS1-RuvBL2 interaction is RNA-independent, and RuvBL2 binds the RNA-binding domain of the NS1. Infection of interferon (IFN)-deficient Vero cells with wild-type or delNS1 PR8 virus reduced RuvBL2 protein levels and induced apoptosis; delNS1 virus caused more reductions in RuvBL2 protein levels and induced more apoptosis than did wild-type virus. Knockdown of RuvBL2 by siRNAs induced apoptosis and overexpression of RuvBL2 resulted in increased resistance to infection-induced apoptosis in Vero cells. These results suggest that a non-NS1 viral element or elements induce apoptosis by suppressing RuvBL2 protein levels, and the NS1 inhibits the non-NS1 viral element-induced apoptosis by maintaining RuvBL2 abundance in infected cells in the absence of IFN influence. In contrast to Vero cells, infection of IFN-competent A549 cells with PR8 virus caused reductions in RuvBL2 protein levels but did not induce apoptosis. Concomitantly, pretreatment of Vero cells with a recombinant IFN resulted in resistance to infection-induced apoptosis. These results demonstrate that the infection-induced, RuvBL2-regulated apoptosis in infected cells is counterbalanced by IFN survival signals. Our results reveal a novel mechanism underlying the infection-induced apoptosis that can be modulated by the NS1 and type I IFN signaling in IAV-infected 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
    先前的研究表明,Reptin在肝细胞癌中过表达,并且对于体外增殖和细胞存活是必需的。然而,其在体内的病理生理作用尚不清楚。我们旨在使用肝脏Reptin敲除模型(ReptinLKO)研究Reptin在再生后肝细胞增殖中的作用。有趣的是,部分肝切除术后36小时,ReptinLKO小鼠的肝细胞增殖受到强烈损害,与细胞周期蛋白A表达和mTORC1和MAPK信号传导降低相关,导致肝脏再生受损。此外,在ReptinLKO模型中,我们观察到与非典型肝再生相关的Reptin无效逐渐消失.肥大和增殖的肝细胞逐渐取代ReptinKO肥大的肝细胞。最后,我们的结果表明,Reptin是体内肝细胞增殖和肝脏再生所必需的,并且在肝细胞存活和肝脏稳态中起着至关重要的作用.
    Previous studies have shown that Reptin is overexpressed in hepatocellular carcinoma and that it is necessary for in vitro proliferation and cell survival. However, its pathophysiological role in vivo remains unknown. We aimed to study the role of Reptin in hepatocyte proliferation after regeneration using a liver Reptin knock-out model (ReptinLKO ). Interestingly, hepatocyte proliferation is strongly impaired in ReptinLKO mice 36 h after partial hepatectomy, associated with a decrease of cyclin-A expression and mTORC1 and MAPK signalling, leading to an impaired liver regeneration. Moreover, in the ReptinLKO model, we have observed a progressive loss of Reptin invalidation associated with an atypical liver regeneration. Hypertrophic and proliferative hepatocytes gradually replace ReptinKO hypotrophic hepatocytes. To conclude, our results show that Reptin is required for hepatocyte proliferation in vivo and liver regeneration and that it plays a crucial role in hepatocyte survival and liver homeostasis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    脂肪细胞分化是脂肪组织发育的重要组成部分,与肥胖和肥胖相关疾病密切相关。在这项研究中,我们发现PPARγ的表达,在3T3-L1细胞分化的第5-7天,RUVBL2和脂联素同时明显增加。PPARγ过表达或PPARγ激活剂促进脂联素运输和分泌,并在3T3-L1细胞成脂分化过程中上调RUVBL2表达以及AS160磷酸化。持续的RUVBL2过表达也增强了脂联素的聚合和分泌,相比之下,RUVBL2敲低降低脂联素分泌。Further,PPARγ显著增强RUVBL2启动子活性和转录。RUVBL2启动子对PPARγ结合位点的逐渐缺失和突变表明,位于-804/-781bp的PPARγ结合基序是RUVBL2启动子活性所需的必需成分。染色质免疫沉淀(ChIP)测定确定PPARγ可以直接与RUVBL2启动子DNA相互作用。一起来看,这些数据表明PPARγ促进表达,通过转录激活RUVBL2聚合和分泌脂联素,加速3T3-L1细胞分化。
    Adipocyte differentiation is an essential part of adipose tissue development, and is closely related to obesity and obesity-related diseases. In this study, we found that the expression of PPARγ, RUVBL2 and Adiponectin were concurrently obviously increased in the 5th-7th day of 3T3-L1 cell differentiation. PPARγ overexpression or the PPARγ activator facilitated Adiponectin trafficking and secretion and upregulated RUVBL2 expression as well as AS160 phosphorylation during adipogenic differentiation of 3T3-L1 cells. Consistently RUVBL2 overexpression also enhanced the polymerization and secretion of Adiponectin, in contrast, RUVBL2 knockdown reduced Adiponectin secretion. Further, PPARγ significantly enhanced RUVBL2 promoter activity and transcription. The progressive deletions and mutations of RUVBL2 promoter for PPARγ binding sites suggested that the PPARγ binding motif situated at -804/-781 bp is an essential component required for RUVBL2 promoter activity. Chromatin immunoprecipitation (ChIP) assays determined that PPARγ can directly interact with the RUVBL2 promoter DNA. Taken together, these data suggest that PPARγ promotes the expression, polymerization and secretion of Adiponectin by activating RUVBL2 transcriptionally, which accelerates 3T3-L1 cell differentiation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    在癌症基因组图谱(TCGA)研究中,在多种癌症中鉴定了组蛋白修饰酶和组蛋白变体的突变。然而,在确定表观遗传因素在头颈部鳞状细胞癌(HNSCC)中的作用方面几乎没有进展和理解。这里,我们报告了RUVBL1(TIP49a)的鉴定,TIP60组蛋白修饰复合物的一个组成部分,在HNSCC中被扩增和过表达。RUVBL1在将组蛋白变体H2AZ掺入染色质中发挥关键作用,从而调节参与分化的关键基因的转录。癌细胞增殖和侵袭。H2AZ也在HNSCC肿瘤中过表达,从而调节RUVBL1/H2AZ依赖性转录程序。包括TCGA和单细胞HNSCC数据在内的多个队列的患者数据分析表明RUVBL1过表达是不良预后标志物并预测不良存活。体外实验表明RUVBL1/H2AZ在HNSCC细胞中具有促增殖作用。RUVBL1与分化程序呈负相关,与致癌程序呈正相关,使其成为HNSCC患者肿瘤发生的关键因素和脆弱的治疗靶标。
    Mutations in histone modifying enzymes and histone variants were identified in multiple cancers in The Cancer Genome Atlas (TCGA) studies. However, very little progress and understanding has been made in identifying the contribution of epigenetic factors in head and neck squamous cell carcinoma (HNSCC). Here, we report the identification of RUVBL1 (TIP49a), a component of the TIP60 histone modifying complex as being amplified and overexpressed in HNSCC. RUVBL1 plays a key role in incorporating histone variant H2AZ in chromatin thereby regulating transcription of key genes involved in differentiation, cancer cell proliferation and invasion. H2AZ is also overexpressed in HNSCC tumors thereby regulating RUVBL1/H2AZ dependent transcriptional programs. Patient data analysis of multiple cohorts including TCGA and single cell HNSCC data indicated RUVBL1 overexpression as a poor prognostic marker and predicts poor survival. In vitro experiments indicate a pro-proliferative role for RUVBL1/H2AZ in HNSCC cells. RUVBL1 inversely correlates with differentiation program and positively correlates with oncogenic programs, making it a key contributor to tumorigenesis and a vulnerable therapeutic target in HNSCC patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号