E2F Transcription Factors

  • 文章类型: Journal Article
    E2F转录因子家族,其成员由E2F1-E2F8基因编码,在细胞周期中起着关键作用,凋亡,新陈代谢,stemness,转移,老化,血管生成,肿瘤促进或抑制,和其他生物过程。E2Fs的活性在多个水平上受到调控,翻译后修饰是一种重要的调控机制。有许多类型的翻译后修饰,其中磷酸化,乙酰化,甲基化,泛素化,SUMOylation,Neddylation,和聚(ADP-核糖基)是在E2F家族的背景下最常研究的。E2F家族蛋白的翻译后修饰调节其生物活性,稳定性,本地化,以及与其他生物分子的相互作用,影响细胞增殖,凋亡,DNA损伤,等。,从而在生理和病理过程中发挥作用。值得注意的是,这些修改并不总是单独起作用,而是形成一个互动的监管网络。目前,几种靶向翻译后修饰的药物正在研究或临床应用,其中蛋白水解靶向嵌合体和分子胶可以靶向E2Fs。本文旨在总结E2F家族成员的不同PTM在生理状态和癌症中的作用和调节机制,并简要讨论其临床意义和潜在的治疗用途。
    The E2F transcription factor family, whose members are encoded by the E2F1-E2F8 genes, plays pivotal roles in the cell cycle, apoptosis, metabolism, stemness, metastasis, aging, angiogenesis, tumor promotion or suppression, and other biological processes. The activity of E2Fs is regulated at multiple levels, with posttranslational modifications being an important regulatory mechanism. There are numerous types of posttranslational modifications, among which phosphorylation, acetylation, methylation, ubiquitination, SUMOylation, neddylation, and poly(ADP-ribosyl)ation are the most commonly studied in the context of the E2F family. Posttranslational modifications of E2F family proteins regulate their biological activity, stability, localization, and interactions with other biomolecules, affecting cell proliferation, apoptosis, DNA damage, etc., and thereby playing roles in physiological and pathological processes. Notably, these modifications do not always act alone but rather form an interactive regulatory network. Currently, several drugs targeting posttranslational modifications are being studied or clinically applied, in which the proteolysis-targeting chimera and molecular glue can target E2Fs. This review aims to summarize the roles and regulatory mechanisms of different PTMs of E2F family members in the physiological state and in cancer and to briefly discuss their clinical significance and potential therapeutic use.
    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
    受调控的细胞周期进程确保稳态并预防癌症。在增殖细胞中,E3泛素连接酶后酶促进复合物/环体(APC/C)避免了过早的S期进入,尽管降解抑制G1-S进展的APC/C底物尚不完全清楚。APC/C在退出细胞周期的停滞细胞中也很活跃,但目前尚不清楚APC/C是否维持所有类型的逮捕。这里,通过表达APC/C抑制剂,EMI1,我们表明APC/C活性对于防止由药理学细胞周期蛋白依赖性激酶4和6(CDK4/6)抑制(Palbociclib)阻止的细胞中的S期进入至关重要。因此,抑制细胞周期基因表达需要活性蛋白降解。抑制APC/C的快速而强大的阻止旁路的机制涉及CDK以非典型的顺序作用,以失活视网膜母细胞瘤介导的E2F抑制。使APC/C失活首先引起有丝分裂细胞周期蛋白B的积累,然后促进细胞周期蛋白A的表达。我们认为细胞周期蛋白A是维持阻滞的关键底物,因为抗APC/C细胞周期蛋白A,但不是细胞周期蛋白B,足以诱导S相进入。绕过CDK4/6抑制阻滞的细胞启动DNA复制,来源许可严重减少。同时积累S期许可抑制剂,如细胞周期蛋白A和geminin,使用G1许可激活器破坏了G1-S进展的正常顺序。因此,DNA合成和细胞增殖严重受损。我们的研究结果预测,EMI1表达升高的癌症将倾向于逃避CDK4/6的抑制,未获得许可的S期,并遭受增强的基因组不稳定性。
    Regulated cell cycle progression ensures homeostasis and prevents cancer. In proliferating cells, premature S phase entry is avoided by the E3 ubiquitin ligase anaphasepromoting complex/cyclosome (APC/C), although the APC/C substrates whose degradation restrains G1-S progression are not fully known. The APC/C is also active in arrested cells that exited the cell cycle, but it is not clear whether APC/C maintains all types of arrest. Here, by expressing the APC/C inhibitor, EMI1, we show that APC/C activity is essential to prevent S phase entry in cells arrested by pharmacological cyclin-dependent kinases 4 and 6 (CDK4/6) inhibition (Palbociclib). Thus, active protein degradation is required for arrest alongside repressed cell cycle gene expression. The mechanism of rapid and robust arrest bypass from inhibiting APC/C involves CDKs acting in an atypical order to inactivate retinoblastoma-mediated E2F repression. Inactivating APC/C first causes mitotic cyclin B accumulation which then promotes cyclin A expression. We propose that cyclin A is the key substrate for maintaining arrest because APC/C-resistant cyclin A, but not cyclin B, is sufficient to induce S phase entry. Cells bypassing arrest from CDK4/6 inhibition initiate DNA replication with severely reduced origin licensing. The simultaneous accumulation of S phase licensing inhibitors, such as cyclin A and geminin, with G1 licensing activators disrupts the normal order of G1-S progression. As a result, DNA synthesis and cell proliferation are profoundly impaired. Our findings predict that cancers with elevated EMI1 expression will tend to escape CDK4/6 inhibition into a premature, underlicensed S phase and suffer enhanced genome instability.
    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
    组织修复,免疫防御和癌症进展依赖于静止和增殖之间的重要细胞决定1,2。哺乳动物细胞通过触发正反馈机制增殖3,4。转录因子E2F激活细胞周期蛋白依赖性激酶2(CDK2),这又使E2F抑制剂蛋白视网膜母细胞瘤(Rb)磷酸化和失活。该作用进一步增加E2F活性以表达增殖所需的基因。鉴于正反馈会无意中放大小信号,了解细胞如何保持这种积极的反馈在检查仍然是一个难题。在这里,我们测量了单个细胞中的E2F和CDK2信号变化,发现正反馈机制仅在G1期后期参与。细胞在进行增殖之前在中等E2F活性的可逆状态下花费可变且通常延长的时间。该中间E2F活性与由CDK2或CDK4/CDK6介导的Rb中保守的T373残基的磷酸化量成比例。这种T373磷酸化的Rb仍然与染色质结合,但一旦Rb在许多位点过度磷酸化,就会与染色质分离。完全激活E2F。T373的优先初始磷酸化可以通过其相对较慢的去磷酸化速率来解释。一起,我们的研究确定了中间E2F激活的启动状态,即细胞感知外部和内部信号,并决定是否逆转并退出静止状态或触发启动细胞增殖的正反馈机制.
    Tissue repair, immune defence and cancer progression rely on a vital cellular decision between quiescence and proliferation1,2. Mammalian cells proliferate by triggering a positive feedback mechanism3,4. The transcription factor E2F activates cyclin-dependent kinase 2 (CDK2), which in turn phosphorylates and inactivates the E2F inhibitor protein retinoblastoma (Rb). This action further increases E2F activity to express genes needed for proliferation. Given that positive feedback can inadvertently amplify small signals, understanding how cells keep this positive feedback in check remains a puzzle. Here we measured E2F and CDK2 signal changes in single cells and found that the positive feedback mechanism engages only late in G1 phase. Cells spend variable and often extended times in a reversible state of intermediate E2F activity before committing to proliferate. This intermediate E2F activity is proportional to the amount of phosphorylation of a conserved T373 residue in Rb that is mediated by CDK2 or CDK4/CDK6. Such T373-phosphorylated Rb remains bound on chromatin but dissociates from it once Rb is hyperphosphorylated at many sites, which fully activates E2F. The preferential initial phosphorylation of T373 can be explained by its relatively slower rate of dephosphorylation. Together, our study identifies a primed state of intermediate E2F activation whereby cells sense external and internal signals and decide whether to reverse and exit to quiescence or trigger the positive feedback mechanism that initiates cell proliferation.
    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
    组蛋白脱乙酰酶(HDACs)在转录调控中起着至关重要的作用,并与各种疾病有关。包括癌症.它们参与组蛋白尾部去乙酰化,并与转录抑制规范相关。先前的研究表明,HDAC通过视网膜母细胞瘤(RB)蛋白或通过SIN3B的DREAM复合物募集到细胞周期基因启动子对于细胞周期停滞和退出期间G1/S和G2/M基因抑制至关重要。在这里,我们研究了梦想之间的相互作用,RB,SIN3蛋白,和HDAC在细胞周期基因抑制的背景下。在非增殖HCT116和C2C12细胞中,SIN3B的敲除并不全局地抑制细胞周期基因。SIN3A/B的缺失会适度上调HCT116细胞中的几个细胞周期基因,但与DREAM/RB无关。HDAC抑制在停滞的转化或非转化的细胞中不诱导RB/DREAM靶基因的普遍上调。我们的发现表明,E2F:RB和DREAM复合物可以抑制细胞周期基因,而不依赖于HDAC活性。
    Histone deacetylases (HDACs) play a crucial role in transcriptional regulation and are implicated in various diseases, including cancer. They are involved in histone tail deacetylation and canonically linked to transcriptional repression. Previous studies suggested that HDAC recruitment to cell-cycle gene promoters via the retinoblastoma (RB) protein or the DREAM complex through SIN3B is essential for G1/S and G2/M gene repression during cell-cycle arrest and exit. Here we investigate the interplay among DREAM, RB, SIN3 proteins, and HDACs in the context of cell-cycle gene repression. Knockout of SIN3B does not globally derepress cell-cycle genes in non-proliferating HCT116 and C2C12 cells. Loss of SIN3A/B moderately upregulates several cell-cycle genes in HCT116 cells but does so independently of DREAM/RB. HDAC inhibition does not induce general upregulation of RB/DREAM target genes in arrested transformed or non-transformed cells. Our findings suggest that E2F:RB and DREAM complexes can repress cell-cycle genes without relying on HDAC activity.
    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
    背景:E2F/DP(真核2转录因子/二聚化伴侣)家族蛋白在高等生物的细胞周期发育中起重要作用。E2F/DP家族基因仅在少数植物物种中被报道。然而,迄今为止,尚未报道对茄子E2F/DP基因家族进行全面的全基因组鉴定分析。
    结果:本研究鉴定了8个非冗余的SlE2F/DP基因,这些基因在系统发育分析中被分为7组。所有八个基因具有单个E2F-TDP结构域,并且很少基因具有额外的结构域。在番茄中观察到两个片段重复基因对,除了顺式调控因素,miRNA的靶位点和磷酸化位点在番茄的植物发育和胁迫响应中起着重要的作用。为了探索SlE2F/DP蛋白的三维(3D)模型和基因本体(GO)注释,我们指出了它们推定的转运蛋白活性及其与几种推定配体的相互作用。SlE2F/DP-GFP融合蛋白在细胞核和内质网中的定位表明它们可能在其他生物学功能中起作用。表达研究揭示了大多数SlE2F/DP基因在各种器官中的差异表达模式。此外,E2F/DP基因对非生物胁迫的表达,特别是SlE2F/DP2和/或SlE2F/DP7,响应于热量而上调,盐,冷和ABA处理。此外,SlE2F/DP基因与多个代谢途径共表达分析与防御基因共表达,转录因子等,表明了它们在各种生物过程中的关键作用。
    结论:总体而言,我们的发现为了解SlE2F/DP基因的结构和功能提供了一种方法;通过标记辅助选择或转基因方法可能有助于提高果实的发育和对非生物胁迫的耐受性。
    BACKGROUND: E2F/DP (Eukaryotic 2 transcription factor/dimerization partner) family proteins play an essential function in the cell cycle development of higher organisms. E2F/DP family genes have been reported only in a few plant species. However, comprehensive genome-wide characterization analysis of the E2F/DP gene family of Solanum lycopersicum has not been reported so far.
    RESULTS: This study identified eight nonredundant SlE2F/DP genes that were classified into seven groups in the phylogenetic analysis. All eight genes had a single E2F-TDP domain and few genes had additional domains. Two segmental duplication gene pairs were observed within tomato, in addition to cis-regulatory elements, miRNA target sites and phosphorylation sites which play an important role in plant development and stress response in tomato. To explore the three-dimensional (3D) models and gene ontology (GO) annotations of SlE2F/DP proteins, we pointed to their putative transporter activity and their interaction with several putative ligands. The localization of SlE2F/DP-GFP fused proteins in the nucleus and endoplasmic reticulum suggested that they may act in other biological functions. Expression studies revealed the differential expression pattern of most of the SlE2F/DP genes in various organs. Moreover, the expression of E2F/DP genes against abiotic stress, particularly SlE2F/DP2 and/or SlE2F/DP7, was upregulated in response to heat, salt, cold and ABA treatment. Furthermore, the co-expression analysis of SlE2F/DP genes with multiple metabolic pathways was co-expressed with defence genes, transcription factors and so on, suggested their crucial role in various biological processes.
    CONCLUSIONS: Overall, our findings provide a way to understand the structure and function of SlE2F/DP genes; it might be helpful to improve fruit development and tolerance against abiotic stress through marker-assisted selection or transgenic approaches.
    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
    细胞周期事件由细胞周期蛋白依赖性激酶(CDK)协调以确保稳健的细胞分裂。CDK4/6和CDK2通过响应有丝分裂原信号调节细胞周期的生长1(G1)到合成(S)相变,促进E2F转录和抑制后期促进复合物。我们发现,在G2停滞的细胞中仍然需要这种机制来防止S期后细胞周期退出。这种机制揭示了CDK4/6在维持G2状态中的作用,挑战细胞周期是不可逆的,细胞在通过限制点后不需要有丝分裂原的概念。从G2退出发生在利波毒性应激期间,并由应激激活的蛋白激酶积极介导。在缓解压力时,相当一部分细胞经历了第二轮DNA复制,导致全基因组加倍.
    Cell cycle events are coordinated by cyclin-dependent kinases (CDKs) to ensure robust cell division. CDK4/6 and CDK2 regulate the growth 1 (G1) to synthesis (S) phase transition of the cell cycle by responding to mitogen signaling, promoting E2F transcription and inhibition of the anaphase-promoting complex. We found that this mechanism was still required in G2-arrested cells to prevent cell cycle exit after the S phase. This mechanism revealed a role for CDK4/6 in maintaining the G2 state, challenging the notion that the cell cycle is irreversible and that cells do not require mitogens after passing the restriction point. Exit from G2 occurred during ribotoxic stress and was actively mediated by stress-activated protein kinases. Upon relief of stress, a significant fraction of cells underwent a second round of DNA replication that led to whole-genome doubling.
    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
    E2F转录因子与肿瘤的发展有关,但它们在胃癌(GC)中的潜在机制仍不清楚。这项研究探讨了E2Fs是否决定了GC患者的预后或免疫和治疗反应。
    系统研究了来自癌症基因组图谱(TCGA)的E2F调控模式,并将E2F模式与肿瘤微环境(TME)中细胞浸润的特征相关。主成分分析用于构建基于预后相关差异基因的E2F评分模型,以量化单个肿瘤的E2F调节。然后在患者队列中测试该评分模型以预测免疫疗法的效果。
    基于E2F转录因子在GC中的表达谱,确定了两种不同的E2F调控模式。两个集群之间出现了TME和生存差异。Cluster2组中较低的存活率归因于基质激活导致的有限的免疫功能。然后基于E2F相关预后基因构建E2F评分模型。证据支持E2F评分作为免疫治疗反应的独立和有效的预后因素和预测因子。基因集分析将E2F评分与TME内免疫细胞浸润的特征相关联。免疫治疗队列数据库显示,E2F评分较高的患者表现出更好的生存和免疫反应。
    这项研究发现,GC预后的差异可能与TME中的E2F模式有关。本研究开发的E2F评分系统作为GC患者生存和治疗反应的预测因子具有实用价值。
    UNASSIGNED: E2F transcription factors are associated with tumor development, but their underlying mechanisms in gastric cancer (GC) remain unclear. This study explored whether E2Fs determine the prognosis or immune and therapy responses of GC patients.
    UNASSIGNED: E2F regulation patterns from The Cancer Genome Atlas (TCGA) were systematically investigated and E2F patterns were correlated with the characteristics of cellular infiltration in the tumor microenvironment (TME). A principal component analysis was used to construct an E2F scoring model based on prognosis-related differential genes to quantify the E2F regulation of a single tumor. This scoring model was then tested in patient cohorts to predict effects of immunotherapy.
    UNASSIGNED: Based on the expression profiles of E2F transcription factors in GC, two different regulatory patterns of E2F were identified. TME and survival differences emerged between the two clusters. Lower survival rates in the Cluster2 group were attributed to limited immune function due to stromal activation. The E2F scoring model was then constructed based on the E2F-related prognostic genes. Evidence supported the E2F score as an independent and effective prognostic factor and predictor of immunotherapy response. A gene-set analysis correlated E2F score with the characteristics of immune cell infiltration within the TME. The immunotherapy cohort database showed that patients with a higher E2F score demonstrated better survival and immune responses.
    UNASSIGNED: This study found that differences in GC prognosis might be related to the E2F patterns in the TME. The E2F scoring system developed in this study has practical value as a predictor of survival and treatment response in GC 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
    E2F转录因子(E2F)是一组编码转录因子家族的基因。它们已被确定为参与各种癌症类型的肿瘤进展。然而,对表达水平知之甚少,遗传变异,分子机制,HNSCC中不同E2Fs的预后价值和免疫浸润。在这项研究中,我们利用多个数据库来调查mRNA表达水平,遗传改变,HNSCC患者E2Fs的生物学功能。然后,E2Fs表达及其与发生的关系,进展,预后,并评估了HNSCC患者的免疫细胞浸润。我们发现所有八个E2Fs在HNSCC组织中的表达高于正常组织,E2F1/2/3/4/5/6/8的表达水平也与HNSCC的分期和分级有关。E2F1/2/4/8在HNSCC患者中的异常表达与临床转归有关。在HNSCC患者中,E2Fs的表达与免疫细胞浸润呈统计学相关,而B细胞和CD8T细胞的浸润与较好的OS呈正相关。此外,我们在验证实验中验证了组织水平的E2F2.我们的研究可能为HNSCC患者的免疫治疗靶标和潜在预后生物标志物的选择提供新的见解。
    E2F transcription factors (E2Fs) are a group of genes that encode a family of transcription factors. They have been identified as being involved in the tumor progression of various cancer types. However, little is known about the expression level, genetic variation, molecular mechanism, and prognostic value and immune infiltration of different E2Fs in HNSCC.In this study, we utilized multiple databases to investigate the mRNA expression level, genetic alteration, and biological function of E2Fs in HNSCC patients. Then, the relationship between E2Fs expression and its association with the occurrence, progress, prognosis, and immune cell infiltration in patients with HNSCC was evaluated. We found that all eight E2Fs were higher expressed in HNSCC tissues than in normal tissues, and the expression levels of E2F1/2/3/4/5/6/8 were also associated with the stage and grade of HNSCC. The abnormal expression of E2F1/2/4/8 in HNSCC patients is related to the clinical outcome. The expression of E2Fs was statistically correlated with the immune cell infiltration in HNSCC and the infiltration of B cells and CD8+ T cells were positively associated with better OS in HNSCC patients. Furthermore, we verified the E2F2 at the tissue level in the validation experiment. Our study may provide novel insights into the choice of immunotherapy targets and potential prognostic biomarkers in HNSCC patients.
    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
    鉴于α-檀香醇在多种癌症中的抑瘤作用以及E2F转录因子1(E2F1)作为抗癌研究的重要靶点的作用,本研究探讨了α-桑坦洛尔与E2F1的关系,以及α-桑坦洛尔对肝癌进展的影响及其机制。具体而言,用不同浓度的α-檀香醇处理肝癌细胞。使用Probit回归分析确定α-檀香醇的IC50值。然后,筛选出α-桑他洛尔靶向并在肝癌中差异表达的转录因子。对E2F1及其靶点的临床病理影响进行评价和预测。采用生物信息学方法分析E2F1和高迁移率族蛋白2(HMGB2)在肝癌组织中的表达及其相关性。E2F1和HMGB2对肝癌细胞生物学特性的影响通过功能丧失/获得和分子测定来检测。随着治疗时间的延长,10μmol/L和20μmol/Lα-桑他洛尔对癌细胞存活率的抑制作用均增强(P<0.001)。E2F1和HMGB2在肝癌组织中高表达且呈正相关(P<0.05)。E2F1高表达与肿瘤大、TNM分期高相关(P<0.05)。E2F1敲低促进了α-桑他洛尔剂量依赖性地抑制活力的作用,菌落形成,侵袭和迁移(P<0.05)。此外,E2F1敲除降低了IC50值和HMGB2水平,而HMGB2过表达产生相反的作用。HMGB2过表达和E2F1敲低对α-桑坦洛尔处理的肝癌细胞的IC50值以及对其活力和凋亡的影响相互抵消(P<0.01)。总的来说,阻断E2F1/HMGB2通路增强了α-桑坦洛尔对细胞增殖的干预作用,肝癌细胞的迁移和侵袭。
    In view of the tumor-inhibiting effect of α-santalol in various cancers and the role of E2F transcription factor 1 (E2F1) as an important target for anticancer research, this study investigates the relation between α-santalol and E2F1, as well as the effect of α-santalol on liver cancer progression and the corresponding mechanism. Concretely, liver cancer cells were treated with different concentrations of α-santalol. The IC50 value of α-santalol was determined using Probit regression analysis. Then, transcription factors that are targeted by α-santalol and differentially expressed in liver cancer were screened out. The clinicopathological impact of E2F1 and its targets were evaluated and predicted. The expressions of E2F1 and high-mobility group box 2 (HMGB2) and their correlation in the liver cancer tissues were analyzed by bioinformatics. The effects of E2F1 and HMGB2 on the biological characteristics of liver cancer cells were examined through loss/gain-of-function and molecular assays. With the extension of treatment time, the inhibitory effects of 10 μmol/L and 20 μmol/L α-santalol on cancer cell survival rate were enhanced (P < 0.001). E2F1 and HMGB2 were highly expressed and positively correlated in liver cancer tissues (P < 0.05). High E2F1 expression was correlated with large tumors and high TNM stages (P < 0.05). E2F1 knockdown promoted the effects of α-santalol on dose-dependently inhibiting viability, colony formation, invasion and migration (P < 0.05). Moreover, E2F1 knockdown reduced the IC50 value and HMGB2 level, while HMGB2 overexpression produced opposite effects. HMGB2 overexpression and E2F1 knockdown mutually counteracted their effects on the IC50 value and on the viability and apoptosis of α-santalol-treated liver cancer cells (P < 0.01). Collectively, blocking the E2F1/HMGB2 pathway enhances the intervention effects of α-santalol on the proliferation, migration and invasion of liver cancer 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
    卷曲螺旋α-螺旋杆状蛋白1(CCHCR1)最初被鉴定为银屑病的候选基因,最近被发现与包括COVID-19在内的各种临床疾病有关。CCHCR1位于P体和中心体,但是它在这两个亚细胞结构中的确切作用及其转录控制仍在很大程度上未知。这里,我们发现CCHCR1与其相邻基因共享双向启动子,TCF19.这种双向启动子被G1/S调节转录因子E2F1激活,并且两个基因在细胞周期的G1/S转换过程中共同诱导。荧光素酶报告基因分析表明,短的基因间序列,只有287个bp长,足以诱导两个基因的G1/S,但是由于TCF19和CCHCR1的外显子1的存在,CCHCR1的表达进一步增强。本研究揭示了CCHCR1基因的转录调控,为其功能提供新的视角。这些发现有助于更广泛地了解与CCHCR1相关的疾病,并可能作为这些重要医学领域未来研究的基础基准。
    The coiled-coil alpha-helical rod protein 1 (CCHCR1) was first identified as a candidate gene in psoriasis and has lately been found to be associated with a wide range of clinical conditions including COVID-19. CCHCR1 is located within P-bodies and centrosomes, but its exact role in these two subcellular structures and its transcriptional control remain largely unknown. Here, we showed that CCHCR1 shares a bidirectional promoter with its neighboring gene, TCF19. This bidirectional promoter is activated by the G1/S-regulatory transcription factor E2F1, and both genes are co-induced during the G1/S transition of the cell cycle. A luciferase reporter assay suggests that the short intergenic sequence, only 287 bp in length, is sufficient for the G1/S induction of both genes, but the expression of CCHCR1 is further enhanced by the presence of exon 1 from both TCF19 and CCHCR1. This research uncovers the transcriptional regulation of the CCHCR1 gene, offering new perspectives on its function. These findings contribute to the broader understanding of diseases associated with CCHCR1 and may serve as a foundational benchmark for future research in these vital medical fields.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号