retinoblastoma protein

  • 文章类型: Journal Article
    人乳头瘤病毒(HPV)部分地通过劫持宿主细胞周期和迫使不受控制的细胞分裂而导致肿瘤。虽然有超过200基因型的HPV,15被分类为高风险,并且已经显示出转化受感染的细胞并有助于肿瘤形成。其余的低风险基因型不被认为是致癌的,并导致良性皮肤病变。在高危型HPV中,癌蛋白E7有助于细胞周期调控机制的失调。高风险E7在细胞中的两个保守丝氨酸残基处被酪蛋白激酶2(CK2)磷酸化,并且该磷酸化事件增加了对细胞蛋白(例如肿瘤抑制因子视网膜母细胞瘤(pRb))的结合亲和力。虽然低风险E7具有相似的丝氨酸残基,它在细胞中磷酸化程度较低,结合能力下降。当E7结合亲和力降低时,它不能够促进蛋白质之间复杂的相互作用,因此具有较小的能力,以失调细胞周期。通过比较低危和高危HPV变体的E7蛋白序列,并使用定点突变结合NMR光谱和基于细胞的测定,我们证明了CK2识别序列中存在两个关键的非极性缬氨酸残基,存在于低风险E7中,相对于高风险E7降低丝氨酸磷酸化效率。这导致E7降解视网膜母细胞瘤肿瘤抑制蛋白的能力显著丧失,因此也降低了E7增加细胞增殖和减少衰老的能力。这提供了对当细胞感染高风险与低风险HPV时E7介导的差异结果的额外见解。了解这些致癌差异对于开发HPV诱导的癌症的靶向治疗方案可能很重要。
    The Human papillomavirus (HPV) causes tumors in part by hijacking the host cell cycle and forcing uncontrolled cellular division. While there are >200 genotypes of HPV, 15 are classified as high-risk and have been shown to transform infected cells and contribute to tumor formation. The remaining low-risk genotypes are not considered oncogenic and result in benign skin lesions. In high-risk HPV, the oncoprotein E7 contributes to the dysregulation of cell cycle regulatory mechanisms. High-risk E7 is phosphorylated in cells at two conserved serine residues by Casein Kinase 2 (CK2) and this phosphorylation event increases binding affinity for cellular proteins such as the tumor suppressor retinoblastoma (pRb). While low-risk E7 possesses similar serine residues, it is phosphorylated to a lesser degree in cells and has decreased binding capabilities. When E7 binding affinity is decreased, it is less able to facilitate complex interactions between proteins and therefore has less capability to dysregulate the cell cycle. By comparing E7 protein sequences from both low- and high-risk HPV variants and using site-directed mutagenesis combined with NMR spectroscopy and cell-based assays, we demonstrate that the presence of two key nonpolar valine residues within the CK2 recognition sequence, present in low-risk E7, reduces serine phosphorylation efficiency relative to high-risk E7. This results in significant loss of the ability of E7 to degrade the retinoblastoma tumor suppressor protein, thus also reducing the ability of E7 to increase cellular proliferation and reduce senescence. This provides additional insight into the differential E7-mediated outcomes when cells are infected with high-risk verses low-risk HPV. Understanding these oncogenic differences may be important to developing targeted treatment options for HPV-induced cancers.
    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
    视网膜母细胞瘤,小儿眼部恶性肿瘤,在理解其分子基础和有针对性的治疗方法方面提出了重大挑战。组蛋白脱乙酰酶(HDAC)的活性失调与视网膜母细胞瘤的发病机制有关。影响关键细胞过程,如细胞周期调节或视网膜神经节细胞凋亡。通过它们的脱乙酰酶活性,HDAC对关键的肿瘤抑制因子和癌基因发挥控制作用,影响增殖和细胞死亡之间的微妙平衡。此外,HDAC和视网膜母细胞瘤蛋白通路之间的相互作用,视网膜母细胞瘤病因学的一个关键方面,揭示了影响肿瘤微环境的相互作用的复杂网络。HDAC抑制剂的检查,包括已建立的和新的化合物,提供了恢复乙酰化平衡和阻止视网膜母细胞瘤进展的潜在方法的见解。此外,在视网膜母细胞瘤中表现出不同表达的特定HDAC亚型的鉴定为个性化治疗策略提供了途径,允许针对个体患者概况进行干预。这篇综述集中在HDAC和视网膜母细胞瘤之间复杂的相互关系。阐明控制肿瘤发展和进展的表观遗传机制。对HDAC靶向疗法的探索强调了创新治疗方式在寻求更有效和个性化治疗这种疾病的策略方面的潜力。
    Retinoblastoma, a pediatric ocular malignancy, presents significant challenges in comprehending its molecular underpinnings and targeted therapeutic approaches. The dysregulated activity of histone deacetylases (HDACs) has been associated with retinoblastoma pathogenesis, influencing critical cellular processes like cell cycle regulation or retinal ganglion cell apoptosis. Through their deacetylase activity, HDACs exert control over key tumor suppressors and oncogenes, influencing the delicate equilibrium between proliferation and cell death. Furthermore, the interplay between HDACs and the retinoblastoma protein pathway, a pivotal aspect of retinoblastoma etiology, reveals a complex network of interactions influencing the tumor microenvironment. The examination of HDAC inhibitors, encompassing both established and novel compounds, offers insights into potential approaches to restore acetylation balance and impede retinoblastoma progression. Moreover, the identification of specific HDAC isoforms exhibiting varying expression in retinoblastoma provides avenues for personalized therapeutic strategies, allowing for interventions tailored to individual patient profiles. This review focuses on the intricate interrelationship between HDACs and retinoblastoma, shedding light on epigenetic mechanisms that control tumor development and progression. The exploration of HDAC-targeted therapies underscores the potential for innovative treatment modalities in the pursuit of more efficacious and personalized management strategies for this disease.
    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
    原理:心肌梗塞(MI)是一种严重的全球性临床疾病,普遍存在。成年哺乳动物心脏对损伤的反应产生新的心肌细胞(CMs)的能力有限仍然是开发有效疗法的主要障碍。目前的方法集中在通过细胞周期再入诱导现有CM的增殖。然而,这种方法主要提高细胞周期蛋白依赖性激酶6(CDK6)和DNA含量,缺乏适当的胞质分裂,并导致功能失调的双核CMs的形成。胞质分裂依赖于核糖体生物发生(Ribo-bio),核仁素(Ncl)调节的一个关键过程。我们的目标是确定一种促进DNA合成和胞质分裂的新方法。方法:各种技术,包括RNA/蛋白质测序分析,Ribo-Halo,Ribo-disome,流式细胞术,和心脏特异性肿瘤抑制视网膜母细胞瘤-1(Rb1)基因敲除小鼠,用于评估增殖/细胞周期再入和Ribo-bio/胞质分裂的系列信号传导。超声心动图,共焦成像,和组织学用于评估心功能。结果:与对照小鼠相比,分析显示MI小鼠心脏中Rb1的水平显着升高,circASXL1的水平降低。Rb1的缺失仅诱导细胞周期重新进入,同时增强Ribo-生物调节剂Ncl导致胞质分裂。机械上,生物信息学和损失/增益研究发现circASXL1/CDK6/Rb1调节细胞周期重新进入。此外,Ribo-Halo,Ribo-disome和circRNA下拉测定表明circASXL1通过Ncl/Ribo-bio促进胞质分裂。重要的是,来自脐带间充质干细胞(UMSC-Exo)的外泌体能够通过促进细胞周期折返和Ribo-bio/胞质分裂的协调信号来增强心脏功能。通过在UMSC-Exo中沉默circASXL1来减弱这些作用。结论:circASXL1/CDK6/Rb1/细胞周期折返和circASXL1/Ncl/Ribo-bio/胞质分裂的系列信号在心脏修复中起着至关重要的作用。UMSC-Exo通过以circASXL1依赖性方式刺激CM细胞周期折返和胞质分裂来有效修复梗塞心肌。这项研究提供了针对MI的circASXL1信号网络的创新治疗策略,并提供了增强心脏修复的潜在途径。
    Rationale: Myocardial infarction (MI) is a severe global clinical condition with widespread prevalence. The adult mammalian heart\'s limited capacity to generate new cardiomyocytes (CMs) in response to injury remains a primary obstacle in developing effective therapies. Current approaches focus on inducing the proliferation of existing CMs through cell-cycle reentry. However, this method primarily elevates cyclin dependent kinase 6 (CDK6) and DNA content, lacking proper cytokinesis and resulting in the formation of dysfunctional binucleated CMs. Cytokinesis is dependent on ribosome biogenesis (Ribo-bio), a crucial process modulated by nucleolin (Ncl). Our objective was to identify a novel approach that promotes both DNA synthesis and cytokinesis. Methods: Various techniques, including RNA/protein-sequencing analysis, Ribo-Halo, Ribo-disome, flow cytometry, and cardiac-specific tumor-suppressor retinoblastoma-1 (Rb1) knockout mice, were employed to assess the series signaling of proliferation/cell-cycle reentry and Ribo-bio/cytokinesis. Echocardiography, confocal imaging, and histology were utilized to evaluate cardiac function. Results: Analysis revealed significantly elevated levels of Rb1, bur decreased levels of circASXL1 in the hearts of MI mice compared to control mice. Deletion of Rb1 induces solely cell-cycle reentry, while augmenting the Ribo-bio modulator Ncl leads to cytokinesis. Mechanically, bioinformatics and the loss/gain studies uncovered that circASXL1/CDK6/Rb1 regulates cell-cycle reentry. Moreover, Ribo-Halo, Ribo-disome and circRNA pull-down assays demonstrated that circASXL1 promotes cytokinesis through Ncl/Ribo-bio. Importantly, exosomes derived from umbilical cord mesenchymal stem cells (UMSC-Exo) had the ability to enhance cardiac function by facilitating the coordinated signaling of cell-cycle reentry and Ribo-bio/cytokinesis. These effects were attenuated by silencing circASXL1 in UMSC-Exo. Conclusion: The series signaling of circASXL1/CDK6/Rb1/cell-cycle reentry and circASXL1/Ncl/Ribo-bio/cytokinesis plays a crucial role in cardiac repair. UMSC-Exo effectively repairs infarcted myocardium by stimulating CM cell-cycle reentry and cytokinesis in a circASXL1-dependent manner. This study provides innovative therapeutic strategies targeting the circASXL1 signaling network for MI and offering potential avenues for enhanced cardiac repair.
    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
    视网膜母细胞瘤蛋白在所有后生动物和癌症发展中的增殖和分化等基本细胞决定的信号网络中处于核心地位。免疫染色和生化证据表明,在间期视网膜母细胞瘤蛋白在细胞核中,并且是低磷酸化的,在有丝分裂过程中在细胞质中过度磷酸化。这项研究的目的是在体内观察非患病组织,在有丝分裂过程中,动态的空间和时间核向该蛋白质的细胞质退出,并返回细胞核,以了解其潜在的细胞质功能。使用来自共聚焦显微镜的高分辨率延时图像,我们在植物体内跟踪了拟南芥根中绿色荧光蛋白(GFP)标记的视网膜母细胞瘤相关(RBR)蛋白的直系同源物。RBR蛋白在不到2分钟的时间内,在染色体处于前期之前,从细胞核中的致密聚集体中退出。在有丝分裂期间,像扩散但受控的事件一样,作为较小的颗粒投射到整个细胞质中,直到末期,当子核形成时;RBR与去凝聚的染色体DNA协调地返回到核,在每个相应的核中再次形成新的聚集体,这些聚集体被标记为更大的结构。我们提出,细胞质中的RBR扩散颗粒可能充当传入信号的胞浆传感器,因此,协调与DNA的再聚集是一种机制,通过该机制,RBR遇到的任何新的传入信号都可能导致核转录组上下文的重新配置.细胞质中的小RBR扩散颗粒可能保留拓扑样特性,使它们能够聚集并恢复其核位置,它们也可能是细胞有丝分裂前转录环境的瞬时细胞质储存的一部分,一旦进入细胞核,就可以执行有丝分裂前的转录上下文以及新的转录指令。
    Retinoblastoma protein is central in signaling networks of fundamental cell decisions such as proliferation and differentiation in all metazoans and cancer development. Immunostaining and biochemical evidence demonstrated that during interphase retinoblastoma protein is in the nucleus and is hypophosphorylated, and during mitosis is in the cytoplasm and is hyperphosphorylated. The purpose of this study was to visualize in vivo in a non-diseased tissue, the dynamic spatial and temporal nuclear exit toward the cytoplasm of this protein during mitosis and its return to the nucleus to obtain insights into its potential cytosolic functions. Using high-resolution time-lapse images from confocal microscopy, we tracked in vivo the ortholog in plants the RETINOBLASTOMA RELATED (RBR) protein tagged with Green Fluorescent Protein (GFP) in Arabidopsis thaliana\'s root. RBR protein exits from dense aggregates in the nucleus before chromosomes are in prophase in less than 2 min, spreading outwards as smaller particles projected throughout the cytosol during mitosis like a diffusive yet controlled event until telophase, when the daughter\'s nuclei form; RBR returns to the nuclei in coordination with decondensing chromosomal DNA forming new aggregates again in punctuated larger structures in each corresponding nuclei. We propose RBR diffused particles in the cytoplasm may function as a cytosolic sensor of incoming signals, thus coordinating re-aggregation with DNA is a mechanism by which any new incoming signals encountered by RBR may lead to a reconfiguration of the nuclear transcriptomic context. The small RBR diffused particles in the cytoplasm may preserve topologic-like properties allowing them to aggregate and restore their nuclear location, they may also be part of transient cytoplasmic storage of the cellular pre-mitotic transcriptional context, that once inside the nuclei may execute both the pre mitosis transcriptional context as well as new transcriptional instructions.
    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
    小鼠模型中的功能分析对于建立一组遗传变异参与肿瘤发展是必要的。促进和经济有效地分析多个基因在致癌作用中的作用的建模平台将是有价值的。这里,我们提出了一种通过阳离子聚合物递送的CRISPR/Cas9核糖核蛋白进行肺诱变的创新策略.这种方法允许多个基因的同时失活。我们通过靶向一组肿瘤抑制基因来验证这个系统的有效性,特别是Rb1,Rbl1,Pten,选择Trp53是因为它们有可能导致肺部肿瘤,即小细胞肺癌(SCLC)。气管内施用CRISPR/聚合物纳米颗粒后出现具有人类SCLC组织学和转录组学特征的肿瘤。这些肿瘤在靶向位置的所有四个肿瘤抑制基因中携带功能丧失突变。这些发现在两种不同的纯遗传背景中再现。我们为肺肿瘤发生的简化建模提供了原理证明,以促进潜在癌症相关基因的功能测试。
    Functional analysis in mouse models is necessary to establish the involvement of a set of genetic variations in tumor development. A modeling platform to facilitate and cost-effectively analyze the role of multiple genes in carcinogenesis would be valuable. Here, we present an innovative strategy for lung mutagenesis using CRISPR/Cas9 ribonucleoproteins delivered via cationic polymers. This approach allows the simultaneous inactivation of multiple genes. We validate the effectiveness of this system by targeting a group of tumor suppressor genes, specifically Rb1, Rbl1, Pten, and Trp53, which were chosen for their potential to cause lung tumors, namely small cell lung carcinoma (SCLC). Tumors with histologic and transcriptomic features of human SCLC emerged after intratracheal administration of CRISPR/polymer nanoparticles. These tumors carried loss-of-function mutations in all four tumor suppressor genes at the targeted positions. These findings were reproduced in two different pure genetic backgrounds. We provide a proof of principle for simplified modeling of lung tumorigenesis to facilitate functional testing of potential cancer-related genes.
    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
    患有人乳头瘤病毒(HPV+)相关喉鳞状细胞癌(LSCC)的患者相对于患有HPV阴性(HPV-)肿瘤的患者表现出显著改善的存活率。在这项研究中,作者旨在研究所有已确认的HPV+和HPV-LSCC细胞在体外和体内的放射敏感性.
    从患者获得的肿瘤样本产生原代LSCC细胞。进行实时PCR以确认HPV感染和HPV相关基因(E6和E7)的表达,p53和pRB。克隆性存活测定,西方印迹,流式细胞术用于评估辐射敏感性,凋亡,p53和pRB的表达。使用CRISPR/Cas9技术产生p53和pRB敲除细胞。
    与HPV-细胞相比,HPV+LSCC细胞显示增强的辐射敏感性。辐射诱导HPV+LSCC细胞凋亡,伴随着p53和pRB水平的增加。p53或pRB的基因敲除导致HPV+LSCC细胞中的辐射抗性并减弱辐射诱导的凋亡。体内实验结果相似,其中p53或pRB的敲除降低了荷瘤小鼠的放射敏感性。
    目前的研究结果表明,HPV+LSCC细胞表现出明显的固有辐射敏感性,对应于辐射暴露后细胞凋亡增加。机制研究表明,p53和pRB在HPV+细胞中的表达是放射敏感性所必需的。这些发现强调了一种新的机制,通过该机制,与HPV-LSCC相比,p53和pRB在HPVLSCC的辐射敏感性中起关键作用。
    Patients with Human Papillomavirus (HPV+)-associated Laryngeal Squamous Cell Carcinoma (LSCC) exhibit dramatically improved survival relative to those with HPV-Negative (HPV-) tumors. In this study, the authors aimed to investigate the radiosensitivity of all available confirmed HPV+ and HPV-LSCC cells in vitro and in vivo.
    Primary LSCC cells were generated from tumor specimens obtained from patients. Real-time PCR was performed to confirm HPV infection and the expression of HPV-related genes (E6 and E7), p53, and pRB. Clonogenic survival assays, western blotting, and flow cytometry were used to assess radiation sensitivity, apoptosis, and the expression of p53 and pRB. p53 and pRB knockout cells were generated using CRISPR/Cas9 technology.
    HPV+ LSCC cells displayed enhanced radiation sensitivity compared to HPV- cells. Radiation-induced apoptosis in HPV+ LSCC cells, accompanied by increased levels of p53 and pRB. Knockout of p53 or pRB led to radiation resistance and attenuated radiation-induced apoptosis in HPV+ LSCC cells. In vivo experiments showed similar results, where knockout of p53 or pRB decreased radiosensitivity in tumor-bearing mice.
    The present findings demonstrated that HPV+ LSCC cells displayed obvious inherent radiation sensitivity, corresponding to increased apoptosis following radiation exposure. Mechanism study showed that the expression of p53 and pRB in HPV+ cells are required for radiation sensitivity. These findings highlight a novel mechanism by which p53 and pRB play key roles in the radiation sensitivity of HPV+ LSCC compared to HPV-LSCC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Letter
    暂无摘要。
    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:SGI-1027是公认的DNA甲基转移酶1(DNMT1)抑制剂,早期的研究表明,胃癌(GC)中DNMT1表达失调与视网膜母细胞瘤1(RB1)基因表达呈负相关。尽管有这些知识,SGI-1027在GC细胞中作用的确切机制仍未被充分理解。本研究的主要目的是阐明SGI-1027对GC细胞行为的影响,包括生长和转移潜力等方面,通过干预DNMT1,从而影响RB1基因表达的调控。
    方法:获取正常胃粘膜细胞系GES-1和人胃癌细胞系MKN45后,采用Westernblot(WB)和定量逆转录-聚合酶链反应(qRT-PCR)技术评价RB1和DNMT1在这两种细胞系中的表达水平。随后,MKN45细胞系在含有不同浓度SGI-1027的培养基中培养,并使用WB和qRT-PCR技术重新评估SGI-1027对GC细胞中RB1和DNMT1调节的影响。为了仔细检查SGI-1027对GC细胞的影响,我们利用3-(4,5-二甲基-2-噻唑基)-2,5-二苯基-2H四唑溴化物(MTT)测定来确定细胞增殖,并进行Transwell实验来评估细胞迁移和侵袭能力.在整个过程中,我们还使用WB评估细胞周期相关蛋白(CyclinD1,CyclinE1和CyclinB1)和细胞凋亡相关蛋白(BCL-2相关蛋白X凋亡调节因子(BAX)和B细胞淋巴瘤2凋亡调节因子(BCL-2))的水平.此外,我们将以最佳浓度的SGI-1027培养的MKN45细胞系和MKN45细胞系分别注射入小鼠皮下和通过尾静脉注射5天和10天,将它们划分为模型组,型号+SGI-10275d组,和Model+SGI-102710D组。我们监测了小鼠肿瘤大小和体积的变化,收集肿瘤组织和肺组织进行苏木精和曙红(HE)染色。最后,使用WB和免疫组织化学(IHC)技术检测GC组织中的DNMT1表达水平。此外,使用WB评估GC组织中的RB1表达水平。
    结果:与GES-1细胞相比,MKN45细胞表现出独特的特征谱,其特征在于DNMT1表达增加和RB1表达减少(p<0.05)。然而,在引入SGI-1027后,观察到GC细胞内DNMT1水平显着降低,伴随着RB1基因表达的升高,以25μmol/LSGI-1027为最佳浓度(p<0.05)。功能测定表明,SGI-1027处理的GC细胞表现出明显的抑制增殖特征,迁移,当与未处理的MKN45细胞相比时(p<0.05)。此外,在SGI-1027处理的GC细胞中,CyclinD1,CyclinE1,CyclinB1和BCL-2的水平显着降低,而BAX的表达水平升高(p<0.05)。值得注意的是,在25μmol/LSGI-1027时观察到最明显的影响,进一步强调了其对肿瘤细胞行为的调节作用(p<0.05)。在动物实验中,模型组肿瘤体积显著增加,HE染色结果提示大部分胃组织广泛坏死和明显的肺转移征象,伴随着DNMT1表达增加和RB1基因表达减少。相比之下,SGI-1027组显示胃肿瘤体积减少,坏死减少,肺肿瘤转移减少(p<0.05)。此外,DNMT1的表达在SGI-1027处理的GC细胞中显著降低,而RB1表达增加(p<0.05),进一步证实了SGI-1027对肿瘤生长和转移的抑制作用。
    结论:SGI-1027通过下调DNMT1和促进RB1的表达,有效阻碍GC细胞的增殖和播散。
    BACKGROUND: SGI-1027 is a recognized inhibitor of DNA methyltransferase 1 (DNMT1), and earlier investigations have indicated an inverse correlation between dysregulated DNMT1 expression in gastric cancer (GC) and retinoblastoma 1 (RB1) gene expression. Despite this knowledge, the precise mechanisms underlying the action of SGI-1027 in GC cells remain inadequately comprehended. The primary objective of this study is to elucidate the impact of SGI-1027 on the behavior of GC cells, encompassing aspects such as growth and metastatic potential, by intervening in DNMT1, thereby influencing the regulation of RB1 gene expression.
    METHODS: The acquisition of the normal gastric mucosal cell line GES-1 and the human gastric cancer cell line MKN45 was followed by employing Western blot (WB) and quantitative reverse transcription-polymerase chain reaction (qRT-PCR) techniques to evaluate the expression levels of RB1 and DNMT1 in these two cell lines. Subsequently, the MKN45 cell line was cultured in medium containing varying concentrations of SGI-1027, and the impact of SGI-1027 on the regulation of RB1 and DNMT1 in GC cells was reassessed using WB and qRT-PCR techniques. To scrutinize the effect of SGI-1027 on GC cells, we utilized the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H tetrazolium bromide (MTT) assay to determine cell proliferation and performed Transwell experiments to assess cell migration and invasion capabilities. Throughout this process, we also employed WB to assess the levels of cell cycle-associated proteins (Cyclin D1, Cyclin E1, and Cyclin B1) and proteins related to apoptosis (BCL-2 associated protein X apoptosis regulator (BAX) and B-cell lymphoma 2 apoptosis regulator (BCL-2)). Furthermore, we injected the MKN45 cell line and MKN45 cell line cultured with the optimal concentration of SGI-1027 for 5 days and 10 days into mice subcutaneously and through the tail vein, dividing them into the Model group, Model+SGI-1027 5d group, and Model+SGI-1027 10d group. We monitored changes in tumor size and volume in mice, and tumor tissues as well as lung tissues were collected for hematoxylin and eosin (HE) staining. Finally, DNMT1 expression levels in GC tissues were detected using both WB and immunohistochemistry (IHC) techniques. Additionally, RB1 expression levels in GC tissues were assessed using WB.
    RESULTS: In contrast to GES-1 cells, MKN45 cells displayed a distinctive profile characterized by increased DNMT1 expression and decreased RB1 expression (p < 0.05). However, upon the introduction of SGI-1027, a notable decrease in DNMT1 levels within GC cells was observed, concomitant with an elevation in RB1 gene expression, with 25 μmol/L SGI-1027 identified as the optimal concentration (p < 0.05). Functional assays demonstrated that SGI-1027-treated GC cells exhibited pronounced features of inhibited proliferation, migration, and invasion when compared to untreated MKN45 cells (p < 0.05). Moreover, in SGI-1027-treated GC cells, the levels of Cyclin D1, Cyclin E1, Cyclin B1, and BCL-2 were significantly reduced, while the expression level of BAX increased (p < 0.05). Notably, the most pronounced impact was observed at 25 μmol/L SGI-1027, further underscoring its regulatory effects on tumor cell behavior (p < 0.05). In animal experiments, the Model group exhibited a substantial increase in tumor volume, with HE staining results indicating extensive necrosis in most gastric tissues and noticeable signs of lung metastasis, accompanied by increased DNMT1 expression and decreased RB1 gene expression. In contrast, the SGI-1027 group displayed a reduction in gastric tumor volume, decreased necrosis, and reduced lung tumor metastasis (p < 0.05). Additionally, the expression of DNMT1 was significantly reduced in SGI-1027-treated GC cells, while RB1 expression increased (p < 0.05), further confirming the inhibitory effects of SGI-1027 on tumor growth and metastasis.
    CONCLUSIONS: SGI-1027 effectively hinders the proliferation and dissemination of GC cells by downregulating DNMT1 and promoting the expression of RB1.
    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
    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)

公众号