MutP53

mutp53
  • 文章类型: Journal Article
    表观遗传变化在癌症中很常见,包括异常的DNA甲基化和组蛋白修饰。包括乙酰化或甲基化。启动子区域的DNA甲基化和组蛋白脱乙酰通常伴随着基因沉默,并可能导致癌细胞中肿瘤抑制因子的抑制。据报道,表观遗传途径之间的相互作用可用于更有效地靶向侵袭性癌细胞。特别是那些目前的治疗通常失败,比如胰腺癌。在这项研究中,我们探索了将DNA去甲基化剂5-AZA与HDAC抑制剂SAHA联合治疗胰腺癌细胞系的可能性,专注于mutp53的乙酰化及其稳定性的后果,以及该蛋白与c-myc和BRCA-1的相互作用,这些是癌症生存的关键分子。获得的结果表明,SAHA/5-AZA组合比单一治疗更有效地促进mutp53的降解,上调p21并下调c-Myc和BRCA-1,从而增加胰腺癌细胞的DNA损伤和细胞毒性。
    Epigenetic changes are common in cancer and include aberrant DNA methylation and histone modifications, including both acetylation or methylation. DNA methylation in the promoter regions and histone deacetylation are usually accompanied by gene silencing, and may lead to the suppression of tumor suppressors in cancer cells. An interaction between epigenetic pathways has been reported that could be exploited to more efficiently target aggressive cancer cells, particularly those against which current treatments usually fail, such as pancreatic cancer. In this study, we explored the possibility to combine the DNA demethylating agent 5-AZA with HDAC inhibitor SAHA to treat pancreatic cancer cell lines, focusing on the acetylation of mutp53 and the consequences on its stability, as well as on the interaction of this protein with c-myc and BRCA-1, key molecules in cancer survival. The results obtained suggest that SAHA/5-AZA combination was more effective than single treatments to promote the degradation of mutp53, to upregulate p21 and downregulate c-Myc and BRCA-1, thus increasing DNA damage and cytotoxicity in pancreatic cancer cells.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Preprint
    应激相关分子伴侣系统是癌症治疗中的可行靶标。它在癌组织中普遍上调,并通过稳定数百种癌蛋白和干扰蛋白质复合物的化学计量来实现致瘤性。大多数抑制剂靶向关键组分热休克蛋白90(HSP90)。然而,尽管经典的HSP90抑制剂具有高度的肿瘤选择性,他们在3期临床肿瘤学试验中失败.这些故障至少部分是由于HSP90抑制对负反馈回路的干扰。称为热休克反应(HSR):响应HSP90抑制,有应激诱导的伴侣的补偿性合成,由转录因子热休克因子1(HSF1)介导。我们最近发现,野生型p53(p53)通过p21-CDK4/6-MAPK-HSF1轴抑制HSF1来积极降低HSR。在这里,我们测试了以下假设:在基于HSP90的治疗中,同时p53激活或直接细胞周期抑制会中断有害的HSF1-HSR轴并提高HSP90抑制剂的效率。的确,我们发现临床相关的p53激活剂Idasanutlin在基于HSP90抑制剂的治疗中抑制HSF1-HSR活性.这种组合协同降低了p53-functional结直肠癌(CRC)细胞的细胞活力并加速细胞死亡。鼠肿瘤衍生的类器官和患者衍生的类器官(PDO)。机械上,联合治疗后,人类CRC细胞强烈上调p53相关途径,凋亡,和炎症免疫途径。同样,在小鼠的化学AOM/DSSCRC模型中,双重HSF1-HSP90抑制强烈抑制肿瘤生长和重塑免疫细胞组成,然而,在小鼠和正常粘膜来源的类器官中仅表现出轻微的毒性。重要的是,HSP90抑制下细胞周期蛋白依赖性激酶4和6(CDK4/6)的抑制表型在p53有效的CRC细胞中协同抑制HSR。更重要的是,在p53缺陷(携带mutp53)CRC细胞中,HSP90抑制与CDK4/6抑制剂组合类似地抑制HSF1-HSR系统并减少癌症生长。同样,p53突变的PDO强烈响应双重HSF1-HSP90途径抑制,因此,提供独立于p53状态的靶向CRC的策略。总之,激活p53(在p53高表达的癌细胞中)或抑制CDK4/6(与p53状态无关)为改善基于HSP90的治疗的临床结果和增强结直肠癌治疗提供了新的选择.
    The stress-associated molecular chaperone system is an actionable target in cancer therapies. It is ubiquitously upregulated in cancer tissues and enables tumorigenicity by stabilizing hundreds of oncoproteins and disturbing the stoichiometry of protein complexes. Most inhibitors target the key component heat-shock protein 90 (HSP90). However, although classical HSP90 inhibitors are highly tumor-selective, they fail in phase 3 clinical oncology trials. These failures are at least partly due to an interference with a negative feedback loop by HSP90 inhibition, known as heat-shock response (HSR): in response to HSP90 inhibition there is compensatory synthesis of stress-inducible chaperones, mediated by the transcription factor heat-shock factor 1 (HSF1). We recently identified that wildtype p53 (p53) actively reduces the HSR by repressing HSF1 via a p21-CDK4/6-MAPK-HSF1 axis. Here we test the hypothesis that in HSP90-based therapies simultaneous p53 activation or direct cell cycle inhibition interrupts the deleterious HSF1-HSR axis and improves the efficiency of HSP90 inhibitors. Indeed, we find that the clinically relevant p53 activator Idasanutlin suppresses the HSF1-HSR activity in HSP90 inhibitor-based therapies. This combination synergistically reduces cell viability and accelerates cell death in p53-proficient colorectal cancer (CRC) cells, murine tumor-derived organoids and patient-derived organoids (PDOs). Mechanistically, upon combination therapy human CRC cells strongly upregulate p53-associated pathways, apoptosis, and inflammatory immune pathways. Likewise, in the chemical AOM/DSS CRC model in mice, dual HSF1-HSP90 inhibition strongly represses tumor growth and remodels immune cell composition, yet displays only minor toxicities in mice and normal mucosa-derived organoids. Importantly, inhibition of the cyclin dependent kinases 4 and 6 (CDK4/6) under HSP90 inhibition phenocopies synergistic repression of the HSR in p53-proficient CRC cells. Even more important, in p53-deficient (mutp53-harboring) CRC cells, an HSP90 inhibition in combination with CDK4/6 inhibitors similarly suppresses the HSF1-HSR system and reduces cancer growth. Likewise, p53-mutated PDOs strongly respond to dual HSF1-HSP90 pathway inhibition and thus, providing a strategy to target CRC independent of the p53 status. In sum, activating p53 (in p53-proficient cancer cells) or inhibiting CDK4/6 (independent of the p53 status) provide new options to improve the clinical outcome of HSP90-based therapies and to enhance colorectal cancer therapy.
    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
    UNASSIGNED: Identifying biological markers of colorectal cancer (CRC) development and prognosis and exploring the intrinsic connection between these molecular markers and CRC progression is underway. However, a single molecular tumor marker is often difficult to assess and predict the progression and prognosis of CRC. Consequently, a combination of tumor-related markers is much needed. Ki67, Her-2, and mutant P53 (MutP53) proteins play pivotal roles in CRC occurrence, progression and prognosis.
    UNASSIGNED: Based on the expressions by immunochemistry, we developed a risk model, nomogram and lymph node metastasis model by R software and Pythons to explore the value of these proteins in predicting CRC progression, prognosis, and examined the relationship of these proteins with the CRC clinicopathological features from 755 (training set) and 211 CRC (validation set) patients collected from the hospital.
    UNASSIGNED: We found that Ki67 expression was significantly correlated with T-stage, N-stage, TNM-stage, vascular invasion, organization differentiation, and adenoma carcinogenesis. Moreover, Her-2 expression was significantly correlated with T-stage, N-stage, TNM-stage, vascular and nerve invasion, pMMR/dMMR, signet ring cell carcinoma, and organization differentiation. MutP53 expression was significantly correlated with T-stage, N-stage, TNM-stage, vascular and nerve invasion, adenoma carcinogenesis, signet ring cell carcinoma, organization differentiation, and pMMR/dMMR. Increased expression of each of the protein indicated a poor prognosis. The established risk model based on the three key proteins showed high predictive value for determining the pathological characteristics and prognosis of CRC and was an independent influencer for prognosis. The nomogram prediction model, which was based on the risk model, after sufficient evaluation, showed more premium clinical value for predicting prognosis. Independent cohort of 211 CRC patients screened from the hospital verified the strong predictive efficacy of these models. The utilization of the XGBoost algorithm in a lymph node metastasis model, which incorporates three crucial proteins, demonstrated a robust predictive capacity for lymph node metastasis.
    UNASSIGNED: The risk model, nomogram and lymph node metastasis model have all provided valuable insights into the involvement of these three key proteins in the progression and prognosis of CRC. Our study provides a theoretical basis for further screening of effective models that utilize biological markers of CRC.
    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
    肿瘤相关的p53突变诱导与野生型p53不同的活性,从而导致蛋白质的肿瘤抑制功能丧失。携带p53突变的细胞具有与侵袭相关的更具侵袭性的特征,转移,扩散,细胞存活。通过比较突变p53(mutp53)和突变p53沉默队列的基因表达谱,我们发现FOS相关抗原-1(FRA-1),由FOSL1编码,是mutp53介导的转移的潜在效应物。我们证明FRA-1的表达,间质-上皮转化的看门人,在p53突变的存在下升高。机械上,突变型p53与转录因子ELK1结合并激活FOSL1的启动子,从而促进肺转移。这项研究揭示了突变p53如何促进乳腺癌转移的新见解。
    Tumor-associated p53 mutations induce activities different from wild-type p53, thus causing loss of the protein\'s tumor inhibition function. The cells carrying p53 mutations have more aggressive characteristics related to invasion, metastasis, proliferation, and cell survival. By comparing the gene expression profiles of mutant p53 (mutp53) and mutp53 silenced cohorts, we found that FOS-related antigen-1 (FRA-1), which is encoded by FOSL1, is a potential effector of mutp53-mediated metastasis. We demonstrate that the expression of FRA-1, a gatekeeper of mesenchymal-epithelial transition, is elevated in the presence of p53 mutations. Mechanistically, mutant p53 cooperates with the transcription factor ELK1 in binding and activating the promoter of FOSL1, thus fostering lung metastasis. This study reveals new insights into how mutant p53 contributes to metastasis in breast cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 背景:TP53基因的突变是人类癌症遗传改变中最常见的,导致突变p53蛋白(mutp53)的形成。Mutp53促进增殖,迁移,入侵,和癌细胞的转移。不仅由于mutp53引起了肿瘤发生的开始,而且还发生了癌细胞对化学疗法和放射疗法的抗性。这篇综述旨在总结和讨论突变p53在癌细胞中的肿瘤发生,并介绍目前在临床前和临床阶段正在评估的各种突变p53抑制剂。在靶向p53错义突变上诱导野生型构象的化合物,恢复或增强突变型p53的DNA结合,并抑制癌细胞的生长。此外,临床试验中突变型p53抑制剂的进展和发展被更新.
    结论:开发一种可以成功且有效地靶向突变型p53的癌症治疗方法的进展正处于发展的边缘。突变型p53蛋白不仅引发肿瘤发生,而且在癌细胞中引起对某些化疗或放疗的抗性。进一步支持癌细胞的存活并促进癌细胞的迁移和转移。在这方面,已经开发了许多突变型p53抑制剂,其中一些目前正在临床前水平进行评估,并已确定和讨论.迄今为止,APR-246是进入III期临床试验的最突出的一种。
    BACKGROUND: Mutations in the TP53 gene are the most common among genetic alterations in human cancers, resulting in the formation of mutant p53 protein (mutp53). Mutp53 promotes proliferation, migration, invasion, and metastasis in cancer cells. Not only does the initiation of oncogenesis ensue due to mutp53, but resistance towards chemotherapy and radiotherapy in cancer cells also occurs. This review aims to summarise and discuss the oncogenesis of mutant p53 in cancer cells and introduce the various mutant p53 inhibitors currently being evaluated at the pre-clinical and clinical stages. Compounds that induce the wild-type conformation on the targeted p53 missense mutation, restore or enhance the DNA binding of mutant p53, and inhibit cancer cells\' growth are highlighted. In addition, the progression and development of the mutant p53 inhibitors in clinical trials are updated.
    CONCLUSIONS: The progress of developing a cancer treatment that may successfully and efficiently target mutant p53 is on the verge of development. Mutant p53 proteins not only initiate oncogenesis but also cause resistance in cancer cells to certain chemo or radiotherapies, further endorse cancer cell survival and promote migration as well as metastasis of cancerous cells. With this regard, many mutant p53 inhibitors have been developed, some of which are currently being evaluated at the pre-clinical level and have been identified and discussed. To date, APR-246 is the most prominent one that has progressed to the Phase III clinical trial.
    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:TP53基因的突变是癌细胞中最常见的(占人类癌症的50-60%)遗传改变,表明野生型p53在致癌应激下调节细胞增殖和凋亡中的关键作用。大多数错义突变聚集在DNA结合核心结构域中,破坏DNA结合能力。然而,一些像Y220C这样的突变发生在DNA结合域之外,并且与p53结构不稳定有关.总的来说,这些突变的结果是p53中的单个氨基酸取代和大量功能失调的p53蛋白的产生,因此,允许细胞凋亡的逃逸和肿瘤生长的快速进展。因此,在肿瘤中靶向突变p53以恢复其野生型肿瘤抑制活性具有巨大的转化癌症研究潜力.已经用现代科学技术发现了通过恢复结构变化和/或DNA结合能力来重新激活突变型p53的各种分子。这些化合物包括小分子,各种肽,和植物化学物质。TP53蛋白长期以来被认为是潜在的靶标;然而,用于治疗目的的翻译仍处于起步阶段。该研究全面分析了来自Foeniculumvulgare(茴香)的小植物化学物质的治疗潜力,具有药物相似性和通过分子对接模拟重新激活突变型p53(Y220C)的能力。对接研究和稳定的分子动力学模拟显示胡格林(-8.6kcal/mol),视黄醇(-9.14kcal/mol),和3-硝基氟蒽(-8.43kcal/mol)显着结合突变位点,这表明针对Y220Cmutp53设计药物的可能性。该研究支持这些化合物用于进一步的基于动物的体内和体外研究以验证其功效。
    方法:为了药物再利用,最近,计算机模拟方法有机会排除许多化合物,这些化合物基于其结构部分和与靶大分子的相互作用作为药物的可能性较小。这里的研究利用通过Autodock4.2.6的分子对接和使用Schrodinger2021的分子动力学来寻找能够与突变的TP53蛋白反应的潜在治疗选择。
    BACKGROUND: The mutations in the TP53 gene are the most frequent (50-60% of human cancer) genetic alterations in cancer cells, indicating the critical role of wild-type p53 in the regulation of cell proliferation and apoptosis upon oncogenic stress. Most missense mutations are clustered in the DNA-binding core domain, disrupting DNA binding ability. However, some mutations like Y220C occur outside the DNA binding domain and are associated with p53 structure destabilization. Overall, the results of these mutations are single amino acid substitutions in p53 and the production of dysfunctional p53 protein in large amounts, consequently allowing the escape of apoptosis and rapid progression of tumor growth. Thus, therapeutic targeting of mutant p53 in tumors to restore its wild-type tumor suppression activity has immense potential for translational cancer research. Various molecules have been discovered with modern scientific techniques to reactivate mutant p53 by reverting structural changes and/or DNA binding ability. These compounds include small molecules, various peptides, and phytochemicals. TP53 protein is long thought of as a potential target; however, its translation for therapeutic purposes is still in its infancy. The study comprehensively analyzed the therapeutic potential of small phytochemicals from Foeniculum vulgare (Fennel) with drug-likeness and capability to reactivate mutant p53 (Y220C) through molecular docking simulation. The docking study and the stable molecular dynamic simulations revealed juglalin (- 8.6 kcal/mol), retinol (- 9.14 kcal/mol), and 3-nitrofluoranthene (- 8.43 kcal/mol) significantly bind to the mutated site suggesting the possibility of drug designing against the Y220C mutp53. The study supports these compounds for further animal based in vivo and in vitro research to validate their efficacy.
    METHODS: For the purposes of drug repurposing, recently in-silico methods have presented with opportunity to rule out many compounds which have less probability to act as a drug based on their structural moiety and interaction with the target macromolecule. The study here utilizes molecular docking via Autodock 4.2.6 and molecular dynamics using Schrodinger 2021 to find potential therapeutic options which are capable to reactive the mutated TP53 protein.
    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
    研究表明,野生型(wt)p53抑制癌基因c-Myc,而突变型(mut)p53可能使其反式激活,具有相反的行为,通常发生在wt或mutp53与促进致癌作用的分子/途径的串扰中。即使有报道称mutp53维持c-Myc,c-Myc是否可以反过来影响mutp53表达仍有待研究。在这项研究中,我们发现c-Myc的药理或基因抑制下调mutp53,细胞存活受损,胰腺癌细胞DNA损伤增加.在分子水平上,我们观察到c-Myc抑制降低了甲羟戊酸激酶(MVK)的表达,一种属于甲羟戊酸途径的分子,根据以前的发现,可以控制mutp53的稳定性,从而有助于癌细胞的存活。总之,这项研究揭示了致癌基因之间的另一个犯罪联盟,如c-Myc和mutp53,在肿瘤发生中起关键作用。
    It has been shown that wild-type (wt)p53 inhibits oncogene c-Myc while mutant (mut)p53 may transactivate it, with an opposite behavior that frequently occurs in the crosstalk of wt or mutp53 with molecules/pathways promoting carcinogenesis. Even if it has been reported that mutp53 sustains c-Myc, whether c-Myc could in turn influence mutp53 expression remains to be investigated. In this study, we found that pharmacological or genetic inhibition of c-Myc downregulated mutp53, impaired cell survival and increased DNA damage in pancreatic cancer cells. At the molecular level, we observed that c-Myc inhibition reduced the expression of mevalonate kinase (MVK), a molecule belonging to the mevalonate pathway that-according to previous findings-can control mutp53 stability, and thus contributes to cancer cell survival. In conclusion, this study unveils another criminal alliance between oncogenes, such as c-Myc and mutp53, that plays a key role in oncogenesis.
    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
    与传统的闭壳荧光团相比,自由基阳离子为开发小尺寸且易于制备的红色至NIR荧光团提供了机会。然而,文献中报道的大多数自由基阳离子在水溶液中的稳定性差并且几乎是不发射的。为了应对这一挑战,我们在此开发了深红色发射和水稳定的吡咯自由基阳离子P·-DPA-Zn,可以很容易地通过空气氧化从P-DPA-Zn中生成。深红色发射P·-DPA-Zn可用于成像引导的线粒体靶向将Zn2递送到癌细胞中,以促进突变型p53蛋白降解并消除mutp53表现出的功能获得,包括降低化疗耐药性,抑制癌细胞迁移,减少肿瘤细胞集落和球体形成。因此,水稳定的深红色发射性吡咯自由基阳离子有望用于癌症治疗应用。
    Compared with conventional closed-shell fluorophores, radical cations provide an opportunity for development of red-to-NIR fluorophores with small sizes and easy preparation. However, most radical cations reported in the literature suffer from poor stability in water solution and are almost non-emissive. To tackle this challenge, we herein develop a deep-red-emissive and water-stable pyrrole radical cation P⋅+ -DPA-Zn, which can be easily generated from P-DPA-Zn by air oxidation. The deep-red-emissive P⋅+ -DPA-Zn can be used for imaging-guided mitochondria-targeted delivery of Zn2+ into cancer cells to promote mutant p53 proteins degradation and abrogate mutp53-manifested gain of function, including reduced chemotherapy resistance, inhibited cancer cell migration, decreased tumor cell colony and sphere formation. The water-stable and deep-red emissive pyrrole radical cation is thus promising for cancer theranostic applications.
    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
    HDAC抑制剂(HDACi)代表有前途的抗癌治疗,由于组蛋白和非组蛋白蛋白的乙酰化在癌症中通常失调,并有助于癌症的发生和进展。HDACi也有报道增加DNA损伤剂的细胞毒性,如辐射或顺铂。在这项研究中,我们发现运输安全管理局和,甚至更有效,VPA与AZD2461、PARP1、2和3抑制剂(PARPi)协同诱导DNA损伤并降低胰腺癌细胞存活。在分子水平上,VPA和TSA下调CHK1和RAD51,这与mutp53和HSP70之间的串扰中断有关。此外,VPA和较小程度的TSA重新激活了这些细胞中的wtp53,这有助于CHK1和RAD51的减少。这些结果表明,HDACi和PARPi的组合可能会改善胰腺癌的治疗,它仍然是最具侵袭性和抗治疗性的癌症之一。
    HDAC inhibitors (HDACi) represent promising anti-cancer treatments, as the acetylation of histone and non-histone proteins is often dysregulated in cancer and contributes to cancer onset and progression. HDACi have been also reported to increase the cytotoxicity of DNA-damaging agents, such as radiation or cisplatin. In this study, we found that TSA and, even more effectively, VPA synergized with AZD2461, PARP1, 2 and 3 inhibitor (PARPi) to induce DNA damage and reduce pancreatic cancer cell survival. At a molecular level, VPA and TSA down-regulated CHK1 and RAD51, which is correlated with the interruption of the cross-talk between mutp53 and HSP70. Moreover, VPA and to a lesser extent TSA reactivated wtp53 in these cells, which contributed to CHK1 and RAD51 reduction. These findings suggest that the combination of HDACi and PARPi might improve the treatment of pancreatic cancer, which remains one of the most aggressive and therapy-resistant 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
    p53 is the most frequently mutated or inactivated gene in cancer, as its activity is not reconcilable with tumor onset and progression. Moreover, mutations in the p53 gene give rise to mutant proteins such as p53-R273H that, besides losing the wild type p53 (wtp53) capacity to safeguard genome integrity, may promote carcinogenesis, mainly due to its crosstalk with pro-oncogenic pathways. Interestingly, the activation of oncogenic pathways is interconnected with reactive oxygen species (ROS) and the release of pro-inflammatory cytokines that contribute to create an inflammatory/pro-tumorigenic milieu. In this study, based on experiments involving p53-R273H silencing and transfection, we showed that this mutant p53 (mutp53) promoted cancer cell survival by increasing intracellular ROS level and pro-inflammatory/immune suppressive cytokine release, activating mTOR, reducing autophagy and mitophagy and downregulating uncoupling protein 2 (UCP2). Interestingly, p53-R273H transfection into cancer cells carrying wtp53 induced none of these effects and resulted in p21 upregulation. This suggests that wtp53 may counteract several pro-tumorigenic activities of p53-R273H and this could explain the lower aggressiveness of cancers carrying heterozygous mutp53 in comparison to those harboring homozygous mutp53.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号