Proto-Oncogene Proteins c-mdm2

原癌基因蛋白质 c - mdm2
  • 文章类型: Journal Article
    背景:环状RNA(circularRNAs,circRNAs)已被证明与肿瘤发生和发展有关。然而,circGLIS3(hsa_circ_0002874)在前列腺癌(PCa)中的作用尚未见报道。
    方法:通过对公共数据集的综合分析确定候选circRNA,PCa细胞系,和组织数据。一系列的细胞功能检测,包括CCK-8,集落形成,伤口愈合,和transwell测定进行。随后,RNA测序,RNA免疫沉淀,甲基化RNA免疫沉淀,microRNA下拉,荧光素酶报告分析,和蛋白质印迹用于探索潜在的分子机制。此外,建立异种移植瘤小鼠模型以阐明circGLIS3的功能。
    结果:CircGLIS3,来自亲本GLIS3基因的外显子2,在PCa中被鉴定为与生化复发密切相关的新型致癌circRNA。其表达水平在PCa组织和细胞系以及恩杂鲁胺高抗性细胞中上调。细胞功能测定显示,circGLIS3促进PCa细胞增殖,迁移,和入侵。METTL3介导的N6-甲基腺苷(m6A)修饰通过增强其稳定性来维持其上调。机械上,CircGLIS3激活miR-661上调MDM2,从而调节p53信号通路促进细胞增殖,迁移,和入侵。此外,体外和体内实验,circGLIS3的敲减改善了PCa细胞对ARSI治疗如恩杂鲁胺的反应.
    结论:METTL3介导的m6A修饰circGLIS3通过miR-661/MDM2轴调节p53信号通路,从而促进PCa进展。同时,这项研究揭示了ARSI治疗PCa的一个有希望的潜在靶点.
    BACKGROUND: Circular RNAs (circRNAs) have been shown to be involved in tumorigenesis and progression. However, the role of circGLIS3 (hsa_circ_0002874) in prostate cancer (PCa) has yet not been reported.
    METHODS: Candidate circRNA were determined through comprehensive analysis of public datasets, PCa cell lines, and tissues data. A series of cellular functional assays, including CCK-8, colony formation, wound healing, and transwell assays were performed. Subsequently, RNA sequencing, RNA immunoprecipitation, methylated RNA immunoprecipitation, microRNA pulldown, luciferase reporter assay, and western blot were used to explore the underlying molecular mechanisms. Moreover, the xenograft tumor mouse model was established to elucidate the function of circGLIS3.
    RESULTS: CircGLIS3, derived from exon 2 of the parental GLIS3 gene, was identified as a novel oncogenic circRNA in PCa that was closely associated with the biochemical recurrence. Its expression levels were upregulated in PCa tissues and cell lines as well as enzalutamide high-resistant cells. The cellular functional assays revealed that circGLIS3 promoted PCa cell proliferation, migration, and invasion. METTL3-mediated N6-methyladenosine (m6A) modification maintained its upregulation by enhancing its stability. Mechanically, CircGLIS3 sponged miR-661 to upregulate MDM2, thus regulating the p53 signaling pathway to promote cell proliferation, migration, and invasion. Furthermore, in vitro and in vivo experiments, the knockdown of circGLIS3 improved the response of PCa cells to ARSI therapies such as enzalutamide.
    CONCLUSIONS: METTL3-mediated m6A modification of circGLIS3 regulates the p53 signaling pathway via the miR-661/MDM2 axis, thereby facilitating PCa progression. Meanwhile, this study unveils a promising potential target for ARSI therapy for PCa.
    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:根据世界卫生组织,乳腺癌被广泛认为是几乎普遍的癌症相关死亡的第一或第二原因。细胞周期破坏,以细胞周期蛋白的不受控制的表达形式或由于在负调节蛋白(CDK抑制剂)中的悬浮,被发现会导致乳腺癌.Palbociclib作为特异性CDK4/6抑制剂用于治疗ER+转移性癌症。在这项研究中,我们正在研究palbociclib对乳腺癌细胞的影响,并评估该药物治疗后作为miR-141靶基因的一些细胞周期相关基因的表达变化.我们使用MCF7作为功能性雌激素和非侵入性和MDA-MB-231细胞系作为三阴性类型的乳腺癌和更具侵略性的模型。
    方法:MCF7和MDA-MB-231细胞系在DMEM培养基中培养。计数细胞和测量活力后,使用从MTT获得的IC50以不同剂量施用Palbociclib,在24和72小时的两个时间点给予治疗。从未处理和处理的细胞中提取RNA,最后将RNA转化为cDNA。通过实时PCR研究基因表达变化。使用GraphPadPrism5.01软件进行数据管理和分析。
    结论:在研究的基因中,E2F3基因在任何细胞系和时间点均未受到Palbociclib的显着影响。此外,CCNE1基因的表达被显著抑制。该药物似乎不能显著降低三阴性(MDA-MB-231)癌细胞中MDM2基因的表达;然而,在腔A(MCF-7)细胞中观察到减少。CDKN2A和miR-141基因表达在治疗后显著增加,这可以与palbociclib在增殖抑制方面一致。
    BACKGROUND: According to WHO, Breast cancer is widely considered to be the first or second cause of cancer-related death almost universally. Cell cycle disruption, either in the form of uncontrolled expression of cyclins or because of the suspension in negative regulatory proteins (CDK inhibitors), was found to cause breast cancer. Palbociclib as specific CDK4/6 inhibitor is used for the treatment of ER+ metastatic cancers. In this study, we are looking to investigate the effect of palbociclib on breast cancer cells and evaluate the changes in the expression of some genes involved in the cell cycle as target genes of miR-141 after treatment with this drug. We used MCF7 as functional estrogen and non-invasive and MDA-MB-231 cell lines as triple-negative type of breast cancer and a model for more aggressive.
    METHODS: MCF7 and MDA-MB-231 cell lines were cultured in DMEM medium. After counting cells and measuring viability, Palbociclib was administered at varying doses using the IC50 obtained from MTT, with the treatment given at two time points of 24 and 72 hours. RNA was extracted from untreated and treated cells and RNAs were converted to cDNA in the end. Gene expression changes were investigated by real-time PCR. Data management and analysis were conducted using GraphPad Prism 5.01 software.
    CONCLUSIONS: Among investigated genes, E2F3 gene was not significantly affected by Palbociclib in any of cell lines and time points. Besides, the expression of CCNE1 gene was significantly suppressed. It seems this drug was unable to reduce the expression of MDM2 gene significantly in triple negative (MDA-MB-231) cancer cells; however, a decrease was observed in luminal A (MCF-7) cells. CDKN2A and miR-141 genes expression increased significantly after treatment which can be aligned with palbociclib in proliferation inhibition.
    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
    青少年骨化性纤维瘤(JOF)是颌面部骨骼的罕见良性纤维骨性病变(BFOL),具有局部侵袭性和高复发率。鼠DoubleMinute2(MDM2)是位于染色体12(12q13-15)的癌基因,其抑制肿瘤抑制基因TP53。MDM2基因位点扩增的存在是评估某些肉瘤的有用分子辅助手段,包括低级别髓内骨肉瘤(LGIOS)。JOF和LGIOS具有一些重叠的临床和组织病理学特征。这项研究的目的是使用荧光原位杂交(FISH)评估一系列JOF是否存在MDM2基因位点扩增。
    方法:经IRB批准,对佛罗里达健康大学口腔病理学和外科病理学活检服务档案的机构档案进行了检索.病例由口腔病理学住院医师重新评估,口腔颌面病理学家,还有骨骼和软组织病理学家.选择诊断一致的病例(n=9)进行MDM2检测。通过FISH测试MDM2基因位点扩增被应用于所有检索的病例。
    结果:所有病例经FISH检测MDM2基因位点扩增均为阴性。
    结论:在我们的小系列中,JOF未显示MDM2基因位点异常,LGIOS的特征。这一发现表明JOF具有独特的潜在发病机理。如果在更大的系列中得到证实,在特征重叠的病例中或在活检材料最少的情况下,这些发现可能有助于区分这两个实体.
    Juvenile ossifying fibroma (JOF) is an uncommon benign fibro-osseous lesion (BFOL) of the maxillofacial bones with a locally aggressive nature and a high recurrence rate. Murine Double Minute 2 (MDM2) is an oncogene located at chromosome 12 (12q13-15) that inhibits the tumor suppressor gene TP53. The presence of MDM2 gene locus amplification is a useful molecular adjunct in the evaluation of some sarcomas, including low-grade intramedullary osteosarcoma (LGIOS). JOF and LGIOS have some overlapping clinical and histopathological features. The aim of this study is to evaluate a series of JOF for the presence of MDM2 gene locus amplification using fluorescence in-situ hybridization (FISH).
    METHODS: With IRB approval, a search of the institutional files of the archives of the Oral Pathology and Surgical Pathology biopsy services at the University of Florida Health was performed. The cases were re-evaluated by an oral pathology resident, an oral and maxillofacial pathologist, and a bone and soft tissue pathologist. Cases with consensus in diagnosis were selected (n = 9) for MDM2 testing. Testing by FISH for MDM2 gene locus amplification was applied to all retrieved cases.
    RESULTS: The examined cases were all negative for MDM2 gene locus amplification via FISH testing.
    CONCLUSIONS: In our small series, JOF did not demonstrate MDM2 gene locus abnormality, a characteristic of LGIOS. This finding suggests that JOF has a distinct underlying pathogenesis. If confirmed in a larger series, these findings may be useful in distinguishing these two entities in cases with overlapping features or when minimal biopsy material is available.
    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
    抑制MDM2-p53相互作用被认为是癌症治疗的有效模式。在我们目前的研究中,高斯加速分子动力学(GaMD),深度学习(DL),和结合自由能计算组合在一起以探测非肽抑制剂K23和0Y7以及肽抑制剂PDI6W和PDI与MDM2的结合机制。基于GaMD轨迹的DL方法成功识别了重要的功能域,主要位于螺旋α2和α2',以及α2和α2之间的β链和环。GaMD模拟的后处理分析表明,抑制剂结合会高度影响MDM2的结构灵活性和集体运动。分子力学广义Born表面积(MM-GBSA)和溶剂化相互作用能(SIE)的计算不仅表明计算的束缚自由能的排序与实验结果一致,但也验证了vanderWalls相互作用是导致抑制剂-MDM2结合的主要力量。我们的发现还表明,与非肽抑制剂相比,肽抑制剂与MDM2产生更多的相互作用接触。主成分分析(PCA)和自由能景观(FEL)分析表明,哌啶酮抑制剂0Y7对MDM2的自由能曲线显示出最明显的影响,哌啶酮抑制剂沿主要特征向量显示出更高的波动幅度。通过基于残基的自由能估计揭示的MDM2的热点为针对MDM2的药物设计提供了目标位点。这项研究有望为开发MDM2家族成员的选择性抑制剂提供有用的理论帮助。
    Inhibiting MDM2-p53 interaction is considered an efficient mode of cancer treatment. In our current study, Gaussian-accelerated molecular dynamics (GaMD), deep learning (DL), and binding free energy calculations were combined together to probe the binding mechanism of non-peptide inhibitors K23 and 0Y7 and peptide ones PDI6W and PDI to MDM2. The GaMD trajectory-based DL approach successfully identified significant functional domains, predominantly located at the helixes α2 and α2\', as well as the β-strands and loops between α2 and α2\'. The post-processing analysis of the GaMD simulations indicated that inhibitor binding highly influences the structural flexibility and collective motions of MDM2. Calculations of molecular mechanics-generalized Born surface area (MM-GBSA) and solvated interaction energy (SIE) not only suggest that the ranking of the calculated binding free energies is in agreement with that of the experimental results, but also verify that van der Walls interactions are the primary forces responsible for inhibitor-MDM2 binding. Our findings also indicate that peptide inhibitors yield more interaction contacts with MDM2 compared to non-peptide inhibitors. Principal component analysis (PCA) and free energy landscape (FEL) analysis indicated that the piperidinone inhibitor 0Y7 shows the most pronounced impact on the free energy profiles of MDM2, with the piperidinone inhibitor demonstrating higher fluctuation amplitudes along primary eigenvectors. The hot spots of MDM2 revealed by residue-based free energy estimation provide target sites for drug design toward MDM2. This study is expected to provide useful theoretical aid for the development of selective inhibitors of MDM2 family members.
    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:哮喘的复杂性,以气道炎症和重塑为标志,受缺氧条件的影响。本研究的重点是低氧诱导因子-1α(HIF-1α)和P53泛素化在哮喘加重中的作用。
    方法:采用高通量测序和生物信息学方法鉴定与哮喘进展相关的基因,重点是GO和KEGG通路分析。建立了哮喘小鼠模型,分离气道平滑肌细胞(ASMCs),建立体外缺氧模型。细胞活力,扩散,迁移,并对细胞凋亡进行了评估,以及ELISA和苏木精和伊红(H&E)染色。
    结果:在体内和体外哮喘模型中均观察到HIF-1α的显著增加。HIF-1α上调增强ASMC活力,扩散,和移民,在减少细胞凋亡的同时,主要通过MDM2促进P53泛素化。体内研究显示炎症细胞浸润增加,气道结构改变,通过抑制剂IDF-11,774缓解。
    结论:该研究强调了HIF-1α-MDM2-P53轴在哮喘中的关键作用,表明其作为治疗干预目标的潜力。研究结果表明,调节该途径可以为治疗复杂的哮喘呼吸系统疾病提供新的途径。
    BACKGROUND: Asthma\'s complexity, marked by airway inflammation and remodeling, is influenced by hypoxic conditions. This study focuses on the role of Hypoxia-Inducible Factor-1 Alpha (HIF-1α) and P53 ubiquitination in asthma exacerbation.
    METHODS: High-throughput sequencing and bioinformatics were used to identify genes associated with asthma progression, with an emphasis on GO and KEGG pathway analyses. An asthma mouse model was developed, and airway smooth muscle cells (ASMCs) were isolated to create an in vitro hypoxia model. Cell viability, proliferation, migration, and apoptosis were assessed, along with ELISA and Hematoxylin and Eosin (H&E) staining.
    RESULTS: A notable increase in HIF-1α was observed in both in vivo and in vitro asthma models. HIF-1α upregulation enhanced ASMCs\' viability, proliferation, and migration, while reducing apoptosis, primarily via the promotion of P53 ubiquitination through MDM2. In vivo studies showed increased inflammatory cell infiltration and airway structural changes, which were mitigated by the inhibitor IDF-11,774.
    CONCLUSIONS: The study highlights the critical role of the HIF-1α-MDM2-P53 axis in asthma, suggesting its potential as a target for therapeutic interventions. The findings indicate that modulating this pathway could offer new avenues for treating the complex respiratory disorder of asthma.
    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
    种系TP53致病变体可导致称为Li-Fraumeni(LFS)的癌症易感性综合征。影响其活性的变体可以驱动肿瘤发生改变p53途径,它们的鉴定对于评估个体风险至关重要。本研究探讨了TP53错义变体对其转录因子活性的功能影响。我们选择了七个TP53错义变体(c.129G>C,c.320A>G,c.417G>T,c.460G>A,C,522G>T,c.589G>A和c.997C>T)在有LFS风险的巴西家庭中确定。通过定点诱变产生变体并转染到SK-OV-3细胞中以评估其转录激活能力。变体K139N和V197M在TP53依赖性荧光素酶报告基因测定中显示显著降低的反式激活活性。此外,K139N对CDKN1A和MDM2的表达产生负面影响,并且在辐射诱导的DNA损伤后对GADD45A和PMAIP1的影响有限。变体V197M在所评估的所有靶基因中表现出功能影响和Ser15磷酸化的丧失。K139N和V197M变体在照射后呈现p21水平的降低。我们的数据显示K139N和V197M对p53功能有负面影响,支持将其分类为致病变体。这强调了对种系TP53错义变异进行功能研究的重要性,这些变异被分类为不确定意义的变异,以确保对LFS相关癌症风险的适当管理。
    Germline TP53 pathogenic variants can lead to a cancer susceptibility syndrome known as Li-Fraumeni (LFS). Variants affecting its activity can drive tumorigenesis altering p53 pathways and their identification is crucial for assessing individual risk. This study explored the functional impact of TP53 missense variants on its transcription factor activity. We selected seven TP53 missense variants (c.129G > C, c.320A > G, c.417G > T, c.460G > A, c,522G > T, c.589G > A and c.997C > T) identified in Brazilian families at-risk for LFS. Variants were created through site-directed mutagenesis and transfected into SK-OV-3 cells to assess their transcription activation capabilities. Variants K139N and V197M displayed significantly reduced transactivation activity in a TP53-dependent luciferase reporter assay. Additionally, K139N negatively impacted CDKN1A and MDM2 expression and had a limited effect on GADD45A and PMAIP1 upon irradiation-induced DNA damage. Variant V197M demonstrated functional impact in all target genes evaluated and loss of Ser15 phosphorylation. K139N and V197M variants presented a reduction of p21 levels after irradiation. Our data show that K139N and V197M negatively impact p53 functions, supporting their classification as pathogenic variants. This underscores the significance of conducting functional studies on germline TP53 missense variants classified as variants of uncertain significance to ensure proper management of LFS-related cancer risks.
    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
    肿瘤抑制因子vonHippel-Lindau,pVHL,是一种多方面的蛋白质。一种功能是停靠在缺氧诱导转录因子(HIF)上,并通过泛素化来招募更大的蛋白质复合物,使HIF不稳定。防止血管生成和肿瘤发展。pVHL还与肿瘤抑制因子p53结合以激活特异性p53靶基因。癌基因Mdm2通过将nedd8与pVHL缀合而损害p53-pVHL复合物的形成和下游基因的激活。虽然Mdm2可以影响p53和pVHL,pVHL如何影响Mdm2尚不清楚。像p53体细胞突变,在肾透明细胞癌(RCC)中常见的pVHL中,点突变很明显。在RCC患者中,Mdm2水平升高,我们检查了Mdm2和pVHL之间是否存在关系。TCGA和DepMap分析显示mdm2基因表达在RCC中升高,具有vhl点突变或拷贝数丢失。在pVHL重建或缺失的同源匹配RCC或MEF细胞系中,在pVHL存在下Mdm2降低。此外,通过使用遗传和药理学方法进行分析,我们发现pVHL通过阻断MAPK-Ets信号通路抑制Mdm2基因表达,并阻断Akt介导的Mdm2磷酸化和稳定。Mdm2抑制导致p53-p21途径的增加以阻碍细胞生长。这一发现表明pVHL如何通过调节信号传导途径来限制细胞生长,从而间接影响Mdm2的功能。
    The tumor suppressor von Hippel-Lindau, pVHL, is a multifaceted protein. One function is to dock to the hypoxia-inducible transcription factor (HIF) and recruit a larger protein complex that destabilizes HIF via ubiquitination, preventing angiogenesis and tumor development. pVHL also binds to the tumor suppressor p53 to activate specific p53 target genes. The oncogene Mdm2 impairs the formation of the p53-pVHL complex and activation of downstream genes by conjugating nedd8 to pVHL. While Mdm2 can impact p53 and pVHL, how pVHL may impact Mdm2 is unclear. Like p53 somatic mutations, point mutations are evident in pVHL that are common in renal clear cell carcinomas (RCC). In patients with RCC, Mdm2 levels are elevated, and we examined whether there was a relationship between Mdm2 and pVHL. TCGA and DepMap analysis revealed that mdm2 gene expression was elevated in RCC with vhl point mutations or copy number loss. In pVHL reconstituted or deleted isogenetically match RCC or MEF cell lines, Mdm2 was decreased in the presence of pVHL. Furthermore, through analysis using genetic and pharmacological approaches, we show that pVHL represses Mdm2 gene expression by blocking the MAPK-Ets signaling pathway and blocks Akt-mediated phosphorylation and stabilization of Mdm2. Mdm2 inhibition results in an increase in the p53-p21 pathway to impede cell growth. This finding shows how pVHL can indirectly impact the function of Mdm2 by regulating signaling pathways to restrict cell growth.
    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:小鼠双分2同源物(MDM2)在血液恶性肿瘤中下调p53活性中起关键作用,其过度表达与不良结果相关。
    方法:这项1期研究评估了MDM2抑制剂milademetan的不同给药方案作为单药和联合阿扎胞苷(AZA)治疗复发性或难治性急性髓系白血病或高危骨髓增生异常综合征患者的安全性和有效性。
    结果:74例患者(单药治疗,n=57;milademetan-AZA组合,n=17)被处理。milademetan的最大耐受剂量为160mg,每天一次,用于28天周期的前14-21天作为单一疗法,并在第5-14天与AZA组合。剂量限制性毒性是胃肠道,疲劳,或肾脏/电解质异常。在单药治疗和AZA联合治疗组中,82.5%和64.7%的参与者发生了与milademetan相关的治疗紧急不良事件,分别。单药治疗组的两名参与者(4.2%)达到完全缓解(CR),1例(2.1%)达到CR,血细胞计数恢复不全(CRi)。两名参与者(13.3%)在组合组中获得CRi。新的TP53突变,仅在米拉德美坦单药治疗期间检测到,通过液滴数字聚合酶链反应发现预先存在于标准检测频率以下。
    结论:Milademetan在该人群中的耐受性相对较好;然而,尽管有活动的信号,临床疗效微乎其微。
    BACKGROUND: Mouse double minute-2 homolog (MDM2) plays a key role in downregulating p53 activity in hematologic malignancies, and its overexpression is associated with poor outcomes.
    METHODS: This phase 1 study assessed the safety and efficacy of different dosing regimens of the MDM2 inhibitor milademetan as monotherapy and in combination with azacitidine (AZA) in patients with relapsed or refractory acute myeloid leukemia or high-risk myelodysplastic syndromes.
    RESULTS: Seventy-four patients (monotherapy, n = 57; milademetan-AZA combination, n = 17) were treated. The maximum tolerated dose of milademetan was 160 mg once daily given for the first 14-21 days of 28-day cycles as monotherapy and on Days 5-14 in combination with AZA. Dose-limiting toxicities were gastrointestinal, fatigue, or renal/electrolyte abnormalities. Treatment-emergent adverse events related to milademetan occurred in 82.5% and 64.7% of participants in the monotherapy and AZA combination arms, respectively. Two participants (4.2%) in the monotherapy arm achieved complete remission (CR), and 1 (2.1%) achieved CR with incomplete blood count recovery (CRi). Two participants (13.3%) achieved CRi in the combination arm. New TP53 mutations, detected only during milademetan monotherapy, were found pre-existing below standard detection frequency by droplet digital polymerase chain reaction.
    CONCLUSIONS: Milademetan was relatively well tolerated in this population; however, despite signals of activity, clinical efficacy was minimal.
    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
    Hdm2和Hdm4是调控肿瘤抑制蛋白的结构同源物,.由于一些肿瘤表达野生型p53,Hdm2和Hdm4是抗癌药物的合理靶点,尤其是在表达野生型p53的肿瘤中。Hdm4可以增强和拮抗Tp53的活性,从而在调节p53的活性和稳定性中起关键作用。此外,Hdm2和Hdm4在许多癌症中过度表达,一些表达野生型Tp53。由于实验证据表明野生型Tp53的激活可以通过一些检查点抑制剂增强抗肿瘤活性,针对Hdm2和Hdm4的药物可能是联合检查点抑制剂免疫疗法的有力候选药物.然而,其他证据表明,Hdm2和Hdm4的过表达可能表明对免疫检查点抑制剂的反应较差。这些发现需要仔细检查和审查。在这篇文章中,将对Hdm2/Hdm4伙伴关系进行全面分析。此外,本文将介绍目前针对Hdm2/Hdm4/Tp53合作伙伴关系的分子的药物开发进展。
    Hdm2 and Hdm4 are structural homologs that regulate the tumor suppressor protein, p53. Since some tumors express wild-type p53, Hdm2 and Hdm4 are plausible targets for anticancer drugs, especially in tumors that express wild-type p53. Hdm4 can enhance and antagonize the activity of Tp53, thereby playing a critical role in the regulation of p53\'s activity and stability. Moreover, Hdm2 and Hdm4 are overexpressed in many cancers, some expressing wild-type Tp53. Due to experimental evidence suggesting that the activation of wild-type Tp53 can augment the antitumor activity by some checkpoint inhibitors, drugs targeting Hdm2 and Hdm4 may be strong candidates for combining with checkpoint inhibitor immunotherapy. However, other evidence suggests that the overexpression of Hdm2 and Hdm4 may indicate poor response to immune checkpoint inhibitors. These findings require careful examination and scrutiny. In this article, a comprehensive analysis of the Hdm2/Hdm4 partnership will be conducted. Furthermore, this article will address the current progress of drug development regarding molecules that target the Hdm2/Hdm4/Tp53 partnership.
    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
    在早期胚胎发育过程中,从全能性到多能性的转变是正确发展的基本和关键过程。然而,管理这一过渡的监管机制仍然难以捉摸。这里,我们进行了全面的全基因组CRISPR/Cas9筛选,以研究小鼠胚胎干细胞(mESCs)中的2细胞样细胞(2CLCs)表型.这项努力导致确定了在此过渡期间在确定细胞命运中起关键作用的十种调节剂。值得注意的是,我们的研究表明Mdm2是2CLC的显著负调节因子,由于Mdm2的扰动导致2CLC的比例更高。Mdm2似乎通过其对细胞周期进程和H3K27me3表观遗传修饰的影响来影响细胞命运。总之,我们的CRISPR/Cas9筛选的结果发现了几个在不同水平上调节全能性和多能性具有不同功能的基因,为未来分子研究的潜在目标提供了宝贵的资源。
    During early embryonic development, the transition from totipotency to pluripotency is a fundamental and critical process for proper development. However, the regulatory mechanisms governing this transition remain elusive. Here, we conducted a comprehensive genome-wide CRISPR/Cas9 screen to investigate the 2-cell-like cells (2CLCs) phenotype in mouse embryonic stem cells (mESCs). This effort led to the identification of ten regulators that play a pivotal role in determining cell fate during this transition. Notably, our study revealed Mdm2 as a significant negative regulator of 2CLCs, as perturbation of Mdm2 resulted in a higher proportion of 2CLCs. Mdm2 appears to influence cell fate through its impact on cell cycle progression and H3K27me3 epigenetic modifications. In summary, the results of our CRISPR/Cas9 screen have uncovered several genes with distinct functions in regulating totipotency and pluripotency at various levels, offering a valuable resource for potential targets in future molecular studies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号