AKR1C3

AKR1C3
  • 文章类型: Journal Article
    AST-3424是一种新型的高肿瘤选择性前药。AST-3424被AKR1C3激活以释放有毒的双烷基化部分,AST2660。在这项研究中,我们在体外和体内研究了DNA修复在AST-3424介导的药理活性中的重要作用。我们在这里表明,AST-3424作为单一治疗剂有效对抗癌细胞诱导细胞毒性,DNA损伤,细胞凋亡和细胞周期停滞在G2期以剂量和AKR1C3依赖性方式在p53高的H460(RRID:CVCL_0459)和p53缺陷的HT-29细胞(RRID:CVCL_0320)。G2检查点的消除剂与AST-3424的组合仅在HT-29中具有协同作用,而在H460细胞中不具有协同作用。AST-3424在HT-29细胞中的活性增强是由于通过减弱细胞周期G2停滞和降低RAD51表达而受损的DNA修复能力。此外,我们利用一个BRCA2缺陷细胞系和两个具有BRCA有害突变的PDX模型来研究AST-3424活性的增加.结果表明,AST-3424在缺乏DNA修复蛋白BRCA2的细胞中表现出增强的体外细胞毒性和优异且持久的体内抗肿瘤作用。总之,我们在这里报道,当DNA修复能力降低时,AST-3424的体外和体内活性可以进一步增强,从而为AST-3424在临床上的进一步评估提供了支持证据.
    AST-3424 is a novel and highly tumor-selective prodrug. AST-3424 is activated by AKR1C3 to release a toxic bis-alkylating moiety, AST 2660. In this study, we have investigated the essential role of DNA repair in AST-3424 mediated pharmacological activities in vitro and in vivo. We show here that AST-3424 is effective as a single therapeutic agent against cancer cells to induce cytotoxicity, DNA damage, apoptosis and cell cycle arrest at G2 phase in a dose- and AKR1C3-dependent manner in both p53-proficient H460 (RRID:CVCL_0459) and p53-deficient HT-29 cells (RRID:CVCL_0320). The combination of abrogators of G2 checkpoint with AST-3424 was only synergistic in HT-29 but not in H460 cells. The enhanced activity of AST-3424 in HT-29 cells was due to impaired DNA repair ability via the attenuation of cell cycle G2 arrest and reduced RAD51 expression. Furthermore, we utilized a BRCA2 deficient cell line and two PDX models with BRCA deleterious mutations to study the increased activity of AST-3424. The results showed that AST-3424 exhibited enhanced in vitro cytotoxicity and superior and durable in vivo anti-tumor effects in cells deficient of DNA repair protein BRCA2. In summary, we report here that when DNA repair capacity is reduced, the in vitro and in vivo activity of AST-3424 can be further enhanced, thus providing supporting evidence for the further evaluation of AST-3424 in the clinic.
    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:激活蛋白-1(AP-1)是一个转录因子家族,因其在肿瘤生物学中的广泛参与而受到越来越多的关注。然而,AP-1家族在肺癌演变过程中的作用尚不明确.FBJ鼠骨肉瘤病毒癌基因同源物B(FOSB),典型的AP-1家族成员,以前曾报道,作为一把神秘的双刃剑,在非小细胞肺癌(NSCLC)中扮演着令人困惑的两极分化的角色,其原因和意义值得进一步阐明。
    结果:基于来自TCGA数据库的大型NSCLC队列的生物信息学分析,我们目前的工作发现,众所周知的抑癌基因TP53是破译FOSB两侧的关键代码-其表达表明,携带野生型TP53的NSCLC患者预后为阳性,而携带突变型TP53的NSCLC患者预后为阴性.通过构建一组在不同状态下表达p53的同基因来源的NSCLC细胞,验证了FOSB表达在非小细胞肺癌人群中完全相反的预后效应,与TP53-R248Q突变位点的出现特别有意义。转录组测序表明,FOSB过表达在具有不同遗传背景的TP53和NSCLC细胞中引起多样化的转录组景观,结合RT-qPCR验证,PREX1(TP53-Null),IGFBP5(TP53-WT),AKR1C3和ALDH3A1(TP53-R248Q)分别被鉴定为FOSB的p53依赖性转录靶标。随后,在体外和体内证实了FOSB通过上述选择性转录靶标对NSCLC细胞肿瘤生物学的异质性影响。机制研究表明,野生型或突变型p53可能通过蛋白质-蛋白质相互作用引导FOSB识别并结合不同的启动子序列,以转录激活特定的靶基因。从而对NSCLC的进展和预后产生不同的影响。
    结论:FOSB表达有望作为NSCLC的新的预后生物标志物与TP53的给定遗传背景相结合,FOSB和p53之间的独特相互作用可能作为NSCLC的潜在干预靶点。
    BACKGROUND: Activator protein-1 (AP-1) represents a transcription factor family that has garnered growing attention for its extensive involvement in tumor biology. However, the roles of the AP-1 family in the evolution of lung cancer remain poorly characterized. FBJ Murine Osteosarcoma Viral Oncogene Homolog B (FOSB), a classic AP-1 family member, was previously reported to play bewilderingly two-polarized roles in non-small cell lung cancer (NSCLC) as an enigmatic double-edged sword, for which the reasons and significance warrant further elucidation.
    RESULTS: Based on the bioinformatics analysis of a large NSCLC cohort from the TCGA database, our current work found the well-known tumor suppressor gene TP53 served as a key code to decipher the two sides of FOSB - its expression indicated a positive prognosis in NSCLC patients harboring wild-type TP53 while a negative one in those harboring mutant TP53. By constructing a panel of syngeneically derived NSCLC cells expressing p53 in different statuses, the radically opposite prognostic effects of FOSB expression in NSCLC population were validated, with the TP53-R248Q mutation site emerging as particularly meaningful. Transcriptome sequencing showed that FOSB overexpression elicited diversifying transcriptomic landscapes across NSCLC cells with varying genetic backgrounds of TP53 and, combined with the validation by RT-qPCR, PREX1 (TP53-Null), IGFBP5 (TP53-WT), AKR1C3, and ALDH3A1 (TP53-R248Q) were respectively identified as p53-dependent transcriptional targets of FOSB. Subsequently, the heterogenous impacts of FOSB on the tumor biology in NSCLC cells via the above selective transcriptional targets were confirmed in vitro and in vivo. Mechanistic investigations revealed that wild-type or mutant p53 might guide FOSB to recognize and bind to distinct promoter sequences via protein-protein interactions to transcriptionally activate specific target genes, thereby creating disparate influences on the progression and prognosis in NSCLC.
    CONCLUSIONS: FOSB expression holds promise as a novel prognostic biomarker for NSCLC in combination with a given genetic background of TP53, and the unique interactions between FOSB and p53 may serve as underlying intervention targets for NSCLC.
    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
    人类aldo-keto还原酶家族1成员C3(AKR1C3)表达的改变与多种癌症的不良预后相关。铁凋亡抗性,和代谢性疾病。尽管具有临床意义,AKR1C3的内源性生化作用仍未完全确定。使用非靶向代谢组学,我们确定了由AKR1C3介导的主要转化,其中精胺氧化产物“精子粒”被还原为“精子醇”。“精子引起DNA损伤并激活DNA双链断裂反应,而精子醇在体外诱导自噬。AKR1C3还下调酰基-吡喃酮和吡喃酮-211抑制AKR1C3活性。通过G蛋白偶联受体配体筛选,我们确定吡喃酮-211也是半孤儿受体GPR84的有效激动剂。引人注目的是,哺乳动物脂肪酸合酶在体外产生酰基吡喃酮,这种生产由NADPH调节。一起来看,我们的研究支持AKR1C3在扩展的多胺途径中的调节作用,以及将脂肪酸合成和NADPH水平与GPR84信号传导联系起来的模型.
    Altered human aldo-keto reductase family 1 member C3 (AKR1C3) expression has been associated with poor prognosis in diverse cancers, ferroptosis resistance, and metabolic diseases. Despite its clinical significance, the endogenous biochemical roles of AKR1C3 remain incompletely defined. Using untargeted metabolomics, we identified a major transformation mediated by AKR1C3, in which a spermine oxidation product \"sperminal\" is reduced to \"sperminol.\" Sperminal causes DNA damage and activates the DNA double-strand break response, whereas sperminol induces autophagy in vitro. AKR1C3 also pulls down acyl-pyrones and pyrone-211 inhibits AKR1C3 activity. Through G protein-coupled receptor ligand screening, we determined that pyrone-211 is also a potent agonist of the semi-orphan receptor GPR84. Strikingly, mammalian fatty acid synthase produces acyl-pyrones in vitro, and this production is modulated by NADPH. Taken together, our studies support a regulatory role of AKR1C3 in an expanded polyamine pathway and a model linking fatty acid synthesis and NADPH levels to GPR84 signaling.
    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
    AKR1C3酶在激素和药物代谢中起着至关重要的作用,并与肝癌的异常表达有关,导致肿瘤进展和预后不良。HSA修饰的纳米粒子可以通过增强光动力疗法来调节肿瘤微环境,从而诱导肿瘤细胞凋亡和减轻缺氧。因此,通过构建HSA-RSVNPs载体,探索白藜芦醇对AKR1C3的潜在调控机制,对肝癌的治疗具有重要的理论和临床意义。这项研究的目的是研究通过负载白藜芦醇(RSV)在纳米材料HSA-RSVNPs(纳米颗粒)上靶向调节AKR1C3表达,以减轻肿瘤缺氧并抑制肝细胞癌(HCC)的进展。并探讨其分子机制。使用PubChem数据库和PharmMapper服务器筛选RSV的靶基因。通过GEO数据集分析HCC相关差异表达基因(DEGs),从GeneCards数据库中检索到相关基因,导致三个相交以获得候选DEG。对候选DEGs进行GO和KEGG富集分析,以分析受主要靶基因影响的潜在细胞功能和分子信号通路。使用cytohubba插件筛选按Degree排序的前10个目标基因,并进一步交叉LASSO和随机森林(RF)的结果以获得hub基因。进行hub基因的表达分析及对恶性肿瘤预后的预测。此外,使用PharmMapper构建药效团模型。使用AutoDockTools1.5.6软件进行分子对接模拟,并进行ROC曲线分析以确定核心靶标。通过选择合适的HCC细胞系进行体外细胞实验,用不同浓度的RSV治疗HCC细胞,或使用慢病毒沉默或过表达AKR1C3。CCK-8,克隆形成,流式细胞术,划痕实验,Transwell被用来测量癌细胞的活力,扩散,迁移,入侵,和细胞凋亡,分别。使用海马XF24分析仪分析细胞耗氧率。制备HSA-RSVNP,并对其表征和细胞毒性进行了评估。检测治疗后肝癌细胞的生物学功能变化。使用HepG2细胞系在小鼠中建立HCC皮下异种移植模型。经尾静脉注射HSA-RSVNP,使用控制组集,观察肿瘤生长的变化,NPs的肿瘤靶向,生物安全。TUNEL,在切除的肿瘤组织上进行Ki67和APC-缺氧探针染色以检测肿瘤细胞的增殖。凋亡,和缺氧。慢病毒用于沉默或过表达AKR1C3,同时通过尾静脉注射HSA-RSVNP,以评估AKR1C3对HCC进展中HSA-RSVNP调节的影响。生物信息学分析显示,AKR1C3是RSV调控HCC的重要靶基因,这与HCC患者的预后有关,并且表达上调。体外细胞实验表明,RSV显著抑制HCC细胞的呼吸代谢,抑制它们的扩散,迁移,侵袭和促进细胞凋亡。沉默AKR1C3进一步增强RSV对HCC细胞的毒性。纳米材料的表征和细胞毒性实验证明了HSA-RSVNP的成功构建,对肝癌细胞有较强的抑制作用。在体内,动物实验进一步证实,HSA-RSVNP靶向下调AKR1C3抑制HCC进展和肿瘤缺氧,同时表现出肿瘤靶向性和生物学安全性。HSA-RSVNPs靶向下调AKR1C3可减轻HCC肿瘤缺氧并抑制HCC进展。
    The enzyme AKR1C3 plays a crucial role in hormone and drug metabolism and is associated with abnormal expression in liver cancer, leading to tumor progression and poor prognosis. Nanoparticles modified with HSA can modulate the tumor microenvironment by enhancing photodynamic therapy to induce apoptosis in tumor cells and alleviate hypoxia. Therefore, exploring the potential regulatory mechanisms of resveratrol on AKR1C3 through the construction of HSA-RSV NPs carriers holds significant theoretical and clinical implications for the treatment of liver cancer. The aim of this study is to investigate the targeted regulation of AKR1C3 expression through the loading of resveratrol (RSV) on nanomaterials HSA-RSV NPs (Nanoparticles) in order to alleviate tumor hypoxia and inhibit the progression of hepatocellular carcinoma (HCC), and to explore its molecular mechanism. PubChem database and PharmMapper server were used to screen the target genes of RSV. HCC-related differentially expressed genes (DEGs) were analyzed through the GEO dataset, and relevant genes were retrieved from the GeneCards database, resulting in the intersection of the three to obtain candidate DEGs. GO and KEGG enrichment analyses were performed on the candidate DEGs to analyze the potential cellular functions and molecular signaling pathways affected by the main target genes. The cytohubba plugin was used to screen the top 10 target genes ranked by Degree and further intersected the results of LASSO and Random Forest (RF) to obtain hub genes. The expression analysis of hub genes and the prediction of malignant tumor prognosis were conducted. Furthermore, a pharmacophore model was constructed using PharmMapper. Molecular docking simulations were performed using AutoDockTools 1.5.6 software, and ROC curve analysis was performed to determine the core target. In vitro cell experiments were carried out by selecting appropriate HCC cell lines, treating HCC cells with different concentrations of RSV, or silencing or overexpressing AKR1C3 using lentivirus. CCK-8, clone formation, flow cytometry, scratch experiment, and Transwell were used to measure cancer cell viability, proliferation, migration, invasion, and apoptosis, respectively. Cellular oxygen consumption rate was analyzed using the Seahorse XF24 analyzer. HSA-RSV NPs were prepared, and their characterization and cytotoxicity were evaluated. The biological functional changes of HCC cells after treatment were detected. An HCC subcutaneous xenograft model was established in mice using HepG2 cell lines. HSA-RSV NPs were injected via the tail vein, with a control group set, to observe changes in tumor growth, tumor targeting of NPs, and biological safety. TUNEL, Ki67, and APC-hypoxia probe staining were performed on excised tumor tissue to detect tumor cell proliferation, apoptosis, and hypoxia. Lentivirus was used to silence or overexpress AKR1C3 simultaneously with the injection of HSA-RSV NPs via the tail vein to assess the impact of AKR1C3 on the regulation of HSA-RSV NPs in HCC progression. Bioinformatics analysis revealed that AKR1C3 is an important target gene involved in the regulation of HCC by RSV, which is associated with the prognosis of HCC patients and upregulated in expression. In vitro cell experiments showed that RSV significantly inhibits the respiratory metabolism of HCC cells, suppressing their proliferation, migration, and invasion and promoting apoptosis. Silencing AKR1C3 further enhances the toxicity of RSV towards HCC cells. The characterization and cytotoxicity experiments of nanomaterials demonstrated the successful construction of HSA-RSV NPs, which exhibited stronger inhibitory effects on HCC cells. In vivo, animal experiments further confirmed that targeted downregulation of AKR1C3 by HSA-RSV NPs suppresses the progression of HCC and tumor hypoxia while exhibiting tumor targeting and biological safety. Targeted downregulation of AKR1C3 by HSA-RSV NPs can alleviate HCC tumor hypoxia and inhibit the progression of HCC.
    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: Case Reports
    背景:侵袭性成熟T细胞淋巴瘤(TCL)是一种预后不良的疾病。方法:我们分析了16例随机选择的TCL患者中22种肿瘤细胞功能蛋白的表达。在石蜡包埋的肿瘤组织切片中进行免疫组织化学以确定肿瘤细胞中的蛋白质表达状态。结果:葡萄糖调节蛋白94(GRP94),一种在肿瘤微环境中内质网(ER)应激下充当促生存成分的蛋白质,与生存期缩短显著相关。此外,当GRP94与其他6个因素结合时,观察到显著差异.六个因素是(1)程序性细胞死亡配体1(PD-L1);(2)程序性细胞死亡1(PD-1);(3)醛酮还原酶家族1成员C3(AKR1C3);(4)肿瘤抑制因子P53;(5)葡萄糖调节蛋白78(GRP78),ER应激蛋白;和(6)胸苷磷酸化酶(TP)。基于GRP94和其他六种在肿瘤中表达的因子的组合,我们提出了一种新的TCL预后分类系统(TCLUrayasu分类)。第1组(预后相对良好):GRP94阴性(n=6;中位OS,88个月;p<0.01);第2组(预后不良):GRP94阳性,加上上述六个因素中的两个因素的表达(n=5;中位OS,25个月;p>0.05);第3组(预后极差):GRP94阳性,加上上述六个因素中至少三个的表达(n=5;中位OS,10个月;p<0.01)。结论:因此,TCLUrayasu预后分类可能是一个简单的,有用的,以及创新的分类,这也解释了每种功能蛋白对治疗的抗性机制。如果在更多的患者中验证,TCLUrayasu分类将能够使用选定的抑制剂对每位患者中发现的异常蛋白进行靶向治疗.
    Background: Aggressive mature T-cell lymphoma (TCL) is a disease that carries a poor prognosis. Methods: We analyzed the expression of 22 tumor cell functional proteins in 16 randomly selected patients with TCL. Immunohistochemistry was performed in paraffin-embedded tumor tissue sections to determine the protein expression statuses in tumor cells. Results: Glucose-regulated protein 94 (GRP94), a protein that serves as a pro-survival component under endoplasmic reticulum (ER) stress in the tumor microenvironment, was significantly associated with a shortened survival. Furthermore, significant differences were observed when GRP94 was combined with six other factors. The six factors were (1) programmed cell death-ligand 1 (PD-L1); (2) programmed cell death 1 (PD-1); (3) aldo-keto reductase family 1 member C3 (AKR1C3); (4) P53, a tumor suppressor; (5) glucose-regulated protein 78 (GRP78), an ER stress protein; and (6) thymidine phosphorylase (TP). Based on the combination of GRP94 and the six other factors expressed in the tumors, we propose a new prognostic classification system for TCL (TCL Urayasu classification). Group 1 (relatively good prognosis): GRP94-negative (n = 6; median OS, 88 months; p < 0.01); Group 2 (poor prognosis): GRP94-positive, plus expression of two of the six factors mentioned above (n = 5; median OS, 25 months; p > 0.05); and Group 3 (very poor prognosis): GRP94-positive, plus expression of at least three of the six factors mentioned above (n = 5; median OS, 10 months; p < 0.01). Conclusions: Thus, the TCL Urayasu prognostic classification may be a simple, useful, and innovative classification that also explains the mechanism of resistance to treatment for each functional protein. If validated in a larger number of patients, the TCL Urayasu classification will enable a targeted treatment using selected inhibitors acting on the abnormal protein found in each patient.
    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
    目的:小细胞肺癌(SCLC)是一种高级别神经内分泌肿瘤,其特征是对化疗的初始敏感性,其次是耐药性的发展。SCLC耐药的潜在机制尚未完全阐明。醛酮还原酶家族1成员C3(AKR1C3),已知与多种肿瘤的放化疗抵抗有关。我们旨在评估AKR1C3的预后意义和免疫特征,并探讨其在促进SCLC耐药中的潜在作用。
    方法:81例术后SCLC组织用于分析AKR1C3的预后价值和免疫特征。组织微阵列用于验证AKR1C3在SCLC中的临床意义。AKR1C3对SCLC细胞增殖的影响,迁移,通过CCK-8,伤口愈合试验检测细胞凋亡和肿瘤血管生成,transwell分析,流式细胞术和试管形成测定。
    结果:与其他AKR1C家族基因相比,AKR1C3表现出最高的表达水平,多因素cox回归分析将其确定为SCLC的独立预后因素。接受放化疗的高AKR1C3表达患者的总生存期(OS)明显缩短。此外,AKR1C3参与SCLC肿瘤免疫微环境的调节。沉默AKR1C3导致细胞增殖和迁移的抑制,同时促进SCLC细胞凋亡和减少上皮间质转化(EMT)。
    结论:AKR1C3促进细胞生长和转移,通过诱导SCLC中的EMT和血管生成导致耐药性。
    OBJECTIVE: Small cell lung cancer (SCLC) is a high-grade neuroendocrine tumor characterized by initial sensitivity to chemotherapy, followed by the development of drug resistance. The underlying mechanisms of resistance in SCLC have not been fully elucidated. Aldo-keto reductase family 1 member C3 (AKR1C3), is known to be associated with chemoradiotherapy resistance in diverse tumors. We aim to evaluate the prognostic significance and immune characteristics of AKR1C3 and investigate its potential role in promoting drug resistance in SCLC.
    METHODS: 81 postoperative SCLC tissues were used to analyze AKR1C3 prognostic value and immune features. The tissue microarrays were employed to validate the clinical significance of AKR1C3 in SCLC. The effects of AKR1C3 on SCLC cell proliferation, migration, apoptosis and tumor angiogenesis were detected by CCK-8, wound healing assay, transwell assay, flow cytometry and tube formation assay.
    RESULTS: AKR1C3 demonstrated the highest expression level compared to other AKR1C family genes, and multivariate cox regression analysis identified it as an independent prognostic factor for SCLC. High AKR1C3 expression patients who underwent chemoradiotherapy experienced significantly shorter overall survival (OS). Furthermore, AKR1C3 was involved in the regulation of the tumor immune microenvironment in SCLC. Silencing of AKR1C3 led to the inhibition of cell proliferation and migration, while simultaneously promoting apoptosis and reducing epithelial-mesenchymal transition (EMT) in SCLC.
    CONCLUSIONS: AKR1C3 promotes cell growth and metastasis, leading to drug resistance through inducing EMT and angiogenesis in SCLC.
    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:人醛酮还原酶家族1成员C3(AKR1C3)是参与多种生理代谢过程的重要分子。然而,它的表达,胃癌发生的生物学功能和临床意义尚不清楚。方法:我们从几个公共数据门户和临床样本中收集数据,并系统分析组织和血浆AKR1C3表达的临床意义。然后,我们筛选了预后危险因素,并建立了预测总体生存时间和术后复发风险的新型预后相关列线图模型.使用临床样本进一步评估列线图模型的应用价值。此外,我们通过多组学功能分析和免疫浸润分析,探讨了AKR1C3在胃癌发生和转移中的潜在生物学功能。结果:癌组织中AKR1C3水平降低,但在GC患者血浆中明显升高;AKR1C3在任一样本类型中的表达与多种临床病理特征密切相关。通过结合临床病理因素和AKR1C3水平,我们建立了两个新的列线图模型来预测总生存时间和术后复发风险.多组学功能分析显示,当它的表达失调时,AKR1C3可以通过多种调控模式广泛参与基因表达调控,RNA和蛋白质水平并在癌变和转移中发挥各种关键的生物学作用。此外,AKR1C3的表达与几种免疫细胞类型的浸润有关,预测AKR1C3与几种临床药物相互作用。结论:AKR1C3表达异常与胃癌的发生和免疫治疗反应有关,是胃癌中一个有前途的生物标志物和有效的生物治疗靶点。
    Background: Human aldo-keto reductase family 1 member C3 (AKR1C3) is an important molecule that participates in multiple physiological metabolic processes. However, its expression, biological functions and clinical significance in gastric carcinogenesis are unclear. Methods: We collected data from several public data portals and clinical samples and systematically analyzed the clinical significance of tissue and plasma AKR1C3 expression. Then, we filtered prognostic risk factors and established novel prognosis-related nomogram models for predicting overall survival time and postoperative recurrence risk. The application value of the nomogram models was further assessed using clinical samples. Moreover, we explored the potential biological functions of AKR1C3 in gastric carcinogenesis and metastasis through multiomics functional analysis and immune infiltration analysis. Results: AKR1C3 levels were reduced in cancer tissue but increased significantly in the plasma of GC patients; AKR1C3 expression in either sample type was closely associated with multiple clinicopathological characteristics. By combining clinicopathological factors and AKR1C3 levels, two novel nomogram models were developed to predict overall survival time and postoperative recurrence risk. Multiomics functional analysis revealed that when its expression is dysregulated, AKR1C3 can widely participate in gene expression regulation through multiple regulatory modes at the gene, RNA and protein levels and exert various crucial biological effects in carcinogenesis and metastasis. Moreover, AKR1C3 expression was correlated with the infiltration of several immune cell types, and AKR1C3 was predicted to interact with several clinical drugs. Conclusion: Dysregulated AKR1C3 expression is related to gastric carcinogenesis and immunotherapy response and is a promising biomarker and effective biotherapy target in GC.
    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
    Aldo-酮还原酶家族1成员C3(AKR1C3),也称为5型17β-羟基类固醇脱氢酶(17β-HSD5)或前列腺素F(PGF)合酶,作为雄激素生物合成的关键酶。它催化弱雄激素的转化,雌酮(一种弱雌激素),和PGD2成强效雄激素(睾酮和5α-二氢睾酮),17β-雌二醇(一种有效的雌激素),和11β-PGF2α,分别。AKR1C3激活雄激素受体(AR)信号通路的水平升高,有助于肿瘤复发并赋予对癌症治疗的抵抗力。AKR1C3的过表达作为致癌因子,促进癌细胞增殖,入侵,和转移,并且与癌症患者的不良预后和总生存期相关。抑制AKR1C3在抑制肿瘤进展和克服治疗抗性方面已显示出有效的功效。因此,AKR1C3抑制剂的开发和设计引起了研究人员越来越多的兴趣,近年来取得了重大进展。新型AKR1C3抑制剂,包括基于其结构和框架设计的现有药物的天然产物和类似物,继续在世界各地的实验室中发现和开发。AKR1C3酶已成为癌症进展和治疗抗性的关键参与者,给癌症治疗带来挑战。本文旨在全面分析AKR1C3在肿瘤发展中的作用。它对治疗抗性的影响,以及用于肿瘤治疗的AKR1C3抑制剂开发的最新进展。
    Aldo-Keto Reductase Family 1 Member C3 (AKR1C3), also known as type 5 17β-hydroxysteroid dehydrogenase (17β-HSD5) or prostaglandin F (PGF) synthase, functions as a pivotal enzyme in androgen biosynthesis. It catalyzes the conversion of weak androgens, estrone (a weak estrogen), and PGD2 into potent androgens (testosterone and 5α-dihydrotestosterone), 17β-estradiol (a potent estrogen), and 11β-PGF2α, respectively. Elevated levels of AKR1C3 activate androgen receptor (AR) signaling pathway, contributing to tumor recurrence and imparting resistance to cancer therapies. The overexpression of AKR1C3 serves as an oncogenic factor, promoting carcinoma cell proliferation, invasion, and metastasis, and is correlated with unfavorable prognosis and overall survival in carcinoma patients. Inhibiting AKR1C3 has demonstrated potent efficacy in suppressing tumor progression and overcoming treatment resistance. As a result, the development and design of AKR1C3 inhibitors have garnered increasing interest among researchers, with significant progress witnessed in recent years. Novel AKR1C3 inhibitors, including natural products and analogues of existing drugs designed based on their structures and frameworks, continue to be discovered and developed in laboratories worldwide. The AKR1C3 enzyme has emerged as a key player in carcinoma progression and therapeutic resistance, posing challenges in cancer treatment. This review aims to provide a comprehensive analysis of AKR1C3\'s role in carcinoma development, its implications in therapeutic resistance, and recent advancements in the development of AKR1C3 inhibitors for tumor therapies.
    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
    AST-001是化学合成的无活性氮芥前药,通过醛酮还原酶家族1成员C3(AKR1C3)选择性裂解为细胞毒性氮丙啶(AST-2660)。这项研究的目的是研究前药的药代动力学和组织分布,AST-001及其活性代谢产物,AST-2660,在小鼠中,老鼠,还有猴子.在对Sprague-Dawley大鼠单次和每日一次静脉推注剂量为1.5、4.5和13.5mg/kgAST-001后,对食蟹猴每日一次1小时静脉输注0.5、1.5和4.5mg/kgAST-001后,AST-001表现出剂量依赖性药代动力学,并在输注结束时达到峰值血浆水平。重复给药7天后未观察到显著的积累和性别差异。在老鼠身上,AST-001的半衰期与剂量无关,范围为4.89至5.75h。在食蟹猴中,AST-001的半衰期为1.66-5.56h,并随剂量增加而增加。在Sprague-Dawley大鼠和植入LI6643或LI6280HepG2-GFP肿瘤片段的雌性无胸腺裸鼠中进行的组织分布研究中,AST-001广泛分布于选定的组织。单次静脉注射后,AST-001主要不是作为前药排泄的,AST-001或尿液中的代谢物AST-2660,粪便,还有胆汁.对临床前数据和物种间异速缩放比例的综合分析用于估计AST-001在人类中的药代动力学参数,并导致在首次在人剂量递增研究中建议起始剂量为5mg/m2。
    AST-001 is a chemically synthesized inactive nitrogen mustard prodrug that is selectively cleaved to a cytotoxic aziridine (AST-2660) via aldo-keto reductase family 1 member C3 (AKR1C3). The purpose of this study was to investigate the pharmacokinetics and tissue distribution of the prodrug, AST-001, and its active metabolite, AST-2660, in mice, rats, and monkeys. After single and once daily intravenous bolus doses of 1.5, 4.5, and 13.5 mg/kg AST-001 to Sprague-Dawley rats and once daily 1 h intravenous infusions of 0.5, 1.5, and 4.5 mg/kg AST-001 to cynomolgus monkeys, AST-001 exhibited dose-dependent pharmacokinetics and reached peak plasma levels at the end of the infusion. No significant accumulation and gender differences were observed after 7 days of repeated dosing. In rats, the half-life of AST-001 was dose independent and ranged from 4.89 to 5.75 h. In cynomolgus monkeys, the half-life of AST-001 was from 1.66 to 5.56 h and increased with dose. In tissue distribution studies conducted in Sprague-Dawley rats and in liver cancer PDX models in female athymic nude mice implanted with LI6643 or LI6280 HepG2-GFP tumor fragments, AST-001 was extensively distributed to selected tissues. Following a single intravenous dose, AST-001 was not excreted primarily as the prodrug, AST-001 or the metabolite AST-2660 in the urine, feces, and bile. A comprehensive analysis of the preclinical data and inter-species allometric scaling were used to estimate the pharmacokinetic parameters of AST-001 in humans and led to the recommendation of a starting dose of 5 mg/m2 in the first-in-human dose escalation study.
    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
    甲状腺癌是一种高分化、恶性较差的肿瘤。干扰糖酵解已成为控制癌症进展的有效手段,自噬与糖酵解呈负相关。已证明Aldo-酮还原酶家族1成员C3(AKR1C3)在甲状腺癌组织中高表达,并且较高的AKR1C3表达预示预后较差。我们旨在探讨AKR1C3是否可以通过调节自噬依赖性糖酵解影响甲状腺癌的进展。Westernblot检测甲状腺癌细胞中AKR1C3的表达。然后,在不存在或存在3-甲基腺嘌呤(3-MA)或PMA处理的情况下,通过用对AKR1C3特异的短发夹RNA转染敲低AKR1C3。流式细胞术检测细胞周期和细胞凋亡。免疫荧光染色用于分析LC3B表达。胞外酸化,检测葡萄糖摄取和乳酸分泌。评价体内AKR1C3功能不全对甲状腺癌的致瘤性,将具有AKR1C3敲除的TPC-1细胞皮下注射到裸小鼠中。然后,通过免疫组织化学分析测量肿瘤组织中cyclinD1和Ki67的表达。通过末端脱氧核苷酸转移酶介导的缺口末端标记染色评估细胞凋亡。此外,与细胞周期相关的蛋白质的表达,凋亡,糖酵解,自噬,通过蛋白质印迹评估细胞和肿瘤组织中的细胞外信号调节激酶(ERK)信号传导。在甲状腺癌细胞中观察到高表达的AKR1C3。AKR1C3敲低诱导TPC-1细胞的细胞周期阻滞和凋亡。此外,AKR1C3沉默后,自噬被激活,糖酵解被抑制,3-MA治疗恢复了AKR1C3沉默对糖酵解的影响。进一步的实验表明,AKR1C3功能不全抑制ERK信号传导,PMA应用逆转了TPC-1细胞中AKR1C3沉默诱导的自噬。体内结果表明,AKR1C3敲低抑制裸鼠皮下TPC-1肿瘤的发展,并使ERK信号失活。总的来说,AKR1C3沉默通过失活ERK信号抑制甲状腺癌细胞自噬依赖性糖酵解.
    Thyroid cancer is a highly differentiated and poorly malignant tumor. Interfering with glycolysis has become an effective means of controlling cancer progression and autophagy is negatively correlated with glycolysis. Aldo-keto reductase family 1 member C3 (AKR1C3) has been demonstrated to be highly expressed in thyroid cancer tissue and the higher AKR1C3 expression predicted the worse prognosis. We aimed to explore whether AKR1C3 could affect thyroid cancer progression by regulating autophagy-dependent glycolysis. AKR1C3 expression in thyroid cancer cells was detected by western blot. Then, AKR1C3 was knocked down by transfection with short hairpin RNA specific to AKR1C3 in the absence or presence of 3-methyladenine (3-MA) or PMA treatment. Cell cycle and apoptosis was detected by flow cytometry. Immunofluorescence staining was used to analyze LC3B expression. Extracellular acidification, glucose uptake and lactic acid secretion were detected. To evaluate the tumorigenicity of AKR1C3 insufficiency on thyroid cancer in vivo, TPC-1 cells with AKR1C3 knockdown were injected subcutaneously into nude mice. Then, cyclinD1 and Ki67 expression in tumorous tissues was measured by immunohistochemical analysis. Apoptosis was assessed by terminal-deoxynucleoitidyl transferase mediated nick end labeling staining. Additionally, the expression of proteins related to cell cycle, apoptosis, glycolysis, autophagy, and extracellular signal-regulated kinase (ERK) signaling in cells and tumor tissues was assessed by western blot. Highly expressed AKR1C3 was observed in thyroid cancer cells. AKR1C3 knockdown induced cell cycle arrest and apoptosis of TPC-1 cells. Besides, autophagy was activated and glycolysis was inhibited following AKR1C3 silencing, and 3-MA treatment restored the impacts of AKR1C3 silencing on glycolysis. The further experiments revealed that AKR1C3 insufficiency inhibited ERK signaling and PMA application reversed AKR1C3 silencing-induced autophagy in TPC-1 cells. The in vivo results suggested that AKR1C3 knockdown inhibited the development of subcutaneous TPC-1 tumors in nude mice and inactivated the ERK signaling. Collectively, AKR1C3 silencing inhibited autophagy-dependent glycolysis in thyroid cancer by inactivating ERK signaling.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号