Topoisomerase I

拓扑异构酶 I
  • 文章类型: Journal Article
    结直肠癌(CRC)仍然是消化系统最常见的恶性肿瘤之一。然而,临床使用安全有效的化疗药物的可用性仍然有限。喜树碱(CPT)及其衍生物,虽然被批准用于癌症治疗,由于其低生物利用度和高全身毒性,在临床应用中遇到了重大挑战。在CPT的7位的战略性修饰使得能够开发具有高活性的新型CPT衍生物。在本研究中,一系列含有氨基脲的化合物,氨基硫脲,筛选了在7位作为取代基的酰氨基硫脲。随后评价这些化合物对人胃癌(GC)细胞系AGS和CRC细胞系HCT116的细胞毒性。两个导数,XSJ05(IC50=0.006±0.003μM)和XSJ07(IC50=0.013±0.003μM),表现出显著有效的抗CRC活性,比TPT更好。此外,他们有更好的安全性。体外机制研究表明,XSJ05和XSJ07通过抑制拓扑异构酶I(TopoI)的活性而对CRC细胞增殖发挥抑制作用。这种抑制引发DNA双链断裂,导致DNA损伤并随后导致CRC细胞停滞在G2/M期。最终,细胞经历凋亡。总的来说,这些结果表明,XSJ05和XSJ07具有优异的活性,同时具有良好的安全性,表明它们作为发展CRC疗法的先导化合物的潜力。
    Colorectal cancer (CRC) remains one of the most prevalent malignant tumors of the digestive system, yet the availability of safe and effective chemotherapeutic agents for clinical use remains limited. Camptothecin (CPT) and its derivatives, though approved for cancer treatment, have encountered significant challenges in clinical application due to their low bioavailability and high systemic toxicity. Strategic modification at the 7-position of CPT enables the development of novel CPT derivatives with high activity. In the present study, a series of compounds incorporating aminoureas, amino thioureas, and acylamino thioureas as substituents at the 7-position were screened. These compounds were subsequently evaluated for their cytotoxicity against the human gastric cancer (GC) cell line AGS and the CRC cell line HCT116. Two derivatives, XSJ05 (IC50 = 0.006 ± 0.003 μM) and XSJ07 (IC50 = 0.013 ± 0.003 μM), exhibited remarkably effective anti-CRC activity, being better than TPT. In addition, they have a better safety profile. In vitro mechanistic studies revealed that XSJ05 and XSJ07 exerted their inhibitory effects on CRC cell proliferation by suppressing the activity of topoisomerase I (Topo I). This suppression triggers DNA double-strand breaks, leads to DNA damage and subsequently causes CRC cells to arrest in the G2/M phase. Ultimately, the cells undergo apoptosis. Collectively, these findings indicate that XSJ05 and XSJ07 possess superior activity coupled with favorable safety profiles, suggesting their potential as lead compounds for the development of CRC therapeutics.
    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
    单细胞原生动物寄生虫多尼利什曼原虫是内脏利什曼病(VL)或黑热病的病原体,被忽视的致命寄生虫病.VL的常规治疗由具有几个缺点如毒性的治疗剂组成。高成本,疗效差异和耐药性增加。因此,迫切需要开发针对寄生虫的有效治疗方法。先前的报道表明,类黄酮可以抑制酶利什曼原虫DNA拓扑异构酶I(LdTopILS)。因此,在本研究中第一次,我们透露HSP(黄酮类化合物的天然来源之一),作为一种有效的天然抗利什曼胺化合物,在20mg/kg体重的BALB/c小鼠中具有功效,在预孵育条件下(竞争性),在160μM时抑制LdTopILS的97%活性。它与游离酶结合并且不允许它与底物DNA结合。此外,HSP不稳定DNA-拓扑异构酶I可切割复合物。因此,HSP起催化拓扑异构酶I抑制剂的作用,通过与酶大亚基的Lys269和Thr411结合来抑制完全活性。另一方面,HSP通过细胞活性氧的形成诱导topoI介导的程序性细胞死亡过程,导致线粒体膜电位去极化,然后是核DNA的片段化。因此,本研究阐明了一种天然类黄酮,将来可能是治疗VL的有希望的线索。
    Unicellular protozoan parasite Leishmania donovani is the causative agent for visceral leishmaniasis (VL) or Kala-azar, a neglected fatal parasitic disease. The conventional treatment of VL consists of therapeutic agents having several shortcomings such as toxicity, high cost, efficacy variance and increased drug resistance. Therefore, there is a desperate need to develop an effective treatment against the parasite. Previous reports suggested that flavonoids can inhibit the enzyme Leishmania donovani DNA topoisomerase I (LdTopILS). Therefore, for the first time in this present study, we divulge HSP (one of the natural sources of flavonoids), as a potent natural antileishmanial compound with efficacy in BALB/c mice at 20 mg/kg of body weight, inhibits LdTopILS at 97 % of its activity at 160 μM in preincubation condition (competitively). It binds with free enzyme and does not allow it to bind with the substrate DNA. Moreover, HSP does not stabilize DNA-topoisomerase I cleavable complex. Thus, HSP acts a catalytic topoisomerase I inhibitor, which inhibits complete activity by binding with Lys269 and Thr411 of large subunit of the enzyme. On the other hand, HSP induces the topo I-mediated programmed cell death process by the formation of cellular reactive oxygen species, resulting in depolarization of mitochondrial membrane potential, followed by fragmentation of nuclear DNA. Therefore, the present study illuminates a natural flavonoid that in future might be a promising lead for the treatment of VL.
    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
    TDP1去除转录阻断拓扑异构酶I切割复合物(TOP1ccs),其失活的H493R突变导致神经退行性综合征SCAN1。然而,SCAN1表型的分子机制尚不清楚.这里,我们使用CRISPR-Cas9生成人类SCAN1细胞模型,并显示它们随着基因表达和R环基因组分布的变化而积累TOP1ccs。由于增加的DSB形成和缺乏修复,SCAN1细胞还会在G1细胞群体中积累转录DNA双链断裂(DSB),两者都是由于转录阻断TOP1ccs的失败去除。TDP1活性不足导致DSB产量增加,并且突变的TDP1蛋白的存在通过TDP2依赖性备份途径阻碍DSB修复。这项研究为研究TDP1在生理和病理条件下的功能提供了强大的模型,并揭示了突变的TDP1蛋白的功能获得,这阻止了DSB修复,而不是TDP1活性本身的损失,可能有助于SCAN1的发病机制。
    TDP1 removes transcription-blocking topoisomerase I cleavage complexes (TOP1ccs), and its inactivating H493R mutation causes the neurodegenerative syndrome SCAN1. However, the molecular mechanism underlying the SCAN1 phenotype is unclear. Here, we generate human SCAN1 cell models using CRISPR-Cas9 and show that they accumulate TOP1ccs along with changes in gene expression and genomic distribution of R-loops. SCAN1 cells also accumulate transcriptional DNA double-strand breaks (DSBs) specifically in the G1 cell population due to increased DSB formation and lack of repair, both resulting from abortive removal of transcription-blocking TOP1ccs. Deficient TDP1 activity causes increased DSB production, and the presence of mutated TDP1 protein hampers DSB repair by a TDP2-dependent backup pathway. This study provides powerful models to study TDP1 functions under physiological and pathological conditions and unravels that a gain of function of the mutated TDP1 protein, which prevents DSB repair, rather than a loss of TDP1 activity itself, could contribute to SCAN1 pathogenesis.
    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
    Coumestan代表了分布在许多天然产物中的生物学相关结构基序,相关衍生物的快速构建以及靶标的表征将加速药物化学中先导化合物的发现。在这项工作中,通过双电极恒流电解开发了一种通用且可扩展的8,9-二羟基coumestans方法。两相(水/有机)体系的应用对成功起着至关重要的作用,保护敏感的邻苯醌中间体免受过度氧化。基于结构多样的产品,完成了对coumestan脚手架的初步SAR研究,和化合物3r表现出有效的抗增殖活性和强大的拓扑异构酶I(Top1)抑制活性。进一步的机制研究表明,化合物3r是一种新型的Top1毒物,这可能为Top1靶向抗肿瘤剂的开发开辟了一条途径。
    Coumestan represents a biologically relevant structural motif distributed in a number of natural products, and the rapid construction of related derivatives as well as the characterization of targets would accelerate lead compound discovery in medicinal chemistry. In this work, a general and scalable approach to 8,9-dihydroxycoumestans via two-electrode constant current electrolysis was developed. The application of a two-phase (aqueous/organic) system plays a crucial role for success, protecting the sensitive o-benzoquinone intermediates from over-oxidation. Based on the structurally diverse products, a primary SAR study on coumestan scaffold was completed, and compound 3 r exhibited potent antiproliferative activities and a robust topoisomerase I (Top1) inhibitory activity. Further mechanism studies demonstrates that compound 3 r was a novel Top1 poison, which might open an avenue for the development of Top1-targeted antitumor agent.
    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:喜树碱(CPT),一种具有抗肿瘤特性的五环生物碱,来自喜树。拓扑替康和伊立替康(CPT衍生物)在25年前首次被食品和药物管理局批准用于癌症治疗,并且仍然是今天的关键抗癌药物。然而,它们的使用通常受到临床毒性的限制。尽管进行了广泛的发展努力,许多这些衍生物在临床上没有成功,特别是它们对胰腺癌的有效性仍然很低。
    目的:本研究旨在评估我们实验室合成的CPT衍生物FLQY2的治疗活性,对抗胰腺癌,将其功效和作用机制与已建立的临床药物进行比较。
    方法:使用MTT测定法评估FLQY2对癌细胞的细胞毒性作用。采用患者来源的类器官(PDO)模型来比较FLQY2对各种癌症中现有临床药物的敏感性。通过流式细胞术检测FLQY2对MiaPaca-2胰腺癌细胞凋亡和细胞周期停滞的影响。进行转录组和蛋白质组分析以探索FLQY2抗肿瘤活性的潜在机制。使用蛋白质印迹法确定由FLQY2调节的蛋白质的水平。此外,在胰腺癌异种移植模型中评估了FLQY2的体内抗肿瘤功效.
    结果:FLQY2表现出(1)有效的细胞毒性;(2)与目前的临床药物如吉西他滨相比,在PDO模型中具有优异的肿瘤抑制活性,5-氟尿嘧啶,顺铂,紫杉醇,ivosidenib,infinitinib,和lenvatinib;(3)在胰腺癌异种移植模型中,肿瘤抑制作用明显高于紫杉醇脂质体;(4)强大的抗肿瘤作用,与TOPI和PDK1/AKT/mTOR信号通路的抑制密切相关。体外研究表明,FLQY2抑制细胞增殖,菌落形成,诱导细胞凋亡,并在纳摩尔浓度下引起细胞周期停滞。此外,FLQY2和吉西他滨的组合具有显著的抑制作用和协同作用。
    结论:该研究证实了拓扑异构酶I和PDK1/AKT/mTOR通路参与介导FLQY2治疗MiaPaca-2胰腺癌的抗肿瘤活性。因此,FLQY2具有作为胰腺癌患者的新型治疗选择的潜力。
    BACKGROUND: Camptothecin (CPT), a pentacyclic alkaloid with antitumor properties, is derived from the Camptotheca acuminata. Topotecan and irinotecan (CPT derivatives) were first approved by the Food and Drug Administration for cancer treatment over 25 years ago and remain key anticancer drugs today. However, their use is often limited by clinical toxicity. Despite extensive development efforts, many of these derivatives have not succeeded clinically, particularly in their effectiveness against pancreatic cancer which remains modest.
    OBJECTIVE: This study aimed to evaluate the therapeutic activity of FLQY2, a CPT derivative synthesized in our laboratory, against pancreatic cancer, comparing its efficacy and mechanism of action with those of established clinical drugs.
    METHODS: The cytotoxic effects of FLQY2 on cancer cells were assessed using an MTT assay. Patient-derived organoid (PDO) models were employed to compare the sensitivity of FLQY2 to existing clinical drugs across various cancers. The impact of FLQY2 on apoptosis and cell cycle arrest in Mia Paca-2 pancreatic cancer cells was examined through flow cytometry. Transcriptomic and proteomic analyses were conducted to explore the underlying mechanisms of FLQY2\'s antitumor activity. Western blotting was used to determine the levels of proteins regulated by FLQY2. Additionally, the antitumor efficacy of FLQY2 in vivo was evaluated in a pancreatic cancer xenograft model.
    RESULTS: FLQY2 demonstrated (1) potent cytotoxicity; (2) superior tumor-suppressive activity in PDO models compared to current clinical drugs such as gemcitabine, 5-fluorouracil, cisplatin, paclitaxel, ivosidenib, infinitinib, and lenvatinib; (3) significantly greater tumor inhibition than paclitaxel liposomes in a pancreatic cancer xenograft model; (4) robust antitumor effects, closely associated with the inhibition of the TOP I and PDK1/AKT/mTOR signaling pathways. In vitro studies revealed that FLQY2 inhibited cell proliferation, colony formation, induced apoptosis, and caused cell cycle arrest at nanomolar concentrations. Furthermore, the combination of FLQY2 and gemcitabine exhibited significant inhibitory and synergistic effects.
    CONCLUSIONS: The study confirmed the involvement of topoisomerase I and the PDK1/AKT/mTOR pathways in mediating the antitumor activity of FLQY2 in treating Mia Paca-2 pancreatic cancer. Therefore, FLQY2 has potential as a novel therapeutic option for patients with pancreatic cancer.
    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
    光谱学,生物化学,和计算建模研究已用于评估一组小沟结合(MGB)配体对自身互补DNA序列5'-d(CGCACTAGTGCG)-3'和5'-d(CGCAGTACTGCG)-3'的结合能力。配体经过精心设计,以靶向DNA反应元件,5\'-WGWWCW-3\',几个核受体的结合位点。用三种MGB配体制备的DNA样品的基本1D1HNMR光谱显示出细微的变化,表明每种配体如何与两种DNA序列的双螺旋结构缔合。所研究的配体之间的变化反映在1D1H和31P-{1H}NMR光谱的线形和强度中。这些1DNMR光谱的快速视觉检查被证明有利于提供关于MGB结合分子的有价值的见解。NMR结果与UVDNA变性和分子建模研究的结果一致。NMR光谱和计算分析均表明,所研究的配体以头对尾的方式作为反平行的并排二聚体与小沟结合。此外,与生化研究结果的比较提供了对作用机制的宝贵见解,以及与其结构相关的MGBs的抗肿瘤活性,未来优化MGBs作为治疗剂的必要先决条件。
    Spectroscopic, biochemical, and computational modelling studies have been used to assess the binding capability of a set of minor groove binding (MGB) ligands against the self-complementary DNA sequences 5\'-d(CGCACTAGTGCG)-3\' and 5\'-d(CGCAGTACTGCG)-3\'. The ligands were carefully designed to target the DNA response element, 5\'-WGWWCW-3\', the binding site for several nuclear receptors. Basic 1D 1H NMR spectra of the DNA samples prepared with three MGB ligands show subtle variations suggestive of how each ligand associates with the double helical structure of both DNA sequences. The variations among the investigated ligands were reflected in the line shape and intensity of 1D 1H and 31P-{1H} NMR spectra. Rapid visual inspection of these 1D NMR spectra proves to be beneficial in providing valuable insights on MGB binding molecules. The NMR results were consistent with the findings from both UV DNA denaturation and molecular modelling studies. Both the NMR spectroscopic and computational analyses indicate that the investigated ligands bind to the minor grooves as antiparallel side-by-side dimers in a head-to-tail fashion. Moreover, comparisons with results from biochemical studies offered valuable insights into the mechanism of action, and antitumor activity of MGBs in relation to their structures, essential pre-requisites for future optimization of MGBs as therapeutic agents.
    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
    其广谱的生物活性使苯并咪唑成为药学上的基本药效团。本文描述了新合成的AT特异性荧光双苯并咪唑分子DB2Py(n),其中包含抗生素药物netropsin的吡咯甲酰胺片段。使用吸收的物理化学方法,荧光,和圆二色性光谱显示了双苯并咪唑吡咯与DNA形成复合物的能力。新的DB2Py(n)系列已被证明对人类肿瘤细胞系更具毒性,并且对非肿瘤细胞系的脆弱性较低。双苯并咪唑-吡咯穿透细胞核,影响细胞周期合成(S)期,并在低浓度的无细胞模型中抑制真核拓扑异构酶I。实时肿瘤细胞增殖试验证实了该分子在二聚化后的毒性增强。在具有MDR表型的细胞模型中测试的双苯并咪唑-吡咯的初步细胞毒性数据表明,单体化合物可以克服MDR,与阿霉素相比,二聚化将这种能力削弱到其中间值。在这方面,新合成的细胞毒性结构似乎有希望进一步,深入研究它们与人类肿瘤细胞相关的特性和作用机制,以及用于设计新的AT特异性配体。
    Its broad spectrum of biological activity makes benzimidazole a fundamental pharmacophore in pharmaceutics. The paper describes newly synthesized AT-specific fluorescent bis-benzimidazole molecules DB2Py(n) that contain a pyrrolcarboxamide fragment of the antibiotic drug netropsin. Physico-chemical methods using absorption, fluorescence, and circular dichroism spectra have shown the ability of bis-benzimidazole- pyrroles to form complexes with DNA. The new DB2Py(n) series have turned out to be more toxic to human tumor lines and less vulnerable to non-tumor cell lines. Bis-benzimidazole-pyrroles penetrated the cell nucleus, affected the cell-cycle synthesis (S) phase, and inhibited eukaryotic topoisomerase I in a cellfree model at low concentrations. A real-time tumor cell proliferation test confirmed the molecule\'s enhanced toxic properties upon dimerization. Preliminary cytotoxicity data for the bis-benzimidazole-pyrroles tested in a cell model with a MDR phenotype showed that monomeric compounds can overcome MDR, while dimerization weakens this ability to its intermediate values as compared to doxorubicin. In this respect, the newly synthesized cytotoxic structures seem promising for further, in-depth study of their properties and action mechanism in relation to human tumor cells, as well as for designing new AT-specific ligands.
    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
    槐定碱,源自豆科植物苦参,作为一种新型的抗癌药物,具有一定的药理活性。在这里,设计了一系列新型的N-取代槐定碱衍生物,合成并评价抗癌活性。通过QSAR预测模型,发现苯环作为主要药效团的引入,并在苯环的对位重新引入苯中,有效地提高了槐定碱衍生物的抗癌活性。体外,评估了28种新化合物对四种人肿瘤细胞系(A549,CNE-2,HepG-2和HEC-1-B)的抗癌活性。特别是,化合物26表现出显著的抑制作用,对HepG-2细胞的IC50值为15.6μM,超越顺式-二氯二胺铂(II)。分子对接研究证实,与槐定碱相比,衍生物与DNA拓扑异构酶I表现出更强的结合亲和力。此外,图26显示DNA拓扑异构酶I的显著抑制,并且可以将细胞阻滞在G0/G1期。这项研究为具有抗癌活性的N-取代的槐定碱衍生物的设计和合成提供了有价值的见解。
    Sophoridine, which is derived from the Leguminous plant Sophora alopecuroides L., has certain pharmacological activity as a new anticancer drug. Herein, a series of novel N-substituted sophoridine derivatives was designed, synthesized and evaluated with anticancer activity. Through QSAR prediction models, it was discovered that the introduction of a benzene ring as a main pharmacophore and reintroduced into a benzene in para position on the phenyl ring in the novel sophoridine derivatives improved the anticancer activity effectively. In vitro, 28 novel compounds were evaluated for anticancer activity against four human tumor cell lines (A549, CNE-2, HepG-2, and HEC-1-B). In particular, Compound 26 exhibited remarkable inhibitory effects, with an IC50 value of 15.6 μM against HepG-2 cells, surpassing cis-Dichlorodiamineplatinum (II). Molecular docking studies verified that the derivatives exhibit stronger binding affinity with DNA topoisomerase I compared to sophoridine. In addition, 26 demonstrated significant inhibition of DNA Topoisomerase I and could arrest cells in G0/G1 phase. This study provides valuable insights into the design and synthesis of N-substituted sophoridine derivatives with anticancer activity.
    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
    喜树碱是抑制参与DNA转录和癌细胞生长的hTop1酶的五环天然生物碱。喜树碱结构陷阱促使我们使用结构简化策略设计新的同源物,以减少从五环到四环的环延伸数,同时保持新化合物与DNA碱基对在Top1介导的裂解复合物和水溶性的潜在堆积,以及尽量减少化合物的肝毒性。这项研究的主轴是验证hTop1抑制活性作为一种可能的作用机制,并使用(异喹啉咪唑并喹唑啉)的三个结构面板(A-C),(咪唑并喹唑啉),和(咪唑并异喹啉),分别。DNA弛豫分析确定了五种化合物作为咪唑异喹啉3a的hTop1抑制剂,B,咪唑并喹唑啉12和异喹啉并咪唑并喹唑啉7a,Procedures.在针对不同癌细胞系的MTT细胞毒性试验中,化合物12对HOS骨癌细胞最有效(IC50=1.47μM)。同时,其他抑制剂对任何类型的癌细胞均无检测活性.化合物(12)在HOS癌细胞\'3D-多细胞肿瘤球体模型中表现出巨大的穿透力。对hTop1基因的生物信息学研究发现,TP53细胞增殖基因在hTop1的网络中。使用证明p53表达增加的基因表达测定凭经验证实了该发现。结构简化对化合物12型材的影响,与作为标准抑制剂的阿霉素相比,其特征在于没有急性口服肝毒性,在瑞士白化病雌性小鼠上测量的致死剂量,在LD50=250mg/kg时报告,以及在用化合物12治疗5周后使结肠腺癌肿瘤体积减少75.36%的治疗意义。基于CPT的活性衍生物12和非活性同源物14进入hTop1活性位点的分子对接溶液以及这种MMP的活性削减指导我们建议向化合物12咪唑并喹唑啉核心支架添加HBD和HBA变量,以通过与大沟氨基酸(Asp533,Lys532)形成氢键来增强效力,并保持与小沟氨基酸Arg532的氢键。
    Camptothecin is a pentacyclic natural alkaloid that inhibits the hTop1 enzyme involved in DNA transcription and cancer cell growth. Camptothecin structure pitfalls prompted us to design new congeners using a structure simplification strategy to reduce the ring extension number from pentacyclic to tetracyclic while maintaining potential stacking of the new compounds with the DNA base pairs at the Top1-mediated cleavage complex and aqueous solubility, as well as minimizing compound-liver toxicity. The principal axis of this study was the verification of hTop1 inhibiting activity as a possible mechanism of action and the elaboration of new simplified inhibitors with improved pharmacodynamic and pharmacokinetic profiling using three structure panels (A-C) of (isoquinolinoimidazoquinazoline), (imidazoquinazoline), and (imidazoisoquinoline), respectively. DNA relaxation assay identified five compounds as hTop1 inhibitors belonging to the imidazoisoquinolines 3a,b, the imidazoquinazolines 12, and the isoquinolinoimidazoquinazolines 7a,b. In an MTT cytotoxicity assay against different cancer cell lines, compound 12 was the most potent against HOS bone cancer cells (IC50 = 1.47 μM). At the same time, the other inhibitors had no detectable activity against any cancer cell type. Compound (12) demonstrated great penetrating power in the HOS cancer cells\' 3D-multicellular tumor spheroid model. Bioinformatics research of the hTop1 gene revealed that the TP53 cell proliferative gene is in the network of hTop1. The finding is confirmed empirically using the gene expression assay that proved the increase in p53 expression. The impact of structure simplification on compound 12 profile, characterized by the absence of acute oral liver toxicity when compared to Doxorubicin as a standard inhibitor, the lethal dose measured on Swiss Albino female mice and reported at LD50 = 250 mg/kg, and therapeutic significance in reducing colon adenocarcinoma tumor volume by 75.36 % after five weeks of treatment with compound 12. The molecular docking solutions of the active CPT-based derivative 12 and the inactive congener 14 into the active site of hTop1 and the activity cliffing of such MMP directed us to recommend the addition of HBD and HBA variables to compound 12 imidazoquinazoline core scaffold to enhance the potency via hydrogen bond formation with the major groove amino acids (Asp533, Lys532) as well as maintaining the hydrogen bond with the minor groove amino acid Arg364.
    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
    通过将查尔酮部分引入到harmine中,设计并合成了一系列β-咔啉衍生物。评估合成的衍生物对六种人癌细胞系(MCF-7,MDA-MB-231,HepG2,HT29,A549和PC-3)和一种正常细胞系(L02)的抗增殖活性。其中,化合物G11对MCF-7细胞系表现出有效的抗增殖活性,IC50值为0.34μM。进一步的生物学研究表明,化合物G11抑制MCF-7细胞的集落形成,通过下调迁移相关蛋白MMP-2抑制MCF-7细胞迁移。此外,它可以通过下调Bcl-2和上调Cleaved-PARP诱导MCF-7细胞凋亡,Bax,和磷酸化的Bim蛋白。此外,化合物G11可以作为TopoI抑制剂,影响DNA合成和转录,从而抑制癌细胞增殖。此外,化合物G11在10mg/kg的剂量下抑制4T1同基因移植小鼠的肿瘤生长,抑制率为43.19%,和63.87%在20毫克/千克,不会对小鼠或其器官造成明显的毒性,达到降低毒性和提高疗效的目的。所有这些结果表明G11作为抗肿瘤剂具有巨大的潜力,值得进一步研究。
    A series of β-carboline derivatives were designed and synthesized by introducing the chalcone moiety into the harmine. The synthesized derivatives were evaluated their anti-proliferative activities against six human cancer cell lines (MCF-7, MDA-MB-231, HepG2, HT29, A549, and PC-3) and one normal cell line (L02). Among them, compound G11 exhibited the potent anti-proliferative activity against MCF-7 cell line, with an IC50 value of 0.34 μM. Further biological studies revealed that compound G11 inhibited colony formation of MCF-7 cells, suppressed MCF-7 cell migration by downregulating migration-associated protein MMP-2. In addition, it could induce apoptosis of MCF-7 cells by downregulating Bcl-2 and upregulating Cleaved-PARP, Bax, and phosphorylated Bim proteins. Furthermore, compound G11 can act as a Topo I inhibitor, affecting DNA synthesis and transcription, thereby inhibiting cancer cell proliferation. Moreover, compound G11 inhibited tumor growth in 4T1 syngeneic transplant mice with an inhibition rate of 43.19 % at a dose of 10 mg/kg, and 63.87 % at 20 mg/kg, without causing significant toxicity to the mice or their organs, achieving the goal of reduced toxicity and increased efficacy. All these results indicate of G11 has enormous potential as an anti-tumor agent and merits further investigation.
    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