Natural Products Research Lab, Department of Biochemistry, King George's Medical University, Lucknow 226003, U.P., India.
Department of Chemistry, Shibli National PG College, Azamgarh 276 001, U.P., India.
Anticancer Agents Med Chem. 2022;22(20):3416-3437. doi: 10.2174/1871520622666220204123348.
AIMS: The present study aimed to isolate and characterize chemical compounds from Anthocephalus cadamba Miq. bark and evaluate their anticancer activity by in silico, molecular docking, and in vitro studies. BACKGROUND: Anthocephalus cadamba is a traditionally used Indian medicinal plant. The anticancer and phytochemical properties of this plant remain unexplored except for a few studies. OBJECTIVES: The objective of the study was to evaluate the antiproliferative activity of extract and fractions against breast cancer and prostate cancer cell lines and isolate and characterize active compounds from bio-active guided fractions. Moreover, the anticancer activity of isolated compounds against breast and prostate cancer cell lines was also evaluated, in addition to in silico and molecular docking interactions of isolated compounds with VEGFR2 and PDGFRα target proteins. METHODS: The compounds were isolated and purified with the help of repeated column chromatography, and spectral techniques, such as 1D, 2D NMR, and GC-MS/MS, were used to identify and elucidate the structure of the compounds. Moreover, prediction of activity spectra for substances, physiochemical properties, bioactivity radar prediction, bioactivity score, natural-product likeness, ADME, and toxicity parameters of isolated compounds (AC-1 to AC-4) was performed through various in-silico databases and servers. To evaluate the docking interaction profile and binding energies of compounds, three docking tools were utilized, such as AutoDock, AutoDock Vina, and iGEMDOCK, against two targets VEGFR2 and PDGFRα. MD simulation was performed through ligand and receptor molecular dynamic server (LARMD). RESULTS: It was found that the A. cadamba bark chloroform fraction demonstrated a significant inhibitory effect against MDA-MB-231, MCF-7, and PC-3 cells in a dose-time-dependent manner. The bioassay-guided isolation afforded four molecules AC-1 to AC-4 from chloroform fraction. Moreover, the GC-MS/MS profiling identified fourteen new molecules which were not reported earlier from A. cadamba. The in-silico study showed that the isolated compounds (AC-1 to AC-4) followed Lipinski's rule and had good oral bioavailability. While compound AC-4 had positive bioactivity scores except for kinase inhibitor activity. The ADMET profiling revealed that AC-4 was non-toxic and easily absorbed in the human intestine, and transportable in the blood-brain barrier compared to AC-1, AC-2, AC-3, and standard drug doxorubicin. Molecular docking and MD simulation assessment also signified AC-4 anticancer activity with dual inhibitory action against the target proteins VEGFR2 and PDGFRα amongst the studied compounds. The in vitro cell viability assay of isolated compounds demonstrated that AC-1 showed IC50 (μg/mL) value of 34.96 ±3.91, 47.76±3.80 69.1±4.96, AC-2; 68.26±4.22, 54.03±5.14, >100, AC-3; 35.34±4.14, 51.5±51.5, 70.8±5.25 and AC-4; 44.2±3.57, 24.2±2.67, 51.2±2.54 for MDA-MB-231, MCF-7, and PC-3 cancer cell lines, respectively and compared with standard drug doxorubicin. Moreover, fluorescence microscopy confirmed the apoptogenic property of compounds. We also found that AC-4 exhibited significant intracellular ROS production in breast cancer cells, thereby inducing apoptosis and eventually cell death. CONCLUSION: In conclusion, A. cadamba afforded four pure molecules AC-1 to AC-4 with the identification of fourteen new compounds. The entire in-silico studies concluded that the AC-4 compound had better oral bioavailability, bioactivity score, and ADMET profile among studied molecules. Molecular docking analysis and MD simulation also supported AC-4 dual inhibitory action against both VEGFR2 and PDGFRα receptors. Moreover, the isolated molecules AC-1, AC-2, AC-3, and AC-4 were found to be active against MDA-MB-231, MCF-7, and PC-3 cancer cells. The molecule AC-4 was found to induce ROS-mediated apoptosis in breast cancer cells. It was found that the anticancer inhibitory potentiality of AC-4 is directed to its molecular stereochemistry which specifically binds to the target proteins of breast cancer cells with no toxicological effect. Therefore, AC-4 is suggested to be an effective aspirant for novel drug design and discovery.
目的:本研究旨在从 Anthocephalus cadamba Miq. 树皮中分离和鉴定化学化合物,并通过计算机模拟、分子对接和体外研究评估其抗癌活性。
背景:Anthocephalus cadamba 是一种传统的药用印度植物。除了少数研究外,该植物的抗癌和植物化学特性仍未得到探索。
目的:本研究的目的是评估提取物和馏分对乳腺癌和前列腺癌细胞系的增殖抑制活性,并从生物活性导向馏分中分离和鉴定活性化合物。此外,还评估了分离化合物对乳腺癌和前列腺癌细胞系的抗癌活性,以及分离化合物与 VEGFR2 和 PDGFRα 靶蛋白的计算机模拟和分子对接相互作用。
方法:利用反复的柱层析和光谱技术,如 1D、2D NMR 和 GC-MS/MS,帮助分离和纯化化合物,并用于鉴定和阐明化合物的结构。此外,还通过各种计算机模拟数据库和服务器对分离化合物(AC-1 至 AC-4)的活性光谱预测、理化性质、生物活性雷达预测、生物活性评分、天然产物相似性、ADME 和毒性参数进行了预测。利用 AutoDock、AutoDock Vina 和 iGEMDOCK 三种对接工具,对两个靶点 VEGFR2 和 PDGFRα 进行了对接相互作用和结合能的评估。通过配体和受体分子动力学服务器(LARMD)进行了 MD 模拟。
结果:结果表明,A. cadamba 树皮的氯仿馏分在剂量和时间依赖性方式下对 MDA-MB-231、MCF-7 和 PC-3 细胞具有显著的抑制作用。生物活性导向的分离从氯仿馏分中获得了四个分子 AC-1 至 AC-4。此外,GC-MS/MS 分析鉴定出了 14 种以前从未在 A. cadamba 中报道过的新分子。计算机模拟研究表明,分离出的化合物(AC-1 至 AC-4)符合 Lipinski 规则,具有良好的口服生物利用度。而化合物 AC-4 除了激酶抑制剂活性外,具有阳性的生物活性评分。ADMET 分析表明,AC-4 无毒性,易于在人体肠道吸收,并且在血液-脑屏障中的传输能力优于 AC-1、AC-2、AC-3 和标准药物阿霉素。分子对接和 MD 模拟评估也表明,AC-4 对目标蛋白 VEGFR2 和 PDGFRα 具有双重抑制作用,是一种具有抗癌活性的化合物。分离化合物的体外细胞活力测定表明,AC-1 的 IC50(μg/mL)值为 34.96±3.91、47.76±3.80、69.1±4.96,AC-2 为 68.26±4.22、54.03±5.14、>100,AC-3 为 35.34±4.14、51.5±51.5、70.8±5.25,AC-4 为 44.2±3.57、24.2±2.67、51.2±2.54,分别用于 MDA-MB-231、MCF-7 和 PC-3 癌细胞系,与标准药物阿霉素相比。此外,荧光显微镜证实了化合物的促凋亡特性。我们还发现,AC-4 在乳腺癌细胞中产生了显著的细胞内 ROS 生成,从而诱导凋亡,最终导致细胞死亡。
结论:总之,A. cadamba 提供了四个纯分子 AC-1 至 AC-4,并鉴定了 14 种新化合物。整个计算机模拟研究得出结论,AC-4 化合物在口服生物利用度、生物活性评分和 ADMET 谱方面优于研究中的其他分子。分子对接分析和 MD 模拟也支持 AC-4 对 VEGFR2 和 PDGFRα 受体的双重抑制作用。此外,分离得到的分子 AC-1、AC-2、AC-3 和 AC-4 对 MDA-MB-231、MCF-7 和 PC-3 癌细胞均有活性。分子 AC-4 被发现能诱导乳腺癌细胞中的 ROS 介导的凋亡。结果表明,AC-4 的抗癌抑制潜力与其分子立体化学有关,它能特异性地与乳腺癌细胞的靶蛋白结合,而没有毒理学作用。因此,AC-4 被建议作为一种有效的新型药物设计和发现的候选物。
Nat Prod Commun. 2015-8