Next Article in Journal
Insight into Mechanobiology: How Stem Cells Feel Mechanical Forces and Orchestrate Biological Functions
Previous Article in Journal
The Yin-Yang Regulation of Reactive Oxygen Species and MicroRNAs in Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Synthesis and Evaluation of Anticancer Activity of New 4-Acyloxy Derivatives of Robustic Acid

1
School of Chemistry and Chemical Engineering, Guangxi University, Nanning 530004, China
2
Faculty of Chinese Medicine Science, Guangxi University of Chinese Medicine, Nanning 530222, China
3
College of Pharmacy, Guangxi University of Chinese Medicine, Nanning 530222, China
4
Center for Excellence in Post-Harvest Technologies, North Carolina A&T State University, The North Carolina Research Campus, 500 Laureate Way, Kannapolis 28081, NC, USA
5
Guangxi Institute of Botany, Chinese Academy of Sciences, Guilin 541006, China
6
College of Pharmacy, Guangxi Medical University, Nanning 530021, China
*
Author to whom correspondence should be addressed.
These authors contributed equally.
Int. J. Mol. Sci. 2019, 20(21), 5336; https://doi.org/10.3390/ijms20215336
Submission received: 29 September 2019 / Revised: 20 October 2019 / Accepted: 21 October 2019 / Published: 26 October 2019
(This article belongs to the Section Molecular Pharmacology)

Abstract

:
In the present study, a series of 4-acyloxy robustic acid derivatives were synthesized and characterized for evaluation of their anti-cancer activity. The structures of these derivatives were elucidated by mass spectra (MS) nuclear magnetic resonance spectra (NMR). The single-crystal X-ray diffraction structure of one of these compounds was obtained, for further validation of the target compound structures. The anticancer activities of the target products were evaluated against human leukemic cells HL-60, human non-small cell lung carcinoma cells A-549, human hepatic carcinoma cells SMMC-7721, human hepatocellular carcinoma cells HepG2, and human cervical carcinoma cells Hela. Three compounds among them exhibited potent in-vitro cytotoxicity and excellent DNA topoisomerase I inhibitory activity, even at 0.1 mM concentrations. The most noteworthy observation was the minor toxicity of two of these compounds to normal cells, with an activity similar to the positive control in cancerous cells. A Surflex-Dock docking study was performed to investigate the topoisomerase I activity of all compounds. Of all the other compounds, the most sensitive compound was selected for further investigation of its effect on apoptosis induction and cell cycle regulation in HL-60 cells. Our results suggest that the anticancer effects of these compounds can be attributed to their pharmacological effects on topoisomerase I, cell apoptosis, and cell cycle. These findings suggest that robustic acid derivatives could be used as potential antitumor drugs.

1. Introduction

Coumarin compounds are widely distributed in nature, especially in higher plants [1]. They are proven to possess a wide range of biological activities, with minimal side effects [2]. In particular, pyranocoumarins (including linear type and angular type) are an interesting class of fused coumarins. They are important as photoactive drugs with important pharmacological effects that include anti-inflammatory [3], anti-HIV-1 [4], anti-hepatitis B virus [5], anti-cancer [6], and antinociceptive properties [7]. As the major constituents from Clausena or Angelica genus plants [8,9,10,11], some linear-type pyranocoumarins (shown in Figure 1) with anti-cancer property have become compounds of growing research interest. Examples of such compounds are, clausenidin (1), nordentatin (2), clausarin (3), dentatin (4), decursin (5), decursinol angelate (6), etc. Compounds 1–3 mentioned above are reported to exhibit potent cytotoxicity against human non-small cell lung carcinoma cells (A549), human breast adeno carcinoma cells (MCF-7), human nasopharyngeal carcinoma cells (KB), and multidrug-resistant nasopharyngeal carcinoma cells (KB-VIN) [5]. Compound 4 had significant antitumor activity on MCF-7, human hepatocellular carcinoma cells (HepG2), and human prostate carcinoma cells (PC-3 and LNCaP) through causing the induction of apoptosis [12,13,14]. Compounds 5 and 6 are more cytotoxic to leukemia cancer cells [15,16]. However, detailed information on the cytotoxic mechanism and structure-activity relationships of these linear-type pyranocoumarins is still limited in the literature. Considering the wide spectrum of biological activities of these compounds, the synthesis of new linear pyranocoumarins has drawn significant attention in recent years.
Recently, our group isolated and identified a new linear pyranocoumarin was from Dalbergia benthamii Prain: robustic acid (RA; Figure 2). RA constitutes to about 3.5% w/w of its dry matter and was considered to be one of the major bioactive phytoconstituents of D. benthamii [17]. D. benthamii is used commonly in traditional medicine for the treatment of traumatic injury, as a pain reliever and anti-inflammatory agent, and for regulating menstrual cycle [18]. To further explore and continue our earlier investigation on linear pyranocoumarins, the synthesis of a new series of 4-acyloxy compounds of robustic acid derivatives was carried out. Preliminary cytotoxic activities and related mechanisms such as DNA topoisomerase I (Topo I), cell cycle, and apoptosis were also investigated in this study.

2. Results and Discussion

2.1. Chemistry

The scheme used for the synthesis of 4-acyloxy robustic acid derivatives (2a2j) is illustrated in Scheme 1. The starting material RA was isolated from D. benthamii in our previous study [17]. The target compounds 2a2j were synthesized by nucleophilic substitution from RA and derivatives of acyl chloride. We found that the yields of aromatic acyl chloride compounds were higher than the fatty acyl chloride compounds, which ranged from 41–85%. With the attachment of the withdrawing group on aromatic ring, the reaction time was shortened to less than 1 h and the yield was greatly improved. The structures of compounds were elucidated by, MS, 1H NMR, and 13C NMR. All spectral data were in accordance with the assumed structures, and are listed in the procedure. The ESI-MS indicated that the molecular weights were in accordance with the calculated values. In 1H NMR spectra, the single peaks of two methoxyl groups of the original nucleus appeared at δ 3.79 and δ 3.56, and six hydrogen atom signals of two methyl groups were observed at δ 1.47–1.59 ppm. Two pyran ring hydrogen signals were in the ranges δ 6.02–6.58 and δ 5.50–8.74, respectively. Further verification of the structures of the target products was carried out, and a single crystal for 2f(CCDC 1944893, these data can be obtained free of charge via http://www.ccdc.cam.ac.uk/conts/retrieving.html or the Cambridge Crystallographic Data Centre, 12 Union Road, Cambridge CB2 1EZ, UK; fax: (+44) 1223-336-033; or e-mail: [email protected].) was obtained in ethanol solution. The molecular structure of 2f is shown in Figure 3, and the packing diagram of 2f is depicted in Figure 4. The formation of a twin crystal was observed in the crystal structure of 2f. Most bond lengths in the system fell in the range of single and double bonds and the C–C bond distances in all rings ranged between 1.316(5) and 1.498(6) Å. These values are almost equal to the values of typical bonds of aromatic structure. The crystal packing of the compound is stabilized by π–π stacking.

2.2. Biological Study

2.2.1. Anti-Proliferative Activity Screening

All the synthesized compounds were evaluated for their cytotoxicity effects in five human cancer cell lines and two normal cells. The cell lines included: leukemia cells HL-60, human non-small cell lung carcinoma cells A549, human hepatic carcinoma cells SMMC-7721, hepatocellular carcinoma cells HepG2, cervical carcinoma cells Hela, human normal liver cells LO2, and human normal lung epithelial cells BEAS-2B. The obtained IC50 values of the synthesized compounds, compared to RA and the reference drug (cisplatin and camptothecin (CPT)), are shown in Table 1. Interestingly, the activities of most of the target products were superior to the parent nucleus. Compounds 2d, 2g, and 2i exhibited promising biological activity in almost all cell lines. Of all the cell lines selected, they were more sensitive to HL-60, with IC50 values of 21.04 ± 0.43, 16.63 ± 0.12, and 16.38 ± 0.27 μM, respectively. The obtained IC50 values were close to the positive control cisplatin (14.23 ± 0.56) and CPT (12.25 ± 1.06). In addition, 2d and 2g exhibited moderate activity in Hela cells, with IC50 values of 37.03 ± 0.97 and 27.45 ± 0.38 μM, respectively. Their potential antitumor activity might be related to the attracting group on benzene ring of 4-aroyl. It should be noted that most target compounds could significantly inhibit the proliferation of selected cancerous cells with no damage to normal cells (LO2 and BEAS-2B (IC50 > 100 μM)). However, compound 2g has detrimental effects on LO2 cells, as it exhibited an IC50 value of 20.79 ± 0.97 μM. All compounds were less toxic on non-cancerous cells than positive control. This indicates that these compounds (especially 2d and 2i) have highly selective activity against cancerous cells and are non-toxic to normal human cell lines. They can thus be suggested as potential candidates for development into anti-tumor drugs with high efficiency and low toxicity.

2.2.2. Evaluation of DNA Topoisomerase I (Topo I) Inhibitory Activity and Molecular Docking Study

DNA Topo I is an important enzyme in all living organisms, and it participates in many cellular metabolic processes, such as replication, transcription, recombination, and repair [19]. DNA Topo I has been established as a molecular target of anticancer drugs. A combined study of synthesis and docking for the anti-cancer activity of novel coumarins concluded that coumarin derivatives are potent cytotoxic agents. Docking studies further suggest that all coumarins possess higher binding affinities with DNA Topo I [20]. Currently, discovering novel non-camptothecin inhibitors for DNA Topo I has become an important area of research. Thus, a DNA Topo I inhibitory activity assay was also carried out using the supercoiled DNA unwinding method in this study [21]. Figure 5A shows the conversion of supercoiled plasmid DNA Topo I in the presence of compounds RA and 2a to 2j with camptothecin (CPT) as a positive control. Compounds 2d, 2g, and 2i showed significant inhibitory activities at 10 mM concentration. Compounds 2b, 2c, 2f, and 2h showed moderate inhibitory activities with obscure strips in electrophoresis. Compounds 2a, 2e, and 2j did not show any considerable Topo I inhibitory activity. Therefore, compounds 2d, 2g, and 2i were selected for further quantitative investigation (depicted in Figure 5B). It was found that the inhibition weakened gradually as the concentration of RA reduced. At critical concentrations lower than 10 mM, no inhibitory activity was found. In agarose gel electrophoresis, DNA fragmentation was evident with treatment of 2d, 2g, and 2i, even at low concentrations of 1 mM or 0.1 mM. Note that 2g and 2i exhibited Topo I inhibitory activities higher than that of compound 2d. This correlates well with the cytotoxicity to HL-60, A-549, and SMMC-7721 cells. Thus, we infer that the anti-proliferative activity of 4-acyloxy derivatives of RA to these three cells may be related to Topo I.
To understand the mechanism of Topo I inhibitory activity of the compounds synthesized, molecular docking studies were carried using the Surflex-Dock algorithm of Sybyl-X 2.0 (Tripos Inc., St. Louis, MO, USA). The crystal structure of DNA Topo I in complex with CTP was downloaded from the Protein Data Bank (PDB code: 1T8I). To validate the molecular docking approach, the crystallographic pose of CTP (derived from complex) and the top-ranked docking pose obtained in this study were compared. The two binding poses of CTP had almost perfect superposition, with an RMSD (root-mean-square deviation) value of 0.4438 Å (Figure 6). The obtained RMSD value is far below the well-established tolerance level of 2.0 Å, thus validating the adopted docking methodology [22,23].
After running Surflex-Dock, the binding affinities of protein–ligand complexes were obtained and expressed as total score. All 10 inhibitors were docked into the active site of Topo I, as shown in Figure 7. The predicted binding affinities (total score) of all the compounds containing hydrophobic residue, hydrogen bond, and π–π stacking are listed in Table 2. The results indicate that all synthetic compounds except 2b had better combination with Topo I than the RA mother nucleus. Compound 2g exhibited the highest total score value of 10.02, which was close to the positive control (CPT). The most important residues on the 2g–Topo I complex (Figure 4h) were ILE427, TGP11, DT10, DA113, ARG364, DC112, PRO357, GLU356, LYS374, GLU418, and TYR426. As depicted, the oxygen atom of acyl group of compound 2g made a hydrogen bonding interaction with the hydrogen of the -NH2 group of LYS425 (O–H-LYS425, 2.76 Å). The oxygen atoms in the –NO2 group made three binding interactions with hydrogens of the -NH2 group of MET428 (O–H-MET428, 2.72, 3.06, and 3.33Å). Compounds 2d and 2i also exhibited good binding affinities, with total scores of 8.76 and 8.89. All these compounds exhibited remarkable Topo I inhibitory activity even at low concentration, which is nearly in agreement with our observations of in vitro studies. However, the results of the docking assay did not exactly correlate with the Topo I inhibitory activity. The binding affinity of all compounds were decreasing in sequence of 2g, 2h, 2i, 2c, 2d, 2e, 2f, 2a, 2j, 2b. However, the inhibitory activities of 2h and 2c were not as good as the prediction data. This may be related to other combination modes or other pharmacological mechanisms.

2.2.3. Apoptosis and Cell-Cycle Analysis

Cell-cycle arrest and apoptosis are widely accepted as the main mechanisms of inhibition in tumor formation [19,20]. As HL-60 cells were the most sensitive to all tested compounds, they were selected for testing the most active compound 2i (at 6.25-, 12.5-, and 25-μM doses), for their effects on apoptosis and cell cycle profile after 48 h. The induction of apoptosis by 2i was evident from the nuclear morphologic alterations. These were observed by confocal microscopy after labeling the cells with acridine orange (AO)/ethidium bromide (EB). Representative images from four independent experiments are shown in Figure 8. Compared to the control group, chromatin condensation, nuclear fragmentation, and apoptotic bodies were observed in treated cells at concentrations of 6.25, 12.5, and 25 μM. At the highest concentration of 25 μM, the necrosis cell volume was increased, showing uneven orange-red fluorescence and an unapparent outline.
The extent of apoptosis by flow cytometric analysis of the HL-60 cells labeled with Annexin V-APC/7-AAD was further quantified. For this study, treatment with 2i at doses of 6.25, 12.5, and 25 μM for 48 h was carried out. Such treatments resulted in 34.61%, 52.52%, and 66.17% apoptotic cells, respectively (Figure 9). The apoptosis ratio for control was found to be 1.46%. The obtained results thus indicate that compound 2i suppressed cell proliferation by inducing apoptosis, and a considerable dose dependence was observed at concentrations of 6.25, 12.5, and 25 μM.
Since the induction of apoptosis may be mediated through the regulation of cell cycle, we also examined the effect of 2i on cell cycle perturbations labeled with propidium iodide (PI). Compared to control, 2i treatment resulted in an appreciable arrest of HL-60 cells in the G1 phase of the cell cycle, after 48 h of the treatment (Figure 10). The treatment caused an arrest of 56.48% cells in G1 phase of the cell cycle at a 6.25 µM dose, which further increased to 63.40%and 68.29% at higher doses of 12.5 and 25 µM. However, this increase in G1 cell population was accompanied by a decrease of cell number in S and G2 phases. These results indicated that compound 2i induced a significant cell cycle arrest in G1 phase in a concentration-dependent manner.

3. Materials and Methods

All chemicals used in the study were reagent grade and are commercially available. All yields refer to the yields of isolated products after purification. NMR spectra were measured on a Bruker DRX-500 (1H: 500 MHz, 13C: 126 MHz) using CDCl3 as solvent. Chemical shifts (d) are expressed in parts per million (ppm), and J values are given in hertz (Hz). The mass spectra (MS) were obtained on an Agilent 6210 ESI/TOF MS instrument. Melting points were determined using an X-4 apparatus, and were uncorrected. Samples of compounds 2a–2i are available from the authors.

3.1. Synthesis Methods

General Procedure for the Synthesis of 4-Acyloxy Derivatives of Robustic Acid 2a2j

Robustic acid (0.19 g, 0.5 mmol) was dissolved in CH3CN (10 mL), followed by the addition of acyl chloride derivative (1 mmol) and pyridine (0.161 μL). After 15–210 min refluxing with stirring (monitored by thin-layer chromatography), the reaction mixture was cooled to room temperature. The solvent was removed and recrystallization with ethanol was carried out. The formed crystals (2a2j) were filtered and dried under vacuum.
4-O-Benzoyl-robustic acid (2a): reaction time 2.5 h, white powder, isolated yield 67%, m.p. 266.4–273.4 °C. 1H NMR (500 MHz, CDCl3) δ 8.09–8.02 (m, 2H, ArH), 7.64 (t, J = 7.4 Hz, 1H, ArH), 7.50 (q, J = 7.9, 7.3 Hz, 2H, ArH), 7.42 (d, J = 8.8 Hz, 1H, ArH), 6.89 (d, J = 8.8 Hz, 2H, ArH), 6.70 (s, 1H, ArH), 6.49 (d, J = 10.1 Hz, 1H, pyran ring), 5.70 (d, J = 10.1 Hz, 1H, pyran ring), 3.79 (s, 3H, -OCH3), 3.56 (s, 3H, -OCH3), 1.49 (s, 6H, 2-CH3); 13C NMR (126 MHz, CDCl3) δ 165.53, 163.48, 161.83, 159.52, 157.56, 154.58, 154.30, 133.79, 131.19, 130.77, 130.72, 130.14, 130.12, 128.66, 128.62, 123.75, 122.55, 117.12, 115.88, 115.83, 113.71, 110.48, 108.18, 101.28, 101.12, 63.55, 55.21, 28.21, 28.16. HR-MS (ESI) m/z: calculated for C29H24O7 [M + H]+: 485.1595, found: 485.1599.
4-O-Trichloroacetyl-robustic acid (2b): reaction time 2.0 h, white powder, isolated yield 41%, m.p. 218.2–223.1 °C. 1H NMR (500 MHz, CDCl3) δ 7.34 (d, J = 8.3 Hz, 1H, ArH), 7.29 (s, 1H, ArH), 6.97 (d, J = 8.3 Hz, 2H, ArH), 6.70 (s, 1H, ArH), 6.58 (d, J = 10.0 Hz, 1H, pyran ring), 5.77 (d, J = 10.1 Hz, 1H, pyran ring), 3.87 (s, 3H, -OCH3), 3.84 (s, 2H, -OCH3), 1.51 (s, 6H, 2-CH3); 13C NMR (126 MHz, CDCl3) δ 161.23, 160.10, 158.72, 158.15, 154.13, 153.71, 152.56, 131.39, 131.26, 130.99, 124.02, 121.15, 115.84, 114.02, 113.95, 112.63, 110.54, 108.50, 102.66, 101.31, 64.42, 55.35, 28.20, 28.00. HR-MS (ESI) m/z: calculated for C24H19Cl3O7 [M + H]+: 525.0269, found: 525.0267.
4-O-(p-Ethoxy benzoyl)-robustic acid (2c): reaction time 2.0 h, white crystal, isolated yield 56%; m.p. 226.3–228.7 °C. 1H NMR (500 MHz, CDCl3) δ 7.99 (d, J = 8.9 Hz, 2H, ArH), 7.41 (d, J = 8.8 Hz, 2H, ArH), 6.93 (d, J = 8.9 Hz, 2H, ArH), 6.87 (d, J = 8.8 Hz, 2H, ArH), 6.71–6.64 (m, 1H, ArH), 6.49 (d, J = 9.8 Hz, 1H, pyran ring), 5.69 (d, J = 10.1 Hz, 1H, pyran ring), 4.11 (q, J = 7.0 Hz, 2H, -OCH2), 3.76 (s, 3H, -OCH3), 3.58 (s, 3H, -OCH3), 1.47 (s, 6H, 2-CH3), 1.47 – 1.41 (m, 3H, -CH3); 13C NMR (126 MHz, CDCl3) δ 163.56, 163.24, 161.90, 159.46, 157.50, 154.87, 154.27, 152.38, 132.35, 132.32, 131.21, 130.69, 123.77, 122.69, 120.49, 117.05, 115.92, 114.39, 114.38, 113.64, 112.71, 110.50, 108.12, 104.20, 101.15, 63.88, 63.61, 55.17, 28.18, 28.13, 14.65. HR-MS (ESI) m/z: calculated for C31H28O8 [M + H]+: 529.1857, found: 529.1861.
4-O-(p-Trifluoromethyl benzoyl)robustic acid (2d): reaction time 1.0 h, white crystal, isolated yield 81%; m.p. 225.6–228.5 °C. 1H NMR (500 MHz, CDCl3) δ 1H NMR (500 MHz, CDCl3) δ 8.16 (d, J = 7.9 Hz, 2H, ArH), 7.77 (d, J = 8.1 Hz, 2H, ArH), 7.40 (d, J = 8.8 Hz, 2H, ArH), 6.90 (d, J = 8.9 Hz, 2H, ArH), 6.71 (s, 1H, ArH), 6.48 (d, J = 10.1Hz, 1H, pyran ring), 5.72 (d, J = 10.1 Hz, 1H, pyran ring), 3.79 (s, 3H, -OCH3), 3.55 (s, 3H, -OCH3), 1.50 (s, 6H, -CH3); 13C NMR (126 MHz, CDCl3) δ 162.30, 161.62, 159.66, 157.88, 157.72, 154.53, 154.27, 154.09, 152.03, 131.11, 130.99, 130.95, 130.47, 130.44, 130.44, 125.79, 125.75, 125.72, 125.69, 123.64, 122.30, 117.18, 115.70, 115.66, 113.78, 112.73, 112.69, 110.37, 108.24, 103.50, 101.38, 101.21, 63.53, 55.22, 28.24, 28.14. HR-MS (ESI) m/z: calculated for C30H23F3O7 [M + H]+: 553.1469, found: 553.1469.
4-O-Thenoyl robustic acid (2e): reaction time 2.0 h, brown powder, isolated yield 46%; m.p. 171.5–182.3 °C. 1H NMR (500 MHz, CDCl3) δ 7.37–7.20 (m, 3H, ArH), 6.99–6.93 (m, 1H, ArH), 6.89 (d, J = 8.3 Hz, 2H, ArH), 6.83 (s, 1H, ArH), 6.67 (s, 1H, ArH), 6.57 (d, J = 10.1 Hz, 1H, pyran ring), 5.75 (d, J = 10.1 Hz, 1H, pyran ring), 3.94 (s, 3H, -OCH3), 3.86 (s, 3H, -OCH3), 3.78 (s, 3H, -OCH3), 1.50 (s, 6H, -CH3); 13C NMR (126 MHz, CDCl3) δ 167.02, 161.66, 159.62, 157.63, 154.25, 154.22, 152.18, 131.05, 130.94, 127.52, 126.95, 125.38, 123.62, 122.30, 117.07, 115.76, 113.80, 112.64, 110.37, 108.28, 103.62, 101.33, 63.59, 55.29, 34.71, 28.20, 28.19. HR-MS (ESI) m/z: calculated for C27H22O7S [M+Na]+: 513.0978, found: 513.0985.
4-O-(p-Fluorobenzoyl)robustic acid (2f): reaction time 50 min, white crystal, isolated yield 63%; m.p. 236.8–239.6 °C. 1H NMR (500 MHz, CDCl3) δ 8.14–7.99 (m, 2H, ArH), 7.40 (d, J = 8.8 Hz, 2H, ArH), 7.16 (t, J = 8.6 Hz, 2H, ArH), 6.89 (d, J = 8.8 Hz, 2H, ArH), 6.70 (s, 1H, ArH), 6.49 (d, J = 10.0 Hz, 1H, pyran ring), 5.71 (d, J = 10.1 Hz, 1H, pyran ring-H), 3.78 (s, 3H, -OCH3), 3.57 (s, 3H, -OCH3), 1.49 (s, 6H, -CH3); 13C NMR (126 MHz, CDCl3) δ 167.26, 165.22, 162.51, 161.74, 159.57, 157.63, 154.41, 154.27, 152.18, 132.79, 132.72, 131.15, 130.85, 124.78, 123.70, 122.48, 117.13, 116.05, 115.87, 115.79, 113.71, 112.69, 110.42, 108.18, 103.82, 101.28, 63.55, 55.20, 28.21, 28.14. HR-MS (ESI) m/z: calculated for C29H23FO7 [M + H]+: 503.1501, found: 503.1504.
4-O-(p-Nitrobenzoyl)robustic acid (2g): reaction time 15 min, yellow powder, isolated yield 85%; m.p. 239.5–241.9 °C. 1H NMR (500 MHz, CDCl3) δ 8.35 (d, J = 8.8 Hz, 2H, ArH), 8.22 (d, J = 8.8 Hz, 2H, ArH), 7.39 (d, J = 8.8 Hz, 2H, ArH), 6.90 (d, J = 8.8 Hz, 2H, ArH), 6.72 (s, 1H, ArH), 6.47 (d, J = 10.2 Hz, 1H, pyran ring), 5.73 (d, J = 10.2 Hz, 1H, pyran ring), 3.79 (s, 3H, -OCH3), 3.55 (s, 3H, -OCH3), 1.50 (s, 6H, -CH3); 13C NMR (125 MHz, CDCl3) δ 167.26, 165.22, 162.51, 161.74, 159.57, 157.63, 154.41, 154.27, 152.18, 132.79, 131.15, 130.85, 124.78, 123.70, 122.48, 117.13, 116.05, 115.87, 115.79, 113.71, 112.69, 110.42, 108.18, 103.82, 101.28, 63.55, 55.20, 28.21, 28.14. HR-MS (ESI) m/z: calculated for C29H23NO7 [M + H]+: 530.1446, found: 530.1451.
4-O-(p-Ethylbenzoyl)robustic acid (2h): reaction time 1.5 h, white powder, isolated yield 65%; m.p. 246.9–250.2 °C. 1H NMR (500 MHz, CDCl3) δ 7.97 (d, J = 7.9 Hz, 2H), 7.41 (d, J = 8.4 Hz, 2H, ArH), 7.31 (d, J = 7.9 Hz, 2H, ArH), 6.89 (d, J = 8.4 Hz, 2H, ArH), 6.70 (s, 1H, ArH), 6.49 (d, J = 10.1 Hz, 1H, pyran ring), 5.69 (d, J = 10.1 Hz, 1H, pyran ring), 3.79 (s, 3H, -OCH3), 3.57 (s, 3H, -OCH3), 2.75 (q, J = 7.6 Hz, 2H, -CH2), 1.49 (s, 6H, -CH3), 1.30 (t, J = 7.6 Hz, 3H, -CH3); 13C NMR (125MHz, CDCl3) δ 13C NMR (126 MHz, CDCl3) δ 163.53, 161.89, 159.47, 157.51, 154.72, 154.29, 152.31, 150.84, 131.21, 130.67, 130.30, 128.15, 125.97, 122.64, 117.10, 115.91, 115.87, 113.69, 112.67, 104.05, 101.24, 63.60, 55.20, 29.02, 28.18, 15.01. HR-MS (ESI) m/z: calculated for C31H28O7 [M + H]+: 513.1908, found: 513.1906.
4-O-(4-Bromo-2-fluorobenzoyl) robustic acid (2i): reaction time 30 min, white powder, isolated yield 71%; m.p. 250.8–251.9 °C. 1H NMR (500 MHz, CDCl3) δ 7.82 (t, J = 8.1 Hz, 1H, ArH), 7.51–7.33 (m, 4H, ArH), 6.91 (d, J = 8.5 Hz, 2H, ArH), 6.70 (s, 1H, ArH), 6.50 (d, J = 10.1 Hz, 1H, pyran ring), 5.72 (d, J = 10.1 Hz, 1H, pyran ring), 3.81 (s, 3H, -OCH3), 3.61 (s, 3H, -OCH3), 1.50 (s, 6H, -CH3); 13C NMR (125 MHz, CDCl3) δ 162.79, 161.68, 160.67, 160.13, 160.09, 159.63, 157.66, 154.24, 153.79, 152.11, 133.64, 131.28, 130.86, 127.94, 127.91, 123.86, 122.27, 121.09, 120.88, 117.13, 115.77, 113.69, 112.68, 110.51, 108.17, 103.56, 101.31, 101.18, 63.44, 55.23, 28.26, 28.13. HR-MS (ESI) m/z: calculated for C29H22BrFO7 [M+Na]+: 603.0425, found: 603.0429.
4-O-Acetylrobustic acid (2j): reaction time 3.5 h, white crystal, isolated yield 51%; m.p. 189.9–192.1 °C (lit. [24], m.p. 198–199 °C; lit. [25], m.p. 194–196 °C). 1H NMR (600 MHz, CDCl3) δ 7.35 (d, J = 8.7 Hz, 2H, ArH), 7.02–6.92 (m, 2H, ArH), 6.88 (d, J = 7.9 Hz, 1H, ArH), 6.67 (s, 1H, ArH), 6.57 (d, J = 7.8 Hz, 1H, ArH), 6.02 (s, 1H, pyran ring), 5.74 (d, J = 10.1 Hz, 1H, pyran ring), 3.86 (s, 3H, -OCH3), 3.82 (s, 3H, -OCH3), 2.17 (s, 3H, -COCH3), 1.51 (s, 6H, -CH3); 13C NMR (150 MHz, CDCl3)δ 167.54, 161.74, 159.63, 157.55, 154.28, 154.21, 152.08, 131.11, 130.96, 123.64, 122.63, 116.94, 115.73, 115.69, 113.77, 112.65, 110.39, 108.24, 103.74, 101.35, 101.22, 63.52, 55.28, 28.19, 28.00, 20.46. HR-MS (ESI) m/z: calculated for C24H22O7 [M+Na]+: 445.1258, found: 445.1256.

3.2. Biological Activity

3.2.1. Antiproliferative Activity

The MTS (3-(4,5-dimethylthiazole-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H- tetrazolium) (Promega, Madison, WI, USA) assay was used to determine the IC50 value for all compounds, according to [26]. We used a panel of five human carcinoma cell lines and two normal cells including HL-60, A549, SMMC-7721, HepG2, Hela, LO2 and BEAS-2B. Cells were seeded in 96-well plates at a density of 3 × 103–1.5 × 104 cells/well in 100 μL medium (DMEM or RMPI1640) and incubated for 12–24 h at 37 °C. Subsequently, cells were exposed to drugs in an additional 100 μL medium and were further incubated at 37 °C for 48 h. In brief, 100 μL MTS reagent supplement with 100 μL medium was added directly to the wells and cells were incubated at 37 °C for a minimum of 2 h. Colorimetric measurements were recorded at 492 nm. Background absorbance was corrected using triplicate sets of wells, and cells were incubated at 37 °C for a minimum of 2 h. Colorimetric measurements were recorded at 492 nm. Background absorbance was corrected using triplicate sets of wells containing medium only (no cells), and MTS reagent was added per experimental well. Each experiment was performed at least three times, and values in results are represented as mean ± SD.

3.2.2. Topo I Inhibitory Activity

Supercoiled pBR32 DNA was used as a substrate to determine the Topo I catalytic activity. Commercial samples of Topo I and pBR322 were obtained from Takara Bio Inc. The buffer solutions were prepared using 500 mM KAc, 200 mM Tris-Ac, 100 mM Mg(Ac)2, and 1 mg/mL BSA. The procedure used for determination of enzyme inhibitory activity was similar to one reported by Xu et al. [27].

3.2.3. General Procedure for AO/EB Staining

Apoptosis was determined by nuclear morphology. Cells were fixed and stained with AO/EB according to the manufacturer’s instruction (KGA501, KeyGEN BioTECH, Nanjing, China). HL-60 cells were seeded in six-well plates with sterile cover slips. The concentration of cells used per well was 5 × 105–6 × 106 cells/2 mL. To facilitate growth, the medium was replaced with fresh medium (RPMI1640) plus 10% fetal bovine serum. The fresh medium was supplemented with compound 2i (6.25, 12.50, and 25 μM). Post treatment, 25 μL of cell suspension was collected on a glass slide by inverting the cover slip and stained with 10 μL of AO/EB stain (100 mg/mL). Finally, stained nuclei were observed immediately under a fluorescence microscope (BX41, Nikon, Tokyo, Japan).

3.2.4. General Procedure for Apoptosis Ratio Determination

For determination of apoptosis ratios by application of flow cytometry analysis, the manufacturer’s protocol (KGA1024, KeyGEN BioTECH, Nanjing, China) for Annexin-V APC/7-AAD double-stain assay was used. The HL-60 cells were grown at a density of 5 × 105 cells/mL in six-well plates (Costar), and were treated with 2i (6.25, 12.50, and 25 μM) for 72 h. The cells were trypsinized and washed with PBS. Subsequently, the prepared HL-60 cells (5 × 105 cells/mL) were collected and resuspended in 500 μL of binding buffer containing 5 μL of Annexin V-APC. The suspension was shaken well, and 5 μL of 7-AAD was added to it. The suspension was then incubated for 5–15 min in the dark at room temperature. The suspensions were then analyzed by flow cytometry (FACS Cali-bur, Becton Dickinson, San Jose, CA, USA).

3.2.5. General Procedure for Cell Cycle Analysis

The intercalating blue-excited dye propidium iodide (PI) was used in this study. HL-60 cell cultures were seeded within six-well plates (Costar). Then, the cells were treated with 2i (6.25, 12.50, and 25 μM) in DMEM complete medium for 72 h. After the incubation time elapsed, the harvest procedure was performed as follows: the supernatant medium was transferred to centrifuge tubes. To the medium, 0.25% trypsin (Sigma) was added, until all cells were detached. The cell suspension was then washed twice with PBS and resuspended. About 1 mL of 70% ethanol was quickly added to the cell suspension (5 × 105 cells/mL), which was then incubated at 4 °C overnight. The samples were treated with 100 μL RNase A at 37 °C for 30 min and then stained with 400 μL PI solution. After another 30 min of incubation at 4 °C, the resulting nuclei suspension was analyzed with a flow cytometer (FACS Verse, Becton Dickinson, San Jose, CA, USA) at 488 nm excitation for red fluorescence of PI.

3.3. Molecular Docking

Surflex-Dock in Sybyl 2.0 was used for molecular docking. This system performs molecular docking functions aided by the generation of an idealized active site (Protocol), consisting of dummy atoms that guide the docking process. The crystal structures of DNA-Topo I complex was downloaded from the RCSB website (https://www.rcsb.org/structure/1T8I) (PDB ID: 1T8I). The proteins were then imported into the Surflex-Dock and prepared for docking using the biopolymer preparation tool, according to the following criteria: H-Addition, H-Bond, removal of water molecules, termini treatment, charge, protonation.
The structures of the compounds were sketched using ChemBioOffice version 14.0 by PerkinElmer (Waltham, MA, USA). The alignments of the training set molecules were derived using FlexS in SYBYL. All values were filled with valence, and Gasteiger–Marsili charges were calculated for each compound. Ultimately, ligand docking under the Surflex-Dock GeomX precision was performed to generate a grid of proteins and ligands. The docking results were then imported into LigPlot+ 2.1, and the combinations between compounds and proteins were discussed in terms of H-Bond, conjugate action, and hydrophilic or hydrophobic action.

4. Conclusions

A new series of linear pyranocoumarin derivatives were synthesized, and their anti-cancer activities were evaluated. The biological screening results indicated that most of the synthesized compounds exhibited potent in vitro anti-cancer activity, which were better than original nucleus robustic acid. Among them, compounds 2d, 2g, and 2i were showed excellent anti-cancer activity with respective IC50 values of 21.04 ± 0.43, 16.63 ± 0.12, and 16.38 ± 0.27 μM against HL-60 cells. These values were estimated in comparison to the positive control cisplatin (14.23 ± 0.56 μM) and CPT (12.25 ± 1.06 μM). And these compounds were much less toxic to non-cancerous cells when comparing with the two positive controls. In addition to this, docking studies of 2d, 2g, and 2i were performed, and they all exhibited good binding affinities with relatively high total scores of 8.76, 10.02, and 8.89. The inhibitory activities of the compounds against Topo I indicated that 2g and 2i were showed significant inhibitory activity, even at the low concentration of 0.1 mM. Compound 2d aslo exhibited moderate inhibitory activity. These results indicate that the anti-tumor activity of these three compounds may be related to Topo I inhibitory activity. However, the results of the docking assay of other compounds do not exactly correlate with the Topo I inhibitory and anti-tumor activity. This may be the result of other pharmacological mechanisms. Later, compound 2i was selected as the representative compound for further investigation, due to its properties of suppressing cell proliferation by inducing apoptosis and arresting HL-60 cells in the G1 stage. These findings may contribute to the identification of novel linear pyranocoumarins that can serve as the new lead compounds for anti-tumor drugs.

Author Contributions

L.H. and J.W. contributed to the separation and synthesis of compounds. R.C., J.Z., X.X., and Y.L. performed pharmacological activity testing and docking study of the target products. L.H. and R.C. wrote the manuscript. D.L. contributed to the project proposal and executing the experiment. Y.J. and L.W. contributed to improving the manuscript, providing intellectual input, and editing the language of the manuscript.

Funding

This research received no external funding.

Acknowledgments

This work acknowledges the support of the National Natural Science Foundation of China (No. NSFC81603525, and No. NSFC21562009); Guangxi Natural Science Foundation (No.2017GXNSFAA198171 and 2018GXNSFAA281189); State Key Laboratory Cultivation Base for the Chemistry and Molecular Engineering of Medicinal Resources, Ministry of Science and Technology of China (CMEMR2015-B04 and CMEMR2015-B05); Promotion project of basic ability of young and middle-aged teachers in colleges and universities in Guangxi (No.2017KY0283); Hundred Talents Plan for the Introduction of High-Level Overseas Talents by Guangxi Colleges and Universities; Supported by the Fund of Guangxi Key Laboratory of Functional Phytochemicals Research and Utilization (No.FPRU2016-9).

Conflicts of Interest

The authors declare no conflict of interest in association with this manuscript.

References

  1. Klenkar, J.; Molnar, M. Natural and synthetic coumarins as potential anticancer agents. J. Chem. Pharm. Res. 2015, 7, 1223–1238. [Google Scholar]
  2. Xia, L.X.; Wang, Y.B.; Huang, W.L.; Qian, H. Research advance of anticancer drugs with coumarin structures. Chin. J. New Drugs 2013, 23, 2392–2404. [Google Scholar]
  3. Symeonidis, T.; Fylaktakidou, K.C.; Hadjipavlou-Litina, D.J.; Litinas, K.E. Synthesis and anti-inflammatory evaluation of novel angularly or linearly fused coumarins. Eur. J. Med. Chem. 2009, 44, 5012–5017. [Google Scholar] [CrossRef] [PubMed]
  4. Kongkathip, B.; Kongkathip, N.; Sunthitikawinsakul, A.; Napaswat, C.; Yoosook, C. Anti-HIV-1 constituents from Clausena excavata: Part II. Carbazoles and a pyranocoumarin. Phytother. Res. 2005, 19, 728–731. [Google Scholar] [CrossRef] [PubMed]
  5. Su, C.R.; Yeh, S.F.; Liu, C.M.; Damu, A.G.; Kuo, T.; Chiang, P.; Bastow, K.F.; Lee, K.; Wu, T. Anti-HBV and cytotoxic activities of pyranocoumarin derivatives. Bioorg. Med. Chem. 2009, 17, 6137–6143. [Google Scholar] [CrossRef]
  6. Wu, J.; Fong, W.; Zhang, J.; Leung, C.; Kwong, H.; Yang, M.; Li, D.; Cheung, H. Reversal of multidrug resistance in cancer cells by pyranocoumarins isolated from Radix Peucedani. Eur. J. Pharmacal. 2003, 473, 9–17. [Google Scholar] [CrossRef]
  7. Lima, V.; Silva, C.B.; Mafezoli, J.; Bezerra, M.M.; Moraes, M.O.; Mourão, G.S.; Silva, J.N.; Oliveira, M.C.F. Antinociceptive activity of the pyranocoumarin seselin in mice. Fitoterapla 2006, 77, 574–578. [Google Scholar] [CrossRef]
  8. Lee, H.J.; Lee, H.J.; Lee, E.O.; Lee, J.H.; Lee, K.S.; Kim, K.H.; Kim, S.H.; Lü, J. In vivo Anti-Cancer Activity of Korean Angelica Gigas and its Major Pyranocoumarin Decursin. Am. J. Chin. Med. 2009, 37, 127–142. [Google Scholar] [CrossRef] [Green Version]
  9. Zhang, J.; Li, L.; Jiang, C.; Xing, C.; Kim, S.; Lu, J. Anti-cancer and Other Bioactivities of Korean Angelica gigas Nakai (AGN) and Its Major Pyranocoumarin Compounds. Anti Cancer Agents Med. Chem. (Former. Curr. Med. Chem. Anti Cancer Agents) 2012, 12, 1239–1254. [Google Scholar] [CrossRef]
  10. Huang, L.; Feng, Z.L.; Wang, Y.T.; Lin, L.G. Anticancer carbazole alkaloids and coumarins from Clausena plants: A review. Chin. J. Nat. Med. 2017, 15, 881–888. [Google Scholar] [CrossRef]
  11. Peng, W.W.; Song, W.W.; Tan, N.H. Research Progress On Coumarins from Clausena and Their Pharmacological. Nat. Prod. Res. Dev. 2017, 8, 1428–1438. [Google Scholar]
  12. Arbab, I.A.; Looi, C.Y.; Abdul, A.B.; Cheah, F.K.; Wong, W.F.; Sukari, M.A.; Abdullah, R.; Mohan, S.; Syam, S.; Arya, A. Dentatin Induces Apoptosis in Prostate Cancer Cells via Bcl-2, Bcl-xL, Survivin Downregulation, Caspase-9, -3/7 Activation, and NF-κB Inhibition. Evid. Based Compl. Altern. Med. 2012, 2012, 856029. [Google Scholar] [CrossRef] [PubMed]
  13. Arbab, I.A.; Abdul, A.B.; Sukari, M.A.; Abdullah, R.; Syam, S.; Kamalidehghan, B.; Ibrahim, M.Y.; Taha, M.M.; Abdelwahab, S.I.; Ali, H.M. Dentatin isolated from Clausena excavata induces apoptosis in MCF-7 cells through the intrinsic pathway with involvement of NF-κ B signalling and G0/G1 cell cycle arrest: A bioassay-guided approach. J. Ethnopharmacol. 2013, 145, 343–354. [Google Scholar] [CrossRef] [PubMed]
  14. Andas, A.R.J.; Ahmad Bustamam, A.; Heshu Sulaiman, R.; Mohd Aspollah, S.; Siddig Ibrahim, A.; Nozlena Abdul, S.; Theebaa, A.; Ismail Adam, A. Dentatin from Clausena excavata Induces Apoptosis in HepG2 Cells via Mitochondrial Mediated Signaling. Asian Pac. J. Cancer Prev. 2015, 16, 4311. [Google Scholar] [CrossRef] [PubMed]
  15. Ahn, K.S.; Sim, W.S.; Kim, I.H. Decursin: A cytotoxic agent and protein kinase C activator from the root of Angelica gigas. Planta Med. 1996, 62, 7–9. [Google Scholar] [CrossRef]
  16. Ahn, K.S.; Sim, W.S.; Lee, I.K.; Seu, Y.B.; Kim, I.H. Decursinol angelate: A cytotoxic and protein kinase C activating agent from the root of Angelica gigas. Planta Med. 1997, 63, 360–361. [Google Scholar] [CrossRef]
  17. Wei, J.H.; Huo, L.N.; Huang, M.; Chen, R.; Feng, X. Robustic Acid and its Extraction Method and Application. CN2014105222249, 30 September 2014. [Google Scholar]
  18. Editorial Committee of the National Chinese Medicine Administrative Bureau. Chinese Materia Medica; Shanghai Scientific and Technical Education Publishing House: Shanghai, China, 2005; Volume 4. [Google Scholar]
  19. Pommier, Y. Diversity of DNA topoisomerases I and inhibitors. Biochimie 1998, 80, 255–270. [Google Scholar] [CrossRef]
  20. Ismail, M.M.F.; Rateb, H.S.; Hussein, M.M.M. Synthesis and docking studies of novel benzopyran-2-ones with anticancer activity. Eur. J. Med. Chem. 2010, 45, 3950–3959. [Google Scholar] [CrossRef]
  21. Zhao, L.X.; Moon, Y.S.; Basnet, A.; Kim, E.K.; Jahng, Y.; Park, J.G.; Jeong, T.C.; Cho, W.J.; Choi, S.U.; Chong, O.L. Synthesis, topoisomerase I inhibition and structure–activity relationship study of 2,4,6-trisubstituted pyridine derivatives. Bioorg. Med. Chem. Lett. 2004, 14, 1333–1337. [Google Scholar] [CrossRef]
  22. Hevener, K.E.; Zhao, W.; Ball, D.M.; Babaoglu, K.; Qi, J.; White, S.W.; Lee, R.E. Validation of Molecular Docking Programs for Virtual Screening against Dihydropteroate Synthase. J. Chem. Inf. Model. 2009, 2, 444–460. [Google Scholar] [CrossRef]
  23. Lill, M. Virtual screening in drug design. In Silico Models for Drug Discovery; Humana Press: Totowa, NJ, USA, 2013; Volume 993, pp. 1–12. [Google Scholar]
  24. Olivares, E.M.; Lwande, W.; Monache, F.D.; Bettolo, G.B.M. A pyrano-isoflavone from seeds of Milletia thonningii. Phytochemistry 1982, 21, 1763–1765. [Google Scholar] [CrossRef]
  25. Falshaw, C.P.; Harmer, R.A.; Ollis, W.D.; Wheeler, R.E.; Rao, N.V.S. Natural occurrence of 3-aryl-4-hydroxycoumarins. Part II. Phytochemical examination of Derris scandens (Roxb.) Benth. J. Chem. Soc. C Org. 1969, 3, 374. [Google Scholar] [CrossRef]
  26. Jin, Y.; Chen, Q.; Shi, X.; Lu, Z.; Cheng, C.; Lai, Y.; Zheng, Q.; Pan, J. Activity of triptolide against human mast cells harboring the kinase domain mutant KIT. Cancer Sci. 2009, 100, 1335–1343. [Google Scholar] [CrossRef] [PubMed]
  27. Xu, Y.; Jing, D.; Chen, R.; Haroon, U.R.; Jiang, J.; Liu, X.; Wang, L.; Wang, P. Design, synthesis and evaluation of novel sophoridinic imine derivatives containing conjugated planar structure as potent anticancer agents. Bioorg. Med. Chem. 2018, 26, 4136–4144. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Chemical structures of some linear-type pyranocoumarins.
Figure 1. Chemical structures of some linear-type pyranocoumarins.
Ijms 20 05336 g001
Figure 2. Structure of robustic acid (RA).
Figure 2. Structure of robustic acid (RA).
Ijms 20 05336 g002
Scheme 1. The synthesis of 4-acyloxy robustic acid derivatives (2a2j).
Scheme 1. The synthesis of 4-acyloxy robustic acid derivatives (2a2j).
Ijms 20 05336 sch001
Figure 3. Crystal structure of compound 2f.
Figure 3. Crystal structure of compound 2f.
Ijms 20 05336 g003
Figure 4. View of the packing compound 2f in the cell.
Figure 4. View of the packing compound 2f in the cell.
Ijms 20 05336 g004
Figure 5. DNA topoisomerase I (Topo I) inhibitory activity of (A) CPT, RA, and all target compounds (2a–2j) at 10 mM; (B) Compounds 2d, 2g, and 2i in different concentrations using CPT (0.1 mM) as a positive control.
Figure 5. DNA topoisomerase I (Topo I) inhibitory activity of (A) CPT, RA, and all target compounds (2a–2j) at 10 mM; (B) Compounds 2d, 2g, and 2i in different concentrations using CPT (0.1 mM) as a positive control.
Ijms 20 05336 g005
Figure 6. Result of validation for Topo I inhibitor (CPT) inside the enzyme active site (derived from PDB ID 1T8I).
Figure 6. Result of validation for Topo I inhibitor (CPT) inside the enzyme active site (derived from PDB ID 1T8I).
Ijms 20 05336 g006
Figure 7. The best pose of the binding mode of compounds with DNA Topo I complex (PDB:1T8I).
Figure 7. The best pose of the binding mode of compounds with DNA Topo I complex (PDB:1T8I).
Ijms 20 05336 g007
Figure 8. Morphological observation of 2i with AO/EB double staining under fluorescence microscope (630×) on HL-60 cells. (A) Cells not treated with 2i (control); (B) cells treated with compound 2i at 6.25 μM; (C) cells treated with compound 2i at 12.5 μM; (D) cells treated with compound 2i at 25 μM.
Figure 8. Morphological observation of 2i with AO/EB double staining under fluorescence microscope (630×) on HL-60 cells. (A) Cells not treated with 2i (control); (B) cells treated with compound 2i at 6.25 μM; (C) cells treated with compound 2i at 12.5 μM; (D) cells treated with compound 2i at 25 μM.
Ijms 20 05336 g008
Figure 9. Apoptosis ratio detection of compound 2i for 48 h by Annexin V/PI assay. (A) Not treated with compound 2i (control); (B) treated with compound 2i at 6.25 μM; (C) treated with compound 2i at 12.50 μM for and (D) treated with compound 2i at 25 μM (E) The percentage of quadrants Q2 and Q4 are shown as mean ± SD. Data shown in the figure represent three independent experiments (* p < 0.05, ** p < 0.01).
Figure 9. Apoptosis ratio detection of compound 2i for 48 h by Annexin V/PI assay. (A) Not treated with compound 2i (control); (B) treated with compound 2i at 6.25 μM; (C) treated with compound 2i at 12.50 μM for and (D) treated with compound 2i at 25 μM (E) The percentage of quadrants Q2 and Q4 are shown as mean ± SD. Data shown in the figure represent three independent experiments (* p < 0.05, ** p < 0.01).
Ijms 20 05336 g009
Figure 10. Cell-cycle analysis of HL-60 cells treated with compound 2i for 48 h, by flow cytometry. (A) Not treated with compound 2i (control); (B) treated with compound 2i at 6.25 μM; (C) treated with compound 2i at 12.50 μM; (D) treated with compound 2i at 25 μM; (E) The percentages of each population are shown as mean ± SD. Data shown in the figure are representative of three independent experiments (* p < 0.05, ** p < 0.01).
Figure 10. Cell-cycle analysis of HL-60 cells treated with compound 2i for 48 h, by flow cytometry. (A) Not treated with compound 2i (control); (B) treated with compound 2i at 6.25 μM; (C) treated with compound 2i at 12.50 μM; (D) treated with compound 2i at 25 μM; (E) The percentages of each population are shown as mean ± SD. Data shown in the figure are representative of three independent experiments (* p < 0.05, ** p < 0.01).
Ijms 20 05336 g010
Table 1. IC50 values of the synthesized compounds and controls obtained for anti-proliferative activity on different tumor cell lines (μM).
Table 1. IC50 values of the synthesized compounds and controls obtained for anti-proliferative activity on different tumor cell lines (μM).
CompoundsHL-60A-549SMMC-7721HepG2HelaLO2BEAS-2B
2a>10098.42 ± 4.1840.24 ± 1.38>100>100>100>100
2b>100>100>100>100>100>100>100
2c>100>100>100>100>100>100>100
2d21.04 ± 0.4353.33 ± 1.3736.79 ± 1.0640.12 ± 1.3637.03 ± 0.97>100>100
2e80.19 ± 3.7291.63 ± 2.2168.18 ± 3.9258.99 ± 3.11>100>100>100
2f>100>10066.18 ± 5.7087.27 ± 2.1465.07 ± 1.27>100>100
2g16.63 ± 0.1240.26 ± 0.6230.47 ± 1.5955.87± 0.4227.45 ± 0.3820.79 ± 0.97>100
2h>100>10092.01 ± 6.10>100>100>100>100
2i16.38 ± 0.2737.06 ± 0.6131.58 ± 1.2544.23 ± 0.8967.45 ± 1.24>100>100
2j87.44 ± 7.12>10078.15 ± 4.35>100>100>100>100
RA>100>100>100>100>100>100>100
CPT12.25 ± 1.0615.34 ± 1.2218.9 ± 4.121.3 ± 0.620.5 ± 0.080.030 ± 0.0022.159 ± 0.47
cisplatin14.23 ± 0.5625.40 ± 1.061.32 ± 0.9710.59 ± 1.405.83 ± 0.14<0.00857.66 ± 3.10
Table 2. Interactions between DNA Topo I (from PDB ID 1T8I) and target compounds.
Table 2. Interactions between DNA Topo I (from PDB ID 1T8I) and target compounds.
CompoundAmino AcidTypeHydrophobic ResiduesTotal Score a
2aDA113π–πLYS374, GLU356, DC112, TGP11, DT10, TYR426, ALA351, ASN352, MET428, LYS4258.4481
DC112π–π
DA113Hydrogen Bond
2bTGP11π–πASP533, THR718, TGP11, DT10, GLU356, ASN352, DC112, DG126.9191
DA113π–π
ARG364Hydrogen Bond
DA113Hydrogen Bond
2cTGP11π–πASP533, TGP11, GLU356, DC112, LYS532, HIS632, DT10, ASN722, ASN352, DA1138.9089
DA113π–π
DC112π–π
DT10π–π
THR718Hydrogen Bond
ARG364Hydrogen Bond
2dTGP11π–πASN722, DT10, TGP11, ALN351, ASN352, GLU356, DA113, ASP533, PTR723, ARG488, LYS532, HIS6328.7600
DA113π–π
DT10π–π
ARG364Hydrogen Bond
THR718Hydrogen Bond
2eTGP11π–πASN722, DT10, TGP11, ALN351, ASN352, GLU356, DA113, ASP533, PTR723, ARG488, LYS532, HIS6328.6166
DA113π–π
DC112π–π
ARG364Hydrogen Bond
DA113Hydrogen Bond
2fTGP11π–πDC112, TGP11, THR718, DT10, TRP416, GLU356, LYS354, ASN3528.5897
DA113π–π
DT10π–π
DA113Hydrogen Bond
ARP364Hydrogen Bond
2gDC112π–πILE427, TGP11, DT10, DA113, ARG364, DC112, PRO357, GLU356, LYS374, GLU418, TYR42610.0229
TGP11π–π
DA113π–π
MET428Hydrogen Bond
LYS425Hydrogen Bond
2hDT10π–πDC112, TGP11, THR718, DT10, LYS436, PRO431, ASN352, TRP416, LYS354, GLU3569.2983
TGP11π–π
DA113π–π
DA113Hydrogen Bond
ARG364Hydrogen Bond
2iDA113π–πDT10, TGP11, THR718, DC112, LYS354, GLU356, TRP416, ASN3528.8944
TGP11π–π
DT10π–π
DA113Hydrogen Bond
ARG364Hydrogen Bond
2jTGP11π–πDC112, TYR426, LYS425, ASN352, THR718, DT10, TGP118.4305
DA113π–π
ARG364Hydrogen Bond
DA113Hydrogen Bond
RATGP11π–πARG364, ASP533, TGP11, DA113, ALA351, ASN352, LEU429, MET4287.8183
DA113π–π
DT10Hydrogen Bond
CPTTGP11π–πDC112, DA113, TGP11, DT10, DG1210.3269
DA113π–π
DC112π–π
ASP533Hydrogen Bond
TGP11π–π
a Total score represents the binding affinity in units of -logKd.

Share and Cite

MDPI and ACS Style

Chen, R.; Huo, L.; Jaiswal, Y.; Wei, J.; Li, D.; Zhong, J.; Williams, L.; Xia, X.; Liang, Y. Synthesis and Evaluation of Anticancer Activity of New 4-Acyloxy Derivatives of Robustic Acid. Int. J. Mol. Sci. 2019, 20, 5336. https://doi.org/10.3390/ijms20215336

AMA Style

Chen R, Huo L, Jaiswal Y, Wei J, Li D, Zhong J, Williams L, Xia X, Liang Y. Synthesis and Evaluation of Anticancer Activity of New 4-Acyloxy Derivatives of Robustic Acid. International Journal of Molecular Sciences. 2019; 20(21):5336. https://doi.org/10.3390/ijms20215336

Chicago/Turabian Style

Chen, Rui, Lini Huo, Yogini Jaiswal, Jianhua Wei, Dianpeng Li, Jing Zhong, Leonard Williams, Xing Xia, and Yan Liang. 2019. "Synthesis and Evaluation of Anticancer Activity of New 4-Acyloxy Derivatives of Robustic Acid" International Journal of Molecular Sciences 20, no. 21: 5336. https://doi.org/10.3390/ijms20215336

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop