Open Access Article
Sara P. S. P. Moura
abc,
Marta Cascante
de,
Ismael Rufinof,
Rita C. Guedes
f,
Silvia Marin
*de and
Jorge A. R. Salvador
*abc
aLaboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal. E-mail: salvador@ci.uc.pt; Tel: +351-239-488-479
bCenter for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
cCentre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504 Coimbra, Portugal
dDepartment of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, 08028 Barcelona, Spain. E-mail: silviamarin@ub.edu; Tel: +34-934-9683
eCentro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
fResearch Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, 1649-003 Lisboa, Portugal
First published on 6th October 2025
Novel derivatives of carnosic acid 1 with ester or carbamate groups at C-20 and derivatives with these functional groups combined with benzylic modifications (C-7) were synthesized and evaluated in a colorectal cancer cell line (HCT116). Compound 8, which featured a butyl ester at C-20 and a carbonyl group at C-7, and compound 17, which featured a 2-methylpropyl carbamate at C-20, achieved the best results in HCT116 cells. Compounds 8 and 17 also demonstrated better ability to inhibit the growth of other cancer cell lines than CA 1. In general, the best results were achieved with compound 17, which exhibited higher potency against SW480 cells (IC50 = 6.3 μM). This compound also showed selectivity for cancer cells compared to normal cells. Compound 17 was subjected to additional studies to elucidate the mechanism responsible for its antiproliferative activity in SW480 cells. At 24 h, compound 17 arrested the cell cycle at the G0/G1 phase by decreasing the CDK4/CDK6 levels. It also reduced ROS levels by increasing the expression of SOD2/MnSOD. However, at 48 h, compound 17 induced cell cycle arrest in the S phase and increased ROS levels. At 72 h, compound 17 elevated the ROS levels without inducing cell cycle arrest. Additionally, molecular docking studies showed that compound 17 establishes several interactions with the amino acids of the CDK6 active site. In conclusion, compound 17 is a promising candidate for the development of novel anticancer drugs.
Abietane-type diterpenoids belong to a family of natural compounds isolated from diverse terrestrial plants.4,5 The most abundant group of abietanes found in nature is aromatic abietanes, featuring varying degrees of oxygenation at various positions, an aromatic ring C, and nonfunctionalized A-ring carbons. Carnosic acid (CA) 1, a type of aromatic abietane, is characterized by a tricyclic ring system, a diphenol C-ring, and a carboxylic acid at the C-20 position (Fig. 1). This abietane is found in rosemary leaves (Rosmarinus officinalis) and common sage (Salvia officinalis). In recent years, numerous studies have highlighted various biological activities of CA 1, such as anticancer, antioxidant, anti-inflammatory, antimicrobial, antiadipogenic, antiangiogenic, antidiabetic, cardioprotective, neuroprotective, and gastroprotective activities.4,6–13 Among the biological activities of CA 1, anticancer activity has been the most investigated both in vitro and in vivo and mechanistic studies have revealed that CA 1 can modulate the biological pathways, such as apoptosis, autophagy, cell cycle and oxidative metabolism, involved in cancer development and progression.4,6–9
Although CA 1 has shown promising results in cancer treatment, enhancing its potency, selectivity, and pharmacokinetic properties is essential.14–16 One strategy to achieve this goal, minimally explored in this field, involves the chemical modification of the backbone.5,17–20 Han et al. observed that the anticancer activity of CA 1 against the PANC-1 pancreatic cancer cell line could be enhanced by modifying the carboxylic (C-20) and benzylic (C-7) positions.17 This included introducing methyl ester, amides, or substituted 1,3,4-oxadiazole at the C-20 position, as well as introducing substituents using S-linkers or benzylic oxidation at the C-7 position. In another study, it was observed that incorporating an ester group at C-20 along with o-quinone or quinone methide in the C-ring improved the anticancer activity of CA 1 against P388 murine leukemia cells.18 In a previous study, Moura et al. reported that the introduction of a urea group at the C-20 position improved the anticancer activity of CA 1 in HCT116 colorectal cancer cells.20 While CA 1 exhibited an IC50 of 42 μM, most derivatives showed enhanced potency, with the most active derivatives achieving IC50 values between 9.8–14 μM. Carbamate-containing compounds are gaining increasing attention in the field of medicinal chemistry and drug discovery. Some therapeutic agents containing this functional group, such as mitomycin C, irinotecan, and capecitabine, have been used for cancer treatment.21–23 The carbamate group enhances chemical stability and permeability across the cell membrane, and modulates interactions with target enzymes or receptors, highlighting its significance in pharmaceutical research.
In this study, we synthesized novel CA 1 derivatives incorporating an ester or a carbamate group at C-20 position, as well as derivatives featuring these functional groups associated with chemical modifications at the benzylic position (C-7). The antiproliferative activity of the CA 1 derivatives was initially evaluated in a colorectal cancer (CRC) cell line (HCT116). Subsequently, the derivatives exhibiting the most promising activity were examined against other cancer cell lines (colorectal, melanoma, and pancreatic). The CA 1 derivative with the best results was chosen for further investigation to elucidate the potential mechanism responsible for the observed anticancer activity.
In order to control the reactivity of the hydroxyl groups at the C-11 and C-12 positions, CA 1 was treated with 4-dimethylaminopyridine (DMAP) and acetic anhydride to introduce acetate groups at these positions, resulting in derivative 2 (Scheme 1).20 This derivative with acetate groups at C-11 and C-12 allows for a more selective reaction at other positions, reducing the production of secondary products, and has already been synthesized by other research groups.17,18,24
The carboxylic acid at the C-20 of derivative 2 was esterified using methyl iodide, ethyl iodide, and N-butyl bromide, in the presence of anhydrous potassium carbonate, to give derivatives 3–5, respectively (Scheme 1). Compound 3 was previously synthesized by other groups of researchers using other synthetic routes.17,18,24,25 The successful incorporation of the ester group into derivatives 3–5 was confirmed by the presence of a peak at approximately 175 ppm in the 13C NMR spectra and a band stretch between 1713–1719 cm−1 in the IR spectra. A singlet at 3.51 ppm, a triplet at 1.16 and a triplet at 0.80 ppm were observed in the 1H NMR spectra of compounds 3–5, respectively, corresponding to methyl groups of the ester chains. In 13C NMR spectra of compounds 4 and 5, a peak at approximately 14 ppm was observed for the carbon of the methyl group of the ester chain. Signals of the methylene groups bonded to the oxygen atom of the ester chains in compounds 4 and 5 were observed in the range 3.82–4.06 ppm in the 1H NMR spectra, while the corresponding methylene carbons appeared at 60.93 and 64.96 ppm in the 13C NMR spectra.
The next step aimed to explore modifications at the benzylic position (C-7) of the ester derivatives through the incorporation of a carbonyl group (derivatives 6–8) and subsequent reduction to a hydroxyl group (derivatives 9–11), as depicted in Scheme 1. To the best of our knowledge, among the derivatives 6–11, only compound 6 was previously synthesized by another research group through an alternative synthetic route employing chromium trioxide in an acetic acid solution.17 Derivatives 3–5 underwent oxidation at C-7 by reacting with a mixture of iron sulfate and potassium permanganate to give derivatives 6–8, respectively (Scheme 1). Benzylic oxidation was confirmed by the presence of a peak at approximately 198 ppm in the 13C spectra and a band stretch at approximately 1690 cm−1 in the IR spectra of the derivatives 6–8. After, the derivatives 6–8 underwent reduction of the carbonyl group at C-7 through reaction with sodium borohydride in anhydrous methanol, resulting in the formation of derivatives 9–11, respectively (Scheme 1). The 1H NMR spectra of these derivatives revealed a signal around 4.74 ppm corresponding to the benzylic proton (7-H), while the 13C NMR spectra exhibited a peak around 71 ppm, attributed to the benzylic carbon linked to the hydroxyl group. Additionally, successful reduction was also confirmed by the presence of a broad O–H stretching vibration between 3503 and 3516 cm−1. This reduction generated a new stereogenic center at C-7. Analysis of the NOESY spectrum of compound 10 revealed a spatial correlation between the proton at C-5 (5-H, α-oriented) and the benzylic proton (7-H), allowing the stereochemical assignment of 7-H as α-oriented. Given the structural similarity and identical reaction conditions, this stereochemical configuration can be extended to compounds 9 and 11.
To synthesize the novel carbamate-containing derivatives CA 1, we initially focused on preparing an isocyanate-containing derivative at C-20 (derivative 14) from derivative 2. This derivative was previously synthesized by our research group.20 After, derivative 14 underwent a reaction with some alcohols in THF solution at 40/50 °C, resulting in the formation of carbamate-containing derivatives (15–17), as depicted in Scheme 2. The successful introduction of the carbamate moiety was verified by NMR and IR techniques. In the 1H NMR spectra, the proton signal of the –NH group appeared as a singlet in the 4.72–4.76 ppm range. In the IR spectra, a band was observed in the 1513–1515 cm−1 range, which resulted from a combination of an N–H bending band and a C–N stretching band, and another band was observed in the 3437–3447 cm−1 range relative to the N–H bond stretch. In the 13C NMR spectra, the signal of the carbamate carbonyl group appeared at approximately 154 ppm, and an IR band corresponding to the C
O bond stretch was observed in the characteristic region of 1729–1735 cm−1. In the 1H NMR spectra of derivatives 15–17, the signals referent to the methyl groups of the carbamate chains appeared as a singlet at 3.50 ppm for compound 15, a multiplet at 1.18–1.11 ppm for compound 16, and a doublet at 0.85 ppm for compound 17. The signals of the methylene groups bonded to the oxygen atom in compounds 16 and 17 were observed between 3.53 at 3.99 ppm. In the 13C NMR spectra, the signals of the carbons of the methylene groups appeared at 60.31 and 70.61 ppm.
Similar to the approach used for the ester derivatives, we synthesized carbamate derivatives containing a carbonyl group at the benzylic position (C-7) (derivatives 18 and 19), as illustrated in Scheme 2. To achieve this, derivatives 15 and 16 were treated under the aforementioned reaction conditions, resulting in the formation of derivatives 18 and 19, respectively. Successful benzylic oxidation was confirmed by the signal at approximately 197 ppm in the 13C spectra.
| Compound | Cell line/IC50b (μM) |
|---|---|
| HCT116 | |
| a After 72 h of incubation with increasing concentrations of each compound, IC50 values were calculated based on the results of cell viability obtained using the MTT assay. IC50 values are relative to at least three independent experiments and are expressed as the mean ± SD.b IC50 is the compound concentration necessary to inhibit half of the cell growth.c IC50 value was previously determined by our research team using the same method. | |
| CA 1 | 42 ± 4 |
| 2 | 44 ± 4 |
| 3 | 28 ± 2 |
| 4 | 25 ± 2 |
| 5 | 26 ± 1 |
| 6 | 36 ± 1 |
| 7 | 31 ± 3 |
| 8 | 15.1 ± 0.7 |
| 9 | 37 ± 3 |
| 10 | 34 ± 3 |
| 11 | 27 ± 4 |
| 14 | 28 ± 3 |
| 15 | 55 ± 5 |
| 16 | 37 ± 2 |
| 17 | 17 ± 2 |
| 18 | >50 |
| 19 | 48.2 ± 0.1 |
| Cisplatin | 21 ± 1 (ref. 26)c |
![]() | ||
| Fig. 2 Schematic representation of the main SAR conclusions obtained from the IC50 values of the CA 1 derivatives in HCT116 (A) and SW480 (B) cells. | ||
As presented in Table 1, compounds featuring an ester group at the C-20 position (compounds 3–5) demonstrated enhanced antiproliferative activity compared to CA 1. In fact, there was a decrease in the IC50 values from 42 μM (CA 1) to 28 (3), 25 (4), and 26 μM (5). The length of the ester chain did not seem to influence the potency of the compounds, revealing very similar potency.
In general, modifications at the benzylic position (C-7) in the ester derivatives, namely, the introduction of a carbonyl group (compounds 6–8) or hydroxyl group (compounds 9–11), led to a reduction in antiproliferative activity compared to ester derivatives without modifications at this position (compounds 3–5), as shown in Table 1. However, compound 8 was the only exception, exhibiting an increase in potency, verifying a reduction in the IC50 value from 26 μM (derivative 5) to 15.1 μM (compound 8). Consequently, it can be inferred that the insertion of a carbonyl group at the benzylic position (C-7) was advantageous for the antiproliferative activity of the butyl ester derivative.
In compounds featuring a carbamate moiety at the C-20 position (compounds 15–17), carbamate chain length influenced their antiproliferative activity (Table 1). Carbamate 15, which possesses a methyl chain, exhibited a decrease in activity with an IC50 value (55 μM) higher than that of CA 1 (42 μM). With an increase in chain length, a simultaneous increase in the potency of the compounds was observed. Compound 16, characterized by an ethyl chain, exhibited an IC50 of 37 μM, whereas compound 17, featuring a longer chain (2-methylpropyl), demonstrated even greater potency with an IC50 of 17 μM. Benzylic oxidation of compounds 15 and 16, resulting in the formation of compounds 18 and 19, did not contribute to improved antiproliferative activity (Table 1).
Compound 8, featuring a butyl ester at C-20 and a carbonyl group at benzylic position (C-7), and compound 17, featuring a 2-methylpropyl carbamate, exhibited the most potent antiproliferative activity in HCT116 cells, with IC50 values of 15.1 μM and 17 μM, respectively. These compounds achieved better results than cisplatin, being 1.4- and 1.2-fold more active, respectively. Therefore, compounds 8 and 17 were selected for further evaluation in other cancer cell lines. Different cell lines of the same cancer type carry different genetic mutations, leading to variations in their susceptibility to the same treatment.27,28 Given that the HCT116 cell line is wild-type for p53 and microsatellite unstable, our aim was to investigate the effects of compounds 8 and 17 on CRC cell lines with mutations in the p53 gene and microsatellite stable, such as SW480, SW620, and Caco-2 cells.27 Furthermore, we selected SW480 and SW620 cells because they were derived from the same tumor, but from different locations. We investigated whether the compounds tended to exhibit greater effectiveness in a cell line derived from the primary tumor (SW480) or on a cell line derived from metastasis (SW620). As displayed in Table 2, compounds 8 and 17 demonstrated higher potency against SW480, SW620, and Caco-2 cells than CA 1. Moreover, they achieved better IC50 values in these cell lines than in HCT116 cells, showing a preference for p53-mutated cell lines, particularly SW480 cells. In this line, compound 8 and 17 achieved IC50 values of 6.8 μM and 6.3 μM, respectively, being 3.2-fold and 3.5-fold more potent than CA 1 (Fig. 2B). These two compounds also showed better antiproliferative activity against SW480 cells than cisplatin, being 2.2-fold (compound 8) and 2.4-fold (compound 17) more active. Comparing the results obtained in SW480 and SW620 cells, it is evident that the compounds exhibited greater effectiveness against the primary tumor cells. Further investigations were conducted using compounds 8 and 17 to assess their capacity to inhibit cell growth in other cancer types, specifically pancreatic cancer, and melanoma. As shown in Table 2, it can be inferred that these compounds are also effective against other cancer types.
| Compound | Cell line/IC50b (μM) | |||||
|---|---|---|---|---|---|---|
| Colorectal | Pancreas | Melanoma | Non-tumoral | |||
| SW480 | SW620 | Caco-2 | Mia Paca-2 | A375 | BJ | |
| a After 72 h of incubation with increasing concentrations of each compound, IC50 values were calculated based on the results of cell viability obtained using the MTT assay. IC50 values are relative to at least three independent experiments and are expressed as the mean ± SD. N.D. – not determined.b IC50 is the compound concentration necessary to inhibit half of the cell growth.c IC50 value was acquired from the literature by applying the same method.d IC50 value was previously determined by our research team using the same method. | ||||||
| CA 1 | 21.8 ± 0.7 | 18 ± 2 | 34 ± 2 | 21 ± 1 | 27.6 ± 0.5 | N.D. |
| 8 | 6.8 ± 0.6 | 11 ± 1 | 12.7 ± 0.3 | 12 ± 1 | 8.1 ± 0.5 | N.D. |
| 17 | 6.3 ± 0.5 | 10 ± 1 | 16 ± 2 | 7.5 ± 0.6 | 6.7 ± 0.5 | >50 |
| Cisplatin | 15.2 ± 0.4 (ref. 34)c | 1.4 ± 0.5 (ref. 35)d | 12.5 ± 0.9 (ref. 36)c | 5 ± 1 (ref. 37)c | 3 ± 1 (ref. 38)c | 10 ± 2 (ref. 39)d |
In general, compound 17 achieved the most promising results against almost all cancer cell lines tested, and achieved the best results for SW480 cells, with an IC50 of 6.3 μM (Table 2 and Fig. 3). Therefore, we evaluated this compound in a non-tumoral cell line (BJ fibroblasts) to determine its selectivity for cancer cell lines. As indicated in Table 2, the antiproliferative activity of compound 17 notably decreased in BJ cells, being at least 3–8 times less potent than that in cancer cell lines. These findings highlight the selectivity of compound 17 in cancer cells, showing even greater selectivity than cisplatin. Hence, considering the acquired results, compound 17 was selected for additional biological studies in SW480 cells to gain insight into the mechanisms underlying its anticancer activity.
Our objective was to assess whether the anticancer activity observed in SW480 cells treated with compound 17 results from the activation of apoptosis. Therefore, SW480 cells were treated with 6.3 μM (IC50 value) of compound 17, incubated for 24, 48 or 72 h, and subsequently stained with annexin V and PI for post-analysis using flow cytometry. As shown in Fig. 5, compound 17 did not induce apoptosis in the SW480 cells, suggesting that this mechanism is not responsible for the anticancer activity of compound 17.
Since compound 17 did not induce apoptosis but increased the duplication time of treated cells, we aimed to evaluate whether its anticancer activity was due to cell cycle arrest. To validate this hypothesis, compound 17 was administered at 6.3 μM to SW480 cells and incubated for 24, 48, and 72 h. Following each incubation period, the cells were fixed and permeabilized with 70% cold ethanol and stained with PI before flow cytometry analysis. At 24 h, compound 17 caused cell cycle arrest at the G0/G1 phase, as evidenced by a significant increase in the percentage of SW480 cells, from 54% in the control group to 66% in the treated group (Fig. 6). However, at 48 h, compound 17 induced cell cycle arrest in the S phase, as demonstrated by a significant increase in the percentage of cells from 30% in the control group to 40% in the treated group (Fig. 6). At 72 h, compound 17 had no impact on the cell cycle of SW480 cells, as observed in Fig. 6.
In conclusion, the effect of compound 17 on the cell cycle of SW480 cells varied over time, arresting the cell cycle in the G0/G1 phase at 24 h and in the S phase at 48 h. These results suggest that the cytostatic effect of compound 17 was responsible for its anticancer activity.
In the above studies, compound 17 induced cell cycle arrest in the G0/G1 and S phases in SW480 cells at 24 and 48 h, respectively. To better understand the mechanism underlying this cell cycle arrest, we analyzed the expression of key proteins associated with cell cycle regulation using western blot (Fig. 7). As shown in Fig. 7, at 24 h, compound 17 significantly reduced the levels of CDK4/6 proteins, explaining the observed cell cycle arrest at the G0/G1 phase.46–48 Additionally, a decrease in cyclin A expression was noted at 24 h, possibly due to a reduction in the number of cells in the S phase (Fig. 6). At 48 h, the CDK4/6 proteins levels in SW480 cells treated with compound 17 persisted lower than those in the control group (Fig. 7). This reduction may be attributed to a decrease in the number of cells in the G0/G1 phase (Fig. 6). Surprisingly, at 48 h, compound 17 did not affect the expression level of cyclin A, as shown in Fig. 7. Despite the cell cycle analysis indicating arrest in the S phase at this time point (Fig. 6), cyclin A, which is responsible for S phase progression, did not exhibit a decrease. The arrest of the cell cycle at the S phase may be due to a reduction in the expression of cyclin E, as reported in other studies.49–51 Moreover, a notable decrease in the Rb protein levels was observed in SW480 cells treated with compound 17 at three different time points (24, 48, and 72 h), as observed in Fig. 7. These findings are consistent with those of other studies that have reported a decrease in Rb protein levels linked to cell cycle arrest.52,53 As previous studies have reported no cell cycle changes at 72 h, we did not expect variations in the expression of cell cycle regulatory proteins. However, a reduction in CDK4 and Rb levels was detected after 72 h of treatment with compound 17. It is possible that these alterations may not have reached a magnitude significant enough to induce cell cycle arrest.
In conclusion, compound 17 induced changes in the levels of cell cycle regulatory proteins, explaining the observed cell cycle arrest in the SW480 cells. The G0/G1 phase arrest observed at 24 h was attributed to a decrease in the CDK4/6 protein levels. Additional studies are essential to elucidate the mechanism by which compound 17 arrests the cell cycle during the S phase. A decrease in Rb protein levels also contributed to cell cycle arrest.
Given that compound 17 induced G1 phase arrest at 24 h and reduced CDK6 levels, we hypothesized that its effect on the cell cycle could also involve direct interaction with CDK6, in addition to the downregulation of its levels. Moreover, in our previous studies, a CA 1 derivative was shown to be a potential CDK6 inhibitor.20 Therefore, multiple molecular docking simulations were conducted to elucidate the binding mode of compound 17 and assess its interactions with CDK6. The CDK6 (PDB ID 6OQL) and compound 17 structures were prepared and validated using the methodology described by Moura et al.20 The selection of the optimal protocol, scoring function, and software is detailed elsewhere. The molecular docking results demonstrated that compound 17 adopts an optimal pose at the CDK6 binding site (Fig. 8A), interacting with amino acids ILE19, TYR24, VAL27, VAL77, HIS100, PHE98, VAL101, ASP104, ALA162, GLN149, and LEU152.
![]() | ||
| Fig. 8 Docking studies of compound 17 into the active site of CDK6 (PDB ID 6OQL), as predicted by the GNINA 1.0 docking software. Panel (A) illustrates the optimal docking pose of compound 17, highlighted in cyan, within the CDK6 binding site, with the CDK6 surface highlighted in green. Panel (B) compares the interactions observed in the 6OQL structure with those established by compound 17, noting that five out of seven interactions involve identical amino acid residues. Additionally, an extra hydrogen bond interaction is formed by ILE19. Panel (C) provides a detailed view of the ligand–protein interactions between compound 17 and the amino acid residues within the CDK6 binding pocket. For clarity, only the five residues that simultaneously interact with both compound 17 and the native ligand in the 6OQL structure are shown. *Compound 17 interacts with the amino acid residue ALA162, which is very close to ASP163. | ||
Further analysis of the docking simulations revealed that compound 17 adopts a pose highly similar to that of the ligand present in the X-ray crystal structure of CDK6 used for the docking studies (PDB ID 6OQL). The superposition of compound 17 and the crystallographic ligand is provided in the SI (Fig. S8). Compound 17 forms several interactions that are also present in the CDK6–ligand complex, specifically hydrophobic interactions with amino acid residues ILE19, VAL27, and LEU152, and hydrogen bonds with HIS100 and VAL101 (Fig. 8B and C). The distances between the atoms involved in these interactions are listed in the SI (Table S1). Additionally, compound 17 established a hydrogen bond with ILE19 and formed four extra hydrophobic interactions with the amino acids VAL77, PHE98, ASP104, and GLN149. These interactions contribute to stabilizing its position within the binding pocket, supporting the hypothesis that compound 17 could serve as a potential inhibitor of this enzyme.
Intracellular ROS levels can be quantified using lipophilic and non-fluorescent probes, such as 2′,7′-dichlorodihydrofluorescein diacetate (H2DCFDA).60 To assess the effect of compound 17 on intracellular ROS levels, SW480 cells were treated with compound 17, followed by incubation with the H2DCFDA probe after 24, 48, and 72 h of treatment. After, the cells were subjected to flow cytometry to quantify the fluorescence intensity of 2′,7′-dichlorofluorescein (DCF), as shown in Fig. 9. At 24 h, administration of compound 17 resulted in a reduction in intracellular ROS levels in SW480 cells relative to untreated cells (70 vs. 100%), as depicted in Fig. 9. However, at 48 and 72 h, compound 17 exhibited contrasting effects, causing an increase in intracellular ROS levels in SW480 cells to 157% and 290%, respectively. These results suggest that the effect of compound 17 on intracellular ROS levels is dependent on the time of exposure of the cells to the treatment, exhibiting an antioxidant role at 24 h and a pro-oxidant role at 48 and 72 h. Both effects of compound 17 contributed to the inhibition of SW480 cell growth.
The cellular redox environment regulates the cell cycle, and ROS levels determine whether the cell cycle is negatively or positively regulated.61,62 In our study, there seems to be a potential correlation between ROS levels and the cell cycle phase in which arrest occurs. At 24 h, a reduction in ROS levels could have led to cell cycle arrest at the G0/G1 phase in the SW480 cells. However, at 48 h, compound 17 arrested the cell cycle in another phase (S phase), which may be related to an increase in ROS levels. At 72 h, the concentration of ROS was further elevated; therefore, it was expected to arrest the cell cycle. Nevertheless, as observed in previous studies, no significant effect on the cell cycle was observed at 72 h. Further studies are required to investigate whether ROS levels influence the cell cycle of SW480 cells treated with compound 17.
Our studies revealed that compound 17 affects ROS levels; therefore, we assessed the effect of compound 17 on the expression of SOD2 (MnSOD) in SW480 cells after 24, 48, and 72 h of incubation. As shown in Fig. 10, compound 17 significantly increased the expression of SOD2/MnSOD at 24 and 72 h. At 24 h, overexpression of SOD2/MnSOD led to a decrease in ROS levels. However, after 72 h, the increased expression of SOD2 (MnSOD) was insufficient to reduce ROS levels.
:
1 of silica gel 60 F254 and silica gel 60 purchased from Merck Co. (Rahway, NJ, USA). A Büchi® Mp B-540 apparatus (Büchi®, Flawil, Switzerland) was used to determine the melting points (mp), which were reported without adjustment. A PerkinElmer Spectrum 400 FT-IR/FT-NIR spectrometer (PerkinElmer, Waltham, MA, USA) was used to record the infrared (IR) spectra. Chemical structures were elucidated using 1D nuclear magnetic resonance (NMR) (1H, 13C, and DEPT-135) and 2D NMR (COSY, NOESY, HSQC, and HMBC). The samples were dissolved in CDCl3, and 1H and 13C spectra were recorded at 400 and 100 MHz, respectively, using a Bruker Avance III 400 MHz spectrometer (Bruker, Billerica, MA, USA). The coupling constants (J) are denoted in hertz (Hz), and the chemical shifts (δ) are represented in parts per million (ppm). The 1H and 13C spectra were calibrated at δ 7.26 ppm and δ 77.16 ppm, respectively. Mass spectrometry (MS) was conducted using a Thermo Scientific Finnigan LXQ Linear Ion Trap Mass Spectrometer (Thermo Fisher Scientific, Waltham, MA, USA) with electrospray ionization (ESI). The ESI conditions were set to 5 kV in the positive mode, with a sheath gas flow of 5 U and a capillary temperature of 250 °C. Elemental analysis was conducted using a TruSpec 630-200-200 CHNS analyzer (Leco Corporation, St. Joseph, MI, USA).
3), 2.24 (3H, s, OCOC
3), 1.21 (3H, d, J = 6.9 Hz, CHC
3), 1.14 (3H, d, J = 6.9 Hz, CHC
3), 0.96 (3H, s, C
3), 0.86 (3H, s, C
3); 13C NMR (100 MHz, CDCl3) δ 179.61 (C20), 168.80 (O
OCH3), 168.33 (O
OCH3), 141.49, 140.07, 138.82, 136.93, 132.09, 125.36 (C14), 53.91, 47.70, 41.21, 34.66, 34.17, 32.62, 32.05, 27.52, 23.12, 22.87, 20.68, 20.56, 20.20, 19.96, 18.26. ESI-MS m/z: 439.3 [M + Na]+. Anal. calcd for C24H32O6·0.3C6H14: C, 70.05; H, 8.25. Found: C, 70.5; H, 7.9%.
:
1) to obtain the 3 as a white powder (190.0 mg, 92%). Mp: 158.2–160.0 °C. IR (neat)vmax: 3021, 1773, 1763, 1719, 1221, 1210, 1193, 1181 cm−1. 1H NMR (400 MHz, CDCl3) δ 6.95 (1H, s, 14-H), 3.51 (3H, s, COOC
3), 2.26 (3H, s, OCOC
3), 2.25 (3H, s, OCOC
3), 1.22 (3H, d, J = 6.9 Hz, CHC
3), 1.14 (3H, d, J = 6.9 Hz, CHC
3), 0.96 (3H, s, C
3), 0.74 (3H, s, C
3); 13C NMR (100 MHz, CDCl3) δ 175.31 (C20), 168.76 (O
OCH3), 168.43 (O
OCH3), 141.44, 139.82, 138.83, 136.90, 131.88, 125.24 (C14), 53.76, 51.90, 47.86, 41.27, 34.79, 34.08, 32.60, 32.01, 27.51, 23.16, 22.77, 20.84, 20.54, 20.04, 19.90, 18.46. ESI-MS m/z: 453.3 [M + Na]+, 882.7 [2M + Na]+. Anal. calcd for C25H34O6: C, 69.7; H, 8.0. Found: C, 69.9; H, 8.4%.
:
1) to obtain the 4 as a white powder (122.0 mg, 57%). Mp: 125.2–127.1 °C. IR (neat)vmax: 3032, 1772, 1713, 1205, 1195, 1184, 1173 cm−1. 1H NMR (400 MHz, CDCl3) δ 6.94 (1H, s, 14-H), 4.06 (1H, dq, J = 10.9, 7.2 Hz, COOC
2CH3), 3.88 (1H, dq, J = 10.9, 7.1 Hz, COOC
2CH3), 2.25 (3H, s, OCOC
3), 2.24 (3H, s, OCOC
3), 1.22 (3H, d, J = 6.9 Hz, CHC
3), 1.16 (3H, t, J = 7.1 Hz, COOCH2C
3), 1.14 (3H, d, J = 6.9 Hz, CHC
3), 0.97 (3H, s, C
3), 0.78 (3H, s, C
3). 13C NMR (100 MHz, CDCl3) δ 174.72 (C20), 168.72 (O
OCH3), 168.25 (O
OCH3), 141.39, 139.69, 138.74, 136.92, 132.10, 125.09 (C14), 60.93 (O
H2), 53.70, 47.89, 41.33, 34.69, 34.19, 32.60, 31.95, 27.46, 23.19, 22.76, 20.88, 20.52, 20.15, 20.06, 18.43, 13.98 (OCH2
H3). ESI-MS m/z: 467.4 [M + Na]+, 910.6 [2M + Na]+. Anal. calcd for C26H36O6·0.1C6H14·0.4H2O: C, 69.4; H, 8.4. Found: C, 69.0; H, 8.8%.
:
1) to afford the 5 as a white powder (191.3 mg, 84%). Mp: 86.8–88.1 °C. IR (neat)vmax: 3022, 1780, 1766, 1714, 1204, 1183 cm−1. 1H NMR (400 MHz, CDCl3) δ 6.93 (1H, s, 14-H), 3.94 (1H, dt, J = 10.9, 6.5 Hz, COOC
2CH2), 3.82 (1H, dt, J = 10.9, 6.8 Hz, COOC
2CH2), 2.25 (3H, s, OCOC
3), 2.24 (3H, s, OCOC
3), 1.20 (3H, d, J = 6.9 Hz, CHC
3), 1.14 (3H, d, J = 6.9 Hz, CHC
3), 0.97 (3H, s, C
3), 0.80 (3H, t, J = 7.4 Hz, COOCH2CH2CH2C
3), 0.77 (3H, s, C
3). 13C NMR (100 MHz, CDCl3) δ 174.88 (C20), 168.76 (O
OCH3), 168.22 (O
OCH3), 141.41, 139.76, 138.74, 136.94, 132.06, 125.02 (C14), 64.96 (O
H2), 53.57, 48.01, 41.33, 34.62, 34.19, 32.60, 31.92, 30.34, 27.46, 23.12, 22.92, 20.91, 20.54, 20.11, 20.06, 19.41, 18.54, 13.69 (OCH2CH2CH2C
3). ESI-MS m/z: 495.4 [M + Na]+, 966.6 [2M + Na]+. Anal. calcd for C28H40O6·0.2C6H14·0.1H2O: C, 71.3; H, 8.8. Found: C, 71.0; H, 9.2%.
:
1) to afford the 6 as a white powder (83.8 mg, 82%). Mp: 147.2–149.1 °C. IR (neat)vmax: 3026, 1778, 1727,1688, 1198, 1182, 1171, 1150, 1144 cm−1. 1H NMR (400 MHz, CDCl3) δ 8.06 (1H, s, 14-H), 3.55 (3H, s, COOC
3), 2.30 (3H, s, OCOC
3), 2.29 (3H, s, OCOC
3), 1.26 (3H, d, J = 6.9 Hz, CHC
3), 1.17 (3H, d, J = 6.9 Hz, CHC
3), 0.96 (3H, s, C
3), 0.80 (3H, s, C
3); 13C NMR (100 MHz, CDCl3) δ 197.65 (C7), 173.46 (C20), 168.14 (O
OCH3), 167.96 (O
OCH3), 145.58, 141.72, 141.14, 137.14, 131.42, 124.56 (C14), 52.32, 50.97, 48.70, 41.07, 35.43, 34.02, 33.96, 31.97, 27.83, 22.96, 22.63, 20.75, 20.52, 19.90, 19.44. ESI-MS m/z: 445.1 [M + H]+. Anal. calcd for C25H32O7: C, 67.55; H, 7.3. Found: C, 67.4; H, 7.75%.
:
1) to obtain the 7 as a white powder (72.6 mg, 72%). Mp: 122.4–125.2 °C. IR (neat)vmax: 3039, 1771, 1718, 1691, 1192, 1171, 1159, 1140 cm−1. 1H NMR (400 MHz, CDCl3) δ 8.05 (1H, s, 14-H), 4.09 (1H, dq, J = 10.9, 7.2 Hz, COOC
2CH3), 3.93 (1H, dq, J = 10.9, 7.2 Hz, COOC
2CH3), 2.30 (3H, s, OCOC
3), 2.29 (3H, s, OCOC
3), 1.27 (3H, d, J = 6.9 Hz, CHC
3), 1.17 (3H, d, J = 6.9 Hz, CHC
3), 1.15 (3H, t, J = 7.2 Hz, COOCH2C
3), 0.96 (3H, s, C
3), 0.84 (3H, s, C
3). 13C NMR (100 MHz, CDCl3) δ 197.80 (C7), 172.85 (C20), 168.01 (O
OCH3), 167.96 (O
OCH3), 145.56, 141.62, 141.10, 137.33, 131.48, 124.50 (C14), 61.61 (O
H2), 50.95, 48.76, 41.14, 35.48, 34.09, 33.94, 31.96, 27.82, 22.99, 22.61, 20.81, 20.52, 19.94, 19.69, 13.95 (OCH2
H3). ESI-MS m/z: 459.1 [M + H]+. Anal. calcd for C26H34O7·0.5H2O: C, 66.8; H, 7.55. Found: C, 67.1; H, 8.1%.
:
1) to obtain the 8 as a white powder (77.7 mg, 76%). Mp: 88.2–90.1 °C. IR (neat)vmax: 3012, 1781, 1761, 1714, 1691, 1193, 1173, 1155, 1138 cm−1. 1H NMR (400 MHz, CDCl3) δ 8.05 (1H, s, 14-H), 4.00–3.93 (1H, m, COOC
2CH2), 3.89 (1H, dt, J = 10.8, 6.8 Hz, COOC
2CH2), 2.29 (3H, s, OCOC
3), 2.30 (3H, s, OCOC
3), 1.25 (3H, d, J = 6.9 Hz, CHC
3), 1.18 (3H, d, J = 6.9 Hz, CHC
3), 0.96 (3H, s, C
3), 0.83 (3H, s, C
3), 0.79 (3H, t, J = 7.4 Hz, COOCH2CH2CH2C
3).13C NMR (100 MHz, CDCl3) δ 197.77 (C7), 172.98 (C20), 167.98 (O
OCH3), 167.94 (O
OCH3), 145.54, 141.66, 141.11, 137.33, 131.51, 124.46 (C14), 65.58 (O
H2), 50.88, 48.84, 41.12, 35.53, 34.08, 33.89, 31.94, 30.25, 27.80, 22.92, 22.69, 20.80, 20.51, 19.93, 19.61, 19.34, 13.60 (OCH2CH2CH2C
3). ESI-MS m/z: 487.2 [M + H]+. Anal. calcd for C28H38O7: C, 69.1; H, 7.9. Found: C, 69.0; H, 8.3%.
:
1) to afford the 9 as a light-yellow powder (60.5 mg, 76%). Mp: 89.1–91.8 °C. IR (neat)vmax: 3503, 3025, 1771, 1718, 1202, 1176 cm−1. 1H NMR (400 MHz, CDCl3) δ 7.58 (1H, s, 14-H), 4.78–4.69 (1H, m, 7-H), 3.54 (3H, s, COOC
3), 2.26 (3H, s, OCOC
3), 2.24 (3H, s, OCOC
3), 1.24 (3H, d, J = 6.9 Hz, CHC
3), 1.17 (3H, d, J = 6.9 Hz, CHC
3), 0.97 (3H, s, C
3), 0.75 (3H, s, C
3); 13C NMR (100 MHz, CDCl3) δ 174.86 (C20), 168.56 (O
OCH3), 168.34 (O
OCH3), 140.84, 140.66, 140.12, 140.00, 131.99, 123.58 (C14), 71.03 (C7), 52.11, 51.18, 48.54, 41.00, 34.63, 33.77, 32.45, 29.25, 27.79, 23.04, 22.92, 20.77, 20.53, 19.88 (2C). ESI-MS m/z: 469.3 [M + Na]+. Anal. calcd for C25H34O7·1H2O: C, 64.6; H, 7.8. Found: C, 64.3; H, 8.1%.
:
1) to obtain the 10 as a light-yellow powder (67.1 mg, 81%). Mp: 83.0–85.6 °C. IR (neat)vmax: 3510, 3025, 1772, 1715, 1203, 1184, 1174 cm−1. 1H NMR (400 MHz, CDCl3) δ 7.56 (1H, s, 14-H), 4.79–4.69 (1H, m, 7-H), 4.09 (1H, dq, J = 10.9, 7.2 Hz, COOC
2CH3), 3.91 (1H, dq, J = 10.9, 7.1 Hz, COOC
2CH3), 2.26 (3H, s, OCOC
3), 2.24 (3H, s, OCOC
3), 1.60 (1H, dd, J = 13.0, 1.9 Hz, 5-H), 1.24 (3H, d, J = 6.9 Hz, CHC
3), 1.19 (3H, t, J = 7.2 Hz, COOCH2C
3), 1.17 (3H, d, J = 6.9 Hz, CHC
3), 0.98 (3H, s, C
3), 0.79 (3H, s, C
3). 13C NMR (100 MHz, CDCl3) δ 174.28 (C20), 168.54 (O
OCH3), 168.20 (O
OCH3), 140.76, 140.64, 140.10, 139.98, 132.31, 123.60 (C14), 71.01 (C7), 61.26 (O
H2), 51.23 (C5), 48.57, 41.08, 34.56, 33.89, 32.46, 29.27, 27.78, 23.09, 22.90, 20.84, 20.53, 20.14, 19.94, 14.04 (OCH2
H3). ESI-MS m/z: 483.3 [M + Na]+. Anal. calcd for C26H36O7·0.2C6H14·0.5H2O: C, 67.1; H, 8.2. Found: C, 66.8; H, 8.75%.
:
1) to obtain the 11 as a dark yellow powder (61.0 mg, 78%). Mp: 63.1–65.8 °C. IR (neat)vmax: 3516, 3026, 1773, 1715, 1202, 1183, 1172 cm−1. 1H NMR (400 MHz, CDCl3) δ 7.56 (1H, s, 14-H), 4.74 (1H, dd, J = 10.2, 7.0 Hz, 7-H), 3.98 (1H, dt, J = 10.9, 6.6 Hz, COOC
2CH2), 3.85 (1H, dt, J = 10.8, 6.9 Hz, COOC
2CH2), 2.26 (3H, s, OCOC
3), 2.24 (3H, s, OCOC
3), 1.23 (3H, d, J = 6.8 Hz, CHC
3), 1.17 (3H, d, J = 6.9 Hz, CHC
3), 0.98 (3H, s, C
3), 0.83 (3H, t, J = 7.4 Hz, COOCH2CH2CH2C
3), 0.78 (3H, s, C
3). 13C NMR (100 MHz, CDCl3) δ 174.40 (C20), 168.54 (O
OCH3), 168.16 (O
OCH3), 140.78, 140.63, 140.06, 139.97, 132.26, 123.47 (C14), 70.97 (C7), 65.28 (O
H2), 51.13, 48.64, 41.05, 34.49, 33.87, 32.44, 30.35, 29.32, 27.76, 23.01, 22.98, 20.84, 20.52, 20.06, 19.92, 19.46, 13.74 (OCH2CH2CH2C
3). ESI-MS m/z: 511.3 [M + Na]+. Anal. calcd for C28H40O7: C, 68.8; H, 8.25. Found: C, 68.8; H, 8.7%.
3), 2.29 (3H, s, OCOC
3), 1.22 (3H, d, J = 6.9 Hz, CHC
3), 1.16 (3H, d, J = 6.9 Hz, CHC
3), 1.03 (3H, s, C
3), 0.98 (3H, s, C
3); 13C NMR (100 MHz, CDCl3) δ 168.67 (O
OCH3), 168.10 (O
OCH3), 141.54, 141.20, 139.19, 135.78, 132.22, 125.17 (C14), 123.40 (N
O), 61.07, 53.29, 40.66, 37.32, 33.89, 32.85, 32.31, 27.61, 22.99, 22.93, 21.16, 21.02, 20.56, 19.43, 19.28. ESI-MS m/z: 436.3 [M + Na]+. Anal. calcd for C24H31NO5·0.2C6H14: C, 70.3; H, 7.9; N, 3.25. Found: C, 70.5; H, 7.6; N, 3.2%.
:
1) to obtain the 15 as a light-yellow powder (60.2 mg, 50%). Mp: 164.7–166.4 °C. IR (neat)vmax: 3437, 3060, 1770, 1735, 1515, 1228, 1197, 1176, 1146, 1108 cm−1. 1H NMR (400 MHz, CDCl3) δ 6.91 (1H, s, 14-H), 4.76 (1H, br s, N
), 3.50 (3H, s, OC
3), 2.28 (3H, s, OCOC
3), 2.27 (3H, s, OCOC
3), 1.20 (3H, d, J = 6.9 Hz, CHC
3), 1.14 (3H, d, J = 6.9 Hz, CHC
3), 0.97 (3H, s, C
3), 0.96 (3H, s, C
3); 13C NMR (100 MHz, CDCl3) δ 168.80 (O
OCH3), 168.73 (O
OCH3), 154.48 (NH
O), 141.37, 140.17, 138.88, 136.20, 133.35, 124.73 (C14), 56.34, 53.21, 51.76, 40.79, 34.40, 33.92, 32.96, 31.59, 27.55, 23.13, 22.81, 21.48, 21.40, 20.57, 19.40, 18.12. ESI-MS m/z: 468.3 [M + Na]+, 912.5 [2M + Na]+. Anal. calcd for C25H35NO6·0.1C6H15N·0.3H2O: C, 66.7; H, 8.1; N, 3.3. Found: C, 66.4; H, 8.5; N, 3.1%.
:
1) to afford 16 as a beige-salmon powder (60.6 mg, 32%). Mp: 77.5–79.9 °C. IR (neat)vmax: 3447, 3018, 1775, 1729, 1687, 1514, 1228, 1200, 1174, 1150, 1106 cm−1. 1H NMR (400 MHz, CDCl3) δ 6.91 (1H, s, 14-H), 4.72 (1H, br s, N
), 3.99–3.87 (2H, m, OC
2CH3), 2.28 (3H, s, OCOC
3), 2.27 (3H, s, OCOC
3), 1.20 (3H, d, J = 6.8 Hz, CHC
3), 1.18–1.11 (3H, m, OCH2C
3), 1.14 (3H, d, J = 6.9 Hz, CHC
3), 0.98 (3H, s, C
3), 0.96 (3H, s, C
3); 13C NMR (100 MHz, CDCl3) δ 168.76 (O
OCH3), 168.56 (O
OCH3), 154.15 (NH
O), 141.44, 140.12, 138.91, 135.80, 133.47, 124.68 (C14), 60.31 (O
H2), 55.73, 53.33, 40.79, 34.49, 33.89, 32.98, 31.66, 27.56, 23.10, 22.86, 21.49, 21.35, 20.55, 19.42, 18.14, 14.74 (OCH2
H3). ESI-MS m/z: 482.4 [M + Na]+, 940.5 [2M + Na]+. Anal. calcd for C26H37NO6·0.15C6H15N·0.4H2O: C, 67.0; H, 8.4; N, 3.3. Found: C, 67.2; H, 8.9; N, 2.9%.
:
1) to obtain 17 as a beige-yellow powder (100.5 mg, 57%). Mp: 72.6–74.1 °C. IR (neat)vmax: 3447, 3025, 1776, 1731, 1685, 1513, 1226, 1201, 1174, 1149 cm−1. 1H NMR (400 MHz, CDCl3) δ 6.91 (1H, s, 14-H), 4.74 (1H, br s, N
), 3.75–3.66 (1H, m, OC
2), 3.66–3.53 (2H, m, OC
2 (1H)), 2.26 (6H, s, 2 × OCOC
3), 1.20 (3H, d, J = 7.0 Hz, CHC
3), 1.14 (3H, d, J = 6.9 Hz, CHC
3), 0.98 (3H, s, C
3), 0.96 (3H, s, C
3), 0.85 (6H, d, J = 4.2 Hz, OCH2CH(C
3)2); 13C NMR (100 MHz, CDCl3) δ 168.81 (O
OCH3), 168.53 (O
OCH3), 154.24 (NH
O), 141.40, 140.06, 138.87, 136.34, 133.30, 124.67 (C14), 70.61 (O
H2), 56.12, 53.36, 40.73, 34.63, 33.87, 32.99, 31.73, 27.97, 27.53, 23.14, 22.79, 21.42, 21.32, 20.56, 19.39, 19.17 (2C, OCH2CH (
H3)2), 18.16. ESI-MS m/z: 510.4 [M + Na]+, 996.6 [2M + Na]+. Anal. calcd for C28H41NO6·0.2C6H15N·0.2H2O: C, 68.6; H, 8.75; N, 3.3. Found: C, 68.3; H, 9.3; N, 2.8%.
:
1) to obtain 18 as a white powder (119.2 mg, 65%). Mp: 67.3–69.7 °C. IR (neat)vmax: 3435, 3067, 1773, 1744, 1690, 1508, 1188, 1169, 1142 cm−1. 1H NMR (400 MHz, CDCl3) δ 8.00 (1H, s, 14-H), 4.89 (1H, br s, N
), 3.49 (3H, s, OC
3), 2.30 (3H, s, OCOC
3), 2.30 (3H, s, OCOC
3), 1.27 (3H, d, J = 6.9 Hz, CHC
3), 1.16 (3H, d, J = 6.9 Hz, CHC
3), 1.05 (3H, s, C
3), 0.96 (3H, s, C
3); 13C NMR (100 MHz, CDCl3) δ 197.39 (C7), 167.93 (2 × O
OCH3), 155.22 (NH
O), 145.49, 141.78, 140.93, 136.83, 132.00, 123.76 (C14), 56.41, 52.21, 50.10, 40.44, 35.61, 34.81, 33.63, 32.47, 27.92, 23.06, 22.53, 21.04, 21.01, 20.53, 18.82. ESI-MS m/z: 482.3 [M + Na]+. Anal. calcd for C25H33NO7·0.7H2O: C, 63.6; H, 7.3; N, 3.0. Found: C, 63.75; H, 7.7; N, 2.8%.
:
1) to afford the 19 as a white powder (123.0 mg, 74%). Mp: 67.5–69.2 °C. IR (neat)vmax: 3373, 3026, 1780, 1727, 1684, 1521, 1193, 1170, 1143 cm−1. 1H NMR (400 MHz, CDCl3) δ 8.00 (1H, s, 14-H), 4.86 (1H, br s, N
), 4.00–3.85 (2H, m, OC
2CH3), 2.30 (3H, s, OCOC
3), 2.29 (3H, s, OCOC
3), 1.26 (3H, d, J = 6.9 Hz, CHC
3), 1.17 (3H, d, J = 7.1 Hz, CHC
3), 1.15–1.07 (3H, m, OCH2C
3), 1.05 (3H, s, C
3), 0.96 (3H, s, C
3); 13C NMR (100 MHz, CDCl3) δ 197.55 (C7), 167.98 (O
OCH3), 167.85 (O
OCH3), 154.84 (NH
O), 145.45, 141.70, 140.89, 136.91, 132.07, 123.68 (C14), 60.96 (O
H2), 56.27, 50.14, 40.39, 35.69, 34.86, 33.57, 32.50, 27.86, 23.04, 22.58, 21.00 (2C), 20.53, 18.80, 14.52. ESI-MS m/z: 496.3 [M + Na]+. Anal. calcd for C26H35NO7·0.6H2O: C, 64.5; H, 7.5; N, 2.9. Found: C, 64.45; H, 8.1; N, 2.8%.
:
1) of DMEM (without D-glucose) and Ham's F12 supplemented with 0.5% P/S solution, 10% FBS, and 7.5 mM D-glucose. SW480 and SW620 cells were maintained in DMEM (without D-glucose) supplemented with 1% P/S solution, 5% FBS, and 12.5 mM D-glucose. Cells were maintained in a humidified atmosphere containing 5% CO2 at 37 °C. All studies used subconfluent cell monolayers.
000 cells per well), Caco-2 (4000 cells per well), HCT116 (3000 cells per well), Mia Paca-2 (2000 cells per well), SW480 (6000 cells per well), and SW620 (8000 cells per well) cells were seeded in 96-well plates and incubated for 24 h. Subsequently, the culture medium was switched with a new medium containing DMSO (control) or the compound at a specified concentration. Following a 72 h incubation period, the supernatant was aspirated, and 100 μL of a mixture (1
:
1) of filtered MTT solution (1 mg per mL PBS) and FBS-free medium was added. The plates were then incubated for 1 h at 37 °C. After this time, the supernatant was aspirated, and the formazan crystals were dissolved in 100 μL of DMSO in each well. Relative cell viability was calculated by measuring absorbance at 570 nm using a Benchmark Plus Microplate Reader (Bio-Rad Laboratories, Hercules, CA, USA). Results are presented as the mean ± standard deviation (SD) of IC50 values, determined by nonlinear regression analysis of dose–response curves (log(inhibitor) vs. response with variable slope (four parameters)) using GraphPad Prism 9 software (GraphPad Software, San Diego, CA, USA).
00 × g (20 min at 4 °C). After, the protein concentration on supernatants was quantified using Pierce™ bovine serum albumin standard ampules (2 mg mL−1) and Pierce™ BCA protein assay reagents A and B. A volume of protein extract corresponding to 5 μg of protein was loaded onto 12% SDS – polyacrylamide gels and separated by electrophoresis. The gels were then transferred to PVDF membranes, which were blocked with 5% skim milk in PBS-Tween-20 (0.1%) for 1 h at room temperature. Subsequently, the membranes were incubated with the primary antibodies for 30 mi at room temperature (actin) or overnight at 4 °C (CDK4, CDK6, cyclin A, Rb and SOD2/MnSOD). After incubation, the membranes were washed with PBS-Tween-20 (0.1%) and incubated with anti-mouse secondary antibody (40 min) or anti-rabbit secondary antibody (1 h) at room temperature. Finally, the membranes were washed with PBS-Tween-20 (0.1%) and treated with the Immobilon Western Chemiluminescent HRP Substrate Kit, and autoradiography was performed using an FPM-100A film processor (Fujifilm, Tokyo, Japan). western blot results were obtained from three independent experiments, except for cyclin A at 24 h (n = 2), Rb protein at 24 and 72 h (n = 2), and SOD2 (MnSOD) at 72 h (n = 2).Supplementary information: Selected spectroscopic information, dose–response curves, and additional molecular docking results. See DOI: https://doi.org/10.1039/d5ra02441b.
| This journal is © The Royal Society of Chemistry 2025 |