Abstract
Traditionally, Bidens pilosa L. is an edible herb utilized for various ailments. The study accomplished a complete analysis of B. pilosa extract including UPLC/T-TOF–MS/MS, GC–MS, and in vitro antiproliferative activity, in addition to molecular docking on kinase and aldose reductase enzymes. From GC–MS analysis, the percentage of identified unsaturated fatty acids (FAs) (11.38%) was greater than saturated FAs (8.69%), while the sterols percent (39.92%) was higher than the hydrocarbons percent (6.6%). Oleic and palmitic acids are the major FAs (9.48% and 6.14%, respectively). Phytochemical profile uncovered the presence of quercetin, kaempferol, myricetin, and isorhamnetin aglycones and/or glycoside derivatives alongside apigenin, acacetin, and luteolin derivatives. B. pilosa extract suppressed cell proliferation in a concentration-dependent manner against SNB-19 and SK-MEL-5 cell lines (IC50 1.66 ± 0.06 and 4.04 ± 0.14 mg/mL, respectively). These potentials aligned with the molecular docking results on aldose reductase and kinase enzymes with promising binding affinities (− 5.3 to − 8.89 kcal mol−1). B. pilosa metabolites were found as kinases and aldose reductase inhibitors, which rationalize their antiproliferative activity. Unfortunately, toxicity assessments were not performed to assess the safety of B. pilosa extract. Assessment of the therapeutic efficiency via in vivo and clinical studies is required.
Keywords: Bidens pilosa, Antiproliferative, Docking, Flavonoids, GC–MS, LC–MS/MS
Introduction
The Bidens genus (Asteraceae) comprises approximately 280 species [1]. Bidens pilosa L. is an annual and ruderal herb that grows in tropical and subtropical regions because of its outstanding resistance to unfavorable environmental circumstances [2]. B. pilosa grows with numerous ridged branches (1.5–2 m). Leaves are petioled oppositely pinnate arranged, with hairy serrately ovate leaflets (3–5) [3]. B. pilosa has capitulum inflorescence with white ray petals and yellow centers [3]. The seeds are brown to black and widely spread by wind, adhering to clothes and animal hair allowing its fast growth worldwide.
In the folk medicine, B. pilosa has been used with variable indications between countries. B. pilosa herb was acceptable in Europe for its astringent, diaphoretic, and diuretic effects [3]. In traditional Chinese medicine, B. pilosa is used to manage diabetes, inflammation, dysentery, and pharyngitis [4]. Moreover, B. pilosa is commonly known as Pica˜o preto in Brazil, where it is widely used for treating diabetes, ulcers, inflammation, and infections [5, 6]. In addition, the roots are regarded as applicable in treating malaria [5] and even tumors [7]. In Martinique, the herb decoction is used to manage inflammation and diabetes [1, 8]. In South Africa, the herbal tea of the aerial parts is thought to have anti-allergic and anti-inflammatory effects [9]. In Cuba, B. pilosa is known as an antitumor agent [6]. As dietary supplements, the dried herbs are sold in Taiwan; approximately 4 million dollars per year are marketed for diabetes management [10]. Studies of B. pilosa have shown its antihyperglycemic [11], antihypertensive [12], antiulcer [13], hepatoprotective [14], antipyretic [15], anti-inflammatory [9, 16], anti-leukemic [17], antimalarial [5], antibacterial [18], antioxidant [19], and antitumor effects [1, 15]. It has cytotoxic activities against several types of cancer cells [2]. Based on the reported biological activities, some countries like Brazil include B. pilosa as an official medicinal plant for public use [20]. The use of B. pilosa as a medicinal plant is feasible but further clinical trials and toxicity assessments are still rare.
The major phytoconstituents detected in B. pilosa are polyacetylenes [4, 11, 16], terpenes [18, 21], and flavonoids [2]. Polyacetylenes derivatives, in particular, phenylheptatriyne, are the important bioactive phytoconstituent found in B. pilosa essential oils [21]. Flavones, flavanones, and flavonols aglycones and/or glycosides were identified from B. pilosa, especially quercetin, and kaempferol glycosides [20]. Many scientific reports were proceeded on B. pilosa, owing to its traditional use and known bioactive constituents. Further studies are needed to highlight the action mechanism of its phytoconstituents. Hence, metabolomic profiling of B. pilosa is a basic step for offering undiscovered medicinal uses. In this respect, gas chromatography (GC) and high-resolution ultra-performance liquid chromatography (HR-UPLC) coupled with mass spectrometry (MS) represent prospective analytical analysis for untargeted metabolome report, providing efficient separation asides from the highly sensitive recognition of phytoconstituents. Diverse kinase enzymes activate transcription factors that transcribe numerous carcinogenic markers; consequently, kinase inhibitors may rationalize the antiproliferative activity [22]. Hence, the current study aimed to execute chemical profiling, in vitro antiproliferative assessment of B. pilosa, and verification with computational study on kinase and aldose reductase enzymes.
Material and Methods
Plant Material and Extraction
B. Pilosa L. was collected from Beni-Sueif, Egypt in May 2022. The plant identity was performed in the Faculty of Science, Beni-Sueif University, Beni-Sueif, Egypt. The plant aerial part was washed with tap water, dried under shade (20 days), and ground into a fine powder. The powdered materials (1 kg) were extracted in Soxhlet with aqueous ethanol (70%, 4 × 2 L, 2 h) and then filtered. The filtrate was evaporated (Rotavapor®, BÜCHI, Switzerland) [23].
In Vitro Antiproliferative Assay
The American Type Culture Collection (Manassas, VA, USA) provided SNB-19 (brain cancer cells) and SK-MEL-5 (skin cancer cells). Cell lines were cultured and maintained in RPMI-1640 medium with fetal bovine serum and penicillin/streptomycin (10% and 1%, respectively). The antiproliferation assay of B. Pilosa extract was proceeded by MTT assay [24]. Serial dilutions (100–1.56 mg mL−1) of B. Pilosa extract in DMSO were added to the cells (1 × 103 cells mL−1) which were placed in 96-well plates. Furthermore, MTT (10 µL) was added to each well and incubated at 37 °C for 4 h. Using a microplate ELISA reader (FLUO star Omega, Labtech, Germany), the absorbance (Abs) of the produced formazan was measured (490 nm). Each concentration was examined in triplicates and the statistical analysis was done using GraphPad Prism 6 (La Jolla, CA, USA). The cell viability % = [Abs of tested cells/ Abs of control cells)] × 100. The values of cell viability % were plotted versus the concentrations using non-linear regression analysis of Sigmoidal dose–response curve [25].
GC–MS Analysis
The lipoidal composition of B. pilosa was analyzed using a Trace GC-TSQ-MS (Thermo Scientific, TX, USA) over a capillary column TG-5MS. The column temperature was set at 50 °C (2 min), followed elevated by 5 °C/min to 250 °C and then by 30 °C/min to 300 °C (2 min). The injector and MS transfer line were maintained at 270 and 260 °C, respectively. Samples (1 µl) were injected and carried on Helium (1 mL/min) using an Autosampler AS1300 connected to a GC in split mode. Mass spectra covering m/z 50–650 were obtained and furthermore were compared with databases: NIST 14 and WILEY 09 [26].
HR-UPLC/T-TOF–MS/MS Analysis
The B. pilosa extract was analyzed at the Proteomics and Metabolomics unit of the Children’s Cancer Hospital, Cairo, Egypt. An Exion LC Triple TOF 5600 + system (SCIEX, Framingham, MA, USA) equipped with an X select HSS T3 C18 column (Waters Corporation, CT, USA) was used in negative and positive modes. Extract (50 mg) was dissolved in a working solvent: MilliQ water:methanol:acetonitrile (50:25:25, 50 µL) and then was diluted with the working solvent [23]. Samples (1 µg/µL, 10 µL) were injected using the following: solvent A was ammonium formate buffer (5 mM, pH 8) containing 1% methanol, for the negative mode, while in the positive mode, solvent A was ammonium formate buffer (5 mM, pH 3) containing 1% methanol, and in both modes, solvent B was 100% acetonitrile. The gradient elution (0.3 mL/min) was performed as follows: isocratic 90%:10% (0–1 min), linear from 90%:10% to 10% and 90% (1.1–20.9 min), isocratic 10%:90% (21–25 min), and finally isocratic 90% and 10% (25.1–28 min) of solvents A and B, respectively. The metabolites were recorded by Analyst TF 1.7.1, Peak view 2.2 (SCIEX, Framingham, MA, USA), and MS-DIAL 3.70 softwares [23]. MS (50–1100 m/z) was done on a Triple TOF 5600 + system equipped with a Duo-Spray source operating in the electron spray ionization mode (AB SCIEX, Framingham, MA, USA). The metabolites were characterized by generating the formula with an error limit of 20 ppm and considering Rt, MS2 data, compared to databases and literature [27].
Molecular Docking Study
MOE software (version 2016.10, Chemical Computing Group Inc., Montreal, Canada) was used. The structures of aldose reductase (AKR1B1; PDB: 2IKI) and ribosomal S6 kinase (RSK2 kinase; PDB: 3UBD) were obtained from the RCSB protein data bank. The standard preparation of target protein was applied. The target was validated by redocking the co-crystallized ligands (IDD388 and SL0101 to AKR1B1 and RSK2, respectively) offering low binding energy score (S) and small RMSD value. The essential amino acids were defined. The validated target was used to predict metabolite-target interactions [28].
Results and Discussion
In Vitro Antiproliferative Assay
Recent studies informed that B. pilosa has a promising in vitro and in vivo anticancer activity [29, 17]. However, its action mechanism has not been fully understood. The antiproliferation activity was evaluated for B. pilosa ethanol extract on SNB-19 and SK-MEL-5. The B. pilosa extract produced a decrease in cell viability of SNB-19 and SK-MEL-5 cells (IC50 1.66 ± 0.06 and 4.04 ± 0.14 mg/mL, respectively) comparable to doxorubicin (IC50 2.42 ± 0.08 and 11.82 ± 0.41 µg/mL, respectively).
GC–MS Analysis of the Lipoidal Matter
Quantitation was based on separated compounds’ relative peak area, Rt, and their area percentages. The GC–MS chromatogram revealed the detection of 19 major metabolites (66.59%) (Table 1, Fig. 1). The percentage of identified unsaturated FAs (11.38%) was higher than the percentage of saturated FAs (8.69%). At the same time, sterols were found to be higher than hydrocarbons percent (39.92% and 6.6%, respectively). Oleic acid is the major unsaturated FA (6.14%). However, palmitic and stearic acids are the major saturated FAs (6.14% and 2.55%, respectively). The percentage of identified sterols was higher than that of hydrocarbons (39.92% and 6.6%, respectively). Lupene-3,28-diol/botulin is the major sterol, followed by sitostenone (24.95% and 6.17%, respectively) [30].
Table 1.
Identified metabolites detected in B. pilosa lipoidal matter using GC–MS analysis
| # | Rt | Name | Formula | M.Wt | Area % |
|---|---|---|---|---|---|
| 1 | 26.20 | Palmitic acid, methyl ester | C17H34O2 | 270 | 2.55% |
| 2 | 26.96 | Palmitic acid | C16H32O2 | 256 | 3.59% |
| 3 | 29.33 | Linoleic acid, methyl ester | C19H34O2 | 294 | 1.32% |
| 4 | 29.47 | Oleic acid, methyl ester | C19H36O2 | 296 | 4.45% |
| 5 | 29.66 | Tetramethyl-hexadecenol | C20H40O | 296 | 1.14% |
| 6 | 29.66 | Methyl-octadecadienol | C19H36O | 280 | 1.14% |
| 7 | 29.99 | Stearic acid, methyl ester | C19H38O2 | 298 | 1.11% |
| 8 | 30.22 | Oleic acid | C18H34O2 | 282 | 5.03% |
| 9 | 30.67 | Stearic acid | C18H36O2 | 284 | 1.44% |
| 10 | 32.77 | Stigmasterol | C29H48O | 412 | 3.34% |
| 11 | 38.41 | Stigmastanol | C29H52O | 416 | 0.67% |
| 12 | 38.89 | Heptatriacotanol | C37H76O | 536 | 1.03% |
| 13 | 39.68 | Stigmastadiene-3-one | C29H46O | 410 | 3.23% |
| 14 | 39.97 | lupene-3,28-diol/betulin | C30H50O2 | 442 | 24.95% |
| 15 | 40.65 | Linoleic acid ethyl ester | C20H36O2 | 308 | 0.58% |
| 16 | 41.06 | Sitostenone | C29H48O | 412 | 6.17% |
| 17 | 41.28 | Cyclolanostan-3-ol, acetate | C32H54O2 | 470 | 1.56% |
| 18 | 41.63 | Tocopherol | C29H50O2 | 430 | 1.48% |
| 19 | 42.68 | Ethyl iso-allocholate | C26H44O5 | 436 | 1.81% |
| % Identified saturated fatty acids | 8.69% | ||||
| % Identified unsaturated fatty acids | 11.38% | ||||
| % Identified sterols | 39.92% | ||||
| % Identified hydrocarbons | 6.6% | ||||
| % of total identified compounds | 66.59% |
Fig. 1.
GC–MS chromatogram of B. pilosa lipoidal matter
UPLC/T-TOF–MS/MS Analysis
LC–MS can analyze a wide range of metabolites; they offer tools for dissecting immense plant biodiversity. The metabolomic profile of B. pilosa was studied using ESI–MS/MS in positive and negative modes. Flavones, flavanones, and flavonols aglycones with their glycosides or glucuronic derivatives were identified, with a high abundance of quercetin derivatives, alongside phenolic and organic acids (Table 2, Fig. 2). The major content of flavonoids reflects the plant’s diverse biological activities.
Table 2.
Identified metabolites in Bidens pilosa extract via UPLC-MS/MS using negative and positive ionization modes
| # | Rt | Metabolites | Formula | [M-H]− | [M + H]+ | Error PPM | MS2 |
|---|---|---|---|---|---|---|---|
| Flavonoids | |||||||
| 1 | 4.80 | Hesperidin | C28H34O15 | 609.1324 | 0.3 | 463, 301, 177, 151 | |
| 2 | 4.86 | Kaempferol-O-hexuronide | C21H18O12 | 461.077 | 0.1 | 285, 257, 135 | |
| 3 | 5.09 | Quercetin-O-hexuronide | C21H18O13 | 477.0676 | 479.1381 | − 6.6 | 301, 179, 151 |
| 4 | 5.33 | Luteolin-di-O-hexoside | C27H30O16 | 609.1437 | 611.18 | 8.6 | 447, 285, 151 |
| 5 | 5.38 | Luteolin-C-hexoside | C21H20O11 | 447.0947 | − 2.9 | 357, 327, 285, 151, 135 | |
| 6 | 5.52 | Baicalein-O-hexuronide | C21H18O11 | 445.077 | 447.1028 | 2.2 | 269, 117 |
| 7 | 5.64 | Quercetin-O-di-hexoside | C27H30O17 | 625.1737 | 627.2105 | 6.1 | 463, 301, 283 |
| 8 | 6.18 | Isoquercitrin | C21H20O12 | 463.0918 | 0.5 | 301, 283,255,151 | |
| 9 | 6.41 | Apigenin-C-hexoside | C21H20O10 | 433.1357 | 0.2 | 343, 313, 271 | |
| 10 | 6.48 | Rutin | C27H30O16 | 611.1806 | 2.2 | 465, 303 | |
| 11 | 6.50 | Eriodictyol-O-hexoside | C21H22O11 | 449.1194 | 0.7 | 287, 151, 135 | |
| 12 | 6.52 | Hyperoside | C21H20O12 | 463.0938 | 465.1223 | 0.8 | 301, 271, 255, 151 |
| 13 | 6.57 | Apigenin-O-hexoside | C21H20O10 | 431.0989 | 433.1263 | − 9.5 | 269, |
| 14 | 6.59 | Maritimetin-O-hexoside | C21H20O11 | 447.0909 | 449.1236 | 4.2 | 285 |
| 15 | 6.59 | Kaempferol-O-neohesperidoside | C27H30O15 | 593.1467 | 0.3 | 285 | |
| 16 | 6.62 | Kaempferol-O-bis-deoxyhexoside | C27H30O14 | 577.1507 | 579.1816 | 6.7 | 431, 285 |
| 17 | 6.66 | Luteolin-O-hexoside | C21H20O11 | 447.0921 | 2.7 | 285 | |
| 18 | 6.66 | Isorhamnetin-O-deoxyhexosyl-hexoside | C28H32O16 | 623.159 | 625.1852 | − 0.2 | 315, 300 |
| 19 | 6.79 | Okanin-O-hexoside | C21H22O11 | 451.1404 | − 0.7 | 289, 271, 179, 163, 153 | |
| 20 | 6.91 | Naringenin-O-hexoside | C21H22O10 | 433.1115 | 435.1358 | 4.5 | 271, 151, 119 |
| 21 | 7.10 | Vitexin-O-deoxyhexoside | C27H30O14 | 577.1575 | − 3.8 | 431, 269 | |
| 22 | 7.16 | Gossypin | C21H20O13 | 479.1038 | 3.8 | 317 | |
| 23 | 7.27 | Syringetin-O-hexoside | C23H24O13 | 507.1129 | 509.1445 | − 15.4 | 492, 345 |
| 24 | 7.30 | Quercetin-O-hexoside | C21H20O12 | 463.1217 | 465.1522 | 0.4 | 301, 283, 135 |
| 25 | 7.46 | Kaempferol-O-pentoside | C20H18O10 | 419.1226 | − 2.5 | 287, 259, 231 | |
| 26 | 7.46 | Quercetin-O-hexosyl-pentoside | C26H28O16 | 597.1898 | − 10.6 | 435, 303 | |
| 27 | 7.54 | Isorhamnetin-O-hexoside | C22H22O12 | 477.1333 | 4.9 | 315, 300, 151 | |
| 28 | 7.89 | Phlorizin | C21H24O10 | 435.1261 | 6.6 | 273, 151, 119 | |
| 29 | 8.63 | Quercetin-O-pentoside | C20H18O11 | 433.1016 | 435.0974 | 8.7 | 301, 193, 161, 151, |
| 30 | 8.66 | Eriodictyol | C15H12O6 | 287.0629 | 289.0796 | 0.5 | 213, 151, 135, 107 |
| 31 | 8.83 | Acacetin-O-rutinoside | C28H32O14 | 591.1729 | − 1.2 | 283, 268 | |
| 32 | 8.95 | Kaempferol-O-(p-coumaroyl)-hexoside | C30H26O13 | 593.1296 | − 0.5 | 447, 285 | |
| 33 | 9.22 | Quercetin | C15H10O7 | 301.0233 | 303.0576 | 1.5 | 255, 193, 151, 135, 121 |
| 34 | 10.13 | Apigenin | C15H10O5 | 269.0447 | 271.0641 | − 3.2 | 159, 151, 133, 117 |
| 35 | 10.31 | Hesperetin | C16H14O6 | 301.0724 | 303.0969 | − 0.4 | 286, 151, 134 |
| 36 | 10.34 | Luteolin | C15H10O6 | 285.0397 | 287.0589 | 3.3 | 257, 177, 151, 133, 107 |
| 37 | 10.56 | Trihydroxy-methoxyflavone | C16H12O6 | 299.0556 | − 1.7 | 284, 256, 151 | |
| 38 | 13.16 | Acacetin | C16H12O5 | 283.0606 | 285.0812 | 0.4 | 268, 151, 131 |
| 39 | 13.43 | Isorhamnetin | C16H12O7 | 315.0881 | 317.076 | 6.8 | 300, 269, 151, 107 |
| 40 | 14.35 | Naringenin | C15H12O5 | 271.0979 | 273.086 | 12.4 | 225, 136, 122 |
| 41 | 19.81 | Rhamnetin | C16H12O7 | 317.122 | 6.1 | 299 | |
| 42 | 19.82 | Kaempferide | C16H12O6 | 301.1492 | 0.8 | ||
| Phenolic acids | |||||||
| 43 | 1.24 | Gentisic acid | C7H6O4 | 153.0193 | 0.8 | 109, 91 | |
| 44 | 1.24 | Caffeic acid | C9H8O4 | 179.055 | − 0.6 | 161, 135, | |
| 45 | 1.73 | Chlorogenic acid | C16H18O9 | 353.0866 | 355.1069 | 3.1 | 191, 179, 161, 135 |
| 46 | 2.52 | Homogenentisic acid | C8H8O4 | 167.0331 | 0.5 | 149, 123, 108 | |
| 47 | 2.77 | Hydroxybenzoic acid | C7H6O3 | 137.0242 | 4.4 | 93, 75 | |
| 48 | 3.37 | Coumaric acid | C9H8O3 | 163.0233 | 165.093 | 10.2 | 119, 101 |
| 49 | 4.26 | Protocatechuic acid | C7H6O4 | 153.0183 | − 4.3 | 135, 109, 91 | |
| 50 | 4.82 | Dihydroxymandelate | C8H8O5 | 183.0079 | 184.9872 | 24.8 | 139, 109 |
| 51 | 5.24 | Sinapic acid-O-hexoside | C17H22O10 | 385.1803 | 8.4 | 223, 205 | |
| 52 | 6.50 | Methoxysalicylic acid | C8H8O4 | 167.0341 | 2 | 152, 123, 108 | |
| 53 | 8.40 | Ferulic acid | C10H10O4 | 193.0501 | 195.1168 | 1.4 | 178, 161, 149, 133 |
| Coumarin | |||||||
| 54 | 1.27 | Hydroxy-Methylcoumarin | C10H8O3 | 175.0423 | 7.9 | 157, 131, 113 | |
| 55 | 2.68 | Esculin | C15H16O9 | 339.0741 | 341.0936 | − 3 | 177, 133 |
| 56 | 4.68 | Dihydroxycoumarin | C9H6O4 | 177.0187 | 179.0312 | 0.9 | 149, 133, 105 |
| 57 | 7.09 | Scopoletin | C10H8O4 | 193.0547 | − 7.1 | 178 | |
| Acids | |||||||
| 58 | 1.01 | Malic acid | C4H6O5 | 133.0145 | − 0.9 | 115, 89, 71 | |
| 59 | 1.04 | Maleic acid | C4H4O4 | 115.0022 | 9.3 | 71 | |
| 60 | 1.06 | Hydroxy-butyric acid | C4H8O3 | 103.002 | 9.4 | 59 | |
| 61 | 1.07 | Lactic acid | C3H6O3 | 89.02383 | − 0.6 | 71 | |
| 62 | 1.11 | Succinic acid | C4H6O4 | 117.0177 | 9 | 99, 73 | |
| 63 | 1.15 | Citrate | C6H8O7 | 191.0556 | − 0.3 | 173, 129, 111, 85 | |
| 64 | 1.19 | Tartrate | C4H6O6 | 149.0448 | 3.2 | 131, 89, 87 | |
| 65 | 1.25 | Citramalate | C5H8O5 | 146.9487 | 3.6 | 129, 85 | |
| 66 | 1.28 | Isopropylmalic acid | C7H12O5 | 175.0605 | − 1 | 157, 131, 113, 69 | |
| 67 | 1.35 | Methylglutaric acid | C6H10O4 | 145.0258 | 13.1 | 127, 109, 101 | |
| 68 | 2.27 | Phenyllactic acid | C9H10O3 | 165.0562 | 1 | 147, 121, 103 | |
| 69 | 4.90 | Quinic acid | C7H12O6 | 191.0559 | 0.9 | 173, 127, 93 | |
| 70 | 5.09 | Shikimic acid | C7H10O5 | 173.0432 | − 4.3 | 155, 137, 131, 111, 93 | |
| Amino acids | |||||||
| 71 | 1.11 | Arginine | C6H14N4O2 | 175.121 | − 4.3 | ||
| 72 | 1.55 | Oxoproline | C5H7NO3 | 130.0488 | 3.9 | ||
| 73 | 1.71 | Hydroxyproline | C5H9NO3 | 130.0862 | 4.2 | ||
| 74 | 2.15 | Phenylalanine | C9H11NO2 | 166.0876 | − 0.6 | ||
| 75 | 2.80 | Tryptophan | C11H12N2O2 | 203.0821 | 1.3 | ||
| 76 | 3.94 | Homoisoleucine | C7H15NO2 | 144.0459 | 0.8 | ||
| Fatty acids | |||||||
| 77 | 12.82 | Hydroxy-hexadecanoic acid | C16H32O3 | 271.2264 | 9.1 | 253, 212 | |
| 78 | 18.48 | Linolenic acid | C18H30O2 | 277.2177 | 0.5 | 233 | |
| Sugar derivatives | |||||||
| 79 | 1.36 | Mannitol | C6H14O6 | 181.0724 | − 0.8 | ||
| 80 | 2.23 | Maltitol | C12H24O11 | 343.1402 | − 0.5 | ||
| 81 | 2.46 | Maltotriose | C18H32O16 | 503.1371 | 3.8 | 341, 179 | |
| 82 | 3.82 | Melibiose | C12H22O11 | 341.0872 | 7.6 | ||
Fig. 2.
Base peak chromatogram obtained from UPLC/T-TOF–MS/MS analysis of B. pilosa extract in negative ionization mode
Flavone Identification
Hesperidin (1) showed a deprotonated molecular ion peak at m/z 609.1324 and a subsequent loss of 146 and 308 Da, indicating the loss of deoxyhexosyl and deoxyhexosyl-hexosyl moieties yielding fragments at m/z 463 and 301 (hesperitin aglycone), respectively, with the characteristic fragments for flavonoids (177 and 151). Moreover, hesperetin (35) showed a deprotonated molecule at m/z 301.0724, besides the fragments of hesperetin aglycone (m/z 151,134). Luteolin-di-O-hexoside (4) showed a deprotonated molecule at m/z 609.1437 and subsequent losses of 162 and 324 Da at m/z 447 and 285 of hexosyl and di-hexosyl moieties, respectively. Luteolin-C-hexoside (5) showed a deprotonated molecule at m/z 447.0947 and the daughter peaks at m/z 357 and 327 representing [M-H-90]− and [M-H-120]−, respectively, indicating C-linkage glycoside; the luteolin aglycone moiety was confirmed at m/z 285 (Fig. 3). Luteolin-O-hexoside (17) showed a deprotonated signal at m/z 447.0921 and a neutral loss of hexosyl moiety at m/z 285. The molecular ion peak [M-H]− of luteolin (36) was noticed at m/z 285.0397, besides the characteristic fragments of luteolin aglycone (m/z 257, 177, 151, 133, and 107) (Table 2) [23].
Fig. 3.
Mass fragments of major classes identified from Bidens pilosa extract: a quercetin-O-hexuronide (3) and b luteolin-C-hexoside (5)
Apigenin-C-hexoside (9) displayed a protonated molecule at m/z 433.1357 and the characteristic peak at m/z 313 [M + H-120]+ indicating C-linkage hexoside; moreover, base peak signal at m/z 271 [M + H-hexosyl]+ signifies apigenin. The MS2 spectrum of the deprotonated molecule at m/z 431.0989 showed a daughter peak at m/z 269 [M-H-sugar]−, with the major fragments of apigenin, which was identified as apigenin-O-hexoside (13). The MS2 spectrum of the deprotonated molecule at m/z 269.0447 showed the loss of B-ring-H2O moiety yielding the daughter ion at m/z 159, besides the fragments at m/z 151, 133, and 117 characteristics for apigenin (34) (Table 2 ) [23].
Baicalein-O-hexuronide (6) showed a deprotonated molecule at m/z 445.0770 and the loss of 176 amu equivalent to hexuronyl moiety resulting in the base peak at m/z 269, representing baicalein aglycone. The MS2 spectrum of a deprotonated molecule at m/z 577.1575 showed two subsequent losses of 146 and 308 amu, representing the deoxyhexosyl and deoxyhexosyl-hexosyl moieties yielding the product ions at m/z 431 and 269, respectively, with the typical fragmentation pattern of vitexin, which was proposed as vitexin-O-deoxyhexoside (21). The deprotonated molecular ion peak of acacetin-O-rutinoside (31) was observed at m/z 591.1729 and the daughter ion at m/z 283, representing losses of 308 amu (rutinosyl moiety). Acacetin (38) showed a deprotonated molecule at m/z 283.0606, besides signals at m/z 151 and 131 representing 1,3A− and 1,3B−, respectively, for acacetin aglycone (Table 2 ) [27].
Flavonol Identification
Quercetin-O-hexuronide (3) was detected by its parent ion at m/z 477.0676 [M-H]− and a fragment at m/z 301 for losing of hexuronyl moiety (176 Da), besides the characteristic fragments of quercetin aglycone m/z 179 and 151 for 1,4B− and 1,3A−, respectively (Fig. 3). The MS2 spectrum of the deprotonated molecule at m/z 625.1737 showed two subsequent losses at m/z 463 and 301 representing [M-H-hexosyl]− and [M-H-di-hexosyl]−, respectively, moreover another fragment at m/z 283 [Ag-H2O]− with the major fragments of quercetin, which was proposed as quercetin-O-di-hexoside (7). Isoquercitrin (8) was detected by its parent ion at m/z 463.0918 [M-H]− and a fragment at m/z 301 [M-H-sugar]−, moreover, characteristic flavonoid fragments at m/z 283, 255, and 151. Rutin (10) was identified by its protonated molecule at m/z 611.1806. Moreover, deoxyhexosyl and deoxyhexosyl-hexosyl moieties were confirmed by the two daughter ions at m/z 465 and 303, respectively. Hyperoside (12) showed a deprotonated molecule at m/z 463.0938 and a loss of hexosyl at m/z 301 with 1,3A− fragment which signifies quercetin aglycone. Quercetin-O-hexoside (23) was identified by its deprotonated molecule at m/z 463.1217; moreover, hexosyl and Ag-H2O moieties were confirmed by the two daughter ions at m/z 301 and 283, respectively, with another fragment at m/z 135 representing 0,2A−. Quercetin-O-hexosyl-pentoside (26) was identified by its protonated molecule at m/z 597.1898; moreover, hexosyl and hexosyl-pentosyl moieties were confirmed by two daughter ions at m/z 435 and 303, respectively. The MS2 spectrum of a deprotonated molecule at m/z 433.1016 showed two product fragments at m/z 301 [M-H-pentosyl]− and 193 [M-H-B-ring]−, besides the characteristic fragments of quercetin, which was suggested as quercetin-O-pentoside (29). The quercetin (33) molecular ion peak [M-H]− was noticed at m/z 301.0233, besides the characteristic fragments of quercetin aglycone (m/z 255, 193, 151, and 135) (Table 2 ) [27].
Kaempferol-O-hexuronide (2) showed a deprotonated molecular ion peak at m/z 461.0770, and a fragment at m/z 285 signifies kaempferol aglycone, moreover its 0,3A− fragment at m/z 135. Kaempferol-O-neohesperidoside (15) was identified by a deprotonated molecule at m/z 593.1467; moreover, neohesperidoside moiety was confirmed by the daughter peak at m/z 285. The MS2 spectrum of a deprotonated molecule at m/z 577.1507 showed two product ions at m/z 431 [M-H-deoxyhexosyl]− and 285 [M-H-di-deoxyhexosyl]−, with the typical fragments of kaempferol, which was suggested as kaempferol-O-bis-deoxyhexoside (16). The molecular ion peak [M + H]+ of kaempferol-O-pentoside (25) was observed at m/z 419.1226, followed by daughter ions at m/z 287 [M + H-pentosyl]+, 259 [M + H-pentosyl-CO]+, and 231 [M + H-pentosyl-2CO]+. Kaempferol-O-(coumaroyl)-hexoside (32) showed a deprotonated molecule at m/z 593.1296 and a fragment at m/z 447 [M-H-coumaroyl]−, with another fragment at m/z 285 [M-H-coumaroyl-hexosyl]− (Table 2 ) [27].
Isorhamnetin-O-deoxyhexosyl-hexoside (18) showed a deprotonated molecule at m/z 623.1590 and the aglycone fragment at m/z 315 [M-H-deoxyhexosyl-hexosyl]−. Isorhamnetin-O-hexoside (27) showed a molecule ion [M-H]− at m/z 477.1333, besides the aglycone base peak at m/z 315. Gossypin (22) showed a deprotonated molecule at m/z 479.1038, and the loss of hexosyl moiety was confirmed by the peak at m/z 317. Syringetin-O-hexoside (23) showed a molecule peak [M-H]− at m/z 507.1129, followed by the signal of syringetin aglycone at m/z 345. The protonated ion peaks [M + H]+ of rhamnetin and kaempferide (41, 42) were noticed at m/z 317.1220 and 301.1492, respectively (Table 2 ).
Flavanone Identification
Eriodictyol-O-hexoside (11) showed a deprotonated molecule at m/z 449.1194, besides the eriodictyol aglycone fragment at m/z 287. Eriodictyol (30) showed a deprotonated molecule at m/z 287.0629, moreover, the characteristic flavonoid fragments at m/z 213, 151, 135, and 107. Naringenin-O-hexoside (20) showed a signal at m/z 433.1115 [M-H]− and a loss of hexosyl moiety at m/z 271, besides the naringenin fragments at m/z 151 and 119 for 1,3A− and 1,3B−, respectively. Naringenin (40) showed a signal at m/z 271.0979 [M-H]−, besides the fragments at m/z 225, 136, and 122) [30]. Maritimetin-O-hexoside (14) showed a signal at m/z 447.0909 [M-H]− and a base peak at m/z 285 [M-H-hexosyl]−. In the same behavior, okanin-O-hexoside (19) and phlorizin (28) were identified (Table 2 ).
Phenolic, Organic, and Amino Acid Identification
The expected losses of 18, 44, and 62 Da, equivalent to the loss of H2O, CO2, and both, respectively, were observed in MS2 fragmentations of acids. Gentisic acid (43) was detected by its deprotonated molecule at m/z 153.0193, moreover ions at m/z 109 and 91. Protocatechuic acid (49) showed a deprotonated molecule at m/z 153.0183 and subsequent losses of H2O, CO2, and both moieties. In the same approach, caffeic, chlorogenic, coumaric, and ferulic acids (44, 45, 48, 53) were identified. The common neutral loss of CO2 (44 Da) was observed in MS2 fragments of organic acids as in succinic and malic acids (58, 62) (Table 2) [27]. Various amino acids were identified such as oxoproline, hydroxyproline, and phenylalanine (Table 2).
Molecular Docking of Quercetin and Phenolic Derivatives on Aldose Reductase Protein
Inhibition of aldose reductase has a role in cancer management by preventing the activation of many transcription factors responsible for producing carcinogenic mediators [31]. The studies informed that the activity of aldose reductase increased in human cancers [32]. Inhibitors of aldose reductase showed a possible increase in the doxorubicin cytotoxic effect with a diminution in its cardiotoxicity [33, 34]. Flavonols mainly quercetin were reported as inhibitors of kinase enzymes [35, 36]. Quercetin derivatives are inhibiting tumors in rats [37]. Thus, molecular docking of B. pilosa metabolites on aldose reductase and kinase enzymes should be performed to rationalize its observed antiproliferative activity. The active pocket of AKR1B1 consists mainly of VAL47, TYR48, GLN49, HIS110, GLN183, and TRP111, where its co-crystalized ligand IDD388 was interacted by two H–H bonding with TYR48 and HIS110 (− 9.87 kcal mol−1 binding affinity). Five major metabolites were docked on aldose reductase protein with low energy of binding affinities (− 5.3 to − 7.2 kcal mol−1) (Table 3). Quercetin-4′-O-glucoside exhibited interactions (− 5.75 kcal mol−1 binding affinities) with four H-bonds between GLN183, HOH1218, ASN160, and HOH1056. The best docking activity was recorded on chlorogenic acid (− 7.20 kcal mol−1) that interacts with three H-bonds, mainly with ASP43, ILE260, and SER210 (Fig. 4).
Table 3.
Docked conformations of quercetin and phenolic derivatives from Bidens pilosa on aldose reductase (AKR1B1; PDB: 2IKI) protein
| Metabolites | ΔGa (kcal mol−1)/No. of interactions | RMSD | Interactions | Distance/E (kcal/mol) |
|---|---|---|---|---|
| Quercetin-3-O-glucuronide | − 5.31/3 | 1.62 | HOH1184 | 2.92/ − 0.8 |
| HOH1050 | 2.69/ − 0.9 | |||
| TRP20 | 4.73/ − 1.5 | |||
| Isoquercitrin | − 5.49/3 | 2.32 | HOH1184 | 3.08/ − 0.9 |
| HOH1050 | 2.88/ − 1.4 | |||
| HOH1056 | 3.05/ − 1.1 | |||
| Quercetin-4′-O-glucoside | − 5.75/5 | 2.14 | GLN183 | 3.53/ − 0.7 |
| HOH1218 | 2.92/ − 1.0 | |||
| ASN160 | 2.96/ − 2.5 | |||
| HOH1056 | 2.80/ − 1.5 | |||
| TYR209 (pi-pi) | 3.69/ − 0.0 | |||
| Chlorogenic acid | − 7.20/3 | 1.46 | ASP43 | 2.79/ − 3.2 |
| ILE260 | 2.97/ − 4.6 | |||
| SER210 | 2.92/ − 2.9 | |||
| Ferulic acid | − 5.3/3 | 1.11 | HIS110 | 2.91/ − 1.8 |
| TRP20 (H-pi) | 4.06 − 0.8 | |||
| TRP20 (H-pi) | 4.03/ − 0.8 |
Fig. 4.
The interaction of quercetin-4′-O-glucoside (yellow) on aldose reductase protein (PDB ID: 2IKI) in the binding pocket: a 2D and b 3D diagrams
On the other hand, the active site of RSK2 consists mainly of PHE79, LYS100, VAL101, LYS103, LEU147, ASP148, GLU197, and LEU200 amino acids. The crystal ligand, SL0101, interacts with three H–H bonds with LYS100, ASP148, and GLU197 (− 9.54 kcal mol−1). The major metabolites were docked on RSK2 protein and demonstrated auspicious binding affinities (− 5.6 to − 8.89 kcal mol−1) (Table 4) with a variety of degrees of interactions. Isoquercitrin exhibited interactions with four H-bonds between SER78, GLU197, and LEU200 (− 8.89 kcal mol−1) (Fig. 5). The low binding energy of interactions between metabolites and aldose reductase and kinase proteins may justify its antiproliferative activity [38]. The phenolic and hydroxyl OH groups in metabolites are essential for interaction acting as a hydrogen bond acceptor-donator. Moreover, it was found that quercetin-4′-O-glucoside occupies the pocket of aldose reductase better than its co-crystallized ligand. Moreover, quercetin-4′-O-glucoside and isoquercitrin exhibited binding scores to targeted enzymes relatively equal to the co-crystallized ligands, expected acts as kinases and aldose reductase inhibitors leading to rationalizing its antiproliferative activity.
Table 4.
Docked conformations of quercetin and phenolic derivatives from Bidens pilosa on ribosomal S6 kinase (RSK2) (PDB: 3UBD) protein
| Metabolites | ΔGa (kcal mol−1)/No. of interactions | RMSD | Interactions | Distance/E (kcal/mol) |
|---|---|---|---|---|
| Quercetin-3-O-glucuronide | − 8.29/3 | 1.25 | GLU197 | 2.65/ − 3.6 |
| LEU200 (pi-H) | 3.89/ − 1.0 | |||
| PHE79 (pi-pi) | 3.85/ − 0.0 | |||
| Isoquercitrin | − 8.89/5 | 1.41 | SER78 | 3.18/ − 0.7 |
| GLU197 | 2.73/ − 4.3 | |||
| GLU197 | 2.66/ − 2.5 | |||
| LEU200 (pi-H) | 3.77/ − 0.8 | |||
| PHE79 (pi-pi) | 3.64/ − 0.0 | |||
| Quercetin-4′-O-glucoside | − 7.22/3 | 1.10 | ASP148 | 2.82/ − 3.4 |
| LYS216 | 2.86/ − 1.9 | |||
| PHE79 (pi-pi) | 3.70/ − 0.0 | |||
| Chlorogenic acid | − 6.48/3 | 2.76 | ASP148 | 3.44/ − 0.8 |
| ASP148 | 2.67/ − 2.7 | |||
| LEU147 (pi-H) | 3.83/ − 1.3 | |||
| Ferulic acid | − 5.62/2 | 0.81 | ASP148 | 2.64/ − 2.4 |
| LEU147 (pi-H) | 4.11/ − 0.7 |
Fig. 5.
The interaction of isoquercitrin (yellow) on RSK2 protein (PDB: 3UBD) in the binding pocket: a 2D and b 3D diagrams
Conclusion
Metabolomic profiling of the B. pilosa revealed its enrichment of flavonoids and phenolic acids besides coumarins, fatty, and amino acids. Moreover, B. pilosa extract showed in vitro antiproliferative activity. Based on in silico study, major metabolites act as inhibitors of kinases and aldose reductase leading to rationalizing its antiproliferative activity. The use of B. pilosa as a medicinal plant is feasible but further clinical trials and toxicity assessments are still rare.
Acknowledgements
The authors extend their appreciation to the Researchers Supporting Project number (RSP2025R366) King Saud University, Riyadh, Saud Arabia.
Author Contribution
Methodology and writing draft manuscript: D.S.A., A.E.E., A.A.E., E.M.K, and S.A.A.; artwork and schemes: D.S.A., H.S.M., S.A.A.; review and editing: D.S.A., A.E.E., A.A.E., E.M.K., and S.A.A.; conceptualization, validation, supervision: A.E.E., E.M.K., S.A.A., and M.M.H. All authors have read and agreed to the published version of the manuscript.
Funding
Open access funding provided by The Science, Technology & Innovation Funding Authority (STDF) in cooperation with The Egyptian Knowledge Bank (EKB).
Data Availability
The data that support the findings of this study are available on request.
Declarations
Ethical Approval
Not applicable.
Competing Interests
The authors declare no competing interests.
Footnotes
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
References
- 1.Xuan, T. D., & Khanh, T. D. (2016). Chemistry and pharmacology of Bidens pilosa: An overview. Journal of Pharmaceutical Investigation,46, 91–132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Chiang, Y.-M., Chuang, D.-Y., Wang, S.-Y., Kuo, Y.-H., Tsai, P.-W., & Shyur, L.-F. (2004). Metabolite profiling and chemopreventive bioactivity of plant extracts from Bidens pilosa. Journal of Ethnopharmacology,95(2–3), 409–419. [DOI] [PubMed] [Google Scholar]
- 3.Mitich, L. W. (1994). Beggarticks. Weed Technology,8(1), 172–175. [Google Scholar]
- 4.Chiang, Y.-M., Chang, C.L.-T., Chang, S.-L., Yang, W.-C., & Shyur, L.-F. (2007). Cytopiloyne, a novel polyacetylenic glucoside from Bidens pilosa, functions as a T helper cell modulator. Journal of Ethnopharmacology,110(3), 532–538. [DOI] [PubMed] [Google Scholar]
- 5.Brandão, M. G. L., Krettli, A. U., Soares, L. S. R., Nery, C. G. C., & Marinuzzi, H. C. (1997). Antimalarial activity of extracts and fractions from Bidens pilosa and other Bidens species (Asteraceae) correlated with the presence of acetylene and flavonoid compounds. Journal of Ethnopharmacology,57(2), 131–138. [DOI] [PubMed] [Google Scholar]
- 6.Valdés, L., Humberto, A., de León Rego, L., et al. (2001). Bidens pilosa Linné. Rev Cuba Plantas Med,6(1), 28–33. [Google Scholar]
- 7.Alvarez, L., Marquina, S., Villarreal, M. L., Alonso, D., Aranda, E., & Delgado, G. (1996). Bioactive polyacetylenes from Bidens pilosa. Planta Medica,62(04), 355–357. [DOI] [PubMed] [Google Scholar]
- 8.Longuefosse, J.-L., & Nossin, E. (1996). Medical ethnobotany survey in Martinique. Journal of Ethnopharmacology,53(3), 117–142. [DOI] [PubMed] [Google Scholar]
- 9.Horiuchi, M., & Seyama, Y. (2006). Antiinflammatory and antiallergic activity of Bidens pilosa L. var. radiata SCHERFF. Journal of Health Science,52(6), 711–717. [Google Scholar]
- 10.Young, P. H., Hsu, Y. J., Yang, W. C., & others. (2010). Bidens pilosa and its medicinal use. In Recent progress in medicial plants/drug plants II vol 28 (Houston, TX, USA: Stadium Press LLC). Stadium Press LLC.
- 11.Chien, S.-C., Young, P. H., Hsu, Y.-J., Chen, C.-H., Tien, Y.-J., Shiu, S.-Y., others. (2009). Anti-diabetic properties of three common Bidens pilosa variants in Taiwan. Phytochemistry, 70(10), 1246–1254. [DOI] [PubMed]
- 12.Dimo, T., Rakotonirina, S. V., Tan, P. V., Azay, J., Dongo, E., & Cros, G. (2002). Leaf methanol extract of Bidens pilosa prevents and attenuates the hypertension induced by high-fructose diet in Wistar rats. Journal of Ethnopharmacology,83(3), 183–191. [DOI] [PubMed] [Google Scholar]
- 13.Alvarez, A., Pomar, F., Sevilla, M. A., & Montero, M. J. (1999). Gastric antisecretory and antiulcer activities of an ethanolic extract of Bidens pilosa L. var. radiata Schult. Bip. Journal of Ethnopharmacology,67(3), 333–340. [DOI] [PubMed] [Google Scholar]
- 14.Yuan, L.-P., Chen, F.-H., Ling, L., Dou, P.-F., Bo, H., Zhong, M.-M., & Xia, L.-J. (2008). Protective effects of total flavonoids of Bidens pilosa L.(TFB) on animal liver injury and liver fibrosis. Journal of Ethnopharmacology,116(3), 539–546. [DOI] [PubMed] [Google Scholar]
- 15.Sundararajan, P., Dey, A., Smith, A., Doss, A. G., Rajappan, M., & Natarajan, S. (2006). Studies of anticancer and antipyretic activity of Bidens pilosa whole plant. African Health Sciences,6(1), 27–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Pereira, R. L. C., Ibrahim, T., Lucchetti, L., da Silva, A. J. R., & de Moraes, V. L. G. (1999). Immunosuppressive and anti-inflammatory effects of methanolic extract and the polyacetylene isolated from Bidens pilosa L. Immunopharmacology,43(1), 31–37. [DOI] [PubMed] [Google Scholar]
- 17.Chang, J.-S., Chiang, L.-C., Chen, C.-C., Liu, L.-T., Wang, K.-C., & Lin, C.-C. (2001). Antileukemic activity of Bidens pilosa L. var. minor (Blume) Sherff and Houttuynia cordata Thunb. The American Journal of Chinese Medicine,29(02), 303–312. [DOI] [PubMed] [Google Scholar]
- 18.Deba, F., Xuan, T. D., Yasuda, M., & Tawata, S. (2008). Chemical composition and antioxidant, antibacterial and antifungal activities of the essential oils from Bidens pilosa Linn. var. Radiata. Food Control,19(4), 346–352. [Google Scholar]
- 19.Yang, H.-L., Chen, S.-C., Chang, N.-W., Chang, J.-M., Lee, M.-L., Tsai, P.-C., & others. (2006). Protection from oxidative damage using Bidens pilosa extracts in normal human erythrocytes. Food and Chemical Toxicology, 44(9), 1513–1521. [DOI] [PubMed]
- 20.Lima Silva, F., Fischer, D. C. H., Fechine Tavares, J., Sobral Silva, M., de Athayde-Filho, P., & Barbosa-Filho, J. M. (2011). Compilation of secondary metabolites from Bidens pilosa L. Molecules,16(2), 1070–1102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Priestap, H. A., Bennett, B. C., & Quirke, J. M. E. (2008). Investigation of the essential oils of Bidens pilosa var. minor, Bidens alba and Flaveria linearis. Journal of Essential Oil Research,20(5), 396–402. [Google Scholar]
- 22.Tammali, R., Srivastava, K. S., & Ramana, V. K. (2011). Targeting aldose reductase for the treatment of cancer. Current Cancer Drug Targets,11(5), 560–571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.El Gizawy, H. A., El-Haddad, A. E., Attia, Y. M., Fahim, S. A., Zafer, M. M., & Saadeldeen, A. M. (2022). In vitro cytotoxic activity and phytochemical characterization (UPLC/T-TOF-MS/MS) of the watermelon (Citrullus lanatus) rind extract. Molecules,27(8), 2480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Mosmann, T. (1983). Rapid colorimetric assay for cellular growth and survival: Application to proliferation and cytotoxicity assays. Journal of Immunological Methods,65(1–2), 55–63. 10.1016/0022-1759(83)90303-4 [DOI] [PubMed] [Google Scholar]
- 25.El-Haddad, A. E., Saadeldeen, A. M., & El-Emam, S. Z. (2019). Anti-angiogenic activity of major phenolics in tamarind assessed with molecular docking study on VEGF kinase proteins. Pakistan Journal of Biological Sciences,22(10), 502–509. [DOI] [PubMed] [Google Scholar]
- 26.Abd El-Kareem, M. S. M., Rabbih, M. A. E. F., Selim, E. T. M., Elsherbiny, E. A. E., & El-Khateeb, A. Y. (2016). Application of GC/EIMS in combination with semi-empirical calculations for identification and investigation of some volatile components in basil essential oil. International Journal of Analytical Mass Spectrometry and Chromatography,4(1), 14–25. [Google Scholar]
- 27.Mounir, R., Alshareef, W. A., El Gebaly, E. A., El-Haddad, A. E., Ahmed, A. M. S., Mohamed, O. G., & others. (2023). Unlocking the power of onion peel extracts: Antimicrobial and anti-inflammatory effects improve wound healing through repressing notch-1/NLRP3/caspase-1 signaling. Pharmaceuticals, 16(10), 1379. [DOI] [PMC free article] [PubMed]
- 28.Gendy, A., Elnagar, M. R., Soubh, A., Al-Mokaddem, A., El-Haddad, A., & El-Sayed, M. K. (2021). Morin alleviates hepatic ischemia/reperfusion-induced mischief: In vivo and in silico contribution of Nrf2, TLR4, and NLRP3. Biomedicine & Pharmacotherapy,138, 111539. [DOI] [PubMed] [Google Scholar]
- 29.Wu, L.-W., Chiang, Y.-M., Chuang, H.-C., Wang, S.-Y., Yang, G.-W., Chen, Y.-H., & Shyur, L.-F. (2004). Polyacetylenes function as anti-angiogenic agents. Pharmaceutical Research,21, 2112–2119. [DOI] [PubMed] [Google Scholar]
- 30.El Gizawy, H. A., El-Haddad, A. E., Saadeldeen, A. M., & Boshra, S. A. (2022). Tentatively identified (UPLC/T-TOF–MS/MS) compounds in the extract of saussurea costus roots exhibit in vivo hepatoprotection via modulation of HNF-1$α$, sirtuin-1, C/ebp$α$, miRNA-34a and miRNA-223. Molecules,27(9), 2802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Tanagala, K. K. K., Baba, A. B., Kowshik, J., Reddy, G. B., & Nagini, S. (2018). Gedunin, a neem limonoid in combination with epalrestat inhibits cancer hallmarks by attenuating aldose reductase-driven oncogenic signaling in SCC131 oral cancer cells. Anti-Cancer Agents in Medicinal Chemistry (Formerly Current Medicinal Chemistry-Anti-Cancer Agents),18(14), 2042–2052. [DOI] [PubMed] [Google Scholar]
- 32.Saraswat, M., Mrudula, T., Kumar, P. U., Suneetha, A., TS, R. R., Srinivasulu, M., & Reddy, B. (2006). Overexpression of aldose reductase in human cancer tissues. Medical Science Monitor: International Medical Journal Of Experimental And Clinical Research, 12(12), CR525-529. [PubMed]
- 33.Sonowal, H., Pal, P., Shukla, K., Saxena, A., Srivastava, S. K., & Ramana, K. V. (2018). Aldose reductase inhibitor, fidarestat prevents doxorubicin-induced endothelial cell death and dysfunction. Biochemical Pharmacology,150, 181–190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Antony, P., & Vijayan, R. (2015). Identification of novel aldose reductase inhibitors from spices: A molecular docking and simulation study. PLoS ONE,10(9), e0138186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Boly, R., Gras, T., Lamkami, T., Guissou, P., Serteyn, D., Kiss, R., & Dubois, J. (2011). Quercetin inhibits a large panel of kinases implicated in cancer cell biology. International Journal of Oncology,38(3), 833–842. [DOI] [PubMed] [Google Scholar]
- 36.Hou, D.-X., & Kumamoto, T. (2010). Flavonoids as protein kinase inhibitors for cancer chemoprevention: Direct binding and molecular modeling. Antioxidants & redox signaling,13(5), 691–719. [DOI] [PubMed] [Google Scholar]
- 37.Devipriya, S., Ganapathy, V., & Shyamaladevi, C. S. (2006). Suppression of tumor growth and invasion in 9,10 dimethyl benz (a) anthracene induced mammary carcinoma by the plant bioflavonoid quercetin. Chemico-biological Interactions,162(2), 106–113. [DOI] [PubMed] [Google Scholar]
- 38.Francis, R., Kalyanaraman, R., Boominathan, V., Parthasarathy, S., Chavaan, A., Ansari, I. A., & Tharumasivam, S. V. (2024). Piperine’s potential in treating polycystic ovarian syndrome explored through in-silico docking. Scientific Reports,14(1), 21834. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
The data that support the findings of this study are available on request.





