Abstract
Fruit wastes are becoming popular as treasures for drug discovery in different classes of therapeutics. This research aimed to investigate the phytochemicals and potential bioactivities, such as antimicrobial, thrombolytic, anti-inflammatory, analgesic, and antidiarrheal properties of commonly available mandarin orange (Citrus reticulata Blanco) peel through experimental and computational techniques. Extensive chromatographic and 1H-NMR spectroscopic analysis was employed to isolate four purified compounds, which were characterized as tangeretin (A), nobiletin (B), limonin (C), and β-sitosterol (D). Furthermore, GC-MS/MS analysis detected over 90 compounds with a notable number of polymethoxyflavones, including nobiletin (29.04%), tangeretin (15.55%), artemetin (8.1%), 6-demethoxytangeretin (1.28%), sinensetin (0.95%), demethylnobiletin (0.14%), pebrellin (0.10%), and salvigenin (0.04%). Dichloromethane soluble fraction (DCMSF) exerted the highest antimicrobial potency Candia albicans against (20 mm zone of inhibition) in the disk diffusion assay method. The aqueous soluble fraction (AQSF) exhibited 34.71% and 48.14% inhibition in hypotonic solution-induced and heat-induced hemolysis in the membrane stabilizing assay. Similarly, the AQSF exhibited the highest anti-thrombotic property with 32.57% clot lysis. The investigated 3 doses of the methanolic extracts (100, 200, and 400 mg/kg body weight) exerted statistically significant in vivo central analgesic effects in a tail-flicking method in a time-dependent manner. Moreover, all the doses exhibited significant efficacy in inhibiting acetic acid-induced abdominal writhing and castor oil-induced diarrheal episodes in mouse models. The molecular docking studies corroborated the existing in vitro and in vivo findings by demonstrating better or comparable binding affinities toward the respective receptors and favorable pharmacokinetic properties and toxicological profiles. The present findings indicate that C. reticulata is a rich source of polymethoxyflavones, demonstrating potential efficacy against microbial infections, thrombosis, inflammation, pain, and diarrhea. Nonetheless, comprehensive phytochemical screening is imperative to identify additional bioactive compounds and evaluate their pharmacological effects against several chronic health conditions, grounded in their traditional uses and current evidence.
Keywords: Mandarin orange (Citrus reticulata Blanco), phytochemical isolation, NMR, GC-MS/MS analysis, antimicrobial properties, anti-inflammatory, analgesics, antidiarrheal
Introduction
The investigation of contemporary synthetic medications is approached with caution due to their known adverse effects. 1 In contrast, conventional herbals are obtaining popularity as they are perceived as environmentally friendly, more natural, and fewer adverse effects. 2 So, despite all of the benefits of existing synthetic medications, scientists carry on to favor nature-based remedies. 3 The unique phytoconstituents of various plant parts contribute to the diverse potential of medicinal species, functional foods, or food byproducts in alleviating and treating various human disorders.3-5 In Indian culture, approximately 80 000 species of medicinal plants have been utilized as traditional medication since ancient times. 6 Plant-based medications serve as the primary resource for medical care for a substantial portion of the global population.7-9 Particularly in low-income nations, where facilities are limitted, traditional therapy and ethnic values play a significant role, making natural remedies an affordable choice for essential healthcare.10,11
Mandarin fruit (Citrus reticulata Blanco), a wide species that belongs to the family Rutaceae valued not only for its appealing flavor but also for its numerous therapeutic properties, including anticancer, antioxidant, anti-hyperlipidemic, anti-inflammatory, and antidiabetic effects.12,13 These health benefits are attributed to the rich presence of phytochemicals such as organic acids, carotenoids, sugars, amino acids, flavonoids, polyphenols, limonoids, and phenolic acids in mandarin fruit. 13 The traditional uses of mandarin fruit extend to laxative, aphrodisiac, antiemetic, astringent, and tonic properties, as well as skin-related benefits.14-16 It is also used as a stomachic and carminative.15,17,18 The pericarp and endocarp possess medicinal qualities, which are employed in the treatment and management of various conditions such as analgesia, anticholesterolemia, anti-inflammatory, antiseptic, anti-asthmatic, carminative, antiscorbutic, expectorant, antitussive, and stomachic.18,19 The immature green exocarp is utilized for alleviating, gastrointestinal distension, liver cirrhosis, liver, hypochondrium, spleen enlargement, as well as chest pains. The seed exhibits carminative and analgesic properties, making it a remedy for hernias, mastitis, lumbago, and discomfort or enlargement of the testicles. 19
In addition to its extensive biological significance, some phytochemical studies revealed that C. reticulata is a treasure trove of bioactive substances. Another report indicates that polymethoxyflavones (PMFs) are plentiful in citrus peels, particularly in mature citrus peels. These peels have been traditionally employed in Chinese medicine for numerous years to alleviate various conditions, including stomach upset, skin inflammation, cough, ringworm infections, muscle pain, and possibly lowering blood pressure.20,21
The current study utilizes Gas Chromatography-Mass Spectrometry (GC-MS) and NMR spectroscopy to identify and isolate bioactive components from methanolic extract of C. reticulata fruits peel. This research additionally explored the antimicrobial, anti-inflammatory, thrombolytic, central and peripheral analgesic, and anti-diarrheal properties of this species. The outcomes may be helpful to create a new way to discover new drugs and enhance the nutraceuticals and medicinal values of this very commonly available fruit peel waste of the C. reticulata.
Methods and Materials
Sample collection and drying
In July 2023, Mandarin oranges (Citrus reticulata Blanco) were procured from the West Dhanmondi local market in Dhaka, Bangladesh. The batch originated from the Yunnan region in southwest China. Following the purchase from the market, the procedure involved manually collecting the fresh peels and subsequently washing them with fresh water. The drying process was facilitated by prolonged exposure to open air. Strict protocols were employed to ensure that the ambient temperature remained below 30°C during the drying regimen. This precaution was taken to prevent any potential degradation and to ensure the preservation of heat-sensitive compounds. The dried fresh peels of C. reticulata are depicted in supplementary Figure S1.
Chemicals and reagents
Analytical-grade reagents and substances were employed in the study. Tween 80 and the suspending medium for the extract were procured from Merck (Germany). Glibenclamide, normal saline solution, and loperamide were supplied by BEXIMCO Pharmaceuticals Ltd., Dhaka, Bangladesh (Additional chemicals included aspirin, diclofenac sodium, and sodium thiopental.).
Extraction
Following the shadow air drying, 500 g of powdered fruit peel was soaked in 2.5 L of methanol within a sealed amber-colored vessel for 15 days with irregular jerking. The resulting solvent mixture was filtered using a clean cotton pad and filter pad. The methanol was then removed using an “EYELA Rotavapor” rotary evaporator at lower pressures and approximately 40°C to yield 58 g of gummy extract from the methanolic extract of C. reticulate peel. This represented 11.6% of the weight of the powdered fruit peel.
Fractionation
Solvent-solvent differentiation of the extract was performed using S. Morris Kupchan’s partitions (1970), as modified by VanWagenen et al. 22 Five grams of crude extract were sequentially treated with water in methanol (1:9), petroleum ether (PESF), dichloromethane (DCMSF), ethyl acetate (EASF), and water (AQSF) to generate 4 distinct fractions. Each fraction was evaporated until completely dried. The yields for the petroleum ether, dichloromethane, ethyl acetate, and water-soluble fractions were 15%, 43%, 24%, and 18%, respectively.
Phytochemical isolation and detection through nuclear magnetic resonance (NMR) technique
Secondary metabolites were isolated from the methanolic crude extract, which was then subsequently purified using chromatographic techniques. The schematic flowchart of complete phytochemical extraction, fractionation, identification, and isolation of phytoconstituents from the C. reticulata is stated in supplementary Figure S2. The size exclusion chromatography used a gradient solvent system, with lipophilic sephadex LH 20 acting as the stationary phase. The ratios of the various solvents utilized as the mobile phase were as follows: n-hexane, dichloromethane (DCM), and methanol in a proportion of 2:5:1; 10% methanol in DCM; 50% methanol in DCM; and 100% methanol (supplementary Table S1). These fractions were thoroughly screened using thin-layer chromatography (TLC) on silica gel-coated metal plates (20 cm × 20 cm) with various solvent solutions. The obtained chromatogram was visually examined under UV illumination at 254 and 366 nm to identify fluorescence quenching. Colored compounds were also recognized after spraying the plate with a 1% vanillin-sulfuric acid solution and heating at 105°C for 2 to 3 minutes. Fractions with similar TLC patterns were combined, yielding 13 sub-fractions. The phytochemicals were then separated and purified from these sub-fractions using repeated preparative thin layer chromatography (PTLC).
A total of 4 compounds (A-D; Figure 1) were isolated and characterized in the study through an extensive chromatographic and spectroscopic methods. Compound D was isolated from sub-fraction F-9 to F-12 (using a solvent mixture of 5% ethyl acetate in toluene), two compounds such as Compounds A (Rf value = 0.68) and B (Rf value = 0.46) were isolated from sub-fraction F-23 to F-28 (using 5% methanol in chloroform). In addition, compound C was isolated from sub-fraction F-30 to F-34 (using 10% methanol in chloroform; Rf value = 0.55; Tables S2 and S3).
Figure 1.
Structures of the identified and characterized compounds through the 1H-NMR and chromatographic technique from the peel extract of Citrus reticulata Blanco.
The total phytochemical isolation procedure and its schematic flowchart can be found in Supplemental Figure S2. Bruker AMX-400 NMR spectrometer was operated to generate 1H-NMR spectra while all spectra were obtained in a deuterated chloroform solvent (CDCl3). The peak values for chemical shift (δ) were standardized to tetramethylsilane (TMS) and the residual signals from the solvent.
GC-MS/MS analysis
The methanol extract of C. reticulata (MECR) was analyzed using the Shimadzu GC/MS-QP2010 ultra, which had an auto-sampler. A 5 MS/HP column (30 m, 0.25 mm, 0.25 m) was used, using pure helium as the mobile phase. The helium’s linear speed was kept at 39 cm/s, with a circulation rate of 1.12 mL/min. The oven temperature raised at a rate of 10°C/min, ranging from 110°C to 280°C, while the needle temperature was set at 250°C. Injection volume of 50 µL was used in split-less mode with a 10:1 ratio. The detector voltage remained at 0.94 kV, while the ambient temperatures for the ion source, MS transfer line, and ion source were held at 200°C and 250°C, respectively. Full-scan mass spectra were obtained at 10 000 µ/s in the (m/z) range of 85 to 500. Peaks and chemical ingredients were identified using a search of the National Institute of Standards and Technology (NIST) database.
Antimicrobial properties
A total of 13 various microbial species, encompassing 5 Gram-positive bacteria (Bacillus subtilis, Bacillus megaterium, Bacillus cereus, Staphylococcus aureus, and Sarcina lutea), 8 Gram-negative bacteria (Salmonella typhi, Escherichia coli, Shigella boydii, Salmonella paratyphi, Shigella dysenteriae, Vibrio mimicus, Vibrio parahemolyticus, and Pseudomonas aeruginosa), along with 5 fungi (Aspergillus niger, Candida albicans, Aspergillus fumigatus, Saccharomyces cerevisiae, and Aspergillus ustus), were procured in their pure culture strains from the Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka, Bangladesh.
The antibacterial efficacy of crude plant extracts and their various fractions was examined using the disk diffusion method. 23 In this process, 6 mm-diameter filter paper disks containing specified quantities of test samples (concentration of 400 µg/disk) were positioned on nutrient agar medium and uniformly inoculated with the respective microorganisms. These disks were then dried and sterilized. Antibiotics permeated the agar gel from a confined source, generating a concentration gradient. Ciprofloxacin standard antibiotic disks served as positive controls, while blank disks were utilized for the negative control. The petri dishes were refrigerated at a low temperature (4°C) for 16 to 24 hours to enhance the diffusion of test samples into adjacent media. 23 Following inversion and additional incubation at 37°C for 24 hours to promote optimal microbial growth around the disks was impeded by the test samples exhibiting antibacterial effects. This hindrance led to the formation of discernible zones of inhibition, and their diameters were measured to assess the antibacterial activity of the test sample.23,24
Anti-inflammatory effects
The crude methanolic extracts and fractions derived from the peel of C. reticulata were evaluated for in vitro anti-inflammatory activity by employing the established techniques as described by previous studies.23,25
Hypotonic solution-induced anti-inflammatory effect
To evaluate the efficiency of MECR and its related extractives in stabilizing human erythrocyte membranes, we followed known techniques from a previous research. 25 Erythrocytes were extracted from a healthy adult weighing 70 kg with fair skin and stored in a sterile container with EDTA for anticoagulation. A buffer solution containing disodium phosphate and its conjugate acid, monosodium phosphate, was created to maintain a pH of 7.4. An isotonic solution (500 mL, 154 mM) was made by dissolving 4.5045 g of NaCl in sterile distilled water, whereas a hypotonic solution (500 mL, 50 mm) was made using 1.4625 g. The erythrocyte suspension was washed 3 times with buffer and isotonic solution before being centrifuged at 3000 rpm for 10 minutes. A stock RBC suspension (0.50 mL), a 10 mM sodium phosphate buffer (4.5 mL), a hypotonic solution (50 mM NaCl), and different quantities of extractives (2.0 mg/mL) or standard acetyl salicyclic acid (ASA) (0.10 mg/mL) were used to create test samples. After a 10-minute incubation at room temperature, the solutions were centrifuged (3000 rpm for 10 minutes) and the supernatants’ absorbance (optical density (OD)) was measured at 540 nm. The proportion of inhibition of hemolysis or stabilization of membrane was estimated by employing the formula stated below:
Where, OD1 and OD2 represent the optical density of hypotonic buffered saline solution (control) and the investigated sample in hypotonic solution, respectively.
Heat-induced anti-inflammatory effect
Two sets of centrifuge tubes were created, each containing 5 mL of isotonic buffer, 1.0 mg/mL of methanolic peel extract, and different fractions. A control tube contained an equal volume of all substances except the test materials. In every well, 30 µL of erythrocyte suspension was added by inverse and gently stirring. One pair of tubes was placed in a 54°C water bath for 20 minutes, while the second set was maintained in a cold bath at 0°C to 5°C. After incubation, the supernatant was centrifuged (1300 rpm for 3 minutes), and the optical density (OD) was measured at 540 nm. The percentage inhibition of hemolysis in the investigations was estimated using the corresponding equation:
Where, OD1, OD2, and OD3 refers to the optical density of the non-heated test sample, the heated test sample, and the heated control sample, respectively.
Thrombolytic properties
In vitro thrombolytic activity was determined using the blood clot lysis technique according to the methods described by Prasad et al. 26 The positive control was a lyophilized streptokinase vial containing 1 500 000 IU while the negative control was distilled water. Healthy volunteers’ venous blood was collected in pre-weighed sterile micro-centrifuge tubes (1 mL/tube), then incubated at 37°C for 45 minutes. Following clot formation, the weight of each clot was determined after full serum extraction. Each micro-centrifuge tube with a pre-weighted clot received a 100 μL aqueous solution from different partitions, in addition to the crude extract. The tubes were incubated for 90 minutes at 37°C and kept in a close observation for clot lysis. The percentage of clot lysis was enumerated by using the following the formula:
Animal study design
The Animal Division of the International Centre for Diarrheal Diseases and Research in Bangladesh (ICDDRB) provided Swiss albino adult mice aged 4 to 5 weeks and weighed 25 to 30 g. Prior to the animal research, these mice were kept in polypropylene cages under conventional climatic settings such as a 55% relative humidity level, 25°C ambient temperature, and a 12-hour light-dark cycle. Prior to the beginning of the experiments, the animals were allotted a week to acclimate to their new environment. During this time, the animals were fed a standard laboratory diet and had unlimited access to water. Their care and handling followed the guidelines set by the Swiss Academy of Medical Sciences (SAMS) and the Swiss Academy of Sciences. The study also complied with the latest Animal Research: Reporting of In Vivo Experiments (ARRIVE) guidelines. 27 The protocols for animal handling and procedures involved in the animal studies were thoroughly reviewed and approved by the Committee on Ethical Compliance in Research at the State University of Bangladesh, Dhaka, Bangladesh (approval number: 2022-01-23/SUB/A-ERC/006).
The study included 3 different doses of the methanol extract of C. reticulata (MECR) at concentrations of 100, 200, and 400 mg/kg body weight, designated as ME-100, ME-200, and ME-400, respectively. The animal subjects were divided into 5 groups, each consisting of 4 mice (n = 4). This sample size was estimated by using the “resource equation” method which was clearly explained in the previous studies.28-30 The groupings were organized as follows:
Group I (n = 4): Normal saline water [0.9% (w/v) NaCl solution BP] as negative control
Group II (n = 4): Standard drug as positive control
Group III (n = 4): 100 mg/kg body weight MECR (ME-100)
Group IV (n = 4): 200 mg/kg body weight MECR (ME-200)
Group V (n = 4): 400 mg/kg body weight MECR (ME-400)
Central analgesic activity
The analgesic effects of MECR were assessed using a heat-based approach and the tail immersion experiment, as described by previous investigation. 31 The positive control group received morphine (15 mg/mL) diluted with saline water to form the standard sample (subcutaneous, 2 mg/kg), as reported in earlier report. 32 The mice received the test drugs orally using a feeding syringe. The experiment involves submerging a mouse’s tail in water heated to 55°C. The Pain Response Time (PRT) or latency duration, which indicates how long it takes for each mouse to move its tail in hot water, was assessed at 0, 30, 60, and 90 minutes after the test samples were administered.
Peripheral analgesic activity
The peripheral analgesic effects of MECR were assessed by applying the acetic acid-induced writhing technique in mice model. 33 At the 0 hour of the experiment, the control group was administered normal saline, while the positive control group received diclofenac sodium (50 mg/kg) as standard. Subsequently, the 3 experimental groups were administered their respective doses of the designated crude extracts. After 60 minutes, all groups received an intraperitoneal injection of 1% glacial acetic acid (0.1 mL; dissolved in normal saline) to induce visceral pain. After 5 minutes of acetic acid injection, the writhing times were counted for 15 minutes. The formula for calculating the percentage of writhing inhibition was as follows:
Where, N = average amount of stomach writhing for every group.
Anti-diarrheal activity
The anti-diarrheal activity of MECR was measured by employing a castor oil-induced diarrhea model in mice, outlined in earlier studies.34,35 According to the experimental design, the positive control group received oral administration of loperamide (50 mg/kg bw). The weighted extracts, after being suspended with 1% Tween-80, were unidirectional handled and then applied to the test groups. The normal control group received only the normal saline water. To induce diarrhea, 1 mL of pure castor oil was supplied to each mouse after 1 hour. Each mouse was kept in a separate cage with blotting paper on the floor. For each test group, the floor lining of individual mouse cage was changed every hour. The number of diarrheal stools generated by each mouse was monitored hourly for up to 4 hours after castor oil delivery. The count of diarrheal feces produced by the mice was recorded at 1-hour intervals over a 4-hour observation period. The percentage inhibition of diarrheal episodes was subsequently calculated using the following equation.
Where, D = Average diarrheal episodes in every group.
Molecular docking
The isolated and purified 4 compounds (A-D) and 8 more compounds (C-79, C-83, C-84, C-86, C-87, C-88, C-90, and C-91) from the obtained compound list characterized by GC-MS/MS technique were chosen for a computational modeling study to predict their receptor binding profiles against the relevant target proteins to support the biological activities of the studied extracts and fractions. Following established procedures from several previous studies, molecular docking of these phytocompounds was conducted using BIOVIA Discovery Studio version 4.5, PyMOL 2.3, and PyRx software packages.36-39
Ligand preparation
A total of 12 phytomolecules were selected as ligands in docking investigation. These chemicals were identified as tangeretin (PubChem CID: 68077), nobiletin (PubChem CID: 72344), limonin (PubChem CID: 179651), β-sitesterol (PubChem CID: 481107734), salvigenin (PubChem CID: 161271), pentamethoxyflavone (PubChem CID: 35028119), 6-demoethoxytangeretin (PubChem CID 629964), stigmasterol (PubChem CID: 5280794), pebrellin (PubChem CID: 632255), artemetin (PubChem CID: 5320351), senensetin (PubChem CID: 145659), dimethylnobiletin (PubChem CID: 358832) from PubChem database (retrieved on May 27, 2024). The standard drugs thrombolytic agent warfarin (PubChem CID: 54678486), cyclooxygenase inhibitor diclofenac (PubChem CID: 3033), antioxidant BHT (PubChem CID: 31404), antidiarrheal molecule loperamide (PubChem CID: 3955), antibacterial agent ciprofloxacin (PubChem CID: 2764) and μ-opioid receptor inhibitor morphine (PubChem CID: 5288826) were downloaded in SDF format. Using the PubChem CIDs of the ligands, a ligand library in PDB format was created following the ligands’ sequential loading into Discovery Studio version 4.5. Using the PM6 semi-empirical approach, the accuracy of molecular interactions for every ligand was enhanced. 40
Protein preparation
To predict the potential antibacterial, thrombolytic, anti-inflammatory, analgesic, and antidiarrheal activities of the ligands, a computational docking approach was employed. Specifically, dihydrofolate reductase (DHFR; PDB ID: 4M6J), tissue plasminogen activator (PDB ID: 1A5H), cyclooxygenase 2 (COX-2; PDB ID: 1CX2), μ-opioid receptor (MOR; PDB ID: 5C1M), and kappa opioid receptor (PDB ID: 6VI4) with their corresponding active sites were selected to assess antibacterial, thrombolytic, peripheral and central analgesic, and antidiarrheal effects, respectively, based on the previously published literature.23,41-43 In order to retrieve the target proteins, protein data bank was searched. After then, the three-dimensional (3D) crystal structures were downloaded and saved in PDB format (retrieved on May 27, 2024). Then the downloaded proteins were processed by using Discovery Studio (version 4.5) and Swiss PDB viewer for cleaning and optimizing purposes, respectively. 44
Protein and ligand interaction
The PyRx software was used for molecular interaction purpose. The cleaned and optimized proteins were imported in the software and selected as macromolecules. The active amino acids were assigned, which were pre-selected according to the literature data and summarized in the Supplemental Table S4. The 3D conformers of the ligands in SDF format were uploaded to the PyRx program and minimized energy using “uff force field.” Then, all ligands were transformed into pdbqt format to determine the best hit. Then, as shown in Supplemental Table S4, the grid box was created, with the proteins’ active binding sites centered inside. The other docking process parameters were kept at their factory default values. The final docked macromolecules and ligands were exported as pdbqt format output files, and the outcomes of the docking analysis were then investigated. PyMOL software was used to combine the stored ligands’ output files and the macromolecule’s pdbqt file in PDB format for further visualization. Discovery Studio Visualizer (version 4.5) was used for visualization and 2D figure development.
Pharmacokinetics and toxicity analysis
The pharmacokinetic parameters related to absorption, distribution, metabolism, excretion (ADME), and toxicity were predicted by employing the well-known online server pkCSM (https://biosig.lab.uq.edu.au/pkcsm/prediction; accessed on May 10, 2024).
Physicochemical and drug-likeliness analysis
All the docked 12 compounds were assessed for their potential drug-like properties according to Lipinski’s rule of 5 (molar refractivity between 40 and 130, molecular weight ⩽ 500; H-bond acceptors < 10; H-bond donors < 5; and LogP ⩽ 5) using the Swiss ADME online tool (www.swissadme.ch; accessed on May 27, 2024).
Statistical analysis
The data processing and findings interpretation were done in accordance with the established protocols explained by Assel et al. 45 The in vitro experimental analysis, graphs and data processing were carried out using Microsoft Excel (version 10.0). Additionally, the treated groups were compared to the control (vehicle) group in order to do the statistical analysis for the in vivo experiments. The mean values, along with their corresponding standard errors of mean (SEM), were represented as mean ± SEM to indicate the outcomes of the in vivo assessments. The results from in vivo testing were examined and analyzed through GraphPad Prism software (version 8.0.2). A one-way ANOVA was conducted on all variables, followed by Student’s t-test. P-values less than .05 were considered statistically significant. The heatmap representing the docking score analysis and the graphs associated with the in vivo studies were generated utilizing GraphPad Prism software.
Results
Phytochemical studies
Four compounds were identified, purified, and characterized during the phytochemical isolation process through various chromatographic techniques and 1H-NMR spectroscopic analysis. These compounds were determined to be tangeretin (compound A), nobiletin (compound B), limonin (compound C), and β-sitosterol (compound D). Additionally, 91 compounds were detected and characterized using GC-MS/MS analysis. Further detailed information is provided in the following sections.
Compounds characterization by NMR technique
Compound A: According to the 1H-NMR spectrum and the summarized data in Table 1, the presence of 5 singlets at δ 3.88, 3.94, 3.96, 4.01, and 4.09 ppm indicated the existence of 15 protons of 5 methoxy groups. Additionally, a singlet at δ 6.60 ppm suggested that compound A have an olefinic proton, a key feature of flavone-type molecules. The presence of 2 doublets at δ 7.15 and 7.85 ppm, with a coupling constant of nearly 8.0 Hz, indicates a 1,4-substituted benzene ring within compound A. Moreover, matching this 1H-NMR spectrum with the existing literature suggested the identification compound A as tangeretin. 46
Compound B: The double doublets at δ 7.56 ppm, doublet at δ 6.88 ppm, and another doublet at δ 7.40 ppm denoted the presence of 1, 3, 4 tri-substituted benzene ring of the compound B. The appearance of 6 singlets at δ 4.9, 4.03, 4.01, 3.97, 3.95, and 3.94 ppm showed the presence of 6 difference methoxy groups within the compound B. In addition, the singlet at δ 6.61 ppm represents 1 olefinic proton. This 1H-NMR spectrum values indicated this molecule might be a polymethoxyflavone containing total 6 methoxy groups. After that, matching the proton NMR values with the previously published literature suggested this molecule as nobiletin. 47
Compound C: According to the 1H-NMR spectrum and the Table 1, the presence of a multiplet at δ 7.40 and δ 6.49 indicated a furan ring within this molecule. Moreover, 3 different singlets at δ 1.08 (3H), δ 1.17 (6H), and δ 1.28 (3H) suggested the existence of 4 branched methyl groups within the molecule. A multiplet at δ 3.98 showed that C-1 was attached to a highly electronegative group. Furthermore, a singlet at δ 4.04 demonstrated that H-15 was attached to an oxygenated carbon moiety. Another singlet at δ 5.46 showed that H-17 was linked to the furan group of this molecule. Two doublets at δ 4.75 (J = 12.8 Hz) and δ 4.45 (J = 13.2 Hz) indicated that both protons of H-19 were connected to an oxygenated carbon moiety. Additionally, various broad doublet (br. d) peaks within the δ 2.21 to δ 2.97 range indicated several protons in the molecule’s skeleton, such as H-2a, H-2b, H-6a, H-6b, H-5, and H-9. This data aligns with previously published proton NMR data in the literature, supporting the conclusion that the compound is limonin. 48
Compound D: The presence of multiplet at δ 5.34 ppm suggested the presence of steroidal olefinic proton at H-6 position. While multiplet at δ 3.58 ppm recommended the H-3 is attached with the highly electro negative moiety. Furthermore, the presence of 2 singlets, 3 doublets, and 1 triplet peaks ranging from 0.67 to 0.99 ppm demonstrated the presence of 6 steroidal methyl groups within the compound D. In addition, by matching the 1H-NMR spectrum of the compound D with the literature 1H-NMR values, it was confirmed as β-sitosterol. 49
Table 1.
The 1H-NMR (400 MHz, CD3OD) and the compared literature spectroscopic data of the isolated compounds A–D from C. reticulata.
Compounds | Mol. formula | Mol. wt. (g/mol) | Positions | 1 H-NMR values δ in ppm (400 MHz, CDCl3) | Reference values | Ref. |
---|---|---|---|---|---|---|
Tangeretin (A) | C20H20O7 | 372.4 | H-3 | 6.60 (1H, s) | 6.69 (1H, s) | Morimoto et al 46 |
H-2’/H-6’ | 7.85 (2H, d, J = 8 Hz) | 7.96 (2H, d, J = 9.0 Hz) | ||||
H-3’/H-5’ | 7.15 (2H, d, J = 8.4 Hz) | 7.10 (2H, d, J = 9.0 Hz) | ||||
5 Methoxy groups | (3.88, 3.94, 3.96, 4.01, 4.09 (Total five s, 15H) | 3.97, 4.03, 4.06, 4.10, 4.18 (Total five s, 15H) | ||||
Nobiletin (B) | C21H22O8 | 402.4 | H-3 | 6.61 (1H, s) | 6.63 (1H, s) | Santos et al 47 |
H-2’ | 7.56 (1H, dd, J = 8.4, 2.0 Hz) | 7.47 (1H, dd, J = 8.5, 2.1 Hz) | ||||
H-3’ | 6.98 (1H, d, J = 8.8 Hz) | 6.99 (1H, d, J = 8.5 Hz) | ||||
H-6’ | 7.40 (1H, d, J = 2 Hz) | 7.42 (1H, d, J = 2.1 Hz) | ||||
6 Methoxy groups | 4.9, 4.03, 4.01, 3.97, 3.95, 3.94, (Total six s, 18H) | 4.10, 3.95, 3.95, 4.03, 3.98, 3.96 (Total six s, 18H) | ||||
Limonin (C) | C26H30O8 | 470.5 | H-1 | 3.98 (1H, m) | 4.03 (1H, m) | Breksa et al 48 |
H-2a | 2.66 (1H, br. d, J = 16.8 Hz) | 2.67 (1H, dd, J = 16.8, 2.0 Hz) | ||||
H-2b | 2.97 (1H, br. d, J = 16.8 Hz) | 2.98 (1H, dd, J = 16.8, 4.0 Hz) | ||||
H-3 | - | - | ||||
H-4 | - | - | ||||
H-5 | 2.21 (1H, br. d, J = 15.2 Hz) | 2.22 (1H, dd, J = 15.8, 3.4 Hz) | ||||
H-6a | 2.42 (1H, br. d, J = 14.8 Hz) | 2.46 (1H, dd, J = 14.4, 3.2 Hz) | ||||
H-6b | 2.86 (1H, br. d, J = 14.8 Hz) | 2.85 (1H, dd, J = 15.8, 14.6 Hz) | ||||
H-7 | - | - | ||||
H-8 | - | - | ||||
H-9 | 2.53 (1H, br. d, J = 11.6 Hz) | 2.55 (1H, dd, J = 12.2, 3.0 Hz) | ||||
H-11 | 1.76-1.88 (2H, m) | 1.72-1.95 (2H, m) | ||||
H-12 | 1.36-1.46 (2H, m) | 1.46-1.58 (2H, m) | ||||
H-14 | - | - | ||||
H-15 | 4.04 (1H, s) | 4.05 (1H, s) | ||||
H-16 | - | - | ||||
H-17 | 5.46 (1H, s) | 5.47 (1H, s) | ||||
H-18, H-25b | 1.17 (6H, s) | 1.18 (6H, s) | ||||
H-19a | 4.75 (1H, d, J = 12.8 Hz) | 4.76 (1H, d, J = 13.0 Hz) | ||||
H-19b | 4.45 (1H, d, J = 13.2 Hz) | 4.46 (1H, d, J = 13.0 Hz) | ||||
H-20 | - | - | ||||
H-21, H-23 | 7.39-7.40 (2H, m) | 7.40-7.41 (2H, m) | ||||
H-22 | 6.49 (1H, m) | 6.34 (1H, m) | ||||
H-24 | 1.08 (3H, s) | 1.08 (3H, s) | ||||
H-25a | 1.28 (3H, s) | 1.29 (3H, s) | ||||
ß-sitosterol (D) | C29H50O | 414.7 | H-3 | 3.58 (1H, m) | 3.53 (1H, tdd) | Majid Shah et al 49 |
H-6 | 5.34 (1H, m) | 5.36 (1H, t, J = 6.4 Hz) | ||||
H-18 | 0.67 (3H, s) | 0.68 (3H, s) | ||||
H-19 | 0.99 (3H, s) | 1.01 (3H, s) | ||||
H-21 | 0.91 (3H, d, J = 6.8 Hz) | 0.93 (3H, d, J = 6.5 Hz) | ||||
H-26 | 0.80 (3H, d, J = 8.4 Hz) | 0.81 (3H, d, J = 6.4 Hz) | ||||
H-27 | 0.84 (3H, d, J = 7.6 Hz) | 0.83 (3H, d, J = 6.4 Hz) | ||||
H-29 | 0.87 (3H, t, J = 6.0 Hz) | 0.84 (3H, t, J = 7.2 Hz) |
Compounds detection by GC-MS analysis
A total of 91 phytoconstituents were predominantly detected in the MECR through GC-MS technique. The spectrum obtained from the GC-MS/MS analysis is available in Figure 2. Among the identified over 90 compounds, a significant number of compounds were polymethoxyflavones. Among these, the following were quantified: nobiletin (29.04%), tangeretin (15.55%), artemetin (8.1%), 6-demethoxytangeretin (1.28%), sinensetin (0.95%), demethylnobiletin (0.14%), pebrellin (0.10%), and salvigenin (0.04%). Furthermore, some notable compounds were also detected as dihydrocarvyl acetate (4.07%), Methyl alpha D-galactopyranoside (3.37%), 3’,5’-demethoxyacetaophenone (2.55%), D-limonene (2.48%), and stigmasterol (0.35%; Table 2). Yet, some phytoconstituents were discovered as least amount such as ß-Elemene, Megastigmatrienone, Octadecanamide, Fenoterol, Salvigenin, and Bicyclon onan-2-one, 8-isopropylidene- (0.04%). Various classes of phytoconstituents such as fatty derivatives (Methyl 3-hydroxydodeca-noate, 2,6,9,11-Dodecatetraenal, 2,6,10-trimethyl-, (E,E,E)-, and Hexane,1- (isopropylidene cyclopropyl)-,) steroids (Stigmasterol), flavonoids (Demethylnobiletin, sinensetin, Pebrellin, and artemetin), and aliphatic cyclic (1,2- Cyclohexanedione, Trimethylene borate, delta-Elemene, and Furaneol) compounds were also identified in this analysis (Table 2).
Figure 2.
GC-MS spectrum of the crude extract obtained from Citrus reticulata Blanco.
Table 2.
The identified and characterized phytoconstituents obtained from GC-MS/MS analysis are presented with details including the compound names, retention time (RT), percentage of area, molecular weight (g/mol), molecular formula, PubChem CID, and their chemical structures drawn using ChemDraw. Additionally, the pharmacological activities of these compounds were extracted from the literature, with the corresponding references cited in the final column.
SL No. | Compound | RT (min) | Area % | Mol. weight (g/mol) | Molecular formula | PubChem CID | Chemical Structure | Pharmacological activity | Ref |
---|---|---|---|---|---|---|---|---|---|
1. | 2,4-Dihydroxy-2,5-Dimethyl-3(2H)-furan-3-one | 5.36 | 0.1 | 144.12 | C6H8O4 | 538 757 |
![]() |
Antioxidant activity | Chukwu et al 50 |
2. | 1,2-Cyclohexanedione | 5.62 | 0.45 | 112.13 | C6H8O2 | 13 006 |
![]() |
N/A | |
3. | D-Limonene | 5.82 | 2.48 | 136.23 | C10 H16 | 440 917 |
![]() |
Antioxidant, antidiabetic, anticancer | Anandakumar et al 51 |
4. | Dihydrocarvyl acetate | 5.935 | 4.07 | 196.29 | C12 H20 O2 | 30 248 |
![]() |
Antimicrobial, antioxidant, insecticidal, antitumor, anti-inflammatory and antidiabetic | Zhang et al 52 |
5. | Emylcamate | 6.145 | 0.47 | 145.2 | C7H15NO2 | 6526 |
![]() |
Antioxidant and anticancer | Onygeme-Okerenta et al 53 |
6. | Gamma-Terpinene | 6.264 | 0.6 | 136.23 | C10 H16 | 7461 |
![]() |
Antimicrobial, antioxidant, hypoglycemic, hypolipidemic, anxiolytic, analgesic, anti-inflammatory, anti-convulsant and anticancer | Laribi et al 54 |
7. | Furaneol | 6.363 | 0.76 | 128.13 | C6H8O3 | 19 309 |
![]() |
Antimicrobial activity | Sung et al 55 |
8. | Thymine | 6.565 | 0.3 | 126.11 | C5H6N2O2 | 1135 |
![]() |
Antibacterial activity | Liu et al 56 |
9. | Phenol, 2-methoxy- | 6.616 | 0.2 | 124.14 | C7H8O2 | 460 |
![]() |
Antioxidant | Fujisawa et al 57 |
10. | Linalool | 6.728 | 0.23 | 154.25 | C10H18O | 6549 |
![]() |
Antimicrobial, antioxidant, hypoglycemic, hypolipidemic, anxiolytic, analgesic, anti-inflammatory, anti-convulsant and anti-cancer. | Laribi et al 54 |
11. | Chrysantenyl 2-methuylbutanoate | 7.05 | 0.06 | 236.35 | C15H24O2 | 91 693 263 |
![]() |
Antioxidant and anticancer | Ambati et al 58 |
12. | 2-Cyclohexen- 1-ol,1-methyl-4-(1-methylethenyl)-,trans- | 7.21 | 0.1 | 152.23 | C10H16O | 155 626 |
![]() |
Antibacterial and antifungal activity | Mehani et al 59 |
13. | Ethanamine,N-ethyl-N-nitroso- | 7.275 | 0.16 | 102.14 | C4H10N2O | 5921 |
![]() |
N/A | |
14. | 4H-Pyran-4- one,2,3-dihydro-3,5-dihydroxy-6-methyl- | 7.403 | 0.55 | 144.12 | C6H8O4 | 119 838 |
![]() |
Antioxidant activity | Chen et al 60 |
15. | Trimethylene borate | 7.47 | 0.5 | 243.9 | C9H18B2O6 | 88 725 |
![]() |
Antimicrobial activity | Zamakshshari et al 61 |
16. | Ribitol | 7.615 | 0.15 | 152.15 | C5H12O5 | 6912 |
![]() |
Anti-diabetic | Montague and Taylor 62 |
17. | Terpinen-4-ol | 7.709 | 0.2 | 154.25 | C10H18O | 11 230 |
![]() |
Antimicrobial activity | Mondello et al 63 |
18. | Bicyclo[2.2.2]o ctane-1.4-diol | 7.779 | 0.25 | 142.2 | C8H14O2 | 559 236 |
![]() |
N/A | |
19. | alpha-Terpineol | 7.859 | 0.35 | 154.25 | C10H18O | 17 100 |
![]() |
Antioxidant, anticancer, anticonvulsant, antiulcer, antihypertensive, anti-nociceptive | Khaleel et al 64 |
20. | 8-Azabicyclo[3.2.0.1]oct-6-en-3-one,8-methyl- | 7.915 | 0.36 | 137.18 | C8H11NO | 565 060 |
![]() |
N/A | |
21. | 5-Methylbenzofuran-2,3-dione | 8.048 | 0.23 | 162.14 | C9H6O3 | 56 974 146 |
![]() |
Anti-inflammatory, antimicrobial, antifungal, antihyperglycemic, analgesic, antiparasitic, and antitumor activities | Khodarahmi et al 65 |
22. | Hexane,1-(isopropylidene cyclopropyl)- | 8.1 | 0.09 | 166.3 | C12H22 | 32 479 |
![]() |
Antioxidant activity | Erwin et al 66 |
23. | 5-(Hydroxymethyl)furfural | 8.208 | 0.84 | 126.11 | C6H6O3 | 237 332 |
![]() |
Antioxidant antitumor anti-inflammatory | Zhao et al 67 |
24. | (-)-Carvone | 8.376 | 0.11 | 150.22 | C10H14O | 439 570 |
![]() |
Antidiabetic, anti-inflammatory, anticancer, neurological, antimicrobial, antiparasitic, antiarthritic, anticonvulsant, and immunomodulatory effects. | Bouyahya et al 68 |
25. | 1,2-Benzenediol, 3-methyl-6-(4-morpholinylmethyl)- | 8.52 | 0.26 | 223.27 | C12H17 NO3 | 11 333 553 |
![]() |
Antimicrobial activity | Naksing et al 69 |
26. | 1-Acetyl-2-(2’-oxo-propyl)-cyclopentane | 8.651 | 0.12 | 168.23 | C10H16 O2 | 538 127 |
![]() |
N/A | |
27. | ß-Cyclocitral | 8.724 | 0.21 | 166.22 | C10H14O2 | 642 490 |
![]() |
Antifungal, anticancer, antimicrobial, and antiplasmodial | Faizan et al 70 |
28. | Thymol | 8.785 | 0.09 | 150.22 | C10H14O | 6989 |
![]() |
Antioxidant, anti-inflammatory,, antiseptic, and antibacterial and antifungal | Marchese et al 71 |
29. | Santolina epoxide | 8.849 | 0.13 | 152.23 | C10H16O | 565 371 |
![]() |
N/A | |
30. | Allylphenyl sulfide | 8.895 | 0.17 | 150.24 | C9H10S | 79 180 |
![]() |
N/A | |
31. | 2-Methoxy-4-vinylphenol | 9.048 | 1.1 | 150.17 | C9H10O2 | 332 |
![]() |
Anti-proliferative anti-inflammatory anticancer activity | Jeong and Jeong 72 |
32. | Phytol | 9.25 | 0.22 | 296.5 | C20H40O | 5 280 435 |
![]() |
Anxiolytic, cytotoxic, antioxidant, anti-inflammatory, antimicrobial activity. | Islam et al 73 |
33. | delta-Elemene | 9.3 | 0.14 | 204.35 | C15H24 | 89 316 |
![]() |
N/A | |
34. | Decanal dimethyl acetal | 9.557 | 0.19 | 202.33 | C12H26O2 | 24 513 |
![]() |
N/A | |
35. | 3-(2-piperidinyl) sulfate | 9.735 | 0.06 | 260.31 | C10H16 N2N2O4S | 205 585 |
![]() |
N/A | |
36. | ß-Elemene | 9.843 | 0.04 | 204.35 | C15H24 | 6 918 391 |
![]() |
Anti-inflammatory and antitumor | Shamsizadeh et al 74 |
37. | 3-Methoxy-4-{3-oxo-3-(pyrrolidin-1-yl)propoxy}ben zaldehyde | 9.93 | 0.09 | 277.31 | C15 H19NO4 | 91 721 113 |
![]() |
N/A | |
38. | 3-Cyclopentylpropionamide, N-methallyl- | 10.06 | 0.10 | 195.3 | C12H21NO | 91 695 995 |
![]() |
N/A | |
39. | Gamma-elemene | 10.219 | 0.12 | 204.35 | C15H24 | 6 432 312 |
![]() |
N/A | |
40. | Alpha-Thujone | 10.355 | 0.17 | 154.25 | C10H18O | 12 304 610 |
![]() |
5-HT3 receptor antagonist | Tamer et al 75 |
41. | Humulene | 10.54 | 0.32 | 204.35 | C15H24 | 5 281 520 |
![]() |
Antioxidant, anticancer | Russo and Marcu 76 |
42. | (2E)-3,7-dimethylocta-2,7-diene-1,6-diol | 10.761 | 0.82 | 170.25 | C10H18O2 | 12 536 844 |
![]() |
Antimicrobial activity | |
43. | Alpha-Farnesene | 10.80S | 0.24 | 204.35 | C15H24 | 5 281 516 |
![]() |
Anti-viral | Habibah et al 77 |
44. | Cis-5-Dodecenoicacid,methylester | 10.915 | 0.51 | 212.33 | C13H24O2 | 14 524 604 |
![]() |
N/A | |
45. | Acetic acid, 2-propyltetrahydropyran-3-yl ester | 10.96 | 0.62 | 186.25 | C10H18O3 | 537 808 |
![]() |
N/A | |
46. | 4,5-Octanediol, 2,7-dimethyl- | 11.14 | 1.21 | 174.28 | C10H22O2 | 549 801 |
![]() |
N/A | |
47. | Dodecanoic acid | 11.29 | 0.92 | 200.32 | C12H24O2 | 3893 |
![]() |
N/A | |
48. | 3’,5’-Dimethoxyaceto phenone | 11.376 | 2.55 | 180.2 | C10H12O3 | 95 997 |
![]() |
N/A | |
49. | Megastigmatrienone | 12.089 | 0.04 | 190.28 | C13H18O | 5 375 190 |
![]() |
N/A | |
50. | Isovaleric acid, eicosyl ester | 12.48 | 0.11 | 382.7 | C25H50O2 | 85 768 020 |
![]() |
N/A | |
51. | Methyl alpha D-Galactopyranoside | 12.901 | 3.37 | 194.18 | C7H14O6 | 76 935 |
![]() |
N/A | |
52. | Methyl 3-hydroxydodecanoate | 13.18 | 1.36 | 230.34 | C13H26O3 | 155 752 |
![]() |
N/A | |
53. | 2,6,9,11-Dodecatetraenal, 2,6,10-trimethyl-, (E,E,E)- | 13.424 | 1.39 | 218.33 | C15H22O | 5 281 534 |
![]() |
N/A | |
54. | 2,3-Anhydro-d-mannosan | 14.336 | 2.25 | 144.12 | C6H8O4 | 88 413 508 |
![]() |
N/A | |
55. | Ethyl 2,2-dimethyl-5-oxoheptanoate | 15.59 | 0.16 | 200.27 | C11H20O3 | 545 903 |
![]() |
N/A | |
56. | Penicillin Roquefort Toxin | 15.845 | 0.06 | 320.3 | C17H20O6 | 440 907 |
![]() |
Antimicrobial, genotoxic and cytotoxic | Dubey et al 78 |
57. | dl-Citrulline | 16.11 | 0.14 | 175.19 | C6H13N3O3 | 833 |
![]() |
N/A | |
58. | n-Hexadecanoic acid | 16.295 | 0.79 | 256.42 | C16H32O2 | 985 |
![]() |
Antioxidant, anti-inflammatory, ,antibacterial, antifungal | Mazumder et al 79 |
59. | Scoparone | 16.63 | 0.24 | 206.19 | C11H10O4 | 8417 |
![]() |
N/A | |
60. | trans-Sinapyl alcohol | 16.845 | 0.11 | 210.23 | C11H14O4 | 5 280 507 |
![]() |
Anti-inflammatory | Choi et al 80 |
61. | Trehalose | 16.987 | 0.26 | 342.3 | C12H22O11 | 7427 |
![]() |
Anti-inflammatory, anti-cancer | Frapporti et al 81 |
62. | Methyl 10- trans,12-cis- octadecadienoate | 18.334 | 0.26 | 294.5 | C19H34O2 | 5 471 014 |
![]() |
N/A | |
63. | 91 215-O ctadecatrienoicacid, methylester, (Z,Z,Z)- | 18.434 | 0.1 | 292.5 | C19H32O2 | 9316 |
![]() |
Anti-cancer | Al-Otaibi et al 82 |
64. | Linoleic acid trimethylsilyl ester | 19.05 | 0.23 | 352.6 | C21H40O2Si | 5 352 430 |
![]() |
Antimicrobial activity, antibacterial activity | Rossellia et al 83 |
65. | 91 215-O ctadecatrienoic acid,(Z,Z,Z)- | 19.141 | 0.43 | 278.4 | C18H30O2 | 5 280 934 |
![]() |
Antibacterial, anti-inflammatory | Al-Otaibi et al 82 |
66. | E,E,Z-1,3,12-Nonadecatriene-5,14-diol | 19.43 | 0.12 | 294.5 | C19H34O2 | 5 364 768 |
![]() |
N/A | |
67. | Benzyl Beta-D-Glucopyranoside | 21.149 | 0.08 | 270.28 | C13H18O6 | 188 977 |
![]() |
N/A | |
68. | (3R,6R)-3-Hydroperoxy-3-methyl-6-(prop-1-en-2-yl)cyclohex-1-ene | 23.101 | 0.11 | 168.23 | C10H16O2 | 6 431 185 |
![]() |
N/A | |
69. | Hexadecanoic acid 2-hydroxy-1-(hydroxymethyl) ethyl ester | 25.119 | 0.48 | 330.5 | C19H38O4 | 129 853 056 |
![]() |
Antioxidant, anti-inflammatory and anthelmintic activities | Ali et al 84 |
70. | Bis(2-ethylhexyl) phthalate | 25.44 | 0.05 | 390.6 | C24H38O4 | 8343 |
![]() |
Antimicrobial activity | Habib and Karim 85 |
71. | Ethyl Cholate | 27.73 | 0.08 | 436.6 | C26H44O5 | 6 452 096 |
![]() |
N/A | |
72. | Glyceryl monolinoleate | 27.85 | 0.27 | 354.5 | C21H38O4 | 5 283 469 |
![]() |
N/A | |
73. | cis,cis,cis- 7,10,13-Hexadecatrienal | 28 | 0.14 | 234.38 | C16H26O | 5 367 366 |
![]() |
N/A | |
74. | cis-11-Eicosenamide | 29.228 | 2.64 | 309.5 | C20H39NO | 5 365 374 |
![]() |
N/A | |
75. | Octadecanamide | 29.539 | 0.04 | 283.5 | C18H37NO | 31 292 |
![]() |
N/A | |
76. | Squalene | 29.663 | 0.08 | 410.7 | C30H50 | 638 072 |
![]() |
Anticancer, antioxidant, | Kim and Karadeniz 86 |
77. | Fenoterol | 30.481 | 0.04 | 303.35 | C17H21NO4 | 3343 |
![]() |
Antibacterial | Soliman et al 87 |
78. | Gamma-Tocopherol | 32.979 | 0.1 | 416.7 | C28H48O2 | 10 740 654 |
![]() |
Antioxidant | Sánchez-Illana et al 88 |
79. | Salvigenin | 33.128 | 0.04 | 328.3 | C18H16O6 | 161 271 |
![]() |
Anti-inflammatory, anti-cancer | Mansourabadi et al 89 |
80. | Bicyclo[4.3.0]n onan-2-one,8-isopropylidene- | 33.469 | 0.04 | 178.27 | C12H18O | 596 085 |
![]() |
N/A | |
81. | Vitamin E | 34.099 | 0.28 | 430.7 | C29H50O | 14 985 |
![]() |
Antioxidant anti-inflammatory | Sánchez-Illana et al 88 |
82. | Methyleriostoate | 34.41.8 | 0.95 | 358.4 | C21H26O5 | 631 241 |
![]() |
N/A | |
83. | Tangeretin (Compound A) | 34.934 | 15.55 | 372.4 | C20H20O7 | 35 028 119 |
![]() |
Anti-inflammatory, anti-proliferative and neuroprotective | Hung et al 90 |
84. | 6-Demethoxytangeretin | 35.283 | 1.28 | 342.3 | C19H18O6 | 629 964 |
![]() |
Anti-inflammatory, anti-cancer | Li et al 91 |
85. | 4-tert-Butylcatechol, bis(trifluoroacetate) | 35.871 | 0.38 | 358.23 | C14H12F6O4 | 91 743 313 |
![]() |
Antioxidant | García-Moreno et al 92 |
86. | Stigmasterol | 36.272 | 0.35 | 412.7 | C29H48O | 5 280 794 |
![]() |
Antimicrobial, anticancer, diuretic, anti-inflammatory, antioxidant | Hossain et al 93 |
87. | Pebrellin | 37.167 | 0.1 | 374.3 | C19H18O8 | 632 255 |
![]() |
Antibacterial | Mamadalieva et al 94 |
88. | Artemetin | 37.546 | 8.1 | 388.4 | C20H20O8 | 5 320 351 |
![]() |
Anti-edematogenic activity | Bayeux et al 95 |
89. | Nobiletin (Compound B) | 38.359 | 29.04 | 402.4 | C21H22O8 | 72 344 |
![]() |
Antioxidant, anticancer, anti-inflammatory, anti-tumor | Li et al 91 |
90. | Sinensetin | 38.661 | 0.95 | 372.4 | C20H20O7 | 145 659 |
![]() |
Anticancer, anti-inflammatory, antioxidant, antimicrobial, anti-obesity, anti-dementia | Han Jie et al 96 |
91. | Demethylnobiletin | 39.594 | 0.14 | 388.4 | C20H20O8 | 358 832 |
![]() |
anti-inflammatory activity | Guo et al 97 |
Antimicrobial properties
Among the fractions DCMSF showed broad spectrum antimicrobial properties. From gram positive bacteria, DCMSF showed 8 mm zone of inhibition at 3 different bacteria such as B. sereus, B. subtilis, and S. aureus while the DCMSF exerted 10 mm zone of inhibition against 3 different gram-negative bacteria such as S. typhi, E. coli, and V. parahemolyticus at the antimicrobial testing assay (Table S4). DCMSF also showed potency at antifungal Sarcina lutea activity, and most effective result was found against C. albicans (20 mm zone of inhibition). However, MECE exhibits the poorest zone of inhibition only active against Sarcina lutea gram negative bacteria (15 mm; Table S4).
Anti-inflammatory properties
Hypotonic solution-induced hemolysis
In comparison with the standard acetyl salicylic acid at a strength of 0.10 mg/mL (61.90% inhibition against erythrocyte membrane lysis), methanolic extractives of C. reticulata peel at a concentration of 2.0 mg/mL revealed a mild protective effect (Figure 3). AQSF, in particular, displayed 34.71% effective inhibition than the other extract.
Figure 3.
Percentage inhibition of hemolysis of different extractives of peel of C. reticulata in hypotonic solution-induced and heat-induced conditions [Here, ME, PE, DCM, EA, and AQ mean the soluble fractions of C. reticulata in methanol, pet ether, dichloromethane, ethyl acetate, aqueous conditions, respectively.].
Abbreviations: ASA, acetyl salicylic acid.
Heat-induced hemolysis
The extractives from the peel of C. reticulata were efficient in erythrocyte lysis triggered by heat according to the outcomes. The inhibition of AQSF was around 48.14%, which is greater than that of MECR (34.39%), EASF (31.91%), PESF (30.68%), and DCMSF (24.28%), and even higher than the standard Acetyl Salicylic Acid (42.0%) under normal conditions (Figure 3).
Thrombolytic properties
Streptokinase (SK), a positive control resulting in a notable 64.55% clot lysis. In contrast, the negative control, treated with distilled water, exhibited minimal clot lysis at 5.64%. However, the AQSF of the peel of C. reticulata demonstrated the highest thrombolytic activity at 32.57%, followed by the EASF at 22.61%, DCMSF at 6.80%, MECR at 6.05%, and PESF at 4.06% (Figure 4).
Figure 4.
Effects of methanolic crude extract and its various solvent fractions of C. reticulata peel against thrombosis [Here, ME, PE, DCM, EA, and AQ mean the soluble fractions of C. reticulata in methanol, pet ether, dichloromethane, ethyl acetate, aqueous conditions, respectively].
Abbreviations: ASA, acetyl salicylic acid.
Central analgesic properties
Figure 5a shows the findings of the central analgesic effects of the MECR using the tail immersion technique. The pain reaction time for every sample showed dose-dependent when compared to the control medication, morphine. The 100 and 200 mg/kg body weight doses demonstrated statistically significant central analgesic effects (P < .05) at the 30-, 60-, and 90-minute observation points. In contrast, the 400 mg/kg dose showed statistically significant central analgesic effects (P < .05) only at the 30-minute observation point (Figure 5a).
Figure 5.
In vivo pharmacological actions of MECR: (a) effects of methanolic crude extracts of C. reticulata peel on the tail immersion time (seconds) of the mice in central analgesic test, (b) effects of the 3 doses of methanolic crude extract of C. reticulata peel on the number of writhing of mice in the peripheral analgesic experiment, (c) effects of the 3 doses of methanolic crude extract of C. reticulata peel on the number of diarrheal episodes of mice after 4 hours of administration of Castor oil in antidiarrheal experiment. The data were expressed as mean ± SEM.
“*,” “**,” “***,” and “****” denote P < .05, P < .01, P < .001, and P < .0001, respectively versus control group.
Peripheral analgesic properties
The findings of the peripheral analgesic effect of methanolic crude extracts (100, 200, and 400 mg/kg body weight) in mice are shown in Figure 5b. The MECR demonstrated significant efficacy in inhibiting acetic acid-induced abdominal writhing in mice by reducing the number of writhing of mice. After 30 minutes of pain induction, the 100 mg/kg extract reduced pain by 32.18%, while the 400 mg/kg dosage inhibited writhing by 44.83% in mice, compared to the standard diclofenac sodium (77.0% inhibition; Figure S3).
Anti-diarrheal properties
Castor oil-induced diarrhea lasted up to 4 hours in the control group. The anti-diarrheal evaluation of MECR indicated a very strongly significant (P < .0001) antidiarrheal action at various dosages (100, 200, and 400 mg/kg; Figure 5c). Among the studied groups, ME 200 and ME 400 groups showed nearly 70% inhibition in diarrheal episodes at 4 hours (Figure S4), whereas the positive control had a 73.91% reduction in diarrheal episodes at 4 hours (Figure S4).
Molecular docking analysis
Molecular docking studies were performed using various computer-based methods to evaluate the bioactivities of compounds generated from Citrus reticulata peel extracts and their solvent fractions. Docking scores, as generated by PyRx, are presented in Figure 6. Supplemental Tables S6–S10 provides detailed information on the amino acids interacting with ligand atoms, including the interaction type, distance, and nature. A lower binding affinity value (kcal/mol) indicates stronger binding strength. Optimal docking predictions were identified based on projected binding affinities with a root mean square deviation (RMSD) value of zero. The following sections describe the inhibitory potential of the isolated compounds against specific enzymes and receptors.
Figure 6.
Heat map revealing the binding affinity (kcal/mol) obtained from in silico molecular docking between the isolated compounds and the target receptors. Here, the ciprofloxacin, warfarin, diclofenac, morphine, and loperamide were used as standard drugs while interactions with 4M6J, 1A5H, 1CX2, 4C1M, and 6VI4, respectively.
Inhibition of dihydrofolate reductase (DHFR)
Regarding dihydrofolate reductase (DHFR; 4M6J), all compounds exhibited similar or better binding affinities (−7.0 to −7.6 kcal/mol) when compared to the standard ciprofloxacin (−7.4 kcal/mol). Among them, C-84 had the highest binding affinity (−7.6 kcal/mol), followed by C-A, C-B, and C-87 (each at −7.5 kcal/mol) toward the DHFR protein (Figure 6). The three-dimensional (3D) and two-dimensional (2D) interactions of the best-hit 4 compounds [tangeretin (Compound A), nobiletin (Compound B), 6-Demethoxytangeretin (Compound 84), and pebrellin (Compound 87)] while interacting with DHFR were shown in the Supplemental Figure S5. Predicted interactions were predominantly conventional hydrogen bonds, with some hydrophobic interactions such as pi-alkyl and alkyl bonds also observed (Supplemental Table S6).
Inhibition of tissue plasminogen activator (t-PA)
In the case of molecular docking of tissue plasminogen activator (1A5H), compounds C-B, C-83, and C-91 showed lower binding affinities (−6.5, −6.8, and −6.7 kcal/mol, respectively) compared to the standard warfarin (−7.5 kcal/mol; Figure 6). However, C-C exhibited the highest binding affinity (−9.5 kcal/mol) among all docked compounds, surpassing warfarin, while the other molecules demonstrated binding affinities close to warfarin (−7.1 to 7.8 kcal/mol; Figure 6). The Supplemental Figure S6 shows the three-dimensional (3D) and two-dimensional (2D) interactions of the best-hit 4 compounds [limonin (Compound C), ß-sitosterol (Compound D), salvigenin (Compound 79), and stigmasterol (Compound 86)] while interacting with t-PA revealing anti-thrombotic properties. The interactions noted included hydrophilic (conventional hydrogen bonds and carbon hydrogen bonds) and hydrophobic interactions (alkyl and pi-alkyl; Table S7).
Inhibition of COX-2 enzyme
For cyclooxygenase 2 (1CX2), all docked molecules showed better binding affinities (−9.7 to −7.5 kcal/mol) compared to the positive control diclofenac (−7.3 kcal/mol; Figure 6). Figure 7 exhibited the three-dimensional (3D) and two-dimensional (2D) interactions of the 4 best-hit compounds [ß-sitosterol (Compound D), salvigenin (Compound 79), 6-Demethoxytangeretin (Compound 84), and stigmasterol (Compound 86)] while interacting with cyclooxygenase 2 (COX 2) revealing peripheral analgesic properties. The interactions were mostly hydrophobic, involving alkyl and pi-alkyl bonds (Table S8).
Figure 7.
Three-dimensional (3D) and two-dimensional (2D) interactions of the 4 best-hit compounds while interacting with cyclooxygenase 2 (COX 2) revealing peripheral analgesic properties. [Here, (a-h) represent the interactions of ß-sitosterol (Compound D), salvigenin (Compound 79), 6-Demethoxytangeretin (Compound 84), and stigmasterol (Compound 86), respectively, while inhibiting the COX-2 protein.].
Inhibition of mu-opioid receptor
When investigating central analgesic properties via molecular docking analysis with the mu-opioid receptor (5C1M), compounds C, D, and 86 demonstrated more efficient binding affinities (<−9.2 kcal/mol) compared to the standard morphine (−8.0 kcal/mol). The other molecules displayed similar efficiencies (−7.6 to −8.0 kcal/mol) to the positive control (Figure 6). Figure 8 showed the three-dimensional (3D) and two-dimensional (2D) interactions of the 4 best-hit compounds [nobiletin (Compound B), limonin (Compound C), ß-sitosterol (Compound D), and stigmasterol (Compound 86)] while interacting with mu-opioid receptor revealing central analgesic properties. Most ligand-protein interactions were hydrophobic, including pi-alkyl and alkyl bonds, with some hydrogen bonds and 1 electrostatic bond also observed (Table S9).
Figure 8.
Three-dimensional (3D) and two-dimensional (2D) interactions of the 4 best-hit compounds while interacting with mu-opioid receptor revealing central analgesic properties. [Here, (a-h) represent the interactions of nobiletin (Compound B), limonin (Compound C), ß-sitosterol (Compound D), and stigmasterol (Compound 86), respectively, while inhibiting the mu-opioid receptor.].
Inhibition of kappa opioid receptor
The compounds ß-sitosterol (C-D) and stigmasterol (C-86) exerted the highest binding affinities (−8.1 kcal/mol) toward the kappa opioid receptor than the standard molecule loperamide (−7.7 kcal/mol) revealing their promising antidiarrheal activities (Figure 6). The Supplemental Figure S7 exhibited the three-dimensional (3D) and two-dimensional (2D) interactions of the 4 best-hit compounds [nobiletin (Compound B), limonin (Compound C), ß-sitosterol (Compound D), and stigmasterol (Compound 86)] while interacting with kappa opioid receptor revealing antidiarrheal properties. The interactions were predominantly hydrophobic, such as pi-alkyl and alkyl bonds (Supplemental Table S10).
ADMET, physicochemical, and drug-likeliness analysis
Poor ADMET profiles are well-known barrier to design effective therapeutic molecule. One of the major challenges in drug development during clinical investigation is addressing these pharmacokinetic characteristics. To overcome these challenges, in silico techniques have been employed to evaluate ADMET properties and predict the drug-likeness of isolated and identified 12 compounds (4 isolated compounds and 8 selected phytocompounds which were detected by GC-MS/MS technique) as potential therapeutic candidates.98-100 The results of these ADMET and drug-likeliness analyses are summarized in Tables 3 and 4.
Table 3.
Summary of the parameters regarding the pharmacokinetic and toxicological properties of the lead identified compounds from the peel of C reticulata.
Properties | Model name (Unit) | C-A | C-B | C-C | C-D | C-79 | C-83 | C-84 | C-86 | C-87 | C-88 | C-90 | C-91 |
---|---|---|---|---|---|---|---|---|---|---|---|---|---|
Absorption | Water solubility (log mol/L) | −4.792 | −4.949 | −4.04 | −6.773 | −3.897 | −4.794 | −4.58 | −6.68 | −3.479 | −4.33 | −4.682 | −4.32 |
Caco2 permeability (log Papp in 10−6 cm/s) | 1.245 | 1.306 | 0.922 | 1.201 | 1.087 | 1.221 | 1.171 | 1.213 | 0.567 | 1.424 | 1.233 | 1.455 | |
Intestinal absorption (human; % Absorbed) | 98.48 | 98.92 | 100 | 94.46 | 95.87 | 98.55 | 98.25 | 94.97 | 97.22 | 100 | 98.58 | 100 | |
Skin Permeability (log Kp) | −2.678 | −2.715 | −2.83 | −2.78 | −2.71 | −2.68 | −2.62 | −2.78 | −2.75 | −2.75 | −2.67 | −2.75 | |
P-glycoprotein substrate | No | No | No | No | Yes | No | No | No | Yes | Yes | No | Yes | |
P-glycoprotein I inhibitor | Yes | Yes | Yes | Yes | Yes | Yes | Yes | Yes | No | Yes | Yes | Yes | |
P-glycoprotein II inhibitor | Yes | Yes | No | Yes | Yes | Yes | Yes | Yes | Yes | Yes | Yes | Yes | |
Distribution | VDss (human; log L/kg) | −0.22 | −0.28 | 0.26 | 0.19 | −0.16 | −0.22 | −0.17 | 0.17 | −0.02 | −0.24 | −0.18 | −0.25 |
Fraction unbound (human; Fu) | 0.188 | 0.179 | 0.145 | 0 | 0.14 | 0.195 | 0.197 | 0 | 0.069 | 0.123 | 0.155 | 0.118 | |
BBB permeability | −1.026 | −1.254 | −0.84 | 0.781 | −0.695 | −1.02 | −0.23 | 0.771 | −1.021 | −1.15 | −1.008 | −1.14 | |
CNS permeability | −3.011 | −3.142 | −3.07 | −1.705 | −2.309 | −3.032 | −2.27 | −1.65 | −3.198 | −3.16 | −3.016 | −3.17 | |
Metabolism | CYP2D6 substrate | No | No | No | No | No | No | No | No | No | No | No | No |
CYP3A4 substrate | Yes | Yes | Yes | Yes | Yes | Yes | Yes | Yes | No | Yes | Yes | Yes | |
CYP1A2 inhibitor | Yes | Yes | No | No | Yes | Yes | Yes | No | Yes | Yes | Yes | Yes | |
CYP2C19 inhibitor | Yes | Yes | No | No | Yes | Yes | Yes | No | Yes | Yes | Yes | Yes | |
CYP2C9 inhibitor | Yes | Yes | No | No | No | Yes | Yes | No | No | Yes | Yes | Yes | |
CYP2D6 inhibitor | No | No | No | No | No | No | No | No | No | No | No | No | |
CYP3A4 inhibitor | Yes | Yes | No | No | Yes | Yes | No | No | Yes | No | No | Yes | |
Excretion | Total Clearance (log ml/min/kg) | 0.78 | 0.789 | 0.088 | 0.628 | 0.703 | 0.422 | 0.791 | 0.618 | 0.625 | 0.706 | 0.794 | 0.703 |
Renal OCT2 substrate | No | No | No | No | Yes | No | No | No | No | No | No | No | |
Toxicity | AMES toxicity | No | No | No | No | No | No | No | No | No | No | No | No |
Max. tolerated dose (human; log mg/kg/day) | 0.385 | 0.443 | −0.51 | −0.621 | 0.202 | 0.4 | 0.295 | −0.66 | 0.293 | 0.335 | 0.208 | 0.336 | |
hERG I inhibitor | No | No | No | No | No | No | No | No | No | No | No | No | |
hERG II inhibitor | No | No | No | Yes | No | No | No | Yes | No | No | No | No | |
Oral rat acute toxicity (LD50; mol/kg) | 2.368 | 2.459 | 3.452 | 2.552 | 2.229 | 2.401 | 2.29 | 2.54 | 2.273 | 2.36 | 2.503 | 2.406 | |
Oral rat chronic toxicity (LOAEL; log mg/kg_bw/day) | 0.944 | 0.82 | 1.911 | 0.855 | 1.307 | 0.95 | 1.095 | 0.872 | 1.987 | 1.025 | 1.131 | 1.028 | |
Hepatotoxicity | No | No | No | No | No | No | No | No | No | No | No | No | |
Skin sensitization | No | No | No | No | No | No | No | No | No | No | No | No | |
T. Pyriformis toxicity (log µg/L) | 0.355 | 0.315 | 0.286 | 0.43 | 0.481 | 0.351 | 0.418 | 0.433 | 0.31 | 0.332 | 0.384 | 0.322 | |
Minnow toxicity (log mM) | 0.144 | 0.686 | 0.446 | −1.802 | 0.856 | 0.326 | −0.2 | −1.68 | 2.331 | 1.842 | 0.908 | 1.842 |
Table 4.
Summary of the parameters regarding the physicochemical properties and drug-likeliness qualities.
Comp. No. | Compound name | Mol weight (g/mol) | No. of rotatable bonds | H-bond acceptor | H-bond donor | Molar refractivity | TPSA (Å2) | Consensus LogPo/w | Lipinski rule | Bioavailability score | |
---|---|---|---|---|---|---|---|---|---|---|---|
Result | Violation | ||||||||||
C-A | Tangeretin | 372.37 | 6 | 7 | 0 | 100.38 | 76.36 | 3.02 | Yes | 0 | 0.55 |
C-B | Nobiletin | 402.39 | 7 | 8 | 0 | 106.87 | 85.59 | 3.02 | Yes | 0 | 0.55 |
C-C | Limonin | 470.51 | 1 | 8 | 0 | 116.17 | 104.57 | 2.55 | Yes | 0 | 0.55 |
C-D | ß-Sitosterol | 414.71 | 1 | 1 | 0 | 133.23 | 20.23 | 7.19 | Yes | 1 | 0.55 |
C-79 | Salvigenin | 328.32 | 4 | 6 | 1 | 89.42 | 78.13 | 2.88 | Yes | 0 | 0.55 |
C-83 | Pentamethoxyflavone | 372.37 | 6 | 7 | 0 | 100.38 | 76.36 | 3.01 | Yes | 0 | 0.55 |
C-84 | 6-Demethoxytangeretin | 342.34 | 5 | 6 | 0 | 93.89 | 67.13 | 3.00 | Yes | 0 | 0.55 |
C-86 | Stigmasterol | 412.69 | 5 | 1 | 1 | 132.75 | 20.23 | 6.97 | Yes | 1 | 0.55 |
C-87 | Pebrellin | 374.34 | 5 | 8 | 2 | 97.93 | 107.59 | 2.43 | Yes | 0 | 0.55 |
C-88 | Artemetin | 388.37 | 6 | 8 | 1 | 102.40 | 96.59 | 2.82 | Yes | 0 | 0.55 |
C-90 | Senensetin | 372.37 | 6 | 7 | 0 | 100.38 | 76.36 | 3.10 | Yes | 0 | 0.55 |
C-91 | Demethylnobiletin | 388.37 | 6 | 8 | 1 | 102.40 | 96.59 | 2.78 | Yes | 0 | 0.55 |
Abbreviations: TPSA, topological polar surface area.
Most of the molecules exhibited similar water solubility (3.89 log mol/L), with the exceptions of C-D, C-86, and C-87, which had solubility values of 6.77, 6.68, and 3.47 log mol/L, respectively (Table 3). Nearly all compounds demonstrated Caco-2 permeability, except for C-87. The phytocompounds also showed comparable human intestinal absorption and skin permeability properties. While C-79, C-87, C-88, and C-91 were not substrates for P-glycoprotein, approximately all other compounds, except for C-87, were able to inhibit P-glycoprotein-I (Table 3). In terms of distribution, most compounds were unbound in a small fraction, except for C-D and C-86, which were fully bounded (Table 3). Compounds C-B and C-87 exhibited higher blood-brain barrier (BBB) permeability compared to other molecules. Additionally, all compounds, except for C-D and C-87 (−1.705 and −1.65, respectively), showed strong central nervous system (CNS) permeability (Table 3). Regarding drug metabolism, all compounds could act as substrates for CYP2D6 and CYP3A4, except for 87 (Table 3). Compounds C, D, and 86 did not show inhibitory action against any types of CYP450 in the docking study, and no compounds were able to inhibit CYP2D6 (Table 3). For renal clearance, most compounds exhibited total clearance (expressed as log mL/kg/min) ranging from 0.422 to 0.789, except for C-C (0.088). All the compounds acted as non-substrates for renal OCT2 (organic cation transporter 2), excluding C-79 (Table 3). The compounds displayed no AMES toxicity, hERG I inhibition, hERG II inhibition, hepatotoxicity, or skin sensitization (Table 3). According to the OECD 423 model, 101 the obtained LD50 values indicated no acute oral toxicity, classifying the substances as class VI (“non-toxic”) under the Oral Toxicity Classification. Furthermore, nearly all compounds adhered to Lipinski’s rule of 5, with a bioavailability score of 0.55 in the drug-likeness analysis. However, compounds D and 86 had an octanol-water partition coefficient log P higher than Lipinski’s recommendation (log P ⩾ 5; Table 4).
Discussion
Fruit plants and vegetables are considered a significant reservoir of bioactive phytochemicals, illustrating various pharmacological properties. Substantial quantities of secondary metabolites are inherent within fruit and vegetable waste or byproducts, thereby the presence of phenolic molecules, dietary fibers, and other biologically active constituents through extraction and isolation techniques can be identified. Scientific research has elucidated the prevalence of phytochemicals and essential nutrients within various components of fruits and vegetables, including peels, seeds, and pulp. 102 The systematic isolation and characterization of these phytochemicals represent a well-recognized approach for identifying bioactive agents derived from fruit waste. Within the phytochemical analysis, several chromatographic methodologies, encompassing column chromatography (CC), thin-layer chromatography (TLC), gas chromatography (GC), and preparative thin-layer chromatography (PTLC), are employed in the purification and refinement of bioactive compounds from botanical extracts. In our study, we conducted a chemical profiling of Citrus reticulata fruit peel utilizing methanol as the solvent, employing gel permeation chromatography, TLC screening, and subsequent PTLC analysis. In our study, NMR approaches was applied to confirm the subsequent isolated phytochemical structures in the investigation. Furthermore, GC-MS technique was employed to comprehensively identify additional phytomolecules from the methanolic extract of C. reticulata peel.
The present research demonstrated the isolation of 4 compounds from C. reticulata, including tangeretin (compound A), nobiletin (compound B), limonin (compound C), and ß-sitosterol (compound D), as depicted in Figure 1. These phytochemicals were also detected from the same species reported in previous studies.103-105 Nobiletin and tangeretin were separated from the peel extract of C. reticulata and reported by Kaushal et al 106 and Mizuno et al, 107 respectively. Additionally, tangeretin and limonin were extracted from the same botanical component and documented by Yaqoob et al. 108 Nevertheless, while ß-sitosterol was previously identified and documented in the stem bark extract, 105 as per our searching experience, this current study represents the first report of its isolation from the C. reticulata peel extract. Furthermore, GC-MS approaches detected and characterized over 90 bioactive compounds, notably, D-limonene (antioxidant, antidiabetic, anticancer), emylcamate (anti-cancer, antioxidant), furaneol (antimicrobial), thymine (antibiotics), ethanamine, N-ethyl-nitroso (anti-cancer), ribitol (anti-microbial), thymol (anti-inflammatory, antiseptic), phytol (cytotoxicity, antioxidant, antimicrobial), ß-elemene (antibacterial), humulene (antifungal, anticancer), trehalose (anti-inflammatory), bis (2-ethylhexyl) phthalate (antimicrobial, cytotoxicity), fenoterol (antimicrobial), salvigenin (anti-inflammatory). In addition, many fatty derivatives such as cis,cis,cis – 7,10,13-hexadecatrienal, cis-11-eicosenamide, 9,12-octadecadienoic acid (Z,Z)-,2,3- dihydroxypropyl ester, and hexadecanoic acid 2-hydroxy1- (hydroxymethyl) ethyl ester were also identified in our phytochemical analysis, which are mainly promising for their antioxidant properties (Table 2).
The assessment of the antibacterial efficacy of the methanolic extract of C. reticulata and its assorted fractions was conducted with the aim of identifying novel candidates for antimicrobial agents. This initiative fueled from the escalating issue of resistance exhibited by numerous strains of bacteria against conventional antibiotics, thereby emphasizing the imperative for alternative therapeutics. Additionally, contemporary antibiotic treatments often entail heightened adverse effects, underscoring the urgency to explore alternative avenues for combating bacterial infections. 109 Microorganisms resistant to antimicrobial agents have been identified as the primary causative agents in an estimated 700 000 fatalities annually worldwide. This alarming figure underscores the urgent need for concerted efforts, including discovering novel antibiotics to address antimicrobial resistance. 110
The present investigation revealed antibacterial efficacy across various extractives of C. reticulata, with notable antimicrobial activity observed specifically within the dichloromethane-soluble fraction. These findings underscore the potential of natural sources such as C. reticulata in pursuing novel antimicrobial agents amid the escalating global challenge of antimicrobial resistance. 111 In addition, the isolated compounds such as tangeretin and nobiletin showed potential antimicrobial activity at several previous studies. 112 Moreover, a previous study of the fruit peel showed that the presence of tangeretin and nobiletin exhibited potent antimicrobial activity. 113 Additionally, the isolated compounds demonstrated notable binding affinities toward the dihydrofolate reductase (DHFR) enzyme, with values ranging from −7.0 to −7.6 kcal/mol. These affinities compare favorably to the standard drug ciprofloxacin (−7.4 kcal/mol). In addition, several compounds such as santolina epoxide, 3-(2-piperidinyl) sulfate, ß-elemene, α-farnesene, dodecanoic acid, megastigmatrienone, 2,3-anhydro-d-mannosan and isovaleric acid, eicosyl ester were identified through the GC-MS analysis which have anti-microbial properties (Table 2). Further investigation is needed to establish the exact mechanism of actions of these phytochemicals from the C. reticulata fruits peel toward microbial infections.
Thrombosis stands as a leading contributor to morbidity and mortality across a diverse range of vascular dysfunctions. 114 By the influence of activated thrombin, fibrinogen undergoes conversion to fibrin, leading to the formation of a thrombus or blood clot. Individuals with occluded veins or arteries receive treatment with fibrinolytic medications, eliminating thrombi by activating tissue plasminogen activator (t-PA) and manifesting a thrombolytic effect. 115 Diminishing platelet aggregation serves as a protective measure against specific conditions, such as atherosclerosis. 116 In this present investigation, the EASF and AQSF of C. reticulata displayed moderate thrombolytic activity (22.61% and 32.57% clot lysis, respectively). Furthermore, a prior study suggests that the identified compound nobiletin could be a promising therapeutic agent for preventing or treating thromboembolic illnesses. 117 Another compound, tangeretin, blocks agonist-induced human platelet activation in a concentration-dependent manner. It hinders agonist-induced integrin αIIbβ3 inside-out and outside-in signaling, intracellular calcium mobilization, and granule secretion. 118 Furthermore, nearly all the selected compounds from C. reticulata for molecular docking exhibited binding affinities toward human tissue plasminogen activator that surpassed those of the standard thrombolytic drug warfarin (−7.5 kcal/mol). The findings from this investigation may contribute to the find out the thrombolytic properties of the fruit peel of C. reticulata.119,120
Substances capable of stabilizing the erythrocyte membrane may also stabilize the lysosomal membrane, thereby exhibiting anti-inflammatory properties by influencing the activity and release of cell mediators due to their structural similarities.25,121 In this context, the AQSF of C. reticulata showcased notable anti-inflammatory efficacy by averting erythrocyte hemolysis in conditions induced by both hypotonic solution and heat. GC-MS analysis revealed the presence of several anti-inflammatory compounds in MECR, including α-farnesene, dl-citrulline, trehalose, ethyl cholate, octadecanamide, salvigenin, vitamin E, sinensetin, and demethylnobiletin. Furthermore, a compound like limonin exhibited potent inhibition of IL-1β-Induced Inflammation, mitigating osteoarthritis inflammation by activating Nrf2. 122 Additionally, all the docking compounds demonstrated the enhanced binding affinities for the cyclooxygenase protein (COX 2), which supports the outcomes of the experimental studies. Plausibly, these molecules might be accountable for the anti-inflammatory activity of C. reticulata. Additional investigations should be conducted to isolate and identify the specific molecules responsible for the membrane-stabilizing properties.
Exploration for pain-alleviating substances is emphasized from natural reservoirs as substitutes for synthetic medications due to their lower adverse effects. 123 The current investigation revealed the analgesic potential of the methanol extract from the peel of C. reticulata using the writhing and tail immersion methods in mice. In our experimental setting, the plant extract mitigated acetic acid-induced pain responses in mice more effectively than heat-induced pain reactions. The findings propose that phytochemicals within the fruit peel extracts of C. reticulata may possess the capability to diminish pain by impeding prostaglandin synthesis.124,125 Some of the isolated and detected compounds of this phytochemical investigation showed high binding affinity than the conventional analgesic medicine morphine and diclofenac. One prior study indicated that the isolated compound β-sitosterol exhibited analgesic effects in hot plate and acetic acid-induced assessments by either inhibiting central opioid receptors or facilitating the release of endogenous opioid peptides, and inhibiting the production of prostaglandins and bradykinins. 126 Furthermore, the GC-MS technique detected certain analgesic compounds such as gamma-Terpinene, Linalool, and Benzofuran, 2,3-dihydro, suggesting the fruit peel of C. reticulata possesses analgesic attributes.
Since time immemorial, medicinal flora has been utilized to address various gastrointestinal (GI) disorders, such as diarrhea. Nonetheless, the safety and effectiveness profiles of the majority of these botanicals have not been scrutinized. As a result, this investigation was done to assess the efficacy of the peel extract of C. reticulata fruits as a potential remedy for diarrhea. Ricinoleic acid, the primary element in castor oil, is known to provoke irritation of the intestinal wall, eliciting peristaltic motion and ultimately leading to diarrhea. 127 The MECR of C. reticulata exhibited significant properties to counteract diarrhea. Even the smaller dosage (100 mg/kg bw) displayed a significant antidiarrheal effect. This exploration illustrated that the anti-diarrheal capabilities of the MECR could be ascribed to the bioactive phytochemicals accountable for the anti-diarrheal effect. 128 Molecular docking displayed that 3 compounds C, D, and 86 had higher binding affinities than the standard drug loperamide. β-Sitosterol, an isolated secondary metabolite from C. reticulata, might assume a significant function in diminishing peristalsis in the GI system, consequently averting GI motility.127,128 While the GC-MS method did not reveal any potent antidiarrheal phytomolecules, according to the literature pharmacological activities records, antioxidant compounds may be instrumental in mitigating intestinal motility. 129 These antioxidant agents could alleviate the impact of castor oil-induced intestinal inflammation while several antioxidant compounds were identified through the GC-MS approach. Furthermore, 2 flavonoid molecules, tangeretin, and nobiletin, possess potent antioxidant attributes.130,131
Limitations and future research
The current study presents certain limitations and gaps. The purified compounds were not pharmacologically investigated. Instead, the study reported their actions computationally to support the pharmacological properties of the MECR and its various solvent fractions. Nevertheless, the study will serve as a foundation for future research focused on the extensive phytochemical isolation of additional polymethoxyflavones and other flavonoids and their pharmacological evaluations. Moreover, it is recommended that the most abundant purified polymethoxyflavones be assessed for their potential effectiveness against chronic disorders, including infections, thrombosis, inflammation, pain, and diarrhea, based on the present evidence. Additionally, the MECR has not been subjected to acute, subacute, or chronic toxicity evaluations in this report, which should be addressed in future studies.
Conclusion
The present study successfully identified and isolated four purified compounds: tangeretin, nobiletin, limonin, and ß-sitosterol, utilizing column chromatography, TLC, and NMR spectroscopic techniques. Additionally, the study detected over 90 phytoconstituents in the fruit peel extract of Citrus reticulata through GC-MS/MS analysis. The study also evaluated the pharmacological properties of the peel extract of C. reticulata, focusing on antibacterial, thrombolytic, anti-inflammatory, analgesic, and antidiarrheal activities. Promising in vitro antimicrobial and anti-inflammatory properties were exhibited by the methanolic fruit peel extract and its various solvent fractions. Statistically significant in vivo peripheral analgesic and antidiarrheal effects were observed with the crude extract at the tested doses (100, 200, and 400 mg/kg bw), where the outcomes were further supported by in silico investigations. In addition, the reported promising compounds demonstrated favorable pharmacokinetics and safety profiles, which are essential for the development of new drugs. The presence of various polymethoxyflavones in this plant underlines the numerous therapeutic benefits of C. reticulata fruit peels. However, additional research is needed to fully understand the precise mechanisms and discover the bioactive compounds that govern the actions of C. reticulata through a combined experimental and computational approach.
Supplemental Material
Supplemental material, sj-pdf-1-nmi-10.1177_11786388251327668 for Phytochemical Isolation and Antimicrobial, Thrombolytic, Anti-inflammatory, Analgesic, and Antidiarrheal Activities from the Shell of Commonly Available Citrus reticulata Blanco: Multifaceted Role of Polymethoxyflavones by Md. Jamal Hossain, Md. Abdus Samadd, Mst. Nusrat Zahan Urmi, Mst. Farzana Yeasmin Reshmi, Md. Shohel Hossen and Mohammad A. Rashid in Nutrition and Metabolic Insights
Acknowledgments
We extend our gratitude to the Department of Pharmacy at the State University of Bangladesh for the laboratory facilities and logistical support essential for conducting this research.
Footnotes
Funding: The author(s) received no financial support for the research, authorship, and/or publication of this article.
The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Ethics: The protocols for animal handling and procedures involved in the animal studies were thoroughly reviewed and approved by the Animal Ethics Committee at the State University of Bangladesh, Dhaka, Bangladesh (approval number: 2022-01-23/SUB/A-ERC/006).
Informed Consent: Not required.
ORCID iDs: Md. Jamal Hossain
https://orcid.org/0000-0001-9706-207X
Md. Abdus Samadd
https://orcid.org/0000-0002-8287-5454
Md. Shohel Hossen
https://orcid.org/0009-0000-8435-9712
Data Availability Statement: Data included in article/supp. material/referenced in article.
Supplemental Material: Supplemental material for this article is available online.
References
- 1. George P. Concerns regarding the safety and toxicity of medicinal plants-an overview. J Appl Pharm Sci. 2011;1:40-44. [Google Scholar]
- 2. Sahoo N, Manchikanti P. Herbal drug regulation and commercialization: an Indian industry perspective. J Altern Complement Med. 2013;19:957-963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Yuan H, Ma Q, Ye L, Piao G. The traditional medicine and modern medicine from natural products. Molecules. 2016;21:559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Anand U, Jacobo-Herrera N, Altemimi A, Lakhssassi N. A comprehensive review on medicinal plants as antimicrobial therapeutics: potential avenues of biocompatible drug discovery. Metabolites. 2019;9:258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Bayrakçeken Güven Z, Saracoglu I, Nagatsu A, Yilmaz MA, Basaran AA. Anti-tyrosinase and antimelanogenic effect of cinnamic acid derivatives from Prunus mahaleb L.: phenolic composition, isolation, identification and inhibitory activity. J Ethnopharmacol. 2023;310:116378. [DOI] [PubMed] [Google Scholar]
- 6. Pandey MM, Rastogi S, Rawat AK. Indian traditional ayurvedic system of medicine and nutritional supplementation. Evid Based Complement Alternat Med. 2013;2013:376327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Atanasov AG, Zotchev SB, Dirsch VM, Supuran CT. Natural products in drug discovery: advances and opportunities. Nat Rev Drug Discov. 2021;20:200-216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Nasim N, Sandeep IS, Mohanty S. Plant-derived natural products for drug discovery: current approaches and prospects. Nucleus. 2022;65:399-411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Riaz M, Rasool N, Bukhari IH, Shahid M, Zubair K, Rizwan U. In vitro antimicrobial, antioxidant, cytotoxicity and GC-MS analysis of Mazus goodenifolius. Molecules. 2012;17:14275-14278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Mesa C, Ranalison O, Randriantseheno LN, Risuleo G. Natural products from Madagascar, socio-cultural usage, and potential applications in advanced biomedicine: a concise review. Molecules. 2021;26:4507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Sultana A, Hossain MJ, Kuddus MR, et al. Ethnobotanical uses, phytochemistry, toxicology, and pharmacological properties of Euphorbia neriifolia linn. against infectious diseases: a comprehensive review. Molecules. 2022;27:4374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Sawamura M, Thi Minh Tu N, Onishi Y, Ogawa E, Choi H-S. Characteristic odor components of citrus reticulata Blanco (Ponkan) cold-pressed oil. Biosci Biotechnol Biochem. 2004;68:1690-1697. [DOI] [PubMed] [Google Scholar]
- 13. Shorbagi M, Fayek NM, Shao P, Farag MA. Citrus reticulata Blanco (the common mandarin) fruit: an updated review of its bioactive, extraction types, food quality, therapeutic merits, and bio-waste valorization practices to maximize its economic value. Food Biosci. 2022;47:101699. [Google Scholar]
- 14. Chopra RN, Nayar SL, Chopra IC. Glossary of Indian Medicinal Plants (including the Supplement). Council of Scientific and Industrial Research. 1986:77-100. [Google Scholar]
- 15. Khan MA, Ali M, Alam P. Phytochemical investigation of the fruit peels of Citrus reticulata Blanco. Nat Prod Res. 2010;24:610-620. [DOI] [PubMed] [Google Scholar]
- 16. Zou Z, Xi W, Hu Y, Nie C, Zhou Z. Antioxidant activity of citrus fruits. Food Chem. 2016;196:885-896. [DOI] [PubMed] [Google Scholar]
- 17. Apraj VD, Pandita NS. Evaluation of skin anti-aging potential of citrus reticulata Blanco peel. Pharmacogn Res. 2016;8:160-168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Sultana HS, Ali M, Panda BP. Influence of volatile constituents of fruit peels of citrus reticulata Blanco on clinically isolated pathogenic microorganisms under in-vitro. Asian Pac J Trop Biomed. 2012;2:S1299-1302. [Google Scholar]
- 19. Liu Y, Heying E, Tanumihardjo SA. History, global distribution, and nutritional importance of citrus fruits. Compr Rev Food Sci Food Saf. 2012;11:530-545. [Google Scholar]
- 20. Li S, Pan MH, Lo CY, et al. Chemistry and health effects of polymethoxyflavones and hydroxylated polymethoxyflavones. J Funct Foods. 2009;1:2-12. [Google Scholar]
- 21. Guo J, Tao H, Cao Y, Ho C-T, Jin S, Huang Q. Prevention of obesity and Type 2 diabetes with aged Citrus Peel (Chenpi) extract. J Agric Food Chem. 2016;64:2053-2061. [DOI] [PubMed] [Google Scholar]
- 22. VanWagenen BC, Larsen R, Cardellina JH, Randazzo D, Lidert ZC, Swithenbank C. Ulosantoin, a potent insecticide from the sponge Ulosa ruetzleri. J Org Chem. 1993;58:335-337. [Google Scholar]
- 23. Rashid PT, Hossain MJ, Zahan MS, et al. Chemico-pharmacological and computational studies of Ophiorrhiza fasciculata D. Don and Psychotria silhetensis hook. f. focusing cytotoxic, thrombolytic, anti-inflammatory, antioxidant, and antibacterial properties. Heliyon. 2023;9:e20100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Bauer AW, Kirby WMM, Sherris JC, Turck M. Antibiotic susceptibility testing by a standardized single disk method. Am J Clin Pathol. 1966;45:493-496. [PubMed] [Google Scholar]
- 25. Debnath PC, Das A, Islam A, Islam MA, Hassan MM, Gias Uddin SM. Membrane stabilization–a possible mechanism of action for the anti-inflammatory activity of a Bangladeshi medicinal plant: Erioglossum rubiginosum (Bara Harina). Pharmacogn J. 2013;5:104-107. [Google Scholar]
- 26. Prasad S, Kashyap RS, Deopujari JY, Purohit HJ, Taori GM, Daginawala HF. Development of an in vitro model to study clot lysis activity of thrombolytic drugs. Thromb J. 2006;4:14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Percie du, Sert N, Hurst V, Ahluwalia A, et al. The ARRIVE guidelines 2.0: updated guidelines for reporting animal research. J Cereb Blood Flow Metab. 2020;40:1769-1777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Bulbul IJ, Hossain MJ, Haque MR, et al. Two rare flavonoid glycosides from Litsea glutinosa (Lour.) CB Rob.: experimental and computational approaches endorse antidiabetic potentiality. BMC Complement Med Ther. 2024;24:69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Mukta MM, Hossain MJ, Akter M, et al. Cardioprotection of water spinach (Ipomoea aquatica), wood apple (Limonia acidissima) and linseed (Linum usitatissimum L.) on doxorubicin-induced cardiotoxicity and oxidative stress in rat model. Nutr Metab Insights. 2023;16:11786388231212116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Rahman MM, Soma MA, Sultana N, et al. Exploring therapeutic potential of Woodfordia fruticosa (L.) Kurz leaf and bark focusing on antioxidant, antithrombotic, antimicrobial, anti-inflammatory, analgesic, and antidiarrheal properties. Health Sci. Rep. 2023;6:e1654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Ezeja M, Omeh Y, Ezeigbo I, Ekechukwu A. Evaluation of the analgesic activity of the methanolic stem bark extract of Dialium guineense (Wild). Ann Med Health Sci Res. 2011;1:55-62. [PMC free article] [PubMed] [Google Scholar]
- 32. Amee KNS, Hossain MJ, Rohoman A, et al. Phytochemical and pharmacological profiling of extracts of pterygota alata (Roxb.) R. Br. Leaves deciphered therapeutic potentialities against pain, hyperglycemia and diarrhea via in vivo approaches. Pharmacol Res Prod. 2024;4:100060. [Google Scholar]
- 33. Deng J, Han J, Chen J, et al. Comparison of analgesic activities of aconitine in different mice pain models. PLoS One. 2021;16:e0249276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Sisay M, Engidawork E, Shibeshi W. Evaluation of the antidiarrheal activity of the leaf extracts of Myrtus communis linn (Myrtaceae) in mice model. BMC Complement Altern Med. 2017;17:103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Shoba FG, Thomas M. Study of antidiarrhoeal activity of four medicinal plants in castor-oil induced diarrhoea. J Ethnopharmacol. 2001;76:73-76. [DOI] [PubMed] [Google Scholar]
- 36. Dallakyan S, Olson AJ. Small-molecule library screening by docking with pyrx. Methods Mol Biol. 2015;1263:243-250. [DOI] [PubMed] [Google Scholar]
- 37. Taher MA, Hossain MJ, Zahan MS, et al. Phyto-pharmacological and computational profiling of Bombax ceiba linn. leaves revealed pharmacological properties against oxidation, hyperglycemia, pain, and diarrhea. Heliyon. 2024;10:e35422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Lekmine S, Benslama O, Kadi K, et al. LC/MS-MS analysis of phenolic compounds in Hyoscyamus albus L. extract: in vitro antidiabetic activity, in silico molecular docking, and in vivo investigation against STZ-induced diabetic mice. Pharmaceuticals. 2023;16:1015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Tallei TE, Fatimawali, Adam AA, et al. Fruit Bromelain-derived peptide potentially restrains the attachment of SARS-CoV-2 variants to hACE2: a pharmacoinformatics approach. Molecules. 2022;27:260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Bikadi Z, Hazai E. Application of the PM6 semi-empirical method to modeling proteins enhances docking accuracy of AutoDock. J Cheminform. 2009;1:15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Farzana M, Hossain MJ, El-Shehawi AM, et al. Phenolic constituents from Wendlandia tinctoria var. grandis (Roxb.) DC. stem deciphering pharmacological potentials against oxidation, hyperglycemia, and diarrhea: phyto-pharmacological and computational approaches. Molecules. 2022;27:5957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Jannat T, Hossain MJ, El-Shehawi AM, et al. Chemical and pharmacological profiling of Wrightia coccinea (roxb. Ex hornem.) sims focusing antioxidant, cytotoxic, antidiarrheal, hypoglycemic, and analgesic properties. Molecules. 2022;27:4024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Khatun MCS, Muhit MA, Hossain MJ, Al-Mansur MA, Rahman SMA. Isolation of phytochemical constituents from Stevia rebaudiana (Bert.) and evaluation of their anticancer, antimicrobial and antioxidant properties via in vitro and in silico approaches. Heliyon. 2021;7:e08475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Kaplan W, Littlejohn TG. Swiss-PDB viewer (deep view). Brief Bioinform. 2001;2:195-197. [DOI] [PubMed] [Google Scholar]
- 45. Assel M, Sjoberg D, Elders A, et al. Guidelines for reporting of statistics for clinical research in urology. BJU Int. 2019;123:401-410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Morimoto M, Kumeda S, Komai K. Insect antifeedant flavonoids from Gnaphalium a ffine D. Don. J Agric Food Chem. 2000;48:1888-1891. [DOI] [PubMed] [Google Scholar]
- 47. Santos JS, Escher GB, da Silva Pereira JM, et al. 1H NMR combined with chemometrics tools for rapid characterization of edible oils and their biological properties. Ind Crops Prod. 2018;116:191-200. [Google Scholar]
- 48. Breksa API, Dragull K, Wong RY. Isolation and identification of the first C-17 limonin epimer, epilimonin. J Agric Food Chem. 2008;56:5595-5598. [DOI] [PubMed] [Google Scholar]
- 49. Majid Shah S, Ullah F, Ayaz M, et al. β-sitosterol from Ifloga spicata (Forssk.)sch. bip. as potential anti-leishmanial agent against leishmania tropica: docking and molecular insights. Steroids. 2019;148:56-62. [DOI] [PubMed] [Google Scholar]
- 50. Chukwu C, Omaka O, Aja P. Characterization of 2,5-dimethyl-2,4-dihydroxy-3(2H) furanone, a flavourant principle from Sysepalum dulcificum. Nat Prod Chem Res. 2017;5:8. doi: 10.4172/2329-6836.1000299 [DOI] [Google Scholar]
- 51. Anandakumar P, Kamaraj S, Vanitha MK. D-limonene: a multifunctional compound with potent therapeutic effects. J Food Biochem. 2021;45:e13566. [DOI] [PubMed] [Google Scholar]
- 52. Zhang L-L, Chen Y, Li Z-J, Li X, Fan G. Bioactive properties of the aromatic molecules of spearmint (Mentha spicata L.) essential oil: a review. Food Funct. 2022;13:3110-3132. [DOI] [PubMed] [Google Scholar]
- 53. Onygeme-Okerenta BM, Minadoki MD, Amadi BA. Phytochemical composition and antiplasmodial potential of fruit extract of Hunteria umbellata on chloroquine-sensitive plasmodium berghei berghei (NK65)-infected Albino Mice. Asian J Emerg. Res. 2023;5:9-21. [Google Scholar]
- 54. Laribi B, Kouki K, M’Hamdi M, Bettaieb T. Coriander (Coriandrum sativum L.) and its bioactive constituents. Fitoterapia. 2015;103:9-26. [DOI] [PubMed] [Google Scholar]
- 55. Sung WS, Jung HJ, Lee IS, Kim HS, Lee DG. Antimicrobial effect of furaneol against human pathogenic bacteria and fungi. J Microbiol Biotechnol. 2006;16:349-354. [Google Scholar]
- 56. Liu Y, Yang K, Jia Y, Shi J, Tong Z, Wang Z. Thymine sensitizes Gram-negative pathogens to antibiotic killing. Front Microbiol. 2021;12:622798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Fujisawa S, Ishihara M, Murakami Y, Atsumi T, Kadoma Y, Yokoe I. Predicting the biological activities of 2-methoxyphenol antioxidants: effects of dimers. In Vivo. 2007;21:181-188. [PubMed] [Google Scholar]
- 58. Ambati R, Phang SM, Ravi S, Aswathanarayana R. Astaxanthin: sources, extraction, stability, biological activities and its commercial applications—a review. Mar Drugs. 2014;12:128-152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Mehani M, Segni L, Terzi V, et al. Antibacterial, antifungal activity and chemical composition study of essential oil of Mentha pepirita from the south Algerian. Der Pharma Chemica. 2015;7:382-387. [Google Scholar]
- 60. Chen Z, Liu Q, Zhao Z, et al. Effect of hydroxyl on antioxidant properties of 2,3-dihydro-3,5-dihydroxy-6-methyl-4 H -pyran-4-one to scavenge free radicals. RSC Adv. 2021;11:34456-34461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Zamakshshari NH, Ahmed IA, Didik NAM, Nasharuddin MNA, Hashim NM, Abdullah R. Chemical profile and antimicrobial activity of essential oil and methanol extract from peels of four Durio zibethinus L. varieties. Biomass Convers Biorefinery. 2023;13:13995-14003. [Google Scholar]
- 62. Montague W, Taylor KW. The role of the pentose phosphate pathway in insulin secretion. In: Falkmer S, Hellman B, Täljedal IB, eds. The Structure and Metabolism of the Pancreatic Islets. Pergamon Press; 1970:263-273. [Google Scholar]
- 63. Mondello F, Fontana S, Scaturro M, et al. Terpinen-4-ol, the main bioactive component of Tea Tree Oil, as an innovative antimicrobial agent against Legionella pneumophila. Pathogens. 2022;11:682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Khaleel C, Tabanca N, Buchbauer G. α-terpineol, a natural monoterpene: a review of its biological properties. Open Chem. 2018;16:349-361. [Google Scholar]
- 65. Khodarahmi G, Asadi P, Hassanzadeh F, Khodarahmi E. Benzofuran as a promising scaffold for the synthesis of antimicrobial and antibreast cancer agents: a review. J Res Med Sci. 2015;20:1094-1104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Erwin, Pratiwi DR, Saputra I, Alimuddin. Antioxidant assay with scavenging DPPH radical of Artocarpus anisophyllus Miq Stem bark extracts and chemical compositions and toxisity evaluation for the most active fraction. Res J Pharm Technol. 2021;14:2819-2823. [Google Scholar]
- 67. Zhao L, Chen J, Su J, et al. In vitro antioxidant and antiproliferative activities of 5-hydroxymethylfurfural. J Agric Food Chem. 2013;61:10604-10611. [DOI] [PubMed] [Google Scholar]
- 68. Bouyahya A, Mechchate H, Benali T, et al. Health benefits and pharmacological properties of carvone. Biomolecules. 2021;11:1803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Naksing T, Teeka J, Rattanavichai W, Pongthai P, Kaewpa D, Areesirisuk A. Determination of bioactive compounds, antimicrobial activity, and the phytochemistry of the organic banana peel in Thailand. J Biosci. 2021;37:1981-3163. [Google Scholar]
- 70. Faizan M, Tonny SH, Afzal S, et al. β-cyclocitral: emerging bioactive compound in plants. Molecules. 2022;27:6845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Marchese A, Orhan IE, Daglia M, et al. Antibacterial and antifungal activities of thymol: a brief review of the literature. Food Chem. 2016;210:402-414. [DOI] [PubMed] [Google Scholar]
- 72. Jeong JB, Jeong HJ. 2-Methoxy-4-vinylphenol can induce cell cycle arrest by blocking the hyper-phosphorylation of retinoblastoma protein in benzo[a]pyrene-treated NIH3T3 cells. Biochem Biophys Res Commun. 2010;400:752-757. [DOI] [PubMed] [Google Scholar]
- 73. Islam MT, Ali ES, Uddin SJ, et al. Phytol: a review of biomedical activities. Food Chem Toxicol. 2018;121:82-94. [DOI] [PubMed] [Google Scholar]
- 74. Shamsizadeh A, Roohbakhsh A, Ayoobi F, Moghaddamahmadi A. Chapter 25 - the role of natural products in the Prevention and treatment of Multiple Sclerosis. In: Watson RR, Killgore WDS, eds. Nutrition and Lifestyle in Neurological Autoimmune Diseases. Academic Press; 2017:249-260. [Google Scholar]
- 75. Tamer CE, Suna S, Özcan-Sinir G. 14 - Toxicological aspects of ingredients used in nonalcoholic beverages. In: Grumezescu AM, Holban AM, eds. Non-alcohol. Woodhead Publishing; 2019:441-481. [Google Scholar]
- 76. Russo EB, Marcu J. Chapter three - cannabis pharmacology: the usual suspects and a few promising leads. Adv Pharmacol. 2017;80:67-134. [DOI] [PubMed] [Google Scholar]
- 77. Habibah A, Zahira G, Angga M, Nishfatul S, Rokhim D. Schematic literature review: content of Α-farnesene compounds as an anti-virus. Manganite: J Chem Edu. 2023;2:24-29. [Google Scholar]
- 78. Dubey MK, Aamir M, Kaushik MS, et al. PR toxin – biosynthesis, genetic regulation, toxicological potential, prevention and control measures: overview and challenges. Front Pharmacol. 2018;9:288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Mazumder K, Nabila A, Aktar A, Farahnaky A. Bioactive variability and in vitro and in vivo antioxidant activity of unprocessed and processed flour of nine cultivars of Australian lupin species: a comprehensive substantiation. Antioxidants. 2020;9:282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Choi J, Shin K-M, Park H-J, et al. Anti-inflammatory and antinociceptive effects of sinapyl alcohol and its glucoside syringin. Planta Med. 2004;70:1027-1032. [DOI] [PubMed] [Google Scholar]
- 81. Frapporti G, Colombo E, Ahmed H, et al. Squalene-based nano-assemblies improve the pro-autophagic activity of trehalose. Pharmaceutics. 2022;14:862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Al-Otaibi SN, Alshammari GM, AlMohanna FH, Al-Khalifa AS, Yahya MA. Antihyperlipidemic and hepatic antioxidant effects of leek leaf methanol extract in high fat diet-fed rats. Life. 2020;13:373-385. [Google Scholar]
- 83. Rossellia S, Maggio A, Formisano C, et al. Chemical composition and antibacterial activity of extracts of Helleborus bocconei ten. subsp. intermedius. Nat Prod Commun. 2007;2:1934578X0700200611. [Google Scholar]
- 84. Ali HAM, Imad HH, Salah AI. Analysis of bioactive chemical components of two medicinal plants (Coriandrum sativum and Melia azedarach) leaves using gas chromatography-mass spectrometry (GC-MS). Afr J Biotechnol. 2015;14:2812-2830. [Google Scholar]
- 85. Habib MR, Karim MR. Antimicrobial and cytotoxic activity of Di-(2-ethylhexyl) phthalate and anhydrosophoradiol-3-acetate isolated from Calotropis gigantea (Linn.) flower. Mycobiology. 2009;37:31-36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Kim SK, Karadeniz F. Chapter 14 - biological importance and applications of squalene and squalane. In: Kim SK, ed. Advances in Food and Nutrition Research. Academic Press;2012:223-233. [DOI] [PubMed] [Google Scholar]
- 87. Soliman M, Mohamed G, Mohamed E. Metal complexes of fenoterol drug: preparation, spectroscopic, thermal, and biological activity characterization. J Therm Anal Calorim. 2009;99:639-647. [Google Scholar]
- 88. Sánchez-Illana JD, Piñeiro-Ramos V, Ramos-Garcia I, Ten-Doménech M, Vento J, Kuligowski. Chapter three - oxidative stress biomarkers in the preterm infant. In: Makowski GS, ed. Advances in Clinical Chemistry. Elsevier; 2021:127-189. [DOI] [PubMed] [Google Scholar]
- 89. Mansourabadi AH, Sadeghi H, Razavi N, Rezvani E. Anti-inflammatory and analgesic properties of salvigenin, Salvia officinalis flavonoid extracted. Adv Herb Med. 2015;1:31-41. [Google Scholar]
- 90. Hung W-L, Chang W-S, Lu W-C, et al. Pharmacokinetics, bioavailability, tissue distribution and excretion of tangeretin in rat. J Food Drug Anal. 2018;26:849-857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Li S, Wang H, Guo L, Zhao H, Ho C-T. Chemistry and bioactivity of nobiletin and its metabolites. J Funct Foods. 2014;6:2-10. [Google Scholar]
- 92. García-Moreno M, Moreno-Conesa M, Rodríguez-López JN, García-Cánovas F, Varón R. Oxidation of 4-tert-butylcatechol and dopamine by hydrogen peroxide catalysed by Horseradisch peroxidase. Biol Chem. 1999;380:689-694. [DOI] [PubMed] [Google Scholar]
- 93. Hossain MJ, Lema KR, Samadd MA, Aktar R, Rashid MA, Al-Mansur MA. Chemical profiling and antioxidant, Anti-Inflammatory, cytotoxic, analgesic, and antidiarrheal activities from the seeds of commonly available red grape (Vitis vinifera L). Nutr Metab Insights. 2024;17:11786388241275100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Mamadalieva NZ, Hussain H, Xiao J. Recent advances in genus Mentha: phytochemistry, antimicrobial effects, and food applications. Food Front. 2020;1:435-458. [Google Scholar]
- 95. Bayeux MC, Fernandes AT, Foglio MA, Carvalho JE. Evaluation of the antiedematogenic activity of artemetin isolated from Cordia curassavica DC. Asian J Med Biol Res. 2002;35:1229-1232. [DOI] [PubMed] [Google Scholar]
- 96. Han Jie L, Jantan I, Yusoff SD, Jalil J, Husain K. Sinensetin: an insight on its pharmacological activities, mechanisms of action and toxicity. Front Pharmacol. 2021;11:553404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Guo S, Wu X, Zheng J, Song M, Dong P, Xiao H. Anti-inflammatory property of 5-Demethylnobiletin (5-hydroxy-6, 7, 8, 3’, 4’-pentamethoxyflavone) and its metabolites in lipopolysaccharide (LPS)-Induced RAW 264.7 cells. Biology. 2022;11:1820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Kandsi F, Elbouzidi A, Lafdil FZ, et al. Antibacterial and antioxidant activity of Dysphania ambrosioides (L.) Mosyakin and clemants essential oils: experimental and computational approaches. Antibiotics. 2022;11:482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Kandsi F, Lafdil FZ, Elbouzidi A, et al. Evaluation of acute and subacute toxicity and LC-MS/MS compositional alkaloid determination of the hydroethanolic extract of Dysphania ambrosioides (L.) Mosyakin and clemants flowers. Toxins. 2022;14:475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Pires DE, Blundell TL, Ascher DB. Ascher, pkCSM: predicting small-molecule pharmacokinetic and toxicity properties using graph-based signatures. J Med Chem. 2015;58:4066-4072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. OECD. Test No. 423: Acute Oral toxicity - Acute Toxic Class Method. Organisation for Economic Co-operation and Development; 2002. Accessed June 5, 2024. https://www.oecd.org/en/publications/test-no-423-acute-oral-toxicity-acute-toxic-class-method_9789264071001-en.html [Google Scholar]
- 102. Hussain H, Mamadalieva NZ, Hussain A, et al. Fruit peels: food waste as a valuable source of bioactive natural products for drug discovery. Curr Issues Mol Biol. 2022;44:1960-1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Liu J, Liu C, Rong Y, Huang G, Rong L. Extraction of limonin from Orange (Citrus reticulata Blanco) seeds by the Flash extraction method. Solvent Extr Res Dev Jpn. 2012;19:137-145. [Google Scholar]
- 104. Nakajima A, Nemoto K, Ohizumi Y. An evaluation of the genotoxicity and subchronic toxicity of the peel extract of Ponkan cultivar ‘Ohta ponkan’ (Citrus reticulata Blanco) that is rich in nobiletin and tangeretin with anti-dementia activity. Regul Toxicol Pharmacol. 2020;114:104670. [DOI] [PubMed] [Google Scholar]
- 105. Tahsin T, Wansi JD, Al-Groshi A, et al. Cytotoxic properties of the stem bark of Citrus reticulata Blanco (Rutaceae): cytotoxic property of stem bark of citrus reticulata Blanco. Phytother Res. 2017;31:1215-1219. [DOI] [PubMed] [Google Scholar]
- 106. Kaushal S, Kalia A, Kaur V. Proximate, mineral, chemical composition, antioxidant and antimicrobial potential of dropped fruits of citrus reticulata Blanco. J Food Meas Charact. 2022;16:4303-4317. [Google Scholar]
- 107. Mizuno H, Yoshikawa H, Usuki T. Extraction of nobiletin and tangeretin from peels of Shekwasha and Ponkan using [C 2 mim][(MeO)(H)PO 2 ] and centrifugation. Nat Prod Commun. 2019;14:1934578X1984581. [Google Scholar]
- 108. Yaqoob M, Aggarwal P, Babbar N. Extraction and screening of kinnow (Citrus reticulata L.) peel phytochemicals, grown in Punjab, India. Biomass Convers Biorefinery. 2023;13:11631-11643. [Google Scholar]
- 109. Uddin TM, Chakraborty AJ, Khusro A, et al. Antibiotic resistance in microbes: history, mechanisms, therapeutic strategies and future prospects. J Infect Public Health. 2021;14:1750-1766. [DOI] [PubMed] [Google Scholar]
- 110. Das R, Rauf A, Mitra S, et al. Therapeutic potential of marine macrolides: an overview from 1990 to 2022. Chem Biol Interact. 2022;365:110072. [DOI] [PubMed] [Google Scholar]
- 111. Kapoor G, Saigal S, Elongavan A. Action and resistance mechanisms of antibiotics: a guide for clinicians. J Anaesthesiol Clin Pharmacol. 2017;33:300-305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Yao X, Zhu X, Pan S, et al. Antimicrobial activity of nobiletin and tangeretin against Pseudomonas. Food Chem. 2012;132:1883-1890. [Google Scholar]
- 113. Jayaprakasha GK, Negi PS, Sikder S, Mohanrao LJ, Sakariah KK. Antibacterial activity of citrus reticulata peel extracts. Z Für Naturforschung C. 2000;55:1030-1034. [DOI] [PubMed] [Google Scholar]
- 114. Memariani Z, Moeini R, Hamedi SS, Gorji N, Mozaffarpur SA. Medicinal plants with antithrombotic property in Persian medicine: a mechanistic review. J Thromb Thrombolysis. 2018;45:158-179. [DOI] [PubMed] [Google Scholar]
- 115. Gupta N, Zhao Y-Y, Evans CE. The stimulation of thrombosis by hypoxia. Thromb Res. 2019;181:77-83. [DOI] [PubMed] [Google Scholar]
- 116. Chakraborty AJ, Uddin TM, Matin Zidan BMR, et al. Allium cepa: a treasure of bioactive phytochemicals with prospective health benefits. Evid Based Complement Alternat Med. 2022;2022:4586318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Lu WJ, Lin KC, Liu CP, et al. Prevention of arterial thrombosis by nobiletin: in vitro and in vivo studies. J Nutr Biochem. 2016;28:1-8. [DOI] [PubMed] [Google Scholar]
- 118. Vaiyapuri S, Ali MS, Moraes LA, et al. Tangeretin regulates platelet function through inhibition of phosphoinositide 3-kinase and cyclic nucleotide signaling. Arterioscler Thromb Vasc Biol. 2013;33:2740-2749. [DOI] [PubMed] [Google Scholar]
- 119. Marder VJ. Recombinant streptokinase: opportunity for an improved agent. Blood Coagul Fibrinolysis. 1993;4:1039-1040. [PubMed] [Google Scholar]
- 120. Wu D-H, Shi G-Y, Chuang W-J, et al. Coiled coil region of streptokinase γ-domain is essential for plasminogen activation. J Biol Chem. 2001;276:15025-15033. [DOI] [PubMed] [Google Scholar]
- 121. Shinde UA, Phadke AS, Nair AM, Mungantiwar AA, Dikshit VJ, Saraf MN. Membrane stabilizing activity—a possible mechanism of action for the anti-inflammatory activity of Cedrus deodara wood oil. Fitoterapia. 1999;70:251-257. [Google Scholar]
- 122. Jin J, Lv X, Wang B, et al. Limonin inhibits IL-1β-Induced inflammation and catabolism in chondrocytes and ameliorates osteoarthritis by activating Nrf2. Oxid Med Cell Longev. 2021;2021:e7292512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Witschi HP. Enhanced tumour development by butylated hydroxytoluene (BHT) in the liver, lung and gastro-intestinal tract. Food Chem Toxicol. 1986;24:1127-1130. [DOI] [PubMed] [Google Scholar]
- 124. Oguntibeju OO. Medicinal plants with anti-inflammatory activities from selected countries and regions of Africa. J Inflamm Res. 2018;11:307-317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Sarmento-Neto J, Do Nascimento L, Felipe C, De Sousa D. Analgesic potential of essential oils. Molecules. 2015;21:E20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Nirmal SA, Pal SC, Mandal SC, Patil AN. Analgesic and anti-inflammatory activity of β-sitosterol isolated from Nyctanthes arbortristis leaves. Inflammopharmacology. 2012;20:219-224. [DOI] [PubMed] [Google Scholar]
- 127. Zewdie KA, Bhoumik D, Wondafrash DZ, Tuem KB, Tuem KB. Evaluation of in-vivo antidiarrhoeal and in-vitro antibacterial activities of the root extract of Brucea antidysenterica JF mill (Simaroubaceae). BMC Complement Med Ther. 2020;20:11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Naher S, Aziz MA, Akter MI, Rahman SMM, Sajon SR, Mazumder K. Anti-diarrheal activity and brine shrimp lethality bioassay of methanolic extract of Cordyline fruticosa (L.) A. Chev. leaves. Clin Phytoscience. 2019;5:15. [Google Scholar]
- 129. Panganamala RV, Miller JS, Gwebu ET, Sharma HM, Cornwell DG. Differential inhibitory effects of vitamin E and other antioxidants on prostaglandin synthetase, platelet aggregation and lipoxidase. Prostaglandins. 1977;14:261-271. [DOI] [PubMed] [Google Scholar]
- 130. Cajas YN, Cañón-Beltrán K, Ladrón de, Guevara M, et al. Antioxidant nobiletin enhances oocyte maturation and subsequent embryo development and quality. Int J Mol Sci. 2020;21:5340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Wang M, Meng D, Zhang P, et al. Antioxidant protection of nobiletin, 5-demethylnobiletin, tangeretin, and 5-demethyltangeretin from citrus peel in Saccharomyces cerevisiae. J Agric Food Chem. 2018;66:3155-3160. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Supplemental material, sj-pdf-1-nmi-10.1177_11786388251327668 for Phytochemical Isolation and Antimicrobial, Thrombolytic, Anti-inflammatory, Analgesic, and Antidiarrheal Activities from the Shell of Commonly Available Citrus reticulata Blanco: Multifaceted Role of Polymethoxyflavones by Md. Jamal Hossain, Md. Abdus Samadd, Mst. Nusrat Zahan Urmi, Mst. Farzana Yeasmin Reshmi, Md. Shohel Hossen and Mohammad A. Rashid in Nutrition and Metabolic Insights