Next Article in Journal
Oxidative Stress and Inflammatory Mediators in Exhaled Breath Condensate of Patients with Pulmonary Tuberculosis. A Pilot Study with a Biomarker Perspective
Next Article in Special Issue
Phenolic Analysis and In Vitro Biological Activity of Red Wine, Pomace and Grape Seeds Oil Derived from Vitis vinifera L. cv. Montepulciano d’Abruzzo
Previous Article in Journal
Mechanistic Aspects of Apiaceae Family Spices in Ameliorating Alzheimer’s Disease
Previous Article in Special Issue
Noble 3,4-Seco-triterpenoid Glycosides from the Fruits of Acanthopanax sessiliflorus and Their Anti-Neuroinflammatory Effects
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

LC-MS Based Analysis and Biological Properties of Pseudocedrela kotschyi (Schweinf.) Harms Extracts: A Valuable Source of Antioxidant, Antifungal, and Antibacterial Compounds

by
Kouadio Ibrahime Sinan
1,*,
Stefano Dall’Acqua
2,*,
Irene Ferrarese
2,
Adriano Mollica
3,
Azzurra Stefanucci
3,
Jasmina Glamočlija
4,
Marina Sokovic
4,
Marija Nenadić
4,
Abdurrahman Aktumsek
1 and
Gokhan Zengin
1,*
1
Physiology and Biochemistry Research Laboratory, Department of Biology, Science Faculty, Selcuk University, Konya 42130, Turkey
2
Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131 Padova, Italy
3
Department of Pharmacy, G. d’Annunzio University of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
4
Laboratory of Mycology, Department of Plant Physiology, Institute for Biological Research “Siniša Stanković”, National Institute of Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia
*
Authors to whom correspondence should be addressed.
Antioxidants 2021, 10(10), 1570; https://doi.org/10.3390/antiox10101570
Submission received: 11 September 2021 / Revised: 29 September 2021 / Accepted: 30 September 2021 / Published: 2 October 2021

Abstract

:
The impact of two extraction solvents on the phenolic composition, antioxidant, and enzymes inhibitory and antimicrobial activities of two parts (leaves and stem bark) of P. kotschyi was studied. Two different LC-DAD-MSn approaches were used to identify and quantify the bioactive compounds in the different extracts. A total of thirty-two compounds were quantified, being the procyanidin the most abundant in stem bark while catechin and flavonoids are most abundant in leaves. Overall, the stem bark extraction using methanol showed higher amounts of total phenolic (131.83 ± 1.81 mg GAE/g) and flavanol (14.14 ± 0.11 mg CE/g) while the leaves extraction using water exhibited stronger levels of total flavonoid (44.95 ± 0.38 mg RE/g) and phenolic acid (63.58 ± 2.00 mg CAE/g). As regards the antioxidant assays, methanol stem bark extracts were characterized by the highest antioxidant activities (DPPH: 1.94 ± 0.01 mmol TE/g, ABTS: 3.31 ± 0.01 mmol TE/g, FRAP: 2.86 ± 0.02 mmol TE/g, CUPRAC: 5.09 ± 0.08 mmol TE/g, phosphomolybdenum: 5.16 ± 0.23 mmol TE/g and metal chelating: 17.12 ± 0.46 mg EDTAE/g). In addition, the methanolic extracts of stem bark had highest impact on acetylcholinesterase (2.54 mg GALAE/g), butyrylcholinesterase (5.48 mg GALAE/g). In contrast, the methanolic extracts of leaves was potent against tyrosinase (77.39 ± 0.21 mg KAE/g) and α-glucosidase (0.97 ± 0.01 mmol ACAE/g), while a higher anti-α–amylase (0.97 ± 0.01 mmol ACAE/g) was observed for water extracts of the same part. All of the tested extracts showed inhibitory effects on elastase, except methanolic leaves extracts. Additionally, the extracts exhibited appreciable antifungal toward A. ochraceus, A. fumigatus, P. ochrochloron, T. viride, and P. funiculosum and promising antibacterial activity against M. flavus, S. aureus, L. monocytogenes, E. coli, P. aeruginosa, E. cloacae, and S. typhimurium. Taken together, the outcomes demonstrated P. kotschyi as a novel source of bioactive molecules of interest with an evident therapeutic value.

1. Introduction

Herbal based medicines and remedies have a lengthy history of use on all continents for the therapy of several complaints and diseases. Medicinal plants are highly diffused, both for the traditional medicines, as well as for the development of new treatments, and scientific studies assessing their composition and efficacy are needed. The available knowledge on the utilization of herbals preparations in folk medicine is large but still too empiric. The evaluation of the bioactivities of complex mixtures as extracts can be a challenge, and the difference in chemical composition can seriously afflict final effects. Many herbal medicines used in traditional preparation are only poorly studied for their chemical content and for their potential usefulness in therapy. On the other hand, the large amount of traditional knowledge that is present in many countries about the medicinal plant is still orally transmitted and there is a risk of losing information. For these reasons, scientific studies on traditional species are urgently needed to evaluate these botanicals as sources of bioactive compounds, as well as to substantiate their importance in the traditional medicine, finally improved knowledge on species, both from a chemical and pharmacological point of view can help biodiversity protection and increase the value of each country’s botanical diversity. Africa is reputed for the exceptional richness of its flora. Based on rigorous ethnobotanical investigations, together with judicious selection of plants, this large botanical biodiversity can be considered a rich source of new natural bioactive compounds being good candidates for the preparation of new drugs. It is well known the importance of the whole natural product in drug discovery and development [1,2]. In this regard, plant derived natural products still play a crucial role [3,4].
Different classes of secondary metabolites present significant pharmacological activities and, thus, to assess the potential bioactivity of botanical species detailed chemical characterization is needed. Secondary metabolite production is needed for plant protection and relationships with other organisms, such as herbivores, phytofages, insects, furthermore, secondary metabolites can help plants in protection due to abiotic or biotic stresses. As example, the protection role of plant chemical compounds, such as polyphenols, against different disorders has been related in several publications [5,6], and numerous new plant derived drugs increased likewise.
The Meliaceae, one of the most studied families, is rich sources of structurally diversified limonoid [7]. This group of molecules draw great interest due to their pharmaceutical properties [8,9,10]. Amongst approximately 1400 species in this family [11], our attention was focused on the species Pseudocedrela kotschyi. P. kotschyi is used in different parts of Africa by traditional healers in treatment of epilepsy, diabetes, diarrhea, rheumatism, aphrodisiac, anemia, and menstruation disorder [12,13]. Some of these properties have been highlighted in several scientific studies. For example, the antibacterial, anticonvulsant, antidiabetic, antifungal, antimalarial, anti-inflammatory, anti-analgesic, antipyretic, antiulcer, antidiarrhoeal, and anticancer activities have been reported [13,14,15,16,17,18,19]. At the same time, the phytochemical screening revealed that roots, stem bark, or leaves contain flavonoids, tannins, saponins, terpenoids, steroids, anthraquinones, and alkaloids [13,20]. In addition, some compounds, such as pseudocedrelin, quercetin, pseudrelones A, B, C, myricetin, stigmasterol, and kotschyins A-C, have been identified in different extracts of the roots, stem bark, or leaves [17,21,22,23,24,25].
As can be noted, the number of publications have been reported on the potential pharmaceutical properties of P. kotschyi. Nevertheless, limited studies carrying on both leaves and stem bark have been done. Likewise, no reports are available on the effects of different extraction solvents on the nature and amount of phytochemicals extracted and the pharmaceutical activities. Generally, when plant-derived phytochemicals or pharmaceutical activities are studied, the solvents used for extraction, as well as the parts must be carefully considered. For these reasons, in this work, two extraction solvents (aqueous vs. methanol) and parts (leaves vs. stem bark) were comparatively evaluated according to their antioxidant, enzyme inhibitory, and antimicrobial activities. LC-MSn analysis was also carried out to determine the phytochemicals composition of the extracts. From our point of view, this is the first attempt that deals with an exhaustive analysis of secondary metabolites extracted from stem bark and leaves of P. kotschyi collected in Côte d’Ivoire.

2. Materials and Methods

2.1. Plant Materials

The collecting of the leaves and stem bark of P. kotschyi were provided in the village of Prikro (city of Brobo, Côte d’Ivoire), in January 2019. The plant was taxonomically identified by a botanist from the national floristic center (Université Félix Houphouet Boigny, Abidjan, Côte d’Ivoire) and representative specimen was deposited at the herbarium. The two parts were dried at 25 °C temperature for two weeks. Then, they were milled in a laboratory and the obtained thin powder were preserved in room temperature until analysis.

2.2. Preparation of Extracts

Maceration: 5 g of leaves or stem bark was extracted with 100 mL of methanol, brought to room temperature and stirred for 24 h. Resulting extracts were filtered and the solvent were evaporated through the use of rotary-evaporator. Infusion: 100 mL boiled water was added to 5 g of each parts. After 20 min the obtained solutions were filtered and lyophilized. All extracts were kept at 4 °C until the further analysis.

2.3. Determination of Secondary Metabolites

For each extract, different spectrophotometrical tests, were carried out to quantify the flavonoid, phenolic acid, flavanol, and phenolic compounds. The details of the different methods are available in our previous manuscripts [26,27]. Rutin (RE), caffeic acid (CE), catechin (CAE), or gallic acid (GAE) were used as standards and results were expressed as milligrams RE, CE, CAE, or GAE per grams dry matter, respectively.

2.4. LC-MSn Analysis

Samples were analyzed using an LC-DAD-MSn system. Briefly, leaves extracts, and stem bark extracts were weighted and extracted (20 mg/mL methanol) and sonicated for 10 min. Samples were centrifuged at 13,000 rpm for 15 min and liquids were used for LC analysis. The LC-MSn system consisted of an Agilent 1260 quaternary pump coupled to both 1260 Agilent diode array detector (DAD) and a Varian MS 500 Mass spectrometer equipped with Electrospray (ESI) ion. MS data were acquired both in positive and negative ion mode, in the m/z range 100 to 2000. Fragmentation pattern of most intense ion species was obtained using the turbo data depending on scanning (TDDS) function of the instrument. MS parameters were as follows: needle voltage, 4.9 kV; shield voltage, 600 V; capillary voltage, 80 V; RF loading, 80%; nebulizing gas pressure, 25 psi (nitrogen); drying gas pressure, 15 psi; drying gas temperature, 300 °C. Identification of compounds was obtained based on comparison with the literature and reference compounds, when available.
The sample solutions were centrifugated for 10 min, then transferred in vial for analysis. An Agilent Eclipse XDB-C18 column (3.0 mm × 150 mm, 3.5 µm) was used as stationary phase.
For mobile phase solvents were water 1% formic acid (A), Acetonitrile (B), and methanol (C). Gradients start with 98% A and 2% B. In 5 min, 90% A, 8% B, and 2% C, then, at 20 min, 70% A, 28% B, and 2% C isocratic up to 25 min. In 5 min, 80% B and 20% C isocratic up to 34 min. The flow rate was 400 µL/min. Injection volume was 10 µL and the temperature was set at 30 °C. Identification of compounds was obtained based on comparison with the literature and reference compounds, when available. Quinic acid, gallic acid, rutin, quercetin 3-O-glucoside, procyanidin B2 were used as external standard and for compound quantification a calibration curve were performed: for rutin at 350 nm and calibration curve was y = 18.03x + 21.1 (R2 = 0.9991), for gallic acid at 280 nm and calibration curve was y = 5.03 x + 0.2 (R2 = 0.9989), for procyanidin B2 at 280 nm y = 4.1x + 0.34 (R2 = 0.9992).
Accurate m/z values were obtained using a Waters Acquity UPLC system coupled to a Waters Xevo G2 Q-TOF MS detector, operating in ESI (-) mode. For chromatographic separation, an Agilent Eclipse plus C18 column (2.1 × 50 mm, 1.8 µm) was used as stationary phase, and a gradient mixture of methanol (A) and 0.1% formic acid in water (B) as mobile phase. The gradient was: 0 min, 2% A; 0.75 min, 2% A; 11 min, 100% A; 13.5 min, 100% A; 14 min, 2% A and isocratic up to 15 min. Flow rate was 0.4 mL/min. MS parameters were as follows: sampling cone voltage, 40 V; source offset, 80 V; capillary voltage, 3500 V; nebulizer gas (N2) flow rate, 800 L/h; desolvation temperature, 450 °C. The mass accuracy and reproducibility were maintained by infusing lockmass (leucine-enkephalin, [M–H] = 554.2620 m/z) thorough Lockspray at a flow rate of 20 μL/min. Centroided data were collected in the m/z range 50 to 1200, and the m/z values were automatically corrected during acquisition using lockmass.

2.5. Determination of Antioxidant Capacity

By referring to Uysal et al. [28], the radical scavenging and reducing power abilities of the extracts were evaluated using ABTS, DPPH, CUPRAC, and FRAP. Likewise, ferrous ion chelating and phosphomolybdenum experiments were performed. The results were given as standard molecules equivalents of Trolox (TE) or ethylenediaminetetraacetic acid (EDTA), respectively.

2.6. Determination of Enzyme Inhibitory Effect

The ability of extracts to inhibit effects of a panel of key enzymes, such as cholinesterases, α-glucosidase, α-amylase, and tyrosinase were investigated according to the protocol described by Grochowski et al. [29]. The enzyme inhibitory activities of extracts were expressed as equivalents of acarbose for α-glucosidase, α-amylase, galatamine for cholinesterases, kojic acid for tyrosinase, and catechin for elastase.

2.7. Antimicrobial Activities

To determine antimicrobial activities of P. reticulatus extracts, microdilution method was performed as done in our earlier paper. In the experiments, Gram-positive (Staphylococcus aureus (ATCC 6538), Listeria monocytogenes (NCTC 7973), and Bacillus cereus (clinical isolate)) and Gram-negative (Salmonella Typhimurium (ATCC 13311), Pseudomonas aeruginosa (ATCC 27853), Enterobacter cloacae (human isolate), and Escherichia coli (ATCC 35210)) bacteria were used. The tested bacteria were obtained from the Institute for Biological Research “Sinisa Stankovic”, Belgrade, Serbia.
Regarding anti-fungal properties, different fungal strains were selected, and they were Aspergillus fumigatus (human isolate), Aspergillus ochraceus (ATCC 12066), A. niger (ATCC 6275), A. versicolor (ATCC 11730) can be considered as significant for aspergilloses. Trichoderma viride (IAM 5061), Penicillium funiculosum (ATCC 36839) are attacking plant and especially pineapples, P. ochrochloron (ATCC 9112), and P. verrucosum var. cyclopium (food isolate) are contaminant of foods, especially meat products.
Obtained results from antibacterial and antifungal assays were evaluated the minimum inhibitory concentration (MIC) and the minimum bactericidal/fungicidal concentration (MBC/MFC) values. In antibacterial activity, streptomycin and ampicillin were selected as standards (1 mg/mL). In antifungal activity, ketoconazole and bifonazole were selected as standards (0.10–0.35 mg/mL).

2.8. Computational Tests

2.8.1. Receptor Preparation

The crystal structure of the enzyme tyrosinase used for the in silico tests has been downloaded from the Protein Data Bank RCSB PDB [30]; Tyrosinase, pdb id: 2Y9X, [31] in complex with tropolone. The raw structure has been polished with UCSF Chimera [32] by removing additional chain, and the B chain was used as crystallographic protein complex. The protein was prepared with the Protein Preparation Wizard [33] implemented in Maestro suite 11.1 [34], by filling missing side chain and loops e-converting seleno-methionines and seleno-cysteines to methionines and cysteines, respectively, generating heteroatoms states at pH 7.4 using Prime [35], using a well-established procedure previously reported by our research group [36,37,38,39] then deleting non-catalytic waters and other co-crystallized molecule, were conducted the H-bond assignment by using PROpKA at pH 7.4, and a minimization of only the hydrogens. The copper ions present in the catalytic pocket of tyrosinase, have been optimized by the automatic generation of optimized metal states. The protein was separated from the crystallographic ligand and used without further modifications for the docking experiments and self-docking validation process.

2.8.2. Self-Docking Validation

In order to assess the suitability of the software GOLD 5.5 [40] for the docking on tyrosinase, a procedure of self-docking was performed as reported previously by Stefanucci et al. [41]. At this stage, the software GOLD 5.5 was configured for self-docking involving the crystallographic tyrosinase inhibition-enzyme complex chain B (2Y9X), as described above. All the scoring functions of GOLD (ASP, PLP, GOLDSCORE, and CHEMSCORE) were considered to conduct the validation tests, performing a self-docking of the crystallographic ligands, and by comparing the root-mean-square deviation (RMSD) of the best docked crystallographic pose with the original crystallographic pose. An area of 15 Å around the co-crystallized ligand was defined as the binding site. At the end of the docking calculations, the ChemScore and GoldScore scoring functions returned with the lowest RMSD value. Both ASP and PLP methods were not able to find any interactions for tropolone with the copper atoms, thus have been discarded [42]. Finally, both Goldscore and Chemscore were able to find a similar pose of tropolone superimposable to the crystallographic one with similar RMSD in the range of 2 to 3 angstroms. Considering that the GoldScore scoring function is defined as the most comprehensive scoring function embedded in GOLD docking software [43], this scoring function has been employed for the docking experiments in this work.

2.8.3. Ligand Preparation

The selected compounds (Table 1) were prepared with LigPrep tool [44] embedded in Maestro 11.1, neutralized at pH 7.4 by Ionizer and minimized with OPLS-3 force field [45] and used for the docking experiment without further modifications.

2.9. Molecular Docking

Dockings of the selected molecules (see Table 1) were performed on tyrosinase by using the software Gold 5.5, developed by the Cambridge University [40]. GoldScore scoring function was employed for the docking calculations. The docking grid was determined by a radius of 15 Angstroms around the crystallographic ligand center, similarly to the previous published works [46,47,48]. As results of the docking experiments, the two substances with the highest Fitness Score “GoldScore” were Prodelphynidin B3 with a score of 68.18 and Procyanidin B3 with a score of 66.74. The best pose for these latter compounds docked to tyrosinase are depicted in Figure 1.

2.10. Data Analysis

A one-way analysis of variance was carried out to investigate significant differences in phytochemical contents, antioxidant, enzyme inhibitory, antifungal, and antibacterial activities in the different extracts (p < 0.05, Turkey’s post hoc test). The computation of Pearson’s correlation coefficients allowed to analyze the relationship between the phytochemical contents and the antioxidant and enzyme inhibitory activities.
Afterwards, the dataset was scaled and subjected to multi-block discriminant analysis for the purpose of investigating the variation of the evaluated biological activities between the two parts, as well as the two extraction solvents. The sensibility and specificity of the two models was evaluated by calculating the AUC value. R v 3.6.2 statistical software was used for the analysis.

3. Results

3.1. Chemical Composition

The concentration of total phenolic, flavonoids, phenolic acid, and flavanol compounds of the different extracts was reported in Table 2. The content of phenolic and flavonoid ranged between 125.67 to 131.83 mg GAE/g and between 0.63 to 44.95 mg RE/g, respectively. Similarly, that of phenolic acid and flavanol varied from 24.88 to 6.58 mg CAE/g and from 5.99 to 14.14 mg CE/g, respectively. Both total phenolic and flavanol contents were maximum in the methanol stem bark extract (131.83 ± 1.81 mg GAE/g and 14.14 ± 0.11 mg CE/g, respectively). By contrast, the content of flavonoids and phenolic acids of the water leaves extract were higher than that of the other extracts (44.95 ± 0.38 mg RE/g and 63.58 ± 2.00 mg CAE/g, respectively).
Thirty-two different phenolic constituents were identified and quantified combining the data of the Q-Tof for the high-resolution mass spectrometry and MSn fragmentation pathways obtained using ion trap. Results are summarized in Table 3, and clearly revealed that both plant parts contain large amount of condensed tannins, mostly prodelphyinidine and procyanidins. Identified compounds were prodelphynidins, procyanidins [49,50], catechin and epicatechin, in bark, while flavonoids in leaves mostly myricetin, quercetin, and kaempferol derivatives [51,52,53]. Identification of compounds was obtained matching accurate mass measurements, fragmentation pathways, and literature [48,49,50,51,52], confirmation of some of the compound was also obtained by co-injection of reference standard. Considering phenolic constituents, the extraction with methanol for the plant part result more favorable. This result indicate a different extraction behavior of the flavonoid glycosides and procyanidin derivatives in water and in methanol, this result can be explained by the different solubility of the compounds in the two solvents, by the presence of other constituents that can interfere with the extraction and that are not measured, such as sugars, olygosaccharides, as well as lipid and waxes, as well as due to the different ability of the water and methanol to penetrate dried plant material.

3.2. In Vitro Antioxidant Properties and Enzyme Inhibitory Activities

Six different in vitro assays including ABTS and DPPH radical scavenging, Ferric and cupric ion reducing, phosphomolybdenum and ferrous ion chelating were used to assess the antioxidant capacity of the prepared extracts. Overall, the antioxidant capacity of methanol stem bark extract was significantly high than that of the other extracts (Table 4). In depth, these activities were enumerated as follows: DPPH: 1.94 ± 0.01 mmol TE/g, ABTS: 3.31 ± 0.01 mmol TE/g, FRAP: 2.86 ± 0.02 mmol TE/g, CUPRAC: 5.09 ± 0.08 mmol TE/g, phosphomolybdenum: 5.16 ± 0.23 mmol TE/g, and metal chelating: 17.12 ± 0.46 mg EDTAE/g. In addition, as seen in DPPH assay, methanol leaves extract also exhibited highest activity.
The inhibitory effects of the extracts against the six key enzymes were shown in Table 4. Among the extract, methanol stem bark exhibited a potent anti-AChE and anti-BChE activities, being 2.54 and 5.48 mg of galatamine acid equivalent/g of sample for AChE and BChE, respectively. Moreover, the anti-tyrosinase activity increase in the following order: leaves-methanol > stem bark-methanol > leaves-water > stem bark-water. Remarkably, except methanol leaves, it was found that all extracts possessed the same anti-elastase activity (leaves-water: 1.98 ± 0.10 mmol CE/g, stem bark-water: 2.08 ± 0.06 mmol CE/g, stem bark-methanol: 2.21 ± 0.12 mmol CE/g). Concerning both anti-hyperglycemia assays, water leaves, and methanol leaves extracts had the strongest anti-amylase (0.97 ± 0.01 mmol ACAE/g) and anti-glucosidase (0.97 ± 0.01 mmol ACAE/g) activities, respectively. No anti-glucosidase activity was observed in water extracts of both studied parts.
Afterward, the contribution of the phytochemical compounds to the antioxidant and enzymes inhibitory activity was determined. As shown in Figure 2, the antioxidant activity and metal chelating ability of P. kotschyi extracts seemed to be link to several phytochemical compounds by considering the Pearson’s coefficient being higher than 0.8. Similarly, both anti-cholinesterase, anti-glucosidase, anti-tyrosinase, and anti-elastase activities were bound with more than one compounds. Interestingly, Pk10, Pk14, Pk15, and Pk21 were significant positive correlation with all of the antioxidant activities (ABTS, DPPH, CUPRAC, FRAP, PPBD, and MCA), as well as anti-cholinesterase activities. Significant correlation was obtained between Amylase and a single compound (Pk32).

3.3. Antifungal and Antibacterial Activities

The antifungal and antibacterial effects of the extracts on the tested eight fungal strains are depicted in Table 5 and Table 6, respectively. The different extracts showed, overall, remarkable fungistatic and fungicidal, as well as bacteriostatic and bactericidal effects to variable extents, depending on the strain in question. Concerning the fungistatic effect, the results revealed that A. versicolor was more sensitive to leaves water and bifonazole, with a MIC value of 0.10 mg/mL. A. ochraceus was more susceptible to water and methanol extract of leaves (MIC = 0.10 mg/mL) as compared to remaining extracts and reference drugs. Though our mind that both extracts of stem bark (MIC = 0.15 mg/mL) were found to be more effective towards the two above mentioned strains (A. versicolor and A. ochraceus) than ketoconazole (MIC = 0.20 mg/mL). In the case of Aspregillus fumigatus, stem bark water had the lowest MIC value (0.11 mg/mL), while the MIC value of stem bark methanol and Bifonazole were equal (0.15 mg/mL). The lowest MIC value for Penicillium ochrochloron was obtained with water extracts of the two part (0.15 mg/mL). Both leaves extracts and stem bark water exhibited the best effectiveness toward P. funiculosum and T. viride than the two reference drugs. In addition, stem bark methanol and bifonazole had similar minimum inhibitory concentration for T. viride (0.15 mg/mL). The efficiency of water and methanol extracts of leaves against A. niger were comparable to that of ketoconazole (0.15 mg/mL); similarly, leaves water, methanol stem bark, water stem bark, and ketoconazole showed similar MIC value for P. verrucosum var. cyclopium. Regarding the fungicidal effect, some tested extracts effectively eliminate studied strains, at different concentrations, some of which were comparable to that of the controls. The best minimum fungicidal concentration (MFC) values were observed for A. fumigatus, P. ochrochloron, P. funiculosum, and T. viride. Indeed, A. fumigatus and T. viride was eliminated by stem bark water at 0.15 mg/mL. 0.21 mg/mL of leaves water allowed to kill P. ochrochloron while 0.20 mg/mL of leaves methanol and 0.21 mg/mL of leaves water were received sufficient to eliminate P. funiculosum. These concentrations were lower than the two reference drugs.
The best bacteriostatic and bactericidal effects for most of the studied bacterial strains was recorded by stem bark methanol. In depth stem bark methanol inhibit growth of M. flavus, S. aureus, L. monocytogenes, E. coli, P. aeruginosa, E. cloacae, and S. typhimurium at MIC value of 0.04, 0.04, 0.04, 0.11, 0.11, 0.03, 0.07 mg/mL, respectively. Similarly, M. flavus, S. aureus, L. monocytogenes, E. coli, P. aeruginosa, E. cloacae and S. typhimurium were found to be sensitive to stem bark methanol at MBC value of 0.07, 0.08, 0.08, 0.15, 0.16, 0.07, and 0.15, respectively. Otherwise, stem bark water exhibited excellent bacteriostatic and bactericidal activities against S. aureus (MIC = 0.03 mg/mL; MBC = 0.07 mg/mL), while leaves methanol remarkable bacteriostatic and bactericidal activities effect toward Listeria monocytogenes (MIC = 0.05 mg/mL, MBC = 0.20 mg/mL). It should be noted that the MIC and MBC values of above mentioned extracts were lower than the two controls. Additionally, the efficiency of some extracts to inhibit the growing or to kill several strains was comparable to one or both reference drugs; illustratively, leaves methanol-ampicillin-streptomycin showed equal MIC value for S. aureus (0.10 mg/mL) while leaves methanol- streptomycin had similar MBC value for the same strain (0.20 mg/mL). The same trend was observed with stem bark water and ampicillin for E. coli (MIC = 0.15 mg/mL), stem bark water, and both controls (MBC = 0.15 mg/mL) and those are just a few examples.
Thereafter, the relationship between phytochemical compounds and the evaluated antifungal and antibacterial activities was investigated. As seen from results (Figure 3 and Figure 4), the MIC and MFC activities of various fungal strains seemed due to the effect of multiple compounds (Pearson’s coefficient being higher than 0.8). Remarkably, the MFC and MIC of T. viride, the MIC of P. funiculosum and P. verrucosum var. cyclopium had significant positive correlation with a largest number of compounds. Contrary to anti-fungal activity, vary few significant positive correlation was found between anti-bacterial activities and chemical compounds. However, the bacteriostatic and bactericidal activities of S. typhimurium, as well as the bacteriostatic activity of E. coli seemed result from the action of a large number of chemical compounds.

3.4. Antioxidant, Enzyme Inhibitory, Antifungal, and Antibacterial Activities Variation among Extraction Solvents and Plant Parts

The results of multi-block discriminant analysis considering parts and extraction solvents as class membership criteria were graphically shown in Figure 5A. looking at the both scatterplots of samples, it is possible to view a quite clear separation among the two parts and extraction solvents, respectively, except the block “Antioxidant Properties” wherein no segregation between the leaves and stem bark was obtained (Figure 5A,B). For the first model (parts), 2 functions, were sufficient to discriminate the two parts for the block “Enzymes Inhibition” and “Antibacterial Activity” while 1 functions, was sufficient to separate the two parts for the block “Antifungal Activity” (Figure 6A). Regarding the block “Antioxidant Properties”, an AUC value of 0.63 was obtained considering 2 functions, suggesting no significant difference between both parts as regards the antioxidant activities (Figure 6A). Concerning the second model (extraction solvent), the optimal number of functions was found to be 1 for the all the blocks (Figure 6B).

3.5. Molecular Docking

A series of ten bioactive natural compounds, found in the extracts of P. kotschyi have been submitted to in silico docking experiments in order to evaluate their affinity for the enzymatic cavity of the enzyme tyrosinase. This enzyme was selected among the other tested ones due to the relevant anti-tyrosinase activity showed by the P. kotschyi extracts. Two substances, namely Procyanidine B3 and Prodelphynidin B3 have been found to have the higher Fitness score calculated by the scoring function GoldScore embedded in the software GOLD 5.5. The scoring function GoldScore ranks the docking results taking in consideration several parameters, it has been optimized for the prediction of ligand binding positions and takes into account factors such as H-bonding energy, van der Waals energy, metal interaction, and ligand torsion strain [54]. From the analysis of the best docking pose of procyanidin B3 it can be observed that the ligand-enzyme complex is stabilized by three hydrogen bonds respectively with Asn81, His85, and Gly280, also the molecule is able to form several π-π stacks to the aromatic side chains of His85, His244, His259. Additionally, the phenolic hydroxyl group of the Procyanidin B3 is able to form a coordination bond to Cu401, directly involved in the mechanism of the enzyme. The other best ranked substance was Prodelphinidin B3. The ligand-enzyme complex with the best pose obtained, is stabilized by two hydrogen bonds, respectively, toward Glu322 and Gly281, several π-π stacks with His85, His244, and His259. Additionally, this molecule was able to establish two coordinative bonds to both the copper atoms present in the enzymatic cavity.

4. Discussion

The antioxidant property of extracts derived from the leaves and stem bark of P. kotschyi was determined by different methods based on different mechanisms. In fact, because no single method can accurately reflects the mechanism of action of all antioxidant molecules contained in a plant extract, Tabart et al. [55] recommend the necessary achievement of different assays at once. Based on the experimental outcomes, the high antioxidant capacity was recorded with methanol stem bark extract, whatever the assay performed. A previous several in vitro studies on different extracts of P. kotschyi disclosed it potential antioxidant activity [16,56,57]. Apart from in vitro assays, the excellent antioxidant property of P. kotschyi was also demonstrated by various in vivo experiments. Indeed, Eleha et al. [58] reported that water leaves and methanol bark extracts of P. kotschyi possesses anti-oxidative effect, comparable to that of vitamin C, against paracetamol induced liver damage of rats. In addition, 48 hourly administration of the aqueous extract of P. kotschyi to pentylenetetrazole-induced kindled rats, allowed to reduce the progression of epileptogenesis through inhibition of oxidative stress [56]. Furthermore through LC-MSn analysis, several well-known antioxidant molecules were identified in the different extracts. The presence of prodelphynidin derivatives in large amount in leaves and bark extract, as well as epicatechin and catechin (Figure 7) can explain the observed antioxidant activity. Strong antioxidant activity is associated with the presence of two or three hydroxyl substituents in the “B” ring of the flavonoid skeleton [59,60,61,62]. Myricetin and quercetin derivatives also can contribute to the antioxidant activity of the leaves extract samples. We studied antioxidant activity of isolated kaempferol and quercetin glycosidic derivatives showing very high activities for quercetin-3-O-rhamnoside and rutin compared to Kempferol-7-O-β-glucopyranoside-3-O-rutinoside [63], as also confirmed by previous investigations comparing kaempferol and quercetin derivatives [62]. Based on the above findings, P. kotschyi leaves and stem bark appears clearly as a valuable source of antioxidant compounds that could replace synthetic antioxidants, such as tertiary butyl hydroquinone, butylated hydroxyanisole and butylated hydroxytoluene, since there have been reported to have health risk and toxicity [64].
Past few years, oxidative stress-related disorders caused by reactive oxygen species (ROS) have deserve special attention. In fact, ROS are generated as a natural by-product during the normal metabolism processes of oxygen and contributes to the protection for living organisms. Nevertheless, excessive production of ROS is detrimental to the body and result in oxidative stress which is involved in pathogenesis of several chronic disorders [65,66,67]. Disorders, such as neurodegenerative, cancer, cardiovascular, diabetes mellitus, are among the most frequent. Several studies have been done in order to determine the subjacent triggering factors, in particular to understand the role of free radicals in the occurrence these disorders, and also to discover potential substances towards preventing the occurrence of oxidative damages [68]. As part of anti-ROS agents, antioxidant, from both synthetic and natural sources, have proved to be efficient to regulate the production of free radicals, to minimize its undesirable effects and, hence, to prevent a wide variety of above mentioned human disorders [69]. As observed above P. kotschyi leaves and stem bark proved to be an excellent source of antioxidant compounds, which could be potent enzyme inhibitory agents against various enzymes involve in non-communicable disorders including diabetes mellitus, Alzheimer’s disease, and skin’s disorders. Thereby, the extract of both parts were screened for their possible anti-alpha-amylase, anti-alpha-glucosidase, anti-cholinesterases, anti-tyrosinase, and anti-elastase effects.
In view of the results, methanol stem bark exhibited the highest anti-cholinesterases activities. Phenolic acid and flavanol compounds contributed significantly to the observed inhibition effects. Subsequent surveys have determined the relevance of both BChE and AchE in the pathophysiology of Alzheimer’s disease and have demonstrated the therapeutic benefit of the inhibition of both BchE and AchE. In fact, it has been reported that AchE activity is reduced by 67% compared to the normal level in the hippocampus and temporal lobe during the progression of Alzheimer’s disease while the BchE activity is gone up to 165%, relative to normal levels [70]. This imbalance in the AChE/BChE ration causes acetylcholine deficit in the brain. Thus, AChE and BChE inhibitors prevent hydrolyze of the acetylcholine and, thereby, improving the deficient cholinergic neurotransmission. Flavonoid derivative have been considered as significant AChE inhibitors (Khan 2018). A few phenolic compound present in P. kotschyi extracts are reported to be potent inhibitors of AchE and BchE. For example, Khan in review indicate that activity may also be improved by increasing the gallation of catechin [71], and recently catechin of green tea have been considered for their Acetyl- and Butyryl-Cholinesterase Inhibition activity sowing that the most promising compound is epigallocatechin gallate [72].
Related to anti-hyperglycemia assays, water leaves extract had the strongest anti-amylase activity while methanol leaves extract showed the highest anti-glucosidase activities. In an experiment carried out on alloxan induced diabetic rats, the reduction in the blood glucose levels of the rats after four days of treatment with P. kotschyi was demonstrated [12]. Another study dealing with the analysis of the biochemical and hematological parameters in alloxan-induced diabetic rats treated with P. kotschyi roots, result on the decrease in blood urea, creatinine, glucose levels, ALT, and AST [13]. From a significant increase in the leukocyte and erythrocyte counts, PCV and Hb values were observed [13]. All these results may explain the traditional use of the plant by traditional healers as anti-diabetes mellitus agents [12]. Furthermore, the presence of proanthocyanidin groups in the extracts provides further evidence for the observed anti-amylase and glucosidase effects. [73] reported that proanthocyanidins significantly ameliorate the high blood sugar, the hyperlipidemia and the oxidative stress in the mouse model of type 2 diabetes. Although treatment of insulin-resistant mice with proanthocyanidin-rich extract derived from longan flower enhanced the expression of glucose transporter 4 and insulin receptor substrat-1 and declined the systolic blood pressure [74].
Additionally, leaves-methanol extract which exhibited the highest tyrosinase inhibitory activity, showed no inhibitory activity with elastase. However, the remaining extracts inhibited elastase without any significance difference between them. The development of new effective cosmetic products from nature to improve the appearance of the skin and delaying premature aging significantly bounced back over the years. In fact, natural-called cosmetics products are very fashionable for consumers throughout the world. This can be explained by the fact that numerous research has shown that herbals contain biomolecules, having a significant effect on skin brightening similarly to sunscreen and skin aging as antioxidant agents. The inhibitors of elastase are useful to overcome the loss of skin sagging and elasticity, while those of tyrosinase allow to limit melanin synthesis and, hence, ensure a breakdown of skin disorders, i.e., hyperpigmentation and melanoma [75]. In this regard, inhibition of elastase and tyrosinase enzymes are considered as a significant targets in cosmetic industry for researches to find various kind of new skin brightening and aging agents. LC-MSn analysis showed that P. kotschyi contained several potential anti-tyrosinase and anti-elastase compounds, in particular proanthocyanidins that could retard the aging and abnormal pigmentation processes, by acting as strong H-donors, oxygen quenchers, or reducing agents [76]. Therefore, P. kotschyi can be used in various sectors of the cosmetic industry as natural additives ingredients.
Foodborne disease is a serious medical issue. Indeed, various well-publicized outbreaks of foodborne acute or chronic disease have alerted researcher’s awareness that adverse microorganisms may be present in food, at the origin of these diseases. In addition to acute gastrointestinal issues, certain foodborne pathogens, through complex interactions with the human immune response, cause diseases that impact various major organ systems [77]. Given that foodborne disease is a main cause of morbidity and mortality, the suppression of causative pathogens presents a unique challenge for scientists, since the emergence of multi-drug resistance microorganisms. Indeed, fungi and bacteria constantly develop mechanisms of resistance to currently available drugs. The discovery of novel and effective antimicrobial drugs is the best approach to overcome fungal and bacterial resistance and develop appropriate new food preservatives or new effective therapeutic agents to combat human infectious diseases caused by microorganisms. For this purpose, the antimicrobial activity of secondary metabolites occurring in medicinal plants has been intensively investigated towards a wide range of fungi and bacteria. In this study, leaves and stem bark extracts of P. kotschyi showed promising antifungal and antibacterial activity. Far better, a few extracts displayed more pronounced antifungal and antibacterial activity than the reference drug used in this study. Illustratively, an interesting fungistatic or fungicidal effect towards A. ochraceus, A. fumigatus, P. ochrochloron, T. viride and P. funiculosum was observed with a MIC and MFC value ranging from 0.10 to 0.11 mg/mL and 0.15 to 0.21 mg/mL, respectively. These findings justify, for the first time, the possibility of using the extracts of P. kotschyi stem bark and leaves for preserving food against spoilage microorganisms or for managing human infections caused by said pathogens. Similarly a good bacteriostatic or bactericidal was observed against M. flavus, S. aureus, L. monocytogenes, E. coli, P. aeruginosa, E. cloacae, and S. typhimurium with a MIC and MBC value varying from 0.3 to 0.7 mg/mL and 0.7 to 0.21 mg/mL, respectively. Alain et al. [78] reported that E. coli had bacteriostatic and bactericidal sensitivity with ethanolic extract of P. kotschyi stem bark at concentration of 0.39 and 0.78 mg/mL, respectively. Additionally, the hydroethanolic extract of stem bark exhibited the highest bacteriostatic and bactericidal effect on S. aureus with MIC and MBC of 0.39 and 0.78 mg/mL, respectively [78]. Similarly, the ethyl acetate extract of P. kotschyi leaves was reported to be active against S. aureus, E. coli and S. pyogens at MIC value of 10 mg/mL [79]. These findings and reports could explain why the root of the P. kotschyi is commonly used as a toothpick for oral hygiene, since it proved to be active towards two enteropathogenic Gram-positive bacteria, i.e., S. auricularis and S. aureus [80]. The antifungal and antibacterial displayed by the studied extracts may be due to the presence of several molecules highlighted in their phytochemical screening. Indeed, the extracts were rich in variety of molecules of antimicrobial proprieties notably proanthocyanidins [81,82]. Mechanisms of action including cell membrane disruption, DNA gyrase and protein konase inhibition, dehydratase inactivation (HpFabZ) and type III secretion inactivation, which contribute to the antibacterial activity of mostly derived plant molecules have been reported [83]. Similarly, plant derived molecules act as antifungal agent by inhibiting the biofilm formation and the morphogenetic switch, activating the apoptotic pathway, increasing reactive oxygen species levels and interfering with the synthesis of different components (i.e., glucan, ergosterol, chitin, glucosamine, and proteins) in fungi [84]. Thereby, it can be said that not only P. kotschyi leaves and stem bark extracts can be used as antimicrobial agents but also in synergism with the commonly used antimicrobial drugs in order to potentiate their efficiency and to decrease synthetic antibiotic dose and hence to reduce their adverse reactions.
The solubility of the bioactive compounds and, in turn, the bioactivities were mostly impacted by the nature of the solvent used. These findings agree with earlier studies reporting the influence of solvents on the nature and content of molecules and their biological activities [85]. Overall, the maximum presence of bioactive compounds, as well as most of the better bioactivities were obtained with the methanol. Nevertheless, bearing in mind the toxicity of methanol, it would be recommender to aqueous solvent, since it contain numerous valuable molecules which have also shown excellent activities. On the other hand, variation on bioactive compounds and bioactivities were observed among the stem bark and leaves. The stem bark appeared to be the part in which pharmaceutical properties is most concentrated. A similar result was obtained by [86], who found that stem bark of Khaya senegalensis was superior source of secondary metabolites and lead excellent pharmaceutical properties, compared with the leaves. These outcomes indicate that the difference of pharmaceutical properties observed between samples P. kotschyi depended not only on the difference in the polarity of extraction solvents but also on the part types. Further, the excellent pharmaceutical properties obtained with both polar solvents confirms the richness of this herbal in polar molecules.

5. Conclusions

In the current study, the water and methanol extracts of leaves and stem bark from P. kotschyi was investigated for their chemical composition and the antioxidant property. Likewise, the capacity to inhibit cholinesterase, amylase, glucosidase, tyrosinase, and elastase activity, as well as the antimicrobial activities were evaluated. LC-MS analysis allowed to identify thirty-two compounds in the tested extracts, prodelphinidin derivative was the most abundant in stem bark, while epicatechin the most abundant in leaves. As far as the bioactivities, the extracts showed remarkable antioxidant properties and promising outcomes for the management of hyperglycemia, Alzheimer’s disease, and skin disorders. The extracts also show the antifungal and antibacterial effects on different microorganism strains. Afterward, it was evidence that the solubility of bioactive compounds and the bioactivities were depended on the extraction solvents and the plant parts. These antioxidant properties, enzymes inhibitory capacities, and antimicrobial activities potential could be, at least in part, ascribed to the phenolic constituents revealed in the extracts by LC-MS analysis. Finally, obtained data suggest that P. kotschyi could be used to promote human health on the understanding that further research regarding it toxicity be assessed.

Author Contributions

Conceptualization, K.I.S., S.D. and G.Z.; methodology, K.I.S., S.D., I.F., A.M., A.S., J.G., M.S. and M.N.; software, K.I.S.; validation, A.M., A.A. and G.Z.; formal analysis, A.A.; investigation, K.I.S. and G.Z.; resources, A.A. and G.Z.; data curation, K.I.S., S.D. and G.Z.; writing—original draft preparation, K.I.S. and S.D.; writing—review and editing, G.Z.; visualization, K.I.S.; supervision, A.A.; project administration, G.Z.; funding acquisition, A.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data is contained within the article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Newman, D.J.; Cragg, G.M. Natural Products as Sources of New Drugs from 1981 to 2014. J. Nat. Prod. 2016, 79, 629–661. [Google Scholar] [CrossRef] [Green Version]
  2. Newman, D.J.; Cragg, G.M. Natural Products as Sources of New Drugs over the Nearly Four Decades from 01/1981 to 09/2019. J. Nat. Prod. 2020, 83, 770–803. [Google Scholar] [CrossRef] [PubMed]
  3. Lautié, E.; Russo, O.; Ducrot, P.; Boutin, J.A. Unraveling Plant Natural Chemical Diversity for Drug Discovery Purposes. Front. Pharmacol. 2020, 11, 397. [Google Scholar] [CrossRef] [PubMed]
  4. Atanasov, A.G.; Zotchev, S.B.; Dirsch, V.M.; Orhan, I.E.; Banach, M.; Rollinger, J.M.; Barreca, D.; Weckwerth, W.; Bauer, R.; Bayer, E.A.; et al. Natural products in drug discovery: Advances and opportunities. Nat. Rev. Drug Discov. 2021, 20, 200–216. [Google Scholar] [CrossRef] [PubMed]
  5. Shakya, A.K. Medicinal plants: Future source of new drugs. Int. J. Herb. Med. 2016, 4, 59–64. [Google Scholar]
  6. Veeresham, C. Natural products derived from plants as a source of drugs. J. Adv. Pharm. Technol. Res. 2012, 3, 200. [Google Scholar] [CrossRef]
  7. Di, Y.T. The advances in the limonoid chemistry of the Meliaceae family. Curr. Org. Chem. 2011, 15, 1363–1391. [Google Scholar]
  8. Alhassan, A.M.; Ahmed, Q.U.; Malami, I.; Zakaria, Z.A. Pseudocedrela kotschyi: A review of ethnomedicinal uses, pharmacology and phytochemistry. Pharm. Biol. 2021, 59, 955–963. [Google Scholar] [CrossRef]
  9. Jolanta, P.; Daniel Jan, S.; Agata, K.; Stanislaw, L. Variability of biological activities of limonoids derived from plant sources. Mini Rev. Org. Chem. 2014, 11, 269–279. [Google Scholar] [CrossRef]
  10. Tan, Q.-G.; Luo, X.-D. Meliaceous limonoids: Chemistry and biological activities. Chem. Rev. 2011, 111, 7437–7522. [Google Scholar] [CrossRef]
  11. Castillo-Sánchez, L.E.; Jiménez-Osornio, J.J.; Delgado-Herrera, M.A. Secondary metabolites of the Annonaceae, Solanaceae and Meliaceae families used as biological control of insects. Trop. Subtrop. Agroecosyst. 2010, 12, 445–462. [Google Scholar]
  12. Georgewill, U.O.; Georgewill, O.A. Effect of extract of Pseudocedrela kotschyi on blood glucose conccentration of alloxan induced diabetic albino rats. East. J. Med. 2009, 14, 17. [Google Scholar]
  13. Ojewale, A.O.; Adekoya, A.O.; Odukanmi, O.A.; Olaniyan, O.T.; Samson, O. Protective effect of ethanolic roots extract of Pseudocedrela kotschyi (Pk) on some hematological and biochemical parameters in alloxan-induced diabetic rats. World J. Pharm. Pharm. Sci. 2013, 2, 852–866. [Google Scholar]
  14. Akah, P.; Nwafor, S.; Okoli, C.; Orji, U. Evaluation of the antiulcer properties of Pseudocedrela kotschyi Stem Bark Extract. Discov. Innov. 2001, 13, 132–135. [Google Scholar]
  15. Akuodor, G.; Essien, A.; Essiet, G.; David-Oku, E.; Akpan, J.; Udoh, F. Evaluation of antipyretic potential of Pseudocedrela kotschyi Schweint. Harms (Meliaceae). Eur. J. Med. Plants 2013, 105–113. [Google Scholar] [CrossRef]
  16. Essiet, G.A.; Christian, A.G.; Ogbonna, A.D.; Uchenna, M.A.; Azubuike, E.J.; Michael, N.E. Antidiarrhoeal and antioxidant properties of ethanol leaf extract of Pseudocedrela kotschyi. J. Appl. Pharm. Sci. 2016, 6, 107–110. [Google Scholar] [CrossRef] [Green Version]
  17. Hay, A.-E.; Ioset, J.-R.; Ahua, K.M.; Diallo, D.; Brun, R.; Hostettmann, K. Limonoid orthoacetates and antiprotozoal compounds from the roots of Pseudocedrela kotschyi. J. Nat. Prod. 2007, 70, 9–13. [Google Scholar] [CrossRef]
  18. Kassim, O.O.; Copeland, R.L.; Kenguele, H.M.; Nekhai, S.; Ako-Nai, K.A.; Kanaan, Y.M. Antiproliferative activities of Fagara xanthoxyloides and Pseudocedrela kotschyi against prostate cancer cell lines. Anticancer Res. 2015, 35, 1453–1458. [Google Scholar]
  19. Otimenyin, S.; Uguru, M.; Atang, B. Antiinflamatory and analgesic activities of Ficus thonningii and Pseudocedrela kotschyi extracts. Niger. J. Pharm. Res. 2004, 3, 82–85. [Google Scholar] [CrossRef]
  20. Alhassan, A.M.; Malami, I.; Abdullahi, M.I. Phytochemical screening and antimicrobial évaluation of stem bark extract of Pseudocedrela kotschyi (Schweinf.) Herms. J. Pharm. Res. Int. 2014, 1937–1944. [Google Scholar] [CrossRef]
  21. Asase, A.; Kokubun, T.; Grayer, R.J.; Kite, G.; Simmonds, M.S.; Oteng-Yeboah, A.A.; Odamtten, G.T. Chemical constituents and antimicrobial activity of medicinal plants from Ghana: Cassia sieberiana, Haematostaphis barteri, Mitragyna inermis and Pseudocedrela kotschyi. Phytother. Res. 2008, 22, 1013–1016. [Google Scholar] [CrossRef]
  22. Atinga, V.; Ahmed, A.; Nuhu, H.; Ayeni, E.; Dauda, G. Phytochemical and Structure Elucidation of Stigmasterol from the Stem Bark of Pseudocedrela kotschyi (Harms)(Meliaceae). Asian J. Res. Med. Pharm. Sci. 2018, 1–7. [Google Scholar] [CrossRef]
  23. Ekong, D.; Olagbemi, E. Novel meliacins (limonoids) from the wood of Pseudocedrela kotschyii. Tetrahedron Lett. 1967, 8, 3525–3527. [Google Scholar] [CrossRef]
  24. Niven, M.L.; Taylor, D.A. Revision of the structure of the limonoid pseudrelone B from Pseudocedrela kotschyii. Phytochemistry 1988, 27, 1542. [Google Scholar] [CrossRef]
  25. Oliver-Bever, B. Medicinal Plants in Tropical West Africa; Cambridge University Press: Cambridge, UK, 1986. [Google Scholar]
  26. Vladimir-Knežević, S.; Blažeković, B.; Bival Štefan, M.; Alegro, A.; Kőszegi, T.; Petrik, J. Antioxidant activities and polyphenolic contents of three selected Micromeria species from Croatia. Molecules 2011, 16, 1454–1470. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Zengin, G.; Aktumsek, A. Investigation of antioxidant potentials of solvent extracts from different anatomical parts of Asphodeline anatolica E. Tuzlaci: An endemic plant to Turkey. Afr. J. Tradit. Complementary Altern. Med. 2014, 11, 481–488. [Google Scholar] [CrossRef] [Green Version]
  28. Uysal, S.; Zengin, G.; Locatelli, M.; Bahadori, M.B.; Mocan, A.; Bellagamba, G.; De Luca, E.; Mollica, A.; Aktumsek, A. Cytotoxic and enzyme inhibitory potential of two Potentilla species (P. speciosa L. and P. reptans Willd.) and their chemical composition. Front. Pharmacol. 2017, 8, 290. [Google Scholar] [CrossRef] [PubMed]
  29. Grochowski, D.M.; Uysal, S.; Aktumsek, A.; Granica, S.; Zengin, G.; Ceylan, R.; Locatelli, M.; Tomczyk, M. In vitro enzyme inhibitory properties, antioxidant activities, and phytochemical profile of Potentilla thuringiaca. Phytochem. Lett. 2017, 20, 365–372. [Google Scholar] [CrossRef]
  30. Berman, H.; Westbrook, J.; Feng, Z.; Gilliland, G.; Bhat, T.; Weissig, H.; Shindyalov, I.; Bourne, P. The Protein Data Bank. Nucleic Acids Res. 2000, 28, 235–242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Ismaya, W.T.; Rozeboom, H.J.; Weijn, A.; Mes, J.J.; Fusetti, F.; Wichers, H.J.; Dijkstra, B.W. Crystal structure of Agaricus bisporus mushroom tyrosinase: Identity of the tetramer subunits and interaction with tropolone. Biochemistry 2011, 50, 5477–5486. [Google Scholar] [CrossRef] [Green Version]
  32. Pettersen, E.F.; Goddard, T.D.; Huang, C.C.; Couch, G.S.; Greenblatt, D.M.; Meng, E.C.; Ferrin, T.E. UCSF Chimera—A visualization system for exploratory research and analysis. J. Comput. Chem. 2004, 25, 1605–1612. [Google Scholar] [CrossRef] [Green Version]
  33. Sastry, G.M.; Adzhigirey, M.; Day, T.; Annabhimoju, R.; Sherman, W. Protein and ligand preparation: Parameters, protocols, and influence on virtual screening enrichments. J. Comput. Aided Mol. Des. 2013, 27, 221–234. [Google Scholar] [CrossRef]
  34. Schrödinger, 2017–3: Maestro; Schrödinger: New York, NY, USA, 2017.
  35. Jacobson, M.P.; Kaminski, G.A.; Friesner, R.A.; Rapp, C.S. Force field validation using protein side chain prediction. J. Phys. Chem. B 2002, 106, 11673–11680. [Google Scholar] [CrossRef] [Green Version]
  36. Placines, C.; Castañeda-Loaiza, V.; João Rodrigues, M.; Pereira, C.G.; Stefanucci, A.; Mollica, A.; Zengin, G.; Llorent-Martínez, E.J.; Castilho, P.C.; Custódio, L. Phenolic Profile, Toxicity, Enzyme Inhibition, In Silico Studies, and Antioxidant Properties of Cakile maritima Scop. (Brassicaceae) from Southern Portugal. Plants 2020, 9, 142. [Google Scholar] [CrossRef] [Green Version]
  37. Llorent-Martínez, E.J.; Zengin, G.; Fernández-de Córdova, M.L.; Bender, O.; Atalay, A.; Ceylan, R.; Mollica, A.; Mocan, A.; Uysal, S.; Guler, G.O.; et al. Traditionally Used Lathyrus Species: Phytochemical Composition, Antioxidant Activity, Enzyme Inhibitory Properties, Cytotoxic Effects, and in silico Studies of L. czeczottianus and L. nissolia. Front. Pharmacol. 2017, 8, 83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Uysal, S.; Aktumsek, A.; Picot, C.M.N.; Sahan, A.; Mollica, A.; Zengin, G.; Fawzi Mahomoodally, M. A comparative in vitro and in silico study of the biological potential and chemical fingerprints of Dorcycinum pentapyllum subsp. haussknechtii using three extraction procedures. New J. Chem. 2017, 41, 13952–13960. [Google Scholar] [CrossRef]
  39. Bender, O.; Llorent-Martínez, E.J.; Zengin, G.; Mollica, A.; Ceylan, R.; Molina-García, L.; Fernández-de Córdova, M.L.; Atalay, A. Integration of in vitro and in silico perspectives to explain chemical characterization, biological potential and anticancer effects of Hypericum salsugineum: A pharmacologically active source for functional drug formulations. PLoS ONE 2018, 13, e0197815. [Google Scholar] [CrossRef]
  40. Jones, G.; Willett, P.; Glen, R.C.; Leach, A.R.; Taylor, R. Development and validation of a genetic algorithm for flexible docking. J. Mol. Biol. 1997, 267, 727–748. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Stefanucci, A.; Dimmito, M.P.; Tenore, G.; Pieretti, S.; Minosi, P.; Zengin, G.; Sturaro, C.; Calò, G.; Novellino, E.; Cichelli, A.; et al. Plant-derived peptides rubiscolin-6, soymorphin-6 and their c-terminal amide derivatives: Pharmacokinetic properties and biological activity. J. Funct. Foods 2020, 73, 104154. [Google Scholar] [CrossRef]
  42. Stefanucci, A.; Lei, W.; Pieretti, S.; Novellino, E.; Dimmito, M.P.; Marzoli, F.; Streicher, J.M.; Mollica, A. On resin click-chemistry-mediated synthesis of novel enkephalin analogues with potent anti-nociceptive activity. Sci. Rep. 2019, 9, 5771. [Google Scholar] [CrossRef] [PubMed]
  43. Locatelli, M.; Yerlikaya, S.; Baloglu, M.C.; Zengin, G.; Altunoglu, Y.C.; Cacciagrano, F.; Campestre, C.; Mahomoodally, M.F.; Mollica, A. Investigations into the therapeutic potential of Asphodeline liburnica roots: In vitro and in silico biochemical and toxicological perspectives. Food Chem. Toxicol. 2018, 120, 172–182. [Google Scholar] [CrossRef]
  44. Schrödinger, 2017–3: LigPrep; Schrödinger: New York, NY, USA, 2017.
  45. Harder, E.; Damm, W.; Maple, J.; Wu, C.; Reboul, M.; Xiang, J.Y.; Wang, L.; Lupyan, D.; Dahlgren, M.K.; Knight, J.L. OPLS3: A force field providing broad coverage of drug-like small molecules and proteins. J. Chem. Theory Comput. 2015, 12, 281–296. [Google Scholar] [CrossRef] [PubMed]
  46. Sadeer, N.B.; Llorent-Martínez, E.J.; Bene, K.; Mahomoodally, M.F.; Mollica, A.; Sinan, K.I.; Stefanucci, A.; Ruiz-Riaguas, A.; Fernández-de Córdova, M.L.; Zengin, G. Chemical profiling, antioxidant, enzyme inhibitory and molecular modelling studies on the leaves and stem bark extracts of three African medicinal plants. J. Pharm. Biomed. Anal. 2019, 174, 19–33. [Google Scholar] [CrossRef] [PubMed]
  47. Llorent-Martínez, E.J.; Zengin, G.; Lobine, D.; Molina-García, L.; Mollica, A.; Mahomoodally, M.F. Phytochemical characterization, in vitro and in silico approaches for three Hypericum species. New J. Chem. 2018, 42, 5204–5214. [Google Scholar] [CrossRef]
  48. Zengin, G.; Lobine, D.; Mollica, A.; Locatelli, M.; Carradori, S.; Mahomoodally, M.F. Multiple pharmacological approaches on Fibigia eriocarpa extracts by in vitro and computational assays. Fundam. Clin. Pharmacol. 2018, 32, 400–413. [Google Scholar] [CrossRef] [PubMed]
  49. Pinto, G.; De Pascale, S.; Aponte, M.; Scaloni, A.; Addeo, F.; Caira, S. Polyphenol Profiling of Chestnut Pericarp, Integument and Curing Water Extracts to Qualify These Food By-Products as a Source of Antioxidants. Molecules 2021, 26, 2335. [Google Scholar] [CrossRef]
  50. Friedrich, W.; Eberhardt, A.; Galensa, R. Investigation of proanthocyanidins by HPLC with electrospray ionization mass spectrometry. Eur. Food Res. Technol. 2000, 211, 56–64. [Google Scholar] [CrossRef]
  51. Kachlicki, P.; Piasecka, A.; Stobiecki, M.; Marczak, Ł. Structural characterization of flavonoid glycoconjugates and their derivatives with mass spectrometric techniques. Molecules 2016, 21, 1494. [Google Scholar] [CrossRef] [Green Version]
  52. Vukics, V.; Guttman, A. Structural characterization of flavonoid glycosides by multi-stage mass spectrometry. Mass Spectrom. Rev. 2010, 29, 1–16. [Google Scholar] [CrossRef]
  53. Fabre, N.; Rustan, I.; de Hoffmann, E.; Quetin-Leclercq, J. Determination of flavone, flavonol, and flavanone aglycones by negative ion liquid chromatography electrospray ion trap mass spectrometry. J. Am. Soc. Mass Spectrom. 2001, 12, 707–715. [Google Scholar] [CrossRef] [Green Version]
  54. Annamala, M.K.; Inampudi, K.K.; Guruprasad, L. Docking of phosphonate and trehalose analog inhibitors into M. tuberculosis mycolyltransferase Ag85C: Comparison of the two scoring fitness functions GoldScore and ChemScore, in the GOLD software. Bioinformation 2007, 1, 339–350. [Google Scholar] [CrossRef] [Green Version]
  55. Tabart, J.; Kevers, C.; Pincemail, J.; Defraigne, J.-O.; Dommes, J. Comparative antioxidant capacities of phenolic compounds measured by various tests. Food Chem. 2009, 113, 1226–1233. [Google Scholar] [CrossRef]
  56. Abubakar, K.; Danjuma, N.; Maiha, B.; Anuka, J.; Magaji, M.; Malami, S. In Vitro and in Vivo Antioxidant Effect of the Stem Bark Extract of Pseudocedrela kotschyi in Pentylenetetrazole-kindled Rats. Niger. J. Pharm. Appl. Sci. Res. 2016, 5, 7–15. [Google Scholar]
  57. Dafam, D.G.; Agunu, A.; Ibrahim, H.; Ojerinde, O.S.; Ohemu, T.L.; Okwori, V.A.; Olotu, P.N.; Ilyas, N. Phytochemical screening and antioxidant activity of Pseudocedrela kotschyi Schweinf Harms (Meliaceae) and Strophanthus sarmentosus DC (Apocynaceae). J. Pharm. Bioresour. 2018, 15, 78–83. [Google Scholar] [CrossRef]
  58. Eleha, S.I.; Ola, M.A.; Oyewole, O.S.; Ganiyu, A.O.; Onaolapo, A.O.; Afodun, A.M. Anti-oxidative and Hepato-protective Effects of Pseudocedrela kotschyi against Paracetamol Induced Liver Damage: A Biochemical and Histological Evaluation in Rats. J. Adv. Med. Pharm. Sci. 2016, 1–11. [Google Scholar] [CrossRef]
  59. Khlebnikov, A.I.; Schepetkin, I.A.; Domina, N.G.; Kirpotina, L.N.; Quinn, M.T. Improved quantitative structure–activity relationship models to predict antioxidant activity of flavonoids in chemical, enzymatic, and cellular systems. Bioorganic Med. Chem. 2007, 15, 1749–1770. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Masek, A.; Chrzescijanska, E.; Latos, M.; Zaborski, M. Influence of hydroxyl substitution on flavanone antioxidants properties. Food Chem. 2017, 215, 501–507. [Google Scholar] [CrossRef] [PubMed]
  61. Pietta, P.-G. Flavonoids as antioxidants. J. Nat. Prod. 2000, 63, 1035–1042. [Google Scholar] [CrossRef]
  62. Van Acker, S.A.; Tromp, M.N.; Griffioen, D.H.; Van Bennekom, W.P.; Van Der Vijgh, W.J.; Bast, A. Structural aspects of antioxidant activity of flavonoids. Free Radic. Biol. Med. 1996, 20, 331–342. [Google Scholar] [CrossRef]
  63. Sut, S.; Dall’Acqua, S.; Sinan, K.I.; Bene, K.; Kumar, G.; Mahomoodally, M.F.; Picot-Allain, C.; Zengin, G. Cola caricifolia (G. Don) K. Schum and Crotalaria retusa L. from Ivory Coast as sources of bioactive constituents. Ind. Crop. Prod. 2020, 147, 112246. [Google Scholar] [CrossRef]
  64. Horbańczuk, O.K.; Kurek, M.A.; Atanasov, A.G.; Brnčić, M.; Brnčić, S.R. The Effect of Natural Antioxidants on Quality and Shelf Life of Beef and Beef Products. Food Technol. Biotechnol. 2019, 57, 439. [Google Scholar] [CrossRef] [PubMed]
  65. Phaniendra, A.; Jestadi, D.B.; Periyasamy, L. Free radicals: Properties, sources, targets, and their implication in various diseases. Indian J. Clin. Biochem. 2015, 30, 11–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Poprac, P.; Jomova, K.; Simunkova, M.; Kollar, V.; Rhodes, C.J.; Valko, M. Targeting free radicals in oxidative stress-related human diseases. Trends Pharmacol. Sci. 2017, 38, 592–607. [Google Scholar] [CrossRef] [PubMed]
  67. Rani, V.; Deep, G.; Singh, R.K.; Palle, K.; Yadav, U.C. Oxidative stress and metabolic disorders: Pathogenesis and therapeutic strategies. Life Sci. 2016, 148, 183–193. [Google Scholar] [CrossRef]
  68. Fernandez-Panchon, M.; Villano, D.; Troncoso, A.; Garcia-Parrilla, M. Antioxidant activity of phenolic compounds: From in vitro results to in vivo evidence. Crit. Rev. Food Sci. Nutr. 2008, 48, 649–671. [Google Scholar] [CrossRef]
  69. Martins, N.; Barros, L.; Ferreira, I.C. In vivo antioxidant activity of phenolic compounds: Facts and gaps. Trends Food Sci. Technol. 2016, 48, 1–12. [Google Scholar] [CrossRef] [Green Version]
  70. Marucci, G.; Buccioni, M.; Dal Ben, D.; Lambertucci, C.; Volpini, R.; Amenta, F. Efficacy of acetylcholinesterase inhibitors in Alzheimer’s disease. Neuropharmacology 2020, 190, 108352. [Google Scholar] [CrossRef]
  71. Khan, H.; Amin, S.; Kamal, M.A.; Patel, S. Flavonoids as acetylcholinesterase inhibitors: Current therapeutic standing and future prospects. Biomed. Pharmacother. 2018, 101, 860–870. [Google Scholar] [CrossRef]
  72. Okello, E.J.; Mather, J. Comparative kinetics of acetyl-and butyryl-cholinesterase inhibition by green tea catechins|relevance to the symptomatic treatment of Alzheimer’s disease. Nutrients 2020, 12, 1090. [Google Scholar] [CrossRef] [Green Version]
  73. Lee, Y.A.; Cho, E.J.; Yokozawa, T. Effects of proanthocyanidin preparations on hyperlipidemia and other biomarkers in mouse model of type 2 diabetes. J. Agric. Food Chem. 2008, 56, 7781–7789. [Google Scholar] [CrossRef]
  74. Tsai, H.-Y.; Wu, L.-Y.; Hwang, L.S. Effect of a proanthocyanidin-rich extract from longan flower on markers of metabolic syndrome in fructose-fed rats. J. Agric. Food Chem. 2008, 56, 11018–11024. [Google Scholar] [CrossRef]
  75. Apraj, V.D.; Pandita, N.S. Evaluation of skin anti-aging potential of Citrus reticulata blanco peel. Pharmacogn. Res. 2016, 8, 160. [Google Scholar] [CrossRef] [Green Version]
  76. Yodkeeree, S.; Thippraphan, P.; Punfa, W.; Srisomboon, J.; Limtrakul, P. Skin anti-aging assays of proanthocyanidin rich red rice extract, oryzanol and other phenolic compounds. Nat. Prod. Commun. 2018, 13, 1934578X1801300812. [Google Scholar] [CrossRef] [Green Version]
  77. Archer, D.L.; Young, F.E. Contemporary issues: Diseases with a food vector. Clin. Microbiol. Rev. 1988, 1, 377–398. [Google Scholar] [CrossRef]
  78. Alain, K.Y.; Oronce, D.O.; Boniface, Y.; Mhudro, Y.; Pascal, A.D.C.; Paul, T.F.; Alain, A.G.; Akpovi, A.; Dominique, S.K. Free radical scavenging and antibacterial potential of two plants extracts (Khaya senegalensis and Pseudocedrela kotschyi) used in veterinary pharmacopoeia in Benin. Elixir Appl. Chem. 2014, 76, 28720–28726. [Google Scholar]
  79. Ayo, R.; Audu, O.; Ndukwe, G.; Ogunshola, A. Antimicrobial activity of extracts of leaves of Pseudocedrela kotschyi (Schweinf.) Harms. Afr. J. Biotechnol. 2010, 9, 7733–7737. [Google Scholar]
  80. Akande, J.; Hayashi, Y. Potency of extract contents from selected tropical chewing sticks against Staphylococcus aureus and Staphylococcus auricularis. World J. Microbiol. Biotechnol. 1997, 14, 235–238. [Google Scholar] [CrossRef]
  81. Rauf, A.; Imran, M.; Abu-Izneid, T.; Patel, S.; Pan, X.; Naz, S.; Silva, A.S.; Saeed, F.; Suleria, H.A.R. Proanthocyanidins: A comprehensive review. Biomed. Pharmacother. 2019, 116, 108999. [Google Scholar] [CrossRef] [PubMed]
  82. De la Iglesia, R.; Milagro, F.I.; Campión, J.; Boqué, N.; Martínez, J.A. Healthy properties of proanthocyanidins. Biofactors 2010, 36, 159–168. [Google Scholar] [CrossRef] [PubMed]
  83. Rempe, C.S.; Burris, K.P.; Lenaghan, S.C.; Stewart, C.N., Jr. The potential of systems biology to discover antibacterial mechanisms of plant phenolics. Front. Microbiol. 2017, 8, 422. [Google Scholar] [CrossRef] [Green Version]
  84. Ansari, M.A.; Anurag, A.; Fatima, Z.; Hameed, S. Natural phenolic compounds: A potential antifungal agent. Microb. Pathog. Strateg. Combat. Sci. Technol. Educ. 2013, 1, 1189–1195. [Google Scholar]
  85. Dirar, A.; Alsaadi, D.; Wada, M.; Mohamed, M.; Watanabe, T.; Devkota, H. Effects of extraction solvents on total phenolic and flavonoid contents and biological activities of extracts from Sudanese medicinal plants. S. Afr. J. Bot. 2019, 120, 261–267. [Google Scholar] [CrossRef]
  86. De la Luz Cádiz-Gurrea, M.; Sinan, K.I.; Zengin, G.; Bene, K.; Etienne, O.K.; Leyva-Jiménez, F.J.; Fernández-Ochoa, Á.; del Carmen Villegas-Aguilar, M.; Mahomoodally, M.F.; Lobine, D. Bioactivity assays, chemical characterization, ADMET predictions and network analysis of Khaya senegalensis A. Juss (Meliaceae) extracts. Food Res. Int. 2020, 139, 109970. [Google Scholar] [CrossRef] [PubMed]
Figure 1. (A) Procyanidin B3; (B) Prodelphynidin B3 docked to tyrosinase.
Figure 1. (A) Procyanidin B3; (B) Prodelphynidin B3 docked to tyrosinase.
Antioxidants 10 01570 g001
Figure 2. Relationship between phytochemical compounds and antioxidant and six key enzyme inhibitory activities.
Figure 2. Relationship between phytochemical compounds and antioxidant and six key enzyme inhibitory activities.
Antioxidants 10 01570 g002
Figure 3. Relationship between phytochemical compounds and antifungal activities.
Figure 3. Relationship between phytochemical compounds and antifungal activities.
Antioxidants 10 01570 g003
Figure 4. Relationship between phytochemical compounds and antibacterial activities.
Figure 4. Relationship between phytochemical compounds and antibacterial activities.
Antioxidants 10 01570 g004
Figure 5. Score plot of the multi-block discriminant analysis on P. kotschyi bioactive antioxidant, enzyme inhibitory, antifungal, and antibacterial activities. (A): Discrimination of the samples according to the parts. (B): Discrimination of the samples according to the solvents of extraction.
Figure 5. Score plot of the multi-block discriminant analysis on P. kotschyi bioactive antioxidant, enzyme inhibitory, antifungal, and antibacterial activities. (A): Discrimination of the samples according to the parts. (B): Discrimination of the samples according to the solvents of extraction.
Antioxidants 10 01570 g005
Figure 6. Goodness of the models assessment by Area Under the Curve average using one-vs-all comparisons. (A): model 1 (Leaves versus Stem bark). (B): model 2 (Methanol versus Water).
Figure 6. Goodness of the models assessment by Area Under the Curve average using one-vs-all comparisons. (A): model 1 (Leaves versus Stem bark). (B): model 2 (Methanol versus Water).
Antioxidants 10 01570 g006
Figure 7. Main components in P. kotschyi extracts.
Figure 7. Main components in P. kotschyi extracts.
Antioxidants 10 01570 g007
Table 1. Docking scores expressed as Gold Fitness Score.
Table 1. Docking scores expressed as Gold Fitness Score.
Compound NameGOLD Fitness Score (Gold Score)
Epigallocatechin54.83
Prodelphynidin B368.18
Procyanidin B252.04
Procyanidin B366.74
Catechin58.02
Epicatechin54.97
Quinic acid50.97
Myricetin-7-glucoside57.64
Myricetin-3-rutinoside53.46
Quercetin-3-O-rutinoside55.53
Table 2. Quantitative phytochemical determinations of P. kotschyi leaves and stem bark extracts *.
Table 2. Quantitative phytochemical determinations of P. kotschyi leaves and stem bark extracts *.
p-ValueLeaves MethanolLeaves WaterStem Bark MethanolStem Bark Water
Total phenolic content (mg GAE/g)0.0001127.18 ± 1.85 b126.81 ± 0.89 b131.83 ± 1.81 a125.67 ± 0.89 b
Total flavonoid content (mg RE/g)0.000143.79 ± 0.30 b44.95 ± 0.38 a0.63 ± 0.17 d3.30 ± 0.14 c
Total phenolic acids content (mg CAE/g)0.000124.88 ± 0.58 c63.58 ± 2.00 a54.44 ± 2.91 b53.76 ± 4.01 b
Total flavanols content (mg CE/g)0.00017.77 ± 0.06 b5.99 ± 0.05 d14.14 ± 0.11 a6.24 ± 0.01 c
* Values are reported as mean ± S.D. GAE: Gallic acid equivalent; RE: Rutin equivalent; CAE: Caffeic acid equivalent; CE: Catechin equivalent. a–d line wise values with same superscripts of this type indicate no significant difference among extracts (p > 0.05).
Table 3. Identified and quantified constituents of P. kotschyi leaves and stem bark extracts using LC-DAD-MSn (mg/g), * indicate confirmed identification by reference compound comparison. Number indicating compounds are the one used for the structure of the main compounds reported in Chart 1 and of LC-MS chromatograms of Chart 2.
Table 3. Identified and quantified constituents of P. kotschyi leaves and stem bark extracts using LC-DAD-MSn (mg/g), * indicate confirmed identification by reference compound comparison. Number indicating compounds are the one used for the structure of the main compounds reported in Chart 1 and of LC-MS chromatograms of Chart 2.
Retention Time (min)Identification[M−H] HR-MSMolecular Formula (Exact Mass)FragmentsLeaves MethanolLeaves WaterStem Bark MethanolStem Bark Water
2.3P1 prodelphynidin gallocatechin-gallocatechin609.1263C30H25O14 (609.1244)441 423 305 297 255n.d.n.d.0.74 ± 0.020.15 ± 0.01
2.5P2 prodelphynidin B3 isomer 1593.1341C30H25O13 (593.1295)467 441 425 407 2891.55 ± 0.010.36 ± 0.010.94 ± 0.020.27 ± 0.01
3.6P3 prodelphynidin gallocatechin-gallocatechin609.1265C30H25O14 (609.1244)441 423 305 297 255n.d.n.d.0.47 ± 0.010.08 ± 0.01
5.2P4 prodelphynidin B3 isomer 2593.1315C30H25O13 (593.1295)467 441 425 407 289n.d.n.d.1.06 ± 0.010.16 ± 0.01
5.7P5 prodelphynidin gallocatechin-gallocatechin609.1274C30H25O14 (609.1244)441 423 305 297 255n.d.n.d.0.97 ± 0.010.09 ± 0.01
6.6P6 epigallocatechin *305.0715C15H13O7 (305.0661)261 219 218 179 165 1253.32 ± 0.020.71 ± 0.011.69 ± 0.020.63 ± 0.01
6.7P7 prodelphynidin B3 isomer 3593.132C30H25O13 (593.1295)467 441 425 407 2891.63 ± 0.010.77 ± 0.029.20 ± 0.020.60 ± 0.01
7.0P8 prodelphinidin B3 isomer 4593.132C30H25O13 (593.1295)467 441 425 407 289n.d.0.77 ± 0.011.35 ± 0.020.31 ± 0.01
7.3P9 prodelphinidin B3593.1332C30H25O13 (593.1295)467 441 425 407 289n.d.0.77 ± 0.021.32 ± 0.020.57 ± 0.01
7.7P10 procyanidin B2 *577.11394C30H25O12 (577.1346)451 425 407 2892.08 ± 0.011.22 ± 0.023.46 ± 0.021.26 ± 0.02
7.9P11 gallocatechin305.0742C15H13O7 (305.0661)261 219 218 179 165 125n.d.0.71 ± 0.011.34 ± 0.010.45 ± 0.01
8.3P12 procyanidin B dimer577.1407C30H25O12 (577.1346)452 425 407 2893.57 ± 0.021.45 ± 0.023.28 ± 0.021.62 ± 0.02
8.6P13 catechin *289.0761C15H13O6(289.0712) --0.12 ± 0.020.21 ± 0.01
9.7P14 procyanidin C1 isomer865.2001C45H37O18 (865.1980)739 713 696 577 451 4070.18 ± 0.020.15 ± 0.030.32 ± 0.020.18 ± 0.01
8.5P15 procyanidin C1 isomer865.1995C45H37O18 (865.1980)739 713 696 577 451 4070.18 ± 0.020.13 ± 0.020.26 ± 0.020.19 ± 0.02
9.4P16 epicatechin *289.0777C15H13O6 (289.0712)245 205 17914.78 ± 0.023.08 ± 0.021.48 ± 0.020.10 ± 0.01
9.4P17 galloyl procyanidin B2729.1489C37H29O16 (729.1456)245 205 1791.47 ± 0.020.47 ± 0.010.95 ± 0.020.11 ± 0.01
11.8P18 catechin gallate441.0823C22H17O10 (441.0821)577 425 407 289ndnd0.37 ± 0.020.09 ± 0.01
12.9P19 acetyl Procyanidin dimer635.1358C32H27O14 (635.1401)576 467 423n.d.n.d.0.28 ± 0.01
total28.713.731.09.1
2.1O1 quinic acid *191.0556C7H11O6 (191.0556)173 127 111 93 853.07 ± 0.023.40 ± 0.014.18 ± 0.024.57 ± 0.01
3.3O2 galloyl glucoside331.0649C13H15O10 (331.0665)271 211 169 1250.11 ± 0.010.02 ± 0.010.27 ± 0.010.02 ± 0.01
- total3.183.424.454.59
9.9F1 myricetin-7-O-galloyl glucoside631.09351C28H23O17 (631.0935)479 326 287 271 1790.31 ± 0.010.20 ± 0.01n.d.n.d.
10.0F2 myrecitin-3-O-rutinoside *625.1385C27H29O17 (625.1405)420 3160.31 ± 0.010.08 ± 0.010.01 ± 0.01n.d.
10.2F3 myrecitin-7-O-glucoside *479.0819C21H19O13 (479.0826)316 271 1794.24 ± 0.020.96 ± 0.020.03 ± 0.01n.d.
10.8F4 quercetin-3-O-glucoside gallate615.0986C28H23O16 (615.0986)463 301 271 255 179 1520.27 ± 0.010.19 ± 0.010.06 ± 0.01n.d.
11.1F5 myricetin-3-O-rutinoside463.0876C21H19O12 (463.0872)316 287 271 1798.92 ± 0.022.10 ± 0.020.12 ± 0.010.08 ± 0.01
12.3F6 quercetin-3-O-rutinoside *447.0925C21H19O11 (447.0927)301 271 179 1513.47 ± 0.020.84 ± 0.020.05 ± 0.01n.d.
12.7F7 myricetin-rhamnoside-glucoside625.1399C27H29O17 (625.1405)479 316 2710.25 ± 0.010.14 ± 0.010.01 ± 0.01n.d.
13.0F8 kaempferol-3-O-rhamnoside *431.0958C21H19O10 (431.0978)285 267 2290.14 ± 0.030.04 ± 0.010.01 ± 0.01n.d.
13.2F9 methoxy-quercetin317.03105C15H9O8 (317.0297)299 179 1510.49 ± 0.020.11 ± 0.01n.d.n.d.
13.6F10 quercetin-3-rhamnosyl-7-glucoside609.1449C27H29O17 (609.1456)463 301 1510.21 ± 0.040.14 ± 0.04n.d.n.d.
15.0F11 quercetin *301.041C15H9O7 (301.3482)179 1510.43 ± 0.010.44 ± 0.02n.d.n.d.
total18.835.240.290.08
nd: not detected.
Table 4. Antioxidant and enzyme inhibitory properties of P. kotschyi extracts *.
Table 4. Antioxidant and enzyme inhibitory properties of P. kotschyi extracts *.
p-ValueLeaves MethanolLeaves WaterStem Bark MethanolStem Bark Water
Antioxidant assaysABTS (mmol TE/g)0.00012.95 ± 0.02 b2.15 ± 0.08 d3.31 ± 0.01 a2.28 ± 0.04 c
DPPH (mmol TE/g)0.00011.91 ± 0.01 a1.65 ± 0.03 b1.94 ± 0.01 a1.59 ± 0.08 b
CUPRAC (mmol TE/g)0.00013.59 ± 0.07 b3.43 ± 0.02 c5.09 ± 0.08 a3.14 ± 0.01 d
FRAP (mmol TE/g)0.00011.86 ± 0.04 c2.11 ± 0.01 b2.86 ± 0.02 a1.66 ± 0.01 d
Phosphomolybdenum (mmol TE/g)0.00013.78 ± 0.23 b3.48 ± 0.04 bc5.16 ± 0.23 a3.25 ± 0.09 c
Metal chelating (mg EDTAE/g)0.00019.28 ± 0.44 c11.56 ± 0.07 b17.12 ± 0.46 a5.36 ± 0.34 d
Enzyme inhibitory assaysAChE inhibition (mg GALAE/g)0.00012.36 ± 0.02 b2.27 ± 0.03 c2.54 ± 0.02 a1.90 ± 0.04 d
BChE inhibition (mg GALAE/g)0.00013.46 ± 0.16 b2.18 ± 0.25 c5.48 ± 0.10 a3.47 ± 0.25 b
Tyrosinase inhibition (mg KAE/g)0.000177.39 ± 0.21 a68.32 ± 0.18 c75.93 ± 0.68 b57.32 ± 0.38 d
Amylase inhibition (mmol ACAE/g)0.00010.72 ± 0.01 b0.97 ± 0.01 a0.67 ± 0.01 c0.36 ± 0.01 d
Glucosidase inhibition (mmol ACAE/g)0.00010.91 ± 0.02 bna0.97 ± 0.01 ana
Elastase inhibition (mmol CE/g)0.00011.07 ± 0.07 b1.98 ± 0.10 a2.21 ± 0.12 a2.08 ± 0.06 a
* Values are reported as mean ± S.D. TE: Trolox equivalent; EDTAE: EDTA equivalent; GALAE: Galatamine equivalent; KAE: Kojic acid equivalent; ACAE: Acarbose equivalent. na: not active. a–d. line wise values with same superscripts of this type indicate no significant difference among extracts (p > 0.05).
Table 5. Antifungal activity of P. kotschyi samples (mg/mL) *.
Table 5. Antifungal activity of P. kotschyi samples (mg/mL) *.
SamplesA.v.A.o.A.n.A.f.P.o.P.f.T.v.P.v.c
Minimum inhibitory concentration (MIC)Leaves Methanol0.21 ± 0.01 a0.10 ± 0.01 c0.15 ± 0.01 b0.20 ± 0.01 a0.20 ± 0.01 b0.15 ± 0.01 b0.10 ± 0.01 c0.30 ± 0.01 a
Leaves Water0.10 ± 0.01 c0.10 ± 0.01 c0.15 ± 0.01 b0.21 ± 0.01 a0.10 ± 0.01 d0.10 ± 0.01 c0.11 ± 0.01 c0.20 ± 0.01 b
Stem Bark Methanol0.15 ± 0.01 b0.15 ± 0.01 b0.20 ± 0.01 a0.15 ± 0.01 b0.20 ± 0.01 b0.20 ± 0.01 a0.15 ± 0.01 b0.20 ± 0.01 b
Stem Bark Water0.15 ± 0.01 b0.15 ± 0.01 b0.21 ± 0.01 a0.11 ± 0.01 c0.15 ± 0.01 c0.15 ± 0.01 b0.11 ± 0.01 c0.20 ± 0.01 b
Ketoconazole0.20 ± 0.01 a0.20 ± 0.01 a0.15 ± 0.01 b0.20 ± 0.01 a1.00 ± 0.01 a0.20 ± 0.01 a1.00 ± 0.01 a0.20 ± 0.01 b
Bifonazole0.10 ± 0.01 c0.15 ± 0.01 b0.10 ± 0.01 c0.15 ± 0.01 b0.20 ± 0.01 b0.20 ± 0.01 a0.15 ± 0.01 b0.10 ± 0.01 c
Minimum fungicidal concentration (MFC)Leaves Methanol0.40 ± 0.01 b0.40 ± 0.01 b0.21 ± 0.01 b0.41 ± 0.01 b0.41 ± 0.01 b0.20 ± 0.01 d0.20 ± 0.01 c0.40 ± 0.01 a
Leaves Water0.21 ± 0.01 d0.20 ± 0.01 d0.20 ± 0.01 b0.41 ± 0.01 b0.21 ± 0.01 e0.21 ± 0.01 d0.20 ± 0.01 c0.41 ± 0.01 a
Stem Bark Methanol0.30 ± 0.01 c0.30 ± 0.01 c0.31 ± 0.01 a0.30 ± 0.01 c0.30 ± 0.01 c0.30 ± 0.01 b0.30 ± 0.01 b0.30 ± 0.01 b
Stem Bark Water0.30 ± 0.01 c0.31 ± 0.01 c0.30 ± 0.01 a0.15 ± 0.01 e0.30 ± 0.01 c0.31 ± 0.01 b0.15 ± 0.01 d0.30 ± 0.01 b
Ketoconazole0.50 ± 0.01 a0.50 ± 0.01 a0.20 ± 0.01 b0.50 ± 0.01 a1.50 ± 0.01 a0.50 ± 0.01 a1.50 ± 0.01 a0.30 ± 0.01 b
Bifonazole0.20 ± 0.01 d0.20 ± 0.01 d0.15 ± 0.01 c0.20 ± 0.01 d0.25 ± 0.01 d0.25 ± 0.01 c0.20 ± 0.01 c0.20 ± 0.01 c
* Different letters indicate the differences in these samples for each fungal strains (p < 0.05). Statistical analysis were individually performed for minimum inhibitory concentration (MIC) and minimum fungicidal concentration (MFC) values. A.v.: Aspergillus versicolor; A.o.: Aspergillus ochraceus; A.n.: Aspergillus niger; A.f.: Aspregillus fumigatus; P.o.: Penicillium ochrochloron; P.f.: Penicillium funiculosum; T.v: Trichoderma viride; P.v.c.: Penicillium verrucosum var. cyclopium.
Table 6. Antibacterial activity of P. kotschyi samples (mg/mL) *.
Table 6. Antibacterial activity of P. kotschyi samples (mg/mL) *.
SamplesB.c.M.f.S.a.L.m.E. coliP.a.E.c.S.t.
Minimum inhibitory concentration (MIC)Leaves Methanol0.15 ± 0.01 b0.31 ± 0.01 a0.10 ± 0.01 b0.05 ± 0.01 c0.30 ± 0.01 b0.30 ± 0.01 a0.61 ± 0.01 a0.60 ± 0.01 a
Leaves Water0.15 ± 0.01 b0.30 ± 0.01 a0.21 ± 0.01 a0.20 ± 0.01 a0.31 ± 0.01 b0.30 ± 0.01 a0.20 ± 0.01 b0.15 ± 0.01 b
Stem Bark Methanol0.11 ± 0.01 c0.04 ± 0.01 d0.04 ± 0.01 c0.04 ± 0.01 d0.11 ± 0.01 d0.11 ± 0.01 c0.03 ± 0.01 e0.07 ± 0.01 d
Stem Bark Water0.20 ± 0.01 a0.31 ± 0.01 a0.03 ± 0.01 c0.21 ± 0.01 a0.40 ± 0.01 a0.15 ± 0.01 b0.15 ± 0.01 c0.16 ± 0.01 b
Streptomycin0.10 ± 0.01 d0.05 ± 0.01 c0.10 ± 0.01 b0.15 ± 0.01 b0.10 ± 0.01 d0.10 ± 0.01 d0.10 ± 0.01 d0.10 ± 0.01 c
Ampicillin0.10 ± 0.01 d0.10 ± 0.01 b0.10 ± 0.01 b0.15 ± 0.01 b0.15 ± 0.01 c0.30 ± 0.01 a0.15 ± 0.01 c0.10 ± 0.01 c
Minimum bactericidal concentration (MBC)Leaves Methanol0.20 ± 0.01 b0.40 ± 0.01 b0.20 ± 0.01 b0.21 ± 0.01 d0.40 ± 0.01 b0.40 ± 0.01 b0.80 ± 0.01 a0.80 ± 0.01 a
Leaves Water0.20 ± 0.01 b0.41 ± 0.01 b0.40 ± 0.01 a0.40 ± 0.01 b0.41 ± 0.01 b0.41 ± 0.01 b0.40 ± 0.01 c0.20 ± 0.01 c
Stem Bark Methanol0.15 ± 0.01 c0.07 ± 0.01 e0.08 ± 0.01 d0.08 ± 0.01 e0.15 ± 0.01 d0.16 ± 0.01 e0.07 ± 0.01 e0.15 ± 0.01 d
Stem Bark Water0.30 ± 0.01 a0.60 ± 0.01 a0.07 ± 0.01 d0.30 ± 0.01 c0.61 ± 0.01 a0.31 ± 0.01 c0.60 ± 0.01 b0.30 ± 0.01 b
Streptomycin0.15 ± 0.01 c0.10 ± 0.01 d0.20 ± 0.01 b0.30 ± 0.01 c0.20 ± 0.01 c0.20 ± 0.01 d0.20 ± 0.01 d0.20 ± 0.01 c
Ampicillin0.15 ± 0.01 c0.15 ± 0.01 c0.15 ± 0.01 c0.50 ± 0.01 a0.20 ± 0.01 c0.50 ± 0.01 a0.20 ± 0.01 d0.20 ± 0.01 c
* Column wise values with different letters (a–e) indicate the differences in these samples for each bacterial strains (p < 0.05). Statistical analysis were individually performed for minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) values. B.c.: Bacillus cereus; M.f.: Micrococcus flavus; S.a.: Staphylococcus aureus; L.m.: Listeria monocytogenes; E. coli: Escherichia coli; P.a.: Pseudomonas aeruginosa; E.c: Enterobacter cloacae; S.t.: Salmonella typhimurium.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Sinan, K.I.; Dall’Acqua, S.; Ferrarese, I.; Mollica, A.; Stefanucci, A.; Glamočlija, J.; Sokovic, M.; Nenadić, M.; Aktumsek, A.; Zengin, G. LC-MS Based Analysis and Biological Properties of Pseudocedrela kotschyi (Schweinf.) Harms Extracts: A Valuable Source of Antioxidant, Antifungal, and Antibacterial Compounds. Antioxidants 2021, 10, 1570. https://doi.org/10.3390/antiox10101570

AMA Style

Sinan KI, Dall’Acqua S, Ferrarese I, Mollica A, Stefanucci A, Glamočlija J, Sokovic M, Nenadić M, Aktumsek A, Zengin G. LC-MS Based Analysis and Biological Properties of Pseudocedrela kotschyi (Schweinf.) Harms Extracts: A Valuable Source of Antioxidant, Antifungal, and Antibacterial Compounds. Antioxidants. 2021; 10(10):1570. https://doi.org/10.3390/antiox10101570

Chicago/Turabian Style

Sinan, Kouadio Ibrahime, Stefano Dall’Acqua, Irene Ferrarese, Adriano Mollica, Azzurra Stefanucci, Jasmina Glamočlija, Marina Sokovic, Marija Nenadić, Abdurrahman Aktumsek, and Gokhan Zengin. 2021. "LC-MS Based Analysis and Biological Properties of Pseudocedrela kotschyi (Schweinf.) Harms Extracts: A Valuable Source of Antioxidant, Antifungal, and Antibacterial Compounds" Antioxidants 10, no. 10: 1570. https://doi.org/10.3390/antiox10101570

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

Article Metrics

Back to TopTop