Next Article in Journal
Intrarenal Arterial Transplantation of Dexmedetomidine Preconditioning Adipose Stem-Cell-Derived Microvesicles Confers Further Therapeutic Potential to Attenuate Renal Ischemia/Reperfusion Injury through miR-122-5p/Erythropoietin/Apoptosis Axis
Next Article in Special Issue
The Current State of Knowledge in Biological Properties of Cirsimaritin
Previous Article in Journal
Bile Acids Induce Alterations in Mitochondrial Function in Skeletal Muscle Fibers
Previous Article in Special Issue
A Comparative Study on Phytochemical Fingerprint of Two Diverse Phaseolus vulgarisvar. Tondino del Tavo and Cannellino Bio Extracts
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Phytochemical Screening, Antioxidant, and Enzyme Inhibitory Properties of Three Prangos Species (P. heyniae, P. meliocarpoides var. meliocarpoides, and P. uechtritzii) Depicted by Comprehensive LC-MS and Multivariate Data Analysis

by
Stefano Dall’Acqua
1,*,
Stefania Sut
1,
Gokhan Zengin
2,*,
Gregorio Peron
3,
Fevzi Elbasan
4,5,
Evren Yildiztugay
4,
Nabeelah Bibi Sadeer
6 and
Mohamad Fawzi Mahomoodally
6,7,8
1
Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131 Padova, Italy
2
Department of Biology, Science Faculty, Selcuk University, Konya 42300, Turkey
3
Department of Molecular and Translational Medicine (DMMT), University of Brescia, Viale Europa 11, 25123 Brescia, Italy
4
Department of Biotechnology, Science Faculty, Selcuk University, Konya 42300, Turkey
5
Department of Soil Science and Plant Nutrition, Selcuk University, Konya 42300, Turkey
6
Department of Health Sciences, Faculty of Medicine and Health Sciences, University of Mauritius, Réduit 80837, Mauritius
7
Center for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Science, Chennai 600077, India
8
Centre of Excellence for Pharmaceutical Sciences (Pharmacen), North West University, Potchefstroom 2520, South Africa
*
Authors to whom correspondence should be addressed.
Antioxidants 2022, 11(9), 1712; https://doi.org/10.3390/antiox11091712
Submission received: 15 July 2022 / Revised: 18 August 2022 / Accepted: 25 August 2022 / Published: 30 August 2022

Abstract

:
The aim of the present study was to identify/quantify bioactive compounds and determine the antioxidant activity and enzyme inhibitory effects of various solvent extracts (n-hexane, ethyl acetate, methanol, and water) of Prangos heyniae H. Duman and M.F. Watson, Prangos meliocarpoides var. meliocarpoides, and Prangos uechtritzii Boiss. and Hausskn. This is the first time such a report has been designed to validate the phytochemical composition and bioactivity (especially enzyme inhibitory properties) of these plants. A combined approach of liquid chromatography (LC) with mass spectrometry (HR-MS and MSn) allowed to identify that P. heyniae contains condensed tannins; P. meliocarpoides is rich in hydrolysable tannins; and P. uechtritzii possesses coumarins, flavonoids, and hydroxycinnamic acids. Different extracts were tested for antioxidant activities using a battery of assays, such as 2,2-diphenyl-1-picrylhydrazyl (DPPH), 2,2-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid) (ABTS), ferric reducing antioxidant power (FRAP), cupric reducing antioxidant capacity (CUPRAC), total antioxidant capacity (TAC) (phosphomolybdenum), and metal chelating. Enzyme inhibitory effects were investigated using acetylcholinesterase (AChE), butyrylcholinesterase (BChE), tyrosinase, α-amylase, and α-glucosidase as target enzymes. The obtained results depended on the extraction solvents used for each Prangos species. The methanol extract of P. meliocarpoides var. meliocarpoides exhibited significant radical scavenging activity (DPPH: 52.27 mg Trolox equivalent (TE)/g; ABTS: 92.84 mg TE/g), the most potent-reducing potential (CUPRAC: 154.04 mg TE/g; FRAP: 104.34 mg TE/g), and high TAC (2.52 mmol TE/g). Moreover, the strongest BChE (7.97 mg galantamine equivalent/g), α-amylase (0.46 mmol acarbose equivalent/g), and tyrosinase (81.15 mg kojic acid equivalent/g) inhibitory effects were observed for the hexane extract of P. meliocarpoides var. meliocarpoides. Correlation analysis showed a significant positive correlation between hydrolysable tannins and antioxidant activities. The same trend was also observed between the same class of compounds and the inhibitory effects on enzymatic activities. These results suggest a principal role of hydrolysable tannins in the observed bioactivities of Prangos. Our results suggested that the tested Prangos species could be valuable as sources of natural agents in the development of health-promoting applications.

1. Introduction

Plants are still significant sources of bioactive constituents, and many of the known secondary metabolites still need a lot of research to assess their potential usefulness as therapeutic agents. In the recent time, the development of new therapeutic agents has considered the so-called drug-repositioning and, in this regard, natural products show great potential because they have been used for various medical purposes for thousands of years [1,2]. Furthermore, many natural compounds have been investigated only for limited bioactivities, and they still can be used to assess new potential effects. Humans have used medicinal herbs empirically, with no rational knowledge of their pharmacological effects or active ingredients, but just learning from the experience. Plant-based rational drug discovery began in the early 19th century [3], trying to combine the chemical composition with bioactivity. Despite the importance of biotechnological drugs and monoclonal antibodies, medicinal plants still offer a unique opportunity to discover novel drugs thanks to their exceptional chemical diversity [4].
Plants are a rich source of chemicals, including flavonoids, anthocyanins, carotenoids, catechins, cinnamic acid derivatives, chalcones, stilbenes, and tocopherols, which can promote health through antioxidant action [5]. Phytochemicals can operate as multiple target molecules and can be valuable sources of health-promoting agents, particularly knowing that the pathophysiology of many illnesses is multifactorial and not caused by a single factor [6]. To ensure product safety, particular attention should be paid to pharmaceutical formulation, extraction and manufacture, and mode of action [7]. In this respect, medicinal plants must be investigated to identify possible antioxidants or enzyme inhibitors for use as nutraceuticals, functional foods, or medications.
The genus Prangos includes 45 species worldwide and the Irano-Turanian region is the gene center of the genus [8]. The genus is represented by 19 species in Turkey and most of them are distributed in the Central and East Anatolia Region [9]. In the literature, several studies have been performed on the members of the genus Prangos. For example, crude extracts or essential oil of P. ferulacea (L.) Lindl. are commonly investigated in terms of its volatile and non-volatile components, cytotoxic activity; antioxidant, antimicrobial, hypoglycemic, acetylcholinesterase, and analgesic activities [10,11,12]; wound healing property [13]; and antiviral activity [14]. Interestingly, a randomized controlled trial was even conducted to determine the effect of a vaginal cream containing P. ferulacea on accelerating the recovery of bacterial vaginosis. The results showed that the cream displayed a positive effect on patients with this type of inflammation [15]. A novel coumarin (yuganin A) identified in the roots of P. pabularia Lindl. exhibited potent effects on the proliferation of B16 melanoma cells [16]. P. haussknechtii Bioss containing coumarins 1 and 2, monoterpenoids, and amino acid derivatives inhibited lipid peroxidation with IC50 values between 43 and 114 μM, and reduced MTT to formazan blue between 48 and 128 μM [17]. Tan, et al. [18] assessed the antibacterial activity of the pyrenylated coumarin 4′-senecioiloxyosthol, identified in the roots of P. hulusii Senol, Yildirim, and Secmen. The results showed that the coumarin exhibited antimicrobial activity against Bacillus subtilis at a minimum inhibitory concentration of 5 µg/mL. The aim of this study is to assess the bioactive compounds and biological activities of three underexplored Prangos species, namely P. heyniae H.Duman and M.F.Watson, P. meliocarpoides var. meliocarpoides, and P. uechtritzii Boiss. and Hausskn. In the literature, the Prangos species have been studied for several biological abilities. For example, Ahmed et al. [19] investigated the total phenolic and DPPH radical scavenging abilities of the methanol and water extracts of the three Prangos species, and the water extract of P. heyniae was the richest in terms of total phenolic level, with 127.33 mg GAE/g. In addition, the extract was the most active one in DPPH radical scavenging ability with the lowest IC50 values (20.96 µg/mL). In a recent study performed by Albayrak et al. [20], two new coumarin glycosides (7-methoxy isoarnottinin 4′-O-β-d-glucopyranoside and 7-methoxy isoarnottinin 4′-O-rutinoside) were isolated from the roots of P. heyniae collected from Turkey. In addition to the studies on the extract of P. heyniae, several studies showed chemical characterization and some biological activities of the essential oil of the species [21,22]. In an earlier study conducted by Oke et al. [23], the radical scavenging and chemical composition of P. meliocarpoides fruit extracts were determined and the water extract was found to be the most active one. Moreover, chlorogenic and rutin were determined to be the main components in their study. The fruit extract had a higher radical scavenging ability as compared with the root and aerial parts in another study [19]. Among the targeted Prangos species, P. uechtritzii has been the most studied one and several studies reported its essential oil composition as well as biological activities [24,25,26]. A recent study by Sevin et al. [27] examined the erectile function of root extract of P. uechtritzii as well as P. heyniae, and the authors reported that oxypeucedanin was the most active coumarin. In our earlier paper [28], the chemical characterization and biological activities of the essential oils of three Prangos species were reported. Although many studies have examined the phytochemical composition and bioactivity of different Prangos species, the three species examined here still need to be evaluated and confirmed by scientists. To the best of our knowledge, this study is the first to report the phytochemical composition, antioxidant effects, and inhibitory activity against α-amylase, α-glucosidase, acetylcholinesterase, butyrylcholinesterase, and tyrosinase enzymes of secondary metabolites extracted from the same species. We believe that the results presented here could fill in the research gap and subsequently open new research avenues, particularly in the development of therapeutic bioproducts.

2. Materials and Methods

2.1. Plant Material and Extraction

The aerial parts of the Prangos species were collected during Summer 2020 from Central Anatolia Region of Turkey. The information of the collection area was reported in our earlier paper [28]. Voucher specimens were deposited at the herbarium of Selcuk University, Science Faculty (Voucher numbers: EY-3039, EY-2998, and EY-3023 for P. heyniae, P. meliocarpoides var. meliocarpoides, and P. uechtritzii, respectively). The plant samples were dried in shade conditions at room temperature for about one week. Then, the powdering procedure was performed using a mill, and the samples were stored in the dark.
Extracts were prepared using n-hexane, ethyl acetate, methanol, and water. Overnight, the plant material (10 g) was macerated at room temperature with 200 mL of solvents (hexane, ethyl acetate, and methanol, individually). Finally, solvents were evaporated from the mixtures. Plant materials (10 g) were extracted in 200 mL of boiling water for 15 min before being filtered. Water extracts were lyophilized. All extracts were stored at 4 °C until further analysis.

2.2. Total Phenolic and Flavonoid Contents

The total phenolic and flavonoid contents were determined using the Folin–Ciocalteu and AlCl3 tests, respectively [29]. The results were presented as gallic acid equivalents (mg GAEs/g dry extract) and rutin equivalents (mg REs/g dry extract) for the assays. The experimental details are given in the supplemental section.

2.3. LC-DAD-MSn

Extracts were dissolved in appropriate solvent: the more lipophilic extracts were dissolved in DMSO, methanol extracts in methanol, and water extracts were dissolved in water/DMSO mixtures. Samples were prepared weighting 10 mg (±0.1 mg) of dried material and dissolved in 10 mL of solvent using an ultrasound bath (WWR, Ultrasonic bath 45 Hz, 60 W, WWR Milano, Italy) for 15 min [30]. Then, the volume was adjusted at 20 mL and solutions were centrifuged at 13,000 rpm prior to being transferred to glass vials for analyses [30].
LC-DAD-MSn analyses were performed using an Agilent 1260 chromatograph equipped with autosampler, column oven, and diode array detector (DAD), all from the 1260 series. At the end of the chromatographic column, a “T” connector was fixed splitting the flow into two identical volumes: one was linked to DAD, while the other was connected to the electro spray (ESI) ion source of a Varian 500 Ion Trap mass spectrometer (MS). The spectrometer operated in both negative and positive ion modes. Data were acquired in the 100–2000 m/z range using the turbo data depending scanning (tdds) function, which allows the recording of the fragmentation of the most abundant ionic species. MS parameters were as follows: needle voltage: 4800 V, capillary voltage: 80 V, drying gas pressure: 25 psi, nebulizer pressure: 40 psi, drying gas temperature: 280 °C, and nebulizer temperature: 270 °C. For the identification of compounds, MSn spectra were acquired and compared with the literature and available online databases (Human Metabolome Database (HMDB) and Food Metabolome Database (FOodB)). Furthermore, MS data were compared with those of reference compounds available in the laboratory or acquired by commercial sources. Quantification of compounds was obtained using selected reference standards for each class of identified constituents. The calibration curves were as follows: imperatorin (detected in MS, positive ion mode, and DAD at 320 nm), reference solutions from 90 to 1.5 µg/mL, y = 35x − 0.320; rutin (detected in MS, negative ion mode, and in DAD at 350 nm), reference solutions from 100 to 1.0 µg/mL, y = 72x − 1.20; ellagic acid (detected in MS, negative ion mode, and in DAD at 254 nm), reference solutions from 100 to 1.0 µg/mL, y = 37.5x − 0.88; and epigallocatechin gallate (detected in MS, negative ion mode, and in DAD at 280 nm), reference solutions from 120 to 1.0 µg/mL, y = 83x − 0.992.
Accurate m/z values of identified metabolites were obtained by UPLC-QToF analysis of the same Prangos extracts. The system used for the analysis was an Acquity UPLC (Waters) coupled with a QToF MS model Xevo G2 (Waters). Agilent Zorbax Eclipse Plus C18 (2.1 × 50 mm, 1.8 μm) was used as the stationary phase, which was kept at a constant temperature of 40 °C. As the mobile phase, a mixture of water with 1% formic acid (A) and acetonitrile (B) was used. The elution gradient started from 0 to 1 min, 98% A; then at 11 min, 15% A; at 16 min, 0% A; and kept isocratic until 20 min. Then, at 21 min, 98% A and 24 min, 98% A. The flow rate was 300 µL/min, and the injection volume was 2 μL. MS data were acquired in both ESI(+) and ESI(−) in the mass range of 50–2000 Da. The sampling cone voltage was adjusted at 40 V and the source offset was 80 V. The capillary voltage was set to 3.5 KV. Nitrogen was used as nebulizer gas at a flow rate of 800 L/h. The desolvation temperature was 450 °C. Mass accuracy and reproducibility were maintained by infusing lock mass (leucine–enkephalin, [M + H]+ = 556.2771 m/z, and [M − H] = 554.2620 m/z) through Lockspray at a flow rate of 20 μL/min. The m/z value of all acquired spectra was automatically corrected during acquisition based on lock mass. MSe experiment was simultaneously performed to collect structural information, setting the collision energy to 30 V.

2.4. Antioxidant Assays

Antioxidant assays were performed using methods that have been previously reported [31,32]. The antioxidant potential was calculated as follows: mg Trolox equivalents (TE)/g extract in the 2,2-diphenyl-1-picrylhydrazyl (DPPH) and 2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) radical scavenging tests; cupric-reducing antioxidant capacity (CUPRAC) and ferric-reducing antioxidant power (FRAP); and mmol TE/g extract in the (MCA). The experimental details are given in the supplemental section.

2.5. Enzyme Inhibitory Assays

The enzyme inhibition experiments were performed based on previously described procedures [31,32]. Amylase and glucosidase inhibition was expressed as mmol acarbose equivalents (ACAEs)/g extract, while acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) inhibition was expressed as mg galanthamine equivalents (GALAEs)/g extract. Tyrosinase inhibition was expressed as mg kojic acid equivalents (KAEs)/g extract. The experimental details are given in the supplemental section.

2.6. Statistical Analysis

This study used ANOVA (Tukey’s test) in order to determine whether there were any differences in the extract levels between the three samples. xlStat was used to carry out the statistical analysis. p-values < 0.05 were considered as statistically significant. Multivariate statistical analyses were performed on chemical and biological data to uncover correlations between chemical constituents of Prangos extracts and their bioactivities, i.e., to describe possible activity biomarkers. Predictive OPLS models were developed using SIMCA (Umetrics, Sweden), and they were validated performing a permutation test (n = 200 permutations). Pearson’s rank correlation test was performed using the open-source MetaboAnalyst platform (https://www.metaboanalyst.ca/, accessed on 14 July 2022). Prior to being analyzed, data were pre-processed as follows: variables with more than 80% missing values were removed, and the remaining missing values were imputed using the KNN algorithm; finally, they were normalized by log-transformation and Pareto scaling.

3. Results and Discussion

3.1. Characterization of Bioactive Secondary Metabolites

The choice of the most suitable extraction solvent is an important step in order to define the quality and yield of extraction of these compounds [33]. Solvents used to extract bioactive chemicals from plants are chosen based on the polarity of the solute of interest, because a solute with comparable polarity to the solvent will be adequately dissolved according to the rule of similarity and intermiscibility (like dissolves like) [34,35]. In this study, the polyphenolic compounds such as phenolics (TPCs) and flavonoids (TFCs) of P. heyniae, P. meliocarpoides var. meliocarpoides, and P. uechtritzii extracted with four solvents of different polarities (water > methanol > ethyl acetate > hexane) were quantified. The results are presented in Table 1.
Overall, we observed a decline in TPC and TFC in the different plant species studied in the following order: P. meliocarpoides var. meliocarpoides (TPC: 44.28 mg GAE/g, and TFC: 44.66 mg RE/g), P. heyniae (TPC: 38.77 mg GAE/g, and 28.75 mg RE/g), and P. uechtritzii (TPC: 31.20 mg GAE/g, and TFC: 28.22 mg RE/g). The most polar solvents, namely water and methanol, were successful in extracting phenolics and flavonoids. As such, it can be said that the compounds extracted were mostly polar. On the other hand, hexane extracted the least number of compounds. A number of publications have reported methanol and water as the most efficient solvents in obtaining a high yield of extracts, as well as in extracting polar bioactive compounds such as phenolics and flavonoids [36,37].
The bioactive compounds in Prangos species were investigated by combining positive and negative ion mode LC-MS (Table 2). The analysis revealed that the extraction of Prangos species yielded extracts containing different classes of secondary metabolites, which we grouped as condensed tannins, hydrolysable tannins, coumarins, flavonoids hydroxycinnamic acid derivatives, and “other constituents”. Schematic representations of the relative abundance of the main constituents are reported in the form of pie-charts in Supplementary Materials Figures S1–S3. The structures of the identified compounds were deduced owing to the HR-MS data as well as by MSn. Epigallocatechin gallate derivatives are characterized by the loss of one or two gallic acid moieties (m/z 152), leading in this latter case to ion m/z 305, corresponding to the epigallocatechin moiety [38,39] (Table 1). Other condensed tannins in P. heyniae were prodelphynidine gallate derivatives, compounds in fact present the molecular formula of C37H30O18, which generate fragment because of −152 Da ascribable to gallic acid moiety. Further fragmentations of the species generated at m/z 611 are ascribable to prodelphynidin dimer gallate. Four different isomeric derivatives are present in P. heyniae. Two further peaks presenting similar fragmentation and showing the loss of two gallic acid moieties have also been detected and annotated as di-galloyl prodelphynidin dimers [40,41,42]. Epigallocatechin gallate and methyl ellagic acid were also detected and their presence was confirmed with an injection of authentic standards. An unusual derivative presenting molecular formula of C14H12O11 was tentatively assigned to Chebulic acid due to the loss of water and CO2,leading to the fragment at m/z 293 that further presented loss of further CO2 moieties leading to the species at m/z 249, 205, and 163; the mass spectra are reported in Supplementary Figure S4. A second peak presenting the same molecular ion and fragmentation pathway was observed and indicated as a chebulic acid isomer (Supplementary Figure S5). These compounds have been identified in the fruits of Terminalia chebula and LC methods have been proposed for their identification [43]. A series of condensed tannins formed by units of epigallo/gallo catechin with or without gallic acid esterification have been also identified [39,44,45]. Furthermore, the peak presenting molecular formula of C44H36O22 was tentatively assigned to the Assamicain chalcan flavan dimer previously identified in Camelia sinensis [46]. Several hydrolysable tannins have been identified specifically in P. meliocarpoides var. meliocarpoides and most of the compounds were ester of gallic acid with glucose, quinic acid; moreover, different derivatives formed by mixed esters of gallic, synaptic, and shikimic acid have been detected, and identification was based on the obtained MS measurements and predicted MS fragmentation from database and software, as well as by comparison with the literature [44,47,48,49,50]. Some coumarin derivatives were identified, especially in the P. meliocarpoides extracts obtained with methanol or water. Coumarins have been reported in Prangos species [8,51], where the identity of compounds was confirmed with the injection of reference purified substances. A series of glycosidic flavonoids, mainly quercetin, myricetin, and isorhamnetin derivatives, were also identified [52,53] and quantified, and they mostly were present in P. uechtritzii.
As clearly shown in Supplementary Figures S1–S3, composition is different considering the various species and extraction solvents. Chromatograms are also given in Supplementary Figures S5 and S6. P. heyniae extracts are characterized by large abundance of condensed tannins (except for the hexane extract, which is rich in 6-hydroxy-3-oxo-alpha ionol), while P. meliocarpoides mostly contains hydrolysable tannins. More complex is the situation of P. uechtritzii, which presents coumarins in the hexane extract, other constituents in the ethyl acetate extract, and flavonoids in methanol and water extracts.
Thus, the three Prangos species differ in composition and the solvent used for the extraction can influence the composition of the obtained products. Positive and negative ion mode LC-MS revealed that the most abundant compounds in the most lipophilic extracts (hexane and ethyl acetate) of P. heyniae are 6-hydroxy-3-oxo-alpha-ionol and the p-hydroxy-benzoic acid rhamnosyl ester. The extraction is influenced by the nature of the solvent and, using methanol and water, other derivatives have been extracted from P. heyniae in higher yields such as epigallocatechin digallate, and galloyl prodelphynidine resulted in more abundant compounds of the methanol and water extracts. The P. heyniae samples also contained the coumarins heraclenin, 8-methoxy coumarin, and marmesin. Considering P. meliocarpoides, all of the different extracts present as the most abundant constituents’ hydrolysable tannins. Nevertheless, chebulic acid (a tricarboxylic acid forming specific ellagitannin derivatives), as well as chebulinic acid (an ellagitannin), were specifically detected only in this species and, to the best of our knowledge, this is the first report of these derivatives in Prangos, while previous identification of such compounds occurred in Terminalia spp and in Phyllantus emblica [54,55]. P. uechtritzii is characterized by the presence of flavonoid glycosides and coumarins, while it contains a negligible amount of condensed or hydrolysable tannin.
Some compounds can be considered characteristic for the different species. For example, condensed tannins, mostly derivatives of epigallocatechin gallate and galloprodelphynidine, are found mainly in P. heyniae. On the other hand, chebulic acid, i.e., a tetracarboxylic acid, was found only in P. meliocarpoides, which is also characterized by the large presence of hydrolysable tannins, which were not detected in the other two species. Considering the coumarins, heraclenin characterized P. heyniae while imperatorin and dentatin were specifically found only in P. meliocarpoides and P. uechtritzii, respectively.
The results revealed that, considering the whole phytochemical profile of extracts obtained from this plant material, methanol and ethyl acetate are the most suitable solvents, despite water and hexane. It is possible that the condensed tannins and epigallocatechin derivatives are not soluble enough, neither in the too nonpolar hexane nor in water. What is notable compared with other Prangos species reported in the literature is the limited amount of detected coumarins, as only low amounts of imperatorin were detected.
A similar behavior was observed for P. meliocarpoides. In fact, the LC-MS analysis revealed that more compounds were detected in the ethyl acetate and methanol extracts. The main detected compounds in this second case were tetragalloyl quinic acid and digalloyltheaflavonin. Marmesin and imperatorin were also detected, showing the presence of different coumarins compared with the previous plant species. Furthermore, condensed tannins as punicalagins; the ellagitannin terchebulin; as well as esters of gallic, shikimic, and synapoyl acid with glucose represented the most notable constituents. P. uechtritzii extracts were characterized by the presence of a large amount of the coumarins 8-methoxy coumarin and marmesin, as well as a significant amount of epigallocatechin-gallate. Furthermore, this species was the only one showing a pattern of glycosidic flavonoids, myricetin, and quercetin.

3.2. Antioxidant Effects

The antioxidant activities of P. heyniae, P. meliocarpoides var. meliocarpoides, and P. uechtritzii extracts were evaluated using different assays. In our model, DPPH and ABTS assays were used to assess radical quenching ability, while FRAP and CUPRAC assays results indicated reducing power. The capacity of extracts to chelate ferrous ions was measured by metal chelating assay, while the phosphomolybdenum test was useful to establish the total antioxidant capacity. Each assay presents strengths and limitations, as detailed elsewhere [56]. The results are presented in Table 1 and Table 3.
Overall, the findings showed that a correlation exist between DPPH, ABTS, FRAP, and CUPRAC assays, except with the metal chelating and phosphomolybdenum assay. Such a correlation was observed in several other studies [56,57,58,59]. The methanolic and aqueous extracts exhibited the highest antioxidant activities in most of the assays, suggesting an important role of the phenolics and flavonoids of the extracts. The polarity-dependent increase in total antioxidant activity and reducing properties indicates that the extraction of strong antioxidant compounds is more favorable in polar solvents [60]. Furthermore, in a previous paper that highly polar solvents, such as methanol, can have a high effectiveness in the extraction of antioxidants [35], supporting our findings.
Among the tested Prangos species, P. meliocarpoides var. meliocarpoides exhibited the strongest antioxidant activity in DPPH (52.27 mg TE/g), ABTS (92.84 mg TE/g), CUPRAC (154.04 mg TE/g), FRAP (104.34 mg TE/g), and phosphomolybdenum (2.52 mmol TE/g) assays. However, P. heyniae displayed the highest metal chelation effects (27.79 mg EDTAE/g) considering metal-chelating properties. Previous studies have also reported a weak correlation between metal-chelating assays and DPPH, ABTS, CUPRAC, and FRAP methods [61,62,63,64]. An explanation of this discrepancy is related to the different chemical reactions involved in the assays. The DPPH, ABTS, CUPRAC, FRAP, and phosphomolybdenum assays are single-electron transfer or hydrogen atom transfer reaction-based assays. On the other hand, the metal-chelating assay involves sequestration of transition metals [56]. This can justify the difference in the results obtained with DPPH, ABTS, CUPRAC, and FRAP methods and metal-chelating tests.

3.3. Enzyme Inhibitory Activities

In the present study, the ability of P. heyniae, P. meliocarpoides var. meliocarpoides, and P. uechtritzii extracts to modulate the activity of enzymes related to Alzheimer’s disease (acetylcholinesterase (AChE) and butyrylcholinesterase (BChE)), diabetes type 2 (α-amylase and α-glucosidase), and skin hyperpigmentation (tyrosinase) was investigated, and the results are presented in Table 4.
Although AChE inhibition is regarded as a very promising technique for symptomatic therapy of Alzheimer’s disease, the involvement of BChE in late Alzheimer’s disease has been established [65,66]. Among the prepared extracts, hexane extracts demonstrated the highest anti-AChE and anti-BChE activities for the three Prangos species, despite that these extracts yielded the lowest phenolic and flavonoid content. In terms of plants, P. heyniae demonstrated the highest inhibitory property against AChE (2.39 mg GALAE/g). However, the three plants exhibited relatively similar activity against BChE, with galantamine equivalent ranging from 7.63 to 7.97 mg GALAE/g. Taken together, the three members of the Prangos genus could be considered as valuable sources of AChE and BChE inhibitors. It is important to highlight that, despite that aqueous extracts possessed high levels of phenolic and flavonoid, the extracts were not good inhibitors of AChE and BChE. In an earlier study by Abbas-Mohammadi et al. [67], who tested twenty-five Iranian plants for AChE inhibition, found the n-hexane fraction of P. ferulacea to be the most active (75.6%) at 50 µg/mL concentration, and the results are consistent with our presented results, where n-hexane extracts were the most active on AChE. In addition, several furanocoumarins were isolated in their study, which showed more potent AChE inhibitory effects with low IC50 values. However, we did not find a good correlation between coumarins and AChE inhibitory properties of the tested extracts. This fact could be explained by the interaction of phytochemicals (antagonistic and so on) in the present study. Thus, we strongly suggested to conduct further studies on the cholinesterase inhibitory properties of isolated compounds from the tested Prangos species. In another study by Bahadori [68], the AChE and BChE inhibitory effects of P. gaubae extracts (n-hexane, dichloromethane, and methanol) were detected, and the dichloromethane extract was the most active on both enzymes (AChE: 2.62 mg GALAE/g; BChE: 3.51 mg GALAE/g). The AChE inhibitory effects for the methanol extracts of P. ferulacea and P. peucadanifolia were also reported as 1.47 and 4.09 mg GALAE/g, respectively [69].
α-Amylase and α-glucosidase inhibitors delay the breakdown of carbohydrates in the small intestine and, as a consequence, decrease the post-prandial blood glucose level, which is considered as an important treatment strategy to manage blood glucose levels in type 2 diabetic patients [70]. As shown in Table 4, the three Prangos species exhibited similar inhibition against α-amylase enzyme (0.40–0.46 mmol ACAE/g). However, the highest anti-glucosidase activity was observed with P. uechtritzii (0.78 mmol ACAE/g), followed by P. meliocarpoides var. meliocarpoides (0.74 mmol ACAE/g) and P. heyniae (0.67 mmol ACAE/g). No activity against α-glucosidase was noted with aqueous extracts. Inhibition of α-amylase and α-glucosidase enzymes has been reported to be an intriguing target for the management of type II diabetes because of less side effects when compared with standard therapies [71,72]. Few studies have been reported in the literature on the amylase and glucosidase inhibitory effects of members of the genus Prangos. Loizzo et al. [73] tested the amylase and glucosidase inhibitory activity of P. asperula extracts (n-hexane, chloroform, and methanol), and the n-hexane extract was the most active for the enzymes, consistent with our findings. However, the best amylase and glucosidase inhibitory activity was detected by dicholoromethane extract (amylase: 0.93 mmol ACAE/g and glucosidase: 20.07 mmol ACAE/g) of P. gaubae [68]. The amylase and glucoside inhibitory properties were also reported for P. ferulacea (amylase: 0.77 mmol ACAE/g and glucoside: 4.45 mmol ACAE/g) and P. peucadanifolia (amylase: 0.83 mmol ACAE/g and glucosidase: 4.97 mmol ACAE/g) [69]. From Figure 1, the observed amylase and glucosidase inhibitory abilities of the tested extracts were also moderately correlated with punicalagins and the compounds were reported as antidiabetic agents in earlier studies [74,75].
Tyrosinase is a rate-limiting enzyme that is responsible for the manufacture of melanin, and it is regarded as a critical therapeutic approach for the treatment of skin hyperpigmentation problems [76]. The methanolic extract of P. meliocarpoides var. meliocarpoides displayed the best tyrosinase inhibitory effect (81.15 mg KAE/g), followed by P. uechtritzii (68.03 mg KAE/g) and P. heyniae (65.20 mg KAE/g). Aqueous extracts exhibited the lowest inhibition against tyrosinase (Table 4). In a previous study by Zengin et al. [69], the tyrosinase inhibitory properties of the methanol extract of P. ferulacea (131.94 mg KAE/g) and P. peucadanifolia (128.54 mg KAE/g) were higher than those of the presented study. The tyrosinase inhibitory properties of the extracts of P. gaubae varied from 16.85 mg KAE/g (in methanol) to 36.33 mg KAE/g (in n-hexane) [68].
Heatmaps were generated to establish possible correlations between the chemical composition of extracts and the observed bioactivities. One of these plots considered flavonoids and coumarins, while the second considered all of the other polyphenols (tannins and other constituents). These heatmaps are reported in Figure 1 and Figure 2, respectively. Clearly, the results show a strong positive correlation between antioxidant activities and all of the different tannins, as well as between prodelphynidine and total flavonoid content. Considering the enzymatic activities, epigallocatechin derivatives were moderately correlated with AChE inhibition, suggesting a potential role of these compounds on this specific enzyme. As reported by Jabir et al. [77], different polyphenols have been considered as potential inhibitors of AChE, but their mode of action should be specifically studied, considering not only the possible different interactions with the active site of the enzyme, but also the potential modification to polyphenol structures due to metabolism of the host. Thus, our data suggest a possible role of gallocatechin derivatives as potential inhibitors of AChE, and they can lead to further studies on these natural chemical compounds.
Furthermore, the OPLS model was generated using the data from the chemical analysis and combining the results of the bioassays. Figure 3 shows the score scatter plot, while the loading plot is shown in Figure 4.
In the score scatter plot (Figure 3), a clear distinction can be observed considering the extracts obtained with the lipophilic solvents, namely hexane and ethyl acetate, and the extract obtained with the more hydrophilic methanol and water, with the first being all in the -x part of the plot and the other at the opposite side. As shown by the loading plot (Figure 4) and in the biplot (Figure 5), the more relevant results related to antioxidant activities are obtained with the methanol and water extracts, while the enzyme inhibitory activities appear to be related mostly to the more lipophilic extracts. This result suggests proceeding with the studies using the water and methanol extracts of Prangos species as sources of antioxidant constituents, while using the lipophilic solvents to extract compounds that can be evaluated as inhibitors of cholinesterase, tyrosinase, amylase, or glucosidase. Considering the other assays, we can observe that CUPRAC appears to be related to the hydrophylic extracts, while the metal-chelating assay results were ascribed to lipophilic extracts. Thus, the application of multivariate analysis can be used to select the proper extraction solvent to improve or to search for compounds or mixtures presenting efficient activity on target assays.

4. Conclusions

In this study, the different phytochemical composition and biological activities (antioxidant and enzyme inhibitory effects) of three Prangos species, namely, P. heyniae, P. meliocarpoides var. meliocarpoides, and P. uechtritzii, are described. The present study is the first report on the detailed chemical profiling and biological activities of the tested species. Thus, this work could provide valuable contributions to the scientific pool for the member of the genus Prangos. For each Prangos species, the chemical profile and biological activity depended on the extraction solvents used. Among these species, P. meliocarpoides var. meliocarpoides yielded the highest phenolic and flavonoid contents in the obtained extracts. Additionally, in terms of biological assays, P. meliocarpoides var. meliocarpoides displayed the most potent antioxidant activity as well as a significant inhibitory property on BChE, α-amylase, and tyrosinase, and these effects appear to be correlated to the presence of hydrolysable tannins. This research work presents valuable preliminary data on the three members of Prangos genus, displaying P. meliocarpoides var. meliocarpoides as the most promising one. However, further investigations such as in vivo bioavailability and toxicity studies need to be performed in the future, before projecting the plant for possible nutraceutical/functional food and/or pharmaceutical applications.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/antiox11091712/s1, Figure S1: Pie charts summarising the relative percentage of phytochemicals identified and quantified in P. heyniae extracts; Figure S2: Pie charts summarising the relative percentage of phytochemicals identified and quantified in P. meliocarpoides extracts; Figure S3: Pie charts summarising the relative percentage of phytochemicals identified and quantified in P.uechtritzii extracts; Figure S4: MSn spectra showing the loss of water and proposed fragmentation for the peak assigned to chebulic acid; Figure S5: MSn spectra showing the loss of water and proposed fragmentation for the peak assigned to chebulic acid.

Author Contributions

Conceptualization, S.D., S.S. and G.Z.; methodology; S.D., S.S. and G.Z., software, S.D., S.S. and G.P.; validation, G.Z., E.Y. and F.E.; formal analysis, G.Z.; investigation, S.D., S.S., G.Z. and N.B.S.; resources, G.Z. and E.Y.; data curation, G.Z.; writing—original draft preparation, S.D., S.S., G.Z., N.B.S. and M.F.M.; writing—review and editing, S.D., S.S. and G.Z.; visualization, G.P.; supervision, S.D.; project administration; G.Z.; funding acquisition, S.D. 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

The data are contained within the article and supplementary materials.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Huang, B.; Zhang, Y. Teaching an old dog new tricks: Drug discovery by repositioning natural products and their derivatives. Drug Discov. Today 2022, 27, 1936–1944. [Google Scholar] [CrossRef] [PubMed]
  2. Rastelli, G.; Pellati, F.; Pinzi, L.; Gamberini, M.C. Repositioning natural products in drug discovery. Molecules 2020, 25, 1154. [Google Scholar] [CrossRef]
  3. Atanasov, A.G.; Waltenberger, B.; Pferschy-Wenzig, E.-M.; Linder, T.; Wawrosch, C.; Uhrin, P.; Temml, V.; Wang, L.; Schwaiger, S.; Heiss, E.H.; et al. Discovery and resupply of pharmacologically active plant-derived natural products: A review. Biotechnol. Adv. 2015, 33, 1582–1614. [Google Scholar] [CrossRef]
  4. Atanasov, A.G.; Zotchev, S.B.; Dirsch, V.M.; Supuran, C.T. Natural products in drug discovery: Advances and opportunities. Nature Rev. Drug Discov. 2021, 20, 200–216. [Google Scholar]
  5. Rammohan, A.; Bhaskar, B.V.; Camilo, A.; Gunasekar, D.; Gu, W.; Zyryanov, G.V. In Silico, In Vitro antioxidant and density functional theory based structure activity relationship studies of plant polyphenolics as prominent natural antioxidants. Arab. J. Chem. 2020, 13, 3690–3701. [Google Scholar] [CrossRef]
  6. Efferth, T.; Koch, E. Complex interactions between phytochemicals. The multi-target therapeutic concept of phytotherapy. Curr. Drug Targets 2011, 12, 122–132. [Google Scholar] [CrossRef]
  7. Sut, S.; Baldan, V.; Faggian, M.; Peron, G.; Dall Acqua, S. Nutraceuticals, a New Challenge for Medicinal Chemistry. Curr. Med. Chem. 2016, 23, 3198–3223. [Google Scholar] [CrossRef] [PubMed]
  8. Mottaghipisheh, J.; Kiss, T.; Tóth, B.; Csupor, D. The Prangos genus: A comprehensive review on traditional use, phytochemistry, and pharmacological activities. Phytochem. Rev. 2020, 19, 1449–1470. [Google Scholar] [CrossRef]
  9. Başer, K.; Kırımer, N. Essential oils of Anatolian Apiaceae—A profile. Nat. Volatiles Essent. Oils 2014, 1, 1–50. [Google Scholar]
  10. Bruno, M.; Ilardi, V.; Lupidi, G.; Quassinti, L.; Bramucci, M.; Fiorini, D.; Venditti, A.; Maggi, F. The nonvolatile and volatile metabolites of Prangos ferulacea and their biological properties. Planta Med. 2019, 85, 815–824. [Google Scholar] [CrossRef]
  11. Bruno, M.; Ilardi, V.; Lupidi, G.; Quassinti, L.; Bramucci, M.; Fiorini, D.; Venditti, A.; Maggi, F. Composition and biological activities of the essential oil from a Sicilian accession of Prangos ferulacea (L.) Lindl. Nat. Prod. Res. 2021, 35, 733–743. [Google Scholar] [CrossRef] [PubMed]
  12. Delnavazi, M.R.; Soleimani, M.; Hadjiakhoondi, A.; Yass, N. Isolation of phenolic derivatives and essential oil analysis of Prangos ferulacea (L.) Lindl. Aerial Parts. Iran. J. Pharm. Res. IJPR 2017, 16, 207–215. [Google Scholar] [PubMed]
  13. Yousefi, K.; Hamedeyazdan, S.; Hodaei, D.; Lotfipour, F.; Baradaran, B.; Orangi, M.; Fathiazad, F. An In Vitro ethnopharmacological study on Prangos ferulacea: A wound healing agent. Bioimpacts 2017, 7, 75–82. [Google Scholar] [CrossRef]
  14. Shokoohinia, Y.; Sajjadi, S.E.; Gholamzadeh, S.; Fattahi, A.; Behbahani, M. Antiviral and cytotoxic evaluation of coumarins from Prangos ferulacea. Pharm. Biol. 2014, 52, 1543–1549. [Google Scholar] [CrossRef]
  15. Azadpour Motlagh, A.M.S.S.; Dolatian, M.P.; Mojab, F.P.; Nasiri, M.P.; Ezatpour, B.P.; Sahranavard, Y.M.; Shakiba, H.B.; Rahimy, B.M.; Ghanati, K. The effect of Prangos ferulacea vaginal cream on accelerating the aecovery of bacterial vaginosis: A randomized controlled clinical trial. Int. J. Community Based Nurs. Midwifery 2018, 6, 100–110. [Google Scholar]
  16. Numonov, S.; Bobakulov, K.; Numonova, M.; Sharopov, F.; Setzer, W.N.; Khalilov, Q.; Begmatov, N.; Habasi, M.; Aisa, H.A. New coumarin from the roots of Prangos pabularia. Nat. Prod. Res. 2018, 32, 2325–2332. [Google Scholar] [CrossRef]
  17. Dissanayake, A.A.; Ameen, B.A.H.; Nair, M.G. Lipid peroxidation and cyclooxygenase enzyme inhibitory compounds from Prangos haussknechtii. J. Nat. Prod. 2017, 80, 2472–2477. [Google Scholar] [CrossRef]
  18. Tan, N.; Yazıcı-Tütüniş, S.; Bilgin, M.; Tan, E.; Miski, M. Antibacterial activities of pyrenylated coumarins from the roots of Prangos hulusii. Molecules 2017, 22, 1098. [Google Scholar] [CrossRef]
  19. Ahmed, J.; Güvenç, A.; Küçükboyaci, N.; Baldemir, A.; Coşkun, M. Total phenolic contents and antioxidant activities of Prangos Lindl.(Umbelliferae) species growing in Konya province (Turkey). Turk. J. Biol. 2011, 35, 353–360. [Google Scholar] [CrossRef]
  20. Albayrak, G.; Demir, S.; Kose, F.A.; Baykan, S. New coumarin glycosides from endemic Prangos heyniae H. Duman & M.F. Watson. Nat. Prod. Res. 2021, 1–13. [Google Scholar] [CrossRef]
  21. Karahisar, E.; Köse, Y.B.; İşcan, G.; Kurkcuoglu, M.; Tugay, O. Chemical composition and anticandidal activity of essential oils obtained from different parts of Prangos heyniae H. Duman & MF Watson. Rec. Nat. Prod. 2022, 16, 74–83. [Google Scholar]
  22. Özek, G.; Bedir, E.; Tabanca, N.; Ali, A.; Khan, I.A.; Duran, A.; Başer, K.H.; Özek, T. Isolation of eudesmane type sesquiterpene ketone from Prangos heyniae H. Duman & MF Watson essential oil and mosquitocidal activity of the essential oils. Open Chem. 2018, 16, 453–467. [Google Scholar]
  23. Oke Altuntas, F.; Aslim, B.; Duman, H. The anti-lipid peroxidative, metal chelating, and radical scavenging properties of the fruit extracts from endemic Prangos meliocarpoides Boiss var. meliocarpoides. Gazi Univ. J. Sci. 2016, 29, 537–542. [Google Scholar]
  24. Uzel, A.; Dirmenci, T.; Celik, A.; Arabaci, T. Composition and antimicrobial activity of Prangos platychlaena and P-uechtritzii. Chem. Nat. Compd. 2006, 42, 169–171. [Google Scholar] [CrossRef]
  25. Ozcan, M.; Bagci, Y.; Akgul, A.; Dural, H.; Novak, J. Chemical composition of the essential oil of Prangos uechtritzii Boiss. et Hausskn. fruits from Turkey. J. Essent. Oil Res. 2000, 12, 183–185. [Google Scholar] [CrossRef]
  26. Ozcan, M. Antifungal effects of Micromeria myrtifolia Boiss. & Hohen. in Boiss. and Prangos uechtritzii Boiss. Hawsskn decoctions. Acta Aliment. 1999, 28, 355–360. [Google Scholar]
  27. Sevin, G.; Alan, E.; Demir, S.; Albayrak, G.; Demiroz, T.; Yetik-Anacak, G.; Baykan, S. Comparative evaluation of relaxant effects of three Prangos species on mouse corpus cavernosum: Chemical characterization and the relaxant mechanisms of action of P. pabularia and (+)-oxypeucedanin. J. Ethnopharmacol. 2022, 284, 114823. [Google Scholar] [CrossRef]
  28. Zengin, G.; Mahomoodally, M.F.; Yıldıztugay, E.; Jugreet, S.; Khan, S.U.; Dall’Acqua, S.; Mollica, A.; Bouyahya, A.; Montesano, D. Chemical composition, biological activities and in silico analysis of essential oils of three endemic Prangos species from Turkey. Molecules 2022, 27, 1676. [Google Scholar] [CrossRef]
  29. 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. Trad. Complement. Alt. Med. 2014, 11, 481–488. [Google Scholar] [CrossRef]
  30. Dall’Acqua, S.; Sinan, K.I.; Sut, S.; Ferrarese, I.; Etienne, O.K.; Mahomoodally, M.F.; Lobine, D.; Zengin, G. Evaluation of antioxidant and enzyme inhibition properties of Croton hirtus L’Hér. extracts obtained with different solvents. Molecules 2021, 26, 1902. [Google Scholar] [CrossRef]
  31. 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]
  32. 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]
  33. Deen, F.; Visvanathan, R.; Liyanage, R. Extraction Methods: Conventional and non-conventional. In Bioactive Compounds from Plant Origin; CRC Press: Boca Raton, FL, USA, 2019; p. 24. [Google Scholar]
  34. Zhang, Q.W.; Lin, L.G.; Ye, W.C. Techniques for extraction and isolation of natural products: A comprehensive review. Chin. Med. 2018, 13, 20. [Google Scholar] [PubMed]
  35. Altemimi, A.; Lakhssassi, N.; Baharlouei, A.; Watson, D.G.; Lightfoot, D.A. Phytochemicals: Extraction, isolation, and identification of bioactive compounds from plant extracts. Plants 2017, 6, 42. [Google Scholar] [CrossRef]
  36. Abarca-Vargas, R.; Peña Malacara, C.F.; Petricevich, V.L. Characterization of chemical compounds with antioxidant and cytotoxic activities in Bougainvillea x buttiana Holttum and Standl, (var. rose) extracts. Antioxidants 2016, 5, 45. [Google Scholar] [CrossRef]
  37. Abdul Razak, M.; Yong, P.; Shah, Z.; Abdullah, L.; Yee, S.; Yaw, I. The effects of varying solvent polarity on extraction yield of Orthosiphon stamineus leaves. J. Appl. Sci. 2012, 12, 1207–1210. [Google Scholar] [CrossRef]
  38. Lee, R.J.; Lee, V.S.; Tzen, J.T.; Lee, M.R. Study of the release of gallic acid from (–)-epigallocatechin gallate in old oolong tea by mass spectrometry. Rapid Commun. Mass Spectrom. 2010, 24, 851–858. [Google Scholar]
  39. De Lima Paula, P.; de Oliveira Lemos, A.S.; Campos, L.M.; Ferreira, T.G.; de Souza, T.F.; Queiroz, L.S.; Guedes, M.C.M.R.; Martins, M.M.; Goulart Filho, L.R.; Macedo, G.C. Pharmacological investigation of antioxidant and anti-inflammatory activities of leaves and branches extracts from Plinia cauliflora (Jaboticaba). J. Ethnopharmacol. 2021, 280, 114463. [Google Scholar] [CrossRef]
  40. Cheng, H.-Y.; Lin, C.-C.; Lin, T.-C. Antiviral properties of prodelphinidin B-2 3′-O-gallate from green tea leaf. Antivir. Chem. Chemother. 2002, 13, 223–229. [Google Scholar] [CrossRef] [Green Version]
  41. Li, M.; Shen, Y.; Ling, T.; Ho, C.-T.; Li, D.; Guo, H.; Xie, Z. Analysis of differentiated chemical components between Zijuan purple tea and Yunkang green tea by UHPLC-Orbitrap-MS/MS Combined with Chemometrics. Foods 2021, 10, 1070. [Google Scholar] [CrossRef] [PubMed]
  42. Kumar, S.; Singh, A.; Kumar, B. Identification and characterization of phenolics and terpenoids from ethanolic extracts of Phyllanthus species by HPLC-ESI-QTOF-MS/MS. J. Pharm. Anal. 2017, 7, 214–222. [Google Scholar] [CrossRef] [PubMed]
  43. Juang, L.J.; Sheu, S.J.; Lin, T.C. Determination of hydrolyzable tannins in the fruit of Terminalia chebula Retz. by high-performance liquid chromatography and capillary electrophoresis. J. Sep. Sci. 2004, 27, 718–724. [Google Scholar] [CrossRef]
  44. Lee, D.Y.; Kim, H.W.; Yang, H.; Sung, S.H. Hydrolyzable tannins from the fruits of Terminalia chebula Retz and their α-glucosidase inhibitory activities. Phytochemistry 2017, 137, 109–116. [Google Scholar] [CrossRef] [PubMed]
  45. Silva, O.; Gomes, E.T.; Wolfender, J.-L.; Marston, A.; Hostettmann, K. Application of high performance liquid chromatography coupled with ultraviolet spectroscopy and electrospray mass spectrometry to the characterisation of ellagitannins from Terminalia macroptera roots. Pharm. Res. 2000, 17, 1396–1401. [Google Scholar] [CrossRef]
  46. Hashimoto, F.; Nonaka, G.-i.; Nishioka, I. Tannins and Related Compounds. LXXVII.: Novel Chalcan-flavan Dimers, Assamicains A, B and C, and a New Flavan-3-ol and Proanthocyanidins from the fresh leaves of Camella sinensis L. var. assamica kitamura. Chem. Pharm. Bull. 1989, 37, 77–85. [Google Scholar] [CrossRef]
  47. Wyrepkowski, C.C.; Gomes da Costa, D.L.M.; Sinhorin, A.P.; Vilegas, W.; De Grandis, R.A.; Resende, F.A.; Varanda, E.A.; Dos Santos, L.C. Characterization and quantification of the compounds of the ethanolic extract from Caesalpinia ferrea stem bark and evaluation of their mutagenic activity. Molecules 2014, 19, 16039–16057. [Google Scholar] [CrossRef] [PubMed]
  48. Singh, A.; Bajpai, V.; Kumar, S.; Sharma, K.R.; Kumar, B. Profiling of gallic and ellagic acid derivatives in different plant parts of Terminalia arjuna by HPLC-ESI-QTOF-MS/MS. Nat. Prod. Comm. 2016, 11, 1934578X1601100227. [Google Scholar]
  49. Ben Said, R.; Arafa, I.H.; Usam, A.M.; Abdullah Sulaiman, A.-A.; Kowalczyk, M.; Moldoch, J.; Oleszek, W.; Stochmal, A. Tentative characterization of polyphenolic compounds in the male flowers of Phoenix dactylifera by liquid chromatography coupled with mass spectrometry and DFT. Int. J. Mol. Sci. 2017, 18, 512. [Google Scholar] [CrossRef]
  50. Barry, K.; Davies, N.; Mohammed, C. Identification of hydrolysable tannins in the reaction zone of Eucalyptus nitens wood by high performance liquid chromatography–electrospray ionisation mass spectrometry. Phytochem. Anal. 2001, 12, 120–127. [Google Scholar] [CrossRef]
  51. Farooq, S.; Dangroo, N.A.; Priya, D.; Banday, J.A.; Sangwan, P.L.; Qurishi, M.A.; Koul, S.; Saxena, A.K. Isolation, cytotoxicity evaluation and HPLC-quantification of the chemical constituents from Prangos pabularia. PLoS ONE 2014, 9, e108713. [Google Scholar]
  52. Schieber, A.; Keller, P.; Streker, P.; Klaiber, I.; Carle, R. Detection of isorhamnetin glycosides in extracts of apples (Malus domestica cv.“Brettacher”) by HPLC-PDA and HPLC-APCI-MS/MS. Phytochem. Anal. 2002, 13, 87–94. [Google Scholar] [CrossRef] [PubMed]
  53. Mosić, M.; Trifković, J.; Vovk, I.; Gašić, U.; Tešić, Ž.; Šikoparija, B.; Milojković-Opsenica, D. Phenolic composition influences the health-promoting potential of bee-pollen. Biomolecules 2019, 9, 783. [Google Scholar] [CrossRef] [PubMed]
  54. Feng, X.-H.; Xu, H.-Y.; Wang, J.-Y.; Duan, S.; Wang, Y.-C.; Ma, C.-M. In Vivo hepatoprotective activity and the underlying mechanism of chebulinic acid from Terminalia chebula fruit. Phytomedicine 2021, 83, 153479. [Google Scholar] [CrossRef] [PubMed]
  55. Singh, E.; Sharma, S.; Pareek, A.; Dwivedi, J.; Yadav, S.; Sharma, S. Phytochemistry, traditional uses and cancer chemopreventive activity of Amla (Phyllanthus emblica): The Sustainer. J. Appl. Pharm. Sci. 2012, 2, 176–183. [Google Scholar]
  56. Bibi Sadeer, N.; Montesano, D.; Albrizio, S.; Zengin, G.; Mahomoodally, M.F. The versatility of antioxidant assays in food science and safety—chemistry, applications, strengths, and limitations. Antioxidants 2020, 9, 709. [Google Scholar] [CrossRef]
  57. Dall’Acqua, S.; Kumar, G.; Sinan, K.I.; Sut, S.; Ferrarese, I.; Mahomoodally, M.F.; Seebaluck-Sandoram, R.; Etienne, O.K.; Zengin, G. An insight into Cochlospermum planchonii extracts obtained by traditional and green extraction methods: Relation between chemical compositions and biological properties by multivariate analysis. Ind. Crops Prod. 2020, 147, 112226. [Google Scholar] [CrossRef]
  58. Lazarova, I.; Zengin, G.; Sinan, K.I.; Aneva, I.; Uysal, S.; Picot-Allain, M.C.N.; Aktumsek, A.; Bouyahya, A.; Mahomoodally, M.F. Metabolomics profiling and biological properties of root extracts from two Asphodelus species: A. albus and A. aestivus. Food Res. Int. 2020, 134, 109277. [Google Scholar] [CrossRef]
  59. Llorent-Martínez, E.J.; Zengin, G.; Sinan, K.I.; Polat, R.; Canlı, D.; Picot-Allain, M.C.N.; Mahomoodally, M.F. Impact of different extraction solvents and techniques on the biological activities of Cirsium yildizianum (Asteraceae: Cynareae). Ind. Crops Prod. 2020, 144, 112033. [Google Scholar] [CrossRef]
  60. Nawaz, H.; Shad, M.A.; Rehman, N.; Andaleeb, H.; Ullah, N. Effect of solvent polarity on extraction yield and antioxidant properties of phytochemicals from bean (Phaseolus vulgaris) seeds. Braz. J. Pharm. Sci. 2020, 56. [Google Scholar] [CrossRef]
  61. Sadeer, N.B.; Rocchetti, G.; Senizza, B.; Montesano, D.; Zengin, G.; Uysal, A.; Jeewon, R.; Lucini, L.; Mahomoodally, M.F. Untargeted metabolomic profiling, multivariate analysis and biological evaluation of the true mangrove (Rhizophora mucronata Lam.). Antioxidants 2019, 8, 489. [Google Scholar] [CrossRef]
  62. Bibi Sadeer, N.; Sinan, K.I.; Cziáky, Z.; Jekő, J.; Zengin, G.; Jeewon, R.; Abdallah, H.H.; Rengasamy, K.R.R.; Fawzi Mahomoodally, M. Assessment of the pharmacological properties and phytochemical profile of Bruguiera gymnorhiza (l.) lam using in vitro studies, in silico docking, and multivariate analysis. Biomolecules 2020, 10, 731. [Google Scholar] [CrossRef] [PubMed]
  63. Dall’Acqua, S.; Sinan, K.I.; Ferrarese, I.; Sut, S.; Bene, K.; Mahomoodally, M.F.; Sadeer, N.B.; Ak, G.; Zengin, G. Chromatographic separation of Breynia retusa (Dennst.) Alston bark, fruit and leaf constituents from bioactive extracts. Molecules 2020, 25, 5537. [Google Scholar] [CrossRef]
  64. 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]
  65. Darvesh, S. Butyrylcholinesterase as a diagnostic and therapeutic target for Alzheimer’s disease. Curr. Alzheimer. Res. 2016, 13, 1173–1177. [Google Scholar] [CrossRef] [PubMed]
  66. DeBay, D.R.; Darvesh, S. Chapter 17-Butyrylcholinesterase as a biomarker in Alzheimer’s disease. In Diagnosis and Management in Dementia; Martin, C.R., Preedy, V.R., Eds.; Academic Press: Cambridge, MA, USA, 2020; pp. 263–280. [Google Scholar]
  67. Abbas-Mohammadi, M.; Moridi Farimani, M.; Salehi, P.; Nejad Ebrahimi, S.; Sonboli, A.; Kelso, C.; Skropeta, D. Acetylcholinesterase-inhibitory activity of Iranian plants: Combined HPLC/bioassay-guided fractionation, molecular networking and docking strategies for the dereplication of active compounds. J. Pharm. Biomed. Anal. 2018, 158, 471–479. [Google Scholar] [CrossRef] [PubMed]
  68. Bahadori, M.B.; Zengin, G.; Bahadori, S.; Maggi, F.; Dinparast, L. Chemical composition of essential oil, antioxidant, antidiabetic, anti-obesity, and neuroprotective properties of Prangos gaubae. Nat. Prod. Comm. 2017, 12, 1934578X1701201233. [Google Scholar] [CrossRef]
  69. Zengin, G.; Sinan, K.I.; Ak, G.; Mahomoodally, M.F.; Paksoy, M.Y.; Picot-Allain, C.; Glamocilja, J.; Sokovic, M.; Jekő, J.; Cziáky, Z.; et al. Chemical profile, antioxidant, antimicrobial, enzyme inhibitory, and cytotoxicity of seven Apiaceae species from Turkey: A comparative study. Ind. Crops Prod. 2020, 153, 112572. [Google Scholar] [CrossRef]
  70. Tundis, R.; Loizzo, M.R.; Menichini, F. Natural products as alpha-amylase and alpha-glucosidase inhibitors and their hypoglycaemic potential in the treatment of diabetes: An update. Mini Rev. Med. Chem. 2010, 10, 315–331. [Google Scholar] [CrossRef]
  71. Papoutsis, K.; Zhang, J.; Bowyer, M.C.; Brunton, N.; Gibney, E.R.; Lyng, J. Fruit, vegetables, and mushrooms for the preparation of extracts with α-amylase and α-glucosidase inhibition properties: A review. Food Chem. 2021, 338, 128119. [Google Scholar] [CrossRef]
  72. Aleixandre, A.; Gil, J.V.; Sineiro, J.; Rosell, C.M. Understanding phenolic acids inhibition of α-amylase and α-glucosidase and influence of reaction conditions. Food Chem. 2022, 372, 131231. [Google Scholar] [CrossRef]
  73. Loizzo, M.R.; Saab, A.M.; Tundis, R.; Menichini, F.; Bonesi, M.; Piccolo, V.; Statti, G.A.; de Cindio, B.; Houghton, P.J.; Menichini, F. In Vitro inhibitory activities of plants used in Lebanon traditional medicine against angiotensin converting enzyme (ACE) and digestive enzymes related to diabetes. J. Ethnopharmacol. 2008, 119, 109–116. [Google Scholar] [CrossRef] [PubMed]
  74. Bellesia, A.; Verzelloni, E.; Tagliazucchi, D. Pomegranate ellagitannins inhibit α-glucosidase activity in vitro and reduce starch digestibility under simulated gastro-intestinal conditions. Int. J. Food Sci.Nutr. 2015, 66, 85–92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Les, F.; Arbonés-Mainar, J.M.; Valero, M.S.; López, V. Pomegranate polyphenols and urolithin A inhibit α-glucosidase, dipeptidyl peptidase-4, lipase, triglyceride accumulation and adipogenesis related genes in 3T3-L1 adipocyte-like cells. J. Ethnopharmacol. 2018, 220, 67–74. [Google Scholar] [CrossRef] [PubMed]
  76. Zolghadri, S.; Bahrami, A.; Hassan Khan, M.T.; Munoz-Munoz, J.; Garcia-Molina, F.; Garcia-Canovas, F.; Saboury, A.A. A comprehensive review on tyrosinase inhibitors. J. Enzym. Inhib. Med. Chem. 2019, 34, 279–309. [Google Scholar] [CrossRef]
  77. Jabir, N.R.; Khan, F.R.; Tabrez, S. Cholinesterase targeting by polyphenols: A therapeutic approach for the treatment of Alzheimer’s disease. CNS Neurosci. Ther. 2018, 24, 753–762. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Correlation heatmap showing positive (red colour) and negative (blue colour) correlations between flavonoids and coumarins and measured bioactivities. Color scale is dependent on Pearson’s r value. Significance levels are indicated by asterisks in the plot. *: p < 0.05; **: p < 0.01; ***: p < 0.001.
Figure 1. Correlation heatmap showing positive (red colour) and negative (blue colour) correlations between flavonoids and coumarins and measured bioactivities. Color scale is dependent on Pearson’s r value. Significance levels are indicated by asterisks in the plot. *: p < 0.05; **: p < 0.01; ***: p < 0.001.
Antioxidants 11 01712 g001
Figure 2. Correlation heatmap showing positive (red colour) and negative (blue colour) correlations between specific polyphenols and measured bioactivities. Color scale is dependent on Pearson’s r value. Significance levels are indicated by asterisks in the plot. *: p < 0.05; **: p < 0.01; ***: p < 0.001.
Figure 2. Correlation heatmap showing positive (red colour) and negative (blue colour) correlations between specific polyphenols and measured bioactivities. Color scale is dependent on Pearson’s r value. Significance levels are indicated by asterisks in the plot. *: p < 0.05; **: p < 0.01; ***: p < 0.001.
Antioxidants 11 01712 g002
Figure 3. OPLS model considering all of the prepared extracts categorized on the basis of the solvents, HEX: hexane, EA: ethyl acetate, M: methanol, and W: water.
Figure 3. OPLS model considering all of the prepared extracts categorized on the basis of the solvents, HEX: hexane, EA: ethyl acetate, M: methanol, and W: water.
Antioxidants 11 01712 g003
Figure 4. OPLS loading plot considering chemical components and biological properties.
Figure 4. OPLS loading plot considering chemical components and biological properties.
Antioxidants 11 01712 g004
Figure 5. Biplot summarising the results of the assays and the different extracts.
Figure 5. Biplot summarising the results of the assays and the different extracts.
Antioxidants 11 01712 g005
Table 1. Total bioactive compounds and total antioxidant capacity (by phosphomolybdenum assay) of the tested extracts (n = 3).
Table 1. Total bioactive compounds and total antioxidant capacity (by phosphomolybdenum assay) of the tested extracts (n = 3).
SpeciesSolventsTPC (mg GAE/g)TFC (mg RE/g)PBD (mmol TE/g)
P. heyniaeHexane17.55 ± 0.16 g3.43 ± 0.14 j1.93 ± 0.02 b
EA21.85 ± 0.36 f12.93 ± 0.35 f2.30 ± 0.10 a
MeOH32.13 ± 0.76 c28.75 ± 0.36 b1.51 ± 0.11 de
Water38.77 ± 0.01 b16.19 ± 0.24 d1.39 ± 0.03 ef
P. meliocarpoides var. meliocarpoidesHexane22.63 ± 0.16 f8.19 ± 0.23 h2.52 ± 0.06 a
EA26.15 ± 1.40 e19.40 ± 0.60 c2.49 ± 0.05 a
MeOH40.03 ± 0.68 b44.66 ± 0.68 a1.82 ± 0.11 bc
Water44.28 ± 0.27 a11.00 ± 0.18 g1.46 ± 0.02 def
P. uechtritziiHexane18.70 ± 0.08 g1.72 ± 0.07 k1.79 ± 0.04 bc
EA25.45 ± 0.12 e5.72 ± 0.05 i2.35 ± 0.17 a
MeOH31.20 ± 0.16 c28.22 ± 0.54 b1.67 ± 0.13 cd
Water29.62 ± 0.07 d14.75 ± 0.30 e1.21 ± 0.03 f
Values are reported as mean ± SD of three parallel experiments. EA: ethyl acetate; MeOH: methanol; TPC: total phenolic content; TFC: total flavonoid content; PBD: phosphomolybdenum; GAE: gallic acid equivalent; RE: rutin equivalent; TE: Trolox equivalent. Different letters indicate significant differences between the tested extracts (p < 0.05).
Table 2. Chemical composition of the tested extracts in positive and negative ion mode (mg/g).
Table 2. Chemical composition of the tested extracts in positive and negative ion mode (mg/g).
M-HMolecular FormulaFragmentsName and ReferencesP. heyniae- HexaneP. heyniae- EAP. heyniae- MeOHP. heyniae- WaterP. meliocarpoides- HexaneP. meliocarpoides-EA P. meliocarpoides-MeOHP. meliocarpoides-WaterP. uechtritzii-Hexane P. uechtritzii-EA P. uechtritzii-MeOH P. uechtritzii-Water
Condensed Tannins
609.088050C29H21O15457 305 249Epigallocatechin 3,5-di-gallate [38,39]0.15 ± 0.010.08 ± 0.010.47 ± 0.020.89 ± 0.05ndndndndndndndnd
609.088050C29H21O15457 305 249Epigallocatechin di-gallate isomer [38,39]0.05 ± 0.010.03 ± 0.017.93 ± 0.053.21 ± 0.05ndndndndndndndnd
M + H
763,151C37H31O18611 595 458 443 425 317 305 287Galloylprodelphinidin isomer 1 [40]ndnd2.33 ± 0.051.85 ± 0.04ndndndndndndndnd
761,1398C37H28O18611 595 458 443 425 317 305 287Galloylprodelphinidin isomer 2 [40]0.01 ± 0.010.01 ± 0.010.31 ± 0.030.25 ± 0.02ndndndndndndndnd
763,1505C37H30O18611 595 458 443 425 317 305 287Galloylprodelphinidin isomer 3 [40]0.01 ± 0.01nd4.66 ± 0.055.79 ± 0.07ndndndndndndndnd
761,1398C37H28O18611 595 458 443 425 317 305 287Galloylprodelphinidin isomer 4 [40]ndnd3.22 ± 0.055.47 ± 0.09ndndndndndndndnd
915,162C44H34O22611 595 458 443 425 317 305 287di-O-galloylprodelphinidin dimer isomer 1 [40]0.01 ± 0.01nd3.11 ± 0.052.81 ± 0.060.34 ± 0.020.23 ± 0.020.48 ± 0.010.11 ± 0.01ndnd0.22 ± 0.030.24 ± 0.02
915,1615C44H34O22611 595 458 443 425 317 305 287di-O-galloylprodelphinidin dimer isomer 2 [40]ndndndndndndndndndndndnd
459,0925C23H20O10303,0579Epigallocatechin-gallate * [41]ndndndndndndndndnd5.09 ± 0.0853.52 ± 0.2231.25 ± 0.44
345,0614C17H13O8315Methylellagic acid * [42,43]ndnd1.62 ± 0.051.27 ± 0.050.93 ± 0.041.86 ± 0.042.43 ± 0.042.04 ± 0.02ndndndnd
357.0461C14H12O11 Chebulic acid [44]ndndndnd0.02 ± 0.010.04 ± 0.020.05 ± 0.010.88 ± 0.01ndndndnd
357.046C14H12O11 Chebulic acid isomer 1 [44]ndndndndnd0.02 ± 0.010.01 ± 0.011.71 ± 0.03ndndndnd
761,136C37H30O18593 423(epi)gallocatechin-gallocatechin-gallate dimer [39,45]nd0.56 ± 0.024.81 ± 0.052.63 ± 0.05ndndndndndndndnd
761,136C37H30O18593 423(epi)gallocatechin-gallocatechin-gallate dimer [39,45]nd0.35 ± 0.013.00 ± 0.051.63 ± 0.05ndndndndndndndnd
761,136C37H30O18593 423(epi)gallocatechin-gallocatechin-gallate dimer [39,45]nd3.12 ± 0.0623.54 ± 0.2512.4 ± 0.11ndndndndndndndnd
761,136C37H30O18593 423(epi)gallocatechin-gallocatechin-gallate dimer [39,45]nd2.22 ± 0.0513.64 ± 0.117.52 ± 0.11ndndndndndndndnd
745,141C37H30O17 Epicatechin-(4beta- > 8)-epigallocatechin 3′-gallate [39,45]nd0.59 ± 0.067.31 ± 0.074.48 ± 0.10ndndndndndndndnd
913,1468C44H33O22761 423(epi)gallocatechin-gallate dimer [39,45]nd3.46 ± 0.1118.02 ± 0.1112.39 ± 0.22ndndndndndndndnd
913,1468C44H33O22761 423(epi)gallocatechin-gallate dimer [39,45]nd1.85 ± 0.099.71 ± 0.118.46 ± 0.10ndndndndndndndnd
745,141C37H30O18 Epicatechin-(4beta- > 8)-epigallocatechin 3′-gallate [39,45]nd0.40 ± 0.055.79 ± 0.086.57 ± 0.05ndndndndndndndnd
745,141C37H30O19 Epicatechin-(4beta- > 8)-epigallocatechin 3′-gallate [39,45]nd0.86 ± 0.055.89 ± 0.091.96 ± 0.02ndndndndndndndnd
915,162C44H36O22457Assamicain [46]nd0.17 ± 0.053.52 ± 0.224.58 ± 0.04ndndndndndndndnd
457,0771C22H17O11331 305 169gallocatechin gallate* [46]0.01 ± 0.011.61 ± 0.057.23 ± 0.097.32 ± 0.05ndndndndndndndnd
457,0771C22H17O11331 305 169epi-gallocatechin gallate*nd1.77 ± 0.058.61 ± 0.116.61 ± 0.09ndndndndndndndnd
915,162C44H36O22457Assamicain B [46]nd0.05 ± 0.017.24 ± 0.095.73 ± 0.07ndndndndndndndnd
TOTAL0.2417.12141.95103.771.292.152.974.73nd5.0953.7431.49
M-H Hydrolisable tannins
801,1135C35H29O23 tetragalloylquinic acid [39,47,48,49]ndnd0.99 ± 0.051.51 ± 0.090.26 ± 0.030.21 ± 0.011.33 ± 0.063.02 ± 0.06ndndndnd
618,0935C27H23O17 3-O-Galloyl-4,6-(S)-HHDP-d-glucose [39,47,48,49]ndndndnd0.19 ± 0.020.16 ± 0.0313.35 ± 0.366.11 ± 0.06ndndndnd
787,0994C34H27O22617 3211,3-Digalloyl-4,6-HHDP-glucose/Heterophyliin A [39,47,48,49]ndndndnd1.28 ± 0.040.01 ± 0.0126.06 ± 0.3512.48 ± 0.12ndndndnd
787,1152C35H30O21 2″,3″,6″-Tris-O-(3,4,5-trihydroxybenzoyl)-3′-Glucosyl-2′,4′,6′-trihydroxyacetophenone [39,47,48,49]ndnd1.71 ± 0.061.16 ± 0.060.22 ± 0.030.15 ± 0.0337.97 ± 0.9918.84 ± 0.17ndndndnd
787,0994C34H27O22617 321Heterophylliin A isomer ndnd1.86 ± 0.051.16 ± 0.090.47 ± 0.05nd36.29 ± 0.8522.62 ± 0.23ndndndnd
801,155C35H29O22 tetragalloylquinic acid [39,47,48,49]ndndndnd8.58 ± 0.060.36 ± 0.0318.63 ± 0.8812.13 ± 0.21ndndndnd
1237,71C55H34O34 Prostratin A or Tirucallin Andndndnd0.11 ± 0.020.04 ± 0.020.32 ± 0.012.11 ± 0.06ndndndnd
1083.0581C48H27O30 alfa-Punicalagin * [48]ndndndnd4.26 ± 0.148.82 ± 0.110.11 ± 0.010.65 ± 0.05ndndndnd
1083.0581C48H27O30 beta-punicalagin * [48]ndndndnd9.12 ± 0.1614.64 ± 0.230.13 ± 0.011.13 ± 0.02ndndndnd
1083.0581C48H27O300601Punicalagin derivative [48]ndndndnd4.72 ± 0.228.40 ± 0.220.32 ± 0.014.93 ± 0.09ndndndnd
1083.0581C48H28O30601Terchebuline [39,45,48]ndndndnd8.71 ± 0.0317.53 ± 0.85ndndndndndnd
637.1052C27H24O18483 465 313trigalloyl glucose [39,47,48,49]ndndndnd0.11 ± 0.030.23 ± 0.091.32 ± 0.023.67 ± 0.04ndndndnd
637.1048C27H24O18483 465 313trigalloyl glucose [39,47,48,49]ndndndnd0.04 ± 0.010.07 ± 0.031.43 ± 0.044.48 ± 0.04ndndndnd
637.1048C27H24O18483 465 313trigalloyl glucose [39,47,48,49]ndndndnd0.08 ± 0.010.13 ± 0.020.37 ± 0.0227.63 ± 0.34ndndndnd
971.1733C43H38O26817 635 465tetragalloyl-syrigylglucose [39,47,48,49]ndndndnd0.01 ± 0.010.07 ± 0.033.43 ± 0.084.37 ± 0.06ndndndnd
637.1045C27H24O18 trigalloyl glucose [39,47,48,49]ndndndnd0.46 ± 0.041.35 ± 0.057.63 ± 0.0928.29 ± 0.33ndndndnd
939.1110C41H30O26 Tellimagrandin II [47,48,49,50]ndndndnd0.05 ± 0.020.04 ± 0.014.77 ± 0.080.19 ± 0.01ndndndnd
989.1483C42H36O28 Methyl neochebulinate [44]ndndndnd0.04 ± 0.030.11 ± 0.0310.02 ± 0.332.07 ± 0.06ndndndnd
787,0997C34H27O22 tetragalloyl glucose [39,47,48,49]ndndndnd0.48 ± 0.050.66 ± 0.0311.15 ± 0.3432.53 ± 0.22ndndndnd
955C41H32O27 Chebulinic acid [44,47,48]ndndndnd2.02 ± 0.072.55 ± 0.0913.5 ± 0.0853.83 ± 0.76ndndndnd
1001.2199C45H45O26909 617 465digalloyl-dishikimoyl-synapoylglucose [39,45,47,49]ndndndndndnd0.55 ± 0.01ndndndndnd
1001.2199C45H45O26910 617 465digalloyl-dishikimoyl-synapoylglucose [39,45,47,49]ndndndndndnd1.54 ± 0.05ndndndndnd
1001.2199C45H45O26911 617 465digalloyl-dishikimoyl-synapoylglucose [39,45,47,49]ndndndndndnd0.74 ± 0.01ndndndndnd
1001.2199C45H45O26912 617 465digalloyl-dishikimoyl-synapoylglucose [39,45,47,49]ndndndndndnd0.37 ± 0.01ndndndndnd
987.1887C43H40O26799 771 617 465 313trigalloyl-syrigyl-syìhykimil-glucose [39,45,47,49]ndndndnd0.11 ± 0.02nd12.29 ± 0.0810.84 ± 0.07ndndndnd
1001.2199C45H45O26913 617 465digalloyl-dishikimoyl-synapoylglucose [39,45,47,49]ndndndndndnd0.42 ± 0.03ndndndndnd
TOTALndnd4.563.8241.2955.52204.04 ± 1.52251.9 ± 0.99ndndndnd
M + H Coumarins
177,0552C10H9O31338-methoxy coumarin * [8,51]0.05 ± 0.010.03 ± 0.010.86 ± 0.050.84 ± 0.040.19 ± 0.020.23 ± 0.035.83 ± 0.111.04 ± 0.020.05 ± 0.010.09 ± 0.0114.39 ± 0.1116.12 ± 0.33
327,0896C18H15O6 4-coumaroyl-4′-hydroxyphenyllactate0.02 ± 0.010.01 ± 0.010.51 ± 0.030.87 ± 0.05ndndndnd0.02 ± 0.010.01 ± 0.013.88 ± 0.072.38 ± 0.06
287,1016C16H15O5269 245 201heraclenin/prangenin * [8,51]ndnd2.07 ± 0.114.14 ± 0.09ndndndndndndndnd
247,097C14H14O4188 1468-(2′,3′-dihydroxyisopentyl)-7-hydroxycoumarin /marmesin* [8,51]ndnd0.74 ± 0.053.25 ± 0.090.52 ± 0.03nd2.41 ± 0.047.06 ± 0.04ndnd4.71 ± 0.065.87 ± 0.11
271,091C16H13O4203 175Imperatorin* [8,51]ndndndnd0.25 ± 0.030.21 ± 0.038.28 ± 0.1114.72 ± 0.33ndndndnd
327,166C20H23O4 Dentatin * [8,51]ndndndndndndndnd4.05 ± 0.031.54 ± 0.032.61 ± 0.053.68 ± 0.11
TOTAL0.070.044.189.110.940.4316.5122.824.121.6425.5928.05
M-H Flavonoid derivatives
785,162C36H32O20 Degalloyltheaflavoninndndndnd2.04 ± 0.032.65 ± 0.112.17 ± 0.093.16 ± 0.22ndndndnd
771,2372C34H42O20625,1824 479,0928 317,0729Typhaneosidendndndnd1.37 ± 0.051.05 ± 0.0830.82 ± 0.5515.9 ± 0.78ndndndnd
433,1171C21H21O10 Apigenin-C-hexosidendndndnd1.26 ± 0.090.01 ± 0.010.02 ± 0.010.28 ± 0.02ndndndnd
601,1193C28H25O15431 3292′-O-Galloylquercitrinndndndnd1.25 ± 0.02129.98 ± 0.6614.72 ± 0.22ndndndnd
585,1269C28H25O14431 415 285Kaempferol-deoxyhexoside-gallate [39,45,49]ndndndnd0.12 ± 0.010.32 ± 0.0319.61 ± 0.449.87 ± 0.08ndndndnd
769.2205C34H42O20605 314Isorhamnetin-7-O-hexoside-3-O-hexosidedeoxyhexoside [39,45,49]ndndndndndndndndndnd0.24 ± 0.020.11 ± 0.01
625.1198C30H26O15448 301Quercetin-7-O-hexoside-caffeoyl [39,45,49]ndndndndndndndndndnd3.08 ± 0.091.4 ± 0.02
625.1196C30H26O15449 301Quercetin-7-O-hexoside-caffeoyl isomer [39,45,49]ndndndndndndndndndnd0.94 ± 0.060.25 ± 0.01
479.0815C21H20O13316 271 179Myricetin-3-O-glucopyranoside *ndndndndndndndndndnd4.52 ± 0.090.71 ± 0.02
769.2204C34H42O20605 314Isorhamnetin-7-O-hexoside-3-O-hexosidedeoxyhexoside [39,45,49]ndndndndndndndndndnd1.54 ± 0.110.71 ± 0.03
769.2200C34H42O20605 314Isorhamnetin-7-O-hexoside-3-O-hexosidedeoxyhexoside [39,45,49]ndndndndndndndndndnd0.78 ± 0.080.43 ± 0.01
609.1464C27H30O16301 271 179Rutin *ndndndndndndndndndnd8.55 ± 0.112.75 ± 0.11
463.0884C21H20O12316 271Myricetin-3-O-rhamnopyranoside *ndndndndndndndndndnd11.7 ± 0.223.07 ± 0.11
463.0887C21H20O12316 271Myricetin-7-O-rhamnopyranoside *ndndndndndndndndndnd7.37 ± 0.131.11 ± 0.22
623.1627C28H32O16461 314 315 299Isorhamnetin-7-O-rhamnopyranosyl-3-O-glucopyranoside *ndndndndndndndndndnd5.51 ± 0.221.86 ± 0.22
463.0969C21H20O12301Quercetin-3-O-galactopyranoside *ndndndnd0.06 ± 0.030.16 ± 0.031.05 ± 0.023.69 ± 0.05ndndndnd
477.0684C21H18O13301 271Quercetin-3-O-glucuronide *ndndndndndndndndndnd16.33 ± 0.9910.17 ± 0.22
623.1638C28H32O16315 300Isorhamnetin-3-O-hexosyl-deoxyhexoside [39,45,49]ndndndndndndndndndnd2.18 ± 0.110.73 ± 0.05
623.1622C28H32O16315 300Isorhamnetin-3-O-rutinoside *ndndndndndndndndndnd5.05 ± 0.091.61 ± 0.07
477.1054C22H22O12314 300 271 255Isorhamnetin-3-O-glucopyranoside *ndndndndndndndndndnd2.02 ± 0.051.4 ± 0.07
463.0854C21H20O12301 271 255 179 151Quercetin-3-O-glucopyranoside *ndndndndndndndndndnd34.3 ± 0.5110.57 ± 0.33
477.1043C22H22O12314 300 271 255Isorhamnetin-3-O-galactopyranoside *ndndndndndndndndndnd10.03 ± 0.113.11 ± 0.05
491.0840C22H20O13315 300 271 255Isorhamnetin-3-O-glucuronide *ndndndndndndndndndnd11.93 ± 0.226.18 ± 0.07
431.0978C21H20O10285Kaempferol-3-O-rhamnoside *ndndndndndndndndndnd8.18 ± 0.081.71 ± 0.06
461.1083C22H21O11314 300Isorhamnetin-7-O-rhamnopyranoside *ndndndndndndndndndnd4.12 ± 0.111.06 ± 0.05
TOTALndndndnd6.10 ± 0.055.19 ± 0.0983.65 ± 0.9947.62 ± 0.08nd2.68 ± 0.03138.03 ± 1.2548.92 ± 0.66
M + H Hydroxycinnamic derivatives nd
339,11C16H19O81911-coumaroyl quinic acid [39,45,49]1.83 ± 0.015.48 ± 0.078.52 ± 0.056.32 ± 0.070.13 ± 0.011.44 ± 0.073.06 ± 0.040.75 ± 0.05ndnd4.11 ± 0.043.91 ± 0.04
M-H ndnd
341,0873C15H17O9179Caffeoyl hexose [39,45,49]nd0.16 ± 0.050.76 ± 0.061.31 ± 0.090.01 ± 0.010.02 ± 0.010.01 ± 0.010.02 ± 0.01ndnd0.08 ± 0.010.23 ± 0.02
501,1619C22H29O13417 399 285 152Clemomandshuricoside Bndndndndndndndndndnd2.09 ± 0.060.55 ± 0.02
TOTAL1.835.649.26 ± 0.107.61 ± 0.090.14 ± 0.051.46 ± 0.063.07 ± 0.050.77 ± 0.03ndnd6.27 ± 0.074.69 ± 0.02
M + H Other compounds ndnd
225,1399C13H20O3147 119 103 79 886-hydroxy-3-oxo-alpha-ionol3.72 ± 0.013.12 ± 0.0510.2 ± 0.2010.8 ± 0.11nd0.89 ± 0.035.05 ± 0.110.08 ± 0.01ndnd6.52 ± 0.056.57 ± 0.02
381,1658C16H29O10364 219 200Prenyl arabinosyl-(1- > 6)-glucoside0.11 ± 0.010.27 ± 0.057.36 ± 0.117.92 ± 0.10ndndndndndnd3.25 ± 0.053.61 ± 0.04
787,1152C35H31O31 2″,3″,6″-Tris-O-(3,4,5-trihydroxybenzoyl)-3′-Glucosyl-2′,4′,6′-trihydroxyacetophenonendnd1.71 ± 0.061.16 ± 0.050.21 ± 0.050.15 ± 0.0337.97 ± 0.5418.84 ± 0.22ndndndnd
285,0974C13H17O7 p-hydroxy-benzoic acid rhamnosyl ester4.92 ± 0.023.83 ± 0.063.52 ± 0.093.65 ± 0.09ndndndnd0.57 ± 0.032.54 ± 0.055.46 ± 0.112.91 ± 0.03
315.1088C14H18O8167glucovanillinndnd2.98 ± 0.092.27 ± 0.11ndndndnd2.21 ± 0.096.78 ± 0.0910.27 ± 0.223.67 ± 0.05
TOTAL8.737.2225.7725.80.21.0443.0218.922.789.3225.516.75
nd: not detected; * identity confirmed by injection of reference compound.
Table 3. Antioxidant properties of the tested extracts (n = 3).
Table 3. Antioxidant properties of the tested extracts (n = 3).
SpeciesSolventsDPPH (mg TE/g)ABTS (mg TE/g)CUPRAC (mg TE/g)FRAP (mg TE/g)MCA (mg EDTAE/g)
P. heyniaeHexane5.42 ± 0.37 gna35.97 ± 1.80 h20.27 ± 0.55 i27.34 ± 0.34 a
EA10.15 ± 0.47 e10.44 ± 0.91 g59.74 ± 0.63 ef31.60 ± 0.62 gh26.18 ± 0.16 ab
MeOH46.65 ± 0.34 c60.98 ± 0.04 d108.43 ± 1.52 c70.21 ± 3.22 d15.53 ± 0.90 e
Water50.66 ± 0.42 b90.74 ± 1.35 a129.89 ± 3.24 b93.14 ± 1.75 b27.79 ± 0.32 a
P. meliocarpoides var. meliocarpoidesHexane4.13 ± 0.33 g8.90 ± 0.95 g55.98 ± 0.84 f28.61 ± 1.00 h26.38 ± 0.15 ab
EA9.70 ± 0.09 e17.72 ± 0.08 f62.26 ± 1.02 e34.22 ± 0.55 g19.91 ± 0.22 cd
MeOH52.27 ± 0.28 a77.88 ± 1.07 b133.19 ± 1.09 b80.79 ± 1.03 c18.55 ± 0.61 d
Water52.01 ± 0.52 ab92.84 ± 0.44 a154.04 ± 2.10 a104.34 ± 1.07 a21.17 ± 0.54 c
P. uechtritziiHexane2.18 ± 0.62 h17.00 ± 0.74 f35.91 ± 1.32 h21.79 ± 0.85 i18.50 ± 1.92 d
EA8.11 ± 0.35 f26.29 ± 0.44 e45.62 ± 2.18 g27.99 ± 1.69 h20.75 ± 0.25 c
MeOH34.55 ± 0.63 d75.03 ± 0.43 c96.69 ± 2.56 d56.31 ± 0.36 f17.88 ± 0.83 d
Water34.52 ± 0.96 d74.74 ± 0.58 c95.43 ± 0.80 d65.86 ± 0.82 e24.57 ± 0.40 b
Values are reported as mean ± SD of three parallel experiments. EA: ethyl acetate; MeOH: methanol; TE: Trolox equivalent; EDTAE: EDTA equivalents. na: not active. Different letters in the same column indicate significant differences between the tested extracts (p < 0.05).
Table 4. Enzyme inhibitory effects of the tested extracts (n = 3).
Table 4. Enzyme inhibitory effects of the tested extracts (n = 3).
SpeciesSolventsAChE
(mg GALAE/g)
BChE
(mg GALAE/g)
Tyrosinase
(mg KAE/g)
Amylase
(mmol ACAE/g)
Glucosidase
(mmol ACAE/g)
P. heyniaeHexane2.39 ± 0.06 a7.83 ± 0.18 a56.07 ± 1.46 ef0.36 ± 0.01 c0.67 ± 0.04 bc
EA1.58 ± 0.38 cd7.64 ± 0.15 ab54.21 ± 1.32 f0.41 ± 0.01 b0.62 ± 0.04 cd
MeOH2.36 ± 0.18 a4.28 ± 0.16 c65.20 ± 0.89 c0.17 ± 0.01 e0.46 ± 0.08 e
Water0.35 ± 0.08 ena17.34 ± 0.38 i0.06 ± 0.01 gna
P. meliocarpoides var. meliocarpoidesHexane1.16 ± 0.27 d7.97 ± 0.06 a81.15 ± 0.19 a0.46 ± 0.01 a0.61 ± 0.02 cd
EAna7.32 ± 0.80 ab59.92 ± 0.96 d0.40 ± 0.02 b0.56 ± 0.01 d
MeOHna3.34 ± 0.46 d70.57 ± 0.59 b0.21 ± 0.01 d0.74 ± 0.01 ab
Water0.19 ± 0.01 ena21.23 ± 1.33 h0.05 ± 0.01 gna
P. uechtritziiHexane2.34 ± 0.12 ab7.63 ± 0.39 ab58.77 ± 1.76 de0.39 ± 0.01 b0.64 ± 0.01 cd
EA2.13 ± 0.18 ab6.91 ± 0.17 b61.03 ± 1.10 d0.40 ± 0.01 b0.59 ± 0.01 cd
MeOH1.76 ± 0.14 bc1.58 ± 0.12 e68.03 ± 0.39 bc0.20 ± 0.01 d0.78 ± 0.01 a
Waterna0.34 ± 0.04 f27.54 ± 1.03 g0.09 ± 0.01 fna
Values are reported as mean ± SD of three parallel experiments. EA: Ethyl acetate; MeOH: Methanol; GALAE: Galantamine equivalent; KAE: Kojic acid equivalent; ACAE: Acarbose equivalent; na: not active. Different letters in the same column indicate significant differences between the tested extracts (p < 0.05).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Dall’Acqua, S.; Sut, S.; Zengin, G.; Peron, G.; Elbasan, F.; Yildiztugay, E.; Bibi Sadeer, N.; Mahomoodally, M.F. Phytochemical Screening, Antioxidant, and Enzyme Inhibitory Properties of Three Prangos Species (P. heyniae, P. meliocarpoides var. meliocarpoides, and P. uechtritzii) Depicted by Comprehensive LC-MS and Multivariate Data Analysis. Antioxidants 2022, 11, 1712. https://doi.org/10.3390/antiox11091712

AMA Style

Dall’Acqua S, Sut S, Zengin G, Peron G, Elbasan F, Yildiztugay E, Bibi Sadeer N, Mahomoodally MF. Phytochemical Screening, Antioxidant, and Enzyme Inhibitory Properties of Three Prangos Species (P. heyniae, P. meliocarpoides var. meliocarpoides, and P. uechtritzii) Depicted by Comprehensive LC-MS and Multivariate Data Analysis. Antioxidants. 2022; 11(9):1712. https://doi.org/10.3390/antiox11091712

Chicago/Turabian Style

Dall’Acqua, Stefano, Stefania Sut, Gokhan Zengin, Gregorio Peron, Fevzi Elbasan, Evren Yildiztugay, Nabeelah Bibi Sadeer, and Mohamad Fawzi Mahomoodally. 2022. "Phytochemical Screening, Antioxidant, and Enzyme Inhibitory Properties of Three Prangos Species (P. heyniae, P. meliocarpoides var. meliocarpoides, and P. uechtritzii) Depicted by Comprehensive LC-MS and Multivariate Data Analysis" Antioxidants 11, no. 9: 1712. https://doi.org/10.3390/antiox11091712

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