Next Article in Journal
Tyk2 Targeting in Immune-Mediated Inflammatory Diseases
Next Article in Special Issue
Extraction, Structural, and Antioxidant Properties of Oligosaccharides Hydrolyzed from Panax notoginseng by Ultrasonic-Assisted Fenton Degradation
Previous Article in Journal
Biomaterials-Enhanced Intranasal Delivery of Drugs as a Direct Route for Brain Targeting
Previous Article in Special Issue
Unveiling the Potentiality of Shikonin Derivatives Inhibiting SARS-CoV-2 Main Protease by Molecular Dynamic Simulation Studies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Antioxidant, Tyrosinase, α-Glucosidase, and Elastase Enzyme Inhibition Activities of Optimized Unripe Ajwa Date Pulp (Phoenix dactylifera) Extracts by Response Surface Methodology

1
Department of Food Science and Biotechnology, Graduate School, Kyungpook National University, Daegu 41566, Republic of Korea
2
Food and Bio-Industry Research Institute, Inner Beauty/Antiaging Center, Kyungpook National University, Daegu 41566, Republic of Korea
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(4), 3396; https://doi.org/10.3390/ijms24043396
Submission received: 20 November 2022 / Revised: 2 February 2023 / Accepted: 6 February 2023 / Published: 8 February 2023

Abstract

:
The Ajwa date (Phoenix dactylifera L., Arecaceae family) is a popular edible fruit consumed all over the world. The profiling of the polyphenolic compounds of optimized unripe Ajwa date pulp (URADP) extracts is scarce. The aim of this study was to extract polyphenols from URADP as effectively as possible by using response surface methodology (RSM). A central composite design (CCD) was used to optimize the extraction conditions with respect to ethanol concentration, extraction time, and temperature and to achieve the maximum amount of polyphenolic compounds. High-resolution mass spectrometry was used to identify the URADP’s polyphenolic compounds. The DPPH-, ABTS-radical scavenging, α-glucosidase, elastase and tyrosinase enzyme inhibition of optimized extracts of URADP was also evaluated. According to RSM, the highest amounts of TPC (24.25 ± 1.02 mgGAE/g) and TFC (23.98 ± 0.65 mgCAE/g) were obtained at 52% ethanol, 81 min time, and 63 °C. Seventy (70) secondary metabolites, including phenolic, flavonoids, fatty acids, and sugar, were discovered using high-resolution mass spectrometry. In addition, twelve (12) new phytoconstituents were identified for the first time in this plant. Optimized URADP extract showed inhibition of DPPH-radical (IC50 = 87.56 mg/mL), ABTS-radical (IC50 = 172.36 mg/mL), α-glucosidase (IC50 = 221.59 mg/mL), elastase (IC50 = 372.25 mg/mL) and tyrosinase (IC50 = 59.53 mg/mL) enzymes. The results revealed a significant amount of phytoconstituents, making it an excellent contender for the pharmaceutical and food industries.

1. Introduction

Antioxidative phenolics found in the tissues of many plant species are thought to be responsible for their medicinal actions. They play a variety of purposes in plants, from structural to defensive [1]. However, studies have demonstrated phenolics’ preventive significance in diabetes, chronic cardiovascular illnesses, cancer, and aging cases [2,3]. Their positive effects on human health have thus far undergone substantial study. The study of polyphenolic compounds is gaining popularity, and the first and most crucial stage in extracting and purifying polyphenolic compounds from plant sources is extraction [4], given that the extraction of polyphenol is influenced by several factors, including the chemical makeup of the sample, the solvent employed, agitation, extraction time, solute/solvent ratio, and temperature [5,6]. Furthermore, phenolic molecules should not be oxidized because they participate in the enzymatic browning reaction and lose their phenol activity and antioxidant capacity [7]. Additionally, phenolic compounds’ structural and physicochemical diversity precludes a uniform extraction methodology and necessitates a unique strategy for each phenolic source [7]. Therefore, it is essential to research extraction conditions to enhance polyphenolic compound yield.
Tyrosinase is the type-3 metalloenzyme most closely related to the formation of melanin [8]. Living organisms naturally produce melanin to protect the skin from UV rays and reactive oxygen species (ROS). Wrinkles and skin hyperpigmentation brought on by too much melanin are urgent problems in the cosmetics industry [9]. Tyrosinase activity modulation has been the main focus of control measures for melanin formation. Because of their structural similarities to the enzyme’s substrate, L-tyrosine, polyphenolic compounds are the source of most tyrosinase inhibitors [9,10]. Furthermore, α-glucosidase is one of the essential enzymes for diabetes mellitus (DM). α-glucosidase hydrolyzes the 1,4-glucosidic bonds of oligosaccharides to create monosaccharides, which are absorbed into the blood from the intestine [11]. As a result, inhibitors of α-glucosidase can significantly lower postprandial hyperglycemia following a mixed-carbohydrate diet and may be used to manage DM. Furthermore, human neutrophil elastase (HNE) is a serine protease with a single polypeptide chain that is stored and secreted by polymorphonuclear neutrophils. It is a member of the elastase-like serine proteases subfamily [12]. Excess extracellular HNE, which can break down structural proteins of the extracellular matrix such as elastin, proteoglycan, collagen, and fibronectin, is brought on by imbalances between NE and its endogenous inhibitors [13]. NE can destroy elements of the coagulation and fibrinolytic pathways, as well as activate matrix metalloproteinases and deactivate their inhibitors. Following this, an excess of HNE may cause a number of pathological illnesses and tissue damage, including rheumatoid arthritis, psoriasis, cystic fibrosis, chronic obstructive lung disease, acute respiratory distress syndrome, pulmonary fibrosis, and pulmonary fibrosis [14,15,16,17]. The ability of the serine protease inhibitors to control the proteolytic activities of the serine proteases makes them vital for restoring the balance between the protease and anti-protease systems, limiting excessive elastin proteolysis, and lowering neutrophil accumulation at inflammatory areas [13,15]. Natural compounds such as polyphenolic compounds (ugonins Q: IC50 = 0.49 μM, quercetin-3-O-glucoside; IC50 = 0.35 μM, 6,8-diprenylorobol; IC50 = 1.3 μM, and amentoflavone; IC50 = 1.27 μM) are primarily found in herbal plants and have been shown to affect elastase release [12,14].
The extraction of phenolic chemicals must be optimized to produce a reliable result. It is generally possible to optimize a process using either empirical or statistical methods. The empirical one-factor-at-a-time technique includes altering one component at a time while keeping the other variables constant [18]. This approach’s fundamental flaw is that it ignores how the variables interact, making it impossible to account for all of a parameter’s impacts on the response. Another burden is that it takes many trials to complete the investigation, which extends the time, expense, reagent, and material consumption [18]. To overcome this challenge, multivariate statistical methods were used to optimize the analytical processes. The response surface methodology (RSM) is one of the most well-known multivariate approaches used in analytical optimization. Intending to optimize the desired response, RSM is a set of statistical and mathematical approaches for creating, developing, and modifying procedures where several variables have an impact. In addition to improving the design of existing products, it can be used to develop, formulate, and build new ones. It explains how the independent variables might affect the processes individually or collectively. In addition to evaluating the effects of independent components, this experimental approach offers a mathematical model that illustrates the chemical or biological processes [18,19].
Ajwa dates (Phoenix dactylifera L., Arecaceae family) are only cultivated in Madinah, Saudi Arabia, and are a popular edible fruit consumed worldwide. It is one of the market’s most expensive and valued cultivars owing to ethnomedical beliefs regarding its health-promoting qualities [20]. It is regarded to have cardioprotective [21], hepatoprotective [22], nephroprotective [23] and constipation-relieving [24] properties and antioxidant, anti-inflammatory, anticancer [25], antifungal, antibacterial, and antiviral activities [26]. In addition, it contains abundant bioactive components such as polyphenols, including phenolic acids, flavonoids, and lignans [20].
To the best of our knowledge, this is the first report that uses RSM to improve the extraction conditions so that more polyphenolic components may be extracted from the pulp of unripe Ajwa dates (URADP). The goal was to obtain the highest polyphenolic content possible from URADP by investigating and optimizing extraction parameters such as extraction temperature and duration, as well as ethanol concentration, using the RSM central composite design (CCD) tool. The RSM-CCD approach’s projected values accurately reflect the actual findings, and this statistical technique can be used to maximize the extraction of URADP polyphenolic compounds.

2. Results and Discussion

Response surface methodology (RSM) is a collection of mathematical and statistical methods built on fitting polynomial equations to experimental data. It accurately describes the behavior of data collection designed to produce statistical predictions. It is better than traditional single-parameter optimization since it takes less time, space, and raw materials [18,19].
Scientific information dealing with optimization of the extraction of polyphenols from unripe Ajwa date pulp (URADP) extracts is very inadequate. Mounting evidence has revealed the optimization of ultrasonic assistance extraction, microwave-assisted extraction, and supercritical fluid extraction procedures that were performed to extract polyphenols from different varieties of dates except from Ajwa dates [4,27,28,29]. To the best of our knowledge, this is the first report dealing with the optimization of heat extraction on individual biologically active polyphenols as dependent variables.

2.1. Fitting of the RSM Models

Table 1 lists the experimental conditions and findings for each extraction scenario. All response variables were transformed into second-order quadratic polynomials to account for extraction factor effects. The statistical significance of the fitted second-order quadratic model equations was assessed using ANOVA. The fitness of the model was evaluated using the regression coefficient (β), adjusted correlation factor (R2), coefficient of variation (CV), and adequate precision (Table 2). The non-significant terms (p > 0.05) were removed to enhance the models’ fit and predictions. p values were used to assess each coefficient’s significance. The model terms were statistically significant, extremely significant, and impressively significant when the p values were less than 0.05, 0.01, and 0.001, respectively.
From Table 2, smaller probability values (p < 0.0001) indicate that the model terms are significant. In general, proceeding with exploration and optimization of a fitted response surface may produce poor or misleading results unless the model exhibits an adequate fit [7]. The developed regression models have a high degree of statistical significance, as indicated by their R2 values (0.9706 and 0.9968). The appropriate precision value is an indicator of the signal-to-noise ratio. It is preferable to have a ratio of >4 [25]. Here, the ratios were 15.9930 and 49.6969, suggesting a sufficient signal, indicating that the model is suitable for this procedure. The coefficient of variation (CV) is a measure of a model’s reproducibility and describes the extent to which the data were dispersed. The CV for total phenolic content (TPC) and total flavonoid content (TFC) of URADP was within the acceptable range (Table 2). Since CV is a measure expressing standard deviation as a percentage of the mean, the small values of CV give better reproducibility. In general, a high CV indicates that variation in the mean value is high and does not satisfactorily develop an adequate response mode [7]. The modified R2 (R2 ≥ 0.80) was well within acceptable limits in this study, showing that the experimental data fit second-order polynomial equations satisfactorily. To demonstrate the interactions between the independent variables, 3D surfaces and contour plots were constructed using multiple linear regression equations. The main and cross-product effects of the independent variables on the response variables are more easily understood from these 3D charts (Figure 1A,B).

2.2. Effect of Extraction Parameters on TPC and TFC

Phenolic chemicals are secondary metabolites that plants produce under oxidative stress and are necessary to adapt to various adverse situations [1]. In the current investigation, TPC was measured using the Folin–Ciocalteu reagent, and it was discovered that the TPC ranged from 5.41 to 23.92 mgGAE/g (Table 1). According to earlier research, the total phenol content of Ajwa fruit ranged from 2.45 to 4.55 mgGAE/g. In contrast, this study found that URADP had a more significant percentage of total phenolic compounds [30,31]. Numerous studies have shown that the extraction solvent is crucial in the extraction of phenolic compounds. Compared to alcoholic extracts, the contents in hydroalcoholic extracts are always higher [32]. In addition, Eid et al. [33] stated that the phenolic content in Ajwa dates is also varied according to the ripening stage. Unripe Ajwa dates contain higher amounts of phenolic content than ripe fruits. Our experimental results also support this statement. In addition, flavonoids are the most abundant polyphenolic compounds found in Ajwa dates with pervasive dispersal. These polyphenolic compounds are mainly present within fruit skins in high concentrations with immense health benefits such as antioxidant and free radical scavenging activities [31,33]. In URADP extracts, TFC ranged from 6.81 to 24.20 mgCAE/g, which also agrees with the previous work [34].
As shown in Table 2, the linear effects of ethanol concentration (X1), extraction temperature (X3), quadratic component of (X12), (X22), and (X32) and interaction of (X1X2), (X1X3) and (X2X3) exhibited significant effects on both TPC and TFC, except for the interaction of (X2X3), which has no significant effect on TPC. In addition, the regression coefficient (β) values verified the effect of extraction parameter on both TPC and TFC in the following order: TPC: X12 > X22 > X3 > X32 > X1X3 > X1X2 > X1 and TFC: X22 > X12 > X32 > X3 > X2X3 > X1X2 ≅ X1X3 > X1 (Table 2). The following second-order polynomial equations shown in Equations (1) and (2) demonstrate the relationships among TPC, TFC and their variables.
T P C ( Y 1 ) = 23.39 0.3838 X 1 + 0.0612 X 2 + 3.06 X 3 4.37 X 1 2 4.10 X 2 2 1.98 X 3 2 0.5475 X 1 X 2 1.61 X 1 X 3 + 0.0825 X 2 X 3
T F C ( Y 2 ) = 23.10 + 0.9656 X 1 0.5854 X 2 + 1.71 X 3 3.78 X 1 2 4.29 X 2 2 2.81 X 3 2 1.22 X 1 X 2 1.22 X 1 X 3 + 1.57 X 2 X 3
Three-dimensional response surface plots (Figure 1A,B) were constructed based on Equations (1) and (2), respectively, and were applied to clarify the interactive effects of the three variables on the TPC and TFC of URADP, respectively. The ethanol concentration (X1), extraction time (X2) and extraction temperature (X3) showed an interactive effect on both TPC and TFC, which increased readily with increasing ethanol concentration up to 60%, extraction time up to 90 min and extraction temperature up to 65 °C, followed by a decrease (Figure 1A,B). This could be because a medium concentration of ethanol may make the solvent more polar and dissolve more polyphenols, both polar and moderately polar ones [4]. Experiments in a previous comparative study revealed that the extraction of polyphenols from green tea leaves using a high hydrostatic pressure procedure augmented with the percentage of ethanol in the solvent; peaked at 50% ethanol and dropped after that [35]. Hence, the extraction of polyphenols in hydroalcoholic solution is highly efficient, as the polyphenols are highly soluble in these solutions. Furthermore, when ethanol is present at a moderate quantity in water, it can disrupt and break the architecture and structure of phospholipids that make up the lipid bilayer of membranes, affecting the penetrability of plant cells and thereby allowing for better extraction and diffusion of the polyphenolic compounds [36].

2.3. Model Validation

The parameters were forecasted using Derringer’s desirability function, allowing for the multivariate analysis to discover the ideal level for all responses in a single extraction [37]. Figure 2 shows the contour plot as a function of ethanol concentration, extraction time and temperature. In this study, the following conditions, (X1, 52%), (X2: 81 min), and (X3, 63 °C), were used to achieve the maximal overall desirability D = 0.977. Under these optimal conditions, the predicted values for TPC and TFC are 23.98 mgGAE/g and 23.39 mgCAE/g, respectively. To verify the sufficiency of the model equations, a triplicate experiment was conducted in the optimal conditions predicted by Derringer’s desire model and it found the TPC and TFC values to be 24.20 ± 0.096 mgGAE/g and 22.92 ± 1.19 mgCAE/g, respectively. As stated in Table 3, the relative standard deviations (RSDs) of TPC and TFC showed that the predicted values for all groups were very similar to the experimental results. This result is also supported by a prior report [38]. The suitability of the response surface methodology model for quantitative predictions was verified by a satisfactory agreement between the predicted and measured values.

2.4. Comparison of Optimized Extraction Condition with Other Extraction Methods Using Different Solvents

To demonstrate the effectiveness of the optimized method in extracting TPC and TFC, a comparative study was performed. As shown in Figure 3A, higher yields of TPC and TFC were obtained using hydroalcoholic solvent in heat extraction instead of methanol, ethanol and water for heat and maceration extraction. The extraction efficiency of TPC and TFC of different solvents and conditions are presented as heat extract with optimized condition (OP) > heat extract with 100% H2O (HW) > heat extract with 100% methanol (HM) > maceration extract with 100% methanol (MM) > heat extract with 100% ethanol (HE) > maceration extract with 100% H2O (MW) > maceration extract with 100% ethanol (ME) and OP > HM > HW > MM > HE > MW > ME, respectively. This result indicated that hydroalcoholic solvent with heat extraction was more efficient than that of other solvents with both heat and maceration techniques. The results also coincided with those obtained for the extraction of TPC and TFC from dates [30,31,32].
In addition, the pharmacological properties, such as antioxidant, tyrosinase, α-glucosidase, and elastase enzyme inhibitory activities, of various URADP extracts were intensively examined to determine their potential for application. Antioxidant components often have a potent ability to scavenge free radicals, preserving DNA and proteins from damage. Therefore, antioxidant chemicals have been utilized to treat a variety of diseases. DPPH and ABTS•+ has been frequently used as a representative reagent for examining the free radical scavenging activities of bioactive compounds. To quantify the antioxidant activities of different extracts/compounds, the concentration of the samples required to scavenge 50% of radicals (IC50) was measured. A smaller IC50 value indicates an increase in free-radical scavenging ability [38].
As anticipated, OP showed the lowest IC50 values (87.56 ± 1.21 mg/mL) for DPPH-, whereas HM had the lowest IC50 values (105.56 ± 0.98 mg/mL) for ABTS-radical scavenging activity. In addition, OP had the lowest IC50 values of 59.53 ± 1.02 mg/mL and 221.59 ± 2.52 mg/mL for tyrosinase and α-glucosidase enzyme inhibition, respectively. In contrast, the IC50 values (299.05 ± 2.52 mg/mL) for elastase enzyme inhibition were achieved by HW. To calculate the correlation between phenols, flavonoids, antioxidant and enzymes inhibition activity of different enriched products, the Pearson coefficient (ρ) method (supplementary data Table S2) was assessed. A negative ρ value (−1) represents the perfect positive correlation between polyphenols, free radical scavenging and enzyme inhibition ability using IC50. The results revealed very strong correlations for DPPH-radical scavenging and tyrosinase inhibition activity (p < 0.01) with TFC and (p < 0.05) for TPC. In contrast, there was no strong correlation shown between polyphenolic content with ABTS–radical scavenging, α–glucosidase and elastase enzyme inhibition activity. These data are in accordance with other studies that show that higher phenol content augments the antioxidant activity [39,40].

2.5. Chemometric Analysis

Chemometric analysis is the process of better understanding chemical information using mathematical and statistical methods. It is also the process of correlating quality characteristics to analytical instrument data. It has been used to investigate the relationship between antioxidant components and the antioxidant potentiality of various plant extracts [41]. This study used two chemometric techniques—principal component analysis (PCA) and hierarchical cluster analysis (HCA)—to find how the extraction method affected TPC, TFC, antioxidant effects, and other enzyme-inhibitory activities of URADP. PCA analysis reduces the dimensions of the data set and analyzes the responses based on the correlation between data samples. PCA could also find the variable that makes the most difference in the data set [41]. The loading plots were used to determine correlations between the study’s variables. The antioxidant activity, TPC, TFC, and other enzyme inhibitory activities were all included in these loading plots (Figure 3C). A total of 64.6% of the data set’s variability was accounted for by the first principal component (PC1), which also had the highest eigenvalue of 4.52. Meanwhile, 20.6% of the variability was represented by the PC2, which had an eigenvalue of 1.44. According to Figure 3C, the TPC and TFC, which point in the opposite direction from the IC50 loading vectors, may have the most significant potential to contribute to DPPH–, ABTS–radical scavenging, and tyrosinase inhibitory capacities. According to Pearson’s correlation analysis, the TPC and TFC were strongly linked with the antioxidant and tyrosine kinase inhibitory actions, supporting the PCA result (supplementary data Table S2). However, neither TPC nor TFC substantially impacted the activities of elastase and α–glucosidase. Additionally, all variables resulting from comparing the first two PCs (Figure 3C) revealed the existence of three different extract sample groups. Due to their high bioactive component concentration, antioxidant, and tyrosinase inhibitory activity, OP and HM made up Cluster I. In contrast, HE, HW, MM, and ME were made up of Cluster II since they had a mixed record regarding bioactive chemicals, antioxidant activity, and enzyme inhibition. Due to its inferior performance in TPC, TFC, antioxidant, and enzyme inhibition potentiality, extract MW made up Cluster III. Based on similarities, HCA was used to classify distinct solvent-based extraction techniques under research (Figure 3D).

2.6. Secondary Metabolites Profiling in URADP by High-Resolution Mass Spectrometry

Secondary metabolites in the URADP extracts were identified using ESI-MS/MS in the negative ionization modes. As indicated in Table 4, seventy (70) compounds were identified in the negative mode using MSn data from the mass of the precursor ion, fragments, recognized fragmentation patterns for the given classes of compounds, and neutral mass loss, and from comparisons with the existing literature and searches in online databases. Furthermore, the significance of these results was determined by finding the confidence level. Level 3 denotes a tentative candidate, whereas level 2 indicates the probable structure of the identified compound [42].

2.6.1. Phenolic Acids

A phenolic acid may lose its methyl (15 Da), hydroxyl (18 Da), or carboxyl (44 Da) moiety to form a specific fragment ion [42,43]. The fragmentation of a phenolic acid glycoside begins with the cleavage of the glycosidic link to yield the m/z of the phenolic acid and the corresponding loss of the sugar molecule (neutral mass loss of 162 Da). Thus, compounds 1–8 were tentatively identified as hydroxybenzoylhexose, coumaroylshikimic acid, vanillic acid glucoside, caffeoylshikimic acid, quinic acid hexoside, 5-feruloylquinic acid, caffeic acid derivatives, sinapic acid hexoside, caffeoyl shikimic acid hexoside, caffeoyl shikimic acid hexoside, and quinic acid derivatives, respectively [44,45]. Previous studies stated that p-coumaric acid, gallic acid and ferulic acid derivatives were the most dominant phenolic compounds in Ajwa dates [33]. In addition, compound 9 was tentatively identified as 1,2-di-(syringoyl)-hexoside with molecular formula (C24H28O14), which yielded a deprotonated ion [M–H] at m/z 539.1377 and generated the following fragment ions: m/z at 359.09 ([M–H–syringoyl moiety]), 341.08 ([M–H–syringoyl moiety–H2O]), 197.04 (syringic acid), and 153.05 because of the loss of a water molecule from ion m/z 197.04 (Figure 4A). Compound 9 has been identified for the first time in URADP.

2.6.2. Flavonoids

Numerous studies demonstrated that each subgroup of flavonoids exhibits a different fragmentation behavior in MS2 analysis. The cleavage of the C-ring bonds (retro-Diels-Alder, i.e., RDA mechanism) produces ions with the A– or B–ring and some part of the C–ring, which is the most common fragmentation of flavonoids, and notable losses of small neutral molecules, such as CO (28 Da), C2H2O (42 Da), COO (44 Da), and 2CO (56 Da). [5,42,43]. Based on a comparison of the fragmentation patterns with those previously published in the literature, compounds 10-15 were identified as luteolin, catechin or epicatechin, chrysoeriol, quercetin, epigallocatechin, and methoxysinensetin, respectively [5,42,43,44,45]. Flavonoids are frequently glycosylated. The glycoside residues can be linked to the O and C atoms of the flavonoids, resulting in O-glycosides, C-glycosides, and O-C-glycosides. The typical fragmentation of O-glycosides produces neutral species corresponding to sugar units (hexoses, 162 Da; deoxyhexoses, 146 Da; pentoses, 132 Da) and an aglycone ion. Conversely, C-glucosides produce a sequence of fragments because of the cleavage of the C–C bonds with the sugar moiety; examples of such fragments are [M–H–60], [M–H–90], and [M–H–120], which serve as the hallmark diagnostic ions of glycone. Furthermore, in the case of O-C mixed glycosides, the cleavage of the O-glycosidic link is frequently observed in the first step [46,47]. Compounds 17, 19, 21–25, 27–30, and 32–40 were identified as naringenin rhamnoside, biochanin A 7-glucoside, afrormosin 7-glucoside, chrysoeriol hexoside, isoquercitrin, epicatechin 4’-glucuronide, isorhamnetin hexoside, luteolin hexosyl sulfate, chrysoeriol hexosyl sulfate, isoquercitrin sulfate, procyanidin B2, luteolin rhamnosyl hexoside, chrysoeriol rhamnosyl hexoside, isorhamnetin rhamnosyl hexoside, isorhamnetin diglucoside quercetin 3-O-rhamnoside 7-O-glucoside, isorhamnetin 3-O-rhamnosyl glucoside, quercetin xylosyl rutinoside, luteolin rhamnosyl dihexoside, quercetin glucosyl-rutinoside, Isorhamnetin rhamnosyl dihexoside and epicatechin-(2α→7,4α→8)-epicatechin glucoside, respectively, based on the similarities observed in the comparison of their fragmentation behaviors and with the behaviors reported in the literature [5,42,43]. The deprotonated molecular ion [M–H] at m/z 515.1611 exhibited MS2 fragment ions at m/z 353.10 by loss of glucosyl (162 Da). The ion at m/z 353.10 further yielded the MS3 ion at m/z 311 and 297.04 through the loss of 42 and 56 Da. Thus, compound 26 was tentatively identified as luteone glucoside, which has been identified for the first time in URADP (Figure 4B). Moreover, the monoisotopic mass [M–H] at m/z 581.2236 yielded a characteristic fragment ion at m/z 419.17 by loss of hexosyl moiety (162 Da), m/z at 265.10 and 247.09 by cleavage between the α- and β-position, followed by loss of H2O confirming the presence of lyoniresinol. It has been also identified for the first time in URADP (Figure 4C). Mounting evidence revealed that the Ajwa date fruit is enriched with active flavonoids and flavonoid glycosides (mainly as O-glycosides), which depend on the different ripening stages, and where significant quantities of quercetin, naringenin, apigenin, luteolin and kaempferol were found using LC-MS/MS techniques [30,31,32,44,45]. Furthermore, hydrolyzable tannins (HTs) are a broad category of polyphenolic compounds found in plants. During mass spectroscopy fragmentation, HTs frequently exhibit neutral losses of galloyl (152 Da). Compounds 18 and 20 have been characterized as epicatechin-3-gallate and epicatechin-3-(3-methylgallate), respectively, based on the MS and MS2 data and previously cited literature and were first identified in URADP [44].

2.6.3. Sugar Molecules

Further, compound 49 was tentatively identified as xylosmaloside with molecular formula C18H20O9, and this compound generated the deprotonated ion [M–H] at m/z 379.1027 and the following mass fragmentation pattern: m/z 343.08 ([M–H–36 Da]), 217.05 ([M–H–162 Da]), 179.05 (xylose) and 161.04 ([M–H–179.05–18 Da]) (Figure 4D). This compound was also identified for the first time in RADP. Compounds 41–48 were confirmed as sugar molecules from comparison of their deprotonated ion mass and fragmentation behaviors with those reported in the literature and online databases [42,43,48,49,50].

2.6.4. Carboxylic Acids and Fatty Acids

From comparisons of the mass and the fragmentation behaviors of the precursor ion based on mass spectroscopic analysis reported in the literature and various online databases [42,43,48,49,50], compounds 50–70 were identified as carboxylic acids and fatty acids (Table 4).

3. Materials and Methods

3.1. Sample Collection and Preparation

A scientific officer at the National Herbarium and Genebank of Saudi Arabia recognized unripe Ajwa date fruits (voucher specimen No. NHG005) obtained from an Ajwa date farm in Al-Madina Al-Munawara, Saudi Arabia, and they were kept in our lab for additional research. Unripe Ajwa date pulp (URADP) was separated, dried outside, chopped into small pieces, and ground in a sterilized laboratory blender (model 7011HS, Osaka Co. Ltd., Kita-Ku, Osaka, Japan). The powdered samples were maintained in an airtight container covered in aluminum foil and chilled before extraction.

3.2. Extraction Methods

Two distinct techniques and three different solvents (ethanol, methanol, and distilled water) were used for solvent extractions. The maceration method was primarily chosen because it is straightforward and inexpensive. In contrast, heat extraction was carried out in anticipation of a shorter extraction time since temperature may aid in breaking the plant cell wall of an empty palm fruit during heat extraction.
As stated by Mollica et al. [51], the maceration process was carried out with continuous stirring. Briefly, the plant materials (10 g) were soaked in 200 mL of the solvents, and extractions were performed with stirring at 250 rpm for 24 h at room temperature. Choi et al. [5] stated that 10 g of the extract and 200 mL of the solvents were used for heat extraction, which was carried out at 60 °C for 1 h. Following the extraction process, each extract was filtered using Whatman no. 1 filter paper (Schleicher & Schuell, Keene, NH, USA). The solvents were then removed using a rotary evaporator (Tokyo Rikakikai Co. Ltd., Tokyo, Japan) at 50 °C and 50 rpm. Finally, the extracts were lyophilized using a freeze dryer (Il-shin Biobase, Goyang, Republic of Korea). Before further research, the URADP extract was kept at −20 °C.

3.3. Total Phenolic Content (TPC) and Total Flavonoid Content (TFC)

The total phenolic content (TPC) and total flavonoid content (TFC) in URADP extracts were determined by the Folin–Ciocalteu test and the aluminum chloride colorimetric method, respectively [39]. The TPC (y = 0.0512x + 0.0018; r2 = 0.9835) and TFC (y = 0.014x + 0.0021; r2 = 0.9994) were determined using the corresponding regression equations for the calibration curves. The TPC was expressed in terms of the gallic acid equivalent (mg)/dry weight sample (g) and the TFC in terms of the catechin equivalent (mg)/dry weight sample (g).

3.4. Antioxidant Assay and Enzyme Inhibitory Effects

The antioxidant and enzyme inhibitory capability of various URADP extracts was evaluated using the procedures outlined in earlier publications [8,39,52,53]. Antioxidant experiments employed ascorbic acid as a positive control. In contrast, specific enzyme inhibitors, including arbutin, acarbose, and epigallocatechin gallate (EGCG), were utilized for the mushroom tyrosinase, α-glucosidase, and elastase enzyme assays, respectively. The percentage inhibition of DPPH- and ABTS-scavenging, mushroom tyrosinase, α-glucosidase, and elastase activity was calculated using Equation (3).
( %   i n h i b i t i o n ) = [ ( 1 A b s s a m p l e A b s c o n t r o l ) ] ×   100
where Abscontrol and Abssample are the absorbance of the control and absorbance of the sample, respectively. Each sample was examined three times. Each sample’s 50% inhibitory concentration (IC50) value was also computed to compare various extraction method efficacies.

3.5. Experimental Design of RSM for the Extraction Process

The hot extraction method was used to optimize the extraction procedure of polyphenolic compounds from URADP. The RSM model was designed to extract phenolic chemicals from URADP using ethanol concentration (X1), extraction duration (X2), and temperature (X3) as independent process factors. Respondent factors included TPC and TFC (Y1 and Y2, respectively). A three-component, five-layer CCD was employed for the extractions (supplementary data Table S1). The second-order polynomial model equation (Equation (4)) describes the link between independent factors and replies.
Y = β 0 + i = 1 n β i X i + i = 1 n β i i X i i 2 + i n 1 j n β i j X i j
where Y is the response variable and Xi and Xj are the independent coded variables; β0 denotes the constant coefficient, and βi, βii, and βij denote the coefficients of linear, quadratic, and interaction effects, respectively. Design Expert 11 was used for the RSM analysis and multiple linear regression (Stat-Ease, Minneapolis, Minnesota, USA). The model’s adequacy was tested using the determination coefficient (R2), the adjusted determination coefficient (Adj.R2), and the lack of fit test. The F value with p < 0.05 indicated statistical significance. The interaction outcome of each factor on the response value was represented in three-dimensional (3D) surface plots.

3.6. Optimal Extraction Condition and Validation of the Model

Derringer’s desire function was used to find the ideal conditions for maximizing all replies in a single experiment. Each response is turned into a unique desirability function ranging from 0 to 1 during this procedure. The component functions are then combined to create a total desirability function. The total desirability function is constructed using the following equation [4].
D = ( d 1 w 1 d 2 w 2 . d n w n ) 1 / w i
Response surface and desirability function analyses were used to determine the optimal extraction parameters. A triple experiment was carried out under ideal conditions, and the average experimental results were compared to the predicted results to verify the validity of the existing model. In addition, the experimental data were contrasted with the values that the model anticipated. Equation (5) was used to determine the relative standard deviation (RSD) and to compare the experimental and projected results.
R S D   ( % ) = S t a n d a r d   d e v i a t i o n   b e t w e e n   p r e d i c t e d   a n d   e x p e r i m e n t a l   v a l u e s M e a n   v a l u e s   b e t w e e n   p r e d i c t e d   a n d   e x p e r i m e n t a l   v a l u e s ×   100
The resulting data were analyzed and optimized for all response circumstances when the RSD% was <10. Additionally, the electrospray ionization mass spectrometry (ESI-MS)/MS profiles of phenolic compounds were found under ideal circumstances.

3.7. Analysis of Chemical Compounds by ESI-MS/MS

The Q-Exactive Orbitrap mass spectrometer (Thermo Fisher Scientific INC., San Jose, CA, USA) was used to conduct the negative (−) mode ESI-MS investigations. A 500 μL graded syringe (Hamilton Company Inc., Reno, NV, USA) and a 15 μL/min syringe pump (Model 11, Harvard, Holliston, MA, USA) were used to immerse the sample in the ESI source. The normal negative mode ESI-MS conditions were as follows: mass resolution of 140,000 (full width at half maximum, FWHM), sheath gas flow rate of 5, seep gas flow rate of 0, auxiliary gas flow rate of 0, spray voltage of 4.20 kV, capillary temperature of 320 °C, S-lens Rf level, and automatic gain control of 5 × 106. The MS/MS studies were performed using the same instrument using three distinct stepwise normalized collision energies (10, 30, and 40) [5]. The Xcalibur 3.1 with Foundation 3.1 (Thermo Fisher Scientific Inc. Rockford, IL, USA) was used to process the collected mass spectral data. The m/z peaks were tentatively identified by comparing their calculated (exact) masses of deprotonated (M–H) adducts with the m/z values and ESI-MS/MS fragmentation patterns from the in-house MS/MS database and online databases such as FooDB [49], METLIN [50], CFM-ID 4.0 [48]. The chemical structure was drawn using ChemDraw Professional 15.0 (PerkinElmer, Waltham, MA, USA).

3.8. Statistical Analysis

All data were reported as the mean ± standard deviation of at least three independent experiments (n = 3), each with three sample replicates. One-way analysis of variance (ANOVA), followed by Dunnett’s multiple comparison test, was executed using SigmaPlot Version 12.5 (Systat Software, Inc., Chicago, IL, USA) to determine statistical significance at p < 0.001, p < 0.01, and p < 0.05. Principle component analysis (PCA) was performed to analyze the effect of the extraction method on TPC, TFC, antioxidant, mushroom tyrosinase, α-glucosidase and elastase enzyme inhibition and to learn the correlations between these variables. PCA was carried out using Minitab Statistical Software (Version 18.0, Minitab Inc., Enterprise Drive State College, PA, USA).

4. Conclusions

This study was the first investigation on optimizing the solvent extraction conditions on URADP using RSM, and high-resolution mass spectroscopic analysis revealed the presence of phenolic acids, flavonoids, lignans, etc. Optimal conditions (52% ethanol, extraction time of 81 min, and extraction temperature of 63 °C) were determined. Under these conditions, the maximum TPC and TFC were obtained as 24.25 mgGAE/g and 23.98 mgCAE/g, respectively. Optimized extract (OP) and heat extract made using 100% methanol (HM) also showed significant antioxidant and anti-tyrosinase enzyme activity compared to other extracts. Furthermore, on the basis of their bioactive components and biological activities, chemometric analysis showed a substantial association between the HM and OP by grouping them together. However, the mechanism underlying URADP’s antioxidant and depigmenting actions is still unknown. The antioxidant and depigmenting actions of URADP are still being confirmed in investigations using cells and animal models. Based on these outcomes, we can conclude that these findings can be used as the basis for a broad commercial application of URADP, a promising candidate for an antioxidant and tyrosinase as enzymatic inhibition functional food, in nutraceutical food and pharmaceutical industries.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms24043396/s1.

Author Contributions

F.A.: methodology, formal analysis, investigation, writing—original draft. M.B.A.: conceptualization, investigation, formal analysis, writing—review and editing. B.-R.S.: formal analysis, investigation; S.-H.L.: conceptualization, methodology, supervision, funding acquisition, project administration, writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the National Research Foundation of Korea (NRF) funded by the Ministry of Science and ICT (2020R1A2C2011495 and 2021R1IA1A01058062).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Upon request, Authors will provide the data.

Acknowledgments

This study was supported by the Education Ministry of the Kingdom of Saudi Arabia (EMSA) for the author Fanar Alshammari, who received financial support for his Ph.D. study project.

Conflicts of Interest

The authors declare that they have no known competing financial interest or personal relationships that could have appeared to influence the work reported in this paper.

References

  1. Simić, V.M.; Rajković, K.M.; Stojičević, S.S.; Veličković, D.T.; Nikolić, N.Č.; Lazić, M.L.; Karabegović, I.T. Optimization of microwave-assisted extraction of total polyphenolic compounds from chokeberries by response surface methodology and artificial neural network. Sep. Purif. Technol. 2016, 160, 89–97. [Google Scholar] [CrossRef]
  2. Bochi, V.C.; Barcia, M.T.; Rodrigues, D.; Speroni, C.S.; Giusti, M.M.; Godoy, H.T. Polyphenol extraction optimisation from Ceylon gooseberry (Dovyalis hebecarpa) pulp. Food Chem. 2014, 164, 347–354. [Google Scholar] [CrossRef] [PubMed]
  3. Samaram, S.; Mirhosseini, H.; Tan, C.P.; Ghazali, H.M.; Bordbar, S.; Serjouie, A. Optimisation of ultrasound-assisted extraction of oil from papaya seed by response surface methodology: Oil recovery, radical scavenging antioxidant activity, and oxidation stability. Food Chem. 2015, 172, 7–17. [Google Scholar] [CrossRef] [PubMed]
  4. Sedraoui, S.; Badr, A.; Barba, M.G.M.; Doyen, A.; Tabka, Z.; Desjardins, Y. Optimization of the Ultrahigh-Pressure–Assisted Extraction of Phenolic Compounds and Antioxidant Activity from Palm Dates (Phoenix dactylifera L.). Food Anal. Methods 2020, 13, 1556–1569. [Google Scholar] [CrossRef]
  5. Choi, H.-J.; Naznin, M.; Alam, M.B.; Javed, A.; Alshammari, F.H.; Kim, S.; Lee, S.-H. Optimization of the extraction conditions of Nypa fruticans Wurmb. using response surface methodology and artificial neural network. Food Chem. 2022, 381, 132086. [Google Scholar] [CrossRef]
  6. Javed, A.; Naznin, M.; Alam, M.B.; Fanar, A.; Song, B.-R.; Kim, S.; Lee, S.-H. Metabolite Profiling of Microwave-Assisted Sargassum fusiforme Extracts with Improved Antioxidant Activity Using Hybrid Response Surface Methodology and Artificial Neural Networking-Genetic Algorithm. Antioxidants 2022, 11, 2246. [Google Scholar] [CrossRef]
  7. Hou, M.; Hu, W.; Wang, A.; Xiu, Z.; Shi, Y.; Hao, K.; Sun, X.; Cao, D.; Lu, R.; Sun, J. Ultrasound-Assisted Extraction of Total Flavonoids from Pteris cretica L.: Process Optimization, HPLC Analysis, and Evaluation of Antioxidant Activity. Antioxidants 2019, 8, 425. [Google Scholar] [CrossRef]
  8. Alam, M.B.; Ahmed, A.; Motin, M.A.; Kim, S.; Lee, S.H. Attenuation of melanogenesis by Nymphaea nouchali (Burm. f) flower extract through the regulation of cAMP/CREB/MAPKs/MITF and proteasomal degradation of tyrosinase. Sci. Rep. 2018, 8, 13928. [Google Scholar] [CrossRef]
  9. Kim, J.H.; Jang, D.H.; Lee, K.W.; Kim, K.D.; Shah, A.B.; Zhumanova, K.; Park, K.H. Tyrosinase Inhibition and Kinetic Details of Puerol A Having But-2-Enolide Structure from Amorpha fruticosa. Molecules 2020, 25. [Google Scholar] [CrossRef]
  10. Kim, D.S.; Cha, S.B.; Park, M.C.; Park, S.A.; Kim, H.S.; Woo, W.H.; Mun, Y.J. Scopoletin Stimulates Melanogenesis via cAMP/PKA Pathway and Partially p38 Activation. Biol. Pharm. Bull. 2017, 40, 2068–2074. [Google Scholar] [CrossRef] [Green Version]
  11. Shah, A.B.; Yoon, S.; Kim, J.H.; Zhumanova, K.; Ban, Y.J.; Lee, K.W.; Park, K.H. Effectiveness of cyclohexyl functionality in ugonins from Helminthostachys zeylanica to PTP1B and α-glucosidase inhibitions. Int. J. Biol. Macromol. 2020, 165, 1822–1831. [Google Scholar] [CrossRef]
  12. Jakimiuk, K.; Gesek, J.; Atanasov, A.G.; Tomczyk, M. Flavonoids as inhibitors of human neutrophil elastase. J. Enzym. Inhib. Med. Chem. 2021, 36, 1016–1028. [Google Scholar] [CrossRef]
  13. Alam, S.R.; Newby, D.E.; Henriksen, P.A. Role of the endogenous elastase inhibitor, elafin, in cardiovascular injury: From epithelium to endothelium. Biochem. Pharm. 2012, 83, 695–704. [Google Scholar] [CrossRef]
  14. Ban, Y.J.; Baiseitova, A.; Nafiah, M.A.; Kim, J.Y.; Park, K.H. Human neutrophil elastase inhibitory dihydrobenzoxanthones and alkylated flavones from the Artocarpus elasticus root barks. Appl. Biol. Chem. 2020, 63, 63. [Google Scholar] [CrossRef]
  15. von Nussbaum, F.; Li, V.M. Neutrophil elastase inhibitors for the treatment of (cardio)pulmonary diseases: Into clinical testing with pre-adaptive pharmacophores. Bioorg. Med. Chem. Lett. 2015, 25, 4370–4381. [Google Scholar] [CrossRef]
  16. Delgado-Rizo, V.; Martínez-Guzmán, M.A.; Iñiguez-Gutierrez, L.; García-Orozco, A.; Alvarado-Navarro, A.; Fafutis-Morris, M. Neutrophil Extracellular Traps and Its Implications in Inflammation: An Overview. Front. Immunol. 2017, 8, 81. [Google Scholar] [CrossRef]
  17. Yang, H.; Biermann, M.H.; Brauner, J.M.; Liu, Y.; Zhao, Y.; Herrmann, M. New Insights into Neutrophil Extracellular Traps: Mechanisms of Formation and Role in Inflammation. Front. Immunol. 2016, 7, 302. [Google Scholar] [CrossRef]
  18. Tabaraki, R.; Nateghi, A. Optimization of ultrasonic-assisted extraction of natural antioxidants from rice bran using response surface methodology. Ultrason. Sonochem. 2011, 18, 1279–1286. [Google Scholar] [CrossRef]
  19. Bezerra, M.A.; Santelli, R.E.; Oliveira, E.P.; Villar, L.S.; Escaleira, L.A. Response surface methodology (RSM) as a tool for optimization in analytical chemistry. Talanta 2008, 76, 965–977. [Google Scholar] [CrossRef]
  20. Yasin, B.R.; El-Fawal, H.A.; Mousa, S.A. Date (Phoenix dactylifera) Polyphenolics and Other Bioactive Compounds: A Traditional Islamic Remedy’s Potential in Prevention of Cell Damage, Cancer Therapeutics and Beyond. Int. J. Mol. Sci. 2015, 16, 30075–30090. [Google Scholar] [CrossRef] [Green Version]
  21. Al-Yahya, M.; Raish, M.; AlSaid, M.S.; Ahmad, A.; Mothana, R.A.; Al-Sohaibani, M.; Al-Dosari, M.S.; Parvez, M.K.; Rafatullah, S. ‘Ajwa’ dates (Phoenix dactylifera L.) extract ameliorates isoproterenol-induced cardiomyopathy through downregulation of oxidative, inflammatory and apoptotic molecules in rodent model. Phytomed. Int. J. Phytother. Phytopharm. 2016, 23, 1240–1248. [Google Scholar] [CrossRef] [PubMed]
  22. Almatroodi, S.A.; Khan, A.A.; Aloliqi, A.A.; Ali Syed, M.; Rahmani, A.H. Therapeutic Potential of Ajwa Dates (Phoenix dactylifera) Extract in Prevention of Benzo(a)pyrene-Induced Lung Injury through the Modulation of Oxidative Stress, Inflammation, and Cell Signalling Molecules. Appl. Sci. 2022, 12, 6784. [Google Scholar] [CrossRef]
  23. Abdelghffar, E.A.; Obaid, W.A.; Mohammedsaleh, Z.M.; Ouchari, W.; Eldahshan, O.A.; Sobeh, M. Ajwa dates (Phoenix dactylifera L.) attenuate cisplatin-induced nephrotoxicity in rats via augmenting Nrf2, modulating NADPH oxidase-4 and mitigating inflammatory/apoptotic mediators. Biomed. Pharmacother. Biomed. Pharmacother. 2022, 156, 113836. [Google Scholar] [CrossRef] [PubMed]
  24. Al-Shahib, W.; Marshall, R.J. The fruit of the date palm: Its possible use as the best food for the future? Int. J. Food Sci. Nutr. 2003, 54, 247–259. [Google Scholar] [CrossRef] [PubMed]
  25. Rahmani, A.H.; Aly, S.M.; Ali, H.; Babiker, A.Y.; Srikar, S.; Khan, A.A. Therapeutic effects of date fruits (Phoenix dactylifera) in the prevention of diseases via modulation of anti-inflammatory, anti-oxidant and anti-tumour activity. Int. J. Clin. Exp. Med. 2014, 7, 483–491. [Google Scholar]
  26. Boulenouar, N.; Marouf, A.; Cheriti, A. Antifungal activity and phytochemical screening of extracts from Phoenix dactylifera L. cultivars. Nat. Prod. Res. 2011, 25, 1999–2002. [Google Scholar] [CrossRef]
  27. Almusallam, I.A.; Ahmed, I.A.M.; Babiker, E.E.; Al Juhaimi, F.Y.; Fadimu, G.J.; Osman, M.A.; Al Maiman, S.A.; Ghafoor, K.; Alqah, H.A. Optimization of ultrasound-assisted extraction of bioactive properties from date palm (Phoenix dactylifera L.) spikelets using response surface methodology. LWT 2021, 140, 110816. [Google Scholar] [CrossRef]
  28. Ghafoor, K.; Sarker, M.Z.I.; Al-Juhaimi, F.Y.; Babiker, E.E.; Alkaltham, M.S.; Almubarak, A.K. Extraction and Evaluation of Bioactive Compounds from Date (Phoenix dactylifera) Seed Using Supercritical and Subcritical CO2 Techniques. Foods 2022, 11, 1806. [Google Scholar] [CrossRef]
  29. Pourshoaib, S.J.; Ghatrami, E.R.; Shamekhi, M.A. Comparing ultrasonic-and microwave-assisted methods for extraction of phenolic compounds from Kabkab date seed (Phoenix dactylifera L.) and stepwise regression analysis of extracts antioxidant activity. Sustain. Chem. Pharm. 2022, 30, 100871. [Google Scholar] [CrossRef]
  30. Al-Turki, S.; Shahba, M.A.; Stushnoff, C. Diversity of antioxidant properties and phenolic content of date palm (Phoenix dactylifera L.) fruits as affected by cultivar and location. J. Food Agric. Environ. 2010, 8, 253–260. [Google Scholar]
  31. Mohamed, S.A.; Awad, M.A.; El-Dengawy, E.-R.F.A.; Abdel-Mageed, H.M.; El-Badry, M.O.; Salah, H.A.; Abdel-Aty, A.M.; Fahmy, A.S. Total phenolic and flavonoid contents and antioxidant activities of sixteen commercial date cultivars grown in Saudi Arabia. RSC Adv. 2016, 6, 44814–44819. [Google Scholar] [CrossRef]
  32. Hamad, I.; AbdElgawad, H.; Al Jaouni, S.; Zinta, G.; Asard, H.; Hassan, S.; Hegab, M.; Hagagy, N.; Selim, S. Metabolic Analysis of Various Date Palm Fruit (Phoenix dactylifera L.) Cultivars from Saudi Arabia to Assess Their Nutritional Quality. Molecules 2015, 20, 13620–13641. [Google Scholar] [CrossRef]
  33. Eid, N.M.S.; Al-Awadi, B.; Vauzour, D.; Oruna-Concha, M.J.; Spencer, J.P.E. Effect of Cultivar Type and Ripening on the Polyphenol Content of Date Palm Fruit. J. Agric. Food Chem. 2013, 61, 2453–2460. [Google Scholar] [CrossRef]
  34. Hassan, S.M.A.; Aboonq, M.S.; Albadawi, E.A.; Aljehani, Y.; Abdel-Latif, H.M.; Mariah, R.A.; Shafik, N.M.; Soliman, T.M.; Abdel-Gawad, A.R.; Omran, F.M.; et al. The Preventive and Therapeutic Effects of Ajwa Date Fruit Extract Against Acute Diclofenac Toxicity-Induced Colopathy: An Experimental Study. Drug Des. Devel. 2022, 16, 2601–2616. [Google Scholar] [CrossRef]
  35. Xi, J.; Wang, B. Optimization of Ultrahigh-Pressure Extraction of Polyphenolic Antioxidants from Green Tea by Response Surface Methodology. Food Bioprocess Technol. 2013, 6, 2538–2546. [Google Scholar] [CrossRef]
  36. Gurtovenko, A.A.; Anwar, J. Interaction of Ethanol with Biological Membranes: The Formation of Non-bilayer Structures within the Membrane Interior and their Significance. J. Phys. Chem. B 2009, 113, 1983–1992. [Google Scholar] [CrossRef]
  37. Derrien, M.; Badr, A.; Gosselin, A.; Desjardins, Y.; Angers, P. Optimization of a green process for the extraction of lutein and chlorophyll from spinach by-products using response surface methodology (RSM). LWT-Food Sci. Technol. 2017, 79, 170–177. [Google Scholar] [CrossRef]
  38. Xu, S.; Li, X.; Liu, S.; Tian, P.; Li, D. Juniperus sabina L. as a Source of Podophyllotoxins: Extraction Optimization and Anticholinesterase Activities. Int. J. Mol. Sci. 2022, 23, 10205. [Google Scholar] [CrossRef]
  39. Alam, M.B.; Ju, M.K.; Lee, S.H. DNA Protecting Activities of Nymphaea nouchali (Burm. f) Flower Extract Attenuate t-BHP-Induced Oxidative Stress Cell Death through Nrf2-Mediated Induction of Heme Oxygenase-1 Expression by Activating MAP-Kinases. Int. J. Mol. Sci. 2017, 18, 2069. [Google Scholar] [CrossRef]
  40. Rebollo-Hernanz, M.; Cañas, S.; Taladrid, D.; Segovia, Á.; Bartolomé, B.; Aguilera, Y.; Martín-Cabrejas, M.A. Extraction of phenolic compounds from cocoa shell: Modeling using response surface methodology and artificial neural networks. Sep. Purif. Technol. 2021, 270, 118779. [Google Scholar] [CrossRef]
  41. Sunarwidhi, A.L.; Hernawan, A.; Frediansyah, A.; Widyastuti, S.; Martyasari, N.W.R.; Abidin, A.S.; Padmi, H.; Handayani, E.; Utami, N.W.P.; Maulana, F.A.; et al. Multivariate Analysis Revealed Ultrasonic-Assisted Extraction Improves Anti-Melanoma Activity of Non-Flavonoid Compounds in Indonesian Brown Algae Ethanol Extract. Molecules 2022, 27, 7509. [Google Scholar] [CrossRef] [PubMed]
  42. Naznin, M.; Badrul Alam, M.; Alam, R.; Islam, S.; Rakhmat, S.; Lee, S.-H.; Kim, S. Metabolite profiling of Nymphaea rubra (Burm. f.) flower extracts using cyclic ion mobility–mass spectrometry and their associated biological activities. Food Chem. 2023, 404, 134544. [Google Scholar] [CrossRef] [PubMed]
  43. Alam, M.B.; Naznin, M.; Islam, S.; Alshammari, F.H.; Choi, H.J.; Song, B.R.; Kim, S.; Lee, S.H. High Resolution Mass Spectroscopy-Based Secondary Metabolite Profiling of Nymphaea nouchali (Burm. f) Stem Attenuates Oxidative Stress via Regulation of MAPK/Nrf2/HO-1/ROS Pathway. Antioxidants 2021, 10, 719. [Google Scholar] [CrossRef] [PubMed]
  44. Najm, O.A.; Addnan, F.H.; Mohd-Manzor, N.F.; Elkadi, M.A.; Abdullah, W.O.; Ismail, A.; Mansur, F.A.F. Identification of Phytochemicals of Phoenix dactylifera L. Cv Ajwa with UHPLC-ESI-QTOF-MS/MS. Int. J. Fruit Sci. 2021, 21, 848–867. [Google Scholar] [CrossRef]
  45. Nematallah, K.A.; Ayoub, N.A.; Abdelsattar, E.; Meselhy, M.R.; Elmazar, M.M.; El-Khatib, A.H.; Linscheid, M.W.; Hathout, R.M.; Godugu, K.; Adel, A.; et al. Polyphenols LC-MS2 profile of Ajwa date fruit (Phoenix dactylifera L.) and their microemulsion: Potential impact on hepatic fibrosis. J. Funct. Foods 2018, 49, 401–411. [Google Scholar] [CrossRef]
  46. 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]
  47. Vukics, V.; Guttman, A. Structural characterization of flavonoid glycosides by multi-stage mass spectrometry. Mass Spectrom. Rev. 2010, 29, 1–16. [Google Scholar] [CrossRef]
  48. Wang, F.; Liigand, J.; Tian, S.; Arndt, D.; Greiner, R.; Wishart, D.S. CFM-ID 4.0: More Accurate ESI-MS/MS Spectral Prediction and Compound Identification. Anal. Chem. 2021, 93, 11692–11700. [Google Scholar] [CrossRef]
  49. Naveja, J.J.; Rico-Hidalgo, M.P.; Medina-Franco, J.L. Analysis of a large food chemical database: Chemical space, diversity, and complexity. F1000Research 2018, 7. [Google Scholar] [CrossRef]
  50. Domingo-Almenara, X.; Guijas, C.; Billings, E.; Montenegro-Burke, J.R.; Uritboonthai, W.; Aisporna, A.E.; Chen, E.; Benton, H.P.; Siuzdak, G. The METLIN small molecule dataset for machine learning-based retention time prediction. Nat. Commun. 2019, 10, 5811. [Google Scholar] [CrossRef]
  51. Mollica, A.; Zengin, G.; Sinan, K.I.; Marletta, M.; Pieretti, S.; Stefanucci, A.; Etienne, O.K.; Jekő, J.; Cziáky, Z.; Bahadori, M.B.; et al. A Study on Chemical Characterization and Biological Abilities of Alstonia boonei Extracts Obtained by Different Techniques. Antioxidants 2022, 11, 2171. [Google Scholar] [CrossRef]
  52. Alam, M.B.; Bajpai, V.K.; Ra, J.S.; Lim, J.Y.; An, H.; Shukla, S.; Quan, K.T.; Khan, I.; Huh, Y.S.; Han, Y.K.; et al. Anthraquinone-type inhibitor of α-glucosidase enhances glucose uptake by activating an insulin-like signaling pathway in C2C12 myotubes. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2019, 129, 337–343. [Google Scholar] [CrossRef]
  53. Zhao, P.; Alam, M.B.; Lee, S.-H. Protection of UVB-Induced Photoaging by Fuzhuan-Brick Tea Aqueous Extract via MAPKs/Nrf2-Mediated Down-Regulation of MMP-1. Nutrients 2019, 11, 60. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The three-dimensional (3D) response surface plots of URADP extraction for (A) TPC and (B) TFC for ethanol concentration, time, and temperature as a function of key interaction factors for RSM.
Figure 1. The three-dimensional (3D) response surface plots of URADP extraction for (A) TPC and (B) TFC for ethanol concentration, time, and temperature as a function of key interaction factors for RSM.
Ijms 24 03396 g001
Figure 2. Desirability surface plot: as a function of (A) ethanol concentration and extraction time; (B) ethanol concentration and extraction temperature; (C) extraction time and temperature.
Figure 2. Desirability surface plot: as a function of (A) ethanol concentration and extraction time; (B) ethanol concentration and extraction temperature; (C) extraction time and temperature.
Ijms 24 03396 g002
Figure 3. The pharmacological activities of the different extracts of URADP. (A) Total phenolic content (TPC) and total flavonoid content (TFC). (B) IC50 values of DPPH–, ABTS– radical scavenging, tyrosinase, α-glucosidase and elastase enzyme inhibition activity. (C) Biplot (scores of samples and load factors of each variable) of the principal component analysis (PCA). (D) Hierarchical cluster analysis (HCA). OP: heat extract with optimized condition, HM: heat extract with 100% methanol, HE: heat extract with 100% ethanol, HW: heat extract with 100% H2O, MM: maceration extract with 100% methanol, ME: maceration extract with 100% ethanol, MW: maceration extract with 100% H2O.
Figure 3. The pharmacological activities of the different extracts of URADP. (A) Total phenolic content (TPC) and total flavonoid content (TFC). (B) IC50 values of DPPH–, ABTS– radical scavenging, tyrosinase, α-glucosidase and elastase enzyme inhibition activity. (C) Biplot (scores of samples and load factors of each variable) of the principal component analysis (PCA). (D) Hierarchical cluster analysis (HCA). OP: heat extract with optimized condition, HM: heat extract with 100% methanol, HE: heat extract with 100% ethanol, HW: heat extract with 100% H2O, MM: maceration extract with 100% methanol, ME: maceration extract with 100% ethanol, MW: maceration extract with 100% H2O.
Ijms 24 03396 g003
Figure 4. Tentative mass fragmentation behavior of 1,2-Di-(syringoyl)-hexoside (A), luteone glucoside (B), lyoniresinol 9-O-glucoside (C) and xylosmaloside (D).
Figure 4. Tentative mass fragmentation behavior of 1,2-Di-(syringoyl)-hexoside (A), luteone glucoside (B), lyoniresinol 9-O-glucoside (C) and xylosmaloside (D).
Ijms 24 03396 g004
Table 1. Central composite design (CCD) for independent variables and corresponding response values (experimental).
Table 1. Central composite design (CCD) for independent variables and corresponding response values (experimental).
RunIndependent VariablesResponses
(X1)(X2)(X3)TPC (Y1)TFC (Y2)
Exp.Pred.Exp.Pred.
110082.5605.41 ± 0.285.7011.02 ± 0.339.90
25082.56023.69 ± 0.4323.3421.05 ± 0.6223.10
3751207013.75 ± 0.5414.1712.9 ± 0.1513.44
450156010.24 ± 0.769.7511.52 ± 0.2510.24
5751205010.21 ± 0.6110.607.83 ± 0.399.31
65082.56023.12 ± 0.1223.3424.20 ± 0.2023.10
7082.5606.53 ± 0.126.736.85 ± 0.166.04
825457017.50 ± 0.6917.6211.52 ± 0.4611.97
925120508.88 ± 0.458.996.81 ± 0.357.39
105082.58023.00 ± 0.4322.8916.05 ± 0.7515.28
115082.56023.10 ± 0.7223.3423.59 ± 0.3623.10
125082.56023.51 ± 0.1623.3424.01 ± 0.4323.10
135082.56023.92 ± 0.5423.3424.01 ± 0.6323.10
142545507.85 ± 0.727.947.85 ± 0.559.25
15501506010.11 ± 0.469.979.25 ± 0.258.14
1675455011.01 ± 0.0411.7415.25 ± 0.8016.05
17251207019.22 ± 0.5819.0015.25 ± 0.9216.38
185082.54010.05 ± 0.189.649.59 ± 0.228.43
195082.56023.10 ± 0.5323.3423.25 ± 0.5923.10
2075457014.58 ± 0.5414.9812.56 ± 0.2713.92
X1: ethanol concentration (%); X2: time (min); X3: temperature (°C); TPC: total phenolic content (mg gallic acid equivalent/g dry weight extract); TFC: total flavonoid content (mg catechin equivalent/g dry weight extract). Exp.: experimental value; Pred.: predicted value.
Table 2. ANOVA for quadratic model.
Table 2. ANOVA for quadratic model.
ANOVA for Quadratic Model for TPC
SourceRCSSDFMSF Valuep Value
Model 843.91993.77347.63<0.0001Significant
Intercept23.39
Linear terms
X1−0.38382.3612.368.740.0144Significant
X20.06120.054210.05420.20100.6635Nonsignificant
X33.06150.311150.31557.25<0.0001Significant
Interaction terms
X1X2−0.54752.4012.408.890.0138Significant
X1X3−1.6120.74120.7476.88<0.0001Significant
X2X30.08250.054410.05440.20190.6628Nonsignificant
Quadratic terms
X12−4.37484.621484.621796.67<0.0001Significant
X22−4.10292.301292.301083.65<0.0001Significant
X32−1.9899.41199.41368.54<0.0001Significant
Lack of Fit 2.0750.41453.320.1071Nonsignificant
Pure error 0.624750.1249
R2 0.9968
Adjusted R2 0.9939
Adeq Precision 49.6969
C.V.% 3.39
ANOVA for quadratic model for TFC
Model 751.10983.4636.64<0.0001Significant
Intercept23.10
Linear terms
X10.965614.92114.926.550.0284Significant
X2−0.58544.9614.962.180.1708Nonsignificant
X31.7146.96146.9620.610.0011Significant
Interaction terms
X1X2−1.2211.93111.935.240.0451Significant
X1X3−1.2211.83111.835.200.0458Significant
X2X31.5719.63119.638.620.0149Significant
Quadratic terms
X12−3.78363.271363.27159.47<0.0001Significant
X22−4.29320.241320.24140.58<0.0001Significant
X32−2.81200.731200.7388.12<0.0001Significant
Lack of Fit 15.8353.172.280.1938Nonsignificant
Pure error 6.9551.39
R2 0.9706
Adjusted R2 0.9441
Adeq Precision 15.9930
C.V.% 10.25
RC: regression coefficient; SS: sum of squares; MS: mean square.
Table 3. Experiment data of the validation of predicted values at optimal extraction conditions of URADP.
Table 3. Experiment data of the validation of predicted values at optimal extraction conditions of URADP.
ResponseExp.Pred.StdRSD (%)
TPC (mgGAE/g)24.25 ± 1.0223.970.200.82
TFC (mgCAE/g)23.98 ± 0.6523.390.421.76
Optimal condition: ethanol concentration (%): 51.97%; time (min): 81.38; temperature (°C): 62.76. Exp.: experimental value; Pred.: predicted value; Std: standard deviation; RSD: relative standard deviation.
Table 4. List of tentative identified compounds of the optimized extract of URADP by electrospray ionization mass spectrometry (ESI-MS)/MS.
Table 4. List of tentative identified compounds of the optimized extract of URADP by electrospray ionization mass spectrometry (ESI-MS)/MS.
GroupNo.Compound NameEFOM
(m/z)
CM
(m/z)
MS/MS (Negative Mode)CECL
Phenolic acids and derivatives14-Hydroxybenzoyl glucoseC13H16O8299.0773299.0766137.02, 163.02202
2Coumaroylshikimic acidC16H16O7319.0824319.0817173.04, 163.03, 145.02202
3Vanillic acid glucosideC14H18O9329.0873329.0872167.03, 152.02, 123.04202
4Caffeoylshikimic acidC16H16O8335.0776335.0772179.01, 161.03, 155.03, 137.05202
5Quinic acid hexosideC13H22O11353.1085353.1084191.05, 173.04, 179.05202
65-Feruloylquinic acidC17H20O9367.1046367.1029191.08, 173.04, 127.01302
5Caffeic acid derivativesC18H18O9377.0885377.0878341.10, 215.03, 179.06, 161.04, 135.05102
6Sinapic acid hexosideC17H22O10385.1141385.1135223.06, 205.05102
7Caffeoyl shikimic acid hexosideC22H26O13497.1297497.1295335.01, 178.02, 161.03, 155.03, 135.02202
8Quinic acid derivativesC19H34O17533.1718533.1718341.10, 191.05302
91,2-di-(syringoyl)-hexoside #C24H28O14539.1377539.1401359.09, 341.08, 197.04, 153.05303
Flavonoids and derivatives10LuteolinC15H10O6285.0405285.0399267.05, 241.03, 151.00, 133.02202
11Catechin/EpicatechinC15H14O6289.0718289.0712245.04, 205.05, 179, 151.04, 137.02202
12ChrysoeriolC13H16O8299.0561299.0555285.03, 153.01, 135.03, 125.03202
13QuercetinC15H10O7301.0354301.0348273.02, 229.05, 179.01, 151.01202
14EpigallocatechinC15H14O7305.0644305.0661287.05, 137.02, 125.02202
15Methoxysinensetin #C21H22O8401.1299401.1236371.11, 339.08, 191.71202
16Epicatechin hydroxybenzoate #C22H18O8409.0924409.0923289.07, 271.06, 137.02, 119.01302
17Naringenin rhamnosideC21H22O9417.1245417.1186271.06, 187.03, 151.00, 119.05202
18Epicatechin-3-gallateC22H18O10441.081441.0821371.04, 273.02, 135.10, 169.02302
19Biochanin A 7-glucoside #C22H22O10445.1195445.1135283.06, 239.03, 211.04, 132.02302
20Epicatechin 3-(-methylgallate) #C23H20O10455.1015455.0978289.02, 183.05, 124.01302
21Afrormosin 7-glucosideC23H24O10459.1354459.1291297.07, 281.04, 267.06202
22Chrysoeriol hexosideC22H22O11461.1085461.1083299.07, 283.02, 269.06202
23IsoquercitrinC21H20O12463.0878463.0876301.05, 268.01, 179.02, 151.01202
24Epicatechin 4’-glucuronide#C21H22O12465.1036465.1033289.15, 151.10, 137.08, 123.10202
25Isorhamnetin hexosideC22H22O12477.1035477.1033315.05, 300.01, 179.05, 151.02202
26Luteone glucosideC26H28O11515.1611515.1553353.10, 311.05, 297.04203
27Luteolin hexosyl sulfateC21H20O14S527.0491527.0495447.05, 285.01, 241.06202
28Chrysoeriol hexosyl sulfateC22H22O14S541.0645541.0652299.05, 284.05, 241.02202
29Isoquercitrin sulfateC21H20O15S543.0441543.0444463.05, 301.01, 179.02, 151.01202
30Procyanidin B2 #C30H26O12577.1347577.1346451.10, 407.07, 289.07, 287.05, 125.02202
31Lyoniresinol 9-glucoside #C28H37O13581.2236581.2234419.17, 265.10, 247.09202
32Luteolin rhamnosyl hexosideC27H30O15593.1507593.1506447.09, 285.03, 153.01, 135.04202
33Chrysoeriol rhamnosyl hexosideC28H32O15607.1669607.1663461.10, 299.05, 153.01, 149.05202
34Isorhamnetin rhamnosyl hexosideC28H32O16623.1617623.1612477.10, 315.05, 299.05, 165.05202
35Isorhamnetin diglucosideC28H32O17639.1563639.1561447.01, 315.01202
36Quercetin xylosyl rutinoside #C32H38O20741.1846741.1878609.14, 301.03102
37Luteolin rhamnosyl dihexosideC33H40O20755.2046755.2034709.16, 593.10, 575.05, 285.01202
38Quercetin glucosyl-rutinosideC33H40O21771.1981771.1983609.14, 591.05, 301.03, 153.02, 125.00202
39Isorhamnetin rhamnosyl dihexosideC34H42O21785.211785.214623.16, 477.10, 315.05202
40Epicatechin-(2α→7,4α→8)-epicatechin glucoside #C36H34O17737.1721737.1718721.02, 577.05, 425.05, 195.02302
Sugar molecules41Ribonic acidC5H10O6165.0421165.0418149.04, 105.01, 87.00, 75.00102
42L-GalactoseC6H12O6179.0572179.0561161.04, 143.03, 113.02, 101.02,102
43Gluconic acidC6H12O7195.0522195.0504177.05, 159.02, 129.05, 98.90102
48SedoheptuloseC7H14O7209.0679209.068191.05, 179.05, 149.04,202
49Xylosmaloside #C18H20O9379.1027379.1029343.08, 217.05, 179.05, 161.04203
Carboxylic acids50Fumaric acidC4H4O4115.005115.003771.01102
51Glutaconic acidC5H6O4129.0203129.0203111.00, 85.02102
52Glutaric acidC5H8O4131.0355131.035113.00, 87.02102
533-Methylglutaconic acidC6H8O4143.0367143.036199.03202
54Methyl glutaric acidC6H10O4145.0521145.0506127.02, 101.02102
552-Hydroxyglutaric acidC5H8O5147.0301147.0299129.01, 99.03102
56Hydroxymethyl glutaric acidC6H10O5161.0459161.0455143.03, 117.05, 99.04102
58Citric acidC6H8O7191.0197191.0197173.00, 129.01, 111.00202
Fatty acids59Palmitic acidC16H32O2255.233255.233237.23, 211.24, 197.22202
60Linolenic acidC18H30O2277.2165277.2169259.20, 233.22, 205.21, 179.25,102
61α-Linoleic acidC18H32O2279.2331279.2330261.22102
62Oleic acidC18H34O2281.2487281.2486263.25, 181.21, 127.25102
63Hydroxy octadecatrienoic acid #C18H30O3293.212293.0216275.22203
64Hydroxy octadecadienoic acidC18H32O3295.2276295.2273277.23202
65Hydroxy octadecenoic acidC18H34O3297.2433297.2429279.23202
66Dihydroxy octadecadienoic acidC18H32O4311.2246311.2239293.22, 275.23202
67Dihydroxy octadecenoic acidC18H34O4313.2381313.2378295.23, 277.25, 183.32202
68Dihydroxy octadecanoic acidC18H36O4315.2538315.2535297.23, 279.25,202
69Trihydroxy octadecadienoic acidC18H32O5327.2176327.2171309.23, 291.25, 273.23202
70Trihydroxy octadecenoic acidC18H34O5329.2346329.2333311.25, 293.26, 275.23202
EF: elemental formula; OM: observed mass; CM: calculated mass; CE: collision energy (eV); CL: confidence level; # First time identification in Ajwa date.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Alshammari, F.; Alam, M.B.; Song, B.-R.; Lee, S.-H. Antioxidant, Tyrosinase, α-Glucosidase, and Elastase Enzyme Inhibition Activities of Optimized Unripe Ajwa Date Pulp (Phoenix dactylifera) Extracts by Response Surface Methodology. Int. J. Mol. Sci. 2023, 24, 3396. https://doi.org/10.3390/ijms24043396

AMA Style

Alshammari F, Alam MB, Song B-R, Lee S-H. Antioxidant, Tyrosinase, α-Glucosidase, and Elastase Enzyme Inhibition Activities of Optimized Unripe Ajwa Date Pulp (Phoenix dactylifera) Extracts by Response Surface Methodology. International Journal of Molecular Sciences. 2023; 24(4):3396. https://doi.org/10.3390/ijms24043396

Chicago/Turabian Style

Alshammari, Fanar, Md Badrul Alam, Bo-Rim Song, and Sang-Han Lee. 2023. "Antioxidant, Tyrosinase, α-Glucosidase, and Elastase Enzyme Inhibition Activities of Optimized Unripe Ajwa Date Pulp (Phoenix dactylifera) Extracts by Response Surface Methodology" International Journal of Molecular Sciences 24, no. 4: 3396. https://doi.org/10.3390/ijms24043396

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