Next Article in Journal
The Impact of Nonalcoholic Fatty Liver Disease on Severe Community-Acquired Pneumonia Outcomes
Next Article in Special Issue
Annona muricata: Comprehensive Review on the Ethnomedicinal, Phytochemistry, and Pharmacological Aspects Focusing on Antidiabetic Properties
Previous Article in Journal
Sympatho-Vagal Dysfunction in Systemic Sclerosis: A Follow-Up Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Phytochemical Composition, Antioxidant, and Anticancer Activities of Sidr Honey: In Vitro and In Silico Computational Investigation

1
Department of Biology, College of Science, University of Hail, P.O. Box 2440, Hail 2440, Saudi Arabia
2
Research Unit: Molecular Biology of Leukemia and Lymphoma, Department of Biochemistry, University of Medecine of Sousse, Sousse 4002, Tunisia
3
Section of Histology—Cytology, University of Medicine of Tunis, University of Tunis El Manar, La Rabta 1007, Road Djebal Lakhdhar, Tunis 1007, Tunisia
4
Department of Histo-Embryology and Cytogenetics, Medicine Faculty of Sfax, University of Sfax, Road of Majida Boulia, Sfax 3029, Tunisia
5
Department of Chemistry, College of Science, University of Hail, P.O. Box 2440, Hail 2440, Saudi Arabia
6
Laboratory of Water, Membranes and Environmental Biotechnologies, Center of Research and Water Technologies, P.O. Box 273, Soliman 8020, Tunisia
7
Laboratory of Galenic and Pharmacological Chemical Development of Drugs, University of Pharmacy, Monastir 5000, Tunisia
8
Laboratory of Genetics, Biodiversity and Valorisation of Bioressources, High Institute of Biotechnology University of Monastir, Monastir 5000, Tunisia
*
Author to whom correspondence should be addressed.
Life 2023, 13(1), 35; https://doi.org/10.3390/life13010035
Submission received: 30 November 2022 / Revised: 12 December 2022 / Accepted: 19 December 2022 / Published: 23 December 2022
(This article belongs to the Special Issue Natural Products on Health and Diseases)

Abstract

:
Cancer is one of the major causes of death worldwide. The repercussions of conventional therapeutic approaches present a challenge in the delivery of new effective treatments. Thus, more attention is being awarded to natural products, mainly honey. Honey could be the basis for the development of new therapies for cancer patients. The aim of this study is to assess the phytochemical profiling, antioxidant, drug-likeness properties, and anticancer activity of Ziziphus honey (ZH) derived from the Hail region of Saudi Arabia. The phytochemical profiling using high resolution-liquid chromatography mass spectrometry (HR-LCMS) revealed 10 compounds belonging to several familial classes and one tripeptide. Potential antioxidant activity was noted as assessed by DPPH (IC50 0.670 mg/mL), ABTS (IC50 3.554 mg/mL), and β-carotene (IC50 > 5 mg/mL). The ZH exerted a notable cytotoxic effect in a dose-dependent manner against three cancer cell lines: lung (A549), breast (MCF-7), and colon (HCT-116), with respective IC50 values of 5.203%, 6.02%, and 7.257%. The drug-likeness investigation unveiled that most of the identified compounds meet Lipinski’s rule. The molecular docking analysis revealed interesting antioxidant and anticancer activities for most targeted proteins and supported the in vitro findings. The Miraxanthin-III compound exhibited the most stabilized interaction. This study provides deeper insights on ZH as prominent source of bioactive compounds with potent antioxidant and anticancer effects.

1. Introduction

Cancer represents a heavy burden on healthcare systems and is a major public concern globally. According to the International Agency for Research on Cancer (IARC), the number of diagnosed cases of cancer is increasing. Nearly 19.3 million new cases were recorded in 2020. Breast, lung, and colorectal cancer are the main types of cancer detected in both males and females, with respective rates of 11.7%, 11.4%, and 10% [1]. The number of new diagnostic cases of cancer may rise to 32.2 million in 2040 [2].
Despite thorough research targeting the prevention of cancer and providing efficient, safe treatments, the disease continues to gain range and meets challenges to overcome this ubiquitous issue. The conventional approaches of treating cancer by chemotherapy, radiation therapy, and surgery often have severe side effects on patients and are very painful. Furthermore, some malignancies may display drug resistance leading to poor prognosis and poor survival among patients. This is why complementary and alternative medicine could be a valuable substitute, offering new potential bioactive molecules with therapeutic effects. Among these natural products is honey, which is often considered one of the best sources of potent phytochemicals exhibiting interesting chemopreventive and chemotherapeutic effects against cancers [3].
Honey has been used in folk medicine and has been known for its healing properties for many years. Prominent therapeutic results have been reported to treat infectious and inflammatory diseases. Recently there has been an increased interest in honey phytochemicals and their pharmacological application in cancers. Several results showed prominent anticancer activity displayed by various honey types [4]. Honey content may vary according to floral origin, the geographical area considering the same floral origin, season, altitude, and species of honeybees [4,5,6]. Monofloral honey derived from the nectar of one plant species has a high virtue compared with polyfloral honey [7]. Saudi Arabia is among the geographic regions rich in valuable monofloral honey, such us ziziphus honey (Ziziphus spp.) (ZH) is also known as Sidr honey [8]. ZH is derived from three main plant species, namely Ziziphus nummularia, Z. mucronata, and Z. spina-christi [9].
Ziziphus spp. is considered a medicinal plant for its therapeutic virtue [10,11,12]. Consequently, valuable phytochemicals or their metabolites could be provided by ZH. Several studies reported interesting antioxidant, anti-inflammatory, and antitumor effects related to the bioactive phytochemical content in ZH. The chemopreventive effect is due mainly to antioxidant bioactive molecules, mainly polyphenols and flavonoids, which may perform scavenging activity against free radicals preventing tumorigenesis processes [13]. The anticancer potential exhibited by ZH is associated with polyphenolic profiles, such as carvacrol and its isomer thymol, found in Ziziphus Spina-Christi honey [14,15]. Both of them include compounds which may induce cytotoxicity, cell cycle arrest, apoptosis, antimetastatic activity, and may lead to inhibition of several signaling pathways and antiproliferative effects [15].
A previous in vivo study highlighted that ZH can display preventive effects against gastric ulceration in rats through antioxidant and anti-apoptotic effects [16]. In addition, it has been proven that honey could be a valuable chemoprotectant or adjuvant during cancer treatment [17]. An in vitro study showed that ZH might improve breast cancer treatment by modulating the expression of genes involved in metastasis in triple-negative breast cancer cell lines [18]. A clinical trial disclosed also that ZH could reduce significantly the severity of radiotherapy-induced mucositis in patients with head and neck cancers [18]. All these finding highlight the chemopreventive and chemotherapeutic effects displayed by ZH.
In a previous study, we studied Acacia honey derived from the Hail region. Interesting results have been reported highlighting the potential use of this honey as a source of bioactive compounds with significant antimicrobial, antioxidant, and anticancer activities, which could imply further pharmacological application. Thus, more honey subtypes should be investigated [19]. This study seeks to assess the biological activities of ZH obtained from the Hail region for the first time. The HR-LCMS method was used to investigate the phytochemical profile of ZH from the Hail geographical floral region. This methodology was used to assess the total phenols, flavonoids, and tannins, and to measure the antioxidant potential. This study will also estimate the anticancer activity of ZH against lung (A549), breast (MCF-7), and colon (HCT-116) cancer cell lines. Finally, a computational study was conducted in order to estimate the potential affinity based on conformation and complementarity between identified compounds and the active binding site of a targeted protein to explore the anticancer and antioxidant effects of ZH [19]. The drug-likeness properties were also assessed.

2. Materials and Methods

2.1. Honey Sampling

The ZH was produced from apiaries in the Hail region and was conserved at 4 °C in dark, dry, and sterile conditions. The monofloral origin was verified by microscopic observation of the honey to check for predominant pollens [20].

2.2. Phytochemical Screening of ZH

2.2.1. Polyphenol Evaluation

The total phenolic compounds in the ZH sample were measured using the Folin-Ciocalteu protocol [21]. A total of 1000 μL of ZH was added to 5 mL of Folin-Ciocalteu reagent (mixed with distilled water 1:10 v/v) and 4 mL (75 g/L) of sodium carbonate. The samples were vortexed for 15 s then left to stand for 30 min at 40 °C for staining. The absorbance was measured at a wavelength of 765 nm using the Thermo Scientific Spectrascan UV 2700 dual-beam spectrophotometer. The total phenolic content of ZH was expressed as mg/g n-propyl gallate equivalent (mg GAE/g).

2.2.2. Flavonoid Estimation

A total of 2 mL of distilled water and a NaNO2 (0.15 mL; 5%) solution were added to 0.5 mL of ZH then incubated at room temperature for 6 min. A solution of AlCl3 (0.15 mL, 1.1%) was added and allowed to stand for 6 minutes. Afterwards, 2 mL of NaOH (4%) then 5 mL of distilled water were added to the previous mixture. The mixture was allowed to stand for 15 min, and the color intensity was measured at 510 nm. The total phenolic content was expressed in mg of quercetin equivalent (QC) per gram [22].

2.2.3. Determination of Total Tannins

A total of 50 μL of ZH was diluted in 1.5 mL of 4% vanillin then added to 750 μL of concentrated HCl. After vigorous stirring, the mixture was incubated for 20 min at room temperature then the absorbance was measured at 500 nm. The total tannin content was expressed in tannic acid equivalent (TAE) [23].

2.3. Antioxidant Activity

The antioxidant activity of ZH was investigated using 3 different approaches, based on electron or hydrogen transfer reactions, to assess scavenging activity: DPPH radical scavenging activity, ABTS radical scavenging activity, and β-carotene-linoleic acid bleaching (BCB) assay [24,25]. The butylated hydroxytoluene (BHT) and ascorbic acid (AA) were used as positive antioxidant sample controls.

2.3.1. DPPH Radical Scavenging Activity

The DPPH (2,2-diphenyl-1-picryl-hydrazyl-hydrate) (Sigma-Aldrich, Milano, Italy) free radical scavenging activities of the ZH samples were determined using an antioxidant assay based on the electron transfer reaction. Several ZH dilutions were incubated with DPPH for 30 min at room temperature. The variation in the color of DPPH was assessed based on spectrophotometric analysis at 515 nm. We used the ascorbic acid as the standard control. The antioxidant activity was calculated using the following formula and expressed as IC50 (mg/mL):
[DPPH scavenging activity (%) = (A0 − A1)/A0 × 100];
where A0 denotes the absorbance of the control and A1 denotes the absorbance of the sample.

2.3.2. ABTS Radical Scavenging Activity

To assess the free radical scavenging activity of ZH, we used ABTS (2, 2-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid); Sigma-Aldrich, Milano, Italy). The ABTS+ was generated by reacting 7 mM ABTS with 2.45 mM K2S2O8 at room temperature under dark conditions. Then, serval ZH dilutions were mixed with 900 μL of the prepared ABTS+ solution and incubated for 30 min. ABTS scavenging activity was calculated using the following formula and expressed as IC50 (mg/mL):
[ABTS scavenging activity (%) = (A0 − A1)/A0 × 100];
where A0 denotes the absorbance of the control and A1 denotes the absorbance of the sample.

2.3.3. β-Carotene/Linoleic Acid Method

The β-carotene method was performed as described by Ikram et al. [26]. An emulsion containing 5 mg β-carotene, 0.30 mL linoleic acid, and 1.5 mL Tween 40 in 650 mL distilled water was prepared. A total of 25 mL from the prepared emulsion was added to 1 mL of ZH then incubated in bath water at 50 °C in the dark. Absorbance of β-carotene was assessed based on spectrophotometric analysis at 470 nm (t = 0 min) and after 2 h of incubation (t = 120 min). The AA and BHT were used as the standard for comparison. The antioxidant activity of ZH was estimated using the following equation: and expressed as IC50 (mg/mL):
[PI% = ( A-β-carotene T120/A-β-carotene t0) × 100];

2.4. MTT Assay

The anticancer activity of ZH was carried out on 3 human cancer cell lines, including lung (A549), breast (MCF-7), and colon (HCT-116) [27]. The National Centre for Cell Science (NCCS) in Pune, India, provided cell lines for use in this study. Doxorubicin (Sigma, Chennai, India) was used as a reference drug. Cells were firstly grown in (Dulbecco’s Modified Eagle Medium) DMEM (MP Biomedicals, Eschwege, Germany) and 10,000 U/mL penicillin and 5 mg/mL streptomycin antibiotic solution (Hi-Media, Mumbai, India) supplemented with 10% fetal bovine serum (FBS) (MP Biomedicals, Germany), then incubated at 37 °C, 5% CO2, and 80% humidity. After the required growth of cells was obtained, cells were treated in triplicate for 24 h with variable concentrations of ZH diluted in complete media at 2%, 4%, 6%, 8%, and 10%, then washed with phosphate buffered saline (PBS) solution. The cultured cells were treated for 4 h with the prepared MTT (3-(4,5-dimethylthiazolyl-2)-2,5 diphenyl tetrazolium bromide) (MP Biomedicals, Germany) solution (5 mg/mL). The supernatant was removed and 100 μL DMSO (Merck, Darmstadt, Germany) was added to each well. Cell viability was determined by measuring the absorbance at 570 nm (ELISA reader (EL10 A, Biobase, Jinan, China)) and the percentage of viable cells was estimated in order to determine the 50% cytotoxic concentration IC50 values for respective cancer cell lines using the following equation:
[% cell viability = (A570 of treated cells/A570 of control cells) × 100%];

2.5. LCMS Analysis

The phytochemistry of the ZH sample was analyzed using a UHPLC-PDA-Detector Mass Spectrophotometer (HR-LCMS 1290 Infinity UHPLC System), Agilent Technologies®, USA [28]. The liquid chromatographic system consisted of a HiP sampler, binary gradient solvent pump, column compartment, and quadrupole time of flight mass spectrometer (MS Q-TOF) with dual Agilent Jet Stream Electrospray (AJS ES) ion source. A total of 10 μL of the sample was injected into the system, followed by separation in SB-C18 column (2.1 × 50 mm, 1.8 μm particle size). The solvents used were 1% formic acid in deionized water (solvent A) and acetonitrile (solvent B). A flow rate of 0.50 mL/min was used, while MS detection was performed in MS Q-TOF. Compounds were identified via their mass spectra and their unique mass fragmentation patterns. Compound Discoverer 2.1, ChemSpider, and PubChem were used as the main tools for the identification of the phytochemical constituents.

2.6. ADMET Analysis

Drug ability and pharmacokinetics were predicted for the different compounds identified in honey based on the ADMET properties (absorption, distribution, metabolism, elimination, and toxicity) using online servers and established commercial packages (http://www.swissadme.ch/) and (http://biosig.unimelb.edu.au/pkcsm/prediction) (accessed on 11 March 2022) as reported in previous studies [29,30,31]. Bioavailability was also studied based on the physicochemical structures of the compounds [29].

2.7. Computational Analysis

Three different receptors (human peroxiredoxin 5 (1hd2), cyclin-dependent kinase 2 (CDK-2, 1di8), and rho-associated protein kinase 1 (ROCK 1, 3twj)) were selected. These receptors were targeted to check the potential antioxidant and anticancer effect of the identified compounds. Their crystalized structures were obtained from the RCSB protein data bank. ChemDraw was used to obtain the chemical structures when needed [30,32]. Following the pre-processing of both ligands and receptors, the binding approach was used based on the CHARMM force field as previously described using vina packages [31,33,34]. The binding scores and calculation of embedding distances and bonding networks were studied as previously described [29,30,31]. The reason behind the selection of these receptors is their involvement in antioxidant and anticancer pathways and they are commonly targeted in pharmaceutical and drug design approaches.

2.8. Statistical Analysis

For statistical analysis, a significance test was carried out among the treatments by one way ANOVA followed by Tukey’s post hoc test at p < 0.05. Statistical analysis was conducted with software GraphPad Prism 5.0.

3. Results

3.1. Phytochemical Profiling

In order to determine the chemical composition of ZH, high resolution liquid chromatography mass spectrometry (HR-LCMS) was carried out. This technique was performed in the separation and identification of the phytoconstituents based on their retention time, database difference (library), experimental m/z, MS/MS fragments, metabolite class, and proposed compounds. MS data were provided in negative and positive ionization mode. We revealed 11 compounds, including sugar acids, amino acid derivatives, alkaloids, fatty acids, and one small protein (Supplementary Materials Figure S1). The complete list is summarized in Table 1.
The chemical structures of all identified compounds are listed in Figure 1.

3.2. MTT Assay

The cytotoxicity of various concentrations of ZH were assessed after 24 h on three cancer cell lines: breast (MCF-7), lung (A549), and colon (HCT-116). Treatment with ZH significantly decreased cell viability in a dose-dependent manner (Figure 1). The IC50 values obtained for breast, colon, and lung cancer were 5.203%, 6.02%, and 7.257%, respectively, while for Doxorubicinin (used as standard) the respective IC50 were 3.5 µg/mL, 1.5 µg/mL, and 6.5 µg/mL (Figure 2 and Figure 3).

3.3. Anti-Oxidant Potential

The assessment of ZH antioxidant properties was carried out using different approaches. The antioxidant and free radical scavenging activity assessed via DPPH and ABTS methods showed nearly similar values with respective IC50 3.45 mg/mL and 3.554 mg/mL. The β-carotene-linoleic acid bleaching reveled an IC50 >5 mg/mL (Table 2). The total phenolic content (TPC), total flavonoids content (TFC), and tannins were illustrated in Table 2.

3.4. ADMET Analysis

The investigation of the ADMET properties of the identified compounds from ZH was carried out via the Swiss-model online server. Several measures of physiochemical properties that project the drug-likeness of compounds was carried out. According to Lipinski’s rule of five, a compound of reported biological activity is considered active with good absorption and permeation when the following criteria are fulfilled: the hydrogen-bond acceptors (H-bond acceptors) must be less than 10, hydrogen-bond donors (H-bond donors) less than five, rotatable bonds (RBN) must be 10 or less, a molecular weight not exceeding 500 Da, and a calculated log P (consensus log po/w) less than five. In our analysis, the in silico ADMET study revealed that most of the identified compounds meet Lipinski’s rule with zero alert as shown in Table 3, Figure 4.

3.5. Docking Analysis

Table 4 shows that all ZH compounds bound to the three targeted receptors with negative free binding energy, but with different scores. The free binding energy ranged between −4.3 and −8.7 kcal/mol. The best binding score was predicted in the 1di8/Miraxanthin-III (compound No. 8) complex. Miraxanthin-III was also found to interact with the 3twj receptor with −8.0 kcal/mol. The molecular interactions of this compound with 1DI8 included four conventional H-bonds and involved seven different residues, which include Asp145 with the lowest distance: 1.829 Å only. A network of hydrophobic bonds further supported the established H-bonds. The identified compounds in honey established one to eight H-bonds, and the highest number of H-bonds was predicted in compounds one and six with 3 twj macromolecules, and compound seven with 1HD2. The diagram of interactions (Figure 5) shows involvement of Asp86, Glu81, Ile10, Leu83, Phe80, Lys33, and Asp145. The latter interacted with a salt bridge bond, which is even stronger than an H-bond. In our study, the number of the closest interacting residues ranged between three and nine. The highest number was predicted in 3TWJ with compound six and in 1DI8 with compounds three, six, seven, nine, and 11. Despite miraxanthin-III (compound eight) interacting with seven residues of 1DI8, it was also deeply embedded in the pocket region of the receptor. In this context, all honey compounds were found to be in close vicinity of all the targeted receptor with a distance of less than three Å. In fact, it exceeded 2.5 only in 3TWJ-compound seven, with a distance equal to 2.556 Å.

4. Discussion

With the increasing rates of cancer globally and drug resistance in severe malignancies, finding new, efficient, and safe therapies is a challenge [29,30]. Natural products, such as honey, could be a valuable source of interesting bioactive compounds with prominent anticancer potential [31,33,35,36]. This activity may vary according to floral origin, geographic region, season, or altitudes [4,5]. More than 200 honey constituents have been identified, including sugars, amino acids, polypeptides, minerals, and vitamins. The biological activities of honey are not only due to their high content of phenolic and flavonoid compounds, but also to other active family of compounds, which should be explored [3,37,38].
In our investigation, profiling of ZH via the LC-MS approach reported 11 dominant phytoconstituents belonging to several families, including sugars, amino acid derivatives, alkaloids, and fatty acids. It has been previously reported that the Ziziphus genus, especially Z. jujuba, are a source of biological compounds such as alkaloids, glycosides, saponins, flavonoids, terpenoids, and phenolic compounds [39,40]. Ziziphus members are known to possess several pharmacological properties, including antioxidant, anticancer, anxiolytic, immunostimulant, antiulcer, anti-inflammatory, cardiovascular, antifertility, contraceptive, antifungal, hypoglycemic, anti-allergic, and anti-diarrheal properties [39]. Additional properties have been reported for some Ziziphus members such as the Z. lotus plant species, described to possess the ability to inhibit copper corrosion in natural seawater [41].
Among the identified sugars, isomaltulose constitutes one of the predominant natural carbohydrates found in honey [42]. This sugar is often used as an alternative sweetener with low glycemic index properties and has potential prebiotic benefits [43]. Among the identified alkaloids, anabasamine was previously reported in the seed extract of the Anabasis aphylla plant. Due to its acetylcholinesterase inhibitor effect, this alkaloid could be used for symptomatic treatment of dementia [44]. A previous study also reported that ZH could contain protein components, even cyclic peptides, with potential medicinal values [45]. These proteins can be used as a marker for floral origin [46]. In our case, we report a tripeptide Asp-Thr-Gly, which could be a specific marker for the floral origin of our investigated ZH. Among identified compounds, we noted amino acids and amino acid derivatives, such as Miraxanthin-III, N-(1-Deoxy-1- fructosyl) leucine, and N-(1-Deoxy-1- fructosyl) phenylalanine. Miraxanthin-III was previously reported in several extracts issued from medicinal plants, mainly Ardisia japonica, Ligusticum chuanxiong, Lippia nodiflora, and Mirabilis jalapa [47], and was classified as an alkaloid within the amine group. Several in silico studies revealed an interesting application for Miraxanthin-III to treat several diseases. It has been shown that this compound could be a prominent alternative for stroke treatment by interacting and activating neuroglobin [48]. Another study provided new insight to use Miraxanthin-III to treat Alzheimer disease by inhibiting proteins that are involved in pathogenesis pathways [47]. This phytocompound could also be used as an alternative treatment for cancer by inhibiting lactate dehydrogenase enzymes leading to decreased cell proliferation and increased sensitivity to chemotherapy agents [49].
Studies revealed that honey may exhibit chemopreventive and therapeutic effects against several cancer forms. Several investigations highlight that honey may display anticancer potential involving different molecular mechanisms, such as cell cycle arrest, apoptosis induction, and activation of mitochondrial pathways, with immunomodulatory and anti-inflammatory effect [3,50,51,52]. ZH has been widely investigated for its healing properties, especially against microbial infectious diseases [53]. Important antioxidant, anti-inflammatory, and antitumor activity has also been reported by several other studies, showing ZH to be an important research topic for alternative treatments of cancer [12]. Through this study, we investigate for the first time the anticancer potential of ZH derived from the Hail region against three cancer cell lines, including breast (MCF-7), lung (A549), and colon (HCT-116). We reported interesting anticancer activity with IC50 values in breast 5.203%, followed by colon 6.02%, in favor of inhibition of proliferation. The inhibition was observed in a dose-dependent manner.
Previous studies investigated the anticancer activity of ZH from Egypt against liver Hep-G2, colon Caco-2, and breast MCF-7 cell lines when treated with 100 µg/mL, and revealed a percentage of IC50 cytotoxicity of 15.94%, 34.22%, and 34.22%, respectively [54].
The in vitro study showed the chemoprotective effects of ZH by improving breast cancer treatment. They revealed that ZH modulates the expression of the two genes MMPs and TIMPs involved in cancer metastasis in triple-negative breast cancer cells (MDA-MB-231) [18]. This finding may explain the antiproliferative effect observed against the breast MCF-7 cell line in our study. Ghramh and colleagues tested three ZHs from different geographical locations in Saudi Arabia and Pakistan and their synthesized silver nanoparticles (AgNPs). They revealed a growth modulation in normal splenic cells and an antiproliferative effect on Hela cancer cell line HepG2 [55]. Moreover, another study which investigated Ziziphus jujube mill honey from China revealed an anticancer effect on HepG2 cells. It showed that this type of honey may block the cell cycle progression at the G0/G1 and induce apoptosis via molecular mechanisms, including DNA damage, p53 expression, and caspase activation [56]. The anticancer potential cannot be related only to phenolic compounds but also to the immunomodulatory effect exhibited by some glycoproteins and glycopeptides [29,57] as previously identified in ZH.
Studies revealed that normal cellular metabolism leads to the accumulation of free radicals, principally reactive oxygen and nitrogen species, that may be enhanced under stressed conditions. This accumulation could give rise to diseases and promote cancers by inducing tumorigenesis processes. Under extreme oxidative stress, the antioxidant agents produced by cells become insufficient [33,36,58]. In this instance, research may turn to the natural antioxidants derived from natural products, especially honey, to overcome oxidative stress and prevent cancers. Studies revealed that honey could be an interesting source of natural antioxidants due to its high content of polyphenols and flavonoids [58,59]. These two compounds add to other molecules, such as vitamins, and some peptides could exhibit chemoprevention activity trough potent free radical scavenging abilities [28,31,57].
In vivo and in vitro assays proved that ZH may improve considerably the oxidative status. Previous studies highlighted that ZH can display in vivo preventive effects against ethanol-induced gastric ulceration in rats by displaying antioxidant and anti-apoptotic effects [16]. Another recent study also demonstrated the gastroprotective effects of Saudi Sidr honey which exhibited high antioxidant activity against acetyl salicylic acid-induced gastric ulcers in rats [8].
An in vivo study showed that the administration of Saudi Sidr honey in rats could prevent the biochemical and histomorphological alteration induced by carbon tetrachloride CCl4 in liver and kidney tissues [60]. All these findings highlight the antioxidant potency that could display the preventative effects of ZH.
Honey content and antioxidant potential may vary according to many factors, including botanical source, region, climatic conditions, seasonal factors, entomological origin, altitude, etc. [61,62]. Thus, this study aimed to investigate the antioxidant potential of identified phytochemicals in ZH derived from the Hail region and to characterize its specificity. Since honey may contain a different group of antioxidants and may display complex oxidation processes [19,60], we used three different approaches based on the electron or hydrogen transfer reaction to assess antioxidant potential: DPPH radical scavenging activity, ABTS radical scavenging activity, and β- carotene-linoleic acid bleaching (BCB) assay. The respective antioxidant potential IC50 for these three approaches were, respectively, 3.450 mg/mL, 3.554 mg/mL, and >5 mg/mL.
In a previous study established by Alotibi et al, exploring the biological activity of several Saudi honeys, including unifloral and multiforal Sidr honey, the rate of TPC was between 1.07–1.73 mg/g and the antioxidant activities using DPPH were between 13.56% and 30%. In these studies, there is no clear correlation between activities displayed by tested honey and total phenolic compounds [63,64]. Another study on Omani honey including 10 ZH samples revealed the absence of correlation between TPC, TFC, and antioxidant activity [65]. In that study, the TFC and TPC ranged between 718–1034 mg/kg and 972–1520 mg/kg, respectively, while the antioxidant IC50 was estimated between 33.8–72.3 mg/mL using the DPPH approach [58].
In our study, the TPC was 3.396 mg/g and the TFC was 0.061 mg/g, and the obtained antioxidant activity IC50 was 3.450 mg/mL using DDPH approaches. Despite the TPC of ZH from the Hail region being lower than Omani ZH, our honey shows higher antioxidant activities. All these findings indicate that the antioxidant activity is not only due to phenolic compounds, but also to other honey constituents that could display antioxidant activity.
We investigated the drug-likeness properties of identified compounds to assess their potential use for drug development. We concluded that the majority of the identified compounds met Lipinsli’s rule. The only compounds that did not meet this rule were still associated with zero alert. Usually, the absence of toxicophores of alarming chemicals indicates the absence of DNS and/or protein damage [30,31,34]. The bioavailability scores showed that all compounds may be physiologically active. Furthermore, BAS scores showed that oral administration is computationally possible (radar figure). Gastrointestinal (GI) and blood-brain-barrier (BBB) were also assessed. Our findings suggest that half of the identified compounds (3, 8–11) have high GI absorption. Most of the compounds were also predicted to be able to penetrate the BBB. This was confirmed by the boiled-egg model (Figure 3). Only four compounds were found to behave as P-glycoprotein (P-gp) substrates. As cytochrome P450 enzymes (CYP) play a key role in drug metabolism and excretion [29,32,34], the major five CYP isoforms (CYP1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3A4) were assessed. While none of the compounds inhibited CYP2C19, CYP2D6 was predicted to be inhibited by three different compounds (three, nine, and 10). Furthermore, compounds one, two, five–eight, and 11 were predicted to inhibit none of the CYP assessed isoforms. Based on the physico-chemical properties, particularly the lipophilicity and molecular weight, skin permeability was evaluated. It ranged between −4.45 and −11.9, which indicated low to moderate permeability [30,32].
The identified compounds in honey were subjected to computational modeling to assess their molecular interactions with some key receptors related to both antioxidant and anticancer activities. Table 4 shows that all compounds bound the three targeted receptors with negative free-binding energy, but with different scores. The free-binding energy ranged between −4.3 and −8.7 kcal/mol. Variation in such score values was reported to be related to the 3D chemical structures of the ligands [30,34,66]. The best binding score was predicted in the 1di8- Miraxanthin- III (compound eight) complex. Miraxanthin-III was also found to interact with the 3twj receptor with −8.0 kcal/mol. The molecular interactions of this compound with 1di8 included four conventional H-bonds and involved seven different residues, which include Asp145 with the lowest distance: 1.829 Å only. Regardless of H-bonds that are commonly evaluated to assess the biological activities of the targeted compounds [29,31,32], a network of hydrophobic bonds was also found within the different studied complexes. This may contribute to the stability of the complexes as reported in several recent in silico studies [29,30,67,68]. Our results exhibit that all identified compounds in honey established one to eight H-bonds. The highest number of H-bonds (n = 8) was predicted in compounds one and six with 3twj macromolecules and compound seven with 1hd2. The corresponding diagram of interactions (Figure 3) showed involvement of Asp86, Glu81, Ile10, Leu83, Phe80, Lys33, and Asp145. The latter interacted with a salt bridge bond, which is even stronger than an H-bond. Interactions with key residues were found to promote biological activities, including antioxidant and anticancer potential in the studied compounds. In our study, the number of the closest interacting residues ranged between three and nine. The highest number was predicted in 3twj with compound six and in 1di8 with compounds three, six, seven, nine, and 11. As CDK-2 and ROCK 1 are involved in the control of cell cycles and are commonly targeted to counteract cancer cell proliferation [30,66], our data may confirm the potential anti-tumoral effect of ZH phytochemicals. Despite miraxanthin-III (compound eight) interacting with seven residues of 1di8, it was also deeply embedded in the pocket region of the receptor. In this context, all honey compounds were found to be in close vicinity of all the targeted receptor with a distance of less than three Å. In fact, it exceeded 2.5 only in 3twj-compound seven with a distance equal to 2.556 Å. Ligand deep embedding was reported to enhance biological activity via increasing the complex stability [30,33,68]. Taken together, these molecular interactions justify that the antioxidant and anticancer potentials of the studied honey are thermodynamically possible, and this could explain the results obtained in vitro.

5. Conclusions

The diversity and complexity of honey phytocompounds and the absence of standardization of its biological activities pose a hindrance to use honey in conventional medicine. For that, extended studies are needed to characterize honey constituents and explore their healing properties for use in alternative medicines. Through our findings, we reported interesting antioxidant activity that could be related to non-phenolic or flavonoid compounds. The ZH also showed a notable cytotoxic effect in a dose-dependent manner against three cancer cell lines in favor of anticancer potential. The computational analysis revealed that most honey compounds present good drug-likeness properties with imminent antioxidant and anticancer effects, mainly for L-Gulonate, Isomaltulose, Semilepidinoside B, and Miraxanthin-III. These findings may support the in vitro observed results. Nonetheless, further studies on the isolation, characterization, and assessment of these biological activities should be conducted to support the above-stated findings. The findings of our investigation proves that ZH could be used for therapeutic purposes to treat cancers.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/life13010035/s1, Figure S1: HR-LC/MS spectrum peak of honey showing the chromatogram intensity against the acquisition time, (A) positive analysis and (B) negative analysis.

Author Contributions

Conceptualization, M.S., N.B. and W.S.H.; methodology, A.H., R.B., M.A. (Mousa Alreshidi) and M.S.; software, R.B., R.H.L. and A.J.S.; validation, Z.S. and M.A. (Mousa Alreshidi); formal analysis, M.A. (Mohd Adnan) and M.S.; investigation, A.H.; data curation, A.H., R.B. and E.N.; writing—original draft preparation, M.S., N.B., E.N., W.S.H. and R.B.; writing—review and editing, M.A. (Mohd Adnan), M.S., N.B., E.N., W.S.H. and R.B.; visualization, M.S.; supervision, W.S.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Scientific Research Deanship at the University of Hail, Hail, Saudi Arabia. Project number RG-21 109.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

We declare no conflict of interest.

References

  1. Ferlay, J.; Colombet, M.; Soerjomataram, I.; Parkin, D.M.; Piñeros, M.; Znaor, A.; Bray, F. Cancer statistics for the year 2020: An overview. Int. J. Cancer 2021, 149, 778–789. [Google Scholar] [CrossRef] [PubMed]
  2. Dyba, T.A.; Randi, G.; Martos, C.; Giusti, F.; Calvalho, R.; Neamtiu, L.; Bettio, M. 1501O Long-term estimates of cancer incidence and mortality for the EU and EFTA countries according to different demographic scenarios. Ann. Oncol. 2021, 32, S1102. [Google Scholar] [CrossRef]
  3. Waheed, M.; Hussain, M.B.; Javed, A.; Mushtaq, Z.; Hassan, S.; Shariati, M.A.; Khan, M.U.; Majeed, M.; Nigam, M.; Mishra, A.P.; et al. Honey and cancer: A mechanistic review. Clin. Nutr. 2019, 38, 2499–2503. [Google Scholar] [CrossRef] [PubMed]
  4. Mohammed, M.E.A.; Alaergani, W.; Suleiman, M.H.; Al-Gramah, H.A. Hydrogen peroxide and dicarbonyl compounds concentration in honey samples from different botanical origins and altitudes in the South of Saudi Arabia. Curr. Res. Nutr. Food Sci. J. 2019, 7, 150–160. [Google Scholar] [CrossRef]
  5. Farooq, R.; Majid, S.; Hanif, A.; Ashraf, A.; Khan, A. Different types of honey and their properties. In Therapeutic Applications of Honey and Its Phytochemicals; Springer: Singapore, 2022; pp. 261–278. [Google Scholar]
  6. Taha, E.K.A.; Al-Kahtani, S.; Taha, R. Comparison of the physicochemical characteristics of sidr (Ziziphus spp.) honey produced by Apis florea F. and Apis mellifera L. J. Apic. Res. 2021, 60, 470–477. [Google Scholar] [CrossRef]
  7. Ali, H.; Khan, S.; Ullah, R.; Khan, B. Fluorescence fingerprints of Sidr honey in comparison with uni/polyfloral honey samples. Eur. Food Res. Technol. 2020, 246, 1829–1837. [Google Scholar] [CrossRef]
  8. Al-Ghamdi, G.; Hussein, R.H.; Al-azragi, R. The antioxidant impact of Saudi sidr honey against acetyl salicylic acid-induced gastric ulcer. Med. Sci. 2020, 24, 1923–1929. [Google Scholar]
  9. Almalki, R.A.; Alzahrani, D.A. Morphological investigation of genus Ziziphus mill.(Rhamnaceae) in saudi arabia. Am. J. Plant Sci. 2018, 9, 2644. [Google Scholar] [CrossRef] [Green Version]
  10. Mishra, V. Evaluation of the antioxidant activity of fruit extracts of indigenous medicinal plant, Zizyphus xylopyrus (Retz.) Willd. Int. J. Green Pharm. 2018, 4, 12. [Google Scholar] [CrossRef]
  11. Soliman, S.; Hamoda, A.M.; El-Shorbagi, A.N.A.; El-Keblawy, A.A. Novel betulin derivative is responsible for the anticancer folk use of Ziziphus spina-christi from the hot environmental habitat of UAE. J. Ethnopharmacol. 2019, 231, 403–408. [Google Scholar] [CrossRef]
  12. Al-Ghamdi, A.A.; Ansari, M.J. Biological and therapeutic roles of Saudi Arabian honey: A comparative review. J. King Saud Univ. -Sci. 2021, 33, 101329. [Google Scholar] [CrossRef]
  13. Zerrouk, S.; Seijo, M.C.; Escuredo, O.; Rodríguez-Flores, M.S. Characterization of Ziziphus lotus (jujube) honey produced in Algeria. J. Apic. Res. 2018, 57, 166–174. [Google Scholar] [CrossRef]
  14. Mărgăoan, R.; Topal, E.; Balkanska, R.; Yücel, B.; Oravecz, T.; Cornea-Cipcigan, M.; Vodnar, D.C. Monofloral Honeys as a Potential Source of Natural Antioxidants, Minerals and Medicine. Antioxidants. 2021, 10, 1023. [Google Scholar] [CrossRef]
  15. Sampaio, L.A.; Pina, L.T.S.; Serafini, M.R.; dos Santos Tavares, D.; Guimarães, A.G. Antitumor effects of carvacrol and thymol: A systematic review. Front. Pharmacol. 2021, 12, 702487. [Google Scholar] [CrossRef] [PubMed]
  16. Taha, M.M.E.; Abdelwahab, S.I.; Elsanousi, R.; Sheikh, B.Y.; Abdulla, M.A.; Babiker, S.E.; Mohamed, E. Effectiveness of Sidr Honey on the prevention of ethanol-induced gatroulcerogenesis: Role of antioxidant and antiapoptotic mechanism. Pharmacogn. J. 2015, 7, 157–164. [Google Scholar] [CrossRef] [Green Version]
  17. Sponghini, A.P.; Rondonotti, D.; Platini, F.; Cena, T.; Ferrante, D.; Stratica, F.; Gennari, A. A Simon’s two-stage design trial evaluating the potential role of a kind of honey in preventing chemotherapy- hematopoietic toxicities. J. Tradit. Complement. Med. 2021, 11, 466–469. [Google Scholar] [CrossRef] [PubMed]
  18. Almeer, R.; Alqarni, A.; Alqattan, S.; Abdi, S.; Alarifi, S.; Hassan, Z.; Semlali, A. Effect of honey in improving breast cancer treatment and gene expression modulation of MMPs and TIMPs in triple-negative breast cancer cells. Pak. J. Zool. 2018, 50, 1999–2007. [Google Scholar] [CrossRef]
  19. Hamadou, W.S.; Bouali, N.; Badraoui, R.; Hadj Lajimi, R.; Hamdi, A.; Alreshidi, M.; Patel, M.; Adnan, M.; Siddiqui, A.J.; Noumi, E.; et al. Chemical Composition and the Anticancer, Antimicrobial, and Antioxidant Properties of Acacia Honey from the Hail Region: The in vitro and in silico Investigation. Evid.-Based Complement. Altern. Med. 2022, 1518511. [Google Scholar] [CrossRef] [PubMed]
  20. Bagameri, L.; Giurgiu, A.I.; Baci, G.M.; Bobiş, O.; Dezmirean, D.S. Microscopical techniques used in melissopalinology for botanical origin of honey determination. Anim. Sci. 2021, 1, 64. [Google Scholar]
  21. Julkunen-Tiitto, R. Phenolic constituents in the leaves of northern willows: Methods for the analysis of certain phenolics. J. Agric. Food Chem. 1985, 33, 213–217. [Google Scholar] [CrossRef]
  22. Haddaji, F.; Papetti, A.; Noumi, E.; Colombo, R.; Deshpande, S.; Aouadi, K.; Adnan, M.; Kadri, A.; Selmi, B.; Snoussi, M. Bioactivities and in silico study of Pergularia tomentosa L. phytochemicals as potent antimicrobial agents targeting type IIA topoisomerase, TyrRS, and Sap1 virulence proteins. Environ. Sci. Pollut. Res. 2021, 28, 25349–25367. [Google Scholar] [CrossRef]
  23. Amanat, A.; Ahmed, A.; Kazmi, A.; Aziz, B. The effect of honey on radiation-induced oral mucositis in head and neck cancer patients. Indian J. Palliat. Care 2017, 23, 317. [Google Scholar] [CrossRef] [PubMed]
  24. Noumi, E.; Snoussi, M.; Anouar, E.H.; Alreshidi, M.; Veettil, V.N.; Elkahoui, S.; Adnan, M.; Patel, M.; Kadri, A.; Aouadi, K.; et al. HR-LCMS-Based metabolite profiling, antioxidant, and anticancer properties of teucrium polium L. methanolic extract: Computational and In Vitro study. Antioxidants 2020, 9, 1089. [Google Scholar] [CrossRef] [PubMed]
  25. Ikram, E.H.K.; Eng, K.H.; Jalil, A.M.M.; Ismail, A.; Idris, S.; Azlan, A.; Nazri, H.S.M.; Diton, N.A.M.; Mokhtar, R.A.M. Antioxidant capacity and total phenolic content of Malaysian underutilized fruits. J. Food Compos. Anal. 2009, 22, 388–393. [Google Scholar] [CrossRef]
  26. Adnan, M.; Siddiqui, A.J.; Hamadou, W.S.; Snoussi, M.; Badraoui, R.; Ashraf, S.A.; Patel, M. Deciphering the molecular mechanism responsible for efficiently inhibiting metastasis of human non-small cell lung and colorectal cancer cells targeting the matrix metalloproteinases by Selaginella repanda. Plants 2021, 10, 979. [Google Scholar] [CrossRef]
  27. Reddy, M.N.; Adnan, M.; Alreshidi, M.M.; Saeed, M.; Patel, M. Evaluation of anticancer, antibacterial and antioxidant properties of a medicinally treasured fern tectaria coadunata with its phytoconstituents analysis by HR-LCMS. Anti-Cancer Agents Med. Chem. 2020, 20, 1845–1856. [Google Scholar] [CrossRef] [PubMed]
  28. Badraoui, R.; Rebai, T.; Elkahoui, S.; Alreshidi, M.; Veettil, V.N.; Noumi, E.; Al-Motair, K.A.; Aouadi, K.; Kadri, A.; De Feo, V.; et al. Allium subhirsutum L. as a Potential Source of Antioxidant and Anticancer Bioactive Molecules: HR-LCMS Phytochemical Profiling, In Vitro and In Vivo Pharmacological Study. Antioxidants 2020, 9, 1003. [Google Scholar] [CrossRef]
  29. Badraoui, R.; Saeed, M.; Bouali, N.; Hamadou, W.S.; Elkahoui, S.; Alam, M.J.; Siddiqui, A.J.; Adnan, M.; Saoudi, M.; Rebai, T. Expression profiling of selected immune genes and trabecular microarchitecture in breast cancer skeletal metastases model: Effect of α–tocopherol acetate supplementation. Calcif. Tissue Int. 2022, 110, 475–488. [Google Scholar] [CrossRef]
  30. Hchicha, K.; Korb, M.; Badraoui, R.; Naïli, H. A novel sulfate-bridged binuclear copper (II) complex: Structure, optical, ADMET and in vivo approach in a murine model of bone metastasis. New J. Chem. 2021, 45, 13775–13784. [Google Scholar] [CrossRef]
  31. Zammel, N.; Jedli, O.; Rebai, T.; Hamadou, W.S.; Elkahoui, S.; Jamal, A.; Alam, J.M.; Adnan, M.; Siddiqui, A.J.; Alreshidi, M.M.; et al. Kidney injury and oxidative damage alleviation by Zingiber officinale: Pharmacokinetics and protective approach in a combined murine model of osteoporosis. 3 Biotech 2022, 12, 112. [Google Scholar] [CrossRef]
  32. Mhadhbi, N.; Issaoui, N.; Hamadou, W.S.; Jahoor, M.A.; Elhadi, A.S.; Adnan, M.; Naϊli, H.; Badraoui, R. Physico- Cjhemical Properties, Pharmacokinetics, Molecular Dcking and In-Vitro Pharmacological Study of a Cobalt(II) Complex Based on 2-Aminopyridine. ChemitrySelect 2022, 7, e202103592. [Google Scholar] [CrossRef]
  33. Akacha, A.; Badraoui, R.; Rebai, T.; Zourgui, L. Effect of Opuntia ficus indica extract on methotrexate-induced testicular injury: A biochemical, docking and histological study. J. Biomol. Struct. Dyn. 2022, 40, 4341–4351. [Google Scholar] [CrossRef]
  34. Badraoui, R.; Adnan, M.; Bardakci, F.; Alreshidi, M.M. Chloroquine and Hydroxychloroquine Interact Differently with ACE2 Domains Reported to Bind with the Coronavirus Spike Protein: Mediation by ACE2 Polymorphism. Molecules 2021, 26, 673. [Google Scholar] [CrossRef] [PubMed]
  35. Mzid, M.; Badraoui, R.; Khedir, S.B.; Sahnoun, Z.; Rebai, T. Protective effect of ethanolic extract of Urtica urens L. against the toxicity of imidacloprid on bone remodeling in rats and antioxidant activities. Biomed. Pharmacother. 2017, 91, 1022–1041. [Google Scholar] [CrossRef]
  36. Rahmouni, F.; Saoudi, M.; Amri, N.; El-Feki, A.; Rebai, T.; Badraoui, R. Protective effect of Teucrium polium on carbon tetrachloride induced genotoxicity and oxidative stress in rats. Arch. Physiol. Biochem. 2018, 124, 1–9. [Google Scholar] [CrossRef] [PubMed]
  37. Badolato, M.; Carullo, G.; Cione, E.; Aiello, F.; Caroleo, M.C. From the hive: Honey, a novel weapon against cancer. Eur. J. Med. Chem. 2017, 142, 290–299. [Google Scholar] [CrossRef]
  38. Rouamba, A.; Compaoré, M.; Kiendrebeogo, M. Molecular targets of honey bee’s products in cancer prevention and treatment. J. Herbmed Pharmacol. 2019, 8, 261–268. [Google Scholar] [CrossRef]
  39. Tripathi, S. Ziziphus jujuba: A phytopharmacological review. Int. J. Res. Dev. Pharm. Life Sci. 2014, 3, 959–966. [Google Scholar]
  40. Lu, Y.; Bao, T.; Mo, J.; Ni, J.; Chen, W. Research advances in bioactive components and health benefits of jujube (Ziziphus jujuba Mill.) fruit. J. Zhejiang Univ. Sci. B 2021, 22, 431–449. [Google Scholar] [CrossRef]
  41. Oukhrib, R.; El Issami, S.; El Ibrahimi, B.; El Mouaden, K.; Bazzi, L.; Bammou, L.; Chaouay, A.; Salghi, R.; Jodeh, S.; Hammouti, B.; et al. Ziziphus lotus as green inhibitor of copper corrosion in natural sea water. Port. Electrochim. Acta 2017, 35, 187–200. [Google Scholar] [CrossRef]
  42. Fletcher, M.T.; Hungerford, N.L.; Webber, D.; Carpinelli de Jesus, M.; Zhang, J.; Stone, I.S.; Zawawi, N. Stingless bee honey, a novel source of trehalulose: A biologically active disaccharide with health benefits. Sci. Rep. 2020, 10, 1–8. [Google Scholar] [CrossRef] [PubMed]
  43. Shyam, S.; Ramadas, A.; Chang, S.K. Isomaltulose: Recent evidence for health benefits. J. Funct. Foods 2018, 48, 173–178. [Google Scholar] [CrossRef]
  44. Lin, S.X.; Curtis, M.A.; Sperry, J. Pyridine alkaloids with activity in the central nervous system. Bioorganic Med. Chem. 2020, 28, 115820. [Google Scholar] [CrossRef]
  45. ALaerjani, W.M.A.; Abu-Melha, S.A.; Khan, K.A.; Ghramh, H.A.; Alalmie, A.Y.A.; Alshareef, R.M.H.; Mohammed, M.E.A. Presence of short and cyclic peptides in Acacia and Ziziphus honeys may potentiate their medicinal values. Open Chem. 2021, 19, 1171–1182. [Google Scholar] [CrossRef]
  46. Zhang, Y.; Wang, Y.; Zhao, H.; Zhang, G.; Peng, D.; Cao, W. Characterization of novel protein component as marker for floral origin of jujube (Ziziphus jujuba Mill.) honey. J. Agric. Food Chem. 2019, 67, 12255–12263. [Google Scholar] [CrossRef] [PubMed]
  47. He, X.; Zhao, L.; Zhong, W.; Chen, H.Y.; Shan, X.; Tang, N.; Chen, C.Y.C. Insight into potent leads for alzheimer’s disease by using several artificial intelligence algorithms. Biomed. Pharmacother. 2020, 129, 110360. [Google Scholar] [CrossRef] [PubMed]
  48. Pangestu, A.P.; Indarto, D.; Balgis. Neuroglobin activator In Silico found from Mirabilis Jalapa for stroke treatment. In AIP Conference Proceedings; AIP Publishing LLC: Melville, NY, USA, 2018; Volume 2021, No. 1; p. 070002. [Google Scholar]
  49. Kusumawati, R.; Nasrullah, A.H.; Pesik, R.N.; Indarto, D. Secondary metabolites of Mirabilis jalapa structurally inhibit Lactate Dehydrogenase A in silico: A potential cancer treatment. In IOP Conference Series: Materials Science and Engineering; IOP Publishing: Bristol, UK, 2018; Volume 333, No. 1; p. 012078. [Google Scholar]
  50. Masad, R.J.; Haneefa, S.M.; Mohamed, Y.A.; Al-Sbiei, A.; Bashir, G.; Fernandez-Cabezudo, M.J.; Al- Ramadi, B.K. The immunomodulatory effects of honey and associated flavonoids in cancer. Nutrients 2021, 13, 1269. [Google Scholar] [CrossRef]
  51. Martinotti, S.; Pellavio, G.; Patrone, M.; Laforenza, U.; Ranzato, E. Manuka honey induces apoptosis of epithelial cancer cells through aquaporin-3 and calcium signaling. Life 2020, 10, 256. [Google Scholar] [CrossRef]
  52. Eteraf-Oskouei, T.; Najafi, M. Uses of natural honey in cancer: An updated review. Adv. Pharm. Bull. 2021, 12, 248–261. [Google Scholar] [CrossRef]
  53. Owayss, A.A.; Elbanna, K.; Iqbal, J.; Abulreesh, H.H.; Organji, S.R.; Raweh, H.S.; Alqarni, A.S. In vitro antimicrobial activities of Saudi honeys originating from Ziziphus spina-christi L. and Acacia gerrardii Benth. trees. Food Sci. Nutr. 2020, 8, 390–401. [Google Scholar] [CrossRef] [Green Version]
  54. Badria, F.; Fathy, H.; Fatehe, A.; Elimam, D.; Ghazy, M. Evaluate the cytotoxic activity of honey, propolis, and bee venom from different localities in Egypt against liver, breast, and colorectal cancer. J. Apither. 2017, 2, 1–4. [Google Scholar] [CrossRef] [Green Version]
  55. Ghramh, H.A.; Ibrahim, E.H.; Kilany, M. Study of anticancer, antimicrobial, immunomodulatory, and silver nanoparticles production by Sidr honey from three different sources. Food Sci. Nutr. 2020, 8, 445–455. [Google Scholar] [CrossRef] [PubMed]
  56. Cheng, N.; Zhao, H.; Chen, S.; He, Q.; Cao, W. Jujube honey induces apoptosis in human hepatocellular carcinoma HepG2 cell via DNA damage, p53 expression, and caspase activation. J. Food Biochem. 2019, 43, e12998. [Google Scholar] [CrossRef]
  57. Mesaik, M.A.; Dastagir, N.; Uddin, N.; Rehman, K.; Azim, M.K. Characterization of immunomodulatory activities of honey glycoproteins and glycopeptides. J. Agric. Food Chem. 2015, 63, 177–184. [Google Scholar] [CrossRef]
  58. Sarv, V.; Venskutonis, P.R.; Bhat, R. The Sorbus spp.—Underutilised plants for foods and nutraceuticals: Review on polyphenolic phytochemicals and antioxidant potential. Antioxidants 2020, 9, 813. [Google Scholar] [CrossRef] [PubMed]
  59. Wang, S.; Meckling, K.A.; Marcone, M.F.; Kakuda, Y.; Tsao, R. Can phytochemical antioxidant rich foods act as anti-cancer agents? Food Res. Int. 2011, 44, 2545–2554. [Google Scholar] [CrossRef]
  60. Al-Yahya, M.; Mothana, R.; Al-Said, M.; Al-Dosari, M.; Al-Musayeib, N.; Al-Sohaibani, M.; Rafatullah, S. Attenuation of CCl4-induced oxidative stress and hepatonephrotoxicity by Saudi Sidr honey in rats. Evid.-Based Complement. Altern. Med. 2013, 2013, 569037. [Google Scholar] [CrossRef] [Green Version]
  61. Cornara, L.; Biagi, M.; Xiao, J.; Burlando, B. Therapeutic properties of bioactive compounds from different honeybee products. Front. Pharmacol. 2017, 8, 412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Suleiman, M.H.; ALaerjani, W.M.A.; Mohammed, M.E.A. Influence of altitudinal variation on the total phenolic and flavonoid content of Acacia and Ziziphus honey. Int. J. Food Prop. 2020, 23, 2077–2086. [Google Scholar] [CrossRef]
  63. Martinello, M.; Mutinelli, F. Antioxidant activity in bee products: A review. Antioxidants 2021, 10, 71. [Google Scholar] [CrossRef] [PubMed]
  64. Alotibi, I.A.; Harakeh, S.M.; Al-Mamary, M.; Mariod, A.A.; Al-Jaouni, S.K.; Al-Masaud, S.; Al- Hindi, R.R. Floral markers and biological activity of Saudi honey. Saudi J. Biol. Sci. 2018, 25, 1369–1374. [Google Scholar] [CrossRef]
  65. Al-Farsi, M.; Al-Amri, A.; Al-Hadhrami, A.; Al-Belushi, S. Color, flavonoids, phenolics and antioxidants of Omani honey. Heliyon 2018, 4, e00874. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Othman, I.M.M.; Gad-Elkareem, M.A.M.; Radwan, H.A.; Badraoui, R.; Aouadi, K.; Kadri, A.; Snoussi, M.; Kadri, A. Synthesis, Structure-Activity Relationship and in silico Studies of Novel Pyrazolothiazole and Thiazolopyridine Derivatives as Prospective Antimicrobial and Anticancer Agents. ChemistrySelect 2021, 6, 7860–7872. [Google Scholar] [CrossRef]
  67. Hamrita, B.; Noumi, B.; Papetti, A.; Badraoui, R.; Bouslama, L.; Ben Tekfa, M.-I.; Hamdi, A.; Patel, M.; Elasbali, A.M.; Adnan, M.; et al. Phytochemical analysis, antioxidant, antimicrobial, and anti-swarming properties of hibiscus sabdariffa L. calyx extracts: In Vitro and In Silico modelling approaches. Evid.-Based Complement. Altern. Med. 2022, 2022, 1252672. [Google Scholar] [CrossRef] [PubMed]
  68. Jedli, O.; Ben-Nasr, H.; Zammel, H.; Rebai, T.; Saoudi, M.; Elkahoui, S.; Jamal, A.; Siddiqui, A.J.; Sulieman, A.E.; Alreshidi, M.M.; et al. Attenuation of ovalbumin-induced inflammation and lung oxidative injury in asthmatic rats by Zingiber officinale extract: Combined in silico and in vivo study on antioxidant potential, STAT6 and TNF-α pathways. 3 Biotech 2022, 12, 191. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Chemical structure of the identified compounds from Ziziphus honey. Names of the compounds are listed in Table 1.
Figure 1. Chemical structure of the identified compounds from Ziziphus honey. Names of the compounds are listed in Table 1.
Life 13 00035 g001
Figure 2. Anticancer activity of honey against A-549, MCF-7, and HCT-116 cancer cells. Error bars indicate SDs (±standard deviation) of three independent experiments. Significance; ** p < 0.01, *** p < 0.001.
Figure 2. Anticancer activity of honey against A-549, MCF-7, and HCT-116 cancer cells. Error bars indicate SDs (±standard deviation) of three independent experiments. Significance; ** p < 0.01, *** p < 0.001.
Life 13 00035 g002
Figure 3. Impact of honey concentration on A-549, MCF-7, and HCT-116 cell line viability.
Figure 3. Impact of honey concentration on A-549, MCF-7, and HCT-116 cell line viability.
Life 13 00035 g003
Figure 4. Bioavailability radars (A) and boiled-egg model (B) of the identified compounds in honey (111).
Figure 4. Bioavailability radars (A) and boiled-egg model (B) of the identified compounds in honey (111).
Life 13 00035 g004
Figure 5. Illustration of 3D H-bonds (A), 3D hydrophobic (B) and 2D diagrams (C) on molecular interactions of the identified compounds in honey and the targeted receptors: 1hd2 (left), adi8 (middle), and 3twj (right), which exhibited the best free-binding energies (−6.6, −8.7 and −8.0 kcal/mol, respectively). The illustrations correspond to the following complexes: 1hd2-compound four, 1di8-compound eight, and 3twj-compound eight.
Figure 5. Illustration of 3D H-bonds (A), 3D hydrophobic (B) and 2D diagrams (C) on molecular interactions of the identified compounds in honey and the targeted receptors: 1hd2 (left), adi8 (middle), and 3twj (right), which exhibited the best free-binding energies (−6.6, −8.7 and −8.0 kcal/mol, respectively). The illustrations correspond to the following complexes: 1hd2-compound four, 1di8-compound eight, and 3twj-compound eight.
Life 13 00035 g005
Table 1. Phytochemical compounds identified by the HR-LCMS technique in Ziziphus honey from the Hail region.
Table 1. Phytochemical compounds identified by the HR-LCMS technique in Ziziphus honey from the Hail region.
Compound NameFamilyRT (mn)MW
(g/mol)
Chemical
Formula
[m/z]−[m/z]+
1L-GulonateSugar Acid1.08196.0576C6 H12 O7195.0503-
2N-(1-Deoxy-1-fructosyl) leucineAmino acid derivative1.238293.147C12 H23 N O7-294.1543
3AnabasamineAlkaloids1.389253.1545C16 H19 N3-276.1435
4N-(1-Deoxy-1- fructosyl) phenylalanineAmino acid derivative1.672327.1307C15 H21 N O7-328.138
5Asp-Thr-GlyTripeptide2.082291.1107C10 H17 N3 O7-292.1178
6IsomaltuloseGlucans2.503342.1158C12 H22 O11-365.104
7Semilepidinoside BGlycoside2.734366.1415C17 H22 N2 O7-367.1491
8Miraxanthin-IIIAmino acid derivative3.67330.1215C17 H18 N2 O5-331.1284
9AnatallineAlkaloid5.935239.1418C15 H17 N3-240.1492
107-hydroxy-10E,16-heptadecadien-8-ynoic acidFatty acid20.175278.1936C17 H26 O3277.1866-
1114-fluoro-myristic acidFatty Acyl27.174246.2024C14 H27 F O2291.2009
RT: Retention time (mn); MW: molecular weight (g/mol); [m/z]−: mass-to-charge ratio in negative ionization mode; [m/z]+: mass-to-charge ratio in positive ionization mode.
Table 2. Antioxidant activities of the ZH sample compared with butylated hydroxytoluene (BHT) and ascorbic acid (AA).
Table 2. Antioxidant activities of the ZH sample compared with butylated hydroxytoluene (BHT) and ascorbic acid (AA).
Ziziphus Honey(BHT)(AA)
Phytochemical Classes
Total Flavonoids Content (mg QE/g)0.061 ± 0.001 A --
Total Tannins Content (mg TAE/g)2.511 ± 0.321 B--
Total Phenols Content (mg GAE/g)3.396 ± 0.019 C--
Antioxidant tests
DPPH IC50 (mg/mL)3.450 ± 0.081 b0.023 ± 3 × 10−4a0.022 ± 5 × 10−4a
ABTS IC50 (mg/mL)3.554 ± 0.139 a0.018 ± 4 × 10−4a0.021 ± 0.001 a
β-carotene IC50 (mg/mL)5 c0.042 ± 3.5 × 10−3b0.017 ± 0.001 a
The letters (a–c) indicate a significant difference between Ziziphus honey results by using the different antioxidant tests compared with standard and drugs according to the Duncan test (p < 0.05). Capital letters (A–C) indicate a significant difference between phytochemical method screening using the Duncan test (p < 0.05).
Table 3. Drug-likeness, pharmacokinetics, and physico-chemical properties of the identified compounds in honey (111) based on absorption, distribution, metabolism, elimination, and toxicity (ADMET) properties.
Table 3. Drug-likeness, pharmacokinetics, and physico-chemical properties of the identified compounds in honey (111) based on absorption, distribution, metabolism, elimination, and toxicity (ADMET) properties.
EntryHoney Compounds
1234567891011
Properties/Lipophilicity/Drug-likeness
Molecular weight 195.15293.31253.34327.33291.26342.30366.37330.34239.32278.39246.36
Num. heavy atoms1320192320232624182017
Num. arom. heavy atoms00126001161200
Fraction Csp30.830.920.380.530.601.000.470.240.330.590.93
Num. rotatable bonds57271056621113
Num. H-bond acceptors783881186333
Num. H-bond donors56066854121
Molar Refractivity36.5968.0381.1778.0963.6068.1688.6892.3875.3283.9871.23
TPSA (Å2)141.28139.4829.02139.48179.05189.53137.29119.2237.8157.5337.30
Consensus Log Po/w−2.87−1.922.33−1.52−2.66−3.49−0.330.751.943.704.25
Lipinski’s RuleYesYesYesYesYesNoYesYesYesYesYes
Bioavailability Score0.560.550.550.550.110.170.550.560.550.850.85
PAINS0 alert0 alert0 alert0 alert0 alert0 alert0 alert0 alert0 alert0 alert0 alert
Pharmacokinetics
GI absorptionLowLowHighLowLowLowLowHighHighHighHigh
BBB permeantNoNoYesNoNoNoNoNoYesYesYes
P-gp substrateNoYesYesNoNoYesNoNoYesNoNo
CYP1A2 inhibitorNoNoNoYesNoNoNoNoNoYesNo
CYP2C19 inhibitorNoNoNoNoNoNoNoNoNoNoNo
CYP2C9 inhibitorNoNoNoNoNoNoNoNoNoYesNo
CYP2D6 inhibitorNoNoYesNoNoNoNoNoYesYesNo
CYP3A4 inhibitorNoNoYesNoNoNoNoNoNoNoNo
Log Kp (cm/s)−9.88−10.21−6.74−10.24−11.90−11.41−9.20−7.32−6.75−5.31−4.45
Table 4. Free binding energy and molecular interactions of the identified compounds in honey with the targeted receptors: 1jij, 1hd2, 1di8, and 3twj.
Table 4. Free binding energy and molecular interactions of the identified compounds in honey with the targeted receptors: 1jij, 1hd2, 1di8, and 3twj.
Target ReceptorBinding Energy (kcal/mol)Molecular Interactions
No. H-BondsNo. Closest
Interacting Residues
Closest
Interacting Residue
Distance (Å)
11HD2−5.067Val942.003
1DI8−5.066Lys331.946
3TWJ−5.788Lys2001.751
21HD2−5.277Ala421.888
1DI8−6.538Asn1322.064
3TWJ−6.356Gly851.879
31HD2−6.013Lys222.193
1DI8−8.429Gln1311.881
3TWJ−7.226Ala2342.385
41HD2−6.676Val752.064
1DI8−7.968Asn1322.001
3TWJ−7.565Lys2001.852
51HD2−5.566Val942.351
1DI8−6.364Asp1452.155
3TWJ−6.877Lys2001.889
61HD2−6.068Gly822.081
1DI8−6.655Glu121.908
3TWJ−7.089Ala2342.238
71HD2−6.387Asn212.093
1DI8−8.059His841.994
3TWJ−7.748Cys2202.556
81HD2−6.256Arg952.095
1DI8−8.747Asp1451.829
3TWJ−8.067Ala1191.943
91HD2−6.144Asp72.036
1DI8−8.339Asp1452.389
3TWJ−7.735Glu242.019
101HD2−5.145Leu962.118
1DI8−6.738Leu831.865
3TWJ−5.356Ala2341.976
111HD2−4.345Leu962.204
1DI8−6.019Lys332.151
3TWJ−5.537Thr2192.092
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

Bouali, N.; Hamadou, W.S.; Badraoui, R.; Lajimi, R.H.; Hamdi, A.; Alreshidi, M.; Adnan, M.; Soua, Z.; Siddiqui, A.J.; Noumi, E.; et al. Phytochemical Composition, Antioxidant, and Anticancer Activities of Sidr Honey: In Vitro and In Silico Computational Investigation. Life 2023, 13, 35. https://doi.org/10.3390/life13010035

AMA Style

Bouali N, Hamadou WS, Badraoui R, Lajimi RH, Hamdi A, Alreshidi M, Adnan M, Soua Z, Siddiqui AJ, Noumi E, et al. Phytochemical Composition, Antioxidant, and Anticancer Activities of Sidr Honey: In Vitro and In Silico Computational Investigation. Life. 2023; 13(1):35. https://doi.org/10.3390/life13010035

Chicago/Turabian Style

Bouali, Nouha, Walid Sabri Hamadou, Riadh Badraoui, Ramzi Hadj Lajimi, Assia Hamdi, Mousa Alreshidi, Mohd Adnan, Zohra Soua, Arif Jamal Siddiqui, Emira Noumi, and et al. 2023. "Phytochemical Composition, Antioxidant, and Anticancer Activities of Sidr Honey: In Vitro and In Silico Computational Investigation" Life 13, no. 1: 35. https://doi.org/10.3390/life13010035

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