Next Article in Journal
Herpetin Promotes Bone Marrow Mesenchymal Stem Cells to Alleviate Carbon Tetrachloride-Induced Acute Liver Injury in Mice
Next Article in Special Issue
Sea Fennel (Crithmum maritimum L.) as an Emerging Crop for the Manufacturing of Innovative Foods and Nutraceuticals
Previous Article in Journal
L-Theanine and Immunity: A Review
Previous Article in Special Issue
Armeria maritima (Mill.) Willd. Flower Hydromethanolic Extract for Cucurbitaceae Fungal Diseases Control
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Chemical Characterisation, Antidiabetic, Antibacterial, and In Silico Studies for Different Extracts of Haloxylon stocksii (Boiss.) Benth: A Promising Halophyte

1
Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Bahauddin Zakariya University, Multan 60000, Pakistan
2
Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
3
Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11495, Saudi Arabia
4
Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, MN 55454, USA
5
Department of Food Development and Food Quality, Institute of Food Science and Human Nutrition, Gottfried Wilhelm Leibniz University Hannover, Am Kleinen Felde 30, 30167 Hannover, Germany
6
Department of Food Science, Faculty of Agriculture, Zagazig University, Zagazig 44519, Egypt
7
School of Food Science and Engineering, South China University of Technology, Guangzhou 510641, China
*
Authors to whom correspondence should be addressed.
Molecules 2023, 28(9), 3847; https://doi.org/10.3390/molecules28093847
Submission received: 26 March 2023 / Revised: 27 April 2023 / Accepted: 28 April 2023 / Published: 1 May 2023

Abstract

:
The objective of the study is to evaluate the chemical characterisation, and biological and in silico potential of Haloxylon stocksii (Boiss.) Benth, an important halophyte commonly used in traditional medicine. The research focuses on the roots and aerial parts of the plant and extracts them using two solvents: methanol and dichloromethane. Chemical characterisation of the extracts was carried out using total phenolic contents quantification, GC-MS analysis, and LC-MS screening. The results exhibited that the aerial parts of the plant have significantly higher total phenolic content than the roots. The GC-MS and LC-MS analysis of the plant extracts revealed the identification of 18 bioactive compounds in each. The biological evaluation was performed using antioxidant, antibacterial, and in vitro antidiabetic assays. The results exhibited that the aerial parts of the plant have higher antioxidant and in vitro antidiabetic activity than the roots. Additionally, the aerial parts of the plant were most effective against Gram-positive bacteria. Molecular docking was done to evaluate the binding affinity (BA) of the bioactive compounds characterised by GC-MS with diabetic enzymes used in the in vitro assay. The results showed that the BA of γ-sitosterol was better than that of acarbose, which is used as a standard in the in vitro assay. Overall, this study suggests that the extract from aerial parts of H. stocksii using methanol as a solvent have better potential as a new medicinal plant and can provide a new aspect to develop more potent medications. The research findings contribute to the scientific data of the medicinal properties of Haloxylon stocksii and provide a basis for further evaluation of its potential as a natural remedy.

1. Introduction

Herbs have been the foundation of nearly all medicinal therapy since the prehistoric era until the era of synthetic drugs in the 19th century [1]. Traditional medicines are used as an alternative therapy to treat human and animal diseases in developing countries. According to WHO, 80% of the population in emerging countries relies on herbs for disease cure [2]. Furthermore, 25% of pharmacopoeial drugs are plant derivatives or semisynthetic compounds [3]. Numerous phytochemicals have been recognised from medicinal plants and scientifically validated for their therapeutic actions [4]. Recently, there has been a significant increase in attention to discovering natural antioxidants as an alternative to synthetic antioxidants for use in medicinal materials or foods that are being restricted due to their carcinogenicity [5]. Thus, screening of new plants has remained an axis of research to find the most potent and effective drug molecule against various diseases. Medicinal plants are widely accepted in various cultures and are known to provide therapeutic benefits with minimal risks of adverse effects as compared to some synthetic products [6]. Plants comprise a diverse number of free radical scavenging moieties, such as flavonoids, anthocyanins, carotenoids, dietary glutathione, vitamins, and endogenous metabolites, and these natural moieties are rich in antioxidant potential [7,8]. Halophytes have the potential as antidiabetic agents and they have been found to possess compounds that can improve insulin sensitivity and regulate blood glucose levels [9].
Many plant species and herbs have a preservative effect, revealing the presence of antioxidant and antimicrobial components in their tissues [10]. The human body generates free radicals as part of natural physiological processes, and they can also be produced by external factors such as smoking and prolonged sun exposure. These free radicals have been associated with various metabolic and degenerative disorders, including diabetes [11]. Plants with good antioxidant potential possess antidiabetic characteristics [12]; therefore, they are becoming a point of interest for researchers as the prevalence of diabetes is rising. These plants might result in a triumph in the fight against this disorder. Antidiabetic medications can manage blood sugar levels through a variety of processes. The hepatic biosynthesis of glucose is reduced, insulin receptor sensitivity is increased, insulin secretion is stimulated, and peripheral glucose absorption is raised. Antidiabetic medications can prolong digestion and intestinal carbohydrate absorption, which aids in regulating blood sugar levels after meals [13]. Recent studies suggest that by delaying digestion, α-amylase and α-glucosidase inhibitors help to regulate blood sugar levels after meals [14]. Acarbose and miglitol, two inhibitors of α-amylase and α-glucosidase, prevented the absorption of carbohydrates from the gut [15]. The importance of in silico studies in natural product chemistry lies in their ability to save time and resources in the drug discovery process, as well as to identify new potential leads for drug development. These studies can also aid in the rational design of new analogues of natural products, to improve their bioactivity, and reduce toxicity [16].
Haloxylon stocksii (Boiss.) Benth belongs to the Chenopodiaceae family and is a type of halophyte plant [17]. H. stocksii seeds show promise as a valuable crop for arid, saline lands due to their potential as a source of edible oil and medicinal properties [18]. Its habitat is in semiarid areas and is widespread in South Asia and the Middle East. It is frequently subjected to salinity, erratic drought, and high temperatures [19]. It is a paniculately branched perennial shrub that is used as animal forage [17]. Haloxylon stocksii has a range of medicinal uses, including treating burns, cuts, internal ulcers, insect stings, and urinary system problems, particularly kidney and bladder stones. Additionally, it is used to alleviate symptoms associated with arthritis, joint disorders, and paralysis of the limbs [20]. Methanolic extracts of H. stocksii possesses anti-inflammatory, analgesic, and antiulcer properties [21]. Several extracts have been found to have hypoglycaemic, hypolipidemic, and antimicrobial properties [22,23,24]. The study of the antioxidant, antibacterial and anti-inflammation potential of this plant can reveal meaningful information regarding its use for human and medicinal purposes; however, intensive screening including antidiabetic and antioxidant potential is inevitable before its medicinal use.
To the best of the authors’ knowledge, there is no comparative study available for phytochemical characterisation, antibacterial, enzymatic inhibition, and molecular docking studies on aerial parts and root extracts of Haloxylon stocksii. This study has reported the phytochemical composition, antidiabetic potential, antibacterial, and molecular docking studies for both aerial parts and root extracts of H. stocksii. The study will provide meaningful insights into the screening of the Haloxylon stocksii plant as a new medicinal plant.

2. Results

2.1. Phytochemical Composition

The phytochemical characterisation of different solvent extracts of H. stocksii was determined by their total phenolic contents (TPC), total flavonoid contents (TFC), gas chromatography-mass spectrometry (GC-MS) analysis, and liquid chromatography-electron spray ionization mass spectrometry (LC-ESI-MS2) screening.

2.1.1. Total Phenolic Contents (TPC)

The TPC was detected at the maximum level in the aerial methanolic extract and least in the roots dichloromethane extract (Table 1).

2.1.2. Total Flavonoid Contents (TFC)

The TFC was determined in the methanolic and dichloromethane extracts of both parts, i.e., in aerial and root parts (Table 1). The TFC was significantly different in all the sample extracts. The highest TFC was exhibited by methanolic aerial parts (99.19 ± 1.14 mg of quercetin equivalent per gram of extract, mg QE/g) and lowest in the roots dichloromethane extract (54.65 ± 0.65 QE/g of extract).

2.1.3. GC-MS Analysis

Due to higher phenolic contents and biological activities, methanolic extracts of aerial parts and roots were subjected to GC-MS for screening of secondary metabolites. This chemical characterisation resulted in the identification of 18 bioactive phytochemicals (Table 2). Some of these bioactive compounds are furfural; 4H-pyran-4-one, 2,3-dihydro-3,5-dihydroxy-6-methyl-; 2-furancarboxaldehyde; 5-(hydroxymethyl)-; 2-furancarboxaldehyde; 5-(hydroxymethyl)-; 1,6-anhydro-β-ᴅ-glucopyranose; phthalic acid; bis(7-methyloctyl) ester; di-n-octyl phthalate; phenol; 2,2′-methylenebis[6-(1, 1-dimethylethyl)-4-(1-methylpropyl); and γ-sitosterol. These compounds belong to the different classes, such as furans, aldehyde, pyrones, anhydrohexose, fatty acid ester.

2.1.4. Phytochemical Screening of Haloxylon stocksii by LC-ESI-MS2

The methanolic extract of aerial parts was subjected to LC-ESI-MS2 in positive mode (Table 3, and Figures S1–S23 in the Supplementary Materials) due to its maximum total phenolic and flavonoid contents compared to other extracts. In this screening, 23 compounds were detected. Among these compounds, 18 bioactive secondary metabolites were identified with the Discoverer 3.3, and 5 were not identified with any compound library. The available toxicity profile of identified compounds is discussed in Table S1.

2.2. Biological Activities of Aerial and Roots Extract of H. stocksii

2.2.1. Antioxidant Activities

The antioxidant activity of different sample extracts of H. stocksii was evaluated by DPPH, ABTS, CUPRAC and FRAP assays (Table 4). The antioxidant results of the extracts showed that aerial parts methanolic extract have maximum antioxidant activity as compared to other extracts (aerial part dichloromethane, roots methanolic, and roots dichloromethane extracts). This should be due to the highest TPC and TFC in the aerial parts methanolic extract as compared to its counterpart extract.

2.2.2. Antibacterial Activity

In the current study, the antibacterial activity of the Haloxylon stocksii plant was determined by measuring its antibacterial activity against pathogenic bacteria. These bacteria include Bacillus subtilis, Bacillus pumilus, Pseudomonas aeruginosa, Staphylococcus epidermidis, Escherichia coli, Bordetella bronchispetica, and Micrococcus luteus, which are the main pathogenic and spoilage organisms. The results of the antibacterial activity of various extracts of Haloxylon stocksii are shown in Table 5.

2.2.3. Antidiabetic Activities

The potential of H. stocksii to alleviate diabetes was investigated in vitro by analysing its ability to inhibit two key enzymes involved in the disease, namely α-amylase and α-glucosidase. Both parts (aerial and roots) of the plant were found to exhibit significant inhibitory activity against these enzymes, indicating their potential as antidiabetic agents. Figure 1 and Table S2 shows the antidiabetic potential of this plant.

2.3. In silico Evaluation

In silico evaluation was done by performing molecular docking, ADME (Absorption, Distribution, Metabolism and Excretion), and toxicological studies.

2.3.1. Molecular Docking (MD) for α-Amylase

MD of GC-MS identified molecules was carried out for α-amylase receptors. In order to find out docking score, binding affinity, and interaction of different compounds at the active sites of α-amylase receptors. Amongst those, the interaction with best docked molecules in terms of binding affinity are represented in Table 4. The docking score for phenol, 2,2′-methylenebis[6-(1, 1-dimethylethyl)-4-(1-methylpropyl) and γ-sitosterol with α-Amylase receptors was (−8.5) and (−8.7), respectively. These findings reveal the potent role of Haloxylon stocksii in α-amylase inhibition. The details of binding affinities and interactive forces of ligands with receptors are given in Table 6 and Figure 2.

2.3.2. Molecular Docking (MD) for α-Glucosidase

MD was done with α-glucosidase receptors to find out the binding score and binding interactions at active sites. For MD, molecules tentatively identified by GC-MS were selected. The binding scores for phenol, 2,2′-methylenebis[6-(1, 1-dimethylethyl)-4-(1-methylpropyl), and γ-sitosterol were −8.1 and −8.9, respectively. This reveals the key role of Haloxylon stocksii in α-glucosidase inhibition. The details of binding interactions, binding forces, and docking scores are given in Table 7 and Figure 3.

2.3.3. ADMET Study

The SwissADME online tool was utilised to investigate the best-docked compounds in addition to the standard drug acarbose. This tool offers valuable insights into the pharmacokinetics, drug-likeness, and physicochemical properties of the compounds (Table 8). The compounds octadecanoic acid methyl ester; 4H-pyran-4-one; 2,3- dihydro-3,5-dihydroxy-6- methyl-; ethyl oleate; 1,6-anhydro-β-ᴅ-glucopyranose; and di-n-octyl phthalate have high gastrointestinal absorption. Eicosanoic acid methyl ester; phthalic acid; bis(7-methyloctyl) ester; phenol; 2,2′-methylenebis[6-(1,1-dimethylethyl)-4-(1- methylpropyl); γ-sitosterol; and acarbose have low gastrointestinal absorption. All the compounds that cannot cross blood–brain barrier have the advantage of lacking CNS adverse effects. 1,6-Anhydro-β-ᴅ-glucopyranose and the standard drug acarbose increase serum concentrations by inhibiting Pgp inhibitors. Phthalic acid, bis(7-methyloctyl) ester, and di-n-octyl phthalate can interact with the metabolism of drugs which involve cytochrome P450 3A4.

2.3.4. Lipinski Rule of Five

Lipinski’s rule of five is a rule that evaluates the chemical compounds having pharmacological or biological properties that may act as orally active drugs in human beings. The definition of the Lipinski rule is that the compounds should follow the following criteria with only exception allowed of the following criterion:
  • The hydrogen bond donors should not be more than 5.
  • The hydrogen bond acceptors should not be more than 10.
  • The molecular mass should not be more than 500 daltons.
  • Lipophilicity should not exceed the value of 5.
In silico study of best-docked compounds disclosed that all the compounds have <5 hydrogen bond donors and <10 hydrogen bond acceptors. The molecular mass of all chemical compounds evaluated is less than 500 daltons. The lipophilicity value of all the observed compounds follow the rule except eicosanoic acid, methyl ester and phthalic acid, bis(7-methyloctyl) ester (Table 9 and Figure 4). So, with these observations, all the compounds can be used as oral drugs.

2.3.5. Toxicity Study

During the in silico study, the toxicity profile of chemical compounds was evaluated by determining LD50, toxicity class, hepatotoxicity, carcinogenicity, immunotoxicity, mutagenicity, and cytotoxicity (Table 10). The toxicity findings were that all the compounds have ≥ 4 toxicity levels. This toxicity level indicates that all the compounds are slightly toxic or practically non-toxic. All the compounds except standard acarbose do not cause hepatotoxicity. Three compounds, namely phthalic acid, bis(7-methyloctyl) ester, and di-n-octyl phthalate, exhibited properties of carcinogenicity. γ-Sitosterol and acarbose displayed immunotoxicity. Only 4H-pyran-4-one, 2,3-dihydro-3,5-dihydroxy-6-methyl- exhibited mutagenicity and no chemical compound showed cytotoxicity.

3. Discussion

Screening of plants used for medicinal purposes has remained one of the most valuable approaches towards the development and provision of new drug candidates. The findings disclosed that there was significant difference (p < 0.05) among the extraction methods as well as the different plant parts, i.e., in aerial parts and roots. Briefly, it was found that AMHS samples had significantly (p < 0.05) high TPC and TFC compared to RMHS, which indicates that the methanol extract of aerial parts has higher TPC and TFC compared to that of the roots. Our results regarding higher TPC and TFC in aerial parts are in line with the literature that also revealed that phenolic compounds were significantly higher in aerial parts as compared to roots [45]. The difference in TPC between the roots and aerial parts could be due the presence of carotenoids in the aerial parts that are missing in the roots. Further, the effectiveness of different extraction methods was evaluated. It was observed that TPC and TFC were higher in the methanol extraction method compared to the dichloromethane extraction method. In addition, the higher TPC and TFC levels in the aerial parts indicate the presence of polyphenolic secondary metabolites, which may be a result of the environmental stress factors that the aerial parts face. This could be the reason that aerial parts face more stress against the environmental factors, which produce more antioxidant compounds as compared to roots, agreeing well with previous studies [46,47]. Peerzada, Khan et al. determined the TPC and TFC (262.6 ± 7.49 mg GAE/g and 79.86 ± 6.02 mg QE/g, respectively) in methanolic extract of the whole plant [48]. These high values may be due to difference in habitat of plant or due to synergistic effect of both aerial parts and roots.
GC-MS analysis is a prevailing analytical technique used in the identification of the components present in plant extracts [49]. This technique is particularly useful in the field of natural product chemistry, where plant extracts are commonly used as a source of bioactive secondary metabolites [50]. One of the main advantages of GC-MS analysis of plant extracts is its high sensitivity and specificity. The technique can detect and identify components present in trace amounts, allowing for the detection of compounds that may have important biological activities even at low concentrations [51]. Additionally, mass spectrometry is used for the characterisation of the molecular weight and structure of the identified components, which is critical for understanding their biological activities and potential therapeutic applications.
Liquid chromatography-mass spectrometry (LC-MS) profiling of plant extracts is an effective analytical approach for identifying the numerous chemical components found in plant extracts. It gives a thorough breakdown of the extract’s phytochemical makeup, including flavonoids, alkaloids, terpenoids, and other bioactive substances. The identification of unidentified chemicals in the extract via LC-MS analysis can also aid in the discovery of novel natural products with potential medicinal uses. In general, LC-MS profiling of plant extract is a useful tool for characterising chemicals obtained from plants, and it can help create new medications and functional food ingredients. The LC-MS screening resulted in the identification of amines, aromatic ketones, phenyl amines, flavonoids, alkaloids, organophosphate esters, fatty acids, and glycerolipids.
Antioxidant properties are of great importance as they determine the suitability and applicability of these plants for medicinal use. Researchers are seeking new plants to assess their effectiveness, as they believe that these are the best alternatives, with the fewest side effects, to medicines. Plant extracts have been recognized as a rich source of antioxidants, which have played a critical role in the prevention of various illnesses, including cancer, cardiovascular diseases, diabetes, and neurodegenerative disorders [52]. The antioxidant property of plant extracts is primarily due to their content of polyphenolic compounds, likely flavonoids, phenolics, and tannins [53]. Numerous studies have reported the antioxidant properties of plant extracts from various sources, including roots, vegetables, and aerial parts [54,55]. Overall, plant extracts are a promising source of antioxidants that could have potential health benefits, although further studies are needed to better understand their bioavailability and potential therapeutic applications. A study conducted by Yaseen et al. (2020) investigated the total antioxidant activity of the acetone, chloroform, acetic acid and propranolol extracts of Haloxylon stocksii [56]. The literature review of some bioactive phytoconstituents, identified with GC-MS and LC-MS, possess antioxidant activities. These bioactive secondary metabolites include triacontanoic acid [57], octadecanoic acid [44], γ-sitosterol [58], and fraxetin [59]. There is no antioxidant comparative study of the aerial and roots part of H. stocksii available in literature by utilising methanol and dichloromethane as extraction solvents. This comparative study is the first one that reports the antioxidant potential of H. stocksii for different parts and with different extraction methods.
The antibacterial potential of plants has great importance regarding their use in medicine as this property mainly involves the reduction of pathogenic microbes [60]. The results of the current study exhibited that aerial parts methanolic extract (AMHS) is more active to Gram-positive bacteria, and roots methanolic extract (RMHS) is more active against Gram-negative bacterial strains. These antibacterial activities may be due to the presence of several compounds which have antimicrobial activity. These compounds include 2-furancarboxaldehyde; 5-(hydroxymethyl)- [28]; 1,6-anhydro-β-ᴅ-glucopyranose [29]; hexadecanoic acid; methyl ester; 9-octadecenoic acid; methyl ester [52]; heptadecanoic acid; 16-methyl ester [35]; phthalic acid; bis(7-methyloctyl) ester [41]; hordenine [61]; fraxetin [62]; piperine [63]; stearamide [64]; and erucamide [65]. Wahab et al. reported the antibacterial activity against some Gram-positive and Gram-negative bacterial strains. Their results revealed that the methanolic extract of the whole plant showed good antibacterial activity against tested strains and was maximum against S. typhi [66]. Anabasis aretioides, a member of Chenopodiaceae, and showed the best activity against Staphylococcus aureus [67].
Type 2 diabetes (T2D) is a prevalent health condition worldwide that results in high blood sugar levels and abnormal carbohydrate metabolism. This disease is a significant contributor to illness and death, and it also places a substantial economic burden on societies [68]. The H. stocksii extracts were evaluated in vitro for antidiabetic activity (α-amylase and α-glucosidase inhibition). The aerial parts extracts were more effective compared to that of the roots. When we compare the two solvents, the methanol extract was more effective as compared to the dichloromethane extract. The higher antidiabetic potential of the methanolic extraction method could be due to the higher TPC of this method. Truong et al. demonstrated that the methanolic extraction method gives maximum extraction yield, highest phenolics and flavonoid contents, and antioxidant activity as compared with ethanol, chloroform, acetone, and dichloromethane extraction methods [69]. In addition, higher polyphenols are found to be effective in diabetes management. The antidiabetic potential of this plant is attributed due to the presence of some bioactive phytocompounds which were identified with GC-MS and LC-MS, such as β-sitosterol [51]; phenol; 2,2′-methylene bis[6-(1,1-dimethylethyl)-4-(1-methylpropyl) [55]; hordenine [70]; and piperine [63]. The compounds having good antioxidant activities are also responsible for antidiabetic activity [56]. The aqueous extract of Chenopodium botrys exhibited α-amylase and α-glucosidase inhibition activity [71]. Atriplex halimus L. significantly increases body weight and decreases blood glucose and hepatic levels in the rats [72]. The results of in vitro antidiabetic activities of plants endorse that oral administration of these extracts, especially aerial parts extracts in non-insulin-dependent diabetes, can reasonably reduce postprandial blood glucose levels; however, future studies, especially in vivo studies, are necessary to confirm their effectiveness.
In silico studies of Haloxylon stocksii have been gaining attention due to their cost-effectiveness and ability to provide insights into the potential biological activities of the plant’s bioactive compounds. Several studies have used computational approaches such as molecular docking, ADME, and toxicological studies to predict the binding affinity and activity of Haloxylon stocksii’s bioactive compounds with various therapeutic targets [55]. In silico studies have identified several compounds with potential antidiabetic, anti-inflammatory, and anticancer activities, such as stigmasterol and β-sitosterol. These studies have also helped to elucidate the mechanism of action of the plant’s bioactive compounds and have guided further experimental studies [73]. Therefore, future studies should aim to combine in silico and in vitro/in vivo experiments to further validate the potential therapeutic activities of Haloxylon stocksii’s bioactive compounds.
Overall, these studies suggest that Haloxylon stocksii could be a potential source of bioactive compounds, natural antioxidants, antibacterial, and antidiabetic and that further research is needed to better understand the bioactive compounds present in the plant extract and their potential therapeutic applications.

4. Materials and Methods

4.1. Plant Identification and Sample Collection

The roots and aerial parts of the plant were collected from sandy cliffs of Tehsil Choti Zarieen District Dera Ghazi Khan in August 2020 Punjab, Pakistan. The plant was identified as Haloxylon stocksii by Zafar Ullah Zafar, Institute of Pure and Applied Biology, Bahauddin Zakariya University Multan, Pakistan (BZU). Voucher no. http://www.theplantlist.org/tpl1.1/record/tro-50307719 (accessed on the 20 May 2020) was given to that plant.

4.2. Extract Preparation of the Plant Material

Firstly, the plant was washed, and shade-dried in two separate portions (i.e., aerial parts and roots) for 6 weeks at 25 °C. Both the plant parts were pulverized roughly to a coarse powder and the extraction of both powdered materials was done by simple maceration procedure separately in dichloromethane (DCM) and methanol (the ratio between powder and solvent was 1:3). Then, filtration was done after 24 h. This procedure was repeated four times with both these solvents. DCM and methanol extracts were dried under reduced pressure in a rotary evaporator (Buchi, Flawil, Switzerland). The DCM extract of roots (50.0 g) and aerial parts (30.8 g) were collected in sample bottles and assigned codes RDHS and ADHS, respectively. Methanol resulted in the yield of roots extract (70.4 g) and aerial parts extract (52.0 g). These dried extracts were collected in the sample bottles and assigned codes RMHS and AMHS, respectively.

4.3. Phytochemical Composition

4.3.1. Estimation of Total Phenolic Content

The determination of total phenolic contents (TPC) and total flavonoid contents (TFC) were done using the Folin–Ciocalteu (FC) and aluminium trichloride colorimetric methods, respectively [44].

4.3.2. GC-MS Analysis

GC-MS screening was done with an Agilent 6890 series and Hewlett Packard 5973 ground sensor. The HP-5MS column (Santa Clara, CA, USA) with 30 m length × 250 µm diameter × 0.25 µm film thickness was used. The temperature of the injection was set at 220 °C up to 240 °C, while the temperature of the oven was programmed from 60 °C to 246 °C at a rate of 3 °C/min. Pure He (helium) gas was used as a carrier. A volume of 1.0 μL of the reconstituted sample extract was injected. The temperature was set between 50 °C and 150 °C at a rate of 3 °C/min, and then raised to 300 °C at a rate of 10 °C/min. The identification of bioactive compounds was performed by tentatively identifying peaks using the NIST 2011 library scanning ranging from 35 to 600 m/z [74].

4.3.3. Phytochemical Screening of Haloxylon stocksii by LC-ESI-MS2

The aerial parts methanolic extract of H. stocksii was subjected to LC-ESI-MS2 analysis for qualitative determination of the secondary metabolites. The sample extract was dissolved in methanol of LC-MS grade. This solution was filtered and then injected by direct syringe method at the flow rate of 5 µL/min. The system consists of a column with dimensions 250 × 2.0 mm ODS-VP C18, 5 µm. The mobile phase used in liquid chromatography comprises the solvent A (0.1 % formic acid) and solvent B (methanol with 0.1 % formic acid). Gradient method of elution with 5 % B (0–5 min), 5–45 % B (90–100 min), 45–5% B (100–101 min) was then used. Afterwards, it was evaluated by Tandem Mass Spectrometry and LTQ XL linear Ion Trap Mass Spectrophotometer having an Electron spray ionization interface. The compounds were identified with Compound Discoverer 3.3. The sample was scanned at positive mode and the scan range of mass was m/z 50–2000. The voltage used was 4.8 KV, and capillary voltage used was 23 V during the analysis. The temperature of the capillary was 350 °C during the scan.

4.4. Biological Profiling

4.4.1. Antioxidant Activities

The roots and aerial parts of the Haloxylon stocksii plant were evaluated for antioxidant potential through different antioxidant assays which include the determination of radical scavenging activity and reducing power. Trolox was used as standard, and results were expressed as milligrams of Trolox equivalents per gram of dry extract (mg TE/g extract). Control was prepared by the same procedure without adding plant extract.

Radical Scavenging Activity

Aerial parts and roots of the plant were analysed for the evaluation of scavenging potential and were evaluated using 1,1-diphenyl-2-picrylhydrazyl (DPPH) and 2,2-azinobis 3-ethylbenzothiazoline-6-sulfonic acid (ABTS) assays. The procedures for above mentioned assays were in line with the literature [75].
  • DPPH Assay
For this radical scavenging assay, the volume of 50 µL stock solutions of the extracts of both parts of the plants were mixed with 200 µL of DPPH solution (0.267 mM). After, these mixtures were incubated in a 96-well microtiter plate at 25 °C for 30 min (in darkness). The absorbance of samples was measured by a microplate reader (BioTek Synergy HT, Winooski, Vermont, USA) at wavelength 517 nm. The blank solution was also assessed according to this procedure.
2.
ABTS Assay
For this reducing power assay, stock solutions of the extracts of both parts of the plants were prepared and diluted with ABTS+ solution until absorbance reached 0.700 ± 0.02 at 734 nm. The volume of 100 µL of extract solutions were mixed with 200 µL of ABTS+ solution in 96-well microplate. This mixture was incubated at 25 °C for 30 min. After this, absorbance of both solutions was measured at wavelength 734 nm. The control solution was also assessed according to this procedure.

Reducing Power Assays

Both parts of the H. stocksii plant were assessed for their reducing capacity by utilising direct reducing antioxidant Cupric Reducing Antioxidant Capacity (CUPRAC) and Ferric Reducing Antioxidant Power (FRAP). Both mentioned assays were performed according to the procedures mentioned in the literature [76].
  • CUPRAC Assay
For performing the CUPRAC assay, stock solutions of the extracts of both parts of the plant were prepared and added to reaction mixture containing neocuproine (200 µL, 7.5 mM), cupric chloride (200 µL, 10 mM), and ammonium acetate buffer (200 µL, 1 M, pH 7.0)]. After this, both solutions were incubated at 25 °C for 30 min, and absorbance was measured at 450 nm. The control solution was also assessed according to this procedure.
2.
FRAP Assay
For performing the FRAP assay, stock solutions of the extracts of both parts of the plants were prepared and added to acetate buffer in 40 mM HCl and ferric chloride (20 mM) in a final concentration with the ratio of 10:1:1 (v/v/v). Absorbance was measured at 593 nm after the incubation for 30 min at 25 °C. Furthermore, a blank solution was also assessed according to this procedure.

4.4.2. Antibacterial Activities

Antibacterial activities of the Haloxylon stocksii plant aerial parts and roots were assessed against seven bacterial stains. Three Gram-negative stains (Pseudomonas aeruginosa ATCC 9027, Escherichia coli ATCC 25922, and Bordetella bronchiseptica ATCC 7319) and four Gram-positive strains (Bacillus subtilis ATCC 1692, Bacillus pumilus ATCC 13835, Staphylococcus epidermidis ATCC 8724, and Micrococcus luteus ATCC 4925) of bacteria. These strains were obtained from Drug Testing Laboratory, Lahore (DTL). Co-amoxiclav was used as standard in this assay. The 24 h old cultures of all seven strains were taken, prepared inoculum by taking few colonies of each bacterium and adding them in test tubes containing broth medium. Tubes were incubated overnight, and colonies were diluted to a cell density of 106 CFU/mL (colony forming unit).

Disc Diffusion Method

This method was used to estimate the antibacterial activity of common infection-causing bacterial strains from the aerial parts and roots of the Haloxylon stocksii plant. The results were measured as zones of inhibition. Solutions were prepared in 10% of DMSO. These solutions were sterilised by filtering with a sterile membrane filter. After this Petri dishes with Mueller Hinton agar were prepared and inoculated with the seven bacterial strains. Positive and negative control discs were made using 10% DMSO and co-amoxiclav (10 µg/disc), respectively. These Petri dishes were incubated at 37 °C for 24 h. Triplicate experiments were performed to minimize the error. The zone of inhibition was measured in millimetres (mm), and greater than 6 mm was considered for antibacterial activity [52].

4.4.3. In vitro Antidiabetic Potential

For the estimation of the in vitro antidiabetic potential in aerial parts and root extracts of H. stocksii, α-amylase and α-glucosidase inhibition assays were performed.
  • α-Amylase inhibitory assay:
The α-amylase inhibition assay of methanolic and DCM extracts of aerial parts and roots was performed according to the method described in the literature with minor changes [73]. The volume of 0.5 mL extracts of both parts was mixed with 0.5 mL of α-amylase solutions with 0.02 M of sodium phosphate buffer (pH 6.9) this mixture was kept for incubation at 25 °C for 10 min, and then the reaction was terminated by adding 1 mL of the colouring agent dinitrosalicylic acid. After this, these test tubes were heated in a warm bath at 100 °C for 5 min. After cooling the reaction mixture till 25 °C, it was diluted with 10 mL of deionized water, and the absorbance of blank and control samples was measured at 540 nm. Acarbose was used as a standard drug. The following equation measured the inhibition of α-amylase.
%   i n h i b i t i o n   o f   α a m y l a s e = ( A b s C o n t r o l A b s S a m p l e ) / ( A b s C o n t r o l ) × 100
  • α-Glucosidase inhibitory assay:
For determination of the antidiabetic potential crude methanolic and DCM extracts of both aerial and root, parts were also analysed using α-glucosidase inhibitory assay [13]. For this analysis, 1 mL of each extracted part was mixed with 100 µL of phosphate buffer of pH 5.9. Then, 50 µL of enzyme α-glucosidase was added and kept at 37 °C for 10 min for incubation. The volume of 50 µL of substrate, i.e., p-nitrophenyl-α-ᴅ-glyco-pyranoside (pNPG) was added to the reaction mixture and the mixture was kept for incubation for a further 30 min at 35 °C. After complete incubation, the absorbance of all samples was measured at 405 nm. The % inhibition was measured by the following formula.
%   i n h a b i t i o n   o f   α g l u c o c i d a s e = 1 ( A b s S a m p l e A b s C o n t r o l ) × 100

4.5. In-Silico Evaluation

In silico evaluation was performed by molecular docking, ADME, and toxicity studies.

4.5.1. Molecular Docking Study

Molecular docking plays a key role in computer-aided drug designs and the development of molecular biology. For molecular docking in the current study, various software tools were employed, such as Autodock Vina, MGL Tools, PyRx, Babel and Discovery Studio. The enzymes α-amylase (PDB DOI: 10.2210/pdb1SMD/pdb, accessed on 7 August 2022) and α-glucosidase (PDB DOI: 10.2210/pdb5ZCB/pdb, accessed on 7 August 2022) were downloaded from Protein Data Bank [77]. The enzymes were prepared by Discovery Studio (Discovery Studio 2021 client). The GC-MS identified compounds along with acarbose (standard drug) were used as ligands and the SDF format of these ligands was downloaded from PubChem. Open Babel was used for the preparation of Ligand. Both prepared receptor and ligands were loaded to vina, embedded in PyRx. These were placed in the active area of the enzyme and the evaluation of outcomes was carried out by using Discovery Studio Visualizer [10].

4.5.2. ADMET Analysis

The ADMET analysis of the compounds were performed with online SwissADME database (http://www.swissadme.ch/, accessed on 15 October 2022) [78].

4.5.3. Toxicity Evaluation

The toxicity of in silico studied compounds were performed by the online tool PROTOX II (https://tox-new.charite.de/protox_II/, accessed on 13 October 2022) [73].

5. Conclusions

The exploration of new plants for drug development is a promising approach in the search for more effective therapies. In this study, Haloxylon stocksii was evaluated for its potential in various parameters including polyphenol contents, antioxidant, antibacterial, and antidiabetic properties. The antidiabetic potential of methanolic extracts was better than that of dichloromethane extracts. The results demonstrate that aerial parts extracts are more effective against-Gram-positive bacteria and root extracts showed more activity against Gram-negative bacteria. These findings suggest that H. stocksii may have significant potential for drug development, particularly in the treatment of infections and metabolic diseases; however, further investigation, including in vitro and in vivo cytotoxicity assays to evaluate the safety of the identified compounds is necessary to fully validate these findings before considering H. stocksii as a viable candidate for drug development. Nonetheless, these results may represent a promising milestone in the medical industry’s search for novel and effective therapies.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/molecules28093847/s1, Figure S1: Retention time peak, fragmentation pattern and chemical structure of N-Methyltyramine; Figure S2: Retention time peak, fragmentation pattern and chemical structure of Acetophenone; Figure S3: Retention time peak, fragmentation pattern and chemical structure of Hordenine; Figure S4: Retention time peak, fragmentation pattern and chemical structure of Ethyl 2-(3-{[4-(4-methylphenyl)-1,3-thiazol-2-yl]methyl}-4-oxo-3,4-dihydrophthalazin-1-yl)acetate; Figure S5: Retention time peak, fragmentation pattern and chemical structure of Fraxetin; Figure S6: Retention time peak, fragmentation pattern and chemical structure of 6,8-dihydroxy-7-methoxy-3-methyl-1H-isochromen-1-one; Figure S7: Retention time peak, and fragmentation pattern of Unknown compound (not identified with library); Figure S8: Retention time peak, fragmentation pattern and chemical structure of nor -3-methylfentanyl; Figure S9: Retention time peak, fragmentation pattern and chemical structure of Moupinamide; Figure S10: Retention time peak, fragmentation pattern and chemical structure of 2-(2,6-dimethoxyphenyl)-5,6-dimethoxy-4H-chromen-4-one; Figure S11: Retention time peak, fragmentation pattern and chemical structure of Piperine: Figure S12: Retention time peak, fragmentation pattern and chemical structure of tris(2-Butoxyethyl) phosphate; Figure S13: Retention time peak, fragmentation pattern and chemical structure of Acetyl tributyl citrate; Figure S14: Retention time peak, and fragmentation pattern of Unknown compound (not identified with library); Figure S15: Retention time peak, and fragmentation pattern of Unknown compound (not identified with library); Figure S16: Retention time peak, fragmentation pattern and chemical structure of Hexadecanamide; Figure S17: Retention time peak, fragmentation pattern and chemical structure of Unknown compound (not identified with library); Figure S18: Retention time peak, fragmentation pattern and chemical structure of 1-Palmitoylglycerol; Figure S19: Retention time peak, fragmentation pattern and chemical structure of (1S)-Tricyclo[7.3.1.0~2,7~]tridec-2(7)- en-13-one; Figure S20: Retention time peak, fragmentation pattern and chemical structure of Stearamide; Figure S21: Retention time peak, fragmentation pattern and chemical structure of Erucamide; Figure S22: Retention time peak, fragmentation pattern and chemical structure of unknown compound (not identified with library); Figure S23: Retention time peak, fragmentation pattern and chemical structure of N,N-Dimethylaniline. Table S1: Safety Profile of LC-MS identified compound; Table S2: α-amylase inhibition and α-glucosidase inhibition of Haloxylon stocksii.

Author Contributions

Conceptualization, H.Y.A. and S.A.; methodology, B.A.G. and H.R.; software, H.R.; validation, M.N.S.; formal analysis, S.A.K. and T.E.; investigation, S.A.K. and T.E.; resources, S.A.K., T.E. and H.Y.A.; data curation, S.A.K. and T.E.; writing—original draft preparation, S.N.R.R. and B.A.G.; writing—review and editing, D.A.K., S.A.K., T.E., H.R. and K.-u.-R.K.; visualization, K.-u.-R.K., S.A.K. and T.E.; supervision, S.A.; project administration, K.-u.-R.K., S.A.K. and T.E.; funding acquisition, H.Y.A., S.A.K. and T.E. All authors have read and agreed to the published version of the manuscript.

Funding

The authors are grateful to the King Saud University, Riyadh, Saudi Arabia for funding this study through project number RSP2023R504.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors acknowledge the King Saud University, Riyadh, Saudi Arabia for supporting this study through project number RSP2023R504.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Samples of the compounds are not available from the authors.

References

  1. Williamson, E.M.; Liu, X.; Izzo, A.A. Trends in use, pharmacology, and clinical applications of emerging herbal nutraceuticals. Br. J. Pharmacol. 2020, 177, 1227–1240. [Google Scholar] [CrossRef]
  2. Bhardwaj, S.; Verma, R.; Gupta, J. Challenges and future prospects of herbal medicine. Int. Res. Med. Heal Sci. 2018, 1, 12–15. [Google Scholar] [CrossRef]
  3. Robina, S.H.; Ahmad, J.; Javed, S.; Adnan, S.M.; Zahir, J.; Shagufa, M. Antimicrobial Activity of Ethyl Acetate, Chloroform and Deionized Water Extract of Leaves of Pteris Cretica. Ann. Romanian Soc. Cell Biol. 2021, 25, 1493–1501. [Google Scholar]
  4. Ahmed, M.; Khan, K.-U.-R.; Ahmad, S.; Aati, H.Y.; Ovatlarnporn, C.; Rehman, M.S.-U.; Javed, T.; Khursheed, A.; Ghalloo, B.A.; Dilshad, R.; et al. Comprehensive Phytochemical Profiling, Biological Activities, and Molecular Docking Studies of Pleurospermum candollei: An Insight into Potential for Natural Products Development. Molecules 2022, 27, 4113. [Google Scholar] [CrossRef]
  5. Jain, C.; Khatana, S.; Vijayvergia, R. Bioactivity of secondary metabolites of various plants: A review. Int. J. Pharm. Sci. Res 2019, 10, 494–504. [Google Scholar]
  6. Tungmunnithum, D.; Thongboonyou, A.; Pholboon, A.; Yangsabai, A.J.M. Flavonoids and other phenolic compounds from medicinal plants for pharmaceutical and medical aspects: An overview. Medicines 2018, 5, 93. [Google Scholar] [CrossRef] [PubMed]
  7. Krishnaiah, D.; Sarbatly, R. A review of the antioxidant potential of medicinal plant species. Food Bioprod. Process. 2011, 89, 217–233. [Google Scholar] [CrossRef]
  8. Jahanban-Esfahlan, A.; Ostadrahimi, A.; Tabibiazar, M.; Amarowicz, R.J.M. A comparative review on the extraction, antioxidant content and antioxidant potential of different parts of walnut (Juglans regia L.) fruit and tree. Molecules 2019, 24, 2133. [Google Scholar] [CrossRef]
  9. Mroczek, A. Phytochemistry and bioactivity of triterpene saponins from Amaranthaceae family. Phytochem. Rev. 2015, 14, 577–605. [Google Scholar] [CrossRef]
  10. Yousuf, M.; Khan, H.M.S.; Rasool, F.; Usman, F.; Ghalloo, B.A.; Umair, M.; Babalghith, A.O.; Kamran, M.; Aadil, R.M.; Al Jaouni, S.K. Chemical Profiling, Formulation Development, In Vitro Evaluation and Molecular Docking of Piper nigrum Seeds Extract Loaded Emulgel for Anti-Aging. Molecules 2022, 27, 5990. [Google Scholar] [CrossRef]
  11. Kumar, S.; Sharma, S.; Vasudeva, N. Review on antioxidants and evaluation procedures. Chin. J. Integr. Med. 2017, 1–12. [Google Scholar] [CrossRef] [PubMed]
  12. Atangwho, I.J.; Egbung, G.E.; Ahmad, M.; Yam, M.F.; Asmawi, M.Z. Antioxidant versus anti-diabetic properties of leaves from Vernonia amygdalina Del. growing in Malaysia. Food Chem. 2013, 141, 3428–3434. [Google Scholar] [CrossRef] [PubMed]
  13. Nisar, R.; Ahmad, S.; Khan, K.-U.-R.; Sherif, A.E.; Alasmari, F.; Almuqati, A.F.; Ovatlarnporn, C.; Khan, M.A.; Umair, M.; Rao, H. Metabolic Profiling by GC-MS, In Vitro Biological Potential, and In Silico Molecular Docking Studies of Verbena officinalis. Molecules 2022, 27, 6685. [Google Scholar] [CrossRef]
  14. Adeloye, J.B.; Aluko, P.A.; Oluwajuyitan, T.D. In vitro α-amylase and α-glucosidase inhibitory activities, antioxidant activity, in vivo glycemic response and nutritional quality of dough meals from Dioscorea alata and Vernonia amygdalina. J. Food Meas. Charact. 2021, 15, 4083–4097. [Google Scholar] [CrossRef]
  15. Nguelefack, T.B.; Fofie, C.K.; Nguelefack-Mbuyo, E.P.; Wuyt, A.K. Multimodal α-glucosidase and α-amylase inhibition and antioxidant effect of the aqueous and methanol extracts from the trunk bark of Ceiba pentandra. BioMed Res. Int. 2020, 2020, 3063674. [Google Scholar] [CrossRef]
  16. Najmi, A.; Javed, S.A.; Al Bratty, M.; Alhazmi, H.A. Modern approaches in the discovery and development of plant-based natural products and their analogues as potential therapeutic agents. Molecules 2022, 27, 349. [Google Scholar] [CrossRef] [PubMed]
  17. Baber, A. Phytochemical and Biological Screening of Haloxylon stocksii. Ph.D. Thesis, The Islmia University of Bahawalpur, Bahawalpur, Pakistan, 2018. [Google Scholar]
  18. Abbas, S.; Rasheed, A.; Soriano, P.; Estrelles, E.; Gul, B.; Hameed, A. Moisture content and oxidative damage determine longevity of the seeds of potential cash crop halophyte Haloxylon stocksii. Plant Biosyst. -Int. J. Deal. All Asp. Plant Biol. 2022, 156, 1478–1484. [Google Scholar] [CrossRef]
  19. Rathore, V.S.; Singh, J.P.; Roy, M.M. Haloxylon stocksii (Boiss.) Benth. et Hook. f., a promising halophyte: Distribution, cultivation and utilization. Genet. Resour. Crop Evol. 2012, 59, 1213–1221. [Google Scholar] [CrossRef]
  20. Babar, A.; Ahmad, S.; Rehman, T.; Sabah, N.U.; Arshad, M.A. A Review on Phytochemical Analysis and Ethnobotanical Uses of Haloxylon stocksii. RADS J. Pharm. Pharm. Sci. 2018, 6, 162–167. [Google Scholar]
  21. Natubhai, P.M.; Pandya, D.S.S.; Rabari, D.H.A. Anti-Inflammatory activity of leaf extracts of salvadora oleoides decne. Int. J. Pharm. Bio. Sci. 2013, 4, 985–993. [Google Scholar]
  22. Zafar, M.I.; Nazli, A.; Riaz, M.A.; Ashraf, M.R.; Iqbal, M.M.; Khan, A.; Iqbal, Z.; Javed, A.; Ahmed, S.; Haq, I.U. Toxic potential of alstonia scholaris and salvadora oleiodes leaves extract against subterranean termites, microtermes obesi (isoptera:termitidae). Int. J. Pharmacogn. 2020, 7, 109–115. [Google Scholar]
  23. Korejo, F.; Noreen, R.; Ali, S.A.; Humayun, F.; Rahman, A.; Sultana, V.; Ehteshamul-Haque, S. Evaluation of antibacterial and antifungal potential of endophytic fluorescent pseudomonas associated with salvadora persica L. and salvadora oleoides decne. Pak. J. Bot. 2017, 49, 1995–2004. [Google Scholar]
  24. Kumar, D.; Parcha, V.; Maithani, A. Combined effect on antihyperlipidemic activity of Bauhinia variegata (linn.) and Salvadora oleoides (decne.) in Triton WR-1339 induced hyperlipidemic rats. World J. Pharm. Sci. 2014, 2, 428–435. [Google Scholar]
  25. Yi, B.-H.; Kim, D.-H. Antioxidant activity of maltol, kojic acid, levulinic acid, furfural, 5-hydroxymethyl furfural, and pyrazine. Korean J. Food Sci. Technol. 1982, 14, 265–270. [Google Scholar]
  26. Jerković, I.; Kasum, A.; Marijanović, Z.; Tuberoso, C.I.G. Contribution to the characterisation of honey-based Sardinian product abbamele: Volatile aroma composition, honey marker compounds and antioxidant activity. Food Chem. 2011, 124, 401–410. [Google Scholar] [CrossRef]
  27. Teoh, Y.P.; Don, M.M. Effect of Temperature on Schizophyllum commune Growth and 4H-pyran-4-one, 2, 3-dihydro-3, 5-dihydroxy-6-methyl-Production using a Bubble Column Bioreactor. Chiang Mai J. Sci. 2015, 42, 539–548. [Google Scholar]
  28. Praveena, P.E.; Periasamy, S.; Kumar, A.; Singh, N. Cytokine profiles, apoptosis and pathology of experimental Pasteurella multocida serotype A1 infection in mice. Res. Vet. Sci. 2010, 89, 332–339. [Google Scholar] [CrossRef]
  29. Nascimento, M.A.D.; Vargas, J.P.C.; Rodrigues, J.G.A.; Leão, R.A.C.; De Moura, P.H.B.; Leal, I.C.R.; Bassut, J.; De Souza, R.O.M.A.; Wojcieszak, R.; Itabaiana, I. Lipase-catalyzed acylation of levoglucosan in continuous flow: Antibacterial and biosurfactant studies. RSC Adv. 2022, 12, 3027–3035. [Google Scholar] [CrossRef]
  30. Shaaban, M.T.; Ghaly, M.F.; Fahmi, S.M. Antibacterial activities of hexadecanoic acid methyl ester and green-synthesized silver nanoparticles against multidrug-resistant bacteria. J. Basic Microbiol. 2021, 61, 557–568. [Google Scholar] [CrossRef]
  31. Jitendra, R.; Sonu, R.; Narayan, S.R. Chemical study of fatty acids in mimusops elengiseeds by GC-MS and comparison of its antibacterial activity with trimethoprim. World J. Pharm. Res. 2015, 4, 1022–1026. [Google Scholar]
  32. Mohamad, O.A.; Li, L.; Ma, J.-B.; Hatab, S.; Xu, L.; Guo, J.-W.; Rasulov, B.A.; Liu, Y.-H.; Hedlund, B.P.; Li, W.-J. Evaluation of the antimicrobial activity of endophytic bacterial populations from Chinese traditional medicinal plant licorice and characterization of the bioactive secondary metabolites produced by Bacillus atrophaeus against Verticillium dahliae. Front. Microbiol. 2018, 9, 924. [Google Scholar] [CrossRef] [PubMed]
  33. Meechaona, R.; Sengpracha, W.; Banditpuritat, J.; Kawaree, R.; Phutdhawong, W. Fatty acid content and antioxidant activity of Thai bananas. Maejo Int. J. Sci. Technol. 2007, 1, 222–228. [Google Scholar]
  34. Othman, A.R.; Abdullah, N.; Ahmad, S.; Ismail, I.S.; Zakaria, M.P. Elucidation of in-vitro anti-inflammatory bioactive compounds isolated from Jatropha curcas L. plant root. BMC Complement. Altern. Med. 2015, 15, 11. [Google Scholar] [CrossRef] [PubMed]
  35. Suseem, S.; Saral, A.M. Analysis on essential fatty acid esters of mushroom pleurotus eous and its antibacterial activity. Asian J. Pharm. Clin. Res. 2013, 6, 188–191. [Google Scholar]
  36. An, K.; Fu, M.; Zhang, H.; Tang, D.; Xu, Y.; Xiao, G. Effect of ethyl oleate pretreatment on blueberry (Vaccinium corymbosum L.): Drying kinetics, antioxidant activity, and structure of wax layer. J. Food Sci. Technol. 2019, 56, 783–791. [Google Scholar] [CrossRef]
  37. Khan, N.; Ali, A.; Qadir, A.; Ali, A.; Warsi, M.H.; Tahir, A.; Ali, A. GC-MS Analysis and Antioxidant Activity of Wrightia tinctoria R. Br. Leaf Extract. J. AOAC Int. 2021, 104, 1415–1419. [Google Scholar] [CrossRef]
  38. Yatsuda, R.; Rosalen, P.; Cury, J.; Murata, R.; Rehder, V.; Melo, L.; Koo, H. Effects of Mikania genus plants on growth and cell adherence of mutans streptococci. J. Ethnopharmacol. 2005, 97, 183–189. [Google Scholar] [CrossRef]
  39. Krishnaveni, M.; Dhanalakshmi, R.; Nandhini, N. GC-MS analysis of phytochemicals, fatty acid profile, antimicrobial activity of Gossypium seeds. Int. J. Pharm. Sci. Rev. Res. 2014, 27, 273–276. [Google Scholar]
  40. Husein, A.I.; Ali-Shtayeh, M.S.; Jamous, R.M.; Jondi, W.J.; Zatar, N.A.-A. Phthalate derivatives are naturally occurring in Arum Palaestinum. Int. J. Curr. Res. Aca. Rev 2014, 2, 195–203. [Google Scholar]
  41. Sivasubramanian, R.; Brindha, P. In-vitro cytotoxic, antioxidant and GC-MS studies on Centratherum punctatum Cass. Int. J. Pharm. Pharm. Sci 2013, 4, e8. [Google Scholar]
  42. Mohammad, A.K.; Mohammad, R.D.; Masumeh, K.; Somayeh, N. The GC-MS analyses of the n-hexane extract of Nitraria schoberi L., its total phenolics and in vitro antioxidant activity. J. Med. Plants Res. 2012, 6, 4874–4878. [Google Scholar]
  43. Izu, G.O.; Adeyi, A.O.; Erukainure, O.L.; Islam, M.S. Gamma-sitosterol–rich fraction from the methanolic extract of Ficus exasperata restores diabetes associated pathophysiological alterations in an alloxan-induced diabetic rats. Biokemistri 2022, 33. Available online: https://ojs.klobexjournals.com/index.php/bkr/article/view/1351/0 (accessed on 25 March 2023).
  44. Dilshad, R.; Ahmad, S.; Aati, H.; Al-qahtani, J.H.; Sherif, A.E.; Hussain, M.; Ghalloo, B.A.; Tahir, H.; Basit, A.; Ahmed, M. Phytochemical Profiling, In Vitro Biological Activities, and In-Silico Molecular Docking Studies of Typha domingensis. Arab. J. Chem. 2022, 15, 104133. [Google Scholar] [CrossRef]
  45. Dores, R.G.; Guimarães, S.F.; Braga, T.V.; Fonseca, M.; Martins, P.M.; Ferreira, T.C. Phenolic compounds, flavonoids and antioxidant activity of leaves, flowers and roots of goat weed. Hortic. Bras. 2014, 32, 486–490. [Google Scholar] [CrossRef]
  46. Goufo, P.; Singh, R.K.; Cortez, I.J.A. A reference list of phenolic compounds (including stilbenes) in grapevine (Vitis vinifera L. ) roots, woods, canes, stems, and leaves. 2020, 9, 398. [Google Scholar]
  47. Grzegorczyk-Karolak, I.; Kuźma, L.; Skała, E.; Kiss, A.K. Hairy root cultures of Salvia viridis L. for production of polyphenolic compounds. Ind. Crop. Prod. 2018, 117, 235–244. [Google Scholar] [CrossRef]
  48. Peerzada, S.; Khan, M.T.; Akhtar, M.F.; Saleem, A.; Hamid, I.; Akhtar, B.; Ali, S.; Ahmed, S.; Raza, M. Phytochemical, anti-inflammatory, anti-nociceptive and cytotoxic basis for the use of Haloxylon stocksii. Pak. J. Pharm. Sci. 2020, 33, 887–894. [Google Scholar]
  49. He, L.; Gao, Y.; Zhao, L. Online coupling of bubbling extraction with gas chromatography-mass spectrometry for rapid quantitative analysis of volatiles in beer. J. Chromatogr. A 2022, 1665, 462800. [Google Scholar] [CrossRef]
  50. Meena, B.R.; Meena, S.; Chittora, D.; Sharma, K. Antifungal efficacy of Thevetia peruviana leaf extract against Alternaria solani and characterization of novel inhibitory compounds by Gas Chromatography-Mass Spectrometry analysis. Biochem. Biophys. Rep. 2021, 25, 100914. [Google Scholar] [CrossRef]
  51. Vander Pyl, C.; Feeney, W.; Arroyo, L.; Trejos, T. Capabilities and Limitations of GC-MS and LC-MS/MS for Trace Detection of Organic Gunshot Residues from Skin Specimens. Forensic Chem. 2023, 51, 100471. [Google Scholar] [CrossRef]
  52. Ghalloo, B.A.; Khan, K.-U.-R.; Ahmad, S.; Aati, H.Y.; Al-Qahtani, J.H.; Ali, B.; Mukhtar, I.; Hussain, M.; Shahzad, M.N.; Ahmed, I. Phytochemical Profiling, In Vitro Biological Activities, and In Silico Molecular Docking Studies of Dracaena reflexa. Molecules 2022, 27, 913. [Google Scholar] [CrossRef] [PubMed]
  53. Basit, A.; Ahmad, S.; Sherif, A.E.; Aati, H.Y.; Ovatlarnporn, C.; Khan, M.A.; Rao, H.; Ahmad, I.; Shahzad, M.N.; Ghalloo, B.A. New mechanistic insights on Justicia vahlii Roth: UPLC-Q-TOF-MS and GC–MS based metabolomics, in-vivo, in-silico toxicological, antioxidant based anti-inflammatory and enzyme inhibition evaluation. Arab. J. Chem. 2022, 15, 104135. [Google Scholar] [CrossRef]
  54. Khursheed, A.; Ahmad, S.; Khan, K.-U.-R.; Tousif, M.I.; Aati, H.Y.; Ovatlarnporn, C.; Rao, H.; Khurshid, U.; Ghalloo, B.A.; Tabassum, S. Efficacy of Phytochemicals Derived from Roots of Rondeletia odorata as Antioxidant, Antiulcer, Diuretic, Skin Brightening and Hemolytic Agents—A Comprehensive Biochemical and In Silico Study. Molecules 2022, 27, 4204. [Google Scholar] [CrossRef]
  55. Al-Qahtani, J.; Abbasi, A.; Aati, H.Y.; Al-Taweel, A.; Al-Abdali, A.; Aati, S.; Yanbawi, A.N.; Khan, M.A.; Ghalloo, B.A.; Anwar, M. Phytochemical, Antimicrobial, Antidiabetic, Thrombolytic, anticancer Activities, and in silico studies of Ficus palmata Forssk. Arab. J. Chem. 2023, 16, 104455. [Google Scholar] [CrossRef]
  56. Yasin, G.; Noor, S.; Irshad, S.; Haq, I.; Fatima, S.; Anwer, I.; Khan, A. Solvents Based Extraction of Antioxidants and their Activity from Some Plants of Cholistan Desert, Pakistan. Int. J. Pharm. Phytopharm. Res. 2020, 10, 70–76. [Google Scholar]
  57. Alqahtani, F.Y.; Aleanizy, F.S.; Mahmoud, A.Z.; Farshori, N.N.; Alfaraj, R.; Al-Sheddi, E.S.; Alsarra, I.A. Chemical composition and antimicrobial, antioxidant, and anti-inflammatory activities of Lepidium sativum seed oil. Saudi J. Biol. Sci. 2019, 26, 1089–1092. [Google Scholar] [CrossRef] [PubMed]
  58. Gupta, R.; Sharma, A.K.; Dobhal, M.; Sharma, M.; Gupta, R. Antidiabetic and antioxidant potential of β-sitosterol in streptozotocin-induced experimental hyperglycemia. J. Diabetes 2011, 3, 29–37. [Google Scholar] [CrossRef]
  59. Balaha, M.; Ahmed, N.; Geddawy, A.; Kandeel, S. Fraxetin prevented sodium fluoride-induced chronic pancreatitis in rats: Role of anti-inflammatory, antioxidant, antifibrotic and anti-apoptotic activities. Int. Immunopharmacol. 2021, 93, 107372. [Google Scholar] [CrossRef]
  60. El Moussaoui, A.; Jawhari, F.Z.; Almehdi, A.M.; Elmsellem, H.; Benbrahim, K.F.; Bousta, D.; Bari, A. Antibacterial, antifungal and antioxidant activity of total polyphenols of Withania frutescens.L. Bioorganic Chem. 2019, 93, 103337. [Google Scholar] [CrossRef]
  61. Kumar, A.; Aswal, S.; Semwal, R.B.; Chauhan, A.; Joshi, S.K.; Semwal, D.K. Role of plant-derived alkaloids against diabetes and diabetes-related complications: A mechanism-based approach. Phytochem. Rev. 2019, 18, 1277–1298. [Google Scholar] [CrossRef]
  62. Minh, T.N.; Xuan, T.D.; Tran, H.-D.; Van, T.M.; Andriana, Y.; Khanh, T.D.; Quan, N.V.; Ahmad, A. Isolation and purification of bioactive compounds from the stem bark of Jatropha podagrica. Molecules 2019, 24, 889. [Google Scholar] [CrossRef]
  63. Haq, I.U.; Imran, M.; Nadeem, M.; Tufail, T.; Gondal, T.A.; Mubarak, M.S. Piperine: A review of its biological effects. Phytother. Res. 2021, 35, 680–700. [Google Scholar] [CrossRef] [PubMed]
  64. Reid, T.; Kashangura, C.; Chidewe, C.; Benhura, M.A.; Stray-Pedersen, B.; Mduluza, T. Characterization of Anti− Salmonella typhi compounds from medicinal mushroom extracts from Zimbabwe. Int. J. Med. Mushrooms 2019, 21, 713–724. [Google Scholar] [CrossRef] [PubMed]
  65. Aisiah, S.; Rini, R.K.; Tanod, W.A.; Fatmawati, F.; Fauzana, N.A.; Olga, O.; Riyadi, P.H. Metabolomic profiling of Jeruju (Acanthus ilicifolius) leaf extract with antioxidant and antibacterial activity on Aeromonas hydrophila growth. J. Appl. Pharm. Sci. 2022, 12, 057–069. [Google Scholar] [CrossRef]
  66. Wahab, A.; Ahmed, E.; Nawaz, S.A.; Sharif, A.; Haq, R.U.; Malik, A.; Choudhary, M.I.; Raza, M. A pharmacological and toxicological evaluation of Haloxylon recurvum. Nat. Prod. Res. 2008, 22, 1317–1326. [Google Scholar] [CrossRef] [PubMed]
  67. Senhaji, S.; Lamchouri, F.; Toufik, H. Phytochemical content, antibacterial and antioxidant potential of endemic plant anabasis aretioïdes coss. & moq.(Chenopodiaceae). BioMed Res. Int. 2020, 2020, 6152932. [Google Scholar]
  68. Sekhon-Loodu, S.; Rupasinghe, H.V. Evaluation of antioxidant, antidiabetic and antiobesity potential of selected traditional medicinal plants. Front. Nutr. 2019, 6, 53. [Google Scholar] [CrossRef]
  69. Truong, D.-H.; Nguyen, D.H.; Ta, N.T.A.; Bui, A.V.; Do, T.H.; Nguyen, H.C. Evaluation of the use of different solvents for phytochemical constituents, antioxidants, and in vitro anti-inflammatory activities of Severinia buxifolia. J. Food Qual. 2019, 2019, 8178294. [Google Scholar] [CrossRef]
  70. Su, S.; Cao, M.; Wu, G.; Long, Z.; Cheng, X.; Fan, J.; Xu, Z.; Su, H.; Hao, Y.; Li, G. Hordenine protects against hyperglycemia-associated renal complications in streptozotocin-induced diabetic mice. Biomed. Pharmacother. 2018, 104, 315–324. [Google Scholar] [CrossRef]
  71. Ozer, M.S.; Sarikurkcu, C.; Tepe, B. Phenolic composition, antioxidant and enzyme inhibitory activities of ethanol and water extracts of Chenopodium botrys. RSC Adv. 2016, 6, 64986–64992. [Google Scholar] [CrossRef]
  72. Chikhi, I.; Allali, H.; Dib, M.E.A.; Medjdoub, H.; Tabti, B. Antidiabetic activity of aqueous leaf extract of Atriplex halimus L. (Chenopodiaceae) in streptozotocin–induced diabetic rats. Asian Pac. J. Trop. Dis. 2014, 4, 181–184. [Google Scholar] [CrossRef]
  73. Aati, H.Y.; Anwar, M.; Al-Qahtani, J.; Al-Taweel, A.; Khan, K.-U.-R.; Aati, S.; Usman, F.; Ghalloo, B.A.; Asif, H.M.; Shirazi, J.H.; et al. Phytochemical Profiling, In Vitro Biological Activities, and In-Silico Studies of Ficus vasta Forssk.: An Unexplored Plant. Antibiotics 2022, 11, 1155. [Google Scholar] [CrossRef] [PubMed]
  74. Ahmed, M.; Ahmad, S.; Aati, H.Y.; Sherif, A.E.; Ashkan, M.F.; Alrahimi, J.; Motwali, E.A.; Tousif, M.I.; Khan, M.A.; Hussain, M. Phytochemical, Antioxidant, Enzyme Inhibitory, Thrombolytic, Antibacterial, Antiviral and In Silico Studies of Acacia jacquemontii Leaves. Arab. J. Chem. 2022, 15, 104345. [Google Scholar] [CrossRef]
  75. Dilshad, R.; Khan, K.-U.-R.; Saeed, L.; Sherif, A.E.; Ahmad, S.; Ovatlarnporn, C.; Nasim, J.; Hussain, M.; Ghalloo, B.A.; Basit, A.; et al. Chemical Composition and Biological Evaluation of Typha domingensis Pers. to Ameliorate Health Pathologies: In Vitro and In Silico Approaches. BioMed Res. Int. 2022, 2022, 8010395. [Google Scholar] [CrossRef]
  76. Shahzad, M.N.; Ahmad, S.; Tousif, M.I.; Ahmad, I.; Rao, H.; Ahmad, B.; Basit, A. Profiling of phytochemicals from aerial parts of Terminalia neotaliala using LC-ESI-MS2 and determination of antioxidant and enzyme inhibition activities. PLoS ONE 2022, 17, e0266094. [Google Scholar] [CrossRef] [PubMed]
  77. Majid, M.; Farhan, A.; Asad, M.I.; Khan, M.R.; Hassan, S.S.U.; Haq, I.-U.; Bungau, S. An Extensive Pharmacological Evaluation of New Anti-Cancer Triterpenoid (Nummularic Acid) from Ipomoea batatas through In Vitro, In Silico, and In Vivo Studies. Molecules 2022, 27, 2474. [Google Scholar] [CrossRef]
  78. Mahnashi, M.H.; Alshahrani, M.A.; Nahari, M.H.; Hassan, S.S.U.; Jan, M.S.; Ayaz, M.; Ullah, F.; Alshehri, O.M.; Alshehri, M.A.; Rashid, U. In-Vitro, In-Vivo, Molecular Docking and ADMET Studies of 2-Substituted 3, 7-Dihydroxy-4H-chromen-4-one for Oxidative Stress, Inflammation and Alzheimer’s Disease. Metabolites 2022, 12, 1055. [Google Scholar] [CrossRef]
Figure 1. (A) α-amylase inhibition activity of extracts and acarbose (standard) and (B) α-glucosidase inhibition activity of extracts and acarbose (Standard). The superscripts a, b, c, d, and e represent significant difference (p < 0.05).
Figure 1. (A) α-amylase inhibition activity of extracts and acarbose (standard) and (B) α-glucosidase inhibition activity of extracts and acarbose (Standard). The superscripts a, b, c, d, and e represent significant difference (p < 0.05).
Molecules 28 03847 g001
Figure 2. Interaction of (A) γ-Sitosterol, (B) phenol, 2,2′-methylenebis[6-(1, 1-dimethylethyl)-4-(1-methylpropyl), and (C) acarbose with α-amylase.
Figure 2. Interaction of (A) γ-Sitosterol, (B) phenol, 2,2′-methylenebis[6-(1, 1-dimethylethyl)-4-(1-methylpropyl), and (C) acarbose with α-amylase.
Molecules 28 03847 g002aMolecules 28 03847 g002b
Figure 3. Interaction of (A) γ-Sitosterol, (B) phenol, 2,2′-methylenebis[6-(1, 1-dimethylethyl)-4-(1-methylpropyl), and (C) acarbose with α-glucosidase.
Figure 3. Interaction of (A) γ-Sitosterol, (B) phenol, 2,2′-methylenebis[6-(1, 1-dimethylethyl)-4-(1-methylpropyl), and (C) acarbose with α-glucosidase.
Molecules 28 03847 g003aMolecules 28 03847 g003b
Figure 4. The bioavailability radar of in silico studied molecules. The pink area indicates oral bioavailability considering polarity, lipophilicity and size of the compounds.
Figure 4. The bioavailability radar of in silico studied molecules. The pink area indicates oral bioavailability considering polarity, lipophilicity and size of the compounds.
Molecules 28 03847 g004aMolecules 28 03847 g004b
Table 1. TPC and TFC values for different extracts (dry weight) of H. stocksii.
Table 1. TPC and TFC values for different extracts (dry weight) of H. stocksii.
ExtractTPC (mg GAE/g Extract)TFC (mg QE/g Extract)
AMHS119.58 ± 2.45 a99.19 ± 1.14 a
ADHS102.65 ± 1.79 b87.54 ± 0.73 b
RMHS91.54 ± 2.65 c65.65 ± 0.65 c
RDHS77.65 ± 1.91 d54.65 ± 0.84 d
Results are expressed as mean ± standard deviation. “AMHS” aerial parts methanolic H. stocksii extract; “ADHS” aerial parts dichloromethane H. stocksii extract; “RMHS” roots methanolic H. stocksii extract; and “RDHS” roots dichloromethane H. stocksii extract. “GAE/g extract” Gallic acid equivalent per gram of extract, “QE/g extract” Quercetin equivalent per gram of extract, the superscripts a, b, c, and d represent significant difference (p < 0.05).
Table 2. GC-MS analysis of aerial parts and roots of H. stocksii.
Table 2. GC-MS analysis of aerial parts and roots of H. stocksii.
Sr. No.Compound NamePlant PartMolecular FormulaM.W.
(g/moL)
Rt (min)Percentage Area (%)Chemical ClassBiological Activity
1FurfuralA, RC5H4O296.082.732.43Furan aldehydeAntimicrobial andantioxidant [25]
22-Furancarboxaldehyde, 5-methyl-A, RC6H6O2110.13.652.38Furans and aldehydeAntimicrobial and antioxidant [26]
34H-Pyran-4-one, 2,3-dihydro-3,5-dihydroxy-6-methyl-A, RC5H6O4130.105.913.80PyronesAntimicrobial and antioxidant [27]
42-Furancarboxaldehyde, 5-(hydroxymethyl)-A, RC6H6O3126.16.6424.05Aryl-aldehudeAntioxidant and antibacterial [28]
51,6-Anhydro-β-ᴅ-glucopyranoseRC6H10O5162.149.225.95AnhydrohexoseAntibacterial [29]
6Hexadecanoic acid, methyl esterA, RC17H34O2270.510.521.13Fatty acid esterAntibacterial and antioxidant [30]
7Pentadecanoic acid, 14-methyl-, methyl esterRC17H34O2270.510.531.70Fatty acid methyl esterAntioxidant and antimicrobial [31]
89-Octadecenoic acid, methyl esterA, RC19H36O2296.511.981.75Fatty acid esterAntibacterial, antioxidant, and
anti-inflammatory [32]
9Octadecanoic acid, methyl esterRC19H38O2298.512.160.29Fatty acid methyl esterCytotoxicity, antioxidant activity, and anti-inflammatory [33,34]
10Heptadecanoic acid, 16-methyl-methyl esterAC19H38O2298.512.150.08Fatty acid esterAntibacterial and antioxidant [35]
11Ethyl OleateA, RC20H38O2310.512.510.31Fatty acid ethyl esterAntioxidant and antimicrobial [36]
12Eicosanoic acid, methyl esterA, RC21H42O2326.613.830.03Fatty acid esterAntimicrobial and antioxidant [37]
131-OctadeceneRC18H36252.513.490.23OctadeceneAntimicrobial [38]
14Phenol, 2,2′-methylenebis[6-(1, 1-dimethylethyl)-4-(1-methylpropyl)AC29H44O2424.714.890.05Phenol
15Docosanoic acid, methyl esterAC23H46O2354.615.870.04Fatty acid methyl esterAntioxidant [39]
16Di-n-octyl phthalateA, RC24H38O4390.616.160.21Phthalate esterAnticancer and antioxidant [40]
17Phthalic acid, bis(7-methyloctyl) esterAC26H42O4418.619.770.18Phthalate esterAntioxidant and antimicrobial [41]
18γ-SitosterolAC29H50O414.721.140.25PhytosterolsAnalgesic, antioxidant, antidiabetic, andantibacterial [42,43,44]
A: aerial parts of H. stocksii. R: roots of H. stocksii.
Table 3. LC-ESI-MS2 profiling of methanolic extract of aerial parts.
Table 3. LC-ESI-MS2 profiling of methanolic extract of aerial parts.
Sr. No.Retention Time (Minutes)M/ZCompound NameMolecular FormulaMolecular MassChemical Class
13.504152.107N-MethyltyramineC9H13NO151.099Amines
23.505121.065AcetophenoneC8H8O120.057Aromatic ketones
34.004166.122HordenineC10H15NO165.115Phenyl amines
410.477420.144Methyl 2-(4-oxo-3-((4-(p-tolyl)thiazol-2-yl)methyl)-3,4-dihydrophthalazin-1-yl)acetateC23H21N3O3 S419.137Acetamides
511.821209.044FraxetinC10H8O5208.037Flavonoids
615.267223.0596,8-Dihydroxy-7-methoxy-3-methyl-1H-isochromen-1-oneC11H10O5222.053Flavonoids
716.846300.122UnknownC17H17NO4299.115
817.395247.179nor-3-MethylfentanylC15H22N2O246.173Opioids
919.106314.138MoupinamideC18H19NO4313.131Amides
1027.591343.1172-(2,6-Dimethoxyphenyl)-5,6-dimethoxy-4H-chromen-4-oneC19H18O6342.109
1129.578286.143PiperineC17H19NO3285.136Alkaloids
1238.689399.251Tris(2-butoxyethyl) phosphateC18H39O7P398.243Phosphate esters
1343.688403.232Acetyl tributyl citrateC20H34O8402.225Organophophate esters
1445.134352.321UnknownC22H41NO2351.314
1545.755309.242UnknownC19H32O3308.235
1646.749256.263HexadecanamideC16H33NO255.256Fatty acid amides
1748.076354.337UnknownC22H43NO2353.329
1848.144331.2841-PalmitoylglycerolC19H38O4330.277Glycerolipid
1949.561191.143(1S)-Tricyclo[7.3.1.0~2,7~]tridec-2(7)- en-13-oneC13H18O190.136Cyclic ketone
2052..221284.294StearamideC18H37NO283.287Long-chain fatty acid
2155.801338.342ErucamideC22H43NO337.334Long-chain fatty acid
2255.152394.347UnkownC28H43N393.339
2356.318122.096N,N-DimethylanilineC8H11N121.089Aromatic amines
“Unknown” Not identified with Discoverer 3.3.
Table 4. DPPH, ABTS, CUPRAC, and FRAP values for different extracts (dry weight) of H. stocksii.
Table 4. DPPH, ABTS, CUPRAC, and FRAP values for different extracts (dry weight) of H. stocksii.
ExtractDPPH (mg TE/g Extract)ABTS (mg TE/g Extract)FRAP (mg TE/g Extract)CUPRAC (mg TE/g Extract)
AMHS145.45 ± 2.94 a98.07 ± 3.47 a231.76 ± 7.69 a410.08 ± 10.51 a
ADHS121.65 ± 3.4 b66.65 ± 2.93 b192.27 ± 5.7 b367.42 ± 8.35 b
RMHS93.41 ± 1.99 c68.07 ± 1.15 b168.67 ± 3.81 c332.65 ± 7.91 c
RDHS74.98 ± 1.28 d52.98 ± 0.82 c145.64 ± 2.6 d296.12 ± 5.84 d
Results are expressed as mean ± standard deviation. “DPPH” 1,1-diphenyl-2-picrylhydrazyl; “ABTS” (2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid)); “FRAP” Ferric ion reducing antioxidant power; “AMHS” aerial parts methanolic H. stocksii extract; “ADHS” aerial parts dichloromethane H. stocksii extract; “RMHS” roots methanolic H. stocksii extract; and “RDHS” roots dichloromethane H. stocksii extract. the superscripts a, b, c, and d represent significant difference (p < 0.05).
Table 5. Antibacterial activity of Haloxylon stocksii extracts by disc difusion method.
Table 5. Antibacterial activity of Haloxylon stocksii extracts by disc difusion method.
Strain NameZI of StdZI of AMHSZI of ADHSZI of RMHSZI of RDHS
Bacillus subtilis23 ± 1.5520 ± 1.3318 ± 0.7117 ± 1.3919 ± 1.62
Bacillus pumilus25 ± 0.7722 ± 1.5116 ± 1.6615 ± 0.5714 ± 1.26
Micrococcus luteus23 ± 0.9516 ± 0.6519 ± 1.5114 ± 1.1914 ± 0.87
Staphylococcus epidermidis24 ± 1.1318 ± 0.8317 ± 1.1517 ± 1.2816 ± 0.63
Escherichia coli22 ± 0.4717 ± 1.2016 ± 1.2519 ± 1.4218 ± 0.96
Bordetella bronchispetica25 ± 1.3112 ± 0.3310 ± 0.4315 ± 1.0914 ± 0.75
Pseudomonas aeruginosaNA9 ± 0.358 ± 0.2117 ± 1.3714 ± 1.16
Results are expressed as mean ± standard deviation. “ZI” zone of inhibition in mm; “Std” standard co-amoxiclav; “AMHS” aerial parts methanolic extract; “ADHS” aerial parts dichloromethane extract; “RMHS” roots methanolic extract; and “RDHS” roots dichloromethane extract.
Table 6. Molecular docking study of H. stocksii against α-amylase.
Table 6. Molecular docking study of H. stocksii against α-amylase.
Sr. No.LigandsBinding AffinityAmino Acid Interactions
Hydrogen BondingPi Alkyl
1Octadecanoic acid, methyl ester−5Gln63Trp58, Trp59, Tyr62, Leu162, Leu165, His101, His305
24H-Pyran-4-one, 2,3-dihydro-3,5-dihydroxy-6-methyl-−5.1Gly309, Asp317, Arg346
39-Octadecenoic acid, methyl ester−5.2Gln63Trp59, Tyr62, Leu162, Leu165
4Eicosanoic acid, methyl ester−5.3 Trp59, Tyr62, Ala198, Leu162, Leu165, His201
51,6-Anhydro-β-ᴅ-glucopyranose−5.8Asn301, Gln302, Gly304, Ile312, Arg346Unfavorable: Arg267
6Phthalic acid, bis(7-methyloctyl) ester−5.9His101, Glu233, Asp300Pi-sigma: Trp59, Leu162
Pi-Alkyl: Trp58, Tyr62, His305
7Di-n-octyl phthalate−6Asp197Pi-sigma: Trp59, Ile235
Alkyl/Pi-Alkyl: Trp58, Tyr62, Leu162, Leu165, Ala198, His305
8Phenol, 2,2′-methylenebis[6-(1, 1-dimethylethyl)-4-(1-methylpropyl)−8.5His305Pi-Anion: Asp300
Pi-sigma: Tyr62
Pi-Pi Stacked: Trp58, Trp59
Alkyl/Pi-Alkyl: Leu165, His299
9γ-Sitosterol−8.7 Alkyl/Pi-Alkyl: Trp59, Leu165, Ala198
Acarbose−6.9Asp197, Glu233, His305, Lys352, Asp356Pro54, Trp58, Trp59, His101, Leu162, Ser163, Leu165, Arg195, Ala198, His299, Asp300, Trp357
Table 7. Molecular docking study of H. stocksii against α-glucosidase.
Table 7. Molecular docking study of H. stocksii against α-glucosidase.
Sr. No.LigandsBinding AffinityAmino Acid Interactions
Hydrogen BondingPi Alkyl
1Pentadecanoic acid, 14-methyl-, methyl ester−5Val380, Asp401, Gly402Val334, Val335, Lys398, Phe397
22-Furancarboxaldehyde, 5-(hydroxymethyl)-−5.2His515Pi-Alkyl: Lys352, Ala514
Amide-Pi Stacked: Phe516
3Ethyl Oleate−5.4Arg457Pi-sigma: Phe463
Alkyl/Pi-Alkyl: Leu95, Arg456
4Eicosanoic acid, methyl ester−5.4Arg347, Gly432His348, Ala349, Lys352, Ala444, Arg450, Ala454
51,6-Anhydro-β-ᴅ-glucopyranose−5.7Arg437, Asp441, Ala451
6Di-n-octyl phthalate−5.6Gly439, Gly581, His515Pi-sigma: Ala349
Alkyl/Pi-Alkyl: Ala43, Leu93
7Phthalic acid, bis(7-methyloctyl) ester−6.2Ser44, Asn443, Arg450Pi-sigma: Phe516
Alkyl/Pi-Alkyl: Tyr41, Lys352, Val435
8Phenol, 2,2′-methylenebis[6-(1, 1-dimethylethyl)-4-(1-methylpropyl)−8.1Asp441Pi-Cation: His348
Alkyl/Pi-Alkyl: Pro442, Ala444, Arg450, Ala451
9γ-Sitosterol−8.9Arg437His348, Ala349, Lys352, Ala444, Ala451, Ala454
Acarbose−6.8Arg17, Asp59, Asn61, Asp381, Pro433, Trp434Asn58, Asp60, Asp378, Asp379, Asp382, Val383, Met435
Table 8. Pharmacokinetic properties of best-docked compounds.
Table 8. Pharmacokinetic properties of best-docked compounds.
Sr. No.Compound NameGastrointestinal AbsorptionBlood–brain Barrier PermeantPgp
Inhibitor
CYP1A2
Inhibitor
CYP2C19
Inhibitor
CYP2C9
Inhibitor
CYP2D6
Inhibitor
CYP3A4 InhibitorLog Kp
(cm/s)
1.Octadecanoic acid, methyl esterHighNoNoYesNoNoNoNo−2.19
2.4H-Pyran-4-one, 2,3-dihydro-3,5-dihydroxy-6- methyl-HighNoNoNoNoNoNoNo−7.44
3.Ethyl OleateHighNoNoYesNoNoNoNo−2.82
4.Eicosanoic acid, methyl esterLowNoNoYesNoNoNoNo−1.69
5.1,6-Anhydro-β-ᴅ-glucopyranoseHighNoYesNoNoNoNoNo−8.82
6.Phthalic acid, bis(7-methyloctyl) esterLowNoNoNoNoNoNoYes−3.61
7.Di-n-octyl phthalateHighNoNoNoNoNoNoYes−2.93
8.Phenol,2,2′-methylenebis[6-(1,1-dimethylethyl)-4-(1- methylpropyl)LowNoYesNoNoNoYesNo−2.60
9.γ-SitosterolLowNoNoNoNoNoNoNo−2.20
10.AcarboseLowNoYesNoNoNoNoNo−16.29
Table 9. Lipinski rule of five and solubility of best-docked compounds.
Table 9. Lipinski rule of five and solubility of best-docked compounds.
Sr. No.Compounds NameLipinski’s ruleSolubility
HBDHBAMWT
(g/moL)
LipophilicityMRLRESOL ClassAli ClassSilicos-IT Class
1.Octadecanoic acid, methyl ester02298.504.8194.731Moderately solublePoorly solublePoorly soluble
2.4H-Pyran-4-one, 2,3- dihydro-3,5-dihydroxy-6- methyl-24144.131.1932.390Very solubleVery solubleSoluble
3.Ethyl Oleate02296.494.7594.261Moderately solublePoorly solublePoorly soluble
4.Eicosanoic acid, methyl ester02326.565.35104.351Poorly solublePoorly solublePoorly soluble
5.1,6-Anhydro-β-ᴅ-glucopyranose35162.141.2732.380Highly solubleHighly solubleSoluble
6.Phthalic acid, bis(7- methyloctyl) ester04418.615.41125.911Poorly solublePoorly solublePoorly soluble
7.Di-n-octyl phthalate04390.564.14116.301Poorly solublePoorly solublePoorly soluble
8.Phenol, 2,2′-methylenebis[6-(1,1-dimethylethyl)-4-(1- methylpropyl)22424.664.95137.251Poorly solublePoorly solublePoorly soluble
9.γ-Sitosterol11414.714.79133.231Poorly solublePoorly solublePoorly soluble
10.Acarbose1419645.600.63136.693Highly solubleHighly solubleSoluble
HBD: hydrogen bond donor; HBA: hydrogen bond acceptor; MWT: molecular weight; MR: molar refractivity; LR: Lipinski rule.
Table 10. Toxicity profiles of best-docked molecules.
Table 10. Toxicity profiles of best-docked molecules.
Sr. No.Compound NamePredicted LD50
(mg/kg)
Predicted Toxicity ClassHepatotoxicityCarcinogenicityImmunotoxicityMutagenicityCytotoxicity
1.Octadecanoic acid, methyl ester50005Not activeNot activeNot activeNot activeNot active
2.4H-Pyran-4-one, 2,3- dihydro-3,5-dihydroxy-6- methyl-5954Not activeNot activeNot activeActiveNot active
3.Ethyl oleate30005Not activeNot activeNot activeNot activeNot active
4.Eicosanoic acid, methyl ester50005Not activeNot activeNot activeNot activeNot active
5.1,6-Anhydro-β-ᴅ-glucopyranose23,0006Not activeNot activeNot activeNot activeNot active
6.Phthalic acid, bis(7- methyloctyl) ester13404Not activeActiveInactiveNot activeNot active
7.Di-n-octyl phthalate13404Not activeActiveNot activeNot activeNot active
8.Phenol, 2,2′-methylenebis[6- (1, 1-dimethylethyl)-4-(1- methylpropyl)34305Not activeNot activeNot activeNot activeNot active
9.γ-Sitosterol8904Not activeNot activeActiveNot activeNot active
10.Acarbose24,0006ActiveNot activeActiveNot activeNot active
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

Rizvi, S.N.R.; Afzal, S.; Khan, K.-u.-R.; Aati, H.Y.; Rao, H.; Ghalloo, B.A.; Shahzad, M.N.; Khan, D.A.; Esatbeyoglu, T.; Korma, S.A. Chemical Characterisation, Antidiabetic, Antibacterial, and In Silico Studies for Different Extracts of Haloxylon stocksii (Boiss.) Benth: A Promising Halophyte. Molecules 2023, 28, 3847. https://doi.org/10.3390/molecules28093847

AMA Style

Rizvi SNR, Afzal S, Khan K-u-R, Aati HY, Rao H, Ghalloo BA, Shahzad MN, Khan DA, Esatbeyoglu T, Korma SA. Chemical Characterisation, Antidiabetic, Antibacterial, and In Silico Studies for Different Extracts of Haloxylon stocksii (Boiss.) Benth: A Promising Halophyte. Molecules. 2023; 28(9):3847. https://doi.org/10.3390/molecules28093847

Chicago/Turabian Style

Rizvi, Syed Nabil Raza, Samina Afzal, Kashif-ur-Rehman Khan, Hanan Y. Aati, Huma Rao, Bilal Ahmad Ghalloo, Muhammad Nadeem Shahzad, Duraiz Ahmed Khan, Tuba Esatbeyoglu, and Sameh A. Korma. 2023. "Chemical Characterisation, Antidiabetic, Antibacterial, and In Silico Studies for Different Extracts of Haloxylon stocksii (Boiss.) Benth: A Promising Halophyte" Molecules 28, no. 9: 3847. https://doi.org/10.3390/molecules28093847

Article Metrics

Back to TopTop