1Research Unit on Medicinal Plants (URPM, 3000, Laghouat), Biotechnology Research Center (CRBt, 2500, Constantine), Algeria;
2Laboratory of Organic Chemistry and Natural Substances, Faculty of Exact Sciences and Computer, University of Djelfa, PO Box 3117, Djelfa 17000, Algéria;
3Biotechnology Research Center (CRBt, 2500, Constantine), Algeria;
4Scientific Consulting, Chemical Engineering, University of Chemical Technology and Metallurgy, 1734 Sofia, Bulgaria;
5Technical Platform of Physico-Chemical Analysis (PTAPC-Laghouat-CRAPC), Laghouat, Algeria;
6Department of Chemical Engineering, University of Laghouat, (3000, Laghouat), Algeria;
7Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
8Department of Biology, University of Laghouat, (3000, Laghouat), Algeria;
9Chemistry Department Faculty, Ferhat abbas Sétif-1 University, 19000 Sétif, Algeria;
10Nuclear Research Centre of Birine, Ain Oussera, Djelfa 17200, Algeria
The chemical composition, antioxidant activities, and α-amylase enzyme inhibitory activity of Algerian Juniperus phoenicea L berries were quantitatively and qualitatively determined in this study. Essential oil (EO) and non-polar crude extracts from cyclohexane and ethyl acetate were prepared, and the chemical profile was determined using GC-MS technique. The predominant compound in the EO was α-pinene (76.03%), while communic acid (23.66% and 22.38%) was the main compound in both non-polar crude extracts. The antioxidant potential of the samples was evaluated using 1,1-diphenyl-2-picrylhydrazyl (DPPH), 2,20-azino-bis(3-ethylbenzothiazoline-6-sulfonicacid)-diammonium salt (ABTS), and phenanthroline. All samples showed weak antioxidant capacity. The antidiabetic effect was assessed in vitro using the α-amylase assay; a strong inhibitory effect against the α-amylase enzyme was detected for both cyclohexane and ethyl acetate extracts with IC50 (IC50 = 186.91 ± 5.74 mg/mL and IC50 = 351.48 ± 0.17 mg/mL, respectively). Finally, an in silico study was performed for both α-amylase and α-glucosidase proteins to enhance our outcomes.
Key words: α-amylase assay, anti-oxidant assays, essential oil, Juniperus phoenicea L berries, molecular docking, non-polar extracts
*Corresponding Authors: Abderrezak Bouchareb, Research Unit on Medicinal Plants (URPM, 3000, Laghouat), Biotechnology Research Center (CRBt, 2500, Constantine), Algeria. Email: [email protected]; Wafa Zahnit, Chemistry Department Faculty, Ferhat abbas Sétif-1 University,19000 Sétif, Algeria. Email: [email protected]; Mohammed Messaoudi, Nuclear Research Centre of Birine, Ain Oussera, Djelfa 17200, Algeria. Email: [email protected]
Received: 30 July 2024; Accepted: 21 October 2024; Published: 1 January 2025
© 2025 Codon Publications
This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0). License (http://creativecommons.org/licenses/by-nc-sa/4.0/)
The medicinal properties of plants have been widely known and used since ancient times due to their beneficial and curative effects. A medicinal plant’s distinct pharmacological qualities stem from the combined effects of its many natural elements. The whole expression of a medicinal plant’s bioactivity is found in its plant complex, which is a biochemical entity (Bouras et al., 2024). In fact, its synergistic and additive effects, increased pharmacological efficacy, or even decreased toxicity, are often not related to a single active component (Severina et al., 2013). Essential oils (EOs) are volatile compounds with a low molecular weight and biological activities synthesized in different plant organs, especially flowers, buds, leaves, branches, stems, seeds, fruits, woods, and roots (Jain et al., 2022). Furthermore, an abundance of active chemicals, including alkaloids, tannins, steroids, glycosides, resins, phenols, volatile oils, and flavonoids, are present in EOs (Amirifar et al., 2022; Asgari et al., 2017).
Nowadays, plant extracts and EOs are sources of advantageous chemical compounds with potential uses in the food, cosmetics, pharmaceutical, and agricultural industries. They are becoming popular natural alternatives to synthetic antioxidants to address consumer concerns about the adverse effects of synthetic antioxidants that have toxic effects on consumers and can lead to various cancers. Therefore, new and affordable sources of natural antioxidants are becoming more widely available in order to preserve and improve customers’ health and ensure food safety (Nastaran et al., 2021; Zahnit et al., 2022). Due to the significant amounts of volatile, aromatic, and bioactive chemicals they contain, EOs and extracts are highly valued in various sectors (Messaoudi et al., 2022; Samadi et al., 2021). Additionally, these effective compounds have inherent antioxidant, enzymatic, and antibacterial characteristics that are crucial for daily life (Bolouri et al., 2022).
Around the world, Juniperus species are frequently used in traditional medicine for a variety of purposes. Among their many uses, they are used as insect repellent, hypoglycemic, carminative, diuretic, antibacterial, antitussive, antifertility, and stomachic. However, they are also favored as treatments for urticaria, rheumatoid arthritis, TB, leukorrhea, fever, diarrhea, jaundice, and urinary tract infections. Berries of the J. phoenicea plant are the source of juniper oil, which is included in various pharmacopeias, including the 8th edition of the European Pharmacopoeia (Ph. Eur. 8). For centuries, J. phoenicea berries and their EOs have been used for both medicinal and cosmetic purposes, due to their complex combination of chemical compounds, mostly aromatics and terpene hydrocarbons, in addition to terpenoids, polyphenolic compounds, alcohols, myrcene, thujone, glycoside acetate-ethers, citronellol, curcumin, geraniol, terpineol, and ketones. According to Amalich et al. (2015), the primary chemical found in the EO of Moroccan J. phoenicea was α-pinene (78.8%), germacrene D (5.42%), 4 trans-data-4(11),7-diene (2.98%), E-caryophyllene (2.77%), α-himachalene (0.9%), and δ- cadinene (0.71%). Also, in the study by Mehira et al. (2021), which compared EO extractions from leaves and a combination of leaves/berries, a significant increase in the amount of chemical composition was found for the combination of leaves/berries. In the same context, El-Sawi et al. (2007) reported, in their study, the chemical composition of Egyptian J. phoenicea berries oil, where they found the major components were α-pinene (39.30%) followed by sabinene (24.29%), trans-pinocarveol (4.27%), β-phellandrene (4.13%), α-terpinyl acetate (3.36%), p-mentha-1,5-diene-8-ol (2.88%), and β-pinene (2.45%). Additionally, Medini et al. (2011) examined the chemical composition and antioxidant activity of the EO of Tunisian J. phoenicea L ripe and unripe berries. They indicated that the major components of the oils were α-pinene (58.61%–77.39%), camphene (0.67%–9.31%), δ-3-carene (0%–10.01%), and trans-verbenol (0%–5.24%). Also, Ennajar et al. (2009) analyzed the chemical composition of J. phoenicea plants collected from the southeastern region of Tunisia. They found that the main components were α-pinene (80.7%), δ-3-carene (4.5%), γ-cadinene (5.1%), β-caryophyllene (2.9%), β-myrcene (1.8%), and germacrene B (1.5%). Another study conducted by Bouyahyaoui et al. (2016) investigated the chemical composition of Algerian J. phoenicea EO from berries. They found that the main constituents were α-pinene (56.6%), α-terpineol, myrcene (2%), verbenone (1.9%), germacrene D (1.5%), β-caryophyllene (1.2%), camphor (1.9%), p-mentha-1,5-dien-8-ol (1.6%), terpinen-4-ol (1.1%), trans-pinocarveol (1.1%), α-phellandrene (1%), and β-pinene (1%). Harhour et al. (2018) analyzed the composition of the EOs of wild J. phoenicea from northern Algeria, including berries and branches. They found that the major components were α-p (40.3%), δ-3-carene (20.1%), α-cedrol (7%), α-terpinolene (4.5%), β-phellandrene (4.1%), β-myrcene (2.8%), α-terpinylacetate (2.7%), β-caryophyllene (1.9%), α-fenchene (1.9%), α-humulene (1.6%), β-pinene (1.5%), and elemol (1.2%).
The novelty of the present study is the enhancement of the chemical data from two non-polar crude extracts (cyclohexane and ethyl acetate). This information was obtained for the first time using GC-MS techniques to the best of our knowledge. The main purpose of this study was to characterize the chemical composition of the EO and non-polar extracts (cyclohexane and ethyl acetate) isolated from J. phoenicea berries. These berries were harvested in the Aflou region of Laghouat (Algerian Sahara). The study aims to assess their biological activities, including anti-diabetic effects (α-amylase activity) and antioxidative properties (DPPH, ABTS, and phenanthroline activities). Additionally, outcomes were further analyzed with molecular docking and absorption, distribution, metabolism, elimination, and toxicity (ADMET) studies. These analyses aimed to investigate the binding affinity of all compounds identified through GC-MS analysis to the active sites of α-amylase and α-glucosidase. Furthermore, the pharmacokinetic properties of each extract were determined through ADMET experiments. Ultimately, the data synthesized through these experiments provided insight into the best-docked molecules.
Ripe berries of Juniperus phoenicea were collected in March 2023 from the region of Aflou (Djebel Amour), located at the center of the Saharan Atlas and at the edge of the Sahara Desert (34°12' N; 2°10' E, 1300–1470 m) in the Laghouat province of southern Algeria. The plant’s identity was verified by the Department of Biological Sciences, Faculty of Science at Laghouat University in Algeria. Voucher specimens were deposited at the Research Unit of Medicinal Plant at the University of Laghouat, Algeria with the identification numbers URPM. Jp.03.23.
The J. phoenicea ripe berries were separated and placed on paper. The samples were dried in two steps: shade-drying and oven-drying.In the case of shade-drying, initially, 700 g of fresh sample was spread over 1 m2 of area and dried in a ventilated place at a temperature of 25 ± 2°C for 3 weeks. The same quantity was then transferred to an oven and dried for 4 days at a temperature of 50°C with 40% ventilation. The plant materials were ground using a basic grinder (IKA-10) with parameters of 3000rpm and a 1 mm sieve to get a homogenous powder.
To calculate the moisture content of the sample, 5 mg of ground fruit was placed in a porcelain mortar in the oven at a temperature of 105°C without ventilation for 24 h. After that, the moisture content (TH%) was determined to be 5.3%.
Juniperus phoenicea ripe berries (300 g) were ground into small pieces and subjectedto hydro-distillation for 3 h in a Clevenger-type apparatus, using 1 L of deionized water. The transparent oil was collected, dried over anhydrous Na2SO4, and stored at 4°C in dark sealed vials for further analysis.
Forty grams (40 g) of dried berry powder was divided into two equal parts (20 g each) and separately extracted by maceration with 100 mL of solvents (cyclohexane and ethyl-acetate) in a 1:5 ratio. This extraction process was repeated three times with fresh solvent each time. The mixture was then filtered through Whatman filter paper no.1. The filtrate was evaporated at 40°C using a rotary evaporator and stored in a refrigerator at 4°C for further analysis.
At the National Center for Biotechnology Research, we used a 96-well microplate reader (PerkinElmer Multimode Plate Reader EnSpire) to measure the activities of various substances. These substances included ascorbic acid, quercetin, α-tocopherol, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), Folin–Ciocalteu reagent (FCR), 1,1-diphenyl-2-picrylhydrazyl (DPPH), 2,20-azino-bis(3-ethylbenzothiazoline-6-sulfonicacid)-diammonium salt (ABTS), phenanthroline, pancreatic α-amylase enzyme (1U), acarbose, starch, sodium phosphate dibasic heptahydrate, sodium phosphate monobasic monohydrate, and hydrochloric acid, all obtained from Sigma-Aldrich. Additionally, we used sodium carbonate, aluminum chloride (AlCl3), iron (III) chloride (FeCl3), sodium bicarbonate (NaHCO3), potassium iodide (KI), potassium persulfate (K2S2O8), and potassium acetate (CH3CO2K), obtained from Biochem Chemopharma. All other chemicals and solvents used were of analytical grade.
The composition of J. phoenicea berries’ EO and crude extracts was determined using a gas chromatography-mass spectrometer system (GC-MS; GC-2010 Shimadzu Corporation, Kyoto, Japan) coupled to a mass spectrometer detector (QP-2020 A, Shimadzu). The GC–MS system utilized an Rxi®-5ms capillary column (Phase: Crossbond® 5% diphenyl/95% dimethyl polysiloxane) with dimensions of 30 m × 0.25 mm and a 0.25 µm film thickness. This column is similar in phase to HP-1ms, HP-1msUI, DB-1ms, DB-5ms, DB-1msUI, Ultra-1, VF-1ms, ZB-1, ZB-1ms and is considered equivalent to USP G1, G2, and G38 phases.
Analyses were conducted using helium (99.995% purity) as a carrier gas at a column flow rate of 1 mL/min with 1 µL of the sample injected in split mode at a ratio of 1:50. The oven program was as follows: injector and interface temperatures were maintained at 250°C and 310°C, respectively. The column temperature was initially set at 50°C fixed for 2 min, then increased to 310°C at a rate of 3°C/min, and then maintained at 310°C for 2 min.
The mass spectrometer conditions included an ionization voltage of 70 eV, ion source temperature of 200°C, and electron ionization mass spectra acquired over the mass range of 45–600 m/z. Component identification was determined by matching mass spectra with Wiley and NIST library data from 2017, standards of the main components, comparing Kovats Retention Indices (KRI) with reference libraries, and relevant literature (Adams, 2001; Barrero et al., 2005). Component concentrations were determined by semi-quantification through peak area integration from GC peaks and applying the correction factors of homologs series of C9-C10 n-alkanes and available authentic standards. For both EO and crude extract, a similar method was used, with only difference being in the sample volume and split mode (0.5 µL for the crude extract and a split ratio of 1:10).
The Folin–Ciocalteu method, slightly modified by Ostadi et al. (2020), was used to assess the total phenolic content, with results expressed in µg of gallic acid per mg of extract.
Following the study by Singleton et al. (1999), total flavonoids were calculated using the aluminum chloride method, with results presented in µg of quercetin per mg of extract.
Antioxidants exists in various classes and forms in plants, including flavonoids, coumarins, benzoic acid derivatives, phenolic compounds, and carotenoids. Multiple spectrophotometric methods are used to measure the total antioxidant capacity and content, providing a comprehensive profile of the antioxidant levels and capacity of the substances being studied. This study evaluated the antioxidant potential of EOs and non-polar extracts using six different methods, such as DPPH•, phenanthroline, and ABTS•+, compared to five standards: BHA, BHT, ascorbic acid, quercetin, and α-tocopherol.
The DPPH assay was determined using the methodology outlined by Moreno et al. (2000). The results were compared to positive standards including BHA, ascorbic acid, quercetin, α-tocopherol, and BHT, with absorbance readings taken at 517 nm. The concentration at which the DPPH radical could be 50% extinguished (IC50) was determined by plotting percentage inhibitions of extinction, as shown in Equation (1). The results indicated that the tested sample exhibited a greater capacity for scavenging free radicals, as evidenced by the decrease in absorbance of the solution:
where, Acontrol is the absorbance of blank and the Asample is that of extract or standard sample.
Oyaizu (1986) utilized the phenanthroline technique to determine the activity, with the findings reported as A0.50 (µg/mL). In order to compare the results obtained, positive standards such as BHA, ascorbic acid, quercetin, α-tocopherol, and BHT were used (Oyaizu, 1986).
The technique used to assess the ABTS•+ activity, and the findings were expressed as IC50, based on the work of Özyürek et al. (2012). BHA, ascorbic acid, quercetin, α-tocopherol, and BHT were utilized as positive standards for comparing the acquired findings.
This method was performed using the iodine/potassium iodide (IKI) method described by Zengin et al. (2014), with a few minor adjustments. A 96-well microplate was incubated for 10 min at 37°C with 50 μL of α-amylase solution (1U) prepared in phosphate buffer [PBS (pH 6.9 supplemented with 6 mM sodium chloride)] combined with 25 μL of each extract dissolved in MeOH, or acarbose as a positive control, at varying concentrations (6.25–400 μg/mL). Subsequently, 50 μL of 0.1% starch solution were added to initiate the reaction. The reaction mixture was then incubated at 37°C for 10 min. The reaction was stopped by adding HCl (25 μL, 1 M) and 100 μL of IKI solution. Similarly, a blank was prepared by adding the sample solution to all reagents without the enzyme α-amylase solution, and acarbose was used as a positive control. Absorbances were measured at 630 nm, with the absorbance of the blank subtracted from that of the samples (Zengin et al., 2014). The α-amylase inhibitory effect was calculated according to Equation (2):
where,
Ac: Absorbance of the mixture of starch + IKI + HCl + MeOH + phosphate buffer solutions
Ae: Absorbance of the mixture of the enzyme + starch + IKI + HCl+ MeOH solutions
As: Absorbance of the mixture of the enzyme + sample + starch + IKI + HCl solutions
Ab: Absorbance of the mixture of the sample + IKI + phosphate buffer solutions.
The crystallographic 3D structures of the studied enzymes: human pancreatic alpha-amylase in complex with montbretin A (PDB ID: 4W93) and sugar beet alpha-glucosidase with acarbose (PDB ID: 3W37) were retrieved from the Protein Data Bank website (http://www.rcsb.org) in “.PDB” format. The native ligands and water molecules were removed, and polar hydrogen atoms and Kollman charges were added using MGL-AutoDockTools (ADT, v1.5.7) (Morris et al., 2009). This procedure was applied to both proteins and then saved as the dockable protein databank extension “.pdbqt” for docking simulations.
The 3D structures of bioactive compounds, detected by GC-MS analysis, were retrieved from the PubChem compound database (https://pubchem.ncbi.nlm.nih.gov/) in “.SDF” format. The collected ligands were then converted to the “.pdbqt” format using AutoDock Tools for molecular docking simulation.
Autodock Vina was used for assessing binding affinity. The grid box centers, X, Y, and Z dimensions, used for the studied targets were (–9.632488, 4.340907, –23.107256) (20x20x20) and (0.108773, –1.916977, –23.053182) (20x20x20) for α-amylase (4W93) and α-glucosidase (3W37), respectively. The best binding pose with the lowest binding energy (kcal/mol) was chosen, and the interaction bonds were visualized in 3D and 2D images using Discovery Studio software. These parameters were applied to all studied compounds, and the top three interacted ones for each extract were graphically presented. The docking results were validated by redocking the native inhibitor into the enzyme’s active region (Trott and Olson, 2010).
The study of drug absorption, distribution, metabolism, excretion, and toxicity (ADMET) is known as pharmacokinetics. Eliminating poor drug candidates is a crucial concept that not only determines the availability of a drug but also helps to prevent issues during in vivo research (Cheng et al., 2012; Yang et al., 2019). In this study, ADMET characteristics were determined using ADMETSAR.
Hydrodistillation was used to extract the EO of J. phoenicea berries, allowing the extraction of 3.33% of the total mass of crude dry plant material., The yellow pale EO of J. phoenicea was compared to yields found in the literature, and Table 1 summarizes the different yields found. The chemical composition of the EO was analyzed by GC-MS, with a total of 50 compounds identified (Table 2) totalling 98%. Oxygenated monoterpenes were the most abundant components, despite the main components being derived from the monoterpenes hydrocarbons α-pinene (76.03%), β-pinene (2.75%), myrcene (1.73%), 3-carene (1.15%), and D-limonene (1.32%). Other compounds from sesquiterpene hydrocarbons such as caryophyllene (3.10%), α-humulene (1.25%), and germacrene D (1.7%) were also present. Most of the compounds identified in the EO of J. phoenicea berries in this study have been previously described for the same subspecies, although in different amounts.
Table 1. Yields and major chemical composition of different Juniperus phoenicea berries.
Region | Yield % | Major components | References |
---|---|---|---|
Algeria (Aflou) | 3.33 | α-Pinene (76.03%), Caryophyllene (3.10%) β-Pinene (2.7%), Myrcene (1.73%), Germacrene D (1.7%) |
This study |
Algeria (Naama) | 2 | α-Pinene (56.6%), α-Terpineol (3.3%) Camphor (1.9%), Verbanone (1.9%), p-Mentha-1,5-dien-8-ol (1.6%). |
Bouyahyaoui et al. (2016) |
Algeria (Mostaganem) | 0.14 | α-Pinene (43.7%), p-Cymene(5.8%) β-Phellandrene (4.6%), α-Terpineol (4.3%), Germacrene D (1.7%). |
Abdelli et al. (2018) |
Algeria (Bouira) | 2.7 | α-Pinene (80.80%), Caryophyllene oxide (3.9%), 3-Carene (1.90%) , β-Pinene (1.8%), α-Humulene oxide (1.6%) | Menaceur et al. (2013) |
Algeria (Ain Defla) | 1.1 | α-Pinene (40.30%), 3-Carene (20.10%), α-Terpinolene (4.5%) α-Cedrol (7%), β-Phellandrene (4.1%) |
Harhour et al. (2018) |
Italy(Sardinia) | 2.54 | α-Pinene (87.54%), 3-Carene(1.23%)β-Phellandrene (2.81%), Myrcene (1.61%), Germacrene D (1.17%). | Angioni et al. (2003) |
Tunisia (Matmata) | Shade drying (2.82%)Sun drying (3.40%) | May (Shade drying): α-Pinene (26.9%), γ-Cadinene(10.2%), Caryophyllene oxide (6.3%), β-Selinene (6.9%), Germacrene B(7.3%).May (Sun drying): α-Pinene (86.4%), γ-Cadinene(4.6%), β-Myrcene (2.2%), γ-Terpinene (1.6%), Germacrene B (1.1%) | Ennajar et al. (2010) |
Tunisia (1-Rimel, 2-Makthar, 3-J.Mansour) | 2–7 | 1− α-Pinene (63.40%), Caryophyllene oxide (3.5%), 3-Carene (10%), Trans-verbenol (2.40%), β-Pinene (2%)2- α-Pinene (69.50%), β-Pinene (2.1%), Camphene (9.3%), Caryophyllene (1.8%), Myrcene (2.1%)3- α-Pinene (77.3%), β-Pinene (2.5%), β-3-Carene (3.7%), β-phellandrene (2.5%), Trans-verbenol (1.60%) | Medini et al. (2011) |
Morocco (Tounfite) | 2.01 | α-Pinene (78.11%), β-Pinene (0.95%), β-Trans-Dauca-4(11),7-diene (2.96%), Caryophyllene (2.77%), Germacrene D (5.42%) | Amalich et al. (2015) |
Table 2. Antioxidant activities with phenolic and flavonoid content of nonpolar extracts and essential oil of Juniperus phoenicea.
Extracts | DPPH. Assay IC50(µg/L) |
Phenoltroline | ABTS.+Assay | Total Phenolics | Total Flavonoids |
---|---|---|---|---|---|
Essentiel Oil | In | In | In | – | – |
Cyclohexane | >800 | >800 | >800 | 19.39 ± 0.61 | 10.96 ± 1.38 |
Ethyl acetate | >800 | >800 | 378.28 ± 5.18 | 20.86 ± 2.44 | 12.93 ± 0.83 |
BHA | 9.11 ± 0.89c | 1.49 ± 0.08ab | 2.98 ± 0.11ab | NT | NT |
BHT | 1.60 ± 0.36a | 2.20 ± 0.04b | 1.31 ± 0.06a | NT | NT |
α-Tocopherol | 19.99 ± 0.74d | 5.78 ± 0.30c | 10.54 ± 0.07b | NT | NT |
Quercetine | 3.40 ± 0.30b | 0.65 ± 0.04a | 2.50 ± 0.06a | NT | NT |
Ascorbic acid | 2.69 ± 0.22b | 8.30 ± 0.76d | 4.04 ± 0.02ab | NT | NT |
The concentration at 50% inhibition and the concentration at 0.50 absorbance, respectively, are referred to IC50 and A0.50 values. By using a linear regression analysis, the IC50 and A0.50 values were determined and expressed as mean ± SD (n = 3). The values in the same columns that have different superscripts (a, b, c, d, e, f, g, or h) differ significantly (p < 0.05).
BHA: utylated hydroxyanisole, BHT: butylated hydroxytoluene, b: reference compounds, BHA: butylated hydroxyanisole, BHT: butylated hydroxytoluene, Nd: not determined, NT: nottested,
*total phenolics (µg gallic acid/mg extract), **total flavonoids (µg quercetin/mg extract)
Statistical study was made in term of n = 3.
The berries used in this study were in the same stage of maturation as those reported by the previous authors. α-Pinene predominated in all examined EOs, with levels ranging from 27% to 87.5%, depending on the local, harvesting season, and amount of plant material. As mentioned in the literature, the phoenicea group of juniper significantly supported these findings. These results are in agreement with those reported by research communities in this field, especially those focusing on this genre in Algeria. The majority of compounds remain the same, with variationsmainly in percentage. Slight differences were reported for some compounds like Dauca-4(11),7-diene that were reported in a study conducted by Amalich et al. (2015).
In the present study, and for the first time to our knowledge, we provide a report on the phytochemical composition of cyclohexane and ethyl acetate extracts from Phoenician juniper. According to a study by Byrne et al. (2016) which categorized green solvents for extractions, separations, formulations, and reaction chemistry, the two solvents selected for this study were the most appropriate based on environmental, health, and safety (EHS) criteria as outlined by the three big pharmaceutical companies Pfizer, GlaxoSmithKline, and Sanofi (Byrne et al., 2016).
Classical maceration was used for 3 days to prepare the crude extract of non-polar solvents (cyclohexane and ethyl acetate) from Phoenician juniper berries. This process allowed the extraction of 7.95% and 8.7,% respectively, of the total mass of the crude dry plant material. The variation in yield was attributed to the polarities of the different compounds in the berries, a variation that has been noted in the literature, as seen in the study by Ennajar et al. (2009). The total flavonoid content was reported as micrograms of quercetin equivalents per milligram of extract (µg QE/mg) and the total phenolic content was reported as micrograms of gallic acid equivalents per milligram of extract (µg GAE/mg). The results are summarized in Table 2.
The chemical composition of cyclohexane and ethyl acetate extracts was analyzed using the same GC-MS. A total of 89 and 82 compounds were identified (Table 3), with a total percentage of identification of 67% and 68%, respectively. Despite the EO, the most abundant components for the non-polar extract are the oxygenated diterpenes, with amounts of 21.34% and 21.95% respectively. The analysis reveals the appearance of new compounds such as sterol (gamma.-Sitosterol), lignan (Anthricin), ether (Methyl tetratriacontyl ether) and ester (Acetic acid, 2,6,6-trimethyl-3-methylene-7-(3-oxobutylidene)oxepan-2-yl ester). For both extracts (cyclohexane and ethyl acetate), the main components were derived from the oxygenated diterpenes such as communic acid (23.66% and 22.38%), abitale (110.75% and 13.22%), abitole (4.25% and 4.60%), and abieta-7,13-diene-3-one (4.02% and 4.55%), respectively. Despite the existence of α-pinene with significant amounts for both extracts with percentages of 13.01% and 16.21 %, which proves the presence of J. phoenicea. The GC-MS analysis of the non-polar extracts reveals chemical variations in the content compared with the EO, noting the presence of diterpene hydrocarbons, oxygenated diterpenes in significant amounts, and sesquiterpene oxygen, which is present in significant quantities as in the EO. The total ion chromatogram of representative samples in which the main compounds are pointed out is displayed in Figure 1. The complete EO composition is shown in Table 2.
Table 3. Chemical composition of Juniperus phoenicea berries essential oils and nonpolar extracts (cyclohexane and ethylacetate); molecular docking score for both enzymes (α-amylase and α-glucosidase).
No | Compounds name | RT (min) |
RIE/RIT | EO Area % |
Extract 1 Area % |
Extract 2 Area% |
Docking scoresa kKcal/mol |
Docking scoresb kcal/mol |
---|---|---|---|---|---|---|---|---|
01 | Tricyclenea | 8.19 | 916/921 | 0.21 | – | – | –5.4 | –5.7 |
02 | α-Thujenea | 8.41 | 921/924 | 0.04 | – | – | –5.9 | –6.2 |
03 | α-Pinenea | 8.78 | 931/932 | 76.03 | 13.01 | 16.21 | –5.4 | –5.9 |
04 | Camphenea | 9.2 | 941/942 | 0.72 | 0.07 | 0.09 | –5.5 | –4.6 |
05 | Thuja-2,4(10)-dienea | 9.42 | 947/953 | 0.30 | – | – | –5.9 | –6.2 |
06 | Sabinena | 10.202 | 967/969 | 0.16 | – | – | –5.9 | –6.3 |
07 | β-Pinenea | 10.31 | 969/974 | 2.75 | 0.67 | 0.75 | –5.4 | –5.8 |
08 | Myrcenea | 10.94 | 985/988 | 1.73 | 0.37 | – | –4.7 | –5.7 |
09 | 3-Carenea | 11.73 | 1004/1008 | 1.15 | 0.87 | 1.02 | –5.6 | –6.8 |
10 | ρ-Cymenea | 12.35 | 1018/1020 | 0.46 | – | 0.09 | –5.9 | –6.2 |
11 | D-Limonenea | 12.54 | 1022/1024 | 1.32 | 0.36 | 0.36 | –5.7 | –6.0 |
12 | Terpinolenea | 15.25 | 1082/1086 | 0.06 | – | – | –5.6 | –5.9 |
13 | α-Pinene oxideb | 15.66 | 1091/1099 | 0.1 | – | – | –5.6 | –4.7 |
14 | Trans-Verbenolb | 16.037 | 1120/1122 | – | 0.40 | – | –5.8 | –4.9 |
15 | α-Campholenalb | 16.95 | 1119/1122 | 0.38 | 0.40 | 0.17 | –5.5 | –5.0 |
16 | Trans-Pinocrveolb | 17.55 | 1132/1135 | 0.67 | 0.52 | 0.52 | –5.9 | –6.2 |
17 | Cis-Verbenolb | 17.65 | 1135/1137 | 0.06 | 0.27 | 0.79 | –5.8 | –6.2 |
18 | Camphorb | 17.75 | 1137/1141 | 0.82 | – | – | –5.4 | –4.6 |
19 | p-Menth-3-en-8-olb | 18.01 | 1142/1145 | 0.09 | – | – | –5.7 | –6.3 |
20 | Trans-Pinocamphoneb | 18.53 | 1154/1158 | 0.06 | – | – | –5.7 | –5.3 |
21 | Borneolb | 18.78 | 1159/1165 | 0.09 | – | – | –5.8 | –4.8 |
22 | p-Mentha-1,5-dien-8-olb | 18.87 | 1161/1166 | 0.04 | – | – | –6.1 | –6.4 |
23 | Terpinen-4-olb | 19.34 | 1171/1174 | 0.22 | – | – | –5.8 | –6.1 |
24 | ρ-Cymen-8-olb | 19.71 | 1179/1176 | 0.11 | – | – | –5.8 | –6.3 |
25 | α-Terpineolb | 19.96 | 1185/1186 | 0.42 | 0.27 | 0.24 | –5.8 | –6.3 |
26 | Myrtenolb | 20.22 | 1190/1194 | 0.19 | – | – | –5.6 | –5.0 |
27 | ρ-allyl- Anisolee | 20.366 | 1193/1195 | 0.08 | – | – | –5.5 | –5.7 |
28 | Verbenoneb | 20.79 | 1202/1204 | 0.14 | – | – | –5.7 | –5.6 |
29 | Bornyl acetateb | 24.323 | 1280/1254 | 0.25 | – | – | –6.0 | –5.3 |
30 | Verbenyl acetateb | 24.78 | 1292/1291 | 0.19 | – | – | –6.0 | –5.2 |
31 | δ−Elemenec | 26.63 | 1333/1135 | 0.07 | – | – | –6.7 | –6.0 |
32 | α-Terpinyl acetateb | 27.13 | 1344/1346 | 0.56 | 0.64 | 0.57 | –6.2 | –6.9 |
33 | α-Copaenec | 28.301 | 1371/1374 | 0.06 | – | – | –7.1 | –6.8 |
34 | β−Elemenec | 28.995 | 1387/1389 | 0.52 | 0.42 | 0.35 | –6.8 | –5.8 |
35 | Cedrenec | 29.407 | 1396/1410 | 0.21 | – | – | –7.7 | –5.6 |
36 | Caryophyllenec | 30.15 | 1414/1417 | 3.10 | 1.62 | 1.45 | –7.4 | –6.0 |
37 | γ-Elemenec | 30.73 | 1428/1434 | 0.10 | 0.18 | 0.14 | –6.5 | –5.4 |
38 | α-Humulenec | 31.563 | 1448/1452 | 1.25 | 0.75 | 0.68 | –7.1 | –6.0 |
39 | γ-Muurolenec | 32.51 | 1471/1478 | 0.30 | 0.22 | 0.22 | –7.2 | –7.1 |
40 | Germacrene Dc | 32.698 | 1475/1484 | 1.70 | 1.05 | 0.74 | –7.4 | –6.4 |
41 | β-Selinenec | 32.89 | 1481/1489 | 0.17 | 0.14 | 0.10 | –7.4 | –6.2 |
42 | trans-Muurola-4(15), 5-dienec | 33.12 | 1486/1493 | 0.10 | – | – | –6.9 | –6.7 |
43 | Valencenec | 33.25 | 1489/1496 | 0.18 | – | – | –7.1 | –7.3 |
44 | α-Muurolenec | 33.47 | 1494/1500 | 0.30 | 0.26 | 0.20 | –7.1 | –5.7 |
45 | δ-Amorphenec | 34.02 | 1508/1511 | 0.13 | – | – | –7.0 | –6.7 |
46 | Cubebolc | – | 0.18 | 0.31 | –7.4 | –7.3 | ||
47 | δ-Cadinenec | 34.39 | 1518/1522 | 0.80 | 0.45 | 0.53 | –7.0 | –6.8 |
48 | Elemold | 35.387 | 1543/1548 | 0.13 | 0.59 | 0.49 | –6.7 | –5.9 |
49 | Germacrene Bd | 35.703 | 1551/1559 | 0.72 | 0.34 | 0.26 | –7.4 | –5.8 |
50 | Caryophyllene oxided | 36.697 | 1577/1582 | 0.53 | 1.64 | 1.64 | –7.1 | –5.8 |
51 | Salvial-4(14)-en-1-oned | 37.140 | 1588/1594 | – | 0.17 | 0.12 | –7.2 | –5.5 |
52 | Humulene epoxide IId | 37.705 | 1603/1608 | 0.12 | 0.71 | 0.63 | –7.5 | –5.8 |
53 | 1-epi-Cubenold | 38.444 | 1623/1641 | 0.08 | – | – | –7.0 | –6.0 |
54 | Isospathulenold | 38.45 | 1623/1630 | – | 0.71 | – | –6.9 | –6.3 |
55 | Aromadendrene epoxided | 39.101 | 1640/1641 | – | 0.16 | 0.14 | –9.1 | –7.5 |
56 | β-Eudesmold | 39.247 | 1644/1650 | – | 0.17 | 0.14 | –8.5 | –7.8 |
57 | Eudesma-4,11-dien-2-old | 39.36 | 1648/nd | – | 1.19 | 0.99 | –7.4 | –6.5 |
58 | Schyobunold | 40.737 | 1684/1688 | – | 0.24 | 0.16 | –6.5 | –5.1 |
59 | 3-Isopropyl-6,7-dimethyltricyclo[4.4.0.0(2,8)] decane-9,10-diole |
41.13 | 1695/1710 | – | 0.25 | – | –8.9 | –7.2 |
60 | Oplopanoned | 42.40 | 1731/1739 | – | 0.54 | 0.32 | –6.9 | –5.6 |
61 | Acetic acid, 2,6,6-trimethyl-3-methylene-7-(3-oxo butylidene)oxepan-2-yl estere | 43.47 | 1762/nd | – | – | 0.18 | –6.8 | –5.8 |
62 | α-Vetivold | 43.49 | 1762/nd | – | 0.30 | – | –7.5 | –6.3 |
63 | Platambind | 46.48 | 1848/nd | – | 0.20 | – | –6.8 | –6.1 |
64 | Manool oxideg | 50.91 | 1984/1987 | – | – | 0.25 | –8.1 | –6.8 |
65 | Abitatrienef | 52.90 | 2047/2055 | – | 0.27 | 0.19 | –9.0 | –8.5 |
66 | Abitadienef | 53.675 | 2071/2087 | – | 0.95 | 0.75 | –9.0 | –8.2 |
67 | 19-norabieta-8,11,13-trieneg | 56.41 | 2164/nd | – | 0.89 | 0.70 | –8.5 | –7.2 |
68 | 4-Hydroxy-18 -nor-abietaneg | 56.60 | 2170/nd | – | 0.27 | – | –8.5 | –7.8 |
69 | Methyl abietateg | 57.628 | 2205/2385 | – | – | 1.89 | –8.4 | –8.0 |
70 | Sandaracopimarinalg | 56.701 | 2174/2184 | – | 0.31 | 0.22 | –8.4 | –7.2 |
71 | Palustraleg | 57.846 | 2213/nd | – | – | 0.88 | –8.5 | –7.5 |
72 | Methyl levopimarateg | 57.628 | 2205/2305 | – | 2.18 | – | –8.7 | –7.6 |
73 | Levopimarinalg | 58.051 | 2220/nd | – | 0.37 | 0.88 | –8.8 | –7.5 |
74 | Androst-5-ene-3, 17-diol, 3-acetate,(3.β.,17.β.)e | 58.56 | 2239/nd | – | 0.17 | – | –10.0 | –8.9 |
75 | Dehydro Abietalg | 58.854 | 2249/2274 | – | 0.70 | 0.72 | –8.7 | –7.9 |
76 | 3-α-hydroxy Manoolg | 59.015 | 2255/2297 | – | 1.03 | 0.90 | –9.0 | –8.2 |
77 | Abieta-8,11,13-trien- 7-oneg | 59.1 | 2257/nd | – | 0.63 | – | –9.1 | –8.0 |
78 | Abietalg | 59.788 | 2282/2313 | – | 10.75 | 13.22 | –8.9 | –7.9 |
79 | Abieta-7,13-dien-3-oneg | 60.16 | 2295/nd | – | 4.02 | 4.55 | –8.9 | –7.3 |
80 | 4-Epiabietolg | 60.36 | 2302/2343 | – | 0.26 | – | –8.5 | –7.7 |
81 | Verticiolg | 60.66 | 2313/nd | – | 0.58 | – | –8.6 | –6.4 |
82 | Dihydro Abietolg | 60.354 | 2302/2368 | – | 0.58 | 0.23 | –8.7 | –8.0 |
83 | Neoabietyl acetateg | 61.075 | 2328/nd | – | 0.51 | 1.24 | –9.0 | –8.6 |
84 | 4-epi- dehydroabietolg | 61.504 | 2344/nd | – | 1.22 | 1.33 | –8.5 | –6.6 |
85 | Communic acidg | 61.81 | 2355/2365 | – | 23.66 | 22.38 | –8.4 | –7.3 |
86 | Bis(2-methoxy phenyl) (isopropoxy) methanee | 62.11 | 2366/nd | – | 2.03 | – | –6.5 | –6.3 |
87 | Abieta-8(14),13(15)-dien-18-alf | 62.114 | 2367/nd | – | – | 1.32 | –8.9 | –7.8 |
88 | Abietolg | 62.447 | 2379/2401 | – | 4.25 | 4.60 | –8.8 | –7.8 |
89 | 16-Norisopimar-7-en- 15-olg | 63.1 | 2402/nd | – | 0.83 | – | –8.6 | –6.9 |
90 | Abietic acidg | 63.798 | 2430/nd | – | – | 1.95 | –8.9 | –7.8 |
91 | Abietateg | 63.87 | 2433/nd | – | 1.07 | – | –8.9 | –7.9 |
92 | Neoabietolg | 64.267 | 2448/2468 | – | – | 0.25 | –9.2 | –8.2 |
93 | Methyl tetratriacontyl ethere | 79.21 | 3084/nd | – | 0.32 | 0.45 | –5.7 | –5.4 |
94 | Anthricine | 82.347 | 3237/nd | – | 0.51 | 0.25 | –8.0 | –7.6 |
95 | Tetrapentacontanee | 83.12 | 3275/nd | – | 0.15 | – | –5.2 | –5.7 |
96 | γ-Sitosterolé | 83.50 | 3295/nd | – | 0.22 | 0.36 | –9.3 | –8.5 |
Monoterpene hydrocarbons – 23.52%; 6.74%; 7.31%
Oxygenated monoterpens – 33.33%; 6.74%; 6.09%
Sesqueterpene hydrocarbons – 29.41%; 10.11%; 12.19%
Oxygenated sesqueterpens – 9.80%; 14.60%; 12.19%
Non-terpene derivative – 1.96%; 6.74%; 4.87%
Diterpene hydrocarbons – 2.25%; 3.65%
Oxygenated diterpenes –21.34%; 21.95%
Totale identified (%) – 98%; 67%; 68%
Not identified (%) – 2%; 32%; 32%
EO: Essential oil; nd: non–determinated; E: Experimentale; T: Theorical;1: Cyclohexane extract; 2: Ethyl acetate extract; a: α-amylase docking scores; b: α-glucosidase docking scores.
Figure 1. GC-MS total ion chromatogrammes from a representative sample of berries of Juniperus phoenicea L. (A) Essential oil. (B) Cyclohexane extract. (C) Ethyl acetate extract.
The preliminary study examined the chemical composition, antioxidants, antidiabetic effects, and in silico modeling of the EO and non-polar extracts of Algerian J. phoenicea berries. The results of this study showed that the EO was characterized by α-pinene, caryophyllene, β-pinene, myrcene, and germacrene D. These results, although slightly different in profile from earlier research, are generally consistent with previous findings (Amalich et al., 2015; Angioni et al., 2003; Medini et al., 2011; Menaceur et al., 2013, Abdelli et al. 2018). The minor differences in EO composition could be attributed to genotype-environment interactions and the phenological stage of the species under study. Plants may produce certain chemical biomolecules as an adaptive response to changing environmental conditions. The results also indicated that the principal elements mentioned above were present in the non-polar extracts. While there were no previous studies mentioning the composition of non-polar extracts, especially for cyclohexane and ethyl acetate, the current outcome was consistent with the literature. The main constituents of most J. phoenicea species include abitale, abitole abieta-7,13-diene-3-one, and α-pinene (Boudiba et al., 2021 Bouyahyaoui et al., 2016; Ennajar et al., 2010).
According to literature data, extracts such as ethyl acetate, ethanol, and methanol solvents had the highest content of total phenols and flavonoids (Bouyahyaoui et al., 2016). This could be explained by the fact that polar solvents have a higher affinity for these components than non-polar solvents. On the other hand, variations in phenolic content might be attributed to different extraction methods, the standard solution used, geographical location, and climate conditions. Overall, the study reported that total phenolic and total flavonoids levels were relatively low.
A total of three in vitro antioxidant tests (DPPH, ABTS, and phenanthroline) were conducted to compare with five standards: BHA, BHT, ascorbic acid, quercetin, and α-tocopherol. The results were obtained using linear regression analysis, and the IC50 and A0.5 values are presented in Table 3 as the mean values ± SD of three measurements. In most tests, the IC50 and A0.50 values differed significantly from the standard ones.
Antioxidants operate through two distinct processes: electron transfer (single electron transfer, SET) and hydrogen donation (hydrogen atom transfer, or HAT). Our investigation utilized several techniques (DPPH•, ABTS•+, and phenanthroline) to gather more data on the antioxidant activity of the examined extracts. The differences between these tests lie in their modes of action and reaction mechanisms.
In the DPPH assay, all samples showed lower activity (IC50 > 800 g/mL) compared to the standards. The lower activity exhibited by all samples was likely due to the high concentration of terpene hydrocarbons (53%) in the EO. Similarly, the non-polar extracts showed lower activity despite containing a high concentration of oxygenated terpene (40.23%–42.68%). This can be attributed to the fact that the inhibitory activity of the DPPH radical is not solely dependent on the total polyphenol content, but rather on specific polyphenols with unique chemical structures (Sánchez-Vioque et al., 2013). In other words, DPPH is highly selective and only reacts with flavonoids that contain hydroxyl groups in the B cycle (Yokozawa et al., 1998), as well as aromatic acids with multiple hydroxyl groups, which were not observed in our case. These results were in agreement with existing literature on similar species, such as the antioxidant activity studies conducted by Ennajar et al. (2009) and Taviano et al. (2011) on methanol and water extracts of five different Turkish Juniperus species, which showed weaker activity compared to the extracts examined in this study (Ennajar et al., 2009; Taviano et al., 2011). On the other hand, compared to the polar extract case by Ghouti et al. (2018) where hydroethanolic and infusion extracts were used, our study demonstrated interesting DPPH activity that was two to three times more effective than Trolox. Additionally, research by Keskes et al. (2014) reported that the methanol extract exhibited a stronger ability to scavenge DPPH radicals (EC50 value = 2 μg/mL).
Regarding the phenanthroline assay, the EO and non-polar extracts of J. phoenicea showed a weak ability to reduce iron with A0.50 value of >200 µg/mL for the high concentration. This is in contrast to the results of standards such as ascorbic acid, quercetin, BHA, BHT, and α-tocopherol, which demonstrated a substantial reducing impact on iron. Our results indicated a low absorbance for both extracts (200 μg/L – 0.35), which is comparable to findings in the literature (Byrne et al., 2016; Menaceur et al., 2013). The phenolic content of the extracts was closely correlated with their poor activity in the DPPH radical scavenging and phenanthroline tests. Numerous investigations have shown a strong positive association between total phenolic content, phenanthroline, and anti-DPPH action.
In the ABTS assay, it was observed that the extracts of J. phoenicea berries exhibited a stronger free radical scavenging capacity compared to DPPH for all samples. This can be attributed to the versatility of the radical cation ABTS, which is soluble in both water and organic solvents. This allows evaluation of anti-radical activity for both hydrophilic and lipophilic compounds. Additionally, the planar form of ABTS makes it more susceptible to attack compared to DPPH, which is more complex (Gülçin, 2010). When compared to the five positive standards, ABTS still showed minimal activity. The percentage of ABTS-scavenging ability of the EO from Tunisian J. pheonicea berries was determined to be 128.7 ± 3.8 μg/mL compared to vitamin C at 1.9 ± 0.9 μg/mL. These results were slightly lower than previous investigations. Despite their low concentration, no correlation was found between phenolic compounds and ABTS•+ activity. Certain monoterpene alcohols, ethers, ketones, aldehydes, and non-polar bioactive substances like pigments, were associated with ABTS•+ activity (Edris, 2007; Merola et al., 2017).
In general, the antioxidant capacity of both EO and non-polar extracts (cyclohexane and ethyl acetate) of J. phoenicea berries was tested in three different assays (DPPH, ABTS, and phenanthroline). It can be concluded that the EO exhibits no antioxidant activity despite increases in concentration. This could be explained by the complex mixture of EOs that may exhibit antioxidant activity different from that of their major components tested alone. Literature reports suggest that the antioxidant activity of EO may result from a complex interaction between different constituents, producing additive, synergistic, or antagonistic effects, even at low concentrations (Ennajar et al., 2010). The non-polar extracts show some antioxidant activities but are still insignificant compared to five standards. This could be due to the low phenolic content and the mixture of extracts preventing the reduction of both DPPH and ABTS, and even forming a complex with phenanthroline for reducing the ferric ion. Table 3 presents the different values obtained in the antioxidant activities.
One of the most crucial approaches to treating type 2 diabetes (T2D) is controlling postprandial hyperglycemia. This is achieved by inhibiting the enzymes that hydrolyze carbohydrates, primarily intestinal α-glucosidase (EC 3.2.1.21.), and pancreatic α-amylase (EC 3.2.1.1.1.). This process may reduce blood levels of glucose in circulation by delaying the gastrointestinal absorption of dietary carbohydrates (Al-Ishaq et al., 2019; Deo et al., 2016; Priscilla et al., 2014). Due to these factors, the present investigation examined our plant’s potential as an antidiabetic by evaluating its inhibitory effect on the enzymatic activity of α-amylase. The primary enzyme in humans, α-amylase, breaks down starch into simple sugars such as dextrin, maltotriose, maltose, and glucose. α-amylase inhibitors are believed to enhance glucose tolerance in diabetic patients, although the activity of the enzyme has not been directly linked to the genesis of diabetes. To improve diabetes management, great effort has been put forward in recent years to discover α-amylase inhibitors that are clinically effective (Loizzo et al., 2007). The results are expressed as mean ± SD and provided in Table 4.
Table 4. Anti-diabetic activities (α-amylase ) of Juniperus phoenicea berries essential oil and non-polar extracts.
Extracts/standard | α-amylase | |
---|---|---|
Max inhibitory % | IC50(μg/mL) | |
Essential oil | 33.47 ± 2.58 | >1600d |
Cyclohexane | 69.59 ± 0.07 | 186.91 ± 5.74d |
Ethyl acetate | 88.71 ± 0.47 | 351.48 ± 0.17d |
Acarbose | 53.05 ± 1.6 | 3650.9 ± 1.7d |
Inhibition rates and IC50 values are expressed as means ±S.D (n = 3). Mean values followed by different letters are significantly different (based on one-way ANOVA followed by Tukey’s multiple comparison tests, Level of Significance: p < 0.0001.
As demonstrated by the results obtained, both extracts showed a strong dose-dependent inhibitory effect against α-amylase, with maximum inhibition rates >75.0% at the high tested dose of 400 μg/mL. Table 4 shows the significant increase in the α-amylase inhibitory effect with an IC50 of 186.91 ± 5.74 μg/mLfor the cyclohexane extract compared to 351.48 ± 0.17 μg/mL for the ethyl acetate extract. This difference is nearly 20 times greater than that of acarbose, used as a positive control. Interestingly, acarbose’s inhibitory efficacy against α-amylase is about 15 times less (3650.9 ± 1.7 μg/mL) than the ethyl acetate extract. However, pure quercetin demonstrated effective inhibition of α-amylase with the lowest IC50 value of 4.3 ± 0.2 μg/mL, surpassing the activity observed in our investigation against α-amylase. These results suggest the strong anti-diabetic properties of both non-polar extracts (cyclohexane and ethyl acetate) of J. phoenicea berries. In contrast, the EO showed a maximum inhibition of 33% at a concentration of 16 mg/L, providing a reliable value compared to the extracts.
Regrettably, the literature lacked significant information about the potential inhibitory effects of Juniperus species against α-amylase, while few reports on J. phoenicea berries’ α-amylase inhibitory activities were published. Interestingly, Loizzo et al. (2007) investigated the α-amylase inhibitory activities of J. oxycedrus berries EO. The bio-assay was adopted and modified based on the work of Conforti et al. (2005). Their results indicated moderate activities of berries EO with IC50 value of >25 µL/mL (Loizzo et al., 2007). In the same context, the work of Orhan et al. (2012) evaluated the hypoglycemic and antidiabetic activity of J. oxycedrus subsp . Oxycedrus berries. They studied the oral administration of berry extracts using in vivo models in normal and glucose-hyperglycemic rats. They indicated that ethanol extract showed a higher and continuous hypoglycemic effect (7.7%–23.6%) in STZ-diabetic rats. For normal rats, the ethanol extract exhibited a moderate hypoglycemic effect (12.8%–13.0%). The n-ButOH extract gives better results, especially for fraction C, which exhibited the most effective antidiabetic activity compared with others between 18.6% and 26.4%. This activity was caused by the presence of bioactive compounds such as shikimic acid (Orhan et al., 2012).
Banerjee et al. (2013) studied the impact of a methanolic extract of J. communis (100 mg/kg, 200 mg/kg) administered orally to streptozotocin nicotinamide-induced diabetic rats. They found that the extract had dose-dependent and significant anti-diabetic and antihyperlipidemic properties, supporting its potential as an effective drug for type-2 diabetes. In another study conducted by Sánchez de Medina et al. (1994) and Swanston-Flatt et al. (1990), it was shown that decoction and infusion of J. communis fruits led to significant decrease in blood glucose levels in both normoglycemic rats and STZ-induced diabetic rats and mice.
Overall, research on the antidiabetic effects of Juniperus species, both in vitro and in vivo, suggests strong inhibitory activity against α-amylase and α-glucosidase. These findings have the potential to improve hyperglycemia in individuals with type 2 diabetics, highlighting the need for further investigation into the mechanisms of action and active components of these plants.
Molecular docking is an increasingly common method in structure-based drug design (SBDD) because it can accurately predict the binding conformation of small molecules on their corresponding target proteins (Wang et al., 2020). This technique facilitates the analysis and visualization of significant intermolecular biological processes, including the binding of ligands to their corresponding receptors and interactions that maintain the stability of the ligand-receptor system. Furthermore, it is possible to rank the “docked” conformations of the simulated ligands by using their binding free energies since molecular docking techniques quantify receptor-ligand binding energies (Huang and Zou, 2010; Kadi et al., 2023).
To understand the binding efficacy of compounds from J. phoenicea L. berries’ EOl and non-polar extracts (cyclohexane and ethyl acetate) to the selected proteins associated with diabetes (α-amylase 4W93 and α-glucosidase 3W37), molecular docking simulations were conducted using Autodock Vina. The native ligands montbretin A and acarbose were used for comparison, and their binding energies were -11.0 and -10.0 kcal/mol, respectively. Among those results, binding energy values varied from -4.6 to -10.0 kcal/mol. The best result in this range was measured in terms of the score value of the native ligand, and the three best compounds from each extract composition were selected.
Our findings showed potent molecules such as Humulene epoxide II, Caryophyllene, and cedrene against α-amylase, valencene, γ-muurolene, and δ-cadinene for α-glucosidase in the EO composition. In the cyclohexane extract, the most potent ones were androst-5-ene-3,17-diol, 3-acetate (3.β.,17.β.), γ-sitosterol and abieta-8,11,13-trien-7-one, for α-amylase and androst-5-ene-3,17-diol,3-acetate (3.β.,17.β.), neoabietyl acetate, and γ-Sitosterol for α-glucosidase. In the ethyl acetate extract, γ-Sitosterol, Neoabietol, and Aromadendrene epoxide showed a potent affinity against α-amylase while abitatriene, γ-sitosterol, and neoabietol for α-glucosidase.
Major compounds for each extract showed good results but this varied for each protein, possibly due to the amino acid construction of each protein. Although there have been fluctuations in this range, the result of diterpene and sesquiterpene is still high compared to monoterpene, reflecting the fact that their structure has many hydroxyl groups that facilitate ligands in forming hydrogen bonds with a free residue of the receptor. Additionally, Table 4 shows the best receptor of these bioactive compounds in J. phoenicea L. berries, α-amylase 4W93 followed by α-glucosidase 3W37. Interaction analysis in 3D and 2D images was performed to understand the binding mode of the studied compounds in interaction with α-amylase 4W93 and α-glucosidase 3W37. Figures 2 and 3 show the different interactions.
Figure 2. The molecular interactions in 2D and 3D images between the studied compounds and the main α-amylase 3W93 using Autodock Vina. Essentail oil: (A) Cedrene, (B) Caryophyllene, (C) Humulene epoxide II. Cyclohexane extract: (D) Androst-5-ene-3,17-diol, 3-acetate, (E) g-Sitosterol, (F) Abieta-8,11,13-trien-7-one. Ethyl acetate extract: (G) g-Sitosterol, (H) Neoabietol, (I) Aromadendrene epoxide.
Figure 3. The molecular interactions in 2D and 3D images between the studied compounds and the main α-glucosidase 4W37 using Autodock Vina. Essential oil: (A) γ-Muurolene, (B) δ-Cadinene, (C) Valencene. Cyclohexane extract : (D) Androst-5-ene-3,17-diol, 3-acetate, (E) Neoabietyl acetate, (F) γ-Sitosterol. Ethyl acetate extract: (G) Abitatriene, (H) Neoabietyl acetate, (I) γ-Sitosterol.
Our results showed that in the case of ethyl acetate extract, γ-Sitosterol is the most interactive ligand with α-amylase 4W93. This interaction is facilitated through two positive ionizable interactions formed with the catalytic triad of hydrogen-donor, with ASP197 and GLU233, five interactions (one π-sigma and four π-alkyl) with TRP59, and one alkyl interaction observed with LEU162. On the other hand, in the case of α-glucosidase 3W37, the most interactive ligand was abitatriene. This interaction involved three interactions with the first ring, one π-anion with ASP568, one intriguing π-sulfur interaction with MET470, and one π-π T-shaped interaction with TRP432.
For the cyclohexane extract, the most interactive ligand in this study, for α-amylase was androst-5-ene-3,17-diol,3-acetate (3.β.,17.β.). It showed two hydrogen donors intracting with HIS201 and GLN63, two π-π interactions with TRP59, and three hydrophobic alkyl interactions, two with ALA198, and one with LEU162. In the case of EO, we observed lower values for α-amylase activity, which was confirmed by molecular docking. Lower values were also registered for both proteins α-amylase 4W93 and α-glucosidase 3W37. Cedrene was identified as the best compound with the highest binding energy with α-amylase, forming two π-sigma interactions with TYR62 and TRP59, and two π-alkyl interactions with TRP58 and TYP92. On the other hand, valencene showed better binding energy with α-glucosidase 3W37, with four interactions with the same amino acid TRP59, two being π-sigma and two being π-alkyl. Table 5 summarizes the different values of binding energies, type, and distances of each interaction for both proteins α-amylase 4W93 and α-glucosidase 3W37.
Table 5. Different values of banding energies, type and distances of each interaction for both proteins α-amylase 4W93 and α-glucosidase 3W37.
Human pancreatic α-amylase is 56 kD protein composed of 496 amino acids as single peptide chains divided into three domains: domain A (residues 1–99 and 169–404), the largest part where the active site is located as a V-shaped cleft in the catalytic domain A that binds to the substrate, domain B (residues 100–168) emerges from domain A and is located between the third β-strand and the α-helix of the β-barrel of the catalytic domain, maintaining the conformation of the enzyme, and domain C (residues 405–496) is an anti-parallel β-sheet of domain A loosely associated with other domains (Brayer et al., 1995; Mótyán et al., 2011; Nahoum et al., 2000; Williams et al., 2012). According to the results of our research, domain A accounts for the majority of contacts with a particular level of efficacy such as those of ASP197 and GLU233 except for the interaction of LEU162 which belongs to domain B and presents a lower hydrophobic interaction.
In the same context, the structures of α-glucosidases are complex. There are four primary domains in this structure: two C-terminal domains, a catalytic domain of the (β/α) 8-barrel, and an N-terminal domain. The inserts 1 and 2 of the catalytic domain are situated next to β-stands 3 and 4, respectively. Except for insert 1, the overall architectures of these subunits are nearly identical. The β-barrel loops generate the active site pocket (Subsite-1) in the catalytic domain, and the residues involved in subsite-1 formation have been extensively conserved throughout the subunits of α-glucosidases (Kashtoh and Baek, 2022). In our study, the most interesting interaction that contributes to enzyme/ligand binding was established with SER497, ARG552, and LYS506, which provide hydrogen-donor interactions in the case of Androst-5-ene-3, 17-diol, 3-acetate, (3.β.,17.β.), Neoabietyl acetate, and γ-Sitosterol.
The compounds (best docked) from each extract were further investigated using the online software ADMETSAR, which provides more knowledge about pharmacokinetic, physiochemical behavior, and druglikeness. ADMET properties of those bioactive compounds, which include Caco-2 cell permeability, brain/blood barrier, human intestinal absorption, AMES mutagenesis, and carcinogenicity, were elucidated in the present study. Results of ADMETSAR were analyzed and tabulated in Tables 6 and 7. The molecular weight of all the bioactive compounds was found to be less than 500, anticipating their easy transportation, absorption, and diffusion. Hydrogen bonding describes drug permeability; poor permeation correlates to more than 5 H-bond donors and 10 H-bond acceptors, which were not detected in our results, indicating the best permeation for all tested compounds. Despite that, not all our tested compounds are in the range of acceptable ALog P value (≤ 5), but still orally absorbed based on the Lipinsky rule, an important consideration of the drug’s likeness. Furthermore, a drug molecule is expected to be in an aqueous solubility range of –1 to –5, and the Log S values of all the selected compounds fall within the range except for the case of Abitatriene, which was observed with a value of -6.20 (Bergenhem, 2011). The different results are shown in Table 6. The determination of pharmacological qualities is crucial to identify bioactive molecules with acceptable pharmaceutical properties and then address their drug availability. This involves researching absorption, distribution, metabolism, excretion, and toxicity (Ghannay et al., 2020).
Table 6. Physicochemical parameters (Lipinski Rule of Five) of the best docked bioactive compounds of Juniperus phoenicea berries.
Molecules | Molecular weight | Number of HBA |
Number of HBD |
Number of Rotation Bond |
ALog P | Log S |
---|---|---|---|---|---|---|
Cedrene | 204.36 | 0 | 0 | 0 | 4.42 | –4.80 |
Humulene epoxyde II | 220.36 | 1 | 0 | 0 | 4.25 | –3.18 |
Caryophyllene | 204.36 | 0 | 0 | 0 | 4.73 | –4.69 |
Valencene | 204.36 | 0 | 0 | 1 | 4.73 | –5.36 |
γ-Muurolene | 204.36 | 0 | 0 | 1 | 4.58 | –5.37 |
δ-Cadinene | 204.36 | 0 | 0 | 1 | 4.73 | –5.25 |
Androst-5-ene-3,17-diol, 3-acetate ,(3.β.,17.β.) | 332.48 | 3 | 1 | 1 | 4.24 | –5.20 |
γ-Sitosterol | 414.72 | 1 | 1 | 6 | 8.02 | –4.70 |
Abieta-8,11,13-trien-7-one | 284.44 | 1 | 0 | 1 | 5.48 | –4.52 |
Neoabietol | 288.47 | 1 | 1 | 1 | 5.26 | –3.95 |
Aromadendrene epoxide | 220.36 | 1 | 0 | 0 | 3.48 | –2.98 |
Neoabietyl acetate | 330.51 | 2 | 0 | 3 | 5.68 | –4.88 |
Abitatriene | 270.46 | 0 | 0 | 1 | 5.84 | –6.20 |
HBA: hydrogen bond acceptor; HBD: hydrogen bond donor; ALog P: compound octanol/water partition coefficient; Log S: solubility coefficient.
Table 7. Pharmacokinetic ADMET profile of the best-docked bioactive compounds of Juniperus phoenicea berries.
Molecules | HIAa | Caco-2b | BBBc | HOBd | Carcinogenicity | Ames | Respiratory |
---|---|---|---|---|---|---|---|
Mutagenesis toxicity | |||||||
V P | V P | V P | V P | V P | V P | V P | |
Cedrene | + 0.9930 | + 0.7991 | + 0.9750 | + 0.6000 | – 0.7500 | – 0.8900 | + 0.5222 |
Humulene epoxyde II | + 0.9923 | + 0.8466 | + 0.9250 | + 0.7286 | – 0.6900 | – 0.7400 | + 0.5778 |
Caryophyllene | + 0.9881 | + 0.8656 | + 0.9250 | + 0.6429 | – 0.6500 | – 1.0000 | – 0.5556 |
Valencene | + 0.9930 | + 0.9131 | + 0.9000 | – 0.5714 | – 0.7900 | – 0.8500 | – 0.7444 |
γ-Muurolene | + 0.9946 | + 0.9185 | + 0.9250 | + 0.6571 | – 0.7700 | – 0.8400 | – 0.6771 |
δ-Cadinene | + 0.9958 | + 0.9654 | + 0.9250 | + 0.7000 | – 0.8000 | – 0.8400 | – 0.6111 |
Androst-5 -ene-3,17-diol, 3- acetate, (3.β.,17.β.) | + 1.0000 | + 0.5732 | + 0.5750 | + 0.7143 | – 1.0000 | – 0.8770 | + 0.8444 |
γ-Sitosterol | + 1.0000 | + 0.5385 | + 0.5750 | + 0.5286 | – 0.9700 | – 0.9000 | + 0.9111 |
Abieta-8, 11,13-trien- 7-one | + 1.0000 | + 0.8710 | + 0.7750 | + 0.6000 | – 0.8500 | – 0.7900 | + 0.5556 |
Neoabietol | + 0.9943 | + 0.9493 | + 0.8750 | – 0.6143 | – 0.8900 | – 0.7900 | + 0.6667 |
Aromadendrene epoxide | + 0.9913 | + 0.8255 | + 0.9500 | + 0.6429 | – 0.7800 | – 0.6353 | – 0.5000 |
Neoabietyl acetate | + 1.0000 | + 0.8454 | + 0.7750 | + 0.5286 | – 0.7900 | – 0.8900 | + 0.5556 |
Abitatriene | + 0.9963 | + 0.9315 | + 0.9500 | + 0.6000 | – 0.7000 | – 0.8200 | – 0.6222 |
HIA: Human Intestine Absorption; Caco-2: permeability value; BBB: Blood Brain Barrier; HOB: Human Oral Bioavailability.
a: (+): more than 30%, (-): less than 30%.
b: (+): High Caco-2 permeability, (-): moderate-poor Caco-2 permeability.
c: (+) High BBB permeability; (−) moderate-poor BBB permeability.
d: (+) High HOB permeability; (−) moderate-poor HOB permeability.
ADMET pharmacokinetic properties reveal that all selected compounds had better Human Intestinal Adsorption (HIA) with a score of > 99%. Greater HIA indicates that the compound could be better absorbed from the intestinal tract upon oral administration. In addition to the HIA, all selected compounds had a positive value of Caco-2 permeability and blood-brain barrier BBB. It is observed that the lower value is registered for both γ-Sitosterol and Androst-5 -ene-3,17-diol, 3-acetate,(3.β.,17.β.). The admetSAR online tool was used to evaluate the toxicological properties of the selected compounds, as the safety of the compounds is an important parameter for becoming a good drug (Cheng et al., 2012), Table 7 presents the different values of the pharmacokinetic profile. In this study, no AMES toxicity and carcinogens were identified as a threat. An exception was observed for respiratory toxicity, where some of those compounds posed a positive threat against the respiratory system, such as cedrene, humulene epoxide II, androst-5-ene-3,17-diol,3-acetate (3.β.,17.β.), abieta-8,11,13-trien-7-one, neoabietol, neoabietyl acetate, and especially γ-sitosterol.
The medicinal plants include natural products such as phytochemicals, used in various medical applications including their antidiabetic effects. Traditionally, Juniperus phoenicea berries extracts are considered promising natural medicinal agents and widely used for the treatment of type 2 diabetes (T2D). The findings obtained in this study are divided into three main axes:
Analysis of the chemical composition of EO, cyclohexane extract, and ethyl acetate extract allowed the identification of50, 60, and 56 compounds, respectively, representing over 98% of the essential oil. Oxygenated monoterpenes are the major components, representing 68% of both non-polar extracts, with oxygenated diterpenes being the major components of the Juniperus phoenicea berries from the Aflou region.
In vitro assays for antidiabetic effects, based on the α-amylase inhibitory capability of both non-polar extracts, exhibited a strong inhibitory effect against the α-amylase enzyme in a dose-dependent manner. Maximum inhibition rates of over 75.0% were observed at the high tested dose of 400 mg/mL against α-amylase, with good IC50 values for both cyclohexane and ethyl acetate extracts (IC50 = 186.91 ± 5.74 mg/mL, IC50 = 351.48 ± 0.17 mg/mL) respectively. In comparison, the EO showed a maximum inhibition rate of 33.0% at the high tested dose.
The study was further enhanced with molecular docking analysis to get a comprehensive understanding of the phenomenon. Various compounds showed promising binding affinity to specific proteins (α-amylase and α-glucosidase), with, all compounds displaying a favorable ADMET profile, indicating their potential to serve as lead compounds in drug discovery. Comprehensive in silico studies, including molecular docking studies, were conducted to evaluate and support the in vitro findings. However, further studies using different antidiabetic models, especially in vivo models, are needed to confirm the beneficial qualities of these extracts.
The authors acknowledge and extend their appreciation to the Researchers Supporting Project Number (RSPD2024R709), King Saud University, Riyadh, Saudi Arabia for funding this study. The authors also would like the Directorate-General for Scientific Research and Technological Development (DGRSDT) of the Algerian Ministry of Higher Education and Scientific Research for their support.
The authors declare no conflict of interest.
This research was funded by King Saud University, Riyadh, Saudi Arabia, Project Number (RSPD2024R709).
Abdelli, W., Bahri, F., Höferl, M., Wanner, J., Schmidt, E. & Jirovetz, L. (2018). Chemical composition, antimicrobial and anti-inflammatory activity of Algerian Juniperus phoenicea essential oils. Natural Product Communications, 13(2), 223–228. 10.1177/1934578X1801300227
Adams, R. P. (2001). Identification of essential oil components by gas chromatography/ quadrupole mass spectrometry. Carol Stream, IL: Allured Publishing Corporation.
Al-Ishaq, R. K., Abotaleb, M., Kubatka, P., Kajo, K. & Büsselberg, D. (2019). Flavonoids and their anti-diabetic effects: Cellular mechanisms and effects to improve blood sugar levels. Biomolecules, 9(9), 430–464. 10.3390/biom9090430
Amalich, S., Zekri, N., N’Dédianhoua, K. S., Fadili, K., Khabbal, Y., Mahjoubi, M. & Zaïr, T. (2015). Chemical characterization and antibacterial evaluation of Juniperus phoenicea L. leaves and fruits’ essential oils from eastern high Atlas (Morocco). International Journal of Innovation and Applied Studies, 13, 881—889.
Amirifar, A., Hemati, A., Asgari Lajayer, B., Pandey, J., & Astatkie, T. (2022). Impact of various environmental factors on the biosynthesis of alkaloids in medicinal plants, In: Aftab, T., editor. Environmental challenges and medicinal plants. Cham, Switzerland: Springer, pp. 229–248.
Angioni, A., Barra, A., Russo, M. T., Coroneo, V., Dessi, S., & Cabras, P. (2003). Chemical composition of the essential oils of Juniperus from ripe and unripe berries and leaves and their antimicrobial activity. Journal of Agricultural and Food Chemistry, 51(10), 3073–3080. 10.1021/jf026203j
Asgari, Lajayer B., Ghorbanpour, M., & Nikabadi, S. (2017). Heavy metals in contaminated environment: Destiny of secondary metabolite biosynthesis, oxidative status and phytoextraction in medicinal plants. Ecotoxicology and Environmental Safety, 145, 377–390. 10.1016/j.ecoenv.2017.07.035
Banerjee, S., Singh, H., & Chatterjee, T. K. (2013). Evaluation of anti-diabetic and anti-hyperlipidemic potential of methanolic extract of Juniperus communis (L.) in streptozotocin-nicotinamide induced diabetic rats. International Journal of Pharmaceutical and Biological Sciences, 4(3), 10–17. https://www.cabidigitallibrary.org/doi/full/10.5555/20133392186
Barrero, A. F., et al. (2005). Chemical composition of the essential oils of Cupressus atlantica Gaussen. Journal of Essential Oil Research, 17(4), 437–439. 10.1080/10412905.2005.9698954
Bergenhem, N. (2011). Preclinical candidate nomination and development. In: Tsaioun, K., Kate, S. A., editors. Admet for medicinal chemists. Singapore: John Wiley and Sons, pp. 399–415.
Boudiba, S., et al. (2021). Anti-quorum sensing and antioxidant activity of essential oils extracted from Juniperus species, growing spontaneously in Tebessa region (East of Algeria). Natural Product Communications, 16(6), 1934578X2110240. 10.1177/1934578X2110240
Bolouri, Parisa., et al. (2022). Applications of essential oils and plant extracts in different industries. Molecules, 27(24), 8999. 10.3390/molecules27248999
Bouras, Y., Atef, C., Cherrada, N., Gheraissa, N., Chenna, D., Elkhalifa, A., et al. (2024). Phytochemical profile and biological activities of Brassica oleracea var. elongata leaf and seed extracts: An in vitro study. Italian Journal of Food Science, 36(4), 193–207. 10.15586/ijfs.v36i4.2691
Bouyahyaoui, A., et al. (2016). Antimicrobial activity and chemical analysis of the essential oil of Algerian Juniperus phoenicea. Natural Product Communications, 11(4). PMid: 27396209.
Brayer, G. D., Luo, Y., & Withers, S. G. (1995). The structure of human pancreatic alpha-amylase at 1.8: A resolution and comparisons with related enzymes. Protein Science, 4(9), 1730–1742. 10.1002/pro.5560040908
Byrne, F. P., et al. (2016). Tools and techniques for solvent selection: Green solvent selection guides. Sustainable Chemical Process, 4, 7. 10.1186/s40508-016-0051-z
Cheng, F., et al. (2012). admetSAR: A comprehensive source and free tool for assessment of chemical ADMET properties. Journal of Chemical Information and Modeling, 52(11), 3099–3105. 10.1021/ci300367a
Conforti, F., Statti, G., Loizzo, M. R., Sacchetti, G., Poli, F., & Menichini, F. (2005). In vitro antioxidant effect and inhibition of alpha-amylase of two varieties of Amaranthus caudatus seeds. Biological and Pharmaceutical Bulletin, 28(6), 1098–1102. 10.1248/bpb.28.1098
Deo, P., et al. (2016). In vitro inhibitory activities of selected Australian medicinal plant extracts against protein glycation, angiotensin converting enzyme (ACE) and digestive enzymes linked to type II diabetes. BMC Complementary and Alternative Medicine, 16, 435–445. :10.1186/s12906-016-1421-5
Edris, A. E. (2007). Pharmaceutical and therapeutic potentials of essential oils and their individual volatile constituents: A review. Phytotherapy Research, 21(4), 308–323. 10.1002/ptr.2072
El-Sawi, S. A., Motawae, H. M., & Ali, A. M. (2007). Chemical composition, cytotoxic activity and antimicrobial activity of essential oils of leaves and berries of Juniperus phoenicea L. grown in Egypt. African Journal of Traditional, Complementary, and Alternative Medicines: AJTCAM, 4(4), 417–426. 10.4314/ajtcam.v4i4.31236
Ennajar, M., Bouajila, J., Lebrihi, A., Mathieu, F., Abderraba, M., Raies, A., et al. (2009). Chemical composition and antimicrobial and antioxidant activities of essential oils and various extracts of Juniperus phoenicea L. (Cupressacees). Journal of Food Science, 74(7), M364–M371. 10.1111/j.1750-3841.2009.01277.x
Ennajar, Monia., et al. (2010). The influence of organ, season and drying method on chemical composition and antioxidant and antimicrobial activities of Juniperus phoenicea L. essential oils. Journal of the Science of Food and Agriculture, 90(3), 462–470. 10.1002/jsfa.3840
Ghannay, S., Kadri, A., & Aouadi, K. (2020). Synthesis, in vitro antimicrobial assessment, and computational investigation of pharmacokinetic and bioactivity properties of novel trifluoromethylated compounds using in silico ADME and toxicity prediction tools. Monatshefte für Chemie, 151, 267–280. 10.1007/s00706-020-02550-4
Ghouti, D., et al. (2018). Phenolic profile and in vitro bioactive potential of Saharan Juniperus phoenicea L., and Cotula cinerea (Del) growing in Algeria. Food & Function Journa,. 9, 4664–4672. 10.1039/C8FO01392F
Gülçin, İlhami. (2010). Antioxidant properties of resveratrol: A structure–activity insight. Innovative Food Science & Emerging Technologies, 11(1), 210–218. 10.1016/j.ifset.2009.07.002
Harhour, A., Brada, M., Fauconnier, M-L., & Lognay, G. (2018). Chemical composition and antioxidant activity of Algerian Juniperus phoenicea essential oil. Natural Product Sciences, 24(2), 125–131. 10.20307/nps.2018.24.2.125
Hongbin, Y., et al. (2019). admetSAR 2.0: Web-service for prediction and optimization of chemical ADMET properties. Bioinformatics, 35(6), 1067–1069. 10.1093/bioinformatics/bty707
Huang, S. Y., & Zou, X. (2010). Advances and challenges in protein-ligand docking. International Journal of Molecular Sciences, 11(8), 3016–3034. 10.3390/ijms11083016
Jain, P. L., Patel, S. R., & Desai, M. A. (2022). Patchouli oil: An overview on extraction method, composition and biological activities. Journal of Essential Oil Research, 34, 1–11. 10.1080/10412905.2021.1955761
Kadi, Imededdine. et al. (2023). Molecular interactions, binding stability, and synergistic inhibition on Acetylcholinesterase activity of Safranin O in combination with Quercetin and Gallic acid: In vitro and in silico study. Journal of Molecular Structure, 1286, 135562. 10.1016/j.molstruc.2023.135562
Kashtoh, Hamdy., & Kwang-Hyun Baek. (2022). Recent updates on phytoconstituent alpha-glucosidase inhibitors: An approach towards the treatment of type two diabetes. Plants,20, 2722. 10.3390/plants11202722
Keskes, Henda, Mnafgui, Kais, Hamden, Khaled, Damak, Mohamed, El Feki, Abdelfattah, & Allouche, Noureddine. (2014). In vitro anti-diabetic, anti-obesity and antioxidant proprieties of Juniperus phoenicea L. leaves from Tunisia. Asian Pacific Journal of Tropical Biomedicine, 4(2), S649–S655. 10.12980/APJTB.4.201414B114
Khodaei, Nastaran, Nguyen, Marina, Minh, Mdimagh, Asma, Bayen, Stéphane, & Karboune, Salwa. (2021). Compositional diversity and antioxidant properties of essential oils: Predictive models. LWT, 138, 110684. 10.1016/j.lwt.2020.110684
Loizzo, Monica R., Tundis, Rosa, Conforti, Filomena, Saab, Antoine, M., Statti, Giancarlo A., & Menichini, Francesco. (2007). Comparative chemical composition, antioxidant and hypoglycaemic activities of Juniperus oxycedrus ssp. oxycedrus L. berry and wood oils from Lebanon. Food Chemistry, 105(2), 572–578. 10.1016/j.foodchem.2007.04.015
Medini, H., et al. (2011). Chemical composition and antioxidant activity of the essential oil of Juniperus phoenicea L. berries. Natural Product Research, 25, 1695–1706. 10.1080/14786419.2010.535168
Mehira, K., et al. (2021). Chemical composition, antioxidant and antibacterial efficiency of essential oils from Algerian Juniperus phoenicea L. against some pathogenic bacteria. Tropical Journal of Natural Product Research (TJNPR), 5(11), 1966–1972. 10.26538/tjnpr/v5i11.13
Menaceur, F., Benchabane, A., Hazzit, M., & Baaliouamer, A. (2013). Chemical composition and antioxidant activity of Algerian Juniperus phoenicea L. extracts. Journal of Biologically Active Products from Nature, 3(1), 87–96. 10.1080/22311866.2013.782754
Messaoudi, M., Rebiai, A., Sawicka, B., Atanassova, M., Ouakouak, H., Larkem, I., et al. (2022). Effect of extraction methods on polyphenols, flavonoids, mineral elements, and biological activities of essential oil and extracts of Mentha pulegium L. Molecules, 27(1), 11. 10.3390/molecules27010011
Moreno, M. I., Isla, M. I., Sampietro, A. R., & Vattuone, M. A. (2000). Comparison of the free radical-scavenging activity of propolis from several regions of Argentina. Journal of Ethnopharmacology, 71(1–2), 109–114. 10.1016/s0378-8741(99)00189-0
Merola, N., Castillo, J., Benavente-García, O., Ros, G., & Nieto, G. (2017). The effect of consumption of citrus fruit and olive leaf extract on lipid metabolism. Nutrients, 9(10), 1062. 10.3390/nu9101062
Morris, G. M., Huey, R., Lindstrom, W., Sanner, M. F., Belew, R. K., & Goodsell, D. S. 2009. AutoDock4 and AutoDockTools4: Automated docking with selective receptor flexibility. Journal of Computational Chemistry, 30, 2785–2791. 10.1002/jcc.21256
Mótyán, János A., Gyémánt, Gyöngyi, Harangi, János, & Bagossi, Péter. (2011). Computer-aided subsite mapping of α-amylases. Carbohydrate Research, 346(3), 410–415. 10.1016/j.carres.2010.12.002
Nahoum, V., et al. (2000). Crystal structures of human pancreatic alpha-amylase in complex with carbohydrate and proteinaceous inhibitors. The Biochemical Journal, 346(1), 201–208. PMid: 10657258; PMCID: PMC1220841.
Orhan, N., Aslan, M., Pekcan, M., Orhan, D. D., Bedir, E., & Ergun, F. (2012). Identification of hypoglycaemic compounds from berries of Juniperus oxycedrus subsp. oxycedrus through bioactivity guided isolation technique. Journal of Ethnopharmacology, 139(1), 110–118. 10.1016/j.jep.2011.10.027
Ostadi, A., Javanmard, A., Amani, Machiani, M., Morshedloo, M. R., Nouraein, M., Rasouli, F., et al. (2020). Effect of different fertilizer sources and harvesting time on the growth characteristics, nutrient uptakes, essential oil productivity and composition of Mentha x piperita L. Industrial Crops and Products, 148, 112290. 10.1016/j.indcrop.2020.112290
Oyaizu, M. (1986). Studies on products of browning reactions: Antioxidative activities of product of browning reaction prepared from glucosamine. Japan Journal of Nutrition, 44, 307–315. 10.5264/eiyogakuzashi.44.307
Özyürek, Mustafa, Güngör, Nilay, Baki, Sefa, Güçlü, Kubilay, & Apak, Reşat. (2012). Development of a silver nanoparticle-based method for the antioxidant capacity measurement of polyphenols. Analytical Chemistry, 84(18), 8052–8059. 10.1021/ac301925b
Pacifico, Severina, et al. (2013). Apolar Laurus nobilis leaf extracts induce cytotoxicity and apoptosis towards three nervous system cell lines. Food and Chemical Toxicology, 62, 628–637. 10.1016/j.fct.2013.09.029
Priscilla, D. H., Roy, D., Suresh, A., Kumar, V., & Thirumurugan, K. (2014). Naringenin inhibits α-glucosidase activity: A promising strategy for the regulation of postprandial hyperglycemia in high fat diet fed streptozotocin induced diabetic rats. Chemico-Biological Interactions, 210, 77–85. 10.1016/j.cbi.2013.12.014
Samadi, Saba., Asgari, Behnam, Lajayer, Moghiseh, Ebrahim, & Rodríguez-Couto, Susana. (2021). Effect of carbon nanomaterials on cell toxicity, biomass production, nutritional and active compound accumulation in plants. Environmental Technology & Innovation, 21, 101323. 10.1016/j.eti.2020.101323
Sánchez de Medina, F., Gámez, M. J., Jiménez, I., Jiménez, J., Osuna, J. I., & Zarzuelo, A. (1994). Hypoglycemic activity of juniper “berries”. Planta Medica, 60(3), 197–200. 10.1055/s-2006-959457
Sánchez-Vioque, R., et al. (2013). Polyphenol composition and antioxidant and metal chelating activities of the solid residues from the essential oil industry. Industrial Crops and Products, 49, 150–159. 10.1016/j.indcrop.2013.04.053
Singleton, Vernon, L., Orthofer, Rudolf, & Lamuela-Raventós, Rosa, M. (1999). Analysis of total phenols and other oxidation substrates and antioxidants by means of folin-ciocalteu reagent. Methods in Enzymology, 299, 152–178. 10.1016/S0076-6879(99)99017-1
Swanston-Flatt, S. K., Day, C., Bailey, C. J., & Flatt, P. R. (1990). Traditional plant treatments for diabetes. Studies in normal and streptozotocin diabetic mice. Diabetologia, 33(8), 462–464. 10.1007/BF00405106
Taviano, M. F., et al. (2011). Antioxidant and antimicrobial activities of branches extracts of five Juniperus species from Turkey. Pharmaceutical Biology, 49(10), 1014–1022. 10.3109/13880209.2011.560161
Trott, O., & Olson, A. J. (2010). AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. Journal of Computational Chemistry, . 31(2), 455–461. 10.1002/jcc.21334
Wang, Zhe, et al. (2020). Combined strategies in structure-based virtual screening. Physical Chemistry Chemical Physics, 22 (6), 3149–3159. 10.1039/C9CP06303J
Williams, L. K., Chunmin Li, Stephen, G. Withers, & Gary, D. Brayer. (2012). Order and disorder: Differential structural impacts of myricetin and ethyl caffeate on human amylase, an antidiabetic target. Journal of Medicinal Chemistry, 55(22), 10177–10186. 10.1021/jm301273u
Yokozawa, T., Chen, C. P., Dong, E., Tanaka, T., Nonaka, G. I., & Nishioka, I. (1998). Study on the inhibitory effect of tannins and flavonoids against the 1,1-diphenyl-2 picrylhydrazyl radical. Biochemical Pharmacology,.. 56(2), 213–222. 10.1016/s0006-2952(98)00128-2
Zahnit, W., Smara, O., Bechki, L., Bensouici, C., Messaoudi, M., Benchikha, N.,... et al. (2022). Phytochemical profiling, mineral elements, and biological activities of Artemisia campestris L. grown in Algeria. Horticulturae, 8(10), 914. 10.3390/horticulturae8100914
Zengin, G., Sarikurkcu, C., Aktumsek, A., Ceylan, R., & Ceylan, O. (2014). A comprehensive study on phytochemical characterization of Haplophyllum myrtifolium Boiss. endemic to Turkey and its inhibitory potential against key enzymes involved in Alzheimer, skin diseases and type II diabetes. Industrial Crops and Products. 53, 244–251. 10.1016/j.indcrop.2013.12.043