Introduction
Medicinal plants have been utilized for thousands of years to cure many different diseases. They are currently used by a large percentage of the population because of their generally reasonable safe application, considerable customer acceptability, and possible multi-purpose functional use [1]. Nowadays, we can state that bioactive components, extracted from plants along with their chemical substitutes, account for more than half of FDA-approved medications and continue to be significant sources for the discovery of novel active pharmaceuticals [2].
Plants that produce essential oils represent a great portion of the abundant natural resource used in a variety of industries, including pharmaceutical, food, and cosmetic fields. This is due to their flavor, smell, and diverse biological activity [3]. One of these remarkable plants is coriander. Coriander (Coriandrum sativum L.) is classified as a member of the Umbelliferae/Apiaceae family that is widely utilized in pharmaceuticals and food industries as a herb and medicine [4]. It is well established that coriander possesses high concentrations of bioactive substances such as linalool and geranyl acetate, which are known for their antioxidant, anti-inflammatory, and antibacterial properties [5–8]. Furthermore, previous research conducted on coriander revealed potential anti-diabetic, hypocholesterolemic, anthelmintic, hepatoprotective, antihypertensive, anticancer [9,10], and interestingly, some studies showed a protective effect against metals poisoning [11,12].
Inflammation is a "host defense" strategy against pathogens that involves increased or excessive production of Reactive Oxygen Species (ROS) by activated pro-inflammatory cytokines and immune cells [13]. ROS aids in clearing tissue invading germs, but when produced in large quantities, it can increase oxidative stress and chronic inflammatory-related diseases [14]. Furthermore, while inflammation causes oxidative stress, the inverse sequence of events is also true. As a result, inflammation and oxidative stress are tightly intercalated [15].
Severe inflammation can increase immune cell activity, which may subsequently, damage tissues and body health [16,17]. When inflammation develops, many pro-inflammatory mediators are overproduced, leading to the escalation of a variety of reactions such as vascular changes and white blood cell responses [18]. Macrophage cells have a vital role in the activation of pro-inflammatory mediators. Toll-like receptor 4 becomes active in cases of infected cells, causing nuclear factor-kappa B (NF-κB) translocation from the cytoplasm into the nucleus [19]. Once NF-κB is active, inflammatory mediators such as interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), Nitric Oxide (NO), and cyclooxygenase-2 (COX-2) are activated [20–23]. However, a recent study showed that coriander oil extract derivatives such as phenolic compounds and flavonoids possess anti-inflammatory action through decreasing the levels of multiple inflammatory cytokines or inflammatory mediators such as IL-1β, IL-6, IL-10, TNF-α, NF-κB, NO, and COX-2 [24].
This study aims to assess the antioxidant, anti-inflammatory, and immunostimulatory activities of different concentrations of Jordanian seed extracted coriander essential oil (JCEO) against RAW246.7 murine macrophages using a variety of well-known immunological assays and anti-oxidant assessment techniques.
Materials and methods
Extraction of coriander essential oil from Jordanian coriander seeds using hydro-distillation and Clevenger apparatus
Coriander seeds were purchased from the Saffron Kingdom herb market in Amman, Jordan. These seeds were cultivated and harvested during 2020–2021. The essential oil from coriander seeds was extracted using a hydro-distillation method and a glass Clevenger apparatus (Borosil, India). The seeds (150-200g) were moderately grinded using the mechanical grinder for 30 seconds before hydro-distillation, to allow more essential oil release. Powdered seeds were mixed with approximately 1500 mL of filtered water and placed in a 2000 mL round-bottom flask to extract the essential oil, followed by heating using a heating mantle (Electrothermal, UK), and condensation using a cooler circulator (Lauda, Austria) for 2-hrs to obtain approximately 1.5-2mL of coriander essential oil [25,26]. The extracted essential oil was collected into dark amber vials and stored at 4±2°C until used for further investigation, Fig 1.
[Figure omitted. See PDF.]
Gas chromatography-mass spectrometry (GC-MS) analysis.
A sample of the CEO was sent to the University of California, San Diego, USA, for analysis using an Agilent 5977B Gas Chromatograph-Mass Selective Detector (GC-MSD). This instrument features electron impact sources connected to an Agilent 7820A GC system, allowing the analysis of sample mixtures. It is particularly effective for routine examination of non-polar small organic compounds with masses ranging from 50 to 1000 amu. The results were compared against the NIST (National Institute of Standards and Technology) library, which contains over 1 million compounds. For the analysis, approximately 1 μL of the CEO sample was diluted in 10 μL of GC-grade n-hexane and then subjected to GC analysis. This was carried out using a Varian Chrompack CP-3800 GC-MS/MS-200 (Saturn, Netherlands) equipped with a DB-5 (5% diphenyl, 95% dimethyl polysiloxane) GC capillary column (30 m × 0.25 mm i.d., 0.25 μm film thickness) and helium as the carrier gas at a flow rate of 0.9 mL/min. Compounds were identified by comparing them to entries in the built-in libraries (NIST and Wiley Co. USA).
Cell culturing and treatment
Murine macrophages RAW 246.7 were provided from the American Type Culture Collection (TIB-71™-ATCC, USA). Cells were defrosted from -80±5°C and placed in a 75cm2 culture flask in an incubator at 37±2°C and 5% CO2. Cells were maintained in Dulbecco’s modified Eagle’s medium (DMEM, Gibco™, USA) supplemented with 10% fetal bovine serum (FBS, Gibco™, USA), and 1% penicillin and streptomycin antibiotic mixture (P/S, Gibco™, USA). The confluence of the cells was continually checked under optical microscopy (Nikon, USA) until it reached 75–80% approximation. Before treatment with JCEO, RAW264.7 cells were seeded at an inoculum of 1×105 cells/well in 96-well cell culture plates and incubated at 37±2°C and 5% CO2 in a humidified incubator (Biosan, China) for 24±2 hrs. The medium (DMEM supplemented with 10% FBS) was removed and cultured cells were washed 3 times with phosphate buffer saline PBS (PBS, Gibco, USA). Then, 100μL of different concentrations of JCEO (0.08, 0.16, 0.3, 0.6, 1.25, and 2.5 mg/mL) was added, and the cells were again incubated overnight at 37±2°C and 5% CO2. Then, the cells were washed again with PBS, and the supernatant was collected for further analysis. Untreated macrophages were utilized as a negative control, while lipopolysaccharides LPS from Pseudomonas aeruginosa (LPS, Sigma Aldrich, L9143) was used as a positive control (1μg/mL).
Viability assessment
To select a range of non-toxic concentrations, the cytotoxicity potential of different concentrations of JCEO was tested in triplicate using 3-(4, 5-dimethyl-2-thiazyl)-2, 5-diphenyl-2H-tetrazolium bromide (MTT) reduction assay [26,27]. Briefly, 100μL of RAW246.7 murine macrophages were seeded at a density of 1×105 cells/well in a 96-well plate with 100μL of fresh supplemented (FBS 10%) DMEM medium per well. Cells were allowed to adhere for 24±2-hrs under standard conditions in a humidified incubator (37±2°C, 5% CO2). The medium was removed after 24±2-hrs of incubation and replaced with 100μL of fresh supplemented (FBS 10%) DMEM medium containing different concentrations (0.08, 0.16, 0.3, 0.6, 1.25, and 2.5 mg/mL) of JCEO. For negative control wells, only 100μL of fresh DMEM medium supplemented with FBS 10% and Penicillin/Streptomycin1% (Gibco, UK) was utilized. For positive control wells, only 100μL of supplemented (FBS 10%) DMEM medium with LPS (1μg/mL) was utilized. Following 24±2-hrs of incubation, 20μL of MTT (5 mg/ml; Sigma Aldrich) was added into each well and further incubated for another 4-hrs in a humidified incubator (37±2°C, 5% CO2). The formation of insoluble purple formazan from yellowish MTT reaction was dissolved by the addition of 100μL of Dimethyl sulfoxide (DMSO, Sigma Aldrich), and the absorbance was measured at 570nm using a Bio-Tek® microplate reader (Bio-Tek, Germany). The formula below was used to calculate the percentage of cell inhibition:
Cell migration assay
RAW246.7 cells with a density of 1×105 cells/well) were seeded in a 96-well plate and grown for 24±2-hrs under standard conditions in a humidified incubator (37±2°C, 5% CO2). The cell monolayer was then scratched vertically with a sterile 10μL pipette tip, washed three times with PBS (Gibco, USA), and incubated with 100μL of supplemented (10% FBS) DMEM medium containing JCEO with different concentrations (0.3, and 0.16 mg/mL). LPS (1μg/mL) was utilized as a positive control, while untreated macrophages were used as a negative control. The plate was then incubated at 37±2°C in a humidified 5% CO2 incubator and the gap closure was observed using an inverted lens microscope (Nikon, USA) after 24±2 hrs, and 48±2-hrs. The gap closure was calculated using the ImageJ software using images taken by iPhone (Apple, USA).
TNF-α assay
Tumor necrosis factor-α (TNF-α) level was determined using a mouse enzyme-linked immunosorbent assay (ELISA) kit (Thermo Fisher, US) according to the manufacturer`s instructions. Treatment of RAW264.7 cells (1×105 cells/well) with different concentrations of JCEO (0.3 and 0.15 mg/mL), side by side with LPS (1μg/mL) as a positive control, while untreated macrophages were used as a negative control. The supernatant was obtained from wells after 6 hr of treatment and the amount of TNF-α in cell-free supernatant was determined at 450 nm.
The DPPH free radical-scavenging assay
In this test, the 2,2-diphenyl-1-picrylhydrazyl (DPPH) radical, which generates a purple solution (λ max = 517 nm), is reduced when it combines with any antioxidant that can contribute a hydrogen atom, generating a yellow-colored diphenyl-picrylhydrazine molecule [28,29]. The DPPH free radical-scavenging activity assessment was carried out using a previously published method [30,31]. JCEO was diluted in methanol to make concentrations range of 100, 50, 25, 12.5, 6.25, 3.13, 1.6, and 0.78 mg/mL. The 180μL reaction mixture contained 90μL of 0.1mM DPPH (dissolved in methanol) and 90μL of JCEO solutions of various concentrations. The test reagent was mixed with the different JCEO concentrations thoroughly in 96-well plates, incubated at 37±2°C for 30 minutes, and the absorbance at 517 nm was measured using a Bio-Tek® microplate reader (Bio-Tek, Germany). The percentage of DPPH antioxidant activity was calculated as follows:
Cytokine release assessment
Supernatants (100μL) were collected from previously seeded cells at 1×105 cells/well density to assess the levels of INF-γ, IL-10, IL-4, and IL-2 using TH1/TH2 ELISA kit (Thermo Fisher, USA), as per the manufacturer’s protocol. The absorbance readings for each treatment were translated into concentration (ng/mL) using a standard curve of predetermined cytokine concentrations.
Phagocytosis assay
A phagocytosis assay kit (Red E. coli, ab235901, UK) was used to measure phagocytic activity. Following 24±2-hrs previously seeded RAW267.4 cells at a density of 1×105 cells/well, followed by treatment of 0.6, 0.3, and 0.16 mg/mL of JCEO, seeded cells were rinsed three times with PBS. A volume of 5μL of red fluorescence E. coli was placed in each well, and incubated at 37±2°C, 5% CO2 for 2-hrs. Wells were rinsed again three times with 100μL PBS, and fluorescence was measured using a Bio-Tek® microplate reader (Bio-Tek, Germany) at 509⁄533nm.
Fluorescence imaging using DAPI stain
RAW246.7 murine cells were seeded overnight on coverslips inside a 6-well plate (1×105 cells/well). The next day the cells were treated with different concentrations of JCEO (0.6, 0.3, and 0.16 mg/mL) for 24±2-hrs at 37±2°C incubator. After 24-hrs of incubation, the cells were washed three times using 100μL PBS to remove all DMEM residual before the addition of E. coli particles (5μL of E. coli in 100μL of media in each well) for 2-hrs. After applying the E. coli for 2-hrs, the media was removed and the cells were rinsed with 100μL PBS three times, before they were fixed using 4% formaldehyde prepared in PBS. For staining and imaging purposes, the 4% formaldehyde was discarded, and the cells were washed again with 100μL PBS three times. Two drops of DAPI (4’,6-diamidine-20’-phenyl indole dihydrochloride) solution (abcam, UK) was added to the slide, and the coverslip was removed from the 6-well plate using forceps to place it above the DAPI solution for fluorescence imaging using confocal laser scanning microscope (Confocal LSM 780, ZEISS, Germany).
Nitrate concentration assay
RAW264.7 macrophages (1×105 cells/well) were previously seeded in 96-wells and pre-treated with LPS (1μg/mL) for 24±2-hrs, and then treated with different concentrations of JCEO (0.6, 0.3, and 0.16 mg/mL) for another 24±2-hrs. The collected supernatant was used to measure the nitrite levels by Griess reagent using a Total NO/Nitrate/Nitrite Parameter Assay kit (ParameterTM, R&D Systems, USA) as per the manufacturer’s protocol. The absorbance was detected using a Bio-Tek® microplate reader (Bio-Tek, Germany) at 550 nm.
Statistical analysis
Data analyses were accomplished by using the GraphPad Prism 9 (GraphPad Software, Inc., San Diego, CA, United States). Data is presented as mean ± SD. Statistical significance was determined using one-way analysis of variance (ANOVA) with Tukey’s post-hoc test for multiple group comparisons. All statistical analyses were based on a p-value < 0.05 level of significance.
Results
Essential oil characterization
The amount of the CEO obtained from each 100 g coriander seeds was 0.313 (±0.01) mL, (n = 3). GCMS analysis (S1 File) of the hydro-distilled oil from the Coriander seeds revealed 6 identified major volatile compounds: α-pinene (terpene), limonene (aliphatic hydrocarbon), linalool (terpene alcohol), camphor (terpene ketone), o-cymene and γ-terpinene (hydrocarbons), as demonstrated in Table 1.
[Figure omitted. See PDF.]
Viability assessment
RAW 246.7 murine macrophage cells (1x105 cells/well) were treated with different concentrations of JCEO (0.02, 0.04, 0.08, 0.16, 0.30, 0.60, 1.25 and 2.50mg/mL). The results of the MTT assay showed that concentrations > 0.3 mg/mL of CEO were considerably cytotoxic, as the viability of cells was lower than the accepted viability limit in this study (80%). As demonstrated in Fig 2, a significant increase in the viability percentage of RAW 246.7 was observed at 0.3mg/mL (****p <0.0001). In other words, the percentage had reached 191.85% compared to untreated negative control cells. It was also higher than that of the positive control LPS (1μg/mL), which reached a percentage of 105.38% compared to negative control cells (untreated macrophages).
[Figure omitted. See PDF.]
This experiment was conducted using six replicates for each treatment. The lower acceptable limit of cell viability in this study was 80% as indicated by the red dashed line. Data in the figure is represented as mean± SD, (****p <0.0001, * p<0.05).
Migration assay results
RAW264.7 murine macrophages were evaluated in a 96-well pre-seeded plate for 24±2 hrs, and 48±2-hrs after treatment to see the effect of different concentrations of JCEO (0.16, 0.3, 0.6 mg/mL) on cell migration and gap closure. The two concentrations of JCEO 0.3, and 0.6mg/mL treated cells showed the most substantial and maximal gap closure after 24±2 hours when compared to 0.16 mg/mL of JCEO, LPS (1μg/mL), and the negative control (untreated macrophages). However, the concentrations of 0.16, 0.3, and LPS showed a full gap closure after 48 hours, as shown in Fig 3.
[Figure omitted. See PDF.]
This experiment was conducted using ten replicates for each treatment (**p <0.01). Data in the figure is represented as mean± SD.
TNF-α assay
TNF-α was measured in the supernatants of previously seeded RAW264.7 murine macrophages that were treated with different concentrations of JCEO (0.6, 0.3, and 0.16 mg/mL), or LPS (1μg/mL) as a positive control, versus the negative untreated macrophages. The results indicate that there was a significant increase in TNF-α in LPS-activated RAW264.7 cells compared to JCEO (0.6, 0.3, 0.16mg/mL) treated cells and to the negative untreated control (****p <0.0001), as seen in Fig 4. In addition, there was an increase in TNF-α concentration in 0.6 mg/mL JCEO treated cells compared to the untreated macrophages.
[Figure omitted. See PDF.]
This experiment was conducted using three replicates for each treatment (*p <0.05, ****p <0.0001). Data in the figure represent mean± SD.
The DPPH radical scavenging assay
DPPH radical scavenging assay was conducted to examine the effect of different concentrations of JCEO (0.78, 1.6, 3.13, 6.25, 12.5, 25, 50, 100 mg/mL) as antioxidants. As can be seen in Fig 5, the highest free radical scavenging activity was shown by JCEO with a concentration of 100mg/mL (98.7%) followed by 50mg/mL (84.4%), 25mg/mL (60.4%), and 12.5 mg/mL (52.2%). It was clear that a reduction in JCEO concentrations resulted in a decrease in antioxidant activity. However, the concentrations of JCEO below 6.26 mg/mL showed no significant change.
[Figure omitted. See PDF.]
Each value was conducted using four replicates for each concentration tested. Data in the figure represented as mean± SD.
Cytokine release assessment
The immuno-modulatory effect of different concentrations of JCEO (0.6, 0.3, and 0.16mg/mL) was evaluated by measuring levels of INF-γ (Fig 6A), IL-10 (Fig 6B), IL-4 (Fig 6C), and IL-2 (Fig 6D) using the TH1/TH2 assay kit. The treatment of JCEO induced changes in cytokine levels in the supernatant of RAW264,7 macrophages. Briefly, 0.3 mg/mL JCEO was able to increase IL‐10, IL-4, and IL-2 levels in RAW264.7 murine macrophages compared to 0.6 and 0.16 JCEO treated cells (**p <0.01, ***p <0.001), as shown in Fig 6B–6D, respectively. In addition, 0.16 mg/mL JCEO treated cells increased IL-2 levels in the supernatant of macrophages compared to 0.6mg/mL of JCEO (**p <0.01), as shown in Fig 6D.
[Figure omitted. See PDF.]
A) INF-γ, B) IL-10, C) IL-4, D) IL-2. This experiment was conducted using a duplicate for each treatment (**p <0.01, ***p <0.001). Data in the figure represented as mean± SD.
Phagocytosis assay–fluorescence imaging
Phagocytosis activity of RAW264.7 cells was measured using fluorescence intensity of phagocytosis after treatment of JCEO with different concentrations (0.16, 0.3, 0.6 mg/mL) when compared to the negative untreated control and the LPS positive control. The 0.3mg/mL concentration of JCEO was seen to have a higher significant mean of fluorescence intensity compared to untreated control and similar intensity to the LPS positive control as shown in Fig 7A (****p <0.0001). Confocal imaging in Fig 7B, confirms the phagocytosis induction in comparison to the control.
[Figure omitted. See PDF.]
A) The mean fluorescence intensity of pHrodo™ Red E. coli BioParticles™ after phagocytosis. This experiment was conducted using a duplicate for each treatment. Data in the figure is represented as mean± SD, ****p <0.0001. B) Confocal microscopy images were taken at 10x magnification using confocal LSM 780.
Nitrate concentration assay
Different concentrations of JCEO (0.16, 0.4, 0.6 mg/mL) were investigated for the regulation of NO production in LPS-stimulated macrophages, since it is vital to regulate the overproduction of NO, which generates pro-inflammatory responses, in pro-inflammatory conditions. The NO levels were determined using the Griess reagent. RAW264.7 cells were treated with 1 μg/mL of LPS as positive control whereas negative control cells were left untreated. NO production was calculated as nitrite concentration in the supernatant of cells. It was noticed that different concentrations of JCEO (0.6, .3, 0.16 mg/mL) inhibited the production of NO compared to LPS-treated cells (*p <0.05), as demonstrated in Fig 8.
[Figure omitted. See PDF.]
This experiment was conducted using a duplicate for each treatment (*p <0.05). Data in the figure are represented as mean± SD.
Discussion
Over the last decade, essential oils have gained attention as a source of bioactive compounds, with many potential health benefits for the essential oil and their bioactive constituents [32–35]. Scholars across the world have attempted to study the chemical composition of essential oils from coriander seeds, using different methodologies [36]. The essential oil content is around 0.18 to 1.40% (v/w) of the coriander seed [37]. The major component that has been previously reported in the oil is linalool, in the range of 60–70% of total seed oil [37,38]. That range is dependent on the region of cultivation, degree of fruit ripening, and time of harvesting [37,39–43]. As seen in our results, Jordanian coriander essential oil (JCEO) extracted from seeds had a high scavenging percentile of free radicals at higher concentrations reaching up to 100mg/mL of oil concentration with 98.7% percentile and decreases with reducing the concentration of JCEO. This shows how our test results are in alignment with previous data on antioxidant activity showing that some essential oils like Allium cepa L., Citrus aurantium, Myrtus communis, Coriandrum sativum L., and Eucalyptus oleosa revealed a clear antioxidant potential but displayed different ranges of efficacy [40,43]. Essential oils extracted from aerial parts of coriander, specifically the one cultivated in European countries, possessed the highest free radical scavenging capacity using the DPPH scavenging assay [44–48]. The findings of our study suggest that the essential oil from Jordanian coriander seeds in particular could be of interest in the potential prevention of lipid oxidation and binding to reactive oxygen species (ROS) in the human body, preventing oxidation damage or oxidative stress [48,49]. Such natural antioxidants could serve as physical barriers, inhibiting the access of ROS to biological sites and binding to damaging metallic ions [48]. Moreover, linalool is often conveyed as the main factor of the coriander essential oil bioactivity [49], as it was previously proposed that mono- and sesquiterpenes present in the essential oil were responsible for the antioxidant and anti-inflammatory activities [49,50].
JCEO significantly induced an anti-inflammatory effect that was seen in cytokine assay through monitoring the release of IL-10 and IL-4 in the supernatant of treated murine macrophages, where 0.3mg/mL of JCEO had the most noteworthy upstroke of cytokines tested. In addition, LPS-induced macrophages releasing NO were treated with 0.3mg/mL and 0.16mg/mL JCEO having the most limiting concentrations to the release of NO into supernatant. Meanwhile, TNF-α and INF-γ have been seen to be low and insignificant levels in our study outcomes. This can be allied to many studies that have confirmed that reactive oxygen species (ROS) are involved in response to inflammation [51,52]. Thus, the enhanced generation of ROS exacerbates inflammation and leads to tissue injury; therefore, preventing the NF-κB signaling pathway, MAPK pathways, TNF-α, INF-γ, and ROS generation might be a potential strategy to attenuate inflammatory diseases [51,53]. Our results are in agreement with results from a study about coriander oil components showing high anti-inflammatory activity with IC50 of 6.25μM [51,52]. This study demonstrated an inhibitory effect of JCEO on nitrite oxide (NO) level, the LPS-stimulated generations of ROS, and the inflammatory cytokines like IL-6 and TNF-α [51,52]. Moreover, the use of coriander as an anti-inflammatory agent is marked in one of the traditional formulations from Sri Lanka, containing coriander seeds as one of its primary components. Administration of this formula inhibited carrageenan-induced rat paw edema and augmented pain tolerance in rats by 57% after 1-hr treatment as evaluated by the hot plate test [35,54–57]. This formula was examined on patients suffering from rheumatoid arthritis for 3 months and it was noted that there was an improvement in pain, inflammation, and mobility without any adverse effects on liver functions and gastrointestinal functions [57]. Another study on a multi-herbal formulation, consisting of coriander as one of the constituents, confirmed the inhibitory effect against inflammatory bowel disease [58]. The topical anti-inflammatory effect of coriander oil was also reported by 40 human volunteers using formulation as lipo-lotion supplemented with 0.5% and 1% of coriander oil. CEO-supplemented Lipo-lotion effectively lowered the UV-induced erythema but was less effective than hydrocortisone. Yet, coriander oil showed a minor anti-inflammatory effect with good skin tolerance at both concentrations [53].
It was comprehended in our study that the treatment of JCEO was safe for RAW264.7 murine macrophages’ viability below concentrations of 0.6mg/mL. At a concentration of 0.3mg/mL, it was perceived as nontoxic and might be considered as metabolic enhancer according to MTT results- in terms of number of cells and/or intracellular activity. This was correlated to the scratch-gap closure overnight using the scratch wound assay. For the viability results, usually MTT dye is reduced by active mitochondria in living cells [59], however, some data suggest the reduction process occurs in the intracellular vesicles (such as endosomes or lysosomes), which later contribute to the needle-like aggregates of MTT formazan at the cell surface [59,60]. In our study results pertaining the high surge in the MTT assay at 0.3mg/mL JCEO treatment suggesting higher intracellular metabolism activity [59,60].
To consider coriander essential oil safety, there have been inconsistent reports on the mutagenicity of coriander components. As reported in one study, coriander ethanolic seeds extract failed to show any mutagenicity in rat embryo fibroblasts [61]. The principal component of coriander essential oil, linalool, was found to be antimutagenic at 25 mg/mL, and it did not encourage any chromosomal alterations in the Chinese hamster fibroblast assay [62]. Yet, the mutagenicity of coriander has been reported with an extract of coriander fruit showing mutagenic activity in the Ames assay involving some Salmonella typhimurium strains (TA98 and TA100) [61,63,64]. On the other hand, for the toxicity associated with the topical application of the coriander oil and its components, there seems to be adequate suggestions that the essential oil does not irritate the skin, not even after applying it for 48-hrs in a closed patch test with human volunteers [61,65,66].
To conclude, it is important to consider the presence of diverse range of compounds in the CEO which act in synergism to produce such bioactivities [67]. In that regard, examining the mechanisms of action of the JCEO constituents, may help towards the understanding of the interconnection between the composition and the related biological activity.
Conclusion
The seed-extracted JCEO demonstrated a significant dose-response in free radical scavenging, reaching up to 98.7% at 100 mg/mL. Additionally, JCEO exhibited strong anti-inflammatory effects, with 0.3 mg/mL inducing the highest cytokine release. It also significantly inhibited LPS-induced NO and TNF-α production at 0.3 mg/mL and 0.16 mg/mL, respectively. Notably, 0.3 mg/mL of JCEO was the least toxic to RAW264.7 murine macrophages and may act as a potent anti-inflammatory agent. Collectively, these findings highlight JCEO’s potential as a therapeutic compound with strong antioxidant and anti-inflammatory properties.
Supporting information
S1 File.
https://doi.org/10.1371/journal.pone.0297250.s001
(DOC)
Acknowledgments
The authors would like to thank the Applied Science Private University of Jordan for supporting this work.
References
1. 1. Farnsworth N.R. and Soejarto D.D., Global importance of medicinal plants. The conservation of medicinal plants, 1991. 26(26): p. 25–51.
* View Article
* Google Scholar
2. 2. Xie T., et al., Review of natural product databases. Cell proliferation, 2015. 48(4): p. 398–404. pmid:26009974
* View Article
* PubMed/NCBI
* Google Scholar
3. 3. Dakhlaoui S., et al., Combined effect of essential oils from Lavender (Lavandula officinalis L.) aerial parts and coriander (Coriandrum sativum L.) seeds on antioxidant, anti-diabetic, anti-cancer and anti-inflammatory activities. Journal of Essential Oil Bearing Plants, 2022. 25(1): p. 188–199.
* View Article
* Google Scholar
4. 4. Dillasamola D., Aldi Y., and Kolobinti M., The effect of coriander ethanol extract (Coriandrum sativum L.) against phagocytosis activity and capacity of the macrophage cells and the percentage of leukocyte cells in white male mice. Pharmacognosy Journal, 2019. 11(6).
* View Article
* Google Scholar
5. 5. Das S., Pradhan C., and Pillai D., Dietary coriander (Coriandrum sativum L) oil improves antioxidant and anti-inflammatory activity, innate immune responses and resistance to Aeromonas hydrophila in Nile tilapia (Oreochromis niloticus). Fish & Shellfish Immunology, 2023. 132: p. 108486. pmid:36513321
* View Article
* PubMed/NCBI
* Google Scholar
6. 6. Mechchate H., et al., Antioxidant, anti-inflammatory and antidiabetic proprieties of LC-MS/MS identified polyphenols from coriander seeds. Molecules, 2021. 26(2): p. 487. pmid:33477662
* View Article
* PubMed/NCBI
* Google Scholar
7. 7. Pathak Nimish L., et al., Phytopharmacological properties of Coriander sativum as a potential medicinal tree: an overview. J Appl Pharm Sci, 2011. 1(4): p. 20–5.
* View Article
* Google Scholar
8. 8. Ahmed E.H., Abadi R.S., and Mohammed A.M., Phytochemical screening, chemical composition and antioxidant activity of seeds essential oil of Coriandrum sativum L. from the Sudan. International journal of Herbal medicine, 2018. 6(1): p. 1–4.
* View Article
* Google Scholar
9. 9. Asgarpanah J. and Kazemivash N., Phytochemistry, pharmacology and medicinal properties of Coriandrum sativum L. African Journal of Pharmacy and Pharmacology, 2012. 6(31): p. 2340–2345.
* View Article
* Google Scholar
10. 10. Padmaa M., Coriandrum sativum linn: a review. Pharmacol online Newsl, 2009. 3: p. 561–573.
* View Article
* Google Scholar
11. 11. Kumar V.M., et al., Protective role of Coriandrum sativum seed extract against lead-induced oxidative stress in rat liver and kidney. Current Trends in Biotechnology and Pharmacy, 2013. 7(2): p. 650–664.
* View Article
* Google Scholar
12. 12. Sharma V., Kansal L., and Sharma A., Prophylactic efficacy of Coriandrum sativum (Coriander) on testis of lead-exposed mice. Biological trace element research, 2010. 136: p. 337–354. pmid:19902160
* View Article
* PubMed/NCBI
* Google Scholar
13. 13. Chatterjee S., Oxidative stress, inflammation, and disease, in Oxidative stress and biomaterials. 2016, Elsevier. p. 35–58.
14. 14. Nakkala J.R., et al., Immunomodulatory biomaterials and their application in therapies for chronic inflammation-related diseases. Acta Biomaterialia, 2021. 123: p. 1–30. pmid:33484912
* View Article
* PubMed/NCBI
* Google Scholar
15. 15. Pérez-Torres I., Guarner-Lans V., and Rubio-Ruiz M.E., Reductive stress in inflammation-associated diseases and the pro-oxidant effect of antioxidant agents. International journal of molecular sciences, 2017. 18(10): p. 2098. pmid:28981461
* View Article
* PubMed/NCBI
* Google Scholar
16. 16. Cárdeno A., et al., The unsaponifiable fraction of extra virgin olive oil promotes apoptosis and attenuates activation and homing properties of T cells from patients with inflammatory bowel disease. Food chemistry, 2014. 161: p. 353–360. pmid:24837962
* View Article
* PubMed/NCBI
* Google Scholar
17. 17. Lee J.-W., et al., Anti-inflammatory effect of Rhodiola crenulata extracts through the down-regulation of MyD88 dependent pathway and induction of autophagy. Journal of Functional Foods, 2020. 64: p. 103703.
* View Article
* Google Scholar
18. 18. Golia E., et al., Inflammation and cardiovascular disease: from pathogenesis to therapeutic target. Current atherosclerosis reports, 2014. 16: p. 1–7. pmid:25037581
* View Article
* PubMed/NCBI
* Google Scholar
19. 19. Daddaoua A., et al., The nutritional supplement Active Hexose Correlated Compound (AHCC) has direct immunomodulatory actions on intestinal epithelial cells and macrophages involving TLR/MyD88 and NF-κB/MAPK activation. Food chemistry, 2013. 136(3–4): p. 1288–1295.
* View Article
* Google Scholar
20. 20. Campestrini L.H., et al., Alkaline conditions better extract anti-inflammatory polysaccharides from winemaking by-products. Food research international, 2020. 131: p. 108532. pmid:32247498
* View Article
* PubMed/NCBI
* Google Scholar
21. 21. Hoskin R.T., et al., Blueberry polyphenol-protein food ingredients: the impact of spray drying on the in vitro antioxidant activity, anti-inflammatory markers, glucose metabolism and fibroblast migration. Food chemistry, 2019. 280: p. 187–194. pmid:30642485
* View Article
* PubMed/NCBI
* Google Scholar
22. 22. Kim E.-A., et al., Tuberatolide B isolated from Sargassum macrocarpum inhibited LPS-stimulated inflammatory response via MAPKs and NF-κB signaling pathway in RAW264. 7 cells and zebrafish model. Journal of Functional Foods, 2019. 52: p. 109–115.
* View Article
* Google Scholar
23. 23. Chansiw N., et al., Quercetin-rich ethanolic extract of Polygonum odoratum var pakphai leaves decreased gene expression and secretion of pro-inflammatory mediators in lipopolysaccharide-induced murine RAW264. 7 macrophages. Molecules, 2022. 27(12): p. 3657. pmid:35744785
* View Article
* PubMed/NCBI
* Google Scholar
24. 24. El-Sayed S. and Ahmed S.Y., Effects of coriander seeds powder (Coriandrum sativum) as feed supplements on growth performance parameters and immune response in albino rats. Int. J. Livest. Res, 2017. 7(2): p. 54–63.
* View Article
* Google Scholar
25. 25. Afifi F., Kasabri V., and Abaza I., GC-MS composition and antiproliferative activity of Inula graveolens (L.) Desf. essential oil. Arabian Journal of Medicinal and Aromatic Plants, 2015. 1(1): p. 57–66.
* View Article
* Google Scholar
26. 26. Hasen E., The Potential Effects of the Essential Oil of Coriander Seeds on Bacterial Biofilm and Immune Cells. Jordan Journal of Pharmaceutical Sciences, 2023. 16(2): p. 473–473.
* View Article
* Google Scholar
27. 27. Bag A. and Chattopadhyay R.R., Evaluation of synergistic antibacterial and antioxidant efficacy of essential oils of spices and herbs in combination. PloS one, 2015. 10(7): p. e0131321. pmid:26132146
* View Article
* PubMed/NCBI
* Google Scholar
28. 28. Brand-Williams W., Cuvelier M.-E., and Berset C., Use of a free radical method to evaluate antioxidant activity. LWT-Food science and Technology, 1995. 28(1): p. 25–30.
* View Article
* Google Scholar
29. 29. Brand-Williams W., Use of a free radical method to evaluate antioxidant activity. Food Sci. Technol., 1999. 28: p. 1231–1237.
* View Article
* Google Scholar
30. 30. Dahal R.H., et al., Development of multifunctional cosmetic cream using bioactive materials from Streptomyces sp. T65 with synthesized mesoporous silica particles SBA-15. Antioxidants, 2020. 9(4): p. 278.
* View Article
* Google Scholar
31. 31. Dahal R.H., Shim D.S., and Kim J., Development of actinobacterial resources for functional cosmetics. Journal of Cosmetic Dermatology, 2017. 16(2): p. 243–252. pmid:28097821
* View Article
* PubMed/NCBI
* Google Scholar
32. 32. Ni Z.-J., et al., Recent updates on the chemistry, bioactivities, mode of action, and industrial applications of plant essential oils. Trends in Food Science & Technology, 2021. 110: p. 78–89.
* View Article
* Google Scholar
33. 33. Yeshi K. and Wangchuk P., Essential oils and their bioactive molecules in healthcare, in Herbal biomolecules in healthcare applications. 2022, Elsevier. p. 215–237.
34. 34. Patil B.S., et al., Bioactive compounds: historical perspectives, opportunities, and challenges. Journal of agricultural and food chemistry, 2009. 57(18): p. 8142–8160. pmid:19719126
* View Article
* PubMed/NCBI
* Google Scholar
35. 35. Sahib N.G., et al., Coriander (Coriandrum sativum L.): A potential source of high‐value components for functional foods and nutraceuticals‐A review. Phytotherapy Research, 2013. 27(10): p. 1439–1456. pmid:23281145
* View Article
* PubMed/NCBI
* Google Scholar
36. 36. Singh G., et al., Studies on essential oils, Part 41. Chemical composition, antifungal, antioxidant and sprout suppressant activities of coriander (Coriandrum sativum) essential oil and its oleoresin. Flavour and fragrance journal, 2006. 21(3): p. 472–479.
* View Article
* Google Scholar
37. 37. Al-Khayri J.M., et al., Essential Oil from Coriandrum sativum: A review on Its Phytochemistry and Biological Activity. Molecules, 2023. 28(2). pmid:36677754
* View Article
* PubMed/NCBI
* Google Scholar
38. 38. Hasen E., et al., Synergistic Effect of Coriander Sativum Essential Oil and Gentamicin against Biofilm Formation of Some Pathogenic Bacteria. Jordan Journal of Biological Sciences 2024. 17(4).
* View Article
* Google Scholar
39. 39. Micić D., et al., Essential oils of coriander and sage: Investigation of chemical profile, thermal properties and QSRR analysis. Industrial Crops and Products, 2019. 138: p. 111438.
* View Article
* Google Scholar
40. 40. Ayanoğlu F., et al., Seed yields, yield components and essential oil of selected coriander (Coriandrum sativum L.) lines. Journal of Herbs, Spices & Medicinal Plants, 2002. 9(2–3): p. 71–76.
* View Article
* Google Scholar
41. 41. Ramezani S., Rasouli F., and Solaimani B., Changes in essential oil content of coriander (Coriandrum sativum L.) aerial parts during four phonological stages in Iran. Journal of essential oil bearing plants, 2009. 12(6): p. 683–689.
* View Article
* Google Scholar
42. 42. Wahba H.E., Abd Rabbu H.S., and Ibrahim M.E., Evaluation of essential oil isolated from dry coriander seeds and recycling of the plant waste under different storage conditions. Bulletin of the National Research Centre, 2020. 44(1): p. 192.
* View Article
* Google Scholar
43. 43. Raveau R., et al., Chemical composition, antioxidant and anti-inflammatory activities of clary sage and coriander essential oils produced on polluted and amended soils-phytomanagement approach. Molecules, 2021. 26(17): p. 5321. pmid:34500751
* View Article
* PubMed/NCBI
* Google Scholar
44. 44. Berka-Zougali B., et al., Comparative study of essential oils extracted from Algerian Myrtus communis L. leaves using microwaves and hydrodistillation. International journal of molecular sciences, 2012. 13(4): p. 4673–4695. pmid:22606003
* View Article
* PubMed/NCBI
* Google Scholar
45. 45. Hsouna A.B., et al., Characterization of essential oil from Citrus aurantium L. flowers: antimicrobial and antioxidant activities. Journal of Oleo Science, 2013. 62(10): p. 763–772. pmid:24088513
* View Article
* PubMed/NCBI
* Google Scholar
46. 46. Marzoug H.N.B., et al., Eucalyptus oleosa essential oils: chemical composition and antimicrobial and antioxidant activities of the oils from different plant parts (stems, leaves, flowers and fruits). Molecules, 2011. 16(2): p. 1695–1709. pmid:21330958
* View Article
* PubMed/NCBI
* Google Scholar
47. 47. Ye C.-L., Dai D.-H., and Hu W.-L., Antimicrobial and antioxidant activities of the essential oil from onion (Allium cepa L.). Food control, 2013. 30(1): p. 48–53.
* View Article
* Google Scholar
48. 48. Miguel M.G., Antioxidant and anti-inflammatory activities of essential oils: a short review. Molecules, 2010. 15(12): p. 9252–9287. pmid:21160452
* View Article
* PubMed/NCBI
* Google Scholar
49. 49. Duarte A., et al., Antioxidant properties of coriander essential oil and linalool and their potential to control Campylobacter spp. Food Control, 2016. 61: p. 115–122.
* View Article
* Google Scholar
50. 50. Kumar R., Sharma S., and Pathania V., Effect of shading and plant density on growth, yield and oil composition of clary sage (Salvia sclarea L.) in north western Himalaya. Journal of essential oil research, 2013. 25(1): p. 23–32.
* View Article
* Google Scholar
51. 51. Yuan R., et al., Novel compounds in fruits of coriander (Coşkuner & Karababa) with anti-inflammatory activity. Journal of Functional Foods, 2020. 73: p. 104145.
* View Article
* Google Scholar
52. 52. Wu T.-T., et al., Suppressive effects of extracts from the aerial part of Coriandrum sativum L. on LPS-induced inflammatory responses in murine RAW 264.7 macrophages. Journal of the Science of Food and Agriculture, 2010. 90(11): p. 1846–1854. pmid:20549653
* View Article
* PubMed/NCBI
* Google Scholar
53. 53. Reuter J., et al., Anti‐inflammatory potential of a lipolotion containing coriander oil in the ultraviolet erythema test. JDDG: Journal der Deutschen Dermatologischen Gesellschaft, 2008. 6(10): p. 847–851. pmid:18371049
* View Article
* PubMed/NCBI
* Google Scholar
54. 54. Patwardhan, B., EVIDENCE BASED AYURVEDA AND HERBAL MEDICINE.
55. 55. Ksouri R., et al., Medicinal halophytes: potent source of health promoting biomolecules with medical, nutraceutical and food applications. Critical reviews in biotechnology, 2012. 32(4): p. 289–326. pmid:22129270
* View Article
* PubMed/NCBI
* Google Scholar
56. 56. Bashir S. and Safdar A., Coriander seeds: ethno-medicinal, phytochemical and pharmacological profile. Science of Spices and Culinary Herbs-Latest Laboratory, Pre-clinical, and Clinical Studies, 2020. 2: p. 39–64.
* View Article
* Google Scholar
57. 57. Thabrew M.I., Dharmasiri M., and Senaratne L., Anti-inflammatory and analgesic activity in the polyherbal formulation Maharasnadhi Quathar. Journal of ethnopharmacology, 2003. 85(2–3): p. 261–267. pmid:12639750
* View Article
* PubMed/NCBI
* Google Scholar
58. 58. Jagtap A., Shirke S., and Phadke A., Effect of polyherbal formulation on experimental models of inflammatory bowel diseases. Journal of ethnopharmacology, 2004. 90(2–3): p. 195–204. pmid:15013181
* View Article
* PubMed/NCBI
* Google Scholar
59. 59. Berridge M.V., Herst P.M., and Tan A.S., Tetrazolium dyes as tools in cell biology: new insights into their cellular reduction. Biotechnology annual review, 2005. 11: p. 127–152. pmid:16216776
* View Article
* PubMed/NCBI
* Google Scholar
60. 60. Liu Y., et al., Mechanism of cellular 3‐(4, 5‐dimethylthiazol‐2‐yl)‐2, 5‐diphenyltetrazolium bromide (MTT) reduction. Journal of neurochemistry, 1997. 69(2): p. 581–593. pmid:9231715
* View Article
* PubMed/NCBI
* Google Scholar
61. 61. Burdock G.A. and Carabin I.G., Safety assessment of coriander (Coriandrum sativum L.) essential oil as a food ingredient. Food and chemical Toxicology, 2009. 47(1): p. 22–34. pmid:19032971
* View Article
* PubMed/NCBI
* Google Scholar
62. 62. Ishidate M. Jr., et al., Primary mutagenicity screening of food additives currently used in Japan. Food Chem Toxicol, 1984. 22(8): p. 623–36. pmid:6381265
* View Article
* PubMed/NCBI
* Google Scholar
63. 63. Bhat R., Mutagenic/Antimutagenic potential of Spices of common use in Indian diet. 2003, Goa University.
64. 64. Hong C.-E., Ji S.-T., and Lyu S.-Y., Absence of mutagenicity in three Nigerian medicinal plants-bidens pilosa, Cleistopholis paterns and Tetrapleura tetraptera. Tropical Journal of Pharmaceutical Research, 2011. 10(2).
* View Article
* Google Scholar
65. 65. Eidi M. and Eidi A., Effect of Coriander (Coriandrum sativum L.) seed ethanol extract in experimental diabetes, in Nuts and Seeds in Health and Disease Prevention. 2011, Elsevier. p. 395–400.
66. 66. Api A.M., Research Institute for Fragrance Materials, in Encyclopedia of Toxicology (Third Edition), Wexler P., Editor. 2014, Academic Press: Oxford. p. 93.
67. 67. Freires I.d.A., et al., Coriandrum sativum L.(coriander) essential oil: antifungal activity and mode of action on Candida spp., and molecular targets affected in human whole-genome expression. PLoS One, 2014. 9(6): p. e99086. pmid:24901768
* View Article
* PubMed/NCBI
* Google Scholar
Citation: Omar A, Barakat M, Alzaghari LF, Abdulrazzaq SB, Hasen E, Chellappan DK, et al. (2024) The effect of Jordanian essential oil from coriander seeds on antioxidant, anti-inflammatory, and immunostimulatory activities using RAW 246.7 murine macrophages. PLoS ONE 19(8): e0297250. https://doi.org/10.1371/journal.pone.0297250
About the Authors:
Amin Omar
Roles: Conceptualization, Investigation, Methodology, Project administration, Writing – original draft, Writing – review & editing
Affiliation: Faculty of Pharmacy, Department of Pharmaceutics and Pharmaceutical Sciences, Applied Science Private University, Amman, Jordan
Muna Barakat
Roles: Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Project administration, Resources, Software, Supervision, Validation, Visualization, Writing – original draft, Writing – review & editing
E-mail: [email protected]
Affiliation: Faculty of Pharmacy, Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman, Jordan
ORICD: https://orcid.org/0000-0002-7966-1172
Lujain F. Alzaghari
Roles: Data curation, Formal analysis, Investigation, Methodology, Project administration, Visualization, Writing – original draft, Writing – review & editing
Affiliation: Faculty of Pharmacy, Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman, Jordan
Shaymaa B. Abdulrazzaq
Roles: Data curation, Formal analysis, Methodology, Writing – original draft, Writing – review & editing
Affiliation: Department of Chemical and Pharmaceutical Sciences and Biotechnology, Chemical Sciences Division, Chemistry Interdisciplinary Project (ChIP), University of Camerino, Camerino (Macerata), Italy
Eliza Hasen
Roles: Investigation, Methodology, Writing – original draft, Writing – review & editing
Affiliation: Faculty of Pharmacy, Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman, Jordan
Dinesh Kumar Chellappan
Roles: Validation, Visualization, Writing – original draft, Writing – review & editing
Affiliation: Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
Mohammad A. A. Al-Najjar
Roles: Conceptualization, Investigation, Methodology, Project administration, Resources, Software, Supervision, Validation, Visualization, Writing – original draft, Writing – review & editing
Affiliation: Faculty of Pharmacy, Department of Pharmaceutics and Pharmaceutical Sciences, Applied Science Private University, Amman, Jordan
ORICD: https://orcid.org/0000-0003-0001-6902
[/RAW_REF_TEXT]
[/RAW_REF_TEXT]
[/RAW_REF_TEXT]
[/RAW_REF_TEXT]
[/RAW_REF_TEXT]
[/RAW_REF_TEXT]
1. Farnsworth N.R. and Soejarto D.D., Global importance of medicinal plants. The conservation of medicinal plants, 1991. 26(26): p. 25–51.
2. Xie T., et al., Review of natural product databases. Cell proliferation, 2015. 48(4): p. 398–404. pmid:26009974
3. Dakhlaoui S., et al., Combined effect of essential oils from Lavender (Lavandula officinalis L.) aerial parts and coriander (Coriandrum sativum L.) seeds on antioxidant, anti-diabetic, anti-cancer and anti-inflammatory activities. Journal of Essential Oil Bearing Plants, 2022. 25(1): p. 188–199.
4. Dillasamola D., Aldi Y., and Kolobinti M., The effect of coriander ethanol extract (Coriandrum sativum L.) against phagocytosis activity and capacity of the macrophage cells and the percentage of leukocyte cells in white male mice. Pharmacognosy Journal, 2019. 11(6).
5. Das S., Pradhan C., and Pillai D., Dietary coriander (Coriandrum sativum L) oil improves antioxidant and anti-inflammatory activity, innate immune responses and resistance to Aeromonas hydrophila in Nile tilapia (Oreochromis niloticus). Fish & Shellfish Immunology, 2023. 132: p. 108486. pmid:36513321
6. Mechchate H., et al., Antioxidant, anti-inflammatory and antidiabetic proprieties of LC-MS/MS identified polyphenols from coriander seeds. Molecules, 2021. 26(2): p. 487. pmid:33477662
7. Pathak Nimish L., et al., Phytopharmacological properties of Coriander sativum as a potential medicinal tree: an overview. J Appl Pharm Sci, 2011. 1(4): p. 20–5.
8. Ahmed E.H., Abadi R.S., and Mohammed A.M., Phytochemical screening, chemical composition and antioxidant activity of seeds essential oil of Coriandrum sativum L. from the Sudan. International journal of Herbal medicine, 2018. 6(1): p. 1–4.
9. Asgarpanah J. and Kazemivash N., Phytochemistry, pharmacology and medicinal properties of Coriandrum sativum L. African Journal of Pharmacy and Pharmacology, 2012. 6(31): p. 2340–2345.
10. Padmaa M., Coriandrum sativum linn: a review. Pharmacol online Newsl, 2009. 3: p. 561–573.
11. Kumar V.M., et al., Protective role of Coriandrum sativum seed extract against lead-induced oxidative stress in rat liver and kidney. Current Trends in Biotechnology and Pharmacy, 2013. 7(2): p. 650–664.
12. Sharma V., Kansal L., and Sharma A., Prophylactic efficacy of Coriandrum sativum (Coriander) on testis of lead-exposed mice. Biological trace element research, 2010. 136: p. 337–354. pmid:19902160
13. Chatterjee S., Oxidative stress, inflammation, and disease, in Oxidative stress and biomaterials. 2016, Elsevier. p. 35–58.
14. Nakkala J.R., et al., Immunomodulatory biomaterials and their application in therapies for chronic inflammation-related diseases. Acta Biomaterialia, 2021. 123: p. 1–30. pmid:33484912
15. Pérez-Torres I., Guarner-Lans V., and Rubio-Ruiz M.E., Reductive stress in inflammation-associated diseases and the pro-oxidant effect of antioxidant agents. International journal of molecular sciences, 2017. 18(10): p. 2098. pmid:28981461
16. Cárdeno A., et al., The unsaponifiable fraction of extra virgin olive oil promotes apoptosis and attenuates activation and homing properties of T cells from patients with inflammatory bowel disease. Food chemistry, 2014. 161: p. 353–360. pmid:24837962
17. Lee J.-W., et al., Anti-inflammatory effect of Rhodiola crenulata extracts through the down-regulation of MyD88 dependent pathway and induction of autophagy. Journal of Functional Foods, 2020. 64: p. 103703.
18. Golia E., et al., Inflammation and cardiovascular disease: from pathogenesis to therapeutic target. Current atherosclerosis reports, 2014. 16: p. 1–7. pmid:25037581
19. Daddaoua A., et al., The nutritional supplement Active Hexose Correlated Compound (AHCC) has direct immunomodulatory actions on intestinal epithelial cells and macrophages involving TLR/MyD88 and NF-κB/MAPK activation. Food chemistry, 2013. 136(3–4): p. 1288–1295.
20. Campestrini L.H., et al., Alkaline conditions better extract anti-inflammatory polysaccharides from winemaking by-products. Food research international, 2020. 131: p. 108532. pmid:32247498
21. Hoskin R.T., et al., Blueberry polyphenol-protein food ingredients: the impact of spray drying on the in vitro antioxidant activity, anti-inflammatory markers, glucose metabolism and fibroblast migration. Food chemistry, 2019. 280: p. 187–194. pmid:30642485
22. Kim E.-A., et al., Tuberatolide B isolated from Sargassum macrocarpum inhibited LPS-stimulated inflammatory response via MAPKs and NF-κB signaling pathway in RAW264. 7 cells and zebrafish model. Journal of Functional Foods, 2019. 52: p. 109–115.
23. Chansiw N., et al., Quercetin-rich ethanolic extract of Polygonum odoratum var pakphai leaves decreased gene expression and secretion of pro-inflammatory mediators in lipopolysaccharide-induced murine RAW264. 7 macrophages. Molecules, 2022. 27(12): p. 3657. pmid:35744785
24. El-Sayed S. and Ahmed S.Y., Effects of coriander seeds powder (Coriandrum sativum) as feed supplements on growth performance parameters and immune response in albino rats. Int. J. Livest. Res, 2017. 7(2): p. 54–63.
25. Afifi F., Kasabri V., and Abaza I., GC-MS composition and antiproliferative activity of Inula graveolens (L.) Desf. essential oil. Arabian Journal of Medicinal and Aromatic Plants, 2015. 1(1): p. 57–66.
26. Hasen E., The Potential Effects of the Essential Oil of Coriander Seeds on Bacterial Biofilm and Immune Cells. Jordan Journal of Pharmaceutical Sciences, 2023. 16(2): p. 473–473.
27. Bag A. and Chattopadhyay R.R., Evaluation of synergistic antibacterial and antioxidant efficacy of essential oils of spices and herbs in combination. PloS one, 2015. 10(7): p. e0131321. pmid:26132146
28. Brand-Williams W., Cuvelier M.-E., and Berset C., Use of a free radical method to evaluate antioxidant activity. LWT-Food science and Technology, 1995. 28(1): p. 25–30.
29. Brand-Williams W., Use of a free radical method to evaluate antioxidant activity. Food Sci. Technol., 1999. 28: p. 1231–1237.
30. Dahal R.H., et al., Development of multifunctional cosmetic cream using bioactive materials from Streptomyces sp. T65 with synthesized mesoporous silica particles SBA-15. Antioxidants, 2020. 9(4): p. 278.
31. Dahal R.H., Shim D.S., and Kim J., Development of actinobacterial resources for functional cosmetics. Journal of Cosmetic Dermatology, 2017. 16(2): p. 243–252. pmid:28097821
32. Ni Z.-J., et al., Recent updates on the chemistry, bioactivities, mode of action, and industrial applications of plant essential oils. Trends in Food Science & Technology, 2021. 110: p. 78–89.
33. Yeshi K. and Wangchuk P., Essential oils and their bioactive molecules in healthcare, in Herbal biomolecules in healthcare applications. 2022, Elsevier. p. 215–237.
34. Patil B.S., et al., Bioactive compounds: historical perspectives, opportunities, and challenges. Journal of agricultural and food chemistry, 2009. 57(18): p. 8142–8160. pmid:19719126
35. Sahib N.G., et al., Coriander (Coriandrum sativum L.): A potential source of high‐value components for functional foods and nutraceuticals‐A review. Phytotherapy Research, 2013. 27(10): p. 1439–1456. pmid:23281145
36. Singh G., et al., Studies on essential oils, Part 41. Chemical composition, antifungal, antioxidant and sprout suppressant activities of coriander (Coriandrum sativum) essential oil and its oleoresin. Flavour and fragrance journal, 2006. 21(3): p. 472–479.
37. Al-Khayri J.M., et al., Essential Oil from Coriandrum sativum: A review on Its Phytochemistry and Biological Activity. Molecules, 2023. 28(2). pmid:36677754
38. Hasen E., et al., Synergistic Effect of Coriander Sativum Essential Oil and Gentamicin against Biofilm Formation of Some Pathogenic Bacteria. Jordan Journal of Biological Sciences 2024. 17(4).
39. Micić D., et al., Essential oils of coriander and sage: Investigation of chemical profile, thermal properties and QSRR analysis. Industrial Crops and Products, 2019. 138: p. 111438.
40. Ayanoğlu F., et al., Seed yields, yield components and essential oil of selected coriander (Coriandrum sativum L.) lines. Journal of Herbs, Spices & Medicinal Plants, 2002. 9(2–3): p. 71–76.
41. Ramezani S., Rasouli F., and Solaimani B., Changes in essential oil content of coriander (Coriandrum sativum L.) aerial parts during four phonological stages in Iran. Journal of essential oil bearing plants, 2009. 12(6): p. 683–689.
42. Wahba H.E., Abd Rabbu H.S., and Ibrahim M.E., Evaluation of essential oil isolated from dry coriander seeds and recycling of the plant waste under different storage conditions. Bulletin of the National Research Centre, 2020. 44(1): p. 192.
43. Raveau R., et al., Chemical composition, antioxidant and anti-inflammatory activities of clary sage and coriander essential oils produced on polluted and amended soils-phytomanagement approach. Molecules, 2021. 26(17): p. 5321. pmid:34500751
44. Berka-Zougali B., et al., Comparative study of essential oils extracted from Algerian Myrtus communis L. leaves using microwaves and hydrodistillation. International journal of molecular sciences, 2012. 13(4): p. 4673–4695. pmid:22606003
45. Hsouna A.B., et al., Characterization of essential oil from Citrus aurantium L. flowers: antimicrobial and antioxidant activities. Journal of Oleo Science, 2013. 62(10): p. 763–772. pmid:24088513
46. Marzoug H.N.B., et al., Eucalyptus oleosa essential oils: chemical composition and antimicrobial and antioxidant activities of the oils from different plant parts (stems, leaves, flowers and fruits). Molecules, 2011. 16(2): p. 1695–1709. pmid:21330958
47. Ye C.-L., Dai D.-H., and Hu W.-L., Antimicrobial and antioxidant activities of the essential oil from onion (Allium cepa L.). Food control, 2013. 30(1): p. 48–53.
48. Miguel M.G., Antioxidant and anti-inflammatory activities of essential oils: a short review. Molecules, 2010. 15(12): p. 9252–9287. pmid:21160452
49. Duarte A., et al., Antioxidant properties of coriander essential oil and linalool and their potential to control Campylobacter spp. Food Control, 2016. 61: p. 115–122.
50. Kumar R., Sharma S., and Pathania V., Effect of shading and plant density on growth, yield and oil composition of clary sage (Salvia sclarea L.) in north western Himalaya. Journal of essential oil research, 2013. 25(1): p. 23–32.
51. Yuan R., et al., Novel compounds in fruits of coriander (Coşkuner & Karababa) with anti-inflammatory activity. Journal of Functional Foods, 2020. 73: p. 104145.
52. Wu T.-T., et al., Suppressive effects of extracts from the aerial part of Coriandrum sativum L. on LPS-induced inflammatory responses in murine RAW 264.7 macrophages. Journal of the Science of Food and Agriculture, 2010. 90(11): p. 1846–1854. pmid:20549653
53. Reuter J., et al., Anti‐inflammatory potential of a lipolotion containing coriander oil in the ultraviolet erythema test. JDDG: Journal der Deutschen Dermatologischen Gesellschaft, 2008. 6(10): p. 847–851. pmid:18371049
54. Patwardhan, B., EVIDENCE BASED AYURVEDA AND HERBAL MEDICINE.
55. Ksouri R., et al., Medicinal halophytes: potent source of health promoting biomolecules with medical, nutraceutical and food applications. Critical reviews in biotechnology, 2012. 32(4): p. 289–326. pmid:22129270
56. Bashir S. and Safdar A., Coriander seeds: ethno-medicinal, phytochemical and pharmacological profile. Science of Spices and Culinary Herbs-Latest Laboratory, Pre-clinical, and Clinical Studies, 2020. 2: p. 39–64.
57. Thabrew M.I., Dharmasiri M., and Senaratne L., Anti-inflammatory and analgesic activity in the polyherbal formulation Maharasnadhi Quathar. Journal of ethnopharmacology, 2003. 85(2–3): p. 261–267. pmid:12639750
58. Jagtap A., Shirke S., and Phadke A., Effect of polyherbal formulation on experimental models of inflammatory bowel diseases. Journal of ethnopharmacology, 2004. 90(2–3): p. 195–204. pmid:15013181
59. Berridge M.V., Herst P.M., and Tan A.S., Tetrazolium dyes as tools in cell biology: new insights into their cellular reduction. Biotechnology annual review, 2005. 11: p. 127–152. pmid:16216776
60. Liu Y., et al., Mechanism of cellular 3‐(4, 5‐dimethylthiazol‐2‐yl)‐2, 5‐diphenyltetrazolium bromide (MTT) reduction. Journal of neurochemistry, 1997. 69(2): p. 581–593. pmid:9231715
61. Burdock G.A. and Carabin I.G., Safety assessment of coriander (Coriandrum sativum L.) essential oil as a food ingredient. Food and chemical Toxicology, 2009. 47(1): p. 22–34. pmid:19032971
62. Ishidate M. Jr., et al., Primary mutagenicity screening of food additives currently used in Japan. Food Chem Toxicol, 1984. 22(8): p. 623–36. pmid:6381265
63. Bhat R., Mutagenic/Antimutagenic potential of Spices of common use in Indian diet. 2003, Goa University.
64. Hong C.-E., Ji S.-T., and Lyu S.-Y., Absence of mutagenicity in three Nigerian medicinal plants-bidens pilosa, Cleistopholis paterns and Tetrapleura tetraptera. Tropical Journal of Pharmaceutical Research, 2011. 10(2).
65. Eidi M. and Eidi A., Effect of Coriander (Coriandrum sativum L.) seed ethanol extract in experimental diabetes, in Nuts and Seeds in Health and Disease Prevention. 2011, Elsevier. p. 395–400.
66. Api A.M., Research Institute for Fragrance Materials, in Encyclopedia of Toxicology (Third Edition), Wexler P., Editor. 2014, Academic Press: Oxford. p. 93.
67. Freires I.d.A., et al., Coriandrum sativum L.(coriander) essential oil: antifungal activity and mode of action on Candida spp., and molecular targets affected in human whole-genome expression. PLoS One, 2014. 9(6): p. e99086. pmid:24901768
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
© 2024 Omar et al. This is an open access article distributed under the terms of the Creative Commons Attribution License: http://creativecommons.org/licenses/by/4.0/ (the “License”), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
Abstract
Coriander (Coriandrum sativum L.) is a member of the Umbelliferae/Apiaceae family and one of the well-known essential oil-containing plants, in which the seeds are used in traditional medicine, and as flavoring in food preparation. Knowing the diverse chemical components of different parts of the plant, this work aims to investigate the antioxidant, the anti-inflammatory, and the immunostimulatory modulator effects of the Jordanian C. sativum’s seed extracted essential oil (JCEO). Coriander oil extract was prepared by hydro-distillation method using the Clevenger apparatus. Different concentrations of coriander oil were examined by using DPPH radical scavenging assay, MTT assay, pro-inflammatory cytokine (Tumor Necrosis Factor-TNF-alpha) production in RAW264.7 murine macrophages in addition, scratch-wound assessment, NO level examination, Th1/Th2 assay, phagocytosis assay, and fluorescence imaging using DAPI stain were conducted. JCEO had a potential metabolic enhancer effect at a concentration of 0.3 mg/mL on cell viability with anti-inflammatory activities via increasing cytokines like IL-10, IL-4, and limiting NO, INF-γ, and TNF-α release into cell supernatant. Antioxidant activity was seen significantly at higher concentrations of JCEO reaching 98.7% when using 100mg/mL and minimally reaching 50% at 12.5mg/mL of the essential oil. Treated macrophages were able to attain full scratch closure after 48-hrs at concentrations below 0.3mg/mL. The seed-extracted JCEO showed significant free radical scavenging activity even at lower dilutions. It also significantly induced an anti-inflammatory effect via an increase in the release of cytokines but reduced the LPS-induced NO and TNF-α production at 0.16–0.3mg/mL. In summary, coriander essential oil demonstrated antioxidant, anti-inflammatory, and immunostimulatory effects, showcasing its therapeutic potential at specific concentrations. The findings underscore its safety and metabolic enhancement properties, emphasizing its promising role in promoting cellular health.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer