Next Article in Journal
Physicochemical Properties and Cytoprotective Effects on PC12 Cells of Polysaccharides from Belamcanda chinensis (L.) DC. Obtained via a Gradient Ethanol Precipitation Method
Next Article in Special Issue
Therapeutic Potential of Myricitrin in a db/db Mouse Model of Type 2 Diabetes
Previous Article in Journal
Microwave-Heating-Assisted Synthesis of Ultrathin and Ultralong Hydroxyapatite Nanowires Using Biogenic Creatine Phosphate and Their Derived Flexible Bio-Paper with Drug Delivery Function
Previous Article in Special Issue
Zephycandidine A and Synthetic Analogues—Synthesis and Evaluation of Biological Activity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Chemical Analysis and Antioxidant Activities of Resin Fractions from Pistacia lentiscus L. var. Chia in Neuroblastoma SH-SY5Y Cells

by
Achilleas Georgantopoulos
1,
Foteini D. Kalousi
1,
Federica Pollastro
2,
Ioannis Tsialtas
1,
Natasa P. Kalogiouri
3 and
Anna-Maria G. Psarra
1,*
1
Department of Biochemistry and Biotechnology, University of Thessaly, Biopolis, 41500 Larissa, Greece
2
Department of Pharmaceutical, University of Eastern Piedmont, 28100 Novara, Italy
3
Laboratory of Analytical Chemistry, School of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
*
Author to whom correspondence should be addressed.
Molecules 2025, 30(5), 997; https://doi.org/10.3390/molecules30050997
Submission received: 12 January 2025 / Revised: 14 February 2025 / Accepted: 19 February 2025 / Published: 21 February 2025
(This article belongs to the Special Issue Natural Compounds for Disease and Health II)

Abstract

Chios mastiha is the natural aromatic resin of Pistacia lentiscus L. var. Chia, Anacardiaceae, which is exclusively cultivated in the southern part of the Greek island of Chios. Chios mastiha (P. lenticonus/Chios mastiha) is well-known for its distinctive taste and aroma and has been known since ancient times due to its healing properties in gastrointestinal and inflammatory disorders and because of its anti-bacterial and anti-fungal activities. In this study, the chemical composition, applying LC-QTOF-MS/MS analysis, and the antioxidant activities of three different polarity P. lenticonus/Chios mastiha fractions, apolar, medium polar, and polar, were characterized in human neuroblastoma SH-SY5Y cells. Chemical analysis of the fractions unveiled new components of P. lenticonus/Chios mastiha, mainly fatty acids compounds, known for their antioxidant activity and regulatory effects on lipid metabolism. By applying the MTT assay and confocal microscopy analysis, we showed that P. lenticonus/Chios mastiha fractions, especially the apolar and medium polar fractions, enriched in triterpenes and fatty acids, caused suppression of the H2O2-induced reduction in cell viability, ROS production, and depolarization of the mitochondrial membrane potential, in SH-SY5Y cells. Moreover, Western blot analysis revealed that apolar fraction, enriched in fatty acids, induced expression of the PPARα, which is well-known for its antioxidant activities and its crucial role in lipid metabolism. Induction of PPARα, a GR target gene, was also accompanied by an increase in GR protein levels. Enhanced antioxidant activities of the apolar fraction may be correlated with its chemical composition, enriched in fatty acids and triterpenoids. Thus, our results indicate the neuroprotective actions of P. lenticonus/Chios mastiha fractions, highlighting their potential application as neuroprotective agents in neurodegenerative diseases.

Graphical Abstract

1. Introduction

Pistacia lentiscus L. var. Chia (Mastic Tree) belongs to the Anacardiaceae family. Even though P. lentiscus is distributed throughout the Mediterranean, the Chia variety is exclusively cultivated on the Greek island of Chios, particularly in the southern part of the island, due to the climatic and soil conditions that prevail there. Chios mastiha, or Chios mastic gum (P. lenticonus/Chios mastiha), is the natural aromatic resin derived from the trunk and branches of the Pistacia lentiscus L. var. Chia. P. lenticonus/Chios mastiha has been harvested for over 2500 years; it is widely used as a food flavoring additive and in beverages and it has many applications in producing natural chewing gum, skin-, dental-, and cosmetic products. Most importantly, P. lenticonus/Chios mastiha has been known since ancient times for its healing properties in gastrointestinal and inflammatory disorders [1]. In 2015 P. lenticonus/Chios mastiha was awarded by the European Medicine Agency as a natural medicine for skin inflammation and dyspeptic disorders [1,2]. Moreover, anti-cancer [3,4,5] and cardiovascular protective activities [6,7] of P. lenticonus/Chios mastiha have been proposed. These actions are attributed to the anti-bacterial, anti-fungal [8] anti-inflammatory [9,10,11,12] antilipemic, antioxidant, and apoptotic [7,13,14] activities of P. lenticonus/Chios mastiha and its essential oil.
P. lenticonus/Chios mastiha’s therapeutic actions on gastrointestinal and inflammatory disorders, namely inflammatory bowel diseases, are associated with the P. lenticonus/Chios mastiha’s protective anti-inflammatory effects on intestinal epithelial cells [11,15]. Anti-inflammatory actions of P. lenticonus/Chios mastiha are also beneficial for other cell types [9,10,16], including cardiovascular cells, where the antioxidant activities of P. lenticonus/Chios mastiha are suggested to play a crucial role. Specifically, it has been shown that treatment with P. lenticonus/Chios mastiha improves the total antioxidant status of patients with nonalcoholic fatty liver disease (NAFLD) [17] and the antioxidant activity of oxidized low-density lipoprotein (LDL) in peripheral blood mononuclear cell (PBMC) cells, suppressing the main causative factor of atherosclerosis [6]. Increased reactive oxygen species (ROS) levels are responsible for many pathological conditions primarily for neurodegenerative diseases [18] and cancer. In this context, the apoptotic and antiproliferative properties of P. lenticonus/Chios mastiha in neuroblastoma SH-SY5Y and SK-N-BE cells [19] have been reported. Moreover, due to the fundamental role of mitochondria in ROS production and energy metabolism, maintenance of mitochondria functionality is crucial for both cancer and neurodegenerative diseases [20]. Thus, further investigation of the neuroprotective actions of P. lenticonus/Chios mastiha and its potential anti-cancer activity on neuroblastoma cells, with emphasis on its effect on the preservation of mitochondrial functionality could be of great importance.
Biological activities of P. lenticonus/Chios mastiha are associated with its chemical composition. To this point, chemical analyses of P. lenticonus/Chios mastiha, applying gas spectrometry, mass spectrometry, and NMR analysis, have revealed that the sticky texture of the resin is attributed to a component named β-polymyrcene, which constitutes approximately 25–30% of the total composition. The rest, 65–70% of the P. lenticonus/Chios mastiha is enriched in terpenoids [21,22,23]. Thus, terpenoids such as masticadienonic, isomasticadienonic acid, moronate, oleanolate, and oleanonate, have been identified in the acidic fraction of P. lenticonus/Chios mastiha. Moreover, triterpenic compounds like oleanolic aldehyde, 28-norolean-17-en-3-one, β-amyrone, isomasticadienolic aldehyde, tirucallol, and dammaradienone, lupeol, 24Z-masticadienonic acid methyl ester, 24Z-isomasticadienonic acid methyl ester keto-oleanolic aldehyde, and oleanolic aldehyde, have been detected in neutral fractions or medium polar fractions of P. lenticonus/Chios mastiha [12,14]. In addition, other phenolic and flavonoid compounds such as gallic acid, caffeic acid, α-terpinolene, linalool, benzoic acid, phenylacetic acid, and tyrosol have been detected, although in smaller amounts [12,13,21,24,25].
The structural similarity of triterpenoids to the steroid hormone glucocorticoid that acts through its cognate receptor, the glucocorticoid receptor (GR) [26,27,28], indicates that triterpenoids could interact with glucocorticoid signaling, affecting many glucocorticoid-mediated biological actions, including energy metabolism, inflammation, and apoptosis. In this context, we have recently shown that P. lenticonus/Chios mastiha essential oil, as well as different polarity fractions from Chios Mastic tree leaves and resin, enriched in triterpenoids, could exert anti-inflammatory actions and regulation of apoptosis [12,14,29], affecting both glucocorticoid receptor activity and protein levels. The association of P. lenticonus/Chios mastiha enrichment in triterpenoids and phenol compounds with its anti-inflammatory, anti-microbial, and apoptotic activities is also supported by data in the literature [6,7,30,31,32,33,34,35,36].
In this study, LC-QTOF-MS/MS analysis of three different polarity fractions, apolar (a), medium polar (mp), and polar (p) from P. lenticonus/Chios mastiha, diluted in DMSO, was applied with the aim of identifying additional unidentified P. lenticonus/Chios mastiha’s components of chemical origin, other than terpenoids and phenolic compounds. In addition, the possible neuroprotective actions of P. lenticonus/Chios mastiha’s fractions were evaluated by investigating their possible antioxidant activities, their effect on mitochondrial functionality, and their interference with steroid receptors signaling in human neuroblastoma SH-SY5Y cells. In consequence, assessment of the P. lenticonus/Chios mastiha fractions’ effects on the H2O2-induced reduction in SH-SY5Y cell viability, depolarization of the mitochondrial membrane, ROS production, and regulation of GR and antioxidant factor levels, applying MTT assay, confocal microscopy, and Western blot analysis was performed. To the best of our knowledge, this is the first time that new compounds such as palmitic, stearic, and myristic acids in P. lenticonus/Chios mastiha’s different polarity fractions were detected. Notably, the study revealed the presence of 37 compounds in P. lenticonus/Chios mastiha’s fractions, identified through suspect and non-target screening. Most importantly, the antioxidant activity of P. lenticonus/Chios mastiha fractions in SH-SY5Y cells was revealed, highlighting their potential application as neuroprotective agents.

2. Results

2.1. Chemical Characterization of the Three Different Polarity Fractions of P. lenticonus/Chios Mastiha

Previous studies on the chemical characterization of P. lenticonus/Chios mastiha revealed its enrichment in terpenoid compounds [14,37,38,39]. Although extensive studies have focused on terpenoid compound identification [14,37,38,39,40,41], the P. lenticonus/Chios mastiha composition involves unidentified compounds that remain to be characterized. To this effort, HPLC-QTOF-MS/MS analysis was performed in three different polarity P. lenticonus/Chios mastiha fractions (ap, mp, p). The analysis revealed a plethora of masses m/z (Table 1 and Table 2 and Supplementary data, Tables S1–S3, Figures S1 and S2). The identification of the suspect compounds was carried out according to Karadimou et al. [42].
Twenty-eight compounds were identified through suspect screening (Table 1) and are presented along with their literature-reported biological activities in Supplementary data, Table S1. Non-target identification (Supplementary data, Table S3) was performed using the SCIEX Natural Products Library with a Library Match Score above 50.0, according to Mitsikaris et al. [43]. All the tentative compounds were semi-quantified using the calibration curve of caffeic acid and were expressed as equivalents. Nine compounds were identified through non-target screening (Supplementary data, Table S2). Tentatively identified compounds involve fatty acids (Table 1), whose presence in P. lenticonus/Chios mastiha is reported for the first time. Specifically, the enrichment of fatty acids, such as palmitic acid, stearic, myristic, arachidonic, and pentadecanoic acid was observed in the ap fraction (Table 1). The exclusive enrichment of the coniferaldehyde compound in the ap was also tentatively identified. Moreover, to the best of our knowledge, the enrichment of mp and p fractions in compounds such as betulinic acid, euscaphic acid (triterpenoid with antioxidant activity), flavidin, luteolin glucoside, and fatty acids such as α-linolenic acid, ricinoleic acid, crepenynic acid, and gamma-linolenic acid is revealed for the first time (Table 1). In summary, except for oleanolic acid, octyl formate, 6,7-dihydroxylinalool, α-irone, methoxycinnamic, and masticadecanoic acid, which were previously identified as P. lenticonus/Chios mastiha components [1,21], the compounds in Table 1 are reported for the first time as P. lenticonus/Chios mastiha’s components. Furthermore, nine non-target new compounds were detected in different polarity fractions from P. lenticonus/Chios mastiha (Table 2). Interestingly, unequivocal molecular formulas were attributed to approximately 250 detected features (Supplementary Data, Table S3), highlighting P. lenticonus/Chios mastiha’s rich composition.

2.2. Different Polarity Fractions from P. lenticonus/Chios Mastiha Induced Resistance to H2O2-Triggered Reduction in Cell Viability of SH-SY5Y Cells

To assess the antioxidant activity and the potential neuroprotective effects of the different polarity fractions from P. lenticonus/Chios mastiha, their possible defensive actions on H2O2-induced reductions in cell viability of SH-SY5Y cells, by applying the MTT assay, were investigated. Firstly, we focused on setting up experimental conditions that lead to approximately 30% H2O2-induced reduction in the cell viability of SH-SY5Y cells. A moderate (20–30%) and not a severe reduction in cell viability by H2O2 could allow us to observe any possible protective effects achieved through pretreatment with potential antioxidant factors. Thus, the effects of 50 μΜ to 500 μΜ H2O2 treatment for 6 h, and 1 mM or 2 mM H2O2 treatment for 3 h on SH-SY5Y cell viability (Figure 1A) were examined. As shown in Figure 1A, the incubation of SH-SY5Y cells with 500 μΜ H2O2 for 6 h caused approximately 20% reduction in cell viability, whereas incubation of the cells with 1 mM H2O2 for 3 h caused more than 60% reduction in cell viability. Thus, incubation of the cells with 700 μΜ H2O2 for 6 h was considered an optimal condition to study the possible antioxidant activity of the different polarity fractions from P. lenticonus/Chios mastiha. Then, the MTT assay was applied in SH-SY5Y cells pretreated with different polarity fractions from P. lenticonus/Chios mastiha, at concentrations of 10 and 20 μg/mL for the ap fraction, 5 and 10 μg/mL for the mp fraction, and 10 and 20 μg/mL of p fraction for 24 h, and subsequently subjected to 700 μΜ H2O2 treatment for an additional 6 h. As shown in Figure 1B, after 24 h of incubation, 10 and 20 μg/mL of ap fraction, 5 and 10 μg/mL of mp fraction, and 10 and 20 μg/mL of p fraction did not cause any statistically significant reduction in cell viability of SH-SY5Y cells. Moreover, as was expected, incubation of SH-SY5Y cells with 700 μΜ H2O2 for 6 h caused an approximately 30% statistically significant reduction in cell viability compared to control cells. Interestingly, statistically significant resistance to H2O2-induced reduction in cell viability was observed in cells pre-incubated with 10 μg/mL or 20 μg/mL of ap fraction for 24 h and then subjected to 700 μΜ H2O2 treatment (for 6 h). Resistance to H2O2-induced reduction in cell viability was also observed, although to a lower extent, with pretreatment of the cells using 5 μg/mL of the mp fraction and 10 μg/mL of the p fraction.

2.3. Protective Effect of the Different Polarity P. lenticonus/Chios Mastiha Fractions on H2O2-Induced ROS Production in SH-SY5Y Cells

The potential antioxidant activity of the different polarity P. lenticonus/Chios mastiha fractions was estimated by measuring ROS production in H2O2-treated cells. ROS production was evaluated by assessing DCF staining in SH-SY5Y cells pretreated or not with the P. lenticonus/Chios mastiha’s fractions and subsequently subjected to H2O2-induced ROS production. Confocal microscopy images of the DCF dye, an indicator of ROS production, were taken in live SH-SY5Y cells. As shown in Figure 2, treatment of SH-SY5Y cells with 700 μΜ H2O2 for 6 h caused a statistically significant 2.5-fold increase in DCF staining compared to control cells. Interestingly, pretreatment of SH-SY5Y cells with the P. lenticonus/Chios mastiha’s fractions, for 24 h, at the indicated concentrations, provided them with resistance to H2O2-induced ROS production, leading to a statistically significant decrease in DCF staining compared to that in H2O2-treated cells. In accordance with the results from the MTT assay, the ap fraction showed the highest antioxidant activity.

2.4. Maintenance of the Mitochondrial Functionality by the Different Polarity P. lenticonus/Chios Mastiha Fractions upon Conditions of H2O2-Induced Oxidative Stress

The protective effect of the P. lenticonus/Chios mastiha’s fractions on H2O2-induced oxidative stress and reduction in cell viability of SH-SY5Y neuroblastoma cells leads us to evaluate the possible protective effect of the P. lenticonus/Chios mastiha’s fractions on mitochondrial functionality. Thus, mitochondrial membrane depolarization was assessed in SH-SY5Y cells pretreated or not with P. lenticonus/Chios mastiha fractions and subsequently subjected to H2O2 treatment, using the JC-1 dye. JC-1 monomer/aggregate (green/red) staining was assessed by applying confocal microscopy analysis. As shown in Figure 3, H2O2 treatment of the cells caused increased depolarization of the mitochondrial membrane as indicated by the increased ratio of the green to red staining compared to control vehicle-treated cells. Incubation of SH-SY5Y cells with the ap, mp, and p fractions of P. lenticonus/Chios mastiha, at concentrations of 10 μg/mL, 5 μg/mL, and 10 μg/mL, respectively, did not cause any mitochondrial depolarization, in accordance with results from the MTT assay. On the contrary, green/red JC1 staining was lower in the presence of ap and p fractions compared to control vehicle-treated cells, indicating the potential beneficial effect of the ap and p fractions on the preservation of the mitochondrial membrane potential. Interestingly, preincubation of the H2O2-treated SH-SY5Y cells with the different polarity fractions of P. lenticonus/Chios mastiha led to the maintenance of the mitochondrial membrane potential even in the presence of the oxidizing agent.

2.5. Regulation of GR, PPARα και Bcl-2 Protein Levels by the Apolar Fraction

Taking into consideration the fatty acid detection in P. lenticonus/Chios mastiha’s fractions and the crucial role of PPARα, a fatty acid-activating factor, in the regulation of many cellular functions, including lipid metabolism, antioxidant, and anti-inflammatory actions, the effect of P. lenticonus/Chios mastiha’s resin fractions on the modulation of PPARα protein levels was assessed (Figure 4). Western blot analysis of PPARα in extracts of SH-SY5Y cells pretreated with the ap, mp, and p P. lenticonus/Chios mastiha’s fractions and subsequently subjected to H2O2 treatment revealed a dose-dependent increase (1.3 to 2.6 folds increase) in PPARα protein levels. The highest activity was observed by the ap fraction. Considerable induction of PPARα levels was also observed with the 700 μΜ H2O2 treatment. A similar dose-dependent effect, by the ap and mp fractions, was also observed on GR protein levels, whose activity is proposed to be regulated by triterpenes and to constitute an activator of PPARα expression [44,45]. Evaluation of P. lenticonus/Chios mastiha fractions on the anti-apoptotic Bcl-2 molecule showed a moderate increase (20%) in the Bcl-2 protein levels by the ap fraction and approximately 20% decrease by the mp fraction.

3. Discussion

P. lenticonus/Chios mastiha is well-known for its enrichment in terpenoids [37,38,40,41]. In this study, we attempted to assess in depth the chemical composition of P. lenticonus/Chios mastiha concerning the presence of other chemical groups, such as fatty acids and unidentified compounds in three different polarity fractions (ap, mp, and p). Thus, HPLC-QTOF-MS/MS analysis was applied. Results from the analysis revealed 22 new components, mainly fatty acid compounds in P. lenticonus/Chios mastiha. Enrichment of the apolar and medium polar fraction with fatty acids such as palmitic, stearic, arachidonic, and pentadecanoic acids—compounds known to be involved in cellular signaling, affecting, among others, lipid metabolism and inflammatory responses [46,47,48,49,50]—was detected. To our knowledge, this is the first time that the enrichment of P. lenticonus/Chios mastiha in such fatty acids has been uncovered, shedding light on the further identification of the P. lenticonus/Chios mastiha chemical composition and the interpretation of the molecular mechanisms of many of the P. lenticonus/Chios mastiha’s biological actions, including anti-inflammatory activities and regulation of fatty acid metabolism. Moreover, the presence and enrichment of the p fraction with betulinic acid and euscaphic acid, terpenoids with known antioxidant and anti-inflammatory activities [51,52,53,54], as well as bioactive fatty acids such as linolenic acid, ricinoleic, and crepenynic acid, were revealed, unwrapping the presence of both saturated and non-saturated fatty acids in the p fraction of P. lenticonus/Chios mastiha. Furthermore, the presence of coniferaldehyde, an organic compound with many biological activities, such as anti-inflammatory, antioxidant, cytoprotective [55,56,57,58,59,60], as well as neuroprotective via Nrf2 activation [61], was detected in apolar fraction. Interestingly, a variety of unidentified compounds were also detected in P. lenticonus/Chios mastiha’s fractions, which remains to be identified. The plethora of identified and non-identified molecules in P. lenticonus/Chios mastiha unveiled its unique chemical composition, which is closely related to the plethora of its biological activities.
To further establish a link between P. lenticonus/Chios mastiha’s chemical composition and its biological activities, this study focuses on the investigation of the antioxidant activities of P. lenticonus/Chios mastiha; the antioxidant and neuroprotective activities of the three different polarity mastiha fractions—ap, mp, and p—were evaluated in SH-SY5Y cells. Emphasis was placed on the elucidation of the effect of P. lenticonus/Chios mastiha’s different polarity fractions on mitochondrial origin ROS production, mitochondrial functionality, the modulation of regulatory factors involved in these processes, and the association of the chemical composition of the fractions with their biological activities.
Evaluation of the potential cytoprotective actions of the P. lenticonus/Chios mastiha’s fractions on the H2O2-induced oxidative stress in SH-SY5Y cells revealed that the P. lenticonus/Chios mastiha’s ap fraction exerted the highest antioxidant activity, exhibiting statistically significant resistance to H2O2-induced reduction in cell viability of SH-SY5Y cells, which was accompanied by resistance to H2O2-induced increase in ROS production and in mitochondrial membrane depolarization. Enrichment of the apolar fraction with triterpenoids may be responsible for its antioxidant activity. The terpenoid-related antioxidant activities of P. lenticonus/Chios mastiha are supported by previous observations demonstrating that triterpenes of P. lenticonus/Chios mastiha are responsible for the decrease in LDL oxidation [13], reduction in several oxidative stress biomarkers [15], and enhancement of the antioxidant status in NAFLD obese patients [17]. Interestingly, results from the chemical analysis of this study uncovered enrichment of apolar fraction with fatty acids, such as arachidonic, palmitic, stearic acid, and pentadecanoic acid, which constitute signaling molecules with known antioxidant and/or neuroprotective activities [36,49,60,62,63,64,65,66,67]. Thus, the fatty acid components in the ap fraction of the P. lenticonus/Chios mastiha may also contribute to its antioxidant activity in SH-SY5Y neuroblastoma cells. In addition, enhanced antioxidant activities of the apolar fraction may be associated with its enrichment of the potential neuroprotective factor coniferaldehyde (55), which is first demonstrated as a P. lenticonus/Chios mastiha compound in this study, as well as with the presence of the octyl formate compound, demonstrated as a mitochondrial biogenesis factor [68].
Resistance to H2O2-induced decreases in SH-SY5Y cell viability, increases in ROS production, and mitochondrial membrane depolarization was also observed by the mp fraction, which was enriched in fatty acids, as shown in this study, and in triterpenes such as lupeol, oleanolic aldehyde, and triterpenoid methyl esters such as 24Z-masticadienonic acid methyl ester and 24Z-isomasticadienonic acid methyl ester, as we have previously shown [12]. The considerable antioxidant activities of the medium polar fraction may be attributed to fatty acids and to lupeol, which has been proposed as an antioxidant and anti-inflammatory agent with a potential impact on oxidative stress resistance in Alzheimer’s disease [34,69], as well as to its enrichment in the methoxycinnamic acid, as revealed by this study. Methoxycinnamic acid is proposed, among others, as a neuroprotective factor [70].
Structural similarities of triterpenoids with glucocorticoids [27] prompted us to evaluate whether the antioxidant activity of P. lenticonus/Chios mastiha’s fractions is related to the interference of their components with glucocorticoid signaling. We observed that the ap fraction caused an increase in GR protein levels, which was accompanied by an increase in PPARα that constitutes a GR target [44,45]. This observation shed light on the molecular mechanism of the potent antioxidant activity of ap and mp fractions of P. lenticonus/Chios mastiha since PPARα is considered an antioxidant factor, inducing among others catalase expression, and its expression is known to be triggered in the presence of antioxidant factors [71,72,73,74]. Apolar fraction-induced increase in PPARα protein level may be associated with the enrichment of the fraction with palmitic and stearic fatty acids, which are known as PPARα agonists and induce PPARα gene expression [46,75,76]. Arachidonic acid, an ap fraction identified compound, is known to interact with the anti-inflammatory effects of GR signaling [77] and may also contribute to the ap-induced anti-inflammatory actions [12].
Polar fraction of P. lenticonus/Chios mastiha also showed antioxidant activity, via reversal of the H2O2-induced increase in ROS production and mitochondrial membrane depolarization in SH-SY5Y cells. Since polar fraction is enriched in phenolic compounds, its antioxidant activity may be associated with these compounds, well-known for their antioxidant activity [78]. Antioxidant—the neuroprotective actions of polar fraction may also be associated with its enrichment in luteolin glucoside and gamma-linolenic acid compounds, as revealed in this study, compounds that have been reported to exert neuroprotection [79,80].

4. Conclusions

To conclude, in this study the characterization of 31 new components, mainly fatty acid compounds, such as palmitic, stearic, arachidonic, and pentadecanoic acids, and compounds such as octyl formate and coniferaldehyde in three different polarity fractions of P. lenticonus/Chios mastiha was achieved. Moreover, the presence of 250 non-target compounds was uncovered, highlighting the great abundance of bioactive molecules in P. lenticonus/Chios mastiha and strengthening the potential of its pharmaceutical exploitation. Most importantly, the potent neuroprotective activity of the P. lenticonus/Chios mastiha, accomplished by the preservation of the mitochondrial functionality and regulation of antioxidant and anti-inflammatory factors expression, upon oxidative stress induction in neuroblastoma cells, was documented. Precisely, the antioxidant activity of the three different polarity mastiha fractions, ap, mp, and polar, was revealed in human neuroblastoma SH-SY5Y cells. Apolar fraction enriched in antioxidant factors such as triterpenes, fatty acids, octyl formate, and coniferaldehyde exhibited the highest antioxidant–neuroprotective activities, as documented by the enhanced suppression of the H2O2-induced reduction in SH-SY5Y cell viability, the induction of the antioxidant and anti-inflammatory factors PPARα and GR expression, the reduction in ROS production, and the resistance to the depolarization of the mitochondrial membrane potential, in H2O2-treated neuroblastoma SH-SY5Y cells. Antioxidant activity, although to a lower extent, was also observed by the mp and p fractions, which can be attributed to their enrichment in terpenoids and phenolic compounds, respectively. Moreover, the identification of neuroprotective factors such as methoxycinnamic and leteolin glucoside and gamma-linolenic acid compounds, as components of the mp and p fraction, respectively, was achieved in this study. Their presence may be correlated with the observed antioxidant activity of the mp and p fractions. Taking into account the seriousness of neurodegenerative diseases, the global trend in increasing the number of patients with this health impairment, alongside the world trend in using natural products to improve health or to face mental illness and neurodegeneration [81], the results from the study uncover and highlight the potential application of P. lenticonus/Chios mastiha as a neuroprotective agent in neurodegenerative diseases.

5. Materials and Methods

5.1. Chemicals

Dulbecco’s Modified Eagle Medium (DMEM), Trypsin, Fetal Bovine Serum (FBS), MitoTracker Red CMX Ros (CMX), the 6-carboxy-2′,7′-dichlorodihydrofluorescein diacetate (6-carboxy H2DCFDA), and the (5,5′,6,6′-tetrachloro-1,1′,3,3′-tetraethylbenzimidazolylcarbocyanine iodide) JC-1, were obtained from Thermo Fisher Scientific (GmbH, Basel, Switzerland). The Molecular protein weight marker was purchased from ProteinTech (Rosemont, IL, USA). Cocktail protease inhibitors were purchased from Roche (Mannheim, Germany). Silica gel 60 (70–230 mesh) and Celite® 545 particle size 0.02–0.1 mm, pH 10 (100 g/L, H2O, 20 °C), used for vacuum filtration, were purchased from Macherey–Nagel (Düren, Germany). Solvents were from Sigma-Aldrich (Darmstadt, Germany) and were used without any further purification unless stated otherwise. All other chemicals, including H2O2, were purchased from Sigma-Aldrich (Darmstadt, Germany).

5.2. Chios Mastiha Fractionation and Chemical Characterization

P. lenticonus/Chios mastiha was offered by the “Chios Mastiha Growers Association” (https://www.gummastic.gr/en/, accessed on 18 February 2025), which is the certified organization for the production, identification, and commercial exploitation of the P. lenticonus/Chios mastiha. P. lenticonus/Chios mastiha’s three different polarity fractions were obtained as previously described [12,14]. Details on the methodology applied are presented in the Supplementary Materials (Supplementary Data S1). The methodology applied for the chemical characterization of the three different polarity fractions of P. lenticonus/Chios mastiha is followed. Chromatographic analysis was performed on an ExionAC LC system (SCIEX, Framingham, MA, USA) equipped with a controller, two pumps, a degasser, and an auto-sampler. The X500R Q-TOF mass spectrometer (SCIEX, Framingham, MA, USA) was equipped with an electrospray ionization (ESI) turboVTM source and operated in negative ionization mode. TOF-MS and TOF-MS/MS data were acquired using a data-dependent acquisition ESI mode. For the separation of the analytes, a Fortis C18 column (100 mm length, 2.1 mm i.d, 2.6 μm particle size) purchased from Fortis (Cheshire, United Kingdom) was used, thermostated at 40 °C. The mobile phase consisted of: (A) 0.1% (v/v) formic acid in water and (B) 0.1% (v/v) formic acid in methanol. The flow rate was initially set at 0.2 mL min−1, increased to 0.4 mL min−1 at 15 min, and decreased to 0.2 mL min−1 at 16 min. The elution program was as follows: 0–4 min, 99–61%A; 4–12 min, 61–5% A; 12–15 min, 5% A; 15–16 min, 5–99% A; 16–20 min, 99% A. The QTOF-MS system was equipped with an ESI interface, operating in negative mode with the following settings: spray voltage, 4500 V; heater gas temperature, 55 °C; declustering potential, 80 V. The MS/MS spectra were obtained at a collision energy of 45 eV and a collision energy spread of 15 eV. External calibration was performed before analysis with a cluster solution provided by SCIEX. Additionally, the calibration solution was injected at the beginning of each run for internal calibration and once every five samples during batch acquisition. Mass spectra were recorded in the m/z range from 50 to 1000 at an accumulation time of 0.25 s. MS/MS experiments were conducted in data-dependent acquisition mode at an accumulation time of 0.08 s for the 10 most abundant precursor ions per full scan. Sample acquisition was monitored by SCIEX OS software Version 2.0.1., released in 2019 (© 2019 AB Sciex). Extraction ion chromatograms were generated using SCIEX OS software Version 2.0.1. The non-target screening was carried out using Analytics SCIEX OS software Version 2.0.1. and the SCIEX Natural Products Library.

5.3. Cell Culture

SH-SY5Y cells were used in the study. SH-SY5Y is a human neuroblastoma cell line widely used as an in vitro model for a variety of neurobiological analyses [82]. SH-SY5Y cells were obtained from the American Type Culture Collection (ATCC) and were cultured in DMEM 1g/L glucose, with phenol red, 10% v/v FBS, 2 mM L-glutamine, and 100 units/mL penicillin/streptomycin at 37 °C and 5% CO2. For live cell fluorescence microscopy analysis, cells were maintained in low glucose (1g/L), without phenol red DMEM, supplemented with 2 mM L-glutamine and 100 units/mL penicillin/streptomycin.

5.4. Antibodies

For Western blot analysis, specific antibodies against Bcl-2, PPARα, GR, and β-actin were used. Monoclonal antibodies against GR, PPARα, and β-actin were obtained from Santa Cruz Biotechnology (Europe Inc., Heidelberg, Germany) and ProteinTech (Rosemont, IL, USA), respectively. Polyclonal antibodies against Bcl-2 were purchased from Cell Signaling Technology (Leiden, The Netherlands).

5.5. Cell Viability Assay-MTT

To investigate the effects of different polarity fractions—ap, mp, and p (based on the gradually increased polarity of the solvents used in the fractionation: petroleum ether solvent for ap fraction, ethyl acetate solvent for mp fraction, and tetrahydrofuran solvent for p fraction)—from P. lenticonus/Chios mastiha on the cell viability of SH-SY5Y cells, either in the presence or absence of H2O2, an MTT assay was applied as previously described [83]. Briefly, 8 × 103 cells were plated in a 96-well plate and cultured in low glucose (1 g/L) DMEM, supplemented with 10% v/v FBS, 2 mM L-glutamine, and 100 units/mL penicillin/streptomycin for 24 h. Then, cells were treated with 10 and 20 μg/mL of ap and p P. lenticonus/Chios mastiha’s fractions and with 5 and 10 μg/mL of mp fraction (diluted in DMSO) for 24 h. Incubation with 700 μΜ H2O2 for 6 h followed. Control, vehicle-treated cells were incubated with 1/1000 v/v DMSO for 24 h as well. Then, MTT reagent was added at a final concentration of 0.5 μg/mL for 3 h at 37 °C. Finally, the produced formazan crystals were dissolved in 100% isopropanol, upon shaking and the absorbance was measured at 570 nm and 690 nm, as a reference, in a multimode plate reader (EnSpire, PerkinElmer, Beaconsfield, UK). Relative cell viability was expressed as the viability of the cells treated with the indicated concentrations of the respective different polarity P. lenticonus/Chios mastiha fractions (ap, mp, and p) and/or H2O2, compared to the cell viability of the vehicle-untreated (control) cells. The viability of control cells was set to 1.

5.6. Electrophoresis and Western Blot Analysis

For Western blot analysis, 25 × 104 SH-SY5Y cells were grown in 6-well plates in low glucose (1g/L glucose) DMEM supplemented with 10% v/v FBS, 2 mM L-glutamine, and 100 units/mL penicillin/streptomycin for 48 h. Then, the cells were incubated with 20 μg/mL and 10 μg/mL of ap and p fractions of P. lenticonus/Chios mastiha, and with 10 μg/mL and 5 μg/mL of the medium polar fraction, for 24 h. Subsequently, SH-SY5Y cells were further incubated with 700 μΜ H2O2 for 6 h, or 1/1000 v/v DMSO (control vehicle-treated cells). Then, cells were washed with PBS (4 °C) and lysed in lysis buffer (20 mM Tris-HCl pH: 7.5, 250 mM NaCl, 0.5 % v/v Triton-X, 3 mM EDTA, supplemented also with DTT, PMSF, and cocktail of protease inhibitors). Bradford assay was applied for protein determination [12]. Then, cell extracts were electrophoresed in discontinuous SDS-PAGE and Western blotting with specific antibodies against Bcl-2, GR, PPARα, and β-actin, as previously described [84]. Enhanced chemiluminescence was used for the detection of the protein bands. Quantification of protein band density was performed by the ImageJ program (v1.52). β-actin band intensity was used for the normalization of the results. Relative protein levels were expressed as band intensity normalized against the respective band’s intensity of β-actin. Relative protein levels in control vehicle-treated cells were set to 1 [29].

5.7. Intracellular ROS Measurement

To evaluate the antioxidant activity of different polarity fractions from P. lenticonus/Chios mastiha, total ROS production measurements were conducted in SH-SY5Y cells pretreated with the ap, mp, and p fractions and then subjected to H2O2-induced oxidative stress. The 6-carboxy-2′,7′-dichlorodihydrofluorescein diacetate, (6-carboxy H2DCFDA) dye (Thermo Fisher Scientific) was used for ROS detection. The cell-permeant H2DCFDA is a non-fluorescent indicator for reactive oxygen species (ROS) in cells. Upon cleavage of the acetate groups by intracellular esterases and oxidation, the nonfluorescent H2DCFDA is converted to the highly fluorescent 2′,7′-dichlorofluorescein (DCF). Thus, for ROS production measurement, 1 × 105 SH-SY5Y cells were plated on 60 × 15 mm culture dishes and cultured in low glucose (1 g/L) DMEM, supplemented with 10% v/v FBS, 2 mM L-glutamine, and 100 units/mL penicillin/streptomycin for 48 h. Then, cells were incubated with 10 μg/mL of ap, and p fractions or with 5 μg/mL of mp fraction for 24 h. Vehicle (1/1000 v/v DMSO)-treated cells were used as control. Subsequently, cells were subjected to H2O2 (700 μΜ) or vehicle (1/1000 H2O) treatment for 6 h. Then, the cell culture medium was replaced with low glucose (1 g/L glucose), without phenol red DMEM, supplemented with 2 mM L-glutamine and 100 units/mL penicillin/streptomycin, containing 10 μΜ H2DCFDA dye, 200 nM CMX and 1 μg/mL Hoecsht-33342 at 37 °C for 30 min. Finally, cells were washed in DMEM without phenol red and subjected to live cell imaging using confocal microscopy analysis. Fluorescence excitation of the oxidized DCF-DA dye was at 488 nm, and fluorescence emission was measured at 530–550 nm using a Zeiss LSM 800 confocal microscope [85].

5.8. Analysis of the Mitochondrial Membrane Potential

To assess the ability of P. lenticonus/Chios mastiha’s fractions to reverse the H2O2-induced mitochondrial membrane depolarization of SH-SY5Y cells, mitochondrial membrane potential changes were measured using (5,5′,6,6′-tetrachloro-1,1′,3,3′-tetraethylbenzimidazolylcarbocyanine iodide) JC-1 dye. JC-1 is a lipophilic cationic dye that can enter mitochondria, where it accumulates and forms reversible red fluorescent J-aggregates. In apoptotic cells, the JC-1 dye also enters the mitochondria but to a lesser degree since the inside of the mitochondria is less negative. Under such circumstances, JC-1 does not reach the appropriate concentration to trigger the formation of J aggregates, thus retaining it as a green, fluorescent dye. Based on that, an assessment of the green/red fluorescence ratio depicts the state of the mitochondrial membrane polarization [86]. For the JC-1 assay, 1 × 105 SH-SY5Y cells were plated on 60 × 15 mm culture dishes and cultured in low glucose DMEM (1 g/L glucose), supplemented with 10% v/v FBS, 2 mM L-glutamine, and 100 units/mL penicillin/streptomycin for 48 h. Then, cells were treated with 10 μg/mL of ap and p fractions, or with 5 μg/mL of mp fraction for 24 h or with 1/1000 v/v DMSO (control cells), and subsequently cultured in the presence or absence of 700 μΜ H2O2 for 6 h. Finally, cells were incubated with 1 μg/mL Hoechst-33342 and 2 μΜ JC-1, without phenol red-DMEM culture medium, at 37 °C for 30 min. Then, DMEM was replaced with a fresh medium, and cells were subjected to confocal microscopy live imaging analysis. Green JC-1 fluorescence was measured at an excitation of 488 nm, and the emission was measured at 494–535 nm. The red fluorescent J-aggregates were measured at an excitation of 560 nm, and the emission wavelength was at 560–590 nm, using a Zeiss LSM 800 confocal microscope [85].

5.9. Statistical Analysis

Results are expressed as mean ± SD. Data were analyzed by one-way analysis of variance (one-way ANOVA) or two-way ANOVA followed by Tukeys’s post hoc test using StatPlus LE Software, version 7.0 (AnalystSoft, Brandon, FL, USA). Differences were considered significant at a two-tailed p value < 0.05.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/molecules30050997/s1. Supplementary Data S1: Protocol of P. lenticonus/Chios mastiha fractionation [87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183,184,185,186,187,188,189,190,191,192,193,194,195,196,197,198,199,200,201,202,203,204,205,206,207,208,209,210,211,212,213]. Table S1: Suspect compounds identified in different polarity fractions from P. lenticonus/Chios mastiha by HPLC-QTOF-MS/MS analysis, their chemical classification, and biological activities. Table S2: Non-target screening results, identified by HPLC-QTOF-MS/MS analysis, in different polarity fractions from mastiha, their chemical classification, and biological activities. Table S3: Unequivocal molecular formulas and hit scores of non-target compounds identified in apolar, medium polar, and polar fractions from P. lenticonus/Chios mastiha assigned by SCIEX OS software Version 2.0.1. and the SCIEX Natural Products Library. Figure S1: Extracted Ion Chromatograms (EICs), MS, and MS/MS spectra of suspect compounds; Figure S2: Extracted Ion Chromatograms (EICs), MS, and MS/MS spectra of non-target compounds. References [87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183,184,185,186,187,188,189,190,191,192,193,194,195,196,197,198,199,200,201,202,203,204,205,206,207,208,209,210,211,212,213] are cited in the Supplementary Materials.

Author Contributions

Conceptualization, A.-M.G.P.; Methodology, A.-M.G.P., A.G., N.P.K., and F.P.; Validation A.G., I.T., and F.D.K.; Investigation, A.G., F.D.K., F.P., and N.P.K.; Formal Analysis, A.G., F.D.K., I.T., A.-M.G.P., and N.P.K.; Resources, A.-M.G.P. and F.P.; Data Curation, A.-M.G.P., F.P. and N.P.K.; Writing—Original Draft Preparation, A.-M.G.P. and A.G.; Writing—Review and Editing, A.-M.G.P., A.G., and N.P.K.; Visualization, A.G.; Supervision, A.-M.G.P.; Project Administration, A.-M.G.P.; Funding Acquisition, A.-M.G.P., and F.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research was partially supported by the “Chios Gum Mastic Growers Association”, the “mastihashop” and by the Postgraduate Programs “Application of Molecular Biology-Genetics-Diagnostic Biomarkers” and “Biotechnology-Quality assessment in Nutrition and the Environment”, Department of Biochemistry and Biotechnology, University of Thessaly (to A.-M.G.P.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data, tables, and figures are original. Details on data analysis are available from the corresponding author upon reasonable request.

Acknowledgments

The authors would like to thank the Interdisciplinary Agri-Food Center, Aristotle University of Thessaloniki (KEAGRO-AUTH) for providing access to the equipment of the unit.

Conflicts of Interest

The authors declare no conflicts of interest. The founders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, and in the decision to publish the result.

References

  1. Pachi, V.K.; Mikropoulou, E.V.; Gkiouvetidis, P.; Siafakas, K.; Argyropoulou, A.; Angelis, A.; Mitakou, S.; Halabalaki, M. Traditional uses, phytochemistry and pharmacology of Chios mastic gum (Pistacia lentiscus var. Chia, Anacardiaceae): A review. J. Ethnopharmacol. 2020, 254, 112485. [Google Scholar] [CrossRef] [PubMed]
  2. Soulaidopoulos, S.; Tsiogka, A.; Chrysohoou, C.; Lazarou, E.; Aznaouridis, K.; Doundoulakis, I.; Tyrovola, D.; Tousoulis, D.; Tsioufis, K.; Vlachopoulos, C.; et al. Overview of Chios Mastic Gum (Pistacia lentiscus) Effects on Human Health. Nutrients 2022, 14, 590. [Google Scholar] [CrossRef]
  3. Balan, K.V.; Demetzos, C.; Prince, J.; Dimas, K.; Cladaras, M.; Han, Z.; Wyche, J.H.; Pantazis, P. Induction of apoptosis in human colon cancer HCT116 cells treated with an extract of the plant product, Chios mastic gum. In Vivo 2005, 19, 93–102. [Google Scholar] [PubMed]
  4. Dimas, K.S.; Pantazis, P.; Ramanujam, R. Review: Chios mastic gum: A plant-produced resin exhibiting numerous diverse pharmaceutical and biomedical properties. In Vivo 2012, 26, 777–785. [Google Scholar]
  5. He, M.L.; Yuan, H.Q.; Jiang, A.L.; Gong, A.Y.; Chen, W.W.; Zhang, P.J.; Young, C.Y.; Zhang, J.Y. Gum mastic inhibits the expression and function of the androgen receptor in prostate cancer cells. Cancer 2006, 106, 2547–2555. [Google Scholar] [CrossRef] [PubMed]
  6. Dedoussis, G.V.; Kaliora, A.C.; Psarras, S.; Chiou, A.; Mylona, A.; Papadopoulos, N.G.; Andrikopoulos, N.K. Antiatherogenic effect of Pistacia lentiscus via GSH restoration and downregulation of CD36 mRNA expression. Atherosclerosis 2004, 174, 293–303. [Google Scholar] [CrossRef]
  7. Triantafyllou, A.; Chaviaras, N.; Sergentanis, T.N.; Protopapa, E.; Tsaknis, J. Chios mastic gum modulates serum biochemical parameters in a human population. J. Ethnopharmacol. 2007, 111, 43–49. [Google Scholar] [CrossRef]
  8. Paraschos, S.; Mitakou, S.; Skaltsounis, A.L. Chios gum mastic: A review of its biological activities. Curr. Med. Chem. 2012, 19, 2292–2302. [Google Scholar] [CrossRef]
  9. Zhou, L.; Satoh, K.; Takahashi, K.; Watanabe, S.; Nakamura, W.; Maki, J.; Hatano, H.; Takekawa, F.; Shimada, C.; Sakagami, H. Re-evaluation of anti-inflammatory activity of mastic using activated macrophages. In Vivo 2009, 23, 583–589. [Google Scholar]
  10. Loizou, S.; Paraschos, S.; Mitakou, S.; Chrousos, G.P.; Lekakis, I.; Moutsatsou, P. Chios mastic gum extract and isolated phytosterol tirucallol exhibit anti-inflammatory activity in human aortic endothelial cells. Exp. Biol. Med. 2009, 234, 553–561. [Google Scholar] [CrossRef]
  11. Papada, E.; Forbes, A.; Amerikanou, C.; Torovic, L.; Kalogeropoulos, N.; Tzavara, C.; Triantafillidis, J.K.; Kaliora, A.C. Antioxidative Efficacy of a Pistacia Lentiscus Supplement and Its Effect on the Plasma Amino Acid Profile in Inflammatory Bowel Disease: A Randomised, Double-Blind, Placebo-Controlled Trial. Nutrients 2018, 10, 1779. [Google Scholar] [CrossRef]
  12. Kalousi, F.D.; Pollastro, F.; Karra, A.G.; Tsialtas, I.; Georgantopoulos, A.; Salamone, S.; Psarra, A.G. Regulation of Energy Metabolism and Anti-Inflammatory Activities of Mastiha Fractions from Pistacia lentiscus L. var. chia. Foods 2023, 12, 1390. [Google Scholar] [CrossRef] [PubMed]
  13. Andrikopoulos, N.K.; Kaliora, A.C.; Assimopoulou, A.N.; Papapeorgiou, V.P. Biological activity of some naturally occurring resins, gums and pigments against in vitro LDL oxidation. Phytother. Res. 2003, 17, 501–507. [Google Scholar] [CrossRef]
  14. Kalousi, F.D.; Pollastro, F.; Christodoulou, E.C.; Karra, A.G.; Tsialtas, I.; Georgantopoulos, A.; Salamone, S.; Psarra, A.G. Apoptotic, Anti-Inflammatory Activities and Interference with the Glucocorticoid Receptor Signaling of Fractions from Pistacia lentiscus L. var. chia Leaves. Plants 2022, 11, 934. [Google Scholar] [CrossRef] [PubMed]
  15. Papada, E.; Gioxari, A.; Amerikanou, C.; Forbes, A.; Tzavara, C.; Smyrnioudis, I.; Kaliora, A.C. Regulation of faecal biomarkers in inflammatory bowel disease patients treated with oral mastiha (Pistacia lentiscus) supplement: A double-blind and placebo-controlled randomised trial. Phytother. Res. 2019, 33, 360–369. [Google Scholar] [CrossRef]
  16. Kaliora, A.C.; Stathopoulou, M.G.; Triantafillidis, J.K.; Dedoussis, G.V.; Andrikopoulos, N.K. Chios mastic treatment of patients with active Crohn’s disease. World J. Gastroenterol. 2007, 13, 748–753. [Google Scholar] [CrossRef] [PubMed]
  17. Kanoni, S.; Kumar, S.; Amerikanou, C.; Kurth, M.J.; Stathopoulou, M.G.; Bourgeois, S.; Masson, C.; Kannt, A.; Cesarini, L.; Kontoe, M.S.; et al. Nutrigenetic Interactions Might Modulate the Antioxidant and Anti-Inflammatory Status in Mastiha-Supplemented Patients with NAFLD. Front. Immunol. 2021, 12, 683028. [Google Scholar] [CrossRef]
  18. Checa, J.; Aran, J.M. Reactive Oxygen Species: Drivers of Physiological and Pathological Processes. J. Inflamm. Res. 2020, 13, 1057–1073. [Google Scholar] [CrossRef] [PubMed]
  19. Piccolella, S.; Nocera, P.; Carillo, P.; Woodrow, P.; Greco, V.; Manti, L.; Fiorentino, A.; Pacifico, S. An apolar Pistacia lentiscus L. leaf extract: GC-MS metabolic profiling and evaluation of cytotoxicity and apoptosis inducing effects on SH-SY5Y and SK-N-BE(2)C cell lines. Food Chem. Toxicol. 2016, 95, 64–74. [Google Scholar] [CrossRef] [PubMed]
  20. Galluzzi, L.; Kepp, O.; Trojel-Hansen, C.; Kroemer, G. Mitochondrial control of cellular life, stress, and death. Circ. Res. 2012, 111, 1198–1207. [Google Scholar] [CrossRef]
  21. Assimopoulou, A.N.; Papageorgiou, V.P. GC-MS analysis of penta- and tetra-cyclic triterpenes from resins of Pistacia species. Part I. Pistacia lentiscus var. Chia. Biomed. Chromatogr. 2005, 19, 285–311. [Google Scholar] [CrossRef] [PubMed]
  22. Paraschos, S.; Magiatis, P.; Mitakou, S.; Petraki, K.; Kalliaropoulos, A.; Maragkoudakis, P.; Mentis, A.; Sgouras, D.; Skaltsounis, A.L. In vitro and in vivo activities of Chios mastic gum extracts and constituents against Helicobacter pylori. Antimicrob. Agents Chemother. 2007, 51, 551–559. [Google Scholar] [CrossRef] [PubMed]
  23. Ottria, R.; Xynomilakis, O.; Casati, S.; Abbiati, E.; Maconi, G.; Ciuffreda, P. Chios Mastic Gum: Chemical Profile and Pharmacological Properties in Inflammatory Bowel Disease: From the Past to the Future. Int. J. Mol. Sci. 2023, 24, 12038. [Google Scholar] [CrossRef]
  24. Vallianou, I.; Peroulis, N.; Pantazis, P.; Hadzopoulou-Cladaras, M. Camphene, a plant-derived monoterpene, reduces plasma cholesterol and triglycerides in hyperlipidemic rats independently of HMG-CoA reductase activity. PLoS ONE 2011, 6, e20516. [Google Scholar] [CrossRef] [PubMed]
  25. Magiatis, P.; Melliou, E.; Skaltsounis, A.L.; Chinou, I.B.; Mitaku, S. Chemical composition and antimicrobial activity of the essential oils of Pistacia lentiscus var. chia. Planta Med. 1999, 65, 749–752. [Google Scholar] [CrossRef]
  26. Timmermans, S.; Souffriau, J.; Libert, C. A General Introduction to Glucocorticoid Biology. Front. Immunol. 2019, 10, 1545. [Google Scholar] [CrossRef]
  27. Georgatza, D.; Gorgogietas, V.A.; Kylindri, P.; Charalambous, M.C.; Papadopoulou, K.K.; Hayes, J.M.; Psarra, A.G. The triterpene echinocystic acid and its 3-O-glucoside derivative are revealed as potent and selective glucocorticoid receptor agonists. Int. J. Biochem. Cell Biol. 2016, 79, 277–287. [Google Scholar] [CrossRef]
  28. Karra, A.G.; Konstantinou, M.; Tzortziou, M.; Tsialtas, I.; Kalousi, F.D.; Garagounis, C.; Hayes, J.M.; Psarra, A.G. Potential Dissociative Glucocorticoid Receptor Activity for Protopanaxadiol and Protopanaxatriol. Int. J. Mol. Sci. 2018, 20, 94. [Google Scholar] [CrossRef]
  29. Georgantopoulos, A.; Vougioukas, A.; Kalousi, F.D.; Tsialtas, I.; Psarra, A.G. Comparative Studies on the Anti-Inflammatory and Apoptotic Activities of Four Greek Essential Oils: Involvement in the Regulation of NF-kappaBeta and Steroid Receptor Signaling. Life 2023, 13, 1534. [Google Scholar] [CrossRef]
  30. Boutemine, I.M.; Amri, M.; Dorgham, K.; Amir, Z.C.; Benazzouz, S.; Ameur, F.; Layaida, K.; Yssel, H.; Touil-Boukoffa, C. Beneficial role of Pistacia lentiscus aqueous extract in experimental colitis: Anti-inflammatory and potential therapeutic effects. Inflammopharmacology 2021, 29, 1225–1239. [Google Scholar] [CrossRef]
  31. Cui, H.; Li, X.; An, X.R.; Liu, W.; Yuan, T. Masticadienonic acid from Chios mastic gum mitigates colitis in mice via modulating inflammatory response, gut barrier integrity and microbiota. Phytomedicine 2023, 108, 154518. [Google Scholar] [CrossRef]
  32. Lee, W.; Yang, E.J.; Ku, S.K.; Song, K.S.; Bae, J.S. Anti-inflammatory effects of oleanolic acid on LPS-induced inflammation in vitro and in vivo. Inflammation 2013, 36, 94–102. [Google Scholar] [CrossRef]
  33. Dharmappa, K.K.; Kumar, R.V.; Nataraju, A.; Mohamed, R.; Shivaprasad, H.V.; Vishwanath, B.S. Anti-inflammatory activity of oleanolic acid by inhibition of secretory phospholipase A2. Planta Med. 2009, 75, 211–215. [Google Scholar] [CrossRef]
  34. Park, J.S.; Rehman, I.U.; Choe, K.; Ahmad, R.; Lee, H.J.; Kim, M.O. A Triterpenoid Lupeol as an Antioxidant and Anti-Neuroinflammatory Agent: Impacts on Oxidative Stress in Alzheimer’s Disease. Nutrients 2023, 15, 3059. [Google Scholar] [CrossRef] [PubMed]
  35. Dai, D.; Zeldin, D.C.; Blaisdell, J.A.; Chanas, B.; Coulter, S.J.; Ghanayem, B.I.; Goldstein, J.A. Polymorphisms in human CYP2C8 decrease metabolism of the anticancer drug paclitaxel and arachidonic acid. Pharmacogenetics 2001, 11, 597–607. [Google Scholar] [CrossRef] [PubMed]
  36. Xu, B.; Bai, L.; Chen, L.; Tong, R.; Feng, Y.; Shi, J. Terpenoid natural products exert neuroprotection via the PI3K/Akt pathway. Front. Pharmacol. 2022, 13, 1036506. [Google Scholar] [CrossRef] [PubMed]
  37. Stamou, P.; Gianniou, D.D.; Trougakos, I.P.; Mitakou, S.; Halabalaki, M.; Kostakis, I.K.; Skaltsounis, A.L. Anti-Inflammatory Activity of the Major Triterpenic Acids of Chios Mastic Gum and Their Semi-Synthetic Analogues. Biomolecules 2024, 14, 1618. [Google Scholar] [CrossRef]
  38. Said, S.A.; Fernandez, C.; Greff, S.; Torre, F.; Derridj, A.; Gauquelin, T.; Mevy, J.P. Inter-population variability of terpenoid composition in leaves of Pistacia lentiscus L. from Algeria: A chemoecological approach. Molecules 2011, 16, 2646–2657. [Google Scholar] [CrossRef]
  39. Floris, S.; Di Petrillo, A.; Pintus, F.; Delogu, G.L. Pistacia lentiscus: Phytochemistry and Antidiabetic Properties. Nutrients 2024, 16, 1638. [Google Scholar] [CrossRef] [PubMed]
  40. Yu, Y.H.; Feng, Y.P.; Liu, W.; Yuan, T. Diverse Triterpenoids from Mastic Produced by Pistacia lentiscus and Their Anti-Inflammatory Activities. Chem. Biodivers 2022, 19, e202101012. [Google Scholar] [CrossRef] [PubMed]
  41. Kahaer, G.; Abdulla, R.; Wu, T.; Aisa, H.A. Systematic qualitative analysis of terpenes in mastic (Pistacia lentiscus L.) extract and their fragmentations by UHPLC-Q-Orbitrap-HRMS. Phytochem. Anal. 2024, 35, 1072–1087. [Google Scholar] [CrossRef]
  42. Karadimou, C.; Petsa, E.; Ouroumi, N.A.; Papadakis, E.N.; Kontoudakis, N.; Theocharis, S.; Mourtzinos, I.; Menkissoglu-Spiroudi, U.; Kalogiouri, N.P.; Koundouras, S. Exploration of the anthocyanin and proanthocyanidin profile of Greek red grape skins belonging to Vradiano, Limnio, and Kotsifali cultivars, analyzed by a novel LC-QTOF-MS/MS method. Phytochem. Anal. 2024, 35, 1781–1793. [Google Scholar] [CrossRef] [PubMed]
  43. Mitsikaris, P.D.; Kostas, S.; Mourtzinos, I.; Menkissoglu-Spiroudi, U.; Papadopoulos, A.; Kalogiouri, N.P. Investigation of Rosa species by an optimized LC-QTOF-MS/MS method using targeted and non-targeted screening strategies combined with multivariate chemometrics. Phytochem. Anal. 2024, 35, 1100–1111. [Google Scholar] [CrossRef] [PubMed]
  44. Lemberger, T.; Staels, B.; Saladin, R.; Desvergne, B.; Auwerx, J.; Wahli, W. Regulation of the peroxisome proliferator-activated receptor alpha gene by glucocorticoids. J. Biol. Chem. 1994, 269, 24527–24530. [Google Scholar] [CrossRef]
  45. Wang, L.; Cai, Y.; Jian, L.; Cheung, C.W.; Zhang, L.; Xia, Z. Impact of peroxisome proliferator-activated receptor-alpha on diabetic cardiomyopathy. Cardiovasc. Diabetol. 2021, 20, 2. [Google Scholar] [CrossRef] [PubMed]
  46. Kamata, S.; Oyama, T.; Saito, K.; Honda, A.; Yamamoto, Y.; Suda, K.; Ishikawa, R.; Itoh, T.; Watanabe, Y.; Shibata, T.; et al. PPARalpha Ligand-Binding Domain Structures with Endogenous Fatty Acids and Fibrates. iScience 2020, 23, 101727. [Google Scholar] [CrossRef]
  47. Hostetler, H.A.; Petrescu, A.D.; Kier, A.B.; Schroeder, F. Peroxisome proliferator-activated receptor alpha interacts with high affinity and is conformationally responsive to endogenous ligands. J. Biol. Chem. 2005, 280, 18667–18682. [Google Scholar] [CrossRef] [PubMed]
  48. Varga, T.; Czimmerer, Z.; Nagy, L. PPARs are a unique set of fatty acid regulated transcription factors controlling both lipid metabolism and inflammation. Biochim. Biophys. Acta 2011, 1812, 1007–1022. [Google Scholar] [CrossRef] [PubMed]
  49. Iversen, L.; Kragballe, K. Arachidonic acid metabolism in skin health and disease. Prostaglandins Other Lipid Mediat. 2000, 63, 25–42. [Google Scholar] [CrossRef]
  50. Venn-Watson, S.; Schork, N.J. Pentadecanoic Acid (C15:0), an Essential Fatty Acid, Shares Clinically Relevant Cell-Based Activities with Leading Longevity-Enhancing Compounds. Nutrients 2023, 15, 4607. [Google Scholar] [CrossRef] [PubMed]
  51. Fulda, S. Betulinic acid: A natural product with anticancer activity. Mol. Nutr. Food Res. 2009, 53, 140–146. [Google Scholar] [CrossRef] [PubMed]
  52. Costa, J.F.; Barbosa-Filho, J.M.; Maia, G.L.; Guimaraes, E.T.; Meira, C.S.; Ribeiro-dos-Santos, R.; de Carvalho, L.C.; Soares, M.B. Potent anti-inflammatory activity of betulinic acid treatment in a model of lethal endotoxemia. Int. Immunopharmacol. 2014, 23, 469–474. [Google Scholar] [CrossRef] [PubMed]
  53. Dai, W.; Dong, P.; Liu, J.; Gao, Y.; Hu, Y.; Lin, H.; Song, Y.; Mei, Q. Euscaphic acid inhibits proliferation and promotes apoptosis of nasopharyngeal carcinoma cells by silencing the PI3K/AKT/mTOR signaling pathway. Am. J. Transl. Res. 2019, 11, 2090–2098. [Google Scholar] [PubMed]
  54. Shi, C.; Li, Z.; Wu, Y.; Li, X.; Li, Y.; Wei, J.; Li, J.; Zhang, Y.; Li, L. Euscaphic acid and Tormentic acid protect vascular endothelial cells against hypoxia-induced apoptosis via PI3K/AKT or ERK 1/2 signaling pathway. Life Sci. 2020, 252, 117666. [Google Scholar] [CrossRef]
  55. Kim, S.Y.; Lee, H.J.; Nam, J.W.; Seo, E.K.; Lee, Y.S. Coniferyl aldehyde reduces radiation damage through increased protein stability of heat shock transcriptional factor 1 by phosphorylation. Int. J. Radiat. Oncol. Biol. Phys. 2015, 91, 807–816. [Google Scholar] [CrossRef] [PubMed]
  56. Kim, K.M.; Heo, D.R.; Kim, Y.A.; Lee, J.; Kim, N.S.; Bang, O.S. Coniferaldehyde inhibits LPS-induced apoptosis through the PKC alpha/beta II/Nrf-2/HO-1 dependent pathway in RAW264.7 macrophage cells. Environ. Toxicol. Pharmacol. 2016, 48, 85–93. [Google Scholar] [CrossRef] [PubMed]
  57. Karamac, M.; Koleva, L.; Kancheva, V.D.; Amarowicz, R. The Structure-Antioxidant Activity Relationship of Ferulates. Molecules 2017, 22, 527. [Google Scholar] [CrossRef] [PubMed]
  58. Yi, B.; Hu, L.; Mei, W.; Zhou, K.; Wang, H.; Luo, Y.; Wei, X.; Dai, H. Antioxidant phenolic compounds of cassava (Manihot esculenta) from Hainan. Molecules 2011, 16, 10157–10167. [Google Scholar] [CrossRef]
  59. Wang, Y.; Gao, Y.; Li, X.; Sun, X.; Wang, Z.; Wang, H.; Nie, R.; Yu, W.; Zhou, Y. Coniferyl Aldehyde Inhibits the Inflammatory Effects of Leptomeningeal Cells by Suppressing the JAK2 Signaling. BioMed Res. Int. 2020, 2020, 4616308. [Google Scholar] [CrossRef] [PubMed]
  60. Park, J.M.; Park, J.E.; Park, J.S.; Leem, Y.H.; Kim, D.Y.; Hyun, J.W.; Kim, H.S. Anti-inflammatory and antioxidant mechanisms of coniferaldehyde in lipopolysaccharide-induced neuroinflammation: Involvement of AMPK/Nrf2 and TAK1/MAPK/NF-kappaB signaling pathways. Eur. J. Pharmacol. 2024, 979, 176850. [Google Scholar] [CrossRef]
  61. Dong, Y.; Stewart, T.; Bai, L.; Li, X.; Xu, T.; Iliff, J.; Shi, M.; Zheng, D.; Yuan, L.; Wei, T.; et al. Coniferaldehyde attenuates Alzheimer’s pathology via activation of Nrf2 and its targets. Theranostics 2020, 10, 179–200. [Google Scholar] [CrossRef] [PubMed]
  62. Al-Khayri, J.M.; Sahana, G.R.; Nagella, P.; Joseph, B.V.; Alessa, F.M.; Al-Mssallem, M.Q. Flavonoids as Potential Anti-Inflammatory Molecules: A Review. Molecules 2022, 27, 2901. [Google Scholar] [CrossRef]
  63. Avalos, Y.; Hernandez-Caceres, M.P.; Lagos, P.; Pinto-Nunez, D.; Rivera, P.; Burgos, P.; Diaz-Castro, F.; Joy-Immediato, M.; Venegas-Zamora, L.; Lopez-Gallardo, E.; et al. Palmitic acid control of ciliogenesis modulates insulin signaling in hypothalamic neurons through an autophagy-dependent mechanism. Cell Death Dis. 2022, 13, 659. [Google Scholar] [CrossRef]
  64. To, N.B.; Nguyen, Y.T.; Moon, J.Y.; Ediriweera, M.K.; Cho, S.K. Pentadecanoic Acid, an Odd-Chain Fatty Acid, Suppresses the Stemness of MCF-7/SC Human Breast Cancer Stem-Like Cells through JAK2/STAT3 Signaling. Nutrients 2020, 12, 1663. [Google Scholar] [CrossRef] [PubMed]
  65. Ysrafil, Y.; Sapiun, Z.; Slamet, N.S.; Mohamad, F.; Hartati, H.; Damiti, S.A.; Alexandra, F.D.; Rahman, S.; Masyeni, S.; Harapan, H.; et al. Anti-inflammatory activities of flavonoid derivates. ADMET DMPK 2023, 11, 331–359. [Google Scholar] [CrossRef]
  66. Venn-Watson, S.K.; Butterworth, C.N. Broader and safer clinically-relevant activities of pentadecanoic acid compared to omega-3: Evaluation of an emerging essential fatty acid across twelve primary human cell-based disease systems. PLoS ONE 2022, 17, e0268778. [Google Scholar] [CrossRef]
  67. Wang, Z.J.; Li, G.M.; Tang, W.L.; Yin, M. Neuroprotective effects of stearic acid against toxicity of oxygen/glucose deprivation or glutamate on rat cortical or hippocampal slices. Acta Pharmacol. Sin. 2006, 27, 145–150. [Google Scholar] [CrossRef]
  68. Charlot, A.; Morel, L.; Bringolf, A.; Georg, I.; Charles, A.L.; Goupilleau, F.; Geny, B.; Zoll, J. Octanoic Acid-Enrichment Diet Improves Endurance Capacity and Reprograms Mitochondrial Biogenesis in Skeletal Muscle of Mice. Nutrients 2022, 14, 2721. [Google Scholar] [CrossRef]
  69. Gupta, R.; Sharma, A.K.; Sharma, M.C.; Dobhal, M.P.; Gupta, R.S. Evaluation of antidiabetic and antioxidant potential of lupeol in experimental hyperglycaemia. Nat. Prod. Res. 2012, 26, 1125–1129. [Google Scholar] [CrossRef] [PubMed]
  70. Rychlicka, M.; Rot, A.; Gliszczynska, A. Biological Properties, Health Benefits and Enzymatic Modifications of Dietary Methoxylated Derivatives of Cinnamic Acid. Foods 2021, 10, 1417. [Google Scholar] [CrossRef]
  71. Georgiadis, I.; Karatzas, T.; Korou, L.M.; Katsilambros, N.; Perrea, D. Beneficial health effects of Chios Gum Mastic and peroxisome proliferator-activated receptors: Indications of common mechanisms. J. Med. Food 2015, 18, 1–10. [Google Scholar] [CrossRef]
  72. Toyama, T.; Nakamura, H.; Harano, Y.; Yamauchi, N.; Morita, A.; Kirishima, T.; Minami, M.; Itoh, Y.; Okanoue, T. PPARalpha ligands activate antioxidant enzymes and suppress hepatic fibrosis in rats. Biochem. Biophys. Res. Commun. 2004, 324, 697–704. [Google Scholar] [CrossRef] [PubMed]
  73. Shin, M.H.; Lee, S.R.; Kim, M.K.; Shin, C.Y.; Lee, D.H.; Chung, J.H. Activation of Peroxisome Proliferator-Activated Receptor Alpha Improves Aged and UV-Irradiated Skin by Catalase Induction. PLoS ONE 2016, 11, e0162628. [Google Scholar] [CrossRef] [PubMed]
  74. Muzio, G.; Barrera, G.; Pizzimenti, S. Peroxisome Proliferator-Activated Receptors (PPARs) and Oxidative Stress in Physiological Conditions and in Cancer. Antioxidants 2021, 10, 1734. [Google Scholar] [CrossRef]
  75. Popeijus, H.E.; van Otterdijk, S.D.; van der Krieken, S.E.; Konings, M.; Serbonij, K.; Plat, J.; Mensink, R.P. Fatty acid chain length and saturation influences PPARalpha transcriptional activation and repression in HepG2 cells. Mol. Nutr. Food Res. 2014, 58, 2342–2349. [Google Scholar] [CrossRef]
  76. Lo Verme, J.; Fu, J.; Astarita, G.; La Rana, G.; Russo, R.; Calignano, A.; Piomelli, D. The nuclear receptor peroxisome proliferator-activated receptor-alpha mediates the anti-inflammatory actions of palmitoylethanolamide. Mol. Pharmacol. 2005, 67, 15–19. [Google Scholar] [CrossRef]
  77. Malcher-Lopes, R.; Franco, A.; Tasker, J.G. Glucocorticoids shift arachidonic acid metabolism toward endocannabinoid synthesis: A non-genomic anti-inflammatory switch. Eur. J. Pharmacol. 2008, 583, 322–339. [Google Scholar] [CrossRef] [PubMed]
  78. Zeb, A. Concept, mechanism, and applications of phenolic antioxidants in foods. J. Food Biochem. 2020, 44, e13394. [Google Scholar] [CrossRef] [PubMed]
  79. Benadiba, M.; Miyake, J.A.; Colquhoun, A. Gamma-linolenic acid alters Ku80, E2F1, and bax expression and induces micronucleus formation in C6 glioma cells in vitro. IUBMB Life 2009, 61, 244–251. [Google Scholar] [CrossRef]
  80. Kou, J.J.; Shi, J.Z.; He, Y.Y.; Hao, J.J.; Zhang, H.Y.; Luo, D.M.; Song, J.K.; Yan, Y.; Xie, X.M.; Du, G.H.; et al. Luteolin alleviates cognitive impairment in Alzheimer’s disease mouse model via inhibiting endoplasmic reticulum stress-dependent neuroinflammation. Acta Pharmacol. Sin. 2022, 43, 840–849. [Google Scholar] [CrossRef] [PubMed]
  81. Cui, D.; Chen, Y.; Ye, B.; Guo, W.; Wang, D.; He, J. Natural products for the treatment of neurodegenerative diseases. Phytomedicine 2023, 121, 155101. [Google Scholar] [CrossRef] [PubMed]
  82. Lopez-Suarez, L.; Awabdh, S.A.; Coumoul, X.; Chauvet, C. The SH-SY5Y human neuroblastoma cell line, a relevant in vitro cell model for investigating neurotoxicology in human: Focus on organic pollutants. Neurotoxicology 2022, 92, 131–155. [Google Scholar] [CrossRef] [PubMed]
  83. Tsialtas, I.; Georgantopoulos, A.; Karipidou, M.E.; Kalousi, F.D.; Karra, A.G.; Leonidas, D.D.; Psarra, A.M.G. Anti-Apoptotic and Antioxidant Activities of the Mitochondrial Estrogen Receptor Beta in N2A Neuroblastoma Cells. Int. J. Mol. Sci. 2021, 22, 7620. [Google Scholar] [CrossRef]
  84. Psarra, A.M.G.; Sekeris, C.E. Glucocorticoids induce mitochondrial gene transcription in HepG2 cells Role of the mitochondrial glucocorticoid receptor. BBA-Mol. Cell Res. 2011, 1813, 1814–1821. [Google Scholar] [CrossRef]
  85. Tsialtas, I.; Gorgogietas, V.A.; Michalopoulou, M.; Komninou, A.; Liakou, E.; Georgantopoulos, A.; Kalousi, F.D.; Karra, A.G.; Protopapa, E.; Psarra, A.G. Neurotoxic effects of aluminum are associated with its interference with estrogen receptors signaling. Neurotoxicology 2020, 77, 114–126. [Google Scholar] [CrossRef] [PubMed]
  86. Sivandzade, F.; Bhalerao, A.; Cucullo, L. Analysis of the Mitochondrial Membrane Potential Using the Cationic JC-1 Dye as a Sensitive Fluorescent Probe. Bio-Protocol 2019, 9, e3128. [Google Scholar] [CrossRef]
  87. Pisha, E.; Chai, H.; Lee, I.-S.; Chagwedera, T.E.; Farnsworth, N.R.; Cordell, G.A.; Beecher, C.W.; Fong, H.H.; Kinghorn, A.D.; Brown, D.M.; et al. Discovery of betulinic acid as a selective inhibitor of human melanoma that functions by induction of apoptosis. Nat. Med. 1995, 1, 1046–1051. [Google Scholar] [CrossRef] [PubMed]
  88. Lou, H.; Li, H.; Zhang, S.; Lu, H.; Chen, Q. A Review on Preparation of Betulinic Acid and Its Biological Activities. Molecules 2021, 26, 5583. [Google Scholar] [CrossRef] [PubMed]
  89. Armah, F.A.; Annan, K.; Mensah, A.Y.; Amponsah, I.K.; Tocher, D.A.; Habtemariam, S. Erythroivorensin: A novel anti-inflammatory diterpene from the root-bark of Erythrophleum ivorense (A Chev.). Fitoterapia 2015, 105, 37–42. [Google Scholar] [CrossRef] [PubMed]
  90. Kim, S.J.; Quan, H.Y.; Jeong, K.J.; Kim, D.Y.; Kim, G.W.; Jo, H.K.; Chung, S.H. Beneficial Effect of Betulinic Acid on Hyperglycemia via Suppression of Hepatic Glucose Production. J. Agric. Food Chem. 2013, 62, 434–442. [Google Scholar] [CrossRef] [PubMed]
  91. Yoon, J.J.; Lee, Y.J.; Han, B.H.; Choi, E.S.; Kho, M.C.; Park, J.H.; Ahn, Y.M.; Kim, H.Y.; Kang, D.G.; Lee, H.S. Protective effect of betulinic acid on early atherosclerosis in diabetic apolipoprotein-E gene knockout mice. Eur. J. Pharmacol. 2017, 796, 224–232. [Google Scholar] [CrossRef]
  92. Gai, H.; Zhou, F.; Zhang, Y.; Ai, J.; Zhan, J.; You, Y.; Huang, W. Coniferaldehyde ameliorates the lipid and glucose metabolism in palmitic acid-induced HepG2 cells via the LKB1/AMPK signaling pathway. J. Food Sci. 2020, 85, 4050–4060. [Google Scholar] [CrossRef] [PubMed]
  93. Jeong, N.-H.; Lee, S.; Choi, Y.-A.; Song, K.-S.; Kim, S.-H. Inhibitory Effects of Euscaphic Acid in the Atopic Dermatitis Model by Reducing Skin Inflammation and Intense Pruritus. Inflammation 2022, 45, 1680–1691. [Google Scholar] [CrossRef] [PubMed]
  94. Jayaprakasha, G.K.; Rao, L.J.; Sakariah, K.K. Antioxidant activities of flavidin in different in vitro model systems. Bioorganic Med. Chem. 2004, 12, 5141–5146. [Google Scholar] [CrossRef]
  95. Wang, K.-L.; Yu, Y.-C.; Hsia, S.-M. Perspectives on the Role of Isoliquiritigenin in Cancer. Cancers 2021, 13, 115. [Google Scholar] [CrossRef]
  96. Hirchaud, F.; Hermetet, F.; Ablise, M.; Fauconnet, S.; Vuitton, D.A.; Prétet, J.-L.; Mougin, C. Isoliquiritigenin Induces Caspase-Dependent Apoptosis via Downregulation of HPV16 E6 Expression in Cervical Cancer Ca Ski Cells. Planta Medica 2013, 79, 1628–1635. [Google Scholar] [CrossRef]
  97. Caporali, S.; De Stefano, A.; Calabrese, C.; Giovannelli, A.; Pieri, M.; Savini, I.; Tesauro, M.; Bernardini, S.; Minieri, M.; Terrinoni, A. Anti-Inflammatory and Active Biological Properties of the Plant-Derived Bioactive Compounds Luteolin and Luteolin 7-Glucoside. Nutrients 2022, 14, 1155. [Google Scholar] [CrossRef] [PubMed]
  98. Samec, M.; Liskova, A.; Koklesova, L.; Samuel, S.M.; Zhai, K.; Buhrmann, C.; Varghese, E.; Abotaleb, M.; Qaradakhi, T.; Zulli, A.; et al. Flavonoids against the Warburg phenotype—Concepts of predictive, preventive and personalised medicine to cut the Gordian knot of cancer cell metabolism. EPMA J. 2020, 11, 377–398. [Google Scholar] [CrossRef] [PubMed]
  99. Kang, K.A.; Piao, M.J.; Ryu, Y.S.; Hyun, Y.J.; Park, J.E.; Shilnikova, K.; Zhen, A.X.; Kang, H.K.; Koh, Y.S.; Jeong, Y.J.; et al. Luteolin induces apoptotic cell death via antioxidant activity in human colon cancer cells. Int. J. Oncol. 2017, 51, 1169–1178. [Google Scholar] [CrossRef] [PubMed]
  100. Junior, R.F.d.A.; Eich, C.; Jorquera, C.; Schomann, T.; Baldazzi, F.; Chan, A.B.; Cruz, L.J. Ceramide and palmitic acid inhibit macrophage-mediated epithelial–mesenchymal transition in colorectal cancer. Mol. Cell. Biochem. 2020, 468, 153–168. [Google Scholar] [CrossRef] [PubMed]
  101. Yu, G.; Luo, H.; Zhang, N.; Wang, Y.; Li, Y.; Huang, H.; Liu, Y.; Hu, Y.; Liu, H.; Zhang, J.; et al. Loss of p53 Sensitizes Cells to Palmitic Acid-Induced Apoptosis by Reactive Oxygen Species Accumulation. Int. J. Mol. Sci. 2019, 20, 6268. [Google Scholar] [CrossRef] [PubMed]
  102. Mancini, A.; Imperlini, E.; Nigro, E.; Montagnese, C.; Daniele, A.; Orrù, S.; Buono, P. Biological and Nutritional Properties of Palm Oil and Palmitic Acid: Effects on Health. Molecules 2015, 20, 17339–17361. [Google Scholar] [CrossRef]
  103. Innis, S.M. Palmitic Acid in Early Human Development. Crit. Rev. Food Sci. Nutr. 2015, 56, 1952–1959. [Google Scholar] [CrossRef]
  104. Acosta-Montaño, P.; Rodríguez-Velázquez, E.; Ibarra-López, E.; Frayde-Gómez, H.; Mas-Oliva, J.; Delgado-Coello, B.; Rivero, I.A.; Alatorre-Meda, M.; Aguilera, J.; Guevara-Olaya, L.; et al. Fatty Acid and Lipopolysaccharide Effect on Beta Cells Proteostasis and its Impact on Insulin Se-cretion. Cells 2019, 8, 884. [Google Scholar] [CrossRef] [PubMed]
  105. Liu, Z.; Xiao, M.; Mo, Y.; Wang, H.; Han, Y.; Zhao, X.; Yang, X.; Liu, Z.; Xu, B. Emerging roles of protein palmitoylation and its modifying enzymes in cancer cell signal transduction and cancer therapy. Int. J. Biol. Sci. 2022, 18, 3447–3457. [Google Scholar] [CrossRef] [PubMed]
  106. Librán-Pérez, M.; Pereiro, P.; Figueras, A.; Novoa, B. Antiviral activity of palmitic acid via autophagic flux inhibition in zebrafish (Danio rerio). Fish Shellfish. Immunol. 2019, 95, 595–605. [Google Scholar] [CrossRef]
  107. Syed, I.; Lee, J.; Moraes-Vieira, P.M.; Donaldson, C.J.; Sontheimer, A.; Aryal, P.; Wellenstein, K.; Kolar, M.J.; Nelson, A.T.; Siegel, D.; et al. Palmitic Acid Hydroxystearic Acids Activate GPR40, Which Is Involved in Their Beneficial Effects on Glucose Homeostasis. Cell Metab. 2018, 27, 419–427. [Google Scholar] [CrossRef]
  108. Hammarstedt, A.; Syed, I.; Vijayakumar, A.; Eliasson, B.; Gogg, S.; Kahn, B.B.; Smith, U. Adipose tissue dysfunction is associated with low levels of the novel Palmitic Acid Hydroxystearic Acids. Sci. Rep. 2018, 8, 15757. [Google Scholar] [CrossRef] [PubMed]
  109. Yore, M.M.; Syed, I.; Moraes-Vieira, P.M.; Zhang, T.; Herman, M.A.; Homan, E.A.; Patel, R.T.; Lee, J.; Chen, S.; Peroni, O.D.; et al. Discovery of a Class of Endogenous Mammalian Lipids with Anti-Diabetic and Anti-inflammatory Effects. Cell 2014, 159, 318–332. [Google Scholar] [CrossRef]
  110. Khan, A.A.; Alanazi, A.M.; Jabeen, M.; Chauhan, A.; Abdelhameed, A.S. Design, synthesis and in vitro anticancer evaluation of a stearic acid-based ester conjugate. Anticancer Res. 2013, 33, 2517–2524. [Google Scholar]
  111. Fekete, K.; Györei, E.; Lohner, S.; Verduci, E.; Agostoni, C.; Decsi, T. Long-chain polyunsaturated fatty acid status in obesity: A systematic review and meta-analysis. Obes. Rev. 2015, 16, 488–497. [Google Scholar] [CrossRef]
  112. Hodson, L.; Skeaff, C.M.; Fielding, B.A. Fatty acid composition of adipose tissue and blood in humans and its use as a biomarker of dietary intake. Prog. Lipid Res. 2008, 47, 348–380. [Google Scholar] [CrossRef]
  113. Ren, J.; Chung, S.H. Anti-inflammatory effect of alpha-linolenic acid and its mode of action through the inhibition of nitric oxide production and inducible nitric oxide synthase gene expression via NF-kappaB and mitogen-activated protein kinase pathways. J. Agric. Food Chem. 2007, 55, 5073–5080. [Google Scholar] [CrossRef]
  114. Fan, N.; Fusco, J.L.; Rosenberg, D.W. Antioxidant and Anti-Inflammatory Properties of Walnut Constituents: Focus on Personalized Cancer Prevention and the Microbiome. Antioxidants 2023, 12, 982. [Google Scholar] [CrossRef]
  115. Yan, H.; Zhang, S.; Yang, L.; Jiang, M.; Xin, Y.; Liao, X.; Li, Y.; Lu, J. The Antitumor Effects of α-Linolenic Acid. J. Pers. Med. 2024, 14, 260. [Google Scholar] [CrossRef] [PubMed]
  116. Azrad, M.; Turgeon, C.; Demark-Wahnefried, W. Current Evidence Linking Polyunsaturated Fatty Acids with Cancer Risk and Progression. Front. Oncol. 2013, 3, 224. [Google Scholar] [CrossRef] [PubMed]
  117. Vieira, C.; Evangelista, S.; Cirillo, R.; Lippi, A.; Maggi, C.A.; Manzini, S. Effect of ricinoleic acid in acute and subchronic experimental models of inflammation. Mediat. Inflamm. 2000, 9, 223–228. [Google Scholar] [CrossRef] [PubMed]
  118. Pabiś, S.; Kula, J. Synthesis and Bioactivity of (R)-Ricinoleic Acid Derivatives: A Review. Curr. Med. Chem. 2016, 23, 4037–4056. [Google Scholar] [CrossRef]
  119. Minto, R.E.; Blacklock, B.J. Biosynthesis and function of polyacetylenes and allied natural products. Prog. Lipid Res. 2008, 47, 233–306. [Google Scholar] [CrossRef]
  120. Xu, T.; Tripathi, S.K.; Feng, Q.; Lorenz, M.C.; Wright, M.A.; Jacob, M.R.; Mask, M.M.; Baerson, S.R.; Li, X.-C.; Clark, A.M.; et al. A Potent Plant-Derived Antifungal Acetylenic Acid Mediates Its Activity by Interfering with Fatty Acid Homeostasis. Antimicrob. Agents Chemother. 2012, 56, 2894–2907. [Google Scholar] [CrossRef]
  121. Rengachar, P.; Bhatt, A.N.; Polavarapu, S.; Veeramani, S.; Krishnan, A.; Sadananda, M.; Das, U.N. Gamma-Linolenic Acid (GLA) Protects against Ionizing Radiation-Induced Damage: An In Vitro and In Vivo Study. Biomolecules 2022, 12, 797. [Google Scholar] [CrossRef]
  122. Das, U.N.; Rao, K.P. Effect of γ-linolenic acid and prostaglandins E1 on gamma-radiation and chemical-induced genetic damage to the bone marrow cells of mice. Prostaglandins Leukot. Essent. Fat. Acids 2006, 74, 165–173. [Google Scholar] [CrossRef] [PubMed]
  123. Leaver, H.A.; Wharton, S.B.; Bell, H.S.; Leaver-Yap, I.M.M.; Whittle, I. Highly unsaturated fatty acid induced tumour regression in glioma pharmacodynamics and bioavaila-bility of gamma linolenic acid in an implantation glioma model: Effects on tumour biomass, apoptosis and neuronal tissue histology. Prostaglandins Leukot Essent Fatty Acids 2002, 67, 283–292. [Google Scholar] [CrossRef]
  124. Das, U.N. Gamma-linolenic acid therapy of human glioma-a review of in vitro, in vivo, and clinical studies. Med. Sci. Monit. 2007, 13, RA119–RA131. [Google Scholar]
  125. Miyake, J.A.; Gomes, R.N.; Colquhoun, A. Gamma-Linolenic acid alters migration, proliferation and apoptosis in human and rat glioblastoma cells. Prostaglandins Other Lipid Mediat. 2020, 150, 106452. [Google Scholar] [CrossRef]
  126. Miyake, J.A.; Benadiba, M.; Colquhoun, A. Gamma-linolenic acid inhibits both tumour cell cycle progression and angi-ogenesis in the orthotopic C6 glioma model through changes in VEGF, Flt1, ERK1/2, MMP2, cyclin D1, pRb, p53 and p27 protein expression. Lipids Health Dis. 2009, 8. [Google Scholar] [CrossRef]
  127. Oepen, K.; Özbek, H.; Schüffler, A.; Liermann, J.C.; Thines, E.; Schneider, D. Myristic Acid Inhibits the Activity of the Bacterial ABC Transporter BmrA. Int. J. Mol. Sci. 2021, 22, 13565. [Google Scholar] [CrossRef] [PubMed]
  128. Contreras, C.M.; Rodríguez-Landa, J.F.; García-Ríos, R.I.; Cueto-Escobedo, J.; Guillen-Ruiz, G.; Bernal-Morales, B. Myristic Acid Produces Anxiolytic-Like Effects in Wistar Rats in the Elevated Plus Maze. Bio. Med. Res. Int. 2014, 2014, 1–8. [Google Scholar] [CrossRef]
  129. Javid, S.; Ather, H.; Hani, U.; Siddiqua, A.; Ansari, S.M.A.; Shanmugarajan, D.; Kumar, H.Y.; Arivuselvam, R.; Purohit, M.N.; Kumar, B.R.P. Discovery of Novel Myristic Acid Derivatives as N-Myristoyltransferase Inhibitors: Design, Synthesis, Analysis, Computational Studies and Antifungal Activity. Antibiotics 2023, 12, 1167. [Google Scholar] [CrossRef]
  130. Prasath, K.G.; Alexpandi, R.; Parasuraman, R.; Pavithra, M.; Ravi, A.V.; Pandian, S.K. Anti-inflammatory potential of myristic acid and palmitic acid synergism against systemic candidiasis in Danio rerio (Zebrafish). Biomed. Pharmacother. 2020, 133, 111043. [Google Scholar] [CrossRef]
  131. Alonso-Castro, A.J.; Serrano-Vega, R.; Gutiérrez, S.P.; Isiordia-Espinoza, M.A.; Solorio-Alvarado, C.R. Myristic acid reduces skin inflammation and nociception. J. Food Biochem. 2021, 46, e14013. [Google Scholar] [CrossRef]
  132. Pompéia, C.; Lopes, L.; Miyasaka, C.; Procópio, J.; Sannomiya, P.; Curi, R. Effect of fatty acids on leukocyte function. Braz. J. Med Biol. Res. 2000, 33, 1255–1268. [Google Scholar] [CrossRef]
  133. Brash, A.R. Arachidonic acid as a bioactive molecule. J. Clin. Investig. 2001, 107, 1339–1345. [Google Scholar] [CrossRef] [PubMed]
  134. Tallima, H.; El Ridi, R. Arachidonic acid: Physiological roles and potential health benefits—A review. J. Adv. Res. 2018, 11, 33–41. [Google Scholar] [CrossRef] [PubMed]
  135. Tokuda, H.; Kontani, M.; Kawashima, H.; Kiso, Y.; Shibata, H.; Osumi, N. Differential effect of arachidonic acid and docosahexaenoic acid on age-related decreases in hippocampal neurogenesis. Neurosci. Res. 2014, 88, 58–66. [Google Scholar] [CrossRef] [PubMed]
  136. Fukaya, T.; Gondaira, T.; Kashiyae, Y.; Kotani, S.; Ishikura, Y.; Fujikawa, S.; Kiso, Y.; Sakakibara, M. Arachidonic acid preserves hippocampal neuron membrane fluidity in senescent rats. Neurobiol. Aging 2007, 28, 1179–1186. [Google Scholar] [CrossRef] [PubMed]
  137. Antollini, S.S.; Barrantes, F.J. Fatty Acid Regulation of Voltage- and Ligand-Gated Ion Channel Function. Front. Physiol. 2016, 7, 573. [Google Scholar] [CrossRef]
  138. Wang, Z.-J.; Liang, C.-L.; Li, G.-M.; Yu, C.-Y.; Yin, M. Neuroprotective effects of arachidonic acid against oxidative stress on rat hippocampal slices. Chem. Interactions 2006, 163, 207–217. [Google Scholar] [CrossRef]
  139. Pérez, R.; Matabosch, X.; Llebaria, A.; Balboa, M.A.; Balsinde, J. Blockade of arachidonic acid incorporation into phospholipids induces apoptosis in U937 promonocytic cells. J. Lipid Res. 2006, 47, 484–491. [Google Scholar] [CrossRef]
  140. Trostchansky, A.; Wood, I.; Rubbo, H. Regulation of arachidonic acid oxidation and metabolism by lipid electrophiles. Prostaglandins Other Lipid Mediat. 2020, 152, 106482. [Google Scholar] [CrossRef]
  141. Trostchansky, A.; Rubbo, H. Anti-inflammatory signaling actions of electrophilic nitro-arachidonic acid in vascular cells and astrocytes. Arch. Biochem. Biophys. 2017, 617, 155–161. [Google Scholar] [CrossRef]
  142. Sala, A.; Proschak, E.; Steinhilber, D.; Rovati, G.E. Two-pronged approach to anti-inflammatory therapy through the modulation of the arachidonic acid cascade. Biochem. Pharmacol. 2018, 158, 161–173. [Google Scholar] [CrossRef]
  143. Wang, T.; Fu, X.; Chen, Q.; Patra, J.K.; Wang, D.; Wang, Z.; Gai, Z. Arachidonic Acid Metabolism and Kidney Inflammation. Int. J. Mol. Sci. 2019, 20, 3683. [Google Scholar] [CrossRef] [PubMed]
  144. Wang, Y.; Wei, X.; Xiao, X.; Hui, R.; Card, J.W.; Carey, M.A.; Wang, D.W.; Zeldin, D.C. Arachidonic acid epoxygenase metabolites stimulate endothelial cell growth and angiogenesis via mito-gen-activated protein kinase and phosphatidylinositol 3-kinase/Akt signaling pathways. J. Pharmacol. Exp. Ther. 2005, 314, 522–532. [Google Scholar] [CrossRef] [PubMed]
  145. Li, N.; Liu, J.-Y.; Qiu, H.; Harris, T.R.; Sirish, P.; Hammock, B.D.; Chiamvimonvat, N. Use of Metabolomic Profiling in the Study of Arachidonic Acid Metabolism in Cardiovascular Disease. Congest. Hear. Fail. 2011, 17, 42–46. [Google Scholar] [CrossRef] [PubMed]
  146. Sonnweber, T.; Pizzini, A.; Nairz, M.; Weiss, G.; Tancevski, I. Arachidonic Acid Metabolites in Cardiovascular and Metabolic Diseases. Int. J. Mol. Sci. 2018, 19, 3285. [Google Scholar] [CrossRef] [PubMed]
  147. Tavolari, S.; Bonafe, M.; Marini, M.; Ferreri, C.; Bartolini, G.; Brighenti, E.; Manara, S.; Tomasi, V.; Laufer, S.; Guarnieri, T. Licofelone, a dual COX/5-LOX inhibitor, induces apoptosis in HCA-7 colon cancer cells through the mi-tochondrial pathway independently from its ability to affect the arachidonic acid cascade. Carcinogenesis 2008, 29, 371–380. [Google Scholar] [CrossRef] [PubMed]
  148. Colombero, C.; Cárdenas, S.; Venara, M.; Martin, A.; Pennisi, P.; Barontini, M.; Nowicki, S. Cytochrome 450 metabolites of arachidonic acid (20-HETE, 11,12-EET and 14,15-EET) promote pheo-chromocytoma cell growth and tumor associated angiogenesis. Biochimie 2020, 171, 147–157. [Google Scholar] [CrossRef]
  149. Shao, J.; Wang, H.; Yuan, G.; Chen, Z.; Li, Q. Involvement of the arachidonic acid cytochrome P450 epoxygenase pathway in the proliferation and invasion of human multiple myeloma cells. Peer.J. 2016, 4, e1925. [Google Scholar] [CrossRef]
  150. Pozzi, A.; Popescu, V.; Yang, S.; Mei, S.; Shi, M.; Puolitaival, S.M.; Caprioli, R.M.; Capdevila, J.H. The Anti-tumorigenic Properties of Peroxisomal Proliferator-activated Receptor α Are Arachidonic Acid Epoxygenase-mediated. J. Biol. Chem. 2010, 285, 12840–12850. [Google Scholar] [CrossRef]
  151. Venn-Watson, S.; Lumpkin, R.; Dennis, E.A. Efficacy of dietary odd-chain saturated fatty acid pentadecanoic acid parallels broad associated health benefits in humans: Could it be essential? Sci. Rep. 2020, 10, 8161. [Google Scholar] [CrossRef]
  152. Shafaghat, A. Antioxidant, Antimicrobial Activities and Fatty Acid Components of Flower, Leaf, Stem and Seed of Hypericum scabrum. Nat. Prod. Commun. 2011, 6, 1739–1742. [Google Scholar] [CrossRef]
  153. Singh, D.; Mehghini, P.; Rodriguez-Palacios, A.; Di Martino, L.; Cominelli, F.; Basson, A.R. Anti-Inflammatory Effect of Dietary Pentadecanoic Fatty Acid Supplementation on Inflammatory Bowel Disease in SAMP1/YitFc Mice. Nutrients 2024, 16, 3031. [Google Scholar] [CrossRef] [PubMed]
  154. Wang, X.; Ye, X.-L.; Liu, R.; Chen, H.-L.; Bai, H.; Liang, X.; Zhang, X.-D.; Wang, Z.; Li, W.-L.; Hai, C.-X. Antioxidant activities of oleanolic acid in vitro: Possible role of Nrf2 and MAP kinases. Chem. Biol. Interact. 2010, 184, 328–337. [Google Scholar] [CrossRef]
  155. Wang, X.; Liu, R.; Zhang, W.; Zhang, X.; Liao, N.; Wang, Z.; Li, W.; Qin, X.; Hai, C. Oleanolic acid improves hepatic insulin resistance via antioxidant, hypolipidemic and anti-inflammatory effects. Mol. Cell. Endocrinol. 2013, 376, 70–80. [Google Scholar] [CrossRef]
  156. Dzubak, P.; Hajduch, M.; Vydra, D.; Hustova, A.; Kvasnica, M.; Biedermann, D.; Markova, L.; Urban, M.; Sarek, J. Pharmacological activities of natural triterpenoids and their therapeutic implications. Nat. Prod. Rep. 2006, 23, 394–411. [Google Scholar] [CrossRef]
  157. Allouche, Y.; Warleta, F.; Campos, M.; Sánchez-Quesada, C.; Uceda, M.; Beltrán, G.; Gaforio, J.J. Antioxidant, Antiproliferative, and Pro-apoptotic Capacities of Pentacyclic Triterpenes Found in the Skin of Olives on MCF-7 Human Breast Cancer Cells and Their Effects on DNA Damage. J. Agric. Food Chem. 2010, 59, 121–130. [Google Scholar] [CrossRef]
  158. Du, Y.; Ko, K.M. Oleanolic Acid Protects against Myocardial Ischemia-Reperfusion Injury by Enhancing Mitochondrial Antioxidant Mechanism Mediated by Glutathione and α-Tocopherol in Rats. Planta Medica 2005, 72, 222–227. [Google Scholar] [CrossRef]
  159. Tang, Z.Y.; Li, Y.; Tang, Y.T.; Ma, X.D.; Tang, Z.Y. Anticancer activity of oleanolic acid and its derivatives: Recent advances in evidence, target profiling and mechanisms of action. Biomed. Pharmacother. 2022, 145, 112397. [Google Scholar] [CrossRef]
  160. Gupta, S.; Kalani, K.; Saxena, M.; Srivastava, S.K.; Agrawal, S.K.; Suri, N.; Saxena, A.K. Cytotoxic Evaluation of Semisynthetic Ester and Amide Derivatives of Oleanolic Acid. Nat. Prod. Commun. 2010, 5, 1567–1570. [Google Scholar] [CrossRef]
  161. Roohbakhsh, A.; Parhiz, H.; Soltani, F.; Rezaee, R.; Iranshahi, M. Molecular mechanisms behind the biological effects of hesperidin and hesperetin for the prevention of cancer and cardiovascular diseases. Life Sci. 2015, 124, 64–74. [Google Scholar] [CrossRef]
  162. Parhiz, H.; Roohbakhsh, A.; Soltani, F.; Rezaee, R.; Iranshahi, M. Antioxidant and Anti-Inflammatory Properties of the Citrus Flavonoids Hesperidin and Hesperetin: An Updated Review of their Molecular Mechanisms and Experimental Models. Phytotherapy Res. 2014, 29, 323–331. [Google Scholar] [CrossRef]
  163. de Oliveira, J.M.P.F.; Santos, C.; Fernandes, E. Therapeutic potential of hesperidin and its aglycone hesperetin: Cell cycle regulation and apoptosis induction in cancer models. Phytomedicine. 2019, 73, 152887. [Google Scholar] [CrossRef]
  164. Fontana, A.; Spolaore, B.; de Laureto, P.P. The biological activities of protein/oleic acid complexes reside in the fatty acid. Biochim. Biophys. Acta (BBA)-Proteins Proteom. 2013, 1834, 1125–1143. [Google Scholar] [CrossRef] [PubMed]
  165. Carrillo, C.; Cavia Camarero, M.D.M.; Alonso-Torre, S.R. Antitumor effect of oleic acid; mechanisms of action: A review. Nutr. Hosp. 2012, 27, 1860–1865. [Google Scholar] [CrossRef]
  166. Carrillo, C.; Cavia Mdel, M.; Alonso-Torre, S. Role of oleic acid in immune system; mechanism of action; a review. Nutr. Hosp. 2012, 27, 978–990. [Google Scholar] [CrossRef]
  167. Masner, M.; Lujea, N.; Bisbal, M.; Acosta, C.; Kunda, P. Linoleic and oleic acids enhance cell migration by altering the dynamics of microtubules and the remodeling of the actin cytoskeleton at the leading edge. Sci. Rep. 2021, 11, 14984. [Google Scholar] [CrossRef]
  168. Benjamin, S.; Spener, F. Conjugated linoleic acids as functional food: An insight into their health benefits. Nutr. Metab. 2009, 6, 1–13. [Google Scholar] [CrossRef] [PubMed]
  169. Kolar, M.J.; Konduri, S.; Chang, T.; Wang, H.; McNerlin, C.; Ohlsson, L.; Härröd, M.; Siegel, D.; Saghatelian, A. Linoleic acid esters of hydroxy linoleic acids are anti-inflammatory lipids found in plants and mammals. J. Biol. Chem. 2019, 294, 10698–10707. [Google Scholar] [CrossRef]
  170. Cahoon, E.B.; Li-Beisson, Y. Plant unusual fatty acids: Learning from the less common. Curr. Opin. Plant Biol. 2020, 55, 66–73. [Google Scholar] [CrossRef]
  171. Scott, S.; Cahoon, E.B.; Busta, L. Variation on a theme: The structures and biosynthesis of specialized fatty acid natural products in plants. Plant J. 2022, 111, 954–965. [Google Scholar] [CrossRef]
  172. Lin, W.-C.; Hsu, K.-C.; You, M.-F.; Lee, K.-H.; Chi, C.-H.; Chen, J.-Y. Octanoic acid promotes clearance of antibiotic-tolerant cells and eradicates biofilms of Staphylococcus aureus isolated from recurrent bovine mastitis. Biofilm 2023, 6, 100149. [Google Scholar] [CrossRef] [PubMed]
  173. Zhang, H.; Dolan, H.L.; Ding, Q.; Wang, S.; Tikekar, R.V. Antimicrobial action of octanoic acid against Escherichia coli O157:H7 during washing of baby spinach and grape tomatoes. Food Res. Int. 2019, 125, 108523. [Google Scholar] [CrossRef]
  174. Mączka, W.; Duda-Madej, A.; Grabarczyk, M.; Wińska, K. Natural Compounds in the Battle against Microorganisms—Linalool. Molecules 2022, 27, 6928. [Google Scholar] [CrossRef] [PubMed]
  175. Pereira, I.; Severino, P.; Santos, A.C.; Silva, A.M.; Souto, E.B. Linalool Bioactive Properties and Potential Applicability in Drug Delivery Systems. Colloid Surface Biointerface 2018, 171, 566–578. [Google Scholar] [CrossRef]
  176. Liu, X.; Cai, J.; Chen, H.; Zhong, Q.; Hou, Y.; Chen, W.; Chen, W. Antibacterial activity and mechanism of linalool against Pseudomonas aeruginosa. Microb. Pathog. 2020, 141, 103980. [Google Scholar] [CrossRef]
  177. Elbe, H.; Ozturk, F.; Yigitturk, G.; Baygar, T.; Cavusoglu, T. Anticancer activity of linalool: Comparative investigation of ultrastructural changes and apoptosis in breast cancer cells. Ultrastruct. Pathol. 2022, 46, 348–358. [Google Scholar] [CrossRef]
  178. Zielińska-Błajet, M.; Feder-Kubis, J. Monoterpenes and Their Derivatives—Recent Development in Biological and Medical Applications. Int. J. Mol. Sci. 2020, 21, 7078. [Google Scholar] [CrossRef]
  179. Koziol, A.; Stryjewska, A.; Librowski, T.; Salat, K.; Gawel, M.; Moniczewski, A.; Lochynski, S. An Overview of the Pharmacological Properties and Potential Applications of Natural Monoterpenes. Mini-Reviews Med. Chem. 2015, 14, 1156–1168. [Google Scholar] [CrossRef]
  180. Khusnutdinova, E.F.; Sinou, V.; Babkov, D.A.; Kazakova, O.; Brunel, J.M. Development of New Antimicrobial Oleanonic Acid Polyamine Conjugates. Antibiotics 2022, 11, 94. [Google Scholar] [CrossRef]
  181. Castellano, J.M.; Ramos-Romero, S.; Perona, J.S. Oleanolic Acid: Extraction, Characterization and Biological Activity. Nutrients 2022, 14, 623. [Google Scholar] [CrossRef]
  182. Vuorinen, A.; Seibert, J.; Papageorgiou, V.P.; Rollinger, J.M.; Odermatt, A.; Schuster, D.; Assimopoulou, A.N. Pistacia lentiscus Oleoresin: Virtual Screening and Identification of Masticadienonic and Isomasticadi-enonic Acids as Inhibitors of 11beta-Hydroxysteroid Dehydrogenase 1. Planta Med. 2015, 81, 525–532. [Google Scholar] [CrossRef]
  183. Meng, Q.; Li, J.; Wang, C.; Shan, A. Biological function of resveratrol and its application in animal production: A review. J. Anim. Sci. Biotechnol. 2023, 14, 1–23. [Google Scholar] [CrossRef] [PubMed]
  184. Bhat, K.P.L.; Kosmeder, J.W., 2nd; Pezzuto, J.M. Biological effects of resveratrol. Antioxid. Redox Signal 2001, 3, 1041–1064. [Google Scholar] [CrossRef] [PubMed]
  185. Yu, X.; Jia, Y.; Ren, F. Multidimensional biological activities of resveratrol and its prospects and challenges in the health field. Front. Nutr. 2024, 11, 1408651. [Google Scholar] [CrossRef]
  186. Santos, J.A.; de Carvaho, G.S.; Oliveira, V.; Raposo, N.R.; da Silva, A.D. Resveratrol and Analogues: A Review of Antioxidant Activity and Applications to Human Health. Recent Patents Food Nutr. Agric. 2013, 5, 144–153. [Google Scholar] [CrossRef] [PubMed]
  187. Płowuszyńska, A.; Gliszczyńska, A. Recent Developments in Therapeutic and Nutraceutical Applications of p-Methoxycinnamic Acid from Plant Origin. Molecules 2021, 26, 3827. [Google Scholar] [CrossRef]
  188. Adisakwattana, S. Cinnamic Acid and Its Derivatives: Mechanisms for Prevention and Management of Diabetes and Its Complications. Nutrients 2017, 9, 163. [Google Scholar] [CrossRef] [PubMed]
  189. Yoon, B.K.; Jackman, J.A.; Valle-González, E.R.; Cho, N.-J. Antibacterial Free Fatty Acids and Monoglycerides: Biological Activities, Experimental Testing, and Therapeutic Applications. Int. J. Mol. Sci. 2018, 19, 1114. [Google Scholar] [CrossRef]
  190. Kim, J.J.; Kim, H.K. Antioxidant and Antibacterial Activity of Caprylic Acid Vanillyl Ester Produced by Lipase-Mediated Transesterification. J. Microbiol. Biotechnol. 2020, 31, 317–326. [Google Scholar] [CrossRef]
  191. Altinoz, M.A.; Ozpinar, A.; Seyfried, T.N. Seyfried, Caprylic (Octanoic) Acid as a Potential Fatty Acid Chemotherapeutic for Glio-blastoma. Prostaglandins Leukot Essent Fatty Acids 2020, 159, 102142. [Google Scholar] [CrossRef]
  192. Okere, I.C.; McElfresh, T.A.; Brunengraber, D.Z.; Martini, W.; Sterk, J.P.; Huang, H.; Chandler, M.P.; Brunengraber, H.; Stanley, W.C. Differential effects of heptanoate and hexanoate on myocardial citric acid cycle intermediates following ischemia-reperfusion. J. Appl. Physiol. 2006, 100, 76–82. [Google Scholar] [CrossRef] [PubMed]
  193. Huang, C.B.; Alimova, Y.; Myers, T.M.; Ebersole, J.L. Short- and medium-chain fatty acids exhibit antimicrobial activity for oral microorganisms. Arch. Oral Biol. 2011, 56, 650–654. [Google Scholar] [CrossRef]
  194. Wisetsai, A.; Lekphrom, R.; Bua-art, S.; Suebrasri, T.; Boonlue, S.; Tontapha, S.; Amornkitbamrung, V.; Senawong, T.; Schevenels, F.T. Scalarane Sesterterpenoids with Antibacterial and Anti-Proliferative Activities from the Mushroom Ne-onothopanus nambi. Molecules 2021, 26, 7667. [Google Scholar] [CrossRef]
  195. Zhang, C.; Liu, Y. Targeting cancer with sesterterpenoids: The new potential antitumor drugs. J. Nat. Med. 2015, 69, 255–266. [Google Scholar] [CrossRef]
  196. Jarboe, L.R.; Royce, L.A.; Liu, P. Understanding biocatalyst inhibition by carboxylic acids. Front. Microbiol. 2013, 4, 272. [Google Scholar] [CrossRef] [PubMed]
  197. Kallscheuer, N.; Polen, T.; Bott, M.; Marienhagen, J. Reversal of beta-oxidative pathways for the microbial production of chemicals and polymer building blocks. Metab. Eng. 2017, 42, 33–42. [Google Scholar] [CrossRef]
  198. Zhang, B.; Du, H.; Sun, M.; Wu, X.; Li, Y.; Wang, Z.; Xiao, Y.; Peng, F. Comparison of lauric acid and 12-hydroxylauric acid in the alleviation of drought stress in peach (Prunus persica (L.) Batsch). Front. Plant Sci. 2022, 13, 1025569. [Google Scholar] [CrossRef] [PubMed]
  199. Deen, A.; Visvanathan, R.; Wickramarachchi, D.; Marikkar, N.; Nammi, S.; Jayawardana, B.C.; Liyanage, R. Chemical composition and health benefits of coconut oil: An overview. J. Sci. Food Agric. 2020, 101, 2182–2193. [Google Scholar] [CrossRef] [PubMed]
  200. Liang, C.; Gao, W.; Ge, T.; Tan, X.; Wang, J.; Liu, H.; Wang, Y.; Han, C.; Xu, Q.; Wang, Q. Lauric Acid Is a Potent Biological Control Agent That Damages the Cell Membrane of Phytophthora sojae. Front. Microbiol. 2021, 12. [Google Scholar] [CrossRef] [PubMed]
  201. Takagi, T.; Fujiwara-Tani, R.; Mori, S.; Kishi, S.; Nishiguchi, Y.; Sasaki, T.; Ogata, R.; Ikemoto, A.; Sasaki, R.; Ohmori, H.; et al. Lauric Acid Overcomes Hypoxia-Induced Gemcitabine Chemoresistance in Pancreatic Ductal Adenocarci-noma. Int. J. Mol. Sci. 2023, 24, 7506. [Google Scholar] [CrossRef]
  202. Aihara, K.-I.; Azuma, H.; Akaike, M.; Ikeda, Y.; Yamashita, M.; Sudo, T.; Hayashi, H.; Yamada, Y.; Endoh, F.; Fujimura, M.; et al. Disruption of Nuclear Vitamin D Receptor Gene Causes Enhanced Thrombogenicity in Mice. J. Biol. Chem. 2004, 279, 35798–35802. [Google Scholar] [CrossRef] [PubMed]
  203. Pilz, S.; Tomaschitz, A.; Drechsler, C.; Zittermann, A.; Dekker, J.M.; Marz, W. Vitamin D Supplementation: A Promising Approach for the Prevention and Treatment of Strokes. Curr. Drug Targets 2011, 12, 88–96. [Google Scholar] [CrossRef] [PubMed]
  204. Wang, T.J.; Pencina, M.J.; Booth, S.L.; Jacques, P.F.; Ingelsson, E.; Lanier, K.; Benjamin, E.J.; D’Agostino, R.B.; Wolf, M.; Vasan, R.S. Vitamin D Deficiency and Risk of Cardiovascular Disease. Circulation 2008, 117, 503–511. [Google Scholar] [CrossRef]
  205. Pilkey, N.G.; Novosel, O.; Roy, A.; Wilson, T.E.; Sharma, J.; Khan, S.; Kapuria, S.; Adams, M.A.; Holden, R.M. Does Native Vitamin D Supplementation Have Pleiotropic Effects in Patients with End-Stage Kidney Disease? A Systematic Review of Randomized Trials. Nutrients 2023, 15, 3072. [Google Scholar] [CrossRef] [PubMed]
  206. Nguyen, T.D.; Prykhodko, O.; Hållenius, F.F.; Nyman, M. Monovalerin and trivalerin increase brain acetic acid, decrease liver succinic acid, and alter gut microbiota in rats fed high-fat diets. Eur. J. Nutr. 2018, 58, 1545–1560. [Google Scholar] [CrossRef]
  207. Nguyen, T.D.; Hållenius, F.F.; Lin, X.; Nyman, M.; Prykhodko, O. Monobutyrin and Monovalerin Affect Brain Short-Chain Fatty Acid Profiles and Tight-Junction Protein Expression in ApoE-Knockout Rats Fed High-Fat Diets. Nutrients 2020, 12, 1202. [Google Scholar] [CrossRef] [PubMed]
  208. Onrust, L.; Van Driessche, K.; Ducatelle, R.; Schwarzer, K.; Haesebrouck, F.; Van Immerseel, F. Valeric acid glyceride esters in feed promote broiler performance and reduce the incidence of necrotic en-teritis. Poult. Sci. 2018, 97, 2303–2311. [Google Scholar] [CrossRef]
  209. Zhao, L.; Zheng, L. A Review on Bioactive Anthraquinone and Derivatives as the Regulators for ROS. Molecules 2023, 28, 8139. [Google Scholar] [CrossRef]
  210. Berillo, D.; Kozhahmetova, M.; Lebedeva, L. Overview of the Biological Activity of Anthraquinons and Flavanoids of the Plant Rumex Species. Molecules 2022, 27, 1204. [Google Scholar] [CrossRef] [PubMed]
  211. Feng, S.; Wang, W. Bioactivities and Structure–Activity Relationships of Natural Tetrahydroanthraquinone Compounds: A Review. Front. Pharmacol. 2020, 11, 799. [Google Scholar] [CrossRef]
  212. Wang, X.; Fu, H.Y.; He, W.; Xiang, Y.T.; Yang, Z.C.; Kuang, Y.; Yang, S.X. Synthesis and Antibacterial Activity Evaluation of Biphenyl and Dibenzofuran Derivatives as Potential An-timicrobial Agents against Antibiotic-Resistant Bacteria. Curr. Issues Mol. Biol. 2022, 44, 4087–4099. [Google Scholar] [CrossRef]
  213. Wang, Y.; Huang, Q.; Zhang, L.; Zheng, C.; Xu, H. Biphenyls in Clusiaceae: Isolation, structure diversity, synthesis and bioactivity. Front. Chem. 2022, 10, 987009. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Resistance to H2O2-induced reduction in neuroblastoma SH-SY5Y cells viability, by the different polarity P. lenticonus/Chios mastiha’s fractions. (A) Relative cell viability, assessed by MTT assay, of SH-SY5Y cells treated with 50–500 μΜ H2O2 for 6 h and 1 mM or 2 mM H2O2 for 3 h. Viability of vehicle-treated (1/1000 v/v DMSO) control cells was set as 1. (B) Relative cell viability of SH-SY5Y cells pretreated or not with ap, mp, and p P. lenticonus/Chios mastiha’s fractions for 24 h and/or not subsequently subjected to 700 μΜ H2O2 for 6 h. Viability of control (vehicle-treated, 1/1000 v/v DMSO) cells was set as 1. Relative cell viability is expressed as the viability of the cells pretreated with various concentrations of the ap, mp, and p fractions compared to the viability of the control cells. Data were analyzed by 1-way ANOVA (Figure 1A) and 2-way ANOVA (Figure 1B), respectively, and are expressed as mean ± SD (n = 4, n = 5), ** p < 0.01 *** p < 0.001.
Figure 1. Resistance to H2O2-induced reduction in neuroblastoma SH-SY5Y cells viability, by the different polarity P. lenticonus/Chios mastiha’s fractions. (A) Relative cell viability, assessed by MTT assay, of SH-SY5Y cells treated with 50–500 μΜ H2O2 for 6 h and 1 mM or 2 mM H2O2 for 3 h. Viability of vehicle-treated (1/1000 v/v DMSO) control cells was set as 1. (B) Relative cell viability of SH-SY5Y cells pretreated or not with ap, mp, and p P. lenticonus/Chios mastiha’s fractions for 24 h and/or not subsequently subjected to 700 μΜ H2O2 for 6 h. Viability of control (vehicle-treated, 1/1000 v/v DMSO) cells was set as 1. Relative cell viability is expressed as the viability of the cells pretreated with various concentrations of the ap, mp, and p fractions compared to the viability of the control cells. Data were analyzed by 1-way ANOVA (Figure 1A) and 2-way ANOVA (Figure 1B), respectively, and are expressed as mean ± SD (n = 4, n = 5), ** p < 0.01 *** p < 0.001.
Molecules 30 00997 g001
Figure 2. Reduced ROS production in SH-SY5Y cells pretreated with the P. lenticonus/Chios mastiha’s fractions and subsequently subjected to H2O2-induced oxidative stress. (A) Representative confocal microscopy single images of SH-SY5Y cells stained with Hoechst-33342 (nuclear staining), MitoTracker CMXRos (mitochondrial staining in living cells, Red), DCF (ROS production dye, Green) upon pretreatment with 10 μg/mL ap, 5 μg/mL mp and 10 μg/mL p P. lenticonus/Chios mastiha’s fractions for 24 h and/or not subsequently subjected to 700 μΜ H2O2 for 6 h. Vehicle-treated, control cells were incubated with 1/1000 v/v DMSO. Representative images were taken with Zeiss LSM-800 confocal microscope in 40× objective (B) Quantification analysis of DCF staining per CMX staining of single cells. The ratio of DCF/CMX staining in control cells was set as 1. Data were analyzed by 2-way ANOVA (Figure 2) and relative DCF fluorescence density is expressed as mean ± SD (n = 60–80), *** p < 0.001.
Figure 2. Reduced ROS production in SH-SY5Y cells pretreated with the P. lenticonus/Chios mastiha’s fractions and subsequently subjected to H2O2-induced oxidative stress. (A) Representative confocal microscopy single images of SH-SY5Y cells stained with Hoechst-33342 (nuclear staining), MitoTracker CMXRos (mitochondrial staining in living cells, Red), DCF (ROS production dye, Green) upon pretreatment with 10 μg/mL ap, 5 μg/mL mp and 10 μg/mL p P. lenticonus/Chios mastiha’s fractions for 24 h and/or not subsequently subjected to 700 μΜ H2O2 for 6 h. Vehicle-treated, control cells were incubated with 1/1000 v/v DMSO. Representative images were taken with Zeiss LSM-800 confocal microscope in 40× objective (B) Quantification analysis of DCF staining per CMX staining of single cells. The ratio of DCF/CMX staining in control cells was set as 1. Data were analyzed by 2-way ANOVA (Figure 2) and relative DCF fluorescence density is expressed as mean ± SD (n = 60–80), *** p < 0.001.
Molecules 30 00997 g002
Figure 3. Antioxidant activities of the different polarity fractions of P. lenticonus/Chios mastiha’s contribute to the maintenance of the mitochondrial membrane potential in H2O2-treated SH-SY5Y cells. (A) Representative images of JC-1 staining (JC-1 dimers-red aggregates; JC-1 monomers-green fluorescence) and Hoechst-33342 staining (nuclear staining) in human neuroblastoma SH-SY5Y cells, pretreated or not with 10 μg/mL ap, 5 μg/mL mp and 10 μg/mL p P. lenticonus/Chios mastiha’s fractions for 24 h and then, subjected or not to 700 μΜ H2O2 for 6 h. Vehicle-treated, control cells were incubated with 1/1000 v/v DMSO. Representative images were taken with a Zeiss LSM-800 confocal microscope in 40× objective. (B) Quantification of green to red staining. Data were analyzed by 2-way ANOVA and JC-1 fluorescence relative ratio (JC-1 monomers fluorescence density to relative JC-1 dimers fluorescence density per single cell) is expressed as mean ± SD (n = 35–50), * p < 0.05, ** p < 0.01, *** p < 0.001. JC-1 fluorescence ratio of control cells was set as 1.
Figure 3. Antioxidant activities of the different polarity fractions of P. lenticonus/Chios mastiha’s contribute to the maintenance of the mitochondrial membrane potential in H2O2-treated SH-SY5Y cells. (A) Representative images of JC-1 staining (JC-1 dimers-red aggregates; JC-1 monomers-green fluorescence) and Hoechst-33342 staining (nuclear staining) in human neuroblastoma SH-SY5Y cells, pretreated or not with 10 μg/mL ap, 5 μg/mL mp and 10 μg/mL p P. lenticonus/Chios mastiha’s fractions for 24 h and then, subjected or not to 700 μΜ H2O2 for 6 h. Vehicle-treated, control cells were incubated with 1/1000 v/v DMSO. Representative images were taken with a Zeiss LSM-800 confocal microscope in 40× objective. (B) Quantification of green to red staining. Data were analyzed by 2-way ANOVA and JC-1 fluorescence relative ratio (JC-1 monomers fluorescence density to relative JC-1 dimers fluorescence density per single cell) is expressed as mean ± SD (n = 35–50), * p < 0.05, ** p < 0.01, *** p < 0.001. JC-1 fluorescence ratio of control cells was set as 1.
Molecules 30 00997 g003
Figure 4. Regulation of the GR, PPARα, and Bcl-2 protein levels by the three different polarity fractions (ap, mp, p) of P. lenticonus/Chios mastiha. SH-SY5Y cells were treated with 20 μg/mL and 10 μg/mL ap and p P. lenticonus/Chios mastiha’s fractions and 10 μg/mL and 5 μg/mL with mp fraction for 24 h and subsequently treated with 700 μΜ H2O2 for 6 h. Vehicle cells were incubated with 1/1000 v/v DMSO. Western blot analysis results were expressed as the ratios of the GR, PPARα, and Bcl-2 band intensity normalized against the respective band intensity of the β-actin. Relative protein levels in control cells were set to 1.
Figure 4. Regulation of the GR, PPARα, and Bcl-2 protein levels by the three different polarity fractions (ap, mp, p) of P. lenticonus/Chios mastiha. SH-SY5Y cells were treated with 20 μg/mL and 10 μg/mL ap and p P. lenticonus/Chios mastiha’s fractions and 10 μg/mL and 5 μg/mL with mp fraction for 24 h and subsequently treated with 700 μΜ H2O2 for 6 h. Vehicle cells were incubated with 1/1000 v/v DMSO. Western blot analysis results were expressed as the ratios of the GR, PPARα, and Bcl-2 band intensity normalized against the respective band intensity of the β-actin. Relative protein levels in control cells were set to 1.
Molecules 30 00997 g004
Table 1. Suspect compounds by HPLC-QTOF-MS/MS analysis in the P. lenticonus/Chios mastiha’s different polarity fractions (ap, mp, and p). Normalized concentrations (μg/L) and their relative enrichment are presented.
Table 1. Suspect compounds by HPLC-QTOF-MS/MS analysis in the P. lenticonus/Chios mastiha’s different polarity fractions (ap, mp, and p). Normalized concentrations (μg/L) and their relative enrichment are presented.
CompoundsQuantitative Measurement (μg/L)Relative Enrichment
Apolar, apMedium Polar, mpPolar, pApolar, apMedium Polar, mpPolar, p
Coniferaldehyde103.9-----
Palmitic acid9174.31768.31102.98.31.61.0
Stearic acid13,951.42437.41620.98.61.51.0
Myristic acid395.769.379.85.71.01.2
Arachidonic acid25.97.00.643.211.71.0
Pentadecanoic acid246.248.938.86.31.31.0
Betulinic acid2119.711,462.911,725.71.05.45.5
Euscaphic Acid40.4199.6482.01.04.911.9
Flavidin30.7265.9148.11.08.74.8
Luteolin glucoside0.42.22.31.05.55.8
α-Linolenic acid23.055.7103.61.02.44.5
Hesperidin4.96.05.31.01.21.1
Oleic acid2502.31443.11131.42.21.31.0
Linoleic acid267.3248.33201.11.01.3
Caprylic acid54.135.431.71.71.11.0
Nebraskanic acid47.550.065.81.01.11.4
Octyl formate122.254.740.63.01.31.0
6,7-dihydro-7-hydroxylinalool64.151.353.71.21.01.04
α-irone10.318.513.51.01.81.3
Oleanonic acid29,571.432,200.025,114.31.21.31.0
Resveratrol5.76.46.71.01.11.2
Isoliquiritigenin di-glucoside3.10.13.131.01.031.0
Methoxycinnamic acid10.318.513.51.01.81.3
Masticadecanoic acid29,571.432,200.025,114.31.21.31.0
Gamma linolenic acid23.055.7103.61.02.44.5
Oleanolic acid601.12920.02885.71.04.94.8
Ricinoleic acid23.055.7103.61.02.44.5
Crepenynic acid23.055.7103.61.02.44.5
Table 2. Non-target compounds in the different polarity P. lenticonus/Chios mastiha’s fractions as revealed by non-target analysis using the SCIEX Natural Products Library and their chemical classification.
Table 2. Non-target compounds in the different polarity P. lenticonus/Chios mastiha’s fractions as revealed by non-target analysis using the SCIEX Natural Products Library and their chemical classification.
Non-Target CompoundsNon-Target Compounds Chemical Classification
Ethyl 2-acetyl heptanoatestraight-chain fatty acid
Sesterstatinsesterterpenoid
Cyclohexanecarboxylic acidorganic compound
1,2-Hydroxylauric acidmedium-chain fatty acid
Dodecanedioic acidsaturated aliphatic dicarboxylic acid
Tisocalcitateorganic compound (vitamin D derivative)
Trivalerin(ester of valeric acid) straight-chain saturated fatty acid
Frangulin Banthraquinone
4,4′,6,6′-Tetra-tert-butyl-2,2′-biphenolaromatic hydrocarbon
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Georgantopoulos, A.; Kalousi, F.D.; Pollastro, F.; Tsialtas, I.; Kalogiouri, N.P.; Psarra, A.-M.G. Chemical Analysis and Antioxidant Activities of Resin Fractions from Pistacia lentiscus L. var. Chia in Neuroblastoma SH-SY5Y Cells. Molecules 2025, 30, 997. https://doi.org/10.3390/molecules30050997

AMA Style

Georgantopoulos A, Kalousi FD, Pollastro F, Tsialtas I, Kalogiouri NP, Psarra A-MG. Chemical Analysis and Antioxidant Activities of Resin Fractions from Pistacia lentiscus L. var. Chia in Neuroblastoma SH-SY5Y Cells. Molecules. 2025; 30(5):997. https://doi.org/10.3390/molecules30050997

Chicago/Turabian Style

Georgantopoulos, Achilleas, Foteini D. Kalousi, Federica Pollastro, Ioannis Tsialtas, Natasa P. Kalogiouri, and Anna-Maria G. Psarra. 2025. "Chemical Analysis and Antioxidant Activities of Resin Fractions from Pistacia lentiscus L. var. Chia in Neuroblastoma SH-SY5Y Cells" Molecules 30, no. 5: 997. https://doi.org/10.3390/molecules30050997

APA Style

Georgantopoulos, A., Kalousi, F. D., Pollastro, F., Tsialtas, I., Kalogiouri, N. P., & Psarra, A.-M. G. (2025). Chemical Analysis and Antioxidant Activities of Resin Fractions from Pistacia lentiscus L. var. Chia in Neuroblastoma SH-SY5Y Cells. Molecules, 30(5), 997. https://doi.org/10.3390/molecules30050997

Article Metrics

Back to TopTop