Next Article in Journal
Evaluation of Cotton (Gossypium hirsutum L.) Leaf Abscission Sensitivity Triggered by Thidiazuron through Membership Function Value
Next Article in Special Issue
Validation of Qualitative Broth Volatilization Checkerboard Method for Testing of Essential Oils: Dual-Column GC–FID/MS Analysis and In Vitro Combinatory Antimicrobial Effect of Origanum vulgare and Thymus vulgaris against Staphylococcus aureus in Liquid and Vapor Phases
Previous Article in Journal
Application of Exogenous Phytohormones at Silking Stage Improve Grain Quality under Post-Silking Drought Stress in Waxy Maize
Previous Article in Special Issue
Prostanthera (Lamiaceae) as a ‘Cradle of Incense’: Chemophenetics of Rare Essential Oils from Both New and Forgotten Australian ‘Mint Bush’ Species
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Effect of Essential Oils on the Release of TNF-α and CCL2 by LPS-Stimulated THP‑1 Cells

by
Maria Graça Miguel
1,*,
Carina Isabel da Silva
1,
Luana Farah
2,
Fernão Castro Braga
2 and
Ana Cristina Figueiredo
3
1
Mediterranean Institute for Agriculture, Environment and Development (MED), Departamento de Química e Farmácia, Faculdade de Ciências e Tecnologia, Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
2
Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, Pampulha, 31.270-901 Belo Horizonte, Brazil
3
Centro de Estudos do Ambiente e do Mar (CESAM Lisboa), Faculdade de Ciências da Universidade de Lisboa, Centro de Biotecnologia Vegetal (CBV), DBV, C2, Piso 1, Campo Grande, 1749-016 Lisboa, Portugal
*
Author to whom correspondence should be addressed.
Plants 2021, 10(1), 50; https://doi.org/10.3390/plants10010050
Submission received: 21 November 2020 / Revised: 14 December 2020 / Accepted: 16 December 2020 / Published: 28 December 2020

Abstract

:
Plants and their constituents have been used to treat diverse ailments since time immemorial. Many plants are used in diverse external and internal formulations (infusions, alcoholic extracts, essential oils (EOs), etc.) in the treatment of inflammation-associated diseases, such as those affecting the respiratory tract or causing gastrointestinal or joint problems, among others. To support the traditional uses of plant extracts, EOs have been assessed for their alleged anti-inflammatory properties. However, the effect of EOs on the release of cytokines and chemokines has been much less reported. Considering their traditional use and commercial relevance in Portugal and Angola, this study evaluated the effect of EOs on the in vitro inhibition of the cytokine tumor necrosis factor-α (TNF-α) and the chemokine (C-C motif) ligand 2 (CCL2) by lipopolysaccharide (LPS)-stimulated human acute monocytic leukemia cells (THP-1 cells). Twenty EOs extracted from eighteen species from seven families, namely from Amaranthaceae (Dysphania ambrosioides), Apiaceae (Foeniculum vulgare), Asteraceae (Brachylaena huillensis, Solidago virgaurea), Euphorbiaceae (Spirostachys africana), Lamiaceae (Lavandula luisieri, Mentha cervina, Origanum majorana, Satureja montana, Thymbra capitata, Thymus mastichina, Thymus vulgaris, Thymus zygis subsp. zygis), Myrtaceae (Eucalyptus globulus subsp. maidenii, Eucalyptus radiata, Eucalyptus viminalis) and Pinaceae (Pinus pinaster) were assayed for the release of CCL2 and TNF-α by LPS-stimulated THP-1 cells. B. huillensis, S. africana, S. montana, Th. mastichina and Th. vulgaris EOs showed toxicity to THP-1 cells, at the lowest concentration tested (10 μg/mL), using the tetrazolium dye assay. The most active EOs in reducing TNF-α release by LPS-stimulated THP-1 cells were those of T. capitata (51% inhibition at 20 μg/mL) and L. luisieri (15–23% inhibition at 30 μg/mL and 78–83% inhibition at 90 μg/mL). L. luisieri EO induced a concentration-dependent inhibition of CCL2 release by LPS‑stimulated THP-1 cells (23%, 54% and 82% inhibition at 10, 30 and 90 μg/mL, respectively). These EOs are potentially useful in the management of inflammatory diseases mediated by CCL2 and TNF‑α, such as atherosclerosis and arthritis.

Graphical Abstract

1. Introduction

Since ancient times, man has used the plant kingdom as a source for clothing, construction, fuel, food, spices and medicines, as well as for poisons. Nowadays, around half the pharmaceutical drugs used in developed countries, such as aspirin, are of plant origin [1]. Traditional medicine is still the main source of health care for 80% of the people in developing countries, where medicinal plants are commonly used for the treatment of several ailments, notably inflammatory diseases.
Acute inflammation is a short-term reaction which is essential for survival after an infection or a physical injury. On the other hand, chronic inflammation, promoted by social, environmental and lifestyle factors (diet, smoking, alcoholism, inactivity), may trigger diverse long-term illnesses such as cardiovascular disease, cancer, diabetes mellitus, chronic kidney disease, nonalcoholic fatty liver disease and autoimmune and neurodegenerative disorders [2]. These diseases and lifestyles are associated with atherosclerosis, the early detection of which is based on peripheral artery and carotid artery thickness. In a recent study of the epidemiological burden caused by carotid atherosclerosis, Song et al. [3] estimated that, in 2020, the prevalence of increased carotid intima-media thickness in people aged 30 to 79 years was about 28%, equivalent to approximately 1070 million cases worldwide.
In Portugal, recent studies showed that as many as 740,000 adults are affected by atherosclerosis [4]. Data on other inflammatory diseases in Portugal, such as rheumatic diseases [5], showed that women (64%) are more affected by rheumatic diseases, including rheumatoid arthritis, than men (47%). These diseases are underdiagnosed in Portugal and are responsible for disability and absenteeism at work, with the consequent individual, social and economic costs [5]. Much less is known about inflammatory diseases in other Portuguese-speaking countries. There are no studies on the prevalence of rheumatic diseases in Angola [6], although the occurrence of rheumatic fever, rheumatoid arthritis or systemic lupus erythematosus is known [7].
Acute inflammation can be treated by using oral nonsteroidal anti-inflammatory drugs (NSAIDs), despite being associated with adverse gastrointestinal and cardiovascular effects [8,9]. It would be desirable to identify natural plant products with anti-inflammatory properties but with fewer adverse effects. Despite the traditional application of plants, their effects have not always been proven by scientific evidence. On the other hand, scientific research sometimes provides evidence of biological activities for which the plants in question had never been traditionally used.
Plant extracts, such as essential oils (EOs), have been used in traditional medicine as anti-inflammatories, digestives, diuretics, expectorants and sedatives, along with other applications (Table 1). Nowadays, in addition to their use in aromatherapy, essential oils find application in cosmetics, cleaning products, fragrances, foods and beverages. Essential oils have been reported to show several biological properties, including antimicrobial, antioxidant, anti-inflammatory and anticancer properties, among others [10,11,12]. Particularly relevant is EOs’ anti-inflammatory activity, either by inhibiting several enzymes, such as oxygenases, nitric oxide synthases and peroxidases, or by inducing the release of pro-inflammatory cytokines, like interleukins and tumor necrosis factor-α (TNF‑α) [10].
Chemokines constitute a family of chemoattractant cytokines. These are small heparin-binding proteins involved in atherosclerosis by promoting directed migration of inflammatory cells. Chemokine (C-C motif) ligand 2 (CCL2), also known as monocyte chemoattractant protein-1 (MCP-1), has been detected in atherosclerotic lesions [34]. CCL2 is also a potent mediator of chronic inflammation, triggering, for instance, inflammation in rheumatoid arthritis [35]. In addition, inflammatory response is characterized by increased production of tumor necrosis factor-α (TNF-α) [35]. TNF-α, interleukin (IL)-1 and IL-6, secreted by macrophages, lymphocytes, natural killer cells and vascular smooth muscle cells, are considered pro-atherogenic cytokines [36]. Despite the reported anti-inflammatory potential of several EOs (Table 2), their effect on the release of CCL2 is much less reported than the release of TNF-α.
Essential oils are gaining commercial relevance in several countries, such as Portugal or Angola, as an additional source of income in a context of a more sustainable use of the local flora. Nevertheless, despite these essential oils being traded, national or internationally, for specific markets, it is ever more important for their added value to gather scientific support for their alleged biological properties. Given the traditional and commercial use of EOs for medicinal and cosmetic purposes and the knowledge of the ability, of their monoterpene and sesquiterpene constituents, to act as anti-inflammatories [56,57], the present work evaluated twenty EOs obtained from eighteen plant species collected in Portugal and Angola (Table 1) for their effect on the release of CCL2 (MCP‑1) and TNF-α by lipopolysaccharide (LPS)-stimulated THP-1 cells.

2. Material and Methods

2.1. Plant Material

Collective and/or individual samples, from cultivated and wild-growing medicinal and aromatic plants, were collected from mainland Portugal (Table 3). As a rule, the plant material was collected during the local producers’ harvesting season. For herbaceous species, this was usually at the flowering phase, whereas for trees, it was at landscaping time. If not immediately extracted, the plant material was stored at −20 °C until essential oil (EO) isolation. Dried aerial parts from commercially available products sold in local herbal shops were also analyzed, as well as the essential oils isolated from oleoresin, in the case of Pinus pinaster, and from the two species from Angola (Table 3). A total of twenty essential oils isolated from eighteen species from the Amaranthaceae, Apiaceae, Asteraceae, Euphorbiaceae, Lamiaceae, Myrtaceae and Pinaceae families were tested. A voucher specimen of each plant species, collected from the wild state condition, was deposited in the Herbarium of the Botanical Garden of Lisbon University, Lisbon, Portugal. For commercial plant material, a reference sample from each plant is retained at the CBV laboratory and is available upon request.

2.2. Extraction and Chemical Analysis of the Essential Oils

Essential oils were extracted by hydrodistillation for 3 h, using a Clevenger-type apparatus, according to the European Pharmacopoeia [59], and stored at −20 °C until analysis. The EOs were analyzed by gas chromatography (GC) for component quantification and gas chromatography coupled to mass spectrometry (GC-MS) for component identification.

2.2.1. Gas Chromatography (GC)

Gas chromatographic analyses were performed using a Perkin Elmer Clarus 400 gas chromatograph equipped with two flame ionization detectors (FIDs), a data handling system and a vaporizing injector port into which two columns of different polarities were installed: a DB-1 fused-silica column (polydimethylsiloxane, 30 m × 0.25 mm i.d., film thickness 0.25 µm; J & W Scientific Inc., Rancho Cordova, CA, USA) and a DB-17HT fused-silica column ((50% phenyl)-methylpolysiloxane, 30 m × 0.25 mm i.d., film thickness 0.15 µm; J & W Scientific Inc.). The oven temperature was programmed from 45 to 175 °C, at 3 °C/min, and subsequently at 15 °C/min up to 300 °C, and then held isothermal for 10 min; injector and detector temperatures were 280 °C and 300 °C, respectively; the carrier gas, hydrogen, was adjusted to a linear velocity of 30 cm/s. The samples were injected using a split sampling technique, ratio 1:50. The volume of injection was 0.1 µL of n-pentane-essential oil solution (1:1). The percentage composition of the volatiles was computed, by the normalization method from the GC peak areas, and calculated as the mean values of two injections, from each sample, without using the response factors.

2.2.2. Gas Chromatography-Mass Spectrometry (GC‑MS)

The GC‑MS unit consisted of a Perkin Elmer Clarus 600 gas chromatograph, equipped with a DB‑1 fused-silica column (30 m × 0.25 mm i.d., film thickness 0.25 µm; J & W Scientific, Inc.), and interfaced with a Perkin Elmer 600T mass spectrometer (software version 5.4.2.1617, Perkin Elmer, Shelton, CT, USA). Injector and oven temperatures were as above; transfer line temperature, 280 °C; ion source temperature, 220 °C; the carrier gas, helium, was adjusted to a linear velocity of 30 cm/s; split ratio, 1:40; ionization energy, 70 eV; scan range, 40–300 u; scan time, 1 s. The identity of the components was assigned by comparison of their retention indices, relative to n‑alkane indices and GC‑MS spectra from a lab‑made library, created with reference essential oils, laboratory-synthesized components, laboratory-isolated compounds and commercially available standards.

2.3. In Vitro Inhibition of TNF-α and CCL2

This assay was performed according to Campana et al. [60]. Briefly, THP-1 cells (ATCC TIB-202) were cultivated in RPMI 1640 medium supplemented with 0.05 mM 2-mercaptoethanol, 10% fetal bovine serum (FBS), 100 U/mL penicillin and 100 μg/mL gentamicin at 37°C in an atmosphere containing 5% CO2. The medium was renewed twice a week when the cell concentration reached 1.0 × 106 cells/mL. The cells were transferred to a 96-well microplate at a concentration of 100,000 cells per well and incubated for 18 h with RPMI supplemented with 1% FBS to initiate serum starvation, which was kept throughout the experiment.
The cells were pre-treated with EOs at three concentrations for 3 h. To determine each EO working concentration, the toxicity of the EOs on THP-1 cells was accessed by measuring cell viability using a 3‑(4,5-dimethylthiazol-2-yl)-2,5‑diphenyltetrazolium bromide (MTT) method and untreated cells as the reference for viability [61]. EOs were considered nontoxic for the THP-1 cell line, and adequate for further analysis, when cell viability was higher than 90%. The EO concentrations ranged from 3 µg/mL to 90 µg/mL (Dysphania ambrosioides, Eucalyptus globulus, E. radiata, E. viminalis, Foeniculum vulgare, Lavandula stoechas, Mentha cervina, Origanum majorana, Pinus pinaster, Solidago virgaurea, Thymus mastichina, Th. pulegioides (Thymus abbreviated to Th., to avoid confusion with T. from Thymbra), Th. vulgaris), from 3 µg/mL to 30 µg/mL (Brachylaena huillensis, Satureja montana, Spirostachys africanus, Th. zygis) and from 5 µg/mL to 90 µg/mL (Thymbra capitata) (Table 4).
Lipopolysaccharide (LPS) from Escherichia coli 0111:B4 (Sigma), added at 200 ng/mL, was employed as the inflammatory stimulus. The plate was incubated at 37°C overnight. After this period, the plate was centrifuged (1800 g, 5 min, 16°C), the supernatant collected and TNF-α release measured using the cytokine-specific sandwich quantitative ELISA according to the manufacturer’s instructions (TNF-α duo set, DY210, R&D Systems, Minneapolis, MN, USA). CCL2 release was measured using the cytokine-specific sandwich quantitative ELISA according to the manufacturer’s instructions (Human CCL2/MCP-1 duo set, DY279, R&D Systems, Minneapolis, MN, USA). Dexamethasone was employed as positive control (0.3 μM). The statistical significance of differences was calculated employing the software GraphPad Prism, version 5.0 (GraphPad Software Inc., San Diego, CA, USA), using ordinary one-way ANOVA/Newman–Keuls multiple comparison test. All the experiments were performed in triplicate.

3. Results and Discussion

3.1. Chemical Composition of the Essential Oils

All essential oils were fully chemically characterized. Table 3 reports only their main constituents (≥10%), since, in some cases, duly marked in Table 3, the detailed composition was previously reported, or their composition was overall very similar to data from prior studies. In the case of EOs from Angola, the complexity of the EOs still requires additional characterization for the full identification of some minor components.
Although commercialized as Lavandula stoechas L., the presence of necrodane derivatives, such as 5-methylene-2,3,4,4-tetramethylcyclopent-2-enone (18%) in the analyzed essential oil, undoubtedly indicated that the butterfly lavender tested was L. luisieri and not L. stoechas (Table 3). The chemical composition of L. stoechas essential oil is characterized by large variations in fenchone, camphor and 1,8‑cineole amounts, whereas necrodane derivatives are characteristic of L. luisieri [23,24,62]. Even though some variations in their contents were observed, the remaining essential oil compositions were in accordance with previous studies carried out with Foeniculum vulgare [15,63], Mentha cervina ([25] and references therein), Origanum majorana [64], Satureja montana [58,65], Thymbra capitata, Thymus mastichina, Th. pulegioides, Th. vulgaris and Th. zygis subsp. zygis [28,64,65,66], Eucalyptus species [67,68] and Pinus pinaster [33,69].
Although a few studies evaluated the EO composition from Brachylaena huillensis aerial parts, only three studies reported the essential oil composition from the wood or saw powder of this species [16,17,70]. Although no detailed composition has been reported, α‑amorphene was the dominant constituent in the studies of Klein and Schmidt [70] and of Maitai et al. [16] (17% and 15%, respectively), whereas β-caryophyllene (19%) was the major constituent described by Oliva et al. [17]. In the present study, α‑amorphene was the second main component, together with gleenol (both 6%), whereas β-caryophyllene was found only in trace amounts. Baarschers et al. [20] reported the isolation of diterpenes from Spirostachys africana wood, but no previous studies addressed the EO composition from the wood.

3.2. In Vitro Inhibition of TNF‑α Release by LPS-Stimulated THP-1 Cells

The potential anti-inflammatory activity of essential oils (EOs) was investigated by measuring TNF‑α release by lipopolysaccharide (LPS)-stimulated THP-1 cells by employing an immunoassay. The toxicity of the EOs on THP-1 cells was accessed to determine the adequate EO working concentrations. When the cell viability of THP-1 cells was higher than 90%, samples were considered non-cytotoxic and adequate for further analysis. According to the availability of EO, at least three concentrations were checked for each essential oil (Table 4). Data in Table 4 also include information on EOs which were not assessed further due to being toxic (ND) to differentiate them from those that showed no inhibition (NI).
The EOs of T. capitata, L. luisieri, F. vulgare and D. ambrosioides significantly reduced TNF-α release by LPS-stimulated THP-1 cells, in comparison to the control cells. From these four EOs, those of T. capitata and L. luisieri were the most effective. T. capitata EO showed an inhibition percentage of TNF-α release of 51 ± 7% at 20 μg/mL, whereas that of L. luisieri EO was 23 ± 1% at 30 μg/mL and 83 ± 8% at 90 μg/mL (Table 4). These inhibition percentages were higher than those of D. ambrosioides EO (49 ± 2% at 90 μg/mL), or F. vulgare EO (22 ± 2% at 90 μg/mL) (Table 4).
The potential anti-inflammatory activity of L. luisieri, F. vulgare and T. capitata EOs has been previously reported using different in vitro and in vivo models (Table 2), but as far as we know, the anti-inflammatory potential of D. ambrosioides EO has not been addressed to date. Recently, the anti-inflammatory activity of alcoholic or hydroalcoholic extracts of D. ambrosioides was reported as showing the ability to reduce interleukin 6 (IL-6), myeloperoxidase (MPO), nitric oxide (NO) and adenosine-deaminase (ADA) activity and TNF-α and, therefore, they are potentially useful in wound healing and in the treatment of arthritic processes [13,71]. The oxygen-containing monoterpene ascaridole was identified as a constituent of D. ambrosioides ethanolic extract by Grassi et al. [13], a compound also identified in the essential oils evaluated in the present work (Table 3).
Despite carvacrol being the main compound of T. capitata EO (Table 3), this phenol-like oxygen-containing monoterpene may not be the only compound accountable for T. capitata EO activity. Indeed, other carvacrol-rich EOs, such as those of S. montana and Th. zygis (Table 3), were not able to reduce TNF‑α release. The presence of antagonists in these EOs can also not be ignored. Moreover, it is relevant to highlight the important role of the minor compounds and/or some of the compounds’ enantiomeric ratio in the overall activity of EOs. Often overlooked, these factors can contribute to synergistic or antagonistic actions determining differences in the EOs’ activities [56,72]. These results make it difficult to predict the effect of different species’ essential oils that share the same major component for TNF-α release.
Th. pulegioides and, particularly, Th. vulgaris EOs, with thymol, an isomer of carvacrol as the main component (Table 3), were toxic for THP-1 cells, even at lower concentrations (Table 4). Th. vulgaris EO has been reported to show anti-inflammatory activity, including the capacity of reducing TNF-α release, this activity being related solely to the higher carvacrol content [37,49,50,52]. On the other hand, Th. zygis and Th. vulgaris EOs, which have thymol as the main constituent, have been reported to decrease TNF-α secretion by human macrophages derived from THP‑1 monocytes and activated by oxidized (ox)‑LDLs [51]. Dexamethasone at 0.3 μM had > 90% inhibition.

3.3. In Vitro Inhibition of CCL2 Release by LPS-Stimulated THP-1 Cells

Inflammatory changes in arterial lesions are characterized by the recruitment and activation of monocytes/macrophages, which are regulated by CCL2. This chemoattractant cytokine has been shown to play a vital role in the initiation and progression of arteriosclerotic lesions in experimental animals [73]. The effect of the essential oils on CCL2 release by LPS-stimulated THP‑1 cells was also evaluated.
Of the four essential oils with the ability to inhibit CCL2 release, only L. luisieri EO had remarkable activity, with inhibition percentages of 23 ± 1%, 54 ± 3% and 82 ± 12% at 10, 30 and 90 µg/mL, respectively (Table 4). The major compound of L. luisieri EO, 5-methylene-2,3,4,4-tetramethylcyclopent-2-enone, a necrodane derivative, may have contributed to this activity, along with 1,8-cineole. Nevertheless, the absence of the activity of other EOs in which 1,8-cineole was also present, even in a much higher percentage, such as Th. mastichina or Eucalyptus EOs (Table 3), may suggest that 5‑methylene-2,3,4,4-tetramethylcyclopent-2-enone plays an important role in the inhibition of both TNF‑α and CCL2 release (Table 4). The inhibitory activities elicited by D. ambrosioides, S. virgaurea or B. huillensis EOs were much lower (Table 4). Dexamethasone at 0.3 μM had > 90% inhibition.
Reports regarding the action of essential oils and/or their main components on the production of CCL2 are scarce. Limonene isolated from Citrus junos EO was able to inhibit CCL2 production on diesel exhaust particle (DEP)-stimulated human eosinophilic leukemia HL-60 clone 15 cells [74]. Artemisia argyi EO, mainly constituted by 1,8-cineole (33%), camphor (17%), (‑)‑borneol (13%) and α‑thujone (13%), reduced TNF-α, IL-6, IFN-β and CCL2 in LPS-induced RAW264.7 macrophages [75]. Xiao Qing Long Tang essential oil was able to suppress CCL2, IL-1β, IL-6, IL-10 and TNF-α expression and production by LPS-stimulated RAW264.7 cells [76]. In addition, Park et al. [77] also reported that (‑)‑linalool was able to inhibit microglial migration induced by CCL2, a chemokine released by oxygen-glucose deprivation/reoxygenation (OGD/R) in cortical cells from 17-day-old embryos of Sprague-Dawley rats.
Along with L. luisieri EO, the ascaridole- and iso-ascaridole-rich D. ambrosioides EO was also able to reduce CCL2 release by LPS-stimulated THP-1 cells, as observed for TNF-α, although in a weaker manner (Table 4). The absence of these compounds in the remaining non-active EOs may suggest that these volatile compounds have an important role in the suppression of some inflammatory processes in which TNF-α and CCL2 are involved. Despite the traditional application of D. ambrosioides as a vermifuge and against vomiting [14], this is the first report on the effect of its essential oil on the release of the pro-inflammatory cytokine TNF-α and the chemokine CCL2. For this reason, this EO and its main component ascaridole, and/or its isomers, should be further investigated to explore their anti-inflammatory activity.

4. Conclusions

Inflammatory disorders are usually treated with steroidal anti-inflammatory drugs (SAIDs) or non-SAIDs (NSAIDs). Nevertheless, because these drugs present multiple negative side effects, it is important to assess and validate the use of other potential anti-inflammatory agents, namely, essential oils. Moreover, some of these essential oils are by-products from landscaping activities or other industries, thus constituting an added value to countries’ local flora.
This study suggests that T. capitata and L. luisieri EOs, mainly constituted by carvacrol and 5‑methylene-2,3,4,4-tetramethylcyclopent-2-enone and 1,8-cineole, respectively, were the most effective to inhibit TNF‑α release by LPS-stimulated THP-1 cells, whereas only L. luisieri EO had the ability to inhibit CCL2 release by LPS-stimulated THP-1 cells.
EOs’ chemical complexity and variability (existence of chemotypes and/or the enantiomeric ratio of some components), their hydrophobicity and, sometimes, their scarcity, have been considered some of the limitations to their use in diverse formulations. Nevertheless, EOs are Generally Regarded as Safe (GRAS), and the knowledge on their biological properties should be further explored, in solo formulations and in combination therapies, as potential anti-inflammatory agents. This approach would contribute to the goal of decreasing the use of SAIDs and, therefore, preventing or diminishing these drugs’ adverse side effects.

Author Contributions

M.G.M., L.F., C.I.d.S., F.C.B. and A.C.F.: Conceptualization, methodology, formal analysis and investigation, M.G.M., F.C.B., L.F., C.I.d.S. and A.C.F.: writing—original draft preparation, review and editing; M.G.M., F.C.B. and A.C.F.: funding acquisition. All authors have read and agreed to the published version of the manuscript.

Funding

The European Commission under the Seventh Framework Programme (FP7) of the European Union, Marie Curie International Research Staff Exchange Scheme (MC-IRSES). Project PEOPLE MC-IRSES, FP7-PEOPLE-2011-IRSES, PIRSES-GA-2011-295251 Fundação para a Ciência e Tecnologia (FCT/MCTES), FEDER, PT2020 PA, Compete 2020. Projects MED UIDB/05183/2020 and CESAM UIDP/50017/2020 + UIDB/50017/2020.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

All authors are indebted to Wolfgang Kreis, from the Friedrich Alexander Universität Erlangen, Nürnberg, Germany, as the main supervisor of the international project under which part of this work was performed (DIGITALIS: The genus Digitalis: Molecular taxonomy, preservation, active constituents and therapeutic applications. Project PEOPLE MC-IRSES, FP7-PEOPLE-2011-IRSES, PIRSES-GA-2011-295251, funded by the European Commission under the Seventh Framework Programme (FP7) of the European Union, Marie Curie International Research Staff Exchange Scheme (MC-IRSES)). Maria Graça Miguel and Carina Isabel da Silva are grateful for the grants under research contract PEOPLE MC‑IRSES, FP7-PEOPLE-2011-IRSES, PIRSES-GA-2011-295251. The authors acknowledge the Instituto da Conservação da Natureza e das Florestas (ICNF) and particularly Eng João Sanches (Mata Experimental do Escaroupim (MEE) from the Centro Nacional de Sementes Florestais (CENASEF)) for kindly allowing the sampling of all studied species from MEE. This study was partially funded by Fundação para a Ciência e Tecnologia (FCT/MCTES), under MED UIDB/05183/2020, and CESAM UIDP/50017/2020 + UIDB/50017/2020, FEDER, PT2020 PA and Compete 2020.

Convention on Biodiversity

The authors obtained, and acknowledge, the appropriate authority to access plant samples, other than commercially available plant material, essential oils or oleoresin, used for research as required under the framework of the United Nations Convention on Biodiversity.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

CCL2Chemokine (C-C motif) ligand 2
COX-2Cyclooxygenase-2
DFDry, flowering phase aerial parts
DLDry leaves
DVDry, vegetative phase aerial parts
EOsEssential oils
FBSFetal bovine serum
FFFresh, flowering phase aerial parts
FLFresh leaves from fruiting phase
IC50Half-maximal inhibitory concentration
LDLLow-density lipoprotein
LPSLipopolysaccharide
MCP-1Monocyte chemoattractant protein-1
MEEMata Experimental do Escaroupim
MTT3-(4,5-Dimethylthiazol-2-yl)-2,5 diphenyltetrazolium bromide
THP-1Human acute monocytic leukemia cell line
TNF-αTumor necrosis factor-α

References

  1. Vane, J.R.; Botting, R.M. The mechanism of action of aspirin. Thromb. Res. 2003, 110, 255–258. [Google Scholar] [CrossRef]
  2. Furman, D.; Campisi, J.; Verdin, E.; Carrera-Bastos, P.; Targ, S.; Franceschi, C.; Ferrucci, L.; Gilroy, D.W.; Fasano, A.; Miller, G.W.; et al. Chronic inflammation in the etiology of disease across the life span. Nat. Med. 2019, 25, 1822–1832. [Google Scholar] [CrossRef] [PubMed]
  3. Song, P.; Fang, Z.; Wang, H.; Cai, Y.; Rahimi, K.; Zhu, Y.; Fowkes, F.G.R.; Fowkes, F.J.I.; Rudan, I. Global and regional prevalence, burden, and risk factors for carotid atherosclerosis: A systematic review, meta-analysis, and modelling study. Lancet Glob. Health 2020, 8, e721–e729. [Google Scholar] [CrossRef]
  4. CCAP. Custo e Carga da Aterosclerose em-Portugal.Centro de Estudo de Medicina Baseada na Evidência da FML, Centro de Estudos Aplicados da Universidade Católica e Sociedade Portuguesa de Aterosclerose. 2019. Available online: https://spaterosclerose.org/highlights-de-2018/item/274-custo-e-carga-da-aterosclerose-em-portugal.html (accessed on 13 October 2020). (In Portuguese)
  5. Branco, J.C.; Faustino, A.; Carvalho, B.; Araújo, F.; Canhão, H.; Brito, I.; da Silva, J.A.P.; Costa, J.A.; Costa, L.; Maurício, L.; et al. Rede Nacional de especialidade hospitalar e de referenciação de reumatologia. 2015. Available online: https://www.sns.gov.pt/wp-content/uploads/2016/05/rede-referencia%C3%A7%C3%A3o-hospitalar-reumatologia.pdf (accessed on 13 October 2020). (In Portuguese)
  6. Usenbo, A.; Kramer, V.; Young, T.; Musekiwa, A. Prevalence of arthritis in Africa: A systematic review and meta-analysis. PLoS ONE 2015, 10, e0133858. [Google Scholar] [CrossRef]
  7. SESPA. Secretário de Estado para a Saúde Pública de Angola. 2018. Available online: http://jornaldeangola.sapo.ao/sociedade/angola_so_tem_oito_medicos_formados_em_reumatologia (accessed on 13 October 2020). (In Portuguese).
  8. Boudreault, J.; Desmeules, F.; Roy, J.-S.; Dionne, C.; Frémont, P.; MacDermid, J.C. The efficacy of oral non-steroidal anti-inflammatory drugs for rotator cuff tendinopathy: A systematic review and meta-analysis. J. Rehabil. Med. 2014, 46, 294–306. [Google Scholar] [CrossRef] [Green Version]
  9. Wallace, J.L. Eicosanoids in the gastrointestinal tract. Br. J. Pharmacol. 2019, 176, 1000–1008. [Google Scholar] [CrossRef] [Green Version]
  10. Miguel, M.G. Antioxidant and anti-inflammatory activities of essential oils: A short review. Molecules 2010, 15, 9252–9287. [Google Scholar] [CrossRef] [Green Version]
  11. Dandlen, S.; Lima, A.; Mendes, M.; Miguel, M.; Faleiro, M.L.; Sousa, M.; LG, P.; JG, B.; Figueiredo, A. Antimicrobial activity, cytotoxicity and intracellular growth inhibition of Portuguese Thymus essential oils. Rev. Bras. Farmacog. 2011, 21, 1012–1024. [Google Scholar] [CrossRef] [Green Version]
  12. De Lima, V.T.; Vieira, M.C.; Kassuya, C.A.L.; Cardoso, C.A.L.; Alves, J.M.; Foglio, M.A.; de Carvalho, J.E.; Formagio, A.S.N. Chemical composition and free radical-scavenging, anticancer and anti-inflammatory activities of the essential oil from Ocimum kilimandscharicum. Phytomedicine 2014, 21, 1298–1302. [Google Scholar] [CrossRef]
  13. Grassi, L.T.; Malheiros, A.; Meyre-Silva, C.; Buss, Z.S.; Monguilhott, E.D.; Fröde, T.S.; da Silva, K.A.B.S.; de Souza, M.M. From popular use to pharmacological validation: A study of the anti-inflammatory, anti-nocicpetive and healing effects of Chenopodium ambrosioides extract. J. Ethnopharmacol. 2013, 145, 127–138. [Google Scholar] [CrossRef] [Green Version]
  14. Alonso-Castro, A.J.; Domínguez, F.; Ruiz-Padilla, A.J.; Campos-Xolalpa, N.; Zapata-Morales, J.R.; Carranza-Alvarez, C.; Maldonado-Miranda, J.J. Medicinal plants from North and Central America and the Caribbean considered toxic for humans: The other side of the coin. Evid. Based Complement. Alternat. Med. 2017, 2017, 9439868. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Miguel, M.G.; Cruz, C.; Faleiro, L.; Simões, M.T.F.; Figueiredo, A.C.; Barroso, J.G.; Pedro, L.G. Foeniculum vulgare essential oils: Chemical composition, antioxidant and antimicrobial activities. Nat. Prod. Commun. 2010, 5, 319–328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Maitai, C.K.; Talalaj, S.; Talalaj, D. Aromatic Plants of East Africa; Department of Pharmacy College of Health Sciences, University of Nairobi: Nairobi, Kenya, 1983. [Google Scholar]
  17. Oliva, M.M.; Demo, M.S.; Malele, R.S.; Mutayabarwa, C.K.; Mwangi, J.W.; Thoithi, G.N.; Kibwage, I.O.; Faillaci, S.M.; Scrivanti, R.L.; Lopez, A.G.; et al. Essential oil of Brachylaena hutchinsii Huthch from Tanzania: Antimicrobial activity and composition. East. Central Afr. J. Pharm. Sci. 2003, 6, 61–63. [Google Scholar]
  18. Mrema, J.P. Conservation of Brachylaena huillensis O.Hoffm (Asteraceae) in Dindili Forest Reserve, Morogoro, Tanzania. Ph.D. Thesis, Addis Ababa University, Addis Ababa, Ethiopia, 2006. [Google Scholar]
  19. Minciullo, P.L.; Calapai, G.; Miroddi, M.; Mannucci, C.; Chinou, I.; Gangemi, S.; Schmidt, R.J. Contact dermatitis as an adverse reaction to some topically used European herbal medicinal products—Part 4: Solidago virgaurea—Vitis vinifera. Contact Dermat. 2017, 77, 67–87. [Google Scholar] [CrossRef]
  20. Baarschers, W.H.; Horn, D.H.; Johnson, L.R.F. The structure of some diterpenes from tambooti wood, Spirostachys africana Sond. J. Chem. Soc. 1962, 10, 4046–4055. [Google Scholar] [CrossRef]
  21. Mathabe, M.C.; Hussein, A.A.; Nikolova, R.V.; Basson, A.E.; Meyer, J.J.M.; Lall, N. Antibacterial activities and cytotoxicity of terpenoids isolated from Spirostachys africana. J. Ethnopharmacol. 2008, 116, 194–197. [Google Scholar] [CrossRef]
  22. Mukandiwa, L.; McGaw, L.J.; Eloff, J.N.; Naidoo, V. Extracts of four plant species used traditionally to treat myiasis influence pupation rate, pupal mass and adult blow fly emergence of Lucilia cuprina and Chrysomya marginalis (Diptera: Calliphoridae). J. Ethnopharmacol. 2012, 143, 812–818. [Google Scholar] [CrossRef]
  23. Zuzarte, M.; Gonçalves, M.J.; Cruz, M.T.; Cavaleiro, C.; Canhoto, J.; Vaz, S.; Pinto, E.; Salgueiro, L. Lavandula luisieri essential oil as a source of antifungal drugs. Food Chem. 2012, 135, 1505–1510. [Google Scholar] [CrossRef]
  24. Figueiredo, A.C.; Pedro, L.G.; Barroso, J.G.; Trindade, H.; Sanches, J.; Oliveira, C.; Correia, M. Lavandula luisieri (Rozeira) Rivas-Martínez e Lavandula pedunculata (Mill.) Cav. Agrotec.
  25. Rodrigues, L.; Duarte, A.; Figueiredo, A.C.; Brito, L.; Teixeira, G.; Moldão, M.; Monteiro, A. Chemical composition and antibacterial activity of the essential oils from the medicinal plant Mentha cervina L. grown in Portugal. Med. Chem. Res. 2012, 21, 3485–3490. [Google Scholar] [CrossRef]
  26. Costa, D.C.; Costa, H.S.; Albuquerque, T.G.; Ramos, F.; Castilho, M.C.; Sanches-Silva, A. Advances in phenolic compounds analysis of aromatic plants and their potential applications. Trends Food Sci. Technol. 2015, 45, 336–354. [Google Scholar] [CrossRef]
  27. Mastelić, J.; Jerković, I. Gas chromatography-mass spectrometry analysis of free and glycoconjugated aroma compounds of seasonally collected Satureja montana L. Food Chem. 2003, 80, 135–140. [Google Scholar] [CrossRef]
  28. Figueiredo, A.C.; Barroso, J.G.; Pedro, L.G.; Salgueiro, L.; Miguel, M.G.; Faleiro, M.L. Portuguese Thymbra and Thymus species volatiles: Chemical composition and biological activities. Cur. Pharm. Des. 2008, 14, 3120–3140. [Google Scholar] [CrossRef] [PubMed]
  29. Fernandes, A.S.F.; Barros, L.; Carvalho, A.M.; Ferreira, I.C.F.R. Lipophilic and hydrophilic antioxidants, lipid peroxidation, inhibition and radical scavenging activity of two Lamiaceae food plants. Eur. J. Lip. Sci. Technol. 2010, 112, 1115–1121. [Google Scholar] [CrossRef]
  30. Figueiredo, A.C.; Pedro, G.; Barroso, J.G.; Sanches, J.; Correia, M. Óleos essenciais de espécies de Eucalyptus. Agrotec 2013, 8, 96–100. [Google Scholar]
  31. Mahumane, G.D.; van Vuuren, S.F.; Kamatou, G.; Sandasi, M.; Viljoen, A.M. Chemical composition and antimicrobial activity of Eucalyptus radiata leaf essential oil, sampled over a year. J. Essent. Oil Res. 2016, 28, 475–488. [Google Scholar] [CrossRef]
  32. Tolmacheva, A.A.; Rogozhin, E.A.; Deryabin, D.G. Antibacterial and quorum sensing regulatory activities of some traditional Eastern-European medicinal plants. Acta Pharm. 2014, 64, 173–186. [Google Scholar] [CrossRef] [Green Version]
  33. Figueiredo, A.C.; Pedro, L.G.; Barroso, J.G.; Trindade, H.; Sanches, J.; Oliveira, C.; Correia, M. Pinus pinaster Aiton e Pinus pinea L. Agrotec 2014, 12, 23–27. [Google Scholar]
  34. Takeya, M.; Yoshimura, T.; Leonard, E.J.; Takahashi, K. Detection of monocyte chemoattractant protein-1 in human atherosclerotic lesions by an anti-monocyte chemoattractant protein-1 monoclonal antibody. Hum. Pathol. 1993, 24, 534–539. [Google Scholar] [CrossRef]
  35. Bakheet, S.A.; Alrwashied, B.S.; Ansari, M.A.; Nadeem, A.; Attia, S.M.; Alanazi, M.M.; Aldossari, A.A.; Assiri, M.A.; Mahmood, H.M.; Al-Mazroua, H.A.; et al. CXC chemokine receptor 3 antagonist AMG487 shows potent anti-arthritic effects on collagen-induced arthritis by modifying B cell inflammatory profile. Immunol. Lett. 2020, 225, 74–81. [Google Scholar] [CrossRef]
  36. Tousoulis, D.; Oikonomou, E.; Economou, E.K.; Crea, F.; Kaski, J.C. Inflammatory cytokines in atherosclecrosis: Current therapeutic approaches. Eur. Heart J. 2016, 37, 1723–1735. [Google Scholar] [CrossRef] [Green Version]
  37. Aazza, S.; Lyoussi, B.; Megías, C.; Cortés-Giraldo, I.; Vioque, J.; Figueiredo, A.C.; Miguel, M.G. Anti-oxidant, anti-inflammatory and anti-proliferative activities of Moroccan commercial essential oils. Nat. Prod. Commun. 2014, 9, 587–594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Rezayat, S.M.; Dehpour, A.-R.; Motamed, S.M.; Yazdanparast, M.; Chamanara, M.; Rashidian, A.; Sahebgharani, M. Foeniculum vulgare essential oil ameliorates acetic-induced colitis in rats through the inhibition of NF-kB pathway. Inflammopharmacology 2018, 26, 851–859. [Google Scholar] [CrossRef] [PubMed]
  39. Arantes, S.; Candeias, F.; Lopes, O.; Lima, M.; Pereira, M.; Tinoco, T.; Cruz-Morais, J.; Martins, M.R. Pharmacological and toxicological studies of essential oil of Lavandula stoechas subsp. luisieri. Planta Med. 2016, 82, 1266–1273. [Google Scholar] [CrossRef] [PubMed]
  40. Rufino, A.T.; Ribeiro, M.; Sousa, C.; Judas, F.; Salgueiro, L.; Cavaleiro, C.; Mendes, A.F. Evaluation of the anti-inflammatory, anti-catabolic and pro-anabolic effects of E-caryophyllene, myrcene and limonene in a cell model of osteoarthritis. Eur. J. Pharmacol. 2015, 750, 141–150. [Google Scholar] [CrossRef]
  41. Rufino, A.T.; Ferreira, I.; Judas, F.; Salgueiro, L.; Lopes, M.C.; Cavaleiro, C.; Mendes, A.F. Differential effects of the essential oils of Lavandula luisieri and Eryngium duriaei subsp. juresianum in cell models of two chronic inflammatory diseases. Pharm. Biol. 2015, 53, 1220–1230. [Google Scholar] [CrossRef] [Green Version]
  42. Arranz, E.; Jaime, L.; López de las Hazas, M.C.; Reglero, G.; Santoyo, S. Supercritical fluid extraction as an alternative process to obtain essential oils with anti-inflammatory properties from marjoram and sweet basil. Ind. Crops Prod. 2015, 67, 121–129. [Google Scholar] [CrossRef]
  43. Aazza, S.; El-Guendouz, S.; Miguel, M.G.; Antunes, M.D.; Faleiro, M.L.; Correia, A.I.; Figueiredo, A.C. Antioxidant, anti-inflammatory and anti-hyperglycamic activities of essential oils from Thymbra capitata, Thymus albicans, Thymus caespititius, Thymus carnosus, Thymus lotocephalus and Thymus mastichina from Portugal. Nat. Prod. Commun. 2016, 11, 1029–1038. [Google Scholar] [CrossRef] [Green Version]
  44. Carrasco, A.; Perez, E.; Cutillas, A.-B.; Martinez-Gutierrez, R.; Tomas, V.; Tudela, J. Origanum vulgare and Thymbra capitata essential oils from Spain: Determination of aromatic profile and bioactivities. Nat. Prod. Commun. 2016, 11, 113–120. [Google Scholar] [CrossRef]
  45. Wei, A.; Shibamoto, T. Antioxidant/lipoxygenase inhibitory activities and chemical compositions of selected essential oils. J. Agric. Food Chem. 2010, 58, 7218–7225. [Google Scholar] [CrossRef]
  46. Tsai, M.-L.; Lin, C.-C.; Lin, W.-C.; Yang, C.-H. Antimicrobial, antioxidant, and anti-inflammatory activities of essential oils from five selected herbs. Biosci. Biotech. Bioch. 2011, 75, 1977–1983. [Google Scholar] [CrossRef] [Green Version]
  47. Abdelli, W.; Bahri, F.; Romane, A.; Höferl, M.; Wanner, J.; Schmidt, E.; Jirovetz, L. Chemical composition and anti-inflammatory activity of Algerian Thymus vulgaris essential oil. Nat. Prod. Commun. 2017, 12, 611–614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Fachini-Queiroz, F.C.; Kummer, R.; Estevão-Silva, C.F.; Carvalho, N.D.B.; Cunha, J.M.; Grespan, R.; Bersani-Amado, C.A.; Cuman, R.K.N. Effects of thymol and carvacrol, constituents of Thymus vulgaris L. essential oil, on the inflammatory response. Evid. Based Complement. Alternat. Med. 2012, 2012, 657026. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Hotta, M.; Nakata, R.; Katsukawa, M.; Hori, K.; Takahashi, S.; Inoue, H. Carvacrol, a component of thyme oil, activates PPARα and Gamma and suppresses COX-2 expression. J. Lip. Res. 2010, 51, 132–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Bukovská, A.; Čikoš, Š.; Juhás, Š.; Il’Ková, G.; Rehák, P.; Koppel, J. Effects of a combination of thyme and oregano essential oils on TNBS-induced colitis in mice. Mediat. Inflamm. 2007, 23296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Ocaña, A.; Reglero, G. Effects of thyme extract oils (from Thymus vulgaris, Thymus zygis and Thymus hyemalis) on cytokine production and gene expression of oxLDL-stimulated THP-1 macrophages. J. Obes. 2012, 2012, 104706. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Juhás, Š.; Bujňáková, D.; Rehák, P.; Čikoš, Š.; Veselá, J.; Il’Ková, G.; Koppel, J. Anti-inflammatory effects of thyme essential oil in mice. Acta Vet. Brno 2008, 77, 327–334. [Google Scholar] [CrossRef] [Green Version]
  53. Cutillas, A.-B.; Carrasco, A.; Martinez-Gutierrez, R.; Tomas, V.; Tudela, J. Thyme essential oils from Spain: Aromatic profile ascertained by GC-MS, and their antioxidant, anti-lipoxygenase and antimicrobial activities. J. Food Drug Anal. 2018, 26, 529–544. [Google Scholar] [CrossRef] [Green Version]
  54. Tümen, I.; Guragac, F.T.; Keles, H.; Reunanen, M.; Kupeli-Akkol, E. Characterization and wound repair potential of essential oil Eucalyptus globulus Labill. Fresenius Environ. Bull. 2017, 26, 6390–6399. [Google Scholar]
  55. Tümen, I.; Akkol, E.K.; Taştan, H.; Süntar, I.; Kurtca, M. Research on the antioxidant, wound healing, and anti-inflammatory activities and the phytochemical composition of maritime pine (Pinus pinaster Ait). J. Ethnopharmacol. 2018, 211, 235–246. [Google Scholar] [CrossRef]
  56. Sá, R.C.S.; Andrade, L.N.; de Sousa, D.P. A review on anti-inflammatory activity of monoterpenes. Molecules 2013, 18, 1227–1254. [Google Scholar]
  57. Sá, R.C.S.; Andrade, L.N.; de Sousa, D.P. Sesquiterpenes from essential oils and anti-inflammatory activity. Nat. Prod. Commun. 2015, 10, 1767–1774. [Google Scholar]
  58. Faria, J.M.S.; Sena, I.; Ribeiro, B.; Rodrigues, A.M.; Maleita, C.M.N.; Abrantes, I.; Bennett, R.N.; Mota, M.; Figueiredo, A.C. First report on Meloidogyne chitwoodi hatching inhibition activity of essential oils and essential oils fractions. J. Pest. Sci. 2016, 89, 207–217. [Google Scholar] [CrossRef]
  59. Council of Europe (COE). European Directorate for the Quality of Medicines. European Pharmacopoeia, 7th ed.; COE: Strasbourg, France, 2007.
  60. Campana, P.R.V.; Mansur, D.S.; Gusman, G.S.; Ferreira, D.; Teixeira, M.M.; Braga, F.C. Anti-TNF-α activity of Brazilian medicinal plants and compounds from Ouratea semiserrata. Phytother. Res. 2015, 29, 1509–1515. [Google Scholar] [CrossRef] [PubMed]
  61. Mosmann, T. Rapid colorimetric assay for cellular growth and survival: Application to proliferation and cytotoxicity assays. J. Immunol. Methods 1983, 65, 55–63. [Google Scholar] [CrossRef]
  62. Matos, F.; Miguel, M.G.; Duarte, J.; Venâncio, F.; Moiteiro, C.; Correia, A.I.D.; Figueiredo, A.C.; Barroso, J.G.; Pedro, L.G. Antioxidant capacity of the essential oils from Lavandula luisieri, L. stoechas ssp. lusitanica, L. stoechas ssp. lusitanica x L. luisieri and L. viridis grown in Algarve (Portugal). J. Essent. Oil Res. 2009, 21, 327–336. [Google Scholar] [CrossRef]
  63. Lopes, V.R.; Barata, A.M.; Farias, R.; Mendes, M.D.; Lima, A.S.; Pedro, L.G.; Barroso, J.G.; Figueiredo, A.C. Morphological and essential oil variability from nine Portuguese fennel (Foeniculum vulgare Mill.) accessions. Acta Hort. 2010, 860, 33–50. [Google Scholar] [CrossRef]
  64. Faria, J.M.S.; Barbosa, P.; Bennett, R.N.; Mota, M.; Figueiredo, A.C. Bioactivity against Bursaphelenchus xylophilus: Nematotoxics from essential oils, essential oils fractions and decoction waters. Phytochemistry 2013, 94, 220–228. [Google Scholar] [CrossRef] [Green Version]
  65. Barbosa, P.; Faria, J.M.S.; Mendes, M.D.; Dias, L.S.; Tinoco, M.T.; Barroso, J.G.; Pedro, L.G.; Figueiredo, A.C.; Mota, M. Bioassays against pinewood nematode: Assessment of a suitable dilution agent and screening for bioactive essential oils. Molecules 2012, 17, 12312–12329. [Google Scholar] [CrossRef] [Green Version]
  66. Figueiredo, A.C.; Barroso, J.G.; Pedro, L.G. Volatiles from Thymbra and Thymus Species of the Western Mediterranean Basin, Portugal and Macaronesia. Nat. Prod. Commun. 2010, 5, 1465–1476. [Google Scholar] [CrossRef] [Green Version]
  67. Faria, J.M.S.; Lima, A.S.; Mendes, M.D.; Leiria, R.; Geraldes, D.A.; Figueiredo, A.C.; Trindade, H.; Pedro, L.G.; Barroso, J.G.; Sanches, J. Eucalyptus from Mata Experimental do Escaroupim (Portugal): Evaluation of the essential oil composition from sixteen species. Acta Hortic. 2011, 925, 61–66. [Google Scholar] [CrossRef]
  68. Miguel, M.G.; Gago, C.; Antunes, M.D.; Lagoas, S.; Faleiro, M.L.; Megías, C.; Cortés-Giraldo, I.; Vioque, J.; Figueiredo, A.C. Antibacterial, antioxidant and antiproliferative activities of Corymbia citriodora and eight Eucalyptus species essential oils. Medicines 2018, 5, 61. [Google Scholar] [CrossRef] [Green Version]
  69. Rodrigues, A.M.; Mendes, M.D.; Lima, A.S.; Barbosa, P.M.; Ascensão, L.; Barroso, J.G.; Pedro, L.G.; Mota, M.M.; Figueiredo, A.C. Pinus halepensis, Pinus pinaster, Pinus pinea and Pinus sylvestris essential oils chemotypes and monoterpene hydrocarbon enantiomers, before and after inoculation with the pinewood nematode Bursaphelenchus xylophilus. Chem. Biodivers. 2017, 14, e1600153. [Google Scholar] [CrossRef] [PubMed]
  70. Klein, E.; Schmidt, W. Structure of brachyl oxide. J. Agric. Food Chem. 1971, 19, 1115–1117. [Google Scholar] [CrossRef]
  71. Pereira, W.S.; da Silva, G.P.; Vigliano, M.V.; Leal, N.R.F.; Pinto, F.A.; Fernandes, D.C.; Santos, S.V.M.; Martino, T.; Nascimento, J.R.; de Azevedo, A.P.S.; et al. Anti-arthritic properties of crude extract from Chenopodium ambrosioides L. leaves. J. Pharm. Pharmacol. 2018, 70, 1078–1091. [Google Scholar] [CrossRef] [PubMed]
  72. Figueiredo, A.C. Biological properties of essentials oils and volatiles. Sources of variability. Nat. Volatiles Essent. Oils 2017, 4, 1–13. [Google Scholar]
  73. Ni, W.; Kitamoto, S.; Ishibashi, M.; Usui, M.; Inoue, S.; Huasa, K.-I.; Zhao, Q.; Nishida, K.-I.; Takeshita, A.; Eghashira, K. Monocyte chemoattractant protein-1 is an essential inflammatory mediator in angiotensin II-induced progression of established atherosclerosis in hypercholesterolemic mice. Arter. Thromb. Vasc. Biol. 2004, 24, 534–539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Hirota, R.; Roger, N.N.; Nakamura, H.; Song, H.-S.; Sawamura, M.; Suganuma, N. Anti-inflammatory effects of limonene from Yuzu (Citrus junos Tanaka) essential oil on eosinophilis. J. Food Sci. 2010, 75, H87–H92. [Google Scholar] [CrossRef]
  75. Chen, L.-L.; Zhang, H.J.; Chao, J.; Liu, J.F. Essential oil of Artemisia argyi suppresses inflammatory responses by inhibiting JAK/STATs activation. J. Ethnopharmacol. 2017, 204, 107–117. [Google Scholar] [CrossRef]
  76. Luo, G.; Kong, J.; Cheng, B.C.-Y.; Zhao, H.; Fu, X.-Q.; Yan, L.-S.; Ding, Y.; Liu, Y.-L.; Pan, S.-Y.; Zhang, S.-F.; et al. Xiao Qing Long Tang essential oil exhibits inhibitory effects on the release of pro-inflammatory mediators by suppressing NF-alphaB, AP-1, and IRF3 signalling in the lipopolysaccharide-stimulated RAW264.7 cells. Rsc Adv. 2019, 9, 12977–12989. [Google Scholar] [CrossRef] [Green Version]
  77. Park, H.; Seol, G.H.; Ryu, S.; Choi, I.-Y. Neuroprotective effects of (-)-linalool against oxygen-glucose deprivation-induced neuronal injury. Arch. Pharmacal Res. 2016, 39, 555–564. [Google Scholar] [CrossRef]
Table 1. Some of the traditional applications of the species studied in the present work.
Table 1. Some of the traditional applications of the species studied in the present work.
Family/Plant SpeciesCommon Names
(pt/en)
Medicinal UseOther UsesReference
Amaranthaceae
Dysphania ambrosioides (L.) Mosyakin & Clemants (= Chenopodium ambrosioides L.)Quenopódio/WormseedAgainst respiratory, gastrointestinal and joint inflammatory disordersVermifuge, emetic[13,14]
Apiaceae/Umbelliferae
Foeniculum vulgare Mill.Funcho/FennelAgainst respiratory and gastrointestinal inflammatory disordersCulinary (seasoning)[15]
Asteraceae/Compositae
Brachylaena huillensis O. Hoffm. (= Brachylaena hutchinsii Hutch., Brachylaena mullensis O.Hoffm.)Muhuhu */Silver oakAgainst schistosomiasis and roots against diabetesFirewood, charcoal, timber, poles, posts, tool handles, carving. Perfumery (essential oil distilled from wood)[16,17,18]
Solidago virgaurea L.Vara-de-ouro/European goldenrod or woundwortExternal and internal against urinary inflammatory disordersCosmetic[19]
Euphorbiaceae
Spirostachys africana Sond. [= Excoecaria africana (Sond.) Müll.Arg., Excoecaria synandra Pax, Excoecariopsis synandra (Pax) Pa, Sapium africanum (Sond.) Kuntze, Spirostachys synandra (Pax) Pax, Stillingia africana (Sond.) Baill.]Tambooti **External to treat myiasis, internal against gastrointestinal inflammatory disordersUse of wood in furniture[20,21,22]
Lamiaceae/Labiatae
Lavandula luisieri (Rozeira) Rivas-MartínezRosmaninho/butterfly lavenderExternal and internal against respiratory, circulatory, gastrointestinal and joint inflammatory disordersOrnamental, aromatic, cosmetic, culinary (seasoning)[23,24]
Mentha cervina L.Poejo fino/Hart’s pennyroyalExternal and internal against respiratory and gastrointestinal inflammatory disordersAromatic, culinary (seasoning)[25]
Origanum majorana L.Oregão/MarjoramExternal and internal against nervous, respiratory and gastrointestinal inflammatory disordersAromatic, culinary (seasoning)[26]
Satureja montana L.Segurelha/Winter savoryExternal and internal against nervous, respiratory and gastrointestinal inflammatory disordersCulinary (seasoning)[27]
Thymbra capitata (L.) Cav. [= Thymus capitatus Hoffms. et Link., Thymus creticus Brot., Corydothymus capitatus
Rechenb. f., Satureja capitata L.]
Tomilho-de-Creta/Conehead thymeExternal and internal against spasms and nervous, respiratory and gastrointestinal disordersAromatic, culinary (seasoning)[28]
Thymus mastichina (L.) L.Bela-luz/Spanish marjoramExternal and internal against nervous, respiratory, gastrointestinal and joint inflammatory disordersAromatic, culinary (seasoning)[28]
Thymus pulegioides L.Serpão/Broad-leaved thyme, lemon thymeExternal and internal against nervous, respiratory and gastrointestinal inflammatory disordersAromatic, culinary (seasoning)[29]
Thymus vulgaris L.Tomilho/thymeExternal and internal against nervous, respiratory and gastrointestinal inflammatory disordersOrnamental, aromatic, culinary (seasoning)[19]
Thymus zygis Loefl. ex L. subsp. zygisErva-de-Santa-Maria/Spanish red thymeExternal and internal against nervous, circulatory, respiratory and gastrointestinal inflammatory disordersAromatic, culinary (seasoning)[28]
Myrtaceae
Eucalyptus globulus subsp. maidenii (F.Muell.) J.B.Kirkp.Eucalipto/Maiden’s gumExternal and internal against circulatory, respiratory and gastrointestinal inflammatory disordersTimber, fuel, paper pulp. Aromatic, culinary (seasoning)[30]
Eucalyptus radiata A.Cunn. ex DC.Eucalipto/Narrow-leaved peppermint eucalyptusExternal and internal against mouth, respiratory and gastrointestinal inflammatory disorders [31]
Eucalyptus viminalis Labill.Eucalipto/Manna gumInternal against respiratory inflammatory disordersDeodorant[32]
Pinaceae
Pinus pinaster AitonPinheiro-bravo/Maritime pineExternal for circulatory problems, and internal against respiratory, gastrointestinal and joint inflammatory disordersTimber and oleoresin production[33]
pt/en: Official two-letter codes of Portuguese and English languages, respectively. * African name adopted in Portuguese. ** African name given to the wood and adopted in Portuguese and English.
Table 2. Previously reported anti-inflammatory activity of the essential oils (EOs) from the species under study.
Table 2. Previously reported anti-inflammatory activity of the essential oils (EOs) from the species under study.
EO/EO Components’ Anti-Inflammatory ActivityFamily/SpeciesReference
Apiaceae
Foeniculum vulgareEO inhibition of 5-lipoxygenase (IC50 = 0.04 mg/mL). Fenchone inhibition of 5-lipoxygenase (IC50 = 0.02 mg/mL).[37]
EO (200 and 400 mg/kg) decreased the activity of mieloperoxidase (MPO) and the expression of TNF-α in the colon tissue previously submitted to acetic acid solution (acute colitis), and inhibited acetic acid-induced expression of p‑NF-kB p65 protein.[38]
Lamiaceae
Lavandula luisieriEO (50–200 mg/kg) inhibition of paw edema (31–83%) induced by carrageenan administered in male Wistar rats.[39]
EO (25 μg/mL) nitric oxide (NO) inhibition (75%) in IL-1β induced primary chondrocyte.[40]
EO reduction of iNOS in human chondrocytes and intestinal cell line C2BBe1 (54.9 and 81.0%, respectively) and phosphorylated IkB-α (87.4% and 62.3%, respectively).[41]
Origanum majoranaEO (10 μg/mL) diminished the TNF-α, IL‑1β, IL-6, IL-10 and COX-2 secretion and NFκB gene expression after activation of THP-1 cells by lipopolysaccharide or human ox – LDL. The activity was attributed to cis-sabinene hydrate and terpinen‑4‑ol.[42]
Thymbra capitataEO inhibition of 5-lipoxygenase (IC50 = 0.1 mg/mL).[43]
EO inhibition of 5-lipoxygenase (IC50 = 0.2 mg/mL).[44]
Thymus mastichinaEO inhibition of 5-lipoxygenase (IC50 = 0.7 mg/mL).[43]
Thymus vulgarisEO inhibition of 5-lipoxygenase (IC50 = 0.19 μg/mL).[37]
EO (0.5 μg/mL) inhibition (80%) of 5-lipoxygenase.[45]
EO inhibition of 5-lipoxygenase (IC50 = 0.005 μg/mL). EO reduced the TNF-α, IL-1β, IL-8 secretion levels of THP-1 cells.[46]
EO (400 mg/kg, after 6 h) reduced (50.4–58.4%) carrageenan-induced paw edema in mice.[47]
Carvacrol (10 mg/ear) reduced ear edema. Carvacrol (10 mg/ear) inhibited the activity of myeloperoxidase (MPO) (43.8%). Carvacrol (0.3–90 μg/mL) reduced (20.07–52.23%) neutrophil migration in response to fMLP stimulation. EO (750 mg/kg) and carvacrol (100–400 mg/kg) exerted inhibited leukocyte migration to the injury site. Carvacrol (0.3‑90μg/mL) reduced LTB4 stimulation (19.8–61.1%).[48]
EO and carvacrol suppressed lipopolysaccharide-induced COX-2 mRNA and protein expression in human macrophage-like U937 cells.[49]
EO (moderate concentration) decreased the mRNA levels of IL-1β, IL-6, granulocyte-macrophage colony stimulating factor (GM-CSF) and TNF-α, and lowered the amount of IL-1β and IL-6 proteins in animal models of colitis.[50]
EO reduced production and gene expression of the pro-inflammatory mediators TNF‑α, IL-1B and IL-6 and increased the parameters on the anti-inflammatory IL-10 cytokine.[51]
EO (5000 ppm) decreased paw edema and ear swelling, inhibited the total mRNA IL‑1β expression in the mouse colon.[52]
Thymus zygis subsp. zygisEO thymol type inhibition of 5-lipoxygenase (IC50 = 54 – 73 μL/L).EO linalool type inhibition of 5-lipoxygenase (IC50 = 299 – 402 μL/L).[53]
EO reduced production and gene expression of the pro-inflammatory mediators TNF‑α, IL-1B and IL-6 and increased the parameters on the anti-inflammatory IL-10 cytokine.[51]
Myrtaceae
Eucalyptus globulusEO inhibition of 5-lipoxygenase (IC50 = 0.16 mg/mL).[37]
subsp. maideniiEO (0.5μg/mL) inhibition (50%) of lipoxygenase.[45]
EO (200 mg/kg) inhibited by 28.8% the inflammatory phase of wound healing (Whittle method).[54]
Pinaceae
Pinus pinasterEO (100 mg/kg dose) inhibition (30.3%) of paw edema in the Whittle method using carrageenan.[55]
EO: Essential oil. IC50: Half-maximal inhibitory concentration. LDL: Low-density lipoprotein. COX‑2: Ciclo‑oxigenase-2.
Table 3. List of the evaluated species, sampling year, plant part used for hydrodistillation, plant source, essential oil yield and main components (≥10%).
Table 3. List of the evaluated species, sampling year, plant part used for hydrodistillation, plant source, essential oil yield and main components (≥10%).
Family/SpeciesCodeSampling DatePlant PartCollection Place/Source #EO yield (%, v/w)Main Components (≥10%)
Amaranthaceae
Dysphania ambrosioidesDa2013FFMonsaraz0.56iso-Ascaridole 51, ascaridole 16
Apiaceae
Foeniculum vulgareFv2013DVHerbal shop α-Pinene 27, trans-anethole 18, Limonene 11
Fv s2013SeedsHerbal shop1.16Methyl chavicol 79, limonene 12
Asteraceae
Brachylaena huillensisBh2013EOAngolan.a.Copaen-15-ol * 14
Bh *2013EO*Angolan.a.Copaen-15-ol * 12
Solidago virgaureaaSv2013FFPinheiro da Cruz0.72β-Pinene 22, α-pinene 21, germacrene D 15, limonene 12
Euphorbiaceae
Spirostachys africana 2013EOAngolan.a.Stachenone * 28, Diosphenol (2) * 38
Lamiaceae
Lavandula luisieria,bLl2013DFHerbal shop0.445-Methylene-2,3,4,4-tetramethylcyclopent-2-enone 18, 1,8-cineole 16
Mentha cervinaMc2013DVHerbal shop1.54Pulegone 76
Origanum majoranaaOm2013DVHerbal shop0.98Terpinen-4-ol 18, carvacrol 17, γ‑terpinene 13, carvacrol methyl ether 13
Satureja montanaaSm2013DVHerbal shop1.48Carvacrol 77
Thymbra capitataTc2013FVAlgarve0.89Carvacrol 71
Thymus mastichinaThm2013FFBragança1.351,8-Cineole 69
Thymus pulegioidesaThp2013DLHerbal shop0.49Thymol 32, ρ-cymene 22
Thymus vulgarisaThv2013DVHerbal shop1.20Thymol 45, ρ-cymene 21, γ‑terpinene 16
Thymus zygis subsp. zygis aThzz2013FFBragança0.71Carvacrol 45, ρ-cymeme 22, γ‑terpinene 17
Myrtaceae
Eucalyptus globulus subsp. maideniiEg2013FLMEE3.20α-Pinene 15, 1,8-Cineole 46, Limonene 23
Eucalyptus radiataEr2012FLMEE7.201,8-Cineole 49
Eucalyptus viminalisEv2012FLMEE2.50α-Pinene 10, 1,8-Cineole 69
Pinaceae
Pinus pinasterPp2013OleoresinNazaré29.76α-Pinene 62, β-pinene 23
# Unless otherwise specified, the collection place was in Portugal. a Detailed composition of EOs reported in Faria et al. [58]. b Commercialized as Lavandula stoechas L. n.a.: Information not available. * Identification based on mass spectra only. DF: Dry, flowering phase aerial parts. DL: Dry leaves. DV: Dry, vegetative phase aerial parts. EO: Essential oil supplied by the producer, obtained from the wood. EO*: In-lab re-distilled essential oil supplied by the producer, due to some turbidity of the original sample. FF: Fresh, flowering phase aerial parts. FL: Fresh leaves from fruiting phase. MEE: Mata Experimental do Escaroupim.
Table 4. Inhibition of TNF-α and CCL2 production by lipopolysaccharide (LPS)-activated THP-1 monocytic cells elicited by the evaluated essential oils (EOs).
Table 4. Inhibition of TNF-α and CCL2 production by lipopolysaccharide (LPS)-activated THP-1 monocytic cells elicited by the evaluated essential oils (EOs).
Family/Species and ControlConcentrations
(µg/mL)
TNF-α Inhibition
(% ± S.D., n = 3)
CCL2 Inhibition
(% ± S.D., n = 3)
ControlLPS (200 ng)2428.1 ± 587.8 a2382.3 ± 1480.8 a
DMSO (0.1%)96.5 ± 13.9 a24.1 ± 14.7 a
Amaranthaceae
Dysphania ambrosioides9048.6 ± 2.1 ***15.6 ± 0.7 ***
3030.9 ± 1.5 *9.4 ± 0.3 **
1013.6 ± 1.07.5 ± 0.3 *
Apiaceae
Foeniculum vulgare9022.3 ± 1.9 ***NI
300.5 ± 0.0NI
10NINI
Asteraceae
Brachylaena huillensis30NI18.8 ± 2.3 **
(re-distilled EO)104.4 ± 0.89.0 ± 0.5
3NI5.4 ± 0.2
Brachylaena huillensis30NDND
10NDND
3NDND
Solidago virgaurea90NDND
305.0 ± 0.2ND
10NI4.9 ± 0.1 *
3ND8.0 ± 0.1 **
Euphorbiaceae
Spirostachys africanus30NDND
10NDND
3NDND
Lamiaceae
Lavandula luisieri9082.9 ± 8.2 ***82.0 ± 12.4 ***
3023.2 ± 1.1 ***54.3 ± 3.0 ***
102.5 ± 0.122.7 ± 1.0 ***
Mentha cervina90NINI
30NINI
10NI2.5 ± 0.0
Origanum majonara90NINI
30NINI
10NI4.8 ± 0.1
Satureja montana30NDND
10NDND
3NI0.2 ± 0.0
Thymbra capitata30NDNI
2051.1 ± 6.5 ***ND
1029.5 ± 1.7 ***0.4 ± 0.0
59.1 ± 0.1 *ND
Thymus mastichina90NDND
30NDND
10NDND
Thymus pulegioides90NDND
30NI0.9 ± 0.0
10NI8.4 ± 0.5
Thymus vulgaris90NDND
30NDND
10NDND
Thymus zygis ssp. sygis30NI8.9 ± 1.0
10NI2.7 ± 0.2
3NI0
Myrtaceae
Eucapyptus globulus subsp.906.5 ± 0.41.7 ± 0.0
maidenii304.4 ± 0.3NI
102.4 ± 0.2NI
Eucalyptus radiata9012.0 ± 0.1 *NI
30NINI
10NINI
Eucalyptus viminalis903.3 ± 0.2NI
300.2 ± 0.0NI
10NI1.4 ± 0.0
Pinaceae
Pinus pinaster (oleoresin)90NDND
306.3 ± 0.1NI
106.8 ± 0.3NI
a Inflammatory mediator production (absolute values in pg/mL). NI: No inhibition. ND: Not determined due to toxicity (cell viability ≤ 90%). * p ≤ 0.05; ** p ≤ 0.01; *** p ≤ 0.001: Indicates significant inhibition of TNF-α or CCL2 release in comparison to LPS-stimulated cells (ordinary one-way ANOVA/Newman–Keuls multiple comparison test: GraphPad Prism).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Miguel, M.G.; da Silva, C.I.; Farah, L.; Castro Braga, F.; Figueiredo, A.C. Effect of Essential Oils on the Release of TNF-α and CCL2 by LPS-Stimulated THP‑1 Cells. Plants 2021, 10, 50. https://doi.org/10.3390/plants10010050

AMA Style

Miguel MG, da Silva CI, Farah L, Castro Braga F, Figueiredo AC. Effect of Essential Oils on the Release of TNF-α and CCL2 by LPS-Stimulated THP‑1 Cells. Plants. 2021; 10(1):50. https://doi.org/10.3390/plants10010050

Chicago/Turabian Style

Miguel, Maria Graça, Carina Isabel da Silva, Luana Farah, Fernão Castro Braga, and Ana Cristina Figueiredo. 2021. "Effect of Essential Oils on the Release of TNF-α and CCL2 by LPS-Stimulated THP‑1 Cells" Plants 10, no. 1: 50. https://doi.org/10.3390/plants10010050

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop