Next Article in Journal
Antioxidant and Antimicrobial Effects of Baby Leaves of Amaranthus tricolor L. Harvested as Vegetable in Correlation with Their Phytochemical Composition
Next Article in Special Issue
Phytochemical Composition and Antimicrobial Activity of Essential Oil from the Leaves of Artemisia vulgaris L.
Previous Article in Journal
Synthesis of Enantiomerically Enriched Protected 2-Amino-, 2,3-Diamino- and 2-Amino-3-Hydroxypropylphosphonates
Previous Article in Special Issue
Chemical Composition, Enantiomeric Distribution, Antimicrobial and Antioxidant Activities of Origanum majorana L. Essential Oil from Nepal
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Anti-Trypanosomatidae Activity of Essential Oils and Their Main Components from Selected Medicinal Plants

by
María Bailén
1,*,
Cristina Illescas
1,
Mónica Quijada
1,
Rafael Alberto Martínez-Díaz
1,†,
Eneko Ochoa
2,
María Teresa Gómez-Muñoz
3,
Juliana Navarro-Rocha
4 and
Azucena González-Coloma
5
1
Department of Preventive Medicine, Public Health and Microbiology, Faculty of Medicine, Universidad Autónoma de Madrid, 28049 Madrid, Spain
2
Research and Development Division, AleoVitro, 48160 Derio, Spain
3
Department of Animal Health, Faculty of Veterinary Sciences, University Complutense of Madrid, 28040 Madrid, Spain
4
Centro de Investigación y Tecnología Agroalimentaria de Aragón, Unidad de Recursos Forestales, 50059 Zaragoza, Spain
5
Instituto de Ciencias Agrarias, CSIC, 28006 Madrid, Spain
*
Author to whom correspondence should be addressed.
Deceased.
Molecules 2023, 28(3), 1467; https://doi.org/10.3390/molecules28031467
Submission received: 16 January 2023 / Revised: 28 January 2023 / Accepted: 30 January 2023 / Published: 2 February 2023

Abstract

:
Kinetoplastida is a group of flagellated protozoa characterized by the presence of a kinetoplast, a structure which is part of a large mitochondria and contains DNA. Parasites of this group include genera such as Leishmania, that cause disease in humans and animals, and Phytomonas, that are capable of infecting plants. Due to the lack of treatments, the low efficacy, or the high toxicity of the employed therapeutic agents there is a need to seek potential alternative treatments. In the present work, the antiparasitic activity on Leishmania infantum and Phytomonas davidi of 23 essential oils (EOs) from plants of the Lamiaceae and Asteraceae families, extracted by hydrodistillation (HD) at laboratory scale and steam distillation (SD) in a pilot plant, were evaluated. The chemical compositions of the EOs were determined by gas chromatography-mass spectrometry. Additionally, the cytotoxic activity on mammalian cells of the major components from the most active EOs was evaluated, and their anti-Phytomonas and anti-Leishmania effects analyzed. L. infantum was more sensitive to the EOs than P. davidi. The EOs with the best anti-kinetoplastid activity were S. montana, T. vulgaris, M. suaveolens, and L. luisieri. Steam distillation increased the linalyl acetate, β-caryophyllene, and trans-α-necrodyl acetate contents of the EOs, and decreased the amount of borneol and 1,8 cineol. The major active components of the EOs were tested, with thymol being the strongest anti-Phytomonas compound followed by carvacrol. Our study identified potential treatments against kinetoplastids.

Graphical Abstract

1. Introduction

Kinetoplastida is a group of flagellated protozoa characterized by the presence of a particular structure, known as a kinetoplast, which is located in the single mitochondrion of the cell and is composed of mitochondrial DNA. Despite kinetoplastids having similarities in their genomic organization and cellular structures, they comprise a large variety of organisms, many of them pathogenic, and transmitted by different vectors, causing different diseases in multiple hosts [1]. Parasites of the genera Leishmania [2] and Trypanosoma [3] cause disease in humans, while pathogens of the genus Phytomonas are capable of infecting plants [4].
Leishmania spp. causes leishmaniasis, a disease affecting populations with low resources [5]. In spite of its great epidemiological importance, leishmaniasis is considered by WHO as a neglected disease [6]. Treatment is based on pharmacological therapies; however, many of the currently employed drugs present high toxicities, decreased efficacies, difficulties of administration, and high costs, and the emergence of resistant strains has been reported [7].
Phytomonas comprises a wide range of parasite species which infect a diversity of vascular plants, many of them of great economic importance, such as cashew, coffee, oil palm, tomato, orange, and grape, among others [8]. The main symptoms of the disease caused by these parasites are leaf chlorosis and root atrophy [9]. Currently, there are no effective treatments and the strategies for infection control are the felling of diseased plants or the removal of infected plant material [4]. Additionally, Phytomonas share similar structural components and similarities in life cycle with other kinetoplastids such as Leishmania or Trypanosoma, which allows them to be used a laboratory safe model in the search for new anti-kinetoplastids compounds [8].
Natural products are chemical compounds produced by living organisms that have been used in traditional medicine for thousands of years, still serving today as the basis for many pharmaceuticals [10]. They are environmentally friendly due to their biodegradability and low toxicity to mammals [11]. EOs are plant-derived natural products consisting of complex mixtures of volatile, lipophilic, low molecular weight compounds, with terpenoids and phenylpropanoids among their most common constituents [12], in which hydrocarbons and oxygenated compounds stand out [13]. The composition of EOs may be influenced by several factors, such as water availability, soil composition, climate, nutrients, organ used for extraction, pathogen attack, or type of extraction [14]. They play an important ecological role in plants by attracting pollinators and beneficial insects, protecting from heat and cold, and being used as chemical defenses against pests, pathogenic microorganisms, or vertebrate predators [15]. EOs have pharmacological interest because of their biocidal effects on pathogenic microorganisms, among other properties [16]. They present a broad pharmacological spectrum that includes analgesic, sedative, anti-inflammatory, antispasmodic, antimicrobial, antiprotozoal, and anthelmintic properties [16,17], as well as insecticidal effects [18], and can be used to overcome toxicity and the emergence of drug-resistant pathogens associated with routine treatments [14].
Moreover, promising effects have been observed from the combination of natural products and approved antileishmanial drugs. Meglumine antimoniate combined with capsaicin or piperine produced synergistic effects on L. infantum promastigotes and amastigotes, which can help to reduce the amount of antimonials administered and its associated toxicity [19]. Another example was the effect of an ethanolic extract of Moringa oleifera in combination with amphotericin B on L. major amastigotes and promastigotes [20].
Several studies have determined that EOs from plant species belonging to the genera Artemisia [21], Thymus, Lavandula [14], Salvia, and Origanum [22], among others [23], exhibit antiparasitic properties against multiple protozoa of the kinetoplastida class.
There is a need to seek potential alternative treatments against Leishmania sp. and Phytomonas sp., among other kinetoplastids, considering the low efficacy or high toxicity of some of the current treatments against Trypanosomatids. In the present work, the antiparasitic activity of 23 EOs from plants of the Lamiaceae (Salvia, Thymus, Lavandula, Satureja, Origanum, Mentha, and Rosmarinus) and Asteraceae families (Ditrichia, Santolina, and Tanacetum), some extracted by two different methods (laboratory-scale hydrodistillation and pilot plant-scale steam distillation), were evaluated against Leishmania infantum and Phytomonas davidi. Additionally, the cytotoxic activity on mammalian cells of the major constituents from the most active EOs was evaluated, and their anti-Phytomonas and anti-Leishmania effects analyzed.

2. Materials and Methods

2.1. Plants and Essential Oil Extraction

The EOs studied in this work were obtained from four species of the Asteraceae family and seventeen species of the Lamiaceae family. The plant species Tanacetum vulgare, Lavandula x intermedia “Grosso”, Salvia blancoana, and Thymus mastichina were cultivated in distinct locations of Spain (see Table S1 for locations and voucher numbers). The plant species were identified by Dr. Daniel Gomez, IPE-CSIC, and the seeds were deposited at the CITA (Centro de Investigación y Tecnología Agroalimentaria de Aragón, Unidad de Recursos Forestales, Zaragoza, Spain) germplasm bank. Location, identification, and voucher numbers of the remaining species have been previously reported by Bailén et al. [24].
The EOs were obtained at the Research Center and Food Technology (CITA-Aragón, Spain) using two different methods, hydrodistillation (HD) and steam distillation (SD), as described by Bailén et al. [24]. Aerial plant parts were collected at the flowering stage between 2016 and 2019. The hydrodistillation was carried out in triplicate with 100 g of dried aerial plant parts and 1 L of water for 1 h in a Clevenger-type. The oils were dried over MgSO4, filtered and stored at 4 °C until used. Pilot plant steam distillation was carried out on the fresh biomass of the plants (60 kg total fresh plant biomass) harvested at the flowering stage. A stainless-steel pilot extraction plant equipped with a pressure reducing valve was used as described. The pressure of the work was 0.5 bar. The hydrolate (aqueous phase) was decanted from the essential oil collected in the condensation section, and filtered.

2.2. EO Analysis

The EOs were analyzed by gas chromatography-mass spectrometry (GC-MS) using a Shimadzu GC-2010 Plus coupled to a Shimadzu GCMS-QP2010-Ultra mass detector with an electron impact ionization source at 70 eV and a Single Quadrupole analyzer, and employing Helium as carrier gas. The samples were injected by an automatic injector (AOC-20i). Chromatography was carried out with a Teknokroma TRB-5 (95%) Dimethyl- (5%) diphenylpolysiloxane capillary column, 30 × 0.25 mm ID and 0.25 μm phase thickness. The working conditions used were: Split mode injection using 1 µL of sample with a split ratio (20:1) employing a Shimadzu AOC-20i automatic injector, injector temperature 300 °C, transfer line temperature connected to the mass spectrometer 250 °C, and ionization source temperature 220 °C. The initial column temperature was 70 °C, heating up to 290 °C at 6 °C/min, and leaving at 290 °C for 15 min. All the samples (4 g/µL) were previously dissolved in 100% dichloromethane (DCM) for injection.
The mass spectra and retention time were used to identify the compounds by comparison with those found in the Wiley database (Wiley 275 Mass Spectra Database, 2001) and NIST 17 (NIST/EPA/NIH Mass Spectral Library), while the relative area percentages of all peaks obtained in the chromatograms were used for quantification. Identification of necrodanes in L. luisieri was performed with our own database (CSIC), built by the injection of pure compounds isolated from this plant [25]. In addition, to confirm the identities of the constituents, the retention index of marker constituents of known EOs were used.

2.3. Pure Compounds

After analyzing the composition of the EOs by GC-MS, thymol, carvacrol, γ-terpinene and p-cymene, major components of EOs with anti-Phytomonas activity, were selected for further studies. Some of the major compounds that were excluded were not identified, not commercially available, or not easy to obtain or isolate. Pure compounds (monoterpenes) were obtained from commercial sources. Thymol (≥98.5%) was obtained from Sigma Aldrich (Madrid, Spain), γ-terpinene (97%) and p-cymene (99%) from Acros Organics (Madrid, Spain), and carvacrol (98%) from Fluka (Madrid, Spain).

2.4. Anti-Parasitic Activity In Vitro

Anti-Phytomonas (AP) and anti-Leishmania (AL) activity studies were performed on promastigote forms of P. davidi ATCC® 30287™, isolated from Euphorbia heterophylla [26] and L. infantum “JPC” (MCAN/ES/98/LLM-722), kindly donated by Dr. J.M Requena from CBM-CSIC. The P. davidi promastigotes were cultured in LIT medium and the L. infantum JPC in RPMI medium, supplemented with 10% and 15% heat-inactivated fetal calf serum (FCS), respectively, at 28 °C. RPMI was also supplemented with 10 μg/mL of hemin (Acros Organics, Madrid, Spain). Parasites in the logarithmic growth phase were distributed in 96-well flat-bottom plates (95 μL of culture/well). EOs and compounds in DMSO were tested in quadruplicate at various concentrations for 24 h (Phytomonas) and 48 h (Leishmania) at several concentrations (EOs at 800, 400 and 200, 100, 50 and 25 µg/mL; and pure compounds at 100, 50, 25, 10 and 1 μg/mL). Amphotericin B was used as a reference drug. The parasite viability was analyzed by a modified MTT colorimetric assay method [27]. The percentage of AP activity and AL activity was calculated as growth inhibition using the following formula: % AP or AL = 100 − [(Ap − Ab) ÷ (Ac − Ab)] × 100; Ap is the absorbance of the tested product, Ab the absorbance of the blank, and Ac the absorbance of the control wells (not treated).

2.5. Cytotoxicity of Pure Compounds

African green monkey kidney cells (Vero cells) were maintained in Dulbecco’s modified Eagle’s minimal essential medium (DMEM) supplemented with 10% fetal calf serum and 1% penicillin/streptomycin (Fisher Scientific, Madrid, Spain) at 37 °C under a humidified atmosphere of 5% CO2/95% air.
Cells seeded in 96-well flat-bottom microplates with 100 μL medium per well (initial densities 104 cells per well) were exposed for 48 h to serial dilutions (100, 75, 50, 25, 10 and 1 µg/mL) of the tested compounds in DMSO (< 1% final concentration). The cell viability was analyzed by the MTT colorimetric assay method, and the purple-colored formazan precipitate was dissolved with 100 μL of DMSO [24].

2.6. Statistical Analysis

The data were analyzed using STATGRAPHICS Centurion XIX (https://www.statgraphics.com, accessed on 5 September 2022). A parametric bivariate correlation analysis was performed between the main components of the EOs, present in a proportion higher than 5%, and the AL or AP activity (IC50).
The cells’ and promastigotes’ viabilities were tested with each compound in a dose-response experiment, to calculate their relative potency (CC50 or IC50 value). IC50 (μg/mL) expresses the dose of EOs or pure compounds needed to produce 50% mortality of promastigotes, while CC50 (μg/mL) expresses the dose of compounds necessary to produce 50% mortality of Vero cells.
The selectivity index was calculated for the AP or AL of pure compounds, using the formula SI = IC50/CC50. Compounds with an SI higher than one were considered as potential anti-Phytomonas or anti-Leishmania compounds, since they are more toxic for kinetoplastids than for mammalian cells.

3. Results

3.1. Antiprotozoal Activity of EOs from Lamiaceae and Asteraceae Plants

The activity of 23 EOs from 17 selected species (domesticated or undergoing domestication) belonging to the Asteraceae and Lamiaceae families were evaluated against the kinetoplastids P. davidi and L. infantum. All of these EOs were obtained at laboratory scale by hydrodistillation (HD). EOs from the domesticated plants cultivated in the field (a total of 17) were also extracted at a semi-industrial scale, representative of a potential commercial situation, by steam distillation (SD) (Table 1).
Eight EOs, from five Lavandula species, were tested against Leishmania. Six of them showed high antileishmanial activities at 800 µg/mL (AL > 70%), one had a moderate effect (AP: 50–70%), and one was not active (Figure 1A and Figure S1A). The most effective Lavandula EOs were L. luisieri 1 (SD) and L. luisieri 2 (HD and SD), followed by L. mallete (SD) and L. intermedia “Abrial” (SD). The EOs with the highest anti-Phytomonas activity were L. intermedia “Super”, L. luisieri 1 (HD), and L. mallete (HD). Overall, EOs obtained by SD from Lavandula sp. were more active against Leishmania and those extracted by HD were more active against Phytomonas.
EOs belonging to S. montana, M. suaveolens, and R. officinalis had effects on both parasites. Differences related to the EO extraction method were observed for R. officinalis against both parasites (Figure 1 and Figure S1). S. montana EOs had the highest antiprotozoal activity against both parasites and the AP activity was maintained even at 100 µg/mL. Among the four EOs from Salvia spp., two were highly active against L. infantum, and two had no activity (Table 1). The EOs with the largest effects were S. hybrid (HD) and S. officinalis (SD). Worse results were obtained against Phytomonas, with three EOs having moderate effects and one showing no activity. S. officinalis was the Salvia species with the highest anti-Trypanosomatidae activity. The EOs from Origanum and Thymus spp. showed high or moderate activity against Leishmania. The best AL effects were obtained for T. zygis, and T. vulgaris (SD EOs). On P. davidi, T. vulgaris (HD) showed the highest activity at 200 µg/mL. No activity against Phytomonas was found for T. mastichina, T. zygis (HD), or O. majorana.
The EOs from the Asteraceae (Tanacetum, Santolina, and Ditrichia) showed moderate antiprotozoal activities, except for T. vulgare which showed no activity on P. davidi and L. infantum.
Overall, antiparasitic results of the tested EOs showed that L. infantum was more sensitive to the EOs than P. davidi (50% vs. 30% active EOs: IC50 < 500 μg/mL). The most active EOs against P. davidi (IC50 < 100 μg/mL) were S. montana (SD), T. vulgaris (HD) and S. montana (HD), whilst for L. infantum they were M. suaveolens (SD), L. luisieri 2 (SD), and L. luisieri 1 (SD). In general, the EOs extracted by SD performed better against L. infantum (100% SD EOs).

3.2. Chemical Composition of Essential Oils

EOs with an IC50 lower than 200 μg/mL were selected for further analysis (both extraction methods), adding to a total of six species and seven EOs (Table 2 and Table S2; Figures S2–S14). Only components with a proportion higher than 5% were considered.
Camphor, trans-α-necrodyl acetate, and fenchone were predominant in L. Luisieri (1 and 2), with fenchone being more effectively extracted by HD and trans-α-necrodyl acetate by SD. For M. suaveolens, the main components were piperitenone and piperitenone oxide, which accounted for more than 60% of the total composition. Piperitenone was more abundant in the EO obtained by HD and piperitenone oxide in the EO obtained by SD. Among the components of S. hybrid HD, 1,8 cineol, camphor, and trans-bornyl acetate appeared in higher proportions.
In the chemical composition of S. montana, thymol, p-cymene, and carvacrol stand out, with thymol and p-cymene being more abundant when extracted by HD and carvacrol when the extraction was done by SD. Thymol and p-cymene were also the main components of T. vulgaris and T. zygis. Linalyl acetate was also an abundant component, but only for T. zygis SD EO.
Overall, HD extraction increased borneol and 1,8 cineol, whilst SD extraction favored linalyl acetate, β-caryophyllene, and trans-α-necrodyl acetate.
A parametric bivariate correlation analysis was performed between the main components of the EOs and the AL and AP activities. There were two significant correlations for AL and three for AP (p < 0.05) (Table 3). Among the compounds with significant correlations, piperitenone oxide was directly associated with the AL activity (negative correlation between compound abundance and IC50 of the EO), whereas p-cymene, thymol, and carvacrol were directly associated with the AP activity of the EOs. Only α-terpineol was inversely associated with AL activity (positive correlation between compound abundance and IC50 of the EO).

3.3. Anti-Phytomonas and Cytotoxic Activity of Pure Compounds

A total of four compounds (Figure 2), selected from the chemical composition of the most active EOs, were tested on L. infantum and P. davidi, and also their cytotoxic effects on Vero cells were evaluated (Table 4). Amphotericin B was used as a reference drug. Data previously reported from these compounds on Leishmania sp. have been included in Table 4. Only compounds 1 (thymol) and 2 (carvacrol) showed AP activities with moderate effects, with 1 (thymol) being the compound with the highest AP activity (IC50: 45 µg/mL). The highest SI was observed for thymol (SI: 2.22). Compounds 1 (thymol) and 2 (carvacrol) were also the terpenes with the highest selectivity indexes (SI: 10.20 and 13.85 respectively) on L. infantum. p-Cymene (4) was the only terpene previously tested on other Leishmania species (L. chagasi) [28]. None of the tested compounds were cytotoxic against Vero cells.

4. Discussion

Lamiaceae is an important family, with a variety of aromatic and medicinal genera such as Rosmarinus, Origanum, Thymus, Lavandula, Mentha, and Satureja, among many others [31]. The most active EOs found in our study for both kinetoplastids, L. infantum and P. davidi, belong to this family, highlighting EOs from M. suaveolens and S. montana. The Asteraceae plant family has emerged as a new source of trypanocidal and leishmanicidal compounds [32]. However, the Asteraceae EOs tested in this study only had moderate effects on both targets.
Overall, the results of the tested EOs in our study showed that L. infantum was more sensitive to the EOs than P. davidi, probably because Phytomonas is a plant parasite, and thus it is more adapted to the chemical defensive components of plants. These results agree with those found by Sainz et al. when comparing the activity of various EOs on T. cruzi and P. davidi [27]. The activity of EOs on Phytomonas sp. have been less studied than the leishmanicidal effects. Even so, EOs from other plant families have been found to have anti-Phytomonas activity. EOs from Varronia curassavica genotypes have antiprotozoal activity against Phytomonas serpens causing alterations in the permeabilization of the cytoplasmic membrane [33]. Lantana camara EOs, extracted at different harvesting times, displayed trypanocidal activity on P. serpens [34], and EOs from Hyssopus officinalis showed antiprotozoal activity on T. cruzi and P. davidi [35]. However, this is the first report on the phytomonacidal effects of the EOs studied here.
M. suaveolens is an aromatic species usually employed in traditional medicine, with reported cytotoxic, antioxidant, anti-inflammatory, antifungal, antiviral, and insecticidal properties [36]. Our results indicate that M. suaveolens EO had the highest antileishmanial effects of all the tested EOs, as well as moderate effects on Phytomonas. This EO contained high proportions of piperitenone oxide and piperitenone. Piperitenone oxide could be involved in the antileishmanial activity, as it has been previously been reported to have trypanocidal, insecticidal, and schistosomicidal effects [37,38,39]. There are also other species of the genus Mentha with antileishmanial properties on promastigotes of various species of Leishmania. EOs from M. australis and M. microphylla have antileishmanial effects on L. major [40], M. pulegium on L. major, L. infantum and L. tropica [41], M. x piperita on L. infantum and L. donovani [42,43], and M. cervina on L. infantum [42,44]. However, none of the mentioned EOs had piperitenone oxide as a major component.
Another plant species with strong anti-kinetoplastid activity in our study was S. montana. The genus Satureja comprises numerous species of shrubs or aromatic herbs with acaricidal, insecticidal, and antiparasitic properties [45]. Satureja montana EO has interesting properties against bacteria, fungi, viruses, helminths, insects, and protozoa [24,46,47,48,49]. EOs from Satureja khuzestanica, Satureja bakhtiarica, and Satureja punctata demonstrated antileishmanial effects on L. major, L. donovani, and L. aethiopica previously [50,51,52]. S. montana EO (HD and SD) was active against both parasites, with P. davidi being more sensitive. Thymol and carvacrol, which correlated with AP activity, were the main components of the Satureja EO.
EOs from two populations of L. luisieri (1 and 2) had the strongest effects among the tested lavandulas on L. infantum. Essential oils from plants of the Lavandula genus have reported acaricidal, antibacterial, antifungal, antioxidant, and anti-parasitic effects [53]. EOs from L. angustifolia, L. stoechas, L. viridis, and L. luisieri had reported effects on L. major, L. infantum, and L. tropica [42,53,54,55]. However, the effect found here was weaker than that observed by Machado et al. on L. infantum promastigotes [53]. Trans-α-necrodyl acetate is one of the major components of L. luisieri EOs. Therefore, necrodane derivatives should be further studied to verify their antileishmanial properties. L. luisieri EOs can produce leishmanicidal effects through different mechanisms, but mainly through unleashed apoptosis, with phosphatidylserine externalization, mitochondrial membrane potential loss, and G0/G1 phase cell cycle arrest being the most remarkable aspects involved in apoptosis [53]. On P. davidi, the studied Lavandula EOs only had moderate effects, with L. intermedia “Super” being the most active.
T. vulgaris HD EO, showing a remarkable anti-Phytomonas activity, had the highest content of thymol (48%) of all the tested EOs. As mentioned before, thymol correlated with AP activity. EOs from T. zygis, Thymus capitelatus, and T. mastichina have been reported to have antileishmanial effects on L. infantum promastigotes [42,56].
Among the genus Salvia, the EOs from S. hybrid and S. officinalis showed important antileishmanial effects, while S. blancoana EOs had moderate action against P. davidi. EOs from various species of Salvia have been previously tested on Phytomonas and Leishmania, with moderate effects [17,22].
In this study, the EO (HD and SD) from R. officinalis showed a moderate effect on L. infantum. Previous reports have showed strong antileishmanial effects of R. officinalis EOs on L. major, L. tropica, and L infantum [41,54]. Variations in the chemical composition of the EOs could explain these differences.
Origanum majorana is a shrub found in Asia and in the Mediterranean area, with reported antibacterial, antifungal, antiparasitic, anthelmintic, and antiviral activities [57,58,59]. EOs from O. majorana and O. dubium have been reported to have antimalarial effects on mice, reducing the parasitemia and increasing their life span [57]. Also, EOs from O. majorana, O. virens, and O. vulgare have been reported to have anti-Phytomonas effects on P. davidi, with O. virens having the strongest effect [22]. Other antileishmanial effects were reported from O. virens EO on L. infantum [42], and O. vulgare EO on L. amazonensis, L. panamensis, and L. braziliensis [60,61].
In our study, thymol (1) and carvacrol (2) correlated with AP activity, and were active when tested on P. davidi, while p-cymene was not. Thymol and carvacrol are p-cymene derivatives, and therefore their amounts in EOs are usually correlated, explaining the correlation of this compound with AP effects. Compounds 1 and 2 are common in EOs with activity against promastigotes and amastigotes of L. infantum [29]. Thymol has been reported to have anti-Phytomonas [22] and leishmanicidal properties on promastigotes of L. infantum [29], promastigotes and amastigotes of L. infantum, and amastigotes of L. donovani [62]. The anti-Leishmania and anti-Phytomonas activities of carvacrol and thymol depend on the presence of the phenolic hydroxyl group, as observed before by Silva et al. [63]. The lack of the phenolic hydroxyl group in the precursor p-cymene is associated with the absence of antiprotozoal effects. Also, thymol has been used as a starting compound to obtain derivatives with stronger antileishmanial effects [64].
The biological properties of EOs can be determined by their major compounds, but minor compounds modulate these effects. Synergistic effects between EO components have been observed before. A mixture 1:4 of lupenone and β-caryophyllene oxide presented better antileishmanial activity and lower cytotoxicity than β-caryophyllene oxide alone [65]. Also, the combination between ascaridole and carvacrol produced synergistic effects on L. amazonensis [66].

5. Conclusions

EOs with better anti-kinetoplastid activity were S. montana, T. vulgaris, M. suaveolens, and L. luisieri. They are good sources of thymol, carvacrol, trans-α-necrodyl acetate, and piperitenone oxide. Further studies should be performed with trans-α-necrodyl acetate and piperitenone oxide to corroborate their potential activity against L. infantum and P. davidi. Thymol and carvacrol were the best anti-kinetoplastid compounds of this study. The synthesis of new compounds, using carvacrol and thymol as starting compounds, could be a strategy for the search for new anti-trypanosomatid compounds as alternatives to the current treatments, due to their anti-Phytomonas and antileishmanial effects.
Phytomonas is a plant parasite that can cause important economic losses and that lacks an effective treatment. Our study identifies potential treatments against this pathogen, and extraction methods which potentiate the concentration of specific components. The potential use of EOs, and their main components, as new alternatives for the treatment of animal trypanosomatid diseases, including leishmaniasis, could lie in their use as alternative treatments or in combination with approved treatments, to increase the efficiency and diminish the toxic effects of the current therapeutic protocols.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/molecules28031467/s1, Table S1. Plant species selected, localization, coordinates, and voucher numbers. Table S2. Chemical composition of the EOs from the most active species: L. luisieri (1 and 2) (Ll1 and Ll2), M. suaveolens (Ms); S. hybrid (Sh), S. montana (Sm), T. vulgaris (Tv), and T. zygis (Tz). RI, Retention index; RT, Retention time; HD, EOs obtained by hydrodistillation; SD, EOs obtained by steam distillation. Figure S1. Percentage of antiprotozoal activity of EOs obtained by hydrodistillation (HD) and steam distillation (SD) from L. x intermedia “Abrial” (Lab), L. x intermedia “Grosso” (Lg), L. x intermedia “Super” (Lsu), L. lanata (Ll), L. angustifolia (La), L. mallete (Lm), T. mastichina (Tm), O. virens (Ov), S. blancoana (Sb), S. officinalis (So), S. sclarea (Ss), R. officinalis (Ro), S. chamaecyparissus (Sc), T. vulgare (Tav), D. graveolens (Dg). A. Anti-Leishmania activity. B. Anti-Phytomonas activity of EOs. Figure S2. Chromatograms of the most active species EOs: L. luisieri (1 and 2) (Ll1 and Ll2), M. suaveolens (Ms); S. hybrid (Sh), S. montana (Sm), T. vulgaris (Tv), and T. zygis (Tz); HD, EOs obtained by hydrodistillation; SD, EOs obtained by steam distillation.

Author Contributions

Conceptualization: A.G.-C., R.A.M.-D., M.B. and M.T.G.-M.; methodology: C.I., M.Q., J.N.-R., E.O., A.G.-C., R.A.M.-D., M.B. and M.T.G.-M.; formal analysis: C.I., M.Q., M.B. and M.T.G.-M.; investigation: C.I., M.Q., J.N.-R. and E.O.; resources: A.G.-C., J.N.-R., E.O., R.A.M.-D., M.B. and M.T.G.-M.; data curation: A.G.-C., M.B. and M.T.G.-M.; writing original draft preparation: C.I., M.Q. and M.B.; writing, review and editing: A.G.-C., M.B. and M.T.G.-M.; supervision: A.G.-C., M.T.G.-M. and M.B.; funding acquisition: A.G.-C., R.A.M.-D. and M.T.G.-M. All authors have read and agreed to the published version of the manuscript.

Funding

This work has been partially financed by Grants PR108/20-08 (Santander-UCM), PID2020-114207RB-I00 and PID2019-106222RB-C31/SRA (Spanish Ministry of Science and Innovation).

Data Availability Statement

Not applicable.

Acknowledgments

This article is dedicated to RM-D who sadly passed away in 2021. We want to thank Jose María Requena for his generous donation of L. infantum promastigotes and Rubén Muñoz for his kind assistance in conducting identification of chemical components of essential oils.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Samples of the compounds… are available from the authors.

References

  1. Stuart, K.; Brun, R.; Croft, S.; Fairlamb, A.; Gürtler, R.E.; McKerrow, J.; Reed, S.; Tarleton, R. Kinetoplastids: Related protozoan pathogens, different diseases. J. Clin. Investig. 2008, 118, 1301. [Google Scholar] [CrossRef]
  2. Sunter, J.; Gull, K. Shape, form, function and Leishmania pathogenicity: From textbook descriptions to biological understanding. Open Biol. 2017, 7, 170165. [Google Scholar] [CrossRef]
  3. Yurchenko, V.; Butenko, A.; Kostygov, A.Y. Genomics of Trypanosomatidae: Where we stand and what needs to be done? Pathogens 2021, 10, 1124. [Google Scholar] [CrossRef]
  4. Jaskowska, E.; Butler, C.; Preston, G.; Kelly, S. Phytomonas: Trypanosomatids adapted to plant environments. PLoS Pathog. 2015, 11, e1004927. [Google Scholar] [CrossRef]
  5. Georgiadou, S.P.; Makaritsis, K.P.; Dalekos, G.N. Leishmaniasis revisited: Current aspects on epidemiology, diagnosis and treatment. J. Transl. Int. Med. 2015, 3, 43–50. [Google Scholar] [CrossRef]
  6. Torres-Guerrero, E.; Quintanilla-Cedillo, M.R.; Ruiz-Esmenjaud, J.; Arenas, R. Leishmaniasis: A review. F1000Research 2017, 6, 750. [Google Scholar] [CrossRef]
  7. Sasidharan, S.; Saudagar, P. Leishmaniasis: Where are we and where are we heading? Parasitol. Res. 2021, 120, 1541–1554. [Google Scholar] [CrossRef]
  8. Santos, A.L.S.; d’Avila-Levy, C.M.; Elias, C.G.R.; Vermelho, A.B.; Branquinha, M.H. Phytomonas serpens: Immunological similarities with the human Trypanosomatid pathogens. Microbes Infect. 2007, 9, 915–921. [Google Scholar] [CrossRef]
  9. Lopes, A.H.; Souto-Padrón, T.; Dias, F.A.; Gomes, M.T.; Rodrigues, G.C.; Zimmermann, L.T.; Alves e Silva, T.L.; Vermelho, A.B. Trypanosomatids: Odd organisms, devastating diseases. Open Parasitol. J. 2010, 4, 30–59. [Google Scholar] [CrossRef]
  10. Ginsburg, H.; Deharo, E. A Call for using natural compounds in the development of new antimalarial treatments—An introduction. Malar. J. 2011, 10, S1. [Google Scholar] [CrossRef] [Green Version]
  11. Figueiredo, A.C.; Barroso, J.G.; Pedro, L.G.; Scheffer, J.J.C. Factors affecting secondary metabolite production in plants: Volatile components and essential oils. Flavour Fragr. J. 2008, 23, 213–226. [Google Scholar] [CrossRef]
  12. Rehman, R.; Hanif, M.A.; Mushtaq, Z.; Al-Sadi, A.M. Biosynthesis of essential oils in aromatic plants: A review. Food Rev. Int. 2016, 32, 117–160. [Google Scholar] [CrossRef]
  13. Tongnuanchan, P.; Benjakul, S. Essential oils: Extraction, bioactivities, and their uses for food preservation. J. Food Sci. 2014, 79, R1231–R1249. [Google Scholar] [CrossRef]
  14. Fampa, P.; Florencio, M.; Santana, R.C.; Rosa, D.; Soares, D.C.; Leonel De Matos Guedes, H.; Cordeiro Da Silva, A.; Siqueira, D.; Chaves, A.; Pinto-Da-Silva, L.H. Anti-Leishmania effects of volatile oils and their isolates. Rev. Bras. Farmacogn. 2021, 31, 561–578. [Google Scholar] [CrossRef]
  15. Pavela, R.; Benelli, G.; Pavoni, L.; Bonacucina, G.; Cespi, M.; Cianfaglione, K.; Bajalan, I.; Morshedloo, M.R.; Lupidi, G.; Romano, D.; et al. Microemulsions for delivery of apiaceae essential oils—Towards highly effective and eco-friendly mosquito larvicides? Ind. Crops Prod. 2019, 129, 631–640. [Google Scholar] [CrossRef]
  16. Bunse, M.; Daniels, R.; Gründemann, C.; Heilmann, J.; Kammerer, D.R.; Keusgen, M.; Lindequist, U.; Melzig, M.F.; Morlock, G.E.; Schulz, H.; et al. essential oils as multicomponent mixtures and their potential for human health and well-being. Front. Pharmacol. 2022, 13, 2645. [Google Scholar] [CrossRef]
  17. Andrade, M.A.; Azevedo, C.D.S.; Motta, F.N.; Santos, M.L.D.; Silva, C.L.; Santana, J.M.D.; Bastos, I.M.D. Essential Oils: In vitro activity against Leishmania amazonensis, cytotoxicity and chemical composition. BMC Complement. Altern. Med. 2016, 16, 444. [Google Scholar] [CrossRef]
  18. Kalaivani, K.; Senthil-Nathan, S.; Murugesan, A.G. biological activity of selected Lamiaceae and Zingiberaceae plant essential oils against the dengue vector Aedes aegypti L. (Diptera: Culicidae). Parasitol. Res. 2012, 110, 1261–1268. [Google Scholar] [CrossRef]
  19. Vieira-Araújo, F.M.; Macedo Rondon, F.C.; Pinto Vieira, Í.G.; Pereira Mendes, F.N.; Carneiro de Freitas, J.C.; Maia de Morais, S. Sinergism between alkaloids piperine and capsaicin with meglumine antimoniate against Leishmania infantum. Exp. Parasitol. 2018, 188, 79–82. [Google Scholar] [CrossRef]
  20. Hammi, K.M.; Essid, R.; Tabbene, O.; Elkahoui, S.; Majdoub, H.; Ksouri, R. Antileishmanial activity of Moringa oleifera leaf extracts and potential synergy with Amphotericin B. S. Afr. J. Bot. 2020, 129, 67–73. [Google Scholar] [CrossRef]
  21. Abad, M.J.; Bedoya, L.M.; Apaza, L.; Bermejo, P. The Artemisia L. Genus: A review of bioactive essential oils. Molecules 2012, 17, 2542–2566. [Google Scholar] [CrossRef]
  22. Guzmán, M.; González-Coloma, A.; Fe Andrés, M.; Navarro-Rocha, J.; Martínez-Díaz, R.A. Biological evaluation of essential oils from selected medicinal plants and their main components against Phytomonas davidi (Kinetoplastea: Trypanosomatidae). Chem. Biodivers. 2020, 17, e2000521. [Google Scholar] [CrossRef]
  23. Monzote, L.; Geroldinger, G.; Tonner, M.; Scull, R.; de Sarkar, S.; Bergmann, S.; Bacher, M.; Staniek, K.; Chatterjee, M.; Rosenau, T.; et al. Interaction of ascaridole, carvacrol, and caryophyllene oxide from essential oil of Chenopodium ambrosioides L. with mitochondria in Leishmania and other eukaryotes. Phytother. Res. 2018, 32, 1729–1740. [Google Scholar] [CrossRef]
  24. Bailén, M.; Díaz-Castellanos, I.; Azami-Conesa, I.; Alonso Fernández, S.; Martínez-Díaz, R.A.; Navarro-Rocha, J.; Gómez-Muñoz, M.T.; González-Coloma, A. Anti-Trichomonas gallinae activity of essential oils and main compounds from Lamiaceae and Asteraceae plants. Front. Vet. Sci. 2022, 9, 981763. [Google Scholar] [CrossRef]
  25. Julio, L.F.; Barrero, A.F.; Herrador Del Pino, M.M.; Arteaga, J.F.; Burillo, J.; Andres, M.F.; Díaz, C.E.; González-Coloma, A. Phytotoxic and nematicidal components of Lavandula luisieri. J. Nat. Prod. 2016, 79, 261–266. [Google Scholar] [CrossRef]
  26. Postell, F.J.; Mcghee, R.B. An Ultrastructural study of Phytomonas davidi Lafont (Trypanosomatidae)1. J. Protozool. 1981, 28, 78–83. [Google Scholar] [CrossRef]
  27. Sainz, P.; Andrés, M.F.; Martínez-Díaz, R.A.; Bailén, M.; Navarro-Rocha, J.; Díaz, C.E.; González-Coloma, A. Chemical composition and biological activities of Artemisia pedemontana subsp. assoana essential oils and hydrolate. Biomolecules 2019, 9, 558. [Google Scholar] [CrossRef]
  28. Escobar, P.; Leal, S.M.; Herrera, L.V.; Martinez, J.R.; Stashenko, E. Chemical composition and antiprotozoal activities of Colombian lippia spp. essential oils and their major components. Mem. Inst. Oswaldo Cruz 2010, 105, 184–190. [Google Scholar] [CrossRef]
  29. Youssefi, M.R.; Moghaddas, E.; Tabari, M.A.; Moghadamnia, A.A.; Hosseini, S.M.; Hosseini Farash, B.R.; Ebrahimi, M.A.; Mousavi, N.N.; Fata, A.; Maggi, F.; et al. In vitro and in vivo effectiveness of carvacrol, thymol and linalool against Leishmania infantum. Molecules 2019, 24, 2072. [Google Scholar] [CrossRef]
  30. Leal, S.M.; Pino, N.; Stashenko, E.E.; Martínez, J.R.; Escobar, P. Antiprotozoal activity of essential oils derived from Piper spp. grown in Colombia. J. Essent. Oil Res. 2013, 25, 512–519. [Google Scholar] [CrossRef]
  31. Zaman, W.; Ye, J.; Saqib, S.; Liu, Y.; Shan, Z.; Hao, D.; Chen, Z.; Xiao, P. Predicting potential medicinal plants with phylogenetic topology: Inspiration from the research of traditional Chinese medicine. J. Ethnopharmacol. 2021, 281, 114515. [Google Scholar] [CrossRef]
  32. Moraes Neto, R.N.; Setúbal, R.F.B.; Higino, T.M.M.I.; Brelaz-De-Castro, M.C.A.; da Silva, L.C.N.; dos Santos Aliança, A.S. Asteraceae plants as sources of compounds against leishmaniasis and Chagas disease. Front. Pharmacol. 2019, 10, 477. [Google Scholar] [CrossRef]
  33. Silva, K.P.; de Carvalho Santos, T.A.; Moutinho, B.L.; da Silva, R.S.; dos Santos Pinto, V.; Blank, A.F.; Corrêa, C.B.; Scher, R.; Fernandes, R.P.M. Using Varronia curassavica (Cordiaceae) essential oil for the biocontrol of Phytomonas serpens. Ind. Crops Prod. 2019, 139, 111523. [Google Scholar] [CrossRef]
  34. Pereira, K.L.G.; Nizio, D.A.D.C.; de Lima, P.C.N.; Fernandes, R.P.M.; Arrigoni-Blank, M.D.F.; de Sá Filho, J.C.F.; Nascimento, L.F.D.A.; de Souza, V.T.; Silva, K.P.; Blank, A.F. Seasonal variance in the chemical composition of essential oils from Lantana camara accessions and their trypanocidal activity on Phytomonas serpens. Bol. Latinoam. Caribe Plantas Med. Aromat. 2022, 21, 737–756. [Google Scholar] [CrossRef]
  35. Ben Hamida, N.; Martínez-Díaz, R.A.; Hela, M.; Msaada, K.; Ouerghi, Z.; Andres, M.F.; González-Coloma, A. Effect of salinity on the antiparasitic activity of hyssop essential oil. J. Essent. Oil Res. 2020, 32, 74–83. [Google Scholar] [CrossRef]
  36. Božovic, M.; Pirolli, A.; Ragno, R. Mentha suaveolens Ehrh. (Lamiaceae) essential oil and its main constituent piperitenone oxide: Biological activities and chemistry. Molecules 2015, 20, 8605–8633. [Google Scholar] [CrossRef]
  37. Menezes, L.R.A.; Santos, N.N.; Meira, C.S.; Ferreira Dos Santos, J.A.; Guimarães, E.T.; Soares, M.B.P.; Nepel, A.; Barison, A.; Costa, E.V. A new source of (R)-limonene and rotundifolone from leaves of Lippia pedunculosa (Verbenaceae) and their trypanocidal properties. Nat. Prod. Commun. 2014, 9, 737–739. [Google Scholar] [CrossRef]
  38. Matos-Rocha, T.J.; dos Santos Cavalcanti, M.G.; Barbosa-Filho, J.M.; Lúcio, A.S.S.C.; Veras, D.L.; Feitosa, A.P.S.; de Siqueira Júnior, J.P.; de Almeida, R.N.; Marques, M.O.M.; Alves, L.C.; et al. In vitro evaluation of schistosomicidal activity of essential oil of Mentha x villosa and some of its chemical constituents in adult worms of Schistosoma mansoni. Planta Med. 2013, 79, 1307–1312. [Google Scholar] [CrossRef]
  39. Lima, T.C.; da Silva, T.K.M.; Silva, F.L.; Barbosa-Filho, J.M.; Marques, M.O.M.; Santos, R.L.C.; Cavalcanti, S.C.D.H.; de Sousa, D.P. Larvicidal activity of Mentha x villosa Hudson essential oil, rotundifolone and derivatives. Chemosphere 2014, 104, 37–43. [Google Scholar] [CrossRef]
  40. Ibrahim, S.R.M.; Abdallah, H.M.; Mohamed, G.A.; Farag, M.A.; Alshali, K.Z.; Alsherif, E.A.; Ross, S.A. Volatile oil profile of some Lamiaceous plants growing in Saudi Arabia and their biological activities. J. Biosci. 2017, 72, 35–41. [Google Scholar] [CrossRef]
  41. Bouyahya, A.; Et-Touys, A.; Bakri, Y.; Talbaui, A.; Fellah, H.; Abrini, J.; Dakka, N. Chemical composition of Mentha pulegium and Rosmarinus officinalis essential oils and their antileishmanial, antibacterial and antioxidant activities. Microb. Pathog. 2017, 111, 41–49. [Google Scholar] [CrossRef]
  42. Machado, M.; Santoro, G.; Sousa, M.C.; Salgueiro, L.; Cavaleiro, C. Activity of essential oils on the growth of Leishmania infantum promastigotes. Flavour Fragr. J. 2010, 25, 156–160. [Google Scholar] [CrossRef]
  43. Zheljazkov, V.D.; Cantrell, C.L.; Astatkie, T.; Hristov, A. Yield, content, and composition of peppermint and spearmints as a function of harvesting time and drying. J. Agric. Food Chem. 2010, 58, 11400–11407. [Google Scholar] [CrossRef]
  44. Zarenezhad, E.; Agholi, M.; Ghanbariasad, A.; Ranjbar, A.; Osanloo, M. A nanoemulsion-based nanogel of citrus limon essential oil with leishmanicidal activity against Leishmania tropica and Leishmania major. J. Parasit. Dis. 2021, 45, 441–448. [Google Scholar] [CrossRef]
  45. Ebadollahi, A.; Jalali Sendi, J.; Ziaee, M.; Krutmuang, P. Acaricidal, insecticidal, and nematicidal efficiency of essential oils isolated from the Satureja genus. Int. J. Environ. Res. Public Health 2021, 18, 6050. [Google Scholar] [CrossRef]
  46. Jafari, F.; Farmani, F.; Zomorodian, K.; Moein, M.; Faridi, P.; Zarshenas, M.M. A Study on essential oil chemical compositions, antioxidant, and antimicrobial activities of native and endemic Satureja species growing in Iran. Pharm. Chem. J. 2018, 52, 63–68. [Google Scholar] [CrossRef]
  47. Tepe, B.; Cilkiz, M.A. Pharmacological and phytochemical overview on Satureja. Pharm. Biol. 2015, 54, 375–412. [Google Scholar] [CrossRef]
  48. Faria, J.M.S.; Rodrigues, A.M.; Sena, I.; Moiteiro, C.; Bennett, R.N.; Mota, M.; Figueiredo, A.C. Bioactivity of Ruta graveolens and Satureja montana essential oils on Solanum tuberosum hairy roots and Solanum tuberosum hairy roots with Meloidogyne chitwoodi co-cultures. J. Agric. Food Chem. 2016, 64, 7452–7458. [Google Scholar] [CrossRef]
  49. Navarro-Rocha, J.; Andrés, M.F.; Díaz, C.E.; Burillo, J.; González-Coloma, A. Composition and biocidal properties of essential oil from pre-domesticated Spanish Satureja montana. Ind. Crops Prod. 2020, 145, 111958. [Google Scholar] [CrossRef]
  50. Kheirandish, F.; Chegeni, R.; Delfan, B.; Jabari, M.; Ebrahimzadeh, F.; Rashidipour, M. The cytotoxic and antileishmanial effects of Satureja khuzestanica essential oil. Herb. Med. J. 2016, 1, 11–17. [Google Scholar] [CrossRef]
  51. Mohammadpour, G.; Marzony, E.T.; Farahmand, M. Evaluation of the anti-Leishmania major activity of Satureja bakhtiarica essential oil in vitro. Nat. Prod. Commun. 2012, 7, 133–136. [Google Scholar] [CrossRef]
  52. Tariku, Y.; Hymete, A.; Hailu, A.; Rohloff, J. Essential-oil composition, antileishmanial, and toxicity study of Artemisia abyssinica and Satureja punctata ssp. punctata from Ethiopia. Chem. Biodivers. 2010, 7, 1009–1018. [Google Scholar] [CrossRef]
  53. Machado, M.; Martins, N.; Salgueiro, L.; Cavaleiro, C.; Sousa, M.C. Lavandula luisieri and Lavandula viridis essential oils as upcoming anti-protozoal agents: A key focus on leishmaniasis. Appl. Sci. 2019, 9, 3056. [Google Scholar] [CrossRef]
  54. Shokri, A.; Saeedi, M.; Fakhar, M.; Morteza-Semnani, K.; Keighobadi, M.; Teshnizi, S.H.; Kelidari, H.R.; Sadjadi, S. Antileishmanial activity of Lavandula angustifolia and Rosmarinus officinalis essential oils and nano-emulsions on Leishmania major (MRHO/IR/75/ER). Iran. J. Parasitol. 2017, 12, 622–631. [Google Scholar]
  55. Bouyahya, A.; Et-Touys, A.; Abrini, J.; Talbaoui, A.; Fellah, H.; Bakri, Y.; Dakka, N. Lavandula stoechas essential oil from Morocco as novel source of antileishmanial, antibacterial and antioxidant activities. Biocatal. Agric. Biotechnol. 2017, 12, 179–184. [Google Scholar] [CrossRef]
  56. Machado, M.; Dinis, A.M.; Santos-Rosa, M.; Alves, V.; Salgueiro, L.; Cavaleiro, C.; Sousa, M.C. Activity of Thymus capitellatus volatile extract, 1,8-cineole and borneol against Leishmania Species. Vet. Parasitol. 2014, 200, 39–49. [Google Scholar] [CrossRef]
  57. Güler, E.; Özbilgin, A.; Becer, E.; Hanoğlu, A.; Şanlidağ, T. An endemic plant of Cyprus, Origanum majorana: Is it a new alternative natural product for malaria treatment? Mikrobiyol. Bul. 2020, 54, 463–478. [Google Scholar] [CrossRef]
  58. Méabed, E.M.H.; El- Sayed, N.M.; Abou-Sreea, A.I.B.; Roby, M.H.H. Chemical analysis of aqueous extracts of Origanum majorana and Foeniculum vulgare and their efficacy on Blastocystis spp. cysts. Phytomedicine 2018, 43, 158–163. [Google Scholar] [CrossRef]
  59. Babili, F.E.; Bouajila, J.; Souchard, J.P.; Bertrand, C.; Bellvert, F.; Fouraste, I.; Moulis, C.; Valentin, A. Oregano: Chemical analysis and evaluation of its antimalarial, antioxidant, and cytotoxic activities. J. Food Sci. 2011, 76, C512–C518. [Google Scholar] [CrossRef]
  60. Teles, A.M.; Rosa, T.D.D.S.; Mouchrek, A.N.; Abreu-Silva, A.L.; da Silva Calabrese, K.; Almeida-Souza, F. Cinnamomum zeylanicum, Origanum vulgare, and Curcuma longa essential oils: Chemical composition, antimicrobial and antileishmanial activity. Evid. Based Complement. Altern. Med. 2019, 2019, 2421695. [Google Scholar] [CrossRef]
  61. Sanchez-Suarez, J.F.; Riveros, I.; Delgado, G. Evaluation of the leishmanicidal and cytotoxic potential of essential oils derived from ten Colombian plants. Iran. J. Parasitol. 2013, 8, 129. [Google Scholar]
  62. Tasdemir, D.; Kaiser, M.; Demirci, B.; Demirci, F.; Hüsnü Can Baser, K. Antiprotozoal activity of Turkish Origanum onites essential oil and its components. Molecules 2019, 24, 4421. [Google Scholar] [CrossRef] [Green Version]
  63. Silva, A.R.S.T.; Scher, R.; Santos, F.V.; Ferreira, S.R.; Cavalcanti, S.C.H.; Correa, C.B.; Bueno, L.L.; Alves, R.J.; Souza, D.P.; Fujiwara, R.T.; et al. Leishmanicidal activity and structure-activity relationships of essential oil constituents. Molecules 2017, 22, 815. [Google Scholar] [CrossRef]
  64. De Morais, S.M.; Vila-Nova, N.S.; Bevilaqua, C.M.L.; Rondon, F.C.; Lobo, C.H.; de Alencar Araripe Noronha Moura, A.; Sales, A.D.; Rodrigues, A.P.R.; de Figuereido, J.R.; Campello, C.C.; et al. Thymol and eugenol derivatives as potential antileishmanial agents. Bioorg. Med. Chem. 2014, 22, 6250–6255. [Google Scholar] [CrossRef]
  65. Polanco-Hernández, G.M.; Giménez-Turba, A.; Salamanca, E.; Getti, G.; Rai, R.; Acosta-Viana, K.Y.; Ermilo Arana-Argáez, V.; Torres-Romero, J.C.; Fernández-Martín, K.G.; Segura-Campos, M.R.; et al. Leishmanicidal activity and immunomodulatory effect of a mixture of lupenone and β-caryophyllene oxide. Rev. Bras. Farmacogn. 2021, 31, 199–206. [Google Scholar] [CrossRef]
  66. Pastor, J.; García, M.; Steinbauer, S.; Setzer, W.N.; Scull, R.; Gille, L.; Monzote, L. Combinations of ascaridole, carvacrol, and caryophyllene oxide against Leishmania. Acta Trop. 2015, 145, 31–38. [Google Scholar] [CrossRef]
Figure 1. Percentage of antiprotozoal activity of EOs with IC50 < 200 µg/mL obtained by hydrodistillation (HD) and steam distillation (SD) from L. luisieri 1 and 2 (Ll1 and Ll2), T. vulgaris (Tv), T. zygis (Tz), O. majorana (Om), S. hybrid (So), S. montana (Sm), and M. suaveolens (Ms). (A) Anti-Leishmania activity. (B) Anti-Phytomonas activity of EOs.
Figure 1. Percentage of antiprotozoal activity of EOs with IC50 < 200 µg/mL obtained by hydrodistillation (HD) and steam distillation (SD) from L. luisieri 1 and 2 (Ll1 and Ll2), T. vulgaris (Tv), T. zygis (Tz), O. majorana (Om), S. hybrid (So), S. montana (Sm), and M. suaveolens (Ms). (A) Anti-Leishmania activity. (B) Anti-Phytomonas activity of EOs.
Molecules 28 01467 g001aMolecules 28 01467 g001b
Figure 2. Chemical structures of the tested monoterpenes.
Figure 2. Chemical structures of the tested monoterpenes.
Molecules 28 01467 g002
Table 1. Effects of the tested EOs from Lamiaceae and Asteraceae species on L. infantum and P. davidi (IC50) a.
Table 1. Effects of the tested EOs from Lamiaceae and Asteraceae species on L. infantum and P. davidi (IC50) a.
FamilyGenusSpeciesExtraction MethodL. infantum (IC50)P. davidi (IC50)
AsteraceaeTanacetumTanacetum vulgareHD325.5 (300.0, 353.1)>800
SantolinaSantolina chamaecyparissusHD451.2 (391.4–520.2)599.2 (580.7–618.3)
DitrichiaDitrichia graveolensHD350.8 (303.4–405.4)341.5 (288.8–403.8)
SD306.8 (246.7–381.5)>800
LamiaceaeLavandulaLavandula lanataHD>800>800
Lavandula luisieri 1HD>800572.5 (557.5–587.9)
SD<200439.5 (394.8–489.3)
Lavandula luisieri 2HD247.4 (227.7–268.8)573.9 (485.6–678.2)
SD74.3 (61.6–89.5)>800
Lavandula angustifoliaHD461.2 (402.2–528.8)>800
SD512.2 (477.1–550.0)>800
Lavandula x intermedia “Abrial”HD634.0 (528.5–760.7)>800
SD319.1 (294.8–345.3)>800
Lavandula x intermedia “Super”HD466.5 (404.2–538.3)343.8 (259.3–455.8)
SD640.7 (603.6–680.1)>800
Lavandula x intermedia “Grosso”HD>800496.6 (417.9–590.1)
SD633.8 (595.0–675.0)>800
Lavandula malleteHD397.9 (362.4–436.9436.5 (367.4–518.6)
SD293.2 (233.8–367.6)>800
OriganumOriganum majoranaHD547.9 (521.4–575.7)>800
Origanum virensHD781.2 (742.7–821.7)507.8 (486.8–529.7)
SD742.6 (664.7–829.6)521.9 (499.0–545.8)
RosmarinusRosmarinus officinalisHD718.9 (667.1–774.7)599.6 (495.4–725.7)
SD367.1 (307.8–437.7)446.5 (321.7–619.7)
SaturejaSatureja montanaHD170.9 (154.0–189.6)83.1 (67.2–102.7)
SD194.1 (176.7–213.3))49.7 (46.0–53.7)
MenthaMentha suaveolensHD117.9 (101.3–137.4)431.6 (404.5–460.6)
SD88.2 (73.2–106.4)396.0 (374.0–419.3)
SalviaSalvia officinalisHD680.8 (621.8–745.4)>800
SD<200329.3 (280.6–386.4)
Salvia hybridHD184.7 (154.8–220.3)406,3 (387.2–426,3)
Salvia blancoanaHD>800514.2 (474.1–557.7)
Salvia sclareaHD>800>800
ThymusThymus mastichinaHD743.7 (682.4–810.5)>800
Thymus vulgarisHD>80043.3 (33.5–56.0)
SD375.6 (331.8–425.1)426.4 (369.9–491.4)
Thymus zygisHD141.8 (99.1–202.8)>800
SD433.0 (394.9–474.7)182.3 (155.7–213.4)
a IC50 (μg/mL) = concentration needed to produce 50% promastigote mortality. SD: steam distillation. HD: hydrodistillation. Light grey: IC50 < 200 μg/mL; Dark grey: IC50: 200–500 μg/mL.
Table 2. Main components of the EOs from the most active species (abundance ≥ 5%).
Table 2. Main components of the EOs from the most active species (abundance ≥ 5%).
Plant SpeciesEMCompounds (% Relative Abundance)
M. suaveolensHDpiperitenone (53%), piperitenone oxide (23%), limonene (6%)
SDpiperitenone oxide (37%), piperitenone (21%), limonene (7%), germacrene D (7%), β -caryophyllene (6%)
L. luisieri 1 *HDfenchone (20%), camphor (13%), trans-α-necrodyl acetate (12%), lavandulyl acetate (6%), α-pinene (6%), 1,8-cineole (5%)
SDcamphor (35%), trans-α-necrodyl acetate (19%), lavandulyl acetate (6%), α-pinene (5%)
L. luisieri 2 *HDcamphor (49%), trans-α-necrodyl acetate (13%), lavandulol (6%)
SDtrans-α-necrodyl acetate (18%), lavandulol (8%), germacrene D (8%), camphor (5%)
S. hybrid * HD1,8-cineole (21%), camphor (14%), trans-bornyl acetate (13%), β-pinene (11%), camphene (7%)
S. montana * HDcarvacrol (33%), p-cymene (18%), thymol (17%), γ-terpinene (12%)
SDcarvacrol (41%), p-cymene (12%), γ-terpinene (12%), thymol (7%), β-caryophyllene (6%)
T. vulgaris * HDthymol (43%), p-cymene (22%), γ-terpinene (6%)
SDcarvacrol (41%), p-cymene (31%), thymol (28%), β-caryophyllene (6%)
T. zygis * HDthymol (39%), p-cymene (18%), γ-terpinene (9%), linalool (5%), borneol (6%), carvacrol (5%)
SDthymol (21%), linalyl acetate (18%), linalool (12%), p-cymene (8%), carvacrol (5%), α-bisabolol (5%)
EM: extraction method; HD: EOs obtained by hydrodistillation; SD: EOs obtained by steam distillation; *: previously reported [25].
Table 3. Pearson correlation coefficients between main components of EOs and AL and AP activities.
Table 3. Pearson correlation coefficients between main components of EOs and AL and AP activities.
CompoundAL ActivityAP Activity
PCCpPCCp
p-cymene−0.0480.773−0.4410.005
α-terpineol0.4080.0100.2470.129
piperitenone oxide−0.3380.035−0.1460.377
thymol−00290.859−0.3960.013
carvacrol−02420.138−0.5170.001
PCC: Pearson correlation coefficient.
Table 4. Effects of the tested compounds on P. davidi, L. infantum, and Vero cells (IC50 and CC50 respectively).
Table 4. Effects of the tested compounds on P. davidi, L. infantum, and Vero cells (IC50 and CC50 respectively).
CompoundVero Cells [25]P. davidiL. infantum
CC50 (µg/mL) aIC50 (µg/mL) bSI cIC50 (µg/mL) bSI c
thymol (1)≈10045.0 (38.4–52.7)2.227.22 (6.22–8.62) [29]13.85
carvacrol (2)>10079.1 (71.9–86.9)1.269.8 (8.51–11.7) [29]10.20
α-terpineol (3)>100>1001nd-
p-cymene (4)>100>1001>100 [28]0.67
amphotericin B-0.2 (0.1–0.2)-0.01 [30]10.000
a CC50 (μg/mL) = concentration needed to produce 50% Vero cell mortality; b IC50 (μg/mL) = concentration needed to produce 50% trophozoite mortality, c SI: Selectivity index.; nd: not determined.
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

Bailén, M.; Illescas, C.; Quijada, M.; Martínez-Díaz, R.A.; Ochoa, E.; Gómez-Muñoz, M.T.; Navarro-Rocha, J.; González-Coloma, A. Anti-Trypanosomatidae Activity of Essential Oils and Their Main Components from Selected Medicinal Plants. Molecules 2023, 28, 1467. https://doi.org/10.3390/molecules28031467

AMA Style

Bailén M, Illescas C, Quijada M, Martínez-Díaz RA, Ochoa E, Gómez-Muñoz MT, Navarro-Rocha J, González-Coloma A. Anti-Trypanosomatidae Activity of Essential Oils and Their Main Components from Selected Medicinal Plants. Molecules. 2023; 28(3):1467. https://doi.org/10.3390/molecules28031467

Chicago/Turabian Style

Bailén, María, Cristina Illescas, Mónica Quijada, Rafael Alberto Martínez-Díaz, Eneko Ochoa, María Teresa Gómez-Muñoz, Juliana Navarro-Rocha, and Azucena González-Coloma. 2023. "Anti-Trypanosomatidae Activity of Essential Oils and Their Main Components from Selected Medicinal Plants" Molecules 28, no. 3: 1467. https://doi.org/10.3390/molecules28031467

Article Metrics

Back to TopTop