Next Article in Journal
Ultrasonic Assisted-Reflux Synergistic Extraction of Camptothecin and Betulinic Acid from Camptotheca acuminata Decne. Fruits
Next Article in Special Issue
Myrcia splendens (Sw.) DC. (syn. M. fallax (Rich.) DC.) (Myrtaceae) Essential Oil from Amazonian Ecuador: A Chemical Characterization and Bioactivity Profile
Previous Article in Journal
Labradorins with Antibacterial Activity Produced by Pseudomonas sp.
Previous Article in Special Issue
The Effects of Selected Sesquiterpenes from Myrica rubra Essential Oil on the Efficacy of Doxorubicin in Sensitive and Resistant Cancer Cell Lines
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

In Vitro Anti-Leishmanial Activity of Essential Oils Extracted from Vietnamese Plants

by
Thanh Binh Le
1,2,*,
Claire Beaufay
1,
Duc Trong Nghiem
3,
Marie-Paule Mingeot-Leclercq
4 and
Joëlle Quetin-Leclercq
1,*
1
GNOS Research Group, Louvain Drug Research Institue, Université Catholique de Louvain, Bruxelles 1200, Belgium
2
Department of Pharmacognosy, Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hoan Kiem, Hanoi 100000, Vietnam
3
Department of Botany, Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hoan Kiem, Hanoi 100000, Vietnam
4
TFAR Research Group, Louvain Drug Research Institue, Université Catholique de Louvain, Bruxelles 1200, Belgium
*
Authors to whom correspondence should be addressed.
Molecules 2017, 22(7), 1071; https://doi.org/10.3390/molecules22071071
Submission received: 4 May 2017 / Revised: 21 June 2017 / Accepted: 22 June 2017 / Published: 27 June 2017
(This article belongs to the Special Issue Essential Oils: Chemistry and Bioactivity)

Abstract

:
Leishmania mexicana is one of the pathogens causing cutaneous leishmaniasis which is associated with patient morbidity. In our researches for new safe and effective treatments, thirty-seven essential oils (EOs) extracted from Vietnamese plants were screened in vitro for the first time on Leishmania mexicana mexicana (Lmm) promastigotes at the maximum concentration of 50 nL/mL. Active EOs were also analyzed for cytotoxicity on mammalian cell lines (WI38, J774) and their selectivity indices (SI) were calculated. Their composition was determined by GC-MS and GC-FID. Our results indicated that EOs extracted from Cinnamomum cassia, Zingiber zerumbet, Elsholtzia ciliata and Amomum aromaticum, possessed a moderate anti-leishmanial activity, with IC50 values of 2.92 ± 0.08, 3.34 ± 0.34, 8.49 ± 0.32 and 9.25 ± 0.64 nL/mL respectively. However, they also showed cytotoxicity with SI < 10. The most promising EO was extracted from Ocimum gratissimum, displaying an IC50 of 4.85 ± 1.65 nL/mL and SI > 10. It contained 86.5% eugenol, which was demonstrated to be effective on Lmm with IC50 of 2.57 ± 0.57 nL/mL and not toxic on mammalian cells, explaining the observed activity.

1. Introduction

Cutaneous leishmaniasis (CL), the most common form of leishmaniasis, largely affects poor and developing countries in Africa, the Mediterranean Basin, the Middle East, central Asia (the Old World) and South America (the New World). The estimated number of new CL cases worldwide ranges from 0.7 million to 1.3 million annually [1].
CL is caused by protozoan parasites of the Leishmania (L.) genus. Infected female phlebotomine sand flies inject these parasites from their proboscis to human when they take their blood meals. Skin lesions, the clinical signs of CL, develop within several weeks or months after exposure. These lesions are sometimes self-healing without treatment, or become chronic. In cases of ulcers healing, they leave permanent deep scars, which often cause a serious social prejudice. Conversely, if the disease becomes chronic, there is a high risk of severe bacterial infection. CL in the New World, mainly caused by L. mexicana, L. amazonensis, L. venezuelensis, L. braziliensis, L. guyanensis, L. panamensis, and L. peruviana, is usually more severe and lasts longer than that in the Old World, which is caused by L. tropica, L. major, L. aethiopica, L. infantum and L. donovani. Moreover, CL due to L. mexicana, L. amazonensis and L. panamensis can develop to diffuse forms. Lesions can spread from the skin to the mucosal surfaces of the nose or the mouth, leading to chronic stuffiness, bleeding, and inflamed mucosa or sores. In advanced cases, it can lead to the ulcerative destruction of the nose (such as perforation of the nasal septum), mouth, and pharynx [1,2].
Drugs are the only therapeutic option for the treatment of CL as there is no vaccine at the moment. However, existing drugs, such as pentavalent antimonials and miltefosine, have serious drawbacks in terms of safety, efficacy, cost and difficulty in administration [3,4]. Moreover, diffuse CL initially responding to the standard treatment relapses and becomes unresponsive to further treatments [2]. Research on a safer, more effective, and shorter-course treatment for CL is therefore urgent. Nevertheless, the development of new medicines for treatment of this disease is challenging because of the variety of parasite species, pathology and immune responses [2].
Essential oils (EOs), also known as volatile oils, are natural products formed by a mixture of volatile compounds produced by many plants. They are known to be biologically active, mainly possessing antibacterial, antifungal, and antioxidant properties. There are more and more studies on EOs biological activity because they are usually devoid of long-term genotoxic risks [5]. Interestingly, topical application, the most frequent way to administer EOs, is recommended to be explored for the development of new anti-leishmanial drugs targeting CL because of their effectiveness and safety [2]. Several EOs showed interesting potential to be used as new anti-parasitic drugs such as those extracted from Chenopodium ambrosioides L. (a synonym of Dysphania ambrosioides (L.) Mosyakin & Clemant) [6,7] or Bixa orellana L. [8]. However, EOs are complex mixtures of several compounds and their compositions vary according to many factors, such as plant’s environment and growing conditions, methods of harvesting, extraction and storage. Moreover, the major component of an EO can also vary in different chemotypes of the same plant species. This chemical variability can influence their activities or adverse effects. Therefore, a clear knowledge of the EO composition is necessary [9].
With the aim of discovering new natural products against Leishmania mexicana mexicana and developing active EOs or compounds for topical application, 37 Vietnamese plants were selected for anti-leishmanial investigation in our work. It is the first time that these Vietnamese EOs were tested on this Leishmania species. Moreover, this study also presents the results of chemical composition analyses of the most interesting samples.

2. Results

A primary screening was performed at concentrations of 50 and 25 nL/mL for the 37 EOs. The results showed that among them five EOs, extracted from Amomum aromaticum, Cinnamomum cassia, Elsholtzia ciliata, Ocimum gratissimum, and Zingiber zerumbet, displayed a promising effect against Leishmania mexicana mexicana (Lmm) promastigotes with less than 1% viable parasites at the lower concentration tested (Table 1).
These selected EOs were further analyzed for dose-response activity to calculate IC50 value and for cytotoxicity on mammalian cells. The results are summarized in Table 2. All selected EOs revealed high potential against promastigote form of Lmm with IC50 values lower than 10 nL/mL. However, four of them also showed toxicity on mammalian cells, as indicated by their SI < 10. Our data demonstrated that the most active and selective EO was extracted from Ocimum gratissimum with an IC50 value on Lmm promastigotes of 4.85 ± 1.65 nL/mL and no toxicity towards mammalian cells at the highest tested concentration (50 nL/mL), as evidenced by more than 80% viable cells after 72 h of incubation.
To define their composition and further determine active compounds, the five selected EOs were analyzed by GC-MS and GC-FID (Table 3). Identified compounds accounted for more than 90% for each EO. Eucalyptol (55.2%), trans-cinnamaldehyde (83.6%), citral (neral and geranial) (40.2%), eugenol (86.5%), and zerumbone (60.3%) were characterized as the main components of EOs extracted from A. aromaticum, C. cassia, E. ciliata, O. gratissimum, and Z. zerumbet, respectively.
It is important to point out that eugenol is the major compound (86.5%) of the EO extracted from O. gratissimum, which was shown to be the most selective active sample. In order to understand the activity of this EO, in vitro anti-leishmanial activity and cytotoxicity of eugenol were studied. Interestingly, the IC50 value against Lmm promastigotes of eugenol was 2.57 ± 0.57 nL/mL (=2.72 µg/mL, 15.67 µM) and more than 80% of mammalian cells were living after 72 h when incubated at the maximal concentration used (50 nL/mL).

3. Discussion

This study analyzed the in vitro activity of 37 EOs extracted from Vietnamese plants against Leishmania mexicana mexicana. According to the classification of anti-parasitic activity from literature, extracts that showed effect against parasites with IC50 value ≤2 µg/mL (or 2 µM for pure compounds) are cited as having good activity, those with IC50 between 2 and 20 µg/mL (or micro molar for pure compounds) are considered as having a moderate activity, while those with higher IC50 value were considered as less interesting [11]. In this study, the primary screening was performed against Lmm promastigotes at concentrations of 25 and 50 nL/mL in order to quickly identify interesting EOs (1 nL is a little less than 1 µg depending on the density of the EO) [11]. Our data showed that percentages of viable parasites incubated with the lower concentration of five EOs, extracted from A. aromaticum, C. cassia, E. ciliata, O. gratissimum, and Z. zerumbet, were lower than 1%, meaning that these EOs revealed a potential activity. Indeed, the IC50 values of these selected EOs were lower than 10 nL/mL in the second analysis.
To have a general look at anti-leishmanial activity and cytotoxicity of the five more active EOs and to understand their observed effects, we compiled literature data of EOs extracted from similar plant species and also of their major components in Table 4.
As shown in this table, among the five selected samples, EOs obtained from A. aromaticum, C. cassia, and E. ciliata were analyzed here for the first time for their leishmanicidal effect. In our experiment, they showed IC50 values of 9.25 ± 0.64, 2.29 ± 0.08, and 8.49 ± 0.32 nL/mL, respectively against Lmm promastigotes. Unfortunately, this activity was not very selective on Leishmania as SI values compared to non-cancer mammalian cells (WI38) and cancer cells (J774) were approximately 5 and 2, respectively.
Two EOs extracted from O. gratissimum and Z. zerumbet were already investigated for anti-leishmanial activity [13,14,15,16] however on other L. species. The EO extracted from fresh leaves of O. gratissimum collected in Brazil did not reveal interesting effects against both L. amazonensis and L. chagasi. On the contrary, in our study, the EO extracted from O. gratissimum collected in Vietnam demonstrated notable activity against Lmm promastigotes, with an IC50 value of 4.85 ± 1.65 nL/mL. These results indicated a specific activity of this EO related to parasite species (Leishmania mexicana mexicana) and/or collection place and composition. Another important feature of this EO is its absence of cytotoxicity as the percentages of viable WI38 and J774 cells at the maximal tested concentration (50 nL/mL) were higher than 80%. We therefore selected the EO extracted from O. gratissimum as the most active and selective sample. Regarding the EO extracted from fresh rhizomes of Z. zerumbet, its activity against L. donovani was in the same range than what we found against Lmm promastigotes (IC50 = 3.34 ± 0.34 nL/mL). Unfortunately, we also observed cytotoxicity of this EO on WI38 and J774 cells as shown by SI values of 1.10 and 0.72, respectively.
Eucalyptol, the major compound of the EO extracted from A. aromaticum (55.2%), was not active against the different tested L. species [17,18]. Other components present in our A. aromaticum EO, such as citral (neral and geranial, 9.6%), 2-(E)-decenal (5.3%), α-terpineol (2.8%), β-pinene (2.6%) and α-pinene (2.1%) may be therefore responsible, in part, of the observed activity. Among these compounds, 2-(E)-decenal and α-pinene have already shown leishmanicidal effects. IC50 values of 2-(E)-decenal against L. donovani promastigotes and axenic amastigotes were 7.85 ± 0.28 and 2.47 ± 0.25 µg/mL respectively [29], and IC50 value of α-pinene against L. amazonensis promastigotes, L. amazonensis axenic amastigotes and L. infantum promastigotes were 19.7, 16.1 and 45.94 µg/mL respectively [17,30].
Citral was characterized as the main component of the EO extracted from E. ciliata (40.2%). It showed a moderate activity against L. donovani, L. amazonensis and a weak effect against L. infantum, L. tropica and L. major [20,21,22]. However, it is important to point out that this compound is not selective on Leishmania as shown by the toxicity on kidney epithelial and J774 cells [20,21]. Our results indicated activity on parasites and mammalian cells of the EO extracted from E. ciliata in the same range than citral with IC50 value of 8.49 ± 0.32 nL/mL and SI of 5.58 (compared to WI38) and 1.56 (compared to J774). Nevertheless, citral being present at only 40.2% in the E. ciliata EO, other compounds should also have anti-leishmanial activity as β-(E)-ocimene (14.0%), linalool (8.3%), 1-octen-3-ol (7.1%) and β-(E)-farnesene (6.2%). Dutra et al. have already examined leishmanicidal effect of linalool on L. infantum chagasi. However when axenic amastigotes were treated with linalool, IC50 was 550 µg/mL [24].
EO extracted from C. cassia contained 83.6% of trans-cinnamaldehyde. The anti-leishmanial activity of this compound is unknown but its toxicity on two human cell lines was determined [19]. In our sample, the high percentage of cinnamaldehyde may explain the toxicity of this EO on both mammalian cell lines, WI38 and J774, with IC50 of 14.19 ± 0.54 and 6.26 ± 0.80 nL/mL respectively. Given the known cytotoxicity of this compound and its high concentration in the C. cassia EO, we did not find interesting to further analyze its anti-parasitic activity.
As mentioned previously, eugenol was characterized as the major compound (86.5%) of the most interesting EO extracted from our Vietnamese sample of O. gratissimum. Literature data indicated a moderate effect on L. amazonensis and a less interesting activity against L. infantum chagasi of eugenol [23,24,25]. On L. donovani, along with a moderate in vitro activity, in vivo effect of eugenol (emulsified) was determined. The intra-peritoneal administration of an eugenol emulsion at the dose of 75 mg/kg b.w. for 10 consecutive days decreased by 87.01 ± 5.85 and 86.68 ± 5.42% parasitic load in spleen and liver, respectively, in 8-weeks infected BALB/c mice. Moreover, a significant reduction in spleen size, and spleen and liver weights were also found at this dose [26]. Our results further support the anti-leishmanial activity of this compound with IC50 value of 2.57 ± 0.57 nL/mL (=2.72 µg/mL or 15.67 µM) against Lmm promastigotes. Interestingly, eugenol was not toxic on both non-cancer and cancer mammalian cells in our models at the highest analyzed concentration (50 nL/mL). From these results, and its high percentage in the O. gratissimum EO, it can be concluded that activity of eugenol can explain the anti-leishmanial activity of this EO.
Zerumbone, accounting for 60.3% of the EO extracted from fresh rhizomes of Z. zerumbet, was reported to be active against L. donovani, however it revealed toxicity on human leukemia cells HL-60 [27,28]. These data can explain the anti-leishmanial activity but also cytotoxicity of the Z. zerumbet EO found in our study.
Mechanisms of anti-leishmanial activity of these EOs and their major compounds are not well known. Usually, effects were analyzed on the morphology of treated parasites or as a consequence of immunostimulatory activities.
Most of the studies used transmission electron microscopy (TEM) and scanning electron microscopy (SEM) to analyze the morphology of EOs treated parasites. The EO extracted from O. gratissimum caused considerable mitochondrial swelling in L. amazonensis promastigotes at 135 µg/mL and amastigotes at 100 µg/mL [13], swelling of cell body, flagellar pocket and mitochondria in L. chagasi promastigotes at 50 µg/mL [15]. Ultrastructure alterations were also observed in L. amazonenesis and L. infantum promastigotes treated with citral at concentrations of 8.0 and 42 µg/mL respectively [21,22]. Mukherjee et al. detected morphological alterations by SEM in L. donovani promastigotes treated with 9.36 µM of zerumbone associated with induced ROS-mediated apoptosis [27].
Using other experiments, citral at the concentration of 42 µg/mL was shown to trigger programmed cell death of L. infantum promastigotes, as indicated by the externalization of phosphatidylserine, loss of mitochondrial membrane potential and cell-cycle arrest at the G(0)/G(1) phase [22].
Regarding immunostimulatory activity, an increase of nitric oxide produced by infected macrophages has been suggested to be responsible for the activity of the O. gratissimum EO against L. amazonensis at 100 and 150 µg/mL [13]. Islamuddin et al. explored the synergic effect between eugenol emulsion and the immune system. In BALB/mice infected by L. donovani, treatment with 75 mg/kg b.w. of eugenol emulsion enhanced IFN-γ and IL-2 serum levels, as well as increased CD4+ and CD8+ T cell population and expanded IFN-γ producing CD4+ and CD8+ splenic T lymphocytes [26].
Concerning the mechanism of eugenol activity, although no data are available on Lmm promastigotes, some experiments were carried out on bacteria and fungi. Eugenol at concentrations of 5.3 and 10.6 mg/mL was reported to significantly damage both the cell wall and membrane of the treated Gram-negative and Gram-positive bacteria [31]. Khan et al. observed damaging effects of eugenol at 200 µg/mL on cell wall, cell membrane, cytoplasmic contents and other membranous structures of treated Candida albicans [32]. Indeed, because of the lipophilicity of eugenol, it could easily diffuse between the fatty acyl chains of lipid bilayers modifying the fluidity, integrity and permeability of cell membranes [31]. We now intend to analyze possible effects of this compound on leishmanial membranes.

4. Materials and Methods

4.1. Plants Collection

Thirty-seven plants were collected from different areas of Vietnam in November 2014 and from May to August 2015. They were identified by matching with literature and herbarium specimen at the Botanical Department, Hanoi University of Pharmacy, Vietnam. The information of sample name, genus, species, family, and collector name is given in the supplementary material.

4.2. Essential Oils Extraction

Fresh samples were extracted by hydro-distillation using a modified Clavenger apparatus for two hours. Each essential oil obtained from hydro-distillation was dried using sodium sulfate, filtered and kept refrigerated prior to analysis. Stock solutions at the concentration of 20 µL/mL were prepared in DMSO and diluted further in culture medium to achieve a maximum final DMSO concentration of 0.25%.

4.3. Culture Maintenance

Promastigote form of Leishmania mexicana mexicana (Lmm, MHOM/BZ/84/BEL46) was grown in SDM 79 medium (Life technologies, Ghent, Belgium) supplemented with 15% heat-inactivated fetal bovine serum and 0.2% hemin. The culture was maintained at 28 °C in 5% CO2 incubator. The human non-cancer fibroblast cell line WI38 (ATCC Number CCL-75 from LGC Standards, Middlesex, UK) and macrophage-like murine cell line J774 (ECACC Number 91051511 from Public Health England, Salisbury, UK) were grown in DMEM and RPMI medium (Gibco from Thermo Fisher Scientific, Merelbeke, Belgium or Sigma-Aldrich, Bornem, Belgium), respectively, supplemented with 10% fetal bovine serum and penicillin-streptomycin (100 UI/mL). The culture was maintained at 37 °C in 5% CO2 incubator.

4.4. Anti-leishmanial Assay

The Lmm promastigote density was counted on a haemocytometer and adjusted to 105 parasites/mL. The assays were performed in 96-well plates. For primary screening (repeated two times at concentrations of 50 and 25 nL/mL in triplicate) essential oils were diluted in culture medium from the stock solutions (20 µL/mL). Each well was filled with 50 μL of the diluted essential oil and 50 μL of the Lmm promastigote culture (total volume 100 μL). After 72 h of incubation, 10 µL Alamar blue (Thermo Fisher Scientific, diluted with PBS at the ratio 1:1) was added to each well and the plates were further incubated for 4 h. Fluorescence was measured on a spectrophotometer (SpectraMax-Molecular Devices, Berkshire, UK) at 530 nm excitation and 590 nm emission wavelengths. Pentamidine was tested as standard drug. The essential oils that inhibited more than 50% the growth of Lmm promastigotes at the concentration of 25 nL/mL in the primary screening were submitted to a second analysis for accurate IC50 determination. Selected essential oils were screened at least three times at concentrations ranging from 50 to 0.02 nL/mL in duplicate. IC50 values were calculated from dose response growth inhibition curves by Microsoft excel files.

4.5. Cytotoxicity Assay

The essential oils which were analyzed for IC50 value on anti-leishmanial assay were also analyzed for cytotoxicity against non-cancer (WI38) and cancer (J774) mammalian cells. The culture of cells was diluted with medium to the adequate density of 5 × 103 cells/mL and then 180 μL was added into each well of 96-well plates. After 24 h of incubation, 20 μL of essential oils diluted in culture medium was added to each well (to obtain concentrations ranging from 50−0.02 nL/mL) for further 72 h of incubation. After removing the medium, 100 μL of MTT was added to each well and the plates were further incubated for 45 minutes. 100 μL of DMSO was used to dissolve formed formazan crystals after MTT removing. Absorbance was measured on a spectrophotometer (SpectraMax-Molecular Devices, Berkshire, UK) at 570 nm with a reference wavelength at 620 nm. All experiments were made at least two times in triplicate. The computation of the IC50 values was performed with GraphPad Prism 5.0 (GraphPad Software Inc., San Diego, CA, USA). The selectivity index (SI) values were calculated using the formula below:
SI = IC50 for mammalian cell/IC50 for protozoan parasite

4.6. Essential Oil Analysis

The GC-MS analyses were carried out on a TRACE GC 2000 series (Thermo-Quest, Rodano, Italy), with a DB-WAX capillary column (30 m × 0.25 mm × 0.25 µm) using the following operating conditions: injection volume: 1 µL (TBME solution); injection mode: splitless; injector temperature: 230 °C; oven temperature: increased from 45 °C (held on 5 min) to 250 °C (held on 5 min) at 3 °C/min; helium was used as a carrier gas at a constant flow of 1.3 mL/min; detector temperatures: 260 °C; ion source: 70 eV. The oil components were identified using linear retention indices in relation to a series of fatty acid methyl esters (C5–C27), pure compounds and NIST mass spectral library (matching coefficient > 700). GC-FID was done using the same column and conditions on a FOCUS GC (Thermo Finnigan, Milan, Italy) with modifications of injection mode (split at ratio 1:50) and detector temperature (250 °C). Percentage of compounds was calculated by the normalization procedure.

5. Conclusions

The present study analyzes for the first time the anti-leishmanial activity of 37 essential oils extracted from Vietnamese plants. Those extracted from A. aromaticum, C. cassia and E. ciliata are shown here for the first time to be effective on a Leishmania species, while the effects of O. gratissimum and Z. zerumbet EOs are reported for the first time on L. mexicana species. More than 90% of their contents was characterized. Eugenol was identified as the major compound of the O. gratissimum EO, the most active and selective one. This compound, showing a moderate anti-leishmanial activity and low cytotoxicity in the tested models, can explain this EO activity. However, further results are necessary before developing it in the treatment of cutaneous leishmaniasis such as in vivo assessment in Lmm infected animals and determination of its mode of action.

Supplementary Materials

Supplementary materials are available online.

Acknowledgments

The authors gratefully acknowledge Vietnamese Ministry of Education and Training and Belgian National Fund for Scientific Research (FNRS) (PDR T.0190.13) for financial support. We wish to thank Marie-France Hérent and Marie-Christine Fayt for skillful technical assistance in GC and all the members of the Pharmacognosy group from LDRI, UCL, Belgium, for helpful scientific advices and support.

Author Contributions

Thanh Binh Le directly collected plants, performed chemical, anti-leishmanial activity and cytotoxicity experiments, analyzed the data; Claire Beaufay participated in analyzes of the data and discussions during the work; Duc Trong Nghiem was responsible for the plants identification; Marie-Paule Mingeot-Leclercq participated in discussions during the work; Joëlle Quetin-Leclercq was responsible for the conception of this work. They also wrote this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Centers for Disease Control and Prevention (CDC). Parasites-Leishmaniasis. Available online: https://www.cdc.gov/parasites/leishmaniasis/ (accessed on 8 June 2017).
  2. WHO Expert Committee. Control of the Leishmaniases; WHO Technical Report Series, No. 949; World Health Organization: Geneva, Switzerland, 2010. [Google Scholar]
  3. Drugs for Neglected Diseases initiative (DNDi). Diseases & Projects-Leishmaniasis. Available online: http://www.dndi.org/diseases-projects/leishmaniasis/ (accessed on 8 June 2017).
  4. Schmidt, T.J.; Khalid, S.A.; Romanha, A.J.; Alves, T.M.; Biavatti, M.W.; Brun, R.; da Costa, F.B.; de Castro, S.L.; Ferreira, V.F.; de Lacerda, M.V.; et al. The potential of secondary metabolites from plants as drugs or leads against protozoan neglected diseases-part I. Curr. Med. Chem. 2012, 19, 2128–2175. [Google Scholar] [CrossRef] [PubMed]
  5. Bakkali, F.; Averbeck, S.; Averbeck, D.; Idaomar, M. Biological effects of essential oils—A review. Food. Chem. Toxicol. 2008, 46, 446–475. [Google Scholar] [CrossRef] [PubMed]
  6. Monzote, L.; García, M.; Pastor, J.; Gil, L.; Scull, R.; Maes, L.; Cos, P.; Gille, L. Essential oil from Chenopodium ambrosioides and main components: Activity against Leishmania, their mitochondria and other microorganisms. Exp. Parasitol. 2014, 136, 20–26. [Google Scholar] [CrossRef] [PubMed]
  7. Monzote, L.; Pastor, J.; Scull, R.; Gille, L. Antileishmanial activity of essential oil from Chenopodium ambrosioides and its main components against experimental cutaneous leishmaniasis in BALB/c mice. Phytomedicine 2014, 21, 1048–1052. [Google Scholar] [CrossRef] [PubMed]
  8. Monzote, L.; García, M.; Scull, R.; Cuellar, A.; Setzer, W.N. Antileishmanial activity of the essential oil from Bixa orellana. Phytother. Res. 2014, 28, 753–758. [Google Scholar] [CrossRef] [PubMed]
  9. Montoro, P.; Masullo, M.; Piacente, S.; Pizza, C. Extraction, sample preparation, and analytical methods for quality issues of essential oils. In Aromatherapy: Basic Mechanisms and Evidence-Based Clinical Use; Bagetta, G., Cosentino, M., Sakurada, T., Eds.; CRC Press: Boca Raton, FL, USA, 2015; p. 106. ISBN 978-1-4822-4663-6. [Google Scholar]
  10. Hérent, M.F.; de Bie, V.; Tilquin, B. Determination of new retention indices for quick identification of essential oils compounds. J. Pharm. Biomed. Anal. 2007, 43, 886–892. [Google Scholar] [CrossRef] [PubMed]
  11. Bero, J.; Kpoviessi, S.; Quetin-Leclercq, J. Anti-parasitic activity of essential oils and their constituents against Plasmodium, Trypanosoma and Leishmania. In Novel Plant Bioresource: Applications in Food, Medicine and Cosmetic; Gurib-Fakim, A., Ed.; John Wiley & Sons: Oxford, UK, 2014; pp. 445–469. ISBN 978-1-118-46061-0. [Google Scholar]
  12. Choi, M.S.; Choi, B.S.; Kim, S.H.; Pak, S.C.; Jang, C.H.; Chin, Y.W.; Kim, Y.M.; Kim, D.I.; Jeon, S.; Koo, B.S. Essential Oils from the Medicinal Herbs Upregulate Dopamine Transporter in Rat Pheochromocytoma Cells. J. Med. Food. 2015, 18, 1112–1120. [Google Scholar] [CrossRef] [PubMed]
  13. Ueda-Nakamura, T.; Mendonça-Filho, R.R.; Morgado-Díaz, J.A.; Korehisa Maza, P.; Prado Dias Filho, B.; Aparício Garcia Cortez, D.; Alviano, D.S.; Rosa Mdo, S.; Lopes, A.H.; Alviano, C.S.; et al. Antileishmanial activity of Eugenol-rich essential oil from Ocimum gratissimum. Parasitol. Int. 2006, 55, 99–105. [Google Scholar] [CrossRef] [PubMed]
  14. Kpadonou Kpoviessi, B.G.H.; Kpoviessi, D.S.S.; Yayi Ladekan, E.; Gbaguidi, F.; Frédérich, M.; Moudachirou, M.; Quetin-Leclercq, J.; Accrombessi, G.C.; Bero, J. In vitro antitrypanosomal and antiplasmodial activities of crude extracts and essential oils of Ocimum gratissimum Linn from Benin and influence of vegetative stage. J. Ethnopharmacol. 2014, 155, 1417–1423. [Google Scholar] [CrossRef] [PubMed]
  15. Oliveira, V.C.S.; Moura, D.M.S.; Lopes, J.A.D.; de Andrade, P.P.; da Silva, N.H.; Figueiredo, R.C.B.Q. Effects of essential oils from Cymbopogon citratus (DC) Stapf., Lippia sidoides Cham., and Ocimum gratissimum L. on growth and ultrastructure of Leishmania chagasi promastigotes. Parasitol. Res. 2009, 104, 1053–1059. [Google Scholar] [CrossRef] [PubMed]
  16. Singh, C.B.; Chanu, S.B.; Kh, L.; Swapana, N.; Cantrell, C.; Ross, S.A. Chemical composition and biological activity of the essential oil of rhizome of Zingiber zerumbet (L.) Smith. J. Pharmacogn. Phytochem. 2014, 3, 130–133. Available online: http://handle.nal.usda.gov/10113/62328 (accessed on 8 June 2017).
  17. 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]
  18. 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] [PubMed]
  19. Behar, R.Z.; Davis, B.; Wang, Y.; Bahl, V.; Lin, S.; Talbot, P. Identification of toxicants in cinnamon-flavored electronic cigarette refill fluids. Toxicol. In Vitro 2014, 28, 198–208. [Google Scholar] [CrossRef] [PubMed]
  20. Zheljazkov, V.D.; Cantrell, C.L.; Tekwani, B.; Khan, S.I. Content, composition, and bioactivity of the essential oils of three basil genotypes as a function of harvesting. J. Agric. Food Chem. 2008, 56, 380–385. [Google Scholar] [CrossRef] [PubMed]
  21. Santin, M.R.; dos Santos, A.O.; Nakamura, C.V.; Dias Filho, B.P.; Ferreira, I.C.P.; Ueda-Nakamura, T. In vitro activity of the essential oil of Cymbopogon citratus and its major component (citral) on Leishmania amazonensis. Parasitol. Res. 2009, 105, 1489–1496. [Google Scholar] [CrossRef] [PubMed]
  22. Machado, M.; Pires, P.; Dinis, A.M.; Santos-Rosa, M.; Alves, V.; Salgueiro, L.; Cavaleiro, C.; Sousa, M.C. Monoterpenic aldehydes as potential anti-Leishmania agents: Activity of Cymbopogon citratus and citral on L. infantum, L. tropica and L. major. Exp. Parasitol. 2012, 130, 223–231. [Google Scholar] [CrossRef] [PubMed]
  23. Xavier, F.J.S.; Rodrigues, K.A.D.F.; de Oliveira, R.G.; Lima Junior, C.G.; Rocha, J.D.; Keesen, T.S.; de Oliveira, M.R.; Silva, F.P.; Vasconcellos, M.L. Synthesis and in vitro anti Leishmania amazonensis biological screening of Morita-Baylis-Hillman Adducts prepared from eugenol, thymol and carvacrol. Molecules 2016, 21, 1483. [Google Scholar] [CrossRef] [PubMed]
  24. Dutra, F.L.; Oliveira, M.M.; Santos, R.S.; Silva, W.S.; Alviano, D.S.; Vieira, D.P.; Lopes, A.H. Effects of linalool and eugenol on the survival of Leishmania (L.) infantum chagasi within macrophages. Acta Trop. 2016, 164, 69–76. [Google Scholar] [CrossRef] [PubMed]
  25. 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.; 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] [PubMed]
  26. Islamuddin, M.; Chouhan, G.; Want, M.Y.; Ozbak, H.A.; Hemeg, H.A.; Afrin, F. Immunotherapeutic potential of eugenol emulsion in experimental visceral leishmaniasis. PLoS Negl. Trop. Dis. 2016, 10, e0005011. [Google Scholar] [CrossRef] [PubMed]
  27. Mukherjee, D.; Singh, C.B.; Dey, S.; Mandal, S.; Ghosh, J.; Mallick, S.; Hussain, A.; Swapana, N.; Ross, S.A.; Pal, C. Induction of apoptosis by zerumbone isolated from Zingiber zerumbet (L.) Smith in protozoan parasite Leishmania donovani due to oxidative stress. Braz. J. Infect. Dis. 2016, 20, 48–55. [Google Scholar] [CrossRef] [PubMed]
  28. Huang, N.C.; Chien, T.Y.; Chen, L.G.; Wang, C.C. Antitumor effects of zerumbone from Zingiber zerumbet in P-388D1 cells in vitro and in vivo. Planta Med. 2005, 71, 219–224. [Google Scholar] [CrossRef] [PubMed]
  29. Donega, M.A.; Mello, S.C.; Moraes, R.M.; Jain, S.K.; Tekwani, B.L.; Cantrell, C.L. Pharmacological activities of cilantro’s aliphatic aldehydes against Leishmania donovani. Planta Med. 2014, 80, 1706–1711. [Google Scholar] [CrossRef] [PubMed]
  30. Rodrigues, K.A.D.F.; Amorim, L.V.; Dias, C.N.; Moraes, D.F.C.; Carneiro, S.M.P.; Carvalho, F.A.D.A. Syzygium cumini (L.) Skeels essential oil and its major constituent α-pinene exhibit anti-Leishmania activity through immunomodulation in vitro. J. Ethnopharmacol. 2015, 160, 32–40. [Google Scholar] [CrossRef] [PubMed]
  31. Kamatou, G.P.; Vermaak, I.; Viljoen, A.M. Eugenol-From the remote Maluku Islands to the international market place: A review of a remarkable and versatile molecule. Molecules 2012, 17, 6953–6981. [Google Scholar] [CrossRef] [PubMed]
  32. Khan, M.S.A.; Ahmad, I.; Cameotra, S.S. Phenyl aldehyde and propanoids exert multiple sites of action towards cell membrane and cell wall targeting ergosterol in Candida albicans. AMB Express 2013, 3, 54. [Google Scholar] [CrossRef] [PubMed]
Sample Availability: Samples of essential oils are available from the authors.
Table 1. Viability of Lmm promastigotes in the presence of 25 nL/mL EO.
Table 1. Viability of Lmm promastigotes in the presence of 25 nL/mL EO.
Plant SpeciesEO Obtained FromViability of Lmm promastigotes (%) (Average ± Standard Deviation)
Ageratum conyzoides (L.) L.leaves89.84 ± 1.58
Alpinia galanga (L.) Willd.rhizomes93.02 ± 4.90
Amomum aromaticum Roxb.fruits0.27 ± 0.05
Amomum schmidtii (K. Schum.) Gagnep.rhizomes96.82 ± 1.13
Anethum graveolens L.fruits96.53 ± 1.26
Artemisia annua L.leaves92.32 ± 1.55
Blumea lanceolaria (Roxb.) Druceleaves93.59 ± 1.24
Cinnamomum cassia (L.) J. Preslstem barks0.48 ± 0.01
Clausena indica (Dalzell) Oliv.leaves95.07 ± 7.40
Coriandrum sativum L.fruits94.47 ± 2.38
Curcuma longa L.rhizomes92.90 ± 1.61
Curcuma zedoaria (Christm.) Roscoerhizomes99.53 ± 0.23
Dysphania ambrosioides (L.) Mosyakin & Clemantleaves, fruits92.69 ± 1.93
Elsholtzia blanda (Benth.) Benth.leaves94.70 ± 6.12
Elsholtzia ciliata (Thunb.) Hyl.leaves0.38 ± 0.00
Elsholtzia communis (Collett & Hemsl.) Dielsleaves97.24 ± 2.19
Elsholtzia penduliflora W. W. Sm.leaves97.88 ± 0.37
Eucalyptus camaldulensis Dehnh.leaves101.13 ± 0.54
Hedychium coronarium J.Koenigrhizomes91.79 ± 4.60
Hyptis suaveolens (L.) Poit.leaves67.32 ± 1.97
Illicium verum Hook. f.fruits93.35 ± 5.29
Kaempferia galanga L.rhizomes100.00 ± 7.47
Litsea cubeba (Lour.) Pers.fruits96.00 ± 5.18
Litsea cubeba (Lour.) Pers.leaves94.67 ± 2.99
Melaleuca alternifolia (Maiden & Betch) Cheelleaves87.61 ± 4.93
Melaleuca cajuputi Powellleaves92.53 ± 3.17
Ocimum gratissimum L.leaves0.13 ± 0.01
Ocimum tenuiflorum L.leaves89.08 ± 5.23
Piper sarmentosum Roxb.leaves81.43 ± 14.12
Platycladus orientalis (L.) Francoleaves92.41 ± 16.49
Plectranthus amboinicus (Lour.) Spreng.leaves82.06 ± 1.50
Pluchea indica (L.) Less.leaves88.07 ± 4.37
Pogostemon cablin (Blanco) Benth.leaves93.64 ± 1.51
Vitex trifolia L.leaves88.70 ± 3.34
Zingiber montanum (J.Koenig) Link ex A.Dietr.rhizomes88.36 ± 1.59
Zingiber officinale Roscoerhizomes94.29 ± 3.83
Zingiber zerumbet (L.) Roscoe ex Sm.rhizomes0.19 ± 0.05
Table 2. In vitro anti-leishmanial activity, cytotoxicity and selectivity indices of the five selected EOs.
Table 2. In vitro anti-leishmanial activity, cytotoxicity and selectivity indices of the five selected EOs.
Plant SpeciesAnti-Leishmanial Activity (IC50 nL/mL) Average ± Standard DeviationCytotoxicity (IC50 nL/mL) Average ± Standard Deviation
WI38SI (WI38)J774SI (J774)
Amomum aromaticum9.25 ± 0.6447.31 ± 0.305.1122.68 ± 3.222.45
Cinnamomum cassia2.92 ± 0.0814.19 ± 0.544.856.26 ± 0.802.14
Elsholtzia ciliata8.49 ± 0.3247.38 ± 1.645.5813.21 ± 1.481.56
Ocimum gratissimum4.85 ± 1.65>50>10.3>50 >10.3
Zingiber zerumbet3.34 ± 0.343.68 ± 0.341.102.41 ± 0.040.72
Pentamidine0.04 ± 0.006 *
Camptothecin 0.13 ± 0.02 * 0.01 ± 0.01 *
* concentration in µg/mL.
Table 3. Chemical composition of the five selected EOs.
Table 3. Chemical composition of the five selected EOs.
No.CompoundsRIRelative Percentages (%)Identification
A. aromaticumC. cassiaE. ciliataO. gratissimumZ. zerumbet
1α-Pinene5352.1---2.3MS, [10], Co-GC
2Camphene573−576t---8.0MS, [10]
3β-Pinene619−6212.6-0.1-0.2MS, [10], Co-GC
4Sabinene634−635t-t0.2-MS, [10]
53-Carene664−6650.1---1.1MS
6α-Phellandrene682----0.1MS, Co-GC
7Myrcene684--0.20.20.4MS, [10]
8α-Terpinene697---t-MS, [10], Co-GC
9Limonene719−7200.8-4.1-1.0MS, [10], Co-GC
10Eucalyptol730-74055.2---1.6MS, [10], Co-GC
11(Z)-β-Ocimene758−760--0.75.4-MS
12γ-Terpinene765−766--t0.1tMS, [10], Co-GC
13(E)-β-Ocimene773−778--14.00.2-MS
14p-Cymene787−7920.6ttt0.2MS, [10], Co-GC
15Terpinolene800−801--tt0.1MS, [10], Co-GC
16Octanal8120.2----MS
17(E)-3-Hexen-1-ol acetate837--t--MS
185-Hepten-2-one, 6-methyl-855−8580.2-1.1--MS, Co-GC
19α-Pinene oxide875--0.1--MS
20allo-Ocimene887---0.1-MS, Co-GC
211-Octen-1-yl acetate897--1.0--MS
22(Z)-3-Hexen-1-ol902−904--0.5t-MS, Co-GC
23Fenchone907----0.1MS, Co-GC
243-Octanol915--0.3--MS, [10]
25(E)-2-Octenal9450.6----MS
26cis-Linalool oxide (furanoid)961t----MS, Co-GC
271-Octen-3-ol971--7.1--MS, [10]
28cis-Sabinene hydrate980---0.3-MS
29Cyclosativene987-0.1---MS
30trans-Linalool oxide (furanoid)988t----MS, Co-GC
31Citronellal990--t--MS, [10], Co-GC
32α-Copaene998−1003-4.2-0.2-MS
33Camphor1020−1021--0.4-2.1MS, [10], Co-GC
34β-Bourbonene1023---0.2-MS
35Benzaldehyde1027-1.0t--MS
36β-Cubebene1044---0.1-MS
37Linalool1060−10650.7-8.30.10.3MS, [10], Co-GC
38Terpinen-1-ol10770.1----MS
39Bornyl acetate1086----0.1MS, Co-GC
40trans-α-Bergamotene1091−1092-tt--MS, [10]
41β-Elemene1093−1094-0.2-0.1-MS
42β-Caryophyllene1098−1101-0.13.01.30.7MS, [10], Co-GC
43Terpinen-4-ol1108−11130.9tt0.20.2MS, [10], Co-GC
44Acetophenone1149--0.7--MS
45(E)-2- Decenal11535.3----MS
46α-Humulene1166−1173-ttt9.2MS, [10], Co-GC
47(E)-β-Farnesene1178--6.2--MS
48γ-Muurolene1187−1188-0.5-t-MS
49Neral1187−11883.2-16.8--MS, Co-GC
50Methyl geranate1197--0.2--MS
51α-Terpineol1201−12072.8-0.4-0.2MS, [10], Co-GC
52Borneol1205----0.2MS, [10], Co-GC
53Germacrene D1207---3.4-MS, [10]
54Geranyl formate12120.6----MS
55β-Selinene1214-0.1---MS
56Neryl acetate1218--t--MS, [10], Co-GC
57α-Muurolene1224-0.8---MS
58Geranial12386.4-23.4--MS
59β-Cadinene1253−1254---0.20.1MS
60δ-Cadinene1257-2.0---MS
61Geranyl acetate1259−12630.9-0.7--MS, [10], Co-GC
62Citronellol1272--0.5--MS, [10], Co-GC
63Benzenepropanal1273-1.7---MS
64cis-Cadina-1,4-diene1278-0.5---MS
65Isogeraniol1289--0.2--MS
66Nerol1304--3.5--MS, [10], Co-GC
67cis-Isogeraniol1311--0.4--MS
68(E)-2,6-dimethyl-3,5,7-octatriene-2-ol1323−1324--t0.1-MS
69cis-Calamenene1325-0.5---MS
70Geraniol1350−13542.4-3.1--MS, [10], Co-GC
71(E)-2-Dodecenal13591.5----MS
72cis-Cinnamaldehyde1382-2.4---MS
73Indane-4-carboxalde-hyde14661.7----MS
74Caryophyllene oxide1466−1501--0.60.23.6MS, [10], Co-GC
75Humulene epoxide II1522----2.2MS
76trans-Cinnamaldehyde1534-83.6---MS, Co-GC
77(E)-Nerolidol1533−15381.2-0.5--MS, [10], Co-GC
78Cubenol1552-0.2---MS
79Elemol15710.2----MS
80Cinnamyl acetate1633-0.9---MS
81Eugenol1657---86.5-MS, Co-GC
82α-Muurolol1669-0.1---MS
83β-Eudesmol1706----0.2MS
84α-Cadinol1710---0.1-MS
85Cinnamyl alcohol1761-0.2---MS, Co-GC
86Zerumbone1831----60.3MS
87o-Methoxy-cinnamaldehyde1902-0.3---MS
88trans-Phytol2085--0.16--MS, Co-GC
Total identified 90.399.498.399.294.5
t: trace (peak area less than 0.05%); RI: the retention index was calculated using a homologous series of fatty acid methyl esters C5–C27; MS: mass spectra (matching coefficient >700 compared with NIST database); Co-GC: co-injection with pure compound.
Table 4. Profile of in vitro anti-leishmanial activity and cytotoxicity of the five more active EOs and their major compounds.
Table 4. Profile of in vitro anti-leishmanial activity and cytotoxicity of the five more active EOs and their major compounds.
Plants/CompoundsAnti-Leishmanial ActivityCytotoxicityRefs.
L. Species-FormIC50 (µg/mL)Cell LineIC50 (µg/mL)
A. aromaticumND ND
C. cassiaND ND
E. ciliataND PC12 rat pheochromocytoma cells>50[12]
O. gratissimumL. amazonensis-proma.135CHO125.00 ± 1.68[13,14]
L. amazonensis-ama.100WI38165.51 ± 6.81[13,14]
L. chagasi-proma.80 [15]
Z. zerumbetL. donovani-proma.4.62ND [16]
EucalyptolL. infantum-proma.>100Vero cell63.49[17]
L. infantum-ama.>100 [17]
L. infantum-proma.>400 [18]
L. tropica-proma.>400 [18]
L. major-proma.>400 [18]
Cinnamal-dehydeND human embryonic stem cells4.88[19]
human pulmonary fibroblasts5.28[19]
CitralL. donovani-proma.19kidney epithelial cell22.4[20]
L. amazonensis-proma.8.0 ± 0.06J77450.0 ± 0.10[21]
L. amazonensis-ama.25 ± 0.29 [21]
L. infantum-proma.42 [22]
L. tropica-proma.34 [22]
L. major-proma.36 [22]
EugenolL. amazonensis-proma.12.65red blood cell>65.6[23]
L. infantum chagasi-proma.500BALB/c peritoneal macrophages300[24]
L. infantum chagasi-axe. ama.220 [24]
L. infantum chagasi infected macrophages100 [24]
L. infantum chagasi-proma.56.13 ± 2.09 [25]
L. infantum chagasi-ama.20.81 ± 1.59 [25]
Eugenol (emulsified)L. donovani-proma.8.43 ± 0.96murine macrophages>200[26]
L. donovani-ama.5.05 ± 1.72 [26]
ZerumboneL. donovani-proma.2.04HL-602.27[27,28]
ND: not determined; proma.: promastigote; ama.: intracellular amastigote; axe. ama.: axenic amastigote.

Share and Cite

MDPI and ACS Style

Le, T.B.; Beaufay, C.; Nghiem, D.T.; Mingeot-Leclercq, M.-P.; Quetin-Leclercq, J. In Vitro Anti-Leishmanial Activity of Essential Oils Extracted from Vietnamese Plants. Molecules 2017, 22, 1071. https://doi.org/10.3390/molecules22071071

AMA Style

Le TB, Beaufay C, Nghiem DT, Mingeot-Leclercq M-P, Quetin-Leclercq J. In Vitro Anti-Leishmanial Activity of Essential Oils Extracted from Vietnamese Plants. Molecules. 2017; 22(7):1071. https://doi.org/10.3390/molecules22071071

Chicago/Turabian Style

Le, Thanh Binh, Claire Beaufay, Duc Trong Nghiem, Marie-Paule Mingeot-Leclercq, and Joëlle Quetin-Leclercq. 2017. "In Vitro Anti-Leishmanial Activity of Essential Oils Extracted from Vietnamese Plants" Molecules 22, no. 7: 1071. https://doi.org/10.3390/molecules22071071

Article Metrics

Back to TopTop