Skip to main content
Erschienen in: BMC Complementary Medicine and Therapies 1/2016

Open Access 01.12.2016 | Research article

Essential oils: in vitro activity against Leishmania amazonensis, cytotoxicity and chemical composition

verfasst von: Milene Aparecida Andrade, Clênia dos Santos Azevedo, Flávia Nader Motta, Maria Lucília dos Santos, Camila Lasse Silva, Jaime Martins de Santana, Izabela M. D. Bastos

Erschienen in: BMC Complementary Medicine and Therapies | Ausgabe 1/2016

Abstract

Background

The current chemotherapy for cutaneous leishmaniosis (CL) has a series of drug limitations such as toxic side effects, long duration, high costs and drug resistance, which requires the development of new drugs or effective alternatives to the CL treatment. Essential oils (EOs) are complex mixtures of secondary metabolites from various plants. It has been shown that several EOs, or their constituents, have inhibitory activity against protozoa. Thus, this study aims to evaluate the biological activity of different essential oils (EOs) on Leishmania (L.) amazonensis promastigotes forms, as well as their cytotoxicity on mammalian cells and chemical composition.

Methods

Sixteen EOs were evaluated by mean of IC50/24 h and cytotoxicity against L6 cells (CC50/24 h) using Resazurin assay. Only those EOs that presented better results for IC50/24 h were submitted to GC–MS analysis to determine their chemical constitution.

Results

The EO from Cinnamodendron dinisii, Matricaria chamomilla, Myroxylon peruiferum, Salvia sclarea, Bulnesia sarmientoi, Ferula galbaniflua, Siparuna guianensis and Melissa officinalis were the most active against L. amazonensis with IC50/24 h ranging from 54.05 to 162.25 μg/mL. Analysis of EOs by GC–MS showed mainly the presence of β-farnesene (52.73 %) and bisabolol oxide (12.09 %) for M. chamomilla; α-copaene (13.41 %), safrole (8.35 %) and δ-cadinene (7.08 %) for M. peruiferum; linalool (28.80 %) and linalyl acetate (60.08 %) for S. sclarea; guaiol (48.29 %) and 2-undecanone (19.49 %) for B. sarmientoi; ethyl phthalate (13.09 %) and methyl-8-pimaren-18-oate (41.82 %) for F. galbaniflua; and neral (37.18 %) and citral (5.02 %) for M. officinalis.

Conclusion

The EO from F. galbaniflua showed to be effective against L. amazonensis promastigotes forms and presented low cytotoxic activity against L6 cells. Thus, it represents a strong candidate for future studies aiming its molecular activity on these pathogenic parasites.

Background

Leishmaniasis, the third most important vector-borne diseases, is caused by a protozoan parasite of the genus Leishmania, which is transmitted to human by the bite of sand flies. Leishmaniasis represents a complex disease with diverse clinical manifestations and poses a public health problem since it is a neglected tropical disease with current high worldwide incidence [1, 2]. Globally, more than 12 million individuals are infected, with another 350 million at risk of infection, and nearly 2 million new cases are reported annually worldwide [3]. The disease is prevalent in 16 developed and 72 developing countries; nevertheless 90 % of cases are reported in three regions: Sudan/Ethiopia/Kenya, India/Bangladesh/Nepal and Brazil with as many as 0.02 to 0.04 million deaths every year [3, 4].
Leishmaniasis can be divided into three forms, varying in severity from self-healing cutaneous lesions, dermatological ulcers in cutaneous leishmaniasis (CL), destructive form of mucocutaneous leishmaniasis, to deadly form of visceral leishmaniasis (VL) [5]. CL is characterized by ulcers on the skin that are often formed at the site of the insect vector bite. Those ulcers can undergo metastasis of the nasopharyngeal mucosa developing to tissue destruction, depending on the species of Leishmania involved [6]. Leishmania (Viannia) braziliensis and Leishmania (V.) panamensis are responsible for cases of mucocutaneous leishmaniasis in the Americas, although L. (V.) guyanensis and L. (L.) amazonensis have been identified, especially, in immuno-compromised hosts [6].
The first-line drugs for systemic treatment of leishmaniasis are parenterally administered antimonials such as the sodium stibogluconate (Pentostam®) and the N-methyl glucamine antimoniate (Glucantime®) [7, 8] generally required for the treatment of CL in the New World due to the risk of mucosal involvement [9]. This current chemotherapy presents several issues such as high cost, difficult administration and elevated toxicity, associated with serious side effects [10], for instance musculoskeletal pain, gastrointestinal disturbances, mild to moderate headache, electrocardiographic QTs interval prolongation and mild to moderate increase of liver and pancreatic enzymes [11]. Second-line drug Pentamidine and amphotericin B are not widely used due to their high toxicity and cost. Miltefosine, the first oral anti-leishmanial drug, is the treatment of choice for diffuse cutaneous leishmaniasis and New World cutaneous leishmaniasis caused by Leishmania braziliensis but increasing resistance to this drug has been notified [12].
All antileishmanial drugs except miltefosine have to be administrated parenterally. Most of these drugs are toxic, requires prolonged hospitalization and close monitoring, which makes the treatment costly and beyond the reach of most patients. Consequently, the development of alternative therapies is a priority for the treatment of leishmaniasis. As a strategy, the investigation of extracts and compounds, with biological activity, isolated from plants and used in traditional medicine is a promising in the research field for compounds with potential action for the prophylaxis and chemotherapy of CL [13].
Essential oils (EOs) are complex mixtures of secondary metabolites isolated from plants. In these mixtures, there are 10–60 constituents at different concentrations, but usually only 2–3 major constituents determine the biological properties of the EO [14]. Those compounds and their constituents present a broad pharmacological spectrum, and they are used as analgesics, sedatives, anti-inflammatory, and anti-spasmodic drugs, as well as antimicrobials, antiprotozoals and antihelmintics [13, 15, 16]. It has been shown that several EOs or their constituents have inhibitory activity on protozoa, especially Leishmania [1719]. For instance, Santos and colleagues demonstrated that copaiba oil from Copaifera martii is a safer, shorter, less-expansive, and more easily administered antileishmanial drug [18]. Therefore, the purpose of this present work was to analyze the effect of sixteen EOs biological potential on L. amazonensis promastigotes forms and L6 cells and chemical constitution, by GC-MS, of those EOs that showed better leishmanicidal results.

Methods

Essential oils

EOs of Litsea cubeba fruits; Lavandula officinalis, Matricaria chamomilla and Cananga odorata flowers; Elettaria cardamomum seeds; Cinnamomum camphora, Myroxylon peruiferum and Bulnesia sarmientoi barks; Ferula galbaniflua resin; Salvia sclarea, Foeniculum officinalis, Cordia verbenaceae, and Melissa officinalis leaves; Pelargonium graveolens leaves and stems were purchased from QUINARI Cosmetic and Fragrances Inc. (Maringá-PR, Brazil) with lot number 0717/05209/F. EOs of Cinnamodendron dinisii and Siparuna guianensis leaves were obtained as described by Andrade et al. [20].
Firstly, EOs and Amphotericin B 250 μg/mL (Sigma-Aldrich, St. Louis, USA) were diluted in dimethylsulfoxide (DMSO) at 100 mg/mL and 50 μg/mL, respectively. For use, the stock was diluted 5:100 in either Schneider (Sigma-Aldrich) or RPMI-1640 (Sigma-Aldrich) media (sub stock). DMSO final concentration in the experiments never exceeded 0.5 %, a concentration that is not harmful to parasites and L6 cells [13]. Stocks were stored at 4 °C in the dark, to avoid degradation [21]. The sub stock was freshly prepared before use.

Chemical composition of EOs

Gas chromatography–mass spectrometry (GC–MS) analysis was performed using a Shimadzu GC-2010 gas chromatograph coupled with GCMS-QP2010 Plus equipped with auto sampler (model AOC-20i, Shimadzu, Columbia, MD, USA) and GC–MS Solution software. Investigation was performed with a Rtx-5MS capillary column (30 mm × 0.25 mm × 0.25 μm) at programmed temperature ranging from 60 to 250 °C at 3 °C/min. Analysis conditions were: injector temperature 250 °C, ion source interface temperature 300 °C, analysis of masses between 40–350 m/z, electron impact at 70 eV, column head pressure at constant pressure of 59 kPa, column flow 1.02 mL/min, gas linear velocity: 36.8 cm/s, carrier gas: helium, injected volume 1 μL (1:1000 in hexane) in splitless. Constituents of EOs were identified by comparing their mass spectral pattern and retention indexes (RI) relative to a standard n-alkane series (C9–C24) with those known in the literature and the Wiley W9N08 database [13, 22].

Parasites and culture conditions

The promastigotes forms of L. amazonensis (strain MHOM/BR/77/LTB0016) were maintained at 28 °C in Schneider medium supplemented with 10 % fetal bovine serum (FBS) and 100 μg/mL gentamicin, with weekly passages. For the screening of EOs biological potential, promastigotes were collected from cultures at the mid-log phase of growth (3-day-old culture). The parasite strain was obtained from Fiocruz-COLPROT (Coleção de Protozoários da Fiocruz).

Antileishmanial activity of essential oils in vitro

EOs serial dilutions, from 500 to 31.25 μg/mL, were prepared on a 96-well cell culture plate. Afterwards, 1.35 × 106 parasite/mL culture resuspended in 150 μL were added to the plates and incubated for 24 h at 28 °C. After this period, 20 μL of Resazurin solution were added to a 2 mM final concentration in all wells [23, 24]. The plates were incubated for further 4 h at 37 °C followed by the fluorescence measurement under 570 nmex/595 nmem in the microplate reader SpectraMax M5 (Molecular Devices, Sunnyvale, CA, USA).
In vitro experiment was performed in triplicate and repeated twice independently. DMSO was used as control in the same final concentration found in each dilution. Amphotericin B was used as positive control at final concentration ranging from 312.5 to 19.56 ng/mL.
The percentage of viable promastigotes cells was determined by the equation [25]:
$$ \%\mathrm{P}=\left(100\times \mathrm{F}\mathrm{a}\right)/\mathrm{F}\mathrm{c} $$
Where %P: percentage of viable promastigotes cells; Fc: control fluorescence units; Fa: fluorescence units emitted by the analyzed samples (with inhibitor).

Cytotoxic activity of essential oils in vitro

Uninfected L6 cell monolayers were washed with Phosphate Buffered Saline (PBS) for 5 min at 37 °C, washed with RPMI medium pH 7.4 + 2.5 % FBS, centrifuged at 200 g for 10 min at 4 °C, resuspended in the same medium and, finally, seeded into 96-well plates (5 × 104 cells/well). Plates were incubated at 37 °C for 24 h, then the medium was removed and cells were washed with PBS. Diluted EOs were added to overnight-adhered L6 cells and incubated for more 24 h at 37 °C. Cell viability was assessed by 2 mM Resazurin as described above.
The percentage of viable cells was determined by the equation [25]:
$$ \%\mathrm{V}=\left(100\times \mathrm{F}\mathrm{a}\right)/\mathrm{F}\mathrm{c} $$
Where %V is the percentage of viable cells, Fc: control fluorescence units; Fa: fluorescence units emitted by the analyzed samples (with inhibitor). The selectivity index (SI) was calculated by dividing CC50 for the IC50.

Statistical analysis

For both in vitro EOs biological potential, a randomized complete block design (RBD) test was used, with 5 concentrations, 3 repetitions and 2 experiments (blocks) for each sample. The statistical program used was SISVAR [26]. Data were submitted to analysis of variance and the averages compared by Scott-Knott test and regression, both 5 % probability. The adjusted equations were used to calculate the concentration needed to cripple 50 % of L6 cells (CC50) or 50 % of the parasites (IC50).

Results

Antileishmanial and cytotoxic activity of essential oils in vitro

Growth inhibitory activity by the selected EOs was performed on L. amazonensis promastigotes forms at concentrations ranging from 30 to 500 μg/mL. In the test, the EOs of L. cubeba, E. cardamomum, L. officinalis, C. camphora and C. odorata did not show activity at 500 μg/mL (Table 1). Lower concentrations of the remaining EOs were then evaluated to estimate the IC50/24 h (Table 1). The most effective EO was of the one from S. guianensis (48.55 ± 3.64 μg/mL), followed by C. dinisii (54.05 ± 4.88 μg/mL), M. chamomilla (60.16 ± 4.24 μg/mL), C. verbenaceae (64.75 ± 2.04 μg/mL), B. sarmientoi (85.56 ± 3.38 μg/mL), F. galbaniflua (95.70 ± 1.82 μg/mL), M. officinalis (132.02 ± 3.14 μg/mL), M. peruiferum (162.25 ± 1.57 μg/mL), S. sclarea (325.92 ± 8.58 μg/mL), F. officinalis (328, 28 ± 6,80 μg/mL) and P. graveolens (363.71 ± 6.77 μg/mL). The IC50/24 h of Amphotericin B was 0.83 ± 0.03 μg/mL (Table 1).
Table 1
EOs biological potential and selectivity indexes (SI) for L. amazonensis (IC50/24 h) promastigotes and L6 cells (CC50/24 h)
Essential oils
L. amazonensis
L6 cells
SIg
IC50a ± DP (μg/mL)
CC50 f ± DP (μg/mL)
Litsea cubeba
NIe
180.72 ± 1.37
-
Matricaria chamomilla
60.16 ± 4.24
173.04 ± 1.24
2.87
Elettaria cardamomum
>500.00b
439.57 ± 2.27
Lavandula officinalis
>500.00
377.56 ± 8.91
Cinnamomum camphora
>500.00
>500.00
Myroxylon peruiferum
162.25 ± 1.57
160.80 ± 1.62
0.99
Salvia sclarea
325.92 ± 8.58
375.37 ± 3.62
1.15
Bulnesia sarmientoi
85.56 ± 3.38
163.46 ± 1.77
1.91
Ferula galbaniflua
95.70 ± 1.82
377.26 ± 2.71
3.94
Pelargonium graveolens
363.71 ± 6.77
368.39 ± 3.90
1.01
Cananga odorata
NI
142.80 ± 1.76
Foeniculum officinalis
328. 28 ± 6.80
368.27 ± 3.81
1.12
Cordia verbenaceae
64.75 ± 2.04
130.00 ± 1.08
2.01
Melissa officinalis
132.02 ± 3.14
297.45 ± 1.32
2.25
Siparuna guianensis
48.55 ± 3.64
78.02 ± 1.19
1.60
Cinnamodendron dinisii
54.05 ± 4.88
106.31 ± 2.23
1.97
Anfotericina Bc
(0.083 ± 0.003 μg/mL)
NI
DMSOd
NI
NI
a) IC50 ± DP: the concentration able to cripple 50 % of the parasites ± standard deviation
b) > 500.00: IC50 greater than the highest concentration tested
c) Amphotericin B - positive control
d) DMSO - negative control
e) NI: no inhibition
f) CC50: the concentration able to cripple 50 % of cells after 24 h of treatment ± standard deviation
g) Selectivity index - SI = CC50 L6 / IC50 promastigotes
The cytotoxicity against L6 cells and L. amazonensis were compared using the selectivity index (SI) (Table 1). Higher values of SI means more promising compounds for developing antileishmanial drugs. The SI measures the compound’s level of selectivity towards L. amazonensis. Evaluation of cytotoxicity showed that the least cytotoxic EO was that of C. camphora (CC50/24 h = >˃500.00 μg/mL), followed by E. cardamomum (439.57 ± 2.27 μg/mL), L. officinalis (377.56 ± 8.91 μg/mL), F. galbaniflua (377.26 ± 2.71 μg/mL), S. sclarea (375.37 ± 3.62 μg/mL), P. graveolens (368.39 ± 3.90 μg/mL), F. officinalis (368.27 ± 3.81 μg/mL), M. officinalis (297.45 ± 1.32 μg/mL), L. cubeba (180.72 ± 1.37 μg/mL), M. chamomilla (173.04 ± 1.24 μg/mL), B. sarmientoi (163.46 ± 1.77 μg/mL), M. peruiferum (160.80 ± 1.62 μg/mL), C. odorata (142.80 ± 1.76 μg/mL), C. verbenaceae (130.00 ± 1.08 1.77 μg/mL), C. dinisii (106.31 ± 2.23 μg/mL) and the most cytotoxic EO was of the one from S. guianensis (78.02 ± 1.19 μg/mL) (Table 1).
EOs with higher selectivity indexes were those from F. galbaniflua (3.94), M. chamomilla (2.87) and M. officinalis (2.25), but all were more cytotoxic and less selective than Amphotericin B, because the reference drug did not show CC50/24 h value.

Chemical composition

The GC-MS analyses were performed for EOs that showed the lower values of IC50 and/or the higher SI values (Table 2). The analysis identified the main constituents as β-farnesene (52.73 %), bisabolol oxide (12.09 %), α-farnesene (10.34 %) for M. chamomilla; α-copaene (13.41 %), guaiol (9.35 %), safrole (8.35 %) and δ-cadinene (7.08 %) for M. peruiferum; linalool (28.80 %) and linalyl acetate (60.08 %) for S. sclarea; guaiol (48.29 %) and 2-undecanone (19.49 %) for B. sarmientoi; ethyl phthalate (13.09 %) and methyl-8-pimaren-18-oate (41.82 %) for F. galbaniflua; and neral (37.18 %) and geranial (5.02 %) for M. officinalis. According to Andrade et al. [20], EO from C. dinisii fresh leaves is composed mainly by α-pinene (35.41 %), β-pinene (17.81 %), sabinene (12.01 %) and bicyclogermacrene (7.59 %). EO from S. guianensis fresh leaves contains β-myrcene (13.14 %), germacrene-D (8.68 %) and bicyclogermacrene (16.71 %).
Table 2
Chemical composition of selected essential oils
Content (%)
IRc
IRl
Component
Matricaria chamomilla
Myroxylon peruiferum
Salvia sclarea
Bulnesia sarmientoi
Ferula galbaniflua
Melissa officinalis
Siparuna guianensis a
Cinnamodendron dinisiia a
930
932
α-pinene
1.83
35.41
958
963
sabinene
12.01
961
970
β-pinene
17.34
17.81
969
971
6-metil-5-hepten-2-one
3.88
975
0980
β-myrcene
1.23
13.14
1.46
1018
1024
1,8-cineole
8.71
3.71
4.37
1086
1092
linalool
28.80
2.67
1135
1130
α-terpineol
5.14
1231
1235
neral
37.18
1247
1252
linalyl acetate
60.08
2.64
1263
1264
geranial
52.02
1282
1285
safrole
8.35
1287
1293
2-undecanone
19.49
1.69
1374
1374
α-copaene
13.41
1428
1432
trans-α-bergamotene
3.48
1443
1439
(+)-aromadendrene
2.27
1.26
1451
1455
(E)-β-farnesene
52.73
1455
1458
allo-aromandrendene
5.24
1470
1474
y-gurjunene
5.29
1476
1482
germancrene-D
3.42
8.68
1478
1479
ar-curcumene
5.05
1481
1487
β-selinene
3.27
1488
1497
bicyclogermancrene
16.71
7.59
1504
1505
(E,E)-a-farnesene
10.34
1504
1507
β-bisabolene
2.09
1510
1511
δ-amorfene
6.59
1520
1522
δ-cadinene
7.08
1.04
0.14
1576
1577
(−)-spathulenol
5.79
4.16
1.88
1594
diethyl phthalate
13.09
1601
1600
Guaiol
9.35
48.29
1630
(−)-sinularene
5.81
1652
1649
β-eudesmol
2.10
1656
1656
bisabolol oxide B
12.09
1657
1658
t-cadinol
1,35
4.14
1685
1685
α-bisabolol
9.83
3.35
1732
camazulene
2.30
elixene
5.87
methyl 8 (14)-pimaren-18- oate
41.82
NI
9.39
  
Total identified (%)
90.72
79.38
94.05
88.85
81.83
93.08
55.25
80.67
IRl literature retention rate [22], IRc retention ratio calculated by Kovats’ equation. a) Described by Andrade et al. [20]

Discussion

According to the classification of cytotoxicity and antileishmanial activity for extracts and fractions derived from plants and natural products defined by Study Program and Disease Control [27], the evaluated EOs are classified as moderately toxic (100 < CC50 ≤ 1000 μg/mL), except the EO from S. guainensis, which was classified as toxic (10 < CC50 ≤ 100 μg/mL). Regarding the antileishmanial activity only EOs from S. guianensis, C. dinisii, M. chamomilla, C. verbenaceae, B. sarmientoi, F. galbaniflua and M. officinalis are considered moderately active (50 < IC50 ≤ 150 μg/mL). The others are considered not active.
Considering the chemical composition of the EO from M. chamomilla, (E)-β-farnesene and (E,E)-α-farnesene were found as the major compound group representing 73.07 % of the total composition. These results corroborate with those reported by Machado et al. [28] that found farnesene derivatives as the most representative constituents (22 %) and their bioassays using EO from Lantana camara revealed a significant leishmanicidal activity against L. amazonensis (IC50/72 h = 0.25 μg/mL), except for the cytotoxic activity, in which the authors obtained high values on Brine shrimp (CC50 10 μg/mL). Subsequently, Gawde et al. [29] observed that the chemical composition of M. chamomilla was similar to the one found in our study (β-farnesene, α-bisabolol oxide B, chamazulene) but no leishmanicidal activity on L. donavani was observed.
Studies on the chemical composition and biological activity of M. peruiferum EO are scarce. The literature reports (E) and (Z)-nerolidol, α-bisabolol and (E, E)-farnesol as its major components [30] but those compounds were not identified in the present study. Santos et al. [18] reported high levels of α-copaene in EO from Copaifera reticulata as well as for EO from M. peruiferum. The last one showed growth inhibitory activity for L. amazonensis with IC50/72 h values of 5 μg/mL for promastigotes and low cytotoxicity on J774G8 macrophages.
Ghannadi and Amree [31] have already described the EO composition obtained from the fresh oleogum resin and latex of Iranian F. galbaniflua (synonym F. gummosa) and the main constituents of this monoterpene rich oil were β-pinene (58.8 %). Other studies also indicate β-pinene as the major compound from the fresh oleogum resin and latex of this same specie [32, 33], which corroborates our results. The presence of methyl 8-(14)-pimaren-18-ate, a diterpene esters hydrocarbons, has been reported on rosin, a solid form of resin obtained from pines and some other plants; and also in the Cretaceous resins from India and Myanmar [34, 35]. To our knowledge, there is no antileishmanial activity reports related to this EO to date.
Rodilla et al. [36] determined the chemical composition of EO from B. sarmientoi. In accordance with our work, they identified guaiol as its major component. Studies with EO from Endlicheria bracteolata, which has 72.12 % of guaiol in its composition, showed IC50 of 7.93 μg/mL for L. amazonensis and presented a CC50 of 15.14 μg/mL for J774.G8 macrophages [37]. The antileishmanial activity may be attributed to the presence of a hydroxyl group of alcohol characteristics in the guaiol, especially in the exocyclic portion of the molecule [36].
The presence of linalyl acetate and linalool as the major compounds in S. sclarea EO (total of 88.88 %) corroborate to the results presented by Pitarokili et al. [38] that evaluated the EO composition of S. sclarea originated from two localities in Greece, and by Kuźma et al. [39] that evaluated the EO composition from S. sclarea plants generated in vitro. On the other hand, antileishmanial activity of linalool-rich EO from leaves of Croton cajucara against L. amazonensis was previously evaluated by Rosa et al. [40], they were able to demonstrate morphological changes in L. amazonensis promastigotes when treated with 15 ng/mL of that EO. In this study the cell lysis was observed within 1 h, indicating that the antileishmanial activity observed is directly related to the presence of linalool, due to the existence of a hydroxyl group in the organic alcohol function.
As in our study, the presence of the isomers of citral, neral and geranial are constantly reported in the chemical composition of the EO from M. officinalis [4143]. Regarding the antileishmanial activity, Mikus et al. [44] reported an IC50/72 h of 7 μg/mL for L. major, a CC50/72 h of 25.5 μg/mL in HL-60 cells and SI of 3.6, higher than those observed in our study. Another study has already showed that citral presents activity against T. cruzi, possibly by inducing cell membrane lysis with leakage of cytoplasm [45].
The EO from C. dinisii and S. guianensis showed weak inhibitory effect on the protozoan T. cruzi with values of IC50/24 h = 209.30 μg/mL and 282.93 mg/mL, respectively. These values are higher when compared to those obtained in the study for L. amazonensis, 54.05 and 48.55 μg/mL, respectively [25].
The mechanism of action by which EOs inhibits parasite growth is still not well known, but previous studies have suggested that structural and morphological changes are caused by drugs that inhibit ergosterol synthesis, or interact with the membrane ergosterol [19, 46]. Other studies indicated that the activity of essential oils on parasites is mainly due to terpene composition. Terpenes are responsible for the hydrophobic characteristic of EOs, thus allowing their diffusion through the parasite cell membrane, affecting intracellular metabolic pathways and organelles [47].

Conclusion

F. galbaniflua EO is effective against L. amazonensis promastigotes forms and has low cytotoxic activity. Thus, it represents a strong candidate for future studies in order to comprehend its biological activity agaisnt L. amazonensis.
The promising results of this study offer prospects for further research, as the evaluation of the antileishmanial potential of the major compounds and the elucidation of their molecules may, in the future, contribute to the discovery of effective drugs derived from plants for the treatment of parasitic diseases.

Acknowledgements

The autors are indebted with Central Analítica do Instituto de Química da Universidade de Brasília (CAIQ/UnB) for GC-MS analysis.

Funding

This work was supported by Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES): Programa Nacional de Incentivo a Pesquisa em Parasitologia Básica (CAPES grant no: 23038.005298/2011-83); Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq Chamada 79/2013-MCTI/CNPq/FNDCT); Fundação de Amparo à Pesquisa do Distrito Federal (FAP-DF- 193.001.076/2015); PGCTS-FCE/UNB; Financiadora de Estudos e Projetos (FINEP); Decanato de Pesquisa e Pós-graduação/UnB (DPP/UnB).

Availability of data and materials

The datasets supporting the conclusions of this article are included within the article.

Authors’ contributions

IMDB and JMS conceived the research idea. MAA, CSA and CLS conducted the experiment, analyzed and interpreted the data as well as prepared the first draft. MLS supported the conduct of the analysis and interpreted the data of CG-MS. IMDB, JMS, FNM, and CSA critically read and revised the paper. All authors have read and approved the paper before its final submission.

Competing interests

The authors declare that they have no competing interests.
Not applicable.
Not applicable.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
2.
Zurück zum Zitat de Vries HJ, Reedijk SH, Schallig HD. Cutaneous leishmaniasis: recente developments in diagnosis and management. Am J Clin Dermatol. 2015;16:99–109.CrossRefPubMedPubMedCentral de Vries HJ, Reedijk SH, Schallig HD. Cutaneous leishmaniasis: recente developments in diagnosis and management. Am J Clin Dermatol. 2015;16:99–109.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Alvar J, Vélez ID, Bern C, Herrero M, Desjeux P, Cano J, et al. Leishmaniasis worldwide and global estimates of its incidence. PloS One. 2012;7:e35671.CrossRefPubMedPubMedCentral Alvar J, Vélez ID, Bern C, Herrero M, Desjeux P, Cano J, et al. Leishmaniasis worldwide and global estimates of its incidence. PloS One. 2012;7:e35671.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Name RQ, Borges KT, Nogueira LSC, Sampaio JHD, Tauil PL, Sampaio RNR. Clinical, epidemiological and therapeuthic study of 402 patients with american cutaneous leishmaniasis attended at University Hospital of Brasilia, DF, Brazil. Ann Bras Dermatol. 2005;80:249–54.CrossRef Name RQ, Borges KT, Nogueira LSC, Sampaio JHD, Tauil PL, Sampaio RNR. Clinical, epidemiological and therapeuthic study of 402 patients with american cutaneous leishmaniasis attended at University Hospital of Brasilia, DF, Brazil. Ann Bras Dermatol. 2005;80:249–54.CrossRef
9.
Zurück zum Zitat Minodier P, Parola P. Cutaneous leishmaniasis treatment. Travel Med Infect Dis. 2007;5:150–8.CrossRefPubMed Minodier P, Parola P. Cutaneous leishmaniasis treatment. Travel Med Infect Dis. 2007;5:150–8.CrossRefPubMed
10.
Zurück zum Zitat Reithinger R, Dujardin JC, Louzir H, Pirmez C, Alexander B, Brooker S. Cutaneous leishmaniasis. Lancet Infect Dis. 2007;7:581–96.CrossRefPubMed Reithinger R, Dujardin JC, Louzir H, Pirmez C, Alexander B, Brooker S. Cutaneous leishmaniasis. Lancet Infect Dis. 2007;7:581–96.CrossRefPubMed
11.
Zurück zum Zitat Ben Salah A, Ben Messaoud N, Guedri E, Zaatour A, Ben Alaya N, Bettaieb J. Topical paromomycin with or without gentamicin for cutaneous leishmaniasis. N Engl J Med. 2013;368:524–32.CrossRefPubMed Ben Salah A, Ben Messaoud N, Guedri E, Zaatour A, Ben Alaya N, Bettaieb J. Topical paromomycin with or without gentamicin for cutaneous leishmaniasis. N Engl J Med. 2013;368:524–32.CrossRefPubMed
12.
Zurück zum Zitat Utaile M, Kassahun A, Abebe T, Hailu A. Susceptibility of clinical isolates of Leishmania aethiopica to miltefosine, paromomycin, amphotericin B and sodium stibogluconate using amastigote-macrophage in vitro model. Exp Parasitol. 2013;134:68–75.CrossRefPubMed Utaile M, Kassahun A, Abebe T, Hailu A. Susceptibility of clinical isolates of Leishmania aethiopica to miltefosine, paromomycin, amphotericin B and sodium stibogluconate using amastigote-macrophage in vitro model. Exp Parasitol. 2013;134:68–75.CrossRefPubMed
13.
Zurück zum Zitat Azeredo CMO, Santos TG, Maia BHLNS, Soares MJ. In vitro biological evaluation of eight different essential oils against Trypanosoma cruzi, with emphasis on Cinnamomum verum essential oil. BMC Complementary Altern Med. 2014;14:309–15.CrossRef Azeredo CMO, Santos TG, Maia BHLNS, Soares MJ. In vitro biological evaluation of eight different essential oils against Trypanosoma cruzi, with emphasis on Cinnamomum verum essential oil. BMC Complementary Altern Med. 2014;14:309–15.CrossRef
14.
Zurück zum Zitat Bakkali F, Averbeck S, Averbeck D, Idaomar M. Biological effects of essential oils - A review. Food Chem Toxicol. 2008;46:446–75.CrossRefPubMed Bakkali F, Averbeck S, Averbeck D, Idaomar M. Biological effects of essential oils - A review. Food Chem Toxicol. 2008;46:446–75.CrossRefPubMed
15.
Zurück zum Zitat Singh G, Maurya S, Lampasona MP, Catalan CAN. A comparison of chemical, antioxidant and antimicrobial studies of cinnamon leaf and bark volatile oils, oleoresins and their constituents. Food Chem Toxicol. 2007;45:1650–61.CrossRefPubMed Singh G, Maurya S, Lampasona MP, Catalan CAN. A comparison of chemical, antioxidant and antimicrobial studies of cinnamon leaf and bark volatile oils, oleoresins and their constituents. Food Chem Toxicol. 2007;45:1650–61.CrossRefPubMed
16.
Zurück zum Zitat Victoria FN, Lenardão EJ, Savegnago L, Perin G, Jacob RG, Alves D, Da Silva WP, Motta AS, Nascente OS. Essential oil of the leaves of Eugenia uniflora L.: antioxidant and antimicrobial properties. Food Chem Toxicol. 2012;50:2668–74.CrossRefPubMed Victoria FN, Lenardão EJ, Savegnago L, Perin G, Jacob RG, Alves D, Da Silva WP, Motta AS, Nascente OS. Essential oil of the leaves of Eugenia uniflora L.: antioxidant and antimicrobial properties. Food Chem Toxicol. 2012;50:2668–74.CrossRefPubMed
17.
Zurück zum Zitat Ueda-Nakamura T, Mendonça-Filho RR, Morgado-Díaz JA, Maza PK, Prado Dias Filho B, Cortez DAD, Alviano DS, Rosa MS, Lopes AH, Alviano CS, Nakamura CV. Antileishmanial activity of eugenol-rich essential oil from Ocimum gratissimum. Parasitol Int. 2006;55:99–105.CrossRefPubMed Ueda-Nakamura T, Mendonça-Filho RR, Morgado-Díaz JA, Maza PK, Prado Dias Filho B, Cortez DAD, Alviano DS, Rosa MS, Lopes AH, Alviano CS, Nakamura CV. Antileishmanial activity of eugenol-rich essential oil from Ocimum gratissimum. Parasitol Int. 2006;55:99–105.CrossRefPubMed
18.
Zurück zum Zitat Santos AO, Ueda-Nakamura T, Dias Filho BP, Veiga Junior VF, Pinto AC, Nakamura CV. Effect of Brazilian copaiba oils on Leishmania amazonensis. J Ethnopharmacol. 2008;120:204–8.CrossRefPubMed Santos AO, Ueda-Nakamura T, Dias Filho BP, Veiga Junior VF, Pinto AC, Nakamura CV. Effect of Brazilian copaiba oils on Leishmania amazonensis. J Ethnopharmacol. 2008;120:204–8.CrossRefPubMed
19.
Zurück zum Zitat Medeiros MGF, Silva AC, Citó AMGL, Borges AR, Lima SG, Lopes JAD, Figueiredo RCBQ. In vitro antileishmanial activity and cytotoxicity of essential oil from Lippia sidoides Cham. Parasitol Int. 2011;60:237–41.CrossRefPubMed Medeiros MGF, Silva AC, Citó AMGL, Borges AR, Lima SG, Lopes JAD, Figueiredo RCBQ. In vitro antileishmanial activity and cytotoxicity of essential oil from Lippia sidoides Cham. Parasitol Int. 2011;60:237–41.CrossRefPubMed
20.
Zurück zum Zitat Andrade MA, Cardoso MG, Andrade J, Silva LF, Teixeira ML, Resende JV, Figueiredo ACS, Barroso J. Chemical Composition and Antioxidant Activity of Essential Oils from Cinnamodendron dinisii Schwacke and Siparuna guianensis Aublet. Antioxidants. 2013;2:384–97.CrossRefPubMedPubMedCentral Andrade MA, Cardoso MG, Andrade J, Silva LF, Teixeira ML, Resende JV, Figueiredo ACS, Barroso J. Chemical Composition and Antioxidant Activity of Essential Oils from Cinnamodendron dinisii Schwacke and Siparuna guianensis Aublet. Antioxidants. 2013;2:384–97.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Guimarães LGL, Cardoso MG, Zacaroni LM, Lima RK, Pimentel F, Morais AR. Influência da luz e da temperatura sobre a oxidação do óleo essencial de capim-limão (Cymbopogon citratus (D.C.) STAPF). Quim Nova. 2008;31:1476–80.CrossRef Guimarães LGL, Cardoso MG, Zacaroni LM, Lima RK, Pimentel F, Morais AR. Influência da luz e da temperatura sobre a oxidação do óleo essencial de capim-limão (Cymbopogon citratus (D.C.) STAPF). Quim Nova. 2008;31:1476–80.CrossRef
22.
Zurück zum Zitat Adams RP. Identification of Essential oil Components by Gas Chromatography/ Mass Spectroscopy. 4th ed. IL: Allured Publishing Corporation; 2007. Adams RP. Identification of Essential oil Components by Gas Chromatography/ Mass Spectroscopy. 4th ed. IL: Allured Publishing Corporation; 2007.
23.
Zurück zum Zitat O’Brien J, Wilson I, Orton T, Pognan F. Investigation of the Alamar Blue (resazurin) fluorescent dye for the assessment of mammalian cell cytotoxicity. Eur J Biochem. 2000;267:5421–6.CrossRefPubMed O’Brien J, Wilson I, Orton T, Pognan F. Investigation of the Alamar Blue (resazurin) fluorescent dye for the assessment of mammalian cell cytotoxicity. Eur J Biochem. 2000;267:5421–6.CrossRefPubMed
24.
Zurück zum Zitat Raz B, Iten M, Grether-Buhler Y, Kaminsky R, Brun R. The Alamar Blue assay to determine drug sensitivity of African trypanosomes (T. b. rhodesiense and T. b. gambiense) in vitro. Acta Trop. 1997;68:139–47.CrossRefPubMed Raz B, Iten M, Grether-Buhler Y, Kaminsky R, Brun R. The Alamar Blue assay to determine drug sensitivity of African trypanosomes (T. b. rhodesiense and T. b. gambiense) in vitro. Acta Trop. 1997;68:139–47.CrossRefPubMed
25.
Zurück zum Zitat Andrade MA, Cardoso MG, Gomes MS, Azeredo CMO, Batista LR, Soares MJ, Rodrigues LMA, Figueiredo ACS. Biological Activity of the Essential Oils from Cinnamodendron dinisii and Siparuna guianensis. Braz J Microbiol. 2015;46:189–94.CrossRefPubMedPubMedCentral Andrade MA, Cardoso MG, Gomes MS, Azeredo CMO, Batista LR, Soares MJ, Rodrigues LMA, Figueiredo ACS. Biological Activity of the Essential Oils from Cinnamodendron dinisii and Siparuna guianensis. Braz J Microbiol. 2015;46:189–94.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Machado RRP, Valente Júnior W, Lesche B, Coimbra ES, Souza NB, Abramo C, et al. Essential oil from leaves of Lantana camara: a potential source of medicine against leishmaniasis. Rev Bras Farmacogn. 2012;22:1011–7.CrossRef Machado RRP, Valente Júnior W, Lesche B, Coimbra ES, Souza NB, Abramo C, et al. Essential oil from leaves of Lantana camara: a potential source of medicine against leishmaniasis. Rev Bras Farmacogn. 2012;22:1011–7.CrossRef
29.
Zurück zum Zitat Gawde A, Cantrell CL, Zheljazkov VD, Astatkie T, Schlegel V. Steam distillation extraction kinetics regression models to predict essential oil yield, composition, and bioactivity of chamomile oil. Ind Crops Prod. 2014;58:61–7.CrossRef Gawde A, Cantrell CL, Zheljazkov VD, Astatkie T, Schlegel V. Steam distillation extraction kinetics regression models to predict essential oil yield, composition, and bioactivity of chamomile oil. Ind Crops Prod. 2014;58:61–7.CrossRef
31.
Zurück zum Zitat Ghannadi A, Amree S. Volatile oil constituents of Ferula gummosa Boiss. from Kashan, Iran. J Essent Oil Res. 2002;14:420–42.CrossRef Ghannadi A, Amree S. Volatile oil constituents of Ferula gummosa Boiss. from Kashan, Iran. J Essent Oil Res. 2002;14:420–42.CrossRef
32.
Zurück zum Zitat Kouyakhi ET, Naghavi MR, Alayhs M. Study of the essential oil variation of Ferula gummosa samples from Iran. Chem Nat Compounds. 2008;44:124–6.CrossRef Kouyakhi ET, Naghavi MR, Alayhs M. Study of the essential oil variation of Ferula gummosa samples from Iran. Chem Nat Compounds. 2008;44:124–6.CrossRef
33.
Zurück zum Zitat Sahebkar A, Iranshahi M. Volatile Constituents of the Genus Ferula (Apiaceae): A Review. J Essent Oil-Bear Plants. 2011;14:504–31.CrossRef Sahebkar A, Iranshahi M. Volatile Constituents of the Genus Ferula (Apiaceae): A Review. J Essent Oil-Bear Plants. 2011;14:504–31.CrossRef
35.
Zurück zum Zitat Dutta S, Mallick M, Kumar K, Mann U, Greenwood PF. Terpenoid composition and botanical affinity of Cretaceous resins from India and Myanmar. Int J Coal Geol. 2011;85:49–55.CrossRef Dutta S, Mallick M, Kumar K, Mann U, Greenwood PF. Terpenoid composition and botanical affinity of Cretaceous resins from India and Myanmar. Int J Coal Geol. 2011;85:49–55.CrossRef
36.
Zurück zum Zitat Rodilla JM, Silva LA, Martinez N, Lorenzo D, Davyt D, Castillo L, Giménez C, Cabrera R, González-Coloma A, Zrostlíkováf J, Dellacassa E. Advances in the identification and agrochemical importance of sesquiterpenoids from Bulnesia sarmientoi essential oil. Ind Crops Prod. 2011;33:497–503.CrossRef Rodilla JM, Silva LA, Martinez N, Lorenzo D, Davyt D, Castillo L, Giménez C, Cabrera R, González-Coloma A, Zrostlíkováf J, Dellacassa E. Advances in the identification and agrochemical importance of sesquiterpenoids from Bulnesia sarmientoi essential oil. Ind Crops Prod. 2011;33:497–503.CrossRef
37.
Zurück zum Zitat Rottini MM, Amaral ACF, Ferreira JLP, Silva JRA, Taniwaki, Souza CSF, d'Escoffier LN, Almeida-Souza F, Hardoim DJ, Costa SCG, Calabrese KS. In vitro evaluation of (−) α-bisabolol as a promising agent against Leishmania amazonensis. Exp Parasitol. 2015;148:66–72.CrossRefPubMed Rottini MM, Amaral ACF, Ferreira JLP, Silva JRA, Taniwaki, Souza CSF, d'Escoffier LN, Almeida-Souza F, Hardoim DJ, Costa SCG, Calabrese KS. In vitro evaluation of (−) α-bisabolol as a promising agent against Leishmania amazonensis. Exp Parasitol. 2015;148:66–72.CrossRefPubMed
38.
Zurück zum Zitat Pitarokili D, Couladis M, Petsikos-Panayotarou N, Tzakou O. Composition and antifungal activity on soil-borne pathogens of the essential oil of Salvia sclarea from Greece. J Agric Food Chem. 2002;50:6688–91.CrossRefPubMed Pitarokili D, Couladis M, Petsikos-Panayotarou N, Tzakou O. Composition and antifungal activity on soil-borne pathogens of the essential oil of Salvia sclarea from Greece. J Agric Food Chem. 2002;50:6688–91.CrossRefPubMed
39.
Zurück zum Zitat Kuźma L, Kalemba D, Różalski M, Różalska B, Więckowska-Szakiel M, Krajewska U, Wysokińska H. Chemical composition and biological activities of essential oil from Salvia sclarea plants regenerated in vitro. Molecules. 2009;14:1438–47.CrossRefPubMed Kuźma L, Kalemba D, Różalski M, Różalska B, Więckowska-Szakiel M, Krajewska U, Wysokińska H. Chemical composition and biological activities of essential oil from Salvia sclarea plants regenerated in vitro. Molecules. 2009;14:1438–47.CrossRefPubMed
40.
Zurück zum Zitat Rosa MSS, Mendonça-Filho RR, Bizzo HR, Rodrigues IA, Soares RMA, Souto-Padrón T, Alviano CS, Lopes AHCS. Antileishmanial activity of a linalool-rich essential oil from Croton cajucara. Antimicrob Agents Chemother. 2003;47:1895–901.CrossRefPubMedCentral Rosa MSS, Mendonça-Filho RR, Bizzo HR, Rodrigues IA, Soares RMA, Souto-Padrón T, Alviano CS, Lopes AHCS. Antileishmanial activity of a linalool-rich essential oil from Croton cajucara. Antimicrob Agents Chemother. 2003;47:1895–901.CrossRefPubMedCentral
41.
Zurück zum Zitat Sadraei H, Ghannadib A, Malekshahia K. Relaxant effect of essential oil of Melissa officinalis and citral on rat ileum contractions. Fitoterapia. 2003;74:445–52.CrossRefPubMed Sadraei H, Ghannadib A, Malekshahia K. Relaxant effect of essential oil of Melissa officinalis and citral on rat ileum contractions. Fitoterapia. 2003;74:445–52.CrossRefPubMed
42.
Zurück zum Zitat Mimica-Dukic N, Bozin B, Sokovic M, Simin N. Antimicrobial and Antioxidant activities of Melissa officinalis L. (Lamiaceae) essential oil. J Agric Food Chem. 2004;52:2485–9.CrossRefPubMed Mimica-Dukic N, Bozin B, Sokovic M, Simin N. Antimicrobial and Antioxidant activities of Melissa officinalis L. (Lamiaceae) essential oil. J Agric Food Chem. 2004;52:2485–9.CrossRefPubMed
43.
Zurück zum Zitat Silva S, Sato A, Lage CLS, Gil S, Silva RA, Azevedo, Almeida D, Esquibel MA. Essential oil composition of Melissa officinalis L. in vitro produced under the influence of growth regulators. J Braz Chem Soc. 2005;16:1387–90.CrossRef Silva S, Sato A, Lage CLS, Gil S, Silva RA, Azevedo, Almeida D, Esquibel MA. Essential oil composition of Melissa officinalis L. in vitro produced under the influence of growth regulators. J Braz Chem Soc. 2005;16:1387–90.CrossRef
44.
Zurück zum Zitat Mikus J, Harkenthal M, Steverding D, Reichling J. In vitro effect of essential oils and isolated mono- and sesquiterpenes on Leishmania major and Trypanosoma brucei. Planta Med. 2000;66:366–8.CrossRefPubMed Mikus J, Harkenthal M, Steverding D, Reichling J. In vitro effect of essential oils and isolated mono- and sesquiterpenes on Leishmania major and Trypanosoma brucei. Planta Med. 2000;66:366–8.CrossRefPubMed
45.
Zurück zum Zitat Santoro GF, Cardoso MG, Guimarães LGL, Freire JM, Soares MJ. Antiproliferative effect of the essential oil of Cymbopogon citratus (DC) Stapf (lemongrass) on intracelular amastigotes, bloodstream trypomastigotes and culture epimastigotes of Trypanosoma cruzi (Protozoa: Kinetoplastida). Parasitology. 2007;13:1649–56. http://www.ncbi.nlm.nih.gov/pubmed/17686189. Accessed 05 Apr 2015. Santoro GF, Cardoso MG, Guimarães LGL, Freire JM, Soares MJ. Antiproliferative effect of the essential oil of Cymbopogon citratus (DC) Stapf (lemongrass) on intracelular amastigotes, bloodstream trypomastigotes and culture epimastigotes of Trypanosoma cruzi (Protozoa: Kinetoplastida). Parasitology. 2007;13:1649–56. http://​www.​ncbi.​nlm.​nih.​gov/​pubmed/​17686189. Accessed 05 Apr 2015.
46.
Zurück zum Zitat Brenzan MA, Nakamura CV, Filho BPD, Ueda-Nakamura T, Young MCM, Cortez DAG. Antileishmanial activity of crude extract and coumarin from Calophyllum brasiliense leaves against Leishmania amazonensis. Parasitol Res. 2007;101:715–22.CrossRefPubMed Brenzan MA, Nakamura CV, Filho BPD, Ueda-Nakamura T, Young MCM, Cortez DAG. Antileishmanial activity of crude extract and coumarin from Calophyllum brasiliense leaves against Leishmania amazonensis. Parasitol Res. 2007;101:715–22.CrossRefPubMed
47.
Zurück zum Zitat Borges AR, Aires JR, Higino TM. Trypanocidal and cytotoxic activities of essential oils from medicinal plants of Northeast of Brazil. Exp Parasitol. 2012;132:123–8.CrossRefPubMed Borges AR, Aires JR, Higino TM. Trypanocidal and cytotoxic activities of essential oils from medicinal plants of Northeast of Brazil. Exp Parasitol. 2012;132:123–8.CrossRefPubMed
Metadaten
Titel
Essential oils: in vitro activity against Leishmania amazonensis, cytotoxicity and chemical composition
verfasst von
Milene Aparecida Andrade
Clênia dos Santos Azevedo
Flávia Nader Motta
Maria Lucília dos Santos
Camila Lasse Silva
Jaime Martins de Santana
Izabela M. D. Bastos
Publikationsdatum
01.12.2016
Verlag
BioMed Central
Erschienen in
BMC Complementary Medicine and Therapies / Ausgabe 1/2016
Elektronische ISSN: 2662-7671
DOI
https://doi.org/10.1186/s12906-016-1401-9

Weitere Artikel der Ausgabe 1/2016

BMC Complementary Medicine and Therapies 1/2016 Zur Ausgabe