Skip to main content
Advertisement
  • Loading metrics

Assessment of sesquiterpene lactones isolated from Mikania plants species for their potential efficacy against Trypanosoma cruzi and Leishmania sp.

  • Laura C. Laurella ,

    Contributed equally to this work with: Laura C. Laurella, Natacha Cerny

    Roles Data curation, Formal analysis, Investigation, Methodology, Writing – original draft

    Affiliation Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Farmacognosia, Buenos Aires, Argentina

  • Natacha Cerny ,

    Contributed equally to this work with: Laura C. Laurella, Natacha Cerny

    Roles Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Writing – original draft

    Affiliation CONICET—Universidad Nacional de Luján, Instituto de Ecología y Desarrollo Sustentable (INEDES), Luján, Argentina

  • Augusto E. Bivona,

    Roles Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Writing – original draft

    Affiliations Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Buenos Aires, Argentina, Instituto de Estudios de la Inmunidad Humoral (IDEHU), UBA-CONICET, Buenos Aires, Argentina, CONICET- Universidad de Buenos Aires, Instituto de Microbiología y Parasitología Médica—CONICET (IMPaM), Facultad de Medicina, Piso 13, Buenos Aires, Argentina

  • Andrés Sánchez Alberti,

    Roles Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Writing – original draft

    Affiliations Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Buenos Aires, Argentina, Instituto de Estudios de la Inmunidad Humoral (IDEHU), UBA-CONICET, Buenos Aires, Argentina, CONICET- Universidad de Buenos Aires, Instituto de Microbiología y Parasitología Médica—CONICET (IMPaM), Facultad de Medicina, Piso 13, Buenos Aires, Argentina

  • Gustavo Giberti,

    Roles Conceptualization, Methodology, Writing – original draft, Writing – review & editing

    Affiliations Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Farmacognosia, Buenos Aires, Argentina, CONICET–Universidad de Buenos Aires, Instituto de Química y Metabolismo del Fármaco—CONICET (IQUIMEFA), Buenos Aires, Argentina

  • Emilio L. Malchiodi,

    Roles Methodology, Supervision, Writing – review & editing

    Affiliations Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Buenos Aires, Argentina, Instituto de Estudios de la Inmunidad Humoral (IDEHU), UBA-CONICET, Buenos Aires, Argentina, CONICET- Universidad de Buenos Aires, Instituto de Microbiología y Parasitología Médica—CONICET (IMPaM), Facultad de Medicina, Piso 13, Buenos Aires, Argentina

  • Virginia S. Martino,

    Roles Conceptualization, Supervision, Writing – review & editing

    Affiliation CONICET–Universidad de Buenos Aires, Instituto de Química y Metabolismo del Fármaco—CONICET (IQUIMEFA), Buenos Aires, Argentina

  • Cesar A. Catalan,

    Roles Conceptualization, Supervision, Writing – review & editing

    Affiliation CONICET–Universidad Nacional de Tucumán, Instituto de Química del Noroeste—CONICET (INQUINOA), Ayacucho 471 (T4000INI), San Miguel de Tucumán, Argentina

  • María Rosario Alonso,

    Roles Conceptualization, Data curation, Methodology, Writing – original draft

    Affiliation CONICET–Universidad de Buenos Aires, Instituto de Química y Metabolismo del Fármaco—CONICET (IQUIMEFA), Buenos Aires, Argentina

  • Silvia I. Cazorla ,

    Roles Conceptualization, Investigation, Methodology, Supervision, Writing – original draft, Writing – review & editing

    vsulsen@ffyb.uba.ar (VPS); silcazorla@yahoo.es (SIC).

    ‡ SIC and VPS also contributed equally to this work.

    Affiliations CONICET- Universidad de Buenos Aires, Instituto de Microbiología y Parasitología Médica—CONICET (IMPaM), Facultad de Medicina, Piso 13, Buenos Aires, Argentina, CONICET–Centro de Referencia para Lactobacilos (CERELA), Batalla de Chacabuco 145, San Miguel de Tucumán, Argentina

  • Valeria P. Sülsen

    Roles Conceptualization, Investigation, Methodology, Project administration, Supervision, Writing – original draft, Writing – review & editing

    vsulsen@ffyb.uba.ar (VPS); silcazorla@yahoo.es (SIC).

    ‡ SIC and VPS also contributed equally to this work.

    Affiliations Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Farmacognosia, Buenos Aires, Argentina, CONICET–Universidad de Buenos Aires, Instituto de Química y Metabolismo del Fármaco—CONICET (IQUIMEFA), Buenos Aires, Argentina

Abstract

Four sesquiterpene lactones, mikanolide, deoxymikanolide, dihydromikanolide and scandenolide, were isolated by a bioassay-guided fractionation of Mikania variifolia and Mikania micrantha dichloromethane extracts. Mikanolide and deoxymikanolide were the major compounds in both extracts (2.2% and 0.4% for Mikania variifolia and 21.0% and 6.4% for Mikania micrantha respectively, calculated on extract dry weight). Mikanolide, deoxymikanolide and dihydromikanolide were active against Trypanosoma cruzi epimastigotes (50% inhibitory concentrations of 0.7, 0.08 and 2.5 μg/mL, for each compound respectively). These sesquiterpene lactones were also active against the bloodstream trypomastigotes (50% inhibitory concentrations for each compound were 2.1, 1.5 and 0.3 μg/mL, respectively) and against amastigotes (50% inhibitory concentrations for each compound were 4.5, 6.3 and 8.5 μg/mL, respectively). By contrast, scandenolide was not active on Trypanosoma cruzi. Besides, mikanolide and deoxymikanolide were also active on Leishmania braziliensis promastigotes (50% inhibitory concentrations of 5.1 and 11.5 μg/mL, respectively). The four sesquiterpene lactones were tested for their cytotoxicity on THP 1 cells. Deoxymikanolide presented the highest selectivity index for trypomastigotes (SI = 54) and amastigotes (SI = 12.5). In an in vivo model of Trypanosoma cruzi infection, deoxymikanolide was able to decrease the parasitemia and the weight loss associated to the acute phase of the parasite infection. More importantly, while 100% of control mice died by day 22 after receiving a lethal T. cruzi infection, 70% of deoxymikanolide-treated mice survived. We also observed that this compound increased TNF-α and IL-12 production by macrophages, which could contribute to control T. cruzi infection.

Author summary

Chagas' disease and Leishmaniasis are parasitic diseases that affect especially poor people in developing countries. They are caused by the protozoan parasites Trypanosoma cruzi and different Leishmania species, respectively. According to the World Health Organization they are considered, among others, Neglected Tropical Diseases. The drugs currently in use for the treatment of these parasitoses are not at all effective and have severe drawbacks. Nature has proved to be a rich source of bioactive compounds. Among antiparasitic drugs, the sesquiterpene lactone artemisinin and the alkaloid quinine and their derivatives are used nowadays for human malaria treatment. Sequiterpenes lactones, present mainly in species from Asteraceae family, are interesting compounds due their pharmacological properties. This group of compounds has shown trypanocidal and leishmanicidal activity and are considered as promising leads for antiparasitic drug discovery.

Introduction

Chagas’ disease is caused by the protozoan parasite Trypanosoma cruzi, and even though over 100 years have passed since its discovery, this disease still represents a serious problem for global health [1]. Thus, this disease, which was traditionally limited to Latin America, has crossed these boundaries and spread worldwide. Increased travelling and migration of individuals from endemic to non-endemic areas has presented a worrisome scenario for congenital infection, blood transfusions and organ transplantations [2, 3]. Trypanosoma cruzi infects a wide range of mammalian hosts, including humans. About six to seven million people are known to be affected by this infection [4]. The alarming numbers would be even greater if the underdiagnosed cases were taken in account. Vector control programs have not been completely successful in preventing parasite transmission [5].

Leishmaniasis is another protozoan parasitic disease caused by Leishmania spp., which presents a worldwide distribution. According to World Health Organization, 300000 cases of visceral leishmaniasis are reported annually, with 200000 deaths One million cases of cutaneous leishmaniasis have been reported in the last five years and 310 million people are at risk of acquiring this parasitosis [6]. Cutaneous leishmaniasis is the most common clinical form in Argentina caused by L. (V.) braziliensis and L. (L.) amazonensis, with some recent reports about human and canine leishmaniasis [7]. Available anti-parasitic drugs for both Chagas’ disease and leishmaniasis are not sufficiently safe or effective [8,9] and no protective vaccines have been developed so far [10].

Natural products are an interesting source of new drugs that might, in the near future, replace current medications, which are known to have severe side effects. Sesquiterpene lactones are a group of natural compounds, mainly found in the Asteraceae family, which show interesting biological activities such as antitumoral, antiinflammatory, antibacterial and antiparasitic, among others [11]. Some of the most representative compounds with antiparasitic activity are artemisinin, parthenolide, costunolide, helenalin, mexicanin, psilostachyins and cynaropicrin [1214].

The Mikania genus (Asteraceae) is found in the tropics of America and Asia and many of its species which are known with the common name of “guaco” are used for treating fever, rheumatism, colds and respiratory diseases, as well as for snake bites and scorpion stings. In recent years, there has been an increasing interest in the study of species from the genus Mikania, since many of its species have been reported to have a wide range of bioactivities [15].

Mikania variifolia Hieron. and M. micrantha Kunth are species native to South America. These species grow in the Northeastern region of Argentina, South of Brazil, Paraguay and Uruguay. In particular, M. micrantha is considered a very invasive weed, growing in cattle fields [16] and has spread throughout Asia. It is used in popular medicine as vulnerary and as antidote [17]. In previous investigations, the isolation of sesquiterpene lactones, flavonoids and caffeoylquinic acids esters have been reported [1821]. Antimicrobial and antiviral activities have also been described for this species [22]. We have previously reported the antiprotozoal and antiviral activities of extracts from M. micrantha. The organic extract of this species has proved to be active against T. cruzi epimastigotes and Leishmania braziliensis promastigotes. The bioassay-guided fractionation of this extract led to the identification of two fractions with trypanocidal activity which showed the presence of sesquiterpene lactones [23].

In this work we describe the isolation of the active constituents of M. variifolia and M. micrantha and the anti-Trypanosoma cruzi and antileishminial activity of the isolated compounds.

Materials and methods

Plant material

The aerial parts of Mikania variifolia Hieron. (Asteraceae) were collected in December 2012 in the province of Entre Rios, Argentina. A voucher specimen (BAF 788) was deposited at the Herbarium of the Museo de Farmacobotánica—Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires. The aerial parts of Mikania micrantha Kunth (Asteraceae) were collected in April 2011 in the province of Tucumán, Argentina. A voucher specimen (LIL609699) was deposited at the Herbarium of Instituto Miguel Lillo, Facultad de Ciencias Naturales, Universidad Nacional de Tucumán.

Parasites

Trypanosoma cruzi epimastigotes (RA strain) were grown in a biphasic medium. Cultures were routinely maintained by weekly passages at 28°C. T. cruzi bloodstream trypomastigotes (RA strain) and the recombinant Tulahuen strain expressing β-galactosidase (Tul-β-Gal) were obtained from infected CF1 mice by cardiac puncture at the peak of parasitemia on day 15 post-infection [24]. Trypomastigotes were routinely maintained by infecting 21-day-old CF1 mice.

Leishmania braziliensis promastigotes (MHOM/BR/75/M2903 strain) were grown in liver infusion tryptose medium LIT, which was prepared as follows: 5 g/L liver infusion (Sigma 2023-072K1066), 5 g/L tryptose (Britania), 2 g/L glucose (Sigma), 68 mM NaCl, 5.4 mM KCl, 22 mM HPO4Na2, supplemented with 20 mg/L hemin (Sigma) and 10% (vol/vol) fetal calf serum (FCS) (Internegocios). Culture maintenance was performed by weekly passages at 26°C.

Preparation of extracts

The extraction of the aerial parts of M. variifolia (400 g) and M. micrantha (100 g) was done by maceration with dichloromethane (8L and 1L, respectively) at room temperature. The organic extracts were filtered and taken to dryness.

Bioassay-guided fractionation of M. variifolia organic extract

The extract residue of M. variifolia was suspended in ethanol:water (70:30) (50 mL) and partitioned successively with hexane (3x40 mL) and dichloromethane (3x40 mL). The dichloromethane fraction (3.5 g) was subjected to open-column chromatography over silica gel 60 with a gradient of dichloromethane/ethyl acetate/methanol, collecting fractions of 70 mL each. Fractions were then combined according to their TLC profile on Silica gel 60 F254 using hexane:ethyl acetate (1:1) as mobile phase, into six final fractions (MV1–MV6). These fractions were subsequently tested for trypanocidal activity against T. cruzi epimastigotes. Compound 1 precipitated from fraction MV3 (eluted with dichloromethane:ethyl acetate (3:1). Compound 2 precipitated from MV4 eluted with dichloromethane:ethyl acetate (2:1). The supernatant from this fraction was subjected to chromatography on a Silica gel column eluted with hexane:ethyl acetate (1:1) and compound 3 precipitated from one of the fractions.

Isolation of compounds from Mikania micrantha

The fractionation procedure of M. micrantha active extract was described previously [23]. The active fractions MM3 and MM4 were combined (1.25 g) and subjected to Silica gel column (51 cm x 3.5 cm, 110 g) chromatography eluted with a gradient of hexane, dichloromethane, ethyl acetate and methanol. Fractions of 20 mL were collected. Compound 1 and compound 2 precipitated from fractions eluted with dichloromethane:ethyl acetate (95:5) and dichloromethane:ethyl acetate (90:10), respectively. Compound 3 crystallized from the supernatant of fractions eluted with dichloromethane:ethyl acetate (90:10). The resulting mother liquors were subjected to preparative TLC on Silica gel 60 F254 using hexane:ethyl acetate (1:1) as mobile phase to obtain compound 4.

Compound identification

The isolated compounds were identified by proton and carbon nuclear magnetic resonance (1HNMR) and 13CNMR, (Bruker Avance 500) (500 MHz, in CDCl3), heteronuclear single quantum correlation (HSQC), heteronuclear multiple bond correlation (HMBC), correlated spectroscopy (COSY), electron impact mass spectrometry (EI-MS) (Thermo Scientific EM/DSQII), infrared spectroscopy (IRS) (Bruker FT-IR IFS 25) and by comparison with literature data and reference compounds. The purity of isolated compounds was checked by HPLC/DAD using a Varian Pro Star instrument equipped with a reversed-phase column Phenomenex–KinetexXB-C18 (250 mm x 4.6 mm and 5 μdp), a Rheodyne valve (20 μL) and a photo diode array detector set at 210 nm. The chromatographic system used was that reported by Laurella et al. [23].

HPLC analysis of M. micrantha and M. variifolia dichloromethane extract and quantification of bioactive compounds

For HPLC analysis, samples were eluted with a gradient of water (A) and acetonitrile (B) from 100% A to 0% A in 60 min and back to initial conditions. A flow rate of 1 mL/min was employed and the separation was done at room temperature (18–25°C). Data were analysed with Varian Star 5.5 (USA). Mikania micranta and M. variifolia organic extracts and pure isolated compounds (used as reference) were dissolved in methanol and water (9:1) at a concentration of 5 mg/mL and 1 mg/mL respectively. Water employed to prepare working solution was of ultrapure quality (Milliq). Methanol and acetonitrile (HPLC) J.T. Baker were used.

In vitro trypanocidal activity

The evaluation of T. cruzi epimastigotes growth inhibition was performed by the [3H] thymidine uptake assay [25]. Briefly, exponentially growing epimastigotes were adjusted to a cell density of 1.5 × 106 parasites/mL in fresh medium. Parasites were allowed to grow for 72 h at 28°C in medium with or without different concentrations of each compound (0–5 μg/mL) in triplicate. Percent of inhibition was calculated as {100 − [(cpm of treated parasites/ cpm of untreated parasites) × 100]}. The compound concentration at which the parasite growth was inhibited by 50% (IC50) was determined after 72 h. Benznidazole was used as positive control.

The trypanocidal effect of the purified compounds was also tested on bloodstream trypomastigotes. Briefly, mouse blood containing trypomastigotes was diluted in complete RPMI -1640 Medium (Sigma-Aldrich R6504- Batch 029K83102) with 10% (vol/vol) fetal calf serum (FCS) (Internegocios) to adjust the parasite concentration to 3 × 105/mL. Parasites were seeded in duplicate in a 96-well microplate in the presence of each compound (0–50 μg/mL) or controls and incubated at 4 °C for 24 h. The number of remaining living parasites in each sample was determined in 5 μL of cell suspension diluted 1/5 in lysis buffer (0.75% NH4Cl, 0.2% Tris, pH 7.2) and counted in a Neubauer chamber. Benznidazole was used as positive control.

The effect of each compound on intracellular forms of T. cruzi was assayed using β-galactosidase transfected parasites [25]. Briefly, 96-well plates were seeded with Raw 264.7 cells at 5x103 per well in 100 mL of culture medium and incubated for 24 h at 37 °C in a 5% CO2 atmosphere. Cells were washed and infected with transfected blood trypomastigotes expressing β-galactosidase at a parasite/cell ratio of 10:1. After 12 h of co- culture, plates were washed twice with PBS to remove unbound parasites and each pure compound was added at different concentrations (0.001–50 μg/mL) in 150 μL of fresh complete RPMI medium without phenol red (Gibco, Rockville, MD). Controls included infected untreated cells (100% infection control) and uninfected cells (0% infection control). The assay was developed 5 days later by the addition of chlorophenol red-β-D-galactopyranoside (CPRG) (100 mM) and 1% Nonidet P40. Plates were then incubated for 4–6 h at 37 °C and the absorbance was measured at 595 nm in a microplate reader (Bio-Rad Laboratories, Hercules, CA). The percentage inhibition was calculated as 100–{[(absorbance of treated infected cells)/(absorbance of untreated infected cells) x 100} and the IC50 value was estimated. Benznidazole was used as positive control.

In vitro leishmanicidal activity

The growth inhibition of Leishmania braziliensis promastigotes was evaluated by the MTT method. Parasites (5x106) were settled at a final volume of 150 μL in a flat-bottom 96-well microplate and cultured at 37°C in a 5% CO2 atmosphere in the absence or presence of increasing concentrations of the pure compounds. After 72 h, 3-(4,5- dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) was added at a final concentration of 1.5 mg/mL. Plates were incubated for 2 h at 37°C. The purple formazan crystals formed were completely dissolved by adding 150 μL of ethanol and the absorbance was read at 595 nm in a microplate reader. The inhibition percentage was calculated as {100 − [(DO595nm of treated parasites/ DO595nm of untreated parasites) × 100]}. Compounds were tested at 0–50 μg/mL. Amphotericin B was used as positive control.

Host cell toxicity

The human monocyte leukemia THP1 (ATCC TIB202) cell line were obtained from the vendor. Cells were thawed, expanded and settled at a concentration 5x105 in a final volume of 150 μL in a flat-bottom 96-well microplate and cultured at 37°C in a 5% CO2 atmosphere in the absence or presence of increasing concentrations of the pure compounds. After 24 h, the cell viability was determined by the trypan blue exclusion method in the absence and presence of increasing concentrations of the compounds [26]. The 50% cytotoxic concentration (CC50) and the selectivity index (SI = CC50/IC50) were determined for each compound for T. cruzi trypomastigotes and amastigotes.

Cytokine production

RAW 264.7 (ATCC TIB71) macrophage cell line were obtained from ATCC and cultured (1×105) in duplicate in 24-well plates in the presence of 2.5 or 25 μg/mL of Compound 3, LPS (10 μg/mL) plus INF-γ (0.30 μg/mL) (positive control), or medium alone (negative control). After 24 h, supernatants were collected and stored at -80 °C until cytokine analysis. IL-12 and TNF-α levels were determined by sandwich ELISA (BD Pharmingen, and R&D System, Minneapolis, MN, respectively) according to manufacturer’s instructions. Supplied standards were used to generate a standard curve.

Animal model of T. cruzi infection

Inbred male Balb/c mice were nursed at the Departamento de Microbiología, Facultad de Medicina, Universidad de Buenos Aires. Groups of six Balb/c male mice (6 to 8 weeks old) were infected with 1000 bloodstream T. cruzi trypomastigotes by the intraperitoneal route. Five days after infection, the presence of circulating parasites was confirmed by the microhematocrit method (Sülsen et al., 2013). Mice were treated daily with compound 3 or benznidazole (1 mg/kg of body weight/day) for five consecutive days (days 4 to 8 post-infection) by the intraperitoneal route. Drugs were resuspended in DMSO and then adjust to concentration in 0.1 M phosphate buffered saline (PBS, pH 7.2); this vehicle was also employed as a negative control. Levels of parasitemia were monitored every 2 days in 5 μL of blood diluted 1:5 in lysis buffer (0.75% NH4Cl, 0.2% Tris, pH 7.2) by counting parasites in a Neubauer chamber. The number of animal deaths was recorded daily.

The weight of each animal was evaluated during the acute phase of infection. Results were expressed as the ratio between the weight on each day and the weight registered the day of the infection multiplied by 100.

Ethics statement

Animal experiments were approved by the Review Board of Ethics of Universidad de Buenos Aires, Facultad de Medicina (Argentina), with the No 2943/2013 and conducted in accordance with the Guide for the Care and Use of Laboratory Animals of the National Research Council [27].

Statistical analysis

Results are presented as means ± SEM. GraphPad Prism 5.0 software (GraphPad Software Inc., San Diego, CA) was employed to carry out calculations. To calculate the IC50 values, the percentages of inhibition were plotted against the drug concentration and fitted with a straight line determined by a linear regression (Sigma Plot 10 software). Results presented are representative of three to four independent experiments.

Parasitemia and weight loss were analyzed using a non parametric test: Mann-Whitney test. The survival curves were analyzed with a log rank test.

The statistical significance was determined by one-way analysis of variance (ANOVA) performed with the GraphPad Prism 5.0 software (GraphPad Software Inc., San Diego, CA). Comparisons were referred to the control group. P values <0.05 were considered significant.

Results

Trypanocidal activity and fractionation of Mikania variifolia dichloromethane extract

The Mikania variifolia dichloromethane extract was evaluated for its trypanocidal activity on T. cruzi epimastigotes. At 100 μg/mL, the extract caused a 97.2% of inhibition of parasite growth. Two active fractions were obtained by bioassay-guided fractionation which caused inhibitions of 99.3% and 96.1%, respectively at 10 μg/mL. Three compounds (1–3) were isolated from these fractions.

Isolation and identification of compounds from fractions 3 and 4 of Mikania micrantha

From the active fractions (MM3 and MM4) of the organic extract of Mikania micrantha, four compounds (1–4) were isolated. Compounds 1–4 were identified by spectroscopic methods and by comparison with literature data [18]. Compound 1 was identified as mikanolide; compound 2 as dihydromikanolide, compound 3 as deoxymikanolide and compound 4 as scandenolide (Fig 1).

thumbnail
Fig 1. Chemical structures of mikanolide, dihydromikanolide, deoxymikanolide and scandenolide.

https://doi.org/10.1371/journal.pntd.0005929.g001

Quantification of mikanolide and deoxymikanolide in M. micrantha and M. variifolia dichloromethane extracts

Two major peaks (rt = 21.4 and 19.9 min) appeared in the HPLC chromatogram of M. micrantha and M. variifolia dichloromethane extracts, corresponding to mikanolide and deoxymikanolide, respectively. The content of these sesquiterpene lactones is shown in Table 1.

thumbnail
Table 1. Quantification of deoxymikanolide and mikanolide in M. micrantha and M. variifolia extracts.

https://doi.org/10.1371/journal.pntd.0005929.t001

Trypanosoma cruzi growth inhibition by isolated compounds

The trypanocidal activity of the isolated compounds was analyzed on T. cruzi epimastigotes. All the compounds were active, with 50% inhibitory concentration (IC50) values of 0.08, 0.7 and 2.5 μg/mL, for deoxymikanolide, mikanolide and dihydromikanolide, respectively. Conversely, scandenolide was not active against epimastigotes (IC50 = 137.2 μg/mL) (Fig 2). For benznidazole, an IC50 of 1.7 μg/mL was registered (S1 Fig).

thumbnail
Fig 2. Inhibition of T. cruzi epimastigotes by mikanolide, dihydromikanolide, deoxymikanolide and scandenolide.

Epimastigotes were adjusted at 1.5x106 parasites/mL and cultured for 72 h at 28°C in the presence of the compounds at a final concentration ranging from 0–50 μg/mL. Growth inhibition of parasites was evaluated by a [3H] thymidine uptake assay. The percentage of inhibition was calculated as 100-[(cpm of treated parasites)/(cpm of untreated parasites)]x100. Values represent mean ± SEM from three independent experiments carried out in triplicate.

https://doi.org/10.1371/journal.pntd.0005929.g002

The trypanocidal activity against bloodstream trypomastigotes was then analyzed for each drug. As shown in Fig 3, an important decrease in the remaining live parasite count was observed for dihydromikanolide, deoxymikanolide and mikanolide, in comparison with controls (IC50 = 0.3, 1.5 and 2.1 μg/mL, respectively). The reference drug (benznidazole) showed an IC50 of 10.4 μg/mL (S1 Fig).

thumbnail
Fig 3. Trypanocidal activities against T. cruzi trypomastigotes of mikanolide, dihydromikanolide, deoxymikanolide and scandenolide.

Bloodstream trypomastigotes were cultured in duplicate in the presence of 0 to 50 μg/mL of mikanolide, dihydromikanolide, deoxymikanolide and scandenolide. Cultures were done in 96-well plates with 3 x 105 parasites/mL during 24 h, and the remaining live parasites were counted in a Neubauer chamber. Symbols represent mean ± SEM.

https://doi.org/10.1371/journal.pntd.0005929.g003

Finally, the inhibition of amastigotes replication was tested on transgenic T. cruzi expressing the β-galactosidase. Mikanolide, deoxymikanolide and dihydromikanolide were active against this replicative form of the parasite with IC50 values of 4.5, 6.3 and 8.5 μg/mL, respectively (Fig 4). The IC50 value for benznidazole was 1.1μg/mL.

thumbnail
Fig 4. Inhibition of T. cruzi amastigotes by mikanolide, dihydromikanolide, deoxymikanolide and scandenolide.

Mammalian cells (5x103 cells/well) were seeded in 96 well plates and infected 24 h later with transfected trypomastigotes expressing β-galactosidase. After 24 h of co-culture, plates were washed and compounds were added at 0–50 μg/mL in 150 μL medium. On day 6 post-infection, the assays were developed by the addition of CPRG (100 mM) and Nonidet P-40 (1%). Plates were incubated for 6 h and quantified at 570 nm. Controls included infected untreated cells (100% infection control). The percentage of inhibition was calculated as 100-{[(Absorbance of treated infected cells)/(Absorbance of untreated infected cells)]x100}.

https://doi.org/10.1371/journal.pntd.0005929.g004

Scandenolide showed IC50 values of 68.4 and 60.6 μg/mL on trypomastigotes and amastigotes, respectively, therefore it was not active against this replicative form.

Antileishmanial activity of isolated compounds

A moderate activity against L. braziliensis promastigotes was observed with mikanolide, deoxymikanolide and dihydromikanolide (IC50 values of 5.1, 11.5 and 57.1 μg/mL, respectively). By contrast, no activity was registered for scandenolide (IC50 = 252.0 μg/mL) (Fig 5). The reference drug, amphotericin B, presented an IC50 value of 0.26 μg/mL (S2 Fig).

thumbnail
Fig 5. Leishmanicidal activity on L. braziliensis promastigotes of mikanolide, dihidromikanolide, deoxymikanolide and scandenolide.

Parasites were cultured for 72 h in the presence of the compounds (0–50 μg/mL). Growth inhibition of parasites was evaluated by a MTT assay. The percent of inhibition was calculated as {100 − [(DO595nm of treated parasites/ DO595nm of untreated parasites) × 100]}. Values represent mean ± SEM from three independent experiments carried out in triplicate.

https://doi.org/10.1371/journal.pntd.0005929.g005

Cytotoxicity

The in vitro cytotoxic effect of each compound was evaluated on a human cell line by the trypan blue exclusion method. Viable human cells were incubated in the absence and presence of increasing concentrations of the compounds. Deoxymikanolide presented a CC50 value of 79.37 μg/mL, being the SI for T. cruzi trypomastigotes and amastigotes 54 and 12.5, respectively. These SI values were higher than those obtained for mammalian cells. CC50 values for dihydromikanolide and mikanolide were 12.98 and 22.3 μg/mL, respectively. Selectivity indexes for these STLs were 50 and 1.52 (dihydromikanolide), and 10.66 and 4.96 (mikanolide), for trypomastigotes and amastigotes, respectively. Selectivity indexes of the compounds for L. braziliensis were lower than 10 (Table 2).

thumbnail
Table 2. Cytotoxicity on human monocyte leukemia THP1 and selectivity indexes of the isolated compounds.

https://doi.org/10.1371/journal.pntd.0005929.t002

Treatment of Trypanosoma cruzi infected mice with deoxymikanolide

Since deoxymikanolide presented good selectivity on differents T. cruzi stages, this compound was selected for an in vivo study. Balb/c mice were infected with a lethal dose of T. cruzi (RA strain) and treated for 5 consecutive days with either deoxymikanolide or the vehicle. As shown in Fig 6A, infected mice that received deoxymikanolide presented a lower blood parasitemia, as compared to controls. Moreover, in terms of area under the parasitemia curve, a 1.5-fold reduction was observed in deoxymikanolide-treated mice with respect to controls; whereas a 2.3-fold was observed for benznidazole-treated mice (S1 Fig). Interestingly, the treatment with deoxymikanolide was able to reduce the weight loss observed during the acute phase of infection, as compared to control mice (p = 0.028, Mann-Whitney test) at 20 days post infection (dpi) (Fig 6B). More importantly, in deoxymikanolide-treated mice, a significant decrease in the mortality caused by T.cruzi infection was observed (p = 0.02, Log-rank test). While nearly 70% of deoxymikanolide-treated mice survived the acute phase of infection, 100% mortality was observed in control mice by day 22 post-infection (Fig 6C). These results highlight the in vivo efficacy of deoxymikanolide to induce the killing of circulating trypomastigotes during the acute infection.

thumbnail
Fig 6. In vivo deoxymikanolide trypanocidal activity.

Balb/c mice infected with a lethal dose of T. cruzi were treated for 5 consecutive days (days 4 to 8 post-infection) with either deoxymikanolide or with vehicle (phosphate buffered saline). Parasitemia (A), body weight loss (B), and Survival (C), during the acute phase of the parasite infection were analyzed. Results presented are representative of three independent experiments. *p = 0.028, Mann- Whitney test.

https://doi.org/10.1371/journal.pntd.0005929.g006

Cytokine production

We further studied in vitro the ability of deoxymikanolide to modulate the host’s immune response, particularly, macrophage cells, which are the first line of defense against T. cruzi infection. Upon macrophage stimulation with 25 μg/mL deoxymikanolide, a significant increase in the secretion of TNF-α was observed as compared with the negative control (p<0.01). Moreover, a slight increase in the IL-12 production was observed when deoxymikanolide was used at 2 μg/mL and a higher production rate was observed when this STL was tested at 25 μg/mL (Fig 7).

thumbnail
Fig 7. Cytokine secretion induced by deoxymikanolide on macrophages.

Cells (1×105cells/mL) were cultured in the presence of deoxymikanolide (2 and 25 μg/mL) for 24 h. Cells were incubated with LPS (1 μg/mL) plus INF-γ (1000 pg/mL) as positive control of macrophage IL-12 and TNF-α production. Cytokines were assayed by ELISA. Data are expressed as mean ± SEM and are representative of two independent experiments.

https://doi.org/10.1371/journal.pntd.0005929.g007

Discussion

Four sesquiterpene lactones of the germacranolide type have been isolated and identified from M. variifolia and M. micrantha organic extracts. Mikanolide, dihydromikanolide and deoxymikanolide were identified in M. variifolia, while the same compounds together with scandenolide were found to be present in M. micrantha. All the compounds, with the exception of scandenolide, showed high activity on T. cruzi epimastigotes. Mikanolide, deoxymikanolide and dihydromikanolide were also active against trypomastigote and amastigote forms.

Due to the existence of coinfections with both T. cruzi and Leishmania spp., the isolated compounds were also tested in vitro against L. braziliensis, the main agent of tegumentary leishmaniasis in Argentina [28]. Mikanolide and deoxymikanolide showed significant activity against L. braziliensis promastigotes, while dihydromikanolide displayed moderate activity. On the other hand, scandenolide was not active against this parasite stage.

The major determinant for the antiprotozoal activity of STLs would be the presence of an α,β- unsaturated lactone group in the sesquiterpene lactone structure [29]. Nevertheless, some of α-santonin derivatives, lacking the exomethylene group, have shown trypanocidal activity [30]. Steric and electronic factors could also influence the anti-T. cruzi activity of STLs [31].

The four isolated compounds differ from each other in the number of epoxy groups, in the presence or absence of the exocyclic double bond and in the presence of an OAc group at C-3. In comparing the results of activity obtained for mikanolide and dihydromikanolide on the different T. cruzi stages, the absence of the exocyclic double bond in dihydromikanolide with respect to mikanolide, does not seem to affect its activity against this parasite. Despite the different number of epoxy groups present in mikanolide and deoxymikanolide structures, both compounds showed similar activity on this parasite.

On the other hand, the presence of the exocyclic double bond seems to be important for the leishmanicidal activity, since mikanolide and deoxymikanolide were active, while dihydromikanolide was moderately active.

Scandenolide did not show any antiprotozoal activity, suggesting that the presence of an OAc at C-3 could be responsible for the decrease of such activity.

A strong correlation between biological activity and cytotoxicity of STLs has been reported [29]. Although many of them display a considerable cytotoxic activity against mammalian cells, some of these compounds show more selectivity against the parasites [31]. Deoxymikanolide presented a CC50 that was approximately four times higher than mikanolide, thus proving to be more selective for the parasite.

There are previous reports describing the presence of mikanolide, dihydromikanolide, deoxymikanolide and scandenolide in M. micrantha [18]. Although these sesquiterpene lactones have been evaluated for other activities [20, 22, 32], this is the first study that describes their trypanocidal and leishmanicidal effects. Herein, the STLs of M. variifolia have been isolated for the first time.

An optimal response to treatment of trypanosomatid diseases is strongly linked to an efficient immune response. This response is mediated by the activation of macrophages with consequent production of nitrogen and oxygen intermediates that are toxic to the parasite, as well as the activation of T helper and cytotoxic cells producing INF-γ [10, 3336]. The biological activities of lactones, including sesquiterpenoids are largely due to their inhibitory effects on the activity of a plethora of enzymes of prokaryotic organisms and eukaryotic cells [37]. Sesquiterpene lactones have proved to be potent inhibitors of different transcription factors [38, 39]. The best recognized interference of this type of compounds is the inhibition of multiple factors within the nuclear factor-κB (NF-κB) signaling system [40, 41]. These effects are presumed to be related to molecular mechanisms determining the immune activity of sesquiterpene lactones. In that sense, it has been previously reported that a guaianolide type sesquiterpene lactone with a lactone-diol moiety stimulates production of interleukin-8 (IL-8) [42] and up-regulates the lipopolysaccharide (LPS)-induced production of pro-inflammatory cytokines such as IL-6 and tumor necrosis factor-α (TNF-α). Harmatha et al. [43], have demonstrated an increase in NO production and in cytokines such as IFN-γ, IL-1β, IL-6, VEGF and GM-CSF in a range of low concentrations of 10 nM-10 mM by a guaianolide type sesquiterpene lactone. Based on its activity and selectivity, we selected deoxymikanolide for the evaluation of its immunomodulatory activity on macrophage cells, which are the first line of defense against T. cruzi and Leishmania infection. The results obtained suggest that, besides the antiprotozoal activity that deoxymikanolide has per se, this compound could stimulate the host´s immune system through the secretion of cytokines, thus exerting a protective effect against parasite infection.

Conclusion

Four sesquiterpene lactones, mikanolide, deoxymikanolide, dihydromikanolide and scandenolide were isolated from M. variifolia and M. micrantha. With the exception of scandenolide, all the sesquiterpene lactones showed trypanocidal and leishmanicidal activities. Deoxymikanolide was the most promising compound based on its activity, selectivity and its capacity to stimulate cytokine production. Besides, this compound was also active in a murine model of T. cruzi infection. This finding makes it an interesting lead molecule which may be useful for the development of new drugs, alone or in combination with actual therapy, for the treatment of trypanosomatid diseases, in order to reduce side effects.

Supporting information

S1 Fig.

Trypanocidal activity of the reference drug benznidazol determined by in vitro assays against: epimastigotes (A), trypomastigotes (B) and amastigotes (C) of T. cruzi. In vivo treatment with benznidazol in a murine model of T. cruzi infection (D). Area under curve (AUC) was determined. *p<0.01, ***p<0.001.

https://doi.org/10.1371/journal.pntd.0005929.s001

(TIF)

S2 Fig. Leishmanicidal in vitro activity of the reference drug amphotericin B against promastigotes of L. braziliensis.

https://doi.org/10.1371/journal.pntd.0005929.s002

(TIF)

Acknowledgments

This investigation is part of the activities within the “Research Network Natural Products against Neglected Diseases” (ResNet NPND): http://www.resnetnpnd.org/.

References

  1. 1. Andrade DV, Gollob KJ, Dutra WO. Acute Chagas disease: new global challenges for an old neglected disease. PLoS Negl Trop Dis. 2014; 8(7):e3010. pmid:25077613
  2. 2. Moncayo A, Silveira A. Current epidemiological trends for Chagas disease in Latin America and future challenges in epidemiology, surveillance and health policy. Mem Inst Oswaldo Cruz. 2009; 104:17–30. pmid:19753454
  3. 3. Sánchez LV, Ramírez JD. Congenital and oral transmission of American trypanosomiasis: an overview of physiopathogenic aspects. Parasitology. 2013; 140: 147–159. pmid:23010131
  4. 4. World Health Organization. Fact sheet Chagas disease (American trypanosomiasis). 2017. Available from: http://www.who.int/mediacentre/factsheets/fs340/en/.
  5. 5. Camargo EP. Perspectives of vaccination in Chagas disease revisited. Mem Inst Oswaldo Cruz. 2009; 104:1275–1280.
  6. 6. World Health Organization. Lesihmaniasis. 2017. Available from: http://www.who.int/leishmaniasis/en/.
  7. 7. Gould IT, Perner MS, Santini MS, Saavedra SB, Bezz G, Maglianese MI, et al. Leishmaniasis visceral en la argentina notificación y situación vectorial (2006–2012). Medicina. 2013; 73:104–110.
  8. 8. Espuelas S, Plano D, Nguewa P, Font M, Palop JA, Irache JM, et al. Innovative lead compounds and formulation strategies as newer kinetoplastid therapies. Curr Med Chem. 2012; 19:4259–4288. pmid:22834813
  9. 9. Drugs for Neglected Disease Initiative (DNDi). About Leishmaniasis. 2017. Available from: http://www.dndi.org/diseases-projects/leishmaniasis/.
  10. 10. Cazorla SI, Frank FM, Malchiodi EL. Vaccination approaches against Trypanosoma cruzi infection. Expert Rev Vaccines. 2009; 8:921–935. pmid:19538117
  11. 11. Chaturvedi D. Sesquiterpene lactones: Structural diversity and their biological activities. In: Tiwari B, Mishra B, editors. Opportunity, challenge and scope of natural products in medicinal chemistry; 2011. pp. 313–334.
  12. 12. Saeidnia S, Gohari AR, Haddadi A. Biogenic trypanocidal sesquiterpenes: lead compounds to design future trypanocidal drugs—a mini review. DARU J Pharm Sci. 2013; 21:1–9.
  13. 13. Schmidt T, Khalid S, Romanha A, Alves T, Biavatti M, Brun R, 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. pmid:22414103
  14. 14. da Silva CF, Batista D da G, De Araújo JS, Batista MM, Lionel J, de Souza EM, et al. Activities of psilostachyin A and cynaropicrin against Trypanosoma cruzi in vitro and in vivo. Antimicrob Agents Chemother. 2013; 57:5307–5314. pmid:23939901
  15. 15. Rufatto LC, Gower A, Schwambach J, Moura S. Genus Mikania: chemical composition and phytotherapeutical activity. Rev Bras Farmacogn. 2012; 22:1384–1403.
  16. 16. Marzocca A. Vademecum de Malezas Medicinales. Buenos Aires: Orientacion Gráfica Editor; 1997.
  17. 17. Toursarkissian M. Plantas Medicinales de la Argentina. Buenos Aires: Ed. Hemisferio Sur; 1980.
  18. 18. Cuenca MDR, Bardon A, Catalán CAN, Kokke WCMC. Sesquiterpene lactones from Mikania micrantha. J Nat Prod. 1988; 51:625–626. pmid:21401180
  19. 19. Wei X, Huang H, Wu P, Cao H, Ye W. Phenolic constituents from Mikania micrantha. Biochem Syst Ecol. 2004; 32:1091–1096.
  20. 20. Ríos EV, León A, Chávez MI, Torres Y, Ramírez-Apan MT, Toscano RA, et al. Sesquiterpene lactones from Mikania micrantha and Mikania cordifolia and their cytotoxic and anti-inflammatory evaluation. Fitoterapia. 2014; 94:155–163. pmid:24565963
  21. 21. Xu Q, Xie H, Xiao H, Wei X. Phenolic constituents from the roots of Mikania micrantha and their allelopathic effects. J Agric Food Chem. 2013; 61:7309–7314. pmid:23822807
  22. 22. Li Y, Li J, Wang XX, Cao AC. Antimicrobial constituents of the leaves of Mikania micrantha H.B.K. PLoS One. 2013; 8(10):e76725. pmid:24098556
  23. 23. Laurella LC, Frank FM, Sarquiz A, Alonso MR, Giberti G, Cavallaro L, et al. In vitro evaluation of antiprotozoal and antiviral activities of extracts from argentinean Mikania species. Scientific World Journal. 2012; 2012:121253. pmid:22919289
  24. 24. Buckner FS, Verlinde CL, La Flamme AC, Van Voorhis WC. Efficient technique forscreening drugs for activity against Trypanosoma cruzi using parasites express-ing beta-galactosidase. Antimicrob Agents Chemother. 1996; 40:2592–2597. pmid:8913471
  25. 25. Sülsen VP, Cazorla SI, Frank FM, Laurella LC, Muschietti LV, Catalán CV, et al. Natural terpenoids from Ambrosia species are active in vitro and in vivo against human pathogenic trypanosomatids. PLoS Negl Trop Dis. 2013; 7:e2494. pmid:24130916
  26. 26. Sülsen VP, Cazorla SI, Frank FM, Redko F, Anesini C, Coussio J, et al. Trypanocidal and leishmanicidal activities of flavonoids from Argentine medicinal plants. Am J Trop Med Hyg. 2007; 77:654–659. pmid:17978066
  27. 27. National Research Council. Guide for the care and use of laboratory animals. 8th ed. Washington DC: National Academy Press; 2011.
  28. 28. Frank FM, Fernández MM, Taranto N, Cajal SP, Margni RA, Soccol V, et al. Characterization of human infection by Leishmania spp. in the Northwest of Argentina: immune response, double infection with T. cruzi and species of Leishmania involved. Parasitology. 2003; 129:1–9.
  29. 29. Schmidt T, Nour A, Khalid S, Kaiser M, Brun R. Quantitative structure -antiprotozoal activity relationships of sesquiterpene lactones. Molecules. 2009; 14: 2062–2076. pmid:19513006
  30. 30. Otoguro K, Iwatsuki M, Ishiyama A, Namatame M, Nishihara-Tukashima A, Kiyohara H, et al. In vitro antitrypanosomal activity of plant terpenes against Trypanosoma brucei. Phytochemistry. 2011; 72:2024–2030. pmid:21843897
  31. 31. Fabian L, Sülsen V, Frank F, Cazorla S, Malchiodi E, Martino V, et al. In silico study of structural and geometrical requirements of natural sesquiterpene lactones with trypanocidal activity. Mini Rev Med Chem. 2013; 13:1407–1414. pmid:23815577
  32. 32. But PP, He ZD, Ma SC, Chan YM, Shaw PC, Ye WC, et al. Antiviral constituents against respiratory viruses from Mikania micrantha. J Nat Prod. 2009; 72:925–928. pmid:19267453
  33. 33. Aliberti JC, Souto JT, Marino AP, Lannes-Vieira J, Teixeira MM, Farber J, et al. Modulation of chemokine production and inflammatory responses in interferon-gamma- and tumor necrosis factor-R1-deficient mice during Trypanosoma cruzi infection. Am J Pathol. 2001; 58:1433–1440.
  34. 34. Kumar S, Tarleton RL. Antigen-specific Th1 but not Th2 cells provide protection from lethal Trypanosoma cruzi infection in mice. J Immunol. 2001; 166:4596–4603. pmid:11254717
  35. 35. Croft SL, Vivas L, Brooker S. Recent advances in research and control of malaria, leishmaniasis, trypanosomiasis and schistosomiasis. East Mediterr Health J. 2001; 9:518–533.
  36. 36. Frank FM, Petray PB, Cazorla SI, Muñoz MC, Corral RS, Malchiodi EL. Use of a purified Trypanosoma cruzi antigen and CpG oligodeoxynucleotides for immunoprotection against a lethal challenge with trypomastigotes. Vaccine. 2003; 22:77–86. pmid:14604574
  37. 37. Konaklieva MI, Plotkin BJ. Lactones: generic inhibitors of enzymes? Mini Rev Med Chem. 2005; 5:73–95. pmid:15638793
  38. 38. Dirsch VM, Stuppner H, Ellmerer-Müller EP, Vollmar AM. Structural requirements of sesquiterpene lactones to inhibit LPS-induced nitric oxide synthesis in RAW 264.7 macrophages. Bioorg Med Chem. 2000; 8:2747–2753. pmid:11131166
  39. 39. Gertsch J, Sticher O, Schmidt T, Heilmann J. Influence of helenanolide-type sesquiterpene lactones on gene transcription profiles in Jurkat T cells and human peripheral blood cells: anti-inflammatory and cytotoxic effects. Biochem Pharmacol. 2003; 66:2141–2153. pmid:14609739
  40. 40. Siedle B, García-Piñeres AJ, Murillo R, Schulte-Mönting J, Castro V, Rüngeler P, et al. Quantitative structure-activity relationship of sesquiterpene lactones as inhibitors of the transcription factor NF-kappaB. J Med Chem. 2004; 47:6042–6054. pmid:15537359
  41. 41. Salminen A, Lehtonen M, Suuronen T, Kaarniranta K, Huuskonen J. Terpenoids: natural inhibitors of NF-kappaB signaling with anti-inflammatory and anticancer potential. Cell Mol Life Sci. 2008; 65:2979–2999. pmid:18516495
  42. 42. Gewirtz AT, Rao AS, Simon PO Jr, Merlin D, Carnes D, Madara JL, et al. Salmonella typhimurium induces epithelial IL-8 expression via Ca(2+)-mediated activation of the NF-kappaB pathway. J Clin Invest. 2000; 105:79–92. pmid:10619864
  43. 43. Harmatha J, Buděšínský M, Vokáč K, Kostecká P, Kmoníčková E, Zídek Z.Trilobolide and related sesquiterpene lactones from Laser trilobum possessing immunobiological properties. Fitoterapia. 2013; 89:157–166. pmid:23747321