Next Article in Journal
Phytochemicals and Regulation of NF-kB in Inflammatory Bowel Diseases: An Overview of In Vitro and In Vivo Effects
Next Article in Special Issue
Chemical Characterization and Leishmanicidal Activity In Vitro and In Silico of Natural Products Obtained from Leaves of Vernonanthura brasiliana (L.) H. Rob (Asteraceae)
Previous Article in Journal
The Combined Effect of Licorice Extract and Bone Marrow Mesenchymal Stem Cells on Cisplatin-Induced Hepatocellular Damage in Rats
Previous Article in Special Issue
Curcumin Decreases Viability and Inhibits Proliferation of Imatinib-Sensitive and Imatinib-Resistant Chronic Myeloid Leukemia Cell Lines
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

In Vitro Activity of Essential Oils from Piper Species (Piperaceae) against Tachyzoites of Toxoplasma gondii

by
Adalberto Alves Pereira Filho
1,*,
Mariana Maciel Cunha
1,
Mariana Alves Stanton
2,
Lydia Fumiko Yamaguchi
2,
Massuo Jorge Kato
2,*,† and
Érica S. Martins-Duarte
1,*,†
1
Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
2
Department of Fundamental Chemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, São Paulo, Brazil
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Submission received: 21 November 2022 / Revised: 16 December 2022 / Accepted: 2 January 2023 / Published: 6 January 2023
(This article belongs to the Special Issue The Natural Products in the Treatment and Prevention of Diseases)

Abstract

:
Toxoplasmosis is a tropical and neglected disease caused by the parasitic protozoa Toxplasma gondii. Conventional treatment with sulfadiazine and pyrimethamine plus folinic acid, has some drawbacks, such as inefficacy in the chronic phase, toxic side effects, and potential cases of resistance have been observed. In this study, the activity of essential oils (EOs) from three Piper species and their main constituents, including α-Pinene (Piper lindbergii and P. cernuum), β-Pinene (P. cernuum), and dillapiole (P. aduncum), were evaluated against tachyzoites of T. gondii. α-Pinene was more active [(IC50 0.3265 (0.2958 to 0.3604) μg/mL)] against tachyzoites than P. lindbergii EO [0.8387 (0.6492 to 1.084) μg/mL]. Both α-Pinene and P. lindbergii EO exhibited low cytotoxicity against NHDF cells, with CC50 41.37 (37.64 to 45.09) µg/mL and 83.80 (75.42 to 91.34) µg/mL, respectively, suggesting they could be of potential use against toxoplasmosis.

1. Introduction

Toxoplasma gondii (Nicolle and Manceaux, 1909) is an apicomplexan parasite present in approximately one-third of the human population worldwide. In addition to humans, it can infect practically all warm-blooded vertebrates. This zoonotic infection represents an important public health problem in human and veterinary medicine [1]. Transmission occurs mainly by ingestion of oocysts in the environment and of tissue cysts in raw or undercooked meat. Although most people affected are asymptomatic, serious cases can occur in congenitally infected newborns and in immunocompromised patients [2,3].
Toxoplasmosis treatment usually consists of a combination of sulfadiazine and pyrimethamine plus folinic acid. This combination has a synergic action and traditionally shows good results in the acute stage of infection [4,5]. However, these and the other currently recommended drugs for toxoplasmosis treatment have limitations. A notable limitation is that one of the mechanisms of action involves the reduction innucleic acid synthesis, which makes a teratogenic drug. Adverse effects, resistance, and intolerance against these and other known treatments are commonly reported in the literature. In addition, all drugs are inefficient against the chronic phase of infection. These limitations affect the success of the treatments, mainly in immunocompromised patients and in ocular and congenital cases, which raises the need for new treatment options [6,7,8,9,10,11].
In the search for alternatives to the treatment of toxoplasmosis, essential oils (EOs) can represent an excellent source of mixtures of biologically active natural products. In this context, the genus Piper L. (Piperaceae) is one of the most diverse and widely distributed plant groups in pantropical regions, with approximately 1000 species worldwide and several reported cases of use in traditional medicine. Piper species are aromatic plants and have many metabolites with demonstrated biological effects on human health, and therefore they represent a potential source of pharmacologically active substances.
In addition, EOs from Piper species have demonstrated antiparasitic activity against several medically important protozoa. Here, it is worth reporting some studies that have already been carried out against Plasmodium falciparum [12,13], Trypanosoma cruzi [12,13,14,15,16], and several species of the genus Leishmania [15,17]. Despite the absence of studies using EOs from the Piper genus against T. gondii, it is known that the ethanol extract from Piper betle L. resulted in 100% survival of infected mice with this apicomplexan parasite [18].
Due to the urgent need to find and develop new treatments for toxoplasmosis, the objective of the present work was to evaluate the potential of EOs from species of the genus Piper (Piperaceae) and some of their major compounds for use against T. gondii.

2. Materials and Methods

2.1. Parasites

The highly virulent T. gondii RH strain (Type I) was maintained in vitro through serial passages in 25 cm2 culture flasks of confluent neonatal normal human dermal fibroblast cells (NHDF (Lonza®, Catalog: CC-2509)) in complete RPMI medium (RPMI 1640 medium, Gibco, 2% fetal bovine serum, 4 mM of glutamine, 100 U/mL of penicillin, 100 μg/mL of streptomycin and 25 μg/mL of fungizone, Gibco). Culture flasks were maintained at 37 °C and 5% CO2.

2.2. Plant Materials and Extraction of Essential Oils

The leaves of three species of plants belonging to the genus Piper with their respective identification vouchers in parentheses: Piper aduncum L. (K-0057), Piper cernuum Vell. (K-0137) and Piper lindbergii C.DC. (K-2325), were collected in the period of January to June 2018. The vouchers were identified by Dr. Eric Tepe and deposited at the Herbarium of USP, University of São Paulo for identification, and all collections were made under permits #59161-1 and 010/2018-R from SISBIO (Sistema de Autorização e Informação em Biodiversidade) and Fundação Serra do Japi, respectively.
The EOs were extracted from fresh leaves of each species, submitted to hydrodistillation in a Clevenger-type apparatus for 4 h, using 300–500 g of fresh leaves and 500 mL of distilled water [19,20]. The EOs were collected and dried with anhydrous sodium sulfate and stored in amber bottles in a refrigerator at 4 °C until the experiments and analysis by GC-MS and their main constituents were identified based on library search, retention index (RI), and use of standard compounds when available, and expressed as relative percentage of each constituent according to the method described [21].
The major compounds from EOs characterized by GC-MS and used in this study were: α-Pinene (Sigma-Aldrich: 147524) and β-Pinene (Sigma-Aldrich: 402753) and were acquired commercially. Pure dillapiole was obtained by fractionation using the Isolera Flash Chromatography system (Biotage INC) according to the method described [21].

2.3. Antiproliferative RH Strain Tachyzoites Delayed-Death Assays

NHDF cells were maintained in 6-well culture plates at 37 °C and 5% CO2. After verifying the formation of NHDF monolayers, the old medium was removed and a new complete RPMI-1640 medium was added, adding either EOs of Piper or the major compounds once, at final concentrations of 0.5, 1, 2.5, 5, 10, 15, 20 μg/mL. Subsequently, previously counted tachyzoites of the RH strain were added to achieve an amount of 800 tachyzoites/well. The plates were left for 7 days at 37 °C and 5% CO2. After verifying the infection of NHDF monolayers by T. gondii tachyzoites, the culture medium was discarded, and the plates were fixed with 99% ethanol and stained with crystal violet to visualize plaque formation. The plates were then photographed on an Alpha DigiDocphoto document. Images were processed with ImageJ software to determine plaque area. The areas of each well of the plate treated with EOs of Piper or major compounds were compared with the control treated only with 0.1% DMSO and the percentages of proliferations were calculated to obtain the respective 50% effective concentrations (EC50) (Figure 1).

2.4. Invasion Assay of NHDF Cells by T. gondii Tachyzoites

Cultures of the RH strain that had completely lysed an NHDF monolayer were recollected into a centrifuge tube. Additionally, 1 × 106 extracellular tachyzoites resuspended in RPMI medium were preincubated for 2 h at 37 °C and 5% CO2 based on the EC50 of each compound or of the vehicle (DMSO 0.1%). Parasites were centrifuged at 6000 rpm × 5 min three times, resuspended in RPMI medium, and then allowed to invade NHDF cell monolayers previously grown on coverslips in 24-well plates by incubation for 2 h at 37 °C. The cultures were then washed to remove extracellular parasites and fixed with Bouin for 20 min, washed again with 70% alcohol, and stained with solutions 2 and 3 of the Fast Panoptic kit (Laborclin) for 45 and 10 s, respectively. The glass slides were observed under the optical microscope, cells were counted, and the percentage of infected cells was calculated from the ratio of infected cells to uninfected cells.

2.5. Host Cell Viability

The viability of NHDF cells was evaluated by MTS/PMS colorimetric assay. The cells were treated once with Piper EOs or their major compounds (dillapiole, α-Pinene, and β-Pinene) at final concentrations of 12.5, 25, 50, 100, and 200 μg/mL. For that, fibroblasts were seeded in 96-well plates at a density of 1 × 105 cells/well. After semi-confluence, cells were incubated with complete RPMI medium in the absence or presence of EOs or of single compounds for 7 days at 37 °C and 5% CO2. At the end of the incubation time, the cells were washed with sterile PBS (pH 7.2), and the wells filled with 100 μL of PBS + 10 mM of glucose and 20 μL of the MTS/PMS reagent (20:1), from a stock solution of 2 mg/mL MTS and 0.92 mg/mL PMS diluted in PBS (Promega, Madison WI, USA). After 2 h of incubation, the absorbance of the samples was detected at 490 nm using the Versamax ELISA plate reader (Molecular Devices) and cytotoxicity was calculated as the percentage of viable cells in relation to control cells. The cytotoxic concentration of 50% (CC50) for the host cells was determined, and the selective index (SI) was calculated as the ratio of CC50/EC50 [22].

2.6. Statistical Analysis

The data were organized in spreadsheets using Microsoft Excel software (Office 2007). The obtained data were subjected to non-linear regression to obtain 50% effective concentrations (EC50) and cytotoxic concentration (CC50), with 95% confidence intervals using the GraphPad Prism 7.0 software (GraphPad Inc., San Diego, CA, USA). Comparisons between groups (control group and treated groups) were carried out using Kruskal–Wallis followed by Dunn’s posthoc test. Statistical significance was defined as p < 0.05.

3. Results

3.1. Chemical Composition of Essential Oils (EO)

The chemical analysis of the EOs obtained from Piper aduncum, P. cernuum and P. lindbergii identified 34, 35, and 44 compounds, respectively, as shown in Table 1. In summary, the major compounds were identified as phenylpropanoids, sesquiterpenes, and monoterpenes. From the analysis by GC-MS, some major compounds of the EOs were selected to be used in the tests: α-Pinene, β-Pinene, and Dillapiole.

3.2. In Vitro Antiproliferative Effect of Piper EOs and Major Compounds on RH Strain Tachyzoites

The antiproliferative effect of Piper EOs and three major compounds against tachyzoites of the T. gondii RH strain were evaluated after 7 days of treatment under the same conditions as described above in Section 2.3. All compounds and EOs evaluated were able to inhibit the proliferation of T. gondii and had their effective concentrations calculated (Table 2). The EO from P. lindbergii was the most active mixture and inhibited the proliferation of T. gondii with an EC50 = 0.839 µg/mL, in contrast to the P. cernuum EO, which presented the highest EC50 = 3.687 µg/mL. As for the major components, α-Pinene showed the lowest EC50 = 0.326 µg/mL, followed by β-Pinene EC50 = 1.145 µg/mL, and finally dillapiole with EC50= 4.827 µg/mL. Excepting P. cernuum EO, all tested compounds and EOs were able to inhibit T. gondii proliferation in a dose-dependent manner (Figure 2A–F).

3.3. Cytotoxicity Evaluation of Piper EOs and Major Compounds in NHDF Cells

To verify whether Piper EOs and major compounds inhibit T. gondii proliferation without exerting a secondary toxic effect against the host cell, NHDF monolayers were treated with different concentrations (12.5–220 µg/mL) for 7 days under the same conditions as in Section 2.5 above. The selectivity index (SI) showed that Piper EOs and major compounds have a wide selectivity index ranging from 46 to 126 (Table 3). P. lindbergii and α-Pinene showed the highest levels of selectivity, 99 and 126 SI, respectively. Except for Dillapiole, which affected the cytotoxicity in a dose-dependent manner, all the other compounds and EOs showed cytotoxicity variation that did not follow a linear pattern (Figure 3A–F).

3.4. Evaluation of the Effect of Major Compounds on the Invasion of NHDF Cells by T. gondii Tachyzoites of the RH Strain

To verify whether single major compounds showed influence on the invasion of NHDF cells by T. gondii, 106 tachyzoites were pre-treated under the same conditions as in Section 2.4 above. Invasion assays were also performed to evaluate the behavior of T. gondii after the pre-treatment with the major compounds tested here, to verify if they could harm this important stage of the parasite’s cycle. Treatment with α-Pinene led to the lowest rates of invasion, with averages of 5.5% at the highest concentration tested. In contrast, dillapiole showed low effects on T. gondii invasion of NHDF cells (Figure 4).

4. Discussion

Studies focusing on natural products and their anti-T.gondii effects represent alternatives for the treatment of toxoplasmosis. Herein, we describe the in vitro activity of the EOs from three species of genus Piper and their main constituents: α-Pinene, β-Pinene, and Dillapiole against tachyzoites of T. gondii.
In general, the ability of species of the genus Piper to show activity against parasites of medical importance is well known. However, some works need to be highlighted. A classic example is the EO from Piper aduncum that showed activity against Plasmodium falciparum, Trypanosoma cruzi, Trypanosoma brucei, and Leishmania infantum [23].
Several studies have demonstrated the antiparasitic effects of the genus Piper on medically important protozoa, but on Toxoplasma gondii there are only reports of the use of ethanolextracts from P. betle, P. nigrum and P. sarmentosum [18]. Our study revealed that EOs of Piper species and some of their constituents were active against T. gondii.
Analysis of the chemical composition of the EOs of three Piper species (Table 1) and comparisons to the anti-T. gondii activities suggest that, in the case of the P. lindbergii EO, the monoterpene α-Pinene could account for this potential. That is evidenced by the expressive amount of α-Pinene found in P. lindbergii (61.67%), compatible with their lowest EC50 values (Table 2). On the other hand, although P. aduncum contained 81.01% of Dillapiole in its composition, it had the second lowest EC50 among the three Piper species tested. Interestingly, isolated Dillapiole had the lowest EC50 suggesting other minor compounds in P. aduncum EO could account for the high activities against this parasite. Finally, although P. cernuum EO has both β-Pinene and α-Pinene (which showed anti- T. gondii activity), it performed worse than P. aduncum EO against T. gondii. That also suggests that some other compound in the P. cernuum may have a negative effect on the activity of this oil against T. gondii. EOs are complex mixtures of monoterpenes, sesquiterpenes, and phenylpropanoids, and the main components can act synergistically or antagonistically with minor constituents to modulate each other’s activity; consequently, we found different results than would be expected for single compounds [24].
Toxoplasma gondii is an obligate intracellular protozoan, and the host cell invasion process is crucial for parasite viability and infection establishment [25,26]. Thus, we sought to understand whether the pre-treatment of T. gondii tachyzoites with major compounds would be able to interfere with this essential process for the success of the parasitic infection. The pre-treatment of tachyzoites for 2 h with the major compounds of Piper EOs showed a decrease in the percentage of invading parasites, especially for β-Pinene and α-Pinene treatments. Similarly, a study using oleoresins from Copaifera reticulata, C. duckei, and C. pubiflora on T. gondii tachyzoites for one hour (with 64 and 32 μg/mL) significantly reduced parasite invasion compared to the control group [27].
In studies involving the search for new potential drugs for the treatment of toxoplasmosis, it is important to investigate their cytotoxicity given that the parasite is an obligatory intracellular organism. Moreover, in vitro toxicity tests are necessary for the initial phase of the discovery of promising drugs, as they constitute an essential tool to exclude compounds with cytotoxic properties [28,29]. The higher the value of the SI selectivity index obtained, the less toxic to the host and more selective to the parasite the compound is [30]. The selectivity indexes found in this work were highly satisfactory (99 for P. lindbergii and 126 for α-Pinene) since the selectivity index between parasites and host cells must reach at least 20 [31,32]. The selectivity potential of the EO of the genus Piper is already well known since previous works have demonstrated such action, either with the EO of P. aduncum with a high index of selectivity for trypomastigotes of Trypanosoma cruzi [12] or even against the promastigotes of Leishmania major, L. mexicana, L. braziliensis and L. donovani with a favorable selectivity index against peritoneal macrophages from BALB/c mice [33].
The present work opens future perspectives to explore the Piper EOs in the search for new compounds against toxoplasmosis and highlights that complex mixtures need to be further studied to understand the effects and interactions between their constituents on biological activity. In addition, it highlights α-Pinene as a promising compound for in vivo testing and against T. gondii bradyzoites.

5. Conclusions

The EO from P. lindbergii and its major monoterpene α-Pinene showed excellent anti-T. gondii activity, displaying a negative influence on the invasion of the parasite in the studied model, with a good selectivity index. These findings support future studies with these compounds using in vivo models of activity against T. gondii to search for new compounds and targets for the development of alternatives for the treatment of toxoplasmosis.

Author Contributions

A.A.P.F., É.S.M.-D. designed and carried out the research. A.A.P.F., M.J.K., M.M.C., L.F.Y. and M.A.S. interpreted the data and contributed to writing the manuscript. A.A.P.F., M.M.C. and É.S.M.-D. contributed to methodology and investigation. M.J.K. and É.S.M.-D. supervised the work. All authors have read and agreed to the published version of the manuscript.

Funding

Support for this work was provided by the Brazilian agencies: Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP) (2014/50316-7), PRPq UFMG 09/2019 and Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data areavailable in the manuscript.

Acknowledgments

The authors would like to thank Rosalida Estevan Nazar Lopes for their valuable technical assistance.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Daher, D.; Shaghlil, A.; Sobh, E.; Hamie, M.; Hassan, M.E.; Moumneh, M.B.; Itani, S.; El Hajj, R.; Tawk, L.; El Sabban, M.; et al. Comprehensive Overview of Toxoplasma Gondii-Induced and Associated Diseases. Pathogens 2021, 10, 1351. [Google Scholar] [CrossRef]
  2. Dunay, I.R.; Gajurel, K.; Dhakal, R.; Liesenfeld, O.; Montoya, J.G. Treatment of Toxoplasmosis: Historical Perspective, Animal Models, and Current Clinical Practice. Clin. Microbiol. Rev. 2018, 31, e00057-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Aguirre, A.A.; Longcore, T.; Barbieri, M.; Dabritz, H.; Hill, D.; Klein, P.N.; Lepczyk, C.; Lilly, E.L.; McLeod, R.; Milcarsky, J.; et al. The One Health Approach to Toxoplasmosis: Epidemiology, Control, and Prevention Strategies. EcoHealth 2019, 16, 378–390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Robert-Gangneux, F.; Dardé, M.-L. Epidemiology of and Diagnostic Strategies for Toxoplasmosis. Clin. Microbiol. Rev. 2012, 25, 264–296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Wei, H.-X.; Wei, S.-S.; Lindsay, D.S.; Peng, H.-J. A Systematic Review and Meta-Analysis of the Efficacy of Anti-Toxoplasma Gondii Medicines in Humans. PLoS ONE 2015, 10, e0138204. [Google Scholar] [CrossRef]
  6. Ben-Harari, R.R.; Goodwin, E.; Casoy, J. Adverse Event Profile of Pyrimethamine-Based Therapy in Toxoplasmosis: A Systematic Review. Drugs RD 2017, 17, 523–544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Teil, J.; Dupont, D.; Charpiat, B.; Corvaisier, S.; Vial, T.; Leboucher, G.; Wallon, M.; Peyron, F. Treatment of Congenital Toxoplasmosis: Safety of the Sulfadoxine-Pyrimethamine Combination in Children Based on a Method of Causality Assessment. Pediatr. Infect. Dis. J. 2016, 35, 634–638. [Google Scholar] [CrossRef]
  8. Ovung, A.; Bhattacharyya, J. Sulfonamide Drugs: Structure, Antibacterial Property, Toxicity, and Biophysical Interactions. Biophys. Rev. 2021, 13, 259–272. [Google Scholar] [CrossRef]
  9. Silva, L.A.; Fernandes, M.D.; Machado, A.S.; Reis-Cunha, J.L.; Bartholomeu, D.C.; Almeida Vitor, R.W. Efficacy of Sulfadiazine and Pyrimetamine for Treatment of Experimental Toxoplasmosis with Strains Obtained from Human Cases of Congenital Disease in Brazil. Exp. Parasitol. 2019, 202, 7–14. [Google Scholar] [CrossRef]
  10. Oliveira, C.B.; Meurer, Y.S.; Andrade, J.M.; Costa, M.E.; Andrade, M.M.; Silva, L.A.; Lanza, D.C.; Vítor, R.W.; Andrade-Neto, V.F. Pathogenicity and Phenotypic Sulfadiazine Resistance of Toxoplasma Gondii Isolates Obtained from Livestock in Northeastern Brazil. Mem. Inst. Oswaldo Cruz 2016, 111, 391–398. [Google Scholar] [CrossRef]
  11. Montazeri, M.; Mehrzadi, S.; Sharif, M.; Sarvi, S.; Tanzifi, A.; Aghayan, S.A.; Daryani, A. Drug Resistance in Toxoplasma Gondii. Front. Microbiol. 2018, 9, 2587. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Villamizar, L.H.; Cardoso, M.D.G.; Andrade, J.D.; Teixeira, M.L.; Soares, M.J. Linalool, a Piper Aduncum Essential Oil Component, Has Selective Activity against Trypanosoma Cruzi Trypomastigote Forms at 4 °C. Mem. Inst. Oswaldo Cruz 2017, 112, 131–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Esperandim, V.; da Silva Ferreira, D.; Sousa Rezende, K.; Magalhães, L.; Medeiros Souza, J.; Pauletti, P.; Januário, A.; da Silva de Laurentz, R.; Bastos, J.; Símaro, G.; et al. In Vitro Antiparasitic Activity and Chemical Composition of the Essential Oil Obtained from the Fruits of Piper Cubeba. Planta Med. 2013, 79, 1653–1655. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Ceole, L.F.; Cardoso, M.D.G.; Soares, M.J. Nerolidol, the Main Constituent of Piper Aduncum Essential Oil, Has Anti- Leishmania Braziliensis Activity. Parasitology 2017, 144, 1179–1190. [Google Scholar] [CrossRef]
  15. Moura do Carmo, D.F.; Amaral, A.C.F.; Machado, G.M.C.; Leon, L.L.; Silva, J.R.D.A. Chemical and Biological Analyses of the Essential Oils and Main Constituents of Piper Species. Molecules 2012, 17, 1819–1829. [Google Scholar] [CrossRef] [PubMed]
  16. Houël, E.; Gonzalez, G.; Bessière, J.-M.; Odonne, G.; Eparvier, V.; Deharo, E.; Stien, D. Therapeutic Switching: From Antidermatophytic Essential Oils to New Leishmanicidal Products. Mem. Inst. Oswaldo Cruz 2015, 110, 106–113. [Google Scholar] [CrossRef] [Green Version]
  17. Varela, M.T.; Lima, M.L.; Galuppo, M.K.; Tempone, A.G.; de Oliveira, A.; Lago, J.H.G.; Fernandes, J.P.S. New Alkenyl Derivative from Piper Malacophyllum and Analogues: Antiparasitic Activity against Trypanosoma Cruzi and Leishmania Infantum. Chem. Biol. Drug Des. 2017, 90, 1007–1011. [Google Scholar] [CrossRef]
  18. Leesombun, A.; Boonmasawai, S.; Shimoda, N.; Nishikawa, Y. Effects of Extracts from Thai Piperaceae Plants against Infection with Toxoplasma Gondii. PLoS ONE 2016, 11, e0156116. [Google Scholar] [CrossRef] [Green Version]
  19. Aziz, Z.A.A.; Ahmad, A.; Setapar, S.H.M.; Karakucuk, A.; Azim, M.M.; Lokhat, D.; Rafatullah, M.; Ganash, M.; Kamal, M.A.; Ashraf, G.M. Essential Oils: Extraction Techniques, Pharmaceutical And Therapeutic Potential—A Review. Curr. Drug Metab. 2018, 19, 1100–1110. [Google Scholar] [CrossRef]
  20. Fanela, T.L.M.; Baldin, E.L.L.; Pannuti, L.E.R.; Cruz, P.L.; Crotti, A.E.M.; Takeara, R.; Kato, M.J. Lethal and Inhibitory Activities of Plant-Derived Essential Oils against Bemisia Tabaci Gennadius (Hemiptera: Aleyrodidae) Biotype B in Tomato. Neotrop. Entomol. 2016, 45, 201–210. [Google Scholar] [CrossRef]
  21. Pereira Filho, A.A.; Pessoa, G.C.D.; Yamaguchi, L.F.; Stanton, M.A.; Serravite, A.M.; Pereira, R.H.M.; Neves, W.S.; Kato, M.J. Larvicidal Activity of Essential Oils From Piper Species against Strains of Aedes Aegypti (Diptera: Culicidae) Resistant to Pyrethroids. Front. Plant Sci. 2021, 12, 685864. [Google Scholar] [CrossRef] [PubMed]
  22. Martins-Duarte, E.S.; Portes, J.D.A.; da Silva, R.B.; Pires, H.S.; Garden, S.J.; de Souza, W. In Vitro Activity of N-Phenyl-1,10-Phenanthroline-2-Amines against Tachyzoites and Bradyzoites of Toxoplasma Gondii. Bioorg. Med. Chem. 2021, 50, 116467. [Google Scholar] [CrossRef] [PubMed]
  23. Gutiérrez, Y.; Montes, R.; Scull, R.; Sánchez, A.; Cos, P.; Monzote, L.; Setzer, W.N. Chemodiversity Associated with Cytotoxicity and Antimicrobial Activity of Piper Aduncum Var. Ossanum. Chem. Biodivers. 2016, 13, 1715–1719. [Google Scholar] [CrossRef]
  24. Nunes, T.A.L.; Costa, L.H.; De Sousa, J.M.S.; De Souza, V.M.R.; Rodrigues, R.R.L.; Val, M.D.C.A.; da Cunha Pereira, A.C.T.; Ferreira, G.P.; Da Silva, M.V.; Da Costa, J.M.A.R.; et al. Eugenia Piauhiensis Vellaff. Essential Oil and γ-Elemene Its Major Constituent Exhibit Antileishmanial Activity, Promoting Cell Membrane Damage and in Vitro Immunomodulation. Chem. Biol. Interact. 2021, 339, 109429. [Google Scholar] [CrossRef]
  25. Mammari, N.; Halabi, M.A.; Yaacoub, S.; Chlala, H.; Dardé, M.-L.; Courtioux, B. Toxoplasma Gondii Modulates the Host Cell Responses: An Overview of Apoptosis Pathways. BioMed Res. Int. 2019, 2019, 6152489. [Google Scholar] [CrossRef] [Green Version]
  26. Sanchez, S.G.; Besteiro, S. The Pathogenicity and Virulence of Toxoplasma Gondii. Virulence 2021, 12, 3095–3114. [Google Scholar] [CrossRef] [PubMed]
  27. Teixeira, S.C.; de Souza, G.; Borges, B.C.; de Araújo, T.E.; Rosini, A.M.; Aguila, F.A.; Ambrósio, S.R.; Veneziani, R.C.S.; Bastos, J.K.; Silva, M.J.B.; et al. Copaifera Spp. Oleoresins Impair Toxoplasma Gondii Infection in Both Human Trophoblastic Cells and Human Placental Explants. Sci. Rep. 2020, 10, 15158. [Google Scholar] [CrossRef]
  28. Indrayanto, G.; Putra, G.S.; Suhud, F. Validation of In-Vitro Bioassay Methods: Application in Herbal Drug Research. In Profiles of Drug Substances, Excipients and Related Methodology; Elsevier: Cambridge, MA, USA; Academic Press: Cambridge, MA, USA, 2021; Volume 46, pp. 273–307. ISBN 978-0-12-824127-1. [Google Scholar]
  29. Atanasov, A.G.; Zotchev, S.B.; Dirsch, V.M.; Supuran, C.T. Natural Products in Drug Discovery: Advances and Opportunities. Nat. Rev. Drug Discov. 2021, 20, 200–216. [Google Scholar] [CrossRef]
  30. Krivogorsky, B.; Grundt, P.; Yolken, R.; Jones-Brando, L. Inhibition of Toxoplasma Gondii by Indirubin and Tryptanthrin Analogs. Antimicrob. Agents Chemother. 2008, 52, 4466–4469. [Google Scholar] [CrossRef] [Green Version]
  31. Evans, S.M.; Casartelli, A.; Herreros, E.; Minnick, D.T.; Day, C.; George, E.; Westmoreland, C. Development of a High Throughput in Vitro Toxicity Screen Predictive of High Acute in Vivo Toxic Potential. Toxicol. In Vitro 2001, 15, 579–584. [Google Scholar] [CrossRef]
  32. Nwaka, S.; Hudson, A. Innovative Lead Discovery Strategies for Tropical Diseases. Nat. Rev. Drug Discov. 2006, 5, 941–955. [Google Scholar] [CrossRef] [PubMed]
  33. Monzote, L.; García, M.; Montalvo, A.M.; Scull, R.; Miranda, M. Chemistry, Cytotoxicity and Antileishmanial Activity of the Essential Oil from Piper Auritum. Mem. Inst. Oswaldo Cruz 2010, 105, 168–173. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Overview of the assay used to evaluate the in vitro antiproliferative effect of Piper sp. essential oils and dillapiole, α-Pinene, and β-Pinene on Toxoplasma gondii tachyzoites of the RH lineage. The chemical structures of dillapiole, α-Pinene, and β-Pinene were obtained from the website ChemSpider.
Figure 1. Overview of the assay used to evaluate the in vitro antiproliferative effect of Piper sp. essential oils and dillapiole, α-Pinene, and β-Pinene on Toxoplasma gondii tachyzoites of the RH lineage. The chemical structures of dillapiole, α-Pinene, and β-Pinene were obtained from the website ChemSpider.
Metabolites 13 00095 g001
Figure 2. Antiproliferative effects of essential oils from three Piper species (AC) and dillapiole, β-Pinene and α-Pinene (DF) on tachyzoites of the T. gondii RH strain 7 days after treatment. Kruskal–Wallisfollowed by Dunn’s posthoc test was performed (** p < 0.001, and *** p < 0.0001 among the compared groups).
Figure 2. Antiproliferative effects of essential oils from three Piper species (AC) and dillapiole, β-Pinene and α-Pinene (DF) on tachyzoites of the T. gondii RH strain 7 days after treatment. Kruskal–Wallisfollowed by Dunn’s posthoc test was performed (** p < 0.001, and *** p < 0.0001 among the compared groups).
Metabolites 13 00095 g002
Figure 3. Cytotoxic effects of Piper sp. essential oils (AC) and dillapiole, β-Pinene, and α-Pinene (DF) on NHDF cells. Viability was compared by Kruskal–Wallis, followed by Dunn’s posthoc test (*** p < 0.0001 among the compared groups).
Figure 3. Cytotoxic effects of Piper sp. essential oils (AC) and dillapiole, β-Pinene, and α-Pinene (DF) on NHDF cells. Viability was compared by Kruskal–Wallis, followed by Dunn’s posthoc test (*** p < 0.0001 among the compared groups).
Metabolites 13 00095 g003
Figure 4. Effect of pretreatment with Dillapiole, β-Pinene, and α-Pinene (AC) on T. gondii tachyzoites during NHDF cell invasion. The number of infected cells in each group was compared to controls by Kruskal–Wallis, followed by Dunn’s posthoc tests (* p < 0.01, and ** p < 0.001 among the compared groups).
Figure 4. Effect of pretreatment with Dillapiole, β-Pinene, and α-Pinene (AC) on T. gondii tachyzoites during NHDF cell invasion. The number of infected cells in each group was compared to controls by Kruskal–Wallis, followed by Dunn’s posthoc tests (* p < 0.01, and ** p < 0.001 among the compared groups).
Metabolites 13 00095 g004
Table 1. Chemical composition of the essential oils of three Piper species.
Table 1. Chemical composition of the essential oils of three Piper species.
CompoundsRIaRIbPADPLDPCE
α-Pinene S9329320.261.716.6
Camphene S947946-1.70.1
β-Pinene S9759740.31.411.5
β-Myrcene S992988-0.31.0
α-Phellandrene S100410020.1-0.2
2-Carene S101010080.1-0.2
α-Terpinene10161014--4.5
p-Cymene S102410200.11.09.2
Limonene S103910240.15.30.8
(Z)-β-Ocimene S103910321.6-0.1
(E)-β-Ocimene S104910443.4-0.3
γ-Terpinene105910540.2-9.9
α-Terpinolene S108810860.4-2.7
Linalool S11001095-1.6-
(E)-4,8-Dimethyl-1,3,7-nonatriene (DMNT) S11171114--0.3
Camphor S11441141-1.1-
Terpinen-4-ol11781174-0.10.3
α-Terpineole11911186-2.30.3
Oxygenated monoterpene *1209-0.10.1-
(+)-Piperitone125512490.7--
δ-Elemene133913350.11.10.4
α-Cubebene13521345-0.10.3
α-Ylangene137413730.1--
α-Copaene S137813740.26.42.1
β-Bourbonene13871387-0.20.6
β-Elemene139413890.20.34.4
α-Gurjunene S141214090.1-0.1
(E)-β-Caryophyllene S142214170.80.57.0
β-Gurjunene143214310.20.60.5
(+)-Aromadendrene S14421439-0.20.4
α-Humulene S145714520.90.22.1
(-)-Alloaromadendrene S14641458-0.90.1
Dehydro-aromadendrane14661460-0.9-
γ-Muurolene14801479-1.30.5
Germacrene D148414812.7-5.2
β-Selinene14901490-0.30.5
α-Selinene150014981.40.1-
Bicyclogermacrene150015002.3-10.7
α-Muurolene150315000.11.0-
α-Bulnesene151015090.2-0.7
γ-Cadinene151715130.11.20.4
δ-Cadinene152215221.2--
(E)-Cadina-1.4-diene15271533-1.30.3
Germacrene B156115590.2-0.1
(E)-Nerolidol S156615610.1-0.9
Palustrol15721567--0.1
Spathulenol158115770.10.30.7
(-)-Caryophyllene oxide S15871582-3.40.8
Veridiflorol159615920.30.50.6
β-Asarone16231616-0.3-
Dillapiole S1632162081.0-0.1
epi-α-Muurolol164616400.30.60.6
Torreyol16501644-0.30.4
α-Cadinol16591652-0.91.2
Apiole S168616770.2--
RIa—Retention Index calculated against RIa. RIa: Retention index calculated against C8–C40 n-alkanes using an HP-5 ms column. RIb: Retention index values from the literature (Adams, 2007). PAD: Piper aduncum; PLD: Piper lindbergii; PCE: Piper cernuum. S Compound identity confirmed with an authentic standard. The remaining compounds were identified by comparing the RI and mass spectra with the Adams and Wiley databases (see text for details). * Unidentified compound.
Table 2. Evaluation of effective concentrations (EC50) of Piper sp. Essential oils (EOs) and major compounds on tachyzoites of the T. gondii RH strain.
Table 2. Evaluation of effective concentrations (EC50) of Piper sp. Essential oils (EOs) and major compounds on tachyzoites of the T. gondii RH strain.
EO/CompoundEC50 (95% Confidence Intervals) µg/mLR
Piper aduncum1.749 (1.498 to 2.043)0.9807
P. cernuum3.687 (2.190 to 6.208)0.7144
P. lindbergii0.839 (0.6492 to 1.084)0.9358
Dillapiole4.287 (3.511 to 5.234)0.9331
β-Pinene1.145 (1.008 to 1.300)0.9836
α-Pinene0.326 (0.295 to 0.360)0.9805
Table 3. Evaluation of cytotoxicity concentrations (CC50) of Piper sp. Essential oils (EOs) and major compounds in NHDF cells. SI = selectivity index.
Table 3. Evaluation of cytotoxicity concentrations (CC50) of Piper sp. Essential oils (EOs) and major compounds in NHDF cells. SI = selectivity index.
EO/CompoundCC50 (95% Confidence Intervals) µg/mLRSI
Piper aduncum169.8 (135.6 to 212.8)0.807497
Piper cernuum172.1 (125.6 to 235.7)0.682646
Piper lindbergii83.80 (75.42 to 91.34)0.999899
Dillapiole210.8 (194.6 to 228.3)0.990649
β-Pinene70.78 (53.22 to 94.12)0.968261
α-Pinene41.37 (37.64 to 45.09)0.9991126
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Pereira Filho, A.A.; Cunha, M.M.; Alves Stanton, M.; Fumiko Yamaguchi, L.; Jorge Kato, M.; Martins-Duarte, É.S. In Vitro Activity of Essential Oils from Piper Species (Piperaceae) against Tachyzoites of Toxoplasma gondii. Metabolites 2023, 13, 95. https://0-doi-org.brum.beds.ac.uk/10.3390/metabo13010095

AMA Style

Pereira Filho AA, Cunha MM, Alves Stanton M, Fumiko Yamaguchi L, Jorge Kato M, Martins-Duarte ÉS. In Vitro Activity of Essential Oils from Piper Species (Piperaceae) against Tachyzoites of Toxoplasma gondii. Metabolites. 2023; 13(1):95. https://0-doi-org.brum.beds.ac.uk/10.3390/metabo13010095

Chicago/Turabian Style

Pereira Filho, Adalberto Alves, Mariana Maciel Cunha, Mariana Alves Stanton, Lydia Fumiko Yamaguchi, Massuo Jorge Kato, and Érica S. Martins-Duarte. 2023. "In Vitro Activity of Essential Oils from Piper Species (Piperaceae) against Tachyzoites of Toxoplasma gondii" Metabolites 13, no. 1: 95. https://0-doi-org.brum.beds.ac.uk/10.3390/metabo13010095

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

Article Metrics

Back to TopTop