Next Article in Journal
Research Progress on Plant Shaker K+ Channels
Previous Article in Journal
A SEPALLATA MADS-Box Transcription Factor, SlMBP21, Functions as a Negative Regulator of Flower Number and Fruit Yields in Tomato
Previous Article in Special Issue
Corchorus tridens L.: A Review of Its Botany, Phytochemistry, Nutritional Content and Pharmacological Properties
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Exploring Southern Ecuador’s Traditional Medicine: Biological Screening of Plant Extracts and Metabolites

1
Institute for Regenerative Medicine and Biotherapy (IRMB), Université de Montpellier National Institute of Health and Medical Research (INSERM), 34295 Montpellier, France
2
Nephrology Department CHRU Montpellier, Institute for Regenerative Medicine and Biotherapy (IRMB), Université de Montpellier, 34295 Montpellier, France
3
Departamento de Química, Universidad Técnica Particular de Loja, San Cayetano Alto, s/n. AP: 11 01 608, Loja 1101608, Ecuador
*
Author to whom correspondence should be addressed.
Plants 2024, 13(10), 1422; https://doi.org/10.3390/plants13101422
Submission received: 8 March 2024 / Revised: 8 May 2024 / Accepted: 10 May 2024 / Published: 20 May 2024
(This article belongs to the Special Issue Plant Extracts)

Abstract

:
Ecuador stands as a nation inheriting a profound ancestral legacy in the utilization of medicinal plants, reflective of the rich biodiversity embraced by various ethnic groups. Despite this heritage, many of these therapeutic resources remain insufficiently explored concerning their toxicity and potential pharmacological effects. This study focused on a comprehensive evaluation of cytotoxicity and the potential subcellular targets within various extracts and nine isolated metabolites from carefully selected medicinal plants. Assessing their impact on the breast cancer cell line (MCF7), we subsequently examined the most active fractions for effects on the cell cycle, microtubule network, centrosome duplication, γH2AX foci, and E-cadherin. The investigated crude extracts and isolated compounds from Ecuadorian medicinal plants demonstrated cytotoxic effects, influencing diverse cellular pathways. These findings lend credence to the traditional uses of Ecuadorian medicinal plants, which have served diverse therapeutic purposes. Moreover, they beckon the exploration of the specific chemicals, whether in isolation or combination, responsible for these observed activities.

1. Introduction

According to the World Health Organization (WHO), more than 80% of the world’s population uses some form of traditional and complementary medicine at the primary level. Traditional and complementary medicine is defined by the WHO as comprising health practices, approaches, knowledge, and beliefs that incorporate plant-based medicines, spiritual therapies, manual techniques, and exercises. These are based on cultural traditions of healing that have been handed down from generation to generation and influenced by factors such as history, personal attitudes, and philosophy. Both traditional Chinese medicine as well as Ayurvedic practices maintained the knowledge and preserved the practice necessary for their survival [1].
For millions of people, herbal treatments, traditional remedies, and traditional medical practices represent the main, and sometimes only, source of health care. They are also culturally accepted. In “Western” civilization, the practice of traditional medicine was gradually lost and considered contrary to a rationalist application of patient care.
We can consider that the first obstacle to this dogma was brought by the description [2] then of the power [3] of the placebo effect and its necessary evaluation in the approval of any new drug.
The World Health Organization (WHO) now has a Traditional Medicine Strategy that supports the integration of these practices in national health systems [4]. In 2018, 98 countries, mostly from the African, South-east, South American, and Pacific regions but also including Canada, Germany, Switzerland, the UK, and Norway, had a national policy level for traditional medicine [4].
Natural products derived from plants play a role in the area of chemical medicine by providing a reservoir of secondary metabolites that have pharmacological actions. In the southern Andes of Ecuador, which has particularly rich biodiversity, a large variety of medicinal plants have been found to have biological activity as antioxidants and have anti-inflammatory, anti-parasitic, and many other pharmacological characteristics [5,6,7,8,9].
In the area of cancer, there is an urgent need for novel therapeutic agents to minimize the harmful side effects of cytotoxic chemotherapies, among which is the emergence of antibiotic-resistant bacteria that develop due to the weakened immune systems that render patients more sensitive to infections. In response to this pressing issue, a multifaceted approach is being pursued, encompassing diverse forms of therapy. This strategy involves a combination of therapies such as chemotherapy, immunotherapy with monoclonal antibodies [10,11] and immune system “boosters”, and radiation or complementary therapies. The synergy between these drugs is more potent than the efficacy of each one in isolation. These observations open the reflection on the utilization of complementary therapies based on the ancient recipes and traditional knowledge [12,13,14,15,16] compatible with approved therapeutic protocols.
Based on this assumption, we took advantage of the extensive botanical biodiversity coupled with the traditional medicinal knowledge to screen various plant extracts for their cytotoxic potential.
For decades, products isolated from the plant kingdom such as taxol, vinca alkaloids, etoposides, and others have been established as reference molecules due to their notable anti-cancer efficacy [17]. Moreover, numerous studies have demonstrated the anti-tumoral potential of essentials oils [18] or various plant extracts [19,20].
The observed cytotoxicity of widely therapeutically used plant-derived drugs may affect diverse molecular pathways [21]. Cytotoxic compounds target, directly, the DNA of the cells and act as inhibitors of topoisomerase I (Camptothecin) or topoisomerase II [22]. The dynamics of microtubules are disrupted by molecules binding to tubulin, thus blocking the cell cycle [23]. Natural HDAC inhibitors that reactivate epigenetically silenced genes in cancer cells are promising candidates [19]. It is now admitted that the combination of drugs or multi-acting drugs for the simultaneous targeting of various pathways will be an option for new treatments [24]. These observations motivated us to investigate the impact of raw and complex extracts from plants used in traditional medicine on cell behavior, aiming to assess their potential in complementary treatments.
In this study, we focused on evaluating the potential targets of the selected plant extracts. To achieve this, we opted to investigate their influence on (1) the equilibrium of cell cycle phases, (2) microtubule network organization, (3) centrosome duplication, (4) DNA damage, and (5) E-Cadherin architecture. The selection of plants and their specific parts was based on comprehensive ethnobotanical, ethnopharmacological, and ethno-medical records developed in previous years [6,7,25,26,27].

2. Results

2.1. Screening the Extracts

Plants were carefully chosen and collected based on local traditional medicinal practices and knowledge. When the traditional uses were clearly indicated, a brief description of these uses is provided in Table 1. A total of 71 plant extracts from two sampling campaigns involving 56 plant species were evaluated for their toxicity against the MCF7 breast cancer cell line. Concentrations of 20, 50, and 100 µg mL−1 for each extract, dissolved in DMSO, were selected arbitrarily for the initial screening. For the sake of clarity, only concentrations of 20 and 100 µg mL−1 are presented in the tables.
The outcomes of this evaluation are documented in the aforementioned tables. The selection of extracts for further experimentation was based on several factors. The initial criterion was the extracts’ ability to reduce the survival rate of MCF7 cell lines to less than 40% at a concentration of 50 µg/mL. An exception was made for the hydro-alcoholic extract from Piper ecuadorense Sodiro because of the wide use of this plant by local communities. Additionally, the solubility of the extracts in DMSO was taken into account. Sixteen extracts (numbered from one to sixteen) were then chosen and listed in Table 2.

2.2. Determination of the Cytotoxicity of the Selected Extracts

The survival of the MCF7 cell line was assessed following a 72 h exposure to concentrations ranging from 6.25 to 100 µg/mL of each extract. The IC50 was determined from this dataset as the concentration that prompts a 50% survival rate (see Section 4). These findings are outlined in Table 2. Notably, a majority of these plants boast recognized medicinal properties, which will be expounded upon below.

2.3. Description of Recognized Traditional Therapeutic Uses

Cestrum sendtnerianum C. Mart. (flowers) is used as a purgative and to treat head pain, stomach pain, fever, gangrene, influenza, internal infections, rheumatism, cough [4], postpartum relapse [25], and cold and to relieve inflammation in children after excessive exposure to sun [28].
Croton lechleri Müll. Arg. (latex) is used to treat hepatic pain and dermatitis; serves as a disinfectant; plays roles in the healing of wounds and diuretics [6]; acts against diarrhea, insect bites [5], gastritis, inflammation of the intestines, skin blemishes, and pimples; serves as an anti-parasitic and antiseptic; and is used to treat ulcers, throat infections, and gingivitis [29].
Gaiadendron punctatum (Ruiz & Pav.) G. Don (leaves) is used to treat bronchitis, hepatic pain, influenza, and cough; serves as a hair tonic [6]; fights strong cough [25]; and is also used as a hair tonic. It is used in case of measles and smallpox or for help in insomnia and to decrease disease relapses (recaída in Spanish) after childbirth [28].
Garcinia macrophylla Mart. (leaves and bark) is used for the relief of pains in the body and to treat external inflammation [25].
Huperzia columnaris B. Øllg. (aerial part): The fresh whole plant is used to treat liver and kidney diseases, fever, inflammation, and colds [30]. According to the main cultural tradition of the Saraguro people, this plant is mainly used as an intestinal purgative, especially to cure various supernatural diseases such as vaho de agua (exposure to water-vapors), espanto, and susto (startlement and fright) [31].
Huperzia kuesteri (Nessel) B. Øllg. (aerial part): The fresh whole plant treats liver and kidney diseases, fever, inflammations and colds [30]. According to Saraguro, people could take baths to relieve pain in the waist and backache, treat colds, give baths after childbirth, and use the plant as a purgative and to cure various supernatural diseases.
Hernandulcin: this [6-(1′-hydroxy-1′, 5 dimethyl-4′-hexenyl)-3-methyl-2-cyclohexenone] isolated product is 1000 times sweeter than sugar with any toxicity [33,34].
Myrcianthes fragrans (Sw.) McVaugh (leaves): It is used as an infusion to treat respiratory problems, the inflammation of the throat and gums, tonsillitis, and stomatitis, and is used to treat vaginal infections [35]. Besides medicinal uses, the leaves have also been employed as natural aromatic ingredients in the traditional Ecuadorian drink colada morada, which is drunk on the Day of the Dead (on 2 November each year).
Phyla strigulosa (M. Martens & Galeotti) Moldenke (leaves and flowers): This plant is used to treat stomachache [29], cramps, diarrhea in children, and intestinal infections and serves as a tonic.
Pinostrobin [(2S)-5-hydroxy-7-methoxy-2-phenyl-2,3-dihydrochromen-4-one]:
This substance was proposed as being nontoxic for the MCF7 cell line (IC50> 50 µM) but proposed as a topoisomerase 1 inhibitor highlighting the therapeutic potential of pinostrobin as an anti-proliferative agent [36].
Piper ecuadorense Sodiro (leaves): This plant helps fight hangover, acts as a disinfectant, and helps in the healing of wounds [6]. This plant is used in mythological cases as mal aire (bad air) [25].
Piper pseudochurumayu (Kunth) C. DC. (leaves): This plant is used to provide analgesic, diuretic, digestive, dermatological, anthelmintic, antirheumatic, and antidiarrheal effects and treat respiratory infections [9].
Stereocaulon ramulosum (Sw.) Raeusch. (aerial part) is used to treat external infections as its antibiotic activity has been reported [32].

2.4. Identification of Potential Target for the Selected Extracts

2.4.1. Cell Cycle

We first studied the effect of selected extracts on the equilibrium of cell cycle phases. In Figure 1 and the Table 3 are presented the significance of the cell cycle phases and their ratio including the SubG1 upon incubation with 25 µg/mL of each extract.

2.4.2. Microtubule

The microtubule network is part of the cellular cytoskeleton involved in the maintenance of the cell shape, mobility, and intracellular trafficking [37], and it is also the main component of the mitotic spindle. Any disturbance in the microtubule dynamics leads to cell cycle blockage and cell death [38]. Figure 2’s lane A illustrates a selected example of an observed effect on the microtubule network after the treatment using the extracts (here, in 13 and 3). The effects of the 16 extracts and DMSO vehicle are presented in Supplementary Figure S1.

2.4.3. Centrosomes

Centrosomes are the microtubule-organizing centers (MTOCs) of eukaryotic cells [39]. The progress of mitosis depends on their duplication and their migration on either side of the nucleus during the G2 phase of the cell cycle.
Centrosome amplification or abnormality in their duplication lead to numerous diseases including cancer [40]. Upon the addition of the selected extracts, we evaluated the centrosomal integrity as follows. Typical features observed after incubation with compounds 1 or 4 are represented in Figure 2, lane B. The effects of the 16 extracts and DMSO vehicle are presented in Supplementary Figure S2.

2.4.4. DNA Damage Analysis

Maintaining DNA integrity is vital for cellular survival. However, DNA damage can occur depending on the cellular environment. Common types of damage include single- or double-strand breaks induced by irradiation, reactive oxygen species, or chemical agents. Additionally, cross-linking between strands and base damage caused by chemical modifications or adduct formation are frequent occurrences. In healthy cells, DNA repair mechanisms can often overcome such damage and restore DNA integrity [41].
DNA damage due to double-strand breakage is associated with the formation of γ-H2AX foci at the site of a break. This is one of the markers of the genotoxicity induced by a treatment or the environment of the cell. Genetic instability is one of the important factors that could drive cells to a tumoral phenotype [42]. Figure 2, lane C illustrates the detection of γ–H2AX foci observed in the cell nucleus after the treatment of the cell culture with extract 4 or 10. The effects of the 16 extracts and DMSO vehicle are presented in Supplementary Figure S3.

2.4.5. E-Cadherin Test

E-Cadherin is a transmembrane protein regulating epithelial cell–cell adhesions that drives cellular proliferation and tissue morphogenesis. Thus, its expression is implicated in tumor progression and metastasis [43]. Its interaction with the F-actin network through the cadherin–catenin complex contributes also to dynamic cell movements in response to physical changes in the cells’ environment. When observed, the effect of the addition of plant extract to the culture medium was presented (Figure 2, lane D). The effects of the 16 extracts and DMSO vehicle are presented in Figure S4.

3. Discussion

All plants included in this study were chosen due to their utilization in the traditional medicine practices of southern Ecuador. The traditional uses of the plants from which extracts were prepared are described in the results section. Furthermore, certain extracts selected for the second phase of this study have been previously investigated for their pharmacological or medicinal properties, albeit without specifying their targets. For instance, ethanol extract from Cestrum sendtenerianum (1) was found to contain steroidal saponins exhibiting modest cytotoxic activity [44]. Croton lechleri (2), commonly known as Dragon’s blood, has been extensively studied, revealing diverse pharmacological benefits [5] including anti-tumoral effects [45], the management of diarrhea associated with AIDS or cancer treatments [16,46], and dermatological disease [47]. Cytotoxic compounds were extracted from Garcinia macrophylla (4) with potential anti tumoral properties [48]. An Acetyl cholinesterase inhibitory potential was demonstrated for preparations from Huperzia columnaris [31]. The essential oil of Myrcianthes fragrans (8) exhibited antimicrobial activity [34] and showed cytotoxic activity against the Hep G2 cell line [49]. Both the antifungal activity of raw extracts from Piper ecuadorense (14) [50,51] as well as larvicidal and antimalarial [52] activities of essential oil or extracts of Piper pseudochurumayu (15) have been demonstrated. Lastly, antimicrobial activity was found in an extract derived from Stereocaulon sp. (16) Ecuadorian lichen [53].
Following an extensive screening of extracts from plants traditionally employed in folk medicine and a subsequent selection of the most potent compounds, this study was conducted to enhance our understanding of some of these extracts by investigating their impact on potential cellular targets.
Table 3 illustrates the cell-cycle phase proportion after the culturing of MCF7 cells in the presence of 25 µg/mL of selected extracts. A well-described moderate increase in the G0/G1 phase due to DMSO [54] was observed. A 10% increase in the G0/G1 phase with a decrease in the S1 and G2/M phases was induced by compounds 4 and 5, reflecting a possible blockage in the S phase entry. A strong increase in the proportion of cells in a Sub G1 phase, which are often referred to as apoptotic cells, is induced after incubation with the dilution of compounds 8, 12, 13, and 15 without direct links with their overall toxicity (IC50) (Table 2). Then, incubation with 25 µg/mL of plant extract was investigated for its effect on cellular substructures, namely the microtubular network, centrosomes, DNA double-strand break, and E-cadherin assembly.
Among the extracts tested, extracts 3 (Figure 2, lane A, center) and 10 induced a disappearance of polymerized tubulin assemblies, but surprisingly, this effect was associated with strong toxicity only for extract 10. Treatments with extracts 6, 7, 8, 9, 12, 13 (Figure 2, lane A, right) and 16 resulted in a mis-organization of the microtubule with shorter filaments and a less dense network. Other extracts exerted no or slight effects on the microtubule network.
Concerning centrosomes that are the center for cellular microtubule organization, their behavior was investigated under the pression of the extracts (Figure 2, lane B). The average number of centrosomes visualized per cell calculated as described varied from 0.22 after treatment with extract 5 to 0.7 in the untreated condition. A significant decrease in the number of centrioles resulted from treatments with extracts 4 (Figure 2, lane B, right), 5, 6, and 14, but this phenomenon was without correlation with toxicity or cell-cycle perturbation.
We evaluated the DNA breaks induced by the presence of the extracts in the cell culture medium through the visualization of the γ-H2AX foci into the cell nucleus. In Figure 2C, we evidence that the number of foci per cell nucleus was clearly increased after treatment with extracts 2, 4 (Figure 2, lane C, center), 8, and 14 and strongly increased after treatment with extract 10 (Figure 2, lane C, right).
Lastly, we analyzed the cell–cell adhesion parameter through the membrane expression of the E-cadherin protein. Upon vehicle-only addition, E-cadherin was, as expected, clearly expressed at the basal poles of the cells and concentrated at the cell–cell interface (Figure 2, lane D left). Extracts 6, 10 and 15 clearly abolish the participation of E-cadherin in the membrane architecture whereas the others slightly modify its intracellular distribution.
Among the sixteen extracts tested, extracts 2, 6, and 10 exhibited the strongest cytotoxicity. For extract 10 from H. columnaris, the toxicity could be due to apoptosis following microtubule perturbation, the induction of double-strand breakage in the DNA, and the loss of cell–cell adhesion. For extract 6 from H. kuesteri, toxicity maybe related to centrosome duplication and apoptosis. In contrast, the analysis of the targets studied in this study did not allow us to explain the toxicity of extract 2 from Croton lechleri.

4. Materials and Methods

4.1. Chemical Compounds Studied in this Article

The following chemical compounds were included in our study: tricin (PubChem CID: 5281702); serratenediol (PubChem CID: 164947); 21-episerratenediol (PubChem CID: 12309682); serratenediol-3-O-acetate, pinostrobin (PubChem CID: 73201), pallidine (PubChem CID: 12313923), and O-methylpallidine (PubChem CID: 10405046); the flavone 5-hydroxy-4′,7-dimethoxyflavone (apigenin 7,4′-dimethyl ether) (PubChem CID: 5281601), and hernandulcin (PubChem CID: 125608).

4.2. Origin of Plant Material for Obtaining Extract Preparation

For the current study, various parts of medicinal plants, including leaves, flowers, roots, fruits, bark, latex, and aerial parts (leaves and stems), were collected between March 2009 and July 2017. This collection took place in the Loja and Zamora Chinchipe provinces, located in the southern region of Ecuador. These provinces are home to three significant traditional cultures of the South of Ecuador: Campesinos, Shuar, and Saraguros, as shown in Figure 3.
Plant identification was overseen by Bolivar Merino, Curator of the Herbarium Loja (HUNL) at the Universidad Nacional de Loja (UNL), who cross-referenced them with reference samples stored in the Herbarium. Voucher specimens of these plants were deposited at the Department of Chemistry of Universidad Técnica Particular de Loja (UTPL).
This collection process was carried out under the scientific investigation permission of the Ministry of Environment of Ecuador (MAE) under reference number No. 001-IC-FLO-DBAP-VS-DRLZCH-MA. Notably, the plant Phyla strigulosa was collected in March 2013 from the parish of Mejeche, canton Yantzaza, in province of Zamora Chinchipe, Ecuador. After collection, it was cultivated in a greenhouse at the conservation garden of Tumbaco (Pichincha province). Leaves were collected and selected by the Instituto Nacional de Investigaciones Agropecuarias (INIAP), and a botanical sample was prepared and assigned the voucher number MT-KN-111.
Ethnobotanical details, including the plant’s scientific name and family, common name in Spanish or Kichwa language, traditional medicinal use, laboratory extraction method, and the survival ratio of MCF7 cells in the presence of 20 or 100 µg/mL of each extract, are presented in Table 1 and Table 2. The systematic and nomenclature for each species were aligned with the Catalogue of the Vascular Plants of Ecuador [55] and the scientific names were cross-referenced with the database of http://www.theplantlist.org/ (accession date: 16 June 2017) [56].

4.3. Preparation of Extracts

All organic solvents used to extract the plants (EtOH, MeOH, MeCl2, EtOAc, and hexane) were reagent-grade and had been purchased from Sigma-Aldrich (St. Louis, MO, USA).

4.3.1. Crude Extracts

Air-dried (35 °C) and milled plant parts selected (leaves, flowers, roots, fruits, bark, or latex) from each plant were separately extracted at one time at room temperature for two weeks with pure solvent reagent grade or a mixture of solvents (hexane, dichloromethane, ethyl acetate, methanol, ethanol, ethanol 70%, or ethanol 80%) as mentioned. Each filtrate was evaporated to dryness under reduced pressure at 40 °C to obtain 71 crude extracts that were kept in sealed amber glass vials at 4 °C until analysis.

4.3.2. Lyophilized Extracts

The traditional aqueous preparation of guabiduca (Piper crassinervium Kunth), used as water of time, tonic, and diuretic, was purchased in a community assembly in the sector Kiim, in Zamora Chinchipe. The infusion was filtered, centrifuged, frozen at −40 °C, and lyophilized under vacuum in Labconco, model 7754047, series 10083033 (Kansas City, MI, USA) equipment for 72 h until dry powder was obtained. Dry extract was weighed, placed in a vial, labeled, and stored at −20 °C until its use.

4.3.3. Alkaloid Extracts

Approximately 200–300 g of each selected dried plant (Table 1) was exhaustively extracted with a hydro-alcoholic mixture MeOH-H2O (80:20), then the solvent was removed via vacuum distillation, obtaining a dry extract (total extract), which was subjected to an acid-based extraction to obtain the alkaloid fraction [26, 31]. In this case, the total extract was suspended in dilute aqueous sulfuric acid (2% v/v), the suspension was filtered to separate the precipitated solids, and the process was repeated until negative reaction to the Dragendorff´s reagent. The combined aqueous phases were alkalinized until pH 11 by the addition of concentrated ammonia and extracted with chloroform until a negative reaction to the Dragendorff´s reagent occurred. Then, the combined organic phases were distilled under reduced pressure to remove the solvent and obtain the total alkaloid fraction of each plant selected for this study. The extracts of alkaloids were stored in dark flasks at 4 °C until they were analyzed.

4.3.4. Pure Compounds

The pure compounds comprised three serratane triterpenoids, serratenediol (serrat-14-en-3β,21α-diol) (1), serratenediol-3-O-acetate (2), and 21-episerratenediol (serrat-14-en-3β,21β-diol) (3), that were isolated in a previous study from the aerial part of Huperzia crassa [31]. The flavone tricin (5, 7, 4′-trihydroxy-3′, 5′-dimethoxyflavone) (4) was isolated from the aerial part of Huperzia brevifolia as previously described [26,31]. The flavone pinostrobin (2s)-5-hydroxy-7-methoxyflavanone), compound (5), was isolated from the Piper ecuadorense species [50]. The alkaloids pallidine (6) and O-methylpallidine (7) were isolated from the aerial part of Croton elegans [57]. The flavone 5-hydroxy-4′,7-dimethoxyflavone (Apigenin 7,4′-dimethyl ether) (8) was isolated from Piper peltatum [58]. At the end, the compound hernandulcin ((6S)-6-[(2S)-2-hydroxy-6-methylhept-5-en-2-yl]-3-methylcyclohex-2-en-1-one), a sesquiterpene (9), was isolated from the species Phyla strigulosa [59].
The structures of the compounds (19) (see Figure 4) were identified on the basis of extensive spectroscopic analysis. The complete isolation process for each compound previously indicated is detailed in the reported bibliography.
The biological activity of the crude extracts obtained in Ecuador (Table 1) was evaluated in the Institut de Recherche enCancérologie de Montpellier, France. Two permits from the Ministry of the Environment of Ecuador (MAE) were used to send the extracts (crude and lyophilized) and pure compounds to France (MAE, No. 013-2014-IC-FAU-DPL-MA and MAE, No. 015-2014-IC-FAU-DPL-MA) under the authorization of scientific research project No. 047-IC-FLO-DPL-MA.

4.4. Biological Experiments

MCF7 HTB-22 cell line was from ATCC (Manassas, VA, USA), cell culture media and chemical reagents have been purchased from local distributors of Sigma-Aldrich (St. Louis, MO, USA).

4.4.1. Cell Culture

All experiments were conducted on the MCF7 cell line, which was treated the day after plating in RPMI medium containing 10% FCS. Cells were exposed to the selected compounds for 72 h before being analyzed. DMSO was utilized for all treatments, with a final concentration of 1%. Each experiment was performed a minimum of twice.

4.4.2. Cytotoxicity Evaluation

The viability of cells after concentration-dependent treatments was determined using the standard sulfo-rhodamine B assay, which measures cellular protein content. Cells were seeded at 5 × 103 cells/well in 96-well plates. After treatment, cell monolayers were washed in phosphate buffer saline (PBS), fixed with 50% (w/v) trichloroacetic acid, and stained for 30 min in 0.4% sulfo-rhodamine B solution. The excess dye was then removed through washing repeatedly with 1% (v/v) acetic acid. The protein-bound dye was dissolved in 10 mM Tris base solution for OD determination at 540 nm using a microplate reader. IC50 values were determined from a nonlinear regression model using the online GNUPLOT package (www.ic50.tk, www.gnuplot.info, accessed on 30 April 2022).

4.4.3. Cell-Cycle Analysis

The cell-cycle analysis was performed on MCF7 cell line seeded at 4 × 105 cells/well in 6-well plate. After treatment, the cells were trypsinized, washed twice with chilled PBS, and spun in a cold centrifuge at 600 g for 5 min. The resulting pellet was fixed by resuspending it in 500 µL of chilled PBS followed by dropwise addition of 1.5 mL cold (−20 °C) ethanol. The cells were washed again twice with PBS, centrifuged at 600 g for 5 min, and resuspended in 500 µL PBS containing 100 µg/mL RNAse and 40 µg/mL propidium iodide. The staining reaction was allowed to proceed for 2 h at 37 °C. The DNA fluorescence was analyzed on a Gallios flow cytometer (Beckman Coulter France SAS, Villepinte, France). The results were analyzed using FlowJo cell-cycle analysis software (www.flowjo.com).

4.4.4. Immunofluorescence Microscopy Analysis

Cells were seeded onto glass coverslips at 105 cells/well in 6-well plates. Twenty-four hours later, cells were incubated for 72 h with 25 µg/mL of each extract excepted 12.5 µg/mL for extract 2. After treatment, the cells were washed with PBS. For the visualization of microtubule network and centrosomes, the cells were fixed with cold (−20 °C) methanol for 10 min, gradually rehydrated with PBS, and incubated for 2 h at 37 °C with mouse anti-β-tubulin (clone TUB 2.1, T4026, Sigma, St. Louis, MO, USA) or rabbit anti-γ-tubulin (T3559, Sigma) primary antibodies diluted 1:500 in PBS-BSA 0.1%. For the visualization of γ-H2AX foci and E-cadherin, the cells were fixed with 3.7% paraformaldehyde plus 1% methanol for 5 min at 37 °C, permeabilized in PBS-Triton 0.1% for 5 min at 25 °C, washed in PBS, and then incubated for 1 h at 25 °C with mouse anti-phospho-histone H2A.X (Ser139) (clone JBW301, 05-636, Sigma) primary antibody diluted 1:1000 or mouse anti-cadherin (clone36E BD biosciences 610182) diluted 1/100 in PBS-Tween20 0.1% for 1 h at 37 °C. Subsequently, the coverslips were washed with PBS, incubated with anti-mouse or rabbit rhodamine-conjugated secondary antibodies (Rockland) diluted 1:200 in PBS-BSA 0.1% for 1 h at 37 °C. DAPI (4′, 6-diamidino-2-phenylindole) staining was then performed for 15 min followed by further PBS washing, air drying, and embedding in Mowiol-containing mounting medium. Fluorescence was detected using a Leica DM-RM microscope. Images were acquired with objective magnification 63× and processed using GIMP software version 2.10.34.

4.4.5. Evaluation of the Effects on Centrosomes and γH2AX Foci

For this assessment, a minimum of three and up to six images were analyzed in a double-blinded manner for each condition. For centrosomes, the total number of centrosomes was divided by the cell number identified by their DAPI-stained nuclei. An average of 12 cells per image was counted for each analysis. The same methodology was applied for the counting of γH2AX foci.

5. Perspectives

In order to attain a more profound comprehension of the underlying mechanisms responsible for the tumoral cell-line toxicity observed in these plant extracts, it becomes imperative to undertake the tasks of isolating, characterizing, and evaluating pure compounds. Nevertheless, recent demonstrations of the “cocktail” effect, resulting from the synergistic potential of the combined presence of chemicals in concentrations that are individually inert [60,61], are a compelling impetus for us to intensify our efforts to study the complex formulations prepared by traditional healers from plant mixtures using the technical capabilities of modern medicine and biology [62,63,64,65].

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/plants13101422/s1, Figure S1: Microtubule network upon incubation with extracts; Figure S2: Centrosomal gTub localization upon incubation with the extracts; Figure S3: Nuclear gH2aX foci—formation upon incubation with the extracts; Figure S4: Cadherine localization upon incubation with extracts.

Author Contributions

All authors (N.B., C.L. and C.A.) contributed to the study reported in this paper: conceptualization, investigation, methodology, data curation, formal analysis, resources, validation, writing—review and editing, writing—original draft, and writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the SENESCYT (National Secretary of Higher Education, Science, Technology, and Innovation from Ecuador) under the SENECYT-PROMETEO program (SENECYT 2015-2016) and Scientific Research Project of Universidad Técnica Particular de Loja (PROY_PROY_ARTIC_QU_2022_3652).

Data Availability Statement

Data will be made available on request.

Acknowledgments

The authors thank Nadia Vie (ICM Montpellier) for the FACS experiments and Drs Silvia Gonzalez, Omar Malagon, and Gianluca Gilardoni (UTPL) for their support. We thank Dayana Vidal, Jorge Ramírez, and Claudia Herrera from the UTPL Department of Chemistry for providing some of the pure compounds evaluated in this study and the National Institute for Agricultural Research from Ecuador (INIAP) for providing the species Phyla strigulosa.

Conflicts of Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

References

  1. Shen, Y. History, Present and Prospect of World Traditional Medicine (In 2 Volumes), 1st ed.; World Scientific: Singapore, 2023. [Google Scholar]
  2. Graves, T.C. Commentary on a Case of Hystero-epilepsy with delayde puberty treated with testicular extract. Lancet 1920, 196, 1134–1135. [Google Scholar] [CrossRef]
  3. Beecher, H.K. The powerful placebo. J. Am. Med. Assoc. 1995, 159, 1602–1606. [Google Scholar] [CrossRef] [PubMed]
  4. World Health Organization. WHO Traditional Medicine Strategy: 2014–2023, 1st ed.; WHO Library Cataloguing: Hong Kong SAR, China, 2013. [Google Scholar]
  5. Jones, K. Review of Sangre de Drago (Croton lechleri)—A south American tree sap in the treatment of diarrhea, inflammation, insect bites, viral infections, and wounds: Traditional uses to clinical research. J. Altern. Complement. Med. 2003, 9, 877–896. [Google Scholar] [CrossRef]
  6. Tene, V.; Malagón, O.; Finzi, P.V.; Vidari, G.; Armijos, C.; Zaragoza, T. An ethnobotanical survey of medicinal plants used in Loja and Zamora-Chinchipe, Ecuador. J. Ethnopharmacol. 2007, 111, 63–81. [Google Scholar] [CrossRef] [PubMed]
  7. Malagón, O.; Ramírez, J.; Andrade, M.J.; Morocho, V.; Armijos, C.; Gilardoni, G. Phytochemistry and Ethnopharmacology of the Ecuadorian Flora. A Review. Nat. Prod. Commun. 2016, 11, 297–314. [Google Scholar] [CrossRef] [PubMed]
  8. Armijos, C.; Quisatagsi, E.V.; Cuenca, M.; Cuenca-Camacho, S.; Bailón-Moscoso, N. The cytotoxic principle of Bejaria resinosa from Ecuador. J. Pharmacogn. Phytochem. 2015, 4, 268–272. [Google Scholar]
  9. Armijos, C.; Ramírez, J.; Salinas, M.; Vidari, G.; Suárez, A.I. Pharmacology and Phytochemistry of Ecuadorian Medicinal Plants: An Update and Perspectives. Pharmaceuticals 2021, 14, 1145. [Google Scholar] [CrossRef] [PubMed]
  10. Yu, S.; Li, A.; Liu, Q.; Yuan, X.; Xu, H.; Jiao, D.; Pestell, R.G.; Han, X.; Wu, K. Recent advances of bispecific antibodies in solid tumors. J. Hematol. Oncol. 2017, 10, 155. [Google Scholar] [CrossRef]
  11. Trenevska, I.; Li, D.; Banham, A.H. Therapeutic Antibodies against Intracellular Tumor Antigens. Front. Immunol. 2017, 8, 1001. [Google Scholar] [CrossRef] [PubMed]
  12. Greene, C.; Barlesi, B.; Tarroza-David, S.; Friedlander, T. Improved Control of Tyrosine Kinase Inhibitor-Induced Diarrhea with a Novel Chloride Channel Modulator: A Case Report. Oncol. Ther. 2021, 9, 247–253. [Google Scholar] [CrossRef]
  13. Harrison, F.; Roberts, A.E.; Gabrilska, R.; Rumbaugh, K.P.; Lee, C.; Diggle, S.P. A 1000-Year-Old Antimicrobial Remedy with Antistaphylococcal Activity. mBio 2015, 6, 10–1128. [Google Scholar] [CrossRef] [PubMed]
  14. Tu, Y. Artemisinin-A Gift from Traditional Chinese Medicine to the World (Nobel Lecture). Angew. Chem. Int. Ed. Engl. 2016, 55, 10210–10226. [Google Scholar] [CrossRef]
  15. Dettweiler, M. American Civil War plant medicines inhibit growth, biofilm formation, and quorum sensing by multidrug-resistant bacteria. Sci. Rep. 2019, 9, 7692. [Google Scholar] [CrossRef] [PubMed]
  16. Gao, J.J.; Tan, M.; Pohlmann, P.R.; Swain, S.M. HALT-D: A Phase II Evaluation of Crofelemer for the Prevention and Prophylaxis of Diarrhea in Patients With Breast Cancer on Pertuzumab-Based Regimens. Clin. Breast Cancer. 2017, 17, 76–78. [Google Scholar] [CrossRef]
  17. Zhong, C.; Wall, N.R.; Zu, Y.; Sui, G. Therapeutic Application of Natural Medicine Monomers in Cancer Treatment. Curr. Med. Chem. 2017, 24, 3681–3697. [Google Scholar] [CrossRef] [PubMed]
  18. Mohamed Abdoul-Latif, F.; Ainane, A.; Houmed Aboubaker, I.; Mohamed, J.; Ainane, T. Exploring the Potent Anticancer Activity of Essential Oils and Their Bioactive Compounds: Mechanisms and Prospects for Future Cancer Therapy. Pharmaceuticals 2023, 16, 1086. [Google Scholar] [CrossRef]
  19. Akone, S.H.; Ntie-Kang, F.; Stuhldreier, F.; Ewonkem, M.B.; Noah, A.M.; Mouelle, S.E.M.; Müller, R. Natural Products Impacting DNA Methyltransferases and Histone Deacetylases. Front. Pharmacol. 2020, 11, 992. [Google Scholar] [CrossRef]
  20. Calderón-Montaño, J.M.; Martínez-Sánchez, S.M.; Jiménez-González, V.; Burgos-Morón, E.; Guillén-Mancina, E.; Jiménez-Alonso, J.J.; Díaz-Ortega, P.; Garcia, F.; Aparicio, A.; López-Lázaro, M. Screening for Selective Anticancer Activity of 65 Extracts of Plants Collected in Western Andalusia, Spain. Plants 2020, 10, 2193. [Google Scholar] [CrossRef] [PubMed]
  21. Dehelean, C.A.; Marcovici, I.; Soica, C.; Mioc, M.; Coricovac, D.; Iurciuc, S.; Cretu, O.M.; Pinzaru, I. Plant-Derived Anticancer Compounds as New Perspectives in Drug Discovery and Alternative Therapy. Molecules 2021, 26, 1109. [Google Scholar] [CrossRef]
  22. Le, T.T.; Wu, M.; Lee, J.H.; Bhatt, N.; Inman, J.T.; Berger, J.M.; Wang, M.D. Etoposide promotes DNA loop trapping and barrier formation by topoisomerase II. Nat. Chem. Biol. 2008, 19, 641–650. [Google Scholar] [CrossRef]
  23. Pasquier, E.; Kavallaris, M. Microtubules: A dynamic target in cancer therapy. IUBMB Life 2008, 60, 165–170. [Google Scholar] [CrossRef] [PubMed]
  24. Kumar, B.; Singh, S.; Skvortsova, I.; Kumar, V. Promising Targets in Anti-cancer Drug Development: Recent Updates. Curr. Med. Chem. 2017, 24, 4629–4752. [Google Scholar] [CrossRef] [PubMed]
  25. Andrade, J.; Lucero Mosquera, H.; Armijos, C. Ethnobotany of Indigenous Saraguros: Medicinal Plants Used by Community Healers “Hampiyachakkuna” in the San Lucas Parish, Southern Ecuador. BioMed. Res. Int. 2017, 2017, 9343724. [Google Scholar] [CrossRef] [PubMed]
  26. Armijos, C.; Ponce, J.; Ramírez, J.; Gozzini, D.; Finzi, P.V.; Vidari, G. An Unprecedented High Content of the Bioactive Flavone Tricin in Huperzia Medicinal Species Used by the Saraguro in Ecuador. Nat. Prod. Commun. 2016, 11, 273–274. [Google Scholar]
  27. Armijos, C.; Cota, I.; González, S. Traditional medicine applied by the Saraguro yachakkuna: A preliminary approach to the use of sacred and psychoactive plant species in the southern region of Ecuador. J. Ethnobiol. Ethnomed. 2014, 10, 26. [Google Scholar] [CrossRef] [PubMed]
  28. Aguirre, Z.; Yaguana, C.; Merino, B. Plantas Medicinales de la Zona Andina de la Provincia de Loja, 1st ed.; Herbario y Jardín Botánico “Reinaldo Espinosa”: Loja, Ecuador, 2014; p. 193. [Google Scholar]
  29. De la Torre, L.; Navarette, H.; Muriel, P.; Macia, M.J.; Balslev, H. Enciclopedia de las Plantas Útiles del Ecuador, 1st ed.; Herbario QCA de la Escuela de Ciencias Biologicas de la Pontifica Universidad Católica del Ecuador, Quito, Ecuador & Herbario AAU del Departamento de Ciencias Biologicas de la Universidad Aarhus: Aarhus, Denmark, 2008. [Google Scholar]
  30. Bussmann, R.W.; Sharon, D. Traditional medicinal plant use in Loja province, Southern Ecuador. J. Ethnobiol. Ethnomed. 2006, 2, 44. [Google Scholar] [CrossRef] [PubMed]
  31. Armijos, C.; Gilardoni, G.; Amay, L.; Lozano, A.; Bracco, F.; Ramírez, J.; Bec, N.; Larroque, C.; Finzi, P.V.; Vidari, G. Phytochemical and ethnomedicinal study of Huperzia species used in the traditional medicine of Saraguros in Southern Ecuador; AChE and MAO inhibitory activity. J. Ethnopharmacol. 2016, 193, 546–554. [Google Scholar] [CrossRef] [PubMed]
  32. Hickey, B.J.; Lumsden, A.J.; Cole, A.L.J.; Walker, J.R.L. Antibiotic compounds from new zealand plants: Methyl haematommate, an anti-fungal agent from Stereocaulon ramulosum. N. Z. Nat. Sci. 1990, 17, 49–53. [Google Scholar]
  33. Compadre, C.M.; Pezzuto, J.M.; Kinghorn, A.D.; Kamath, S.K. Hernandulcin: An intensely sweet compound discovered by review of ancient literature. Science 1985, 227, 417–419. [Google Scholar] [CrossRef]
  34. Kaneda, N.; Lee, I.S.; Gupta, M.P.; Soejarto, D.D.; Kinghorn, A.D. (+)-4 beta-hydroxyhernandulcin, a new sweet sesquiterpene from the leaves and flowers of Lippia dulcis. J. Nat. Prod. 1992, 55, 1136–1141. [Google Scholar] [CrossRef]
  35. Armijos, C.; Valarezo, E.; Cartuche, L.; Zaragoza, T.; Finzi, P.V.; Mellerio, G.G.; Vidari, G. Chemical composition and antimicrobial activity of Myrcianthes fragrans essential oil, a natural aromatizer of the traditional Ecuadorian beverage colada morada. J. Ethnopharmacol. 2018, 225, 319–326. [Google Scholar] [CrossRef]
  36. Jadaun, A.; Subbarao, N.; Dixit, A. Allosteric inhibition of topoisomerase I by pinostrobin: Molecular docking, spectroscopic and topoisomerase I activity studies. J. Photochem. Photobiol. B 2017, 167, 299–308. [Google Scholar] [CrossRef] [PubMed]
  37. Muroyama, A.; Lechler, T. Microtubule organization, dynamics and functions in differentiated cells. Development 2017, 144, 3012–3021. [Google Scholar] [CrossRef] [PubMed]
  38. Wordeman, L.; Vicente, J.J. Microtubule Targeting Agents in Disease: Classic Drugs, Novel Roles. Cancers 2021, 13, 5650. [Google Scholar] [CrossRef]
  39. Forth, S.; Kapoor, T.M. The mechanics of microtubule networks in cell division. J. Cell Biol. 2017, 216, 1525–1531. [Google Scholar] [CrossRef]
  40. Song, S.; Jung, S.; Kwon, M. Expanding roles of centrosome abnormalities in cancers. BMB Rep. 2023, 56, 216–224. [Google Scholar] [CrossRef]
  41. Huang, R.; Zhou, P.K. DNA damage repair: Historical perspectives, mechanistic pathways and clinical translation for targeted cancer therapy. Signal Transduct. Target. Ther. 2021, 6, 254. [Google Scholar] [CrossRef]
  42. Alhmoud, J.F.; Woolley, J.F.; Al Moustafa, A.E.; Malki, M.I. DNA Damage/Repair Management in Cancers. Cancers 2020, 12, 1050. [Google Scholar] [CrossRef] [PubMed]
  43. Na, T.Y.; Schecterson, L.; Mendonsa, A.M.; Gumbiner, B.M. The functional activity of E-cadherin controls tumor cell metastasis at multiple steps. Proc. Natl. Acad. Sci. USA 2020, 117, 5931–5937. [Google Scholar] [CrossRef]
  44. Haraguchi, M.; Mimaki, Y.; Motidome, M.; Morita, H.; Takeya, K.; Itokawa, H.; Yokosuka, A.; Sashida, Y. Steroidal saponins from the leaves of Cestrum sendtenerianum. Phytochemistry 2000, 55, 715–720. [Google Scholar] [CrossRef]
  45. Alonso-Castro, Á.J.; Ortíz-Sánchez, E.; Domínguez, F.; López-Toledo, G.; Chávez, M.I.; De Jesús Ortiz-Tello, A.; García-Carrancá, A. Antitumor effect of Croton lechleri Mull. Arg. (Euphorbiaceae). J. Ethnopharmacol. 2012, 140, 438–442. [Google Scholar] [CrossRef]
  46. Chordia, P.; MacArthur, R.D. Crofelemer, a novel agent for treatment of non-infectious diarrhea in HIV-infected persons. Expert. Rev. Gastroenterol. Hepatol. 2013, 7, 591–600. [Google Scholar] [CrossRef] [PubMed]
  47. Pona, A.; Cline, A.; Kolli, S.S.; Taylor, S.L.; Feldman, S.R. Review of future insights of Dragon’s Blood in dermatology. Dermatol. Ther. 2019, 32, e12786. [Google Scholar] [CrossRef]
  48. Pardo-Andreu, G.L.; Núñez-Figueredo, Y.; Tudella, V.G.; Cuesta-Rubio, O.; Rodrigues, F.P.; Pestana, C.R.; Uyemura, S.A.; Leopoldino, A.M.; Alberici, L.C.; Curti, C. The anti-cancer agent guttiferone-A permeabilizes mitochondrial membrane: Ensuing energetic and oxidative stress implications. Toxicol. Appl. Pharmacol. 2011, 253, 282–289. [Google Scholar] [CrossRef] [PubMed]
  49. Setzer, W.N.; Setzer, M.C.; Moriarity, D.M.; Bates, R.B.; Haber, W.A. Biological Activity of the Essential Oil of Myrcianthes sp. nov. “Black Fruit” from Monteverde, Costa Rica. Planta Med. 1999, 65, 468–469. [Google Scholar] [CrossRef]
  50. Ramírez, J.; Cartuche, L.; Morocho, V.; Aguilar, S.; Malagon, O. Antifungal activity of raw extract and flavanons isolated from Piper ecuadorense from Ecuador. Rev. Bras. Farmacogn. 2013, 23, 370–373. [Google Scholar] [CrossRef]
  51. Valarezo, E.; Flores-Maza, P.; Cartuche, L.; Ojeda-Riascos, S.; Ramírez, J. Phytochemical profile, antimicrobial and antioxidant activities of essential oil extracted from Ecuadorian species Piper ecuadorense sodiro. Nat. Prod. Res. 2021, 35, 6014–6019. [Google Scholar] [CrossRef]
  52. Garavito, G.; Rincón, J.; Arteaga, L.; Hata, Y.; Bourdy, G.; Giménez, A.; Pinzón, R.; Deharo, E. Antimalarial activity of some Colombian medicinal plants. J. Ethnopharmacol. 2006, 107, 460–462. [Google Scholar] [CrossRef]
  53. Matvieieva, N.A.; Pasichnyk, L.A.; Zhytkevych, N.V.; Pabón Garcés Galo, J.; Pidgorskyi, V.S. Antimicrobial activity of extracts from ecuadorian lichens. Mikrobiolohichnyi Zhurnal 2015, 77, 23–27. [Google Scholar] [CrossRef]
  54. Wei, F.; Zhao, L.; Jing, Y. Mechanisms underlying dimethyl sulfoxide-induced cellular migration in human normal hepatic cells. Environ. Toxicol. Pharmacol. 2020, 80, 103489. [Google Scholar] [CrossRef]
  55. Jorgensen, P.; Leon-Yanez, S. Catalogue of the Vascular Plants of Ecuador; Missouri Garden Press: St. Louis, MO, USA, 1999. [Google Scholar]
  56. Plantlist. Plantlist 2013. Available online: http://www.theplantlist.org (accessed on 16 June 2017).
  57. Herrera, C.; Pérez, Y.; Morocho, V.; Armijos, C.; Malagón, O.; Brito, B.; Tacán, M.; Cartuche, L.; Gilardoni, G. Preliminary phytochemical study of the ecuadorian plant Croton elegans Kunth (Euphorbiaceae). J. Chil. Chem. Soc. 2018, 63, 3875–3877. [Google Scholar] [CrossRef]
  58. Vidal, D.C.E. Aislamiento, Caracterización y Actividad biológica de Metabolitos Secundarios a Partir de la Sspecie Piper peltatum. Ph.D. Thesis, Biochemistry and Pharmacy, Universidad Tecnica Particular Loja, Loja, Ecuador, 2013. [Google Scholar]
  59. Vega, M.; Brito, B.; Malagon, O. Application of qNMR in the Characterization of Hernandulcin in the Species Phyla strigulosa; Congresso SILAE: Carthagena, Colombia, 2017. [Google Scholar]
  60. Hamid, N.; Junaid, M.; Pei, D.S. Combined toxicity of endocrine-disrupting chemicals: A review. Ecotoxicol. Environ. Saf. 2021, 215, 112136. [Google Scholar] [CrossRef] [PubMed]
  61. Kong, W.Y.; Ngai, S.C.; Goh, B.H.; Lee, L.H.; Htar, T.T.; Chuah, L.H. Is Curcumin the Answer to Future Chemotherapy Cocktail? Molecules 2021, 26, 4329. [Google Scholar] [CrossRef] [PubMed]
  62. Huang, Y.; Lü, J.; Zhao, Q.; Chen, J.; Wang, D.; Lin, M.; Zheng, H. Potential Therapeutic Mechanism of Traditional Chinese Medicine on Diabetes in Rodents: A Review from an NMR-Based Metabolomics Perspective. Molecules 2022, 27, 5109. [Google Scholar] [CrossRef] [PubMed]
  63. Li, W.; Zhao, F.; Yang, J.; Pan, J.; Qu, H. Development of a comprehensive method based on quantitative 1H NMR for quality evaluation of Traditional Chinese Medicine injection: A case study of Danshen Injection. J. Pharm. Pharmacol. 2022, 74, 1006–1016. [Google Scholar] [CrossRef] [PubMed]
  64. Wang, X.J.; Ren, J.L.; Zhang, A.H.; Sun, H.; Yan, G.L.; Han, Y.; Liu, L. Novel applications of mass spectrometry-based metabolomics in herbal medicines and its active ingredients: Current evidence. Mass. Spectrom. Rev. 2019, 38, 380–402. [Google Scholar] [CrossRef]
  65. Wen, X.; Shi, J.; Tan, W.; Jiang, H.; Wang, D.; Su, J.; Yang, G.; Zhang, B. Effects of aromatherapy and music therapy on patients’ anxiety during MRI examinations: A randomized controlled trial. Eur. Radiol. 2023, 33, 2510–2518. [Google Scholar] [CrossRef]
Figure 1. Description of cell cycle and illustration of a typical FACS experiment for the study of the extract effect on the cell cycle of MCF7 cell line incubated with each compound at 25 µg/mL for 72 h.
Figure 1. Description of cell cycle and illustration of a typical FACS experiment for the study of the extract effect on the cell cycle of MCF7 cell line incubated with each compound at 25 µg/mL for 72 h.
Plants 13 01422 g001
Figure 2. Illustration of the effects on cellular substructures observed after incubation of the MCF7 cell line in RPMI, 10% FCS at 37 °C, 5% CO2 supplemented with DMSO 1% or plant extracts at 25 µg/mL. Only two pictures for each cell structure are presented and are representative of the feature observed. For each lane, the following are shown: left pictures—reference cells cultured in the presence of 1% DMSO; center pictures illustrate a ‘moderate effect’ on the structure, and right pictures show the strongest observed effect. Label of the compound referring to its identity is inserted in each panel. Lane A: Microtubular network, Lane B: centrosomal substructure localized by white arrows, Lane C: gH2AX-positive DNA double-strand breaks localized with white arrows except in the right panel where they are to numerous. Lane D: E-Cadherin assembly. For clarity, merged picture with DAPI nuclear staining is not presented.
Figure 2. Illustration of the effects on cellular substructures observed after incubation of the MCF7 cell line in RPMI, 10% FCS at 37 °C, 5% CO2 supplemented with DMSO 1% or plant extracts at 25 µg/mL. Only two pictures for each cell structure are presented and are representative of the feature observed. For each lane, the following are shown: left pictures—reference cells cultured in the presence of 1% DMSO; center pictures illustrate a ‘moderate effect’ on the structure, and right pictures show the strongest observed effect. Label of the compound referring to its identity is inserted in each panel. Lane A: Microtubular network, Lane B: centrosomal substructure localized by white arrows, Lane C: gH2AX-positive DNA double-strand breaks localized with white arrows except in the right panel where they are to numerous. Lane D: E-Cadherin assembly. For clarity, merged picture with DAPI nuclear staining is not presented.
Plants 13 01422 g002
Figure 3. Settlement of healer communities from the south region of Ecuador, in Loja and Zamora Chinchipe cantons, that traditionally use plants that were of concern in this study.
Figure 3. Settlement of healer communities from the south region of Ecuador, in Loja and Zamora Chinchipe cantons, that traditionally use plants that were of concern in this study.
Plants 13 01422 g003
Figure 4. Pure compounds isolated from Ecuadorian plants.
Figure 4. Pure compounds isolated from Ecuadorian plants.
Plants 13 01422 g004
Table 1. Identification, traditional names, and usage of plants chosen and collected during the two campaigns. Plant parts used and secondary metabolite extraction conditions are presented. MCF 7 cell line survival after 72 h is reported in terms of percentage of surviving cells. Each plant extract identified as ‘selected compound’ is listed in Table 2 and subjected to further investigation to assess its impact on cellular pathways.
Table 1. Identification, traditional names, and usage of plants chosen and collected during the two campaigns. Plant parts used and secondary metabolite extraction conditions are presented. MCF 7 cell line survival after 72 h is reported in terms of percentage of surviving cells. Each plant extract identified as ‘selected compound’ is listed in Table 2 and subjected to further investigation to assess its impact on cellular pathways.
Plants and Compounds EvaluatedFamilyCommon NameTraditional UsesExtraction ConditionProduct (µg/mL)MCF Cell Line Survival Ratio (%)
Control substances:
RPMI media-----100
DMSO-----100
TAXOL 1000 nM----0.818
TAXOL 10 nM----0.00826
TAXOL 1nM----0.000894
Pure compounds evaluated:
Serratenediol-3-O-acetate (C32H5203)-Pure compound isolated from Huperzia crassa species-4896
Tricin (5, 7, 4′-trihydroxy-3′, 5′-dimethoxyflavone) C17H14O7-Pure compound isolated (Flavone) from various species from Huperzia genderPubChem CID:
5281702
3388
5-hydroxy-4′,7-dimethoxyflavone (apigenin 7,4′-dimethyl ether) C17H14O5-Pure compound isolated from Piper peltatum L.PubChem CID:
5281601
3027
21-episerratenediol (serrat-14-en-3β,21β-diol)
(C30H50O2)
-Pure compound isolated from Huperzia crassaPubChem CID:
12309682
10047
2097
Serratenediol
(serrat-14-en-3β,21α- diol) C30H50O2
Pure compound isolated from Huperzia crassaPubChem CID: 164947100109
20100
Pinostrobin (2s)-5-hydroxy-7-methoxyflavanone) C16H14O4 Pure compound isolated from Piper ecuadorensePubChem CID: 7320110060
2037
Pallidine (2-hydroxy-3,6-dimethoxy-17-methyl-5,6,8,14-tetradehydromorphinan-7-one)
C19H21NO4
Pure compound isolated from Croton elegansPubChem CID: 1231392310074
20103
O-methylpallidine ((1S,9S)-4,5,13-trimethoxy-17-methyl-17-azatetracyclo[7.5.3.01,10.02,7]heptadeca-2,4,6,10,13-pentaen-12-one) C20H23NO4 Pure compound isolated from Croton elegansPubChem CID: 10405046100108
20107
Hernandulcin ((6S)-6-[(2S)-2-hydroxy-6-methylhept-5-en-2-yl]-3-methylcyclohex-2-en-1-one) C15H24O2 Pure compound isolated from Phyla strigulosaPubChem CID: 12560810010
2067
Species evaluated/Herbarium voucher
Acanthoxanthium spinosum (L.) Fourr./PPN-as-039AsteraceaeCasamarucha, cardo de tres puntasTreat conditions of the prostate and kidneys and urinary tract infection (oral testimony); anti-inflammatory and blood purifier [28]EtOAc extract (leaves)10031
20103
MeCl2 extract (leaves)10014
2093
Baccharis obtusifolia Kunth/PPN-as-014AsteraceaeChilca, chilca redonda, shadánAntimycotic, cold, rheumatism [6]MeOH extract (leaves)10084
2094
Croton elegans Kunth/HUTPL536EuphorbiaceaeMosqueraAnti-inflammatory; powerful purgative; treatment of rheumatism, neuralgia, and bronchitis [9]Alkaloid fraction from MeOH-H2O extract (8:2) (leaves)10046
2080
Cinchona officinalis L./PPN-ry-002RubiaceaeCascarilla, quinaStomach pain; fever; malaria; antimycotic [6]EtOH extract (bark)10090
2093
Clusia alata Triana & Planch./HUTPL5081ClusiaceaeDucoGastritis [28]MeOH extract (leaves)10018
2091
Croton lechleri Müll. Arg./PPN-eu-003EuphorbiaceaeSangre de dragoSelected compoundLatex1005
204
Renealmia alpinia (Rottb.) Maas./HUTPL 11186ZingiberaceaeKumpíaThe leaves are used to treat rheumatism; a blue pigment is obtained from the fruit [29]Lyophilized aqueous fruit extract1006
2071
Garcinia macrophylla Mart./HUTPL3841ClusiaceaeShoraSelected compoundMeOH extract (leaves)10014
2053
Huperzia brevifolia (Grev. & Hook.) Holub/PPNIc-10LycopodiaceaeWaminga verdeLiver and kidney diseases, fever, inflammation, colds [30]Alkaloid fraction from MeOH-H2O extract (8:2) (leaves)1004
2071
Huperzia columnaris B. Øllg./PPNIc-09LycopodiaceaeWaminga osoSelected compoundAlkaloid fraction from MeOH-H2O extract (8:2) (leaves)1003
2042
Huperzia compacta (Hook.) Trevis./PPNIc-02LycopodiaceaeWaminga rojaActs as a purgative and to treat supernatural diseases [31]Alkaloid fraction from MeOH-H2O extract (8:2) (leaves)10090
2094
Huperzia crassa (Humb. & Bonpl. ex Willd.) Rothm./PPNIc-05LycopodiaceaeWaminga amarillaTo treat the itching of the body [29]Alkaloid fraction from MeOH-H2O extract (8:2) (leaves)10082
2098
Huperzia espinosana B. Øllg/PPNIc-08LycopodiaceaeWaminga oso warmiLiver and kidney diseases, fever, inflammation, colds [30]Alkaloid fraction from MeOH-H2O extract (8:2) (leaves)1004
2078
Huperzia kuesteri (Nessel) B. Øllg./Ly-HK-001LycopodiaceaeWaminga verde grandeSelected compoundAlkaloid fraction from MeOH-H2O extract (8:2) (leaves)1004
2073
Huperzia tetragona (Hook. & Grev.) Trevis./PPNIc-04LycopodiaceaeTrencilla rojaTreatment of elephantiasis and leprosy and to treat supernatural diseases [31]Alkaloid fraction from MeOH-H2O extract (8:2) (leaves)10081
2093
Hypericum lancioides Cuatrec./PP-hy-001HypericaceaeBura buraAntidepressant effects; antioxidant, antimicrobial, and antiviral properties MeCl2 extract (aerial part)10023
2088
Lycopodium complanatum L./Ly-001-08LycopodiaceaeGateador, trencillaIn bathrooms during the postpartum period for bone pain in childrenAlkaloid fraction from MeOH-H2O extract (8:2) (leaves)10051
2076
Loricaria thuyoides (Lam.) Sch. Bip./PPN-as-044AsteraceaeUshkuchakiUsed in baths after childbirth to treat hip pain and cold and treat the mal aire (bad air) [20]EtOAc extract (leaves and stem)10093
20100
Ludwigia peruviana (L.) H. Hara./PPN-on-003OnagraceaeMejoranaHepatic pain, diuretic, kidney problems [6]MeOH-H2O extract (9:1) (leaves and stem)10054
2097
Macrocarpaea lenae J. R. Grant/PPN-gn-003GentianaceaeTabaco de cerroFever or cold caused by cold air or strong winds locally known as mal aire (bad air) [25]Alkaloid fraction from MeOH-H2O extract (8:2) (flowers and leaves)10052
2093
Sarcorhachis sydowii Trel./Pi-003-010PiperaceaeIntiwaskaThe infusion of the leaves is drunk to treat stomach pain [29]MeOH-H2O extract (9:1) (leaves and stem)10056
2096
Lyophilized aqueous leaves and stem extract10031
2093
Oreopanax andreanus Marchal/PPN-ar-003AraliaceaePumamakiDisinfectant, healing of wounds, dermatitis [6]MeOH extract (leaves)100102
2097
Curcuma longa L./HUTPL 14333ZingiberaceaeCúrcuma, urmeric, perenchiThe plant is traditionally known for its fungicidal and bactericidal properties [9]Lyophilized aqueous tuber extract10087
2085
Piper pseudochurumayu (Kunth) C. DC./PPN-pi-009PiperaceaeMatico, ámbarámbarSelected compoundMeOH extract (leaves and stem)1005
2054
Siparuna eggersii Hieron./PPN-mn-001MonimiaceaeMonte del osoStrokes, diabetes, fractured bones, rheumatism, kidney problems [6]MeOH extract (leaves and stem)10039
2095
Piper crassinervium Kunth/PPN-pi-002PiperaceaeGuabiducaDiabetes, gastritis, prostate problems [6]Lyophilized aqueous leaf extract10096
20106
Juglans neotropica Diels/PPN-ju-001JuglandaceaeNogalRheumatism, hepatic pain in postpartum bath [25]Lyophilized aqueous leaf extract1005
20106
Tropaeolum tuberosum Ruiz & Pav./PPN-tr-001TropaeolaceaeMashuaProstate [25]Lyophilized aqueous tuber juice100103
20105
Valeriana pyramidalis Kunth./FT991ValerianaceaeValerianaTo treat nerves, heart, liver, and kidney problems [29]Lyophilized roots exudate10080
20105
Piper ecuadorense Sodiro/PPN-pi-007 PiperaceaeMatico grande, tiklilin grande, matico del monteSelected compoundEtOH-H2O extract (7:3) (leaves)10018
2054
Selected compoundMeOH extract (leaves)10024
20100
Alibertia sp.RubiaceaeMatiriThe fruits of several Alibertia species are edible [29]MeOH extract (leaves)10043
2085
Artemisia sodiroi Hieron./PPN-as-021AsteraceaeAjenjoInternal inflammation, stomach pain, hepatic pain, fever, internal infections, kidney problems, cough [6]MeOH extract (leaves)10071
20111
Artocarpus altilis (Parkinson) Fosberg/PPN-mo-003MoraceaeFruto del panDiabetes, high cholesterol [6]MeOH extract (leaves)10090
20104
Bejaria resinosa Mutis ex L.f./PPN-er-002EricaceaePayama, pena pena, pena de cerroTo treat nervous system problems, swollen wounds and inflammations of the genital organs, as well liver diseases and cancer [8]MeOH extract (leaves)1006
2089
Brugmansia suaveolens (Willd.) Bercht. & J. Presl/PPNso-021SolanaceaeFloripondio rosado, guando rosadoTo treat rheumatic pain [27]EtOH-H2O (8:2) (flowers)10094
20110
Brugmansia versicolor Lagerh./PPN-so-027SolanaceaeFloripondio, guandoTo treat headache and inflammation and swelling from blows and act as psychoactive plant [29]Alkaloid fraction from MeOH-H2O extract (8:2) (flowers)10084
20125
Centropogon comosus Gleason/HUTPL 11342CampanulaceaeMotepelaWash insect bites (oral testimony)EtOH-H2O (7:3) (leaves)10094
20107
Cestrum sendtnerianum C. Mart./PPN-so-003SolanaceaeSauco negroSelected compoundEtOH-H2O (7:3) (flowers)1006
2047
Purgative, head pain, stomach pain, fever, gangrene, influenza, internal infections, rheumatism, cough [6]MeOH extract (leaves and flowers)100149
20115
Clusia alata Triana & Planch/HUTPL5081ClusiaceaeDucoGastritis [28]MeOH extract (fruits)1009
2085
Gallesia integrifolia (Spreng.) Harms/PPN-ph-001PhytolaccaceaePalo de ajoArthritis, strokes, rheumatism [6]MeOH extract (bark)1007
2088
MeOH extract (leaves)100106
20107
Gaiadendron punctatum (Ruiz & Pav.) G. Don/PPN-lo-001LoranthaceaeVioleta de cerro, violeta de campoStrong cough [25]EtOH extract (flowers)10028
2073
Selected compoundEtOH extract (leaves)1005
2022
Gaultheria erecta Vent/PPN-er-008EricaceaeMote pelaThe fruits are edible [25]EtOH-H2O (7:3) (flowers)10014
2078
Huperzia weberbaueri (Hieron. & Herter ex Nessel) Holub/PPNIc-07LycopodiaceaeWaminga sucaPurgative and to treat supernatural diseases [31]Hexane extract (aerial part)10075
2093
MeOH extract (aerial part)10058
20100
Hesperomeles ferruginea (Pers.) Benth./HUTPL4010RosaceaeQuiqueThe fruits can be used as foods [29]EtOH-H2O (7:3) (fruits)10073
20100
EtOH-H2O (7:3) (leaves)10017
20100
Ilex guayusa Loes./PPN-aq-001AquifoliaceaeGuayusaGastritis, relaxant, increasing woman’s fertility [6]EtOH-H2O (7:3) (leaves)1005
2092
Iresine herbstii Hook./PPN-am-001AmaranthaceaeEscancelFever, relaxant, kidney [6]Lyophilized aqueous (leaves and stems)1008
2078
EtOH-H2O (7:3) (leaves and stems)1005
2093
Lupinus semperflorens Hartw. ex Benth./HUTPL4786FabaceaeChocho silvestre, taure de cerro, aspa chochoFever and stomach painMeOH extract (leaves and stems)10056
20127
Salvia pichinchensis Benth/PPN-la-014LamiaceaeMatico negro, matico grande de cerroTo treat the infection of external wounds and for curing kidney and liver disorders [9]EtOH-H2O (7:3) (leaves and stems)1009
20122
Myrcianthes fragrans (Sw.) McVaugh/PPN-my-008MyrtaceaeArrayán aromático, saco, wawall (kichwa)Selected compoundMeOH extract (leaves)1006
2063
Oreopanax ecuadorensis Seem./PPN-ar-001AraliaceaePumamaquiHeadache [6]MeOH extract (leaves)10087
20100
Oreopanax eriocephalus Harms/HUTPL 4901AraliaceaeMaqui-maquiAnti-inflammatory and antibacterial properties [9]MeOH extract (leaves and flowers)10087
2096
Otholobium mexicanum (L. f.) J.W. Grimes/PPN-fa-005FabaceaeCulén, teculénStomach pain, diarrhea, indigestions, contraceptive [6]EtOAc extract (leaves and flowers)1004
2093
Phyla strigulosa (M. Martens & Galeotti) Moldenke/MT-KN-111VerbenaceaeBuscapina, novalgina Selected compoundEtOAc extract1006
2061
Selected compoundHexane extract (leaves and flowers)1007
2015
Stomachache [29], cramps, diarrhea in children, and intestinal infections; to act as tonicLyophilized aqueous leaves and flowers extract10013
20102
Selected compoundMeOH extract (leaves and flowers)1006
2038
Cestrum racemosum Ruiz & Pav./PPN-so-010SolanaceaeSauco, sauco de montaña, sauco blancoTooth decay, headache, stomach pain, fever, gastritis [6]MeOH extract (leaves and stem)10024
2091
Stereocaulon ramulosum (Sw.) Raeusch./MUTPL-AB-0650StereocaulaceaeMusgoExternal infections, antibiotic [32]EtOAc extract (aerial part)1007
2071
Selected compoundMeCl2 (aerial part)1007
2061
Echinopsis pachanoi (Britton & Rose) Friedrich & G.D. Rowley/PPN-cb-001CactaceaeSan Pedro cactus with 5 ribs/San pedrilloTo induce visions (oral and inhaled administration), to act as a purgative, to treat supernatural diseases, to treat anxiety, and serve as an anti-inflammatory or wound disinfectant [27]Lyophilized from the aqueous extract 10041
2094
San Pedro cactus with 7 ribs/San pedrilloLyophilized from the aqueous extract 1007
2068
San Pedro cactus with 9 ribs/San pedrilloLyophilized from the aqueous extract 10019
2066
Table 2. IC50 of the extracts selected from the screening. The toxicity of each compound is measured as described in the Materials and Methods section. IC50 is defined as the concentration deduced from these data as the concentration that leaves 50% of cells alive. Identification, family, common name of the plant, and extraction mode are also reported.
Table 2. IC50 of the extracts selected from the screening. The toxicity of each compound is measured as described in the Materials and Methods section. IC50 is defined as the concentration deduced from these data as the concentration that leaves 50% of cells alive. Identification, family, common name of the plant, and extraction mode are also reported.
New LabelPlant IdentityCommon NameExtraction ModeIC50 vs. MCF7 Cell Line (µg/mL)Error Bar (µg/mL)Error Bar (%)
1Cestrum sendtnerianum C. Mart. Sauco negroEtOH-H2O
(70:30)
36.803.629.82
2Croton lechleri Müll. Arg.Sangre de dragoLatex5.630.000.00
3Gaiadendron punctatum (Ruiz & Pav.) G. DonVioleta de campo, violeta de cerroEtOH 15.620.201.28
4Garcinia macrophylla Mart.ShoraMeOH36.721.203.28
5Huperzia columnaris B. Øllg.Waminga osoAlkaloid fraction27.351.816.62
6Huperzia kuesteri (Nessel) B. Øllg.Waminga verde grandeEtOAc5.393.2359.80
7Hernandulcin ((6S)-6-[(2S)-2-hydroxy-6-methylhept-5-en-2-yl]-3-methylcyclohex-2-en-1-one)-Pub Chem CID: 12560829.954.6415.50
8Myrcianthes fragrans (Sw.) McVaughArrayán aromático, saco, wawallMeOH36.025.6215.60
9Phyla strigulosa (M. Martens & Galeotti) MoldenkeNovalgina,
buscapina
EtOAc39.531.032.59
10Hexane10.270.232.21
11MeOH28.835.6619.64
12Pinostrobin ((2s)-5-hydroxy-7-methoxyflavone)-Pub Chem CID:7320123.853.8216.01
13Piper ecuadorense SodiroMatico grande, tiklilin grande, matico del monteEtOH-H2O
(70:30)
79.822.322.90
14MeOH30.6921.8871.28
15Piper pseudochurumayu (Kunth) C. DC.Matico, ámbar ámbarMeOH21.391.145.31
16Stereocaulon ramulosum (Sw.) Raeusch.MusgoMeCl219.263.2616.91
Table 3. Proportion of each cell cycle phase of the MCF7 cell line treated for 72 h with 25 µg/mL of each extract dissolved in RPMI, 10% FCS.
Table 3. Proportion of each cell cycle phase of the MCF7 cell line treated for 72 h with 25 µg/mL of each extract dissolved in RPMI, 10% FCS.
Extract From:LabelG0/G1SG2/MApoptosis
RPMI63.016.220.90.6
DMSO (1%)76.410.711.51.8
Cestrum sendtnerianum (flower)174.412.112.51.7
Croton lechleri273.012.913.41.5
G. punctatum (levaes)372.812.414.30.9
Garcinia macrophylla482.67.95.93.8
H. columnaris583.27.87.42.0
H. kuesteri664.415.39.812.7
Hernandulcin775.511.96.29.4
Myrcianthes fragrans866.916.37.512.6
Phyla strigulosa973.013.18.28.1
Phyla strigulosa1070.014.410.76.8
Phyla strigulosa1170.615.48.88.1
Pinostrobin1269.814.56.411.8
Piper ecuadorense1369.015.07.512.0
Piper ecuadorense1472.413.87.88.6
Piper pseudochurumayu1569.714.29.59.1
Stereocaulon ramulosum1672.513.29.17.4
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

Bec, N.; Larroque, C.; Armijos, C. Exploring Southern Ecuador’s Traditional Medicine: Biological Screening of Plant Extracts and Metabolites. Plants 2024, 13, 1422. https://doi.org/10.3390/plants13101422

AMA Style

Bec N, Larroque C, Armijos C. Exploring Southern Ecuador’s Traditional Medicine: Biological Screening of Plant Extracts and Metabolites. Plants. 2024; 13(10):1422. https://doi.org/10.3390/plants13101422

Chicago/Turabian Style

Bec, Nicole, Christian Larroque, and Chabaco Armijos. 2024. "Exploring Southern Ecuador’s Traditional Medicine: Biological Screening of Plant Extracts and Metabolites" Plants 13, no. 10: 1422. https://doi.org/10.3390/plants13101422

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