Next Article in Journal
DNA Sequence Analysis of an Inversion Hot Spot in Lobeliaceae Plastomes
Next Article in Special Issue
Biotechnological Intervention and Secondary Metabolite Production in Centella asiatica L.
Previous Article in Journal
Assessing the Impact of Geographical Distribution and Genetic Diversity on Metabolic Profiles of a Medicinal Plant, Embelia ribes Burm. f.
Previous Article in Special Issue
Anacardium occidentale Bark as an Antidiabetic Agent
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

In Vitro Cytotoxic Activity of Methanol Extracts of Selected Medicinal Plants Traditionally Used in Mexico against Human Hepatocellular Carcinoma

by
Joel H. Elizondo-Luévano
1,
Ricardo Gomez-Flores
2,*,
María J. Verde-Star
1,
Patricia Tamez-Guerra
2,
César I. Romo-Sáenz
2,
Abelardo Chávez-Montes
1,
Nancy E. Rodríguez-Garza
1 and
Ramiro Quintanilla-Licea
1,*
1
Departamento de Química, Facultad de Ciencias Biológicas (FCB), Universidad Autónoma de Nuevo León (UANL), San Nicolás de los Garza, N.L., Monterrey 66455, Mexico
2
Departamento de Microbiología e Inmunología, Facultad de Ciencias Biológicas (FCB), Universidad Autónoma de Nuevo León (UANL), San Nicolás de los Garza, N.L., Monterrey 66455, Mexico
*
Authors to whom correspondence should be addressed.
Plants 2022, 11(21), 2862; https://doi.org/10.3390/plants11212862
Submission received: 14 August 2022 / Revised: 24 October 2022 / Accepted: 25 October 2022 / Published: 27 October 2022
(This article belongs to the Special Issue Medicinal Plant Extracts)

Abstract

:
Medicinal plants are traditionally used in Mexico to treat diseases such as cancer. The present study aimed to evaluate the cytotoxic, antioxidant, and anti-hemolytic activity of 15 plants of ethnopharmacological use in Mexico. For this, plant methanol extracts were prepared by the Soxhlet method, after which their cytotoxic activity was evaluated against human hepatocellular carcinoma (HEP-G2) and monkey kidney epithelial (Vero) cells by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) reduction colorimetric assay. The selectivity index (SI) of each extract was then determined by the IC50 ratio of normal to tumor cells. We showed that Ruta chalepensis extract possessed an IC50 of 1.79 µg/mL and 522.08 µg/mL against HEP-G2 and Vero cells, respectively, resulting in an SI of 291.50. Furthermore, antioxidant activity was evaluated by the 1,1-diphenyl-2-picrylhydrazyl (DPPH) radical scavenging technique, where the best antioxidant potential was shown by the Heterotheca inuloides extract (IC50 = 19.24 µg/mL). Furthermore, the hemolytic potential was determined against human erythrocytes, which showed that the extracts with the highest anti-hemolytic activity were Smilax aspera (IC50 = 4.41 µg/mL) and Amphipterygium adstringens (IC50 = 5.35 µg/mL). In conclusion, we observed that R. chalepensis methanol extract possesses cytotoxic activity against HEP-G2 cells, without affecting non-tumorigenic Vero cells. Our results indicated the antitumor potential of medicinal plants used in Mexico.

1. Introduction

According to the World Health Organization, cancer is one of the leading causes of death worldwide, with 19.3 million cases and 10 million deaths in 2020 [1]. Among the most critical cancers, hepatocellular carcinoma (HCC) is the most frequent primary liver neoplasm globally, requiring a multidisciplinary approach for diagnosis, treatment, and prevention. This pathology is one of the major causes of cancer-related deaths globally [2]. HCC prognosis is complex because of the competing risk imposed by underlying cirrhosis and the presence of malignancy. For this reason, there is a constant search for better diagnostic, screening, and therapy strategies to improve the prognosis and treatment of this neoplasm [3]. Identification of serum biomarkers is currently used in predictive HCC, since some of them are essential as screening tools for therapy selection, surveillance, diagnosis, prognosis, and post-treatment evaluation [4].
Ultrasound surveillance allows diagnosis at early stages, when the tumor may be curable by resection, liver transplantation, or ablation, and a 5-year survival of more than 50% can be achieved.[5]. Liver transplantation may be the most useful for people who are not candidates for resection. However, shortage of donors limits its applicability [6]. In addition, the extreme toxicity of most anticancer drugs prompts the search for new drugs that may prevent or slow cancer growth with less toxicity and more safety [7]. Therefore, developing new, safe, and more specific biological targets is essential, especially for the most aggressive tumors [1].
Humans have been using plants as a source of medicine since ancient times, and several modern drugs are derived from medicinal plants [8,9]. Alternative drugs are conventionally applied in many types of cancers to reduce chemotherapy’s toxicity and adverse effects. A variety of phytonutrients found in fruits, herbs, and spices act as potent cancer prevention agents by preventing the overproduction of toxic chemicals in the body [7]. It is well recognized that more than 70% of anticancer drugs have a natural origin, and more than 3000 plants with antitumor properties have been widely studied [10]. The scientific community has shown that cancer patients often use alternative therapies, particularly medicinal plants, which have served as the basis for discovering new anticancer drugs [11]. Among the most significant examples of plant-derived anticancer agents are vincristine, which has activity against leukemia, Hodgkin’s and non-Hodgkin’s lymphoma, and solid cancers such as breast and lung cancer, vinblastine with activity against breast, renal, and lymphoma cancer, docetaxel against breast and lung cancer, and paclitaxel against ovarian, breast, lung, and bladder cancer [12].
Plants are a source of bioactive molecules with antibacterial, antiparasitic, healing, analgesic, anti-inflammatory, and tissue repairing properties, representing a potential alternative in cancer treatment [13,14,15]. Approximately 80% of developing countries’ world population adopts plants to treat various ailments [16]. Mexico has used medicinal plants since pre-Hispanic times and is considered one of the leading countries with broad plant diversity. About 30% of Mexican patients diagnosed with cancer use plants as complementary medicine [17].
In the last two decades, research concerning natural medicinal products has increased due to the discovery of new molecules with pharmaceutical interest based on ethnopharmacological knowledge. However, more than 90% of plant species have not yet been studied [18]. The present study aimed to evaluate the in vitro antitumor, antioxidant, and anti-hemolytic activities of methanol extracts of Amphipterygium adstringens, Argemone mexicana, Artemisia ludoviciana, Cymbopogon citratus, Heterotheca inuloides, Jatropha dioica, Justicia spicigera, Larrea tridentata, Mimosa tenuiflora, Psacalium peltatum, Pseudognaphalium obtusifolium, Ruta chalepensis, Semialarium mexicanum, Smilax aspera, and Tagetes lucida. These plants were selected based on ethnomedical reports showing their use in different regions of Mexico, particularly against cancer [10].
A. adstringens possesses cytotoxic activity against prostate adenocarcinoma (PC-3), ovarian adenocarcinoma (OVCAR-3), and colon adenocarcinoma (HT-29) cell lines, whereas A. mexicana is used to treat stomach problems, intestinal infections, and parasites [19], and in vitro, it inhibits lung (A549), colon (HT-2), and human larynx (HLaC) cell lines growth [20]. In addition, A. ludoviciana is used in combination with antibiotics to treat tuberculosis [21] but has not been reported against cancer. Furthermore, C. citratus essential oil has shown cytotoxic activity against HT-29 and Caco-2 human colorectal adenocarcinoma cell lines [22], whereas H. inuloides has traditionally been used to treat a wide range of diseases in Mexico, such as rheumatism, inflammation, and muscle pain, as well as dental diseases and gastrointestinal disorders [23]. J. dioica extracts have been evaluated in vitro in mouse peripheral blood cells, mouse fibroblasts, human hepatocytes, and pig epithelial cells with low or nil toxicity [24] but their antitumor potential has not yet been investigated. In addition, J. spicigera is traditionally used in Mexico to treat dysentery, diabetes, leukemia, and anemia, and its ethanol extract, as well as that of T. lucida, showed cytotoxic effect against human adenocarcinoma HeLa cells [25], whereas L. tridentata is recognized for its various traditional uses [26]. Nordihydroguaiaretic acid, one of L. tridentata most studied secondary metabolites, has shown biological activity against reactive oxygen species, activation of the endogenous antioxidant response mediated by nuclear factor erythroid 2, and inhibition of lipoxygenases [27].
In addition, M. tenuiflora is popularly used because of its antimicrobial and antifungal properties due to its flavonoids and tannins [28]. Plants of the Asteraceae family, such as P. peltatum and P. obtusifolium, possess sesquiterpenes such as cacalol and cacalone that increase insulin levels and keep hypoglycemic, anti-inflammatory, and antioxidant activities [29]. R. chalepensis is one of the most widely used plants in Mexico and worldwide, whose phytochemical compounds have shown antiparasitic, antifungal, antioxidant, antibacterial, and cytotoxic activities [30]. In addition, the root bark of S. mexicanum was reported to possess a cytotoxic effect against the human breast cancer cell line MCF-7, with promising results [31]. Studies on the chemical components of Smilax species such as S. aspera have revealed the presence of steroidal saponins, flavonoids, phenylpropanoids, and stilbenoids, with antifungal, anti-inflammatory, and cytotoxic activities against different cell lines [32].
In this investigation, we evaluated the cytotoxic action of plant extracts against the human HCC cell line HEP-G2, as compared with that on normal monkey kidney epithelial cells (Vero cells) to determine cancer selectivity. HCC HEP-G2 cells are widely used in cytotoxicity studies because they are easy to maintain in culture [33] and they preserve hepatic functions similar to those of primary human hepatocytes [34]. They are useful to evaluate regulation of enzymes that metabolize pharmacological agents [35]. One of the advantages of using the Vero cell line is that it indefinitely multiplies, whereas primary liver cells have a limited lifespan and die after a number of generations; therefore, the Vero cell line is easy to handle and represents an unlimited source of self-replication with a relatively high degree of homogeneity [36].
Therefore, based on epidemiological data and empirical knowledge, these medicinal plants, commonly used by ethnic groups in Mexico, were studied to validate their therapeutic properties. This investigation contributes to the knowledge and dissemination of traditional Mexican medicine and provides information for further studies of bioactive molecules present in plants that may be useful in treating human hepatocarcinoma.

2. Results

2.1. Plant Identification

Table 1 shows plants used in the present study, which were identified by the FCB taxonomist with their corresponding identification voucher numbers, the family to which they belong, their common name, the parts used to perform the extractions, and the extraction yields obtained for each extract. These plants were selected based on empirical or scientific knowledge of their use in Mexico. The plant with the highest extraction yield was A. adstringens, with 38.36%, and the plant with the lowest extraction yield was S. mexicanum, with an extraction percentage of 10.52.

2.2. Cytotoxic Activity

Table 2 shows the cytotoxicity results of the extracts against tumor and normal cells and the SI obtained for each extract. HEP-G2 cells were compared against Vero cells since these are adherent cells. The extracts with the highest cytotoxic activity against the HEP-G2 cell line were those of J. spicigera (IC50 = 2.92 µg/mL) and R. chalepensis (IC50 = 1.79 µg/mL), which statistically presented similar activity, followed by J. dioica (IC50 = 12.34 µg/mL) and A. adstringens (IC50 = 41.77 µg/mL). The other extracts presented IC50 greater than 350 µg/mL against HEP-G2 cells. Vincristine positive control showed cytotoxicity percentages of 11.27 and 19.65 against Vero and HEP-G2 cell lines, respectively, after 48 h incubation. Extracts with SIs higher than three were classified as promising. This value indicates that the extract is three times more cytotoxic for the tumor cell line, compared with the normal cell line [37]. The highest SI’s against HEP-G2 cells corresponded to R. chalepensis, J. spicigera, J. dioica, and A. adstringens extracts, with an SI of 291.50, 18.84, 5.72, and 4.74, respectively. R. chalepensis extract showed the highest SI. The selectivity behavior of extracts provides information on being selectively cytotoxic to cancer cells and less cytotoxic to normal cells. Extracts with this behavior will be considered suitable for the bidirected isolation of their phytocompounds.

2.3. Antioxidant Activity

The extract with the highest antioxidant activity was H. inuloides, with an IC50 = 19.24 µg/mL. The other extracts showed IC50 above 500 µg/mL (Table 3). The extract with the lowest antioxidant activity was S. mexicanum, with an IC50 = 1639.79 µg/mL. However, none of the extracts was significantly better than the positive control.

2.4. Hemolytic and Anti-Hemolytic Activity

The in vitro hemolytic test was commonly used in the pharmaceutical industry to screen therapies throughout the early stages of clinical development [38]. In the tests of the hemolytic activity of the methanol extracts against human erythrocytes, it was found that the extract with the highest hemolytic activity was A. adstringens (IC50 = 203.62 µg/mL), followed by A. adstringens (IC50 = 545.74 µg/mL). However, the other extracts resulted in IC50 above 600 µg/mL. Specifically, the extracts corresponding to J. spicigera, M. tenuiflora, P. peltatum, and P. obtusifolium were the least hemolytic with IC50 above 2500 µg/mL (Table 4).
The AAPH radical-induced hemolysis protection assay, which causes lipid peroxidation of normal red blood cells [39], and is considered an ex vivo model to demonstrate the high antioxidant potential of some plant species [40]. When the anti-hemolytic activity was evaluated, it was found that the extracts with the highest anti-hemolytic activity were S. aspera (IC50 = 4.41 µg/mL), A. adstringens (IC50 = 5.35 µg/mL), H. inuloides (IC50 = 5.42 µg/mL), and P. peltatum (IC50 = 5.92 µg/mL), which statistically behaved in the same manner (Table 4). The extract with the lowest anti-hemolytic activity was L. tridentata (IC50 = 777.85 µg/mL).

3. Discussion

Cancer is the second leading cause of death in Mexico. Most cases are detected in advanced stages, and the Mexican population commonly applies natural therapies. More than 300 complementary and alternative medicine treatments are currently used in Mexican folklore [17]. Some studies have shown that plants of the Anacardiaceae [41], Papaveraceae [42], Compositae [43], Euphorbiaceae [44], Acanthaceae [45], and Rutaceae [30] families have antioxidant, anti-inflammatory, antiparasitic, antimicrobial, and anticancer properties [46,47,48]. The search for new compounds with antitumor potential represents a priority in the pharmaceutical area due to new variants resistant to conventional antineoplasic drugs [10].
Medicinal plants produce bioactive compounds with various biological functions that possess a high content of flavonoids, coumarins, carbohydrates, tannins, sterols, quinones, and alkaloids, as observed in R. chalepensis, J. spicigera, J. dioica, and A. adstringens, which showed high selectivity against tumor cells. These compounds may be an effective alternative for new anticancer molecules [49]. In our study, the species showing in vitro antineoplasic effects against HEP-G2 cells were R. chalepensis (SI = 291.50), J. spicigera (SI = 18.84), J. dioica (SI = 5.72), and A. adstringens (SI = 4.74). When the SI value of plant extracts is greater than two, it is accepted that those have a selective toxic effect on cancer cells; on the other hand, an SI value of less than two shows low toxicity on cancer cells [50]; therefore, plant extracts with an SI less than two were considered to have no cytotoxic activity against HEP-G2 cells.
Our study demonstrated that R. chalepensis was cytotoxic against the human tumor cell line HEP-G2 and showed the highest SI compared with the other evaluated plants (Table 2). Alkaloids and furanocoumarins are among the primary metabolites identified in plants from the Rutaceae family [51,52]. These metabolites may be responsible for the cytotoxic activity we observed, since several studies evidenced the cytotoxic effect of R. chalepensis extracts against HeLa, MCF-7, and human squamous cell carcinoma (A431) cell lines were due to furanoacridones and acridone alkaloids (arborinin and evoxanthin) [53,54]. R. chalepensis has also been shown to possess antioxidant and hypoglycemic properties, protecting against lipid peroxidation through inhibiting α-amylase and α-glucosidase enzymes [52,55].
J. spicigera showed an SI index of 18.84 when evaluated in Vero and HEP-G2 cells, since it did not show significant toxicity in Vero cells (IC50 = 54.91 μg/mL) compared with HEP-G2 cells (IC50 = 2.92 μg/mL). Thus, we may consider it an excellent candidate to investigate against other tumor cell lines. Its cytotoxic activity on human breast cancer cell lines T47D, MCF-7, and HeLa cells with IC50 < 30 μg/mL has been previously reported [25,56]. Glycosides of kaempferol, such as kaempferitrin, were isolated from this plant [57,58] and identified as the primary metabolite that exerted anti-oxidant, anti-inflammatory, proapoptotic, cardio-protective, and anti-cancer activities [59].
The root of J. dioica is popular in Mexican herbal medicine to treat various diseases. It has also shown cytotoxic, antimicrobial, and antiviral activities. Components found in J. dioica are mainly diterpenes, such as citlalitrione, riolozane, 6-epiriolozatrione, citlalitrione, and jatrophatrione, and riolozatrione [60,61,62]. When we evaluated the cytotoxic activity of J. dioica, we found that the methanol extract has potent activity against HEP-G2 (IC50 = 12.34 μg/mL) cells. We also determined that the extract of J. dioica showed an SI of 5.72 (Table 2). Diterpenes are probably responsible for the cytotoxic activity since abietane diterpenoids have demonstrated in vitro cytotoxic activity against HL-60 and A549 cell lines and high antioxidant activity against Trolox [63]. In addition to the cytotoxic activity of the diterpene lriolozatrione, it possesses antiviral activity against HSV-1 and HSV-2 [64]. We also determined its antihemolytic potential against the AAPH radical, making it a plant of interest for future studies.
A. adstringens extract showed cytotoxic activity against HEP-G2 (IC50 = 41.77 µg/mL) with an SI of 4.74 against Vero cells. A. adstringens is traditionally used to treat gastric ulcers and cancer. This plant is rich in anacardic acids, 6-pentadecyl acid being the most abundant, which has been determined to be cytotoxic against the human colorectal adenocarcinoma Caco-2 [65], the human gastric adenocarcinoma AGS, and the chronic myeloid leukemia K562 [66], without affecting human PBMC [66]. In another study, the antiproliferative effect of the methanol extract of A. adstringens bark against the human ovarian adenocarcinoma OVCAR-3, the melanoma UACC-62, the colorectal adenocarcinoma HT-29, the prostate adenocarcinoma PC-3, the glioblastoma multiforme U251, the lung carcinoma NCI-H460, and the renal adenocarcinoma 786-O was determined, which ranged from 4.4 µg/mL to 28.0 µg/mL [67]. Such results agreed with our study, indicating the potential activity of A. adstringens extract against different cancer cells and showing high anti-hemolytic activity. Hence, it is essential to continue with studies of this plant, including its mechanisms. Furthermore, A. mexicana antitumor activity against MCF-7 and MDA-MB-231 breast cancer cell lines through the induction of the Estrogen receptor alpha (Erα) gene has been previously reported [68]. This plant is rich in alkaloids, which have significantly reduced cytotoxicity to the human colorectal adenocarcinoma cell line SW480 [69]. The other methanol extracts had cytotoxic activity against Vero cells without significantly affecting HEP-G2 cells. These extracts had SIs from 0.02 to 1.48. However, some of these plants have been ethnopharmacologically used to treat other types of cancer cell lines, such as HT29, K-562, HCT-15, SW-480, and MCF-7 cells [10,11]. However, our results showed that those plants did not have cytotoxic activity against HEP-G2 cells.
Regarding the antioxidant activity related to the DPPH free radical scavenging effect, the samples with an IC50 ≤ 50 µg/mL were considered to have a relevant antioxidant activity [70]. Some natural components such as phenols, alkaloids, and terpenoids in medicinal plants possess antioxidant activity and are considered necessary because of their numerous health benefits, as these molecules can scavenge free radicals [40]. Although the in vitro evaluation of free radicals is useful to demonstrate the antioxidant potential of a sample, we suggest the need to show the effects in a more complex physiological system to confirm the antioxidant potential of natural extracts in an ex vivo model, such as the assay for protection against AAPH radical-induced hemolysis in human erythrocytes [70,71].
Only H. inuloides extract showed significant antioxidant and anti-hemolytic activity compared with the other evaluated extracts, showing IC50 of 19.24 μg/mL and 5.42 μg/mL, respectively. These data agree with a study conducted in 2010, showing the antioxidant activity of its methanol extract [72]. However, this extract did not have a similar effect than the positive control, which had an IC50 = 7.23 μg/mL. The other extracts showed IC50 > than 500 µg/mL. Therefore, they were not considered to have high antioxidant activity. H. inuloides has been traditionally used to treat a wide range of diseases in Mexico, such as rheumatism, topical skin inflammation, and muscle pain, among other conditions associated with inflammatory processes or pain. Reports regarding the toxicity of H. inuloides are limited to date [23]. On the other hand, we found anti-hemolytic activity and hemolytic potential with IC50 of 28.29 μg/mL and 738.73 μg/mL, respectively, for R. chalepiensis. However, we did not find an antioxidant activity as effective as ascorbic acid or H. inuloides extract.
We observed the antioxidant activity of J. spicigera extract by the DPPH method with an IC50 of 924.92 μg/mL, whereas Baqueiro-Peña in 2017 [45] determined an IC50 of 880.00 μg/mL. We found only one report regarding the evaluation of J. spicigera extracts on human erythrocytes by the AAPH method, where they reported an antihemolytic activity with an IC50 = 13.5 µg/mL. However, they performed an extraction with a solvent mixture of dichloromethane:methanol (1:1), which may have resulted in a different outcome than ours. Therefore, this is the first report regarding the antihemolytic activity of J. spicigera methanol extract with an IC50 of 81.08 μg/mL. In addition, they reported antioxidant activity with IC50 = 35.8 µg/mL, as determined by the DPPH method.
The antioxidant (IC50 = 312.50 μg/mL) and anti-hemolytic activity of A. mexicana methanol extract at 400 μg/mL has been reported [20]. However, berberine, one of its main components, was observed to have an antioxidant effect with IC50 = 168.18 μg/mL but had a deficient anti-hemolytic activity, which decreased as the concentration increased [47]. In our study, we determined that A. mexicana extract possesses DPPH uptake potential with IC50 = 565.98 μg/mL and a low hemolytic activity (IC50 = 973.88 μg/mL). This difference may be due to the concentration of secondary metabolites in the extract or by the area and date of collection of the plant since these factors may cause diversity in the concentration of metabolites in the plant [73].
The inhibitory activity of the extracts against AAPH radicals was related to the most effective extracts of A. adstringens (IC50 = 5.35 ± 1.63 µg/mL), H. inuloides (IC50 = 5.42 ± 0.89 µg/mL), P. peltatum (IC50 =5.92 ± 1.29 µg/mL), and S. aspera (IC50 = 4.41 ± 0.69 µg/mL) since they did not present statistically significant differences. The extract of L. tridentata (IC50 = 777.85 ± 18.58 µg/mL) showed the lowest antihemolytic activity. The protective role against hemolysis by AAPH has been attributed to the polyphenol content of plants because they interact with erythrocyte membrane components by preventing oxidation of membrane proteins and lipids via hydrogen bonds [71].

4. Materials and Methods

4.1. Chemicals and Reagents

All chemicals and solvents were of analytical grade. Dulbecco’s modified Eagle medium (DMEM), 1% antibiotic/antimycotic solution, and fetal bovine serum (FBS) (Gibco BRL, Grand Island, NY, USA) were used. Dimethyl sulfoxide (DMSO), 2,2′-azobis (2-methylpropionamidine) dihydrochloride (AAPH), 2,2-diphenyl-1-picrylhydrazyl (DPPH), methyl alcohol (MeOH), 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), ferric chloride, sodium hydroxide (NaOH), and sulfuric acid were obtained from Sigma-Aldrich (St. Louis, MO, USA). Vincristine sulfate (VS) salt was purchased from Hospira Inc. (Lake Forest, IL, USA).

4.2. Plant Material and Extraction

Plants used in this study were identified by Professor Dr. Marco A. Guzman Lucio, Chief of the Herbarium of FCB at UANL, Nuevo León, México, who was assigned a voucher number to each specimen (Table 1). Specimens were deposited in the FCB-UANL herbarium. The names and botanical families of species were taxonomically validated using WorldFlora Online, http://www.worldfloraonline.org (accessed on 1 August 2022). Plant extraction was performed by subjecting 60 g of milled dry plant material to 600 mL of absolute MeOH in Soxhlet equipment for 48 h [51]. Extracts were then filtered, rotaevaporated, and stored at 4 °C in amber bottles until use. The following Formula (1) was used to calculate the extraction yield percentage [13]:
%   Yield = Final   weight Initial   weight × 100

4.3. Tumor and Normal Cells

Human hepatocellular carcinoma (HEP-G2; ATCC HB-8065) and monkey kidney epithelial cells (Vero; ATCC CCL-81) were grown in DMEM supplemented with 10% heat-inactivated FBS and 1% antibiotic/antifungal solution (referred as a complete DMEM culture medium) [74]. Cells were cultured at 37 °C in an atmosphere of 5% CO2 and 95% air. Before evaluating the extracts, cells were incubated for 24 h for adaptation. To test the cytotoxicity of plants, cells were treated with methanol extracts at concentrations ranging from 31.25 µg/mL to 1000 µg/mL of each extract in a final 200 µL for 48 h. The positive control consisted of 0.05 µg/mL of VS, whereas the negative control was culture medium alone. A colorimetric assay determined cell viability by adding 15 µL of MTT/well (final concentration of 500 µg/mL) and incubating for three hours. Formazan crystals were dissolved with 80 µL/well of DMSO, and optical densities (OD) were measured at 570 nm on a MULTISKAN GO microplate reader (Thermo Fisher Scientific, Waltham, MA, USA). Cell viability was determined using the following Formula (2):
%   Cell   viability = OD   Treatment OD   Negative   control × 100
Finally, the selectivity index (SI) was determined for each extract evaluated. SI are used to assess cytotoxic potential relative to toxicity to normal cells, where high SI indicates high potency and low cellular toxicity [75]. They were obtained for each extract evaluated after dividing the IC50 (half maximal inhibitory concentration) by the specific IC50 against HEP-G2 tumor cells after 72 h incubation [76]. According to Bezivin et al. (2003), the selectivity index is interesting in the case of values greater than three [37]. Thus, in our study, any sample that has an SI value greater than three was considered to have high selectivity. SI was calculated using the following Formula (3):
SI = IC 50   Normal   Cells   Value IC 50   Tumor   Cells   Value

4.4. Antioxidant Activity

The antioxidant potential of 31.25 µg/mL to 500 µg/mL plant extracts was tested by the DPPH radical scavenging assay [77]. For this, we incubated 100 μL of 125 μM DPPH to 100 μL of each sample for 30 min at 37 °C in darkness. ODs at 517 nm were then measured in a spectrophotometer Genesys 20 (Thermo-Fisher Scientific, Waltham, MA, USA), using 50 µM ascorbic acid as a positive control (C+) and MeOH as a blank. The reduction percentage was calculated using the following Formula (4):
%   DPPH   reduction = OD   Treatment OD   Blank × 100
IC50 values were calculated as the concentration of sample required to scavenge 50% of DPPH.

4.5. Hemolytic Activity Test

The in vitro hemoglobin denaturation and hemolysis test is recommended and used for a battery of tests aimed at checking the toxicity of different products in the pharmaceutical industry, which are useful to examine therapies throughout the early stages of clinical development [78]. The hemolytic activity of plant extracts was evaluated by the hemolysis test [79]. Ten milliliters of human blood from healthy donors was collected by venipuncture in tubes with anticoagulant (EDTA), after which blood was centrifuged to separate erythrocytes from plasma at 1500 rpm (15 min at 25 °C) and washed three times with 10 mL of PBS (pH 7.4). Erythrocytes were then suspended at 5% v/v in PBS for the tests. For the hemolysis assay, the erythrocytes suspension was mixed with the extracts dissolved in PBS at concentrations ranging from 100 μg/mL to 1000 μg/mL in 2 mL Eppendorf (Eppendorf® AG, Hamburg, Germany) microcentrifuge tubes for 30 min at 37 °C, protected from light. After this, treatments were centrifuged at 13,000 rpm (3 min at 4 °C) and 200 μL of supernatant fluids were taken and placed on a transparent plastic 96 flat-bottomed wells microplate. The degree of hemolysis was then determined by measuring ODs at 540 nm, whereas the IC50 values were calculated as the concentration of sample required to hemolyze 50% of human red blood cells. The percentage of hemolysis was calculated with the following Formula (5):
%   Hemolysis = OD   Treatment OD   Positive   control × 100

4.6. Anti-Hemolytic Activity Test

The inhibitory property of many antioxidants on AAPH radical-induced hemolysis has been widely studied and documented [47,77,80,81]. Hemolysis of erythrocytes is commonly used as an ex vivo model in the study of the alteration of cell membranes induced by ROS, since the thermolysis of the azoic compound AAPH generates a constant flow of peroxyl radicals at 37 °C, causing hemolysis of erythrocytes [80]. The anti-hemolytic activity was determined by the AAPH inhibition test, as previously reported [81]. For the hemolysis inhibition assay, the same steps were performed as for the hemolysis assay but in this case, the AAPH (150 mM) radical prepared in PBS was used as a hemolysis inducer (positive control). To evaluate the erythrocyte protection effect of the extracts, the previously obtained erythrocyte suspension was incubated in 2 mL Eppendorf microcentrifuge tubes with different concentrations of the extract 100 μg/mL to 1000 μg/mL plus AAPH. The mixture was set at 37 °C for 3 h at 250 rpm and protected from light. Next, the mixture was centrifuged at 13,000 rpm (5 min at 4 °C) and 200 μL of supernatant fluids were taken and placed in a transparent plastic 96 flat-bottomed wells microplate. The percentage of anti-hemolytic activity was calculated with the following Formula (6):
%   Anti hemolytic   Activity = 1 ( OD   Treatment OD   Positive   control   ) × 100

4.7. Statistical Analysis

Statistical analyses were performed using the Graph Pad Prism 6 program (GraphPad Software Inc., San Diego, CA, USA). One-way analysis of variance was used to determine the significant difference between evaluated concentrations. The post-hoc Tukey test was used to determine the difference between treatment means. The Probit test was used to calculate the IC50 values. All data represent means ± SD of triplicate determinations from at least three independent experiments (p < 0.05).

5. Conclusions

The use of medicinal plants demonstrates the biotechnological potential of plants as a source of molecules with cytotoxic activity. Plant methanol extracts used in this study showed promising selectivity indexes, particularly R. chalepensis extract, which demonstrated its cytotoxic effect in vitro, making it a suitable candidate for future research in cancer therapies. Our study provides scientific validation of the antitumor potential of some plants commonly used in Mexico and Latin America. In addition, we showed their hemolytic or anti-hemolytic potential for the first time, suggesting that they may be promising for developing new therapies against cancer, as some of them were not toxic for red cells. Our results may contribute to the safe ethnopharmacological use of medicinal plants, which stimulates research on their bioactive compounds against cancer and the mechanisms associated with their biological activity.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/plants11212862/s1.

Author Contributions

Conceptualization, J.H.E.-L. and R.Q.-L.; methodology, R.G.-F.; software, M.J.V.-S.; validation, C.I.R.-S. and R.Q.-L.; formal analysis, M.J.V.-S.; investigation, N.E.R.-G. and A.C.-M.; resources, R.G.-F. and M.J.V.-S.; data curation, C.I.R.-S. and A.C.-M.; writing—original draft preparation, J.H.E.-L.; writing—review and editing, J.H.E.-L. and R.G.-F.; visualization, J.H.E.-L. and N.E.R.-G.; supervision, R.Q.-L.; project administration, P.T.-G.; funding acquisition, R.G.-F. and P.T.-G. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the Consejo Nacional de Ciencia y Tecnología (CONACYT, Mexico), grant 935405 (CVU: 418935) to J.H.E.L., grant 808132 (CVU: 1006989) to N.E.R.-G., and grant 877783 (CVU: 445572) to C.I.R.-S. R.G.-F., P.T.-G., A.C.-M., and R.Q.-L. was supported by the Programa de Apoyo a la Investigación Científica y Tecnológica (PAICYT-UANL 307-CN-2022).

Institutional Review Board Statement

The laboratory animal study was conducted according to the guidelines of the Declaration of Helsinki and approved by the Ethics Committee of the UANL, registration number CI-09-2022. The letter of Informed Consent for donors of human biological sample material and the Institutional Review Board Approval is attached as supplementary material.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The datasets generated or analyzed during the present study are available from the corresponding author.

Acknowledgments

We would like to thank the crew of the Laboratorio de Inmunología y Virología and Laboratorio de Fitoquímica of FCB-UANL for supporting this research.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Matasar, M.J.; Zelenetz, A.D. Overview of lymphoma diagnosis and management. Radiol. Clin. N. Am. 2008, 46, 175–198, vii. [Google Scholar] [CrossRef] [PubMed]
  3. Kulik, L.; El-Serag, H.B. Epidemiology and Management of Hepatocellular Carcinoma. Gastroenterology 2019, 156, 477–491.e1. [Google Scholar] [CrossRef]
  4. Piñero, F.; Dirchwolf, M.; Pessôa, M.G. Biomarkers in hepatocellular carcinoma: Diagnosis, prognosis and treatment response assessment. Cells 2020, 9, 1370–1396. [Google Scholar] [CrossRef] [PubMed]
  5. Marengo, A.; Rosso, C.; Bugianesi, E. Liver cancer: Connections with obesity, fatty liver, and cirrhosis. Annu. Rev. Med. 2016, 67, 103–117. [Google Scholar] [CrossRef] [PubMed]
  6. Forner, A.; Llovet, J.M.; Bruix, J. Hepatocellular carcinoma. Lancet 2012, 379, 1245–1255. [Google Scholar] [CrossRef]
  7. Radha Abbas Hasoon, M.; Jawad Kadhim, N. Improvement of the selectivity index (SI) and cytotoxicity activity of doxorubicin drug by Panax ginseng plant extract. Arch. Razi Inst. 2021, 76, 659–666. [Google Scholar] [CrossRef] [PubMed]
  8. Zaman, W.; Ye, J.; Ahmad, M.; Saqib, S.; Shinwari, Z.K.; Chen, Z. Phylogenetic exploration of traditional Chinese medicinal plants: A case study on Lamiaceae (angiosperms). Pakistan J. Bot. 2022, 54, 1033–1040. [Google Scholar] [CrossRef]
  9. Zaman, W.; Ahmad, M.; Zafar, M.; Amina, H.; Lubna; Saqib, S.; Ullah, F.; Ayaz, A.; Bahadur, S.; Park, S. Diversity of medicinal plants used as male contraceptives: An initiative towards herbal contraceptives. Indian J. Tradit. Knowl. 2022, 21, 616–624. [Google Scholar]
  10. Alonso-Castro, A.J.; Villarreal, M.L.; Salazar-Olivo, L.A.; Gomez-Sanchez, M.; Dominguez, F.; Garcia-Carranca, A. Mexican medicinal plants used for cancer treatment: Pharmacological, phytochemical and ethnobotanical studies. J. Ethnopharmacol. 2011, 133, 945–972. [Google Scholar] [CrossRef] [PubMed]
  11. Jacobo-Herrera, N.J.; Jacobo-Herrera, F.E.; Zentella-Dehesa, A.; Andrade-Cetto, A.; Heinrich, M.; Pérez-Plasencia, C. Medicinal plants used in Mexican traditional medicine for the treatment of colorectal cancer. J. Ethnopharmacol. 2016, 179, 391–402. [Google Scholar] [CrossRef]
  12. Da Rocha, A.B.; Lopes, R.M.; Schwartsmann, G. Natural products in anticancer therapy. Curr. Opin. Pharmacol. 2001, 1, 364–369. [Google Scholar] [CrossRef]
  13. Cárdenas Garza, G.R.; Elizondo Luévano, J.H.; Bazaldúa Rodríguez, A.F.; Chávez Montes, A.; Pérez Hernández, R.A.; Martínez Delgado, A.J.; López Villarreal, S.M.; Rodríguez Rodríguez, J.; Sánchez Casas, R.M.; Castillo Velázquez, U.; et al. Benefits of cardamom (Elettaria cardamomum (L.) Maton) and turmeric (Curcuma longa L.) extracts for their applications as natural anti-inflammatory adjuvants. Plants 2021, 10, 1908–1924. [Google Scholar] [CrossRef] [PubMed]
  14. Elizondo-Luévano, J.H.; Castro-Ríos, R.; Sánchez-García, E.; Hernández-García, M.E.; Vargas-Villarreal, J.; Rodríguez-Luis, O.E.; Chávez-Montes, A. In vitro study of antiamoebic activity of methanol extracts of Argemone mexicana on trophozoites of Entamoeba histolytica HM1-IMSS. Can. J. Infect. Dis. Med. Microbiol. 2018, 2018, 1–8. [Google Scholar] [CrossRef] [Green Version]
  15. Hernández-Martinez, H.C.; Gómez-Flores, R.; Tamez-Guerra, P.; Quintanilla-Licea, R.; Samaniego-Escamilla, M.A.; Monreal-Cuevas, E.; Tamez-Guerra, R.; Rodriguez-Padilla, C. Antitumor activity of Pachycereus marginatus (DC.) Britton Rose extracts against murine lymphoma L5178Y-R and skin melanoma B16F10 cells. J. Med. Plants Res. 2016, 10, 635–639. [Google Scholar] [CrossRef] [Green Version]
  16. Rai, P.K.; Lalramnghinglova, H. Ethnomedicinal plant resources of Mizoram, India: Implication of traditional knowledge in health care system. Ethnobot. Leafl. 2010, 14, 274–305. [Google Scholar]
  17. Gerson-Cwilich, R.; Serrano-Olvera, A.; Villalobos-Prieto, A. Complementary and alternative medicine (CAM) in Mexican patients with cancer. Clin. Transl. Oncol. 2006, 8, 200–207. [Google Scholar] [CrossRef]
  18. Helmstädter, A.; Staiger, C. Traditional use of medicinal agents: A valid source of evidence. Drug Discov. Today 2014, 19, 4–7. [Google Scholar] [CrossRef]
  19. Molina-Garza, Z.J.; Bazaldúa-Rodríguez, A.F.; Quintanilla-Licea, R.; Galaviz-Silva, L. Anti-Trypanosoma cruzi activity of 10 medicinal plants used in northeast Mexico. Acta Trop. 2014, 136, 14–18. [Google Scholar] [CrossRef]
  20. Hussain, T.; Bajpai, S.; Saeed, M.; Moin, A.; Alafnan, A.; Khan, M.; Kamal, M.A.; Ganash, M.; Ashraf, G.M. Potentiating effect of ethnomedicinal plants against proliferation on different cancer cell lines. Curr. Drug Metab. 2018, 19, 584–595. [Google Scholar] [CrossRef] [PubMed]
  21. Gálvez Romero, J.L.; Parada Sosa, C.M.; Burgoa, G.L.; Lorenzo Leal, A.C.; El Kassis, E.G.; Bautista Rodríguez, E.; Paredes Juárez, G.A.; Hernández, L.R.; Bach, H.; Juárez, Z.N. Antimycobacterial, cytotoxic, and anti-inflammatory activities of Artemisia ludoviciana. J. Ethnopharmacol. 2022, 293, 115249. [Google Scholar] [CrossRef]
  22. Dolghi, A.; Buzatu, R.; Marcovici, I.; Pinzaru, I.; Dehelean, C.A.; Dobrescu, A.; Olaru, F.; Popescu, G.A.; Navolan, D.; Cretu, O.M.; et al. Phytochemical analysis and in vitro cytotoxic activity against colorectal adenocarcinoma cells of Hippophae rhamnodies L., Cymbopogon citratus (D.C.) Stapf, and Ocimum basilicum L. essential oils. Plants 2021, 10, 2752–2768. [Google Scholar] [CrossRef]
  23. Rodríguez-Chávez, J.L.; Egas, V.; Linares, E.; Bye, R.; Hernández, T.; Espinosa-García, F.J.; Delgado, G. Mexican Arnica (Heterotheca inuloides Cass. Asteraceae: Astereae): Ethnomedical uses, chemical constituents and biological properties. J. Ethnopharmacol. 2017, 195, 39–63. [Google Scholar] [CrossRef]
  24. Araujo-Espino, D.I.; Zamora-Perez, A.L.; Zúñiga-González, G.M.; Gutiérrez-Hernández, R.; Morales-Velazquez, G.; Lazalde-Ramos, B.P. Genotoxic and cytotoxic evaluation of Jatropha dioica Sessé ex Cerv. by the micronucleus test in mouse peripheral blood. Regul. Toxicol. Pharmacol. 2017, 86, 260–264. [Google Scholar] [CrossRef] [PubMed]
  25. Vega-Avila, E.; Espejo-Serna, A.; Alarcón-Aguilar, F.; Velasco-Lezama, R. Cytotoxic activity of four Mexican medicinal plants. Proc. West. Pharmacol. Soc. 2009, 52, 78–82. Available online: https://www.researchgate.net/profile/Adolfo-Espejo/publication/51841133_Cytotoxic_Activity_of_Four_Mexican_Medicinal_Plants/links/0fcfd50fd574cd355c000000/Cytotoxic-Activity-of-Four-Mexican-Medicinal-Plants.pdf (accessed on 14 August 2022).
  26. Sharma, A.; Flores-Vallejo, R.d.C.; Cardoso-Taketa, A.; Villarreal, M.L. Antibacterial activities of medicinal plants used in Mexican traditional medicine. J. Ethnopharmacol. 2017, 208, 264–329. [Google Scholar] [CrossRef] [PubMed]
  27. Reyes-Melo, K.Y.; Galván-Rodrigo, A.A.; Martínez-Olivo, I.E.; Núñez-Mojica, G.; Ávalos-Alanís, F.G.; García, A.; Del Rayo Camacho-Corona, M. Larrea tridentata and its biological activities. Curr. Top. Med. Chem. 2021, 21, 2352–2364. [Google Scholar] [CrossRef]
  28. Ferreira, T.L.; Evangelista, A.J.J. Mimosa tenuiflora’s antimicrobial activity on bacteria and fungi from medical importance: An integrative review. Arch. Microbiol. 2021, 203, 3399–3406. [Google Scholar] [CrossRef]
  29. Jiménez-Estrada, M.; Huerta-Reyes, M.; Tavera-Hernández, R.; Alvarado-Sansininea, J.J.; Alvarez, A.B. Contributions from Mexican flora for the treatment of diabetes mellitus: Molecules of Psacalium decompositum (A. Gray) H. Rob & Brettell. Molecules 2021, 26, 2892–2915. [Google Scholar] [CrossRef]
  30. Nahar, L.; El-Seedi, H.R.; Khalifa, S.A.M.; Mohammadhosseini, M.; Sarker, S.D. Ruta essential oils: Composition and bioactivities. Molecules 2021, 26, 4766–4796. [Google Scholar] [CrossRef]
  31. Maldonado-Cubas, J.; San Martin-Martínez, E.; Quiroz-Reyes, C.N.; Casañas-Pimentel, R.G. Cytotoxic effect of Semialarium mexicanum (Miers) Mennega root bark extracts and fractions against breast cancer cells. Physiol. Mol. Biol. Plants Int. J. Funct. Plant Biol. 2018, 24, 1185–1201. [Google Scholar] [CrossRef]
  32. Tian, L.W.; Zhang, Z.; Long, H.L.; Zhang, Y.J. Steroidal saponins from the genus Smilax and their biological activities. Nat. Products Bioprospect. 2017, 7, 283–298. [Google Scholar] [CrossRef] [Green Version]
  33. Wang, J.B.; Qin, W.; Yang, Z.; Shen, S.; Ma, Y.; Wang, L.Y.; Zhuo, Q.; Gong, Z.L.; Huo, J.S.; Chen, C. Optimization of three-dimensional culture conditions of HepG2 cells with response surface methodology based on the VitroGel system. Biomed. Environ. Sci. 2022, 35, 688–698. [Google Scholar] [CrossRef]
  34. Guo, L.; Dial, S.; Shi, L.; Branham, W.; Liu, J.; Fang, J.L.; Green, B.; Deng, H.; Kaput, J.; Ning, B. Similarities and differences in the expression of drug-metabolizing enzymes between human hepatic cell lines and primary human hepatocytes. Drug Metab. Dispos. 2011, 39, 528–538. [Google Scholar] [CrossRef] [Green Version]
  35. Wilkening, S.; Stahl, F.; Bader, A. Comparison of primary human hepatocytes and hepatoma cell line hepg2 with regard to their biotransformation properties. Drug Metab. Dispos. 2003, 31, 1035–1042. [Google Scholar] [CrossRef]
  36. Weerapreeyakul, N.; Junhom, C.; Barusrux, S.; Thitimetharoch, T. Induction of apoptosis in human hepatocellular carcinoma cells by extracts of Lannea coromandelica (Houtt.) Merr. and Diospyros castanea (Craib) Fletcher. Chinese Med. 2016, 11, 1–10. [Google Scholar] [CrossRef] [Green Version]
  37. Bézivin, C.; Tomasi, S.; Lohezic-Le Devehat, F.; Boustie, J. Cytotoxic activity of some lichen extracts on murine and human cancer cell lines. Phytomedicine 2003, 10, 499–503. [Google Scholar] [CrossRef]
  38. Parvathy, C.R.; Praseetha, P.K. Carbon quantum dot induced hemolysis and anti-angiogenesis in proliferating cancers with Vitis vinifera as the source material. Vegetos 2022, 1–9. [Google Scholar] [CrossRef]
  39. Arora, A.; Nair, M.G.; Strasburg, G.M. Structure-activity relationships for antioxidant activities of a series of flavonoids in a liposomal system. Free Radic. Biol. Med. 1998, 24, 1355–1363. [Google Scholar] [CrossRef]
  40. Dkhil, M.A.; Delic, D.; El Enshasy, H.A.; Abdel Moneim, A.E. Medicinal plants in therapy: Antioxidant activities. Oxid. Med. Cell. Longev. 2016, 2016, 7468524. [Google Scholar] [CrossRef]
  41. Remila, S.; Atmani-Kilani, D.; Delemasure, S.; Connat, J.-L.; Azib, L.; Richard, T.; Atmani, D. Antioxidant, cytoprotective, anti-inflammatory and anticancer activities of Pistacia lentiscus (Anacardiaceae) leaf and fruit extracts. Eur. J. Integr. Med. 2015, 7, 274–286. [Google Scholar] [CrossRef]
  42. Bouyahya, A.; Guaouguaou, F.-E.; El Omari, N.; El Menyiy, N.; Balahbib, A.; El-Shazly, M.; Bakri, Y. Anti-inflammatory and analgesic properties of Moroccan medicinal plants: Phytochemistry, in vitro and in vivo investigations, mechanism insights, clinical evidences and perspectives. J. Pharm. Anal. 2022, 12, 35–57. [Google Scholar] [CrossRef]
  43. Zheng, X.; Wang, W.; Piao, H.; Xu, W.; Shi, H.; Zhao, C. The genus Gnaphalium L. (Compositae): Phytochemical and pharmacological characteristics. Molecules 2013, 18, 8298–8318. [Google Scholar] [CrossRef]
  44. Wong-Paz, J.E.; Contreras-Esquivel, J.C.; Rodríguez-Herrera, R.; Carrillo-Inungaray, M.L.; López, L.I.; Nevárez-Moorillón, G.V.; Aguilar, C.N. Total phenolic content, in vitro antioxidant activity and chemical composition of plant extracts from semiarid Mexican region. Asian Pac. J. Trop. Med. 2015, 8, 104–111. [Google Scholar] [CrossRef] [Green Version]
  45. Baqueiro-Peña, I.; Guerrero-Beltrán, J. Physicochemical and antioxidant characterization of Justicia spicigera. Food Chem. 2017, 218, 305–312. [Google Scholar] [CrossRef]
  46. Elizondo-Luevano, J.H.; Verde-Star, J.; González-Horta, A.; Castro-Ríos, R.; Hernández-García, M.E.; Chávez-Montes, A. In vitro effect of methanolic extract of Argemone mexicana against Trichomonas vaginalis. Korean J. Parasitol. 2020, 58, 135–145. [Google Scholar] [CrossRef]
  47. Elizondo-Luévano, J.H.; Castro-Ríos, R.; López-Abán, J.; Gorgojo-Galindo, O.; Fernández-Soto, P.; Vicente, B.; Muro, A.; Chávez-Montes, A. Berberine: A nematocidal alkaloid from Argemone mexicana against Strongyloides venezuelensis. Exp. Parasitol. 2021, 220, 1–7. [Google Scholar] [CrossRef]
  48. Orozco-Flores, A.A.; Valadez-Lira, J.A.; Covarrubias-Cárdenas, K.E.; Pérez-Trujillo, J.J.; Gomez-Flores, R.; Caballero-Hernández, D.; Tamez-Guerra, R.; Rodríguez-Padilla, C.; Tamez-Guerra, P. In vitro antitumor, pro-inflammatory, and pro-coagulant activities of Megalopyge opercularis J.E. Smith hemolymph and spine venom. Sci. Rep. 2020, 10, 1–10. [Google Scholar] [CrossRef]
  49. Stevigny, C.; Bailly, C.; Quetin-Leclercq, J. Cytotoxic and antitumor potentialities of aporphinoid alkaloids. Curr. Med. Chem. Anti-Cancer Agents 2005, 5, 173–182. [Google Scholar] [CrossRef] [Green Version]
  50. Ozsoylemez, O.D.; Ozcan, G. Effects of Colchicum baytopiorum leaf extract on cytotoxicity and cell death pathways in C-4 I and Vero cell lines. J. BUON Off. J. Balk. Union Oncol. 2021, 26, 1135–1147. [Google Scholar]
  51. Bazaldúa-Rodríguez, A.F.; Quintanilla-Licea, R.; Verde-Star, M.J.; Hernández-García, M.E.; Vargas-Villarreal, J.; Garza-González, J.N. Furanocoumarins from Ruta chalepensis with amebicide activity. Molecules 2021, 26, 3684–3694. [Google Scholar] [CrossRef]
  52. Loizzo, M.R.; Falco, T.; Bonesi, M.; Sicari, V.; Tundis, R.; Bruno, M. Ruta chalepensis L. (Rutaceae) leaf extract: Chemical composition, antioxidant and hypoglicaemic activities. Nat. Prod. Res. 2018, 32, 521–528. [Google Scholar] [CrossRef]
  53. Varamini, P.; Soltani, M.; Ghaderi, A. Cell cycle analysis and cytotoxic potential of Ruta graveolens against human tumor cell lines. Neoplasma 2009, 56, 490–493. [Google Scholar] [CrossRef] [Green Version]
  54. Réthy, B.; Zupkó, I.; Minorics, R.; Hohmann, J.; Ocsovszki, I.; Falkay, G. Investigation of cytotoxic activity on human cancer cell lines of arborinine and furanoacridones isolated from Ruta graveolens. Planta Med. 2007, 73, 41–48. [Google Scholar] [CrossRef]
  55. Günaydin, K.; Savci, S. Phytochemical studies on Ruta chalepensis (LAM.) Lamarck. Nat. Prod. Res. 2005, 19, 203–210. [Google Scholar] [CrossRef]
  56. Jacobo-Salcedo, M.D.R.; Alonso-Castro, A.J.; Salazar-Olivo, L.A.; Carranza-Alvarez, C.; Gonzaĺez-Espińdola, L.A.; Domińguez, F.; Maciel-Torres, S.P.; Garciá-Lujan, C.; Gonzaĺez-Martińez, M.D.R.; Goḿez-Sańchez, M.; et al. Antimicrobial and cytotoxic effects of Mexican medicinal plants. Nat. Prod. Commun. 2011, 6, 1925–1928. [Google Scholar] [CrossRef] [Green Version]
  57. Ángeles-López, G.E.; González-Trujano, M.E.; Rodríguez, R.; Déciga-Campos, M.; Brindis, F.; Ventura-Martínez, R. Gastrointestinal activity of Justicia spicigera Schltdl. in experimental models. Nat. Prod. Res. 2021, 35, 1847–1851. [Google Scholar] [CrossRef]
  58. Zapata-Morales, J.R.; Alonso-Castro, A.J.; Domínguez, F.; Carranza-Álvarez, C.; Castellanos, L.M.O.; Martínez-Medina, R.M.; Pérez-Urizar, J. Antinociceptive activity of an ethanol extract of Justicia spicigera. Drug Dev. Res. 2016, 77, 180–186. [Google Scholar] [CrossRef]
  59. Patel, D.K. Pharmacological activities and therapeutic potential of kaempferitrin in medicine for the treatment of human disorders: A review of medicinal importance and health benefits. Cardiovasc. Hematol. Disord. Drug Targets 2021, 21, 104–114. [Google Scholar] [CrossRef]
  60. Melchor-Martínez, E.M.; Silva-Mares, D.A.; Torres-López, E.; Waksman-Minsky, N.; Pauli, G.F.; Chen, S.-N.; Niemitz, M.; Sánchez-Castellanos, M.; Toscano, A.; Cuevas, G.; et al. Stereochemistry of a second riolozane and other diterpenoids from Jatropha dioica. J. Nat. Prod. 2017, 80, 2252–2262. [Google Scholar] [CrossRef]
  61. Castro-Ríos, R.; Melchor-Martínez, E.M.; Solís-Cruz, G.Y.; Rivas-Galindo, V.M.; Silva-Mares, D.A.; Cavazos-Rocha, N.C. HPLC method validation for Jatropha dioica extracts analysis. J. Chromatogr. Sci. 2020, 58, 445–453. [Google Scholar] [CrossRef]
  62. Vargas-Segura, A.I.; Silva-Belmares, S.Y.; Segura-Ceniceros, E.P.; Ascacio-Valdés, J.A.; Méndez-González, L.; Ilyina, A. Screening and characterization of medicinal plants extracts with bactericidal activity against Streptococcus mutans. Nat. Prod. Res. 2020, 34, 2672–2676. [Google Scholar] [CrossRef]
  63. Li, P.; Li, L.; Zhu, Q.; Xu, M. Abietane diterpenoids isolated from Clerodendrum bracteatum and their antioxidant and cytotoxic activities. Molecules 2021, 26, 4870–4876. [Google Scholar] [CrossRef]
  64. Silva-Mares, D.; Torres-López, E.; Rivas-Estilla, A.M.; Cordero-Pérez, P.; Waksman-Minsky, N.; Rivas-Galindo, V.M. Plants from northeast Mexico with anti-HSV activity. Nat. Prod. Commun. 2013, 8, 297–298. [Google Scholar] [CrossRef]
  65. Knauth, P.; Acevedo-Hernández, G.J.; Cano, M.E.; Gutiérrez-Lomelí, M.; López, Z. In vitro bioactivity of methanolic extracts from Amphipterygium adstringens (Schltdl.) Schiede ex Standl., Chenopodium ambrosioides L., Cirsium mexicanum DC., Eryngium carlinae F. Delaroche, and Pithecellobium dulce (Roxb.) Benth. Used in Traditional Medi. Evid. Based Complement. Altern. Med. 2018, 2018, 1–11. [Google Scholar] [CrossRef] [Green Version]
  66. Alam-Escamilla, D.; Estrada-Muñiz, E.; Solís-Villegas, E.; Elizondo, G.; Vega, L. Genotoxic and cytostatic effects of 6-pentadecyl salicylic anacardic acid in transformed cell lines and peripheral blood mononuclear cells. Mutat. Res. Genet. Toxicol. Environ. Mutagen. 2015, 777, 43–53. [Google Scholar] [CrossRef]
  67. Rodriguez-Garcia, A.; Peixoto, I.T.A.; Verde-Star, M.J.; De La Torre-Zavala, S.; Aviles-Arnaut, H.; Ruiz, A.L.T.G. in vitro antimicrobial and antiproliferative activity of Amphipterygium adstringens. Evid. Based Complement. Altern. Med. 2015, 2015, 1–7. [Google Scholar] [CrossRef] [Green Version]
  68. Lambertini, E.; Piva, R.; Khan, M.T.H.; Lampronti, I.; Bianchi, N.; Borgatti, M.; Gambari, R. Effects of extracts from Bangladeshi medicinal plants on in vitro proliferation of human breast cancer cell lines and expression of estrogen receptor alpha gene. Int. J. Oncol. 2004, 24, 419–423. [Google Scholar] [CrossRef]
  69. Singh, S.; Verma, M.; Malhotra, M.; Prakash, S.; Singh, S.; Verma, M.; Malhotra, M.; Prakash, S.; Singh, T.D. Cytotoxicity of alkaloids isolated from Argemone mexicana on SW480 human colon cancer cell line. Pharm. Biol. 2016, 54, 740–745. [Google Scholar] [CrossRef]
  70. De La Cruz-Jiménez, L.; Hernández-Torres, M.A.; Monroy-García, I.N.; Rivas-Morales, C.; Verde-Star, M.J.; Gonzalez-Villasana, V.; Viveros-Valdez, E. Biological activities of seven medicinal plants used in Chiapas, Mexico. Plants 2022, 11, 1790–1801. [Google Scholar] [CrossRef]
  71. Sangkitikomol, W. Antioxidants in Thai herb, vegetable and fruit inhibit hemolysis and Heinz body formation in human erythrocytes. In Oxidative Stress—Environmental Induction and Dietary Antioxidants; Lushchak, V., Ed.; InTech: Shanghai, China, 2012; pp. 289–306. [Google Scholar]
  72. Coballase-Urrutia, E.; Pedraza-Chaverri, J.; Camacho-Carranza, R.; Cárdenas-Rodríguez, N.; Huerta-Gertrudis, B.; Medina-Campos, O.N.; Mendoza-Cruz, M.; Delgado-Lamas, G.; Espinosa-Aguirre, J.J. Antioxidant activity of Heterotheca inuloides extracts and of some of its metabolites. Toxicology 2010, 276, 41–48. [Google Scholar] [CrossRef]
  73. Bourvellec, C.L.; Bureau, S.; Renard, C.M.G.C.; Plenet, D.; Gautier, H.; Touloumet, L.; Girard, T.; Simon, S. Cultivar and year rather than agricultural practices affect primary and secondary metabolites in apple fruit. PLoS ONE 2015, 10, e0141916. [Google Scholar] [CrossRef] [PubMed]
  74. Ramírez-Villalobos, J.M.; Romo-Sáenz, C.I.; Morán-Santibañez, K.S.; Tamez-Guerra, P.; Quintanilla-Licea, R.; Orozco-Flores, A.A.; Romero-Arguelles, R.; Tamez-Guerra, R.; Rodríguez-Padilla, C.; Gomez-Flores, R. In vitro tumor cell growth inhibition induced by Lophocereus marginatus (DC.) S. Arias and Terrazas endophytic fungi extracts. Int. J. Environ. Res. Public Health 2021, 18, 9917–9925. [Google Scholar] [CrossRef]
  75. Effects, A. Nanoemulsions of Jasminum humile L. and Jasminum grandiflorum L. Essential Oils: An Approach to Enhance Their Cytotoxic and Antiviral Effects. Molecules 2022, 27, 36–39. [Google Scholar] [CrossRef]
  76. Al-Qubaisi, M.; Rozita, R.; Yeap, S.K.; Omar, A.R.; Ali, A.M.; Alitheen, N.B. Selective cytotoxicity of goniothalamin against hepatoblastoma HepG2 cells. Molecules 2011, 16, 2944–2959. [Google Scholar] [CrossRef] [Green Version]
  77. Elizondo-Luévano, J.H.; Pérez-Narváez, O.A.; Sánchez-García, E.; Castro-Ríos, R.; Hernández-García, M.E.; Chávez-Montes, A. In-vitro effect of Kalanchoe daigremontiana and its main component, quercetin against Entamoeba histolytica and Trichomonas vaginalis. Iran. J. Parasitol. 2021, 16, 394–401. [Google Scholar] [CrossRef]
  78. Pérez Marqués, U.; Murillo, G.; Tur, E.; Vinardell, M.P.; García Simón, G.; Pascual, J.R. Evaluación de la irritación ocular mediante un ensayo de hemólisis y desnaturalización de la hemoglobina in vitro. Rev. Toxicol. 2003, 20, 193–198. [Google Scholar]
  79. Elizondo-Luévano, J.H.; Hernández-García, M.E.; Pérez-Narváez, O.A.; Castro-Ríos, R.; Chávez-Montes, A. Berberina, curcumina y quercetina como potenciales agentes con capacidad antiparasitaria. Rev. Biol. Trop. 2020, 68, 1241–1249. [Google Scholar] [CrossRef]
  80. Pieroni, L.G.; De Rezende, F.M.; Ximenes, V.F.; Dokkedal, A.L. Antioxidant activity and total phenols from the methanolic extract of Miconia albicans (Sw.) Triana leaves. Molecules 2011, 16, 9439–9450. [Google Scholar] [CrossRef]
  81. Elizondo-Luévano, J.H.; Castro-Ríos, R.; Vicente, B.; Fernández-Soto, P.; López-Aban, J.; Muro, A.; Chávez-Montes, A.; Chávez-Montes, A. In vitro antischistosomal activity of the Argemone mexicana methanolic extract and its main component berberine. Iran. J. Parasitol. 2021, 16, 91–100. [Google Scholar] [CrossRef]
Table 1. Plants used in this study and percentage of extraction yields.
Table 1. Plants used in this study and percentage of extraction yields.
Plant SpeciesFamilyMexican Common
Name
Evaluated
Plant Part
Voucher NumberYield %
Amphipterygium adstringens
(Schltdl.) Standl.
AnacardiaceaeCuachalalate Bark3064238.36
Argemone mexicana L.PapaveraceaeChicaloteLeaves2912711.26
Artemisia ludoviciana Nutt.CompositaeEstafiateLeaves3064318.39
Cymbopogon citratus (DC.) Stapf.PoaceaeZacate limónLeaves3064423.04
Heterotheca inuloides Cass.CompositaeMexican arnicaFlowers3064620.49
Jatropha dioica SesséEuphorbiaceaeSangre de dragónRoot3064815.97
Justicia spicigera Schltdl.AcanthaceaeMuicleLeaves3064913.18
Larrea tridentata (Sessé & Moc. ex DC.) CovilleZygophyllaceaeGobernadoraLeaves3065013.17
Mimosa tenuiflora (Willd.) Poir.LeguminosaeTepezcohuiteBark3065110.84
Psacalium peltatum (Kunth) Cass.CompositaeMatariqueLeaves3065210.93
Pseudognaphalium obtusifolium (L.) Hilliard & B.L.Burtt.CompositaeGordoloboLeaves3065316.99
Ruta chalepensis L. RutaceaeRudaRoot3065419.39
Semialarium mexicanum
(Miers) Mennega
CelastraceaeCancerinaBark3064710.52
Smilax aspera L.SmilacaceaeZarzaparrillaLeaves3065513.14
Tagetes lucida Cav.CompositaeHierbanís or YerbanizBark3065620.63
Table 2. Cytotoxic activity of Mexican plant extracts against HEP-G2 tumor cells and Vero non-tumor cells.
Table 2. Cytotoxic activity of Mexican plant extracts against HEP-G2 tumor cells and Vero non-tumor cells.
Plant ExtractHEP-G2
IC50 (µg/mL)
Vero
IC50 (µg/mL)
SI
Amphipterygium adstringens41.77 ± 6.18 c197.98 ± 4.71 e4.74
Argemone mexicana820.78 ± 20.81 h245.41 ± 13.05 g0.29
Artemisia ludoviciana1034.76 ± 12.01 i,j197.37 ± 2.79 e0.19
Cymbopogon citratus1560.01 ± 23.26 j24.47 ± 3.79 a0.02
Heterotheca inuloides1002.08 ± 14.81 i36.95 ± 8.22 b0.04
Jatropha dioica12.34 ± 3.12 b70.59 ± 9.73 d5.72
Justicia spicigera2.92 ± 0.54 a54.91 ± 7.60 c18.84
Larrea tridentata403.05 ± 13.72 d214.64 ± 1.63 f0.53
Mimosa tenuiflora809.64 ± 19.72 h467.59 ± 13.72 h0.58
Psacalium peltatum975.81 ± 15.19 i54.91 ± 4.94 c0.06
Pseudognaphalium obtusifolium690.05 ± 10.45 g61.98 ± 2.82 c,d0.09
Ruta chalepensis1.79 ± 0.38 a522.08 ± 29.96 i291.50
Semialarium mexicanum527.10 ± 20.87 e20.75 ± 3.16 a0.04
Smilax aspera393.05 ± 15.06 d582.11 ± 31.14 i1.48
Tagetes lucida607.93 ± 11.15 f780.62 ± 9.27 j1.28
Data are means ± SD (p < 0.05) of the IC50 in µg/mL for each extract, against the evaluated cell lines. Different letters within the same column are significantly different, analyzed by the post-hoc Tukey test. The selectivity index (SI) represents IC50 for the normal cell line (Vero) between the IC50 for the cancerous cell line (HEP-G2) after 72 h. Vincristine sulfate was used as a positive control at a concentration of 0.05 µg/mL, as detailed in the Section 4.
Table 3. Antioxidant activity of methanol plant extracts by the DPPH radical scavenging method.
Table 3. Antioxidant activity of methanol plant extracts by the DPPH radical scavenging method.
Plant ExtractDPPH Assay (IC50 in µg/mL)
Amphipterygium adstringens504.89 ± 34.33 c
Argemone mexicana565.98 ± 17.60 c
Artemisia ludoviciana723.33 ± 25.92 d,e
Cymbopogon citratus690.4 ± 26.37 d
Heterotheca inuloides19.24 ± 2.11 b
Jatropha dioica681.18 ± 15.64 d
Justicia spicigera924.92 ± 30.83 f
Larrea tridentata665.41 ± 31.70 d
Mimosa tenuiflora547.66 ± 28.87 c
Psacalium peltatum520.52 ± 15.69 c
Pseudognaphalium obtusifolium528.67 ± 25.78 c
Ruta chalepensis859.85 ± 25.08 e
Semialarium mexicanum1,639.79 ± 35.74 g
Smilax aspera936.5 ± 11.19 f
Tagetes lucida550.85 ± 16.09 c
Ascorbic acid (positive control)7.23 ± 0.03 a
Data are means ± SD (p < 0.05) of the IC50 in µg/mL for each evaluated extract. Different letters within the same column are significantly different, analyzed by the post-hoc Tukey test.
Table 4. Hemolytic and anti-hemolytic activities of Mexican plant extracts on human erythrocytes.
Table 4. Hemolytic and anti-hemolytic activities of Mexican plant extracts on human erythrocytes.
Plant ExtractHemolytic ActivityAnti-Hemolytic Activity
IC50 (µg/mL)
Amphipterygium adstringens203.62 ± 11.96 a5.35 ± 1.63 a
Argemone mexicana973.88 ± 38.46 f101.60 ± 10.21 d
Artemisia ludoviciana746.39 ± 12.80 d50.31 ± 7.64 c
Cymbopogon citratus606.82 ± 19.12 c32.01 ± 4.74 b
Heterotheca inuloides835.73 ± 23.73 e5.42 ± 0.89 a
Jatropha dioica545.74 ± 8.76 b72.92 ± 4.85 d
Justicia spicigera˃2500 81.08 ± 8.10 d
Larrea tridentata741.71 ± 12.80 d777.85 ± 18.58 f
Mimosa tenuiflora˃2500 71.24 ± 6.47 d
Psacalium peltatum˃2500 5.92 ± 1.29 a
Pseudognaphalium obtusifolium˃2500 143.17 ± 19.29 e
Ruta chalepensis738.73 ± 20.74 d28.29 ± 2.31 b
Semialarium mexicanum1976.75 ± 26.06 f41.32 ± 8.27 b c
Smilax aspera625.17 ± 13.11 c4.41 ± 0.69 a
Tagetes lucida843.84 ± 31.93 e62.48 ± 8.52 c
Data are mean ± SD (p < 0.05) of the IC50 in µg/mL for each extract evaluated. Different letters within the same column are significantly different, analyzed by the post-hoc Tukey test. As IC50 was above 2500 µg/mL, these values were not considered for the Tukey analysis.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Elizondo-Luévano, J.H.; Gomez-Flores, R.; Verde-Star, M.J.; Tamez-Guerra, P.; Romo-Sáenz, C.I.; Chávez-Montes, A.; Rodríguez-Garza, N.E.; Quintanilla-Licea, R. In Vitro Cytotoxic Activity of Methanol Extracts of Selected Medicinal Plants Traditionally Used in Mexico against Human Hepatocellular Carcinoma. Plants 2022, 11, 2862. https://doi.org/10.3390/plants11212862

AMA Style

Elizondo-Luévano JH, Gomez-Flores R, Verde-Star MJ, Tamez-Guerra P, Romo-Sáenz CI, Chávez-Montes A, Rodríguez-Garza NE, Quintanilla-Licea R. In Vitro Cytotoxic Activity of Methanol Extracts of Selected Medicinal Plants Traditionally Used in Mexico against Human Hepatocellular Carcinoma. Plants. 2022; 11(21):2862. https://doi.org/10.3390/plants11212862

Chicago/Turabian Style

Elizondo-Luévano, Joel H., Ricardo Gomez-Flores, María J. Verde-Star, Patricia Tamez-Guerra, César I. Romo-Sáenz, Abelardo Chávez-Montes, Nancy E. Rodríguez-Garza, and Ramiro Quintanilla-Licea. 2022. "In Vitro Cytotoxic Activity of Methanol Extracts of Selected Medicinal Plants Traditionally Used in Mexico against Human Hepatocellular Carcinoma" Plants 11, no. 21: 2862. https://doi.org/10.3390/plants11212862

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