Next Article in Journal
Development and Characterization of a Novel α-Synuclein-PEST H4 Cell Line for Enhanced Drug Screening in α-Synucleinopathies
Previous Article in Journal
Dysregulated miRNAs Targeting Adiponectin Signaling in Colorectal Cancer
Previous Article in Special Issue
Hesperetin-Enhanced Metformin to Alleviate Cognitive Impairment via Gut–Brain Axis in Type 2 Diabetes Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Metabolomic and Pharmacological Approaches for Exploring the Potential of Tanacetum parthenium L. Root Culture as a Source of Bioactive Phytochemicals

by
Aurelio Nieto-Trujillo
1,
Rosendo Luria-Pérez
2,
Francisco Cruz-Sosa
3,
Carmen Zepeda-Gómez
4,
María G. González-Pedroza
4,
Cristina Burrola-Aguilar
1,
Armando Sunny
5,
José Correa-Basurto
6,
José A. Guerrero-Analco
7,
Juan L. Monribot-Villanueva
7 and
María Elena Estrada-Zúñiga
1,*
1
Centro de Investigación en Recursos Bióticos, Facultad de Ciencias, Universidad Autónoma del Estado de México, Carretera Toluca-Ixtlahuaca km 14.5, San Cayetano, Toluca 50295, Mexico
2
Unidad de Investigación en Enfermedades Oncológicas, Hospital Infantil de México Federico Gómez, Dr. Márquez No 162, Col. Doctores, Cuauhtémoc, Mexico City 06720, Mexico
3
Departamento de Biotecnología, Universidad Autónoma Metropolitana-Unidad Iztapalapa, Av. Ferrocarril San Rafael Atlixco No 186, Leyes de Reforma 1ra Sección, Iztapalapa, Mexico City 09310, Mexico
4
Facultad de Ciencias, Universidad Autónoma del Estado de México, Campus El Cerrillo, Piedras Blancas, Carretera Toluca-Ixtlahuaca km. 15.5, Toluca 50200, Mexico
5
Centro de Investigación en Ciencias Biológicas Aplicadas, Facultad de Ciencias, Universidad Autónoma del Estado de México, Carretera Toluca-Ixtlahuaca km 14.5, San Cayetano, Toluca 50295, Mexico
6
Laboratorio de Modelado Molecular, Bioinformática y Diseño de Fármacos, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Salvador Díaz Mirón S/N, Casco de Santo Tomás, Mexico City 11340, Mexico
7
Laboratorio de Química de Productos Naturales, Red de Estudios Moleculares Avanzados, Instituto de Ecología, A.C., Carretera Antigua a Coatepec 351, El Haya, Xalapa 91073, Mexico
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(15), 7209; https://doi.org/10.3390/ijms26157209
Submission received: 20 June 2025 / Revised: 17 July 2025 / Accepted: 21 July 2025 / Published: 25 July 2025
(This article belongs to the Special Issue Plant Phenolic Accumulation and Application in Human Diseases)

Abstract

Tanacetum parthenium (Asteraceae) has been traditionally used worldwide for medicinal purposes, and some of its therapeutic uses have been attributed to the pharmacological effects of its secondary metabolites. The root culture of this species might represent a sustainable source of several pharmacologically active compounds. The biomass of a root T. parthenium culture was extracted with methanol and fractionated using column chromatography. Three selected fractions (4TP, 5TP, and 8TP) were analyzed via spectrophotometric, chromatographic, and mass spectrometry techniques and in vitro pharmacological assays. The greatest values for total phenolic and phenolic acid contents and antibacterial activity against Escherichia coli were determined for 4TP. The highest values for total flavonoid and sesquiterpene lactone contents, antioxidant potential, and α-amylase inhibitory effect were determined for 8TP. The antibacterial effect against Staphylococcus aureus was not significantly different among the three fractions. The root culture of T. parthenium is a potential source of several metabolites, such as phenolic acids, fatty acids, coumarins, sesquiterpenoids, and triterpenoids, which are capable of exerting α-amylase inhibition and antioxidant, antibacterial, and cytotoxic effects. Among eight phenolic compounds detected and quantified in the fractions, chlorogenic acid was the most abundant.

Graphical Abstract

1. Introduction

In vitro cultures of medicinal plant producers of secondary metabolites have gained significant attention in recent decades since they are a biotechnological tool representing a sustainable source of compounds able to exert pharmacological effects [1,2,3]. In vitro culture consists of several techniques for cultivating cells, tissues, organs, or plantlets under relative control of genetics, metabolism, and environmental growth conditions [1]. Among the applications of plant in vitro culture are the production of bioactive secondary metabolites because this method offers major advantages over whole plants, such as allowing uniform yield and quality of plant-derived products, shorter and continuous production cycles, efficient recovery of bioactive compounds, lack of environmental or genetic contamination due to controlled aseptic conditions, and scalable production from laboratory volumes to industrial bioreactors [4,5]. However, achieving industrial-scale production in a bioreactor implies great previous background research in flasks, laboratory volume bioreactors, and scale-up to industrial volumes, focused on determining the factors that influence biomass production and enhancing the yield production and recovery processes of specific secondary metabolites with pharmacological potential proven for designing drugs [6,7].
In recent years, the potential of in vitro cultures of medicinal plants as a source of biopharmaceutical compounds has been researched by determining the biological effects of their extracts via in vitro and in vivo pharmacological tests, e.g., antioxidant, antibacterial, cytotoxic, and antidiabetic effects [8]. Assessing the antioxidant capability of plant extracts has been widely reported since the overproduction of reactive oxygen species is responsible for the pathogenesis of many nontransmissible diseases, such as atherosclerosis, cancer, diabetes, inflammation, liver injury, aging, cardiovascular and neurodegenerative diseases, and others [9,10,11]. The antioxidant effect of these extracts has been related to phenolic compounds and terpenoid secondary metabolites because of their ability to scavenge free radicals [9,12,13]. Depending on their structure and chemical nature, these secondary metabolites might also exhibit other pharmacological activities, e.g., antibacterial, cytotoxic, and antidiabetic activities [9].
In addition, metabolomics can be a complementary tool for the plant in vitro cultures [14] to examine its potential to produce several pharmacologically active secondary metabolites. In recent decades, untargeted metabolomics has gained great importance in identifying the metabolites produced by organisms under specific conditions [15]. In medicinal plants, untargeted metabolomics can be applied to identify a vast number of secondary metabolites as a “fingerprint” related to pharmacological effects, which might also contribute to obtaining chemical quality control [15,16,17]. The putative identification of metabolites obtained during untargeted metabolomics may establish the basis for carrying out targeted metabolomics to confirm the identification and quantification of potential secondary biomarkers, such as those demonstrated in dark tea (Camelia sinensis) [18].
Tanacetum parthenium L. (Asteraceae) is an aromatic and perennial herb that occurs in the Caucasian region and southern Europe, but it was introduced and naturalized in other regions of Africa, Asia, and America [19]. This plant has been traditionally used worldwide for medicinal purposes, but it is strongly associated with inflammation and pain [20,21]. Several of the therapeutic properties reported for this species have been attributed to their secondary metabolites, as they have proven to have pharmacological effects; among those secondary metabolites, terpenes (sesquiterpene lactones and essential oils) and phenolic compounds (flavonoids and phenolic acids) are highlighted [20,21,22,23]. The sesquiterpene lactones reported for T. parthenium include nearly thirty compounds, such as artecanin, balchanin, canin, costunolide, parthenolide (PTN), reynosin, santamarine, and tanacetin, among others; PTN has been reported to be the most abundant [20] and the most pharmacologically important because of its significant anti-inflammatory, neuroprotective, cardioprotective, and anticancer effects [20,24,25]. While some flavonoids of this species, such as tanetin, quercetin, apigenin, luteolin, chrysoeriol, santin, and kaempferol derivatives, among others [20], have been found to be involved in anticancer, antioxidant, or anti-inflammatory activities [24,25]. Finally, essential oil has been reported to be responsible for antimicrobial activity, and some examples of this type of secondary metabolite reported in this species include camphor, farnesol, trans-chrysanthenyl acetate, camphene, borneol, p-cymene, and bornyl acetate; however, camphor has been reported as the most abundant compound [20,26,27,28,29].
Several works reporting T. parthenium in vitro cultures, whether focused on the propagation of this species or aimed at producing bioactive secondary metabolites, have focused mainly on PTN production because of its pharmacological effects, especially its anticancer activity [30,31,32,33,34,35,36,37,38]. In previous work, Nieto-Trujillo et al. [36] demonstrated that a T. parthenium root culture produced chlorogenic acid, caffeic acid, salicylic acid, and PTN, but their production depended on the time of culture; the largest amounts of all those secondary metabolites (1.347 ± 0.159 mg chlorogenic acid/g; 0.851 ± 0.099 mg caffeic acid/g; 0.19 ± 0.028 mg salicylic acid/g, and 0.0231 ± 0.011 mg PTN/g) were produced after thirty-two days of growth; however, in all those previous reports of this species, no attempts were made to assess pharmacological effects, or untargeted metabolomics for chemical profiling. Thus, to increase knowledge of our previous work, by Nieto-Trujillo et al. [36], we consider that combining these two methods could probably be useful for exploring the potential of a root culture of T. parthenium as a source of bioactive phytochemicals.
This work aimed to estimate the potential of Tanacetum parthenium root culture to produce several phytochemicals that have antioxidant, antibacterial, cytotoxic, and α-amylase effects in vitro through the phytochemical and pharmacological characterization of three fractions from a methanolic extract.

2. Results

2.1. Selection of Fractions

Phytochemical qualitative screening of phenolic compounds, flavonoids, and sesquiterpene lactones in a total of ten fractions obtained after the fractionation of the methanolic extract of T. parthenium root culture revealed a main intensity in precipitate positive response or color for three fractions, coded 4TP, 5TP, and 8TP; thus, these fractions were selected for further experiments.

2.2. The Three Fractions Exhibited Pharmacological Activity

The 4TP, 5TP, and 8TP fractions significantly affected the antioxidant, antibacterial, cytotoxic activities, and inhibition of α-amylase enzyme, generally depending on the tested concentration and type of fraction (Figure 1a–f). Significant differences in antioxidant and α-amylase inhibitory effects between the three fractions were determined, with the greatest values occurring for both effects in the 8TP fraction (IC50 of 2.3 μg/mL for scavenging the DPPH radical; 10.15% enzyme inhibition at a concentration of 10 µg/mL) (Figure 1a,b). The control tested for the antioxidant assay (MeOH) could not scavenge the DPPH radical. When the results of the enzyme inhibition were expressed as mg of acarbose equivalents per gram of fraction (mg AE/gF), 8TP had the highest value (23.36 ± 3.3 mg AE/gF) compared with 4TP and 5TP (12.12 ± 3.27 and 4.80 ± 1.73 mg AE/gF, respectively). A comparison between the 8TP fraction and the positive control (acarbose) regarding the effect of α-amylase inhibition revealed that the fraction with a significantly lower percentage of inhibition than that observed in the positive control (IC50 of 2.48 ± 0.31 μg acarbose/mL). All fractions significantly caused the inhibition of the growth of both bacteria at all the tested concentrations, but the highest percentage of bacteria inhibited against E. coli was found at 6 μg/disk, which was significantly different among the fractions. Thus, the percentage of bacteria inhibited at 6 μg/disk concentration decreased in the order 4TP > 8TP > 5TP (corresponding to 27.0%, 20.4%, and 14.8%, respectively) (Figure 1c). For S. aureus, the highest inhibition percentage for any fraction was reached at 8 μg/disk, and the three fractions were not significantly different (for 18.80–18.93%) (Figure 1d). Since the percentage inhibition against these bacteria was estimated for the positive control (antibiotic), the results showed that the fractions possessed a lower antibacterial effect than the antibiotic drug reference. Finally, for all the fractions, the cellular viability of the Ramos RA-1 lymphoma cells significantly decreased at concentrations higher than 20 μg/mL, with the best cytotoxic effect found at 40 μg/mL (cellular viability of approximately 35% for 4TP and 15% for 5TP and 8TP) (Figure 1e). A cytotoxic activity comparison between fractions determined the 5TP fraction as the best (IC50 of 26.37 ± 1.31 μg/mL against 30.91 ± 0.76 and 30.30 ± 0.94 μg/mL, for 4TP and 8TP, respectively). By contrasting the cytotoxic effect of the fractions with that of the positive control (1 nM vincristine), the fractions were found to be more toxic since this drug reference caused a cellular viability of 72.61% ± 1.45%. Moreover, the combination of each fraction at different concentrations with 1 nM vincristine caused enhanced cytotoxicity on the cells compared to the results obtained for the fractions alone; this effect depended on the type of fraction and its concentration (Figure 1f). The best cytotoxic effect of the combination of the fraction and vincristine mixture occurred for 4TP and 5TP at 40 μg/mL, which caused almost 5% cellular viability (Figure 1e). A comparison of the IC50 values for each fraction combined with 1 nM vincristine showed no significant differences (17.88 ± 1.41, 16.71 ± 1.26, and 18.45 ± 1.99 μg/mL for 4TP, 5TP, and 8TP with 1 nM vincristine, respectively).

2.3. Phytochemical Characterization of the Three Fractions

2.3.1. Total Secondary Metabolite Contents Profile of Fractions

The phytochemical characterization of the 4TP, 5TP, and 8TP fractions, estimated by determining the total secondary metabolite contents (TPC, TPAC, TFC and TSLC), revealed significant differences (Figure 2a). The 4TP fraction had the highest values for TPC and TPAC (194.2 mg GAE/gF and 143.6 mg VBE/gF, respectively; Figure 2a), while the 8TP fraction had the highest values for TFC and TSLC (11.9 mg QE/gF and 14.7 mg PTNE/gF, respectively; Figure 2a).

2.3.2. Differential Abundance of Compounds Between the Three Fractions from Untargeted Metabolomics

After processing the chromatographic data obtained from the LC-MS analysis by the Molecular Feature Extraction algorithm, based on spectrometric features (retention time_m/z ratio values) of the chromatographic, 97 compounds were distributed among the 4TP, 5TP, and 8TP fractions. The three fractions exhibited different chromatographic profiles, with several notable chromatographic peaks eluted from 20.5 to 23 min of retention time in the negative (Figure 2b) and positive (Figure 2c) ionization modes by electrospray. The Venn diagram showed a differential distribution of those compounds between fractions; 64, 33, and 37 signals were detected in the 4TP, 5TP, and 8TP fractions, respectively, with 44, 6, and 16 signals observed solely in the 4TP, 5TP, and 8TP fractions, respectively; at the same time, six signals were found in common in the three fractions (Figure 2d). The PCA, which was used to determine the relationship of the compound abundances between the three fraction replicates, indicated high (64.1%) variation (Figure 2e), which was also confirmed with PLS-DA (63%; Figure 2f), the VIP score (Figure 2g), and a heatmap (Figure 2h).

2.3.3. The Phytochemical and Pharmacological Activities and Compound Abundances of the Three Fractions Were Correlated

The phytochemical profiles (TPC, TPAC, TFC, and TSLC) of the three fractions were correlated with their pharmacological effects; these correlations and their types (positive or negative) were significantly influenced by the type of secondary metabolite (Figure 3a). TPC and TPAC were positively correlated with antioxidant, antibacterial, and cytotoxic effects in the following order from high to moderate: antioxidant > antibacterial against E. coli > antibacterial against S. aureus > cytotoxic (Figure 3a). TFC was significantly correlated with α-amylase inhibition and cytotoxic effects in the following order from high to moderate: α-amylase inhibition > cytotoxic (Figure 3a). Moreover, the TSLC was significantly correlated, in a moderate way, with the α-amylase inhibition effect (Figure 3a). When every fraction was correlated with the phytochemical profile and pharmacological effects, more moderate positive correlations were observed in the 4TP fraction than in the 5TP and 8TP fractions (Figure 3b). In addition, approximately 30% of the total 97 compounds showed a positive correlation with TFC, TSLC, α-amylase inhibition, and cytotoxic effects. Nearly 60% also had a positive correlation with TPC; TPAC; and antioxidant, antibacterial, and cytotoxic effects, and the remaining percentage was not correlated with any total secondary metabolite content but was correlated with antibacterial effects against S. aureus (Figure 3c). When the abundances of the top compounds were correlated with every pharmacological effect or total secondary metabolite content and compared among the fractions, it was observed that the fractions had different compounds able to exert particular pharmacological effects and even the same compound contributing to different pharmacological effects (Figure 3d); thus, the compounds showing the greatest values were selected for putative identification.

2.3.4. Putative Identification of Metabolites Correlated with Pharmacological Effects

According to the putative identification, some of the compounds that predominantly contributed to the pharmacological effects of the 4TP and 5TP fractions were fatty acids and derivatives (octanoic acid, oleic acid, palmitoleoyl ethanolamide, behenic acid, lignoceric acid, and cerebronic acid), triterpenoid (camelledionol), retinoid (isoacritretin), piperazine (1-piperazinecarbodithioic acid), coumarin (pectachol), acetate salt (sodium diacetate), and lysophosphatidylethanolamine. In contrast, in the 8TP fraction, the compounds contributing to pharmacological effects were mostly amino acids and derivatives (L-proline, glycyl-glycine, and β-alanine betaine), retinoid (isoacritretin), bicyclic monoterpenoid (piperochromenoic acid), sesquiterpenoid (gamma-eudesmol rhamnoside), triterpenoid (spirosta-3,5-diene), hydroxy coumarin (gerberinol), cholesterol ester and lysophosphatidylcholine (Figure 4a, Table S1, and Figure S1). In addition, the putative identification of the compounds that exhibited the highest VIP scores from the PLS-DA showed that at least seven compounds, fatty acids (behenic acid, lignoceric acid, and cerebronic acid), bicyclic monoterpenoid (piperochromenoic acid), piperazine (1-piperazinecarbodithioic acid), triterpenoid (spirosta-3,5-diene), and acetate salt (sodium diacetate), were correlated with the pharmacological effects of the fractions (Figure 4a,b, Tables S1 and S2, and Figure S1). The other compounds identified among those found in the PLS-DA to have the highest VIP scores were choline, 2-succinylbenzoate, variabilin, chlorogenic acid and isochlorogenic acid b, among others (Figure 4b, Table S2, and Figure S1).

2.3.5. Targeted Metabolomics in Fractions

Based on the untargeted analysis, phenolics targeted metabolomics allowed for the identification of a total of four hydroxybenzoic acids (4-hydroxybenzoic acid, gallic acid, gentisic acid, and protocatechuic acid), one caffeoylquinic acid (chlorogenic acid), two flavonoids (kaempferol 3-O-glucoside and quercetin 3-glucoside) and L-phenylalanine amino acid (Figure 5a), which were detected and quantified in the 4TP, 5TP, and 8TP fractions (Figure 5b and Table S3). Some compounds showed concentration differences among the three fractions (Figure 5c). However, the most abundant compound was chlorogenic acid, and the concentration was greater in the 5TP fraction (37,902.8 µg/gF) than in the 4TP and 8TP fractions (4204.7 and 4119.2 µg/gF, respectively); the rest of the compounds were detected at low concentration in the three fractions, except for phenylalanine, which was detected at concentrations of 119.14, 22.64 and 3.22 µg/gF for 8TP, 5TP and 4TP, respectively (Table S3). According to the correlations between phytochemical and pharmacological effects and compound abundances (Figure 3c), chlorogenic acid (rt_m/z of 23.916_353.3427) abundance was strongly positively correlated with the TPC, TPAC, IC50, and antibacterial effect against E. coli (0.97, 0.96, 0.96 and 0.85, respectively), and it was moderately positively correlated with the cytotoxic effect (0.61) and antibacterial effect against S. aureus (0.44) (Figure 3c).

3. Discussion

3.1. Fractions Obtained from Tanacetum parthenium Root Culture Showed Increased Total Secondary Metabolite Contents Regarding Crude Extracts

The results obtained in this work highlight that the root culture of T. parthenium represents a source of several secondary metabolites, exhibiting that a wide range of pharmacological activities and combining conventional and cutting-edge approaches such as untargeted metabolomics allowed for the identification of some of the phenolic compounds and terpenoids responsible for those effects. Although targeted metabolomics contributed to confirming the putative identification of these compounds, most focused on phenolic compounds because they exhibited high values in phytochemical analysis, and showed pharmacological effects (Table 1). Several reports on tissue cultures of T. parthenium have been published on targeted metabolomics for PTN since its production is influenced by genotype, type of organ (higher in aerial parts), developmental stage, cultivation, and environmental factors [32,39,40,41]. Several reports of the in vitro cultures of this species have demonstrated that PTN production depends on the type of culture (higher in organ cultures than in cell cultures), carbon source, and plant growth regulator (PGR) [34,42]. Other reports have focused on detecting other secondary metabolites, such as those in hairy roots culture, where the production of spiroketalenolethers [32] and PTN [31] was described. In callus cultures, phenolic compounds (scopoletin and isofraxidin coumarins) [33] or terpenoids, such as PTN [32,34], sitosterol and stigmasterol [30], and germacrene D [31], were reported; however, in those works, pharmacological assays and untargeted metabolomics, were not carried out, limiting the understanding of the full pharmacological potential and the identification of unknown bioactive compounds.
Moreover, testing fractions from a raw extract for pharmacological effects implemented in this work increases the total secondary metabolite content, which enhances pharmacological effects compared to previous reports about whole plants or tissue cultures. In this work, the highest values determined for total secondary metabolites were as follows: TPC, 194.2 mg GAE/gF; TPAC, 143.6 mg VBE/g; TFC, 11.9 mg QE/gF; and TSLC, 14.7 mg PTNE/g. For instance, in T. parthenium grown in a greenhouse and open field, under adequate-water conditions, the TPC (64.7 and 110.5 mg GAE/g, respectively) in the aerial parts was greater than that observed under reduced watering (33.45 and 93.85 mg GAE/g, respectively) [39]. The amount of TPC reported in the essential oil of T. parthenium was 152.2 mg GAE/g, while the TFC was 70.2 mg QE/g [43]. An 80% alcohol extract of the golden variety of T. parthenium had a TPC of 21.21 mg GAE/g, which was higher than that reported for T. vulgare; this extract showed a strong antioxidant effect and was potentially attributed to luteolin; PTN [44]; and caffeoylquinic acids, such as 3,5-dicaffeoylquinic acid, 3,4-dicaffeoyl-quinic acid, and 4,5-dicaffeoyl-quinic acid (comprising isochlorogenic acid A, B and C, respectively) [44]. The TPC of the shoots and roots of the in vitro plantlets changed after 32 days of culture, and it was enhanced when the plantlets were grown in PGR-enriched culture media (0.27 µM α-naphtalenacetic acid with 2.32 µM kinetin); the greatest TPC (62.54 mg GAE/g) was observed in the shoots from the plantlets treated with PGR-enriched culture for 10 days [35]. In a root culture, the TPC also changed after 32 days of culture, and the highest value (42.52 mg GAE/g) was determined at the end of the experiment (32 days) [35]. The PTN concentration in the plant material of T. parthenium maintained in vitro was 5 × 10−3% in long-term cell culture, while in leaves from regenerated cultured shoots, it was 1.08%; moreover, in glasshouse-grown plant material, it was 2.77% in apical leaves, 2.3% in mature flowerheads, and 6.5 × 10−3% in roots [32]. The hydroethanolic extract from the different organs (aerial parts, flower heads, and leaves) of T. parthenium had different phytochemical profiles. The highest total content of several secondary metabolites, such as polyphenols (67.41 mg GAE/mL), flavonoids (19.33 mg catechin equivalent/mL), and phenolic acids (5.10 mg caffeic acid equivalent/mL), as well as the highest antioxidant potential, were observed in the flowers. In contrast, the extract of aerial parts had the highest content of condensed tannins (22.89 mg delphinidin/mL) and terpenoids (54.41 mg linalool/mL), and also exhibited the most effective antimicrobial activity [45]. In addition, an HPLC analysis showed that caffeoylquinic acids (e.g., chlorogenic acid at 3.98 mg/mL; isochlorogenic acids A, B, and C) were present at higher concentrations (5.33, 1.28 and 0.59 mg/mL, respectively) than flavonoids (apigenin, quercetin, santin, and kaempferol-3-rutinoside at 0.05, 0.06, 0.32, and 0.4 mg/mL, respectively) [45].

3.2. Fractions Obtained from Tanacetum parthenium Root Culture Exhibited Enhanced Pharmacological Activity as Compared to Crude Extracts

Regarding the antioxidant effect, in the present study, IC50 of 2.3, 6.11, and 14.84 µg/mL were achieved for the 8TP, 5TP and 4TP fractions, respectively, indicating a better activity than that reported for the extracts. For instance, an IC50 of 33.22 µg/mL has been reported for the 70% methanol extract of T. parthenium using the radical DPPH, from which phenolic acids (ferulic acid) and flavonoids (apigenin, luteolin, luteolin-7-O-glucoside, kaempferol, and chrysophanol) were isolated and determined to be potent antioxidant agents (IC50 values from 6.44-16.23 µM) based on the DPPH assay [46]. The essential oil of T. parthenium had an IC50 of 30.23 µg/mL according to the DPPH assay; it was suggested that the high DPPH scavenging activity was attributed to the content of monoterpenes and sesquiterpene alcohols, e.g., camphor (the most abundant at 43.97%), chrysanthenyl acetate, germacrene D and B, and farnesol [43]. The 80% alcohol extract of the golden variety of this species showed a strong antioxidant effect (84% of DPPH free radical-scavenging activity) that was potentially attributed to luteolin; PTN [44]; and caffeoylquinic acids, such as isochlorogenic acid B and C [44]. The superior antioxidant capacity of the fractions examined in this study highlights their relevance for future applications.
The α-amylase inhibitory effect of extracts obtained from species belonging to the Tanacetum genus has been reported. For example, the methanol and ethyl acetate extracts of different organs (leaf + stem, capitulate, and herb) of T. haussknechtii showed from 263.3 to 320.7 mg of acarbose equivalents per gram of extract (mg AE/g) in the methanolic extract, and 258.8 to 356.9 mg AE/g in the ethyl acetate extract. The greatest inhibitory effect (356.9 mg AE/g) determined in capitula was attributed to caffeoylquinic acid and flavonoids, which are reported to have antidiabetic effects [47]. Similarly, the type of extract obtained from the aerial parts of T. poteriifolium influenced α-amylase inhibition, with the greatest inhibitory effect (0.7 mmol AE/g ~451.9 mg AE/g) observed in the ethyl acetate extract compared to that in the MeOH (0.51 mmol AE/g ~322.8 mg AE/g) and water (0.10 mmol AE/g ~64.5 mg AE/g) extracts; this inhibitory effect was correlated with TFC (0.91), although this effect was observed as poor [48]. The type of organ (flower head, aerial parts and roots) of T. macrophyllum and T. speciosa influenced the inhibitory effect of the 80% MeOH extract on α-amylase; in both species, the greatest inhibitory effect was detected for flower heads (0.65 mmol AE/g ~419.6 mg AE/g for T. macrophyllum, and 0.58 mmol AE/g ~374.4 mg AE/g for T. speciosa) [49]. Additionally, the type of organ (flowers, stems, and aerial parts) of T. vulgare and its extracts (prepared with hexane, EtOH/water, and infusion) influenced α-amylase inhibition and the highest effect was detected in the hexane extracts of flowers and stems (0.53 and 0.50 mmol AE/g ~342.1 and 322.8 mg AE/g; the authors argued that this effect might be attributed to phenolics and sesquiterpene lactones [50]. Finally, the inhibition of α-amylase was not affected by the extraction method (maceration, Soxhlet extraction, ultrasound, microwave extraction, and accelerated extraction) with ethanol used in the aerial parts of T. parthenium; the inhibitory effect ranged from 0.51 to 0.56 mmol AE/g ~329.2 to 361.5 mg AE/g [48]. In the present study, the enzyme inhibition levels shown by all the fractions were 23.36, 12.12, and 4.80 mg AE/gF for 8TP, 4TP, and 5TP, respectively, which are capable of inhibiting the enzyme at 10.15, 5.27, and 2.09% at 10 μg/mL, respectively; these values were lower than the IC50 of acarbose (2.48 ± 0.31 μg/mL). However, in previous reports, the α-amylase inhibition activity of the samples was expressed as the acarbose equivalent per gram, but data about that percentage were not shown. Enzyme inhibition is considered a therapeutic strategy for managing type II diabetes, where α-amylase is a blank target because it is related to the breakdown of sugars in the intestine [48].
Moreover, T. parthenium has been proven to have antibacterial effects against E. coli and S. aureus, which was also observed in the present work, where the fractions of methanolic extract provoked the highest inhibition of 27.02% in E. coli after treatment with fraction 4TP at 6 µg per disk and approximately 18% in S. aureus after the exposure to the three fractions tested at 8 µg per disk. While the essential oil of T. parthenium has a proven antibacterial effect, it had different results among the types of bacteria tested. An inhibition rate of 89.44% was shown for S. aureus (at a 20 µL dilution with ethanol at a 1:5 ratio, applied per disk), while against E. coli, no inhibition was observed [26]; the authors attributed this effect to the presence of camphor. Similarly, Ghavam [30] reported an inhibition rate of 38.8% in S. aureus with 10 µL of essential oil per well, but against E. coli, there was no inhibition effect. In another study, Shafaghat et al. [29] demonstrated that the essential oil of T. parthenium, at 30 µL per disk, inhibited the growth of E. coli by 68.10%, while for S. aureus, it inhibited growth by 89.44%, and the authors attributed the effect to the presence of the oxygenated compounds. Izadi et al. [28] reported 64.91% growth inhibition for S. aureus, 68.42% growth inhibition for E. coli ATCC 25923, and 74.46% growth inhibition for E. coli ATCC 157 using essential oil at 2.5 µL per disk. Polatoğlu et al. [51] reported a minimum inhibitory concentration (MIC) of 125 µg essential oil/mL for E. coli and S. aureus.
Several species of the Tanacetum genus have demonstrated cytotoxic effects on cancer cell lines. The methanol extracts of T. macrophyllum, T. vulgare, and T. corymbosum inhibited cell growth by 69.87, 77.68, and 93.71%, respectively, at 200 µg/mL in HeLa cells (human cervical cancer) after 24 h of exposure [52]. The IC50 of the T. erzincanense methanol extract in the MCF-7 (breast adenocarcinoma), HepG2 (hepatocellular carcinoma), and HT-29 (human colorectal adenocarcinoma) cell lines were 41.3, 46.9, and 63.8 µg/mL, respectively, after 48 h of exposure; in this extract the TPC was 64.4 mg GAE/g and the TFC was 62.2 mg QE/g; and both had high positive correlations (0.795 and 0.671, respectively) with cytotoxic activity [53]. The IC50 of the T. vulgare leaf methanol extract in the HeLa cell line after 72 h of exposure was approximately 50 µg/mL; this cytotoxic activity was attributed to its phenolic acid content [54]. The IC50 for PTN after 48 h of exposure was reported to be 1.26 µg/mL for the Raji cell line (Epstein–Barr virus-positive lymphoma) [55], 2.1 µg/mL for SiHa (human cervical cancer) cells, and 2.4 µg/mL for MCF-7 cells. Another study determined an IC50 of 5 µg/mL after 72 h of exposure to PTN in the MT2 cell line (lymphoma cells infected with human T-cell leukemia virus type 1) [56]. In the present study, the three fractions showed cytotoxic effects on the RA-1 cell line (human B non-Hodgkin lymphoma), with IC50 values of 26.37, 30.91 and 30.30 µg/mL for 5TP, 4TP and 8TP, respectively. These data revealed that the fractions obtained in this work had better cytotoxic effects than the complete extracts of other species of the Tanacetum genus; however, compared to those of PTN, the fractions obtained in this study had lower cytotoxic activity. In addition, the results of the present work, in which the fractions were combined with 1 nM vincristine (added 6 h before completing the 24 h incubation) might cause chemosensitization because the cytotoxic effect of the fractions alone was enhanced, provoking IC50 values of 16.71, 17.88 and 18.45 µg/mL, for 5TP, 4TP, and 8TP, respectively. Chemosensitizing can occur due to the synergistic or additive effect of the secondary metabolite profile, which can be greater than the sum of the individual actions given by different individual action mechanisms [57].

3.3. Correlations Among Pharmacological Activities, Phytochemical Profiles, and Untargeted Metabolomics of Fractions Enabled the Identification of Metabolites

For this work, determining the correlation among pharmacological activities, phytochemical characterization, and untargeted metabolomics proved useful for identifying secondary metabolites, such as phenolic compounds, mainly phenolic acids and terpenoids, that are strongly associated with the pharmacological activity of the three fractions. Additionally, this approach allows for the detection of other metabolites potentially participating in biological effects, such as fatty acids (Table 1, Tables S1 and S2). Although T. parthenium has been described as a promising source of bioactive phytochemicals related to its medicinal uses, its chemical composition is limited [58]. Recent studies have reported large amounts of succinic, palmitic, linoleic, and linolenic acids (4.89, 2.15, 1.71, and 1.07 mg/g, respectively) and benzoic acid (1.89%) [58]. Furthermore, Rezaei et al. [44] reported thirteen fatty acids, such as palmitic acid (the most abundant at 57.27%), myristic acid, lauric acid, capric acid, palmitoleic acid, oleic acid, linoleic acid, linolenic acid, arachidic acid, behenic acid, erucic acid, and lignoceric acid, in the essential oil of T. parthenium. This species has also been recognized as a source of sesquiterpenes, monoterpenes, flavonoid glycosides (e.g., quercetin, 6-hydroxykaempferol 3,6-dimethyl ether, and 6-methoxykaempferol 3-methyl ether), triterpenoids (e.g., oleanolic acid methyl ester), phytosterols closely resembling cholesterol (e.g., campesterol, fucosterol, β-sitosterol, and stigmasterol), fatty acids (e.g., palmitic, myristic, lauric, linoleic, and capric acids), phenolic acids (e.g., isochlorogenic, chlorogenic, syringic, ferulic, sinapic, vanillic, and p-coumaric acids), coumarins (e.g., isofraxidin and 9-epipectachol B), and pyrethrin, among others [20]. In this work, several of those metabolites were identified (Table 1, Tables S1 and S2), and they have been credited in the literature for their biological effects (Table 1), supporting the results observed through the analysis carried out in this research. In addition, the metabolites that exhibited the highest VIP scores from the PLS-DA could be used as chemical biomarkers of the fractions, especially the compounds that were also correlated with pharmacological effects, such as chlorogenic acid, which might be useful for quality control for studying and developing drugs, nutraceuticals, or standardized extracts, among others, from the three fractions.
Table 1. Pharmacological activities reported in the literature for metabolites identified in the three fractions obtained from the methanolic extract of the root biomass of T. parthenium culture.
Table 1. Pharmacological activities reported in the literature for metabolites identified in the three fractions obtained from the methanolic extract of the root biomass of T. parthenium culture.
CompoundPharmacological ActivityReference
Biological EffectMICIC50% Inhibition
4-hydroxybenzoic acidAntibacterial against E. coli125 µg/mL [59]
Antibacterial against S. aureus62.5 µg/mL
α-Amylase inhibition 3.552 mg/mL [60]
Antioxidant 321.72 µg/mL for DPPH [59]
Cytotoxic 20.8 µg/mL in MCF-7
Behenic acidCytotoxic 7.52 µM in HepG2 [61]
11.86 µM in MCF-7
12.28 µM in PC3
Antioxidant ˃1 mg/mL for DPPH [62]
Chlorogenic acidAntibacterial against E. coli6 mg/mL [63]
Antibacterial against S. aureus3 mg/mL
α-Amylase inhibition 9.1 µg/mL [64]
Antioxidant 51.23 µg/mL for DPPH
Cytotoxic 52% at 100 µM in 2OS [65]
27% at 100 µM in MG-63
Gallic acidAntibacterial against E. coli1 µg/mL [66]
Antibacterial against S. aureus1 µg/mL
α-Amylase inhibition 1.09 µg/mL [67]
Antioxidant 5.73 µM for DPPH
Cytotoxic 50 µM in SMMC-7721 [68]
80 µM in HL-60
4 µM in k562
40 µM in Wehi231
80 µM in HeLa
Gentisic acidAntibacterial against E. coli4 mg/mL [69]
Antibacterial against S. aureus4.15 mg/mL
α-Amylase inhibition 2.07 mg/mL [70]
Antioxidant 7.6 µM for DPPH [71]
Cytotoxic 14 mM in HepG2 [69]
Isochlorogenic acid bAntioxidant EC50 9.4 µg/mL for DPPH [72]
Kaempferol-3-O-glucosideAntibacterial against E. coli1.25 µg/mL [73]
Antibacterial against S. aureus0.625 µg/mL
Antioxidant1.25 µg/mL for DPPH
Lignoceric acidAntibacterial against E. coli˃1 mg/mL [62]
Antibacterial against S. aureus˃1 mg/mL
Antioxidant ˃1 mg/mL for DPPH
Oleic acidAntibacterial against E. coli0.512 mg/mL
Antibacterial against S. aureus0.256 mg/mL
Antioxidant 0.5 mg/mL for DPPH
Protocatechuic acidAntibacterial against E. coli2.5 mg/mL [63]
Antibacterial against S. aureus0.45 mg/mL [74]
α-Amylase inhibition 1.12 µg/mL [67]
Antioxidant 8.29 µM for DPPH
Cytotoxic 55% at 8 µmol/L in MCF-7 [75]
60% at 8 µmol/L in A549
45% at 8 µmol/L in HepG2
42% at 8 µmol/L in HeLa
65% at 8 µmol/L in LNCaP
Quercetin-3-glucosideAntioxidant 2.39 mM TEAC [76]
Cytotoxic 70.44% at 50 µg/mL in HeLa[77]
Sodium diacetateAntibacterial against E. coli0.31% (w/v) [78]
Antibacterial against S. aureus0.31% (w/v)
VariabilinCytotoxic 87.74 µM in PC3 [58]
38.08 µM in MCF-7
˃100 µM in HT-29
DPPH, 2,2-Diphenyl-1-picrylhydrazyl (DPPH data shown in this table are related to the corresponding assay); EC50, half maximal effective concentration; IC50, half maximal inhibitory concentration; MIC, minimum inhibitory concentration; TEAC Trolox equivalent antioxidant capacity. The cancer cell lines described in this table are: A549 human lung cancer; C6 glioma; CaCo-2 human colorectal cancer; CAL27 epithelial cell line isolated from tongue; Calu-6 human lung cancer; CEM leukemia; H3255 lung cancer; HCT-116 human colorectal cancer; HeLa cervical; HepG2 hepatocellular carcinoma; HL-60 leukemia; HSC-2 oral squamous cell carcinoma; HSG human salivary gland adenocarcinoma; HT-29 colorectal cancer; k562 leukemia; L1210 leukemia; LNCaP human prostate cancer; MCF-7 breast cancer; MG-63 osteosarcoma; PC3 human prostate cancer; SMMC-7721 hepatoma; U2OS osteosarcoma; U937 leukemia; Wehi231 lymphoma.

3.4. Future Perspectives of the Findings

Medicinal plants are a source of natural compounds that possess diverse chemical structures and are important for the pharmaceutical industry to produce food supplements and cosmetics, among other products. Moreover, they are bioactive principles for complementary and alternative medicines; thus, the identification of key chemical biomarkers (active or purely analytical) and quality confirmation must be carried out [79]. Given that the 4TP fraction described in this work showed an outstanding abundance of fatty acids, it could be tested for applications in the food and cosmetic industries [80].
Since the 1960s, the industrial production of phytochemicals through plant tissue culture has been examined worldwide. Interest in commercial application has promoted extensive research on the production of different types of secondary metabolites as potential drugs. However, despite those efforts in research, only a few commercial applications have been achieved [81]. Thus, different strategies, such as screening productive cell lines, implementing cultures in bioreactors, eliciting secondary metabolism, hairy root culture, micropropagation, biotransformation, and metabolic engineering [81], have been developed to increase in vitro productivity and improve the efficiency of secondary metabolite production. In this work, plant tissue culture combined with analytical techniques related to untargeted and targeted metabolomics was demonstrated to represent an alternative source for researching cell lines that can produce several bioactive compounds that may be identified as key chemical biomarkers. Further research must be carried out on the fractions obtained from the methanolic extract of the root culture of T. parthenium, particularly for 4TP, because of the great pharmacological effects that were exerted, such as increasing the productivity and efficiency of producing the pharmacologically active metabolites, e.g., chlorogenic acid, since it was determined that this metabolite was detected at high concentration. Chlorogenic acid has been reported to be distributed among different species; some of the main sources of this compound are green coffee (7–8 g/100 g for Coffea canephora and 4–6 g/100 g for C. arabica) [82], apple (0.41–1.16 mg/g), prunes (1.3–3.9 g/100 g), tomato (21.30–240.16 µg/g), and carrot (0.3–18.8 mg/g) [82]; in this work, the 5TP fraction was 37.9 mg/g. This secondary metabolite can produce several biological effects, such as antioxidant, anti-inflammatory, liver and kidney protector, antibacterial, antitumor, nervous system protector, and sugar and lipid metabolism regulator effects. Due to its biological activities, it has received great attention, mainly as an antioxidant for medicinal and food purposes [82]. In addition, the concentration of isochlorogenic acid may also be high due to the correlations established between abundance and phytochemical and pharmacological effects for this compound (rt_m/z of 5.599_515.1196); its high positive correlations with TPC, TPAC, and IC50 and antibacterial effect against E. coli (0.97, 0.96, 0.96, and 0.85, respectively) and its moderate positive correlations with cytotoxic effect (0.62) and antibacterial effect against S. aureus (0.43) (Figure 3c). However, further research on the fractions obtained from the methanolic extract of the root culture of T. parthenium is needed to validate the putative identification of these compounds detected through untargeted metabolomics and to quantify them.

4. Materials and Methods

4.1. Root Biomass Production

Half-strength Murashige and Skoog culture media for the cultivation of suspended roots were prepared by adding 40 g/L D-glucose (Sigma-Aldrich, St. Louis, MO, USA), 150 mg/L ascorbic, 100 mg/L citric acid (Sigma-Aldrich, St. Louis, MO, USA), and 3 mg/L α-naphtalenacetic acid (Sigma-Aldrich, St. Louis, MO, USA) [36]. The culture medium pH was adjusted to 5.8 and then sterilized by autoclaving at 121 °C for 18 min. Roots (12 g fresh weight) were inoculated in a 1000 mL Erlenmeyer flask with 200 mL of liquid culture medium; the resulting root culture was placed in an orbital shaker at 110 rpm, and incubated at 25 ± 2 °C under white fluorescent light (50 µmol m−2 s−1) under a 16 h/8 h light/dark photoperiod for 32 days. The harvested biomass was filtered with a vacuum pump, rinsed with distilled water, and dried in an oven for 24 h at 60 °C.

4.2. Obtaining Fractions and Phytochemical Analysis

4.2.1. Methanolic Extraction from Root Biomass

Dried root biomass (3 g) was extracted with 500 mL of methanol (MeOH) for 30 min in an ultrasonic homogenizer (Ningbo YinZhou Sjia Lab Equipment Co., LTD, model SJIA-950 W, Ningbo, China) configured at 20 kHz and 50 °C, operating for 9 s on and 6 s off. The extract was filtered using a vacuum pump and filter paper and then concentrated on a rotary evaporator (Büchi Labortechnik AG, R-300, Flawil, Switzerland) until dry.

4.2.2. Fractionation of Methanolic Extract

Fractionation was performed on a chromatographic column employing silica gel (mesh size 60–200, Sigma-Aldrich, St. Louis, MO, USA) as the stationary phase, and the polarity was increased by mixing (A) acetonitrile and (B) MeOH. Ten different fractions were obtained with a volume of 100 mL each. An aliquot of every fraction (1 mL) was qualitatively analyzed with Baljet, ferric chloride, and sodium hydroxide tests to detect sesquiterpene lactones, phenolic compounds, and flavonoids, respectively [83]. The fractions with the most visually perceptible results (coded as 4TP, 5TP, and 8TP) were chosen for quantitative phytochemical analysis and for in vitro assays. The selected fractions (4TP, 5TP, and 8TP) were concentrated in a rotary evaporator (Büchi Labortechnik AG, R-300, Flawil, Switzerland) until almost dry and brought to dryness in an oven at 40 °C for 24 h.

4.3. In Vitro Assays of Selected Fractions

4.3.1. Antioxidant Assay

The half maximal inhibitory concentration (IC50) of free radical scavenging was determined with 2,2-diphenyl-1-picrylhydrazyl (DPPH) radical to evaluate the antioxidant effect of the 4TP, 5TP, and 8TP fractions according to the methods of Nieto-Trujillo et al. [83]. A 0.1 mM methanolic solution of DPPH (Sigma-Aldrich, St. Louis, MO, USA) was prepared. Moreover, three drops of dimethyl sulfoxide (DMSO; Sigma-Aldrich, St. Louis, MO, USA) were added to each fraction, which was subsequently dissolved in MeOH to prepare different concentrations (0.1–10 mg/mL). The assay involved the combination of 1.8 mL of 0.1 mM DPPH with 0.3 mL of different concentrations of each fraction or MeOH (a blank control of the fractions). This mixture was incubated for 15 min in the dark at room temperature. Finally, the absorbance was measured at 515 nm in a UV-Vis spectrophotometer (Thermo Scientific, model Evolution 60S, Waltham, MA, USA). Every measurement was made in triplicate (n = 3). The results are expressed as the amount (in µg/mL) of the fraction capable of inhibiting the DPPH radical at 50% (IC50).

4.3.2. Antibacterial Assay

The assay consisted of the Kirby–Bauer method and was tested via disk diffusion against Staphylococcus aureus (ATCC 25923) and Escherichia coli (ATCC 25922). For both strains, the bacterial inoculum was prepared in 5 mL of trypticase soy broth (Becton, Dickinson and Company, Sparks, MD, USA), adjusted to 0.5 on the McFarland scale to 1 × 106 CFU/mL, and incubated at 37 °C for 24 h. Subsequently, 80 μL of inoculum was spread onto Müller–Hinton agar (Becton, Dickinson and Company, Sparks, MD, USA) plates using an L-shaped plastic rod, and sterile filter paper disks were placed. The fractions (4TP, 5TP, and 8TP) were dissolved in 3% (v/v) DMSO (Sigma-Aldrich, St. Louis, MO, USA) in MeOH to prepare solutions at concentrations of 100, 200, 400, 600 and 800 μg/mL; 10 µL of each solution was added per disk to achieve concentrations of 1, 2, 4, 6, and 8 μg/disk. Vancomycin (Sigma-Aldrich, St. Louis, MO, USA) and chloramphenicol (Sigma-Aldrich, St. Louis, MO, USA) (1 μg/disk) were used as positive inhibitory controls for S. aureus and E. coli, respectively, while 3% DMSO (Sigma-Aldrich, St. Louis, MO, USA) in MeOH was used as a blank control for the fractions, while sterile water was used as a negative control. The plates were incubated at 37 °C for 24 h, after which the inhibition halo was measured in mm using a Vernier caliper. This trial was performed with seven repeats per triplicate for each concentration against each bacterial strain (n = 21). The data are shown as a percentage of inhibition according to the following formula [83]:
%   i n h i b i t i o n = Z H I   o f   f r a c t i o n     Z H I   o f   n e g a t i v e   c o n t r o l Z H I   o f   p o s i t i v e   c o n t r o l     Z H I   o f   n e g a t i v e   c o n t r o l     100
where ZHI corresponds to zone halo inhibition in mm.

4.3.3. Cytotoxicity Assay

The human B non-Hodgkin lymphoma (Burkitt) cell line (Ramos RA-1) (ATCC, CRL-1596) was cultured in RPMI 1640 Advanced medium (Invitrogen®, Waltham, MA, USA) supplemented with 5% fetal bovine serum (FBS; Invitrogen®, Waltham, MA, USA), 1% antibiotic–antifungal mixture containing 10,000 U/mL penicillin G, 10 mg/mL streptomycin, and 25 μg/mL amphotericin B (Invitrogen®, Waltham, MA, USA). A total of 50,000 cells per well were cultured in 96-well plates with Advanced-RPMI 1640 medium and 2% FBS. The different fractions were dissolved in 0.01% (v/v) DMSO (Sigma-Aldrich, St. Louis, MO, USA) in culture media at final concentrations of 1, 5, 10, 20, 30, and 40 μg/mL, alone or in combination with 1 nM vincristine (Sigma-Aldrich, St. Louis, MO, USA). The controls for this assay consisted of 0.01% (v/v) DMSO in culture medium (negative control) and 1 nM vincristine (positive control). 50 μL of cells and 50 μL of fraction/control solution were inoculated into each well. The cell-inoculated plates were incubated for 24 h at 37 °C and 5% CO2. Treatments consisting of the combination of vincristine with the fraction, the vincristine solution was added 6 h before 24 h of incubation. Afterward, the exclusion staining method was used to count the dead and living cells. Thus, 20 μL was taken from each well of the plate, previously resuspended, and mixed with 80 μL of trypan blue (Invitrogen®, Waltham, MA, USA); then, 10 μL was placed on each side of a Neubauer chamber, and viable cells were counted on an inverted microscope (Olympus IX73, Evident Scientific, Inc., Waltham, MA, USA) with a 40× objective. The resulting data were used to estimate the cellular viability, expressed as a percentage of viable cells relative to total cells according to the following formula [83]:
C e l l u l a r   v i a b i l i t y   ( % ) = N u m b e r   o f   t o t a l   c e l l s     N u m b e r   o f   d e a d   c e l l s N u m b e r   o f   t o t a l   c e l l s     100
In addition, for each fraction, the results of this assay were used to determine the IC50 value using the equation for calculating the percentage of cell viability versus the concentration tested.

4.3.4. α-Amylase Inhibitory Assay

The procedure consisted of testing the inhibitory effect of the selected fractions at different concentrations on α-amylase enzyme activity, according to Nieto-Trujillo et al. [83]. For this assay, 2 U of porcine α-amylase enzyme (Sigma-Aldrich, St. Louis, MO, USA) was dissolved in phosphate buffer solution (PBS); 0.5% starch (Sigma-Aldrich, St. Louis, MO, USA) was prepared in distilled water; and reactive dinitrosalicylic acid (DNSA; Sigma -Aldrich, St. Louis, MO, USA) was also used. The different fractions were dissolved in 0.01% (v/v) DMSO and distilled water to obtain 10 μg/mL solutions. Acarbose (Sigma-Aldrich, St. Louis, MO, USA) was used as a positive control, and distilled water was used as a negative control. The absorbance was measured at 540 nm in a UV-Vis spectrophotometer (Thermo Scientific, model evolution 60S, Waltham, MA, USA) using water as a blank. The absorbance results are expressed as a percentage of inhibition according to the following formula [83]:
% I n h i b i t i o n = b a b     100
where:
a = A b s o r b a n c e   T 1     ( A b s o r b a n c e   T 2     A b s o r b a n c e   T 3 )
b = A b s o r b a n c e   T 4 A b s o r b a n c e   T 5
The treatments related to variables coded T1 to T5 are shown in Table S4.
In addition, different water solutions of acarbose (0.1, 0.65, 1.25, 2.5, 3.5 and 5 μg/mL) were prepared to construct a calibration curve (y = 15.141x + 12.316, R2 = 0.9408), from which the IC50 was estimated. This calibration curve was used to express the results of the α-amylase inhibition as mg of acarbose equivalents per gram of fraction (mg AE/gF) [83].

4.4. Phytochemical Analysis of the Selected Fractions

Total Secondary Metabolite Contents

The total contents of phenolic compounds, phenolic acids, flavonoids, and sesquiterpene lactones (TPC, TPAC, TFC, and TSLC, respectively) were determined according to the methods of Nieto-Trujillo et al. [83] using gallic acid, quercetin, and verbascoside standards (Sigma-Aldrich, St. Louis, MO, USA), respectively, to construct the calibration curves. The results are expressed as milligrams of gallic acid, quercetin, or verbascoside equivalents per gram of fraction (mg GAE/gF, mg QE/gF, or mg VBE/gF), respectively. TSLC was conducted according to Salapovic et al. [84] using PTN as a standard (Sigma-Aldrich, St. Louis, MO, USA). The TSLC results are expressed as milligrams of PTN equivalents per gram of fraction (mg PTNE/gF). The absorbance measurements were carried out in triplicate (n = 3) using a UV-Vis spectrophotometer (Thermo Scientific, model evolution 60S, Waltham, MA, USA).

4.5. Metabolomics of Fractions

4.5.1. Untargeted Metabolomics

The 4TP, 5TP, and 8TP fractions were analyzed via liquid chromatography coupled with mass spectrometry (LC-MS) to perform untargeted metabolomics. The LC-MS system consisted of a 1290 infinity Agilent ultrahigh resolution liquid chromatograph (UPLC) coupled to a 6545 Agilent quadrupole time-of-flight mass spectrometer (Q-TOF/MS) (Agilent Technologies, Santa Clara, CA, USA). Chromatographic separations were performed on an ACQUITY UPLC BEH C18 column (1.7 µm, 2.1 × 50 mm; Agilent Technologies, Santa Clara, CA, USA). The mobile phase consisted of (A) water with 0.1% formic acid and (B) MeOH with 0.1% formic acid, both of which were of mass spectrometry grade. The gradient elution profile was as follows: 90% in the first 2 min A, 2–10 min 90–40% A, 10–15 min 40% A, 15–20 min 40–10%, 20–25 min 10% A, 25–27 min 10–90% A, and 27–31 min 90% A. The flow rate was set to 0.4 mL/min. The column temperature was 40 °C. The injection volume was 10 µL. All samples were analyzed with three replicates. The mass spectrometer used a dual AJS ESI ion source. The ESI source was operated in negative and positive mode with a gas temperature of 300 °C, cone gas (N2) flow rate of 10 L/min, nebulizer pressure of 20 psi, sheath gas temperature of 300 °C, sheath gas flow rate of 10 L/min, and skimmer offset of 65 V with a fragmentor of 120 V. The mass spectrum data were acquired with a mass range from 100 to 3000 m/z. Multiple mass spectrophotometric scanning modes, including full scanning, product ion scanning, and positive/negative scanning were used for qualitative analysis. MS/MS experiments were performed to determine fragmentation patterns by means of the acquisition mode untargeted MS and a fixed collision energy of 15 eV. During this analytical study, quality control samples (seven reference masses) were analyzed to assess the variance observed in the acquired data.
Chromatographic data were processed using MassHunter Qualitative Analysis Software (version B.08.00; Agilent Technologies, Santa Clara, CA, USA). The Molecular Feature Extraction algorithm extracted compounds from the raw data. The MetaboAnalyst bioinformatic platform (https://www.metaboanalyst.ca/, accessed on 20 August 2024) was used to analyze the untargeted data via the statistical analysis module; a principal component analysis (PCA), a partial least squares discriminant analysis (PLS-DA), a variable importance in projection (VIP), and a hierarchical clustering heatmap were generated [85]. In addition, the data obtained through untargeted metabolomics analysis (rt_m/z signals and their abundances based on peak intensities) were correlated with the results of the total secondary metabolite contents of the fractions and the in vitro assays by testing a Pearson correlation; those m/z signals correlated with a pharmacological effect were putatively identified with the FooDB Version 1.0 database (https://foodb.ca/, accessed 15 October 2024). The mass spectra of those putatively identified compounds are shown in the Supplementary Material (Figure S1).

4.5.2. Targeted Metabolomics

The confirmatory identification of selected phenolic compounds obtained through untargeted metabolomics was performed using targeted metabolomics. This procedure was performed according to the procedure reported by Monribot-Villanueva et al. [86]. Briefly, each fraction was dissolved in MeOH with 0.1% formic acid at 50 mg/mL, filtered through 0.2 µm PTFE membranes and placed in 2 mL UPLC vials. The samples were analyzed in a 1290 infinity Agilent UPLC coupled to a 6460 Agilent triple quadrupole mass spectrometer (Agilent Technologies, Santa Clara, CA, USA). Chromatographic separations were performed on an ECLIPSE PLUS C18 column (1.8 µm, 2.1 × 50 mm; Agilent Technologies, Santa Clara, CA, USA). The mobile phase consisted of (A) water with 0.1% formic acid and (B) acetonitrile with 0.1% formic acid, both of which were MS grade. The gradient elution profile, regarding the mobile phase A, was as follows: 99–50% in the first 30 min, 30–35 min 50–1%, 35–39 min 1%, 39–40 min 1–99%, and 40–45 min 99%. The flow rate was set to 0.3 mL/min. The column temperature was 40 °C. The injection volume was 2 µL. All the samples were analyzed with three replicates. The mass spectrometer was operated at a gas temperature of 300 °C, gas flow rate of 5 L/min, nebulizer pressure of 45 psi, sheath gas temperature of 250 °C, sheath gas flow rate of 11 L/min, capillary voltage (positive and negative) of 3500 V and nozzle voltage (positive and negative) of 500 V. The analytical conditions for identifying and quantifying phenolic compounds are shown in Table S5. The concentrations of the identified compounds are expressed in µg/g of the dry fraction. In addition, the MetaboAnalyst bioinformatic platform (https://www.metaboanalyts.ca/, accessed on 20 November 2024) was used for building a heatmap through a statistical analysis module to compare the concentrations of all the quantified phenolic compounds among the three fractions.

4.6. Statistical Analysis

The total secondary metabolite contents (TPC, TPAC, TFC, and TSLC) and in vitro assay results (IC50 for the antioxidant assay, percentage inhibition of bacteria and α-amylase, and percentage of viability in RA-1) were statistically analyzed through ANOVA, followed by a multiple media/median comparison test. NCSS software (2007 version) was used for all the ANOVA and Pearson’s correlation analyses, and R 4.4.2 software was used for obtaining heatmaps; in all the statistical analyses, a p < 0.05 was assumed to indicate significant differences.

5. Conclusions

The root culture of T. parthenium is a potential source of several metabolites, such as phenolic acids, fatty acids, coumarins, sesquiterpenoids, and triterpenoids, which are capable of exerting α-amylase inhibition and antioxidant, antibacterial, and cytotoxic effects. Because of the pharmacological effects of these fractions, further research must be carried out to estimate their functionality as a source of bioactive principles in several applications, such as the development of drugs and the formulation of cosmetics and nutraceuticals, among others. The potential of plant tissue culture as a source of several pharmacologically active compounds could be estimated by in vitro bioassays combined with analytical techniques such as untargeted and targeted metabolomics, which might enhance the current limitation in establishing commercial applications of plant tissue culture because it is aimed at producing key bioactive compounds.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms26157209/s1.

Author Contributions

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

Funding

The authors thank the Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCyT) for providing funding through grant number 522545 for the Posdoctoral Fellowship 2020–2022 of A.N.-T. at the Universidad Autónoma del Estado de México with the following projects: “Evaluación de la actividad antibacteriana y antidiabética in vitro de fracciones del extracto metanólico obtenido de una línea cellular de las plantas medicinales Arnica montana, Buddleja cordata y Tanacetum parthenium” and “Fraccionamiento biodirigido de extractos de líneas celulares de Arnica montana, Buddleja cordata y Tanacetum parthenium, a través de la actividad antibacteriana y antidiabética”. In addition, we thank this university for partially supporting the experimental procedures through Project No. 6769/2022CIB: “Establecimiento de cultivos celulares de plantas medicinales en un biorreactor”. Additionally, the authors thank the Consejo Mexiquense de Ciencia y Tecnología (COMECYT) for partially supporting the experimental procedures through Project No. 6415/2021E and FICDTEM-2021-048: “Metabolómica y actividades biológicas de las fracciones resultantes de la purificación parcial de un extracto metanólico, obtenido de cultivos in vitro de plantas medicinales”. Finally, J.A.G.-A. (CVU 43039) thanks CONAHCYT for his fellowship grant (854285) during his sabbatical leave.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data that support the findings of this study are available from the corresponding author on request.

Acknowledgments

M.E.E.-Z. and C.Z.-G. thank the Universidad Autónoma del Estado de México for supporting their sabbatical leave. The authors thank Abigail Ramírez-Cruz, a bachelor’s thesis student in Biotechnology from the Facultad de Ciencias, Universidad Autónoma del Estado de México, for integrating some of her thesis’s results, related to antibacterial activity, into this global work.

Conflicts of Interest

The authors affirm that they have no conflicts of interest regarding the completion of this work and its subsequent publication.

Abbreviations

The following abbreviations are used in this manuscript:
4TP, 5TP or 8TPFractions obtained from the methanolic extract of the root biomass of the T. parthenium culture
AJSAgilent jet stream
ATCCAmerican Type Culture Collection
CFUColony-forming units
DMSODimethyl sulfoxide
DNSADinitrosalicylic acid
DPPH2,2-Diphenyl-1-picrylhydrazyl
EC50Half maximal effective concentration
ESIElectrospray ionization
FBSFetal bovine serum
IC50Half maximal inhibitory concentration
LC-MSLiquid chromatography coupled with mass spectrometry
MeOHMethanol
mg AE/gFMilligrams of acarbose equivalents per gram of fraction
mg GAE/gFMilligrams of gallic acid equivalents per gram of fraction
mg QE/gFMilligrams of quercetin equivalents per gram of fraction
mg VBE/gFMilligrams of verbascoside equivalents per gram of fraction
mg PTNE/gFMilligrams of parthenolide equivalents per gram of fraction
MICMinimum inhibitory concentration
MSMass spectrometry
m/zMass charge signal
PBSPhosphate buffer solution
PCAPrincipal component analysis
PGRPlant growth regulators
PLS-DAPartial least squares discriminant analysis
PTNParthenolide
Q-TOF/MSQuadrupole time-of-flight mass spectrometer
rtRetention time in minutes
TFCTotal flavonoids content
TPACTotal phenolic acid content
TPCTotal phenolic compound content
TSLCTotal sesquiterpene lactone content
UPLCUltrahigh resolution liquid chromatograph
VIPVariable importance in projection
ZHIZone halo inhibition

References

  1. Twaij, B.M.; Jazar, Z.H.; Hasan, M.N. Trends in the use of tissue culture, applications and future aspects. Int. J. Plant Biol. 2020, 11, 8385. [Google Scholar] [CrossRef]
  2. Abdulhafiz, F.; Mohammed, A.; Hanif, R.M.F.; Kari, Z.A.; Wei, L.S.; Goh, K.W. Plant cell culture technologies: A promising alternatives to produce high-value secondary metabolites. Arab. J. Chem. 2022, 15, 104161. [Google Scholar] [CrossRef]
  3. Selwal, N.; Rahayu, F.; Herwati, A.; Latifah, E.; Supriyono; Suhara, C.; Kade, S.I.B.; Makarti, M.W.; Khurshid, W.A. Enhancing secondary metabolite production in plants: Exploring traditional and modern strategies. J. Agr. Food Res. 2023, 14, 100702. [Google Scholar] [CrossRef]
  4. Marchev, A.S.; Yordanova, Z.; Georgiev, M.I. Green (cell) factories for advanced production of plant secondary metabolites. Crit. Rev. Biotechnol. 2020, 40, 443–458. [Google Scholar] [CrossRef]
  5. Krasteva, G.; Georgiev, V.; Pavlov, A. Recent applications of plant cell culture technology in cosmetics and foods. Eng. Life Sci. 2021, 21, 68–76. [Google Scholar] [CrossRef]
  6. Rohini, M.R.; Rajasekharan, P.E. Scale-up production of bioactive compounds using bioreactors. In Nutraceuticals Production from Plant Cell Factory; Belwal, T., Ed.; Springer: Singapore, 2022; pp. 69–81. [Google Scholar]
  7. Verdú-Navarro, F.; Moreno-Cid, J.A.; Weiss, J.; Egea-Cortines, M. The advent of plant cells in bioreactors. Front. Plant Sci. 2023, 14, 1310405. [Google Scholar] [CrossRef]
  8. Chandran, H.; Meena, M.; Barupal, T.; Sharma, K. Plant tissue culture as a perpetual source for production of industrially important bioactive compounds. Biotechnol. Rep. 2020, 26, e00450. [Google Scholar] [CrossRef] [PubMed]
  9. Kozłowska, M.; Ścibisz, I.; Przybył, J.L.; Laudy, A.E.; Majewska, E.; Tarnowska, K.; Małajowicz, J.; Ziarno, M. Antioxidant and antibacterial activity of extracts from selected plant material. Appl. Sci. 2022, 12, 9871. [Google Scholar] [CrossRef]
  10. Muscolo, A.; Mariateresa, O.; Giulio, T.; Mariateresa, R. Oxidative stress: The role of antioxidant phytochemicals in the prevention and treatment of diseases. Int. J. Mol. Sci. 2024, 25, 3264. [Google Scholar] [CrossRef]
  11. Marino, A.; Battaglini, M.; Moles, N.; Ciofani, G. Natural antioxidant compounds as potential pharmaceutical tools against neurodegenerative diseases. ACS omega 2022, 7, 25974–25990. [Google Scholar] [CrossRef]
  12. Gulcin, İ. Antioxidants and antioxidant methods: An updated overview. Arch. Toxicol. 2020, 94, 651–715. [Google Scholar] [CrossRef]
  13. Platzer, M.; Kiese, S.; Herfellner, T.; Schweiggert-Weisz, U.; Miesbauer, O.; Eisner, P. Common trends and differences in antioxidant activity analysis of phenolic substances using single electron transfer based assays. Molecules 2021, 26, 1244. [Google Scholar] [CrossRef]
  14. Bansal, Y.; Mujib, A.; Mamgain, J.; Syeed, R.; Mohsin, M.; Nafees, A.; Dewir, Y.H.; Mendler-Drienyovszki, N. Integrated GC-MS and UPLC-ESI-QTOF-MS based untargeted metabolomics analysis of in vitro raised tissues of Digitalis purpurea L. Front. Plant Sci. 2024, 15, 1433634. [Google Scholar] [CrossRef]
  15. Patel, M.K.; Pandey, S.; Kumar, M.; Haque, M.I.; Pal, S.; Yadav, N.S. Plants metabolome study: Emerging tools and techniques. Plants 2021, 10, 2409. [Google Scholar] [CrossRef]
  16. Waris, M.; Kocak, E.; Gonulalan, E.M.; Demirezer, L.O.; Kır, S.; Nemutlu, E. Metabolomics analysis insight into medicinal plant science. TrAC Trends Anal. Chem. 2022, 157, 116795. [Google Scholar] [CrossRef]
  17. Abdelhafez, O.H.; Othman, E.M.; Fahim, J.R.; Desoukey, S.Y.; Pimentel-Elardo, S.M.; Nodwell, J.R.; Schirmeister, T.; Tawfike, A.; Abdelmohsen, U.R. Metabolomics analysis and biological investigation of three Malvaceae plants. Phytochem. Anal. 2020, 31, 204–214. [Google Scholar] [CrossRef]
  18. Shen, S.; Huang, J.; Li, T.; Wei, Y.; Xu, S.; Wang, Y.; Ning, J. Untargeted and targeted metabolomics reveals potential marker compounds of an tea during storage. LWT 2022, 154, 112791. [Google Scholar] [CrossRef]
  19. Maggi, F. Feverfew (Tanacetum parthenium L.) Sch. Bip.). In Nonvitamin and Nonmineral Nutritional Supplements; Mohammad, N.S., Sanchez, S.A., Eds.; Elsevier: Amsterdam, Netherlands, 2019; pp. 223–225. [Google Scholar]
  20. Pareek, A.; Suthar, M.; Rathore, G.S.; Bansal, V. Feverfew (Tanacetum parthenium L.): A systematic review. Pharmacogn. Rev. 2011, 5, 103–110. [Google Scholar] [CrossRef]
  21. Kashkooe, A.; Jalali, A.; Zarshenas, M.M.; Hamedi, A. Exploring the phytochemistry, signaling pathways, and mechanisms of action of Tanacetum parthenium (L.) Sch.Bip.: A comprehensive literature review. Biomedicines 2024, 12, 2297. [Google Scholar] [CrossRef]
  22. Binh, H.H.; Van Nghia, V. Chemical composition, antimicrobial, and anticancer activity of essential oil from Tanacetum parthenium (L.) Sch.Bip leaves cultivated in Dalat, Vietnam. Chem. Biodivers. 2025, e202403430. [Google Scholar] [CrossRef]
  23. Nithin, B.; Revathi, S.; Penabaka, V.; Chandra, P. Review on Tanacetum parthenium. Int. J. Indig. Herbs Drugs 2025, 10, 7–13. [Google Scholar] [CrossRef]
  24. Rabito, M.F.; Almeida, M.B.; Iglesias, A.H.; Paula, F.; Silva, B.P.; Cortez, D.A.; Nixdorf, S.L.; Ferreira, I.C.P. Development and validation of a method for simultaneous determination of bioactive compounds of Tanacetum parthenium (L.) Schultz-Bip. J. Braz. Chem. Soc. 2014, 10, 1824–1831. [Google Scholar] [CrossRef]
  25. Long, C.; Sauleau, P.; David, B.; Lavaud, C.; Cassabios, V.; Ausseil, F.; Massiot, G. Bioactive flavonoids of Tanacetum parthenium revisited. Phytochemistry 2003, 64, 567–569. [Google Scholar] [CrossRef]
  26. Freund, R.R.A.; Gobrecht, P.; Fischer, D.; Arndt, H.D. Advances in chemistry and bioactivity of parthenolide. Nat. Prod. Rep. 2020, 37, 441–565. [Google Scholar] [CrossRef]
  27. Izadi, Z.; Esna-Ashri, M.; Piri, K.; Davoodi, P. Chemical composition and antimicrobial activity of feverfew (Tanacetum parthenium) essential oil. Int. J. Agric. Biol. 2010, 12, 759–763. [Google Scholar]
  28. Izadi, Z.; Aghaalikhani, M.; Esna-Ashari, M.; Davoodi, P. Determining chemical composition and antimicrobial activity of feverfew (Tanacetum parthenium L.) essential oil on some microbial strains. Zahedan J. Res. Med. Sci. 2013, 15, 8–13. [Google Scholar]
  29. Shafaghat, A.; Ghorban-Dadras, O.; Mohammadhosseini, M.; Akhavan, M.; Shafaghatlonbar, M.; Panahi, A. A comparative study on chemical composition and antimicrobial activity of essential oils from Tanacetum parthenium (L.) Schultz. Bip. and Tanacetum punctatum (Desr.) Grierson. leaves from Iran. J. Essent. Oil-Bear. Plants 2017, 20, 1143–1150. [Google Scholar] [CrossRef]
  30. Ghavam, M. Tripleurospermum disciforme (C.A.Mey.) Sch.Bip., Tanacetum parthenium (L.) Sch.Bip, and Achillea biebersteinii Afan.: Efficiency, chemical profile, and biological properties of essential oil. Chem. Biol. Technol. Agric. 2021, 8, 45. [Google Scholar] [CrossRef]
  31. Banthorpe, D.V.; Brown, G.D. Growth and secondary metabolism in cell cultures of Tanacetum, Mentha and Anethum species in buffered media. Plant Sci. 1990, 67, 107–113. [Google Scholar] [CrossRef]
  32. Brown, G.D. Production of anti-malarial and anti-migraine drugs in tissue culture of Artemisia annua and Tanacetum parthenium. Acta Hortic. 1993, 330, 269–276. [Google Scholar] [CrossRef]
  33. Stojakowska, A.; Kisiel, W. Acetylenes in Agrobacterium rhizogenes transformed root cultures of Tanacetum parthenium. Pol. J. Chem. 1997, 71, 509–512. [Google Scholar]
  34. Sy, L.-K.; Brown, G.D. Coniferaldehyde derivatives from tissue culture of Artemisia annua and Tanacetum parthenium. Phytochemistry 1999, 50, 781–785. [Google Scholar] [CrossRef]
  35. Rateb, M.E.M.; El-Hawary, S.S.; El-Shamy, A.M.; Yousef, E.M.A. Production of parthenolide in organ and callus cultures of Tanacetum parthenium (L.). Afr. J. Biotechnol. 2007, 6, 1306–1316. [Google Scholar]
  36. Nieto-Trujillo, A.; Buendía-González, L.; García-Morales, C.; Román-Guerrero, A.; Cruz-Sosa, F.; Estrada-Zúñiga, M.E. Phenolic compounds and parthenolide production from in vitro cultures of Tanacetum parthenium. Rev. Mex. Ing. Quim. 2017, 16, 371–383. [Google Scholar]
  37. Pourianezhad, F.; Rahnama, H.; Mousavi, A.; Khosrowshahli, M.; Mafakheri, S. Effects of combined elicitors on parthenolide production and expression of parthenolide synthase (TpPTS) in Tanacetum parthenium hairy root culture. Plant Biotechnol. Rep. 2019, 13, 211–218. [Google Scholar] [CrossRef]
  38. Mahood, H.E.; Abbas, M.K.; Zahid, N.A. Micropropagation of feverfew (Tanacetum parthenium) and quantification of parthenolide content in its micropropagated and conventionally grown plants. Horticulturae 2022, 8, 50. [Google Scholar] [CrossRef]
  39. Saray, L.A.; Ahmadabadi, M.; Kamran, R.V. Efficient tissue culture and regeneration systems for the medicinal plant Tanacetum parthenium. Braz. Arch. Biol. Technol. 2022, 65, e22210264. [Google Scholar] [CrossRef]
  40. Fonseca, J.M.; Rushing, J.W.; Rajapakse, N.C.; Thomas, R.L.; Riley, M.B. Phenolics and parthenolide levels in feverfew (Tanacetum parthenium) are inversely affected by environmental factors. J. Appl. Hort. 2008, 10, 36–39. [Google Scholar] [CrossRef]
  41. Majdi, M.; Charnikhova, T.; Bouwmeester, H. Genetical, developmental and spatial factors influencing parthenolide and its precursor costunolide in feverfew (Tanacetum parthenium L. Schulz Bip.). Ind. Crops Prod. 2013, 47, 270–276. [Google Scholar] [CrossRef]
  42. Hojati, M.; Modarres-Sanavy, S.A.M.; Tahmasebi, E.S.; Majdi, M.; Ghanati, F.; Farzadfar, S. Differential deployment of parthenolide and phenylpropanoids in feverfew plants subjected to divalent heavy metals and trans-cinnamic acid. Plant Soil 2016, 399, 41–59. [Google Scholar] [CrossRef]
  43. El-Shamy, A.M.; El-Hawary, S.S.; Rateb, M.E. Quantitative estimation of parthenolide in Tanacetum parthenium (L.) Schultz-Bip. cultivated in Egypt. J. AOAC Int. 2007, 90, 21–27. [Google Scholar] [CrossRef]
  44. Rezaei, F.; Jamei, R.; Heidari, R. Evaluation of the phytochemical and antioxidant potential of aerial parts of Iranian Tanacetum parthenium. Pharm. Sci. 2017, 23, 136–142. [Google Scholar] [CrossRef]
  45. Wu, C.; Chen, F.; Wang, X.; Kim, H.J.; He, G.Q.; Haley-Zitlin, V.; Huang, G. Antioxidant constituents in feverfew (Tanacetum parthenium) extract and their chromatographic quantification. Food Chem. 2006, 96, 220–227. [Google Scholar] [CrossRef]
  46. Michalak, M.; Stryjecka, M.; Żarnowiec, P.; Zagórska-Dziok, M.; Kiełtyka-Dadasiewicz, A. Chemical composition of extracts from various parts of feverfew (Tanacetum parthenium L.) and their antioxidant, protective, and antimicrobial activities. Int. J. Mol. Sci. 2024, 25, 12179. [Google Scholar] [CrossRef]
  47. Hwang, S.H.; Kim, H.-Y.; Quispe, Y.N.G.; Wang, Z.; Zuo, G.; Lim, S.S. Aldose reductase, protein glycation inhibitory and antioxidant of Peruvian medicinal plants: The case of Tanacetum parthenium L. and its constituents. Molecules 2019, 24, 2010. [Google Scholar] [CrossRef] [PubMed]
  48. Yur, S.; Tekin, M.; Göger, F.; Başer, K.H.C.; Özek, T.; Özek, G. Composition and potential of Tanacetum haussknechtii Bornm. Grierson as antioxidant and inhibitor of acetylcholinesterase, tyrosinase, and α-amylase enzymes. Int. J. Food Prop. 2017, 29, S2359–S2378. [Google Scholar] [CrossRef]
  49. Zengin, G.; Cvetanović, A.; Gašić, U.; Stupar, A.; Bulut, G.; Şenkardes, I.; Dogan, A.; Sinan, K.I.; Uysal, S.; Aumeeruddy-Elalfi, Z.; et al. Modern and traditional extraction techniques affect chemical composition and bioactivity of Tanacetum parthenium (L.) Sch. Bip. Ind. Crops Prod. 2020, 146, 112202. [Google Scholar] [CrossRef]
  50. Ak, G.; Gevrenova, R.; Sinan, K.I.; Zengin, G.; Zheleva, D.; Mahomoodally, M.F.; Senkardes, I.; Brunetti, L.; Leone, S.; Di Simone, S.C.; et al. Tanacetum vulgare L. (Tansy) as an effective bioresource with promising pharmacological effects from natural arsenal. Food Chem. Toxicol. 2021, 153, 112268. [Google Scholar] [CrossRef]
  51. Polatoğlu, K.; Demirci, F.; Demirci, B.; Gören, N.; Can, B.K.H. Antibacterial activity and the variation of Tanacetum parthenium (L.) Schultz Bip. essential oils from Turkey. J. Oleo Sci. 2010, 59, 177–184. [Google Scholar] [CrossRef]
  52. Ivănescu, B.; Tuchilus, C.; Corciova, A.; Lungu, C.; Mihai, C.T.; Gheldiu, A.M.; Vlase, L. Antioxidant, antimicrobial and cytotoxic activity of Tanacetum vulgare, Tanacetum corymbosum and Tanacetum macrophyllum extracts. Farmacia 2018, 66, 282–288. [Google Scholar]
  53. Yapici, I.; Altay, A.; Sarikaya, B.Ö.; Korkmaz, M.; Atila, A.; Gülçin, I.; Köksal, E. In vitro antioxidant and cytotoxic activities of extracts of endemic Tanacetum erzincanense together with phenolic content by LC-ESI-QTOF-MS. Chem. Biodivers. 2021, 18, e2000812. [Google Scholar] [CrossRef] [PubMed]
  54. Devrnja, N.; Anđelković, B.; Aranđelović, S.; Radulović, S.; Soković, M.; Krstić-Milošević, D.; Ristić, M.; Ćalić, D. Comparative studies on the antimicrobial and cytotoxic activities of Tanacetum vulgare L. essential oil and methanol extracts. S. Afr. J. Bot. 2017, 111, 212–221. [Google Scholar] [CrossRef]
  55. Li, Y.; Zhang, Y.; Fu, M.; Yao, Q.; Zhuo, H.; Lu, Q.; Niu, X.; Zhang, P.; Pei, Y.; Zhang, K. Parthenolide induces apoptosis and lytic cytotoxicity in Epstein-Barr virus-positive Burkitt lymphoma. Mol. Med. Rep. 2012, 6, 477–482. [Google Scholar] [CrossRef]
  56. Kouhpaikar, H.; Sadeghian, M.H.; Rafatpanah, H.; Kazemi, M.; Iranshahi, M.; Delbari, Z.; Khodadadi, F.; Ayatollahi, H.; Rassouli, F.B. Synergy between parthenolide and arsenic trioxide in adult T-cell leukemia/lymphoma cell in vitro. Iran J. Basic Med. Sci. 2020, 23, 616–622. [Google Scholar]
  57. Chen, T.; Xiao, Z.; Liu, X.; Wang, T.; Wang, Y.; Ye, F.; Su, J.; Yao, X.; Xiong, L.; Yang, D.-H. Natural products for combating multidrug resistance in cancer. Pharmacol. Res. 2024, 202, 107099. [Google Scholar] [CrossRef]
  58. Lerata, M.S.; D’Souza, S.; Sibuyi, N.R.; Dube, A.; Meyer, M.; Samaai, T.; Beukes, D.R. Encapsulation of variabilin in stearic acid solid lipid nanoparticles enhances its anticancer activity in vitro. Molecules 2020, 25, 830. [Google Scholar] [CrossRef] [PubMed]
  59. Jansi, R.S.; Khusro, A.; Agastian, P.; Almutairi, M.H.; Almutairi, B.O. Bioassay-guided fractionation and biological activities of antimycin A and 4-hydroxybenzoic acid isolated from Nocardiopsis sp. strains LC-9. BioResources 2024, 19, 7673–7697. [Google Scholar] [CrossRef]
  60. Guan, L.; Long, H.; Ren, F.; Li, Y.; Zhang, H. A structure-activity relationship study of the inhibition of α-amylase by benzoic acid and its derivatives. Nutrients 2022, 14, 1931. [Google Scholar] [CrossRef]
  61. Samra, R.M.; Soliman, A.F.; Ashour, A.; Al-Karmalawy, A.A.; Hassan, M.A.; Zaghloul, A.M. Bioassay-guided isolation of a new cytotoxic ceramide from Cyperus rotundus L. S. Afr. J. Bot. 2021, 139, 210–216. [Google Scholar] [CrossRef]
  62. Kofi, A.; Stephen, G.; Francis, A. Antibacterial and radical scavenging activity of fatty acids from Paullinia pinnata L. Pharmacogn. Mag. 2009, 5, S1–S4. [Google Scholar]
  63. Chai, B.; Jiang, W.; Hu, M.; Wu, Y.; Si, H. In vitro synergistic interactions of protocatechuic acid and chlorogenic acid in combination with antibiotics against animal pathogens. Synergy 2019, 9, 100055. [Google Scholar] [CrossRef]
  64. Oboh, G.; Agunloye, O.M.; Adefegha, S.A.; Akinyemi, A.J.; Ademiluyi, A.O. Caffeic and chlorogenic acids inhibit key enzymes linked to type 2 diabetes (in vitro): A comparative study. J. Basic Clin. Physiol. Pharmacol. 2015, 26, 165–170. [Google Scholar] [CrossRef]
  65. Salzillo, A.; Ragone, A.; Spina, A.; Naviglio, S.; Sapio, L. Chlorogenic acid enhances doxorubicin-mediated cytotoxic effect in osteosarcoma cells. Int. J. Mol. Sci. 2021, 22, 8586. [Google Scholar] [CrossRef] [PubMed]
  66. Zhang, Y.; Pu, C.; Tang, W.; Wang, S.; Sun, Q. Gallic acid liposomes decorated with lactoferrin: Characterization, in vitro digestion and antibacterial activity. Food Chem. 2019, 293, 315–322. [Google Scholar] [CrossRef]
  67. Adeniyi, B.A.; Adetoye, A.; Ayeni, F.A. Antibacterial activities of lactic acid bacteria isolated from cow faeces against potential enteric pathogens. Afr. Health Sci. 2015, 15, 888–895. [Google Scholar] [CrossRef]
  68. Subramanian, A.P.; John, A.A.; Vellayappan, M.V.; Balaji, A.; Jaganathan, S.K.; Supriyanto, E.; Yusof, M. Gallic acid: Prospects and molecular mechanisms of its anticancer activity. RSC Adv. 2015, 5, 35608–35621. [Google Scholar] [CrossRef]
  69. Cunha, L.B.; Lepore, E.D.; Medeiros, C.C.B.; Sorrechia, R.; Pietro, R.C.L.R.; Corrêa, M.A. Can gentisic acid serve as a high-performance antioxidant with lower toxicity for a promising new topical application? Life 2024, 14, 1022. [Google Scholar] [CrossRef]
  70. Mechchate, H.; Es-safi, I.; Al Kamaly, O.M.; Bousta, D. Insight into gentisic acid antidiabetic potential using in vitro and in silico approaches. Molecules 2021, 26, 1932. [Google Scholar] [CrossRef]
  71. Villaño, D.; Fernández-Pachón, M.S.; Moyá, M.L.; Troncoso, A.M.; García-Parrilla, M.C. Radical scavenging ability of polyphenolic compounds towards DPPH free radical. Talanta 2007, 71, 230–235. [Google Scholar] [CrossRef] [PubMed]
  72. Xu, J.G.; Hu, Q.P.; Liu, Y. Antioxidant and DNA-protective activities of chlorogenic acid isomers. J. Agric. Food Chem. 2012, 60, 11625–11630. [Google Scholar] [CrossRef] [PubMed]
  73. Taiwo, F.O.; Oyedeji, O.; Osundahunsi, M.T. Antimicrobial and antioxidant properties of kaemferol-3-O-glucoside and 1-(4-hydroxyphenyl)-3-phenylprpan-1-one isolated from the leaves of Annona muricate (Linn.). J. Pharm. Res. Int. 2019, 26, 1–13. [Google Scholar] [CrossRef]
  74. Ajiboye, T.O.; Habibu, R.S.; Saidu, K.; Haliru, F.Z.; Ajiboye, H.O.; Aliyu, N.O.; Ibitoye, O.B.; Uwazie, J.N.; Muritala, H.F.; Bello, S.A.; et al. Involvement of oxidative stress in protocatechuic acid-mediated bacterial lethality. Microbiol. Open 2017, 6, e472. [Google Scholar] [CrossRef] [PubMed]
  75. Yin, M.C.; Lin, C.C.; Wu, H.C.; Tsao, S.M.; Hsu, C.K. Apoptotic effects of protocatechuic acid in human breast, lung, liver, cervix, and prostate cancer cells: Potential mechanism of action. J. Agric. Food Chem. 2009, 57, 6468–6473. [Google Scholar] [CrossRef]
  76. Kim, H.; Jeong, K.; Jung, S. Molecular dynamics simulations on the coplanarity of quercetin backbone for the antioxidant activity of quercetin-3-monoglycoside. Bull. Korean Chem. Soc. 2006, 27, 325–328. [Google Scholar] [CrossRef]
  77. Nile, A.; Nile, S.H.; Shin, J.; Park, G.; Oh, J.-W. Quercetin-3-glucoside extracted from apple pomace induces cell cycle arrest and apoptosis by increasing intracellular ROS levels. Int. J. Mol. Sci. 2021, 22, 10749. [Google Scholar] [CrossRef] [PubMed]
  78. Tangwatcharin, P.; Teemeesuk, W. Control of pathogenic bacteria in cooked duck blood curd using sodium diacetate and sodium chloride. Curr. Appl. Sci. Technol. 2019, 19, 306–312. [Google Scholar]
  79. Muyumba, N.W.; Mutombo, S.C.; Sheridan, H.; Nachtergael, A.; Duez, P. Quality control of herbal drugs and preparations: The methods of analysis, their relevance and applications. Talanta Open 2021, 4, 100070. [Google Scholar] [CrossRef]
  80. Fasina, O.O.; Hallman, H.; Craig-Schmidt, M.; Clements, C. Predicting temperature-dependence viscosity of vegetable oils from fatty acid composition. J. Am. Oil Chem. Soc. 2006, 83, 899–903. [Google Scholar] [CrossRef]
  81. Zhou, L.G.; Wu, J.Y. Development and application of medicinal plant tissue cultures for production of drugs and herbal medicinals in China. Nat. Prod. Rep. 2006, 23, 789–810. [Google Scholar] [CrossRef]
  82. Farah, A.; de Paula Lima, J. Consumption of chlorogenic acids through coffee and health implications. Beverages 2019, 5, 11. [Google Scholar] [CrossRef]
  83. Nieto-Trujillo, A.; Cruz-Sosa, F.; Luria-Pérez, R.; Gutiérrez-Rebolledo, G.A.; Román-Guerrero, A.; Burrola-Aguilar, C.; Zepeda-Gómez, C.; Estrada-Zúñiga, M.E. Arnica montana cell culture establishment, and assessment of its cytotoxic, antibacterial, α-amylase inhibitor, and antioxidant in vitro bioactivities. Plants 2021, 10, 2300. [Google Scholar] [CrossRef]
  84. Salapovic, H.; Geier, J.; Reznicek, G. Quantification of sesquiterpene lactones in Asteraceae plant extracts: Evaluation of their allergenic potential. Sci. Pharm. 2013, 81, 807–818. [Google Scholar] [CrossRef] [PubMed]
  85. Morales-Merida, B.E.; Grimaldi-Olivas, J.C.; Cruz-Mendívil, A.; Villicaña, C.; Valdez-Torres, J.B.; Heredia, J.B.; León-Chan, R.G.; Lightbourn-Rojas, L.A.; Monribot-Villanueva, J.L.; Guerrero-Analco, J.A.; et al. Integrating proteomics and metabolomics approaches to elucidate the mechanism of responses to combined stress in the bell pepper (Capsicum annuum). Plants 2024, 13, 1861. [Google Scholar] [CrossRef] [PubMed]
  86. Monribot-Villanueva, J.L.; Altúzar-Molina, A.; Aluja, M.J.A.; Zamora-Briseño, J.; Elizalde-Contreras, J.M.; Bautista-Valle, M.V.; Arellano, J.; Sánchez-Martínez, D.E.; Rivera-Reséndiz, F.; Vázquez-Rosas-Landa, M.; et al. Integrating proteomics and metabolomics approaches to elucidate the ripening process in white Psidium guajava. Food Chem. 2022, 367, 130656. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Pharmacological activities of the Ijms 26 07209 i001 4TP, Ijms 26 07209 i002 5TP, and Ijms 26 07209 i003 8TP fractions obtained from the methanolic extract of the root biomass of a T. parthenium culture. (a) Antioxidant, inhibitory effect of (b) α-amylase, on (c) E. coli, (d) S. aureus, cytotoxic effect against the Ramos RA-1 lymphoma cell line (e) alone and (f) combined with 1 nM vincristine. The data are presented as the means ± SD. In (a,b), the letters indicate significant differences at the 5% level of probability; from (c) to (e), * indicates significant differences at the 5% level of significance compared to the negative control, and ** indicates the treatments showing the lowest values at the 5% level of significance compared to the negative control. The corresponding controls tested for those assays were as follows: (a) MeOH could not scavenge DPPH radical; (b) the positive control (acarbose) had an IC50 of 2.48 ± 0.31 μg/mL; (c,d) the negative control for both antibacterial assays was 3% DMSO, while the positive control for E. coli was 1 μg chloramphenicol/disk and the positive control for S. aureus was 1 μg vancomycin/disk; and (e) the negative control was the culture medium (which induced a cellular viability of 94.71% ± 1.5%), while 1 nM vincristine was used as a positive control (reducing cellular viability at 72.61% ± 1.45%).
Figure 1. Pharmacological activities of the Ijms 26 07209 i001 4TP, Ijms 26 07209 i002 5TP, and Ijms 26 07209 i003 8TP fractions obtained from the methanolic extract of the root biomass of a T. parthenium culture. (a) Antioxidant, inhibitory effect of (b) α-amylase, on (c) E. coli, (d) S. aureus, cytotoxic effect against the Ramos RA-1 lymphoma cell line (e) alone and (f) combined with 1 nM vincristine. The data are presented as the means ± SD. In (a,b), the letters indicate significant differences at the 5% level of probability; from (c) to (e), * indicates significant differences at the 5% level of significance compared to the negative control, and ** indicates the treatments showing the lowest values at the 5% level of significance compared to the negative control. The corresponding controls tested for those assays were as follows: (a) MeOH could not scavenge DPPH radical; (b) the positive control (acarbose) had an IC50 of 2.48 ± 0.31 μg/mL; (c,d) the negative control for both antibacterial assays was 3% DMSO, while the positive control for E. coli was 1 μg chloramphenicol/disk and the positive control for S. aureus was 1 μg vancomycin/disk; and (e) the negative control was the culture medium (which induced a cellular viability of 94.71% ± 1.5%), while 1 nM vincristine was used as a positive control (reducing cellular viability at 72.61% ± 1.45%).
Ijms 26 07209 g001
Figure 2. Differential compounds identified between the Ijms 26 07209 i001 4TP, Ijms 26 07209 i002 5TP, and Ijms 26 07209 i003 8TP fractions obtained from the methanolic extract of the root biomass of the T. parthenium culture. (a) Total secondary metabolite contents: phenolic, phenolic acid, flavonoid, and sesquiterpene lactone (TPC, TPAC, TFC, and TSLC, respectively); LC-MS chromatogram obtained via electrospray ionization in (b) negative ionization mode and (c) positive ionization mode; (d) Venn diagram; (e) PCA; (f) PSL-DA; (g) VIP score of the top 20 compounds; and (h) heatmap of all identified compounds. In (a), data show the means ± SD; letters indicate significant differences at the 5% probability level, and the results for TPC, TPAC, TFC, and TSLC are expressed as milligrams of gallic acid, verbascoside, quercetin, or parthenolide equivalents per gram of fraction, respectively (mg GAE/gF, mg VBE/gF, mg QE/gF, or mg PTNE/gF). The Venn diagram in (d) was constructed online at http://www.interactivenn.net/, accessed on 20 August 2024. Data from (e) to (h) were analyzed for the rt_m/z signal and its abundance (peak intensity) obtained from LC-MS results.
Figure 2. Differential compounds identified between the Ijms 26 07209 i001 4TP, Ijms 26 07209 i002 5TP, and Ijms 26 07209 i003 8TP fractions obtained from the methanolic extract of the root biomass of the T. parthenium culture. (a) Total secondary metabolite contents: phenolic, phenolic acid, flavonoid, and sesquiterpene lactone (TPC, TPAC, TFC, and TSLC, respectively); LC-MS chromatogram obtained via electrospray ionization in (b) negative ionization mode and (c) positive ionization mode; (d) Venn diagram; (e) PCA; (f) PSL-DA; (g) VIP score of the top 20 compounds; and (h) heatmap of all identified compounds. In (a), data show the means ± SD; letters indicate significant differences at the 5% probability level, and the results for TPC, TPAC, TFC, and TSLC are expressed as milligrams of gallic acid, verbascoside, quercetin, or parthenolide equivalents per gram of fraction, respectively (mg GAE/gF, mg VBE/gF, mg QE/gF, or mg PTNE/gF). The Venn diagram in (d) was constructed online at http://www.interactivenn.net/, accessed on 20 August 2024. Data from (e) to (h) were analyzed for the rt_m/z signal and its abundance (peak intensity) obtained from LC-MS results.
Ijms 26 07209 g002
Figure 3. Correlations between total secondary metabolite contents (TPC, TPAC, TFC, and TSLC), pharmacological activities (antioxidant, antibacterial, cytotoxic, and α-amylase inhibition), and compound abundances among the 4TP, 5TP, and 8TP fractions obtained from the methanolic extract of the T. parthenium root culture biomass. Heatmap showing correlation values between pharmacological activities and total secondary metabolite content (a) among all three fractions and (b) by every fraction, (c) heatmap showing correlation values between compound abundance and pharmacological activities and total secondary metabolite content in the three fractions, and (d) heatmap showing correlation values between compound abundance and pharmacological activities in every fraction (showing the top correlations for Ijms 26 07209 i001 4TP, Ijms 26 07209 i002 5TP, and Ijms 26 07209 i003 8TP). Values ranging from −1 to 1 represent the value of each correlation.
Figure 3. Correlations between total secondary metabolite contents (TPC, TPAC, TFC, and TSLC), pharmacological activities (antioxidant, antibacterial, cytotoxic, and α-amylase inhibition), and compound abundances among the 4TP, 5TP, and 8TP fractions obtained from the methanolic extract of the T. parthenium root culture biomass. Heatmap showing correlation values between pharmacological activities and total secondary metabolite content (a) among all three fractions and (b) by every fraction, (c) heatmap showing correlation values between compound abundance and pharmacological activities and total secondary metabolite content in the three fractions, and (d) heatmap showing correlation values between compound abundance and pharmacological activities in every fraction (showing the top correlations for Ijms 26 07209 i001 4TP, Ijms 26 07209 i002 5TP, and Ijms 26 07209 i003 8TP). Values ranging from −1 to 1 represent the value of each correlation.
Ijms 26 07209 g003aIjms 26 07209 g003b
Figure 4. Compounds from the Ijms 26 07209 i001 4TP, Ijms 26 07209 i002 5TP, and Ijms 26 07209 i003 8TP fractions obtained from the methanolic extract of the root biomass of the T. parthenium culture correlated with pharmacological effects, and those compounds were determined mainly by the VIP score. (a) Putatively identified compounds in the 4TP, 5TP, and 8TP fractions showing correlation values related to pharmacological effects; (b) putatively identified compounds showing the main values of the VIP score.
Figure 4. Compounds from the Ijms 26 07209 i001 4TP, Ijms 26 07209 i002 5TP, and Ijms 26 07209 i003 8TP fractions obtained from the methanolic extract of the root biomass of the T. parthenium culture correlated with pharmacological effects, and those compounds were determined mainly by the VIP score. (a) Putatively identified compounds in the 4TP, 5TP, and 8TP fractions showing correlation values related to pharmacological effects; (b) putatively identified compounds showing the main values of the VIP score.
Ijms 26 07209 g004
Figure 5. Compounds were detected and quantified through targeted metabolomics from the 4TP, 5TP, and 8TP fractions obtained from the methanolic extract of the root biomass of the T. parthenium culture. (a) The chemical structures of amino acid (L-phenylalanine), hydroxybenzoic acids (4-hydroxybenzoic acid, gallic acid, gentisic acid, and protocatechuic acid), caffeoylquinic acid (chlorogenic acid), and flavonoids (kaempferol 3-O-glucoside and quercetin 3-glucoside); (b) chromatogram and (c) heatmap of the concentrations of phenolic compounds identified through targeted metabolomics in the 4TP, 5TP, and 8TP fractions. The chemical structure images in (a) were obtained from the PubChem database (https://pubchem.ncbi.nlm.nih.gov/, accessed on 15 October 2024); in (c), the Euclidan distance and Ward algorithm were used for sample clustering.
Figure 5. Compounds were detected and quantified through targeted metabolomics from the 4TP, 5TP, and 8TP fractions obtained from the methanolic extract of the root biomass of the T. parthenium culture. (a) The chemical structures of amino acid (L-phenylalanine), hydroxybenzoic acids (4-hydroxybenzoic acid, gallic acid, gentisic acid, and protocatechuic acid), caffeoylquinic acid (chlorogenic acid), and flavonoids (kaempferol 3-O-glucoside and quercetin 3-glucoside); (b) chromatogram and (c) heatmap of the concentrations of phenolic compounds identified through targeted metabolomics in the 4TP, 5TP, and 8TP fractions. The chemical structure images in (a) were obtained from the PubChem database (https://pubchem.ncbi.nlm.nih.gov/, accessed on 15 October 2024); in (c), the Euclidan distance and Ward algorithm were used for sample clustering.
Ijms 26 07209 g005
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

Nieto-Trujillo, A.; Luria-Pérez, R.; Cruz-Sosa, F.; Zepeda-Gómez, C.; González-Pedroza, M.G.; Burrola-Aguilar, C.; Sunny, A.; Correa-Basurto, J.; Guerrero-Analco, J.A.; Monribot-Villanueva, J.L.; et al. Metabolomic and Pharmacological Approaches for Exploring the Potential of Tanacetum parthenium L. Root Culture as a Source of Bioactive Phytochemicals. Int. J. Mol. Sci. 2025, 26, 7209. https://doi.org/10.3390/ijms26157209

AMA Style

Nieto-Trujillo A, Luria-Pérez R, Cruz-Sosa F, Zepeda-Gómez C, González-Pedroza MG, Burrola-Aguilar C, Sunny A, Correa-Basurto J, Guerrero-Analco JA, Monribot-Villanueva JL, et al. Metabolomic and Pharmacological Approaches for Exploring the Potential of Tanacetum parthenium L. Root Culture as a Source of Bioactive Phytochemicals. International Journal of Molecular Sciences. 2025; 26(15):7209. https://doi.org/10.3390/ijms26157209

Chicago/Turabian Style

Nieto-Trujillo, Aurelio, Rosendo Luria-Pérez, Francisco Cruz-Sosa, Carmen Zepeda-Gómez, María G. González-Pedroza, Cristina Burrola-Aguilar, Armando Sunny, José Correa-Basurto, José A. Guerrero-Analco, Juan L. Monribot-Villanueva, and et al. 2025. "Metabolomic and Pharmacological Approaches for Exploring the Potential of Tanacetum parthenium L. Root Culture as a Source of Bioactive Phytochemicals" International Journal of Molecular Sciences 26, no. 15: 7209. https://doi.org/10.3390/ijms26157209

APA Style

Nieto-Trujillo, A., Luria-Pérez, R., Cruz-Sosa, F., Zepeda-Gómez, C., González-Pedroza, M. G., Burrola-Aguilar, C., Sunny, A., Correa-Basurto, J., Guerrero-Analco, J. A., Monribot-Villanueva, J. L., & Estrada-Zúñiga, M. E. (2025). Metabolomic and Pharmacological Approaches for Exploring the Potential of Tanacetum parthenium L. Root Culture as a Source of Bioactive Phytochemicals. International Journal of Molecular Sciences, 26(15), 7209. https://doi.org/10.3390/ijms26157209

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