Next Article in Journal
Oxysterol-Induced Inflammation in Human Diseases: Strategies for Treatment with Natural Compounds and Synthetic Molecules
Previous Article in Journal
Urolithin A Attenuates Periodontitis in Mice via Dual Anti-Inflammatory and Osteoclastogenesis Inhibition: A Natural Metabolite-Based Therapeutic Strategy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Phytochemical Screening and Biological Activities of Lippia multiflora Moldenke

by
Dorcas Tlhapi
1,*,
Ntsoaki Malebo
2,
Idah Tichaidza Manduna
1,
Monizi Mawunu
3,4 and
Ramakwala Christinah Chokwe
5
1
Centre for Applied Food Sustainability and Biotechnology, Faculty of Health and Environmental Sciences, Central University of Technology, Bloemfontein 9300, South Africa
2
Centre for Innovation in Learning and Teaching, Central University of Technology, Bloemfontein 9300, South Africa
3
Department of Agronomy, Polytechnic Institute, Kimpa Vita University, Uíge P.O. Box 77, Angola
4
Department of Biology, Faculty of Science and Technology, University of Kinshasa, Kinshasa P.O. Box 190, Congo
5
Department of Chemistry, College of Science Engineering and Technology, University of South Africa, Johannesburg 1710, South Africa
*
Author to whom correspondence should be addressed.
Molecules 2025, 30(13), 2882; https://doi.org/10.3390/molecules30132882
Submission received: 27 May 2025 / Revised: 2 July 2025 / Accepted: 3 July 2025 / Published: 7 July 2025

Abstract

Lippia multiflora Moldenke is widely used in Angola, on the African continent, and beyond for the treatment of many health conditions such as hypertension, enteritis, colds, gastrointestinal disturbances, stomachaches, jaundice, coughs, fevers, nausea, bronchial inflammation, conjunctivitis, malaria, and venereal diseases. However, there is limited literature about the active compounds linked with the reported biological activities. This study aims to assess the chemical profiles, antioxidant properties, and the cytotoxicity effects of the roots, stem bark, and leaves of L. multiflora. Chemical characterization of the crude extracts was assessed through quantification of total phenolic and flavonoid contents followed by Q exactive plus orbitrap™ ultra-high-performance liquid chromatography-mass spectrometer (UHPLC-MS) screening. The correlation between the extracts and the correlation between the compounds were studied using the multivariate analysis. Principal component analysis (PCA) loading scores and principal component analysis (PCA) biplots and correlation plots were used to connect specific compounds with observed biological activities. The antioxidant activities of the crude extracts were carried out in vitro using DPPH (2,2-diphenyl-1-picrylhydrazyl) free radical scavenging and reducing power assays, while the in vitro toxicology of the crude extracts was evaluated using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. A total of twenty constituents were characterized and identified using the UHPLC–Q/Orbitrap/MS. The methanol leaf extract showed the highest antioxidant activity in the DPPH free radical scavenging activity (IC50 = 0.559 ± 0.269 μg/mL); however, the stem bark extract had the highest reducing power (IC0.5 = 0.029 ± 0.026 μg/mL). High phenolic and flavonoid content was found in the dichloromethane leaf extract (32.100 ± 1.780 mg GAE/g) and stem bark extract (624.153 ± 29.442 mg QE/g), respectively. The results show the stem bark, methanol leaf, and dichloromethane leaf extracts were well-tolerated by the Vero cell line at concentrations up to 50 µg/mL. However, at 100 µg/mL onward, some toxicity was observed in the root, methanol leaf, and dichloromethane leaf extracts. The UHPLC–Q/Orbitrap/MS profiles showed the presence of terpenoids (n = 5), flavonoids (n = 5), phenols (n = 4), alkaloids (n = 3), coumarins (n = 1), fatty acids (n = 1), and organic acids (n = 1). According to several studies, these secondary metabolites have been reported and proven to be the most abundant for antioxidant potential. The identified flavonoids (catechin, quercitrin, and (−)-epigallocatechin) and phenolic compound (6-gingerol) can significantly contribute to the antioxidant properties of different plant parts of L. multiflora. The research findings obtained in this study provide a complete phytochemical profile of various parts of L. multiflora that are responsible for the antioxidant activity using UHPLC–Q/Orbitrap/MS analysis. Furthermore, the results obtained in this study contribute to the scientific information or data on the therapeutic properties of Lippia multiflora and provide a basis for further assessment of its potential as a natural remedy.

Graphical Abstract

1. Introduction

Lippia multiflora Moldenke belongs to the Verbenaceae family, which includes about ~200 known species of small trees, shrubs, and grasses. It is usually found in tropical West African regions, Central American countries, and South American countries [1,2,3]. Lippia multiflora is commonly known as “Ti-tree, bush tea”, “Gambian tea bush”, or “healer herb” (in English), “thé de Gambie’’ (in French), “bunsurun fadama” or “godon kada” (in Hausa), “sre Numum” (in Akan), “naasuruu’’ (in Ga), “afudoti or afu” (in Ewe), and “Bulukutu” (in Kikongo) [4,5,6,7]. It is an aromatic, perennial plant with more or less woody stems growing up to 2.7 to 4.0 m in height [4]. Aerial and underground parts of L. multiflora are commonly used by traditional healers in Angola and other African countries for the treatment of health conditions such as hypertension, enteritis, cleaning lymph, chest pain, diabetes, colds, gastrointestinal disturbances, stomach aches, jaundice, coughs, fevers, nausea, bronchial inflammation, conjunctivitis, malaria, and venereal diseases [5,7,8,9].
Ethnopharmacological studies have shown antimalarial, antiviral, antifungal, analgesic, antipyretic, anti-inflammatory, antimicrobial, anti-diarrheal, and antifungal activities from Lippia multiflora [10,11,12,13]; these activities are usually attributed to the presence of alkaloids, terpenes, saponins, glycosides, flavonoids, and tannins [4]. Despite L. multiflora being used in traditional herbal medicine, the molecular basis underlying its therapeutic effects remains largely unexplored. Previous studies on the phytochemistry of Lippia multiflora have revealed very few isolated compounds, such as monoterpenoids (carvacrol and geniposide) and phenolic compounds (leucoseptoside A, alyssonoside, isoverba phenolscoside, verbascoside, samioside, isonuomiside A, and nuomioside A), using classical methods [14,15]. According to literature review studies [14,15], the pharmacological properties of these compounds isolated from L. multiflora have never been studied. Furthermore, the extracts and secondary metabolites of this plant species remain less explored with regard to their antioxidant and cytotoxic activities. Highlighting a need to identify, quantify, and characterize the metabolite profile of different organs of Lippia multiflora fully. Phytochemical screening allows for the identification of different classes of phytoconstituents within plant products, which could have potential pharmaceutical applications.
The current study shows that phytochemical analysis using UHPLC–Q/Orbitrap/MS enables a faster quantification, characterization, and identification of various secondary metabolites with remarkable sensitivity and accuracy compared to classical phytochemical techniques, which are time-consuming and sensitive, and consume a high amount of solvents. In addition, the quantity of chemical compounds isolated from plants is often small; thus, conducting structure elucidation of the isolated compounds from plants can be difficult to carry out using classical methods and requires precision. The distribution of antioxidants in different organs of the plant species has been evaluated using the DPPH (2,2-diphenyl-1-picrylhydrazyl) free radical scavenging and reducing power assays. Natural antioxidants in a daily diet have shown substantial benefits for human health and different diseases by reducing the effect of reactive oxygen or nitrogen compounds [16].
Oxidative stress is caused by the imbalance between the production of reactive oxygen species (ROS) and the body’s antioxidant defense mechanisms [17]. Free radicals that are released by cells often result in the degradation of proteins and DNA that facilitates diseases such as Alzheimer’s, cancer, neurodegenerative diseases, diabetes, age-related degeneration conditions, inflammation, rheumatoid arthritis, and atherosclerosis [18]. Antioxidants protect deoxyribonucleic acid (DNA) from oxidation to lower the risk of developing degenerative and chronic conditions [19]. Previous research studies have uncovered that herbal or traditional medicinal plants are good sources of antioxidants due to the fact that they are rich in flavonoids and phenolic compounds [20]. These compounds can protect humans from high levels of chelate metal ions, scavenge free radicals, and inhibit lipid peroxidation and free radicals [21,22]. A study conducted by Dabire et al. (2015) confirmed that the leaf crude extracts of L. multiflora have potent antioxidant activity against DPPH (2,2-diphenyl-1-picrylhydrazyl) free radical scavenging assays [23]. However, the antioxidant activity of the crude stem bark and root extract from L. multiflora has never been explored. Therefore, this study aimed at exploring the phytochemical and pharmacological properties of L. multiflora. It focuses on the crude leaf, root, and stem bark extracts, using UHPLC–Q/Orbitrap/MS, and reports on the antioxidant and cytotoxicity assessments.

2. Results and Discussion

2.1. Total Flavonoid Contents (TFCs) and Total Phenolic Contents (TPCs)

Natural antioxidants from plants are usually found in the form of phenolic compounds such as tocopherols, phenolic acids, flavonoids, etc. [24,25]. Several studies have shown that the antioxidant activity of medicinal plants could be attributed to their high content of flavonoids and phenolic compounds [26,27,28]. Phenolic compounds play a critical role in scavenging free radicals, reducing tocopherol radicals, activating antioxidant enzymes, chelating metals, and inhibiting enzymes that cause oxidation reactions, thus protecting cells from oxidative damage [21]. Flavonoids play a significant role in scavenging most oxidizing molecules, including various free radicals (peroxy radicals, hydroxyl radicals, and superoxide anion radicals) and singlet oxygen [22]. The different plant organs in L. multiflora were screened for the total contents of phenolics and flavonoids that might have contributed to their antioxidant activities. The total flavonoid content of the crude extracts was determined using the aluminum chloride (AlCl3) colorimetric method. Quercetin was used to formulate a standard curve (y = 0.0865x + 3.3762; R2 = 0.9973; Figure S1, Supplementary Material). The highest flavonoid content was found in methanol stem bark extract (624.153 ± 29.442 mg QE/g, p < 0.05), followed by methanol root extract (593.72 ± 57.918 mg QE/g, p < 0.05), dichloromethane leaf extract (305.666 ± 117.401 mg QE/g, p < 0.05), and methanol leaf extract (37.173 ± 6.443 mg QE/g, p < 0.05; see Figure 1). Our findings are in agreement with previous research studies that have shown that methanol stem bark extracts have high flavonoid content compared to the other different parts of the plant [29,30,31,32]. Moreover, other studies have proven that the antioxidant properties of several medicinal plant extracts are due to the high quality of the flavonoids or synergistic interactions of flavonoids with other various metabolites [33,34,35,36,37]. Therefore, our results confirm that the flavonoids found in the stem bark extracts of L. multiflora contributed to the potent antioxidant activities. Furthermore, the total phenolic content in the crude extracts was estimated using Folin–Ciocalteu’s reagent. Gallic acid was used to formulate a calibration curve (y = 1.0372x + 1.2487; R2 = 0.9076; see Figure S2, Supplementary Material). The results in Figure 1 show that total phenolic compounds in the extracts varied widely, ranging from 8.313 ± 3.179 to 32.100 ± 1.780 mg GAE/g with a descending order of dichloromethane leaf > root > methanol leaf > stem bark. The dichloromethane leaf extract exhibited the highest total phenolic content (32.100 ± 1.780 mg GAE/g, p < 0.05), while the stem bark extract (8.313 ± 3.179 mg GAE/g, p < 0.05) exhibited the lowest total phenolic content in comparison with the other crude extracts, as shown in Figure 1. The results obtained in this study support the result of a previous study conducted by Rouamba et al. (2024), who reported that the total phenolic content in the dichloromethane extract of L. multiflora leaves extracted by the maceration method was 96.70 ± 1.70 mg GAE/g (p < 0.05) [38]. The phenolic content of any plant is directly linked to its antioxidant properties. Phenolic compounds act as hydrogen donors and reducing agents and are capable of scavenging free radicals [39]. The presence of a noticeably good amount of phenolics in the leaf extracts of L. multiflora might contribute significantly to the antioxidant properties. These properties could be the reason why this plant species is used in various traditional herbal drugs. Furthermore, in other previous studies, the total phenolic contents of methanol and ethanol extracts of L. multiflora leaf was 462.20 ± 10.90 mg GAE/g (p < 0.05) and 71.17 ± 0.2 µg GAE/g [23,38], which is quite higher than that of the present study. Therefore, it can be suggested that the concentration of phenols that affect the antioxidant activities of the plant depends on the extraction methods used, the choice of extraction solvent, the metabolite content of each plant species, and other conditions [39,40]. Consequently, comparable with the data in the existing literature for other raw extracts of the plant products [41], our findings indicated that the contents of phenolics and flavonoids, as well as the reducing power and radical scavenging activities, might be due to the phenolic acids and flavonoids found in various extracts of L. multiflora.

2.2. Phytochemical Screening of the Crude Extracts of the Root, Leaf, and Stem Bark Using UHPLC–Q/Orbitrap/MS Analysis

The identification of the chemical constituents was carried out by UHPLC–Q/Orbitrap/MS. Both negative (ESI (−), Figure S3, Supplementary Material) and positive (ESI (+), Figure S4, Supplementary Material) modes were evaluated; however, the negative (ESI (−)) mode was chosen for additional sample investigation since it resulted in a larger abundance of ions. A total of twenty constituents from different plant parts of L. multiflora have been identified and characterized by comparing their spectral information with values in the existing literature. UHPLC–Q/Orbitrap/MS data for the identified chemical constituents, namely, the main product ions, the molecular ion [M − H], retention time, and their fragmentation ions, are provided in Table 1. Different classes of metabolites, such as terpenoids (15), flavonoids (610), phenols (1114), alkaloids (1517), coumarin (18), fatty acids (19), and organic acids (20), were identified. The UHPLC–Q/Orbitrap/MS profiles showed that various Lippia multiflora plant parts generated both similar and dissimilar compounds. Many of these compounds were mostly found in the leaf rather than in the stem and root extracts.

2.2.1. Terpenoids

Compound 1 (Figure S5, Supplementary Material) with a precursor ion at m/z 483.12 [M − H] and the molecular formula C30H44O5 was identified as poricoic acid B; this molecule produced a product ion at m/z 483, which is similar to results previously reported in the literature [42]. Compound 2 (at m/z 455.30) exhibited a molecular ion peak that corresponded to boswellic acid alpha (Figure S6, Supplementary Material), which was verified using two fragment ions [M − H] at m/z 454 and 453 [43]. However, compounds 3 (Rt = 10.06 min) and 4 (Rt = 1.55 min), with [M − H] ions at m/z 469.11 (Figure S7, Supplementary Material) and m/z 501.11 (Figure S8, Supplementary Material), respectively, were identified as eburicoic acid (C31H50O3) and medicagenic acid (C30H46O6), respectively [44,45]. Compound 5 (Figure S9, Supplementary Material), with an [M − H] ion at m/z 525.30, which produced a fragmentation at m/z 525, was identified as albiflorin by comparison with the reference [46].

2.2.2. Flavonoids

The MS-MS spectra of compounds 6 (Figure S10, Supplementary Material), 7 (Figure S11, Supplementary Material), 8 (Figure S12, Supplementary Material), 9 (Figure S13, Supplementary Material), and 10 (Figure S14, Supplementary Material) revealed fragment ions at m/z 259,231; m/z 259,243; m/z 267; m/z 311; and m/z 455,303, respectively. These data are similar to the published data on catechin (C15H14O6), epicatechin (C15H14O6), formononetin-7-O-glucoside (Ononin, C22H22O9), 3′,4′-dimethoxy-7-hydroxyflavone (C17H14O5), and quercitrin (C21H20O11), respectively [47,48,49,50].

2.2.3. Phenolic Compounds

A total of four phenolic metabolites were identified based on the MS/MS fragment ions (m/z) observed, respective to the precursor or parent ions observed. The identification of these phenolic metabolites in the Lippia multiflora extract was executed by comparing the spectral data with data in the literature. Compounds 11 (m/z 293.18 [M − H]), 12 (m/z 277.20 [M − H]), 13 (m/z 295.23 [M − H]), and 14 (m/z 305.92 [M − H]) were identified as 6-gingerol (C17H26O4, Figure S15, Supplementary Material), 6-paradol (C17H26O3, Figure S16, Supplementary Material), esmolol (C16H25NO4, Figure S17, Supplementary Material), and (−)-epigallocatechin (C15H14O7, Figure S18, Supplementary Material), respectively [48,51]. The MS-MS spectra of compounds 11, 12, and 14 showed fragment ions at m/z 248, 237, and 209; m/z 283, 233, and 205; and m/z 289 and 221, respectively, whereas peak 13 did not show recognizable MS fragment ions.

2.2.4. Alkaloids

A total of three alkaloid compounds were identified in various plant parts of Lippia multiflora. Compound 15 (Figure S19, Supplementary Material) was tentatively assigned as isomajdine (C23H28N2O6); fragmentation from [M − H] ions at m/z 397, 381, 369, 243, and 231 [52]. Based on the comparison of the MS data and retention times with those reported in the literature [53], compounds 16 (m/z 339.12 [M − H], Figure S20, Supplementary Material) and 17 (m/z 339.20 [M − H], Figure S21, Supplementary Material) were identified as yohimbinic acid (C20H24N2O3) and reserpic acid (C22H28N2O5).

2.2.5. Coumarins and Others

Compound 18 (Figure S22, Supplementary Material) with a precursor ion at m/z 339.07 [M − H] and the molecular formula C15H16O9 was identified as 6,7-dihydroxycoumarin-6-glucoside (Esculin), a coumarin glucoside. This molecule produced a product ion at m/z 338 from the precursor ion of m/z 339.07 [54]. In addition to this, one fatty acid compound was identified with m/z 307.19 [M − H] as a precursor ion and confirmed as dihomolinoleic acid (compound 19, Figure S23, Supplementary Material). Compound 19 generated fragment ions at m/z 289 and 290 [55]; the spectral data are similar to the values in the literature. Compound 20 (Rt = 0.35 min), with [M − H] ions at m/z 229.14 (Figure S24, Supplementary Material), was identified as dodecanedioic acid (C12H22O4), an organic acid. Compound 20 generated fragment ions at m/z 213; the data were confirmed by comparison with previous research [56].

2.3. Comparison of Analytical Equipment Used for Compound Identification in This Study with the Existing Literature

Table 2 summarizes differences in the analytical equipment used for compound identification in this study (exp) with the existing literature (lit). Desmiaty et al. (2020) used an UPLC-MS XEVO G2-XS QTOF to identify the presence of poricoic acid B (1) from the crude stem bark methanol extract of Rubus fraxinifolius Poir. [42]. Zhang et al. (2021) structurally characterized poricoic acid B (1) isolated from the crude ethyl acetate poriae cutis extract of Poria cocos (Schw.) Wolf using a UHPLC-QTOF-MS/MS and nuclear magnetic resonance (1H-NMR and 13C-NMR) spectroscopy [57]. Mannino et al. (2016) identified boswellic acid alpha (2) from the methanol gum resins of Boswellia sacra Flueck and Boswellia serrata Roxb by HPLC-DAD-ESI-MS/MS [43]. Compound (3) was tentatively characterized by UPLC-QTOF-MS from the ethanol cactus, cladodes, and callus of Opuntia ficus-indica (L.) Mill. [44]. In another study by Sheth et al. (1967), eburicoic acid (3) isolated from the petroleum ether stem decay extract of Fomitopsis pinicola (Sw.) P.Karst. was chemically characterized by means of FTIR and NMR spectroscopy [58]. Compound (4) was characterized as medicagenic acid from the root of Medicago truncatula Gaertn. using liquid chromatography coupled to negative-ion electrospray ionization Fourier transform ion cyclotron resonance mass spectrometry (LC ESI FT-ICR MS) [45]. However, Timbekova et al. (1996) identified and characterized medicagenic acid (4) from the leaves and root extracts of Medicago sativa L. by gas–liquid chromatography (GLC) and nuclear magnetic resonance (1H-NMR and 13C-NMR) spectroscopy [59]. Wang et al. (2016) detected the presence of albiflorin (5) in the crude ethanol extracts of aerial and underground plant parts of Cinnamomi ramulus, Paeoniae radix Alba, Jujubae fructus, Glycyrrhizae radix et Rhizoma preparata cum melle, and Zingiberis rhizoma Recens by LC-Q-TOF-MS and LC-IT-MS [46]. On the other hand, Wu et al. (2021) used HPLC–DAD and HPLC–DAD–ESI–MS to identify albiflorin (5) in the crude ethanol extracts (root, stem, leaves, and flowers) of Paeonia lactiflora Pall. [60]. Liu et al. (2021) used an UHPLC-Q-Orbitrap HRMS to identify catechin (6) in the traditional Chinese medicine decoction [47]. The herbal mixture contained Pericarpium citri Reticulatae, Radix asteris, Radix scutellariae, Semen armeniaceae Amarum, Fructus aurantii Immaturus, Rhizoma belamcandae, Radix glycyrrhizae Praeparata, Agastachis herba, Rhizoma pinelliae, Gypsum fibrosum, Rhizoma dioscoreae, Poria cocos, Asari herba, Polyporus flos Farfarae, Alismatis rhizoma, Cinnamoni ramulus, and Ephedrae herba macerated in deionized water before being decocted [47]. Furthermore, Abdel-Aal et al. (2022) separated and quantified catechin (6) using an UPLC in the aqueous tea leaves of Camellia sinensis (L.) Kuntze; the identity of catechin (6) was confirmed using liquid chromatography mass spectrometry (LC-MS/MS) [61]. Araujo et al. (2020) screened and identified epicatechin (7) from the aqueous leaves, pulp, and seed extracts of Eugenia calycina Cambess by LC-ESI-MS/MS [48]. Compound (8) was separated using an HPLC/DAD from the methanolic leaves and flower extracts of Sulla (Hedysarum coronarium L.); the chemical structure of formononetin-7-O-glucoside (Ononin, 8) was identified by LC-ESI-MS [49]. Conversely, Zgórka et al. (2022) identified and quantified formononetin-7-O-glucoside (Ononin, 8) from the aqueous–ethanolic leaves and flower extracts of Zigzag clover (Trifolium medium L.) using advanced analytical techniques such as RP-LC/PDA/ESI-QTOF/MS-MS spectroscopy [62]. Tine et al. (2017) used LC-MS/MS to characterize quercitrin (10) from the methanolic plant organs from separated parts (root barks, trunk barks, stem, leaves, and fruits) of Zanthoxylum zanthoxyloides (Lam.) Zepern. & Timler [50]. In another study, Parolin Trindade et al. (2025) characterized quercitrin (10) from the crude ethanolic leaf extract of Lippia origanoides Kunth by UHPLC-ESI-QTOF-MS/MS [63]. Araujo et al. (2020) screened and identified 6-gingerol (11), 6-paradol (12), and esmolol (13) from the aqueous leaves, pulp, and seed extracts of Eugenia calycina Cambess by LC-ESI-MS/MS [48]. Compound (14) was tentatively identified as (−)-epigallocatechin from a Japanese knotweed rhizome bark extract by HPLC-MS, SEC-HPLC-UV, and HPTLC [64]. Gülçin et al. (2012) isolated and characterized isomajdine (15) from the crude methanol root extract of Vinca herbacea Waldst. and Kit by NMR [65]. In addition, Kumar et al. (2016) used HPLC–ESI–QTOF–MS/MS to determine the chemical structure of yohimbic acid (16) and reserpic acid (17) from the crude ethanol root extracts of six Rauwolfia species (R. vomitoria, R. verticillata, R. tetraphylla, R. serpentina, R. micrantha, and R. hookeri) [53]. In another study conducted by Wang et al. (2014), yohimbic acid (16) was identified from Pausinystalia johimbe (K. Schum.) Pierre ex Beille by UHPLC–UV–MS/MS [66]. Compound (18) was tentatively characterized by HR-LCMS from the crude methanol extracts (leaves, young stems, and roots) of Oroxylum indicum (L.) Vent [54]. The chemical structures of eicosadieneoic acid (19) and dodecanedioic acid (20) were determined by LC-Q-TOF-MS and UHPLC–Q-exactive orbitrap MS from the methanol and ethyl acetate seed extracts of Nigella sativa L. and the water rhizome extracts of Senecio cannabifolius Less., respectively [55,56].

2.4. Multivariate Analysis

The phytochemical data were analyzed using the PAST 4.03 software to assess the variance and correlation between the extracts and the correlation between the compounds. The compounds were given code names (C1C20) according to their order in Table 1. The variance and correlation between the extracts were assessed using principal component analysis and hierarchical clustering. In contrast, the correlation between the compounds that were tentatively identified was evaluated using the correlation plot. Principal component 1 (PC1) and principal component 2 (PC2) explain 52.23% and 30.89% of the variance, respectively (Figure 2A). The most influential compounds along PC1 were C9, C10, C11, C13, C16, C18, and C4, C5, and C19, which had a positive and negative influence, respectively. While C6, C7, and C15 had a positive influence, and C3 and C19 had a negative influence on PC2, as shown in Table 3. This means that variance along PC1 and PC2 is due to the presence or absence of these compounds in the extracts. The PCA biplot shows that DL and ML are positively correlated to each other but are negatively correlated to R and S. This is because C10 and C18 were tentatively identified in these extracts while they were not detected in R and S. Figure 2B shows that DL and ML are more similar to each other than they are to other extracts. This is due to the fact that C10 and C18 were identified in these extracts, while they were not identified in the other extracts. At the same time, the same is true for R and S. Figure 2C, however, shows that R and S correlate more to DL than to ML. This is because ML contains C11, C13, and C16, while these compounds were not identified in the other extracts. Figure 2D shows that C9, C11, C13, and C16 are highly correlated; this is because these compounds were all identified in R, S, and DL. C18 and C10 are correlated because they were identified in DL and ML but not in R and S. C5 and C4 were identified in S, while not in the other extracts. C7 and C6 were identified in S and DL but not in R and ML; hence, they have a high correlation. C10 and C18 are negatively correlated to C19; this is due to the fact that C10 and C18 were identified in ML and DL, while C19 was not. Furthermore, C19 was identified in R and S, while C18 and C10 were not identified in these extracts.

2.5. Antioxidant Activity

All samples exhibited strong free radical scavenging and reducing power activities (Table 4). The methanol leaf extract exhibited high radical scavenging activity with an IC50 value of 0.559 ± 0.269 µg/mL (p < 0.05), whereas the root extract exhibited the lowest radical scavenging activity (IC50 = 25.108 ± 19.798 µg/mL, p < 0.05). In contrast, the stem bark extract exhibited high reducing power activity with an IC0.5 value of 0.029 ± 0.026 µg/mL (p > 0.05), while the methanol leaf extract exhibited the lowest reducing power activity (0.710 ± 1.220 µg/mL (p > 0.05)), as shown in Table 4. The presence of the polyphenols and flavonoids identified in the methanol leaf and stem bark extracts could have contributed to the high antioxidant properties of these extracts, as shown in Table 4. The reason for the high radical scavenging activity of the methanol leaf extract as compared to the other extracts could be that it contains C9, C11, C13, and C16, whose antioxidant properties have been reported before, while the other extracts do not. This is also shown by Table 3 and Figure 2A, where it is observed that these compounds have a positive influence on PC1, meaning extracts with a high PC1 score contain these compounds. The compounds C4 and C5, which have also shown antioxidant activity before, were only identified in the stem bark extract, which could be the reason why this extract exhibited high reducing power activity compared to the others. These compounds have a negative influence on PC1; this means extracts with a low PC1 score contain these compounds. Additionally, the correlation of these compounds is also shown by Figure 2D. Wang et al. (2003) reported that 6-gingerol (11) isolated from the rhizome of Zingiber officinale Roscoe exhibited a high DPPH radical scavenging activity, with an EC50 value of 23.07 µg/mL (p < 0.05) [67]. However, Jug et al. (2021) showed that (−)-epigallocatechin (14), tentatively identified from a Japanese knotweed rhizome bark extract, exhibited high DPPH radical scavenging activity, with an IC50 value of 1.5 µg/mL−1 [64]. Grzesik et al. (2018) reported that catechin (6) found in the stem bark extract (Table 1) has potent antioxidant activities against DPPH radical scavenging and reducing power activities, with IC50 and IC0.5 values ranging from 0.965 ± 0.015 mol TE/mol to 3.965 ± 0.067 mol TE/mol (p < 0.05) and 0.793 ± 0.004 mol TE/mol to 1.032 ± 0.007 mol TE/mol (p < 0.05), respectively [68]. Furthermore, Picos-Salas et al. (2024) confirmed that the presence of quercitrin (10), quantified using an ultra-performance liquid chromatography mass spectrometry (UPLC-MS) analysis, contributed to the antioxidant activity of the methanolic stems, leaves, and flowers of Lippia graveolens Kunth using the oxygen radical absorbance capacity (ORAC) and trolox equivalent antioxidant capacity (TEAC) assays [69]. However, González Güereca et al. (2007) reported that quercitrin (10) isolated from the ethyl acetate stem of Lippia graveolens HBK var. berlandieri Schauer by silica gel column chromatography showed a high DPPH radical scavenging activity, with an IC50 value of 11.24 ± 1.10 µg/mL [70]. With that being said, it can be concluded that 6-gingerol (11), (−)-epigallocatechin (14), catechin (6), and quercitrin (10) play a significant role in the antioxidant activities of the methanol leaf and stem bark extracts. In addition, the findings obtained in this study are not in line with previous reports conducted by Dabire et al. (2015), who demonstrated that the dichloromethane leaf extracts of L. multiflora (IC50 = 175.60 ± 0.1 µg/mL) extracted by cold maceration had the lowest antioxidant activity using the 2,2-diphenyl-1-picrylhydrazine (DPPH) scavenging assay [23]. Dabire et al. (2015) further showed that the ethyl acetate leaf extracts of L. multiflora had the highest antioxidant activity, with an IC50 value of 23.68 ± 0.2 µg/mL [23]. These observations prove that the activity of a plant-extracted product depends not only on the existence of bioactive constituents but also on the possible interactions with other compounds present in the extract, the solvent used for extraction, and environmental factors (such as humidity, different soil characteristics, light intensity, temperature, rainfall, height above MSL, and change of season) [71,72,73]. The findings of this research also showed that the crude extracts had higher antioxidant activities compared to quercetin, gallic acid, and ascorbic acid using both 2,2-diphenyl-1-picrylhydrazine (DPPH) scavenging and reducing power methods. This shows that L. multiflora could be a great antioxidant agent useful in the treatment and management of free radicals.

2.6. Comparison of the Methods Used to Determine the Antioxidant Activity of Extracts Obtained in This Study with the Existing Literature

Table 5 summarizes differences in the methods used to determine the antioxidant activity of extracts obtained in this study (exp) with the existing literature (lit). Desmiaty et al. (2020) confirmed that the presence of poricoic acid B (1), identified using a liquid chromatography equipped with mass spectrometry (Waters UPLC–MS XEVO G2–XS QTOF, Milford, MA, USA) analysis, contributed to the antioxidant activity of the crude methanol stem bark extracts of Rubus fraxinifolius Poir. (IC50 = 63.04 ppm) using DPPH (2,2-diphenyl-1-picrylhydrazyl) free radical assays [42]. Gupta et al. (2022) reported that boswellic acid (2) alpha, characterized using high-performance liquid chromatography (HPLC), contributed to the antioxidant activity of the methanol oleo-gum resin extract of Boswellia serrata Roxb. with percent inhibition (70.7–78.4%) and (67.79–87.57%) at concentrations of 100 μg/mL and 500 μg/mL using DPPH (2,2-diphenyl-1-picrylhydrazyl) free radical scavenging and ABTS (2,2′-azino-bis(3-ethylbenzothiazoline- 6 sulfonic acid)) assays [74]. Wu et al. (2021) showed that albiflorin (5) found in the ethanol extracts (root, stem, leaves, and flowers) of Paeonia lactiflora Pall. could be responsible for the antioxidant effectiveness of this plant species against DPPH (IC50 = 55.05 ± 0.15 μg/mL) free radical scavenging and ABTS (IC50 = 33.10 ± 0.41 μg/mL) assays [60]. A study conducted by Grzesik et al. (2018) showed that catechin (6) and epicatechin (7) had the highest ABTS scavenging capacity and ferric reducing antioxidant power (FRAP), with IC50 values of 3.965 ± 0.067 mol TE/mol and 0.793 ± 0.004 mol TE/mol and 2.800 ± 0.051 mol TE/mol and 0.917 ± 0.004 mol TE/mol, compared to other metabolites [68]. Zgórka et al. (2022) documented that formononetin-7-O-glucoside (Ononin, 8) obtained in the aqueous–ethanolic leaves and flower extracts of Zigzag clover (Trifolium medium L.) had higher antioxidant activity against ABTS radical (IC50 = 30.30 ± 0.18 µg/mL) and DPPH radical (IC50 = 30.18 ± 0.37 µg/mL) [62]. Tine et al. (2017) showed that the presence of quercitrin (10) was responsible for the antioxidant activities of the methanolic trunk barks (IC50 = 33.32 ± 0.88 µg/mL) and leaves (IC50 = 24.96 ± 0.32 µg/mL) of Zanthoxylum zanthoxyloides (Lam.) Zepern. & Timler using the ABTS radical method [50,69,70]. Furthermore, Wang et al. (2003) revealed that 6-gingerol (11) isolated from the rhizome of Zingiber officinale Roscoe displayed a high DPPH radical scavenging activity, with an EC50 value of 23.07 µg/mL [67]. Jug et al. (2021) showed that (−)-epigallocatechin (14), tentatively identified from a Japanese knotweed rhizome bark extract, exhibited high DPPH radical scavenging activity with an IC50 value of 1.5 µg/mL−1 [64]. Gülçin et al. (2012) reported that isomajdine (15) isolated and characterized from the crude methanol root extract of Vinca herbacea Waldst. and Kit exhibited antioxidant activity against ABTS scavenging (EC50 = 34.11 μg/mL), DMPD scavenging (EC50 = 32.09 μg/mL), and ferrous ion (Fe2+) chelating activity (EC50 = 10.05 μg/mL) [65].

2.7. Pearson Correlation Analysis Between Phytochemicals (TFC and TPC) and Their Antioxidant Activities

As shown in Table 6, a Pearson correlation was conducted between phytochemicals (TFC and TPC) and their antioxidant capacities to understand their relationship. The results illustrate a significant strong negative correlation from the reducing power with TFC (r = −0.956). DPPH activity showed a strong positive correlation with the TFC value (r = 0.619). The weak negative correlation (r = −0.019) between RP and TPC indicates no relation between reducing power and the total phenolic content. Additionally, the weak negative correlation (r = −0.111) between TPC and TFC indicates no relation between the total phenolic content and total flavonoid content. There was a moderate negative correlation (r = −0.406) between RP and DPPH activity. On the other hand, DPPH activity showed a weak relationship (r = 0.071) with TPC.

2.8. Cytotoxicity

Cytotoxicity was evaluated using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay against Vero cells (derived from the kidney of an African green monkey) at varying concentrations of 50 µg/mL, 100 µg/mL, 250 µg/mL, and 500 µg/mL (Figure 3). The inhibitory concentration value (IC50, Table 7) was evaluated from the percentage (%) of cell viability compared to the positive control (doxorubicin, Table 7 and Figure 3). The results show the stem bark, methanol leaf, and dichloromethane leaf extracts were well-tolerated by the Vero cell line at concentrations up to 50 µg/mL. On the other hand, at 100 µg/mL onward, some toxicity was observed in the root, methanol leaf, and dichloromethane leaf extracts, except in the stem bark (Figure 3). This might be due to the fact that the stem bark contains medicagenic acid (4) and albiflorin (5) which the other extracts do not. The only difference between the stem bark extract and the other extracts is also shown in the PCA loading scores (Table 3), where C4 and C5 have a negative influence on PC1, meaning samples with high PC scores do not contain these compounds. Furthermore, certain compounds present in the root, stem bark, and leaf extracts of Lippia multiflora (Table 1), such as eburicoic acid (3) [75], catechin (6) [76], epicatechin (7) [76], 6-gingerol (11) [77], (−)-epigallocatechin (14) [78], isomajdine (15) [65], and boswellic acid alpha (2) [79], are known to have various beneficial activities on the cytotoxicity of African green monkey (Vero) kidney cells. Therefore, these compounds may therefore be partly responsible for the observed activity. According to the literature, the toxicity of Lippia multiflora has not been determined using Vero cells.

3. Materials and Methods

3.1. General Experimental Procedure

All solvents and chemicals used in this study were of analytical grade. African green monkey (Vero) kidney cells (ATCC CCL-81) were purchased from Cytiva Hyclone, Marlborough, MA, USA. Minimal essential medium (MEM), Earle’s balanced salt solution (EBSS), L-glutamine, fetal bovine serum (FBS), penicillin/streptomycin (P/S), trypsin/EDTA, and trypan blue were from (Cytiva Hyclone, Marlborough, MA, USA). Dichloromethane, methanol, formic acid, and acetonitrile were purchased from Sigma-Aldrich (St. Louis, MO, USA). Dimethyl sulfoxide (DMSO), ascorbic acid, quercetin, gallic acid, 2,2-diphenyl-1-picrylhydrazyl (DPPH), potassium hexacyanoferrate [K3Fe (CN)6], trichloroacetic acid, ferric chloride, Folin–Ciocalteu, sodium carbonate (Na2CO3), aluminum chloride, and thiazolyl blue tetrazolium bromide were purchased from Sigma-Aldrich (Darmstadt, Germany). Doxorubicin was purchased from Merck (Darmstadt, Germany).

3.2. Sampling and Extraction

The various plant parts (root, stem bark, and leaf) of Lippia multiflora were collected at Uíge Province, Angola. Identification of the species was carried out by Mr. Monizi Mawunu of the Department of Agronomy, Polytechnic Institute, Kimpa Vita University, Angola. A voucher herbarium specimen, NM 01, was assigned, and the voucher was deposited in the herbarium of the Department of Agronomy. The samples were air-dried for four weeks and then ground to a fine powder using a POLYMIX® lab mill with blade grinding, PX-MFC 90 D (Westdene, Bloemfontein, South Africa). Approximately ~220.22 g ground root and ~213.55 g stem bark of Lippia multiflora were each macerated in methanol (2 L) for two days, respectively. The stem bark and root extracts were filtered and then concentrated at 45 °C using a Büchi® Rotavapor® (Sigma-Aldrich, St. Louis, MO, USA) to obtain 4.19 g and 2.65 g of solid crude extracts. Moreover, ~217.58 g ground leaves of Lippia multiflora were macerated in dichloromethane (2 L) and methanol (2 L) for two days. The leaf extracts were filtered and then concentrated at 45 °C using a Büchi® Rotavapor® (Sigma-Aldrich, St. Louis, MO, USA) to obtain 3.53 g dichloromethane extract and 6.56 g methanol extract.

3.3. Total Flavonoid Content (TFC)

The total flavonoid content (TFC) of the extracts was determined by the aluminum chloride (AlCl3) method in Tlhapi (2018) [80]. The crude extracts (100 µL) were added in triplicate in 96-well plates containing 80 µL of distilled water, followed by an addition of aluminum chloride (2%, 100 µL) solution. The mixture was left to stand for 30 min, and the absorbance was read at 420 nm using a SoftMaxR Pro 6 (version 6.3) microplate reader. Quercetin was used as a standard to convert the absorbance of the crude extract to total flavonoid content expressed in milligram quercetin equivalent per gram of the crude extract (mg QE/g). The total flavonoid content of the crude extracts was determined using the aluminum chloride (AlCl3) colorimetric method. Quercetin was used to formulate a standard curve (y = 0.0865x + 3.3762; R2 = 0.9973; Figure S1, Supplementary Material).
The extracts were calculated using the following equation:
C = c × V/m
  • C = total amount of compounds.
  • c = concentration of standard from the standard curve (mg/mL).
  • V = volume of the extract (mL).
  • m = weight of extract (g).

3.4. Total Phenolic Content (TPC)

The total polyphenolic contents (TPCs) of the extracts were determined by the Folin–Ciocalteu method as seen in Tlhapi (2018) [80]. The crude extracts (100 µL) were added in triplicate to a 96-well plate containing 80 µL of distilled water. A Folin–Ciocalteu reagent (10%; 20 µL) and 60 µL of 7% sodium carbonate (Na2CO3) solution were added to the mixture, respectively. The absorbance was read at 760 nm using a SoftMaxR Pro 6 (version 6.3) microplate reader. The gallic acid standard curve was used to convert the absorbance of the crude extracts to total phenolic content expressed in milligram gallic acid equivalent per gram of the crude extract (mg GAE/G). The total phenolic content in crude extracts was estimated using Folin–Ciocalteu’s reagent. Gallic acid was used to formulate a calibration curve (y = 1.0372x + 1.2487; R2 = 0.9076; see Figure S2, Supplementary Material). The extracts were calculated using Equation (1).

3.5. UHPLC-Q Exactive-Orbitrap-MS Analysis

The crude extracts were identified using ultra-high-performance liquid chromatography coupled with quadrupole-orbitrap MS (UHPLC-Q-orbitrap MS, Thermo Fisher Scientific, Waltham, MA, USA). LC-ESI-Orbitrap-MS analysis was executed using a 4.6 mm × 150 mm, particle size 3.5 μm C18 analytical column to separate the extracts. Mobile phases were formic acid (0.1 percent v/v) in water (solvent A) and formic acid (0.1 percent v/v) in acetonitrile (solvent B). The linear gradient elution began with five percent of solvent B and increased to a hundred percent of solvent B for twenty minutes. A sample injection volume of ten microliters, a mobile phase flow rate of 0.9 mL min−1, and a column temperature of 25 °C were used. Data processing was determined using the XCaliber (Thermo Fisher Scientific Inc., Waltham, MA, USA, version 3.0 software). Both negative (ESI (−), Figure S3, Supplementary Material) and positive (ESI (+), Figure S4, Supplementary Material) modes were evaluated; however, the negative (ESI (−)) mode was chosen for additional sample analysis since it resulted in a larger abundance of ions. Moreover, the mass spectrometry raw data files were converted to an open-source format (.mzXML) using ProteoWizard tool MSConvertGUI (version 3.0.24164-38d6037) software. The mzXML files were evaluated using MZmine version 3.9.0 software; centroid techniques such as mass detection, chromatograph building, and peak deconvolution were used to process settings of MZmine version 3.9.0. The aligned peak list, which contained the retention time, m/z values, and peak heights in each sample, was exported in CSV format. The exported file was uploaded to MassBank, an online database for compound identification. Phytochemicals with a match of ≥80% in the database were considered; the identified compounds were extracted from the sample chromatogram data for further validation.

3.6. Antioxidant Activity Assays

3.6.1. Free Radical Scavenging Assay (DPPH)

The DPPH free radical scavenging ability was evaluated for the crude extracts using the DPPH (2,2-diphenyl-1-picrylhydrazyl) assay using a modified spectrophotometric method of Motamed and Naghibi (2010) [81]. The crude extracts (100 µL) were added to a 96-well plate containing distilled water (100 µL). After serial dilution, a 0.3 M DPPH/methanol (200 µL) solution was added to the mixture. The mixture was left at room temperature for 30 min, and the absorbance was measured using a SoftMaxR Pro 6 (version 6.3) microplate reader at 517 nm. All the crude extracts were tested in triplicate.
The percentage of radical scavenging was calculated by the following formula:
% Free RSA = [(ADPPH − Asample)/(ADPPH)] × 100
where ADPPH is the absorbance of the DPPH solution and Asample is the absorbance of the samples and DPPH. The ability of the crude extracts to inhibit 50% of the free radical (IC50) was extrapolated from a graph of % RSA against concentration.

3.6.2. Reducing Power

The reducing capacity of the crude extracts was estimated as described by Pereira et al. (2013) [82]. Fifty microliters of the crude extracts were mixed with fifty microliters of sodium phosphate buffer (0.2 M, pH 6.6). After serial dilution, a potassium hexacyanoferrate (1% aqueous, 50 µL) solution was added. A trichloroacetic acid (10%, 50 µL) solution was added after 20 min of incubation at 50 °C. Eighty microliters of each mixture was moved to another 96-well plate containing eighty microliters of distilled water. Ferric chloride (0.1%, w/v; 16 µL) was added, and the absorbance was measured using a SoftMaxR Pro 6 (version 6.3) microplate reader at 700 nm. All the crude extracts were tested in triplicate.

3.7. Cytotoxicity Assay

3.7.1. Cell Culture

African green monkey (Vero) kidney cells (ATCC CCL-81, Cellonex, Johannesburg, South Africa) were cultured in Minimal Essential Medium (MEM), which consisted of Earle’s Balanced Salt Solution (EBSS) and L-Glutamine (2.0 mM) (Cytiva Hyclone, Marlborough, MA, USA) and was supplemented with 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin (P/S, Biowest, Nuaille, France). These cells were incubated for 48 h at 37 °C in a 95% air and 5% CO2 humidified incubator (Nϋve, Ankara, Turkey). The cells were trypsinized with 0.25% trypsin/EDTA (Cytiva Hyclone, Marlborough, MA, USA) and split at a ratio of 1:5 for further passaging at 70 to 80% confluency. Trypan blue 0.4% (Cytiva Hyclone, Marlborough, MA, USA) on an automated cell counter (NanoEntek, Guro-gu, Seoul, Republic of Korea) was used to check the cell viability, and only suspensions with a cell viability higher than 90% were used for the cytotoxicity assay.

3.7.2. Cytotoxicity Assay

The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) cytotoxicity assay was determined as described by Mariri et al. (2024) [83]. The cytotoxic effects of the crude extracts of Lippia multiflora were evaluated against Vero cells. The Vero cells were seeded at a density of ten thousand cells on the 96-well microliter plates, and the cells were allowed to attach overnight. After incubation, the cells were treated with varying concentrations of 50 µg/mL, 100 µg/mL, 250 µg/mL, and 500 µg/mL dissolved in dimethyl sulfoxide and further diluted in fresh culture medium. In each test, the concentration of dimethyl sulfoxide in the medium did not surpass 0.5% (negative control). Doxorubicin was used as a positive control at varying concentrations of 0.67 µg/mL, 1.35 µg/mL, 2.71 µg/mL, and 5.43 µg/mL. The plates were further incubated for two days; thereafter, the culture medium containing tested samples was discarded and replaced by two hundred microliters of fresh medium with thirty microliters of thiazolyl blue tetrazolium bromide (5 mg/mL) dissolved in phosphate buffered saline (PBS). After 4 h of incubation, the culture medium was gently aspirated, and the formazan crystals were dissolved in fifty microliters of dimethyl sulfoxide. The mixture was kept in the dark for 15 min, and the absorbance was measured spectrophotometrically at 570 nm on a SpectraMax iD3 multi-mode microplate reader (Winooski, VT, USA). The cell viability was calculated as the percentage (%) of negative control (0.5% dimethyl sulfoxide; see Equation (3)).
The extracts were calculated using the following equation:
C e l l   v i a b i l i t y % = A b s s a m p l e A b s B l a n k A b s n e g a t i v e   c o n t r o l A b s B l a n k × 100
where Abs is the absorbance.

3.8. Statistical Analysis

Statistical analysis was carried out using the IBM SPSS Statistics package, version 22 (Chicago, IL, USA). For the antioxidant, total flavonoid content, total phenolic content, and cytotoxicity tests, the measured data were obtained in triplicate, and the data were expressed as mean ± standard deviation. Mean differences of the crude extracts were assessed by a one-way analysis of variance (ANOVA) with Duncan’s Multiple Range Test (DMRT) in the antioxidant, total flavonoid content, and total phenolic content tests; the difference with p < 0.05 was considered statistically significant. A principal component analysis (PCA) biplot was prepared with both Y and X scaling at 10 to −10 and 95% ellipses; the eigenvalue scale was also used. Hierarchical clustering (prepared with the X scaling at 0 to 5, Y scaling at 0 to −0.3223, and 95% ellipses), classical clustering plot, neighbor-joining cluster plot, and the correlation plot were performed using the PAST 4.03 software. A Pearson correlation analysis was conducted using the IBM SPSS Statistics package, version 22 (Chicago, IL, USA), to evaluate the relationship between phytochemicals (TFC and TPC) and their antioxidant activities. All the extracts were analyzed in triplicate for the PCA and correlation plot methods. For cytotoxicity, the data were shown as the Vero cell number ± standard deviation at varying concentrations. Doxorubicin was used as a positive control drug standard for the cytotoxicity assay.

4. Conclusions

In this study, the phytochemical composition, total flavonoid content, total phenolic content, and the antioxidant and cytotoxic potentials of different organs of Lippia multiflora were evaluated. UHPLC–Q/Orbitrap/MS was used to detect 20 metabolites. The metabolite profiles showed that different plant parts of L. multiflora produce similar and dissimilar metabolites. Furthermore, the multivariate analysis of the data obtained showed that the chemistry of the root extracts and stem bark extracts is similar and different from the chemistry of the methanol leaf extracts and dichloromethane leaf extracts. The study demonstrated that the leaf and stem bark crude extracts had a greater antioxidant capacity compared to the root extract. The study also demonstrated that the root extract was more toxic at varying concentrations compared to the stem bark and leaf extracts. Furthermore, the efficient antioxidant activities were supported by the presence of a good amount of total flavonoid and phenolic content in the extracts. Our results highlight that this species provides a promising source for obtaining phytoconstituents that might be used to develop drugs that can be used in the treatment of infectious diseases. Bioassay-guided fractionation and isolation of phytochemicals are required to discover other bioactive compounds contributing to the biological activities of L. multiflora. Finally, in vitro and in vivo toxicological and pharmacological studies using different in vitro and in vivo cytotoxicity assays must be performed to confirm the safety of the identified compounds before considering Lippia multiflora as a viable candidate for drug development. Nonetheless, our results may represent a promising milestone in the medical or pharmaceutical industry’s search for new and effective therapies.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/molecules30132882/s1, Figure S1: Regression equation of quercetin calibration curve; Figure S2: Regression equation of gallic acid calibration curve; Figure S3: UHPLC–Q/Orbitrap/MS HRMS chromatograms with relative abundance and retention time (min) of (A) root; (B) stem bark; (C) dichloromethane leaf; and (D) methanol leaf extracts of Lippia multiflora obtained in negative mode electrospray ionization; Figure S4: UHPLC–Q/Orbitrap/MS HRMS chromatograms with relative abundance and retention time (min) of (A) root; (B) stem bark; (C) dichloromethane leaf; and (D) methanol leaf extracts of Lippia multiflora obtained in positive mode electrospray ionization; Figure S5: UHPLC–Q/Orbitrap/MS HRMS spectrum showing fragments of poricoic acid B (1); Figure S6: UHPLC–Q/Orbitrap/MS HRMS spectrum showing fragments of boswellic acid alpha (2); Figure S7: UHPLC–Q/Orbitrap/MS HRMS spectrum showing fragments of eburicoic acid (3); Figure S8: UHPLC–Q/Orbitrap/MS HRMS spectrum showing fragments of medicagenic acid (4); Figure S9: UHPLC–Q/Orbitrap/MS HRMS spectrum showing fragments of albiflorin (5); Figure S10: UHPLC–Q/Orbitrap/MS HRMS spectrum showing fragments of catechin (6); Figure S11: UHPLC–Q/Orbitrap/MS HRMS spectrum showing fragments of epicatechin (7); Figure S12: UHPLC–Q/Orbitrap/MS HRMS spectrum showing fragments of formononetin-7-O-glucoside (Ononin) (8); Figure S13: UHPLC–Q/Orbitrap/MS HRMS spectrum showing fragments of 3′,4′-dimethoxy-7-hydroxyflavone (9); Figure S14: UHPLC–Q/Orbitrap/MS HRMS spectrum showing fragments of quercitrin (10); Figure S15: UHPLC–Q/Orbitrap/MS HRMS spectrum showing fragments of 6-gingerol (11); Figure S16: UHPLC–Q/Orbitrap/MS HRMS spectrum showing fragments of 6-paradol (12); Figure S17: UHPLC–Q/Orbitrap/MS HRMS spectrum showing fragments of esmolol (13); Figure S18: UHPLC–Q/Orbitrap/MS HRMS spectrum showing fragments of (−)-epigallocatechin (14); Figure S19: UHPLC–Q/Orbitrap/MS HRMS spectrum showing fragments of isomajdine (15); Figure S20: UHPLC–Q/Orbitrap/MS HRMS spectrum showing fragments of yohimbinic acid (16); Figure S21: UHPLC–Q/Orbitrap/MS HRMS spectrum showing fragments of reserpic acid (17); Figure S22: UHPLC–Q/Orbitrap/MS HRMS spectrum showing fragments of 6,7-dihydroxycoumarin-6-glucoside (Esculin) (18); Figure S23: UHPLC–Q/Orbitrap/MS HRMS spectrum showing fragments of eicosadieneoic acid (19); Figure S24: UHPLC–Q/Orbitrap/MS HRMS spectrum showing fragments of dodecanedioic acid (20).

Author Contributions

Conceptualization, D.T., N.M., I.T.M., and M.M.; methodology, D.T.; UHPLC–Q/Orbitrap/MS analysis, R.C.C.; writing—original draft preparation, D.T.; writing—review and editing, D.T., N.M., I.T.M., M.M., and R.C.C. All authors have read and agreed to the published version of the manuscript.

Funding

This work was financially supported by Central University of Technology, Free State’s University Research Grants and Scholarship Committee (RGSC). The German Federal Ministry of Education and Research (BMBF)/Southern African Science Service Centre for Climate Change and Adaptative Land Management (SASSCAL) is also acknowledged for funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article and Supplementary Materials.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Oladimeji, F.A.; Orafidiya, L.O.; Okeke, I.N. Physical properties and antimicrobial activities of leaf essential oil of Lippia multiflora Moldenke. Int. J. Aromather. 2004, 14, 162–168. [Google Scholar] [CrossRef]
  2. Samba, N.; Aitfella-Lahlou, R.; Nelo, M.; Silva, L.; Coca, R.; Rocha, P.; Lopez Rodilla, J.M. Chemical composition and antibacterial activity of Lippia multiflora Moldenke essential oil from different regions of Angola. Molecules 2020, 26, 155. [Google Scholar] [CrossRef]
  3. Ngaibi, J.; Kandeda, A.K.; Nguezeye, Y.; Wangbara, T.A.; Gaoudji, L.; Taiwe, G.S.; Bum, E.N. Antiepileptic and anti-inflammatory effects of Lippia multiflora moldenke (Verbenaceae) in mice model of chronic temporal lobe epilepsy induced by pilocarpine. Heliyon 2024, 10, e39483. [Google Scholar] [CrossRef]
  4. Folashade, K.O.; Omoregie, E.H. Essential oil of Lippia multiflora Moldenke: A review. J. Appl. Pharm. Sci. 2012, 2, 15–23. [Google Scholar]
  5. Mawunu, M.; António, D.; Vita, P.; Ngbolua, K.N.; Luyeye, L.; Ndiku, L.; Luzolawo, M.P.; Francisco, N.M. Ethnobotanical Survey of Herbal Teas Consumed in Uíge Province, Angola: Part 1. Ethnobot. Res. Appl. 2023, 26, 23. [Google Scholar] [CrossRef]
  6. Mawunu, M.; Makuntima, P.; Masidivinga, L.; Lautenschläger, T.; Luyindula, N.; Ngbolua, K.N.; Lukoki, L. First Survey on the Edible Non-Wood Forest Products Sold in Uíge Province, Northern Angola. Eur. J. Agric. Food Sci. 2020, 2, 6. [Google Scholar] [CrossRef]
  7. Lautenschläger, T.; Monizi, M.; Pedro, M.; Mandombe, J.L.; Bránquima, M.F.; Heinze, C.; Neinhuis, C. First large-scale ethnobotanical survey in the province of Uíge, Northern Angola. J. Ethnobiol. Ethnomed. 2018, 14, 513. [Google Scholar] [CrossRef]
  8. Oladimeji, F.A.; Orafidiya, O.O.; Ogunniyi, T.A.B.; Adewunmi, T.A. Pediculocidal and scabicidal properties of Lippia multiflora essential oil. J. Ethnopharmacol. 2000, 72, 305–311. [Google Scholar] [CrossRef]
  9. Sakyiamah, M.M.; Baffour, P.K.; Atta-Adjei, P.; Bolah, P.; Ehun, E.; Opare, C.; Brew-Daniels, H.; Appiah, A.A. Effect of seasonal variations on the secondary metabolites and antioxidant activities of Bridelia ferruginea, Lippia multiflora, and Azadirachta indica leaves. J. Pharmacogn. Phytother. 2024, 16, 1–13. [Google Scholar]
  10. Abena, A.A.; Atipo-Ebata, J.K.; Diatewa, M. Psychopharmacological properties of crude extract and essential oil of Lippia multiflora. L’encephale 2001, 27, 360–364. [Google Scholar]
  11. Abena, A.A.; Diatewa, M.; Gakosso, G.; Gbeassor, M.; Hondi-Assah, T.H.; Ouamba, J.M. Analgesic, antipyretic and anti-inflammatory effects of essential oil of Lippia multiflora. Fitoterapia 2003, 74, 231–236. [Google Scholar] [CrossRef]
  12. Oussou, K.R.; Yolou, S.; Boti, J.B.; Guessennd, K.N.; Kanko, C.; Ahibo, C.; Casanova, J. Etude chimique et activité antidiarrheique des huiles essentielles de deux plantes aromatiques de la pharmacopée ivoirienne. Eur. J. Sci. Res. 2008, 24, 94–103. [Google Scholar]
  13. Masunda, A.T.; Inkoto, C.L.; Masengo, C.A.; Bongili, S.B.; Basilua, J.P.K.; Legbiye, E.M.; Mpiana, P.T. Traditional uses, physical properties, phytochemistry and bioactivity of Lippia multiflora Moldenke (Verbenaceae): A Mini-review. Dis. Phytomed. 2020, 7, 19–26. [Google Scholar]
  14. Koïta, K.; Baissac, Y.; Sanon, E.; Campa, C.; Sankara, P. Phenolics from Lippia multiflora Moldenke as potential bioactive agents against peanut pathogens. Res. J. Agric. Sci. 2017, 7, 267–276. [Google Scholar]
  15. Kunle, O.; Okogun, J.; Egamana, E.; Emojevwe, E.; Shok, M. Antimicrobial activity of various extracts and carvacrol from Lippia multiflora leaf extract. Phytomedicine 2003, 10, 59–61. [Google Scholar] [CrossRef] [PubMed]
  16. Xu, D.P.; Li, Y.; Meng, X.; Zhou, T.; Zhou, Y.; Zheng, J.; Zhang, J.J.; Li, H.B. Natural antioxidants in foods and medicinal plants: Extraction, assessment and resources. Int. J. Mol. Sci. 2017, 18, 96. [Google Scholar] [CrossRef]
  17. Collin, F. Chemical basis of reactive oxygen species reactivity and involvement in neurodegenerative diseases. Int. J. Mol. Sci. 2019, 20, 2407. [Google Scholar] [CrossRef]
  18. Demirci-Cekic, S.; Özkan, G.; Avan, A.N.; Uzunboy, S.; Çapanoğlu, E.; Apak, R. Biomarkers of oxidative stress and antioxidant defense. J. Pharm. Biomed. Anal. 2022, 209, 114477. [Google Scholar] [CrossRef]
  19. Limmongkon, A.; Janhom, P.; Amthong, A.; Kawpanuk, M.; Nopprang, P.; Poohadsuan, J.; Somboon, T.; Saijeen, S.; Surangkul, D.; Srikummool, M.; et al. Antioxidant activity, total phenolic, and resveratrol content in five cultivars of peanut sprouts. Asian Pac. J. Trop. Biomed. 2017, 7, 332–338. [Google Scholar] [CrossRef]
  20. Škrovánková, S.; Mišurcová, L.; Machů, L. Antioxidant activity and protecting health effects of common medicinal plants. Adv. Food Nutr. Res. 2012, 67, 75–139. [Google Scholar]
  21. Engwa, G.A. Free radicals and the role of plant phytochemicals as antioxidants against oxidative stress-related diseases. In Phytochemicals: Source of Antioxidants and Role in Disease Preventio; Intech: New York, NY, USA, 2018; Volume 7, pp. 49–74. [Google Scholar]
  22. Simunkova, M.; Barbierikova, Z.; Jomova, K.; Hudecova, L.; Lauro, P.; Alwasel, S.H.; Alhazza, I.; Rhodes, C.J.; Valko, M. Antioxidant vs. prooxidant properties of the flavonoid, kaempferol, in the presence of Cu (II) ions: A ROS-scavenging activity, fenton reaction and DNA damage study. Int. J. Mol. Sci. 2021, 22, 1619. [Google Scholar] [CrossRef]
  23. Dabire, C.M.; Bationo, R.K.; Hema, A.; Nebie, R.C.; Pale, E.; Dhanabal, S.P.; Nacro, M. Total phenolics content, flavonoids profiling and antioxidant activity of Lippia multiflora leaves extracts from Burkina Faso. Asian J. Plant Sci. Res. 2015, 5, 28–33. [Google Scholar]
  24. Guo, J.; Zhang, D.; Yu, C.; Yao, L.; Chen, Z.; Tao, Y.; Cao, W. Phytochemical analysis, antioxidant and analgesic activities of Incarvillea compacta Maxim from the Tibetan plateau. Molecules 2019, 24, 1692. [Google Scholar] [CrossRef] [PubMed]
  25. Nwozo, O.S.; Effiong, E.M.; Aja, P.M.; Awuchi, C.G. Antioxidant, phytochemical, and therapeutic properties of medicinal plants: A review. Int. J. Food Prop. 2023, 26, 359–388. [Google Scholar] [CrossRef]
  26. Burlec, A.F.; Pecio, Ł.; Mircea, C.; Cioancă, O.; Corciovă, A.; Nicolescu, A.; Oleszek, W.; Hăncianu, M. Chemical profile and antioxidant activity of Zinnia elegans Jacq. fractions. Molecules 2019, 24, 2934. [Google Scholar] [CrossRef] [PubMed]
  27. Burlec, A.F.; Pecio, Ł.; Kozachok, S.; Mircea, C.; Corciovă, A.; Vereștiuc, L.; Cioancă, O.; Oleszek, W.; Hăncianu, M. Phytochemical profile, antioxidant activity, and cytotoxicity assessment of Tagetes erecta L. flowers. Molecules 2021, 26, 1201. [Google Scholar] [CrossRef] [PubMed]
  28. Mehmood, A.; Javid, S.; Khan, M.F.; Ahmad, K.S.; Mustafa, A. In vitro total phenolics, total flavonoids, antioxidant and antibacterial activities of selected medicinal plants using different solvent systems. BMC Chem. 2022, 16, 64. [Google Scholar] [CrossRef]
  29. Itam, A.; Wulandari, A.; Rahman, M.M.; Ferdinal, N. Preliminary phytochemical screening, total phenolic content, antioxidant and cytotoxic activities of Alstonia scholaris R. Br leaves and stem bark extracts. J. Pharm. Sci. Res. 2018, 10, 518–522. [Google Scholar]
  30. Okselni, T.; Santoni, A.; Dharma, A.; Efdi, M. Determination of antioxidant activity, total phenolic content, and total flavonoid content of roots, stem bark, and leaves of Elaeocarpus mastersii king. Rasayan J. Chem. 2018, 11, 1211–1216. [Google Scholar] [CrossRef]
  31. Syaputra, N.D.; Dewi, K.C.; Lili, W.; Agung, M.U.K. Total phenolic, flavonoid content and antioxidant capacity of stem bark, root, and leaves methanolic extract of Rhizophora mucronata Lam. World News Nat. Sci. 2019, 26, 118–128. [Google Scholar]
  32. Gupta, S.; Acharya, R.; Gamit, R.V.; Shukla, V.J. Quantitative analysis of tannins, alkaloids, phenols, and flavonoids in Ficus semicordata leaf, stem, stem bark, root, and fruit powder. J. Indian. Sys. Med. 2021, 9, 171–174. [Google Scholar] [CrossRef]
  33. Dirar, A.I.; Alsaadi, D.H.M.; Wada, M.; Mohamed, M.A.; Watanabe, T.; Devkota, H.P. Effects of extraction solvents on total phenolic and flavonoid contents and biological activities of extracts from Sudanese medicinal plants. S. A. J. Bot. 2019, 120, 261–267. [Google Scholar] [CrossRef]
  34. Neupane, P.; Lamichhane, J. Estimation of total phenolic content, total flavonoid content and antioxidant capacities of five medicinal plants from Nepal. Vegetos 2020, 33, 360–366. [Google Scholar] [CrossRef]
  35. Dias, M.C.; Pinto, D.C.; Silva, A.M. Plant flavonoids: Chemical characteristics and biological activity. Molecules 2021, 26, 5377. [Google Scholar] [CrossRef]
  36. Yu, M.; Gouvinhas, I.; Rocha, J.; Barros, A.I. Phytochemical and antioxidant analysis of medicinal and food plants towards bioactive food and pharmaceutical resources. Sci. Rep. 2021, 11, 10041. [Google Scholar] [CrossRef] [PubMed]
  37. Shen, N.; Wang, T.; Gan, Q.; Liu, S.; Wang, L.; Jin, B. Plant flavonoids: Classification, distribution, biosynthesis, and antioxidant activity. Food Chem. 2022, 383, 132531. [Google Scholar] [CrossRef]
  38. Rouamba, A.; Badini, D.; Compaoré, E.; Ouédraogo, V.; Kiendrebeogo, M. Lippia multiflora leaves extracts enhance cefotaxime bactericidal effects and quench the biofilm formation in methicillin-resistant Staphylococcus aureus ATCC 43300. Avicenna J. Med. Biotechnol. 2024, 16, 193. [Google Scholar] [CrossRef]
  39. Phuyal, N.; Jha, P.K.; Raturi, P.P.; Rajbhandary, S. Total phenolic, flavonoid contents, and antioxidant activities of fruit, seed, and bark extracts of Zanthoxylum armatum DC. Sci. World J. 2020, 2020, 8780704. [Google Scholar] [CrossRef]
  40. Koraqi, H.; Petkoska, A.T.; Khalid, W.; Sehrish, A.; Ambreen, S.; Lorenzo, J.M. Optimization of the extraction conditions of antioxidant phenolic compounds from strawberry fruits (Fragaria x ananassa Duch.) using response surface methodology. Food Anal. Methods 2023, 16, 1030–1042. [Google Scholar] [CrossRef]
  41. Saeed, N.; Khan, M.R.; Shabbir, M. Antioxidant activity, total phenolic and total flavonoid contents of whole plant extracts Torilis leptophylla L. BMC Complement. Alt. Med. 2012, 12, 221. [Google Scholar]
  42. Desmiaty, Y.; Saputri, F.C.; Hanafi, M.; Prastiwi, R.; Elya, B. Anti-elastase, anti-tyrosinase and anti-oxidant of Rubus fraxinifolius stem methanolic extract. Pharmacogn. J. 2020, 12, 271–275. [Google Scholar] [CrossRef]
  43. Mannino, G.; Occhipinti, A.; Maffei, M.E. Quantitative determination of 3-O-acetyl-11-keto-β-boswellic acid (AKBA) and other boswellic acids in Boswellia sacra Flueck and Boswellia serrata Roxb. Molecules 2016, 21, 1329. [Google Scholar] [CrossRef] [PubMed]
  44. Nam, D.G.; Yang, H.S.; Bae, U.J.; Park, E.; Choi, A.J.; Choe, J.S. The Cactus (Opuntia ficus-indica) Cladodes and Callus extracts: A study combined with LC-MS metabolic profiling, in-silico, and in-vitro analyses. Antioxidants 2023, 12, 1329. [Google Scholar] [CrossRef]
  45. Pollier, J.; Morreel, K.; Geelen, D.; Goossens, A. Metabolite profiling of triterpene saponins in Medicago truncatula hairy roots by liquid chromatography Fourier transform ion cyclotron resonance mass spectrometry. J. Nat. Prod. 2011, 74, 1462–1476. [Google Scholar] [CrossRef]
  46. Wang, S.; Wang, H.; Liu, Y.; Wang, Y.; Fan, X.; Cheng, Y. Rapid discovery and identification of anti-inflammatory constituents from traditional Chinese medicine formula by activity index, LC-MS, and NMR. Sci. Rep. 2016, 6, 31000. [Google Scholar] [CrossRef]
  47. Liu, W.; Huang, J.; Zhang, F.; Zhang, C.C.; Li, R.-S.; Wang, Y.-L.; Wang, C.-R.; Liang, X.M.; Zhang, W.D.; Yang, L. Comprehensive profiling and characterization of the absorbed components and metabolites in mice serum and tissues following oral administration of Qing-Fei-Pai-Du decoction by UHPLC-Q-Exactive-Orbitrap HRMS. Chin. J. Nat. Med. 2021, 19, 305–320. [Google Scholar] [CrossRef]
  48. Araujo, N.M.P.; Arruda, H.S.; Dos Santos, F.N.; de Morais, D.R.; Pereira, G.A.; Pastore, G.M. LC-MS/MS screening and identification of bioactive compounds in leaves, pulp and seed from Eugenia calycina Cambess. Food Res. Int. 2020, 137, 109556. [Google Scholar] [CrossRef] [PubMed]
  49. Tava, A.; Biazzi, E.; Ronga, D.; Mella, M.; Doria, F.; D’Addabbo, T.; Candido, V.; Avato, P. Chemical identification of specialized metabolites from sulla (Hedysarum coronarium L.) collected in southern Italy. Molecules 2021, 26, 4606. [Google Scholar] [CrossRef]
  50. Tine, Y.; Yang, Y.; Renucci, F.; Costa, J.; Wélé, A.; Paolini, J. LC-MS/MS analysis of flavonoid compounds from Zanthoxylum zanthoxyloides extracts and their antioxidant activities. Nat. Prod. Commun. 2017, 12, 1865–1868. [Google Scholar] [CrossRef]
  51. Ali, A.; Bashmil, Y.M.; Cottrell, J.J.; Suleria, H.A.; Dunshea, F.R. LC-MS/MS-QTOF screening and identification of phenolic compounds from Australian grown herbs and their antioxidant potential. Antioxidants 2021, 10, 1770. [Google Scholar] [CrossRef]
  52. Picardo, M.; Sanchís, J.; Núñez, O.; Farré, M. Suspect screening of natural toxins in surface and drinking water by high performance liquid chromatography and high-resolution mass spectrometry. Chemosphere 2002, 261, 127888. [Google Scholar] [CrossRef] [PubMed]
  53. Kumar, S.; Singh, A.; Bajpai, V.; Srivastava, M.; Singh, B.P.; Kumar, B. Structural characterization of monoterpene indole alkaloids in ethanolic extracts of Rauwolfia species by liquid chromatography with quadrupole time-of-flight mass spectrometry. J. Pharm. Anal. 2016, 6, 363–373. [Google Scholar] [CrossRef]
  54. Smitha, C.K.; Udayan, P.S. GC-MS and HR-LCMS fingerprinting of various parts of Oroxylum indicum (L.) Vent. A comparative phytochemical study based on plant part substitution approach. J. Pharmacogn. Phytochem. 2020, 9, 1817–1824. [Google Scholar]
  55. Servi, H.; Kisa, Ö.; Aysal, A.I.; ERKÖSE GENÇ, G.; Şatana, D. Chemical profile by LC-Q-TOF-MS of Nigella sativa seed extracts and in vitro antimicrobial activity on bacteria which are determined resistance gene and isolated from nosocomial infection. J. Res. Pharm. 2022, 26, 287. [Google Scholar] [CrossRef]
  56. Li, J.; Chen, Y.; Yu, K.; Zhang, M.; Li, Q.; Tang, S.; Liu, Y.; Li, H.; Zhang, Z. Rapid chemical characterization and pharmacological mechanism of Fining Granules in the treatment of chronic bronchitis based on UHPLC–Q-exactive orbitrap mass spectrometer and network pharmacology. Heliyon 2024, 10, e31804. [Google Scholar] [CrossRef] [PubMed]
  57. Zhang, L.; Yin, M.; Feng, X.; Ibrahim, S.A.; Liu, Y.; Huang, W. Anti-inflammatory activity of four triterpenoids isolated from poriae cutis. Foods 2021, 10, 3155. [Google Scholar] [CrossRef] [PubMed]
  58. Sheth, K.; Catalfomo, P.; Sciuchetti, L.A. Isolation and identification of eburicoic acid from Fomes pinicola. J. Pharm. Sci. 1967, 56, 1656–1658. [Google Scholar] [CrossRef]
  59. Timbekova, A.E.; Isaev, M.I.; Abubakirov, N.K. Chemistry and biological activity of triterpenoid glycosides from Medicago sativa. In Saponins Used in Food and Agriculture; Waller, G.R., Yamasaki, K., Eds.; Advances in Experimental Medicine and Biology; Springer: Boston, MA, USA, 1996; Volume 405, pp. 171–182. [Google Scholar] [CrossRef]
  60. Wu, Y.; Jiang, Y.; Zhang, L.; Zhou, J.; Yu, Y.; Zhou, Y.; Kang, T. Chemical profiling and antioxidant evaluation of Paeonia lactiflora Pall. “Zhongjiang” by HPLC–ESI–MS combined with DPPH assay. J. Chromatogr. Sci. 2021, 59, 795–805. [Google Scholar] [CrossRef]
  61. Abdel-Aal, E.S.M.; Rabalski, I.; Mats, L.; Rai, I. Identification and quantification of anthocyanin and catechin compounds in purple tea leaves and flakes. Molecules 2022, 27, 6676. [Google Scholar] [CrossRef]
  62. Zgórka, G.; Maciejewska-Turska, M.; Makuch-Kocka, A.; Plech, T. In vitro evaluation of the antioxidant activity and chemopreventive potential in human breast cancer cell lines of the standardized extract obtained from the aerial parts of zigzag clover (Trifolium medium L.). Pharmaceuticals 2022, 15, 699. [Google Scholar] [CrossRef]
  63. Parolin Trindade, G.; Perez Pinheiro, G.; Silva, A.A.R.; Porcari, A.D.M.; Sawaya, A.C.H.F. Sources of variation of the chemical composition of Lippia origanoides Kunth (Verbenaceae). Nat. Prod. Res. 2025, 1–10. [Google Scholar] [CrossRef] [PubMed]
  64. Jug, U.; Naumoska, K.; Vovk, I. (−)-Epicatechin—An important contributor to the antioxidant activity of Japanese knotweed rhizome bark extract as determined by antioxidant activity-guided fractionation. Antioxidants 2021, 10, 133. [Google Scholar] [CrossRef]
  65. Gülçin, I.L.; Beydemir, Ş.; Topal, F.; Gagua, N.; Bakuridze, A.; Bayram, R.; Gepdiremen, A. Apoptotic, antioxidant and antiradical effects of majdine and isomajdine from Vinca herbacea Waldst. and kit. J. Enzym. Inhib. Med. Chem. 2012, 27, 587–594. [Google Scholar] [CrossRef]
  66. Wang, Y.H.; Avula, B.; Wang, M.; Sagi, S.; Cohen, P.A.; Maller, G.; DeSouza, R.; Khan, I.A. Determination of caffeine, yohimbic acid, corynanthine, yohimbine and rauwolscine from yohimbe plants and products using LC-UV-MS/MS. Planta Medica 2014, 80, 32. [Google Scholar] [CrossRef]
  67. Wang, C.C.; Chen, L.G.; Lee, L.T.; Yang, L.L. Effects of 6-gingerol, an antioxidant from ginger, on inducing apoptosis in human leukemic HL-60 cells. In Vivo (Athens Greece) 2003, 17, 641–645. [Google Scholar] [PubMed]
  68. Grzesik, M.; Naparło, K.; Bartosz, G.; Sadowska-Bartosz, I. Antioxidant properties of catechins: Comparison with other antioxidants. Food Chem. 2018, 241, 480–492. [Google Scholar] [CrossRef] [PubMed]
  69. Picos-Salas, M.A.; Leyva-López, N.; Bastidas-Bastidas, P.D.J.; Antunes-Ricardo, M.; Cabanillas-Bojórquez, L.A.; Angulo-Escalante, M.A.; Heredia, J.B.; Gutiérrez-Grijalva, E.P. Supercritical CO2 extraction of naringenin from Mexican oregano (Lippia graveolens): Its antioxidant capacity under simulated gastrointestinal digestion. Sci. Rep. 2024, 14, 1146. [Google Scholar] [CrossRef]
  70. González Güereca, M.C.; Soto Hernández, M.; Kite, G.; Martinez Vazquez, M. Antioxidant activity of flavonoids from the stem of the Mexican oregano (Lippia graveolens HBK var. berlandieri Schauer). Rev. Fitotec. Mex. 2007, 30, 43–49. [Google Scholar]
  71. Dhami, N.; Mishra, A.D. Phytochemical variation: How to resolve the quality controversies of herbal medicinal products? J. Herb. Med. 2015, 5, 118–127. [Google Scholar] [CrossRef]
  72. Gololo, S.S.; Shai, L.J.; Agyei, N.M.; Mogale, M.A. Effect of seasonal changes on the quantity of phytochemicals in the leaves of three medicinal plants from Limpopo province, South Africa. J. Pharm. Phytother. 2016, 8, 168–172. [Google Scholar]
  73. Tlhapi, D.; Ramaite, I.; Anokwuru, C.; van Ree, T.; Madala, N.; Hoppe, H. Effects of seasonal variation on phytochemicals contributing to the antimalarial and antitrypanosomal activities of Breonadia salicina using a metabolomic approach. Heliyon 2024, 10, e24068. [Google Scholar] [CrossRef]
  74. Gupta, M.; Singh, S.; Luqman, S.; Saikia, D.; Thomas, M.; Rout, P.K. Correlation of boswellic acids with antiproliferative, antioxidant and antimicrobial activities of topographically collected Boswellia serrata oleo-gum-resin. Phytomed. Plus 2022, 2, 100289. [Google Scholar] [CrossRef]
  75. Su, Y.C.; Liu, C.T.; Chu, Y.L.; Raghu, R.; Kuo, Y.H.; Sheen, L.Y. Eburicoic acid, an active triterpenoid from the fruiting bodies of basswood cultivated Antrodia cinnamomea, induces ER stress-mediated autophagy in human hepatoma cells. J. Trad. Complement. Med. 2012, 2, 312–322. [Google Scholar] [CrossRef]
  76. Meneses-Gutiérrez, C.L.; Hernández-Damián, J.; Pedraza-Chaverri, J.; Guerrero-Legarreta, I.; Téllez, D.I.; Jaramillo-Flores, M.E. Antioxidant capacity and cytotoxic effects of catechins and resveratrol oligomers produced by enzymatic oxidation against T24 human urinary bladder cancer cells. Antioxidants 2019, 8, 214. [Google Scholar] [CrossRef] [PubMed]
  77. de Lima, R.M.T.; Dos Reis, A.C.; de Oliveira Santos, J.V.; de Oliveira Ferreira, J.R.; Braga, A.L.; de Oliveira Filho, J.W.G.; de Menezes, A.A.P.M.; da Mata, A.M.O.F.; de Alencar, M.V.O.B.; do Nascimento Rodrigues, D.C.; et al. Toxic, cytogenetic and antitumor evaluations of [6]-gingerol in non-clinical in vitro studies. Biomed. Pharmacother. 2019, 115, 108873. [Google Scholar] [CrossRef]
  78. Weisburg, J.H.; Weissman, D.B.; Sedaghat, T.; Babich, H. In vitro cytotoxicity of epigallocatechin gallate and tea extracts to cancerous and normal cells from the human oral cavity. Basic. Clin. Pharmacol. Toxicol. 2004, 95, 191–200. [Google Scholar] [CrossRef]
  79. Jamshidi-Adegani, F.; Ghaemi, S.; Al-Hashmi, S.; Vakilian, S.; Al-Kindi, J.; Rehman, N.U.; Alam, K.; Al-Riyami, K.; Csuk, R.; Arefian, E.; et al. Comparative study of the cytotoxicity, apoptotic, and epigenetic effects of Boswellic acid derivatives on breast cancer. Sci. Rep. 2022, 12, 19979. [Google Scholar] [CrossRef] [PubMed]
  80. Tlhapi, B.D. Isolation and Structure Elucidation of Bioactive Compounds from Rauvolfia caffra Sond. Master’s Thesis, University of Venda, Thohoyandou, South Africa, 2018. [Google Scholar]
  81. Motamed, S.M.; Naghibi, F. Antioxidant activity of some edible plants of the Turkmen Sahra region in northern Iran. Food Chem. 2010, 119, 1637–1642. [Google Scholar] [CrossRef]
  82. Pereira, O.R.; Macias, R.I.; Perez, M.J.; Marin, J.J.; Cardoso, S.M. Protective effects of phenolic constituents from Cytisus multiflorus, Lamium album L. and Thymus citriodorus on liver cells. J. Funct. Foods 2013, 5, 1170–1179. [Google Scholar] [CrossRef]
  83. Mariri, N.G.; Mongalo, N.I.; Makhafola, T.J. The in vitro cytotoxicity, genotoxicity and LC-ToF-MS profiling of four South African plants with good antifungal activity. S. Afr. J. Bot. 2024, 174, 446–455. [Google Scholar] [CrossRef]
Figure 1. Total flavonoid content and total phenolic content of crude extracts. A different superscript letter indicates significant differences using a one-way ANOVA at p < 0.05. Data (n = 3) expressed as mean ± standard deviation. In the total flavonoid content test: a—methanol leaf extract was significantly different from the stem bark extract, root extract, and dichloromethane leaf extract; b—dichloromethane leaf extract was significantly different to all the crude extracts; c—stem bark extract was not significantly different from the root extract. In the total phenolic content test: a—methanol leaf extract was not significantly different from the stem bark extract; b—root extract was significantly different from the stem bark extract, methanol leaf extract, and dichloromethane leaf extract; c—dichloromethane leaf extract was significantly different from all the crude extracts.
Figure 1. Total flavonoid content and total phenolic content of crude extracts. A different superscript letter indicates significant differences using a one-way ANOVA at p < 0.05. Data (n = 3) expressed as mean ± standard deviation. In the total flavonoid content test: a—methanol leaf extract was significantly different from the stem bark extract, root extract, and dichloromethane leaf extract; b—dichloromethane leaf extract was significantly different to all the crude extracts; c—stem bark extract was not significantly different from the root extract. In the total phenolic content test: a—methanol leaf extract was not significantly different from the stem bark extract; b—root extract was significantly different from the stem bark extract, methanol leaf extract, and dichloromethane leaf extract; c—dichloromethane leaf extract was significantly different from all the crude extracts.
Molecules 30 02882 g001
Figure 2. (A) PCA biplot of the extracts and the compounds that were tentatively identified. (B) The classical clustering plot of the extracts. (C) Neighbor-joining cluster plot of the extract. (D) Correlation plot of the compounds that were tentatively identified in the extracts.
Figure 2. (A) PCA biplot of the extracts and the compounds that were tentatively identified. (B) The classical clustering plot of the extracts. (C) Neighbor-joining cluster plot of the extract. (D) Correlation plot of the compounds that were tentatively identified in the extracts.
Molecules 30 02882 g002
Figure 3. Cytotoxicity of Lippia multiflora extracts against Vero cells at varying concentrations of 50 µg/mL, 100 µg/mL, 250 µg/mL, and 500 µg/mL. Mean differences of samples were assessed by a one-way analysis of variance (ANOVA) with Duncan’s Multiple Range Test (DMRT); the difference with p < 0.05 was considered statistically significant. Error bars indicate the standard deviation of triplicate values conducted as a single experiment. All experiments were carried out in triplicate (n = 3), and data represent the mean ± standard deviation of two independent experiments. Data for doxorubicin (positive control) at varying concentrations of 0.67 µg/mL, 1.35 µg/mL, 2.71 µg/mL, and 5.43 µg/mL are shown for comparison.
Figure 3. Cytotoxicity of Lippia multiflora extracts against Vero cells at varying concentrations of 50 µg/mL, 100 µg/mL, 250 µg/mL, and 500 µg/mL. Mean differences of samples were assessed by a one-way analysis of variance (ANOVA) with Duncan’s Multiple Range Test (DMRT); the difference with p < 0.05 was considered statistically significant. Error bars indicate the standard deviation of triplicate values conducted as a single experiment. All experiments were carried out in triplicate (n = 3), and data represent the mean ± standard deviation of two independent experiments. Data for doxorubicin (positive control) at varying concentrations of 0.67 µg/mL, 1.35 µg/mL, 2.71 µg/mL, and 5.43 µg/mL are shown for comparison.
Molecules 30 02882 g003
Table 1. UHPLC–Q/Orbitrap/MS data for compounds from various extracts of L. multiflora. Qualitative identification of compounds in L. multiflora extracts (R, roots; S, stem bark; DL, dichloromethane leaf; ML, methanol leaf; √, present; X, absent) by UHPLC–Q/Orbitrap/MS analysis.
Table 1. UHPLC–Q/Orbitrap/MS data for compounds from various extracts of L. multiflora. Qualitative identification of compounds in L. multiflora extracts (R, roots; S, stem bark; DL, dichloromethane leaf; ML, methanol leaf; √, present; X, absent) by UHPLC–Q/Orbitrap/MS analysis.
Compound
No.
Rt
(min)
Theoretical
Mass
[M − H]
(m/z)
Observed
Mass
[M − H]
(m/z)
Molecular
Formula
MS/MS Fragment
Ions
(m/z)
Compound
Name
Compound
Class
RSDLMLReferences
11.50483.31483.12C30H44O5 483Poricoic acid BTriterpenoid[42]
21.45455.35455.30C30H48O3 454, 453Boswellic acid alphaPentacyclic terpenoid[43]
310.06469.36469.11C31H50O3 469Eburicoic acidTriterpenoid XXX[44]
41.55501.32501.11C30H46O6485, 421Medicagenic acidTriterpenoidsXXX[45]
51.98525.16525.30C23H28O11525AlbiflorinMonoterpenoid glycosideXXX[46]
60.60289.07289.18C15H14O6259, 231CatechinFlavonoidXX[47]
70.60289.10289.18C15H14O6259, 243EpicatechinFlavonoidXX[48]
82.12267.02267.07C22H22O9 267Formononetin-7-O-glucoside (Ononin)Isoflavone glycoside[49]
92.88297.23297.24C17H14O53113′,4′-Dimethoxy-7-hydroxyflavoneFlavonoidXXXMassBank-BS-BS003750
101.97447.09447.09C21H20O11 455, 303QuercitrinFlavonoid XX[50]
111.46293.17293.18C17H26O4248, 237, 2096-GingerolPhenolic compoundXXX[48]
122.33277.18277.20C17H26O3283, 233, 2056-ParadolPhenolic ketone[48]
132.58295.17295.23C16H25NO4-EsmololPhenolic compoundXXX[48]
141.51305.06305.92C15H14O7289, 221(−)-EpigallocatechinPolyphenol[51]
150.65427.18427.18C23H28N2O6397, 381, 369, 243, 231IsomajdineIndole AlkaloidXXX[52]
160.23339.17339.12C20H24N2O3321, 297Yohimbic acidMonoterpene indole alkaloidsXXX[53]
171.46399.22339.20C22H28N2O5325, 309, 304Reserpic acidMonoterpene indole alkaloids[53]
181.52339.06339.07C15H16O93386,7-Dihydroxycoumarin-6-glucoside (Esculin)Coumarin glucosideXX[54]
191.83307.26307.19C20H36O2289, 290Eicosadieneoic acidPolyunsaturated fatty acidXX[55]
200.35229.14229.14C12H22O4213Dodecanedioic acidOrganic acid[56]
Table 2. Comparison of analytical equipment used for compound identification in this study (exp) with the existing literature (lit).
Table 2. Comparison of analytical equipment used for compound identification in this study (exp) with the existing literature (lit).
Compound
No.
Identified CompoundMolecular
Formula
Compound
Class
Plant Part(s)
(exp)
Plant Part(s)
(lit)
Method of Identification
(exp)
Method of Identification
(lit)
References
1Poricoic acid BC30H44O5 TriterpenoidRoot, stem bark, leavesStem bark, poriae cutisUHPLC–Q/Orbitrap/MSUPLC-MS XEVO G2-XS QTOF, UHPLC-QTOF-MS/MS, 1H-NMR, 13C-NMR[42,57]
2Boswellic acid alphaC30H48O3 Pentacyclic terpenoidRoot, stem bark, leavesResinsUHPLC–Q/Orbitrap/MSHPLC-DAD-ESI-MS/MS, HPLC[43]
3Eburicoic acidC31H50O3 Triterpenoid RootCactus, cladodes, callus, stem decayUHPLC–Q/Orbitrap/MSUPLC-QTOF-MS, FTIR, 1H-NMR, 13C-NMR[44,58]
4Medicagenic acidC30H46O6TriterpenoidsStem barkRoot, leavesUHPLC–Q/Orbitrap/MSLC ESI FT-ICR MS, FTIR, 1H-NMR, 13C-NMR[45,59]
5AlbiflorinC23H28O11Monoterpenoid glycosideStem barkWhole plant, roots, stems, leaves, flowersUHPLC–Q/Orbitrap/MSLC-Q-TOF-MS, LC-IT-MS, HPLC–DAD, HPLC–DAD–ESI–MS[46,60]
6CatechinC15H14O6FlavonoidStem bark, leavesWhole plantUHPLC–Q/Orbitrap/MSUHPLC-Q-Orbitrap HRMS, UPLC, LC-MS/MS[47,61]
7EpicatechinC15H14O6FlavonoidStem bark, leavesPulp, seed, leavesUHPLC–Q/Orbitrap/MSLC-ESI-MS/MS, [48]
8Formononetin-7-O-glucoside (Ononin)C22H22O9 Isoflavone glycosideRoot, stem bark, leavesLeaves, flowersUHPLC–Q/Orbitrap/MSLC-ESI-MS, HPLC/DAD,
RP-LC/PDA-ESI-QTOF-MS/MS
[49,62]
93′,4′-Dimethoxy-7-hydroxyflavoneC17H14O5FlavonoidLeavesN.R.UHPLC–Q/Orbitrap/MSLC-ESI-QTOFMassBank-BS-BS003750
10QuercitrinC21H20O11 Flavonoid LeavesRoot barks, trunk barks, stem, leaves, fruitsUHPLC–Q/Orbitrap/MSLC-MS/MS, UHPLC-ESI-QTOF-MS/MS[50,63]
116-GingerolC17H26O4Phenolic compoundLeavesPulp, seed, leavesUHPLC–Q/Orbitrap/MSLC-ESI-MS/MS, LC-MS/MS Q-TOF[48]
126-ParadolC17H26O3Phenolic ketoneRoot, stem bark, leavesPulp, seed, leavesUHPLC–Q/Orbitrap/MSLC-ESI-MS/MS, LC-MS/MS Q-TOF[48]
13EsmololC16H25NO4Phenolic compoundLeavesPulp, seed, leavesUHPLC–Q/Orbitrap/MSLC-ESI-MS/MS, LC-MS/MS Q-TOF[48]
14(−)-EpigallocatechinC15H14O7PolyphenolRoot, stem bark, leavesRhizome barkUHPLC–Q/Orbitrap/MSHPLC-MS, SEC-HPLC-UV, HPTLC[64]
15IsomajdineC23H28N2O6Indole AlkaloidLeavesRootUHPLC–Q/Orbitrap/MSNMR[65]
16Yohimbic acid C20H24N2O3Monoterpene indole alkaloidsLeavesRootUHPLC–Q/Orbitrap/MSHPLC–ESI–QTOF–MS/MS, UHPLC–UV–MS/MS[53,66]
17Reserpic acidC22H28N2O5Monoterpene indole alkaloidsRoot, stem bark, leavesRootUHPLC–Q/Orbitrap/MSHPLC–ESI–QTOF–MS/MS[53]
186,7-Dihydroxycoumarin-6-glucoside (Esculin)C15H16O9Coumarin glucosideLeavesRoot, young stem, leavesUHPLC–Q/Orbitrap/MSHR-LCMS[54]
19Eicosadieneoic acidC20H36O2Polyunsaturated fatty acid Root, stem bark SeedsUHPLC–Q/Orbitrap/MSLC-Q-TOF-MS[55]
20Dodecanedioic acidC12H22O4Organic acidRoot, stem bark, leavesRhizomesUHPLC–Q/Orbitrap/MSUHPLC–Q-exactive orbitrap MS[56]
Table 3. The PCA loading scores.
Table 3. The PCA loading scores.
CompoundsPC1PC2PC3
C1000
C2000
C3−0.279210−0.73822−0.61406
C4−0.679120.103980.72662
C5−0.679120.103980.72662
C6−0.527580.829610.18275
C7−0.527580.0829610.18275
C8000
C90.88841−0.219730.40304
C100.829940.54927−0.097482
C110.88841−0.219730.40304
C12000
C130.88841−0.219730.40304
C14000
C150.0699230.85397−0.5156
C160.88841−0.0219730.40304
C17000
C180.829940.54927−0.097482
C19−0.82994−0.549270.097482
C20000
Table 4. Antioxidant activity of Lippia multiflora extracts.
Table 4. Antioxidant activity of Lippia multiflora extracts.
SamplesDPPH IC50 (µg/mL)Reducing Power IC0.5 (µg/mL)
Root extract25.108 ± 19.798 ab0.218 ± 0.157 a
Stem bark extract5.031 ± 2.940 a0.029 ± 0.026 a
Methanol leaf extract0.559 ± 0.269 a0.710 ± 1.220 a
Dichloromethane leaf extract6.299 ± 0.495 a0.326 ± 0.210 a
Ascorbic acid44.308 ± 9.813 b0.355 ± 0.393 a
Gallic acid37.340 ± 17.529 ab8.210 ± 11.257 a
Quercetin37.361 ± 21.356 b3.760 ± 2.979 a
Notes: A different superscript letter indicates significant differences using a one-way ANOVA at p < 0.05. Data (n = 3) expressed as mean ± standard deviation. For DPPH (2,2-diphenyl-1-picrylhydrazyl) free radical scavenging activity: a—stem bark crude extract was not significantly different from the methanol leaf extract and dichloromethane leaf extract; b—ascorbic acid was not significantly different from quercetin; ab—root extract was not significantly different from all crude extracts, ascorbic acid, gallic acid, and quercetin. For reducing power activity: a—root extract was not significantly different from all crude extracts, ascorbic acid, gallic acid, and quercetin.
Table 5. Comparison of the methods used to determine the antioxidant activity of extracts obtained in this study (exp) with the existing literature (lit).
Table 5. Comparison of the methods used to determine the antioxidant activity of extracts obtained in this study (exp) with the existing literature (lit).
Compound
No.
Identified CompoundMolecular
Formula
Compound
Class
Plant Part(s)
(exp)
Plant Part(s)
(lit)
Antioxidant Assay
(exp)
Antioxidant Assay
(lit)
References
1Poricoic acid BC30H44O5 TriterpenoidRoot, stem bark, leavesStem barkDPPH, RPADPPH[42]
2Boswellic acid alphaC30H48O3 Pentacyclic terpenoidRoot, stem bark, leavesResinsDPPH, RPADPPH, ABTS[74]
3Eburicoic acidC31H50O3 Triterpenoid Root-DPPH, RPA--
4Medicagenic acid C30H46O6TriterpenoidsStem bark-DPPH, RPA--
5AlbiflorinC23H28O11Monoterpenoid glycosideStem barkRoot, stem, leaves, flowersDPPH, RPADPPH, ABTS[60]
6CatechinC15H14O6FlavonoidStem bark, leavesN.R.DPPH, RPAABTS, FRAP[68]
7EpicatechinC15H14O6FlavonoidStem bark, leavesN.R.DPPH, RPAABTS, FRAP[68]
8Formononetin-7-O-glucoside (Ononin)C22H22O9 Isoflavone glycosideRoot, stem bark, leavesLeaves, flowersDPPH, RPADPPH, ABTS[62]
93′,4′-Dimethoxy-7-hydroxyflavoneC17H14O5FlavonoidLeaves-DPPH, RPA--
10QuercitrinC21H20O11 Flavonoid LeavesTrunk barksDPPH, RPAABTS, ORAC, TEAC[50,69,70]
116-GingerolC17H26O4Phenolic compoundLeavesRhizomesDPPH, RPADPPH[67]
126-ParadolC17H26O3Phenolic ketoneRoot, stem bark, leaves-DPPH, RPA--
13EsmololC16H25NO4Phenolic compoundLeaves-DPPH, RPA--
14(−)-EpigallocatechinC15H14O7PolyphenolRoot, stem bark, leavesRhizome barkDPPH, RPADPPH[64]
15IsomajdineC23H28N2O6Indole AlkaloidLeavesRootDPPH, RPAABTS, DMPD, Fe2+[65]
16Yohimbic acidC20H24N2O3Monoterpene indole alkaloidsLeaves-DPPH, RPA--
17Reserpic acidC22H28N2O5Monoterpene indole alkaloidsRoot, stem bark, leaves-DPPH, RPA--
186,7-Dihydroxycoumarin-6-glucoside (Esculin)C15H16O9Coumarin glucosideLeaves-DPPH, RPA--
19Eicosadieneoic acidC20H36O2Polyunsaturated fatty acid Root, stem bark, -DPPH, RPA--
20Dodecanedioic acidC12H22O4Organic acidRoot, stem bark, leaves-DPPH, RPA--
Notes: N.R.—not reported; DPPH—DPPH (2,2-diphenyl-1-picrylhydrazyl) free radical scavenging assay; RPA—reducing power assay; ABTS—2,2′-azino-bis(3-ethylbenzothiazoline-6 sulfonic acid) assay; FRAP—ferric reducing antioxidant power; DMPD—N,N dimethyl-p-phenylenediamine radical scavenging; Fe2+—ferrous ions chelating activity; ORAC—oxygen radical absorbance capacity; and TEAC—Trolox equivalent antioxidant capacity.
Table 6. Pearson’s correlation of phytochemicals (TFC and TPC) and their antioxidant activities.
Table 6. Pearson’s correlation of phytochemicals (TFC and TPC) and their antioxidant activities.
TestTFCTPCDPPHRP
TFC1
TPC−0.111 *1
DPPH0.619 *0.071 *1
RP−0.956−0.019−0.4061
Notes: Correlation coefficient (r) between the phytochemicals (TFC and TPC) with antioxidant capacity (DPPH and RP). If the r value is close to +1 or −1, it indicates a strong positive relationship. If r is near 0, it indicates a weak or no relationship. * Correlation is significant at p < 0.05. Abbreviations: TFC—total flavonoid content; TPC—total phenolic content; DPPH—DPPH (2,2-diphenyl-1-picrylhydrazyl) free radical scavenging assay; and RP—reducing power capacity.
Table 7. Inhibitory concentrations (IC50 in µg/mL) of extracts of the root, stem bark, and leaf of Lippia multiflora and doxorubicin (positive control) in Vero monkey kidney cell lines.
Table 7. Inhibitory concentrations (IC50 in µg/mL) of extracts of the root, stem bark, and leaf of Lippia multiflora and doxorubicin (positive control) in Vero monkey kidney cell lines.
SamplesVero IC50 (µg/mL)
Root extract37.15 ± 2.61 b
Stem bark extract273.60 ± 1.70 e
Methanol leaf extract74.22 ± 2.33 c
Dichloromethane leaf extract132.90 ± 4.52 d
Doxorubicin (positive control)4.97 ± 0.83 a
Notes: The values above are presented by mean ± standard deviation of three replicates (n = 3). Values with different superscript letters in each column are significantly different using one-way ANOVA at p < 0.05.
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

Tlhapi, D.; Malebo, N.; Manduna, I.T.; Mawunu, M.; Chokwe, R.C. Phytochemical Screening and Biological Activities of Lippia multiflora Moldenke. Molecules 2025, 30, 2882. https://doi.org/10.3390/molecules30132882

AMA Style

Tlhapi D, Malebo N, Manduna IT, Mawunu M, Chokwe RC. Phytochemical Screening and Biological Activities of Lippia multiflora Moldenke. Molecules. 2025; 30(13):2882. https://doi.org/10.3390/molecules30132882

Chicago/Turabian Style

Tlhapi, Dorcas, Ntsoaki Malebo, Idah Tichaidza Manduna, Monizi Mawunu, and Ramakwala Christinah Chokwe. 2025. "Phytochemical Screening and Biological Activities of Lippia multiflora Moldenke" Molecules 30, no. 13: 2882. https://doi.org/10.3390/molecules30132882

APA Style

Tlhapi, D., Malebo, N., Manduna, I. T., Mawunu, M., & Chokwe, R. C. (2025). Phytochemical Screening and Biological Activities of Lippia multiflora Moldenke. Molecules, 30(13), 2882. https://doi.org/10.3390/molecules30132882

Article Metrics

Back to TopTop