Next Article in Journal
Is It Reliable to Take the Molecular Docking Top Scoring Position as the Best Solution without Considering Available Structural Data?
Next Article in Special Issue
Three New Highly Oxygenated Germacranolides from Carpesium Divaricatum and Their Cytotoxic Activity
Previous Article in Journal
Discovery of Pyrazolo[4,3-c]quinolines Derivatives as Potential Anti-Inflammatory Agents through Inhibiting of NO Production
Previous Article in Special Issue
Anticancer Profiling for Coumarins and Related O-Naphthoquinones from Mansonia gagei against Solid Tumor Cells In Vitro
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Phenolic Composition and Bioactivity of Lavandula pedunculata (Mill.) Cav. Samples from Different Geographical Origin

1
Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal
2
Institute for Biological Research “Siniša Stanković”, Department of Plant Physiology, University of Belgrade, Bulevar Despota Stefana 142, 11000 Belgrade, Serbia
3
Banco Português de Germoplasma Vegetal, Instituto Nacional de Investigação Agrária e Veterinária, I.P. (INIAV, I.P.), Quinta S. José, S. Pedro de Merelim, 4700-859 Braga, Portugal
*
Author to whom correspondence should be addressed.
Molecules 2018, 23(5), 1037; https://doi.org/10.3390/molecules23051037
Submission received: 9 April 2018 / Revised: 24 April 2018 / Accepted: 27 April 2018 / Published: 28 April 2018
(This article belongs to the Collection Bioactive Compounds)

Abstract

:
The aim of this study was to characterize the phenolic composition and evaluate the bioactivity of several samples of Lavandula pedunculata (Mill.) Cav, and to compare aqueous and hydroethanolic extracts. Plant materials were obtained by growing some accessions (seed samples) of various wild populations from different regions of Portugal conserved at the Portuguese Genebank in Braga. Phenolic compounds were analised by HPLC-DAD-ESI/MSn, antioxidant potential through in vitro assays (DPPH radical scavenging activity, reducing power and inhibition of lipid peroxidation), cytotoxicity on tumor cells (MCF-7, NCI-H460, HeLa and HepG2) and non-tumor (PLP2) cells, anti-inflammatory activity in rat RAW 264.7 macrophages, by the ability to inhibit NO production and antimicrobial potential by the microdilution method with INT dye (iodonitrotetrazolium chloride). Thirteen compounds were identified, being salvianolic acid B, rosmarinic acid and luteolin-7-O-glucuronide, the main compounds present, with values ranging between 44.3–582, 50.9–550, and 24.36–101.5 mg/g extract, respectively. L. pedunculata aqueous extract revealed a higher antioxidant potential (EC50 values between 14 to 530 μg/mL), which could be related to its higher concentration in phenolic compounds; however, the hydroethanolic extract showed a higher anti-inflammatory (lower EC50 values than 124 μg/mL) potential and antiproliferative capacity (lower GI50 values than 34 μg/mL). Thus, this study highlights the bioactive effects of this species and opens up possibilities of uses in food and pharmaceutical formulations. However, there are potential differences in such properties according to geographical origin of plant material, as in general, the samples from Alentejo presented higher results in all the bioactivities, compared with Trás-os-Montes samples.

Graphical Abstract

1. Introduction

Bioactive compounds are molecules that have health benefits on living organisms, tissues or cells, being comprised of a wide range of classes, such as vitamins, carotenoids (lycopene, β-carotene and xanthophylls) and phenolic compounds [1]. These compounds are present in a wide range of foods (e.g., fruits, vegetables, plants and other food products) and its ingestion enables the induction of positive effects in the prevention and treatment of many human diseases, such as anxiety, insomnia, anorexia, cough and bronchitis [2].
Throughout the years, phenolic compounds have been largely studied in several foods, namely edible plants, due to their therapeutic capabilities in pathologies related to the nervous and gastrointestinal systems [3]; they also exhibit antioxidant, antitumor, anti-inflammatory and antiviral properties [1]. These molecules are characterized by being a very heterogeneous group of a great variety of compounds, such as phenolic acids, coumarins, flavonoids, stilbenes, hydrolysable and condensed tannins, lignans and lignins [3,4]. Flavonoids are one of the most abundant groups within phenolic compounds; their main mechanism of action goes through their ability of capturing reactive oxygen species and chelate metal ions, having a distinct antioxidant action [4]. Thus, the ingestion of antioxidants can significantly control the severity of chronic diseases, providing a close relationship between the uptake of free radicals and the involvement of endocrine responses. Therefore, the inclusion of antioxidants in the diet for the prevention of chronic diseases and for an improvement of general health, has been an important research target in recent decades [5].
Lavandula pedunculata (Mill.) Cav. (common English name French lavender and known in Portugal as rosmaninho, arçã, rosmaninho maior or lavanda), is a species belonging to the Lamiaceae which is native to the Iberian Peninsula, North Africa and Turkey [6,7,8]. In the Iberian Peninsula, this species has a widespread distribution and significant ethnobotanical uses documented [6,9]. It is considered the most resistant of all species of the Lavandula genus, well adapted to continental climates, especially to annual variation in temperature (hot summers and cold winters), growing in altitudes up to 1700 m [6,9]. In the context of traditional medicine, L. pedunculata has been used in infusions for internal and external applications, mainly recommended for the respiratory and digestive systems and as a therapeutic agent with antiseptic action for cleaning wounds [6,10].
Wild plant genetic resources, particularly of medicinal and aromatic species, have received increased interested worldwide. Besides L. pedunculata traits and potential use in different domains (e.g., as source of bioactive compounds), this endemic species had poor representation in germplasm collections and has not been comprehensively characterized [8]. Therefore, the Portuguese Genebank (BPGV) carried out in 2009, several collecting missions of Lavandula germoplasm in different regions of the national territory, including L. pedunculata [8]. The main purpose was to collect plant diversity, promoting ex situ conservation and providing morphological, molecular, chemical, and biochemical information (germplasm characterization); the seeds of several wild populations (genebank accessions) were stored at the BPGV facilities, in Braga, Portugal [8].
As bioactive molecules are often produced in response to changes in the surrounding environment, the same plant species growing at different sites may have different concentrations of the most promising compounds, or even different compounds in their constitution [11]. Germplasm accessions conserved at genebanks might generate very good matrices to evaluate useful characteristics and bioactive properties, because they correspond to stored plant materials of the same species but of different provenance. Moreover, genebanks procedures, such as germplasm characterization, provide the description of plant germplasm and determine the expression of highly heritable characters ranging from morphological, agronomical, chemical features to bioactive properties or molecular markers.
The aim of the present study was to evaluate the phenolic profile of the hydroethanolic and aqueous extracts of thirteen Lavandula pedunculata (Mill.) Cav. samples, whose seeds originated from different regions of Portugal, and are conserved ex-situ in the Portuguese Genebank. The bioactive properties were explored in terms of antioxidant, anti-inflammatory, cytotoxicity and antimicrobial potential. Furthermore, the study also addressed L. pedunculata germoplasm characterization (e.g., chemical and biochemical characteristics), contributing with relevant data about the studied accessions, enabling eventual discrimination among them, resulting in better insight about the composition of the germplasm collection and its genetic diversity.

2. Results

2.1. Phenolic Compounds

Data related to the phenolic compounds identification (retention time, λmax in the visible region, molecular ion, main fragment ions in MS2, and tentative identification) obtained by HPLC-DAD-ESI/MSn analysis of Lavandula pedunculata (Mill.) Cav. samples are presented in Table 1. All samples presented a similar profile (Figure 1), revealing the presence of thirteen phenolic compounds, in both hydroethanolic and aqueous extracts, being nine identified as phenolic acids (mainly caffeic acid dimers, trimers and tetramers) and four flavonoids (mainly luteolin and eriodictyol glycoside derivatives).
Caffeic acid (compound 4), luteolin-7-O-glucuronide (compound 8) and rosmarinic acid (compound 10) were positively identified by comparison with commercial standards. Rosmarinic acid has been one of the main compounds previously identified in Lavandula species [3,12,13,14,15]. Compounds 1 and 2, 3 and 7 were identified as caffeic, p-coumaric and rosmarinic acid hexosides, based on the respective fragment ions released at m/z 179 [caffeic acid-H], 163 [coumaric acid-H] and 359 [rosmarinic acid-H] after loss of a hexosyl moiety (−162 mu). With the exception of rosmarinic acid hexoside, the mentioned compounds have been found in Lavandula x intermedia Emeric ex Loiseleur) Waste [12].
Compound 9 ([M − H] at m/z 719) released a main MS2 fragment at m/z 359 ([M − 2H]2−, rosmarinic acid), which allowed its identification as sagerinic acid [16]. Compound 11 ([M − H] at m/z 537) presented a similar UV spectrum and fragmentation pattern, consistent with the caffeic acid trimer lithospermic acid A [16,17,18]. Other identities, with the same molecular weight (salvianolic acids H/I), were discarded because they present quite a different fragmentation pattern [18,19]. Thus, being assigned to lithospermic acid A. Compound 13 ([M − H] at m/z 717) presented a fragmentation pattern with successive losses of 198 mu (danshensu) or 180 mu (caffeic acid) units, coherent with salvianolic acid B (also known as lithospermic acid B) [18,19]. These compounds have not been previously identified in Lavandula species, to the best of the authors’ knowledge.
The remaining compounds correspond to glycosylated flavones. Compound 5 ([M − H] at m/z 623) MS2 fragments revealed the alternative loss of hexosyl (m/z at 461; −162 u) and glucuronyl (m/z at 285; −176 u) residues, indicating location of each residue on different positions of the aglycone. No information about the identity of the sugar moieties and location on the aglycone could be obtained, so, the compound was identified as luteolin-O-hexosyl-O-glucuronide [20]. Compound 6 ([M − H] at m/z 463) was identified as a eriodictyol derivative, due to its UV-Vis spectra and MS fragmentation. This peak revealed the loss of glucuronyl (−176 u), therefore being assigned as eriodictyol-O-glucuronide [21]. Compound 12 ([M − H] at m/z 623) presented 42 u higher then compound 8, thus being assigned to methylluteolin-O-glucuronide [21]. Also, to the best of our knowledge, these compounds have not been previously identified in Lavandula species.
In a previous study performed by Costa et al. [3], using polar extracts from Lavandula pedunculata subsp. lusitanica, six different phenolic compounds were detected (3-O-caffeoylquinic acid, 4-O-caffeoylquinic acid, 5-O-caffeoylquinic acid, rosmarinic acid, luteolin and apigenin), nonetheless, only rosmarinic acid was the common phenolic acid identified, but revealed smaller amounts. However, also in this case, this compound proved to be the majority, in the hydroethanolic and ethanolic extracts. The differences observed between the different samples could be explained, due to the influence that the geographical area has on the chemical composition of plants, namely at the level of soil composition, climacteric environment, air humidity and daily sun exposure [11]. Moreover, to the best of the authors’ knowledge, and with the exception of the previous mentioned authors, there are no more reports describing the phenolic composition of L. pedunculata (Mill.) Cav.
Phenolic acids represent a significant part of this phenolic profile (Table 2), highlighting salvianolic acid B (compound 13) and rosmarinic acid (compound 10) as the major phenolic compounds present in both types of extracts. Regarding flavonoids, luteolin-7-O-glucuronide (compound 8) was the main compound present (Table 2). In general, the aqueous extract revealed the highest content in phenolic compounds, revealing sample 8 (Ponte de Sôr, Portalegre) the highest concentration in total phenolic acids, flavonoids and total phenolic compounds for both the studied extracts. This difference could be explained to the chemical composition that each of the seeds initially presented and due to their different geographic origin.

2.2. Antioxidant Potential

The antioxidant activity of L. pedunculata hydroethanolic and aqueous extracts was evaluated using different in vitro assays (DPPH radical-scavenging activity, reducing power, inhibition of β-carotene bleaching and inhibition of lipid peroxidation—TBARS) and the results are present in Table 3. All samples, for both of the studied extracts, revealed antioxidant potential, EC50 values between 530–14 μg/mL and 1833–17 μg/mL for aqueous and hydroethanolic extract, respectively. In general, there is not a large divergence regarding EC50 values between the hydroethanolic and aqueous extracts. Accordingly, for aqueous extracts, sample 4 (Bragança) revealed the highest DPPH scavenging activity (EC50 = 68.0 μg/mL), while sample 2 (Évora) revealed the highest reducing power (EC50 = 51 μg/mL). For the β-carotene bleaching inhibition assay, samples 2 (Évora) and 10 (Castelo de Vide, Portalegre), with values of 253 and 236 μg/mL, respectively, revealed the highest potential. On the other hand, sample 1 (Marvão, Portalegre) revealed the highest antioxidant potential for the TBARS inhibition assay (14 μg/mL).
Otherwise, hydroethanolic extracts of samples 10 (Castelo de Vide, Portalegre), 11 (Elvas, Portalegre), 12 (Castelo de Vide, Portalegre), and 13 (Bragança) showed the lowest EC50 values for the β-carotene bleaching inhibition assay, presenting values of 252, 190, 223 and 214 μg/mL, respectively. Additionally, in the hydroethanolic extracts, samples 8 (Ponte de Sôr, Portalegre), 9 (Évora), and 2 (Évora) showed the highest antioxidant capacity for the DPPH scavenging activity, reducing power and TBARS inhibition assays, respectively. The highest EC50 values (lowest antioxidant activity) were detected in sample 13 (Bragança) for DPPH scavenging activity (257 μg/mL) and reducing power (216 μg/mL), in sample 8 for β-carotene bleaching inhibition (1833 μg/mL), and in samples 6 (Portalegre) and 11 (Elvas, Portalegre) in TBARS inhibition (63.5 and 62 μg/mL, respectively).
These results are in agreement with those found by Costa et al. [3], where the antioxidant potential L. pedunculata was evaluated by TBARS assay and at the highest concentration tested (5 mg/mL), the infusion, water and water:ethanol extracts completely prevented MDA production.
In a study performed by Pereira et al. [15], the antioxidant activity of different extracts (n-hexane, dichloromethane, ethyl acetate, methanol and water) of L. pedunculata was evaluated regarding the lipid peroxidation index (%), obtaining values that ranged from 26 to 41%. Moreover, Ferreira et al. [23] tested the ethanolic extracts and decoctions of L. pedunculata using the DPPH and β-carotene inhibition methods, in which these samples revealed antioxidant activity, presenting values between 93 and 20 mg/mL, respectively. Baptista et al. [24], also studied the antioxidant potential for the essential oils and extracts of two native Portuguese Lavandula species (L. stoechas subsp. luisieri and L. pedunculata) and considering the results obtained, there is some similarity in the results of the methanolic and aqueous extract, in comparison with the concentrations obtained in our study.

2.3. Anti-Inflammatory Activities

The in vitro anti-inflammatory results are present in Table 3. In general, hydroethanolic extracts revealed more promising anti-inflammatory potential (EC50 values ranging from 216 to 124 μg/mL), showing lower EC50 values than the aqueous extracts (EC50 values ranging between 140 to 301 μg/mL). Moreover, there were several samples that did not show anti-inflammatory activity (EC50 > 400 μg/mL), being the absence of this bioactivity higher for the aqueous samples. The samples that presented this activity were samples 4 (Bragança), 5 (Arronches. Portalegre), 7 (Nisa, Portalegre), 8 (Ponte de Sôr, Portalegre), 9 (Évora), and 13 (Bragança) in hydroethanolic extract, and samples 1 (Marvão, Portalegre), 2 (Évora), 3 (Vila Viçosa, Évora), 6 (Portalegre), 9 (Évora), 10 (Castelo de Vide, Portalegre), 11 (Elvas, Portalegre), 12 (Castelo de Vide, Portalegre), and 13 (Bragança) for the aqueous extracts.
Sample 6 hydroethanolic extract (Portalegre) presented the highest anti-inflammatory potential (124 μg/mL), while for the aqueous extract, sample 8 (Ponte de Sôr, Portalegre) revealed the highest efficiency, presenting a concentration of 140 μg/mL. Similarly, Algieri et al. [25] studied the anti-inflammatory activity of L. stoechas hydromethanolic extract, which also exhibited a significantly inhibition of carrageenan-induced paw edema in mice. Therefore, the hydroethanolic extracts could also be considered as a good source of anti-inflammatory molecules.

2.4. Cytotoxicity

Table 4 summarizes the effects of L. pedunculata hydroethanolic and aqueous extracts (infusions) on the inhibition of the growth of four human tumor cell lines (MCF-7, NCI-H460, HeLa and HepG2). In this study, it was evident the cytotoxic activity of L. pedunculata in all the cell lines tested, ranging values between 374–82 μg/mL and 340–34 μg/mL, for aqueous and hydroethanolic extracts, respectively. Comparing the two types of studied extracts, the aqueous extracts showed anti-proliferative potential in more samples and in more cell lines, however it was the hydroethanolic extract that, overall, showed a higher cytotoxic potential, i.e., lower GI50 values (34 μg/mL). Sample 6 (Portalegre) hydroethanolic extract presented the best cytotoxic potential in MCF-7 (values ranged from 53 to 236 μg/mL) and HepG2 (values between from 34 and 212 μg/mL) cell lines. On the other hand, sample 12 (Castelo de Vide, Portalegre) showed a higher antiproliferative capacity in NCI-H460 cell line (GI50 values = 119 μg/mL); and sample 10 (Castelo de Vide, Portalegre) showed a lower GI50 value in the HeLa cell line (62 μg/mL). In hydroethanolic extract, sample 6, 12 and 13 (Bragança) revealed lower antiproliferative capacity in NCI-H460 (340 μg/mL), MCF-7 (236 μg/mL) and HeLa (222 μg/mL) cell lines, respectively; in HepG2 cell line were samples 7 (Nisa, Portalegre) and 13 that evidenced a lower, with GI50 values of 212 and 204 μg/mL, respectively.
On the other hand, the aqueous extract of sample 9 (Évora) revealed a better cytotoxic potential in MCF-7 and HepG2 cell lines, with values of 150 and 118 μg/mL, respectively. In the NCI-H460 cell line, the values ranged from 374 and 113 μg/mL, highlighting sample 4 (Bragança) with the highest antiproliferative activity. Meanwhile, for HeLa cell line, sample 12 (Castelo de Vide, Portalegre) showed the lowest GI50 value (82 μg/mL).
The cytotoxicity in non-tumor cells were tested using a primary culture cell line obtained from porcine liver (PLP2 cell culture) and was used as a preliminary toxicity model for normal cells. None of the samples showed toxicity (GI50 > 400 μg/mL), with the exception of hydroethanolic extract of sample 12 (Castelo de Vide, Portalegre), and aqueous extracts of sample 7 (Nisa, Portalegre), 8 (Ponte de Sôr, Portalegre) and 9 (Évora). However, the presented concentrations are much higher than those obtained for the antiproliferative inhibition using the studied tumor cell lines.
Despite being the same plant species, it is normal the existence of different results. The geographical origin of the seeds is one of the factors that can directly influence the chemical and nutritional composition of plants due to biotic and abiotic factors [11], thus, in this case, as the seeds were harvested from different areas, this factor could lead to the observed oscillations in the bioactive potential.
There are few cytotoxicity studies carried out using this species, however the results described in literature revealed lower cytotoxic potential, when compared to our results. According to Pereira et al. [15], the results obtained with L. pedunculata methanolic extracts suggest that for concentrations up to 15 µg/mL and an incubation period of 24 h, no relevant cytotoxic effects were observed. Tang et al. [26], tested five human tumor cell lines (NB4, A549, SHSY5Y, PC3, and MCF7) using the methanolic extract of L. angustifolia, showed a weak inhibitory activity against the tested human tumor cell lines (IC50 values ranging from 2.2–8.2 mM).

2.5. Antimicrobial Activity

The results on antibacterial and antifungal activities of L. pedunculata hydroethanolic and aqueous extracts are presented in Table 5. The samples were tested against a panel of eight bacteria and fungi strains, specifically selected on the basis of their importance to public health.
For antibacterial activity of the hydroethanolic extracts, samples 1 (Marvão, Portalegre) and 5 (Arronches, Portalegre) revealed a higher activity (lower MICs) for all the studied strains. In these cases, MIC values range from 20 µg/mL in sample 1 for the Staphylococcus aureus strain and 40 µg/mL in sample 5 for the Enterobacter cloacae strain. The lowest MBC values were observed for samples 1 and 3 (Vila Viçosa, Évora) in the inhibition of Staphylococcus aureus strain (40 µg/mL). In the evaluation of L. pedunculata aqueous extracts, sample 5 showed the lowest MIC and MBC, being more potent for Bacillus cereus strain (MIC = 75 µg/mL and MBC = 150 µg/mL), along with the MBC of sample 8 (Ponte de Sôr, Portalegre) in Escherichia coli strain (also 150 µg/mL). These results are in agreement with previous studies, where other plants species also showed antimicrobial activity: for example, Castanea sativa Mill. flowers [27], Alnus rugosa L. aerial parts [28], and Veronica urticifolia Jacq. [29]. Nikolic et al. [30] studied the essential oils of different Thymus species, such as T. serpyllum, T. algeriensis and T. vulgaris, which showed antimicrobial activity. However, to the best of the author’s’ knowledge there are no studies reporting the antibacterial activity of the studied Lavandula species.
Considering the antifungal activity, for hydroethanolic extracts, all samples revealed inhibitory and fungicidal activity. Sample 3 showed the best MIC values for all the tested strains, with the exception of Aspergillus versicolor and Aspergillus ochraceus, where samples 2 and 9 revealed the highest potential, respectively. Regarding the MFC, in general, most of the samples revealed promising results, being better for sample 13 aqueous extract. In the case of aqueous extracts, only a few samples showed activity in all the studied strains, meanwhile samples 2, 11, 12 and 13 showed inhibitory and fungicidal potential for all the studied fungi strains. Samples 9 and 11 presented the highest inhibitory capacity for all the fungal strains analysed. The best fungicide capacity was evident in sample 11 against Aspergillus versicolor strain.
The antifungal potential is, also, in agreement with other studies regarding antifungal activity of other plant species, which were carried out by other authors, such as Castanea sativa [27] and Alnus rugosa [28]. Once more, to the best of the authors’ knowledge, there are no previous studies on the antifungal activity of L. pedunculata.

3. Materials and Methods

3.1. Samples and Samples Preparation

The studied samples concern plant materials (the top 20 cm of the flowering stems with inflorescences) randomly harvested in each of thirteen field plots of Lavandula pedunculata (Mill.) Cav. (botanical family Lamiaceae) grown at the Portuguese Genebank (Banco Português de Germoplasma Vegetal, BPGV).
The Portuguese Genebank conserves ex situ, using cold temperatures (e.g., 5–6 °C), a collection of medicinal and aromatic plants, including species of the genus Lavandula. The col, lection consists of seed samples (accessions) of wild specimens that have been randomly collected in different natural populations within several Portuguese regions [8].
For this study, the stored seeds of thirteen accessions of L. pedunculata, of the BPGV collection were sown and the plantlets transplanted outdoor to thirteen field plots, in BPGV farm at S. Pedro de Merelim, Braga, Northern Portugal (GPS coordinates 41°34’28.01’’ N; 8°27’09.21’’ S).
In 2015, the flowering parts in blossom of each accession grown were harvested, kept in paper bags at −20 °C, corresponding individually to a sample. Each sample of plant material (approximately 200 g) was subsequently lyophilized (FreeZone 4.5, Labconco, Kansas City, MO, USA), reduced to a fine dried powder (~20 mesh) and mixed to obtain a homogenate sample, corresponding to a total of thirteen different samples.
The thirteen studied accessions of L. pedunculata were gathered wild in two Portuguese regions, Alentejo and Trás-os-Montes (Table 6), altitude range between 100 and 1000 m [8]. This species has several synonymies such as Lavandula eliasii Sennen, Lavandula pedunculata subsp. pedunculata, Lavandula stoechas subsp. pedunculata (Mill.) Samp. ex Rozeira, Lavandula stoechas subsp. lusitanica (Chaytor) Rozeira, and Stoechas pedunculata Mill. [9].
The hydroethanolic extracts were obtained from the lyophilized plant material. The dried sample was extracted following a procedure previously described by Pereira et al. [31]. A solution of ethanol/water (80:20 v/v) (25 mL) was added to 1 g of sample and stirring for 1 h (25 °C at 150 rpm) and subsequently filtered through Whatman No. 4 paper. The residue was then re-extracted with an additional portion of 25 mL of the same solution (25 °C at 150 rpm) during 1 h. The combined extracts were evaporated to remove the ethanolic fraction (at 40 °C) and then the water was frozen and lyophilized.
The aqueous extracts (infusions) preparation was established according a study performed by Pereira et al. [31]. Powdered samples (2 g) were added to 200 mL of boiling distilled water, left to stand at room temperature for 5 min, and then filtered under reduced pressure, frozen and lyophilized.

3.2. Standards and Reagents

Acetonitrile 99.9% was of HPLC grade from Fisher Scientific (Lisbon, Portugal). Phenolic compound standards (caffeic acid ≥ 99%, p-coumaric acid ≥ 90%; hesperetin ≥ 99%, luteolin-7-O-glucoside ≥ 99%, quercetin-3-O-glucoside ≥ 99%, rosmarinic acid ≥ 99% HPLC purity) were from Extrasynthese (Genay, France). Trypan blue, lipopolysaccharide (LPS), dexamethasone, acetic acid, formic acid, ellipticine, sulforhodamine B (SRB), trichloroacetic acid (TCA) and Tris were all purchased from Sigma-Aldrich (St. Louis, MO, USA). Dulbecco’s modified Eagle’s medium was purchased from HyClone. RAW264.7 cells were acquired from ECACC (‘‘European Collection of Animal Cell Culture”) (Salisburg, UK), Griess reagent system kit was purchased from Promega, Fetal bovine serum (FBS), L-glutamine, Hank’s balanced salt solution (HBSS), trypsin-EDTA (ethylenediaminetetraacetic acid), penicillin/streptomycin solution (100 U/mL and 100 mg/mL, respectively), RPMI-1640 and DMEM media were from Hyclone (Logan, UT, USA). Water was treated in Milli-Q water purification system (TGI Pure Water Systems, Greenville, SC, USA).

3.3. Analysis of Phenolic Compounds

The lyophilized extracts were re-dissolved at 5 mg/mL in ethanol/water (20:80, v/v) and 100% water for hydroethanolic and aqueous extract, respectively, and filtered through a 0.22-µm disposable LC filter disk. Chromatographic analyses were performed in a Dionex Ultimate 3000 UPLC (Thermo Scientific, San Jose, CA, USA) system equipped with a diode array detector (DAD) coupled to an electrospray ionization mass detector (LC-DAD-ESI/MSn) as previously described by Bessada et al. [32].
Chromatographic separation was achieved with a Waters Spherisorb S3 ODS-2 C18 (3 μm, 4.6 mm × 150 mm, Waters, Milford, MA, USA) column thermostatted at 35 °C. The solvents used were: (A) 0.1% formic acid in water; (B) acetonitrile (HPLC purity). The elution gradient established was isocratic 15% B (5 min), 15% B to 20% B (5 min), 20-25% B (10 min), 25–35% B (10 min), 35–50% B (10 min), and re-equilibration of the column, using a flow rate of 0.5 mL/min. Double online detection was carried out in the DAD using 280, 330, 370 and 520 nm as preferred wavelengths and in a mass spectrometer (MS) connected to HPLC system via the DAD cell outlet.
MS detection was performed in negative mode, using a Linear Ion Trap LTQ XL mass spectrometer (Thermo Finnigan, San Jose, CA, USA) equipped with an ESI source. Nitrogen served as the sheath gas (50 psi); the system was operated with a spray voltage of 5 kV, a source temperature of 325 °C, a capillary voltage of −20 V. The tube lens offset was kept at a voltage of −66 V. The full scan covered the mass range from m/z 100 to 1500. The collision energy used was 35 (arbitrary units). Data acquisition was carried out with Xcalibur® data system (Thermo Finnigan, San Jose, CA, USA).
Compounds were carefully identified comparing the obtained information: retention times, UV-vis and mass spectra, with those obtained from standard compounds, when available. Otherwise, compounds were tentatively identified comparing the obtained information with available data reported in the literature. For the quantification, a calibration curve was constructed for each available phenolic standard based on the UV signal (maximum absorption of each standard compound lambda max). When a commercial standard was not available, the quantification was performed through the calibration curve of the most similar available standard. The results were expressed as mg/g of extract.

3.4. Evaluation of the Antioxidant Activity

The lyophilized extracts were re-dissolved in ethanol/water (80:20 v/v) and water for hydroethanolic and aqueous extracts, respectively (final concentration 10 mg/mL), and submitted to various in vitro colorimetric assays (DPPH radical-scavenging activity, reducing power, inhibition of β-carotene bleaching and inhibition of lipid peroxidation—TBARS).
DPPH radical-scavenging activity was performed using an ELX800 Microplate Reader (Bio-Tek). The reaction mixture in each one of the 96-wells consisted of one of the different concentrations of the extracts (30 µL) and the methanolic solution containing DPPH radicals (6 × 10−5 mol/L, 270 µL). The mixture was left to stand for 60 min in the dark. The reduction of the DPPH radical was determined by measuring the absorption at 515 nm. The radical scavenging activity (RSA) was calculated by the equation: % RSA = [(ADPPH − AS)/ADPPH] × 100 (AS is the absorbance of the solution when the sample extract has been added at a particular level and ADPPH is the absorbance of the DPPH solution). The extract concentration providing 50% of the radicals scavenging activity (EC50) was calculated from the graph of RSA percentage against extract concentration.
Reducing power evaluation was performed using the Microplate Reader cited above. The different concentrations of the extracts (0.5 mL) were mixed with sodium phosphate buffer (200 mmol/L, pH 6.6, 0.5 mL) and potassium ferricyanide (1% w/v, 0.5 mL). For each concentration, the mixture was incubated at 50 °C during 20 min, and then was added trichloroacetic acid (10% w/v, 0.5 mL). The mixture (0.8 mL) was poured in the 48-wells, as were deionized water (0.8 mL) and ferric chloride (0.1% w/v, 0.16 mL), and was measured the absorbance at 690 nm. The extract concentration providing 0.5 of absorbance (EC50) was calculated from the graph of absorbance at 690 nm against extract concentrations.
In inhibition of β-carotene bleaching, a solution of β-carotene was previously prepared dissolving β-carotene (2 mg) in chloroform (10 mL) and 2 mL of this solution were pipetted into a round-bottom flask. After chloroform removed (at 40 °C under vacuum) linoleic acid (40 mg), Tween 80 emulsifier (400 mg), and distilled water (100 mL) were added with vigorous shaking. Aliquots (4.8 mL) of this emulsion were transferred into different tubes containing different concentrations of the extracts (0.2 mL). The tubes were shaken and incubated (at 50 °C) in a water bath. As soon as the emulsion was added to each tube, the zero-time absorbance was measured at 470 nm. β-carotene bleaching inhibition was calculated using the equation: β-carotene content after 2 h of assay/initial β-carotene content) × 100. The extract concentration providing 50% antioxidant activity (EC50) was calculated by interpolation from the graph of β-carotene bleaching inhibition percentage against extract concentration.
Lipid peroxidation inhibition was evaluated using a porcine (Sus scrofa) brains (obtained from official slaugh-tered animals). This is homogenized with Polytron in an ice-cold Tris–HCl buffer (20 mM, pH 7.4) to produce a 1:2, w/v brain tissue homogenate which was centrifuged (3000× g; 10 min). Analiquot (100 μL) of the supernatant was incubated with the sample solutions (200 μL) in the presence of FeSO4 (10 mM; 100 μL) and ascorbic acid (0.1 mM; 100 μL) at 37 °C during 1 h. The reaction was stopped by the addition of trichloroacetic acid (28%, w/v, 500 μL), and then thiobarbituric acid (TBA, 2%, w/v, 380 μL). The mixture was then heated at 80 °C during 20 min. After centrifugation (3000× g; 10 min), the color intensity of the malondialdehyde (MDA)–TBA complex in the supernatant was measured (at 532 nm). The inhibition ratio (%) was calculated using the formula: inhibition ratio (%) = [(A − B)/A] × 100% (A were the absorbance of the control and B were the absorbance of sample solution). The extract concentration providing 50% of antioxidant activity (EC50) was calculated from the graph of TBARS formation inhibition against extract concentrations.
All the results were expressed in EC50 values (µg/mL) and trolox was used as positive control [33].

3.5. Evaluation of the Cytotoxicity

3.5.1. General

For cytotoxicity evaluation all lyophilized extracts were re-dissolved in water, with a final solution of 8 mg/mL and diluted to different concentrations. The assay was performed according a procedure described by Guimarães et al. [34]. Ellipticine was used as positive control and the results were calculated as GI50 values (sample concentration that inhibited 50% of the net cell growth).

3.5.2. In Tumor Cell Lines

Four human tumor cell lines were used to acee the cytotoxicity evaluation: MCF-7 (breast adenocarcinoma), NCI-H460 (non-small cell lung cancer), HeLa (cervical carcinoma) and HepG2 (hepatocellular carcinoma). Cells were routinely maintained as adherent cell cultures in RPMI-1640 medium containing 10% heat-inactivated FBS (MCF-7, NCI-H460 and HCT-15) and 2 mM glutamine or in DMEM supplemented with 10% FBS, 2 mM glutamine, 100 U/mL penicillin and 100 mg/mL streptomycin (HeLa and HepG2 cells), at 37 °C, in a humidified air incubator containing 5% CO2. The cell lines were plated an appropriate density (1.0 × 104 cells/well) in 96-well plates and allowed to attach for 24 h. Cells were then treated for 48 h with several extract concentrations. Following this incubation period, the adherent cells were fixed by adding cold 10% trichloroacetic acid (TCA, 100 μL) and incubated during 60 min at 4 °C. Plates were then washed with deionized water and dried; sulforhodamine B solution (0.1% in 1% acetic acid, 100 μL) was then added to each plate well and incubated during 30 min at room temperature. Unbound SRB was removed by washing with 1% acetic acid. Plates were air-dried, the bound SRB was solubilized with 10 mM Tris (200 μL) and the absorbance was measured at 540 nm in the microplate reader cited above. The results were expressed in GI50 values; sample concentration that inhibited 50% of the net cell growth [34].

3.5.3. In Non-Tumor Cells

For the hepatotoxicity evaluation, a cell culture was prepared from a freshly harvested porcine liver obtained from a local slaughter house, and it was designed as PLP2. Briefly, the liver tissues were rinsed in hank’s balanced salt solution containing 100 U/mL penicillin, 100 µg/mL streptomycin and divided into 1 × 1 mm3 explants. Some of these explants were placed in 25 cm2 tissue flasks in DMEM medium supplemented with 10% fetal bovine serum, 2 mM nonessential amino acids and 100 U/mL penicillin, 100 mg/mL streptomycin and incubated at 37 °C with a humidified atmosphere containing 5% CO2. The medium was changed every two days. Cultivation of the cells was continued with direct monitoring every two to three days using a phase contrast microscope. Before confluence was reached, cells were subcultured and plated in 96-well plates at a density of 1.0 × 104 cells/well, and cultivated in DMEM medium with 10% FBS, 100 U/mL penicillin and 100 µg/mL streptomycin [34].

3.6. Evaluation of the Anti-Inflammatory Activity

For the cells treatment, the mouse macrophage-like cell line RAW 264.7 was cultured in DMEM medium supplemented with 10% heat-inactivated fetal bovine serum, glutamine and antibiotics at 37 °C under 5% CO2, in humidified air. For each experiment, cells were detached with a cell scraper. In the experiment cell density of 5 × 105 cells/mL was used, and the proportion of dead cells was less than 5% according to the Trypan blue dye exclusion test. Cells were seeded in 96-well plates at 150,000 cells/well and allowed do attach to the plate overnight. Subsequently, cells were treated with the several concentrations of each extract during 1 h. Dexamethasone (50 µM) was used as a positive control for the experiment. The following step was the stimulation with LPS (1 µg/mL) for 18 h. The effect of all the tested samples in the absence of LPS was also analyzed, to observe if they induced changes in Nitric oxide (NO) basal levels. In negative controls, no LPS was added. Both extracts and LPS were dissolved in supplemented DMEM.
Anti-inflammatory assay was performed in concentration range 400–125 μg/mL and dexamethasone (50 μM) was used as a positive control. The mouse macrophage-like cell line RAW 264.7 stimulated with LPS was used in the assay. Nitric oxide (NO) production was studied with Griess Reagent System kit. Results were expressed as EC50 values (μg/mL) equal to the sample concentration providing a 50% inhibition of NO production [35].

3.7. Evaluation of the Antimicrobial Activity

3.7.1. Antibacterial Activity

Escherichia coli (ATCC (American type culture collection) 35210), Enterobacter cloacae (ATCC 35030), Salmonella typhimurium (ATCC 13311), and Pseudomonas aeruginosa (ATCC 27853), were the used Gram-negative bacteria used, while Listeria monocytogenes (NCTC (National collection of type cultures) 7973), Micrococcus flavus (ATCC 10240), Bacillus cereus (clinical isolate), Staphylococcus aureus (ATCC 6538), were the used Gram-positive bacteria. The minimum inhibitory (MIC) and minimum bactericidal (MBC) concentrations were determined by the microdilution method. Each fresh overnight culture of bacteria was adjusted spectrophotometrically (625 nm) to a concentration of 1 × 105 CFU/mL. Dilutions of inocula were cultured on solid medium to verify the absence of contamination and check the validity of each inoculum. Different dilutions of the aqueous extract were added to the wells containing 100 μL of Tryptic Soy Broth (TSB) and afterwards, 10 μL of inoculum was added to all wells. The microplates were incubated for 24 h at 37 °C. The MIC of the samples was detected following the addition of 40 μL of iodonitrotetrazolium chloride (INT) (0.2 mg/mL) and incubation at 37 °C for 30 min. The lowest concentration that produced a significant inhibition (around 50%) of the growth of the bacteria in comparison with the positive control was identified as the MIC. MICs, obtained from the susceptibility testing of various bacteria to tested extracts were determined also by a colorimetric microbial viability assay based on the reduction of the INT colour and compared with a positive control for each bacterial strain. MBC were determined by serial sub-cultivation of 10 μL into microplates containing 100 μL of TSB. The lowest concentration that showed no growth after this sub-culturing was read as the MBC [36]. The results were expressed in mg/mL.

3.7.2. Antifungal Activity

Aspergillus ochraceus (ATCC 12066), Aspergillus versicolor (ATCC 11730), Aspergillus niger (ATCC 6275), Aspergillus fumigatus (ATCC 1022), Trichoderma viride (IAM (Culture Collection, Center for Cellular and Molecular Research, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Japan) 5061), Penicillium funiculosum (ATCC 36839), Penicillium ochrochloron (ATCC 9112) and Penicillium verrucosum var. cyclopium (food isolate) were used. Fungal spores were washed from the surface of agar plates with sterile 0.85% saline containing 0.1% Tween 80 (v/v). The spore suspension was adjusted with sterile saline to a concentration of approximately 1.0 × 105 in a final volume of 100 μL per well. The inocula were stored at 4 °C for further use. Dilutions of each inoculum were cultured on solid MA to verify the absence of contamination and to check the validity of the inoculum. MIC determination was also performed by a serial dilution technique using 96-well microtitre plates. The investigated sample was dissolved in water and added to broth malt medium with a fungal inoculum. The microplates were incubated during 72 h at 28 °C. The lowest concentrations without visible growth (as assessed using a binocular microscope) were defined as the MICs. The minimum bactericidal and fungicidal concentrations (MBC and MFC) were determined by serial sub-cultivation of 2 μL in microtitre plates containing 100 μL of malt broth per well and further incubation for 72 h at 28 °C. The lowest concentration with no visible growth was defined as the MFC, indicating 99.5% killing of the original inoculum. Streptomycin and ampicillin, bifonazole and ketoconazole (in a range of 0.01 to 5 mg/mL) were used as positive controls and for negative control 5% DMSO was used [36]. The results were expressed in μg/mL.

3.8. Statistical Analysis

In this study all the assays were carried out in triplicate. The results are expressed as mean values and standard deviation (SD). All results were analyzed using one-way analysis of variance (ANOVA) followed by Tukey’s HSD Test with α = 0.05. These analyses were carried out using IBM SPSS Statistics, Version 23.0. (IBM Corp., Armonk, New York, USA).

4. Conclusions

Overall, the study of natural matrices allows a greater knowledge of their chemical composition and, therefore, a more adequate applicability. The present study allowed to deep knowledge on the phenolic profile of L. pedunculata, since samples with different origins were studied. In general, most of the samples revealed diverse bioactive properties, such as, cytotoxicity, antioxidant, anti-inflammatory and antimicrobial activity. The aqueous extracts showed a higher concentration of phenolic compounds and a higher antioxidant activity; nevertheless, the hydroethanolic extracts exhibited a higher anti-inflammatory potential in most of the samples, as also a higher antiproliferative capacity and antimicrobial activity. This may be due to the fact that hydroethanolic extracts contains other compounds (non-phenolic compounds), which could be correlated with these bioactivities.
Besides the great importance of L. pedunculata bioactive properties, these results also highlight the existing variances between samples from different geographical origin. Further studies are required to correlate biotic and abiotic factors with the chemical composition of the specimens.
The studied plant materials were obtained from previously conserved seeds (accessions) of different L. pedunculata populations, of the Portuguese Genebank collection. These outcomes contributed to data on important chemical characteristics, which might distinguish accessions within a species. Such characterization provides essential information assuring the best utilization of the conserved germplasm to the final users, meeting the goal of ex situ conservation of germplasm in genebanks.

Author Contributions

I.C.F.R.F. and A.M.C. designed the experiments. A.M.C., A.M.B., V.L. and F.R. collaborated in the supply of samples. C.L.L. and E.P. performed all the experimental assays with collaboration of L.B. (phenolic compounds analysis), R.C. (cytotoxicity and anti-inflammatory activity) and M.S. (antimicrobial activity). I.C.F.R.F., A.M.C., C.L.L., E.P., and L.B. collaborated in the manuscript elaboration.

Acknowledgments

The authors are grateful to the Foundation for Science and Technology (FCT, Portugal) and FEDER under Program PT2020 for financial support to CIMO (UID/AGR/00690/2013), L. Barros and R. Calhelha contracts. The authors are grateful to FEDER-Interreg España-Portugal programme for financial support through the project 0377_Iberphenol_6_E.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Komes, D.; Belščak-cvitanović, A.; Horžić, D. Phenolic composition and antioxidant properties of some traditionally used medicinal plants affected by the extraction time and hydrolysis. Phytochem. Anal. 2011, 22, 172–180. [Google Scholar] [CrossRef] [PubMed]
  2. Kaefer, C.M.; Milner, J.A. The role of herbs and spices in cancer prevention. J. Nutr. Biochem. 2008, 19, 347–361. [Google Scholar] [CrossRef] [PubMed]
  3. Costa, P.; Gonçalves, S.; Valentão, P.; Andrade, P.B.; Almeida, C.; Nogueira, J.M.F.; Romano, A. Metabolic profile and biological activities of Lavandula pedunculata subsp. lusitanica (Chaytor) Franco: Studies on the essential oil and polar extracts. Food Chem. 2013, 141, 2501–2506. [Google Scholar] [CrossRef] [PubMed]
  4. Port, S.; Chisté, R.C.; Godoy, H.T.; Prado, M.A. The phenolic compounds and the antioxidant potential of infusion of herbs from the Brazilian Amazonian region. Food Res. Int. 2013, 53, 875–881. [Google Scholar]
  5. Bajpai, V.K.; Agrawal, P.; Bang, B.H.; Park, Y.-H. Phytochemical analysis, antioxidant and antilipid peroxidation effects of a medicinal plant, Adhatoda vasica. Front. Life Sci. 2015, 8, 305–312. [Google Scholar] [CrossRef]
  6. Quer, P.F. Plantas Medicinales—El Dioscorides Renovado; Ediciones Península: Barcelona, Spain, 1999. [Google Scholar]
  7. Zuzarte, M.R.; Dinis, A.M.; Cavaleiro, C.; Salgueiro, L.R.; Canhoto, J.M. Trichomes, essential oils and in vitro propagation of Lavandula pedunculata (Lamiaceae). Ind. Crops Prod. 2010, 32, 580–587. [Google Scholar] [CrossRef]
  8. Lopes, V.; Barata, A.M. Ex situ morphological assessment of wild Lavandula populations in Portugal. Arab. J. Med. Aromat. Plants 2017, 3, 87–100. [Google Scholar]
  9. Morales, R. Lavandula L. In Flora Iberica; Castroviejo, S., Ed.; Real Jardín Botánico, CSIC: Madrid, Spain, 2010; Volume 12, pp. 484–496. [Google Scholar]
  10. Neves, J.M.; Matosa, C.; Moutinho, C.; Queiroz, G.; Gomes, L.R. Ethnopharmacological notes about ancient uses of medicinal plants in Trás-os-Montes (northern of Portugal). J. Ethnopharmacol. 2009, 124, 270–283. [Google Scholar] [CrossRef] [PubMed]
  11. Santos, E.S.; Abreu, M.M.; Saraiva, J.A. Mutielemental concentration and physiological responses of Lavandula pedunculata growing in soils developed on different mine. Environ. Pollut. 2016, 213, 43–52. [Google Scholar] [CrossRef] [PubMed]
  12. Torras-Claveria, L.; Jauregui, O.; Bastida, J.; Codina, C.; Viladomat, F. Antioxidant Activity and Phenolic Composition of Lavandin (Lavandula x intermedia Emeric ex Loiseleur) Waste. J. Agric. Food Chem. 2007, 55, 8436–8443. [Google Scholar] [CrossRef] [PubMed]
  13. Costa, P.; Gonçalves, S.; Valentão, P.; Andrade, P.B.; Romano, A. Accumulation of phenolic compounds in in vitro cultures and wild plants of Lavandula viridis L’Hér and their antioxidant and anti-cholinesterase potential. Food Chem. Toxicol. 2013, 57, 69–74. [Google Scholar] [CrossRef] [PubMed]
  14. Nunes, R.; Pasko, P.; Tyszka-Czochara, M.; Szewczyk, A.; Szlosarczyk, M.; Carvalho, I.S. Antibacterial, antioxidant and anti-proliferative properties and zinc content of five south Portugal herbs. Pharm. Biol. 2016, 55, 114–123. [Google Scholar] [CrossRef] [PubMed]
  15. Pereira, F.; Baptista, R.; Ladeiras, D.; Madureira, A.M.; Teixeira, G.; Rosado, C.; Rijo, P. Production and characterization of nanoparticles containing methanol extracts of Portuguese Lavenders. Measurement 2015, 74, 170–177. [Google Scholar] [CrossRef]
  16. Barros, L.; Dueñas, M.; Dias, M.I.; Sousa, M.J.; Santos-Buelga, C.; Ferreira, I.C.F.R. Phenolic profiles of cultivated, in vitro cultured and commercial samples of Melissa officinalis L. infusions. Food Chem. 2013, 136, 1–8. [Google Scholar] [CrossRef] [PubMed]
  17. Chen, H.; Zhang, Q.; Wang, X.; Yang, J.; Wang, Q. Qualitative analysis and simultaneous quantification of phenolic compounds in the aerial Parts of Salvia miltiorrhiza by HPLCDAD and ESI/MSn. Phytochem. Anal. 2011, 22, 247–257. [Google Scholar] [CrossRef] [PubMed]
  18. Zeng, G.; Xiao, H.; Liu, J.; Liang, X. Identification of phenolic constituents in Radix Salvia miltiorrhizae by liquid chromatography/electrospray ionization mass spectrometry. Rapid Commun. Mass Spectrom. 2006, 20, 499–506. [Google Scholar] [CrossRef] [PubMed]
  19. Ruan, M.; Li, Y.; Li, X.; Luo, J.; Kong, L. Qualitative and quantitative analysis of the major constituents in Chinese medicinal preparation Guan-Xin-Ning injection by HPLC-DAD-ESI-MSn. J. Pharm. Biomed. Anal. 2012, 59, 184–189. [Google Scholar] [CrossRef] [PubMed]
  20. Pereira, C.; Barros, L.; Carvalho, A.M.; Santos-Buelga, C.; Ferreira, I.C.F.R. Infusions of artichoke and milk thistle represent a good source of phenolic acids and flavonoids. Food Funct. 2015, 6, 56. [Google Scholar] [CrossRef] [PubMed]
  21. Lech, K.; Witkoś, K.; Jaros, M. HPLC–UV–ESI MS/MS identification of the color constituents of sawwort (Serratula tinctoria L.). Anal. Bioanal. Chem. 2014, 406, 3703–3708. [Google Scholar] [CrossRef] [PubMed]
  22. Correa, R.C.G.; Souza, A.H.P.; Calhelha, R.C.; Barros, L.; Glamoclija, J.; Sokovic, M.; Peralta, R.M.; Bracht, M.; Ferreira, I.C.F.R. Bioactive formulations prepared from fruiting bodies and submerged culture mycelia of the Brazilian edible mushroom Pleurotus ostreatoroseus Singer. Food Funct. 2015, 6, 2155–2164. [Google Scholar] [CrossRef] [PubMed]
  23. Ferreira, A.; Proença, C.; Serralheiro, M.L.M.; Araújo, M.E.M. The in vitro screening for acetylcholinesterase inhibition and antioxidant activity of medicinal plants from Portugal. J. Ethnopharmacol. 2006, 108, 31–37. [Google Scholar] [CrossRef] [PubMed]
  24. Baptista, R.; Madureira, A.M.; Jorge, R.; Adão, R.; Duarte, A.; Duarte, N.; Lopes, M.M.; Teixeira, G. Antioxidant and Antimycotic Activities of Two Native Lavandula Species from Portugal. Evid.-Based Complement. Altern. Med. 2015, 2015, 1–10. [Google Scholar] [CrossRef] [PubMed]
  25. Algieri, F.; Rodriguez-Nogales, A.; Vezza, T.; Garrido-Mesa, J.; Garrido-Mesa, N.; Utrilla, M.P.; González-Tejero, M.R.; Casares-Porcel, M.; Molero-Mesa, J.; Contreras, M.M.; et al. Anti-inflammatory activity of hydroalcoholic extracts of Lavandula dentata L. and Lavandula stoechas L. J. Ethnopharmacol. 2016, 190, 142–158. [Google Scholar] [CrossRef] [PubMed]
  26. Tang, S.; Shia, J.; Liu, C.; Zhang, J.; Xang, N.; Liu, X.; Chen, Y.; You, J.; Shen, O.; Miao, M.; et al. Three new arylbenzofurans from Lavandula angustifolia and their bioactivities. Phytochem. Lett. 2017, 19, 60–63. [Google Scholar] [CrossRef]
  27. Carocho, M.; Calhelha, R.C.; Queirozc, M.J.R.Q.P.; Bento, A.; Morales, P.; Sokovic, M.; Ferreira, I.C.F.R. Infusions and decoctions of Castanea sativa flowers a seff ective antitumor and antimicrobial matrices. Ind. Crops Prod. 2014, 62, 42–46. [Google Scholar] [CrossRef]
  28. Rashed, K.; Ciric, A.; Glamoclijab, J.; Calhelha, R.C.; Ferreira, I.C.F.R.; Sokovic, M. Antimicrobial and cytotoxic activities of Alnus rugosa L. aerial parts and identification of the bioactive components. Ind. Crops Prod. 2014, 59, 189–196. [Google Scholar]
  29. Živkovic, J.; Barreira, J.C.M.; Stojkovic, D.; Cebovic, T.; Santos-Buelga, C.; Maksimovic, Z.; Ferreira, I.C.F.R. Phenolic profile, antibacterial, antimutagenic and antitumour evaluation of Veronica urticifolia Jacq. J. Funct. Foods 2014, 9, 192–201. [Google Scholar] [CrossRef]
  30. Nikolic, M.; Glamoclija, J.; Ferreira, I.C.F.R.; Calhelha, R.C.; Fernandes, A.; Markovic, T.; Markovic, D.; Giwelie, A.; Sokovic, M. Chemical composition, antimicrobial, antioxidant and antitumor activity of Thymus serpyllum L., Thymus algeriensis Boiss. and Reut and Thymus vulgaris L. essential oils. Ind. Crops Prod. 2014, 52, 183–190. [Google Scholar] [CrossRef]
  31. Pereira, E.; Barros, L.; Calhelha, R.C.; Dueñas, M.; Carvalho, A.M.; Santos-Buelga, C.; Ferreira, I.C.F.R. Bioactivity and phytochemical characterization of Arenaria montana L. Food Funct. 2014, 5, 1848. [Google Scholar] [CrossRef] [PubMed]
  32. Bessada, S.M.F.; Barreira, J.C.M.; Barros, L.; Ferreira, I.C.F.R.; Oliveira, M.B.P.P. Phenolic profile and antioxidant activity of Coleostephus myconis (L.) Rchb.f.: An underexploited and highly disseminated species. Ind. Crops Prod. 2016, 89, 45–51. [Google Scholar] [CrossRef]
  33. Martins, N.; Barros, L.; Dueñas, M.; Santos-Buelga, C.; Ferreira, I.C.F.R. Characterization of phenolic compounds and antioxidant properties of Glycyrrhiza glabra L. rhizomes and roots. RSC Adv. 2015, 5, 26991–26997. [Google Scholar] [CrossRef]
  34. Guimarães, R.; Barros, L.; Dueñas, M.; Calhelha, R.C.; Carvalho, A.M.; Santos-Buelga, C.; Queiroz, M.J.R.P.; Ferreira, I.C.F.R. Infusion and decoction of wild German chamomile: Bioactivity and characterization of organic acids and phenolic compounds. Food Chem. 2013, 136, 947–954. [Google Scholar] [CrossRef] [PubMed]
  35. Taofiq, O.; Calhelha, R.C.; Heleno, S.; Barros, L.; Martins, A.A.; Santos-Buelga, C.; Queiroz, M.J.R.P.; Ferreira, I.C.F.R. The contribution of phenolic acids to the anti-inflammatory activity of mushrooms: Screening in phenolic extracts, individual parent molecules and synthesized glucuronated and methylated derivatives. Food Res. Int. 2015, 76, 821–827. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Carocho, M.; Barros, L.; Calhelha, R.C.; Ćirić, A.; Soković, M.; Santos-Buelga, C.; Ferreira, I.C.F.R. Melissa officinalis L. decoctions as functional beverages: A bioactive approach and chemical characterization. Food Funct. 2015, 6, 2240–2248. [Google Scholar] [CrossRef] [PubMed]
Sample Availability: Samples of the compounds are available from the authors.
Figure 1. Chromatographic profile of sample 3 (Vila Viçosa, Évora) recorded at 280 nm, the peak numbers correspond to the compounds listed in Table 1.
Figure 1. Chromatographic profile of sample 3 (Vila Viçosa, Évora) recorded at 280 nm, the peak numbers correspond to the compounds listed in Table 1.
Molecules 23 01037 g001
Table 1. Retention time (Rt), wavelengths of maximum absorption in the visible region (λmax), mass spectral data and tentative identification of phenolic compounds in L. pedunculata samples.
Table 1. Retention time (Rt), wavelengths of maximum absorption in the visible region (λmax), mass spectral data and tentative identification of phenolic compounds in L. pedunculata samples.
PeakRt (min)λmax (nm)[M-H] (m/z)MS2 (m/z)Tentative IdentificationIdentification Type
15.97320341179(100)Caffeic acid hexoside[12], DAD/MS
26.57320341179(100)Caffeic acid hexoside[12], DAD/MS
38.65310325163(100)p-Coumaroyl hexoside[12], DAD/MS
410.16320179119(100)Caffeic acidStandard/DAD/MS
513.22332623461(27), 285(100)Luteolin-O-hexosyl-O-glucuronide[20], DAD/MS
616.34284,330 sh463287(100)Eriodictyol-O-glucuronide[21], DAD/MS
717.64320521359(100), 197(8), 179(14), 161(3), 135(5)Rosmarinic acid hexoside[22], DAD/MS
818.68347461285(100)Luteolin-7-O-glucuronideStandard/DAD/MS
919.23283,338 sh719539(29), 521(18), 359(100), 179(32), 161(5), 135(5)Sangerinic acid[16], DAD/MS
1022.29325359197(27), 179(37), 161(100), 135(5)Rosmarinic acid Standard/DAD/MS
1123.19288,328 sh537493(18), 359(100), 313(11), 295(5), 197(3), 179(3)Lithospermic acid A[17,18,19], DAD/MS
1224.65340475299(100), 284(68)Methylluteolin-O-glucuronide[21], DAD/MS
1325.75309,338sh717537(47), 519(17), 493(40), 359(97), 339(10), 321(8), 313(17), 295(100), 197(7), 179(27)Salvianolic acid B[18,19], DAD/MS
Table 2. Phenolic compounds quantification (mg/g of extract, mean ± SD) in L. pedunculata hydroethanolic and aqueous extracts.
Table 2. Phenolic compounds quantification (mg/g of extract, mean ± SD) in L. pedunculata hydroethanolic and aqueous extracts.
12345678910111213
Hydroethanol Extract
Peak 10.28 ± 0.01 f0.034 ± 0.003 k0.179 ± 0.001 itr0.210 ± 0.005 h0.23 ± 0.01 g0.11 ± 0.04 j2.44 ± 0.02 a0.37 ± 0.01 d0.44 ± 0.01 c0.277 ± 0.004 f0.59 ± 0.02 b0.33 ± 0.02 e
Peak 20.18 ± 0.02 h0.079 ± 0.003 j0.31 ± 0.001 d0.120 ± 0.003 i0.22 ± 0.02 g0.295 ± 0.001 e0.24 ± 0.01 f1.87 ± 0.02 a0.40 ± 0.01 c0.31 ± 0.02 de0.22 ± 0.01 fg0.54 ± 0.02 b0.115 ± 0.001 i
Peak 30.77 ± 0.01 f1.002 ± 0.001 d0.80 ± 0.002 f0.50 ± 0.01 h0.95 ± 0.02 e0.98 ± 0.01 de1.48 ± 0.03 a1.27 ± 0.07 b1.16 ± 0.03 c0.98 ± 0.03 de0.80 ± 0.01 f1.26 ± 0.01 b0.57 ± 0.03 g
Peak 40.63 ± 0.02 i0.805 ± 0.002 f0.88 ± 0.001 d0.639 ± 0.002 hi1.01 ± 0.02 c0.85 ± 0.02 e1.14 ± 0.02 b1.49 ± 0.03 a1.00 ± 0.04 c0.802 ± 0.001 f0.72 ± 0.03 g1.12 ± 0.04 b0.65 ± 0.02 h
Peak 52.05 ± 0.03 f2.33 ± 0.03 c2.68 ± 0.01 b1.37 ± 0.01 k1.846 ± 0.001 g1.84 ± 0.01 g1.599 ± 0.004 i1.54 ± 0.01 j2.22 ± 0.01 d2.695 ± 0.003 a2.09 ± 0.01 e2.06 ± 0.01 f1.698 ± 0.003 h
Peak 60.137 ± 0.003 htr0.40 ± 0.01 c0.74 ± 0.01 b0.082 ± 0.003 itr0.060 ± 0.001 j0.782 ± 0.003 atr0.299 ± 0.005 d0.150 ± 0.003 g0.231 ± 0.004 e0.17 ± 0.01 f
Peak 71.18 ± 0.01 g1.379 ± 0.001 de1.52 ± 0.01 b0.93 ± 0.02 i1.40 ± 0.05 cd1.355 ± 0.004 ef1.39 ± 0.04 cde1.78 ± 0.06 a1.51 ± 0.09 b1.32 ± 0.01 f1.38 ± 0.05 de1.43 ± 0.03 c1.047 ± 0.002 h
Peak 813.10 ± 0.01 l19.93 ± 0.02 e21.68 ± 0.03 c22.35 ± 0.06 b12.15 ± 0.02 m17.37 ± 0.02 i18.62 ± 0.01 g20.66 ± 0.01 d19.7 ± 0.1 f15.60 ± 0.02 k17.57 ± 0.01 h16.54 ± 0.02 j24.36 ± 0.04 a
Peak 91.99 ± 0.01 e2.34 ± 0.05 c2.25 ± 0.04 d2.90 ± 0.01 a1.77 ± 0.08 g1.97 ± 0.01 ef2.38 ± 0.03 bc2.4 ± 0.1 b2.84 ± 0.08 a2.20 ± 0.05 d1.93 ± 0.01 f1.13 ± 0.04 i1.41 ± 0.03 h
Peak 1021.4 ± 0.40 j34.4 ± 0.4 e38.5 ± 0.60 b16.9 ± 0.5 k36.0 ± 0.2 c32.5 ± 0.6 f26.11 ± 0.05 h50.9 ± 0.9 a30.4 ± 0.4 g35.2 ± 0.6 d23.5 ± 0.2 i17.0 ± 0.6 k7.5 ± 0.2 l
Peak 112.89 ± 0.01 k4.27 ± 0.06 f5.4 ± 0.4 c4.00 ± 0.09 gh3,52 ± 0.08 j3.546 ± 0.002 ij3.69 ± 0.09 i6.4 ± 0.1 a5.15 ± 0.04 d4.4 ± 0.1 e3.95 ± 0.1 h4.1 ± 0.6 fg6.0 ± 0.01 b
Peak 122.520 ± 0.001 i2.73 ± 0.02 h5.09 ± 0.02 a3.57 ± 0.01 c1.78 ± 0.01 k2.41 ± 0.01 j2.76 ± 0.02 gnd3.14 ± 0.01 f2.41 ± 0.01 j5.06 ± 0.01 b3.41 ± 0.03 e3.52 ± 0.001 d
Peak 1320.2 ± 0.50 h36.8 ± 0.3 b30.9 ± 0.5 d14.89 ± 0.04 j30.27 ± 0.04 e30.9 ± 0.8 d29.8 ± 0.4 f44.3 ± 0.2 a34.1 ± 0.5 c30.6 ± 0.4 de25.9 ± 0.4 g16.7 ± 0.5 i8.7 ± 0.01 k
TPA50 ± 1 h81 ± 1 b81 ± 2 b40.8 ± 0.4 j75.4 ± 0.3 d72.6 ± 0.2 e66 ± 1 f113 ± 1 a77 ± 1 c76.351 ± 0.003 c59 ± 1 g43.97 ± 0.04 i26.3 ± 0.5 k
TF17.81 ± 0.01 l25.0 ± 0.01 e29.85 ± 0.03 a28.04 ± 0.04 c15.86 ± 0.03 m21.62 ± 0.01 j23.04 ± 0.01 g22.988 ± 0.002 h25.1 ± 0.1 d21.00 ± 0.02 k24.86 ± 0.01 f22.2 ± 0.1 i29.75 ± 0.02 b
TPC68 ± 1 k106 ± 1 c111 ± 2 b68.9 ± 0.5 i91.3 ± 0.3 g94.2 ± 0.2 f89 ± 1 h136 ± 1 a102 ± 1 d97.36 ± 0.02 e84 ± 1 i66.17 ± 0.02 l56.1 ± 0.5 m
Aqueous Extract
Peak 11.18 ± 0.06 d0.87 ± 0.04 g1.02 ± 0.02 e0.59 ± 0.02 h1.31 ± 0.01 b1.20 ± 0.06 cd1.23 ± 0.01 c7.9 ± 0.1 a1.19 ± 0.06 d0.91 ± 0.05 fg0.96 ± 0.04 f1.02 ± 0.05 e0.28 ± 0.04 i
Peak 22.36 ± 0.02 h1.97 ± 0.04 j2.65 ± 0.01 f2.55 ± 0.08 g2.31 ± 0.01 hi3.8 ± 0.1 c3.83 ± 0.03 c5.03 ± 0.07 a2.37 ± 0.05 h2.28 ± 0.05 i4.3 ± 0.01 b3.19 ± 0.03 d3.06 ± 0.08 e
Peak 33.5 ± 0.1 d3.18 ± 0.03 g3.002 ± 0.002 h1.52 ± 0.06 l3.22 ± 0.02 f3.83 ± 0.02 c4.19 ± 0.06 a4.13 ± 0.03 b2.98 ± 0.04 h3.30 ± 0.03 e2.77 ± 0.08 j2.19 ± 0.09 k2.90 ± 0.05 i
Peak 43.25 ± 0.006 f3.46 ± 0.08 e2.917 ± 0.004 h2.80 ± 0.03 i3.29 ± 0.04 f3.14 ± 0.08 g3.99 ± 0.08 b3.87 ± 0.05 c3.18 ± 0.04 g2.80 ± 0.04 i2.61 ± 0.05 j3.63 ± 0.01 d4.35 ± 0.04 a
Peak 56.49 ± 0.02 c6.71 ± 0.02 b8.76 ± 0.01 a3.681 ± 0.003 k5.18 ± 0.04 f5.28 ± 0.01 d4.1 ± 0.10 i4.45 ± 0.02 g3.32 ± 0.02 l3.79 ± 0.01 j4.34 ± 0.01 h2.95 ± 0.03 m5.22 ± 0.03 e
Peak 612.83 ± 0.04 d9.36 ± 0.02 h8.87 ± 0.03 i16.73 ± 0.04 a8.6 ± 0.1 k10.8 ± 0.1 g11.6 ± 0.10 f12.9 ± 0.1 d8.17 ± 0.02 l12.97 ± 0.04 c13.31 ± 0.05 b8.8 ± 0.1 j12.26 ± 0.02 e
Peak 72.52 ± 0.01 cd2.41 ± 0.09 e3.33 ± 0.01 a1.6 ± 0.2 h2.45 ± 0.08 de2.26 ± 0.04 f2.38 ± 0.07 e2.83 ± 0.01 b2.61 ± 0.03 c3.3 ± 0.2 a2.16 ± 0.09 g2.74 ± 0.01 b2.11 ± 0.06 g
Peak 870.81 ± 0.04 g64.31 ± 0.02 k76.4 ± 0.1 d101.5 ± 0.1 a42.12 ± 0.01 m66.8 ± 0.1 i65.9 ± 0.1 j84.1 ± 0.1 c73.56 ± 0.09 f67.35 ± 0.08 h73.8 ± 0.1 e53.6 ± 0.2 l99.43 ± 0.02 b
Peak 94.4 ± 0.1 ef3.26 ± 0.03 i4.4 ± 0.4 ef6.6 ± 0.1 a3.73 ± 0.03 h4.3 ± 0.5 g4.6 ± 0.1 e5.2 ± 0.1 d6.2 ± 0.5 b5.7 ± 0.4 c3.30 ± 0.01 i4.3 ± 0.30 g5.8 ± 0.3 c
Peak 10290.0 ± 0.4 e293 ± 3 d353.0 ± 0.3 b190 ± 2 j288.6 ± 0.2 f318.0 ± 0.7 c211 ± 1 i550 ± 3 a266.3 ± 0.7 g291 ± 4 e218.4 ± 0.5 h52.2 ± 0.6 k27.1 ± 0.3 l
Peak 1111.5 ± 0.3 i13.9 ± 0.4 g16.37 ± 0.06 c15.1 ± 0.4 d9.8 ± 0.3 j12.2 ± 0.6 h9.63 ± 0.02 j16.8 ± 0.2 b14.83 ± 0.07 de14.7 ± 0.4 e14.14 ± 0.09 f16.9 ± 0.1 b26.2 ± 0.6 a
Peak 1211.25 ± 0.03 e7.75 ± 0.02 j16.08 ± 0.01 b15.1 ± 0.1 c4.45 ± 0.04 k8.54 ± 0.04 i8.9 ± 0.1 hnd10.9 ± 0.1 f8.51 ± 0.02 i19.8 ± 0.1 a9.73 ± 0.02 g13.7 ± 0.1 d
Peak 13381 ± 5 d390 ± 2 c366.7 ± 0.9 e212.9 ± 0.4 k309.9 ± 0.4 h405 ± 3 b279 ± 2 j582 ± 1 a343 ± 2 f340 ± 4 g305.7 ± 0.5 i62.8 ± 0.3 l47.7 ± 0.8 m
TPA699 ± 5 d712 ± 1 c753.4 ± 0.7 b433 ± 3 j621.6 ± 0.8 g754 ± 3 b519 ± 3 i1177 ± 3 a643 ± 2 f664 ± 9 e554.3 ± 0.1 h148.9 ± 0.6 k119 ± 1 l
TF101.4 ± 0.2 e88.13 ± 0.05 j110.06 ± 0.01 d137.0 ± 0.2 a60.3 ± 0.1 l91.4 ± 0.1 h90.4 ± 0.2 i101.4 ± 0.2 e96.0 ± 0.2 f92.6 ± 0.1 g111.2 ± 0.1 c75.1 ± 0.3 k130.6 ± 0.2 b
TPC801 ± 5 d801 ± 1 d863.5 ± 0.7 b570 ± 3 j681.9 ± 0.7 g845 ± 3 c610 ± 3 i1278 ± 3 a739 ± 2 f756 ± 9 e665.6 ± 0.2 h224 ± 1 l250 ± 1 k
Notes: nd-not detected; tr-traces. TPA—Total phenolic acids; TF—Total flavonoids; TPC—Total phenolic compounds; Peaks: 1—caffeic acid hexoside; 2—caffeic acid hexoside; 3—p-coumaroyl hexoside; 4—caffeic acid; 5—luteolin-O-hexosyl-O-glucuronide; 6—eriodictyol-O-glucuronide; 7—rosmarinic acid hexoside; 8—luteolin-7-O-glucuronide; 9—sangerinic acid; 10—rosmarinic acid; 11—lithospermic acid A; 12—methylluteolin-O-glucuronide; 13—salvianolic acid B. Standard calibration curves: caffeic acid (y = 388,345x + 406,369, R² = 0.994; LOD = 0.78 μg/mL; LOQ = 1.97 μg/mL) (compounds 1, 2 and 4); p-coumaric acid (y = 301,950x + 6966.7, R² = 0.999; LOD = 0.68 μg/mL; LOQ = 1.61 μg/mL) (compound 3); luteolin-7-O-glucoside (y = 4087x + 72,589, R² = 0.999; LOD = 0.21 μg/mL; LOQ = 0.74 μg/mL) (compounds 5, 8 and 12); hesperetin (y = 34156x + 268,027, R² = 0.999; LOD = 0.42 µg/mL; LOQ = 0.87 µg/mL) (compound 6); and rosmarinic acid (y = 191,291x − 652,903, R² = 0.999; LOD = 0.15 µg/mL; LOQ = 0.68 µg/mL) (compounds 7, 9, 10, 11 and 13). The results were analyzed using one-way analysis of variance (ANOVA) followed by Tukey’s HSD Test, and in each row and for each extract (hydroethanolic or aqueous extracts) different letters (a to m) mean significant differences among total compounds (p < 0.05).
Table 3. Antioxidant activity and anti-inflammatory potential of L. pedunculata hydroethanolic and aqueous extracts (mean ± SD).
Table 3. Antioxidant activity and anti-inflammatory potential of L. pedunculata hydroethanolic and aqueous extracts (mean ± SD).
12345678910111213
Hydroethanolic Extracts
Antioxidant Activity (EC50 Values, μg/mL)
DPPH scavenging activity142 ± 6 ef139 ± 3 ef139 ± 2 ef212 ± 8 b137 ± 3 f139 ± 1 f150 ± 3 d87 ± 2 g150 ± 2 d146 ± 8 de181 ± 2 c140 ± 5 ef257 ± 7 a
Reducing power110 ± 1 f98 ± 1 g117.9 ± 0.5 e149 ± 2 b97 ± 2 g107 ± 2 f129 ± 1 d72 ± 1 h67 ± 1 i135 ± 9 c133 ± 3 cd133 ± 1 cd216 ± 6 a
β-carotene bleaching inhibition1009 ± 88 d821 ± 34 e1062 ± 65 d516 ± 75 f1176 ± 40 c1578 ± 77 b541 ± 43 f1833 ± 178 a372 ± 18 g252 ± 7 h190 ± 20 h223 ± 3 h214 ± 10 h
TBARS inhibition28 ± 1 g17 ± 1 i26 ± 1 h35 ± 1 e38.25 ± 0.01 d63.5 ± 0.1 a44.56 ± 0.01 c25.57 ± 0.02 h38.2 ± 0.3 d48 ± 2 b62 ± 4 a27.7 ± 0.05 gh31.4 ± 0.1 f
Anti-Inflammatory Potential (EC50 Values, µg/mL)
Nitric oxide (NO) production190 ± 3 c198 ± 5 b190 ± 6 c>400>400124 ± 8 f>400>400>400216 ± 12 a171 ± 3 d162 ± 4 e>400
Aqueous Extracts
Antioxidant Activity (EC50 Values. μg/mL)
DPPH scavenging activity109 ± 4 f99 ± 2 g99 ± 1 g68.0 ± 0.5 i99 ± 2 g115 ± 1 e125 ± 2 d69 ± 3 h98 ± 3 g115 ± 2 e133 ± 3 c144 ± 6 b191 ± 2 a
Reducing power101 ± 2 f51 ± 1 i113 ± 4 e137 ± 4 b99 ± 2 f93 ± 3 g111 ± 2 e73 ± 1 h110 ± 2 e125 ± 4 cd122 ± 6 d130 ± 6 c167 ± 1 a
β-carotene bleaching inhibition274 ± 10 f253 ± 14 fg267 ± 9 f385 ± 13 d472 ± 14 b475 ± 19 b422 ± 25 c388 ± 21 d452 ± 12 b236 ± 14 g305 ± 4 e391 ± 25 d530 ± 21 a
TBARS inhibition14 ± 1 i24.8 ± 0.4 de16 ± 1 h29 ± 1 c21.8 ± 0.4 fg22 ± 1 f21.15 ± 0.01 g21.2 ± 0.2 g21 ± 1 g39.1 ± 0.1 a25 ± 1 d24.1 ± 0.4 e36 ± 1 b
Anti-Inflammatory Potential (EC50 Values. µg/mL)
Nitric oxide (NO) production>400>400>400301 ± 7 a237 ± 7 b>400205 ± 7 c140 ± 5 d>400>400>400>400>400
Notes: EC50: Extract concentration corresponding to 50% of antioxidant activity or 0.5 of absorbance in reducing power assay. Trolox (positive control) EC50 values: 41 µg/mL (reducing power), 42 µg/mL (DPPH scavenging activity), 18 µg/mL (β-carotene bleaching inhibition) and 23 µg/mL (TBARS inhibition). Anti-inflammatory activity is expressed as EC50 values corresponding to 50% of inhibition of the NO production in comparison with the negative control (100% of NO production). Dexamethasone (positive control) EC50 values: 16 µg/mL. The results were analyzed using one-way analysis of variance (ANOVA) followed by Tukey’s HSD Test, and in each row different letters (a to i) mean significant differences (p < 0.05).
Table 4. Cytotoxic properties of L. pedunculata hydroethanolic and aqueous extracts (mean ± SD).
Table 4. Cytotoxic properties of L. pedunculata hydroethanolic and aqueous extracts (mean ± SD).
12345678910111213
Hydroethanolic Extracts
Tumor Cell Lines (GI50 Values, µg/mL)
MCF-761 ± 2 f57 ± 3 fg58 ± 2 fg115 ± 9 c82 ± 8 d53 ± 5 g70 ± 2 e61 ± 4 f60 ± 4 f53 ± 3 g61 ± 3 f236 ± 8 a212 ± 1 b
NCI-H460285 ± 22 bc275 ± 22 c277 ± 10 c>400>400340 ± 21 a>400>400>400>400297 ± 6 b119 ± 7 e241 ± 23 d
HeLa70 ± 5 de65.0 ± 0.2 fgh69 ± 2 def200 ± 14 c216 ± 7 b66.2 ± 0.2 efgh67 ± 2 efgh63.3 ± 0.1 gh73.1 ± 0.3 d62.2 ± 0.6 h65 ± 2 fgh68 ± 3 efg222 ± 6 a
HepG282 ± 4 e67 ± 1 f94 ± 6 d144 ± 10 c191 ± 16 b34 ± 3 g212 ± 19 a67 ± 10 f100 ± 3 d62 ± 5 f65 ± 1 f203 ± 14 a204 ± 4 a
Non-Tumor Cells (GI50 Values, µg/mL)
PLP2>400>400>400>400>400>400>400>400>400>400>400291 ± 11>400
Aqueous Extracts
Tumor Cell Lines (GI50 Values, µg/mL)
MCF-7>400270 ± 14 c267 ± 2 c287 ± 8 b267.7 ± 0.3 c222 ± 14 e223 ± 6 e185 ± 16 g150 ± 9 h290 ± 12 b256.2 ± 0.6 d270 ± 14 f342 ± 14 a
NCI-H460329 ± 15 c256 ± 5 e334 ± 15 c113 ± 9 i142 ± 12 h226 ± 14 f188 ± 12 g183 ± 11 g258 ± 14 e245 ± 13 e349 ± 5 b293 ± 12 d374 ± 7 a
HeLa>400>400310.2 ± 0.4 b343.92 ± 0.01 a253 ± 23 d262 ± 14 d199 ± 12 f159 ± 16 g286 ± 4 c195 ± 11 f224 ± 11 e82 ± 14 h298 ± 17 bc
HepG2315 ± 6 a254 ± 14 c251 ± 14 c293 ± 10 b>400211 ± 17 d194 ± 15 e179 ± 14 f118 ± 8 h256 ± 4 c191 ± 143 ef148 ± 1 g324 ± 6 a
Non-Tumor Cells (GI50 Values, µg/mL)
PLP2>400>400>400>400>400>400349 ± 12 b362 ± 21 a240 ± 16 c>400>400>400>400
MCF-7: breast carcinoma; NCI-H460: non-small lung cancer; HeLa: cervical carcinoma; HepG2: hepatocellular carcinoma; GI50 values—concentration that inhibited 50% of the net cell growth. Ellipticine (positive control) GI50 values: 1.21 µg/mL (MCF-7), 1.03 µg/mL (NCI-H460), 0.91 µg/mL (HeLa), 1.10 µg/mL (HepG2) and 2.29 µg/mL (PLP2). The results were analyzed using one-way analysis of variance (ANOVA) followed by Tukey’s HSD Test, and in each row different letters (a to h) mean significant differences (p < 0.05).
Table 5. Antibacterial (MIC and MBC, μg/mL) and antifungal (MIC and MFC, μg/mL) activities of L. pedunculata hydroethanolic and aqueous extracts.
Table 5. Antibacterial (MIC and MBC, μg/mL) and antifungal (MIC and MFC, μg/mL) activities of L. pedunculata hydroethanolic and aqueous extracts.
Antibacterial Activity
Hydroethanolic Extracts
12345678910111213SA
MICMICMICMICMICMICMICMICMICMICMICMICMICMICMIC
MBCMBCMBCMBCMBCMBCMBCMBCMBCMBCMBCMBCMBCMBCMBC
B.c.507575755075757575757575200100250
7515015015075150150150150150150150300200400
M.f.7510010010075100100100100100100300200200250
150150150150150150150150150150150450300300400
S.a.20403015010015015015020015020045020040250
407540300150300300300150300300600300100450
L.m.100150100150100150150150150150200450200200400
150300150300150300300300300300300600300300500
E.c.7510010010075100100100100100100200450200400
150150150150150150150150150150150300600300500
En.cl.40404050404010075757575200450200250
757575757575150150150150150300600300500
P.a.100150100150100150150150150150150300150200750
1503001503001503003003003003003004503003001200
S.t.150150150200100200200150200200200150150250400
300300300300150300300300300300300300300500750
Aqueous Extracts
12345678910111213SA
MICMICMICMICMICMICMICMICMICMICMICMICMICMICMIC
MBCMBCMBCMBCMBCMBCMBCMBCMBCMBCMBCMBCMBCMBCMBC
B.c.20030015030075200150200150150150300150100250
300450300450150300300300300300300450300200400
M.f.200300200300200300200200200150200450200200250
300450300450300450300300300300300600300300400
S.a.30030030030045030020020015030030030030040250
450450600450600450300300300450450600600100450
L.m.300300300300450300300300150300300300300200400
600600600600600450450450300450450600600300500
E.c.20020020020020020020010020045020045020020040
300300300300300300300150300600300900300300500
En.cl.150200200200200150200150150200150200150200250
300300300300300300300300300300300300300300500
P.a.300450300300300300300150150300300300300200750
4506006006006004506004503004506006006003001200
S.t.450450300300300300300300150300300300300250400
600600600600600600600450300450600600600500750
Antifungal Activity
Hydroethanolic Extracts
12345678910111213KB
MICMICMICMICMICMICMICMICMICMICMICMICMICMICMIC
MFCMFCMFCMFCMFCMFCMFCMFCMFCMFCMFCMFCMFCMFCMFC
A.fum.7515075751004015075100150150200200250150
15030015015015075300150150300300300300500200
A.v.40304040754040407515075150150200100
757515075150757575150300150300300500200
A.o.4040504075405040207540100200150150
75757575150757510040150753003002000200
A.n.507540751507515075150150200150200200150
7515075150300150300150300300300300300500200
T.v.2040203075303050155030751001000150
407540401504040752075401501501000200
P.f.757540752007575407575150200200200200
150150751503001515075150150300300300500250
P.o.40403040100404015040757530302500200
757540751507575200751501504504503500250
P.v.c.751505010020075150150150150150300300200100
15030075150300150300300300300300450450300200
Aqueous Extracts
12345678910111213KB
MICMICMICMICMICMICMICMICMICMICMICMICMICMICMIC
MFCMFCMFCMFCMFCMFCMFCMFCMFCMFCMFCMFCMFCMFCMFC
A.fum.300300600-600300--600600300300450250150
6006001200-1200600--9001200450600900500200
A.v.300300300600300300300300300200100150200200100
6006006001200600600600600600450200300300500200
A.o.600900---600300-2003001503002001500150
12001200---1200600-4506003004504502000200
A.n.-300---600--300-200450450200150
-600---1200--450-450900900500200
T.v.600300600600-600--1003002004504501000150
120060012001200-1200--3006004506006001000200
P.f.600300600-600-----450450450200200
12006001200-1200-----600900900500250
P.o.900450600-600---200-4504504502500200
12006001200-1200---450-6009009003.500.25
P.v.c.-600600-----200450450450450200100
-12001200-----450900600900900300200
Notes: MIC: Minimal Inhibitory Concentration; MBC: Minimal Bactericidal Concentration; MFC: Minimal Fungicide Concentration; S: Streptomycin; A: Ampicillin; K: Ketoconazole; B: Bifonazole; B.c.: Bacillus cereus; M.f.: Micrococcus flavus; S.a.: Staphylococcus aureus; L.m.: Listeria monocytogenes; E.c.: Escherichia coli; En. cl.: Enterobacter cloacae; P.a.: Pseudomonas aeruginosa; S.t.: Salmonella typhimirium; A.fum.: Aspergillus fumigatus; A.v.: Aspergillus versicolor; A.o.: Aspergillus ochraceus; A.n.: Aspergillus niger; T.v.: Trichoderma viride; P.f.: Penicillium funiculosum; P.o.: Penicillium ochrochloron; P.v.c.: Penicillium verrucosum var. cyclopium.
Table 6. Geographical information about the studied L. pedunculata samples.
Table 6. Geographical information about the studied L. pedunculata samples.
SamplesBPGV Accession NumberOriginSite Data Altitude (m)
110378Marvão. Portalegre333
209845Évora198
309838Vila Viçosa. Évora420
411290Bragança810
510372Arronches. Portalegre375
610400Portalegre375
710418Nisa. Portalegre291
810387Ponte de Sôr. Portalegre175
910391Évora155
1010412Castelo de Vide. Portalegre358
1110369Elvas. Portalegre267
1210379Castelo de Vide. Portalegre353
1311308Bragança667
More information in Lopes and Barata [9]; Accessions passport data documented and available at Grin-Global database (http://bpgv.iniav.pt).

Share and Cite

MDPI and ACS Style

Lopes, C.L.; Pereira, E.; Soković, M.; Carvalho, A.M.; Barata, A.M.; Lopes, V.; Rocha, F.; Calhelha, R.C.; Barros, L.; Ferreira, I.C.F.R. Phenolic Composition and Bioactivity of Lavandula pedunculata (Mill.) Cav. Samples from Different Geographical Origin. Molecules 2018, 23, 1037. https://doi.org/10.3390/molecules23051037

AMA Style

Lopes CL, Pereira E, Soković M, Carvalho AM, Barata AM, Lopes V, Rocha F, Calhelha RC, Barros L, Ferreira ICFR. Phenolic Composition and Bioactivity of Lavandula pedunculata (Mill.) Cav. Samples from Different Geographical Origin. Molecules. 2018; 23(5):1037. https://doi.org/10.3390/molecules23051037

Chicago/Turabian Style

Lopes, Catarina L., Eliana Pereira, Marina Soković, Ana Maria Carvalho, Ana Maria Barata, Violeta Lopes, Filomena Rocha, Ricardo C. Calhelha, Lillian Barros, and Isabel C.F.R. Ferreira. 2018. "Phenolic Composition and Bioactivity of Lavandula pedunculata (Mill.) Cav. Samples from Different Geographical Origin" Molecules 23, no. 5: 1037. https://doi.org/10.3390/molecules23051037

Article Metrics

Back to TopTop