Next Article in Journal
Advances in the Production of PBCA Microparticles Using a Micromixer with HH-Geometry in a Microfluidic System
Previous Article in Journal
Antibacterial and Wound Healing Activity In Vitro of Individual and Combined Extracts of Tagetes nelsonii Greenm, Agave americana and Aloe vera
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Phytochemical Profiling and Biological Activities of Two Helianthemum Species Growing in Greece

by
Evgenia Panou
1,
Konstantia Graikou
1,
Nikolaos Tsafantakis
1,
Fanourios-Nikolaos Sakellarakis
2 and
Ioanna Chinou
1,*
1
Laboratory of Pharmacognosy and Chemistry of Natural Products, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, 15771 Athens, Greece
2
Society for the Protection of Prespa, Agios Germanos, 53077 Florina, Greece
*
Author to whom correspondence should be addressed.
Sci. Pharm. 2024, 92(3), 42; https://doi.org/10.3390/scipharm92030042
Submission received: 20 May 2024 / Revised: 29 July 2024 / Accepted: 7 August 2024 / Published: 8 August 2024

Abstract

:
Helianthemum nummularium (HN) and Helianthemum oelanticum subsp. incanum (HO) are plant species, among Cistaceae, that are highly distributed in the Mediterranean region. In the current study, extracts of the aerial parts from both species have been analyzed phytochemically. The non-polar extract analysis resulted in the identification of 15 compounds in each species, mainly terpene and fatty acid derivatives, through GC–MS. The methanolic extract analysis, conducted through UHPLC–MS/MS, led to the identification of 39 metabolites in HN and 29 in HO, respectively, the majority of which were phenolics. Among the identified compounds, several have also been isolated and structurally determined (from HN: rutin, linoleic acid, gallic acid, and isoquercetin, and from HO: quercetin-3-O-(2″-O-galloyl)-galactopyranoside, methyl gallate, catechin-3-O-glucopyranoside, and astragalin, while hyperoside, and cis- and trans-tiliroside have been determined in both species). Furthermore, the methanolic extracts of HN and HO displayed a high total phenolic content (177.2 mg GA/g extract and 150.6 mg GA/g extract, respectively) and considerable free-radical scavenging activity against the DPPH radical (94.6% and 94.0% DPPH inhibition, respectively). Antimicrobial testing showed stronger inhibition of HN against Gram (+) bacterial strains (MIC values 0.07–0.15 mg/mL), while both extracts exhibited low tyrosinase-inhibitory activity. Considering the lack of studies conducted on the chemistry and biological activities of the genus Helianthemum, the chemical characterization of extracts could contribute to new sources of bioactive metabolites to be explored and exploited for further potential applications such as food and/ or the cosmetic industry.

1. Introduction

The genus Helianthemum (Cistaceae) is a monophyletic lineage with about 136 heliophytic species and subspecies of evergreen or semi-evergreen shrubs, subshrubs, and annual herbs [1,2]. It is the largest and most diverse genus in the Cistaceae family, thriving from sea level up to approximately 3000 m in various substrates, including limestone, dolomite, marl, gypsum, saline, and sandy soils [2]. This genus ranges from the Macaronesia region (North Atlantic archipelagos) to Central Asia, spanning the Mediterranean floristic region and Europe, with its highest diversity of species concentrated in the western Mediterranean area [2].
Helianthemum species have been traditionally used as natural remedies in folk medicine in Spain, Turkey, and America [3,4]. The aerial parts of different Helianthemum species have been used as anti-inflammatory, antiulcerogenic, antidiabetic, wound-healing, antiprotozoal, antimicrobial, analgesic, and psychiatric remedies [3,5,6]. In general, the plants of this genus are used in preparations of decoctions and teas for ingestion for gastrointestinal problems [3], while extracts are embodied into poultices and ointments for direct application on infected wounds or burns [7]. The conducted pharmacological studies have confirmed the benefits of the species regarding their traditional use and have enriched their biological activities known to date by also adding strong antioxidant, cytotoxic, and spasmolytic effects [3,8,9].
Regarding the phytochemistry of Helianthemum species, the majority of the literature data primarily focus on polar extract analysis, while limited studies have been conducted on essential oils [10,11]. The most characteristic chemical class of the genus is the phenolic derivatives including a variety of phenolic acids (derivatives of benzoic acid or hydroxycinnamic acid) [12,13,14], flavonoids [9,12,14,15], and polyphenols (hydrolyzed tannins and proanthocyanidins) [6,16], while to a lesser extent, lignans and coumarin derivatives have been reported [12,13]. In addition to these compounds, other chemical categories like aliphatic derivatives (aliphatic and fatty acids), sterols, and alkaloids are present in some species [12,13,16].
H. nummularium (L.) Mill (HN) is a species that consists of evergreen, dwarf shrubs that produce multiple racemes of yellow or pink flowers in early summer [2]. It has been traditionally used as part of the flower remedies, a popular form of complementary medicine often employed to address psychological issues and stress [12,17]. H. oelanticum subsp. incanum (synonym of H. canum) (HO) is a perennial dwarf shrub [2], with traditional use as an anti-inflammatory and wound-healing medicine, especially in the eastern Mediterranean region [18].
Both plants are widely distributed in Greece, where the Helianthemum genus is represented by 11 species [19]. Βoth studied species are native to Greece and are also encountered in the Prespa Lakes’ region, a protected National Park northwest of Greece with an exceptional richness and diversity of habitats, fauna, and flora unique to Greece and Europe [20].
According to our knowledge, no Helianthemum species growing in Greece have been previously studied. Therefore, the objective of this study was to conduct a comprehensive phytochemical analysis of the aerial parts of H. nummularium and H. oelanticum subsp. incanum, encompassing both non-polar and polar extract analysis, as well as the isolation and identification of their most abundant metabolites. Additionally, evaluating antioxidant, antimicrobial, and tyrosinase-inhibitory activities can provide insights into new sources of bioactive extracts.

2. Materials and Methods

2.1. Chemicals and Reagents

For the extraction, all solvents were purchased from Merck (Darmstadt, Germany) (c-hexane, dichloromethane, methanol). For the column chromatography (CC) stationary phases, silica gel (Kieselgel 60 H Merck) and microcrystalline cellulose (cellulose microcrystalline, Merck) were used, with gradient elution with the solvent mixtures indicated in each case. The solvents used were HPLC grade and were purchased from Fisher Chemical (Fisher Scientific, Loughborough, Leics, UK). For all column chromatographic procedures, fractionation was monitored via TLC: Merck silica gel 60 F254 (0.2 mm layer thickness), Merck RP-18 F254S, and Merck cellulose. For preparative thin-layer chromatography (prepTLC), 60 F254 (Merck) silica gel was used. Detection on TLC plates was enabled using UV light (254 and 366 nm), H2SO4–vanillin spray reagent on silica gel, followed by heating, and Naturstoff spray reagent on cellulose. For the biological assays, the chemicals and reagents were purchased from Merck (Darmstadt, Germany) (Folin–Ciocalteu reagent, gallic acid, sodium carbonate (Na2CO3)), Carlo Erba Reagents (Val-de-Reuil, France) (dimethyl sulfoxide (DMSO)), and Glentham Life Sciences (Corsham, UK) (2,2-diphenyl-1-picrylhydrazyl (DPPH•).

2.2. Plant Material

The crude plant materials (aerial parts of H. nummularium (HN) and H. oelanticum subsp. incanum (HO)) were collected from the Prespa Lake Park during the flowering period in 2021 (Table 1) and were botanically identified by F.N. Sakellarakis (Society for the Protection of Prespa, Agios Germanos, Greece). The samples were kept in a shaded and dry environment until fully dried, then ground using a laboratory mill and stored in darkness at room temperature.

2.3. Preparation of Extracts

The air-dried aerial parts of HN (52 g) and HO (66 g) were successively extracted with solvents of increasing polarity: cyclohexane (C-hex), dichloromethane (DCM), and methanol (MeOH) (3 times in each solvent with 500 mL) for 24 h at room temperature, receiving three extracts from each species (HN-C/HO-C cyclohexane extracts, HN-D/HO-D, dichloromethane extracts, HN-M/HO-M methanolic extracts). The extracts were evaporated under reduced pressure to dryness.

2.4. GC–MS Analysis

The chemical compositions of the cyclohexane and dichloromethane extracts were analyzed using GC–MS. Analysis was conducted using an Agilent Technologies Gas Chromatograph 7820A connected to an Agilent Technologies 5977B mass spectrometer system (Agilent, Santa Clara, CA, USA) with electron impact (EI) ionization at 70 eV. The gas chromatograph featured a split/splitless injector and an HP5MS capillary column (30 m, 0.25 mm internal diameter, and 0.25 µm film thickness). For cyclohexane extracts, the temperature program started at 60 °C for 5 min, increased at 3 °C/min to 280 °C, and held for 15 min, totaling 93 min. For dichloromethane extracts, the program started at 60 °C, increased at 3 °C/min to 300 °C, and held for 10 min, totaling 90 min. Helium was used as the carrier gas at a flow rate of 0.7 mL/min, with an injection volume of 2 µL, a split ratio of 1:10, and an injector temperature of 280 °C. Compounds were identified by comparing mass spectra with the Wiley Registry of Mass Spectral Data.

2.5. UHPLC–HRMS Analysis

Analyses of methanolic extracts were performed on a UHPLC–HRMS/MS Orbitrap Q-Exactive platform (Thermo Scientific, San Jose, CA, USA). A full scan in centroid mode with a mass range of 100–1200 Da was utilized. HRMS data at a resolution of 70,000 were collected in both negative and positive ionization modes under the following conditions: capillary temperature at 320 °C, spray voltage at 2.7 kV, S-lens RF level at 50 V, sheath gas flow at 40 arb units, auxiliary gas flow at 5 arb units, and auxiliary gas heater temperature at 50 °C. Separations were performed using a Hypersil Gold UPLC C18 reverse-phase column (2.1 × 100 mm, 1.9 µm; Thermo Fisher Scientific, San Jose, CA, USA). The mobile phase consisted of solvent A: ultra-pure H2O with 0.1% (v/v) FA, and solvent B: MeOH with 0.1% (v/v) FA. A gradient method with a total run time of 18 min was employed: 0–1 min, 5% B (isocratic); 1–8 min, 50–50% B (linear gradient); 8–13 min, 100% B (isocratic for column cleaning); 14–16 min, 95–5% B (linear gradient); and 16–18 min, 5% B (isocratic for column equilibration). The column temperature was maintained at 40 °C and the sample tray temperature at 10 °C, with a flow rate of 0.3 mL/min and an injection volume of 5 µL.

2.6. Fractionation and Purification Procedures of Methanolic Extracts

2.6.1. H. nummularium (HN-M)

HN-M (2.00 g) was subjected to column chromatography with microcrystalline cellulose and eluted with CH3COOH 3% up to 30% (gradient analysis); 219 fractions (HNA1–HNA219) were collected. Fractions HNA8–HNA14 (6.3 mg) were identified as quercetin-3-O-α-L-rhamnopyranosyl-(1→6)-β-D-glucopyranose (rutin) and fractions HNA15-HN27 (3.8 mg) identified as gallic acid, all of which were eluted with CH3COOH 3%. Fractions HNA214–HNA215 were eluted with CH3COOH 30% and were further extracted with ethyl acetate. From the ethyl acetate residue, linoleic acid (3.5 mg) was obtained. Furthermore, fractions HNA36–HNA148 (215 mg) were subjected to silica-60 gel column chromatography eluted with DCM: MeOH (98:2–80:20) (gradient method) to afford 176 fractions (HNB1–HNB176). Fractions HNB5-HNB45 (22.6 mg) were eluted with DCM: MeOH (95:5) and identified as the racemic mixture of cis- and trans-kaempferol-3-O-β-D-(6″-p-coumaroyl-glucopyranoside (cis/trans-tiliroside). Fractions HNB152–HNB159 (7.8 mg) were eluted with DCM: MeOH (90:10) and identified as the mixture of metabolites quercetin-3-O-β-D-glucopyranoside (isoquercetin)/quercetin-3-O-β-D-galactopyranoside (hyperoside).

2.6.2. H. oelanticum subsp. incanum (HO-M)

HO-M (1.15 g) was subjected to column chromatography with microcrystalline cellulose and eluted with CH3COOH 5% up to 30% (gradient analysis); 300 fractions (HOA1–HOA300) were collected. Fraction HOA39 (2.9 mg) was eluted with CH3COOH 5% and was identified as quercetin-3-O-(2″-O-galloyl)-β-D-galactopyranoside. Fractions HOA25-HOA38 (25.9 mg) were subjected to a preparative TLC silica plate (EtOAc (100):AcOH (11):FA (11):H2O (26)) and three bands were received. The first band was the racemic mixture of cis- and trans-kaempferol-3-O-β-D-(6″-p-coumaroyl-glucopyranoside (cis/trans-tiliroside) (7.6 mg), the second band was the metabolite kaempferol-3-O-β-D-glucopyranoside (astragalin) (4.2 mg) and the third band was identified as quercetin-3-O-β-D-galactopyranoside (hyperoside) (4.5 mg). Furthermore, fractions HOA8–HOA14 (225 mg) were subjected to silica-60 gel column chromatography eluted with CHCl3: MeOH (90:10-80:20) (gradient analysis) to afford 162 fractions (HOB1–HOB162). Fractions HOB4-HOMB5 (6.1 mg) were identified as methyl gallate and fractions HOB50–HOB61 (6.9 mg) were identified as catechin-3-O-β-D-glucopyranoside, all of which were eluted with CHCl3: MeOH (90:10).

2.7. Nuclear Magnetic Resonance (NMR)

One-dimensional (1H-NMR, 13C-NMR) and two-dimensional NMR spectra (COSY, HSQC, HMBC) were recorded on a Bruker Avance III 400 MHz (Bruker BioSpin, Rheinstetten, Germany) spectrometer using methanol-d4 (D024ES, Methanol-d4, Eurisotop, Chembiotin S.A., Voula, Greece) as the solvent. Chemical shifts are reported in ppm relative to the solvent in which the spectra were recorded [1H: δ (CD3OD) = 3.31 ppm; 13C:δ (CD3OD) = 49.15 ppm].

2.8. Total Phenolic Content (TPC)

The total phenolic content of the samples was determined by the Folin–Ciocalteu method [21]. A 96-well plate was used to dilute 25 µL of various concentrations (4, 2, 1 mg/mL) of extract or standard solutions of gallic acid (2.5, 5, 10, 12.5, 20, 25, 40, 50, 80, 100 µg/mL) in dimethylsulfoxide (DMSO). Both the extract and standard solutions were mixed with 125 µL of 10% Folin–Ciocalteu solution, followed by the addition of 100 µL of 7.5% sodium carbonate. The plate was incubated in darkness at room temperature for 30 min. Absorbance was measured at 765 nm using a TECAN Infinite m200 PRO multimode reader (Tecan Group, Männedorf, Switzerland). All measurements were performed in triplicate, and the mean values were plotted on a gallic acid calibration curve. The total phenolic content was expressed as mg gallic acid equivalent (GAE) per gram of dry extract.

2.9. DPPH (2,2-DiPhenyl-1-PicrylHydrazyl) Assay

The antioxidant activity of the samples was evaluated by DPPH (1,1-diphenyl-2-picrylhydrazyl) radical scavenging activity, according to the literature [21]. In a 96-well plate, 10 µL of various concentrations (4, 2, 1, 0.5, 0.25, 0.125, 0.0625 mg/mL) of extracts dissolved in DMSO were added to 190 µL of DPPH solution (12.4 mg/100 mL in ethanol). The plate was then incubated in darkness at room temperature for 30 min. Absorbance was measured at 517 nm using an Infinite M200 Pro TECAN photometer (Tecan Group, Männedorf, Switzerland). All evaluations were performed in triplicate, with gallic acid serving as the positive control (IC50 = 2.6 µg/mL). The percentage inhibition of the DPPH radical for each dilution was calculated using the following formula: % inhibition = [(A-B) − (C-D)]/(A-B) × 100, where A is the control (without sample), B is the blank (without sample, without DPPH), C is the sample, and D is the blank sample (without DPPH). Samples were evaluated in triplicate at final concentrations of 200, 100, 50, 25, 12.5, and 6.25 µg/mL, with results expressed as means ± standard deviation (n = 3).

2.10. In Vitro Tyrosinase Assay

The methanolic extracts were investigated for their ability to inhibit the oxidation of L-DOPA (L-3,4-dihydroxyphenylalanine) to dopaquinone and subsequently to dopachrome by the enzyme tyrosinase according to the literature [21]. The fractions were dissolved in DMSO (10 mg/mL) and then diluted in 1/15 M phosphate buffer (NaH2PO4/Na2HPO4), pH 6.8, ensuring that the final DMSO concentration in each well did not exceed 2%. In 96-well plates, 40 µL of sample in the phosphate buffer (1/15 M, pH 6.8) was mixed with 80 µL of the same buffer and 40 µL of mushroom tyrosinase (92 Units/mL) in the buffer. The mixture was incubated for 10 min at 25 °C, followed by the addition of 40 µL of 2.5 mM L-DOPA in the same buffer. The plates were then incubated for 5 min at 25 °C, and the absorbance of each well was measured at 475 nm. Extracts were tested in triplicate at a concentration of 300 µg/mL, with blank samples for each fraction also measured. Kojic acid was used as a positive control at a final concentration of 2 µg/mL, resulting in 52.84 ± 2.39% inhibition.

2.11. Antimicrobial Activity

A total of eleven microorganisms were assayed, including four Gram-positive bacteria: Staphylococcus aureus (ATCC 25923), Staphylococcus epidermidis (ATCC 12228), Streptococcus mutans (ATCC 31989), and Streptococcus viridans (ATCC 19952); four Gram-negative bacteria: Escherichia coli (ATCC 25922), Enterobacter cloacae (ATCC 13047), Klebsiella pneumoniae (ATCC 13883), and Pseudomonas aeruginosa (ATCC 227853); and three pathogenic fungi: Candida albicans (ATCC 10231), Candida tropicalis (ATCC 13801), and Candida glabrata (ATCC 28838). The antimicrobial activity was tested using the micro-dilution broth method to determine the minimal inhibitory concentration (MIC), following the guidelines of the European Committee on Antimicrobial Susceptibility Testing (EUCAST). Sterile 96-well microtiter plates were prepared by dispensing 100 µL per well of appropriately diluted extracts in broth medium, with two-fold dilutions to achieve final concentrations ranging from 0.50 to 10 mg/mL. Microbial inoculums, prepared in sterile 0.85% NaCl to match the turbidity of the 0.5 McFarland standard, were added to the wells to achieve final densities of 1.5 × 106 CFU/mL for bacteria and 5 × 104 CFU/mL for yeasts. The plates were then incubated at 37 °C for 24 h. MICs were determined visually as the lowest concentration of the extracts that completely inhibited the growth of the reference microbial strains. Each microplate included a DMSO control (at a final concentration of 10%), a positive control (inoculum without tested samples), and a negative control (tested samples without inoculum). Standard antibiotics, including netilmicin (at concentrations of 4–88 µg/mL) for bacteria and sanguinarine for oral bacteria, were used as controls. For fungi, 5-flucytosine (at concentrations of 0.5–25 µg/mL) and amphotericin B (at concentrations of 30, 15, and 10 µg/mL) were used as controls (Sanofi Paris, France, Diagnostics Pasteur). Pure solvent was also used as a blind control. All experiments were repeated three times, and results were expressed as mean values [22].

2.12. Statistical Analysis

All the experiments were conducted in triplicate and average values along with their standard deviations (SDs) were calculated using the Excel software package (Microsoft Office 2021).

3. Results

3.1. Identification of Metabolites from the Non-Polar Extracts (HN-C, HO-C, HN-D, and HO-D)

GC–MS analysis of HN-C, HO-C, HN-D, and HO-D is given in Table 2 and Table 3. Twelve and nine compounds were tentatively identified from the cyclohexane extracts as well as four and six compounds from the dichloromethane extracts.

3.2. Identification of Metabolites from Methanolic Extracts

Based on the chromatographic analysis of the methanolic extracts HN-M and HO-M, ten metabolites were isolated from the studied species through several chromatographic techniques and identified through NMR spectral analysis in comparison with the literature data: quercetin-3-O-α-L-rhamnopyranosyl-(1→6)-β-D-glucopyranose (rutin), gallic acid, linoleic acid, cis/trans-kaempferol-3-O-β-D-(6″-p-coumaroyl-glucopyranoside (cis/trans-tiliroside), quercetin-3-O-β-D-glucopyranoside (isoquercetin), quercetin-3-O-β-D-galactopyranoside (hyperoside), quercetin-3-O-(2″-O-galloyl)-β-D-galactopyranoside, kaempferol-3-O-β-D-glucopyranoside (astragalin), methyl gallate, and catechin-3-O-β-D-glucopyranoside. Furthermore, mass spectral analysis was performed to identify the bioactive compounds in both studied extracts (Figures S1–S4). Thirty-nine and twenty-nine compounds (Table 4) were tentatively identified from HN-M and HO-M extracts, respectively, using UHPLC–HRMS and comparing the mass spectral data with the literature.

3.3. Determination of Total Phenolic Content (TPC), DPPH Free Radical, and Tyrosinase-Inhibitory Activity

The total phenolic content was measured in the methanolic extracts HN-M and HO-M using the DPPH assay. Tyrosinase-inhibitory effects of methanolic extracts were tested according to the L-DOPA oxidation method. The results of the determination of the TPC as well as the DPPH free radical and tyrosinase-inhibition rates are presented in Table 5.

3.4. Antimicrobial Activity

The methanolic extracts were evaluated for their antimicrobial activity via the dilution method. The results, expressed as the minimum growth inhibitory concentrations (MICs) of the extracts and the reference antimicrobial agents, are shown in Table 6.

4. Discussion

Despite being the largest genus in the Cistaceae family, with an exceptionally diverse distribution and a history in folk medicine, only limited data concerning the phytochemical profile and biological activities of the Helianthemum species have been identified in the literature. We proceeded with an integrated phytochemical analysis of the species HN and HO, growing wild in the Prespa Lakes region, northwest of Greece, including non-polar and polar extracts of their aerial parts.
Cyclohexane (HN-C, HO-C) and dichloromethane extracts (HN-D, HO-D) were analyzed through GC–MS and the cyclohexane extracts showed the presence of monoterpenes, probably attributed to their volatile content received through the extraction procedure. In both species, the most notable monoterpenes were α-pinene, sabinene, myrcene, δ-3-carene, and limonene, with slight quantitative differences (Table 2). Specifically, in HΝ-C, α-pinene (35.77%), sabinene (12.05%), myrcene (2.70%), δ-3-carene (6.47%), and limonene (28.95%) were detected, while in HO-C, α-pinene (40.77%), sabinene (14.74%), myrcene (2.97%), δ-3-carene (7.18%), and limonene (28.93%) were detected. On the other hand, the volatile fraction of the dichloromethane extracts differed chemically (Table 3). In HN-D, the fatty acids palmitic and linoleic acid were detected, while in HO-D, hydrocarbons and palmitic acid were detected. In both species, the sterol derivatives β- and γ-sitosterol were identified.
Only a few studies have been conducted on the volatile compounds of Helianthemum species, and these concern essential oils from the aerial parts of H. kahiricum Del. from Iran [10] and H. canum (L.) Baumg. from Turkey [11]. The essential oil from H. kahiricum consisted mainly of the fatty acids like palmitic acid, myristic acid, lauric acid, and linoleic acid, as well as oxygenated monoterpenes and oxygenated sesquiterpenes [10]. The essential oil from H. canum appeared to contain myristicin (29.4%) as the dominant component [11], which was not detected in the studied species in the current study. Other reported minor compounds were saturated fatty acids (palmitic acid, myristic acid, lauric acid), hydrocarbons, sesquiterpenes, oxygenated sesquiterpenes, and monoterpenes similar to those found in the studied cyclohexane extracts (α-pinene, myrcene, limonene, 1,8-cineole, (E)-ocimene, camphor, bornyl acetate) [11].
The presence of sterols has been previously reported in the species H. sessiliflorum, presenting the isolation of β-sitosterol and daucosterol [13], and in H. ruficomum, referring to β-sitosterol and stigmasterol isolations [15].
Overall, our analysis of the chemical composition of the non-polar extracts HN-C, HO-C, HN-D, and HO-D have revealed the presence of monoterpenes, fatty acids, and hydrocarbons similar to those found in essential oils from the genus [10,11], as well as sterols that have been reported in other Helianthemum species [13,15].
According to the UHPLC–HRMS study of the methanolic extracts (HN-M, HO-M) (Table 4), the majority of the determined metabolites are new for the species to the best of our knowledge. There were significant similarities in both studied samples in terms of qualitative analysis. Thirty-nine and twenty-nine compounds were identified from HN-M and HO-M extracts, respectively, based to the comparison of mass spectral data with the literature. In both species, phenolic derivatives including phenolic acids, flavonoids, and polyphenols were abundant, while other chemical categories (such as fatty acids) were also detected.
Regarding phenolic acids, benzoic and hydroxycinnamic acid derivatives were present in both plants (4-hydroxybenzoic acid, dihydroxybenzoic acid isomers, gallic acid, methyl gallate, chlorogenic acid, 5-O-feruloylquinic acid, syringoyl dihexoside). Previous studies in various Helianthemum species including H. canum [4], H. nummularium [9], H. lippii [12,14], H. sessilifolium [13], H. ruficomum [15], and H. getulum [57], have also confirmed the existence of phenolic acids in their aerial parts. Dihydroxybenzoic acid hexoside and sinapic acid hexoside, most likely glycosylated derivatives, were detected in HN-M, marking the first report of their presence in the genus. Similarly, metabolites such as the galloylshikimic acid isomer and O-galloylquinic acid, identified in HO-M alongside caffeic acid derivatives, caffeic acid hexoside, and dicaffeoylglyceride, were detected in both studied plants and have also been reported for the first time in the genus. Among the detected phenolic acid derivatives, gallic acid and methyl gallate were isolated from HN-M and HO-M, respectively. Common phenolic acid derivatives from HN-M, HO-M, and previously studied Helianthemum spp. are presented in Table S1.
Flavonoids, especially flavonol derivatives, were detected in both species such as quercetin and kaempferol derivatives. Rutin (quercetin-3-O-α-L-rhamnopyranosyl-(1→6)-β-D-glucopyranose) was detected and isolated from HN-M, while it has been previously reported in the same species [17] as well as in H. lippii [12] and H. ruficomum [15]. Furthermore, isoquercetin (quercetin-3-β-D-glucopyranoside) was isolated from HN-M, previously reported in this species [9], as well as in H. sessilifolium [13] and H. lippii [14]. Hyperoside (quercetin-3-β-D-galactopyranoside) was isolated from both studied species, and it has been previously detected in H. nummularium [17], but it was isolated from HO for the first time.
Among the kaempferol derivatives, the racemic mixture of cis-/trans-tiliroside was isolated from both studied species. This metabolite is one of the most widespread in the genus, having been reported previously in many species such as H. nummularium [9,17], H. lippii [12], H. getulum [57], H. ruficomum [15], H. sessilifolium [13], and H. kahiricum [58]. Kaempferol coumaroyl rutinoside, identified in both studied species, and kaempferol rutinoside, detected in HN-M, appeared for the first time in the species.
Interestingly, galloyl-flavonol derivatives were detected in both plants for the first time in the genus. Specifically, quercetin galloylhexoside, and myricetin-O-galloylhexoside were identified in both species, while from HO-M, the quercetin galloylhexoside was isolated and identified as quercetin-3-O-(2″-O-galloyl)-galactopyranoside. In HO-M, kaempferol galloyl hexoside was also detected. Galloylflavonols have been previously reported in only two species in the whole Cistaceae family: Fumana montana Pomel. [59] and Cistus salvifolius [60].
The flavan derivative catechin-3-O-β-D-glucopyranoside was isolated from HO-M, not previously reported in the species, while in the Helianthemum genus, only catechin rutinoside has been detected so far in H. lippii [12]. In HN-M, epigallocatechin was detected with previous reference in H. lippii [12], H. getulum [57], and H. sessilifolium [13]. Common flavonoid derivatives from HN-M, HO-M, and previously studied Helianthemum spp. are presented in Table S2.
Hydrolyzed and condensed tannins were detected in both studied species. Galloyl-HHDP-hexoside, was found in HO-M, digalloyl-hexoside was detected in both species, and the pedunculagin isomer was found in HΝ-M. These polyphenols are reported for the first time in the genus. The flavan-3-ol dimers (epi)gallocatechin-O-galloyl(epi)gallocatechin and procyanidin dimer were also detected in HΝ-M. In the genus Helianthemum, the presence of polyphenols has been confirmed in H. helianthemoides [6], H. lippii [12], and H. ordosicum, from which gallocatechin-(4a → 8)-epigallocatechin was identified [16]. Ellagitannins and gallotannins have been reported in the genus Cistus, with galloyl-HHDP-hexoside and digalloyl-hexoside found in C. creticus L., and the compound pedunculagin in C. incanus L. [60].
Apart from the abovementioned phenolic derivatives, several fatty acid derivatives were detected in the studied species, including oxo-dihydroxy-octadecenoic acid, which is an oxidated oleic acid derivative; DGMG (digalactosylmonoacylglycerol) (18:3) galactolipid, which is a digalactosyl alpha-linolenic acid derivative; and lysophosphatidylinositol (LPI) 18:2, which belongs to the class of lysophospholipids. These compounds are most probably cellular components with either a structural role as components of cell membranes or organelles such as chloroplasts, or they may have a functional role in participating in the regulation of biological functions by transmitting or transferring signals through transcript G-protein-coupled receptors (GPCRs) [52,54,61]. To the best of our knowledge, there are no previous reports in the Cistaceae family regarding these compounds, while their presence has been reported in the fruits of Corylus avellana (Betulaceae) [54] and the leaves and fruits of Castanea sativa (Fagaceae) [52]. From this class of derivatives, linoleic acid was isolated from HN-M in the present study.
The last chemical class detected was hydroxycinnamate amides, with the detection of the metabolite N, N″-di-p-coumaroylspermidine in HN-M. Nitrogen-containing secondary metabolites are not prevalent in the genus Helianthemum, nor the wider family of Cistaceae. However, the presence of alkaloids has been reported in H. ordosicum, from which the alkaloid 7-phenyl-pyrazino[1,2-a] azepine was isolated [16]. In a recent study on the aerial parts of H. lippii, the metabolite spermatheridine was detected through GC–MS analysis [7], while the presence of alkaloids in H. lippii was additionally confirmed by detection testing in the Laib et al. study [14]. N,N″-di-p-coumaroylspermidine was detected in both the negative and positive ionization modes with parent ions at m/z 436 and 438, respectively, at a retention time of 19.15 min. (Figure S5). In the figure below (Figure 1), the chemical structure for this compound identified in HN-M, reported for the first time in Cistaceae family, is presented.
The antioxidant capacity of the methanolic extracts of the studied species has been determined. The extracts displayed high TPC values, with 177.2 mg GA/g extract for HN-M and 150.6 mg GA/g extract for HO-M. The DPPH assay revealed similarly high inhibition for both species, with maximum inhibition occurring at 0.2 mg/mL (94.6% for HN-M and 94.0% for HO-M). The IC50 was observed at 0.25 mg/mL for both species. In the Baldemir et al. study, the methanolic extract of H. canum (HO) displayed a higher total phenolic content with a TPC value of 284.13 mg GA/g extract, and the IC50 for the DPPH assay was 0.19 mg/mL [4]. Pirvu et al. estimated the scavenging effect of the 70% ethanolic extract of H. nummularium with the chemiluminescence assay and indicated IC50 was 1.27 μg/mL, using gallic acid (IC50 0.85 μg/mL) as a positive control [17].
Since Helianthemum species have been used traditionally for dermal applications, with no supportive literature referring to in vitro assays on skin disorders, we demonstrated a screening on the extracts’ inhibitory activity against tyrosinase due to the lack of such data and/or experiments on the genus Helianthemum.
The assayed extracts exhibited low tyrosinase-inhibitory activity with inhibition percentages of 42.7% for HN-M and 8.5% for HO-M at the concentration of 300 µg/mL. Comparing both results, the higher inhibition for HN-M could be attributed to the presence of rutin with known anti-tyrosinase activity, which was not identified in the HO-M extract [62].
The antimicrobial activity against free-living microbial strains is expressed in terms of MIC in mg/mL and is shown in Table 6. The methanolic extracts of both species noted stronger inhibition against the Gram-positive bacterial strains S. aureus, S. epidermidis, S. mutans, and S. viridans, while between the two species, HN-M showed lower MIC values (0.07 and 0.08 mg/mL against S. epidermidis and S. viridans, respectively). Moderate-to-weak activities were exhibited against the examined Candida spp. and Gram-negative bacterial strains P. aeruginosa, K. pneumoniae, E. cloacae, and E. coli (MIC range of 0.72–1.00 mg/mL). However, inhibition in fungal strains was slightly higher compared to that of Gram-negative bacteria.
According to the literature, quercetin glycosides (hyperoside, isoquercetin, rutin) and kaempferol derivatives (tiliroside, kaempferol rutinoside) combined with gallic and/or caffeic phenylcarboxylic acid derivates may contribute to the exerted microbial inhibition [17]. Both studied species have been previously evaluated in terms of their antimicrobial activities. Baldemir et al. reported, that the methanolic extract of H. canum (HO) showed strong inhibition against Bacillus sp. strains with an MIC value of 0.62 mg/mL. In contrast, the MIC values for E. coli, S. aureus, and P. aeruginosa were found to be comparatively higher (1.25 and <2.5 mg/mL) than the MICs for the same strains in our study [4]. In the study of Pirvu et al., the 70% ethanolic extract of HN was evaluated using the dilution method, exerting strong activity against S. aureus and E. coli, with inhibition zones of 15.5 and 21.5 mm, respectively [17].
The antimicrobial activity and the ability of phenolic compounds to scavenge free radicals is a property that makes these compounds extremely attractive. Phenolic extracts, including flavonol compounds such as glycosides of quercetin and kaempferol, and phenolic acids such as gallic acid and its derivatives have been shown to constitute interesting tools as natural preservatives, increasing the shelf life and stability of fresh food products [63,64]. Moreover, several reports indicate an antimicrobial effect of flavonols identified in the studied extracts including kaempferol-3-O-rutinoside (HO-M), rutin (HN-M), isoquercitrin (HN-M), and hyperoside (HN-M, HO-M) [17,63]. Dietary polyphenols have demonstrated the ability to modulate gut microbiota composition and function, as well as interfere with bacterial quorum sensing and exhibiting prebiotic-like activity [65]. Taken together, these data indicate extracts rich in these compounds can be a valuable source for functional food products, food supplements, and natural food preservatives.
The antioxidant and antimicrobial activities of phenolic compounds are also of cosmeceutical significance. The use of antioxidants in cosmetics helps to mitigate oxidative damage, offering an effective approach for preventing and treating premature aging, while the antimicrobial activity aims to minimize the degradation of a product caused by microorganisms [66]. The inhibition of the microbial proliferation of S. aureus, S. epidermis, and S. mutans, especially from HN-M, alongside its high antioxidant and moderate tyrosinase-inhibitory activity suggest some potential application in cosmetics.

5. Conclusions

The current study conducted an integrated phytochemical analysis of the aerial parts of the Helianthemum species HN and HO. The analysis included the identification of metabolites from the non-polar extracts (GC–MS) and the chemical profile from methanolic extracts through UHPLC–HRMS, while the latter were evaluated for their total phenolic content, and antioxidant, tyrosinase-inhibition, and antimicrobial activities. Both species have limited references towards their phytochemistry and biopotential, while they consist of a considerable source of natural remedies and are used traditionally against various health disorders. The current study is the first comprehensive analysis for Helianthemum species growing wild in Greece.
Both studied species seem to bear essential oil in their aerial parts, which is in accordance with a previous study on volatile compounds from HO. Specifically, HN-C appeared to have a characteristic odor, with no previous reports on its volatile compounds, making it an interesting candidate for such an analysis. The chemical profile from the methanolic extracts showed similarities with other species of the genus previously studied, with the most prominent chemical class being the phenolic derivatives, something that was also confirmed by the TPC determination. However, new derivatives from the species and the genus were identified, such as galloyl flavonol glycosides, flavonoid and catechin glucosides, caffeic acid derivatives, and polyphenols, while several compounds are reported for the first time in the family like the hydroxycinnamate amide, N,N”-di-p-coumaroylspermidine, and the fatty acid derivatives DGMG (18:3) galactolipid and lysophosphatidylinositol (LPI) 18:2. Although the compounds were tentatively identified by comparing the mass spectral data with the literature, the isolation of some of them (quercetin-3-O-(2″-O-galloyl)-galactopyranoside, catechin-3-O-β-D-glucopyranoside) further confirms their existence.
In our effort to broaden, the already tested biological properties, we proceeded to the evaluation of the tyrosinase-inhibitory activity. This is in addition to the antioxidant and antimicrobial activities, both of which showed promising results with high inhibition percentages for the free radical and low MIC values against Gram-positive microbial strains. Although the inhibition of tyrosinase was moderate for HN and low for HO, the noticeable difference between them indicates qualitative and/or quantitative differences that have a crucial impact on the ability to interact with the enzyme, representing a hypothesis that could be further studied.
Overall, the rich phenolic content of both species alongside their high antioxidant and antimicrobial activities indicates that species of this widespread genus can consist of new sources of bioactive compounds to be used as natural food preservatives or as part of functional food products, food supplements, and cosmeceutical products.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/scipharm92030042/s1, Figure S1: LC chromatogram (-cESI) of H. nummularium by UHPLC–HRMS analysis; Figure S2: LC chromatogram (+cESI) of H. nummularium by UHPLC–HRMS analysis; Figure S3: LC chromatogram (-cESI) of H. oelanticum subsp. incanum by UHPLC–HRMS analysis; Figure S4: LC chromatogram (+cESI) of H. oelanticum subsp. incanum by UHPLC–HRMS analysis. Figure S5. (a) ESI (-) and (b) ESI (+) mass spectrum of N, N″-di-p-coumaroylspermidine; Table S1. Phenolic acids of the genus Helianthemum according to the literature; Table S2. Flavonoids of genus Helianthemum according to the literature.

Author Contributions

Conceptualization, I.C.; methodology, E.P., K.G. and N.T.; formal analysis, E.P.; investigation, E.P. and N.T.; resources, F.-N.S.; data curation, E.P. and K.G.; writing—original draft preparation, E.P.; writing—review and editing, E.P., K.G., F.-N.S. and I.C.; supervision, I.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Society for the Protection of Prespa through PoliPrespa, project grant number SARG17131.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article and the Supplementary Materials.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AcOHAcetic acid
C-hexCyclohexane
CFUColony-forming unit
DCMDichloromethane
DMSODimethyl sulfoxide
DPPH2,2-diphenyl-1-picrylhydrazyl
EtOAcEthyl acetate
FAFormic acid
GAGallic acid
GAEGallic acid equivalent
GC–MSGas chromatography–mass spectrometry
HNH. nummularium
HN-CH. nummularium cyclohexane extract
HN-DH. nummularium dichloromethane extract
HN-MH. nummularium methanolic extract
HOH. oelanticum subsp. incanum
HO-CH. oelanticum subsp. incanum cyclohexane extract
HO-DH. oelanticum subsp. incanum dichloromethane extract
HO-MH. oelanticum subsp. incanum methanolic extract
MeOHMethanol
MICMinimum inhibitory concentration
UHPLC–MS/MSUltra-high-performance liquid chromatography–MS/MS

References

  1. Martín-Hernanz, S.; Martínez-Sánchez, S.; Albaladejo, R.G.; Lorite, J.; Arroyo, J.; Aparicio, A. Genetic diversity and differentiation in narrow versus widespread taxa of Helianthemum (Cistaceae) in a hotspot: The role of geographic range, habitat, and reproductive traits. Ecol. Evol. 2019, 9, 3016–3029. [Google Scholar] [CrossRef] [PubMed]
  2. Arrington, J.M.; Kubitzki, K. Cistaceae. En: The Families and Genera of Vascular Plants. In IV Flowering Plants Dicotyledons Malvales, Capparales and Non-Betalain Caryophyllales; Kubitzki, K., Ed.; Springer: Berlin/Heidelberg, Germany, 2003; pp. 62–70. [Google Scholar]
  3. Rubio-Moraga, Á.; Argandoña, J.; Mota, B.; Pérez, J.; Verde, A.; Fajardo, J.; Gómez-Navarro, J.; Castillo-López, R.; Ahrazem, O.; Gómez-Gómez, L. Screening for polyphenols, antioxidant and antimicrobial activities of extracts from eleven Helianthemum taxa (Cistaceae) used in folk medicine in south-eastern Spain. J. Ethnopharmacol. 2013, 148, 287–296. [Google Scholar] [CrossRef] [PubMed]
  4. Baldemir, A.; Gökşen, N.; Ildız, N.; Karatoprak, G.Ş.; Koşar, M. Phytochemical Profile and Biological Activities of Helianthemum canum l. Baumg. from Turkey. Chem. Biodivers. 2017, 14, e1700052. [Google Scholar] [CrossRef] [PubMed]
  5. Thaler, K.; Kaminski, A.; Chapman, A.; Langley, T.; Gartlehner, G. Bach flower remedies for psychological problems and pain: A systematic review. BMC Complement. Altern. Med. 2009, 9, 16. [Google Scholar] [CrossRef] [PubMed]
  6. Wafa, N.; Sofiane, G. In-vitro Antioxidant and anti-inflammatory activities valorisation of tannin crude extract of Helianthemum helianthemoïdes (Desf.) Grosser. J. Drug Deliv. Ther. 2020, 10, 135–139. [Google Scholar] [CrossRef]
  7. Alshammari, S.O.; Mahmoud, S.Y. Bioactive compounds of methanolic extract of Helianthemum lippii grows in Hafr Al-Batin region, northeastern Saudi Arabia. Acta Fytotech. Zootech. 2022, 25, 60–66. [Google Scholar]
  8. Djemam, N.; Lassed, S.; Gül, F.; Altun, M.; Monteiro, M.; Menezes-Pinto, D.; Benayache, S.; Benayache, F.; Zama, D.; Demirtas, I.; et al. Characterization of ethyl acetate and n-butanol extracts of Cymbopogon schoenanthus and Helianthemum lippii and their effect on the smooth muscle of the rat distal colon. J. Ethnopharmacol. 2020, 252, 112613. [Google Scholar] [CrossRef] [PubMed]
  9. Agostini, M.; Hininger-Favier, I.; Marcourt, L.; Boucherle, B.; Gao, B.; Hybertson, B.M.; Bose, S.K.; McCord, J.M.; Millery, A.; Rome, M.; et al. Phytochemical and Biological Investigation of Helianthemum nummularium, a High-Altitude Growing Alpine Plant Overrepresented in Ungulates Diets. Planta Med. 2020, 86, 1185–1190. [Google Scholar]
  10. Javidnia, K.; Nasiri, A.; Miri, R.; Jamalian, A. Composition of the Essential Oil of Helianthemum kahiricum Del. from Iran. J. Essent. Oil Res. 2007, 19, 52–53. [Google Scholar] [CrossRef]
  11. Gökşen, N.; Demirci, B.; Baldemir, A.; Koşar, M. Essential oil composition of Helianthemum canum (L.) Baumg. (Cistaceae) growing in Turkey. Asian Soc. Pharmacogn. 2017, 1, 5–10. [Google Scholar]
  12. Plescia, F.; Venturella, F.; D’Anneo, A.; Catania, V.; Gargano, M.L.; Polito, G.; Schillaci, D.; Palumbo Piccionello, A.; Lauricella, M.; Venturella, G.; et al. Phytochemical-rich extracts of Helianthemum lippii possess antimicrobial, anticancer, and anti-biofilm activities. Plant Biosyst. Int. J. Deal. All Asp. Plant Biol. 2022, 156, 1314–1324. [Google Scholar] [CrossRef]
  13. Benabdelaziz, I.; Haba, H.; Lavaud, C.; Harakat, D.; Benkhaled, M. Lignans and Other Constituents from Helianthemum sessiliflorum Pers. Rec. Nat. Prod. 2015, 9, 342–348. [Google Scholar]
  14. Laib, I.; Djahra, A.B. Phytochemical investigation of Helianthemum lippii l. aerial Dum.Cours part and evaluation for its antioxidant activities. Int. J. Sec. Metabol. 2022, 9, 229–237. [Google Scholar] [CrossRef]
  15. Chemam, Y.; Benayache, S.; Marchioni, E.; Zhao, M.; Mosset, P.; Benayache, F.; McPhee, D.J. On-line screening, isolation and identification of antioxidant compounds of Helianthemum ruficomum. Molecules 2017, 22, 239. [Google Scholar] [CrossRef]
  16. Wang, Q.; Wu, R.; Wu, J.; Dai, N.; Han, N. A New Alkaloid from Helianthemum Ordosicum: A New Alkaloid from Helianthemum Ordosicum. Magn. Reson. Chem. 2015, 53, 314–316. [Google Scholar] [CrossRef]
  17. Pirvu, L.; Nicu, I. Polyphenols Content, Antioxidant and Antimicrobial Activity of Ethanol Extracts from the Aerial Part of Rock Rose (Helianthemum nummularium) Species. J. Agric. Sci. Technol. A 2017, 7, 61–67. [Google Scholar]
  18. Küpeli Akkol, E.; Kosar, M.; Baldemir, A.; Şeker Karatoprak, G.; Demirpolat, E.; Betul Yerer Aycan, M.; Süntar, I.; Ilgün, S. The Wound-Healing Potential of the Endemic Plant Helianthemum canum (L.) Baumg: Preclinical Studies Supported with Phytochemical Profiling. Chem. Biodivers. 2023, 20, e202301529. [Google Scholar] [CrossRef]
  19. Dimopoulos, P.; Raus, T.; Bergmeier, E.; Constantinidis, T.; Iatrou, G.; Kokkini, S.; Strid, A.; Tzanoudakis, D. Vascular Plants of Greece: An Annotated Checklist; Botanic Garden and Botanical Museum Berlin-Dahlem: Berlin, Germany, 2013; Volume 31, pp. 1–371. [Google Scholar]
  20. Strid, A.; Bergmeier, E.; Fotiadis, G. Flora and Vegetation of the Prespa National Park, Greece; Bilingual Edition: Greek—English; Aage, V., Ed.; Jensen Charity Foundation, by the Prespa Preservation Society and Vlassi Bros Publications: Athens, Greece, 2020. [Google Scholar]
  21. Klontza, V.; Graikou, K.; Cheilari, A.; Kasapis, V.; Ganos, C.; Aligiannis, N.; Chinou, I. Phytochemical Study on Seeds of Paeonia clusii subsp. rhodia—Antioxidant and Anti-Tyrosinase Properties. Int. J. Mol. Sci. 2023, 24, 4935. [Google Scholar] [CrossRef] [PubMed]
  22. Pyrgioti, E.; Graikou, K.; Aligiannis, N.; Karabournioti, S.; Chinou, I. Qualitative analysis related to palynological characterization and biological evaluation of propolis from Prespa National Park (Greece). Molecules 2022, 27, 7018. [Google Scholar] [CrossRef]
  23. Yang, W.; Min, Y.; Man, L.; Dezhi, K.; Rui, S.; Xiaowei, S.; Kerong, Z.; Qiao, W.; Zhang, L. A Practical Strategy for the Characterization of Coumarins in Radix Glehniae by Liquid Chromatography Coupled with Triple Quadrupole-Linear Ion Trap Mass Spectrometry. J. Chromatogr. 2010, 1217, 4587–4600. [Google Scholar] [CrossRef]
  24. Gouveia, S.; Castilho, P.C. Helichrysum monizii Lowe: Phenolic Composition and Antioxidant Potential: H. monizii Phenolic Composition and Antioxidant Potential. Phytochem. Anal. 2012, 23, 72–83. [Google Scholar] [CrossRef] [PubMed]
  25. Hooi Poay, T.; Sui Kiong, L.; Cheng Hock, C. Characterization of galloylated cyanogenic glucosides and hydrolysable tannins from leaves of Phyllagathis rotundifolia by LC-ESI-MS/MS. Phytochem. Anal. 2011, 22, 516–525. [Google Scholar] [CrossRef] [PubMed]
  26. Bujor, O.C.; Le Bourvellec, C.; Volf, I.; Popa, V.I.; Dufour, C. Seasonal Variations of the Phenolic Constituents in Bilberry (Vaccinium myrtillus L.) Leaves, Stems and Fruits, and Their Antioxidant Activity. Food Chem. 2016, 213, 58–68. [Google Scholar] [CrossRef] [PubMed]
  27. Maggi, F.; Lucarini, D.; Papa, F.; Peron, G.; Dall'Acqua, S. Phytochemical Analysis of the Labdanum-Poor Cistus Creticus subsp. eriocephalus (Viv.) Greuter et Burdet Growing in Central Italy. Biochem. Syst. Ecol. 2016, 66, 50–57. [Google Scholar] [CrossRef]
  28. Gruz, J.; Novák, O.; Strnad, M. Rapid analysis of phenolic acids in beverages by UPLC–MS/MS. Food Chem. 2008, 111, 789–794. [Google Scholar] [CrossRef]
  29. Singh, A.; Bajpai, V.; Kumar, S.; Sharma, K.R.; Kumar, B. Profiling of Gallic and Ellagic Acid Derivatives in Different Plant Parts of Terminalia arjuna by HPLC-ESI-QTOF-MS/MS. Nat. Prod. Commun. 2016, 11, 239–244. [Google Scholar] [CrossRef] [PubMed]
  30. Chan, C.L.; Gan, R.Y.; Shah, N.P.; Corke, H. Enhancing Antioxidant Capacity of Lactobacillus acidophilus—Fermented Milk Fortified with Pomegranate Peel Extracts. Food Biosci. 2018, 26, 185–192. [Google Scholar] [CrossRef]
  31. Boulekbache-Makhlouf, L.; Meudec, E.; Mazauric, J.P.; Madani, K.; Cheynier, V. Qualitative and Semi-Quantitative Analysis of Phenolics in Eucalyptus globulus Leaves by High-Performance Liquid Chromatography Coupled with Diode Array Detection and Electrospray Ionisation Mass Spectrometry: HPLC-DAD-ESI/MS Analysis of Phenolics of Eucalyptus globulus Leaves. Phytochem. Anal. 2013, 24, 162–170. [Google Scholar] [PubMed]
  32. Yeo, J.; Shahidi, F. Identification and quantification of soluble and insoluble-bound phenolics in Lentil hulls using HPLC-ESI-MS/MS and their antioxidant potential. Food Chem. 2020, 315, 126202. [Google Scholar] [CrossRef]
  33. Jaiswal, R.; Sovdat, T.; Vivan, F.; Kuhnert, N. Profiling and characterization by LC-MS n of the chlorogenic acids and hydroxycinnamoyl shikimate esters in mate (Ilex paraguariensis). J. Agric. Food Chem. 2010, 58, 5471–5484. [Google Scholar] [CrossRef]
  34. Finimundy, T.C.; Pereira, C.; Dias, M.I.; Caleja, C.; Calhelha, R.C.; Sokovic, M.; Stojković, D.; Carvalho, A.M.; Rosa, E.; Barros, L.; et al. Infusions of Herbal Blends as Promising Sources of Phenolic Compounds and Bioactive Properties. Molecules 2020, 25, 2151. [Google Scholar] [CrossRef] [PubMed]
  35. Ma, C.; Xiao, S.Y.; Li, Z.G.; Wang, W.; Du, L.J. Characterization of Active Phenolic Components in the Ethanolic Extract of Ananas Comosus, L. Leaves Using High-Performance Liquid Chromatography with Diode Array Detection and Tandem Mass Spectrometry. J. Chromatogr. 2007, 1165, 39–44. [Google Scholar] [CrossRef] [PubMed]
  36. Steingass, C.B.; Glock, M.P.; Schweiggert, R.M.; Carle, R. Studies into the Phenolic Patterns of Different Tissues of Pineapple (Ananas comosus [L.] Merr.) Infructescence by HPLC-DAD-ESI-MS n and GC-MS Analysis. Anal. Bioanal. Chem. 2015, 407, 6463–6479. [Google Scholar] [CrossRef]
  37. Mastino, P.M.; Marchetti, M.; Costa, J.; Juliano, C.; Usai, M. Analytical Profiling of Phenolic Compounds in Extracts of Three Cistus Species from Sardinia and Their Potential Antimicrobial and Antioxidant Activity. Chem. Biodivers. 2021, 18, e2100053. [Google Scholar] [CrossRef]
  38. Tittikpina, N.K.; Katragunta, K.; Avula, B.; Ali, Z.; Khan, I.A. Strategy for the Quality Control of Herbal Preparations Made of Sarcocephalus latifolius: Development and Validation of a UHPLC-PDA Method for Quantification of Angustoline and Strictosamide and Chemical Profiling Using LC-QTOF. Phytochem. Anal. 2023, 34, 105–126. [Google Scholar] [CrossRef]
  39. Xiang, J.; Zhang, M.; Apea-Bah, F.B.; Beta, T. Hydroxycinnamic Acid Amide (HCAA) Derivatives, Flavonoid C-Glycosides, Phenolic Acids and Antioxidant Properties of Foxtail Millet. Food Chem. 2019, 295, 214–223. [Google Scholar] [CrossRef]
  40. Wang, R.D.; Su, G.H.; Wang, L.; Xia, Q.; Liu, R.; Lu, Q.; Zhang, J.L. Identification and Mechanism of Effective Components from Rape (Brassica napus L.) Bee Pollen on Serum Uric Acid Level and Xanthine Oxidase Activity. J. Funct. Foods 2018, 47, 241–251. [Google Scholar] [CrossRef]
  41. Zhang, H.; Liu, R.; Lu, Q. Separation and Characterization of Phenolamines and Flavonoids from Rape Bee Pollen, and Comparison of Their Antioxidant Activities and Protective Effects Against Oxidative Stress. Molecules 2020, 25, 1264. [Google Scholar] [CrossRef]
  42. Vukics, V.; Guttman, A. Structural Characterization of Flavonoid Glycosides by Multi-Stage Mass Spectrometry: MS Characterization of Flavonoid Glycosides. Mass Spectrom. Rev. 2010, 29, 1–16. [Google Scholar] [CrossRef]
  43. Guimarães, R.; Barros, L.; Dueñas, M.; Carvalho, A.M.; Queiroz, M.J.R.; Santos-Buelga, C.; Ferreira, I.C. Characterization of phenolic compounds in wild fruits from Northeastern Portugal. Food Chem. 2013, 141, 3721–3730. [Google Scholar] [CrossRef]
  44. Fougère, L.; Da Silva, D.; Destandau, E.; Elfakir, C. TLC-MALDI-TOF-MS-Based Identification of Flavonoid Compounds Using an Inorganic Matrix. Phytochem. Anal. 2019, 30, 218–225. [Google Scholar] [CrossRef]
  45. Sobral, F.; Calhelha, R.C.; Barros, L.; Dueñas, M.; Tomás, A.; Santos-Buelga, C.; Vilas-Boas, M.; Ferreira, I.C. Flavonoid Composition and Antitumor Activity of Bee Bread Collected in Northeast Portugal. Molecules 2017, 22, 248. [Google Scholar] [CrossRef]
  46. Bunse, M.; Lorenz, P.; Stintzing, F.C.; Kammerer, D.R. Characterization of Secondary Metabolites in Flowers of Sanguisorba officinalis L. by HPLC-DAD-MSn and GC/MS. Chem. Biodivers. 2020, 17, e1900724. [Google Scholar] [CrossRef]
  47. Karapandzova, M.; Stefkov, G.; Cvetkovikj, I.; Stanoeva, J.P.; Stefova, M.; Kulevanova, S. Flavonoids and Other Phenolic Compounds in Needles of Pinus peuce and Other Pine Species from the Macedonian Flora. Nat. Prod. Commun. 2015, 10, 1934578X1501000. [Google Scholar]
  48. Llorent-Martínez, E.J.; Spínolaa, V.; Gouveiac, S.; Castilho, P.C. HPLC-ESI-MSn characterization of phenolic compounds, terpenoid saponins, and other minor compounds in Bituminaria bituminosa. Ind. Crop. Prod. 2015, 69, 80–90. [Google Scholar] [CrossRef]
  49. Farid, M.M.; Marzouk, M.M.; Hussein, S.R.; Elkhateeb, A.; Abdel-Hameed, E.S. Comparative Study of Posidonia oceanica L.: LC/ESI/MS Analysis, Cytotoxic Activity and Chemosystematic Significance. J. Mater. Environ. Sci. 2018, 9, 1676–1682. [Google Scholar]
  50. Simirgiotis, M.J.; Schmeda-Hirschmann, G. Direct identification of phenolic constituents in Boldo Folium (Peumus boldus Mol.) infusions by high-performance liquid chromatography with diode array detection and electrospray ionization tandem mass spectrometry. J. Chromatogr. A 2010, 1217, 443–449. [Google Scholar] [CrossRef]
  51. Masullo, M.; Cerulli, A.; Pizza, C.; Piacente, S. Pouteria Lucuma Pulp and Skin: In Depth Chemical Profile and Evaluation of Antioxidant Activity. Molecules 2021, 26, 5236. [Google Scholar] [CrossRef]
  52. Cerulli, A.; Napolitano, A.; Hošek, J.; Masullo, M.; Pizza, C.; Piacente, S. Antioxidant and in vitro preliminary anti-inflammatory activity of Castanea sativa (Italian Cultivar “Marrone di Roccadaspide” PGI) burs, leaves, and chestnuts extracts and their metabolite profiles by LC-ESI/LTQ Orbitrap/MS/MS. Antioxidants 2021, 10, 278. [Google Scholar] [CrossRef]
  53. Rush, M.D.; Rue, E.A.; Wong, A.; Kowalski, P.; Glinski, J.A.; van Breemen, R.B. Rapid Determination of Procyanidins Using MALDI-TOF/TOF Mass Spectrometry. J. Agric. Food Chem. 2018, 66, 11355–11361. [Google Scholar] [CrossRef]
  54. Napolitano, A.; Cerulli, A.; Pizza, C.; Piacente, S. Multiclass polar lipid profiling in fresh and roasted hazelnut (Corylus avellana cultivar “Tonda di Giffoni”) by LC-ESI/LTQ Orbitrap/MS/MSn. Food Chem. 2018, 269, 125–135. [Google Scholar] [CrossRef]
  55. Hsu, F.F.; Turk, J. Electrospray Ionization Multiple-Stage Linear Ion-trap Mass Spectrometry for Structural Elucidation of Triacylglycerols: Assignment of Fatty Acyl Groups on the Glycerol Backbone and Location of Double Bonds. J. Am. Soc. Mass Spectrom. 2010, 21, 657–669. [Google Scholar] [CrossRef] [PubMed]
  56. Wang, W.; Yang, R.; Chen, H.; Wu, Y.; Lin, S.; Yuan, S.; Pan, W.; Jia, A.Q. Chemical Constituents of Smilax riparia and their Tumoral Cytotoxicities. Chem. Nat. Compd. 2020, 56, 228–234. [Google Scholar] [CrossRef]
  57. Terfassi, S.; Dauvergne, X.; Cérantola, S.; Lemoine, C.; Bensouici, C.; Fadila, B.; Christian, M.; Marchioni, E.; Benayache, S. First report on phytochemical investigation, antioxidant and antidiabetic activities of Helianthemum getulum. Nat. Prod. Res. 2022, 36, 2806–2813. [Google Scholar] [CrossRef] [PubMed]
  58. Bouzergoune, F.; Bitam, F.; Aberkane, M.C.; Mosset, P.; Fetha, M.N.H.; Boudjar, H.; Aberkane, A. Preliminary Phytochemical and Antimicrobial Activity Investigations on the Aerial Parts of Helianthemum kahiricum. Chem. Nat. Compd. 2013, 49, 751–752. [Google Scholar] [CrossRef]
  59. Laraoui, H.; Long, C.; Haba, H.; Benkhaled, M. New methylated flavonol glucosides from Fumana montana Pomel. Nat. Prod. Res. 2013, 27, 770–775. [Google Scholar] [CrossRef]
  60. Lukas, B.; Bragagna, L.; Starzyk, K.; Labedz, K.; Stolze, K.; Novak, J. Polyphenol Diversity and Antioxidant Activity of European Cistus creticus L. (Cistaceae) Compared to Six Further, Partly Sympatric Cistus Species. Plants 2021, 10, 615. [Google Scholar] [CrossRef]
  61. Kihara, Y.; Mizuno, H.; Chun, J. Lysophospholipid receptors in drug discovery. Exp. Cell Res. 2015, 333, 171–177. [Google Scholar] [CrossRef] [PubMed]
  62. Si, Y.-X.; Liu, B.; Lu, J.-Y.; Liu, S.-S.; Ma, Q.-M.; Su, Z.-X. An integrated study of tyrosinase inhibition by rutin: Progress using a computational simulation. J. Biomol. Struct. Dyn. 2012, 29, 999–1012. [Google Scholar] [CrossRef]
  63. Gervasi, T.; Calderaro, A.; Barreca, D.; Tellone, E.; Trombetta, D.; Ficarra, S.; Smeriglio, A.; Mandalari, G.; Gattuso, G. Biotechnological Applications and Health-Promoting Properties of Flavonols: An Updated View. Int. J. Mol. Sci. 2022, 23, 1710. [Google Scholar] [CrossRef]
  64. Choubey, S.; Varughese, L.R.; Kumar, V.; Beniwal, V. Medicinal Importance of Gallic Acid and Its Ester Derivatives: A Patent Review. Pharm. Pat. Anal. 2015, 4, 305–315. [Google Scholar] [CrossRef] [PubMed]
  65. Kumar Singh, A.; Cabral, C.; Kumar, R.; Ganguly, R.; Rana, H.K.; Gupta, A.; Lauro, M.R.; Carbone, C.; Reis, F.; Pandey, A.K. Beneficial Effects of Dietary Polyphenols on Gut Microbiota and Strategies to Improve Delivery Efficiency. Nutrients 2019, 11, 2216. [Google Scholar] [CrossRef] [PubMed]
  66. De Lima Cherubim, D.J.; Buzanello Martins, C.V.; Fariña, L.O.; Da Silva De Lucca, R.A. Polyphenols as Natural Antioxidants in Cosmetics Applications. J. Cosmet. Dermatol. 2020, 19, 33–37. [Google Scholar] [CrossRef] [PubMed]
Figure 1. N,N″-di-p-coumaroylspermidine identified by UHPLC–MS/MS.
Figure 1. N,N″-di-p-coumaroylspermidine identified by UHPLC–MS/MS.
Scipharm 92 00042 g001
Table 1. Collection data of the two studied species H. nummularium and H. oelanticum subsp. incanum.
Table 1. Collection data of the two studied species H. nummularium and H. oelanticum subsp. incanum.
SpeciesAbbreviationDateLocationElevation (m)
Helianthemum nummularium (L.) Mill.HN05/2021Mt. Devas, woodland
Quercus macedonica, Juniperus excelsa and Carpinus sp.,
Prespa National Park, NW Greece
1067
Helianthemum oelandicum (L.) DC. in Lam. and DC. subsp. incanum (L.) BonnierHO1065
Table 2. Volatile metabolites of the extracts HN-C and HO-C using GC–MS.
Table 2. Volatile metabolites of the extracts HN-C and HO-C using GC–MS.
CompoundArea (%)
HN-CHO-C
α-Thujene0.63-
α-Pinene35.7740.77
Sabinene12.0514.74
β-Pinene1.03-
Myrcene2.702.97
δ-3-Carene6.477.18
p-Cymene1.451.04
Limonene28.9528.93
γ-Terpinene1.251.11
Terpinolene1.431.25
Thujone isomer<0.1<0.1
Palmitic acid methyl ester0.76-
Table 3. Volatile metabolites of the extracts HN-D and HO-D using GC–MS.
Table 3. Volatile metabolites of the extracts HN-D and HO-D using GC–MS.
CompoundArea (%)
HN-DHO-D
1-Nonadecene-7.74
Octadecane-5.63
Eicosane-19.46
Hexadecanoic acid (Palmitic acid)1.351.42
9,12-Octadecenoic acid (Linoleic acid)1.14-
γ-Sitosterol1.111.19
β-Sitosterol0.830.90
Table 4. Metabolites of the methanolic extracts of HN-M and HO-M using UHPLC–HRMS in negative and positive modes.
Table 4. Metabolites of the methanolic extracts of HN-M and HO-M using UHPLC–HRMS in negative and positive modes.
Rt (min)Adduct IonsObserved m/zMS/MSMassMolecular FormulaIdentified CompoundsDetermined by
HN-MHO-M
3.543.51[M − H]169169, 125170C7H6O5Gallic acid[12], NMR
4.02 [M + H]+365203, 185364C17H16O9Xanthotoxol glucopyranoside[12,23]
4.054.03[(M − H) + HCO2H]387341, 179, 119, 101, 89342C15H18O9Caffeic acid hexoside[24]
4.29 [M − H]312312, 184, 183313 Methylgallate derivative[25]
14.16 [M − H]315179, 153, 152, 108316C13H16O9Dihydroxybenzoic acid hexoside[26]
14.44[M − H]343191, 169344C14H16O10O-galloylquinic acid[27]
14.60[M − H]153153, 109154C7H6O4Dihydroxybenzoic acid[28]
14.8814.88[M − H]183 184C8H8O5Methyl gallate[12], NMR
14.93 [M − H]455409, 325, 307, 265, 205, 163, 456 p-Coumaric acid derivative[26]
15.38 [M − H]305179,174, 139, 137, 125306C15H14O7(Epi)gallocatechin[27]
15.47 [M + H]+355 354C16H18O9Chlorogenic acid[12]
15.90 [M − H]153153, 135, 109, 65154C7H6O4Dihydroxybenzoic acid[28]
15.95[M − H]325169, 125326C14H14O9Galloylshikimic acid isomer[29]
16.12 [M − H]783783, 765, 301, 275, 247, 169784C34H24O22Pedunculagin isomer
(Ellagitannin)
[30,31]
16.28[M − H]451451, 329, 313, 289, 271452C21H24O11Catechin-3-O-glucopyranoside[32], NMR
16.6616.64[M − H]483 484C20H20O14Digalloyl-hexoside[29]
17.0517.05[M − H]431385, 223, 205, 161, 153, 432C17H22O10Sinapic acid hexoside[26]
17.16 [M − H]367367, 193, 134368C17H20O95-O-Feruloyl-quinic acid[33]
17.35 [M − H]337191, 163, 119338C16H18O8Coumaroyl-quinic acid[34]
17.57[M − H]633633, 463, 301, 275, 257, 245634C27H22O18Galloyl-HHDP-hexoside[30,31]
17.6417.64[(M − H) + HCO2H]461415, 269. 161, 101416C21H20O9Dicaffeoyl glycerol[35]
18.47[(M − H) +HCO2H]567567, 521, 359, 341, 329, 179, 522C21H30O15Syringyl dihexoside[36]
18.5218.52[M − H]631631, 479, 317, 179632C28H24O17Myricetin-O-galloyl-hexoside[37]
18.67 [M − H]463463, 301, 300, 271, 255464C21H20O12Isoquercetin[38], NMR
18.68 [M − H]609301, 300, 271610C27H30O16Rutin[38], NMR
18.82 [M − H]761609, 305762C37H30O18(Epi)gallocatechin-O-galloyl(epi)gallocatechin[33]
19.15 [M − H]436436, 316, 273, 145, 119437C25H31N3O4N, N″-di-p-coumaroylspermidine[39,40,41]
19.23 [M − H]593593, 571, 447, 384, 327, 285, 594C27H30O15Kaempferol rutinoside[12,42]
19.2819.31[M − H]463463, 301, 300, 271, 255464C21H20O12Hyperoside [38], NMR
19.31 [M − H]615463, 301, 300, 271, 255, 169616C28H24O16Quercetin galloylhexoside[43]
19.33[M − H]615463, 301, 300, 271, 255, 243616C28H24O16Quercetin-3-O-(2″-O-galloyl)-galactopyranoside[43], NMR
19.76[M − H]447285, 284, 255, 227448 Kaempferol-3-O-glucopyranoside[38], NMR
19.80[M − H]599517, 447, 429, 415, 301, 285600C28H24O15Kaempferol galloyl hexoside[44]
19.8519.85[M − H]137137, 93138C7H6O34-Hydroxybenzoic acid[28]
19.9419.90[M − H]447315, 299, 284, 255, 227448C21H20O11Isorhamnetin-pentoside[45]
20.25[M − H]625463, 301626C27H30O17Quercetin dihexoside[46]
20.40[M − H]477301478C21H18O13Quercetin glucuronide[46]
21.22 [(M − H) + HCO2H]493447, 399, 315, 161448C21H20O11Isorhamnetin pentoside isomer[47]
21.43 [M − H]419153, 152, 108, 109420 Procatechuic acid derivative[24]
21.9721.95[M − H]593593, 447, 307, 285, 255, 145, 594C30H26O13cis/trans-Tiliroside[12,47], NMR
22.2422.24[M − H]593 594C30H26O13cis/trans-Tiliroside
23.3923.41[M − H]327327, 291, 239, 229, 211328C18H32O5Oxo-dihydroxy-octadecenoic acid[48]
24.1224.11[M − H]329329, 229, 211330 Linoleic acid derivative[49]
25.3625.40[M − H]739739, 593, 453, 285, 255, 227, 740C36H36O17Kaempferol coumaroyl rutinoside[47]
25.63[M − H]739593, 285, 284, 255, 227, 145740C36H36O17Kaempferol coumaroyl rutinoside isomer[47]
26.60 [M − H]593593, 315, 277, 241, 152594C27H30O15Isorhamnetin deoxyhexosyl-pentoside[50]
27.7127.70[(M − H) + HCO2H]721675, 415, 397, 277, 235, 179676C33H56O14DGMG (18:3)
galactolipid
[51,52]
27.79 [M − H]577577, 441, 299, 225, 94, 80578C30H26O12Procyanidin dimer[53]
28.1928.16[M − H]595415, 315, 279, 214, 152596C27H49O12PLysophospatidylinositol
18:2
[52,54]
29.45 [(M − H) + HCO2H]647601, 571,341, 323, 277, 265 Fatty acid derivative[55]
32.37 [M − H]279 280C18H32O2Linoleic acid[56], NMR
Table 5. Determination of TPC, DPPH free radical, and tyrosinase-inhibitory activity of the methanolic extracts of HN-M and HO-M and standards gallic acid (for DPPH assay), and kojic acid (for tyrosinase-inhibition assay).
Table 5. Determination of TPC, DPPH free radical, and tyrosinase-inhibitory activity of the methanolic extracts of HN-M and HO-M and standards gallic acid (for DPPH assay), and kojic acid (for tyrosinase-inhibition assay).
SamplesTPC
(mg GAE/g)
% Inhibition of DPPH•Tyrosinase % Inhibition
200 μg/mL100 μg/mL50 μg/mL25 μg/mL12.5 μg/mL6.25 μg/mL300 µg/mL
HN-M177.21 ± 2.3594.58 ± 0.1186.93 ± 2.4559.40 ± 0.6748.81 ± 1.5819.66 ± 2.2516.05 ± 0.9742.72 ± 0.61
HO-M150.63 ± 4.7293.96 ± 1.0085.66 ± 4.3857.11 ± 2.8447.83 ± 2.2515.84 ± 2.5413.95 ± 1.238.46 ± 1.12
Gallic acid (2.6 μg/mL)53.56 ± 2.47
Kojic acid (2.0 μg/mL) 52.84 ± 2.39
Table 6. Antimicrobial activity of the extracts HN-M and HO-M measured in terms of the MIC (mg/mL).
Table 6. Antimicrobial activity of the extracts HN-M and HO-M measured in terms of the MIC (mg/mL).
Sample/StandardsS. aureusS. epidermidisP. aeruginosaK. pneumoniaeE. cloacaeE. coliS. mutansS. viridansC. albicansC. tropicalisC. glabrata
HN-M0.100.080.750.820.930.780.120.070.670.700.79
HO-M0.120.100.720.971.000.800.150.120.700.770.90
Netilmicin3.5 × 10−34 × 10−38.5 × 10−37.7 × 10−37.5 × 10−39 × 10−3
Sanguinarine 0.0170.013
5-flucytocine 0.12 × 10−31.2 × 10−310 × 10−3
Amphotericin B 1.5 × 10−30.7 × 10−30.25 × 10−3
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

Panou, E.; Graikou, K.; Tsafantakis, N.; Sakellarakis, F.-N.; Chinou, I. Phytochemical Profiling and Biological Activities of Two Helianthemum Species Growing in Greece. Sci. Pharm. 2024, 92, 42. https://doi.org/10.3390/scipharm92030042

AMA Style

Panou E, Graikou K, Tsafantakis N, Sakellarakis F-N, Chinou I. Phytochemical Profiling and Biological Activities of Two Helianthemum Species Growing in Greece. Scientia Pharmaceutica. 2024; 92(3):42. https://doi.org/10.3390/scipharm92030042

Chicago/Turabian Style

Panou, Evgenia, Konstantia Graikou, Nikolaos Tsafantakis, Fanourios-Nikolaos Sakellarakis, and Ioanna Chinou. 2024. "Phytochemical Profiling and Biological Activities of Two Helianthemum Species Growing in Greece" Scientia Pharmaceutica 92, no. 3: 42. https://doi.org/10.3390/scipharm92030042

Article Metrics

Back to TopTop