Next Article in Journal
Simple Webserver-Facilitated Method to Design and Synthesize Artificial miRNA Gene and Its Application in Engineering Viral Resistance
Next Article in Special Issue
Seaweed Extracts as an Effective Gateway in the Search for Novel Antibiofilm Agents against Staphylococcus aureus
Previous Article in Journal
Ethnobotanical History: Duckweeds in Different Civilizations
Previous Article in Special Issue
Comparative LC–LTQ–MS–MS Analysis of the Leaf Extracts of Lantana camara and Lantana montevidensis Growing in Egypt with Insights into Their Antioxidant, Anti-Inflammatory, and Cytotoxic Activities
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Caucasian Dragonheads: Phenolic Compounds, Polysaccharides, and Bioactivity of Dracocephalum austriacum and Dracocephalum botryoides

1
Laboratory of Medical and Biological Research, Institute of General and Experimental Biology, Siberian Division, Russian Academy of Science, 670047 Ulan-Ude, Russia
2
Department of Pharmacognosy, Azerbaijan Medical University, Anvar Gasimzade Street 14, AZ1022 Baku, Azerbaijan
3
Department of Biochemistry and Biotechnology, North-Eastern Federal University, 58 Belinsky Street, 677027 Yakutsk, Russia
*
Author to whom correspondence should be addressed.
Plants 2022, 11(16), 2126; https://doi.org/10.3390/plants11162126
Submission received: 25 July 2022 / Revised: 10 August 2022 / Accepted: 12 August 2022 / Published: 15 August 2022

Abstract

:
Dracocephalum botryoides Steven and Dracocephalum austriacum L. are unexplored species of the Dracocephalum genus (Lamiaceae family) with a distribution in the Caucasus, where they are used in folk medicine and local cuisine. There are no data on the chemical composition of these Dracocephalum species. In this study, the application of a liquid chromatography-mass spectrometry technique for the metabolite profiling of methanol extracts from herbs and roots of D. austriacum and D. botryoides resulted in the identification of 50 compounds, including benzoic acid derivatives, phenylpropanoids, flavonoids and lignans. Water-soluble polysaccharides of the herbs and roots of D. austriacum and D. botryoides were isolated and characterized as mostly pectins with additive arabinogalactan-protein complexes and starch-like compounds. The antioxidant potential of the studied extracts of Dracocephalum and selected phenolics and water-soluble polysaccharides were investigated via radical-scavenging and ferrous (II) ion chelating assays. This paper demonstrates that herbs and roots of D. austriacum and D. botryoides are rich sources of metabolites and could be valuable plants for new biologically active products. To the best of our knowledge, this is the first study of whole plant metabolites and their antioxidant activity in D. austriacum and D. botryoides.

1. Introduction

For thousands of years, medicinal plants have been a valuable source of therapeutic agents, and they remain an important basis for the discovery of modern medicines [1]. The practice of herbal medicine builds on indigenous knowledge of the use of native plants for both the prevention and treatment of diseases. Local people have developed effective methods for identifying, collecting, using, and conserving medicinal plants and their habitats [2]. The implementation of ethnopharmacological and phytochemical studies on the study of plants from the local flora is an important task in the search for promising medicinal raw materials [3].
The Caucasus is a floristically diverse region and is of interest to modern researchers due to its extensive thickets of medicinal plants and the widespread use of local plant resources as medicinal plant raw materials [4]. Plants of the Lamiaceae family, and the Dracocephalum genus (dragonhead) in particular, are among the most used in folk medicine in the Caucasus [5,6]. There are 74 species in the Dracocephalum genus [7], with about 30 species of economic significance and used as medicinal plants [8]. This genus is of interest to researchers due to its wide range of biological activities, such as anti-inflammatory [9,10], antibacterial [11], antioxidant [12], etc. The genus Dracocephalum is represented in the Caucasus, in particular Azerbaijan, by six species, among which Dracocephalum botryoides Steven and Dracocephalum austriacum L. are of special interest as they have not been investigated (Figure 1) [13]. Dracocephalum botryoides is a perennial plant with numerous simple and branched pubescent stems, 10–20 cm in height; ovate-rounded leaves, 1–1.5 cm long and 0.8–1.2 cm wide; and purple flowers on short pedicels with false whorls. It grows on rocky slopes, near streams and on rocks in the alpine belt, at an altitude of 2500–3600 m. Dracocephalum austriacum is a perennial plant with single or several pubescent stems, 20–60 cm in height; linear or lanceolate leaves, 2–3 cm long and 1–2.5 mm wide; and dark purple flowers on short pedicels with false whorls. It grows on limestone and rocky slopes, on steppe and subalpine meadows up to 2400 m [14].
Freshly harvested D. austriacum and D. botryoides are applied as a spice in Azerbaijani cuisine, and the fresh herb of D. botryoides is used in vegetable salads. These Dracocephalum species are used in the preparation of the famous Azerbaijani dish kükü, which is made by some local people from eggs, fresh herbs, butter, and milk [15]. Moreover, one of the features of the first dishes of Azerbaijani cuisine is their use for medicinal purposes. Thus, xəmiraşı flour soup, with the addition of dragonhead, was used in Azerbaijani villages in ancient times to treat diseases of the respiratory and gastrointestinal tracts [16]. Furthermore, an herb infusion of D. botryoides was applied in liver diseases, gastritis, and ulcers, while an herb infusion of D. austriacum was employed as an anti-inflammatory and wound healing remedy in Azerbaijani folk medicine [17].
There is information about the essential oils for both Dracocephalum species [18] and germacrone for D. botryoides [19]. However, the chemical data known about the Dracocephalum genus makes it possible to characterize some species of this genus as known and valuable sources of bioactive components. Earlier phytochemical studies of the genus Dracocephalum reported the presence of flavonoids, terpenoids, alkaloids, lignans, coumarins, polysaccharides, and cyanogenic glycosides [9,20]. An increased interest in phenolic compounds was noted when analysing chemical information on various Dracocephalum species, which can be explained by their good antioxidant activity [12,21,22,23].
As part of the ongoing work involving the metabolomic study of the Dracocephalum genus [24,25,26,27], we realized the first analysis of herb and root extracts of D. austriacum and D. botryoides using high-performance liquid chromatography with diode array and electrospray triple quadrupole mass detection (HPLC-PDA-ESI-QQQ-MS). Water-soluble polysaccharides (WSPS) of the herb and roots of D. austriacum and D. botryoides were also investigated. As most of the metabolites found in D. austriacum and D. botryoides were phenolic compounds, the antioxidant potential of the studied extracts of Dracocephalum, as well as WSPS and selected phenolic compounds, was studied using five in vitro models. To the best of our knowledge, this is the first study of whole plants metabolites and their antioxidant activity for D. austriacum and D. botryoides.

2. Results and Discussion

2.1. LC-MS Profiles of Herb and Root Extracts of Two Dracocephalum Species

Chromatographic profiles of herbs and roots of D. botryoides and D. austriacum were realized by high-performance liquid chromatography with photodiode array and electrospray ionization mass spectrometric detection (HPLC-PDA-ESI-QQQ-MS). Compounds of both Dracocephalum species were identified after a precise interpretation of the chromatographic and spectral data (using retention times and ultraviolet-visible spectra/mass spectral patterns, respectively) in comparison with reference standards and literature data. The obtained HPLC-PDA-ESI-QQQ-MS chromatograms of herb and root extracts from D. austriacum and D. botryoides revealed the presence of 50 compounds with interpretable data (Figure 2 and Figure 3), details of which are shown in Table 1.

2.1.1. Benzoic Acid Derivatives, Phenylpropanoids and Lignans

Two derivatives of benzoic acid {4-hydroxybenzoic acid 4-O-glucoside (8), 4-hydroxybenzoic acid O-hexoside-O-malonyl ester (16)} were found in the herb of D. botryoides. 4-Hydroxybenzoic acid 4-O-glucoside (8) was identified using the reference standard. The mass spectrometric analysis of compound 16 demonstrated the loss of malonyl (86 a.m.u.) and hexose fragments (162 a.m.u.), and the remaining fragment with m/z 299 was related to the 4-hydroxybenzoic acid O-hexoside moiety. The assumed structure of compound 16 was found to be 4-hydroxybenzoic acid O-hexoside-O-malonyl ester. Compound 8 has not been found previously in the Dracocephalum genus but has been detected in Nandina domestica before [28].
Seventeen compounds were determined as phenylpropanoids, separated into danshensu (3) and its derivatives {danshensu O-hexoside (1) and danshensu O-acetyl ester (4)}; caffeic (15) and quinic acid derivatives {caftaric acid (2), 4-O-caffeoylquinic acid (5), 5-O-caffeoylquinic acid (11), 3-O-caffeoylquinic acid (12), caffeic acid O-hexoside (13) and 1,3-di-O-caffeoylquinic acid (14)}, derivatives of p-coumaric acid {benzyl O-p-coumaroyl-O-desoxyhexoside-O-hexoside (38) and benzyl O-p-coumaroyl-O-hexoside (39)} and derivatives of rosmarinic acid (30) {lithospermic acid B O-hexoside (31), rosmarinic acid di-O-methyl ester (47), lithospermic acid A (33) and lithospermic acid B (32)}. The presence of danshensu (3) in the herb and roots of D. botryoides was established by comparison with the reference standard. The mass spectrometric analysis of 1 and 4 demonstrated the loss of a carbohydrate fragment of hexose (162 a.m.u.) and an acetyl fragment (42 a.m.u.), leaving the fragment with m/z 197, related to the danshensu moiety. The provisional structures of 1 and 4 were found to be danshensu O-hexoside and danshensu O-acetyl ester, respectively. Trace amounts of danshensu O-hexoside (1) was revealed in the herb of D. austriacum, while danshensu O-acetyl ester was detected in the roots of D. botryoides. The presence of danshensu and its derivatives in Dracocephalum species has been revealed for the first time; previously, danshensu was detected in the genus Salvia [29] and Orthosiphon [30].
The identification of caffeic (15) and quinic acids derivatives, such as monocaffeoylated 3-O-(12), 4-O-(5), 5-O-(11) and dicaffeoylated 1,3-di-O-caffeoylquinic acids (14), was carried out by comparing the retention times, UV- and mass-spectra with reference standards. The authentication of caftaric acid (2), a derivative of caffeic and tartaric acids, was realised in the same way. Component 13 produced a deprotonated ion with m/z 341 and a dehexosylated fragment with m/z 179, characteristic for caffeic acid O-hexoside. Caffeic acid (15) and its hexoside (13) were revealed in the herb and roots of D. austriacum. Caftaric acid (2) was also detected in the herb of D. austriacum. Monocaffeoylated 3-O-(12), 4-O-(5) and 5-O-caffeoylquinic acids (11) were revealed in both investigated Dracocephalum species, while 1,3-di-O-caffeoylquinic acid (14) was found in trace amounts in the herb and roots of D. botryoides only. Caffeic acid derivatives are characteristic compounds for Dracocephalum species. Caffeic acid (15) was previously revealed in D. moldavica [21], D. peregrinum [31], D. ruyschiana [32] and D. palmatum [24]. 3-O-caffeoylquinic acid (12) was discovered in D. peregrinum [31], D. tanguticum [33] and D. palmatum [24], while 5-O-caffeoylquinic acid (11) was previously found in D. palmatum [24]. Caftaric acid was also detected earlier in D. palmatum [24]. There are no literature data on the early detection of 4-O-caffeoylquinic acid (5) in other species of the Dracocephalum genus.
Rosmarinic acid (30) and its derivatives, lithospermic acid A (33) and lithospermic acid B (salvianolic acid B, 32), were identified by comparing their retention times and UV- and mass spectrometric data with reference standards. Derivatives of rosmarinic acid were described as lithospermic acid B O-hexoside (31) and rosmarinic acid di-O-methyl ester (47). Compounds 3032 were revealed in both investigated Dracocephalum species, while components 33 and 47 were detected only in D. austriacum. Rosmarinic acid (30) has previously been found in D. moldavica [21], D. kotschyi [34], D. palmatum [25] and D. forrestii [35]. Lithospermic acid B (32) was previously detected in D. forrestii [35] and D. palmatum [25], while lithospermic acid A (33) was revealed in D. forrestii [36].
In the mass spectra of compounds 38 and 39, we found the primary loss of the neutral fragment with m/z 146, related to the p-coumaroyl moiety (m/z 561 [M-H] → 415; m/z 415 [M-H] → 269), and the remaining fragment with m/z 269 was related to the benzoyl-hexose. The provisional structures of 38 and 39 were found to be benzyl O-p-coumaroyl-O-desoxyhexoside-O-hexoside and benzyl O-p-coumaroyl-O-hexoside, respectively. Both compounds were detected in the herb of D. austriacum.
Four lignans {schizotenuin A (46), nepetamultin A O-hexosides (48, 49) and nepetamultin A (50)} were detected in the herbs and roots of D. austriacum and D. botryoides. Schizotenuin A (46) was identified using the reference standard and was revealed in the herb of D. austriacum in trace amounts. Benzofuran lignan nepetamultin A (50) was identified in the herbs and roots of D. austriacum and D. botryoides by comparison with the reference standard. Compounds 48 and 49 had UV spectra typical of caffeic acid derivatives (λmax 290 and 321 nm), and their mass spectra showed the primary loss of the particle with m/z 162, related to hexose, and the remaining fragment, with m/z 743, was related to nepetamultin A and the loss of the particle with m/z 212 due to the loss of hydroxyhydrocafeic acid. The supposed structures of 49 and 50 were found to be nepetamultin A O-hexosides. Compounds 46 and 50 have not previously been found in the Dracocephalum genus, although schizotenuin A has been detected in Lycopus lucidus [37] and Schizonepeta tenuifolia [38], while nepetamultin A has been revealed in Nepeta multifida [39].

2.1.2. Flavonoids

Twenty-seven flavonoid compounds were determined in herbs and roots of D. austriacum and D. botryoides, belonging to one of five groups, depending on their aglycone structure: flavones luteolin (12 components), apigenin (6 components) and acacetin (3 components), and flavanones eriodictyol (3 components) and naringenin (3 components). The luteolin derivatives were the largest group with 12 compounds including non-acylated and acylated derivatives. Seven non-acylated compounds were identified using the reference standards as luteolin 7,4′-di-O-rutinoside (10), luteolin 7-O-rutinoside-4′-O-glucoside (cynarotriside, 17), luteolin 7-O-rutinoside (scolymoside, 19), luteolin 7-O-glucoside (cynaroside, 20), luteolin 4′-O-glucoside (22), luteolin 3′-O-glucoside (24) and luteolin (37). The most diverse composition of non-acylated luteolin derivatives was noted in the herb of D. austriacum (10, 17, 19, 20, 22, 24 and 37), while only two compounds (10 and 22) were found in the roots. Compounds 37, 19, 20 and 22 were present in the herb of D. botryoides, while compounds 20 and 22 were detected in the roots of this species. The remaining compounds gave an aglycone ion with m/z 285 in the mass spectra, and the loss of hexose fragments (m/z 162) and desoxyhexose (m/z 146) was observed in a 3:2 ratio (luteolin tri-O-hexoside-di-O-desoxyhexoside, 6 and 7) and in a 1:1 ratio (luteolin O-desoxyhexoside-O-hexoside, 21). Components 6 and 7 were detected in the herb of D. austriacum, while 21 was revealed in herb and roots of D. austriacum and in the herb of D. botryoides. The acylated luteolin derivative luteolin 7-O-(6″-O-acetyl)-glucoside (27) was identified by comparison with the reference standard. Compound 34 gave an aglycone ion with m/z 285 in the mass spectra, and the consistent loss of particles with m/z 42, related to acetyl, and the loss of a hexose fragment (m/z 162) was observed. The provisional structure of 34 was found to be luteolin 7-O-(2″,6″-di-O-acetyl)-hexoside. Compounds 27 and 34 were revealed in the herb and roots of D. botryoides. Luteolin was previously discovered in D. moldavica [39], D. peregrinum [31], D. kotschyi [22], D. subcapitatum [40], D. rupestre [41], D. tanguticum [42] and D. palmatum [25]. Two rare di-O-glucosides of luteolin, 10 and 17, were previously found in D. palmatum [24], while luteolin 7-O-rutinoside was previously detected in D. peregrinum [31] and D. palmatum [24]. Luteolin 7-O-glucoside was previously revealed in D. rupestre [43], D. peregrinum [31], D. moldavica [21], D. kotschyi [44], D. tanguticum [42] and D. palmatum [25], while compound 22 was previously detected in D. palmatum [24]. Luteolin 3′-O-glucoside has been found in D. thumiflorum [45] and D. nutans [46], while the acylated luteolin derivative 27 was previously discovered in D. palmatum [27].
The flavanone eriodictyol (36) and its derivatives eriodictyol 7-O-rutinoside (9) and eriodictyol 7-O-glucoside (18) were identified by comparing their retention times, UV and ESI-MS patterns with reference standards. Compounds 9, 18 and 36 were found in the herb of D. botryoides. Eriodictyol was previously identified in D. peregrinum [31], D. palmatum [25], D. rupestre [47] and D. tanguticum [42]; eriodictyol 7-O-rutinoside (9) was revealed in D. palmatum [24], while eriodictyol 7-O-glucoside (18) was detected in D. rupestre [9] and D. palmatum [25]. The identification of the flavanone naringenin (40) and its derivatives naringenin 7-O-rutinoside (23) and naringenin 7-O-glucoside (26) was carried out by comparison of retention times, UV- and mass-spectra with reference compounds. Naringenin 7-O-rutinoside and naringenin were detected in the herb of D. botryoides, while naringenin 7-O-glucoside was found in both the herb and roots of this species. Compounds 40 and 26 were previously revealed in D. rupestre [9] and D. palmatum [25], while compound 23 was discovered in D. forrestii [48].
The apigenin group of flavones consisted of non-acylated and acylated derivatives. After comparison with reference standards, the non-acylated compounds were identified as apigenin (41), apigenin 7-O-rutinoside (25) and apigenin 7-O-glucoside (28). Apigenin was revealed in the herb of D. botryoides, and apigenin 7-O-rutinoside was detected in the herb and roots of D. austriacum, while apigenin 7-O-glucoside was found only in the herb of this species. The acylated apigenin derivatives apigenin 7-O-(4″-malonyl-6″-O-acetyl)-glucoside (42) and apigenin 7-O-(6″-O-acetyl)-glucoside (44) were identified by comparison with the reference standard. Compound 45 gave an aglycone ion with m/z 269 in the mass spectra, and the consistent loss of particles with m/z 42, related to acetyl, and the loss of a hexose fragment (m/z 162) was observed. The provisional structure of compound 45 was found to be apigenin O-hexoside-di-O-acetyl ester. All acylated apigenin derivatives (42, 44 and 45) were revealed in the herb of D. botryoides. Apigenin was previously discovered in D. rupestre [9], D. moldavica [49], D. kotschyi [10], D. palmatum [25], D. polychaetum [50] and D. oligadenium [51], and apigenin 7-O-rutinoside has been detected in D. heterophyllum [52] and D. palmatum [24]. Apigenin 7-O-glucoside was found previously in D. kotschyi [53], D. palmatum [25], D. tanguticum [54] and D. moldavica [55], and apigenin 7-O-(6″-O-acetyl)-glucoside was previously revealed in D. palmatum [27], while apigenin 7-O-(4″-malonyl-6″-O-acetyl)-glucoside has not previously been found in any species of the genus Dracocephalum but has been identified in Matricaria chamomilla [56].
The acacetin group of flavones was represented by three compounds. Acacetin 7-O-glucoside (43) was identified by comparing its retention time, UV and ESI-MS pattern with the reference standard. The remaining compounds showed the loss of hexose fragments with m/z 162 or fragments of desoxyhexose (m/z 146) in ratios of 3:1 (acacetin tri-O-hexoside-O-desoxyhexoside, 29) and 1:1 (acacetin O-desohyhexoside-O-hexoside, 35) in the mass spectra. Compound 35 was found in the herb and roots of D. austriacum, while compound 29 was found only in the herb of D. austriacum. Meanwhile, compound 43 was detected in herbs of both D. austriacum and D. botryoides. Acacetin 7-O-glucoside was previously identified in D. foetidum [57], D. tanguticum [58], D. moldavica [58], D. peregrinum [31], D. kotschyi [53] and D. palmatum [24].
As a result of the chromatographic research of D. austriacum and D. botryoides, 50 metabolites of various chemical groups were identified. This study demonstrates that the whole plant of both Dracocephalum species are characterized by a specific accumulation of phenolic metabolites. The highest content of phenolic compounds was typically found in the herb of both Dracocephalum species, while the roots were characterized by the lowest phenolic diversity. Flavonoid derivatives of luteolin and eriodictyol (luteolin 7-O-glucoside, luteolin 7-O-(6″-O-acetyl)-glucoside, luteolin 4′-O-glucoside and eriodictyol 7-O-glucoside) were the main compounds in herb of D. botryoides, while luteolin 4′-O-glucoside and 5-O-caffeoylquinic acid dominated in herb of D. austriacum. The roots of D. austriacum and D. botryoides had a similar chemical profile, with a predominance of the phenylpropanoids rosmarinic acid and lithospermic acid B. This may affect the biological properties of these species, particularly the antioxidant activity of dragonhead extracts, since the activity of diverse phenolic groups is known to be variable [59,60,61].
Table 1. Retention times (t), ultraviolet (UV), and mass spectral (ESI-MS) data of compounds 150 were found in leaves and roots of D. austriacum and D. botryoides, in addition to their content (mg/g of dry plant weight, in brackets S.D.).
Table 1. Retention times (t), ultraviolet (UV), and mass spectral (ESI-MS) data of compounds 150 were found in leaves and roots of D. austriacum and D. botryoides, in addition to their content (mg/g of dry plant weight, in brackets S.D.).
Not, minCompound [Ref.]Ident. Level aUV
Spectrum, λmax, nm
ESI-MS,
[M-H], m/z
ESI-MS,
Additional Ions, m/z
D. austriacumD. botryoides
HerbRootsHerbRoots
12.00Danshensu O-hexoside2280359197, 179, 135trace b
22.52Caftaric acid1328311179, 149, 1350.14 (0.00)
32.78Danshensu1280197179, 135 tracetrace
43.08Danshensu O-acetyl ester2280239197, 179, 135 tracetrace
55.284-O-Caffeoylquinic acid1328353191, 179, 173, 1350.58 (0.02)0.14 (0.00)trace1.02 (0.04)
65.51Luteolin tri-O-hexoside-
di-O-desoxyhexoside
2263, 3331063901, 755, 593, 447, 285trace
75.62Luteolin tri-O-hexoside-
di-O-desoxyhexoside
2263, 3331063901, 755, 593, 447, 2850.08 (0.00)
85.834-Hydroxybenzoic acid 4-O-glucoside1256299137 2.43 (0.10)
95.98Eriodictyol 7-O-rutinoside (eriocitrin)1287595449, 287 trace
106.22Luteolin 7, 4′-di-O-rutinoside1265, 333901755, 593, 447, 285trace0.11 (0.00)
116.425-O-Caffeoylquinic acid1328353191, 1653.14 (0.12)0.09 (0.00)0.52 (0.02)trace
126.743-O-Caffeoylquinic acid1328353191, 179, 135trace 0.08 (0.00)trace
136.81Caffeic acid O-hexoside2328341179, 1350.79 (0.03)0.33 (0.02)
147.001,3-Di-O-Caffeoylquinic acid1328515353, 191, 179, 135 tracetrace
157.09Caffeic acid13271791350.47 (0.03)0.58 (0.02)
167.114-Hydroxybenzoic acid O-hexoside-O-malonyl ester2256385299, 137 1.80 (0.07)
177.31Luteolin 7-O-rutinoside-4′-O-glucoside (cynarotriside)1265, 334755593, 447, 2850.11 (0.00)
187.47Eriodictyol 7-O-glucoside1287449287 4.83 (0.24)
197.72Luteolin 7-O-rutinoside (scolymoside)1255, 346593447, 2850.14 (0.00) 0.06 (0.00)
207.98Luteolin 7-O-glucoside (cynaroside)1256, 3454472850.09 (0.00) 22.14 (0.92)1.29 (0.06)
218.21Luteolin O-desoxyhexoside-O-hexoside2269, 337593447, 2851.73 (0.07)trace0.15 (0.00)
228.51Luteolin 4′-O-glucoside1269, 33744728518.36 (0.73)trace3.79 (0.15)0.91 (0.03)
238.71Naringenin 7-O-rutinoside (narirutin)1289579433, 271 1.47 (0.06)
248.82Luteolin 3′-O-glucoside1268, 3424472851.20 (0.05)
259.01Apigenin 7-O-rutinoside1265, 335577431, 2690.67 (0.03)0.93 (0.04)
269.04Naringenin 7-O-glucoside1289433271 0.58 (0.02)trace
279.24Luteolin 7-O-(6″-O-acetyl)-glucoside1256, 344489447, 285 4.22 (0.17)2.46 (0.10)
289.28Apigenin 7-O-glucoside1266, 336431269trace
299.31Acacetin tri-O-hexoside-
O-desoxyhexoside
2268, 333915445, 283trace
309.53Rosmarinic acid1329359719, 395, 179, 1610.98 (0.04)7.39 (0.27)1.19 (0.04)10.85 (0.44)
319.72Lithospermic acid B O-hexoside2250, 290, 306, 330879717, 537, 519, 179trace 1.29 (0.05)
329.96Lithospermic acid B1251, 289, 305, 329717537, 519, 1791.27 (0.06)12.73 (0.63)0.07 (0.0)11.39 (0.46)
3310.04Lithospermic acid A1253, 289, 310537493, 295trace1.68 (0.07)
3410.09Luteolin 7-O-(2″,6″-di-O-acetyl)-hexoside2256, 344531489, 447, 285 1.04 (0.02)trace
3510.74Acacetin O-desoxyhexoside-O-hexoside2268, 332591445, 283trace0.76 (0.02)
3610.91Eriodictyol1287287 trace
3711.48Luteolin1256, 346285 trace 0.04 (0.00)
3811.53Benzyl O-p-coumaroyl-O-desoxyhexoside-O-hexoside2316561415, 2690.83 (0.03)
3912.02Benzyl O-p-coumaroyl-O-hexoside2316415269trace
4012.06Naringenin1289271 trace
4112.58Apigenin1266, 337269 0.09 (0.0)
4213.31Apigenin 7-O-(4″-malonyl-6″-O-acetyl)-glucoside1266, 335559431, 269 0.02 (0.00)
4314.48Acacetin 7-O-glucoside1268, 335445283trace 0.08 (0.00)
4415.52Apigenin 7-O-(6″-O-acetyl)-glucoside1265, 336473431, 269 0.07 (0.00)
4516.50Apigenin O-hexoside-di-
O-acetyl ester
2265, 336515431, 269 trace
4616.82Schizotenuin A1289, 3207151431, 535, 357trace
4718.69Rosmarinic acid di-O-methyl ester2330387359, 179, 161trace
4823.53Nepetamultin A O-hexoside2290, 321905743, 531, 355 0.69 (0.02)
4924.00Nepetamultin A O-hexoside2290, 321905743, 531, 3550.14 (0.00)trace
5025.08Nepetamultin A1290, 320743531, 3550.35 (0.02)trace0.52 (0.01)trace
a Identification levels: (1) identified compounds after comparison of UV, mass-spectral data, and retention time with reference standards; (2) putatively annotated compounds after comparison of UV and mass-spectral data with literature data [62]. b traces—<LOQ (limit of quantification).

2.2. Water-Soluble Polysaccharides of Herb and Roots of Two Dracocephalum Species

The fractions of WSPS were obtained from the herb and root samples of D. austriacum and D. botryoides by hot (90 °C) water extraction, followed by triple ethanol precipitation, deproteinization, ion-exchange resin and polyamide purification. The pure WSPS gave 0.6–1.2% yield (of dry plant weight) and were completely soluble in hot water (Table 2). The total carbohydrate content of the WSPS varied from 88.3% (WSPS of herb of D. botryoides) to 92.8% (WSPS of roots of D. austriacum), including a content of 25.3–49.2% uronic acids in herb WSPS and 11.2–12.0% uronic acids in root WSPS. All fractions were protein positive despite the deproteinization procedure used; the lowest protein level was 1.0% (WSPS of roots of D. botryoides) and the highest was 1.8% (WSPS of herb of D. austriacum). Only the root WSPS gave a positive iodine reaction, indicating the presence of starch-like polymers opposite negative resorcinol and Fehling’s reactions typical for fructans and mannans. The β-glucosyl Yariv reagent showed the precipitation of all four WSPS, indicating the formation of arabinogalactan-protein complexes. The monosaccharide composition study showed that herb WSPS had the highest content of galacturonic acid (23.6–48.0 mol%), arabinose (12.1–27.4 mol%) and galactose (16.2–20.5 mol%), and root WSPS had the highest content of arabinose (21.3–25.5 mol%), galactose (23.9–24.1 mol%) and glucose (22.6–25.6 mol%).
The Fourier-transform infrared (FTIR) spectral patterns of WSPS from herbs of D. austriacum and D. botryoides showed the typical pectin bands (Figure 4). There were intense bands of stretching vibrations of the esterified carboxylic groups (1714–1741 cm−1) and free carboxyls (1600–1606 cm−1), as well as bands of asymmetric and symmetric vibrations of the methyl esters (1437 cm−1), O-C-O fragments of esters (1260–1262, 1407–1418 cm−1) and C-H vibrations of the pyran rings (1327–1323 cm−1) [63]. The 900–1200 cm−1 region included vibration bands of the pyran rings and the glycoside bonds of pectins at 1140, 1099, 1080–1084, 1050–1054, 1021 and 976 cm−1 [64]. The presence of α-linkages and aldopyranoses were confirmed by the strong absorption bands at 831–840 cm−1 and 886 cm−1 in the anomeric region of the FTIR spectra, respectively. The FTIR spectral properties of WSPS from roots of D. austriacum and D. botryoides were generally close to that of the herb samples, except in the 900–1200 cm−1 region, and the anomeric region showed shapes specific for the starch-like polymers.
The spectra of WSPS in the ultraviolet region had shoulders at 270–280 nm and 320–331 nm, indicating the presence of phenolic fragments (Figure 5). The pure polysaccharides (like apple pectin) did not have maxima in the UV spectrum. The shortwave band was most likely the secondary benzoic band (B-band) caused by the p-hydroxy-phenyl fragments, while the longwave band was a K-band, owing to the n ⟶ π* transitions of the p-coumaroyl-like fragments. The ionization of phenolic functional groups by alkaline additives (NaOH) resulted in the appearance of additional maxima at 275–290, 310–339, 369–373 and 394–395 nm, typical for the simple phenolic compounds in alkaline media.
Alkaline hydrolysis of the WSPS of herbs and roots of D. austriacum and D. botryoides resulted in the release of 17 phenolic compounds, which were identified as benzoic acids, benzoic aldehydes and hydroxycinnamates using HPLC-DAD-MS (Table 3). The main phenolics in the WSPS of D. austriacum were p-methoxybenzoic acid (19.3–22.4%), vanillic acid (18.6–19.3%), vanillin (8.4–11.6%) and p-coumaric acid (10.7–15.2%), and the main phenolics in the WSPS of D. botryoides were p-methoxybenzoic acid (25.7–31.1%), vanillic acid (25.7–28.3%), veratric acid (14.7–19.7%) and vanillin (10.3–11.6%).
The WSPS of herbs and roots of D. austriacum and D. botryoides, therefore, had a complex structure consisting of at least three fragments of carbohydrate, protein, and phenolic nature. The polysaccharides were mostly pectins with additive arabinogalactan-protein complexes and starch-like compounds, linked with phenolic fragments of benzoic acids, benzoic aldehydes and hydroxycinnamates.
Dragonhead polysaccharides are still poorly researched; currently, there are only data relating to polysaccharides of the herb of D. palmatum, which are pectin, arabinogalactans, and a starch mixture [24], and information about polysaccharides of D. moldavica [20]. Dracocephalum membership in the subtribe Nepetinae, tribe Mentheae, which is included in the subfamily Nepetoideae of the Lamiaceae family, demonstrates the taxonomical proximity to Agastache, Cedronella, Nepeta, Glechoma, Hymenocrater and Meehania species [65]. Even so, polysaccharides have not been studied in the above genera. The closest known data about carbohydrate polymers belongs to the Menthinae and Salviinae subtribes, where galacturonans, glucans and arabinogalactans were found as components of Lycopus [66], Mentha [67,68], Origanum [69], Prunella [70], Rosmarinus [71], Salvia [72] and Thymus [73], indicating the prevalence of these polymers in the Mentheae tribe that Dracocephalum represents. The high phenolic content of polysaccharides of D. austriacum and D. botryoides seems unusual for Lamiaceae plants but is common for many other plants, showing the presence of cross-connected pectin-phenol and hemicellulose-lignin complexes [74]. Some of polyphenol linked polysaccharides are bioactive polymers with antioxidant, antidiabetic, and antitumor properties [75].

2.3. Antioxidant Activity of Extracts of D. austriacum and D. botryoides, Selected Phenolics and Water-Soluble Polysaccharides

Phenols are widely known as protective compounds against free radicals and toxic metals [76,77,78], so we decided to study the antioxidant potential of extracts of D. austriacum and D. botryoides as possible radical scavengers and metal chelators. Five well-known antioxidant assays were used to study the scavenging properties against 2,2-diphenyl-1-picrylhydrazyl radicals (DPPH), 2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) cation radicals (ABTS•+), hydroxyl radicals and superoxide anion radicals, as well as the ferrous (II) ion chelating ability [79]. Seven selected compounds representing various phenolic groups detected in D. austriacum and D. botryoides were analysed, as well as WSPS. Trolox was used as a reference substance (Table 4).
The studied extracts of Dracocephalum demonstrated good scavenging effects against synthetic free radicals, such as DPPH and ABTS•+. The herb extract of D. botryoides was the most active, with IC50 values of 28.63 and 25.20 µg/mL for DPPH and ABTS•+ radicals, respectively, while roots of this species showed the lowest IC50 value among the extracts of Dracocephalum (40.67 and 36.14 µg/mL, respectively). Individual phenylpropanoids rosmarinic acid and lithospermic acid B showed superior scavenging activity against the DPPH radical, with IC50 values of 6.22 and 9.08 µg/mL, respectively, while the flavonoids eriodictyol 7-O-glucoside and luteolin 7-O-glucoside demonstrated maximal inhibition of the ABTS•+ radical (IC50 7.39 and 8.26 µg/mL, respectively). The WSPS in Dracocephalum scavenged DPPH and ABTS•+ radicals with low efficiency (IC50 102.85 and 82.79 µg/mL, respectively, for root WSPS D. botryoides). The same parameters for Trolox were 7.03 and 3.44 µg/mL, respectively. The ability of extracts of Dracocephalum to scavenge oxygen radicals (hydroxyl radicals and superoxide anion radicals) was moderate, with values prevailing for herb extract of D. botryoides (IC50 103.28 and 85.67 µg/mL, respectively). Significant scavenging activity against OH radicals was found for luteolin 7-O-glucoside and luteolin 7-O-(6″-acetyl)-glucoside (53.29 and 55.73 µg/mL, respectively). Rosmarinic acid and lithospermic acid B demonstrated effective inactivation of O2•− radicals (15.38 and 16.27 µg/mL, respectively). The potential of root WSPS from D. botryoides to scavenge OH and O2•− radicals was good (IC50 105.73 and 73.68 µg/mL, respectively). The maximum Fe2+-chelating ability was found with herb extract of D. botryoides (0.51 mM Fe2+-ions/g), while rosmarinic acid and eriodictyol 7-O-glucoside had the best chelating properties among the individual compounds (3.72 and 3.18 mM Fe2+-ions/g, respectively). The herb WSPS of D. austriacum demonstrated the best iron-chelating activity (4.31 mM Fe2+-ions/g). The same parameter for Trolox was 1.53 mM Fe2+-ions/g.
Previous studies have been conducted on the antioxidant activity of other Dracocephalum species. The water-soluble extract of D. moldavica revealed DPPH radical scavenging activity, with an IC50 of 445.90 μg/mL, and superoxide anion radical scavenging activity (IC50 467.20 μg/mL) [21]. The methanol extract of D. kotschyi inhibited DPPH, with IC50 values of 60.69 and 51.54 µg/mL for two-year and six-year-old samples, respectively [12]. The IC50 values for the DPPH, ABTS•+ and OH radical-scavenging activities for the extract of D. rupestre were 50.01, 43.62 and 28.59 µg/mL, respectively [80], and the methanol extract of shoots of D. polychaetum had an IC50 value of 5600 µg/mL in the DPPH inhibiting assay [50].
The data obtained in five in vitro assays demonstrated a good efficacy for the herb extract of D. botryoides as an antioxidant agent. This trend is not surprising at all; the presence of strong antioxidants, such as phenylpropanoids and flavonoids in the herb extract of D. botryoides have led to such results. The antioxidant activity of herb and root extracts of D. botryoides and D. austriacum, as well as their water-soluble polysaccharides, have been shown here for the first time.

3. Conclusions

In this paper, the whole plants of D. austriacum and D. botryoides were shown to be natural accumulators of diverse metabolites of a phenolic and non-phenolic nature. The metabolites of these plant species were investigated for the first time using HPLC-PDA-ESI-QQQ-MS, and the water-soluble polysaccharides of roots of D. austriacum and D. botryoides were studied. The dragonhead polysaccharides were mainly found to be pectins with additive arabinogalactan-protein complexes and starch-like compounds, linked with phenolic fragments of benzoic acids, benzoic aldehydes and hydroxycinnamates. The phenolic diversity in herbs and roots of D. austriacum and D. botryoides implies the presence of antioxidant properties, which were confirmed by five in vitro assays (DPPH, ABTS•+, OH, O2•− radical-scavenging activity and Fe2+-chelating activity). Thus, the information presented highlights the potential of the herbs and roots of D. austriacum and D. botryoides for possible future plant remedies or sources of new functional products.

4. Materials and Methods

4.1. Plant Material and Chemicals

Samples of Dracocephalum austriacum (herb and roots) were collected during the flowering period in the subalpine meadow in Azerbaijan in 5 locations, 10 samples from each near Gryz, Guba region (16.VI.2020, 41°21′93.2″ N, 48°24′55.8″ E, 1900 m a.s.l.; voucher No Az/LDR-0620/74-95). Samples of D. botryoides (herb and roots) were collected during the flowering period in alpine meadow in Azerbaijan in 5 locations, 10 samples from each near Khinalig, Guba region (5.VII.2020, 41°19′24.9″ N, 48°12′07.0″ E, 2850 m a.s.l.; voucher No Az/LDR-0720/87-114). The species were authenticated by authors Javanshir I. Isaev (Azerbaijan Medical University, Baku, Azerbaijan) and Daniil N. Olennikov (IGEB SB RAS, Ulan-Ude, Russia). Plant material was dried in the ventilated heat oven at 40 °C within 8–10 days and stored at 3–4 °C before analysis.
The reference compounds were purchased from BioCrick Co., Ltd. (Chengdu Tianfu, Sichuan, China); ChemFaces (Wuhan, Hubei, China); Extrasynthese (Lyon, France); PhytoLab GmbH & Co. KG (Vestenbergsgreuth, Germany); Sigma–Aldrich (St. Louis, MO, USA) or isolated in our laboratory (Table S1). Selected chemicals were from Biosupplies Australia Ply Ltd. (Victoria, Australia): Yariv reagent kit (Cat. No. 100-4); Sigma–Aldrich (St. Louis, MO, USA): acetonitrile for HPLC (Cat. No. 34851, ≥99.9%), anisic aldehyde (Cat. No. A88107, ≥98%), anthrone (Cat. No. 319899, ≥97%), arabinose (Cat. No. A3256, ≥99%), 2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) diammonium salt (Cat. No. A1888, ≥ 98%), trans-cinnamic acid (Cat. No. C80857, ≥99%), p-coumaric acid (Cat No. C9008, ≥98%), 3,5-dimethylphenol (Cat. No. 144134, ≥99%), 2,2-diphenyl-1-picrylhydrazyl radical (Cat. No. 281689, ≥97%), Fehling’s reagent I (Cat. No. 36018), trans-ferulic acid (Cat. No. 128708, ≥99%), ferrous sulphate (Cat. No. PHR1483), Folin–Ciocalteu′s phenol reagent (Cat No. 47641), formic acid (Cat. No. F0507, ≥95%), fucose (Cat. No. F8150, ≥98%), galactose (Cat. No. G0750, ≥99%), galacturonic acid monohydrate (Cat. No. 48280, ≥97%), gentisic acid (Cat. No. 149357, ≥98%), glucose (Cat. No. G8270, ≥99.5%), glucuronic acid (Cat. No. G5269, ≥98%), 4-hydroxybenzoic acid (Cat. No. H20059, ≥99%), isovanillic acid (Cat. No. 220108, ≥97%), trans-isoferulic acid (Cat. No. 05407, ≥98%), lithium perchlorate (Cat. No. 205281, ≥95%), mannose (Cat. No. M8574, ≥99%), p-methoxycinnamic acid (Cat. No. M13807, ≥99%), methanol (Cat. No. 322415, ≥99.8%), 4-methoxybenzoic acid (Cat. No. 117390, ≥99%), perchloric acid (Cat. No. 244252, ≥70%), protocatechuic acid (Cat. No. 03930590, ≥99%), rhamnose (Cat. No. W373011, ≥99%), resorcinol (Cat. No. 398047, ≥99%), ribose (Cat. No. R7500, ≥99%), sodium carbonate (Cat. No. S7795, ≥99%), trolox (Cat. No. 238813, ≥97%), vanillic acid (Cat. No. H36001, ≥97%), vanillin (Cat. No. V1104, ≥99%), veratric acid (Cat. No. D131806, ≥99%), syringic acid (Cat. No. S6881, ≥95%), xylose (Cat. No. X1500, ≥99%).

4.2. Plant Extracts Preparation

To prepare plant extracts for HPLC analysis, the ground plant material (particle size 0.125 μm; weight 200 mg) was treated with 70% methanol (2 mL) three times by sonication (ultrasonic bath, 30 min, 50 °C, ultrasound power 100 W, frequency 35 kHz). The obtained liquid extracts were centrifuged at 20 °C (6000× g, 10 min) and the supernatants were filtered through 0.22 μm syringe filters into the measuring flask (10 mL). The final volume was 10 mL with 70% methanol. The obtained methanol extracts were stored at 2 °C before treatment. Before analysis, internal standard (methanol solutions) was added to samples in a ratio of 1:1: herb extracts—3′,4′-di-O-acetyl-cis-khellactone (5 μg/mL), root extracts—3′,4′-di-O-acetyl-cis-khellactone (5 μg/mL).
The extracts of herbs and roots of D. austriacum and D. botryoides for the antioxidant study were prepared from the ground samples (50 g), treated by the 70% methanol (1 L) in an ultrasonic bath (30 min, 45 °C, ultrasound power 100 W, frequency 35 kHz), filtered, reduced in a vacuum until dryness and stored (−20 °C) before analysis. The yields of the extracts from D. austriacum were 23.4% for the herb sample, 19.8% for the root sample. The yields of the extracts from D. botryoides were 21.7% for the herb sample and 18.1% for the root sample.

4.3. High-Performance Liquid Chromatography with Photodiode Array Detection and Electrospray Ionization Triple Quadrupole Mass Spectrometric Detection (HPLC-PDA-ESI-QQQ-MS) Metabolite Profiling

High-performance liquid chromatography with photodiode array detection and electrospray ionization triple quadrupole mass spectrometric detection (HPLC-PDA-ESI-QQQ-MS) was used for the quantitative profiling of the extracts from D. austriacum and D. botryoides. Liquid chromatograph LC-20 Prominence coupled with photodiode array detector SPD-M30A (wavelength range 200–600 nm), triple-quadrupole mass spectrometer LCMS 8050 (all Shimadzu, Columbia, MD, USA) and GLC Mastro C18 column (2.1 × 150 mm, 3 μm; Shimadzu, Kyoto, Japan) was used. The two-component eluent system of eluent A (0.5% formic acid in water) and B (0.5% formic acid in acetonitrile) used the gradient program for the separation (%B): 0–4 min 4%, 4–6 min 4–10%, 6–9 min 10–15%, 9–15 min 15–25%, 15–20 min 25–38%, 20–25 min 38–70%, 25–30 min 70–88%, 30–40 min 88–4%, 40–50 min 4%. The injection volume was 1 μL and the flow rate was 120 μL/min. The UV-Vis spectra were recorded by an SPD-M20A photodiode detector (spectral range 200–600 nm) equipped with a post-column derivatization reactor. The used temperature levels were 300 °C in the ESI interface, 250 °C in the desolvation line, and 400 °C in the heat block. The flow values were 3 L/min for the nebulizing gas (N2), 10 L/min for the heating gas (air), and 0.3 mL/min for the collision-induced dissociation gas (Ar). The source voltage of mass spectra was 3 kV and the collision energy −10–30 eV (negative ionization) by the scanning range of m/z 80–1900. The LC-MS system was managed by LabSolution’s workstation software equipped with the inner LC-MS library. The integrated analysis of retention time, ultraviolet and mass spectra data after comparison with the reference standards and literature data was used for the identification of metabolites [62].

4.4. HPLC-PDA-ESI-QQQ-MS Metabolite Quantification

To quantify 50 metabolites of herbs and roots of D. austriacum and D. botryoides, the HPLC-PDA-ESI-QQQ-MS conditions were used (Section 4.3). Totally, 34 reference standards were separately weighed (10 mg) and dissolved in the methanol-DMSO mixture (1:1) in volumetric flasks (10 mL) preparing the stock solution (1000 µg/mL) used for the calibration curve building. The calibration solution (1, 10, 25, 50, 100 µg/mL) chromatographed in known HPLC-PDA-ESI-QQQ-MS conditions and mass spectral data was used to create ‘concentration–mass spectrometric peak area’ correlation. The principal validation criteria including correlation coefficients (r2), standard deviation (SYX), limits of detection (LOD), limits of quantification (LOQ), and linear ranges were found using the known method [81] (Table S2). Five HPLC runs were sufficient for the quantitative analyses and the results were expressed as mean value ± standard deviation (S.D.).

4.5. Polysaccharides Extraction and Analysis

The samples of dried milled herb and roots of D. austriacum and D. botryoides (100 g) were added to distilled water (1.0 L), heated on a boiled water bath (1 h) and after cooling to room temperature water extracts were filtered under reduced pressure and concentrated down in vacuo to 200 mL. The concentrated residues were mixed with 95% ethanol (1:5) and after 2 h the precipitates were centrifuged at 3000× g. The crude polysaccharide fractions were redissolved in 200 mL of water. The Sevag method [82] was applied for deproteination and was followed by dialysis for 48 h against distilled water using dialysis tubes with an MW-cut off of 2 kDa (Sigma–Aldrich, St. Louis, MO, USA). The non-dialysed parts were loaded on to a KU-2-8 cation-exchange resin column (H+-form, 200 g; Closed Joint-Stock Company Tokem, Kemerovo, Russia) which were eluted with 2 L of distilled water. Eluates were concentrated in vacuo up to 200 mL and then liophylized. The resulting water-soluble polysaccharides were whitish powders.
For estimation of total carbohydrate content modified anthrone-H2SO4 spectrophotometric assay with glucose as a standard compound was used [83]. To evaluate content of uronic acids 3,5-dimethylphenol method was applied calculated as galacturonic acid [84]. The Bradford method using Coomassie G250 was employed for determination of protein content [85]. Phenolic content was analysed using the Folin–Ciocalteu method [86]. Reactions of water-soluble polysaccharides solutions with iodine, resorcinol, Yariv and Fehling’s reagents were carried out accordingly [87,88,89]. The IR spectra were registered in a spectral range of 4000–600 cm−1 using a FT-801 Fourier-transform infrared spectrometer (Simex, Novosibirsk, Russia) coupled with a single reflection ATR device. Alkaline hydrolysis procedure and HPLC-DAD-MS analysis conditions of the released products were as described by us earlier [90].

4.6. Antioxidant Activity

Microplate spectrophotometric assays were applied to investigate the scavenging activity of extracts of D. austriacum and D. botryoides, selected phenolics and water-soluble polysaccharides against 2,2-diphenyl-1-picrylhydrazyl radicals (DPPH) [25], 2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) cation radicals (ABTS•+) [24], superoxide anion radicals (O2•−) [91], hydroxyl radicals (OH) [91]. Ferrous (II) ion chelating activity was investigated by spectrophotometric assay [92]. Trolox was used as a reference standard (1–100 μg/mL in methanol). The results of DPPH, ABTS•+, O2•−, OH assays were determined as IC50 (the half maximal inhibitory concentration) and calculated graphically using ‘concentration (μg/mL or mg/mL)–antioxidant activity (%)’ correlations. All the analyses were carried out five times, and the data are expressed as the mean value ± standard deviation (S.D.)

4.7. Statistical Analysis

Statistical analyses were performed by one-way analysis of variance, and the significance of the mean difference was determined by Duncan’s multiple range test. Differences at p < 0.05 were considered statistically significant. The results are presented as mean values ± standard deviations (S.D.). The linear regression analysis and generation of calibration graphs were conducted using Advanced Grapher 2.2 (Alentum Software Inc., Ramat-Gan, Israel).

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/plants11162126/s1. Table S1: Reference standards used for the qualitative and quantitative analysis by HPLC-PDA-ESI-QQQ-MS assays. Table S2: Regression equations, correlation coefficients, standard deviation, limits of detection, limits of quantification and linear ranges for 34 reference standards.

Author Contributions

Conceptualization, N.I.K., D.N.O. and J.I.I.; methodology, N.I.K. and G.S.J.; software, N.I.K. and N.K.C.; validation, D.N.O.; formal analysis, J.I.I. and G.S.J.; investigation, N.I.K., D.N.O. and G.S.J.; resources, D.N.O. and N.K.C.; data curation, N.I.K.; writing—original draft preparation, N.I.K.; writing—review and editing, D.N.O.; visualization, J.I.I.; supervision, G.S.J.; project administration, N.K.C.; funding acquisition, N.I.K., D.N.O. and J.I.I. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Ministry of Education and Science of Russia, grant numbers 121030100227-7; FSRG-2020-0019.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Acknowledgments

The authors acknowledge the Buryat Research Resource Center for the technical support in chromatographic and mass-spectrometric research.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Yeung, A.W.K.; Heinrich, M.; Atanasov, A.G. Ethnopharmacology—A bibliometric analysis of a field of research meandering between medicine and food science? Front. Pharmacol. 2018, 9, 215. [Google Scholar] [CrossRef] [PubMed]
  2. Sheng-Ji, P. Ethnobotanical approaches of Traditional medicine studies: Some experiences from Asia. Pharm. Biol. 2001, 39, 74–79. [Google Scholar] [CrossRef]
  3. Süntar, I. Importance of ethnopharmacological studies in drug discovery: Role of medicinal plants. Phytochem. Rev. 2020, 19, 1199–1209. [Google Scholar] [CrossRef]
  4. Mamedov, N.A.; Mehdiyeva, N.P.; Craker, L.E. Medicinal plants used in Traditional medicine of the Caucasus and North America. J. Med. Act. Plants 2015, 4, 42–66. [Google Scholar] [CrossRef]
  5. Ibadullayeva, S.J.; Mamedova, S.E.; Sultanova, Z.R.; Movsumova, N.V.; Jafarli, I.A. Medicinal plants of Azerbaijan flora used in the treatment of certain diseases. Afr. J. Pharm. Pharmacol. 2010, 4, 545–548. [Google Scholar] [CrossRef]
  6. Miraldi, E.; Ferri, S.; Mostaghimi, V. Botanical drugs and preparations in the traditional medicine of West Azerbaijan (Iran). J. Ethnopharmacol. 2001, 75, 77–87. [Google Scholar] [CrossRef]
  7. The Plant List. Available online: http://www.theplantlist.org/1.1/browse/A/Lamiaceae/Dracocephalum/#statistics (accessed on 14 August 2022).
  8. Song, E.; Choi, J.; Gwon, H.; Lee, K.Y.; Choi, S.G.; Islam, M.A.; Chun, J.; Hwang, J. Phytochemical profile and antioxidant activity of Dracocephalum moldavica L. seed extracts using different extraction methods. Food Chem. 2021, 350, 128531. [Google Scholar] [CrossRef] [PubMed]
  9. Zeng, Q.; Jin, H.-Z.; Qin, J.-J.; Fu, J.-J.; Hu, X.-J.; Liu, J.-H.; Yan, L.; Chen, M.; Zhang, W.-D. Chemical constituents of plants from the genus Dracocephalum. Chem. Biodiv. 2010, 7, 1911–1929. [Google Scholar] [CrossRef]
  10. Sadraei, H.; Asghari, G.; Khanabadi, M.; Minaiyan, M. Anti-inflammatory effect of apigenin and hydroalcoholic extract of Dracocephalum kotschyi on acetic acid-induced colitis in rats. Res. Pharm. Sci. 2017, 12, 322–329. [Google Scholar] [CrossRef]
  11. Stappen, I.; Wanner, J.; Tabanca, N.; Wedge, D.E.; Ali, A.; Kaul, V.K.; Lal, B.; Jaitak, V.; Gochev, V.K.; Schmidt, E.; et al. Chemical composition and biological activity of essential oils of Dracocephalum heterophyllum and Hyssopus officinalis from Western Himalaya. Nat. Prod. Commun. 2015, 10, 133–138. [Google Scholar] [CrossRef]
  12. Moradi, H.; Ghavam, M.; Tavili, A. Study of antioxidant activity and some herbal compounds of Dracocephalum kotschyi Boiss. in different ages of growth. Biotechnol. Rep. 2020, 25, e00408. [Google Scholar] [CrossRef]
  13. Galushko, A.I. The Flora of the North Caucasus; Rostov University Publishing House: Rostov-on-Don, Russia, 1980; Volume I, pp. 37–38. [Google Scholar]
  14. Komarov, V.L. Flora of the U.S.S.R.; AN U.S.S.R.: Moscow, Russia, 1954; Volume XX, pp. 444–473. [Google Scholar]
  15. Qashqai, K. Azerbaijani Cuisine; Olma Media Group: Moscow, Russia, 2011; Volume I, pp. 97–100. [Google Scholar]
  16. Rumanovskaya, E.Y. Azerbaijani Cuisine; Dilya: St. Petersburg, Russia, 2001; Volume I, p. 134. [Google Scholar]
  17. Sokolov, P.D. Plant Resources of the U.S.S.R.; Nauka: St. Petersburg, Russia, 1991; Volume VI, pp. 19–24. [Google Scholar]
  18. Azimova, S.S.; Glushenkova, A.I. Lipids, Lipophilic Components and Essential Oils from Plant Sources; Springer: Berlin/Heidelberg, Germany, 2012; Volume I, pp. 403–404. [Google Scholar] [CrossRef]
  19. Barrero, A.F.; Herrador, M.M.; Arteaga, P.; Catalán, J.V. Germacrone: Occurrence, synthesis, chemical transformations and biological properties. Nat. Prod. Commun. 2008, 3, 567–576. [Google Scholar] [CrossRef]
  20. Hu, Z.; Wang, J.; Jin, L.; Duan, Y.; Zhang, X.; Sun, J.; Zhou, W.; Li, G. Isolation and structural characterization of two polysaccharides from Dracocephalum moldavica and their anti-complementary activity. Chem. Biodiv. 2022, 19, e202200294. [Google Scholar] [CrossRef]
  21. Dastmalchi, K.; Dorman, H.J.D.; Koşar, M.; Hiltunen, R. Chemical composition and in vitro antioxidant evaluation of a water-soluble Moldavian balm (Dracocephalum moldavica L.) extract. LWT 2007, 40, 239–248. [Google Scholar] [CrossRef]
  22. Kamali, M.; Khosroyar, S.; Kamali, H.; Ahmadzadeh, S.T.; Mohammadi, A. Phytochemical screening and evaluation of antioxidant activities of Dracocephalum kotschyi and determination of its luteolin content. Avic. J. Phytomed. 2016, 6, 425–433, 27516983. [Google Scholar]
  23. Weremczuk-Jeżyna, I.; Grzegorczyk-Karolak, I.; Frydrych, B.; Królicka, A.; Wysokińska, H. Hairy roots of Dracocephalum moldavica: Rosmarinic acid content and antioxidant potential. Acta Physiol. Plant. 2013, 35, 2095–2103. [Google Scholar] [CrossRef]
  24. Olennikov, D.N.; Chirikova, N.K.; Kashchenko, N.I.; Gornostai, T.G.; Selyutina, I.Y.; Zilfikarov, I.N. Effect of low temperature cultivation on the phytochemical profile and bioactivity of Arctic plants: A case of Dracocephalum palmatum. Int. J. Mol. Sci. 2017, 18, 2579. [Google Scholar] [CrossRef] [PubMed]
  25. Olennikov, D.N.; Chirikova, N.K.; Okhlopkova, Z.M.; Zulfugarov, I.S. Chemical composition and antioxidant activity of Tánara Ótó (Dracocephalum palmatum Stephan), a medicinal plant used by the North-Yakutian nomads. Molecules 2013, 18, 14105–14121. [Google Scholar] [CrossRef]
  26. Olennikov, D.N.; Chirikova, N.K. Dracopalmaside, a new flavonoid from Dracocephalum palmatum. Chem. Nat. Comp. 2015, 51, 1067–1069. [Google Scholar] [CrossRef]
  27. Olennikov, D.N.; Chirikova, N.K.; Kim, E.; Kim, S.W.; Zul′fugarov, I.S. New glycosides of eriodictyol from Dracocephalum palmatum. Chem. Nat. Comp. 2018, 54, 860–863. [Google Scholar] [CrossRef]
  28. Masuda, T.; Fujita, N.; Odaka, Y.; Takeda, Y.; Yonemori, S.; Nakamoto, K.; Kuninaga, H. Tyrosinase inhibitory activity of ethanol extracts from medicinal and edible plants cultivated in Okinawa and identification of a water-soluble inhibitor from the leaves of Nandina domestica. Biosci. Biotechnol. Biochem. 2007, 71, 2316–2320. [Google Scholar] [CrossRef] [PubMed]
  29. Lu, Y.; Foo, L.Y. Polyphenolics of Salvia—A review. Phytochemistry 2002, 59, 117–140. [Google Scholar] [CrossRef]
  30. Guo, Z.; Liang, X.; Xie, Y. Qualitative and quantitative analysis on the chemical constituents in Orthosiphon stamineus Benth. using ultra high-performance liquid chromatography coupled with electrospray ionization tandem mass spectrometry. J. Pharm. Biomed. Anal. 2019, 164, 135–147. [Google Scholar] [CrossRef] [PubMed]
  31. Dai, L.M.; Zhao, C.C.; Jin, H.-Z.; Tang, J.; Shen, Y.-H.; Li, H.-L.; Peng, C.-Y.; Zhang, W.-D. A new ferulic acid ester and other constituents from Dracocephalum peregrinum. Arch. Pharm. Res. 2008, 31, 1325–1329. [Google Scholar] [CrossRef] [PubMed]
  32. Kakasy, A.; Füzfai, Z.; Kursinszki, L.; Molnár-Perl, I.; Lemberkovics, É. Analysis of non-volatile constituents in Dracocephalum species by HPLC and GC-MS. Chromatographia 2006, 63, S17–S22. [Google Scholar] [CrossRef]
  33. Yang, X.; Liu, Y.; Chen, T.; Wang, N.; Li, H.; Zhao, J.; Li, Y. HSCCC Separation of three main compounds from the crude extract of Dracocephalum tanguticum by using dimethyl sulfoxide as cosolvent. J. Chromatogr. Sci. 2021, 59, 175–181. [Google Scholar] [CrossRef]
  34. Fattahi, M.; Nazeri, V.; Torras-Claveria, L.; Sefidkon, F.; Cusido, R.M.; Zamani, Z.; Palazon, J. A new biotechnological source of rosmarinic acid and surface flavonoids: Hairy root cultures of Dracocephalum kotschyi Boiss. Ind. Crops Prod. 2013, 50, 256–263. [Google Scholar] [CrossRef]
  35. Weremczuk-Jeżyna, I.; Skała, E.; Olszewska, M.A.; Kiss, A.K.; Balcerczak, E.; Wysokińska, H.; Kicel, A. The identification and quantitative determination of rosmarinic acid and salvianolic acid B in hairy root cultures of Dracocephalum forrestii W.W. Smith. Ind. Crops Prod. 2016, 91, 125–131. [Google Scholar] [CrossRef]
  36. Weremczuk-Jeżyna, I.; Kuźma, Ł.; Kiss, A.K.; Grzegorczyk-Karolak, I. Effect of cytokinins on shoots proliferation and rosmarinic and salvianolic acid B production in shoot culture of Dracocephalum forrestii W. W. Smith. Acta Physiol. Plant. 2018, 40, 189. [Google Scholar] [CrossRef]
  37. Kashchenko, N.I.; Olennikov, D.N. Glycosides of rosmarinic acid from Nepeta multifida. Chem Nat Comp. 2022, 58, 274–278. [Google Scholar] [CrossRef]
  38. Matsuta, M.; Kanita, R.; Saito, Y.; Yamashita, A. The 3α-hydroxysteroid dehydrogenase inhibitory active flavonoids and phenylpropanoids from Schizonepeta spikes. Nat. Med. 1996, 50, 204–211. [Google Scholar]
  39. Olennikov, D.N. New benzofuran lignans from Nepeta multifida. Chem. Nat. Comp. 2021, 57, 818–822. [Google Scholar] [CrossRef]
  40. Saeidnia, S.; Gohari, A.R.; Ito, M.; Kiuchi, F.; Honda, G. Bioactive constituents from Dracocephalum subcapitatum (O. Kuntze) Lipsky. Z. Für Nat. C 2005, 60, 22–24. [Google Scholar] [CrossRef] [PubMed]
  41. Ren, D.M.; Qu, Z.; Wang, X.N.; Shi, J.; Lou, H.X. Simultaneous determination of nine major active compounds in Dracocephalum rupestre by HPLC. J. Pharm. Biomed. Anal. 2008, 48, 1441–1445. [Google Scholar] [CrossRef] [PubMed]
  42. Wang, S.Q.; Han, X.Z.; Li, X.; Ren, D.M.; Wang, X.N.; Lou, H.X. Flavonoids from Dracocephalum tanguticum and their cardioprotective effects against doxorubicin-induced toxicity in H9c2 cells. Bioorg. Med. Chem. Lett. 2010, 20, 6411–6415. [Google Scholar] [CrossRef] [PubMed]
  43. Ren, D.M.; Lou, H.X.; Ma, B.; Ji, M. Determination of luteolin-7-O-glycoside in the herb of Dracocephalum rupestra by HPLC. China J. Chin. Mater. Med. 2004, 29, 860–862. [Google Scholar]
  44. Gohari, A.R.; Saeidnia, S.; Matsuo, K.; Uchiyama, N.; Yagura, T.; Ito, M.; Kiuchi, F.; Honda, G. Flavonoid constituents of Dracocephalum kotschyi growing in Iran and their trypanocidal activity. J. Nat. Med. 2003, 57, 250–252. [Google Scholar]
  45. Litvinenko, V.I.; Sergienko, T.A. Dracocephaloside—A new flavonoid glycoside from Dracocephalum thymiflorum L. Chem. Nat. Comp. 1965, 1, 108–109. [Google Scholar] [CrossRef]
  46. Shamyrina, A.A.; Peshkova, V.A.; Shergina, N.I. Dracocephaloside from Dracocephalum nutans. Chem. Nat. Comp. 1978, 14, 688. [Google Scholar] [CrossRef]
  47. Lou, H.; Jing, X.; Ren, D.; Wei, X.; Zhang, X. Eriodictyol protects against H2O2-induced neuron-like PC12 cell death through activation of Nrf2/ARE signaling pathway. Neurochem. Int. 2012, 61, 251–257. [Google Scholar] [CrossRef] [PubMed]
  48. Li, S.-M.; Yang, X.-W.; Li, Y.-L.; Shen, Y.-H.; Feng, L.; Wang, Y.-H.; Zeng, H.-W.; Liu, X.-H.; Zhang, C.-S.; Long, C.-L.; et al. Chemical constituents of Dracocephalum forrestii. Planta Med. 2009, 75, 1591–1596. [Google Scholar] [CrossRef]
  49. Gu, H.F.; Chen, R.Y.; Sun, Y.H.; Liu, F. Studies on chemical constituents from herb of Dracocephalum moldavica. China J. Chin. Mater. Med. 2004, 29, 232–234, 15706847. [Google Scholar]
  50. Pouraboli, I.; Nazari, S.; Sabet, N.; Sharififar, F.; Jafari, M. Antidiabetic, antioxidant, and antilipid peroxidative activities of Dracocephalum polychaetum shoot extract in streptozotocin-induced diabetic rats: In vivo and in vitro studies. Pharm. Biol. 2016, 54, 272–278. [Google Scholar] [CrossRef]
  51. Ashrafian, S.; Farimani, M.M.; Sonboli, A.; Ashrafian, H.; Kabiri, H.; Rezadoost, H. Simultaneous characterization of nine isolated flavonoids in Iranian Dracocephalum species and in silico study of their inhibitory properties against MTH1 enzyme. S. Afr. J. Bot. 2022, 146, 254–261. [Google Scholar] [CrossRef]
  52. Shi, Q.Q.; Dang, J.; Wen, H.X.; Yuan, X.; Tao, Y.-D.; Wang, Q.-L. Anti-hepatitis, antioxidant activities and bioactive compounds of Dracocephalum heterophyllum extracts. Bot. Stud. 2016, 57, 16. [Google Scholar] [CrossRef]
  53. Heydari, P.; Yavari, M.; Adibi, P.; Asghari, G.; Ghanadian, S.M.; Dida, G.O.; Khamesipour, F. Medicinal properties and active constituents of Dracocephalum kotschyi and its significance in Iran: A systematic review. Evid. Based Complement. Alternat. Med. 2019, 2019, 9465309. [Google Scholar] [CrossRef]
  54. Chen, T.; Yang, X.; Wang, S.; Cheng, S.; Li, H.; Wei, Y.; Yan, S.; Song, Z.; Yang, F.; Liu, Y.; et al. Separation of five flavone glycosides including two groups with similar polarities from Dracocephalum tanguticum by a combination of three high-speed counter-current chromatography modes. J. Sep. Sci. 2022, 45, 468–476. [Google Scholar] [CrossRef]
  55. Yang, L.-N.; Xing, J.-G.; He, C.-H.; Wu, T.; Wu, T. The phenolic compounds from Dracocephalum moldavica L. Biochem. Syst. Ecol. 2014, 54, 19–22. [Google Scholar] [CrossRef]
  56. Olennikov, D.N.; Kashchenko, N.I. New acylated apigenin glycosides from edge flowers of Matricaria chamomilla. Chem. Nat. Comp. 2016, 52, 996–999. [Google Scholar] [CrossRef]
  57. Selenge, E.; Murata, T.; Tanaka, S.; Sasaki, K.; Batkhuu, J.; Yoshizaki, F. Monoterpene glycosides, phenylpropanoids, and acacetin glycosides from Dracocephalum foetidum. Phytochemistry 2014, 101, 91–100. [Google Scholar] [CrossRef]
  58. Li, Q.; Liu, Y.; Han, L.; Liu, J.; Liu, W.; Feng, F.; Zhang, J.; Xie, N. Chemical constituents and quality control of two Dracocephalum species based on high-performance liquid chromatographic fingerprints coupled with tandem mass spectrometry and chemometrics. J. Sep. Sci. 2016, 39, 4071–4085. [Google Scholar] [CrossRef] [PubMed]
  59. Kumar, S.; Pandey, A.K. Chemistry and biological activities of flavonoids: An overview. Sci. World J. 2013, 2013, 162750. [Google Scholar] [CrossRef]
  60. Ullah, A.; Munir, S.; Badshah, S.L.; Khan, N.; Ghani, L.; Poulson, B.G.; Emwas, A.H.; Jaremko, M. Important flavonoids and their role as a therapeutic agent. Molecules 2020, 25, 5243. [Google Scholar] [CrossRef] [PubMed]
  61. Olennikov, D.N.; Tankhaeva, L.M.; Stolbikova, A.V.; Petrov, E.V. Phenylpropanoids and polysaccharides from Plantago depressa and P. media growing in Buryatia. Chem. Nat. Comp. 2011, 47, 165–169. [Google Scholar] [CrossRef]
  62. Sumner, L.W.; Amberg, A.; Barrett, D.; Beale, M.H.; Beger, R.; Daykin, C.A.; Fan, T.W.-M.; Fiehn, O.; Goodacre, R.; Griffin, J.L.; et al. Proposed minimum reporting standards for chemical analysis Chemical Analysis Working Group (CAWG) Metabolomics Standards Initiative (MSI). Metabolomics 2007, 3, 211–221. [Google Scholar] [CrossRef] [PubMed]
  63. Synytsya, A.; Čopíková, J.; Matéjka, P.; Machovic, V. Fourier transform Raman and infrared spectroscopy of pectins. Carbohydr. Polym. 2003, 54, 97–106. [Google Scholar] [CrossRef]
  64. Čopíková, J.; Černá, M.; Novotná, M.; Kaasová, J.; Synytsya, A. Application of FT-IR spectroscopy in detection of food hydrocolloids in confectionery jellies and food supplements. Czech J. Food Sci. 2001, 19, 51–56. [Google Scholar] [CrossRef]
  65. Zhao, F.; Chen, Y.P.; Salmaki, Y.; Drew, B.T.; Wilson, T.C.; Scheen, A.C.; Celep, F.; Bräuchler, C.; Bendiksby, M.; Wang, Q.; et al. An updated tribal classification of Lamiaceae based on plastome phylogenomics. BMC Biol. 2021, 19, 2. [Google Scholar] [CrossRef]
  66. Yang, X.; Lv, Y.; Tian, L.; Zhao, Y. Composition and systemic immune activity of the polysaccharides from an herbal tea (Lycopus lucidus Turcz). J. Agricult. Food Chem. 2010, 58, 6075–6080. [Google Scholar] [CrossRef]
  67. Olennikov, D.N.; Tankhaeva, L.M. Lamiaceae carbohydrates. I. Pectinic substances and hemicelluloses from Mentha × piperita. Chem. Nat. Comp. 2007, 43, 501–507. [Google Scholar] [CrossRef]
  68. Olennikov, D.N.; Tankhaeva, L.M. Lamiaceae carbohydrates. II. Water-soluble polysaccharides from Mentha × piperita. Chem. Nat. Comp. 2007, 43, 648–651. [Google Scholar] [CrossRef]
  69. Olennikov, D.N.; Tankhaeva, L.M. Lamiaceae carbohydrates. III. Water-soluble polysaccharides from Origanum vulgare. Chem. Nat. Comp. 2008, 44, 630–631. [Google Scholar] [CrossRef]
  70. Feng, L.; Jia, X.-B.; Shi, F.; Chen, Y. Identification of two polysaccharides from Prunella vulgaris L. and evaluation on their anti-lung adenocarcinoma activity. Molecules 2010, 15, 5093–5103. [Google Scholar] [CrossRef]
  71. De Almeida Gonçalves, G.; de Sá-Nakanishi, A.B.; Comar, J.F.; Bracht, L.; Dias, M.I.; Barros, L.; Bracht, A. Water soluble compounds of Rosmarinus officinalis L. improve the oxidative and inflammatory states of rats with adjuvant-induced arthritis. Food Funct. 2008, 9, 2328–2340. [Google Scholar] [CrossRef] [PubMed]
  72. Capek, P.; Hříbalová, V.; Švandová, E.; Ebringerová, A.; Sasinková, V.; Masarová, J. Characterization of immunomodulatory polysaccharides from Salvia officinalis L. Int. J. Biol. Macromol. 2003, 33, 113–119. [Google Scholar] [CrossRef]
  73. Chun, H.; Jun, W.J.; Shin, D.H.; Hong, B.S.; Cho, H.Y.; Yang, H.C. Purification and characterization of anti-complementary polysaccharide from leaves of Thymus vulgaris L. Chem. Pharm. Bull. 2001, 49, 762–764. [Google Scholar] [CrossRef] [PubMed]
  74. Mnich, E.; Bjarnholt, N.; Eudes, A.; Harholt, J.; Holland, C.; Jørgensen, B.; Larsen, F.H.; Liu, M.; Manat, R.; Meyer, A.S.; et al. Phenolic cross-links: Building and de-constructing the plant cell wall. Nat. Prod. Rep. 2020, 37, 919–961. [Google Scholar] [CrossRef]
  75. Siemińska-Kuczer, A.; Szymańska-Chargot, M.; Zdunek, A. Recent advances in interactions between polyphenols and plant cell wall polysaccharides as studied using an adsorption technique. Food Chem. 2022, 30, 131487. [Google Scholar] [CrossRef] [PubMed]
  76. Olennikov, D.N.; Kashchenko, N.I.; Chirikova, N.K.; Vasil’eva, A.G.; Gadimli, A.I.; Isaev, J.I.; Vennos, C. Caffeoylquinic acids and flavonoids of fringed sagewort (Artemisia frigida Willd.): HPLC-DAD-ESI-QQQ-MS Profile, HPLC-DAD quantification, in vitro digestion stability, and antioxidant capacity. Antioxidants 2019, 8, 307. [Google Scholar] [CrossRef]
  77. Lakey-Beitia, J.; Burillo, A.M.; La Penna, G.; Hegde, M.L.; Rao, K.S. Polyphenols as potential metal chelation compounds against Alzheimer’s disease. J. Alzheimers. Dis. 2021, 82, S335–S357. [Google Scholar] [CrossRef] [PubMed]
  78. Olennikov, D.N.; Chirikova, N.K.; Tankhaeva, L.M. Phenolic compounds of Scutellaria baicalensis Georgi. Russ. J. Bioorg. Chem. 2010, 36, 816–824. [Google Scholar] [CrossRef]
  79. Gulcin, İ. Antioxidants and antioxidant methods: An updated overview. Arch. Toxicol. 2020, 94, 651–715. [Google Scholar] [CrossRef] [PubMed]
  80. Zhu, C.-S.; Liu, K.; Wang, J.-L.; Li, J.-F.; Liu, M.-F.; Hao, N.; Lin, Y.-X.; Xiao, Z.-F. Antioxidant activities and hepatoprotective potential of Dracocephalum rupestre Hance extract against CCl4-induced hepatotoxicity in Kunming mice. J. Food Biochem. 2017, 42, e12484. [Google Scholar] [CrossRef]
  81. Olennikov, D.N.; Chirikova, N.K.; Kashchenko, N.I.; Nikolaev, V.M.; Kim, S.W.; Vennos, C. Bioactive phenolics of the genus Artemisia (Asteraceae): HPLC-DAD-ESI-TQ-MS/MS profile of the Siberian species and their inhibitory potential against α-amylase and α-glucosidase. Front. Pharmacol. 2018, 9, 756. [Google Scholar] [CrossRef]
  82. Sevag, M.G.; Lackman, D.B.; Smolens, J. The isolation of the components of Streptococcal nucleoproteins in serologically active form. J. Biol. Chem. 1938, 124, 425–436. [Google Scholar] [CrossRef]
  83. Olennikov, D.N.; Tankhaeva, L.M.; Samuelsen, A.B. Quantitative analysis of polysaccharides from Plantago major using the Dreywood method. Chem. Nat. Comp. 2006, 42, 265–268. [Google Scholar] [CrossRef]
  84. Usov, A.T.; Bilan, M.I.; Klochkova, N.G. Polysaccharides of algae. 48. Polysaccharide composition of several calcareous red algae: Isolation of alginate from Corallina pilulitara P. et R. (Rhodophyta, Corallinaceae). Bot. Mar. 1995, 35, 43–51. [Google Scholar] [CrossRef]
  85. Bradford, M.M. A rapid and sensitive method for the quantification of microgram quantities of protein utilizing the principle of protein-dye binding. Anal. Biochem. 1976, 76, 248–254. [Google Scholar] [CrossRef]
  86. Chew, Y.-L.; Goh, J.-K.; Lim, Y.-Y. Assessment of in vitro antioxidant capacity and polyphenolic composition of selected medicinal herbs from Leguminosae family in Peninsular Malaysia. Food Chem. 2009, 116, 13–18. [Google Scholar] [CrossRef]
  87. Olennikov, D.N.; Stolbikova, A.V.; Rokhin, A.V.; Khobrakova, V.B.; Tankhaeva, L.M. Polysaccharides from Fabaceae. V. α-Glucan from Sophora flavescens roots. Chem. Nat. Comp. 2011, 47, 1–6. [Google Scholar] [CrossRef]
  88. Olennikov, D.N.; Tankhaeva, L.M. A quantitative assay for total fructans in burdock (Arctium spp.) roots. Russ. J. Bioorg. Chem. 2011, 37, 893–898. [Google Scholar] [CrossRef]
  89. Togola, A.; Inngjerdingen, M.; Diallo, D.; Barsett, H.; Rolstad, B. Polysaccharides with complement fixing and macrophage stimulation activity from Opilia celtidifolia, isolation and partial characterization. J. Ethnopharmacol. 2007, 115, 423–431. [Google Scholar] [CrossRef] [PubMed]
  90. Olennikov, D.N.; Kirillina, C.S.; Chirikova, N.K. Water-soluble melanoidin pigment as a new antioxidant component of fermented willowherb leaves (Epilobium angustifolium). Antioxidants 2021, 10, 1300. [Google Scholar] [CrossRef] [PubMed]
  91. Olennikov, D.N.; Chirikova, N.K.; Vasilieva, A.G.; Fedorov, I.A. LC-MS profile, gastrointestinal and gut microbiota stability and antioxidant activity of Rhodiola rosea herb metabolites: A comparative study with subterranean organs. Antioxidants 2020, 9, 526. [Google Scholar] [CrossRef] [PubMed]
  92. Olennikov, D.N.; Kashchenko, N.I.; Chirikova, N.K. A novel HPLC-assisted method for investigation of the Fe2+-chelating activity of flavonoids and plant extracts. Molecules 2014, 19, 18296–18316. [Google Scholar] [CrossRef]
Figure 1. Dracocephalum austriacum (a) and D. botryoides (b) in their natural habitat in the flowering stage (Azerbaijan, Gryz, Guba region).
Figure 1. Dracocephalum austriacum (a) and D. botryoides (b) in their natural habitat in the flowering stage (Azerbaijan, Gryz, Guba region).
Plants 11 02126 g001
Figure 2. High-performance liquid chromatography with photodiode array detection (HPLC-PDA) chromatograms (270 nm) of herb and root extracts of Dracocephalum austriacum (DA). Compounds are numbered as listed in Table 1. IS—3′,4′-di-O-acetyl-cis-khellactone (5 μg/mL).
Figure 2. High-performance liquid chromatography with photodiode array detection (HPLC-PDA) chromatograms (270 nm) of herb and root extracts of Dracocephalum austriacum (DA). Compounds are numbered as listed in Table 1. IS—3′,4′-di-O-acetyl-cis-khellactone (5 μg/mL).
Plants 11 02126 g002
Figure 3. High-performance liquid chromatography with photodiode array detection (HPLC-PDA) chromatograms (270 nm) of herb and root extracts of Dracocephalum botryoides (DB). Compounds are numbered as listed in Table 1. IS—3′,4′-di-O-acetyl-cis-khellactone (5 μg/mL).
Figure 3. High-performance liquid chromatography with photodiode array detection (HPLC-PDA) chromatograms (270 nm) of herb and root extracts of Dracocephalum botryoides (DB). Compounds are numbered as listed in Table 1. IS—3′,4′-di-O-acetyl-cis-khellactone (5 μg/mL).
Plants 11 02126 g003
Figure 4. FTIR spectra of water-soluble polysaccharides of herbs and roots of D. austriacum and D. botryoides.
Figure 4. FTIR spectra of water-soluble polysaccharides of herbs and roots of D. austriacum and D. botryoides.
Plants 11 02126 g004
Figure 5. UV-Vis spectra of water-soluble polysaccharides of herbs and roots of D. austriacum and D. botryoides.
Figure 5. UV-Vis spectra of water-soluble polysaccharides of herbs and roots of D. austriacum and D. botryoides.
Plants 11 02126 g005
Table 2. Characteristics of water-soluble polysaccharides of D. austriacum and D. botryoides herb and roots.
Table 2. Characteristics of water-soluble polysaccharides of D. austriacum and D. botryoides herb and roots.
ParameterWSPS of
D. austriacum Herb
WSPS of
D. austriacum Roots
WSPS of
D. botryoides Herb
WSPS of
D. botryoides Roots
Yield, % 11.20.90.70.6
Carbohydrate content, % 290.5 ± 2.792.8 ± 2.488.3 ± 2.592.1 ± 2.8
Uronic acid content, % 249.2 ± 1.912.0 ± 0.425.3 ± 0.911.2 ± 0.4
Protein content, % 21.8 ± 0.01.7 ± 0.01.4 ± 0.01.0 ± 0.0
Phenolic content, % 23.3 ± 0.11.4 ± 0.04.0 ± 0.14.3 ± 0.1
Iodine reactionnegativepositivenegativepositive
Resorcinol reactionnegativenegativenegativenegative
Fehling’s reactionnegativenegativenegativenegative
β-Glucosyl Yariv reactionpositivepositivepositivepositive
Monosaccharide composition:
Arabinose, mol%12.121.327.425.5
Galactose, mol%16.224.120.523.9
Glucose, mol%6.322.613.425.6
Fucose, mol%0.61.80.41.1
Mannose, mol%3.08.34.47.7
Ribose, mol%0.00.00.00.0
Rhamnose, mol%10.86.48.13.8
Xylose, mol%0.50.10.40.1
Galacturonic acid, mol%48.012.623.610.0
Glucuronic acid, mol%2.42.91.72.4
1 Percentage of dry plant weight. 2 Percentage of polysaccharide fraction weight.
Table 3. Phenolic compounds released after alkaline hydrolysis of water-soluble polysaccharides of herbs and roots of D. austriacum and D. botryoides.
Table 3. Phenolic compounds released after alkaline hydrolysis of water-soluble polysaccharides of herbs and roots of D. austriacum and D. botryoides.
CompoundPercentage of Compound, % of Total Phenolic Content
D. austriacum HerbD. austriacum RootsD. botryoides HerbD. botryoides Roots
Benzoic acid1.2 ± 0.02.5 ± 0.10.2 ± 0.00.1 ± 0.0
p-Hydroxybenzoic acid1.1 ± 0.00.8 ± 0.00.1 ± 0.00.1 ± 0.0
p-Methoxybenzoic acid19.3 ± 0.422.4 ± 0.531.1 ± 0.625.7 ± 0.5
Protocatechuic acid0.5 ± 0.00.2 ± 0.00.1 ± 0.00.3 ± 0.0
Gentisic acid0.2 ± 0.00.5 ± 0.00.5 ± 0.00.3 ± 0.0
Vanillic acid18.6 ± 0.419.3 ± 0.425.7 ± 0.528.3 ± 0.6
Isovanillic acid2.5 ± 0.01.4 ± 0.01.1 ± 0.00.9 ± 0.0
Veratric acid7.2 ± 0.19.3 ± 0.214.7 ± 0.319.7 ± 0.4
Syringic acid0.2 ± 0.03.4 ± 0.10.6 ± 0.01.4 ± 0.0
Anisic aldehyde6.2 ± 0.17.2 ± 0.13.1 ± 0.12.2 ± 0.0
Vanillin8.4 ± 0.111.6 ± 0.211.6 ± 0.210.3 ± 0.02
Cinnamic acid2.6 ± 0.11.1 ± 0.00.3 ± 0.00.9 ± 0.0
p-Coumaric acid15.2 ± 0.210.7 ± 0.22.5 ± 0.11.4 ± 0.0
p-Methoxycinnamic acid9.2 ± 0.25.1 ± 0.13.9 ± 0.16.2 ± 0.1
Caffeic acid0.7 ± 0.00.1 ± 0.00.0 ± 0.00.0 ± 0.0
Ferulic acid3.8 ± 0.13.9 ± 0.13.6 ± 0.11.9 ± 0.0
Isoferulic acid1.9 ± 0.00.4 ± 0.00.8 ± 0.00.2 ± 0.0
Table 4. Bioactivity of extracts of D. austriacum and D. botryoides, selected phenolics and water-soluble polysaccharides (WSPS) in five antioxidant assays a.
Table 4. Bioactivity of extracts of D. austriacum and D. botryoides, selected phenolics and water-soluble polysaccharides (WSPS) in five antioxidant assays a.
ObjectDPPHbABTS+ bOH• bO2− bFeCA c
D. austriacum herb extract37.15 ± 0.74 viii35.81 ± 0.88 v125.67 ± 3.79 vii108.26 ± 4.32 ix0.42 ± 0.02 iii
D. austriacum root extract35.19 ± 0.71 viii29.64 ± 0.72 iv139.09 ± 4.19 viii99.14 ± 3.82 ix0.38 ± 0.01 ii
D. botryoides herb extract28.63 ± 0.54 vii25.20 ± 0.50 iv103.28 ± 3.09 v85.67 ± 3.45 vii0.51 ± 0.02 iv
D. botryoides root extract40.67 ± 0.82 ix36.14 ± 0.93 v164.11 ± 4.99 ix135.24 ± 5.44 x0.27 ± 0.01 i
5-O-Caffeoylquinic acid7.63 ± 0.15 iii9.04 ± 0.17 iii68.25 ± 1.73 ii49.11 ± 1.47 ii2.28 ± 0.09 vii
Luteolin 7-O-glucoside9.20 ± 0.18 iv8.26 ± 0.15 ii53.29 ± 1.33 i60.83 ± 1.83 iv2.84 ± 0.11 ix
Luteolin 7-O-(6″-acetyl)-glucoside10.27 ± 0.22 v9.63 ± 0.20 iii55.73 ± 1.30 i67.09 ± 2.01 v2.42 ± 0.10 vii
Luteolin 4′-O-glucoside45.60 ± 0.87 x67.19 ± 1.28 vi123.14 ± 2.97 vii170.81 ± 5.18 xi1.65 ± 0.06 vi
Eriodictyol 7-O-glucoside12.04 ± 0.25 vi7.39 ± 0.14 i82.17 ± 2.05 iii53.02 ± 1.63 iii3.18 ± 0.12 ix
Rosmarinic acid6.22 ± 0.14 i25.73 ± 0.52 iv95.16 ± 2.47 iv15.38 ± 0.42 i3.72 ± 0.15 x
Lithospermic acid B9.08 ± 0.17 iv36.28 ± 0.71 v105.61 ± 2.50 v16.27 ± 0.49 i2.87 ± 0.12 ix
D. austriacum herb WSPS189.42 ± 3.78 xiii150.03 ± 3.75 ix197.83 ± 5.90 x89.57 ± 3.11 vii4.31 ± 0.18 xi
D. austriacum root WSPS>500214.53 ± 5.35 x251.12 ± 7.53 xi129.11 ± 4.56 x1.19 ± 0.04 v
D. botryoides herb WSPS141.15 ± 2.80 xii105.63 ± 2.67 viii129.63 ± 3.80 vii82.56 ± 2.88 vii2.39 ± 0.09 vii
D. botryoides root WSPS102.85 ± 2.07 xi82.79 ± 2.06 vii105.73 ± 3.14 v73.68 ± 2.57 vi1.10 ± 0.05 v
Trolox d7.03 ± 0.14 ii3.44 ± 0.69 v108.69 ± 0.04 v, vi165.14 ± 4.95 xi1.53 ± 0.06 vi
a DPPH—2,2-diphenyl-1-picrylhydrazyl radical scavenging capacity; ABTS+—2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) cation radical scavenging capacity; O2—superoxide radical scavenging capacity; OH—hydroxyl radical scavenging capacity; FeCA—Fe2+-ions chelating activity. b IC50, μg/mL. c mM Fe2+-ions/g. d Reference compound. Averages ± standard deviation (S.D.) were obtained from five different experiments. Values with different numbers (i–xiii) in each column indicate statistically significant differences among groups at p < 0.05 by one-way ANOVA.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kashchenko, N.I.; Jafarova, G.S.; Isaev, J.I.; Olennikov, D.N.; Chirikova, N.K. Caucasian Dragonheads: Phenolic Compounds, Polysaccharides, and Bioactivity of Dracocephalum austriacum and Dracocephalum botryoides. Plants 2022, 11, 2126. https://doi.org/10.3390/plants11162126

AMA Style

Kashchenko NI, Jafarova GS, Isaev JI, Olennikov DN, Chirikova NK. Caucasian Dragonheads: Phenolic Compounds, Polysaccharides, and Bioactivity of Dracocephalum austriacum and Dracocephalum botryoides. Plants. 2022; 11(16):2126. https://doi.org/10.3390/plants11162126

Chicago/Turabian Style

Kashchenko, Nina I., Gunay S. Jafarova, Javanshir I. Isaev, Daniil N. Olennikov, and Nadezhda K. Chirikova. 2022. "Caucasian Dragonheads: Phenolic Compounds, Polysaccharides, and Bioactivity of Dracocephalum austriacum and Dracocephalum botryoides" Plants 11, no. 16: 2126. https://doi.org/10.3390/plants11162126

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop