Next Article in Journal
Natural Antioxidants from Seeds and Their Application in Meat Products
Next Article in Special Issue
The Combined Effect of Mediterranean Shrubland Pasture and the Dietary Administration of Sage By-Products on the Antioxidant Status of Segureña Ewes and Lambs
Previous Article in Journal
The Sorbus spp.—Underutilised Plants for Foods and Nutraceuticals: Review on Polyphenolic Phytochemicals and Antioxidant Potential
Previous Article in Special Issue
Chemical Composition, Antimicrobial Properties of Siparuna guianensis Essential Oil and a Molecular Docking and Dynamics Molecular Study of its Major Chemical Constituent
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Health-Promoting Effects of Thymus Phenolic-Rich Extracts: Antioxidant, Anti-inflammatory and Antitumoral Properties

by
Andrea F. Afonso
1,2,
Olívia R. Pereira
3 and
Susana M. Cardoso
1,*
1
LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
2
Public Health Laboratory of Bragança, Local Health Unit, Rua Eng. Adelino Amaro da Costa, 5300-146 Bragança, Portugal
3
Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal
*
Author to whom correspondence should be addressed.
Antioxidants 2020, 9(9), 814; https://doi.org/10.3390/antiox9090814
Submission received: 7 August 2020 / Revised: 26 August 2020 / Accepted: 28 August 2020 / Published: 1 September 2020

Abstract

:
Thymus genus comprises numerous species that are particularly abundant in the West Mediterranean region. A growing body of evidence suggests that many of these species are a rich source of bioactive compounds, including phenolic compounds such as rosmarinic acid, salvianolic acids and luteolin glycosides, able to render them potential applications in a range of industrial fields. This review collects the most relevant studies focused on the antioxidant, anti-inflammatory and anti-cancer of phenolic-rich extracts from Thymus plants, highlighting correlations made by the authors with respect to the main phenolic players in such activities.

1. Introduction

Thymus is a Latin name of the plant genus, which is considered to derive from the Greek word thyo (perfume) by some authors, while others consider the Greek word thymos (courage, strength) [1]. Thymus genus belongs to Lamiaceae family and includes about 350 aromatic species that are distributed around the world, with particularly abundancy in the West Mediterranean region [2,3]. These species are perennial and are characterized as being herbaceous subshrubs or shrubs with 10 to 30 cm tall, containing small and simple leaves, ramified and prostrated branches and big clusters of pink, white, cream or violet flowers [2].
Thymus plants are claimed to be endowed of distinct health beneficial properties, including antimicrobial, antioxidant, anti-inflammatory, cardioprotective, neuroprotective, anticarcinogenic and hypoglycemic activities [4,5,6,7]. Although these benefits have been mostly associated with essential oils [8,9,10,11], nowadays, Thymus phenolic-rich extracts usually obtained with polar solvents, are an attractive target for the screening of bioactive compounds for possible industrial applications in distinct fields, including food, cosmetics or pharmaceutical industries [6,12]. Among them, counteraction of oxidative stress events is a central issue underlying the valorization of these plants.
Oxidative stress is generated when the balance between reactive species formation and their elimination is disrupted. Under this biological condition, there is an overproduction of reactive oxygen species (ROS) such as superoxide (O2-), hydroxyl radical (OH) and hydrogen peroxide (H2O2), as well as of reactive nitrogen species (RNS) like nitric oxide (NO) and peroxynitrite (ONOO¯), overall leading to several mitochondrial and cellular damages. These events are closely associated to aging processes [13,14] and to the physiopathology of several oxidative-related diseases [15,16,17]. To minimize the oxidative damage, enzymatic and non-enzymatic antioxidants are involved. The first include superoxide dismutase (SOD), which catalyzes the conversion of O2- to H2O2, which in its turn, can be neutralized by catalase or glutathione peroxidase (GPx), while non-enzymatic antioxidants enclose, among others, vitamins A, C and E, the glutathione reduced form (GSH) and β-carotene [16,17,18,19]. Oxidative stress also plays a critical role in the triggering and progression of inflammation, through the overproduction of ROS and RNS. In this process, the enzyme NO synthase (iNOS) is responsible for the production of NO, which is a key free radical in inflammatory events. Globally, these oxidative agents result from activation of immunological cells that can also generate proinflammatory agents [18,20], namely chemokines, interleukins (IL) and interferons (INF), which are majorly regulated by the transcriptional factor kappa B (NF-κB). The activation of adherent molecules by chemokines and complement split products is also triggered, enabling leukocytes migration into tissue spaces and the release of pro-inflammatory cytokines (e.g., IL-1β, IL-6, IL-12 and tumor necrosis factor (TNF-α)), that in turn stimulate and amplify inflammation [18,20,21]. Moreover, there is the activation of arachidonic acid, whose metabolism generates prostaglandins and tromboxanes by the cyclooxygenases pathway and leukotrienes by lipoxygenase pathway, stimulated by the enzymes cyclooxygenases (constitutive COX-1 and inducible COX-2) and by 5-lipoxygenase (5-LOX), respectively [22,23,24].
Notably, oxidative stress and chronic inflammation are also known to contribute to malignant transformation of cells associated with tumor cell proliferation and tumor progression, as well as angiogenesis and tissue remodeling, induction of neoplastic transformation and metastasis [16,25,26]. The cancerous state develops when the normal tissue homeostasis is broken, resulting in an imbalance between cellular proliferation and programmed cell death (apoptosis). It is known that apoptosis can be induced by activation of the caspase cascade through death receptors pathway or the mitochondrial death signaling, whose members are either proapoptotic (Bax, Bad) or antiapoptotic (Bcl-2, Bcl-XL) [27,28,29]. Therefore, the induction of programed cell death in cancer cells is one of the strategies used in the development of anti-cancer drugs [24,30,31,32] and, in particular, plants used in traditional forms have been the focus of attention as plant-based foods, whose regular consumption may be correlated with a lower incidence of cancer [33,34,35,36].
In recent years, numerous studies have reported the bioactivities of different species belonging to the Thymus genus. This review represents the first collection of data regarding the antioxidant, anti-inflammatory and antitumoral properties of phenolic-rich extracts from Thymus, highlighting its health-benefits and relevance as potential agents with pharmacological activity.

2. Antioxidant Activity

The antioxidant activity of Thymus species has been closely associated with their phenolic richness and/or specific phenolic composition [37] and hence, many authors have previously screened the antioxidant potential of polar extracts. The majority of such studies used one or several chemical methods, including the (i) scavenging of stable free radicals such as DPPH (2.2-diphenyl-1-picrylhydrazyl), ABTS●+ (2.2′-azino-bis-3-ethylbenzthiazoline-6-sulphonic acid), OH, O2- and NO; (ii) the reduction of metal ions from the higher to the lower oxidation state by reducing power method (RP) and ferric reducing antioxidant power assay (FRAP); (iii) the oxygen radical absorbance capacity (ORAC), which measures the radical chain breaking ability of antioxidants by the inhibition of peroxyl radical (RO2)–induced oxidation and trolox equivalence antioxidant capacity (TEAC); (iv) the inhibition of lipid peroxidation by ß-carotene bleaching test; and (v) the inhibition of lipid peroxidation by decreasing the thiobarbituric acid reactive substances (TBARS). The extensive use of such assays is mainly attributed to their simplicity and quickness of execution [38,39]. In general, these methods are accepted as a first approach to screen the antioxidant potential of the samples when used combined (because there is no universal method for antioxidant ability screening) and compared to positive controls [4,40,41,42,43,44]. Studies complying such criteria are summarized in Table 1.
Since Thymus vulgaris is a widespread species, its polar extracts have been explored for a range of biological effects, including antioxidant ability. Among the reported data for this species, water, methanolic and hydroalcoholic extracts of this plant origin were reported to have DPPH EC50 values in the range of 1.8 and 44.7 μg/mL [4,57,59]. Moreover, phenolic-rich extracts from T. vulgaris were shown to be good ABTS●+ (EC50 = 0.7–13.8 μg/mL) [57,59] and OH (EC50 = 0.24 μg/mL) [57] scavenging agents (Table 1).
The promising antioxidant potential of many others Thymus extracts were also previously emphasized by distinct authors. In particular, a methanolic extract of Thymus nummularius, also known as Thymus pseudopulegioides, was claimed to have higher DPPH scavenging ability (EC50 = 5.7 μg/mL) than 3,5-di-tert-butyl-4 hydroxytoluene (BHT) (EC50 = 47.1 μg/mL), and to be 1.5 times more promising than this standard in ABTS●+ and ß-carotene bleaching inhibition methods [44]. Moreover, hydromethanolic extracts of Thymus capitatus [50] and Thymus mastichina [37], which presented rosmarinic acid as major phenolic constituent, were shown to have high antioxidant potential based in DPPH (18–149 mg TE/g DW) and FRAP assays (30–154 mg TE/g DW).
In addition, a significant scavenging activity of ABTS●+ (expressed as mmol Trolox eq./g extract) was observed for hydroethanolic/aqueous decoction extracts of Thymus pulegioides (1.53/1.58) [61], Thymus citriodorus (1.52/1.21) and T. vulgaris (0.92/0.79) [51], T. mastichina (1.48/0.96) [54], and Thymus zygis subsp. zygis (1.08/0.76) [60]. These authors also noted the high inhibitory ability of some of these extracts (at 0.1 mg/mL) against hydroxyl radical in the presence of EDTA, namely the decoctions of T. zygis subsp. zygis (66%) [60] and T. citriodorus (37.9%) [51], as well the hydroethanolic and decoctions of T. mastichina (43.2 and 48.5%, respectively) [54] and T. pulegioides (34.8 and 34.0%, respectively) [61].
Favorable DPPH and reducing power results were also been observed by our group for decoctions of Thymus herba-barona, Thymus pseudolanuginosus, Thymus caespititius, T. pulegioides, Thymus fragantissimus and T. zygis [40,41], for which the EC50 values were 1.4–2.0 higher than those of the reference commercial compounds. Among these six extracts, those rich in flavones (T. pulegioides and T. pseudolanuginosus) tended to have superior DPPH scavenging ability and reducing Fe ability. In fact, the DPPH scavenging ability of these extracts surpassed that recently described for decoction and infusion extracts of Thymus sipyleus Boiss. subsp. rosulans (Borbas) Jalas (EC50 values of 43.5±1.02 and 87.38±1.73 μg/mL, corresponding to about 1.6–3.2-fold of the ascorbic acid). Interestingly, the authors disclosed a greater antioxidant ability of these polar solvent extracts in comparison to non-polar ones [56]. Different results were described for aqueous extracts of Thymus atlanticus, T. zygis and Thymus satureioides based on ABTS+ scavenging assay, which corresponded to about 7.5–8.5 times those of the standard ascorbic acid [62].
In opposition to chemical assays, to our knowledge, only a few authors used cellular and/or in vivo models to screen the antioxidant potential of Thymus polar extracts (Table 1). Under this context, hydroethanolic extracts of T. citriodorus (50 μg/mL) were shown to reduce significantly the intracellular ROS formation in a potassium dichromate-stimulated HepG2 cells [52]. Moreover, Khouya et al. [63] reported the protection of T. satureioides against oxidative damage, by demonstrating that the addition of its crude extract or subsequent fractions (dichloromethane, ethyl acetate, methanol or aqueous) to erythrocyte suspensions resulted in an increase in haemolysis half-time (1.2 to 2.1-fold of potency compared to ascorbic acid). As regard to in vivo studies, it was only recently that Righi et al. [46] reported that the treatment of Swiss albinos mice with Thymus algeriensis hydroalcoholic extracts at 800 mg/kg bw resulted in an improvement of the antiradical ability of the plasma, as well as catalase and glutathione levels (at 400 and 800 mg/kg bw), paralleled by a decrement on the levels of malondialdehyde (200 and 400 mg/kg bw). In addition, El-Boshy et al. [58] demonstrated that supplementation of lead (Pb) intoxicated rats with ethanolic extracts from T. vulgaris could ameliorate antioxidant parameters and the overall toxic effects.
Notably, some authors evaluated the antiradical/antioxidant activity of Thymus polar extracts with regard to their phenolic content and to the presence of specific phenolic compounds [37,51], particularly with the major components (e.g., rosmarinic acid, caffeic acid, luteolin-O-glucoside, apigenin, naringenin) [37,43,44,50]. A recent study [51] focused the antioxidant potential of T. vulgaris and T. citriodorus, described a high correlation with their polyphenol levels with their Trolox equivalent antioxidant capacity (TEAC) (e.g., rosmarinic acid (r = 0.874), eriodictyol-glucoside (r = 0.759), luteolin-hexuronide (r = 0.734, p < 0.0157), and chrysoeriol-hexoside (r = 0.771)) [51]. Amongst phenolic compounds, rosmarinic acid is frequently the principal phenolic acid in the said extracts and it is believed to play a central role in their antioxidant abilities [37,44]. In fact, this caffeic acid dimer was shown to be sixteen times better than α-tocopherol [44] and twice better than trolox [4] in DPPH-scavenging assay. It also exerts higher reducing power capacity (EC50 = 2.7 μg/mL) than trolox (EC50 = 6.6 μg/mL) [4] and higher ABTS●+ scavenging ability than BHT [44]. Despite that, according to Ertas et al. [44], this is less efficient that BHT in protecting against lipid peroxidation (EC50 value was 1.2-fold higher in the ß-carotene bleaching inhibition assay). Moreover, rosmarinic acid derivatives together with other non-flavonoids and flavonoid compounds (in particular luteolin, apigenin and its derivatives) may also contribute to the antioxidant capacity of Thymus extracts, since they are good antiradical/antioxidants as evaluated in chemical and cellular models. Namely, luteolin and apigenin (50 μg/mL) were demonstrated to drastically decrease the levels of intracellular ROS under oxidative-stress conditions on a hepatic cellular model [52]. Moreover, luteolin was highlighted by its high ability to prevent lipid peroxidation events when compared to rosmarinic acid [4].

3. Anti-Inflammatory Activity

A growing body of experimental evidence suggests that plant extracts or their bioactive compounds (e.g., phenolic compounds) have potential therapeutic application in the treatment of several inflammatory diseases, through distinct mechanisms, including the inhibition of pro-inflammatory cytokines, intercellular adhesion molecules and enzymes (e.g., iNOS, COX-2, 5-LOX), as well as, transcription factors, such as NF-κB [20,64,65,66,67], prevention of protein denaturation and stabilization of the potential of erythrocyte membrane [62] or control of the levels of radicals like NO [4,40]. Distinct authors evaluated the anti-inflammatory potential of Thymus polar extracts towards such events (Table 2).
The capacity to inhibit key enzymes in inflammation was demonstrated for extracts of different Thymus species. Among them, diethyl ether, ethyl acetate and n-butanol extracts of Thymus camphoratus, Thymus carnosus and T. mastichina [49] and methanolic extracts of T. algeriensis and Thymus fontanesii [68] were demonstrated to possess high inhibitory capacity against 5-LOX, with the last two showing similar potency to that of the positive control zileuton, and about one third of the potency of celecoxibe, with regard to the ability to inhibit COX-2 [68]. Moreover, the anti-inflammatory activity of aqueous extracts of T. atlanticus, T. zygis and T. satureioides was evidenced because of their potency to prevent protein denaturation and cellular lysis in a dose-dependent manner [62], as shown in Table 2. Among them, the authors underlined the erythrocyte-protective effect of T. atlanticus, which was superior to that of indomethacin (IC50 of 93.28 vs 97.83 μg/mL). Albeit the authors did not analyze the phenolic composition of the samples, they suggested that the anti-inflammatory effects were due to its high quantity of polyphenols found in these Thymus species [62].
The ability of natural compounds in scavenging the radical NO is frequently estimated through chemical assays and this is also the case of Thymus polar extracts. Among the distinct studies, one must emphasize the potential of ethanolic extracts from six Thymus species that showed EC50 NO scavenging results 1-3 times higher than the tested standard compound (Trolox), with the best results being obtained for the T. pulegioides and Thymus longicaulis species (EC50 of 69.8 and 71.6 μg/mL, respectively) [4]. In addition, T. herba-barona, T. pseudolanuginosus and T. caespititius decoctions, which were rich in caffeic acid derivatives and flavones, were all reported as good NO scavenging agents, from which that from T. caespititius origin had similar potency as ascorbic acid [40].
Studies on cellular models also support the hypothesis that Thymus phenolic-rich extracts possess anti-inflammatory activity. Among them, the work of Ustuner and colleagues [56] underlined the anti-inflammatory potential of T. sipyleus Boiss. subsp. rosulans (Borbas) Jalas aqueous extract through reducing the overproduction of NO and the expression of TNF-α, as attested in lipopolysaccharide (LPS)-activated RAW 264.7 macrophages. Using the same model, Silva et al. [60] demonstrated that 50 µg/mL of hydroethanolic and aqueous extracts of T. zygis decreased the NO levels by ~89% and 48%, respectively, while the aqueous extracts of T. carnosus Boiss. lowered the levels of this radical in a dose-dependent manner, reaching 90% inhibition for 200 μg/mL [69]. Besides, an hydroalcoholic extract of T. longicaulis (rich in rosmarinic acid) was shown to effectively inhibit COX-2 gene expression in THP-1 macrophages [42] and the aqueous extract of T. vulgaris were attested to decrease the release of the cytokine IL-8 for both H2O2 or TNFα-stimulated peripheral blood lymphocytes (PBL) [71]. In addition, a commercial propylene glycol extract of T. vulgaris was reported to inhibit the production of cytokines IL-1β and TNF-α in LPS-activated RAW 264.7 macrophages after exposure to different concentrations of propylene glycol extracts [72].
Notably, some authors emphasized a straight correlation between the high levels of phenolic acids and/or flavonoids in the extracts with their antioxidant/anti-inflammatory activities [37,71]. Among non-flavonoids, rosmarinic acid (in general the major phenolic in Thymus polar extracts) and its derivatives are probably positive contributors to anti-inflammatory activities. In fact, rosmarinic acid was demonstrated to be effective (in vitro and in vivo) against the production and mRNA expression of TNF-α, IL-1β, IL-6, COX-2 and iNOS, and to downregulate proteins involved in the signaling NF-kB-pathway [73]. Moreover, ethyl rosmarinate, a structurally-modified rosmarinic acid, was claimed to be a potent inhibitor against lung inflammation, based on NO and PGE2 inhibition and also by decreasing of mRNA and protein expression of iNOS and COX-2, as demonstrated in LPS-induced alveolar macrophages [74]. In the same line, flavonoids have also been shown to inhibit the expression of various enzyme systems, inflammatory mediators (e.g., molecules adhesion, cytokines and chemokines) and molecules involved in signal transduction pathways implicated in the inflammatory response [7,24,75]. In particular, luteolin (a common flavone in Thymus plants in general), was reported to inhibit the expression of COX-2 and TNF-α secretion in IFN-γ-stimulated RAW264.7 macrophage cells, and to inhibit the phosphorylation of signal transducer and activator of transcription (STAT)-3, a crucial transcription factor of proliferation and activation of T cells [76].
Opposing to the antioxidant capacity, in vivo anti-inflammatory potential of diverse thyme polar extracts have been reported on animal models of acute inflammation (e.g., paw edema induced by carrageenan, ear edema induced by toxics as croton oil, measurement of myeloperoxidase in leucocytes and total numbers of polymorphonuclear leukocytes (LPM)) [77,78,79]. Among the reported bibliography of Thymus plants, the work of Khouya et al. [43] underlined that the aqueous extracts of T. satureioides, T. atlanticus and T. zygis (at 900 µg/ear) could suppress croton oil-induced mice ear edema, and in addition, the last two (at 50 mg/kg) also inhibited carrageenan-induced rat paw edema, with similar potency to that of the non-steroidal anti-inflammatory drug indomethacin [43]. More recently, the same authors showed that the application of the dichloromethane and ethyl acetate fractions of T. satureioides (1 mg/ear) caused significant decreases in ear edema volume after 4 h (1.8–2.1-fold that of positive control indomethacin (600 μg/ear)), while oral treatment with a crude extract (60 mg/kg) inhibited significantly the carrageenan-induced rat paw oedema, during the first phase (1–5 h) [63]. In the same model, a methanolic fraction of Thymus praecox subsp. skorpilii var. skorpilii showed moderate anti-inflammatory effect [70]. Moreover, the intraperitoneal administration of an ethanolic extract of T. vulgaris (50 and 100 mg/kg) in a rat model of experimental autoimmune encephalomyelitis (EAE), was able to decrease the cytokines production by splenocytes (IFN-γ, IL-6, and IL-17), while the amount of TGF-ß secretion was significantly higher [80]. The administration of hydromethanolic extracts of T. algeriensis and T. fontanesii (600 mg/kg) resulted in the reduction (62% and 52%) on carrageenan-induced leukocyte migration into the peritoneal cavity in mice model, while diclofenac (20 mg/kg) and dexamethasone (2 mg/kg) caused 39% and 30% of reduction, respectively [68].
It is possible that major phenolic constituents of Thymus plants may contribute to their anti-inflammatory effects since either rosmarinic acid, caffeic acid and flavonoides were previously reported as to effectively counteract inflammatory events in vivo models [73,76,77,81,82,83]. In particular, rosmarinic acid suppressed LPS-induced proinflammatory cytokine [73,77], inhibited the expression of COX-2 and iNOS protein in mice disease-inflammatory models [73,84], and reduced the myeloperoxidase activity compared with the LPS-treated cells, suggesting the neutrophil infiltration, which indirectly reflect the degree of inflammation [77]. Likewise, the flavone luteolin suppressed inflammatory signals including TNF-α, NF-kB, MCP-1 and IL-1β in LPS-induced cells, attenuating acute renal injury in mice [81]. In addition, the anti-inflammatory effects of luteolin on autoimmune thyroiditis mouse model was demonstrated by the reduction of infiltrating lymphocytes and downregulation of IL-6/STAT1 and STAT3 signaling pathway in thyroid glands [76]. In turn, administration of apigenin exerted a beneficial role in acute mouse pancreatitis, reducing necrosis to 100% and a significant inhibition of TNF-α expression [85].
The overall data focus on the anti-inflammatory potential of Thymus extracts and phenolic compounds as well [24], emphasize their potential useful in the development of alternative drugs for the treatment of a variety of diseases settled on oxidative and/or inflammatory events, including autoimmune diseases, diabetes, cancers and ageing.

4. Anticancer Activity

The antitumoral potency of Thymus extracts has been studied, underlying their potential as sources of chemotherapeutic agents, particularly in in vitro cellular models. As for drugs in general, the majority of these studies used the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay as a first approach method to estimate cytotoxicity/growth-inhibitory activity of extracts in cancer cell lines (Table 3). This colorimetric method measures the activity of mitochondrial enzymes that reduce MTT to purple formazan, and this conversion is often used as an estimative of viable cells [30,59]. Similar principle methods are also applied by the authors, including those based on the reduction of 2,3-bis-(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide (XTT) [42,86]; or sulforhodamine B (SRB) assay, which is based on the ability of SRB to bind to protein components of cells under mild acidic conditions and that can be extracted under basic conditions, given an estimation of cell mass (extrapolated to growth activity) [87,88]. In general, literature data is presented as IC50 or GI50 values (inhibition concentration or grow inhibition, respectively), which both correspond to sample concentration able to inhibit 50% of the cell grow.
The antitumoral activity of the most widespread Thymus species, namely T. vulgaris and/or Thymus serpyllum, were previously screened. Commercial aqueous extracts from these two plant species were described to exhibit cytotoxic ability towards adriamycin-resistant human breast cancer cell line (MCF-7/Adr) [89]. Amongst representative compounds of the extracts (caffeic acid, rosmarinic acid, lithospermic acid, luteolin-7-O-glucuronide, luteolin-7-O-rutinoside and eriodictiol-7-O-rutinoside), rosmarinic acid and lithospermic acid were demonstrated to be the most active [89]. Notably, plant extracts (including T. vulgaris and T. serpyllum) proved to be more toxic against the cancer resistant cells than the analyzed phenolics (that exhibited greater toxicity against the non-resistant cells), suggesting the existence of synergistic or additive effects between the extracts’ components [89] (Table 3).
Moreover, methanolic extracts of Thymus schimperi, containing high amounts of the flavone luteolin and derivatives, required doses of 50–100 μg/mL to inhibit the growth of human gastric adenocarcinoma AGS cells by 50% (IC50 = 88 μg/mL) [91]. In addition, Galasso et al. [42] reported that T. longicaulis extracts, containing high amounts of quercetin-O-hexoside, luteolin-O-hexoside and salvianolic acid K, caused a decrease of cell viability on breast cancer MDA-MB-23, colon carcinoma HCT-116 and lung fibroblast MRC-5 cells, equal to or even higher than those observed for vinblastine, a known anti-cancer agent [25]. Besides, Esmaeili-Mahani et al. [30] reported that polar extracts of Thymus caramanicus (at 150–200 μg/mL), could reduce the viability of human breast adenocarcinoma MCF-7 for about 60% and interestingly, its concomitant treatment (40 and 80 μg/mL extract) with the vincristine effectively potentiated the effect of the anticancer drug. Aqueous extracts of T. satureioides, mainly composed of rosmarinic acid and of luteolin glucoside, exhibited an IC50 value of 37.5 µg/mL in the same cell line [43]. The cytotoxicity capacity was also demonstrated for methanolic extracts of T. serpyllum, which caused a viability decrement of two human breast cancer cell lines (MCF-7 and MDAMB-231) after 72 h exposure (IC50 of 509 and 276 μg/mL, respectively), disclosing less anti-proliferation ability in human normal breast cells (MCF-10A), which further proved the anticancer property of this extract [92]. Polar extracts obtained from T. carnosus Boiss. were also tested for their cytotoxicity in MCF-7, BT-474 and RAW 264.7 cells using Alamar Blues assay, with best results being obtained for the hydroethanolic extracts rich in salvianolic acids and rosmarinic acid [69]. T. zygis extracts, containing similar phenolic composition, displayed promising results in Caco-2 and HepG2 cells (IC50 = 85.01 and 82.19 µg/mL, respectively), especially at 48 h of exposure [60]. The same group reported high cytotoxic ability of aqueous decoction and hydroethanolic extracts from different Thymus species towards Caco-2, namely T. pulegioides (IC50 of 82.25 and 105.44 µg/mL respectively) [61] and T. citriodorus (IC50 of 159.40 and 114.6 µg/mL respectively) [51], albeit both were less active against HepG2 cells [51]. In addition, aqueous decoction/hydroethanolic extracts of T. mastichina (rich in salvianolic acid derivatives) required low doses to inhibit the same cells growth by 50% (IC50 of 95.65 and 51.30 μg/mL towards Caco-2; 285.03 and 180.10 μg/mL towards HepG2 cells, respectively) [54]. Interestingly, T. vulgaris, the most studied and widely used thyme species worldwide, showed the lowest cytotoxic activity against Caco-2 and HepG2 cells after 48 h, for aqueous decoction (IC50 values of 376.8 and > 500 µg/mL, respectively) and hydroethanolic extracts (IC50 values of 442.45 and 254.25 µg/mL, respectively) [51].
In addition, Gordo et al. [88] reported that the dichloromethane and ethanol extracts of the aerial parts of T. mastichina were able to inhibit the viability of the HCT colon cancer cell line (IC50 value of 2.8 μg/mL and 12.0 μg/mL, respectively). Moreover, in this study, the authors also described that ursolic acid and a mixture (1:1) of oleanolic acid and ursolic acid, isolated in the chromatographic procedures, exhibited high cytotoxicity (IC50 of 6.8 and 2.8 μg/mL, respectively), suggesting that phenolic compounds maybe related to the protective effect of this thyme extract against colon cancer cells [88]. Discrepant results were reported recently for an ethanolic extract of T. algeriensis Boiss. and Reut. since this was ineffective in suppressing cancer cell growth (required a concentration greater than 10 000 μg/mL for achieving 50% reduction of cancer cell lines survival) [45]. In turn, other study using a methanolic extract from the same species, rich in gallic and vanillic acids, reported an effective protection against in vitro Multiple Myeloma cell line (U266) proliferation in a dose-dependent manner [90].
The DNA repair ability is very important, as DNA damage can lead to carcinogenesis if replication proceeds without a proper repair. The DNA-protective effect of plant extracts against DNA strand breaks induced by the oxidant agents H2O2 and 2,3-dimethoxy-1,4-naphthoquinone (DMNQ) has been performed by the comet assay, based in single cell gel electrophoresis [59,93]. Kozics et al. reported the protective effects of ethanolic extract of T. vulgaris (rich in rosmarinic acid) against DNA damage induced by strong oxidants, achieved the highest DNA-protective capacity at concentrations of 0.5 and 0.1 mg/mL, respectively [59].
Important effects of Thymus extracts in key mechanisms of tumor formation/progression (e.g., apoptosis and angiogenesis) have also been attested, both in in vitro and in vivo models. Although less studied than essential oils [8], phenolic-rich extracts from Thymus have been evaluated for effects on apoptosis of proliferative cells by cycle analysis, mostly through the flow cytometry method [94]. Some studies also investigated the apoptosis induction of cancer cells through activation of the caspase cascade or by disruption in the balance between pro- and antiapoptotic Bcl-2 family members, after exposure to thyme polar extracts. For example, Al-Menhali et al. [95] registered moderate apoptotic capacity of aqueous extracts of T. vulgaris, which were shown to increase the activity of caspase 3/7 on human colon carcinoma cells HCT116 in a concentration-dependent manner (3.5%, 36.1% and 54.7% increase in the activity of caspase 3/7 in cells treated with 0.2, 0.4, or 0.6 mg/mL, respectively) [95]. In addition, Esmaeili-Mahani et al. [30] highlighted the anticancer activity of T. caramanicus hydroalcoholic extracts, based on their ability to induce apoptosis in human breast cancer cells MCF-7 through the increment of caspase-3 and Bax and decreasing of Bcl-2 expression. These T. caramanicus extracts (200–250 µg/mL) also potentiated the cytotoxicity of vincristine, which was accompanied by an increase in the levels of cleaved caspase-3 [30]. Moreover, treatment of MDA-MB-231 breast cancer cells for 72 h with a methanolic extract of T. serpyllum gradually increased the caspase 3/7 enzyme activity (increased 1.6-, 2.2-, and 3-fold at 10, 250, and 500 μg/mL of extract, respectively) [92]. Less promising results were reported by Caprioli and coworkers [94] for the apoptotic effect of an ethanolic extract of Thymus lanceolatus (rich in rosmarinic acid), which required a concentration of 500 μg/mL for a significant increase in programmed cell death of chronic myelogenous leukemia K562 cells (11.5% and 14.2% at 48 and 72 h, respectively), with no significant effect towards colorectal adenocarcinoma CaCo-2 and neuroblastoma SH-SY5Y cells.
Once the inhibition of angiogenesis has been recognized to be advantageous for prevention of inflammation and neoplastic growth [33,96], methanolic extracts of T. capitatus (100 μg/mL) were shown to inhibit significantly micro-vessel formation in rat aortic rings [97]. Still, as the authors did not find significant effects against proliferation and migration of the human endothelial EA.hy926 cells after extract exposure, they concluded that the in vivo observed anti-angiogenic effects must occur thorough an indirect mechanisms of action [97].
The activity of enzymes involved in inflammation, such as COX-2 and its product PGE-2 (known as carcinogenesis promoters), and others involved in enhancing detoxification, like as GPx, SOD or catalase [34,35], and cytokines or others immunological mediators, have also been used for evaluation of the antitumoral activity of Thymus extracts [42,59]. With this regard, ethanolic extracts of T. vulgaris (1 and 0.5 mg/mL), with high amounts of rosmarinic acid, were described to induce a significant increase of GPx activity in human HepG2 cells, while SOD activity was significantly lowered with respect to the control [59]. Likewise, hydroalcoholic extracts of T. longicaulis, particularly rich in rosmarinic acid and methylapigenin, exhibited a strong anti-inflammatory and anticancer effectiveness through inhibition of COX-2 gene expression on human leukemia cell line THP-1 macrophages [42]. These results suggest that the downregulation of COX-2 expression/activity and growth inhibition of tumor cells are closely associated, determining the relationships of anti-inflammatory, antioxidant and cytotoxic potential [42,98].
Moreover, a water extract of Thymus pannonicus was demonstrated to cause a significant increase in the activity of xanthine oxidase, glutathione reductase and GPx in Ehrlich ascites carcinoma mice cells (EACs), having the authors concluded that the induction of oxidative stress may play an important role in antitumor properties of the extract [99]. The authors also demonstrated that this Thymus extract caused a strong tumor growth inhibition in mice, which was followed by a high percentage of apoptotic/necrotic cells in a dose dependent manner, as well as significant decrease (96–98%) in the EAC volume [99]. The extract was particularly rich in rosmarinic acid, salvianolic acid H, catechin derivatives and glycosidic derivatives of luteolin and apigenin. On another study, Caproli and coworkers also reported that an ethanolic extract of T. lanceolatus, rich in rosmarinic acid and flavonoids, was an efficient ROS scavenger, exhibiting a good antioxidant activity (since it decreases the ROS generation by about 30% in SH-SY5Y (neuroblastoma) and 70% in CaCo-2 (colorectal adenocarcinoma) and K562 (chronic myelogenous leukemia) cells [94].

5. Conclusions

The Thymus genus is a rich source of secondary metabolites with claimed health benefits. For the last years, phenolic-rich extracts from distinct Thymus species have been the focus of attention and screened for distinct bioactive properties of interest for application in industrial fields like food, pharmaceutical and cosmetics. This review highlights the biological activities of phenolic-rich extracts from various Thymus species, with emphasis devoted to antioxidant, anti-inflammatory and anticancer properties. Globally, the documented data revealed that Thymus extracts, rich in phenolic acids and flavonoids compounds, may have enormous ability to scavenge radicals and reduce inflammatory events. Importantly, several studies disclose that thyme extracts have the capacity to inhibit diverse proinflammatory mediators, such as cytokines (IL-1β, IL-6, IL-12, INF-γ, TNF-α), enzymes (iNOS, COX-2) and others, which are majorly regulated by the transcriptional factor NF-κB, whose activation is also associated with increased ROS generation. Moreover, there is also a considerable amount of work reflecting the promising cytotoxic activity of some Thymus phenolic-riche extracts on tumor cell lines, as well as their ability as regulators of the tumor cell cycle, leading to programed cell death or inhibiting angiogenesis. Hence, the evidenced health-benefits of Thymus extracts as antioxidant and anti-inflammatory agents corroborate their potential applications for pharmaceutical purposes, including the development of antitumoral compounds.
Regardless that, one must remark that so far, most of the gathered investigation has been based on in vitro experiments whereas there is a huge lack of in vivo studies, in order to be able to confirm the beneficial effects of Thymus polar extracts on human health.

Author Contributions

A.F.A. contribution to writing the original draft; O.R.P. and S.M.C. contribution to supervision and writing—reviewing and editing. All authors have read and agreed to the published version of the manuscript.

Funding

Science and Technology Foundation/Ministry of Education and Science (FCT/MEC) funded the Associated Laboratory for Green Chemistry (LAQV) of the Network of Chemistry and Technology (REQUIMTE) (UIDB/50006/2020) and CIMO (UIDB/00690/2020), through national funds and, where applicable, co-financed by FEDER, within the Portugal 2020. Project AgroForWealth (CENTRO-01-0145-FEDER-000001), funded by Centro2020, through FEDER and PT2020, financed the research contract of Susana M. Cardoso.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Morales, R. The History, Botany and Taxonomy of the Genus Thymus. In Thyme, the Genus Thymus; Stahl-Biskup, E., Sáez, F., Eds.; Taylor & Francis: London, UK, 2002; ISBN 0415284880. [Google Scholar]
  2. Pereira, O.R.; Cardoso, S.M. Overview on Mentha and Thymus Polyphenols. Curr. Anal. Chem. 2013, 9, 382–396. [Google Scholar] [CrossRef] [Green Version]
  3. Horwath, A.B.; Grayer, R.J.; Keith-Lucas, D.M.; Simmonds, M.S.J. Chemical characterisation of wild populations of Thymus from different climatic regions in southeast Spain. Biochem. Syst. Ecol. 2008, 36, 117–133. [Google Scholar] [CrossRef]
  4. Kindl, M.; Blažeković, B.; Bucar, F.; Vladimir-Knežević, S. Antioxidant and anticholinesterase potential of six thymus species. Evidence-based Complement. Altern. Med. 2015, 2015, 403950. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Iauk, L.; Acquaviva, R.; Mastrojeni, S.; Amodeo, A.; Pugliese, M.; Ragusa, M.; Loizzo, M.R.; Menichini, F.; Tundis, R. Antibacterial, antioxidant and hypoglycaemic effects of Thymus capitatus (L.) Hoffmanns. et Link leaves’ fractions. J. Enzyme Inhib. Med. Chem. 2015, 30, 360–365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Ozkan, G.; Kamiloglu, S.; Ozdal, T.; Boyacioglu, D.; Capanoglu, E. Potential use of Turkish medicinal plants in the treatment of various diseases. Molecules 2016, 21, 257. [Google Scholar] [CrossRef]
  7. Kumar, S.; Pandey, A.K. Chemistry and biological activities of flavonoids: An overview. Sci. World J. 2013, 2013, 162750. [Google Scholar] [CrossRef] [Green Version]
  8. Kubatka, P.; Uramova, S.; Kello, M.; Kajo, K.; Samec, M.; Jasek, K.; Vybohova, D.; Liskova, A.; Mojzis, J.; Adamkov, M.; et al. Anticancer Activities of Thymus vulgaris L. in Experimental Breast Carcinoma In Vivo and In Vitro. Int. J. Mol. Sci. 2019, 20, 1749. [Google Scholar] [CrossRef] [Green Version]
  9. Nikolić, M.; Glamočlija, J.; Ferreira, I.C.F.R.; Calhelha, R.C.; Fernandes, Â.; Marković, T.; Marković, D.; Giweli, A.; Soković, M. Chemical composition, antimicrobial, antioxidant and antitumor activity of Thymus serpyllum L., Thymus algeriensis Boiss. and Reut and Thymus vulgaris L. essential oils. Ind. Crops Prod. 2014, 52, 183–190. [Google Scholar] [CrossRef]
  10. Grulova, D.; Scognamiglio, M.; Snoussi, M.; Feo, V. Studies on Chemical Composition, Antimicrobial and Antioxidant Activities of Five Thymus vulgaris L. Essential Oils. Molecules 2015, 20, 12016–12028. [Google Scholar] [CrossRef] [Green Version]
  11. Figueiredo, A.C.; Barroso, J.G.; Pedro, L.G.; Salgueiro, L.; Miguel, M.G.; Faleiro, M.L. Portuguese Thymbra and Thymus species volatiles: chemical composition and biological activities. Curr. Pharm. Des. 2008, 14, 3120–3140. [Google Scholar] [CrossRef]
  12. Brewer, M.S. Natural Antioxidants: Sources, Compounds, Mechanisms of Action, and Potential Applications. Compr. Rev. Food Sci. Food Saf. 2011, 10, 221–247. [Google Scholar] [CrossRef]
  13. Lagouge, M.; Larsson, N.-G. The role of mitochondrial DNA mutations and free radicals in disease and ageing. J. Intern. Med. 2013, 273, 529–543. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Sánchez-Rodríguez, C.; Cuadrado, E.; Riestra-Ayora, J.; Sanz-Fernández, R. Polyphenols protect against age-associated apoptosis in female rat cochleae. Biogerontology 2018, 19, 159–169. [Google Scholar] [CrossRef] [PubMed]
  15. Mangge, H. Antioxidants, inflammation and cardiovascular disease. World J. Cardiol. 2014, 6, 462–477. [Google Scholar] [CrossRef]
  16. He, L.; He, T.; Farrar, S.; Ji, L.; Liu, T.; Ma, X. Antioxidants Maintain Cellular Redox Homeostasis by Elimination of Reactive Oxygen Species. Cell. Physiol. Biochem. 2017, 44, 532–553. [Google Scholar] [CrossRef]
  17. Lin, D.; Xiao, M.; Zhao, J.; Li, Z.; Xing, B.; Li, X.; Kong, M.; Li, L.; Zhang, Q.; Liu, Y.; et al. An overview of plant phenolic compounds and their importance in human nutrition and management of type 2 diabetes. Molecules 2016, 21, 1374. [Google Scholar] [CrossRef]
  18. Biswas, S.K. Does the Interdependence between Oxidative Stress and Inflammation Explain the Antioxidant Paradox? Oxid. Med. Cell. Longev. 2016, 2016, 17–19. [Google Scholar] [CrossRef] [Green Version]
  19. Guillaumet-Adkins, A.; Yañez, Y.; Peris-Diaz, M.D.; Calabria, I.; Palanca-Ballester, C.; Sandoval, J. Epigenetics and Oxidative Stress in Aging. Oxid. Med. Cell. Longev. 2017, 2017, 9175806. [Google Scholar] [CrossRef]
  20. Catarino, M.D.; Talhin, O.; Rabahi, A.; Silva, A.M.S.; Cardoso, S.M. The Anti-inflammatory Potential of Flavonoids: Mechanistic Aspects. In Studies in Natural Products Chemistry; Atta-ur-Rahman, F., Ed.; Elsevier: Amsterdam, The Netherlands, 2016; pp. 65–99. [Google Scholar]
  21. Goldsby, R.A.; Kindt, T.J.; Osborne, B.A.; Kuby, J. Leukocyte Migration and Inflammation. In Kuby Immunology, 6th ed.; W.H. Freeman and Company: New York, NY, USA, 2007; pp. 327–349. [Google Scholar]
  22. Bisht, R.; Bhattacharya, S.; Jaliwala, Y.A. COX and LOX inhibitory potential of Abroma augusta and Desmodium gangeticum. J. Phytopharm. 2014, 3, 168–175. [Google Scholar]
  23. Martel-Pelletier, J.; Lajeunesse, D.; Reboul, P.; Pelletier, J.P. Therapeutic role of dual inhibitors of 5-LOX and COX, selective and non-selective non-steroidal anti-inflammatory drugs. Ann. Rheum. Dis. 2003, 62, 501–509. [Google Scholar] [CrossRef]
  24. Ginwala, R.; Bhavsar, R.; Chigbu, D.G.I.; Jain, P. Potential Role of Flavonoids in Treating Chronic Inflammatory Diseases with a Special Focus on the Anti-Inflammatory Activity of Apigenin. Antioxidants 2019, 8, 35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Grimm, E.A.; Sikora, A.G.; Ekmekcioglu, S. Molecular pathways: Inflammation-associated nitric-oxide production as a cancer-supporting redox mechanism and a potential therapeutic target. Clin. Cancer Res. 2013, 19, 5557–5563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. De Oliveira, G.A.; Cheng, R.Y.S.; Ridnour, L.A.; Basudhar, D.; Somasundaram, V.; McVicar, D.W.; Monteiro, H.P.; Wink, D.A. Inducible Nitric Oxide Synthase in the Carcinogenesis of Gastrointestinal Cancers. Antioxid. Redox Signal. 2017, 26, 1059–1077. [Google Scholar] [CrossRef] [PubMed]
  27. Huang, S.; He, X. The role of microRNAs in liver cancer progression. Br. J. Cancer 2011, 104, 235–240. [Google Scholar] [CrossRef] [Green Version]
  28. Yang, B.F.; Lu, Y.J.; Wang, Z.G. MicroRNAs and apoptosis: Implications in the molecular therapy of human disease. Clin. Exp. Pharmacol. Physiol. 2009, 36, 951–960. [Google Scholar] [CrossRef] [PubMed]
  29. Laulier, C.; Lopez, B.S. The secret life of Bcl-2: Apoptosis-independent inhibition of DNA repair by Bcl-2 family members. Mutat. Res. Rev. Mutat. Res. 2012, 751, 247–257. [Google Scholar] [CrossRef]
  30. Esmaeili-Mahani, S.; Falahi, F.; Yaghoobi, M.M. Proapoptotic and antiproliferative effects of Thymus caramanicus on human breast cancer cell line (MCF-7) and its interaction with anticancer drug vincristine. Evidence-based Complement. Altern. Med. 2014, 2014, 893247. [Google Scholar] [CrossRef] [Green Version]
  31. Wang, W.-H.; Hsuan, K.-Y.; Chu, L.-Y.; Lee, C.-Y.; Tyan, Y.-C.; Chen, Z.-S.; Tsai, W.-C. Anticancer effects of Salvia miltiorrhiza alcohol extract on oral squamous carcinoma cells. Evidence-based Complement. Altern. Med. 2017, 2017. [Google Scholar] [CrossRef]
  32. Ye, Y.T.; Zhong, W.; Sun, P.; Wang, D.; Wang, C.; Hu, L.M.; Qian, J.Q. Apoptosis induced by the methanol extract of Salvia miltiorrhiza Bunge in non-small cell lung cancer through PTEN-mediated inhibition of PI3K/Akt pathway. J. Ethnopharmacol. 2017, 200, 107–116. [Google Scholar] [CrossRef]
  33. Aravindaram, K.; Yang, N.; Biotechnology, A. Anti-Inflammatory Plant Natural Products for Cancer Therapy. Rom. Biotechnol. Lett. 2010, 76, 1103–1117. [Google Scholar] [CrossRef] [Green Version]
  34. Cerella, C.; Sobolewski, C.; Dicato, M.; Diederich, M. Targeting COX-2 expression by natural compounds: A promising alternative strategy to synthetic COX-2 inhibitors for cancer chemoprevention and therapy. Biochem. Pharmacol. 2010, 80, 1801–1815. [Google Scholar] [CrossRef] [PubMed]
  35. Liu, R.H. Dietary bioactive compounds and their health implications. J. Food Sci. 2013, 78, 18–25. [Google Scholar] [CrossRef] [PubMed]
  36. Ravishankar, D.; Rajora, A.K.; Greco, F.; Osborn, H.M.I. Flavonoids as prospective compounds for anti-cancer therapy. Int. J. Biochem. Cell Biol. 2013, 45, 2821–2831. [Google Scholar] [CrossRef] [PubMed]
  37. Méndez-Tovar, I.; Sponza, S.; Asensio-S-Manzanera, M.C.; Novak, J. Contribution of the main polyphenols of Thymus mastichina subsp: mastichina to its antioxidant properties. Ind. Crops Prod. 2015, 66, 291–298. [Google Scholar] [CrossRef]
  38. Orłowska, M.; Pytlakowska, K.; Mrozek-Wilczkiewicz, A.; Musioł, R.; Waksmundzka-Hajnos, M.; Sajewicz, M.; Kowalska, T. A Comparison of Antioxidant, Antibacterial, and Anticancer Activity of the Selected Thyme Species by Means of Hierarchical Clustering and Principal Component Analysis. Acta Chromatogr. 2016, 28, 207–221. [Google Scholar] [CrossRef]
  39. Krishnaiah, D.; Sarbatly, R.; Nithyanandam, R. A review of the antioxidant potential of medicinal plant species. Food Bioprod. Process. 2011, 89, 217–233. [Google Scholar] [CrossRef]
  40. Afonso, A.F.; Pereira, O.R.; Neto, R.T.; Silva, A.M.S.; Cardoso, S.M. Health-promoting effects of Thymus herba-barona, Thymus pseudolanuginosus, and Thymus caespititius decoctions. Int. J. Mol. Sci. 2017, 18, 1879. [Google Scholar] [CrossRef]
  41. Afonso, A.F.; Pereira, O.R.; Válega, M.; Silva, A.M.S.; Cardoso, S.M. Metabolites and biological activities of Thymus zygis, Thymus pulegioides, and Thymus fragrantissimus grown under organic cultivation. Molecules 2018, 23, 1514. [Google Scholar] [CrossRef] [Green Version]
  42. Galasso, S.; Pacifico, S.; Kretschmer, N.; Pan, S.P.; Marciano, S.; Piccolella, S.; Monaco, P.; Bauer, R. Influence of seasonal variation on Thymus longicaulis C. Presl chemical composition and its antioxidant and anti-inflammatory properties. Phytochemistry 2014, 107, 80–90. [Google Scholar] [CrossRef]
  43. Khouya, T.; Ramchoun, M.; Hmidani, A.; Amrani, S.; Harnafi, H.; Benlyas, M.; Zegzouti, Y.F.; Alem, C. Anti-inflammatory, anticoagulant and antioxidant effects of aqueous extracts from Moroccan thyme varieties. Asian Pac. J. Trop. Biomed. 2015, 5, 636–644. [Google Scholar] [CrossRef] [Green Version]
  44. Ertas, A.; Boga, M.; Yilmaz, M.A.; Yesil, Y.; Tel, G.; Temel, H.; Hasimi, N.; Gazioglu, I.; Ozturk, M.; Ugurlu, P. A detailed study on the chemical and biological profiles of essential oil and methanol extract of Thymus nummularius (Anzer tea): Rosmarinic acid. Ind. Crops Prod. 2015, 67, 336–345. [Google Scholar] [CrossRef]
  45. Rezzoug, M.; Bakchiche, B.; Gherib, A.; Roberta, A.; Kilinçarslan, Ö.; Mammadov, R.; Bardaweel, S.K. Chemical composition and bioactivity of essential oils and Ethanolic extracts of Ocimum basilicum L. and Thymus algeriensis Boiss. & Reut. from the Algerian Saharan Atlas. BMC Complement. Altern. Med. 2019, 19, 1–10. [Google Scholar]
  46. Righi, N.; Boumerfeg, S.; Fernandes, P.A.R.; Deghima, A.; Baali, F.; Coelho, E.; Cardoso, S.M.; Coimbra, M.A.; Baghiani, A. Thymus algeriensis Bioss & Reut: Relationship of phenolic compounds composition with in vitro/in vivo antioxidant and antibacterial activity. Food Res. Int. 2020, 136, 109500. [Google Scholar] [CrossRef] [PubMed]
  47. Jaouadi, R.; Cardoso, S.M.; Silva, A.M.S.; Ben Hadj Yahia, I.; Boussaid, M.; Zaouali, Y. Variation of phenolic constituents of Tunisian Thymus capitatus (L.) Hoff. et Link. populations. Biochem. Syst. Ecol. 2018, 77, 10–15. [Google Scholar] [CrossRef]
  48. Jaouadi, R.; Silva, A.M.S.; Boussaid, M.; Yahia, I.B.H.; Cardoso, S.M.; Zaouali, Y. Differentiation of phenolic composition among tunisian Thymus algeriensis boiss. Et reut. (Lamiaceae) populations: Correlation to bioactive activities. Antioxidants 2019, 8, 515. [Google Scholar] [CrossRef] [Green Version]
  49. Albano, S.M.; Miguel, M.G. Biological activities of extracts of plants grown in Portugal. Ind. Crops Prod. 2011, 33, 338–343. [Google Scholar] [CrossRef]
  50. Skendi, A.; Irakli, M.; Chatzopoulou, P. Analysis of phenolic compounds in Greek plants of Lamiaceae family by HPLC. J. Appl. Res. Med. Aromat. Plants 2017, 6, 62–69. [Google Scholar] [CrossRef]
  51. Taghouti, M.; Martins-Gomes, C.; Félix, L.M.; Schäfer, J.; Santos, J.A.; Bunzel, M.; Nunes, F.M.; Silva, A.M. Polyphenol composition and biological activity of Thymus citriodorus and Thymus vulgaris: Comparison with endemic Iberian Thymus species. Food Chem. 2020, 331, 127362. [Google Scholar] [CrossRef] [PubMed]
  52. Pereira, O.R.; Macias, R.I.R.; Perez, M.J.; Marin, J.J.G.; Cardoso, S.M. Protective effects of phenolic constituents from Cytisus multiflorus, Lamium album L. and Thymus citriodorus on liver cells. J. Funct. Foods 2013, 5, 1170–1179. [Google Scholar] [CrossRef]
  53. Barros, L.; Heleno, S.A.; Carvalho, A.M.; Ferreira, I.C.F.R. Lamiaceae often used in Portuguese folk medicine as a source of powerful antioxidants: Vitamins and phenolics. LWT Food Sci. Technol. 2010, 43, 544–550. [Google Scholar] [CrossRef]
  54. Taghouti, M.; Martins-Gomes, C.; Schäfer, J.; Santos, J.A.; Bunzel, M.; Nunes, F.M.; Silva, A.M. Chemical characterization and bioactivity of extracts from Thymus mastichina: A thymus with a distinct salvianolic acid composition. Antioxidants 2020, 9, 34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Fernandes, Â.S.F.; Barros, L.; Carvalho, A.M.; Ferreira, I.C.F.R. Lipophilic and hydrophilic antioxidants, lipid peroxidation inhibition and radical scavenging activity of two Lamiaceae food plants. Eur. J. Lipid Sci. Technol. 2010, 112, 1115–1121. [Google Scholar] [CrossRef]
  56. Ustuner, O.; Anlas, C.; Bakirel, T.; Ustun-alkan, F.; Sigirci, B.D.; Ak, S.; Akpulat, H.A.; Donmez, C.; Koca-caliskan, U. In Vitro Evaluation of Antioxidant, Anti-Inflammatory, Antimicrobial and Wound Healing Potential of Thymus Sipyleus Boiss. Subsp. Rosulans (Borbas) Jalas. Molecules 2019, 24, 3353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Brahmi, N.; Scognamiglio, M.; Pacifico, S.; Mekhoukhe, A.; Madani, K.; Fiorentino, A.; Monaco, P. 1H NMR based metabolic profiling of eleven Algerian aromatic plants and evaluation of their antioxidant and cytotoxic properties. Food Res. Int. 2015, 76, 334–341. [Google Scholar] [CrossRef]
  58. El-Boshy, M.E.; Refaat, B.; Qasem, A.H.; Khan, A.; Ghaith, M.; Almasmoum, H.; Mahbub, A.; Almaimani, R.A. The remedial effect of Thymus vulgaris extract against lead toxicity-induced oxidative stress, hepatorenal damage, immunosuppression, and hematological disorders in rats. Environ. Sci. Pollut. Res. 2019, 26, 22736–22746. [Google Scholar] [CrossRef]
  59. Kozics, K.; Klusová, V.; Srančíková, A.; Mučaji, P.; Slameňová, D.; Hunáková, Ľ.; Kusznierewicz, B.; Horváthová, E. Effects of Salvia officinalis and Thymus vulgaris on oxidant-induced DNA damage and antioxidant status in HepG2 cells. Food Chem. 2013, 141, 2198–2206. [Google Scholar] [CrossRef]
  60. Silva, A.M.; Martins-Gomes, C.; Souto, E.B.; Schäfer, J.; Santos, J.A.; Bunzel, M.; Nunes, F.M. Thymus zygis subsp. zygis an endemic portuguese plant: Phytochemical profiling, antioxidant, anti-proliferative and anti-inflammatory activities. Antioxidants 2020, 9, 482. [Google Scholar] [CrossRef]
  61. Taghouti, M.; Martins-Gomes, C.; Schäfer, J.; Félix, L.M.; Santos, J.A.; Bunzel, M.; Nunes, F.M.; Silva, A.M. Thymus pulegioides L. as a rich source of antioxidant, anti-proliferative and neuroprotective phenolic compounds. Food Funct. 2018, 9, 3617–3629. [Google Scholar] [CrossRef]
  62. Hmidani, A.; Dine, E.; Bouhlali, T.; Khouya, T.; Ramchoun, M. Antioxidant, anti-inflammatory and anticoagulant activities of three Thymus species grown in southeastern Morocco. Futur. J. Pharm. Sci. 2019, 5. [Google Scholar] [CrossRef] [Green Version]
  63. Khouya, T.; Ramchoun, M.; Hmidani, A.; El moualij, B.; Amrani, S.; Harnafi, H.; Benlyas, M.; Zegzouti, Y.F.; Nazih, E.H.; Ouguerram, K.; et al. Acute toxicity and antiproliferative and procoagulant activities of fractions derived from Thymus satureioides of the Moroccan High Atlas. South African J. Bot. 2019, 121, 568–576. [Google Scholar] [CrossRef]
  64. Debnath, T.; Kim, D.H.; Lim, B.O. Natural products as a source of anti-inflammatory agents associated with inflammatory bowel disease. Molecules 2013, 18, 7253–7270. [Google Scholar] [CrossRef]
  65. Lopez-Lazaro, M. Distribution and Biological Activities of the Flavonoid Luteolin. Mini-Reviews Med. Chem. 2009, 9, 31–59. [Google Scholar] [CrossRef] [PubMed]
  66. Leyva-Lopez, N.; Gutierrez-Grijalva, E.P.; Ambriz-Perez, D.L.; Basilio Heredia, J. Flavonoids as cytokine modulators: A possible therapy for inflammation-related diseases. Int. J. Mol. Sci. 2016, 17, 921. [Google Scholar] [CrossRef]
  67. Azab, A.; Nassar, A.; Azab, A.N. Anti-inflammatory activity of natural products. Molecules 2016, 21, 1321. [Google Scholar] [CrossRef]
  68. Sobeh, M.; Rezq, S.; Cheurfa, M.; Abdelfattah, M.A.O.; Rashied, R.M.H.; El-Shazly, A.M.; Yasri, A.; Wink, M.; Mahmoud, M.F. Thymus algeriensis and Thymus fontanesii: Chemical composition, in vivo antiinflammatory, pain killing and antipyretic activities: A comprehensive comparison. Biomolecules 2020, 10, 599. [Google Scholar] [CrossRef] [Green Version]
  69. Martins-Gomes, C.; Taghouti, M.; Schäfer, J.; Bunzel, M.; Silva, A.M.; Nunes, F.M. Chemical characterization and bioactive properties of decoctions and hydroethanolic extracts of Thymus carnosus Boiss. J. Funct. Foods 2018, 43, 154–164. [Google Scholar] [CrossRef]
  70. Taşkın, T.; Çam, M.E.; Taşkın, D.; Rayaman, E. In vitro and In vivo biological activities and phenolic characterization of Thymus praecox subsp. skorpilii var. skorpilii. J. Food Meas. Charact. 2019, 13, 536–544. [Google Scholar] [CrossRef]
  71. Chohan, M.; Naughton, D.P.; Jones, L.; Opara, E.I. An investigation of the relationship between the anti-inflammatory activity, polyphenolic content, and antioxidant activities of cooked and in vitro digested culinary herbs. Oxid. Med. Cell. Longev. 2012, 2012, 627843. [Google Scholar] [CrossRef] [Green Version]
  72. De Oliveira, J.R.; de Jesus Viegas, D.; Martins, A.P.R.; Carvalho, C.A.T.; Soares, C.P.; Camargo, S.E.A.; Jorge, A.O.C.; de Oliveira, L.D. Thymus vulgaris L. extract has antimicrobial and anti-inflammatory effects in the absence of cytotoxicity and genotoxicity. Arch. Oral Biol. 2017, 82, 271–279. [Google Scholar] [CrossRef] [Green Version]
  73. Wei, Y.; Chen, J.; Hu, Y.; Lu, W.; Zhang, X.; Wang, R.; Chu, K. Rosmarinic Acid Mitigates Lipopolysaccharide-Induced Neuroinflammatory Responses through the Inhibition of TLR4 and CD14 Expression and NF-kappaB and NLRP3 Inflammasome Activation. Inflammation 2018, 41, 732–740. [Google Scholar] [CrossRef]
  74. Thammason, H.; Khetkam, P.; Pabuprapap, W.; Suksamrarn, A.; Kunthalert, D. Ethyl rosmarinate inhibits lipopolysaccharide-induced nitric oxide and prostaglandin E2 production in alveolar macrophages. Eur. J. Pharmacol. 2018, 824, 17–23. [Google Scholar] [CrossRef] [PubMed]
  75. Tuñón, M.J.; García-Mediavilla, M.V.; Sánchez-Campos, S.; González-Gallego, J. Potential of flavonoids as anti-inflammatory agents: modulation of pro-inflammatory gene expression and signal transduction pathways. Curr. Drug Metab. 2009, 10, 256–271. [Google Scholar] [CrossRef] [PubMed]
  76. Xia, N.; Chen, G.; Liu, M.; Ye, X.; Pan, Y.; Ge, J.; Mao, Y.; Wang, H.; Wang, J.; Xie, S. Anti-inflammatory effects of luteolin on experimental autoimmune thyroiditis in mice. Exp. Ther. Med. 2016, 12, 4049–4054. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Jiang, K.; Ma, X.; Guo, S.; Zhang, T.; Zhao, G.; Wu, H.; Wang, X.; Deng, G. Anti-inflammatory Effects of Rosmarinic Acid in Lipopolysaccharide-Induced Mastitis in Mice. Inflammation 2018, 41, 437–448. [Google Scholar] [CrossRef]
  78. Joe, Y.; Zheng, M.; Kim, H.J.; Kim, S.; Uddin, M.J.; Park, C.; Ryu, D.G.; Kang, S.S.; Ryoo, S.; Ryter, S.W.; et al. Salvianolic acid B exerts vasoprotective effects through the modulation of heme oxygenase-1 and arginase activities. J. Pharmacol. Exp. Ther. 2012, 341, 850–858. [Google Scholar] [CrossRef] [Green Version]
  79. Eddouks, M.; Chattopadhyay, D.; Zeggwagh, N.A. Animal models as tools to investigate antidiabetic and anti-inflammatory plants. Evidence-based Complement. Altern. Med. 2012, 2012, 142087. [Google Scholar] [CrossRef]
  80. Mahmoodi, M.; Ayoobi, F.; Aghaei, A.; Rahmani, M.; Taghipour, Z. Biomedicine & Pharmacotherapy Beneficial effects of Thymus vulgaris extract in experimental autoimmune encephalomyelitis: Clinical, histological and cytokine alterations. Biomed. Pharmacother. 2019, 109, 2100–2108. [Google Scholar] [CrossRef]
  81. Xin, S.; Yan, H.; Ma, J.; Sun, Q.; Shen, L. Protective Effects of Luteolin on Lipopolysaccharide-Induced Acute Renal Injury in Mice. Med. Sci. Monit. 2016, 22, 5173–5180. [Google Scholar] [CrossRef] [Green Version]
  82. Gamaro, G.D.; Suyenaga, E.; Borsoi, M.; Lermen, J.; Pereira, P.; Ardenghi, P. Effect of rosmarinic and caffeic acids on inflammatory and nociception process in rats. ISRN Pharmacol. 2011, 2011, 451682. [Google Scholar] [CrossRef] [Green Version]
  83. Rahbardar, M.G.; Amin, B.; Mehri, S.; Mirnajafi-Zadeh, S.J.; Hosseinzadeh, H. Rosmarinic acid attenuates development and existing pain in a rat model of neuropathic pain: An evidence of anti-oxidative and anti-inflammatory effects. Phytomedicine 2018, 40, 59–67. [Google Scholar] [CrossRef]
  84. Jin, B.R.; Chung, K.S.; Cheon, S.Y.; Lee, M.; Hwang, S.; Noh Hwang, S.; Rhee, K.J.; An, H.J. Rosmarinic acid suppresses colonic inflammation in dextran sulphate sodium (DSS)-induced mice via dual inhibition of NF-κB and STAT3 activation. Sci. Rep. 2017, 7, 1–11. [Google Scholar] [CrossRef] [PubMed]
  85. Charalabopoulos, A.; Davakis, S.; Lambropoulou, M.; Papalois, A.; Simopoulos, C.; Tsaroucha, A. Apigenin Exerts Anti-inflammatory Effects in an Experimental Model of Acute Pancreatitis by Down-regulating TNF-α. In Vivo 2019, 33, 1133–1141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Berrington, D.; Lall, N. Anticancer activity of certain herbs and spices on the cervical epithelial carcinoma (HeLa) cell line. Evidence-based Complement. Altern. Med. 2012, 2012, 564927. [Google Scholar] [CrossRef] [Green Version]
  87. Vichai, V.; Kirtikara, K. Sulforhodamine B colorimetric assay for cytotoxicity screening. Nat. Protoc. 2006, 1, 1112–1116. [Google Scholar] [CrossRef] [PubMed]
  88. Gordo, J.; Máximo, P.; Cabrita, E.; Lourenço, A.; Oliva, A.; Almeida, J.; Filipe, M.; Cruz, P.; Barcia, R.; Santos, M.; et al. Thymus mastichina: Chemical Constituents and their Anti-cancer Activity. Nat. Prod. Commun. 2012, 7, 1491–1494. [Google Scholar] [CrossRef] [PubMed]
  89. Berdowska, I.; Zieliński, B.; Fecka, I.; Kulbacka, J.; Saczko, J.; Gamian, A. Cytotoxic impact of phenolics from Lamiaceae species on human breast cancer cells. Food Chem. 2013, 141, 1313–1321. [Google Scholar] [CrossRef]
  90. Fatma, G.; Issam, S.; Rawya, S.; Najla, H.; Ahmed, L. Antioxidant Potential of Four Species of Natural Product and Therapeutic Strategies for Cancer through Suppression of Viability in the Human Multiple Myeloma Cell Line U266. Biomed. Environ. Sci. 2019, 32, 22–33. [Google Scholar] [CrossRef]
  91. Desta, K.T.; Kim, G.S.; El-Aty, A.M.A.; Raha, S.; Kim, M.B.; Jeong, J.H.; Warda, M.; Hacımüftüoğlu, A.; Shin, H.C.; Shim, J.H.; et al. Flavone polyphenols dominate in Thymus schimperi Ronniger: LC–ESI–MS/MS characterization and study of anti-proliferative effects of plant extract on AGS and HepG2 cancer cells. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2017, 1053, 1–8. [Google Scholar] [CrossRef]
  92. Bozkurt, E.; Atmaca, H.; Kisim, A.; Uzunoglu, S.; Uslu, R.; Karaca, B. Effects of Thymus serpyllum Extract on Cell Proliferation, Apoptosis and Epigenetic Events in Human Breast Cancer Cells. Nutr. Cancer 2012, 64, 1245–1250. [Google Scholar] [CrossRef]
  93. Ramos, A.A.; Azqueta, A.; Pereira-Wilson, C.; Collins, A.R. Polyphenolic compounds from Salvia species protect cellular DNA from oxidation and stimulate DNA repair in cultured human cells. J. Agric. Food Chem. 2010, 58, 7465–7471. [Google Scholar] [CrossRef]
  94. Caprioli, G.; Maggi, F.; Bendif, H.; Miara, M.D.; Cinque, B.; Lizzi, A.R.; Brisdelli, F.; Celenza, G. Thymus lanceolatus ethanolic extract protects human cells from t-BHP induced oxidative damage. Food Funct. 2018, 9, 3665–3672. [Google Scholar] [CrossRef] [PubMed]
  95. Al-Menhali, A.; Al-Rumaihi, A.; Al-Mohammed, H.; Al-Mazrooey, H.; Al-Shamlan, M.; Al-Jassim, M.; Al-Korbi, N.; Hussein Eid, A. Thymus vulgaris (Thyme) Inhibits Proliferation, Adhesion, Migration, and Invasion of Human Colorectal Cancer Cells. J. Med. Food 2015, 18, 54–59. [Google Scholar] [CrossRef] [PubMed]
  96. Bruno, A.; Pagani, A.; Pulze, L.; Albini, A.; Dallaglio, K.; Noonan, D.M.; Mortara, L. Orchestration of angiogenesis by immune cells. Front Oncol 2014, 4, 131. [Google Scholar] [CrossRef] [PubMed]
  97. Abdallah, Q.; Al-Deeb, I.; Bader, A.; Hamam, F.; Saleh, K.; Abdulmajid, A. Anti-angiogenic activity of Middle East medicinal plants of the Lamiaceae family. Mol. Med. Rep. 2018, 18, 2441–2448. [Google Scholar] [CrossRef] [PubMed]
  98. Jaksevicius, A.; Carew, M.; Mistry, C.; Modjtahedi, H.; Opara, E.I. Inhibitory effects of culinary herbs and spices on the growth of HCA-7 colorectal cancer cells and their COX-2 expression. Nutrients 2017, 9, 1051. [Google Scholar] [CrossRef] [Green Version]
  99. Ćebović, T.; Arsenijević, J.; Drobac, M.; Živković, J.; Šoštarić, I.; Maksimović, Z. Potential use of deodorised water extracts: Polyphenol-rich extract of Thymus pannonicus as a chemopreventive agent. J. Food Sci. Technol. 2018, 55, 560–567. [Google Scholar] [CrossRef] [Green Version]
Table 1. Antioxidant properties of phenolic-rich extracts of distinct Thymus species.
Table 1. Antioxidant properties of phenolic-rich extracts of distinct Thymus species.
Thymus PlantsOriginSolvent Extraction (Major Components or TPC)Results of Screen AssayRef
T. algeriensisAlgeriaEtOH (DHA, Rut, Epi)DPPH (EC50, mg/mL) = 1.56 (EtOH), 1.68 (BHA), 0.002 (AA)/ABTS (EC50, mg/mL) = 1.74 (EtOH), 0.003 (Tlx), 0.001 (AA)[45]
AlgeriaMeOH-H2O (RA, CaffeoylRA, Kaemp, Eri-Glc)Plasma antioxidant capacity (DPPH, 800 mg/kg bw): 22% of Inhib (treated group)/6 % (non-treated group)
Ferric reducing ability of plasma (FRAP, 800 mg/kg bw): 908 µM FeSO4 eq/mL (treated group)/405 µM FeSO4/mL (non-treated group)
↑ CAT activity; ↑ GSH levels (400 and 800 mg/kg bw); ↓ MDA levels (200 and 400 mg/kg bw)
[46]
TunisiaMeOH (Carvacrol, RA, Tetramethyl-scutellarein, Kaemp-O-Hexu)DPPH (EC50, μg/mL): 8.9–68.8/β-carot bleach (mg/mL): 0.03–1.81/FRAP (mmol Fe2+/L): 0.3–20.6[47,48]
T. atlanticusMoroccoH2O (RA, CaffA, Quer)DPPH (EC50, μg/mL) = 120 (H2O), 510 (Tlx)/FRAP (mmol Tlx/g extract) = 40.0 (H2O), 44.3 (Tlx)[43]
T. caespititiusPortugalH2O (RA, Lut-O-Glr)DPPH (EC50, μg/mL) = 13.8 (H2O), 6.90 (AA)/RP (EC50, μg/mL) = 39.3 (H2O), 16.30 (BHA)/β-carot bleach (EC50 μg/mL) = 6.1 (H2O), 0.4 (BHA)[40]
T. camphoratusPortugalDE, EA, n-but, H2O [TPC (GAE mg/mL) = 10.77 (DE), 10.21 (EA), 6.62 (n-but), 1.82 (H2O)]DPPH (EC50 μg/mL): 3.1 (DE), 2.7 (EA), 6.4 (n-but), 3.2 (H2O)/O2- scav (EC50 μg/mL): 7.8 (DE), 11.0 (n-but), 9.5 (H2O)[49]
T. capitatusGreece70% MeOH (RA, FA, Nar, Lut); H2O (RA, CaffA, Epi, Epig)DPPH (mg TE/g DW): 56.2/ABTS (mg TE/g DW): 75.2/FRAP (mg TE/g DW): 76.1[50]
T. carnosusPortugalDE, EA, n-but, H2O [TPC (GAE mg/mL) = 3.55 (DE), 5.97 (EA), 2.99 (n-but), 1.24 (H2O)]DPPH (EC50 μg/mL): 4.0 (DE), 3.0 (EA), 5.2 (n-but), 3.6 (H2O)/O2- scav (EC50 μg/mL): 12.3 (DE), 8.9 (EA), 8.9 (n-but), 13.6 (H2O)[49]
T. citriodorusPortugalEtOH-H2O (RA, SA I, Lut-O-Hexu)
H2O (RA, Lut-O-Hexu)
ABTS (EC50 mmol Tlx eq./g extract): 1.52 (EtOH-H2O); 1.21 (H2O)/OH scav (% inhib, 1 mg/mL): 37.97 (H2O)[51]
EtOH-H2O (RA, Lut-Glr Api-Glr)Intracellular ROS formation on HepG2 cells (EtOH-H2O 50 μg/mL): ↓ 21% (at 5 μM potassium dichromate-stimulated cells); ↓ 20% (at 25 μM potassium dichromate-stimulated cells)[52]
T. fragrantissimusPortugalH2O (RA, Lut-O-Glr, CaffeoylRA)DPPH (EC50, μg/mL) = 12.9 (H2O), 6.90 (AA)/RP (EC50, μg/mL) = 33.4 (H2O), 16.30 (BHA)[41]
T. herba-baronaPortugalH2O (RA, Lut-O-Glr, CaffeoylRA, SA B)DPPH (EC50, μg/mL) = 11.6 (H2O), 6.90 (AA)/RP (EC50, μg/mL) = 35.1 (H2O), 16.30 (BHA)/β-carot bleach (EC50 μg/mL) = > 26.7 (H2O), 0.4 (BHA)[40]
T. longicaulisCroatiaEtOH (PC: THA = 5.41%; TFlav = 0.40%)DPPH (EC50 μg/mL): 3.01 (EtOH), 0.66 (RA),0.73 (Lut), 1.67 (Tlx)/RP (EC50 μg/mL): 11.8 (EtOH), 2.67 (RA), 4.51 (Lut), 6.64 (Tlx)/TBARS (EC50 μg/mL): 34.3 (EtOH), 21.1 (RA), 2.03 (Lut)[4]
ItalyEtOH-H2O (RA, SA K, Lut-O-Hex, Quer-O-Hex)DPPH (EC50, μg/mL): 9.5 (H2O-MeOH), 5.1 (Tlx)/ABTS (EC50, μg/mL): 9.5 (H2O-MeOH), 5.1 (Tlx)/RP (μM TE/g extract): 475 (H2O-MeOH)/ORAC (μM TE/g extract): 776.5 (H2O-MeOH)[42]
T. mastichinaPortugalDE, EA, n-but, H2O [TPC (GAE mg/mL) = 26.28 (DE), 19.50 (EA), 9.74 (n-but), 2.23 (H2O)]DPPH (EC50 μg/mL): 2.7 (DE), 3.7 (EA), 4.0 (n-but), 3.9 (H2O)/O2- scav (EC50 μg/mL): 10.0 (DE), 4.9 (EA), 6.9 (n-but), 12.2 (H2O)[49]
MeOH, H2O [TPC (mg GAE/g) = 165.29; TF (mg CE/g) = 83.85]DPPH (EC50 mg/mL): 0.69 (MeOH), 2.57 (H2O), 0.04 (Tlx)/RP (EC50 mg/mL): 0.23 (MeOH), 0.7 (H2O), 0.03 (Tlx)/β-carot bleach (EC50 mg/mL): 0.9 (MeOH), 0.003 (Tlx)/TBARS (EC50 mg/mL): 0.43 (MeOH), 0.004 (Tlx)[53]
PortugalEtOH-H2O (RA, SA I)
H2O (RA, SA I)
ABTS (EC50 mmol Tlx eq./g extract): 1.48 (EtOH-H2O); 0.96 (H2O)/OH scav (% inhib, 1 mg/mL): 43.22 (EtOH-H2O); 48.52 (H2O)[54]
Spain50% MeOH (RA, CaffA, Lut, Lut-Glc)DPPH (mg TE/g DW): 18–149/FRAP (mg TE/g dw): 30–154, different populations[37]
T. nummulariusTurkeyMeOH (QA, RA, Lut, Kaemp)DPPH (EC50 μg/mL): 5.73 (MeOH), 1.21 (RA), 47.1 (BHT), 19.6 (α-Toc)/ABTS (EC50 μg/mL): 7.1 (MeOH), 1.7 (RA), 10.9 (BHT)/β-carot bleach (EC50 μg/mL): 6.54 (MeOH), 12.1 (RA), 9.95 (BHT)[44]
T. praecox subsp. polytrichus, T. serpyllum subsp. serpyllum, T. striatusCroatiaEtOH T. praecox [PC: THA = 54.39%; TFlav = 0.24%)/EtOH T. serpyllum (PC: THA = 4.36%; TFlav = 0.4%)/EtOH T. striatus (PC: THA = 3.35%; TFlav = 0.15%)DPPH (EC50 μg/mL): 3.4 (T. praecox), 4.06 (T. striatus), 6.01 (T. serpyllum) 0.73 (Lut), 1.67 (Tlx)/RP (EC50 μg/mL): 15.1 (T. praecox), 14.7 (T. striatus), 14.5 (T. serpyllum), 2.67 (RA), 4.51 (Lut), 6.64 (Tlx)/TBARS (EC50 μg/mL): 78.7 (T. praecox), 63.0 (T. striatus), 80.0 (T. serpyllum), 21.1 (RA), 2.03 (Lut)[4]
T. pseudolanuginosusPortugalH2O (RA, Lut-O-Glr, SA B)DPPH (EC50, μg/mL) = 10.9 (H2O), 6.90 (AA)/RP (EC50, μg/mL) = 32.2 (H2O), 16.30 (BHA)/β-carot bleach (EC50 μg/mL) = 2.4 (H2O), 0.4 (BHA)[40]
T. pulegioidesCroatiaEtOH (PC: THA = 6.17%; TFlav = 0.42%)DPPH (EC50 μg/mL): 4.18 (EtOH), 0.66 (RA), 0.73 (Lut), 1.67 (Tlx)/RP (EC50 μg/mL): 11.4 (EtOH), 2.67 (RA), 4.51 (Lut), 6.64 (Tlx)/TBARS (EC50 μg/mL): 34.8 (EtOH), 21.1 (RA), 2.03 (Lut)[4]
PortugalMeOH [TPC (mg GAE/g) = 210.49; TFlav (mg CE/g) = 128.24; TFlol (mg QE/g) = 126.74)]DPPH (EC50 μg/mL): 680/RP (EC50 μg/mL): 490/β-carot bleach (EC50 μg/mL): 30/TBARS (EC50 μg/mL): 220[55]
PortugalH2O (RA, Lut-O-Glr, CaffeoylRA)DPPH (EC50, μg/mL) = 9.5 (H2O), 6.90 (AA)/RP (EC50, μg/mL) = 30.7 (H2O), 16.30 (BHA)[41]
T. satureioidesMoroccoH2O (RA, CaffA, Quer)DPPH (EC50, μg/mL) = 440 (H2O), 510 (Tlx)/FRAP (mmol Tlx/g extract) = 40.1 (H2O), 44.3 (Tlx)[43]
T. sipyleus Boiss. subsp. rosulansMoroccoH2O [TPC (mg GAE/g) = 147.6 (decoction), 118.5 (infusion)]DPPH (EC50, μg/mL) = 43.5 (decoction), 87.38 (infusion), 27.63 (AA)[56]
T. vulgarisAlgeriaMeOH-H2O (TPC (mg GAE/100g DW) = 81.5; RA, Flav)DPPH (EC50 μg/mL): 1.78/ABTS (EC50 μg/mL): 0.69/OH scav (EC50 μg/mL): 0.24[57]
CroatiaEtOH (PC: THA = 3.58%; TFlav = 0.24%)DPPH (EC50 μg/mL): 5.6 (EtOH), 0.66 (RA),0.73 (Lut), 1.67 (Tlx)/RP (EC50 μg/mL): 14.1 (EtOH), 2.67 (RA), 4.51 (Lut), 6.64 (Tlx)/TBARS (EC50 μg/mL): 69.6 (EtOH), 21.1 (RA), 2.03 (Lut)[4]
EgyptEtOH [TPC (mg/g) = 212; TFlav (mg/g) = 85)Protective antioxidant effects against lead intoxicated rats: ↓ GSH, GPx and CAT levels; ↑ MDA levels[58]
PortugalEtOH-H2O (RA, SA I, Lut-O-Hexu)
H2O (RA, Lut-O-Hexu)
ABTS (EC50 mmol Tlx eq./g extract): 0.92 (EtOH-H2O); 0.79 (H2O)/OH scav (% inhib, 1 mg/mL): 9.58 (H2O)[51]
SlovakiaH2O, MeOH (RA, SA K isomer, Lut-Hex, Eri-Glc)DPPH (EC50, μg/mL): 44.7 (MeOH), 2.79 (Que)/ABTS (EC50 μg/mL): 13.8 (MeOH), 49.6 (H2O), 1.17 (Que)[59]
T. zygisMoroccoH2O (RA, CaffA, Quer)DPPH (EC50, μg/mL): 440 (H2O), 510 (Tlx)/FRAP (mmol Tlx/g extract): 65.0 (H2O), 44.3 (Tlx)[43]
PortugalH2O (RA, CaffeoylRA)]DPPH (EC50, μg/mL) = 12.6 (H2O), 6.90 (AA)/RP (EC50, μg/mL) = 33.7 (H2O), 16.30 (BHA)[41]
EtOH-H2O (RA, SA I, SA K)
H2O (RA, Lut-O-Hex, Lut-O-Hexu)
ABTS (EC50 mmol Tlx eq./g extract): 1.08 (EtOH-H2O); 0.76 (H2O)/OH scav (% inhib, 1 mg/mL): 66.28[60]
AA—ascorbic acid; ABTS—2,2′-azino-bis-(3-ethylbenzothiazoline-6-sulphonic acid) radical scavenging assay; BHA—2(3)-t-butyl-4-hydroxyanisole; BHT—3.5-di-tert-butyl-4 hydroxytoluene; CaffA/CaffA deriv—caffeic acid/caffeic acid derivative; CE—chatequin equivalents; β-carot bleach—β-carotene/linoleic acid bleaching inhibition assay; CAT—catalase; DE—diethyl ether; DHA—dihydroxybenzoic acid; DPPH—1,1-diphenyl-2-picrylhydrazyl radical scavenging assay; DW—dry weight; EA—ethyl acetate; Epi—epicatechin; Epig—epigallocatechine; Eri—eriodictyol; EtOH—ethanol; FA- ferulic acid; Flav—flavonoids; FRAP—ferric reducing antioxidant power; GAE—gallic acid equivalent; Glc—glucoside; Glr—glucuronide; GPx—glutathione peroxidase; GSH—reduced glutathione; Hex—hexoside; Hexu—hexuronide; Kaemp—kaempferol; Lut—luteolin; MDA—malonaldehyde; MeOH—methanol; Nar—naringin; n-but—n-butanol; ND—not determined; OH scav—hydroxyl radical scavenging assay; O2- scav—O2- radical scavenging assay; ORAC—oxygen radical absorbance capacity; QA—quinic acid; Quer—quercetin; QE—quercetin equivalents; RA—rosmarinic acid; RP—reducing power; Rut—rutin; SA—salvianolic acid; TBARS—thiobarbituric acid reactive substances assay; TE—trolox equivalente; TFlav—total flavonoids; TFlol—total flavanols; THA—total hydroxycinnamic acids; Tlx—trolox; TPC—total phenolic compounds; PC—phenolic compounds; α-Toc—α-tocopherol.
Table 2. Anti-inflammatory properties of phenolic-rich extracts of distinct Thymus species.
Table 2. Anti-inflammatory properties of phenolic-rich extracts of distinct Thymus species.
Plant SpeciesOriginSolvent Extraction (Major Components or TPC)Screen AssayEffectRef
T. algeriensisAlgeriaMeOH-H2O (SA K, RA-Glc, Lut-Glr)COX Inhib
5-LOX Inhib
Carrageenan-Induced Hind-Paw Edema Model
Carrageenan-Induced Leukocyte Migration (LeucM)
EC50 (μM) = COX-1: 12.4 (MeOH-H2O), 4.06 (dic); COX-2: 0.05 (MeOH-H2O), 0.06 (cel); 5-LOX: 2.7 (MeOH-H2O), 3.2 (zil)
Paw Edema: MeOH-H2O (200 mg/kg) = ↓~15%
LeucM: MeOH-H2O (600 mg/kg) = ↓62%;
dic (20 mg/kg) = ↓39%; dexa (2 mg/kg) = ↓30%
[68]
T. atlanticusMoroccoH2O (RA, CaffA, Quer)Croton oil-induced mice ear edema/Carrageenan-induced rat paw edemaAt 900 µg/ear after 8 h: ↓ 84.6% (in comparison to ind)/At 50 mg/kg after 5 h: ↓ 9.5% (in comparison to ind)[43]
H2O (ND)Inhib of denaturation of bovine serum albumin/Inhib of erythrocyte lysisEC50 μg/mL: 122.9 (H2O), 86.07 (ind)/EC50 μg/mL: 93.28 (H2O), 97.83 (ind)[62]
T. caespititiusPortugalH2O (RA, Lut-O-Glr)5-LOX inhib/NO scav5-LOX (EC50 μg/mL): 590.5 (H2O), 7.8 (AA)/NO (EC50 μg/mL): 229.7 (H2O), 228.0 (AA)[40]
T. camphoratus, T. carnosusPortugalDE, EA, n-but [TPC (GAE mg/mL) = 10.77 (DE), 10.21 (EA), 6.62 (n-but)]5-LOX inhib5-LOX (EC50 μg/mL): 29.9 (DE), 27.4 (EA), 28.0 (n-but) (T. camphoratus); 23.5 (DE), 29.6 (EA), 18.3 (n-but) (T. carnosus)[49]
T. carnosus Boiss.PortugalH2O (SA A isomer, SA K)
EtOH-H2O (SA A isomer, SA K, RA)
NO scav
in LPS-induced macrophages
H2O (200 μg/mL) = ↓ 90% of control; EtOH-H2O (15 μg/mL) = ↓ 75% of control[69]
T. fontanesiiAlgeriaMeOH-H2O (Carnosol, Salvigenin)COX Inhib, 5-LOX Inhib/Carrageenan-Induced Hind-Paw Edema Model/Carrageenan-Induced Leukocyte Migration (LeucM)EC50 (μM) = COX-1: 12.88 (MeOH-H2O), 4.06 (dic); COX-2: 0.04 (MeOH-H2O), 0.06 (cele); 5-LOX: 2.5 (MeOH-H2O), 3.2 (zil)/Paw Edema: MeOH-H2O (600 mg/kg) and dic (20 mg/kg) = ↓ 44%/LeucM: MeOH-H2O (600 mg/kg) = ↓ 52%; dic (20 mg/kg) = ↓ 39%; dexa (2 mg/kg) = ↓ 30%[68]
T. herba-baronaPortugalH2O (RA, Lut-O-Glr, CaffeoylRA, SA B)5-LOX inhib/NO scav5-LOX (EC50 μg/mL): 840.8 (H2O), 7.8 (AA)/NO (EC50 μg/mL): 286.1 (H2O), 228.0 (AA)[40]
T. longicaulisItalyEtOH-H2O (RA, SA K, Lut-O-Hex, Quer-O-Hex)COX-2 gene expression on THP-1 cellsAt 50 µg/mL: ↓ 42% (extract collected in october)[42]
T. mastichinaPortugalDE, EA, n-but [TPC (GAE mg/mL) = 10.77 (DE), 10.21 (EA), 6.62 (n-but)]5-LOX inhib5-LOX (EC50 μg/mL): 62.5 (DE), 53.1 (EA), 30.5 (n-but)[49]
T. praecox subsp. skorpilii var. skorpiliiTurkeyMeOH fraction (CA, Lut-O-Glc, 3-O-feruloylQA, Quer-O-Hex)Carrageenan-induced paw edema% inhib (MeOH f, 100 mg/kg) = 4.26% (1 h), 67.67% (2 h), 52.07% (3 h), 65.75% (4 h); % inhib (ind 5, mg/kg) = 34.04% (1 h), 88.65% (2 h), 82.89% (3 h), 88.63% (4 h)[70]
T. pseudolanuginosusPortugalH2O (RA, Lut-O-Glr, SA B)5-LOX inhib/NO scav5-LOX (EC50 μg/mL): 813.6 (H2O), 7.8 (AA)/NO (EC50 μg/mL): 298.98 (H2O), 228.0 (AA)[40]
T. satureioidesMoroccoH2O (ND)Inhib of denaturation of bovine serum albumin/Inhib of erythrocyte lysisEC50 μg/mL: 181.42 (H2O), 86.07 (ind)/
EC50 μg/mL: 204.41 (H2O), 97.83 (ind)
[62]
H2O (RA, Lut-7-Glc, Hesp)Croton oil-induced mice ear edemaAt 900 µg/ear after 8 h: ↓ 29.7% (in comparison to ind)[43]
T. sipyleus Boiss. subsp. rosulansMoroccoH2O [TPC (mg GAE/g) = 147.6 (decoction), 118.5 (infusion)]Inhib of NO and TNF-α production in LPS-induced macrophagesAt 50 μg/mL: ↓ NO: 50.86% (decoction) and 47.79% (infusion); ↓ TNFα: 49.76% (decoction) and 54.79% (infusion)[56]
T. vulgarisUnited KingdomH2O [TPC (mg GAE/g herb) = ~20]IL-8 release of PBLs prior to stimulation by TNFα and H2O2↓IL-8 release in 35 and 37% upon stimulation of TNF-α and H2O2, respectively[71]
BrazilPG (Thymol, Carvacrol, Linalool, Geranoil, Citral, Tannins,
Organic acids, Flavonoids)
Cytokines production by LPS-induced RAW264.7 macrophagesIL-1β (pg/mL) = 28, 2, 2 at 25, 50 and 100 mg/mL extract, respectively; TNF-α (pg/mL) = 4466, 824, 12 at 25, 50 and 100 mg/mL extract, respectively[72]
EgyptEtOH [TPC (mg/g) = 212;
TFlav (mg/g) = 85)]
Lead intoxicated ratsProtective effects against lead intoxicated rats: ↑ IL-1β, IL-6 and TNF-α levels; ↓ IL-10 and (IFN)-γ levels[58]
T. zygisMoroccoH2O (RA, CaffA, Lut-7-O-Glc)Croton oil-induced mice ear edema/Carrageenan-induced rat paw edemaAt 900 µg/ear after 8 h: ↓ 70% (in comparison to ind)/At 50 mg/kg after 5 h: ↓ 3.7% (in comparison to ind 10 mg/kg)[43]
H2O (ND)Inhib of denaturation of bovine serum albumin/Inhib of erythrocyte lysisEC50 μg/mL: 133.25 (H2O), 86.07 (ind)/EC50 μg/mL: 156.20 (H2O), 97.83 (ind)[62]
PortugalEtOH-H2O (RA, SA I, SA K); H2O (RA, Lut-O-Glc, L-O-Hexu)NO scav/Inhib of NO production in LPS-induced macrophagesNO scav (% inhib): 29.32 (H2O)/At 50 µg/mL: ~89% (EtOH), 48% (H2O-H2O)[60]
CA—chlorogenic acid; CaffA—caffeic acid; Celecoxibe—cele; COX-2—cyclooxygenase; DE—diethyl ether; Dex—dexamethasone; Dic—diclofenac; EA—ethyl acetate; EtOH—ethanol; GAE—allic acid equivalent; Glc—glucoside; Glr—glucuronide; Gly—glucosyl; Hesp—hesperetin; Hex—hexoside; Hexu—hexuronide; IFN—interferon γ; IL—interleukin; LOX—lipoxygenase; Inhib—Inhibition; Ind—indomethacin; LPS—lipopolysaccharide; Lut—luteolin; MeOH—methanol; n-but—n-butanol; ND—not determined; NO—nitric oxide; PBL—peripheral blood lymphocytes; PG—propylene glycol; QA—quinic acid; Quer—quercetin; RA—rosmarinic acid; SA—salvianolic acid; Scav—scavenging; TFlav—total flavonoids; THP-1—human monocytic leukemia cell line; TNF-α—tumor necrosis factor-α; TFlav—total flavonoids; TPC—total phenolic compounds; Zil—zileuton.
Table 3. Anticancer properties of phenolic-enriched extracts of distinct Thymus species.
Table 3. Anticancer properties of phenolic-enriched extracts of distinct Thymus species.
Plant SpeciesOriginSolvent extraction (Major Components or TPC)Screen AssayEffectRef
T. algeriensisNIMeOH (GallicAc, VanillicAc)CViab (MTT) on U266 cell lineCVI (%): ~15%[90]
T. caramanicusIranEtOH-H2O (Carvacrol, Thymol, Borneol, Cymene)CViab (MTT) on MCF-7 cells/Biochemical markers of apoptosis and cell proliferation (Western blot)CVI (%) = MCF-7: 85% (EtOH-H2O), 85% (Vin), 65% (EtOH-H2O+Vin) after 40 µg/mL extract; MCF-7: 70% (EtOH-H2O), 85% (Vin), 50% (EtOH-H2O+Vin), after 80 µg/mL extract/Western blot = MCF-7 after 200 µg/mL extract: ↑ caspase 3, ↑ bax, ↓Bcl2, ↓ cyclin D1[30]
T. carnosus Boiss.PortugalH2O (SA A isomer, SA K)
EtOH-H2O (SA A isomer, SA K, RA)
CViab (Alamar Blue) on MCF-7, BT-474, RAW 264.7CVI (IC50 µg/mL, H2O) = MCF-7: 841.28 (24 h); 735.18 (48 h); BT-474: 533.87 (24 h); 603.86 (48 h); RAW 264.7: 603.07 (24 h); 223.22 (48 h);
CVI (IC50 µg/mL, EtOH-H2O) = MCF-7: 86.87 (24 h); 74.37 (48 h); BT-474: 39.91 (24 h); 34.45 (48 h); RAW 264.7: 24.80 (24 h); 28.20 (48 h)
[69]
T. citriodorusPortugalEtOH-H2O (RA, SA I, Lut-O-Hexu)
H2O (RA, Lut-O-Hexu)
CViab (Alamar Blue) on Caco-2 and HepG2CVI (IC50 µg/mL, EtOH-H2O) = Caco-2: 128.2 (24 h); 114.6 (48 h); HepG2: > 500 (24 h); >500 (48 h); CVI (IC50 µg/mL, H2O) = Caco-2: 223.7 (24 h); 159.4 (48 h); HepG2: >500 (24 h); >500 (48 h)[51]
T. mastichinaPortugalEtOH-H2O; H2O (RA, SA A isomer, Lut-Hex, Quer-Hex)CViab (Alamar Blue) on Caco-2 and HepG2 cellsCVI (IC50 µg/mL, EtOH-H2O) = Caco-2: 71.18 (24 h); 51.30 (48 h); HepG2: 264.60 (24 h); 180.10 (48 h); CVI (IC50 µg/mL, H2O) = Caco-2: 220.60 (24 h); 95.65 (48 h); HepG2: >500 (24 h); 285.03 (48 h)[54]
T. pulegioidesPortugalEtOH-H2O; H2O (RA, Lut-O-Hexu, Eri-O-Hexu, Chr-Hex)CViab (Alamar Blue) on Caco-2 cellsCVI (IC50 µg/mL) = 82.25 (H2O); 105.44 (EtOH-H2O)[61]
T. satureioidesMoroccoH2O (RA, Lut-O-Glc)CViab (MTT) on MCF-7CVI (IC50 µg/mL) = 37.5 ± 4.02[43]
T. schimperiEthiopiaMeOH 70% (Lut, Lut-7-O-Glc, Lut-7-O-xy)CViab (MTT) on AGS and HepG2 cellsCVI (IC50, µg/mL) = AGS: 88, after 50–100 µg/mL extract; HepG2: 326, after 200–400 µg/mL extract/CVI (%) = AGS: 38%, HepG2: 35%[91]
T. serpyllum (Ts), T. vulgaris (Tv)PolandTs H2O (RA, CaffA, LAc, Lut-7-O-Glr, Lut-7-O-Rut, Eri-7-O-Rut)
Tv H2O (RA, CaffA, Lut-7-O-Glr, Lut-7-O-Rut, Eri-7-O-Rut)
CViab (MTT) on MCF-7/Adr or wt cellsCVI (IC50, µg/mL) = MCF-7/Adr: 399 (Ts), 407 (Tv)/(IC50, mM) = MCF-7/Adr: 0.81 (RA), 1.26 (LAc), 1.81 (CaffA), 1.87 (Lut-7-O-Glr), 4.2 (Lut-7-O-Rut), 2.6 (Erd-7-O-Rut), 5.8 (Ab); MCF-7/wt: 0.74 (RA), 1.09 (Lut-7-O-Glr), 0.45 (LAc), 1.36 (CaffA), 18.2 (Lut-7-O-Rut), 1.71 (Eri-7-O-Rut), >1000 (Ab)CVI (%) = MCF-7/Adr: 60% (Ts, Tv), MCF-7/wt: 30% (Ts), 60% (Tv) at 500 mg/L extract/MCF-7/Adr: 86% (RA), 26% (Lut-7-O-Glr), MCF-7/wt: 92% (RA), 80% (LAc), 54% (Lut-7-O-Glr), at 1.25 mM[89]
T. serpyllumTurkeyMeOH (ND)CViab (XTT) on MCF-7 and MDA-MB-231 cells/DNA fragmentation/caspase 3/7 enzyme activityCVI (%) MCF-7: ↓ 15, 32, 42.5%, and 71 at 10, 100, 500, and 1000 μg/mL of extract, respectively
CVI (%) MDA-MB-231: ↓ 22, 33, 66, and 75 at 10, 100, 500, and 1000 μg/mL of extract, respectively
DNA fragmentation of MDA-MB-231 cells: ↑ 16%, 30%, and 55% at 10, 100, 250, and 500 μg/mL extract
Caspase 3/7 enzyme activity: ↑ 1.6-, 2.2-, 3-fold at 10, 250, 500 μg/mL
[92]
T. vulgarisBrazilPG (Thymol, Carvacrol, Linalool, Geranoil, Citral, Tannins,
Organic Acids, Flavonoids)
CViab (MTT, NR and CVA) on RAW 264.7, FMM-1, MCF-7 and HeLa cellsCVI, MTT (%) = RAW 264.7 and FMM-1: ↓ at 25, 50 and 100 mg/mL; MCF-7: ↓ at 50 and 100 mg/mL; HeLa cells: ↓ at 100 mg/mL extract
CVI, NR (%) = RAW 264.7: ↓ at 50 and 100 mg/mL; FMM-1: ↓ no significant; MCF-7: ↓ at 25, 50 and 100 mg/mL; HeLa cells: ↓ at 50 and 100 mg/mL extract
CVI, CVA (%): RAW 264.7: ↓ at 50 and 100 mg/mL; FMM-1: ↓ at 25, 50 and 100 mg/mL; MCF-7: ↓ at 100 mg/mL; HeLa cells: ↓ at 50 mg/mL extract
[72]
PortugalEtOH-H2O (RA, SA I, Lut-O-Hexu)
H2O (RA, Lut-O-Hexu)
CViab (Alamar Blue) on Caco-2 and HepG2 cellsCVI (IC50 µg/mL, EtOH-H2O) = Caco-2: >500 (24 h); 442.45 (48 h); HepG2: 495.05 (24 h); 254.25 (48 h)/CVI (IC50 µg/mL, H2O) = Caco-2: >500 (24 h); 376.8 (48 h); HepG2: >500 (24 h); >500 (48 h)[51]
SlovakiaEtOH (RA, SA K isomer, Lut-Hex, Api-Glr)CViab (MTT), DNA damage (comet assay); enzymatic activity of tumor HepG2 cellsCVI (IC50, mg/mL) = 4.3/HepG2 DNA damage induced by H2O2 and DMNQ: ↓ after 0.5 mg/mL extracts, at 24 h/Enzymatic activity: ↑ GPx, ↓ SOD, after 1 and 0.5 mg/mL extracts[59]
South AfricaAcet (ND)CViab (XTT) on HeLa and non-tumor Vero cellsCVI (IC50, µg/mL) = HeLa: >200, 0.002 (AmD); Vero: 138, 0.027 (AmD)[86]
T. zygis subsp. zygisPortugalEtOH-H2O (RA, SA I, SA K)CViab (Alamar Blue) on Caco-2 and HepG2 cellsCVI (IC50 µg/mL) = Caco-2: 85.01 ± 15.10; HepG2: 82.19 ± 2.46 µg/mL[60]
Ac—acid; Acet—acetone; Adr—adriamycin-resistant; AGS—human gastric adenocarcinoma cells; AmD—actinomycin D; Ab—arbutin; BT-474—breast cancer cell line; CaffA—caffeic acid; CaCo-2—colon adenocarcinoma cell line; Chr—chrysoeriol; CViab—cell viability; CVA—crystal violet assay; CVI—cell viability inhibition; DMNQ—2,3-dimethoxy-1,4-naphthoquinone; Eri—eriodictiol; FMM-1—human gingival fibroblasts; Glc—glucoside; Glr—glucuronide; GPx—glutathione peroxidase; Hex—hexoside; Hexu—hexuronide; HeLa—cervical carcinoma cells; HepG2—hepatocellular carcinoma cells; LAc—lithospermic acid; Lut—luteolin, MeOH—methanol; MCF-7—human breast carcinoma cells; MDA-MB-231—breast cancer cell line; MTT—3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide; ND—not determined; NI—not indicated; NR—neutral red assay; PG—propylene glycol; Quer—quercetin; RA—rosmarinic acid; Rut—rutinoside; SA—salvianolic acid; SOD—superoxide dismutase; U266—multiple myeloma cell line; Vin—vincristine; Wt—wild-type; Xy—xyloside; XTT—2,3-bis-(2-methoxy-4- nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide.

Share and Cite

MDPI and ACS Style

Afonso, A.F.; Pereira, O.R.; Cardoso, S.M. Health-Promoting Effects of Thymus Phenolic-Rich Extracts: Antioxidant, Anti-inflammatory and Antitumoral Properties. Antioxidants 2020, 9, 814. https://doi.org/10.3390/antiox9090814

AMA Style

Afonso AF, Pereira OR, Cardoso SM. Health-Promoting Effects of Thymus Phenolic-Rich Extracts: Antioxidant, Anti-inflammatory and Antitumoral Properties. Antioxidants. 2020; 9(9):814. https://doi.org/10.3390/antiox9090814

Chicago/Turabian Style

Afonso, Andrea F., Olívia R. Pereira, and Susana M. Cardoso. 2020. "Health-Promoting Effects of Thymus Phenolic-Rich Extracts: Antioxidant, Anti-inflammatory and Antitumoral Properties" Antioxidants 9, no. 9: 814. https://doi.org/10.3390/antiox9090814

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