Next Article in Journal
Standardization of the Optimum Effects of Indole 3-Butyric Acid (IBA) to Control Root Knot Nematode, Meloidogyne enterolobii, in Guava (Psidium guajava L.)
Next Article in Special Issue
The Role of Natural and Semi-Synthetic Compounds in Ovarian Cancer: Updates on Mechanisms of Action, Current Trends and Perspectives
Previous Article in Journal
Effects of Long-Term Intervention with Losartan, Aspirin and Atorvastatin on Vascular Remodeling in Juvenile Spontaneously Hypertensive Rats
Previous Article in Special Issue
Synthesis and In Vitro Anticancer Evaluation of Flavone—1,2,3-Triazole Hybrids
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Potential of Phenolic Compounds in Medicinal Plants—Natural Health Products for Human Health

by
Wenli Sun
*,† and
Mohamad Hesam Shahrajabian
Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2023, 28(4), 1845; https://doi.org/10.3390/molecules28041845
Submission received: 6 January 2023 / Revised: 11 February 2023 / Accepted: 13 February 2023 / Published: 15 February 2023

Abstract

:
Phenolic compounds and flavonoids are potential substitutes for bioactive agents in pharmaceutical and medicinal sections to promote human health and prevent and cure different diseases. The most common flavonoids found in nature are anthocyanins, flavones, flavanones, flavonols, flavanonols, isoflavones, and other sub-classes. The impacts of plant flavonoids and other phenolics on human health promoting and diseases curing and preventing are antioxidant effects, antibacterial impacts, cardioprotective effects, anticancer impacts, immune system promoting, anti-inflammatory effects, and skin protective effects from UV radiation. This work aims to provide an overview of phenolic compounds and flavonoids as potential and important sources of pharmaceutical and medical application according to recently published studies, as well as some interesting directions for future research. The keyword searches for flavonoids, phenolics, isoflavones, tannins, coumarins, lignans, quinones, xanthones, curcuminoids, stilbenes, cucurmin, phenylethanoids, and secoiridoids medicinal plant were performed by using Web of Science, Scopus, Google scholar, and PubMed. Phenolic acids contain a carboxylic acid group in addition to the basic phenolic structure and are mainly divided into hydroxybenzoic and hydroxycinnamic acids. Hydroxybenzoic acids are based on a C6-C1 skeleton and are often found bound to small organic acids, glycosyl moieties, or cell structural components. Common hydroxybenzoic acids include gallic, syringic, protocatechuic, p-hydroxybenzoic, vanillic, gentistic, and salicylic acids. Hydroxycinnamic acids are based on a C6-C3 skeleton and are also often bound to other molecules such as quinic acid and glucose. The main hydroxycinnamic acids are caffeic, p-coumaric, ferulic, and sinapic acids.

1. Introduction

Medicinal plants are very important worldwide, both when used alone and as a supplement to traditional medication [1,2,3,4,5]. For many years, humans have employed plants as a source of food, flavoring, and medicines [6,7,8,9,10]. Various parts of medicinal plants such as seeds, leaves, flowers, fruits, stems, and roots are rich sources of bioactive compounds [11,12,13]. Bioactive compounds should be considered as important dietary supplements [14,15,16,17,18,19]. Polyphenols are a group of secondary metabolites involved in the hydrogen peroxide scavenging in plant cells [20]. Phenolic compounds are second only to carbohydrates in abundance in higher plants, and they display a great variety of structures, varying from derivatives of simple phenols to complex polymeric materials such as lignin [21,22,23,24,25,26]. Phenolic compounds are known for their notable potential activity against various human viruses, and phenolic compounds also have immunomodulatory and anti-inflammatory activity [27]. The most abundant phenolic compounds are phenolic monoterpenes (carvacrol and thymol) and diterpenes (carnosol, carnosic acid, and methyl carnosate), hydroxybenzoic acids (p-hydroxybenzoic, protocatechuic, gallic, vanillic, catechol, and ellagic), phenylpropanoic acids (p-coumaric, caffeic, rosmarinic, chlorogenic, ferulic, cryptochlorogenic, and neochlorogenic), phenylpropenes (eugenol), coumarins (herniarin and coumarin), flavanoes (naringenin, eriocitrin, naringin, and hesperidin), flavones (apigenin, apigetrin, genkwanin, luteolin, luteolin 7-glucuronide, cynaroside, scolymoside, salvigenin, and cirsimaritin), and flavanols (catechin, astragalin, kaempferol, methyl ethers, quercetin, hyperoside, isoquercetin, miquelianin, and rutin) [28,29].
Plant phenolics are considered promising antibiofilm and antifungal agents [30,31]. Diaz et al. [32] also reported that the levels of phenolic and flavonoid compounds were correlated with the anti-inflammatory and antioxidant activities of medicinal plants. Tukun et al. [33] reported that phenolic content is significantly connected to antioxidant activity, and halophytes have high content of nutrients and phenolic metabolites. Some of the most important phenolic compounds recognized from medicinal plants are syringic acid and gallic acid from Moringa oleifera [34]; gallic acid, vanillic acid, 4-hydroxybenzoic acid, and syringic acid from Peganum harmala [35]; rosmarinic acid from Rosmarinus officinalis L. and Mentha canadensis L. [36]; vanillin from Thymus vulgaris [37]; caffeic acid and p-coumaric acid from Ocimum basilicum L., Thymus vulgaris L., Salvia officinalis L., and Origanum vulgare L. [36]; piceatannol glucoside, resveratroloside, and piceid from Polygonum cuspidatum [38]; trans-rhapontin, cis-rhapontin, and trans-desoxyrhaponticin from Rheum tanguticum Maxim. Ex Balf. [39]; herniarin from Matricaria chamomilla [40]; kayeassamin I, mammeasin E, and mammeasin E from Mammea siamensis [41]; scopoletin, fraxetin, aesculetin, fraxin, and aesculin from Fraxinus rhynchophylla [42]; phyllanthin, niranthin, hypophyllanthin, nirtetralin, virgastusin, heliobuphthalmin lactone, and bursehernin from Phyllanthus amarus [43]; schisanchinin A, schisanchinin B, schisanchinin C, and schisanchinin D from Schisandra chinensis [44]; 7-methyljuglone from Drosera rotundifolia [45], rhein, physcion, chrysophanol, emodin, and aloe-emodin from Rheum palmatum and Rheum hotaoense [46]; curcumin, demethoxycurcumin, and bis-demethoxycurcumin from Curcuma longa [47]; luteolin, apigenin, orientin, apigenin-O-glucuronide, and luteolin-O-glycoside from Origanum majorana [48]; glycitein, genistein, formononetin, daidzein, prunetin, biochanin A and daidzin, and genistin from Medicago spp. [49]; kaempferol 3-O-glucoside and isorhamnetin 3-O-galactoside from Tephrosia vogelii [50]; rutin, kaempferol 3-O-rhamnoside, and quercetin 3-O-glucoside from M. oleifera [34]; gallocatechin and catechin from Mentha pulegium [48]; taxifolin, taxifolin methyl ether, and dihydrokaempferide from Origanum majorana [48]; hesperidin, naringenin-O-rhamnoglucoside, and isosakuranetin-O-rutinoside from Mentha pulegium [48]; and punicalagin, pedunculagin I, granatin A, ellagic acid, ellagic acid pentoside, ellagic acid glucoside, and punigluconin from Punica granatum [51]. Phenolic phytochemicals include flavonoids, flavonols, flavanols, flavanones, flavones, phenolic acids, chalcones, isoflavones, tannins, coumarins, lignans, quinones, xanthones, curcuminoids, stilbenes, cucurmin, phenylethanoids, and several other plant compounds, owing to the hydroxyl group bonded directly to an aromatic hydrocarbon group [52]. The classes of phenolic compounds in plants are shown in Table 1.
Phenolic acids include two subgroups, i.e., hydroxybenzoic and hydroxycinnamic acids [53]. Hydroxybenzoic acids consist of gallic, p-hydroxybenzoic, vanillic, protocatechuic, and syringic acid, which, in common, have the C6-C1 structure [53]. Hydroxycinnamic acids, on the other hand, are aromatic compounds with a three-carbon side chain (C6-C3), with caffeic, p-coumaric, ferulic, and sinapic acids being the most common [52]. Gallic acid is present in cloves (Eugenia caryophyllata Thunb.), while protocatechuic acid can be found in coriander (Coriandrum sativum L.), dill (Anethum graveolens L.), and star anise (Illicium verum Hook. f.) [54]. Caffeic acid is found among others in parsley (Petroselinum crispum L.), ginger (Zingiber officinale Rosc.), and sage (Salvia officinalis L.), and p-coumaric acid is found in oregano (Origanum vulgare L.), basil (Ocimum basilicum L.), and thyme (Thymus vulgaris L.) [54]. Some samples of hydroxybenzoic and hydrozycinnamic acids are presented in Table 2.
Flavonoids include the largest group of plant phenolics, responsible for over half of the eight thousand naturally occurring phenolic constituents [55,56]. Flavonoids are low molecular weight compounds, including fifteen carbon atoms, arranged in a C6-C3-C6 configuration [53]. The genetic structure of main classes of flavonoids are shown in Table 3.
Phenolic phytochemicals play a variety of protective roles against abiotic stresses, such as UV light, or abiotic stresses, namely predator and pathogen attacks [57]. Phenolic phytochemicals are utilized by humans to treat several ailments including bacterial, protozoal, fungal, and viral infections, inflammation, diabetes, and cancer. Biosynthesis and accumulation of polyphenol and other secondary metabolites in plants is considered as an evolutionary reaction of biochemical pathways under adverse environmental influences, i.e., biotic/abiotic limitations, including increased salinity and drought stress [58,59,60]. Some of the extraction methodologies of phenolic components from medicinal and aromatic plants are maceration, digestion, infusion, decoction, Soxhlet extraction, percolation, aqueous alcoholic extraction by fermentation, counter-current extraction, ultrasound extraction, supercritical fluid extraction, and phytonics stage. The principle factors shaping the production of phenolic components are the water supplied to plants and the time of stress exposure, and, among the various quantification methods, HPLC and colorimetric tests are the most utilized to quantify the phenolic compounds analyzed [61]. Djeridane et al. [62] reported that the phenolics in medicinal plants provide substantial antioxidant activity. A positive, significant linear connection between antioxidant activity and total phenolic content revealed that phenolic components were the dominant antioxidant constituents in medicinal plants [63,64]. Various groups of tests on phenolics indicated significant mean alterations in radical scavenging activity; tannins demonstrated the strongest activity, while most quinones, isoflavones, and lignans tested revealed the weakest activity [65,66]. The most abundant flavone in Cytisus multiflorus is the chrysin derivative, Kaempferol-3-O-rutinoside is the major flavonol in Malva sylvestris, and Quercetin-3-O-rutinoside is the principle flavonol in Sambucus nigra [66]. Nepeta italica subsp. cadmea and Teucrium sandrasicum are rich in phenolics, which indicated antioxidant and cytotoxic properties [67]. Through LC-ESI-MS analysis, five phenolic acids (quinic acid, syringic acid, gallic acid, p-coumaric acid, and trans-ferulic acid) and five flavonoids (catechin, epicatechin, quercetrin, rutin, and naringenin) were predominant and common in some desert shrubs of Tunisian flora (Pituranthos tortuosus, Ephedra alata, Retama raetam, Ziziphus lotus, Calligonum comosum, and Capparis spinosa) [68].
The main phenolic compounds in Matico (Piper angustifolium R.), Guascas (Galinsoga parviflora), and Huacatay were chlorogenic acid and hydroxycinnamic acid derivatives [69]. High phenolic and antioxidant activity-containing medicinal plants and species such as Chanca Piedra (Phyllanthus nirui L.), Yerba Mate (Ilex paraguariensis St-Hil), Zarzaparrilla (Smilax officinalis), and Huacatay (Tagetes minuta) have the highest anti-hyperglycemia-relevant in vitro α-glucosidase inhibitory activities with no effect on α-amylase [69]. Nineteen phenolic compounds from different groups are used in wound treatment, and the compounds are tyrosol, curcumin, hydroxytyrosol, luteolin, rutin, chrysin, kaempferol, quercetin, icariin, epigallocatechin gallate, morin, silymarin, taxifolin, hesperidin, naringin, puerarin, isoliquiritin, genistein, and daidzein [70,71,72,73]. The most important identified phenolics in Phlomis angustissima and Phlomis fruticosa, medicinal plants from Turkey, by RP-HPLC-DAD were hesperidin, catechin, kaempferol, epicatechin, eupatorin, and epigallocatechin, and chlorogenic, syringic, vanillic, p-coumaric, ferulic, and benzoic acids [74]. Quercetin of Cordia dichotoma G. Forst. (Lashusa) is the most notable phytoconstituent responsible for the therapeutic efficacy [75]. Vanillic acid, nepetin, verbascoside, and hispidulin, of Clerodendrum petasites S. Moore (CP) were chosen as potential phenolic active compounds in Thai traditional medicine for the treatment of different kinds of skin diseases [76,77,78]. Bouyahya et al. [79] reported that compounds such as terpenoids, alkaloids, flavonoids, phenolic acids, and fatty acids of Arbutus unedo L., Thymus capitatus managed diabetes by several mechanisms such as enzymatic inhibition, interference with glucose and lipid metabolism signaling pathways, and the inhibition and the activation of gene expression involved in glucose homeostasis.
Grewia tenax, Terminalia sericea, Albizia anthelmintica, Corchorus tridens, and Lantana camara are frequently used to treat gastroenteritis and include higher total phenolic and flavonoid contents in Namibia [80,81,82,83,84,85]. The most important phenolics identified from pomegranate are punicalin, gallic acid, ellagic acid, pyrogallol, salycillic acid, coumaric acid, vanillic acid, sesamin, and caffeic [86], and phenolic compounds have been discovered to have inhibitory effects again α-glucosidase activities [87]. Two new phenolics, leucoxenols A and B, were obtained and identified as major secondary metabolites from the leaves of Syzygium leucoxylon [88]. Phenolics are main phytochemicals found in Cyathea species, and Cyathea has been considered to be a potential source of novel cancer therapeutic compounds [89]. Purified phenolic compounds from the bark of Acacia nilotica showed insecticidal potential against Spodoptera litura, and they could provide substitutes to synthetic pesticides for controlling various pests [90]. Bellumori et al. [91] reported that the roots of Acmella oleracea L. had about twice as many phenols as the aerial parts, and caffeic acid derivatives were the main phenolic compounds in roots and aerial parts. Kaempferol was found as the most abundant phenolic compound in basil leaf extract after using an HPLC-UC method (61.4 mg.kg−1) [92]. Apple fruit (Annona squamosa L.) has a specific spatial distribution of microbes and phenolics, its peel phenolics contain antimicrobial activity against several Gram-positive bacteria, and its peel phenolics had a growth-promoting effect toward autochthonous yeasts [93,94,95,96]. The phenolic contents of Cyathea dregei (root and leaves), Felicia erigeroides (leaves and stems), Felicia erigeroides (leaves and stems), Hypoxis colchicifolia (leaves), Hypoxis colchicifolia (leaves), and Senna petersiana (leaves) have shown high antimicrobial and cyclooxygenase (COX) inhibitory activities [97].
The most important techniques for analysis of phenolic compounds and extracts are nuclear magnetic resonance (NMR), high performance liquid chromatography (HPLC) with ultraviolet-visible (UV-Vis) or photodiode array (PDA) detector or coupled to mass spectrometry (MS), derivatization (silylation, alkylation, etc.) as well as gas chromatography (GC) or GC-MS analysis, phytochemical screening such as total flavonoid content (TFC), total phenolic content (TPC), etc., and antioxidant potential tests such as 2,2-dipehnyl-1-picrylhydrazyl (DPPH), etc. [97,98,99,100,101,102,103,104,105,106,107]. Solid-liquid extraction (SLE) is one of the main methods for extraction of phenolic compounds, specially syringic acid, catechin, and p-coumaric acid, which is simple, well established, and widely used [108]. Ultrasound-assisted extraction (UAE) is often used for extraction of gallic acid and rutin, which is easy to execute, uses inexpensive equipment, and consumes less solvents, and has fast extraction, good extraction yield, and low impacts on the environment [109]. Supercritical fluid extraction (SFE) usually applies for gallic acid, anthocyanin, and protocatechuic acid, which has high selectivity, cheaper and safer solvent, easily controlled extraction conditions, environmental friendliness, low operating temperature, and easy separation of solvent from solutes [110]. Microwave-assisted extraction (MAE) is used for extraction of 3-caffeoylquinic acid, 5-caffeoylquinic acid, and ellagic acid, which has short extraction time and low solvent consumption [111]. Pressurized liquid extraction (PLE) applies for extraction of rutin and quercetin, which consumes fewer organic solvents, has higher probability to avoid organic solvents by using water only, and is fast and efficient [112]. For extraction of proanthocyanidin, naringin, and hesperidin, enzyme-assisted extraction (EAE) is proposed, which is safe and green and does not need complex paraphernalia [113]. Key points about phenolic acids and their derivatives are shown in Table 4. This work aims to provide an overview of phenolic compounds and flavonoids as potential sources of pharmaceutical and medical application from recently published studies, as well as some interesting directions for future research.

2. The Important Health Benefits of Phenolic Components

Flavonoids and phenolics are commonly known as the largest phytochemical molecules with antioxidant characteristics [124]. Traditional Chinese medicinal plants that contain phenolic acids and flavonoids have shown high antioxidant activity. Nepeta italica subsp. Cadmea and Teucrium sandrasicum are rich in phenolic, tannin, and flavonoids content, which showed antioxidant and cytotoxic properties. Bauhinia variegata L. contained flavonoid compounds and revealed antioxidant properties against oxidative damage by radical neutralization, iron binding, and decreasing power abilities [125]. The rhizome extracts of Polygonatum verticillatum (L.) All. exhibited antioxidant activity, which is connected to the level of phenolic composition [126]. Singh and Yadav [127] have reported that, among medicinal plants, oregano, clove, thyme, and rosemary contain the highest amounts of phenolic compounds. Flavan-3-ol oligomers and monomers were potent antioxidant compounds abundantly identified in Camellia fangchengensis [128].
Bellis perennis L. was rich in phenolic compounds, and it can be used for wounds, cancer, inflammation, and eye diseases [129]. A total of 27 kinds of phenolic compounds were identified by HPLC-ESI-QTOF-MS/MS, and okra (Abelmoschus esculentus) polyphenols exhibited great antioxidant activity in vitro [130]. The Althaea officinalis extracts showed stronger antioxidant activity and excellent α-glucosidase, 5-lipoxygenase, and nitric oxide inhibitory properties [131]. Dendrobium densiflorum was rich in flavonoid, alkaloid, and antioxidant activity, Acampe papillosa was rich in total phenol, total tannin, and total saponin content, and Coelogyne nitida exhibited higher antioxidant activity because of its higher quercetin content [132]. Cirak et al. [133] showed that Achillea arabica Kotschy is an important source of natural antioxidants. The antioxidant property and bioactive constituents from the fruits of Aesculus indica (Wall. Ex Cambess.) Hook, which were quercetin and mandelic acid, were the major bioactive molecules with notable antioxidant properties to decrease oxidative stress caused by reactive oxygen species (ROS) [134]. The phytochemical compounds and biological activity of Pinus cembra L. contain higher concentration of total phenolics and flavonoids than that of needle extract, and its bark extract showed better ability as a free radical scavenger [135]. Higher antioxidant activity in normal-tannin lentil seed coats than low-tannin ones was reported; kaempferol tetraglycoside was dominant in low-tannin seed coats, and procyanidins, kaempferol tetraglycoise, and catechin-3-O-glucoside in normal-tannin has been found [136]. Zhang et al. [137] also reported that antioxidant activity and prebiotic impacts were positively correlated for oat phenolic compounds. 3,4-dihydroxybenzoic, rutin, vanillic acid, and quercetin were detected from aqueous extracts of azendjar and taamriouth figs, and a dark peel variety consisted of more phenolics and exerted a higher antioxidant capacity [138]. Although gallic acid was the most important compound in carob (Ceratonia siliqua L.) pulp extract, geographic origin strongly influenced the contents of bioactive compounds and antioxidant activities [139].
Asplenium nidus L. contained gliricidin 7-O-hexoside and quercetin-7-O-rutinoside that can fight against three pathogens, i.e., Proteus vulgaris Hauser, Proteus mirabilis Hauser, and Pseudomonas aeruginosa (Schroeter) Migula [140]. Flavones, which were extracted from the root of Scutellaria baicalensis Georgi, were proven as potential antibacterial agents against Propionibacterium acnes-induced skin inflammation both in in vitro and in vivo models [141]. Kaempferol that was isolated from the Impatiens balsamina L. exhibited potential activity to inhibit the growth of P. acnes [142]. Phenolics from kernel extract Mangifera indica L. also showed anti-acne properties to inhibit the growth of P. acnes [143]. Medicinal plants such as Albizia procera, Atalantia monophylla, Asclepias curassavica, Azima tetracantha, Cassia fistula, Costus speciosus, Cinnamomum verum, Nymphaea stellata, Osbeckia chinensis, Punica granatum, Piper argyrophyllum, Tinospora cordifolia, and Toddalia asiatica have shown antifungal activity [144]. The strictinin isolated from the leaves of Camellia sinensis var. assamica (J.W. Mast.) Kitam was a good substitute for antibacterial activities [145]. Phenolic compounds, especially flavonoids, have long been reported as chemopreventive factors in cancer therapy [146,147,148]. The extract of Curcuma longa L. rhizome has been suggested as a promising source of natural active compounds to fight against malignant melanoma due to its potential anticancer property in the B164A5 murine melanoma cell line [149]. Glircidia 7-O-hexoside and Quercetin 7-O-rutinoside, which were flavonoids isolated from the medicine fern (Asplenium nidus), were also proposed as potential chemopreventives against human hepatoma HepG2 and human carcinoma HeLa cells [140]. Quercetin can induce miR-200b-3p to regulate the mode of self-renewing divisions of the tested pancreatic cancer [150], and a soy isoflavone genistein inhibited the activation of the nuclear factor kappa B (NF-KB) signaling pathway that maintains the balance of cell survival and apoptosis; this soy isoflavone could also take its action to fight against cell growth, apoptosis, and metastasis, including epigenetic modifications in prostate cancer [151]. Curcumin exhibits anticancer impacts towards skin cancers, as this phenolic can influence the cell cycle by acting as a pro-apoptotic agent [152]. Curcumin acts as a non-selective cyclic nucleotide phosphodiesterase (PDE) inhibitor to inhibit melanoma cell proliferation, which is associated with epigenetic integrator UHRF1 [153]. Curcumin inhibited proliferation of the selected cell lines in prostate cancer and induced apoptosis of the cancer cells with a dose-dependent response [154].
The cardioprotective impacts from various kinds of phenolics and flavonoids occurring in medicinal plants have been investigated in many studies [155,156]. Many phenolic and flavonoid compounds have been studied and had reported their cardioprotective properties via different mechanisms including inhibition of ROS generation, apoptosis, mitochondrial dysfunction, NF-KB, p53, and DNA damage both in vitro and in vivo, and clinical studies [157]. Kaempferol, luteolin, rutin, and resveratrol showed their efficacy against doxorubicin-induced cardiotoxicity [158,159]. Isorhamnetin provided a cardioprotective effect against cardiotoxicity of doxorubicin and potentiated the anticancer efficacy of this drug [160]. The total phenolic and flavonoid contents of the aqueous fraction from Marrubium vulgare L. have effects on ischemia-reperfusion injury of rat hearts, which proved that the aqueous fraction from M. vulgare had cardioprotective potential [156]. Aspalathin and phenylpyruvic acid-2-O-β-D-glucoside, two of the major compounds from Aspalathus linearis (Burm.f.) R. Dahlgren, were demonstrated as potential protective compounds to protect myocardial infarction caused by chronic hyperglycemia [155]. Puerarin is a potential isoflavone that was reported as an interesting candidate for cardioprotection by protecting myocardium from ischemia and reperfusion damage by means of opening the Ca2+-activated K+ channel and activating the protein kinase C [161]. Quercetin, hesperidin, apigenin, and luteolin were reported as flavonoids containing potential anti-inflammatory impacts [162]. The flavonoids and phenolic compounds of Phyllanthus acidus leaves could be correlated with the analgesic, antioxidant, and anti-inflammatory activities [163]. Hydroxytyrosol and quercetin 7-O-α-L-rhamnopyranoside exhibited anti-inflammatory activity through lowering the levels of TNF-α, and hydroxytyrosol and caffeic acid showed significant anti-inflammatory activity at 100 μm by reducing the release of NO in LPS-stimulated macrophages comparable to positive control indomethacin [164].
The most important chemical compounds extracted from ethanol of Cardiospermum halicacabum were chrysoeriol, kaempferol, apigenin, luteolin, methyl 3,4-dihydroxybenzoate, 4-hydroxybenzoic acid, quercetin, hydroquinone, protocatechuic acid, gallic acid, and indole 3-carboxylic acid, which have shown high anti-inflammatory and antioxidant activities [165]. The most important phenolic components with antiviral effects against COVID-19 were curcumin, Theaflavin-3,3′-digallate, EGCG, Paryriflavonol A, Resveratrol, Quercetin, Luteolin, Scutellarein, Myricetin, and Forsythoside A [166]. In traditional Persian medicinal science, medicinal plants such as Glycyrrhiza glabra L., Rheum palmatum L., Punica granatum L., and Nigella sativa L. have been introduced for treating respiratory disorders and infections because of their phenolic compounds [167]. The anti-inflammatory activity of polyphenolic compounds in Gaillardia grandiflora Hort. Ex Van Houte and Gaillardia pulchella Foug from Egypt were reported [168]. Anti-inflammatory properties of two medicinal plant species, Bidens engleri O.E. Schulz from Asteraceae family as well as Boerhavia erecta L. from Nyctaginaceae family, were identified and reported in various fractions [169]. Plantago subulata has shown anti-inflammatory properties on macrophages and a protective effect against H2O2 injury [170]. Phenolic content changes with aromatic and medicinal plant species and extraction method used [171]. Astilbin, a dihydroflavonol, from Smilax glabra Roxb significantly inhibited nitric oxide production, tumor necrosis factor-α (TNF-α), and mRNA expression of inducible nitric oxide synthase in the tested cells [172]. Apigenin is a main flavone with skin protective impact against UV light; this flavone can be identified in various edible medicinal plants or plants-derived beverages, e.g., beer, red wine, and chamomile tea [173,174]. Quercetin is a flavonol that can be discovered in apple peel, onion skin, and Hypericum perforatum L. leaves [175]. Silymarin, a standardized extract of flavonolignans from the milk thistle (Silybum marianum (L.) Gaernt.) fruits, consists of silybin, a principle active component [176]. Genistein is a soybean isoflavone that was also reported as photoprotective molecule against photocarcinogenesis by inhibiting UV-induced DNA damage in human skin-equivalent in vitro model [177]. Equol is considered as an isoflavonoid metabolite from isoflavone daidzein or genistein produced by gut microflora [178,179]. Genistein is an obvious example of an interesting choice of a flavonoid phytoestrogen for improving endothelial roles in postmenopausal women with MetS [180]. A chrysin derivative was the most abundant flavone in Cytisus multiflorus, quercetin-3-O-rutinoside was the main flavonol in Sambucus nigra, and kaempferol-3-O-rutinoside was the main flavonol in Malva sylvestris [181]. Biological properties of phenolic compounds are presented in Table 5.

3. Hydroxybenzoic Acids (Gallic Acid and Protocatechuic Acid)

Hydroxybenzoic acids (HBAs) are antioxidant phytochemicals found in many medicinal plants and are efficient for prevention of various human diseases [247,248]. Joshi et al. [249] reported that 4-hydroxybenzoic acid (4HBA) is a potential antidiabetic, anticancer, antifungal, antioxidant, and cardioprotective, etc. Piper garagaranum C. DC contains prenylated hydroxybenzoic acids, and prenylated hydroxybenzoic acids indicated anti-inflammatory characteristics, as determined in murine macrophage assays [250].

3.1. Gallic Acid

Gallic acid is one of the most abundant polyphenols identified in nature [251,252]. Behera et al. [253] reported that gallic acid reveals antioxidant or free radical scavengers in adipocyte proliferation. Gallic acid is found in a wide range of natural plants, it is associated with the health of human beings, and it has well-documented anticancer, antibacterial, anti-inflammatory, and antifungal activities [254,255]. Gallic acid in Emblica officinalis mediated antidiabetic potential and delineated the upregulation of pAkt, PPAR-γ, and Glut4 through gallic acid-mediated antidiabetic properties, thus providing potent therapy for diabetes [256]. Gallic acid inhibited about 44–57% of the total CaOx crystal formations, and it is a promising agent with antiurolithiatic properties for the treatment and prevention of urinary or kidney stones [257]. Gallic acid supplementation adjusted serum lipid metabolism by decreasing serum triglyceride, fat digestibility, and bacteroidetes/firmicutes ratio [258]. Gallic acid prevents the development and occurrence of gastric precancerous lesions (GPL) by inhibiting the Wnt/β-catenin signaling pathway and then suppressing the epithelial–mesenchymal transition (EMT) process [259]. Gallic acid is a direct thrombin inhibitor with a platelet aggregation inhibitory effect [260]. Gallic acid shows significant binding and disruption of protease structure, and gallic acid has a potential phytotherapeutic effect against fungal protease, which is a notable virulence factor [261]. Gallic acid can boost gut microbiota alterations connected with cardiovascular disease (CVD) and suggests that males suffering from atherosclerosis may benefit from gallic acid supplementation, as this polyphenol partially restored microbiome dysbiosis [262]. Gallic acid could decrease the noxious impacts of diclofenac (DIC) on the antioxidant defense system and renal tissue [263].

3.2. Protocatechuic Acid

Protocatechuic acid (3,4-dihydroxybenzoic acid) is a natural phenolic acid, and one of the chief metabolites of complex polyphenols [264]. It can be identified in many plants such as bran and grain brown rice, particularly in the scales of onion, plums, grapes, gooseberries, and nuts such as ordinary almonds [265,266]. Da-Costa-Roch et al. [267] and Adedara et al. [268] reported that protocatechuic acid can be found in many medicinal plants, especially Hibiscus sabdariffa L. (Hs, roselle; Malvaceae). Protocatechuic acid has different activities such as neuroprotective activities, antiosteoporotic activities, antitumor activities, and the protective effects against hepatotoxic and nephrotoxic activities [269,270]. It has also antibacterial, antiulcer, anti-aging, antidiabetic, anticancer, antiviral, antifibrotic, analgesic, anti-inflammatory, anti-atherosclerotic, and cardiac activity [271,272]. Protocatechuic acid from bitter melon (Momordica charantia) alleviates cisplatin-induced oxidative renal damage, which proves it has protective activity against anticancer drug-induced oxidative nephrotoxicity [273]. Protocatechuis acid inhibits Cd-induced neurotoxicity in rats, increases the Nrf2 signaling pathway, and exhibits anti-apoptotic and anti-inflammatory activities [274]. Veronica montana has protocatechuic acid as the main phenolic molecule, and it kills bacteria by affecting its cytoplasmic membrane [275].

4. Hydroxycinnamic Acids (p-Coumaric Acid, Caffeic Acid, Ferulic Acid, Sinapic Acid)

Hydroxycinnamic acid derivatives are a notable class of polyphenols found in vegetables, fruits, and medicinal plants, and extensively consumed in human diet [276,277]. Hydroxycinnamic acids significantly contribute to antioxidant capacity [278]. Hydroxycinnamic acids are widely found in plants and their products such as cereals, fruits, coffee, vegetables, etc. [279,280].

4.1. p-Coumaric Acid

p-Coumaric acid is a plant metabolite with antioxidant and anti-inflammatory impacts [281,282]. p-Coumaric acid boosts hepatic fatty acid oxidation and fecal lipid excretion, and it affects inflammatory and insulin resistance-related adipokines. p-Coumaric acid stimulates electrical factors of biological and model lipid membranes [283].

4.2. Caffeic Acid

Caffeic acid (3,4-dihydroxycinnamic acid) has been known as an important source of natural antioxidants in different agricultural products [284,285]. It has immense use in cancer treatment [286,287], and it could be known as an important natural antioxidant [288]. Caffeic acid can induce apoptosis in cancer cells through increasing ROS levels and impairing mitochondrial function, and it also benefits from reducing aggressive behavior of tumors via suppressing metastasis [289]. Caffeic acid has anti-inflammatory and antioxidant properties against 6-propyl-thiouracil (PTU)-induced hypothyroidism [290]. Meinhart et al. [291] reported that higher sums of mono-caffeoylquinic acids were found in mulberry, quince, and bilberry, and the dicaffeoylquinic acids sum was higher in granadilla, passion fruit, and kumquat. It is a phenolic compound extensively discovered in commonly consumed foods such as apples, pears, and coffee [292]. The biosynthesis pathway of caffeic acid can be categorized into two modules, (1) L-tyrosine is synthesized from carbon sources via the glycolytic pathway, the pentose phosphate pathway, and the shikimate pathway; (2) caffeic acid is generated by the continuous deamination and hydroxylation of L-tyrosine [293]. Trifan et al. [294] found that caffeic acid oligomers reported in Symphytum officinale L. root may contribute to the anti-inflammatory activity for which comfrey preparations are used in traditional medicine. Caffeic acid phenethyl ester extracted from Rhodiola sacra could provide health benefits, decreasing the magnitude of the inflammatory process triggered by endotoxin shock and the production of inflammatory mediators [295]. Caffeic acid from the leaves of Annona coriacea have shown antidepressant-like impacts, which involve important neurotransmitter systems [296]. Spagnol et al. [297] reported that caffeic acid presented antioxidant activity greater than ascorbic acid and trolox. Caffeic acid regulates lipogenesis-related protein expression in high-fat diet (HFD)-fed mice, alleviates endotoxemia and the proinflammatory response in HFD-fed mice, and attenuates gut microbiota dysbiosis in HFD-fed mice [298]. Caffeic acid decreases oxidative stress levels in the hippocampus and regulates microglial activation in the hippocampus [299].

4.3. Ferulic Acid

Ferulic acid (4-hydroxy-3-methoxycinnamic acid) is a polyphenol that is widely known for its therapeutic potential, showing anti-aging, anti-inflammatory, and neuroprotective impacts [300,301]. The ferulic acid molecule reveals cis-trans isomerism, with the most abundant form in nature being the trans isomer, and both isomers have proven results in the treatment of several pathologies such as diabetes, cancer, and neurodegenerative and cardiac diseases [302]. Ferulic acid is important for the synthesis of significant chemical molecules such as coniferyl alcohol, di ferulic acid, vanillin, synaptic, and curcumin, as well as for giving the cell wall stiffness [303]. Ferulic acid can be applied as an antioxidant to prevent damage from ultraviolet (UV) radiation and skin carcinogenesis [304]. It is ample in numerous fruits and vegetables, including bananas, eggplant, citrus fruits, and cabbage, as well as in seeds and leaves [305,306]. In Chinese medicinal science, ferulic is normally joined with polysaccharides by covalent bonds in various plant cell walls such as cereal bran and regarded as the main bioactive compound of Angelica sinensis, chuanxiong rhizoma, and ferula [307], and it has several biological activities such as anti-apoptosis, anticancer, antioxidant, and anti-inflammatory impacts [308]. Free ferulic acid is related to the natural content of ferulic acid in herbs, and total ferulic acid refers to the sum of free ferulic acid plus the amount of related hydrolyzed components [309,310]. Angelica sinensis is a perennial herbaceous species that creates the bioactive metabolite ferulic acid [311,312]. The ferulic compounds of Salvia officinalis could be useful as a safe natural source for estrogenic characteristics [313]. Singh et al. [314] indicated that ferulic acid is a phenol derivative from natural sources and applied it as a potential pharmacophore that exerts multiple pharmacological properties such as neuroprotection, Aβ aggregation modulation, antioxidant, and anti-inflammatory. Ferulic acid increases cerebellar functional and histopathological changes induced by diabetes, which can be attributed to its antioxidative effect and its ability to modulate nitric oxide synthase (NOS) isoforms [315]. Ramar et al. [316] showed that ferulic acid and resveratrol revealed antioxidant as well as antidiabetic effects, consequently modulating liver, kidney, and pancreas damage caused by alloxan-induced diabetes, possibly via inhibition of the proinflammatory factor, NF-KB. Ferulic acid treatment prevents radiation-induced lipid peroxidation and DNA damage and restores antioxidant status and histopathological alterations in experimental animals [317]. Hu et al. [318] found that ferulic acid could alleviate inflammation and oxidative stress. Ferulic acid can inhibit cancer proliferation through various mechanisms, including changing the cancer cell cycle, inducing apoptosis, and regulating proteins involved in cell proliferation [319], and ferulic acid could be used as a potential official adjuvant for breast cancer treatment [320].

4.4. Sinapic Acid

Sinapic acid, a widely prevalent hydroxycinnamic acid, contains numerous biological activities related to its antioxidant property [321,322]. It protects lysosomes and prevents lysosomal dysfunction [323]. Saeedavi et al. [324] reported that sinapic acid may be a new therapeutic potential to treat allergic asthma through suppressing T-helper 2 immune responses. Sinapic acid phenethyl ester boosts gene expression related to the cholesterol metabolic process [325]. Hu et al. [326] indicated that sinapic acid can be utilized as an effective chemo preventive agent against lung carcinogenesis. It can also alleviate blood glucose levels by improving insulin production in pancreatic β-cells, and it can exhibit an antioxidative impact by suppressing lipid peroxidation and increasing the activity of antioxidant enzymes [327]. Sinapic acid significantly increases caspase-3 activity and inhibits cell invasion, and it has anticancer impacts on prostate cancer cells [328]. Sinapic acid pretreatment mitigates renal impairment and structural injuries through the downregulation of oxidative/nitrosative stress, inflammation, and apoptosis in the kidney [329]. Raish et al. [330] indicated the ability of sinapic acid to restore the antioxidant system and to suppress oxidative stress, pro-inflammatory cytokines, extracellular matrix, and TGF-β, and showed that sinapic acid treatment (10 and 20 mg/kg) significantly ameliorated bleomycin (BML)-induced lung injuries. Singh and Verman [331] revealed that sinapic acid increases streptozotocin (STZ)-induced cognitive impairment by ameliorating oxidative stress and neuro inflammation in the cortex and hippocampus. Sinapic acid can modulate the redox state in high-fat diet (HFD) rats [332].

5. The Health Benefits of Coumarins (Umbelliferone, Esculetin, Scopoletin)

Coumarins (2H-chromen-2-one ring) with the molecular formula C9H6O2 are an important group of natural compounds and are used as additives in both cosmetics and foods [333], and they constituent a notable class of heterocyclic compounds with the characteristic benzo-α-pyrone moiety in its structure [334]. Coumarin has been reported to have antibacterial, anticancer, antioxidant, anti-inflammatory, anticoagulant, and anti-Alzheimer’s disease (AD) activities [335,336]. Coumarin derivatives are found naturally as secondary metabolites in more than 150 species of plants and in over 30 plant families such as Clusiaceae, Umbelliferae, Guttiferae, Rutaceae, Oleaceae, Fabaceae, and many more [337]. Seo et al. [338] reported that different coumarins were identified from the roots of Angelica dahurica using NMR spectroscopy, and each coumarin revealed remarkable differences in content and inhibitory effect. Kassim et al. [339] indicated that the good antioxidant activity of Melicope glabra (Rutaceae) is because of umbelliferone, glabranin, and scopoletin. Coumarin-based compounds extracted from the medicinal plants are shown in Table 6.

5.1. Umbelliferone

Umbelliferone is a 7-hydroxycoumarin and an isomer of caffeic acid [341], and it has been reported for different pharmacological activities against numerous diseases such as cancer [342]. The plant sources of umbelliferone are Acacia nilotica, Angelica decursiva, Aegle marmelos, Artemesia tridentata, Aster praelatus, Balsamocitrus camerunensis, Chamomilla recutita, Citrus aurantium, Cirtus natsudaidai, Citrus paradise, Coriandrum sativum, Diospyros oocarpa, Diplostephium foliosissimum, Dystaenia takeshimana, Edgeworthia chrysantha, Edgeworthia gardneri, Eriostemon apiculatus, Ferula communis, Ferula communis, Ferula assafoetida, Fructus Aurantii, Glycyrrhiza glabra, Angelica archangelica, Haplophyllum villosum, Harbouria trachypleura, Haplopappus desertzcola, Haplophyllum patavinum, Hydrangea chinensis, Hydrangea macrophylla, Hieracium pilosella, Ipomoea mauritiana, Justicia pectoralis, Matricaria recutita, Melicope glabra, Musa spp., Parkinsonia aculeata, Peucedanum praeruptorum, Picea abies, Potentilla evestita, Rhododendron lepidotum, Platanus acerifolia, Selaginella stautoniana, Saussurea eopygmaea, Stellera chamaejasme, and Typha domingensis [343]. It has been reported to have antioxidant, anti-inflammatory, free radical scavenging, and antihyperglycemic properties [344], and antifungal characteristics [345]. Althunibat et al. [346] reported that umbelliferone prevented isoproterenol cardiotoxicity in rats, and it decreased isoproterenol-induced oxidative stress and inflammation. Kutlu et al. [347] reported that umbelliferone has a strong antioxidant and anti-inflammatory effect on sepsis, and it can be considered as a new treatment for organ dysfunction. Umbelliferone ameliorates atopic dermatitis (AD)-associated symptoms and inflammation via regulation of various signaling pathways, suggesting that umbelliferone might be a potential therapeutic of AD [348]. Umbelliferone downregulates TGF-β1 levels in kidney tissue and it may promote kidney function and ameliorate renal oxidative stress [349]. Mohamed et al. [350] indicated that umbelliferone ameliorated oxidative stress-related hepatotoxicity via its ability to augment cellular antioxidant defenses by activating Nrf2-mediated HO-1 expression. Umbelliferone exhibits anticancer impacts on human oral carcinoma (KB) cell lines, with the increased generation of intracellular reactive oxygen species (ROS) triggering oxidative stress-mediated depolarization of mitochondria [351]. Umbelliferone has gastric protective activity in vivo, and it has antidiarrheal activity in vivo [352].

5.2. Esculetin

Esculetin (6,7-dihydroxycoumarin), a natural coumarin derived from herbs, has shown different pharmacological activities [353]. Kadakol et al. [354] reported that esculetin, a naturally occurring 6,7-dihydroxy derivative of coumarin, has revealed its potential function in various non-communicable diseases (NCDs) including obesity, diabetes, renal failure, cardiovascular disease, cancer, and neurological disorders. Esculetin reduced both chronic and acute topic skin inflammation, and mitigated inflammation by suppressing infiltration of inflammatory cells [355]. It can be found in many medicinal plants such as Artemisia capillaris, Matricaria chamomilla L., Artemisia scoparia, Citrus limonia, Cortex Fraxini, and Ceratostigma willmottianum [356,357,358]. Esculetin supplementation could protect against development of non-alcoholic fatty liver in diabetes via regulation of glucose, lipids, and inflammation [359]. The esculetin protects human hepatoma HepG2 cells from hydrogen peroxide-induced oxidative injury, and the production is provided via the induction of protective enzymes as part of an adaptive response mediated by Nrf2 nuclear accumulation [360]. Esculetin prevents progressive renal fibrosis under insulin resistance (IR) and type 2 diabetic nephropathy (T2D) conditions, and it decreases oxidative stress in the kidney under IR and T2D conditions [361]. Esculetin has the ability to suppress tumor growth and metastasis via Axin2 suppression, which can be an attractive therapeutic strategy for the treatment of metastatic colorectal cancer (CRC) [362]. Esculetin treatment decreased neurological defects and improved cognitive impairments in transient bilateral common carotid artery occlusion (tBCCAO)-treated mice, and the mechanism underlying the pharmacological impacts of esculetin involved its action on mitochondrial autophagy and the apoptosis triggered by mitochondrial oxidative stress via mediation of mitochondrial fragmentation during transient cerebral ischaemia and reperfusion injury [363]. Zhang et al. [364] reported that esculetin could be a potential therapeutic drug for the treatment of hepatic fibrosis by inducing stellate cell senescence. Wang et al. [365] indicated that esculetin is safe and reliable, is easy to be absorbed by the body, and can be synthesized in a variety of ways. Esculetin inhibits the pyroptosis of microvascular endothelial cells through the NF-KB/NLFP3 signaling pathway and is expected to be conducive in treating pyroptosis-related diseases [366]. Esculetin directly binds to hnRNPA1 and decreases the concentration of hnRNPA1 in endometrial cancer cells, and it downregulates the levels of BCL-XL and XIAP expression, resulting in apoptosis and an arrest in proliferation [367]. Esculetin inhibits clear cell renal cell carcinoma growth in a dose- and time-dependent manner, and it induces apoptosis and cell cycle arrest [368]. Esculetin could be used as a dietary therapy for the prevention of alcoholic liver disease, and it can markedly prevent ethanol-induced liver injury in mice [369].

5.3. Scopoletin

Scopoletin (6-methoxyl-7-hydroxy coumarin) has a phenolic hydroxyl structure and is a member of the coumarin family [370]. It has a long history of use for its medicinal characteristics in traditional Chinese medicine [371]. Scopoletin is one of the main bioactive components of Convolvulus prostratus Forssk, known to have a role in acetylcholinesterase inhibitor, antimicrobial, memory enhancer, and antioxidative properties [372]. It is a major component of noni (Morinda citrifolia L.), which contributes to the anti-inflammatory, antioxidative, immunomodulatory, and hepatoprotective properties [373]. Scopoletin could be a potential phagocytic enhancer, and it can increase immunity through enhancing macrophage phagocytic capabilities [374]. Scopoletin improved vancomycin-induced renal injury via restoring the antioxidant defense system [375]. Scopoletin reduces non-alcoholic fatty liver disease in high-fat diet-fed mice [376]. It has been reported that scopoletin could exert a positive impact on anti-aging related to autophagy via modulation of p53 in human lung fibroblasts [377].

6. The Health Benefits of Stilbenes (Resveratrol, Piceatannol, Pterostilbene)

Stilbenes (based on the 1,2-diphenylethylene skeleton) are a group of plant polyphenols with rich structural and bioactive diversity [378]. They originate from plant families such as Vitaceae, Gnetaceae, Leguminaceae, and Dipterocarpaceae, and, structurally, they have a C6-C2-C6 skeleton, normally with two isomeric forms [379,380]. They have wonderful potential for anti-inflammatory, antiviral, anticancer, and antioxidant activities, as well as an application as cosmetic materials, coloring agents, and dietary supplements [381,382,383]. Wine and grapes are the main dietary source of stilbenes [384]. These compounds are synthetized by plants in response to abiotic or biotic stress situations [385]. Most stilbene compounds reveal antimicrobial properties, acting as phytoalexins in response to pathogen or herbivore attack [386]. Phytochemical phenols of stilbene families indicated good stability at elevated temperatures [387].

6.1. Resveratrol

Resveratrol (3,5,4′-trihydroxy-trans-stilbene) is a plant polyphenol, extensively popularized during the last decades, owing to its promising beneficial effects on human health [388]. It is a famous non-flavonoid polyphenol, related to the family of stilbenes whose structure consists of two phenolic rings linked by a double bond, which promotes two isomeric conformations: trans- and cis-resveratrol [389,390]. Resveratrol’s cis-isomer is unstable, and its trans-isomer contains greater stability, but converts to the cis-isomer under exposure to high pH or UV light [391,392], with heat increasing the degradation process [391]. It exists in many traditional herbs, and in several types of fruits, especially in the muscadine grape, red wine, cranberry, lingonberry, and redcurrant [393], and roots of various plant species including Polygonum cuspidatum and rhubarb (Rheun rhapontiicum) [394]. It is also useful in common age-related diseases such as cancer, cardiovascular diseases, type 2 diabetes, and neurological conditions, and it has also positive impacts on metabolism and can boost the lifespan of various organisms [395]. Resveratrol supplementation can be considered as an adjuvant therapy for relieving inflammation [396]. It has great potency in treating cardiovascular diseases [397]. Resveratrol attenuates kidney damage in malignant hypertension rats, and it can increase glomerular filtration while decreases proteinuria [398]. It inhibits the release of proinflammatory cytokines and leads to the release of anti-inflammatory cytokines, and it scavenges free radicals and upregulates antioxidant enzymes [399]. Chowdhury et al. [400] indicated that resveratrol treatment indicated beneficial impacts on preventing oxidative stress and fibrosis in the kidneys of high-fat (HF) diet-fed rats, probably by modulating the gene expression of oxidative stress and inflammation-related parameters and enzymes. Resveratrol can downregulate the pro-inflammatory cytokine release decreasing lung injury [401]. Resveratrol-containing fruits could be a promising substitute for the management of Alzheimer’s disease [402]. It can be more effective in cardiotoxicity prevention [403]. Polygonum cuspidatum is an important medicinal plant in China and a rich source of resveratrol compounds, which is a secondary metabolite formed in the long-term evolution procedure of plants to increase their response to adverse environments such as pathogens and ultraviolet radiation [404]. As an anticancer parameter, resveratrol promotes apoptosis in hepatocellular carcinoma cells [405]. Bhaskara et al. [406] reported that resveratrol is a potential reducing factor that can prevent carcinogenesis due to its antioxidant abilities, and it acts as an immunomodulatory agent for treating cancer. Resveratrol can exhibit anti-aging activity through a variety of signaling pathways [407]. Resveratrol shows potent anti-rotavirus efficacy in vitro and in vivo, and it blocks viral structural expression and genomic RNA synthesis [408]. Resveratrol oligomers from Paeonia suffruticosa indicate neuroprotective effects in vitro and in vivo by regulating cholinergic, antioxidant, and anti-inflammatory pathways, and they may have promising applications in the treatment of Alzheimer’s disease [409]. Resveratrol is also involved in neurodegenerative diseases (NDs) with multiple neuroprotective activities [410]. Antimicrobial activity of resveratrol against many bacteria and fungi has been reported, such as antimicrobial activity against Gram-positive bacteria such as Bacillus cereus, Bacillus megaterium, Staphylococcus aureus, Enterococcus faecalis, Enterococcus faecium, Mycobacterium tuberculosis, Mycobacterium smegmatis, Streptococcus pneumoniae, Streptococcus pyogenes, Propionibacterium acnes, and Listeria monocytogenes; against Gram-negative bacteria such as Escherichia coli, Klebsiella pneumoniae, Salmonella enterica serovar Typhimurium, Pseudomonas aeruginosa, Helicobacter pylori, Arcobacter butzleri, Arocobacter cryaerophilus, Haemophilus ducreyi, Neisseria gonorrhoeae, Neisseria meningitidis, Vibrio cholerae, Fuscobacterium nucleatum, Campylobacter jejuni, and Campylobacter coli; and against fungi such as Trichophyton mentagrophytes, Trichophyton tonsurans, Trichophyton rubrum, Epidermophyton floccosum, Microsporum gypseum, Candida albicans, Saccharomyces cerevisiae, Botrytis cinerea, and Trichosporon beigelii [411]. Resveratrol has powerful anticancer characteristics in different cancer cells and organs such as pancreatic cancer, colorectal cancer, gastric cancer, esophageal cancer, hepatocellular cancer, oral cancer, and biliary tract cancer [412]. Resveratrol decreases damage to pancreatic tissue via suppression of calcium overload; it suppresses calcium overload and, thereby, decreases trypsinogen activation, oxidative stress, mitochondrial dysfunction, and disorders, and it also reduces damage to other organs such as lung and heart by decreasing microcirculatory dysfunction [413].

6.2. Piceatannol

Piceatannol (3,4,3′,5′-tetrahydroxy-trans-stilbene), a natural polyphenolic stilbene, has pleiotropic pharmacological potentials [414]. It can be found in different kinds of fruits and vegetables such as blueberries, grapes, and passion fruit [415]. Piceatannol is a metabolite of resveratrol found in red wine, which prevents cardiac hypertrophy in rat neonatal cardiomyocytes [416]. It has previously been known as an antileukemic principle, which has been shown to be an inhibitor of protein-tyrosine kinase activity [417]. It has been reported that its low water-solubility and bioavailability could limit its application in both food and pharmaceutical fields [418]. Piceatannol, compared with the renowned resveratrol, is a better anticancer factor and a superior agent with other biological properties [419]. Piceatannol lightened oxidative injury and collagen synthesis in lung tissues during pulmonary fibrosis, and it suppressed the activation and collagen synthesis of TGF-β-induced lung fibroblasts [420]. It appears to be an appropriate nutritional or pharmacological biomolecule that modulates effector T cell functions, namely cytokine production, differentiation, and proliferation [421]. Piceatannol attenuates fat accumulation in steatosis-induced HepF2 cells, it suppressed lipogenesis and fatty acid uptake in steatosis-induced HepG2 hepatocytes, and it suppressed fatty acid-induced oxidative stress [422]. It shows anti-aggregation activity, and it increases catalase and glutathione peroxidase activity [423]. It can also be considered as a potential chemotherapeutic factor in the treatment of leukemia, but it may be connected with the risk of multi-drug resistance [424]. Passion fruit seed extract and piceatannol could exert anticancer activity via human glyoxalase I (GLO I) inhibition [425]. Piceatannol is a promising medication for preventing acute liver failure and the mechanisms may be associated to its inhibitory impacts on ER stress, inflammation, and oxidative stress [426]. Piceatannol has a potential inhibitory activity against human glyoxapase I (GLO I), and it inhibits the proliferation of GLO I-dependent human lung cancer [427]. It protects ARPE-19 cells against apoptosis induced by photo-oxidation, and the protective effect of piceatannol is because of the activation of the Nrf2/NQO1 pathway [428]. Piceatannol is a potent enhancer of cisplatin-induced apoptosis, and it reveals the potential for clinical development for the treatment of ovarian cancer [429]. It has been reported that piceatannol significantly decreases the degree of bovine serum albumin (BSA) glycosylation, and this suggests its potential impact on preventing the progression of diabetes mellitus [430].

6.3. Pterostilbene

Pterostilbene, a dimethyl ester derivative of resveratrol, may act as a cytotoxic and anticancer factor [431]. It primarily exists in blueberries, grapevines, and heartwood of red sandalwood [432,433]. Phenolic resveratrol, pterostilbene has been reported to have antifungal activity against a broad range of important phytopathogenic fungi such as Leptosphaeria maculans and Peronophythora litchii [434]. It is an anti-inflammatory and antioxidant agent with preventive effects toward skin disorders, and its anticancer impacts include inducing necrosis, apoptosis, and autophagy [435]. It can alleviate hepatic damage and oxidative stress and increase hepatic antioxidant function in piglets [436]. It possesses the abilities of antiproliferation, reversing epithelial to mesenchymal transition (EMT), and suppression of cancer stemness, and it could suppress tumor growth and inhibit the metastasis of tumor cells to livers and lungs with therapeutic safety in BALB/C mice [437].

7. The Important Health Benefits of Lignan (Sesamin)

Lignans are naturally occurring compounds produced and accumulated in different edible and medicinal plants, which can be subdivided bio-synthetically into neolignan and lignans [438,439]. Lignans, as the notable subgroup of phenylpropanoids, are involved in the plant defense responses to numerous biotic and abiotic stresses [440]. Lignans, with different biological activities, such as antitumor, antibacterial, antioxidant, and antiviral activities, are generally distributed in nature and mostly exist in the xylem of plants [441,442]. The level of lignans varies between plant parts of all species [443].
Sesamin, a major lignan derived from sesame seeds, has several benefits and medicinal characteristics [444]. It exerts various pharmacological impacts, such as prevention of hyperlipidemia, hypertension, and carcinogenesis, as well as anticancer and chemopreventive activity in vitro and in vivo [445,446], and antioxidant and anti-inflammatory characteristics [447,448]. Plants reported to contain sesamin are Paulownia tomentosa Staud., Phyllarthron comorense, Justicia simplex, Hyptis tomentosa, Anacyclus pyrethrum, Artemisia absinthium, Artemisia gorgonum, Chrysanthemum cinerariaefolium, C. frutescens, C. indicum, Diotis maritima, Eupatorium ageratina, E. ritonia, E. fleischmannia, Otanthus maritimus, Aptosimum spinescens, Gmelina arborea Roxb., Acanthopanax senticosus, A. sessiliflorum, Eleutherococcus divaricatus, Asarum sieboldii, Aristolochia cymbifera, Alnus glutinosa, Salicomia europaea, Austrocedrus chilensis, Evodia micrococca, Fagara xanthoxyloides, Fagara tessmannii, Fagara heitzii, Micromelum minutum, Melicope glabra, Spiranthera odoratissima, Flindersia pubescens, Zanthoxylum naranjillo, Zanthoxylum tingoassuiba, Zanthoxylum piperitum, Zanthoxylum nitidum, Zanthoxylum flavum, Zanthoxylum alatum Roxb., Zanthoxylum bungeanum, Ginkgo biloba, Machilus glaucescens, Ocotea usambarensis, Aiouea trinervis Meisn., Talauma hodgsonii, Magnolia spp., Picea abies, Macropiper excelsum, Piper sarmentosum, Sesamum indicum, S. radiatum, S. mulayanum, S. malabaricum, S. alatum, S. angustifolium, S. angolense, S. calycinum, Anemopsis californica, Quercus frainetto Ten., Vernicia fordii, Jatropha curcas, Larrea tridentata, Morinda citrifolia, Glossostemon bruguieri, Ligustrum japonicum, and Triclisia sacleuxii [449]. Sesamin could boost the proliferation and adhesion of intestinal probiotics, leading to modulating gut microbiota, which provided the basis for sesamin as a food-borne functional parameter for improving intestinal health [450]. Sesamin suppressed breast cancer proliferation, and it downregulated programmed death ligand 1 (PD-L1) expression, which is mediated by NF-KB and AKT [451]. Sesamin increased osteoblast differentiation by the increase of type I collagen (COL1A1) and alkaline phosphatase (ALP) gene expression as well as ALP activity [452]. Sesamin ameliorated lead-induced neuroinflammation in rats, and decreased accumulation of lead in blood and neuronal tissues of rats [453]. It ameliorated polymorphonuclear neutrophils infiltration and exudate volume [454]. Majdalawieh et al. [455] reported that sesamin can potentially be utilized as an effectual adjuvant therapeutic agent in ameliorating tumor development and progression, and it could be utilized in the prevention and treatment of different types of cancer. It has been reported that sesamin promoted diabetes-induced neuroinflammation in rats, exhibited neurotrophic supportive action in diabetic rats, and prevented neuronal loss in diabetic rats [456]. Sesamin has a chondroprotective effect through inhibition of proteoglycans (PGs) degradation induced by IL-1beta and inhibition of collagen degradation [457].

8. The Health Benefits of Condensed Tannins or Proanthocyanidins (Procyanidin B1)

Proanthocyanidins, also known as condensed tannins [458,459], belong to the oldest of plant secondary metabolites, and these constituents are widespread in woody plants, but are also discovered in certain forages, as well as fruits, seeds, nuts, and bark [460,461]. Yu et al. [462] reported that proanthocyanidins were prevalent in lotus seed coats. They can be categorized into three groups according to their component units and the linkages between them: procyanidins, prodelphinidins, and propelargonidins [463]. The biological activity of plant proanthocyanidins is associated with their chemical concentration and structure [464]. Proanthocyanidins from Pinus thunbergii mainly included catechin/epicatechin, and they showed significant antioxidant capacity [465]. Proanthocyanidins in tea, black currant, grapes, bilberry, pine bark, cranberry, and peanut skin may lead to a decrease in the oxidative stress (ROS), induce lower iNOS and COX-2 over-expression, then lower inflammation, and, lastly, show activities against diabetes, asthma, neuropathologies, cardiovascular ailments, obesity, and cancer [466]. The precursors of proanthocyanidins are produced by the phenyl propanoid pathway in the cytosol and are converted to the vacuole, where they polymerize to create proanthocyanidins [467]. They have various bioactivities, such as anticancer, antibacterial, and antioxidant [468]. Proanthocyanidins stimulate antioxidant capacity and increase resistance against oxidative stress-induced senescence in fruits after harvest [469].
Procyanidins are associated with the class of natural products known as proanthocyanidins or condensed polyphenols [470]. They have been reported to reveal broad advantages to human health and are applied in the prevention of cancers, diabetes, cardiovascular diseases, etc. [471]. They are structurally diverse constituents and can be divided into monomeric, oligomeric, or polymeric variants associated with degree of polymerization, which plays a role in manifesting various impacts that are associated with human health [471]. The anti-digestion and antioxidant impacts of grape seed procyanidins have been proven [472]. Procyanidin B1 is also a promising liver cancer antitumor drug [473] (Na et al., 2020). Procyanidins increase the glycometabolism and decrease the secretion of inflammatory factors of postpartum mice with gestational diabetes mellitus (GDM) [474].

9. The Health Benefits of Curcuminoids (Curcumin, Demethoxycurcumin, Bisdemethoxycurcumin)

9.1. Curcuminoids

Curcuminoids are a group of polyphenol coloring constituents that exist in the plant species Curcuma, such as Curcuma longa, C. Wenyujin, C. zedoaria, etc. [475,476]. They are synthesized in turmeric from cinnamic acid precursors obtained via the phenylpropanoid biosynthetic pathway, and there are three different precursors, namely curcuminoids biosynthesis-cinnamic acid, ferulic acid, and coumaric acid [477]. Ramirez-Ahumada et al. [478] reported that curcuminoid synthase activity in turmeric crude protein extracts converts feruloyl-CoA into curcumin. Curcumins are the commercially available component in curcuminoids, as the principle constituents, and the other two, demethoxycurcumin and bisdemethoxycurcumin, as minor components [479,480]. Curcumin and demethoxycurcumin are distinctive because of the phenylmethoxy group [481]. Curcuminoids share important pharmacological characteristics possessed by turmeric, a distinguished curry spice, considered as an important factor in Alzheimer’s disease [482]. It has been reported that curcuminoids of turmeric can be considered as a modern medicine for the treatment of knee osteoarthritis [483] as well as a potential anticancer agent [484]. Zhou et al. [485] also reported that turmeric rhizomes exhibit versatile biological activities such as a significant anticancer property. Three curcuminoids, namely curcumin, demethoxycurcumin, and bisdemethoxycurcumin, in turmeric were found and were shown to contain significant synergistic anticancer activities [486]. Curcuminoids rescued neurotoxin-induced inflammatory gene expression and rescued neurotoxin-induced apoptotic gene expression, and individual curcuminoids showed significant function useful for Alzheimer’s disease [482].

9.2. Curcumin

Curcumin (bis-α,β-unsaturated β-diketone), also known as diferuloylmethane, is a hydrophobic polyphenol obtained from the rhizome of the perennial herb genus Curcuma, which belongs to the ginger family (Zingiberaceae) and consists of species such as Curcuma longa, Curcuma amada, Curcuma aromatic, Curcuma zedoaria, and Curcuma raktakanta [487,488]. Curcumins contain different medicinal values such as antioxidant, anti-pulmonary fibrosis, anti-inflammation, antiviral, and chronic obstructive pulmonary disease impacts, and attractively docked with multi-target molecular proteins related to diabetes [489,490,491,492,493,494]. Curcumin is insoluble in water and easily efficient in organic solvents [495]; the active functional groups of curcumin can be oxidized by electron transfer and hydrogen abstraction [496], and curcumin is more durable in acidic to neutral conditions than in alkaline circumstances [495,496,497]. Curcumin, as an enzyme inhibitor, has proper structural characteristics including a flexible backbone, hydrophobic nature, and different available hydrogen bond (H-bond) donors and acceptors [498]. Curcumin is stable to heat but is light-sensitive and produces singlet oxygen and other reactive oxygen species (ROS) when exposed to the sun, which is also a photodynamic and photobiological property of curcumin [499]. Curcumin decreases inflammation by inhibiting lipopolysaccharide-induced nuclear factor-KB (NF-KB) p65 translocation and mitogen-activated protein kinase activation in dendritic cells [500]. Curcumin decreases morphine dependence in rats through an inhibitory influence on neuroinflammation and a decline in the expression of μ-opioid receptors in the prefrontal cortex [501]. Curcumin influences synaptic plasticity genes (Arc and Fmr1) to decrease amnesia [502]. Xie et al. [503] reported that curcumin together with photodynamic therapy have been confirmed as effective in many kinds of cancer cells in vitro and animal models. It has been extensively applied in cancer treatment because of its ability to trigger cell death and suppress metastasis [504]. Mahjoob and Stochaj [505] reported that curcumin improves aging-related cellular and organ dysfunctions. Curcumin can be a promising antifatigue substitute for improving exercise performance [506]. Its derivatives have anti-inflammatory actions for drug repurposing in traumatic brain injury (TBI), but their molecular targets are not clear [507].

9.3. Demethoxycurcumin

Demethoxycurcumin is one of the principle active compounds of curcuminoids discovered in turmeric powder, which is used as a spice in Asian cooking and traditional medicine [508]. Recent studies reveal that demethoxycurcumin has various biological activities including antioxidant, anti-inflammation, and anticancer activities [509,510,511]. Lin et al. [512] reported that demethoxycurcumin is the most active constituent against various kinds of breast cancer cell lines and induces apoptosis and autophagy. Demethoxycurcumin, a natural derivative of curcumin, revealed stronger inhibitory activity on nitric oxide and tumor necrosis factor-α production in comparison with curcumin in lipopolysaccharide-activated rat primary microglia [513]. Demethoxycurcumin remitted the inflammation of nucleus pulposus cells without overt cytotoxic impacts [514].

9.4. Bisdemethoxycurcumin

Bisdemethoxycurcumin is a demethoxy derivative of curcumin and is much more stable than curcumin in physiological media [514,515,516]. It can scavenge free radicals and control cellular redox balance because of its antioxidant property [517,518], and it has potential anti-allergic effects [519]. Mahattanadul et al. [520] reported that bisdemethoxycurcumin’s antiulcer impacts might be because of its characteristics of decreasing gastric acid secretion and increasing the mucosal defensive mechanism via suppression of inducible nitric oxide synthase (iNOS)-mediated inflammation. Bisdemethoxycurcumin inhibits human pancreatic α-amylase (HPA) [521].

10. Conclusions

Phenolic compounds are one of the most important types of compounds with an important role in growth and reproduction, providing protection against pathogens and predators, and they could be the main determinant of antioxidant potential of foods. Phenolics are a heterogeneous collection of compounds generated as secondary metabolites in plants. Phenolic compounds are aromatic or aliphatic compounds with at least one aromatic ring to which one or more OH groups are connected. They are subdivided into different groups depending on the number of phenolic rings that they possess and the structural elements joined to them. They are naturally occurring compounds present in several foods such as cereals, fruits, vegetables, and beverages. Polyphenols can also be found in dried legumes and chocolate. The distribution of phenolic compounds in plant tissues and cells change considerably according to the type of chemical compound. They also contribute towards the color and sensory characteristics of fruits and vegetables. Different classes of phenolic compounds in plants are simple phenolics, benzoquinones, hydroxybenzoic acids, acetophenones, phenylacetic acids, hydroxycinnamic acids, phenylpropanoids, naphthoquinones, xanthones, stilbenes, anthraquinones, flavonoids, isoflavonoids, lignans, neolignans, biflavonoids, lignins, and condensed tannins. Hydroxybenzoic acids are gallic acid and Protocatechuic acid. Hydroxycinnamic acids are p-coumaric acid, caffeic acid, ferulic acid, sinapic acid, and other components such as coumarins (umbelliferone, esculetin, scopoletin, resveratrol, piceatannol, pterostilbene), curcuminoids (curcumin, demethoxycurcumin, bisdemethoxycurcumin), condensed tannins or proanthocyanidins (procyanidin B1), and lignan (sesamin). From a human physiological viewpoint, phenolic compounds are important in defense responses such as antioxidant, anti-aging, antiproliferative, and anti-inflammatory. High phenolic activity in many species could prove to be beneficial towards human health if included as part of food designs for a healthy diet.
Flavonoids are the largest group of natural phenolic compounds, and, based on the differences in the pyran ring, flavonoids can be divided into flavones, isoflavones, flavanonols, flavonols, flavanones, flavan-3-ols, and anthocyanidins. They can be subdivided into different subgroups on the basis of the carbon of the C ring on which the B ring is attached and the degree of unsaturation and oxidation of the C ring. Flavonoids in which the B ring is linked in position 3 of the C ring are called isoflavones. Those in which the B ring is linked in position 4 are called neoflavonoids, while those in which the B ring is linked in position 2 can be further subdivided into several subgroups on the basis of the structural characteristics of the C ring. The most prominent health benefits of phenolic compounds are antioxidant activity, anti-inflammatory properties, antifungal activity, antimicrobial activity, antibacterial properties, anti-coronavirus activities, neuroprotective potential, appropriate for skin health, suitable for wound healing, and anticancer activities. Flavonoids, a group of natural substances with variable phenolic structure, are found in vegetables, fruits, grains, bark, stems, roots, flowers, wine, and tea. Flavonoids are considered as an important constituent in different pharmaceutical, medicinal, nutraceutical, and cosmetic applications. They belong to a class of low-molecular-weight phenolic compounds that are extensively distributed in the plant kingdom. Future research is needed to determine the pharmaceutical benefits of phenolic and flavonoid compounds of medicinal plants, especially traditional Chinese medicinal plants, and to gain a better understanding of these chemical compounds in medicinal plants and herbs. It is also important to increase analytic techniques to allow the collection of more data on excretion and absorption.

Author Contributions

W.S., writing—original draft preparation; M.H.S., writing—original draft preparation and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Natural Science Foundation of Beijing, China (Grant No.M21026). This research was also supported by the National Key R&D Program of China (Research grant 2019YFA0904700).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Marmitt, D.; Shahrajabian, M.H. Plant species used in Brazil and Asia regions with toxic properties. Phytother. Res. 2021, 35, 4703–4726. [Google Scholar] [CrossRef] [PubMed]
  2. Shahrajabian, M.H.; Sun, W.; Cheng, Q. Clinical aspects and health benefits of ginger (Zingiber officinale) in both traditional Chinese medicine and modern industry. Acta Agric. Scand. B Soil Plant Sci. 2019, 69, 546–556. [Google Scholar] [CrossRef]
  3. Shahrajabian, M.H.; Sun, W.; Shen, H.; Cheng, Q. Chinese herbal medicine for SARS and SARS-CoV-2 treatment and prevention, encouraging using herbal medicine for COVID-19 outbreak. Acta Agric. Scand. B Soil Plant Sci. 2020, 70, 437–443. [Google Scholar] [CrossRef]
  4. Shahrajabian, M.H.; Sun, W. Using sumac (Rhus coriaria L.), as a miraculous spice with outstanding pharmacological activities. Not. Sci. Biol. 2022, 14, 11118. [Google Scholar] [CrossRef]
  5. Shahrajabian, M.H.; Sun, W. Medicinal plants, economical and natural agents with antioxidant activity. Curr. Nutr. Food. Sci. 2022, 18, 1. [Google Scholar] [CrossRef]
  6. Shahrajabian, M.H.; Sun, W.; Cheng, Q. Product of natural evolution (SARS, MERS, and SARS-CoV-2); deadly diseases, from SARS to SARS-CoV-2. Hum. Vaccines Immunother. 2020, 17, 62–83. [Google Scholar] [CrossRef] [PubMed]
  7. Shahrajabian, M.H.; Sun, W.; Soleymani, A.; Cheng, Q. Traditional herbal medicines to overcome stress, anxiety and improve mental health in outbreaks of human coronaviruses. Phytother. Res. 2020, 35, 1237–1247. [Google Scholar] [CrossRef] [PubMed]
  8. Marmitt, D.; Shahrajabian, M.H.; Goettert, M.I.; Rempel, C. Clinical trials with plants in diabetes mellitus therapy: A systematic review. Expert Rev. Clin. Pharmacol. 2021, 14, 735–747. [Google Scholar] [CrossRef]
  9. Sun, W.; Shahrajabian, M.H.; Cheng, Q. Barberry (Berberis vulgaris), a medicinal fruit and food with traditional and modern pharmaceutical uses. Isr. J. Plant Sci. 2021, 68, 61–71. [Google Scholar] [CrossRef]
  10. Sun, W.; Shahrajabian, M.H.; Cheng, Q. Fenugreek cultivation with emphasis on historical aspects and its uses in traditional medicine and modern pharmaceutical sciences. Mini Rev. Med. Chem. 2021, 21, 724–730. [Google Scholar] [CrossRef] [PubMed]
  11. Sun, W.; Shahrajabian, M.H.; Cheng, Q. Anise (Pimpinella anisum L.), a dominant spice and traditional medicinal herb for both food and medicinal purposes. Cogent Biol. 2019, 5, 1673688. [Google Scholar] [CrossRef]
  12. Barragan-Zarate, G.S.; Lagunez-Rivera, L.; Solano, R.; Carranza-Alvarez, C.; Hernandez-Benavides, D.M.; Vilarem, G. Validation of the traditional medicinal use of a Mexican endemic orchid (Prosthechea karwinskii) through UPLC-ESI-qTOF-MS/MS characterization of its bioactive compounds. Heliyon 2022, 8, e09867. [Google Scholar] [CrossRef]
  13. Shahrajabian, M.H. Medicinal herbs with anti-inflammatory activities for natural and organic healing. Curr. Org. Chem. 2021, 25, 2885–2901. [Google Scholar] [CrossRef]
  14. Shahrajabian, M.H.; Sun, W.; Cheng, Q. Exploring Artemisia annua L., artemisinin and its derivatives, from traditional Chinese wonder medicinal science. Not. Bot. Horti Agrobot. 2020, 48, 1719–1741. [Google Scholar] [CrossRef]
  15. Shahrajabian, M.H.; Sun, W.; Cheng, Q. Chemical components and pharmacological benefits of Basil (Ocimum basilicum): A review. Int. J. Food Prop. 2020, 23, 1961–1970. [Google Scholar] [CrossRef]
  16. Shahrajabian, M.H.; Sun, W.; Cheng, Q. Traditional herbal medicine for the prevention and treatment of cold and flu in the autumn of 2020, overlapped with COVID-19. Nat. Prod. Commun. 2020, 15, 1431. [Google Scholar] [CrossRef]
  17. Shahrajabian, M.H.; Sun, W. Sustainable approaches to boost yield and chemical constituents of aromatic and medicinal plants by application of biostimulants. Recent Adv. Food Nutr. Agric. 2022, 13, 72–92. [Google Scholar] [CrossRef]
  18. Shahrajabian, M.H.; Sun, W. Importance of thymoquinone, sulforaphane, phloretin, and epigallocatechin and their health benefits. Lett. Drug Des. Discov. 2022, 19. ahead of print. [Google Scholar] [CrossRef]
  19. Shahrajabian, M.H.; Sun, W. Survey on medicinal plants and herbs in traditional Iranian medicine with antioxidant, antiviral and antimicrobial, and antiinflammation properties. Lett. Drug Des. Discov. 2022, 19. ahead of print. [Google Scholar] [CrossRef]
  20. Supritha, P.; Radha, K.V. Estimation of phenolic compounds present in the plant extracts using high pressure liquid chromatography, antioxidant properties, and its antibacterial activity. Indian J. Pharm. Educ. Res. 2018, 52, 321–326. [Google Scholar] [CrossRef]
  21. Amessis-Ouchemoukh, N.; Madani, K.; Fale, P.L.V.; Serralheiro, M.L.; Araujo, M.E.M. Antioxidant capacity and phenolic contents of some Mediterranean medicinal plants and their potential role in the inhibition of cyclooxygenase-1 and acetylcholinesterase activities. Ind. Crop. Prod. 2014, 53, 6–15. [Google Scholar] [CrossRef]
  22. El-Haci, I.A.; Bekkara, F.A.; Mazari, W.; Gherib, M. Phenolic content and antioxidant activity of some organic extracts of endemic medicinal plant Anabasis aretioides Coss. & Moq. From Algerian Sahara. Pharmacogn. J. 2013, 5, 108–112. [Google Scholar] [CrossRef]
  23. Subedi, L.; Timalsena, S.; Duwadi, P.; Thapa, R.; Paudel, A.; Parajuli, K. Antioxidant activity and phenol and flavonoid contents of eight medicinal plants from Western Nepal. J. Tradit. Chin. Med. 2014, 34, 584–590. [Google Scholar] [CrossRef] [Green Version]
  24. Skotti, E.; Anastasaki, E.; Kanellou, G.; Polissiou, M.; Tarantilis, P.A. Total phenolic content, antioxidant activity and toxicity of aqueous extracts from selected Greek medicinal and aromatic plants. Ind. Crop. Prod. 2014, 53, 46–54. [Google Scholar] [CrossRef] [Green Version]
  25. Jimenez, N.; Carrillo-Hormaza, L.; Pujol, A.; Alzate, F.; Osorio, E.; Lara-Guzman, O. Antioxidant capacity and phenolic content of commonly used anti-inflammatory medicinal plants in Colombia. Ind. Crop. Prod. 2015, 70, 272–279. [Google Scholar] [CrossRef]
  26. Cisneros, H.S.; Bertiller, M.B.; Furlong, J.J.P.; Carrera, A.L. Similar structural complexity of phenols in plant morphotypes with contrasting soluble phenol concentration and richness in arid rangelands of Patagonia. Flora 2022, 295, 152134. [Google Scholar] [CrossRef]
  27. Tirado-Kulieva, V.A.; Hernandez-Martinez, E.; Choque-Rivera, T.J. Phenolic compounds versus SARS-CoV-2: An update on the main findings against COVID-19. Heliyon 2022, 8, e10702. [Google Scholar] [CrossRef] [PubMed]
  28. Sun, W.; Shahrajabian, M.H.; Cheng, Q. Natural dietary and medicinal plants with anti-obesity therapeutics activities for treatment and prevention of obesity during lock down and in post-COVID-19 era. Appl. Sci. 2021, 11, 7889. [Google Scholar] [CrossRef]
  29. Sun, W.; Shahrajabian, M.H.; Lin, M. Research progress of fermented functional foods and protein factory-microbial fermentation technology. Fermentation 2022, 8, 688. [Google Scholar] [CrossRef]
  30. Macharia, J.M.; Mwangi, R.W.; Rozmann, N.; Zsolt, K.; Varjas, T.; Uchechukwu, P.O.; Wagara, I.N.; Raposa, B.L. Medicinal plants with anti-colorectal cancer bioactive compounds: Potential game-changers in colorectal cancer management. Biomed. Pharmacother. 2022, 153, 113383. [Google Scholar] [CrossRef]
  31. Jaiswal, N.; Kumar, A. HPLC in the discovery of plant phenolics as antifungal molecules against Candida infection related biofilms. Microchem J. 2022, 179, 107572. [Google Scholar] [CrossRef]
  32. Diaz, P.; Jeong, S.C.; Lee, S.; Khoo, C.; Koyyalamudi, S.R. Antioxidant and anti-inflammatory activities of selected medicinal plants and fungi containing phenolic and flavonoids compounds. Chin. Med. 2012, 7, 26. [Google Scholar] [CrossRef] [Green Version]
  33. Tukun, A.B.; Shaheen, N.; Banu, C.P.; Mohiduzzaman, M.D.; Islam, S.; Begum, M. Antioxidant capacity and total phenolic contents in hydrophilic extracts of selected Bangladeshi medicinal plants. Asian Pac. J. Trop. Med. 2014, 7 (Suppl. 1), S568–S573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Manguro, L.O.A.; Lemmen, P. Phenolics of Moringa oleifera leaves. Nat. Prod. Res. 2007, 21, 56–68. [Google Scholar] [CrossRef]
  35. Sodaeizadeh, H.; Rafieiolhossaini, M.; Havlik, J.; van Damme, P. Allelopathic activity of different plant parts of Peganum harmala L. and identification of their growth inhibitors substances. Plant Growth Regul. 2009, 59, 227–236. [Google Scholar] [CrossRef]
  36. Shah, B.; Cai, Y.Z.; Sun, M.; Corke, H. Antioxidant capacity of 26 spice extracts and characterization of their phenolic constituents. J. Agric. Food Chem. 2005, 53, 7749–7759. [Google Scholar] [CrossRef] [Green Version]
  37. Koksal, E.; Bursal, E.; Gulcin, I.; Korkmaz, M.; Caglayan, C.; Goren, A.C.; Alwasel, S.H. Antioxidant activity and polyphenol content of Turkish thyme (Thymus vulgaris) monitored by liquid chromatography and tandem mass spectrometry. Int. J. Food Prop. 2017, 20, 514–525. [Google Scholar] [CrossRef] [Green Version]
  38. Vastano, B.C.; Chen, Y.; Zhu, N.Q.; Ho, C.T.; Zhou, Z.Y.; Rosen, R.T. Isolation and identification of stilbenes in two varieties of Polygonum cuspidatum. J. Agric. Food 2000, 48, 253–256. [Google Scholar] [CrossRef]
  39. Zhao, X.H.; Han, F.; Li, Y.L.; Yue, H.L. Preparative isolation and purification of three stilbene glycosides from the Tibetan medicinal plant Rheum tanguticum Maxim. Ex Balf. by high-speed counter-current chromatography. Phytochem. Anal. 2013, 24, 171–175. [Google Scholar] [CrossRef]
  40. Ahmad, A.; Misra, L.N. Isolation of herniarin and other constituents from Matricaria chamomilla flowers. Int. J. Pharmacogn. 1997, 35, 121–125. [Google Scholar] [CrossRef]
  41. Morikawa, T.; Luo, F.L.; Manse, Y.; Sugita, H.; Saeki, S.; Chaipech, S.; Pongiriyadacha, Y.; Muraoka, O.; Ninomiya, K. Geranylated coumarins from Thai medicinal plant Mammea siamensis with testosterone 5 alpha-reductase inhibitory activity. Front. Chem. 2020, 8, 199. [Google Scholar] [CrossRef] [Green Version]
  42. Thuong, P.T.; Hung, T.M.; Ngoc, T.M.; Ha, D.T.; Min, B.S.; Kwak, S.J.; Kang, T.S.; Choi, J.S.; Bae, K. Antioxidant activities of coumarins from Korean medicinal plants and their structure-activity relationships. Phytother. Res. 2010, 24, 101–106. [Google Scholar] [CrossRef] [PubMed]
  43. Singh, M.; Tiwari, N.; Shanker, K.L.; Verma, R.K.; Gupta, A.K.; Gupta, M.M. Two new lignans from Phyllanthus amarus. J. Asian Nat. Prod. Res. 2009, 11, 562–568. [Google Scholar] [CrossRef] [PubMed]
  44. Hu, D.; Yang, Z.Y.; Yao, X.C.; Wang, H.; Han, N.; Liu, Z.H.; Wang, Y.; Yang, J.Y.; Yin, J. Dibenzocyclooctadiene lignans from Schisanra chinensis and their inhibitory activity on No production in lipopolysaccharide-activated microglia cells. Phytochemistry 2014, 104, 72–78. [Google Scholar] [CrossRef] [PubMed]
  45. Baranyai, B.; Backer, C.; Reich, C.; Lindequist, U. The production of 7-methyljuglone, plumbagin, and quercetin in wild and cultivated Drosera rotundifolia and Drosera intermedia. Mires Peat 2016, 18, 1–8. [Google Scholar] [CrossRef]
  46. Tian, K.; Zhang, H.G.; Chen, X.G.; Hu, Z.D. Determination of five anthraquinones in medicinal plants by capillary zone electrophoresis with beta-cyclodextrin addition. J. Chromatogr. A 2006, 1123, 134–137. [Google Scholar] [CrossRef]
  47. Ali, I.; Haque, A.; Saleem, K. Separation and identification of curcuminoids in turmeric powder by HPLC using phenyl coloumn. Anal. Methods 2014, 6, 2526–2536. [Google Scholar] [CrossRef]
  48. Taamalli, A.; Arraez-Roman, D.; Abaza, L.; Iswaldi, I.; Fernandez-Gutierrez, A.; Zarrouk, M.; Segura-Carretero, A. LC-MS-based metabolite profiling of methanolic extracts from the medicinal and aromatic species Mentha pulegium and Origanum majorana. Phytochem Anal. 2015, 26, 320–330. [Google Scholar] [CrossRef]
  49. Rodrigues, F.; Almeida, I.; Sarmento, B.; Amaral, M.H.; Oliveira, M. Study of the isoflavone content of different extracts of Medicago spp. as potential active ingredient. Ind. Crop. Prod. 2014, 57, 110–115. [Google Scholar] [CrossRef]
  50. Stevenson, P.C.; Kite, G.C.; Lewis, G.P.; Forest, F.; Nyirenda, S.P.; Belmain, S.R.; Sileshi, G.W.; Veitch, N.C. Disntinct chemotypes of Tephrosia vogelii and implications for their use in pest control and soil enrichment. Phytochemistry 2012, 78, 135–146. [Google Scholar] [CrossRef]
  51. Russo, M.; Fanali, C.; Tripodo, G.; Dugo, P.; Muleo, R.; Dugo, L.; de Gara, L.; Mondello, L. Analysis of phenolic compounds in different parts of pomegranate (Punica granatum) fruit by HPLC-PDA-ESI/MS and evaluation of their antioxidant activity: Application to different Italian varieties. Anal. Bioanal. Chem. 2018, 410, 3507–3520. [Google Scholar] [CrossRef] [PubMed]
  52. Bravo, L. Polyphenols: Chemistry, dietary sources, metabolism, and nutritional significance. Nutr. Rev. 1998, 56, 317–333. [Google Scholar] [CrossRef] [PubMed]
  53. Balasundram, N.; Sundram, K.; Samman, S. Phenolic compounds in plants and agri-industrial by-products: Antioxidant activity, occurrence, and potential uses. Food Chem. 2006, 99, 191–203. [Google Scholar] [CrossRef]
  54. De la Rosa, L.A.; Moreno-Escamilla, J.O.; Rodrigo-Garcia, J.; Alvarez-Parrilla, E. Chapter 12—Henolic Compounds. In Postharvest Physiology and Biochemistry of Fruits and Vegetables; Yahia, E.M., Ed.; Woodhead Publishing: Sawston, UK, 2019; pp. 253–271. [Google Scholar] [CrossRef]
  55. Soleymani, A.; Shahrajabian, M.H.; Naranjani, L. Yield and yield components of berseem clover cultivars in low nitrogen fertilizer input farming. J. Food Agric. Environ. 2011, 9, 281–283. [Google Scholar]
  56. Soleymani, A.; Shahrajabian, M.H.; Naranjani, L. Effect of planting dates and different levels of nitrogen on seed yield and yield components of nuts sunflower (Helianthus annuus L.). Afr. J. Agric. Res. 2013, 8, 5802–5805. [Google Scholar] [CrossRef]
  57. Paine, C.S.; van Staden, J.; Finnie, J.F. Elicitation of phenolic compounds of medicinal value in plants. S. Afr. J. Bot. 2017, 109, 362. [Google Scholar] [CrossRef]
  58. Salem, M.M.; Davidorf, F.H.; Abdel-Rahman, M.H. In vitro anti-uveal melanoma activity of phenolic compounds from the Egyptian medicinal plant Acacia nilotica. Fitoterapia 2011, 82, 1279–1284. [Google Scholar] [CrossRef]
  59. Carbonell-Capella, J.M.; Buniowska, M.; Barba, F.J.; Esteve, M.J.; Frigola, A. Analytical methods for determining bioavailability and bioaccessibility of bioactive compounds from fruits and vegetables: A review. Compr. Rev. Food Sci. Food Saf. 2014, 13, 155–171. [Google Scholar] [CrossRef]
  60. Rein, M.J.; Renouf, M.; Cruz-Hernandez, C.; Actis-Goretta, L.; Thakkar, S.K.; da Silva Pinto, M. Bioavailability of bioative food compounds: A challenging journey of bioefficacy. Br. J. Clin. Pharmacol. 2013, 75, 588–602. [Google Scholar] [CrossRef] [Green Version]
  61. Albergaria, E.T.; Oliveira, A.F.M.; Albuquerque, U.P. The effect of water deficit stress on the composition of phenolic compounds in medicinal plants. S. Afr. J. Bot. 2020, 131, 12–17. [Google Scholar] [CrossRef]
  62. Djeridane, A.; Yousfi, M.; Nadjemi, B.; Boutassouna, D.; Stocker, P.; Vidal, N. Antioxidant activity of some algerian medicinal plants extracts containing phenolic compounds. Food Chem. 2006, 97, 654–660. [Google Scholar] [CrossRef]
  63. Cai, Y.; Luo, Q.; Sun, M.; Corke, H. Antioxidant activity and phenolic compounds of 112 trasitional Chinese medicinal plants associated with anticancer. Life Sci. 2004, 74, 2157–2184. [Google Scholar] [CrossRef] [PubMed]
  64. Chirinos, R.; Pedreschi, R.; Rogez, H.; Larondelle, Y.; Campos, D. Phenolic compounds contents and antioxidant activity in plants with nutritional and/or medicinal properties from the Peruvian Andean region. Ind. Crop. Prod. 2013, 47, 145–152. [Google Scholar] [CrossRef]
  65. Cai, Y.-Z.; Sun, M.; Xing, J.; Luo, Q.; Corke, H. Structure—Radical scavenging activity relationships of phenolic compounds from traditional Chinese medicinal plants. Life Sci. 2006, 78, 2872–2888. [Google Scholar] [CrossRef] [PubMed]
  66. Barros, L.; Duenas, M.; Carvalho, A.M.; Ferreira, I.C.F.R.; Santos-Buelga, C. Characterization of phenolic compounds in flowers of wild medicinal plants from Northeastern Portugal. Food Chem. Toxicol. 2012, 50, 1576–1582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Kaska, A.; Cicek, M.; Mammadov, R. Biological activities, phenolic constituents and mineral element analysis of two endemic medicinal plants from Turkey: Nepeta italica subsp. cadmea and Teucrium sandrasicum. S. Afr. J. Bot. 2019, 124, 63–70. [Google Scholar] [CrossRef]
  68. Benabderrahim, M.A.; Yahia, Y.; Bettaieb, I.; Elfalleh, W.; Nagaz, K. Antioxidant activity and phenolic profile of a collection of medicinal plants from Tunisian arid and Saharan regions. Ind. Crop. Prod. 2019, 138, 111427. [Google Scholar] [CrossRef]
  69. Ranilla, L.G.; Kwon, Y.-I.; Apostolidis, E.; Shetty, K. Phenolic compounds, antioxidant activity and in vitro inhibitory potential against key enzymes relevant for hyperglycemia and hypertension of commonly used medicinal plants, herbs, and species in Latin America. Bioresour. Technol. 2010, 101, 4676–4689. [Google Scholar] [CrossRef]
  70. Ng, M.H.; Nu/man, A.H. Investigation on the use of deep eutectic solvent with microwave assistance for the extraction of ferulic acid from palm pressed fibre. Curr. Res. Green Sustain. Chem. 2021, 4, 100155. [Google Scholar] [CrossRef]
  71. Chahal, S.; Kaur, H.; Lekhak, M.M.; Shekhawat, M.S.; Goutam, U.; Singh, S.K.; Ochatt, S.J.; Kumar, V. Meta-topolin-mediated regeneration and accumulation of phenolic acids in the critically endangered medicinal plant Crinum malabaricum (Amaryllidaceae): A potent source of galanthamine. S. Afr. J. Bot. 2022, 149, 853–859. [Google Scholar] [CrossRef]
  72. Mssillou, I.; Bakour, M.; Slighoua, M.; Laaroussi, H.; Saghrouchni, H.; Amrati, F.E.-Z.; Lyoussi, B.; Derwich, E. Investigation on wound healing effect of Mediterranean medicinal plants and some related phenolic compounds: A review. J. Ethnopharmacol. 2022, 298, 115663. [Google Scholar] [CrossRef]
  73. Silva, R.F.D.; Carneiro, C.N.; Sousa, C.B.D.C.; Gomez, F.J.V.; Espino, M.; Boiteux, J.; Fernandez, M.D.I.A.; Silva, M.F.; Dias, F.D.S. Sustainable extraction bioactive compounds procedures in medicinal plants based on the principles of green analytical chemistry: A review. Microchem. J. 2022, 175, 107184. [Google Scholar] [CrossRef]
  74. Tarhan, L.; Urek, R.O.; Oner, A.; Nakiboglu, M. Evaluation of phenolic profiles, antioxidant activities, and cytotoxic and apoptotic potentials of Phlomis angustissima and Phlomis fruticosa, medicinal plants from Turkey. Eur. J. Integr. Med. 2022, 55, 102188. [Google Scholar] [CrossRef]
  75. Raghuvanshi, D.; Sharma, K.; Verma, R.; Kumar, D.; Kumar, H.; Khan, A.; Valko, M.; Almoar, S.Y.; Alwasel, S.H.; Nepovimova, E.; et al. Phytochemistry, and pharmacological efficacy of Cordia dichotoma G. Forst. (Lashuda): A therapeutic medicinal plant of Himachal Pradesh. Biomed. Pharmacother. 2022, 153, 113400. [Google Scholar] [CrossRef] [PubMed]
  76. Thitilertdecha, P.; Rowan, M.G.; Guy, R.H. Topical formulation and dermal delivery of active phenolic compounds in the Thai medicinal plant-Clerodendrum petasites S. Moore. Int. J. Pharm. 2015, 478, 39–45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Sprea, R.M.; Caleja, C.; Pinela, J.; Finimundy, T.C.; Calhelha, R.C.; Kostic, M.; Sokovic, M.; Prieto, M.A.; Pereira, E.; Amaral, J.S.; et al. Comparative study on the phenolic composition and in vitro bioactivity of medicinal and aromatic plants from the Lamiaceae family. Food Res. Int. 2022, 161, 111875. [Google Scholar] [CrossRef] [PubMed]
  78. Garcia-Perez, P.; Ayuso, M.; Lozano-Milo, E.; Pereira, C.; Dias, M.I.; Ivanov, M.; Calhelha, R.C.; Sokovic, M.; Ferreira, I.C.F.R.; Barros, L.; et al. Phenolic profiling and in vitro bioactivities of three medicinal Bryophyllum plants. Ind. Crop. Prod. 2021, 162, 113241. [Google Scholar] [CrossRef]
  79. Bouyahya, A.; El Omari, N.; Elmenyiy, N.; Guaouguaou, F.-E.; Balahbib, A.; Belmehdi, O.; Salhi, N.; Imtara, H.; Mrabti, H.N.; El-Shazly, M.; et al. Moroccan antidiabetic medicinal plants: Ethnobotanical studies, phytochemical bioactive compounds, preclinical investigations, toxicological validations and clinical evidences; challenges, guidance, and perspectives for future management of diabetes worldwide. Trends Food Sci. Technol. 2021, 115, 147–254. [Google Scholar] [CrossRef]
  80. Shatri, A.M.N.; Mumbengegwi, D.R. Ethonmedicinal use and phytochemical analysis of medicinal plants used to treat gastrointestinal conditions by Awambo people in Iikokola village, Namibia. Sci. Afr. 2022, 18, e01428. [Google Scholar] [CrossRef]
  81. Pesaraklu, A.; Radjabian, T.; Salami, S.A. Methyl jasmonate and Ag+ effective elicitors for enhancement of phenolic acids contents in Salvia officinalis and Salvia verticillata, as two traditional medicinal plants. S. Afr. J. Bot. 2021, 141, 105–115. [Google Scholar] [CrossRef]
  82. Tavakoli, M.; Esfahani, M.T.; Soltani, S.; Karamian, R.; Aliarabi, H. Effects of ecological factors on phenolic compounds in Salvia multicaulis Vahl (Lamiaceae). Biochem. Syst. Ecol. 2022, 104, 104484. [Google Scholar] [CrossRef]
  83. Milevskaya, V.V.; Prasad, S.; Temerdashev, Z.A. Extraction and chromatographic determination of phenolic compounds from medicinal herbs in the Lamiaceae and Hypericaceae families: A review. Microchem. J. 2019, 145, 1036–1049. [Google Scholar] [CrossRef]
  84. Fawole, O.A.; Opara, U.L. Stability of total phenolic concentration and antioxidant capacity of extracts from pomegranate co-products subjected to in vitro digestion. BMC Complement. Altern. Med. 2016, 16, 358. [Google Scholar] [CrossRef] [Green Version]
  85. Guerrero, R.F.; Aliano-Gonzalez, M.J.; Puertas, B.; Richard, T.; Cantos-Villar, E. Comparative analysis of stilbene concentration in grapevine shoots of thirteen Vitis during a three-year study. Ind. Crop. Prod. 2020, 156, 112852. [Google Scholar] [CrossRef]
  86. Maphetu, N.; Unuofin, J.O.; Masuku, N.P.; Olisah, C.; Lebelo, S.L. Medicinal uses, pharmacological activities, phytochemistry, and the molecular mechanisms of Punica granatum L. (pomegranate) plant extracts: A review. Biomed. Pharmacother. 2022, 153, 113256. [Google Scholar] [CrossRef]
  87. Topalovic, A.; Knezevic, M.; Ivanovic, L.; Gacnik, S.; Mikulic-Petkovsek, M. Phytochemical screening of wild pomegranate (Punica granatum L.) juices from the market. J. Food Compos. Anal. 2021, 100, 103933. [Google Scholar] [CrossRef]
  88. Shamsudin, K.J.; Phan, C.-S.; Kulip, J.; Hatai, K.; Vairappan, C.S.; Kamada, T. Leucoxenols A and B, two new phenolics from Bornean medicinal plant Syzygium leucoxylon. J. Asian Nat. Prod. Res. 2019, 21, 435–441. [Google Scholar] [CrossRef]
  89. Chaparro-Hernandez, I.; Rodriguez-Ramirez, J.; Barriada-Bernal, L.G.; Mendez-Lagunas, L. Tree ferns (Cyatheaceae) as a source of phenolic compounds—A review. J. Herb. Med. 2022, 35, 100587. [Google Scholar] [CrossRef]
  90. Gautam, S.; Chimni, S.S.; Arora, S.; Sohal, S.K. Toxic effects of purified phenolic compounds from Acacia nilotica against common cutworm. Toxicon 2021, 203, 22–29. [Google Scholar] [CrossRef] [PubMed]
  91. Bellumori, M.; Zonfrillo, B.; Maggini, V.; Bogani, P.; Gallo, E.; Firenzuoli, F.; Mulinacci, N.; Innocenti, M. Acmella oleracea (L.) R.K. Jansen: Alkylamides and phenolic compounds in aerial parts and roots of in vitro seedlings. J. Pharm. Biomed. Anal. 2022, 220, 114991. [Google Scholar] [CrossRef] [PubMed]
  92. Parkes, R.; McGee, D.; McDonnell, A.; Gillespie, E.; Touzet, N. Rapid screening of phenolic compounds in extracts of photosynthetic organisms separated using C18 monolithic column based HPLC-UV method. J. Chromatogr. B 2022, 1213, 123521. [Google Scholar] [CrossRef]
  93. Tlais, A.Z.A.; Rantsiou, K.; Filannino, P.; Cocolin, L.S.; Cavoski, I.; Gobbetti, M.; Cagno, R.D. Ecological linkages between biotechnologically relevant autochthonous microorganisms and phenolic compounds in sugar apple fruit (Annona squamosa L.). Int. J. Food Microbiol. 2022, 387, 110057. [Google Scholar] [CrossRef]
  94. Sytar, O.; Hemmerich, I.; Zivcak, M.; Rauh, C.; Brestic, M. Comparative analysis of bioactive phenolic compounds composition from 26 medicinal plants. Saudi J. Biol. Sci. 2018, 25, 631–641. [Google Scholar] [CrossRef] [Green Version]
  95. Kasmi, S.; Hamdi, A.; Atmani-Kilani, D.; Debbache-Benaida, N.; Jaramillo-Carmona, S.; Rodriguez-Arcos, R.; Jimenez-Araujo, A.; Ayouni, K.; Atmani, D.; Guillen-Bejarano, R. Characterization of phenolic compounds isolated from the Fraxinus angustifolia plant and several associated bioactivities. J. Herb. Med. 2021, 29, 100485. [Google Scholar] [CrossRef]
  96. Hossain, M.A.; Shah, M.D.; Gnanaraj, C.; Iqbal, M. In vitro total phenolics, flavonoids contents and antioxidant activity of essential oil, various organic extracts from the leaves of tropical medicinal plant Tetrastigma from Sabah. Asian Pac. J. Trop. Med. 2011, 4, 717–721. [Google Scholar] [CrossRef] [Green Version]
  97. Aremu, A.O.; Ndhlala, A.R.; Fawole, O.A.; Light, M.E.; Finnie, J.F.; van Staden, J. In vitro pharmacological evaluation and phenolic content of ten South African medicinal plants used as anthelmintics. S. Afr. J. Bot. 2010, 76, 558–566. [Google Scholar] [CrossRef] [Green Version]
  98. Golkar, P.; Fotoohi, A. Preliminar phytochemical screening of the phenolic compounds and antioxidant activity of six Plantago species from Iran. J. Plant Process Funct. 2021, 10, 1–10. [Google Scholar] [CrossRef]
  99. Tauchen, J.; Bortil, L.; Huml, L.; Miksatkova, P.; Doskocil, I.; Marsik, P.; Villegas, P.P.P.; Flores, Y.B.; van Damme, P.V.; Lojka, B.; et al. Phenolic composition, antioxidant and anti-proliferative activities of edible and medicinal plants from the Peruvian Amazon. Rev. Bras. Farmacogn. 2016, 26, 728–737. [Google Scholar] [CrossRef]
  100. Stalikas, C.D. Extraction, separation, and detection methods for phenolic acids and flavonoids. J. Sep. Sci. 2007, 30, 3268–3295. [Google Scholar] [CrossRef] [PubMed]
  101. Khalafallah, A.K.; Suleiman, S.A.; Yousef, A.H.; El-Kaniz, N.A.A.; Mohamed, A. Prenylated flavonoids from Tephrosia apollinea. Chin. Chem. Lett. 2009, 20, 1465–1468. [Google Scholar] [CrossRef]
  102. Rebey, I.B.; Bourgou, S.; Debez, I.B.; Karoui, I.J.; Sellami, I.H.; Msaada, K.; Limam, F.; Marzouk, B. Effects of extraction solvents and provenances on phenolic contents and antioxidant activities of cumin (Cuminmum cyminum L.) seeds. Food Bioprocess Technol. 2012, 5, 2827–2836. [Google Scholar] [CrossRef]
  103. Rayyan, S.; Fossen, T.; Andersen, Q.M. Flavone C-Glycosides from seeds of fenugreek, Trigonella foenum-graecum L. J. Agric. Food Chem. 2010, 58, 7211–7217. [Google Scholar] [CrossRef]
  104. Dona, A.C.; Kyriakides, M.; Scott, F.; Shephard, E.A.; Varshavi, D.; Veselkov, K.; Everett, J.R. A guide to the identification of metabolites in NMR-based metabonomics/metabolomics experiments. Comput. Struct. Biotechnol. J. 2016, 14, 135–153. [Google Scholar] [CrossRef] [Green Version]
  105. Arceusz, A.; Wesolowski, M.; Konieczynski, P. Methods for extraction and determination of phenolic acids in medicinal plants: A review. Nat. Prod. Commun. 2013, 8, 1821–1829. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Castillo-Lopez, R.I.; Leon-Felix, J.; Angulo-Escalante, M.A.; Gutierrez-Dorado, R.; Muy-Rangel, M.D.; Heredia, J.B. Nutritional and phenolic characterization of Moringa oleifera leaves grown in Sinaloa, Mexico. Pak. J. Bot. 2017, 49, 161–168. [Google Scholar] [CrossRef] [Green Version]
  107. Xu, C.-C.; Wang, B.; Pu, Y.-Q.; Tao, J.-S.; Zhang, T. Advances in extraction and analysis of phenolic compounds from plant materials. Chin. J. Nat. Med. 2017, 15, 721–731. [Google Scholar] [CrossRef]
  108. Koleva, V.; Simeonov, E. Solid liquid extraction of phenolic and flavonoid compounds from Cotinus coggygria and concentration by nanofiltration. Chem. Biochem. Eng. Q. 2014, 28, 545–551. [Google Scholar] [CrossRef]
  109. Ameer, K.; Shahbaz, H.M.; Kwon, J.H. Green extraction methods for polyphenols from plant matrices and their byproducts: A review. Compr. Rev. Food Sci. Food Saf. 2017, 16, 295–315. [Google Scholar] [CrossRef] [Green Version]
  110. Pimentel-Moral, S.; Borras-Linares, I.; Lozano-Sanchez, J.; Arraez-Roman, D.; Martinez-Ferez, A.; Segura-Carretero, A. Supercritical CO2 extraction of bioactive compounds from Hibiscus sabdariffa. J. Supercrit. Fluids 2019, 147, 213–221. [Google Scholar] [CrossRef]
  111. Dahmoune, F.; Nayak, B.; Moussi, K.; Remini, H.; Madani, K. Optimization of microwave-assisted extraction of polyphenols from Myrtus communis L. leaves. Food Chem. 2015, 166, 585–595. [Google Scholar] [CrossRef] [PubMed]
  112. Fernandez-Ponce, M.T.; Parjikolaei, B.R.; Lari, H.N.; Casas, L.; Mantell, C.; de la Ossa, E.J.M. Pilot-plant scale extraction of phenolic compounds from mango leaves using different green techniques: Kinetic and scale up study. Chem. Eng. J. 2016, 299, 420–430. [Google Scholar] [CrossRef]
  113. Kitryte, V.; Povilaitis, D.; Kraujaliene, V.; Sulniute, V.; Pukalskas, A.; Venskutonis, P.R. Fractionation of sea buckthorn pomace and seeds into valuable components by using high pressure and enzyme-assisted extraction methods. LWT Food Sci. Technol. 2017, 85, 534–538. [Google Scholar] [CrossRef]
  114. Santos, E.L.; Maia, B.H.L.N.S.; Ferriani, A.P.; Teixeira, S.D. Flavonoids: Classification, Biosynthesis and Chemical Ecology. In Flavonoids-From Biosynthesis to Human Health; Justino, G., Ed.; IntechOpen: London, UK, 2017; pp. 3–16. [Google Scholar]
  115. Niesen, D.B.; Hessler, C.; Seeram, N.P. Beyong resveratrol: A review of natural stilbenoids identified from 2009–2013. J. Berry Res. 2013, 3, 181–196. [Google Scholar] [CrossRef] [Green Version]
  116. Chong, J.; Poutaraud, A.; Hugueney, P. Metabolism and roles of stilbenes in plants. Plant Sci. 2009, 177, 143–155. [Google Scholar] [CrossRef]
  117. Chang, Z.; Zhang, Q.; Liang, W.; Zhou, K.; Jian, P.; She, G.; Zhang, L. A Comprehensive Review of the Structure Elucidation of Tannins from Terminalia Linn. Evidence-Based Complement. Altern. Med. 2019, 2019, 8623909. [Google Scholar] [CrossRef] [Green Version]
  118. Fraga-Corral, M.; Garcia-Oliveira, P.; Pereira, A.G.; Lourenco-Lopes, C.; Jimenez-Lopez, C.; Prieto, M.A.; Simal-Gandara, J. Technological application of tannin-based extracts. Molecules 2020, 25, 614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Babula, P.; Adam, V.; Havel, L.; Kizek, R. Noteworthy secondary metabolites naphthoquinones—Their occurrence, pharmacological properties and analysis. Curr. Pharm. Anal. 2009, 5, 47–68. [Google Scholar] [CrossRef]
  120. Venugopala, K.N.; Rashmi, V.; Odhav, B. Review on natural coumarin lead compounds for their pharmacological activity. BioMed Res. Int. 2013, 2013, 963248. [Google Scholar] [CrossRef] [Green Version]
  121. Itokawa, H.; Shi, Q.; Akiyama, T.; Morris-Natschke, S.L.; Lee, K.H. Recent advances in the investigation of curcuminoids. Chin. Med. 2008, 3, 11. [Google Scholar] [CrossRef] [Green Version]
  122. Amalraj, A.; Pius, A.; Gopi, S. Biological activities of curcuminoids, other biomolecules from turmeric and their derivatives—A review. J. Tradit. Complement. Med. 2017, 7, 205–233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Rodriguez-Garcia, C.; Sanchez-Quesada, C.; Toledo, E.; Delgado-Rodriguez, M.; Gaforio, J.J. Naturally lingnan-rich foods: A dietary tool for health promotion? Molecules 2019, 24, 917. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Choudhary, P.; Guleria, S.; Sharma, N.; Salaria, K.H.; Chalotra, R.; Ali, V.; Vyas, D. Comparative phenolic content and antioxidant activity of some medicinal plant extracts prepared by choline chloride based green solvents and methanol. Curr. Res. Green Sustain. Chem. 2021, 4, 100224. [Google Scholar] [CrossRef]
  125. Mishra, A.; Sharma, A.K.; Kumar, S.; Saxena, A.K.; Pandey, A.K. Bauhinia variegata leaf extracts exhibit considerable antibacterial, antioxidant, and anticancer activities. BioMed Res. Int. 2013, 2013, 915436. [Google Scholar] [CrossRef] [Green Version]
  126. Kumar Singh, S.; Patra, A. Evaluation of phenolic composition, antioxidant, anti-inflammatory and anticancer activities of Polygonatum verticillatum (L.). J. Integr. Med. 2018, 16, 273–282. [Google Scholar] [CrossRef] [PubMed]
  127. Singh, N.; Yadav, S.S. A review on health benefits of phenolics derived from dietary spices. Curr. Res. Food Sci. 2022, 5, 1508–1523. [Google Scholar] [CrossRef] [PubMed]
  128. Meng, X.-H.; Liu, C.; Zhug, L.Z.; Zhu, L.-F.; Yang, S.-X.; Zhu, H.-T.; Wang, D.; Yang, C.-R.; Zhang, Y.-J. Antioxidative Flavan-3-ol Dimers from the Leaves of Camellia fangchengensis. J. Agric. Food Chem. 2018, 66, 247–254. [Google Scholar] [CrossRef] [PubMed]
  129. Karakas, F.P.; Turker, A.U.; Karakas, A.; Mshvildadze, V.; Pichette, A.; Legault, J. I vitro cytotoxic, antibacterial, anti-inflammatory and antioxidant activities and phenolic content in wild-grown flowers of common daisy—A medicinal plant. J. Herb. Med. 2017, 8, 31–39. [Google Scholar] [CrossRef]
  130. Wang, L.; Li, N.; Yu, S.; Zhou, J. Enhancing caffeic acid production in Escherichia coli by engineering the biosynthesis pathway and transporter. Bioresour. Technol. 2023, 368, 128320. [Google Scholar] [CrossRef] [PubMed]
  131. Xue, T.-T.; Yang, Y.-G.; Tang, Z.-S.; Duan, J.-A.; Song, Z.-X.; Hu, X.-H.; Yang, H.-D.; Xu, H.-B. Evaluation of antioxidant, enzyme inhibition, nitric oxide production inhibitory activities and chemical profiles of the active extracts from the medicinal and edible plant: Althaea officinalis. Food Res. Int. 2022, 156, 111166. [Google Scholar] [CrossRef]
  132. Natta, S.; Mondol, S.A.; Pal, K.; Mandal, S.; Sahana, N.; Pal, R.; Pandit, G.K.; Alam, B.K.; Das, S.S.; Biswas, S.S.; et al. Chemical composition, antioxidant activity and bioactive constituents of six native endangered medicinal orchid species from north-eastern Himalayan region of India. S. Afr. J. Bot. 2022, 150, 248–259. [Google Scholar] [CrossRef]
  133. Cirak, C.; Radusiene, J.; Raudone, L.; Vilkickyte, G.; Seyis, F.; Marksa, M.; Ivanauskas, L.; Yayla, F. Phenolic compounds and antioxidant activity of Achillea arabica populations. S. Afr. J. Bot. 2022, 147, 425–433. [Google Scholar] [CrossRef]
  134. Zahoor, M.; Shafiq, S.; Ullah, H.; Sadiq, A.; Ullah, F. Isolation of quercetin and mandelic acid from Aesculus indica fruit and their biological activities. BMC Biochem. 2018, 19, 5. [Google Scholar] [CrossRef] [Green Version]
  135. Lungu, C.; Tuchilus, C.; Aprotosoaie, A.C.; Oprea, A.; Malterud, K.E.; Miron, A. Chemical, antioxidant and antimicrobial investigations of Pinus cembra L. bark and needles. Molecules 2011, 16, 7773–7788. [Google Scholar] [CrossRef] [Green Version]
  136. Pathiraja, D.; Wanasundara, J.P.D.; Elessawy, F.M.; Purves, R.W.; Vandenberg, A.; Shand, P.J. Water-soluble phenolic compounds and their putative antioxidant activities in the seed coats from different lentin (Lens culinaris) genotypes. Food Chem. 2023, 407, 135145. [Google Scholar] [CrossRef] [PubMed]
  137. Zhang, Y.; Li, Y.; Ren, X.; Zhang, X.; Wu, Z.; Liu, L. The positive correlation of antioxidant activity and prebiotic effect about oat phenolic compounds. Food Chem. 2023, 402, 134231. [Google Scholar] [CrossRef]
  138. Kebal, L.; Pokajewicz, K.; Djebli, N.; Mostefa, N.; Poliwoda, A.; Wieczorek, P.P. HPLC-DAD profile of phenolic compounds and in vitro antioxidant activity of Ficus carica L. fruits from two Algerian varieties. Biomed. Pharmacother. 2022, 155, 113738. [Google Scholar] [CrossRef] [PubMed]
  139. Richane, A.; Rim, B.M.; Wided, M.; Riadh, K.; Khaoula, A.; Nizar, M.; Hanen, B.I. Variability of phenolic compounds and antioxidant activities of ten Ceratonia siliqua L. provenances. Biochem. Syst. Ecol. 2022, 104, 104486. [Google Scholar] [CrossRef]
  140. Jarial, R.; Thakur, S.; Sakinah, M.; Zularisam, A.W.; Sharad, A.; Kanwar, S.S.; Singh, L. Potent anticancer, antioxidant and antibacterial activities of isolated flavonoids from Asplenium nidus. J. King Saud. Univ. Sci. 2018, 30, 185–192. [Google Scholar] [CrossRef]
  141. Tsai, P.J.; Huang, W.C.; Hsieh, M.C.; Sung, P.J.; Kuo, Y.H.; Wu, W.H. Flavones isolated from Scutellariae radix suppress Propionibacterium acnes-induced cytokine production in vitro and in vivo. Molecules 2016, 21, 15. [Google Scholar] [CrossRef] [Green Version]
  142. Lim, Y.-H.; Kim, I.-H.; Seo, J.-J. In vitro activity of kaempferol isolated from the Impatiens balsamina alone and in combination with erythromycin or clindamycin against Propionibacterium acnes. J. Microbiol. 2007, 45, 473–477. [Google Scholar]
  143. Poomanee, W.; Chaiyana, W.; Mueller, M.; Viernstein, H.; Khunkitti, W.; Leelapornpisid, P. In vitro investigation of anti-acne properties of Mangifera indica L. kernel extract and its mechanism of action against Propionibacterium acnes. Anaerobe 2018, 52, 64–74. [Google Scholar] [CrossRef]
  144. Duraipandiyan, V.; Ignacimuthu, S. Antifungal activity of traditional medicinal plants from Tamil Nadu, India. Asian Pac. J. Trop. Biomed. 2011, 1, S204–S215. [Google Scholar] [CrossRef]
  145. Hsieh, S.K.; Xu, J.R.; Lin, N.H.; Li, Y.C.; Chen, G.H.; Kuo, P.C.; Chen, W.Y.; Tzen, J.T.C. Antibacterial and laxative activities strictinin isolated from Pu er tea (Camellia sinensis). J. Food Drug Anal. 2016, 24, 722–729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Ahmed, S.I.; Hayat, M.Q.; Tahir, M.; Mansoor, Q.; Ismail, M.; Keck, K.; Bates, R.B. Pharmacologically active flavonoids from the anticancer, antioxidant, and antimicrobial extracts of Cassia angustifolia Vahl. BMC Complement. Altern Med. 2016, 16, 460. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Block, V.; Patterson, B.; Subar, A. Fruit, vegetables, and cancer prevention: A review of the epidemiological evidence. Nutr. Cancer 1992, 18, 1–29. [Google Scholar] [CrossRef]
  148. Brusselmans, K.; de Schrijver, E.; Heyns, W.; Verhoeven, G.; Swinnen, J.V. Epigallocatechin-3-gallate is a potent natural inhibitor of fatty acid synthase in intact cells and selectively induces apoptosis in prostate cancer cells. Int. J. Cancer 2003, 106, 856–862. [Google Scholar] [CrossRef]
  149. Danciu, C.; Vlaia, L.; Fetea, F.; Hancianu, M.; Coricovac, D.E.; Ciurlea, S.A.; Soica, C.M.; Marincu, I.; Vlaia, V.; Dehelean, C.A.; et al. Evaluation of phenolic profile, antioxidant, and anticancer potential of two main representants of Zingiberaceae family against B164A5 murie melanoma cells. Biol. Res. 2015, 48, 1. [Google Scholar] [CrossRef] [Green Version]
  150. Nwaeburu, C.C.; Abukiwan, A.; Zhao, Z.; Herr, I. Quercetin-induced miR-200b-3p regulates the mode of self-renewing divisions in pancreatic cancer. Mol. Cancer 2017, 16, 23. [Google Scholar] [CrossRef] [Green Version]
  151. Adjakly, M.; Ngollo, M.; Boiteux, J.P.; Bignon, Y.J.; Guy, L.; Bernard-Gallon, D. Genistein and daidzein: Different molecular effects on prostate cancer. Anticancer Res. 2013, 33, 39–44. [Google Scholar] [CrossRef]
  152. Dazialo, M.; Mierziak, J.; Korzun, U.; Preisner, M.; Szopa, J.; Kulma, A. The potential of plant phenolics in prevention and therapy of skin disorders. Int. J. Mol. Sci. 2016, 17, 160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Abusnina, A.; Keravis, T.; Yougbare, I.; Bronner, C.; Lugnier, C. Anti-proliferative effect of curcumin on melanoma cells is mediated by PDE1A inhibition that regulates the epigenetic integrator UHRF1. Mol. Nutr. Food Res. 2011, 55, 1677–1689. [Google Scholar] [CrossRef] [PubMed]
  154. Ide, H.; Lu, Y.; Noguchi, T.; Muto, S.; Okada, H.; Kawato, S.; Horie, S. Modulation of AKr1C2 by curcumin decreases testosterone production in prostate cancer. Cancer Sci. 2018, 109, 1230–1238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Dludla, P.V.; Joubert, E.; Muller, C.J.F.; Louw, J.; Johnson, R. Hyperglycemia-induced oxidative stress and heart disease-cardioprotective effects of rooibos flavonoids and phenylpyruvic acid-2-O-β-D-glucoside. Nutr. Metab. 2017, 14, 45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Garjani, A.; Tila, D.; Hamedeyazdan, S.; Vaez, H.; Rameshrad, M.; Pashaii, M.; Fathiazad, F. An investigation on cardioprotective potential of Marrubium vulgare aqueous fraction against ischaemia-reperfusion injury in isolated rat heart. Folia Morphol. 2017, 76, 361–371. [Google Scholar] [CrossRef] [Green Version]
  157. Razvani-Azarkhiavi, K.; Iranshahy, M.; Sahebkar, A.; Shirani, K.; Karimi, G. The protective role of phenolic compounds against doxorubicin-induced cardiotoxicity: A comprehensive review. Nutr. Cancer 2016, 68, 892–917. [Google Scholar] [CrossRef]
  158. Han, X.; Gao, S.; Cheng, Y.; Sun, Y.; Liu, W.; Tang, L.; Ren, D. Protective effect of naringenin-7-O-glucoside against oxidative stress induced by doxorubicin in H9c2 cardiomyocytes. Biosci. Trends 2012, 6, 19–25. [Google Scholar] [CrossRef] [Green Version]
  159. Morrison, D.K. MAP kinase pathways. Cold Spring Harb. Perspect. Biol. 2012, 4, a011254. [Google Scholar] [CrossRef] [Green Version]
  160. Sun, J.; Sun, G.; Meng, X.; Wang, H.; Luo, Y.; Qin, M.; Ma, B.; Wang, M.; Cai, D.; Guo, P.; et al. Isorhamnetin protects against doxorubicin-induced cardiotoxicity in vivo and in vitro. PLoS ONE 2013, 8, e64526. [Google Scholar] [CrossRef]
  161. Gao, Q.; Yang, B.; Ye, Z.-C.; Wang, J.; Bruce, I.C.; Xia, Q. Opening the calcium-activated potassium channel participates in the cardioprotective effect of puerarin. Eur. J. Pharmacol. 2007, 574, 179–184. [Google Scholar] [CrossRef]
  162. Kumar, S.; Pandey, A.K. Chemistry and biological activities of flavonoids: An overview. Sci. World J. 2013, 2013, 162750. [Google Scholar] [CrossRef] [Green Version]
  163. Chakraborty, R.; Biplab, D.; Devanna, N.; Sen, S. Antiinflammatory, antinociceptive and antioxidant activities of Phyllanthus acidus L. extracts. Asian Pac. J. Trop. Biomed. 2012, 2, S953–S961. [Google Scholar] [CrossRef]
  164. Kirmizibekmez, H.; Inan, Y.; Reis, R.; Sipahi, H.; Goren, A.C.; Yesilada, E. Phenolic compounds from the aerial parts of Clematis viticella L. and their in vitro anti-inflammatory activities. Nat. Prod. Res. 2019, 33, 2541–2544. [Google Scholar] [CrossRef] [PubMed]
  165. Cheng, H.-L.; Zhang, L.-J.; Liang, Y.-H.; Hsu, Y.-W.; Lee, I.-J.; Liaw, C.-C.; Hwang, S.-Y.; Kuo, Y.-H. Antiinflammatory and antioxidant flavonoids and phenols from Cardiospermum halicacabum. J. Tradit. Complement. Med. 2013, 3, 33–40. [Google Scholar] [CrossRef] [Green Version]
  166. Augusti, P.R.; Conterato, G.M.M.; Denardin, C.C.; Prazeres, I.D.; Serra, A.T.; Bronze, M.R.; Emanuelli, T. Bioactivity, bioavailability, and gut microbiota transformations of dietary phenolic compounds: Implications for COVID-19. J. Nutr. Biochem. 2021, 97, 108787. [Google Scholar] [CrossRef] [PubMed]
  167. Setayesh, M.; Karimi, M.; Zargaran, A.; Abousaidi, H.; Shahesmaeili, A.; Amiri, F.; Hasheminasab, F.S. Efficacy of a persian herbal medicine compound on coronavirus disease 2019 (COVID-19): A randomized controlled trial. Integr. Med. Res. 2022, 11, 100869. [Google Scholar] [CrossRef]
  168. Moharram, F.A.; El Dib, R.A.E.M.; Marzouk, M.S.; El-Shenawy, S.M.; Ibrahim, H.A. New apigenin glycoside, polyphenolic constituents, anti-inflammatory and hepatoprotective activities of Gaillardia grandiflora and Gaillardia pulchella aerial parts. Pharmacogn. Mag. 2017, 13, S244–S249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Compaore, M.; Bakasso, S.; Meda, R.; Nacoulma, O. Antioxidant and anti-inflammatory activities of fractions from Bidens engleri O.E. Schulz (Asteraceae) and Boerhavia erecta L. (Nyctaginaceae). Medicines 2018, 5, 53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Genc, Y.; Harput, U.S.; Saracoglu, I. Active compounds isolated from Plantago subulata L. via wound healing and antiinflammatory activity guided studies. J. Ethnopharmacol. 2019, 241, 112030. [Google Scholar] [CrossRef]
  171. Skendi, A.; Irakli, M.; Chatzopoulou, P.; Bouloumpasi, E.; Biliaderis, C.G. Phenolic extracts from solid wastes of the aromatic plant essential oil industry: Potential uses in food applications. Food Chem. Adv. 2022, 1, 100065. [Google Scholar] [CrossRef]
  172. Lu, C.L.; Zhu, Y.F.; Hu, M.M.; Wang, D.M.; Xu, X.J.; Lu, C.J.; Zhu, W. Optimization of astilbin extraction from the rhizome of Smilax glabra, and evaluation of its anti-inflammatory effect and probably underlying mechanism in lipopolysaccharide-induced RAW264.7 macrophages. Molecules 2015, 20, 625–644. [Google Scholar] [CrossRef]
  173. Gerhauser, C. Beer constituents as potential cancer chemopreventive agents. Eur. J. Cancer. 2005, 41, 1941–1954. [Google Scholar] [CrossRef] [PubMed]
  174. McKay, D.L.; Blumberg, J.B. A review of the bioactivity and potential health benefits of chamomile teas (Matricaria recutita L.). Phytother. Res. 2006, 20, 519–530. [Google Scholar] [CrossRef]
  175. Wach, A.; Pyrzynska, K.; Biesaga, M. Quercetin content in some food and herbal samples. Food Chem. 2007, 100, 699–704. [Google Scholar] [CrossRef]
  176. Bijak, M. Silybin, a major bioactive component of milk thistle (Silybum marianum L. Gaernt.)—Chemistry, bioavailability, and metabolism. Molecules 2017, 22, 1942. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Moore, J.O.; Wang, Y.; Stebbins, W.G.; Gao, D.; Zhou, X.; Phelps, R.; Lebwohl, M.; Wei, H. Photoprotective effect of isoflavone genistein on ultraviolet B-induced pyrimidine dimer formation and PCNA expression in human reconstituted skin and its implications in dermatology and prevention of cutaneous carcinogenesis. Carcinogenesis 2006, 27, 1627–1635. [Google Scholar] [CrossRef]
  178. Widyarini, S. Protective effect of the isoflavone equol against DNA damage induced by ultraviolet radiation to hairless mouse skin. J. Vet. Sci. 2006, 7, 217–223. [Google Scholar] [CrossRef] [Green Version]
  179. Setchell, K.D.R.; Clerici, C. Equol: History, chemistry, and formation. J. Nutr. 2010, 140, 1355S–1362S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Irace, C.; Marini, H.; Bitto, A.; Altavilla, D.; Polito, F.; Adamo, E.B.; Arcoraci, V.; Minutoli, L.; Di Benedetto, A.; Di Vieste, G. Genistein and endothelial function in postmenopausal women with metabolic syndrome. Eur. J. Clin. Investig. 2013, 43, 1025–1031. [Google Scholar] [CrossRef]
  181. Shahrajabian, M.H.; Petropoulos, S.A.; Sun, W. Survey of the influences of microbial biostimulants on horticultural crops: Case studies and successful paradigms. Horticulturae 2023, 9, 1–24. [Google Scholar] [CrossRef]
  182. Atun, S.; Handayani, S.; Rakhmawati, A.; Aini Purnamaningish, N.; An Naila, B.I.; Lestari, A. Study of potential phenolic compounds from stems of Dendrophthoe falcata (Loranthaceae) plant as antioxidant and antimicrobial agents. Orient. J. Chem. 2018, 34, 2342–2349. [Google Scholar] [CrossRef] [Green Version]
  183. Teixeira, T.S.; Vale, R.C.; Almeida, R.R.; Ferreira, T.P.S.; Guimaraes, G.L. Antioxidant potential and its correlation with the contents of phenolic compounds and flavonoids of methanolic extracts from different medicinal plants. Rev. Virtual Química 2017, 9, 1546–1559. [Google Scholar] [CrossRef]
  184. Xie, G.F.; Xu, X.Y.; Zhou, X.L.; Liu, Y.L.; Zhao, Z.B. Changes in phenolic profiles and antioxidant activity in rabbiteye blue berries during ripening. Int. J. Food Prop. 2019, 22, 320–329. [Google Scholar]
  185. Onder, A.; Izgi, M.N.; Cinar, A.S.; Zengin, G.; Yilmaz, M.A. The characterization of phenolic compounds via LC-ESI-MS/MS, antioxidant, enzyme inhibitory activities of Salvia absconditiflora, Salvia sclarea, and Salvia palaestina: A comparative analysis. S. Afr. J. Bot. 2022, 150, 313–322. [Google Scholar] [CrossRef]
  186. Tebii, S.O.; Debbache-Benaida, N.; Kadri, N.; Kadi, R.; Zaidi, S. A novel strategy to improve the recovery of phenolic compounds from Pistacia lentiscus L. fruits using design-based statistical modeling for ultrasound-deep eutectic solvents extraction and the evaluation of their antioxidant potential. Sustain. Chem. Pharm. 2023, 31, 100933. [Google Scholar] [CrossRef]
  187. Jiang, R.; Su, G.; Chen, X.; Chen, S.; Li, Q.; Xie, B.; Zhao, Y. Esculetin inhibits endometrial cancer proliferation and promotes apoptosis via hnRNPA1 to downregulate BCLXL and XIAP. Cancer Lett. 2021, 521, 308–321. [Google Scholar] [CrossRef] [PubMed]
  188. Ma, Y.-L.; Wang, Y.; Wu, Z.-F.; Mei, J.; Zhang, W.-Q.; Shang, Y.-F.; Thakur, K.; Wei, Z.-J. Exploring the effect of in vitro digestion on the phenolics and antioxidant activity of Lycium barbarum fruit extract. Food Biosci. 2023, 51, 102255. [Google Scholar] [CrossRef]
  189. Bahadori, M.B.; Zengin, G.; Dinparast, L.; Eskandani, M. The health benefits of three Hedgenettle herbal teas (Stachys byzantina, Stachys inflata, and Stachys lavandulifolia)- profiling phenolic and antioxidant activities. Eur. J. Integr. Med. 2020, 36, 101134. [Google Scholar] [CrossRef]
  190. Muddathir, A.M.; Yamauchi, K.; Batubara, I.; Mohieldin, E.A.M.; Mitsunaga, T. Anti-tyrosinase, total phenolic content and antioxidant activity of selected Sudanese medicinal plants. S. Afr. J. Bot. 2017, 109, 9–15. [Google Scholar] [CrossRef]
  191. Zekeya, N.; Ibrahim, M.; Mamiro, B.; Ndossi, H.; Kilonzo, M.; Mkangara, M.; Chacha, M.; Chilongola, J.; Kideghesho, J. Potential of natural phenolic antioxidant compounds from Bersama abyssinica (Meliathacea) for treatment of chronic diseases. Saudi J. Biol. Sci. 2022, 29, 103273. [Google Scholar] [CrossRef]
  192. Lin, Y.; Tang, D.; Liu, X.; Cheng, J.; Wang, X.; Guo, D.; Zou, J.; Yang, H. Phenolic profile and antioxidant activity of longan pulp of different cultivars from South China. LWT 2022, 165, 113698. [Google Scholar] [CrossRef]
  193. Cesar, L.-R.J.; Javier, H.; Fernando, A.-Z.J.; Carlos, V.; Enrique, R.-Z.R.; Efrain, A.; Evelin, M.-B.; Inocencio, H.-C.; Luis, O.J.; Zaira, D.; et al. Identification of the main phenolic compounds responsible for the antioxidant activity of Litsea glaucescens Kunth. S. Afr. J. Bot. 2022, 147, 208–214. [Google Scholar] [CrossRef]
  194. Acemi, R.K.; Acemi, A.; Cakir, M.; Polat, E.G.; Ozen, F. Preliminary screening the antioxidant potential of in vitro-propagated Amsonia orientalis: An example to sustainable use of rare medicinal plants in pharmaceutical studies. Sustain. Chem. Pharm. 2020, 17, 100302. [Google Scholar] [CrossRef]
  195. Limmongkon, A.; Nopprang, P.; Chaikeandee, P.; Somboon, T.; Wongshaya, P.; Pilaisangsuree, V. LC-MS/MS profiles and interrlationships between the anti-inflammatory activity, total phenolic content, and antioxidant potential of Kalasin 2 cultivar peanut sprout crude extract. Food Chem. 2018, 239, 569–578. [Google Scholar] [CrossRef]
  196. Vetuschi, A.; Battista, N.; Pompili, S.; Cappariello, A.; Prete, R.; Taticchi, A.; Selvaggini, R.; Latella, G.; Corsetti, A.; Sferra, R. The antiinflammatory and antifibrotic effect of olive phenols and Lactiplantibacillus plantarum IMC513 in dextran sodium sulfate-induced chronic colitis. Nutrition 2022, 94, 111511. [Google Scholar] [CrossRef]
  197. Demir, T.; Akpinar, O.; Kara, H.; Gungor, H. Phenolic profile and investigation of biological activities of Allium scorodoprasum L. subsp. rotundum. Food Biosci. 2022, 46, 101548. [Google Scholar] [CrossRef]
  198. Malik, J.; Tauchen, J.; Landa, P.; Kutil, Z.; Marsik, P.; Kloucek, P.; Havlik, J.; Kokoska, L. In vitro antiinflammatory and antioxidant potential of root extracts from Ranunculaceae species. S. Afr. J. Bot. 2017, 109, 128–137. [Google Scholar] [CrossRef]
  199. Kim, H.-J.; Park, C.-G.; Varghese, R.; Lee, J.Y.; Kim, Y.O.; Sung, G.-H. In-vitro antioxidative, antiinflammatory properties of Aurea helianthus leaf extract a Korean traditional medicinal plant. Saudi J. Biol. Sci. 2017, 24, 1943–1947. [Google Scholar] [CrossRef] [PubMed]
  200. Boga, M.; Ersoy, E.; Ozkan, E.E.; Cinar, E.; Kara, E.M.; Canturk, Y.Y.; Zengin, G. Volatile and phenolic profiling of a traditional medicinal plant, Hypericum empetrifolium with in vitro biological activities. J. Ethnopharmacol. 2021, 272, 113933. [Google Scholar] [CrossRef]
  201. Abirami, S.; Raj, B.E.; Soundarya, T.; Kannan, M.; Sugapriya, D.; Al-Dayan, N.; Mohammed, A.A. Exploring antifungal activities of acetone extract of selected Indian medicinal plants against human dermal fungal pathogens. Saudi J. Biol. Sci. 2021, 28, 2180–2187. [Google Scholar] [CrossRef]
  202. Barros, L.; Alves, C.T.; Dueñas, M.; Silva, S.; Oliveira, R.; Carvalho, A.M.; Henriques, M.; Santos-Buelga, C.; Ferreira, I.C.F.R. Characterization of phenolic compounds in wild medicinal flowers from Portugal by HPLC-DAD-ESI/MS and evaluation of antifungal properties. Ind. Crop. Prod. 2013, 44, 104–110. [Google Scholar] [CrossRef] [Green Version]
  203. Alishir, A.; Yu, J.S.; Park, M.; Kim, J.-C.; Pang, C.; Kim, J.K.; Jang, T.S.; Jung, W.H.; Kim, K.H. Ulmusakidian, a new coumarin glycoside and antifungal phenolic compounds from the root bark of Ulmus davidiana var. japonica. Bioorg. Med. Chem. Lett. 2021, 36, 127828. [Google Scholar] [CrossRef]
  204. Karimi, A.; Meiners, T. Antifungal activity of Zataria multiflora Boiss. Essential oils and changes in volatile compound composition under abiotic stress conditions. Ind. Crop. Prod. 2021, 171, 113888. [Google Scholar] [CrossRef]
  205. Munir, N.; Jiaz, W.; Altaf, I.; Naz, S. Evaluation of antifungal and antioxidant potential of two medicinal plants: Aconitum heterophyllum and Polygonum bistorta. Asian Pac. J. Trop. Biomed. 2014, 4, S639–S643. [Google Scholar] [CrossRef] [Green Version]
  206. Soleimani, M.; Arzani, A.; Arzani, V.; Roberts, T.H. Phenolic compounds and antimicrobial properties of mint and thyme. J. Herb. Med. 2022, 36, 100604. [Google Scholar] [CrossRef]
  207. Ghimire, B.K.; Seong, E.S.; Yu, C.Y.; Kim, S.-H.; Chung, I.-M. Evaluation of phenolic compounds and antimicrobial activities in transgenic Codonopsis lanceolata plans via overexpression of the γ-tocopherol methyltransferase (γ-tmt) gene. S. Afr. J. Bot. 2017, 109, 25–33. [Google Scholar] [CrossRef]
  208. Sruthi, P.; Roopavathi, C.; Naidu, M.M. Profiling of phenolics in cashew nut (Anacardium occidentale L.) testa and evaluation of their antioxidant and antimicrobial properties. Food Biosci. 2023, 51, 102246. [Google Scholar] [CrossRef]
  209. Muhammad, H.; Qasim, M.; Ikram, A.; Versiani, M.A.; Tahiri, I.A.; Yasmeen, K.; Abbasi, M.W.; Azeem, M.; Ali, S.T.; Gul, B. Antioxidant and antimicrobial activities of Ixora coccinea root and quantification of phenolic compounds using HPLC. S. Afr. J. Bot. 2020, 135, 71–79. [Google Scholar] [CrossRef]
  210. Oussaid, S.; Chibane, M.; Madani, K.; Amrouche, T.; Achat, S.; Dahmoune, F.; Houali, K.; Rendueles, M.; Diaz, M. Optimization of the extraction of phenolic compounds from Scripus holoschoenus using a simplex centroid design for antioxidant and antibacterial applications. LWT Food Sci. Technol. 2017, 86, 635–642. [Google Scholar] [CrossRef]
  211. Boussoussa, H.; Khacheba, I.; Djeridane, A.; Mellah, N.; Yousfi, M. Antibacterial activity from Rhanterium adpressum flowers extracts depending on seasonal variations. Ind. Crop. Prod. 2016, 83, 44–47. [Google Scholar] [CrossRef]
  212. Jang, J.Y.; Shin, H.; Lim, J.W.; Ahn, J.H.; Jo, Y.H.; Lee, K.Y.; Hwang, B.Y.; Jung, S.-J.; Kang, S.Y.; Lee, M.K. Comparison of antibacterial activity and phenolic constituents of bark, lignum, leaves, and fruit of Rhus verniciflua. PLoS ONE 2018, 13, e0200257. [Google Scholar] [CrossRef]
  213. Lin, T.; Huang, L.; Cheng, N.; Wang, Y.; Ning, Z.; Huang, S.; Wu, Y.; Chen, T.; Su, S.; Lin, Y. The in vitro and in vivo antibacterial activities of unilorous honey from a medicinal plant, Scrophularia ningpoensis Hemsl., and characterization of its chemical profile with UPLC-MS/MS. J. Ethnopharmacol. 2022, 296, 115499. [Google Scholar] [CrossRef]
  214. Guadie, A.; Dakone, D.; Unbushe, D.; Wang, A.; Xia, S. Antibacterial activity of selected medicinal plants used by traditional healers in Genta Meyche (Southern Ethiopia) for the treatment of gastrointestinal disorders. J. Herb. Med. 2020, 22, 100338. [Google Scholar] [CrossRef]
  215. Frankova, A.; Vistejnova, L.; Merinas-Amo, T.; Leheckova, Z.; Doskocil, I.; Soon, J.W.; Kudera, T.; Laupua, F.; Alonso-Moraga, A.; Kokoska, L. In vitro antibacterial activity of extracts from Samoan medicinal plants and their effect on proliferation and migration of human fibroblasts. J. Ethnopharmacol. 2021, 264, 113220. [Google Scholar] [CrossRef] [PubMed]
  216. Zahedipour, F.; Hosseini, S.A.; Sathyapalan, T.; Majeed, M.; Jamialahmadi, T.; Al-Rasadi, K.; Banach, M.; Sahebkar, A. Potential effects of curcumin in the treatment of COVID-19 infection. Phytother. Res. 2020, 34, 2911–2920. [Google Scholar] [CrossRef] [PubMed]
  217. Lung, J.; Lin, Y.-S.; Yang, Y.-H.; Chou, Y.-L.; Shu, L.-H.; Cheng, Y.-C.; Liu, H.T.; Wu, C.-Y. The potential chemical structure of anti-SARS-CoV-2 RNA-dependent RNA polymerase. J. Med. Virol. 2020, 92, 693–697. [Google Scholar] [CrossRef]
  218. Ghosh, R.; Chakraborty, A.; Biswas, A.; Chowdhuri, S. Evaluation of green tea polyphenols as novel corona virus (SARS CoV-2) main protease (Mpro) inhibitors—An in silico docking and molecular dynamics simulation study. J. Biomol. Struct. Dyn. 2021, 39, 4362–4374. [Google Scholar] [CrossRef] [PubMed]
  219. Xiao, T.; Cui, M.; Zheng, C.; Wang, M.; Sun, R.; Gao, D.; Bao, J.; Ren, S.; Lin, J.; Li, X. Myricetin inhibits SARS-CoV-2 viral replication by targeting M-pro and ameliorates pulmonary inflammation. Front. Pharmacol. 2021, 12, 1012. [Google Scholar] [CrossRef]
  220. Davella, R.; Gurrapu, S.; Mamidala, E. Phenolic compounds as promising drug candidates against COVID-19 an integrated molecular docking and dynamics simulation study. Mater. Today 2022, 51, 522–527. [Google Scholar] [CrossRef]
  221. Lin, C.-W.; Tsai, F.-J.; Tsai, C.-H.; Lai, C.-C.; Wang, L.; Ho, T.-Y.; Hsieh, C.-C.; Chao, P.-D.L. Anti-SARS coronavirus 3C-like protease effects of Isatis indigotica root and plant-derived phenolic compounds. Antivir. Res. 2005, 68, 36–42. [Google Scholar] [CrossRef]
  222. Weng, J.-R.; Lin, C.-S.; Lai, H.-C.; Lin, Y.-P.; Wang, C.-Y.; Tsai, Y.-C.; Wu, K.-C.; Huang, S.-H.; Lin, C.-W. Antiviral activity of Sambucus Formosana Nakai ethanol extract and related phenolic acid constituents against human coronavirus NL63. Virus Res. 2019, 273, 197767. [Google Scholar] [CrossRef]
  223. Wei, L.; Liao, Z.; Ma, H.; Wei, J.; Peng, C. Antioxidant properties, anti-SARS-CoV-2 study, collagenase and elastase inhibition effects, anti-human lung cancer potential of some phenolic compounds. J. Indian Chem. Soc. 2022, 99, 100416. [Google Scholar] [CrossRef]
  224. Granato, D.; Fidelis, M.; Haapakoski, M.; Lima, A.D.S.; Viil, J.; Hellstrom, J.; Ratsep, R.; Kaldmae, H.; Bleive, U.; Azevedo, L.; et al. Enzyme-assisted extraction of anthocyanins and other phenolic compounds from blackcurrant (Ribes nigrum L.) press cake: From processing to bioactivities. Food Chem. 2022, 391, 133240. [Google Scholar] [CrossRef]
  225. Conde, C.; Escribano, B.M.; Luque, E.; Aguilar-Luque, M.; Feijóo, M.; Ochoa, J.; Latorre, M.; Giraldo, A.; Lillo, R.; Aguera-Morales, E.; et al. The protective effect of extra-virgin olive oil in the experimental model of multiple sclerosis in the rat. Nutr. Neurosci. 2020, 23, 37–48. [Google Scholar] [CrossRef]
  226. Ibrahim, N.; Tadj, N.M.I.; Sarker, R.; Mohamed, I.N. The potential mechanisms of the neuroprotective actions of oil palm phenolics: Implications for neurodegenerative diseases. Molecules 2020, 25, 5159. [Google Scholar] [CrossRef]
  227. Yang, L.; Wang, Z.-M.; Wang, Y.; Li, R.-S.; Wang, F.; Wang, K. Phenolic constituents with neuroprotective activities from Hypericum wightianum. Phytochemistry 2019, 16, 112049. [Google Scholar] [CrossRef] [PubMed]
  228. Qneibi, M.; Hanania, M.; Jaradat, N.; Emwas, N.; Radwan, S. Inula viscosa (L.) Greuter, phytochemical composition, antioxidant, total phenolic content, total flavonoids content and neuroprotective effects. Eur. J. Integr. Med. 2021, 42, 101291. [Google Scholar] [CrossRef]
  229. Camargo, A.; Dalmagro, A.P.; Rebelo, A.M.; Reinke, C.K.; Zeni, A.L.B. Phenolic profile, antidepressant-like and neuroprotective effects of Maclura tinctoria leaves extract. Nat. Prod. Res. 2021, 36, 4692–4695. [Google Scholar] [CrossRef]
  230. Nunes, A.R.; Rodrigues, A.L.M.; Queiroz, D.B.D.; Vieira, I.G.P.; Neto, J.F.C.; Calixto Junior, J.T.; Tintino, S.R.; de Morais, S.M.; Coutinho, H.D.M. Photoprotective potential of medicinal plants from Cerrado biome (Brazil) in relation to phenolic content and antioxidant activity. J. Photochem. Photobiol. B Biol. 2018, 189, 119–123. [Google Scholar] [CrossRef]
  231. Ajjoun, M.; Kharchoufa, L.; Merrouni, I.A.; Elachouri, M. Moroccan medicinal plants traditionally used for the treatment of skin diseases: From ethnobotany to clinical trials. J. Ethnopharmacol. 2022, 297, 115532. [Google Scholar] [CrossRef]
  232. Li, Y.; Huang, J.; Lu, J.; Ding, Y.; Jiang, L.; Hu, S.; Chen, J.; Zeng, Q. The role and mechanism of Asian medicinal plants in treating skin pigmentary disorders. J. Ethnopharmacol. 2019, 245, 112173. [Google Scholar] [CrossRef]
  233. Maya-Cano, D.A.; Arango-Varela, S.; Santa-Gonzalez, G.A. Phenolic compounds of blueberries (Vaccinium spp.) as a protective strategy against skin cell damage induced by ROS: A review of antioxidant potential and antiproliferative capacity. Heliyon 2021, 7, e06297. [Google Scholar] [CrossRef]
  234. Xavier-Santos, J.B.; Passos, J.G.R.; Gomes, J.A.S.; Cruz, J.V.C.; Alves, J.S.F.; Garcia, V.B.; da Silva, R.M.; Lopes, N.P.; Araujo-Junior, R.F.; Zucolotto, S.M.; et al. Topical gen containin phenolic-rich extract from Ipomoea pes-capre leaf (Convolvulaceae) has anti-inflammatory, wound healing, and antiophidic properties. BioMed Pharmacother. 2022, 149, 112921. [Google Scholar] [CrossRef] [PubMed]
  235. Moglad, E.H.; Hamad, A.M.; Fatima, F.; Seshadri, V.D.; Naz, M. Antimicrobial and wound healing activities of certain Sudanese medicinal plants. Saudi J. Biol. Sci. 2020, 27, 1766–1772. [Google Scholar] [CrossRef] [PubMed]
  236. Ghuman, S.; Ncube, B.; Finnie, J.F.; McGaw, L.J.; Njoya, E.M.; Coopoosamy, R.M.; van Staden, J. Antioxidant, anti-inflammatory and wound healing properties of medicinal plant extracts used to treat wounds and dermatological disorders. S. Afr. J. Bot. 2019, 126, 232–240. [Google Scholar] [CrossRef]
  237. Nigussie, D.; Makonnen, E.; Tufa, T.B.; Brewster, M.; Legesse, B.A.; Fekadu, A.; Davey, G. Systematic review of Ethiopian medicinal plants used for their anti-inflammatory and wound healing activities. J. Ethnopharmacol. 2021, 276, 114179. [Google Scholar] [CrossRef] [PubMed]
  238. Yazarlu, O.; Iranshahi, M.; Kashani, H.R.K.; Reshadat, S.; Habtemariam, S.; Iranshahy, M.; Hasanpour, M. Perspective on the application of medicinal plants and natural products in wound healing: A mechanistic review. Pharmacol. Res. 2021, 174, 105841. [Google Scholar] [CrossRef] [PubMed]
  239. Asante-Kwatia, E.; Adjei, S.; Jibira, Y.; Gyimah, L.; Adjei-Hinneh, G.; Amponsah, I.K.; Mensah, A.Y. Amphimas pterocarpoides harms.: An evaluation of flavonoids and phenolic contents, wound healing, anthelmintic and antioxidant activities of the leaves and stem bark. Heliyon 2021, 7, e08261. [Google Scholar] [CrossRef] [PubMed]
  240. Enriquez-Ochoa, D.; Sanchez-Trasvina, C.; Hernandez-Sedas, B.; Mayolo-Deloisa, K.; Zavala, J.; Rito-Palomares, M.; Valdez-Garcia, J.E. Aqueous two-phase extraction of phenolic compounds from Sedum dendroideum with antioxidant activity and anti-proliferative properties against breast cancer cells. Sep. Purif. Technol. 2020, 251, 117341. [Google Scholar] [CrossRef]
  241. Eroglu, E.; Girin, S.N. A unique phenolic extraction method from oil macerate of Hypericum perforatum using DMSO: Assessment of in vitro anticancer activity, LC-MS/MS profile, total phenolic content and antioxidant capacity. S. Afr. J. Bot. 2021, 139, 6–11. [Google Scholar] [CrossRef]
  242. Al-Qahtani, J.; Abbasi, A.; Aati, H.Y.; Al-Taweel, A.; Al-Abdali, A.; Atai, S.; Yanbawi, A.N.; Khan, M.A.; Ghalloo, B.A.; Anwar, M.; et al. Phytochemical, antimicrobial, antidiabetic, thrombolytic, anticancer activities, and in silico studies of Ficus palmata Forssk. Arab. J. Chem. 2023, 16, 104455. [Google Scholar] [CrossRef]
  243. Sulaiman, C.T.; Deepak, M.; Praveen, T.K.; Lijini, K.R.; Salman, M.; Kumari, S.; Balachandran, I. Metabolite profiling and anti-cancer activity of two medicinally important Euphorbia species. Med. Omics 2022, 7, 100018. [Google Scholar] [CrossRef]
  244. Salehi, B.; Vlaisavljevic, S.; Adetunji, C.O.; Adetunji, J.B.; Kregiel, D.; Antolak, H.; Pawlikowska, E.; Uprety, Y.; Mileski, K.S.; Devkota, H.P.; et al. Plants of the genus Vitis: Phenolic compounds, anticancer properties and clinical relevance. Trends Food Sci. Technol. 2019, 91, 362–379. [Google Scholar] [CrossRef]
  245. Tauchen, J.; Huml, L.; Bortl, L.; Doskocil, I.; Jarosova, V.; Marsik, P.; Frankova, A.; Peralta, Z.M.C.; Zans, M.-E.C.; Havlik, J.; et al. Screening of medicinal plants traditionally used in Peruvian Amazon for in vitro antioxidant and anticancer potential. Nat. Prod. Res. 2019, 33, 2718–2721. [Google Scholar] [CrossRef] [PubMed]
  246. Khalid, M.; Amayreh, M.; Sanduka, S.; Salah, Z.; Al-Rimawi, F.; Al-Mazaideh, G.M.; Alanezi, A.A.; Wedian, F.; Alasmari, F.; Shalayel, M.H.F. Assessment of antioxidant, antimicrobial, and anticancer activities of Sisymbrium officinale plant extract. Heliyon 2022, 8, e10477. [Google Scholar] [CrossRef]
  247. Ogawa, S.; Takafuji, K.; Tsubuku, S.; Horie, Y.; Ikegawa, S.; Higashi, T. Isotope-coded derivatization based LC/ESI-MS/MS methods using a pair of novel reagents for quantification of hydroxycinnamic acids and hydroxybenzoic acids in fermented brown rice product. J. Pharm. Biomed. Anal. 2017, 142, 162–170. [Google Scholar] [CrossRef]
  248. Ossipov, V.; Zubova, M.; Nechaeva, T.; Zagoskina, N.; Salminen, J.-P. The regulating effect of light on the content of flavan-3-ols and derivatives of hydroxybenzoic acids in the callus culture of the tea plant, Camellia sinensis L. Biochem. Syst. Ecol. 2022, 101, 104384. [Google Scholar] [CrossRef]
  249. Joshi, A.N.; Chandrakar, A.K.; Wasewar, K.L. Extractive separation of 4-hydroxybenzoic acid from aqueous solution using nontoxic and conventional solvents. Chem. Data Collect. 2021, 36, 100782. [Google Scholar] [CrossRef]
  250. Saenz-Castillo, A.; Sanabria-Chinchilla, J.; Garcia-Pineres, A.J.; Tamayo-Castillo, G. Bioactivity of prenylated hydroxybenzoic acids from Piper garagaranum C. DC. Phytochem. Lett. 2022, 47, 28–33. [Google Scholar] [CrossRef]
  251. Osamudiamen, P.M.; Oluremi, B.B.; Oderinlo, O.O.; Aiyelaagbe, O.O. Trans-resveratrol, piceatannol and gallic acid: Potent polyphenols isolated from Mezoneuron benthamianum effective as anticaries, antioxidant and cytotoxic agents. Sci. Afr. 2020, 8, e00244. [Google Scholar] [CrossRef]
  252. Asdaq, S.M.B.; Alamri, A.S.; Alsanie, W.F.; Alhomrani, M.; Yasmin, F. Potential benefits of gallic acid as skeletal muscle relaxant in animal experimental models. Saudi J. Biol. Sci. 2021, 28, 7575–7580. [Google Scholar] [CrossRef]
  253. Behera, P.K.; Devi, S.; Mittal, N. Therapeutic potential of gallic acid in obesity: Considerable shift. Obes. Med. 2023, 37, 100473. [Google Scholar] [CrossRef]
  254. Gong, W.; Wang, R.; Huang, H.; Hou, Y.; Wang, X.; He, W.; Gong, X.; Hu, J. Construction of double network hydrogels using agarose and gallic acid with antibacterial and anti-inflammatory properties for wound healing. Int. J. Biol. Macromol. 2023, 227, 698–710. [Google Scholar] [CrossRef] [PubMed]
  255. Tan, Q.; An, X.; Pan, S.; Zhen, S.; Hu, Y.; Hu, X. A facile and sensitive ratiometric fluorescent sensor for determination of gallic acid. Microchem. J. 2022, 172 Pt B, 106922. [Google Scholar] [CrossRef]
  256. Variya, B.C.; Bakrania, A.K.; Patel, S.S. Antidiabetic potential of gallic acid from Emblica officinalis: Improved glucose transporters and insulin sensitivity through PPAR-γ and Akt signaling. Phytomedicine 2020, 73, 152906. [Google Scholar] [CrossRef] [PubMed]
  257. Cechinel-Zanchett, C.C.; Mariano, L.N.B.; Schlickmann, F.; Cechinel-Filho, V.; de Souza, P. In vitro effects of two bioactive compounds, gallic acid and methyl gallate, on urolithiasis Efecto in vitro de dos compuestos bioactivos, el acido galico y el galato de metilo, sobre la urolitiasis. Actas Urol. Esp. 2021, 45, 604–608. [Google Scholar] [CrossRef]
  258. Yang, K.; Jian, S.; Guo, D.; Wen, C.; Xin, Z.; Zhang, L.; Kuang, T.; Wen, J.; Yin, Y.; Deng, B. Fecal microbiota and metabolomics revealed the effect of long-term consumption of gallic acid on canine lipid metabolism and gut health. Food Chem. 2022, 15, 100377. [Google Scholar] [CrossRef]
  259. Liao, W.; Wen, Y.; Wang, J.; Zhao, M.; Iv, S.; Chen, N.; Li, Y.; Wang, L.; Zheng, Q.; Mou, Y.; et al. Gallic acid alleviates gastric precancerous lesions through inhibition of epithelial mesenchymal transition via Wnt/β-catenin signaling pathway. J. Ethnopharmacol. 2023, 302 Pt A, 115885. [Google Scholar] [CrossRef]
  260. Zhang, Y.; Wang, X.; Lu, B.; Gao, Y.; Zhang, Y.; Li, Y.; Niu, H.; Fan, L.; Pang, Z.; Qiao, Y. Functional and binding studies of gallic acid showing platelet aggregation inhibitory effect as a thrombin inhibitor. Chin. Herb. Med. 2022, 14, 303–309. [Google Scholar] [CrossRef]
  261. Hasan, Z.; Islam, A.; Khan, L.A. Spectroscopic investigations on fungal aspartic protease as target of gallic acid. Int. J. Biol. Macromol. 2022, 228, 333–345. [Google Scholar] [CrossRef]
  262. Clark, M.; Centner, A.M.; Ukhanov, V.; Nagpal, R.; Salazar, G. Gallic acid ameliorates atherosclerosis and vascular senescence and remodels the microbiome in a sex-dependent manner in ApoE−/− mice. J. Nutr. Biochem. 2022, 110, 109132. [Google Scholar] [CrossRef]
  263. Moradi, A.; Abolfathi, M.; Javadian, M.; Heidarian, E.; Roshanmehr, H.; Khaledi, M.; Nouri, A. Gallic acid exerts nephroprotective, anti-oxidative stress, and anti-inflammatory effects against diclofenac-induced renal injury in malerats. Arch. Med. Res. 2021, 52, 380–388. [Google Scholar] [CrossRef]
  264. Erukainure, O.L.; Hafizur, R.M.; Choudhary, M.I.; Adhikari, A.; Mesaik, A.M.; Atolani, O.; Banerjee, P.; Preissner, R.; Muhammad, A.; Islam, S. Anti-diabetic effect of the ethyl acetate fraction of Clerodendrum volubile: Protocatechuic acid suppresses phagocytic oxidative burst and modulates inflammatory cytokines. Biomed. Pharmacother. 2017, 86, 307–315. [Google Scholar] [CrossRef] [PubMed]
  265. Dare, R.G.; Oliveira, M.M.; Truiti, M.C.T.; Nakamura, C.V.; Ximenes, V.F.; Lautenschlager, S.O.S. Abilities of protocatechuic acid and its alkylesters, ethyl and heptyl protocatechuates, to counteract UVB-induced oxidative injuries and photoaging in fibroblasts L929 cell line. J. Photochem. Photobiol. B Biol. 2020, 203, 111771. [Google Scholar] [CrossRef] [PubMed]
  266. Krishna, P.U.N.; Muraleedharan, K. Metal chelation ability of protocatechuic acid anion with 210Po84; A theoretical insight. Comput. Theor. Chem. 2022, 1220, 113996. [Google Scholar] [CrossRef]
  267. Da-Costa-Rocha, I.; Bonnlaender, B.; Sievers, H.; Pischel, I.; Heinrich, M. Hibiscus sabdariffa L.—A phytochemical and pharmacological review. Food Chem. 2014, 165, 424–443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  268. Adedara, I.A.; Omole, O.; Okpara, E.S.; Fasina, O.B.; Ayeni, M.F.; Ajayi, O.M.; Busari, E.O.; Farombi, E.O. Impact of prepubertal exposure to dietary protocatchuic acid on the hypothalamic-pituitary-testicular axis in rats. Chem. Biol. Interact. 2018, 290, 99–109. [Google Scholar] [CrossRef]
  269. Salama, A.; Elgohary, R.; Amin, M.M.; Elwahab, S.A. Immunomodulatory effect of protocatechuic acid on cyclophosphamide induced brain injury in rat: Modulation of inflammosomes NLRP3 and SIRT1. Eur. J. Pharmacol. 2022, 932, 175217. [Google Scholar] [CrossRef]
  270. Song, J.; He, Y.; Luo, C.; Feng, B.; Ran, F.; Xu, H.; Ci, Z.; Xu, R.; Han, L.; Zhang, D. New progress in the pharmacology of protocatechuic acid: A compounds ingested in daily foods and herbs frequently and heavily. Pharmacol. Res. 2020, 161, 105109. [Google Scholar] [CrossRef]
  271. Antony, F.M.; Wasewar, K. Effect of temperature on equilibria for physical and reactive extraction of protocatechuic acid. Heliyon 2020, 6, e03664. [Google Scholar] [CrossRef]
  272. Chen, J.; Dai, X.; Jiang, C.; Fu, Y.; Jiang, T.; Tang, L.; Wang, L.; Wang, Q.; Huang, G.; Cao, J. One new protocatechuic acid methyl ester and one enantiomeric pair of dihydroflavones isolated from Phymatopteris hastata. Phytochem. Lett. 2021, 43, 130–134. [Google Scholar] [CrossRef]
  273. Yamabe, N.; Park, J.Y.; Lee, S.; Cho, E.-J.; Lee, S.; Kang, K.S.; Hwang, G.S.; Kim, S.-N.; Kim, H.Y.; Shibamoto, T. Protective effects of protocatechuic acid against cisplatin-induced renal damage in rats. J. Funct. Foods 2015, 19 Pt A, 20–27. [Google Scholar] [CrossRef]
  274. Al-Olayan, E.M.; Aloufi, A.S.; Al-Amri, O.D.; El-Habit, O.H.; Moneim, A.E.A. Protocatechuic acid mitigates cadmium-induced neurotoxicity in rats: Role of oxidative stress, inflammation and apoptosis. Sci. Total Environ. 2020, 723, 137969. [Google Scholar] [CrossRef] [PubMed]
  275. Stojkovic, D.S.; Zivkovic, J.; Sokovic, M.; Glamoclija, J.; Ferreira, I.C.F.R.; Jankovic, T.; Maksimovic, Z. Antibacterial activity of Veronica montana L. extract and protocatechuic acid incorporated in a food system. Food Chem. Toxicol. 2013, 55, 209–213. [Google Scholar] [CrossRef] [PubMed]
  276. Yuan, Y.; Xiang, J.; Zheng, B.; Sun, J.; Luo, D.; Li, P.; Fan, J. Diversity of phenolics including hydroxycinnamic acid amide derivatives phenolic acids contribute to antioxidant properties of proso millet. LWT 2022, 154, 112611. [Google Scholar] [CrossRef]
  277. Schroter, D.; Baldermann, S.; Schreiner, M.; Witzel, K.; Maul, R.; Rohn, S.; Neugart, S. Natural diversity of hydroxycinnamic acid derivatives, flavonoid glycosides, carotenoids and chlorophylls in leaves of six different amaranth species. Food Chem. 2018, 267, 376–386. [Google Scholar] [CrossRef]
  278. Xiang, J.; Zhang, M.; Apea-Bah, F.; Beta, T. Hydroxycinnamic acid amide (HCAA) derivatives, flavonoid C-glycosides, phenolic acids and antioxidant properties of foxtail millet. Food Chem. 2019, 295, 214–223. [Google Scholar] [CrossRef]
  279. Bijalwan, V.; Ali, U.; Kesarwani, A.K.; Yadav, K.; Mazumder, K. Hydroxycinnamic acid bound arabinoxylans from millet brans-structural features and antioxidant activity. Int. J. Biol. Macromol. 2016, 88, 296–305. [Google Scholar] [CrossRef] [PubMed]
  280. Ribas-Agusti, A.; Martin-Belloso, O.; Soliva-Fortuny, R.; Elez-Martinez, P. Enhancing hydroxycinnamic acids and flavan-3-ol contents by pulsed electric fields without affecting quality attributes of apple. Food Res. Int. 2019, 121, 433–440. [Google Scholar] [CrossRef]
  281. Bal, S.S.; Leishangthem, G.D.; Sethi, R.S.; Singh, A. p-coumaric acid ameliorates fipronil induced liver injury in mice through attenuation of structural changes, oxidative stress and inflammation. Pestic. Biochem. Physiol. 2022, 180, 104997. [Google Scholar] [CrossRef]
  282. Combes, J.; Imatoukene, N.; Couvreur, J.; Godon, B.; Fojcik, C.; Allais, F.; Lopez, M. An optimized semi-defined medium for p-coumaric acid production in extractive fermentation. Process. Biochem. 2022, 122 Pt 2, 357–362. [Google Scholar] [CrossRef]
  283. Naumowicz, M.; Kusaczuk, M.; Kruszewski, M.A.; Gal, M.; Kreowski, R.; Cechowska-Pasko, M.; Kotynska, J. The modulating effect of lipid bilayer/p-coumaric acid interactions on electrical properties of model lipid membranes and human glioblastoma cells. Bioorg. Chem. 2019, 92, 103242. [Google Scholar] [CrossRef] [PubMed]
  284. Grodzicka, M.; Pena-Gonzalez, C.E.; Ortega, P.; Michlewska, S.; Lozano, R.; Bryszewska, M.; Mata, F.J.D.I.; Ionov, M. Heterofunctionalized polyphenolic dendrimers decorated with caffeic acid: Synthesis, characterization and antioxidant activity. Sustain. Mater. Technol. 2022, 33, e00497. [Google Scholar] [CrossRef]
  285. Kfoury, M.; Geagea, C.; Ruellan, S.; Greige-Gerges, H.; Fourmentin, S. Effect of cyclodextrin and cosolvent on the solubility and antioxidant activity of caffeic acid. Food Chem. 2019, 278, 163–169. [Google Scholar] [CrossRef]
  286. Raviadaran, R.; Ng, M.H.; Chandran, D.; Ooi, K.K.; Manickam, S. Stable W/O/W multiple nanoemulsion encapsulating natural tocotrienols and caffeic acid with cisplatin synergistically treated cancer cell lines (A549 and HEP G2), and reduced toxicity on normal cell line (HEK 293). Mater. Sci. Eng. C 2021, 121, 111808. [Google Scholar] [CrossRef] [PubMed]
  287. Tabakam, G.T.; Kodama, T.; Donfack, A.R.N.; Nguekeu, Y.M.M.; Nomin-Erdene, B.; Htoo, Z.P.; Do, K.M.; Ngouela, S.A.; Tene, M.; Morita, H.; et al. A new caffeic acid ester and a new ceramide from the roots of Eriosema glomeratum. Phytochem. Lett. 2021, 45, 82–87. [Google Scholar] [CrossRef]
  288. Salsabila, R.; Perdani, M.S.; Kitakawa, N.S.; Hermansyah, H. Production of methyl caffeate as an intermediate product to produce caffeic acid phenethyl ester by esterification using cation-exchange resin. Energy Rep. 2020, 6, 528–533. [Google Scholar] [CrossRef]
  289. Mirzaei, S.; Gholami, M.H.; Zabolian, A.; Saleki, H.; Farahani, M.V.; Hamzehlou, S.; Bakhtiari Far, F.; Sharifzadeh, S.O.; Samarghandian, S.; Khan, H.; et al. Caffeic acid and its derivatives as potential modulators of oncogenic molecular pathways: New hope in the fight against cancer. Pharmacol. Res. 2021, 171, 105759. [Google Scholar] [CrossRef]
  290. Kar, A.; Panda, S.; Singh, M.; Biswas, S. Regulation of PTU-induced hypothyroidism in rats by caffeic acid primarily by activating thyrotropin receptors and by inhibiting oxidative stress. Phytomed. Plus 2022, 2, 100298. [Google Scholar] [CrossRef]
  291. Meinhart, A.D.; Damin, F.M.; Caldeirao, L.; Filho, M.D.J.; da Silva, L.C.; Constant, L.D.S.; Filho, J.T.; Wagner, R.; Godoy, H.T. Chlorogenic and caffeic acids in 64 fruits consumed in Brazil. Food Chem. 2019, 286, 51–63. [Google Scholar] [CrossRef]
  292. Silva, A.D.S.; Pereira-de-Morais, L.; da Silva, R.E.R.; Dantas, D.D.M.; Milfont, C.G.B.; Gomes, M.F.; Araujo, I.M.; Kerntopf, M.R.; Menezes, I.R.A.D.; Barbosa, R. Pharmacological screening of the phenolic compound caffeic acid using rat aorta, uterus and ileum smooth muscle. Chem. Biol. Interact. 2020, 332, 109269. [Google Scholar] [CrossRef]
  293. Wang, X.; Liu, X.; Shi, N.; Zhang, Z.; Chen, Y.; Yan, M.; Li, Y. Response surface methodology optimization and HPLC-ESI-QTOF-MS/MS analysis on ultrasonic-assisted extraction of phenolic compounds from okra (Abelmoschus esculentus) and their antioxidant activity. Food Chem. 2023, 405 Pt B, 134966. [Google Scholar] [CrossRef] [PubMed]
  294. Trifan, A.; Skalicka-Wozniak, K.; Granica, S.; Czerwinska, M.E.; Kruk, A.; Marcourt, L.; Wolfender, J.-L.; Wolfram, E.; Esslinger, N.; Grubelnik, A.; et al. Symphytum officinale L.: Liquid-liquid chromatography isolation of caffeic acid oligomers and evaluation of their influence on pro-inflammatory cytokine release in LPS-stimulated neutrophils. J. Ethnopharmacol. 2020, 262, 113169. [Google Scholar] [CrossRef] [PubMed]
  295. Jung, W.-K.; Lee, D.-Y.; Kim, J.-H.; Choi, I.; Park, S.-G.; Seo, S.-K.; Lee, S.-W.; Lee, C.-M.; Park, Y.-M.; Jeon, Y.-J.; et al. Anti-inflammatory activity of caffeic acid phenethyl ester (CAPE) extracted from Rhodiola sacra against lipopolysaccharide-induced inflammatory responses in mice. Process. Biochem. 2008, 43, 783–787. [Google Scholar] [CrossRef]
  296. Monteiro, A.B.; Rodrigues, C.K.D.S.; Nascimento, E.P.D.; Sales, V.D.S.; Delmondes, G.D.A.; Costa, M.H.N.D.; Oliveira, V.A.P.D.; Morais, L.P.D.; Boligon, A.A.; Barbosa, R.; et al. Anxiolytic and antidepressant-like effects of Annona coriacea (Mart.) and caffeic acid in mice. Food Chem. Toxicol. 2020, 136, 111049. [Google Scholar] [CrossRef]
  297. Spagnol, C.M.; Assis, R.P.; Brunetti, I.L.; Isaac, V.L.B.; Salgado, H.R.N.; Correa, M.A. In vitro methods to determine the antioxidant activity of caffeic acid. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2019, 219, 358–366. [Google Scholar] [CrossRef]
  298. Mu, H.-N.; Zhou, Q.; Yang, R.-Y.; Tang, W.-Q.; Li, H.-X.; Wang, S.-M.; Li, J.; Chen, W.-X.; Dong, J. Caffeic acid prevents non-alcoholic fatty liver disease induced by a high-fat diet through gut microbiota modulation in mice. Food Res. Int. 2021, 143, 110240. [Google Scholar] [CrossRef]
  299. Koga, M.; Nakagawa, S.; Kato, A.; Kusumi, I. Caffeic acid reduces oxidative stress and microglial activation in the mouse hippocampus. Tissue Cell 2019, 60, 14–20. [Google Scholar] [CrossRef]
  300. Bao, Y.; Chen, Q.; Xie, Y.; Tao, Z.; Jin, K.; Chen, S.; Bai, Y.; Yang, J.; Shan, S. Ferulic acid attenuates oxidative DNA damage and inflammatory responses in microglia induced by benzo(a)pyrene. Int. Immunopharmacol. 2019, 77, 105980. [Google Scholar] [CrossRef]
  301. Calabrese, E.J.; Agathokleous, E.; Calabrese, V. Ferulic acid and hormesis: Biomedical and environmental implications. Mech. Ageing Dev. 2021, 198, 111544. [Google Scholar] [CrossRef]
  302. Pinheiro, P.G.; Santiago, G.M.P.; Silva, F.E.F.D.; Araujo, A.C.J.D.; Oliveira, C.R.T.D.; Freitas, P.R.; Rocha, J.E.; Neto, J.B.D.A.; da Silva, M.M.C.; Tintino, S.R.; et al. Ferulic acid derivatives inhibiting Staphylococcs aureus tetK and MsrA efflux pumps. Biotechnol. Rep. 2022, 34, e00717. [Google Scholar] [CrossRef]
  303. Raj, N.D.; Singh, D. A critical appraisal on ferulic acid: Biological profile, biopharmaceutical challenges nano formulations. Health Sci. Rev. 2022, 5, 100063. [Google Scholar] [CrossRef]
  304. Zhang, L.-W.; Al-Sywayeh, S.A.; Hsieh, P.-W.; Fang, J.-Y. A comparison of skin delivery of ferulic acid and its derivatives: Evaluation of their efficacy and safety. Int. J. Pharm. 2010, 399, 44–51. [Google Scholar] [CrossRef]
  305. Ali, S.A.; Saifi, M.a.; Pulivendala, G.; Godugu, C.; Talla, V. Ferulix acid ameliorates the progression of pulmonary fibrosis via inhibition of TGF-β/smad signalling. Food Chem. Toxicol. 2021, 149, 111980. [Google Scholar] [CrossRef]
  306. Cao, C.-N.; Liu, C.-F.; Zhao, L.; Rao, G.-W. New insight into the photoinduced wavelenght dependent decay mechanisms of the ferulic acid system on the excited states. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2020, 240, 118565. [Google Scholar] [CrossRef]
  307. Rosa, L.; Jordao, N.; Soares, N.; de Mesquita, J.; Monteiro, M.; Teodoro, A. Pharmacokinetic, antiproliferative and apoptotic effects of phenolic acids in human colon adenocarcinoma cells using in vitro and in silico approaches. Molecules 2018, 23, 2569. [Google Scholar] [CrossRef] [Green Version]
  308. Wang, X.; He, Y.; Tian, J.; Muhammad, I.; Liu, M.; Wu, C.; Xu, C.; Zhang, X. Ferulic acid prevents aflatoxin B1-induced liver injury in rats via inhibiting cytochrome P450 enzyme, activating Nrf2/GST pathway and regulating mitochondrial pathway. Ecotoxicol. Environ. Saf. 2021, 224, 112624. [Google Scholar] [CrossRef] [PubMed]
  309. Pazo-Cepeda, M.V.; Aspromonte, S.G.; Alonso, E. Extraction of ferulic acid and feruloylated arabinoxylo-oligosaccharides from wheat bran using pressurized hot water. Food Biosci. 2021, 44, 101374. [Google Scholar] [CrossRef]
  310. Singh, S.; Arthur, R.; Upadhayay, S.; Kumar, P. Ferulic acid ameliorates neurodegeneration via the Nrf2/ARE signalling pathways: A review. Pharmacol. Res. Mod. Chin. Med. 2022, 5, 100190. [Google Scholar] [CrossRef]
  311. Li, J.; Wang, C.; Chen, X.; Huang, M.; Fu, Q.; Li, R.; Wang, Y.; Li, C.; Zhao, P.; Xie, Y.; et al. A non-enzymatic photoelectrochemical sensor based on g-C3N4@CNT heterojunction for sensitive detection of antioxidant gallic acid in food. Food Chem. 2022, 389, 133086. [Google Scholar] [CrossRef]
  312. Wang, Q.-H.; Qin, S.-W.; Jiang, J.-G. Improvement effects of esculetin on the formation and development of atherosclerosis. Biomed. Pharmacother. 2022, 150, 113001. [Google Scholar] [CrossRef] [PubMed]
  313. Sabry, M.M.; Abdel-Rahman, R.F.; El-Shenawy, S.; Hassan, A.M.; El-Gayed, S.H. Estrogenic activity of Sage (Salvia officinalis L.) aerial parts and its isolated ferulic acid in immature ovariectomized female rats. J. Ethnopharmacol. 2022, 282, 114579. [Google Scholar] [CrossRef] [PubMed]
  314. Singh, Y.P.; Rai, H.; Singh, G.; Singh, G.K.; Mishra, S.; Kumar, S.; Srikrishna, S.; Modi, G. A review on ferulic acid and analogs based scaffolds for the management of Alzheimers disease. Eur. J. Med. Chem. 2021, 215, 113278. [Google Scholar] [CrossRef] [PubMed]
  315. Elhessy, H.M.; Eltahry, H.; Erfan, O.S.; Mahdi, M.R.; Hazem, N.M.; El-Shahat, M.A. Evaluation of the modulation of nitric oxide synthase expression in the cerebellum of diabetic albino rats and the possible protective effect of ferulic acid. Acta Histochem. 2020, 122, 151633. [Google Scholar] [CrossRef]
  316. Ramar, M.; Manikandan, B.; Raman, T.; Priyadarsini, A.; Palanisamy, S.; Velayudam, M.; Munusamy, A.; Prabhu, N.M.; Vaseeharan, B. Protective effect of ferulic acid and resveratrol against alloxan-induced diabetesin mice. Eur. J. Pharmacol. 2012, 690, 226–235. [Google Scholar] [CrossRef] [PubMed]
  317. Shanthakumar, J.; Karthikeyan, A.; Bandugula, V.R.; Prasad, N.R. Ferulic acid, a dietary phenolic acid, modulates radiation effects in Swiss albino mice. Eur. J. Pharmcol. 2012, 691, 268–274. [Google Scholar] [CrossRef] [PubMed]
  318. Hu, R.; Wu, S.; Li, B.; Tan, J.; Yan, J.; Wang, Y.; Tang, Z.; Liu, M.; Fu, C.; Zhang, H.; et al. Dietary ferulic acid and vanillic acid on inflammation, gut barrier function and growth performance in lipopolysaccharide-challened piglets. Anim. Nutr. 2022, 8, 144–152. [Google Scholar] [CrossRef]
  319. Alam, M.A.; Sernia, C.; Brown, L. Ferulic acid improves cardiovascular and kidney structure and function in hypertensive rats. J. Cardiovasc. Pharmacol. 2013, 61, 240–249. [Google Scholar] [CrossRef]
  320. Cheng, W.-J.; Zhang, P.-P.; Luo, Q.-Q.; Deng, S.-M.; Jia, A.-Q. The chemosensitizer ferulic acid enhances epirubicin-induced apoptosis in MDA-MB-231 cells. J. Funct. Foods. 2020, 73, 104130. [Google Scholar] [CrossRef]
  321. Von Danwitz, A.; Schulz, C. Effects of dietary rapeseed glucosinolates, sinapic acid and phytic acid on feed intake growth performance and fish health in turbot (Psetta maxima L.). Aquaculture 2020, 516, 734624. [Google Scholar] [CrossRef]
  322. Kaur, J.; Mehta, V.; Kaur, G. Preparation, development and characterization of Leucaena leucocephala galactomannan (LLG) conjugated sinapic acid: A potential colon targeted prodrug. Int. J. Biol. Macromolecul. 2021, 178, 29–40. [Google Scholar] [CrossRef]
  323. Roy, S.J.; Prince, P.S.M. Protective effects of sinapic acid on lysosomal dysfunction in isoproterenol induced myocardial infarcted rats. Food Chem. Toxicol. 2012, 50, 3984–3989. [Google Scholar] [CrossRef] [PubMed]
  324. Saeedavi, M.; Goudarzi, M.; Mehrzadi, S.; Basir, Z.; Hasanvand, A.; Hosseinzadeh, A. Sinapic acid ameliorates airway inflammation in murine ovalbumin-induced allergic astham by reducing Th2 cytokine production. Life Sci. 2022, 307, 120858. [Google Scholar] [CrossRef]
  325. Basque, A.; Touaibia, M.; Martin, L.J. Sinapic and ferulic acid phenethyl esters increase the expression of steroidogenic genes in MA-10 tumor Leydig cells. Toxicol. In Vitro 2023, 86, 105505. [Google Scholar] [CrossRef] [PubMed]
  326. Hu, X.; Geetha, R.V.; Surapaneni, K.M.; Veeraraghavan, V.P.; Chinnathambi, A.; Alahmadi, T.A.; Manikandan, V.; Manokaran, K. Lung cancer induced by Benzo(A)Pyrene: Chemo-protective effect of sinapic acid in swiss albino mice. Saudi J. Biol. Sci. 2021, 28, 7125–7133. [Google Scholar] [CrossRef]
  327. Altindag, F.; Ragetli, M.C.; Ozdek, U.; Koyun, N.; Alhalboosi, J.K.I.; Elasan, S. Combined treatment of sinapic acid and ellagic acid attenuates hyperglycemia in streptozotocin-induced diabetic rats. Food Chem. Toxicol. 2021, 156, 112443. [Google Scholar] [CrossRef]
  328. Eroglu, C.; Avci, E.; Vural, H.; Kurar, E. Anticancer mechanism of Sinapic acid in PC-3 and LNCaP human prostate cancer cell lines. Gene 2018, 671, 127–134. [Google Scholar] [CrossRef] [PubMed]
  329. Ansari, M.A.; Raish, M.; Ahmad, A.; Ahmad, S.F.; Mudassar, S.; Mohsin, K.; Shakeel, F.; Korashy, H.M.; Bakheet, S.A. Sinapic acid mitigates gentamicin-induced nephrotoxicity and associated oxidative/nitrosative stress, apoptosis, and inflammation in rats. Life Sci. 2016, 165, 1–8. [Google Scholar] [CrossRef] [PubMed]
  330. Raish, M.; Ahmad, A.; Ansari, M.A.; Ahad, A.; Al-Jenoobi, F.I.; Al-Mohizea, A.M.; Khan, A.; Ali, N. Sinapic acid ameliorates bleomycin-induced lung fibrosis in rats. Biomed. Pharmacother. 2018, 108, 224–231. [Google Scholar] [CrossRef]
  331. Singh, D.; Verma, R.K. Sinapic acid mitigates intracerebroventricular streptozotocin induced oxidative stress and neuro-inflammatory changes in rats. J. Neurol. Sci. 2019, 405, 109. [Google Scholar] [CrossRef]
  332. Yang, C.; Deng, Q.; Xu, J.; Wang, X.; Hu, C.; Tang, H.; Huang, F. Sinapic acid and resveratrol alleviate oxidative stress with modulation of gut microbiota in high-fat diet-fed rats. Food Res. Int. 2019, 116, 1202–1211. [Google Scholar] [CrossRef] [PubMed]
  333. Bezerra, J.J.L.; Pinheiro, A.A.V.; Barreto, E.D.O. Medicinal plants used in the treatment of asthma in different regions of Brazil: A comprehensive review of ethnomedicinal evidence, preclinical pharmacology and clinical trials. Phytomed. Plus 2022, 2, 100376. [Google Scholar] [CrossRef]
  334. Borah, B.; Dwivedi, K.D.; Kumar, B.; Chowhan, L.R. Recent advances in the microwave- and ultrasound-assisted green synthesis of coumarin-heterocycles. Arab. J. Chem. 2022, 15, 103654. [Google Scholar] [CrossRef]
  335. Alagesan, V.; Ramalingam, S.; Kim, M.; Venugopal, S. Antioxidant activity guided isolation of a coumarin compound from Ipomoea pes-caprea (Convolvulaceae) leaves acetone extract and its biological and molecular docking studies. Eur. J. Integr. Med. 2019, 32, 100984. [Google Scholar] [CrossRef]
  336. Cakmakci, E.; Ozdemir, M.; Sen, F.; Bulut, M.; Talcin, B. Vegetable oil-based, coumarin-containing antibacterial thermosets with improved thermal stability via copper-free thermal stability via copper-free thermal azide-alkyne click polymerization. Ind. Crop. Prod. 2022, 182, 114870. [Google Scholar] [CrossRef]
  337. Dandriyal, J.; Singla, R.; Kumar, M.; Jaitak, V. Recent developments of C-4 substituted coumarin derivatives as anticancer agents. Eur. J. Med. Chem. 2016, 119, 141–168. [Google Scholar] [CrossRef] [PubMed]
  338. Seo, W.D.; Kim, J.Y.; Ryu, H.W.; Kim, J.H.; Han, S.-I.; Ra, J.-E.; Seo, K.H.; Jang, K.C.; Lee, J.H. Identification and characterisation of coumarins from the roots of Angelica dahurica and their inhibitory effects against cholinesterase. J. Funct. Foods 2013, 5, 1421–1431. [Google Scholar] [CrossRef]
  339. Kassim, N.K.; Rahmani, M.; Ismail, A.; Sukari, M.A.; Ee, G.C.L.; Nasir, N.M.; Awang, K. Antioxidant activity-guided separation of coumarins and lignan from Melicope glabra (Rutaceae). Food Chem. 2013, 139, 87–92. [Google Scholar] [CrossRef] [PubMed]
  340. Sharma, M.; Vyas, V.K.; Bhatt, S.; Ghate, M.D. Therapeutic potential of 4-substitued coumarins: A conspectus. Eur. J. Med. Chem. Rep. 2022, 6, 100086. [Google Scholar] [CrossRef]
  341. Salau, V.F.; Erukainure, O.L.; Ibeji, C.U.; Koorbanally, N.A.; Islam, S. Umbelliferone stimulates glucose uptake; modulates gluconeogenic and nucleotide-hydrolyzing enzymes activities, and dysregulated lipid metabolic pathways in isolated psoas muscle. J. Funct. Food 2020, 67, 103847. [Google Scholar] [CrossRef]
  342. Seong, S.H.; Ali, Y.; Jung, H.A.; Choi, J.S. Umbelliferone derivatives exert neuroprotective effects by inhibiting monoamine oxidase A, self-amyloidβ aggregation, and lipid peroxidation. Bioorg. Chem. 2019, 92, 103293. [Google Scholar] [CrossRef]
  343. Mazimba, O. Umbelliferone: Sources, chemistry and bioactivities review. Bull. Fac. Pharm. Cairo Univ. 2017, 55, 223–232. [Google Scholar] [CrossRef]
  344. Kundu, M.; Chatterjee, S.; Ghosh, N.; Manna, P.; Das, J.; Sil, P.C. Tumor targeted delivery of umbelliferone via a smart mesoporous silica nanoparticles controlled-release drug delivery system for increased anticancer efficiency. Mater. Sci. Eng. C 2020, 116, 111239. [Google Scholar] [CrossRef]
  345. Pan, L.; Li, X.; Jin, H.; Yang, X.; Qin, B. Antifungal activity of umbelliferone derivatives: Synthesis and structure-activity relationships. Microb. Pathog. 2017, 104, 110–115. [Google Scholar] [CrossRef] [PubMed]
  346. Althunibat, O.Y.; Abduh, M.S.; Abukhalil, M.H.; Aladaileh, S.H.; Hanieh, H.; Mahmoud, A.M. Umbelliferone prevents isoproterenol-induced myocardial injury by upregulating Nrf2/HO-1 signaling, and attenuating oxidative stress, inflammation, and cell death in rats. Biomed. Pharmacother. 2022, 149, 112900. [Google Scholar] [CrossRef] [PubMed]
  347. Kutlu, Z.; Celik, M.; Bilen, A.; Halici, Z.; Yildirim, S.; Karabulut, S.; Karakaya, S.; Bostanlik, D.F.; Aydin, P. Effects of umbelliferone isolated from the Ferulago pauciradiata Boiss. & Heldr. plant on cecal ligation and puncture-induced sepsis model in rats. Biomed. Pharmacother. 2020, 127, 110206. [Google Scholar] [CrossRef] [PubMed]
  348. Lim, J.-Y.; Lee, J.-H.; Lee, D.-H.; Lee, J.-H.; Kim, D.-K. Umbelliferone reduces the expression of inflammatory chemokines in HaCaT cells and DNCB/DFE-induced atopic dermatitis symptoms in mice. Int. Immunopharmacol. 2019, 75, 105830. [Google Scholar] [CrossRef]
  349. Garud, M.S.; Kulkarni, Y.A. Attenuation of renal damage in type I diabetic rats by umbelliferone—A coumarin derivative. Pharmacol. Rep. 2017, 69, 1263–1269. [Google Scholar] [CrossRef]
  350. Mohamed, M.R.; Emam, M.A.; Hassan, N.S.; Mogadem, A.I. Umbelliferone and daphnetin ameliorate carbon tetrachloride-induced hepatotoxicity in rats via nuclear factor erythroid 2-related factor 2-mediated heme oxygenase-2 expression. Environ. Toxicol. Pharmacol. 2014, 38, 531–541. [Google Scholar] [CrossRef]
  351. Vijayalakshmi, A.; Sindhu, G. Umbelliferone arrest cell cycle at G0/G1 phase and induces apoptosis in human oral carcinoma (KB) cells possibly via oxidative DNA damage. Biomed. Pharmacother. 2017, 92, 661–671. [Google Scholar] [CrossRef]
  352. Cruz, L.F.; de Figueiredo, G.F.; Pedro, L.P.; Amorin, Y.M.; Andrade, J.T.; Passos, T.F.; Rodrigues, F.F.; Souza, I.L.A.; Goncalves, T.P.R.; Lima, L.A.R.D.S.; et al. Umbelliferone (7-hydroxycoumarin): A non-toxic antidiarrheal and antiulcerogenic coumarin. Biomed. Pharmacother. 2020, 129, 110432. [Google Scholar] [CrossRef]
  353. Park, C.; Jin, C.-Y.; Kim, G.-Y.; Choi, I.-W.; Kwon, T.K.; Choi, B.T.; Lee, S.J.; Lee, W.H.; Choi, Y.H. Induction of apoptosis by esculetin in human leukemia U937 cells through activation of JNK and ERK. Toxicol. Appl. Pharmacol. 2008, 227, 219–228. [Google Scholar] [CrossRef] [PubMed]
  354. Kadakol, A.; Sharma, N.; Kulkarni, Y.A.; Gaikwad, A.B. Esculetin: A phytochemical endeavor fortifying effect against non-communicable diseases. Biomed. Pharmacother. 2016, 84, 1442–1448. [Google Scholar] [CrossRef]
  355. Jeong, N.-H.; Yang, E.-J.; Jin, M.; Lee, J.Y.; Choi, Y.-A.; Park, P.-H.; Lee, S.-R.; Kim, S.-U.; Shin, T.-Y.; Kwon, T.K.; et al. Esculetin from Fraxinus rhynchopylla attenuates atopic skin inflammation by inhibiting the expression of inflammatory. Int. Immunopharmacol. 2018, 59, 209–216. [Google Scholar] [CrossRef]
  356. Prabakaran, D.; Ashokkumar, N. Antihyperglycemic effect of esculetin modulated carbohydrate metabolic enzymes activities in streptozotocin induced diabetic rats. J. Funct. Food 2012, 4, 776–783. [Google Scholar] [CrossRef]
  357. Prabakaran, D.; Ashokkumar, N. Protective effect of esculetin on hyperglycemia-mediated oxidative damage in the hepatic and renal tissues of experimental diabetic rats. Biochimie 2013, 95, 366–373. [Google Scholar] [CrossRef]
  358. Yang, J.; Han, J.; Li, Y.; Dong, B. Esculetin inhibits the apoptosis in H9c2 cardiomyocytes via the MAPK signaling pathway following hypoxia/reoxygenation injury. Biomed. Pharmacother. 2017, 88, 1206–1210. [Google Scholar] [CrossRef]
  359. Choi, R.-Y.; Ham, J.R.; Lee, M.-K. Esculetin prevents non-alcoholic fatty liver in diabetic mice fed high-fat diet. Chem. Biol. Interact. 2016, 260, 13–21. [Google Scholar] [CrossRef]
  360. Subramaniam, S.R.; Ellis, E.M. Esculetin-induced protection of human hepatoma HepG2 cells against hydrogen peroxide is associated with the Nrf2-dependent induction of the NAD(P)H: Quinone oxidoreductase 1 gene. Toxicol. Appl. Pharmacol. 2011, 250, 130–136. [Google Scholar] [CrossRef]
  361. Kadakol, A.; Malek, V.; Goru, S.K.; Pandey, A.; Sharma, N.; Gaikwad, A.B. Esculetin ameliorates insulin resistance and type 2 diabetic nephropathy through reversal of histone H3 acetylation and H2A lysine 119 monoubiquitination. J. Funct. Food 2017, 35, 256–266. [Google Scholar] [CrossRef]
  362. Kim, W.K.; Byun, W.S.; Chung, H.-J.; Oh, J.; Park, H.J.; Choi, J.S.; Lee, S.K. Esculetin suppresses tumor growth and metastasis by targeting Axin2/E-cadherin axis in colorectal cancer. Biochem. Pharmacol. 2018, 152, 71–83. [Google Scholar] [CrossRef]
  363. Xu, B.; Zhu, L.; Chu, J.; Ma, Z.; Fu, Q.; Wei, W.; Deng, X.; Ma, S. Esculetin improves cognitive impairments induced by transient cerebral ischaemia and reperfusion in mice via regulation of mitochondrial fragmentation and mitophagy. Behav. Brain Res. 2019, 372, 112007. [Google Scholar] [CrossRef] [PubMed]
  364. Zhang, M.; Damba, T.; Wu, Z.; Serna-Salas, S.; Buist-Homan, M.; Faber, K.N.; Moshage, H. Bioactive coumarin-derivative esculetin decreases hepatic stellate cell activation via induction of cellular senescence via the PI3K-Akt-GSK3β pathway. Food Biosci. 2022, 50, 102164. [Google Scholar] [CrossRef]
  365. Wang, J.; Lai, X.; Yuan, D.; Liu, Y.; Wang, J.; Liang, Y. Effects of ferulic acid, a major component of rice bran, on proliferation, apoptosis, and autophagy of HepG2 cells. Food Res. Int. 2022, 161, 111816. [Google Scholar] [CrossRef] [PubMed]
  366. Ren, W.; Zhou, Q.; Yu, R.; Liu, Z.; Hu, Y. Esculetin inhibits the pyroptosis of microvascular endothelial cells through NF-KB/NLRP3 signaling pathway. Arch. Biochem. Biophys. 2022, 720, 109173. [Google Scholar] [CrossRef] [PubMed]
  367. Jiang, Y.; Fang, Z.; Leonard, W.; Zhang, P. Phenolic compounds in Lycium berry: Composition, health benefits and industrial applications. J. Funct. Food 2021, 77, 104340. [Google Scholar] [CrossRef]
  368. Duan, J.; Shi, J.; Ma, X.; Xuan, Y.; Li, P.; Wang, H.; Fan, Y.; Gong, H.; Wang, L.; Pang, Y.; et al. Esculetin inhibits proliferation, migration, and invasion of clear cell renal cell carcinoma cells. Biomed. Pharmacother. 2020, 125, 110031. [Google Scholar] [CrossRef] [PubMed]
  369. Lee, J.; Yang, J.; Jeon, J.; Jeong, H.S.; Lee, J.; Sung, J. Hepatoprotective effect of esculetin on ethanol-induced liver injury in hman HepG2 cells and C57BL/6J mice. J. Funct. Food 2018, 40, 536–543. [Google Scholar] [CrossRef]
  370. Dou, Y.; Tong, B.; Wei, Z.; Li, Y.; Xia, Y.; Dai, Y. Scopoletin suppresses IL-6 production from fibroblast-like synoviocytes of adjuvant arthritis rats induced by IL-1β stimulation. Int. Immunopharmacol. 2013, 17, 1037–1043. [Google Scholar] [CrossRef]
  371. Connell, B.J.; Saleh, M.C.; Rajagopal, D.; Saleh, T.M. UPEI-400, a conjugate of lipoic acid and scopoletin, mediates neuroprotection in a rat model of ischemia/reperfusion. Food Chem. Toxicol. 2017, 100, 175–182. [Google Scholar] [CrossRef]
  372. Rutul, R.; Amar, S.; Mithil, P.; Sushil, K.; Singh, A.; Parth, D.; Ghanshyam, P.; Mistri, J.; Subhash, N. Study of dynamics of genes involved in biosynthesis and accumulation of scopoletin at different growth stages of Convolvulus prostratus Forssk. Phytochemistry 2021, 182, 112594. [Google Scholar] [CrossRef] [PubMed]
  373. Tasfiyati, A.N.; Antika, L.D.; Dwei, R.T.; Spetama, A.W.; Sabarudin, A.; Ernawati, T. An experimental design approach for the optimization of scopoletin extraction from Morinda citrifolia L. using accelerated solvent extraction. Talanta 2022, 238, 123010. [Google Scholar] [CrossRef] [PubMed]
  374. Alkorashy, A.I.; Doghish, A.S.; Abulsoud, A.I.; Ewees, M.G.; Abdelghany, T.M.; Elshafey, M.M.; Elkhatib, W.F. Effect of scopoletin on phagocytic activity of U937-derived human macrophages: Insights from transcriptomic analysis. Genomics 2020, 112, 3518–3524. [Google Scholar] [CrossRef] [PubMed]
  375. Khalaf, M.M.; Hassan, S.M.; Sayed, A.M.; Abo-Youssef, A.M. Ameliorate impacts of scopoletin against vancomycin-induced intoxication in rat model through modulation of Keap1-Nrf2/HO-1 and IKBα-P65 NF-KB/P38 MAPK signaling pathways: Molecular study, molecular docking evidence and network pharmacology analysis. Int. Immunophrmacol. 2022, 102, 108382. [Google Scholar] [CrossRef] [PubMed]
  376. Ham, J.R.; Lee, H.-I.; Choi, R.-Y.; Sim, M.-O.; Choi, M.-S.; Kwon, E.-Y.; Yun, K.W.; Kim, M.-J.; Lee, M.-K. Anti-obesity and anti-hepatosteatosis effects of dietary scopoletin in high-fat diet fed mice. J. Funct. Food 2016, 25, 433–446. [Google Scholar] [CrossRef]
  377. Nam, H.; Kim, M.-M. Scopoletin has a potential activity for anti-aging via autophagy in human lung fibroblasts. Phytomedicine 2015, 22, 362–368. [Google Scholar] [CrossRef]
  378. Oyama, T.; Yoshimori, A.; Ogawa, H.; Shirai, Y.; Abe, H.; Kamiya, T.; Tanuma, S.-I. The structural differences between mushroom and human tyrosinase cleared by investigating the inhibitory activities of stilbenes. J. Mol. Struct. 2023, 1272, 134180. [Google Scholar] [CrossRef]
  379. Avendano-Godoy, J.; Ortega, E.; Urrutia, M.; Escobar-Avello, D.; Luengo, J.; Baer, D.V.; Mardones, C.; Gomez-Gaete, C. Prototype of nutraceutical products from microparticles loaded with stilbenes extracted from grape cane. Food Bioprod. Process. 2022, 134, 19–29. [Google Scholar] [CrossRef]
  380. Fu, Y.-H.; Hou, Y.-D.; Duan, Y.-Z.; Sun, X.-Y.; Chen, S.-Q. Six undescrived derivatives of stilbene isolated from Lindera reflexa hemsl. And their anti-tumor and anti-inflammatory activities. Fitoterapia 2022, 163, 105331. [Google Scholar] [CrossRef]
  381. Choi, Y.S.; Yoon, D.H.; Kim, S.Y.; Kim, C.S.; Lee, K.R. Stilbene oligomers from the stems of Parthenocissus tricuspidata and their potential anti-neuroinflammatory and neuroprotective activity. Tetrahedron Lett. 2021, 71, 153027. [Google Scholar] [CrossRef]
  382. Brianceau, S.; Turk, M.; Vitrac, X.; Vorobiev, E. High voltage electric discharges assisted extraction of phenolic compounds from grape stems: Effect of processing parameters on flavan-3-ols, flavonols and stilbenes recovery. Innov. food Sci. Emerg. Technol. 2016, 35, 67–74. [Google Scholar] [CrossRef]
  383. Robb, E.L.; Stuart, J.A. The stilbenes resveratrol, pterostilbene and piceid affect growth and stress resistance in mammalian cells via a mechanism requiring estrogen receptor beta and the induction of Mn-superoxide dismutase. Phytochemistry 2014, 98, 164–173. [Google Scholar] [CrossRef] [PubMed]
  384. Fernandez-Marin, M.I.; Guerrero, R.F.; Garcia-Parrilla, M.C.; Puertas, B.; Richard, T.; Rodriguez-Werner, M.A.; Winterhalter, P.; Monti, J.-P.; Cantos-Villar, E. Isorhapontigenin: A novel bioactive stilbene from wine grapes. Food Chem. 2012, 135, 1353–1359. [Google Scholar] [CrossRef] [PubMed]
  385. Benbouguerra, N.; Hornedo-Ortega, R.; Garcia, F.; Khawand, T.E.; Saucier, C.; Richard, T. Stilbenes in grape berries and wine and their potential role as anti-obesity agents: A review. Trends Food Sci. Technol. 2021, 112, 362–381. [Google Scholar] [CrossRef]
  386. Guerrero-Solao, J.; Jaramillo-Morales, O.A.; Jimenez-Cabrera, T.; Urrutia-Hernandez, T.A.; Chehue-Romero, A.; Olvera-Hernandez, E.G.; Bautista, M. Punica protopunica Balf., the forgotten sister of the common pomegranate (Punica granatum L.): Features and medicinal properties—A review. Plants 2020, 9, 1214. [Google Scholar] [CrossRef] [PubMed]
  387. Runeberg, P.; Ryabukhin, D.; Lagerquist, L.; Rahkila, J.; Eklund, P. Transformations and antioxidative activities of lignans and stilbenes at high temperatures. Food Chem. 2023, 4040, 134641. [Google Scholar] [CrossRef] [PubMed]
  388. Thriumalaisamy, R.; Bhuvaneswari, M.; Haritha, S.; Jeevarathna, S.; Janani, K.S.S.; Suresh, K. Curcumin, Naringenin and resveratrol from natural plant products hold promising solutions for modern world diseases—A recent review. S. Afr. J. Bot. 2022, 151, 567–580. [Google Scholar] [CrossRef]
  389. Pinilla-Penalver, E.; Garcia-Bejar, B.; Contento, A.M.; Rios, A. Graphene quantum dots an efficient nanomaterial for enhancing the photostability of trans-resveratrol in food samples. Food Chem. 2022, 386, 132766. [Google Scholar] [CrossRef]
  390. Pirola, L.; Frojdo, S. Resveratrol: One molecule, many targets IUBMB. Life 2008, 60, 323–332. [Google Scholar] [CrossRef]
  391. Zupancic, S.; Lavric, Z.; Kristl, J. Stability and solubility of trans-resveratrol are strongly influenced by pH and temperature. Eur. J. Pharm. Biopharm. 2015, 93, 196–204. [Google Scholar] [CrossRef] [PubMed]
  392. Vian, M.A.; Tomao, V.; Gallet, S.; Coulomb, P.O.; Lacombe, J.M. Simple and rapid method for cis- and trans-resveratrol and piceid isomers determination in wine by high-performance liquid chromatography using chromolith columns. J. Chromatogr. A 2005, 1085, 224–229. [Google Scholar] [CrossRef]
  393. Shen, L.; Ji, H.-F. Reciporocal interactions between resveratrol and gut microbiota deepen our understanding of molecular mechanisms underlying its health benefits. Trends Food Sci. Technol. 2018, 81, 232–236. [Google Scholar] [CrossRef]
  394. Nicolau, A.L.A.; Peres, G.B.; Silva, J.D.S.; Nunes, S.H.; Fortes, T.M.L.; Suffredini, I.B. Pilot project. Resveratrol intake by physical active and sedentary older adult women and blood pressure. Exp. Gerontol. 2022, 166, 111883. [Google Scholar] [CrossRef]
  395. Marques, F.Z.; Markus, M.A.; Morris, B.J. Resveratrol: Cellular actions of a potent natural chemical that confers a diversity of health benefits. Int. J. Biochem. Cell Biol. 2009, 41, 2125–2128. [Google Scholar] [CrossRef] [PubMed]
  396. Shoura, S.M.S.; Naghsh, N.; Moslemi, E.; Kayvani, Z.; Moridpour, A.H.; Musazadeh, V.; Dehghan, P. Can resveratrol supplementation affect biomarkers of inflammation and oxidative stress? An umbrella meta-analysis. J. Funct. Food 2022, 99, 105360. [Google Scholar] [CrossRef]
  397. Guo, S.; Zhou, Y.; Xie, X. Resveratrol inhibiting TGF/ERK signaling pathway can improve atherosclerosis: Backgrounds, mechanisms and effects. Biomed. Pharmacother. 2022, 155, 113775. [Google Scholar] [CrossRef]
  398. Grujic-Milanovic, J.; Jacevic, V.; Miloradovic, Z.; Milanovic, S.D.; Jovovic, D.; Ivanov, M.; Karanovic, D.; Vajic, U.-J.; Mihailovic-Stanojevic, N. Resveratrol improved kidney function and structure in malignantly hypertensive rats by restoration of antioxidant capacity and nitric oxide bioavailability. Biomed. Pharmacother. 2022, 154, 113642. [Google Scholar] [CrossRef]
  399. Nath, J.; Roy, R.; Sathyamoorthy, Y.K.; Paul, S.; Goswami, S.; Chakravarty, H.; Paul, R.; Borah, A. Resveratrol as a therapeutic choice for traumatic brain injury: An insight into its molecular mechanism of action. Brain Disord. 2022, 6, 100038. [Google Scholar] [CrossRef]
  400. Chowdhury, F.I.; Yasmin, T.; Akter, R.; Islam, N.; Hossain, M.M.; Khan, F.; Aldhahrani, A.; Soliman, M.M.; Subhan, N.; Haque, A.; et al. Resveratrol treatment modulates several antioxidant and anti-inflammatory genes expression and ameliorated oxidative stress mediated fibrosis in the kidneys of high-fat diet-fed rats. Saudi Pharm. J. 2022, 30, 1454–1463. [Google Scholar] [CrossRef]
  401. Van Brummelen, R.; Brummelen, A.C.V. The potential role of resveratrol as supportive antiviral in treating conditions such as COVID-19—A formulator’s perspective. Biomed. Pharmacother. 2022, 148, 112767. [Google Scholar] [CrossRef]
  402. Kotta, S.; Aldawsari, H.M.; Badr-Eldin, S.M.; Alhakamy, N.A.; Shadab, M. Coconut oil-based resveratrol nanoemulsion: Optimization using response surface methodology, stability assessment and pharmacokinetic evaluation. Food Chem. 2021, 357, 129721. [Google Scholar] [CrossRef]
  403. Monahan, D.S.; Flaherty, E.; Hameed, A.; Duffy, G.P. Resveratrol significantly improves cell survival in comparison to dexrazoxane and carvedilol in a h9c2 model of doxorubicin induced cardiotoxicity. Biomed. Pharmacother. 2021, 140, 111702. [Google Scholar] [CrossRef]
  404. Dai, M.; Yuan, D.; Lei, Y.; Li, J.; Ren, Y.; Zhang, Y.; Cang, H.; Gao, W.; Tang, Y. Expression, purification and structural characterization of resveratrol synthase from Polygonum cuspidatum. Protein Expr. Purif. 2022, 191, 106024. [Google Scholar] [CrossRef] [PubMed]
  405. Catalogna, G.; Moraca, F.; DAntona, L.; Dattilo, V.; Perrotti, G.; Lupia, A.; Costa, G.; Ortuso, F.; Iuliano, R.; Trapasso, F.; et al. Review about the multi-target profile of resveratrol and its implication in the SGK1 inhibition. Eur. J. Med. Chem. 2019, 183, 111675. [Google Scholar] [CrossRef]
  406. Bhaskara, V.K.; Mittal, B.; Mysorekar, V.V.; Amaresh, N.; Simal-Gandara, J. Resveratrol, cancer and cancer stem cells: A review on past to future. Curr. Res. Food Sci. 2020, 3, 284–295. [Google Scholar] [CrossRef]
  407. Chu, S.-H.; Yang, D.; Wang, Y.-P.; Yang, R.; Qu, L.; Zeng, H.-J. Effect of resveratrol on their repair kidney and brain injuries and its regulation on klotho gene in D-galactose-induced aging mice. Bioorg. Med. Chem. Lett. 2021, 40, 127913. [Google Scholar] [CrossRef] [PubMed]
  408. Huang, H.; Liao, D.; Zhou, G.; Zhu, Z.; Cui, Y.; Pu, R. Antiviral activities of resveratrol against rotavirus in vitro and in vivo. Phytomedicine 2020, 77, 153230. [Google Scholar] [CrossRef] [PubMed]
  409. Liu, S.; Li, Y.; Yi, F.; Liu, Q.; Chen, N.; He, X.; He, C.; Xiao, P. Resveratrol oligomers from Paeonia suffruticosa protect mice against cognitive dysfunction by regulation cholinergic, antioxidant and anti-inflammatory pathways. J. Ethnopharmacol. 2020, 260, 112983. [Google Scholar] [CrossRef]
  410. Fantacuzzi, M.; Amoroso, R.; Carradori, S.; Filippis, B.D. Resveratrol-based compounds and neurodegenration: Recent insight in multitarget therapy. Eur. J. Med. Chem. 2022, 233, 114242. [Google Scholar] [CrossRef]
  411. Vestergaard, M.; Ingmer, H. Antibacterial and antifungal properties of resveratrol. Int. J. Antimicrob. Agents 2019, 53, 716–723. [Google Scholar] [CrossRef]
  412. Roshani, M.; Jafari, A.; Loghman, A.-H.; Sheida, A.H.; Taghavi, T.; Tamehri Zadeh, S.S.; Hamblin, M.R.; Homayounfal, M.; Mirzaei, H. Application of resveratrol in the treatment of gastrointestinal cancer. Biomed. Pharmacother. 2022, 153, 113274. [Google Scholar] [CrossRef] [PubMed]
  413. Agah, S.; Akbari, A.; Sadeghi, E.; Morvaridzadeh, M.; Basharat, Z.; Palmowski, A.; Heshmati, J. Resveratrol supplementation and acute pancreatitis: A comprehensive review. Biomed. Pharmacother. 2021, 137, 111268. [Google Scholar] [CrossRef]
  414. Algandaby, M.M.; Al-Sawahli, M.M. Augmentation of anti-proliferative, pro-apoptotic and oxidant profiles induced by piceatannol in human breast carcinoma MCF-7 cells using zein nanostructures. Biomed. Pharmacother. 2021, 138, 111409. [Google Scholar] [CrossRef]
  415. Takei, M.; Umeyama, A.; Arihara, S.; Matsumoto, H. Effect of piceatannol in human monocyte-derived dendritic cells in vitro. J. Pharm. Sci. 2005, 94, 974–982. [Google Scholar] [CrossRef] [PubMed]
  416. Kee, H.J.; Park, S.; Kang, W.; Lim, K.S.; Kim, J.H.; Ahn, Y.; Jeong, M.H. Piceatannol attenuates cardiac hypertrophy in an animal model through regulation of the expression and binding of the transcription factor GATA binding factor 6. FEBS Lett. 2014, 588, 1529–1536. [Google Scholar] [CrossRef] [Green Version]
  417. Geahlen, R.; McLaughlin, J.L. Piceatannol (3,4,3′,5′-tetrahydroxy-trans-stilbene) is a naturally occuring protein-tyroisne kinase inhibitor. Biochem. Biophys. Res. Commun. 1989, 164, 241–245. [Google Scholar] [CrossRef]
  418. Messiad, H.; Amira-Guebailia, H.; Houache, O. Reversed phase high performance liquid chromatography used for the physicochemical and thermodynamic characterization of piceatannol/β-cyclodextrin complex. J. Chromatogr. B 2013, 926, 21–27. [Google Scholar] [CrossRef]
  419. Wang, D.; Zhang, Y.; Zhang, C.; Gao, L.; Li, J. Piceatannol pretreatment alleviates acute cardiac injury via regulating PI3K-Akt-eNOS signaling in H9c2 cells. Biomed. Pharmacother. 2019, 109, 886–891. [Google Scholar] [CrossRef] [PubMed]
  420. Tieyuan, Z.; Ying, Z.; Xinghua, Z.; Huimin, W.; Huagang, L. Piceatannol-mediated JAK2/STAT3 signaling pathway inhibition contributes to the alleviation of oxidative injury and collagen synthesis during pulmonary fibrosis. Int. Immunopharmacol. 2022, 111, 109107. [Google Scholar] [CrossRef] [PubMed]
  421. Kim, D.-H.; Lee, Y.-G.; Park, H.-J.; Lee, J.-A.; Kim, H.J.; Hwang, J.-K.; Choi, J.-M. Piceatannol inhibits effector T cell functions by suppressing TcR signaling. Int. Immunopharmacol. 2015, 25, 285–292. [Google Scholar] [CrossRef]
  422. Yang, J.S.; Tongson, J.; Kim, K.-H.; Park, Y. Piceatannol attenuates fat accumulation and oxidative stress in steatosis-induced HepG2 cells. Curr. Res. Food Sci. 2020, 3, 92–99. [Google Scholar] [CrossRef] [PubMed]
  423. Gerszon, J.; Wojtala, M.; Michlewska, S.; Rodacka, A. Piceatannol effectively counteracts glyceraldehyde-3-phosphate dehydrogenase aggregation and nuclear translocation in hippocampal cells. J. Funct. Food 2019, 58, 180–188. [Google Scholar] [CrossRef]
  424. Siedleck-Kroplewska, K.; Slebioda, T.; Kmiec, Z. Induction of autophagy, apoptosis and acquisition of resistance in response to piceatannol toxicity in MOLT-4 human leukemia cells. Toxicol In Vitro 2019, 59, 12–25. [Google Scholar] [CrossRef]
  425. Yamamoto, T.; Sato, A.; Takai, Y.; Yoshimori, A.; Umehara, M.; Ogino, Y.; Inada, M.; Shimada, N.; Nishida, A.; Ichida, R.; et al. Effect of piceatannol-rich passion fruit seed extract on human glyoxalase I-mediated cancer cell growth. Biochem. Biophys. Rep. 2019, 20, 100684. [Google Scholar] [CrossRef] [PubMed]
  426. Wen, J.; Lin, H.; Zhao, M.; Tao, L.; Yang, Y.; Xu, X.; Jia, A.; Zhang, J.; Weng, D. Piceatannol attenuates D-Ga1N/LPS-induced hepatoxicity in mice: Involvement of ER stress, inflammation and oxidative stress. Int. Immunopharmacol. 2018, 64, 131–139. [Google Scholar] [CrossRef]
  427. Takasawa, R.; Akahane, H.; Tanaka, H.; Shimada, N.; Yamamoto, T.; Uchida-Maruki, H.; Sai, M.; Yoshimori, A.; Tanuma, S.-I. Piceatannol, a natural trans-stilbene compound, inhibits human glyoxalase I. Bioorg. Med. Chem. Lett. 2017, 27, 1169–1174. [Google Scholar] [CrossRef] [PubMed]
  428. Lu, B.; Sun, T.; Li, W.; Sun, X.; Yao, X.; Sun, X. Piceatannol protects ARPE-19 cells against vitamin A dimer-mediated photo-oxidative damage through activation of Nrf2/NQO1 signaling. J. Funct. Food 2016, 26, 739–749. [Google Scholar] [CrossRef]
  429. Farrand, L.; Byun, S.; Kim, J.Y.; Im-Aram, A.; Lee, J.; Lim, S.; Lee, K.W.; Suh, J.-Y.; Lee, H.J.; Tsang, B.K. Piceatannol enhances cisplatin sensitivity in ovarian cancer via modulation of p53, X-linked inhibitor of apoptosis protein (XIAP), and mitochondrial fission. J. Biol. Chem. 2013, 288, 23740–23750. [Google Scholar] [CrossRef] [Green Version]
  430. Xu, X.; Zhao, M.; Han, Q.; Wang, H.; Zhang, H.; Wang, Y. Effects of piceatannol on the structure and activities of bovine serum albumin: A multi-spectral and molecular modeling studies. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2020, 228, 117706. [Google Scholar] [CrossRef]
  431. Ren, G.; Rimando, A.M.; Mathews, S.T. AMPK activation by pterostilbene contributes to suppression of hepatic gluconeogenic gene expression and glucose production in H4IIE cells. Biochem. Biophys. Res. Commun. 2018, 498, 640–645. [Google Scholar] [CrossRef] [PubMed]
  432. Rodriguez-Bonilla, P.; Lopez-Nicolas, J.M.; Mendez-Cazorla, L.; Garcia-Carmona, F. Development of a reversed phase high performance liquid chromatography method based on the use of cyclodextrins as mobile phase additives to determine pterostilbene in blueberries. J. Chromatogr. B 2011, 879, 1091–1097. [Google Scholar] [CrossRef]
  433. Azzolini, M.; Mattarei, A.; Spina, M.L.; Fanin, M.; Chiodarelli, G.; Romio, M.; Zoratti, M.; Paradisi, C.; Biasutto, L. New natural amino acid-bearing prodrugs boost pterostilbene’s oral pharmacokinetic and distribution profile. Eur. J. Pharm. Biopharm. 2017, 115, 149–158. [Google Scholar] [CrossRef]
  434. Xu, D.; Qiao, F.; Xi, P.; Lin, Z.; Jiang, Z.; Romanazzi, G.; Gao, L. Efficacy of pterostilbene suppression of postharvest gray mold in table grapes and potential mechanisms. Postharvest Biol. Technol. 2022, 183, 111745. [Google Scholar] [CrossRef]
  435. Chen, R.-J.; Lee, Y.-H.; Yeh, Y.-L.; Wu, W.-S.; Ho, C.-T.; Li, C.-Y.; Wang, B.-J.; Wang, Y.-J. Autophagy-inducing effect of pterostilbene: A prospective therapeutic/preventive option for skin diseases. J. Food Drug Anal. 2017, 25, 125–133. [Google Scholar] [CrossRef]
  436. Zhang, H.; Chen, Y.; Chen, Y.; Ji, S.; Jia, P.; Xu, J.; Li, Y.; Wang, T. Pterostilbene attenuates liver injury and oxidative stress in intrauterine growth-retarded weanling piglets. Nutrition 2021, 81, 110940. [Google Scholar] [CrossRef] [PubMed]
  437. Ma, K.; Chen, H.; Wang, K.; Han, X.; Zhang, Y.; Wang, H.; Hu, Z.; Wang, J. Pterostilbene inhibits the metastasis of TNBC via suppression of β-catenin-mediated epithelial to mesenchymal transition and stemness. J. Funct. Food 2022, 96, 105219. [Google Scholar] [CrossRef]
  438. Zalesak, F.; Bon, D.J.-Y.D.; Pospisil, J. Lignans and neolignans: Plant secondary metabolites as a reservoir of biologically active substances. Pharmacol. Res. 2019, 146, 104284. [Google Scholar] [CrossRef]
  439. Su, J.-H.; Wang, M.-Q.; Zhu, L.-P.; Yang, W.-Q.; Jiang, S.-Q.; Zhao, Z.-X.; Sun, Z.-H. Coumarins and lignans from the roots of Zanthoxylum nitidum. Biochem. Syst. Ecol. 2022, 101, 104399. [Google Scholar] [CrossRef]
  440. Samari, E.; Chashmi, N.A.; Ghanati, F.; Sajedi, R.H.; Gust, A.A.; Haghdoust, F.; Sharifi, M.; Fuss, E. Interactions between second messengers, SA and MAPK6 signaling pathways lead to chitosan-induced lignan production in Linum album cell culture. Ind. Crop. Prod. 2022, 177, 114525. [Google Scholar] [CrossRef]
  441. Tannous, S.; Haykal, T.; Dhaini, J.; Hodroj, M.H.; Rizk, S. The anti-cancer effect of flaxseed lignan derivatives on different acute myeloid leukemia cancer cells. Biomed. Pharmacother. 2020, 132, 110884. [Google Scholar] [CrossRef]
  442. Torres-Moreno, H.; Lopez-Romero, J.C.; Vidal-Gutierrez, M.; Rodriguez-Martinez, K.; Robles-Zepeda, R.E.; Vilegas, W.; Velarde-Rodrigez, G.M. Seasonality impact on the anti-inflammatory, antiproliferative potential and the lignan composition of Bursera microphylla. Ind. Crop. Prod. 2022, 184, 115095. [Google Scholar] [CrossRef]
  443. Kalinova, J.P.; Maresova, I.; Triska, J.; Vrchotova, N. Distribution of lignans in Panicum miliaceum, Fagopyrum esculentum, Fagopyrum tataricum, and Amaranthus hypochondriacus. J. Food Compos. Anal. 2022, 106, 104283. [Google Scholar] [CrossRef]
  444. Rangkadilok, N.; Pholphana, N.; Mahidol, C.; Wongyai, W.; Saengsooksree, K.; Nookabkaew, S.; Satayavivad, J. Variation of sesamin, sesamolin, and tocopherols in sesame (Sesamum indicum L.) seeds and oil products in Thailand. Food Chem. 2010, 122, 724–730. [Google Scholar] [CrossRef]
  445. Anju, V.T.; Busi, S.; Ranganathan, S.; Ampasala, D.R.; Kumar, S.; Suchiang, K.; Kumavath, R.; Dyavaiah, M. Sesamin and sesamolin rescues Caenorhabditis elegans from pseudomonas aeruginosa infection through the attenuation of quorum sensing regulated virulence factors. Microb. Pathog. 2021, 155, 104912. [Google Scholar] [CrossRef] [PubMed]
  446. Dong, Z.-Y.; Wei, L.; Lu, H.-Q.; Zeng, Q.-H.; Meng, F.-C.; Wang, G.-W.; Lan, X.-Z.; Liao, Z.-H.; Chen, M. Ptechoosines A and B: Two new sesamin-type sesquilignans with antiangiogenic activity from Pterocephalus hookeri (C.B. Clarke) Hoeck. Fitoterapia 2021, 151, 104886. [Google Scholar] [CrossRef]
  447. Dalibalta, S.; Majdalawieh, A.F.; Manjikian, H. Health benefits of sesamin on cardiovascular disease and its associated risk factors. Saudi Pharm. J. 2020, 28, 1276–1289. [Google Scholar] [CrossRef]
  448. Hata, N.; Hayashi, Y.; Okazawa, A.; Ono, E.; Satake, H.; Kobayashi, A. Comparison of sesamin contents and CYP81Q1 gene expressions in aboveground vegetative organs between two Japanese sesame (Sesamum indicum L.) varieties differing in seed sesamin contents. Plant Sci. 2010, 178, 510–516. [Google Scholar] [CrossRef]
  449. Dossou, S.S.K.; Xu, F.-T.; Dossa, K.; Zhou, R.; Zhao, Y.-Z.; Wang, L.-H. Antioxidant lignans sesamin and sesamolin in sesame (Sesamum indicum L.): A comprehensive review and future prospects. J. Integr. Agric 2022. [Google Scholar] [CrossRef]
  450. Wang, M.; Liu, P.; Kong, L.; Xu, N.; Lei, H. Promotive effects of sesamin on proliferation and adhesion of intestinal probiotics and its mechanism of action. Food Chem. Toxicol. 2021, 149, 112049. [Google Scholar] [CrossRef] [PubMed]
  451. Kongtawelert, P.; Wudtiwai, B.; Shwe, T.H.; Pothacharoen, P.; Phitak, T. Inhibition of programmed death ligand 1 (PD-L1) expression in breast cancer cells by sesamin. Int. Immunopharmacol. 2020, 86, 106759. [Google Scholar] [CrossRef]
  452. Wanachewin, O.; Klangjorhor, J.; Pothacharoen, P.; Phitak, T.; Laohapoonrungsee, A.; Pruksakorn, D.; Kongtawelert, P. The promoting effects of sesamin on osteoblast differentiation of human mesenchumal stem cells. J. Funct. Foods. 2015, 14, 395–406. [Google Scholar] [CrossRef]
  453. Rashno, M.; Sarkaki, A.; Ghaderi, S.; Khoshnam, S.E. Sesamin: Insights into its protective effects against lead-induced learning and memory deficits in rats. J. Trace Elem. Med. Biol. 2022, 72, 126993. [Google Scholar] [CrossRef]
  454. Ye, H.; Sun, L.; Li, J.; Wang, Y.; Bai, J.; Wu, L.; Han, Q.; Yang, Z.; Li, L. Sesamin attenuates carrageenan-induced lung inflammation through upregulation of A20 and TAX1BP1 in rats. Int. Immunopharmacol. 2020, 88, 107009. [Google Scholar] [CrossRef] [PubMed]
  455. Majdalawieh, A.F.; Massri, M.; Nasrallah, G.K. A comprehensive review on the anti-cancer properties and mechanisms of action of sesamin, a lignan in sesame seeds (Sesamum indicium). Eur. J. Pharmacol. 2017, 815, 512–521. [Google Scholar] [CrossRef] [PubMed]
  456. Ghaderi, S.; Rashno, M.; Nesari, A.; Khoshnam, S.E.; Sarkaki, A.; Khorsandi, L.; Farbood, Y.; Rashidi, K. Sesamin alleviates diabetes-associated behavioral deficits in rats: The role of inflammatory and neurotrophic factors. Int. Immunopharmacol. 2021, 92, 107356. [Google Scholar] [CrossRef]
  457. Phitak, T.; Potacharoen, P.; Settakorn, J.; Poompimol, W.; Caterson, B.; Kongtawelert, P. Chondroprotective and anti-inflammatory effects of sesamin. Phytochemistry 2012, 80, 77–88. [Google Scholar] [CrossRef] [PubMed]
  458. Ju, Y.-I.; Yang, L.; Yue, X.-F.; He, R.; Deng, S.-L.; Yang, X.; Liu, X.; Fang, Y.-L. The condensed tannin chemistry and astringency properties of fifteen Vitis davidii Foex grapes and wines. Food Chem. 2021, 11, 100125. [Google Scholar] [CrossRef]
  459. Han, S.; Kong, X.; Xia, S.; Jia, B.; Dong, M.; Gan, L. UV light-driven controlled photodegradation of condensed tannins from larch bark. Ind. Crop. Prod. 2022, 177, 114403. [Google Scholar] [CrossRef]
  460. Abhijit, S.; Tripathi, S.J.; Rao, B.S.S.; Devi, S.A. Grape seed proanthocyanidin extract and swimming training enhances neuronal number in dorso-medial prefrontal cortex in middle-aged male rats by alleviating oxidative stress. J. Funct. Food 2020, 64, 103693. [Google Scholar] [CrossRef]
  461. Sangiovanni, E.; Piazza, S.; Vrhovsek, U.; Fumagalli, M.; Khalilpour, S.; Masuero, D.; Lorenzo, C.D.; Colombo, L.; Mattivi, F.; Fabiani, E.D.; et al. A bio-guided approach for the development of a chestnut-based proanthocyanidin-enriched nutraceutical with potential anti-gastritis properties. Pharmacol. Res. 2018, 134, 145–155. [Google Scholar] [CrossRef]
  462. Yu, G.-H.; Fang, Y. Resveratrol attenuates atherosclerotic endothelial injury through the Pin1/Notch1 pathway. Toxicol. Appl. Pharmacol. 2022, 446, 116047. [Google Scholar] [CrossRef]
  463. Tao, W.; Pan, H.; Jiang, H.; Wang, M.; Ye, X.; Chen, S. Extraction and identification of proanthocyanidins from the leaves of persimmon and loquat. Food Chem. 2022, 372, 130780. [Google Scholar] [CrossRef] [PubMed]
  464. Zhang, X.; Song, X.; Hu, X.; Chen, F.; Ma, C. Health benefits of proanthocyanidins linking with gastrointestinal modulation: An updated review. Food Chem. 2023, 4040, 134596. [Google Scholar] [CrossRef]
  465. Yang, J.; Chen, J.; Hao, Y.; Liu, Y. Identification of the DPPH radical scavenging reaction adducts of ferulic acid and sinapic acid and their structure-antioxidant activity relationship. LWT 2021, 146, 111411. [Google Scholar] [CrossRef]
  466. Rauf, A.; Imran, M.; Abu-Izneid, T.; Ul-Haq, I.; Patel, S.; Pan, X.; Naz, S.; Silva, A.S.; Saeed, F.; Suleria, H.A.R. Proanthocyanidins: A comprehensive review. Biomed. Pharmacother. 2019, 116, 108999. [Google Scholar] [CrossRef] [PubMed]
  467. Rippin; Sharma, A.K.; Beniwal, V. Biosynthesis and medicinal applications of proanthocyanidins: A recent update. Biocatal. Agric. Biotechnol. 2022, 45, 102500. [Google Scholar] [CrossRef]
  468. Morimoto, H.; Hatanaka, T.; Narusaka, M.; Narusaka, Y. Molecular investigation of proanthocyanidin from Alpinia zerumbet against the influenza A virus. Fitoterapia 2022, 158, 105141. [Google Scholar] [CrossRef] [PubMed]
  469. Shen, W.; Li, W.; Shao, Y.; Zeng, J. Proanthocyanidin delays litchi peel browning by inhibiting ethylene biosynthesis, respiratory metabolism, and phenol oxidase activities. Sci. Hortic. 2023, 309, 111677. [Google Scholar] [CrossRef]
  470. Rohr, G.E.; Meier, B.; Sticher, O. Analysis of procyanidins. Stud. Nat. Prod. Chem. 2000, 21, 497–570. [Google Scholar] [CrossRef]
  471. Yang, H.; Tuo, X.; Wang, L.; Tundis, R.; Portillo, M.P.; Simal-Gandara, J.; Yu, Y.; Zou, L.; Xiao, J.; Deng, J. Bioactive procyanidins from dietary sources: The relationship between bioactivity and polymerization degree. Trends Food Sci. Technol. 2021, 111, 114–127. [Google Scholar] [CrossRef]
  472. Zhou, P.; Zhang, L.; Li, W.; Zhang, S.; Luo, L.; Wang, J.; Sun, B. In vitro evaluation of the anti-digestion and antioxidant effects of grape seed procyanidins according to their degrees of polymerization. J. Funct. Food 2018, 49, 85–95. [Google Scholar] [CrossRef]
  473. Na, W.; Ma, B.; Shi, S.; Chen, Y.; Zhang, H.; Zhan, Y.; An, H. Procyanidin B1, a novel and specific inhibitor of Kv10.1 channel, suppresses the evolution of hepatoma. Biochem. Pharmacol. 2020, 178, 114089. [Google Scholar] [CrossRef]
  474. Liu, Y.; Sun, R.; Li, X.-P.; Wu, L.; Chen, H.; Shen, S.; Li, Y.; Wei, Y.; Deng, G. Procyanidins and its metabolites by gut microbiome improves insulin resistance in gestational diabetes mellitus mice model via regulating NF-KB and NLRP3 inflammasome pathway. Biomed. Pharmacother. 2022, 151, 113078. [Google Scholar] [CrossRef]
  475. Chen, R.; Hu, T.; Wang, M.; Hu, Y.; Chen, S.; Wei, Q.; Yin, X.; Xie, T. Functional characterization of key polyketide synthases by integrated metabolome and transcriptome analysis on curcuminoid biosynthesis in Curcuma wenyujin. Syn. Sys. Biotechnol. 2022, 7, 849–861. [Google Scholar] [CrossRef] [PubMed]
  476. Wu, J.-N.; Tu, Q.-K.; Xiang, X.-L.; Shi, Q.-X.; Chen, G.-Y.; Dai, M.-X.; Zhang, L.-J.; Yang, M.; Song, C.-W.; Huang, R.-Z.; et al. Changes in curcuminoids between crude and processed turmeric based on UPLC-QTOF-MS/MS combining with multivariate statistical analysis. Chin. J. Anal. Chem. 2022, 50, 100108. [Google Scholar] [CrossRef]
  477. El-Hawaz, R.; Tharayil, N.; Bridges, W.; Adelberg, J. Mineral nutrition of Curcuma longa L. in bioreactors affects subsequent development of curcuminoids following transfer to the greenhouse. Ind. Crop. Prod. 2016, 83, 186–193. [Google Scholar] [CrossRef]
  478. Ramirez-Ahumada, M.D.; Timmermann, B.N.; Gang, D.R. Biosynthesis of curcuminoids and gingerols in turmeric (Curcuma longa) and ginger (Zingiber officinale): Identification of curcuminoid synthase and hydroxycinnamoyl-CoA thioesterases. Phytochemistry 2006, 67, 2017–2029. [Google Scholar] [CrossRef]
  479. Aarthi, S.; Suresh, J.; Leela, N.K.; Prasath, D. Multi environment testing reveals genotype-environment interaction for curcuminoids in turmeric (Curcuma longa L.). Ind. Crop. Prod. 2020, 145, 112090. [Google Scholar] [CrossRef]
  480. Abouheif, S.A.; Sallam, S.M.; El Shoafy, S.M.; Kassem, F.F.; Shawky, E. A green extraction approach using natural deep eutectic solvents enhances the in-vivo bioavailability of curcuminoids from turmeric extracts. Ind. Crop. Prod. 2022, 189, 115790. [Google Scholar] [CrossRef]
  481. Wang, H.; Luo, J.; Zhang, Y.; He, D.; Jiang, R.; Xie, X.; Yang, Q.; Li, K.; Xie, J.; Zhang, J. Phospholipid/hydroxypropyl-β-cyclodextrin supramolecular complexes are promising candidates for efficient oral delivery of curcuminoids. Int. J. Pharm. 2020, 582, 119301. [Google Scholar] [CrossRef]
  482. Ahmed, T.; Gilani, A.-H. A comparative study of curcuminoids to measure their effect on inflammatory and apoptotic gene expression in an Aβ plus ibotenic acid-infused rat model of Alzheimer’s disease. Brain Res. 2011, 1400, 1–18. [Google Scholar] [CrossRef]
  483. Monton, C.; Charoenchai, L.; Sukasaeree, J.; Sueree, L. Quantitation of curcuminoid contents, dissolution profile, and volatile oil content of turmeric capsules produced at some secondary government hospitals. J. Food Drug Anal. 2016, 24, 493–499. [Google Scholar] [CrossRef] [Green Version]
  484. Solani, L.N.; Nelson, G.L.; Ronayne, C.T.; Lueth, E.A.; Foxley, M.A.; Jonnalagadda, S.K.; Gurrapu, S.; Mereddy, V.R. Synthesis, in vitro, and in vivo evaluation of novel functionalized quaternary ammonium curcuminoids as potential anti-cancer agents. Bioorg. Med. Chem. Lett. 2015, 25, 5777–5780. [Google Scholar] [CrossRef] [PubMed]
  485. Zhou, J.-L.; Zheng, J.-Y.; Cheng, X.-Q.; Xin, G.-Z.; Wang, S.-L.; Xie, T. Chemical markers, knockout coupled with UHPLC-HRMS-based metabolomics reveals anti-cancer integration effect of the curcuminoids of turmeric (Curcuma longa L.) on lung cancer cell line. J. Pharm. Biomed. Anal. 2019, 175, 112738. [Google Scholar] [CrossRef] [PubMed]
  486. Wei, M.-M.; Zhao, S.-J.; Dong, X.-M.; Wang, Y.-J.; Fang, C.; Wu, P.; Song, G.-Q.; Gao, J.-N.; Huang, Z.-H.; Xie, T.; et al. A combination index and glycoproteomics-based approach revealed synergistic anticancer effects of curcuminoids of turmeric against prostate cancer PC3 cells. J. Ethonpharmacol. 2021, 267, 113467. [Google Scholar] [CrossRef] [PubMed]
  487. Feng, Y.; Li, B.; Yang, L.; Liu, Y. Co-amorphous delivery systems based on curcumin and hydroxycinnamic acids: Stabilization, solubilization, and controlled release. LWT 2022, 170, 114091. [Google Scholar] [CrossRef]
  488. Vafaeipour, Z.; Razavi, B.M.; Hosseinzadeh, H. Effects of turmeric (Curcuma longa) and its constituent (Curcumin) on the metabolic syndrome: An updated review. J. Integr. Med. 2022, 20, 193–203. [Google Scholar] [CrossRef]
  489. Aguiar, D.D.; Gonzaga, A.C.R.; Teofilo, A.L.H.; Miranda, F.A.; Perez, A.D.C.; Duarte, I.D.G.; Romero, T.R.L. Curcumin induces peripheral antinociception by opioidergic and cannabinoidergic mechanism: Pharmacological evidence. Life Sci. 2022, 293, 120279. [Google Scholar] [CrossRef]
  490. Fu, Y.-S.; Chen, T.-H.; Weng, L.; Huang, L.; Lai, D.; Weng, C.-F. Pharmacological properties and underlying mechanisms of curcumin and prospects in medicinal potential. Biomed. Pharmacother. 2021, 141, 111888. [Google Scholar] [CrossRef]
  491. Yixuan, L.; Qaria, M.A.; Sivasamy, S.; Jianzhong, S.; Daochen, Z. Curcumin production and bioavailability: A comprehensive review of curcumin extraction, synthesis, biotransformation and delivery systems. Ind. Crop. Prod. 2021, 172, 114050. [Google Scholar] [CrossRef]
  492. Wang, Y.; Xu, S.; Han, C.; Wang, L.; Zheng, Q.; Wang, S.; Huang, Y.; Wei, S.; Qin, Q. Curcumin inhibits Singapore grouper iridovirus infection through multiple antiviral mechanisms. Aquaculture 2023, 562, 738870. [Google Scholar] [CrossRef]
  493. Liu, Z.; Shi, B.; Wang, Y.; Xu, Q.; Gao, H.; Ma, J.; Jiang, X.; Yu, W. Curcumin alleviates aristolochic acid nephropathy based on SIRT1/Brf2/HO-1 signaling pathway. Toxicology 2022, 479, 153297. [Google Scholar] [CrossRef] [PubMed]
  494. Lu, K.-H.; Lu, P.W.-A.; Lin, C.-W.; Yang, S.-F. Curcumin in human osteosarcoma: From analogs to carriers. Drug Discov. Today 2023, 28, 103437. [Google Scholar] [CrossRef]
  495. Ming, T.; Tao, Q.; Tang, S.; Zhao, H.; Yang, H.; Liu, M.; Ren, S.; Xu, H. Curcumin: An epigenetic regulator and its application in cancer. Biomed. Pharmacother. 2022, 156, 113956. [Google Scholar] [CrossRef] [PubMed]
  496. Priyadarsini, K.I. The chemistry of curcumin: From extraction to therapeutic agent. Molecules 2014, 19, 20091–20112. [Google Scholar] [CrossRef] [Green Version]
  497. Zagury, Y.; David, S.; Edelman, R.; Brill, R.H.; Livney, Y.D. Sugar beet pectin as a natural carrier for curcumin, a water-insoluble bioactive for food and beverage enrichment: Formation and characterization. Innov. Food Sci. Emerg. Technol. 2021, 74, 102858. [Google Scholar] [CrossRef]
  498. Ezati, M.; Ghavamipour, F.; Adibi, H.; Pouraghajan, K.; Arab, S.S.; Sajedi, R.H.; Khodarahmi, R. Design, synthesis, spectroscopic characterizations, antidiabetic, in silico and kinetic evaluation of novel curcumin-fused aldohexoses. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2023, 285, 121806. [Google Scholar] [CrossRef] [PubMed]
  499. Elanthendral, G.; Shobana, N.; Meena, R.; Prakash, P.; Samrot, A.V. Utilizing pharmacological properties of polyphenolic curcumin in nanotechnology. Biocatal. Agric. Biotechnol. 2021, 38, 102212. [Google Scholar] [CrossRef]
  500. Jena, A.B.; Dash, U.C.; Duttaroy, A.K. An in silico investigation on the interactions of curcumin and epigallocatechin-3-gallate with NLRP3 inflammasome complex. Biomed. Pharmacother. 2022, 156, 113890. [Google Scholar] [CrossRef]
  501. Sheikholeslami, M.A.; Parvardeh, S.; Ghafghazi, S.; Sabetkasaei, M. Curcumin attenuates morphine dependence by modulating μ-opioid receptors and glial cell-activated neuroinflammation in rat. Neuropeptides 2022, 98, 102318. [Google Scholar] [CrossRef]
  502. Bhavani, T.; Gautam, A. Expression analysis of synaptic plasticity genes in curcumin-treated amnesic mice model. Mater. Today Proc. 2023, 73, 307–311. [Google Scholar] [CrossRef]
  503. Xie, L.; Ji, X.; Zhang, Q.; Wei, Y. Curcumin combined with photodynamic therapy, promising therapies for the treatment of cancer. Biomed. Pharmacother. 2022, 146, 112567. [Google Scholar] [CrossRef] [PubMed]
  504. Hashemi, M.; Mirzaei, S.; Barati, M.; Hejazi, E.S.; Kakavand, A.; Entezari, M.; Salimimoghadam, S.; Kalbasi, A.; Rashidi, M.; Taheriazam, A.; et al. Curcumin in the treatment of urological cancers: Therapeutic targets, challenges and prospects. Life Sci. 2022, 309, 120984. [Google Scholar] [CrossRef]
  505. Mahjoob, M.; Stochaj, U. Curcumin nanoformulations to combat aging-related diseases. Ageing Res. Rev. 2021, 69, 101364. [Google Scholar] [CrossRef] [PubMed]
  506. Chen, Y.; Wang, J.; Jing, Z.; Ordovas, J.M.; Wang, J.; Shen, L. Anti-fatigue and anti-oxidant effects of curcumin supplementation in exhaustive swimming mice via Nrf2/Keap1 signal pathway. Curr. Res. Food Sci. 2022, 5, 1148–1157. [Google Scholar] [CrossRef] [PubMed]
  507. Sahebkar, A.; Sathyapalan, T.; Guest, P.C.; Barreto, G.E. Identification of difluorinated curcumin molecular targets linked to traumatic brain injury pathophysiology. Biomed. Pharmacother. 2022, 148, 112770. [Google Scholar] [CrossRef]
  508. Huang, W.-T.; Larsson, M.; Lee, Y.-C.; Liu, D.-M.; Chiou, G.-Y. Dual drug-loaded biofunctionalized amphiphilic chitosan nanoparticles: Enhanced synergy between cisplatin and demethoxycurcumin against multidrug-resistant stem-like lung cancer cells. Eur. J. Pharm. Biopharm. 2016, 109, 165–173. [Google Scholar] [CrossRef]
  509. Gullaiya, S.; Nagar, A.; Dubey, V.; Singh, V.; Kumar, A.; Tiwari, P.; Agrawal, S.S. Modulation of disease related immune events by demethoxycurcumin against autoimmune arthritis in rats. Biomed. Aging Pathol. 2013, 3, 7–13. [Google Scholar] [CrossRef]
  510. Yodkeeree, S.; Ampasavate, C.; Sung, B.; Aggarwal, B.B.; Limtrakul, P. Demethoxycurcumin suppresses migration and invasion of MDA-MB-231 human breast cancer cell line. Eur. J. Pharmacol. 2010, 627, 8–15. [Google Scholar] [CrossRef]
  511. Kumar, R.; Lal, N.; Nemaysh, V.; Luthra, P.M. Demethoxycurcumin mediated targeting of MnSOD leading to activation of apoptotic pathway and inhibition of Akt/NF-KB survival signalling in human glioma U87 MG cells. Toxicol. Appl. Pharmacol. 2018, 345, 75–93. [Google Scholar] [CrossRef]
  512. Lin, H.-Y.; Lin, J.-N.; Ma, J.-W.; Yang, N.-S.; Ho, C.-T.; Kuo, S.-C.; Way, T.-D. Demethoxycurcumin induces autophagic and apoptotic responses on breast cancer cells in photodynamic therapy. J. Funct. Food 2015, 12, 439–449. [Google Scholar] [CrossRef]
  513. Zhang, L.; Wu, C.; Zhao, S.; Yuan, D.; Lian, G.; Wang, X.; Wang, L.; Yang, J. Demethoxycurcumin, a natural derivative of curcumin attenuates LPS-induced pro-inflammatory responses through down-regulation of intracellular ROS-related MAPK/NF-KB signaling pathways in N9 microgia induced by lipopolysaccharide. Int. Immunopharmacol. 2010, 10, 331–338. [Google Scholar] [CrossRef]
  514. Lu, B.; Chen, X.; Chen, H.; Li, Q.; Li, H.; Xu, Y.; Li, Y.; Shen, X.; Jiang, R. Demethoxycurcumin mitigates inflammatory responses in lumbar disc herniation via MAPK and NF-KB pathways in vivo and in vitro. Int. Immunopharmacol. 2022, 108, 108914. [Google Scholar] [CrossRef] [PubMed]
  515. Mehanny, M.; Hathout, R.M.; Geneidi, A.S.; Mansour, S. Bisdemethoxycurcumin loaded polymeric mixed micelles as potential anti-cancer remedy: Preparation, optimization and cytotoxic evaluation in a HepG-2 cell model. J. Mol. Liq. 2016, 214, 162–170. [Google Scholar] [CrossRef]
  516. Paul, M.; Manikanta, K.; Hemshekhar, M.; Sundaram, M.S.; Naveen, S.; Ramesh, T.N.; Kemparaju, K.; Girish, K.S. Bisdemethoxycurcumin promotes apoptosis in human platelets via activation of ERK signaling pathway. Toxicol. In Vitro 2020, 63, 104743. [Google Scholar] [CrossRef]
  517. Liu, J.; Wang, Q.; Omari-Siaw, E.; Adu-Frimpong, M.; Liu, J.; Xu, X.; Yu, J. Enhanced oral bioavailability of bisdemethoxycurcumin-loaded self-microemulsifying drug delivery system; Formulation design, in vitro and in vivo evaluation. Int. J. Pharm. 2020, 590, 119887. [Google Scholar] [CrossRef] [PubMed]
  518. Zhang, J.; Han, H.; Zhang, L.; Wang, T. Dietary bisdemethoxycurcumin supplementation attenuates lipopolysaccharide-induced damages on intestinal redox potential and redox status of broilers. Poult. Sci. 2021, 100, 101061. [Google Scholar] [CrossRef]
  519. Fu, M.; Fu, S.; Ni, S.; Wang, D.; Hong, T. Inhibitory effects of bisdemethoxycurcumin on mast cell-mediated allergic diseases. Int. Immunopharmacol. 2018, 65, 182–189. [Google Scholar] [CrossRef]
  520. Mahattanadul, S.; Nakamura, T.; Panichayupakaranant, P.; Phdoongsombut, N.; Tungsinmunkong, K.; Bouking, P. Comparative antiulcer effect of bisdemethoxycurcumin and curcumin in a gastric ulcer model system. Phytomedicine 2009, 16, 342–351. [Google Scholar] [CrossRef]
  521. Ponnusamy, S.; Zinjarde, S.; Bhargava, S.; Rajamohanan, P.R.; RaviKumar, A. Discovering bisdemethoxycurcumin from Curcuma longa rhizome as a potent small molecule inhibitor of human pancreatic α-amylase, a target for type-2 diabetes. Food Chem. 2012, 135, 2638–2642. [Google Scholar] [CrossRef]
Table 1. Classes of phenolic compounds in plants [53].
Table 1. Classes of phenolic compounds in plants [53].
ClassStructure
Simple phenolics, benzoquinonesC6
Hydroxybenzoic acidsC6-C1
Acetophenones, phenylacetic acidsC6-C2
Hydroxycinnamic acids, phenylpropanoids (coumarins, isocoumarins, chromones, chromenes)C6-C3
NapthoquinonesC6-C4
XanthonesC6-C1-C6
Stilbenes, anthraquinonesC6-C2-C6
Flavonoids, isoflavonoidsC6-C3-C6
Lignans, neolignans(C6-C3)2
Biflavonoids(C6-C3-C6)2
Lignins(C6-C3)n
Condensed tannins (proanthocyanidins or flavolans)(C6-C3-C6)n
Table 2. Examples of hydroxybenzoic and hydroxycinnamic acids.
Table 2. Examples of hydroxybenzoic and hydroxycinnamic acids.
Phenolic AcidsExamplesMolecular Formula
Hydroxybenzoic acidsGallic acid
Protocatechuic acid
C7H6O5
C7H6O4
Hydroxycinnamic acidsp-coumaric acid
Caffeic acid
Ferulic acid
Sinapic acid
C9H8O3
C9H8O4
C10H10O4
C11H12O5
Other components
CoumarinsUmbelliferone
Esculetin
Scopoletin
C9H6O3
C9H6O4
C10H8O4
StilbenesResveratrol
Piceatannol
Pterostilbene
C14H12O3
C14H12O4
C16H16O3
CurcuminoidsCurcumin
Demethoxycurcumin
Bisdemethoxycurcumin
C21H20O6
C20H18O5
C19H16O4
Condensed tannins or proanthocyanidinsProcyanidin B1C30H26O12
LignanSesaminC20H18O6
Table 3. Generic structure of major classes of flavonoids.
Table 3. Generic structure of major classes of flavonoids.
Flavonoids Molecular Formula
FlavonesApigenin
Luteolin
Chrysin
C15H10O5
C15H10O6
C15H10O4
FlavonolsKaempferol
Quercetin
Isorhamnetin
C15H10O6
C15H10O7
C16H12O7
FlavanonesNaringenin
Eriodictyol
Hesperetin
C15H12O5
C15H12O6
C16H14O6
Flavanols C15H14O2
Anthocyanidin C15H11O+
FlavanonolsTaxifolin
Aromadendrin
C15H12O7
C15H12O6
Flavan-3-olsGallocatechin
Catechin
C15H14O7
C15H14O6
IsoflavonesGenistein
Daidzein
Formononetin
C15H10O5
C15H10O4
C16H12O4
Table 4. Important points about phenolic acids and their derivatives.
Table 4. Important points about phenolic acids and their derivatives.
The Derivatives of Phenolic AcidsKey PointsReferences
FlavonoidsThe largest group of natural phenolic compounds.[54,114]
Their structure is based on a 15-carbon phenyl benzopyran skeleton (C6-C3-C6, i.e., A-C-B rings).[54,114]
Based on differences in the pyran ring, flavonoids can be categorized into flavones, isoflavones, flavanonols, flavonols, flavanones, flavan-3-ols, and anthocyanidins.[54,114]
The majority occur as glycosides, except for flavan-3-ols, which are rarely glycosylated.[54,114]
Different patterns of hydroxylation and methylation of the A and B rings consequently result in a variety of compounds for each flavonoid category.[54,114]
Flavones have a double bond between C-2 and C-3, a keto function in C-4, and the B ring is attached at C-2.[54,114]
The most common flavonoes in medicinal and aromatic plants are luteolin, apigenin, and glycosides.[54,114]
In isoflavones, the B ring is attached at C-3 and the main components are daidzein, genistein, and glycitein.[54,114]
Flavonols are flavones bearing a hydroxyl group at C-3, such as kaempferol, quercetin, and myricetin.[54,114]
In flavanones, the C-ring has no double bond between C2 and C3, such as in naringenin, eriodictyol, and hesperetin.[54,114]
Flavanonols, also called dihydroflavonols, have the same saturated C-ring as flavanones but are hydroxylated at C-3.[54,114]
Flavan-3-ols, also referred to as flavanols, also contain a saturated C-ring, but lack the keto group at C-4, and are hydroxylated at C-3, such as catechin and gallocatechin, or as oligomers and polymers.[54]
In anthocyanidins, the C-ring lacks the keto group at C-4, is hydroxylated at C-3, and, uniquely, has two double bonds forming the flavylium cation, such as in cyanidin, petunidin, malvidin, pelargonidin, peonidin, and delphinidin.[54]
StilbenesThey are based on 1,2-diphenylethylene, which has a C6-C2-C6 skeleton.[115]
They can be found as aglycones, monomers, oligomers, or glycosylated derivatives.[116]
TanninsTannins are high molecular weight polyphenolic compounds.[117,118]
They can be synthesized as a defensive mechanism in response to pathogen attack and abiotic stresses such as UV radiation.[117,118]
Based on their structures, tannins in plants can be classified into mainly hydrolysable tannins and condensed tannins, also known as proanthocyanidins.[117,118]
Hydrolysable tannins are built based on gallic acid and are divided into the gallotannins and ellagitannins.[117,118]
QuinonesThey contain a di-one or di-ketone group.[119]
They are distinguished into benzoquinones and naphthoquinones and are based on their derivative molecules.[119]
They may occur as monomers, dimers, trimers, glycosides, or in reduced forms.[119]
CoumarinsThey may occur in a free or glycosylated state.[120]
They are divided into six categories, namely simple coumarins, furanocoumarins, dihydrofuranocoumarins, pyranocoumarins, phenylcoumarins, and bicoumarins.[120]
CurcuminoidsThey widely occur in Curcuma spp., especially in the rhizomes of Curcuma longa (turmeric).[121,122]
There are three major curcuminoids, namely curcumin, demethoxycurcumin, and bis-demethoxycurcumin.[121,122]
The structure of curcumin consists of a keto-enol tautomeric unsaturated chain linking two aromatic rings bearing a hydroxyl and methoxy group.[121,122]
LigninsLignans consist of two phenylpropane units joined together by a β-β′ bond.[123]
They are divided into eight categories, namely dibenzylbutyrolactols, dibenzocyclooctadienes, dibenzylbutanes, dibenzylbutyrolactones, arylnaphthalene, aryl-tetralins, furans, and furofurans.[123]
Table 5. Biological activities of phenolic compounds.
Table 5. Biological activities of phenolic compounds.
Health BenefitsKey PointsReferences
Antioxidant activity* The stem of Dendrophthoe falcata (Loranthaceae) plant had a high content of phenolic and flavonoid compounds and very high antioxidant activities.[182]
* The phenolic compounds of Buchenavia tetraphylla, Buchenavia tomentosa, and Lippia sidoides provided the main contributions to the antioxidant potential.[183]
* The total phenolic, flavonoid, and antioxidant capacity of all blueberry cultivars increased nonlinearly with ripening.[184]
* Cynaroside, rosmarinic acid, cosmosiin, luteolin, apigenin, and acacetin were the main components in ethyl acetate extracts of Salvia absconditiflora, Salvia sclarea, and Salvia palaestina with antioxidant activity.[185]
* Phenolic compounds from Pistacia lentiscus L. black fruits exhibited potent antioxidant properties.[186]
* Lycium berries of different species contained a total of 186 phenolic compounds, which exhibited potent antioxidant activities.[187,188]
* Stachys species contained important bioactive phenolics and had promising antioxidant impacts.[189]
* Acacia nilotica pods and bark had potent total phenolic content, antioxidant activity, and tyrosinase inhibitory properties.[190]
* Bersama abyssinica (Meliathacea) was rich in phenolic compounds, flavonoids and coumarin, and 7,8-Dimethoxycoumarin with high antioxidant activity.[191]
* Epicatechin was the main monomeric polyphenol in the profile of longan phenolics.[192]
* Epicatechin, quercetin 3-O-rhamnoside, and kaempferol were responsible for the high antioxidant activity of Litsea glaucescens.[193]
* The water extract of Amsonia orientalis leaves exhibited promising antioxidant activity when used at low concentration.[194]
* The ethanolic extract of Amsonia orientalis leaves had the highest phenolic substance content and 2,2-diphenyl-1-picrylhydrazyl (DPPH) scavenging activity.[194]
Anti-inflammatory activity* A variety of phenolic compounds and stilbene derivatives in different parts of germinated peanut suggested that the peanut sprout exerted high anti-inflammatory effects and may be related to the polyphenolic content and antioxidant properties.[195]
* Fermented olive cream and Lactiplantibacillus (Lpb.) plantarum IMC513 reduced proinflammatory cytokine levels.[196]
* Allium scorodoprasum L. subsp. rotundum extract showed high anti-inflammatory inhibitory effects against xanthine oxidase activity.[197]
* Helleborus purpurascens demonstrated the strongest anti-inflammatory potential, especially because of fatty acids.[198]
* Thalictrum minus possessed combined anti-inflammatory and antioxidant effects.[198]
* The leaf of Aurea helianthus demonstrated strong anti-inflammatory activity that reduced NO production.[199]
Antifungal activity* Hypericum empetrifolium aerial parts extract (HEA) exhibited antifungal activity against Candida tropicalis with 19.53 μg/mL.[200]
* Allium sativum extract revealed strong antifungal activity effects against Curvularia spp., Trichophyton spp., and Geotrichum spp.[201]
* Rosa micrantha flowers extract revealed fungicide effects in Candida glabrata.[202]
* Phenolic compounds of Ulmus davidiana var. japonica showed antifungal activity against Cryptococcus neoformans and Candida albicans.[203]
* Zataria multiflora essential oils could act as natural fungicides; carvacrol and thymol chemotypes of Zataria multiflora inhibited five important fungal plant pathogens.[204]
* Aconitum heterophyllum and Polygonum bistorta exhibited significant antimicrobial and antioxidant activity.[205]
Antimicrobial activity* The antimicrobial activities of mint and thyme were due to a wide range of diverse phenolics such as menthone, menthyl acetate, menthol, terpenes, and thyme.[206]
* Phenolic compounds of Codonopsis lanceolata plants exhibited notable antimicrobial activity.[207]
* Phenolic compounds of cashew (Anacardium occidentale L.) compounds identified included mainly flavanols, which showed high antimicrobial activity.[208]
* Ixora coccinea Linn. root contained bioactive phenolic compounds including pyrocatechol, catechin, and chlorogenic acid with potent antimicrobial effects.[209]
Antibacterial activity* The ethyl acetate fraction of Scirpus holoschoenus showed the highest antioxidant activity and antibacterial effect for Staphylococcus aureus and Bacillus subtilis.[210]
* Rhanterium adpressum showed antibacterial activity.[211]
* The lignum of Rhus verniciflua contained high content of phenolic compounds with less urushiols, which suggests efficient antibacterial activity with less toxicity.[212]
* Phenolic compounds of Scrophularia ningpoensis Hemsl. showed antibacterial activity.[213]
* Flavonoids, saponin, alkaloids, tannins, steroids, and terpenoids of Solanum incanum L. and Harrisonia abyssinica Oliv. exhibited antibacterial activity.[214]
* The phenolic extracts from Cerbera manghas, Commelina diffusa, Peperomia pellucida, Kleinhovia hospita, Mikania micrantha, Homalanthus nutans, Psychotria insularum, Phymatosorus scolopendria, Piper graeffei, and Schizostachyum glaucifolium exhibited antibacterial activities.[215]
Anti-Coronavirus Properties* Curcumin has been suggested as a potential treatment choice for patients with COVID-19 because it inhibits ACE2 and suppresses the entry of SARS-CoV-2 into the cells.[216]
* Theaflavin, the compound responsible for the orange/black color of black tea, is a potent inhibitor of the RNA polymerase of SARS-CoV-2.[217]
* Catechin gallate and gallocatechin gallate also showed high inhibitory activity against SARS-CoV-2 N protein in a concentration-dependent manner and affected virus replication.[218]
* Myricetin could be further tested and developed as a potential SARS-CoV-2 antiviral.[219]
* The phenolic compounds Kadsurenin L. and Methysticin of Piper nigrum are candidate ligands for inhibiting COVID-19.[220]
* Plant-derived phenolic compounds of Isatis indigotica root were frequently used for the prevention of SARS during the SARS outbreaks in east Asia.[221]
* Among phenolic acid constituents, chlorogenic acid, caffeic acid, and gallic acid of Sambucus Formosana Nakai reduced cytopathicity and virus yield in HCoV-NL63-infected cells.[222]
* Some phenolic compounds such as diethylstilbestrol, enterodiol, enterolactone, flavokawain A, flavokawain B, and flavokawain C showed excellent to good inhibitory activities against collagenase, elastase enzymes, and SARS-CoV-2.[223]
* The phenolic compounds of blackcurrant (Ribes nigrum L.) had antiviral activity in Coxsackievirus A9 and human coronavirus HCoV-OC-43.[224]
Neuroprotective potential* Hydroxytyrosol obtained from olive exhibited neuroprotective impacts on multiple chronic neurodegenerative diseases including Alzheimer’s, Parkinson’s, and multiple sclerosis.[225]
* The protective impacts of oil palm phenolics against neurodegenerative diseases have been recently identified.[226]
* Phenolic compounds of Hypericum wightianum, namely Hyperwightin E and petiolin G, revealed noticeable neuroprotection at 10 μM.[227]
* Inula viscosa (L.) Greuter has high total phenolics and flavonoids and demonstrated neuroprotective properties.[228]
* Maclura tinctoria leaf aqueous extract contained high phenolic components, and it has been found that neuroprotective effects of it could be associated with the presence of the phenolic compounds identified.[229]
Skin health* Phenolic compounds from Lippia microphylla and Dimorphandra gardneriana presented a high sun protector factor because of the presence of sakuranetin flavonoids and quercetin glycosides.[230]
* Among Moroccan medicinal plants, Allium cepa L., Chamaeleon gummifer (L.) Cass, and Salvia rosmarinus Schleid. Mill. leaves were the most commonly used for different types of skin diseases.[231]
* Panax ginseng C.A. Meyer and Nardostachys chinensis Bat. have been confirmed for the treatment of skin pigmentary disorders.[232]
* The protective effects on skin cells associated with blueberry phenolic compounds that included inhibition of proliferation and cell cycle arrest in malignant cells decreased oxidized macromolecules, down-regulated inflammatory cytokine genes, and mitigated oxidative stress.[233]
Wound healing* Gel containing Ipomoea pes-caprae (Ipc) phenolic-rich leaf extract accelerated the wound-healing process.[234]
* Loranthus acaciae exhibited high phenolic contents and wound healing activities.[235]
* Haworthia limifolia showed excellent wound-healing properties because of high phenolic contents.[236]
* Lawsonia inermis and Azadirachta indica are well known for wound healing.[237]
* Aloe vera (Aloe barbadensis) is one of the oldest medicinal plants with wound healing activity for a variety of skin disorders including burns as well as infections and diabetic dermal wounds.[238]
* Amphimas pterocarpoides leaves and stem bark have high phenolic and flavonoid contents, and it has been proven that leaf and stem bark ointments increased the rate of wound healing in rats.[239]
Anticancer activity* Sedum dendroideum showed anti-proliferative activity in breast cancer cells.[240]
* Hypericum perforatum extract exhibited a remarkable total phenol content, which showed high anticancer activity.[241]
* Ficus palmata Forssk. exhibited high total phenolic and flavonoids contents, which showed its high anticancer activity.[242]
* Euphorbia thymifolia and Euphorbia hirta showed anticancer activity against ascites carcinoma in mice models.[243]
* Vitis vinifera L. contained high phenolic components, which can be considered as a promising potential for an anticancer drug.[244]
* Phenolic compounds and alkaloid compounds of Dysphania ambrosioides might have significantly contributed to anticancer activity.[245]
* Sisymbrium officinale had considerable phenolic and flavonoids contents, which was why it showed anticancer activity.[246]
Table 6. Coumarin-based compounds obtained from the medicinal plants used by various ancient medical systems [340].
Table 6. Coumarin-based compounds obtained from the medicinal plants used by various ancient medical systems [340].
CompoundsMolecular FormulaPharmaceutical Activity
6-hydroxy-4-methoxy-5-methylcoumarinC11H10O4Microtubule stabilizing agent
(+)-CalanolideC22H26O5Anti-HIV agent
InophyllumC25H24O5Anti-HIV agent
TheraphinC22H28O6Anticancer agent
Antimalarial agent
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Sun, W.; Shahrajabian, M.H. Therapeutic Potential of Phenolic Compounds in Medicinal Plants—Natural Health Products for Human Health. Molecules 2023, 28, 1845. https://doi.org/10.3390/molecules28041845

AMA Style

Sun W, Shahrajabian MH. Therapeutic Potential of Phenolic Compounds in Medicinal Plants—Natural Health Products for Human Health. Molecules. 2023; 28(4):1845. https://doi.org/10.3390/molecules28041845

Chicago/Turabian Style

Sun, Wenli, and Mohamad Hesam Shahrajabian. 2023. "Therapeutic Potential of Phenolic Compounds in Medicinal Plants—Natural Health Products for Human Health" Molecules 28, no. 4: 1845. https://doi.org/10.3390/molecules28041845

Article Metrics

Back to TopTop