Next Article in Journal
Modelling Nannochloropsis gaditana Growth in Reactors with Different Geometries, Determination of Kinetic Parameters and Biochemical Analysis in Response to Light Intensity
Next Article in Special Issue
The Effects of a Natural Polyphenol Extract from Sugarcane (Saccharum officinarum) on the Growth, Survival, and Feed Conversion Efficiency of Juvenile Black Tiger Shrimp (Penaeus monodon)
Previous Article in Journal
Experimental Research on In Situ Uniaxial Tensile Response of Silica-Based PLC Optical Splitters
Previous Article in Special Issue
Comparative Study of Potato (Solanum tuberosum L.) and Sweet Potato (Ipomoea batatas L.): Evaluation of Proximate Composition, Polyphenol Content, Mineral and Antioxidant Activities
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Natural Sources, Pharmacological Properties, and Health Benefits of Daucosterol: Versatility of Actions

1
Laboratory of Histology, Embryology and Cytogenetic, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat 10100, Morocco
2
Laboratory of Organic Chemistry, Catalysis and Environment, Department of Chemistry, Faculty of Sciences, Ibn Tofail University, B.P.:133, Kenitra 14000, Morocco
3
Laboratory of Human Pathologies Biology, Department of Biology, Mohammed V University in Rabat, Rabat 10106, Morocco
4
Environment and Health Team, Polydisciplinary Faculty of Safi, Cadi Ayyad University, Sidi Bouzid B.P. 4162, Morocco
5
Molecular Modeling, Materials, Nanomaterials, Water and Environment Laboratory, CERNE2D, Department of Chemistry, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco
6
Molecular Engineering, Valorization and Environment Team, Polydisciplinary Faculty of Taroudant, Ibn Zohr University, Agadir B.P. 32/S, Morocco
7
Laboratory of Pharmacology, National Agency of Medicinal and Aromatic Plants, Taouanate 34025, Morocco
8
Laboratoire d’Ingénierie des Matériaux Organométalliques, Moléculaires et Environnement, Sidi Mohamed Ben Abdellah University, Fez B.P. 1796, Morocco
9
Department of Biology, Science Faculty, Selcuk University, Campus of Alaeddin Keykubat, Konya 42130, Turkey
10
Department of Food, Nutrition and Packaging Sciences, Clemson University, Clemson, SC 29634, USA
11
Centro Tecnológico de la Carne de Galicia, Rúa Galicia No. 4, Parque Tecnológico de Galicia, San Cibrao das Viñas, 32900 Ourense, Spain
12
Facultade de Ciencias, Universidade de Vigo, Área de Tecnoloxía dos Alimentos, 32004 Ourense, Spain
13
Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Via Pansini, 5, 80131 Naples, Italy
14
Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Naples, Italy
*
Authors to whom correspondence should be addressed.
Appl. Sci. 2022, 12(12), 5779; https://doi.org/10.3390/app12125779
Submission received: 30 April 2022 / Revised: 26 May 2022 / Accepted: 2 June 2022 / Published: 7 June 2022

Abstract

:
Daucosterol is a saponin present in various natural sources, including medicinal plant families. This secondary metabolite is produced at different contents depending on species, extraction techniques, and plant parts used. Currently, daucosterol has been tested and explored for its various biological activities. The results reveal potential pharmacological properties such as antioxidant, antidiabetic, hypolipidemic, anti-inflammatory, immunomodulatory, neuroprotective, and anticancer. Indeed, daucosterol possesses important anticancer effects in many signaling pathways, such as an increase in pro-apoptotic proteins Bax and Bcl2, a decrease in the Bcl-2/Bax ratio, upregulation of the phosphatase and tensin homolog (PTEN) gene, inhibition of the PI3K/Akt pathway, and distortion of cell-cycle progression and tumor cell evolution. Its neuroprotective effect is via decreased caspase-3 activation in neurons and during simulated reperfusion (OGD/R), increased IGF1 protein expression (decreasing the downregulation of p-AKT3 and p-GSK-3b4), and activation of the AKT5 signaling pathway. At the same time, daucosterol inhibits key glucose metabolism enzymes to keep blood sugar levels within normal ranges. Therefore, this review describes the principal research on the pharmacological activities of daucosterol and the mechanisms of action underlying some of these effects. Moreover, further investigation of pharmacodynamics, pharmacokinetics, and toxicology are suggested.

1. Introduction

Daucosterol [(3β)-stigmast-5-en-3-yl β-D-glucopyranoside], a natural β-sitosterol glucoside, is a saponin phytosterol belonging to different families and genera found in several countries such as China, Vietnam, Mexico, Algeria, Tunisia, Italy, Iran, Cameroon, Saudi Arabia, Nigeria, Korea, and India [1,2,3,4].
Daucosterol is the major component of several plant extracts, namely Chinese Fallopia cillinerve root extract [1], Dendrobium huoshanense and Dendrobium officinale stem extract [5], and Hyssopus cuspidatus Boriss aerial part extract [6]. In addition, this natural agent was found to be the main component of aerial part extracts of Centaurea resupinata subsp. dufourii [7] and Ononis mitissima L. [8] from Algeria. It was also detected at high concentrations in leaf and stem extracts of Prangos ferulacea from Iran [9], aerial part extracts of Cassia italic collected from Saudi Arabia [3], and leaf extracts of Ficus deltoidea from Indonesia [10] and Dioscorea batatas collected from Korea [11]. Furthermore, daucosterol is the principal component of aerial part extracts of Astragalus tanae found in Italy [12] and Landolphia owariensis collected from Nigeria [13], as well as whole plant and root extracts of Rheum turkestanicum from Iran [14,15]. Other investigations have reported high contents of daucosterol in plant extracts, namely Archidendron clypearia harvested in Vietnam [16] and Parasenecio pseudotaimingasa leaf extract [17] and Grewia optiva Drummond ex Burret stem bark extract in Pakistan [18].
Concerning the purification and isolation of daucosterol from plants, several methods have been used as spectroscopic techniques, including NMR and FT-IR, on the stems and leaves of Prangos ferulacea [9] and the aerial parts of Cassia italica [3] and Ononis mitissima L. [8]. NMR (1H NMR, 13C NMR, COSY, HSQC, and HMBC) and mass spectroscopy (ESI–MS) have been used to elucidate daucosterol in Centaurea resupinata subsp. Dufourii [7]. This molecule has been elucidated from Dioscorea opposite with silica gel column chromatography (SGCC) using CHCl3-MeOH [19], from Phyllenthus emblica L. with thin layer chromatography (TLC) [20], from Portulaca oleracea L. [21] and Eriobotrya fragrans Champ [22] with 1H and 13C aided by HMQC, and from Arctotis arctotoides [23] using NMR (COSY, NOESY, HMQC, and HMBC) and mass spectra.
Several studies have highlighted the pharmacological properties of daucosterol, including chemopreventive [24,25,26], neuroprotective [27,28,29,30], antioxidant [7,8,9], anti-inflammatory [31], antidiabetic [32], inhibition and interactions of alpha-amylase by daucosterol from the peel of Chinese water chestnut [33], and immunomodulatory [34]. Immunoregulatory activity by daucosterol, a β-sitosterol glycoside, induces a protective Th1 immune response against disseminated Candidiasis in mice [35]. Regarding its chemopreventive potential, daucosterol was found to possess important anticancer activity on various tumor cell lines and could be considered as one of the novel pharmacological treatment strategies for cancer: for breast adenocarcinoma through different cellular and molecular mechanisms, such as an increase in pro-apoptotic protein Bax and Bcl2, a decrease in the Bcl-2/Bax ratio, upregulation of the PTEN gene, inhibition of the PI3K/Akt pathway, loss of mitochondrial membrane potential and cytochrome c (Cyt c) [36,37], repression of cell migration and invasion, and induction of cell death by cell-cycle arrest and apoptosis [38,39]; for lung cancer by increasing reactive oxygen species (ROS) level and promoting intrinsic apoptotic cell death on A549 cells mediated by increased expression of caspase-3, caspase-9, Bax, PARP inactivation, Cyt c release, and diminished bcl-2 protein expression; and for hepatocellular carcinoma by inhibiting the proliferation, migration, and invasion of hepatocellular carcinoma cells via Wnt/β-Catenin signaling [40]. Daucosterol has also been shown to exert a neuroprotective action by decreasing caspase-3 activation in neurons treated with oxygen–glucose deprivation and simulated reperfusion (OGD/R), as well as increasing the expression level of IGF1 protein, reducing the downregulation of p-AKT3 and p-GSK-3b4, thus activating the AKT5 signal pathway [28]. Further, daucosterol showed a neuroprotective property against H2O2-induced oxidative stress mediated through downregulation of MAPK pathways, minimizing ROS, and upregulation of antioxidant gene expression (HO-1, CAT, and SOD2) [41]. It could play a key role as an antioxidant by fighting against free radicals [3,8,9], which lead to a potential reduction of the DPPH (2,2-diphenyl-1-picrylhydrazyl) radical. This molecule can also act as an antidiabetic agent; it could be considered a great candidate to prevent hyperglycemia due to its antidiabetic activity through the inhibition of α-glucosidase [42] and α-amylase enzymes [33]. In addition, this molecule has been shown to act as a promising anti-inflammatory drug that notably decreases nitric oxide (NO) content in different inflammatory tests [43,44]. Furthermore, other studies have demonstrated that daucosterol possesses immunomodulatory potential [34]. It was able to regulate the population and activation of immune cells, including Treg cells, macrophages, B1 cells, and NK cells in colitis mediated by suppressing the release of inflammatory cytokines such as IL-6, TNF-α, IFN-γ, and IL-1β [31]. Further, this natural compound has shown potent lipolysis activity and could be used by physicians in the management of certain types of disease, thus conferring medicinal principles [35]. It was found to exhibit hypolipidemic actions with a sharp decrease in serum total cholesterol, triglyceride, and low-density lipoprotein cholesterol (LDL-C) levels [45] Nevertheless, daucosterol did not show antibacterial activity [23].
This review has attempted to compile a complete and current understanding of the origin, phytochemical, biological, and pharmacological process analysis of the daucosterol molecule, providing an overview to explain its mechanism of action (in vitro and in vivo) and its future applications in drug discovery, particularly for neuroprotective and chemopreventive effects.

2. Sources of Daucosterol

Daucosterol (Figure 1), a natural sterol, is a glucoside of β-sitosterol, mainly synthesized by plants (Table 1). Daucosterol is the major component of Chinese Fallopia cillinerve root extract [1], Dendrobium huoshanense stem extract, and Dendrobium officinale [5]. The richness of the compound depends on geographical factors and the plant part used. Hechtia glomerata Zucc from Mexico is characterized by its richness in daucosterol [2]. Additionally, daucosterol is the major compound in Crataegus gracilior flowers [46,47], Litsea cubeba extract [4], and Chinese plants Eleocharis dulcis [33] and Ipomoea batatas [25]. Other works have mentioned the richness of Cameroonian plants such as Crateva adansonii (stem bark and leaves) in this compound [24,48].
Hyssopus cuspidatus Boriss aerial part extract showed that daucosterol is the major compound of this plant collected in China [6], as well as of Dioscorea batatas collected in Korea [11] and Ficus deltoidea leaf extract harvested from Indonesia [10]. Daucosterol is the major compound of the extract of Astragalus tanae aerial parts collected in Italy [12], of the fruit extract of Acanthopanax sessiliflorus (Rupr. and Maxim.), collected in China [49], of the aerial part extracts of Centaurea resupinata subsp. Dufourii [7] and Ononis mitissima L. [8] collected in Algeria. It has also been detected in the extracts of Prangos ferulacea leaves and stems collected in Iran [9] and the extract of Cassia italic aerial parts collected in Saudi Arabia [3].
Daucosterol is an isolate of different plants of the caprifoliaceae family harvested in China: Dipsacus chinensis, Dipsacus asperoides, Dipsacus japonicas, Dipsacus kangdigensis, and Dipsacus daliensis [50]. Analysis of Rheum turkestanicum whole plant and root extract showed that daucosterol is the major compound of this plant collected in Iran [14,15], of Heracleum persicum [15] and Landolphia owariensis collected from Nigeria [13], and of Streptocaulon griffithii [51] and Streptocaulon griffithii whole plant extract [51].
Richness in daucosterol has also been recorded in other plant extracts, namely Archidendron clypearia collected in Vietnam [16], Shibataea chinensis Nakai leaves from China [52], and Morus alba leaves from Korea [35]. Likewise, other extracts have been characterized by the dominance of this compound, such as the roots of Geranium collinum [53] and the leaves of Lasianthus hartii [54], Salvia syriaca, Sedum caeruleum, Adenophora triphylla, Pulicaria inuloides, Salvia miltiorrhiza, Salvia officinalis, and Rosa canina L. [27,28,55,56,57,58].
Indeed, daucosterol is the major compound of many plant extracts, such as extracts of the whole plant Clematis heracleifolia collected in Korea [59], aerial parts of Dorema glabrum Fisch. and C.A. Mey. collected in Iran [60], Artemisia apiacea [34,61], seeds of Juglans regia [28], peels and pulps of Pyrus spp. [62], salvia sahendica, Punica granatum, Helicteres isora L., Ceiba pentandra L., Eria spicata, Lysimachia clethroides, and Randia dumetorum [63,64,65,66,67,68].
Daucosterol has been isolated from Litchi chinensis seed extract [69], Parasenecio pseudotaimingasa leaf extract [17], Grewia optiva Drummond ex Burret stem bark extract collected in Pakistan [18], Urtica angustifolia leaf, root, and stem extracts [70], and Salvia limbata aerial part extracts from Iran, Turkey, and Afghanistan [71]. Further, it has been found in stem extracts of Lindera glauca [72], Sphallerocarpus gracilis, Hypericum ascyron L., Paeonia lactiflora, Paeonia suffruticosa, Alangium kurzi, Boerhaavia diffusa, and Cassia mimosoides var. [72,73,74,75,76,77,78].
Furthermore, daucosterol is among the major compounds of Phyllenthus emblica L. [20], Mitragyna speciose [79], Astragalus membranaceus [80], stem bark and twig extracts of Bennettiodendron leprosipes [81], Flacourtia ramontchi [81], Alchornea cordifolia (Schumach. and Thonn.) Müll. Arg. [82], Flueggea virosa [83], Eriobotrya fragrans [22], Portulaca oleracea L. [21], Pteridium aquilinum [84], and Brassica campestris ssp rapa [85].
This has also been observed with extracts of Penthorum chinense, Arctotis arctotoides, Selinum cryptotaenium, Embelia ribes, Punica granatum, Dioscorea opposita, Junellia aspera, Sitophilus oryzae, Astragalus mongholicus Bunge, Hemiphragma heterophyllum, Dendrobium moniliforme, Punica granatum, Nepeta cataria L. var. citriodora [19,23,86,87,88,89,90,91,92,93,94,95], Euphorbia altotibetic [96], Rhodiola sachalinensis roots [97], Gnetum montanum [98], and Ajania fruticulosa aerial parts.
Table 1. Sources of Daucosterol.
Table 1. Sources of Daucosterol.
Plant
Family
CountryParts UsedReferences
Fallopia cillinerve
Polygonaceae
ChinaRoots[1]
Litsea cubeba
Lauraceae
VietnamNot reported[4]
Hechtia glomerata Zucc
Bromeliaceae
MexicoLeaves[2]
Eleocharis dulcis
Cyperaceae
ChinaNot reported[33]
Ipomoea batatas
Convolvulaceae
ChinaNot reported[25]
Dendrobium huoshanense
Orchidaceae
ChinaStems[5]
Dendrobium officinale
Orchidaceae
Crataegus gracilior
Rosaceae
MexicoFlowers[46]
Hyssopus cuspidatus Boriss.
Lamiaceae
ChinaAerial parts[6]
Ficus deltoidea
Moraceae
IndonesiaLeaves[10]
Dioscorea batatas
Dioscoreaceae
KoreaNot reported[11]
Crateva adansonii
Capparaceae
CameroonStem bark[24]
Leaves[48]
Astragalus tanae
Fabaceae
ItalyAerial parts[12]
Acanthopanax sessiliflorus (Rupr. and Maxim.) Seem.
Araliaceae
ChinaFruits[49]
Centaurea resupinata subsp. dufourii
Asteraceae
AlgeriaAerial parts[7]
Ononis mitissima L.
Fabaceae
AlgeriaAerial parts[8]
Prangos ferulacea
Apiaceae
IranLeaves and stems[9]
Cassia italica
Fabaceae
Saudi ArabiaAerial parts[3]
Dipsacus chinensis
Caprifoliaceae
ChinaRoots[50]
Dipsacus asperoides
Caprifoliaceae
Dipsacus japonicas
Caprifoliaceae
Dipsacus kangdigensis
Caprifoliaceae
Dipsacus daliensis
Caprifoliaceae
Rheum turkestanicum
Polygonaceae
IranWhole plant
Roots
[14,15]
Heracleum persicum
Apiaceae
IranWhole plant[15]
Landolphia owariensis
Apocynaceae
NigeriaLeaves[13]
Streptocaulon griffithii
Asclepiadaceae
ChinaNot reported[51]
Streptocaulon griffithii
Asclepiadaceae
ChinaWhole plant[51]
Archidendron clypearia
Fabaceae
VietnamWhole plant[16]
Shibataea chinensis Nakai
Gramineae
ChinaLeaves[52]
Morus alba
Moraceae
KoreaLeaves[35]
Geranium collinum
Geraniaceae
ChinaRoots[53]
Lasianthus hartii
Rubiaceae
ChinaLeaves[54]
Salvia syriaca
Lamiaceae
IranRoots[55]
Sedum caeruleum
Crassulaceae
AlgeriaAerial parts[56]
Adenophora triphylla
Campanulaceae
KoreaNot reported[27]
Pulicaria inuloides
Asteraceae
YemenAerial parts[57]
Salvia miltiorrhiza
Lamiaceae
ChinaRoots[28]
Salvia officinalis
Lamiaceae
ChinaRoots[28]
Rosa canina L.
Rosaceae
IranFruits[58]
Dorema glabrum Fisch. and C.A. Mey.
Apiaceae
IranAerial parts[60]
Clematis heracleifolia
Ranunculaceae
KoreaWhole plant[59]
Artemisia apiacea
Asteraceae
KoreaNot reported[61,99]
Pyrus spp.
Rosaceae
ChinaPeels and pulps[62]
Salvia sahendica
Lamiaceae
IranAerial parts[36]
Punica granatum
Lythraceae
TunisiaFlowers[64]
Helicteres isora L.
Sterculiaceae
IndiaFruits[65]
Ceiba pentandra L.
Bombacaceae
Seeds
Eria spicata
Orchidaceae
ChinaWhole plant[68]
Lysimachia clethroides
Primulaceae
ChinaAerial parts[66]
Randia dumetorum
Rubiaceae
Not reportedBark[67]
Litchi chinensis
Sapindaceae
ChinaSeeds[69]
Parasenecio pseudotaimingasa
Asteraceae
KoreaLeaves[17]
Grewia optiva Drummond ex Burret
Tiliaceae
PakistanStem bark[18]
Urtica angustifolia
Urticaceae
ChinaLeaves, roots, and stems[74]
Salvia limbata
Lamiaceae
Iran, Turkey, and AfghanistanAerial parts[71]
Lindera glauca
Lauraceae
KoreaStems[72]
Sphallerocarpus gracilis
Apiaceae
ChinaRoots[73]
Hypericum ascyron L.
Hypericaceae
ChinaWhole plant[74]
Paeonia lactiflora
Paeoniaceae
KoreaRoots[75]
Paeonia suffruticosa
Paeoniaceae
Root bark
Alangium kurzi
Alangiaceae
IndonesiaStem bark[76]
Boerhaavia diffusa
Nyctaginaceae
NigeriaLeaves[77]
Cassia mimosoides var. nomame Makino
Fabaceae
KoreaSeeds[78]
Phyllenthus emblica L.
Phyllanthaceae
ChinaFruits[20]
Mitragyna speciosa
Rubiaceae
AmericaLeaves[79]
Astragalus membranaceus
Fabaceae
KoreaRoots[80]
Bennettiodendron leprosipes
Flacourtiaceae
ChinaStem bark and twigs[81]
Flacourtia ramontchi
Flacourtiaceae
Bark and twigs
Alchornea cordifolia (Schumach. and Thonn.) Müll. Arg.
Euphorbiaceae
BelgiumLeaves and root bark[82]
Flueggea virosa
Euphorbiaceae
ChinaTwigs and leaves[83]
Eriobotrya fragrans Champ
Rosaceae
ChinaFruits and leaves[22]
Portulaca oleracea L.
Portulacaceae
EgyptNot reported[21]
Pteridium aquilinum
Pteridaceae
ChinaNot reported[84]
Brassica campestris ssp rapa
Brassicaceae
KoreaRoots[85]
Penthorum chinense
Penthoraceae
ChinaWhole plant[86]
Arctotis arctotoides
Asteraceae
Not reportedNot reported[23]
Selinum cryptotaenium
Umbelliferae
ChinaRoots[87]
Embelia ribes
Myrsinaceae
ChinaRoots[88]
Punica granatum
Lythraceae
ChinaFlowers[89]
Dioscorea opposita
Dioscoreaceae
ChinaAerial parts[19]
Junellia aspera
Verbenaceae
SpainAerial parts[90]
Sitophilus oryzae
Curculionidae
Not reported
Astragalus mongholicus Bunge
Fabaceae
ChinaRoots[91]
Hemiphragma heterophyllum
Scrophulariaceae
ChinaWhole plant[92]
Dendrobium moniliforme
Orchidaceae
ChinaStems[95]
Punica granatum
Lythraceae
ChinaSeeds[93]
Nepeta cataria L. var. citriodora
Lamiaceae
PolandSeeds[94]
Euphorbia altotibetic
Euphorbiaceae
ChinaWhole plant[96]
Rhodiola sachalinensis
Crassulaceae
KoreaRoots[97]
Gnetum montanum
Gnetaceae
ChinaNot reported[98]
Ajania fruticulosa
Asteraceae
ChinaAerial parts[100]
Rhodiola fastigiata
Crassulaceae
ChinaRhizomes[101]
Jatropha curcas
Euphorbiaceae
ChinaRoots[102]
Gentiana algida
Gentianaceae
ChinaWhole plant[103]
Gentiana siphonantha
Gentianaceae
ChinaRhizomes and roots[104]
Gentiana scabra Bunge
Gentianaceae
ChinaRoots[105]
Rabdosia coetsa
Lamiaceae
ChinaLeaves[106]
Artemisia sieversiana
Asteraceae
ChinaAerial parts[105]
Inula racemosa
Asteraceae
Roots
Amanoa oblongifolia
Euphorbiaceae
PeruStem bark[107]
Rhodiola rosea L.
Crassulaceae
RussiaRhizomes[108]
Acanthopanax sessiliflorum
Araliaceae
Not reportedRoots[109]
Leaves

3. Extraction and Characterization

Many research groups have isolated and purified daucosterol from various medicinal plants [3,7,49], as showed in Table 2.
Spectroscopic techniques such as NMR, FT-IR, and elemental analysis have been used to elucidate the structure of daucosterol isolated and purified from the leaves and stems of Prangos ferulacea [9], as well as the aerial parts of Cassia italica [3] and Ononis mitissima L. [8]. NMR techniques (1H NMR, 13C NMR, COSY, HSQC, and HMBC) and mass spectroscopy (ESI-MS) have also been used to elucidate this molecule from Centaurea resupinata subsp. Dufourii [7], while spectroscopic techniques such as ESI/MS, 1H- and 13C-NMR have been used for its purification from the fruit of Acanthopanax sessiliflorus (Rupr. and Maxim.) Seem. [49]; SGCC with further purification with MeOH was the characterization method for Hyssopus cuspidatus Boriss. aerial parts [6]. Similarly, daucosterol was elucidated from many plants such as Rheum turkestanicum [14], Morus alba using NMR by 1H, 13C, DEPT, COSY, HSQC, and HMBC NMR [35], and from Pulicaria inuloides [57], Salvia syriaca [55], Adenophora triphylla using 1H-NMR, 13C-NMR; and electron-impact (EI) mass spectra [27] for Sedum caeruleum [56], Rosa canina L. [58], Dorema glabrum Fisch. and C.A. Mey. [60], Salvia sahendica [36], Pyrus spp., Punica granatum, Helicteres isora L., and Ceiba pentandra L. [62,64,65]. The complete interpretation of 1H and 13C NMR spectra using 2D NMR (HSQC, HMBC, and DQF-COSY) allowed the identification of daucosterol in extracts of Litchi chinensis [69], Randia dumetorum [67], and Lysimachia clethroides [66]. TLC, IR, and ESI-MS spectra have been used for identification in Sphallerocarpus gracilis [73] and Hypericum ascyron L. [74].
It was obtained from Grewia optiva Drummond ex Burret [18], Boerhaavia diffusa [77], and Astragalus membranaceus [80] using 1D and 2D NMR (HMQC, HMBC, COSY, NOESY, and J-resolved) with EI and HRMS; from Phyllenthus emblica L. [20] using SGCC and thin-layer chromatography (TLC); and from Portulaca oleracea L. [21] and Eriobotrya fragrans Champ [22] using 1H and 13C aided with HMQC. Besides, daucosterol was found in Alchornea cordifolia (Schumach. and Thonn.) Müll. Arg. [82] and Arctotis arctotoides [23] using NMR (COSY, NOESY, HMQC and HMBC). Daucosterol was eluted from Dioscorea opposite with SGCC using CHCl3-MeOH [19], with 13C-NMR from Astragalus mongholicus Bunge and Punica granatum [91,93], and with a combination of spectral methods (IR, EIMS, H and 2CNMR, DEPT, COSY, NOESY, and HETCOR) from Artemisia sieversiana [105].

4. Evaluation of Biological Properties

4.1. Antioxidant Activity

Daucosterol is an antioxidant molecule with an essential role in the fight against free radicals (Table 3). Abdollahnezhad et al. [9] demonstrated that this constituent has potent antioxidant properties, with significant activity using the DPPH (2, 2-diphenyl-1-picrylhydrazyl) free radical assay. Indeed, it showed a potential reduction of DPPH radicals, with 50% of inhibition (IC50 = 490 ± 2.9 µg/mL) and a percentage inhibition of 19.7% at 100 mg/mL [3] and a potential reduction with IC50 = 27.3 ± 0.015 µg/mL in another, similar study [8]. Other methods such as H2O2, FTC, FRAP, and PPM showed DPPH radical inhibition equal to 31.18 ± 0.5%, 37.12 ± 0.44%, 122.23 ± 0.014 µg EAA/mg ex, and 16.44 ± 0.0012 µg EAA/mg ex, respectively [8]. Several studies have confirmed this potential for DPPH radical inhibition with varying results: IC50 = 36.263 ± 0.005 µg/mL [7], IC50 = 155.0 ± 0.5 μM [14], IC50 = 224.1 ± 8.2 µg/mL [60], IC50 = 6.0 ± 0.1 mg/L [64], EC50 > 250 µg/mL [69], IC50 = 11.42 ± 0.07 µg/mL [66], IC50 = 108.14 ± 9.54 µg GAE/ mL [73], and IC50 = 38.86 µg/mL [20]. However, Shomirzoeva and collaborators demonstrated that daucosterol does not affect the DPPH radical [6]. On the other hand, it inhibited the activity of the ABTS radical with a potential inhibition at 50% equal to 4.8 ± 0.04 mg/mL and a percentage inhibition of 27% at the concentration of 0.04 mg/mL [110], plus EC50 = 143.4 µg/mL [69], IC50 = 9.02 ± 0.11 µg/mL [66], IC50 = 1.66 ± 0.15 µmol TE/g of DW [73], and IC50 = 0.71 ± 0.01 µg/mL [20].

4.2. Anticancer Activity

Several works have shown that daucosterol possesses important anticancer activity on various tumor cell lines and could be considered one of the novel pharmacological treatment strategies for cancer (Table 4). Esmaeili et al. [36] isolated this molecule from Salvia sahendica to assess its apoptotic and anti-proliferative activity against human breast adenocarcinoma MCF-7 cells using MTT assay, lactate dehydrogenase (LDH) leakage assay, and flow cytometry. They found that daucosterol inhibits cell proliferation and induces cytotoxicity in MCF-7 cells, including PARP proteolytic activity, DNA fragmentation, and cell morphological changes. Indeed, at 80 μM, maximum inhibition of cell growth and survival was approximately 60%. Anti-cancer drug mechanisms move from the subcellular to the molecular level by increasing the levels of the pro-apoptotic proteins (Bax and Bcl2) and decreasing the Bcl-2/Bax ratio, upregulating the PTEN gene to inhibit the PI3K/Akt pathway, inducing the loss of mitochondrial membrane potential, and by Cyt c release in breast cancer cells [110,112,113]. On the other hand, Zhao et al. [110] assessed the anticancer effect of daucosterol against human breast cancer cell line MCF-7 and gastric cancer cell lines MGC803, BGC823, and AGS. The results demonstrated that this compound exerts an important antiproliferative activity against the studied cell lines, with IC50 values of 16.95, 19.96, 3.13, and 24.19 μM, respectively. This effect was induced by autophagy in a ROS-dependent manner. Wang et al. [38] recorded an IC50 value of 26.6 μM on human colon cancer cell line HCT-116. Daucosterol can repress cell migration and cell invasion in these cells and cause cell death by cell-cycle arrest and apoptosis. Indeed, it significantly inhibited the proliferation of human lung cancer cell line A549, with an IC50 value of 17.46 μg/mL targeting multiple checkpoints, including cell-cycle arrest at the G2/M phase and induction of cell apoptosis [112]. Using MCF-7, MDA-MB-231, and 4T1 breast cancer cells, Han et al. [37] revealed that daucosterol exhibits antitumor activities, with an IC50 = 53.27 μg/mL on MCF-7 and IC50 > 1000 on MDA-MB-231 and 4T1. This molecule also inhibited tumor growth in vivo using MCF-7 xenografts in nude mice by decreasing the expression of Bcl-xl, Bcl-2, and XIAP, increasing Bax, Bad, inducing the activation of caspase-dependent apoptosis in tumor tissues, and inactivation of the upstream PI3K/Akt/NF-κB pathway. Moreover, Gao et al. [114] reported that daucosterol inhibits cell proliferation, induces cell-cycle arrest, and promotes autophagy-dependent apoptosis in human prostate cancer cell lines (PC3 and LNCap) via activation of JNK signaling. Indeed, this phytosterol strongly inhibited the growth of human lung cancer cell line A549, with an IC50 value of 20.9 μM, by increasing ROS levels and promoting intrinsic apoptotic cell death of A549 cells. This effect was mediated by increased expression of caspase-3, caspase-9, and Bax, PARP inactivation, Cyt c release, and diminished expression of bcl-2 protein (Figure 2) [26]. Recently, Han et al. [25] evaluated the antiproliferative effect of daucosterol extracted from sweet potatoes on three breast cancer cell lines (MCF-7, MDA-MB-231, and 4T1) and nontumorigenic breast epithelial cell line MCF-10A using MTT assay. The results showed that this compound inhibited the proliferation of breast cancer MCF-7 cells by inactivating the phosphoinositide 3-kinase/protein kinase B pathway, but had only weak effects on the proliferation of MDA-MB-231, 4T1, and MCF-10A cells. Nguedia and coworkers [24] investigated antitumor effects in vivo using the environmental carcinogen 7,12-dimethylbenz[a]anthracene (DMBA). The results demonstrated that daucosterol reduced, at all doses, tumor volume as well as proteins and cancer antigen 1. It also exhibited an antioxidant effect by decreasing malondialdehyde (MDA) levels and increasing catalase activity. In another study, it inhibited LNCaP, DU145, and PC3 prostate carcinoma cell growth and proliferation via downregulated cell cycle proteins (cdk1, cdk2, pcdk1, cyclin A and B), downregulated anti-apoptotic proteins (Akt, pAKT, and Bcl-2), and upregulated the pro-apoptotic protein Bax [114,115,116]. On the other hand, the in vivo anticancer effect of daucosterol on primary tumor growth and pulmonary metastasis was studied using a BALB/c mouse model of a breast tumor. After 35 days of treatment, daucosterol suppressed primary tumor growth, reduced the number of lung metastases, and delayed the trend of increasing numbers of captured CTCs in the blood circulation [117]. Using a murine H22 hepatoma allograft model in ICR mice, Zhao et al. [110] showed that daucosterol treatment inhibits tumor growth in vivo by inducing intracellular ROS generation and subsequent autophagic cell death.

4.3. Neuroprotective Activity

Some studies have shown that daucosterol exhibits neuroprotective effects [29,30,40,41] (Table 5). Indeed, Jiang and collaborators [40] showed that this molecule exhibits a neuroprotective action that reduces neuronal loss and cell apoptosis by diminishing caspase-3 activation in oxygen–glucose deprivation and simulated reperfusion (OGD/R)-treated neurons. It also increased the expression level of IGF1 protein and diminished the downregulation of p-AKT3 and p-GSK-3b4, thus activating the AKT5 signal pathway. Moreover, daucosterol decreased the downregulation of the anti-apoptotic proteins Mcl-1 and Bcl-2 and diminished the expression level of the pro-apoptotic protein Bax. In another study, Chung et al. [41] investigated the neuroprotective effects of daucosterol on H2O2-induced cell death of human brain neuroblastoma SK-N-SH cells using MTT and lactate dehydrogenase (LDH) assays, annexin-V/PI double-staining, and flow cytometry. Therefore, daucosterol exhibited neuroprotective activity against H2O2-induced oxidative stress through downregulation of MAPK pathways, minimizing ROS, and upregulation of antioxidant genes (HO-1, CAT, and SOD2) (Figure 3). Moreover, oral administration of daucosterol ameliorated amyloid beta-induced learning and memory impairment in rats by inhibiting beta-amyloid-induced hippocampal ROS production, and prevented beta-amyloid-induced hippocampal neuronal damage and restored hippocampal synaptophysin expression level [29]. Recently, Zhang et al. [30] investigated the neuroprotective effects of daucosterol in a cerebral ischemia/reperfusion (I/R) rat model. Accordingly, daucosterol attenuated brain damage and neuronal cell apoptosis caused by I/R injury by upregulating the PI3K/Akt/mTOR signaling pathway and suppressing iNOS expression in the ischemic zone.

4.4. Anti-Inflammatory Activity

Using in vivo inflammation experiments (e.g., xylene-induced ear edema and carrageenan-induced paw), edema could be mediated by inflammatory factors [118], such as serotonin, histamine, prostaglandins, and bradykinin. Huang et al. [119] investigated the anti-inflammatory activity of two sterols and two triterpenes extracted from Pyrus bretschneideri Rehd., including daucosterol, β-sitosterol, ursolic acid, and oleanolic acid. Mice treated orally with the isolated compounds showed significant dose-dependent inhibition of edema compared to control mice [119].
Daucosterol isolated from the leaves of Liriodendron chinensis could be a natural anti-inflammatory agent due to its strong inhibitory effect on the activated inflammatory cells. As observed in the experiment performed by Yang et al. [43], this compound could significantly decrease the NO content of LPS-induced rat peritoneal macrophages [43].
In another study, Kim et al. [120] used LPS-treated RAW 264.7 macrophage cells to assess the anti-inflammatory action of phytochemicals, including daucosterol, isolated from fermented bark of Acanthopanax sessiliflorus (FAS). The anti-inflammatory responses to FAS revealed decreased NO production and inhibition of COX-2, iNOS, collagenase, and pro-inflammatory cytokines (TNF-α and IL-6) (Figure 4) [120]. Additionally, Bui et al. tested the anti-inflammatory activity of bioactive compounds, including daucosterol, extracted from Sanchezia speciosa Leonard’s leaf ethanol extract. This research reported that daucosterol might have anti-inflammatory action against heat-induced protein denaturation, with moderate inhibition (IC50 = 245.59 ± 3.17 µg/mL) compared to 3-methyl-1Hbenz[f]indole-4,9-dione, which exhibited the strongest inhibition (IC50 = 193.70 ± 5.24 µg/mL) [121].
The pharmacological effects of daucosterol as an anti-inflammatory agent isolated from the leaves and root bark of Alchornea cordifolia was also tested using a mouse ear edema model in the study carried out by Mavar-Manga and collaborators. At a dose of 90 g/cm2, daucosterol was found to be more active (50% inhibition)—along with acetyl aleuritolic acid, N1, N2 diisopentenyl guanidine, and N1,N2,N3-triisopentenyl guanidine—than indomethacin. However, β-sitosterol and di(2-ethylhexyl) phthalate were less effective [82].
In addition, daucosterol could suppress dextran sulfate sodium (DSS)-induced colitis in mice. Colitis is an inflammatory response of the large bowel, which may be of infectious or autoimmune origin [122]. Jang et al. [31] found daucosterol decreased DSS-induced ROS production and the expression of pro-inflammatory cytokines, including TNF-α, IL-6, and IL-1β [31] (Figure 4). These outcomes indicate that daucosterol may be useful as a candidate in folk medicine and for adopting new therapeutics as an alternative for treating several inflammatory diseases associated with excessive NO production.

4.5. Immunomodulatory Effects

Recently, modulation of the immune system has become a promising way to treat several human pathologies. Indeed, the reactivity of our immune system can be decreased due to several risk factors. This decrease induces a disequilibrium between immune response and disorders, including pathogenic microbial infections. In several ways, reinforcement of the immune system can establish homeostasis and remove these disorders. Moreover, it is well established that regulatory T cells (Treg) have a significant contribution to managing immune homeostasis. In particular, some previous works have indicated that Treg cells possess an essential function in inhibiting colitis-induced inflammatory responses by regulating inflammation and suppressing the release of inflammatory cytokines through modulating different genes such as NF-κB, TNF-α, interleukins (IL-1 and IL-6), and chemokines [123].
Jang et al. investigated the immunomodulatory effects of daucosterol in a mouse model of DSS-induced colitis. This research demonstrated that this molecule could regulate the population and activation of immune cells, including Treg cells, macrophages, B1 cells, and NK cells in colitis. These cells are involved in several diseases (malignant tumors, infections, mucosal immunity, allergies, etc.). They also found that this bioactive compound suppresses ROS and colitis-induced inflammatory cytokines, such as IL-6, TNF-α, IFN-γ, and IL-1β, and is associated with downregulation of macrophage infiltration and upregulation of Treg cell number [31].
To discover new immunoregulatory approaches using natural drugs isolated from medicinal plants to fight against illnesses caused by Candida albicans, Lee et al. evaluated the possible immunomodulatory activity of daucosterol against disseminated Candidiasis in mice by regulating the immune response. They observed that splenic CD4+ T cells (DSCD4T) in daucosterol-treated mice produce IFN-γ and IL-2 cytokines more abundantly than cytokines IL-4 and IL-10, inducing the polarization of CD4+ T cells towards a Th1-type immune response [34]. However, some studies reported that the release of Th1 cytokine would enhance host resistance to disseminated candidiasis by improving the killing capacity of different immune cells, including activated macrophages, NK cells, and cytotoxic T cells against cells infected with C. albicans [124,125]. Furthermore, Yang et al. [40] proved the immunocompetent activity of daucosterol isolated from the leaves of Liriodendron chinensis. They demonstrated that daucosterol, among other bioactive compounds, markedly decreased the NO content of LPS-induced rat peritoneal macrophages and decreased splenic lymphocyte proliferation in mice [43]. These assessments could pave the way for new immunoregulatory strategies exerted by this biologically active compound for the treatment of numerous immunologic pathologies.

4.6. Antidiabetic Activity

Daucosterol extracted from the chloroform fraction of Swertia longifolia Boiss., was tested among other examined compounds in the study performed by Saeidnia and collaborators (Table 6). The results showed that daucosterol had the highest inhibitory activity against the digestive enzyme α-amylase (57.5 ± 3.1% in a 10 mg/mL) [126]. In the same context, the inhibitory activity of daucosterol purified from the peel of Chinese water chestnuts was evaluated in the study conducted by Gu et al. [33] against the same enzyme (Table 6). Using fluorescence quenching, enzyme inhibition, and molecular docking models, results showed that three saponins from Chinese water chestnut bark exhibit a potent α-amylase inhibitory effect, and that daucosterol was the major inhibitory factor of this enzyme with a mixed-mode [33].
To find effective strategies for the treatment of diabetes using α-glucosidase inhibitors, Sheng et al. [42] assessed the antidiabetic effect of five natural drugs isolated from the flowers of Musa spp. (Baxijiao), including ferulic acid, vanillic acid, β-sitosterol, daucosterol, and 9-(4′-hydroxyphenyl)-2-methoxyphenalen-1-one, against this enzyme. Daucosterol exhibited an excellent α-glucosidase inhibitory action, with an IC50 value of 247.35 mg/L and an inhibition constant of 2.34 mg/L. β-sitosterol and 9-(4ꞌ-hydroxyphenyl)-2-methoxyphenalen-1-one also showed potent α-glucosidase inhibiting activity [42]. Similarly, Numonov and colleagues investigated the antidiabetic effect of ten bioactive compounds, including daucosterol isolated from the root of Geranium collinum [53]. Using molecular docking, they observed that daucosterol could play a key role in inhibiting human α-glucosidase, while polyphenolic agents are probably involved in inhibiting PTP-1B.
On the other hand, Ivorra and collaborators investigated the possible effects of daucosterol and its aglycone (β-sitosterol) as an antihyperglycemic and insulin releaser [128]. The action of these agents on plasma insulin and glucose levels in normal and hyperglycemic rats after oral treatment was found to increase fasting plasma insulin levels. Additionally, they noted that both compounds enhance oral glucose tolerance and improve glucose-induced insulin release. In addition, Asghari et al. [58] tested the capacity of D-glucono-1,4-lactone and daucosterol isolated from Rosa canina fruits to inhibit α-glucosidase using the substrate p-nitrophenyl-α-D-glucopyranoside (pNPG). The findings revealed that both agents contribute to the inhibition of α-glucosidase with a high effect; the IC50 values of D-glucono-1,4-lactone and daucosterol on yeast α-glucosidase were 6.5 ± 2.0 and 13.3 ± 1.9 µM, respectively [58].
From the perspective of characterizing and isolating the antidiabetic molecules present in the leaves of Costus pictus, Benny et al. [128] observed that β-sitosterol-3-O-β-D-glucoside extracted from Costus pictus leaves exhibited a significant dose-dependent hypoglycemic action in rats after enteric coating compared to the uncoated compound [128]. The inhibitory effects on α-glucosidase in vitro and the increase in postprandial glycemia in maltose-loaded mice by the saponin component of Eleocharis dulcis bark were investigated in a recent study by Gu et al. [33]. Using NMR spectroscopy, three saponins were detected: campesterol glucoside, daucosterol, and stigmasterol glucoside. All three saponins were found to exert potent α-glucosidase inhibition, with IC50 values of 10.03 mg/L (campesterol glucoside), 7.68 mg/L (stigmasterol glucoside), and 5.67 mg/L (daucosterol), which were notably greater than that of acarbose (91.50 mg/L). Further, daucosterol showed the greatest inhibitory capacity against α-glucosidase [33]. Based on these findings, daucosterol could be considered an excellent candidate to prevent hyperglycemia due to its antidiabetic activity through the inhibition of α-glucosidase and α-amylase. However, further in vivo and in vitro investigations are warranted to elucidate the mechanisms involved.

4.7. Hypolipidemic Activity

In the objective to investigate the lipolysis effect of phytosterols isolated from mulberry (Morus alba) leaves, Li et al. observed that daucosterol exhibits a concentration-dependent lipolysis activity with very high potency. They also hypothesized that doctors could use daucosterol to manage certain types of disease, which can confer medicinal principles [35]. Sashidhara et al. [129] screened for natural resources with hypolipidemic activity in Bauhinia racemosa leaves using high-fat diet (HFD)-fed hamsters. The results showed that Bauhinia racemosa leaf ethanolic extract exhibits a strong hypolipidemic action due to its content of sitosterol-β-D-glucoside and other phytoconstituents, which could act additively or synergistically. The authors also indicated that Bauhinia racemosa leaf alcoholic extract rich in daucosterol could be used as a herbal drug to treat hypercholesterolemia and hypertriglyceridemia, a comprehensive approach to hypercholesterolemia and hypertriglyceridemia dyslipidemia management [129].
In addition, red yeast rice (RYR) used in traditional Chinese medicine could be an important product to prevent hyperlipidemia and dyslipidemia due to its content of more than 101 chemical agents, including daucosterol. According to experiments, RYR has been shown to significantly reduce total cholesterol, triglyceride, and low-density lipoprotein cholesterol (LDL-C) levels and increase high-density lipoprotein cholesterol (HDL-C) levels [130]. The underlying pathways of drug action involve increased mRNA levels of the farnesoid X receptor and peroxisome-proliferator-gamma-activated receptor, key receptors in cholesterol metabolism and bile acid homeostasis [131].
Khan and Hossain extracted two bioactive molecules, β-sitosterol glycoside and scopoletin, from Ipomoea digitate root ethanolic extract. The tuberous root of this plant, mainly used in traditional medicine, has been shown to exhibit hypolipidemic action, potently lowering serum total cholesterol and LDL-C [45]. In the in vivo study performed by Mironova and Kalashnikova [132] on animals suffering from hypercholesterolemia, treatment with β-sitosterol β-d-glucoside reduced β-lipoproteins by 46% and blood cholesterol by 31% while normalizing the cholesterol/phospholipid level, whereas administration of β-daucosterol did not affect the β-lipoprotein and cholesterol levels in the blood serum in control animals. In experimental hypercholesterolemia, the suspected mechanism of lipid-lowering activity of the drug is the stimulation of phospholipid synthesis [132]. Further studies investigating the hypolipidemic capacity of daucosterol are needed to clarify the different pathways implicated in this pharmacological property to prevent the various diseases associated with lipid disorders.

5. Limitations and Perspectives

Here we have reported the different natural sources and the benefits and pharmacological properties of daucosterol. According to the literature, this molecule exhibited remarkable biological activities in vitro and in vivo, and therefore may be a key candidate in drug development. Indeed, its anticancer and neuroprotective effects are promising; elucidating these different mechanisms of action may play a crucial role in the development of new drugs. However, different limitations of this study should be mentioned. The first is the lack of data related to pharmacodynamic action of daucosterol, which makes it difficult to trace and create clear molecular target pathways regarding its mechanism of action. The second limitation is related to the lack of studies assesses the toxicity and safety of daucosterol in vivo. Moreover, understanding the pharmacokinetics and pharmacodynamics of daucosterol is necessary for its incorporation as a drug to treat many diseases; on the other hand, toxicological investigations are needed to validate its safety; the results of these investigations can also inform many approaches for the development of new drugs. Moreover, the investigation of daucosterol in combination with other drugs is also limited because no work has been reported in this direction.

Author Contributions

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

Funding

This research received no external funding.

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. Cao, X.; Zhuang, H.; Jiang, H. Study on the Chemical Constituents of a Plant in the Soil of Taibai Mountain. Int. J. Sci. 2021, 8, 4. [Google Scholar]
  2. Stefani, T.; Morales-San Claudio, P.D.C.; Rios, M.Y.; Aguilar-Guadarrama, A.B.; González-Maya, L.; Sánchez-Carranza, J.N.; González-Ferrara, M.; Camacho-Corona, M. del R. UPLC–QTOF–MS Analysis of Cytotoxic and Antibacterial Extracts of Hechtia Glomerata Zucc. Nat. Prod. Res. 2022, 36, 644–648. [Google Scholar] [CrossRef] [PubMed]
  3. Al-Haidari, R.A.; Al-Oqail, M.M. New Benzoic Acid Derivatives from Cassia Italica Growing in Saudi Arabia and Their Antioxidant Activity. Saudi Pharm. J. 2020, 28, 1112–1117. [Google Scholar] [CrossRef]
  4. Quach, N.T.; Nguyen, Q.H.; Vu, T.H.N.; Le, T.T.H.; Ta, T.T.T.; Nguyen, T.D.; Van Doan, T.; Van Nguyen, T.; Dang, T.T.; Nguyen, X.C.; et al. Plant-Derived Bioactive Compounds Produced by Streptomyces Variabilis LCP18 Associated with Litsea cubeba (Lour.) Pers as Potential Target to Combat Human Pathogenic Bacteria and Human Cancer Cell Lines. Braz. J. Microbiol. 2021, 52, 1215–1224. [Google Scholar] [CrossRef]
  5. Wan, J.; Gong, X.; Wen, C.; Wei, Y.; Kong, L.; Han, B.; Ouyang, Z. First Systematic Analysis for Chemical Composition by HPLC-ESI-MSn and Antioxidant Activity Comparison of Dendrobium Huoshanense and Dendrobium Officinale. Res. Sq. 2020; in review. [Google Scholar] [CrossRef]
  6. Shomirzoeva, O.; Li, J.; Numonov, S.; Atolikshoeva, S.; Aisa, H.A. Chemical Components of Hyssopus Cuspidatus Boriss.: Isolation and Identification, Characterization by HPLC-DAD-ESI-HRMS/MS, Antioxidant Activity and Antimicrobial Activity. Nat. Prod. Res. 2020, 34, 534–540. [Google Scholar] [CrossRef]
  7. Bouzghaia, B.; Moussa, M.T.B.; Goudjil, R.; Harkat, H.; Pale, P. Chemical Composition, in Vitro Antioxidant and Antibacterial Activities of Centaurea Resupinata Subsp. Dufourii (Dostál) Greuter. Nat. Prod. Res. 2021, 35, 4734–4739. [Google Scholar] [CrossRef]
  8. Besbas, S.; Mouffouk, S.; Haba, H.; Marcourt, L.; Wolfender, J.-L.; Benkhaled, M. Chemical Composition, Antioxidant, Antihemolytic and Anti-Inflammatory Activities of Ononis mitissima L. Phytochem. Lett. 2020, 37, 63–69. [Google Scholar] [CrossRef]
  9. Abdollahnezhad, H.; Bahadori, M.B.; Pourjafar, H.; Movahhedin, N. Purification, Characterization, and Antioxidant Activity of Daucosterol and Stigmasterol from Prangos Ferulacea. Lett. Appl. Biosci. 2021, 10, 2174–2180. [Google Scholar]
  10. Murni, A.; Yuliansih, P.; Mohamad, K.; Kita, M.; Hanif, N. A Glucosylated Steroid from the Active Fraction of Indonesian Ficus Deltoidea Leaves. AIP Conf. Proc. 2020, 2243, 030015. [Google Scholar] [CrossRef]
  11. Park, M.-K.; Cho, S.; Ahn, T.-K.; Kim, D.-H.; Kim, S.-Y.; Lee, J.-W.; Kim, J.-I.; Seo, E.-W.; Son, K.-H.; Lim, J.-H. Immunomodulatory Effects of β-sitosterol and Daucosterol Isolated from Dioscorea batatas on LPS-stimulated RAW 264.7 and TK-1 Cells. J. Life Sci. 2020, 30, 359–369. [Google Scholar] [CrossRef]
  12. Kavtaradze, N.; Alaniya, M.; Masullo, M.; Cerulli, A.; Piacente, S. New Flavone Glycosides from Astragalus Tanae Endemic to Georgia. Chem. Nat. Compd. 2020, 56, 70–74. [Google Scholar] [CrossRef]
  13. Ibekwe, N.N.; Akoje, V.O.; Igoli, J.O. Phytochemical Constituents of the Leaves of Landolphia Owariensis. Trop. J. Nat. Prod. Res. 2019, 3, 261–264. [Google Scholar] [CrossRef]
  14. Dehghan, H.; Salehi, P.; Amiri, M.S. Bioassay-Guided Purification of α-Amylase, α-Glucosidase Inhibitors and DPPH Radical Scavengers from Roots of Rheum Turkestanicum. Ind. Crops Prod. 2018, 117, 303–309. [Google Scholar] [CrossRef]
  15. Dehghan, H. Isolation of 17 Antidiabetic Compounds from Two Iranian Plants. Org. Agric. 2019. Available online: http://herb.bonyadhamayesh.ir/fa/ (accessed on 29 April 2022).
  16. Duong, N.T.; Vinh, P.D.; Thuong, P.T.; Hoai, N.T.; Thanh, L.N.; Bach, T.T.; Nam, N.H.; Anh, N.H. Xanthine Oxidase Inhibitors from Archidendron Clypearia (Jack.) I.C. Nielsen: Results from Systematic Screening of Vietnamese Medicinal Plants. Asian Pac. J. Trop. Med. 2017, 10, 549–556. [Google Scholar] [CrossRef] [PubMed]
  17. Cho, E.J.; Choi, J.Y.; Lee, K.H.; Lee, S. Isolation of Antibacterial Compounds from Parasenecio Pseudotaimingasa. Hortic. Environ. Biotechnol. 2012, 53, 561–564. [Google Scholar] [CrossRef]
  18. Uddin, G.; Siddiqui, B.S.; Alam, M.; Sadat, A.; Ahmad, A.; Uddin, A. Chemical Constituents and Phytotoxicity of Solvent Extracted Fractions of Stem Bark of Grewia Optiva Drummond Ex Burret. Middle-East. J. Sci. Res. 2011, 8, 85–91. [Google Scholar]
  19. Ma, C.; Wang, W.; Chen, Y.-Y.; Liu, R.-N.; Wang, R.-F.; Du, L.-J. Neuroprotective and Antioxidant Activity of Compounds from the Aerial Parts of Dioscorea Opposita. J. Nat. Prod. 2005, 68, 1259–1261. [Google Scholar] [CrossRef]
  20. Luo, W.; Zhao, M.; Yang, B.; Shen, G.; Rao, G. Identification of Bioactive Compounds in Phyllenthus emblica L. Fruit and Their Free Radical Scavenging Activities. Food Chem. 2009, 114, 499–504. [Google Scholar] [CrossRef]
  21. Elkhayat, E.S.; Ibrahim, S.R.M.; Aziz, M.A. Portulene, a New Diterpene from Portulaca oleracea L. J. Asian Nat. Prod. Res. 2008, 10, 1039–1043. [Google Scholar] [CrossRef]
  22. Hong, Y.; Qiao, Y.; Lin, S.; Jiang, Y.; Chen, F. Characterization of Antioxidant Compounds in Eriobotrya Fragrans Champ Leaf. Sci. Hortic. 2008, 118, 288–292. [Google Scholar] [CrossRef]
  23. Sultana, N.; Afolayan, A.J. A Novel Daucosterol Derivative and Antibacterial Activity of Compounds from Arctotis Arctotoides. Nat. Prod. Res. 2007, 21, 889–896. [Google Scholar] [CrossRef]
  24. Nguedia, M.Y.; Tueche, A.B.; Yaya, A.J.G.; Yadji, V.; Ndinteh, D.T.; Njamen, D.; Zingue, S. Daucosterol from Crateva Adansonii DC (Capparaceae) reduces 7,12-dimethylbenz(a)anthracene-induced mammary tumors in Wistar rats. Environ. Toxicol. 2020, 35, 1125–1136. [Google Scholar] [CrossRef]
  25. Han, B.; Jiang, L.; Jiang, P.; Zhou, D.; Jia, X.; Li, X.; Ye, X. Daucosterol Linolenate from Sweet Potato Suppresses MCF7-Xenograft-Tumor Growth through Regulating PI3K/AKT Pathway. Planta Med. 2020, 86, 767–775. [Google Scholar] [CrossRef]
  26. Rajavel, T.; Banu Priya, G.; Suryanarayanan, V.; Singh, S.K.; Pandima Devi, K. Daucosterol Disturbs Redox Homeostasis and Elicits Oxidative-Stress Mediated Apoptosis in A549 Cells via Targeting Thioredoxin Reductase by a P53 Dependent Mechanism. Eur. J. Pharmacol. 2019, 855, 112–123. [Google Scholar] [CrossRef]
  27. Chung, M.J.; Lee, S.; Park, Y.I.; Kwon, K.H. Antioxidative and Neuroprotective Effects of Extract and Fractions from Adenophora triphylla. J. Korean Soc. Food Sci. Nutr. 2016, 45, 1580–1588. [Google Scholar] [CrossRef]
  28. Jiang, Y.; Zhang, L.; Rupasinghe, H. The Anticancer Properties of Phytochemical Extracts from Salvia Plants. Bot. Targets Ther. 2016, 6, 25–44. [Google Scholar] [CrossRef] [Green Version]
  29. Ji, Z.-H.; Xu, Z.-Q.; Zhao, H.; Yu, X.-Y. Neuroprotective Effect and Mechanism of Daucosterol Palmitate in Ameliorating Learning and Memory Impairment in a Rat Model of Alzheimer’s Disease. Steroids 2017, 119, 31–35. [Google Scholar] [CrossRef]
  30. Zhang, H.; Song, Y.; Feng, C. Improvement of Cerebral Ischemia/Reperfusion Injury by Daucosterol Palmitate-Induced Neuronal Apoptosis Inhibition via PI3K/Akt/MTOR Signaling Pathway. Metab. Brain Dis. 2020, 35, 1035–1044. [Google Scholar] [CrossRef]
  31. Jang, J.; Kim, S.-M.; Yee, S.-M.; Kim, E.-M.; Lee, E.-H.; Choi, H.-R.; Lee, Y.-S.; Yang, W.-K.; Kim, H.-Y.; Kim, K.-H.; et al. Daucosterol Suppresses Dextran Sulfate Sodium (DSS)-Induced Colitis in Mice. Int. Immuno Pharmacol. 2019, 72, 124–130. [Google Scholar] [CrossRef]
  32. Gu, Y.; Yang, X.; Shang, C.; Phuong Thao, T.T.; Koyama, T. Inhibitory Properties of Saponin from Eleocharis Dulcis Peel against α-Glucosidase. RSC Adv. 2021, 11, 15400–15409. [Google Scholar] [CrossRef]
  33. Gu, Y.; Yang, X.; Shang, C.; Thao, T.T.P.; Koyama, T. Inhibition and Interactions of Alpha-Amylase by Daucosterol from the Peel of Chinese Water Chestnut (Eleocharis dulcis). Food Funct. 2021, 12, 8411–8424. [Google Scholar] [CrossRef]
  34. Lee, J.-H.; Lee, J.Y.; Park, J.H.; Jung, H.S.; Kim, J.S.; Kang, S.S.; Kim, Y.S.; Han, Y. Immunoregulatory Activity by Daucosterol, a β-Sitosterol Glycoside, Induces Protective Th1 Immune Response against Disseminated Candidiasis in Mice. Vaccine 2007, 25, 3834–3840. [Google Scholar] [CrossRef]
  35. Li, K.; Lee, M.L.; Que, L.; Li, M.; Kang, J.S.; Choi, Y.H.; Kim, K.M.; Jung, J.-C.; Hwang, D.Y.; Choi, Y.W. Lipolysis Effect of Daucosterol Isolated from Mulberry (Morus alba) Leaves. J. Life Sci. 2017, 27, 1500–1506. [Google Scholar] [CrossRef]
  36. Esmaeili, M.A.; Farimani, M.M. Inactivation of PI3K/Akt Pathway and Upregulation of PTEN Gene Are Involved in Daucosterol, Isolated from Salvia Sahendica, Induced Apoptosis in Human Breast Adenocarcinoma Cells. S. Afr. J. Bot. 2014, 93, 37–47. [Google Scholar] [CrossRef] [Green Version]
  37. Han, B.; Jiang, P.; Liu, W.; Xu, H.; Li, Y.; Li, Z.; Ma, H.; Yu, Y.; Li, X.; Ye, X. Role of Daucosterol Linoleate on Breast Cancer: Studies on Apoptosis and Metastasis. J. Agric. Food Chem. 2018, 66, 6031–6041. [Google Scholar] [CrossRef]
  38. Wang, G.-Q.; Gu, J.-F.; Gao, Y.-C.; Dai, Y.-J. Daucosterol Inhibits Colon Cancer Growth by Inducing Apoptosis, Inhibiting Cell Migration and Invasion and Targeting Caspase Signalling Pathway. Bangladesh J Pharm. 2016, 11, 395. [Google Scholar] [CrossRef] [Green Version]
  39. Mir, S.A.; Dar, L.A.; Ali, T.; Kareem, O.; Rashid, R.; Khan, N.A.; Chashoo, I.A.; Bader, G.N. A Review on Its Phytochemistry and Pharmacology. Edible Plants Health Dis. 2022, 327–348. [Google Scholar] [CrossRef]
  40. Zeng, J.; Liu, X.; Li, X.; Zheng, Y.; Liu, B.; Xiao, Y. Daucosterol Inhibits the Proliferation, Migration, and Invasion of Hepatocellular Carcinoma Cells via Wnt/β-Catenin Signaling. Molecules 2017, 22, 862. [Google Scholar] [CrossRef]
  41. Chung, M.J.; Lee, S.; Park, Y.I.; Lee, J.; Kwon, K.H. Neuroprotective Effects of Phytosterols and Flavonoids from Cirsium Setidens and Aster Scaber in Human Brain Neuroblastoma SK-N-SH Cells. Life Sci. 2016, 148, 173–182. [Google Scholar] [CrossRef]
  42. Sheng, Z.; Dai, H.; Pan, S.; Wang, H.; Hu, Y.; Ma, W. Isolation and Characterization of an α-Glucosidase Inhibitor from Musa spp. (Baxijiao) Flowers. Molecules 2014, 19, 10563–10573. [Google Scholar] [CrossRef] [Green Version]
  43. Yang, N.; Wang, L.; Zhang, Y. Immunological Activities of Components from Leaves of Liriodendron Chinensis. Chin. Herb. Med. 2015, 7, 279–282. [Google Scholar] [CrossRef]
  44. Taheri, Y.; Quispe, C.; Herrera-Bravo, J.; Sharifi-Rad, J.; Ezzat, S.M.; Merghany, R.M.; Cho, W.C. Urtica dioica-derived phytochemicals for pharmacological and therapeutic applications. Evid.-Based Complem. Alter. Med. 2022, 2022, 4024331. [Google Scholar] [CrossRef]
  45. Jain, V.; Verma, S.K.; Katewa, S.S. Therapeutic Validation of Ipomoea digitata Tuber (Ksheervidari) for its Effect on Cardio-Vascular Risk Parameters. Ind. J. Trad. Knowl. 2011, 617–623. Available online: http://hdl.handle.net/123456789/12823 (accessed on 29 April 2022).
  46. Torres-Ortiz, D.A.; Eloy, R.; Moustapha, B.; César, I.-A.; Edmundo, M.-S.; Jesús Eduardo, C.-R.; Dulce María, R.-P. Vasorelaxing Effect and Possible Chemical Markers of the Flowers of the Mexican Crataegus Gracilior. Nat. Prod. Res. 2020, 34, 3522–3525. [Google Scholar] [CrossRef]
  47. Hernández-Pérez, A.; Bah, M.; Ibarra-Alvarado, C.; Rivero-Cruz, J.F.; Rojas-Molina, A.; Rojas-Molina, J.I.; Cabrera-Luna, J.A. Aortic relaxant activity of Crataegus gracilior Phipps and identification of some of its chemical constituents. Molecules 2014, 19, 20962–20974. [Google Scholar] [CrossRef]
  48. Zingue, S.; Gbaweng Yaya, A.J.; Michel, T.; Ndinteh, D.T.; Rutz, J.; Auberon, F.; Maxeiner, S.; Chun, F.K.-H.; Tchinda, A.T.; Njamen, D.; et al. Bioguided Identification of Daucosterol, a Compound That Contributes to the Cytotoxicity Effects of Crateva Adansonii DC (Capparaceae) to Prostate Cancer Cells. J. Ethnopharmacol. 2020, 247, 112251. [Google Scholar] [CrossRef]
  49. Chen, L.; Xin, X.; Feng, H.; Li, S.; Cao, Q.; Wang, X.; Vriesekoop, F. Isolation and Identification of Anthocyanin Component in the Fruits of Acanthopanax Sessiliflorus (Rupr. and Maxim.) Seem. by Means of High Speed Counter Current Chromatography and Evaluation of Its Antioxidant Activity. Molecules 2020, 25, 1781. [Google Scholar] [CrossRef] [Green Version]
  50. CHEN Da-xia1, 2 Compare and Cluster Analysis of Six Chemical Constituents in Dipsacus Medicinal Plants. Nat. Prod. Res. Dev. 2019, 30, 132. [CrossRef]
  51. Zhang, X.; Wang, K.-W.; Wang, H. Chemical Constituents of Streptocaulon Griffithii. Chem. Nat. Compd. 2018, 54, 803–805. [Google Scholar] [CrossRef]
  52. Kuang, Y.; Yang, S.X.; Sampietro, D.A.; Zhang, X.F.; Tan, J.; Gao, Q.X.; Liu, H.W.; Ni, Q.X.; Zhang, Y.Z. Phytotoxicity of Leaf Constituents from Bamboo (Shibataea chinensis Nakai) on Germination and Seedling Growth of Lettuce and Cucumber. Allelopath. J. 2017, 40, 133–142. [Google Scholar] [CrossRef]
  53. Numonov, S.; Edirs, S.; Bobakulov, K.; Qureshi, M.N.; Bozorov, K.; Sharopov, F.; Setzer, W.N.; Zhao, H.; Habasi, M.; Sharofova, M.; et al. Evaluation of the Antidiabetic Activity and Chemical Composition of Geranium Collinum Root Extracts—Computational and Experimental Investigations. Molecules 2017, 22, 983. [Google Scholar] [CrossRef]
  54. Yang, D.; Zhang, C.; Liu, X.-X.; Zhang, Y.; Wang, K.; Cheng, Z.-Q. Chemical Constituents and Antioxidant Activity of Lasianthus Hartii. Chem. Nat. Compd. 2017, 53, 390–393. [Google Scholar] [CrossRef]
  55. Bahadori, M.B.; Dinparast, L.; Valizadeh, H.; Farimani, M.M.; Ebrahimi, S.N. Bioactive Constituents from Roots of Salvia Syriaca L.: Acetylcholinesterase Inhibitory Activity and Molecular Docking Studies. S. Afr. J. Bot. 2016, 106, 1–4. [Google Scholar] [CrossRef]
  56. Bensouici, C.; Kabouche, A.; Karioti, A.; Öztürk, M.; Duru, M.E.; Bilia, A.R.; Kabouche, Z. Compounds from Sedum Caeruleum with Antioxidant, Anticholinesterase, and Antibacterial Activities. Pharm. Biol. 2016, 54, 174–179. [Google Scholar] [CrossRef]
  57. Galala, A.A.; Sallam, A.; Abdel-Halim, O.B.; Gedara, S.R. New Ent-Kaurane Diterpenoid Dimer from Pulicaria Inuloides. Nat. Prod. Res. 2016, 30, 2468–2475. [Google Scholar] [CrossRef]
  58. Asghari, B.; Salehi, P.; Farimani, M.M.; Ebrahimi, S.N. α-Glucosidase Inhibitors from Fruits of Rosa canina L. Rec. Nat. Prod. 2015, 9, 276–283. [Google Scholar]
  59. Kim, M.A.; Kim, M.J.; Chun, W.; Kwon, Y. Chemical Constituents and Their Acetylcholinesterase Inhibitory Activity of Underground Parts of Clematis heracleifolia. Korean J. Pharmacogn. 2015, 46, 6–11. [Google Scholar]
  60. Delnavazi, M.-R.; Hadjiakhoondi, A.; Delazar, A.; Ajani, Y.; Tavakoli, S.; Yassa, N. Phytochemical and Antioxidant Investigation of the Aerial Parts of Dorema Glabrum Fisch. and C.A. Mey. Iran. J. Pharm. Res. 2015, 14, 925–931. [Google Scholar]
  61. Lee, J.; Weon, J.B.; Yun, B.-R.; Eom, M.R.; Ma, C.J. Simultaneous Determination Three Phytosterol Compounds, Campesterol, Stigmasterol and Daucosterol in Artemisia Apiacea by High Performance Liquid Chromatography-Diode Array Ultraviolet/Visible Detector. Pharm. Mag. 2015, 11, 297–303. [Google Scholar] [CrossRef] [Green Version]
  62. Li, X.; Wang, T.; Zhou, B.; Gao, W.; Cao, J.; Huang, L. Chemical Composition and Antioxidant and Anti-Inflammatory Potential of Peels and Flesh from 10 Different Pear Varieties (Pyrus spp.). Food Chem. 2014, 152, 531–538. [Google Scholar] [CrossRef]
  63. Wang, C.; Wang, J.; Gao, M.; Gao, P.; Gao, D.; Zhang, H.; Qiao, M. Radix Ranunculi ternati: Review of its chemical constituents, pharmacology, quality control and clinical applications. J. Pharm Pharmacol. 2022. [Google Scholar] [CrossRef]
  64. Bekir, J.; Mars, M.; Vicendo, P.; Fterrich, A.; Bouajila, J. Chemical Composition and Antioxidant, Anti-Inflammatory, and Antiproliferation Activities of Pomegranate (Punica granatum) Flowers. J. Med. Food 2013, 16, 544–550. [Google Scholar] [CrossRef]
  65. Loganayaki, N.; Siddhuraju, P.; Manian, S. Antioxidant Activity and Free Radical Scavenging Capacity of Phenolic Extracts from Helicteres Isora L. and Ceiba Pentandra L. J. Food Sci. Technol. 2013, 50, 687–695. [Google Scholar] [CrossRef] [Green Version]
  66. Wei, J.; Zhang, Y.; Kang, W. Antioxidant and A-Glucosidase Inhibitory Compounds in Lysimachia Clethroides. AJPP 2012, 6, 3230–3234. [Google Scholar] [CrossRef] [Green Version]
  67. Singh, R. Chemical investigation and in vitro cytotoxic activity of randia dumetorum lamk bark. Int. J. Chem. Sci. 2012, 10, 1374–1382. [Google Scholar]
  68. Wang, L.; Wu, M.; Huang, J.; Wang, J.; Chen, Y.; Yin, B. Chemical Constituents of Eria Spicata. Chem. Nat. Compd. 2012, 48, 168–169. [Google Scholar] [CrossRef]
  69. Guo, J.; Chen, J.; Lin, L.; Xu, F. Five Chemical Constituents of the Ethyl Acetate Fraction from Ethanol Extract of Semen Litchi. JMPR 2012, 6, 168–170. [Google Scholar] [CrossRef]
  70. Zhang, H.; Yan, X.; Jiang, Y.; Han, Y.; Zhou, Y. The Extraction, Identification and Quantification of Hypoglycemic Active Ingredients from Stinging Nettle (Urtica angustifolia). Afr. J. Biotechnol. 2011, 10, 9428–9437. [Google Scholar] [CrossRef]
  71. Saeidnia, S.; Gohari, A.R.; Malmir, M.; Moradi, A.F.; Ajani, Y. Tryptophan and Sterols from Salvia limbata. J. Med. Plants 2011, 10, 41–47. [Google Scholar]
  72. Huh, G.-W.; Park, J.-H.; Shrestha, S.; Lee, Y.-H.; Ahn, E.-M.; Kang, H.-C.; Baek, N.-I. Sterols from Lindera Glauca Blume Stem Wood. J. Appl. Biol. Chem. 2011, 54, 309–312. [Google Scholar] [CrossRef]
  73. Gao, C.; Lu, Y.; Tian, C.; Xu, J.; Guo, X.; Zhou, R.; Hao, G. Main Nutrients, Phenolics, Antioxidant Activity, DNA Damage Protective Effect and Microstructure of Sphallerocarpus Gracilis Root at Different Harvest Time. Food Chem. 2011, 127, 615–622. [Google Scholar] [CrossRef]
  74. Kang, W.-Y.; Song, Y.-L.; Zhang, L. α-Glucosidase Inhibitory and Antioxidant Properties and Antidiabetic Activity of Hypericum Ascyron L. Med. Chem. Res. 2011, 20, 809–816. [Google Scholar] [CrossRef]
  75. Koo, Y.K.; Kim, J.M.; Koo, J.Y.; Kang, S.S.; Bae, K.; Kim, Y.S.; Chung, J.-H.; Yun-Choi, H.S. Platelet Anti-Aggregatory and Blood Anti-Coagulant Effects of Compounds Isolated from Paeonia Lactiflora and Paeonia Suffruticosa. Die Pharm. Int. J. Pharm. Sci. 2010, 65, 624–628. [Google Scholar] [CrossRef]
  76. Nur, S.M.; Soemitro, S.; Bahti, H.H.; Tarigan, P.; Shen, Y.M.; Rumampuk, R.J. Chemical Constituents from the Steam Bark of Alangium Kurzi, Craib. Indones. J. Chem. 2010, 1, 128–130. [Google Scholar] [CrossRef]
  77. Olaleye, M.T.; Akinmoladun, A.C.; Ogunboye, A.A.; Akindahunsi, A.A. Antioxidant Activity and Hepatoprotective Property of Leaf Extracts of Boerhaavia Diffusa Linn against Acetaminophen-Induced Liver Damage in Rats. Food Chem. Toxicol. 2010, 48, 2200–2205. [Google Scholar] [CrossRef]
  78. Park, J.-H.; Kwon, S.-J. Isolation of Daucosterol and Naphthalene glucoside from Seeds of Cassia mimosoides var. nomame Makino. Korean J. Plant Resour. 2009, 22, 26–30. [Google Scholar]
  79. León, F.; Habib, E.; Adkins, J.E.; Furr, E.B.; McCurdy, C.R.; Cutler, S.J. Phytochemical Characterization of the Leaves of Mitragyna Speciosa Grown in USA. Nat. Prod. Commun. 2009, 4, 1934578X0900400705. [Google Scholar] [CrossRef] [Green Version]
  80. Jeong, J.-S.; Lee, J.-H.; Lee, S.-H.; Kang, S.-S.; Jeong, C.-S. Suppressive Actions of Astragali Radix (AR) Ethanol Extract and Isolated Astragaloside I on HCl/Ethanol-Induced Gastric Lesions. Biomol. Ther. 2009, 17, 62–69. [Google Scholar] [CrossRef] [Green Version]
  81. Chai, X.-Y.; Ren, H.-Y.; Xu, Z.-R.; Bai, C.-C.; Zhou, F.-R.; Ling, S.-K.; Pu, X.-P.; Li, F.-F.; Tu, P.-F. Investigation of Two Flacourtiaceae Plants: Bennettiodendron Leprosipes and Flacourtia Ramontchi. Planta Med. 2009, 75, 1246–1252. [Google Scholar] [CrossRef] [PubMed]
  82. Mavar-Manga, H.; Haddad, M.; Pieters, L.; Baccelli, C.; Penge, A.; Quetin-Leclercq, J. Anti-Inflammatory Compounds from Leaves and Root Bark of Alchornea Cordifolia (Schumach. and Thonn.) Müll. Arg. J. Ethnopharmacol. 2008, 115, 25–29. [Google Scholar] [CrossRef] [PubMed]
  83. Wang, G.-C. Chemical Constituents from Flueggea Virosa: Chemical Constituents from Flueggea Virosa. Chin. J. Nat. Med. 2008, 6, 251–253. [Google Scholar] [CrossRef]
  84. Chen, Y.; Zhao, Y.; Hu, Y.; Wang, L.; Ding, Z.; Liu, Y.; Wang, J. Isolation of 5-Hydroxypyrrolidin-2-One and Other Constituents from the Young Fronds of Pteridium Aquilinum. J. Nat. Med. 2008, 62, 358–359. [Google Scholar] [CrossRef] [PubMed]
  85. Bang, M.-H.; Lee, D.-Y.; Oh, Y.-J.; Han, M.-W.; Yang, H.-J.; Chung, H.-G.; Jeong, T.-S.; Lee, K.-T.; Choi, M.-S.; Baek, N.-I. Development of Biologically Active Compounds from Edible Plant Sources XXII. Isolation of Indoles from the Roots of Brassica campestris ssp rapa and their hACAT Inhibitory Activity. Appl. Biol. Chem. 2008, 51, 65–69. [Google Scholar]
  86. Zhang, T.; Chen, Y.-M.; Zhang, G.-L. Novel Neolignan from Penthorum Chinense. J. Integr. Plant Biol. 2007, 49, 1611–1614. [Google Scholar] [CrossRef]
  87. Rao, G.-X.; Gao, Y.-L.; Lin, Y.-P.; Xiao, Y.-L.; Li, S.-H.; Sun, H.-D. Chemical Constituents of Selinum Cryptotaenium. J. Asian Nat. Prod. Res. 2006, 8, 273–275. [Google Scholar] [CrossRef]
  88. Lin, P.; Li, S.; Wang, S.; Yang, Y.; Shi, J. A Nitrogen-Containing 3-Alkyl-1,4-Benzoquinone and a Gomphilactone Derivative from Embelia Ribes. J. Nat. Prod. 2006, 69, 1629–1632. [Google Scholar] [CrossRef] [PubMed]
  89. Wang, R.; Wang, W.; Wang, L.; Liu, R.; Ding, Y.; Du, L. Constituents of the Flowers of Punica granatum. Fitoterapia 2006, 77, 534–537. [Google Scholar] [CrossRef]
  90. Pungitore, C.R.; García, M.; Gianello, J.C.; Sosa, M.E.; Tonn, C.E. Insecticidal and Antifeedant Effects of Junellia Aspera (Verbenaceae) Triterpenes and Derivatives on Sitophilus Oryzae (Coleoptera: Curculionidae). J. Stored Prod. Res. 2005, 41, 433–443. [Google Scholar] [CrossRef]
  91. Yu, D.-H.; Bao, Y.-M.; Wei, C.-L.; An, L.-J. Studies of chemical constituents and their antioxidant activities from Astragalus mongholicus Bunge. Biomed. Environ. Sci. 2005, 18, 297–301. [Google Scholar]
  92. Yang, M.-F.; Li, Y.-Y.; Li, B.-G.; Zhang, G.-L. A New Monoterpene Glycoside from Hemiphragma heterophyllum. J. Integr. Plant Biol. 2004, 46, 1454. [Google Scholar]
  93. Wang, R.-F.; Xie, W.-D.; Zhang, Z.; Xing, D.-M.; Ding, Y.; Wang, W.; Ma, C.; Du, L.-J. Bioactive Compounds from the Seeds of Punica Granatum (Pomegranate). J. Nat. Prod. 2004, 67, 2096–2098. [Google Scholar] [CrossRef]
  94. Klimek, B.; Modnicki, D. Terpenoids and sterols from Nepeta cataria L. var. citriodora (Lamiaceae). Acta Pol. Pharm. 2005, 62, 231–235. [Google Scholar] [PubMed]
  95. Bi, Z.-M.; Wang, Z.-T.; Xu, L.-S. Chemical Constituents of Dendrobium Moniliforme. Acta Bot. Sin. Engl. Ed. 2004, 46, 124–126. [Google Scholar]
  96. Li, P.; Feng, Z.X.; Ye, D.; Huan, W.; Da Gang, W.; Dong, L.X. Chemical Constituents from the Whole Plant of Euphorbia Altotibetic. Helv. Chim. Acta 2003, 86, 2525–2532. [Google Scholar] [CrossRef]
  97. Song, E.-K.; Kim, J.-H.; Kim, J.-S.; Cho, H.; Nan, J.-X.; Sohn, D.-H.; Ko, G.-I.; Oh, H.; Kim, Y.-C. Hepatoprotective Phenolic Constituents of Rhodiola Sachalinensis on Tacrine-Induced Cytotoxicity in Hep G2 Cells. Phytother. Res. 2003, 17, 563–565. [Google Scholar] [CrossRef] [PubMed]
  98. Xiang, W.; Jiang, B.; Li, X.-M.; Zhang, H.-J.; Zhao, Q.-S.; Li, S.-H.; Sun, H.-D. Constituents of Gnetum Montanum. Fitoterapia 2002, 73, 40–42. [Google Scholar] [CrossRef]
  99. Lee, S.; Kim, K.S.; Jang, J.M.; Park, Y.; Kim, Y.B.; Kim, B.-K. Phytochemical Constituents from the Herba of Artemisia Apiacea. Arch. Pharm. Res. 2002, 25, 285–288. [Google Scholar] [CrossRef]
  100. Meng, J.C.; Hu, Y.F.; Chen, J.H.; Tan, R.X. Antifungal Highly Oxygenated Guaianolides and Other Constituents from Ajania Fruticulosa. Phytochemistry 2001, 58, 1141–1145. [Google Scholar] [CrossRef]
  101. Jin-rui, C.; Lin-gang, Q.; Min, L.; Si-ping, J.; Yu, K.; Zhong-wu, M.; Guan-fu, H. Studies on the Chemical Constituents of Rhodiola Fastigita S. H. Fu. J. Integr. Plant Biol. 1991, 33. Available online: https://www.jipb.net/EN/Y1991/V33/I1/ (accessed on 12 October 2021).
  102. Ling-yi, K.; Zhi-da, M.; Jian-xia, S.; Rui, F. Chemical Constituents from Roots of Jatropha Curcas. J. Integr. Plant Biol. 1996, 38. Available online: https://www.jipb.net/EN/Y1996/V38/I2/ (accessed on 12 October 2021).
  103. Tan, R.X.; Wolfender, J.-L.; Ma, W.G.; Zhang, L.X.; Hostettmann, K. Secoiridoids and Antifungal Aromatic Acids from Gentiana Algida. Phytochemistry 1996, 41, 111–116. [Google Scholar] [CrossRef]
  104. Tan, R.X.; Kong, L.D. Secoiridoids from Gentiana Siphonantha. Phytochemistry 1997, 46, 1035–1038. [Google Scholar] [PubMed]
  105. Tan, R.X.; Tang, H.Q.; Hu, J.; Shuai, B. Lignans and Sesquiterpene Lactones from Artemisia Sieversiana and Inula Racemosa. Phytochemistry 1998, 49, 157–161. [Google Scholar] [CrossRef]
  106. Wen-Wu, L.I.; Bo-Gang, L.I.; Li-Sheng, D.; Yao-Zu, C. The Chemical Constituents of Rabdosia Coetsa. J. Integr. Plant Biol. 1998, 40. Available online: https://www.jipb.net/EN/Y1998/V40/I5/ (accessed on 12 October 2021).
  107. Fang, X.; Nanayakkara, N.P.D.; Phoebe, C.H.; Pezzuto, J.M.; Kinghorn, A.D.; Farnsworth, N.R. Plant Anticancer Agents XXXVII. Constituents of Amanoa Oblongifolia. Planta Med. 1985, 51, 346–347. [Google Scholar] [CrossRef]
  108. Kurkin, V.A.; Zapesochnaya, G.G.; Shchavlinskii, A.N. Terpenoids of the Rhizomes OfRhodiola Rosea. Chem. Nat. Compd. 1985, 21, 593–597. [Google Scholar] [CrossRef]
  109. Elyakova, L.A.; Dzizenko, A.K.; Sova, V.V.; Elyakov, G.B. The Isolation of Daucosterol FromAcanthopanax Sessiliflorum. Chem. Nat. Compd. 1966, 2, 26–27. [Google Scholar] [CrossRef]
  110. Zhao, C.; She, T.; Wang, L.; Su, Y.; Qu, L.; Gao, Y.; Xu, S.; Cai, S.; Shou, C. Daucosterol Inhibits Cancer Cell Proliferation by Inducing Autophagy through Reactive Oxygen Species-Dependent Manner. Life Sci. 2015, 137, 37–43. [Google Scholar] [CrossRef] [PubMed]
  111. Hou, J.P.; Wu, H.; Wang, Y.; Weng, X.C. Isolation of Some Compounds from Nutmeg and Their Antioxidant Activities. Czech J. Food Sci. 2012, 30, 164–170. [Google Scholar] [CrossRef] [Green Version]
  112. Rajavel, T.; Mohankumar, R.; Archunan, G.; Ruckmani, K.; Devi, K.P. Beta Sitosterol and Daucosterol (Phytosterols Identified in Grewia Tiliaefolia) Perturbs Cell Cycle and Induces Apoptotic Cell Death in A549 Cells. Sci. Rep. 2017, 7, 3418. [Google Scholar] [CrossRef] [Green Version]
  113. Gao, P.; Huang, X.; Liao, T.; Li, G.; Yu, X.; You, Y.; Huang, Y. Daucosterol Induces Autophagic-Dependent Apoptosis in Prostate Cancer via JNK Activation. BioSci. Trends Adv. Publ. 2019, 13, 160–167. [Google Scholar] [CrossRef] [Green Version]
  114. El Omari, N.; Bakrim, S.; Bakha, M.; Lorenzo, J.M.; Rebezov, M.; Shariati, M.A.; Bouyahya, A. Natural bioactive compounds targeting epigenetic pathways in cancer: A review on alkaloids, terpenoids, quinones, and isothiocyanates. Nutrients 2021, 13, 3714. [Google Scholar] [CrossRef]
  115. Bouyahya, A.; Mechchate, H.; Oumeslakht, L.; Zeouk, I.; Aboulaghras, S.; Balahbib, A.; El Omari, N. The role of epigenetic modifications in human cancers and the use of natural compounds as epidrugs: Mechanistic pathways and pharmacodynamic actions. Biomolecules 2022, 12, 367. [Google Scholar] [CrossRef]
  116. El Omari, N.; Bakha, M.; Imtara, H.; Guaouguaoua, F.E.; Balahbib, A.; Zengin, G.; Bouyahya, A. Anticancer mechanisms of phytochemical compounds: Focusing on epigenetic targets. Environ. Sci. Poll. Res. 2021, 28, 47869–47903. [Google Scholar] [CrossRef]
  117. Jia, M.; Pang, S.; Liu, X.; Mao, Y.; Wu, C.; Zhang, H. Effect of Dietary Phytochemicals on the Progression of Breast Cancer Metastasis Based on the in Vivo Detection of Circulating Tumor Cells. J. Funct. Foods 2020, 65, 103752. [Google Scholar] [CrossRef]
  118. Scholl, S.; Augustin, A.; Loewenstein, A.; Rizzo, S.; Kuppermann, B.D. General pathophysiology of macular edema. Eur. J. Ophthalmol. 2011, 21 (Suppl. S6), 10–19. [Google Scholar] [CrossRef]
  119. Huang, L.-J.; Gao, W.-Y.; Li, X.; Zhao, W.-S.; Huang, L.-Q.; Liu, C.-X. Evaluation of the in Vivo Anti-Inflammatory Effects of Extracts from Pyrus Bretschneideri Rehd. J. Agric. Food Chem. 2010, 58, 8983–8987. [Google Scholar] [CrossRef]
  120. Kim, M.J.; Wang, H.S.; Lee, M.W. Anti-Inflammatory Effects of Fermented Bark of Acanthopanax Sessiliflorus and Its Isolated Compounds on Lipopolysaccharide-Treated RAW 264.7 Macrophage Cells. Evid. Based Complement. Altern. Med. 2020, 2020, e6749425. [Google Scholar] [CrossRef]
  121. Bui Thanh, T.; Vu Duc, L.; Nguyen Thanh, H.; Nguyen Tien, V. In Vitro Antioxidant and Anti-Inflammatory Activities of Isolated Compounds of Ethanol Extract from Sanchezia Speciosa Leonard’s Leaves. J. Basic Clin. Physiol. Pharm. 2017, 28, 79–84. [Google Scholar] [CrossRef]
  122. de Carvalho, A.T.; Paes, M.M.; Cunha, M.S.; Brandão, G.C.; Mapeli, A.M.; Rescia, V.C.; Oesterreich, S.A.; Villas-Boas, G.R. Ethnopharmacology of Fruit Plants: A Literature Review on the Toxicological, Phytochemical, Cultural Aspects, and a Mechanistic Approach to the Pharmacological Effects of Four Widely Used Species. Molecules 2020, 25, 3879. [Google Scholar] [CrossRef]
  123. Randhawa, P.K.; Singh, K.; Singh, N.; Jaggi, A.S. A Review on Chemical-Induced Inflammatory Bowel Disease Models in Rodents. Korean J. Physiol. Pharm. 2014, 18, 279–288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Han, Y.; Lee, J.-H. Berberine Synergy with Amphotericin B against Disseminated Candidiasis in Mice. Biol. Pharm. Bull. 2005, 28, 541–544. [Google Scholar] [CrossRef] [Green Version]
  125. Herzyk, D.J.; Gore, E.R.; Polsky, R.; Nadwodny, K.L.; Maier, C.C.; Liu, S.; Hart, T.K.; Harmsen, A.G.; Bugelski, P.J. Immunomodulatory Effects of Anti-CD4 Antibody in Host Resistance against Infections and Tumors in Human CD4 Transgenic Mice. Infect. Immun. 2001, 69, 1032–1043. [Google Scholar] [CrossRef] [Green Version]
  126. Saeidnia, S.; Ara, L.; Hajimehdipoor, H.; Read, R.W.; Arshadi, S.; Nikan, M. Chemical Constituents of Swertia Longifolia Boiss. with α-Amylase Inhibitory Activity. Res. Pharm. Sci. 2016, 11, 23–32. [Google Scholar]
  127. Ivorra, M.; D’ocon, M.P.; Payá, M.; Villar, Á. Antihyperglycemic and Insulin-Releasing Effects of Beta-Sitosterol 3-Beta-D-Glucoside and Its Aglycone, Beta-Sitosterol. Arch. Int. De Pharmacodyn. Et De Ther. 1988, 296, 224–231. [Google Scholar]
  128. Benny, M.; Antony, B.; Aravind, A.; Gupta, K.; Joseph, B.; Benny, I. Purification and characterization of anti-hyperglycemic bioactive molecule from costus pictus d. don. Int. J. Pharm. Sci. Res. 2020, 11, 2075–2081. [Google Scholar] [CrossRef]
  129. Sashidhara, K.V.; Singh, S.P.; Srivastava, A.; Puri, A. Main Extracts and Hypolipidemic Effects of the Bauhinia Racemosa Lam. Leaf Extract in HFD-Fed Hamsters. Nat. Prod. Res. 2013, 27, 1127–1131. [Google Scholar] [CrossRef] [PubMed]
  130. Zhu, B.; Qi, F.; Wu, J.; Yin, G.; Hua, J.; Zhang, Q.; Qin, L. Red Yeast Rice: A Systematic Review of the Traditional Uses, Chemistry, Pharmacology, and Quality Control of an Important Chinese Folk Medicine. Front. Pharm. 2019, 10, 1449. [Google Scholar] [CrossRef] [Green Version]
  131. Zhou, W.; Guo, R.; Guo, W.; Hong, J.; Li, L.; Ni, L.; Sun, J.; Liu, B.; Rao, P.; Lv, X. Monascus Yellow, Red and Orange Pigments from Red Yeast Rice Ameliorate Lipid Metabolic Disorders and Gut Microbiota Dysbiosis in Wistar Rats Fed on a High-Fat Diet. Food Funct. 2019, 10, 1073–1084. [Google Scholar] [CrossRef]
  132. Mironova, V.N.; Kalashnikova, L.A. [Hypolipidemic action of beta-D-glycoside beta-sitosterol in the rat]. Farm. Toksikol. 1982, 45, 45–47. [Google Scholar]
Figure 1. Chemical structure of Daucosterol.
Figure 1. Chemical structure of Daucosterol.
Applsci 12 05779 g001
Figure 2. Anticancer mechanisms of Daucosterol. The arrow indicates increasing or decreasing.
Figure 2. Anticancer mechanisms of Daucosterol. The arrow indicates increasing or decreasing.
Applsci 12 05779 g002
Figure 3. Neuroprotective effects of Daucosterol. The arrow indicates increasing or decreasing.
Figure 3. Neuroprotective effects of Daucosterol. The arrow indicates increasing or decreasing.
Applsci 12 05779 g003
Figure 4. Anti-inflammatory mechanisms of Daucosterol. The arrow indicates increasing, decreasing or inhibiting.
Figure 4. Anti-inflammatory mechanisms of Daucosterol. The arrow indicates increasing, decreasing or inhibiting.
Applsci 12 05779 g004
Table 2. Extraction and characterization of Daucosterol.
Table 2. Extraction and characterization of Daucosterol.
Plant
Family
Parts UsedExtraction MethodExtraction ParametersPurification MethodYieldsReferences
Prangos ferulacea
Apiaceae
Leaves and stemsMaceration750 g extracted by 3 L of n-hexane, dichloromethan, ethyl acetate, and methanolNMR and FT-IR-[9]
Cassia italica
Fabaceae
Aerial partsNot reportedNot reportedNMR and HMBC-[3]
Ononis mitissima L.
Fabaceae
Aerial partsNot reportedNot reported1D and 2D NMR, mass spectrometry-[8]
Centaurea resupinata subsp. dufourii
Asteraceae
Aerial partsNot reportedNot reportedNMR techniques (1H NMR, 13C NMR, COSY, HSQC, HMBC) and mass spectroscopy (ESI-MS)-[7]
Acanthopanax sessiliflorus (Rupr. and Maxim.) Seem.
Araliaceae
FruitsNot reportedNot reportedElectro-spray ionization/mass spectrometry (ESI/MS), 1H- and 13C-NMR-[49]
Hyssopus cuspidatus Boriss.
Lamiaceae
Aerial partsNot reported17 kg were extracted in triplicate with EtOH (75%) at room temperature (50 L each time).
The crude EtOH extract was concentrated under reduced pressure, followed by suspension in water and successive extraction with petroleum ether, ethyl acetate, and n-butanol
Silica gel column chromatography (SGCC) and further purified with MeOH15 mg[6]
Rheum turkestanicum
Polygonaceae
RootsMaceration3.8 kg extracted with 8 L of n-hexane (24 h × 2)1H-, 13C-, 2D NMR, EI-MS, and single-crystal X-ray diffraction-[14]
Morus alba
Moraceae
LeavesUltrasound1933.76 g extracted in triplicate and successively with n-hexane, EtOAc, and MeOH in sonicator at room temperature (1 h)NMR, using 1H, 13C, DEPT, COSY, HSQC, and HMBC NMR-[35]
Pulicaria inuloides
Asteraceae
Aerial partsNot reported-1H-NMR, 13C-NMR, and HMQC-[57]
Salvia syriaca
Lamiaceae
RootsMaceration2.2 kg of powdered material were extracted with acetone (3 × 10 L) by maceration at room temperaturePreparative thin layer chromatography (TLC) (CHCl3–MeOH (85:15))45 mg[55]
Adenophora triphylla
Campanulaceae
Not reportedNot reportedNot reported1H-NMR, 13C-NMR, and EI mass spectra-[27]
Sedum caeruleum
Crassulaceae
Aerial partsNot reported1500 g of powder was extracted with 80% MeOH.
After evaporating the methanol under vacuum, the residue was dissolved in water and extracted with petroleum ether, chloroform, ethyl acetate, and butanol
UV, 1D, 2D NMR, and MS7.2 mg[56]
Rosa canina L.
Rosaceae
FruitsMaceration1.6 kg of powder was extracted with 4 L of n-hexane, ethyl acetate, acetone, and methanol1H- and 13C-NMR-[58]
Dorema glabrum Fisch. and C.A. Mey.
Apiaceae
Aerial partsMaceration0.8 kg was macerated with methanol (4 L × 5) at room temperatureUV and 1H, 13C-NMR-[60]
Salvia sahendica
Lamiaceae
Aerial partsmaceration3 kg extracted with Me2CO (7 × 5 L)1H and 13C NMR-[36]
Pyrus spp.
Rosaceae
Peels and pulpsNot reported10 g extracted with methanol: water (6:4), acid (solvent A) and 1% (v/v) formic acid in acetonitrile (solvent B)--[62]
Punica granatum
Lythraceae
FlowersNot reported5 g of powder was extracted with 80% ethanol--[64]
Helicteres isora L.
Sterculiaceae
FruitsNot reportedPowdered material was extracted by stirring with 50 mL of 50% methanol at 25 °C for 24 h and centrifuged at 7000 rpm for 10 min.
The pellet was re-extracted with an additional 50 mL of 50% methanol
--[65]
Ceiba pentandra L.
Bombacaceae
SeedsNot reportedPowdered material was extracted by stirring with 50 mL of 50% methanol at 25 °C for 24 h and centrifuged at 7000 rpm for 10 min.
The pellet was re-extracted with an additional 50 mL of 50% methanol
--[65]
Litchi chinensis
Sapindaceae
SeedsNot reported10 kg exhaustively extracted three times with 95% ethanol (50 L) at room temperature--[69]
Randia dumetorum
Rubiaceae
BarkNot reported-13C-NMR spectra using 2D NMR (HSQC, HMBC, and DQF-COSY) [67]
Lysimachia clethroides
Primulaceae
Aerial partsNot reported6.75 kg extracted three times with 75% alcohol for 7 days at room temperature.
The concentrated extract (480 g), after evaporation of the solvent in a vacuum pump, was suspended in water and extracted with petroleum ether, EtOAC, and n-BuOH
1H and 13C NMR-[66]
Urtica angustifolia
Urticaceae
Leaves, roots, and stemsDecoction1 kg was extracted with water (90 °C, 20 BV) 3 times (15 min/time)TLC, IR, and ESI-MS spectral-[70]
Sphallerocarpus gracilis
Apiaceae
RootsNot reported---[73]
Hypericum ascyron L.
Hypericaceae
Whole plantNot reported450 g was refluxed three times with petroleum ether, EtOAC, and MeOH for 2 h13C-NMR20.9 mg[74]
Grewia optiva Drummond ex Burret.
Tiliaceae
Stem barkNot reported7 kg was extracted three times with ethanol at room temperature.
The combined ethanolic extracts were partitioned between EtOAc and water.
The EtOAc layer was washed with H2O, dried (anhydrous Na2SO4), and evaporated under reduced pressure to give a gummy residue that was further fractionated into petroleum ether soluble and insoluble fractions
1D and 2D NMR (HMQC, HMBC, COSY, NOESY, and J-resolved) and EI and HRMS6 mg[18]
Boerhaavia diffusa
Nyctaginaceae
LeavesMaceration150 g extracted with 1 L of distilled water for 24 h [77]
Astragalus membranaceus
Fabaceae
RootsNot reported17.8 kg was chopped into small pieces and refluxed with 70% ethanol for 3 h at 70–80 °C--[80]
Phyllenthus emblica L.
Phyllanthaceae
FruitsNot reported1100 g extracted with 95% ethanol at room temperature for 7 daysSGCC and TLC-[20]
Portulaca oleracea L.
Portulacaceae
Not reportedNot reported1450 g extracted with CHCl31H and 13C aided with HMQC-[21]
Eriobotrya fragrans Champ
Rosaceae
Fruits and leavesNot reported4.5 kg extracted with 95% EtOH three times for 3 days with 2 shakings per day13C-NMR20.8 mg[22]
Alchornea cordifolia (Schumach. and Thonn.) Müll. Arg.
Euphorbiaceae
Leaves and root barkNot reported-1D and 2D NMR spectra were recorded in CDCl3-[82]
Arctotis arctotoides
Asteraceae
Not reported NMR (COSY, NOESY, HMQC, and HMBC) and mass spectra [23]
Dioscorea opposita
Dioscoreaceae
Aerial partsNot reported4 kg extracted with 95% ethanol under reflux.
The ethanol crude extract (115 g) was suspended in water and partitioned successively with petroleum ether, EtOAc, and n-butanol
Silica gel column chromatography using CHCl3-MeOH mixtures (95:5 f 3:1)300 mg[19]
Astragalus mongholicus Bunge
Fabaceae
RootsMaceration10 kg extracted with 90% ethanol at room temperature.
The alcoholic solution was concentrated under vacuum
The concentrated extract was diluted with water.
The water solution was successively extracted with petroleum ether, ethyl acetate, and, finally, n-butanol
13C-NMR-[91]
Punica granatum
Punicaceae
SeedsNot reported4 kg extracted with 95% ethanol under reflux.
The ethanol crude extract (489 g) was suspended in water and partitioned successively with petroleum ether, EtOAc, and n-butanol
1H and 13C NMR205 mg[93]
Artemisia sieversiana
Asteraceae
Aerial partsNot reported14 kg extracted twice for 37 h at room temperature with MeOHCombination of spectral methods (IR, EIMS, H and 2CNMR, DEPT, COSY, NOESY, and HETCOR)64 mg[105]
Table 3. Antioxidant effects of Daucosterol.
Table 3. Antioxidant effects of Daucosterol.
Methods UsedKey ResultsReferences
DPPHIC50 = 490 ± 2.9 µg/Ml[9]
DPPHRSA% = 19.7% at 100 mg/mL[3]
DPPHIC50 = 27.3 ± 0.015 µg/mL[8]
H2O2I% = 31.18 ± 0.5%
FTCI% = 37.12 ± 0.44%
FRAPEC50 = 122.23 ± 0.014 µg EAA/mg ex
PPMEC50 = 16.44 ± 0.0012 µg EAA/mg ex
DPPHIC50 = 36.263 ± 0.005 µg/mL[7]
DPPHNo effect[6]
DPPHIC50 = 155.0 ± 0.5 μM[14]
DPPHIC50 = 224.1 ± 8.2 µg/mL[60]
DPPHIC50 = 6.0 ± 0.1 mg/L[64]
ABTSIC50 = 4.8 ± 0.04 mg/mL[111]
ABTSI% = 27% at 0.04 mg/mL
FRAPEC50 = 1.09 ± 0.12 mg/mL
DPPHEC50 > 250 µg/mL[69]
ABTSEC50 = 143.4 µg/mL
DPPHIC50 = 11.42 ± 0.07 µg/mL[66]
ABTSIC50 = 9.02 ± 0.11 µg/mL
DPPHIC50 = 108.14 ± 9.54 µg GAE/mL[73]
FRAPIC50 = 4.91 ± 0.39 µmol Fe(II)/g of DW
ABTSIC50 = 1.66 ± 0.15 µmol TE/g of DW
DPPHIC50 = 38.86 µg/mL[20]
ABTSIC50 = 0.71 ± 0.01 µg/mL
Table 4. Anticancer effects of Daucosterol.
Table 4. Anticancer effects of Daucosterol.
Cell LinesKey ResultsReferences
Human breast adenocarcinoma MCF-7Suppressed the proliferation of MCF-7 cells
Induced the cytotoxicity of MCF-7 cells
Modulated Bax, Bcl2, and PARP
Reduced the mitochondrial membrane potential
Increased the levels of cytochrome c (Cyt c) released
Upregulated the PTEN gene and inhibited the PI3K/Akt pathway
Decreased the intracellular GSH content
[36]
Human breast cancer MCF-7IC50 = 16.95 μM
Inhibited colony formation of MCF-7 cells
Induced autophagy
Induced the conversion and aggregation of LC3-II
Increased the expression of Beclin-1
[110]
Gastric cancer MGC803IC50 = 19.96 μM
Induced autophagy
Induced the conversion and aggregation of LC3-II
Increased the expression of Beclin-1
Gastric cancer BGC823IC50 = 3.13 μM
Inhibited colony formation of BGC823 cells
Increased ROS production
Induced autophagy
Induced the conversion and aggregation of LC3-II
Increased the expression of Beclin-1
Gastric cancer AGSIC50 = 24.19 μM
Induced autophagy
Induced the conversion and aggregation of LC3-II
Increased the expression of Beclin-1
Human colon cancer HCT-116IC50 = 26.6 μM at 24 h
IC50 = 47.3 μM at 48 h
Decreased the percentage of migrated cells by 14.4% at 100 μM
Increased the percentage of apoptotic cells by 74.2% at 100 μM
Induced cell-cycle arrest at sub-G1 phase
[38]
Human lung cancer A549Inhibited the proliferation of A549 cells
IC50 = 17.46 μg/mL at 48 h
Perturbed cell cycle
Induced apoptotic cell death
[112]
Human HCC HepG2;
Human HCC SMMC-7721
Reduced the proliferation of HepG2 and SMMC-7721 cells
Decreased cell migration and invasion abilities of both cells
Reduced the levels of β-catenin and p-β-catenin
Suppressed the expression of Wnt/ β-catenin signaling proteins
[40]
Breast cancer MCF-7;
MCF-7 xenografts in nude mice
IC50 = 53.27 μg/mL
Inhibited cell viability in ER-positive MCF-7 cells
Induced apoptosis in MCF-7 cells
Diminished the expression of Bcl-xl, Bcl-2, and XIAP
Increased Bax, Bad, and activated caspase
Inactivated the upstream PI3K/Akt/NF-κB pathway
[37]
Breast cancer MDA-MB-231IC50 > 1000 μg/mL
Breast cancer 4T1IC50 > 1000 μg/mL
Blocked metastasis progression
Decreased the number of visible metastasis foci
Inhibited metastasis size distribution in lung tissue
Nontumorigenic breast epithelial MCF-10ANo cytotoxicity
Human prostate cancer (PC3 and LNCap)Inhibited cell proliferation
Induced cell-cycle arrest
Promoted apoptosis and autophagy
Increased phosphorylation of c-Jun N-terminal kinase (JNK)
[113]
Human lung cancer A549IC50 = 20.9 Μm
Inhibited the growth of A549 cells
Increased reactive oxygen species (ROS) level
Promoted intrinsic apoptotic cell death
Increased the expression of caspase-3, caspase-9, Bax, PARP inactivation, and Cyt c release
Diminished the expression of Bcl-2 protein
Inhibited the thioredoxin (TrxR) redox system
[26]
Breast cancer MCF-7;
MCF-7 xenografts in nude mice
Induced cytotoxicity
Decreased the proliferation rates
Increased the number of apoptotic cells
Activated the expressions of caspase 3 and PARP1
Reduced the tumor volume
Decreased the levels of CEA, CA125, and CA153
Increased the expression of cleaved caspase 3
Decreased the BCL-2 and VEGF
Downregulated the expression of PI3K/Akt
Repressed insulin-induced PI3K/Akt activation
[25]
MDA-MB-231Exhibited a weak effect on cell proliferation
4T1 nontumorigenicExhibited a weak effect on cell proliferation
Breast epithelial MCF-10AExhibited a weak effect on cell proliferation
7,12-dimethylbenz(a)anthracene-induced mammary tumors in Wistar ratsReduced tumor volume
Decreased the levels of protein and malondialdehyde (MDA)
Reduced cancer antigen (CA) 15-3 level
Decreased MDA levels
Increased catalase activity
Reduced proliferation of mammary duct cells
[24]
Prostate carcinoma LNCaPInhibited cell growth and proliferation[48]
Prostate carcinoma DU145Inhibited cell growth and proliferation
Increased the number of late apoptotic cells
Increased the number of cells in S phase
Decreased the number of G0/G1 cells
Downregulated the cell-cycle proteins (cdk1, pcdk1, cyclin A and B)
Prostate carcinoma PC3Inhibited cell growth and proliferation
Increased the number of late apoptotic cells
Downregulated the cell-cycle proteins (cdk1, pcdk1, cyclin A and B)
Downregulated cdk2
Downregulated Akt, pAKT, and Bcl-2 proteins
Upregulated the pro-apoptotic protein Bax
Breast tumor BALB/c mouse modelSuppressed primary tumor growth
Reduced lung metastases
Increased the number of captured CTCs in the blood circulation
[117]
Murine H22 hepatoma allograft model in ICR miceInhibited murine hepatoma H22 cell growth
Induced intracellular ROS generation
Induced autophagy
[110]
Table 5. Neuroprotective effects of Daucosterol.
Table 5. Neuroprotective effects of Daucosterol.
Experimental ApproachesKey ResultsReferences
Oxygen–glucose deprivation/reperfusion-mediated injury in OGD/R modelReduced neuronal loss and apoptotic rate
Suppressed caspase-3 activity
Upregulated the expression of IGF1 protein
Activated the AKT signal pathway
Diminished the downregulation of the Mcl-1 and Bcl-2
Decreased the expression level of protein Bax
[40]
H2O2-induced cell death of human brain neuroblastoma SK-N-SH cellsInhibited cell death and LDH activity
Reduced intracellular ROS levels by 37.7%
Reduced H2O2-induced apoptotic cell death
Reduced H2O2-mediated fragmented DNA
Increased CAT and SOD2 mRNA levels
Attenuated H2O2-induced phosphorylation of p38 and JNK
[41]
Cerebral ischemia/reperfusion (I/R) rat modelDecreased apoptotic cell death
Suppressed iNOS expression in ischemic zone
Upregulated the PI3K/Akt/mTOR signaling pathway
[30]
Table 6. Antidiabetic activities of daucosterol.
Table 6. Antidiabetic activities of daucosterol.
Experimental ApproachesKey ResultsReferences
α-amylase inhibitorsInhibition = 57.5 ± 3.1% in a 10 mg/mL[126]
α-glucosidase inhibitorsIC50 = 247.35 mg/L
Inhibition constant = 2.34 mg/L
[42]
Normal and hyperglycemic ratsIncreased fasting plasma insulin levels
Enhanced oral glucose tolerance
Improved glucose-induced insulin release
[127]
Molecular dockingInhibited human α-glucosidase[53]
α-glucosidase inhibitorsIC50 = 13.3 ± 1.9 µM[58]
Glucose tolerance testSignificant hypoglycemic activity[128]
α-glucosidase inhibitorsIC50 = 5.67 mg/L[33]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

El Omari, N.; Jaouadi, I.; Lahyaoui, M.; Benali, T.; Taha, D.; Bakrim, S.; El Menyiy, N.; El Kamari, F.; Zengin, G.; Bangar, S.P.; et al. Natural Sources, Pharmacological Properties, and Health Benefits of Daucosterol: Versatility of Actions. Appl. Sci. 2022, 12, 5779. https://doi.org/10.3390/app12125779

AMA Style

El Omari N, Jaouadi I, Lahyaoui M, Benali T, Taha D, Bakrim S, El Menyiy N, El Kamari F, Zengin G, Bangar SP, et al. Natural Sources, Pharmacological Properties, and Health Benefits of Daucosterol: Versatility of Actions. Applied Sciences. 2022; 12(12):5779. https://doi.org/10.3390/app12125779

Chicago/Turabian Style

El Omari, Nasreddine, Imane Jaouadi, Manal Lahyaoui, Taoufiq Benali, Douae Taha, Saad Bakrim, Naoual El Menyiy, Fatima El Kamari, Gökhan Zengin, Sneh Punia Bangar, and et al. 2022. "Natural Sources, Pharmacological Properties, and Health Benefits of Daucosterol: Versatility of Actions" Applied Sciences 12, no. 12: 5779. https://doi.org/10.3390/app12125779

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

Article Metrics

Back to TopTop