Next Article in Journal
Special Issue “Recent Advances in Precision Nanomedicine for Cancer”
Next Article in Special Issue
Profiling Anticancer and Antioxidant Activities of Phenolic Compounds Present in Black Walnuts (Juglans nigra) Using a High-Throughput Screening Approach
Previous Article in Journal
Water-Tree Resistability of UV-XLPE from Hydrophilicity of Auxiliary Crosslinkers
Previous Article in Special Issue
Structure and Physical Properties of Cardamonin: A Spectroscopic and Computational Approach
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Sephadex® LH-20, Isolation, and Purification of Flavonoids from Plant Species: A Comprehensive Review

by
Javad Mottaghipisheh
1,* and
Marcello Iriti
2,*
1
Department of Pharmacognosy, Faculty of Pharmacy, University of Szeged, Eötvös u. 6, 6720 Szeged, Hungary
2
Department of Agricultural and Environmental Sciences, Milan State University, via G. Celoria 2, 20133 Milan, Italy
*
Authors to whom correspondence should be addressed.
Molecules 2020, 25(18), 4146; https://doi.org/10.3390/molecules25184146
Submission received: 20 August 2020 / Revised: 2 September 2020 / Accepted: 8 September 2020 / Published: 10 September 2020
(This article belongs to the Special Issue Bioactive Phenolic and Polyphenolic Compounds)

Abstract

:
Flavonoids are considered one of the most diverse phenolic compounds possessing several valuable health benefits. The present study aimed at gathering all correlated reports, in which Sephadex® LH-20 (SLH) has been utilized as the final step to isolate or purify of flavonoid derivatives among all plant families. Overall, 189 flavonoids have been documented, while the majority were identified from the Asteraceae, Moraceae, and Poaceae families. Application of SLH has led to isolate 79 flavonols, 63 flavones, and 18 flavanones. Homoisoflavanoids, and proanthocyanidins have only been isolated from the Asparagaceae and Lauraceae families, respectively, while the Asteraceae was the richest in flavones possessing 22 derivatives. Six flavones, four flavonols, three homoisoflavonoids, one flavanone, a flavanol, and an isoflavanol have been isolated as the new secondary metabolites. This technique has been able to isolate quercetin from 19 plant species, along with its 31 derivatives. Pure methanol and in combination with water, chloroform, and dichloromethane have generally been used as eluents. This comprehensive review provides significant information regarding to remarkably use of SLH in isolation and purification of flavonoids from all the plant families; thus, it might be considered an appreciable guideline for further phytochemical investigation of these compounds.

Graphical Abstract

1. Introduction

Flavonoids are considered as one of the most varied phenolic compounds. Different derivatives of these secondary metabolites, which are naturally synthesized in higher plants and microbial sources, possess extensive beneficial properties for human health. Many studies have assessed pharmacological and bioactivities of these compounds. Along with those effects, the importance of these compounds is mainly related to their ability in the scavenging of free radicals, hence possessing high antioxidant activity [1,2].
Isolation of flavonoids have been majorly carried out by hiring chromatographic methods. These techniques have been developed by the Noble laureates in chemistry at 1952 Archer John Porter Martin and Richard Laurence Millington Synge for their invention of partition chromatography [3]. These methods are the most remarkable separation techniques, which are extensively applied in natural product chemistry analysis for both analytical and preparative purposes. The chromatographic methods functionally separate the mixtures according to physical properties of their constituents.
Silica gel chromatography has been widely used in the isolation and characterization of these compounds. They can be separated according to the polarity, while normal phase and reversed phase (C18 silica gel) are applied for isolation of low to moderate polar and high polar flavonoids (e.g., glycosylated forms), respectively. The ability of polyamide to make hydrogen bonds with hydroxyl groups of flavonoids, depending on the numbers and positions of this groups, are the basis of the isolation process in this technique. The recent advanced techniques, including High-performance liquid chromatography (HPLC), high-speed counter current chromatography (HSCCC), molecular imprinting technology (MIT), droplet counter-current chromatography (DCCC), along with older methods medium pressure liquid chromatography (MPLC), circular liquid chromatography (CLC), and centrifugal preparative thin layer chromatography (CPTLC), are the applied techniques to isolate flavonoids [2,4,5]. However, GC-MS, HPLC-ESI-Q-TOF-MS, HPLC-PdAD-ESI-MS/MS, LC-MS, LC-MS/MS, and ultra-HPLC (UHPLC), have mostly been employed to analytical analysis of flavonoids from plant species [5]. The acidity of the extraction solvents have been reported a direct correlation with flavonoid contents, whereas in a study on Vitis vinifera, total flavonoid contents enhanced (from 20.63 to 46.77 mg/g) by addition of HCl (from 0 to 1%) [6]. In another analytical research, the content of spinacetin 3-gentiobioside was the highest in a pH of 2.5, compared to the applied acidity media (7.25 and 12), with the same other parameters in the extraction procedure [7].
Size-exclusion chromatography is considered a kind of partition chromatography, which is able to isolate compounds according to various molecular sizes. Gel-permeation, gel-exclusion, gel filtration, and molecular-sieve chromatography are the alternative definitions of this technique. The diameter and pore size of packed materials, choosing a proper eluent as mobile phase, and length of the used column are the significant parameters for effectively separation of a mixture by size exclusion chromatography [8].
The first application of size-exclusion procedure refers to separation of peptides from amino acids on a column packed with starch [9]. The Pharmacia company (Stockholm, Sweden) developed dextran crosslinked with epichlorohydrin with tradename of “Sephadex®” [10]. Initially, Sephadex® comprised irregular particles, then was synthesized as porous spheres [11].
Nuclear magnetic resonance (NMR), mass spectrometry (MS), spectrophotometric ultra-violet (UV), and infrared (IR) techniques providing spectroscopic spectra of flavonoids, have been exploited to elucidate the flavonoid structures. Moreover, their physical characteristics such as melting point (mp), circular dichroism (CD), and optical rotatory power ([α]D) can be applied to identify the flavonoids [5].
However, the new techniques have facilitated isolation and identification of flavonoids from specifically plant resources, the classical methods particularly Sephadex® LH-20 (SLH) has been widely utilized, due to being inexpensive, convenient, rapid, and efficient. For the first time, this review comprehensively gathered all the existing information about the application of SLH in isolation or purification of diverse range of flavonoid derivatives, where this method has been used as the last separation step. The keywords of “flavonoid” and “Sephadex” have been applied to search the correlated published data through databases including PubMed and Web of Science (last search: 27 June 2020).

2. Isolation of Various Flavonoid Classifications by Sephadex® LH-20

Overall, 190 flavonoid derivatives have been isolated or purified by utilization of SLH from 40 various plant families. This method has been able to isolate or purify seven major flavonoid classifications, including flavan and isoflavan, flavanone, flavanol and isoflavanol, flavone and isoflavone, flavonol, homoisoflavonoid, and proanthocyanidin derivatives (Table S1). In general, one flavan and an isoflavan, 18 flavanones, eight flavanols and one isoflavanol, 63 flavones and five isoflavones, 79 flavonols, 10 homoisoflavonoids, and three proanthocyanidins have been isolated and identified. The basic chemical structures of the flavonoid classifications have been illustrated in Figure 1.
The most isolated flavonoids have been identified from Asteraceae family including 37 different flavonoids, besides the plants belonging to Moraceae and Poaceae with 27 and 24 possessed more flavonoids, respectively (Figure 2). The species in families Asteraceae with 22 flavones, and Asteraceae and Fabaceae with 13 flavonols were the richest; whilst whole 10 and three isolated homoisoflavonoids and proanthocyanidins have been isolated from Asparagaceae and Lauraceae families, respectively.
Overall, 17 different eluents have been applied to isolate/purify flavonoids through SLH column. Pure methanol and its mixtures specifically in combination with water have been exploited as the most prevalent eluents in isolation of 71 and 67 flavonoids, respectively (Table 1).

2.1. Flavan and Isoflavan Derivatives

Among 15 isolated flavonoids from chloroform extract of Dalbergia cochinchinensis herb, one flavan namely 6,4′-dihydroxy-7-methoxy-flavan (1) and an isoflavan mucronulatol (2) have been purified by applying SLH with dichloromethane‒methanol (1:1) as the eluting solvent [12].

2.2. Flavanone Derivatives

Dihydrowogonin (3) is a 5,7-dihydroxy-8-methoxyflavanone which has been isolated from dichloromethane extract of Chenopodium procerum aerial parts. Methanol was applied as solvent system to isolate the mentioned flavanone via SLH [13].
Isolation of naringenin (4) has been carried out from three plant species. n-Butanol extract obtained from Paulownia tomentosa bark by applying methanol‒water (1:1, 1:3) [14], chloroform fraction of Dalbergia cochinchinensis herb with dichloromethane‒water (1:1) [12], and ethyl acetate extract gained from wooden part of Populus davidiana by using methanol‒water (3:1, 1:1, 1:3) as elution solvents [15]. One glycosylated derivative of naringenin called naringenin 7-O-β-glucopyranoside (syn. prunin) (5) has been furtherly isolated from hydro-methanolic extract (80%) of leaf and flower of Hawthorn (Crataegus spp.) by increasing ratio of methanol (40 to 70%) in water by applying SLH [16].
Jung et al. [17] subjected ethyl acetate extract of root bark of Morus alba to isolate major constituents. Flavanones including sanggenol Q (6), sanggenol F (7) [17], a new compound sanggenon U (8), and kuwanon E (9) by using methanol‒water (8:2), along with euchrenone a7 (10) with methanol‒water (7:3) as SLH eluents have been isolated and purified [18]. In another study, three other flavanones namely sanggenon J (11), sanggenon F (12), and sanggenol A (13) have also been isolated from the root bark ethyl acetate extract of M. alba, where the samples were eluted with methanol and methanol‒water (1:1) as eluents through SLH [19].
Dichloromethane‒methanol (1:1) has been used as eluent to isolate pinocembrin (14) from ethyl acetate extract of Corema album and petroleum ether fraction of Dalbergia cochinchinensis herb. This compound is a 5,7-dihydroxyflavanone and has been extracted from honey, propolis, ginger roots, etc. were reported as a potential natural drug to treat ischemic stroke, and for its anti-inflammatory and neuroprotective effects [20,21].
Liquiritigenin (15) and alpinetin (16) from chloroform, and 7,8-dihydroxyflavanone (17) from ethyl acetate extracts of Dalbergia cochinchinensis herb have been previously isolated by eluting dichloromethane‒methanol (1:1) through SLH [12]. Methanolic extract obtained from the aerial parts of Taraxacum mongolicum have been chromatographed and finally two flavanones hesperidin (18) and 4′,5,7-trihydroxy-3′-methoxyflavanone (19) were purified by SLH (eluent: methanol) [22]. Another aglycone flavanone (2S)-homoeriodictyol (20) has been furtherly isolated from methanolic extract of the whole parts of Dendrobium ellipsophyllum applying SLH eluting with acetone [23].

2.3. Flavanol and Isoflavanol Derivatives

In general, eight flavanol (2128) and a novel isoflavanol (29) have been isolated and purified by SLH as the final chromatographic step from families including Oleaceae, Moraceae, and Fabaceae. Phytochemical investigation of ethyl acetate extract of Chionanthus retusus flowers led to the isolation of aromadendrin (21) and taxifolin (syn. dihydroquercetin) (22) using SLH (eluent: methanol‒water 8:2) as the last separation step [24]. The aforementioned aglycone flavanols (21,22), along with two glycosylated taxifolin namely taxifolin 7-glucoside (23) and 6-p-hydroxybenzyl taxifolin-7-O-β-d-glucoside (24), and two other aglycones gericudranin E (27) and gericudranin C (28), have been furtherly isolated from Cudrania tricuspidata aqueous extract of bark utilizing methanol‒water (1:1) as eluting solvent [25].
2,3-trans-Dihydromorin (25) [19] and a novel flavanol (2R,3S)-guibourtinidol-3-O-α-d-apiofuranosyl-(1→6)-O-β-d-glucopyranoside (26) [26] have been previously isolated from ethyl acetate and n-butanol extracts of Morus alba root barks via gel filtration SLH column with methanol and methanol‒water (3:2) as eluents, respectively. Awouafack et al. [27] hired SLH to isolate a new isoflavanol namely kotstrigoisoflavanol (29) from methanolic extract of Kotschya strigosa fruit.

2.4. Flavone and Isoflavone Derivatives

The 63 flavone (3092) and five isoflavone (9397) derivatives isolated by using SLH as the last separation stage illustrated that this technique plays an effective role in extraction of these compounds.
The simple flavone (30) and its derivative 4′-hydroxy-5-methoxyflavone (33) have been isolated from Imperata cylindrica, whilst ethyl acetate extracts of rhizome were finally chromatographed via SLH with dichloromethane‒methanol (1:1) as eluent system [28]. Ethyl acetate and ethanolic extracts gained from stem bark and aerial parts of Albizzia julibrissin and Athrixia phylicoides were extracted to isolate two aglycone flavones of 3′,4′,7-trihydroxyflavone (31) [29] and 5-hydroxy-6,7,8,3′,4′,5′-hexamethoxyflavon-3-ol (32) [30], respectively, through a separation procedure with SLH (eluent: methanol).
A well-known flavone luteolin (34) (3,4,5,7-tetrahydroxy flavone) possessing several health benefits, such as anti-cancer [31], cardio-protective [32], anti-inflammation, and anti-allergy [33] effects, has been isolated and purified from 11 plant species applying SLH as the final step: from hydroethanolic (70%) extract of Brachychiton acerifolius leaf (eluent: methanol‒water 1:1) [34], ethyl acetate extracts of Thymus praecox aerial part [35], Ginko biloba leaf (eluent: methanol) [36], Rosmarinus officinalis sprig (eluent: methanol‒water 1:1) [37], Chamaemelum nobile flower (eluent: methanol‒dichloromethane 1:1) [38], Populus davidiana wood (eluent: methanol‒water 3:1, 1:1, 1:3) [15], and Solenostemon monostachys aerial part (eluent: n-hexane‒ethyl acetate 3:7, 2:8, 1:9; ethyl acetate; ethyl acetate‒methanol 1:9, 2:8, 4:6, 5:5) [39], aqueous fraction of Phlomis bruguieri aerial part (eluent: n-hexane‒MeOH‒acetone 30:60:10) [40], methanolic extracts of Taraxacum mongolicum aerial part (eluent: methanol) [22] and Dendrobium ellipsophyllum whole plant part (eluent: acetone) [23], and n-butanol extract of xylem part of Populus tomentosa (eluent: methanol‒water 1:1, 1:3) [41].
Moreover, 7-methoxy luteolin (35) has been isolated from ethyl acetate extract of Onopordum alexandrinum seeds via SLH as the final step with methanol‒water (9:1) as eluting solvent [42]. Overall, five glycosylated luteolin (3640) have been purified applying gel filtration chromatography. Orientin (36) which is luteolin 8-C-glucoside was finally isolated by SLH (eluent: methanol) from petroleum ether extract of Indocalamus latifolius leaf [43].
Cynaroside (37) as luteolin 7-O-β-d-glucoside have been previously isolated from six plant species: ethyl acetate extracts of Tridax procumbens whole part [44] and Salvia macrosiphon aerial part (eluent: methanol) [45], hydro-methanolic (80%) portion of Tilia rubra leaf (eluent: methanol‒water 8:2), hydroethanolic extracts of leaf of Olea europaea (eluent: ethanol 0–50% in water) [46] and Brachychiton acerifolius (eluent: methanol‒water 1:1) [34], and chloroform extract obtained from Citrus unshiu peel (eluent: methanol‒water 1:1) [47].
From the methanolic extract of Taraxacum mongolicum aerial part eluting with methanol through SLH, luteolin-7-O-β-d-galactopyranoside (38) and luteolin-7-O-β-d-glucopyranoside (39) [22], and luteolin-4′-O-β-glucoside (40) from hydroethanolic (50%) extract of Olea europaea leaf (eluent: ethanol 0‒50% in water) [46] have been furtherly isolated.
Apigenin (41), characterized as 4′,5,7,-trihydroxyflavone is considered as a natural flavone, and rich in several fruits, vegetables and medicinal plants possessing numerous pharmacological potencies, such as anti-inflammatory, antioxidant, antibacterial, antiviral, antidiabetic, antidepressant, and anticancer activities, and the treatment of amnesia and Alzheimer’s disease, and insomnia [48,49,50,51,52]. SLH has been capable to isolate this natural product from hydroethanolic extracts of Brachychiton acerifolius leaf (eluent: methanol‒water 1:1) [34] and Saccharum officinarum sugarcane top (eluent: chloroform‒methanol 1:1) [53], ethyl acetate fractions of Chamaemelum nobile flowers (eluent: methanol‒dichloromethane 1:1) [38], and Solenostemon monostachys aerial part (eluent: n-hexane‒ethyl acetate 3:7, 2:8, 1:9; ethyl acetate; ethyl acetate‒methanol 1:9, 2:8, 4:6, 5:5) [39], n-butanol extract of xylem of Populus tomentosa (eluent: methanol‒water 1:1, 1:3) [41], and aqueous extract of Phlomis bruguieri aerial part (eluent: n-hexane‒methanol‒acetone 30:60:10) [40].
From leaf hydroethanolic (70%) extract of Brachychiton acerifolius, apigenin-7-O-α-rhamnosyl (1→2)-β-D-glucuronide (42), apigenin-7-O-β-d-glucoside (43), and apigenin-7-O-β-d-glucuronide (44) have been isolated eluting with methanol‒water (1:1) [34]. Nonetheless, apigenin-7-O-β-d-glucoside (43) were isolated from ethyl acetate extracts of aerial parts of Thymus praecox [35] and Salvia macrosiphon (eluent: methanol) [45] as two Lamiaceae species; moreover, apigenin-7-O-β-d-glucuronide (44) was purified from n-butanol fraction of Erigeron multiradiatus whole part (eluent: chloroform‒methanol 1:1) [54].
SLH was applied as the last chromatographic step in isolation of vitexin (45) (apigenin 8-C-glucoside) from hydroethanolic (60%) and petroleum ether extracts obtained from Desmodium adscendens [55] and Indocalamus latifolius [43] leaves, where methanol (20 to 100%) in water and pure methanol were used as eluting solvents, respectively.
Vitexin 2”-O-xyloside (46) and its iso-derivative namely isovitexin 2”-O-xyloside (48) have been formerly isolated from Desmodium adscendens leaf hydroethanolic (60%) extract utilizing methanol (20 to 100%) in water as eluent [55]; however, isovitexin (47) is apigenin-6-C-glucoside that has been isolated from ethanolic extract of Croton zambesicus leaf with ethyl acetate in methanol (10 to 100%) as eluting solvent in SLH [56].
Gohari et al. [45] isolated apigenin-7,4′-dimethyl ether (49) by finally exploiting SLH (eluent: methanol) from ethyl acetate extract fractionated from Salvia macrosiphon aerial part. From ethyl acetate extract of Aquilaria sinensis seeds, 7,4′-dimethylapigenin-5-O-xylosylglucoside (50) and 7,4′-dimethyl-5-O-glucosideflavonoide (55) eluting with methanol‒water (7:3), along with hydroxylgenkwanin (51), lethedoside A (52), 5,7-dihydroxyl-4′-methoxyflavone (53), and 7,3′-dimethyl-4′-hydroxyl-5-O-glucosideflavonoide (54) using methanol as eluent have been isolated and purified via SLH [57].
In another investigation, amentoflavone (56) was isolated from Ginko biloba leaf ethyl acetate extract by application of methanol as eluent [36]. Hispidulin (57) has been isolated from ethyl acetate extracts of sprig and flower of Rosmarinus officinalis [37] and Chamaemelum nobile [38] utilizing methanol‒water (2:1) and methanol‒dichloromethane (1:1), respectively. Root bark of Morus alba has been previously partitioned and its ethyl acetate extract was subjected to separation of their phytoconstituents, finally through methanol‒water (8:2) as eluent in SLH, 2 known flavones kuwanon T (58) and sanggenon J (59), in addition, two novel secondary metabolites sanggenon V (60) and sanggenon W (61) have been isolated accordingly [18].
Several other flavone derivatives have been also isolated and purified from different soluble-extracts of the species by SLH: hypoletin-7-O-β-d-xylopyranoside (62) from leaf ethyl acetate extract of Thuja orientalis (eluent: methanol) [58], galangin (63) from herb chloroform fraction of Dalbergia cochinchinensis (eluent: methanol‒dichloromethane 1:1) [12], 3′-geranyl-3-prenyl-2′,4′,5,7-tetrahydroxyflavone (64) from ethyl acetate extract of Morus alba root bark (eluent: methanol‒water 1:1) [19], pectolinarigenin (65) from chloroform fraction of Cirsium Japonicum aerial part [59], scutellarein-7-O-β-glucuronide (66) from Erigeron multiradiatus n-butanol aerial part extract [54], cirsimaritin (67), cirsilinelol (68), and eupatilin (69) from chloroform extract of aerial part of Centaurea bruguierana [60], eluting with chloroform‒methanol (1:1), and eupafolin (70) from ethyl acetate extract of Chamaemelum nobile aerial part (eluent: methanol‒dichloromethane 1:1) [38].
Tricin (71) is 5,7,4′-trihydroxy-3′,5′-dimethoxyflavone, comprising many valuable bio- and pharmacological properties [61], and it has been isolated from leaf ethyl acetate extract of Sasa senanensis (eluent: methanol‒water 6:4) [62], bract hydroethanolic (95%) fraction of Zea mays [63], and aqueous extract of Phlomis bruguieri aerial part (eluent: n-hexane‒methanol‒acetone 3:6:1) [40].
The application of SLH on hydroethanolic (95%) extract of Zea mayes bract has led to isolation of three tricin glucosides including tricin-5-O-β-d-glucopyranoside (72), tricin-7-O-β-d-glucopyranoside (73), and novel flavone namely tricin-7-O-[β-d-apifuranosyl (1→2)]-β-d-glucopyranoside (74) [63]. Tricin-7-O-β-d-glucopyranoside (73) has been furtherly isolated from two other Poaceae species Avena sativa [64] and Indocalamus latifolius [43], while a hydroethanolic (95%) fraction of bran and methanolic extract of leaf have been eluted by methanol in SLH column, respectively.
A new secondary metabolite 4′-methoxy-luteolin-7-phosphate (75) has been formerly isolated by hiring SLH (eluent: n-hexane‒methanol‒acetone 3:6:1) from aerial part aqueous extract of Phlomis bruguieri [40]. From an Asteraceae species Santolina chamaecyparissus nepetin (76) (eluent: methanol) was purified, where the dichloromethane extract of its aerial part was subjected to chromatographic procedure [65].
Isoetin (77) and its glycosylated analogous including isoetin-7-O-β-d-glucopyranosyl-2′-O-α-l-arabinopyranoside (79), isoetin-7-O-β-d-glucopyranosyl-2′-O-α-d-arabinopyranoside (80), and isoetin-7-O-β-d-glucopyranosyl-2′-O-α-d-xyloypyranoside (81), along with genkwanin (82) and genkwanin-4′-O-β-d-lutinoside (83), have been isolated and purified by applying SLH as the last separation stage (eluent: methanol) from methanol extract of Taraxacum mongolicum aerial part [22]. Notably, a novel flavone isoetin 2′-methyl ether (78) (5,7,4′,5′-tetrahydroxy-2′-methoxyflavone) has been isolated from Bauhinia galpinii, where the ethyl acetate extract of the leaf were applied by using acetone‒methanol (1:1) as eluting solvent via SLH [66].
By subjecting hydroethanolic (50%) extract of sugarcane top part of Saccharum officinarum to various chromatographic methods, albanin A (84), australone A (85), and 5′-geranyl-5,7,2′,4′-tetrahydroxy-flavone (86) have been finally isolated by exploiting chloroform‒methanol (1:1) and pure methanol as eluting solvents in SLH column [53]. In another study, methanolic extract obtained from whole part of Dendrobium ellipsophyllum were subjected to SLH (eluent: acetone) and chrysoeriol (87) was consequently isolated [23]. Xuan et al. [28] isolated 4′-methoxyflavone-6-O-β-d-glucopyranoside (88) for the first time in the nature from rhizome ethyl acetate extract of Imperata cylindrica by SLH (eluent: methanol), whereas 5-hydroxyflavone (89) was furtherly isolated from its petroleum ether extract by using dichloromethane‒methanol (1:1) as eluting mixture.
Three other flavones have also been isolated by SLH: texasin 7-O-β-d-glucopyranoside (90) from ethyl acetate extract of Leptadenia pyrotechnica aerial part [67], tilianin (91) from hydroethanolic (95%) extract of Avena sativa bran (eluent: methanol) [64], and 5-hydroxy-6,7,3′,4′-tetramethoxyflavone (92) from flower chloroform extract of Citrus aurantium (eluent: chloroform‒methanol 1:1) [68].
Moreover, 5 isoflavone derivatives have been isolated by SLH as the last separation procedure. Formononetin-7-O-β-d-glucosy1 [1,2,3,4,5,6] glucoside (94) and tectoridin (95) have been purified from ethyl acetate extract of Maackia amurensis bark (eluent: methanol‒water 6:4) [69], whilst formononetin (93) was extracted from Aquilaria sinensis stem ethyl acetate extract (eluent: methanol) [57]. Sphaerobioside (96) and a well-known isoflavone genistein (97) have been previously isolated by SLH eluting with methanol‒water (1:1) and methanol, respectively, from aqueous root fractions of Cudrania tricuspidata [25].

2.5. Flavonol Derivatives

SLH played a key role in isolation or purification of flavonoids specifically flavonol derivatives. The performed studies reported that SLH has been applied for isolation or purification of 79 different flavonol derivatives (98176), while quercetin (98) with its analogous (99129), and kaempferol (130) and its analogous (131152) were the most identified compound.
Quercetin (98) (3,3′,4′,5,7-pentahydroxyflavone, C15H10O7) is considered as one of the most beneficial flavonols and renowned for its antioxidant, anticancer, anti-inflammatory, and antiviral properties and endothelium-dependent vasodilation, and blood lipid-lowering effects [70,71,72,73]. SLH gel filtration chromatography has been able to isolate and purify quercetin (98). Nineteen studies reported the successful isolation of this compound from 19 diverse species by using SLH. According to the literature, it seems the ethyl acetate fractions of various plant species are the richest extracts in case of quercetin (98) content.
The calix part of Fragaria ananassa was solvent-solvent partitioned and finally by utilizing SLH (eluent: methanol‒water 6:4), quercetin (98) was isolated from the ethyl acetate extract [74]. The ethyl acetate extract of Gynura divaricate leaf has been furtherly subjected to isolate their major secondary metabolites, and the abovementioned compound was isolated by chloroform‒methanol (1:1) as an eluent system [75]. By eluting methanol through SLH column, quercetin (98) has been isolated from Sarcopyramis bodinieri ethyl acetate extract [76]. Quercetin (98) has also been isolated from ethyl acetate extracts of Chionanthus retusus flower (eluent: methanol‒water 8:2) [24], Tamarix hohenackeri aerial parts (eluent: methanol) [77], whole part of Pteris vittata (eluent: chloroform‒methanol 1:1) [78], Populus davidiana wood eluting with methanol‒water (3:1, 1:1, 1:3) [15], and from aerial part of Halimodendron halodendron (eluent: chloroform‒methanol 1:1) [79].
Several researchers isolated quercetin (98) from alcoholic extracts of different species via SLH as the last chromatographic step. Abou Zeid et al. [34] isolated this flavonol from hydroethanolic (70%) extract of Brachychiton acerifolius leaf eluting by methanol‒water (1:1) as eluent. In other phytochemical studies on Byrsocarpus coccineus (Connaraceae family) [80], Juniperus chinensis (Cupressaceae family) [81], and Paulownia tomentosa (Scrophulariaceae family) [14], this compound has been isolated from n-butanol fraction of the leaf, herb, and bark, while methanol, chloroform‒methanol (4:1), and methanol‒water (1:1) were applied as eluents, respectively. The methanolic extracts of Cheilanthes tenuifolia whole part [82] and Taraxacum mongolicum aerial part [22] have been exploited to isolate this phytochemical eluting with methanol (0 to 60%) in water and pure methanol, respectively.
Hydroalcoholic fractions of some species have been previously applied for isolation and purification of quercetin (98): hydro-methanolic (70%) extracts of leaf and aerial part of Albizia amara [83] and Allium porrum [84] eluting via methanol and methanol‒water (6:4), respectively, and hydro-ethanolic (50%) extract of sugarcane top part of Saccharum officinarum (eluent: chloroform‒methanol 1:1) [53]. Furthermore, this aglycone flavonol has been isolated from stem aqueous extract of Bauhinia strychnifolia using methanol as eluent in SLH gel filtration method [85]. Among all isolated quercetin derivatives (99129) by applying SLH, two aglycones, including 3-O-methylquercetin (99) and 3,3′-di-O-methylquercetin (100) have been isolated from the ethyl acetate extract of Halimodendron halodendron (Fabaceae) aerial part with mixture eluting solvents of chloroform‒methanol (1:1) [79].
Rutin (101) (syn. quercetin-3-O-α-rhamnosyl (1→6)-β-d-glucoside or 3′,4′,5,7-tetrahydroxy-flavone-3-rutinoside), as a well-renown dietary flavonoid, has been reported to possess several remarkable pharmacological benefits, such as in the treatment of Parkinson’s, and Alzheimer’s diseases, and myocardial infraction, along with anti-depressant, antihypertensive, anti-allergic, antioxidant, and anticancer properties [86,87,88]. However, this compound has been isolated by different methods, specifically solid-phase extraction and counter-current chromatography, and the size exclusion technique has also been applied to isolate this compound [86]. By utilization of SLH as the final purification phase, rutin (101) has been isolated from hydroethanolic (70%) and aqueous extracts of leaf and fruit of Brachychiton acerifolius [34] and Cinnamomum zeylanicum [89], respectively, by using methanol‒water (1:1), and from whole part methanolic fraction of Cheilanthes tenuifolia eluting with methanol (0 to 60%) in water [82].
Application of SLH has led to isolation of quercetin-3-O-β-6′’-(p-coumaroyl) glucopyranoside-3′-methyl ether (102) (syn. helichrysoside-3′-methyl ether) from ethanolic leaf extract of Croton zambesicus with chloroform (10 to 60%) in methanol as eluent [56]. Two glycosylated quercetin analogous quercetin 3-β-d-glucoside (103) and quercetin 3-O-α-arabinoside (104) have been isolated using n-butanol and ethyl acetate extracts of Byrsocarpus coccineus leaf, respectively, in which methanol was as eluting solvent [80].
From leaf ethyl acetate fractions of Bauhinia galpinii and Dryopteris filix-mas have been finally isolated quercetin-3-O-β-galactopyranoside (105) [66] and quercetin-3-O-α-l-rhamnopyranoside (106) [90] exploiting acetone‒methanol (1:1) and pure methanol as eluting solvent systems, respectively. Quercetin-3-O-α-l-rhamnopyranoside (106) has been furtherly isolated from aqueous and n-butanol extracts of flower and leaf of Cinnamomum zeylanicum [89] and Curcuma longa [91], respectively, by using methanol‒water as eluent mixture in SLH gel filtration column. The leaf n-butanol extract of Ficus exasperate was extracted and quercetin-3-O-β-rhamnoside (107) accordingly isolated via SLH (eluent: toluene‒ethanol 7:3) [92]. By utilization of methanol as eluting solvent through SLH column, quercetin-3-O-glucopyranoside (108) has been isolated from leaf methanolic and n-butanol extracts of Indocalamus latifolius [43] and Sambucus ebulus [93], respectively. Another glycosylated quercetin derivative namely quercetin-3-O-β-d-glucuronide (109) has been obtained by SLH from n-butanol, ethanol, and ethyl acetate extracts of leaf, leaf, and stem parts of Curcuma longa [91], Eugenia jambos [94], and Nelumbo nucifera [95], while methanol‒water (8:2), ethanol‒water (7:3), and methanol were applied as eluents, respectively.
In similar studies, quercetin-3-O-sambubioside (110) and quercetin-3-O-sophoroside (112) have been isolated from n-butanol and hydroethanolic (70%) extracts of Eriobotrya japonica [96] and Poacynum hendersonii [97] leaves, respectively, using methanol as solvent in SLH. Moreover, quercetin 3-O-gentiobioside (111) has been finally extracted by application of SLH from hydro-methanolic (70%) fraction of Albizia amara leaf [83] and n-butanol extract of Oryza sativa grain [98] eluting with methanol‒water.
Hydro-methanolic (70%) extracts have been previously obtained from Albizia amara leaf [83] and aerial part of Allium porrum [84], then by using methanol‒water as eluents, quercetin 3-O-α-rhamnopyranoside (113) has been isolated and identified. Three flavonol glucosides consist of quercetin-3-O-α-l-rhap-(1→2)-[α-l-rhap-(1→6)]-β-d-galactopyranoside (114), quercetin-3-O-α-l-rhap-(1→6)-β-d-galactopyranoside (115), and quercetin-3-O-α-l-rhap-(1→2)-α-l-rhamnopyranoside (116) have been isolated from Curcuma longa leaf n-butanol extracts, eluting with methanol‒water (1:1) [91]. Moreover, phytochemical analysis of hydro-methanolic (70%) extract of Allium porrum aerial part was finally led to isolation of quercetin-3-O-β-glucopyranosyl-7-O-α-rhamnopyranoside (117) and quercetin-4′-O-β-glucopyranoside (118), by using methanol‒water as eluting solvents with ratios of 2:8 and 4:6, respectively [84].
Shi et al. [22] isolated quercetin-3,7-di-O-β-d-di-glucopyranoside (119), quercetin-3′,4′,7-trimethyl ether (120), and quercetin-7-O-[β-d-glucopyranosyl(1→6)-β-d-glucopyranoside] (121) from methanolic extract of aerial part of Taraxacum mongolicum (eluent: methanol). From ethyl acetate extracts of two plant species belonging to Asteraceae family including Onopordum alexandrinum seed and Tridax procumbens whole part, quercimeritrin (syn. quercetin-7-O-glucoside) (122) [42] and quercetin-7-O-β-d-glucopyranosyl-(2→1)-α-l-rhamnose (123) [44] have been isolated, respectively; moreover, quercimeritrin (122) was isolated from aqueous extract of Cudrania tricuspidata bark eluting with methanol‒water (1:1) [25].
SLH gel filtration method has also been used for isolation and purification of other glycosylated quercetin derivatives: dihydroquercetin 7-O-β-d-glucoside (124) from leaf n-butanol extract of Curcuma longa (eluent: methanol‒water 1:1) [91], quercetrin (syn. quercetin 3-O-rhamnoside) (125) from leaf butanol extract of Camellia japonica eluting with chloroform‒methanol (1:1) [99], and isoquercetin (syn. quercetin 3-β-O-glucoside) (126) from ethyl acetate fraction of Dorema glabrum aerial part (eluent: methanol‒water 8:2) [100].
Quercitrin (syn. quercetin-3-rhamnoside) (127) has been formerly isolated from Thuja orientalis leaf ethyl acetate extract [58], bran hydroethanolic (95%) extract of Avena sativa [64], and Eriobotrya japonica leaf n-butanol extract [96], whilst methanol was used as eluting solvent. Isoquercitrin (syn. quercetin 3-O-β-d-glucopyranoside) (128) has been previously isolated and identified from n-butanol extracts of Phyllanthus reticulatus leaf [101] and Juniperus chinensis herb [81], using methanol‒water (1:1) and methanol for eluting, respectively. This compound has also been isolated from hydroethanolic (70%) and ethyl acetate fractions yielded from leaves of Poacynum hendersonii [97] and Thuja orientalis [58], respectively, eluting with methanol. Hiring SLH eluting with toluene‒ethanol (7:3) has been concluded to isolate isoquercitrin-6-O-4-hydroxybenzoate (129) from n-butanol extract of Ficus exasperate leaf [92].
Kaempferol (130) (3,4′,5,1-tetrahydroxyflavoune) is an aglycone flavonol which is naturally occurred in many plants’ parts through the phenylpropanoid pathway [102,103]. Pharmacological and biological activities of this nutraceutical compound have been extensively studied and reported to possess significant antiproliferative, cytotoxicity, anti-inflammatory, antioxidant, and antidiabetic activities [104,105,106,107,108,109].
This valuable compound has been isolated from 11 different plant species by employing SLH as the last chromatographic step. Ethyl acetate extracts might be considered as the richest fractions in kaempferol (130) content: from Fragaria ananassa calyx (eluent: acetone‒water 2:1) [74], Gynura divaricate leaf (eluent: chloroform‒methanol 1:1) [75], Gingko biloba leaf (eluent: methanol) [36], Chionanthus retusus flower (eluent: methanol‒water 7:3) [24], Populus davidiana wood (methanol‒water 3:1, 1:1, 1:3) [15], and Leptadenia pyrotechnica aerial parts [67]. Kaempferol (130) has been furtherly isolated from hydro-methanolic (70%) extracts of Albizia amara leaf [83] and Allium porrum aerial part [84] eluting with methanol and methanol‒water (8:2), respectively. From leaf hydroethanolic (70%) extract of Brachychiton acerifolius applying methanol‒water (1:1) as eluting solvent [34], and aqueous fractions of Zygophyllum dumosum shoot [110] and Cudrania tricuspidata bark [25] with methanol through SLH, kaempferol (130) have been also isolated.
Among 23 kaempferol derivatives (131152), only 7,4′-dimethoxykaempferol (131) has been isolated as aglycone analogue from aerial part ethyl acetate extract of Tamarix hohenackeri (Tamaricaceae family) using methanol as eluent in SLH column [77]. The leaf ethanolic and ethyl acetate extracts of Croton zambesicus and Gingko biloba have been previously subjected to various chromatographic methods, and tiliroside (syn. kaempferol-3-O-β-6′’(p-coumaroyl)-glucopyranoside) (132) [56] and kaempferol 3-O-rhamnopyranoside (133) [36] have been accordingly isolated via chloroform‒methanol (9:1) and methanol as SLH eluent, respectively.
Among all the isolated secondary metabolites from leaf n-butanol extract of Curcuma longa, kaempferol-3-O-α-l-rhamnopyranoside (134) has been identified as a glycosylated flavonol exploiting methanol‒water (8:2) for eluting of samples in SLH [91]. Kaempferin (syn. afzelin, Kaempferol-3-rhamnoside) (135) has been previously isolated from two plant species of Eriobotrya japonica [96] and Thuja orientalis [58], whereas their leaves n-butanol and ethyl acetate extracts were chromatographed by SLH with methanol, respectively. Methanol has been used as eluting solvent in isolation and purification of Kaempferol-3-rutinoside (136) from Sideroxylon foetidissimum leaf petroleum ether extract [111] and kaempferol 3-O-α-arabinoside (137) from ethanolic fraction of Opuntia dilleniid flower [112].
Kaouadji et al. [113] isolated kaempferol 3-O-α-l-(2-E-p-coumaroyl rhamnopyranoside) (138) and kaempferol 3-O-α-l-(2-Z-p-coumaroyl rhamnopyranoside) (139) from ethyl acetate extract of buds of Platanus acerifolia by SLH (eluent: methanol). In a phytochemical investigation carried out on Nelumbo nucifera, the ethyl acetate extract of stem by utilization of methanol in SLH gel filtration column kaempferol 3-O-α-l-rhamnopyranosyl-(1→6)-β-d-glucopyranoside (140), kaempferol 3-O-β-(2″-O-α-rhamnosyl)-glucuronide (141), and kaempferol 3-O-α-l-rhamnopyranosyl-(1→2)-β-d-glucopyranoside (142), and kaempferol 3-O-β-d-glucuronopyranoside (143) have been isolated and purified [95].
SLH has been able to isolate astragalin (syn. kaempferol 3-O-β-d-glucopyranoside) (144) from five plant species. Hydro-methanolic (70%) and hydroethanolic (95%) extracts of Allium porrum aerial part [84] and bran part of Avena sativa [64] have been applied to isolate astragalin (144) applying methanol‒water (6:4) and methanol as eluting solvent, respectively. Furthermore, aerial parts ethyl acetate extracts of Leptadenia pyrotechnica [67] and Dorema glabrum [100], and Fragaria ananassa calyx (eluent: acetone‒water 7:3) [74] comprised the aforementioned compound.
From aerial part ethyl acetate extract of Leptadenia pyrotechnica kaempferol-3-O-α-l-rhamnopyranosyl (1″′→6″)-O-β-d-glucopyranoside (145) and kaempferol-3-O-β-d-glucopyranosyl (1″′→6″)-O-β-d-glucopyranoside (146) [67], whereas kaempferol 3-O-(3″-E-p-coumaroyl)-α-l-rhamnopyranoside (147) and kaempferol 3-O-(2″-O-E-p-coumaroyl)-β-d-glucopyranoside (148) were also isolated and identified from bran hydroethanolic (95%) extract of Avena sativa (eluent: methanol) [64].
Rayyan et al. [16] reported isolation of a novel kaempferol glucoside, namely 8-methoxykaempferol 3-O-(6″-malonyl-β-glucopyranoside) (149) from hydro-methanolic (80%) extract of leaf and flower parts of Crataegus spp. (Hawthorn) by increasing ratio of methanol (40 to 70%) in water using SLH.
According to previously performed phytochemical studies, three other glycosylated kaempferol derivatives have been furtherly isolated and purified by SLH gel filtration: kaempferol 7-O-glucoside (150) from seed ethyl acetate extract of Onopordum alexandrinum (eluent: methanol‒water 9:1) [42], kaempferol 7-O-β-glucopyranoside (151) from hydro-methanolic (70%) fraction of Allium porrum aerial part (eluent: methanol‒water 6:4) [84], and kaempferol 7-O-α-l-rhamnopyranoside (152) from bran hydro-ethanolic (95%) extract of Avena sativa (eluent: methanol) [64].
Isorhamnetin (153) has been isolated by using SLH eluting with methanol‒water (8:2) from hydro-methanolic (70%) extract of Allium porrum aerial parts [84]. Three isorhamnetin glucosides consist of isorhamnetin 3-O-β-d-rutinoside (154) from aerial part ethyl acetate extract of Halimodendron halodendron (eluent: chloroform‒methanol 1:1) [79] and flower ethanolic fraction of Opuntia dillenii (eluent: methanol) [112], isorhamnetin 3-O-monoglucoside (155) from n-butanol extract of Sambucus ebulus leaf (eluent: methanol) [93], along with isorhamnetin 3-O-β-d-glucopyranoside (156) from Dorema glabrum aerial part ethyl acetate extract (eluent: methanol‒water 8:2) [100].
Exploiting SLH by eluting acetone‒methanol (1:1), myricetin (syn. 3,5,7,3′,4′,5′-hexahydroxyflavone) (157) and myricetin-3-O-β-galactopyranoside (160) have been isolated from ethyl acetate extract of Bauhinia galpinii leaf [66]. Moreover, from stamen ethyl acetate, a fraction of Nelumbo nucifera, myricetin 3′,5′-dimethylether 3-O-β-d-glucopyranoside (158) (eluent: methanol) [114], and a novel secondary metabolite myricetin 7-methylether 3-O-xylopyranosylsyl-(1→2)-α-rhamnopyranoside (159) have been previously isolated and identified from Eugenia jambos ethanolic extract of the leaf (eluent: ethanol‒water 3:7) [94].
By eluting methanol‒water and pure methanol through SLH column, myricitrin (syn. myricetin 3-O-α-rhamnopyranoside) (161) has been isolated from hydro-methanolic (70%) and ethyl acetate extracts of Albizia amara [83] and Thuja orientalis [58], respectively. Another study reported SLH was able to isolate penduletin (162) and chrysosplenol D (163) from aerial part methanolic extract of Plectranthus cylindraceus [115].
More flavonol derivatives have also been isolated and purified from ethyl acetate extracts of diverse plant species: sexangularetin (164) from calyx of Fragaria ananassa eluting with methanol‒water (4:1) [74], a new natural product brassicin-4′-O-β-d-glucopyranoside (165) via increasing acetone ratio (33 to 100%) in water from Oryza sativa spp. japonica grain [116], 5,7,3′-trimethyl-4′-methoxyl-3-O-β-d-flavonoid glucoside (166) and 8,3′-dihydroxyl-3,7,4′-trimethoxy-6-O-β-d-flavonoid glucoside (167) from whole part of Tridax procumbens [44], a novel phytochemical ptevon-3-d-glucoside (168) from Pterocarpus indicus leaf (eluent: dichloromethane‒methanol 1:1) [117], leonurusoide E (170) from Leonurus japonicus eluting with methanol‒water (4:6) [118], dillenetin (172) from Tamarix hohenackeri aerial part (eluent: methanol‒water) [77], and tamarixetin 3-O-rhamnopyranoside (175) from Firmiana simplex stem bark (eluent: methanol) [119].
Furthermore, sophoflavescenol (169) from root dichloromethane extract of Sophora flavescens (eluent: dichloromethane‒methanol) [120], 5,4′-dihydroxyflavone-3,6-di-O-β-d-glucoside-7-O-β-d-glucuronide (171) from Carthamus tinctorius aqueous flower fraction (eluent: water) [121], 7-hydroxy-6-methoxyflavone (173) from herb chloroform extract of Dalbergia cochinchinensis (dichloromethane‒methanol 1:1) [12], 3-O-demethyldigicitrin (174) from ethanolic extract of Athrixia phylicoides aerial part (eluent: methanol), and artemitin (176) from methanolic fraction of Taraxacum mongolicum [22] have been formerly isolated and purified by application of SLH as the final separation step.

2.6. Homoisoflavonoid Derivatives

Homoisoflavonoids are naturally occurred mostly in Asparagaceae and Fabaceae families. Several research groups reported their antimicrobial, antidiabetic, cytotoxic, anticancer, anti-inflammatory, antimutagenic, etc. properties [122,123].
In a previous phytochemical investigation performed on hydroethanolic (60%) extract obtained from the rhizome of Polygonatum odoratum (Asparagaceae family), SLH gel filtration column eluted by acetonitrile‒methanol (1:1) was applied to isolate 10 homoisoflavonoids (177186) including three novel natural products of (3R)-5,7-dihydroxy-8-methyl-3-(2′,4′-dihydroxybenzyl)-chroman-4-one (177), (3R)-5,7-dihydroxy-8-methyl-3-(4′-hydroxybenzyl)-chroman-4-one (181), and (3R)-5,7-dihydroxy-3-(2′-hydroxy-4′-methoxybenzyl)-chroman-4-one (182); whereas (3R)-5,7-dihydroxy-6-methoxy-8-methyl-3-(2′,4′-dihydroxybenzyl)-chroman-4-one (178), (3R)-5,7-dihydroxy-3-(4′-hydroxybenzyl)-chroman-4-one (179), (3R)-5,7-dihydroxy-8-methoxy-3-(2′-hydroxy-4′-methoxybenzyl)-chroman-4-one (180), (3R)-5,7-dihydroxy-6-methyl-3-(4′-hydroxybenzyl)-chroman-4-one (183), (3R)-5,7-dihydroxy-6-methyl-8-methoxy-3-(4′-hydroxybenzyl)-chroman-4-one (184), (3R)-5,7-dihydroxy-6,8-dimethyl-3-(4′-hydroxybenzyl)-chroman-4-one (185), and (3R)-5,7-dihydroxy-6-methyl-8-methoxy-3-(4′-methoxybenzyl)-chroman-4-one (186) have been isolated as known homoisoflavonoid analogous [124].

2.7. Proanthocyanidins

Proanthocyanidins are condensed tannins, considered as the end product of flavonoid biosynthetic pathway with various health characteristic advantages, for instance, antioxidant, anticancer, antidiabetic, neuroprotective, and antimicrobial potencies, and the treatment of cardiovascular disease [125,126].
Utilization of SLH has led to isolate 3 proanthocyanidin derivatives from two Lauraceae species. Cinnamtannin B1 (syn. epicatechin-(2β→O-7,4β→8)-epicatechin-(4β→8) epicatechin) (187) has been isolated from herb ethyl acetate extract of Lindera glauca [127] and aqueous fraction of Cinnamomum zeylanicum fruit [89] by hiring methanol‒water as eluting solvent. Huh et al. [127] reported isolation of two proanthocyanidins of cinnamtannin D1 (188) and procyanidin A1 (189) from herb ethyl acetate of Lindera glauca by application of SLH eluting with methanol‒water 1:1, and 1:1 to 5:1 of this solvent mixture, respectively.

3. Conclusions

Nowadays, new separation techniques have been established to facilitate analysis of natural products. The isolation and purification of flavonoids as one of the most valuable natural compounds have been carried out by applying several classical and recently developed methods. SLH as a type of size-exclusion chromatography (syn. gel filtration) has been broadly used in isolation or purification of flavonoid analogous.
The present context overviewed the role of SLH in isolation or purification of flavonoids as the final chromatographic step. This review for the first time provides valuable information about the classification of isolated flavonoids, plant families and species, the used plant parts and extracts, and applied eluents utilized in SLH gel filtration chromatography.
In brief, SLH has been able to isolate or purify 189 flavonoids categorized in seven classes, mostly comprised of 79 flavonols and 63 flavones. Notably, six flavones (60,61,74,75,78,88), four flavonols (149,159,165,168), three homoisoflavonoids (177,181,182), one flavanone (8), a flavanol (26), and an isoflavanol (29) have been isolated as the novel secondary metabolites in nature. The Asteraceae possessing 22 flavone and 13 flavonol derivatives has been documented as the richest plant family, which was subjected to finally SLH for isolation or purification of their flavonoids. Homoisoflavonoids and proanthocyanidins have been only isolated from the Asparagaceae and Lauraceae families, respectively. Furthermore, methanol and methanol‒water has been majorly applied as eluents to perform the separation process.
In general, the flavonoids have mainly been isolated or purified from hydro-alcoholic and ethyl acetate extracts. The flavan and isoflavan compounds have been isolated from chloroform extracts. Eleven flavanones (from totally 18), were isolated from ethyl acetate extracts, while methanol fractions were applied for isolation of four derivatives. Aqueous extracts were the richest in flavanol compounds (six of eight). Thirty flavones from ethyl acetate extracts, 17 derivatives from hydro-ethanolic (50 to 95%), and 11 compounds from methanolic extracts have been isolated or purified.
From totally 79 flavonols, combination of methanol and water have majorly been used for isolation or purification of the most flavonols, where 19 compounds from different ratios, and 15 derivatives from the equivalent ratio (1:1) of this mixture were isolated. Hydro-ethanolic (60%) extract was applied for isolation of all homoisoflavonoids, while all four proanthocyanidins were isolated from ethyl acetate extracts.

Supplementary Materials

The following are available online at https://www.mdpi.com/1420-3049/25/18/4146/s1, Table S1. Isolated or purified flavonoid derivatives by utilizing Sephadex® LH-20 from diverse plant families.

Author Contributions

Conceptualization, methodology, and writing was carried out by J.M. and M.I. revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Panche, A.N.; Diwan, A.D.; Chandra, S.R. Flavonoids: An overview. J. Nutr. Sci. 2016, 5, 1–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Feng, W.; Hao, Z.; Li, M. Isolation and structure identification of flavonoids. In Flavonoids—From Biosynthesis to Human Health; InTech: London, UK, 2017. [Google Scholar]
  3. Amarowicz, R.; Kolodziejczyk, P.P.; Pegg, R.B. Chromatographic separation of phenolic compounds from rapeseed by a Sephadex LH-20 column with ethanol as the mobile phase. J. Liq. Chromatogr. Relat. Technol. 2003, 26, 2157–2165. [Google Scholar] [CrossRef]
  4. Markham, K.R. Isolation techniques for flavonoids. In The Flavonoids; Springer US: Boston, MA, USA, 1975; pp. 1–44. [Google Scholar]
  5. Chávez-González, M.L.; Sepúlveda, L.; Verma, D.K.; Luna-García, H.A.; Rodríguez-Durán, L.V.; Ilina, A.; Aguilar, C.N. Conventional and emerging extraction processes of flavonoids. Processes 2020, 8, 434. [Google Scholar] [CrossRef] [Green Version]
  6. Putnik, P.; Kovačević, D.B.; Radojčin, M.; Dragović-Uzelaca, V. Influence of acidity and extraction time on the recovery of flavonoids from grape skin pomace optimized by Response Surface Methodology. Chem. Biochem. Eng. Q. J. 2017, 30, 455–464. [Google Scholar] [CrossRef]
  7. Roselló-Soto, E.; Martí-Quijal, F.; Cilla, A.; Munekata, P.; Lorenzo, J.; Remize, F.; Barba, F. Influence of temperature, solvent and pH on the selective extraction of phenolic compounds from tiger nuts by-products: Triple-TOF-LC-MS-MS characterization. Molecules 2019, 24, 797. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Hameed, B.S.; Bhatt, C.S.; Nagaraj, B.; Suresh, A.K. Chromatography as an efficient technique for the separation of diversified nanoparticles. In Nanomaterials in Chromatography; Elsevier: Amsterdam, The Netherlands, 2018; pp. 503–518. [Google Scholar]
  9. Lindqvist, B.; Storgårds, T. Molecular-sieving properties of starch. Nature 1955, 175, 511–512. [Google Scholar] [CrossRef]
  10. Hong, P.; Koza, S.; Bouvier, E.S.P. Size-exclusion chromatography for the analysis of protein biotherapeutics and their aggregates. J. Liq. Chromatogr. Relat. Technol. 2012, 35, 2923–2950. [Google Scholar] [CrossRef] [Green Version]
  11. Flodin, P.G.M. Process for Preparing Hydrophilic Copolymerization and Product Obtained Thereby. U.S. Patent 3208994A, 28 September 1965. [Google Scholar]
  12. Liu, R.; Wen, X.; Shao, F.; Zhang, P.; Huang, H.; Zhang, S. Flavonoids from heartwood of Dalbergia cochinchinensis. Chinese Herb. Med. 2016, 8, 89–93. [Google Scholar] [CrossRef]
  13. Bergeron, C.; Marston, A.; Hakizamungu, E.; Hostettmann, K. Antifungal constituents of Chenopodium procerum. Int. J. Pharmacogn. 1995, 33, 115–119. [Google Scholar] [CrossRef]
  14. Si, C.L.; Lu, Y.Y.; Qin, P.P.; Sun, R.C.; Ni, Y.H. Phenolic extractives with chemotaxonomic significance from the bark of Paulownia tomentosa var. Tomentosa. BioResources 2011, 6, 5086–5098. [Google Scholar]
  15. Si, C.-L.; Xu, Q.; Li, S.-M.; Liu, Z. Phenolic compounds from Populus davidiana wood. Chem. Nat. Compd. 2009, 45, 634–636. [Google Scholar] [CrossRef]
  16. Rayyan, S.; Fossen, T.; Nateland, H.S.; Andersen, Ø.M. Isolation and identification of flavonoids, including flavone rotamers, from the herbal drug ‘crataegi folium cum flore’ (hawthorn). Phytochem. Anal. 2005, 16, 334–341. [Google Scholar] [CrossRef] [PubMed]
  17. Jung, J.-W.; Ko, W.-M.; Park, J.-H.; Seo, K.-H.; Oh, E.-J.; Lee, D.-Y.; Lee, D.-S.; Kim, Y.-C.; Lim, D.-W.; Han, D.; et al. Isoprenylated flavonoids from the root bark of Morus alba and their hepatoprotective and neuroprotective activities. Arch. Pharm. Res. 2015, 38, 2066–2075. [Google Scholar] [CrossRef] [PubMed]
  18. Jung, J.-W.; Park, J.-H.; Lee, Y.-G.; Seo, K.-H.; Oh, E.-J.; Lee, D.-Y.; Lim, D.-W.; Han, D.; Baek, N.-I. Three new isoprenylated flavonoids from the root bark of Morus alba. Molecules 2016, 21, 1112. [Google Scholar] [CrossRef]
  19. Liu, B.; Yan, T.; Xiao, J.; Wang, X. α-Glucosidase inhibitors and antioxidants from root bark of Morus alba. Chinese Herb. Med. 2018, 10, 331–335. [Google Scholar] [CrossRef]
  20. Lan, X.; Wang, W.; Li, Q.; Wang, J. The natural flavonoid pinocembrin: Molecular targets and potential therapeutic applications. Mol. Neurobiol. 2016, 53, 1794–1801. [Google Scholar] [CrossRef] [Green Version]
  21. Lan, X.; Han, X.; Li, Q.; Li, Q.; Gao, Y.; Cheng, T.; Wan, J.; Zhu, W.; Wang, J. Pinocembrin protects hemorrhagic brain primarily by inhibiting toll-like receptor 4 and reducing M1 phenotype microglia. Brain. Behav. Immun. 2017, 61, 326–339. [Google Scholar] [CrossRef] [Green Version]
  22. Shi, S.; Zhao, Y.; Zhou, H.; Zhang, Y.; Jiang, X.; Huang, K. Identification of antioxidants from Taraxacum mongolicum by high-performance liquid chromatography–diode array detection–radical-scavenging detection–electrospray ionization mass spectrometry and nuclear magnetic resonance experiments. J. Chromatogr. A 2008, 1209, 145–152. [Google Scholar] [CrossRef]
  23. Tanagornmeatar, K.; Chaotham, C.; Sritularak, B.; Likhitwitayawuid, K.; Chanvorachote, P. Cytotoxic and anti-metastatic activities of phenolic compounds from Dendrobium ellipsophyllum. Anticancer Res. 2014, 34, 6573–6580. [Google Scholar]
  24. Lee, Y.-G.; Lee, H.; Jung, J.-W.; Seo, K.-H.; Lee, D.Y.; Kim, H.-G.; Ko, J.-H.; Lee, D.-S.; Baek, N.-I. Flavonoids from Chionanthus retusus (Oleaceae) flowers and their protective effects against glutamate-induced cell toxicity in HT22 cells. Int. J. Mol. Sci. 2019, 20, 3517. [Google Scholar] [CrossRef] [Green Version]
  25. Zheng, Z.-P.; Liang, J.-Y.; Hu, L.-H. Water-soluble constituents of Cudrania tricuspidata (Carr.) Bur. J. Integr. Plant Biol. 2006, 48, 996–1000. [Google Scholar] [CrossRef]
  26. Park, J.-H.; Jung, Y.-J.; Jung, J.-W.; Shrestha, S.; Lim, D.W.; Han, D.; Baek, N.-I. A new flavonoid glycoside from the root bark of Morus alba L. Nat. Prod. Res. 2014, 28, 1859–1863. [Google Scholar] [CrossRef] [PubMed]
  27. Awouafack, M.D.; Tchuenguem, R.T.; Ito, T.; Dzoyem, J.P.; Tane, P.; Morita, H. A new isoflavanol from the fruits of Kotschya strigosa (Fabaceae). Helv. Chim. Acta 2016, 99, 321–324. [Google Scholar] [CrossRef]
  28. Xuan, L.; Bin-Feng, Z.; Li, Y.; Gui-Xin, C.; Zheng-Tao, W. Two new chromones and a new flavone glycoside from Imperata cylindrica. Chin. J. Nat. Med. 2013, 11, 77–80. [Google Scholar]
  29. Jung, M.J.; Chung, H.Y.; Kang, S.S.; Choi, J.H.; Bae, K.S.; Choi, J.S. Antioxidant activity from the stem bark of Albizzia julibrissin. Arch. Pharm. Res. 2003, 26, 458–462. [Google Scholar] [CrossRef] [PubMed]
  30. Mavundza, E.J.; Tshikalange, T.E.; Lall, N.; Hussein, A.A.; Mudau, F.N.; Meyer, J.J.M. Antioxidant activity and cytotoxicity effect of flavonoids isolated from Athrixia phylicoides. J. Med. Plants Res. 2010, 4, 2584–2587. [Google Scholar]
  31. Imran, M.; Rauf, A.; Abu-Izneid, T.; Nadeem, M.; Shariati, M.A.; Khan, I.A.; Imran, A.; Orhan, I.E.; Rizwan, M.; Atif, M.; et al. Luteolin, a flavonoid, as an anticancer agent: A review. Biomed. Pharmacother. 2019, 112, 108612. [Google Scholar] [CrossRef]
  32. Luo, Y.; Shang, P.; Li, D. Luteolin: A flavonoid that has multiple cardio-protective effects and its molecular mechanisms. Front. Pharmacol. 2017, 8, 692. [Google Scholar] [CrossRef] [Green Version]
  33. Lin, Y.; Shi, R.; Wang, X.; Shen, H.-M. Luteolin, a flavonoid with potential for cancer prevention and therapy. Curr. Cancer Drug Targets 2008, 8, 634–646. [Google Scholar] [CrossRef]
  34. Abou-Zeid, A.H.; Farag, M.A.; Hamed, M.A.A.; Kandil, Z.A.A.; El-Akad, R.H.; El-Rafie, H.M. Flavonoid chemical composition and antidiabetic potential of Brachychiton acerifolius leaves extract. Asian Pac. J. Trop. Biomed. 2017, 7, 389–396. [Google Scholar] [CrossRef]
  35. Erenler, R.; Sen, O.; Yildiz, I.; Aydın, A. Antiproliferative activities of chemical constituents isolated from Thymus praecox subsp. grossheimii (ronniger) jalas. Rec. Nat. Prod. 2016, 10, 766–770. [Google Scholar]
  36. Hyun, S.K.; Jung, H.A.; Chung, H.Y.; Choi, J.S. In vitro peroxynitrite scavenging activity of 6-hydroxykynurenic acid and other flavonoids from Gingko biloba yellow leaves. Arch. Pharm. Res. 2006, 29, 1074–1079. [Google Scholar] [CrossRef] [PubMed]
  37. Hyun, H.B.; Shrestha, S.; Boo, K.H.; Cho, S.K. Evaluation of antioxidant potential of ethyl acetate fraction of Rosmarinus officinalis L. and its major components. J. Korean Soc. Appl. Biol. Chem. 2015, 58, 715–722. [Google Scholar] [CrossRef]
  38. Sándor, Z.; Mottaghipisheh, J.; Veres, K.; Hohmann, J.; Bencsik, T.; Horváth, A.; Kelemen, D.; Papp, R.; Barthó, L.; Csupor, D. Evidence supports tradition: The in vitro effects of roman chamomile on smooth muscles. Front. Pharmacol. 2018, 9, 323. [Google Scholar] [CrossRef]
  39. Taiwo, B.J.; Obuotor, E.; Onawunmi, G.O.; Ogundaini, A.O. Radical scavenging compounds from the aerial parts of Solenostemon monostachys briq (Lamiaceae). African J. Tradit. Complement. Altern. Med. 2015, 12, 140. [Google Scholar] [CrossRef] [Green Version]
  40. Sajjadi, S.; Delazari, Z.; Aghaei, M.; Ghannadian, M. Flavone constituents of Phlomis bruguieri Desf. with cytotoxic activity against MCF-7 breast cancer cells. Res. Pharm. Sci. 2018, 13, 422. [Google Scholar] [PubMed]
  41. Si, C.-L.; Lu, Y.-Y.; Zhang, Y.; Xu, J.; Qin, P.-P.; Sun, R.-C.; Ni, Y.-H. Antioxidative low molecular weight extractives from triploid Populus tomentosa xylem. BioResources 2011, 6, 232–242. [Google Scholar]
  42. Abdallah, W.E.; Elsayed, W.M.; Hassanein, H.D.; Shams, K.A.; Ismail, S.I.; Ali, S.M. Phytochemical and biological investigations of Onopordum alexandrinum seeds. Res. J. Pharm. Biol. Chem. Sci. 2015, 6, 361–374. [Google Scholar]
  43. Tan, J.; Shenghua, L.; Zeng, J.; Wu, X. Anti-oxidant and hepatoprotective activities of total flavonoids of Indocalamus latifolius. Bangladesh J. Pharmacol. 2015, 10, 779. [Google Scholar] [CrossRef]
  44. Cui, H.-X.; Zhang, L.-S.; Yan, H.-G.; Yuan, K.; Jin, S.-H. Constituents of flavonoids from Tridax procumbens L. and antioxidant activity. Pharmacogn. Mag. 2020, 16, 201. [Google Scholar]
  45. Gohari, A.R.; Ebrahimi, H.; Saeidnia, S.; Foruzani, M.; Ebrahimi, P.; Ajani, Y. Flavones and flavone glycosides from Salvia macrosiphon Boiss. Iran. J. Pharm. Res. IJPR 2011, 10, 247–251. [Google Scholar] [PubMed]
  46. Komaki, E.; Yamaguchi, S.; Maru, I.; Kinoshita, M.; Kakehi, K.; Ohta, Y.; Tsukada, Y. Identification of anti-α-amylase components from olive leaf extracts. Food Sci. Technol. Res. 2003, 9, 35–39. [Google Scholar] [CrossRef] [Green Version]
  47. Hwang, J.-H.; Yang, H.-S.; Ra, K.S.; Park, S.S.; Yu, K.-W. Intestinal immune system-modulating activity through peyer’s patch of flavonoid glycoside purified from Citrus unshiu peel. J. Food Biochem. 2013, 37, 151–160. [Google Scholar] [CrossRef]
  48. Salehi, B.; Venditti, A.; Sharifi-Rad, M.; Kręgiel, D.; Sharifi-Rad, J.; Durazzo, A.; Lucarini, M.; Santini, A.; Souto, E.; Novellino, E.; et al. The therapeutic potential of apigenin. Int. J. Mol. Sci. 2019, 20, 1305. [Google Scholar] [CrossRef] [Green Version]
  49. Shankar, E.; Goel, A.; Gupta, K.; Gupta, S. Plant flavone apigenin: An emerging anticancer agent. Curr. Pharmacol. Reports 2017, 3, 423–446. [Google Scholar] [CrossRef]
  50. Ali, F.; Rahul-Naz, F.; Jyoti, S.; Siddique, Y.H. Health functionality of apigenin: A review. Int. J. Food Prop. 2017, 20, 1197–1238. [Google Scholar] [CrossRef]
  51. Yan, X.; Qi, M.; Li, P.; Zhan, Y.; Shao, H. Apigenin in cancer therapy: Anti-cancer effects and mechanisms of action. Cell Biosci. 2017, 7, 50. [Google Scholar] [CrossRef] [Green Version]
  52. Zhou, X.; Wang, F.; Zhou, R.; Song, X.; Xie, M. Apigenin: A current review on its beneficial biological activities. J. Food Biochem. 2017, 41, e12376. [Google Scholar] [CrossRef]
  53. Sun, J.; He, X.-M.; Zhao, M.-M.; Li, L.; Li, C.-B.; Dong, Y. Antioxidant and nitrite-scavenging capacities of phenolic compounds from sugarcane (Saccharum officinarum L.) tops. Molecules 2014, 19, 13147–13160. [Google Scholar] [CrossRef]
  54. Luo, P.; Zhang, Z.; Yi, T.; Zhang, H.; Liu, X.; Mo, Z. Anti-inflammatory activity of the extracts and fractions from Erigeron multiradiatus through bioassay-guided procedures. J. Ethnopharmacol. 2008, 119, 232–237. [Google Scholar] [CrossRef]
  55. Zielińska-Pisklak, M.A.; Kaliszewska, D.; Stolarczyk, M.; Kiss, A.K. Activity-guided isolation, identification and quantification of biologically active isomeric compounds from folk medicinal plant Desmodium adscendens using high performance liquid chromatography with diode array detector, mass spectrometry and multidiment. J. Pharm. Biomed. Anal. 2015, 102, 54–63. [Google Scholar] [CrossRef] [PubMed]
  56. Aderogba, M.A.; McGaw, L.J.; Bezabih, M.; Abegaz, B.M. Isolation and characterisation of novel antioxidant constituents of Croton zambesicus leaf extract. Nat. Prod. Res. 2011, 25, 1224–1233. [Google Scholar] [CrossRef] [PubMed]
  57. CHEN, D.; BI, D.; SONG, Y.-L.; TU, P.-F. Flavanoids from the stems of Aquilaria sinensis. Chin. J. Nat. Med. 2012, 10, 287–291. [Google Scholar] [CrossRef]
  58. Lee, E.H. Flavonoids from the leaves of Thuja orientalis inhibit the aldose reductase and the formation of advanced glycation endproducts. J. Korean Soc. Appl. Biol. Chem. 2009, 52, 448–455. [Google Scholar] [CrossRef]
  59. Lu, M.; Kong, Q.; Xu, X.; Lu, H.; Lu, Z.; Yu, W.; Zuo, B.; Su, J.; Guo, R. Pectolinarigenin—A flavonoid compound from Cirsium Japonicum with potential anti-proliferation activity in MCF- 7 breast cancer cell. Trop. J. Pharm. Res. 2014, 13, 225. [Google Scholar] [CrossRef] [Green Version]
  60. Mirzahosseini, G.; Manayi, A.; Khanavi, M.; Safavi, M.; Salari, A.; Madjid-Ansari, A.; San’ati, H.; Vazirian, M. Bio-guided isolation of Centaurea bruguierana subsp. belangerana cytotoxic components. Nat. Prod. Res. 2019, 33, 1687–1690. [Google Scholar] [CrossRef] [PubMed]
  61. Zhou, J.-M.; Ibrahim, R.K. Tricin—A potential multifunctional nutraceutical. Phytochem. Rev. 2010, 9, 413–424. [Google Scholar] [CrossRef]
  62. Matsuta, T.; Sakagami, H.; Satoh, K.; Kanamoto, T.; Terakubo, S.; Nakashima, H.; Kitajima, M.; Oizumi, H.; Oizumi, T. Biological activity of luteolin glycosides and tricin from Sasa senanensis Rehder. In Vivo 2011, 25, 757–762. [Google Scholar]
  63. Mu-Xin, Z.; LIU, Y.-Y.; Wei, S.; Xiao-Hong, Y.; Guang-Shu, W. Isolation and identification of novel flavonoids from the bract of Zea mays L. Chem. J. Chin. Univ. 2011, 32, 2554–2557. [Google Scholar]
  64. ZHANG, W.-K.; XU, J.-K.; ZHANG, L.; DU, G.-H. Flavonoids from the bran of Avena sativa. Chin. J. Nat. Med. 2012, 10, 110–114. [Google Scholar] [CrossRef]
  65. Sala, A.; Recio, M.C.; Giner, R.M.; Manez, S.; Rios, J.L. Anti-phospholipase A(2) and anti-inflammatory activity of Santolina chamaecyparissus. LIFE Sci. 2000, 66, PL35–PL40. [Google Scholar] [CrossRef]
  66. Ahmed, A.S.; Moodley, N.; Eloff, J.N. Bioactive compounds from the leaf extract of Bauhinia galpinii (Fabaceae) used as antidiarrhoeal therapy in southern Africa. S. Afr. J. Bot. 2019, 126, 345–353. [Google Scholar] [CrossRef]
  67. Youssef-Moustafa, A.M.; Khodair, A.I.; Saleh, M.A. Isolation, structural elucidation of flavonoid constituents from Leptadenia pyrotechnica and evaluation of their toxicity and antitumor activity. Pharm. Biol. 2009, 47, 539–552. [Google Scholar] [CrossRef] [Green Version]
  68. Zhao, H.-Y.; Yang, L.; Wei, J.; Huang, M.; Jiang, J.-G. Bioactivity evaluations of ingredients extracted from the flowers of Citrus aurantium L. var. amara Engl. Food Chem. 2012, 135, 2175–2181. [Google Scholar] [CrossRef] [PubMed]
  69. Kim, S.B.; Hwang, S.H.; Wang, Z.; Yu, J.M.; Lim, S.S. Rapid identification and isolation of inhibitors of rat lens aldose reductase and antioxidant in Maackia amurensis. Biomed Res. Int. 2017, 2017, 1–10. [Google Scholar]
  70. Anand-David, A.V.; Arulmoli, R.; Parasuraman, S. Overviews of biological importance of quercetin: A bioactive flavonoid. Pharmacogn. Rev. 2016, 10, 84–89. [Google Scholar]
  71. Formica, J.V.; Regelson, W. Review of the biology of quercetin and related bioflavonoids. Food Chem. Toxicol. 1995, 33, 1061–1080. [Google Scholar] [CrossRef]
  72. Rauf, A.; Imran, M.; Khan, I.A.; Ur-Rehman, M.-; Gilani, S.A.; Mehmood, Z.; Mubarak, M.S. Anticancer potential of quercetin: A comprehensive review. Phyther. Res. 2018, 32, 2109–2130. [Google Scholar] [CrossRef]
  73. Deng, Q.; Li, X.X.; Fang, Y.; Chen, X.; Xue, J. Therapeutic potential of quercetin as an antiatherosclerotic agent in atherosclerotic cardiovascular disease: A review. Evidence-Based Complement. Altern. Med. 2020, 2020, 1–12. [Google Scholar] [CrossRef]
  74. Baek, Y.-S.; Song, N.-Y.; Nam, T.-G.; Kim, D.-O.; Kang, H.-C.; Kwon, O.-K.; Baek, N.-I. Flavonoids from Fragaria ananassa calyx and their antioxidant capacities. J. Korean Soc. Appl. Biol. Chem. 2015, 58, 787–793. [Google Scholar] [CrossRef]
  75. Cao, S.; Wan, C.; Yu, Y.; Zhou, S.; Tian, S. Isolation and identification of phenolic compounds from Gynura divaricata leaves. Pharmacogn. Mag. 2011, 7, 101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Huang, W.; Wan, C.; Zhou, S. Quercetin - a flavonoid compound from Sarcopyramis bodinieri var. delicate with potential apoptotic activity in HepG2 liver cancer cells. Trop. J. Pharm. Res. 2013, 12, 529–533. [Google Scholar]
  77. Liao, J.; Tang, Y.; Tan, C.; Ni, H.; Wu, X.; Li, N.; Jia, X.; Xing, Y.; Zhang, P. ACE and platelet aggregation inhibitors from Tamarix hohenackeri Bunge (host plant of Herba Cistanches) growing in Xinjiang. Pharmacogn. Mag. 2014, 10, 111. [Google Scholar] [CrossRef] [PubMed]
  78. Lin, L.; Huang, X.; Lv, Z. Isolation and identification of flavonoids components from Pteris vittata L. Springerplus 2016, 5, 1649. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Wang, J.; Gao, H.; Zhao, J.; Wang, Q.; Zhou, L.; Han, J.; Yu, Z.; Yang, F. Preparative separation of phenolic compounds from Halimodendron halodendron by high-speed counter-current chromatography. Molecules 2010, 15, 5998–6007. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Ahmadu, A.A.; Hassan, H.S.; Abubakar, M.; Akpulu, I.N. Flavonoid glycosides from Byrsocarpus coccineus leaves. Schum and Thonn (Connaraceae). African J. Tradit. Complement. Altern. Med. 2007, 4, 257–260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Lim, J.P.; Song, Y.C.; Kim, J.W.; Ku, C.H.; Eun, J.S.; Leem, K.H.; Kim, D.K. Free radical scavengers from the heartwood of Juniperus chinensis. Arch. Pharm. Res. 2002, 25, 449–452. [Google Scholar] [CrossRef]
  82. Jarial, R.; Shard, A.; Thakur, S.; Sakinah, M.; Zularisam, A.W.; Rezania, S.; Kanwar, S.S.; Singh, L. Characterization of flavonoids from fern Cheilanthes tenuifolia and evaluation of antioxidant, antimicrobial and anticancer activities. J. King Saud Univ. Sci. 2018, 30, 425–432. [Google Scholar] [CrossRef]
  83. Kassem, M.E.S.; Ibrahim, L.F.; Hussein, S.R.; El-Sharawy, R.; El-Ansari, M.A.; Hassanane, M.M.; Booles, H.F. Myricitrin and bioactive extract of Albizia amara leaves: DNA protection and modulation of fertility and antioxidant-related genes expression. Pharm. Biol. 2016, 54, 2404–2409. [Google Scholar] [CrossRef] [Green Version]
  84. Mahmoud, A.H.; Foda, D.S.; Ibrahim, N.E.; El-Hagrassi, A.M.; Yousef, O.M.H. Secondary metabolites and biological activities of Allium porrum L. attack Ehrlich ascites carcinoma in mice. Egypt. J. Chem. 2019, 62, 211–227. [Google Scholar]
  85. Yuenyongsawad, S.; Bunluepuech, K.; Wattanapiromsakul, C.; Tewtrakul, S. Anti-cancer activity of compounds from Bauhinia strychnifolia stem. J. Ethnopharmacol. 2013, 150, 765–769. [Google Scholar] [CrossRef]
  86. Gullón, B.; Lú-Chau, T.A.; Moreira, M.T.; Lema, J.M.; Eibes, G. Rutin: A review on extraction, identification and purification methods, biological activities and approaches to enhance its bioavailability. Trends Food Sci. Technol. 2017, 67, 220–235. [Google Scholar] [CrossRef]
  87. Al-Dhabi, N.A.; Arasu, M.V.; Park, C.H.; Park, S.U. An up-to-date review of rutin and its biological and pharmacological activities. EXCLI J. 2015, 14, 59–63. [Google Scholar]
  88. Ganeshpurkar, A.; Saluja, A.K. The pharmacological potential of rutin. Saudi Pharm. J. 2017, 25, 149–164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Jayaprakasha, G.K.; Ohnishi-Kameyama, M.; Ono, H.; Yoshida, M.; Jaganmohan-Rao, L. Phenolic constituents in the fruits of Cinnamomum zeylanicum and their antioxidant activity. J. Agric. Food Chem. 2006, 54, 1672–1679. [Google Scholar] [CrossRef] [PubMed]
  90. Erhirhie, E.O.; Emeghebo, C.N.; Ilodigwe, E.E.; Ajaghaku, D.L.; Umeokoli, B.O.; Eze, P.M.; Ngwoke, K.G.; Okoye, F.B.C. Dryopteris filix-mas (L.) Schott ethanolic leaf extract and fractions exhibited profound anti-inflammatory activity. Avicenna J. Phytomed. 2019, 9, 396–409. [Google Scholar] [PubMed]
  91. Jiang, C.-L.; Tsai, S.-F.; Lee, S.-S. Flavonoids from Curcuma longa leaves and their NMR assignments. Nat. Prod. Commun. 2015, 10, 63–66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Taiwo, B.; Igbeneghu, O. Antioxidant and antibacterial activities of flavonoid glycosides from Ficus Exasperata Vahl-Holl (Moraceae) leaves. Afr. J. Tradit. Complement. Altern. Med. 2014, 11, 97. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Yesilada, E.; Gürbüz, İ.; Toker, G. Anti-ulcerogenic activity and isolation of the active principles from Sambucus ebulus L. leaves. J. Ethnopharmacol. 2014, 153, 478–483. [Google Scholar] [CrossRef] [PubMed]
  94. Nawwar, M.A.; Hashem, A.N.; Hussein, S.A.; Swilam, N.F.; Becker, A.; Haertel, B.; Lindequist, U.; El-Khatib, A.; Linscheid, M.W. Phenolic profiling of an extract from Eugenia jambos L. (Alston)—The structure of three flavonoid glycosides—Antioxidant and cytotoxic activities. Pharmazie 2016, 71, 162–168. [Google Scholar]
  95. Lim, S.S.; Jung, Y.J.; Hyun, S.K.; Lee, Y.S.; Choi, J.S. Rat lens aldose reductase inhibitory constituents of Nelumbo nucifera stamens. Phyther. Res. 2006, 20, 825–830. [Google Scholar] [CrossRef] [PubMed]
  96. Jung, H.A.; Park, J.C.; Chung, H.Y.; Kim, J.; Choi, J.S. Antioxidant flavonoids and chlorogenic acid from the leaves of Eriobotrya japonica. Arch. Pharm. Res. 1999, 22, 213–218. [Google Scholar] [CrossRef] [PubMed]
  97. Li, H.; Liu, Y.; Yi, Y.; Miao, Q.; Liu, S.; Zhao, F.; Cong, W.; Wang, C.; Xia, C. Purification of quercetin-3-O-sophoroside and isoquercitrin from Poacynum hendersonii leaves using macroporous resins followed by Sephadex LH-20 column chromatography. J. Chromatogr. B 2017, 1048, 56–63. [Google Scholar] [CrossRef] [PubMed]
  98. Lee, D.-Y.; Shrestha, S.; Seo, W.D.; Lee, M.-H.; Jeong, T.-S.; Cho, J.H.; Song, Y.C.; Kang, H.W.; Rho, Y.-D.; Baek, N.-I. Structural and quantitative analysis of antioxidant and low-density lipoprotein-antioxidant flavonoids from the grains of sugary rice. J. Med. Food 2012, 15, 399–405. [Google Scholar] [CrossRef]
  99. Azuma, C.M.; Santos, F.C.S.; Lago, J.H.G. Flavonoids and fatty acids of Camellia japonica leaves extract. Rev. Bras. Farmacogn. 2011, 21, 1159–1162. [Google Scholar] [CrossRef] [Green Version]
  100. 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. & C.A. mey. Iran. J. Pharm. Res. 2015, 14, 925–931. [Google Scholar]
  101. Lam, S.-H.; Wang, C.-Y.; Chen, C.-K.; Lee, S.-S. Chemical investigation of Phyllanthus reticulatus by HPLC-SPE-NMR and conventional methods. Phytochem. Anal. 2007, 18, 251–255. [Google Scholar] [CrossRef]
  102. Mottaghipisheh, J.; Mahmoodi-Sourestani, M.; Kiss, T.; Horváth, A.; Tóth, B.; Ayanmanesh, M.; Khamushi, A.; Csupor, D. Comprehensive chemotaxonomic analysis of saffron crocus tepal and stamen samples, as raw materials with potential antidepressant activity. J. Pharm. Biomed. Anal. 2020, 184, 113183. [Google Scholar] [CrossRef]
  103. Ashrafizadeh, M.; Tavakol, S.; Ahmadi, Z.; Roomiani, S.; Mohammadinejad, R.; Samarghandian, S. Therapeutic effects of kaempferol affecting autophagy and endoplasmic reticulum stress. Phyther. Res. 2020, 34, 911–923. [Google Scholar] [CrossRef]
  104. Rasouli, H.; Farzaei, M.H.; Khodarahmi, R. Polyphenols and their benefits: A review. Int. J. Food Prop. 2017, 1–42. [Google Scholar] [CrossRef] [Green Version]
  105. Yang, C.; Yang, W.; He, Z.; He, H.; Yang, X.; Lu, Y.; Li, H. Kaempferol improves lung ischemia-reperfusion injury via antiinflammation and antioxidative stress regulated by SIRT1/HMGB1/NF-κB Axis. Front. Pharmacol. 2020, 10, 1635. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Chen, A.Y.; Chen, Y.C. A review of the dietary flavonoid, kaempferol on human health and cancer chemoprevention. Food Chem. 2013, 138, 2099–2107. [Google Scholar] [CrossRef] [Green Version]
  107. Ren, J.; Lu, Y.; Qian, Y.; Chen, B.; Wu, T.; Ji, G. Recent progress regarding kaempferol for the treatment of various diseases (review). Exp. Ther. Med. 2019, 18, 2759–2776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Calderon-Montano, J.M.; Burgos-Moron, E.; Perez-Guerrero, C.; Lopez-Lazaro, M. A review on the dietary flavonoid kaempferol. Mini Rev. Med. Chem. 2011, 11, 298–344. [Google Scholar] [CrossRef]
  109. Imran, M.; Salehi, B.; Sharifi-Rad, J.; Aslam-Gondal, T.; Saeed, F.; Imran, A.; Shahbaz, M.; Tsouh-Fokou, P.V.; Umair-Arshad, M.; Khan, H.; et al. Kaempferol: A key emphasis to its anticancer potential. Molecules 2019, 24, 2277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Ouf, S.A.; Abdel-Hady, F.K.; El-Gamal, M.H.; Shaker, K.H. Isolation of antifungal compounds from some Zygophyllum species and their bioassay against two soil-borne plant pathogens. Folia Microbiol. 1994, 39, 215–221. [Google Scholar] [CrossRef]
  111. Erosa-Rejón, G.; Peña-Rodríguez, L.; Sterner, O. Isolation of kaempferol-3-rutinoside from the leaf extract of Sideroxylon foetidissimum subsp. Gaumeri. Rev. Latinoam. Química 2010, 38, 7–11. [Google Scholar]
  112. Ahmed, M.S.; El-Tanbouly, N.D.; Islam, W.T.; Sleem, A.A.; El-Senousy, A.S. Antiinflammatory flavonoids from Opuntia dillenii (Ker-Gawl) Haw. flowers growing in Egypt. Phyther. Res. 2005, 19, 807–809. [Google Scholar] [CrossRef]
  113. Kaouadji, M.; Morand, J.-M.; Garcia, J. Further acylated kaempferol rhamnosides from Platanus acerifolia buds. J. Nat. Prod. 1993, 56, 1618–1621. [Google Scholar] [CrossRef]
  114. Jung, H.A.; Kim, J.E.; Chung, H.Y.; Choi, J.S. Antioxidant principles of Nelumbo nucifera stamens. Arch. Pharm. Res. 2003, 26, 279–285. [Google Scholar] [CrossRef]
  115. al Musayeib, N.M.; Amina, M.; Mohamed, G.A.; Ibrahim, S.R.M. Plectranol A, a new sesquiterpene from Plectranthus cylindraceus growing in Saudi Arabia. Lett. Org. Chem. 2017, 14, 4–7. [Google Scholar] [CrossRef]
  116. Cho, J.-G.; Song, N.-Y.; Nam, T.-G.; Shrestha, S.; Park, H.-J.; Lyu, H.-N.; Kim, D.-O.; Lee, G.; Woo, Y.-M.; Jeong, T.-S.; et al. Flavonoids from the grains of C1/R-S transgenic rice, the transgenic Oryza sativa spp. japonica, and their radical scavenging activities. J. Agric. Food Chem. 2013, 61, 10354–10359. [Google Scholar] [PubMed]
  117. Hartati, S.; Angelina, M.; Meilawati, L.; Dewijanti, I.D. Isolation and characterization of compounds from the leaves of Pterocarpus indicus Willd and their antioxidant activity. Ann. Bogor. Trop. Gen. Bot. 2016, 20, 13–18. [Google Scholar]
  118. Leem, H.H.; Lee, G.Y.; Lee, J.S.; Lee, H.; Kim, J.H.; Kim, Y.H. Soluble epoxide hydrolase inhibitory activity of components from Leonurus japonicus. Int. J. Biol. Macromol. 2017, 103, 451–457. [Google Scholar] [CrossRef] [PubMed]
  119. Kim, J.; Yang, H.; Cho, N.; Kim, B.; Kim, Y.; Sung, S. Hepatoprotective constituents of Firmiana simplex stem bark against ethanol insult to primary rat hepatocytes. Pharmacogn. Mag. 2015, 11, 55. [Google Scholar]
  120. Jung, H.A.; Jin, S.E.; Park, J.-S.; Choi, J.S. Antidiabetic complications and anti-alzheimer activities of sophoflavescenol, a prenylated flavonol from Sophora flavescens, and its structure-activity relationship. Phyther. Res. 2011, 25, 709–715. [Google Scholar] [CrossRef]
  121. Li, L.; Yang, Y.; Hou, X.; Gu, D.; Ba, H.; Abdulla, R.; Wu, G.; Xin, X.; Aisa, H.A. Bioassay-guided separation and purification of water-soluble antioxidants from Carthamus tinctorius L. by combination of chromatographic techniques. Sep. Purif. Technol. 2013, 104, 200–207. [Google Scholar] [CrossRef]
  122. Castelli, M.V.; López, S.N. Homoisoflavonoids: Occurrence, biosynthesis, and biological activity. In Studies in Natural Products Chemistry; Elsevier: Amsterdam, The Netherlands, 2017; pp. 315–354. [Google Scholar]
  123. Abegaz, B.M.; Kinfe, H.H. Naturally occurring homoisoflavonoids: Phytochemistry, biological activities, and synthesis (Part II). Nat. Prod. Commun. 2019, 14. [Google Scholar] [CrossRef] [Green Version]
  124. Zhou, X.; Yuping, Z.; Zhao, H.; Liang, J.; Zhang, Y.; Shi, S. Antioxidant homoisoflavonoids from Polygonatum odoratum. Food Chem. 2015, 186, 63–68. [Google Scholar] [CrossRef]
  125. Rauf, A.; Imran, M.; Abu-Izneid, T.; Iahtisham-Ul, H.; Patel, S.; Pan, X.; Naz, S.; Sanches-Silva, A.; Saeed, F.; Rasul-Suleria, H.A.; et al. Proanthocyanidins: A comprehensive review. Biomed. Pharmacother. 2019, 116, 108999. [Google Scholar] [CrossRef]
  126. Cos, P.; Bruyne, T.; Hermans, N.; Apers, S.; Berghe, D.; Vlietinck, A. Proanthocyanidins in health care: Current and new trends. Curr. Med. Chem. 2004, 11, 1345–1359. [Google Scholar] [CrossRef] [PubMed]
  127. Huh, G.-W.; Park, J.-H.; Kang, J.-H.; Jeong, T.-S.; Kang, H.C.; Baek, N.-I. Flavonoids from Lindera glauca Blume as low-density lipoprotein oxidation inhibitors. Nat. Prod. Res. 2014, 28, 831–834. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The basic chemical structures of the flavonoid classes isolated or purified by applying Sephadex® LH-20.
Figure 1. The basic chemical structures of the flavonoid classes isolated or purified by applying Sephadex® LH-20.
Molecules 25 04146 g001
Figure 2. Number of flavonoid derivatives isolated or purified by Sephedex® LH-20.
Figure 2. Number of flavonoid derivatives isolated or purified by Sephedex® LH-20.
Molecules 25 04146 g002
Table 1. Abundant of flavonoids isolated/purified by using different eluents on Sephadex® LH-20.
Table 1. Abundant of flavonoids isolated/purified by using different eluents on Sephadex® LH-20.
Flavonoid ClassesEluent Systems
ABCDEFGHIJKLMNOPQ
Flavan & Isoflavan 2
Flavanone1 5 49
Flavanol & Isoflavanol 27
Flavone & Isoflavone1 1 89 12 282242
Flavonol 33 38 13726 2
Homoisoflavonoids 10
Proanthocyanidin 3
A: acetone; B: acetone-H2O; C: acetone-MeOH; D: acetone-H2O; E: acetonitrile‒MeOH; F: CH2Cl2; G: CH2Cl2–MeOH; H: CHCl3–MeOH; I: EtOAc; J: EtOAc‒MeOH; K: EtOH-H2O; L: H2O; M: MeOH; N: MeOH-H2O; O: n-hexane‒MeOH‒acetone; P: n-hexane-EtOAc; EtOAc; EtOAc–MeOH; Q: toluene‒EtOH.

Share and Cite

MDPI and ACS Style

Mottaghipisheh, J.; Iriti, M. Sephadex® LH-20, Isolation, and Purification of Flavonoids from Plant Species: A Comprehensive Review. Molecules 2020, 25, 4146. https://doi.org/10.3390/molecules25184146

AMA Style

Mottaghipisheh J, Iriti M. Sephadex® LH-20, Isolation, and Purification of Flavonoids from Plant Species: A Comprehensive Review. Molecules. 2020; 25(18):4146. https://doi.org/10.3390/molecules25184146

Chicago/Turabian Style

Mottaghipisheh, Javad, and Marcello Iriti. 2020. "Sephadex® LH-20, Isolation, and Purification of Flavonoids from Plant Species: A Comprehensive Review" Molecules 25, no. 18: 4146. https://doi.org/10.3390/molecules25184146

Article Metrics

Back to TopTop