Next Article in Journal
Studying the Suitability of Nineteen Lignins as Partial Polyol Replacement in Rigid Polyurethane/Polyisocyanurate Foam
Previous Article in Journal
Geochemical Characteristics and Significance of Organic Matter in Hydrate-Bearing Sediments from Shenhu Area, South China Sea
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Dioscorea spp.: Comprehensive Review of Antioxidant Properties and Their Relation to Phytochemicals and Health Benefits

by
Aušra Adomėnienė
and
Petras Rimantas Venskutonis
*
Department of Food Science and Technology, Kaunas University of Technology, Radvilėnų˛ pl. 19, LT-50254 Kaunas, Lithuania
*
Author to whom correspondence should be addressed.
Submission received: 1 February 2022 / Revised: 25 March 2022 / Accepted: 8 April 2022 / Published: 17 April 2022
(This article belongs to the Section Natural Products Chemistry)

Abstract

:
Dioscorea, consisting of over 600 species, is the most important genus in the Dioscoreaceae family; however, the practically used plants, which are commonly called yam, are restricted to a remarkably smaller number of species. Numerous studies have reported the high nutritional value of yam, particularly as an alternative source of starch and some important micronutrients. Several Dioscorea species are widely used for various medicinal purposes as well. In many studies, the bioactivities and health benefits of Dioscorea extracts and other preparations have been related to the presence of phytochemicals, which possess antioxidant properties; they are related mainly to radical-scavenging capacity in chemical assays and positive effects on the endogenous antioxidant system in cell-based and in vivo assays. Considering the increasing number of publications on this topic and the absence of comprehensive and focused review papers on antioxidant potential, this article summarizes the results of studies on the antioxidant properties of Dioscorea spp. and their relation to phytochemicals and health benefits. A comprehensive survey of the published articles has revealed that the majority of studies have been performed with plant tubers (rhizomes, roots), while reports on leaves are rather scarce. In general, leaf extracts demonstrated stronger antioxidant potential than tuber preparations. This may be related to the differences in phytochemical composition: saponins, phenanthrenes and, for some pigment-rich species (purple yams), anthocyanins are important constituents in tubers, while phenolic acids and flavonoids are characteristic phytochemicals in the leaves. The review may assist in explaining ethnopharmacological knowledge on the health benefits of Dioscorea plants and their preparations; moreover, it may foster further studies of poorly investigated species, as well as their wider application in developing new functional foods and nutraceuticals.

1. Introduction

The role of fruits, vegetables and botanicals, as natural and sustainable sources of valuable nutrients and health-beneficial substances, has been very important for humankind since ancient times. Due to the vast biodiversity in the plant kingdom and the large number of poorly investigated plants, the trend in the search, evaluation and application of natural preparations—as well as the development of new products—remains an important issue in various industries, including foods, pharmaceuticals and cosmetics. Moreover, the importance of this topic is increasing in the era of functional foods, nutraceuticals and personalized nutrition. In addition, the shift in consumer preferences towards ‘naturalness’ has also played an important role in the popularity of plant origin preparations.
Dioscoreaceae, with nine genera and about 715 species, is considered one of the earliest families of the plant kingdom (Magnoliophyta) [1]. In terms of uses, Dioscorea, consisting of over 600 species, is the most important genus in this family. The plants are perennial herbaceous and dicotyledonous, and bloom with faint flowers and have large roots and/or rhizomes. Dioscorea plants are common in tropical and subtropical regions; however, many of them have successfully adapted to different habitats and, nowadays, are common in other climatic regions as well. Regardless, a large number of existing species—practically used plants, which are commonly called yam—are restricted to a smaller number of species; among them are D. batatas, D. japonica, D. bulbifera, D. opposita, D. tokoro, D. nipponica and D. alata [2]. For instance, D. bulbifera L. (air potato) is native in Asia, North Florida [3], and Georgia (www.gaeppc.org/list/ [4], accessed on 11 November 2021); D. alata L. (water yam, winged/white yam) originates from Southeast Asia [5] but is also found in Africa (FAOSTAT, Database 2021 [6], accessed on 11 November 2021), Georgia (www.gaeppc.org/list/ [4], accessed on 11 November 2021), and America (www.eddmaps.org [7], accessed on 12 November 2021); D. oppositifolia, syn. D. opposita, D. batata, D. polystachya Turcz. (Chinese yam), is native to Asia and is also found in eastern North America.
It should be noted that the occurrence of invasive plant species, albeit beneficial ones, poses a threat to native species and habitats. For example, D. oppositifolia (syn. D. polystachya) is recognized in North America (www.iucngish.org/gisd/, accessed on 22 November 2021, Global Invasive Species Database, 2021 [8]) as a forest or agricultural weed, which can compete with the native species, especially in coastal habitats; while widespread in Africa, D. alata is one of the most valued foods. In general, due to the nutritional and therapeutic benefits and economic income, the contribution of yam to food security is significant in some regions, especially in sub-Saharan Africa [9]. Dioscorea spp. are also known for its association with low-cost food culture, traditional Chinese medicine, modern western medicine, and the pharmaceutical industry. The peculiarities of the diet and traditions of this genus of plant species in many cultures are closely related to the historical and religious aspects of life (FAOSTAT, Database, 2021 [6]), which began much earlier than scientific research. It should be noted that different yam species accumulate a large variety of biologically active compounds and, therefore, various anatomical parts of the plants have been used for different purposes. For instance, D alata, D. japonica Thunb., and the rhizomes of D. schimperiana [10,11,12] are valued as a source of starch in the development of flour, pasta, and desserts.
Traditional medicine mentions such species as D. bulbifera in the treatment of goiter, skin infections, and oncological diseases [13]; D. nipponica in the treatment of arthritis, cough, asthma, and circulatory disorders [14]; D. villosa (wild yam) in the reduction of intestinal disorders [15]; and D. birmanica in the treatment of chronic diseases, whose development is associated with long-term oxidative stress [16]. Food supplements used in modern medicine as natural alternatives to hormone replacement therapy, in the form of powders, capsules, and herbal extracts, are made from the roots of D. villosa; a traditional herbal preparation made from D. nipponica root is used to regulate the immune system and reduce pain and inflammation [14], while the saponins extracted from D. septembola are used in China to treat gout [17].
In addition to the well-known species with high nutritional, economic and medicinal value, it is worth mentioning the other less-known but valuable species that can enrich the diet and improve health, namely: D. birmanica Prain and Burkill [16]; D. hamiltonii Hook. f syn. D. persimilis [18,19]; D. hispida [20]; D. pubera Blume; D. wallichii Hook [21], and others. Some species are poisonous but have healing properties, e.g., D. hispida, which is a promising alternative to conventional chemotherapy [22,23].
Secondary metabolites identified in Dioscorea spp. are often used in physiological and molecular studies. The well-known natural steroid saponin diosgenin has been used in the pharmaceutical industry as a chemical model in the development and/or complete synthesis of hormonal drugs. This metabolite is also reported to reduce oxidative stress damage, may protect the myocardium from ischemia-induced injury [24], and modulates the intestinal microbiota [24,25]. Dioscin and gracillin are also very important metabolites, demonstrating anticancer [26,27] and antioxidant effects. Trillin, found in D. nipponica [28], D. oppositifolia, and D. hamiltonii [29], is a pharmacologically viable steroid saponin with a broad spectrum of physiological effects in the cells, e.g., increasing superoxide dismutase (SOD) activity, and lowering lipid peroxidation activity and the oxidative stress response [28].
From nutritional and economic points of view, carbohydrates are likely the most important constituents of yam tubers, which are used in the development of foods as bulk ingredients; however, their content in the leaves is rather small. On the other hand, the leaves, as a rule, accumulate larger amounts of bioactive phytochemicals, e.g., polyphenolic antioxidants, including phenolic acids (chlorogenic, syringic, vanillic, p-hydroxybenzoic, p-coumaric), flavonoids (quercetin, rutin, kaempferol, catechin, anthocyanins), etc. Aromatic phenanthrene derivatives, demonstrating antifungal [30] and antioxidant effects [31,32], have also been reported in different species, e.g., D. batatas [30,32], D. rotundata [30], and D. communis [31].
As may be judged from the records present in various databases (Table 1), Dioscorea plants have been widely studied, and the published results have been reviewed in numerous articles.
The topic of Dioscorea antioxidants has also been the focus of many studies; however, the reviews on this topic are less abundant and fragmental.
Obidiegwu et al. [9] published the most comprehensive review on the nutritional and therapeutic potential of the Dioscorea genus. This review covers the nutritional value (starch, fiber, protein, fat, minerals), the profile of bioactive compounds, the therapeutic potential and health benefits (anti-inflammatory, anticancer, anti-diabetic, anti-obesity and anti-hypercholesterolaemic and antimicrobial activities), the management of degenerative disease, menopausal symptoms, and its use as a pharmaceutical excipient. Regarding bioactive compounds, the review includes separate sections on steroidal saponin, dioscorin, alkaloids, flavonoids, phenolic acids and others, while the antioxidant properties of yam are discussed in a very short section. In addition, it refers to seven publications from 2019, while in the period of 2019–2021, fifty-four new articles related to Dioscorea antioxidants have been published.
Padhan and Panda [33], in their review on underutilized and neglected yams, emphasized their potential for improving nutritional security and providing health benefits. The authors focused their article on the ethnobotany of Dioscorea spp., which are consumed by tribal people, and briefly highlighted their nutritional, anti-nutritional, and pharmacological properties. Kumar et al. [34] reviewed the traditional uses and ethnopharmacological potential of wild edible Dioscorea tubers by the local people of the Similipal Biosphere Reserve in India, whereas George et al. [35] highlighted the importance of African yam bean. Salehi et al. [36] reviewed Dioscorea plants as rich in nutrients and pharmacologically valuable constituents; however, their antioxidant activities were summarized in a very short section.
Some reviews were focused on individual Dioscorea species and important groups of nutrients and bioactives. For instance, Bhujbal et al. [37] reviewed the pharmacological activity of D. floribunda, and Semwal et al. [38] focused on the traditional uses, bioactive compounds and biological activities of D. deltoidea Wall. ex Griseb. Huang et al. [39] focused their paper on the isolation, structure and bioactivities of yam mucilage polysaccharides and included a short section on antioxidant properties, while Anwar et al. [40] included yam tubers in their review on water-soluble non-starch polysaccharides of root and tuber crops. Zhang et al. [41] focused their review on the development of proteins and peptides from Dioscorea tubers with therapeutic potential. Petropoulos et al. [42] reviewed the antioxidant properties and health benefits of colored root vegetables, which included a section on yam pigments as well. Finally, Yang et al. [43] reviewed the pharmacological activities of dioscin, and Toth et al. [44] reviewed those of phenanthrenes.
The survey of published review articles reveals that a comprehensive review on Dioscorea antioxidant properties has not been published until now, while the pool of data on this topic is quite large. Therefore, the main task of this review was to collect the available results on Dioscorea antioxidant potential, the presence of the main chemical groups of antioxidants, and their relations to health benefits. It should be noted that ethnopharmacological properties and medicinal uses are not covered in this review unless they are closely related to the antioxidant activities.

2. In Vitro Antioxidant Potential and Total Amounts of Phytochemicals

2.1. Evaluation of Crude Extracts and Their Fractions

The antioxidant properties of Dioscorea spp. have been studied using various in vitro chemical methods, cell cultures and in vivo assays, mainly using rat and mouse models. The results of the in vitro chemical assays are summarized in Table 2.
It can be observed that antioxidant properties have been determined using several chemical assays and expressed in various units, e.g., the equivalent amount of the reference antioxidant (trolox, ascorbic acid, catechin, α-tocopherol); the fresh (pfw) and dry weight (pdw) of plant material and/or extract (edw); the effective inhibitory concentration (IC50, in some cases abbreviated as EC50) values of the extract or its solution; or the percentage inhibition at the applied extract concentrations. In the latter case, the results are very difficult to compare because it is necessary to know the procedure of the sample preparation for the assays, particularly their concentrations in the solutions. Therefore, if the reader would like to know more details, it is suggested to download the full article. The values expressed in the reference antioxidant equivalents in pdw/pfw and/or edw are the most convenient; the former indicates the antioxidant potential of the whole plant material used in the assay, while the latter is directly related to the antioxidant properties of the isolated extracts and fractions.
This section also includes the results reported for the total phenolic content (TPC), total flavonoid content (TFC), total monomeric anthocyanins (TAC) and total saponins (TS) (Table 3). These values are determined mainly using spectrophotometric methods, and—depending on the plant material (species, cultivar), its composition, processing and extractions methods—may be rather indicative. It can be clearly observed from the recently reported values for MeOH extracts of D. bulbifera stem tubers (Table 3) that TPC and TFC values were determined to be remarkably lower than the total flavonols, which belong to one of the flavonoid subclasses [92]. Adeniran and Sonibare [67] reported somewhat similar findings; TFC values were remarkably higher than TPC ones in the four analyzed Dioscorea samples. TPC measurement with the Folin–Ciocalteu reagent is based on a single-electron-transfer reaction, which can proceed within the reaction system and present various nonphenolic compounds as well. In addition, the total content values in many publications are presented together with radical scavenging and other antioxidant capacity assays; therefore, it is reasonable to review all these values in one chapter. Nevertheless, the values of total phytochemical content are summarized separately in Table 3.
Several studies compared the antioxidant potential of different Dioscorea spp. and cultivars of the same species and demonstrated that the differences may be quite remarkable. For instance, Padhan et al. [101] reported the flavonoid, total antioxidant capacity and in vitro antioxidant activity of eight wild (D. oppositifolia, D. hamiltonii, D. bulbifera, D. pubera, D. pentaphylla, D. wallichii, D. glabra, D. hispida) and one cultivated (D. alata) yam tuber from Koraput (India): TPC, TFC, and total antioxidant capacity ranged from 2.19 to 9.62 mg GAE/g pdw, 0.62–0.85 mg QE/g pdw and 1.63–5.59%, respectively; meanwhile, the IC50 values were 77.9–1164, 101.2–1031.6, 27.0–1022.6 and 47.1–690 µg/mL for DPPH, ABTS•+, O2, and NO scavenging capacity, respectively. The TPC and TFC values of D. deltoidea, D. prazeri, D. bulbifera, D. pentaphylla, D. esculenta, and six cultivars of D. alata were in the ranges of 115.31–1628.50 mg/100 g pdw and 17.57–148.46 mg/100 g pdw, with D. prazeri being the richest source of polyphenolics [102]. Bhandari and Kawabata [96] reported that the TPC of D. bulbifera, D. versicolor, D. deltoidei, and D. triphylla from Nepal ranged from 13 ± 1 to 166 ± 10 mg/100 g fpw, while the differences in antioxidant characteristics were less remarkable, particularly in chelating ferrous ion, reducing power, and total antioxidant capacity. Jimenez-Montero and Silvera [103] compared six varieties of D. bulbifera (air potato) from Panama and found that the variations in ascorbic acid, antioxidant capacity, and TPC were not remarkable (6.0–8.1 mg/100 g, IC50 7.85–8.87, and 175–201 mg GAE/100 g, respectively). However, the cultivars of the same species may differ significantly. For instance, the TPC, TFC, and TAC values of the flour of the Kulonprogo cultivar of D. alata, determined by extraction with acid and with 5% acetic acid water, were higher than those of the Malang cultivar [49]. Alcohol and ester extracts of yellow fresh-cut yam (D. opposita) exhibited higher ORAC and DPPH scavenging capacity than white fresh-cut yam [69].
The majority of studies used yam; however, some of them were performed with different anatomical parts of Dioscorea as well, including the flesh, peels and leaves. Thus, Alsawalha et al. [104] reported that MeOH extract from D. villosa leaf powder demonstrated strong antioxidant capacity in a FRAP and DPPH scavenging assay in a dose-dependent manner; at a concentration of 0.1 mg/mL, the inhibition of DPPH was >90%, while the optical density in the FRAP assay was 1.3 (2 for ascorbic acid). Mondal et al. [105] compared MeOH and EA extracts of D. pentaphylla leaves and found that TPC, DPPH and NO scavenging was higher for the former, while the latter had almost 4-fold higher TFC and TAC.
Yams contain various nutrients and phytochemicals, which differ in their chemical structures, molecular weight, polarity and other characteristics. Some studies fractionated yam materials and extracts using different polarity solvents. Duan et al. [60] reported only slight differences between n-BuOH and EA extracts of D. batatas, which were evaluated using different in vitro antioxidant assays. In the case of a thermally treated product, EA extract was more effective in chelating ferrous ion and scavenging NO, while n-BuOH extract better inhibited linoleic acid peroxidation [59]. Duan et al. [56] compared yam (D. batatas) extracts isolated with 70% MeOH, 70% EtOH and CF–MeOH (2:1, v/v). MeOH extract was most effective at chelating ferrous ion, NO scavenging and the β-carotene bleaching assay, while CF–MeOH extract better inhibited linoleic acid peroxidation. Ghosh et al. [64] extracted the bulbs of D. bulbifera with cold 70% EtOH, and sequentially with PE, EA and MeOH; the latter extract contained higher TPC, while the EA fraction was richer in TFC (Table 3). The antioxidant properties of EA and alcoholic extracts were stronger compared with the PE extract. Park et al. [89] extracted bulbils of Danma (D. japonica) and Jangma (D. batatas) with MeOH, and the crude extracts were re-extracted with CF, EA, n-BuOH, and water. Xia et al. [106] compared antioxidant properties of PE, EA and n-BuOH fractions of D. nipponica, which were obtained from a dried 60% EtOH extract. The fractions inhibited DPPH, O2, and OH in a concentration range of 2.5–12.5 mg/mL; however, the dependence between radical inhibition percentage and extract concentration was far from linear. Adeniran et al. [107] successively extracted the peel and flesh of D. dumetorum, D. hirtiflora, and D. bulbifera with the increasing polarity solvents HX, DCM, EA, and MeOH. The MeOH extract of D. hirtiflora flesh showed the highest radical-scavenging capacity, while TPC and TFC were higher in the EA extract of D. bulbifera flesh. Among D. quinqueloba extracts isolated with water, MeOH, EtOH, and EA, the EtOH extract was the strongest DPPH scavenger, whereas EA extract better scavenged ABTS•+; both values increased with the addition of water-soluble chitosan [97]. Chen et al. [53] fractionated water solution of dried crude EtOH extract from Taiwan yams (D. alata) via sequential extraction with HX, CF, EA, and n-BuOH. Based on the IC50 values, the peel extracts better scavenged DPPH than the flesh products. The EA fraction of the peel of Tainung yam was a particularly strong antioxidant; its EC50 value was only 14.5 µg/mL, which was lower than that of ascorbic acid (21.4 µg/mL). In addition, the authors applied pressurized solvents and revealed that hot pressurized EtOH was superior to hot pressurized water in extracting the DPPH scavenging compounds [53].
Only one study was found on the use of biotechnological methods for obtaining antioxidants from yams. Jirakiattikul et al. [108] applied in vitro propagation of D. birmanica shoots; however, the TPC and DPPH scavenging values were remarkably higher in the mother rhizomes than in the in vitro produced shoots (259.67 ± 7.34 vs. 44.24 ± 8.47 mg GAE/g and 11.42 ± 3.28 vs. 53.67 ± 4.16 EC50 µg/mL dry extract, respectively).

2.2. Evaluation of Polysaccharides and Some Other Compounds

Several studies reported antioxidant properties in yam polysaccharides. The pre-purified (dialyzed) polysaccharide of air yam (D. bulbifera) was approx. a 4-fold stronger antioxidant in ABTS•+, according to a decolorization assay, than the crude (non-dialyzed) polysaccharide; in contrast, in DPPH and FRAP assays, the latter showed a several-times stronger antioxidant capacity [65]. It can be noted that the reported results are rather strange, because the mechanisms of all the radical-scavenging reactions were based on electron transfer (hydrogen atom transfer may proceed with DPPH as well). Most likely, extended studies using more specific assays would be required to more precisely clarify the mechanisms of action of yam polysaccharides in the redox systems. Zhang et al. [105] precipitated yam polysaccharide from the centrifugate of D. opposita homogenate with 75% EtOH, and afterwards, degraded it into three low MW derivatives; these were strong antioxidants against OH (IC50 = 187, 82 and 55 µg/mL) and O2 (IC50 was 241, 138 and 125 µg/mL), while the activity of raw polysaccharide was lower. The polysaccharide partially prevented the cyclophosphamide-induced damage, and its effect was probably due to high anti-lipid peroxidation activity and radical-scavenging effects. A previously unknown water-soluble polysaccharide was isolated from the roots of a well-known edible and medicinal plant in China, D. opposite. Its antioxidant capacity IC50 values against DPPH and PTIO were 2.1 ± 0.1 and 1.6 ± 0.1 mg/mL, respectively [109]. Zhao et al. [72] compared ultrasound-assisted, cold, warm, and hot water extraction for the isolation of polysaccharides, and determined that cold water gave the highest uronic acid yield; its DPPH scavenging, α-glucosidase and α-amylase inhibition activities were the best. The MW of polysaccharides recovered using ultrasound-assisted extraction was small, while its reducing power and FRAP were the best. The extracts of tuber pulp from the three D. alata taxa had significant anti-Fenton reaction activity, which was related to the polysaccharide mucilage [110]. Most recently, the antioxidant activity of mucilage isolated from D. opposita was evaluated using DPPH-scavenging and FRAP assays; at a 5.0 mg/mL concentration, it inhibited 68.57% of radicals, while FRAP was remarkably lower compared to ascorbic acid [111]. The OH-scavenging capacity of diosgenin obtained from D. composita by direct hydrolysis-extraction, under biphasic reaction conditions (1.5% yield), was higher than that of ascorbic acid when the concentration of the scavenger was higher than 2.5 × 10−3 mmol/L [112]. Various solvents have been used for the extraction and fractionation of antioxidants from Dioscorea. Kwon et al. [45] determined antioxidant, antithrombin, and antimicrobial activities of MeOH extracts prepared from two D. batatas cultivars, two D. alata, one D. bulbifera, and one D. nipponica. Ramos-Escudero et al. [113] reported the TAC, TPC, and antioxidant activity of the pigments extracted from D. trifida.

2.3. Evaluation of Proteins and Their Hydrolysis Products

Several studies reported antioxidant and other bioactivities of yam mucilage, proteins and their hydrolysates. Hou et al. [114] purified the storage protein of yam (D. batatas) tuber dioscorin (32 kDa), which scavenged DPPH (8–46%) at 5.97–47.80 nmol; dioscorin decreased the intensities of the electron spin resonance (EPR) signals used for DPPH and OH detection in a dose-dependent manner. Hou et al. [115] extracted and partially purified mucilage from a D. batatas tuber: its IC50 in DPPH, OH and anti-lipid peroxidation and anti-human low-density lipoprotein peroxidation tests were 0.86 mg/mL, 22 µg/mL and 145.46 µg/mL, respectively, using BHT, GSH, or ascorbic acid for comparisons. The intensities of the EPR signals decreased with increasing yam mucilage amounts (IC50 was 1.62 mg/mL). The mucilage also inhibited the angiotensin-converting enzyme (ACE) [116]. Nagai and co-authors [81,85,117,118,119,120] studied proteins isolated from D. opposita tuber mucilage tororo. Firstly, they obtained viscous water extract containing 280 mg/mL of protein, which demonstrated the main bands determined by SDS-PAGE at MW ~31 kDa (reducing conditions) or ~33 kDa (non-reducing conditions), and scavenged O2 (84.1 ± 6.57%), OH (79.4 ± 6.42%), and DPPH (38.2 ± 3.14%) [61]. Afterwards they treated the extract with protease or mannanase and noted that the viscosity highly depends on the interactions between certain soluble proteins with mannan [121]. They purified a soluble viscous protein, dioscorin (~200 kDa), from the mucilage using a chromatographic procedure [111] and measured its antioxidant properties. The protein effectively scavenged OH (IC50 = 195.1 μg/mL) and O2 (IC50 = 92.7 μg/mL) [120]. Autolysate and trypsin hydrolysate from yam (D. opposita) ichyoimo tubers exhibited extremely high antioxidant and ACE-inhibitory activities [119]. The lyophilized powder of enzymatic hydrolysates from mucilage tororo effectively scavenged O2 and OH, particularly when trypsin was used for hydrolysis [118]. The hydrolysates of a yam tsukuneimo tuber mucilage tororo at a 100 mg/mL concentration significantly prolonged the induction period of linoleic acid auto-oxidation (similar to 5 mM ascorbic acid), and scavenged O2, OH, and DPPH [85]. Liu et al. [122] also purified dioscorin from D. alata and D. batatas using DE-52 ion-exchange chromatography and reported antioxidant properties in the protein and its hydrolysates using various assays. Oh and Lim [123] isolated glycoprotein from D. batatas, which has been traditionally used as a folk medicine and health food in Korea; it had an optimal radical-scavenging capacity at an acidic and neutral pH and at temperatures up to 85 °C, while in the presence of Ca2+, Mn2+, Mg2+, and EDTA, its activity reduced.

2.4. Effects of Processing and Various Treatments

Processing techniques such as blanching, thermal treatment, soaking, oven-drying, and freeze-drying had different effects on the content of phenolics, DPPH scavenging and polyphenol oxidase activity in D. esculenta and D. bulbifera tuber flour [124]. Chung et al. [88] reported the effects of blanching, drying and extraction with 50% EtOH, hot water, and water on the antioxidant activities of Taiwanese yam (D. alata). The peel exhibited much higher antioxidant activities. Blanching by immersing the peel in 85 °C water for 30 s caused significant reduction in the antioxidant activities of all peel extracts. Generally speaking, freeze-dried peel maintained higher antioxidant activities than hot air-dried peel. More recently, Santos et al. [90] reported that the freeze-dried purple yam (D. trifida) powder had higher antioxidant capacity and higher TPC and TAC values, compared with the products dried using a refractance window at 70, 80, and 90 °C for 40 min. Compared with the native starch, the antioxidant activity of the starch–palmitic acid complex, prepared using the reaction of different concentrations (0.1–5.0%) of palmitic acid and yam (D. opposita) starch, significantly increased [125]. D. bulbifera tuber extract was used in the synthesis of novel platinum–palladium bimetallic nanoparticles along with individual platinum and palladium nanoparticles. Bimetallic nanoparticles demonstrated enhanced DPPH, O2, NO, and OH scavenging capacity and more pronounced effects on the HeLa cancer cell death [126]. There was no particle size influence on the content of phenolics (0.27–2.82% dw) or anthocyanin (2.25–15.27 mg/100 g dw) in light purple, purple and dark purple cultivars, or carotene (23.75–132.12 mg/100 g dw) in yellow and orange cultivars of D. alata [99]. Adedayo et al. [47] reported the effects of processing on the TPC, TFC and antioxidant characteristics of white (D. rotundata) and water yam (D. alata) flour (Table 3). Jaleel et al. [127] determined that paclobutrazol (PBZ) treatments had a profound effect on antioxidant metabolism and caused an enhancement in both nonenzymatic and enzymatic antioxidant potentials under treatments in white yam (D. rotundata). Djeukeu et al. [11] used 10–60% of the dried yam (D. schimperiana) flour produced by the traditional and modified process (boiled for polyphenol oxidase inhibition before drying) methods in pasta; a yam ingredient increased TPC, ABTS•+ scavenging, FRAP, and RSA values up to 2.9, 3.80, 14.48, and 2.21-fold; conversely in the case of the modified process, all the values were higher than for the traditional method. The content of total soluble phenolics in the roots of minimally processed yam (commercial Dioscorea spp.) did not change significantly during 14 days of storage at 5 °C (16.9–17.8 mg/g FW) or at 10 °C (16.7–17.5 mg/g FW) [128]. Lu et al. [98] reported an increase in the content of polyphenols, flavonoids, and anthocyanins in the tuber flesh and peel of five commercial yams using arbuscular mycorrhizal fungi treatment. Ratnaningsih et al. [49] compared immersion in water, Na-bisulfite, and ascorbic acid as browning inhibitors of D. alata flour, and found that the treatment provided better TAC, TPC and TFC values.

2.5. Application of Dioscorea spp. Ingredients for Increasing Product Antioxidant Properties

Yam flour has been used in the formulations of various foods. Adeloye et al. [10] reported that the addition of water yam (D. alata) flour in the formula of dough at 10–40%, together with plantain and bitter leaves, increased the TPC from 1.26 to 4.44 mg GAE/g, the radical-scavenging capacity from 14.73 to 48.66 mg/g, and the FRAP from 8.57 to 42.52 mg AAE/g, in a dose-dependent manner. The substitution of up to 40% of wheat flour in bread formulation with purple yam (D. alata) flour, containing 220 mg GAE of TPC, 640 mg AAE DPPH scavenging capacity, and 16 mg CGE of TAC in 100 g, showed liking scores in all bread sensory attributes similar to those of the control [129]. The addition of 2–8% D. japonica powder—containing 11.16% protein, 0.73% fat, and 76.39% carbohydrate—into red bean sweet jelly desert ‘Yangaeng’ increased the TPC from 14.58 to 71.54 mg GAE/100 g, and DPPH scavenging from 5.84 to 63.98%, in a dose-dependent manner [12]. Extrusion processing increased the TBARS inhibition ability for all yams (D. alata var. Tai-nun and var. Ta-shan, D. doryophora var. Hang-chun and commercial yam flour) and generally had no negative impact on DPPH, ABTS•+ scavenging, ORAC, and Fe2+ chelating for most yams in corn–yam extrudates [51]. Extruded yam flour (D. alata) containing 61.20 ± 0.02 mg GAE/g phenolics was used in the formulation of instant drinks [130]. Yams (D. alata) added at 5%, 10%, and 15% to Chinese sausages caused lower thiobarbituric acid (TBA) values during 21 days of storage [131]. Li et al. [132] tested up to 25% of Chinese yam (D. opposita) flour in bread formula. The content of flavonoids increased in the bread from 0.162 to 0.516 mg RE/g, and allantoin from 0.05 to 1.063 mg/g, while the IC50 value of antioxidant capacity decreased from 124.14 to 101.36 mg/mL. On the other hand, it should be noted that the TPC values increased less significantly, from 0.467 to 0.491 mg GAE/g, which is in some degree of disagreement with the total flavonoids, which belong to the group of phenolics.

3. In Vitro Assays in Cell Cultures and In Vivo Assays with Animals

Quite a few studies tested Dioscorea preparations in cell cultures and in vivo assays with animals, mainly rats and mice. The results of these studies are summarized in Table 4.
The effects of yam extracts, purified fractions, and individual compounds on various biomarkers were reported, including important factors of endogenous antioxidant defense in the cells, namely superoxide dismutase (SOD), glutathione peroxidase (GPx), catalase (CAT) and reduced glutathione (GSH). This table also includes brief information on chemical assays of enzyme inhibition, which are related to various physiological effects. It should be noted that many authors related the beneficial effects of Dioscorea preparations in cell and animal assays with their antioxidant properties—which were determined using the chemical assays (Table 2)—and the presence of various classes of phytochemicals (Table 3).
The extracts isolated with different polarity solvents were tested in various cell assays. EtOH extraction of dried, ground D. birmanica (locally called Hua-Khao Yen-Neua) rhizomes yielded 10.56% of the extract, with TPC and TFC values of 170.85 ± 3.02 mg GAE/g and 132.55 ± 3.59 mg CE/g, respectively [16]. The authors suggested that the identified phytochemicals from the extract, which scavenged radicals and moderately chelated metals, may be responsible for various bioactivities in the cells. Water fraction isolated by reflux-extraction of dried D. cirrhosa powder, and afterwards separated by AB-8 macroporous resin with water, 60% and 95% EtOH demonstrated numerous bioactivities in cells as well [148]. The extracts of aerial bulblets of D. japonica inhibited the induction of inflammatory mediators and the expression of iNOS and COX-2 in the cells [149].
Among the five products separated from the purple Chinese yam (D. alata) extracts, the EA fraction of flesh and peel exerted the best antioxidant activity and exhibited excellent antiglycation ability by suppressing different stages of the glycation cascade; they also exhibited cytoprotective effects on methylglyoxal-induced oxidative damage in HepG2 cells [52]. Steroidal glycosides, spirostans, of D. villosa (zingiberensis saponin I, dioscin, deltonin, and progenin III) were found to be cytotoxic, whereas, furostans (huangjiangsu A, pseudoprotodioscin, methyl protobioside, protodioscin, and protodeltonin) were non-cytotoxic. While the treatment of HepG2 cells with compounds prior to H2O2 exposure effectively increased cell viability in a concentration-dependent manner, some of them increased GSH level and decreased intracellular ROS generation [150]. 2,7-Dihydroxy-4,6-dimethoxy phenanthrene, which was isolated from yam (D. batatas) peel extract with 95% EtOH, was found to be a strong antioxidant and anti-inflammatory agent in bio-guided fractionation assays with HepG2-ARE cells [136]. MeOH extract of D. bulbifera leaves had significantly higher antioxidant activity than EA and HX extracts in DPPH, FRAP, and ABTS•+ assays, and demonstrated a pronounced cytotoxic effect in MDA-MB-231 and MCF-7 cell lines [63]. Ren et al. [151] isolated two new norsesquiterpenoids, dioscopposin A and dioscopposin B, as well as 21 known compounds from the stems and leaves of D. oppositifolia, and evaluated their estrogenic activity using MCF-7 cells; eight of these compounds inhibited cell proliferation. Antioxidant capacity was correlated with phenolic content in the cytoprotective effect on HUVEC viability assay, and D. glabra extract was more active than D. alata against H2O2-induced oxidative stress [152].
The in vivo studies were focused on the various physiological effects, and some of them reported beneficial effects of Dioscorea preparations in diabetic animals. Potential effects of ethanol extract isolated from a traditional Chinese medicine, D. zingiberensis, against testicular tissue damage via activation of the nuclear factor erythroid 2 (Nrf2) were demonstrated in a diabetic mouse model [153]. A polysaccharide, which was isolated from D. opposita roots with acidified water-extraction following precipitation with EtOH and chromatographic purification, increased SOD activity in alloxan-induced diabetic rats and mouse models at higher doses, and had strong hypoglycemic activity [154]. Fermentable water-soluble polysaccharides of purple and yellow water yam (D. alata) produced short-chain fatty acids, and also demonstrated hypoglycemic effects in rats with alloxan-induced glucose levels [134]. Treatment with crude yam (D. batatas) powder, water extract and allantoin effectively ameliorated antioxidant stress in streptozotocin (STZ)-induced diabetic rats by decreasing MDA formation and increasing SOD/GPx activities [140]. The administration of yam (D. batatas) rhizome aqueous extract and allantoin at high doses had numerous health benefits in the in vivo studies with high-fat-diet (HFD)/STZ-induced diabetic mice [137]. It is worth mentioning that the traditional Chinese medicine Rehmannia Six Formula for treating diabetes mellitus includes D. opposita, D. alata, and D. batatas [155].
Many in vivo studies reported antioxidant effects and their beneficial contribution to regulating inflammatory events and protecting against damage to important biomolecules through excessive formation of ROS. Several studies reported antioxidant defense-related benefits for gastrointestinal health. Ethanol extracts of D. batatas peel had various effects on human colonic epithelial HCT116 cells, including a reduction in the level of ROS in H2O2-challenged cells. Oral supplementation inhibited colon inflammation through the upregulation of antioxidant enzymes and subsequent downregulation of inflammatory proteins [156]. D. batatas flesh and peel extracts, obtained with water or 60% or 95% EtOH, were tested in vivo; pre-treatment with 60% EtOH flesh extract before ulcer induction significantly decreased the expression of biomarkers of oxidative stress [138]. Park et al. [18] also reported positive effects of crude mucin and saponins from the Dioscorea rhizome on rats induced with gastric ulcers using alcohol [18]. In the studies with gastric ulcer rats, the administration of crude Dioscorea rhizoma extract activated the antioxidant enzyme CAT and increased the levels of SOD and GPX, while the production of MDA, congestion, and hemorrhage of tissue decreased [157]. An anthocyanin-rich fraction with five identified compounds isolated from D. alata produced potent anti-inflammatory effects in the mouse model of inflammatory bowel disease (IBD) at 80 mg/kg bw; anthocyanins may reduce intestinal permeability, in part, by preserving the ultrastructural integrity of the epithelial mucosa [17]. Yam (D. batatas) increased erythrocyte levels of glutathione, GPx, and CAT in azoxymethane (AOM)-induced colon carcinogenesis in male F344 rats; in addition, the colonic mucosal gene expression of SOD and GPx were up-regulated, and the expression of inflammatory mediators was suppressed [158].
The excessive formation of ROS is also related with the initial phases of carcinogenesis. Yellow pigment from freshly cut yam (D. opposita) exhibited a protective effect against OH-induced DNA damage, which was attributed to the high OH-scavenging activity exerted by the pigment [70]. In addition, Liu et al. [75] determined that all the peel extracts of D. opposita had a better effect in the ROS scavenging assay and a stronger antitumor property in the mouse models than in flesh. D. bulbifera polysaccharides attenuated cyclophosphamide-heightened oxidative stress in mice; therefore, combination use can potentially enhance the cardiotoxin (CTX) anti-tumor effect and can attenuate CTX-induced immunosuppression and oxidative stress in U14 cervical-tumor-bearing mice [159].
Several studies focused on the evaluation of Dioscorea preparations as cardioprotective agents. Thus, total saponins from D. nipponica, D. panthaica, and D. zingiberensis lowered MDA levels and increased SOD, CAT, GPx, and T-AOC activities in rats; moreover, they demonstrated cardioprotective effects, as was determined by the less severe histological damage [146]. Isoproterenol (ISO)-induced rats pretreated with a flavonoid-rich fraction of D. bulbifera (150 mg/kg) exhibited ameliorated lipid peroxidation and, thereby, enhanced antioxidant status, as evidenced by the increase in the GSH content and the activity of antioxidant enzymes, which also suggest a cardioprotective effect [160]. Feng et al. [24] demonstrated that diosgenins (L, M, H) were the main metabolites produced by rat intestinal microflora from D. nipponica; moreover, when administered orally at up to 80 mg/kg, they protected the myocardium against ischemic insult through increasing enzymatic and nonenzymatic antioxidant (SOD, CAT, GPx, T-AOC) levels in vivo, and by decreasing oxidative stress damage. The intra-peritoneal administration of a steroidal saponin isolated from the rhizomes of D. nipponica, trillin, in rats exerted beneficial effects in improving the levels of lipid peroxidation and SOD activity [28].
Various other health benefits of yam preparations have also been reported. Total steroid saponins (TSS) of a D. zingiberensis rhizome containing 36 quantified compounds [161] alleviated arthritic progression in adjuvant-induced arthritis (AIA) rats in vivo, which correlated with suppression of the overproduction of inflammatory cytokines, oxidant stress makers, eicosanoids, and inflammatory enzymes, while the release of SOD and IL-10 increased [19]. Zhou et al. [145] reported that monosodium-urate-crystals-induced gouty arthritis in rats may be treated with Chinese herbal medicine (D. nipponica) by regulating lysosomal enzymes, antioxidant capacities and the NALP3 inflammasome; the authors demonstrated that the total saponin groups could increase the activities of GPx and SOD, and reduce the activities of several enzymes and the content of several important inflammatory biomarkers. Orally administered TS from D. nipponica remarkably decreased MDA, iNOS and NO levels in mice, while the levels of GSH, GSH-Px and SOD increased [162]. Water extract from D. opposita significantly increased plasma SOD activity and decreased plasma MDA concentration in hypertensive rats [163]. EtOH (95%) extract from Taiwanese yam (D. japonica, var. pseudojaponica) tubers decreased the LPS-induced NO production and expressions of iNOS and COX-2 in RAW264.7 cells; moreover, they had effects on the activities of CAT, SOD, and GPx, as well as on the levels of MDA in the edematous-paw mice [142]. The TPC, TFC, and radical-scavenging capacities of the extract were determined (Table 2 and Table 3). In the livers of mice treated with the EA fraction of D. bulbifera rhizome, the level of LPO increased remarkably, while the SOD, GPx, GST, GR, and glutamate-cysteine ligase (GCL) of the hepatic tissues all decreased conspicuously [164]. Yam (D. pseudojaponica) was also found to increase the activities of SOD and GPx, and to decrease the MDA level in the brains of D-gal-treated mice [165]. Rats fed with a high-cholesterol diet supplemented with either 0.1% or 0.5% diosgenin, for 6 weeks, exhibited a decrease in TBARS, while SOD in the plasma and liver, GSH-Px in the erythrocytes, and CAT in the erythrocytes and liver were significantly increased in the 0.5% diosgenin group [166]. The activity of SOD and CAT was significantly higher in a probucol and Dioscorea rhizome group of rabbits than in the control group fed chow containing 0.5% cholesterol and 10% corn oil [167]. The pre-intake of Chinese (D. alata) and Japanese (D. japonica) yam containing polyphenols and flavonoids significantly alleviated subsequent LPS-induced oxidative injury in mice by decreasing lipid oxidation levels and fibronectin production, and elevating SOD activity [100]. Strong antioxidant (+)-catechin, which was present in the extract of D. bulbifera bulbils, was suggested as a major bioactive constituent responsible for wound healing by promoting the cell proliferation and cell migration of human fibroblasts [62]. Yam (D. oppositifolia) extract demonstrated antioxidant and protective effects on the liver of rainbow trout [168], and an extract of Chinese yam (D. opposita) activated the antioxidant defense system in oxidatively damaged TM3 cells and KDS-Yang rats [147].

4. Phytochemicals of Dioscorea

Phytochemicals were studied in various Dioscorea spp., with saponins, phenanthrenes, and anthocyanins being the most widely investigated compounds. Therefore, this section does not discuss the very widely investigated steroidal saponins diosgenin and dioscin: several recently published reviews are available on the health benefits of these compounds (204–210). This section includes only short comments on the most recently-published data, which are summarized in Table 5. The structures of some secondary metabolites important for Dioscorea are given in Figure 1.
Flavonoids and phenolic acids are important and are the most widely studied antioxidants in many plants. The analysis of anthocyanins using LCMS-IT-TOF mass spectrometry in the vines of purple yams (D. alata) revealed that two investigated accessions from 8 months old had cyanidin or peonidin nuclei, while their glycosides were nonacylated, monoacylated, or diacylated with sinapic or ferulic acid. Cyanidin 3-(6-sinapoyl gentiobioside (alatanin C) was the major yam anthocyanin [93]. The compounds identified in the colored tubers of purple sachapapa (D. trifida) were derived from peonidin (Pn), cyanidin (Cy), and pelargonidin (Pg) aglycones, most of them showing the same substitution pattern, anthocyanin 3-acylglucoside-5-glucoside, where the acyl residue consisted of a hydroxycinnamic acid [113].
Some studies applied more modern extraction methods for Dioscorea. Among the 17 phenolic compounds—quantified in D. glabra leaf extracts isolated using microwave-assisted extraction—rosmarinic acid was the major one (22.31 ± 1.33 mg/g edw), while rutin was the dominant flavonoid, followed by quercetin; the latter compound was found in the highest quantity (8.66 ± 0.29 mg/g DW) in D. alata leaves [152]. Ochoa et al. [92] optimized ultrasound-assisted extraction (UAE) conditions for ethanolic anthocyanin-rich extracts from purple yam (D. alata); the best results were obtained at 60 °C for 10 min with 60% amplitude and an EtOH-to-water ratio of 80:20. Using SuperPro Designer® software (v. 8.5) (Intelligen Inc., Scotch Plains, NJ, USA) the authors determined that the cost of manufacturing decreased from 950.52 to 124.08 USD/kg of extract when the extractor capacity increased from 5 to 500 L; they concluded that UAE would be economically feasible when the selling price is above 170 USD/kg. It is evident that there is a need to expand the studies on using advanced techniques such supercritical and subcritical fluid, pressurized liquid extraction, enzyme assisted, and other methods for the recovery of bioactive constituents. The preservation of heat-sensitive antioxidants, meeting green chemistry principles, and sustainability aspects should be taken into account.
Phenolic acids and flavonoids (in total, 21 compounds) were quantified in 80% ethanol extract isolated from D. alata tubers at ambient temperature. Kaempferol (9.219 mg/100 g pdw) and myricetin (4.613 mg/100 g pdw) were major quantitative constituents, while the content of other identified phenolics was less than 1 mg/100 g pdw [94]. Anthocyanins are the main pigments in the purple yams, while according to the recent report of Zhao et al. [69], the yellow pigments of fresh-cut yam (D. opposita) slices were composed primarily of bisdemethoxycurcumin (73.7%) and two other unknown compounds.
Chaniad et al. [62] partitioned crude EtOH extract of D. bulbifera bulbils with HX, CF, EA, and water, and separated these fractions using chromatography; this resulted in the isolation of 8-epidiosbulbin E acetate; 15,16-epoxy-6α-O-acetyl-8β-hydroxy-19-nor-clero-13; 14-diene-17,12;18,2-diolide; sitosterol-β-D-glucoside; 3,5-dimethoxyquercetin; (+)-catechin; quercetin; kaempferol; allantoin; 2,4,3′,5′-tetrahydroxybibenzy; 2,4,6,7-tetrahydroxy-9,10 dihydrophenanthrene; and myricetin. Some of these compounds inhibited NO, DPPH, and OH.
Boudjada et al. [31] isolated 7 phenanthrene and dihydrophenanthrene derivatives from D. communis: some of them inhibited acetylcholinesterase (IC50 = 69.41 μg/mL) and butyrylcholinesterase (IC50 = 11.40, 14.60 and 14.34 μg/mL). Paul et al. [21] reported sex-specific variations in phytochemicals and the antimicrobial potentiality of D. alata, D. hamiltonii, D. oppositifolia, D. pubera, and D. wallichii tubers. Some studies reported phytochemicals in different yam anatomical parts. Yu et al. [171] compared phytochemicals in the Chinese yam (D. polystachya) tuber flesh and tuber cortex. Among the 38 detected compounds, dehydroepiandrosterone was more abundant in the flesh, while allantoin and flavonoids were more abundant in the cortex. In addition, the authors determined that the levels of allantoin, adenosine, and glutamine increased with the growing years. 2,7-Dihydroxy-4,6-dimethoxy phenanthrene, 6,7-dihydroxy-2,4-dimethoxy phenanthrene and 6-hydroxy-2,4,7-trimethoxyphenanthrene (batatasin I) were identified in the EtOH extracts of yam tuber (D. batatas) peel, while in the flesh, they were not detected; these phenanthrene derivatives scavenged DPPH and ABTS•+ [54]. Yang et al. [80] isolated and identified 19 phytochemicals (dihydrostilbenes, phenanthrenes, diarylheptanoids, and apigenin) in the CF-soluble fraction of D. opposita, and some of them scavenged DPPH and O2.
Itharat et al. [175] isolated eight compounds from an EtOH extract of D. membranacea, two naphthofuranoxepins (dioscorealides A and B); a 1,4-phenanthraquinone (dioscoreanone); three steroids (β-sitosterol, stigmasterol and β-D-sitosterol glucoside); and two steroid saponins, diosgenin-(3-O-α-L-rhamnopyranosyl (1→2)-β-D-glucopyranoside and diosgenin 3-O-β-D-glucopyranosyl (1→3)-β-D-glucopyranoside). Their DPPH scavenging capacity at 0.1 mg/mL ranged from 0.8 to 33.1%. Nine compounds were isolated from the 75% ethanol extract of D. septemloba rhizomes, including new phenanthropyran, dioscorone B; moreover, a new phenanthrene named 2,20,6,60—tetramethoxy-4,40,7,70-tetrahydroxy-1,10-biphenanthrene showed excellent DPPH-scavenging activities, with IC50 values of 0.07 ± 0.10 μM and 0.13 ± 0.09 μM, respectively [172]. Using UPLC-DAD-Q-TOF-MS/MS, Zhao et al. [29] detected 42 and 38 compounds in methanol extracts of D. oppositifolia (Chinese yam) and D. hamiltonii, respectively, while 14 of them were quantified using HPLC. In vitro propagation of D. birmanica shoots resulted in 0.37 ± 0.03% (w/w) of diosgenin-3-O-a-l-rhamnopyranosyl (1→2)–β-d-glucopyranoside, while its content in mother rhizomes was 3.27 ± 0.04% [108]. The β-Carotene content of MPYF (prepared using a modified process, boiled for polyphenol oxidase inhibition before drying)-substituted pasta was significantly higher compared to TPYF (prepared using a traditional process) samples [11]. One new bibenzyl and one new diarylheptanone and diobulbinone A, together with sixteen known compounds, were isolated form the rhizomes of D. bulbifera; this was accomplished by suspending it in H2O, successively extracting with 80% EtOH, petroleum ether, and ethyl acetate, and further fractionating with silica gel, Sephadex LH-20, and ODS [173]. It was reported that Dioscorea saponin (DS) can increase the exopolysaccharide production of tuber melanosporum [176].
Only a few reports are available on the analysis of Dioscorea extracts using gas chromatography including volatile constituents. 2-Acetyl furan and 2-acetylpyrrole had the highest odor activity (FD ≥ 128) factors in the roasted white yam (D. rotundata), while other compounds among the 29 aroma-active constituents with the FD factors ≥ 32 were 2-methylpyrazine, ethyl furfural, and 5-hydroxy methyl furfural [177]. Dey at al. [133] reported mainly higher fatty acids and their esters in an MeOH extract of freeze-dried tubers of D. alata’s various secondary metabolites. Alsawalha et al. [104] performed GC–MS analysis of an MeOH extract of D. villosa leaf powder; however, the identification results of the detected compounds, based on the raw MS software report, are too preliminary.

5. Conclusions

Numerous studies reported antioxidant properties in extracts and fractions isolated from different anatomical parts of Dioscorea spp. The majority of the published results were obtained in studies on plant tubers/rhizomes, while reports on leaves are rather scarce. Some studies focused on plant bulbils as well. Antioxidant potential studies applied mainly to in vitro chemical and bioassays using cell cultures, while in vivo studies with animal models were also available, albeit in smaller numbers. Some of the reported studies supported radical-scavenging capacity and other antioxidant activities of Dioscorea by investigating the phytochemical composition of the extracts and fractions, isolated using various solvents and methods. In general, leaf extracts were stronger antioxidants than rhizome/root preparations. Moreover, several studies demonstrated that tuber peel accumulate more antioxidants than its flesh. The antioxidant potential of different anatomical parts of Dioscorea may be related to their phytochemical composition. Steroidal saponins, phenanthrenes and anthocyanins (in purple yams) are important constituents in tubers, while phenolic acids and flavonoids are the main antioxidants determined in the leaves. Several studies also reported radical-scavenging properties in the mucilage, purified oligosaccharides and protein hydrolysis products obtained from rhizomes. Several in vivo studies using various animal models demonstrated the anti-inflammatory effects of Dioscorea preparations, which may be related to the increased levels of endogenous cell antioxidants, namely SOD, CAT, GPx, and reduced lipid peroxidation.
The reported data may assist in explaining ethnopharmacological knowledge regarding the health benefits of Dioscorea preparations. Moreover, such knowledge may foster a wider application of Dioscorea plants in developing new functional foods and nutraceuticals. In addition, it may be noted that, until now, published articles have focused mainly on the several most widely used species. A large number of Dioscorea spp. remain poorly investigated, and present a large pool for further studies.

Supplementary Materials

The following supporting information can be downloaded at: https://0-www-mdpi-com.brum.beds.ac.uk/article/10.3390/molecules27082530/s1. The list of abbreviations.

Author Contributions

P.R.V.: conceptualization, collecting and sorting out published materials, writing the draft manuscript (50%), data curation, reviewing and editing (100%); A.A.: compiling tables and figures, writing the draft manuscript (50%). 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 interests.

References

  1. Christenhusz, M.J.M.; Byng, J.W. The number of known plants species in the world and its annual increase. Phytotaxa 2016, 261, 201–217. [Google Scholar] [CrossRef] [Green Version]
  2. Kim, J.W.; Yang, M.H.; Yoon, K.D.; Chin, Y.W. Phytochemical and pharmacological profiles of Dioscorea species in Korea, China and Japan. Korean J. Pharmacogn. 2009, 40, 257–279. [Google Scholar]
  3. Lake, E.C.; Smith, M.C.; Dray, F.A., Jr.; Pratt, P.D. Ecological host-range of Lilioceris cheni (Coleoptera: Chrysomelidae), a biological control agent of Dioscorea bulbifera. Biol. Control 2015, 85, 18–24. [Google Scholar] [CrossRef]
  4. Georgia Exotic Pest Plant Council: GA-EPPC. List of Non-Native Invasive Plants in Georgia—Georgia Exotic Pest Plant Council. Available online: http://www.gaeppc.org/list/ (accessed on 11 November 2021).
  5. Lim, T.K. Edible Medicinal and Non-Medicinal Plants: Modified Stems, Roots, Bulbs; Springer: Berlin/Heidelberg, Germany, 2016; Volume 10, pp. 218–225. [Google Scholar]
  6. Food and Agricultural Organization of the United Nations (FAO). Available online: http://www.fao.org/ (accessed on 11 November 2021).
  7. Early Detection and Distribution Mapping System. (EDDMapS). Available online: http://www.eddmaps.org (accessed on 12 November 2021).
  8. Global Invasive Species Database Species Profile: Dioscorea oppositifolia. Available online: http://www.iucngisd.org/gisd/species.php?sc=296 (accessed on 22 November 2021).
  9. Obidiegwu, J.E.; Lyons, J.B.; Chilaka, C.A. The Dioscorea Genus (Yam)—An Appraisal of Nutritional and Therapeutic Potentials. Foods 2020, 9, 1304. [Google Scholar] [CrossRef]
  10. Adeloye, J.B.; Aluko, P.A.; Oluwajuyitan, T.D. In vitro α-amylase and α-glucosidase inhibitory activities, antioxidant activity, in vivo glycemic response and nutritional quality of dough meals from Dioscorea alata and Vernonia amygdalina. J. Food Meas. 2021, 15, 4083–4097. [Google Scholar] [CrossRef]
  11. Djeukeu, W.A.; Gouado, I.; Leng, M.S.; Vijaykrishnaraj, M.; Prabhasankar, P. Effect of dried yam flour (Dioscorea schimperiana) on cooking quality, digestibility profile and antioxidant potential of wheat based pasta. Food Meas. 2017, 11, 1421–1429. [Google Scholar] [CrossRef]
  12. Hwang, S.-J. Antioxidant activities and quality characteristics of yanggaeng added with Yam (Dioscorea japonica Thunb) powder. Culin. Sci. Hosp. Res. 2021, 27, 178–188. [Google Scholar]
  13. Kundu, B.B.; Vanni, K.; Farheen, A.; Jha, P.; Pandey, D.K.; Kumar, V. Dioscorea bulbifera L. (Dioscoreaceae): A review of its ethnobotany, pharmacology and conservation needs. S. Afr. J. Bot. 2020, 140, 365–374. [Google Scholar] [CrossRef]
  14. Zhou, Q.; Yu, D.H.; Zhang, C.; Liu, S.M.; Lu, F. Total saponins from Discorea nipponica ameliorate urate excretion in hyperuricemic mice. Planta Med. 2014, 80, 1259–1268. [Google Scholar] [CrossRef]
  15. Dutta, B. Food and medicinal values of certain species of Dioscorea with special reference to Assam. J. Pharmacogn. Phytochem. 2015, 3, 15–18. [Google Scholar]
  16. Surarit, W.; Nuengchamnong, N.; Hansakul, P. Protective effects of Dioscorea birmanica extract against oxidative stress-induced damage in cultured normal hepatocytes and its phytochemical constituents. Food Biosci. 2021, 41, 101030. [Google Scholar] [CrossRef]
  17. Chen, Y.; Chen, X.; Xiang, T.; Sun, B.; Luo, H.; Liu, M.; Chen, Z.; Zhang, S.; Wang, C. Total saponins from Dioscorea septemloba Thunb. reduce serum uric acid levels in rats with hyperuricemia through OATP1A1 up-regulation. J. Huazhong Univ. Sci. Technol. Med. Sci. 2016, 36, 237–242. [Google Scholar] [CrossRef] [PubMed]
  18. Park, Y.M.; Lim, J.H.; Jeong, H.J.; Seo, E.W. Protective effects of crude mucin and saponin from Dioscorea rhizoma on gastric ulcer induced by alcohol in rats. J. Life Sci. 2014, 24, 1200–1208. [Google Scholar] [CrossRef]
  19. Zhang, X.; Ito, Y.; Liang, J.; Liu, J.; He, J.; Sun, W. Therapeutic effects of total steroid saponin extracts from the rhizome of Dioscorea zingiberensis C.H.Wright in Freund’s complete adjuvant induced arthritis in rats. Int. Immunopharmacol. 2014, 23, 407–416. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Yuniastuti, A.; Iswari, R.S.; Susanti, R. Antioxidant Acitivity in Various Processed Products of Inferior Local Tubers (Dioscorea sp. L.). KnE Life Sci. 2017, 3, 35–40. [Google Scholar] [CrossRef] [Green Version]
  21. Paul, C.; Debnath, A.; Chakraborty, K.; Ghosh, S.; Bhattacharjee, A.; Debnath, B. Sex-specific variations in phytochemicals and antimicrobial potentiality of Dioscorea. Future J. Pharm. Sci. 2020, 6, 60. [Google Scholar] [CrossRef]
  22. Hazrin-Chong, N.H.; Azeem, A.M.; Nik Mat Zin, N.N.I.; Shah Mat Lazim, M.A. Effectiveness of Dioscorea hispida Dennst as antibacterial and antibiofilm agent. Malays. J. Microbiol. 2018, 14, 462–467. [Google Scholar] [CrossRef]
  23. Miah, M.M.; Das, P.; Ibrahim, Y.; Shajib, M.S.; Rashid, M.A. In vitro antioxidant, antimicrobial, membrane stabilization and thrombolytic activities of Dioscorea hispida Dennst. Eur. J. Integr. Med. 2018, 19, 121–127. [Google Scholar] [CrossRef]
  24. Feng, J.F.; Tang, Y.N.; Ji, H.; Xiao, Z.G.; Zhu, L.; Yi, T. Biotransformation of Dioscorea nipponica by Rat Intestinal Microflora and Cardioprotective Effects of Diosgenin. Oxidative Med. Cell. Longev. 2017, 2017, 1–9. [Google Scholar] [CrossRef]
  25. Dong, M.; Meng, Z.; Kuerban, K.; Qi, F.; Liu, J.; Wei, Y.; Wang, Q.; Jiang, S.; Feng, M.; Ye, L. Diosgenin promotes antitumor immunity and PD-1 antibody efficacy against melanoma by regulating intestinal microbiota. Cell Death Dis. 2018, 9, 1039. [Google Scholar] [CrossRef] [Green Version]
  26. Min, H.Y.; Jang, H.J.; Park, K.H.; Hyun, S.Y.; Park, S.J.; Kim, J.H.; Son, J.; Kang, S.S.; Lee, H.Y. The natural compound gracillin exerts potent antitumor activity by targeting mitochondrial complex II. Cell Death Dis. 2019, 10, 810. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Tao, X.; Yin, L.; Xu, L.; Peng, J. Dioscin: A diverse acting natural compound with therapeutic potential in metabolic diseases, cancer, inflammation and infections. Pharmacol. Res. 2018, 137, 259–269. [Google Scholar] [CrossRef] [PubMed]
  28. Wang, T.; Choi, R.C.Y.; Li, J.; Bi, C.W.C.; Ran, W.; Chen, X.; Dong, T.T.X.; Bi, K.; Tsim, K.W.K. Trillin, a steroidal saponin isolated from the rhizomes of Dioscorea nipponica, exerts protective effects against hyperlipidemia and oxidative stress. J. Ethnopharmacol. 2012, 139, 214–220. [Google Scholar] [CrossRef] [PubMed]
  29. Zhao, C.; Zhou, B.; Miao, J.; Li, X.; Jing, S.; Zhang, D.; Yijia Wang, J.; Li, X.; Huang, L.; Gao, W. Multicomponent analysis and activities for evaluation of Dioscorea oppositifolia and Dioscorea hamiltonii. Food Agric. Immunol. 2019, 30, 1148–1161. [Google Scholar] [CrossRef] [Green Version]
  30. Kovács, A.; Vasas, A.; Hohmann, J. Natural phenanthrenes and their biological activity. Phytochemistry 2008, 69, 1084–1110. [Google Scholar] [CrossRef]
  31. Boudjada, A.; Touil, A.; Bensouici, C.; Bendif, H.; Rhouati, S. Phenanthrene and dihydrophenanthrene derivatives from Dioscorea communis with anticholinesterase, and antioxidant activities. Nat. Prod. Res. 2019, 33, 3278–3282. [Google Scholar] [CrossRef]
  32. Kim, J.S.; Lim, J.; Han, D.; Gu, M.J.; Oh, J.; Lee, J.S. Anti-inflammatory and antioxidant effects of phenanthrene derivatives isolated from Dioscorea batatas Decne. Free Radic. Biol. Med. 2019, 139, S10–S57. [Google Scholar]
  33. Padhan, B.; Panda, D. Potential of neglected and underutilized yams (Dioscorea spp.) for improving nutrional security and health Benefits. Front. Pharmacol. 2020, 11, 496. [Google Scholar] [CrossRef] [Green Version]
  34. Kumar, S.; Das, G.; Shin, H.S.; Patra, J.K. Dioscorea spp. (A Wild Edible Tuber): A Study on Its Ethnopharmacological Potential and Traditional Use by the Local People of Similipal Biosphere Reserve, India. Front. Pharmacol. 2017, 8, 52. [Google Scholar] [CrossRef] [Green Version]
  35. George, T.T.; Obilana, A.O.; Oyeyinka, S.A. The prospects of African yam bean: Past and future importance. Heliyon 2020, 6, e05458. [Google Scholar] [CrossRef]
  36. Salehi, B.; Sener, B.; Kilic, M.; Sharifi-Rad, J.; Naz, R.; Yousaf, Z.; Mudau, F.N.; Fokou, P.V.T.; Ezzat, S.M.; El Bishbishy, M.H.; et al. Dioscorea Plants: A Genus Rich in Vital Nutra-pharmaceuticals—A Review. Iran. J. Pharm. Res. 2019, 18, 68–89. [Google Scholar] [PubMed]
  37. Bhujbal, A.S.; Baokar, S.; Mane, K.; Patil, G.; Patil, R.; Jain, P.; Pandey, A. Review on Pharmacological Activity of Dioscorea floribunda. J. Pharm. Res. Int. 2021, 33, 191–198. [Google Scholar] [CrossRef]
  38. Semwal, P.; Painuli, S.; Cruz-Martins, N. Dioscorea deltoidea wall. Ex Griseb: A review of traditional uses, bioactive compounds and biological activities. Food Biosci. 2021, 41, 100969. [Google Scholar] [CrossRef]
  39. Huang, R.; Xie, J.; Yu, Y.; Shen, M. Recent progress in the research of yam mucilage polysaccharides: Isolation, structure and bioactivities. Int. J. Biol. Macromol. 2020, 155, 1262–1269. [Google Scholar] [CrossRef] [PubMed]
  40. Anwar, M.; Birch, E.J.; Ding, Y.; Bekhit, A.E. Water-soluble non-starch polysaccharides of root and tuber crops: Extraction, characteristics, properties, bioactivities, and applications. Crit. Rev. Food Sci. Nutr. 2020, 9, 1–33. [Google Scholar] [CrossRef] [PubMed]
  41. Zhang, L.; Ng, T.B.; Lam, J.K.W.; Wang, S.W.; Lao, L.; Zhang, K.Y.; Sze, S.C.W. Research and Development of Proteins and Peptides with Therapeutic Potential from Yam Tubers. Curr. Prot. Pept. Sci. 2019, 20, 277–284. [Google Scholar] [CrossRef]
  42. Petropoulos, S.A.; Sampaio, S.L.; Di Gioia, F.; Tzortzakis, N.; Rouphael, Y.; Kyriacou, M.C.; Ferreira, I. Grown to be Blue-Antioxidant Properties and Health Effects of Colored Vegetables. Part I: Root Vegetables. Antioxidants 2019, 8, 617. [Google Scholar] [CrossRef] [Green Version]
  43. Yang, L.; Ren, S.; Xu, F.; Ma, Z.; Liu, X.; Wang, L. Recent Advances in the Pharmacological Activities of Dioscin. BioMed Res. Int. 2019, 2019, 5763602. [Google Scholar] [CrossRef] [Green Version]
  44. Tóth, B.; Hohmann, J.; Vasas, A. Phenanthrenes: A Promising Group of Plant Secondary Metabolites. J. Nat. Prod. 2018, 81, 661–678. [Google Scholar] [CrossRef]
  45. Kwon, J.B.; Kim, M.S.; Sohn, H.Y. Evaluation of antimicrobial, antioxidant, and antithrombin activities of the rhizome of various Dioscorea species. Korean J. Food Preserv. 2010, 17, 391–397. [Google Scholar]
  46. Chaudhury, S.; Habibur Rahaman, C.; Singh, H.; Chaudhuri, K.; Seal, T. Nutritional and Medicinal Importance of Dioscorea glabra R. Baron, a Potent Wild Edible Plant Consumed by the Lodha Tribal Community of West Bengal, India. Curr. Nutr. Food Sci. 2020, 16, 284–295. [Google Scholar] [CrossRef]
  47. Adedayo, B.C.; Ademiluyi, A.O.; Oboh, G.; Akindahunsi, A.A. Interaction of aqueous extracts of two varieties of Yam tubers (Dioscorea spp.) on some key enzymes linked to type 2 Diabetes in vitro. Int. J. Food Sci. Technol. 2012, 47, 703–709. [Google Scholar] [CrossRef]
  48. Larief, R.; Dirpan, A. Theresia Purple Yam Flour (Dioscorea alata L.) Processing Effect on Anthocyanin and Antioxidant Capacity in Traditional Cake “Bolu Cukke” Making. IOP Conf. Ser. Earth Environ. Sci. 2018, 207, 012043. [Google Scholar] [CrossRef]
  49. Ratnaningsih, R.; Richana, N.; Suzuki, S.; Fujii, Y. Effect of soaking treatment on anthocyanin, flavonoid, phenolic content and antioxidant activities of Dioscorea alata flour. Indones. J. Chem. 2018, 18, 656. [Google Scholar] [CrossRef] [Green Version]
  50. Anisuzzman, M.; Zilani, M.N.H.; Khushi, S.S.; Asaduzzman, M. Antioxidant, antibacterial potential and HPLC analysis of Dioscorea alata Bulb. Indones. J. Pharm. 2016, 27, 9–14. [Google Scholar]
  51. Chiu, H.W.; Peng, J.C.; Tsai, S.J.; Lui, W.B. Effect of extrusion processing on antioxidant activities of corn extrudates fortified with various Chinese Yams (Dioscorea sp.). Food Bioprocess Technol. 2012, 5, 2462–2473. [Google Scholar] [CrossRef]
  52. Guo, X.; Sha, X.; Cai, S.; Wang, O.; Ji, B. Antiglycative and antioxidative properties of ethyl acetate fraction of Chinese Purple Yam (Dioscorea alata L.) extracts. Food Sci. Technol. Res. 2015, 21, 563–571. [Google Scholar] [CrossRef] [Green Version]
  53. Chen, P.Y.; Tu, Y.X.; Wu, C.T.; Jong, T.T.; Chang, C.M.J. Continuous hot pressurized solvent extraction of 1,1-Diphenyl-2-picrylhydrazyl free radical scavenging compounds from Taiwan Yams (Dioscorea alata). J. Agric. Food Chem. 2004, 52, 1945–1949. [Google Scholar] [CrossRef]
  54. Kim, M.; Gu, M.J.; Lee, J.G.; Chin, J.; Bae, J.S.; Hahn, D. Quantitative analysis of bioactive phenanthrenes in Dioscorea batatas Decne peel, a discarded biomass from postharvest processing. Antioxidants 2019, 8, 541. [Google Scholar] [CrossRef] [Green Version]
  55. Duan, Y.; Kim, G.H.; Kim, H.S. Antioxidant activities of various extracts from Korean Yam (Dioscorea batatas DECNE.). J. Korean Appl. Sci. Technol. 2016, 33, 324–332. [Google Scholar] [CrossRef]
  56. Duan, Y.; Kim, G.H.; Kim, H.S. Determination of bioactive compounds and antioxidant activity of Yam (Dioscorea batatas DECNE.) on thermal treatment. J. Korean Appl. Sci. Technol. 2015, 32, 302–311. [Google Scholar] [CrossRef]
  57. Duan, Y.; Kim, G.H.; Seong, J.H.; Chung, H.S.; Kim, H.S. Antioxidant activities of n-butanol and ethyl acetate extracts from Yam (Dioscorea batatas DECNE.). J. Korean Appl. Sci. Technol. 2015, 32, 599–606. [Google Scholar] [CrossRef]
  58. Duan, Y.; Kim, H.S.; Kim, G.H. Evaluation of antioxidative activity of Korean Yam (Dioscorea batatas DECNE.) by n-butanol and ethyl acetate extracts. J. Korean Appl. Sci. Technol. 2015, 32, 312–319. [Google Scholar] [CrossRef]
  59. Duan, Y.; Kim, G.H.; Kim, H.S. Study on antioxidant activity of thermal treatment Yam (Dioscorea batatas DECN) by n-butanol and ethyl acetate extracts. J. Korean Appl. Sci. Technol. 2015, 32, 631–638. [Google Scholar]
  60. Duan, Y.; Kim, G.H.; Kim, H.S. Investigation of n-butanol and ethyl acetate Extracts from thermal treatment Yam (Dioscorea batatas DECNE.) for they antioxidant activity. J. Korean Appl. Sci. Technol. 2015, 32, 363–371. [Google Scholar] [CrossRef]
  61. Odeghe, O.; Adikwu, E.; Ojiego, C. Phytochemical and antioxidant assessments of Dioscorea bulbifera stem tuber. Biomed. Biotechnol. Res. J. 2021, 4, 305–311. [Google Scholar] [CrossRef]
  62. Chaniad, P.; Tewtrakul, S.; Sudsai, T.; Langyanai, S.; Kaewdana, K. Anti-inflammatory, wound healing and antioxidant potential of compounds from Dioscorea bulbifera L. bulbils. PLoS ONE 2020, 15, e0243632. [Google Scholar] [CrossRef]
  63. Mainasara, M.M.; Abu Bakar, M.F.; Md Akim, A.; Linatoc, A.C.; Abu Bakar, F.I.; Ranneh, Y.K.H. Secondary Metabolites, Antioxidant, and Antiproliferative Activities of Dioscorea bulbifera Leaf Collected from Endau Rompin, Johor, Malaysia. Evid.-Based Complement. Altern. Med. 2021, 2021, 1–10. [Google Scholar] [CrossRef]
  64. Ghosh, S.; Derle, A.; Ahire, M.; More, P.; Jagtap, S.; Phadatare, S.D.; Patil, A.B.; Jabgunde, A.M.; Sharma, G.K.; Shinde, V.S.; et al. Phytochemical analysis and free radical scavenging activity of medicinal plants Gnidia glauca and Dioscorea bulbifera. PLoS ONE 2013, 8, e82529. [Google Scholar] [CrossRef] [Green Version]
  65. Savi, A.; Calegari, G.C.; Santos, V.A.Q.; Pereira, E.A.; Teixeira, S.D. Chemical characterization and antioxidant of polysaccharide extracted from Dioscorea bulbifera. J. King Saud Univ. Sci. 2020, 32, 636–642. [Google Scholar] [CrossRef]
  66. Jo, H.M.; Kim, H.Y.; Heo, Y.J.; Song, D.H.; Kim, G.J.; Kim, I.S.; Yoon, J.A.; Chung, K.H.; An, J.H. Quality characteristics and antioxidant activity of Dioscorea bulbifera, under various oligosaccharide pre-soaking condition. Korean J. Food Nutr. 2017, 30, 1176–1183. [Google Scholar]
  67. Adeniran, A.A.; Sonibare, M.A. In vitro antioxidant activity, brine shrimp lethality and assessment of bioactive constituents of three wild Dioscorea species. Food Meas. 2017, 11, 685–695. [Google Scholar] [CrossRef]
  68. Adomėnienė, A.; Kazernavičiūtė, R.; Venskutonis, P.R. Vaistinių augalų paieška cukrinio diabeto prevencijai. Health Sci. 2020, 30, 65–68. [Google Scholar] [CrossRef]
  69. Zhao, L.; Wang, D.; Ma, Y.; Zhang, Y.; Zhao, X. Yellow pigment formation, pigment composition, and quality of fresh-cut yam (Dioscorea opposita) slices. RSC Adv. 2020, 10, 1104–1113. [Google Scholar] [CrossRef] [Green Version]
  70. Zhao, L.; Zhao, X.; Ma, Y.; Zhang, Y.; Wang, D. DNA Binding Characteristics and Protective Effects of Yellow Pigment from Freshly Cut Yam (Dioscorea opposita). Molecules 2020, 25, 175. [Google Scholar] [CrossRef] [Green Version]
  71. Park, S.H. Convergence dietary effects and antioxidant activity of Poria cocos, Dioscorea opposita, Nelumbo nucifera and Euryale ferox. J. Digit. Converg. 2016, 14, 583–590. [Google Scholar] [CrossRef] [Green Version]
  72. Zhao, C.; Li, X.; Miao, J.; Jing, S.; Li, X.; Huang, L.; Gao, W. The effect of different extraction techniques on property and bioactivity of polysaccharides from Dioscorea hemsleyi. Int. J. Biol. Macromol. 2017, 102, 847–856. [Google Scholar] [CrossRef]
  73. Luo, D. Identification of structure and antioxidant activity of a fraction of polysaccharide purified from Dioscorea nipponica Makino. Carbohydr. Polym. 2008, 71, 544–549. [Google Scholar] [CrossRef]
  74. Luo, D. Structural investigation of a polysaccharide (DMB) purified from Dioscorea nipponica Makino. Carbohydr. Polym. 2014, 103, 261–266. [Google Scholar] [CrossRef]
  75. Liu, Y.; Li, H.; Fan, Y.; Man, S.; Liu, Z.; Gao, W.; Wang, T. Antioxidant and antitumor activities of the extracts from Chinese Yam (Dioscorea opposite Thunb.) Flesh and peel and the effective Compounds. J. Food Sci. 2016, 81, H1553–H1564. [Google Scholar] [CrossRef]
  76. Mondal, M.; Hossain, M.M.; Das, N.; Rahman, M.A.; Uddin, N.; Hasan, M.R.; Alam, M.J.; Islam, M.N.; Wahed, T.B.; Kundu, S.K. Investigation of bioactivities of methanolic and ethyl acetate extracts of Dioscorea pentaphylla leaf along with its phenolic composition. Food Meas. 2019, 13, 622–633. [Google Scholar] [CrossRef]
  77. Kumar, S.; Mahanti, P.; Singh, N.R.; Rath, S.K.; Jena, P.K.; Patra, J.K. Antioxidant activity, antibacterial potential and characterization of active fraction of Dioscorea pentaphylla L. tuber extract collected from Similipal Biosphere Reserve, Odisha, India. Braz. J. Pharm. Sci. 2018, 53, e17006. [Google Scholar] [CrossRef]
  78. Kim, S.; Choi, S.C.; Youn, Y.H.; Ko, C.I.; Ha, Y.S.; Lee, I.A. Antioxidant and anti-inflammatory effects of Dioscorea japonica and Chenopodium album. J. Soc. Cosmet. Sci. Korea 2017, 43, 337–347. [Google Scholar]
  79. Mentha, M. Chromatographic fingerprinting, antioxidant, and anti-inflammatory potential of Dioscorea villosa (wild yam) leaves. Int. J. Green Pharm. 2018, 12, 102–106. [Google Scholar]
  80. Yang, M.H.; Yoon, K.D.; Chin, Y.W.; Park, J.H.; Kim, J. Phenolic compounds with radical scavenging and cyclooxygenase-2 (COX-2) inhibitory activities from Dioscorea opposita. Bioorg. Med. Chem. 2009, 17, 2689–2694. [Google Scholar] [CrossRef]
  81. Nagai, T.; Suzuki, N.; Nagashima, T. Antioxidative activity of water extracts from the yam (Dioscorea opposita Thunb.) tuber mucilage tororo. Eur. J. Lipid Sci. Technol. 2006, 108, 526–531. [Google Scholar] [CrossRef]
  82. Leng, M.S.; Tobit, P.; Demasse, A.M.; Wolf, K.; Gouado, I.; Ndjouenkeu, R.; Schweigert, F.J. Nutritional and anti-oxidant properties of yam (Dioscorea schimperiana) based complementary food formulation. Sci. Afr. 2019, 5, e00132. [Google Scholar] [CrossRef]
  83. Kim, K.M.; Kang, M.K.; Kim, J.S.; Kim, G.C.; Choi, S.Y. Physicochemical composition and antioxidant activities of Korean Dioscorea species. J. East Asian Soc. Diet. Life 2015, 25, 880–886. [Google Scholar] [CrossRef]
  84. Kim, H.S.; Duan, Y.; Ryu, J.Y.; Kim, S.W.; Jang, S.H. Chemical Composition of Thermal Treatment Yam (Dioscorea batatas DECNE.). J. Korean Appl. Sci. Technol. 2015, 32, 7–15. [Google Scholar] [CrossRef]
  85. 85 Nagai, T.; Suzuki, N.; Kai, N.; Tanoue, Y. Functional properties of autolysate and enzymatic hydrolysates from yam tsukuneimo (Dioscorea opposita Thunb.) tuber mucilage tororo: Antioxidative activity and antihypertensive activity. J. Food Sci. Technol. 2014, 51, 3838–3845. [Google Scholar] [CrossRef] [Green Version]
  86. Kim, J.S.; In, J.; Jang, H.S.; Sohn, H.Y. Evaluation of antimicrobial, antithrombin, and antioxidant activity of Dioscorea batatas Decne. Microbiol. Biotechnol. Lett. 2009, 37, 133–139. [Google Scholar]
  87. Ahn, S.M.; Jang, H.S.; Kwun, I.S.; Sohn, H.Y. Evaluation of Antimicrobial, Antithrombin, and Antioxidant Activity of Aerial Bulbils of Dioscorea batatas Decne. Microbiol. Biotechnol. Lett. 2009, 37, 266–272. [Google Scholar]
  88. Chung, Y.C.; Chiang, B.H.; Wei, J.H.; Wang, C.K.; Chen, P.C.; Hsu, C.K. Effects of blanching, drying and extraction processes on the antioxidant activity of yam (Dioscorea alata). Int. J. Food Sci. Technol. 2008, 43, 859–864. [Google Scholar] [CrossRef]
  89. Park, J.; Lee, J.H.; Bang, K.S. Evaluation of antioxidant capacity and antimutagen activity of bulbil extracts of the Dioscorea japonica Decaisne and Dioscorea batatas Decaisne. Korean J. Plant Res. 2012, 25, 200–208. [Google Scholar] [CrossRef] [Green Version]
  90. Santos, S.D.J.L.; Canto, H.K.F.; da Silva, L.H.M.; Rodrigues, A.M.D.C. Characterization and properties of purple yam (Dioscorea trifida) powder obtained by refractance window drying. Dry. Technol. 2020, 1–11. [Google Scholar] [CrossRef]
  91. Ramos-Escudero, F.; Muñoz, A.M.; Alvadaro-Ortiz, U.C.; Yáñez, J.A. Anthocyanins, polyphenols, antioxidant activity of purple sachapapa (Dioscorea trifida L.) and evaluation of lipid peroxidation in human serum. Rev. Soc. Química Perú 2010, 76, 61–72. [Google Scholar]
  92. Ochoa, S.; Durango-Zuleta, M.M.; Felipe Osorio-Tobón, J. Techno-economic evaluation of the extraction of anthocyanins from purple yam (Dioscorea alata) using ultrasound-assisted extraction and conventional extraction processes. Food Bioprod. Process. 2020, 122, 111–123. [Google Scholar] [CrossRef]
  93. Srivichai, S.; Hongsprabhas, P. Profiling anthocyanins in Thai Purple Yams (Dioscorea alata L.). Int. J. Food Sci. 2020, 2020, 1–10. [Google Scholar] [CrossRef]
  94. Chaudhury, S.; Habibur Rahaman, C.; Singh, H.; Chaudhuri, K.; Pillai, B.; Seal, T. Dioscorea alata: A potent wild edible plant consumed by the Lodha Tribal community of West Bengal, India. J. Pharmacogn. Phytochem. 2018, 7, 654–663. [Google Scholar]
  95. Chen, T.; Hu, S.; Zhang, H.; Guan, Q.; Yang, Y.; Wang, X. Anti-inflammatory effects of Dioscorea alata L. anthocyanins in a TNBS-induced colitis model. Food Funct. 2017, 8, 659–669. [Google Scholar] [CrossRef]
  96. Bhandari, M.R.; Kawabata, J. Organic acid, phenolic content and antioxidant activity of wild yam (Dioscorea spp.) tubers of Nepal. Food Chem. 2004, 88, 163–168. [Google Scholar] [CrossRef]
  97. Lee, Y.K.; Jung, B.; Chung, S. The Effect of Water-soluble chitosan on enhancing antioxidant activity of Dioscorea quinqueloba Thunb. extract. J. Chitin Chitosan 2015, 20, 196–201. [Google Scholar]
  98. Lu, F.C.; Lee, C.Y.; Wang, C.L. The influence of arbuscular mycorrhizal fungi inoculation on yam (Dioscorea spp.) tuber weights and secondary metabolite content. PeerJ 2015, 3, e1266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Lula, N.; Wirakartakusumah, A.M.; Andarwulan, N.; Purnomo, H.E.; Noda, T.; Ishiguro, K. Chemical Characterization of flour fractions from five Yam (Dioscorea alata) cultivars in Indonesia. J. Eng. Technol. Sci. 2015, 47, 92–103. [Google Scholar]
  100. Hsu, C.C.; Huang, Y.C.; Yin, M.C.; Lin, S.J. Effect of Yam (Dioscorea alata) compared to Dioscorea japonica) on gastrointestinal function and antioxidant activity in mice. J. Food Sci. 2006, 71, S513–S516. [Google Scholar] [CrossRef]
  101. Padhan, B.; Nayak, J.K.; Panda, D. Natural antioxidant potential of selected underutilized wild yams (Dioscorea spp.) for health benefit. J. Food Sci. Technol. 2020, 57, 2370–2376. [Google Scholar] [CrossRef]
  102. Barman, P.; Bhat, K.V.; Geeta, R. Phylogenetic analysis of Indian Dioscorea and comparison of secondary metabolite content with sampling across the tree. Genet. Resour. Crop Evol. 2018, 65, 1003–1012. [Google Scholar] [CrossRef]
  103. Jimenez-Montero, M.; Silvera, S.S. Nutritional evaluation of air potato (Dioscorea bulbifera L.) grown in Panama. Arch. Latinoam. Nutr. 2017, 67, 62–67. [Google Scholar]
  104. Alsawalha, M.; Al-Subaie, A.M.; Al-Jindan, R.; Bolla, S.R.; Salahuddin, M.F.; Veeraraghavan, V.P.; Sen, D.; Balakrishna, J.P.; Ravi, P.; Joseph, J.; et al. Dioscorea villosa leaf extract enhances in vitro wound healing and expression of extra cellular matrix factors transforming growth factor-beta 1 and collagen-1 in L929 cell lines. Pharmacogn. Mag. 2019, 15, 483–494. [Google Scholar]
  105. Zhang, Z.; Wang, X.; Liu, C.; Li, J. The degradation, antioxidant and antimutagenic activity of the mucilage polysaccharide from Dioscorea opposita. Carbohydr. Polym. 2016, 150, 227–231. [Google Scholar] [CrossRef]
  106. Xia, G.; Liu, W.; Song, J.; Zuo, J.; Zhang, Q.; Xue, C.; Zang, H.; Li, Y. Effective composition extraction and antioxidant activity of Dioscorea nipponica. Int. J. Agric. Biol. 2018, 20, 2251–2256. [Google Scholar]
  107. Adeniran, A.A.; Sonibare, M.A.; Rajacharya, G.H.; Kumar, S. Assessment of genetic fidelity of Dioscorea bulbifera L. and Dioscorea hirtiflora Benth. and medicinal bioactivity produced from the induced tuberous roots. Plant Cell Tissue Organ Cult. 2018, 132, 343–357. [Google Scholar] [CrossRef]
  108. Jirakiattikul, Y.; Rithichai, P.; Songsri, O.; Ruangnoo, S.; Itharat, A. In vitro propagation and bioactive compound accumulation in regenerated shoots of Dioscorea birmanica Prain & Burkill. Acta Physiol. Plant. 2016, 38, 249. [Google Scholar]
  109. Zhi, F.; Yang, T.L.; Wang, Q.; Jiang, B.; Wang, Z.P.; Zhang, J.; Chen, Y.Z. Isolation, structure and activity of a novel water-soluble polysaccharide from Dioscorea opposita Thunb. Int. J. Biol. Macromol. 2019, 133, 1201–1209. [Google Scholar] [CrossRef]
  110. Wang, T.S.; Lii, C.K.; Chang, J.Y.; Kuo, C.I.; Chen, H.P. Anti-Fenton reaction activity of three taxa of water yam (Dioscorea alata L.). Int. J. Food Sci. Technol. 2007, 42, 1107–1113. [Google Scholar] [CrossRef]
  111. Ma, F.; Li, X.; Ren, Z.; Särkkä-Tirkkonen, M.; Zhang, Y.; Zhao, D.; Liu, X. Effects of concentrations, temperature, pH and co-solutes on the rheological properties of mucilage from Dioscorea opposita Thunb. and its antioxidant activity. Food Chem. 2021, 360, 130022. [Google Scholar] [CrossRef]
  112. Shi, G.; Li, F.; Wang, G.; Lei, X.; Guo, T. Antioxidant activity of diosgenin from Dioscorea. Oxid. Commun. 2016, 39, 91–98. [Google Scholar]
  113. Ramos-Escudero, F.; Santos-Buelga, C.; Pérez-Alonso, J.J.; Yáñez, J.A.; Dueñas, M. HPLC-DAD-ESI/MS identification of anthocyanins in Dioscorea trifida L. yam tubers (purple sachapapa). Eur. Food Res. Technol. 2010, 230, 745–752. [Google Scholar] [CrossRef]
  114. Hou, W.C.; Lee, M.H.; Chen, H.J.; Liang, W.L.; Han, C.H.; Liu, Y.W.; Lin, Y.H. Antioxidant activities of dioscorin, the storage protein of yam (Dioscorea batatas Decne) tuber. J. Agric. Food Chem. 2001, 49, 4956–4960. [Google Scholar] [CrossRef]
  115. Hou, W.C.; Hsu, F.L.; Lee, M.H. Yam (Dioscorea batatas) tuber mucilage exhibited antioxidant activities in vitro. Planta Med. 2002, 68, 1072–1076. [Google Scholar] [CrossRef]
  116. Lee, M.H.; Lin, Y.S.; Lin, Y.H.; Hsu, F.L.; Hou, W.C. The mucilage of yam (Dioscorea batatas Decne) tuber exhibited angiotensin converting enzyme inhibitory activities. Bot. Bull. Acad. Sin. 2003, 44, 267–273. [Google Scholar]
  117. Nagai, T.; Suzuki, N.; Nagashima, T. Autolysate and enzymatic hydrolysates fro yam (Dioscorea opposita Thunb.) tuber mucilage tororo have antioxidant and angiotensin I-converting enzyme inhibitory activities. J. Food Agric. Environ. 2007, 5, 39–43. [Google Scholar]
  118. Nagai, T.; Nagashima, T.; Suzuki, N. Purification and Partial Characterization of Major Viscous Protein from Yam (Dioscorea opposita Thunb.) Tuber Mucilage tororo. Int. J. Food Prop. 2007, 10, 515–526. [Google Scholar] [CrossRef]
  119. Nagai, T.; Suzuki, N.; Tanoue, Y.; Kai, N. Antioxidant and antihypertensive activities of autolysate and enzymatic hydrolysates from yam (Dioscorea opposita Thunb) ichyoimo tubers. J. Food Agric. Environ. 2007, 5, 64–68. [Google Scholar]
  120. Nagai, T.; Nagashima, T. Functional properties of dioscorin, a soluble viscous protein from Japanese yam (Dioscorea opposita Thunb.) tuber mucilage Tororo. Z. Nat. C 2006, 61, 792–798. [Google Scholar] [CrossRef]
  121. Myoda, T.; Matsuda, Y.; Suzuli, T.; Nakagawa, T.; Nagai, T.; Nagashima, T. Identification of soluble proteins and interaction with mannan in mucilage of Dioscorea opposita Thunb. (Chinese yam tuber). Food Sci. Technol. Res. 2006, 12, 299–302. [Google Scholar] [CrossRef] [Green Version]
  122. Liu, Y.H.; Liang, H.J.; Cheng, H.C.; Liu, Y.W.; Hou, W.C. Comparisons of in vitro antioxidant activities of storage proteins in tuber of two Dioscorea species. Bot. Stud. 2006, 47, 231–237. [Google Scholar]
  123. Oh, P.S.; Lim, K.T. Antioxidant activity of Dioscorea batatas Decne glycoprotein. Eur. Food Res. Technol. 2008, 226, 507–515. [Google Scholar] [CrossRef]
  124. Setyawan, N.; Maninang, J.S.; Suzuki, S.; Fujii, Y. Variation in the physical and functional properties of Yam (Dioscorea spp.) Flour produced by different processing techniques. Foods 2021, 10, 1341. [Google Scholar] [CrossRef]
  125. Li, X.; Gao, X.; Lu, J.; Mao, X.; Wang, Y.; Feng, D.; Cao, J.; Huang, L.; Gao, W. Complex formation, physicochemical properties of different concentration of palmitic acid yam (Dioscorea pposita Thunb.) starch preparation mixtures. Food Sci. Technol. 2019, 101, 130–137. [Google Scholar] [CrossRef]
  126. Ghosh, S.; Nitnavare, R.; Dewle, A.; Tomar, G.B.; Chippalkatti, R.; More, P.; Kitture, R.; Kale, S.; Bellare, J.; Chopade, B.A. Novel platinum-palladium bimetallic nanoparticles synthesized by Dioscorea bulbifera: Anticancer and antioxidant activities. Int. J. Nanomed. 2015, 10, 7477–7490. [Google Scholar]
  127. Jaleel, C.A.; Manivannan, P.; Gomathinayagam, M.; Sridharan, R.; Panneerselvam, R. Responses of antioxidant potentials in Dioscorea rotundata Poir. following paclobutrazol drenching. Comptes Rendus Biol. 2007, 330, 798–805. [Google Scholar] [CrossRef] [PubMed]
  128. Simões, A.D.N.; Freire, C.S.; Silva, E.F.D.; Barros Junior, A.P.; Ferreira-Silva, S.L. Quality of minimaly processed Yam (Dioscorea sp.) stored at two different temperatures. Rev. Caatinga 2016, 29, 25–36. [Google Scholar] [CrossRef] [Green Version]
  129. Singthonh, J. Functional properties of purple Yam (Dioscorea alata) flou. Suranaree J. Sci. Technol. 2018, 25, 165–176. [Google Scholar]
  130. Pacheco-Delahaye, E.; Techeira, N.; García, A.D. Elaboration and evaluation of powders for instant drinks based upon extruded flour of yam (Dioscorea alata). Rev. Chil. Nutr. 2008, 35, 452–459. [Google Scholar]
  131. Tan, F.J.; Liao, F.Y.; Jhan, Y.J.; Liu, D.C. Effect of replacing pork backfat with yams (Dioscorea alata) on quality characteristics of Chinese sausage. J. Food Eng. 2007, 79, 858–863. [Google Scholar] [CrossRef]
  132. Li, Q.M.; Li, Y.; Zou, J.H.; Guo, S.Y.; Wang, F.; Yu, P.; Su, X.J. Influence of Adding Chinese Yam (Dioscorea opposita Thunb.) Flour on Dough Rheology, Gluten Structure, Baking Performance, and Antioxidant Properties of Bread. Foods 2020, 9, 256. [Google Scholar] [CrossRef] [Green Version]
  133. Dey, P.; Roy Chowdhuri, S.; Sarkar, M.P.; Chaudhuri, T.K. Evaluation of anti-inflammatory activity and standardisation of hydro-methanol extract of underground tuber of Dioscorea alata. Pharm. Biol. 2016, 54, 1474–1482. [Google Scholar] [CrossRef] [Green Version]
  134. Estiasih, T.; Umaro, D.; Harijono. Hypoglycemic effect of crude water soluble polysaccharide extracted from tubers of purple and yellow water yam (Dioscorea alata L.) on alloxan-induced hyperglycemia Wistar rats. Prog. Nutr. 2018, 20, 59–67. [Google Scholar]
  135. Chen, C.T.; Wang, Z.H.; Hsu, C.C.; Lin, H.H.; Chen, J.H. Taiwanese and Japanese yam (Dioscorea spp.) extracts attenuate doxorubicin-induced cardiotoxicity in mice. J. Food Drug Anal. 2017, 25, 872–880. [Google Scholar] [CrossRef] [Green Version]
  136. Lim, J.S.; Hahn, D.; Gu, M.J.; Oh, J.; Lee, J.S.; Kim, J.S. Anti-inflammatory and antioxidant effects of 2, 7-dihydroxy-4, 6-dimethoxy phenanthrene isolated from Dioscorea batatas Decne. Appl. Biol. Chem. 2019, 62, 29. [Google Scholar] [CrossRef]
  137. Ma, J.; Meng, X.; Liu, Y.; Yin, C.; Zhang, T.; Wang, P.; Park, Y.K.; Jung, H.W. Effects of a rhizome aqueous extract of Dioscorea batatas and its bioactive compound, allantoin in high fat diet and streptozotocin-induced diabetic mice and the regulation of liver, pancreas and skeletal muscle dysfunction. J. Ethnopharmacol. 2020, 259, 112926. [Google Scholar] [CrossRef] [PubMed]
  138. Byeon, S.; Oh, J.; Lim, J.; Lee, J.; Kim, J.S. Protective effects of Dioscorea batatas glesh and peel extracts against Ethanol-Induced gastric ulcer in mice. Nutrients 2018, 10, E1680. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Jin, M.; Suh, S.J.; Yang, J.H.; Lu, Y.; Kim, S.J.; Kwon, S.; Jo, T.H.; Kim, J.W.; Park, Y.I.; Ahn, G.W.; et al. Anti-inflammatory activity of bark of Dioscorea batatas DECNE through the inhibition of iNOS and COX-2 expressions in RAW264.7 cells via NF-κB and ERK1/2 inactivation. Food Chem. Toxicol. 2010, 48, 3073–3079. [Google Scholar] [CrossRef]
  140. Go, H.K.; Rahman, M.D.; Kim, G.B.; Na, C.S.; Song, C.H.; Kim, J.S.; Kim, S.J.; Kang, H.S. Antidiabetic Effects of Yam (Dioscorea batatas) and its active constituent, allantoin, in a rat Model of streptozotocin-Induced diabetes. Nutrients 2015, 7, 8532–8544. [Google Scholar] [CrossRef]
  141. Silva do Nascimento, E.; Anaya, K.; de Oliveira, J.M.C.; de Lacerda, J.T.J.G.; Miller, M.E.; Dias, M.; Mendes, M.A.; de Azevedo Lima Pallone, J.; Weis Arns, C.; Juliano, M.A.; et al. Identification of bioactive peptides released from in vitro gastrointestinal digestion of yam proteins (Dioscorea cayennensis). Food Res. Int. 2021, 143, 110286. [Google Scholar] [CrossRef]
  142. Chiu, C.S.; Deng, J.S.; Chang, H.Y.; Chen, Y.C.; Lee, M.M.; Hou, W.C.; Lee, C.Y.; Huang, S.S.; Huang, G.J. Antioxidant and anti-inflammatory properties of taiwanese yam (Dioscorea japonica Thunb. var. pseudojaponica (Hayata) Yamam.) and its reference compounds. Food Chem. 2013, 141, 1087–1096. [Google Scholar] [CrossRef]
  143. Hsu, W.H.; Lee, B.H.; Pan, T.M. Protection of Monascus-Fermented Dioscorea against DMBA-Induced Oral Injury in Hamster by Anti-inflammatory and Antioxidative Potentials. J. Agric. Food Chem. 2010, 58, 6715–6720. [Google Scholar] [CrossRef]
  144. Hwang, J.T.; Park, K.S.; Ryuk, J.A.; Kim, H.J.; Ko, B.S. Development of an Oriental Medicine Discrimination Method through Analysis of Steroidal Saponins in Dioscorea nipponica Makino and Their Anti-Osteosarcoma Effects. Molecules 2019, 24, 4022. [Google Scholar] [CrossRef] [Green Version]
  145. Zhou, Q.; Yu, D.; Zhang, N.; Liu, S. Anti-inflammatory Effect of Total Saponin Fraction from Dioscorea nipponica Makino on Gouty Arthritis and Its Influence on NALP3 Inflammasome. Chin. J. Integr. Med. 2019, 25, 663–670. [Google Scholar] [CrossRef]
  146. Tang, Y.N.; He, X.C.; Ye, M.; Huang, H.; Chen, H.L.; Peng, W.L.; Zhao, Z.Z.; Yi, T.; Chen, H.B. Cardioprotective effect of total saponins from three medicinal species of Dioscorea against isoprenaline-induced myocardial ischemia. J. Ethnopharmacol. 2015, 175, 451–455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Yu, S.; Han, B.; Bai, X.; Liu, S.; Xing, X.; Zhao, D.; Liu, M.; Wang, S. The cold-soaking extract of Chinese yam (Dioscorea opposita Thunb.) protects against erectile dysfunction by ameliorating testicular function in hydrocortisone-induced KDS-Yang rats and in oxidatively damaged TM3 cells. J. Ethnopharmacol. 2020, 263, 113223. [Google Scholar] [CrossRef] [PubMed]
  148. Liu, C.; Lu, D.; You, J.; Lu, Y.; Sun, J.; Pan, J.; Wang, Y.; Wang, A.; Lan, Y.; Li, Y.; et al. Efficacy of water fraction from Dioscorea cirrhosa on oxidative stress and apoptosis in H9c2 cardiomyocytes induced by H2O2. J. Tradit. Chin. Med. 2021, 41, 51–58. [Google Scholar]
  149. Hwang, K.A.; Hwang, Y.J.; Kim, H.S.; Hwang, H.J.; Song, J.; Kim, Y.J. Anti-inflammatory effect of aerial bulblets of Dioscorea japonica Thunb extract through inhibition of NF-κB and MAPK signalling pathway in RAW 264.7. J. Chin. Med. Assoc. 2019, 82, 251–255. [Google Scholar] [CrossRef]
  150. Siddiqiu, M.A.; Ali, Z.; Chittiboyina, A.G.; Khan, I.A. Hepatoprotective effect of steroidal glycosides from Dioscorea villosa on hydrogen peroxide-Induced hepatotoxicity in HepG2 Cells. Front. Pharmacol. 2018, 9, 797. [Google Scholar] [CrossRef] [Green Version]
  151. Ren, Y.J.; Cao, Y.G.; Zeng, M.N.; Zhang, B.B.; Liu, Y.L.; Wang, M.N.; Zhang, Y.L.; Feng, W.S.; Zheng, X.K. Two new norsesquiterpenoids with estrogenic activity from the stems and leaves of Dioscorea oppositifolia L. Nat. Prod. Res. 2019, 35, 3018–3025. [Google Scholar] [CrossRef]
  152. Zhou, L.; Shi, X.; Ren, X.; Qin, Z. Chemical composition and antioxidant activity of phenolic compounds from Dioscorea (Yam) leaves. Pak. J. Pharm. Sci. 2018, 31, 1031–1038. [Google Scholar]
  153. Zhou, J.; Xi, Y.; Zhang, J.; Tang, J.; Zhou, X.; Chen, J.; Nie, C.; Zhu, Z.; Ma, B. Protective effect of Dioscorea zingiberensis ethanol extract on the disruption of blood–testes barrier in high-fat diet/streptozotocin-induced diabetic mice by upregulating ZO-1 and Nrf2. Andrologia 2020, 52, e13508. [Google Scholar] [CrossRef]
  154. Fan, Y.; He, Q.; Luo, A.; Wang, M.; Luo, A. Characterization and antihyperglycemic activity of a polysaccharide from Dioscorea opposita Thunb roots. Int. J. Mol. Sci. 2015, 16, 6391–6401. [Google Scholar] [CrossRef] [Green Version]
  155. Poon, T.Y.C.; Ong, K.L.; Cheung, B.M.Y. Review of the effects of the traditional Chinese medicine Rehmannia Six Formula on diabetes mellitus and its complications. J. Diabetes 2011, 3, 184–200. [Google Scholar] [CrossRef]
  156. Lim, J.S.; Oh, J.; Byeon, S.; Lee, J.S.; Kim, J.S. Protective Effect of Dioscorea batatas peel extract against intestinal inflammation. J. Med. Food 2018, 21, 1204–1217. [Google Scholar] [CrossRef]
  157. Seo, E.W.; Lim, J.H.; Park, Y.M. Effect of Dioscorea rhizoma on gastritis by acute gastric ulcer in rats. Korean J. Plant Res. 2015, 28, 1–8. [Google Scholar]
  158. Son, I.S.; Lee, J.S.; Lee, J.Y.; Kwon, C.S. Antioxidant and anti-inflammatory effects of yam (Dioscorea batatas Decne.) on Azoxymethane-induced colonic Aberrant Crypt Foci in F344 rats. Prev. Nutr. Food Sci. 2014, 19, 82–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Cui, H.; Li, T.; Wang, L.; Su, Y.; Xian, C.J. Dioscorea bulbifera polysaccharide and cyclophosphamide combination enhances anti-cervical cancer effect and attenuates immunosuppression and oxidative stress in mice. Sci. Rep. 2016, 5, 19185. [Google Scholar] [CrossRef] [Green Version]
  160. Jayachandran, K.S.; Vasanthi, R.H.; Rajamanickam, R.G. Flavonoid Rich Fraction of Dioscorea bulbifera Linn. (Yam) Enhances Mitochondrial Enzymes and Antioxidant Status, Thereby Protects Heart from Isoproterenol Induced Myocardial Infarction. Curr. Pharm. Biotechnol. 2010, 11, 887–894. [Google Scholar] [CrossRef]
  161. Zhang, X.; Liang, J.; Liu, J.; Zhao, Y.; Gao, J.; Sun, W.; Ito, Y. Quality control and identification of steroid saponins from Dioscorea zingiberensis C. H. Wright by fingerprint with HPLC-ELSD and HPLC-ESI-Quadrupole/Time-of-fight tandem mass spectrometry. J. Pharm. Biomed. Anal. 2014, 1, 46–59. [Google Scholar] [CrossRef] [Green Version]
  162. Yu, H.; Zheng, L.; Yin, L.; Xu, L.; Qi, Y.; Han, X.; Xu, Y.; Liu, K.; Peng, J. Protective effects of the total saponins from Dioscorea nipponica Makino against carbon tetrachloride-induced liver injury in mice through suppression of apoptosis and inflammation. Int. Immunopharmacol. 2014, 2, 233–244. [Google Scholar] [CrossRef]
  163. Amat, N.; Amat, R.; Abdureyim, S.; Hoxur, P.; Osman, Z.; Mamut, D.; Kijjoa, A. Aqueous extract of Dioscorea opposita Thunb. normalizes the hypertension in 2K1C hypertensive rats. BMC Complement. Altern. Med. 2014, 14, 36. [Google Scholar] [CrossRef] [Green Version]
  164. Wang, J.; Ji, L.; Liu, H.; Wang, Z. Study of the hepatotoxicity induced by Dioscorea bulbifera L. rhizome in mice. Biosci. Trends 2010, 4, 79–85. [Google Scholar]
  165. Chiu, C.S.; Deng, J.S.; Hsieh, M.T.; Fan, M.J.; Lee, M.M.; Chueh, F.S.; Han, C.K.; Lin, Y.C.; Peng, W.H. Yam (Dioscorea pseudojaponica Yamamoto) ameliorates cognition deficit and attenuates oxidative damage in senescent mice induced by D-galactose. Am. J. Chin. Med. 2009, 37, 889–902. [Google Scholar] [CrossRef]
  166. Son, I.S.; Kim, J.H.; Sohn, H.Y.; Son, K.H.; Kim, J.S.; Kwon, C.S. Antioxidative and hypolipidemic effects of diosgenin, a steroidal saponin of Yam (Dioscorea spp.), on high-cholesterol fed rats. Biosci. Biotechnol. Biochem. 2007, 71, 3063–3071. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Chang, W.C.; Yu, Y.M.; Wu, C.H.; Tseng, Y.H.; Wu, K.Y. Reduction of oxidative stress and atherosclerosis in hyperlipidemic rabbits by Dioscorea rhizome. Can. J. Physiol. Pharmacol. 2005, 83, 423–430. [Google Scholar] [CrossRef] [PubMed]
  168. Wang, F.; Liu, F.; Wang, X.; Chen, W.; Meng, X. Transcriptomic responses to yam (Dioscorea oppositifolia L.) extract dietary supplementation in rainbow trout (Oncorhynchus mykiss) liver. Aquac. Res. 2020, 51, 932–945. [Google Scholar] [CrossRef]
  169. Szakiel, A.; Grabarczyk, M.; Pączkowski, C.; Mieczkowski, A. Comparison of the profiles of non-glycosylated triterpenoids from leaves of plants of selected species of genus Dioscorea. Phytochem. Lett. 2017, 20, 350–355. [Google Scholar] [CrossRef]
  170. Yang, G.Y.; Song, J.N.; Chang, Y.Q.; Wang, L.; Zheng, Y.G.; Zhang, D.; Guo, L. Natural deep eutectic solvents for the extraction of bioactive steroidal saponins from Dioscoreae nipponicae rhizoma. Molecules 2021, 26, 2079. [Google Scholar] [CrossRef]
  171. Yu, Y.; Guo, X.; Li, X.; Dai, D.; Xu, X.; Ge, X.; Li, Y.; Yang, T. Organ- and Age-Specific Differences of Dioscorea polystachya Compounds Measured by UPLC-QTOF/MS. Chem. Biodiver. 2021, 18, e2000856. [Google Scholar] [CrossRef]
  172. He, T.; Liu, Y.; Xiong, R.F.; Li, X.Y.; Wang, S.F.; Cheng, C.R. A new phenanthropyran and a new biphenanthrene from the rhizomes of Dioscorea septemloba and their antioxidant activities. Nat. Prod. Res. 2019, 34, 2202–2207. [Google Scholar] [CrossRef]
  173. Liu, H.; Tsim, K.W.K.; Chou, G.X.; Wang, J.M.; Ji, L.L.; Wang, Z.T. Phenolic Compounds from the Rhizomes of Dioscorea bulbifera. Chem. Biodivers. 2011, 8, 2110–2116. [Google Scholar] [CrossRef]
  174. 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]
  175. Itharat, A.; Plubrukan, A.; Kaewpradub, N.; Chuchom, T.; Ratanasuwan, P.; Houghton, P.J. Selective cytotoxicity and antioxidant effects of compounds from Dioscorea membranacea rhizomes. Nat. Prod. Commun. 2007, 2, 643–648. [Google Scholar] [CrossRef] [Green Version]
  176. Liu, X.; Zhang, Z.; Xu, L.; Shi, W.; Sun, Q.; Ding, C.; Zheng, H.; Wang, F. Dioscorea saponin transforms the structure of truffle exo-polysaccharide and enhances its antioxidant activity. LWT 2020, 127, 109417. [Google Scholar] [CrossRef]
  177. Lasekan, O.; Teoh, L.S. Contribution of aroma compounds to the antioxidant properties of roasted white yam (Dioscorea rotundata). BMC Chem. 2019, 13, 133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. The structures of the main important Dioscorea secondary metabolites.
Figure 1. The structures of the main important Dioscorea secondary metabolites.
Molecules 27 02530 g001
Table 1. Search results for available publications on Dioscorea (accessed on 21 December 2021).
Table 1. Search results for available publications on Dioscorea (accessed on 21 December 2021).
Search WordsWoSScience DirectPubMed
TotalReviewTotalReviewTotalReview
Dioscorea (topic)47792185171553166561
Dioscorea (title)215224508293716
Dioscorea + antioxidant (topic + topic)509262574120312
Dioscorea + antioxidant (title + topic)237710921081
Dioscorea + antioxidant (title + title)731na *na00
* na, not applicable.
Table 2. Antioxidant characteristics of various Dioscorea spp.
Table 2. Antioxidant characteristics of various Dioscorea spp.
Species: Plant PartSolvent, Analyzed ProductAntioxidant Characteristics Ref.
D. alata (2)/D. bulbifera/D. batatas/D. nipponica: rhizomeMeOH-EDPPH (IC50, µg/mL): 142.30 ± 2.58
and 486.43 ± 8.45/421.70 ± 17.24/432.66 ± 8.07
and 403.16 ± 14.59/371.64 ± 12.59
(AA, 10.77 ± 0.13; α-Toc, 40.24 ± 6.98; BHT, 11.92 ± 10.67)
[45]
D. alata: dried tubersEtOH (80%)-EDPPH (IC50, mg/gm dw): 0.603 ± 0.010
ABTS•+ (IC50, mg/gm dw): 0.136 ± 0.001
[46]
D. alata: flour/pasteH2O 1:10 (w/v)DPPH (IC50, mg/mL): 18.52 ± 2.1/17.86 ± 0.9
ABTS•+ (mmol TE/100 g): 1.04 ± 0.00/1.15 ± 0.01
Fe3+ RP (mmol AAE/100 g): 0.96 ± 0.06/1.18 ± 0.10
OH (%): 49.37 ± 1.52/53.88 ± 4.59
Fe2+ chelating (%): 11.13 ± 2.77/18.10 ± 1.38
[47]
D. alata: purple yamEffect of processingDPPH (%): 79.08 (raw), 61.75 (blanched), 40.75 (washed), 32.16 (dried), 30.01 (flour)[48]
D. alata: flour of tubersH2O + AcA (5%)-E DPPH (EC50, mg/mL): 2.55 to 8.70, depending on origin and treatment [49]
D. alata: bulbMeOH-EDPPH (IC50, μg/mL): 14.68 (AA 24.95)
Fe3+ RP (max absorption at 1 mg/mL): 1.317; BHT 1.472
[50]
D. alata (2 var);
D. doryophora;
commercial
Yam flourORAC (μmol TE/g): Tai-nung, 119.34; Ta-shan, 65.42
Hang-chun 60.63
103.17
[51]
D. alata: dried tubers of Chinese purple yamEtOH (80%)-E: flesh/peelDPPH (IC50, µg de/mL): 183.4 ± 5.3/47.7 ± 2.7;
FRAP (mg FeSO4·7H2O/g de): 86.5 ± 1.6/144.5 ± 8.5
ABTS•+ (mg TE/g de): 108.1 ± 2.8/357.7 ± 8.4
O2 (IC50, μg de/mL): 457.5 ± 20.8/162.2 ± 9.8
ChelA (%): 24.3 ± 1.6/21.4 ± 0.2
[52]
EA fr: flesh/peelDPPH: 13.9 ± 1.1/8.5 ± 0.2; FRAP: 630.1 ± 19.4/534.4 ± 35.3
ABTS•+: 1326.1 ± 17.3/1578.1 ± 15.5; O2: 78.2 ± 1.2/30.3 ± 0.3
ChelA: 15.1 ± 1.1/8.5 ± 1.0
BuOH fr: flesh/peelDPPH: 245.1 ± 32.1/43.8 ± 3.3; FRAP: 83.9 ± 5.3/162.1 ± 3.0; ABTS•+: 111.8 ± 5.5/448.4 ± 9.0; O2: 333.2 ± 10.6/147.9 ± 4.1
ChelA: 0.0 ± 0.1/34.3 ± 3.2
HX fr: flesh/peelDPPH: 632.0 ± 26.9/55.0 ± 0.9; FRAP: 42.3 ± 1.2/97.3 ± 11.2; ABTS•+: 39.8 ± 1.6/242.3 ± 14.4; O2: 1387.5 ± 110.3/237.9 ± 12.2; ChelA: 32.0 ± 2.2/34.1 ± 1.2
Remaining H2O fr: flesh/peelDPPH: 1532.7 ± 123/225.1 ± 12.9; FRAP: 8.5 ± 0.5/29.1 ± 1.7; ABTS•+: 12.2 ± 0.3/74.0 ± 2.9; O2: 874.5 ± 18.2/461.1 ± 14.1
ChelA: 50.4 ± 5.1/29.1 ± 0.6
D. alata: tubers (3 cultivars from Taiwan)Hot pressurized EtOH-E: flesh/peel DPPH (EC50, µg/mL): ND/86.6–305.4[53]
fr of HX/CF/EA/BuOH/H2O Flesh: 328–2360/144.6–549.4/112.8–343.6/288.1–1526/ND
Peel: 133.0–932.7/45.8–136.6/14.5–38.8/67.2–678.3/ND
D. batatas: tubers, 3 purified phenanthrenesEtOH (95%)-E flesh/peelDPPH (IC50, mg/mL): 7.68/0.944
ABTS•+ (IC50, mg/mL): 3.43/0.771
[54]
2,7-dOH-4,6-dMetOP/6,7-dOH -2,4-dMetOP/6-OH-2,4,7-tMetOPDPPH (IC50, mg/mL): 0.0645/0.154/0.566
ABTS•+ (IC50, mg/mL): 0.0482/0.153/0.297
D. batatas: dried and smashed into raw yam mealsEtOH (70%)/MeOH (70%)/CF: MeOH (2:1)DPPH (IC50, mg/mL): 1.22 ± 0.03/1.34 ± 0.02/0.71 ± 0.00
ABTS•+ (IC50, mg/mL): 2.08 ± 0.20/2.24 ± 0.10/1.15 ± 0.05
FRAP (μM Fe2+ at 1.0 mg/mL): 108.3 ± 0.0/91.67 ± 0.48/220.6 ± 0.7
[55]
EtOH (70%)/MeOH (70%)/CF: MeOH (2:1)Fe2+ chelating: 0.12 ± 0.02/0.09 ± 0.01/0.97 ± 0.03
NO: 0.46 ± 0.02/0.45 ± 0.00/0.55 ± 0.02
β-carotene bleaching: 0.15 ± 0.04/0.07 ± 0.01/0.14 ± 0.01
LPI: 0.50 ± 0.01/0.58 ± 0.00/0.05 ± 0.01(IC50, mg/mL for all)
[56]
D. batatas: raw yamBuOH/EAABTS•+ (IC50, mg/mL): 0.70 ± 0.01/0.45 ± 0.01
DPPH (IC50, mg/mL): 0.50 ± 0.00/0.34 ± 0.01; FRAP (μM Fe2+) ~347/~560 at 1 mg/mL
[57]
D. batatas: raw yamBuOH/EAFe2+ chelating: 0.64 ± 0.01/0.70 ± 0.01
NO: 0.53 ± 0.01/0.26 ± 0.03;
NO2 scavenging: 1.92 ± 0.03/3.92 ± 1.00
β-carotene bleaching: 0.08 ± 0.01/0.05 ± 0.01
LPI: 0.02 ± 0.01/0.01 ± 0.00 (IC50, mg/mL for all assays)
[58]
D. batatas: thermally treated yamBuOH/EAABTS•+ (IC50, mg/mL) 1.31 ± 0.05/0.98 ± 0.02
DPPH (IC50, mg/mL) 0.70 ± 0.00/0.74 ± 0.01
FRAP (μM Fe2+): 237.86 ± 4.07/244.05 ± 9.37
[59]
D. batatas: tubers (thermally treated yam)BuOH/EAFe2+ chelating: 0.11 ± 0.02/0.81 ± 0.01
NO: 0.73 ± 0.01/0.45 ± 0.02
NO2 scavenging: 3.78 ± 1.24/3.87 ± 0.40
β-carotene bleaching: 0.11 ± 0.01/0.11 ± 0.00
LPI: 0.02 ± 0.01/0.05 ± 0.01 (IC50, mg/mL for all assays)
[60]
D. birmanica: freeze-dried rhizomesEtOH (95%) extract (EC50, μg/mL) DPPH, 8.53 ± 1.32; ABTS•+, 21.56 ± 1.72; O2, 50.91 ± 0.39; NO, 26.93 ± 4.79; LPI: 33.37 ± 2.88.
FRAP (mg Fe2+ eq/g extract): 406.96 ± 11.33
Fe2+ chelating (EC50, mg/mL): 1.06 ± 0.03
[16]
D. bulbifera: tuberMeOH (80%)DPPH (IC50, μg/mL): 261.09; AA, 10.65; O2, 2089.3; Q, 17.01[61]
D. bulbifera: bulbilsEtOH/H2O crude extractsDPPH (IC50, μM): 34.14 ± 0.68/13.20 ± 0.77
OH (IC50, μM): 79.00 ± 0.78/>100
[62]
CF/EA/H2O fr of EtOHDPPH: 13.35 ± 0.37/14.00 ± 0.36/18.83 ± 0.46
OH: >100/37.04 ± 0.50/39.31 ± 0.42
D. bulbifera: leavesMeOH/EA/HX extractsDPPH (%): 79.0 ± 0.31/23.2 ± 0.05/11.5 ± 0.31
ABTS•+ (mg AAE/g): 65.6 ± 0.35/59.5 ± 0.10/14.9 ± 0.05
FRAP (mM/Fe2+g dw): 31.34 ± 2.06/10.98 ± 0.64/9.50 ± 0.48
[63]
D. bulbifera: bulbPE/EA/MeOH (sequentially)/EtOH (70%)DPPH (%): 61.82 ± 1.55/82.79 ± 1.24/76.11 ± 1.26/80.64 ± 1.24
O2 (%): 26.88 ± 1.28/57.60 ± 0.81/59.75 ± 0.98/54.76 ± 1.20
O2 (%): 28.30 ± 0.36/59.24 ± 1.44/59.65 ± 1.41/57.34 ± 1.41
NO (%): 20.57 ± 0.57/54.55 ± 0.21/57.59 ± 0.64/49.85 ± 0.16
OH (%): 44.51 ± 0.49/66.67 ± 0.73/76.11 ± 1.26/64.23 ± 1.25
[64]
D. bulbifera: tubersPre-purified/crude polysaccharideDPPH (mg IE/g): 0.28 ± 0.01/0.94 ± 0.02
ABTS•+ (mg TE/g): 623.33 ± 4.71/165.00 ± 2.36
FRAP (mM Fe2+/g): 0.175 ± 0.001/1.056 ± 0.001
[65]
D. bulbifera: tubersPre-soaking in 0–10% oligosaccharideDPPH (0.1 mL): ~17–27%
ABTS•+ (0.1 mL): ~23–45% (determined from figure)
[66]
D. hirtiflora: tubersSuccessively DCM/EA/MeOHDPPH (IC50, μg/mL): 49.7 ± 0.97/11.9 ± 0.85/11.8 ± 0.23[67]
D. dumetorum89.0 ± 5.10/103.2 ± 6.9/137 ± 5.90
D. bulbifera (mauve)46.7 ± 1.57/14.6 ± 0.90/29.9 ± 0.68
D. bulbifera (yellow)57.7 ± 1.32/64.1 ± 0.89/68.6 ± 8.50 (AA, 6.90; GA, 8.60)
D. caucasica: freeze-dried leavesEtOH (70%)DPPH (mg TE/g edw): 279 ± 4
ABTS•+ (mg TE/g edw): 880 ± 10
[68]
D. communis: rhizomeDE/EA crude extractsDPPH: 8.7 ± 0.9/40.1 ± 0.2; ABTS•+, 7.6 ± 0.0/11.6 ± 0.0; FRAP, 44.8 ± 3.6/79.0 ± 0.0; CUPRAC, 10.01 ± 0.2/20.8 ± 0.5 (all IC50, µg/mL)[31]
3 purified phenanthrenesDPPH, <200/61.2 ± 1.1/6.0 ± 0.2; ABTS•+, <200/19.60 ± 0.0/2.4 ± 0.1; FRAP, <50/<50/9.9 ± 1.0; CUPRAC, <200/194.0 ± 0/15.0 ± 0.5 (all IC50, µg/mL)
D. opposita: fresh-cut yamH2O/MeOH/EA-EORAC (µmol TE/g): ~33/63/48 (white); ~29/96/73 (yellow)
DPPH (mmol TE/g): ~11/10/8.5 (white); ~9/15/8.5 (yellow) (determined from the figures)
[69]
D. opposita: purified yellow pigmentMeOH-Amberlite XAD-7-Sep-Pak C18OH scavenging (IC50 mg/mL): 0.098 ± 0.032 (AA, 1.21 ± 0.0)[70]
D. hamiltonii: herbMeOH-E Fe3+ RP (mg AAE/g): 3.30 at 0.5 g/mL[29]
D. opposita: herbMeOH-EFe3+ RP (mg AAE/g): 4.71 at 0.5 g/mL[29]
D. opposita: herbal medicine productEtOH (70%)-E
5 g/50 mL
DPPH (%, 0.5 mL): 43.2 ± 2.35; ABTS•+ (%, 20 μL): 40.01 ± 3.0; SOD (%, 0.2 mL): 39.97 ± 8.87[71]
D. hemsleyi: rhizomeCold/warm/hot H2O extracted polysaccharidesDPPH (IC50, mg/mL): 4.56 ± 0.15/6.95 ± 0.13/8.85 ± 0.16
Fe3+ RP (mg AAE/g): 42.98 ± 0.79/31.78 ± 0.35/25.64 ± 0.24
FRAP (mg AAE/g): 13.88 ± 0.54/8.91 ± 0.18/5.70 ± 0.05
[72]
D. nipponica: rhizomeMeOH-E DPPH (µg/mL): 371.64 ± 12.30[45]
D. nipponica: rhizomeH2O-soluble polysaccharideOH scavenging (%): 3.35–43.73 at 0.25–4 mg
O2 (%): 27.5–35.52 at 0.25 mg and 2 mg
[73,74]
D. nipponica: leavesEtOH (70%)-E(mg TE/g edw) DPPH: 415 ± 9; ABTS•+: 659 ± 4 [68]
D. opposita: rhizome,
flesh/peel
Hot H2O-EDPPH (IC50, μg/mL): 1008.62 ± 5.96/374.85 ± 6.78
OH (IC50, μg/mL): 1267.04 ± 5.13/744.25 ± 3.46
[75]
EtOH (80%)-EDPPH (IC50, μg/mL): 897.14 ± 4.73/415.74 ± 3.79
OH (IC50, μg/mL): 1155.00 ± 9.64/845.21 ± 14.66
D pentaphylla: leavesEA (80%)/MeOHDPPH (IC50, μg/mL): 135.12 ± 0.95/85.61 ± 0.64
NO (IC50, µg/mL): 195.78 ± 0.29/68.13 ± 0.26
[76]
D pentaphylla: tubersMeOH/Ac crude extractsDPPH (EC50, μg/mL): 82.07 ± 0.08/89.41 ± 0.39
Metal chelating (EC50, μg/mL): 81.47 ± 0.36/86.52 ± 0.55
[77]
D. japonica: tubersEtOH (70%)DPPH: at 0.1–10 mg/mL from 18.01 to 89.64%[78]
D. villosa: leavesH2O/MeOH-EDPPH (IC50 μg edw/mL): 21.36/40.24[79]
D. rotundata: flour/pasteH2O 1:10 (w/v)DPPH (IC50, mg/mL): 19.26 ± 2.4/19.56 ± 1.5
ABTS•+ (mmol TE/100 g): 0.79 ± 0.00/1.31 ± 0.08
Fe3+ RP (mmol AAE/100 g): 0.93 ± 0.58/1.45 ± 0.47
OH (%): 56.71 ± 1.51/63.72 ± 2.31
Fe2+ chelating (%): 8.33 ± 2.78/11.11 ± 5.56
[47]
D. opposita: rhizomes18 compounds from MeOH-EDPPH (EC50, µg/mL): 12.3 ± 0.2->100 (AA = 19.2)
O2 (EC50, µg/mL): 38.8 ± 1.3->100 (AA = 16.7)
[80]
D. opposita: tuber mucilageH2O-E(%) DPPH 38.2 ± 3.14/O2, 84.1 ± 6.57/OH, 79.4 ± 6.42;
1 mM AA: 94.7 ± 3.21/89.9 ± 5.31/16.1 ± 0.64
[81]
D. schimperianaMeOH (60%)ABTS•+ (MTE/100 gMF): 0.153 (yellow); 0.218 (with red dot); 0.151 (red fleshed)[82]
D. bulbiferaEtOH (70%)DPPH (%): 64.81 ± 2.80; ABTS (%): 72.44 ± 5.28[83]
D. polystachya DPPH (%): 77.09 ± 0.00; ABTS (%): 99.89 ± 1.60
D. batatas DPPH (%): 75.74 ± 0.94; ABTS (%): 83.66 ± 9.03
D. quinqueloba DPPH (%): 84.46 ± 0.41; ABTS (%): 95.56 ± 0.96
D. batatas: thermally treated yamMeOH (70%)/EtOH (70%)/CF: MeOH (2:1)DPPH (IC50, mg/mL): 0.56/0.52/0.43
ABTS•+ (IC50, mg/mL): 1.28/1.05/0.60
[84]
D. opposita: tuber mucilageAutolysis/hydrolysis (pepsin/trypsin/papain)O2- (all in % at 100 mg/mL): 60.2 ± 4.01/82.2 ± 5.95 (p)/56.0 ± 4.36 (t)/98.5 ± 3.54 (p)/52.6 ± 4.18
OH: 90.4 ± 5.25/91.2 ± 5.86 (p)/91.2 ± 5.50 (t)/91.6 ± 5.92 (p)/67.6 ± 4.34 DPPH: 75.2 ± 4.77/61.7 ± 4.03 (p)/87.1 ± 5.04 (t)/70.2 ± 4.89 (p)/87.6 ± 2.75 (all in mmol α-Toc)
[85]
D. batatas: yamMeOH-E and fractions/HX/EA/BuOH/H2ODPPH (IC50, µg/mL): 602.2 ± 71.92/510.6 ± 25.02/80.5 ± 12.37/263.0 ± 56.47/>1000[86]
Arial bulbilsMeOH-E and fractions/HX/EA/BuOH/H2ODPPH (IC50, µg/mL): 376.3 ± 32.18/180.9 ± 24.77/38.1 ± 5.82/161.4 ± 32.14/>1000
AA: 15.2 ± 2.96; BHT 18.6 ± 4.05; VitE 35.6 ± 5.12
[87]
D. alata: yamEtOH (50%)/hot H2O/H2O DPPH (mg α-Toc eq/g): 4.14 ± 0.01/3.71 ± 0.03/3.37 ± 0.04 (peel); 0.73 ± 0.07/0.72 ± 0.03/0.22 ± 0.03 (flesh)
Fe3+ RP (mg GAE/g): 41.3 ± 0.8/28.7 ± 0.4/30.6 ± 0.4 (peel); 0.58 ± 0.00/0.83 ± 0.02/0.86 ± 0.01 (flesh)
[88]
D. japonica: bulbilMeOH (80%)-E: CF/EA/BuOH/H2O Fr/AADPPH (µg): 200.8 ± 7.9: 38.8 ± 3.6/14.8 ± 0.6/75.4 ± 1.6/>1000/3.3
ABTS•+ (mg): 2.3 ± 0.2: 0.5 ± 0.02/0.13 ± 0.02/0.93 ± 0.1/9.4 ± 0.5/1.2 µg
[89]
D. batatas: bulbil DPPH (µg): 84.0 ± 2.6: 23.6 ± 2.0/9.2 ± 0.2/27.6 ± 0.8/>1000/3.3
ABTS•+ (mg): 0.9 ± 0.04: 0.3 ± 0.04/0.09 ± 0.04/0.42 ± 0.04/7.6 ± 0.4/1.2 µg
D. trifida: dried tubers ABTS•+ (µmol TE/100 g): 131.14 ± 5.49 to 174.52 ± 0.78 (depending on drying method)[90]
D. trifida: tubers (IC50, mg/mL) DPPH: 7.44; ABTS•+: 0.54; O2: 13.67[91]
Abbreviations: AA—ascorbic acid; AAE—ascorbic acid equivalent; Ac—acetone; ABTS•+—2,2′-azinobis-(3-ethylbenzothiazoline-6-sulfonate) scavenging capacity; AcA—acetic acid; AAE—ascorbic acid equivalent; BHT—butyl hydroxy toluene; BuOH—n-butanol; CF—chloroform; ChelA—chelating activity; CUPRAC—cupric reducing antioxidant capacity; DCM—dichloromethane; de—dried extract; DE—diethyl ether; DPPH—(2,2-diphenyl-1-picrylhydrazyl) scavenging capacity; dw—dry weight; E—extract; EA—ethyl acetate; edw—extract dry weight; eq—equivalent; EtOH—ethanol; Fe2+,Fe3+—ferric ion; fr—fraction; FRAP—ferric reducing antioxidant power (determined using ferric-tripyridyltriazine complex); Fe3+ RP—ferric reducing power (determined using potassium ferricyanide complex); GA—gallic acid; GAE—gallic acid equivalents; H2O—water; HX—hexane; IC50—effective concentration reducing 50% of the radicals present in the reaction; LPI—lipid peroxidation inhibition; MeOH—methanol; ND—not detected; NO2nitrogen dioxide radical; NO—nitric oxide scavenging activity; O2—superoxide anion scavenging activity; O2—superoxide radical-scavenging activity; OH—hydroxyl radical-scavenging activity; ORAC—oxygen radical absorbance capacity; pdw—plant dry weight; PE—petrol ether; Q—quercetin; SOD—superoxide dismutase; TE—Trolox (6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid) equivalent; α-Toc—α-tocopherol; 2,7-dOH-4,6-dMetOP—2,7-dihydroxy-4,6-dimethoxy phenanthrene; 6,7-dOH-2,4-dMetOP—6,7-dihydroxy-2,4-dimethoxy phenanthrene; 6-OH-2,4,7-tMetOP—6-hydroxy-2,4,7-trimethoxyphenanthrene (See Supplementary Materials abbreviations).
Table 3. Total content of various groups of compounds in Dioscorea spp.
Table 3. Total content of various groups of compounds in Dioscorea spp.
Species: Plant Part SolventsCharacteriticsRef.
D. alata: tubersMeOHTPC: male 12.21 ± 0.82; female: 17.53 ± 1.30
TFC: male 14.80 ± 0.69; female 9.17 ± 0.3
[21]
EtOH (20%) fr.TS (%): male: 0.48 ± 0.06; female: 0.93 ± 0.17
D. alata: soaked flour of tubersH2O + AcA (5%): in H2O/NaHSO3 0.2%)/AA (0.1%)TPC: 11.6 ± 0.31/12.7 ± 0.67/11.5 ± 0.13
TFC: 7.0 ± 0.14/7.4 ± 0.21/7.3 ± 0.23
TAC (mg CGE/100 g): 46.7 ± 3.35/74.3 ± 3.83/50.0 ± 2.26
[49]
D. alata: tubersH2OTPC: 1.00–3.85; TFC: 0.60–1.60; TAC: 0.10–0.90[93]
D. alata: yamEtOH (50%)/hot H2O/H2O TPC (peel): 11.14 ± 0.30/6.09 ± 0.08/6.60 ± 0.13
TPC (flesh): 0.25 ± 0.01/0.40 ± 0.02/0.24 ± 0.00
[88]
D. alata: tubersEtOH (80%) TPC (mg GAE/g edw): 63.85 ± 1.83; TFC (mg RE/g edw): 8.21 ± 0.02; TFL (mg QE/g edw): 17.23 ± 0.19[94]
D. alata: purple yamEffect of processingTAC (mg/100 g): 38.12 (raw), 36.73 (blanched), 32.63 (washed), 29.29 (dried), 27.27 (flour)[48]
D. glabra: tubers TPC (mg/100 g): 335.64 ± 3.92; TFC: 65.73; C: 23.49 ± 0.0413; H2O-soluble B: 0.036 to 4.159[46]
D. alata: tubersMeOH (50%) + 0.1% HClTAC: 3.32 mg/g[95]
D. alata: tubersMeOH (80%) TPC (mg GAE/g edw): flesh 48.3 ± 4.1; peel 194.8 ± 14.6[52]
EA frTPC (mg GAE/g edw): flesh 479.5 ± 33.1; peel 695.1 ± 35.1
D. alata: tubersMeOH TPC (mg GAE/g edw): 222.99; TFC (mg QE/g edw): 98.95[50]
D. batatas: tubers;
thermally treated yam
EtOH (70%)/MeOH (70%)/
CHCl3:MeOH (2:1)
TPC (mg CE/g edw): 43.38 ± 0.66/37.62 ± 0.88/67.17 ± 0.12[55]
D. batatas: thermally treated TS (mg/g dw): 42.52 ± 1.88
TT (mg CE/g dw): 14.95 ± 0.98
[56]
D. batatas: raw yamBuOH/EATPC (mg CE/g edw): 78.68 ± 0.58/111.88 ± 0.66[57]
D. batatas: tubers thermally treatedBuOH/EATPC (mg GE/g): 53.83 ± 1.00/51.63 ± 2.63[59]
D. batatas: aerial bulbilsMeOH-E: and fr HX/EA/BuOH/H2OTPC (mg/g): 60.60: 12.63/27.48/17.18/4.03
TFC (mg/g): 16.4: 32.1/70.1/34.2/1.38
[87]
D. batatas: thermally treated yamMeOH (70%)/EtOH (70%)/CF:MeOH (2:1)TPC: 63.63 ± 0.33/69.47 ± 1.00/97.49 ± 0.66[84]
D. batatas: rhizomeMeOH: Jang-Ma/Dang-MaTPC (mg/g): 34.86 ± 0.15/45.84 ± 0.34; TFC (mg/g): 6.67 ± 0.22/7.33 ± 0.14; Total sugar (mg/g): 281.96 ± 0.08/140.86 ± 0.21[45]
D. alata: rhizomeMeOH: Dungkun-Daema/Jasak-MaTPC (mg/g): 87.05 ± 0.0.11/27.98 ± 0.25; TFC (mg/g): 12.67 ± 0.34/7.75 ± 0.23; Total sugar (mg/g): 184.98 ± 0.14/107.61 ± 0.32[45]
D. batatas: yamMeOH-E: and fr HX/EA/BuOH/H2OTPC (mg/g): 5.05: 2.61/48.31/8.49/3.81
TFC (mg/g): 4.85: 4.85/42.55/1.71/0.66
[86]
D. hispida:MeOH-E: fr PE/CTC/DCM/H2OTPC: 160.65 ± 0.18: 280.09 ± 0.54/287.50 ± 0.71/68.98 ± 1.43/22.99 ± 0.54[23]
D. bulbifera: stem tuberMeOH (80%)TPC (mg GAE/mg edw): 0.243 ± 0.052; nontannins, 0.632 ± 0.048; tannins, 0.259 ± 0.034.
TFL (mg QE/mg edw) 1.399 ± 0.075; TFC, 0.060 ± 0.025
[61]
D. bulbifera: rhizomeMeOH: Buchae-Ma TPC (mg/g): 51.11 ± 0.16; TFC (mg/g): 10.33 ± 0.09; total sugar (mg/g): 179.79 ± 0.14[45]
D. nipponica: rhizomeMeOH: Dungkun-MaTPC (mg/g): 52.08 ± 0.24; TFC (mg/g): 13.99 ± 0.11; total sugar (mg/g): 147.67 ± 0.09[45]
D. birmanica: rhizomeEtOH (95%). TPC (mg GAE/g e): 170.85 ± 3.02
TFC (mg CE/g e): 132.55 ± 3.59
[16]
D. birmanica: rhizomeEtOH (95%).TPC (mg GAE/g e): 170.85 ± 3.02
TFC (mg CE/g e): 132.55 ± 3.59
[16]
D. bulbifera: tubersMeOH TPC (mg/100 g FW): 67.17 ± 0.12[96]
D. bulbifera: tubersSoaked in 0–10% oligosaccharide solutionTPC (mg GAE/g): 1.37 ± 0.3–1.41 ± 0.1
TFC (mg RE/g): 0.97 ± 0.1–1.04 ± 0.1
[66]
D. bulbifera: bulbPE/EA/MeOH (sequentially)/EtOH (70%) TPC (mg/mL): 49.22 ± 0.80/98.00 ± 1.17/145.4 ± 3.29/85.89 ± 1.16
TFC (mg/mL): 4.95 ± 0.1/27.86 ± 0.18/12.76 ± 0.48/12.10 ± 0.05
[64]
D. hamiltonii: tuberMeOH TPC: male 41.40 ± 2.94; female: 50.70 ± 2.49 [21]
TFC: male: 25.67 ± 0.93; female: 36.67 ± 0.99
EtOH (20%) and its fractionTS (%): male 0.95 ± 0.14; female 1.16 ± 0.18
D. hamiltonii (syn D. persimilis): herbsMeOHTPC (µg/mL GAE): 158.21; TFC (μg/mL CE): 72.3 [29]
Vanillin-CH3COOH and HClO4 mixture 1:5 (v/v)TS (dioscin equivalents): 257.8 μg/mL
D. hispidaMeOH-E: fr PE/CTC/DCM/H2OTPC: 160.65 ± 0.18: 280.09 ± 0.54/287.50 ± 0.71/68.98 ± 1.43/22.99 ± 0.54[23]
D. oppositifolia: tubersMeOHTPC: male: 11.03 ± 0.60; female: 13.65 ± 0.36
TFC: male: 7.21 ± 0.99; female: 15.03 ± 1.08
[21]
EtOH (20%) and its fractionTS (%): male: 0.45 ± 0.09; female: 0.81 ± 0.15
D. opposita: tuber mucilageAutolysis/hydrolysisTPC (mg/g powder): 6.4 ± 0.08/15.3 ± 1.60 (pepsin)/11.2 ± 1.34 (trypsin)/7.4 ± 0.09 (papain)[85]
D. pubera: tubersMeOHTPC (mg GAE/gm dw): male: 31.76 ± 0.21; female: 21.83 ± 2.5 [21]
EtOH (20%) and its fractionTFC (mg CAE/gm): male: 19.68 ± 1; female: 22.17 ± 0.2
TS (%): male: 0.88 ± 0.23; female: 0.92 ± 0.17
D. oppositifolia (syn D. opposita): herbMeOH-ETPC (µg/mL GAE): 297.03; TFC (μg/mL CE) 49.6[29]
Vanillin-CH3COOH and HClO4 mixture 1:5 (v/v)TS (μg/mL dioscin equivalents): 475.5
D. opposita: rhizomeHot H2O TPC: flesh 1.77 ± 0.67; peel:10.97 ± 0.21; TFC (mg rutin/g eq): flesh: 1.03 ± 0.15; peel 1.77 ± 0.07; TC (mg/g): flesh: 324.90 ± 0.82; peel:123.50 ± 0.80[75]
EtOH (80%) Flesh: TPC 7.77 ± 0.10; TFC (mg RE/g extract): 1.20 ± 0.10; TC (mg/g extract) 23.63 ± 0.45
Peel: TPC: 15.40 ± 0.10; TFC (mg RE/g extract): 2.62 ± 0.15; TC (mg/g extract) 17.60 ± 0.20
D. pentaphylla: leavesMeOH TPC (mgGAE/g): 213.89 ± 3.93; TFC (mg QE/g): 41.5 ± 2.12[76]
EATPC (mgGAE/g): 76.39 ± 3.54; TFC (mg QE/g): 147.5 ± 3.54
D. schimperiana: tubers flourEtOH (20%) and its fraction; pasta with 60% yam flourTPC: traditional process, 2.86 ± 0.02; modified process, 5.04 ± 0.03[11]
D. schimperiana:MeOH (60%) TPC (mg/100 g): 10 (yellow); 8 (with red dot); 8 (red fleshed)[82]
D. trifida: tubersCommonly used methodsTPC (mg GAE/100 g): 187.09–513.67 ± 9.49
Total carbohydrate (%): 81.75 ± 0.24
Total starch (%): 74.11 ± 0.55
[90]
EtOH (95%):HCl 85:15 (v/v)TAC (mg C-3-Glc/100 g): 159.11–281.10 ± 0.01
D. trifida: tubersDry powders TPC (mg GAE/100 g dw): 166.10 ± 1.52; TFC (mg QE/100 g dw) 27.63 ± 2.69; TT (mg GAL/100 g dw)
9.62 ± 0.084; TAC (mg C-3-Glc/100 g dw): 21.59 ± 1.47
[91]
D. wallichii: dried powderMeOH (male/female)TPC: 10.73 ± 0.25/9.73 ± 0.28
TFC (mg CE/gm): 20.6 ± 0.6/26.00 ± 2.14
[21]
D. quinquelobaH2O/MeOH/EtOH/
EA
TPC (mg/g): 10.16/10.48/14.67/9.91
TFC (mg/g): 7.58/9.91/10.58/16.02
[97]
D. rotundata: flour/pasteH2O 1:10 (w/v)TPC (mg GAE/g): 1.56 ± 0.04/1.34 ± 0.02
TFC (mg QE/g): 0.16 ± 0.01/0.08 ± 0.01
[47]
D. alata: flour/paste TPC (mg GAE/g): 1.38 ± 0.03/1.12 ± 0.02
TFC (mg QE/g): 0.18 ± 0.01/0.10 ± 0.02
D. japonica: tubersEtOH (70%)TPC: 35.15 mg GAE/100 g edw[78]
Dioscorea spp.: tubers, 5 cultivarsInoculated with 6 spp. of arbuscular mycorrhizal fungi;
flesh/peel
TPC (mg/kg): Tainung 1: 21.3-44.5/39.0-52.7; Tainung 2: 19.8–38.3/43.9–54.5; Ercih: 11.1–16.9/40.4–50.4[98]
TFC (mg/kg): Tainung 1: 4.1–8.6/9.9–15.1; Tainung 2: 4.4–6.9/9.6-10.4; Ercih: 4.5–5.6/7.8–10.9; Zigyuxieshu: 4.9–6.5/9.3–11.8
Tainung 5: 4.4–5.7/8.3–10.9
TAC (flesh/peel, mg/kg): Tainung 1, 2, Ercih: nd; Zigyuxieshu: 0.83–1.08/1.93–2.54; Tainung 5: 0.33–0.76/1.52–2.42
D. bulbiferaEtOH (70%)TPC: 2.23 ± 0.03; TFC (mg RE/g): 1.99 ± 0.17[83]
D. polystachya TPC: 3.65 ± 0.11; TFC (mg RE/g): 2.62 ± 0.20
D. batatas TPC: 2.25 ± 0.19; TFC (mg RE/g): 1.57 ± 0.06
D. quinqueloba TPC: 9.50 ± 0.38; TFC (mg RE/g): 1.30 ± 0.16
D. batatas: rawBuOH/EATPC (mg CAE/g-E): 78.68 ± 0.58/111.88 ± 0.66[57]
D. alata: flour of 5 cultivarsSmall/medium/large particle size fractions Phenols (%): 0.27–1.39/0.52–2.82/0.48–2.20
TAC (mg/100 g): nd-14.20/nd-15.27/2.25–13.07
Carotenoids (µg/100 g): nd-132.12/nd-129.8/nd-123.1
[99]
D. alata: tubers TPC (mg GAE/100 g): 157.7 ± 7.5; TFC (mg CE/100 g): 190.4 ± 10.9[100]
D. japonica: tubers TPC (mg GAE/100 g): 206.4 ± 12.8; TFC (mg CE/100 g): 178.2 ± 8.3[100]
D. bulbifera: tuber TPC (mg GAE/100 g FW): 166 ± 10[96]
D.versicolor: tuber TPC (mg GAE/100 g FW): 41 ± 5
D. deltoidea: tuber TPC (mg GAE/100 g FW): 15 ± 2
D. triphylla: tuber TPC (mg GAE/100 g FW): 13 ± 1
D. japonica: bulbilMeOH (80%): fr CF/EA/BuOH/H2OTPC: 2.2 ± 0.1: 11.5 ± 0.4/33.9 ± 1.8/3.9 ± 0.1/2.4 ± 0.1[89]
D. batatas: bulbil TPC: 3.9 ± 0.2: 19.6 ± 0.8/39.1 ± 2.2/7.4 ± 0.4/5.8 ± 0.2
D. hirtiflora: tubersSuccessively DCM/EA/MeOHTPC (mg GAE/g): 0.25 ± 0.01/8.9 ± 0.69/10.1 ± 0.35
TFC (mg QE/g): ND/24.2 ± 0.43/28.1 ± 0.35
[67]
D. dumetorum TPC (mg GAE/g): 1.75 ± 0.02/0.81 ± 0.003/1.04 ± 0.02
TFC (mg QE/g): 8.58 ± 0.14/12.4 ± 0.43/9.6 ± 0.21
D. bulbifera: mauve TPC (mg GAE/g): 1.91 ± 0.02/14.0 ± 0.41/5.99 ± 0.09
TFC (mg QE/g): 26.1 ± 0.29/74.4 ± 0.41/54.5 ± 0.73
D. bulbifera: yellow TPC (mg GAE/g): 1.0 ± 0.10/12.6 ± 0.34/0.99 ± 0.01
TFC (mg QE/g): 7.87 ± 1.10/52.0 ± 0.14/29.3 ± 0.02
Abbreviations: AA—ascorbic acid; AcA—acetic acid; BuOH—n-butanol; C—vitamin C; CE—catechin equivalents; CF—chloroform; CGE—cyanidin glucoside equivalents; C-3-Glc—cianidina-3-galactosıd; CTC—carbon tetrachloride; B—vitamin B; DCM—dichloromethane; dw—dry weight; E—extract; EA—ethyl acetate; edw—extract dry weight; eq—equivalent; EtOH—ethanol; fr—fraction; FW—fresh weight; GAE—gallic acid equivalents; HX—hexane; MeOH—methanol; NaHSO3—Na-bisulfide; pdw—plant dry weight; PE—petrol ether; pfw—plant fresh weight; Q—quercetin; QE—quercetin equivalents; RE—rutin equivalents; TAC—total anthocyanin content; TFC—total flavonoid content; TFL—total flavonol content; TPC—total phenol content; TS—total saponins; TT—total tanin; α-Toc—α-tocopherol.
Table 4. Bioactivities of Dioscorea species reported by the in vitro cell studies and in vivo animal studies.
Table 4. Bioactivities of Dioscorea species reported by the in vitro cell studies and in vivo animal studies.
Species PreparationStudy DesignResultsRef.
D. alata: tuberH2O-E in plantain and bitter leaf meal Rat model, 2.0 g dough meal food, consumed within 25 min. Blood glucose and GI ↓: the potential to be used as functional foods to alleviate postprandial hyperglycemia [10]
D. alata: tuberMeOH-E Antimicrobial activityEffective against the Gram-positive bacteria Streptococcus pneumoniae and fungi Candida albicans[21]
D. alata: freeze-dried powderContaining antho-cyaninsTNBS-inducted colitis mice; DACNs at 20, 40, and 80 mg/kg for 3 days, intra-rectallyThe levels of pro-inflammatory cytokines, TNF-α and IFN-γ ↓. May be applied as a potential food supplement in inflammatory bowel disease (IBD) therapy[95]
D. alata: tuberMeOH-EAntibacterial activity using disc diffusion assayEffective against the bacteria Staphylococcus epidermidis and Gram-negative bacteria: Shigella dysenteriae, Shigella flexneri[50]
D. alata: tuberMeOH (70%)-E Mice spleen lymphocytes cellsAnti-inflammatory effect by inhibition of the NO and TNF-α expression. [133]
D. alata: tuberH2O (WSP)Alloxan-induced hyperglycemic rats; 400 mg/kg bw/day, 4 weeks, orallyFasting blood glucose gradually decreased[134]
D. alata tuberEtOH and H2O Doxorubicin-induced cardiac damage mice; daily dose 30 mg/mL for 4 weeks; orallyRegulate NF-kB expression at the transcriptional level; cardiac levels of TBARS, ROS, inflammatory factors, the expression of NF-kB, blood pressure ↑; SOD, GPx activity ↑. [135]
D. alata: purple yamEtOH (80%)-E; EA fr of peel and fleshMethylglyoxal-induced HepG2 cellsExtracts strengthened antioxidant defense system [52]
Antiglycation activity in vitroCould inhibit the formation of dicarbonyl compounds in a dose-dependent manner.
D. batatas: peelEtOH (95%), DDP LPS-induced RAW 264.7 cell model2, 7-dihydroxy-4, 6-dimethoxy phenanthrene suppressed LPS-induced expression of cytosolic iNOS and COX-2. Could exert anti-inflammatory activity by suppressing NF-κB signaling pathway.[136]
D. batatas: rhizomeH2OSTZ-induced diabetic mice; at 500 or 1000 mg/kg 1/day for 4 weeks Glucose and leptin, total cholesterol, triglycerides, low-density lipoprotein cholesterol ↓.
Expression of antioxidant enzymes, and mitochondrial-induced biogenetic factors in the liver, pancreas, and muscle tissue ↑.
[137]
Allantoin at 20 or 50 mg/kg/day for 4 weeks; orally
D. batatas: flesh and peelEtOH (60% and 95% w/w); H2OEtOH-induced gastric ulcer in mice; a single dose 100 or 200 mg/kg bw, orallyExtracts dissolved in: 5% Tween-80, 10% polyethylene glycol, 10% DMSO, and 10% EtOH saline: inflammatory factors, NO and IL-6, in the serum; COX-2 expression in the gastric tissue ↓[138]
D. batatas: barkEtOH (BDB)Anti-inflammatory activity in LPS-induced RAW 264.7 cellsNO production (dose-dependent); iNOS protein induction; regulates inflammation by inhibiting the COX-2 pathway[139]
D. batatas: yamH2O-E; powderSTZ-induced diabetic rats; at 2500 or 1000 mg/kg daily doses for 1-month, orallyFasting blood glucose and HbAlc ↓; the serum antioxidant activities of tGSH, GSH and SOD ↑; lipid malondialdehyde (MDA), oxidized glutathione (GSSG) ↓[140]
D. birmanica: rhizomeEtOH-E SNP-induced oxidative stress in liver BNL CL.2 cellsElevated cell viability in a dose-dependent manner. Can ameliorate oxidative stress[16]
D. bulbifera: powder of bulbilsEtOH and H2O-E Cell culture model; 1–100 μg/mL ELow cytotoxic effect on the cells [62]
CF, EA, H2O frLPS-induced RAW macrophage 264.7 cellsMild anti-inflammatory activity
D. bulbifera: leavesMeOH-EMCF-7 and MDA-MB-231 breast cancer cells Cytotoxic effect in cell lines; prompts apoptosis at various stages and a significant decrease in viable cells[63]
D. cayennensis: tubersProtein conc. 64%Antibacterial activityNo inhibition of Salmonella sp. and Lysteria monocytogenes; effective against E. coli [141]
D. hamiltonii: tuberMeOH-E Antimicrobial activity Effective against Streptococcus pneumoniae[21]
D. hamiltonii (D. persimilis): herbMeOH-EXylene-induced ear edema damage mice; 2 and 6 g/kg, 5 days, orallyDecreased the level the inflammatory cytokines and reduced oxidative stress[29]
D. hemsleyi: poly-saccharidesWarm and hot H2O Anti-hyperglycemic activityEffectively inhibits α-amylase, α-glucosidase [72]
D. japonicaEtOH-E λ-carrageenan-induced paw edema mice; 0.5 and 1.0 g/kg, orallyMDA, NO, TNF-α ↓ after 5th hour; 1.0 g/kg decreased the developments of carr-induced paw edema after 5th hour [142]
Acute toxicity, at 10 g/kg No toxicity observed
LPS-induced RAW 264.7 cellsNo effects on viability; suppressed LPS-induced production of NO, TNF-α, expression of iNOS and COX-2
D. japonica: yam tubersEtOH and H2O-E Doxorubicin-induced cardiac damage in mice; a daily dose of 30 mg/mL (w/v) for 4 weeks; orallyMight regulate NF-kB expression at the transcriptional level; cardiac levels of TBARS, ROS, inflammatory factors, expression of NF-kB; blood pressure ↓; SOD, GPX activity ↑[135]
Monascus-fermented D. (red mold D. (RMD))
root
EtOH (95% w/w) dissolved in mineral oilDMBA-induced hamster buccal pouch carcinogenesis; RMD extracts (50, 100, 200 mg/kg bw, paint for 14 weeks on days alternate to DMBA painting) Anti-inflammatory and antioxidative activity. Inhibit the pro-inflammatory cytokines TNF-R, IL-1β, IL-6, and IFN-γ, which in turn, leads to oxidative stress. [143]
D. nipponicaEtOH (70%)-E Anti-osteosarcoma activity Induced apoptosis in human osteosarcoma cells line U2OS[144]
D. nipponica: rhizomeSaponins MSU-inducted gouty arthritis mice; TS at 100, 300, 900 mg/kg every 24 h for 7 days, orally TS (dioscin, protodioscin, pseudo protodioscin) might restore production of pro-inflammatory cytokines TNF-α, pro-interleukins IL-1β and IL-8 to the normal conditions, regulating antioxidant capacities and NALP3 inflammasome. [145]
D. nipponica: rhizomeEtOH (80%)-E and diosgenin in 1% carboxyl methyl-cellulose ISO-induced myocardial ischemia model in rats; diosgenin at 20, 40, 80 mg/kg for 3 days.
500 mg/kg for 3 days, orally, after ISO injection
Diosgenin protects the myocardium against ischemic insult through increasing enzymatic and nonenzymatic antioxidant levels; decreasing oxidative stress damage;
SOD, CAT, GPx activity ↑; lipid peroxidation ↓
Confirms hypothesis that intestinal bacteria produce diosgenin from D. nipponica extract.
[24]
Dry precipate of saponins ISO-induced myocardial ischemia model in rats; 150 and 300 mg/kg for 3 days, orally, both before and after ISO injectionSOD, CAT, GPx, total antioxidant capacity (T-AOC) activity ↑; can protect the myocardium against ischemic insult [146]
D. nipponica: rhizome Dry precipitate of saponinsRat model; single dose of 160 mg/kg intragastricallyDiosgenin was one of the main metabolites found in plasma and feces. The extract can play an essential role in cardioprotective efficacy.[146]
D. nipponicaCrude drug with saponins Potassium oxonate-induced hyperuricemic mice; 60, 300, 600 mg/kg every 24 h for 6 days, before induction Total saponins (dioscin, protodioscin, pseudo protodioscin) from RDN had uricosuric effect and could enhance urate excretion and reduce the serum urate levels [14]
D. nipponica:
rhizome
MeOH-EAntibacterial activityEffective against Bacillus subtilis, Staphylococcus aureus, Proteus vulgaris, Salmonella typhimurium[45]
D. opposita: yam Fresh-cut,
MeOH → yellow powder
OH-induced DNA damage Can protect against DNA damage (IC50 0.098 ± 0.032 mg/mL); provides a theoretical basis for the application of YP in food and drug industry.[70]
D. oppositifoliaMeOH-EXylene-induced ear edema damage mice; 2 and 6 g/kg, for 5 days; orallyThe inflammatory cytokines, TNF-α, IL-6; oxidative stress ↓[29]
D. oppositifolia: tubersMeOH-E Antimicrobial activityEffective against Klebsiella pneumoniae, Shigella dysenteriae, Candida albicans, Candida tropicalis[21]
D. opposita:
Chinese yam
Cold-soaking extract (CYCSE) HCT-induced rat; CYCSE 60 mg/kg and 80 mg/kg for 10 days
H2O2-induced Leydig cells (TM3)
Can stimulate the NO/cGMP pathway and protect against induced erectile dysfunction; may protect testis morphology, increase TM3 cell proliferation and stimulate testosterone secretion. Suppressed TGF-β1 in injured cells.
Can protect against damage from the oxidative stress response.
[147]
D. panthaica:
rhizome
Precipate of saponins DP-EISO-induced myocardial ischemia model in rats; 150 and 300 mg/kg, 3 days; orallySOD, CAT GPx, total antioxidant capacity (T-AOC) activity ↑; can protect the myocardium against ischemic insult [146]
D. purpurea: tuberEtOH and H2O Doxorubicin-induced cardiac damage in mice; a daily dose of 30 mg/mL (w/v) for 4 weeks; orallySOD and GPx activity ↑; might regulate NF-kB expression at the transcriptional level; blood pressure, the cardiac levels of TBARS, ROS, and inflammatory factors, the expression of NF kappa B↓. [135]
D. pentaphylla: tuber MeOHAntibacterial activity using disc diffusion assayEffective against Streptococcus mutans, Streptococcus pyogenes, Vibrio cholerae, Shigella flexneri, Salmonella typhi. [77]
D. pubera BlumeMeOHAntimicrobial activity Effective against Streptococcus pneumoniae, Klebsiella pneumoniae, Escherichia coli, Shigella dysenteriae, Candida albicans, Candida tropicalis [21]
D. wallichiiMeOHAntimicrobial activityEffective against Klebsiella pneumoniae, Shigella dysenteriae and fungus Candida tropicalis [21]
D. zingiberensis: rhizome Total steroid saponins (TSS)Adjuvant-induced arthritis (AIA) rat; 50, 100, and 200 mg/kg 1/day, every 3 days, respectively, from day 0 to day 28, orally The levels of pro-inflammatory cytokines IL-1, IL-1β, IL-6, IL-10, and TNF-α ↓; suppressed production of oxidant stress makers: NO, MDA; could protect an injured ankle joint from further deterioration. [19]
LPS-induced RAW
264.7 macrophage cells
TSS suppresses NF-κB activation by inhibiting the phosphorylation of p65 and IκBα
D. villosa: leaf MeOHMouse fibroblast L929 cell lineNo cytotoxic effect. Scratch assay: expression of Collagen-1; induction of migration of fibroblasts to the wound site ↑.[104]
Abbreviations: AA—ascorbic acid; AIA—adjuvant-induced arthritis; BDB—bark of D. batatas DECNE; cGMP—cyclic 3′,5′-monophosphate; COX-2—cyclooxygenase-2; CYCSE—Chinese yam cold-soaking extract; DACNs—anthocyanins; DP-E—D. panthaica extract; DMBA—7,12-dimethylbenz-[a]anthracene; DMSO—Dimethyl sulfoxide; E—extract; EA—ethyl acetate; EtOH—ethanol; GI—glycemic index; GSH—reduced glutathione; H2O2—hydrogen peroxide; HbAlc—glycated hemoglobin; HCT—hydrocortisone; IBD—inflammatory bowel disease; IFN-γ—interferon-gamma; (IL)-8—interleukin; iNOS—inducible nitric oxide synthase; ISO— isoprenaline; LPS—lipopolysaccharide; MDA—malondialdehyde; MeOH—methanol; NF kappa B—nuclear factor kappa B; NF-κB—nuclear factor-κB; NO—nitric oxide; Nrf2—nuclear factor-erythroid 2-related factor 2; PGE2—prostaglandin E2; RDN—rhizoma D. nipponica; RMD—red mold dioscorea; ROS—reactive oxygen species; SNP—sodium nitroprusside; SOD—superoxide dismutase; STZ—streptozotocin; TBARS—thiobarbituric acid-reacting substances; TGF-β1—transforming growth factor-β1; tGSH—total glutathione; TNBS—trinitrobenzenesulfonic acid; TNF-a—tumor necrosis factor-a; TS—total saponins; TSS—total steroid saponins; YP—yellow powder.
Table 5. Phytochemicals in Dioscorea spp.
Table 5. Phytochemicals in Dioscorea spp.
Species: Plant Part SolventsCompoundsRef.
D. alata: tubers MeOH Vitamins (mg/g): male: C, 13.49 ± 3.64; B1, 1.14 ± 0.16; B2, 1.75 ± 0.26; female: C, 18.26 ± 1.37; B1, 1.26 ± 0.11; B2, 1.87 ± 0.2[21]
D. alata: tubers EtOH (50%) with 0.1% HClAnthocyanins: alatanin C (cyanidin 3-(6-sinapoyl gentiobioside); cyanidin-3-diglucoside; cyanidin-3,5-diglucoside; alatanins B, C; alatanin E, D, F isomers[93]
D. alata (purple yam): dried tubers EtOH (80%) Phenolic acids (mg/100 g mdw): galic 0.482 ± 0.057; 4-hydroxy benzoic 0.192 ± 0.0024; syringic 0.899 ± 0.0022; sinapic 0.202 ± 0.0501; chlorogenic 0.451 ± 0.0038; ferulic 0.089 ± 0.0005. Flavonoids (mg/100 g mdw): quercetin 0.687 ± 0.0030, apigenin 0.210 ± 0.0041; kaempferol 9.219 ± 0.0043[94]
D. glabra: tubers mg/100 gm: C, 23.49 ± 0.0413; H2O-soluble B, 0.036 to 4.159 [46]
D. alata (purple yam):
freeze-dried tubers
MeOH (50%) with 0.1% HClAnthocyanins (% peak area): cyanidin-3,5-diglucoside (31.22); cyanidin-3-diglucoside-5-celery glycosides (28.77); delphinidin-3-glucose-5-rutinoside (16.36); delphinidin-3-glucoside (12.18), delphinidin-3,5-diglucoside (11.31) [95]
D. alata: freeze-dried tubersMeOH (70%)Phenolic acids (mg/g dw): galic 29.34; 4-hydroxy benzoic 6.48; syringic 2.94; p-coumaric 2.53; myricetin 42.39[133]
D. alata: tubersMeOHMyricetin, gallic acid, ellagic acid, vanillic acid, syringic acid, epicatechin, vanillin, p-coumaric acid, trans-cinnamic acid and kaempferol.[50]
D. batatas: freeze-dried flesh and peel of tubersEtOH (95%) Phenanthrenes (mg/100 g dw): peel: 2,7-dihydroxy-4,6-dimethoxy 47.35 ± 0.25; 6,7-dihydroxy-2,4-dimethoxy 29.29 ± 0.08; 6-hydroxy-2,4,7-trimethoxy (batatasin I) 35.85 ± 0.12 [54]
D. batatas: yam Thermally treated mealsVitamins (mg/100 g): E, 8.3; C, 3.5; B1, 2.1, B2, 0.03[84]
D. bulbifera: flesh and peel of bulbils/tubers MeOHPhenolic acids (µg/g dw). Flesh: gallic 1.69 ± 0.13, isovanillic 1.02 ± 0.06, protocatechuic 0.15 ± 0.01. Peel: gallic 2.30 ± 0.20, isovanillic 0.18 ± 0.02, protocatechuic 0.10 ± 0.02
Flavonoids (μg/g dw). Flesh: catechin: 46.1 ± 0.75, quercetin 0.08 ± 0.01. Peel: catechin: 8.50 ± 1.01, quercetin 0.27 ± 0.05
Ascorbic acid (µg/g dw). Flesh: 26.4 ± 0.51. Peel: 25.03 ± 3.82
[107]
EAPhenolic acids (µg/g dw). Flesh: gallic 0.32 ± 0.14, isovanillic 1.71 ± 0.05, protocatechuic 0.13 ± 0.007. Peel: gallic 0.27 ± 0.03, isovanillic 0.22 ± 0.09, protocatechuic 0.20 ± 0.07
Flavonoids (μg/g dw). Flesh: catechin 108.3 ± 0.69, quercetin 0.99 ± 0.05. Peel: catechin 23.1 ± 0.22, quercetin 1.36 ± 0.16
Ascorbic acid (µg/g dw). Flesh: 4.52 ± 1.18. Peel: 2.8 ± 0.09
D. japonica: leaves DE extract fraction Total triterpenoids (including esters) (mg/g d.w.): Tokyo 734.71; Kanagawa 716.55[169]
D. caucasica: leaves DETotal of triterpenoids (including esters) (mg/g dw): 1492.56 [169]
D. hispida: leaves DE extract fractionTotal of triterpenoids (including esters) (mg/g dw): 704.11[169]
D. quinquelobata: leaves DE extract fraction Total of triterpenoids (including esters) (mg/g dw): 467.29[169]
D. purpurea: leaves DE extract fractionTotal of triterpenoids (including esters) (mg/g dw): 628.54[169]
D. nipponica: leaves DE extract fractionTotal of triterpenoids (including esters) (mg/g dw): 837.83[169]
D. hamiltonii: dried powder of tubersMeOH Vitamins (mg/g): male: ascorbic acid 10.31 ± 2.75, thiamine 1.15 ± 0.09, riboflavin 0.82 ± 0.07; female: ascorbic acid 12.7 ± 3.64, thiamine 1.03 ± 0.16, riboflavin 0.98 ± 0.12[21]
D. hamiltonii (syn D. persimilis): dry herb powderMeOH; HPLCPhenolic acids (µg/g d.w): gallic 6.24 ± 0.07; protocatechuic 0.65 ± 0.02; chlorogenic 0.93 ± 0.03; syringic 26.26 ± 0.42; p-coumaric 0.96 ± 0.05
Flavonoids (µg/g dw): catechin 17.69 ± 0.03; rutin 7.07 ± 0.22; quercetol 6.8 ± 0.17; kaempferol 5.92 ± 0.13.
Saponin content (µg/g dw): protogracillin 60.21 ± 1.04; dioscin 18.21 ± 0.54; diosgenin 25.00 ± 0.08; trillin 107.08 ± 1.12
[29]
D. hirtiflora: flesh/peelMeOHPhenolic acids (µg/g dw): gallic 0.34 ± 0.00/0.73 ± 0.35, isovanillic 0.28 ± 0.04/0.30 ± 0.06, protocatechuic 0.13 ± 0.02/0.13 ± 0.03.
Flavonoids (μg/g dw): catechin 6.91 ± 0.21/4.00 ± 0.23, quercetin 0.47 ± 0.14/0.28 ± 0.009
Ascorbic acid (µg/g d.w): 5.88 ± 0.57/12.0 ± 0.61
[107]
EAPhenolic acids (µg/g dw): gallic 0.19 ± 0.03/0.24 ± 0.04, isovanillic 0.88 ± 0.03/0.93 ± 0.09, protocatechuic 0.42 ± 0.02/0.20 ± 0.02.
Flavonoids (μg/g d.w): catechin 23.7 ± 0.42/5.19 ± 0.50, quercetin 0.42 ± 0.09/1.57 ± 0.26.
Ascorbic acid (µg/g d.w): 4.26 ± 0.39/2.72 ± 0.22
D. nipponica: rhizomesNADES containing 30% H2O Steroidal saponins (%): protodioscin 79.90, protogracillin 68.12, pseudoprotodioscin 67.27, pseudoprotogracillin 74.8 [170]
D. nipponica: freeze-dried rhizomesEtOH (70%)Saponins (mg/g): protodioscin 159.983 ± 0.064; protogracillin 4.250 ± 0.024; pseudoprotodioscin 13.821 ± 0.037; dioscin 22.999 ± 0.121[144]
D. opposita: rhizome Hot H2O Phenolic acids (μg/g): gallic 3.56 ± 0.13; chlorogenic 6.77 ± 0.06; vanillic 8.49 ± 0.36; syringic 2.95 ± 0.14; p-coumaric 16.90 ± 0.17.
Flavonoids (μg/g): epicatechin 7.37 ± 0.24; phlorizin 18.90 ± 0.48
[75]
EtOH (80%)Phenolic acids (μg/g): gallic 3.10 ± 0.4; chlorogenic 7.92 ± 0.42; vanillic 12.59 ± 0.51; syringic 6.78 ± 0.46; p-coumaric 16.39 ± 0.37
Flavonoids (μg/g): rutin 9.48 ± 0.40; epicatechin 28.39 ± 0.57; phlorizin 20.29 ± 0.34
D. oppositifolia.: stems, leaves AC (50%) → DCM Norsesquiterpenoids: dioscopposin A, dioscopposin B[151]
D. oppositifolia (syn D. opposita) (Chinese yam): herbMeOHPhenolic acids (μg/g dw): gallic 3.67 ± 0.10; protocatechuic 0.69 ± 0.02; chlorogenic 1.20 ± 0.04; vanillic 2.08 ± 0.05; syringic 37.35 ± 0.49; p-coumaric 1.65 ± 0.04.
Flavonoids (μg/g dw): rutin 11.98 ± 0.16; quercetol 27.76 ± 0.12; kaempferol 18.65 ± 0.08; catechin 4.45 ± 0.07.
Saponins (μg/g dw): protogracillin 154.45 ± 2.56; dioscin 23.64 ± 0.27; diosgenin 26.02 ± 0.05; trillin 77.61 ± 0.10
[29]
D. quinquelobata:
rhizomes
EtOH (70%)Steroidal saponins (mg/g): protodioscin 3.496 ± 0.018, protogracillin 5.945 ± 0.020, pseudoprotodioscin ND.,
dioscin 10.002 ± 0.051, gracillin 9.011 ± 0.098
[144]
D. pentaphylla: leaves MeOHGallic acid, rutin, quercetin[76]
D. polystachya; tubers MeOH (70%)Dehydroepiandrosterone, allantoin, 5-hydroxy-7-methoxyflavanone, arnebinone, dioscin, protodioscin[171]
D. pubera: tubers MeOHVitamin content (mg/g): male: ascorbic acid 14.29 ± 2.38, thiamine 0.85 ± 0.07, riboflavin 1.02 ± 0.08; female: ascorbic acid 15.88 ± 1.37, thiamine 0.99 ± 0.11, riboflavin 0.94 ± 0.14[21]
D. septemloba: rhizomesEtOH (75%)Phenanthropyran: dioscorone B, phenanthrene: 2,2′,6,6′-tetramethoxy-4,4′7,7′-tetrahydroxy-1,1′-biphenanthrenes[172]
D. septemloba: rhizomesEtOH (70%)Steroidal saponins (mg/g): protodioscin 8.959 ± 0.014, protogracillin 9.902 ± 0.061, pseudoprotodioscin ND, dioscin 9.822 ± 0.014, gracillin 7.123 ± 0.031[144]
D. wallichii: dried powderMeOHVitamins (mg/g): male: ascorbic acid 9.52 ± 2.38, thiamine 1.25 ± 0.13, riboflavin 1.18 ± 0.1; female: ascorbic acid 12.7 ± 1.4, thiamine 1.11 ± 0.12, riboflavin 1.13 ± 0.24 [21]
D. bulbifera: rhizomesPurified from EtOH (80%) E with EA, Sephadex LH-20, and ODSC22 ω-hydroxy fatty acid, 3-hydroxy-5-methoxybenzoic acid, various phenanthrene derivatives and flavonoids[173]
D. trifida: yam tubers Pelargonidin, cyanidin, peonidin glycosides and other derivatives[113]
D. opposita Aromatic benzyl compounds, dihydrostilbenes, phenanthrenes: diarylheptanoids, apigenin[80]
D. opposita: aerial partsEtOH6,7-dihydroxy-2-methoxy-1,4-phenanthrenedione; chrysoeriol 4′-O-â-D-glucopyranoside, chrysoeriol 7-O-â-D-glucopyranoside, alternanthin, daucosterol [174]
D. communis Herorensol, 2,3,4-trimethoxy-7,8-methylenedioxyphenanthrene, 2,4-dimethoxy-7,8-methylendioxy-3-phenanthrenol, chrysotoxene, 2,4,8-trimethoxy-3,7-phenanthrenediol, orchinol, and lusianthridin 7[31]
D. schimperiana: yellow-fleshed/yellow with red dot/red-fleshed MeOH (60%)µg/100 g: α-Toc 538.66/275.11/554.86; lutein: 18.06/16.35/18.22; zeaxanthin: 11.62/6.70/11.68; β-kryptoxanthin: 2.37/6.12/2.39; β-carotene: 212.23/197.04/212.98; β-carotene: 560.94/462.30/562.91; lycopene: 0.83/0.84/0.84;
Pro-vit A carotenoids: 787.15/672.16/789.95
[82]
D. batatas: thermally treated (mg/100 g dw) Chlorophyll a/b: 0.43 ± 0.01/0.75 ± 0.02; lycopene 0.30 ± 0.00; phytic acid 1.04 ± 0.42[56]
Abbreviations: AC—acetone; C—vitamin C; DCM—dichloromethane; DE—diethyl ether; dw—dry weight; EA—ethyl acetate; EtOH—ethanol; FW—fresh weight; HCl—hydrochloric acid; MeOH—methanol; NADES—natural deep eutectic solvent; ND—not detected; α-Toc—α-tocopherol.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Adomėnienė, A.; Venskutonis, P.R. Dioscorea spp.: Comprehensive Review of Antioxidant Properties and Their Relation to Phytochemicals and Health Benefits. Molecules 2022, 27, 2530. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules27082530

AMA Style

Adomėnienė A, Venskutonis PR. Dioscorea spp.: Comprehensive Review of Antioxidant Properties and Their Relation to Phytochemicals and Health Benefits. Molecules. 2022; 27(8):2530. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules27082530

Chicago/Turabian Style

Adomėnienė, Aušra, and Petras Rimantas Venskutonis. 2022. "Dioscorea spp.: Comprehensive Review of Antioxidant Properties and Their Relation to Phytochemicals and Health Benefits" Molecules 27, no. 8: 2530. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules27082530

Article Metrics

Back to TopTop