Next Article in Journal
Rational Fabrication of Defect-Rich and Hierarchically Porous Fe-N-C Nanosheets as Highly Efficient Oxygen Reduction Electrocatalysts for Zinc-Air Battery
Next Article in Special Issue
Stilbenes, a Versatile Class of Natural Metabolites for Inflammation—An Overview
Previous Article in Journal
Enhanced Solid-State Fluorescence of Flavin Derivatives by Incorporation in the Metal-Organic Frameworks MIL-53(Al) and MOF-5
Previous Article in Special Issue
Study on the Cellular Anti-Inflammatory Effect of Torularhodin Produced by Sporidiobolus pararoseus ZQHL Isolated from Vinegar Fungus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Dioscorea spp.: Bioactive Compounds and Potential for the Treatment of Inflammatory and Metabolic Diseases

1
Key Laboratory of Molecular Pharmacology and Drug Evaluation, School of Pharmacy, Yantai University, Yantai 264005, China
2
Physiology Group, Department of Basic and Applied Medical Sciences, Ghent University, 9000 Ghent, Belgium
3
School of Pharmacy, Binzhou Medical University, Yantai 264003, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2023, 28(6), 2878; https://doi.org/10.3390/molecules28062878
Submission received: 27 February 2023 / Revised: 18 March 2023 / Accepted: 21 March 2023 / Published: 22 March 2023

Abstract

:
Dioscorea spp. belongs to the Dioscoreaceae family, known as “yams”, and contains approximately 600 species with a wide distribution. It is a major food source for millions of people in tropical and subtropical regions. Dioscorea has great medicinal and therapeutic capabilities and is a potential source of bioactive substances for the prevention and treatment of many diseases. In recent years, increasing attention has been paid to the phytochemicals of Dioscorea, such as steroidal saponins, polyphenols, allantoin, and, in particular, polysaccharides and diosgenin. These bioactive compounds possess anti-inflammatory activity and are protective against a variety of inflammatory diseases, such as enteritis, arthritis, dermatitis, acute pancreatitis, and neuroinflammation. In addition, they play an important role in the prevention and treatment of metabolic diseases, including obesity, dyslipidemia, diabetes, and non-alcoholic fatty liver disease. Their mechanisms of action are related to the modulation of a number of key signaling pathways and molecular targets. This review mainly summarizes recent studies on the bioactive compounds of Dioscorea and its treatment of inflammatory and metabolic diseases, and highlights the underlying molecular mechanisms. In conclusion, Dioscorea is a promising source of bioactive components and has the potential to develop novel natural bioactive compounds for the prevention and treatment of inflammatory and metabolic diseases.

1. Introduction

Nowadays, metabolic diseases, a non-communicable disease, are a major health hazard in the modern world and have become a global problem [1]. The epidemic of metabolic disorders, such as obesity, type 2 diabetes mellitus (T2DM), non-alcoholic fatty liver disease (NAFLD), and cardiovascular diseases, poses a great challenge to global human health [2]. Chronic inflammation plays a key role in the initiation, propagation, and progression of metabolic disorders [3]. Therefore, it is necessary to find suitable compounds to control inflammation, reduce metabolic disorders, and thus prevent the progression of related diseases [4]. Due to the limitations of modern drug therapy, such as gastrointestinal discomfort, hypoglycemia, liver and kidney damage, there is an increasing emphasis on finding effective medicine from natural plants (e.g., Coptis chinensis, Embelia ribes, Cleome) to reduce oxidative stress and treat inflammation, diabetes, cardiovascular diseases, and metabolic disorders [5,6,7,8,9].
Dioscorea is the largest genus in the Dioscoreaceae family, with about 600 species, mainly distributed in tropical and subtropical regions of Southeast Asia, Africa, and Central and South America. Dioscorea species are commonly known as yam worldwide, mostly with underground tubers or rhizomes, and are a major food source for millions of people in tropical and subtropical regions [10,11]. It is considered the fourth most important tuber crop after potato, cassava, and sweet potato, contributing about 10% of the world’s root and tuber production [12]. In addition to their importance as traditional starchy staples, various species of Dioscorea have been domesticated and widely cultivated for nutritional and medicinal purposes [13]. Dioscorea tuber contains secondary metabolites that provide additional health benefits [14]. In fact, Dioscorea has good ethnobotanical value. In different ethnic communities and geographical regions, different species of Dioscorea are used by local people to treat health problems, such as hemorrhoids, coughs, diabetes, rheumatism, stomach pains, diarrhea, and skin infections [15,16].
Dioscorea species contain a large number of bioactive compounds, such as phenols, flavonoids, saponins, anthocyanins, carotenoids, allantoins, and water-soluble polysaccharides [12,17]. Modern research has confirmed that Dioscorea has a variety of pharmacological activities, such as improving the cardiovascular system, and regulating immune function, as well as anti-tumour, anti-bacterial, anti-inflammatory, and anti-diabetic activities [18,19,20]. Phytochemicals derived from Dioscorea, such as polysaccharides, diosgenin, polyphenols, and allantoin, have been widely used for the treatment of inflammatory and metabolic disorders [21,22].
There have been many reports on the phytochemical and anti-inflammatory activities of Dioscorea in the past, as well as studies on the treatment of metabolic diseases, such as obesity and diabetes. However, reported reviews on Dioscorea have focused on single species, such as D. alata, D. nipponica Makino, D. bulbifera, or generalized pharmacological activities [17,19,20], and there has not been a comprehensive review on anti-inflammatory activity and the treatment of metabolic diseases of Dioscorea, which is not conducive to relevant studies. Therefore, we have mainly selected the research results on the bioactive compounds, anti-inflammatory activity, and treatment of metabolic disorders of Dioscorea in the last five years. This is beneficial for the reader to quickly understand the latest research progress of Dioscorea and provides an updated reference for researchers to facilitate the development of medicine against inflammatory and metabolic diseases.

2. Methodology

In order to review the bioactive components and potential to treat inflammatory and metabolic diseases of Dioscorea, we searched databases, such as PubMed (https://pubmed.ncbi.nlm.nih.gov, accessed on 17 March 2023), Web of Science (http://apps.webofknowledge.com/, accessed on 17 March 2023), and China National Knowledge Infrastructure (http://www.cnki.net, accessed on 17 March 2023). The literature selection is shown in the PRISM flow chart (Figure 1).

3. Bioactive Compounds

Plant-based secondary metabolites/bioactive compounds play an important role in the prevention of different human diseases [23]. Dioscorea is rich in bioactive compounds, such as polysaccharides, steroidal saponins, polyphenols, and allantoin. Figure 2 shows the structures of the main bioactive compounds in Dioscorea. It is noteworthy that there are significant differences in the content of bioactive compounds between intra- and interspecies, cultivated and wild varieties [24,25]. The contents of main bioactive compounds of Dioscorea are summarized in Table 1.

3.1. Polysaccharides

Polysaccharides are the main active component of yam. It is mainly composed of mannose, xylose, arabinose, glucose, and galactose [33]. The various biological activities of yam polysaccharides, such as antioxidant, hypoglycemic, and anti-tumor activities, have attracted the interest of researchers [34,35]. Currently, some yam polysaccharides have been isolated and purified. However, different yam species and different extraction methods yielded polysaccharides with different structures and biological activities [36]. Different concentrations of yam polysaccharides and their derivatives purified by hot water extraction and column chromatography had the same ability to scavenge hydroxyl radicals as Vitamin C. Additionally, the antioxidant activity of the polysaccharides and its derivatives against superoxide anion and lipid peroxidation reached the level of Vitamin C [37]. In addition, three polysaccharides purified from Chinese Huaishan yams had the ability to inhibit α-glucosidase and the relative value-added rate of B16 mouse melanoma cells [38].
The water-soluble polysaccharides (WSP) of Dioscorea hispida extracted by aqueous extraction, papain-assisted extraction, and tempeh inoculum-assisted extraction had hypoglycemic effects. Among them, tempeh inoculum-assisted WSP extract had the highest hypoglycemic activity, while papain-assisted WSP extract had the highest glucose absorption inhibition [39]. Another study used hot water extraction, acid extraction, hot-compressed water extraction, and enzyme-assisted extraction of polysaccharides from the peel of the Dioscorea opposita Thunb. The polysaccharide yields, hypoglycemic activity, and antioxidant activity of these four extraction methods were compared. It was shown that hot-compressed water extraction and acid extraction gave the highest polysaccharide yields, and acid extraction showed the highest antioxidant activity and α-amylase inhibitory activity [34]. Optimization of the ultrafiltration-assisted extraction of yam polysaccharides using response surface methodology exhibited not only antioxidant activity but also dose-dependent inhibition of proliferation of human gastric gland cancer cells in vitro [40].
A new study shows that the heteropolysaccharide (DOP-2) isolated from Dioscorea opposita can modulate the metabolic disorders caused by benzopyrene (BaP). DOP-2 can improve BaP-induced liver injury by enhancing the activity of immune and antioxidant enzymes, exhibiting hepatoprotective activity. Moreover, DOP-2 can modulate the concentration of organic acids to increase the fecal absorption of BaP [41].

3.2. Steroidal Saponins

Steroidal saponins are important secondary metabolites of Dioscorea. Currently, more than 100 steroidal saponins have been isolated from various Dioscorea species, such as stigmastanol, furostanol, spirostanol, and cholestanol [42]. The major saponins are dioscin, gracillin (spirostanol glyclosides), protodioscin, and protogracillin (furostanol glycosides) [26]. Hydrolysis of diosgenin derivatives yields diosgenin, which are an important source of corticosteroids, anti-inflammatory, androgen, estrogen, and contraceptive drugs in the pharmaceutical industry [43,44]. Diosgenin are in high demand in the market. There are 137 species of Dioscorea. containing diosgenin, of which 41 species contain more than 1% diosgenin and have great utilization value [45]. More and more studies show that steroidal saponins have a wide range of biological activities, such as anticancer [46,47], antifungal [48], anti-inflammatory [49,50], hypolipidemic [51], and cardioprotective [52]. Lee et al. [53] isolated 15 steroidal saponins from tubers and leaves of Dioscorea esculenta, including 4 furanosteroid glycosides and 11 spirostanol glycosides. More importantly, four novel steroidal saponins were identified. A study analyzed the total saponin content of 1151 materials of six yam cultivars and the powder of five wild yam (D. villosa). The results showed that the total saponin content ranged from 37.36–129.97 mg/g. The average total saponin content was 42.15 mg/g for D. cayenensis, 17.65 mg/g for D. esculenta, and 17.44 mg/g for D. rotundata, with D. cayenensis having the highest total saponin content (78.31 mg/g). No steroidal saponins were found in the other three cultivars (D. alata, D. bulbifera, and D. dumetorum) [29].
Nazir et al. [54] reported for the first time the effective extraction method, solvent system, drying process and seasonal variation of diosgenin content of Dioscorea deltoidea. Drying the plant material in a cool place was the best drying method with the highest diosgenin content of 1.43% dry weight (DW). Microwave-assisted extraction with 50% ethanol yielded the highest amount of diosgenin in a short time (within 3 min) The tubers collected in December had the highest diosgenin content of 1.19% DW. In addition, latitude, annual parity temperature, and maximum annual precipitation affected steroid saponin biosynthesis. RDA analysis showed that environmental factors limited the total variance of steroid saponin biosynthesis (phytosterols, diosgenin, and steroid saponins) by 64.67% [55].

3.3. Polyphenols

Polyphenols, as secondary metabolites of plants, are a class of phytochemicals with potential health-promoting effects. Polyphenols are classified into flavonoids and non-flavonoids (phenolic acids) [56]. Zhang et al. [57] used high performance liquid chromatography–high resolution electrospray ionization mass spectrometry to identify seven phenolic compounds extracted from purple yam (Dioscorea alata L). This study not only confirmed the presence of ferulic and mustard acids, but also identified previously unidentified phenolic compounds, including vanillic acid, caffeic acid, p-coumaric acid, kaempferol, and quercetin hydrate. In addition, gallic acid, rutin and quercetin were confirmed to be present in the leaf extract of Dioscorea pentaphylla [58]. Jing et al. [59] isolated 13 monocyclic phenols and two flavonoids (formononetin and (+)-Catechin) from D. collettii. Among them, two benzylacetone derivatives, three phenylpropanoids, and formononetin were isolated for the first time from the genus Dioscorea.
Two flavonols, caryatin and 3′-O-methyl caryatin, isolated from the tubers of D. nummularia accounted for more than 92% of the stable methanolic extract by Fel et al. [28]. This study further determined the content of these two flavonols in the tubers and bulbils of 411 local varieties and hybrids of eight edible yam species. The results showed that these substances varied considerably at intra- and interspecific levels. Among them, caryatin in D. bulbifera, D. alata, and D. nummularia accounted for more than 90% of the total antioxidant activity of the methanolic extract of the tubers. Du et al. [60] isolated two new phenols and six known phenols from Dioscorea zingiberensis. One of the known phenols with a trihydroxy-diarylheptane backbone could mediate mitochondrial dysfunction by reducing ATP depletion and reactive oxygen species production, thereby preventing sodium taurocholate-induced pancreatic acinar cells necrosis. Zhou et al. [61] isolated and identified 17 phenolic compounds from Dioscorea leaves. The major phenolic compound was rosmarinic acid with the highest content of 22.31 ± 1.33 mg/g DW in the leaves of Dioscorea glabra Roxb. Rutin was the major flavonoid, followed by quercetin. Rutin was highest in the leaves of Dioscorea alata, with 8.66 ± 0.29 mg/g DW. Leaf extracts showed good antioxidant activity against hydrogen peroxide-induced oxidative stress.

3.4. Allantoin

Allantoin is one of the secondary metabolites of yam. Allantoin has antidiabetic and antioxidant activity and promotes wound healing [17]. Lebot et al. [29] detected six cultivars (D. alata, D. bulbifera, D. dumetorum, D. cayenensis, D. esculenta, and D. rotundata) for allantoin content ranging from 0.23 to 22.35 mg/g DW. A study on age-specific differences in Dioscorea polystachya showed that four-year-old tubers with the highest allantoin content were the best choice for industrial production of large quantities of allantoin [62]. Studies have shown that allantoin derived from Dioscorea batatas significantly reduced body weight and plasma biochemical parameters, and inhibited structural damage to the liver, pancreas, and skeletal muscle in mice with high-fat-diet (HFD)- and streptozotocin (STZ)-induced diabetes [63].

3.5. Alkaloids

Alkaloids are the largest group of secondary metabolites, consisting of nitrogen bases synthesized from amino acids. The alkaloids present in Dioscorea tuber are dioscorine, dihydrodioscorine, dioscoretine, and dumetorine [64,65]. The content of alkaloids in D. belophylla was 0.68 mg/100 g [30]. D. alata and D. esculenta contained significant amounts of alkaloids at 1.64 mg/100 g and 1.89 mg/100 g, respectively [31]. Another study showed that the alkaloid content in different Dioscorea species varied from 7.2 to 16 mg/100 g [24]. Alkaloid derivatives have analgesic, antitussive, and antibacterial pharmacological activities [12]. However, one of the major alkaloids in yam, dioscorine, is a toxic isoquinoline alkaloid. Dioscorine can be effectively removed by soaking in 1.0 M sodium chloride solution for 5 days compared to the traditional detoxification method of peeling and slicing and soaking in running river water for seven days [17,66]. In addition, Haji et al. [67] designed a detoxification device based on a water cycle operation and found that a rotating operation of the water cycle was more effective in removing doscorine.

3.6. Phenanthrene Derivatives

Phenanthrene is a group of polycyclic aromatic hydrocarbons naturally occurring in various plants. Kim et al. [32] quantified the contents of three phenanthrenes 2,7-dihydroxy-4,6-dimethoxyphenanthrene, 6,7-dihydroxy-2,4-dimethoxyphenanthrene, and batatasin in the pericarp of D. batatas, D. polystachya, D. quinqueloba, and D. bulbifera. The highest content of 2,7-dihydroxy-4,6-dimethoxyphenanthrene was 173.69 μg/g in D. quinqueloba. D. polystachya was rich in 6,7-dihydroxy-2,4-dimethoxyphenanthrene and batatasin at 166.99 μg/g and 419.73 μg/g, respectively. It has been shown that 2,7-dihydroxy-4,6-dimethoxyphenanthrene isolated from D. batatas Decne peel extract has antioxidant and anti-inflammatory activities [68]. In addition, 2,7-dihydroxy-4,6-dimethoxyphenanthrene isolated from D. opposita and D. batatas Decne peel extracts had protective properties against intestinal mucosal damage and inhibited lipopolysaccharide (LPS)-induced inflammatory responses in BV2 microglia [69,70]. Phenanthrene and dihydrophenanthrene derivatives isolated from D. communis were shown to possess anticholinesterase and antioxidant activities [71].

4. Anti-Inflammatory Activity

Chronic inflammatory diseases are recognized as the most important cause of death, with over 50% of deaths attributable to inflammation-related diseases, such as ischemic heart disease, cancer, diabetes, and NAFLD [72]. The bioactive compounds of Dioscorea, such as phenanthrene derivatives, steroidal saponins, polysaccharides and polyphenols, exert anti-inflammatory activity and have excellent effects on a wide range of inflammatory diseases [68,73,74].

4.1. Enteritis

Yam polysaccharides are not digested by gastrointestinal fluids. Most of the indigestible yam polysaccharides are degraded and utilized by intestinal microorganisms, promoting the production of short-chain fatty acids (SCFAs) and the relative abundance of probiotic bacteria [75]. In LPS-stimulated cocultured Caco-2/Raw264.7 cells, both yam polysaccharides and fecal fermented yam polysaccharides significantly inhibited the levels of inflammatory mediators nitrogen oxide (NO)/Inducible nitric oxide synthase (iNOS), tumor necrosis factor-α (TNF-α) and interleukin-1 beta (IL-1β), and increased the expression of intestinal tight junction protein 1 (ZO-1) and Occludin. Among them, the anti-inflammatory and intestinal barrier-protective effects of fecal fermented yam polysaccharides were superior to those of yam polysaccharides [74].
Chen et al. [76] reported for the first time that anthocyanins (DACNs) isolated from Dioscorea alata L. tubers exhibited anti-inflammatory activity in a trinitrobenzene sulfonic acid (TNBS)-induced colitis model in mice. Compared with the TNBS group, 80 mg kg−1 DACNs significantly reduced disease scores in mice with inflammatory bowel disease (IBD). DACNs significantly promoted the expression of colonic tight junction-related proteins. Similarly, DACNs significantly reduced the concentrations of myeloperoxidase and iNOS in colonic tissues, as well as TNF-α and interferon-γ (IFN-γ) in serum. It was shown that Dioscorea batatas peel extract not only reduced nitric oxide production and expression of pro-inflammatory proteins in LPS-stimulated RAW264.7 cells, but also decreased the level of reactive oxygen species in hydrogen peroxide-stimulated HCT116 cells. Moreover, yam bark extract promoted nuclear factor(erythroid-derived 2)-like 2 (Nrf2)-mediated antioxidant enzyme expression and inhibited nuclear factor kappa B (NF-κB)-mediated inflammatory responses in macrophages, thereby attenuating dextran sodium sulfate (DSS)-induced acute colitis in mice [77]. 6,7-Dihydroxy-2,4-dimethoxyphenanthrene (CYP4) isolated and purified from Chinese yam peel could inhibit extracellular signal-regulated kinase 1/2 (ERK1/2), NF-κB p65, pNF-κB, and cyclooxygenase-2 (COX-2) expression in colon tissue to inhibit DSS-induced intestinal mucosal injury in mice. It was shown that the daily intake of 120 mg/(kg·d) of Chinese yam peel extract and 4 mg/(kg·d) of CYP4 had good preventive effect on acute colitis in mice [69].

4.2. Arthritis

Gouty arthritis (GA) is a very common form of inflammatory arthritis. Controlling inflammation is the key to preventing gouty arthritis [78]. NOD-, LRR-, and pyrin-domain containing protein 3 (NLRP3) inflammatory vesicles induce inflammation and pyrophosphate cell death in response to pathogens and endogenous activators. Excessive activation of NLRP3 inflammatory vesicles leads to the development of inflammatory diseases and cancer [79]. In vitro and in vivo studies have shown that dioscin has anti-inflammatory effects on sodium urate (MSU)-mediated gouty arthritis. The underlying molecular mechanism may be that dioscin reduces inflammation by reducing the production of pro-inflammatory cytokines and inhibiting the activation of inflammasome NLRP3 and Toll-like receptor 4 (TLR 4)/NF-κB signaling pathway [80]. In addition, in vivo studies in mice with GA found that total yam saponins, the main components of which are dioscin, protodioscin, and pseudo protodioscin, exert anti-inflammatory effects for GA by regulating the expression of some key genes in the neutrophilic alkaline phosphatase-3 (NALP3), caspase-1, apoptosis-associated speck-like (ASC), and mitogen-activated protein kinase (MAPK)-peroxisome proliferator-activated receptor γ (PPARγ) signaling pathways [81,82]. Diosgenin from Dioscorea nipponica Makino had a therapeutic effect on collagen-induced arthritis in mice. Dioscorea exhibited anti-arthritic activity by down-regulating the differentiation of Th17 cells, significantly reducing the Th3 cell ratio in CD17+ T cells, IL-17 and IL-6 levels, and RORat expression [83].

4.3. Dermatitis

Natural saponins have anti-inflammatory effects. Jegal et al. [84] investigated the effect of Gracillin, the main saponin isolated from the rhizome of Dioscorea quinqueloba, on atopic dermatitis (AD). It was found that in 2,4-dinitrochlorobenzene (DNCB)-induced AD mice, gracillin effectively alleviated AD symptoms (redness, itchiness, swelling, and skin lichenification). In addition, the impaired skin barrier and skin hydration were significantly improved. Gracillin also inhibited the overproduction of IL-4 in vitro and in vivo. In addition to AD, paeonol, the bioactive phenol in Dioscorea japonica Thunb., plays an effective role in dry skin diseases. In an acetone–ether–water (AEW)-treated mouse model of dry skin, paeonol attenuated skin inflammatory responses and scratching behavior and reduced CXCR3-driven expression of spinal astrocyte activity-dependent genes [85].

4.4. Acute Pancreatitis

Acute pancreatitis (AP) is one of the common causes of hospitalization for gastrointestinal diseases and has a very high morbidity and mortality rate [86]. Endoplasmic reticulum stress (ERS) and Gasdermin D (GSDMD) affect AP. GSDMD accumulates in the endoplasmic reticulum of the glandular alveolar cells, which exacerbates the local pancreatic symptoms and systemic inflammation of AP. The mechanism of action of diosgenin derivative on AP was investigated by L-arginine-induced AP mouse model and mouse pancreatic alveolar cell model in vitro. It was shown that diosgenin derivatives isolated from Dioscorea zingiberensis could act as GSDMD inhibitors and inhibit L-arginine-induced AP by regulating GSDMD in the endoplasmic reticulum through the thioredoxin-interacting protein (TXNIP)/hypoxia-inducible factor-1α (HIF-1α) pathway [87]. Moreover, diosgenin were protective against pancreatic follicular cell injury in vivo and in vitro. Dihydrodiosgenin (5 or 10 mg/kg) had inhibitory effects on Tauro-induced AP, caerulein-induced AP, and palmitoleic acid plus ethanol-induced AP. The potential mechanism of action is to prevent excessive inflammatory responses by restoring mitochondrial function in the pancreas and inhibiting the phosphoinositide 3-kinase gamma (PI3Kγ)/protein kinase B (Akt) pathway. More importantly, this ameliorated acute lung injury associated with pancreatitis [88].

4.5. Neuroinflammation

Neuroinflammation is caused by endogenous or exogenous injury activating microglia. Dioscorea nipponica rhizome ethanol extract (DNRE) and dioscin significantly inhibited LPS-induced microglia activation and reduced the levels of pro-inflammatory factors (IL-1β, IL-6) and inflammatory enzymes (iNOS, COX-2). The dioscin of DNRE may be an inhibitor of p65 phosphorylation and translocation, and inhibition of transcription of multiple inflammatory cytokines and has a preventive effect on neuroinflammation [89,90]. Another study showed that 6,7-dihydroxy-2,4-dimethoxyphenanthrene (DHDMP), a phenanthrene compound isolated from Dioscorea batatas Decne, also significantly reduced the production of pro-inflammatory mediators and inhibited nuclear translocation of NF-κB and phosphorylation of p38 MAPK in BV2 cells [70].
Overall, these studies support the anti-inflammatory activity of Dioscorea extracts against enteritis, arthritis, dermatitis, pancreatitis, and neuroinflammation. The bioactive substances exerting anti-inflammatory effects are mainly yam polysaccharides, polyphenols, phenanthrene derivatives, and steroidal saponin derivatives. The anti-inflammatory mechanism is mainly inhibition of pro-inflammatory cytokine production and modulation of inflammation-related signaling pathways.

5. Prevention and Treatment of Metabolic Diseases

Metabolic diseases, such as obesity, T2DM, dyslipidemia, and NAFLD, are a serious threat to human health. The search for natural active compounds of plant origin to prevent and treat metabolic diseases is a hot research topic today. A growing number of studies have shown that the bioactive compounds of Dioscorea play an important role in the prevention and treatment of metabolic diseases (Table 2). The therapeutic effects of Dioscorea on metabolic diseases are shown in Figure 3.

5.1. Obesity

Obesity is a chronic multisystem disease that leads to a reduced life expectancy, reduced quality of life, and disability, especially for people with cardiovascular disease, diabetes, and cancer [107,108]. It was shown that the addition of Dioscorea batatas extract to the diet for 14 weeks significantly inhibited HFD-induced weight gain in mice. Supplementation with yam extract inhibited the expression of adipogenic transcription factor and its target gene cluster of differentiation 36 (CD36), reducing visceral fat accumulation and suppressing HFD-induced obesity. In addition, yam extract reduced the levels of pro-inflammatory cytokines (TNF-α, IL-6, monocyte chemoattractant protein-1 (MCP-1)) in HFD-induced adipose tissue [91]. Dioscorea japonica propagules promote fat oxidation and utilization and inhibits cholesterol synthesis. For carbohydrate metabolism, the propagules inhibit glycolysis and glycogen synthesis and promote gluconeogenesis. In summary, the propagules regulate fat metabolism and carbohydrate metabolism to inhibit disorders of carbohydrate and fat metabolism in high fat-loaded mice [92].
Jeong et al. [93] reported for the first time the anti-obesity effect of the n-butanol extract of D. oppositifolia. The main components of the extract were 3,5-dimethoxy-2,7-phenanthrenediol and (3R,5R)-3,5-dihydroxy-1,7-bis(4-hydroxyphenyl)-3,5-heptanediol. The results showed that the extract inhibited dietary fat absorption and increased fecal fat excretion to reduce body weight, and adipose tissue weight in HFD-induced obese mice. Du et al. [94] reported metabolic studies on the anti-obesity effects of diarylheptanoid, isolated and identified from Dioscorea zingiberensis. Diarylheptanoid inhibits differentiation and fat accumulation in 3T3-L1 cells by balancing energy metabolism and regulating lipid metabolism. This involves multiple aspects of metabolism, including regulation of tricarboxylic acid (TCA) cycle and glycolysis, disruption of amino acid metabolism, inhibition of purine metabolism, and reduction of ab initio synthesis of fatty acids and lipids.

5.2. Dyslipidemia

Dyslipidemia is an imbalance of plasma lipids, such as total cholesterol (TC), triglyceride (TG), low-density lipoprotein (LDL), and high-density lipoprotein (HDL), caused by abnormal lipid metabolism, which may lead to obesity, diabetes, and cardiovascular disease [109]. A study showed that resistant starch (RS) from purple yam (Dioscorea alata L.) reduced body weight, liver weight, and adipose tissue weight in hyperlipidemia-induced hamsters. RS improved lipid composition and regulated lipid metabolism by decreasing TC, TG, and LDL-C concentrations and increasing HDL-C concentrations [95]. In addition, Chinese yam polysaccharides also have anti-hyperlipidemic activity and can significantly reduce LDL and TC levels [96]. Dioscin, diosgenin and pseudoprotodioscin are phytoestrogens that are structurally similar to estrogens, with antioxidant, anti-inflammatory, anti-adipogenic, and inhibitory effects on atherosclerosis associated with estrogen deficiency [97,98,110]. Dioscin was found to not only regulate lipid metabolic homeostasis, but also attenuate postmenopausal atherosclerosis in HFD and ovariectomy (HFD-OVX)-induced low-density lipoprotein receptor-deficient (LDLR-/-) mice by inhibiting oxidative stress, inflammation, and apoptosis in part dependent on the peroxisome proliferator-activated receptor-γ coactivator 1-α (PGC-1α)/estrogen receptor alpha (Erα) pathway [98].
The Di’ao Xinxuekang (DXXK), extracted from Dioscorea nipponica Makino, attenuated HFD-induced hyperlipidemia and atherogenesis in mice, probably through inhibition of the proprotein convertase subtilisin/kexin type 9 (PCSK9) signaling pathway [99]. Shi et al. [100] reported that Shuangyu Tiaozhi granules (STG), consisting of two herbs from the genus Dioscorea, reduced hypercholesterolemia and hepatic cholesterol accumulation. 3-Hydroxy-3-methylglutaryl-CoA reductase (HMGCR) is the rate-limiting enzyme in a key step of catalytic cholesterol synthesis. STG inhibits the expression of HMGCR and sterol regulatory element-binding protein-2 (SREBP-2) and increases plasma LDL-C clearance to lower cholesterol levels, thereby slowing the onset and progression of coronary heart disease.

5.3. Diabetes

Diabetes is a metabolic disease characterized by high blood glucose. The prevalence of diabetes is 10.5% (536.6 million people) in 2021 and is expected to rise to 12.2% in 2045, by which time it will affect 783.2 million people worldwide [111,112]. T2DM is the most prevalent metabolic disorder characterized by insulin resistance, which results from chronic hyperglycemia and inadequate response of peripheral tissues to circulating insulin [113]. Dioscorea extracts for the treatment of diabetes and its complications, including diabetic nephropathy, diabetic liver disease, diabetic neuropathy, diabetic vasculopathy, diabetic reproductive dysfunction, diabetic eye disease, and diabetic hearing impairment, have been widely reported [63,114,115,116,117]. Dioscorea esculenta administration for eight weeks was reported to significantly inhibit the increase in plasma insulin and fasting glucose levels in T2DM rats. In addition, metabolic insulin-sensitive glucose metabolic clearance was increased, and insulin resistance was improved [101]. A novel Dioscorea opposita Thunb polysaccharide-zinc inclusion complex was shown to reduce blood glucose and insulin levels, as well as improve lipid levels and oxidative stress in STZ-induced diabetic rats [102].
A study has shown that dioscin from Dioscoreae nipponicae rhizomes improves insulin resistance by reducing hyperglycemia and hyperlipidemia. Dioscin promotes glycogen synthesis and inhibits gluconeogenesis and lipogenesis by modulating the miR-125a-5p/STAT3 signaling pathway, thus significantly alleviating the glucolipid metabolism disorders of T2DM [103]. Chinese yam polysaccharides reduced body weight in obese mice. Additionally, it improved insulin resistance by reducing serum inflammatory factors 1L-1β, 1L-10, and leptin and increasing insulin sensitivity [96]. Dioscorea and its extracts have the potential to become functional foods and drugs for the treatment of T2DM.

5.4. Non-Alcoholic Fatty Liver Disease

NAFLD is a metabolic stress liver injury that is closely associated with obesity, insulin resistance, T2DM, atherosclerosis, and metabolic syndrome, and is a major cause of cirrhosis and hepatocellular carcinoma [118,119]. Dioscin significantly attenuated hepatic lipid accumulation and improved serum and hepatic biochemical index levels in vitro and in vivo [98,103]. Dioscin modulates the expression levels of downstream proteins, including promoting the expression of silent information regulator of transcription 1 (SIRT1), AMP-activated protein kinase (AMPK), carnitine palmitoyltransferase (CPT), forkhead box O 1 (FoxO1), and adipose triglyceride lipase (ATGL), and inhibiting the expression of SREBP-1c, FAS, and SCD to suppress lipid metabolism. It was shown that dioscin significantly improves hepatic lipid metabolism and reduces TG accumulation to prevent NAFLD by activating the START1/AMPK signaling pathway [104]. Diosgenin activates the AMPK pathway and inhibits LXRα to suppress high glucose-induced TG accumulation, thereby preventing hepatic lipid accumulation in vitro and in vivo. Diosgenin also improved HFD-induced liver function disturbance [105]. In addition, fecal metabolomics analysis showed that diosgenin improved dysbiosis of gut microbiota HFD-induced NAFLD rats, which was significantly associated with lipid and amino acid metabolism [106].
In short, Dioscorea extracts, such as polyphenols, polysaccharides, resistant starch, dioscin, and diosgenin elements, have excellent preventive and therapeutic effects on obesity, dyslipidemia, diabetes, and NAFLD. Bioactive extracts of Dioscorea ameliorate metabolic disorders by modulating relevant signaling pathways and molecular targets, thus inhibiting the damage of metabolic diseases.

6. Conclusions

Dioscorea is not only the main source of food, but also a potential source of bioactive compounds. Dioscorea is rich in beneficial bioactive compounds, such as yam polysaccharides, diosgenin, dioscin, allantoin, and polyphenols. A growing number of in vitro and in vivo studies have demonstrated the utility of these bioactive compounds in the prevention and treatment of inflammation, obesity, diabetes, dyslipidemia, and NAFLD. Various signaling pathways associated with oxidative stress and inflammation, such as NF-κB, NLRP3, MAPK-PPARγ, PI3Kγ/Akt, PGC-1α/Erα, and START1/AMPK, are the main targets of action of the bioactive compounds. In summary, Dioscorea is a promising medicinal and edible plant, which deserves more intensive research. In the future, the isolation and identification of natural bioactive compounds of Dioscorea should be considered. Moreover, it is necessary to investigate the role and mechanism of these natural bioactive compounds in anti-inflammatory and preventive metabolic diseases. As a medicinal and edible plant, Dioscorea has the potential to be developed into functional foods for the prevention and treatment of inflammatory and metabolic diseases.

Author Contributions

Conceptualization, S.Z.; methodology, F.Z.; data curation, S.T. (Siyu Tao); writing—original draft preparation, Z.W.; writing—review and editing, Z.W. and Q.M.; supervision, G.H.; project administration, S.T. (Shenpeng Tan). All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Natural Science Foundation of China, grant number 81473104.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Saklayen, M.G. The Global Epidemic of the Metabolic Syndrome. Curr. Hypertens. Rep. 2018, 20, 12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Su, Z.; Guo, Y.; Huang, X.; Feng, B.; Tang, L.; Zheng, G.; Zhu, Y. Phytochemicals: Targeting Mitophagy to Treat Metabolic Disorders. Front. Cell Dev. Biol. 2021, 9, 686820. [Google Scholar] [CrossRef]
  3. Baker, R.G.; Hayden, M.S.; Ghosh, S. NF-κB, Inflammation, and Metabolic Disease. Cell Metab. 2011, 13, 11–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Gasmi, A.; Mujawdiya, P.K.; Noor, S.; Lysiuk, R.; Darmohray, R.; Piscopo, S.; Lenchyk, L.; Antonyak, H.; Dehtiarova, K.; Shanaida, M.; et al. Polyphenols in Metabolic Diseases. Molecules 2022, 27, 6280. [Google Scholar] [CrossRef] [PubMed]
  5. Xu, X.; Yi, H.; Wu, J.; Kuang, T.; Zhang, J.; Li, Q.; Du, H.; Xu, T.; Jiang, G.; Fan, G. Therapeutic Effect of Berberine on Metabolic Diseases: Both Pharmacological Data and Clinical Evidence. Biomed. Pharmacother. 2021, 133, 110984. [Google Scholar] [CrossRef] [PubMed]
  6. Sharma, V.; Gautam, D.N.S.; Radu, A.-F.; Behl, T.; Bungau, S.G.; Vesa, C.M. Reviewing the Traditional/Modern Uses, Phytochemistry, Essential Oils/Extracts and Pharmacology of Embelia ribes Burm. Antioxidants 2022, 11, 1359. [Google Scholar] [CrossRef]
  7. Khuntia, A.; Martorell, M.; Ilango, K.; Bungau, S.G.; Radu, A.-F.; Behl, T.; Sharifi-Rad, J. Theoretical Evaluation of Cleome Species’ Bioactive Compounds and Therapeutic Potential: A Literature Review. Biomed. Pharmacother. 2022, 151, 113161. [Google Scholar] [CrossRef] [PubMed]
  8. Behl, T.; Upadhyay, T.; Singh, S.; Chigurupati, S.; Alsubayiel, A.M.; Mani, V.; Vargas-De-La-Cruz, C.; Uivarosan, D.; Bustea, C.; Sava, C.; et al. Polyphenols Targeting MAPK Mediated Oxidative Stress and Inflammation in Rheumatoid Arthritis. Molecules 2021, 26, 6570. [Google Scholar] [CrossRef] [PubMed]
  9. Behl, T.; Bungau, S.; Kumar, K.; Zengin, G.; Khan, F.; Kumar, A.; Kaur, R.; Venkatachalam, T.; Tit, D.M.; Vesa, C.M.; et al. Pleotropic Effects of Polyphenols in Cardiovascular System. Biomed. Pharmacother. 2020, 130, 110714. [Google Scholar] [CrossRef]
  10. Li, H.; Zhang, X.; Wang, J. Progress of Basic Research on Dioscorea spp. in China. Econ. For. Res. 1999, 17, 44–48. [Google Scholar]
  11. Asiedu, R.; Sartie, A. Crops That Feed the World 1. Yams: Yams for Income and Food Security. Food Secur. 2010, 2, 305–315. [Google Scholar] [CrossRef]
  12. Padhan, B.; Panda, D. Potential of Neglected and Underutilized Yams (Dioscorea spp.) for Improving Nutritional Security and Health Benefits. Front. Pharmacol. 2020, 11, 496. [Google Scholar] [CrossRef] [Green Version]
  13. Shan, N.; Wang, P.; Zhu, Q.; Sun, J.; Zhang, H.; Liu, X.; Cao, T.; Chen, X.; Huang, Y.; Zhou, Q. Comprehensive Characterization of Yam Tuber Nutrition and Medicinal Quality of Dioscorea opposita and D. alata from Different Geographic Groups in China. J. Integr. Agric. 2020, 19, 2839–2848. [Google Scholar] [CrossRef]
  14. Epping, J.; Laibach, N. An Underutilized Orphan Tuber Crop—Chinese Yam: A Review. Planta 2020, 252, 58. [Google Scholar] [CrossRef] [PubMed]
  15. 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] [PubMed]
  16. 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] [PubMed] [Green Version]
  17. Lebot, V.; Lawac, F.; Legendre, L. The Greater Yam (Dioscorea alata L.): A Review of Its Phytochemical Content and Potential for Processed Products and Biofortification. J. Food Compos. Anal. 2023, 115, 104987. [Google Scholar] [CrossRef]
  18. Adomėnienė, A.; Venskutonis, P.R. Dioscorea spp.: Comprehensive Review of Antioxidant Properties and Their Relation to Phytochemicals and Health Benefits. Molecules 2022, 27, 2530. [Google Scholar] [CrossRef] [PubMed]
  19. Ou-yang, S.; Jiang, T.; Zhu, L.; Yi, T. Dioscorea nipponica Makino: A Systematic Review on Its Ethnobotany, Phytochemical and Pharmacological Profiles. Chem. Cent. J. 2018, 12, 57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. 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. South Afr. J. Bot. 2021, 140, 365–374. [Google Scholar] [CrossRef]
  21. 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] [PubMed]
  22. 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]
  23. 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]
  24. Padhan, B.; Biswas, M.; Panda, D. Nutritional, Anti-Nutritional and Physico-Functional Properties of Wild Edible Yam (Dioscorea spp.) Tubers from Koraput, India. Food Biosci. 2020, 34, 100527. [Google Scholar] [CrossRef]
  25. Otegbayo, B.O.; Oguniyan, D.J.; Olunlade, B.A.; Oroniran, O.O.; Atobatele, O.E. Characterizing Genotypic Variation in Biochemical Composition, Anti-Nutritional and Mineral Bioavailability of Some Nigerian Yam (Dioscorea spp.) Land Races. J. Food Sci. Technol. 2018, 55, 205–216. [Google Scholar] [CrossRef] [PubMed]
  26. Kwon, H.-J.; Choi, S.-H.; Yoo, C.-S.; Choi, H.-Y.; Lee, S.-E.; Park, Y.-D. Development of an Analytical Method for Yam Saponins Using HPLC with Pulsed Amperometric Detection at Different Column Temperatures: Liquid Chromatography. J. Sep. Sci. 2013, 36, 690–698. [Google Scholar] [CrossRef]
  27. Lebot, V.; Malapa, R.; Molisalé, T. Development of HP-TLC Method for Rapid Quantification of Sugars, Catechins, Phenolic Acids and Saponins to Assess Yam (Dioscorea spp.) Tuber Flour Quality. Plant Genet. Resour. 2019, 17, 62–72. [Google Scholar] [CrossRef]
  28. Fel, B.; Baudouin, A.; Fache, F.; Czarnes, S.; Lebot, V.; Legendre, L. Caryatin and 3′-O-Methylcaryatin Contents in Edible Yams (Dioscorea spp.). J. Food Compos. Anal. 2021, 102, 104010. [Google Scholar] [CrossRef]
  29. Lebot, V.; Faloye, B.; Okon, E.; Gueye, B. Simultaneous Quantification of Allantoin and Steroidal Saponins in Yam (Dioscorea spp.) Powders. J. Appl. Res. Med. Aromat. Plants 2019, 13, 100200. [Google Scholar] [CrossRef]
  30. Poornima, G.N.; Ravishankar, R.V. Evaluation of Phytonutrients and Vitamin Contents in a Wild Yam, Dioscorea belophylla (Prain) Haines. Afr. J. Biotechnol. 2009, 8, 971–973. [Google Scholar]
  31. Senanayake, S.; Ranaweera, K.; Bamunuarachchi, A.; Gunaratne, A. Proximate Analysis And Phytochemical And Mineral Constituents In Four Cultivars Of Yams And Tuber Crops In Sri Lanka. Trop. Agric. Res. Ext. 2013, 15, 32. [Google Scholar] [CrossRef]
  32. Kim, H.; Cao, T.Q.; Yeo, C.; Shin, S.H.; Kim, H.; Hong, D.-H.; Hahn, D. Development and Validation of Quantitative Analysis Method for Phenanthrenes in Peels of the Dioscorea Genus. J. Microbiol. Biotechnol. 2022, 32, 976–981. [Google Scholar] [CrossRef] [PubMed]
  33. Chen, Y.-F.; Zhu, Q.; Wu, S. Preparation of Oligosaccharides from Chinese Yam and Their Antioxidant Activity. Food Chem. 2015, 173, 1107–1110. [Google Scholar] [CrossRef] [PubMed]
  34. Zhao, Z.; Wang, L.; Ruan, Y.; Wen, C.; Ge, M.; Qian, Y.; Ma, B. Physicochemical Properties and Biological Activities of Polysaccharides from the Peel of Dioscorea opposita Thunb. Extracted by Four Different Methods. Food Sci. Hum. Wellness 2023, 12, 130–139. [Google Scholar] [CrossRef]
  35. 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]
  36. 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]
  37. Zhou, S.; Huang, G.; Chen, G. Extraction, Structural Analysis, Derivatization and Antioxidant Activity of Polysaccharide from Chinese Yam. Food Chem. 2021, 361, 130089. [Google Scholar] [CrossRef]
  38. Zhu, Y.; Yang, L.; Zhang, C.; Tian, Y.; Zhang, F.; Li, X. Structural and Functional Analyses of Three Purified Polysaccharides Isolated from Chinese Huaishan-Yams. Int. J. Biol. Macromol. 2018, 120, 693–701. [Google Scholar] [CrossRef] [PubMed]
  39. Estiasih, T.; Sunarharum, W.B.; Rahmawati, A. Hypoglycemic Activity of Water Soluble Polysaccharides of Yam (Dioscorea hispida Dents) Prepared by Aqueous, Papain, and Tempeh Inoculum Assisted Extractions. Int. J. Nutr. Food Eng. 2012, 6, 878–884. [Google Scholar]
  40. Xue, H.Y.; Li, J.R.; Liu, Y.G.; Gao, Q.; Wang, X.W.; Zhang, J.W.; Tanokura, M.; Xue, Y.L. Optimization of the Ultrafiltration-Assisted Extraction of Chinese Yam Polysaccharide Using Response Surface Methodology and Its Biological Activity. Int. J. Biol. Macromol. 2019, 121, 1186–1193. [Google Scholar] [CrossRef] [PubMed]
  41. Bu, W.; Dai, W.; Liu, H.; Bu, H.; Ju, X.; Li, R.; Yuan, B. Structural Characterization of a Polysaccharide from Dioscorea opposita and Assessment of Its Hepatoprotective Activity. Process Biochem. 2022, 120, 156–168. [Google Scholar] [CrossRef]
  42. Salehi, B.; Sener, B.; Kilic, M.; Sharifi-Rad, J.; Naz, R.; Mudau, F.N.; Fokou, P.V.T.; Ezzat, S.M.; Bishbishy, M.H.E.; Taheri, Y.; et al. Dioscorea Plants: A Genus Rich in Vital Nutra- Pharmaceuticals—A Review. Iran. J. Pharm. Res. 2019, 18, 68–89. [Google Scholar] [PubMed]
  43. Yoon, K.D.; Kim, J. Preparative Separation of Dioscin Derivatives from Dioscorea villosa by Centrifugal Partition Chromatography Coupled with Evaporative Light Scattering Detection. J. Sep. Sci. 2008, 31, 2486–2491. [Google Scholar] [CrossRef]
  44. Sautour, M.; Mitaine-Offer, A.C.; Lacaille-Dubois, M.A. The Dioscorea Genus: A Review of Bioactive Steroid Saponins. J. Nat. Med. 2007, 61, 91–101. [Google Scholar] [CrossRef]
  45. Shen, L.; Xu, J.; Luo, L.; Hu, H.; Meng, X.; Li, X.; Chen, S. Predicting the Potential Global Distribution of Diosgenin-Contained Dioscorea Species. Chin. Med. 2018, 13, 58. [Google Scholar] [CrossRef] [PubMed]
  46. 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]
  47. Okubo, S.; Ohta, T.; Shoyama, Y.; Uto, T. Steroidal Saponins Isolated from the Rhizome of Dioscorea tokoro Inhibit Cell Growth and Autophagy in Hepatocellular Carcinoma Cells. Life 2021, 11, 749. [Google Scholar] [CrossRef] [PubMed]
  48. Yang, L.; Liu, X.; Zhuang, X.; Feng, X.; Zhong, L.; Ma, T. Antifungal Effects of Saponin Extract from Rhizomes of Dioscorea panthaica Prain et Burk against Candida albicans. Evid. Based Complement. Alternat. Med. 2018, 2018, 6095307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Xue, Z.; Cao, Z.; Jin, M.; Zhang, X.; Wang, X.; Dou, J.; Zhu, Y.; Ito, Y.; Guo, Z. New Steroid Saponins from Dioscorea zingiberensis Yam and Their Medicinal Use against I/R via Anti-Inflammatory Effect. Food Funct. 2021, 12, 8314–8325. [Google Scholar] [CrossRef]
  50. 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]
  51. Wang, T.; Choi, R.; Li, J.; Li, J.; Bi, C.; Zang, L.; Liu, Z.; Dong, T.; Bi, K.; Tsim, K. Antihyperlipidemic Effect of Protodioscin, an Active Ingredient Isolated from the Rhizomes of Dioscorea nipponica. Planta Med. 2010, 76, 1642–1646. [Google Scholar] [CrossRef]
  52. 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. Oxid. Med. Cell. Longev. 2017, 2017, 4176518. [Google Scholar] [CrossRef] [PubMed]
  53. Lee, H.J.; Watanabe, B.; Nakayasu, M.; Onjo, M.; Sugimoto, Y.; Mizutani, M. Novel Steroidal Saponins from Dioscorea esculenta (Togedokoro). Biosci. Biotechnol. Biochem. 2017, 81, 2253–2260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Nazir, R.; Kumar, V.; Dey, A.; Pandey, D.K. HPTLC Quantification of Diosgenin in Dioscorea deltoidea: Evaluation of Extraction Efficacy, Organ Selection, Drying Method and Seasonal Variation. South Afr. J. Bot. 2021, 138, 386–393. [Google Scholar] [CrossRef]
  55. Hou, L.; Li, S.; Tong, Z.; Yuan, X.; Xu, J.; Li, J. Geographical Variations in Fatty Acid and Steroid Saponin Biosynthesis in Dioscorea zingiberensis Rhizomes. Ind. Crops Prod. 2021, 170, 113779. [Google Scholar] [CrossRef]
  56. Di Lorenzo, C.; Colombo, F.; Biella, S.; Stockley, C.; Restani, P. Polyphenols and Human Health: The Role of Bioavailability. Nutrients 2021, 13, 273. [Google Scholar] [CrossRef] [PubMed]
  57. Zhang, J.; Tian, H.; Zhan, P.; Du, F.; Zong, A.; Xu, T. Isolation and Identification of Phenolic Compounds in Chinese Purple Yam and Evaluation of Antioxidant Activity. LWT 2018, 96, 161–165. [Google Scholar] [CrossRef]
  58. 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. J. Food Meas. Charact. 2019, 13, 622–633. [Google Scholar] [CrossRef]
  59. Jing, S.S.; Wang, Y.; Li, X.J.; Li, X.; Zhao, W.S.; Zhou, B.; Zhao, C.C.; Huang, L.Q.; Gao, W.Y. Phytochemical and Chemotaxonomic Studies on Dioscorea collettii. Biochem. Syst. Ecol. 2017, 71, 10–15. [Google Scholar] [CrossRef]
  60. Du, D.; Jin, T.; Zhang, R.; Hu, L.; Xing, Z.; Shi, N.; Shen, Y.; Gong, M. Phenolic Compounds Isolated from Dioscorea zingiberensis Protect against Pancreatic Acinar Cells Necrosis Induced by Sodium Taurocholate. Bioorg. Med. Chem. Lett. 2017, 27, 1467–1470. [Google Scholar] [CrossRef]
  61. 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]
  62. 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. Biodivers. 2021, 18, e2000856. [Google Scholar] [CrossRef] [PubMed]
  63. 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]
  64. Nimenibo-Uadia, R.; Oriakhi, A. Proximate, Mineral and Phytochemical Composition of Dioscorea dumetorum Pax. J. Appl. Sci. Environ. Manag. 2017, 21, 771. [Google Scholar] [CrossRef] [Green Version]
  65. Corley, D.G.; Tempesta, S.; Iwu, M. Convulsant Alkaloids from Dioscorea dumetorum. Tetrahedron Lett. 1985, 26, 1615–1618. [Google Scholar] [CrossRef]
  66. Kamaruddin, Z.H.; Sapuan, S.M.; Yusoff, M.Z.M.; Jumaidin, R. Rapid Detection and Identification of Dioscorine Compounds in Dioscorea hispida Tuber Plants by LC-ESI-MS. BioResources 2020, 15, 5999–6011. Available online: https://jtatm.textiles.ncsu.edu/index.php/BioRes/article/view/BioRes_15_3_5999_Kamaruddin_Rapid_Detection_Identification_Discorine (accessed on 12 March 2023). [CrossRef]
  67. Haji, R.; Mohd, H.; Mohd, N.; Noordin, A.; Anuar, M.F. Development of Automatic Alkaloid Removal System for Dioscorea hispida. Front. Sci. 2011, 16–20. Available online: http://eprints.unisza.edu.my/id/eprint/3049 (accessed on 12 March 2023).
  68. 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] [Green Version]
  69. Li, Q.; Li, K.; Hu, T.; Liu, F.; Liao, S.; Zou, Y. 6,7-Dihydroxy-2,4-Dimethoxyphenanthrene from Chinese Yam Peels Alleviates DSS-Induced Intestinal Mucosal Injury in Mice via Modulation of the NF-κB/COX-2 Signaling Pathway. J. Agric. Food Chem. 2021, 69, 4720–4731. [Google Scholar] [CrossRef]
  70. Lim, J.S.; Oh, J.; Yun, H.S.; Lee, J.S.; Hahn, D.; Kim, J.-S. Anti-Neuroinflammatory Activity of 6,7-Dihydroxy-2,4-Dimethoxy Phenanthrene Isolated from Dioscorea batatas Decne Partly through Suppressing the P38 MAPK/NF-κB Pathway in BV2 Microglial Cells. J. Ethnopharmacol. 2022, 282, 114633. [Google Scholar] [CrossRef]
  71. 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]
  72. Furman, D.; Campisi, J.; Verdin, E.; Carrera-Bastos, P.; Targ, S.; Franceschi, C.; Ferrucci, L.; Gilroy, D.W.; Fasano, A.; Miller, G.W.; et al. Chronic Inflammation in the Etiology of Disease across the Life Span. Nat. Med. 2019, 25, 1822–1832. [Google Scholar] [CrossRef] [PubMed]
  73. Zhang, X.; Xue, Z.; Zhu, S.; Guo, Y.; Zhang, Y.; Dou, J.; Zhang, J.; Ito, Y.; Guo, Z. Diosgenin Revealed Potential Effect against Cerebral Ischemia Reperfusion through HIKESHI/HSP70/NF-κB Anti-Inflammatory Axis. Phytomedicine 2022, 99, 153991. [Google Scholar] [CrossRef]
  74. Bai, Y.; Zhou, Y.; Zhang, R.; Chen, Y.; Wang, F.; Zhang, M. Gut Microbial Fermentation Promotes the Intestinal Anti-Inflammatory Activity of Chinese Yam Polysaccharides. Food Chem. 2023, 402, 134003. [Google Scholar] [CrossRef] [PubMed]
  75. Gill, P.A.; van Zelm, M.C.; Muir, J.G.; Gibson, P.R. Review Article: Short Chain Fatty Acids as Potential Therapeutic Agents in Human Gastrointestinal and Inflammatory Disorders. Aliment. Pharmacol. Ther. 2018, 48, 15–34. [Google Scholar] [CrossRef] [Green Version]
  76. 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] [PubMed]
  77. 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]
  78. Zhang, M.; Zhang, X.; Tian, T.; Zhang, Q.; Wen, Y.; Zhu, J.; Xiao, D.; Cui, W.; Lin, Y. Anti-Inflammatory Activity of Curcumin-Loaded Tetrahedral Framework Nucleic Acids on Acute Gouty Arthritis. Bioact. Mater. 2022, 8, 368–380. [Google Scholar] [CrossRef]
  79. Biasizzo, M.; Kopitar-Jerala, N. Interplay Between NLRP3 Inflammasome and Autophagy. Front. Immunol. 2020, 11, 591803. [Google Scholar] [CrossRef] [PubMed]
  80. Han, J.; Shi, G.; Li, W.; Xie, Y.; Li, F.; Jiang, D. Preventive Effect of Dioscin against Monosodium Urate-Mediated Gouty Arthritis through Inhibiting Inflammasome NLRP3 and TLR4/NF-kB Signaling Pathway Activation: An In Vivo and In Vitro Study. J. Nat. Med. 2021, 75, 37–47. [Google Scholar] [CrossRef]
  81. 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]
  82. Zhou, Q.; Yu, D.H.; Zhang, N.; Wang, X.Y.; Zhang, N.; Lin, F.F.; Liu, S.M. Therapeutic Effects of Total Saponins From Dioscorea nipponica Makino on Gouty Arthritis Based on the MAPK-PPARγ Signaling Pathway: An In Vitro Study. Nat. Prod. Commun. 2020, 15, 1934578X2090449. [Google Scholar] [CrossRef] [Green Version]
  83. Gao, Y.; Wang, Y.; Song, H.; Guo, Y.; Xing, E.; Zhao, X.; Li, W.; Wang, J.; Yu, C. Effect of Diosgenin on T-Helper 17 Cells in Mice with Collagen-Induced Arthritis. Pharmacogn. Mag. 2020, 16, 486. [Google Scholar]
  84. Jegal, J.; Park, N.J.; Jo, B.G.; Bong, S.K.; Jegal, H.; Yang, M.; Kim, S.N. Anti-Atopic Properties of Gracillin Isolated from Dioscorea quinqueloba on 2,4-Dinitrochlorobenzene-Induced Skin Lesions in Mice. Nutrients 2018, 10, 1205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Wang, W.; Li, Q.; Zhao, Z.; Liu, Y.; Wang, Y.; Xiong, H.; Mei, Z. Paeonol Ameliorates Chronic Itch and Spinal Astrocytic Activation via CXCR3 in an Experimental Dry Skin Model in Mice. Front. Pharmacol. 2022, 12, 805222. [Google Scholar] [CrossRef] [PubMed]
  86. Barlass, U.; Dutta, R.; Cheema, H.; George, J.; Sareen, A.; Dixit, A.; Yuan, Z.; Giri, B.; Meng, J.; Banerjee, S.; et al. Morphine Worsens the Severity and Prevents Pancreatic Regeneration in Mouse Models of Acute Pancreatitis. Gut 2017, 67, 600–602. [Google Scholar] [CrossRef]
  87. Zhang, C.; Niu, H.; Wan, C.; Yu, X.; Xin, G.; Zhu, Y.; Wei, Z.; Li, F.; Wang, Y.; Zhang, K.; et al. Drug D, a Diosgenin Derive, Inhibits L-Arginine-Induced Acute Pancreatitis through Meditating GSDMD in the Endoplasmic Reticulum via the TXNIP/HIF-1α Pathway. Nutrients 2022, 14, 2591. [Google Scholar] [CrossRef]
  88. Shen, Y.; Wen, L.; Zhang, R.; Wei, Z.; Shi, N.; Xiong, Q.; Xia, Q.; Xing, Z.; Zeng, Z.; Niu, H.; et al. Dihydrodiosgenin Protects against Experimental Acute Pancreatitis and Associated Lung Injury through Mitochondrial Protection and PI3Kγ/Akt Inhibition: Dydio Protects against Acute Pancreatitis. Br. J. Pharmacol. 2018, 175, 1621–1636. [Google Scholar] [CrossRef] [Green Version]
  89. Azam, S.; Kim, Y.-S.; Jakaria, M.; Yu, Y.-J.; Ahn, J.-Y.; Kim, I.-S.; Choi, D.-K. Dioscorea nipponica Makino Rhizome Extract and Its Active Compound Dioscin Protect against Neuroinflammation and Scopolamine-Induced Memory Deficits. Int. J. Mol. Sci. 2022, 23, 9923. [Google Scholar] [CrossRef]
  90. Giridharan, S.; Srinivasan, M. Mechanisms of NF-κB P65 and Strategies for Therapeutic Manipulation. J. Inflamm. Res. 2018, 11, 407–419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Song, H.Y. Dioscorea batatas Extract Attenuates High-Fat Diet-Induced Obesity in Mice by Decreasing Expression of Inflammatory Cytokines. Med. Sci. Monit. 2015, 21, 489–495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Shinozaki, F.; Kamei, A.; Watanabe, Y.; Yasuoka, A.; Shimada, K.; Kondo, K.; Arai, S.; Kondo, T.; Abe, K. Propagule Powder of Japanese Yam (Dioscorea japonica) Reduces High-Fat Diet-Induced Metabolic Stress in Mice through the Regulation of Hepatic Gene Expression. Mol. Nutr. Food Res. 2020, 64, 2000284. [Google Scholar] [CrossRef]
  93. Jeong, E.J.; Jegal, J.; Ahn, J.; Kim, J.; Yang, M.H. Anti-Obesity Effect of Dioscorea oppositifolia Extract in High-Fat Diet-Induced Obese Mice and Its Chemical Characterization. Biol. Pharm. Bull. 2016, 39, 409–414. [Google Scholar] [CrossRef] [Green Version]
  94. Du, D.; Gu, H.; Djukovic, D.; Bettcher, L.; Gong, M.; Zheng, W.; Hu, L.; Zhang, X.; Zhang, R.; Wang, D.; et al. Multiplatform Metabolomics Investigation of Antiadipogenic Effects on 3T3-L1 Adipocytes by a Potent Diarylheptanoid. J. Proteome Res. 2018, 17, 2092–2101. [Google Scholar] [CrossRef] [PubMed]
  95. Li, T.; Teng, H.; An, F.; Huang, Q.; Chen, L.; Song, H. The Beneficial Effects of Purple Yam (Dioscorea alata L.) Resistant Starch on Hyperlipidemia in High-Fat-Fed Hamsters. Food Funct. 2019, 10, 2642–2650. [Google Scholar] [CrossRef] [PubMed]
  96. Cheng, Z.; Hu, M.; Tao, J.; Yang, H.; Yan, P.; An, G.; Wang, H. The Protective Effects of Chinese Yam Polysaccharide against Obesity-Induced Insulin Resistance. J. Funct. Foods 2019, 55, 238–247. [Google Scholar] [CrossRef]
  97. Sun, B.; Yang, D.; Yin, Y.; Xiao, J. Estrogenic and Anti-Inflammatory Effects of Pseudoprotodioscin in Atherosclerosis-Prone Mice: Insights into Endothelial Cells and Perivascular Adipose Tissues. Eur. J. Pharmacol. 2020, 869, 172887. [Google Scholar] [CrossRef]
  98. Yang, Q.; Wang, C.; Jin, Y.; Ma, X.; Xie, T.; Wang, J.; Liu, K.; Sun, H. Disocin Prevents Postmenopausal Atherosclerosis in Ovariectomized LDLR-/- Mice through a PGC-1α/ERα Pathway Leading to Promotion of Autophagy and Inhibition of Oxidative Stress, Inflammation and Apoptosis. Pharmacol. Res. 2019, 148, 104414. [Google Scholar] [CrossRef]
  99. Qu, L.; Li, D.; Gao, X.; Li, Y.; Wu, J.; Zou, W. Di’ao Xinxuekang Capsule, a Chinese Medicinal Product, Decreases Serum Lipids Levels in High-Fat Diet-Fed ApoE–/– Mice by Downregulating PCSK9. Front. Pharmacol. 2018, 9, 1170. [Google Scholar] [CrossRef] [Green Version]
  100. Shi, J.; Li, R.; Liu, Y.; Lu, H.; Yu, L.; Zhang, F. Shuangyu Tiaozhi Granule Attenuates Hypercholesterolemia through the Reduction of Cholesterol Synthesis in Rat Fed a High Cholesterol Diet. BioMed Res. Int. 2019, 2019, 4805926. [Google Scholar] [CrossRef] [Green Version]
  101. Sato, K.; Fujita, S.; Iemitsu, M. Dioscorea esculenta—Induced Increase in Muscle Sex Steroid Hormones Is Associated with Enhanced Insulin Sensitivity in a Type 2 Diabetes Rat Model. FASEB J. 2017, 31, 793–801. [Google Scholar] [CrossRef] [Green Version]
  102. Zhang, Y.; Khan, M.Z.H.; Yuan, T.; Zhang, Y.; Liu, X.; Du, Z.; Zhao, Y. Preparation and Characterization of D. opposita Thunb Polysaccharide-Zinc Inclusion Complex and Evaluation of Anti-Diabetic Activities. Int. J. Biol. Macromol. 2019, 121, 1029–1036. [Google Scholar] [CrossRef]
  103. Xu, L.N.; Yin, L.H.; Jin, Y.; Qi, Y.; Han, X.; Xu, Y.W.; Liu, K.-X.; Zhao, Y.Y.; Peng, J.-Y. Effect and Possible Mechanisms of Dioscin on Ameliorating Metabolic Glycolipid Metabolic Disorder in Type-2-Diabetes. Phytomedicine 2020, 67, 153139. [Google Scholar] [CrossRef] [PubMed]
  104. Yao, H.; Tao, X.; Xu, L.; Qi, Y.; Yin, L.; Han, X.; Xu, Y.; Zheng, L.; Peng, J. Dioscin Alleviates Non-Alcoholic Fatty Liver Disease through Adjusting Lipid Metabolism via SIRT1/AMPK Signaling Pathway. Pharmacol. Res. 2018, 131, 51–60. [Google Scholar] [CrossRef] [PubMed]
  105. Cheng, S.; Liang, S.; Liu, Q.; Deng, Z.; Zhang, Y.; Du, J.; Zhang, Y.; Li, S.; Cheng, B.; Ling, C. Diosgenin Prevents High-Fat Diet-Induced Rat Non-Alcoholic Fatty Liver Disease through the AMPK and LXR Signaling Pathways. Int. J. Mol. Med. 2017, 41, 1089–1095. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Zhou, Y.; Li, R.; Zheng, Y.; Song, M.; Zhang, S.; Sun, Y.; Wei, M.; Fan, X. Diosgenin Ameliorates Non-Alcoholic Fatty Liver Disease by Modulating the Gut Microbiota and Related Lipid/Amino Acid Metabolism in High Fat Diet-Fed Rats. Front. Pharmacol. 2022, 13, 854790. [Google Scholar] [CrossRef]
  107. Sarma, S.; Sockalingam, S.; Dash, S. Obesity as a Multisystem Disease: Trends in Obesity Rates and Obesity-Related Complications. Diabetes Obes. Metab. 2021, 23, 3–16. [Google Scholar] [CrossRef] [PubMed]
  108. Blüher, M. Metabolically Healthy Obesity. Endocr. Rev. 2020, 41, bnaa004. [Google Scholar] [CrossRef] [Green Version]
  109. Pappan, N.; Rehman, A. Dyslipidemia. StatPearls. 2022. Available online: https://www.ncbi.nlm.nih.gov/books/NBK560891/ (accessed on 13 February 2023).
  110. Morais, I.C.P.d.S.; Moura, I.J.L.; Sabino, C.K.B.; Nicolau, L.A.D.; Souza, F.d.M.; Silva-Filho, J.C.d.; Oliveira, R.d.C.M.; Medeiros, J.V.R.; Lima, S.G.d.; Oliveira, A.P.d. Cardiovascular Effect of Diosgenin in Ovariectomized Rats. J. Med. Food 2019, 22, 248–256. [Google Scholar] [CrossRef]
  111. Cho, N.H.; Shaw, J.E.; Karuranga, S.; Huang, Y.; da Rocha Fernandes, J.D.; Ohlrogge, A.W.; Malanda, B. IDF Diabetes Atlas: Global Estimates of Diabetes Prevalence for 2017 and Projections for 2045. Diabetes Res. Clin. Pract. 2018, 138, 271–281. [Google Scholar] [CrossRef]
  112. Sun, H.; Saeedi, P.; Karuranga, S.; Pinkepank, M.; Ogurtsova, K.; Duncan, B.B.; Stein, C.; Basit, A.; Chan, J.C.N.; Mbanya, J.C.; et al. IDF Diabetes Atlas: Global, Regional and Country-Level Diabetes Prevalence Estimates for 2021 and Projections for 2045. Diabetes Res. Clin. Pract. 2022, 183, 109119. [Google Scholar] [CrossRef]
  113. Yaribeygi, H.; Sathyapalan, T.; Atkin, S.L.; Sahebkar, A. Molecular Mechanisms Linking Oxidative Stress and Diabetes Mellitus. Oxid. Med. Cell. Longev. 2020, 2020, 8609213. [Google Scholar] [CrossRef] [Green Version]
  114. Guo, C.; Ding, G.; Huang, W.; Wang, Z.; Xiao, W. Total Saponin of Dioscoreae hypoglaucae Rhizoma Ameliorates Streptozotocin-Induced Diabetic Nephropathy. Drug Des. Devel. Ther. 2016, 2016, 799–810. [Google Scholar] [CrossRef] [Green Version]
  115. 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] [PubMed]
  116. Gan, Q.; Wang, J.; Hu, J.; Lou, G.; Xiong, H.; Peng, C.; Zheng, S.; Huang, Q. The Role of Diosgenin in Diabetes and Diabetic Complications. J. Steroid Biochem. Mol. Biol. 2020, 198, 105575. [Google Scholar] [CrossRef] [PubMed]
  117. Hajiabolhassan, F.; Tavanai, E. Diabetes-Induced Auditory Complications: Are They Preventable? A Comprehensive Review of Interventions. Eur. Arch. Otorhinolaryngol. 2021, 278, 3653–3665. [Google Scholar] [CrossRef]
  118. Powell, E.E.; Wong, V.W.-S.; Rinella, M. Non-Alcoholic Fatty Liver Disease. Lancet 2021, 397, 2212–2224. [Google Scholar] [CrossRef]
  119. Cui, Y.; Wang, Q.; Chang, R.; Zhou, X.; Xu, C. Intestinal Barrier Function–Non-Alcoholic Fatty Liver Disease Interactions and Possible Role of Gut Microbiota. J. Agric. Food Chem. 2019, 67, 2754–2762. [Google Scholar] [CrossRef]
Figure 1. PRISMA flow chart presenting literature data selection.
Figure 1. PRISMA flow chart presenting literature data selection.
Molecules 28 02878 g001
Figure 2. The structures of the main bioactive compounds in Dioscorea.
Figure 2. The structures of the main bioactive compounds in Dioscorea.
Molecules 28 02878 g002
Figure 3. The therapeutic effects of Dioscorea on metabolic diseases.
Figure 3. The therapeutic effects of Dioscorea on metabolic diseases.
Molecules 28 02878 g003
Table 1. The contents of main bioactive compounds in Dioscorea.
Table 1. The contents of main bioactive compounds in Dioscorea.
CompoundsContentSpeciePlant PartRef.
Protodioscin13.5–14.9 mg/gD. nipponica [26]
Protogracillin7.7–8.4 mg/gTuber
Dioscin2.3–3.8 mg/g
Gracillin0.7–1.2 mg/g
Gallic acid1.34–2.35 mg/g DWD. alataTubers and bulbils[17]
Epicatechin0.45–10.71 mg/g DWD. alataTubers and bulbils[17]
Catechins25.18 mg/gD. bulbiferaBulbils[27]
6.96 mg/gD. bulbiferaTubers
0.32 mg/gD. esculentaTubers
Phenolic acids4.33 mg/gD. bulbiferaTubers
4.87 mg/gD. alataTubers
9.55 mg/gD. nummulariaTubers
Caryatin1030 µg/g DWD. alata, D. bulbifera, D. cayenensis, D. dumetorumacc, D. esculentaacc, D. nummularia acc, D. pentaphyllaTubers[28]
3′-O-Methyl caryatin457 µg/g DW
Allantoin4.23–20.8 mg/gD. alata, D. bulbifera, D. cayenensis, D. dumetorum, D. esculenta, D. rotundataPowders[29]
0.68 mg/100 gD. belophyllaTubers[30]
1.64 mg/100 gD. alataTubers[31]
1.89 mg/100 gD. esculentaTubers[31]
Alkaloid7.2–16 mg/100 g DWD. oppositifolia, D. hamiltonii, D. pubera, D. wallichii, D. hispida, D. pentaphylla, D. bulbifera, D. glabra, D. alataTubers[24]
2,7-Dihydroxy-4,6-dimethoxyphenanthrene9.79–173.69 μg/gD. batatas, D. polystachya, D. quinqueloba, D. bulbiferaPeels[32]
6,7-Dihydroxy-2,4-dimethoxyphenanthrene46.65–166.99 μg/gD. batatas, D. polystachya
Batatasin97.19–419.73 μg/gD. batatas, D. polystachya
DW: dry weight.
Table 2. The therapeutic effects of Dioscorea on specific metabolic diseases.
Table 2. The therapeutic effects of Dioscorea on specific metabolic diseases.
SpeciesMetabolic DiseasesStudy TypeMain ResultsBioactive CompoundsRef.
D. batatas rhizomeObesityHFD-induced miceDownregulated the adipogenic transcription factor and its target gene (CD36)
Decreased the expression of proinflammatory cytokines (TNF-α, MCP-1, and IL-6)
-[91]
D. Japonica propagulesObesityHigh-fat-loaded miceSuppressed carbohydrate and fat metabolism disorders-[92]
D. oppositifolia rhizomesObesityHFD-induced obese miceSuppressed feeding efficiency and fat absorption3,5-dimethoxy-2,7-phenanthrenediol (3R,5R)-3,5-dihydroxy-1,7-bis(4-hydroxyphenyl)-3,5-heptanediol[93]
D. zingiberensis rhizomesDyslipidemia3T3-L1 cellsInhibited the differentiation and lipid accumulation of 3T3-L1 cellsDiarylheptanoid[94]
D. alata tubersDyslipidemiaHyperlipidemic hamstersAmeliorated lipid metabolism in association with gut microbiota modulationResistant starch[95]
Chinese yams rhizomesHyperlipidemia
Insulin resistance
Obesity-induced insulin resistance and hyperlipidemia in miceLowered the levels of LDL, cholesterol, leptin and IL-1β in serum, and down-regulated the expression of MMP-3 in visceral fat tissuesPolysaccharides[96]
Shanghai Winherb Medical S & T Development (Shanghai, China)AtherosclerosisOvariectomized ApoE-/- mice
Human umbilical vein
endothelial cells and Macrophages
Increased the level of ERα and eNOS protein
Suppress TNFα expression
Antiadipogenic effects
Pseudoprotodioscin[97]
Chenguang biotechnology Co. Ltd., (baoji, ChinaAtherosclerosisHFD-OVX-treated LDLR-/- miceInhibited postmenopausal Atherosclerosis via inhibiting oxidative stress, inflammation, apoptosis and promoting autophagy partly through PGC-1α/ERα pathwayDisocin[98]
D. nipponica Makino rhizomesLipid disorder
Atherosclerosis
High-fat diet-fed ApoE-/- miceReduce the levels of three major modifiable lipid risk factors, LDL-C, HDL-C, and TG
Inhibited PCSK9/LDLR signaling pathway
-[99]
Dioscoreae rhizomesHypercholesterolemiaHypercholesterolemic rat modelsDecreased body weight gain, liver weight ratio, serum lipids levels and hepatic lipids accumulation-[100]
D. esculenta tubersDiabetesType 2 diabetes rat modelIncreased muscle sex steroid hormone levels and decreased insulin resistanceDiosgenin[101]
D. opposita ThunbDiabetesSTZ-induced diabetic ratsDecreased the glucose and insulin levels and MDA contentsPolysaccharides[102]
D. nipponica rhizomesT2DMInsulin-induced HepG2 cells
Palmitic acid-induced AML12 cells
High-fat diet- and streptozotocin-induced T2DM rats
Inhibited miR-125a-5p/STAT3 signaling pathway and alleviate glycolipid metabolic disorderDioscin[103]
Shanghai Tauto Biochemical Technology Co., Ltd. (Shanghai, China)NAFLDMice models of NAFLDAlleviated liver lipid accumulation symptoms and improved the levels of serum and hepatic biochemical parametersDioscin[104]
-NAFLDHFD-induced NAFLD ratAmeliorated the hepatic lipid accumulation and HFD-induced liver function disturbanceDiosgenin[105]
Beijing gersion Bio-Technology Co., Ltd. (Beijing, China)NAFLDHigh-fat diet-fed NAFLD ratsSuppressed excessive weight gain, reduced serum levels of total cholesterol and triglycerides, and decreased liver fat accumulationDiosgenin[106]
Abbreviations: HFD: high-fat-diet; CD36: cluster of differentiation 36; TNF-α: tumor necrosis factor-α; MCP-1: monocyte chemoattractant protein-1, IL-6: interleukin-6; LDL: low-density lipoprotein; MMP-3: matrix metalloproteinase-3; eNOS: endothelial nitric oxide synthase; Erα: estrogen receptor alpha; PGC-1α: peroxisome proliferator-activated receptor-γ coactivator 1-α; HDL-C; high-density lipoprotein cholesterol; TG: triglyceride; HFD-OVX: HFD and ovariectomy; LDLR-/-: low-density lipoprotein receptor deficient; PCSK9: proprotein convertase subtilisin/kexin type 9; STZ: streptozotocin; MDA: malondialdehyde; AML12: alpha mouse liver 12; T2DM: type 2 diabetes mellitus; NAFLD: non-alcoholic fatty liver disease.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wang, Z.; Zhao, S.; Tao, S.; Hou, G.; Zhao, F.; Tan, S.; Meng, Q. Dioscorea spp.: Bioactive Compounds and Potential for the Treatment of Inflammatory and Metabolic Diseases. Molecules 2023, 28, 2878. https://doi.org/10.3390/molecules28062878

AMA Style

Wang Z, Zhao S, Tao S, Hou G, Zhao F, Tan S, Meng Q. Dioscorea spp.: Bioactive Compounds and Potential for the Treatment of Inflammatory and Metabolic Diseases. Molecules. 2023; 28(6):2878. https://doi.org/10.3390/molecules28062878

Chicago/Turabian Style

Wang, Zhen, Shengnan Zhao, Siyu Tao, Guige Hou, Fenglan Zhao, Shenpeng Tan, and Qingguo Meng. 2023. "Dioscorea spp.: Bioactive Compounds and Potential for the Treatment of Inflammatory and Metabolic Diseases" Molecules 28, no. 6: 2878. https://doi.org/10.3390/molecules28062878

Article Metrics

Back to TopTop