Skip to main content
Oxidative Medicine and Cellular Longevity logoLink to Oxidative Medicine and Cellular Longevity
. 2022 May 29;2022:1035441. doi: 10.1155/2022/1035441

Diosgenin: An Updated Pharmacological Review and Therapeutic Perspectives

Prabhakar Semwal 1,, Sakshi Painuli 1, Tareq Abu-Izneid 2, Abdur Rauf 3,, Anshu Sharma 4, Sevgi Durna Daştan 5,6, Manoj Kumar 7, Mohammed M Alshehri 8, Yasaman Taheri 9, Rajib Das 10, Saikat Mitra 10, Talha Bin Emran 11,12, Javad Sharifi-Rad 9,13,, Daniela Calina 14,, William C Cho 15,
PMCID: PMC9168095  PMID: 35677108

Abstract

Plants including Rhizoma polgonati, Smilax china, and Trigonella foenum-graecum contain a lot of diosgenin, a steroidal sapogenin. This bioactive phytochemical has shown high potential and interest in the treatment of various disorders such as cancer, diabetes, arthritis, asthma, and cardiovascular disease, in addition to being an important starting material for the preparation of several steroidal drugs in the pharmaceutical industry. This review aims to provide an overview of the in vitro, in vivo, and clinical studies reporting the diosgenin's pharmacological effects and to discuss the safety issues. Preclinical studies have shown promising effects on cancer, neuroprotection, atherosclerosis, asthma, bone health, and other pathologies. Clinical investigations have demonstrated diosgenin's nontoxic nature and promising benefits on cognitive function and menopause. However, further well-designed clinical trials are needed to address the other effects seen in preclinical studies, as well as a better knowledge of the diosgenin's safety profile.

1. Introduction

Currently, innovative approaches are being adopted to combat the onset and progression of human health problems [13]. One strategy involves using compounds derived from edible dietary plants, seeds and traditional medicinal herbs that have an impact on not only treating but also preventing cancer, either alone or along with existing treatment approaches [46]. Natural medicines which are derived from medicinal herbs or different vegetables or plants and fruits with promising health benefits are proved to be effective in treating and also preventing a wide range of human ailments, including obesity, neurological disorders, cancer, metabolic syndromes, cardiovascular disease, and diabetes [710]. A recent review by Newman and Cragg [11] reported the various natural products as a source of safe drugs for the treatment of various ailments in humans [11]. Over the decades, natural compounds derived from Chinese herbs have been used to a large extent in traditional practices of medicine. There are natural compounds that can be taken from plants, such as the anticancer medicine paclitaxel from Taxus brevifolia quercetin found in numerous vegetables and fruits [12, 13] and the antimalarial medication artemisinin from Artemisia apiacea, which have a wide spectrum of pharmacological effects [14].

Sapogenins are a group of chemicals that are found in a variety of natural products in glycoside form and enhance overall health. The most effective bioactive chemicals derived from natural product sources are steroidal sapogenins (otherwise known as spirostans) [15, 16]. The majority of steroidal sapogenins show pharmacological action in vitro and in preclinical animal models [15, 17]. Sapogenins have been the subject of several clinical investigations, which are either finished or ongoing [18, 19]. Diosgenin is a steroidal sapogenin that can be found in the Rhamnaceae, Liliaceae, Scrophulariaceae, Dioscoreaceae, Amaryllidaceae, Solanaceae, Leguminosae, and Agavaceae families [2023]. It is plentiful in Rhizoma polgonati, Smilax china, Dioscorea villosa, Trigonella foenum-graecum, and Dioscorea rhizome, among other plants with medicinal values. In the pharmaceutical sector, diosgenin is considered to be a fundamental material for starting the steroidal medicines to synthesize. One of the leading causes of death in the whole world is considered to be chronic diseases, and despite recent advancements in the procedures of treatment, synthetic medications still exhibit grating side effects along with chemoresistance, restricting the usage of these medications.

Phytochemicals have gained popularity due to their low risk of negative effects [8, 24]. Diosgenin is a phytochemical that has acquired prominence due to its usefulness in the treatment of lethal diseases such as nervous system disorders, diabetes, cancer, arthritis, asthma, cardiovascular disease, and others [25]. Scientists have used it in the treatment of inflammation, malignancies, hyperlipidemia, and infections as it has a wide spectrum of therapeutic attributes and pharmacological actions [26]. Diosgenin has been investigated extensively for the management and treatment of cancer [17], skin illnesses [27], cardiovascular illnesses [28], diabetes mellitus [29], atherosclerosis [30], and osteoporosis [31].

Diosgenin has been largely talked of and written about for its huge potential in pharmacological studies, as well as the intriguing rudimentary means of action, validating and expanding the comprehensive knowledge gained through its conventional use. Several mechanistic and preclinical studies have been conducted in this context, primarily during the last two decades, to better apprehend the true advantages and significance of diosgenin against manifold illnesses [32, 33]. Overall, the findings of multiple investigations suggest that diosgenin could be used as a novel multi-target-based therapeutic or chemopreventive drug for a variety of chronic diseases. As a result, it has become an agenda of great interest to develop effective ways to derive diosgenin from various natural resources, furthermore to establish medication dosage forms to grant its administration [34, 35]. Recently, the role of diosgenin in diabetic and cardiac diseases has been reviewed, but a holistic review of various biological activities based on preclinical and clinical studies is still missing in the literature to the best of our literature review [36, 37]. The fundamental objective of this review is to summarize the in-depth pharmacological activity moving from preclinical to clinical evidence and safety issues, to upcoming strategies to overcome present limitations.

2. Review Methodology

To conduct this comprehensive study, the most relevant studies on the pharmacological properties of diosgenin were analyzed. We searched for scientific publications published in journals electronic databases such as PubMed/MEDLINE, DOAJ, Scopus, Web of Science, and SciFinder, using the next MeSH terms: “Diosgenin/analogs & derivatives,” “Diosgenin/pharmacology,” “Diosgenin/therapeutic use,” “Humans,” Anti-Asthmatic Agents/pharmacology” “Chronic Disease/prevention & control,” “Cardiovascular Diseases/drug therapy,” “Inflammation/drug therapy,” “Neoplasms/drug therapy,” “Nervous System Diseases/drug therapy” “Saponins/pharmacology,” “Saponins/therapeutic use,” “Saponins/toxicity,” “steroids,” and “Signal Transduction/drug effects.” Studies that explained the molecular mechanisms, signaling pathways, molecular targets, and well-defined doses of pharmacological experiments were included. Duplicates, experimental pharmacological studies involving other test substances, and homeopathic preparations were excluded. The most relevant information was summarized in tables and images. The taxonomy of plants has been validated according to The Plant List [38, 39] and the chemical formulas verified with the PubChem database [40].

3. Sources

The compound diosgenin was discovered by Fuji and Matsukawa in 1936 [41], and its application in the synthesis of cortisone and other drugs was reported by Marker and coworkers [42]. The Dioscorea genus is the main source of diosgenin, and ~137 species of this genus contain diosgenin [26] including Dioscorea nipponica [43], Dioscorea zingiberensis [44], Dioscorea composita [45], and Dioscorea deltoidea [46, 47]. It is also isolated from other botanicals including Trigonella foenum-graecum, Costus speciosus, Tribulus terrestris L., Rhizoma polgonati, and Paris polyphylla (Table 1). The compound diosgenin is mostly produced from the hydrolysis process of steroidal saponins in the presence of strong acid, base, or enzyme catalyst, while microbial transformation technics are gaining much attention due to their highly specific nature, low cost, and environmentally friendly conditions [48].

Table 1.

Botanical sources of diosgenin.

SN Botanical name Family References
1 Trigonella foenum-graecum Fabaceae Arya and Kumar [49]
2 Costus speciosus Costaceae Selim and Al Jaouni [50]
3 Tribulus terrestris L Zygophyllaceae Wang et al., [51]
4 Smilax china L. Smilacaceae Yin et al. [52]
5 Rhizoma polgonation Asparagaceae Chen et al. [53]
6 Helicteres isora L. Malvaceae Deshpande and Bhalsing [21]
7 Paris polyphylla Melanthiaceae Gupta et al. [54]

4. Biosynthesis

Microwave-assisted extraction (MAE) and ultrasound-assisted extraction (UAE) methods were used for a better yield of diosgenin by using different solvent systems including acetone, ethanol, hexane, and petroleum ether with different concentrations (40, 60, 80, and 100%) and treatment time (MAE: 1.5, 3.0, 4.5, and 6.0 min; UAE: 30, 40, 50, and 60 min). The result of this study indicates that different parameters such as solvent type, concentration, treatment time, and extraction method have a significant impact on diosgenin extraction (yield). The UAE method (21.48%, 40.37 mg/100 g) showed better yield and diosgenin content compared to the MAE method (7.38%, 35.50 mg/100 g) at 80% ethanol concentration at 6 and 60 min, respectively [49]. In the industry of pharmaceutical sector, most hormonal medicines have diosgenin as their precursor. This makes diosgenin a classic precursor which is generated mostly by Dioscorea species. The mechanisms underlying the emergence and evolution or progression of biosynthesis of diosgenin in plants are unknown. The geneses of the diosgenin biosynthesis pathway, as well as its evolution, in yam, were validated in recent experimental research. The authors used a variety of diosgenin biosynthesis routes in this study [55] (Figure 1).

Figure 1.

Figure 1

The biosynthetic pathways of diosgenin. Dioscorea zingiberensis, for diosgenin biosynthesis uses two P450 genes, DzinCYP94D144 and DzinCYP90G6, the orthologs of PpCYP94D108 and PpCYP90G4, which, from cholesterol, catalyze the formation of diosgenin.

Metabolic engineering has also been used to produce diosgenin from yeast. Because cholesterol biosynthesis is more efficient in animal cells than in plant cells, researchers have created a strain with yeast chassis that produces cholesterol by integrating animal genes [56]. As a common precursor for triterpenoid biosynthesis, 2,3-oxidosqualene could be produced in significant amounts from it; Saccharomyces cerevisiae strain BY-T3 which was previously modified was chosen as the starting strain [57]. First, Cheng and coauthors discovered that positive selection-driven P450 gene neofunctionalization and duplication were significant in the diosgenin biosynthesis pathway formation. They discovered that CpG islands, which developed to maintain the balance of carbon flux between the production of diosgenin and starch and modulate or control gene expression in the diosgenin pathway, were responsible for diosgenin enrichment in the yam lineage. Finally, we heterologously produced diosgenin to 10 mg/L in yeast that was genetically modified by combining genes from plants, mammals, and yeast [55].

5. Pharmacological Properties of Diosgenin: Underlying Molecular Mechanisms and Signaling Pathways

5.1. Neuroprotective

The oxidative stress caused by the reactive oxygen stress leads to various ailments such as neurological disorders, cardiovascular diseases, and cancer [5860]. Health benefits of diosgenin administration are most commonly determined as a neuroprotective agent in terms of cognitive effects [6163], neuroinflammation [51, 6467], multiple sclerosis [68, 69], spinal cord injury [70, 71], stroke and thrombosis [72], and neuropathic pain [43, 7375]. Few recent investigations based on diosgenin and related aspects have been studied through in silico, in vitro, and in vivo approaches and discussed here. Regarding this, a new structure of the neuronal network has been developed to calculate the solubility of the diosgenin compound. The ordinary neural network modeling was used to improve the calculation accuracy and estimate the solubility of diosgenin in the n-alkanols with more carbon atoms due to its rational design concept [76].

In another study, diosgenin was evaluated against cerebral ischemia-reperfusion injury through in silico (proteome dynamic approach), in vitro, and in vivo methods. The authors investigated 5043 regulatory proteins from the brain samples and different signaling pathways [77]. Screening of small molecules for targeting the fungal virulence factors without any effect on viability was conducted by Aaron and the group [78]. They demonstrated that Cryptococcus neoformans (Cn) was prohibited from crossing the blood-brain barrier via suppressing the proteolytic activity of Mpr 1 with maintaining Cn viability. In another word, out of 240 compounds, diosgenin with two other compounds significantly inhibited Mpr 1 proteolytic activity (IC50: <10 μM) without any cell toxicity and blocked Cn crossing the BBB [78].

Cheng et al. evaluated the antiapoptotic effects of diosgenin in D-galactose-induced ageing brain (cerebral cortical apoptosis) [79]. A total of 36 male (12-week-old) Wistar rats were recruited and divided into four groups (control: 1 mg/kg/day of saline, i.p.; DD0: 150 mg/kg/day of D-galactose, i.p.; DD10: D-galactose +10 of diosgenin; and DD50: D-galactose+50 mg/kg/day of diosgenin orally) and treated up to 8 weeks. The overall results of this study showed the suppressing (D-galactose-induced neuronal Fas-dependent and mitochondria-dependent apoptotic pathways) and enhancing (Bcl-2 family-associated prosurvival and IGF-1-PI3K-AKT survival pathways) effects of diosgenin, which may trigger neuroprotective effects against D-galactose-induced ageing brain [79].

The neuroprotective effects of diosgenin in the diabetic mice model were also evaluated by Leng and the group [80]. All mice (male C57) were fed for up to 8 weeks (high fat diet) and intraperitoneally injected with streptozotocin (dose: 100 mg/kg for 2 days). Eligible mice were dived into four groups including control (n = 6), diabetic group (n = 6), low-dose diosgenin group (n = 6, 50 mg/kg), and high-dose diosgenin group (n = 6, 100 mg/kg). The main outcome of this study indicates that diosgenin significantly reduced the level of blood glucose and increased the body weight of diabetic mice. Diosgenin attenuated the level of MDA (in dose-dependent manner) but increased the activity of antioxidant enzymes (superoxide dismutase (SOD) and glutathione peroxidase (GPx)) and expression of signaling pathways (nuclear factor-erythroid factor 2-related factor (Nrf2), heme oxygenase (HO), and NAD(P)H dehydrogenase [quinone] (NQO)-1) in diabetic mice. These signaling pathways were involved in its neuroprotective activities [80].

Oyelaja-Akinsipo et al. investigated the neuroprotective and glucose-lowering ability of compound diosgenin in hyperglycemia-mediated cerebral ischemic brain injury using the zebra-fish model of type II diabetes mellitus [81]. Diabetes was developed in an experimental model by using streptozotocin (20 mg/kg b.w.) for 28 days, and two different doses (20 and 40 mg/kg b.w) of diosgenin were used. The results of this study indicated that diosgenin significantly decreased the concentration of glucose from 175.87 to 105.68 mg/d/L and 82.06 mg/d/L in both doses. The compound treatment increased the body weight and growth in diabetic zebra fishes (p < 0.05 and p < 0.001) and also enhanced the catalytic activity. Its activity also protects the brain from the possibility of hyperglycemic-mediated brain injury and apoptotic brain cell death [81].

The most representative studies on neuroprotective activities of diosgenin have been presented in Table 2.

Table 2.

Pharmacological studies regarding diosgenin's neuroprotective effect with mechanism of action.

Tested compounds Model Potential mechanisms References
Diosgenin C6 rat glioma cells
in vitro
↓ the dosage regimen of TMZ, ↑ MMP-2, ↑ apoptosis Rajesh et al. [82]
Diosgenin 5XFAD mice
in vivo
↓amyloid plaques, ↓neurofibrillary tangles in the cerebral cortex and hippocampus, ↑1,25D3-MARRS Tohda et al. [83]
Diosgenin ddY mice
in vivo
Memory enhancement effects mediated by 1,25D3-MARRS-triggered axonal growth, ↑ 1,25D3-MARRS Tohda et al. [84]
Diosgenin Transgenic 2576 mice
in vivo
↓AChE, ↓Bax/Bcl-2, ↓amyloid plaques production in the granule cells, ↑ NGF, ↑ SOD, p75 (NTR) Koh et al. [85]
Diosgenin Senescent mice induced via D-galactose
in vivo
↑learning and memory ability, ↑ SOD, ↑ GSH-Px, ↓ MDA level, ↑ endogenous antioxidant enzymatic activities Chiu et al. [61]
Diosgenin-rich extract Senescent mice induced via D-galactose
in vivo
↑learning and memory ability, ↑ SOD, ↑ GSH-Px, ↓ MDA level, ↑ endogenous antioxidant enzymatic activities Chiu et al. [86]
Diosgenin Sprague-Dawley rats
in vivo
↓ neuronal death rate, ↓pro-inflammatory cytokines, ↑impaired neurological functions at 100 and 200 mg/kg, ↑ IkBα, ↓ p65, ↓NF-κB Zhang et al. [87]
Diosgenin + curcumin (bivalent) MC65 neuroblastoma cells
in vitro
Antioxidant, anti-oligomerization ↓ amyloid-β oligomer formation Chojnacki et al. [88]
Diosgenin Sprague-Dawley rats with lipopolysaccharide
in vivo
↓TLR/NF-κB ↓ TLR2, ↓TLR4, ↓NF-κB Li et al. [89]
Diosgenin carbamate derivatives D-galactose aging mice
in vivo
Anti-inflammatory, anti-oxidant, β- amyloid, ↓ NO ↓ IL-1β, IL-6, TNF-α Yang et al. [90]
Arginyl–diosgenin Neuroinflammation model using BV2 cells induced by LPS C57BL/6 mice
in vitro/in vivo
↓activation of microglia, microphages, ↓CD4+ T cell proliferation, ↓Th1/Th17 cell differentiation, ↓ NO, ↓ iNOS, ↓ COX-2, ↓PGD2, ↓IL-6, ↓IL-1β, ↓ TNF-α Cai et al. [65]
Diosgenin Primary murine microglial cell line BV-2
in vitro
↓pro-inflammatory M1 markers via activation of microglia and without affecting M2 makers, ↓IκB-α, ↓ERK, ↓ MAPK, ↓p38 Wang et al. [51]
Diosgenin SH-SY5Y cell line H9c2 cell line
in vitro
Protective effects against SH-SY5Y cells, ↓angiogenesis at high concentration, ↓apoptosis Cai et al. [91]
Diosgenin Trimethyltin-injected transgenic 2576 mice
in vivo
Neuroprotective effects against different brain damages via NGF biosynthesis stimulation, ↓ AChE, ↓ Bax/Bcl-2, ↑ NGF, ↑ SOD Koh et al. [85]

Symbols: ↑ = increased; ↓ = decreased.

5.2. Anticancer

Cancer is the most dangerous disease that affects people around the globe, with one in every six deaths due to cancer [9294]. Different therapeutic tactics, including radiotherapy, chemotherapy, and laser-based therapy, are currently under practice [59, 95, 96]. A series of experiments on cytotoxicity aspects of diosgenin was conducted by several researchers worldwide. In a recent study, cytotoxic effects of standardized extracts, fraction and their compounds (Trigonella foenum-graecum) were evaluated against human cancer cells (SKOV-3, HeLa, and MOLT-4 cells). The steroid saponins fraction (C) showed strongest cytotoxic activity on cancer cells (IC50: 3.94 (HaCaT), 3.91 (HeLa), 3.97 (SKOV-3), and 7.75 (MOLT-4)). The fraction significantly increased reactive oxygen species production and caspases activity in the cells [97].

A total of 28 diosgenin amino acid ester derivatives (3a-3g and 7a-7g) were designed and synthesized by Ma et al. and evaluated for their cytotoxicity against six human cancer cells including K562, T24, MNK45, HepG2, A549, and MCF-7 [98]. The majority of derivatives displayed cytotoxic potential against these six tumor cells. Out of 28 derivatives, compound 7g exhibited significant cytotoxicity against the K562 cells (IC50: 4.41 μM) compared to diosgenin (IC50: 30.04 μM). Compound 7 also triggered K562 cells apoptosis through mitochondria-related pathways. The cytotoxicity of two different extracts of Paris polyphylla rhizomes (ethanol extract and diosgenin rich extract) were evaluated against human breast cancer cells (MCF-7 and MDA-MB-231), cervical cancer cells (HeLa), and Hep-2 cell lines. The diosgenin-rich extract significantly reduced the proliferation of all cancerous cells, and the maximum activity was observed in MCF-7 cells. Diosgenin-rich extract triggered upregulation of Bax and downregulation of Bcl-2 and BIRC5 pre-mRNA transcripts of genes [54].

Yin and coworkers developed and synthesized thirty-two new diosgenin derivatives and evaluated their cytotoxic activity against three human cancer cells (A549, MCF-7, and HepG2). Among them, compounds 8, 18, 26, and 30 were more potent compared to diosgenin. Compound 8 showed strong and low cytotoxic activity against HepG2 cells (IC50: 1.9 μM) and low L02 cells (IC50: 18.6 μM), respectively. Additionally, compound 8 induces G0/G1 cell cycle arrest and apoptosis in HepG2 cells. A molecular docking study also suggested that p38α-MAPK is a suitable target for compound 8 and fits well its active site [99]. Two new azasteroids were synthesized from diosgenin through the modification in the A and B rings and evaluated for their antiproliferative activity [100].

Diosgenin significantly inhibited the cell viability and motility of breast cancer cells and stimulate apoptosis via suppression of S-phase kinase-associated protein Skp-2 in breast cancer cells [101]. Although NF-κB promotes cancer initiation and development, but some reports demonstrate its role in tumor suppression [102]. Part of this several studies has been conducted to investigate the role of diosgenin, and its derivatives against different types of cancers are presented in Figure 2 and Table 3.

Figure 2.

Figure 2

Diagram with cellular targets and molecular mechanisms involved in anticancer effect of diosgenin. Abbreviations and symbols: ↑: increased; ↓: decreased; MMPs: matrix metalloproteinases; abbreviated mRNA: messenger RNA; TIMP: metallopeptidase inhibitor 1; AKT: serine/threonine kinase 1; PI3K: phosphatidylinositol-3-kinase; ERK: extracellular signal-regulated kinase; NF-κB: nuclear factor kappa-light-chain-enhancer of activated B cells; VEGF: vascular endothelial growth factor; DNA: deoxyribonucleic acid; TNFR1: tumor necrosis factor receptor 1; TNFα: tumor necrosis factor; PGE2: prostaglandin E2; COX: cyclooxygenase; LOX: 5-lipoxygenase; LTB4: leukotriene B4; PARP-1: poly(ADP-ribose) polymerase 1; ERα: estrogen receptor alpha; Src: proto-oncogene tyrosine-protein kinase; Cyt C: cytochrome C.

Table 3.

Anticancer properties of diosgenin and their derivatives in different types of cancer.

Model/cancer cell lines/IC50 Mechanism Pharmacological action References
MCF-7
Hs578T
IC50 = 0–40 μM
G2/M phase arrest, ↓cyclin B, ↓ Cdc2, ↓ Bcl-2
↑caspase 3
Regulation of the level of proteins which triggered cell cycle blockade at the G2/M phase Liao et al. [103]
MDA-MB-231
IC50 = 5 μM
Cdc42
Vav2
↓ cancer cell migration
↓actin polymerization
↓Vav2 phosphorylation
↓Cdc42 activation
He et al. [104]
MCF-7
IC50 = 10 − 30 μM
caspases-3,8, ↓PARP1, ↑Fas-L, ↓Survivin, ↓ERα, ↓cyclin D1, ↓c-Myc, ↓p-Src, ↓ ERK½, ↓p38 ↓expression of ER-α
↑apoptosis via extrinsic pathway
Chun et al. [105]
MCF-7
MDA-MB-231
IC50 = 1.15 − 5.76 μM
GATA3, DNMT3A, ZFPM2, E-cadherin, TET2, TET3, TET1, vimentin, MMP9 Diosgenin mediated pathways modulate the GATA3 expression at transcription and translation Aumsuwan et al. [106]
MDA-MB-231
MDA-MB-453
T47D
IC50 = 0 − 8 μg/mL
↓ Bcl-2
↓ cIAP-1
↓ McI-1
↑ apoptosis via downregulation of proteins related with inhibition
↑apoptotic process
Kim et al. [107]
MCF-7
MCF-10
MDA-231
IC50 = 20 − 30 μM
↓ pAKT (Ser473), ↓ AKT kinase activity, ↓ p-GSK3β, ↓ Raf, ↓ p-MEKs 1/2, ↓ p-MEKs 3/6, ↓ MEK-1, ↓ MEK-4, ↑ pElk-1, ↑ p21, ↓ XIAP, ↓ Bcl-2, ↓ Cdk-2, ↓ Survivin, ↓ cyclin D1, ↓ NF-κBp65, ↓ p65, ↑ IκB-α, ↓ pElk-1, ↑ Bax, ↓ NF-κB, ↑ caspase-3 ↑ G1 cell cycle arrest apoptosis in MCF-7 and MDA-231 cells while did not cause in MCF-10A cells Srinivasan et al. [108]
HuCCT1
QBC939
HuH28
SK-ChA1
RBE
Mz-ChA-1
IC50 = 0 − 40 μM
↑Bax/Bcl-2, ↑p21, ↑caspase-3, ↑ pARP-1,↓CyclinB1
↑Cyt c, ↑GSK3β-PY216
↓ GSK3β-PS9
G2/M phase arrest
↑apoptosis
The compound suppressed cholangiocarcinoma cells and triggered
Mao et al. [109]
HT-29
HCT-116
IC50 = 40 μM
↑ PGE2, ↑ COX-2, ↑ 5-LOX, ↑ LTB4 ↑ apoptosis in both cancer cell lines Lepage et al. [110]
HCT-116 ↑ ROS, ↑ Ca2+, ↑ NO, ↑ iNOS, ↑ DNA damage, ↑ Gna11, ↑ATP6V0C, ↑Ppp2r5e, ↑COX6C ↑mRNA The compound triggered mitochondrial damage and G2/M cell cycle arrest Chen et al. [111]
HeLa
CaSki
↑ caspases-3, 8, 9 Diosgenin and its glycoside derivatives showed strong anticancer activity with low necrotic activity and selective action Hernández-Vázquez et al. [112]
HeLa
SiHa
IC50 = 1.25 − 5.0 μg/mL
↑ ROS, ↑ Ca2+, DNA damage, ↑ Bid, Bcl-2, ↓ Bcl-xL, ↑ caspases-3, 9, ↑ Bax, ↑ Bak, ↑ p53 ↑ apoptosis
↓ cell proliferation, ↑DNA damage in both cell lines via modulation of protein level
Zhao et al. [113]
HeLa ↑ apoptosis, ↑ caspase-3 and -9 activity, ↓ Bcl-2 The compound significantly induced apoptosis in a dose and time-dependent manner Cai et al. [114]
HeLa
IC50 = 0 − 40 μM
G2/M phase, ↑ apoptosis, ↑ ROS The compound significantly inhibits cell proliferation, transformed cell morphology, arrests the cell cycle, and regulates apoptosis via death receptor and mitochondrial pathways. Ma et al. [115]
KYSE510
IC50 = 0.5 − 20 μM
G1/S arrest, ↑ apoptosis, ↑ cleaved caspase-9, ↑ Bax, ↑ Cyt c, ↑ ROS, ↓ Bcl-2 Peroxiredoxins 1 and 6 play an important role in compound induced apoptosis Zhiyu et al. [116]
NOZ
SGC996
IC50 = 0 − 8 μM
↓ ROS-mediated PI3K/AKT ↑ apoptosis via inhibition of reactive oxygen species-mediated PI3K/AKT signaling Song et al. [117]
MGC-803
MKN-45
IC50 = 1.25 − 5.0 μg/mL
↑ ROS, ↑ Ca2+, ↑ RBM-3, ↑ GALR-2, ↓ CliC-3, ↓ Bcl-2, ↑ Bax, ↑ caspase-3, 9,↑ MAPKs, ↓ CAP-1, ↓ Tribbles-2 Anticancer effects against human gastric cancer via inducing cell apoptosis, DNA damage, etc. Zhao et al. [118]
SGC-7901
IC50 = 0.65 − 2.6 μg/mL
↑ Fas, ↑ FasL, ↑ TNFR1, ↑ TNF-α, ↓ Bcl-2, ↑ Bax, ↑ Bak, ↓ bid, ↓ Bcl-xL, ↑ p53 mRNA
↑ caspase-3, 8
Anticancer activity Hu et al. [119]
HGC-27
MGC-803
SGC-7901
IC50 = 0 − 60 μmol
↓proliferation
↓ HOTAIR
↓Hox
↓proliferation of gastric cancer cells Ma et al. [120]
C6 allograft
IC50 = 1.25 − 5.0 μg/mL
↑ROS, ↑Ca2+, ↑MDA, ↑NO, ↑GSSG, ↓GSH, ↓Bcl-2, Bcl-xL, ↑Bak, ↑Bax, ↑caspase-3, 9 Anticancer activity Lv et al. [121]
HepG2
IC50 = 0 − 100 μM
↓TAZ
β-catenin
↓ cell growth, ↑apoptosis, ↑ apoptosis, ↑G2/M phase arrest Chen et al. [122]
HepG2
SMMC-7721
IC50 = 0 − 100 μM
G2/M phase arrest, ↑ DDX3, ↓mRNA, ↓ cyclin D1, ↑p21, ↑E-cadherin, ↓ Notch-1, ↓β-catenin ↓ cell growth
↑apoptosis via upregulation of DDX3
Yu et al. [123]
HepG2
IC50 = 0 − 40 μM
↑ caspase-3, 8, 9
↑ Bax, ↓Bcl-2, ↓Bid, ↑ROS, ↑ASK1
↓ cell growth
↑apoptosis in HepG2 cells via Bcl-2 protein-mediated pathways
Kim et al. [124]
Bel-7402
IC50 = 0.25 − 63 μmol/L
↑ TP53, ↑ Bax, ↓ Bcl-2
↑ caspase-3
↓cell growth
↑apoptosis via modulation of protein expression
Zhang et al. [125]
HepG2
IC50 = 0 − 20 μM
↑ apoptosis, G2/M phase arrest, ↓ cyclin B1, ↑ Bax, ↑ Bcl-2 ↓cell growth
↑apoptosis via modulation of protein expression
Wang et al. [126]
DU145
IC50 = 0.2 − 100 μg/mL
↑ LC3-II, ↑ caspase-9, ↓ PI3K, ↓AKT, ↓mTOR, ↑ Beclin-1, ↓ Bcl-2 ↓cell growth
↑apoptosis, ↑autophagy due to inhibition of mitochondrial pathways
Nie et al. [127]
DU145
IC50 = 33 ng/mL
↓vimentin, ↓ Mdm2, ↓ c-Met, ↓ ERK
↓AKT, ↓mTOR
↑ apoptosis
↓ HGF induced
↑ Mdm2, ↑vimentin
↓phosphorylation of Akt, mTOR
Chang et al. [128]
PC-3
IC50 = 0 − 30 μM
↓ MMP 2,7,9, ↓mRNA, ↓ EMMPRIN, ↑ TIMP, ↓ AKT, ↓ PI3K, ↓ ERK, ↓c-JNK, ↓NF-κB, ↓VEGF ↓ cancer cell growth
↑apoptosis via modulation of signaling pathways
Chen et al. [129]
PC-3
IC50 = 0 − 100 μM
G2/M phase arrest, ↓ NEDD4, ↓ p73, ↑ LATS1, ↓ p-AKT, ↓ TAZ ↓cell growth, ↑apoptosis cell cycle arrest Zhang et al. [130]
PC3
IC50 = 250 − 1000 μM
↑ [Ca2+]i, ↑ Mn2+ Significant anticancer activity Sun et al. [131]

5.3. Antiatherosclerosis

Atherosclerosis is a disease of the middle and large arteries characterized by the formation in the inner tunic and middle deposits of atheromatous plaques, which contain accumulations of LDL-cholesterol, lipophage, and sometimes calcifications on former lesions that prevent normal blood flow through the vessel. It is a disease in which plaque accumulates inside the arteries and can trigger serious problems including heart failure, stroke, or death [132134]. The pathogenic mechanisms are complex, involving lipid peroxidation, oxidative stress, inflammation, or altered immune response, causing aging and degenerative brain damage [135137].

The antiatherosclerosis potential of diosgenin and its derivatives were studied by few researchers. In this context, the curative effects of diosgenin on macrophage cholesterol metabolism and its mechanism were investigated by Lv and the group [30]. The diosgenin treatment significantly enhanced the expression of ATP-binding cassette transporter A1 (ABCA1) protein without any effect on liver X receptor α levels. Additionally, diosgenin treatment also inhibits aortic atherosclerosis progression via downregulation of miR-19b proteins in THP-1 macrophages/MPM-derived foam cells (Figure 3). The in vivo study of the diosgenin compound and its impact on Wistar rats treated with an atherogenic diet were performed by Binesh and coworkers. In this study, the atherogenic diet triggered the inflammatory mediators in the heart, liver, and brain via upregulation of COX-2, TNF-α, and NFkBp65 levels of the rats, while diosgenin treatment downregulated the level of these inflammatory markers and inhibit the development of atherosclerosis (Figure 3) [64]. The same research group reported the downregulation of NF-κB expression and polarization of macrophages by diosgenin treatment [138]. In another study, compound dioscin was evaluated for its inhibitory activity against atherosclerosis and postmenopausal atherosclerosis in ovariectomized LDLR-/-mice [139, 140].

Figure 3.

Figure 3

Schematic diagram showing the beneficial effect in atherosclerosis, thus preventing cardiovascular and neurodegenerative diseases. Abbreviations: Cox-2: cyclooxygenase-2; TNF-α: tumor necrosis factor-alpha; NF-κB: nuclear factor kappa-light-chain-enhancer of activated B cells.

5.4. Antiasthmatic

Asthma is a long-term condition which results in inflammation of the lower respiratory tract and affects children and adults. Junchao and coworkers evaluated the molecular mechanism of anti-trachea inflammatory effects produced by diosgenin via interactions with glucocorticoid receptor alpha [141]. They used ovalbumin-induced asthmatic mice and primary tracheal epithelial cells as experimental models. The results demonstrated that diosgenin significantly reduces the secretion of different inflammatory factors including tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6 via upregulation of glucocorticoid receptors, secretory leukocyte protease inhibitor, glucocorticoid-induced leucine zipper, mitogen-activated protein kinases (MAPK) phosphatase 1, and downregulation of heat shock proteins (HSP70).

5.5. Hepatoprotective

Liver disease can have genetic causes or it can be caused by a variety of factors that affect the liver, such as viruses or alcohol consumption [142144]. Obesity is also associated with liver disease. Over time, liver damage results in scarring (cirrhosis), which can lead to liver failure, a life-threatening condition [145, 146]. Under this topic, Xie et al. [147] reported on the impact of diosgenin on transforming growth factor (TGF)-β1-induced hepatic stellate cells and its mechanism of action for antifibrotic effects. The results of this study showed that diosgenin significantly inhibited the proliferation of TGF-β1-induced hepatic stellate cells and reduced the expression of collagen I and alpha-smooth muscle action as well as the expression of TGF-β receptors I and II. Additionally, diosgenin also downregulated the expression of TGF-β1-induced phosphorylation of Smad3 in hepatic stellate cells and displayed potential effects to treat liver fibrosis. In another study, Zhang and coworkers described the dioscin and its effect on alcoholic liver fibrosis (in vitro and in vivo). The results demonstrated that dioscin improved the condition of alcoholic liver fibrosis via modulation of toll-like receptor 4/myeloid differentiation primary response 88/NF-κB signaling pathway [148]. Other important studies related to diosgenin and its derivatives have been presented in Table 4 and shown in Figure 4.

Table 4.

Different biological activities of diosgenin and their derivatives with the mechanism of action.

Disease Experimental model Mechanism Pharmacological action References
Cardiac fibrosis Rat cardiac fibrosis cells
in vitro
IC50 = 0 − 10 μM
α-SMA
↓ TGF-β1
↓ p-Smad3
↓ proliferation of Ang II-induced cardiac fibrosis, ECM synthesis of rat cardiac fibrosis and expression of TGF-beta 1 and Smad3 phosphorylation in cardiac fibrosis cells Zhou et al., [149]
Pulmonary hypertension ICR mice
in vivo
Dose = 0.1 − 10 mg/kg
↓ NF-κB
↓p50/p65
↓ MAPK/p38
↓ iNOS
Pretreatment with diosgenin significantly suppresses the LPS-induced NF-κB, MAPK/p38 activation protective effects against acute lung injury or sepsis Gao et al., [150]
Graves disease BALB/c mice
in vivo
Dose = 20 − 100 mg/kg/day
↓ mRNA, ↓IGF-1, ↓NF-κB, ↓ cyclin D1
↓PCNA
Diosgenin treatment significantly reduces the TT4 level and thyroid size without affecting TRAb in graves' disease mice Cai et al., [151]
Diabetes Albino rats
in vivo
Dose =5-10 mg/kg/day
↓serum glucose, ↓ MDA, ↑GSH, ↑ SOD, ↑ GPx, ↓ protein carbonyl
↑ catalase
↓ blood glucose,
↓LDL cholesterol
↓cardiovascular risk
Kalailingam et al. [28]
Wistar rats
in vivo
Dose = 15 − 60 mg/kg/day
↓ serum glucose Diosgenin significantly reduced the blood glucose, increase the insulin blood level Saravanan et al. [152]
Swiss mice
in vivo
Dose = 1 mg/mL
α-amylase
α-glucosidase
Diosgenin demonstrated significant antidiabetic activity Ghosh et al., [153]
Osteoporosis Ovariectomized rats ↓ RANKL
↑ OPG
Diosgenin demonstrated significant antiosteoporotic activity compared to OVX control Zhang et al. [154]
Arthritis C57BL/6 mice
in vivo
↓ p-JAK2, ↓ p-STAT3, ↓ SDH, ↓COX, ↓ SOD, ↑ Bax Diosgenin treatment significantly inhibited the apoptosis and upregulated the mitochondrial oxidative stress capacity of chondrocytes in experimental mice with osteoarthritis Liu et al. [155]
Cardiotoxicity Male Balb/c mice
in vivo
Dose = 130 mg/kg/day
↓TBARS, ↓ ROS, ↓ caspase-3, ↓ NF-κB, ↑ cGMP, ↑ cAMP, ↓ PDE5 Cardioprotective
Diosgenin displayed antioxidant, anti-apoptotic, cGMP modulation activities
Chen et al. [156]

Figure 4.

Figure 4

Summarized scheme showing the most representative biological activities of the diosgenin. Abbreviations and symbols: ↑: increase; ↓: decrease; T4: thyroxine; LDL: low-density lipoprotein; OPG: osteoprotegerin; RANKL: receptor activator of nuclear factor kappa beta; IL: interleukin; TNF-α: tumor necrosis factor-alpha.

6. Clinical Studies

A clinical pilot study was conducted to investigate the efficacy and safety profile of diallyl thiosulfinate associated with nuciferine and diosgenin in the cure of primary and secondary erectile dysfunction. A total of 143 candidates (age 18-39 Y) were selected and treated orally with nuciferine and diosgenin for up to three months (single dose, alternative days). After three months of treatment, each candidate was screened in terms of the international index of erectile-5, premature ejaculation diagnostic tool, and male sexual health questionnaire. A significant enhancement was observed in terms of the international index of erectile-5 (8.7 vs. 14.01; p < 0.001) when compared with baseline and follow-up visits. The overall observation of the study indicates that these three compounds are capable to enhance the control of ejaculation in candidates suffering from premature and erectile dysfunction without any side effects [157].

In another study, Tohda et al. evaluated the impact of diosgenin-rich yam extract on synaptic loss and memory dysfunction using a transgenic mouse model of Alzheimer's. A placebo-controlled, randomized, double-blind, cross-over study was performed on 28 healthy volunteers (age: 20–81 years) and randomly treated with yam extract or placebo (12-weeks intake and 6-week washout period). The Japanese version of the Repeatable Battery for the Assessment of Neuropsychological Status (RBANS) test and the adverse effects were evaluated. The diosgenin-rich yam extract consumption (12 weeks) significantly increased the synaptic fluency or enhance cognitive function without any side effects [158].

The standardized multinutrient supplement including folic acid, selenium, vitamin E, catechins, glycyrrhizin, diosgenin, damiana, and omega-3-fatty acids showed beneficial effects on in vitro fertilization/intracytoplasmic sperm injection in terms of embryo quality [159]. The curative effects of Dioscorea villosa extract on menopausal systems, lipids, and sex hormones were evaluated [160].

A placebo-controlled, randomized, double-blind, cross-over study was conducted on 23 healthy women suffering from symptoms of menopause. All candidates were treated with wild yam cream or placebo for up to three months, and no significant adverse effects were observed in both the treatments. Additionally, no changes were also recorded in weight, systolic or diastolic blood pressure, total serum cholesterol, triglyceride, high-density lipoprotein cholesterol, glucose, estradiol, or serum [160].

7. Toxicological Profile: Safety and Side Effects

Only a few articles have acknowledged diosgenin's toxicity, even though there has been substantial research on its part and maneuver in the treatment along with prevention of cancer and further different chronic disorders. A study was published in 2009 on breast cancer cells by Srinivasan and coworkers where it was shown that diosgenin modulates AKT to regulate breast cancer cell survival and that this drug has no effect on normal breast epithelial cells (MCF-10A) except for its selective toxicity to cancer cells [108].

Steroidal saponins of different doses from D. zingiberensis were given to mice that were experimental in another in vitro investigation. Diosgenin was the metabolite and the main component of these saponins. There were no harmful effects up to a level of 562.5 mg/kg, according to the findings. However, steroidal saponins, which include diosgenin, displayed deleterious consequences and even death in a dose-dependent manner at dosages of 1125 mg/kg and higher. Interestingly, the steroidal saponins dosage, which is traditional, is 510 mg/kg/day, implying that steroidal saponins, together with diosgenin, have no significant toxicity at this dosage [161]. A recent study also discovered that diosgenin derivatives had antithrombotic properties. In vivo experiments revealed that they appeared to be protective and comparable to aspirin, with a decreased risk of bleeding and less stomach mucosal injury [162]. Furthermore, investigations have shown that diosgenin has a modest inhibitory impact on cytochrome P450 enzymes (CYPs), suggesting diosgenin fused with any other medicine would be safe to consider that they would have no toxicity [163]. These investigations demonstrated that diosgenin and its derivatives are nontoxic and have underlined their utility in the medicaments of chronic disorders including cancer.

8. Therapeutic Perspectives and Limitations

Diosgenin is a natural compound mostly found in Dioscorea species and starting material for the commercial synthesis of different steroids including cortisone, pregnenolone, and progesterone. The natural antioxidant compound diosgenin possesses different biological activities such as anticancer [164, 165], antidiabetic [28, 36, 152, 166169], multiple sclerosis [68, 69], and spinal cord injury [70, 71] and helps in the management of these diseases. Therefore, diosgenin could be functional and helpful in the treatment and prevention of a variety of disorders. The identification of diosgenin-specific targets, on the other hand, is critical for further validating its use in the treatment and elimination of diseases.

The main therapeutic limitations of diosgenin are represented by its low bioavailability; therefore, a special emphasis is being paid to the production of nanoformulations or conjugate complexes to improve the compound's bioavailability and pharmacokinetic features to develop it into a possible medicine. The potential of this chemical, its analogs, or combinations of this molecule with others has previously been demonstrated; however, carrier systems such as nanoparticles must be developed to govern diosgenin to the location where it works, boosting effectiveness and lowering adverse impacts. For example, encapsulated diosgenin PCL [poly(caprolactone)]-pluronic nanoparticles (PCL-F68-D-NPs) were developed by the nanoprecipitation method to improve performance in brain cancer therapy [170]. The developed nanoparticles (PCL-F68-D-NPs) displayed significant cytotoxicity against U87-MG cells compared to free diosgenin. Additionally, developed nanoparticles demonstrated suitable properties in terms of size distribution, stability, morphology, chemical and mechanical properties, encapsulation, and loading efficiency [170].

9. Overall Conclusions

A steroidal saponin, diosgenin, and its chemical and structural variants are useful in the treatment of a great variety of chronic conditions, including cardiovascular disease, several forms of lethal malignancies, nervous system problems, and autoimmune diseases. They have gotten a lot of interest from researchers all across the world. Diosgenin and its derivatives have been shown to have pharmacological benefits against cancer, diabetes, osteoporosis, Alzheimer's disease, and stroke in several investigations. Diosgenin has been shown to act on several molecular targets that are essential players in the occurrence and incidence of many serious disorders. Its multitargeting capability allows it to influence multiple molecular targets and signaling pathways at the same time. Diosgenin has an edge over the most commercial medicinal medicines available today because of this property. Furthermore, investigations demonstrating its nontoxic nature significantly promote the inclusion of this medicine in additional clinical studies or trials in the forthcoming days. Overall, diosgenin demonstrated great promise in the treatment and prevention of a variety of chronic diseases; nevertheless, additional clinical research is needed in the nearing days to come to confirm the preclinical findings and demonstrate the effective and secure usage of these purely natural compounds.

Acknowledgments

The authors acknowledge that some of the components used in figures are taken from freepik and pngtree (<a href='https://www.freepik.com/vectors/heart'>Heart vector created by macrovector - https://www.freepik.com</a>; <a href='https://pngtree.com/so/real-heart-clipart'>real heart clipart png from http://pngtree.com</a>).

Contributor Information

Prabhakar Semwal, Email: semwal.prabhakar@gmail.com.

Abdur Rauf, Email: mashaljcs@yahoo.com.

Javad Sharifi-Rad, Email: javad.sharifirad@gmail.com.

Daniela Calina, Email: calinadaniela@gmail.com.

William C. Cho, Email: chocs@ha.org.hk.

Data Availability

The data used to support the findings of this study are available from the corresponding author upon request.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

Authors' Contributions

Conceptualization and design were performed by J.S.-R., P.S., W.C.C., and A.R.; validation, investigation, data curation, and writing were performed by P.S., S.P., T.A.-I., A.R., A.S., M.K., J.S.-R., M.M.A., R.D., Y.T., S.M., S.D.D., T.B.E., and W.C.C.; review and editing were performed by J.S.-R., D.C., P.S., W.C.C., and A.R. and contributed to the final revision and major edits on the manuscript. All the authors read and approved the final manuscript.

References

  • 1.Scheau C., Caruntu C., Badarau I. A., et al. Cannabinoids and inflammations of the gut-lung-skin barrier. Journal of Personalized Medicine . 2021;11(6):p. 494. doi: 10.3390/jpm11060494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Tsoukalas D., Fragkiadaki P., Docea A., et al. Association of nutraceutical supplements with longer telomere length. International Journal of Molecular Medicine . 2019;44(1):218–226. doi: 10.3892/ijmm.2019.4191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Tsoukalas D., Zlatian O., Mitroi M., et al. A novel nutraceutical formulation can improve motor activity and decrease the stress level in a murine model of middle-age animals. Clinical Medicine . 2021;10(4):p. 624. doi: 10.3390/jcm10040624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Alshehri M. M., Quispe C., Herrera-Bravo J., et al. A review of recent studies on the antioxidant and anti-infectious properties of Senna plants. Oxidative Medicine and Cellular Longevity . 2022;2022:38. doi: 10.1155/2022/6025900.6025900 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Quetglas-Llabrés M. M., Quispe C., Herrera-Bravo J., et al. Pharmacological properties of Bergapten: mechanistic and therapeutic aspects. Oxidative Medicine and Cellular Longevity . 2022;2022:10. doi: 10.1155/2022/8615242.8615242 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 6.Sharifi-Rad J., Quispe C., Bouyahya A., et al. Ethnobotany, phytochemistry, biological activities, and health-promoting effects of the genus Bulbophyllum. Evidence-based Complementary and Alternative Medicine . 2022;2022:15. doi: 10.1155/2022/6727609.6727609 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Popović-Djordjević J., Quispe C., Giordo R., et al. Natural products and synthetic analogues against HIV: a perspective to develop new potential anti-HIV drugs. European Journal of Medicinal Chemistry . 2022;233:p. 114217. doi: 10.1016/j.ejmech.2022.114217. [DOI] [PubMed] [Google Scholar]
  • 8.Salehi B., Sestito S., Rapposelli S., et al. Epibatidine: a promising natural alkaloid in health. Biomolecules . 2019;9:p. 6. doi: 10.3390/biom9010006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Salehi B., Sharifi-Rad J., Capanoglu E., et al. Cucurbita plants: from farm to industry. Applied Sciences-Basel . 2019;9:p. 21. [Google Scholar]
  • 10.Taheri Y., Quispe C., Herrera-Bravo J., et al. Urtica dioica-derived phytochemicals for pharmacological and therapeutic applications. Evidence-based Complementary and Alternative Medicine . 2022;2022:30. doi: 10.1155/2022/4024331.4024331 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Newman D. J., Cragg G. M. Natural products as sources of new drugs over the nearly four decades from 01/1981 to 09/2019. Journal of Natural Products . 2020;83(3):770–803. doi: 10.1021/acs.jnatprod.9b01285. [DOI] [PubMed] [Google Scholar]
  • 12.Babaei F., Mirzababaei M., Nassiri-ASL M. Quercetin in food: possible mechanisms of its effect on memory. Journal of Food Science . 2018;83(9):2280–2287. doi: 10.1111/1750-3841.14317. [DOI] [PubMed] [Google Scholar]
  • 13.Sharifi-Rad J., Quispe C., Patra J. K., et al. Paclitaxel: application in modern oncology and nanomedicine-based cancer therapy. Oxidative Medicine and Cellular Longevity . 2021;2021:24. doi: 10.1155/2021/3687700.3687700 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Tu Y. Artemisinin—a gift from traditional Chinese medicine to the world (Nobel lecture) Angewandte Chemie International Edition . 2016;55(35):10210–10226. doi: 10.1002/anie.201601967. [DOI] [PubMed] [Google Scholar]
  • 15.Jesus M., Martins A. P., Gallardo E., Silvestre S. Diosgenin: recent highlights on pharmacology and analytical methodology. Journal of Analytical Methods in Chemistry . 2016;2016:16. doi: 10.1155/2016/4156293.4156293 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Yang S.-F., Weng C.-J., Sethi G., Hu D.-N. Natural bioactives and phytochemicals serve in cancer treatment and prevention. Evidence-based complementary and alternative medicine . 2013;2013 doi: 10.1155/2013/698190.698190 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Sethi G., Shanmugam M., Warrier S., et al. Pro-apoptotic and anti-cancer properties of diosgenin: a comprehensive and critical review. Nutrients . 2018;10(5):p. 645. doi: 10.3390/nu10050645. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Abbas G., Rauf K., Mahmood W. Saponins: the phytochemical with an emerging potential for curing clinical depression. Natural Product Research . 2015;29(4):302–307. doi: 10.1080/14786419.2014.942661. [DOI] [PubMed] [Google Scholar]
  • 19.Man S., Gao W., Zhang Y., Huang L., Liu C. Chemical study and medical application of saponins as anti-cancer agents. Fitoterapia . 2010;81(7):703–714. doi: 10.1016/j.fitote.2010.06.004. [DOI] [PubMed] [Google Scholar]
  • 20.Avula B., Wang Y. H., Ali Z., Smillie T. J., Khan I. A. Chemical fingerprint analysis and quantitative determination of steroidal compounds from Dioscorea villosa, Dioscorea species and dietary supplements using UHPLC-ELSD. Biomedical Chromatography . 2014;28(2):281–294. doi: 10.1002/bmc.3019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Deshpande H. A., Bhalsing S. R. Isolation and characterization of diosgenin from in vitro cultured tissues of Helicteres isora L. Physiology and Molecular Biology of Plants . 2014;20(1):89–94. doi: 10.1007/s12298-013-0205-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Yang F., Liang Y., Xu L., et al. Exploration in the cascade working mechanisms of liver injury induced by total saponins extracted from Rhizoma Dioscorea bulbifera. Biomedicine & Pharmacotherapy . 2016;83:1048–1056. doi: 10.1016/j.biopha.2016.08.017. [DOI] [PubMed] [Google Scholar]
  • 23.Yuan B., Byrnes D. R., Dinssa F. F., Simon J. E., Wu Q. Identification of polyphenols, glycoalkaloids, and saponins in Solanum scabrum berries using HPLC-UV/Vis-MS. Journal of Food Science . 2019;84(2):235–243. doi: 10.1111/1750-3841.14424. [DOI] [PubMed] [Google Scholar]
  • 24.Salehi B., Prakash Mishra A., Nigam M., et al. Ficus plants: state of the art from a phytochemical, pharmacological, and toxicological perspective. Phytotherapy Research . 2021;35(3):1187–1217. doi: 10.1002/ptr.6884. [DOI] [PubMed] [Google Scholar]
  • 25.Parama D., Boruah M., Kumari Y., et al. Diosgenin, a steroidal saponin, and its analogs: Effective therapies against different chronic diseases. Life Sciences . 2020;260:p. 118182. doi: 10.1016/j.lfs.2020.118182. [DOI] [PubMed] [Google Scholar]
  • 26.Cai B., Zhang Y., Wang Z., et al. Therapeutic potential of diosgenin and its major derivatives against neurological diseases: recent advances. Oxidative Medicine and Cellular Longevity . 2020;2020:16. doi: 10.1155/2020/3153082.3153082 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Kim J. E., Go J., Koh E. K., et al. Diosgenin effectively suppresses skin inflammation induced by phthalic anhydride in IL-4/Luc/CNS-1 transgenic mice. Bioscience, Biotechnology, and Biochemistry . 2016;80(5):891–901. doi: 10.1080/09168451.2015.1135040. [DOI] [PubMed] [Google Scholar]
  • 28.Kalailingam P., Kannaian B., Tamilmani E., Kaliaperumal R. Efficacy of natural diosgenin on cardiovascular risk, insulin secretion, and beta cells in streptozotocin (STZ)-induced diabetic rats. Phytomedicine . 2014;21(10):1154–1161. doi: 10.1016/j.phymed.2014.04.005. [DOI] [PubMed] [Google Scholar]
  • 29.Hua S., Li Y., Su L., Liu X. Diosgenin ameliorates gestational diabetes through inhibition of sterol regulatory element-binding protein-1. Biomedicine & Pharmacotherapy . 2016;84:1460–1465. doi: 10.1016/j.biopha.2016.10.049. [DOI] [PubMed] [Google Scholar]
  • 30.Lv Y.-C., Yang J., Yao F., et al. Diosgenin inhibits atherosclerosis via suppressing the MiR-19b-induced downregulation of ATP-binding cassette transporter A1. Atherosclerosis . 2015;240(1):80–89. doi: 10.1016/j.atherosclerosis.2015.02.044. [DOI] [PubMed] [Google Scholar]
  • 31.Chiang S.-S., Chang S.-P., Pan T.-M. Osteoprotective effect of Monascus-fermented dioscorea in ovariectomized rat model of postmenopausal osteoporosis. Journal of Agricultural and Food Chemistry . 2011;59(17):9150–9157. doi: 10.1021/jf201640j. [DOI] [PubMed] [Google Scholar]
  • 32.Huang B., du D., Zhang R., et al. Synthesis, characterization and biological studies of diosgenyl analogues. Bioorganic & Medicinal Chemistry Letters . 2012;22(24):7330–7334. doi: 10.1016/j.bmcl.2012.10.086. [DOI] [PubMed] [Google Scholar]
  • 33.Yan C., You-Mei T., Su-Lan Y., et al. Advances in the pharmacological activities and mechanisms of diosgenin. Chinese Journal of Natural Medicines . 2015;13(8):578–587. doi: 10.1016/S1875-5364(15)30053-4. [DOI] [PubMed] [Google Scholar]
  • 34.Jiang S., Fan J., Wang Q., et al. Diosgenin induces ROS-dependent autophagy and cytotoxicity via mTOR signaling pathway in chronic myeloid leukemia cells. Phytomedicine . 2016;23(3):243–252. doi: 10.1016/j.phymed.2016.01.010. [DOI] [PubMed] [Google Scholar]
  • 35.Yang H., Yin H., Shen Y., et al. A more ecological and efficient approach for producing diosgenin from Dioscorea zingiberensis tubers via pressurized biphase acid hydrolysis. Journal of Cleaner Production . 2016;131:10–19. doi: 10.1016/j.jclepro.2016.05.030. [DOI] [Google Scholar]
  • 36.Gan Q., Wang J., Hu J., et al. The role of diosgenin in diabetes and diabetic complications. The Journal of Steroid Biochemistry and Molecular Biology . 2020;198:p. 105575. doi: 10.1016/j.jsbmb.2019.105575. [DOI] [PubMed] [Google Scholar]
  • 37.Li X., Liu S., Qu L., et al. Dioscin and diosgenin: insights into their potential protective effects in cardiac diseases. Journal of Ethnopharmacology . 2021;274:p. 114018. doi: 10.1016/j.jep.2021.114018. [DOI] [PubMed] [Google Scholar]
  • 38.PLANTLIST, T. 2021, http://www.theplantlist.org/
  • 39.Heinrich M., Appendino G., Efferth T., et al. Best practice in research - overcoming common challenges in phytopharmacological research. Journal of Ethnopharmacology . 2020;246:p. 112230. doi: 10.1016/j.jep.2019.112230. [DOI] [PubMed] [Google Scholar]
  • 40.PUBCHEM. PubChem; 2021. Available: https://pubchem.ncbi.nlm.nih.gov/ [Google Scholar]
  • 41.Fujii K., Matsukawa T. Saponins and sterols. 8. Saponin of Dioscorea tokoro makino. Journal of the Pharmaceutical Society of Japan . 1936;56:408–414. [Google Scholar]
  • 42.Marker R. E., Wagner R. B., Ulshafer P. R., Wittbecker E. L., Goldsmith D. P. J., Ruof C. H. Sterols. CLVII. Sapogenins. LXIX.1Isolation and structures of thirteen new steroidal sapogenins. New sources for known sapogenins. Journal of the American Chemical Society . 1943;65(6):1199–1209. doi: 10.1021/ja01246a051. [DOI] [Google Scholar]
  • 43.Kang T. H., Moon E., Hong B. N., et al. Diosgenin from Dioscorea nipponica ameliorates diabetic neuropathy by inducing nerve growth factor. Biological and Pharmaceutical Bulletin . 2011;34(9):1493–1498. doi: 10.1248/bpb.34.1493. [DOI] [PubMed] [Google Scholar]
  • 44.Zhang Y., Tang L., An X., Fu E., Ma C. Modification of cellulase and its application to extraction of diosgenin from Dioscorea zingiberensis CH Wright. Biochemical Engineering Journal . 2009;47(1-3):80–86. doi: 10.1016/j.bej.2009.07.006. [DOI] [Google Scholar]
  • 45.Sánchez G. L., Acevedo J. C. M., Soto R. R. Spectrophotometric determination of diosgenin in Dioscorea composita following thin-layer chromatography. Analyst . 1972;97(1161):973–976. doi: 10.1039/an9729700973. [DOI] [PubMed] [Google Scholar]
  • 46.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 African Journal of Botany . 2021;138:386–393. doi: 10.1016/j.sajb.2020.12.027. [DOI] [Google Scholar]
  • 47.Semwal P., Painuli S., Cruz-Martins N. Dioscorea deltoidea wall. Ex Griseb: a review of traditional uses, bioactive compounds and biological activities. Food Bioscience . 2021;41, article 100969 doi: 10.1016/j.fbio.2021.100969. [DOI] [Google Scholar]
  • 48.Zhu Y., Zhu H., Qiu M., Zhu T., Ni J. Investigation on the mechanisms for biotransformation of saponins to diosgenin. World Journal of Microbiology and Biotechnology . 2014;30(1):143–152. doi: 10.1007/s11274-013-1429-7. [DOI] [PubMed] [Google Scholar]
  • 49.Arya P., Kumar P. Comparison of ultrasound and microwave assisted extraction of diosgenin from Trigonella foenum graceum seed. Ultrasonics Sonochemistry . 2021;74:p. 105572. doi: 10.1016/j.ultsonch.2021.105572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Selim S., Al Jaouni S. Anti-inflammatory, antioxidant and antiangiogenic activities of diosgenin isolated from traditional medicinal plant, Costus speciosus (Koen ex. Retz.) Sm. Natural Product Research . 2016;30(16):1830–1833. doi: 10.1080/14786419.2015.1065493. [DOI] [PubMed] [Google Scholar]
  • 51.Wang S., Wang F., Yang H., Li R., Guo H., Hu L. Diosgenin glucoside provides neuroprotection by regulating microglial M1 polarization. International Immunopharmacology . 2017;50:22–29. doi: 10.1016/j.intimp.2017.06.008. [DOI] [PubMed] [Google Scholar]
  • 52.Yin J., Yoon K. H., Hwang Y. J., Lee J. Y., Shin H. S., Lee M. W. Quantitative Analysis of (+)-Catechin and Diosgenin from Smilax china L. Rhizome. Korean Journal of Pharmacognosy . 2015;46:189–194. [Google Scholar]
  • 53.Chen L.-L., Verpoorte R., Yen H. R., et al. Effects of processing adjuvants on traditional Chinese herbs. Journal of Food and Drug Analysis . 2018;26(2):S96–S114. doi: 10.1016/j.jfda.2018.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Gupta D. D., Mishra S., Verma S. S., et al. Evaluation of antioxidant, anti-inflammatory and anticancer activities of diosgenin enriched Paris polyphylla rhizome extract of Indian Himalayan landraces. Journal of Ethnopharmacology . 2021;270:p. 113842. doi: 10.1016/j.jep.2021.113842. [DOI] [PubMed] [Google Scholar]
  • 55.Cheng J., Chen J., Liu X., et al. The origin and evolution of the diosgenin biosynthetic pathway in yam. Plant communications . 2021;2(1):p. 100079. doi: 10.1016/j.xplc.2020.100079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Souza C. M., Schwabe T. M. E., Pichler H., et al. A stable yeast strain efficiently producing cholesterol instead of ergosterol is functional for tryptophan uptake, but not weak organic acid resistance. Metabolic Engineering . 2011;13(5):555–569. doi: 10.1016/j.ymben.2011.06.006. [DOI] [PubMed] [Google Scholar]
  • 57.Dai Z., Liu Y., Sun Z., et al. Identification of a novel cytochrome P450 enzyme that catalyzes the C-2α hydroxylation of pentacyclic triterpenoids and its application in yeast cell factories. Metabolic Engineering . 2019;51:70–78. doi: 10.1016/j.ymben.2018.10.001. [DOI] [PubMed] [Google Scholar]
  • 58.Kirtonia A., Sethi G., Garg M. The multifaceted role of reactive oxygen species in tumorigenesis. Cellular and Molecular Life Sciences . 2020;77(22):4459–4483. doi: 10.1007/s00018-020-03536-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Mitrut P., Docea A. O., Kamal A. M., et al. Colorectal Cancer and Inflammatory Bowel Disease . Intechopen; 2016. [Google Scholar]
  • 60.Sharifi-Rad J., Quispe C., Durazzo A., et al. Resveratrol' biotechnological applications: enlightening its antimicrobial and antioxidant properties. Journal of Herbal Medicine . 2022;32:p. 100550. doi: 10.1016/j.hermed.2022.100550. [DOI] [Google Scholar]
  • 61.Chiu C.-S., Chiu Y. J., Wu L. Y., et al. Diosgenin ameliorates cognition deficit and attenuates oxidative damage in senescent mice induced by D-galactose. The American Journal of Chinese Medicine . 2011;39(3):551–563. doi: 10.1142/S0192415X11009020. [DOI] [PubMed] [Google Scholar]
  • 62.Turchan-Cholewo J., Liu Y., Gartner S., et al. Increased vulnerability of ApoE4 neurons to HIV proteins and opiates: protection by diosgenin and L-deprenyl. Neurobiology of Disease . 2006;23(1):109–119. doi: 10.1016/j.nbd.2006.02.005. [DOI] [PubMed] [Google Scholar]
  • 63.Wang L., et al. Trillium tschonoskii maxim saponin mitigates D-galactose-induced brain aging of rats through rescuing dysfunctional autophagy mediated by Rheb-mTOR signal pathway. Biomedicine & Pharmacotherapy . 2018;98:516–522. doi: 10.1016/j.biopha.2017.12.046. [DOI] [PubMed] [Google Scholar]
  • 64.Binesh A., Devaraj S. N., Halagowder D. Atherogenic diet induced lipid accumulation induced NFκB level in heart, liver and brain of Wistar rat and diosgenin as an anti-inflammatory agent. Life Sciences . 2018;196:28–37. doi: 10.1016/j.lfs.2018.01.012. [DOI] [PubMed] [Google Scholar]
  • 65.Cai B., Seong K. J., Bae S. W., et al. Water-soluble arginyl–diosgenin analog attenuates hippocampal neurogenesis impairment through blocking microglial activation underlying NF-κB and JNK MAPK signaling in adult mice challenged by LPS. Molecular Neurobiology . 2019;56(9):6218–6238. doi: 10.1007/s12035-019-1496-3. [DOI] [PubMed] [Google Scholar]
  • 66.Du L., Zhang Y., Chen Y., Zhu J., Yang Y., Zhang H.-L. Role of microglia in neurological disorders and their potentials as a therapeutic target. Molecular Neurobiology . 2017;54(10):7567–7584. doi: 10.1007/s12035-016-0245-0. [DOI] [PubMed] [Google Scholar]
  • 67.Yang R., Chen W., Lu Y., et al. Dioscin relieves endotoxemia induced acute neuro-inflammation and protect neurogenesis via improving 5-HT metabolism. Scientific Reports . 2017;7:1–13. doi: 10.1038/srep40035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Liu W., Zhu M., Yu Z., et al. Therapeutic effects of diosgenin in experimental autoimmune encephalomyelitis. Journal of Neuroimmunology . 2017;313:152–160. doi: 10.1016/j.jneuroim.2017.10.018. [DOI] [PubMed] [Google Scholar]
  • 69.Xiao L., Guo D., Hu C., et al. Diosgenin promotes oligodendrocyte progenitor cell differentiation through estrogen receptor-mediated ERK1/2 activation to accelerate remyelination. Glia . 2012;60(7):1037–1052. doi: 10.1002/glia.22333. [DOI] [PubMed] [Google Scholar]
  • 70.Chen X.-B., Wang Z. L., Yang Q. Y., et al. Diosgenin glucoside protects against spinal cord injury by regulating autophagy and alleviating apoptosis. International Journal of Molecular Sciences . 2018;19(8):p. 2274. doi: 10.3390/ijms19082274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Chen X.-B., Zhu M.-Y., Qin F.-R., et al. Effect of extract of Trillium tschonoskii Maxim on ciliary neurotropic factor and its receptor α in rats suffering from spinal cord injury. Medical Journal of Chinese People's Liberation Army . 2015;40:622–626. [Google Scholar]
  • 72.Zhu S., Tang S., Su F. Dioscin inhibits ischemic stroke-induced inflammation through inhibition of the TLR4/MyD88/NF-κB signaling pathway in a rat model. Molecular Medicine Reports . 2018;17(1):660–666. doi: 10.3892/mmr.2017.7900. [DOI] [PubMed] [Google Scholar]
  • 73.Kiasalari Z., Rahmani T., Mahmoudi N., Baluchnejadmojarad T., Roghani M. Diosgenin ameliorates development of neuropathic pain in diabetic rats: involvement of oxidative stress and inflammation. Biomedicine & Pharmacotherapy . 2017;86:654–661. doi: 10.1016/j.biopha.2016.12.068. [DOI] [PubMed] [Google Scholar]
  • 74.Lee B.-K., Kim C.-J., Shin M.-S., Cho Y. S. Diosgenin improves functional recovery from sciatic crushed nerve injury in rats. Journal of exercise rehabilitation . 2018;14(4):566–572. doi: 10.12965/jer.1836340.170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Zhao W. X., Wang P. F., Song H. G., Sun N. Diosgenin attenuates neuropathic pain in a rat model of chronic constriction injury. Molecular Medicine Reports . 2017;16(2):1559–1564. doi: 10.3892/mmr.2017.6723. [DOI] [PubMed] [Google Scholar]
  • 76.Lv H., Tian D. Designing and optimizing a parallel neural network model for predicting the solubility of diosgenin in n-alkanols. Chinese Journal of Chemical Engineering . 2021;29:288–294. doi: 10.1016/j.cjche.2020.09.009. [DOI] [Google Scholar]
  • 77.Zhang X., Wang X., Xue Z., Zhan G., Ito Y., Guo Z. Prevention properties on cerebral ischemia reperfusion of medicine food homologous Dioscorea yam-derived diosgenin based on mediation of potential targets. Food Chemistry . 2021;345:p. 128672. doi: 10.1016/j.foodchem.2020.128672. [DOI] [PubMed] [Google Scholar]
  • 78.Aaron P. A., Vu K., Gelli A. An Antivirulence approach for preventing Cryptococcus neoformans from crossing the blood-brain barrier via novel natural product inhibitors of a fungal metalloprotease. MBio . 2020;11(4) doi: 10.1128/mBio.01249-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Cheng S. M., Ho Y. J., Yu S. H., et al. Anti-apoptotic effects of diosgenin in D-galactose-induced aging brain. The American Journal of Chinese Medicine . 2020;48(2):391–406. doi: 10.1142/S0192415X20500202. [DOI] [PubMed] [Google Scholar]
  • 80.Leng J., Li X., Tian H., et al. Neuroprotective effect of diosgenin in a mouse model of diabetic peripheral neuropathy involves the Nrf2/HO-1 pathway. BMC complementary medicine and therapies . 2020;20(1):p. 126. doi: 10.1186/s12906-020-02930-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Oyelaja-Akinsipo O. B., Dare E. O., Katare D. P. Protective role of diosgenin against hyperglycaemia-mediated cerebral ischemic brain injury in zebrafish model of type II diabetes mellitus. Heliyon . 2020;6(1):p. e03296. doi: 10.1016/j.heliyon.2020.e03296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Rajesh Y., Biswas A., Kumar U., et al. Targeting NFE2L2, a transcription factor upstream of MMP-2: A potential therapeutic strategy for temozolomide resistant glioblastoma. Biochemical Pharmacology . 2019;164:1–16. doi: 10.1016/j.bcp.2019.03.025. [DOI] [PubMed] [Google Scholar]
  • 83.Tohda C., Urano T., Umezaki M., Nemere I., Kuboyama T. Diosgenin is an exogenous activator of 1,25D3-MARRS/Pdia3/ERp57 and improves Alzheimer's disease pathologies in 5XFAD mice. Scientific Reports . 2012;2(1):p. 535. doi: 10.1038/srep00535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Tohda C., Lee Y.-A., Goto Y., Nemere I. Diosgenin-induced cognitive enhancement in normal mice is mediated by 1,25D3-MARRS. Scientific Reports . 2013;3(1):p. 3395. doi: 10.1038/srep03395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Koh E.-K., Yun W. B., Kim J. E., et al. Beneficial effect of diosgenin as a stimulator of NGF on the brain with neuronal damage induced by Aβ-42 accumulation and neurotoxicant injection. Laboratory Animal Research . 2016;32(2):105–115. doi: 10.5625/lar.2016.32.2.105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Chiu C.-S., Deng J. S., Hsieh M. T., et al. Yam (Dioscorea pseudojaponica Yamamoto) ameliorates cognition deficit and attenuates oxidative damage in senescent mice induced by D-galactose. The American Journal of Chinese Medicine . 2009;37(5):889–902. doi: 10.1142/S0192415X09007296. [DOI] [PubMed] [Google Scholar]
  • 87.Zhang X., Xue X., Zhao J., et al. Diosgenin attenuates the brain injury induced by transient focal cerebral ischemia-reperfusion in rats. Steroids . 2016;113:103–112. doi: 10.1016/j.steroids.2016.07.006. [DOI] [PubMed] [Google Scholar]
  • 88.Chojnacki J. E., Liu K., Saathoff J. M., Zhang S. Bivalent ligands incorporating curcumin and diosgenin as multifunctional compounds against Alzheimer's disease. Bioorganic & Medicinal Chemistry . 2015;23(22):7324–7331. doi: 10.1016/j.bmc.2015.10.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Li B., Xu P., Wu S., et al. Diosgenin attenuates lipopolysaccharide-induced Parkinson’s disease by inhibiting the TLR/NF-κB pathway. Journal of Alzheimer's Disease . 2018;64(3):943–955. doi: 10.3233/JAD-180330. [DOI] [PubMed] [Google Scholar]
  • 90.Yang G. X., Huang Y., Zheng L. L., et al. Design, synthesis and evaluation of diosgenin carbamate derivatives as multitarget anti-Alzheimer’s disease agents. European Journal of Medicinal Chemistry . 2020;187:p. 111913. doi: 10.1016/j.ejmech.2019.111913. [DOI] [PubMed] [Google Scholar]
  • 91.Cai D., Qi J., Yang Y., et al. Design, synthesis and biological evaluation of diosgenin-amino acid derivatives with dual functions of neuroprotection and angiogenesis. Molecules . 2019;24(22):p. 4025. doi: 10.3390/molecules24224025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.GBD 2019 Colorectal Cancer Collaborators. Global, regional, and national burden of colorectal cancer and its risk factors, 1990-2019: a systematic analysis for the Global Burden of Disease Study 2019. The Lancet Gastroenterology & Hepatology . 2022:p. S2468. doi: 10.1016/S2468-1253(22)00044-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Sharifi-Rad J., Quispe C., Butnariu M., et al. Chitosan nanoparticles as a promising tool in nanomedicine with particular emphasis on oncological treatment. Cancer Cell International . 2021;21(1):318–318. doi: 10.1186/s12935-021-02025-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Zahra K. F., Lefter R., Ali A., et al. The involvement of the oxidative stress status in cancer pathology: a double view on the role of the antioxidants. Oxidative Medicine and Cellular Longevity . 2021;2021:25. doi: 10.1155/2021/9965916.9965916 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Docea A. O., Mitruţ P., Grigore D., Pirici D., Călina D. C., Gofiţă E. Immunohistochemical expression of TGF beta (TGF-β), TGF beta receptor 1 (TGFBR1), and Ki67 in intestinal variant of gastric adenocarcinomas. Romanian Journal of Morphology and Embryology . 2012;53(3 Supplement):683–692. [PubMed] [Google Scholar]
  • 96.Zlatian O. M., Comanescu M. V., Rosu A. F., et al. Histochemical and immunohistochemical evidence of tumor heterogeneity in colorectal cancer. Romanian Journal of Morphology and Embryology . 2015;56:175–181. [PubMed] [Google Scholar]
  • 97.Stefanowicz-Hajduk J., Król-Kogus B., Sparzak-Stefanowska B., Kimel K., Ochocka J. R., Krauze-Baranowska M. Cytotoxic activity of standardized extracts, a fraction, and individual secondary metabolites from fenugreek seeds against SKOV-3, HeLa and MOLT-4 cell lines. Pharmaceutical Biology . 2021;59(1):422–435. doi: 10.1080/13880209.2021.1903047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Ma L., Zhang J., Wang X., et al. Design and synthesis of diosgenin derivatives as apoptosis inducers through mitochondria-related pathways. European Journal of Medicinal Chemistry . 2021;217:p. 113361. doi: 10.1016/j.ejmech.2021.113361. [DOI] [PubMed] [Google Scholar]
  • 99.Yin H., Zhang M. J., An R. F., et al. Diosgenin derivatives as potential antitumor agents: synthesis, cytotoxicity, and mechanism of action. Journal of Natural Products . 2021;84(3):616–629. doi: 10.1021/acs.jnatprod.0c00698. [DOI] [PubMed] [Google Scholar]
  • 100.Martínez-Gallegos A. A., Guerrero-Luna G., Ortiz-González A., Cárdenas-García M., Bernès S., Hernández-Linares M. G. Azasteroids from diosgenin: synthesis and evaluation of their antiproliferative activity. Steroids . 2021;166:p. 108777. doi: 10.1016/j.steroids.2020.108777. [DOI] [PubMed] [Google Scholar]
  • 101.Liu Y., Zhou Z., Yan J., Wu X., Xu G. Diosgenin exerts antitumor activity via downregulation of Skp2 in breast cancer cells. BioMed Research International . 2020;2020 doi: 10.1155/2020/8072639.8072639 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Puar Y. R., Shanmugam M. K., Fan L., Arfuso F., Sethi G., Tergaonkar V. Evidence for the involvement of the master transcription factor NF-κB in cancer initiation and progression. Biomedicine . 2018;6:p. 82. doi: 10.3390/biomedicines6030082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Liao W.-L., Lin J.-Y., Shieh J.-C., et al. Induction of G2/M phase arrest by diosgenin via activation of Chk1 kinase and Cdc25C regulatory pathways to promote apoptosis in human breast cancer cells. International Journal of Molecular Sciences . 2020;21:p. 172. doi: 10.3390/ijms21010172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.He Z., Chen H., Li G., et al. Diosgenin inhibits the migration of human breast cancer MDA-MB-231 cells by suppressing Vav2 activity. Phytomedicine . 2014;21(6):871–876. doi: 10.1016/j.phymed.2014.02.002. [DOI] [PubMed] [Google Scholar]
  • 105.Chun J., Han L., Xu M. Y., Wang B., Cheng M. S., Kim Y. S. The induction of apoptosis by a newly synthesized diosgenyl saponin through the suppression of estrogen receptor-α in MCF-7 human breast cancer cells. Archives of Pharmacal Research . 2014;37(11):1477–1486. doi: 10.1007/s12272-013-0279-z. [DOI] [PubMed] [Google Scholar]
  • 106.Aumsuwan P., Khan S. I., Khan I. A., et al. The anticancer potential of steroidal saponin, dioscin, isolated from wild yam (Dioscorea villosa) root extract in invasive human breast cancer cell line MDA-MB-231 in vitro. Archives of Biochemistry and Biophysics . 2016;591:98–110. doi: 10.1016/j.abb.2015.12.001. [DOI] [PubMed] [Google Scholar]
  • 107.Kim E.-A., Jang J. H., Lee Y. H., et al. Dioscin induces caspase-independent apoptosis through activation of apoptosis-inducing factor in breast cancer cells. Apoptosis . 2014;19(7):1165–1175. doi: 10.1007/s10495-014-0994-z. [DOI] [PubMed] [Google Scholar]
  • 108.Srinivasan S., Koduru S., Kumar R., Venguswamy G., Kyprianou N., Damodaran C. Diosgenin targets Akt-mediated prosurvival signaling in human breast cancer cells. International Journal of Cancer . 2009;125(4):961–967. doi: 10.1002/ijc.24419. [DOI] [PubMed] [Google Scholar]
  • 109.Mao X.-M., Zhou P., Li S. Y., et al. Diosgenin suppresses cholangiocarcinoma cells via inducing cell cycle arrest and mitochondria-mediated apoptosis. Oncotargets and Therapy . 2019;Volume 12:9093–9104. doi: 10.2147/OTT.S226261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Lepage C., Liagre B., Cook-Moreau J., Pinon A., Beneytout J.-L. Cyclooxygenase-2 and 5-lipoxygenase pathways in diosgenin-induced apoptosis in HT-29 and HCT-116 colon cancer cells. International Journal of Oncology . 2010;36(5):1183–1191. doi: 10.3892/ijo_00000601. [DOI] [PubMed] [Google Scholar]
  • 111.Chen H., Xu L., Yin L., et al. iTRAQ-based proteomic analysis of dioscin on human HCT-116 colon cancer cells. Proteomics . 2014;14(1):51–73. doi: 10.1002/pmic.201300101. [DOI] [PubMed] [Google Scholar]
  • 112.Hernández-Vázquez J. M. V., López-Muñoz H., Escobar-Sánchez M. L., et al. Apoptotic, necrotic, and antiproliferative activity of diosgenin and diosgenin glycosides on cervical cancer cells. European Journal of Pharmacology . 2020;871:p. 172942. doi: 10.1016/j.ejphar.2020.172942. [DOI] [PubMed] [Google Scholar]
  • 113.Zhao X., Tao X., Xu L., et al. Dioscin induces apoptosis in human cervical carcinoma HeLa and SiHa cells through ROS-mediated DNA damage and the mitochondrial signaling pathway. Molecules . 2016;21(6):p. 730. doi: 10.3390/molecules21060730. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Cai J., Liu M., Wang Z., Ju Y. Apoptosis induced by dioscin in Hela cells. Biological and Pharmaceutical Bulletin . 2002;25(2):193–196. doi: 10.1248/bpb.25.193. [DOI] [PubMed] [Google Scholar]
  • 115.Ma Y.-L., Zhang Y. S., Zhang F., et al. Methyl protodioscin from Polygonatum sibiricum inhibits cervical cancer through cell cycle arrest and apoptosis induction. Food and Chemical Toxicology . 2019;132:p. 110655. doi: 10.1016/j.fct.2019.110655. [DOI] [PubMed] [Google Scholar]
  • 116.Zhiyu W., Yue C., Neng W., et al. Dioscin induces cancer cell apoptosis through elevated oxidative stress mediated by downregulation of peroxiredoxins. Cancer Biology & Therapy . 2012;13(3):138–147. doi: 10.4161/cbt.13.3.18693. [DOI] [PubMed] [Google Scholar]
  • 117.Song X., Wang Z., Liang H., et al. Dioscin induces gallbladder cancer apoptosis by inhibiting ROS-mediated PI3K/AKT Signalling. International Journal of Biological Sciences . 2017;13(6):782–793. doi: 10.7150/ijbs.18732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Zhao X., Xu L., Zheng L., et al. Potent effects of dioscin against gastric cancer in vitro and in vivo. Phytomedicine . 2016;23(3):274–282. doi: 10.1016/j.phymed.2016.01.012. [DOI] [PubMed] [Google Scholar]
  • 119.Hu M., Xu L., Yin L., et al. Cytotoxicity of dioscin in human gastric carcinoma cells through death receptor and mitochondrial pathways. Journal of Applied Toxicology . 2013;33(8):712–722. doi: 10.1002/jat.2715. [DOI] [PubMed] [Google Scholar]
  • 120.Ma T., Wang R.-P., Zou X. Dioscin inhibits gastric tumor growth through regulating the expression level of lncRNA HOTAIR. BMC Complementary and Alternative Medicine . 2016;16(1):p. 383. doi: 10.1186/s12906-016-1360-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Lv L., Zheng L., Dong D., et al. Dioscin, a natural steroid saponin, induces apoptosis and DNA damage through reactive oxygen species: A potential new drug for treatment of glioblastoma multiforme. Food and Chemical Toxicology . 2013;59:657–669. doi: 10.1016/j.fct.2013.07.012. [DOI] [PubMed] [Google Scholar]
  • 122.Chen Z., Xu J., Wu Y., et al. Diosgenin inhibited the expression of TAZ in hepatocellular carcinoma. Biochemical and Biophysical Research Communications . 2018;503(3):1181–1185. doi: 10.1016/j.bbrc.2018.07.022. [DOI] [PubMed] [Google Scholar]
  • 123.Yu H., Liu Y., Niu C., Cheng Y. Diosgenin increased DDX3 expression in hepatocellular carcinoma. American Journal of Translational Research . 2018;10(11):3590–3599. [PMC free article] [PubMed] [Google Scholar]
  • 124.Kim D. S., Jeon B. K., Lee Y. E., Woo W. H., Mun Y. J. Diosgenin induces apoptosis in HepG2 cells through generation of reactive oxygen species and mitochondrial pathway. Evidence-based Complementary and Alternative Medicine . 2012;2012:8. doi: 10.1155/2012/981675.981675 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Zhang G., Zeng X., Zhang R., et al. Dioscin suppresses hepatocellular carcinoma tumor growth by inducing apoptosis and regulation of TP53, BAX, BCL2 and cleaved CASP3. Phytomedicine . 2016;23(12):1329–1336. doi: 10.1016/j.phymed.2016.07.003. [DOI] [PubMed] [Google Scholar]
  • 126.Wang G., Chen H., Huang M., et al. Methyl protodioscin induces G2/M cell cycle arrest and apoptosis in HepG2 liver cancer cells. Cancer Letters . 2006;241(1):102–109. doi: 10.1016/j.canlet.2005.10.050. [DOI] [PubMed] [Google Scholar]
  • 127.Nie C., Zhou J., Qin X., et al. Diosgenin-induced autophagy and apoptosis in a human prostate cancer cell line. Molecular Medicine Reports . 2016;14(5):4349–4359. doi: 10.3892/mmr.2016.5750. [DOI] [PubMed] [Google Scholar]
  • 128.Chang H.-Y., Kao M. C., Way T. D., Ho C. T., Fu E. Diosgenin suppresses hepatocyte growth factor (HGF)-induced epithelial–mesenchymal transition by down-regulation of Mdm2 and vimentin. Journal of Agricultural and Food Chemistry . 2011;59(10):5357–5363. doi: 10.1021/jf200598w. [DOI] [PubMed] [Google Scholar]
  • 129.Chen P.-S., Shih Y. W., Huang H. C., Cheng H. W. Diosgenin, a steroidal saponin, inhibits migration and invasion of human prostate cancer PC-3 cells by reducing matrix metalloproteinases expression. PLoS One . 2011;6(5) doi: 10.1371/journal.pone.0020164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Zhang J., Xie J.-J., Zhou S.-J., et al. Diosgenin inhibits the expression of NEDD4 in prostate cancer cells. American Journal of Translational Research . 2019;11:3461–3471. [PMC free article] [PubMed] [Google Scholar]
  • 131.Sun G. C., Jan C. R., Liang W. Z. Exploring the impact of a naturally occurring sapogenin diosgenin on underlying mechanisms of Ca2+ movement and cytotoxicity in human prostate cancer cells. Environmental Toxicology . 2020;35(3):395–403. doi: 10.1002/tox.22876. [DOI] [PubMed] [Google Scholar]
  • 132.Calina D., Buga A. M., Mitroi M., et al. The treatment of cognitive, behavioural and motor impairments from brain injury and neurodegenerative diseases through cannabinoid system modulation-evidence from in vivo studies. Journal of Clinical Medicine . 2020;9(8):p. 2395. doi: 10.3390/jcm9082395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Hossain R., Quispe C., Herrera-Bravo J., et al. Neurobiological promises of the bitter diterpene lactone andrographolide. Oxidative Medicine and Cellular Longevity . 2022;2022:9. doi: 10.1155/2022/3079577.3079577 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Mavroudis I., Kazis D., Chowdhury R., et al. Post-Concussion Syndrome and Chronic Traumatic Encephalopathy: Narrative Review on the Neuropathology, Neuroimaging and Fluid Biomarkers. Diagnostics . 2022;12(3):p. 740. doi: 10.3390/diagnostics12030740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Buga A. M., Docea A. O., Albu C., et al. Molecular and cellular stratagem of brain metastases associated with melanoma. Oncology Letters . 2019;17(5):4170–4175. doi: 10.3892/ol.2019.9933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Ciobica A., Padurariu M., Curpan A., et al. Minireview on the connections between the neuropsychiatric and dental disorders: current perspectives and the possible relevance of oxidative stress and other factors. Oxidative Medicine and Cellular Longevity . 2020;2020:13. doi: 10.1155/2020/6702314.6702314 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Siokas V., Aloizou A. M., Tsouris Z., et al. ADORA2A rs5760423 and CYP1A2 rs762551 polymorphisms as risk factors for Parkinson's disease. Journal of Clinical Medicine . 2021;10(3):p. 381. doi: 10.3390/jcm10030381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Binesh A., Devaraj S. N., Devaraj H. Expression of chemokines in macrophage polarization and downregulation of NFκB in aorta allow macrophage polarization by diosgenin in atherosclerosis. Journal of Biochemical and Molecular Toxicology . 2020;34:p. e22422. doi: 10.1002/jbt.22422. [DOI] [PubMed] [Google Scholar]
  • 139.Wang P., He L.-Y., Shen G.-D., Li R.-L., Yang J.-L. Inhibitory effects of Dioscin on atherosclerosis and foam cell formation in hyperlipidemia rats. Inflammopharmacology . 2017;25(6):633–642. doi: 10.1007/s10787-017-0341-4. [DOI] [PubMed] [Google Scholar]
  • 140.Yang Q., Wang C., Jin Y., et al. 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. Pharmacological Research . 2019;148:p. 104414. doi: 10.1016/j.phrs.2019.104414. [DOI] [PubMed] [Google Scholar]
  • 141.Junchao Y., Zhen W., Yuan W., et al. Anti- trachea inflammatory effects of diosgenin from Dioscorea nipponica through interactions with glucocorticoid receptor α. Journal of International Medical Research . 2016;45:101–113. doi: 10.1177/0300060516676724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Cioboată R., Găman A., Traşcă D., et al. Pharmacological management of non-alcoholic fatty liver disease: atorvastatin versus pentoxifylline. Experimental and Therapeutic Medicine . 2017;13(5):2375–2381. doi: 10.3892/etm.2017.4256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Docea A. O., Gofita E., Calina D., Ioan Z. S., Valcea D. I., Mitrut P. Autoimmune disorders due to double antiviral therapy with peginterferon and ribavirin in patients with hepatitis C virus infection. Farmácia . 2016;64:605–611. [Google Scholar]
  • 144.Tsoukalas D., Fragoulakis V., Sarandi E., et al. Targeted metabolomic analysis of serum fatty acids for the prediction of autoimmune diseases. Frontiers in Molecular Biosciences . 2019;6 doi: 10.3389/fmolb.2019.00120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Jain D., Chaudhary P., Varshney N., et al. Tobacco smoking and liver cancer risk: potential avenues for carcinogenesis. Journal of Oncology . 2021;2021:11. doi: 10.1155/2021/5905357.5905357 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Painuli S., Quispe C., Herrera-Bravo J., et al. Nutraceutical Profiling, Bioactive Composition, and Biological Applications of Lepidium sativum L. Oxidative Medicine and Cellular Longevity . 2022;2022:20. doi: 10.1155/2022/2910411.2910411 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Xie W. L., Jiang R., Shen X. L., Chen Z. Y., Deng X. M. Diosgenin attenuates hepatic stellate cell activation through transforming growth factor-β/Smad signaling pathway. International Journal of Clinical and Experimental Medicine . 2015;8(11):20323–20329. [PMC free article] [PubMed] [Google Scholar]
  • 148.Liu M., Xu Y., Han X., et al. Dioscin alleviates alcoholic liver fibrosis by attenuating hepatic stellate cell activation via the TLR4/MyD88/NF-κB signaling pathway. Scientific Reports . 2015;5:18038–18038. doi: 10.1038/srep18038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Zhou H. T., Yu X. F., Zhou G. M. Diosgenin inhibits angiotensin II-induced extracellular matrix remodeling in cardiac fibroblasts through regulating the TGF-β1/Smad3 signaling pathway. Molecular Medicine Reports . 2017;15(5):2823–2828. doi: 10.3892/mmr.2017.6280. [DOI] [PubMed] [Google Scholar]
  • 150.Gao M., Chen L., Yu H., Sun Q., Kou J., Yu B. Diosgenin down-regulates NF-κB p65/p50 and p38MAPK pathways and attenuates acute lung injury induced by lipopolysaccharide in mice. International Immunopharmacology . 2013;15(2):240–245. doi: 10.1016/j.intimp.2012.11.019. [DOI] [PubMed] [Google Scholar]
  • 151.Cai H., Wang Z., Zhang H. Q., et al. Diosgenin relieves goiter via the inhibition of thyrocyte proliferation in a mouse model of Graves' disease. Acta Pharmacologica Sinica . 2014;35(1):65–73. doi: 10.1038/aps.2013.133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Saravanan G., Ponmurugan P., Deepa M., Senthilkumar B. Modulatory effects of diosgenin on attenuating the key enzymes activities of carbohydrate metabolism and glycogen content in streptozotocin-induced diabetic rats. Canadian Journal of Diabetes . 2014;38(6):409–414. doi: 10.1016/j.jcjd.2014.02.004. [DOI] [PubMed] [Google Scholar]
  • 153.Ghosh S., More P., Derle A., et al. Diosgenin from Dioscorea bulbifera: novel hit for treatment of type II diabetes mellitus with inhibitory activity against α-amylase and α-glucosidase. PLoS One . 2014;9(9):p. e106039. doi: 10.1371/journal.pone.0106039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Zhang Z., Song C., Fu X., et al. High-dose diosgenin reduces bone loss in ovariectomized rats via attenuation of the RANKL/OPG ratio. International Journal of Molecular Sciences . 2014;15(9):17130–17147. doi: 10.3390/ijms150917130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Liu J., He X., Zhen P., Zhou S., Li X. Protective effect of diosgenin on chondrocytes mediated by JAK2/STAT3 signaling pathway in mice with osteoarthritis. Zhejiang da xue xue bao. Yi xue ban= Journal of Zhejiang University. Medical Science . 2016;45:453–460. doi: 10.3785/j.issn.1008-9292.2016.09.02. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Chen C.-T., Wang Z. H., Hsu C. C., Lin H. H., Chen J. H. In vivo protective effects of diosgenin against doxorubicin-induced cardiotoxicity. Nutrients . 2015;7(6):4938–4954. doi: 10.3390/nu7064938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Cai T., Cocci A., Cito G., et al. The role of diallyl thiosulfinate associated with nuciferine and diosgenin in the treatment of premature ejaculation: A pilot study. Archivio Italiano di Urologia, Andrologia . 2018;90(1):59–64. doi: 10.4081/aiua.2018.1.59. [DOI] [PubMed] [Google Scholar]
  • 158.Tohda C., Yang X., Matsui M., et al. Diosgenin-rich yam extract enhances cognitive function: a placebo-controlled, randomized, double-blind, crossover study of healthy adults. Nutrients . 2017;9(10):p. 1160. doi: 10.3390/nu9101160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Nouri K., Walch K., Weghofer A., Imhof M., Egarter C., Ott J. The impact of a standardized oral multinutrient supplementation on embryo quality in in vitro fertilization/intracytoplasmic sperm injection: a prospective randomized trial. Gynecologic and Obstetric Investigation . 2017;82(1):8–14. doi: 10.1159/000452662. [DOI] [PubMed] [Google Scholar]
  • 160.Komesaroff P. A., Black C. V., Cable V., Sudhir K. Effects of wild yam extract on menopausal symptoms, lipids and sex hormones in healthy menopausal women. Climacteric . 2001;4(2):144–150. doi: 10.1080/cmt.4.2.144.150. [DOI] [PubMed] [Google Scholar]
  • 161.Qin Y., Wu X., Huang W., et al. Acute toxicity and sub-chronic toxicity of steroidal saponins from Dioscorea zingiberensis CH Wright in rodents. Journal of Ethnopharmacology . 2009;126(3):543–550. doi: 10.1016/j.jep.2009.08.047. [DOI] [PubMed] [Google Scholar]
  • 162.Zheng H., Wei Z., Xin G., et al. Preventive effect of a novel diosgenin derivative on arterial and venous thrombosis in vivo. Bioorganic & Medicinal Chemistry Letters . 2016;26(14):3364–3369. doi: 10.1016/j.bmcl.2016.05.032. [DOI] [PubMed] [Google Scholar]
  • 163.Manda V. K., Avula B., Ali Z., et al. Characterization of in vitro ADME properties of diosgenin and dioscin from Dioscorea villosa. Planta Medica . 2013;79:p. 1421. doi: 10.1055/s-0033-1350699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Kaskiw M. J., Tassotto M. L., Mok M., et al. Structural analogues of diosgenyl saponins: synthesis and anticancer activity. Bioorganic & Medicinal Chemistry . 2009;17(22):7670–7679. doi: 10.1016/j.bmc.2009.09.046. [DOI] [PubMed] [Google Scholar]
  • 165.Raju J., Mehta R. Cancer chemopreventive and therapeutic effects of diosgenin, a food saponin. Nutrition and Cancer . 2009;61(1):27–35. doi: 10.1080/01635580802357352. [DOI] [PubMed] [Google Scholar]
  • 166.Kanchan D. M., Somani G. S., Peshattiwar V. V., Kaikini A. A., Sathaye S. Renoprotective effect of diosgenin in streptozotocin induced diabetic rats. Pharmacological Reports . 2016;68(2):370–377. doi: 10.1016/j.pharep.2015.10.011. [DOI] [PubMed] [Google Scholar]
  • 167.Khosravi Z., Sedaghat R., Baluchnejadmojarad T., Roghani M. Diosgenin ameliorates testicular damage in streptozotocin-diabetic rats through attenuation of apoptosis, oxidative stress, and inflammation. International Immunopharmacology . 2019;70:37–46. doi: 10.1016/j.intimp.2019.01.047. [DOI] [PubMed] [Google Scholar]
  • 168.Roghani-Dehkordi F., Roghani M., Baluchnejadmojarad T. Diosgenin mitigates Streptozotocin diabetes-induced vascular dysfunction of the rat aorta: the involved mechanisms. Journal of Cardiovascular Pharmacology . 2015;66(6):584–592. doi: 10.1097/FJC.0000000000000308. [DOI] [PubMed] [Google Scholar]
  • 169.Sato K., Fujita S., Iemitsu M. Acute administration of diosgenin or dioscorea improves hyperglycemia with increases muscular steroidogenesis in STZ-induced type 1 diabetic rats. The Journal of Steroid Biochemistry and Molecular Biology . 2014;143:152–159. doi: 10.1016/j.jsbmb.2014.02.020. [DOI] [PubMed] [Google Scholar]
  • 170.Rabha B., Bharadwaj K. K., Baishya D., Sarkar T., Edinur H. A., Pati S. Synthesis and characterization of diosgenin encapsulated poly-ε-caprolactone-pluronic nanoparticles and its effect on brain cancer cells. Polymers . 2021;13(8):p. 1322. doi: 10.3390/polym13081322. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

The data used to support the findings of this study are available from the corresponding author upon request.


Articles from Oxidative Medicine and Cellular Longevity are provided here courtesy of Wiley

RESOURCES