Fact-checked by Grok 2 weeks ago

Protodioscin

Protodioscin is a naturally occurring steroidal classified as a furostanol , with the molecular formula C₅₁H₈₄O₂₂, found in various plant species including , species (such as Dioscorea nipponica and Dioscorea collettii), fenugreek (Trigonella foenum-graecum), and Trillium govanianum. Chemically, protodioscin features a furostanol aglycone backbone linked to a trisaccharide chain at the 3-OH position and a glucose at the 26-OH group, distinguishing it from related compounds like dioscin and methylprotodioscin. It is a major bioactive constituent in , where its concentration can reach up to 1530 mg/100 g in aerial parts, varying by plant part, geographic origin, and extraction method. In , protodioscin-rich extracts from plants like Polygonatum sibiricum have been used in Chinese herbal formulations for their purported tonic and restorative effects. Protodioscin has garnered significant research interest for its pharmacological activities, including anticancer effects through induction of in tumor cells (e.g., HL-60 cells with IC₅₀ values of 1.9–6.8 μg/mL) and upregulation of stress pathways like JNK/p38 in models. It also demonstrates properties by promoting mucosal healing and epithelial proliferation in models of intestinal inflammation, as well as anti-hyperlipidemic effects by reducing triglycerides, , and LDL levels in hyperlipidemic rats. Additionally, protodioscin exhibits activity via free radical scavenging and has been linked to improved in animal studies through enhanced testosterone production and pathways, though human clinical trials show mixed results on testosterone elevation and erectile function improvement. Other reported benefits include antidiabetic effects via better and , activity, and potential cardiovascular protection through increased expression for reverse transport. Despite these promising activities, protodioscin's low in some formulations remains a challenge for therapeutic applications.

Chemistry

Structure and Formula

Protodioscin has the C₅₁H₈₄O₂₂ and a of 1,049.2 g/mol. It is a furostanol featuring a steroidal aglycone with a furostane core and an open . The consists of the trisaccharide chain α-L-Rha-(1→4)-[α-L-Rha-(1→2)]-β-D-Glc attached at position 3 of the aglycone (25R)-26-[(β-D-glucopyranosyl)oxy]-3β,5,6β-trihydroxyfurost-5-ene, along with a 26-O-β-D-Glc moiety on the furostanol . The aglycone core resembles diosgenin but exists in the furostanol form with hydroxyl groups at positions 3β, 5, and 6β, forming the base for the glycosidic attachments; the sugar moieties include two units branching on a central glucose at the 3-position and a terminal glucose at the 26-position of the . This arrangement creates a complex structure where the nucleus (rings A-D) connects to the branched , enhancing its amphiphilic properties typical of . A common synonym for protodioscin is (3β,5,6β)-26-(β-D-glucopyranosyloxy)-22-hydroxyfurost-5-ene-3-yl O-α-L-rhamnopyranosyl-(1→2)-β-D-glucopyranosyl-(1→4)-O-[α-L-rhamnopyranosyl-(1→2)]-β-D-glucopyranoside.

Physical and Chemical Properties

Protodioscin is typically isolated as a white to off-white crystalline . This appearance facilitates its handling in laboratory settings and pharmaceutical preparations. Its physical form derives from the steroidal structure, which contributes to its solid state at room temperature. The compound exhibits good solubility in polar organic solvents, including approximately 10 mg/mL in , 20 mg/mL in DMSO, and 5 mg/mL in , but shows poor in , with values around 0.3 mg/mL in aqueous (pH 7.2) when initially dissolved in DMSO. It is slightly soluble in and . The of protodioscin is reported as 190–192 °C. Protodioscin demonstrates stability under neutral conditions and can be stored for at least four years at -20 °C. However, it is sensitive to , a process that cleaves the glycosidic bonds linking the moieties to the aglycone, ultimately yielding diosgenin as the primary product. The specific [α]_D is approximately -75.3° (c = 0.61, ) to -88.4° (c = 0.01, ). For identification and structural confirmation, protodioscin displays characteristic spectroscopic features. In (ESI-MS), it exhibits a prominent deprotonated molecular peak at m/z 1049 [M-H]^-. (NMR) data in pyridine-d_5 reveal key signals, such as δ_C 100.2 for the anomeric carbon of the 3-O-glucose and δ_H 4.96 (d, J = 6.60 Hz) for its proton, aiding in the verification of the trisaccharide chain attached to the furostanol core.

Occurrence

Natural Sources

Protodioscin is a steroidal primarily isolated from the fruits and leaves of , where it constitutes a significant portion of the plant's saponin content, ranging from 0.5% to 2% dry weight in aerial parts. Concentrations are highest in the aerial parts of this species, with reported levels up to 1.53% in samples from regions like . is native to warm temperate and tropical regions worldwide, including the , Asia, and , and has become widely distributed in drier temperate areas globally. Other notable natural sources include species of the genus , such as , Dioscorea tokoro, Dioscorea nipponica, and Dioscorea collettii, where protodioscin is concentrated in the rhizomes and often co-occurs with related compounds like dioscin and methyl protodioscin. In Trigonella foenum-graecum (fenugreek), protodioscin is present in the seeds, contributing to the plant's bioactive profile. Additionally, it has been identified in the underground parts of Trillium erectum and Trillium govanianum. Protodioscin is also found in Asparagus officinalis, particularly in white asparagus spears and shoots, with concentrations reaching up to 1.04% dry weight near the rhizome. Various yam varieties within the Dioscorea genus similarly contain protodioscin in their tubers and rhizomes, though levels vary by species and cultivation conditions. These plants have historically been utilized in traditional medicine for their saponin-rich compositions.

Biosynthesis

Protodioscin, a steroidal , is biosynthesized in through the mevalonate (MVA) pathway and the methylerythritol () pathway, which converge to produce isopentenyl diphosphate () and dimethylallyl diphosphate (DMAPP) as precursors for synthesis. These precursors are condensed to form geranylgeranyl diphosphate, which leads to via farnesyl diphosphate synthase and , followed by epoxidation to 2,3-oxidosqualene by epoxidase. The oxidosqualene is then cyclized to cycloartenol or by oxidosqualene cyclase, and subsequent modifications yield , the primary backbone for protodioscin. From , cytochrome P450-mediated oxidations at positions such as C-22 and C-26 produce furostanol aglycones like diosgenin, the aglycone core of protodioscin. Key enzymes in this pathway include oxidases, which catalyze critical and oxidation steps on the skeleton; notable examples are CYP90 family members like CYP90A1 and CYP90B, which facilitate C-3 oxidation and side-chain modifications leading to the furostanol structure. Glycosyltransferases, particularly UDP-glycosyltransferases (UGTs) such as UGT80A2 and UGT73CR1, attach sugar moieties including glucose and to the aglycone, forming the characteristic trisaccharide chains of protodioscin. Additional enzymes like cycloartenol synthase and obtusifoliol 14α-demethylase initiate and refine the framework early in the pathway. Transcriptome studies in plants such as Asparagus officinalis have identified upregulation of genes involved in protodioscin biosynthesis, including those encoding HMG-CoA synthase (HMGS), 1-deoxy-D-xylulose-5-phosphate synthase (DXS), and various UGTs, with higher expression in tissues accumulating protodioscin. Similar gene upregulation, particularly of CYP90A1 and UGT families, occurs in and species, correlating with steroidal production. In zingiberensis, UGTs like those responsible for rhamnosylation show tissue-specific expression linked to diosgenin . Putative biosynthetic steps include sequential at the C-3 hydroxyl group with a β-D-glucopyranosyl-(1→2)-β-D-galactopyranosyl unit and at the C-26 position with an α-L-rhamnopyranosyl-(1→4)-α-L-rhamnopyranosyl-(1→4)-β-D-glucopyranosyl chain, completing the protodioscin . Under environmental stress, furostanol can convert to spirostanol forms via activity, which cleaves the C-26 glucose to enable ring closure, potentially enhancing stability. Environmental factors such as drought stress influence protodioscin accumulation, with water deficit increasing levels in by upregulating biosynthetic genes and enzymes, aiding plant adaptation. Other stresses like and elicitors similarly boost steroidal saponin production across producing plants.

Pharmacology

Mechanism of Action

Protodioscin interacts with the pathway by increasing immunoreactivity in specific tissues, such as the paraventricular nucleus of the , where chronic administration of extract containing protodioscin elevated immunoreactivity by approximately 58%. This effect is likely secondary to elevated levels of testosterone and (DHT), achieved through stimulation of (LH) release from the , which in turn promotes production. Additionally, protodioscin supports steroidogenesis in Leydig cells by upregulating key enzymes involved in testosterone , including 17β-hydroxysteroid (17β-HSD), thereby enhancing the conversion of precursors to active androgens; this helps restore enzyme activity diminished by toxic insults. The subsequent conversion of testosterone to DHT is facilitated by activity in target tissues, amplifying androgenic signaling. In vascular tissues, extracts containing protodioscin promote release by enhancing endothelial activity in the corpus cavernosum, leading to relaxation and via the nitric oxide/cGMP pathway; this process is endothelium-dependent and can be inhibited by nitric oxide synthase blockers like L-NAME. Regarding cancer-related mechanisms, protodioscin induces and promotes cell cycle arrest at the in cell lines through activation of JNK and p38 signaling pathways.

Biological Activities

Protodioscin exhibits effects in animal models, enhancing and erectile function. In normal and castrated rats, administration of protodioscin-containing extract increased mounting frequency, intromission frequency, and intracavernosal pressure while reducing mount and intromission latencies. These improvements were comparable to those observed with testosterone supplementation, indicating protodioscin's potential to support sexual behavior parameters. Regarding hormonal modulation, protodioscin elevates serum levels of testosterone, (DHT), and (DHEAS) in rats. In castrated rats treated orally with extract containing protodioscin (5 mg/kg for 30 days), testosterone levels increased by 25%, with similar elevations in DHT and DHEAS observed in other rodent models. However, protodioscin shows minimal impact on levels, as estrogen concentrations remained largely unchanged or slightly decreased in treated groups compared to controls. Protodioscin demonstrates anticancer potential through against various cancer cells . It inhibits proliferation and induces in cell lines such as MOLT-4, with growth inhibition concentrations (GI50) of ≤2.0 µM. In colon cancer cells like HCT-116 and SW-620, protodioscin similarly suppresses growth at low micromolar concentrations. For , protodioscin reduces cell proliferation, migration, motility, and invasion in lines such as 5637 and T24, while promoting arrest and . In cardiovascular models, protodioscin reduces LDL and markers of in hyperlipidemic rats. Oral administration (10-20 mg/kg) significantly lowered serum total , triglycerides, and LDL-C levels while increasing HDL-C, thereby mitigating lipid accumulation and related vascular risks. Protodioscin displays activity by suppressing paw in models. In rats with complete Freund's adjuvant-induced , protodioscin (50-200 mg/kg) dose-dependently reduced paw swelling, ankle inflammation, and levels of COX-2, IL-1β, TNF-α, IL-6, and , comparable to sodium. Protodioscin exhibits activity through free radical scavenging. It has been associated with antidiabetic effects by improving and in animal models. Additionally, protodioscin shows antimicrobial activity and potential cardiovascular protection via increased expression of ATP-binding cassette transporter A1 () for reverse transport.

Uses

Traditional Uses

Protodioscin, a steroidal saponin found in plants such as Tribulus terrestris, has been utilized in traditional medicine through these plant sources for various ethnopharmacological purposes. In Ayurveda, Tribulus terrestris, known as Gokshura in Sanskrit, has been employed since ancient times for enhancing vitality, treating urinary disorders, and acting as an aphrodisiac; it is referenced in classical texts like the Charaka Samhita, dating to approximately 300 BCE, where it is described for its invigorating and diuretic effects. In Traditional Chinese Medicine, the plant is used similarly for addressing chest pain, heart-related issues, dizziness, and expelling kidney stones, often prepared to support overall energy and urinary health. Native American tribes have traditionally used species of , which contain protodioscin, particularly (known as Beth root), for reproductive health concerns, including easing , regulating menstrual irregularities, and providing pain relief during labor and gynecological issues. In Middle Eastern and European folklore, both ( foenum-graecum) and —sources of protodioscin—have been applied to address impotence and infertility in both men and women, with fenugreek seeds valued for hormonal regulation and sexual vitality, while served as a tonic for erectile function and reproductive disorders. Traditional preparations of these protodioscin-containing plants often involved decoctions or powders derived from fruits, leaves, or roots, administered to enhance stamina and treat conditions like ; for instance, Tribulus terrestris decoctions (25–50 ml daily) and powders (1–3 grams daily) were commonly used in Ayurvedic practice for urinary tract support and invigorating effects. The cultural significance of Gokshura underscores its role as an emblem of rejuvenation in Sanskrit traditions, symbolizing the plant's reputed ability to promote physical and vital energy. These historical applications have influenced the development of modern herbal supplements derived from and related plants for similar vitality-enhancing purposes.

Modern Applications

Protodioscin is a key active compound in standardized extracts of used in dietary supplements, often formulated with 40–60% total and varying protodioscin content (e.g., 10–20% in some commercial products) to support applications in , enhancement, and athletic performance. These extracts are marketed as natural aids for muscle building and sexual health, drawing from the plant's historical use in , though they are classified as dietary supplements and not approved as drugs by regulatory authorities such as the FDA. Recommended dosages for these Tribulus terrestris extracts range from 250–1500 mg per day, commonly administered in capsule form to facilitate consistent intake. In pharmaceutical contexts, protodioscin-containing extracts from are under investigation as potential alternatives to phosphodiesterase-5 (PDE5) inhibitors for managing , with ongoing exploration of their role in sexual health formulations. Veterinary applications include the incorporation of protodioscin-rich Tribulus terrestris extracts into animal feeds to enhance reproductive performance in livestock, such as improving fertility in sheep and pigs. Standardization of protodioscin in commercial products relies on (HPLC) methods, including HPLC-UV and HPLC-MS/MS techniques, to accurately quantify its concentration and ensure product quality.

Research

Preclinical Studies

Preclinical research on protodioscin has employed assays with various lines and rodent models to explore its pharmacological potential, focusing on sexual, anticancer, and metabolic effects. Studies in male rats have investigated protodioscin's influence on , particularly through extracts of standardized to protodioscin content. Oral administration of 5 mg/kg protodioscin-containing extract daily for 8 weeks in castrated rats increased testosterone levels by 25% compared to controls and enhanced copulatory parameters, including higher mount and intromission frequencies and shorter mount, intromission, and post-ejaculatory latencies. These improvements were attributed to androgenic effects, potentially involving increased expression. Similar benefits on sexual behavior were observed after shorter durations of 7–14 days in intact rats, with elevated testosterone supporting aphrodisiac-like activity. In vitro evaluations of protodioscin's anticancer properties have targeted human tumor cell lines, demonstrating antiproliferative and pro-apoptotic actions. Against cells, protodioscin inhibited growth in 5637 and T24 lines with IC50 values of 72.6 μM and 63.4 μM, respectively, after 24-hour exposure, while inducing G2/M phase arrest and via JNK and p38 signaling activation. Protodioscin's metabolic effects have been assessed in models of , where it ameliorated . In high-fat diet- and streptozotocin-induced diabetic rats, oral doses of 20–40 mg/kg protodioscin for 12 weeks significantly reduced fasting blood glucose levels (p < 0.01) and improved renal function markers, indicating potential antidiabetic activity through glucose-lowering mechanisms. Common preclinical models include Sprague-Dawley rats for sexual and diabetic studies, C57BL/6 mice for metabolic evaluations, and human-derived cell lines such as T24/5637 (bladder) for anticancer screening. However, efficacy often requires relatively high doses (e.g., 20–40 mg/kg in vivo or 50–100 μM in vitro), raising concerns about translational relevance to humans due to pharmacokinetic differences.

Clinical Studies

Clinical studies on protodioscin, primarily investigated through extracts containing 40-60% protodioscin, have yielded mixed results regarding its effects on libido and erectile dysfunction (ED). A systematic review of five randomized controlled trials (RCTs) involving 279 women with sexual dysfunction found significant improvements in overall sexual function scores, including desire, arousal, lubrication, satisfaction, and pain, after 1-3 months of supplementation (doses 250-750 mg/day), with low certainty of evidence due to methodological limitations. In men, a 2025 meta-analysis of RCTs (total n=543) reported improvements in International Index of Erectile Function (IIEF) scores for ED (mean difference 3.23 for IIEF-5, p<0.00001), but results were inconsistent across studies with small samples (n=30-172). A 2021 RCT in 30 male CrossFit athletes (770 mg/day for 6 weeks) showed no changes in body composition despite some hormonal shifts. Hormonal effects of protodioscin-rich extracts remain inconsistent, particularly for testosterone levels. The same 2025 meta-analysis indicated no significant overall increase in total testosterone compared to placebo in men with ED, though two RCTs in hypogonadal men (n=30-70, 750 mg/day, 12 weeks) reported modest rises of 60-70 ng/dL. In women, a systematic review noted a significant testosterone increase (mean difference 6.60 ng/dL) in premenopausal participants after 3 months, but no change in postmenopausal women; benefits were more pronounced for alleviating postmenopausal sexual symptoms like hypoactive sexual desire disorder. A 2014 systematic review up to that year concluded no testosterone elevation in healthy humans, highlighting variability based on baseline levels and population. Safety profiles in clinical trials support good tolerability. Across RCTs reviewed (doses up to 1.5 g/day for up to 90 days), no serious adverse events were reported, with no significant changes in liver or kidney function markers; minor gastrointestinal issues occurred rarely and similarly to placebo. Key trials include RCTs on for infertility, such as a 2016 study (n=65 infertile men, 250 mg/day protodioscin-equivalent extract) showing improved sperm concentration, motility, and liquefaction time after treatment. Another RCT (n=180 men with sexual dysfunction, 750 mg/day, 3 months) demonstrated enhanced overall sexual function without hormonal alterations. Research gaps persist, including small sample sizes (often n<100), short durations (typically <12 weeks), and lack of protodioscin-specific dosing standardization, limiting generalizability and calling for larger, longer-term RCTs.

Safety

Adverse Effects

Protodioscin, a steroidal saponin primarily found in Tribulus terrestris extracts, is generally well-tolerated at recommended doses, with adverse effects reported as mild and uncommon in clinical studies. Common gastrointestinal side effects include stomach cramps, nausea, and diarrhea, which are typically mild and occur at low rates similar to placebo in randomized trials. These effects have been noted in less than 5% of participants across multiple clinical evaluations of Tribulus terrestris extracts standardized to protodioscin content. Hormonal-related adverse effects are rare and primarily associated with high doses mimicking androgenic activity. Isolated cases include gynecomastia in a young male using as a body-building supplement, resolving after discontinuation. Acne has also been reported sporadically at elevated doses due to potential androgen mimicry. Allergic reactions, such as rash or itching, may occur in sensitive individuals, though these are infrequent and often linked to general herbal supplement intolerance rather than protodioscin specifically. Case reports have described prostate enlargement in long-term users of , potentially attributable to 's influence on hormonal pathways, but these findings remain unconfirmed and require further investigation. Overall, adverse effects are uncommon at doses below 1 g per day of protodioscin-containing extracts and typically resolve upon discontinuation. Protodioscin-containing supplements should be avoided during pregnancy, as animal studies suggest potential harm to fetal development, and during breastfeeding due to insufficient safety data. Caution is advised in children and individuals with hormone-sensitive conditions, such as prostate cancer, due to possible androgenic effects.

Toxicology and Interactions

Protodioscin, a steroidal saponin primarily derived from plants like Tribulus terrestris, exhibits low acute toxicity in preclinical models. In rats administered a spray-dried extract of T. terrestris containing protodioscin, the median lethal dose (LD50) exceeded 5000 mg/kg body weight following a single oral dose, with no observed mortality, behavioral changes, or histopathological abnormalities in major organs such as the liver and kidneys over a 14-day observation period. Similarly, acute oral administration of a protodioscin-rich extract from Trigonella foenum-graecum at 2000 mg/kg body weight produced no clinical signs of toxicity, mortality, or disruptions in hematological and biochemical parameters, confirming its low toxicity profile. Subchronic exposure studies further support protodioscin's safety margin. In a 28-day repeated-dose study using T. terrestris extract in rats at doses up to 1500 mg/kg/day, no deaths occurred, and there were no significant alterations in body weight, organ weights, , serum biochemistry, or , establishing a (NOAEL) of at least 1000 mg/kg/day. Chronic effects, however, warrant caution in humans, particularly with overuse of -based supplements. Case reports document elevations in liver enzymes such as (ALT) and aspartate aminotransferase (AST) following prolonged high-dose intake, alongside rare instances of kidney strain in individuals with predisposing factors, including acute renal linked to bile cast nephropathy. Protodioscin may interact with certain medications due to its potential influence on hormonal, vascular, and metabolic pathways. It could enhance the effects of testosterone therapies by promoting the conversion of testosterone to via saponin-mediated mechanisms, necessitating monitoring for amplified androgenic activity. Concurrent use with antihypertensives may potentiate reduction through increased production, raising the risk of . Caution is also advised with antidiabetic agents, as protodioscin's glucose-lowering potential could lead to when combined. In the , extracts are monitored for content to ensure safety, with recommendations for clear labeling of preparation methods and active components to mitigate risks from variability. Mild gastrointestinal upset has been occasionally reported with supplement use, though this is not indicative of systemic .