Salidroside is a natural phenolicglycoside, chemically known as 2-(4-hydroxyphenyl)ethyl β-D-glucopyranoside, with the molecular formula C₁₄H₂₀O₇ and a molecular weight of 300.31 g/mol.[1] It is primarily isolated from the roots and rhizomes of various Rhodiola species, such as Rhodiola rosea, R. crenulata, and R. sachalinensis, which thrive in high-altitude regions like the Himalayas.[2] As a key bioactive constituent in traditional Chinese medicines prepared from these adaptogenic plants, salidroside contributes to their reputed ability to enhance stress resistance and overall vitality.[3]This compound has garnered significant attention for its multifaceted pharmacological profile, demonstrating potent antioxidant properties by scavenging reactive oxygen species (ROS) and mitigating oxidative stress.[2] Salidroside also exhibits neuroprotective effects, protecting against neuronal damage in models of Alzheimer's disease, Parkinson's disease, and cerebral ischemia.[3] In the cardiovascular system, it offers protection against atherosclerosis and hypoxia, while displaying anti-inflammatory and anti-cancer activities.[2] Additionally, salidroside shows promise in anti-aging, anti-diabetic, and anti-fibrotic applications, with preclinical studies highlighting its role in reducing aging markers and improving insulin sensitivity.[3] As of 2025, recent clinical studies have explored its potential in enhancing exercise performance and sports nutrition, alongside applications in rheumatoid arthritis and fertility.[4][5][6]Despite its therapeutic potential, salidroside's clinical translation remains limited by challenges such as low bioavailability, though ongoing research explores synthetic production via engineered microbes and novel delivery systems to enhance its efficacy.[2] Its biosynthesis in plants involves the conversion of tyrosine to tyrosol followed by glycosylation, underscoring opportunities for biotechnological optimization.[3]
Introduction
Definition and Chemical Identity
Salidroside is a naturally occurring tyrosolglucoside, chemically known as p-tyrosol β-D-glucopyranoside, and is classified as a phenyl ethanol glycoside or phenylethanoid glycoside.[7][8][9] This compound represents a glycosylated derivative of tyrosol, where a β-D-glucose moiety is attached to the phenolic ethanol backbone, contributing to its solubility and biological stability in plant tissues.[10]The molecular formula of salidroside is C₁₄H₂₀O₇, with a molar mass of 300.31 g/mol.[1][11] It is identified by the CAS registry number 10338-51-9 and the PubChem Compound ID (CID) 159278.[1][11] These identifiers confirm its unique chemical identity as a bioactive phenolicglycoside derived from tyrosine metabolism.[12]As a key active constituent in species of the Rhodiola genus, such as Rhodiola rosea and Rhodiola crenulata, salidroside plays a central role in the adaptogenic properties of these plants, which have been utilized in traditional medicine for enhancing stress resistance and vitality.[13][14] Its presence in the rhizomes and roots underscores its importance as a primary phenolic compound contributing to the pharmacological profile of Rhodiola extracts.[14]
Historical Background
Salidroside, originally known as rhodioloside, has roots in the traditional medicinal uses of Rhodiola species, particularly Rhodiola rosea, which have been employed for centuries across various cultures to combat fatigue, stress, and related ailments. In Traditional Chinese Medicine, Rhodiola rosea, referred to as "Hong Jing Tian," has been utilized since ancient times as an adaptogen to enhance resistance to physical and mental stress, alleviate asthenia, and promote vitality, often incorporated into formulations for improving endurance and recovery from exhaustion.[15] Similarly, in Russian folk medicine, the plant was valued as a stimulant to reduce fatigue, boost physical and mental performance, and support overall resilience, with records dating back to the 18th century for treating anxiety, depression, and stress-induced weakness. In Scandinavian traditions, Rhodiola has been used to enhance physical and mental performance and improve tolerance to environmental stressors.[16]The scientific recognition of salidroside began in the mid-20th century amid Soviet efforts to identify adaptogenic compounds from native plants. In 1967, Soviet researchers A.T. Troshchenko and G.A. Kutikova first isolated salidroside from the roots of Rhodiola rosea and Rhodiola quadrifida, identifying it alongside its aglycone tyrosol and naming it rhodioloside based on its phenolic glycoside structure. This isolation occurred as part of broader adaptogen research in the USSR during the 1960s, where Rhodiola extracts were investigated for their potential to enhance nonspecific resistance to stressors, building on traditional uses and aligning with national interests in performance enhancement for athletes, cosmonauts, and laborers.[17]Early pharmacological investigations in the 1970s and 1980s further highlighted salidroside's role within Rhodiola extracts, particularly in demonstrating anti-fatigue effects through animal models. Studies during this period, including those by Saratikov and colleagues, showed that Rhodiola preparations containing salidroside prolonged physical endurance and reduced exhaustion in rats subjected to swimming and other stress tests, attributing these outcomes to the compound's adaptogenic properties that modulated stress responses without significant toxicity.[18] These findings solidified salidroside's prominence as a key bioactive constituent, bridging traditional knowledge with emerging biomedical insights into fatigue mitigation.[17]
Chemical Properties
Molecular Structure
Salidroside, chemically designated as 2-(4-hydroxyphenyl)ethyl-β-D-glucopyranoside, features a tyrosol aglycone—a phenethyl alcohol derivative with a phenolic ring—covalently bound to a glucose unit through a β-glycosidic linkage at the anomeric carbon of the sugar.[19] This structure positions the glucose as a β-D-glucopyranoside, where the aglycone attaches via an oxygen bridge from the C1 position of the glucose to the terminal carbon of the tyrosol's ethyl chain.[20]The molecule's key functional groups encompass the phenolic hydroxyl (-OH) on the para-substituted benzene ring of tyrosol, which imparts antioxidant potential; the ether linkage inherent in the β-glycosidic bond, connecting the aglycone and sugar moieties; and the pyranose ring of the glucoside, bearing additional hydroxyl groups at C2, C3, C4, and C6, along with a hydroxymethyl at C5.[21] These elements define salidroside's polarity and reactivity, distinguishing it from aglycone forms like free tyrosol.[22]Stereochemically, salidroside exhibits the β-anomeric configuration at the glucose C1, ensuring the glycosidic oxygen occupies an equatorial position in the chair conformation of the pyranose ring, which contributes to its stability and biological recognition.[20] In contrast to related Rhodiola compounds like rosavin, which employs a phenylpropanoid cinnamyl alcohol aglycone with glycoside attachment along a propenyl chain, salidroside's phenylethanoid scaffold results in a simpler, more linear architecture.[23]
Physical and Spectroscopic Properties
Salidroside is typically isolated and observed as a white to off-white crystalline powder, facilitating its handling and storage in laboratory settings.[24][25]This compound demonstrates high solubility in polar solvents, achieving up to 60 mg/mL in water at neutral pH, as well as ready solubility in methanol and ethanol; in contrast, it exhibits low solubility in non-polar solvents such as diethyl ether and acetone.[25][26] Its thermal properties include a melting point of 159–160 °C, with the compound remaining stable under neutral pH conditions (around pH 7), though it may hydrolyze under acidic environments.[24][27]Spectroscopic characterization aids in the identification and purity assessment of salidroside. In ultraviolet-visible (UV-Vis) spectroscopy, it exhibits a characteristic absorption maximum at 220 nm, primarily due to the phenolic hydroxyl group in its structure.[28]Nuclear magnetic resonance (NMR) analysis, particularly ^1H NMR in deuterated solvents like DMSO-d_6 or CD_3OD, shows aromatic protons as doublets around δ 6.68–7.02 ppm (J ≈ 8.4 Hz), reflecting the para-substituted benzene ring, with the anomeric proton of the β-glucosidic linkage appearing near δ 5.0 ppm.[29]Infrared (IR) spectroscopy reveals key functional group vibrations, including a broad O–H stretching band at approximately 3400 cm⁻¹ from the phenolic and alcoholic hydroxyls, and C–O stretching at around 1100 cm⁻¹ associated with the glycosidic ether linkage.[30] These spectral features, derived from the tyrosolglucoside backbone, enable reliable analytical confirmation in pharmaceutical and natural product research.[29]
Natural Occurrence and Production
Plant Sources
Salidroside is primarily found in the roots and rhizomes of plants belonging to the genus Rhodiola in the Crassulaceae family, with key species including Rhodiola rosea (also known as golden root), Rhodiola crenulata, and Rhodiola sachalinensis. These arctic-alpine perennials are adapted to harsh, cold climates and serve as the main natural sources for the compound.[2][31]The geographical distribution of these Rhodiola species spans high-altitude regions, including the Arctic tundra, the Himalayan and Altai mountain ranges in Asia, and alpine areas in Europe, with significant diversity in China, Tibet, and Mongolia where over 70 species are documented. Cultivation has extended their availability to North America and parts of Europe to meet demand.[32][2]In Rhodiola rosea roots, salidroside concentrations typically range from 0.5% to 2% of dry weight, though values can reach up to 1.8% under optimal conditions; levels vary by species, altitude, soil nutrients, and harvest timing, with higher elevations often yielding greater amounts due to environmental stress.[33][34][35]
Extraction and Synthesis Methods
Salidroside is primarily obtained through natural extraction from plant sources such as Rhodiola species, where solvent-based methods like methanol or ethanolmaceration are commonly employed to isolate the compound from rhizomes and roots.[36] These traditional solvent extractions involve soaking the plant material in methanol for several hours, followed by filtration and concentration, yielding crude extracts containing salidroside at concentrations typically around 0.5-1% of dry weight.[37] To enhance efficiency, microwave-assisted extraction (MAE) has been developed, utilizing microwave energy at 90°C for 20 minutes to rupture plant cells and accelerate solventpenetration, resulting in higher yields and shorter processing times compared to conventional methods.[38] Additionally, supercritical CO2 extraction with an ethanol modifier operates under high pressure (200-300 bar) and moderate temperatures (40-60°C), offering a green alternative that minimizes solvent residues and achieves salidroside yields up to 1.5% while preserving bioactivity.[39]Chemical synthesis of salidroside typically involves the glycosylation of tyrosol, the aglycone precursor, with a glucose moiety using activated glycosyl donors such as acetobromoglucose in the presence of catalysts like silver oxide or Lewis acids.[40] The process proceeds via the Koenigs-Knorr reaction, where tyrosol reacts with protected glucose (e.g., 2,3,4,6-tetra-O-acetyl-α-D-glucopyranosyl bromide) to form the β-glycosidic bond, followed by deacetylation under basic conditions like sodium methoxide in methanol to yield the free salidroside.[41] Overall yields for this multi-step synthesis range from 40-60%, depending on reaction optimization and purification steps, making it suitable for laboratory-scale production but less economical for large volumes due to the need for protected intermediates.[42]Biotechnological production leverages microbial fermentation in hosts like Escherichia coli or Saccharomyces cerevisiae engineered to express glycosyltransferases such as UGT85A1 from Arabidopsis thaliana, enabling de novo synthesis from glucose or tyrosine precursors.[43] In E. coli, pathways incorporating pyruvate decarboxylase (e.g., ARO10) and alcohol dehydrogenase produce tyrosol intermediates, which are then glycosylated, achieving titers of 56.9 mg/L in optimized shake-flask cultures after 48 hours of fermentation at 30°C.[44] More recent engineering efforts have achieved titers up to 16.8 g/L in bioreactors as of 2024.[45] Yeast-based systems similarly utilize shikimate pathway enhancements and UDP-glucosyltransferases, yielding comparable titers while benefiting from eukaryotic glycosylation machinery for higher fidelity, though scaling to bioreactors remains a focus for industrial viability.[46]Purification of salidroside from crude extracts or fermentation broths often employs high-speed counter-current chromatography (HSCCC), which uses a biphasic solvent system (e.g., ethyl acetate-ethanol-water) to separate based on partition coefficients, achieving purities exceeding 98% without solid stationary phases.[47] Alternatively, silica gel column chromatography with gradient elution (e.g., chloroform-methanol mixtures) provides effective separation of salidroside from impurities like tyrosol and rosavins, routinely attaining >98% purity after recrystallization, though it requires more solvent and time than HSCCC.[48] These methods ensure high-purity salidroside suitable for pharmaceutical applications, with overall recovery rates of 70-90% from initial extracts.[49]
Biosynthesis
Biosynthetic Pathway
Salidroside biosynthesis in plants of the Rhodiola genus primarily occurs through a tyrosine-derived pathway originating from the shikimate pathway, which converts phosphoenolpyruvate and erythrose-4-phosphate into aromatic amino acids, including L-tyrosine. In Rhodiola rosea, the committed step involves the direct conversion of L-tyrosine to 4-hydroxyphenylacetaldehyde (4-HPAA) by the bifunctional enzyme 4-HPAA synthase (Rr4HPAAS), a neofunctionalized pyridoxal 5'-phosphate-dependent enzyme that performs both decarboxylation and deamination without requiring separate monoamine oxidase activity. This step corrects earlier models proposing sequential decarboxylation and oxidation.[50]Subsequently, 4-HPAA is reduced to tyrosol, the aglycone of salidroside, by NADPH-dependent 4-HPAA reductases such as Rr4HPAR1 and Rr4HPAR2, with Rr4HPAR1 exhibiting higher catalytic efficiency (6.9 μmol min⁻¹ mg⁻¹ protein). The final glycosylation step couples tyrosol with UDP-glucose to yield salidroside, catalyzed by a regio-specific UDP-glucosyltransferase (UGT), including RrUGT33 (also termed RrT8GT) in R. rosea with a catalytic efficiency of 420.6 s⁻¹ mM⁻¹, or RcUGT72B14 in Rhodiola crenulata. These UGTs preferentially modify the 8-position of tyrosol, ensuring specificity in phenylethanoid glycoside formation.[50][43]An alternative biosynthetic route, proposed in earlier studies and observed in some Rhodiola species, initiates with decarboxylation of tyrosine to tyramine by tyrosine decarboxylase (TYDC), followed by β-hydroxylation or oxidation of tyramine to 4-HPAA via tyramine oxidase, and subsequent reduction to tyrosol. In certain plants, a parallel pathway derives tyrosol from phenylalanine through deamination by phenylalanine ammonia-lyase (PAL) to cinnamic acid, 4-hydroxylation to p-coumaric acid, and further reduction steps, though this route shows limited contribution to salidroside in Rhodiola.[43][51]The pathway enzymes exhibit tissue-specific regulation, with prominent expression in roots where salidroside accumulates, supporting the plant's adaptation to environmental pressures in high-altitude habitats.[50]
Metabolic Engineering
Metabolic engineering of salidroside production has focused on reconstructing the biosynthetic pathway in heterologous hosts to overcome limitations in natural extraction from Rhodiola plants. Key efforts involve expressing tyrosine decarboxylase (TYDC) and UDP-glucosyltransferase (UGT) enzymes from Rhodiola rosea, such as RrTYDC and RrUGT72B14 or RrT8GT, to convert tyrosine to tyrosol and then to salidroside.[50] In Saccharomyces cerevisiae, co-expression of these enzymes with feedback-insensitive variants of ARO4 and ARO7 has enabled de novo production from glucose, achieving titers up to 732 mg/L in fed-batch fermentation.[52] Similarly, transient expression in Nicotiana benthamiana via Agrobacterium-mediated delivery of RrTYDC and RrT8GT has yielded salidroside at up to 2% dry weight in infiltrated leaves.[50]Further advancements in Escherichia coli have demonstrated higher efficiencies through chromosomal integration and multi-copy expression of the pathway genes, including plant-derived 4-hydroxyphenylacetaldehyde synthase (4HPAAS) and UGT85A1, resulting in salidroside production of 2.42 g/L from glucose in shake flasks and up to 9.34 g/L in scaled fermenters.[53] Optimization strategies include co-expression of cofactor recyclers, such as soybean sucrose synthase (GmSUS) to regenerate UDP-glucose, the essential glycosyl donor for UGT activity, which has boosted glycosylation efficiency and titers to 3.8 g/L in coculture systems with S. cerevisiae.[54] Pathway balancing via CRISPR/Cas9-mediated genome editing has been applied in microbial hosts to fine-tune gene expression and minimize flux to byproducts, though specific implementations for salidroside remain emerging.Recent developments as of 2025 highlight plant-based platforms, with tobacco (Nicotiana tabacum) cell cultures engineered using multi-gene cassettes expressing RrTYDC, Rr4HPAAS, and RrUGT to produce salidroside at approximately 378 μg/g fresh weight from endogenous tyrosine.[55] These yields represent significant improvements over early heterologous efforts, approaching industrial viability.Despite progress, challenges persist, including limited availability of the glycosyl donor UDP-glucose, which competes with cellular metabolism and requires supplementation or recycling modules. Additionally, toxicity from pathway intermediates like tyrosol inhibits host growth, necessitating adaptive evolution or compartmentalization strategies to maintain productivity.[54]
Pharmacological Activities
Antioxidant and Anti-inflammatory Effects
Salidroside demonstrates potent antioxidant activity through direct scavenging of reactive oxygen species (ROS), attributed to its phenolic hydroxyl group, which facilitates electron donation and neutralization of free radicals.[56] Additionally, it activates the Nrf2 signaling pathway, promoting nuclear translocation of Nrf2 and subsequent upregulation of downstream antioxidant enzymes, including superoxide dismutase (SOD) and catalase (CAT).[57] In cellular assays, salidroside exhibits efficacy in reducing oxidative stress.[58]The compound's anti-inflammatory effects involve inhibition of NF-κB activation, which suppresses the transcription of pro-inflammatory genes and reduces the production of cytokines such as tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in lipopolysaccharide (LPS)-stimulated macrophages.[59] Salidroside also modulates mitogen-activated protein kinase (MAPK) and c-Jun N-terminal kinase (JNK) signaling pathways, further attenuating inflammatory responses in activated immune cells.[60]In vitro studies have shown that salidroside protects hepatocytes from hydrogen peroxide (H2O2)-induced oxidative damage by decreasing ROS accumulation and preserving cell viability.[61] In vivo assessments in brain tissue demonstrate its ability to reduce lipid peroxidation, as evidenced by lowered malondialdehyde (MDA) levels following exposure to oxidative stressors.[62]In rodent models, salidroside administered at doses of 20-100 mg/kg effectively diminishes MDA levels, indicating reduced lipid peroxidation and enhanced systemic antioxidant capacity.[63] These mechanisms contribute to its broader adaptogenic properties under stress conditions.[57]
Neuroprotective and Adaptogenic Effects
Recent studies on salidroside and its derivatives demonstrate inhibition of amyloid-β aggregation and monoamine oxidase B (MAO-B) activity, targeting these pathways to mitigate neurodegenerative processes.[64] In models of cerebral ischemia, salidroside protects neuronal cells through upregulation of brain-derived neurotrophic factor (BDNF) expression, reducing excitotoxicity and promoting neural repair in the ischemic periphery,[65] while also regulating glutamate metabolism in astrocytes.[66] These effects contribute to decreased neuronal apoptosis and improved histological outcomes in ischemic brain injury.[62]As an adaptogen, salidroside enhances hypothalamic-pituitary-adrenal (HPA) axis balance and reduces cortisol levels in stress-induced models, thereby improving stress resilience without altering baseline hormone function.[67] It also exerts anti-fatigue effects by activating AMP-activated protein kinase (AMPK) and promoting mitochondrial biogenesis, which supports energy metabolism and endurance in exhaustive exercise paradigms.[68]Salidroside readily crosses the blood-brain barrier, enabling direct modulation of neuronal survival pathways such as PI3K/Akt signaling, which inhibits apoptosis and fosters neuroprotection in various injury models.[69] In antidepressant assays, doses of 10-50 mg/kg reduce immobility time in the forced swim test, indicating enhanced behavioral resilience to stress.[70]In specific disease models, salidroside ameliorates Parkinson's-like symptoms by preserving dopamine levels and protecting dopaminergic neurons from mitochondrial dysfunction in MPTP-induced paradigms.[71] It also demonstrates anti-anxiety effects in the elevated plus-maze test, increasing time spent in open arms following acute administration, which supports its role in modulating emotional behaviors.[72]
Research and Applications
Preclinical Studies
Preclinical investigations have explored salidroside's therapeutic potential in multiple disease models, primarily through in vitro and in vivo experiments using rodents and cell lines. These studies highlight its ability to modulate pathological processes without delving into human applications.In anti-cancer research, salidroside has shown inhibitory effects on tumor growth in xenograft models. For instance, in non-small cell lung cancer xenografts in nude mice, intraperitoneal administration of salidroside at 50 mg/kg every other day significantly reduced tumor volume by promoting apoptosis and modulating the tumor microenvironment, including upregulation of T cells and dendritic cells while suppressing regulatory T cells.[73] Similar antitumor activity was observed in breast cancer xenografts, where salidroside treatment inhibited proliferation and invasion, leading to smaller tumor sizes compared to controls.[74]Cardioprotective effects of salidroside have been evidenced in models of myocardial infarction. In rats subjected to ischemia-reperfusion injury, salidroside pretreatment reduced infarct size and preserved cardiac function by upregulating anti-apoptotic Bcl-2 expression and inhibiting caspase-3 activation.[75] Administration in acute myocardial infarction rat models also attenuated apoptosis and improved systolic performance, with outcomes linked to pathways such as Akt phosphorylation.[76]For anti-diabetic activity, salidroside improved insulin sensitivity in streptozotocin (STZ)-induced diabetic models. In high-fat diet/STZ-treated mice, oral salidroside lowered fasting blood glucose levels by approximately 25-30% and reduced serum insulin, alleviating hyperglycemia and hyperinsulinemia through enhanced insulin signaling.[77] These effects were consistent across diabetic nephropathy models, where salidroside mitigated renal damage while restoring glucose homeostasis.[78]Additional preclinical findings include hepatoprotective benefits against acetaminophen (APAP) toxicity. In APAP-challenged mice, salidroside pretreatment decreased liver enzyme elevations (ALT/AST) and histopathological damage by preserving glutathione levels and inhibiting oxidative stress, resulting in survival rates over 80% compared to untreated controls.[79] In rheumatoid arthritis models, such as collagen-induced arthritis in rats and mice, salidroside reduced joint inflammation and paw swelling in 2025 studies, supporting its anti-arthritic potential through immune modulation.[80]Regarding pharmacokinetics, preclinical data indicate salidroside's oral bioavailability is approximately 32% in rats, with rapid absorption leading to peak plasma concentrations within 25-30 minutes.[81] The plasma half-life ranges from approximately 0.7 to 2 hours, influenced by formulation and species, as observed in rodent models following oral dosing.[82] These properties, including brief references to Nrf2 pathway involvement in oxidative stress reduction, underscore salidroside's suitability for further translational research.[83]
Clinical Trials and Therapeutic Potential
Clinical trials on salidroside in humans remain limited, with the majority of evidence derived from studies on Rhodiola rosea extracts where salidroside serves as a primary bioactive component. Recent investigations have begun to isolate the effects of pure or biosynthetic salidroside, particularly in contexts of physical performance and stress adaptation. A key example is an exploratory randomized, double-blind, placebo-controlled trial conducted in 2024 involving 50 healthy active young adults (aged 21 ± 4 years), who received 60 mg/day of biosynthetic salidroside (30 mg twice daily) or placebo for 16 days. Participants underwent high-intensity intermittent exercise testing, revealing that salidroside significantly enhanced predicted oxygen uptake (p < 0.01), reduced exercise-induced increases in myoglobin as a marker of muscle damage (p = 0.02), and mitigated declines in mood and rises in fatigue observed in the placebo group (p < 0.01 for fatigue-inertia and friendliness scores). Time-to-exhaustion also improved in the salidroside group compared to placebo (p = 0.03).[84]As of 2025, additional studies on fermented salidroside have confirmed efficacy in sports nutrition, enhancing performance and recovery in active individuals.[4]These findings indicate salidroside's potential as an adaptogen for reducing fatigue and supporting recovery in athletic populations, with stable mood maintenance during intense physical demands. No serious adverse events were reported, supporting the compound's tolerability at low doses. Broader human data on psychiatric applications, such as anxiety and stress reduction, are referenced in reviews but primarily stem from Rhodiola-based interventions rather than isolated salidroside.[85]Therapeutic potential extends to adjunctive roles in stress-related conditions, informed by the exercise trial's mood-stabilizing effects and preclinical evidence of adaptogenic properties. Emerging preclinical research highlights possibilities in rheumatoid arthritis through anti-inflammatory pathways, including modulation of Th17/Treg balance and cytokine suppression, though human validation is pending. Similarly, in vitro and animal studies suggest neuroprotective benefits, such as MAO-B inhibition for Alzheimer's disease adjunct therapy, but no dedicated human trials have confirmed these yet. Dosing in available human studies is 60 mg/day over 7-16 days, deemed safe with variable efficacy across outcomes, underscoring the need for larger randomized controlled trials to establish broader clinical utility.[6]
Safety and Toxicology
Toxicity Studies
Acute toxicity studies of salidroside in Sprague-Dawley rats have demonstrated a high safety margin, with an oral LD50 exceeding 5000 mg/kg body weight and no observed mortality, clinical signs, or gross pathological changes following single doses up to this level.[86] No organ damage, including to the liver or kidneys, was reported in these assessments, indicating minimal risk from acute exposure.[86]Subchronic toxicity evaluations, including a 28-day repeated oral dose study in male and female Sprague-Dawley rats at doses up to 2000 mg/kg/day, showed no adverse effects on body weight, food consumption, hematology, clinical chemistry, or organ histopathology.[86] Genotoxicity assessments, such as the Ames test using Salmonella typhimurium strains, were negative for mutagenicity at concentrations up to 5000 µg/plate, confirming no DNA-damaging potential.[86] Similarly, no hepatotoxicity was evident, with normal liver enzyme levels (e.g., AST, ALT) and absence of histopathological alterations in the liver.[86] Although a dedicated 90-day study specific to salidroside is limited, related extract studies containing salidroside at equivalent doses support these findings of safety.[87]Reproductive toxicity investigations in pregnant Sprague-Dawley rats administered salidroside at doses ranging from 125 to 500 mg/kg showed no maternal toxicity, embryonic lethality, or teratogenic effects, with normal fetal development and no structural abnormalities observed.[88]Recent studies from 2022 on bioengineered, nature-identical salidroside in rats confirmed its safety profile, revealing no adverse impacts on blood chemistry parameters (e.g., glucose, ureanitrogen) or histopathological features in major organs after repeated dosing up to 2000 mg/kg/day.[86] Overall, preclinical toxicity data align with limited human data suggesting good tolerability, such as a 2024 randomized, double-blind, placebo-controlled trial in healthy adults where 60 mg/day for 16 days resulted in no adverse events and normal blood parameters.[88][89]
Contraindications and Interactions
Due to limited specific data on isolated salidroside, precautions from studies on Rhodiola rosea (which contains salidroside as a major component) suggest avoiding use during pregnancy and breastfeeding to minimize potential risks to the fetus or infant.[90][91]Based on Rhodiola data, individuals with bipolar disorder should avoid salidroside, as it may exacerbate manic episodes through adaptogenic and stimulant-like effects observed in the plant extract.[92][93]For those with autoimmune conditions, such as multiple sclerosis or rheumatoid arthritis, caution and medical monitoring are advised when using salidroside, as Rhodiola may stimulate immune activity and potentially counteract immunosuppressant therapies; salidroside-specific effects remain understudied.[92][94]Salidroside may interact with monoamine oxidase (MAO) inhibitors, potentially potentiating their effects and increasing the risk of tyramine-like hypertensive crises, although in vitro studies show minimal direct MAO-A or MAO-B inhibition by salidroside itself at clinically relevant concentrations.[95][96]It can enhance the sedative effects of central nervous system depressants, such as benzodiazepines or barbiturates, leading to increased drowsiness and impaired coordination, based on observations with Rhodiola.[91]In vitro evaluations indicate that salidroside does not significantly inhibit or induce CYP3A4, suggesting low risk of altering the metabolism of statins like atorvastatin or simvastatin when co-administered.[95]Patients with hepatic impairment should use salidroside with dose adjustments and monitoring, given its partial hepatic metabolism, despite observed hepatoprotective effects in preclinical models.[97][8]When combining salidroside with antidepressants, such as SSRIs or MAOIs, close monitoring is recommended based on Rhodiola data to prevent enhanced side effects, including potential serotonin syndrome, as noted in case reports and interaction warnings for the plant up to 2025; salidroside-specific risks require further study.[91][98]