Resveratrol is a naturally occurring stilbenoid polyphenol with the chemical formula C₁₄H₁₂O₃, characterized by its 3,5,4'-trihydroxystilbene structure, and exists primarily in two isomeric forms: trans-resveratrol, which is more biologically active, and cis-resveratrol.[1][2] It functions as a phytoalexin in plants, produced in response to stress, injury, or pathogen attack to protect against fungal infections and environmental stressors.[3][4]This compound is found in over 70 plant species, with the highest concentrations in the skins of red grapes (Vitis vinifera), where it contributes to the plant's defense mechanism.[3][5] Dietary sources of resveratrol include red and white wines (derived from grape skins during fermentation), berries such as blueberries, cranberries, and mulberries, peanuts, pistachios, apples, plums, and even cocoa and dark chocolate in smaller amounts.[6][5][7] Human intake is typically low, estimated at 0.2–5 mg per day from diet, though supplements can provide higher doses.[5][8]Resveratrol has garnered significant scientific interest due to its diverse biological activities, including potent antioxidant effects that neutralize free radicals and enhance endogenous antioxidant enzymes, as well as anti-inflammatory properties by modulating pathways like NF-κB.[2][4] It activates sirtuin 1 (SIRT1), a protein linked to longevity and metabolic regulation, mimicking the effects of calorie restriction in animal models.[9] Research suggests potential cardioprotective benefits, such as improving endothelial function and reducing low-density lipoprotein oxidation, neuroprotective roles against Alzheimer's and Parkinson's diseases, and anti-cancer effects through apoptosis induction and cell cycle arrest in preclinical studies.[2][4][3]Despite promising in vitro and animal data, clinical evidence in humans remains mixed and inconclusive for many health claims, with bioavailability challenges limiting its efficacy; ongoing studies explore nanoformulations and combinations to enhance absorption.[10][4] Resveratrol's role in the "French Paradox"—the observation of low cardiovascular disease rates despite high-fat diets, attributed partly to red wine consumption—has further popularized its study.[8][11]
Chemistry
Structure and Properties
Resveratrol is a stilbenoidpolyphenol with the molecular formula \ce{C14H12O3}.[1] It features a central stilbene core consisting of two aromatic rings linked by an ethene bridge, with hydroxyl groups attached at the 3, 5, and 4' positions.[1] The compound exists in two geometric isomers due to the configuration around the double bond: the naturally predominant trans (E) form, where the phenyl rings are on opposite sides of the double bond, and the cis (Z) form, where they are on the same side.[12] The transisomer is more stable and bioactive, while the cisisomer can result from photo- or thermal-induced isomerization.[13]Physically, trans-resveratrol appears as a white to off-white crystalline powder.[14] It has a melting point of 253–255 °C.[14] Solubility is limited in water, approximately 30 mg/L at 25 °C, but it dissolves readily in organic solvents such as ethanol (50 mg/mL) and DMSO (16 mg/mL).[15]The stability of resveratrol is influenced by environmental factors, particularly light, heat, and pH.[16]Exposure to UV light or elevated temperatures promotes isomerization from the trans to the cis form, with the trans isomer being more photosensitive.[17] Under alkaline conditions (pH > 8), rapid cis-trans isomerization occurs, whereas the compound remains stable in acidic to neutralpH ranges (pH 2–8).[18] Thermal degradation is minimal below 100 °C but increases at higher temperatures, potentially leading to oxidation products.[19]Spectroscopic techniques are essential for identification and characterization. In UV-Vis spectroscopy, trans-resveratrol exhibits a characteristic absorption maximum at 306 nm (molar absorptivity ε ≈ 42,000 M⁻¹ cm⁻¹ in methanol or ethanol).[20] The cisisomer shows a hypsochromic shift to around 280 nm.[13]Nuclear magnetic resonance (NMR) spectroscopy confirms the structure; in the ¹H NMR spectrum of trans-resveratrol (in DMSO-d₆), the olefinic protons appear as two doublets at δ 6.94–7.09 (two d, J ≈ 16 Hz), indicative of the E configuration, with aromatic protons in the 6.5–7.5 ppm range and OH signals around 9–10 ppm. For the cis form, the olefinic protons are at δ 6.24 (d, J = 11.7 Hz, H-α) and δ 6.51 (d, J = 11.7 Hz, H-β). These data distinguish the isomers and verify purity.[21][22]
Biosynthesis
Resveratrol is synthesized in plants primarily through the phenylpropanoid pathway, a branch of secondary metabolism that begins with the amino acidphenylalanine. The initial step involves the enzymephenylalanine ammonia-lyase (PAL), which catalyzes the non-oxidative deamination of phenylalanine to form trans-cinnamic acid. This is followed by cinnamate 4-hydroxylase (C4H), a cytochrome P450enzyme, which hydroxylates trans-cinnamic acid at the 4-position to yield p-coumaric acid. Subsequently, 4-coumarate:CoA ligase (4CL) activates p-coumaric acid by ligating it with coenzyme A to produce p-coumaroyl-CoA, a key intermediate.[23][24]The committed step in resveratrol biosynthesis is catalyzed by stilbene synthase (STS), a type III polyketide synthase. STS condenses one molecule of p-coumaroyl-CoA with three molecules of malonyl-CoA in a sequential manner. This process involves two decarboxylation events from the malonyl-CoA units and three Claisen-type condensation reactions, followed by a cyclization and aromatization to form the stilbene backbone of trans-resveratrol. The reaction releases free coenzyme A and carbon dioxide as byproducts, resulting in the 3,5,4'-trihydroxystilbene structure characteristic of resveratrol. This pathway is highly conserved across resveratrol-producing plants and represents a diversion from the flavonoid branch of phenylpropanoid metabolism.[23][25]Biosynthesis of resveratrol is tightly regulated at the transcriptional level, particularly in response to environmental stresses such as ultraviolet (UV) radiation, pathogeninfection, and wounding. Stress signals activate signaling cascades involving mitogen-activated protein kinases (MAPKs), which in turn induce transcription factors like those from the WRKY family. WRKY proteins, such as VvWRKY8 in grapevine, bind directly to W-box elements in the promoter regions of STS genes, either activating or repressing their expression depending on the context. For instance, positive regulators like VvWRKY10 promote STS transcription under UV stress, leading to rapid accumulation of resveratrol as a protective response. This inducible regulation ensures that resveratrol production is energy-efficient and targeted to defense needs.[26][27]Evolutionarily, resveratrol serves as a phytoalexin—a low-molecular-weight antimicrobial compound produced de novo in response to stress—contributing to plant defense against fungi, bacteria, and herbivores. The STS gene family has undergone duplication and diversification in lineages like Vitaceae (e.g., grapes) and Fabaceae (e.g., peanuts), enabling widespread distribution across at least 72 plant species in 31 genera belonging to 21 families. This evolutionary adaptation likely arose from the recruitment of chalcone synthase-like enzymes into the stilbene pathway, providing selective advantages in pathogen-rich environments. Seminal studies highlight how resveratrol's phytoalexin role has been conserved, with orthologous pathways in non-grape species underscoring its ancient origins in plant immunity.[28][29]
Natural Occurrence
In Plants
Resveratrol is a stilbenoid phytoalexin primarily produced by various plant species as a defense mechanism against environmental stresses. It occurs naturally in grapes (Vitis vinifera), peanuts (Arachis hypogaea), berries such as blueberries (Vaccinium corymbosum) and cranberries (Vaccinium macrocarpon), and Japanese knotweed (Polygonum cuspidatum). These plants synthesize resveratrol in response to biotic threats, including fungal infections, and abiotic factors like ultraviolet (UV) radiation and mechanical wounding, thereby enhancing their resistance to pathogens and damage.[3][30][31]In grapes, resveratrol serves as a key phytoalexin, particularly against the fungal pathogen Botrytis cinerea, which causes gray mold disease; its accumulation in infected tissues inhibits fungal growth and limits lesion development. Production is triggered by UV irradiation, which can elevate resveratrol levels up to tenfold, and by wounding, activating stress-response pathways that prioritize antimicrobial compound synthesis. Similar protective roles are observed in peanuts and berries, where resveratrol responds to fungal attacks and UV exposure, contributing to the plant's overall defense arsenal. Japanese knotweed, a prolific producer, accumulates high levels of resveratrol in its roots and rhizomes under stress conditions, aiding survival in invasive environments.[32][33][34]Concentrations of resveratrol vary significantly across plant tissues and are influenced by cultivar, environmental stress, and growth conditions; in grapes, levels are notably higher in skins and seeds (typically 50–100 µg/g in skins) compared to the pulp, which contains negligible amounts. Stress factors like fungal infection or UV exposure can increase concentrations by several fold in responsive cultivars, such as those used in viticulture, while genetic variations among cultivars lead to differences in baseline and inducible levels. In peanuts, resveratrol is concentrated in the seed coats, and in berries, it predominates in the skins, with Japanese knotweed exhibiting some of the highest overall contents among these sources. Although trace amounts have been detected in certain fungi and bacteria, such as endophytic microorganisms associated with resveratrol-producing plants, resveratrol is predominantly plant-derived.[30][11][35]
In Foods and Beverages
Resveratrol occurs naturally in several foods and beverages, with the highest concentrations typically found in products derived from grapes. Red wine is a primary source, containing 0.1–14.3 mg/L of resveratrol, largely extracted from the skins of dark grape varieties during fermentation. Grape juice provides lower but notable levels, up to approximately 1.5 mg/L, depending on the grape type and processing. Peanuts represent another key dietary source, with resveratrol content ranging from 0.02–1.92 mg/kg in raw or roasted forms. Berries, such as blueberries and cranberries, contain 0.1–10 mg/kg on a fresh weight basis, varying by species and ripeness. Other sources include mulberries, apples, plums, cocoa, and pistachios.Food processing significantly influences resveratrol levels and isomer composition. In winemaking, alcoholic fermentation with grape skins enhances the concentration of trans-resveratrol compared to white wines, where skins are removed early, resulting in levels below 2.1 mg/L. Cooking and roastingpeanuts can reduce resveratrol content, as observed in peanut butters where levels are substantially lower than in rawpeanuts due to heat and mechanical processing. In grape juices, the cis-isomer often predominates alongside trans-resveratrol and its glucosides, with cis-piceid averaging 0.79 mg/L in red varieties.Typical daily dietary intake of resveratrol in Western populations ranges from 0.1–5 mg, primarily contributed by wine and grape products, though this varies with consumption patterns such as moderate alcohol intake. In assessments of common foods, average intakes were around 0.46 mg/day for women and 0.63 mg/day for men in European cohorts.
Resveratrol was first isolated in 1940 from the roots of the white hellebore plant, Veratrum grandiflorum O. Loes., by Japanese chemist Michio Takaoka during investigations into the phenolic constituents of this toxic species, which had been linked to teratogenic effects in livestock grazing on related Veratrum plants.[36] Takaoka named the compound "resveratrol," derived from the plant's Japanese name, and described its basic properties, though its full structure was not determined at the time. This initial isolation highlighted resveratrol as one of several stilbene derivatives in the plant, but it attracted little attention beyond botanical chemistry circles.In 1963, resveratrol was independently isolated from the roots of Polygonum cuspidatum Sieb. et Zucc. (also known as Japanese knotweed or Ko-jo-kon in traditional Japanese medicine) by Shigenori Nonomura and colleagues, who elucidated its chemical structure as 3,5,4'-trihydroxystilbene through spectroscopic analysis and comparison with known stilbenes. This confirmation marked a key advancement, establishing resveratrol as a trans-stilbene polyphenol with potential phytoestrogenic properties, though early studies focused primarily on its occurrence in medicinal plants rather than biological activity.[2] The Polygonum cuspidatum source became significant for subsequent extractions due to the plant's higher yield compared to Veratrum.Further progress came in 1976 when Peter Langcake and Roger J. Pryce isolated resveratrol from grapevines (Vitis vinifera) and demonstrated its role as a phytoalexin produced in response to fungal infection or injury, confirming the structure through synthesis and providing the first total synthesis verification. Despite these findings, resveratrol remained obscure in scientific literature through the 1980s, with fewer than a dozen publications annually, as interest centered on its botanical distribution rather than pharmacological potential.[36]
Scientific Development
The scientific interest in resveratrol surged in the early 1990s following the popularization of the "French Paradox," a term coined by epidemiologist Serge Renaud to describe the unexpectedly low rates of coronary heart disease in France despite a diet high in saturated fats, attributed in part to moderate red wine consumption containing resveratrol.[37] This connection was first explicitly proposed in 1992 by researchers Edward Siemann and Leroy Creasy, who identified resveratrol in red wine and suggested its antioxidant properties might explain the protective cardiovascular effects observed in epidemiological data. Building on this, research in the late 1990s and early 2000s explored resveratrol's potential mechanisms, culminating in a landmark 2003 study from David Sinclair's laboratory at Harvard Medical School, which demonstrated that resveratrol activates sirtuin proteins (Sir2 in yeast), extending replicative lifespan in yeast cells by up to 70% through enhanced DNA repair and metabolic regulation.The 2000s marked a period of intense hype around resveratrol, driven by preclinical studies suggesting it mimics the benefits of caloric restriction. A pivotal 2006 study from Sinclair's group at Harvard showed that resveratrol supplementation improved healthspan and extended lifespan in mice fed a high-calorie diet, reducing risks of obesity, diabetes, and age-related diseases by activating sirtuin pathways and improving mitochondrial function. This finding sparked a supplement boom, with resveratrol products flooding the market as an anti-aging elixir, fueled by media coverage and celebrity endorsements; global sales of resveratrol supplements grew rapidly, reaching tens of millions in annual revenue by the late 2000s.[38]However, the 2010s brought significant controversies and setbacks, tempering the enthusiasm. Key claims about resveratrol's direct activation of sirtuin-1 (SIRT1) were challenged; a 2005 study by Matt Kaeberlein and colleagues argued that resveratrol's effects in yeast were indirect, not via allosteric activation of Sir2, leading to debates over its mechanism. Further scrutiny arose from retractions of high-profile papers, including over 20 studies by researcher Dipak Das in 2012 due to image manipulation and data fabrication related to resveratrol's cardioprotective effects, eroding trust in some sirtuin-related claims. Despite this, foundational findings on resveratrol's metabolic benefits persisted, prompting a shift toward more rigorous validation.Key milestones in resveratrol research include the initiation of human clinical trials around 2003, with early pharmacokinetic studies confirming its absorption but highlighting rapid metabolism. By the 2020s, attention has focused on clinical limitations, particularly its poor oral bioavailability—often less than 1% due to extensive first-pass metabolism into glucuronides and sulfates—necessitating formulation innovations like micronization or co-administration with piperine to enhance efficacy.[39] A GRAS notice for trans-resveratrol (GRN 224) was submitted to the U.S. FDA in 2007 for use as a food ingredient, but FDA ceased evaluation at the notifier's request. The supplement market has continued to expand, projected to reach approximately $142 million globally by 2025, driven by interest in its potential for metabolic and longevity support.[38] Ongoing research as of 2025 includes advanced nanoformulations and combination therapies to address bioavailability challenges in clinical settings.[40]
Pharmacology
Pharmacokinetics
Resveratrol is rapidly absorbed from the gastrointestinal tract after oral administration, with studies indicating that 70–80% of the dose is absorbed in the intestine via passive diffusion. However, in humans, the oral bioavailability of the parent compound is low, ranging from less than 1% to approximately 5%, primarily due to extensive presystemic metabolism in the gut and liver. Peak plasma concentrations of resveratrol are typically reached within 30–60 minutes post-ingestion.[41][42][43]Following absorption, resveratrol distributes widely throughout the body, with high binding to plasma proteins such as albumin, reported at approximately 98%. Tissue concentrations are notably higher in the liver, kidney, and heart compared to plasma levels, reflecting preferential accumulation in these organs. While resveratrol is lipophilic and capable of crossing the blood-brain barrier, its penetration into the central nervous system is limited by low systemic exposure and rapid biotransformation.[44][45][46]The parent resveratrol undergoes rapid elimination, exhibiting a short plasmahalf-life of about 9–14 minutes in humans. In contrast, its major metabolites, including glucuronides and sulfates, persist longer, with half-lives extending to several hours. Elimination occurs predominantly through fecal excretion (up to 98% of the dose in some animal models, with significant biliary involvement in humans), following phase II conjugation, while urinary excretion accounts for a smaller portion, primarily as metabolites.[47][43][48]Pharmacokinetics of resveratrol are influenced by several factors, including dose, where higher doses result in nonlinear increases in exposure due to saturation of metabolic pathways. The food matrix affects absorption, with co-ingestion of fats enhancing bioavailability by improving solubility. Specialized formulations, such as micronized powders or lipid-based carriers, can significantly improve oral bioavailability by mitigating first-pass effects and increasing gastrointestinal uptake.[39][49][50]
Pharmacodynamics
Resveratrol exerts its biological effects primarily through interaction with multiple molecular targets and signaling pathways at the cellular level. As a polyphenolic compound, it modulates key enzymes and transcription factors involved in cellular stress responses, energy homeostasis, and inflammation. These actions contribute to its pleiotropic effects observed in various experimental models.[4]One of the primary targets of resveratrol is sirtuin 1 (SIRT1), an NAD+-dependent deacetylase that regulates gene expression and cellular processes mimicking calorie restriction. Resveratrol activates SIRT1 by lowering its Michaelis constant for both acetylated substrates and NAD+, thereby enhancing deacetylation activity at low micromolar concentrations. This activation promotes longevity-associated pathways in yeast, worms, and mammalian cells.[51] Additionally, resveratrol phosphorylates AMP-activated protein kinase (AMPK), a central regulator of energy homeostasis, through mechanisms involving increased cytosolic calcium and activation of calcium/calmodulin-dependent protein kinase kinase-β, as well as LKB1-dependent pathways. AMPK activation inhibits anabolic processes and stimulates catabolism, enhancing mitochondrial function.87306-2/fulltext)In terms of antioxidant mechanisms, resveratrol upregulates the Nrf2 pathway, a master regulator of cellular defense against oxidative stress. It promotes Nrf2 nuclear translocation and activation of antioxidant response elements, leading to increased expression of genes encoding enzymes such as heme oxygenase-1 and NAD(P)H quinone dehydrogenase 1. Resveratrol also directly scavenges reactive oxygen species (ROS), reducing oxidative damage in cellular models. Complementing this, resveratrol exhibits anti-inflammatory effects by inhibiting the NF-κB pathway, suppressing its nuclear translocation and DNA binding activity, which in turn reduces pro-inflammatory cytokine production. This inhibition occurs independently of IκB degradation in some contexts.[52][53]Resveratrol further modulates other pathways, acting as a weak phytoestrogen by binding to estrogen receptors (ERα and ERβ) with mixed agonist/antagonist properties, depending on the cellular context and co-activators present. This interaction influences gene transcription related to cell proliferation and survival without strong estrogenic effects in vivo. It also activates endothelial nitric oxide synthase (eNOS) through phosphorylation at Ser1177 and upregulation of its expression, promoting nitric oxide production and vasodilation in endothelial cells. These effects are mediated via PI3K/Akt signaling and SIRT1-dependent deacetylation.[54]The pharmacodynamic effects of resveratrol often follow a hormetic dose-response curve, where low doses (typically 1–10 µM in vitro) elicit beneficial stimulatory responses through adaptive stress pathways, while high doses (>50 µM) may inhibit these effects or induce toxicity by overwhelming cellular defenses. This biphasic pattern is observed across models affecting endpoints like cell survival and antioxidantenzyme induction.[55]
Metabolism
Resveratrol is primarily metabolized through phase II conjugation reactions in the human liver and small intestine, where it undergoes rapid glucuronidation and sulfation shortly after absorption. Glucuronidation is catalyzed by UDP-glucuronosyltransferase enzymes, including UGT1A1 and UGT1A3, which predominantly form resveratrol-3-O-glucuronide, while UGT1A9 contributes to the production of resveratrol-4'-O-glucuronide. Sulfation occurs mainly via the sulfotransferase SULT1A1, yielding resveratrol-3-O-sulfate as a key metabolite. These conjugations represent the dominant metabolic pathways, with studies showing that over 90% of absorbed resveratrol is converted to these phase II metabolites within hours of oral administration.In addition to phase II processes, resveratrol undergoes limited phase I oxidation, primarily through hydroxylation mediated by cytochrome P450 enzymes such as CYP1B1, which converts it to piceatannol by adding a hydroxyl group at the 4-position of the B-ring. Gut microbiota also play a role in metabolism, reducing resveratrol to dihydroresveratrol via microbial ene-reductases and other enzymes in the intestinal lumen. Some of these metabolites, including the glucuronides, retain partial bioactivity comparable to the parent compound in assays for antioxidant, anti-inflammatory, and estrogenic effects, suggesting they may contribute to resveratrol's overall physiological impact.Enterohepatic recirculation of conjugated metabolites, such as resveratrol glucuronides and sulfates, extends systemic exposure by allowing biliary excretion followed by reabsorption in the intestines, as evidenced in rat models and inferred in human pharmacokinetics. Interspecies differences in metabolism are notable, with phase II conjugation occurring more slowly in rodents compared to humans, leading to higher plasma levels and prolonged half-lives of resveratrol in mice and rats, which can affect the translation of preclinical findings to human applications.
Health Research
Cardiovascular Effects
Resveratrol exerts protective effects on the cardiovascular system primarily through endothelial protection and modulation of lipid profiles. It enhances endothelial nitric oxide (NO) production by upregulating endothelial nitric oxide synthase (eNOS) expression and activity, while also reducing oxidative stress that degrades NO, thereby improving vasodilation and vascular function.[56] Additionally, resveratrol inhibits low-density lipoprotein (LDL) oxidation and promotes high-density lipoprotein (HDL) levels, contributing to reduced atherogenic risk and improved lipidhomeostasis.[57]In preclinical models, resveratrol has demonstrated robust anti-atherosclerotic effects. Animal studies, including those in high-fat diet-fed rabbits and apolipoprotein E-deficient mice, show that resveratrol supplementation reduces plaque formation, decreases intimal thickening, and attenuates vascular inflammation by downregulating pro-atherogenic pathways such as NF-κB signaling.[58] These findings highlight its potential in preventing atherosclerosis progression through antioxidant and anti-inflammatory mechanisms.[59]Clinical evidence on resveratrol's cardiovascular benefits remains mixed, with modest improvements observed in specific outcomes. A 2014 meta-analysis of randomized controlled trials involving 247 participants receiving doses above 150 mg daily reported significant reductions in systolic blood pressure (SBP), averaging 2-5 mmHg, particularly at higher doses (1-8 g).[60] However, broader reviews indicate inconsistent effects on overall blood pressure and lipid parameters across human trials, with benefits more pronounced in hypertensive or dyslipidemic subgroups.[60] A 2023 systematic review further noted limited but positive impacts on endothelial function and cardiac remodeling in patients with hypertension, though larger trials are needed for confirmation.[61]Recent investigations from 2024-2025 have explored resveratrol's role in modulating the gut-liver axis to address dyslipidemia. In high-fat diet-induced models, resveratrol preserved gut mucosal integrity, reduced hepatic oxidative stress, and improved lipid profiles by altering microbiota composition and enhancing bile acid metabolism, suggesting indirect cardiovascular benefits through metabolic regulation.[62] A 2025 comprehensive review of dietary polyphenols corroborated these findings, emphasizing resveratrol's potential in ameliorating dyslipidemia via gut-liver interactions in preclinical and early human studies.[63]
Anticancer Potential
Resveratrol has demonstrated potential in cancer prevention and treatment through multiple mechanisms that target key oncogenic processes. It induces cell cycle arrest primarily by activating the tumor suppressor protein p53, which halts progression at the G1/S phase in various cancer cells. Additionally, resveratrol promotes apoptosis by downregulating anti-apoptotic proteins such as Bcl-2 and upregulating pro-apoptotic factors like Bax and caspases. It also inhibits angiogenesis by suppressing vascular endothelial growth factor (VEGF) expression and signaling, thereby limiting tumor vascularization and metastasis. These effects have been observed across diverse cancer types, including breast, colon, and prostate cancers, highlighting resveratrol's pleiotropic anticancer activity.[64]In preclinical models, resveratrol exhibits dose-dependent antiproliferative effects, with IC50 values typically ranging from 10 to 50 µM in breast (e.g., MCF-7 cells), colon (e.g., HCT-116 cells), and prostate (e.g., PC-3 cells) cancer lines. Animal studies further support these findings, showing reduced tumor volume and incidence in xenograft models of these cancers when administered orally or intraperitoneally at doses of 10-50 mg/kg. Resveratrol also synergizes with chemotherapeutic agents, such as doxorubicin, enhancing cytotoxicity and overcoming multidrug resistance in breast and gastric cancer models by modulating efflux pumps and epithelial-mesenchymal transition pathways. For instance, combination therapy lowers the required doxorubicin dose while amplifying apoptosis and reducing tumor growth by up to 70% in vitro and in vivo.[64][65][66][67]Clinical translation of resveratrol's anticancer potential remains limited, with Phase I and II trials establishing safety at doses up to 5 g/day in cancer patients, reporting mild gastrointestinal side effects but no severe toxicity. A 2024 systematic review of adjunctive use in solid tumors indicated modest improvements in response rates when combined with chemotherapy, but no standalone efficacy or regulatory approval for cancer treatment. Challenges include poor bioavailability and variable pharmacokinetics, necessitating higher doses that may not achieve therapeutic tissue levels.[68][69][70]Recent 2025 research has uncovered novel insights into resveratrol's modulation of the gut microbiome to enhance antitumor immunity. In pancreatic cancer models, resveratrol alters microbial composition to increase prostaglandin D2 (PGD2) production, amplifying the efficacy of anti-PD-1 checkpoint inhibitors by boosting CD8+ T-cell infiltration and reducing tumor burden by over 50%. This microbiome-immune axis suggests potential for resveratrol as an immunomodulatory adjunct in immunotherapy-resistant cancers.[71]
Diabetes and Metabolic Effects
Resveratrol has been investigated for its potential to improve glucose regulation and insulin sensitivity through several molecular mechanisms. One key pathway involves the activation of AMP-activated protein kinase (AMPK), which promotes the translocation of glucose transporter type 4 (GLUT4) to the cell membrane, enhancing glucose uptake in insulin-resistant tissues such as skeletal muscle.[72] This effect is particularly evident in models of high-insulin or free fatty acid-induced insulin resistance, where resveratrol restores GLUT4 function via AMPK and related signaling like Akt and IRS-1.[73] Additionally, resveratrol protects pancreatic β-cells from oxidative stress and dedifferentiation, preserving insulin secretion capacity in diabetic conditions.[74] It also modulates adipokine profiles by decreasing leptin expression and secretion while increasing adiponectin levels, thereby improving insulin sensitivity and reducing adipose tissue inflammation.[75] These actions are partly linked to resveratrol's activation of SIRT1, a deacetylase that influences metabolic gene expression.[76]In animal models of type 2 diabetes and obesity, resveratrol supplementation consistently reverses insulin resistance by lowering hepatic glucose production and enhancing peripheral insulin sensitivity.[77] For instance, in high-fat diet-fed rodents and swine, it improves glucose homeostasis, attenuates β-cell loss, and reduces oxidative stress in pancreatic islets.[78] Human studies support these findings, with a 2021 meta-analysis of randomized controlled trials indicating that resveratrol at doses around 500 mg/day significantly reduces HbA1c levels by approximately 0.5% in patients with type 2 diabetes, alongside improvements in fasting glucose and insulin resistance indices.[79] An updated analysis in 2022 confirmed these glycemic benefits, emphasizing dose-dependent effects without major adverse events.[80]Regarding obesity-related metabolism, emerging 2025 preclinical trials highlight resveratrol's influence on gut microbiota as a mechanism for anti-obesity effects, with supplementation in high-fat diet models leading to 5-10% body weight reduction through enhanced microbial diversity and short-chain fatty acid production.[81] This modulation helps mitigate visceral fat accumulation and improves leptin sensitivity in the hypothalamus.[82] In the context of metabolic syndrome, cohort studies demonstrate that resveratrol improves dyslipidemia by lowering total cholesterol and triglycerides, while reducing inflammatory markers like C-reactive protein in affected individuals.[83] These benefits extend to broader inflammation control, supporting its role in managing metabolic dysregulation.[84]
Neurological Effects
Resveratrol demonstrates neuroprotective effects through several mechanisms in the central nervous system, though its direct penetration of the blood-brain barrier (BBB) is limited, primarily occurring via its metabolites such as resveratrol-3-glucuronide and resveratrol-3-sulfate. These metabolites have been detected in cerebrospinal fluid following oral administration, enabling central effects despite resveratrol's poor bioavailability.[85][86]In preclinical models, resveratrol promotes the clearance of amyloid-β peptides via activation of autophagy pathways, reducing amyloid plaque formation. It also inhibits tau hyperphosphorylation by enhancing protein phosphatase 2A activity and interfering with kinase signaling, thereby mitigating neurofibrillary tangle development. These actions contribute to synaptic preservation and reduced neuronal apoptosis in cellular and animal models of neurodegeneration.[87][88][89]Regarding Alzheimer's disease, rodent studies consistently show that resveratrol reduces amyloid-β burden and plaque accumulation in the hippocampus, improving spatial memory and cognitive performance. In human trials, a randomized, double-blind, placebo-controlled study demonstrated that resveratrol treatment stabilized disease progression biomarkers and improved activities of daily living scores in mild-to-moderate cases, with evidence from a 2023meta-analysis indicating enhancements in cognitive function, including trends toward better Mini-Mental State Examination (MMSE) scores.[90][91]For general cognition, a randomized controlled trial in healthy elderly participants found that 200 mg/day of resveratrol for 26 weeks enhanced memoryperformance and hippocampal functional connectivity, alongside reductions in neuroinflammatory markers such as interleukin-6 (IL-6). These effects suggest potential benefits in preventing age-related cognitive decline.[92][93]In Parkinson's disease models, resveratrol protects dopaminergic neurons by preserving dopamine levels, reducing oxidative stress, and inhibiting microglial activation, as evidenced in MPTP-induced rodent paradigms. Human data remain limited, with ongoing trials exploring its safety but no definitive efficacy established yet. Similarly, in Huntington's disease models, resveratrol attenuates mutant huntingtin toxicity, improves motor coordination, and activates neuroprotective pathways like ERK signaling, though clinical translation is preliminary.[94][95][96]
Aging and Lifespan Extension
Resveratrol has been extensively studied for its potential to mimic the effects of caloric restriction (CR), a dietary intervention known to extend lifespan in various organisms by activating pathways that promote cellular resilience and metabolic efficiency. As a CR mimetic, resveratrol activates sirtuin 1 (SIRT1), a NAD+-dependent deacetylase that regulates gene expression associated with longevity. Specifically, SIRT1 deacetylates peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), enhancing its activity and thereby stimulating mitochondrial biogenesis, which improves energymetabolism and reduces oxidative stress during aging. Additionally, resveratrol supports telomere maintenance by upregulating telomerase activity and preserving telomere length, counteracting the progressive shortening that contributes to cellular senescence and age-related decline.[97][98][99]In preclinical models, resveratrol consistently extends lifespan across species. Early studies demonstrated lifespan prolongation in yeast (Saccharomyces cerevisiae) by up to 70% through SIRT1 ortholog activation, while in nematodes (Caenorhabditis elegans) and fruit flies (Drosophila melanogaster), extensions of 20-30% were observed via improved stress resistance and metabolic shifts. A pivotal 2006 study by Baur et al., from David Sinclair's laboratory, showed that resveratrol extended lifespan by approximately 30% in mice fed a high-calorie diet, delaying age-related pathologies such as insulin resistance and inflammation without affecting body weight. These findings positioned resveratrol as a key modulator of aging hallmarks, including mitochondrial dysfunction and genomic instability.[100][101][102]Human evidence, however, remains preliminary and focused on aging biomarkers rather than direct lifespan extension, as long-term trials are infeasible. Reviews from 2024 and 2025 highlight improvements in senescence markers, such as reduced expression of p16 and p21 in peripheral blood mononuclear cells following oral supplementation, indicating delayed cellular aging. Topical resveratrol formulations have shown efficacy in skin aging trials, with clinical studies reporting 10-20% reductions in wrinkle depth and improved elasticity after 4-12 weeks of application, attributed to enhanced collagen synthesis and antioxidant protection. Despite these biomarker shifts, no human data demonstrate lifespan extension, and effects on systemic aging processes require further validation.[103][70][104]Controversies surround resveratrol's translational potential, primarily due to challenges in achieving effective doses in humans equivalent to those in animal models, where supraphysiological levels (e.g., 200-400 mg/kg in mice) exceed safe oral bioavailability in people. These issues underscore the need for optimized formulations to enhance absorption and confirm benefits in human aging.[70][105]
Other Potential Benefits
Resveratrol has demonstrated hepatoprotective effects in models of non-alcoholic fatty liver disease (NAFLD), particularly by reducing elevated liver enzymes and mitigating oxidative stress. In a 2025 study using high-fat diet (HFD)-induced NAFLD rats, resveratrol supplementation significantly lowered serum alanine aminotransferase (ALT) levels by approximately 25%, alongside reductions in aspartate aminotransferase (AST), through enhanced antioxidant enzyme activity such as superoxide dismutase and catalase. This treatment also preserved hepatic glutathione levels, counteracting lipid peroxidation and oxidative damage in liver tissues.[62]The compound exhibits antimicrobial properties, notably by inhibiting bacterial biofilm formation. Resveratrol disrupts biofilm development in Staphylococcus aureus at concentrations 3-4 times below its minimum inhibitory concentration, interfering with extracellular matrix components like polysaccharide intercellular adhesin (PIA) and extracellular DNA release. A 2025 in vitro study confirmed that resveratrol reduces S. aureus biofilm formation by up to 70% via decreased reactive oxygen species production and quorum sensing inhibition. Additionally, resveratrol shows antiviral potential as an adjunct therapy; in 2024 clinical trials for COVID-19 patients, it reduced inflammatory markers such as C-reactive protein and interleukin-6, supporting its role in alleviating cytokine storms without direct antiviral replication inhibition.[106][107][108]Resveratrol influences the gut microbiome in ways that may aid anti-obesity efforts, particularly through modulation of bacterial phyla ratios and the gut-liver axis. Supplementation shifts the Firmicutes/Bacteroidetes ratio toward a healthier profile by increasing Bacteroidetes abundance, which correlates with reduced gut permeability and improved bile acid metabolism in obese models. A 2025 investigation highlighted resveratrol's role in the gut-liver axis, where it restored mucosal integrity in HFD-fed rats, decreasing lipopolysaccharide translocation to the liver and thereby lowering endotoxemia-driven inflammation. These microbiome alterations enhance short-chain fatty acid production, supporting metabolic homeostasis without directly targeting adiposity.[109][62]In inflammatory conditions beyond core metabolic pathways, resveratrol provides relief in experimental endometriosis models by suppressing lesion growth and vascularization. Animal studies show that resveratrol administration reduces the number and volume of endometrial implants by 40-60% in rats, attributed to downregulation of vascular endothelial growth factor and matrix metalloproteinase-9 expression in endometrial tissues. For obesity-related inflammation, resveratrol attenuates adipose tissue macrophage infiltration and pro-inflammatory cytokine release, such as tumor necrosis factor-alpha, in HFD-induced models, thereby improving systemic inflammatory profiles without altering core glucose handling.[110][111][112]
Safety and Adverse Effects
Toxicity and Side Effects
Resveratrol exhibits low acute toxicity in animal models. In rats, the oral median lethal dose (LD50) exceeds 5000 mg/kg body weight, indicating minimal risk of lethality from single high exposures.[113] No cases of humanlethality have been reported, even at doses up to 5 g in clinical settings.[114]Common side effects in humans primarily involve gastrointestinal disturbances, such as nausea and diarrhea, observed at doses exceeding 1 g per day. These effects are dose-dependent and typically mild, resolving upon discontinuation.[115] Resveratrol may also exhibit weak estrogenic activity, prompting caution in individuals with hormone-sensitive conditions like breast or uterine cancers, where it could potentially mimic estrogen and exacerbate symptoms.[115] Rare reports suggest possible estrogen-like effects, though specific instances such as hot flashes in women remain uncommon and unconfirmed in large trials.[116]Chronic exposure data from clinical trials support the safety of resveratrol up to 5 g per day for durations of up to 6 months, with no significant adverse events beyond transient gastrointestinal issues. A 2024 systematic review of human intervention trials confirmed tolerability across various doses and confirmed no evidence of genotoxicity or carcinogenicity in these studies.[117] Animal studies align with this, showing no oncogenic potential at relevant exposures.[118]Special precautions apply to vulnerable populations. In pregnancy, while human data are limited, animal and primate studies indicate potential risks at high doses, including fetal abnormalities and prolonged gestation, warranting avoidance during pregnancy and lactation.[119] Similarly, individuals with hormone-sensitive conditions should consult healthcare providers due to resveratrol's estrogenic properties.[116]
Drug Interactions
Resveratrol exhibits pharmacokinetic interactions primarily through its moderate inhibition of cytochrome P450 enzymes, notably CYP3A4 and CYP2C9, which can alter the metabolism and increase plasma concentrations of co-administered drugs that are substrates for these enzymes.[120] For instance, resveratrol inhibits CYP3A4-mediated metabolism, potentially elevating levels of statins such as simvastatin and atorvastatin, thereby heightening the risk of statin-related adverse effects like myopathy.[121] Similarly, as a CYP2C9 inhibitor, resveratrol increases the systemic exposure of S-warfarin by approximately 49%, enhancing its anticoagulant effects and prolonging prothrombin time and activated partial thromboplastin time in animal models.[122]In terms of pharmacodynamic interactions, resveratrol's antiplatelet properties amplify the bleeding risk when combined with anticoagulants and antiplatelet agents. It inhibits platelet aggregation, synergizing with drugs like warfarin, aspirin, and clopidogrel to potentially increase bruising and hemorrhage incidence.[114] Clinical guidelines recommend that patients on these therapies consult healthcare providers before using resveratrol supplements to monitor international normalized ratio (INR) and bleeding parameters.[123]Resveratrol also interacts with other compounds through synergistic or modulatory effects. It enhances the antioxidant activity of quercetin, a flavonoid, by promoting greater inhibition of oxidative stress in cellular models compared to either compound alone, potentially boosting overall free radical scavenging.[124] Regarding tamoxifen, resveratrol's mixed estrogen receptor agonist/antagonist profile may modulate its efficacy in breast cancer treatment, acting as a selective estrogen receptor modulator that sensitizes antiestrogen-resistant cells while overlapping in estrogenic signaling pathways.[125]Due to CYP3A4 inhibition, resveratrol may theoretically elevate tacrolimus levels in transplant patients, necessitating therapeutic drug monitoring to avoid toxicity.[120]
Related Compounds
Stilbenoids
Stilbenoids are a class of polyphenolic compounds characterized by a 1,2-diphenylethene backbone, with resveratrol (3,5,4'-trihydroxystilbene) serving as a key representative naturally produced in plants as a phytoalexin. Among the structurally related stilbenoids occurring alongside resveratrol, piceid (trans-resveratrol-3-O-β-D-glucopyranoside) stands out as the predominant glycosylated form in grape skins and derived products like wine. In grapes and grape juice, piceid constitutes the major stilbene, often present at concentrations typically 4- to 10-fold higher than the free aglycone resveratrol, depending on grape variety and environmental factors.[126][114] In red wines, total stilbene levels average around 4-5 mg/L, where piceid accounts for the majority, often exceeding resveratrol by 5- to 20-fold in certain cultivars.[127] Upon ingestion, piceid undergoes hydrolysis in the gastrointestinal tract, primarily mediated by β-glucosidase enzymes from gut microbiota such as Bifidobacterium infantis, converting it to the bioactive aglycone resveratrol for absorption.[128]Another notable stilbenoid co-occurring with resveratrol is piceatannol (trans-3,5,4',3'-tetrahydroxystilbene), a tetra-hydroxylated derivative featuring an additional hydroxyl group on the B-ring compared to resveratrol. Piceatannol is found in grapes, particularly in the skins of Vitis vinifera varieties, and is especially abundant in passion fruit (Passiflora edulis) seeds, where it can reach concentrations up to several milligrams per gram of extract.[129] This compound exhibits enhanced biological potency relative to resveratrol, notably as a more effective activator of SIRT1, a NAD+-dependent deacetylase involved in cellular stress responses, with studies demonstrating superior upregulation of SIRT1 expression and downstream signaling at equivalent concentrations.[130][131]These stilbenoids share overlapping antioxidant properties, scavenging reactive oxygen species through phenolic hydroxyl groups, with piceatannol displaying particularly potent radical-quenching activity in assays like DPPH and ABTS, often surpassing resveratrol due to its additional hydroxylation.[132] They frequently co-occur in dietary sources such as red wine, where resveratrol, piceid, and trace piceatannol contribute to the overall polyphenol profile, with levels varying by vinification processes and grape stress responses.[133] Biosynthetically, piceid, piceatannol, and resveratrol derive from the phenylpropanoid pathway in plants, converging at stilbene synthase (STS), which catalyzes the condensation of p-coumaroyl-CoA and malonyl-CoA to form the stilbene core; subsequent glycosylation yields piceid, while cytochrome P450-mediated hydroxylation converts resveratrol to piceatannol.[134] This shared STS-dependent route underscores their coordinated accumulation in response to fungal elicitors or UV stress in grapevines.[135]
Derivatives and Analogs
To address the pharmacokinetic challenges of resveratrol, such as its low oral bioavailability due to rapid metabolism and poor solubility, researchers have developed micronized formulations like SRT501, which reduce particle size to less than 5 μm, resulting in a 3- to 4-fold increase in plasma concentration and area under the curve (AUC) compared to unprocessed resveratrol.[136] This nanoparticle-based approach enhances absorption by increasing surface area for dissolution, allowing higher systemic exposure in preclinical and early clinical studies.[42]Among synthetic analogs, pterostilbene, a dimethylated derivative of resveratrol, exhibits significantly improved bioavailability—approximately 80% versus 20% for resveratrol—owing to its greater lipophilicity, which facilitates better cellular uptake and slower metabolism, leading to a longer half-life of about 105 minutes compared to resveratrol's 14 minutes.[137] This analog has shown preclinical superiority in cancer models, including enhanced inhibition of tumor growth and reduced metastasis in prostate and breast cancer cell lines, attributed to its sustained activation of SIRT1 pathways.[138] Another notable analog, oxyresveratrol, features an additional hydroxyl group on the stilbene backbone, conferring stronger tyrosinase inhibition with an IC50 of 1.2 μM—32-fold more potent than resveratrol—making it a promising candidate for skinhyperpigmentation treatments through reduced melanin synthesis.[139]Further developments include ester conjugates such as resveratrol ferulate esters, synthesized by linking resveratrol to ferulic acid to improve stability and antioxidant capacity, as demonstrated in vitro where these compounds exhibited enhanced free radical scavenging compared to the parent molecule.[140] These modifications aim to reduce first-pass metabolism and extend half-life, with preclinical data indicating superior efficacy in oxidative stress models. Amino acid carbamate prodrugs of resveratrol explore brain-targeted delivery systems to overcome the blood-brain barrier, potentially enhancing neuroprotective effects in neurodegenerative diseases by improving central nervous system penetration.[141] Overall, these derivatives offer advantages like prolonged circulation and targeted potency, though clinical translation remains limited by ongoing safety evaluations.