Next Article in Journal
Fine Particulate Matter (PM2.5) Disrupts Intestinal Barrier Function by Inducing Oxidative Stress and PI3K/AKT-Mediated Inflammation in Caco-2 Cells
Previous Article in Journal
Correction: Wróbel et al. The GPR39 Receptor Plays an Important Role in the Pathogenesis of Overactive Bladder and Corticosterone-Induced Depression. Int. J. Mol. Sci. 2024, 25, 12630
Previous Article in Special Issue
Variation in the Chemical Composition of Small Cranberry (Vaccinium oxycoccos L.) Fruits Collected from a Bog-Type Habitat in Lithuania
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Stilbenes from Vine Extracts: Therapeutic Potential and Mechanisms

by
Luís P. Brás
1,2,
Ângelo Luís
1,
Gregory Chatel
2,*,
Sílvia Socorro
1 and
Ana Paula Duarte
1,*
1
RISE-Health, Department of Medical Sciences, Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
2
EDYTEM, CNRS, University Savoie Mont Blanc, F-73000 Chambéry, France
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(17), 8269; https://doi.org/10.3390/ijms26178269
Submission received: 18 July 2025 / Revised: 12 August 2025 / Accepted: 21 August 2025 / Published: 26 August 2025
(This article belongs to the Special Issue Recent Advances in Medicinal Plants and Natural Products)

Abstract

The wine industry represents a significant economic sector; however, it generates large volumes of waste that can be valorized due to the presence of bioactive compounds, particularly stilbenes. These naturally occurring stilbenes exhibit remarkable potential in the prevention and treatment of various diseases, including cardioprotection, neuroprotection, antidiabetic properties, anti-inflammatory activity, and cancer prevention and therapy. This review discusses biosynthesis, structures, extraction methods, and mechanisms of action of stilbenes, with a particular emphasis on cancer prevention and treatment. Evidence from in vitro, in vivo, and clinical studies demonstrate that stilbenes modulate multiple molecular pathways by promoting apoptosis, inhibiting cell proliferation, and regulating inflammation, oxidative stress, and metabolism. However, the clinical application of stilbenes is limited by their low bioavailability. To overcome this, pharmaceutical formulations have been developed to enhance their stability and bioavailability, reduce side effects, and improve target interactions. These advances are expected to increase the therapeutic efficacy of stilbenes. Furthermore, information on the health benefits of less common stilbenes remains limited, highlighting the need for further research on these compounds.

1. Introduction

Viticulture is one of the most economically and culturally significant agricultural sectors worldwide. Wine is among the most widely produced and consumed beverages, with 94% of global production concentrated in just 29 countries [1]. Global consumption has reached approximately 221 million hectolitres, according to the International Organisation of Vine and Wine (OIV) [2]. Alongside this economic value, the wine industry generates large volumes of waste and byproducts estimated at 20–30% of the initial grape mass during winemaking [3,4].
These residues arise at two key stages: during harvesting (solid waste) and during processing (liquid waste) [5]. Solid waste includes grape pomace (45%), grape stalks (7.5%), grape pips (6%), grape marc (30%), and yeast (41.5%) [6,7,8]. Yeast lees alone account for ~5% of grape weight and contain ethanol, tartaric acid, phenolic compounds, and microbial biomass [9]. The fermentation and pressing steps contribute to an additional 20–25% of total waste [10].
In addition to winery waste, vineyard maintenance also contributes significantly to biomass generation. Annual pruning, essential for balanced plant development, produces an estimated 6–18 million tons of biomass each year, including shoots, branches, and leaves [11,12]. This accumulation results from vineyard renewal aimed at ensuring balanced growth. However, this material is often underutilized despite its potential for bio-based valorization. The biomass derived from vine waste and grape byproducts contains bioactive molecules important for human health [13].
While viticulture brings substantial economic benefit to many countries [14], the growth in grape production has led to increased vineyard surface area and value, along with a rise in the accumulation of both organic and inorganic waste [15,16]. This poses challenges to both economic and ecological management.
From an economic standpoint, inefficiencies persist, i.e., energy from renewable sources is poorly recycled, large quantities of waste are generated, and passive architectural strategies to optimize space and energy efficiency are underused [15,16,17]. Ecologically, the accumulation of waste increases the sector’s environmental footprint, which can be quantified using life cycle assessment (LCA) methodologies [18,19].
Despite these constraints, grapevines and their byproducts are rich in bioactive phytochemicals of potential value to the food, pharmaceutical, and cosmetics industries. These include dietary fiber, phenolics, proteins, and lipids [9,20,21,22,23,24,25,26], as well as stilbenes, which show promise as natural additives, antimicrobial and antioxidant agents, and even fillers in sustainable food packaging materials [15,27,28,29]. Leaves, in particular, contain organic acids, lipids, and polyphenols, and are already exploited in cosmetics [15,30]. Additionally, grapevine residues can be valorized as soil fertilizers or for bioenergy production [31].
The profile of polyphenols present in grape-based materials is subject to considerable fluctuation, reflecting not only genetic diversity and environmental variation, but also the interplay of cultivation and processing strategies. Each step from grape ripening to yeast selection shapes the ultimate spectrum of these compounds, with maceration time standing out for its impact on concentration [16,19]. Despite being a rich reservoir of bioactives like flavonols and anthocyanins, the grape skin’s direct contribution to wine composition is intentionally moderated; winemakers routinely remove these skins early to balance sensory qualities, inadvertently discarding a source of potential nutritional and therapeutic interest. This reality raises the question of how conventional practices might be optimized to both reserve product appeal and maximize the exploitation of health-promoting polyphenols, a challenge still largely unaddressed in mainstream oenology [28].
Grape seeds also contain high concentrations of antioxidants, including fiber, proteins, carbohydrates, lipids, minerals, phytosterols, phenolic compounds, vitamin E, and melatonin [15,32]. These polyphenols display activities relevant to stress response and chronic diseases. Notably, they intervene in cancer-related mechanisms such as tumor cell proliferation, metastasis, and drug resistance [33,34].
Among polyphenols, stilbenes are particularly interesting. As phytoalexins, they protect grapevines from pathogens and are mainly located in woody tissues like pruning canes and stalks [35]. The best known stilbene, trans-resveratrol (5-[(1E)-2-(4-hydroxyphenyl)ethenyl]-1,3-benzenediol; CAS: 501-36-0), has been extensively studied for its antioxidant, cardioprotective, and anticancer properties [36]. Stilbenes are ideal candidates for high-value valorization in the field of cancer prevention and anticancer therapy since they are found in pruning waste [15].
This review explores the therapeutic potential of vine-derived stilbenes particularly those found in pruning residues as anticancer agents. It covers their biosynthesis, extraction, chemical structures, biological activities, and mechanisms of action, while also addressing current limitations and future perspectives for their medical application.

2. Stilbenes from Grapevine: Biosynthesis, Structures, and Extraction

2.1. Polyphenols in Grapevine: General Classification and Biological Roles

Polyphenols, fundamentally, are the product of plant metabolism resulting in molecules that incorporate multiple phenolic motifs [37]. The chemical identity of a phenol, a compound consisting of an aromatic ring bearing at least one hydroxyl group, serves as the foundational structure for this diverse family. However, reducing these compounds to a structural formula overlooks the evolutionary rationale behind their biosynthesis, as well as their often-underestimated functional relevance in plant defense and, potentially, in human health [38].
The sheer heterogeneity of polyphenols, manifesting across grapevine tissues from root to leaf, defies easy classification. While chemists typically subdivide them by structure, highlighting families such as stilbenes or flavonoids, this approach captures only part of a dynamic and context-dependent reality [39,40]. Grape-derived plant material alone is home to a remarkable spectrum of compounds, containing not only well-characterized subclasses but also a long list of less-understood entities. This taxonomic richness, while scientifically intriguing, complicates efforts to standardize bioactive ingredient extraction for practical use [35].
Flavonoids are a class of polyphenols and are defined by a common structure composed of two aromatic rings joined by an oxygenated heterocyclic ring. Based on this core, they are subdivided into nine subclasses: chalcones, aurones, flavones, isoflavones, flavanols, anthocyanins, isoflavonoids, and bioflavonoids. Additionally, polyphenols can form associations with various carbohydrates and organic acids, contributing to their solubility, bioactivity, and interaction with other biomolecules [41].
Natural polyphenols function as secondary metabolites, enabling plants to respond to environmental stressors. These include ultraviolet (UV) radiation, pathogen attacks, nutrient-deficient soils, temperature extremes, drought conditions and herbivory [39,42,43]. As noted by Sies, polyphenol-rich plants and spices, especially those containing flavonoids, have been used for millennia in traditional Eastern medicine, although they are less common in Western therapeutic practices [44].
The extensive scientific literature now demonstrates that dietary polyphenols may confer health benefits across multiple biological systems. Their activities have been linked to protective effects against cancer, neurodegenerative diseases, cardiovascular dysfunctions, metabolic syndrome, diabetes, aging, and chronic inflammation [44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63].

2.2. Focus on Stilbenes: Structure, Diversity, Biosynthesis, and Natural Functions

Stilbenes are a class of polyphenols formed through the condensation of three C2 carbon residues with an activated hydroxycinnamic acid, a mechanism similar to that observed in flavonoid biosynthesis. Structurally, they consist of two aromatic rings linked by an ethylene bridge, forming the core diphenylethylene structure (C6-C2-C6) [64,65]. These compounds exist in both trans and cis isomeric forms, with the trans configuration being more stable and commonly found in nature [64,65,66].
According to the study by Goufo et al., stilbenes can be categorized based on polymerization degree, including monomers, dimers, trimers, tetramers, pentamers, and hexamers [35]. The most widely studied stilbenes such as resveratrol, pterostilbene, and piceatannol exhibit a range of biological properties including antioxidant, anti-inflammatory, and anticancer activities [67,68,69]. Pterostilbene additionally shows analgesic properties, while resveratrol has demonstrated protective effects against atherosclerosis [67,68].
In grapevine, stilbene production is notably induced by mechanical damage, such as pruning of fresh grapevine canes, which stimulates the biosynthesis of compounds like resveratrol and piceatannol [70]. In a study by Guerrero et al., ε-viniferin was identified as the predominant stilbene (26–52%) in one-year-old grapevine canes, followed by other compounds such as trans-resveratrol, piceatannol, pinosylvin, rhapontigenin, pterostilbene, and isorhapontigenin, whose levels varied over time (Figure 1) [71]. These molecules vary structurally, and this variation determines their chemical behavior and bioactivity. For instance, hydroxylated and glycosylated stilbenes have improved water solubility, enhancing incorporation into hydrophilic systems. Methoxylated stilbenes, by contrast, are more lipophilic and bioavailable. Glycosylated forms tend to be less stable and more prone to isomerization than their aglycone counterparts. Ortho-hydroxylated stilbenes, due to the ability to form stable semiquinone radicals, display greater antioxidant, anti-inflammatory, and anticancer effects compared to meta-hydroxylated isomers [72].
Stilbenes occupy a dual role within the plant kingdom, acting both as an ever-present line of defense and as rapid responders to environmental threat. Their involvement in shielding plants from infectious agents or harsh climatic conditions illustrates the evolutionary value of chemical versatility [71,73,74,75]. Not uniquely confined to grapevines, these compounds are distributed across a wide botanical landscape, from staple crops to regional medicinal herbs. The notable presence of pterostilbene in berries and residues underlines the untapped reservoir of such molecules in both dietary and agricultural contexts. This cross-species distribution implicates stilbenes as a convergent solution to common biological challenges but also complicates efforts to establish source-specific therapeutic claims [71,76].
The biosynthesis of stilbenes is catalyzed by stilbene synthase (STS), an enzyme that evolved from chalcone synthases (CHS) through convergent evolution. This enzyme family displays tissue-specific and development-specific expression patterns. For example, STS gene expression is lower in young grapevine leaves but increases significantly in mature leaves and grape skins during ripening especially in varieties such as Cabernet Sauvignon and Norton peaking at harvest time (Figure 2) [73,77].
Grapevine byproducts like pomace and winemaking residues are rich in polyphenols, including stilbenes [78]. In viticultural waste, total stilbene concentrations range from 2400 to 5800 mg/kg dry weight (DW) [71]. Cebrián et al. observed the accumulation of phenolic compounds in grapevine buds after 1, 3, and 6 months of post-pruning storage in two Vitis vinifera varieties [79]. In grapevine canes, trans-resveratrol levels range from 441 to 7532 mg/kg DW, while trans-ε-viniferin concentrations vary from 1218 to 5341 mg/kg DW, depending on cultivar, vintage, and storage conditions.
Dormancy in grapevine, including endodormancy and ecodormancy, which occur during cold climates, significantly influences the biochemical composition of canes [80]. Hybrid cultivars adapted to northern latitudes, such as those grown in Estonia, have been found to accumulate high levels of dietary stilbenes like resveratrol and viniferin [81,82].
Grapevine canes, rich in bioactive stilbenes, demonstrate notable antifungal and antioxidant properties, making them valuable for nutraceutical applications [71]. Bud extracts contain up to 29% stilbenes and their incorporation in winemaking presents an eco-friendly strategy to reduce sulfur dioxide (SO2) use, particularly in white wines [83]. Toasted grapevine canes also enhance the antioxidant properties of wine by contributing high levels of prodelphinidins and stilbenes [84].
Furthermore, grapevine leaves and cane extracts are a source of bioactive antioxidant molecules and other valuable metabolites for pharmaceutical, nutraceutical, and food applications [81,85]. The abundance of raw materials from viticultural waste provides a compelling opportunity for industrial valorization, given the commercial and pharmacological potential of stilbenes [86,87].

2.3. Extraction Methods of Polyphenols and Stilbenes

The pursuit of efficient polyphenol extraction continues to evolve, reflecting the chemical diversity and sensitivity of these compounds. While early protocols relied on harsh conditions and extended processing times, contemporary approaches are trending toward gentler and rapid techniques, such as microwave-assisted extraction (MAE), ultrasound-assisted extraction (UAE), and ultrahigh-pressure extraction, which preserve and selectively recover targets from complex matrices. A critical, though often underestimated, factor in yield and purity is the nature of the solvent system that balances polarity to suit the solubility profile of the molecules of interest. Complexities such as acid–base stability and matrix interference persist, challenging researchers to continually fine-tune protocols for industrial and pharmacological scalability [88].
Conventional methods for extracting stilbenes particularly trans-resveratrol and trans-ε-viniferin from grape canes primarily rely on conventional solid–liquid extraction (CSLE). This involves maceration in a solvent, with or without stirring, under atmospheric pressure and temperatures ranging from ambient to reflux conditions. Extraction efficiency is influenced by multiple factors, including the solvent type, solvent-to-solid ratio, temperature, extraction time, pH, and light exposure. Hydroalcoholic mixtures such as ethanol/water are commonly used due to their efficacy and non-toxicity [89]. Angel et al. showed that ethanol/water mixtures outperform other solvents in yielding stilbenes [90,91]. An increase in the solvent-to-solid ratio generally improves yield, with optimal ratios typically between 5 and 50 mL/g. Higher temperatures also enhance extraction, with optimal results achieved between 20 °C and 80 °C without degrading the compounds. Prolonged extraction times may increase concentration but can also cause compound degradation. Additionally, alkaline conditions and UV light exposure negatively affect stilbene stability.
Emerging techniques such as UAE, MAE, and pressurized solvent extraction (PSE) offer several advantages over conventional methods. UAE employs acoustic cavitation generated by ultrasonic waves to enhance mass transfer, disrupt plant tissues, and increase surface area, thus improving yields and reducing extraction time and energy use [92]. Despite its benefits, UAE requires careful optimization of parameters like frequency, power, and temperature. MAE, on the other hand, uses microwave energy to heat solvents, promoting efficient cell disruption and reducing extraction times. This method enables uniform heating, rapid energy transfer, and lower solvent consumption. Compared to conventional heating, MAE reduces energy waste and enhances extraction of bioactive molecules in an environmentally friendly manner [92,93,94,95,96]. Optimized MAE conditions have been shown to increase stilbene recovery, though comparisons across studies are often difficult due to variability in extraction parameters [33,89]. PSE employs pressures between 10 and 15 MPa to maintain solvents in a liquid state above their boiling points, improving solubility and mass transfer. For stilbenes, ethanol/water (25:75, v/v) at a 30 mL/g ratio, 105 °C, 1 mL/minute flow rate, and 5.2 MPa pressure has demonstrated higher diffusivity and shorter extraction times compared to conventional methods. One study reduced extraction time from 8 h (Soxhlet) to 10 min using PSE, with slightly increased yields [89].
A key trend in extraction science is the shift towards processes that minimize time and energy consumption without compromising yield. As demonstrated by recent comparative studies, techniques leveraging microwave or pressure are especially effective, and may even obviate the need for added solvents when the inherent moisture of the sample is sufficient. Despite universal gains in speed and ecological footprint, not all advanced workflows are equally user-friendly; fine-tuning parameters, particularly for ultrasound-based approaches, remain a nuanced, trial-and-error exercise. This reality tempers the initial enthusiasm for high-tech solutions with the need for context-sensitive optimization in practical settings [36].
Extraction yields are affected by grapevine variety and environmental factors such as UV exposure, soil type, and vineyard practices like leaf removal or fertilization. Even within the same variety, yields may vary due to biological and climatic influences. Post-pruning parameters such as storage duration, cane provenance, and physiological stage also play a significant role in extraction optimization [89].
Trans-resveratrol and trans-ε-viniferin are the dominant stilbenes in grape canes, but other oligomeric stilbenes such as hopeaphenol, ampelopsins, vitisins, pterostilbene, piceatannol, and polydatin are also present and bioactive [97,98,99]. Nonetheless, stilbenes account for only 3.1% of the dry mass of grape canes, meaning over 95% of the biomass remains underutilized after extraction [89].

3. Biological Activities of Stilbenes and Health Implications

3.1. Bioavailability and Metabolism of Stilbenes

Despite their reputation for limited absorption, some stilbenes and their derivatives have been documented to attain concentrations in vivo sufficient for biological relevance. The extent to which this translates into tangible physiological effects, however, is still fiercely debated, and often contingent on the nuances of both compound structure and formulation [100]. However, the most researched stilbene, resveratrol, has a short half-life because to its quick metabolism (14 min), limited oral bioavailability (20–30%), and low water solubility (<0.05 mg/mL) [101,102]. Oral absorption in humans is around 75%, but extensive intestinal and hepatic first-pass metabolism (mainly glucuronidation and sulfation) reduces systemic availability [103]. Stilbenes’ complexation with cyclodextrins improves the phenolic compounds’ solubility but not their bioavailability within target tissues [104]. However, other formulation strategies such as incorporation into bile acid complexes, liposomes, nanoparticles, or co-administration with absorption enhancers like piperine can significantly increase plasma concentrations and tissue delivery [105,106,107,108,109,110].
Pterostilbene exhibits superior oral bioavailability (~80%), attributed to its increased lipophilicity (due to two methoxy groups) and higher metabolic stability (one free hydroxyl), resulting in slower glucuronidation and a longer half-life compared to resveratrol (1.73 vs. 1.48 h) [111,112,113,114,115,116]. Gnetol possesses lower bioavailability (~6.6%) but a longer half-life (4.2 h) and sustains serum levels of its glucuronide metabolite, likely supporting pharmacological activity [117,118]. In contrast, ε-viniferin and other stilbenes (piceatannol, pinostilbene) display very low oral bioavailability (<1%, or 0.8% for ε-viniferin), attributed to poor absorption and/or extensive metabolism [119].
Stilbenes are mainly metabolized via phase II reactions (glucuronidation, sulfation) at phenolic groups [120,121,122,123,124,125,126,127]. In human plasma, phase II conjugates predominate (resveratrol-3-sulfate being the most abundant), whereas in animal models, glucuronide forms are often higher. Some metabolites, especially conjugated forms, may serve as reservoirs, being deconjugated to release active stilbene, and enterohepatic recirculation has been observed. Dihydro-resveratrol, formed by intestinal hydrogenation, retains relevant biological activity, particularly in the colon [128,129].
Tissue distribution studies show that pterostilbene and its sulfate accumulate in tissues, particularly in the central nervous system, unlike resveratrol, whose plasma and tissue levels decrease quickly. Piceatannol and pinostilbene are efficiently glucuronidated and show high hepatic concentrations but low systemic exposure and rapid clearance [130,131,132,133,134]. Piceatannol can also serve as a metabolic product of resveratrol via CYP1B1, suggesting a prodrug relationship [135].
Nanotechnological strategies including PEGylated liposomes and lipid nanocapsules can significantly enhance brain delivery and systemic concentrations of stilbenes, addressing issues of chemical instability and photosensitivity [78,105] and novel soluble galenic formulations also markedly increase resveratrol’s bioavailability and biological effects. In clinical settings, stilbenes are generally well tolerated even at high doses; adverse effects are mild and mostly gastrointestinal, and doses above 1 g/day are avoided due to tolerability limits [136,137,138,139,140].
Metabolite profiles are complex, including a wide array of glucuronide and sulfate forms, as well as hydrogenated and methylated derivatives [120,121,122,123,124,125,126,127,133]. For some stilbenes, tissue- or adipose-localized conjugates may be reconverted in situ, maintaining local activity. Excretion occurs mainly via feces rather than urine, as seen for ε-viniferin. Detection of certain stilbenes (e.g., ε-viniferin) in rat brain tissue indicates the ability to cross the blood–brain barrier, suggesting potential for central nervous system effects [141,142].
In summary, stilbenes demonstrate varied pharmacokinetic profiles, mainly limited by low bioavailability and rapid metabolism. Pterostilbene stands out for its higher oral bioavailability, tissue accumulation, and longer half-life, whereas other stilbenes may benefit from novel delivery systems to enhance clinical efficacy and expand therapeutic potential.

3.2. Antioxidant and Anti-Inflammatory Properties

The preventive effects of stilbenes on various diseases are largely attributed to their antioxidant activities, including anti-cyclooxygenase action and modulation of lipid and lipoprotein metabolism (Figure 3) [77]. Stilbenes counter oxidative stress by inhibiting reactive oxygen and nitrogen species (ROS and RNS), a property influenced by hydroxyl group position and number. Ortho-dihydroxylated compounds, like piceatannol, are more potent due to enhanced stabilization of semiquinone radicals, particularly at the R4’s position of resveratrol. Their antioxidant activity is also modulated by pH and protonation states. While direct radical scavenging is limited in vivo, stilbenes regulate antioxidant enzymes such as catalase, superoxide dimustase (SOD), nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, glutathione peroxidase (GPx), glutathione S-transferase (GST), and NQO. For instance, resveratrol upregulates catalase, GPx1, SOD1, and SOD3, while suppressing NOX2 and NOX4 in ApoE-KO mice [82].
Stilbenes also exert strong anti-inflammatory effects. They inhibit cyclooxygenase (COX) enzymes and nitric oxide synthases, reduce cytokine production, and suppress nuclear factor κB (NF-κB) signaling. Stilbenes such as piceatannol, pterostilbene, pinosylvin, desoxyrhapontigenin, and rhapontigenin inhibit inducible nitric oxide synthase (iNOS), reduce prostaglandin synthesis, and COX-2. Oxyresveratrol and piceatannol also inhibit nitrite production and iNOS, and resveratrol, oxyresveratrol, and piceatannol block NF-κB activation. Pterostilbene inhibits nuclear translocation of NF-κB and reduces pro-inflammatory cytokine production. Piceatannol is more effective than resveratrol in inhibiting COX-2, NF-κB activation, and cytokine production, while pinosylvin inhibits COX-2 with nearly twice the potency of resveratrol. Rhapontigenin is less active than desoxyrhapontigenin due to a missing hydroxyl at R3’ [82].
Piceatannol may contribute to anti-inflammatory responses by activating pathways involving heme oxygenase-1 (HO-1). In human endothelial cells, it upregulates HO-1 expression by activating the nuclear factor erythroid 2-related factor 2 (Nrf2). Inhibition of HO-1 negates piceatannol’s effects, restoring the expression of tumor necrosis factor-α (TNF-α), interleukins-6 (IL-6), and NF-κB and p65 phosphorylation [143]. Piceatannol has also shown the ability to increase HO-1 expression in macrophages, endothelial cells, mammary epithelial cells, rat liver, and neuronal cells [144,145,146,147,148,149]. However, it is worth noting that HO-1’s role in metabolic disorders is not well understood and, in some cases, has shown pro-inflammatory effects [150]. Thus, it is possible that piceatannol’s anti-inflammatory effects are mediated through the regulation of the upstream activator of HO-1, Nrf2 [147].
Several studies have demonstrated piceatannol’s ability to inhibit inflammatory pathways. Free fatty acids (FFAs) promote inflammation, with palmitic acid having pro-inflammatory properties. Piceatannol prevents the inhibitory effects of palmitic acid on insulin receptor substrate 1 (IRS-1) phosphorylation, glucose uptake, eNOS phosphorylation, and nitric oxide production in human endothelial cells [143]. Furthermore, piceatannol reduces lipopolysaccharide (LPS)-induced protein expression of IL-6 and TNF-α, attenuates NF-κB and signal transducer and activator of transcription 3 (STAT3) signaling in macrophages, and prevents IL-6 secretion and STAT3 and STAT5 signaling in human lymphocytes [144,151,152,153].
In addition, resveratrol enhances tetrahydrobiopterin (BH4) synthesis in cell and animal models, showing reduced oxidation [154]. BH4 is reduced by oxidative stress, negatively affecting various physiological functions [155]. Resveratrol can inhibit cyclic adenosine monophosphate (cAMP) phosphodiesterase, thereby increasing cellular levels of cAMP in cell and animal models [156]. Phosphodiesterases (PDEs) are enzymes that degrade cAMP and cyclic guanosine monophosphate (cGMP), which are second messengers involved in regulating numerous genes and cellular functions [157].
Stilbenes, particularly resveratrol, can activate cAMP signaling pathways. For in-stance, resveratrol boosts antioxidant defenses in aging cells, where cAMP induces Nrf2 expression. This pathway is also activated by alpha-melanocyte-stimulating hormone, a hormonal activator of melanocortin receptors coupled to the G-protein–cAMP cascade [158]. Protein kinase A and cAMP response element-binding protein (CREB) mediate Nrf2 activation through cAMP response elements (CREs) biding in their promoters, resulting in Nrf2 and Nrf2-related gene (e.g., glutathione S-transferase pi 1 and NOS) transactivation [159,160,161].
One of the more intriguing avenues in stilbene pharmacology concerns the interplay between resveratrol and energy-sensing molecular machinery. By interfering with phosphodiesterases, resveratrol alters intracellular calcium and thereby activates a cascade that converges on Adenosine monophosphate-activated protein kinase (AMPK), a critical metabolic checkpoint [162]. These events ripple outward, promoting shifts in both NAD+ pools and sirtuin activity, which could, in theory, recalibrate key metabolic regulators implicated in chronic disease. Yet, a challenge persists, i.e., drawing causal links from these signatures in model systems back to consistent outcomes in whole organisms remains elusive [163,164,165].

3.3. Antimicrobial and Antifungal Activities

Stilbenes have demonstrated antibacterial, antiviral, and antifungal activity (Figure 3) [105,166,167,168]. Resveratrol and its analogues act as inhibitors of fungal tyrosinase [106,107]. In addition, stilbenes inhibit fungal cellular respiration and the lipid peroxidation of fungal membranes [136]. The antifungal activity of resveratrol inhibits a broad spectrum of fungi, including Pyricularia oryzae, Plasmopara viticola, Cladosporium cucumerinum, Sphaeropsis sapinea, Phytophthora capsici, Phytophthora colocasiae, Botrytis cinerea, Candida albicans, and Colletotrichum gloeosporioides [137,138,139,169]. Pinosylvin shows the highest efficacy against Gram-negative bacteria, followed by resveratrol, piceatannol, oxyresveratrol, and pterostilbene. Against Gram-positive bacteria, pinosylvin and pterostilbene are more effective than resveratrol. Pterostilbene’s hydrophobicity and methylation improve membrane penetration and antifungal potency up to five times higher than resveratrol in inhibiting conidial germination and in vitro mycelial growth [72].
The first mechanism for antimicrobial activities involves their ability to inhibit conventional targets, such as the cell’s membrane and wall, cell division, DNA, mitochondria, the calmodulin–calcineurin pathway, and the phosphoenolpyruvate (PEP)-dependent phosphotransferase system. The second mechanism relates to their ability to act as antibiofilm and antivirulence agents. Thus, stilbenes inhibit biofilm formation and assist in the eradication of already formed biofilms. Furthermore, another mechanism of stilbenes is their ability to reverse drug resistance through inhibition of alternative targets, target-modifying enzymes, and antibiotic-modifying enzymes [170]. Stilbenes have an antimicrobial mechanism that involves disrupting bacterial membranes, resulting in the leakage of cellular contents and leading to cell death. Pterostilbene, resveratrol, and toremifene (an anticancer agent approved by the Food and Drug Administration (FDA) and synthesized in 1981) have demonstrated antimicrobial activity through cell membrane disruption [171,172,173]. The membrane-associated protein phosphatidylglycerophosphate synthase (PgsA) catalyzes the substitution of cytidine monophosphate with glycerol phosphate to produce phosphatidylglycerol phosphate (PG-P). Subsequently, PG-P is dephosphorylated by PgpP to yield phosphatidylglycerol (PG), which is one of the main components of the cell membrane [174].
Another mechanism involves the action on the bacterial cell wall, which consists of a complex peptidoglycan polymer net composed of N-acetylmuramic acid and N-acetylglucosamine, with an attached pentapeptide. The biosynthesis of bacterial peptidoglycan is catalyzed by sequential Muramyl ligases in the intracellular steps. Stilbenes have the ability to inhibit MurD, an enzyme that catalyzes the reaction from UDP- N-acetylmuramic-Ala to UDP- N-acetylglucosamine-dipeptide [175].
Wall teichoic acids are anionic glycopolymers anchored in the cell walls of Gram-positive bacteria that play critical roles in bacterial physiology. Wall teichoic acids’ biosynthesis initiates with a reaction is catalyzed by the membrane-associated glycosyltransferase TagA [111]. Stilbenes have shown promising activity against Gram-positive bacteria by targeting cell wall teichoic acids [111].
The fungal cell wall is composed of complex components, mainly polysaccharides (chitins, glucans, and mannans), lipids, and proteins. Stilbenes inhibit the activity of chitin synthases [176]. NADPH–cytochrome P450 reductase chemokine receptor (CCR1) is involved in the yeast cell wall. Stilbenes have the ability to inhibit the activity of NADPH–cytochrome P450 reductase CCR1, leading to defects in yeast cell wall integrity [177].
Stilbenes have shown highly promising results in reducing the mycelial growth of phytopathogenic fungi such as Pythium aphanidermatum, Rhizoctonia solani Kuhn, Exserohilum turcicum, and Fusarium oxysporum, as well as the yeast Saccharomyces cerevisiae, in both agar medium and broth microdilution assays, using chlorothalonil as a positive control [178]. A series of resveratrol oligomers were purified from the canes of Vitis vinifera. These compounds were tested against Candida albicans [179,180]. Pterostilbene was shown to be a more potent inhibitor compared to 5-fluorocytosine, a reference antifungal agent used against Candida albicans. Moreover, pterostilbene proved to be a promising inhibitor against 12 non-albicans Candida (NAC) species, including emerging pathogens such as Candida guilliermondii and Candida famata. These findings support the idea that naturally occurring stilbene-type phytoalexins, produced by plants and certain bacterial species, serve as defense mechanisms against pathogens and represent a rich source for the discovery of novel fungicides [181].
Stilbenes can inhibit DNA synthesis. For example, trans-dihydroresveratrol dimer has been shown to block the ATP-binding site of DNA gyrase, inhibiting, therefore, the enzyme’s activity and consequent DNA synthesis [182]. Another mechanism is causing DNA damage. For instance, oxyresveratrol binds directly to DNA, inducing the molecules’ cleavage and subsequent mitochondria-mediated apoptosis in Candida albicans [183]. Resveratrol’s prooxidant activity induces DNA damage through an increase in ROS, malondialdehyde accumulation, and glutathione depletion in Salmonella typhimurium [184].
Beyond their central role in cellular energetics, mitochondria stand at the crossroads of survival and programmed cell death. Loss of membrane integrity precipitates a sequence that leads inevitably to apoptosis [185]. Evidence implicates resveratrol as a potent modulator of this process, which is able to destabilize mitochondrial function and tip the scales towards cell death, as seen in both pathogen and tumor models. This mechanistic insight, while compelling, raises further questions about tissue selectivity and the balance between therapeutic efficacy and toxicity [186].
The cell membrane and wall, DNA, and mitochondria are common targets in antimicrobial therapies and, therefore, stilbenes’ effects on the mentioned cellular structures are of interest when designing new treatment strategies for pathologies caused by microorganisms. For example, resveratrol and piceatannol perform a reversible inhibition in ATPase activity and consequent ATP synthesis in Escherichia coli [187]. Moreover, resveratrol’s antibacterial activity extends to the downregulation of FtsZ expression and inhibition of Z-ring formation, a dynamic structure essential to cell division in prokaryotic organisms since it recruits division-related proteins and directs septal peptidoglycan synthesis [188,189].
Conventional antibiotics are becoming less effective overtime due to an increase in the number of multidrug-resistant microorganisms. Virulence and its associated factors determine pathogenicity and allow pathogens to acquire desired characteristics, such as immune evasion or modulation, colonization, and tissue damage. Toxins are also implicated in pathogenesis and include superantigens, surface proteins, hemolysins, and leukocidins. The pathological mechanism of α-hemolysin (Hla) secreted by Staphylococcus aureus, for example, relies on the direct binding to erythrocytes’ cell membrane and subsequent pore formation and lysis. Thus, resveratrol and trans-stilbene have the ability to inhibit hemolysis caused by Staphylococcus aureus through the repression of the hla gene expression, thereby reducing the microorganism’s virulence [140]. Furthermore, the two-component system SaeRS exerts essential functions in virulence factors’ production [108]. Resveratrol can target the mentioned system, resulting in the downregulation of saeRS and consequent reduction in α-hemolysin secretion [109].
It is estimated that between 40% and 80% of the existing bacteria and archaea reside in biofilms, a macrocolony of microorganisms attached to a surface that constitutes a contributing factor in the development of chronic infections [110,112,113]. Biofilms offer microorganisms an advantage under various environmental challenges [114].
Biofilm formation occurs in five progressive stages, which include initial reversible attachment, irreversible attachment, first layer formation, mushroom- or tower-shaped structure formation, and dispersion and reattachment [112]. Stilbenes are capable of disturbing the hallmarks in biofilm formation, arresting their maturation, and allowing for the elimination of the mentioned structure. For example, resveratrol inhibits the production of flagellin and, consequently, reduces the motility of Proteus mirabilis, thereby preventing swarmer cell invasion into human uroepithelial cells [115]. Resveratrol also impairs the motility and adhesion of Salmonella typhimurium to HeLa cells through downregulation of flagellar genes [190]. Furthermore, resveratrol suppresses fimA and xadA expression, resulting in low levels of afimbrial adhesin and impaired adhesion capacity of Xylella fastidiosa to host cell surfaces [191]. The downregulation of fimbriae-associated gene expression by resveratrol in Porphyromonas gingivalis blocks biofilm production and increases in a directly proportional manner to the stilbene’s concentration [192]. The other stilbene that is able to impair biofilm production and disrupt mature biofilms is pterostilbene, which suppresses filamentation-related gene expression through the Ras/cAMP pathway [180].
Biofilms release planktonic cells or small clusters of pathogens through dispersion in a continuous manner [193]. Stilbenes interfere with biofilm propagation through targeting the enzymes involved, such as micrococcal nuclease, whose production is reduced following resveratrol administration, resulting in more efficient Staphylococcus aureus biofilm clearance [194].

3.4. Anticancer Effects: Preclinical and Clinical Evidence

In a recent epidemiological study, it was estimated that approximately 1,280,000 deaths in the European Union and 618 120 deaths in the United States were caused by cancer in 2025 [120,195]. Cancer is the second leading cause of death since the beginning of the 21st century, preceded by cardiovascular pathologies and followed by diabetes and chronic respiratory diseases [196]. The increase in patient care, therapy efficiency, and awareness of the impact of healthier lifestyles in cardiovascular diseases has caused a drop in its mortality rate, falling behind that of cancer [121,122,197].
Increasing attention has been given to chemoprevention as an alternative approach to cancer control. There is growing evidence that oxidative stress induced by ROS is linked to carcinogenesis at multiple stages [123]. ROS are abundant free radicals in cells and are associated with various degrees of tissue damage. Oxidative stress arises from an imbalance between ROS production and neutralization or elimination by the organisms’ naturally occurring antioxidant machinery. Consequently, oxidative stress can cause multiple issues, such as damage to proteins, lipids, and DNA, triggering or modulating the initiation, promotion, and progression of cancer [124].
Resveratrol has demonstrated potential anticancer activity, first reported by Jang et al., and has sparked significant interest in its chemopreventive and chemotherapeutic properties (Figure 3) [125]. Resveratrol has been tested in both in vitro and in vivo carcinogenesis assays for various cancer types, including leukemia, prostate, breast, lung, colon, ovary, liver, oral cavity, thyroid, and non-melanoma and melanoma skin cancer (Table 1) [126,198,199,200,201,202,203,204,205,206]. The chemopreventive properties of resveratrol rely on the compound’s antioxidant activity. All stages of carcinogenesis are affected by the stilbene’s anticancer activity, with the inhibition of COX-2 representing the major hallmark [125].
To date, three different isoforms of COX have been described, i.e., COX-1, which is expressed in normal tissues and participates in tissue homeostasis, COX-2, which results from overexpression in cases of inflammation or the development of neoplasms, and COX-3, a variant of COX-1 [207]. Many studies show that COX-2 plays a significant role in tumor progression, as its levels are elevated in premalignant and malignant tissues and are associated with a reduced survival rate in cancer patients, making it an unfavorable prognostic factor [116,208,209,210]. Clinical trials suggest that COX-2 inhibitors may be a solution for preventing the development adenomas and, potentially, carcinomas in the colon [207]. However, the clinical efficacy of these inhibitors is questionable due to increased cardiovascular risks [130].
Thus, resveratrol, which has shown positive effects in delaying cancer progression due to its significant potential in inhibiting COX-2, does not demonstrate toxicity effects [127]. Prostaglandins, produced through COX activity, have also been linked to cancer development and progression [116,211].
The increased production of prostaglandins influences the metabolism of carcinogens, tumor cell proliferation, and metastatic potential [131,212]. Thus, the inhibition of prostaglandin synthesis has been proposed as a strategy to prevent tumor development [131,132,211]. Numerous studies confirm that resveratrol has the capacity to inhibit COX-2, thereby reducing the synthesis of prostaglandin E2 (PGE2). Cianciulli et al. reported that resveratrol negatively regulates COX-2 and PGE2 in human intestinal Caco-2 cells treated with LPS, likely related to the inhibition of the NF-κB transcription factor [133]. NF-κB is associated with inflammatory, immune, and oncogenic responses [134].
Resveratrol activates AMPK, effectively preventing tumorigenesis [133,204,213,214]. Studies on redox status and antioxidant mechanisms’ function indicate that resveratrol acts as a potent chemoprotector in in vivo cancer models. In rats treated with the hepatotoxic carcinogen azoxymethane (AOM), which induces oxidative imbalance, resveratrol partially reversed effects such as lipid peroxidation, glutathione (GSH) depletion, and increased NO levels in the liver [215]. Additionally, resveratrol induces the expression of SOD and catalase through a mechanism involving the tumor suppressor gene phosphatase and TENsin homolog (PTEN) and protein kinase B (PKB) signaling pathways. PTEN-mediated inhibition of the phosphoinositide 3-kinase (PI3K)/PKB pathway leads to increased activity of SOD, GSH peroxidase, and catalase [216].
Studies showed that resveratrol induces apoptosis in several cancer types. This pro-apoptotic stimulation is related to changes in the cell cycle, caspase activation, and downregulation of XIAP, Survivin, Bcl-2, and Bcl-xL, and upregulation of Bax, Bak, Bim, Noxa, PUMA, p21, and TRAIL-R2/DR5 [143,144,145,217,218,219,220]. These effects are also correlated with p53 activation [143,144,218,220]. For instance, resveratrol and piceatannol increase cytoplasmic calcium concentrations in human breast cancer cells (MDA-MB-231), activating p53 and triggering pro-apoptotic gene transitions [145]. In prostate cancer cells with p53 mutations (Du145), resveratrol induces p53 phosphorylation, restoring wild-type-like DNA binding and promoting pro-apoptotic events [146,147,148].
Pterostilbene, a natural analog of resveratrol with greater bioavailability [101,146], shares significant similarity with resveratrol but exhibits stronger in vitro antioxidant activity and demonstrates substantial clinical potential across various diseases [149,150,221]. Pterostilbene shows chemopreventive properties against cancer in different in vitro and in vivo assays (Table 1). Studies have demonstrated that pterostilbene, which can act as an active apoptotic agent, also inhibits growth, adhesion, and metastasis development [118,152,153,154]. These effects have been observed in several types of cancer, including breast, lung, stomach, prostate, pancreatic, melanoma, and/or colon cancers, highlighting its role as a chemopreventive agent [152,155,156,157,158,159,217,221,222].
Rimando et al. analyzed the antioxidant activity of pterostilbene, finding that it inhibits preneoplastic lesion formation caused by carcinogenic agents in a rat mammary organ culture model [149]. Moreover, Chiou et al. demonstrated that pterostilbene is more potent than resveratrol in preventing AOM-induced colon tumorigenesis by activating the Nrf2-mediated antioxidant pathway [160].
In a similar experimental model, pterostilbene has shown to regulate the expression of inflammation-related genes such as COX-2 and iNOS [161,223]. Likewise, in immortalized human keratinocytes (HaCaT), pterostilbene enhances Nrf2 nuclear translocation and the expression of Nrf2-dependent oxidative stress-associated molecules, reinforcing the central role of Nrf2 in the chemoprevention promoted by pterostilbene [127]. In HT-29 colon cancer cells, pterostilbene inhibited cytokine induction through the p38 transcription factor pathway, as well as other anti-inflammatory pathways such as ERK, JAK-STAT, NF-κB, c-Jun NH2-terminal kinase, and PI3K [161]. This inhibition is attributed to the reduced expression of COX-2 and iNOS, suggesting that the MAPK cascade activation by p38 is an essential pathway for the anti-inflammatory action of pterostilbene [161].
Furthermore, pterostilbene is equally potent to resveratrol in inhibiting NF-κB, COX-2, iNOS, and activator protein 1 (AP-1) as demonstrated in a skin carcinogenesis model induced by tissue-type plasminogen activator (TPA) in mice [224]. Pterostilbene induces PTEN expression in prostate cancer, leading to decreased levels of miR-106b, miR-17, and miR-20a. The effect of pterostilbene on restoring PTEN mRNA and protein levels to normal values was greater than that of resveratrol, suggesting that pterostilbene exhibits greater in vivo activity due to the methoxy groups occupying the place of hydroxyl groups [225].
In a study involving a mice model of UVB-induced skin carcinogenesis, pterostilbene proved superior to resveratrol in skin damage of acute and chronic nature [127]. This study demonstrated that pterostilbene has anticarcinogenic effects, maintaining skin antioxidant defenses (e.g., GSH peroxidase, catalase, and superoxide dismutase activities and GSH levels) and reducing oxidative damage to DNA, proteins, and lipids induced by UVB radiation [127].
Several studies validate pterostilbene as an efficient anticancer therapeutical agent acting on multiple signaling pathways. In an rat model of AOM-induced colon carcinogenesis, a pterostilbene-enriched diet resulted in reduced aberrant crypt foci, decreased transcriptional activation of COX-2 and iNOS, inhibition of glycogen synthase kinase-3β (GSK-3β) phosphorylation, suppression of the Wnt/β-catenin signaling pathway, and downregulation of cyclin D1, vascular endothelial growth factor (VEGF), and matrix metalloproteinases (MMPs), and activation of Ras, PI3K/PKB, and epidermal growth factor receptor (EGFR) pathways [160]. Additionally, pterostilbene reduces pro-inflammatory cytokines such as IL-1β, IL-4, and TNF-α, as well as nuclear phospho-p65 levels [226].
McComarck et al. demonstrated that pterostilbene inhibits breast cancer cell proliferation stimulated by leptin through reduced JAK/STAT3 signaling [227]. In pancreatic cancer cells treated with pterostilbene, there was an upregulation of pro-apoptotic genes, changes in phosphorylated STAT3 levels, increased antioxidant activity by manganese superoxide dismutase (MnSOD), and enhanced expression of Smac/DIABLO and cytochrome c [152]. Additionally, Liu et al. proposed that pterostilbene inhibits JAK2/STAT3 signaling, reducing the expression of STAT3 target genes, including anti-apoptotic proteins Mcl-1 and Bcl-xL, while increasing proteins related to mitochondrial apoptosis (Bak, Bax, cleaved caspase-3, and cytosolic cytochrome c) and cyclin-dependent kinase inhibitors (e.g., p21 and p27) in osteosarcoma cells [228].
Pterostilbene acts as a chemopreventive agent, with effects extending beyond its anti-inflammatory and antioxidant properties or apoptosis-promoting effects. Studies have proposed that pterostilbene can induce cell death through autophagy [156,158,159,229,230]. Recently, it was observed that pterostilbene induces tumor autophagy through a mechanism dependent on lysosomal membrane permeabilization mediated by Hsp70 [217].
In recent years, the traditional model of cancer progression has been revised to emphasize the importance of tumoral heterogeneity in the development of chemo/radiotherapy resistance and post-treatment relapse. Cancer stem cells (CSCs) are attractive targets due to their self-renewal capacity, ability to generate heterogeneous tumor cell lineages, and high tumorigenicity [231,232,233]. Studies suggest that resveratrol and pterostilbene promote Argonaute-2 expression and activity, a central RNAi component, inhibiting cancer stem cell-like characteristics in breast cancer by increasing tumor-suppressor miRNAs such as miR-200c, miR-141, and miR-16 [234]. Pterostilbene suppresses CSC generation and metastatic potential in various experimental models, modulating epithelial–mesenchymal transition pathways and preventing the enrichment of CD133(+) CSCs in irradiated hepatoma [235,236].
Piceatannol, a hydroxylated analog of resveratrol, exhibits health benefits like resveratrol, though fewer studies are available (Table 1) [237,238,239]. Li et al. found that the anticancer properties of piceatannol may arise from its prooxidant effects due to the presence of copper (Cu)(II), which induces hydroxyl radical formation via Fenton and Haber-–Weiss reactions, leading to DNA damage [240,241]. Piceatannol inhibits hydrogen peroxide (H2O2)-induced NF-κB activation, ceramide, LPS, phorbol 12-myristate 13-acetate, and okadaic acid [242]. Additionally, it inhibits p65 phosphorylation and its nuclear translocation, and TNF-induced Iκ-Bα phosphorylation. Moreover, the hydroxyl groups at the 3′ and 4′ positions appear to play a crucial role in stilbene’s anticancer activity, as the same effects were observed for treatment with resveratrol treatment [242].
Piceatannol could reduce iNOS expression, thereby decreasing NO production. Furthermore, it inhibits COX-2 expression in RAW 264.7 cells stimulated with LPS and BV2 microglia cells [243,244]. Additionally, piceatannol increases protein levels of HO-1 in MCF10A, a human cell line of the mammary epithelium. The underlying mechanism involves the release of Nrf2 from Kelch-like ECH-associated protein 1 and its translocation to the nucleus, with consequent binding to the antioxidant response element, promoting HO-1 expression [245].
The effects of ε-viniferin on cancer prevention and treatment have been studied in over 20 cell models, including neural, breast, skin, liver, lung, bladder, stomach, colon, and hematological cancers. A published study demonstrated that, in approximately half of the total cases, the determined IC50 values for ε-viniferin were below 60 µM, highlighting the compound’s great potential in reducing the growth of cancer cells. Furthermore, comparative approaches on the anticancer activity of resveratrol and ε-viniferin have indicated that the latter is more active in one-third of the total cases [141].
In certain studies, the mechanisms underlying the cytotoxic and antiproliferative activities of ε-viniferin have been explored, with most being linked to apoptosis. This form of programmed cell death is essential for the development of the organism and tissue homeostasis. Apoptosis can be initiated by two main mechanisms: the intrinsic pathway, mediated by mitochondria, and the extrinsic pathway, mediated by death receptors. Most pro-apoptotic stimuli for the intrinsic pathway are associated with the permeabilization of the outer mitochondrial membrane. This process is regulated by a balance between pro- and anti-apoptotic members of the Bcl-2 family, leading to the release of cytochrome c into the cytosol. This apoptotic factor activates caspases and proteases responsible for the apoptotic phenotype [246].
ε-viniferin can induce apoptotic cell death in various tumor cell lines, including leukemia, myeloma, glioma, malignant melanoma, and hepatocellular carcinoma [247,248,249,250,251]. It induces apoptosis by disrupting normal mitochondrial transmembrane potential and activating pro-apoptotic proteases. Caspase-3 activation has been highlighted in leukemia and hepatocellular carcinoma cells [248,252]. Barjot et al. demonstrated that caspase-8 activation by ε-viniferin mediates a significant portion of this compound’s cytotoxic effect in U266 myeloma cell lines. In C6 glioma cells, activation of caspases-8, -9, and -3 has been observed [247,253].
The antiproliferative effect results in cell cycle arrest. ε-viniferin’s ability to halt the cell cycle has been investigated. Barjot et al. analyzed that treatment with ε-viniferin led to an accumulation of U266 myeloma cells in the G2-M phases and a reduction in cells in the G0/G1 phases [247]. Furthermore, ε-viniferin arrests the cell cycle of melanoma cells in the S phase and reduces the percentage of cells in the G1 phase, increasing regulatory proteins that control cell cycle progression through the S phase, such as cyclin E1 [249].
In addition, ε-viniferin, like other resveratrol oligomers, has been studied for its in vitro anticancer activities (Table 1). The cytotoxic activities vary depending on the tumor cell line and the structure of the resveratrol oligomer studied. Ito et al. investigated 18 oligomers ranging from dimers to octamers and tested them on five human tumor cell lines [254]. Seven of these compounds exhibited cytotoxic effects, primarily oligomers with more than four basic resveratrol units. However, other studies have suggested that the number of resveratrol units does not influence cytotoxic activity [255,256,257].
Although larger molecules are more active, their efficacy may be limited due to low bioavailability in vivo studies. To date, no in vivo trials have been conducted to compare ε-viniferin with resveratrol in studying its potential beneficial effects on the promotion, progression, or treatment of cancer. Several studies on ε-viniferin have not isolated the compound but instead used it in plant extracts or compound mixtures. These studies have shown that such mixtures or combinations are sometimes more active than the isolated molecules due to the synergistic effect between compounds [141].
For example, Vineatrol® has demonstrated more efficient antiproliferative effects than isolated resveratrol or ε-viniferin in leukemia and hepatoma cells [248,258]. Additionally, ε-viniferin has been combined with anticancer drugs such as vincristine or cisplatin. Özdemir et al. demonstrated that ε-viniferin can enhance cell sensitivity to vincristine treatment in HepG2 cells [250]. Furthermore, the same author reported a strong apoptotic effect when combining cisplatin with ε-viniferin. Thus, ε-viniferin could potentially be used as a combined treatment with multiple anticancer drugs to reduce drug resistance and lower the required doses, thereby limiting side effects in living organisms [141].
Nivelle et al. achieved very positive results, as they observed reduced toxicity in normal fibroblasts compared to melanoma cancer cell lines [249]. Moreover, in non-transformed hepatocytes (HH4), higher concentrations of the stilbene dimer were required to induce toxicity compared to hepatocellular carcinoma cell lines such as HepG2 and Hep3B [259].
In conclusion, most studies have shown that ε-viniferin can reduce cancer cell growth, inducing apoptosis, and modifying the cell cycle [141]. Antioxidants act as agents that slow cancer progression by scavenging free radicals, inhibiting or preventing oxidative damage, and reducing oxidative stress [260].
Table 1. Overview of in vitro studies on the antitumor activity of representative stilbenes.
Table 1. Overview of in vitro studies on the antitumor activity of representative stilbenes.
StilbenesExperimental In Vitro ModelMethodologyExperimental
Conditions
Significant ResultsRef
ResveratrolD407 cellsH2O2-induced cytotoxicity or MTT assay0, 25, 50, and 100 µM for 24 hResveratrol offered protection to D407 retinal pigment epithelial cells against H2O2-induced cytotoxicity, resulting in reduced cytotoxicity.[261]
N2a cellsFluorescein diacetate assay1.5, 3.125, 6.25, 12.5, 25, 50, and 100 µM for 48 hPlasma membrane integrity was compromised in resveratrol-treated N2a cells, reducing cell viability.[262]
T24 cellsCell Proliferation Kit II50, 100, 150, 200, and 250 µM for 24 hResveratrol exposure led to a decrease in cell viability, which was greater for s higher concentration of the stilbene.
The determined IC50 was 178.73 µM.
[263]
MCF-7 cellsMTS assay20, 40, 60, 80, and 100 µM for 48 hResveratrol was cytotoxic in MCF-7 cells, aligning with the compound’s anticancer properties. Changes in membrane fluidity and, consequently, in cellular signaling pathways were detected.[264]
HRT cellsMTT assay25, 50, 100, 200, and 300 µM for 72 hResveratrol inhibits HRT cell growth
in a concentration-dependent manner.
[265]
MCF-7 cellsSulforhodamine B (SRB) assay15 μg/mL for 24 hCo-treatment with resveratrol and doxorubicin drastically lowered doxorubicin’s IC50 from 0.417 μg/mL to 0.035 μg/mL, showing it to be a good adjuvant in antitumor therapy.[266]
Trans-resveratrolPC-3 cellsMTS assay0, 3, 10, 30, and 100 μM for 72 hTrans-resveratrol exhibited an inhibitory effect on cell viability in PC3 cell line at concentrations ranging from 10 to 100 μM.[267]
MCF-7 cells JURKAT E.6, and THP-1MTT or XTT assay10, 30, 50, 70, 90, and 100 μM for 24, 48, 72, and 96 hAs trans-resveratrol concentrations increased, cell viability showed a greater or lesser pronounced decrease, depending on the cell line. Therefore, the effect of trans-resveratrol depends not only on dose and treatment duration but also on the cell type considered.[268]
MCF-7, Du145, and PC-3 cellsMTT assay1 × 10−15–1×10−3 M for 48 hTrans-resveratrol exhibited a cytotoxic effect on cancer cells at concentrations ranging from 1 × 10−7 to 1 × 10−4 M.[269]
MCF-7 cellsMTT or neutral red uptake (NRU) assay1, 5, 10, 25, and 50 µM for 24 hSolely an administration of trans-resveratrol did not lead to a decrease in cell viability in MCF-7 cell line. However, a pre-treatment with trans-resveratrol conferred protection against rotenone-induced toxicity.[270]
HepG2, Vero, and MCF-7 cellsMTT assay0, 0.2, 0.4, 2, 4, 6.25, 12.5, 25, 50, and 100 µM for 24, 48, 72 hTrans-resveratrol demonstrated cytotoxicity in all mentioned cell types at concentrations equal to or greater than 50 µM after 48 h.[271]
PC12 cellsMTT or NRU assay0, 5, 10, and 25 µM for 24 hTrans-resveratrol decreases the viability of P12 cells, as shown by both performed assays.[272]
HCT-116, HCT-116/p53(−/−), HepG2, and Hep3B cells CellTiter-Blue® and SRB assay0, 1, 10, and 100 µM for 72 hTrans-resveratrol demonstrated a significant ability to decrease cell viability at a concentration of 100 μM after 72 h.[273]
HepG2 cellsWST-1 assay0.5–100 g/mL for 24, 36, and 48 hTrans-resveratrol decreased the viability of HepG2 cells, evidencing the compound’s anticancer activity.
The calculated IC50 was shown to decreased over time.
[273]
MCF-10A, MCF-7, MDA-MB-231, and ZR-75-1 cellsCellTiter-Glo® Luminescent Cell Viability assay1–350 µM for 48 hTrans-resveratrol was proven to be an efficient inhibitor of the cancer cell lines MCF-7, ZR-75-1, and MDA-MB-231, with IC50 values of 68.3 ± 2.6, 82.2 ± 4.8, and 67.6 ± 4.1 µM, respectively. Moreover, it was three times more potent in the MCF-10A cell line, with an IC50 of 20.0 ± 2.9 µM.[274]
SW480 cellsMTT assay30 µM for 48, 72, and 96 hThere was a time- and dose-dependent decline in cell survival.[275]
MCF-7 cellsAnnexin V-FITC and propidium iodide assay6.25–50 µg/mL for 24 hThe viability of MCF-7 cells was suppressed following trans-resveratrol treatment.[276]
ARPE-19 cells, transmitochondrial normal RPE cybrid, and transmitochondrial AMD RPE cybrid cellsMTT assay1000 µM for 48 hViability assays demonstrated that trans-resveratrol has beneficial properties for cybrid cells, increasing their viability compared to untreated cells.[277]
HepG2 cellsMTT assay2.5, 10, 30, 50, 70, 100, 140, and 200 µM for 24, and 48 hTrans-resveratrol has inhibitory effects on the cell viability of HepG2 cells, having greater impact in higher concentrations and prolonged exposure times.[278]
LTC-14 cellsMTT assay0, 0.1, 1, 10, 100, and 1000 µM for 24, and 48 hLTC-14 cells experienced a decrease in cell viability to below 50% in the presence of trans-resveratrol at a concentration of 200 µM.[279]
NCTC clone 929 cellsNRU assay15.63, 31.25, 62.5, 125, and 250 μM for 24 hTrans-resveratrol caused significant cell injury and death with an associated IC50 of 50 μM.[280]
A549 cellsMTT assay0, 5.5, 11, 21.9, 32.9, 43.8, 87.6, 131.4, and 175.2 μM for 24 hTrans-resveratrol treatment led to cell viability inhibition in a dose-dependent manner.
The IC50 was determined to be 85.5 μM.
[281]
PterostilbeneMCF-7, MDA-MD-231, and ZR-75-1MTT assay10 µmol/L, 20 µmol/L, 30 µmol/L (pterostilbene) + 5 µmol/L (Tamoxifen) for 24, 48, and 72 hCombined therapy with pterostilbene and tamoxifen reduced cell viability in all cell lines. A greater decrease in viability was observed for the 24 h treatment.[282]
MDA-MB-231 and T-47D cellsMTT assay10–100 µM for 48 hA decrease in cell viability and significant morphological changes were observed in both cell lines following the treatment with pterostilbene.
The IC50 concentrations for MDA-MB-231 and T-47D cells were 45.7 ± 0.01 and 63.1 ± 0.11 µM, respectively.
[283]
MCF-7, SK-BR-3, and MDA-MB-468 cellsAmerican Type Culture Collection 0–100 µM for 72 hTreatment with pterostilbene arrested cells growth in a dose-dependent manner for all three cell lines, exhibiting a greater impact in MDA-MB-468 cells.
The calculated IC50 values were 87.6 ± 9.0 µM for MCF-7 cells, 64.4 ± 4.6 µM for SK-BR-3 cells, and 45.7 ± 5.2 µM for MDA-MB-468 cells.
[284]
HeLa, CaSki, and SiHa cellsMTT assay0–200 µM for 72 hAll cell lines’ proliferation was inhibited by pterostilbene in a manner that varied in a directly proportional way to concentration.
The IC50 for each cell line was calculated as follows: IC50 = 32.67 µM for HeLa, IC50 = 14.83 µM for CaSki, and IC50 = 34.17 µM for SiHa, indicating growth-inhibitory effects.
[285]
TC-1 mouse cells after co-transformation with HPV16-E6, HPV16-E7, and c-Ha-Ras oncogenesWST-1 assay0–100 µM for 72 hPterostilbene exhibits significant cytotoxicity, evidenced by the formation of cytoplasmic blebs observed after 48 h.
The number of apoptotic cells increased in a dose-dependent manner and the obtained IC50 of pterostilbene was 15.61 µM.
[286]
CL187, C COLO 205, HCT-8, SW480, Lovo, and HCT-116 cellsCell Counting Kit-8 (CCK-8) assay1–100 µM for 24, 48, and 72 hPterostilbene inhibited cellular activity of all cell lines in a dose-dependent manner.
After 72 h of treatment, the IC50 of pterostilbene for most of the cell lines used (except SW480 cells) was determined to be below 30 µM.
[287]
HT-29 cellsSRB assay5–100 µM for 48 hA significant decrease in cell growth was only observed at concentrations equal to or greater than 10 µM. The extent of inhibitory effects was shown to be dependent on pterostilbene dosage.[288]
HT-29 cellsMTT assay 5 and 20 µM for 24 and 48 hTreatment with pterostilbene at 20 μM inhibited the metabolic activity of HT-29 cells up to 80.2 ± 5.9%.[289]
HEC-1A and ECC-1 cellsMTS assay0, 18.75, 37.5, 75, 150, and 300 µM for 48 hPterostilbene treatment significantly reduced cell viability in a dose-dependent manner, with associated IC50 values ranging between 72 and 78 µM for both cell lines.[290]
Kuramochi, Caov-3, OVCAR-4, OVCAR-8, and SKOV3 cellsMTT assay0, 37.5, 75, 150, and 300 µM for 48 hCell viability was markedly reduced by pterostilbene in a dose-related way, with the IC50 for each cell line as follows: 161.2 μM for Kuramochi, 100.6 μM for Caov-3, 143.8 μM for OVCAR-4, 74.8 μM for OVCAR-8, and 95.2 μM for SKOV3 cells.[291]
LNCaP and PC3 cellsMTT assay0, 20, 40, 60, 80, and 100 µM for 48 hPterostilbene reduced cell viability for both cell lines and in a dose-dependent manner.
The IC50 values ranged between 70–80 μM for LNCaP cells, and 80–100 μM for PC3 cells.
[292]
MIA PaCa-2 and PANC-1 cellsMTT assay10–100 μM for 24, 48, and 72 hPterostilbene inhibited cell viability in a dose- and time-dependent manner in both cell lines.
The IC50 concentration values varied depending on cell type and selected time points, with MIA PaCa-2 showing 72 μM at 24 h, 51 μM at 48 h, and 32 μM at 72 h, while PANC-1 showed 84 μM at 24 h, 33 μM at 48 h, and 29 μM at 72 h.
[293]
A375, A549, HT29, and MCF-7 cellsCountess Automated Cell Counter and SRB Toxicology Assay0–200 μM for 24, 48, and 72 hPterostilbene inhibited cell viability in a dose- and time-dependent manner in both cell lines. The IC50 values determined were cell type-dependent, being much lower for HT29 (IC50 = 60.3 mmol/L) and MCF7 (IC50 = 44.0 mmol/L) cells than for A375 (IC50 = 14.7 mmol/L) and A549 (IC50 = 28.6 mmol/L) cells.[217]
11–18, HCC827, HCC4006, H1975, and PC9 cellsMTT assay0–150 μM for 72 hPterostilbene inhibited cell viability in all cell lines, with IC50 values ranging between 23.8 and 40.7 μM.[294]
HepG2 cellsMTT assay and CCK-8 assay12.5–100 µM for 24 hCell viability and proliferation were reduced for all concentrations considered in a dose-dependent manner.[295]
HT29, MKN74, and CT26 cellsMTS assay10, 50, and 100 µM for 48 hPterostilbene reduces cell viability in all three cell lines. The determined IC50 values were 21 µM for CT26, 63 µM for HT29, and 65 µM for MKN74 cells.[296]
CAR cellsMTT assay5, 10, 25, 50, 75, and 100 µM for 24, 48, and 72 hPterostilbene induces cytotoxicity in a time- and dose-dependent manner.
The IC50 values after 24, 48, and 72 h of incubation were 78.26 ± 4.33, 48.04 ± 3.68, and 20.65 ± 4.88 µM, respectively.
[297]
MDA-MB-231 cellsMTT assay1, 5, 20, 30, and 50 µg/mL for 48 hPterostilbene exhibits an inhibitory associated with an IC50 value of 79.5 ± 6.36 µg/mL.[298]
AsPC-1, BxPC-3, MIA PaCa-2, and PANC-1 cellsMTT assay0, 50, 75, 100, 125, and 150 µM for 48 hIncreasing concentrations of pterostilbene reduced viability in all cell lines tested, pointing towards a dose-dependent sensitivity to the mentioned compound.
The IC50 values ranged from 110 to 130 µM.
[299]
MDA-MB-231 cellsMTT assay2.5, 5, 10, 20, 40, and 80 µM for 24 hA 24 h treatment with pterostilbene at 5 μM resulted in a 12% reduction in survival of MDA-MB-231 cells. The IC50, IC80, and IC85 doses against MDA-MB-231 cells were 30.4, 12.1, and 9.7 µM, respectively, confirming selective anticancer toxicity.[300]
GBC-SD, NOZ, and SGC-996 cellsCCK-8 assay0–80 µmol/L for 48 hPterostilbene exhibits cytotoxic effects on all three cell lines. The estimated IC50 for GBC-SD cells was above 80 µmol/L, between 40–60 µmol/L for NOZ cells, and approximately 80 µmol/L for SGC-996 cells.[301]
C6, LN18, LN229, T98G, U87, and HUVECs cellsMTT assay0, 20, 40, 80, and 100 µM for 24, 48, and 72 hPterostilbene inhibited cell viability on C6, LN18, LN229, T98G, and U87 cells. The IC50 values of pterostilbene treatment for 48 h were 30.10 μM for C6 cells, 22.30 μM for LN18 cells, 37.56 μM for LN229 cells, 32.93 μM for T98G cells, and 46.18 μM for U87 cells. Pterostilbene had a minimal impact on HUVEC cells compared to the previously mentioned cell lines.[302]
PiceatannolAGS, SK-MES-1, and J82 cellsMTT assay0–100 µg/mL for 72 hBesides enhancing gemcitabine’s cytotoxic and apoptotic effects, piceatannol actively inhibited SK-MES-1 cell viability. The synergistic combination increased the expression of the Bcl-2 pro-apoptotic protein family.
IC50 concentrations for AGS, SK-MES-1, and J82 cells were 10.8 ± 0.7, 7.64 ± 0.5, and 6.7 ± 0.3 µg/mL, respectively.
[303]
T24 and HT1376 cellsXTT assay0.5, 2.5, 5, and 10 µM for 48 hPiceatannol showed a dose-dependent inhibitory effect on the proliferation of both T24 and HT1376 cell lines. The IC50 values were 3.9 and 4.6 µM, respectively.[304]
HL-60 cellsMTT assay10–200 µM for 24, 48, and 72 hPiceatannol significantly inhibited HL-60 cell growth in a time- and dose-dependent manner. A moderate inhibition of HL-60 cells viability was observed after a 72 h treatment with piceatannol at 10, 20, and 50 µM. The highest inhibition was observed after 24, 48, and 72 h treatment with 100–200 µM concentration range.[305]
WM266-4 and A2058 cellsMTT assay0, 1, 10, 20, 40, 100, and 200 µM for 36 hBoth cell lines exhibited decreased viability following piceatannol treatment.
The calculated IC50 was 29.4 μM for WM266-4 cells, and 15.6 μM for A2058 cells.
[306]
LNCaP, Du145, and PC3M cellsMTS assay1, 5, 10, 25, 50, and 100 µM for 6 days.All cell lines were susceptible to piceatannol treatment, exhibiting declining cellular activity. The IC50 values obtained were 31.7 μM for LNCaP, 23.2 µM for Du145, and 34.6 μM for PC3M cells.[307]
B16 cellsMTT assay5–400 µM for 24 hPiceatannol exhibited cytotoxicity effects, resulting in decreased cell viability.
The obtained IC50 was 1.53 μM.
[308]
U937 cellsMTT assay0–100 µM for 24 hExposure to piceatannol inhibited cell viability, with an associated IC50 of 5 µM.[309]
NCI-H522 cellsWST-8 assay10, 30, 50, 80, and 100 µM for 24, 48, and 72 hPiceatannol treatment notably decreased NCI-H522 cell viability. The IC50 values at each timepoint were 53, 23, and 17 µM, respectively.[310]
Caco-2 and HCT-116 cellsCrystal violet assay12.5, 25, 50, 100, and 200 µM for 24, 48, and 72 hPiceatannol cytotoxic effects led to a decrease in cell viability fin both cell lines after a 72 h treatment.
The obtained IC50 of piceatannol in Caco-2 and HCT-116 cells was 50 µM.
[311]
L1210, K562, and HL-60 cellsTrypan blue dye exclusion0–500 µM for 24 hAll cell lines were sensible to piceatannol’s cytotoxic effects.
The calculated IC50 values of piceatannol were 50 µmol/L, <10 µmol/L, and <20 µmol/L for K562, HL-60, and L1210 cells, respectively.
[312]
RAW 264.7 cellsMTT assay0–50 µg/mL for 48 hPiceatannol exhibits inhibitory activity, with an associated IC50 value of 5.7 µg/mL.[313]
HSG, HL-60 HSC-2, and HSC-3, (tumor cell lines)
HPC, HGF, and HPLF (normal cells line),
MTT assay (HGF, HPC, HPLF, HSC-2, HSC-3, AND HSG)
Trypan blue dye exclusion (HL-60)
10–1000 μM for 24 hPiceatannol exhibits greater inhibitory effects on cancer cells compared to normal cells. The IC50 values for cancer cell lines were 63 µM for HSC-2, 232 µM for HSC-3, 373 µM for HSG, and 11 µM for HL-60 cells. In contrast, the IC50 values for normal cells were 367 µM for HGF, 414 µM for HPC, and >1000 µM for HPLF cells.[314]
SW1990 and PANC-1 cellsCCK-8 assay1, 10, 20, 40, 100, and 200 μM for 72 hPiceatannol inhibited up to 50% cell proliferation for both cell lines.
The IC50 value for SW1990 cells was 30.69 µM, while for PANC-1 cells it was 21.82 µM.
[315]
MOLT-4 cellsNRU assay0.05, 15, 25, 50, and 100 μM for 48 hPiceatannol reduced cellular viability with a calculated IC50 of 45.5 µM.[316]
HeLa cellsMTT assay0–250 µM for 48 hPiceatannol decreased cell viability and the associated IC50 value was 375.20 µM.[317]
Mouse embryonic stem cells (ESCs)MTT assay1–20 µM for 72 hHigh concentrations of piceatannol exhibited cytotoxicity.
The obtained IC50 value was 13.5 µM.
[318]
C6 cells (proliferating and growth arrested)Lowry method1–100 µM for 72 h in proliferating cells and 24 h in growth-arrested cellsPiceatannol exhibits cytotoxic effects on both growth-arrested and proliferating cells. The IC50 concentration for growth-arrested cells was 20 ± 2 µM, while for proliferating cells it was 28 ± 4 µM.[319]
10ScNCr/23, A-431, RAW 264.7, and CCR-CEM cellsTrypan blue dye exclusion0–50 µM for 24 hPiceatannol exhibits inhibitory effects on all cell lines. The IC50 concentration in RAW 264.7 cells were 1.30 ± 0.12 µM.[320]
THP-1 cellsLight microscopy10, 20, 30, 40, and 50 µM for 48 hSignificant cytotoxic effects with noticeable cell shrinkage were observed at concentrations above 30 µM.[321]
(-)-ε-viniferinHSC-2, HSC-3, HCF, HPC, HPLF, HSG, and HL-60 cellsMTT assay in adherent cells
Trypan blue dye exclusion in non-adherent cells
0–1000 µM for 24 hThe four tumor cell lines (HSC-2, HSC-3, HSG, and HL-60) were more sensitive to (-)-ε-viniferin than the remaining normal cell lines.
The IC50 values were 42 µM for HSC-2 cells, 84 µM for HSC-3 cells, 111 µM for HCF cells, 146 µM for HPC cells, 94 µM for HPLF cells, 110 µM for HSG cells, and 31 µM for HL-60 cells.
[314]
P-388 cellsMTT assay0–100 µM for 48 hε-viniferin moderately inhibited cell viability in comparison to hopeaphenol, which exhibited a greater effect.
The IC50 measured at 18.1 ± 0.7 µM.
[255]
HepG2 and Chang cellsMTT assay1.56–200 µg/mL for 72 hNo cytotoxic effect was detected in either cell lines.[322]
(+)-ε-viniferin RAW 264.7 cellsMTT assay1, 5, and 10 µM for 12 hCell viability was significantly reduced to 60% after exposure of 10 µM.
IC50 was not determined.
[323]
trans-ε-viniferin K562, L1210, and HCT116 cellsMTT assay0–50 µM for 48 hNo cytotoxicity was detected.
The IC50 was assumed to be above 50 µM.
[324]
AGS, MRC-5, SK-MES-1, and J82 cellsMTT assay0–100 µg/mL for 72 hCytotoxicity was observed for all cell lines tested.
The IC50 values were 42.6 ± 1.7 µM for AGS cells, 49.9 ± 3 µM for MRC-5 cells, 78.8 ± 3.3 µM in SK-MES-1 cells, for 56.7 ± 1.2 µM in J82 cells.
[303]
Mouse primary astrocytes and neurons co-culture CellTitel 96 ® Aqueous assay1, 5, 10, 20, 50, and 100 µM for 24 hCell viability was significantly reduced when cells were exposed to concentrations of 50 and 100 µM.[325]
AGS, COLO 205, HepG2, HL-60, and HT-29 cellsMTT assay0–100 µg/mL for 48 hDose-dependent cytotoxicity was reported, with a greater effect observed in HL-60 cells.
The determined IC50 values were: 9.3 ± 0.3 µM in AGS cells, 85.5 ± 8.1 µM in COLO 205 cells, 7.7 ± 0.2 µM in HepG2 cells, 5.6 ± 1.4 µM in HL-60 cells, and 13.9 ± 0.1 µM in HT-29 cells.
[326]
Hep3B, HepG2, and HH4 cellsCrystal violet assay0–200 µM for 24, 48, and 72 hIt was more cytotoxic to Hep3B cells and reduced cell quantity in a dose- and time-dependent manner. Higher amounts were required to cause toxicity in HH4 cells.
The IC50 values obtained were the following:
- Hep3B cells: 108.1 ± 31.8 µM (24 h), 73.9 ± 17.3 µM (48 h), 63.1 ± 10.8 µM (72 h).
- HepG2 cells: 140 ± 39.7 µM (24 h), 103.8 ± 19.2 µM (48 h), 94.8 ± 28.3 µM (72 h).
- HH4 cells: >200 µM (24 h), 192.7 ± 21.1 µM (48 h), 177.9 ± 20.5 µM (72 h).
[326]
HepG2 and Caco-2 cellsMTS assay, NRU, and protein content0–100 µg/mL for 24 and 48 hFor every endpohint examined, both cell lines showed a time-dependent decline in cell viability. The IC50 values were:
- HepG2: 28.28 ± 2.15 µg/mL 24 h and 17.85 ± 3.03 µg/mL for 48 h.
- Caco-2 cells: 36.72 ± 3.01 µg/mL for 24 h and 20.63 ± 1.25 µg/mL 48 h.
[327]
trans-ε-viniferin and cis-ε-viniferinHeLa, MCF-7, C6, HepG2, and HT-29 cellsMTT assay0–100 μM for 70 hCis- and trans-ε-viniferin to all cell lines, although greater significance was registered for C6 and HeLa cells.
The IC50 values for trans-ε-viniferin were:
- 20.4 µM in HeLa cells, 44.8 µM in MCF-7 cells, 18.4 µM in C6 cells, 74.3 µM in HepG2 cells, and 88.4 µM in HT-29 cells.
The IC50 values for cis-ε-viniferin were:
- 21.5 µM in HeLa cells, and 47.2 µM in MCF-7 cells, 20.1 µM in C6 cells, 76.2 µM in HepG2 cells, and 90.2 µM in HT-29 cells.
[328]
ε-viniferin WSU-CLL cellsTrypan blue dye exclusion0–100 µM for 24, 48, and 72 hA concentration- and time-dependent decrease in cell viability was observed, with resveratrol overperforming ε-viniferin. Inhibited cell proliferation was accompanied by a reduction in DNA synthesis.
The IC50 value determined at 72 h was 60 µM.
[248]
HL-60 cellsMTT assay10–200 µM for 24 hCell viability decreased in a concentration-dependent manner.
The IC50 was 33 µM.
[329]
HepG2 cellsTrypan blue dye exclusion30 µM for 24, 48, and 72 h. 1, 5, 10, 30, 60, and 100 µM for 48 hAt 60 µM, ε-viniferin completely blocks cell proliferation. After 48 h, the toxicity potential of ε-viniferin was lower than resveratrol. The IC50 for 48 h was 58.4 µM.[258]
SW480 cellsTrypan blue dye exclusion and MTT assay30 µM for 24, 48, 72, and 96 h in trypan blue dye exclusion. 3, 30, 60, and 100 µM for 48 h in coulter counterCells exposed to ε-viniferin grew similarly to the control group, with a reduced growth rate and increasing percentage of cell inhibition. In the MTT assay, no significant inhibition of cell proliferation was recorded.[275]
VSMCs cellsMTS assay10, 20, and 30 µM for 48 hThe potential for arresting cell proliferation rate of ε-viniferin at 20 µM was significantly higher than resveratrol’s at 20 and 30 µM.[330]
SK-MEL-25 and HT-144 cells MTT assay
Trypan blue dye exclusion
25–200 µM for 24, 48, and 72 hBoth melanoma lines showed time- and dose-dependent reduction in survival. The IC50 for 48 h was 60 µM.[249]
C6 cellsWST-1 assay95 and 130 µM 12, 24, and 48 hProliferation decreased at all doses and times tested in C6 cells.[253]
Caco-2 cellsMTT and NRU assays1.56, 3.12, 6.25, 12.5, 25, 50, and 100 µM for 24 hAt and above 25 µM, cell viability in Caco-2 cells decreased. ε-viniferin was slightly more effective than resveratrol.[331]
Vascular endothelial cells (VECs)H2O2-induced cytotoxicity10, 20, and 30 µM for 24 hε-viniferin effectively protected cells from cytotoxic effects of H2O2. A 24 h pre-treatment with ε-viniferin reduced intracellular ROS.[332]
VECsH2O2-induced cytotoxicity5 and 10 µM for 24 hAt 10 μM, a pre-treatment with ε-viniferin conferred VEC with resistance against H2O2-induced oxidative stress.[333]
A2058, A549, HOS, U2OS, and MCF-10A cellsMTT assay1–15 μM for 24, 48, and 72 hε-viniferin exhibited a time- and dose-dependent decrease in the viability of HOS, U2OS, and A549 cells, but not in A2058 cells. [334]
ε-viniferin glucosidePC12 cellsMTT assay0–10 µM for 24 hCell viability was not significantly altered following the exposure to the stilbene.[335]

3.5. Cardiovascular and Metabolic Benefits

Stilbenes act on various factors involved in the bioavailability of NO, such as arginase, dimethylargininase (DDAH), and asymmetric dimethylarginine (ADMA), thereby modulating NO production during oxidative stress [163,336,337,338]. Also, stilbenes influence metabolic pathways like adipogenesis, lipogenesis, lipolysis, thermogenesis, and fatty acid oxidation. They regulate PPARγ, sterol regulatory element-binding protein 1c (SREBP-1c), uncoupling (UCPs), sirtuin 1 (SIRT1), lipoprotein lipase (LPL), fatty acid synthase (FAS), and acetyl-CoA carboxylase (ACC). Piceatannol and pterostilbene modulate lipid metabolism via ACC, SREBP1, PPAR-γ, and PPAR-α [72]. Pterostilbene demonstrates greater in vivo efficiency than resveratrol due to its higher bioavailability and greater capacity to bind three amino acids of PPARα versus two for resveratrol [339]. Resveratrol has vasodilatory properties, enhancing endothelial NO production and increasing the expression of eNOS in rat arteries while decreasing the expression of NADPH oxidase [340,341]. Studies show that resveratrol activates membrane-associated structures, such as estrogen receptors, which trigger an intracellular signaling cascade targeting the AMPK pathway. This activation can phosphorylate eNOS at serine 1177 [342]. Additionally, resveratrol activates SIRT1, reducing acetylation and leading to eNOS activation. Resveratrol improves SIRT1-dependent NO bioavailability induced by insulin in HUVEC cells cultured under high glucose conditions for 48 h [343]. Another study shows that resveratrol suppresses SIRT1 inhibition, reducing eNOS acetylation in HUVEC cells exposed to H2O2-induced oxidative stress [344].
Furthermore, resveratrol increases NO production in blood platelets via the PI3K/protein kinase B (Akt) pathway, activates vasodilator-stimulated phosphoprotein, and inhibits p38MAPK. This leads to reduced platelet activation and ROS production [345]. Resveratrol also dose-dependently increases RNA synthesis of VEGF and eNOS and reduces endothelin secretion in HUVEC cells, lowering blood pressure and improving vasodilation [346]. In human coronary smooth muscle cells incubated with resveratrol, cGMP synthesis increased, activating cGMP-dependent protein kinase, which in turn activated potassium channels, leading to hyperpolarization and relaxation of vascular smooth muscle cells [347].
In vivo studies in rats with accelerated atherosclerosis and metabolic diseases showed improved endothelial dysfunction [57]. In another study using obese Zucker rats, a model for metabolic syndrome, chronic resveratrol administration increased eNOS expression and reduced dyslipidemia and hypertension [348]. Similarly, in hypercholesterolemic rabbits treated with resveratrol, plasma NO concentration increased, and endothelial dysfunction was reduced [349]. However, in estrogen receptor-deficient mice, resveratrol showed no vasorelaxation effects compared to normal mice, demonstrating that estrogen receptors mediate the intracellular effects of resveratrol. Activation of these receptors leads to phosphorylation of Src, Erk ½, and eNOS, resulting in NO synthesis activation [342].
Stilbenes, such as resveratrol found in grapes, need further testing in humans. In a 12-week study where 75 mg/day of resveratrol was administrated to 15 healthy non-obese postmenopausal women, no changes were detected in metabolic functions, such as AMPK signaling, inflammatory markers, insulin, and mitochondrial functions [350]. The absence of effects in humans aligns with previous results in healthy rodents [351], suggesting that resveratrol’s effects are limited to conditions like type II diabetes, dyslipidemia, and obesity [117].
A study on resveratrol’s role in primary and secondary cardiovascular disease prevention involved a small sample size (n = 75) over one year, limiting clinical relevance. Participants were divided into three groups: one received grape extract with 8.1 mg/day of resveratrol for six months followed by an increase in the stilbene’s concentration to 16.2 mg/day for another six months, a second group received grape extract without resveratrol, and the third received a placebo. All participants were on statins and treated per cardiovascular disease prevention guidelines [352]. Results were promising, with the resveratrol-treated group showing improved inflammatory and fibrinolytic profiles in comparison to the remaining groups [352]. Resveratrol provided additional benefits for patients at high risk of cardiovascular disease beyond other phenolic compounds in grape extract, complementing the current primary prevention guidelines [353].
In animal models, resveratrol and pterostilbene demonstrated anti-hyperlipidemic effects, but clinical trials in humans showed no significant changes in low-density lipoprotein (LDL)/high-density lipoprotein (HDL) ratios. A meta-analysis of randomized clinical trials revealed that the use of resveratrol as a supplement did not induce significant alterations in lipid parameters, including LDL, HDL, total cholesterol, and triglycerides [354]. Pterostilbene at high and low doses promotes an increase in LDL without affecting HDL or triglycerides in a placebo-controlled trial [355]. Similarly, grape extract administration did not influence LDL levels [117]. However, in a secondary prevention study of cardiovascular diseases, grape extract with resveratrol (8.1 mg/day for six months, then 16.2 mg/day) in patients with stable coronary artery disease and dietary restrictions showed increased anti-inflammatory adiponectin and reduced plasminogen activator inhibitor-1 (PAI-1) [356].
These findings suggest that resveratrol has cardioprotective effects, improving anti-inflammatory responses and preventing atherothrombotic signaling (Figure 3) [356].
Preclinical data have demonstrated that resveratrol can be utilized in diabetes management through correction of insulin signaling defects, improvement in insulin resistance, and prevention of pancreatic beta cells’ dysfunction [357]. Moreover, resveratrol prevents hyperglycemia in animal models for diabetic pathology by promoting glucose uptake and GLUT4 translocation to the caveolar membrane within the diabetic myocardium [358]. It also improves glucose tolerance and reduces the expression of advanced glycation endproduct (AGE) receptors in the liver and kidneys of diabetic rats [359].
Resveratrol exhibits actions that inhibit reactive oxygen and nitrogen species, such as superoxide anion (O2•−), hydroxyl radical (OH), H2O2, and malondialdehyde (MDA), while increasing levels of antioxidant enzymes like SOD, catalase, and glutathione peroxidase in diabetic animals [360]. It also inhibits pro-inflammatory signaling through NF-κB and reduces the production of inflammatory cytokines, including IL-1β, IL-4, and IL-6 and TNF-α, [360]. Additionally, resveratrol increases insulin sensitivity, glucose tolerance, and mitochondrial biogenesis via AMPK-dependent pathways [361]. A study showed that resveratrol did not achieve the same effects in AMPK-deficient mice, highlighting the essential role of this protein in the metabolic actions of resveratrol [361].
Resveratrol’s role in glycemic control remains slightly uncertain, as some human studies lack significant evidence of the stilbene’s effect on metabolic dysfunctions. Daily co-administration of metformin or glibenclamide and 250 mg of resveratrol for three months improves glycemic parameters in patients with type 2 diabetes, when compared with those receiving therapies lacking the stilbene [362]. As reported by Movahed and colleagues, 1 g/day of resveratrol for 45 days reduced systolic blood pressure, fasting glucose serum levels, and HbA1c [363]. Furthermore, a much lower daily 5 mg dose of resveratrol was found to reduce systolic blood pressure, HbA1c levels, and improve insulin sensitivity, while not affecting the homeostatic model assessment of insulin resistance, in a 28-day long treatment [363].
In contrast, a randomized clinical trial by Thazhath et al., with 500 mg of resveratrol twice daily for five weeks in diet-controlled type 2 diabetes patients, showed no significant effects on glycemic control [364]. No differences were observed in fasting glucose, postprandial glucose, HbA1c, gastric emptying, or glucagon-like peptide-1 (GLP-1) secretion between the resveratrol and placebo groups. Similarly, a six-month treatment did not show metabolic improvements in type 2 diabetes patients [365]. Therefore, the effects of resveratrol on human diabetes remain not fully understood [117].
Pterostilbene has shown promising results in glycemic control in obese rats with insulin resistance, enhancing hepatic glucokinase activity and glucose uptake in skeletal muscle [366]. In vitro, studies indicate that pterostilbene protects pancreatic beta cells from oxidative stress and apoptosis [367]. Pterostilbene, along with other constituents of Pterocarpus marsupium, has demonstrated anti-hyperglycemic properties [368,369]. However, human data on pterostilbene are still limited. One study administered sea buckthorn and blueberry extract to children with type 1 diabetes for two months, resulting in elevated SOD and glutathione peroxidase levels and reduced HbA1c levels [370].
Piceatannol also has glucose-regulating capabilities, increasing glucose disposal. Minakawa et al. observed that piceatannol promotes glucose uptake in cultured myotubes in a dose-dependent manner by activating AMPK and inducing glucose transporter type 4 (GLUT4) translocation [371]. AMPK activators are promising for type 2 diabetes treatment since AMPK stimulates GLUT4 translocation, with skeletal muscle being the primary site of glucose clearance [372,373]. Piceatannol was shown to lower fasting glucose in vivo by activating AMPK in myotubes and minimally impacting insulin secretion in vivo, establishing it as a glucose modulator [371,374,375]. Insulin signaling is modified by piceatannol in different tissues, for instance, it blocked insulin action in adipocytes but improved endothelial function under inflammatory stress [376,377,378].

3.6. Neuroprotective Effects and Cognitive Function

In neurodegenerative diseases (NDs), there is progressive damage to the structure and function of neurons. The three main NDs are Alzheimer’s disease, Parkinson’s disease, and amyotrophic lateral sclerosis. Each presents different clinical signs; however, the pathological processes appear similar, suggesting common biological pathways in the onset and progression of NDs. Factors involved in NDs include aging, lifestyle, and genetics [379]. Generally, neuroinflammation and oxidative stress are the primary causes of neuronal dysfunction and death, along with excitotoxicity, mitochondrial dysfunction, and apoptosis [380]. Oxidative stress in neurodegeneration is correlated with the progression of Parkinson’s and Alzheimer’s diseases [381]. One hypothesis involves the use of natural products due to their centuries-long application in human diseases [382,383,384]. In recent decades, many studies have described the protective effects of natural polyphenols and their active derivatives against various diseases, including cardiovascular diseases, diabetes, cancers, and NDs [385]. Additionally, natural compounds demonstrate great potential as neuroprotective agents for treating NDs due to their inherently multi-target profiles [386,387].
The neuroprotective effects of stilbenes include antioxidants and anti-inflammatory properties (Figure 3) [388,389,390,391,392]. Resveratrol protects neurons from ROS and improves motor coordination in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced parkinsonism in rats by neutralizing hydroxyl radicals [388]. Furthermore, resveratrol improves mitochondrial function, motor coordination, and neuronal survival, inhibits β-secretase, enhances non-amyloidogenic amyloid precursor protein (APP) cleavage, and increases clearance of beta-amyloid (Aβ) peptides [393]. Moreover, resveratrol protects against lipopolysaccharide (LPS)-induced dopaminergic neurodegeneration by inhibiting microglial activation and NF-κB signaling [394]. In Alzheimer’s disease, resveratrol shows therapeutic potential by reducing amyloid plaques in the brain. Marambaud et al. demonstrated that resveratrol may not inhibit Aβ production but promotes proteasome-dependent Aβ degradation [395]. Winemaking residues are a source of piceatannol and vitisinol C, which inhibits Aβ aggregation [396,397].
Pre-treatment with resveratrol in rats subjected to cerebral ischemia reperfusion injury increased Nrf2 and HO-1 levels, reducing oxidative damage during ischemic events within the brain [398]. This pre-treatment also results in improved neurological scores with associated reductions in brain water content and infarct volume. In models of global cerebral ischemia, resveratrol blocked neuronal death by activating PI3K/Akt signaling, downregulating GSK-3β, and regulating CREB (cAMP response element-binding protein) [399].
Resveratrol improves cognition in animal models of vascular dementia, showing effects on cerebral ischemia [400]. In this study, vascular dementia was induced by bilateral common carotid artery occlusion for 8–12 weeks. Resveratrol treatment improved learning and memory scores by reducing malondialdehyde, a lipid peroxidation product, and increasing antioxidant enzyme levels, such as glutathione and SOD, in the cerebral cortex and hippocampus [117,400].
Pterostilbene acts as a neuroprotectant by counteracting glucose-induced damage in neuroblastoma cells, preventing the decline in viable cells and ROS generation in a concentration-dependent manner [389]. It also increased mitochondrial cytochrome C, mitochondrial complex I and III activities, and membrane potential, along with Nrf2, HO-1, and GST levels, offering protection against neuronal oxidative stress [389]. Pterostilbene improves memory by increasing RE1-silencing transcription factor (REST), postsynaptic density protein 95 (PSD-95), and mitochondrial porin 1 in the dentate gyrus of aged rats. Resveratrol improves memory in healthy and diabetic individuals with subclinical impairment but not in full Alzheimer’s cases [393].
Oxyresveratrol has shown neuroprotective effects. Studies in rat cortical neurons demonstrated that it prevented Aβ (25–35)-induced damage by reducing cytosolic Ca2+ levels, inhibiting glutamate release, and lowering ROS generation [401]. Another study by Andrabi et al. demonstrated that, in a rat model of transient middle cerebral artery occlusion, oxyresveratrol significantly reduced cerebral infarct volume and improved subsequent neurological deficits through inhibition of cytochrome C release and caspase-3 activation [402].
In a 52-week long randomized placebo-controlled clinical trial, doses of resveratrol raging between 500 and 1000 mg administered twice daily were well tolerated by Alzheimer’s patients, reducing Aβ40 levels in the cerebrospinal fluid and plasma, although other Alzheimer’s biomarkers were unaffected [403]. Another trial with a daily supplementation of 200 mg of resveratrol and 320 mg of quercetin resulted in an improvement in memory performance when co-administrated in overweight elderly individuals [404].
A recent study showed that a double daily administration of 75 mg of resveratrol, for a period of 14 weeks, improved cerebrovascular function, cognition, and mood in postmenopausal women. Additionally, it enhanced cognitive performance and cerebral blood flow in individuals with type 2 diabetes [405,406]. In conclusion, human studies have demonstrated the beneficial effects of resveratrol in improving memory and cognition in healthy individuals and those with diabetes-related cognitive impairment, but not in Alzheimer’s patients [117].

4. Conclusions and Future Perspectives

The wine industry represents a significant opportunity for obtaining high-value byproducts with potential industrial applications. Vineyard waste is a rich source of compounds with antioxidant effects (specifically stilbenes), which can act as potent reactive free radical scavengers, enzyme activators and/or inhibitors, antibacterial agents, anti-inflammatory agents, and anticancer agents, among other health-beneficial agents. For example, stilbenes inhibit platelet aggregation, possibly through the inhibition of COX-1. They also reduce markers of oxidative stress and inflammation, such as TNF-α and IL-1β, in various models of ischemia reperfusion injury, and inhibit cardiomyocyte hypertrophy through the activation of AMPK. In addition, stilbenes exert beneficial effects in cardiovascular conditions, such as improving glucose tolerance and insulin resistance in animal models of diabetes. Stilbenes exhibit anticancer effects through the inhibition of cancer cell proliferation, oxidative stress, and inflammation, engaging in regulatory roles of cell death mechanisms. From a neurological perspective, stilbenes can upregulate Nrf2 and HO-1 expression to mitigate oxidative damage during cerebral ischemia. Thus, their neuroprotective effects are largely attributed to their antioxidant activity. Stilbenes also exhibit a broad range of functions, including microbicidal and antifungal activities, various effects on the reversal of drug resistance, and biofilms and virulence factors, highlighting the potential of stilbenes as antimicrobial agents. Although the results are promising, most of the evidence comes from in vitro and in vivo studies, with limited clinical trials available to confirm the beneficial effects of stilbenes in humans.
From an industrial perspective, stilbenes have attracted growing interest not only in the pharmaceutical sector but also in the food and cosmetics industries. Their incorporation into biodegradable packaging, natural cosmetics, and functional supplements highlights their potential as multifunctional agents, aligning with the increasing demand for sustainable and bioactive products.
However, to allow large-scale applications, significant advancements are still required in extraction, purification, and formulation technologies. Low water solubility, limited bioavailability, and rapid metabolism remain technical challenges that must be addressed, potentially through innovative strategies such as nanoencapsulation, cyclodextrin complexation, or incorporation into polymeric matrices.
Future prospects for the utilization of stilbenes include the following: the optimization of extraction methods for improved efficiency and selectivity and reduced environmental impact including decreased energy and time consumption, the preservation of stilbene integrity, and the application of green chemistry principles; the development of controlled-release systems for pharmaceutical and cosmetic applications, including the formulation of nanoparticles with stilbenes to enhance bioabsorption and bioavailability for drug delivery; comprehensive toxicological and regulatory evaluations to ensure the safety of stilbene use in consumer products; the exploration of lesser-known stilbenes beyond resveratrol to broaden the range of applications and therapeutic potential, including resveratrol derivatives such as pterostilbene, pinosylvin, ε-viniferin, piceatannol, rhapontigenin, and isorhapontigenin, among others; and integration into circular production chains, contributing to the sustainability of the wine sector and fostering the bioeconomy, thereby fostering eco-design, one of the seven pillars of the circular economy.
In conclusion, stilbenes demonstrate great potential for multiple human health benefits. However, further studies are needed due to the number and complexity of the cellular processes involved. Additionally, because of their low concentrations in foods and associated rapid metabolism and excretion in mammalian organisms, improvements in stability, solubility, and delivery systems are required to enable clinical application. Moreover, there is limited information with regard to the potential beneficial properties of less-studied stilbenes. Therefore, more research is still necessary to uncover the impact of lesser-known stilbenes on health.

Author Contributions

Conceptualization, L.P.B., G.C., S.S. and A.P.D.; investigation, L.P.B.; writing—original draft preparation, L.P.B. and Â.L.; writing—review and editing, L.P.B., Â.L., G.C., S.S. and A.P.D.; supervision, G.C., S.S. and A.P.D.; project administration, S.S. and A.P.D. All authors have read and agreed to the published version of the manuscript.

Funding

This work was developed within the scope of the CICS-UBI (Portugal) core grant (https://doi.org/10.54499/UIDB/00709/2020) and the CICS-UBI programmatic grant (https://doi.org/10.54499/UIDP/00709/2020) with national funds registered in the budget of the FCT. Ângelo Luís acknowledges the Scientific Employment contract (Microbiology), funded by Portuguese Foundation for Science and Technology (FCT), within the scope of DL57/2016 (Reference: DL57/2016/CP1354/CT0010). Luis Brás acknowledges funding from individual PhD fellowships from FCT (UNITA) by project PRT/BD/154423/2022 (DOI https://doi.org/10.54499/PRT/BD/154423/2022).

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
UVUltraviolet
STSStilbene synthase
DWDry weight
MAEMicrowave-assisted extraction
UAEUltrasound-assisted extraction
PSEPressurized solvent extraction
COXCyclooxygenase
Beta-amyloid
ROSReactive oxygen species
RNSReactive nitrogen species
SODSuperoxide dismutase
NADPHNicotinamide adenine dinucleotide phosphate
GPxGlutathione peroxidase
GSTGlutathione-S-transferase
NF-κBNuclear factor κB
eNOSEndothelial nitric oxide synthase
LDLLow-density lipoprotein
SIRT1Sirtuin 1
H2O2Hydrogen peroxide
TNF-αTumor necrosis factor-α
AMPKAdenosine monophosphate-activated protein kinase
LPSLipopolysaccharide
HO-1Heme oxygenase-1
Nrf2Nuclear factor erythroid 2-related factor 2
STAT3Signal transducer and activator of transcription 3
NRUNeutral red uptake
BH4Tetrahydrobiopterin
cAMPCyclic adenosine monophosphate
cGMPCyclic guanosine monophosphate
NDsNeurodegenerative diseases
CCR1Chemokine receptor
PGE2Prostaglandin E2
GSHGlutathione
PTENPhosphatase and TENsin homolog
PKBProtein kinase B
GSK-3βGlycogen synthase kinase-3β
VEGFVascular endothelial growth factor
CSCCancer stem cell
VECsVascular endothelial cells
PI3KPhosphoinositide 3-kinase
AktProtein kinase B
HDLHigh-density lipoprotein

References

  1. International Organisation of Vine and Wine. 2023 World Wine Production—Oiv First Estimates. Available online: https://www.fao.org/3/i7042e/i7042e.pdf (accessed on 11 August 2025).
  2. OIV. State of the World Vine and Wine Sector in 2023; International Organisation of Vine and Wine: Paris, France, 2023. [Google Scholar]
  3. Bustamante, M.A.; Moral, R.; Paredes, C.; Pérez-Espinosa, A.; Moreno-Caselles, J.; Pérez-Murcia, M.D. Agrochemical characterisation of the solid by-products and residues from the winery and distillery industry. Waste Manag. 2008, 28, 372–380. [Google Scholar] [CrossRef]
  4. Bordiga, M.; Travaglia, F.; Locatelli, M.; Arlorio, M.; Coïsson, J.D. Spent grape pomace as a still potential by-product. Int. J. Food Sci. Technol. 2015, 50, 2022–2031. [Google Scholar] [CrossRef]
  5. Yu, J.; Ahmedna, M. Functional components of grape pomace: Their composition, biological properties and potential applications. Int. J. Food Sci. Technol. 2013, 48, 221–237. [Google Scholar] [CrossRef]
  6. Broome, J.C.; Warner, K.D. Agro-environmental partnerships facilitate sustainable wine-grape production and assessment. Calif. Agric. 2008, 62, 133–141. [Google Scholar] [CrossRef]
  7. Zacharof, M.-P. Grape Winery Waste as Feedstock for Bioconversions: Applying the Biorefinery Concept. Waste Biomass Valorization 2017, 8, 1011–1025. [Google Scholar] [CrossRef]
  8. Prozil, S.O.; Evtuguin, D.V.; Lopes, L.P.C. Chemical composition of grape stalks of Vitis vinifera L. from red grape pomaces. Ind. Crops Prod. 2012, 35, 178–184. [Google Scholar] [CrossRef]
  9. Kalli, E.; Lappa, I.; Bouchagier, P.; Tarantilis, P.A.; Skotti, E. Novel application and industrial exploitation of winery by-products. Bioresour. Bioprocess. 2018, 5, 46. [Google Scholar] [CrossRef]
  10. Chowdhary, P.; Gupta, A.; Gnansounou, E.; Pandey, A.; Chaturvedi, P. Current trends and possibilities for exploitation of Grape pomace as a potential source for value addition. Environ. Pollut. 2021, 278, 116796. [Google Scholar] [CrossRef] [PubMed]
  11. Toma, D.-I.; Baroi, A.M.; Din, A.; Vizitiu, D.E.; Fierascu, I.; Fierascu, R.C. Grapevine Plant Waste Utilization in Nanotechnology. AgroLife Sci. J. 2024, 13, 203–216. [Google Scholar] [CrossRef]
  12. Wei, M.; Ma, T.; Ge, Q.; Li, C.; Zhang, K.; Fang, Y.; Sun, X. Challenges and opportunities of winter vine pruning for global grape and wine industries. J. Clean. Prod. 2022, 380, 135086. [Google Scholar] [CrossRef]
  13. Ilyas, T.; Chowdhary, P.; Chaurasia, D.; Gnansounou, E.; Pandey, A.; Chaturvedi, P. Sustainable green processing of grape pomace for the production of value-added products: An overview. Environ. Technol. Innov. 2021, 23, 101592. [Google Scholar] [CrossRef]
  14. Khan, Z.A.; Iqbal, A.; Shahzad, S.A. Synthetic approaches toward stilbenes and their related structures. Mol. Divers. 2017, 21, 483–509. [Google Scholar] [CrossRef] [PubMed]
  15. Baroi, A.M.; Popitiu, M.; Fierascu, I.; Sărdărescu, I.-D.; Fierascu, R.C. Grapevine Wastes: A Rich Source of Antioxidants and Other Biologically Active Compounds. Antioxidants 2022, 11, 393. [Google Scholar] [CrossRef]
  16. Beres, C.; Costa, G.N.S.; Cabezudo, I.; da Silva-James, N.K.; Teles, A.S.C.; Cruz, A.P.G.; Mellinger-Silva, C.; Tonon, R.V.; Cabral, L.M.C.; Freitas, S.P. Towards integral utilization of grape pomace from winemaking process: A review. Waste Manag. 2017, 68, 581–594. [Google Scholar] [CrossRef]
  17. Cuccia, P. Ethics+ economy+ environment= sustainability: Gambero Rosso on the front lines with a new concept of sustainability. Wine Econ. Policy 2015, 4, 69–70. [Google Scholar] [CrossRef]
  18. Iuga, M.; Mironeasa, S. Potential of grape byproducts as functional ingredients in baked goods and pasta. Compr. Rev. Food Sci. Food Saf. 2020, 19, 2473–2505. [Google Scholar] [CrossRef]
  19. Contreras, M.d.M.; Romero-García, J.M.; López-Linares, J.C.; Romero, I.; Castro, E. Residues from grapevine and wine production as feedstock for a biorefinery. Food Bioprod. Process. 2022, 134, 56–79. [Google Scholar] [CrossRef]
  20. Bensid, A.; El Abed, N.; Houicher, A.; Regenstein, J.M.; Özogul, F. Antioxidant and antimicrobial preservatives: Properties, mechanism of action and applications in food—A review. Crit. Rev. Food Sci. Nutr. 2022, 62, 2985–3001. [Google Scholar] [CrossRef]
  21. Vilela, A.; Pinto, T. Grape Infusions: Between Nutraceutical and Green Chemistry. Sustain. Chem. 2021, 2, 441–466. [Google Scholar] [CrossRef]
  22. Teixeira, A.; Baenas, N.; Dominguez-Perles, R.; Barros, A.; Rosa, E.; Moreno, D.A.; Garcia-Viguera, C. Natural Bioactive Compounds from Winery By-Products as Health Promoters: A Review. Int. J. Mol. Sci. 2014, 15, 15638–15678. [Google Scholar] [CrossRef] [PubMed]
  23. Ferrer-Gallego, R.; Silva, P. The Wine Industry By-Products: Applications for Food Industry and Health Benefits. Antioxidants 2022, 11, 2025. [Google Scholar] [CrossRef]
  24. Nunes, M.A.; Rodrigues, F.; Oliveira, M.B.P.P. 11—Grape Processing By-Products as Active Ingredients for Cosmetic Proposes. In Handbook of Grape Processing By-Products; Galanakis, C.M., Ed.; Academic Press: Cambridge, MA, USA, 2017; pp. 267–292. [Google Scholar] [CrossRef]
  25. Silva, A.; Silva, V.; Igrejas, G.; Aires, A.; Falco, V.; Valentão, P.; Poeta, P. Phenolic compounds classification and their distribution in winemaking by-products. Eur. Food Res. Technol. 2023, 249, 207–239. [Google Scholar] [CrossRef]
  26. Constantin, O.E.; Stoica, F.; Rațu, R.N.; Stănciuc, N.; Bahrim, G.E.; Râpeanu, G. Bioactive Components, Applications, Extractions, and Health Benefits of Winery By-Products from a Circular Bioeconomy Perspective: A Review. Antioxidants 2024, 13, 100. [Google Scholar] [CrossRef]
  27. Ianni, A.; Di Maio, G.; Pittia, P.; Grotta, L.; Perpetuini, G.; Tofalo, R.; Cichelli, A.; Martino, G. Chemical–nutritional quality and oxidative stability of milk and dairy products obtained from Friesian cows fed with a dietary supplementation of dried grape pomace. J. Sci. Food Agric. 2019, 99, 3635–3643. [Google Scholar] [CrossRef] [PubMed]
  28. Troilo, M.; Difonzo, G.; Paradiso, V.M.; Summo, C.; Caponio, F. Bioactive Compounds from Vine Shoots, Grape Stalks, and Wine Lees: Their Potential Use in Agro-Food Chains. Foods 2021, 10, 342. [Google Scholar] [CrossRef]
  29. Ahmad, B.; Yadav, V.; Yadav, A.; Rahman, M.U.; Yuan, W.Z.; Li, Z.; Wang, X. Integrated biorefinery approach to valorize winery waste: A review from waste to energy perspectives. Sci. Total Environ. 2020, 719, 137315. [Google Scholar] [CrossRef]
  30. Bordiga, M.; Montella, R.; Travaglia, F.; Arlorio, M.; Coïsson, J.D. Characterization of polyphenolic and oligosaccharidic fractions extracted from grape seeds followed by the evaluation of prebiotic activity related to oligosaccharides. Int. J. Food Sci. Technol. 2019, 54, 1283–1291. [Google Scholar] [CrossRef]
  31. Gabur, G.-D.; Teodosiu, C.; Fighir, D.; Cotea, V.V.; Gabur, I. From Waste to Value in Circular Economy: Valorizing Grape Pomace Waste through Vermicomposting. Agriculture 2024, 14, 1529. [Google Scholar] [CrossRef]
  32. Vitalini, S.; Gardana, C.; Zanzotto, A.; Simonetti, P.; Faoro, F.; Fico, G.; Iriti, M. The presence of melatonin in grapevine (Vitis vinifera L.) berry tissues. J. Pineal Res. 2011, 51, 331–337. [Google Scholar] [CrossRef] [PubMed]
  33. Moreira, M.M.; Barroso, M.F.; Porto, J.V.; Ramalhosa, M.J.; Švarc-Gajić, J.; Estevinho, L.; Morais, S.; Delerue-Matos, C. Potential of Portuguese vine shoot wastes as natural resources of bioactive compounds. Sci. Total Environ. 2018, 634, 831–842. [Google Scholar] [CrossRef]
  34. Peña-Portillo, G.-C.; Acuña-Nelson, S.-M.; Bastías-Montes, J.-M. From Waste to Wealth: Exploring the Bioactive Potential of Wine By-Products—A Review. Antioxidants 2024, 13, 992. [Google Scholar] [CrossRef]
  35. Goufo, P.; Singh, R.K.; Cortez, I. A Reference List of Phenolic Compounds (Including Stilbenes) in Grapevine (Vitis vinifera L.) Roots, Woods, Canes, Stems, and Leaves. Antioxidants 2020, 9, 398. [Google Scholar] [CrossRef] [PubMed]
  36. Zwingelstein, M.; Draye, M.; Besombes, J.-L.; Piot, C.; Chatel, G. trans-Resveratrol and trans-ε-Viniferin in Grape Canes and Stocks Originating from Savoie Mont Blanc Vineyard Region: Pre-extraction Parameters for Improved Recovery. ACS Sustain. Chem. Eng. 2019, 7, 8310–8316. [Google Scholar] [CrossRef]
  37. Zagoskina, N.V.; Zubova, M.Y.; Nechaeva, T.L.; Kazantseva, V.V.; Goncharuk, E.A.; Katanskaya, V.M.; Baranova, E.N.; Aksenova, M.A. Polyphenols in Plants: Structure, Biosynthesis, Abiotic Stress Regulation, and Practical Applications (Review). Int. J. Mol. Sci. 2023, 24, 13874. [Google Scholar] [CrossRef]
  38. Quideau, S.; Deffieux, D.; Douat-Casassus, C.; Pouységu, L. Plant Polyphenols: Chemical Properties, Biological Activities, and Synthesis. Angew. Chem. Int. Ed. 2011, 50, 586–621. [Google Scholar] [CrossRef]
  39. Manach, C.; Scalbert, A.; Morand, C.; Rémésy, C.; Jiménez, L. Polyphenols: Food sources and bioavailability. Am. J. Clin. Nutr. 2004, 79, 727–747. [Google Scholar] [CrossRef]
  40. Bravo, L. Polyphenols: Chemistry, Dietary Sources, Metabolism, and Nutritional Significance. Nutr. Rev. 1998, 56, 317–333. [Google Scholar] [CrossRef]
  41. Zhang, Y.; Li, P.; Cheng, L. Developmental changes of carbohydrates, organic acids, amino acids, and phenolic compounds in ‘Honeycrisp’apple flesh. Food Chem. 2010, 123, 1013–1018. [Google Scholar] [CrossRef]
  42. Dai, J.; Mumper, R.J. Plant Phenolics: Extraction, Analysis and Their Antioxidant and Anticancer Properties. Molecules 2010, 15, 7313–7352. [Google Scholar] [CrossRef]
  43. Barry, T.N.; Forss, D.A. The condensed tannin content of vegetative Lotus pedunculatus, its regulation by fertiliser application, and effect upon protein solubility. J. Sci. Food Agric. 1983, 34, 1047–1056. [Google Scholar] [CrossRef]
  44. Sies, H. Polyphenols and health: Update and perspectives. Arch. Biochem. Biophys. 2010, 501, 2–5. [Google Scholar] [CrossRef]
  45. Kampa, M.; Nifli, A.-P.; Notas, G.; Castanas, E. Polyphenols and cancer cell growth. In Reviews of Physiology, Biochemistry and Pharmacology; Amara, S.G., Bamberg, E., Fleischmann, B., Gudermann, T., Hebert, S.C., Jahn, R., Lederer, W.J., Lill, R., Miyajima, A., Offermanns, S., et al., Eds.; Springer: Berlin/Heidelberg, Germany, 2007; pp. 79–113. [Google Scholar]
  46. Guo, W.; Kong, E.; Meydani, M. Dietary Polyphenols, Inflammation, and Cancer. Nutr. Cancer 2009, 61, 807–810. [Google Scholar] [CrossRef]
  47. Korkina, L.G.; De Luca, C.; Kostyuk, V.A.; Pastore, S. Plant Polyphenols and Tumors: From Mechanisms to Therapies, Prevention, and Protection Against Toxicity of Anti-Cancer Treatments. Curr. Med. Chem. 2009, 16, 3943–3965. [Google Scholar] [CrossRef]
  48. Zhao, B. Natural Antioxidants Protect Neurons in Alzheimer’s Disease and Parkinson’s Disease. Neurochem. Res. 2009, 34, 630–638. [Google Scholar] [CrossRef]
  49. Joseph, J.; Cole, G.; Head, E.; Ingram, D. Nutrition, Brain Aging, and Neurodegeneration. J. Neurosci. 2009, 29, 12795–12801. [Google Scholar] [CrossRef] [PubMed]
  50. Gutierrez-Merino, C.; Lopez-Sanchez, C.; Lagoa, R.; Samhan-Arias, A.K.; Bueno, C.; Garcia-Martinez, V. Neuroprotective Actions of Flavonoids. Curr. Med. Chem. 2011, 18, 1195–1212. [Google Scholar] [CrossRef]
  51. Michalska, M.; Gluba, A.; Mikhailidis, D.P.; Nowak, P.; Bielecka-Dabrowa, A.; Rysz, J.; Banach, M. The role of polyphenols in cardiovascular disease. Med. Sci. Monit. 2010, 16, RA110–RA119. [Google Scholar]
  52. Grassi, D.; Desideri, G.; Croce, G.; Tiberti, S.; Aggio, A.; Ferri, C. Flavonoids, Vascular Function and Cardiovascular Protection. Curr. Pharm. Des. 2009, 15, 1072–1084. [Google Scholar] [CrossRef]
  53. Milne, J.C.; Lambert, P.D.; Schenk, S.; Carney, D.P.; Smith, J.J.; Gagne, D.J.; Jin, L.; Boss, O.; Perni, R.B.; Vu, C.B.; et al. Small molecule activators of SIRT1 as therapeutics for the treatment of type 2 diabetes. Nature 2007, 450, 712–716. [Google Scholar] [CrossRef] [PubMed]
  54. Zunino, S.J. Type 2 Diabetes and Glycemic Response to Grapes or Grape Products1,2. J. Nutr. 2009, 139, 1794S–1800S. [Google Scholar] [CrossRef] [PubMed]
  55. Borriello, A.; Cucciolla, V.; Della Ragione, F.; Galletti, P. Dietary polyphenols: Focus on resveratrol, a promising agent in the prevention of cardiovascular diseases and control of glucose homeostasis. Nutr. Metab. Cardiovasc. Dis. 2010, 20, 618–625. [Google Scholar] [CrossRef]
  56. Ferrari, C.K.B. Functional foods, herbs and nutraceuticals: Towards biochemical mechanisms of healthy aging. Biogerontology 2004, 5, 275–289. [Google Scholar] [CrossRef]
  57. Pearson, K.J.; Baur, J.A.; Lewis, K.N.; Peshkin, L.; Price, N.L.; Labinskyy, N.; Swindell, W.R.; Kamara, D.; Minor, R.K.; Perez, E.; et al. Resveratrol Delays Age-Related Deterioration and Mimics Transcriptional Aspects of Dietary Restriction without Extending Life Span. Cell Metab. 2008, 8, 157–168. [Google Scholar] [CrossRef]
  58. Queen, B.L.; Tollefsbol, T.O. Polyphenols and Aging. Curr. Aging Sci. 2010, 3, 34–42. [Google Scholar] [CrossRef]
  59. Agouni, A.; Lagrue-Lak-Hal, A.-H.; Mostefai, H.A.; Tesse, A.; Mulder, P.; Rouet, P.; Desmoulin, F.; Heymes, C.; Martínez, M.C.; Andriantsitohaina, R. Red Wine Polyphenols Prevent Metabolic and Cardiovascular Alterations Associated with Obesity in Zucker Fatty Rats (Fa/Fa). PLoS ONE 2009, 4, e5557. [Google Scholar] [CrossRef] [PubMed]
  60. Cherniack, E.P. Polyphenols: Planting the seeds of treatment for the metabolic syndrome. Nutrition 2011, 27, 617–623. [Google Scholar] [CrossRef] [PubMed]
  61. Rahman, I.; Chung, S. Dietary polyphenols, deacetylases and chromatin remodeling in inflammation. Pers. Nutr. 2010, 101, 84–94. [Google Scholar]
  62. Kostyuk, V.; Potapovich, A.; De Luca, C. The Promise of Plant Polyphenols as the Golden Standard Skin Anti-Inflammatory Agents. Curr. Drug Metab. 2010, 11, 415–424. [Google Scholar] [CrossRef] [PubMed]
  63. Acquaviva, R.; Iauk, L. Natural polyphenols as anti-inflammatory agents. J. Front. Biosci. 2010, 2, 318–331. [Google Scholar]
  64. Sáez, V.; Gayoso, C.; Riquelme, S.; Pérez, J.; Vergara, C.; Mardones, C.; von Baer, D. C18 core-shell column with in-series absorbance and fluorescence detection for simultaneous monitoring of changes in stilbenoid and proanthocyanidin concentrations during grape cane storage. J. Chromatogr. B 2018, 1074–1075, 70–78. [Google Scholar] [CrossRef]
  65. Waffo-Teguo, P.; Krisa, S.; Richard, T.; Mérillon, J.-M. Grapevine Stilbenes and Their Biological Effects. In Bioactive Molecules and Medicinal Plants; Ramawat, K.G., Merillon, J.M., Eds.; Springer: Berlin/Heidelberg, Germany, 2008; pp. 25–54. [Google Scholar]
  66. Mattivi, F.; Vrhovsek, U.; Malacarne, G.; Masuero, D.; Zulini, L.; Stefanini, M.; Moser, C.; Velasco, R.; Guella, G. Profiling of Resveratrol Oligomers, Important Stress Metabolites, Accumulating in the Leaves of Hybrid Vitis vinifera (Merzling × Teroldego) Genotypes Infected with Plasmopara viticola. J. Agric. Food Chem. 2011, 59, 5364–5375. [Google Scholar] [CrossRef]
  67. Jiang, Y.L.; Liu, Z.P. Natural Products as Anti-Invasive and Anti-Metastatic Agents. Curr. Med. Chem. 2011, 18, 808–829. [Google Scholar] [CrossRef]
  68. Park, E.-J.; Pezzuto, J.M. The pharmacology of resveratrol in animals and humans. Biochim. Biophys. Acta (BBA)—Mol. Basis Dis. 2015, 1852, 1071–1113. [Google Scholar] [CrossRef]
  69. Seyed, M.A.; Jantan, I.; Bukhari, S.N.A.; Vijayaraghavan, K. A Comprehensive Review on the Chemotherapeutic Potential of Piceatannol for Cancer Treatment, with Mechanistic Insights. J. Agric. Food Chem. 2016, 64, 725–737. [Google Scholar] [CrossRef] [PubMed]
  70. Billet, K.; Houillé, B.; Besseau, S.; Mélin, C.; Oudin, A.; Papon, N.; Courdavault, V.; Clastre, M.; Giglioli-Guivarc’h, N.; Lanoue, A. Mechanical stress rapidly induces E-resveratrol and E-piceatannol biosynthesis in grape canes stored as a freshly-pruned byproduct. Food Chem. 2018, 240, 1022–1027. [Google Scholar] [CrossRef] [PubMed]
  71. Guerrero, R.F.; Biais, B.; Richard, T.; Puertas, B.; Waffo-Teguo, P.; Merillon, J.-M.; Cantos-Villar, E. Grapevine cane’s waste is a source of bioactive stilbenes. Ind. Crops Prod. 2016, 94, 884–892. [Google Scholar] [CrossRef]
  72. Navarro-Orcajada, S.; Conesa, I.; Vidal-Sánchez, F.J.; Matencio, A.; Albaladejo-Maricó, L.; García-Carmona, F.; López-Nicolás, J.M. Stilbenes: Characterization, bioactivity, encapsulation and structural modifications. A review of their current limitations and promising approaches. Crit. Rev. Food Sci. Nutr. 2023, 63, 7269–7287. [Google Scholar] [CrossRef]
  73. Chong, J.; Poutaraud, A.; Hugueney, P. Metabolism and roles of stilbenes in plants. Plant Sci. 2009, 177, 143–155. [Google Scholar] [CrossRef]
  74. Bábíková, P.; Vrchotová, N.; Tříska, J.; Kyseláková, M. Content of trans-resveratrol in leaves and berries of interspecific grapevine (Vitis sp.) varieties. Czech J. Food Sci. 2008, 26, S13–S17. [Google Scholar] [CrossRef]
  75. Rusjan, D.; Halbwirth, H.; Stich, K.; Mikulič-Petkovšek, M.; Veberič, R. Biochemical response of grapevine variety ‘Chardonnay’ (Vitis vinifera L.) to infection with grapevine yellows (Bois noir). Eur. J. Plant Pathol. 2012, 134, 231–237. [Google Scholar] [CrossRef]
  76. Parage, C.; Tavares, R.; Réty, S.; Baltenweck-Guyot, R.; Poutaraud, A.; Renault, L.; Heintz, D.; Lugan, R.; Marais, G.A.B.; Aubourg, S.; et al. Structural, Functional, and Evolutionary Analysis of the Unusually Large Stilbene Synthase Gene Family in Grapevine. Plant Physiol. 2012, 160, 1407–1419. [Google Scholar] [CrossRef]
  77. Çetin, E.S.; Altinöz, D.; Tarçan, E.; Göktürk Baydar, N. Chemical composition of grape canes. Ind. Crops Prod. 2011, 34, 994–998. [Google Scholar] [CrossRef]
  78. Ewald, P.; Delker, U.; Winterhalter, P. Quantification of stilbenoids in grapevine canes and grape cluster stems with a focus on long-term storage effects on stilbenoid concentration in grapevine canes. Food Res. Int. 2017, 100, 326–331. [Google Scholar] [CrossRef]
  79. Cebrián, C.; Sánchez-Gómez, R.; Salinas, M.R.; Alonso, G.L.; Zalacain, A. Effect of post-pruning vine-shoots storage on the evolution of high-value compounds. Ind. Crops Prod. 2017, 109, 730–736. [Google Scholar] [CrossRef]
  80. Ben Mohamed, H.; Vadel, A.M.; Geuns, J.M.C.; Khemira, H. Biochemical changes in dormant grapevine shoot tissues in response to chilling: Possible role in dormancy release. Sci. Hortic. 2010, 124, 440–447. [Google Scholar] [CrossRef]
  81. Aaviksaar, A.; Haga, M.; Pussa, T.; Roasto, M.; Tsoupras, G. Purification of resveratrol from vine stems. Proc.-Est. Acad. Sci. Chem. 2003, 52, 155–164. [Google Scholar] [CrossRef]
  82. Püssa, T.; Floren, J.; Kuldkepp, P.; Raal, A. Survey of Grapevine Vitis vinifera Stem Polyphenols by Liquid Chromatography−Diode Array Detection−Tandem Mass Spectrometry. J. Agric. Food Chem. 2006, 54, 7488–7494. [Google Scholar] [CrossRef]
  83. Cruz, S.; Raposo, R.; Ruiz-Moreno, M.J.; Garde-Cerdán, T.; Puertas, B.; Gonzalo-Diago, A.; Moreno-Rojas, J.M.; Cantos-Villar, E. Grapevine-shoot stilbene extract as a preservative in white wine. Food Packag. Shelf Life 2018, 18, 164–172. [Google Scholar] [CrossRef]
  84. Cebrián-Tarancón, C.; Sánchez-Gómez, R.; Salinas, M.R.; Alonso, G.L.; Oliva, J.; Zalacain, A. Toasted vine-shoot chips as enological additive. Food Chem. 2018, 263, 96–103. [Google Scholar] [CrossRef]
  85. Rätsep, R.; Karp, K.; Maante-Kuljus, M.; Aluvee, A.; Kaldmäe, H.; Bhat, R. Recovery of Polyphenols from Vineyard Pruning Wastes—Shoots and Cane of Hybrid Grapevine (Vitis sp.) Cultivars. Antioxidants 2021, 10, 1059. [Google Scholar] [CrossRef]
  86. Pietarinen, S.P.; Willför, S.M.; Ahotupa, M.O.; Hemming, J.E.; Holmbom, B.R. Knotwood and bark extracts: Strong antioxidants from waste materials. J. Wood Sci. 2006, 52, 436–444. [Google Scholar] [CrossRef]
  87. García-Pérez, M.-E.; Royer, M.; Herbette, G.; Desjardins, Y.; Pouliot, R.; Stevanovic, T. Picea mariana bark: A new source of trans-resveratrol and other bioactive polyphenols. Food Chem. 2012, 135, 1173–1182. [Google Scholar] [CrossRef]
  88. Rajbhar, K.; Dawda, H.; Mukundan, U. Polyphenols: Methods of extraction. Sci. Revs. Chem. Commun. 2015, 5, 1–6. [Google Scholar]
  89. Zwingelstein, M.; Draye, M.; Besombes, J.-L.; Piot, C.; Chatel, G. Viticultural wood waste as a source of polyphenols of interest: Opportunities and perspectives through conventional and emerging extraction methods. Waste Manag. 2020, 102, 782–794. [Google Scholar] [CrossRef]
  90. Angelov, G.; Boyadzhieva, S.; Georgieva, S. Rosehip extraction: Process optimization and antioxidant capacity of extracts. Open Chem. 2014, 12, 502–508. [Google Scholar] [CrossRef]
  91. Angelov, G.; Georgieva, S.; Boyadzhieva, S.; Boyadzhiev, L. Optimizing the extraction of globe artichoke wastes. Comptes Rendus De. L’Academie Bulg. Des. Sci. 2015, 68, 1235–1240. [Google Scholar]
  92. Kumar, B.; Bhardwaj, N.; Agrawal, K.; Chaturvedi, V.; Verma, P. Current perspective on pretreatment technologies using lignocellulosic biomass: An emerging biorefinery concept. Fuel Process. Technol. 2020, 199, 106244. [Google Scholar] [CrossRef]
  93. Carvalho, T.M.J. Extraction of Raw Plant Material Using Supercritical Carbon Dioxide. Master’s Thesis, Warsaw University of Technology, Warsaw, Poland, 2016. [Google Scholar]
  94. Mahindrakar, K.V.; Rathod, V.K. Chapter 5—Ultrasound-assisted extraction of lipids, carotenoids, and other compounds from marine resources. In Innovative and Emerging Technologies in the Bio-Marine Food Sector; Garcia-Vaquero, M., Rajauria, G., Eds.; Academic Press: Cambridge, MA, USA, 2022; pp. 81–128. [Google Scholar]
  95. Šimat, V.; Frleta, R.; Čagalj, M.; Skroza, D. Technological and Analytical Aspects of Bioactive Compounds and Nutraceuticals from Marine Algae. In Bioactive Compounds and Nutraceuticals from Dairy, Marine, and Nonconventional Sources; Apple Academic Press: Palm Bay, FL, USA, 2024; pp. 159–196. [Google Scholar]
  96. Maddaloni, M.; Vassalini, I.; Alessandri, I. Green Routes for the Development of Chitin/Chitosan Sustainable Hydrogels. Sustain. Chem. 2020, 1, 325–344. [Google Scholar] [CrossRef]
  97. Schnee, S.; Queiroz, E.F.; Voinesco, F.; Marcourt, L.; Dubuis, P.-H.; Wolfender, J.-L.; Gindro, K. Vitis vinifera Canes, a New Source of Antifungal Compounds against Plasmopara viticola, Erysiphe necator, and Botrytis cinerea. J. Agric. Food Chem. 2013, 61, 5459–5467. [Google Scholar] [CrossRef]
  98. Gabaston, J.; Leborgne, C.; Waffo-Teguo, P.; Valls, J.; Palos Pinto, A.; Richard, T.; Cluzet, S.; Mérillon, J.-M. Wood and roots of major grapevine cultivars and rootstocks: A comparative analysis of stilbenes by UHPLC-DAD-MS/MS and NMR. Phytochem. Anal. 2019, 30, 320–331. [Google Scholar] [CrossRef]
  99. Bavaresco, L.; Fregoni, C. Physiological Role and Molecular Aspects of Grapevine Stilbenic Compounds. In Molecular Biology & Biotechnology of the Grapevine; Roubelakis-Angelakis, K.A., Ed.; Springer: Dordrecht, The Netherlands, 2001; pp. 153–182. [Google Scholar]
  100. El Khawand, T.; Courtois, A.; Valls, J.; Richard, T.; Krisa, S. A review of dietary stilbenes: Sources and bioavailability. Phytochem. Rev. 2018, 17, 1007–1029. [Google Scholar] [CrossRef]
  101. Kapetanovic, I.M.; Muzzio, M.; Huang, Z.; Thompson, T.N.; McCormick, D.L. Pharmacokinetics, oral bioavailability, and metabolic profile of resveratrol and its dimethylether analog, pterostilbene, in rats. Cancer Chemother. Pharmacol. 2011, 68, 593–601. [Google Scholar] [CrossRef]
  102. Marier, J.-F.; Vachon, P.; Gritsas, A.; Zhang, J.; Moreau, J.-P.; Ducharme, M.P. Metabolism and Disposition of Resveratrol in Rats: Extent of Absorption, Glucuronidation, and Enterohepatic Recirculation Evidenced by a Linked-Rat Model. J. Pharmacol. Exp. Ther. 2002, 302, 369–373. [Google Scholar] [CrossRef]
  103. Walle, T. Bioavailability of resveratrol. Ann. N. Y. Acad. Sci. 2011, 1215, 9–15. [Google Scholar] [CrossRef] [PubMed]
  104. Das, S.; Lin, H.-S.; Ho, P.C.; Ng, K.-Y. The Impact of Aqueous Solubility and Dose on the Pharmacokinetic Profiles of Resveratrol. Pharm. Res. 2008, 25, 2593–2600. [Google Scholar] [CrossRef] [PubMed]
  105. Teplova, V.V.; Isakova, E.P.; Klein, O.I.; Dergachova, D.I.; Gessler, N.N.; Deryabina, Y.I. Natural Polyphenols: Biological Activity, Pharmacological Potential, Means of Metabolic Engineering (Review). Appl. Biochem. Microbiol. 2018, 54, 221–237. [Google Scholar] [CrossRef]
  106. Schouten, A.; Wagemakers, L.; Stefanato, F.L.; Kaaij, R.M.v.d.; Kan, J.A.L.v. Resveratrol acts as a natural profungicide and induces self-intoxication by a specific laccase. Mol. Microbiol. 2002, 43, 883–894. [Google Scholar] [CrossRef]
  107. Seppänen, S.K.; Syrjälä, L.; von Weissenberg, K.; Teeri, T.H.; Paajanen, L.; Pappinen, A. Antifungal activity of stilbenes in in vitro bioassays and in transgenic Populus expressing a gene encoding pinosylvin synthase. Plant Cell Rep. 2004, 22, 584–593. [Google Scholar] [CrossRef]
  108. Liu, Q.; Yeo, W.S.; Bae, T. The SaeRS Two-Component System of Staphylococcus aureus. Genes. 2016, 7, 81. [Google Scholar] [CrossRef]
  109. Duan, J.; Li, M.; Hao, Z.; Shen, X.; Liu, L.; Jin, Y.; Wang, S.; Guo, Y.; Yang, L.; Wang, L.; et al. Subinhibitory concentrations of resveratrol reduce alpha-hemolysin production in Staphylococcus aureus isolates by downregulating saeRS. Emerg. Microbes Infect. 2018, 7, 1–10. [Google Scholar] [CrossRef]
  110. Flemming, H.-C.; Wuertz, S. Bacteria and archaea on Earth and their abundance in biofilms. Nat. Rev. Microbiol. 2019, 17, 247–260. [Google Scholar] [CrossRef]
  111. Kattke, M.D.; Gosschalk, J.E.; Martinez, O.E.; Kumar, G.; Gale, R.T.; Cascio, D.; Sawaya, M.R.; Philips, M.; Brown, E.D.; Clubb, R.T. Structure and mechanism of TagA, a novel membrane-associated glycosyltransferase that produces wall teichoic acids in pathogenic bacteria. PLoS Pathog. 2019, 15, e1007723. [Google Scholar] [CrossRef]
  112. Rabin, N.; Zheng, Y.; Opoku-Temeng, C.; Du, Y.; Bonsu, E.; Sintim, H.O. Biofilm Formation Mechanisms and Targets for Developing Antibiofilm Agents. Future Med. Chem. 2015, 7, 493–512. [Google Scholar] [CrossRef]
  113. Ramadan, H.H. Chronic rhinosinusitis and bacterial biofilms. Curr. Opin. Otolaryngol. Head. Neck Surg. 2006, 14, 183–186. [Google Scholar] [CrossRef] [PubMed]
  114. Hall-Stoodley, L.; Costerton, J.W.; Stoodley, P. Bacterial biofilms: From the Natural environment to infectious diseases. Nat. Rev. Microbiol. 2004, 2, 95–108. [Google Scholar] [CrossRef] [PubMed]
  115. Wang, W.-B.; Lai, H.-C.; Hsueh, P.-R.; Chiou, R.Y.-Y.; Lin, S.-B.; Liaw, S.-J. Inhibition of swarming and virulence factor expression in Proteus mirabilis by resveratrol. J. Med. Microbiol. 2006, 55, 1313–1321. [Google Scholar] [CrossRef]
  116. Fernández-Alvarez, A.; Llorente-Izquierdo, C.; Mayoral, R.; Agra, N.; Boscá, L.; Casado, M.; Martín-Sanz, P. Evaluation of epigenetic modulation of cyclooxygenase-2 as a prognostic marker for hepatocellular carcinoma. Oncogenesis 2012, 1, e23. [Google Scholar] [CrossRef]
  117. Akinwumi, B.C.; Bordun, K.-A.M.; Anderson, H.D. Biological Activities of Stilbenoids. Int. J. Mol. Sci. 2018, 19, 792. [Google Scholar] [CrossRef] [PubMed]
  118. Remsberg, C.M.; Yáñez, J.A.; Ohgami, Y.; Vega-Villa, K.R.; Rimando, A.M.; Davies, N.M. Pharmacometrics of pterostilbene: Preclinical pharmacokinetics and metabolism, anticancer, antiinflammatory, antioxidant and analgesic activity. Phytother. Res. 2008, 22, 169–179. [Google Scholar] [CrossRef]
  119. Kim, J.; Min, J.S.; Kim, D.; Zheng, Y.F.; Mailar, K.; Choi, W.J.; Lee, C.; Bae, S.K. A simple and sensitive liquid chromatography–tandem mass spectrometry method for trans-ε-viniferin quantification in mouse plasma and its application to a pharmacokinetic study in mice. J. Pharm. Biomed. Anal. 2017, 134, 116–121. [Google Scholar] [CrossRef]
  120. Santucci, C.; Mignozzi, S.; Levi, F.; Malvezzi, M.; Boffetta, P.; Negri, E.; La Vecchia, C. European cancer mortality predictions for the year 2025 with focus on breast cancer. Ann. Oncol. 2025, 36, 460–468. [Google Scholar] [CrossRef]
  121. Pereira, M.; Peleteiro, B.; Capewell, S.; Bennett, K.; Azevedo, A.; Lunet, N. Changing patterns of cardiovascular diseases and cancer mortality in Portugal, 1980–2010. BMC Public Health 2012, 12, 1126. [Google Scholar] [CrossRef] [PubMed]
  122. Siegel, R.; Naishadham, D.; Jemal, A. Cancer statistics for Hispanics/Latinos, 2012. CA Cancer J. Clin. 2012, 62, 283–298. [Google Scholar] [CrossRef] [PubMed]
  123. Kang, N.J.; Shin, S.H.; Lee, H.J.; Lee, K.W. Polyphenols as small molecular inhibitors of signaling cascades in carcinogenesis. Pharmacol. Ther. 2011, 130, 310–324. [Google Scholar] [CrossRef]
  124. Mena, S.; Ortega, A.; Estrela, J.M. Oxidative stress in environmental-induced carcinogenesis. Mutat. Res./Genet. Toxicol. Environ. Mutagen. 2009, 674, 36–44. [Google Scholar] [CrossRef] [PubMed]
  125. Jang, M.; Cai, L.; Udeani, G.O.; Slowing, K.V.; Thomas, C.F.; Beecher, C.W.W.; Fong, H.H.S.; Farnsworth, N.R.; Kinghorn, A.D.; Mehta, R.G.; et al. Cancer Chemopreventive Activity of Resveratrol, a Natural Product Derived from Grapes. Science 1997, 275, 218–220. [Google Scholar] [CrossRef]
  126. Chatterjee, A.; Ronghe, A.; Singh, B.; Bhat, N.K.; Chen, J.; Bhat, H.K. Natural Antioxidants Exhibit Chemopreventive Characteristics through the Regulation of CNC b-Zip Transcription Factors in Estrogen-Induced Breast Carcinogenesis. J. Biochem. Mol. Toxicol. 2014, 28, 529–538. [Google Scholar] [CrossRef] [PubMed]
  127. Sirerol, J.A.; Feddi, F.; Mena, S.; Rodriguez, M.L.; Sirera, P.; Aupí, M.; Pérez, S.; Asensi, M.; Ortega, A.; Estrela, J.M. Topical treatment with pterostilbene, a natural phytoalexin, effectively protects hairless mice against UVB radiation-induced skin damage and carcinogenesis. Free Radic. Biol. Med. 2015, 85, 1–11. [Google Scholar] [CrossRef]
  128. Alfaras, I.; Juan, M.E.; Planas, J.M. trans-Resveratrol Reduces Precancerous Colonic Lesions in Dimethylhydrazine-Treated Rats. J. Agric. Food Chem. 2010, 58, 8104–8110. [Google Scholar] [CrossRef]
  129. Azorín-Ortuño, M.; Yáñez-Gascón, M.J.; Vallejo, F.; Pallarés, F.J.; Larrosa, M.; Lucas, R.; Morales, J.C.; Tomás-Barberán, F.A.; García-Conesa, M.T.; Espín, J.C. Metabolites and tissue distribution of resveratrol in the pig. Mol. Nutr. Food Res. 2011, 55, 1154–1168. [Google Scholar] [CrossRef]
  130. Roubille, C.; Martel-Pelletier, J.; Davy, J.-M.; Haraoui, B.; Pelletier, J.-P. Cardiovascular adverse effects of anti-inflammatory drugs. Anti-Inflamm. Anti-Allergy Agents Med. Chem. 2013, 12, 55–67. [Google Scholar] [CrossRef]
  131. Levy, G.N. Prostaglandin H synthases, nonsteroidal antiinflammatory drugs, and colon cancer. FASEB J. 1997, 11, 234–247. [Google Scholar] [CrossRef]
  132. Kitasato, A.; Kuroki, T.; Adachi, T.; Ono, S.; Tanaka, T.; Tsuneoka, N.; Hirabaru, M.; Takatsuki, M.; Eguchi, S. A Selective Cyclooxygenase-2 Inhibitor (Etodolac) Prevents Spontaneous Biliary Tumorigenesis in a Hamster Bilioenterostomy Model. Eur. Surg. Res. 2014, 52, 73–82. [Google Scholar] [CrossRef]
  133. Cianciulli, A.; Calvello, R.; Cavallo, P.; Dragone, T.; Carofiglio, V.; Panaro, M.A. Modulation of NF-κB activation by resveratrol in LPS treated human intestinal cells results in downregulation of PGE2 production and COX-2 expression. Toxicol. Vitr. 2012, 26, 1122–1128. [Google Scholar] [CrossRef]
  134. Schwartz, S.A.; Hernandez, A.; Mark Evers, B. The role of NF-κB/IκB proteins in cancer: Implications for novel treatment strategies. Surg. Oncol. 1999, 8, 143–153. [Google Scholar] [CrossRef]
  135. Potter, G.A.; Patterson, L.H.; Wanogho, E.; Perry, P.J.; Butler, P.C.; Ijaz, T.; Ruparelia, K.C.; Lamb, J.H.; Farmer, P.B.; Stanley, L.A.; et al. The cancer preventative agent resveratrol is converted to the anticancer agent piceatannol by the cytochrome P450 enzyme CYP1B1. Br. J. Cancer 2002, 86, 774–778. [Google Scholar] [CrossRef] [PubMed]
  136. Hart, J.H. Role of Phytostilbenes in Decay and Disease Resistance. Annu. Rev. Phytopathol. 1981, 19, 437–458. [Google Scholar] [CrossRef]
  137. Adrian, M.; Jeandet, P. Effects of resveratrol on the ultrastructure of Botrytis cinerea conidia and biological significance in plant/pathogen interactions. Fitoterapia 2012, 83, 1345–1350. [Google Scholar] [CrossRef]
  138. Collado-González, M.; Guirao-Abad, J.P.; Sánchez-Fresneda, R.; Belchí-Navarro, S.; Argüelles, J.-C. Resveratrol lacks antifungal activity against Candida albicans. World J. Microbiol. Biotechnol. 2012, 28, 2441–2446. [Google Scholar] [CrossRef] [PubMed]
  139. Jeandet, P.; Douillet-Breuil, A.-C.; Bessis, R.; Debord, S.; Sbaghi, M.; Adrian, M. Phytoalexins from the Vitaceae: Biosynthesis, Phytoalexin Gene Expression in Transgenic Plants, Antifungal Activity, and Metabolism. J. Agric. Food Chem. 2002, 50, 2731–2741. [Google Scholar] [CrossRef] [PubMed]
  140. Lee, K.; Lee, J.-H.; Ryu, S.Y.; Cho, M.H.; Lee, J. Stilbenes Reduce Staphylococcus aureus Hemolysis, Biofilm Formation, and Virulence. Foodborne Pathog. Dis. 2014, 11, 710–717. [Google Scholar] [CrossRef]
  141. Beaumont, P.; Courtois, A.; Atgié, C.; Richard, T.; Krisa, S. In the shadow of resveratrol: Biological activities of epsilon-viniferin. J. Physiol. Biochem. 2022, 78, 465–484. [Google Scholar] [CrossRef]
  142. Cho, H.S.; Lee, J.-H.; Ryu, S.Y.; Joo, S.W.; Cho, M.H.; Lee, J. Inhibition of Pseudomonas aeruginosa and Escherichia coli O157:H7 Biofilm Formation by Plant Metabolite ε-Viniferin. J. Agric. Food Chem. 2013, 61, 7120–7126. [Google Scholar] [CrossRef]
  143. Frazzi, R.; Valli, R.; Tamagnini, I.; Casali, B.; Latruffe, N.; Merli, F. Resveratrol-mediated apoptosis of hodgkin lymphoma cells involves SIRT1 inhibition and FOXO3a hyperacetylation. Int. J. Cancer 2013, 132, 1013–1021. [Google Scholar] [CrossRef]
  144. Shankar, S.; Chen, Q.; Siddiqui, I.; Sarva, K.; Srivastava, R.K. Sensitization of TRAIL-resistant LNCaP cells by resveratrol (3, 4′, 5 tri-hydroxystilbene): Molecular mechanisms and therapeutic potential. J. Mol. Signal. 2007, 2, 7. [Google Scholar] [CrossRef]
  145. van Ginkel, P.R.; Yan, M.B.; Bhattacharya, S.; Polans, A.S.; Kenealey, J.D. Natural products induce a G protein-mediated calcium pathway activating p53 in cancer cells. Toxicol. Appl. Pharmacol. 2015, 288, 453–462. [Google Scholar] [CrossRef] [PubMed]
  146. Lin, H.-S.; Yue, B.-D.; Ho, P.C. Determination of pterostilbene in rat plasma by a simple HPLC-UV method and its application in pre-clinical pharmacokinetic study. Biomed. Chromatogr. 2009, 23, 1308–1315. [Google Scholar] [CrossRef] [PubMed]
  147. Singh, N.; Nigam, M.; Ranjan, V.; Sharma, R.; Balapure, A.K.; Rath, S.K. Caspase Mediated Enhanced Apoptotic Action of Cyclophosphamide- and Resveratrol-Treated MCF-7 Cells. J. Pharmacol. Sci. 2009, 109, 473–485. [Google Scholar] [CrossRef] [PubMed]
  148. Kai, L.; Samuel, S.K.; Levenson, A.S. Resveratrol enhances p53 acetylation and apoptosis in prostate cancer by inhibiting MTA1/NuRD complex. Int. J. Cancer 2010, 126, 1538–1548. [Google Scholar] [CrossRef]
  149. Rimando, A.M.; Cuendet, M.; Desmarchelier, C.; Mehta, R.G.; Pezzuto, J.M.; Duke, S.O. Cancer Chemopreventive and Antioxidant Activities of Pterostilbene, a Naturally Occurring Analogue of Resveratrol. J. Agric. Food Chem. 2002, 50, 3453–3457. [Google Scholar] [CrossRef]
  150. Stivala, L.A.; Savio, M.; Carafoli, F.; Perucca, P.; Bianchi, L.; Maga, G.; Forti, L.; Pagnoni, U.M.; Albini, A.; Prosperi, E.; et al. Specific Structural Determinants Are Responsible for the Antioxidant Activity and the Cell Cycle Effects of Resveratrol. J. Biol. Chem. 2001, 276, 22586–22594. [Google Scholar] [CrossRef]
  151. McCormack, D.; McFadden, D. Pterostilbene and Cancer: Current Review. J. Surg. Res. 2012, 173, e53–e61. [Google Scholar] [CrossRef]
  152. McCormack, D.E.; Mannal, P.; McDonald, D.; Tighe, S.; Hanson, J.; McFadden, D. Genomic Analysis of Pterostilbene Predicts Its Antiproliferative Effects Against Pancreatic Cancer In Vitro and In Vivo. J. Gastrointest. Surg. 2012, 16, 1136–1143. [Google Scholar] [CrossRef] [PubMed]
  153. Ferrer, P.; Asensi, M.; Segarra, R.; Ortega, A.; Benlloch, M.; Obrador, E.; Varea, M.T.; Asensio, G.; Jordá, L.; Estrela, J.M. Association between Pterostilbene and Quercetin Inhibits Metastatic Activity of B16 Melanoma. Neoplasia 2005, 7, 37–47. [Google Scholar] [CrossRef]
  154. Tolomeo, M.; Grimaudo, S.; Cristina, A.D.; Roberti, M.; Pizzirani, D.; Meli, M.; Dusonchet, L.; Gebbia, N.; Abbadessa, V.; Crosta, L.; et al. Pterostilbene and 3′-hydroxypterostilbene are effective apoptosis-inducing agents in MDR and BCR-ABL-expressing leukemia cells. Int. J. Biochem. Cell Biol. 2005, 37, 1709–1726. [Google Scholar] [CrossRef]
  155. Alosi, J.A.; McDonald, D.E.; Schneider, J.S.; Privette, A.R.; McFadden, D.W. Pterostilbene Inhibits Breast Cancer In Vitro Through Mitochondrial Depolarization and Induction of Caspase-Dependent Apoptosis1. J. Surg. Res. 2010, 161, 195–201. [Google Scholar] [CrossRef]
  156. Wang, Y.; Ding, L.; Wang, X.; Zhang, J.; Han, W.; Feng, L.; Sun, J.; Jin, H.; Wang, X.J. Pterostilbene simultaneously induces apoptosis, cell cycle arrest and cyto-protective autophagy in breast cancer cells. Am. J. Transl. Res. 2012, 4, 44. [Google Scholar]
  157. Moon, D.; McCormack, D.; McDonald, D.; McFadden, D. Pterostilbene induces mitochondrially derived apoptosis in breast cancer cells in vitro. J. Surg. Res. 2013, 180, 208–215. [Google Scholar] [CrossRef] [PubMed]
  158. Chen, R.-J.; Tsai, S.-J.; Ho, C.-T.; Pan, M.-H.; Ho, Y.-S.; Wu, C.-H.; Wang, Y.-J. Chemopreventive Effects of Pterostilbene on Urethane-Induced Lung Carcinogenesis in Mice via the Inhibition of EGFR-Mediated Pathways and the Induction of Apoptosis and Autophagy. J. Agric. Food Chem. 2012, 60, 11533–11541. [Google Scholar] [CrossRef] [PubMed]
  159. Chakraborty, A.; Gupta, N.; Ghosh, K.; Roy, P. In vitro evaluation of the cytotoxic, anti-proliferative and anti-oxidant properties of pterostilbene isolated from Pterocarpus marsupium. Toxicol. Vitr. 2010, 24, 1215–1228. [Google Scholar] [CrossRef]
  160. Chiou, Y.-S.; Tsai, M.-L.; Wang, Y.-J.; Cheng, A.-C.; Lai, W.-M.; Badmaev, V.; Ho, C.-T.; Pan, M.-H. Pterostilbene Inhibits Colorectal Aberrant Crypt Foci (ACF) and Colon Carcinogenesis via Suppression of Multiple Signal Transduction Pathways in Azoxymethane-Treated Mice. J. Agric. Food Chem. 2010, 58, 8833–8841. [Google Scholar] [CrossRef] [PubMed]
  161. Paul, S.; Rimando, A.M.; Lee, H.J.; Ji, Y.; Reddy, B.S.; Suh, N. Anti-inflammatory Action of Pterostilbene Is Mediated through the p38 Mitogen-Activated Protein Kinase Pathway in Colon Cancer Cells. Cancer Prev. Res. 2009, 2, 650–657. [Google Scholar] [CrossRef]
  162. Hardie, D.G.; Ross, F.A.; Hawley, S.A. AMPK: A nutrient and energy sensor that maintains energy homeostasis. Nat. Rev. Mol. Cell Biol. 2012, 13, 251–262. [Google Scholar] [CrossRef]
  163. Reinisalo, M.; Kårlund, A.; Koskela, A.; Kaarniranta, K.; Karjalainen, R.O. Polyphenol Stilbenes: Molecular Mechanisms of Defence against Oxidative Stress and Aging-Related Diseases. Oxidative Med. Cell. Longev. 2015, 2015, 340520. [Google Scholar] [CrossRef]
  164. Cantó, C.; Gerhart-Hines, Z.; Feige, J.N.; Lagouge, M.; Noriega, L.; Milne, J.C.; Elliott, P.J.; Puigserver, P.; Auwerx, J. AMPK regulates energy expenditure by modulating NAD+ metabolism and SIRT1 activity. Nature 2009, 458, 1056–1060. [Google Scholar] [CrossRef]
  165. Wilson, B.J.; Tremblay, A.M.; Deblois, G.v.; Sylvain-Drolet, G.; Giguère, V. An Acetylation Switch Modulates the Transcriptional Activity of Estrogen-Related Receptor α. Mol. Endocrinol. 2010, 24, 1349–1358. [Google Scholar] [CrossRef]
  166. Annunziata, G.; Maisto, M.; Schisano, C.; Ciampaglia, R.; Narciso, V.; Tenore, G.C.; Novellino, E. Resveratrol as a Novel Anti-Herpes Simplex Virus Nutraceutical Agent: An Overview. Viruses 2018, 10, 473. [Google Scholar] [CrossRef]
  167. Kolouchová, I.; Maťátková, O.; Paldrychová, M.; Kodeš, Z.; Kvasničková, E.; Sigler, K.; Čejková, A.; Šmidrkal, J.; Demnerová, K.; Masák, J. Resveratrol, pterostilbene, and baicalein: Plant-derived anti-biofilm agents. Folia Microbiol. 2018, 63, 261–272. [Google Scholar] [CrossRef] [PubMed]
  168. Martelli, G.; Giacomini, D. Antibacterial and antioxidant activities for natural and synthetic dual-active compounds. Eur. J. Med. Chem. 2018, 158, 91–105. [Google Scholar] [CrossRef]
  169. Kumar, S.N.; Nambisan, B. Antifungal Activity of Diketopiperazines and Stilbenes Against Plant Pathogenic Fungi In Vitro. Appl. Biochem. Biotechnol. 2014, 172, 741–754. [Google Scholar] [CrossRef] [PubMed]
  170. Li, X.; Li, Y.; Xiong, B.; Qiu, S. Progress of Antimicrobial Mechanisms of Stilbenoids. Pharmaceutics 2024, 16, 663. [Google Scholar] [CrossRef]
  171. Mattio, L.M.; Dallavalle, S.; Musso, L.; Filardi, R.; Franzetti, L.; Pellegrino, L.; D’Incecco, P.; Mora, D.; Pinto, A.; Arioli, S. Antimicrobial activity of resveratrol-derived monomers and dimers against foodborne pathogens. Sci. Rep. 2019, 9, 19525. [Google Scholar] [CrossRef] [PubMed]
  172. Gerits, E.; Defraine, V.; Vandamme, K.; Cremer, K.D.; Brucker, K.D.; Thevissen, K.; Cammue, B.P.A.; Beullens, S.; Fauvart, M.; Verstraeten, N.; et al. Repurposing Toremifene for Treatment of Oral Bacterial Infections. Antimicrob. Agents Chemother. 2017, 61, 10.1128/aac.01846-01816. [Google Scholar] [CrossRef]
  173. Lim, Y.R.I.; Preshaw, P.M.; Lim, L.P.; Ong, M.M.A.; Lin, H.-S.; Tan, K.S. Pterostilbene complexed with cyclodextrin exerts antimicrobial and anti-inflammatory effects. Sci. Rep. 2020, 10, 9072. [Google Scholar] [CrossRef]
  174. Yang, B.; Yao, H.; Li, D.; Liu, Z. The phosphatidylglycerol phosphate synthase PgsA utilizes a trifurcated amphipathic cavity for catalysis at the membrane-cytosol interface. Curr. Res. Struct. Biol. 2021, 3, 312–323. [Google Scholar] [CrossRef]
  175. Hrast, M.; Rožman, K.; Ogris, I.; Škedelj, V.; Patin, D.; Sova, M.; Barreteau, H.; Gobec, S.; Grdadolnik, S.G.; Zega, A. Evaluation of the published kinase inhibitor set to identify multiple inhibitors of bacterial ATP-dependent mur ligases. J. Enzym. Inhib. Med. Chem. 2019, 34, 1010–1017. [Google Scholar] [CrossRef] [PubMed]
  176. Wu, X.-Z.; Cheng, A.-X.; Sun, L.-M.; Lou, H.-X. Effect of plagiochin E, an antifungal macrocyclic bis(bibenzyl), on cell wall chitin synthesis in Candida albicans. Acta Pharmacol. Sin. 2008, 29, 1478–1485. [Google Scholar] [CrossRef] [PubMed]
  177. Liu, Q.; Guo, X.; Jiang, G.; Wu, G.; Miao, H.; Liu, K.; Chen, S.; Sakamoto, N.; Kuno, T.; Yao, F.; et al. NADPH-Cytochrome P450 Reductase Ccr1 Is a Target of Tamoxifen and Participates in Its Antifungal Activity via Regulating Cell Wall Integrity in Fission Yeast. Antimicrob. Agents Chemother. 2020, 64, 10.1128/aac.00079-00020. [Google Scholar] [CrossRef]
  178. Park, H.B.; Crawford, J.M. Lumiquinone A, an α-Aminomalonate-Derived Aminobenzoquinone from Photorhabdus luminescens. J. Nat. Prod. 2015, 78, 1437–1441. [Google Scholar] [CrossRef]
  179. Houillé, B.; Papon, N.; Boudesocque, L.; Bourdeaud, E.; Besseau, S.; Courdavault, V.; Enguehard-Gueiffier, C.; Delanoue, G.; Guérin, L.; Bouchara, J.-P.; et al. Antifungal Activity of Resveratrol Derivatives against Candida Species. J. Nat. Prod. 2014, 77, 1658–1662. [Google Scholar] [CrossRef]
  180. Li, D.-D.; Zhao, L.-X.; Mylonakis, E.; Hu, G.-H.; Zou, Y.; Huang, T.-K.; Yan, L.; Wang, Y.; Jiang, Y.-Y. In Vitro and In Vivo Activities of Pterostilbene against Candida albicans Biofilms. Antimicrob. Agents Chemother. 2014, 58, 2344–2355. [Google Scholar] [CrossRef]
  181. De Filippis, B.; Ammazzalorso, A.; Amoroso, R.; Giampietro, L. Stilbene derivatives as new perspective in antifungal medicinal chemistry. Drug Dev. Res. 2019, 80, 285–293. [Google Scholar] [CrossRef]
  182. Mora-Pale, M.; Bhan, N.; Masuko, S.; James, P.; Wood, J.; McCallum, S.; Linhardt, R.J.; Dordick, J.S.; Koffas, M.A.G. Antimicrobial mechanism of resveratrol-trans-dihydrodimer produced from peroxidase-catalyzed oxidation of resveratrol. Biotechnol. Bioeng. 2015, 112, 2417–2428. [Google Scholar] [CrossRef]
  183. Kim, S.; Lee, D.G. Oxyresveratrol-induced DNA cleavage triggers apoptotic response in Candida albicans. Microbiology 2018, 164, 1112–1121. [Google Scholar] [CrossRef]
  184. Lee, W.; Lee, D.G. Resveratrol induces membrane and DNA disruption via pro-oxidant activity against Salmonella typhimurium. Biochem. Biophys. Res. Commun. 2017, 489, 228–234. [Google Scholar] [CrossRef] [PubMed]
  185. Bock, F.J.; Tait, S.W.G. Mitochondria as multifaceted regulators of cell death. Nat. Rev. Mol. Cell Biol. 2020, 21, 85–100. [Google Scholar] [CrossRef]
  186. Lee, J.; Lee, D.G. Novel Antifungal Mechanism of Resveratrol: Apoptosis Inducer in Candida albicans. Curr. Microbiol. 2015, 70, 383–389. [Google Scholar] [CrossRef] [PubMed]
  187. Dadi, P.K.; Ahmad, M.; Ahmad, Z. Inhibition of ATPase activity of Escherichia coli ATP synthase by polyphenols. Int. J. Biol. Macromol. 2009, 45, 72–79. [Google Scholar] [CrossRef]
  188. Hwang, D.; Lim, Y.-H. Resveratrol antibacterial activity against Escherichia coli is mediated by Z-ring formation inhibition via suppression of FtsZ expression. Sci. Rep. 2015, 5, 10029. [Google Scholar] [CrossRef] [PubMed]
  189. Barrows, J.M.; Goley, E.D. FtsZ dynamics in bacterial division: What, how, and why? Curr. Opin. Cell Biol. 2021, 68, 163–172. [Google Scholar] [CrossRef]
  190. Lou, F.; Wang, K.; Hou, Y.; Shang, X.; Tang, F. Inhibitory effect of resveratrol on swimming motility and adhesion ability against Salmonella enterica serovar Typhimurium infection. Microb. Pathog. 2023, 184, 106323. [Google Scholar] [CrossRef] [PubMed]
  191. Lee, S.A.; Wallis, C.M.; Rogers, E.E.; Burbank, L.P. Grapevine phenolic compounds influence cell surface adhesion of Xylella fastidiosa and bind to lipopolysaccharide. PLoS ONE 2020, 15, e0240101. [Google Scholar] [CrossRef]
  192. Kugaji, M.S.; Kumbar, V.M.; Peram, M.R.; Patil, S.; Bhat, K.G.; Diwan, P.V. Effect of Resveratrol on biofilm formation and virulence factor gene expression of Porphyromonas gingivalis in periodontal disease. APMIS 2019, 127, 187–195. [Google Scholar] [CrossRef]
  193. Uppuluri, P.; Chaturvedi, A.K.; Srinivasan, A.; Banerjee, M.; Ramasubramaniam, A.K.; Köhler, J.R.; Kadosh, D.; Lopez-Ribot, J.L. Dispersion as an Important Step in the Candida albicans Biofilm Developmental Cycle. PLoS Pathog. 2010, 6, e1000828. [Google Scholar] [CrossRef]
  194. Rosman, C.W.K.; van der Mei, H.C.; Sjollema, J. Influence of sub-inhibitory concentrations of antimicrobials on micrococcal nuclease and biofilm formation in Staphylococcus aureus. Sci. Rep. 2021, 11, 13241. [Google Scholar] [CrossRef]
  195. Siegel, R.L.; Kratzer, T.B.; Giaquinto, A.N.; Sung, H.; Jemal, A. Cancer statistics, 2025. CA Cancer J. Clin. 2025, 75, 10–45. [Google Scholar] [CrossRef]
  196. Jones, S.B. Cancer in the developing world: A call to action. BMJ 1999, 319, 505–508. [Google Scholar] [CrossRef]
  197. Siegel, R.; Ma, J.; Zou, Z.; Jemal, A. Cancer statistics, 2014. CA Cancer J. Clin. 2014, 64, 9–29. [Google Scholar] [CrossRef]
  198. Malhotra, A.; Nair, P.; Dhawan, D.K. Study to Evaluate Molecular Mechanics behind Synergistic Chemo-Preventive Effects of Curcumin and Resveratrol during Lung Carcinogenesis. PLoS ONE 2014, 9, e93820. [Google Scholar] [CrossRef] [PubMed]
  199. Mazué, F.; Delmas, D.; Murillo, G.; Saleiro, D.; Limagne, E.; Latruffe, N. Differential protective effects of red wine polyphenol extracts (RWEs) on colon carcinogenesis. Food Funct. 2014, 5, 663–670. [Google Scholar] [CrossRef] [PubMed]
  200. Back, J.H.; Zhu, Y.; Calabro, A.; Queenan, C.; Kim, A.S.; Arbesman, J.; Kim, A.L. Resveratrol-Mediated Downregulation of Rictor Attenuates Autophagic Process and Suppresses UV-Induced Skin Carcinogenesis. Photochem. Photobiol. 2012, 88, 1165–1172. [Google Scholar] [CrossRef]
  201. Hsieh, T.-c.; Yang, C.-J.; Lin, C.-Y.; Lee, Y.-S.; Wu, J.M. Control of stability of cyclin D1 by quinone reductase 2 in CWR22Rv1 prostate cancer cells. Carcinogenesis 2012, 33, 670–677. [Google Scholar] [CrossRef]
  202. Lin, C.; Crawford, D.R.; Lin, S.; Hwang, J.; Sebuyira, A.; Meng, R.; Westfall, J.E.; Tang, H.-Y.; Lin, S.; Yu, P.-Y.; et al. Inducible COX-2-dependent apoptosis in human ovarian cancer cells. Carcinogenesis 2011, 32, 19–26. [Google Scholar] [CrossRef]
  203. Rajasekaran, D.; Elavarasan, J.; Sivalingam, M.; Ganapathy, E.; Kumar, A.; Kalpana, K.; Sakthisekaran, D. Resveratrol interferes with N-nitrosodiethylamine-induced hepatocellular carcinoma at early and advanced stages in male Wistar rats. Mol. Med. Rep. 2011, 4, 1211–1217. [Google Scholar]
  204. Shrotriya, S.; Tyagi, A.; Deep, G.; Orlicky, D.J.; Wisell, J.; Wang, X.-J.; Sclafani, R.A.; Agarwal, R.; Agarwal, C. Grape seed extract and resveratrol prevent 4-nitroquinoline 1-oxide induced oral tumorigenesis in mice by modulating AMPK activation and associated biological responses. Mol. Carcinog. 2015, 54, 291–300. [Google Scholar] [CrossRef] [PubMed]
  205. Kang, H.J.; Youn, Y.-K.; Hong, M.-K.; Kim, L.S. Antiproliferation and Redifferentiation in Thyroid Cancer Cell Lines by Polyphenol Phytochemicals. JKMS 2011, 26, 893–899. [Google Scholar] [CrossRef]
  206. Tsan, M.-f.; White, J.E.; Maheshwari, J.G.; Chikkappa, G. Anti-leukemia Effect of Resveratrol. Leuk. Lymphoma 2002, 43, 983–987. [Google Scholar] [CrossRef]
  207. Temraz, S.; Mukherji, D.; Shamseddine, A. Potential Targets for Colorectal Cancer Prevention. Int. J. Mol. Sci. 2013, 14, 17279–17303. [Google Scholar] [CrossRef] [PubMed]
  208. Wang, D.; DuBois, R.N. The role of COX-2 in intestinal inflammation and colorectal cancer. Oncogene 2010, 29, 781–788. [Google Scholar] [CrossRef]
  209. Mosalpuria, K.; Hall, C.; Krishnamurthy, S.; Lodhi, A.; Hallman, D.M.; Baraniuk, M.S.; Bhattacharyya, A.; Lucci, A. Cyclooxygenase-2 expression in non-metastatic triple-negative breast cancer patients. Mol. Clin. Oncol. 2014, 2, 845–850. [Google Scholar] [CrossRef] [PubMed]
  210. Wang, D.; Guo, X.-Z.; Li, H.-Y.; Zhao, J.-J.; Shao, X.-D.; Wu, C.-Y. Prognostic significance of cyclooxygenase-2 protein in pancreatic cancer: A meta-analysis. Tumor Biol. 2014, 35, 10301–10307. [Google Scholar] [CrossRef]
  211. Jiao, J.; Ishikawa, T.-O.; Dumlao, D.S.; Norris, P.C.; Magyar, C.E.; Mikulec, C.; Catapang, A.; Dennis, E.A.; Fischer, S.M.; Herschman, H.R. Targeted Deletion and Lipidomic Analysis Identify Epithelial Cell COX-2 as a Major Driver of Chemically Induced Skin Cancer. Mol. Cancer Res. 2014, 12, 1677–1688. [Google Scholar] [CrossRef]
  212. Marnett, L.J. Aspirin and the potential role of prostaglandins in colon cancer. Cancer Res. 1992, 52, 5575. [Google Scholar]
  213. Yi, C.-O.; Jeon, B.T.; Shin, H.J.; Jeong, E.A.; Chang, K.C.; Lee, J.E.; Lee, D.H.; Kim, H.J.; Kang, S.S.; Cho, G.J.; et al. Resveratrol activates AMPK and suppresses LPS-induced NF-κB-dependent COX-2 activation in RAW 264.7 macrophage cells. ACB 2011, 44, 194–203. [Google Scholar] [CrossRef]
  214. Han, Y.; Jiang, C.; Tang, J.; Wang, C.; Wu, P.; Zhang, G.; Liu, W.; Jamangulova, N.; Wu, X.; Song, X. Resveratrol reduces morphine tolerance by inhibiting microglial activation via AMPK signalling. Eur. J. Pain 2014, 18, 1458–1470. [Google Scholar] [CrossRef] [PubMed]
  215. GÜROCAK, Ş.; Karabulut, E.; KARADAĞ SOYLU, N.; ÖZGÖR, D.; Ozkeles, N.; Karabulut, A. Preventive effects of resveratrol against azoxymethane induced damage in rat liver. Asian Pac. J. Cancer Prev. 2013, 14. [Google Scholar] [CrossRef] [PubMed]
  216. Akca, H.; Demiray, A.; Aslan, M.; Acikbas, I.; Tokgun, O. Tumour suppressor PTEN enhanced enzyme activity of GPx, SOD and catalase by suppression of PI3K/AKT pathway in non-small cell lung cancer cell lines. J. Enzym. Inhib. Med. Chem. 2013, 28, 539–544. [Google Scholar] [CrossRef] [PubMed]
  217. Mena, S.; Rodríguez, M.L.; Ponsoda, X.; Estrela, J.M.; Jäättela, M.; Ortega, A.L. Pterostilbene-Induced Tumor Cytotoxicity: A Lysosomal Membrane Permeabilization-Dependent Mechanism. PLoS ONE 2012, 7, e44524. [Google Scholar] [CrossRef]
  218. Casanova, F.; Quarti, J.; da Costa, D.C.F.; Ramos, C.A.; da Silva, J.L.; Fialho, E. Resveratrol chemosensitizes breast cancer cells to melphalan by cell cycle arrest. J. Cell. Biochem. 2012, 113, 2586–2596. [Google Scholar] [CrossRef]
  219. Yu, X.-D.; Yang, J.-l.; Zhang, W.-L.; Liu, D.-X. Resveratrol inhibits oral squamous cell carcinoma through induction of apoptosis and G2/M phase cell cycle arrest. Tumor Biol. 2016, 37, 2871–2877. [Google Scholar] [CrossRef]
  220. Gokbulut, A.A.; Apohan, E.; Baran, Y. Resveratrol and quercetin-induced apoptosis of human 232B4 chronic lymphocytic leukemia cells by activation of caspase-3 and cell cycle arrest. Hematology 2013, 18, 144–150. [Google Scholar] [CrossRef] [PubMed]
  221. McCormack, D.; McFadden, D. A Review of Pterostilbene Antioxidant Activity and Disease Modification. Oxidative Med. Cell. Longev. 2013, 2013, 575482. [Google Scholar] [CrossRef]
  222. Schneider, J.G.; Alosi, J.A.; McDonald, D.E.; McFadden, D.W. Pterostilbene Inhibits Lung Cancer Through Induction of Apoptosis1. J. Surg. Res. 2010, 161, 18–22. [Google Scholar] [CrossRef]
  223. Suh, N.; Paul, S.; Hao, X.; Simi, B.; Xiao, H.; Rimando, A.M.; Reddy, B.S. Pterostilbene, an Active Constituent of Blueberries, Suppresses Aberrant Crypt Foci Formation in the Azoxymethane-Induced Colon Carcinogenesis Model in Rats. Clin. Cancer Res. 2007, 13, 350–355. [Google Scholar] [CrossRef]
  224. Cichocki, M.; Paluszczak, J.; Szaefer, H.; Piechowiak, A.; Rimando, A.M.; Baer-Dubowska, W. Pterostilbene is equally potent as resveratrol in inhibiting 12-O-tetradecanoylphorbol-13-acetate activated NFκB, AP-1, COX-2, and iNOS in mouse epidermis. Mol. Nutr. Food Res. 2008, 52, S62–S70. [Google Scholar] [CrossRef] [PubMed]
  225. Dhar, S.; Kumar, A.; Rimando, A.M.; Zhang, X.; Levenson, A.S. Resveratrol and pterostilbene epigenetically restore PTEN expression by targeting oncomiRs of the miR-17 family in prostate cancer. Oncotarget 2015, 6, 27214. [Google Scholar] [CrossRef]
  226. Paul, S.; DeCastro, A.J.; Lee, H.J.; Smolarek, A.K.; So, J.Y.; Simi, B.; Wang, C.X.; Zhou, R.; Rimando, A.M.; Suh, N. Dietary intake of pterostilbene, a constituent of blueberries, inhibits the β-catenin/p65 downstream signaling pathway and colon carcinogenesis in rats. Carcinogenesis 2010, 31, 1272–1278. [Google Scholar] [CrossRef] [PubMed]
  227. McCormack, D.; Schneider, J.; McDonald, D.; McFadden, D. The antiproliferative effects of pterostilbene on breast cancer in vitro are via inhibition of constitutive and leptin-induced Janus kinase/signal transducer and activator of transcription activation. Am. J. Surg. 2011, 202, 541–544. [Google Scholar] [CrossRef]
  228. Liu, Y.; Wang, L.; Wu, Y.; Lv, C.; Li, X.; Cao, X.; Yang, M.; Feng, D.; Luo, Z. Pterostilbene exerts antitumor activity against human osteosarcoma cells by inhibiting the JAK2/STAT3 signaling pathway. Toxicology 2013, 304, 120–131. [Google Scholar] [CrossRef]
  229. Chen, R.-J.; Ho, C.-T.; Wang, Y.-J. Pterostilbene induces autophagy and apoptosis in sensitive and chemoresistant human bladder cancer cells. Mol. Nutr. Food Res. 2010, 54, 1819–1832. [Google Scholar] [CrossRef]
  230. Chakraborty, A.; Bodipati, N.; Demonacos, M.K.; Peddinti, R.; Ghosh, K.; Roy, P. Long term induction by pterostilbene results in autophagy and cellular differentiation in MCF-7 cells via ROS dependent pathway. Mol. Cell. Endocrinol. 2012, 355, 25–40. [Google Scholar] [CrossRef] [PubMed]
  231. Magee, J.A.; Piskounova, E.; Morrison, S.J. Cancer Stem Cells: Impact, Heterogeneity, and Uncertainty. Cancer Cell 2012, 21, 283–296. [Google Scholar] [CrossRef]
  232. Nguyen, L.V.; Vanner, R.; Dirks, P.; Eaves, C.J. Cancer stem cells: An evolving concept. Nat. Rev. Cancer 2012, 12, 133–143. [Google Scholar] [CrossRef]
  233. Visvader, J.E.; Lindeman, G.J. Cancer Stem Cells: Current Status and Evolving Complexities. Cell Stem Cell 2012, 10, 717–728. [Google Scholar] [CrossRef]
  234. Hagiwara, K.; Kosaka, N.; Yoshioka, Y.; Takahashi, R.-u.; Takeshita, F.; Ochiya, T. Stilbene derivatives promote Ago2-dependent tumour-suppressive microRNA activity. Sci. Rep. 2012, 2, 314. [Google Scholar] [CrossRef]
  235. Mak, K.-K.; Wu, A.T.H.; Lee, W.-H.; Chang, T.-C.; Chiou, J.-F.; Wang, L.-S.; Wu, C.-H.; Huang, C.-Y.F.; Shieh, Y.-S.; Chao, T.-Y.; et al. Pterostilbene, a bioactive component of blueberries, suppresses the generation of breast cancer stem cells within tumor microenvironment and metastasis via modulating NF-κB/microRNA 448 circuit. Mol. Nutr. Food Res. 2013, 57, 1123–1134. [Google Scholar] [CrossRef]
  236. Lee, C.-M.; Su, Y.-H.; Huynh, T.-T.; Lee, W.-H.; Chiou, J.-F.; Lin, Y.-K.; Hsiao, M.; Wu, C.-H.; Lin, Y.-F.; Wu, A.T.H.; et al. BlueBerry Isolate, Pterostilbene, Functions as a Potential Anticancer Stem Cell Agent in Suppressing Irradiation-Mediated Enrichment of Hepatoma Stem Cells. Evid.-Based Complement. Altern. Med. 2013, 2013, 258425. [Google Scholar] [CrossRef]
  237. Hsieh, T.-C.; Bennett, D.J.; Lee, Y.-S.; Wu, E.; Wu, J.M. In Silico and Biochemical Analyses Identify Quinone Reductase 2 as a Target of Piceatannol. Curr. Med. Chem. 2013, 20, 4195–4202. [Google Scholar] [CrossRef] [PubMed]
  238. Murias, M.; Jäger, W.; Handler, N.; Erker, T.; Horvath, Z.; Szekeres, T.; Nohl, H.; Gille, L. Antioxidant, prooxidant and cytotoxic activity of hydroxylated resveratrol analogues: Structure–activity relationship. Biochem. Pharmacol. 2005, 69, 903–912. [Google Scholar] [CrossRef]
  239. Piotrowska, H.; Kucinska, M.; Murias, M. Biological activity of piceatannol: Leaving the shadow of resveratrol. Mutat. Res./Rev. Mutat. Res. 2012, 750, 60–82. [Google Scholar] [CrossRef] [PubMed]
  240. Azmi, A.S.; Bhat, S.H.; Hadi, S.M. Resveratrol–Cu(II) induced DNA breakage in human peripheral lymphocytes: Implications for anticancer properties. FEBS Lett. 2005, 579, 3131–3135. [Google Scholar] [CrossRef] [PubMed]
  241. Sirerol, J.A.; Rodríguez, M.L.; Mena, S.; Asensi, M.A.; Estrela, J.M.; Ortega, A.L. Role of Natural Stilbenes in the Prevention of Cancer. Oxidative Med. Cell. Longev. 2016, 2016, 3128951. [Google Scholar] [CrossRef] [PubMed]
  242. Ashikawa, K.; Majumdar, S.; Banerjee, S.; Bharti, A.C.; Shishodia, S.; Aggarwal, B.B. Piceatannol Inhibits TNF-Induced NF-κB Activation and NF-κB-Mediated Gene Expression Through Suppression of IκBα Kinase and p65 Phosphorylation1. J. Immunol. 2002, 169, 6490–6497. [Google Scholar] [CrossRef]
  243. Islam, S.; Hassan, F.; Mu, M.M.; Ito, H.; Koide, N.; Mori, I.; Yoshida, T.; Yokochi, T. Piceatannol Prevents Lipopolysaccharide (LPS)-Induced Nitric Oxide (NO) Production and Nuclear Factor (NF)-κB Activation by Inhibiting IκB Kinase (IKK). Microbiol. Immunol. 2004, 48, 729–736. [Google Scholar] [CrossRef]
  244. Jin, C.-Y.; Moon, D.-O.; Lee, K.-J.; Kim, M.-O.; Lee, J.-D.; Choi, Y.H.; Park, Y.-M.; Kim, G.-Y. Piceatannol attenuates lipopolysaccharide-induced NF-κB activation and NF-κB-related proinflammatory mediators in BV2 microglia. Pharmacol. Res. 2006, 54, 461–467. [Google Scholar] [CrossRef]
  245. Xueyan, R.; Jia, Y.; Xuefeng, Y.; Lidan, T.; Qingjun, K. Isolation and purification of five phenolic compounds from the Xinjiang wine grape (Vitis Vinifera) and determination of their antioxidant mechanism at cellular level. Eur. Food Res. Technol. 2018, 244, 1569–1579. [Google Scholar] [CrossRef]
  246. Sharma, A.; Boise, L.H.; Shanmugam, M. Cancer Metabolism and the Evasion of Apoptotic Cell Death. Cancers 2019, 11, 1144. [Google Scholar] [CrossRef]
  247. Barjot, C.; Tournaire, M.; Castagnino, C.; Vigor, C.; Vercauteren, J.; Rossi, J.-F. Evaluation of antitumor effects of two vine stalk oligomers of resveratrol on a panel of lymphoid and myeloid cell lines: Comparison with resveratrol. Life Sci. 2007, 81, 1565–1574. [Google Scholar] [CrossRef]
  248. Billard, C.; Izard, J.-C.; Roman, V.; Kern, C.; Mathiot, C.; Mentz, F.; Kolb, J.-P. Comparative Antiproliferative and Apoptotic Effects of Resveratrol, ϵ-viniferin and Vine-shots Derived Polyphenols (Vineatrols) on Chronic B Lymphocytic Leukemia Cells and Normal Human Lymphocytes. Leuk. Lymphoma 2002, 43, 1991–2002. [Google Scholar] [CrossRef]
  249. Nivelle, L.; Aires, V.; Rioult, D.; Martiny, L.; Tarpin, M.; Delmas, D. Molecular analysis of differential antiproliferative activity of resveratrol, epsilon viniferin and labruscol on melanoma cells and normal dermal cells. Food Chem. Toxicol. 2018, 116, 323–334. [Google Scholar] [CrossRef] [PubMed]
  250. Özdemir, F.; Akalın, G.; Şen, M.; Önder, N.I.; Işcan, A.; Kutlu, H.M.; Incesu, Z. Towards Novel Anti-tumor Strategies for Hepatic Cancer: ɛ-Viniferin in Combination with Vincristine Displays Pharmacodynamic Synergy at Lower Doses in HepG2 Cells. OMICS J. Integr. Biol. 2014, 18, 324–334. [Google Scholar] [CrossRef]
  251. Özdemir, F.; İncesu, Z.; Şena, M.; Öndera, N.İ.; Dikme, M. Implications of Enhanced Effectiveness of Vincristine Sulfate/ε-Viniferin Combination Compared to Vincristine Sulfate Only on HepG2 Cells. Dicle Tıp Derg. 2016, 43, 534–541. [Google Scholar]
  252. Tarhan, S.; Özdemir, F.; İncesu, Z.; Demirkan, E.S. Direct and protective effects of single or combined addition of vincristine and ε-viniferin on human HepG2 cellular oxidative stress markers in vitro. Cytotechnology 2016, 68, 1081–1094. [Google Scholar] [CrossRef]
  253. Özdemir, F.; Apaydın, E.; Önder, N.İ.; Şen, M.; Ayrım, A.; Öğünç, Y.; İncesu, Z. Apoptotic effects of ε-viniferin in combination with cis-platin in C6 cells. Cytotechnology 2018, 70, 1061–1073. [Google Scholar] [CrossRef]
  254. Ito, T.; Akao, Y.; Yi, H.; Ohguchi, K.; Matsumoto, K.; Tanaka, T.; Iinuma, M.; Nozawa, Y. Antitumor effect of resveratrol oligomers against human cancer cell lines and the molecular mechanism of apoptosis induced by vaticanol C. Carcinogenesis 2003, 24, 1489–1497. [Google Scholar] [CrossRef]
  255. Muhtadi; Hakim, E.H.; Juliawaty, L.D.; Syah, Y.M.; Achmad, S.A.; Latip, J.; Ghisalberti, E.L. Cytotoxic resveratrol oligomers from the tree bark of Dipterocarpus hasseltii. Fitoterapia 2006, 77, 550–555. [Google Scholar] [CrossRef]
  256. Nur Ainaa, A.; Norizan Ahmat, N.A.; Mashita Abdullah, M.A.; Norrizah Jaafar Sidik, N.J.S.; Syed Ahmad, T. Antioxidant, antimicrobial and cytotoxic activities of resveratrol oligomers of Shorea macroptera dyer. Aust. J. Basic. Appl. Sci. 2012, 6, 431–436. [Google Scholar]
  257. Wibowo, A.; Ahmat, N.; Hamzah, A.S.; Latif, F.A.; Norrizah, J.S.; Khong, H.Y.; Takayama, H. Identification and biological activity of secondary metabolites from Dryobalanops beccarii. Phytochem. Lett. 2014, 9, 117–122. [Google Scholar] [CrossRef]
  258. Colin, D.; Lancon, A.; Delmas, D.; Lizard, G.; Abrossinow, J.; Kahn, E.; Jannin, B.; Latruffe, N. Antiproliferative activities of resveratrol and related compounds in human hepatocyte derived HepG2 cells are associated with biochemical cell disturbance revealed by fluorescence analyses. Biochimie 2008, 90, 1674–1684. [Google Scholar] [CrossRef] [PubMed]
  259. Aja, I.; Ruiz-Larrea, M.B.; Courtois, A.; Krisa, S.; Richard, T.; Ruiz-Sanz, J.-I. Screening of Natural Stilbene Oligomers from Vitis vinifera for Anticancer Activity on Human Hepatocellular Carcinoma Cells. Antioxidants 2020, 9, 469. [Google Scholar] [CrossRef] [PubMed]
  260. Ames, B.N.; Shigenaga, M.K.; Hagen, T.M. Oxidants, antioxidants, and the degenerative diseases of aging. Proc. Natl. Acad. Sci. USA 1993, 90, 7915–7922. [Google Scholar] [CrossRef]
  261. Pintea, A.; Rugină, D.; Pop, R.; Bunea, A.; Socaciu, C.; Diehl, H.A. Antioxidant Effect of Trans-Resveratrol in Cultured Human Retinal Pigment Epithelial Cells. J. Ocul. Pharmacol. Ther. 2011, 27, 315–321. [Google Scholar] [CrossRef]
  262. Ksila, M.; Vejux, A.; Prost-Camus, E.; Durand, P.; Ghzaiel, I.; Nury, T.; Duprey, D.; Meziane, S.; Masmoudi-Kouki, O.; Latruffe, N.; et al. Cytotoxic and Antioxidant Activities of Imine Analogs of Trans-Resveratrol towards Murine Neuronal N2a Cells. Molecules 2022, 27, 4713. [Google Scholar] [CrossRef]
  263. Almeida, T.C.; Seibert, J.B.; Almeida, S.H.d.S.; Amparo, T.R.; Teixeira, L.F.d.M.; Barichello, J.M.; Postacchini, B.B.; Santos, O.D.H.d.; Silva, G.N.d. Polymeric micelles containing resveratrol: Development, characterization, cytotoxicity on tumor cells and antimicrobial activity. Braz. J. Pharm. Sci. 2020, 56, e18411. [Google Scholar] [CrossRef]
  264. Selvaraj, S.; Mohan, A.; Narayanan, S.; Sethuraman, S.; Krishnan, U.M. Dose-Dependent Interaction of trans-Resveratrol with Biomembranes: Effects on Antioxidant Property. J. Med. Chem. 2013, 56, 970–981. [Google Scholar] [CrossRef]
  265. Khayoon, H.A.; Al-Rekabi, F.M. Cytotoxic effect of resveratrol on colorectal cancer cell line. Iraqi J. Vet. Med. 2020, 44, 68–74. [Google Scholar] [CrossRef]
  266. Osman, A.-M.M.; Bayoumi, H.M.; Al-Harthi, S.E.; Damanhouri, Z.A.; ElShal, M.F. Modulation of doxorubicin cytotoxicity by resveratrol in a human breast cancer cell line. Cancer Cell Int. 2012, 12, 47. [Google Scholar] [CrossRef]
  267. Jeon, D.; Jo, M.; Lee, Y.; Park, S.-H.; Phan, H.T.L.; Nam, J.H.; Namkung, W. Inhibition of ANO1 by Cis- and Trans-Resveratrol and Their Anticancer Activity in Human Prostate Cancer PC-3 Cells. Int. J. Mol. Sci. 2023, 24, 1186. [Google Scholar] [CrossRef]
  268. Fernández-Pérez, F.; Belchí-Navarro, S.; Almagro, L.; Bru, R.; Pedreño, M.A.; Gómez-Ros, L.V. Cytotoxic Effect of Natural trans-Resveratrol Obtained from Elicited Vitis vinifera Cell Cultures on Three Cancer Cell Lines. Plant Foods Hum. Nutr. 2012, 67, 422–429. [Google Scholar] [CrossRef]
  269. Anisimova, N.Y.U.; Kiselevsky, M.V.; Sosnov, A.V.; Sadovnikov, S.V.; Stankov, I.N.; Gakh, A.A. Trans-, cis-, and dihydro-resveratrol: A comparative study. Chem. Cent. J. 2011, 5, 88. [Google Scholar] [CrossRef]
  270. Siddiqui, M.; Saquib, Q.; Ahamed, M.; Ahmad, J.; Al-Khedhairy, A.; Abou-Tarboush, F.; Musarrat, J. Effect of trans-resveratrol on rotenone-induced cytotoxicity in human breast adenocarcinoma cells. Toxicol. Int. 2011, 18, 105. [Google Scholar]
  271. Kocsis, Z.; Marcsek, Z.L.; Jakab, M.G.; Szende, B.; Tompa, A. Chemopreventive properties of trans—Resveratrol against the cytotoxicity of chloroacetanilide herbicides in vitro. Int. J. Hyg. Environ. Health 2005, 208, 211–218. [Google Scholar] [CrossRef] [PubMed]
  272. Siddiqui, M.A.; Kashyap, M.P.; Kumar, V.; Al-Khedhairy, A.A.; Musarrat, J.; Pant, A.B. Protective potential of trans-resveratrol against 4-hydroxynonenal induced damage in PC12 cells. Toxicol. Vitr. 2010, 24, 1592–1598. [Google Scholar] [CrossRef]
  273. Leischner, C.; Burkard, M.; Michel, A.; Berchtold, S.; Niessner, H.; Marongiu, L.; Busch, C.; Frank, J.; Lauer, U.M.; Venturelli, S. Comparative Analysis of the Antitumor Activity of Cis- and Trans-Resveratrol in Human Cancer Cells with Different p53 Status. Molecules 2021, 26, 5586. [Google Scholar] [CrossRef] [PubMed]
  274. Miksits, M.; Wlcek, K.; Svoboda, M.; Kunert, O.; Haslinger, E.; Thalhammer, T.; Szekeres, T.; Jäger, W. Antitumor activity of resveratrol and its sulfated metabolites against human breast cancer cells. Planta Medica 2009, 75, 1227–1230. [Google Scholar] [CrossRef] [PubMed]
  275. Marel, A.-K.; Lizard, G.; Izard, J.-C.; Latruffe, N.; Delmas, D. Inhibitory effects of trans-resveratrol analogs molecules on the proliferation and the cell cycle progression of human colon tumoral cells. Mol. Nutr. Food Res. 2008, 52, 538–548. [Google Scholar] [CrossRef]
  276. Filomeni, G.; Graziani, I.; Rotilio, G.; Ciriolo, M.R. trans-Resveratrol induces apoptosis in human breast cancer cells MCF-7 by the activation of MAP kinases pathways. Genes. Nutr. 2007, 2, 295–305. [Google Scholar] [CrossRef]
  277. Nashine, S.; Nesburn, A.B.; Kuppermann, B.D.; Kenney, M.C. Role of Resveratrol in Transmitochondrial AMD RPE Cells. Nutrients 2020, 12, 159. [Google Scholar] [CrossRef]
  278. El-Melegy, M.G.; Eltaher, H.M.; Gaballah, A.; El-Kamel, A.H. Enhanced oral permeability of Trans-Resveratrol using nanocochleates for boosting anticancer efficacy; in-vitro and ex-vivo appraisal. Eur. J. Pharm. Biopharm. 2021, 168, 166–183. [Google Scholar] [CrossRef]
  279. Tsang, S.W.; Zhang, H.; Lin, Z.; Mu, H.; Bian, Z.-X. Anti-fibrotic effect of trans-resveratrol on pancreatic stellate cells. Biomed. Pharmacother. 2015, 71, 91–97. [Google Scholar] [CrossRef]
  280. dos Santos Moreno, C.; Rogero, S.O.; Ikeda, T.I.; Cruz, Á.S.; Rogero, J.R. Resveratrol and radiation biological effects. Int. J. Nutrology 2012, 5, 28–33. [Google Scholar] [CrossRef]
  281. Lucas, I.K.; Kolodziej, H. Trans-resveratrol induces apoptosis through ROS-triggered mitochondria-dependent pathways in A549 human lung adenocarcinoma epithelial cells. Planta Medica 2015, 81, 1038–1044. [Google Scholar] [CrossRef]
  282. Mannal, P.; McDonald, D.; McFadden, D. Pterostilbene and tamoxifen show an additive effect against breast cancer in vitro. Am. J. Surg. 2010, 200, 577–580. [Google Scholar] [CrossRef]
  283. Elsherbini, A.M.; Sheweita, S.A.; Sultan, A.S. Pterostilbene as a Phytochemical Compound Induces Signaling Pathways Involved in the Apoptosis and Death of Mutant P53-Breast Cancer Cell Lines. Nutr. Cancer 2021, 73, 1976–1984. [Google Scholar] [CrossRef]
  284. Wakimoto, R.; Ono, M.; Takeshima, M.; Higuchi, T.; Nakano, S. Differential Anticancer Activity of Pterostilbene Against Three Subtypes of Human Breast Cancer Cells. Anticancer Res. 2017, 37, 6153–6159. [Google Scholar]
  285. Shin, H.J.; Han, J.M.; Choi, Y.S.; Jung, H.J. Pterostilbene Suppresses both Cancer Cells and Cancer Stem-Like Cells in Cervical Cancer with Superior Bioavailability to Resveratrol. Molecules 2020, 25, 228. [Google Scholar] [CrossRef]
  286. Chatterjee, K.; Mukherjee, S.; Vanmanen, J.; Banerjee, P.; Fata, J.E. Dietary Polyphenols, Resveratrol and Pterostilbene Exhibit Antitumor Activity on an HPV E6-Positive Cervical Cancer Model: An in vitro and in vivo Analysis. Front. Oncol. 2019, 9, 352. [Google Scholar] [CrossRef]
  287. Zhang, Y.; Li, Y.; Sun, C.; Chen, X.; Han, L.; Wang, T.; Liu, J.; Chen, X.; Zhao, D. Effect of Pterostilbene, a Natural Derivative of Resveratrol, in the Treatment of Colorectal Cancer through Top1/Tdp1-Mediated DNA Repair Pathway. Cancers 2021, 13, 4002. [Google Scholar] [CrossRef] [PubMed]
  288. Wawszczyk, J.; Jesse, K.; Smolik, S.; Kapral, M. Mechanism of Pterostilbene-Induced Cell Death in HT-29 Colon Cancer Cells. Molecules 2022, 27, 369. [Google Scholar] [CrossRef]
  289. Hsiao, Y.-H.; Chen, N.-C.; Koh, Y.-C.; Nagabhushanam, K.; Ho, C.-T.; Pan, M.-H. Pterostilbene Inhibits Adipocyte Conditioned-Medium-Induced Colorectal Cancer Cell Migration through Targeting FABP5-Related Signaling Pathway. J. Agric. Food Chem. 2019, 67, 10321–10329. [Google Scholar] [CrossRef] [PubMed]
  290. Wen, W.; Lowe, G.; Roberts, C.M.; Finlay, J.; Han, E.S.; Glackin, C.A.; Dellinger, T.H. Pterostilbene, a natural phenolic compound, synergizes the antineoplastic effects of megestrol acetate in endometrial cancer. Sci. Rep. 2017, 7, 12754. [Google Scholar] [CrossRef]
  291. Wen, W.; Lowe, G.; Roberts, C.M.; Finlay, J.; Han, E.S.; Glackin, C.A.; Dellinger, T.H. Pterostilbene Suppresses Ovarian Cancer Growth via Induction of Apoptosis and Blockade of Cell Cycle Progression Involving Inhibition of the STAT3 Pathway. Int. J. Mol. Sci. 2018, 19, 1983. [Google Scholar] [CrossRef]
  292. Lin, V.C.-H.; Tsai, Y.-C.; Lin, J.-N.; Fan, L.-L.; Pan, M.-H.; Ho, C.-T.; Wu, J.-Y.; Way, T.-D. Activation of AMPK by Pterostilbene Suppresses Lipogenesis and Cell-Cycle Progression in p53 Positive and Negative Human Prostate Cancer Cells. J. Agric. Food Chem. 2012, 60, 6399–6407. [Google Scholar] [CrossRef]
  293. Mannal, P.W.; Alosi, J.A.; Schneider, J.G.; McDonald, D.E.; McFadden, D.W. Pterostilbene Inhibits Pancreatic Cancer In Vitro. J. Gastrointest. Surg. 2010, 14, 873–879. [Google Scholar] [CrossRef] [PubMed]
  294. Bracht, J.W.P.; Karachaliou, N.; Berenguer, J.; Pedraz-Valdunciel, C.; Filipska, M.; Codony-Servat, C.; Codony-Servat, J.; Rosell, R. Osimertinib and pterostilbene in EGFR-mutation-positive non-small cell lung cancer (NSCLC). Int. J. Biol. Sci. 2019, 15, 2607–2614. [Google Scholar] [CrossRef]
  295. Guo, L.; Tan, K.; Wang, H.; Zhang, X. Pterostilbene inhibits hepatocellular carcinoma through p53/SOD2/ROS-mediated mitochondrial apoptosis. Oncol. Rep. 2016, 36, 3233–3240. [Google Scholar] [CrossRef]
  296. Mori, S.; Kishi, S.; Honoki, K.; Fujiwara-Tani, R.; Moriguchi, T.; Sasaki, T.; Fujii, K.; Tsukamoto, S.; Fujii, H.; Kido, A.; et al. Anti-Stem Cell Property of Pterostilbene in Gastrointestinal Cancer Cells. Int. J. Mol. Sci. 2020, 21, 9347. [Google Scholar] [CrossRef]
  297. Chang, H.-P.; Lu, C.-C.; Chiang, J.-H.; Tsai, F.-J.; Juan, Y.-N.; Tsao, J.-W.; Chiu, H.-Y.; Yang, J.-S. Pterostilbene modulates the suppression of multidrug resistance protein 1 and triggers autophagic and apoptotic mechanisms in cisplatin-resistant human oral cancer CAR cells via AKT signaling. Int. J. Oncol. 2018, 52, 1504–1514. [Google Scholar] [CrossRef] [PubMed]
  298. Aly, S.; El-Kamel, A.H.; Sheta, E.; El-Habashy, S.E. Chondroitin/Lactoferrin-dual functionalized pterostilbene-solid lipid nanoparticles as targeted breast cancer therapy. Int. J. Pharm. 2023, 642, 123163. [Google Scholar] [CrossRef] [PubMed]
  299. Chen, R.-J.; Lyu, Y.-J.; Chen, Y.-Y.; Lee, Y.-C.; Pan, M.-H.; Ho, Y.-S.; Wang, Y.-J. Chloroquine Potentiates the Anticancer Effect of Pterostilbene on Pancreatic Cancer by Inhibiting Autophagy and Downregulating the RAGE/STAT3 Pathway. Molecules 2021, 26, 6741. [Google Scholar] [CrossRef]
  300. Kumar, V.; Haldar, S.; Das, N.S.; Ghosh, S.; Dhankhar, P.; Sircar, D.; Roy, P. Pterostilbene-isothiocyanate inhibits breast cancer metastasis by selectively blocking IKK-β/NEMO interaction in cancer cells. Biochem. Pharmacol. 2021, 192, 114717. [Google Scholar] [CrossRef]
  301. Tong, C.; Wang, Y.; Li, J.; Cen, W.; Zhang, W.; Zhu, Z.; Yu, J.; Lu, B. Pterostilbene inhibits gallbladder cancer progression by suppressing the PI3K/Akt pathway. Sci. Rep. 2021, 11, 4391. [Google Scholar] [CrossRef] [PubMed]
  302. Gao, H.; Liu, Z.; Xu, W.; Wang, Q.; Zhang, C.; Ding, Y.; Nie, W.; Lai, J.; Chen, Y.; Huang, H. Pterostilbene promotes mitochondrial apoptosis and inhibits proliferation in glioma cells. Sci. Rep. 2021, 11, 6381. [Google Scholar] [CrossRef]
  303. Sáez, V.; Pastene, E.; Vergara, C.; Mardones, C.; Hermosín-Gutiérrez, I.; Gómez-Alonso, S.; Gómez, M.V.; Theoduloz, C.; Riquelme, S.; von Baer, D. Oligostilbenoids in Vitis vinifera L. Pinot Noir grape cane extract: Isolation, characterization, in vitro antioxidant capacity and anti-proliferative effect on cancer cells. Food Chem. 2018, 265, 101–110. [Google Scholar] [CrossRef] [PubMed]
  304. Kuo, P.-L.; Hsu, Y.-L. The grape and wine constituent piceatannol inhibits proliferation of human bladder cancer cells via blocking cell cycle progression and inducing Fas/membrane bound Fas ligand-mediated apoptotic pathway. Mol. Nutr. Food Res. 2008, 52, 408–418. [Google Scholar] [CrossRef] [PubMed]
  305. Morales, P.; Haza, A.I. Selective apoptotic effects of piceatannol and myricetin in human cancer cells. J. Appl. Toxicol. 2012, 32, 986–993. [Google Scholar] [CrossRef]
  306. Du, M.; Zhang, Z.; Gao, T. Piceatannol induced apoptosis through up-regulation of microRNA-181a in melanoma cells. Biol. Res. 2017, 50, 36. [Google Scholar] [CrossRef]
  307. Dias, S.J.; Li, K.; Rimando, A.M.; Dhar, S.; Mizuno, C.S.; Penman, A.D.; Levenson, A.S. Trimethoxy-Resveratrol and Piceatannol Administered Orally Suppress and Inhibit Tumor Formation and Growth in Prostate Cancer Xenografts. Prostate 2013, 73, 1135–1146. [Google Scholar] [CrossRef]
  308. Yokozawa, T.; Kim, Y.J. Piceatannol Inhibits Melanogenesis by Its Antioxidative Actions. Biol. Pharm. Bull. 2007, 30, 2007–2011. [Google Scholar] [CrossRef]
  309. Liu, W.-H.; Chang, L.-S. Piceatannol induces Fas and FasL up-regulation in human leukemia U937 cells via Ca2+/p38α MAPK-mediated activation of c-Jun and ATF-2 pathways. Int. J. Biochem. Cell Biol. 2010, 42, 1498–1506. [Google Scholar] [CrossRef]
  310. Takasawa, R.; Akahane, H.; Tanaka, H.; Shimada, N.; Yamamoto, T.; Uchida-Maruki, H.; Sai, M.; Yoshimori, A.; Tanuma, S.-i. Piceatannol, a natural trans-stilbene compound, inhibits human glyoxalase I. Bioorganic Med. Chem. Lett. 2017, 27, 1169–1174. [Google Scholar] [CrossRef] [PubMed]
  311. Wolter, F.; Clausnitzer, A.; Akoglu, B.; Stein, J. Piceatannol, a Natural Analog of Resveratrol, Inhibits Progression through the S Phase of the Cell Cycle in Colorectal Cancer Cell Lines. J. Nutr. 2002, 132, 298–302. [Google Scholar] [CrossRef]
  312. Ovesná, Z.; Kozics, K.; Bader, Y.; Saiko, P.; Handler, N.; Erker, T.; Szekeres, T. Antioxidant activity of resveratrol, piceatannol and 3,3′,4,4′,5,5′-hexahydroxy-trans-stilbene in three leukemia cell lines. Oncol. Rep. 2006, 16, 617–624. [Google Scholar] [CrossRef]
  313. Duarte, N.; Kayser, O.; Abreu, P.; Ferreira, M.-J.U. Antileishmanial activity of piceatannol isolated from Euphorbia lagascae seeds. Phytother. Res. 2008, 22, 455–457. [Google Scholar] [CrossRef]
  314. Chowdhury, S.A.; Kishino, K.; Satoh, R.; Hashimoto, K.; Kikuchi, H.; Nishikawa, H.; Shirataki, Y.; Sakagami, H. Tumor-specificity and Apoptosis-inducing Activity of Stilbenes and Flavonoids. Anticancer Res. 2005, 25, 2055–2063. [Google Scholar]
  315. Zhang, Y.; Gu, Y.; Xie, J.; Hu, Y. Anti-tumor effect of piceatannol through induction of cell apoptosis via up-regulation of microRNA-125b expression on pancreatic cancer. Int. J. Clin. Exp. Med. 2017, 10, 14495–14502. [Google Scholar]
  316. Siedlecka-Kroplewska, K.; Ślebioda, T.; Kmieć, Z. Induction of autophagy, apoptosis and aquisition of resistance in response to piceatannol toxicity in MOLT-4 human leukemia cells. Toxicol. Vitr. 2019, 59, 12–25. [Google Scholar] [CrossRef] [PubMed]
  317. Liu, T.; Liu, M.; Guo, Q.; Liu, Y.; Zhao, Y.; Wu, Y.; Sun, B.; Wang, Q.; Liu, J.; Han, J. Investigation of binary and ternary systems of human serum albumin with oxyresveratrol/piceatannol and/or mitoxantrone by multipectroscopy, molecular docking and cytotoxicity evaluation. J. Mol. Liq. 2020, 311, 113364. [Google Scholar] [CrossRef]
  318. Arai, D.; Kataoka, R.; Otsuka, S.; Kawamura, M.; Maruki-Uchida, H.; Sai, M.; Ito, T.; Nakao, Y. Piceatannol is superior to resveratrol in promoting neural stem cell differentiation into astrocytes. Food Funct. 2016, 7, 4432–4441. [Google Scholar] [CrossRef]
  319. Rüweler, M.; Gülden, M.; Maser, E.; Murias, M.; Seibert, H. Cytotoxic, cytoprotective and antioxidant activities of resveratrol and analogues in C6 astroglioma cells in vitro. Chem.-Biol. Interact. 2009, 182, 128–135. [Google Scholar] [CrossRef]
  320. Billack, B.; Radkar, V.; Adiabouah, C. In Vitro evaluation of the cytotoxic and anti-proliferative properties of resveratrol and several of its analogs. Cell. Mol. Biol. Lett. 2008, 13, 553–569. [Google Scholar] [CrossRef]
  321. Kang, C.-H.; Moon, D.-O.; Choi, Y.H.; Choi, I.-W.; Moon, S.-K.; Kim, W.-J.; Kim, G.-Y. Piceatannol enhances TRAIL-induced apoptosis in human leukemia THP-1 cells through Sp1- and ERK-dependent DR5 up-regulation. Toxicol. Vitr. 2011, 25, 605–612. [Google Scholar] [CrossRef] [PubMed]
  322. Rohaiza, S.; Yaacob, W.; Din, L.; Nazlina, I. Cytotoxic oligostilbenes from Shorea hopeifolia. Afr. J. Pharm. Pharmacol. 2011, 5, 1272–1277. [Google Scholar] [CrossRef]
  323. Chang, C.-I.; Chien, W.-C.; Huang, K.-X.; Hsu, J.-L. Anti-Inflammatory Effects of Vitisinol A and Four Other Oligostilbenes from Ampelopsis brevipedunculata var. Hancei. Molecules 2017, 22, 1195. [Google Scholar] [CrossRef]
  324. Ha, D.T.; Chen, Q.C.; Hung, T.M.; Youn, U.J.; Ngoc, T.M.; Thuong, P.T.; Kim, H.J.; Seong, Y.H.; Min, B.S.; Bae, K. Stilbenes and oligostilbenes from leaf and stem of Vitis amurensis and their cytotoxic activity. Arch. Pharmacal Res. 2009, 32, 177–183. [Google Scholar] [CrossRef]
  325. Vion, E.; Page, G.; Bourdeaud, E.; Paccalin, M.; Guillard, J.; Rioux Bilan, A. Trans ε-viniferin is an amyloid-β disaggregating and anti-inflammatory drug in a mouse primary cellular model of Alzheimer’s disease. Mol. Cell. Neurosci. 2018, 88, 1–6. [Google Scholar] [CrossRef]
  326. Tian, X.; Guo, S.; Zhang, S.; Li, P.; Wang, T.; Ho, C.-T.; Pan, M.-H.; Bai, N. Chemical characterization of main bioactive constituents in Paeonia ostii seed meal and GC-MS analysis of seed oil. J. Food Biochem. 2020, 44, e13088. [Google Scholar] [CrossRef] [PubMed]
  327. Medrano-Padial, C.; Puerto, M.; Merchán-Gragero, M.d.M.; Moreno, F.J.; Richard, T.; Cantos-Villar, E.; Pichardo, S. Cytotoxicity studies of a stilbene extract and its main components intended to be used as preservative in the wine industry. Food Res. Int. 2020, 137, 109738. [Google Scholar] [CrossRef] [PubMed]
  328. Kim, H.J.; Chang, E.J.; Bae, S.J.; Shim, S.M.; Park, H.D.; Rhee, C.H.; Park, J.H.; Choi, S.W. Cytotoxic and antimutagenic stilbenes from seeds ofPaeonia lactiflora. Arch. Pharmacal Res. 2002, 25, 293–299. [Google Scholar] [CrossRef]
  329. Kang, J.H.; Park, Y.H.; Choi, S.W.; Yang, E.K.; Lee, W.J. Resveratrol derivatives potently induce apoptosis in human promyelocytic leukemia cells. Exp. Mol. Med. 2003, 35, 467–474. [Google Scholar] [CrossRef]
  330. Zghonda, N.; Yoshida, S.; Araki, M.; Kusunoki, M.; Mliki, A.; Ghorbel, A.; Miyazaki, H. Greater Effectiveness of ε-Viniferin in Red Wine Than Its Monomer Resveratrol for Inhibiting Vascular Smooth Muscle Cell Proliferation and Migration. Biosci. Biotechnol. Biochem. 2011, 75, 1259–1267. [Google Scholar] [CrossRef]
  331. Courtois, A.; Garcia, M.; Krisa, S.; Atgié, C.; Sauvant, P.; Richard, T.; Faure, C. Encapsulation of ε-viniferin in onion-type multi-lamellar liposomes increases its solubility and its photo-stability and decreases its cytotoxicity on Caco-2 intestinal cells. Food Funct. 2019, 10, 2573–2582. [Google Scholar] [CrossRef]
  332. Zghonda, N.; Yoshida, S.; Ezaki, S.; Otake, Y.; Murakami, C.; Mliki, A.; Ghorbel, A.; Miyazaki, H. ε-Viniferin Is More Effective Than Its Monomer Resveratrol in Improving the Functions of Vascular Endothelial Cells and the Heart. Biosci. Biotechnol. Biochem. 2012, 76, 954–960. [Google Scholar] [CrossRef]
  333. Wu, C.W.; Nakamoto, Y.; Hisatome, T.; Yoshida, S.; Miyazaki, H. Resveratrol and its dimers ε-viniferin and δ-viniferin in red wine protect vascular endothelial cells by a similar mechanism with different potency and efficacy. Kaohsiung J. Med. Sci. 2020, 36, 535–542. [Google Scholar] [CrossRef]
  334. Huang, C.; Lin, Z.-J.; Lee, C.-J.; Lai, W.-H.; Chen, J.-C.; Huang, H.-C. ε-Viniferin and α-viniferin alone or in combination induced apoptosis and necrosis in osteosarcoma and non-small cell lung cancer cells. Food Chem. Toxicol. 2021, 158, 112617. [Google Scholar] [CrossRef]
  335. Richard, T.; Poupard, P.; Nassra, M.; Papastamoulis, Y.; Iglésias, M.-L.; Krisa, S.; Waffo-Teguo, P.; Mérillon, J.-M.; Monti, J.-P. Protective effect of ε-viniferin on β-amyloid peptide aggregation investigated by electrospray ionization mass spectrometry. Bioorganic Med. Chem. 2011, 19, 3152–3155. [Google Scholar] [CrossRef]
  336. Frombaum, M.; Therond, P.; Djelidi, R.; Beaudeux, J.-L.; Bonnefont-Rousselot, D.; Borderie, D. Piceatannol is more effective than resveratrol in restoring endothelial cell dimethylarginine dimethylaminohydrolase expression and activity after high-glucose oxidative stress. Free Radic. Res. 2011, 45, 293–302. [Google Scholar] [CrossRef] [PubMed]
  337. Woo, A.; Min, B.; Ryoo, S. Piceatannol-3′-O-β-D-glucopyranoside as an active component of rhubarb activates endothelial nitric oxide synthase through inhibition of arginase activity. Exp. Mol. Med. 2010, 42, 524–532. [Google Scholar] [CrossRef] [PubMed]
  338. Yuan, Q.; Peng, J.; Liu, S.-Y.; Wang, C.-J.; Xiang, D.-X.; Xiong, X.-M.; Hu, C.-P.; Li, Y.-J. Inhibitory effect of resveratrol derivative BTM-0512 on high glucose-induced cell senescence involves dimethylaminohydrolase/asymmetric dimethylarginine pathway. Clin. Exp. Pharmacol. Physiol. 2010, 37, 630–635. [Google Scholar] [CrossRef] [PubMed]
  339. Gómez-Zorita, S.; Fernández-Quintela, A.; Lasa, A.; Aguirre, L.; Rimando, A.M.; Portillo, M.P. Pterostilbene, a Dimethyl Ether Derivative of Resveratrol, Reduces Fat Accumulation in Rats Fed an Obesogenic Diet. J. Agric. Food Chem. 2014, 62, 8371–8378. [Google Scholar] [CrossRef]
  340. Spanier, G.; Xu, H.; Xia, N.; Tobias, S.; Deng, S.; Wojnowski, L.; Forstermann, U.; Li, H. Resveratrol reduces endothelial oxidative stress by modulating the gene expression of superoxide dismutase 1 (SOD1), glutathione peroxidase 1 (GPx1) and NADPH oxidase subunit (Nox4). J. Physiol. Pharmacol. 2009, 60, 111–116. [Google Scholar]
  341. Wallerath, T.; Deckert, G.; Ternes, T.; Anderson, H.; Li, H.; Witte, K.; Förstermann, U. Resveratrol, a Polyphenolic Phytoalexin Present in Red Wine, Enhances Expression and Activity of Endothelial Nitric Oxide Synthase. Circulation 2002, 106, 1652–1658. [Google Scholar] [CrossRef]
  342. Frombaum, M.; Le Clanche, S.; Bonnefont-Rousselot, D.; Borderie, D. Antioxidant effects of resveratrol and other stilbene derivatives on oxidative stress and NO bioavailability: Potential benefits to cardiovascular diseases. Biochimie 2012, 94, 269–276. [Google Scholar] [CrossRef]
  343. Yang, J.; Wang, N.; Li, J.; Zhang, J.; Feng, P. Effects of resveratrol on NO secretion stimulated by insulin and its dependence on SIRT1 in high glucose cultured endothelial cells. Endocrine 2010, 37, 365–372. [Google Scholar] [CrossRef]
  344. Arunachalam, G.; Yao, H.; Sundar, I.K.; Caito, S.; Rahman, I. SIRT1 regulates oxidant- and cigarette smoke-induced eNOS acetylation in endothelial cells: Role of resveratrol. Biochem. Biophys. Res. Commun. 2010, 393, 66–72. [Google Scholar] [CrossRef]
  345. Gresele, P.; Pignatelli, P.; Guglielmini, G.; Carnevale, R.; Mezzasoma, A.M.; Ghiselli, A.; Momi, S.; Violi, F. Resveratrol, at Concentrations Attainable with Moderate Wine Consumption, Stimulates Human Platelet Nitric Oxide Production123. J. Nutr. 2008, 138, 1602–1608. [Google Scholar] [CrossRef]
  346. Nicholson, S.K.; Tucker, G.A.; Brameld, J.M. Physiological concentrations of dietary polyphenols regulate vascular endothelial cell expression of genes important in cardiovascular health. Br. J. Nutr. 2009, 103, 1398–1403. [Google Scholar] [CrossRef] [PubMed]
  347. Klinger, J.R. The Nitric Oxide/cGMP Signaling Pathway in Pulmonary Hypertension. Clin. Chest Med. 2007, 28, 143–167. [Google Scholar] [CrossRef] [PubMed]
  348. Rivera, L.; Morón, R.; Zarzuelo, A.; Galisteo, M. Long-term resveratrol administration reduces metabolic disturbances and lowers blood pressure in obese Zucker rats. Biochem. Pharmacol. 2009, 77, 1053–1063. [Google Scholar] [CrossRef] [PubMed]
  349. Zou, J.-G.; Wang, Z.-R.; Huang, Y.-Z.; Cao, K.-J.; Wu, J.M. Effect of red wine and wine polyphenol resveratrol on endothelial function in hypercholesterolemic rabbits. Int. J. Mol. Med. 2003, 11, 317–320. [Google Scholar] [CrossRef]
  350. Yoshino, J.; Conte, C.; Fontana, L.; Mittendorfer, B.; Imai, S.I.; Schechtman, K.B.; Gu, C.; Kunz, I.; Fanelli, F.R.; Patterson, B.W.; et al. Resveratrol Supplementation Does Not Improve Metabolic Function in Nonobese Women with Normal Glucose Tolerance. Cell Metab. 2012, 16, 658–664. [Google Scholar] [CrossRef]
  351. Juan, M.E.; Vinardell, M.P.; Planas, J.M. The Daily Oral Administration of High Doses of trans-Resveratrol to Rats for 28 Days Is Not Harmful. J. Nutr. 2002, 132, 257–260. [Google Scholar] [CrossRef]
  352. Tomé-Carneiro, J.; Gonzálvez, M.; Larrosa, M.; Yáñez-Gascón, M.J.; García-Almagro, F.J.; Ruiz-Ros, J.A.; García-Conesa, M.T.; Tomás-Barberán, F.A.; Espín, J.C. One-Year Consumption of a Grape Nutraceutical Containing Resveratrol Improves the Inflammatory and Fibrinolytic Status of Patients in Primary Prevention of Cardiovascular Disease. Am. J. Cardiol. 2012, 110, 356–363. [Google Scholar] [CrossRef]
  353. Tomé-Carneiro, J.; Gonzálvez, M.; Larrosa, M.; García-Almagro, F.J.; Avilés-Plaza, F.; Parra, S.; Yáñez-Gascón, M.J.; Ruiz-Ros, J.A.; García-Conesa, M.T.; Tomás-Barberán, F.A.; et al. Consumption of a grape extract supplement containing resveratrol decreases oxidized LDL and ApoB in patients undergoing primary prevention of cardiovascular disease: A triple-blind, 6-month follow-up, placebo-controlled, randomized trial. Mol. Nutr. Food Res. 2012, 56, 810–821. [Google Scholar] [CrossRef]
  354. Sahebkar, A. Effects of resveratrol supplementation on plasma lipids: A systematic review and meta-analysis of randomized controlled trials. Nutr. Rev. 2013, 71, 822–835. [Google Scholar] [CrossRef] [PubMed]
  355. Riche, D.M.; Riche, K.D.; Blackshear, C.T.; McEwen, C.L.; Sherman, J.J.; Wofford, M.R.; Griswold, M.E. Pterostilbene on Metabolic Parameters: A Randomized, Double-Blind, and Placebo-Controlled Trial. Evid.-Based Complement. Altern. Med. 2014, 2014, 459165. [Google Scholar] [CrossRef] [PubMed]
  356. Tomé-Carneiro, J.; Gonzálvez, M.; Larrosa, M.; Yáñez-Gascón, M.J.; García-Almagro, F.J.; Ruiz-Ros, J.A.; Tomás-Barberán, F.A.; García-Conesa, M.T.; Espín, J.C. Grape Resveratrol Increases Serum Adiponectin and Downregulates Inflammatory Genes in Peripheral Blood Mononuclear Cells: A Triple-Blind, Placebo-Controlled, One-Year Clinical Trial in Patients with Stable Coronary Artery Disease. Cardiovasc. Drugs Ther. 2013, 27, 37–48. [Google Scholar] [CrossRef]
  357. Szkudelski, T.; Szkudelska, K. Resveratrol and diabetes: From animal to human studies. Biochim. Biophys. Acta (BBA)—Mol. Basis Dis. 2015, 1852, 1145–1154. [Google Scholar] [CrossRef] [PubMed]
  358. Penumathsa, S.V.; Thirunavukkarasu, M.; Zhan, L.; Maulik, G.; Menon, V.P.; Bagchi, D.; Maulik, N. Resveratrol enhances GLUT-4 translocation to the caveolar lipid raft fractions through AMPK/Akt/eNOS signalling pathway in diabetic myocardium. J. Cell. Mol. Med. 2008, 12, 2350–2361. [Google Scholar] [CrossRef]
  359. Moridi, H.; Karimi, J.; Sheikh, N.; Goodarzi, M.T.; Saidijam, M.; Yadegarazari, R.; Khazaei, M.; Khodadadi, I.; Tavilani, H.; Piri, H.; et al. Resveratrol-Dependent Down-regulation of Receptor for Advanced Glycation End-products and Oxidative Stress in Kidney of Rats With Diabetes. Int. J. Endocrinol. Metab. 2015, 13, e23542. [Google Scholar] [CrossRef]
  360. Soufi, F.G.; Mohammad-nejad, D.; Ahmadieh, H. Resveratrol improves diabetic retinopathy possibly through oxidative stress—Nuclear factor κB—Apoptosis pathway. Pharmacol. Rep. 2012, 64, 1505–1514. [Google Scholar] [CrossRef]
  361. Um, J.-H.; Park, S.-J.; Kang, H.; Yang, S.; Foretz, M.; McBurney, M.W.; Kim, M.K.; Viollet, B.; Chung, J.H. AMP-Activated Protein Kinase–Deficient Mice Are Resistant to the Metabolic Effects of Resveratrol. Diabetes 2009, 59, 554–563. [Google Scholar] [CrossRef] [PubMed]
  362. Bhatt, J.K.; Thomas, S.; Nanjan, M.J. Resveratrol supplementation improves glycemic control in type 2 diabetes mellitus. Nutr. Res. 2012, 32, 537–541. [Google Scholar] [CrossRef] [PubMed]
  363. Movahed, A.; Nabipour, I.; Lieben Louis, X.; Thandapilly, S.J.; Yu, L.; Kalantarhormozi, M.; Rekabpour, S.J.; Netticadan, T. Antihyperglycemic Effects of Short Term Resveratrol Supplementation in Type 2 Diabetic Patients. Evid.-Based Complement. Altern. Med. 2013, 2013, 851267. [Google Scholar] [CrossRef] [PubMed]
  364. Thazhath, S.S.; Wu, T.; Bound, M.J.; Checklin, H.L.; Standfield, S.; Jones, K.L.; Horowitz, M.; Rayner, C.K. Administration of resveratrol for 5 wk has no effect on glucagon-like peptide 1 secretion, gastric emptying, or glycemic control in type 2 diabetes: A randomized controlled trial12. Am. J. Clin. Nutr. 2016, 103, 66–70. [Google Scholar] [CrossRef]
  365. Bo, S.; Ponzo, V.; Ciccone, G.; Evangelista, A.; Saba, F.; Goitre, I.; Procopio, M.; Pagano, G.F.; Cassader, M.; Gambino, R. Six months of resveratrol supplementation has no measurable effect in type 2 diabetic patients. A randomized, double blind, placebo-controlled trial. Pharmacol. Res. 2016, 111, 896–905. [Google Scholar] [CrossRef]
  366. Gómez-Zorita, S.; Fernández-Quintela, A.; Aguirre, L.; Macarulla, M.T.; Rimando, A.M.; Portillo, M.P. Pterostilbene improves glycaemic control in rats fed an obesogenic diet: Involvement of skeletal muscle and liver. Food Funct. 2015, 6, 1968–1976. [Google Scholar] [CrossRef]
  367. Bhakkiyalakshmi, E.; Shalini, D.; Sekar, T.V.; Rajaguru, P.; Paulmurugan, R.; Ramkumar, K.M. Therapeutic potential of pterostilbene against pancreatic beta-cell apoptosis mediated through Nrf2. Br. J. Pharmacol. 2014, 171, 1747–1757. [Google Scholar] [CrossRef]
  368. Manickam, M.; Ramanathan, M.; Farboodniay Jahromi, M.A.; Chansouria, J.P.N.; Ray, A.B. Antihyperglycemic Activity of Phenolics from Pterocarpus marsupium. J. Nat. Prod. 1997, 60, 609–610. [Google Scholar] [CrossRef]
  369. Grover, J.K.; Vats, V.; Yadav, S.S. Pterocarpus marsupium extract (Vijayasar) prevented the alteration in metabolic patterns induced in the normal rat by feeding an adequate diet containing fructose as sole carbohydrate. Diabetes Obes. Metab. 2005, 7, 414–420. [Google Scholar] [CrossRef]
  370. Nemes-Nagy, E.; Szőcs-Molnár, T.; Dunca, I.; Balogh-Sămărghiţan, V.; Hobai, Ş.; Morar, R.; Pusta, D.L.; Crăciun, E.C. Effect of a dietary supplement containing blueberry and sea buckthorn concentrate on antioxidant capacity in type 1 diabetic children. Acta Physiol. Hung. 2008, 95, 383–393. [Google Scholar] [CrossRef]
  371. Minakawa, M.; Miura, Y.; Yagasaki, K. Piceatannol, a resveratrol derivative, promotes glucose uptake through glucose transporter 4 translocation to plasma membrane in L6 myocytes and suppresses blood glucose levels in type 2 diabetic model db/db mice. Biochem. Biophys. Res. Commun. 2012, 422, 469–475. [Google Scholar] [CrossRef] [PubMed]
  372. Towler, M.C.; Hardie, D.G. AMP-Activated Protein Kinase in Metabolic Control and Insulin Signaling. Circ. Res. 2007, 100, 328–341. [Google Scholar] [CrossRef]
  373. Saltiel, A.R.; Kahn, C.R. Insulin signalling and the regulation of glucose and lipid metabolism. Nature 2001, 414, 799–806. [Google Scholar] [CrossRef]
  374. Hijona, E.; Aguirre, L.; Pérez-Matute, P.; Villanueva-Millán, M.J.; Mosqueda-Solis, A.; Hasnaoui, M.; Nepveu, F.; Senard, J.M.; Bujanda, L.; Aldámiz-Echevarría, L.; et al. Limited beneficial effects of piceatannol supplementation on obesity complications in the obese Zucker rat: Gut microbiota, metabolic, endocrine, and cardiac aspects. J. Physiol. Biochem. 2016, 72, 567–582. [Google Scholar] [CrossRef]
  375. Oritani, Y.; Okitsu, T.; Nishimura, E.; Sai, M.; Ito, T.; Takeuchi, S. Enhanced glucose tolerance by intravascularly administered piceatannol in freely moving healthy rats. Biochem. Biophys. Res. Commun. 2016, 470, 753–758. [Google Scholar] [CrossRef]
  376. Kershaw, J.; Kim, K.-H. The Therapeutic Potential of Piceatannol, a Natural Stilbene, in Metabolic Diseases: A Review. J. Med. Food 2017, 20, 427–438. [Google Scholar] [CrossRef]
  377. Spalding, K.L.; Arner, E.; Westermark, P.O.; Bernard, S.; Buchholz, B.A.; Bergmann, O.; Blomqvist, L.; Hoffstedt, J.; Näslund, E.; Britton, T.; et al. Dynamics of fat cell turnover in humans. Nature 2008, 453, 783–787. [Google Scholar] [CrossRef]
  378. Jeong, S.O.; Son, Y.; Lee, J.H.; Cheong, Y.K.; Park, S.H.; Chung, H.T.; Pae, H.O. Resveratrol analog piceatannol restores the palmitic acid-induced impairment of insulin signaling and production of endothelial nitric oxide via activation of anti-inflammatory and antioxidative heme oxygenase-1 in human endothelial cells. Mol. Med. Rep. 2015, 12, 937–944. [Google Scholar] [CrossRef] [PubMed]
  379. Jellinger, K.A. General Aspects of Neurodegeneration; Spring: Vienna, Austria, 2003; pp. 101–144. [Google Scholar]
  380. de A Boleti, A.P.; Almeida, J.A.; Migliolo, L. Impact of the metabolic syndrome on the evolution of neurodegenerative diseases. Neural Regen. Res. 2021, 16, 688–689. [Google Scholar] [CrossRef] [PubMed]
  381. Pisoschi, A.M.; Pop, A.; Iordache, F.; Stanca, L.; Predoi, G.; Serban, A.I. Oxidative stress mitigation by antioxidants-an overview on their chemistry and influences on health status. Eur. J. Med. Chem. 2021, 209, 112891. [Google Scholar] [CrossRef]
  382. de Araújo, F.F.; de Paulo Farias, D.; Neri-Numa, I.A.; Pastore, G.M. Polyphenols and their applications: An approach in food chemistry and innovation potential. Food Chem. 2021, 338, 127535. [Google Scholar] [CrossRef] [PubMed]
  383. Olajide, O.A.; Sarker, S.D. Alzheimer’s disease: Natural products as inhibitors of neuroinflammation. Inflammopharmacology 2020, 28, 1439–1455. [Google Scholar] [CrossRef] [PubMed]
  384. Carradori, S.; D’Ascenzio, M.; Chimenti, P.; Secci, D.; Bolasco, A. Selective MAO-B inhibitors: A lesson from natural products. Mol. Divers. 2014, 18, 219–243. [Google Scholar] [CrossRef]
  385. Rahman, M.M.; Rahaman, M.S.; Islam, M.R.; Rahman, F.; Mithi, F.M.; Alqahtani, T.; Almikhlafi, M.A.; Alghamdi, S.Q.; Alruwaili, A.S.; Hossain, M.S.; et al. Role of Phenolic Compounds in Human Disease: Current Knowledge and Future Prospects. Molecules 2022, 27, 233. [Google Scholar] [CrossRef]
  386. Fantacuzzi, M.; Amoroso, R.; Carradori, S.; De Filippis, B. Resveratrol-based compounds and neurodegeneration: Recent insight in multitarget therapy. Eur. J. Med. Chem. 2022, 233, 114242. [Google Scholar] [CrossRef] [PubMed]
  387. Mohd Sairazi, N.S.; Sirajudeen, K.N.S. Natural Products and Their Bioactive Compounds: Neuroprotective Potentials against Neurodegenerative Diseases. Evid.-Based Complement. Altern. Med. 2020, 2020, 6565396. [Google Scholar] [CrossRef]
  388. Lu, K.-T.; Ko, M.-C.; Chen, B.-Y.; Huang, J.-C.; Hsieh, C.-W.; Lee, M.-C.; Chiou, R.Y.Y.; Wung, B.-S.; Peng, C.-H.; Yang, Y.-L. Neuroprotective Effects of Resveratrol on MPTP-Induced Neuron Loss Mediated by Free Radical Scavenging. J. Agric. Food Chem. 2008, 56, 6910–6913. [Google Scholar] [CrossRef]
  389. Yang, Y.; Fan, C.; Wang, B.; Ma, Z.; Wang, D.; Gong, B.; Di, S.; Jiang, S.; Li, Y.; Li, T.; et al. Pterostilbene attenuates high glucose-induced oxidative injury in hippocampal neuronal cells by activating nuclear factor erythroid 2-related factor 2. Biochim. Biophys. Acta (BBA)—Mol. Basis Dis. 2017, 1863, 827–837. [Google Scholar] [CrossRef]
  390. Wang, B.; Liu, H.; Yue, L.; Li, X.; Zhao, L.; Yang, X.; Wang, X.; Yang, Y.; Qu, Y. Neuroprotective effects of pterostilbene against oxidative stress injury: Involvement of nuclear factor erythroid 2-related factor 2 pathway. Brain Res. 2016, 1643, 70–79. [Google Scholar] [CrossRef]
  391. Su, Q.; Pu, H.; Hu, C. Neuroprotection by combination of resveratrol and enriched environment against ischemic brain injury in rats. Neurol. Res. 2016, 38, 60–68. [Google Scholar] [CrossRef]
  392. Fang, L.; Gao, H.; Zhang, W.; Zhang, W.; Wang, Y. Resveratrol alleviates nerve injury after cerebral ischemia and reperfusion in mice by inhibiting inflammation and apoptosis. Int. J. Clin. Exp. Med. 2015, 8, 3219. [Google Scholar]
  393. Teka, T.; Zhang, L.; Ge, X.; Li, Y.; Han, L.; Yan, X. Stilbenes: Source plants, chemistry, biosynthesis, pharmacology, application and problems related to their clinical Application-A comprehensive review. Phytochemistry 2022, 197, 113128. [Google Scholar] [CrossRef]
  394. Zhang, F.; Shi, J.-S.; Zhou, H.; Wilson, B.; Hong, J.-S.; Gao, H.-M. Resveratrol Protects Dopamine Neurons Against Lipopolysaccharide-Induced Neurotoxicity through Its Anti-Inflammatory Actions. Mol. Pharmacol. 2010, 78, 466–477. [Google Scholar] [CrossRef]
  395. Marambaud, P.; Zhao, H.; Davies, P. Resveratrol promotes clearance of Alzheimer’s disease amyloid-beta peptides. J. Biol. Chem. 2005, 280, 37377–37382. [Google Scholar] [CrossRef] [PubMed]
  396. Biais, B.; Krisa, S.; Cluzet, S.; Da Costa, G.; Waffo-Teguo, P.; Mérillon, J.-M.; Richard, T. Antioxidant and Cytoprotective Activities of Grapevine Stilbenes. J. Agric. Food Chem. 2017, 65, 4952–4960. [Google Scholar] [CrossRef] [PubMed]
  397. Chaher, N.; Arraki, K.; Dillinseger, E.; Temsamani, H.; Bernillon, S.; Pedrot, E.; Delaunay, J.-C.; Mérillon, J.-M.; Monti, J.-P.; Izard, J.-C.; et al. Bioactive stilbenes from Vitis vinifera grapevine shoots extracts. J. Sci. Food Agric. 2014, 94, 951–954. [Google Scholar] [CrossRef]
  398. Ren, J.; Fan, C.; Chen, N.; Huang, J.; Yang, Q. Resveratrol Pretreatment Attenuates Cerebral Ischemic Injury by Upregulating Expression of Transcription Factor Nrf2 and HO-1 in Rats. Neurochem. Res. 2011, 36, 2352–2362. [Google Scholar] [CrossRef]
  399. Simão, F.; Matté, A.; Pagnussat, A.S.; Netto, C.A.; Salbego, C.G. Resveratrol prevents CA1 neurons against ischemic injury by parallel modulation of both GSK-3β and CREB through PI3-K/Akt pathways. Eur. J. Neurosci. 2012, 36, 2899–2905. [Google Scholar] [CrossRef]
  400. Ma, X.; Sun, Z.; Liu, Y.; Jia, Y.; Zhang, B.; Zhang, J. Resveratrol improves cognition and reduces oxidative stress in rats with vascular dementia. Neural Regen. Res. 2013, 8, 2050–2059. [Google Scholar]
  401. Ban, J.Y.; Jeon, S.-Y.; Nguyen, T.T.H.; Bae, K.; Song, K.-S.; Seonga, Y.H. Neuroprotective Effect of Oxyresveratrol from Smilacis Chinae Rhizome on Amyloid Beta Protein (25–35)-Induced Neurotoxicity in Cultured Rat Cortical Neurons. Biol. Pharm. Bull. 2006, 29, 2419–2424. [Google Scholar] [CrossRef]
  402. Andrabi, S.A.; Spina, M.G.; Lorenz, P.; Ebmeyer, U.; Wolf, G.; Horn, T.F.W. Oxyresveratrol (trans-2,3′,4,5′-tetrahydroxystilbene) is neuroprotective and inhibits the apoptotic cell death in transient cerebral ischemia. Brain Res. 2004, 1017, 98–107. [Google Scholar] [CrossRef]
  403. Turner, R.S.; Thomas, R.G.; Craft, S.; van Dyck, C.H.; Mintzer, J.; Reynolds, B.A.; Brewer, J.B.; Rissman, R.A.; Raman, R.; Aisen, P.S.; et al. A randomized, double-blind, placebo-controlled trial of resveratrol for Alzheimer disease. Neurology 2015, 85, 1383–1391. [Google Scholar] [CrossRef] [PubMed]
  404. Witte, A.V.; Kerti, L.; Margulies, D.S.; Flöel, A. Effects of Resveratrol on Memory Performance, Hippocampal Functional Connectivity, and Glucose Metabolism in Healthy Older Adults. J. Neurosci. 2014, 34, 7862–7870. [Google Scholar] [CrossRef] [PubMed]
  405. Wong, R.H.X.; Raederstorff, D.; Howe, P.R.C. Acute Resveratrol Consumption Improves Neurovascular Coupling Capacity in Adults with Type 2 Diabetes Mellitus. Nutrients 2016, 8, 425. [Google Scholar] [CrossRef]
  406. Wong, R.H.X.; Nealon, R.S.; Scholey, A.; Howe, P.R.C. Low dose resveratrol improves cerebrovascular function in type 2 diabetes mellitus. Nutr. Metab. Cardiovasc. Dis. 2016, 26, 393–399. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic of the grapevine: stilbenes are primarily located in the wood, roots, branches, and stems, where they help prevent wood degradation and play defensive roles. These molecules exhibit structural variability, which determines their chemical behavior and bioactivity.
Figure 1. Schematic of the grapevine: stilbenes are primarily located in the wood, roots, branches, and stems, where they help prevent wood degradation and play defensive roles. These molecules exhibit structural variability, which determines their chemical behavior and bioactivity.
Ijms 26 08269 g001
Figure 2. Flow diagram of stilbenes’ biosynthesis.
Figure 2. Flow diagram of stilbenes’ biosynthesis.
Ijms 26 08269 g002
Figure 3. Diagram of mechanisms of biological activities of stilbenes.
Figure 3. Diagram of mechanisms of biological activities of stilbenes.
Ijms 26 08269 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Brás, L.P.; Luís, Â.; Chatel, G.; Socorro, S.; Duarte, A.P. Stilbenes from Vine Extracts: Therapeutic Potential and Mechanisms. Int. J. Mol. Sci. 2025, 26, 8269. https://doi.org/10.3390/ijms26178269

AMA Style

Brás LP, Luís Â, Chatel G, Socorro S, Duarte AP. Stilbenes from Vine Extracts: Therapeutic Potential and Mechanisms. International Journal of Molecular Sciences. 2025; 26(17):8269. https://doi.org/10.3390/ijms26178269

Chicago/Turabian Style

Brás, Luís P., Ângelo Luís, Gregory Chatel, Sílvia Socorro, and Ana Paula Duarte. 2025. "Stilbenes from Vine Extracts: Therapeutic Potential and Mechanisms" International Journal of Molecular Sciences 26, no. 17: 8269. https://doi.org/10.3390/ijms26178269

APA Style

Brás, L. P., Luís, Â., Chatel, G., Socorro, S., & Duarte, A. P. (2025). Stilbenes from Vine Extracts: Therapeutic Potential and Mechanisms. International Journal of Molecular Sciences, 26(17), 8269. https://doi.org/10.3390/ijms26178269

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop