Next Article in Journal
Correlation between Serum 25-Hydroxyvitamin D Concentration, Monocyte-to-HDL Ratio and Acute Coronary Syndrome in Men with Chronic Coronary Syndrome—An Observational Study
Previous Article in Journal
Anti-Menopausal Effect of Soybean Germ Extract and Lactobacillus gasseri in the Ovariectomized Rat Model
Previous Article in Special Issue
Are We Ready to Recommend Capsaicin for Disorders Other Than Neuropathic Pain?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Pharmacological Properties of Red Grape Polyphenol Resveratrol: Clinical Trials and Obstacles in Drug Development

1
Department of Basic Sciences, Preparatory Year Deanship, King Faisal University, Al Ahsa 31982, Saudi Arabia
2
Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, India
*
Author to whom correspondence should be addressed.
Nutrients 2023, 15(20), 4486; https://doi.org/10.3390/nu15204486
Submission received: 27 September 2023 / Revised: 18 October 2023 / Accepted: 20 October 2023 / Published: 23 October 2023
(This article belongs to the Special Issue The Action of Bioactive Compounds on Human Health or Disease)

Abstract

:
Resveratrol is a stilbenoid from red grapes that possesses a strong antioxidant activity. Resveratrol has been shown to have anticancer activity, making it a promising drug for the treatment and prevention of numerous cancers. Several in vitro and in vivo investigations have validated resveratrol’s anticancer capabilities, demonstrating its ability to block all steps of carcinogenesis (such as initiation, promotion, and progression). Additionally, resveratrol has been found to have auxiliary pharmacological effects such as anti-inflammatory, cardioprotective, and neuroprotective activity. Despite its pharmacological properties, several obstacles, such as resveratrol’s poor solubility and bioavailability, as well as its adverse effects, continue to be key obstacles to drug development. This review critically evaluates the clinical trials to date and aims to develop a framework to develop resveratrol into a clinically viable drug.

1. Introduction

The natural polyphenol resveratrol (trans-3,5,4′-trihydroxystilbene) is a stilbenoid. In 1939, Takaoka was the first to successfully extract resveratrol from Veratrum grandiflorum. [1,2]. The skin of red grapes contains the highest concentration of resveratrol. It has also been shown that certain foods, including tea, blueberries, pomegranates, almonds, pistachios, and dark chocolate, contain resveratrol (Figure 1) [3,4].
Resveratrol comprises a phenol ring connected to a catechol by an ethylene bridge. Two isomeric variants of resveratrol, cis- and trans-resveratrol, can be distinguished based on their chemical structure (Figure 2). The trans form predominates in terms of frequency, and it has been attributed to a variety of biological actions, including the induction of cell cycle arrest, differentiation, apoptosis, and the inhibition of the proliferation of cancer cells [3,4,5].
Resveratrol has been shown to have a broad range of therapeutic effects, such as its anti-inflammatory, antioxidant, platelet-inhibiting, hyperlipidemic, immunomodulatory, anti-carcinogenic, cardioprotective, vasodilatory, and neuroprotective activity [5,6,7,8,9]. It has been reported that resveratrol can sustain or improve human cerebrovascular functions [10], modulate in vitro angiogenesis by altering vascular endothelial growth factor (VEGF) expression and the formation of new vascular networks [11], stimulate human immune cell functions [12], boost rat cell viability and proliferation [13], reduce mitochondrial respiratory dysfunction, and boost cellular reprogramming in human fibroblasts derived from patients with a mammalian target of rapamycin (mTOR) pathway deficiency [14]. Resveratrol has been shown to also have neuroprotective [15], hepatoprotective [16], and cardioprotective [17,18] effects. In particular, the polyphenol appears to ameliorate the main risk factors of cardiovascular diseases (CVDs) because it can enhance endothelial function, scavenge reactive oxygen species (ROS), reduce inflammation, inhibit platelet aggregation, and improve the lipid profile, among other things [19,20]. In addition, redox-associated mechanisms were suggested as potential mechanisms through which resveratrol exerts its cardioprotective effects. These redox-associated mechanisms include the maintenance of mitochondrial function during hypoxia/reoxygenation-induced oxidative stress [21], the overexpression of antioxidant enzymes, like peroxidase and superoxide dismutase (SOD) [22], and the regulation of nitric oxide (NO) generation [23].
Resveratrol has been demonstrated to be safe for human consumption in several studies [24,25], although there have also been reports of harmful effects of resveratrol in vitro and in vivo [26]. For instance, when resveratrol was given in large quantities, it showed systemic suppression of P450 cytochromes [2]. Several pharmacological interactions involving resveratrol were also discovered and because of the potential for these interactions to reduce the efficacy of the medication, they are considered hazardous [27]. High-dose resveratrol has hormetic effects in vitro (micromolar range in cell culture media) and in vivo (nanomolar range in the blood) [28,29,30], including pro-oxidant effects [29,30,31,32,33,34], but it also has toxic side effects, such as disrupting the thyroid and causing goiter if used for an extended period of time. Thus, it is important to identify the actual biologically effective concentration range at which this compound should be supplemented in human subjects [35,36]. Further studies on pharmacological interactions would allow researchers to address these interactions and understand their cost-to-benefit ratio.

Resveratrol Bioavailability and Metabolism

There are a number of obstacles in the way of resveratrol being used commercially as a pharmaceutical agent, the most significant of which include resveratrol’s limited bioavailability and quick metabolism. These two factors may reduce resveratrol’s [2,37] effects in vivo. With only a few traces of un-metabolized resveratrol detectable in the plasma after an oral dose of 25 mg [37], it is clear that resveratrol has a very limited bioavailability in the body. Although more than 70% of resveratrol is absorbed in the digestive tract, it is rapidly consumed through three separate metabolic routes after ingestion. According to recent research [38], the bioavailability of resveratrol is determined by its extremely quick sulfate conjugation in the intestine/liver (Figure 3).
The fact that resveratrol is only marginally soluble in water (0.05 mg/mL) hinders its absorption. pH and temperature have significant effects on the stability and solubility of resveratrol [39]. In this context, studies found that the solubility of resveratrol is 64 μg/mL at a pH of 1.2, 61 μg/mL at a pH of 6.8, and 50 μg/mL above a pH of 7.4. Once dissolved in water, resveratrol is only stable at ambient temperature or body temperature under acidic conditions; at higher pH levels, the stilbene is destroyed at an exponential rate. We can infer that resveratrol is most stable in its liquid state when kept at a low pH, cool temperature, and away from oxygen and light [39].
After being ingested, resveratrol moves through the body either through passive diffusion or by forming complexes with transporters such as integrins, albumin, and low-density lipoprotein (LDL) [2,40,41]. Although resveratrol seems to be stable in the stomach’s acidic environment, it may be hydrolyzed to oligomeric phenolics or undergo isomeric conversion. In addition, resveratrol’s glycosylation by resident bacteria in the stomach can result in the absorption-competent stilbenoid glucoside piceid (resveratrol-3-O-beta-glucoside) [2,42]. Intestinal and hepatic conjugation processes also contribute to resveratrol modification. Benzoic, phenylacetic, and propionic acids can be metabolized from resveratrol by intestinal bacteria, whereas phase II metabolism in the liver results in glucuronidated, sulfated, and methylated metabolites that are known to retain some of the biological activity of the parent chemical [2,41,43,44].
The affinity of resveratrol for transport proteins is also connected to its biological effects in vivo. There is a lot of evidence that resveratrol can form complexes with plasma transport proteins, such as human serum albumin (HSA) and lipoproteins, which promote resveratrol stability and activity [45,46,47,48]. To enter various tissues, resveratrol forms complexes with HSA [49,50]. HSA is required in circulation to bind resveratrol, transport it, enhance its uptake by cells, and redistribute it to different cell types [2,48]. In this regard, it has previously been established that epigallocatechin gallate (EGCG), another naturally occurring polyphenolic antioxidant from green tea, can also be bound and stabilized by HSA under aqueous physiological conditions. Consequently, HSA and other plasma proteins may play a pivotal role in mediating the in vivo physiologic effects of resveratrol. Dihydro-resveratrol glucuronides, resveratrol glucuronides, and glucosides are all metabolites of resveratrol, and it is known that resveratrol induces its own metabolism, which raises the activity of phase II hepatic detoxifying enzymes. High levels of these metabolites are detected in human plasma and urine [51,52]. This suggests that free resveratrol may be released locally from these metabolites, as its half-life and plasma concentrations were shown to be 10 times higher than the natural resveratrol component [2,44,53].

2. Resveratrol: Pharmacology and Therapeutic Potential

Some of the most prominent biological actions of resveratrol and its therapeutic potential are summarized in Figure 4 and Table 1. The subsequent sub-sections will elucidate the pharmacological effects associated with moderate consumption of resveratrol with special reference to anti-diabetic effects, cardiovascular effects, neuroprotective functions, and anticancer properties.

2.1. Resveratrol in Cardiovascular Health

Heart disease and stroke are the leading causes of death and disability in developed nations [84]. Atherosclerosis is the leading cause of cardiovascular disorders affecting the coronary arteries. Light to moderate alcohol use has been linked to a lower risk of developing type 2 diabetes, increased HDL cholesterol, and decreased lipid oxidative stress. Red wine is superior to other alcoholic beverages in lowering the risk of developing coronary heart disease. It is possible that the synergistic effects of both resveratrol and alcohol come into play in such an action [85].
Resveratrol has been shown in preclinical trials to inhibit LDL oxidation [86]. The effect of red wine on cholesterol is multifaceted, with resveratrol playing a role in hepatic cholesterol and lipoprotein metabolism. The process lowers plasma cholesterol by decreasing cholesterol absorption and its transportation to the liver. Additional effects of resveratrol on cardiovascular disease risk variables include upregulating lipoprotein lipase activity and downregulating low-density lipoprotein circulation [87]. Resveratrol also affects apolipoproteins A and B. In a study, researchers looked at how moderate consumption of red wine, dealcoholized red wine, and gin affected glucose metabolism and lipid profile [88]. Sixty-seven males with a high cardiovascular risk were enrolled in the trial. For four weeks, everyone received 30 g of alcohol each day, which is the same as a standard glass of dealcoholized red wine. A decrease in the Homeostatic Model Assessment for Insulin Resistance (HOMA-IR) and mean-adjusted plasma insulin were observed following wine and dealcoholized wine consumption, while increases in high-density lipoprotein cholesterol, apolipoprotein A-I, and A-II were observed following gin and red wine consumption, and a decrease in lipoprotein was observed following red wine consumption [88]. Paraoxonase 1 is a hydrolytic enzyme that contributes to the protective functions of high-density lipoprotein. A moderate intake of red wine was found to positively alter paraoxonase 1 activity in a healthy Mexican population [89]. A group of researchers [90] looked at the phenomenon of subclinical coronary atherosclerosis. Carotid and femoral artery plaque were measured in this predominantly male sample following polyphenol consumption. Both femoral and carotid subclinical atherosclerosis risk decreased in correlation with increased consumption of flavonoids, while femoral subclinical atherosclerosis risk decreased in correlation with increased consumption of stilbenes. Red wine polyphenols were studied for their potential to counteract age-related declines in vascular function and physical exercise capacity in rats of varying ages (12, 20, and 40 weeks). From week 16 through week 40, rats were treated with either red wine polyphenols or apocynin (an antioxidant and NDPH oxidase inhibitor). Both supplements were found to be effective in reducing endothelial dysfunction, oxidative stress, and abnormal protein expression. Finally, polyphenols in red wine protect against endothelium dysfunction that comes with aging [86]. Seventeen dyslipidaemic postmenopausal women were studied to determine the effects of acute ingestion of red wine and dealcoholized red wine on postprandial lipid and lipoprotein metabolism [86]. Over a six-hour period, acute consumption worsened postprandial lipaemia and increased insulin secretion, but it had no effect on postprandial triglyceride, chylomicrons, or insulin homeostasis. Therefore, it is reasonable to anticipate that long-term use of resveratrol may be good for cardiovascular health [86]. In a particular study, it was shown that moderate consumption of red wine among an elderly population with high cardiovascular risk was associated with a decreased likelihood of metabolic syndrome, abnormal waist circumference, low concentrations of high-density lipoprotein cholesterol, high blood pressure, and hyperglycemia, in comparison to individuals who did not consume red wine [91].

2.2. Resveratrol for the Treatment and Prevention of Cancer

Cancer is a leading cause of death all over the world. Each year, it impacts over 6 million people [86]. Chemoprevention is promising for preventing cancer by utilizing either natural or synthetic drugs, or a combination of the two [92]. Resveratrol present in food and drink is thought to be responsible for lowering cancer risk. Stilbenes have been shown to prevent cancer in cell cultures and animals exposed to cancer cells or carcinogenic substances [86]. Colorectal cancer is the third most common kind of cancer, affecting an estimated 1.8 million individuals annually. In most cases, oncogenic mutations accumulate over time in non-cancerous polyps in the intestinal epithelium lining the colon or rectum. If these benign polyps are not caught early enough, they can develop into malignant adenomatous polyps. This development is significantly influenced by environmental factors like nutrition, smoking, alcohol usage, and inactivity. Several studies [93,94] point to the importance of a healthy diet (such as the Mediterranean diet) as a preventative measure against numerous diseases. Cancer prevention is aided by eating foods high in polyphenols and monounsaturated fats, such as those found in the Mediterranean diet [95].
Resveratrol has been investigated for its apoptotic effects on human colon cancer cells (SNU-C4) [96]. Through chromatin condensation and apoptotic body formation, the results demonstrated that resveratrol (100 g/mL) promoted apoptosis in SNU-C4 cells. Resveratrol was found to decrease Bcl-2 expression while increasing Bax and Caspase-3 expression compared to a control group [86]. In order to prevent colon cancer in animals, scientists looked into resveratrol-rich plant extracts like those found in red wine, pomegranate, white grape, and rosemary [97]. Workshop-made cured pork, which is known to promote colon carcinogenesis, had the extracts added to it. Both normal rats and rats provoked by azoxymethane received supplements for a total of 100 days. The number of mucin-depleted foci per colon was found to decrease in response to dry red wine, pomegranate extract, and tocopherol. Incorporating these extracts into cured meat has been proposed as a means of lowering the risk of colorectal cancer associated with eating processed meat [86,97]. It was also determined whether or not red wine extracts were effective in inhibiting the growth of colon cancer cells in vitro and colonic aberrant crypt foci in vivo [98]. Red wine extracts with greater anti-proliferative activity were examined in cells, and the ability to inhibit the development of aberrant crypt foci in mice was found to be the product of a lengthy vinification procedure. Synergistic anti-proliferative effects were also observed between quercetin and trans-resveratrol [98].

2.3. Resveratrol in Diabetes

The scientific community is becoming increasingly interested in substances that may have anti-diabetic effects. There is hope that such molecules can serve as the foundation for future therapeutic and preventative pharmaceuticals [99]. According to the World Health Organization, almost 500 million individuals will have diabetes mellitus by the year 2025. This condition is part of a more complex metabolic syndrome. Retinal, renal, limb, cardiac, nervous system, and vascular malfunctions, as well as compromised quality of life, and ultimately death, are all associated with this condition [86]. The risk of developing type 2 diabetes is reduced with moderate wine drinking, according to a number of studies [86,100].
In animal studies, simulating type 1 diabetes, a wine concentrate supplemented with natural polyphenols reduced hyperglycemia, brought hemoglobin and erythrocyte counts back to normal, and increased cell survival. Treatment with wine concentrate decreased the activity of catalase and glutathione peroxidase and raised the activity of superoxide dismutase in the plasma of rats with experimental diabetes mellitus [99]. In vitro research [101] looked into the potential anti-diabetic effects of Portuguese red wine. The results demonstrated that both the dealcoholized red wine and the four fractions of red wine produced through solid-phase extraction exhibited potent inhibitory effects against amylase and glucosidase. Monomeric and oligomeric flavan-3-ol molecules are primarily responsible for these actions [86,101]. Researchers examined the effects of co-digesting red wine with models of glucose and whey protein on the digestion, bioavailability, and colonic metabolism of the wine’s polyphenols and constituents. The most significant finding was a decrease in glucose bioaccessibility, which provides more evidence that moderate wine drinking has hypoglycemic effects. Additionally, protein breakdown was slowed, and short-chain fatty acid synthesis (particularly butyric acid) was elevated [102].

2.4. Resveratrol in Neuroprotection

The neuroprotective effects of resveratrol have been the subject of multiple investigations. Pretreatment with resveratrol protected neural stem cells from oxygen–glucose deprivation and activated nuclear factor erythroid 2-related factor 2 (Nrf2) [103]. Piceatoannol, a resveratrol metabolite, prevented glutamate-induced cell death in HT22 neuronal cells [104]. When resveratrol was given to rats, the pre-induction of cerebral ischemia led to the rats’ oxidation indicators dropping, and their superoxide dismutase activity was restored [105]. Glutathione peroxidase and glutathione reductase are necessary for maintaining glutathione in a reduced state. Drinking red wine boosted the enzymes of glutathione metabolism [106]. Red wine powder (freeze-dried with maltodextrin and gum arabic) [107] protects human neuroblastoma SH-SY5Y cell viability when treated with 6-hydroxydopamine. Indeed, red wine powder at a concentration of 150 ng GAE/mL ensured that 88.3% of cells would survive after being exposed to 6-hydroxydopamine cytotoxicity. Polyphenols with numerous hydroxyl groups are effective at preventing the production of mono- and di-adducts that contribute to the formation of advanced glycation end products. This is an effective means of protecting against neurodegenerative disorders [108]. Some of the possible pathways for resveratrol action in different disorders are summarized in Figure 5.

3. Human Clinical Trials of Resveratrol

Resveratrol shows promise as a compound that can help cells keep its metabolic balance. Researchers conducted numerous clinical investigations to verify the therapeutic effects of resveratrol on vascular metabolic illnesses in order to better understand its clinical transformative value. This resulted in 244 finished clinical trials and 27 ongoing trials by the end of 2019 [109]. Numerous diseases and disorders, such as diabetes, obesity, cancer, neurological, and cardiovascular diseases, have been the focus of clinical trials investigating the preventive and therapeutic effects of resveratrol. Preclinical and clinical studies have shown that resveratrol can modulate a wide variety of signaling molecules, including wingless-related integration site (Wnt), nuclear factor-kB (NF-kB), cytokines, caspases, Notch, matrix metalloproteinases (MMPs), 5′-AMP-activated protein kinase (AMPK), intercellular adhesion molecule (ICAM), vascular cell adhesion molecule (VCAM), sirtuin type 1 (SIRT1), tumor necrosis factor α (TNF-α), peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α), insulin-like growth factor 1 (IGF-1), insulin-like growth factor-binding protein (IGFBP-3), ras association domain family 1 isoform A (RASSF-1α), pAkt, vascular endothelial growth factor (VEGF), cyclooxygenase 2 (COX-2), nuclear factor erythroid 2-related factor 2 (Nrf2), and Kelch-like ECH-associated protein 1 (Keap1) [109,110]. The ability of resveratrol to interact with numerous targets, such as kinases, receptors, and signaling molecules, is the likely explanation for its pleiotropic behavior [109,110]. The major resveratrol clinical trials are listed below in Table 2.

4. Resveratrol as an Adjuvant

Contradictory findings in in vivo investigations on resveratrol have been reported, and this discrepancy has been linked to the drug’s low bioavailability, On the other hand, supplementary treatment can help with resveratrol’s subpar bioavailability. Therapeutic benefits can be increased by the synergistic interaction of resveratrol and other bioactive components and micronutrients [137]. This is likely due to an increase in resveratrol bioavailability and a broadening of the metabolic effects of the combined agents. Polyphenols are able to link and interact with other compounds using their hydroxyl groups, which allows them to control the efficacy of other chemicals, including proteins and nutrients [2,138]. Compared to free polyphenols, polyphenol complexes might be more bioavailable, soluble, and absorbable in the small intestine due to their stabilized chemical structure [139]. A potential benefit of combining resveratrol with other treatment modalities is that polyphenol complexes can target numerous metabolic pathways. It is true that resveratrol combined with various therapeutic modalities has been shown to have favorable benefits in a variety of diseases and conditions, including cancer [138].
It has been suggested that the combination of vitamins with polyphenols not only results in synergistic biological effects but also stabilizes, maintains, and supports the action of polyphenols. Combining resveratrol with vitamin D3 has been shown to increase resveratrol’s estrogenic activity and modify ER-mediated transcription [137]. In diabetic nephropathy, resveratrol, and vitamin D3 were found to have synergistic benefits. Combining resveratrol with vitamin D3 has been proven to suppress TNF-α and IL-6 expression more than either medicine alone [140]. The combination of glucan, vitamin C, and resveratrol exhibited a higher suppression of breast and lung tumor growth in in vivo models compared to the individual drugs [138,141].
To combat human papillomavirus (HPV)-positive head and neck squamous cell carcinoma, researchers examined the efficacy of a tri-combination (TriCurin) of three polyphenols (curcumin derived from spice turmeric, resveratrol, and epicatechin gallate from green tea). TriCurin inhibited tumor growth by 85% when administered intratumorally in vivo, and it lowered cell viability, clonogenic survival, and tumor sphere formation in vitro while greatly increasing apoptosis [142]. TriCurin also increased p53 protein levels and decreased HPV16 E6 and E7 [142,143]. In a separate investigation, resveratrol and epicatechin gallate were found to trigger apoptosis in prostate cancer cells at dosages as low as 100 microM [144]. CruciferexTM, a substance derived from cruciferous vegetables, was used in a study of human head and neck squamous carcinoma. CruciferexTM contains a mixture of various polyphenols, including resveratrol. Matrix metalloproteinase (MMP) secretion and cell proliferation were both greatly slowed by the polyphenol mixture [145].
Given the prevalence of cancer and other disorders that require the targeting of several molecular pathways simultaneously, the findings of this study indicate that the combination of polyphenols such as resveratrol, nutrients, and other treatments with additive and/or complementary effects may offer a promising approach to achieve synergistic actions.

5. Resveratrol Based Nanoformulations

Various drug carriers have been tried and are being used to improve the poor bioavailability and stability of resveratrol, resulting in a reduced requirement to consume large resveratrol doses and fewer unwanted effects. These can take the form of emulsions, nanoparticles, or liposomes [146,147]. Lipophilic pharmaceuticals can be better stabilized and bioavailable, water-soluble, safe, biodistributed, and biocompatible when encapsulated in solid lipid nanoparticles [148]. The oral bioavailability of resveratrol was improved by up to 335.7 percent when it was loaded into poly-lactic-co-glycolic acid (PLGA) nanoparticles before being administered to rats [149]. The therapeutic potential and efficacy of resveratrol were further increased by nanoparticle formulations, in particular its in vivo anticancer activity in a variety of cancer types. Tumor size was reduced in studies of gliomas, ovarian cancer, and colorectal cancer when resveratrol was administered [150,151]. High-loading resveratrol-loaded gelatin nanoparticles were found to induce cell death via changes in p53, p21, caspase-3, Bax, Bcl-2, and NF-kB expression when utilized in a coculture setting [152]. Using a rat embryonic cardiomyocyte (H9C2) model, researchers discovered that Curcumin-Resveratrol-mP127 (co-loaded curcumin and resveratrol at a molar ratio of 5:1 in Pluronic® F127 micelles) proved cardioprotective by inhibiting apoptosis and reactive oxygen species (ROS) [153]. In addition to cancer treatment, resveratrol has shown therapeutic effects in treating various diseases, therefore further improvements in resveratrol carrier delivery should help mitigate the negative effects of large dosages of resveratrol.
Children with chronic liver illness have stunted growth and development because of poor nutrient absorption [154]. An increase in morbidity and mortality is related to malnutrition, which is an unfavorable prognostic factor in liver transplantation [155]. Current vitamin E supplementation guidelines recommend giving children D-α-tocopheryl-polyethylene-glycol-succinate (TPGS) orally to increase their chances of survival and overall health, although TPGS alone does not prevent spinocerebellar degeneration or lipid peroxidation [156,157]. Micelles loaded with resveratrol have shown a protective action in the liver, boosting the efficacy of TPGS [158]. Through a phase-solubility analysis, the researchers determined that TPGS was suitable for encapsulating resveratrol in micelles; next, resveratrol TPGS formulations were made through solvent casting and solvent diffusion evaporation. Low polydispersity, a somewhat neutral Zeta potential, and small mean diameters (12 nm) were all characteristics of resveratrol TPGS colloidal dispersions [159]. Infrared spectroscopy and differential scanning calorimetry both validated the formulations’ strong drug loading capacity and stable drug release. Resveratrol TPGSs showed reduced toxicity on HaCaT cells compared to empty TPGSs while maintaining the same level of antioxidant activity as pure resveratrol as measured by the DPPH assay. The antioxidant activity of resveratrol and the reduced surfactant toxicity on normal cells suggest that resveratrol TPGS micelles may be able to overcome the obstacles of conventional liver disease therapy [159,160]. Table 3 details the encouraging results obtained from a variety of nanoformulation methods for increasing resveratrol’s biological activity.

6. Toxicity and Adverse Effects of Resveratrol

Resveratrol is known for its antioxidant and chemopreventive properties. However, some investigations have shown that it may act as a pro-oxidizing agent [4,29,30], which may paradoxically affect disease pathogenesis. Resveratrol’s antioxidant activity is a consequence of ROS scavenging [174,175] and antioxidant defense upregulation [176]. Resveratrol may modulate gene and protein expression through redox-sensitive intracellular pathways in tissues and cells. Thus, gene expression modifications and increased antioxidant defense system action lead to cell survival and adaptability in oxidative environments [4,177,178]. Resveratrol can also be auto-oxidized to semiquinones and the comparatively stable 4′-phenoxyl radical, producing ROS under certain enzymatic conditions [179]. pH and hydroxyl anions or organic bases affect polyphenol oxidative processes [180].
A study examined the dose–time dependency of acute resveratrol injection on lipoperoxidation levels in male rats’ hearts, livers, and kidneys synchronized with a 12 h dark–light cycle. Resveratrol was an antioxidant in the dark and a pro-oxidant in the light, possibly reflecting the changing ratio of pro- and antioxidant activities in various organs during a 24 h cycle or postprandial oxidative burst [181]. Dietary polyphenols, including resveratrol, have impressive antioxidant and cytotoxic properties. Since every antioxidant is a redox agent, it can become a pro-oxidant, causing lipid peroxidation and DNA damage under certain conditions. Thus, pro-oxidant action may contribute to resveratrol’s anticancer and apoptotic activities [182]. Resveratrol’s pro-oxidant action can damage DNA and stop the cell cycle [178].
Resveratrol can influence many pathways simultaneously, resulting in diverse or even opposite biological effects depending on concentration or treatment period. Although a dose-dependent resveratrol pro-oxidative action causes oxidative stress in cells over short periods of time, less cytotoxicity was identified at the same dose but with longer exposure times. This suggests that surviving cells were more resistant to resveratrol-induced damage, which decreased over time [4,183]. Low resveratrol doses (0.1–1.0 μg/mL) increase cell proliferation, but higher doses (10.0–100.0 μg/mL) cause apoptosis and reduce mitotic activity in human tumors and endothelial cells [184]. Studies have shown that resveratrol has a dual effect on HT-29 colon cancer cells, with low concentrations (1 and 10 μmol/L) increasing cell number and higher doses (50 or 100 μmol/L) decreasing cell number and increasing apoptotic or necrotic cell percentage [185].
Resveratrol is interesting for drug research because it does not have any harmful or debilitating side effects. Resveratrol dosages have been varied in in vivo and in vitro experiments. However, the best dose and route must be determined according to the patient’s needs. In addition, resveratrol causes cell death in tumor tissues but not in normal tissues [182]. The tumor-specific absorption of resveratrol is due to variations in cellular targets and gene expression in cancer cells. It has been shown [186] that lower resveratrol levels may be beneficial, but higher amounts kill tumor cells by pro-apoptotic signaling.
It has also been shown that resveratrol causes cell death in tumor tissues while having little to no effect on healthy neighboring tissues [187]. Because of variations in accessible cellular targets and gene expression, resveratrol is tumor-specific in that its absorption by normal cells is significantly lower than cancer cells. It has been hypothesized that modest dosages of resveratrol may have health benefits, whereas high amounts destroy tumor cells through pro-apoptotic actions [186].
The short-term use (1.0 g) of resveratrol appears to be safe. However, patients with nonalcoholic fatty liver disease may have nausea, vomiting, diarrhea, and liver impairment at dosages of 2.5 g or more per day [188]. Curiously, in long-term clinical trials [189], no serious adverse effects were reported. In fact, a single 5 g dose of resveratrol or a part of that dose spread out over numerous days has been shown to be safe and well-tolerated [190]. It is important to note, though, that these findings may be replicable among sick individuals because the research was conducted on healthy populations. Orally administered resveratrol is metabolized by gut microbiota [191], making it difficult to determine which effects are solely due to resveratrol or both resveratrol and its metabolites, further complicating our understanding of resveratrol dose-dependency and administration route [191].
High doses of resveratrol have been shown to inhibit cell growth and trigger apoptosis in normal cells, confirming the compound’s biphasic actions throughout a wide range of concentrations [192]. Rapid activation of mitogen-activated protein kinase (MAPK) by resveratrol is dependent on MEK-1, Src, matrix metalloproteinase, and the epidermal growth factor receptor. Nanomolar doses (i.e., magnitude less than that required for ER genomic activity) and concentrations possibly/transiently obtained in serum after oral red wine ingestion [193] activate MAPK and endothelial nitric-oxide synthase (eNOS). Mice as young as one years old benefit from resveratrol’s anti-aging properties when consumed in low dosages. Mice fed a dosage of 1800 mg/kg of resveratrol died after three to four months [194]. Despite the common occurrence of diarrhea, studies on the steady-state pharmacokinetics and tolerability of 2000 mg trans-resveratrol indicated that it was well-tolerated by healthy subjects [195]. This dosage was given twice a day with meals, quercetin, and alcohol.
Studies highlighting the health advantages of resveratrol all point to the importance of dose and age in eliciting such benefits. Another study that looked at the effects of resveratrol on insulin resistance caused by both aging and re-nutrition found that it increased insulin sensitivity in elderly mice fed a standard diet but had no effect on the insulin resistance status of elderly mice fed a high-protein diet [4]. On the other hand, resveratrol was harmful, lowering aortic distensibility and boosting inflammation and superoxide generation. These results suggest that resveratrol is helpful in a malnourished state of physiological aging, but that it may increase atherogenesis-associated risk factors when combined with high-protein diets in elderly mice, possibly by triggering vascular alterations that are themselves a risk factor for the cardiovascular system [196], which remains to be proven without reasonable doubt.
The biological effects of resveratrol are strongly linked to a hormetic effect (as discussed in the introduction), with low doses generally having beneficial effects and high doses having toxic effects. This biphasic effect on the cellular redox state, which is an antioxidant at low doses and a pro-oxidant at high doses, is believed to be responsible for resveratrol’s hormetic property [2,4]. However, studies on resveratrol have mainly focused on short-term outcomes, leading to controversy [4]. The primary focus should be on resveratrol dosage and interaction with the environment’s redox state, especially when precise redox modulation is needed for physiological function or to prevent harmful effects. More extensive studies in complex models are needed to validate current findings. Despite numerous human and animal studies supporting resveratrol’s beneficial properties, there are not enough clinical studies reporting resveratrol’s harmful effects, and the molecular mechanism of resveratrol’s action needs to be better identified.

7. Conclusions and Future Perspectives

This article provides a summary of the research on the health benefits and mechanism of action behind red grape polyphenol resveratrol, including its effects on cardiovascular disease, cancer prevention and therapy, neuroprotection, and diabetes. Studies on both animals and humans show that resveratrol, when consumed in moderation, can have positive health effects. But in order to make resveratrol more promising pharmaceutically, adjustments must be made to its structure and bioavailability. The potential of resveratrol in the treatment and prevention of various diseases warrants further investigation. Additionally, resveratrol’s biochemical mechanism of action has to be thoroughly elucidated. Most importantly, more standardized clinical trial designs are needed to adequately examine the benefits of resveratrol and establish its mechanisms of therapy and prevention of disease.

Author Contributions

All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Deanship of Scientific Research, Vice Presidency for Graduate Studies and Scientific Research, King Faisal University, Saudi Arabia (Grant No. 4585).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Takaoka, M. Resveratrol, a new phenolic compound, from Veratrum grandiflorum. J. Chem. Soc. Jpn. 1939, 60, 1090–1100. [Google Scholar]
  2. Shaito, A.; Posadino, A.M.; Younes, N.; Hasan, H.; Halabi, S.; Alhababi, D.; Al-Mohannadi, A.; Abdel-Rahman, W.M.; Eid, A.H.; Nasrallah, G.K.; et al. Potential Adverse Effects of Resveratrol: A Literature Review. Int. J. Mol. Sci. 2020, 21, 2084. [Google Scholar] [CrossRef]
  3. Akinwumi, B.C.; Bordun, K.M.; Anderson, H.D. Biological activities of stilbenoids. Int. J. Mol. Sci. 2018, 19, 792. [Google Scholar] [CrossRef] [PubMed]
  4. Salehi, B.; Mishra, A.P.; Nigam, M.; Sener, B.; Kilic, M.; Sharifi-Rad, M.; Fokou, P.V.T.; Martins, N.; Sharifi-Rad, J. Resveratrol: A Double-Edged Sword in Health Benefits. Biomedicines 2018, 6, 91. [Google Scholar] [CrossRef] [PubMed]
  5. Colica, C.; Milanović, M.; Milić, N.; Aiello, V.; De Lorenzo, A.; Abenavoli, L. A Systematic Review on Natural Antioxidant Properties of Resveratrol. Nat. Prod. Commun. 2018, 13, 1934578X1801300923. [Google Scholar] [CrossRef]
  6. Duta-Bratu, C.-G.; Nitulescu, G.M.; Mihai, D.P.; Olaru, O.T. Resveratrol and Other Natural Oligomeric Stilbenoid Compounds and Their Therapeutic Applications. Plants 2023, 12, 2935. [Google Scholar] [CrossRef]
  7. Pannu, N.; Bhatnagar, A. Resveratrol: From enhanced biosynthesis and bioavailability to multitargeting chronic diseases. Biomed. Pharm. 2019, 109, 2237–2251. [Google Scholar] [CrossRef]
  8. Andrade, S.; Ramalho, M.J.; Pereira, M.D.C.; Loureiro, J.A. Resveratrol Brain Delivery for Neurological Disorders Prevention and Treatment. Front. Pharm. 2018, 9, 1261. [Google Scholar] [CrossRef] [PubMed]
  9. Xiao, Q.; Zhu, W.; Feng, W.; Lee, S.S.; Leung, A.W.; Shen, J.; Gao, L.; Xu, C. A Review of Resveratrol as a Potent Chemoprotective and Synergistic Agent in Cancer Chemotherapy. Front. Pharmacol. 2019, 9, 1534. [Google Scholar] [CrossRef]
  10. Rodrigo-Gonzalo, M.J.; González-Manzano, S.; Mendez-Sánchez, R.; Santos-Buelga, C.; Recio-Rodríguez, J.I. Effect of Polyphenolic Complements on Cognitive Function in the Elderly: A Systematic Review. Antioxidants 2022, 11, 1549. [Google Scholar] [CrossRef] [PubMed]
  11. García-Caballero, M.; Torres-Vargas, J.A.; Marrero, A.D.; Martínez-Poveda, B.; Medina, M.Á.; Quesada, A.R. Angioprevention of Urologic Cancers by Plant-Derived Foods. Pharmaceutics 2022, 14, 256. [Google Scholar] [CrossRef]
  12. Zhang, L.Z.; Gong, J.G.; Li, J.H.; Hao, Y.S.; Xu, H.J.; Liu, Y.C.; Feng, Z.H. Dietary resveratrol supplementation on growth performance, immune function and intestinal barrier function in broilers challenged with lipopolysaccharide. Poult. Sci. 2023, 102, 102968. [Google Scholar] [CrossRef] [PubMed]
  13. Moreira-Pinto, B.; Costa, L.; Felgueira, E.; Fonseca, B.M.; Rebelo, I. Low Doses of Resveratrol Protect Human Granulosa Cells from Induced-Oxidative Stress. Antioxidants 2021, 10, 561. [Google Scholar] [CrossRef]
  14. Almannai, M.; El-Hattab, A.W.; Ali, M.; Soler-Alfonso, C.; Scaglia, F. Clinical trials in mitochondrial disorders, an update. Mol. Genet. Metab. 2020, 131, 1–13. [Google Scholar] [CrossRef] [PubMed]
  15. Babylon, L.; Schmitt, F.; Franke, Y.; Hubert, T.; Eckert, G.P. Effects of Combining Biofactors on Bioenergetic Parameters, Aβ Levels and Survival in Alzheimer Model Organisms. Int. J. Mol. Sci. 2022, 23, 8670. [Google Scholar] [CrossRef] [PubMed]
  16. Reda, D.; Elshopakey, G.E.; Mahgoub, H.A.; Risha, E.F.; Khan, A.A.; Rajab, B.S.; El-Boshy, M.E.; Abdelhamid, F.M. Effects of resveratrol against induced metabolic syndrome in rats: Role of oxidative stress, inflammation, and insulin resistance. Evid. Based Complement. Altern. Med. eCAM 2022, 2022, 3362005. [Google Scholar] [CrossRef] [PubMed]
  17. Efimova, S.S.; Ostroumova, O.S. Modulation of the Dipole Potential of Model Lipid Membranes with Phytochemicals: Molecular Mechanisms, Structure–Activity Relationships, and Implications in Reconstituted Ion Channels. Membranes 2023, 13, 453. [Google Scholar] [CrossRef]
  18. Hrelia, S.; Di Renzo, L.; Bavaresco, L.; Bernardi, E.; Malaguti, M.; Giacosa, A. Moderate Wine Consumption and Health: A Narrative Review. Nutrients 2023, 15, 175. [Google Scholar] [CrossRef]
  19. Yang, C.; Tian, X.; Han, Y.; Shi, X.; Wang, H.; Li, H. Extracts of Dunkelfelder Grape Seeds and Peel Increase the Metabolic Rate and Reduce Fat Deposition in Mice Maintained on a High-Fat Diet. Foods 2023, 12, 3251. [Google Scholar] [CrossRef]
  20. Martins, D.; Garcia, L.R.; Queiroz, D.A.R.; Lazzarin, T.; Tonon, C.R.; Balin, P.d.S.; Polegato, B.F.; de Paiva, S.A.R.; Azevedo, P.S.; Minicucci, M.F.; et al. Oxidative Stress as a Therapeutic Target of Cardiac Remodeling. Antioxidants 2022, 11, 2371. [Google Scholar] [CrossRef]
  21. Arinno, A.; Apaijai, N.; Chattipakorn, S.C.; Chattipakorn, N. The roles of resveratrol on cardiac mitochondrial function in cardiac diseases. Eur. J. Nutr. 2021, 60, 29–44. [Google Scholar] [CrossRef]
  22. Chedea, V.S.; Tomoiaga, L.L.; Macovei, S.O.; Magureanu, D.C.; Iliescu, M.L.; Bocsan, I.C.; Buzoianu, A.D.; Vosloban, C.M.; Pop, R.M. Antioxidant/pro-oxidant actions of polyphenols from grapevine and wine by-products-base for complementary therapy in ischemic heart diseases. Front. Cardiovasc. Med. 2021, 8, 750508. [Google Scholar] [CrossRef]
  23. Raj, P.; Thandapilly, S.J.; Wigle, J.; Zieroth, S.; Netticadan, T. A Comprehensive Analysis of the Efficacy of Resveratrol in Atherosclerotic Cardiovascular Disease, Myocardial Infarction and Heart Failure. Molecules 2021, 26, 6600. [Google Scholar] [CrossRef]
  24. Arias-Sánchez, R.A.; Torner, L.; Fenton Navarro, B. Polyphenols and Neurodegenerative Diseases: Potential Effects and Mechanisms of Neuroprotection. Molecules 2023, 28, 5415. [Google Scholar] [CrossRef]
  25. Jomova, K.; Raptova, R.; Alomar, S.Y.; Alwasel, S.H.; Nepovimova, E.; Kuca, K.; Valko, M. Reactive oxygen species, toxicity, oxidative stress, and antioxidants: Chronic diseases and aging. Arch. Toxicol. 2023, 97, 2499–2574. [Google Scholar] [CrossRef]
  26. Li, C.; Wang, Z.; Lei, H.; Zhang, D. Recent progress in nanotechnology-based drug carriers for resveratrol delivery. Drug Deliv. 2023, 30, 2174206. [Google Scholar] [CrossRef] [PubMed]
  27. Iweala, E.J.; Oluwapelumi, A.E.; Dania, O.E.; Ugbogu, E.A. Bioactive Phytoconstituents and Their Therapeutic Potentials in the Treatment of Haematological Cancers: A Review. Life 2023, 13, 1422. [Google Scholar] [CrossRef] [PubMed]
  28. Posadino, A.M.; Giordo, R.; Cossu, A.; Nasrallah, G.K.; Shaito, A.; Abou-Saleh, H.; Eid, A.H.; Pintus, G. Flavin Oxidase-Induced ROS Generation Modulates PKC Biphasic Effect of Resveratrol on Endothelial Cell Survival. Biomolecules 2019, 9, 209. [Google Scholar] [CrossRef]
  29. Wani, S.A.; Khan, L.A.; Basir, S.F. Quercetin and resveratrol ameliorate nickel-mediated hypercontraction in isolated wistar rat aorta. J. Smooth Muscle Res. = Nihon Heikatsukin Gakkai Kikanshi 2022, 58, 89–105. [Google Scholar] [CrossRef] [PubMed]
  30. Roumes, H.; Goudeneche, P.; Pellerin, L.; Bouzier-Sore, A.-K. Resveratrol and Some of Its Derivatives as Promising Prophylactic Treatments for Neonatal Hypoxia-Ischemia. Nutrients 2022, 14, 3793. [Google Scholar] [CrossRef]
  31. Rojas-Aguilar, F.A.; Briones-Aranda, A.; Jaramillo-Morales, O.A.; Romero-Nava, R.; Esquinca-Avilés, H.A.; Espinosa-Juárez, J.V. The Additive Antinociceptive Effect of Resveratrol and Ketorolac in the Formalin Test in Mice. Pharmaceuticals 2023, 16, 1078. [Google Scholar] [CrossRef]
  32. Ramli, I.; Posadino, A.M.; Giordo, R.; Fenu, G.; Fardoun, M.; Iratni, R.; Eid, A.H.; Zayed, H.; Pintus, G. Effect of Resveratrol on Pregnancy, Prenatal Complications and Pregnancy-Associated Structure Alterations. Antioxidants 2023, 12, 341. [Google Scholar] [CrossRef]
  33. Cortés-Espinar, A.J.; Ibarz-Blanch, N.; Soliz-Rueda, J.R.; Bonafos, B.; Feillet-Coudray, C.; Casas, F.; Bravo, F.I.; Calvo, E.; Ávila-Román, J.; Mulero, M. Rhythm and ROS: Hepatic Chronotherapeutic Features of Grape Seed Proanthocyanidin Extract Treatment in Cafeteria Diet-Fed Rats. Antioxidants 2023, 12, 1606. [Google Scholar] [CrossRef] [PubMed]
  34. Jiang, H.; Ni, J.; Hu, L.; Xiang, Z.; Zeng, J.; Shi, J.; Chen, Q.; Li, W. Resveratrol May Reduce the Degree of Periodontitis by Regulating ERK Pathway in Gingival-Derived MSCs. Int. J. Mol. Sci. 2023, 24, 11294. [Google Scholar] [CrossRef] [PubMed]
  35. Danailova, Y.; Velikova, T.; Nikolaev, G.; Mitova, Z.; Shinkov, A.; Gagov, H.; Konakchieva, R. Nutritional Management of Thyroiditis of Hashimoto. Int. J. Mol. Sci. 2022, 23, 5144. [Google Scholar] [CrossRef]
  36. Giuliani, C.; Iezzi, M.; Ciolli, L.; Hysi, A.; Bucci, I.; Di Santo, S.; Rossi, C.; Zucchelli, M.; Napolitano, G. Resveratrol has anti-thyroid effects both in vitro and in vivo. Food Chem. Toxicol. 2017, 107, 237–247. [Google Scholar] [CrossRef]
  37. Fragopoulou, E.; Gkotsi, K.; Petsini, F.; Gioti, K.; Kalampaliki, A.D.; Lambrinidis, G.; Kostakis, I.K.; Tenta, R. Synthesis and Biological Evaluation of Resveratrol Methoxy Derivatives. Molecules 2023, 28, 5547. [Google Scholar] [CrossRef]
  38. Iqbal, I.; Wilairatana, P.; Saqib, F.; Nasir, B.; Wahid, M.; Latif, M.F.; Iqbal, A.; Naz, R.; Mubarak, M.S. Plant Polyphenols and Their Potential Benefits on Cardiovascular Health: A Review. Molecules 2023, 28, 6403. [Google Scholar] [CrossRef]
  39. Sun, W.; Shahrajabian, M.H. Therapeutic Potential of Phenolic Compounds in Medicinal Plants—Natural Health Products for Human Health. Molecules 2023, 28, 1845. [Google Scholar] [CrossRef]
  40. Delmas, D.; Aires, V.; Limagne, E.; Dutartre, P.; Mazue, F.; Ghiringhelli, F.; Latruffe, N. Transport, stability, and biological activity of resveratrol. Ann. N. Y. Acad. Sci. 2011, 1215, 48–59. [Google Scholar] [CrossRef] [PubMed]
  41. Burkon, A.; Somoza, V. Quantification of free and protein-bound trans-resveratrol metabolites and identification of trans-resveratrol-C/O-conjugated diglucuronides—Two novel resveratrol metabolites in human plasma. Mol. Nutr. Food Res. 2008, 52, 549–557. [Google Scholar] [CrossRef] [PubMed]
  42. Rotches-Ribalta, M.; Andres-Lacueva, C.; Estruch, R.; Escribano, E.; Urpi-Sarda, M. Pharmacokinetics of resveratrol metabolic profile in healthy humans after moderate consumption of red wine and grape extract tablets. Pharmacol. Res. 2012, 66, 375–382. [Google Scholar] [CrossRef]
  43. Wang, P.; Sang, S. Metabolism and pharmacokinetics of resveratrol and pterostilbene. Biofactors 2018, 44, 16–25. [Google Scholar] [CrossRef] [PubMed]
  44. Baur, J.A.; Sinclair, D.A. Therapeutic potential of resveratrol: The in vivo evidence. Nat. Rev. Drug Discov. 2006, 5, 493–506. [Google Scholar] [CrossRef]
  45. Rezende, J.P.; Hudson, E.A.; De Paula, H.M.C.; Meinel, R.S.; Da Silva, A.D.; Da Silva, L.H.M.; Pires, A. Human serum albumin-resveratrol complex formation: Effect of the phenolic chemical structure on the kinetic and thermodynamic parameters of the interactions. Food Chem. 2020, 307, 125514. [Google Scholar] [CrossRef] [PubMed]
  46. Fan, Y.; Liu, Y.; Gao, L.; Zhang, Y.; Yi, J. Improved chemical stability and cellular antioxidant activity of resveratrol in zein nanoparticle with bovine serum albumin-caffeic acid conjugate. Food Chem. 2018, 261, 283–291. [Google Scholar] [CrossRef] [PubMed]
  47. Geng, T.; Zhao, X.; Ma, M.; Zhu, G.; Yin, L. Resveratrol-Loaded Albumin Nanoparticles with Prolonged Blood Circulation and Improved Biocompatibility for Highly Effective Targeted Pancreatic Tumor Therapy. Nanoscale Res. Lett. 2017, 12, 437. [Google Scholar] [CrossRef] [PubMed]
  48. Pantusa, M.; Bartucci, R.; Rizzuti, B. Stability of trans-resveratrol associated with transport proteins. J. Agric. Food Chem. 2014, 62, 4384–4391. [Google Scholar] [CrossRef]
  49. Walle, T.; Hsieh, F.; DeLegge, M.H.; Oatis, J.E., Jr.; Walle, U.K. High absorption but very low bioavailability of oral resveratrol in humans. Drug Metab. Dispos. 2004, 32, 1377–1382. [Google Scholar] [CrossRef] [PubMed]
  50. Szymkowiak, I.; Kucinska, M.; Murias, M. Between the Devil and the Deep Blue Sea—Resveratrol, Sulfotransferases and Sulfatases—A Long and Turbulent Journey from Intestinal Absorption to Target Cells. Molecules 2023, 28, 3297. [Google Scholar] [CrossRef]
  51. Wang, Q.; Yu, Q.; Wu, M. Antioxidant and neuroprotective actions of resveratrol in cerebrovascular diseases. Front. Pharmacol. 2022, 13, 948889. [Google Scholar] [CrossRef] [PubMed]
  52. Szczepańska, P.; Rychlicka, M.; Groborz, S.; Kruszyńska, A.; Ledesma-Amaro, R.; Rapak, A.; Gliszczyńska, A.; Lazar, Z. Studies on the Anticancer and Antioxidant Activities of Resveratrol and Long-Chain Fatty Acid Esters. Int. J. Mol. Sci. 2023, 24, 7167. [Google Scholar] [CrossRef] [PubMed]
  53. Trius-Soler, M.; Pratico, G.; Gurdeniz, G.; Garcia-Aloy, M.; Canali, R.; Fausta, N.; Brouwer-Brolsma, E.M.; Andres-Lacueva, C.; Dragsted, L.O. Biomarkers of moderate alcohol intake and alcoholic beverages: A systematic literature review. Genes Nutr. 2023, 18, 7. [Google Scholar] [CrossRef]
  54. Karabekir, S.C.; Ozgorgulu, A. Possible protective effects of resveratrol in hepatocellular carcinoma. Iran. J. Basic Med. Sci. 2020, 23, 71–78. [Google Scholar] [PubMed]
  55. Florio, R.; De Filippis, B.; Veschi, S.; di Giacomo, V.; Lanuti, P.; Catitti, G.; Brocco, D.; di Rienzo, A.; Cataldi, A.; Cacciatore, I.; et al. Resveratrol Derivative Exhibits Marked Antiproliferative Actions, Affecting Stemness in Pancreatic Cancer Cells. Int. J. Mol. Sci. 2023, 24, 1977. [Google Scholar] [CrossRef]
  56. Peterle, L.; Sanfilippo, S.; Borgia, F.; Li Pomi, F.; Vadalà, R.; Costa, R.; Cicero, N.; Gangemi, S. The Role of Nutraceuticals and Functional Foods in Skin Cancer: Mechanisms and Therapeutic Potential. Foods 2023, 12, 2629. [Google Scholar] [CrossRef]
  57. Wang, G.; Hu, Z.; Song, X.; Cui, Q.; Fu, Q.; Jia, R.; Zou, Y.; Li, L.; Yin, Z. Analgesic and Anti-Inflammatory Activities of Resveratrol through Classic Models in Mice and Rats. Evid. Based Complement. Alternat. Med. 2017, 2017, 5197567. [Google Scholar] [CrossRef] [PubMed]
  58. Almatroodi, S.A.; Alsahli, M.A.; Aljohani, A.S.M.; Alhumaydhi, F.A.; Babiker, A.Y.; Khan, A.A.; Rahmani, A.H. Potential Therapeutic Targets of Resveratrol, a Plant Polyphenol, and Its Role in the Therapy of Various Types of Cancer. Molecules 2022, 27, 2665. [Google Scholar] [CrossRef] [PubMed]
  59. De Sá Coutinho, D.; Pacheco, M.T.; Frozza, R.L.; Bernardi, A. Anti-Inflammatory Effects of Resveratrol: Mechanistic Insights. Int. J. Mol. Sci. 2018, 19, 1812. [Google Scholar] [CrossRef]
  60. Silva, A.F.R.; Silva-Reis, R.; Ferreira, R.; Oliveira, P.A.; Faustino-Rocha, A.I.; Pinto, M.d.L.; Coimbra, M.A.; Silva, A.M.S.; Cardoso, S.M. The Impact of Resveratrol-Enriched Bread on Cardiac Remodeling in a Preclinical Model of Diabetes. Antioxidants 2023, 12, 1066. [Google Scholar] [CrossRef] [PubMed]
  61. García-Martínez, B.I.; Ruiz-Ramos, M.; Pedraza-Chaverri, J.; Santiago-Osorio, E.; Mendoza-Núñez, V.M. Hypoglycemic Effect of Resveratrol: A Systematic Review and Meta-Analysis. Antioxidants 2021, 10, 69. [Google Scholar] [CrossRef] [PubMed]
  62. Moon, D.O. A comprehensive review of the effects of resveratrol on glucose metabolism: Unveiling the molecular pathways and therapeutic potential in diabetes management. Mol. Biol. Rep. 2023, 50, 8743–8755. [Google Scholar] [CrossRef] [PubMed]
  63. Rahman, M.H.; Akter, R.; Bhattacharya, T.; Abdel-Daim, M.M.; Alkahtani, S.; Arafah, M.W.; Al-Johani, N.S.; Alhoshani, N.M.; Alkeraishan, N.; Alhenaky, A.; et al. Resveratrol and neuroprotection: Impact and its therapeutic potential in Alzheimer’s disease. Front. Pharmacol. 2020, 11, 619024. [Google Scholar] [CrossRef]
  64. Xiang, L.; Wang, Y.; Liu, S.; Liu, B.; Jin, X.; Cao, X. Targeting Protein Aggregates with Natural Products: An Optional Strategy for Neurodegenerative Diseases. Int. J. Mol. Sci. 2023, 24, 11275. [Google Scholar] [CrossRef] [PubMed]
  65. Dos Santos, M.G.; Schimith, L.E.; Andre-Miral, C.; Muccillo-Baisch, A.L.; Arbo, B.D.; Hort, M.A. Neuroprotective effects of resveratrol in in vivo and in vitro experimental models of parkinson’s disease: A systematic review. Neurotox. Res. 2022, 40, 319–345. [Google Scholar] [CrossRef]
  66. Filardo, S.; Di Pietro, M.; Mastromarino, P.; Sessa, R. Therapeutic potential of resveratrol against emerging respiratory viral infections. Pharmacol. Ther. 2020, 214, 107613. [Google Scholar] [CrossRef] [PubMed]
  67. Abba, Y.; Hassim, H.; Hamzah, H.; Noordin, M.M. Antiviral Activity of Resveratrol against Human and Animal Viruses. Adv. Virol. 2015, 2015, 184241. [Google Scholar] [CrossRef]
  68. Yoon, J.; Ku, D.; Lee, M.; Lee, N.; Im, S.G.; Kim, Y. Resveratrol Attenuates the Mitochondrial RNA-Mediated Cellular Response to Immunogenic Stress. Int. J. Mol. Sci. 2023, 24, 7403. [Google Scholar] [CrossRef] [PubMed]
  69. Carpéné, C.; Les, F.; Cásedas, G.; Peiro, C.; Fontaine, J.; Chaplin, A.; Mercader, J.; López, V. Resveratrol Anti-Obesity Effects: Rapid Inhibition of Adipocyte Glucose Utilization. Antioxidants 2019, 8, 74. [Google Scholar] [CrossRef]
  70. Zhou, L.; Xiao, X.; Zhang, Q.; Zheng, J.; Deng, M. Deciphering the anti-obesity benefits of resveratrol: The “gut microbiota-adipose tissue” axis. Front. Endocrinol. 2019, 10, 413. [Google Scholar] [CrossRef]
  71. Vrânceanu, M.; Hegheş, S.-C.; Cozma-Petruţ, A.; Banc, R.; Stroia, C.M.; Raischi, V.; Miere, D.; Popa, D.-S.; Filip, L. Plant-Derived Nutraceuticals Involved in Body Weight Control by Modulating Gene Expression. Plants 2023, 12, 2273. [Google Scholar] [CrossRef] [PubMed]
  72. Gal, R.; Deres, L.; Toth, K.; Halmosi, R.; Habon, T. The Effect of Resveratrol on the Cardiovascular System from Molecular Mechanisms to Clinical Results. Int. J. Mol. Sci. 2021, 22, 10152. [Google Scholar] [CrossRef]
  73. Kazemirad, H.; Kazerani, H.R. Cardioprotective effects of resveratrol following myocardial ischemia and reperfusion. Mol. Biol. Rep. 2020, 47, 5843–5850. [Google Scholar] [CrossRef]
  74. Menezes-Rodrigues, F.S.; Errante, P.R.; Araujo, E.A.; Fernandes, M.P.P.; Silva, M.M.D.; Pires-Oliveira, M.; Scorza, C.A.; Scorza, F.A.; Taha, M.O.; Caricati-Neto, A. Cardioprotection stimulated by resveratrol and grape products prevents lethal cardiac arrhythmias in an animal model of ischemia and reperfusion. Acta Cir. Bras. 2021, 36, e360306. [Google Scholar] [CrossRef]
  75. Bohara, R.A.; Tabassum, N.; Singh, M.P.; Gigli, G.; Ragusa, A.; Leporatti, S. Recent Overview of Resveratrol’s Beneficial Effects and Its Nano-Delivery Systems. Molecules 2022, 27, 5154. [Google Scholar] [CrossRef] [PubMed]
  76. Wijekoon, C.; Netticadan, T.; Siow, Y.L.; Sabra, A.; Yu, L.; Raj, P.; Prashar, S. Potential Associations among Bioactive Molecules, Antioxidant Activity and Resveratrol Production in Vitis vinifera Fruits of North America. Molecules 2022, 27, 336. [Google Scholar] [CrossRef]
  77. Gu, T.; Wang, N.; Wu, T.; Ge, Q.; Chen, L. Antioxidative stress mechanisms behind resveratrol: A multidimensional analysis. J. Food Qual. 2021, 2021, 5571733. [Google Scholar] [CrossRef]
  78. Zhou, D.D.; Luo, M.; Huang, S.Y.; Saimaiti, A.; Shang, A.; Gan, R.Y.; Li, H.B. Effects and mechanisms of resveratrol on aging and age-related diseases. Oxidative Med. Cell. Longev. 2021, 2021, 9932218. [Google Scholar] [CrossRef] [PubMed]
  79. Pyo, I.S.; Yun, S.; Yoon, Y.E.; Choi, J.W.; Lee, S.J. Mechanisms of aging and the preventive effects of resveratrol on age-related diseases. Molecules 2020, 25, 4649. [Google Scholar] [CrossRef]
  80. Leis, K.; Pisanko, K.; Jundziłł, A.; Mazur, E.; Mêcińska-Jundziłł, K.; Witmanowski, H. Resveratrol as a factor preventing skin aging and affecting its regeneration. Postep. Dermatol. Alergol. 2022, 39, 439–445. [Google Scholar] [CrossRef]
  81. Gowd, V.; Kang, Q.; Wang, Q.; Wang, Q.; Chen, F.; Cheng, K.W. Resveratrol: Evidence for its nephroprotective effect in diabetic nephropathy. Adv. Nutr. 2020, 11, 1555–1568. [Google Scholar] [CrossRef] [PubMed]
  82. Jin, Q.; Liu, T.; Qiao, Y.; Liu, D.; Yang, L.; Mao, H.; Ma, F.; Wang, Y.; Peng, L.; Zhan, Y. Oxidative stress and inflammation in diabetic nephropathy: Role of polyphenols. Front. Immunol. 2023, 14, 1185317. [Google Scholar] [CrossRef] [PubMed]
  83. Cheng, K.; Song, Z.; Chen, Y.; Li, S.; Zhang, Y.; Zhang, H.; Zhang, L.; Wang, C.; Wang, T. Resveratrol Protects Against Renal Damage via Attenuation of Inflammation and Oxidative Stress in High-Fat-Diet-Induced Obese Mice. Inflammation 2018, 42, 937–945. [Google Scholar] [CrossRef] [PubMed]
  84. Ahmadi, A.; Jamialahmadi, T.; Sahebkar, A. Polyphenols and atherosclerosis: A critical review of clinical effects on LDL oxidation. Pharmacol. Res. 2022, 184, 106414. [Google Scholar] [CrossRef] [PubMed]
  85. Castaldo, L.; Narváez, A.; Izzo, L.; Graziani, G.; Gaspari, A.; Di Minno, G.; Ritieni, A. Red Wine Consumption and Cardiovascular Health. Molecules 2019, 24, 3626. [Google Scholar] [CrossRef] [PubMed]
  86. Buljeta, I.; Pichler, A.; Šimunović, J.; Kopjar, M. Beneficial Effects of Red Wine Polyphenols on Human Health: Comprehensive Review. Curr. Issues Mol. Biol. 2023, 45, 782–798. [Google Scholar] [CrossRef]
  87. Rasines-Perea, Z.; Teissedre, P.-L. Grape Polyphenols’ Effects in Human Cardiovascular Diseases and Diabetes. Molecules 2017, 22, 68. [Google Scholar] [CrossRef] [PubMed]
  88. Chiva-Blanch, G.; Urpi-Sarda, M.; Ros, E.; Valderas-Martinez, P.; Casas, R.; Arranz, S.; Guillén, M.; Lamuela-Raventós, R.M.; Llorach, R.; Andres-Lacueva, C.; et al. Effects of red wine polyphenols and alcohol on glucose metabolism and the lipid profile: A randomized clinical trial. Clin. Nutr. 2013, 32, 200–206. [Google Scholar] [CrossRef]
  89. Navarro-García, F.; Ponce-Ruíz, N.; Rojas-García, A.E.; Ávila-Villarreal, G.; Herrera-Moreno, J.F.; Barrón-Vivanco, B.S.; Bernal-Hernández, Y.Y.; González-Arias, C.A.; Medina-Díaz, I.M. The Role of Nutritional Habits and Moderate Red Wine Consumption in PON1 Status in Healthy Population. Appl. Sci. 2021, 11, 9503. [Google Scholar] [CrossRef]
  90. Salazar, H.M.; de Deus Mendonça, R.; Laclaustra, M.; Moreno-Franco, B.; Åkesson, A.; Guallar-Castillón, P.; Donat-Vargas, C. The intake of flavonoids, stilbenes, and tyrosols, mainly consumed through red wine and virgin olive oil, is associated with lower carotid and femoral subclinical atherosclerosis and coronary calcium. Eur. J. Nutr. 2022, 61, 2697–2709. [Google Scholar] [CrossRef]
  91. Tresserra-Rimbau, A.; Medina-Remón, A.; Lamuela-Raventós, R.M.; Bulló, M.; Salas-Salvadó, J.; Corella, D.; Fitó, M.; Gea, A.; Gómez-Gracia, E.; Lapetra, J.; et al. Moderate red wine consumption is associated with a lower prevalence of the metabolic syndrome in the PREDIMED population. Br. J. Nutr. 2015, 113, 121–130. [Google Scholar] [CrossRef] [PubMed]
  92. Surh, Y.J. Cancer chemoprevention with dietary phytochemicals. Nat. Rev. Cancer 2003, 3, 768–780. [Google Scholar] [CrossRef] [PubMed]
  93. Amor, S.; Châlons, P.; Aires, V.; Delmas, D. Polyphenol Extracts from Red Wine and Grapevine: Potential Effects on Cancers. Diseases 2018, 6, 106. [Google Scholar] [CrossRef] [PubMed]
  94. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef]
  95. Liu, L.; Jin, R.; Hao, J.; Zeng, J.; Yin, D.; Yi, Y.; Zhu, M.; Mandal, A.; Hua, Y.; Ng, C.K.; et al. Consumption of the Fish Oil High-Fat Diet Uncouples Obesity and Mammary Tumor Growth through Induction of Reactive Oxygen Species in Protumor Macrophages. AACR 2020, 80, 2564–2574. [Google Scholar] [CrossRef] [PubMed]
  96. Kim, M.-J.; Kim, Y.-J.; Park, H.-J.; Chung, J.-H.; Leem, K.-H.; Kim, H.-K. Apoptotic effect of red wine polyphenols on human colon cancer SNU-C4 cells. Food Chem. Toxicol. 2006, 44, 898–902. [Google Scholar] [CrossRef]
  97. Bastide, N.M.; Naud, N.; Nassy, G.; Vendeuvre, J.-L.; Taché, S.; Guéraud, F.; Hobbs, D.A.; Kuhnle, G.G.; Corpet, D.; Pierre, F.H.F. Red Wine and Pomegranate Extracts Suppress Cured Meat Promotion of Colonic Mucin-Depleted Foci in Carcinogen-Induced Rats. Nutr. Cancer 2017, 69, 289–298. [Google Scholar] [CrossRef]
  98. 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]
  99. Sabadashka, M.; Hertsyk, D.; Strugała-Danak, P.; Dudek, A.; Kanyuka, O.; Kucharska, A.Z.; Kaprelyants, L.; Sybirna, N. Anti-Diabetic and Antioxidant Activities of Red Wine Concentrate Enriched with Polyphenol Compounds under Experimental Diabetes in Rats. Antioxidants 2021, 10, 1399. [Google Scholar] [CrossRef]
  100. Martin, M.A.; Goy, L.; Ramos, S. Protective effects of tea, red wine and cocoa in diabetes. Evidences from human studies. Food Chem. Toxicol. 2017, 109, 302–314. [Google Scholar] [CrossRef]
  101. Xia, X.; Sun, B.; Li, W.; Zhang, X.; Zhao, Y. Anti-diabetic activity phenolic constituents from red wine against α-glucosidase and α-amylase. J. Food Process. Preserv. 2016, 41, 12942. [Google Scholar] [CrossRef]
  102. Tamargo, A.; Cueva, C.; Silva, M.; Molinero, N.; Miralles, B.; Bartolomé, B.; Moreno-Arribas, M.V. Gastrointestinal co-digestion of wine polyphenols with glucose/whey proteins affects their bioaccessibility and impact on colonic microbiota. Food Res. Int. 2022, 155, 111010. [Google Scholar] [CrossRef] [PubMed]
  103. Shen, C.; Cheng, W.; Yu, P.; Wang, L.; Zhou, L.; Zeng, L.; Yang, Q. Resveratrol pretreatment attenuates injury and promotes proliferation of neural stem cells following oxygen-glucose deprivation/reoxygenation by upregulating the expression of Nrf2, HO-1 and NQO1 in vitro. Mol. Med. Rep. 2016, 14, 3646–3654. [Google Scholar] [CrossRef]
  104. Son, Y.; Byun, S.J.; Pae, H.-O. Involvement of heme oxygenase-1 expression in neuroprotection by piceatannol, a natural analog and a metabolite of resveratrol, against glutamate-mediated oxidative injury in HT22 neuronal cells. Amino Acids 2013, 45, 393–401. [Google Scholar] [CrossRef]
  105. 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] [PubMed]
  106. Martínez-Huélamo, M.; Rodríguez-Morató, J.; Boronat, A.; De la Torre, R. Modulation of Nrf2 by Olive Oil and Wine Polyphenols and Neuroprotection. Antioxidants 2017, 6, 73. [Google Scholar] [CrossRef]
  107. Rocha-Parra, D.; Chirife, J.; Zamora, C.; De Pascual-Teresa, S. Chemical Characterization of an Encapsulated Red Wine Powder and Its Effects on Neuronal Cells. Molecules 2018, 23, 842. [Google Scholar] [CrossRef] [PubMed]
  108. Li, Y.; Peng, Y.; Shen, Y.; Zhang, Y.; Liu, L.; Yang, X. Dietary polyphenols: Regulate the advanced glycation end products-RAGE axis and the microbiota-gut-brain axis to prevent neurodegenerative diseases. Crit. Rev. Food Sci. Nutr. 2022, 19, 1–27. [Google Scholar] [CrossRef]
  109. Singh, A.P.; Singh, R.; Verma, S.S.; Rai, V.; Kaschula, C.H.; Maiti, P.; Gupta, S.C. Health benefits of resveratrol: Evidence from clinical studies. Med. Res. Rev. 2019, 39, 1851–1891. [Google Scholar] [CrossRef]
  110. de Vries, K.; Strydom, M.; Steenkamp, V. A Brief Updated Review of Advances to Enhance Resveratrol’s Bioavailability. Molecules 2021, 26, 4367. [Google Scholar] [CrossRef]
  111. Farzin, L.; Asghari, S.; Rafraf, M.; Asghari-Jafarabadi, M.; Shirmohammadi, M. No beneficial effects of resveratrol supplementation on atherogenic risk factors in patients with nonalcoholic fatty liver disease. Int. J. Vitam. Nutr. Res. 2020, 90, 279–289. [Google Scholar] [CrossRef] [PubMed]
  112. Mansur, A.P.; Roggerio, A.; Goes, M.F.S.; Avakian, S.D.; Leal, D.P.; Maranhão, R.C.; Strunz, C.M.C. Serum concentrations and gene expression of sirtuin 1 in healthy and slightly overweight subjects after caloric restriction or resveratrol supplementation: A randomized trial. Int. J. Cardiol. 2017, 227, 788–794. [Google Scholar] [CrossRef]
  113. Amato, B.; Compagna, R.; Amato, M.; Gallelli, L.; de Franciscis, S.; Serra, R. Aterofisiol® in carotid plaque evolution. Drug Des. Dev. Ther. 2015, 9, 3877–3884. [Google Scholar]
  114. Hoseini, A.; Namazi, G.; Farrokhian, A.; Reiner, Ž.; Aghadavod, E.; Bahmani, F.; Asemi, Z. The effects of resveratrol on metabolic status in patients with type 2 diabetes mellitus and coronary heart disease. Food Funct. 2019, 10, 6042–6051. [Google Scholar] [CrossRef]
  115. Diaz, M.; Avila, A.; Degens, H.; Coeckelberghs, E.; Vanhees, L.; Cornelissen, V.; Azzawi, M. Acute resveratrol supplementation in coronary artery disease: Towards patient stratification. Scand. Cardiovasc. J. 2020, 54, 14–19. [Google Scholar] [CrossRef]
  116. Marques, B.; Trindade, M.; Aquino, J.C.F.; Cunha, A.R.; Gismondi, R.O.; Neves, M.F.; Oigman, W. Beneficial effects of acute trans-resveratrol supplementation in treated hypertensive patients with endothelial dysfunction. Clin. Exp. Hypertens. 2018, 40, 218–223. [Google Scholar] [CrossRef]
  117. Biesinger, S.; Michaels, H.A.; Quadros, A.S.; Qian, Y.; Rabovsky, A.B.; Badger, R.S.; Jalili, T. A combination of isolated phytochemicals and botanical extracts lowers diastolic blood pressure in a randomized controlled trial of hypertensive subjects. Eur. J. Clin. Nutr. 2016, 70, 10–16. [Google Scholar] [CrossRef] [PubMed]
  118. Albrecht, T.; Waliszewski, M.; Roca, C.; Redlich, U.; Tautenhahn, J.; Pech, M.; Halloul, Z.; Gögebakan, Ö.; Meyer, D.R.; Gemeinhardt, I.; et al. Two-Year Clinical Outcomes of the CONSEQUENT Trial: Can Femoropopliteal Lesions be Treated with Sustainable Clinical Results that are Economically Sound? Cardiovasc. Interv. Radiol. 2018, 41, 1008–1014. [Google Scholar] [CrossRef]
  119. 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]
  120. Abdollahi, S.; Salehi-Abargouei, A.; Toupchian, O.; Sheikhha, M.H.; Fallahzadeh, H.; Rahmanian, M.; Tabatabaie, M.; Mozaffari-Khosravi, H. The Effect of Resveratrol Supplementation on Cardio-Metabolic Risk Factors in Patients with Type 2 Diabetes: A Randomized, Double-Blind Controlled Trial. Phytother. Res. 2019, 33, 3153–3162. [Google Scholar] [CrossRef]
  121. Brasnyo, P.; Molnar, G.A.; Mohas, M.; Marko, L.; Laczy, B.; Cseh, J.; Mikolas, E.; Szijarto, I.A.; Merei, A.; Halmai, R.; et al. Resveratrol improves insulin sensitivity, reduces oxidative stress and activates the akt pathway in type 2 diabetic patients. Br. J. Nutr. 2011, 106, 383–389. [Google Scholar] [CrossRef]
  122. Poulsen, M.K.; Nellemann, B.; Bibby, B.M.; Stødkilde-Jørgensen, H.; Pedersen, S.B.; Grønbaek, H.; Nielsen, S. No effect of resveratrol on VLDL-TG kinetics and insulin sensitivity in obese men with nonalcoholic fatty liver disease. Diabetes Obes. Metab. 2018, 20, 2504–2509. [Google Scholar] [CrossRef]
  123. Timmers, S.; de Ligt, M.; Phielix, E.; van de Weijer, T.; Hansen, J.; Moonen-Kornips, E.; Schaart, G.; Kunz, I.; Hesselink, M.K.; Schrauwen-Hinderling, V.B.; et al. Resveratrol as Add-on Therapy in Subjects with Well-Controlled Type 2 Diabetes: A Randomized Controlled Trial. Diabetes Care 2016, 39, 2211–2217. [Google Scholar] [CrossRef] [PubMed]
  124. 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 trial. Am. J. Clin. Nutr. 2016, 103, 66–70. [Google Scholar] [CrossRef]
  125. Pollack, R.M.; Barzilai, N.; Anghel, V.; Kulkarni, A.S.; Golden, A.; O’Broin, P.; Sinclair, D.A.; Bonkowski, M.S.; Coleville, A.J.; Powell, D.; et al. Resveratrol Improves Vascular Function and Mitochondrial Number but Not Glucose Metabolism in Older Adults. J. Gerontol. A Biol. Sci. Med. Sci. 2017, 72, 1703–1709. [Google Scholar] [CrossRef] [PubMed]
  126. Boswijk, E.; de Ligt, M.; Habets, M.J.; Mingels, A.M.A.; van Marken Lichtenbelt, W.D.; Mottaghy, F.M.; Schrauwen, P.; Wildberger, J.E.; Bucerius, J. Resveratrol treatment does not reduce arterial inflammation in males at risk of type 2 diabetes: A randomized crossover trial. Nukl. Nucl. Med. 2022, 61, 33–41. [Google Scholar] [CrossRef]
  127. Pecoraro, L.; Zoller, T.; Atkinson, R.L.; Nisi, F.; Antoniazzi, F.; Cavarzere, P.; Piacentini, G.; Pietrobelli, A. Supportive treatment of vascular dysfunction in pediatric subjects with obesity: The OBELIX study. Nutr. Diabetes 2022, 12, 2. [Google Scholar] [CrossRef]
  128. Vors, C.; Couillard, C.; Paradis, M.E.; Gigleux, I.; Marin, J.; Vohl, M.C.; Couture, P.; Lamarche, B. Supplementation with Resveratrol and Curcumin Does Not Affect the Inflammatory Response to a High-Fat Meal in Older Adults with Abdominal Obesity: A Randomized, Placebo-Controlled Crossover Trial. J. Nutr. 2018, 148, 379–388. [Google Scholar] [CrossRef] [PubMed]
  129. Mankowski, R.T.; You, L.; Buford, T.W.; Leeuwenburgh, C.; Manini, T.M.; Schneider, S.; Qiu, P.; Anton, S.D. Higher dose of resveratrol elevated cardiovascular disease risk biomarker levels in overweight older adults—A pilot study. Exp. Gerontol. 2020, 131, 110821. [Google Scholar] [CrossRef] [PubMed]
  130. Kung, H.-C.; Lin, K.-J.; Kung, C.-T.; Lin, T.-K. Oxidative Stress, Mitochondrial Dysfunction, and Neuroprotection of Polyphenols with Respect to Resveratrol in Parkinson’s Disease. Biomedicines 2021, 9, 918. [Google Scholar] [CrossRef] [PubMed]
  131. 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]
  132. Paller, C.J.; Rudek, M.A.; Zhou, X.C.; Wagner, W.D.; Hudson, T.S.; Anders, N.; Hammers, H.J.; Dowling, D.; King, S.; Antonarakis, E.S.; et al. ; et al. A phase i study of muscadine grape skin extract in men with biochemically recurrent prostate cancer: Safety, tolerability, and dose determination. Prostate 2015, 75, 1518–1525. [Google Scholar] [CrossRef] [PubMed]
  133. Kjaer, T.N.; Ornstrup, M.J.; Poulsen, M.M.; Jorgensen, J.O.; Hougaard, D.M.; Cohen, A.S.; Neghabat, S.; Richelsen, B.; Pedersen, S.B. Resveratrol reduces the levels of circulating androgen precursors but has no effect on, testosterone, dihydrotestosterone, psa levels or prostate volume. A 4-month randomised trial in middle-aged men. Prostate 2015, 75, 1255–1263. [Google Scholar] [CrossRef] [PubMed]
  134. Howells, L.M.; Berry, D.P.; Elliott, P.J.; Jacobson, E.W.; Hoffmann, E.; Hegarty, B.; Brown, K.; Steward, W.P.; Gescher, A.J. Phase i randomized, double-blind pilot study of micronized resveratrol (srt501) in patients with hepatic metastases—Safety, pharmacokinetics, and pharmacodynamics. Cancer Prev. Res. 2011, 4, 1419–1425. [Google Scholar] [CrossRef] [PubMed]
  135. Patel, K.R.; Brown, V.A.; Jones, D.J.; Britton, R.G.; Hemingway, D.; Miller, A.S.; West, K.P.; Booth, T.D.; Perloff, M.; Crowell, J.A.; et al. Clinical pharmacology of resveratrol and its metabolites in colorectal cancer patients. Cancer Res. 2010, 70, 7392–7399. [Google Scholar] [CrossRef] [PubMed]
  136. Zhu, W.; Qin, W.; Zhang, K.; Rottinghaus, G.E.; Chen, Y.C.; Kliethermes, B.; Sauter, E.R. Trans-resveratrol alters mammary promoter hypermethylation in women at increased risk for breast cancer. Nutr. Cancer 2012, 64, 393–400. [Google Scholar] [CrossRef]
  137. Uberti, F.; Morsanuto, V.; Aprile, S.; Ghirlanda, S.; Stoppa, I.; Cochis, A.; Grosa, G.; Rimondini, L.; Molinari, C. Biological effects of combined resveratrol and vitamin D3 on ovarian tissue. J. Ovarian Res. 2017, 10, 61. [Google Scholar] [CrossRef]
  138. Feng, Y.; Jin, C.; Lv, S.; Zhang, H.; Ren, F.; Wang, J. Molecular Mechanisms and Applications of Polyphenol-Protein Complexes with Antioxidant Properties: A Review. Antioxidants 2023, 12, 1577. [Google Scholar] [CrossRef]
  139. Gambini, J.; Inglés, M.; Olaso, G.; Lopez-Grueso, R.; Bonet-Costa, V.; Gimeno-Mallench, L.; Mas-Bargues, C.; Abdelaziz, K.M.; Gomez-Cabrera, M.C.; Vina, J.; et al. Properties of Resveratrol: In Vitro and In Vivo Studies about Metabolism, Bioavailability, and Biological Effects in Animal Models and Humans. Oxidative Med. Cell Longev. 2015, 2015, 837042. [Google Scholar] [CrossRef]
  140. Maity, B.; Bora, M.; Sur, D.J.O.P.; Medicine, E. An effect of combination of resveratrol with vitamin D3 on modulation of proinflammatory cytokines in diabetic nephropathy induces rat. Orient. Pharm. Exp. Med. 2018, 18, 127–138. [Google Scholar] [CrossRef]
  141. Vetvicka, V.; Vetvickova, J. Combination of glucan, resveratrol and vitamin C demonstrates strong anti-tumor potential. Anticancer Res. 2012, 32, 81–87. [Google Scholar]
  142. Mukherjee, S.; Debata, P.R.; Hussaini, R.; Chatterjee, K.; Baidoo, J.N.E.; Sampat, S.; Szerszen, A.; Navarra, J.P.; Fata, J.; Severinova, E.; et al. Unique synergistic formulation of curcumin, epicatechin gallate and resveratrol, tricurin, suppresses HPV E6, eliminates HPV+ cancer cells, and inhibits tumor progression. Oncotarget 2017, 8, 60904–60916. [Google Scholar] [CrossRef]
  143. Piao, L.; Mukherjee, S.; Chang, Q.; Xie, X.; Li, H.; Castellanos, M.R.; Banerjee, P.; Iqbal, H.; Ivancic, R.; Wang, X.; et al. TriCurin, a novel formulation of curcumin, epicatechin gallate, and resveratrol, inhibits the tumorigenicity of human papillomavirus-positive head and neck squamous cell carcinoma. Oncotarget 2017, 8, 60025–60035. [Google Scholar] [CrossRef]
  144. Ahmad, K.A.; Harris, N.H.; Johnson, A.D.; Lindvall, H.C.; Wang, G.; Ahmed, K. Protein kinase CK2 modulates apoptosis induced by resveratrol and epigallocatechin-3-gallate in prostate cancer cells. Mol. Cancer 2007, 6, 1006–1012. [Google Scholar] [CrossRef] [PubMed]
  145. Roomi, M.W.; Kalinovsky, T.; Roomi, N.W.; Niedzwiecki, A.; Rath, M. In vitro and in vivo inhibition of human Fanconi anemia head and neck squamous carcinoma by a phytonutrient combination. Int. J. Oncol. 2015, 46, 2261–2266. [Google Scholar] [CrossRef] [PubMed]
  146. Intagliata, S.; Modica, M.N.; Santagati, L.M.; Montenegro, L. Strategies to Improve Resveratrol Systemic and Topical Bioavailability: An Update. Antioxid 2019, 8, 244. [Google Scholar] [CrossRef]
  147. Wan, S.; Zhang, L.; Quan, Y.; Wei, K. Resveratrol-loaded PLGA nanoparticles: Enhanced stability, solubility and bioactivity of resveratrol for non-alcoholic fatty liver disease therapy. R. Soc. Open Sci. 2018, 5, 181457. [Google Scholar] [CrossRef]
  148. Aatif, M. Current Understanding of Polyphenols to Enhance Bioavailability for Better Therapies. Biomedicines 2023, 11, 2078. [Google Scholar] [CrossRef] [PubMed]
  149. Siu, F.Y.; Ye, S.; Lin, H.; Li, S. Galactosylated PLGA nanoparticles for the oral delivery of resveratrol: Enhanced bioavailability and in vitro anti-inflammatory activity. Int. J. Nanomed. 2018, 13, 4133–4144. [Google Scholar] [CrossRef] [PubMed]
  150. Thipe, V.C.; Panjtan Amiri, K.; Bloebaum, P.; Raphael Karikachery, A.; Khoobchandani, M.; Katti, K.K.; Jurisson, S.S.; Katti, K.V. Development of resveratrol-conjugated gold nanoparticles: Interrelationship of increased resveratrol corona on anti-tumor efficacy against breast, pancreatic and prostate cancers. Int. J. Nanomed. 2019, 14, 4413–4428. [Google Scholar] [CrossRef]
  151. Santos, A.C.; Pereira, I.; Magalhaes, M.; Pereira-Silva, M.; Caldas, M.; Ferreira, L.; Figueiras, A.; Ribeiro, A.J.; Veiga, F. Targeting Cancer Via Resveratrol-Loaded Nanoparticles Administration: Focusing on In Vivo Evidence. Aaps J. 2019, 21, 57. [Google Scholar] [CrossRef]
  152. Kumar, A.; Kurmi, B.D.; Singh, A.; Singh, D. Potential role of resveratrol and its nano-formulation as anti-cancer agent. Explor. Target. Anti-Tumor Ther. 2022, 3, 643–658. [Google Scholar] [CrossRef]
  153. Carlson, L.J.; Cote, B.; Alani, A.W.; Rao, D.A. Polymeric micellar co-delivery of resveratrol and curcumin to mitigate in vitro doxorubicin-induced cardiotoxicity. J. Pharm. Sci. 2014, 103, 2315–2322. [Google Scholar] [CrossRef]
  154. Santos, J.L.; Choquette, M.; Bezerra, J.A. Cholestatic Liver Disease in Children. Curr. Gastroenterol. Rep. 2010, 12, 30–39. [Google Scholar] [CrossRef]
  155. Yang, C.H.; Perumpail, B.J.; Yoo, E.R.; Ahmed, A.; Kerner, J.A., Jr. Nutritional Needs and Support for Children with Chronic Liver Disease. Nutrients 2017, 9, 1127. [Google Scholar] [CrossRef] [PubMed]
  156. European Food Safety Agency (EFSA). Opinion of the Scientific Panel on food additives, flavourings, processing aids and materials in contact with food (AFC) related to D-alpha-tocopheryl polyethylene glycol 1000 succinate (TPGS) in use for food for particular nutritional purposes. EFSA J. 2007, 490, 1–20. [Google Scholar]
  157. Thébaut, A.; Nemeth, A.; Le Mouhaër, J.; Scheenstra, R.; Baumann, U.; Koot, B.; Gottrand, F.; Houwen, R.; Monard, L.; de Micheaux, S.L.; et al. Oral Tocofersolan Corrects or Prevents Vitamin E Deficiency in Children with Chronic Cholestasis. J. Pediatr. Gastroenterol. Nutr. 2016, 63, 610–615. [Google Scholar] [CrossRef] [PubMed]
  158. Chimento, A.; De Amicis, F.; Sirianni, R.; Sinicropi, M.S.; Puoci, F.; Casaburi, I.; Saturnino, C.; Pezzi, V. Progress to Improve Oral Bioavailability and Beneficial Effects of Resveratrol. Int. J. Mol. Sci. 2019, 20, 1381. [Google Scholar] [CrossRef]
  159. Zuccari, G.; Alfei, S.; Zorzoli, A.; Marimpietri, D.; Turrini, F.; Baldassari, S.; Marchitto, L.; Caviglioli, G. Increased Water-Solubility and Maintained Antioxidant Power of Resveratrol by Its Encapsulation in Vitamin E TPGS Micelles: A Potential Nutritional Supplement for Chronic Liver Disease. Pharmaceutics 2021, 13, 1128. [Google Scholar] [CrossRef] [PubMed]
  160. Soldatova, Y.V.; Faingold, I.I.; Poletaeva, D.A.; Kozlov, A.V.; Emel’yanova, N.S.; Khodos, I.I.; Chernyaev, D.A.; Kurmaz, S.V. Design and Investigation of New Water-Soluble Forms of α-Tocopherol with Antioxidant and Antiglycation Activity Using Amphiphilic Copolymers of N-Vinylpyrrolidone. Pharmaceutics 2023, 15, 1388. [Google Scholar] [CrossRef]
  161. Vasconcelos, T.; Araújo, F.; Lopes, C.; Loureiro, A.; Das Neves, J.; Marques, S.; Sarmento, B. Multicomponent self-nano emulsifying delivery systems of resveratrol with enhanced pharmacokinetics profile. Eur. J. Pharm. Sci. 2019, 137, 105011. [Google Scholar] [CrossRef] [PubMed]
  162. Calvo-Castro, L.A.; Schiborr, C.; David, F.; Ehrt, H.; Voggel, J.; Sus, N.; Behnam, D.; Bosy-Westphal, A.; Frank, J. The Oral Bioavailability of Trans-Resveratrol from a Grapevine-Shoot Extract in Healthy Humans is Significantly Increased by Micellar Solubilization. Mol. Nutr. Food Res. 2018, 62, e1701057. [Google Scholar] [CrossRef] [PubMed]
  163. Santos, A.C.; Veiga, F.; Sequeira, J.A.D.; Fortuna, A.; Falcão, A.; Souto, E.B.; Pattekari, P.; Ribeiro, C.F.; Ribeiro, A.J. First-time oral administration of resveratrol-loaded layer-by-layer nanoparticles to rats—A pharmacokinetics study. Analyst 2019, 144, 2062–2079. [Google Scholar] [CrossRef]
  164. Yang, C.; Wang, Y.; Xie, Y.; Liu, G.; Lu, Y.; Wu, W.; Chen, L. Oat protein-shellac nanoparticles as a delivery vehicle for resveratrol to improve bioavailability in vitro and in vivo. Nanomedicine 2019, 14, 2853–2871. [Google Scholar] [CrossRef]
  165. Peñalva, R.; Morales, J.; González-Navarro, C.J.; Larrañeta, E.; Quincooces, G.; Peñuelas, I.; Irache, J.; Juan, M. Increased Oral Bioavailability of Resveratrol by Its Encapsulation in Casein Nanoparticles. Int. J. Mol. Sci. 2018, 19, 2816. [Google Scholar] [CrossRef]
  166. Singh, S.K.; Makadia, V.; Sharma, S.; Rashid, M.; Shahi, S.; Mishra, P.R.; Wahajuddin, M.; Gayen, J.R. Preparation and in-vitro/in-vivo characterization of trans-resveratrol nanocrystals for oral administration. Drug Deliv. Transl. Res. 2017, 7, 395–407. [Google Scholar] [CrossRef] [PubMed]
  167. Wu, M.; Zhong, C.; Deng, Y.; Zhang, Q.; Zhang, X.; Zhao, X. Resveratrol loaded glycyrrhizic acid-conjugated human serum albumin nanoparticles for tail vein injection II: Pharmacokinetics, tissue distribution and bioavailability. Drug Deliv. 2020, 27, 81–90. [Google Scholar] [CrossRef]
  168. Katekar, R.; Thombre, G.; Riyazuddin, M.; Husain, A.; Rani, H.; Praveena, K.S.; Gayen, J.R. Pharmacokinetics and brain targeting of trans-resveratrol loaded mixed micelles in rats following intravenous administration. Pharm. Dev. Technol. 2019, 25, 300–307. [Google Scholar] [CrossRef]
  169. Guo, L.; Peng, Y.; Yao, J.; Sui, L.; Gu, A.; Wang, J. Anticancer Activity and Molecular Mechanism of Resveratrol–Bovine Serum Albumin Nanoparticles on Subcutaneously Implanted Human Primary Ovarian Carcinoma Cells in Nude Mice. Cancer Biother. Radiopharm. 2010, 25, 471–477. [Google Scholar] [CrossRef]
  170. Lian, B.; Wu, M.; Feng, Z.; Deng, Y.; Zhong, C.; Zhao, X. Folate-conjugated human serum albumin-encapsulated resveratrol nanoparticles: Preparation, characterization, bioavailability and targeting of liver tumors. Artif. Cells Nanomed. Biotechnol. 2019, 47, 154–165. [Google Scholar] [CrossRef]
  171. Jadhav, P.; Bothiraja, C.; Pawar, A. Resveratrol-piperine loaded mixed micelles: Formulation, characterization, bioavailability, safety and in vitro anticancer activity. RSC Adv. 2016, 6, 112795–112805. [Google Scholar] [CrossRef]
  172. Suktham, K.; Koobkokkruad, T.; Wutikhun, T.; Surassmo, S. Efficiency of resveratrol-loaded sericin nanoparticles: Promising bionanocarriers for drug delivery. Int. J. Pharm. 2018, 537, 48–56. [Google Scholar] [CrossRef]
  173. Hao, J.; Tong, T.; Jin, K.; Zhuang, Q.; Han, T.; Bi, Y.; Wang, J.; Wang, X. Folic acid-functionalized drug delivery platform of resveratrol based on pluronic 127/d-alpha-tocopheryl polyethylene glycol 1000 succinate mixed micelles. Int. J. Nanomed. 2017, 12, 2279–2292. [Google Scholar] [CrossRef]
  174. De la Lastra, C.A.; Villegas, I. Resveratrol as an antioxidant and pro-oxidant agent: Mechanisms and clinical implications. Biochem. Soc. Trans. 2007, 35, 1156–1160. [Google Scholar] [CrossRef]
  175. Pervaiz, S.; Holme, A.L. Resveratrol: Its biologic targets and functional activity. Antioxid. Redox Signal. 2009, 11, 2851–2897. [Google Scholar] [CrossRef]
  176. Martins, L.A.M.; Coelho, B.P.; Behr, G.; Pettenuzzo, L.F.; Souza, I.C.C.; Moreira, J.C.F.; Borojevic, R.; Gottfried, C.; Guma, F.C.R. Resveratrol induces pro-oxidant effects and time-dependent resistance to cytotoxicity in activated hepatic stellate cells. Cell Biochem. Biophys. 2014, 68, 247–257. [Google Scholar] [CrossRef]
  177. Robb, E.L.; Winkelmolen, L.; Visanji, N.; Brotchie, J.; Stuart, J.A. Dietary resveratrol administration increases MnSOD expression and activity in mouse brain. Biochem. Biophys. Res. Commun. 2008, 372, 254–259. [Google Scholar] [CrossRef] [PubMed]
  178. 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. Int. 2009, 182, 128–135. [Google Scholar] [CrossRef]
  179. Li, D.D.; Han, R.M.; Liang, R.; Chen, C.-H.; Lai, W.; Zhang, J.-P.; Skibsted, L.H. Hydroxyl radical reaction with trans-resveratrol: Initial carbon radical adduct formation followed by rearrangement to phenoxyl radical. J. Phys. Chem. B 2012, 116, 7154–7161. [Google Scholar] [CrossRef] [PubMed]
  180. Yang, N.-C.; Lee, C.-H.; Song, T.-Y. Evaluation of resveratrol oxidation in vitro and the crucial role of bicarbonate ions. Biosci. Biotechnol. Biochem. 2010, 74, 63–68. [Google Scholar] [CrossRef] [PubMed]
  181. Gadacha, W.; Ben-Attia, M.; Bonnefont-Rousselot, D.; Aouani, E.; Ghanem-Boughanmi, N.; Touitou, Y. Resveratrol opposite effects on rat tissue lipoperoxidation: Pro-oxidant during day-time and antioxidant at night. Redox Rep. 2009, 14, 154–158. [Google Scholar] [CrossRef] [PubMed]
  182. Giordano, M.E.; Lionetto, M.G. Intracellular Redox Behavior of Quercetin and Resveratrol Singly and in Mixtures. Molecules 2023, 28, 4682. [Google Scholar] [CrossRef] [PubMed]
  183. Nizami, Z.N.; Aburawi, H.E.; Semlali, A.; Muhammad, K.; Iratni, R. Oxidative Stress Inducers in Cancer Therapy: Preclinical and Clinical Evidence. Antioxidants 2023, 12, 1159. [Google Scholar] [CrossRef]
  184. Szende, B.; Tyihak, E.; Kiraly-Veghely, Z. Dose-dependent effect of resveratrol on proliferation and apoptosis in endothelial and tumor cell cultures. Exp. Mol. Med. 2000, 32, 88. [Google Scholar] [CrossRef] [PubMed]
  185. San Hipolito-Luengo, A.; Alcaide, A.; Ramos-Gonzalez, M.; Cercas, E.; Vallejo, S.; Romero, A.; Talero, E.; Sanchez-Ferrer, C.F.; Motilva, V.; Peiro, C. Dual effects of resveratrol on cell death and proliferation of colon cancer cells. Nutr. Cancer 2017, 69, 1019–1027. [Google Scholar] [CrossRef]
  186. Mukherjee, S.; Dudley, J.I.; Das, D.K. Dose-dependency of resveratrol in providing health benefits. Dose Response 2010, 8, 478–500. [Google Scholar] [CrossRef]
  187. Van Ginkel, P.R.; Sareen, D.; Subramanian, L.; Walker, Q.; Darjatmoko, S.R.; Lindstrom, M.J.; Kulkarni, A.; Albert, D.M.; Polans, A.S. Resveratrol inhibits tumor growth of human neuroblastoma and mediates apoptosis by directly targeting mitochondria. Clin. Cancer Res. 2007, 13, 5162–5169. [Google Scholar] [CrossRef]
  188. Brown, V.A.; Patel, K.R.; Viskaduraki, M.; Crowell, J.A.; Perloff, M.; Booth, T.D.; Vasilinin, G.; Sen, A.; Schinas, A.M.; Piccirilli, G.; et al. Repeat dose study of the cancer chemopreventive agent resveratrol in healthy volunteers: Safety, pharmacokinetics and effect on the insulin-like growth factor axis. Cancer Res. 2010, 70, 9003–9011. [Google Scholar] [CrossRef]
  189. 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]
  190. Patel, K.R.; Scott, E.; Brown, V.A.; Gescher, A.J.; Steward, W.P.; Brown, K. Clinical trials of resveratrol. Ann. N. Y. Acad. Sci. 2011, 1215, 161–169. [Google Scholar] [CrossRef]
  191. Bode, L.M.; Bunzel, D.; Huch, M.; Cho, G.S.; Ruhland, D.; Bunzel, M.; Bub, A.; Franz, C.M.; Kulling, S.E. In vivo and in vitro metabolism of trans-resveratrol by human gut microbiota. Am. J. Clin. Nutr. 2013, 97, 295–309. [Google Scholar] [CrossRef]
  192. Ferry-Dumazet, H.; Garnier, O.; Mamani-Matsuda, M.; Vercauteren, J.; Belloc, F.; Billiard, C.; Dupouy, M.; Thiolat, D.; Kolb, J.P.; Marit, G.; et al. Resveratrol inhibits the growth and induces the apoptosis of both normal and leukemic hematopoietic cells. Carcinogenesis 2002, 23, 1327–1333. [Google Scholar] [CrossRef] [PubMed]
  193. Klinge, C.M.; Blankenship, K.A.; Risinger, K.E.; Bhatnagar, S.; Noisin, E.L.; Sumanasekera, W.K.; Zhao, L.; Brey, D.M.; Keynton, R.S. Resveratrol and estradiol rapidly activate MAPK signaling through estrogen receptors alpha and beta in endothelial cells. J. Biol. Chem. 2005, 280, 7460–7468. [Google Scholar] [CrossRef] [PubMed]
  194. 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 lifespan. Cell Metab. 2008, 8, 157–168. [Google Scholar] [CrossRef]
  195. La Porte, C.; Voduc, N.; Zhang, G.; Seguin, I.; Tardiff, D.; Singhal, N.; Cameron, D.W. Steady-state pharmacokinetics and tolerability of trans-resveratrol 2000mg twice daily with food, quercetin and alcohol (ethanol) in healthy human subjects. Clin. Pharmacokinet. 2010, 49, 449–454. [Google Scholar] [CrossRef] [PubMed]
  196. Baron, S.; Bedarida, T.; Cottart, C.H.; Vibert, F.; Vessieres, E.; Ayer, A.; Henrion, D.; Hommeril, B.; Paul, J.L.; Renault, G.; et al. Dual effects of resveratrol on arterial damage induced by insulin resistance in aged mice. J. Gerontol. A Biol. Sci. Med. Sci. 2014, 69, 260–269. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Several resveratrol-containing foodstuffs.
Figure 1. Several resveratrol-containing foodstuffs.
Nutrients 15 04486 g001
Figure 2. The chemical structure of resveratrol (cis and trans forms).
Figure 2. The chemical structure of resveratrol (cis and trans forms).
Nutrients 15 04486 g002
Figure 3. The absorption of resveratrol in the gastrointestinal tract of humans.
Figure 3. The absorption of resveratrol in the gastrointestinal tract of humans.
Nutrients 15 04486 g003
Figure 4. The health benefits of resveratrol consumption in humans.
Figure 4. The health benefits of resveratrol consumption in humans.
Nutrients 15 04486 g004
Figure 5. The current knowledge of the action of resveratrol and its prospective therapeutic mechanisms (↑) up and (↓) down; nuclear factor-kB (NF-kB), matrix metalloproteinases (MMPs), 5′-AMP-activated protein kinase (AMPK), intercellular adhesion molecule (ICAM), vascular cell adhesion molecule (VCAM), sirtuin type 1 (SIRT1), tumor necrosis factor α (TNF-α), peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α), insulin-like growth factor 1 (IGF-1), insulin-like growth factor-binding protein (IGFBP-3), ras association domain family 1 isoform A (RASSF-1α), pAkt, vascular endothelial growth factor (VEGF), cyclooxygenase 2 (COX-2), nuclear factor erythroid 2-related factor 2 (Nrf2), and Kelch-like ECH-associated protein 1 (Keap1), Homeostatic Model Assessment for Insulin Resistance (HOMA-IR), low-density lipoprotein (LDL), high-density lipoprotein (HDL), wingless-related integration site (Wnt), B-cell lymphoma 2 (Bcl-2), Bcl-2-associated X protein (Bax).
Figure 5. The current knowledge of the action of resveratrol and its prospective therapeutic mechanisms (↑) up and (↓) down; nuclear factor-kB (NF-kB), matrix metalloproteinases (MMPs), 5′-AMP-activated protein kinase (AMPK), intercellular adhesion molecule (ICAM), vascular cell adhesion molecule (VCAM), sirtuin type 1 (SIRT1), tumor necrosis factor α (TNF-α), peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α), insulin-like growth factor 1 (IGF-1), insulin-like growth factor-binding protein (IGFBP-3), ras association domain family 1 isoform A (RASSF-1α), pAkt, vascular endothelial growth factor (VEGF), cyclooxygenase 2 (COX-2), nuclear factor erythroid 2-related factor 2 (Nrf2), and Kelch-like ECH-associated protein 1 (Keap1), Homeostatic Model Assessment for Insulin Resistance (HOMA-IR), low-density lipoprotein (LDL), high-density lipoprotein (HDL), wingless-related integration site (Wnt), B-cell lymphoma 2 (Bcl-2), Bcl-2-associated X protein (Bax).
Nutrients 15 04486 g005
Table 1. A summary of resveratrol’s various pharmacological properties.
Table 1. A summary of resveratrol’s various pharmacological properties.
PharmacologyReference
Anticancer[54,55,56]
Analgesic and Anti-inflammatory[57,58,59]
Anti-diabetic[60,61,62]
Neuroprotection[63,64,65]
Antiviral[66,67,68]
Anti-obesity[69,70,71]
Cardioprotection[72,73,74]
Antioxidant[75,76,77]
Anti-aging[78,79,80]
Nephroprotection[81,82,83]
Table 2. A review of the clinical trials on resveratrol’s potential as a therapeutic molecule.
Table 2. A review of the clinical trials on resveratrol’s potential as a therapeutic molecule.
Clinical ConditionCohort Size (Numbers) Resveratrol Dose and DurationPrincipal Outcomes of Resveratrol TreatmentReference
AtherosclerosisIndividuals diagnosed with nonalcoholic fatty liver disease were randomly assigned to either a placebo (n = 25) or resveratrol (n = 25) group600 mg/day, 84 daysPlasma ox-LDL, LDL-C/HDL-C, and LDL-C/ox-LDL levels showed no changes[111]
Individuals in good health were randomly assigned to either a resveratrol (n = 24) or a calorie restriction (n = 24) group500 mg/day, 30 daysA rise in plasma TC and non-HDL cholesterol but no change in plasma TG, HDL-C, LDL-C, or apolipoprotein A1[112]
Randomized groups of patients with carotid stenosis >70% and a request for surgical intervention were given either Cardioaspirin® and Aterofisiol® (n = 107) or Cardioaspirin® and placebo (n = 107)20 mg/day, 25 daysDecreased dry weight of lipid and cholesterol in removed plaques (0.232 ± 0.018 vs. 0.356 ± 0.022; 0.036 ± 0.006 vs. 0.053 ± 0.007 mg/mg dry weight, respectively)[113]
Randomized placebo (n = 28) and resveratrol (n = 28) groups of patients with type 2 diabetes mellitus and coronary heart disease500 mg/day, 30 daysNo change in plasma TG, TC, or LDL-C; HDL-C plasma levels increased; TC/HDL-C plasma levels dropped[114]
Stable coronary artery disease patients (n = 10) were given placebo or resveratrol treatments330 mg/day, 3 daysCoronary artery bypass graft patients had higher FMD than those who had undergone percutaneous coronary intervention, whereas percutaneous coronary intervention patients showed no difference in FMD[115]
HypertensionPatients with hypertension (n = 24) given a placebo or resveratrol300 mg, acute supplementationIncreased FMD in women and individuals with higher LDL-C[116]
Patients with hypertension (n = 18) given a placebo or isolated phytochemicals60 mg/day, 28 daysDecreased diastolic blood pressure[117]
Peripheral artery disease patients were split into two groups and given either a standard balloon angioplasty (n = 75) or a resveratrol drug-coated balloon (n = 78)0.9 µg/mm2, 728 daysTarget lesion revascularization was reduced, and patients were able to walk further after treatment than those who received standard balloon angioplasty[118]
DiabetesA prospective, open-label, randomized controlled experiment involving 62 patients with type 2 diabetes250 mg/day, 90 daysDecreases in hemoglobin A1c, systolic blood pressure, total cholesterol, and total protein indicate better glycemic control[119]
Placebo-treated (n = 38) and resveratrol-treated (n = 38) patients with type 2 diabetes1000 mg/day, 56 daysChanges in plasma HDL-C, TG, TC, and LDL-C were not significant, whereas plasma glucose was reduced[120]
A randomized, placebo-controlled, double-blind investigation of 19 patients with type 2 diabetes5 mg twice daily, 30 daysGlucose and insulin levels dropped, glucose spikes after meals were postponed, and ortho-tyrosine was excreted in the urine[121]
Nonalcoholic fatty liver disease patients who were overweight and randomly assigned to either a placebo (n = 8) or resveratrol (n = 8) group1500 mg/day, 180 daysVery low-density lipoprotein TG secretion. Oxidation, and clearance rates were not affected, neither at baseline nor in response to insulin[122]
Type 2 diabetes patients in whom the disease is under control (n = 17) were given either placebo or resveratrol150 mg/day, 30 daysInsulin sensitivity in the liver and the rest of the body did not change, nor did the amount of fat stored in the liver[123]
Placebo- and resveratrol-treated patients with type 2 diabetes (n = 14)1000 mg/day, 35 daysGlycemic control and glucagon-like peptide 1 secretion did not vary[124]
Treatment with resveratrol or a placebo in elderly people with glucose intolerance (n = 30)2–3 g/day, 42 daysReactive hyperemia index rises, but blood pressure and plasma lipid levels remain unchanged[125]
Diabetic patients at high risk (n = 8) treated with placebo and resveratrol150 mg/day, 34 daysThere was no difference in the absorption of 18F-fluorodeoxyglucose or the inflammation of arteries[126]
ObesityChildren and adolescents with obesity were split into two groups: those who took a resveratrol supplement (n = 16) and those who took a placebo (n = 11)20 mg/day, 180 daysEnhanced hyperemic delta flow 6 months after post-occlusive release[127]
Obese older people (n = 22) were divided into two groups: those given placebo or resveratrol with curcumin200 mg, 30 min before consuming the high-fat mealPost-meal soluble vascular cell adhesion molecule-1 response was reduced, but other inflammatory indicators and adhesion molecules in the blood were unaffected[128]
Placebo (n = 10), 300 mg (n = 10), and 1000 mg (n = 9) resveratrol groups were used to test the effects of resveratrol on the weight and health of older, overweight persons300 and 1000 mg/day, 90 daysThe 1000 mg resveratrol group had higher levels of soluble vascular cell adhesion molecule-1 and total plasminogen activator inhibitor than the 300 mg resveratrol and placebo groups[129]
Neurodegenerative diseases102 people with early-onset Huntington’s disease (HD)40 mg twice a day, 365 daysNot known yet[130]
120 patients with mild to moderate dementia most likely due to Alzheimer’s disease (AD)500 mg/day with dose escalation of up to 1000 mg twice/day, 365 daysNausea, weight loss, and diarrhea are the only reported side effects of resveratrol, which is safe and well-tolerated. CSF Aβ40 and Aβ42 biomarkers show no improvement. Enhanced decline in brain volume[131]
27 people with mild to moderate ADResveratrol, glucose, and malate supp. delivered in grape juice, 365 daysAt modest doses, resveratrol is safe and well-tolerated. The Mini-Mental State Exam and the AD Assessment Scale for Cognition scores did not significantly change[130]
Cancer14 patients with prostate cancer500, 1000, 2000, 3000, or 4000 mg of MPX. Every 500 mg MPX has 4.4 μg resveratrol, 60–930 days (depending on the patient)Increased PSADT[132]
A single-center, randomized, placebo-controlled trial of 66 people with prostate cancer150 mg or 1000 mg daily, 120 daysA drop in androstenedione, and dehydroepiandrosterone (DHEAS). The PSA and prostate size remained unchanged[133]
Phase 1 trial of nine patients with colorectal cancer; randomized, placebo-controlled, double-blind5.0 g SRT501, 14 days before surgeryElevated levels of activated caspase-3 (apoptosis)[134]
Cases of colorectal cancer in 20 patients500 or 1000 mg, 8 days prior to surgeryKi-67 staining decreases, indicating a decrease in tumor cell growth[135]
Randomized, double-blind, placebo-controlled clinical study for breast cancer in 39 people5 or 50 mg twice daily, 90 daysDecreased RASSF-1α methylation[136]
TG: triglyceride; TC: total cholesterol; HDL-C: high-density lipoprotein cholesterol; ox-LDL: oxidized low-density lipoprotein; LDL-C: low-density lipoprotein cholesterol; FMD: flow-mediated dilatation; PSA: prostate-specific antigen; MPX: pulverized muscadine grape skin, which contains resveratrol; PSA: doubling time.
Table 3. Recent nanoformulation-based advances in boosting resveratrol’s biological activity.
Table 3. Recent nanoformulation-based advances in boosting resveratrol’s biological activity.
Nanoformulation MethodStudy ModelOutcomeReference
Resveratrol medication delivery systems based on self-emulsificationIn vitro, in vivo (rats)Enhanced pharmacokinetics, decreased metabolism, and enhanced solubility[161]
Micellar solubilization of resveratrolTwelve healthy volunteers (oral administration)Increased oral bioavailability[162]
Suspension of free resveratrol, resveratrol-filled nanoparticles, and layer-by-layer nanoparticlesIn vivo (Wistar rats, oral administration, 20 mg/kg)Systemic exposure was increased when resveratrol was encapsulated in layer-by-layer nanoparticles with resveratrol nanocores[163]
Nanoparticle delivery method based on oat-shellac proteinsIn vitro, in vivo (rat model)Resveratrol was buffered in the stomach acid and released gradually into the small intestine.
Transport and absorption by cells are enhanced relative to free resveratrol. Enhancement in bioavailability
[164]
Resveratrol nanoencapsulation in caseinIn vitro, in vivo (rats)Oral administration in rats: remained in the gut and reached intestinal epithelium.
Produced high plasma levels of resveratrol (sustained for at least 8 h) and similar results for its metabolites.
Oral bioavailability was 10 times higher compared to an oral solution of resveratrol
[165]
Trans-resveratrol nanocrystalsIn vitro, in vivo (rats)Increased oral bioavailability[166]
Nanoparticles of human serum albumin coupled with glycyrrhizic acid and loaded with resveratrolIn vivo (rats; single-dose tail vein injection)The absorption rate of resveratrol was increased.
High levels of resveratrol were found in most of the rats’ vital organs. The highest levels were found in the liver, suggesting that a delivery method that focuses on the liver could be effective
[167]
Trans-resveratrol-loaded mixed micellesIn vivo (rats; intravenous administration)Enhanced pharmacokinetic parameters.
Improved brain targeting
[168]
Resveratrol bovine serum albumin nanoparticles (RES-BSANP)In vivo (nude mice; intraperitoneal injection)Enhanced dilution and soluble in water. Cancer development was suppressed in hairless mice bearing human ovarian primary tumors[169]
Folate-conjugated HSA nanoparticlesHePG2 liver cancer cellsShowed decreased resveratrol release and increased cytotoxicity[170]
Piperine-loaded mixed micellesMCF-7 breast cancer cellsImproved cytotoxicity[171]
Sericin nanoparticlesCaco-2 cells colorectal cancer cellsStrong cytotoxic against Caco-2 cells[172]
Folic acid-targeted micellesMCF-7 breast cancer cellsIncreased cytotoxicity was achieved due to the sustained release of encapsulated resveratrol provided by the nano-formulation.[173]
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

Farhan, M.; Rizvi, A. The Pharmacological Properties of Red Grape Polyphenol Resveratrol: Clinical Trials and Obstacles in Drug Development. Nutrients 2023, 15, 4486. https://doi.org/10.3390/nu15204486

AMA Style

Farhan M, Rizvi A. The Pharmacological Properties of Red Grape Polyphenol Resveratrol: Clinical Trials and Obstacles in Drug Development. Nutrients. 2023; 15(20):4486. https://doi.org/10.3390/nu15204486

Chicago/Turabian Style

Farhan, Mohd, and Asim Rizvi. 2023. "The Pharmacological Properties of Red Grape Polyphenol Resveratrol: Clinical Trials and Obstacles in Drug Development" Nutrients 15, no. 20: 4486. https://doi.org/10.3390/nu15204486

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