Next Article in Journal
The Effect of Caloric Restriction with and without n-3 PUFA Supplementation on Bone Turnover Markers in Blood of Subjects with Abdominal Obesity: A Randomized Placebo-Controlled Trial
Previous Article in Journal
Human Breast Milk Composition and Function in Human Health: From Nutritional Components to Microbiome and MicroRNAs
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Enhancing Bioavailability of Nutraceutically Used Resveratrol and Other Stilbenoids

by
Ondrej Vesely
1,*,
Simona Baldovska
2 and
Adriana Kolesarova
3,*
1
Department of Food Science, The Faculty of Agrobiology, Food and Natural Resources, The Czech University of Life Sciences Prague, 16500 Prague, Czech Republic
2
AgroBioTech Research Centre, Slovak University of Agriculture in Nitra, 94976 Nitra, Slovakia
3
Department of Animal Physiology, Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, 94976 Nitra, Slovakia
*
Authors to whom correspondence should be addressed.
Nutrients 2021, 13(9), 3095; https://doi.org/10.3390/nu13093095
Submission received: 27 July 2021 / Revised: 29 August 2021 / Accepted: 30 August 2021 / Published: 2 September 2021
(This article belongs to the Section Phytochemicals and Human Health)

Abstract

:
Stilbenoids are interesting natural compounds with pleiotropic in vitro and in vivo activity. Their well-documented biological properties include anti-inflammatory effects, anticancer effects, effects on longevity, and many others. Therefore, they are nowadays commonly found in foods and dietary supplements, and used as a part of treatment strategy in various types of diseases. Bioactivity of stilbenoids strongly depends on different types of factors such as dosage, food composition, and synergistic effects with other plant secondary metabolites such as polyphenols or vitamins. In this review, we summarize the existing in vitro, in vivo, and clinical data from published studies addressing the optimization of bioavailability of stilbenoids. Stilbenoids face low bioavailability due to their chemical structure. This can be improved by the use of novel drug delivery systems or enhancers, which are discussed in this review. Current in vitro and in vivo evidence suggests that both approaches are very promising and increase the absorption of the original substance by several times. However, data from more clinical trials are required.

1. Introduction

Stilbenoids are part of a large group of phytochemicals known as polyphenols. Epidemiological studies have found an association between food consumption and health and longevity. Stilbenoids have become well known due to the compound resveratrol [1]. This molecule is well known for its association in explaining the lower mortality in France compared with the American population. This phytoalexin has helped to focus research on other similar compounds that are based on stilbene and slightly differ only in the numbers of hydroxyl groups, isomerization, glycosylation, styrene bond, methyl groups, prenyl groups, etc. The double bond is responsible for isometry, with the trans- form being more common from a steric point of view. Furthermore, isomerization has a strong influence on biological properties, and therefore it is important to use only the most potent variant in nutraceuticals [2,3,4,5].
Epidemiological studies have tried to find associations between food consumption and health effects in several ways [6]. There is a consensus that the intake of plant-based foods such as vegetables, fruits, nuts, grains, legumes, spices, roots, and leaves has an impact on health, especially in long-term and age-related diseases [7]. As the population ages, diseases such as cardiovascular disease, cancer, and type II and III diabetes become more common, especially in the countries that consume a Western diet [8,9]. The combination of aging and a significant shift in dietary habits from conventional foods to more convenient foods is probably behind the recommendations by a number of organizations such as the U.S. Department of Health and Human Services (DHHS) [10] and WHO/FAO, or country- and region-specific guidelines, namely, the Nordic Nutrition Recommendation, Pan American Health Organization (PAHO), and the Institute of Nutrition of Central America and Panama (INCAP) to incorporate more plants in the diet through the Food-Based Dietary Guidelines [11].
An alternative to plant-based diets are nutraceuticals that contain phytonutrients with biological properties, such as stilbenoids. These compounds are well studied for their anti-inflammatory properties [12], cardioprotective properties [13], and antidiabetic properties [14], all of which are related to age-related diseases. As in vivo experiments do not always correspond to in vitro experiments, more recent studies have focused on determining the effect of the low bioavailability of these compounds.
The aim of this review is to summarize the pharmacokinetics of the best-known stilbenoid resveratrol and the approach to modifying its bioavailability.

2. Chemical Properties

Stilbenoids are naturally occurring phenolic substances, classified as phytoalexins, produced de novo in plants to protect against fungal infection and toxins and also to inhibit bacterial growth [15]. Resveratrol (3,5,4′-trihydroxy-trans-stilbene; RES, Figure 1), the most widely studied stilbenoid, is present in foods such as the skin of grapes, raspberries, blueberries, and peanuts [16]. RES has been known since its isolation from Veratrum grandiflorum by the Japanese scientist Michio Takaoka (of Imperial University of Hokkaido) in 1939 [17]. Its structure consists of two phenolic rings bonded together by a double styrene bond. This double bond is responsible for stereoisomerization, and thus two isomeric forms (cis- and trans-) are known, with the trans- form more common and more biologically active in comparison to cis- form [18]. On the other hand, another study showed the cis- form as a better inhibitor of tumor cells [19], so there is good evidence that isomerization of this molecule has an impact on the bioactivity.
The use of RES as a nutraceutical is limited due to its poor water solubility (<0.05 mg/mL), low oral bioavailability, and high chemical instability. In particular, the problem of low bioavailability of RES is crucial [20,21]. The absorption rate is approximately 75% (25 mg oral dose) [22,23], but it is questionable if this rate will be the same at higher doses. The absorption of RES by different types of cells was confirmed by several research groups [3,24,25,26]. Although the absorption is high, the bioavailability is poor [22]. RES is rapidly transformed by phase II metabolism. RES conjugates are rapidly absorbed through the gastrointestinal tract and are detectable in serum in 30–60 minutes after ingestion [22,23]. Moreover, there is evidence that the circadian rhythm can influence bioavailability [27] as well as the type of meal [28]. The time of ingestion of substances can strongly influence their bioavailability and rate of absorption. Questions regarding the ideal time of drug administration are addressed by chronopharmacokinetic studies [29]. To increase bioavailability, the best time to administer RES is in the morning [27]. The question of whether the intake of 400 mg of resveratrol can be influenced by a high-fat meal was investigated by Vaz-da-Silva et al. [30]. Their results showed that the rate of RES absorption was significantly delayed by the presence of food compared to fasting conditions. However, it did not have any significant effect on overall bioavailability.
Important is the loading or dosage, as shown by another study [31]. RES concentrations vary significantly between studies, from nanomolar to millimolar, and dosage of more than 100 mg/day is considered important. Dose is a significant factor in the manifestation of the effect of RES, with smaller doses having a beneficial effect, while larger doses may be harmful [32]. Simply put, the tolerance to RES is cell-specific and varies depending on various organs; thus, it is difficult to define a safe dose limit. However, clinical studies suggest that relatively high doses of RES (2 g/day or more) begin to have non-life-threatening side effects such as diarrhea, nausea, and hypersensitivity [33], or frontal headache [27]. Finally, the functional and safe window of RES eventually lies somewhere between 100 and 1000 mg/day.

3. Physiological and Therapeutic Effects

Stilbenoids possess numerous biological effects, including cardioprotective, neuroprotective, anti-inflammatory, and antidiabetic properties, and they can play a role in depigmentation, cancer prevention, and treatment [15]. RES exerts anticancer and antiproliferative, anti-aging, and antineoplastic activity in vitro and in vivo [34,35,36]. Antimicrobial activity has also been demonstrated against bacterial, viral, and fungal species, mainly in vitro [37]. Moreover, RES and other stilbenoids have well-studied anti-inflammatory properties in vitro and in vivo [12]. Inflammation is a process vital for survival and it is fundamentally a part of the defense against pathogens and serves as a response to tissue damage. It is normally recognized by immune cells (leukocytes, macrophages, mast cells, and neutrophils), and they release reparative agents at the site of inflammation, including cytokines, histamine, leukotrienes, nitric oxide, and prostaglandins. Three main pathways are known: NF-κB, MAPK, and JAK-STAT. As a result of inflammation, too many free oxygen radicals are produced. With prolonged inflammation, lipids, proteins, and nucleic acids can be damaged, which can lead to various types of diseases, such as atherosclerosis, cancer, inflammatory bowel disease, kidney diseases, lung diseases, liver diseases, and CNS diseases [38]. In chronic inflammation, RES is a promising regulator, protector, and therapeutic agent in the prevention of cardiovascular [39,40] and metabolic diseases, including obesity [40,41,42], as well as reproductive disorders [36,43] and reprotoxicity [44]. Furthermore, this bioactive molecule can extend the lifespan and ameliorate aging-related phenotypes, attenuate the negative effects of a high-calorie diet, mimicking the effects of calorie restriction, and improve cellular function and metabolic health in general [41]. Therefore, stilbenoids, such as RES, possess several physiological actions that can be useful for prevention, mitigation, and treatment of numerous diseases. Even small doses of RES can have an effect, as in the case of 10 mg RES, which significantly decreased insulin resistance via the Akt pathway, which led to improved insulin sensitivity in type II diabetes patients [45].
Although many studies describe the in vitro and in vivo modes of action of RES, some studies experimentally show the opposite action. For example, a study that tried to influence longevity in Drosophila failed [46]. In this case, the authors attributed the results to normal conditions. When Baur et al. attempted to prolong the life of rats, only those fed a high-fat diet had an effect [47]. These examples point to the influence of an individual’s health as a significant factor affecting the potency of RES. A meta-analysis of nine studies ( n = 208; RES dosage, 75–3000 mg/day; duration, minimum of 2 weeks) found no significant changes in body mass index or body weight [48]. As RES should activate adenosine monophosphate-activated protein kinase (AMPK) and thus induce a fasting state, weight loss would be expected. However, this has not been observed by all researchers. Different study results may be due to the differential period of dosing [49].
Thus, we can observe a significant influence of various factors such as age, health, duration and amount of the dose, and diet composition on the observed parameters. Since RES affects many cellular mechanisms, the methodology of the experiments needs to be adapted to the different mechanisms of action.

4. Mechanism of Actions

RES can affect numerous cellular processes via multiple intracellular signaling pathways [50]. Mechanisms of actions include stress signaling pathway (MKP-1) and reduction of oxidative stress and inflammation (iNOS, COX-2 activity, and ROS generation), inflammation factors and malondialdehyde levels [51], as well as inhibition of prohypertrophic molecules, improvement of myocardial Ca2+ handling, and phosphorylation of prosurvival Akt-1 and GSK-3β [52]. Moreover, RES can prevent the expression of endothelial nitric oxide synthase and vascular endothelial growth factor, and suppress phosphorylation of p38 in rats with diabetes-related myocardial infarction. Additionally, the antioxidant activity of RES is associated with anti-aging effect and ability to affect apoptosis, growth, and carcinogenesis [36,43,53]. The binding of RES to various target molecules, which can be intracellular mediators of RES action, includes tubulin, protein kinase C alpha (PKCα), phosphodiesterase-4D [54], adenosine monophosphate kinase, nuclear factor-κB (NF-kB), inflammatory cytokines, antioxidant enzymes, and others [34]. Furthermore, RES can bind and affect DNA methyltransferases and proteins responsible for DNA methylation, which can be involved in epigenetic regulation of oxidative, metabolic, inflammatory, angiogenic, and tumorigenic processes [55].
RES is known as an activator of Sirtuin 1 (SIRT1), a NAD+-dependent histone deacetylase contributing to oxidative stress, aging, metabolism, obesity, and tumors, as well as to DNA methylation [50]. A meta-analysis of 11 randomized controlled trials found a significant influence of RES as a treatment for diabetic parameters such as fasting glucose, insulin, glycated hemoglobin, and insulin resistance in patients with diabetes mellitus (DM) type II [56]. Interestingly, these results are not seen in patients without comorbidities. Researchers have discussed mainly the following mechanisms. First, RES is known as an activator of SIRT 1, thus mimicking the caloric restriction and providing a beneficial effect on glucose control [57,58]. Secondly, RES can increase the expression of the insulin-regulated glucose transporter protein (GLUT4), which is an important factor for modulating insulin resistance [59,60] and improving glucose uptake in the absence of insulin [61]. Because RES can directly alter the secretion of insulin by the pancreas, it may explain the large effect on DM II [62]. All these mechanisms of action can influence a wide array of cellular processes via multiple intracellular signaling pathways.

5. Pharmacokinetic Characteristics of Stilbenoids

Because the efficacy of orally administrated stilbenoids depends on their absorption, metabolism, and tissue distribution, it is important to understand their overall pathway.

5.1. Pharmacokinetics in Animals

To our knowledge, the first study in rats demonstrated absorption of RES from the gastrointestinal tract way back in 1996 [63]. In their experiment, Bertelli et al. added known RES content (6.5 mg/L) in the form of red wine. The results of the study showed that the RES contained in the wine was rapidly absorbed and was detectable within 30 min, peaking at approximately 60 min after ingestion of the wine [64].
Another study in rats [65] described the rapid absorption of RES in blood and serum with a calculated absorption at 50–75%. Moreover, the type of matrix (ethanol, V-8 vegetable cocktail, and white grape juice) did not increase absorption. In addition, trace amounts in the liver, kidney, heart, and spleen were found. Another study confirmed these results in mice but found RES at high concentration in urine and bile [66]. Three hours after administration, RES was detectable in various organs, with the highest concentration in the duodenum, lower in the kidneys, followed by the lungs and liver. In rats, RES is metabolized in the liver by cytochrome P450. Common metabolites are trans-resveratrol-3-O-glucuronide and trans-resveratrol-3-O-sulphate [67]. In another study, differences between dosage and metabolites were found, and higher dosage (300 mg/kg body weight) led to further of trans-resveratrol-3,4’,5-trisulphate [68]. These were detectable in plasma, liver, and kidney samples.
The animal model close to human is pig [69]. In this study, twelve resveratrol and seven dihydroresveratrol metabolites, all of them close to those of previous studies (trans- and cis-resveratrol, nine resveratrol conjugates such as glucuronide, sulphate, and sulphoglucuronide derivatives), the microbiota-derived metabolite dihydroresveratrol, and six conjugates (also glucuronide, sulphate, and sulphoglucuronide derivatives) were detected. Approximately 74.5% (351.6 mg) of the total RES (472 mg) administered was recovered in the form of RES, dihydroresveratrol, and derived metabolites (65.1% along the gastrointestinal tract, 7.7% in urine, 1.2% in bile, and 0.5% in organs). The authors considered that the results obtained may vary depending on the time of sampling. In their study, all the data were obtained six hours after administration of RES.
In summary, RES in animals is absorbed in the intestines, then converted to several metabolites, mainly glucuronide and sulphate, and finally excreted, mainly in the urine and stool. For a more precise approach, it is important to move from animal to clinical evidence.

5.2. Pharmacokinetics in Humans

There are studies on the pharmacokinetics of RES in humans. Goldberg et al. [23] followed up on a previous study [65]. Trans-resveratrol (25 mg/70 kg) was administered in men at four-week intervals in three different matrices: white wine (11.5% ethanol), grape juice, and vegetable juice. RES was present in serum and urine predominantly as glucuronide and sulphate conjugates, reaching peak concentrations approximately 30 min after consumption. Free RES accounted for 1.7% to 1.9%. The study demonstrated human ability to metabolize RES that was not affected by any type of matrix used [23]. A more accurate study used radioactive RES (14C RES) both orally (25 mg (110 µM)) and intravesically (0.2 mg) [22]. Most of the radioactivity after the oral dose was recovered in the urine (53–85%). The recovery in stool was highly variable (0.3–38%). The research group also compared oral dose with the i.v. dose of 0.2 mg; the recovery in urine was 42–83% and in stool 0.6–23%. One volunteer was exposed orally to 100 mg of an unlabeled dose of RES to determine the structure of RES metabolites. Five major metabolites, two monoglucuronides, dihydroresveratrol monoglucuronide, resveratrol monosulphate, and dihydroresveratrol sulphate, were detected in urine. After oral administration, 24% ± 3% of the sulphate conjugates and 13% ± 1% of the glucuronic acid conjugated were excreted in the urine. This study showed that RES is rapidly metabolized and absorbed in humans after consumption. Only 2% free RES can be detected in plasma and serum [22].
The two main reactions for the formations of metabolites in humans are glucuronidation and sulphation. The UDP-glucuronosyltransferase (UGT) family catalyzes the conjugation of RES to glucuronic acid at the 3’ or 4’ hydroxyl group. These enzymes are abundant in intestinal microsomes and in the liver. UGT1A1 and UGT1A9 were found to be predominantly responsible for the formation of 3-O-glucuronide (Km = 149 µM) and 4’-O-glucuronide (Km = 365 µM), respectively [70]. Remarkably, intestinal glucuronide conjugates formed up to 10-fold more compared to the hepatic microsomes. The second pathway commonly used by the body to eliminate polyphenols is sulphation. In the liver, RES is metabolized by sulphotransferase (SULT), where resveratrol-4’-O-sulphate is formed by SULT1E1, and resveratrol-3-O-sulphate is formed by either SULT1A2 or SULT1A3 [71]. In summary, 75% of RES passes through the apical and basolateral membranes by simple diffusion. In cells, RES undergoes rapid and extensive modification by the enzymes UGT and SULT, resulting in glucuronide and sulphate conjugates. The metabolites leave the cell via multidrug resistance protein (MRP 3—basolateral side) or breast cancer resistance protein (BRCP) and MRP2 (apical side) [72,73].
The microbiota plays an important role. The most common metabolites of the human gut microbiota are dihydroresveratrol, 3,4’- dihydroxy-trans-stilbene and lunularin [74]. In the same study, Bode et al. identified two bacterial strains, Slackia equolifaciens and Adlercreutzia equolifaciens, as dihydroresveratrol producers, which means that the composition of microbiota influences the resulting metabolites. When Jarosova et al. attempted to investigate multiple stilbenoids in a similar manner to that of Bode et al., their findings showed dihydroresveratrol as the only one bacterial metabolite [75]. Jose et al. discovered an effect of RES and other stilbenoids on an abundance of specific taxon, Faecalibacterium prausnitzii, which they consider beneficial [76]. This evidence points to the possibility of a prophylactic effect of these substances and the subsequent formation of other metabolites and influence on pharmacokinetics.
Once stilbenoids enter the circulation, they must be transported. In the bloodstream, RES and its metabolites can bind to plasma lipoprotein (LDL) and albumin and thus be transported between tissues. These complexes can be dissociated on cell membranes, leaving RES free to enter cells [77,78]. Most importantly, there is evidence that metabolites of RES can be deconjugated back to RES [79]. In this study, a human aortic endothelial cell line (HAEC) and 0.5–50 µM RES were used. This dose is similar to those obtained after pharmacological intake. However, there is still not enough information about stilbenoids and different cell types, but, as a similar pattern can be observed for other polyphenols, it is likely that this also applies to stilbenoids [80]. The fate of orally ingested RES is shown in Figure 2.
In the end, the largest amount of RES, exceeding the amount required for in vitro activity, can be detected in colon tissues, so the colon can be considered the main target organ. The efficacy in other tissues may depend on its metabolites and the ability to regenerate RES locally or systemically [81].

6. Enhancing the Bioavailability of Stilbenoids

The bioavailability of drugs can be influenced by many factors, namely, effect of pH, acid secretion, delayed gastric emptying, and enzyme activity by the stomach, the release of bile salts by gall bladder, secretion of enzymes into the small intestine and production and secretion of insulin by pancreas, bile salt concentrations, enzyme activity, viscosity and osmolality, potential inhibition of transporters and drug-metabolizing enzymes, amd potential changes in lymphatic uptake by small intestine and colon [82]. The important factor is the food matrix [83]. Proteins, dietary fiber, and minerals can negatively alter the bioavailability of flavonoids, and on the other side, the lipids, digestible carbohydrates, vitamins, alkaloids, carotenoids, etc. are likely to improve flavonoid bioavailability [83]. As flavonoids have a similar molecular structure to that of stilbenoids, we can assume similarities. Moreover, there are ways to increase the bioavailability of stilbenoids, namely, using bioenhancers and novel methods.

6.1. Bioenhancers

Bioenhancers promote and augment the bioavailability of compounds without having any activity with the drug of their own at the dose used. For example, alkaloid piperine has a molecular structure suitable for enzyme inhibition. Piperine, which is a potent inhibitor of drug metabolism, is therefore a strong enhancer of bioavailability. Piperine works through increased gastrointestinal absorption (enhancing solubility, increased blood supply, increased permeability due to epithelial cell modification, through increased microvilli length), reduction of efflux of drugs from the site of action, inhibition of solubilizer attachment, and metabolism reduction [84]. The in vitro, animal, and interventional studies involving stilbenoids and bioenhancers are summarized in Table 1. In vitro experiments using a permeability model show a significant effect of enhancers such as quercetin or piperine. These results are consistent with experiments in animal models. In contrast, they are difficult to apply to results from clinical studies. However, there is still not enough evidence of whether increased bioavailability leads to increased bioefficacy of the target compounds. Wightman et al. used a combination of 250 mg of RES and 20 mg of piperine as bioenhancer. Contrary to in vitro and animal studies, the Cmax concentration of RES alone was 9.98 µM, while with piperine it was only 4.82 µM. Based on results, researchers concluded that co-supplementation of piperine with RES can enhance the bioefficacy of RES with regard to effects on cerebral blood flow in healthy human subjects, and does this without altering the bioavailability of RES, since the plasma concentrations of RES and its metabolites were not different between the treatments [85]. In another study, the combination of 2 g RES and 500 mg quercetin two times a day was used [28]. The combination did not affect the pharmacokinetics of RES. With the current number of studies and the limited number of substances tested, it is not clearly understood as to why the results vary so much at different levels of research. For example, it is possible that although substances inhibit a particular metabolic pathway, probably another persists. Another possibility could be that, due to the complexity of the organism, the enhancer is added in insufficient amounts. More evidence is needed. A simplified operation of the best-known bioenhancers can be seen in Figure 3.
Interestingly, in mice, RES can enhance other biologically active compounds such as apigenin [91]. As resveratrol–apigenin shows, stilbenoids can alter the bioavailability of different compounds. For example, RES, which is generally accepted to be a moderate-to-weak inhibitor of cytochrome P450 (CYPs), is known as an inhibitor of CYP3A4, but inhibition of these enzymes by RES aglycons is not known, although they are more abundant in the body than RES themselves due to metabolism [92]. Because CYP3A is common in small intestine, stilbenoids can alter the first pass metabolism [93]. They also can affect the efflux transporter p-glycoprotein. This ATP-binding cassette transporter (ABC transporter) plays an important role in drug transport in many organs. For example, in the intestine, p-glycoprotein pumps drugs back into the lumen, leading to reduced absorption. P-glycoprotein transporters are more abundant in cancer cells, where they influence survival against anticancer treatment [94]. This transporter is also common in small intestine, liver, and kidney as well as part of the blood brain barrier or placenta [95]. An interesting study using C57BL/6J mice as an animal model combined RES and an alkaloid berberine, which is known for its antidiabetes properties [95]. In this case, the combination of the two agents had a significantly greater effect on cholesterol and low-density lipoprotein cholesterol (27% and 31%, respectively) compared with monotherapy with RES alone (8% and 6%, respectively) and berberine (10% and 10%, respectively). Moreover, RES was able to increase the amount of berberine intracellularly in L02 liver cells. Several studies show that RES can break multidrug resistance by altering p-glycoprotein, and therefore it is better to look at stilbenoids not only as main drugs but also as supportive drugs [96,97,98,99].

6.2. Novel Methods

Novel methods or novel drug delivery systems (NDDSs) are a way to deliver the maximum amount of drug to the target site. This can alter the duration of therapeutic activity and the bioavailability [100]. Stilbenoids can be delivered by different types of systems [101], namely, microspheres [102], microcapsules [103], self-emulsifying drug delivery system (SEDDS) [101,104], self-microemulsifying drug delivery system (SMEDDS) [105,106,107], nanosponges [108], nanoemulsions [109,110], self-nanoemulsifying drug delivery system (SNEDDS) [111,112,113,114], self-double-emulsifying drug delivery system (SDEDDS) [115], liposomes [116,117], nanoparticles [118,119,120,121,122,123,124,125,126], nanostructured lipid carriers [127], nanosuspensions [128], chitosan–gellan nanofiber [129], solidified phospholipid complex [130], spiral dextrin [131], and buccal cyclodextrin delivery system [132]. All delivery systems are extensively studied, but there is little clinical data. Novel methods are more closely discussed in a recent review by Chen et al. [133]. However, more clinical data are needed.
Few clinical studies are known to examine novel methods on humans. In the study by Calvo-Castro et al., a group of 12 subjects were given a single dose of vineatrol (30 mg RES and 75 mg viniferin) in powder or liquid micellar formulation. The maximum total RES concentrations in plasma were 10.6-fold higher when ingesting micellar compared with native vineatrol [134]. Briskey and Rao randomized 39 subjects into four groups and used a novel dispersion agent for screening to enhance RES absorption. The data showed a 2-fold increase in plasma concentration over 24 h and a 3-fold increase in Cmax of both the sulphate and glucuronide conjugates [135].
In vitro, animal, and clinical studies using novel methods to improve stilbenoids uptake promise a significant advance in bioavailability. However, there are currently insufficient studies to confirm and compare the clinical efficacy of different approaches.

7. Conclusions

In summary, stilbenoids exhibit many biological properties both in vitro and in vivo. However, due to their low solubility and bioavailability, clinical studies often do not correspond to in vitro studies. We have shown that stilbenoids are mostly metabolized in the human body to glucuronide and sulphate conjugates in the intestine and liver. These metabolites do not have similar biological properties and therefore, new delivery systems have been developed to improve bioavailability and new research has been conducted on enhancers. Only a few enhancers are known, mainly piperine, but it has not been successful in clinical trials to date. The influence of the food matrix is important for polyphenols, but data are lacking in the stilbenoids.
Finally, we can conclude that there are several promising approaches to increase the bioavailability of stilbenoids, but to date, there is insufficient evidence from human studies, and further research is required. However, recent data suggest that a combination of enhancers and novel drug delivery systems can be very effective.

Author Contributions

Conceptualization, O.V., S.B., and A.K.; writing—original draft preparation, O.V., S.B., and A.K.; editing, O.V., S. B., and A.K..; supervision, A.K. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Ministry of Education, Science, Research and Sport of the Slovak Republic projects APVV-18-0312, DS-FR-19-0049, VEGA 1/0266/20, KEGA 033SPU-4/2021, the Operational Programme Integrated Infrastructure within the project: Demand-driven research for the sustainable and innovative food, Drive4SIFood 313011V336, co-financed by the European Regional Development Fund, and ERASMUS+ Programme of the European Union KA2 2020-1-SK01-KA203-078363.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

We wish to thank Shubhadeep Roychoudhury for language editing and proofreading.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Renaud, S.; De Lorgeril, M. Wine, alcohol, platelets, and the French paradox for coronary heart disease. Lancet 1992, 339, 1523–1526. [Google Scholar] [CrossRef]
  2. Abou-Zeid, L.A.; El-Mowafy, A.M. Differential recognition of resveratrol isomers by the human estrogen receptor-α: Molecular dynamics evidence for stereoselective ligand binding. Chirality 2004, 16, 190–195. [Google Scholar] [CrossRef]
  3. Jarosova, V.; Vesely, O.; Doskocil, I.; Tomisova, K.; Marsik, P.; Jaimes, J.D.; Smejkal, K.; Kloucek, P.; Havlik, J. Metabolism of cis- and trans-Resveratrol and Dihydroresveratrol in an Intestinal Epithelial Model. Nutrients 2020, 12, 595. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Jhanji, M.; Rao, C.N.; Sajish, M. Towards resolving the enigma of the dichotomy of resveratrol: Cis- and trans-resveratrol have opposite effects on TyrRS-regulated PARP1 activation. GeroScience 2020, 43, 1171–1200. [Google Scholar] [CrossRef] [PubMed]
  5. Rius, C.; Abu-Taha, M.; Hermenegildo, C.; Piqueras, L.; Cerda-Nicolas, J.-M.; Issekutz, A.C.; Estañ, L.; Cortijo, J.; Morcillo, E.J.; Orallo, F.; et al. Trans- but Not Cis-Resveratrol Impairs Angiotensin-II–Mediated Vascular Inflammation through Inhibition of NF-κB Activation and Peroxisome Proliferator-Activated Receptor-γ Upregulation. J. Immunol. 2010, 185, 3718–3727. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Freudenheim, J.L. A Review of Study Designs and Methods of Dietary Assessment in Nutritional Epidemiology of Chronic Disease. J. Nutr. 1993, 123, 401–405. [Google Scholar] [CrossRef]
  7. World Health Organization. Healthy Diet. No. WHO-EM/NUT/282/E; World Health Organization Regional Office for the Eastern Mediterranean: Albany, NY, USA, 2019. [Google Scholar]
  8. Chang, A.Y.; Skirbekk, V.F.; Tyrovolas, S.; Kassebaum, N.J.; Dieleman, J.L. Measuring population ageing: An analysis of the Global Burden of Disease Study 2017. Lancet Public Heal. 2019, 4, e159–e167. [Google Scholar] [CrossRef] [Green Version]
  9. Romagnolo, D.F.; Selmin, O.I. Mediterranean Diet and Prevention of Chronic Diseases. Nutr. Today 2017, 52, 208–222. [Google Scholar] [CrossRef] [Green Version]
  10. US Department of Agriculture; US Department of Health and Human Services. Dietary Guidelines for Americans; 2020-2025; US Department of Agriculture; US Department of Health and Human Services: Washington, DC, USA, 2020.
  11. Herforth, A.; Arimond, M.; Álvarez-Sánchez, C.; Coates, J.; Christianson, K.; Muehlhoff, E. A Global Review of Food-Based Dietary Guidelines. Adv. Nutr. 2019, 10, 590–605. [Google Scholar] [CrossRef] [Green Version]
  12. Dvorakova, M.; Landa, P. Anti-inflammatory activity of natural stilbenoids: A review. Pharmacol. Res. 2017, 124, 126–145. [Google Scholar] [CrossRef]
  13. Zordoky, B.N.; Robertson, I.M.; Dyck, J.R. Preclinical and clinical evidence for the role of resveratrol in the treatment of cardiovascular diseases. Biochim. Biophys. Acta Mol. Basis Dis. 2015, 1852, 1155–1177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Oyenihi, O.R.; Oyenihi, A.B.; Adeyanju, A.A.; Oguntibeju, O.O. Antidiabetic Effects of Resveratrol: The Way Forward in Its Clinical Utility. J. Diabetes Res. 2016, 2016, 9737483. [Google Scholar] [CrossRef] [PubMed]
  15. Akinwumi, B.C.; Bordun, K.-A.M.; Anderson, H.D. Biological Activities of Stilbenoids. Int. J. Mol. Sci. 2018, 19, 792. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Tian, B.; Liu, J. Resveratrol: A review of plant sources, synthesis, stability, modification and food application. J. Sci. Food Agric. 2019, 100, 1392–1404. [Google Scholar] [CrossRef] [PubMed]
  17. Takaoka, M.J. Of the Phenolic Substrate of White Hellebore (Veratrum Grandiflorum Loes. Fil.). J. Fac. Sci. Hokkaido Imper. Univ. 1940, 3, 1–16. [Google Scholar]
  18. Anisimova, N.Y.; Kiselevsky, M.V.; Sosnov, A.V.; Sadovnikov, S.V.; Stankov, I.N.; Gakh, A.A. Trans-, cis-, and dihydro-resveratrol: A comparative study. Chem. Central J. 2011, 5, 88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Cardile, V.; Chillemi, R.; Lombardo, L.; Sciuto, S.; Spatafora, C.; Tringali, C. Antiproliferative Activity of Methylated Analogues of E- and Z-Resveratrol. Zeitschrift für Naturforschun 2007, 62, 189–195. [Google Scholar] [CrossRef]
  20. Boocock, D.; Faust, G.E.; Patel, K.R.; Schinas, A.M.; Brown, V.A.; Ducharme, M.P.; Booth, T.D.; Crowell, J.A.; Perloff, M.; Gescher, A.J.; et al. Phase I Dose Escalation Pharmacokinetic Study in Healthy Volunteers of Resveratrol, a Potential Cancer Chemopreventive Agent. Cancer Epidemiol. Biomark. Prev. 2007, 16, 1246–1252. [Google Scholar] [CrossRef] [Green Version]
  21. Smoliga, J.M.; Blanchard, O. Enhancing the Delivery of Resveratrol in Humans: If Low Bioavailability is the Problem, What is the Solution? Molecules 2014, 19, 17154–17172. [Google Scholar] [CrossRef] [PubMed]
  22. Walle, T.; Hsieh, F.; DeLegge, M.H.; Oatis, J.E.; 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] [Green Version]
  23. Goldberg, D.M.; Yan, J.; Soleas, G.J. Absorption of three wine-related polyphenols in three different matrices by healthy subjects. Clin. Biochem. 2003, 36, 79–87. [Google Scholar] [CrossRef]
  24. Willenberg, I.; Michael, M.; Wonik, J.; Bartel, L.C.; Empl, M.T.; Schebb, N.H. Investigation of the absorption of resveratrol oligomers in the Caco-2 cellular model of intestinal absorption. Food Chem. 2015, 167, 245–250. [Google Scholar] [CrossRef] [PubMed]
  25. Kuhnle, G.; Spencer, J.P.; Chowrimootooc, G.; Schroeter, H.; Debnam, E.S.; Srai, S.S.; Rice-Evans, C.; Hahn, U. Resveratrol Is Absorbed in the Small Intestine as Resveratrol Glucuronide. Biochem. Biophys. Res. Commun. 2000, 272, 212–217. [Google Scholar] [CrossRef]
  26. Andlauer, W.; Kolb, J.; Siebert, K.; Fürst, P. Assessment of Resveratrol Bioavailability in the Perfused Small Intes-tine of the Rat. Drugs Exp. Clin. Res. 2000, 26, 47–55. [Google Scholar] [PubMed]
  27. Almeida, L.; Silva, M.V.; Falcão, A.; Soares, E.; Costa, R.; Loureiro, A.I.; Fernandes-Lopes, C.; Rocha, J.-F.; Nunes, T.; Wright, L.; et al. Pharmacokinetic and safety profile of trans-resveratrol in a rising multiple-dose study in healthy volunteers. Mol. Nutr. Food Res. 2009, 53, S7–S15. [Google Scholar] [CrossRef]
  28. La Porte, C.; Voduc, N.; Zhang, G.; Seguin, I.; Tardiff, D.; Singhal, N.; Cameron, B. Steady-State Pharmacokinetics and Tolerability of Trans-Resveratrol 2000 mg Twice Daily with Food, Quercetin and Alcohol (Ethanol) in Healthy Human Subjects. Clin. Pharmacokinet. 2010, 49, 449–454. [Google Scholar] [CrossRef] [PubMed]
  29. Bruguerolle, B. Chronopharmacokinetics. Clin. Pharmacokinet. 1998, 35, 83–94. [Google Scholar] [CrossRef]
  30. Silva, M.V.; Loureiro, A.; Falcao, A.; Nunes, T.; Rocha, J.-F.; Fernandes-Lopes, C.; Soares, E.; Wright, L.; Almeida, L.; Soares-Da-Silva, P. Effect of food on the pharmacokinetic profile of trans-resveratrol. Int. J. Clin. Pharmacol. Ther. 2008, 46, 564–570. [Google Scholar] [CrossRef]
  31. Zhu, X.; Wu, C.; Qiu, S.; Yuan, X.; Li, L.; Zhu, X.; Wu, C.; Qiu, S.; Yuan, X.; Li, L. Effects of resveratrol on glucose control and insulin sensitivity in subjects with type 2 diabetes: Systematic review and meta-analysis. Nutr. Metab. 2017, 14, 60. [Google Scholar] [CrossRef]
  32. Calabrese, E.J.; Mattson, M.P.; Calabrese, V. Resveratrol commonly displays hormesis: Occurrence and biomedical significance. Hum. Exp. Toxicol. 2010, 29, 980–1015. [Google Scholar] [CrossRef]
  33. 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] [Green Version]
  34. Pannu, N.; Bhatnagar, A. Resveratrol: From enhanced biosynthesis and bioavailability to multitargeting chronic diseases. Biomed. Pharmacother. 2018, 109, 2237–2251. [Google Scholar] [CrossRef]
  35. Catalogna, G.; Moraca, F.; D’Antona, L.; Dattilo, V.; Perrotti, G.; Lupia, A.; Costa, G.; Ortuso, F.; Iuliano, R.; Trapasso, F.; et al. Review about the multi-target profile of resveratrol and its implication in the SGK1 inhibition. Eur. J. Med. Chem. 2019, 183, 111675. [Google Scholar] [CrossRef]
  36. Varoni, E.M.; Faro, A.F.L.; Sharifi-Rad, J.; Iriti, M. Anticancer Molecular Mechanisms of Resveratrol. Front. Nutr. 2016, 3, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Vestergaard, M.; Ingmer, H. Antibacterial and antifungal properties of resveratrol. Int. J. Antimicrob. Agents 2019, 53, 716–723. [Google Scholar] [CrossRef] [PubMed]
  38. Chen, L.; Deng, H.; Cui, H.; Fang, J.; Zuo, Z.; Deng, J.; Li, Y.; Wang, X.; Zhao, L. Inflammatory responses and inflammation-associated diseases in organs. Oncotarget 2017, 9, 7204–7218. [Google Scholar] [CrossRef] [Green Version]
  39. Dyck, G.J.B.; Raj, P.; Zieroth, S.; Dyck, J.R.B.; Ezekowitz, J.A. The Effects of Resveratrol in Patients with Cardiovascular Disease and Heart Failure: A Narrative Review. Int. J. Mol. Sci. 2019, 20, 904. [Google Scholar] [CrossRef] [Green Version]
  40. Asgary, S.; Karimi, R.; Momtaz, S.; Naseri, R.; Farzaei, M.H. Effect of resveratrol on metabolic syndrome components: A systematic review and meta-analysis. Rev. Endocr. Metab. Disord. 2019, 20, 173–186. [Google Scholar] [CrossRef] [PubMed]
  41. Springer, M.; Moco, S. Resveratrol and Its Human Metabolites—Effects on Metabolic Health and Obesity. Nutrients 2019, 11, 143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Tabrizi, R.; Tamtaji, O.R.; Lankarani, K.B.; Akbari, M.; Dadgostar, E.; Dabbaghmanesh, M.H.; Kolahdooz, F.; Shamshirian, A.; Momen-Heravi, M.; Asemi, Z. The effects of resveratrol intake on weight loss: A systematic review and meta-analysis of randomized controlled trials. Crit. Rev. Food Sci. Nutr. 2018, 60, 375–390. [Google Scholar] [CrossRef] [PubMed]
  43. Ortega, I.; Duleba, A.J. Ovarian actions of resveratrol. Ann. N. Y. Acad. Sci. 2015, 1348, 86–96. [Google Scholar] [CrossRef]
  44. Kolesarova, A.; Capcarová, M.; Maruniakova, N.; Lukac, N.; Ciereszko, R.E.; Sirotkin, A.V. Resveratrol inhibits reproductive toxicity induced by deoxynivalenol. J. Environ. Sci. Heal. Part A 2012, 47, 1329–1334. [Google Scholar] [CrossRef]
  45. Brasnyó, P.; Molnar, G.A.; Mohás, M.; Markó, L.; Laczy, B.; Cseh, J.; Mikolás, E.; Szijártó, I.A.; Mérei, Á.; 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] [PubMed] [Green Version]
  46. Johnson, A.A.; Riehle, M.A. Resveratrol Fails to Extend Life Span in the MosquitoAnopheles stephensi. Rejuvenation Res. 2015, 18, 473–478. [Google Scholar] [CrossRef] [PubMed]
  47. Baur, J.; Pearson, K.J.; Price, N.; Jamieson, H.A.; Lerin, C.; Kalra, A.; Prabhu, V.V.; Allard, J.S.; López-Lluch, G.; Lewis, K.; et al. Resveratrol improves health and survival of mice on a high-calorie diet. Nature 2006, 444, 337–342. [Google Scholar] [CrossRef]
  48. Christenson, J.; Whitby, S.J.; Mellor, D.; Thomas, J.; Mc Kune, A.; Roach, P.D.; Naumovski, N. The Effects of Resveratrol Supplementation in Overweight and Obese Humans: A Systematic Review of Randomized Trials. Metab. Syndr. Relat. Disord. 2016, 14, 323–333. [Google Scholar] [CrossRef] [PubMed]
  49. Fernández-Quintela, A.; Carpéné, C.; Fernández, M.; Aguirre, L.; Milton-Laskibar, I.; Contreras, J.; Portillo, M.P. Anti-obesity effects of resveratrol: Comparison between animal models and humans. J. Physiol. Biochem. 2016, 73, 417–429. [Google Scholar] [CrossRef]
  50. Li, S.; Chen, M.; Li, Y.; Tollefsbol, T.O. Prenatal epigenetics diets play protective roles against environmental pollution. Clin. Epigenetics 2019, 11, 82. [Google Scholar] [CrossRef]
  51. Bishayee, A.; Barnes, K.F.; Bhatia, D.; Darvesh, A.S.; Carroll, R.T. Resveratrol Suppresses Oxidative Stress and Inflammatory Response in Diethylnitrosamine-Initiated Rat Hepatocarcinogenesis. Cancer Prev. Res. 2010, 3, 753–763. [Google Scholar] [CrossRef] [Green Version]
  52. Riba, A.; Deres, L.; Sumegi, B.; Toth, K.; Szabados, E.; Halmosi, R. Cardioprotective Effect of Resveratrol in a Postinfarction Heart Failure Model. Oxidative Med. Cell. Longev. 2017, 2017, 6819281. [Google Scholar] [CrossRef]
  53. Gambini, J.; Ingles, M.; Olaso, G.; Lopez-Grueso, R.; Bonet-Costa, V.; Gimeno-Mallench, L.; Bargues, C.M.; Abdelaziz, K.M.; Gomez-Cabrera, M.C.; Vina, J.; et al. Properties of Resveratrol:In VitroandIn VivoStudies about Metabolism, Bioavailability, and Biological Effects in Animal Models and Humans. Oxidative Med. Cell. Longev. 2015, 2015, 837042. [Google Scholar] [CrossRef] [Green Version]
  54. Kataria, R.; Khatkar, A. Molecular docking, synthesis, kinetics study, structure–activity relationship and ADMET analysis of morin analogous as Helicobacter pylori urease inhibitors. BMC Chem. 2019, 13, 1–17. [Google Scholar] [CrossRef]
  55. Mirza, S.; Sharma, G.; Parshad, R.; Gupta, S.D.; Pandya, P.; Ralhan, R. Expression of DNA Methyltransferases in Breast Cancer Patients and to Analyze the Effect of Natural Compounds on DNA Methyltransferases and Associated Proteins. J. Breast Cancer 2013, 16, 23–31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Liu, K.; Zhou, R.; Wang, B.; Mi, M.-T. Effect of resveratrol on glucose control and insulin sensitivity: A meta-analysis of 11 randomized controlled trials. Am. J. Clin. Nutr. 2014, 99, 1510–1519. [Google Scholar] [CrossRef] [Green Version]
  57. Borra, M.T.; Smith, B.; Denu, J.M. Mechanism of Human SIRT1 Activation by Resveratrol. J. Biol. Chem. 2005, 280, 17187–17195. [Google Scholar] [CrossRef] [Green Version]
  58. Lagouge, M.; Argmann, C.; Gerhart-Hines, Z.; Meziane, H.; Lerin, C.; Daussin, F.; Messadeq, N.; Milne, J.; Lambert, P.; Elliott, P.; et al. Resveratrol Improves Mitochondrial Function and Protects against Metabolic Disease by Activating SIRT1 and PGC-1α. Cell 2006, 127, 1109–1122. [Google Scholar] [CrossRef] [PubMed]
  59. Vlavcheski, F.; Hartogh, D.J.D.; Giacca, A.; Tsiani, E. Amelioration of High-Insulin-Induced Skeletal Muscle Cell Insulin Resistance by Resveratrol Is Linked to Activation of AMPK and Restoration of GLUT4 Translocation. Nutrients 2020, 12, 914. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Penumathsa, S.V.; Thirunavukkarasu, M.; Zhan, L.; Maulik, G.; Menon, V.P.; Bagchi, D.; Maulik, N. Resveratrol enhances GLUT-4 translocation to the caveolar lipid raft fractions through AMPK/Akt/eNOS signalling pathway in diabetic myocardium. J. Cell. Mol. Med. 2008, 12, 2350–2361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Su, H.-C.; Hung, L.-M.; Chen, J.-K. Resveratrol, a red wine antioxidant, possesses an insulin-like effect in streptozotocin-induced diabetic rats. Am. J. Physiol. Metab. 2006, 290, E1339–E1346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Szkudelski, T. The insulin-suppressive effect of resveratrol—An in vitro and in vivo phenomenon. Life Sci. 2008, 82, 430–435. [Google Scholar] [CrossRef]
  63. Bertelli, A.A.; Giovannini, L.; Stradi, R.; Tillement, J.P. Plasma, urine and tissue levels of trans- and cis-resveratrol (3,4’,5-trihydroxystilbene) after short-term or prolonged administration of red wine to rats. Int. J. Tissue React. 1996, 18, 67–71. [Google Scholar] [PubMed]
  64. Bertelli, A.A.; Giovannini, L.; Stradi, R.; Urien, S.; Tillement, J.P.; Bertelli, A. Kinetics of Trans- and Cis-Resveratrol (3,4’,5-Trihydroxystilbene) after Red Wine Oral Administration in Rats. Int. J. Clin. Pharmacol. Res. 1996, 16, 77–81. [Google Scholar] [CrossRef]
  65. Soleas, G.J.; Angelini, M.; Grass, L.; Diamandis, E.P.; Goldberg, D.M. Absorption of trans-resveratrol in rats. Methods Enzymol. 2001, 335, 145–154. [Google Scholar] [CrossRef] [PubMed]
  66. Vitrac, X.; Desmoulière, A.; Brouillaud, B.; Krisa, S.; Deffieux, G.; Barthe, N.; Rosenbaum, J.; Mérillon, J.-M. Distribution of [14C]-trans-resveratrol, a cancer chemopreventive polyphenol, in mouse tissues after oral administration. Life Sci. 2003, 72, 2219–2233. [Google Scholar] [CrossRef]
  67. Yu, C.; Shin, Y.G.; Chow, A.; Li, Y.; Kosmeder, J.W.; Lee, Y.S.; Hirschelman, W.H.; Pezzuto, J.M.; Mehta, R.G.; van Breemen, R.B. Human, Rat, and Mouse Metabolism of Resveratrol. Pharm. Res. 2002, 19, 1907–1914. [Google Scholar] [CrossRef] [PubMed]
  68. Wenzel, E.; Soldo, T.; Erbersdobler, H.; Somoza, V. Bioactivity and metabolism oftrans-resveratrol orally administered to Wistar rats. Mol. Nutr. Food Res. 2005, 49, 482–494. [Google Scholar] [CrossRef]
  69. Azorín-Ortuño, M.; Yáñez-Gascón, M.J.; Vallejo, F.; Pallarés, F.J.; Larrosa, M.; Lucas, R.; Morales, J.C.; Tomas-Barberan, F.; Conesa, M.T.G.; Espín, J.C. Metabolites and tissue distribution of resveratrol in the pig. Mol. Nutr. Food Res. 2011, 55, 1154–1168. [Google Scholar] [CrossRef] [Green Version]
  70. Brill, S.S.; Furimsky, A.M.; Ho, M.N.; Furniss, M.J.; Li, Y.; Green, A.G.; Green, C.E.; Iyer, L.V.; Bradford, W.W.; Kapetanovic, I.M. Glucuronidation of trans-resveratrol by human liver and intestinal microsomes and UGT isoforms. J. Pharm. Pharmacol. 2006, 58, 469–479. [Google Scholar] [CrossRef]
  71. Planas, J.M.; Alfaras, I.; Colom, H.; Juan, M.E. The bioavailability and distribution of trans-resveratrol are constrained by ABC transporters. Arch. Biochem. Biophys. 2012, 527, 67–73. [Google Scholar] [CrossRef]
  72. Van De Wetering, K.; Burkon, A.; Feddema, W.; Bot, A.; De Jonge, H.; Somoza, V.; Borst, P. Intestinal Breast Cancer Resistance Protein (BCRP)/Bcrp1 and Multidrug Resistance Protein 3 (MRP3)/Mrp3 Are Involved in the Pharmacokinetics of Resveratrol. Mol. Pharmacol. 2008, 75, 876–885. [Google Scholar] [CrossRef] [Green Version]
  73. Kaldas, M.I.; Walle, U.K.; Walle, T. Resveratrol transport and metabolism by human intestinal Caco-2 cells. J. Pharm. Pharmacol. 2003, 55, 307–312. [Google Scholar] [CrossRef] [PubMed]
  74. Bode, L.M.; Bunzel, D.; Huch, M.; Cho, G.-S.; Ruhland, D.; Bunzel, M.; Bub, A.; Franz, C.M.A.P.; Kulling, E.S. In vivo and in vitro metabolism of trans-resveratrol by human gut microbiota. Am. J. Clin. Nutr. 2013, 97, 295–309. [Google Scholar] [CrossRef]
  75. Jarosova, V.; Vesely, O.; Marsik, P.; Jaimes, J.D.; Smejkal, K.; Kloucek, P.; Havlik, J. Metabolism of Stilbenoids by Human Faecal Microbiota. Molecules 2019, 24, 1155. [Google Scholar] [CrossRef] [Green Version]
  76. Jaimes, J.D.; Jarosova, V.; Vesely, O.; Mekadim, C.; Mrazek, J.; Marsik, P.; Killer, J.; Smejkal, K.; Kloucek, P.; Havlik, J. Effect of Selected Stilbenoids on Human Fecal Microbiota. Molecules 2019, 24, 744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Urpi-Sarda, M.; Zamora-Ros, R.; Lamuela-Raventos, R.M.; Cherubini, A.; Jauregui, O.; de la Torre, R.; Covas, M.I.; Estruch, R.; Jaeger, W.; Andres-Lacueva, C. HPLC–Tandem Mass Spectrometric Method to Characterize Resveratrol Metabolism in Humans. Clin. Chem. 2007, 53, 292–299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Delmas, D.; Aires, V.; Limagne, E.; Dutartre, P.; Mazué, F.; Ghiringhelli, F.; Latruffe, N. Transport, stability, and biological activity of resveratrol. Ann. N. Y. Acad. Sci. 2011, 1215, 48–59. [Google Scholar] [CrossRef]
  79. Fernández-Castillejo, S.; Macià, A.; Motilva, M.J.; Catalán, Ú.; Solà, R. Endothelial Cells Deconjugate Resveratrol Metabolites to Free Resveratrol: A Possible Role in Tissue Factor Modulation. Mol. Nutr. Food Res. 2018, 63, e1800715. [Google Scholar] [CrossRef]
  80. Vinson, J.A. Intracellular Polyphenols: How Little We Know. J. Agric. Food Chem. 2019, 67, 3865–3870. [Google Scholar] [CrossRef] [PubMed]
  81. 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] [PubMed]
  82. Koziolek, M.; Alcaro, S.; Augustijns, P.; Basit, A.W.; Grimm, M.; Hens, B.; Hoad, C.L.; Jedamzik, P.; Madla, C.M.; Maliepaard, M.; et al. The mechanisms of pharmacokinetic food-drug interactions—A perspective from the UNGAP group. Eur. J. Pharm. Sci. 2019, 134, 31–59. [Google Scholar] [CrossRef] [PubMed]
  83. Kamiloglu, S.; Tomas, M.; Ozdal, T.; Capanoglu, E. Effect of food matrix on the content and bioavailability of flavonoids. Trends Food Sci. Technol. 2020, in press. [Google Scholar] [CrossRef]
  84. Mhaske, D.B.; Sreedharan, S.; Mahadik, K.R. Role of Piperine as an Effective Bioenhancer in Drug Absorption. Phar-Maceutika Anal. Acta 2018, 9, 591. [Google Scholar]
  85. Wightman, E.L.; Reay, J.; Haskell, C.F.; Williamson, G.; Dew, T.; Kennedy, D. Effects of resveratrol alone or in combination with piperine on cerebral blood flow parameters and cognitive performance in human subjects: A randomised, double-blind, placebo-controlled, cross-over investigation. Br. J. Nutr. 2014, 112, 203–213. [Google Scholar] [CrossRef]
  86. Bailey, H.H.; Johnson, J.J.; Lozar, T.; Scarlett, C.O.; Wollmer, B.W.; Kim, K.; Havinghurst, T.; Ahmad, N. A randomized, double-blind, dose-ranging, pilot trial of piperine with resveratrol on the effects on serum levels of resveratrol. Eur. J. Cancer Prev. 2020, 30, 285–290. [Google Scholar] [CrossRef]
  87. Johnson, J.J.; Nihal, M.; Siddiqui, I.A.; Scarlett, C.O.; Bailey, H.H.; Mukhtar, H.; Ahmad, N. Enhancing the bioavailability of resveratrol by combining it with piperine. Mol. Nutr. Food Res. 2011, 55, 1169–1176. [Google Scholar] [CrossRef] [Green Version]
  88. 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]
  89. Junsaeng, D.; Anukunwithaya, T.; Songvut, P.; Sritularak, B.; Likhitwitayawuid, K.; Khemawoot, P. Comparative pharmacokinetics of oxyresveratrol alone and in combination with piperine as a bioenhancer in rats. BMC Complement. Altern. Med. 2019, 19, 235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Lund, K.C.; Pantuso, T. Combination Effects of Quercetin, Resveratrol and Curcumin on In Vitro Intestinal Absorption. J. Restor. Med. 2014, 3, 112–120. [Google Scholar] [CrossRef] [Green Version]
  91. Lee, J.-A.; Ha, S.K.; Cho, E.; Choi, I. Resveratrol as a Bioenhancer to Improve Anti-Inflammatory Activities of Apigenin. Nutrients 2015, 7, 9650–9661. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Detampel, P.; Beck, M.; Krähenbühl, S.; Huwyler, J. Drug interaction potential of resveratrol. Drug Metab. Rev. 2012, 44, 253–265. [Google Scholar] [CrossRef]
  93. Paine, M.F.; Hart, H.L.; Ludington, S.S.; Haining, R.L.; Rettie, A.E.; Zeldin, D. The human intestinal cytochrome P450 “pie”. Drug Metab. Dispos. 2006, 34, 880–886. [Google Scholar] [CrossRef]
  94. Syed, S.B.; Arya, H.; Fu, I.-H.; Yeh, T.-K.; Periyasamy, L.; Hsieh, H.-P.; Coumar, M.S.; Syed, S.B.; Arya, H.; Fu, I.-H.; et al. Targeting P-glycoprotein: Investigation of piperine analogs for overcoming drug resistance in cancer. Sci. Rep. 2017, 7, 7972. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Thiebaut, F.; Tsuruo, T.; Hamada, H.; Gottesman, M.M.; Pastan, I.; Willingham, M.C. Cellular localization of the multidrug-resistance gene product P-glycoprotein in normal human tissues. Proc. Natl. Acad. Sci. USA 1987, 84, 7735–7738. [Google Scholar] [CrossRef] [Green Version]
  96. Quan, F.; Pan, C.; Ma, Q.; Zhang, S.; Yan, L. Reversal effect of resveratrol on multidrug resistance in KBv200 cell line. Biomed. Pharmacother. 2008, 62, 622–629. [Google Scholar] [CrossRef] [PubMed]
  97. El-Readi, M.Z.; Eid, S.; Abdelghany, A.A.; Al-Amoudi, H.S.; Efferth, T.; Wink, M. Resveratrol mediated cancer cell apoptosis, and modulation of multidrug resistance proteins and metabolic enzymes. Phytomedicine 2018, 55, 269–281. [Google Scholar] [CrossRef] [PubMed]
  98. Huang, F.; Wu, X.-N.; Chen, J.; Wang, W.-X.; Lu, Z. Resveratrol reverses multidrug resistance in human breast cancer doxorubicin-resistant cells. Exp. Ther. Med. 2014, 7, 1611–1616. [Google Scholar] [CrossRef] [PubMed]
  99. Mieszala, K.; Rudewicz, M.; Gomułkiewicz, A.; Ratajczak-Wielgomas, K.; Grzegrzółka, J.; Dziegiel, P.; Borska, S. Expression of genes and proteins of multidrug resistance in gastric cancer cells treated with resveratrol. Oncol. Lett. 2018, 15, 5825–5832. [Google Scholar] [CrossRef]
  100. Devi, P.; Sharma, P.; Rathore, C.; Negi, P. Novel Drug Delivery Systems of Resveratrol to Bioavailability and Therapeutic Effects. IntechOpen 2019. [Google Scholar] [CrossRef] [Green Version]
  101. Balata, G.F.; Eassa, E.; Shamrool, H.; Zidan, S.; Abourehab, M. Self-emulsifying drug delivery systems as a tool to improve solubility and bioavailability of resveratrol. Drug Des. Dev. Ther. 2016, 10, 117–128. [Google Scholar] [CrossRef] [Green Version]
  102. Nam, J.-B.; Ryu, J.-H.; Kim, J.-W.; Chang, I.-S.; Suh, K.-D. Stabilization of resveratrol immobilized in monodisperse cyano-functionalized porous polymeric microspheres. Polymer 2005, 46, 8956–8963. [Google Scholar] [CrossRef]
  103. Shi, G.; Rao, L.; Yu, H.; Xiang, H.; Yang, H.; Ji, R. Stabilization and encapsulation of photosensitive resveratrol within yeast cell. Int. J. Pharm. 2008, 349, 83–93. [Google Scholar] [CrossRef]
  104. Aloisio, C.; Bueno, M.S.; Ponte, M.P.; Paredes, A.; Palma, S.D.; Longhi, M. Development of solid self-emulsifying drug delivery systems (SEDDS) to improve the solubility of resveratrol. Ther. Deliv. 2019, 10, 626–641. [Google Scholar] [CrossRef]
  105. Seljak, K.B.; Berginc, K.; Trontelj, J.; Zvonar, A.; Kristl, A.; Gašperlin, M. A Self-Microemulsifying Drug Delivery System to Overcome Intestinal Resveratrol Toxicity and Presystemic Metabolism. J. Pharm. Sci. 2014, 103, 3491–3500. [Google Scholar] [CrossRef]
  106. Jaisamut, P.; Wanna, S.; Limsuwan, S.; Chusri, S.; Wiwattanawongsa, K.; Wiwattanapatapee, R. Enhanced Oral Bioavailability and Improved Biological Activities of a Quercetin/Resveratrol Combination Using a Liquid Self-Microemulsifying Drug Delivery System. Planta Medica 2020, 87, 336–346. [Google Scholar] [CrossRef]
  107. Luo, X.; Wang, D.; Wang, M.; Deng, S.; Huang, Y.; Xia, Z. Development of phospholipid complex loaded self-microemulsifying drug delivery system to improve the oral bioavailability of resveratrol. Nanomedicine 2021, 16, 721–739. [Google Scholar] [CrossRef]
  108. Ansari, K.A.; Vavia, P.R.; Trotta, F.; Cavalli, R. Cyclodextrin-Based Nanosponges for Delivery of Resveratrol: In Vitro Characterisation, Stability, Cytotoxicity and Permeation Study. AAPS PharmSciTech 2011, 12, 279–286. [Google Scholar] [CrossRef] [Green Version]
  109. Sessa, M.; Balestrieri, M.L.; Ferrari, G.; Servillo, L.; Castaldo, D.; D’Onofrio, N.; Donsì, F.; Tsao, R. Bioavailability of encapsulated resveratrol into nanoemulsion-based delivery systems. Food Chem. 2014, 147, 42–50. [Google Scholar] [CrossRef] [PubMed]
  110. Donsì, F.; Sessa, M.; Mediouni, H.; Mgaidi, A.; Ferrari, G. Encapsulation of bioactive compounds in nanoemulsion- based delivery systems. Procedia Food Sci. 2011, 1, 1666–1671. [Google Scholar] [CrossRef] [Green Version]
  111. Singh, G.; Pai, R.S. Trans-resveratrol self-nano-emulsifying drug delivery system (SNEDDS) with enhanced bioavailability potential: Optimization, pharmacokinetics andin situsingle pass intestinal perfusion (SPIP) studies. Drug Deliv. 2013, 22, 522–530. [Google Scholar] [CrossRef] [Green Version]
  112. Yen, C.-C.; Chang, C.-W.; Hsu, M.-C.; Wu, Y.-T. Self-Nanoemulsifying Drug Delivery System for Resveratrol: Enhanced Oral Bioavailability and Reduced Physical Fatigue in Rats. Int. J. Mol. Sci. 2017, 18, 1853. [Google Scholar] [CrossRef] [Green Version]
  113. Dai, L.; Zhu, W.; Liu, R.; Si, C. Lignin-Containing Self-Nanoemulsifying Drug Delivery System for Enhance Stability and Oral Absorption oftrans-Resveratrol. Part. Part. Syst. Charact. 2018, 35, 1700447. [Google Scholar] [CrossRef]
  114. Md, S.; Alhakamy, N.A.; Aldawsari, H.M.; Ahmad, J.; Alharbi, W.S.; Asfour, H.Z. Resveratrol loaded self-nanoemulsifying drug delivery system (SNEDDS) for pancreatic cancer: Formulation design, optimization and in vitro evaluation. J. Drug Deliv. Sci. Technol. 2021, 64, 102555. [Google Scholar] [CrossRef]
  115. Hu, C.; Wang, Q.; Ma, C.; Xia, Q. Non-aqueous self-double-emulsifying drug delivery system: A new approach to enhance resveratrol solubility for effective transdermal delivery. Colloids Surf. A Physicochem. Eng. Asp. 2016, 489, 360–369. [Google Scholar] [CrossRef]
  116. Ethemoglu, M.; Seker, F.; Akkaya, H.; Kilic, E.; Aslan, I.; Erdogan, C.S.; Yilmaz, B. Anticonvulsant activity of resveratrol-loaded liposomes in vivo. Neuroscience 2017, 357, 12–19. [Google Scholar] [CrossRef]
  117. Basavaraj, S.; Betageri, G.V. Improved oral delivery of resveratrol using proliposomal formulation: Investigation of various factors contributing to prolonged absorption of unmetabolized resveratrol. Expert Opin. Drug Deliv. 2014, 11, 493–503. [Google Scholar] [CrossRef] [PubMed]
  118. Teskač, K.; Kristl, J. The evidence for solid lipid nanoparticles mediated cell uptake of resveratrol. Int. J. Pharm. 2010, 390, 61–69. [Google Scholar] [CrossRef]
  119. Zhang, Q.H.; Xiong, Q.P.; Shi, Y.Y.; Zhang, D.Y. Study on Preparation and Characterization of Resveratrol Solid Li-pid Nanoparticles and Its Anticancer Effects in Vitro. Zhong Yao Cai Zhongyaocai J. Chin. Med. Mater. 2010, 33, 1929–1932. [Google Scholar]
  120. Oganesyan, E.A.; Miroschichenko, I.I.; Vikhrieva, N.S.; Lyashenko, A.A.; Leshkov, S.Y. Use of Nanoparticles to In-crease the Systemic Bioavailability of trans-Resveratrol. Pharm. Chem. J. 2010, 44, 25–27. [Google Scholar] [CrossRef]
  121. Singh, G.; Pai, R.S. In-vitro/in-vivo characterization of trans-resveratrol-loaded nanoparticulate drug delivery system for oral administration. J. Pharm. Pharmacol. 2014, 66, 1062–1076. [Google Scholar] [CrossRef]
  122. Frozza, R.L.; Bernardi, A.; Paese, K.; Hoppe, J.B.; Da Silva, T.; Battastini, A.M.O.; Pohlmann, A.; Guterres, S.; Salbego, C. Characterization of trans-Resveratrol-Loaded Lipid-Core Nanocapsules and Tissue Distribution Studies in Rats. J. Biomed. Nanotechnol. 2010, 6, 694–703. [Google Scholar] [CrossRef]
  123. Lin, Y.F.; Lee, Y.H.; Hsu, Y.H.; Chen, Y.J.; Lin, Y.F.; Cheng, F.-Y.; Chiu, H.W. Resveratrol-Loaded Nanoparticles Conjugated with Kidney Injury Molecule-1 as a Drug Delivery System for Potential Use in Chronic Kidney Disease. Nanomedicine 2017, 12, 2741–2756. [Google Scholar] [CrossRef]
  124. Singh, A.; Ahmad, I.; Ahmad, S.; Iqbal, Z.; Ahmad, F. A novel monolithic controlled delivery system of resveratrol for enhanced hepatoprotection: Nanoformulation development, pharmacokinetics and pharmacodynamics. Drug Dev. Ind. Pharm. 2016, 42, 1524–1536. [Google Scholar] [CrossRef]
  125. Li, C.; Wang, X.; Li, R.; Yang, X.; Zhong, Z.; Dai, Y.; Fan, Q.; Lin, Y.; Zhang, R.; Liang, T.; et al. Resveratrol-loaded PLGA nanoparticles functionalized with red blood cell membranes as a biomimetic delivery system for prolonged circulation time. J. Drug Deliv. Sci. Technol. 2019, 54, 101369. [Google Scholar] [CrossRef]
  126. 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]
  127. Reis, S.; Neves, A.R.; Lúcio, M.; Martins, S.; Lima, J. Novel resveratrol nanodelivery systems based on lipid nanoparticles to enhance its oral bioavailability. Int. J. Nanomed. 2013, 8, 177–187. [Google Scholar] [CrossRef] [Green Version]
  128. Hao, J.; Gao, Y.; Zhao, J.; Zhang, J.; Li, Q.; Zhao, Z.; Liu, J. Preparation and Optimization of Resveratrol Nanosuspensions by Antisolvent Precipitation Using Box-Behnken Design. AAPS PharmSciTech 2014, 16, 118–128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Rostami, M.; Ghorbani, M.; Mohammadi, M.A.; Delavar, M.; Tabibiazar, M.; Ramezani, S. Development of resveratrol loaded chitosan-gellan nanofiber as a novel gastrointestinal delivery system. Int. J. Biol. Macromol. 2019, 135, 698–705. [Google Scholar] [CrossRef]
  130. Liu, C.; Tong, P.; Yang, R.; You, Y.; Liu, H.; Zhang, T. Solidified phospholipid-TPGS as an effective oral delivery system for improving the bioavailability of resveratrol. J. Drug Deliv. Sci. Technol. 2019, 52, 769–777. [Google Scholar] [CrossRef]
  131. Wang, P.-P.; Luo, Z.-G.; Tamer, T.M. Spiral–Dextrin Complex Crystals: Efficient Approach for Colon-Targeted Resveratrol Delivery. J. Agric. Food Chem. 2020, 69, 474–482. [Google Scholar] [CrossRef]
  132. Paczkowska-Walendowska, M.; Dvořák, J.; Rosiak, N.; Tykarska, E.; Szymańska, E.; Winnicka, K.; Ruchała, M.; Cielecka-Piontek, J. Buccal Resveratrol Delivery System as a Potential New Concept for the Periodontitis Treatment. Pharmaceutics 2021, 13, 417. [Google Scholar] [CrossRef]
  133. Chen, Z.; Farag, M.A.; Zhong, Z.; Zhang, C.; Yang, Y.; Wang, S.; Wang, Y. Multifaceted role of phyto-derived polyphenols in nanodrug delivery systems. Adv. Drug Deliv. Rev. 2021, 113870. [Google Scholar] [CrossRef] [PubMed]
  134. 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]
  135. Briskey, D.; Rao, A. Trans-Resveratrol Oral Bioavailability in Humans Using LipiSperse™ Dispersion Technology. Pharmaceutics 2020, 12, 1190. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Chemical structure of trans-resveratrol.
Figure 1. Chemical structure of trans-resveratrol.
Nutrients 13 03095 g001
Figure 2. When resveratrol (RES) reaches the colon, approximately 75% can enter the colonocytes and the remainder can be metabolized by the microbiota into metabolites such as dihydroresveratrol (DiRes) or lunularin (Lun). These may enter the cells or be excreted with the remaining resveratrol in the feces. Colonocytes can convert RES into metabolites via sulphotransferase (SULT) or UDP-glucuronosyltransferase (UGT). These are pumped mainly apically to the lumen via BRCP (breast cancer resistance protein) and MRP2 (multidrug resistance protein) and partly into the circulation via MRP3. When RES or metabolites pass through the cells, they are absorbed by low-density lipoprotein (LDL) or albumin and distributed throughout the body. When they reach the cells, they can be released via LDL-receptor (LDLR) and albumin binding domain (ABP) and subsequently absorbed by the cells. Finally, the cells can deconjugate them.
Figure 2. When resveratrol (RES) reaches the colon, approximately 75% can enter the colonocytes and the remainder can be metabolized by the microbiota into metabolites such as dihydroresveratrol (DiRes) or lunularin (Lun). These may enter the cells or be excreted with the remaining resveratrol in the feces. Colonocytes can convert RES into metabolites via sulphotransferase (SULT) or UDP-glucuronosyltransferase (UGT). These are pumped mainly apically to the lumen via BRCP (breast cancer resistance protein) and MRP2 (multidrug resistance protein) and partly into the circulation via MRP3. When RES or metabolites pass through the cells, they are absorbed by low-density lipoprotein (LDL) or albumin and distributed throughout the body. When they reach the cells, they can be released via LDL-receptor (LDLR) and albumin binding domain (ABP) and subsequently absorbed by the cells. Finally, the cells can deconjugate them.
Nutrients 13 03095 g002
Figure 3. Increasing intestinal permeability with (A) RES alone, (B) quercetin, and (C) piperine. As shown in row, RES (A) diffuses into cells without active assistance, is metabolized by SULT and UGT enzymes to metabolites (ResMet), and only a small volume reaches the basolateral side. Quercetin (B), when added to RES, inhibits ABC transporters (specifically PG, BRCP, and MRP2) and SULT. This combination leads to a decrease in sulphate metabolites and an increase in RES on the basolateral side. If piperine (C) is added, there are not enough studies against MRP, but like quercetin, it is able to inhibit ABC transporters, SULT, and even UGT. This leads to a decrease in sulphate and glucuronide metabolites and an increase in RES.
Figure 3. Increasing intestinal permeability with (A) RES alone, (B) quercetin, and (C) piperine. As shown in row, RES (A) diffuses into cells without active assistance, is metabolized by SULT and UGT enzymes to metabolites (ResMet), and only a small volume reaches the basolateral side. Quercetin (B), when added to RES, inhibits ABC transporters (specifically PG, BRCP, and MRP2) and SULT. This combination leads to a decrease in sulphate metabolites and an increase in RES on the basolateral side. If piperine (C) is added, there are not enough studies against MRP, but like quercetin, it is able to inhibit ABC transporters, SULT, and even UGT. This leads to a decrease in sulphate and glucuronide metabolites and an increase in RES.
Nutrients 13 03095 g003
Table 1. Summary of the in vitro, animal, and interventional studies involving stilbenoids and bioenhancers.
Table 1. Summary of the in vitro, animal, and interventional studies involving stilbenoids and bioenhancers.
CompoundDosageEffectTypeAnimal/CellResultsReference
Resveratrol5 g per personPeak serum concentration 539 ng/mL RES
Metabolites 3-8× higher compared to RES
Clinical studies Low bioavailability[20]
Resveratrol + piperine250 mg RES vs. 250 mg RES + 20 mg PIPNo vs. Increase cerebral blood flowNo altering bioavailability[85]
Resveratrol + piperine2.5 g RES + 0; 5 or 25 mg PIPNo relationship between dose and pharmacokinetic valuesDosage does not alter bioavailability[86]
Resveratrol + quercetin + alcohol2 g 2×/person
+ 500 mg Q
+ 5 mL alcohol
Neither quercetin nor alcohol alter RES.
Fat meal alter RES concentration
Fat meal decreases bioavailability[28]
Resveratrol + piperine100 mg/kg RES vs. 100 mg/kg RES + 10 mg/kg PIP (oral)After comparison maximum serum concentration increased to 1544% Animal studies C57BI/6 miceBeneficial[87]
Resveratrol + piperine30 mg/kgRES with piperine loaded mixed micelles demonstrated 5.7-fold compared to RESWistar albinoBeneficial[88]
Oxyresveratrol + piperine100 mg/kg OXY vs. 100 mg/kg OXY + 10 mg/kg PIP (oral)Piperine mix increased bioavailability of oxyresveratrol 2-foldWistar ratBeneficial[89]
Resveratrol + curcumin + quercetin + piperine50 µM (RES, C, Q)200 nM (PIP)RES received the greatest enhancement in permeability when combined with other agents: quercetin (310%), curcumin (300%), quercetin and curcumin (323%, 350% with piperine). Curcumin also demonstrated increased permeability when combined with quercetin alone (147%) and both quercetin and resveratrol (188%), addition of piperine resulted in a 229% increase in permeability. In vitro/ex vivo studies Caco-2Beneficial[90]
RES: resveratrol, PIP: piperine, OXY: oxyresveratrol, Q: quercetin, C: curcumin.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Vesely, O.; Baldovska, S.; Kolesarova, A. Enhancing Bioavailability of Nutraceutically Used Resveratrol and Other Stilbenoids. Nutrients 2021, 13, 3095. https://doi.org/10.3390/nu13093095

AMA Style

Vesely O, Baldovska S, Kolesarova A. Enhancing Bioavailability of Nutraceutically Used Resveratrol and Other Stilbenoids. Nutrients. 2021; 13(9):3095. https://doi.org/10.3390/nu13093095

Chicago/Turabian Style

Vesely, Ondrej, Simona Baldovska, and Adriana Kolesarova. 2021. "Enhancing Bioavailability of Nutraceutically Used Resveratrol and Other Stilbenoids" Nutrients 13, no. 9: 3095. https://doi.org/10.3390/nu13093095

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