Open access peer-reviewed chapter - ONLINE FIRST

Resveratrol Synthesis, Metabolism, and Delivery: A Mechanistic Treatise

Written By

Fakhar Islam, Umber Shehzadi, Farhan Saeed, Rabia Shabir Ahmad, Muhammad Umair Arshad, Muhammad Sadiq Naseer, Fatima Tariq, Rehman Ali, Sadaf Khurshid, Ghulam Hussain, Aftab Ahmad, Muhammad Afzaal, Rabia Akram, Osman Tuncay Agar, Ali Imran and Hafiz A.R. Suleria

Submitted: 24 April 2023 Reviewed: 10 April 2024 Published: 27 May 2024

DOI: 10.5772/intechopen.114982

Resveratrol - Recent Advances, Application, and Therapeutic Potential IntechOpen
Resveratrol - Recent Advances, Application, and Therapeutic Poten... Edited by Ali Imran

From the Edited Volume

Resveratrol - Recent Advances, Application, and Therapeutic Potential [Working Title]

Dr. Ali Imran and Dr. Hafiz Ansar Suleria

Chapter metrics overview

34 Chapter Downloads

View Full Metrics

Abstract

Resveratrol, a bioactive phytochemical classified as a phytoalexin present in plant sources, is recognized for its distinct characteristics such as anticancer, chemoprotective, chemosensitizer, neuroprotective, anti-inflammatory, and antioxidant properties. Resveratrol is a polyphenol that increases the susceptibility of cancer-resistant cells to chemotherapy. Resveratrol also aids in weight loss by decreasing lipogenesis, the prevention of neurological illnesses, and other topical uses such as the treatment of skin hyperpigmentation. During the past 10 years, resveratrol, a naturally occurring stilbene found in various foods and drinks, has drawn increased interest due to its many health benefits, including its chemo-preventive and anticancer actions. Several naturally occurring resveratrol derivatives can be found in food and share a similar structural makeup with resveratrol. To boost the effectiveness and activity of particular resveratrol features, several resveratrol analogues have also been created by the addition of designated functional groups. Such resveratrol derivatives might provide beneficial cancer therapeutics and cancer chemo-preventive drugs for cancer prevention and therapy. However, the quest for the identification of new analogues with high yield must be explored to extend resveratrol effectiveness. This chapter provides an overview of the most significant resveratrol derivatives used to treat cardiovascular diseases and the methods of their synthesis.

Keywords

  • resveratrol
  • therapeutic potential
  • antioxidants
  • phytochemicals
  • metabolism

1. Introduction

As a consequence of breakthroughs in nutritional research, academics have shown that nutrition plays a vital role in a number of disorders. Many investigations into the molecular effects of phytoconstituents have demonstrated that they are both safe and effective for long-term treatment. Recently, it has been found that several phytochemicals have strong anticancer and anti-arthritic properties. They consist of tea polyphenols, rosmarinic acid, curcumin, and resveratrol [1]. Polyphenol resveratrol may be found in a wide variety of plants [2]. It has cis and trans configurations, which can occasionally change toward one-up difference because a C∙C double bond is present. White squash was where resveratrol was first discovered in the 1940s; since then, it has also been discovered in grapes, peanuts, and Polygonum cuspidatum, among other plants [3].

Because of its abundance, and significant health advantages following ingestion, merlot, a biologically active polyphenolic stilbenoid, has gained interest in the management of medical conditions [4]. Research has been done on this molecule’s antioxidant, anti-inflammatory, and anti-carcinogenic characteristics, and most relate to lowering oxidative stress. Moreover, it has been shown to mediate autophagy, govern critical homeostatic processes in the body, and provide a range of systemic protective advantages [5].

Conventional β-cell cancer treatments including chemotherapy, surgery, and radiation have a number of drawbacks, including the ability to destroy healthy tissue and toxicity and long-term repercussions. Production of bioactives like resveratrol has been suggested as a feasible method of therapy to address these problems [6]. The use of nanoformulations in cancer treatment has been expanded upon to provide targeted distribution and improve valuable benefits. As compared to the way these drugs are typically administered, the use of nanoformulations of biotherapeutics has been shown to boost therapeutic value by increasing absorption and, ultimately, permeability. One such significant advance is the application of immunology [7]. Chimeric antigen receptor (CAR)-T β-cell lymphomas that are malignant have been treated using immunotherapy. Second-generation anti-CD19 medicines have gained prompt regulatory clearance and have been the subject of in-depth study in the treatment of relapse patients. Nevertheless, a few delivery and toxicity-related problems need to be overcome before they may be used more generally [8]. To solve the shortcomings of traditional drug delivery methods, this research focuses on the synthesis of resveratrol and describes the evidence for the related molecular mechanisms underlying these effects. This chapter also discusses the positive advantages of resveratrol, as well as the many hurdles connected with resveratrol’s clinical development and future prospects for therapeutic uses of this agent.

Advertisement

2. Synthesis and derivatives of resveratrol

With researchers’ discovery of organic merlot, several attempts have been undertaken to chemically and biologically develop resveratrol.

2.1 Heck reaction

The C∙C coupling of an engaged olefin with an aryl or vinyl halide is known as the Heck reaction, which is accelerated by palladium in the presence of a base. There are many acceptors and donors that are acceptable for Heck interactions, owing to subsequent advancements in catalytic and heterogeneous catalysts. Resveratrol and its substitutes can only be produced by the Forget interaction [9].

Pd catalysts from various sources must be immobilized onto heterogeneous supports to create pterostilbene. In addition to studying the structure of resveratrol derivatives, important polyphenolic compounds may be synthesized via retrosynthesis methods [10]. Palladium nanoparticles supported on synthetic clay may be able to successfully drive the Heck-Mizoroki C∙C cross-coupling reaction, an essential step. This reaction’s catalyst is strong, stable, and controlled. A tiny number of solvents is required during the purifying process, and the catalyst may be recovered and reused numerous times. Via a decarbonylative Heck process, the phytoalexin resveratrol may be produced. By combining 3,5-dihydroxybenzoic acid and 4-acetoxystyrene with palladium acetate and N,N-bis-(2,6diisopropylphenyl) dihydroimidazolium chloride, resveratrol derivatives were produced during this process [11]. Scientists have successfully synthesized a variety of resveratrol mimics. In experiments on human HL-60 cells, the four-acetoxy derivatives of resveratrol showed enhanced activity (ED50). A study team developed a brand-new, 70% yielding, effective method for producing resveratrol. Researchers improved the synthesis procedures, resulting in a 22–71% increase in overall resveratrol yield [12].

Scientists could synthesize resveratrol in a method that was rapid, simple, and incredibly chemo-, regio-, and stereoselective by dividing the tannin structure into three parts. Some of these derivatives were successful at protecting thymocytes from radioactive material apoptosis, studying of the radioprotective capabilities of resveratrol derivatives. Researchers looked into the manufacturing when utilizing the same procedure but changing the ring structure to a phenol ring [13]. The components demonstrated strong anti-human breast cancer cell activity. Researchers used a number of in vitro and cell-based targets to synthesize resveratrol via the Heck reaction to compare its activity to that of sulphate metabolites. Metabolites of sulphate are frequently less powerful than merlot [14].

2.2 Perkin reaction

Spontaneously, the Perkin synthesis transforms aromatic aldehydes and anhydrides into alpha- and beta-unsaturated carboxylic acids. It requires sodium acetate, an acid, and a base. Phases of the process include condensation, decarboxylation, deprotection, and protection. A product of the regioselective reaction is developed. Späth and Kromp first produced resveratrol using the Perkin reaction, which calls for 1,3-dimethoxy benzaldehyde and sodium salt of p-anisyl acetic acid as the reactants in the presence of acetic anhydride. Takaoka started synthesizing resveratrol after finding it in the root plant Veratrum grandfluorum. The final resveratrol derivative product, which has a structure precisely like a natural substance, was created by decarboxylating quinolone-Cu salt. Many advancements have been noticed [15].

2.3 Wittig reaction

The Wittig reactions use triphenylphosphine, a base, and a byproduct termed triphenylphosphine-oxide to change the main primary alkyl halides, aldehydes/ketones, and an olefin output to make an olefin product. This technique typically resulted in a C∙C double bond. By using the Wittig reaction, we produced trans-resveratrol and investigated how grapes produced resveratrol berries at various phases of growth. Resveratrol was either absent from or present in tiny amounts in the flesh of the fruit; it was generated in the skin cells. The amount of resveratrol in grape skin and the phases of berry development were clearly inversely correlated. If the phosphoric geographical region allies are benzyl alcohols, resveratrol and its analogues are synthesized in a single-pot Wittig-type olefination process. By using the Wittig reaction, we were able to produce trans-resveratrol [16].

Even though the Baeyer process is often employed to create an ethylenic bridge, it produces triphenylphosphine oxide as a byproduct and has poor trans invention yields and/or low E/Z selectivity. As a result, chromatography purifying is necessary for the procedure. Thus, it is crucial to look for rapid processing techniques or efficient catalysts for Wittig interactions [17].

2.4 Other methods

In addition to the typical resveratrol reactions, researchers have found additional ways to make resveratrol and its derivatives. Studies have compiled recent advancements in the Julia-Kocienski response, and the reaction has been recognized as a vital step in the synthesis of resveratrol [18]. The reactants were 3,5-dimethoxybenzyl trimethylsilyl ether and a number of aldehydes. Metallic lithium was created after a sequence of reactions, producing the required reaction result. Scientists have used a conventional Horner-Emmons-Wadsworth reaction or a Sonogashira-type reaction to manufacture resveratrol in huge quantities [13].

Using aluminum chloride induction, researchers recovered resveratrol from grapevine leaves. Also, using this technique, Botrytis cinerea in vineyards may be managed. Researchers have also developed methods for producing resveratrol and its analogues using biosynthesis and biomimicry. Researchers also transformed apples using Agrobacterium tumefaciens, and they found that this transformation can create resveratrol with stable inheritance [19]. Researchers have discovered that when synthesizing resveratrol and its equivalents by chemo-enzymatic reaction, the procedure delivers a sustained produce of the preceding produce. Fifty-seven other strategies have also been employed [20].

Advertisement

3. Resveratrol derivatives

As a result, several variants have been invented to boost this molecule’s effectiveness and stability. There are several publications that go into great lengths about the results and uses of resveratrol derivatives. A brief summary of these substances is given in Table 1. By lowering ROS production and protecting oxidative phosphorylation capability, HS-1793 protected mitochondria from cardiac ischemia/reperfusion injury. These resveratrol compounds therefore have a high therapeutic promise for both CVD and a variety of cancers [28].

NanoformulationsChemical CompositionObservationReference
Resveratrol MicroparticlesUsage of magnesium dihydorixde as a supporting baseImprovement of solubility and subsequently bioavailability[21]
Resveratrol encapsulated with silica carrierEncapsulation of the drug alongside functional silica carriers, matrix-type drug releaseMaintenance of cytotoxic properties, improvement of solubility profile[22]
Resveratrol nanoparticlesLoading of the drug onto a chitosan-pectin coreProvision of sustained drug delivery, improved activity, easy modulation of release by variation of parameters[23]
Resveratrol + Gefitinib cocrystalsCombination of resveratrol with a synthetic chemotherapeutic agentImproved stability as well as solubility, indicating potential for increased clinical usage[24]
Nanocomplexation of resveratrol with nanofibrilsFabricated pea protein isolate (PPI) nanofibrils+ resveratrolSignificantly improved solubility, greater surface area for drug incorporation, greater antioxidant potential even at low doses[25]
Resveratrol loaded nanoparticles (NPs)Chitosan and γ-poly(glutamic acid) (γ-PGA)Improved UV stability and enhanced solubility and antioxidant property[26]
Resveratrol loaded onto nanospongesCombination of resveratrol and oxyresveratrolImproved UV stability, solubility as well as antioxidant effect, as well as a satisfactory toxicity profile[27]

Table 1.

Nanotechnological advancements in resveratrol delivery.

3.1 Absorption of resveratrol

Resveratrol is known to have the characteristics of low water solubility, which is reported to be <0.05 mg/mL owing to its chemical structure. Solubility of resveratrol can be enhanced by introducing organic solvents or alcoholic compounds [29]. The ability of resveratrol to form complexes organically with aliphatic hydrocarbons due to the presence of hydroxyl group is the pathway that can be opted for increasing intestinal absorption as well as permeability of cellular walls of enterocytes. It is also absorbed via passive diffusion and transferred in the circulatory system [30]. Free-form sulphate or glucuronide are the altered forms of resveratrol existing in human blood. Free resveratrol has shown low affinity to albumin, predicting that it is a naturally occurring polyphenol reservoir that can bind to human plasma lipoproteins and enter the cells via portal circulation (Figure 1) [31].

Figure 1.

Absorption of resveratrol.

3.2 Metabolism of resveratrol

The metabolism of resveratrol has been studied and shown using a variety of experimental methodologies. It has been noticed that metabolic enzymes and gut bacteria are both necessary for its biotransformation. Moreover, the amount given, any ongoing medical problems, sex, and tissues all have an impact on the rate of metabolism [32].

Members of the UGT family of enzymes catalyze the conjugation of resveratrol with a glucuronic acid moiety at the 3 or 40 hydroxyl group position, changing the antioxidant’s medicinal properties and accelerating its excretion from the body. Human liver microsomes (HLMs) have a large number of UGT enzymes, allowing them to produce more 3-O-glucuronide than 40-O-glucuronide preferentially. Sulfation is a further mechanism of metabolizing tannin. Human sulfotransferase may sulphate resveratrol to one of three different degrees, resulting in resveratrol-3-O-sulfate, resveratrol-40-Osulfate, and resveratrol-3, 40-O-disulfate, according to a study [33].

Moreover, research has demonstrated that resveratrol injection may be a viable therapy for lupus nephritis in MRL/lpr mice by increasing the activity of FcRIIB, allowing the selective removal of B-cells from the bone marrow and the spleen [34]. Resveratrol inhibits Stat3 at low doses, which inhibits the development and function of tumor-evoked regulatory B-cells. As TGF is a downstream target of Stat3, this prevents it from being able to express itself [35].

3.3 Nanotechnological interventions

Because of the poor bioavailability of resveratrol (said to be less than 0.05 mg/mL), dosage has been severely hampered. When mixing circumstances were ideal, the solubility of resveratrol in peanut oil achieved up to 95% [36]. Resveratrol was able to maintain its antioxidant action and extend its shelf life even in a lipid-based solvent. This suggested that peanut oil may be employed as a drug carrier during the formulation design phase as illustrated in Table 2 [40].

DerivativesApplicationsEffectsReferences
Hydroxylated resveratrol derivativesDihydroxystilbeneAnticancerAntiagiogenic effect, inhibits migration[37]
TetramethoxystilbeneAnticancer,Antioxidative effect, antiangiogenic effect[28]
trimethoxy-benzamidinehypertensionInhibits DNA synthesis[38]
Other resveratrol derivativesMitochondria-targeted resveratrol derivativesAnticancer, mitochondrial regulationEnhance solubility, mitochondria targeting[39]
PterostilbeneAnticancerEnhances bioavailability, antioxidative eff[37]
HexahydroxystilbeneAnticancerNF-KB inhibition, SOD inhibition[28]
Methoxylated resveratrol derivativesTetrahydroxystilbeneIschemic heart diseaseApoptosis regulation, KATP channel opening
Resveratrol triacetateAnticancerCell cycle arrest[39]
Fluorinated stilbenesAnticancerAntiproliferative effect[37]
DigalloylresveratrolAnticancerApoptosis regulation[38]

Table 2.

Resveratrol derivatives effects therapeutically.

The solubility and stability of this phytochemical have been attempted to be increased using a variety of nanotechnological treatments. To extend the time that the drug remains in the body and, as a result, boost bioavailability, many nanoformulations have been utilized [41]. In addition to these benefits, using nanoformulations significantly lessens medication metabolism, principally by decreasing glucuronidation. To enhance the dispersion of this drug and reduce problems with its stability and absorption, nanotechnological interventions present a considerable possibility [42].

Advertisement

4. Resveratrol role in cardiovascular disease

4.1 Antiatherogenic effects of resveratrol

The primary mechanism behind the early onset of atherosclerotic lesions is the transformation of macrophages into foam cells following an excessive ingestion of lipoprotein. It’s noteworthy to notice that resveratrol affects a number of chemical agents involved in the lipid metabolism of macrophages (Figure 2) [43].

Figure 2.

Antiatherogenic effects of RES.

Prostaglandin E2, a significant inflammatory substance, is produced by COX-2 (PGE2). Resveratrol controls COX-2 transcriptional activity, preventing PGE2 generation and limiting the inflammatory response to atherosclerosis. PPAR-c, which also has antiatherogenic effects on smooth muscle cells, endothelial cells, and macrophages, promotes modified low-density lipoprotein (LDL) uptake and macrophage maturation. Hence, PPAR-c agonists may have anti-inflammatory properties that help with atherosclerosis prevention. The LXR activation that resveratrol induces must regulate the atherogenesis process [44]. Transmembrane proteins called ABC transporters hydrolyze ATP, and the energy released makes it easier for molecules to cross cell membranes. The inflammatory cardiovascular aberrations associated with aging have recently been related to resveratrol supplementation [45].

4.2 Anti-inflammatory effects of resveratrol

The relevance of developmental vicissitudes in addition to the outmoded jeopardy influences for the growth of CVD is highlighted by recent research. For example, in elderly persons, inferior irritation related with aging raises the risk of coronary artery infection and stroke. Many studies lend credence to the hypothesis that augmented NAD(P)H oxidase action and excessive mitochondrial reactive oxygen species (ROS) formation cause inflammation and endothelial damage as well as the vascular oxidative stress associated with aging [46].

NO, which is essential for preserving endothelial cell activity, seems to be a key ingredient in developmental vicissitudes. NO becomes inactive due to high superoxide concentrations brought on by oxidative damage brought on by aging. Vasomotor dysfunction is severe, endothelial cell death is elevated, and mitochondrial biogenesis is compromised as a result [47].

Recently, resveratrol supplementation has been recommended as a means of preventing the proatherogenic vascular vicissitudes connected to maturing. Resveratrol increases eNOS expression and enhances NO bioavailability, both of which are consistent with these potential benefits. By correcting the physiological changes brought on by oxidative stress and aging, resveratrol acts as a vasoprotective in animals. Moreover, resveratrol inhibits vascular NADPH oxidases, downregulates the expression of tumor necrosis factor-a in vascular and cardiac tissues, and may also prevent the production of mitochondrial ROS in the vascular system (Figure 3) [48].

Figure 3.

The antihypertensive effects of resveratrol.

4.3 Antihypertensive effects of resveratrol

The defining characteristic of the medical condition hypertension is a rise in arterial blood pressure over time. Almost one-fourth of people globally have hypertension. This therapeutic complaint raises the jeopardy of peripheral ischemic heart attack, vascular infection, and stroke, making it possibly the most significant and treatable risk factor for early mortality worldwide. According to the current study, resveratrol may reduce blood pressure through complex processes involving vasodilation, antioxidative activity, and neovascularization. Resveratrol targets sirtuins, and SIRT 1 is the sirtuin that has garnered the most attention from scientists [49].

The vascular endothelium generates more NO when resveratrol increases SIRT 1 expression, which results in vasodilation. Increased endothelial NO stimulates the production of hemeoxygenase-1 (HO-1), a precursor of bilirubin that also has antihypertensive properties. An endogenous vasopressor is ET-1 [50]. The cascading repercussions are examined, potentially linking the benefits of resveratrol in antiatherogenicity and increased longevity to the renin-angiotensin system [19, 51].

By preventing the generation of reactive oxygen species (ROS), phosphorylating Akt and p38 MAPK, activating IKB-α and NF-KB, and limiting the production of ROS, zinfandel safeguards the function of endothelial cells in the vascular system. Tannins also promote the production of thioredoxin and HO-1, which together have antioxidant and myocardial angiogenesis properties [50].

Resveratrol functions better as a prophylactic drug than a treatment for irreversible vascular remodeling, based on our understanding of how it interacts with healthy vascular endothelial cells. Moreover, resveratrol has a low bioavailability due to its rapid metabolism. Finding a resveratrol substitute with a greater therapeutic potential for treating hypertension is therefore essential [52].

Polyphenol advantages for cardio-protection are unique to the significant positive belongings of resveratrol, even if these advantages have been demonstrated by several current researchers in spontaneous models of heart disease. Animal studies have confirmed the protective effects of resveratrol in preventing cardiac fibrosis, autophagy, apoptosis, and oxidative stress in cardiomyocytes [53]. Resveratrol notably lowers the production of ROS through the activation of SIRT1. Moreover, this chemical encourages the growth of the superoxide dismutase (SOD2) enzyme in the mitochondria, reducing oxidative stress in the mitochondria and the resulting cellular damage. Myocardial hypertrophic is the heart’s natural reaction to hemodynamic tension brought on by numerous physiological and pathological conditions. Nonetheless, chronic hypertrophy is regarded to be an unfit process that finally leads to the organism’s demise because of an elevated effort. There are several ways to explain how resveratrol prevents cardiac enlargement (Table 3) [59].

Study TypeDose and Time PeriodResultsReferences
RCT400 mg daily for 30 daysRES dramatically reduced the levels of VCAM, ICAM, and IL-8.
In those with minimal cardiovascular risk, RES may have preventive benefits against the development of atherosclerosis.
Zhao et al. [54]
Meta-analysis> 300 mg dailySystolic blood pressure was considerably lowered with RES.Fogacci et al. [55]
Meta-analysis> 150 mg dailyRES dramatically lowered the amount of systolic blood pressure.Liu et al. [56]
RCTsingle dose (300 mg)Women’s FMD was considerably elevated by RES upon acute supplementation.Marques et al. [57]
RCT500 mg daily for 4 weeksThe levels of HDL cholesterol, the ratio of total to HDL cholesterol, and glycemic control were all improved by RES.Hoseini et al. [58]

Table 3.

Cardio protective effects of RES.

Resveratrol first slows down the obedience and remodeling of slight veins. In contrast, in resveratrol-treated rats with abdominal aortic bands, pressures overwhelm cardiac hypertrophy, and dysfunction is restored. The reason for these effects may be the rise in eNOS/NO [60]. The resveratrol blocks the serine-threonine kinase AMP-activated protein kinase (LKB1), a downstream signaling molecule, from being inhibited by oxidative stress (AMPK). Last but not least, resveratrol suppresses cardiac transcription of the angiotensin II receptor, AT1a, which helps to stop the development of heart hypertrophy [61].

Resveratrol has also been demonstrated to offer cardioprotective advantages in various situations. Since it results in cardiac enlargement and functional impairment, low ambient temperature is recognized as a substantial CVD jeopardy influence. Treatment with resveratrol successfully inhibits these changes by preventing cardiomyocyte apoptosis [62]. Despite the common occurrence of autophagic dysfunction in diabetics, resveratrol promotes the growth of functional autophagy mechanisms within cells, has a positive impact on this diabetic cardiomyopathy. By preventing the ROS/ERK/TGF-b/periostin pathway from being activated, resveratrol reduces myocardium fibrosis in diabetic mice. Resveratrol also enhances the miR-130a gene, suppresses the miR-34a gene, blocks the miR-34a/Sirt1 gene, lowers oxidative stress, and lowers cardiac fibrosis and inflammation to protect against myocardial traumas brought on by myocardial infarction or hypoxia/reoxygenation damage [63].

As already noted, resveratrol defends the heart from endogenous causes like irritation, dyslipidemia, and endothelial dysfunction. Whether resveratrol also shields the myocardium from external elements like therapeutic medicines or endotoxin lipopolysaccharides has been the subject of more recent studies. Moreover, doxorubicin’s anticancer effects are enhanced by resveratrol by increasing its cellular absorption. These results suggest that resveratrol might reduce cardiotoxicity and work in concert with doxorubicin to kill cancer cells (Figure 4) [64].

Figure 4.

Illustration of the cardioprotective effects of resveratrol.

4.4 Antimetabolic syndrome

The metabolic condition is a lingering criticism that increases the jeopardy of diabetes and cardiovascular syndrome. This disease is characterized by the occurrence of, at the slightest, three of the subsequent health issues: elevated fasting glucose, high blood pressure, high serum triglycerides, high blood sugar, and abdominal (central) obesity. Metabolic syndrome, which affects around one-fourth of adult populations globally, is rather common in modern civilizations [65]. Nowadays, drugs to treat hypercholesterolemia are used as part of the treatment for metabolic syndrome. Insulin performs to be the primary hormone implicated in metabolic syndrome, despite the datum that the pathophysiology of this disorder is incredibly complicated and only slightly altered. According to some professionals, prediabetes and metabolic syndrome are merely two different diagnoses that can only be distinguished by a small number of biomarkers [66].

Many studies suggest that zinfandel might be valuable in treating metabolic disease. Resveratrol exhibited a more significant consequence on the metabolic disease, hepatic oxidative pressure, and insulin sensitivity in rats compared to metformin. Resveratrol therapy reduced blood cholesterol, C-reactive protein, and body mass indices in a pig model. These findings suggest that resveratrol has a beneficial effect on metabolic syndrome risk variables [67]. Resveratrol has the potential to improve cardiovascular health while easing the burden of chronic metabolic disease. In the mice used in the other metabolic syndrome animal model, zinfandel treatment was found to have a positive effect on the liver and skeletal muscle glucose metabolism and result in better control of glucose points [68].

Advertisement

5. Challenges and future perspectives

It has been investigated if resveratrol, a well-known polyphenolic stilbene, may be used to treat a variability of neoplasms, counting B-cell malignancies. It now appears to be a promising molecule for prospective therapeutic usage due to its extensive accessibility and versatility [25].

Forecasts based on epidemiological data from Western Europe and the United States suggest that a low rate of systemic treatment initiation would result in an increase in patients. Moreover, the increased prevalence may be linked to an aging population, toxicity brought on by the accumulation of treatment drugs over time, and an increase in the probability of recurrence [69]. Also, it is vital to understand the genetic influences that subsidize the expansion of disease. Non-Hodgkin lymphoma and autoimmune illnesses are both caused by inherited factors, according to a study by Din et al. An overlay of jeopardy influences may designate that cancer may develop if these genetic abnormalities are not addressed [25]. In a different study, Chang et al. [70] looked into the relationship between hematological malignancies and family history. Despite the absence of a clear link to environmental factors, a significant increase in the risk of incidence persisted if a first-degree relative had a history of cancer by genetics as well. Despite being large wine consumers, these nations have a superior prevalence of B-cell malignancies than the respite of the ecosphere. Resveratrol’s low solubility and stability, which have been demonstrated to directly influence its bioavailability, may be a significant factor. To address these obstacles, new treatment strategies are being used, although development is still sluggish. To prevent the onset of disease and improve the effectiveness of therapy, it has been acknowledged that a variability of aspects in accumulation to alimentary supplements need to be looked at. The factors mentioned above could have a big impact on this [51].

Several pre-clinical studies have been carried out to evaluate the pharmacological effects of resveratrol; nevertheless, significant improvements in clinical research are still needed. This necessitates thorough evaluations of the protection and noxiousness silhouette of this substance, taking into account its connections with additional phytochemicals and synthetic anticancer drugs, as well as clinical study in a range of groups [71]. Its natural sources provide a number of formulation difficulties, including truncated solubility and long-term constancy, for instance, fast universal metabolism, which limits its distribution to aim muscles and affects its absorption. In the end, this makes it more challenging to get the desired therapeutic benefits. Moreover, resveratrol prescription has resulted in nephrotoxicity in multiple myeloma patients; the danger of toxicity linked through abiding dosage for chronic conditions must thus be carefully considered. Before extending its investigation as a possible anticancer drug and opening up new possibilities for diverse usage, these difficulties must be solved [4].

The bioavailability of this drug has been enhanced using a number of nano-strategies as the emphasis on nano technical distribution endures to produce. To overcome problems with medication delivery optimization, several strategies are being employed. The authors have highlighted many of these therapeutic methods to stimulate conversation on the value of these nanoformulations. The poor potency of current preparations is a huge unfulfilled need that could potentially be addressed by manufacturing resveratrol analogues, some of which have been investigated, to greatly boost its anticancer capabilities [51].

The aforementioned is important to begin the protection of resveratrol in gruesome patients, even if it has been proven to be well tolerated in healthy persons at both short- and long-term dosages. Further focused clinical trials with a wider variety of demographics are necessary to completely understand the safety and effectiveness of this chemical by way of its pharmacological probable [51].

Advertisement

6. Conclusion

Using botanical compounds to ward against or perhaps remedy illnesses has grown in popularity in recent years. Resveratrol is a cheap, accessible, and simple-to-acquire small molecule that can be functionalized. It has a variety of pharmacological actions and is not poisonous, making it suitable for industrial application. This chapter focuses on the research of new resveratrol developments, emphasizing the substance’s botanical supplies, production techniques, customization, and possible therapeutic applications. Several protective benefits of resveratrol are seen in CVDs. They include actions that reduce the risk of atherosclerosis, reduce inflammation, lower blood pressure, promote cardio protection, and alter metabolism. With more investigation, it could be viable to employ resveratrol in the diagnosis or prophylaxis of CVDs. For example, we propose that resveratrol might be administered as a preventive medication to individuals with coronary artery hypertension undergoing stent placement, aiming to mitigate complications and reduce mortality associated with the procedure. Resveratrol impersonates calorie constraint in yeast by activating Sirt2, increasing DNA integrity, and lengthening life distance, which substantially implies that this substance might have additional significant impacts on adult wellness.

Advertisement

Acknowledgments

Authors are thankful to the Government College University for providing literature collection facilities. We confirm the final authorship for this manuscript, and we ensure that anyone else who contributed to the manuscript but does not qualify for authorship has been acknowledged with their permission. We acknowledge that all listed authors have made a significant scientific contribution to the research in the manuscript, approved its claims, and agreed to be an author.

Advertisement

Conflict of interest

Authors declare that they have no conflict of interest.

Advertisement

Funding

The authors declare that no funds, grants, or other support were received during the preparation of this manuscript.

Advertisement

Ethical approval

This article does not contain any studies with human participants or animals performed by any of the authors.

Consent to participate

Corresponding and all the co-authors are willing to participate in this manuscript.

Consent for publication

All authors are willing for publication of this manuscript.

Data availability

Even though adequate data has been given in the form of tables and figures, however, all authors declare that if more data required then the data will be provided on request basis.

References

  1. 1. Wijekoon HS, Kim S, Bwalya EC, Fang J, Aoshima K, Hosoya K, et al. Anti-arthritic effect of pentosan polysulfate in rats with collagen-induced arthritis. Research in Veterinary Science. 2019;122:179-185
  2. 2. Vestergaard M, Ingmer H. Antibacterial and antifungal properties of resveratrol. International Journal of Antimicrobial Agents. 2019;53(6):716-723
  3. 3. Elshaer M, Chen Y, Wang XJ, Tang X. Resveratrol: An overview of its anti-cancer mechanisms. Life Sciences. 2018;207:340-349
  4. 4. Wu S-X, Xiong R-G, Huang S-Y, Zhou D-D, Saimaiti A, Zhao C-N, et al. Effects and mechanisms of resveratrol for prevention and management of cancers: An updated review. Critical Reviews in Food Science and Nutrition. 2022;63:12422-12440
  5. 5. Patra S, Mishra SR, Behera BP, Mahapatra KK, Panigrahi DP, Bhol CS, et al. Autophagy-modulating phytochemicals in cancer therapeutics: Current evidences and future perspectives. Seminars in Cancer Biology. 2022;80:205-217
  6. 6. Kashyap D, Tuli HS, Yerer MB, Sharma A, Sak K, Srivastava S, et al. Natural product-based nanoformulations for cancer therapy: Opportunities and challenges. Seminars in Cancer Biology. 2021;69:5-23
  7. 7. Abadi AJ, Mirzaei S, Mahabady MK, Hashemi F, Zabolian A, Hashemi F, et al. Curcumin and its derivatives in cancer therapy: Potentiating antitumor activity of cisplatin and reducing side effects. Phytotherapy Research. 2022;36(1):189-213
  8. 8. Denlinger N, Bond D, Jaglowski S. CAR T-cell therapy for B-cell lymphoma. Current Problems in Cancer. 2022;46(1):100826
  9. 9. Wang G-Z, Shang R, Cheng W-M, Fu Y. Irradiation-induced heck reaction of unactivated alkyl halides at room temperature. Journal of the American Chemical Society. 2017;139(50):18307-18312
  10. 10. Wang G-Z, Shang R, Fu Y. Irradiation-induced palladium-catalyzed decarboxylative heck reaction of aliphatic N-(acyloxy) phthalimides at room temperature. Organic Letters. 2018;20(3):888-891
  11. 11. Martínez AV, García JI, Mayoral JA. An expedient synthesis of resveratrol through a highly recoverable palladium catalyst. Tetrahedron. 2017;73(38):5581-5584
  12. 12. Andrus MB, Liu J. Synthesis of polyhydroxylated ester analogs of the stilbene resveratrol using decarbonylative heck couplings. Tetrahedron Letters. 2006;47(32):5811-5814
  13. 13. Perin G, Barcellos AM, Luz EQ , Borges EL, Jacob RG, Lenardão EJ, et al. Green hydroselenation of aryl alkynes: Divinyl selenides as a precursor of resveratrol. Molecules. 2017;22(2):327
  14. 14. Uzura S, Sekine-Suzuki E, Nakanishi I, Sonoda M, Tanimori S. A facile and rapid access to resveratrol derivatives and their radioprotective activity. Bioorganic & Medicinal Chemistry Letters. 2016;26(16):3886-3891
  15. 15. Solladié G, Pasturel-Jacopé Y, Maignan J. A re-investigation of resveratrol synthesis by Perkins reaction. Application to the synthesis of aryl cinnamic acids. Tetrahedron. 2003;59(18):3315-3321
  16. 16. Alonso F, Riente P, Yus M. Synthesis of resveratrol, DMU-212 and analogues through a novel Wittig-type olefination promoted by nickel nanoparticles. Tetrahedron Letters. 2009;50(25):3070-3073
  17. 17. Tian B, Liu J. Resveratrol: A review of plant sources, synthesis, stability, modification and food application. Agriculture. 2020;100(4):1392-1404
  18. 18. Aissa C. Mechanistic manifold and new developments of the Julia–Kocienski reaction. European Journal of Organic Chemistry. 2009;2009(12):1831-1844
  19. 19. Gómez Baraibar Á, Reichert D, Mügge C, Seger S, Gröger H, Kourist R. A one-pot cascade reaction combining an encapsulated decarboxylase with a metathesis catalyst for the synthesis of bio-based antioxidants. Angewandte Chemie (International Ed. in English). 2016;55(47):14823-14827
  20. 20. Goh YL, Cui YT, Pendharkar V, Adsool VA. Toward resolving the resveratrol conundrum: Synthesis and in vivo pharmacokinetic evaluation of BCP–resveratrol. ACS Medicinal Chemistry Letters. 2017;8(5):516-520
  21. 21. Chung JH, Lee J-S, Lee HGJC, Biointerfaces SB. Resveratrol-loaded chitosan–γ-poly (glutamic acid) nanoparticles: Optimization, solubility, UV stability, and cellular antioxidant activity. Colloids and Surfaces. B, Biointerfaces. 2020;186:110702
  22. 22. Dhakar NK, Matencio A, Caldera F, Argenziano M, Cavalli R, Dianzani C, et al. Comparative evaluation of solubility, cytotoxicity and photostability studies of resveratrol and oxyresveratrol loaded nanosponges. Pharmaceutics. 2019;11(10):545
  23. 23. Sarma S, Agarwal S, Bhuyan P, Hazarika J, Ganguly M. Resveratrol-loaded chitosan–pectin core–shell nanoparticles as novel drug delivery vehicle for sustained release and improved antioxidant activities. Royal Society Open Science. 2022;9(2):210784
  24. 24. Spogli R, Bastianini M, Ragonese F, Iannitti RG, Monarca L, Bastioli F, et al. Solid dispersion of resveratrol supported on magnesium dihydroxide (resv@ mdh) microparticles improves oral bioavailability. Nutrients. 2018;10(12):1925
  25. 25. Ioniţă S, Lincu D, Mitran R-A, Ziko L, Sedky NK, Deaconu M, et al. Resveratrol encapsulation and release from pristine and functionalized mesoporous silica carriers. Pharmaceutics. 2022;14(1):203
  26. 26. Yi J, He Q , Peng G, Fan Y. Improved water solubility, chemical stability, antioxidant and anticancer activity of resveratrol via nanoencapsulation with pea protein nanofibrils. Food Chemistry. 2022;377:131942
  27. 27. Zhai L, Zhang Z, Guo L, Dong H, Yu J, Zhang G. Gefitinib-resveratrol cocrystal with optimized performance in dissolution and stability. Journal of Pharmaceutical Sciences. 2022;111(12):3224-3231
  28. 28. Jeong SH, Hanh TM, Kim HK, Lee SR, Song I-S, Noh SJ, et al. HS-1793, a recently developed resveratrol analogue protects rat heart against hypoxia/reoxygenation injury via attenuating mitochondrial damage. Bioorganic & Medicinal Chemistry Letters. 2013;23(14):4225-4229
  29. 29. Laza-Knoerr AL, Gref R, Couvreur P. Cyclodextrins for drug delivery. Journal of Drug Targeting. 2010;18(9):645-656
  30. 30. Delmas D, Aires V, Limagne E, et al. Transport, stability, and biological activity of resveratrol. Annals of the New York Academy of Sciences. 2011;1215:48-59
  31. 31. Jannin B, Menzel M, Berlot J-P, Delmas D, Lançon A, Latruffe N. Transport of resveratrol, a cancer chemopreventive agent, to cellular targets: Plasmatic protein binding and cell uptake. Biochemical Pharmacology. 2004;68:1113-1118
  32. 32. Wang P, Sang S. Metabolism and pharmacokinetics of resveratrol and pterostilbene. BioFactors. 2018;44(1):16-25
  33. 33. Miksits M, Maier-Salamon A, Aust S, Thalhammer T, Reznicek G, Kunert O, et al. Sulfation of resveratrol in human liver: Evidence of a major role for the sulfotransferases SULT1A1 and SULT1E1. Xenobiotica. 2005;35(12):1101-1119
  34. 34. Jhou J-P, Chen S-J, Huang H-Y, Lin W-W, Huang D-Y, Tzeng S-J. Upregulation of FcγRIIB by resveratrol via NF-κB activation reduces B-cell numbers and ameliorates lupus. Experimental & Molecular Medicine. 2017;49(9):e381-e381
  35. 35. Lee-Chang C, Bodogai M, Martin-Montalvo A, Wejksza K, Sanghvi M, Moaddel R, et al. Inhibition of breast cancer metastasis by resveratrol-mediated inactivation of tumor-evoked regulatory B cells. Journal of Immunology. 2013;191(8):4141-4151
  36. 36. Galiniak S, Aebisher D, Bartusik-Aebisher D. Health benefits of resveratrol administration. Acta Biochimica Polonica. 2019;66(1):13-21
  37. 37. Wu Z, Liu B, Liu J, Zhang Q , Liu J, Chen N, et al. Resveratrol inhibits the proliferation of human melanoma cells by inducing G1/S cell cycle arrest and apoptosis. Molecular Medicine Reports. 2015;11(1):400-404
  38. 38. Ruan B-F, Lu X-Q , Song J, Zhu H-L. Derivatives of resveratrol: Potential agents in prevention and treatment of cardiovascular disease. Current Medicinal Chemistry. 2012;19(24):4175-4183
  39. 39. He S, Yan X. From resveratrol to its derivatives: New sources of natural antioxidant. Current Medicinal Chemistry. 2013;20(8):1005-1017
  40. 40. Li T, Guo Q , Qu Y, Li Y, Liu H, Liu L, et al. Solubility and physicochemical properties of resveratrol in peanut oil. Food Chemistry. 2022;368:130687
  41. 41. Kumar A, Kurmi BD, Singh A, Singh D. Potential role of resveratrol and its nano-formulation as anti-cancer agent. Exploration of Targeted Anti-tumor Therapy. 2022;3(5):643
  42. 42. Li Y, Zhang R, Zhang Q , Luo M, Lu F, He Z, et al. Dual strategy for improving the oral bioavailability of resveratrol: Enhancing water solubility and inhibiting glucuronidation. Journal of Agricultural and Food Chemistry. 2021;69(32):9249-9258
  43. 43. Aviram M, Rosenblat M. Paraoxonases 1, 2, and 3, oxidative stress, and macrophage foam cell formation during atherosclerosis development. Free Radical Biology & Medicine. 2004;37(9):1304-1316
  44. 44. Voloshyna I, Hai O, Littlefield MJ, Carsons S, Reiss AB. Resveratrol mediates anti-atherogenic effects on cholesterol flux in human macrophages and endothelium via PPARγ and adenosine. European Journal of Pharmacology. 2013;698(1-3):299-309
  45. 45. Lin M-T, Yen M-L, Lin C-Y, Kuo M-L. Inhibition of vascular endothelial growth factor-induced angiogenesis by resveratrol through interruption of Src-dependent vascular endothelial cadherin tyrosine phosphorylation. Molecular Pharmacology. 2003;64(5):1029-1036
  46. 46. Donato AJ, Eskurza I, Silver AE, Levy AS, Pierce GL, Gates PE, et al. Direct evidence of endothelial oxidative stress with aging in humans: Relation to impaired endothelium-dependent dilation and upregulation of nuclear factor-κB. Circulation Research. 2007;100(11):1659-1666
  47. 47. Gurusamy N, Ray D, Lekli I, Das DK. Red wine antioxidant resveratrol-modified cardiac stem cells regenerate infarcted myocardium. Journal of Cellular and Molecular Medicine. 2010;14(9):2235-2239
  48. 48. Zhang H, Morgan B, Potter BJ, Ma L, Dellsperger KC, Ungvari Z, et al. Resveratrol improves left ventricular diastolic relaxation in type 2 diabetes by inhibiting oxidative/nitrative stress: In vivo demonstration with magnetic resonance imaging. American Journal of Physiology. Heart and Circulatory Physiology. 2010;299(4):H985-H994
  49. 49. Hsu C-N, Hou C-Y, Chang-Chien G-P, Lin S, Chan JY, Lee C-T, et al. Maternal resveratrol therapy protected adult rat offspring against hypertension programmed by combined exposures to asymmetric dimethylarginine and trimethylamine-N-oxide. The Journal of Nutritional Biochemistry. 2021;93:108630
  50. 50. Huang X, Liu Y, Zou Y, Liang X, Peng Y, McClements DJ, et al. Encapsulation of resveratrol in zein/pectin core-shell nanoparticles: Stability, bioaccessibility, and antioxidant capacity after simulated gastrointestinal digestion. Food Hydrocolloids. 2019;93:261-269
  51. 51. Gupta DS, Gadi V, Kaur G, Chintamaneni M, Tuli HS, Ramniwas S, et al. Resveratrol and its role in the management of B-cell malignancies—A recent update. Biomedicine. 2023;11(1):221
  52. 52. Pauluk D, Padilha AK, Khalil NM, Mainardes RM. Chitosan-coated zein nanoparticles for oral delivery of resveratrol: Formation, characterization, stability, mucoadhesive properties and antioxidant activity. Food Hydrocolloids. 2019;94:411-417
  53. 53. Zhang F, Khan MA, Cheng H, Liang L. Co-encapsulation of α-tocopherol and resveratrol within zein nanoparticles: Impact on antioxidant activity and stability. Journal of Food Engineering. 2019;247:9-18
  54. 54. Zhao W, Li A, Feng X, Hou T, Liu K, Liu B, et al. Metformin and resveratrol ameliorate muscle insulin resistance through preventing lipolysis and inflammation in hypoxic adipose tissue. Cell Signaling. 2016;28(9):1401-1411
  55. 55. Fogacci F, Tocci G, Presta V, Fratter A, Borghi C, Cicero AFG. Effect of resveratrol on blood pressure: A systematic review and meta-analysis of randomized, controlled, clinical trials. Critical Reviews in Food Science and Nutrition. 2019;59(10):1605-1618
  56. 56. Liu Y, Ma W, Zhang P, He S, Huang D. Effect of resveratrol on blood pressure: A meta-analysis of randomized controlled trials. Clinical Nutrition. 2015;34(1):27-34
  57. 57. Marques BCAA, Trindade M, Aquino JCF, Cunha AR, Gismondi RO, Neves MF, et al. Beneficial effects of acute trans-resveratrol supplementation in treated hypertensive patients with endothelial dysfunction. Clinical and Experimental Hypertension. 2018;40(3):218-223
  58. 58. Hoseini A, Namazi G, Farrokhian A, Reiner Ž, Aghadavod E, Bahmani F, et al. The effects of resveratrol on metabolic status in patients with type 2 diabetes mellitus and coronary heart disease. Food and Function. 1 Sep 2019;10(9):6042-6051. DOI: 10.1039/c9fo01075k. Epub 2019 September 5. PMID: 31486447
  59. 59. Liu F, Ma D, Luo X, Zhang Z, He L, Gao Y, et al. Fabrication and characterization of protein-phenolic conjugate nanoparticles for co-delivery of curcumin and resveratrol. Food Hydrocolloids. 2018a;79:450-461
  60. 60. Torres V, Hamdi M, Millán de la Blanca M, Urrego R, Echeverri J, López-Herrera A, et al. Resveratrol–cyclodextrin complex affects the expression of genes associated with lipid metabolism in bovine in vitro produced embryos. Reproduction in Domestic Animals. 2018;53(4):850-858
  61. 61. Liu R, Dai L, Zou Z, Si C. Drug-loaded poly (L-lactide)/lignin stereocomplex film for enhancing stability and sustained release of trans-resveratrol. International Journal of Biological Macromolecules. 2018b;119:1129-1136
  62. 62. da Silva R, Teixeira JA, Nunes WDG, Zangaro GAC, Pivatto M, Caires FJ, et al. Resveratrol: A thermoanalytical study. Food Chemistry. 2017;237:561-565
  63. 63. Liang Q , Ren X, Zhang X, Hou T, Chalamaiah M, Ma H, et al. Effect of ultrasound on the preparation of resveratrol-loaded zein particles. Journal of Food Engineering. 2018;221:88-94
  64. 64. Pineda-Sanabria SE, Robertson IM, Sykes BD. Structure of trans-resveratrol in complex with the cardiac regulatory protein troponin C. Biochemistry. 2011;50(8):1309-1320
  65. 65. Chan GG, Koch CM, Connors LH. Blood proteomic profiling in inherited (ATTRm) and acquired (ATTRwt) forms of transthyretin-associated cardiac amyloidosis. Journal of Proteome Research. 2017;16(4):1659-1668
  66. 66. Singh D, Mendonsa R, Koli M, Subramanian M, Nayak SK. Antibacterial activity of resveratrol structural analogues: A mechanistic evaluation of the structure-activity relationship. Toxicology and Applied Pharmacology. 2019;367:23-32
  67. 67. Chalal M, Vervandier-Fasseur D, Meunier P, Cattey H, Hierso J-C. Syntheses of polyfunctionalized resveratrol derivatives using Wittig and heck protocols. Tetrahedron. 2012;68(20):3899-3907
  68. 68. Fang J-G, Zhou B. Structure−activity relationship and mechanism of the tocopherol-regenerating activity of resveratrol and its analogues. Journal of Agricultural and Food Chemistry. 2008;56(23):11458-11463
  69. 69. Kanas G, Ge W, Quek RG, Keeven K, Nersesyan K, Arnason JE. Epidemiology of diffuse large B-cell lymphoma (DLBCL) and follicular lymphoma (FL) in the United States and Western Europe: Population-level projections for 2020-2025. Leukemia & Lymphoma. 2022;63(1):54-63
  70. 70. Chang ET, Smedby KE, Hjalgrim H, Porwit-MacDonald A, Roos G, Glimelius B, et al. Family history of hematopoietic malignancy and risk of lymphoma. Journal of the National Cancer Institute. 2005;97(19):1466-1474
  71. 71. Ashique S, Afzal O, Yasmin S, Hussain A, Altamimi MA, Webster TJ, et al. Strategic nanocarriers to control neurodegenerative disorders: Concept, challenges, and future perspective. International Journal of Pharmaceutics. 2023;633:122614

Written By

Fakhar Islam, Umber Shehzadi, Farhan Saeed, Rabia Shabir Ahmad, Muhammad Umair Arshad, Muhammad Sadiq Naseer, Fatima Tariq, Rehman Ali, Sadaf Khurshid, Ghulam Hussain, Aftab Ahmad, Muhammad Afzaal, Rabia Akram, Osman Tuncay Agar, Ali Imran and Hafiz A.R. Suleria

Submitted: 24 April 2023 Reviewed: 10 April 2024 Published: 27 May 2024