Abstract
Neurodegeneration is a process leading to the progressive loss of structure and functions of neurons. Many neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease, and Huntington’s disease have shown many common points at the subcellular level. Neurons are metabolically active cells and need a high amount of energy. Mitochondria are known as the energy synthesis center for cells, involved in the synthesis of adenosine triphosphate by oxidative phosphorylation. Rather than just being an energy synthesis center, it has critical importance for many cellular functions such as calcium homeostasis, cell proliferation, cell growth, and apoptosis. In the process of mitochondrial dysfunction, cellular functions are disrupted and cells enter the apoptotic or necrotic pathway. Resveratrol (trans-3,5,4-trihydoxystilbene), a plant-derived polyphenol found in the seed of grapes, berries, peanuts, and wine, has many biological effects such as inhibition of lipid peroxidation, scavenging of free radicals, changes in eicosanoid synthesis, inhibition of platelet aggregation, anti-inflammatory and anticancer activity, and regulation of lipid metabolism. Through the reviewed literature, the current study investigated the protective role of resveratrol in neurodegenerative diseases. Studies show that resveratrol moderates mitochondrial function, redox status, and cellular dynamics in both in vivo and in vitro experimental models of neurodegeneration. Resveratrol suppresses reactive oxygen species production by reducing the activity of complex III due to its competition effect with coenzyme Q. In the present work, we discussed the protective effects of resveratrol on neurodegeneration, neurodegenerative diseases, and the redox biology of the mitochondria.
Main Points
Summary of resveratrol effects on neurodegeneration.
Summary of resveratrol effects on neurodegenerative diseases.
Summary of resveratrol effects on redox biology of the mitochondria.
Introduction
Neurodegenerative disease is a broad definition used to explain a group of diseases distinguished by progressive failure of neuronal performance and structural deterioration.1 Mitochondria are key cytoplasmic organelles that play an important role in many cellular activities such as adenosine triphosphate (ATP) production, oxidative phosphorylation, maintenance of intracellular Ca+2 balance (calcium homeostasis), redox signaling, and apoptotic cell death.2 Neurons are particularly sensitive to reactive oxygen species (ROS) than other organs due to their high oxygen need and poor antioxidant defense. Early stages of neurodegenerative illnesses are critically affected by any breakdown of this regulating system.3 Mitochondrial dysfunction is thought to play a fundamental role in the pathogenesis of neurodegenerative diseases, such as Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), and amyotrophic lateral sclerosis (ALS). Damage to mitochondrial DNA (mtDNA) also plays an important role in the development of many diseases by causing ATP deprivation, leading to oxidative stress, high amount of ROS production, and apoptosis. Numerous antioxidant treatments have been found to shield neurons from oxidative stress by halting the generation of free radicals and preserving normal homeostasis.4 Natural compounds that can modulate bioenergy have the potential to be used as neuroprotective agents. Resveratrol, a plant-derived polyphenol, has protective effects on neurodegenerative diseases with its antioxidant, anti-inflammatory, and metal-chelating properties by crossing the blood–brain barrier.5 It has been shown to be effective in preventing redox degradation in brain cells.6 Furthermore, resveratrol exerts antiapoptotic effects and modulates central nervous system function by modulating mitochondrial function, redox biology and dynamics.6,7 Studies show that antioxidant compounds such as resveratrol can modulate mitochondrial function and dynamics and can have a cytoprotective effect.8 Therefore, this review focuses on the relationship between neurodegenerative diseases, resveratrol, and the redox biology of the mitochondria.
Mitochondrial Biology
Mitochondria are the main energy provider for cellular functions by generating ATP. Mitochondria contain their own pool of mtDNA, which is critical for cell development, functionality, and survival. It has 4 parts: outer membrane, inner membrane, intermembrane space (IMS), and matrix.2 The outer membrane consists of specific transport proteins and porins, and they allow free diffusion of small molecules into IMS. This membrane has many enzymes required for phospholipid and phosphoric acid synthesis, as well as monoamine oxidase, cytochrome c (Cyt c) reductase enzymes that play a catalyst role in the Krebs cycle. The inner membrane shows selective permeability, the electron transport chain (ETC) enzymes and enzymes of oxidative phosphorylation are situated here. They also have a means of transport for particular substances such as ATP, adenosine diphosphate, pyruvate, succinate, malate, citrate, and α-ketoglutarate.9
Generating Energy
Adenosine triphosphate can be produced through cellular respiration, the tricarboxylic acid cycle, ETC, and oxidative phosphorylation.9 To understand mitochondria-related diseases, it would be appropriate to remember the ETC and oxidative phosphorylation pathway.7 In many studies, it has been observed that electrons are transferred from reduced coenzymes to oxygen through a series of carriers in the respiratory system. Electrons flow from negative potential to more positive potential. These carriers, aligned according to their standard redox potentials, yield different complexes. The complexes that make up the protein components of the mitochondrial respiratory chain are multienzyme complexes with their own specific composition.7,9
Oxidative Stress
Under normal physiological conditions, there is a balance between ROS and antioxidants. The deterioration of this balance in favor of ROS, that is, the accumulation of free radicals in the cell or the insufficiency of endogenous defense systems is defined as oxidative stress. Molecular oxygen has 2 unpaired electrons in the parallel spin state. Oxidation reactions occur with a single electron transfer to molecular oxygen by enzymes containing transition metals (such as Fe, Cu) in the organism.10 The ROS includes both free radicals and bound radical molecules such as hydrogen peroxide (H2O2), superoxide radical (O2 .- ), singlet oxygen (1/2 O2), and hydroxyl radical (OH.).10 These free oxygen radicals combine with other radicals or non-radical agents. Thus, it causes many biological effects at the molecular level in the organism.11 Superoxide anion is a highly reactive product generated in the ETC via mitochondrial complexes I and III. As depicted in Figure 1, the passage of O2 _ into the mitochondrial matrix leads to the generation of H2O2 by connecting with 2 protons (H+), a reaction catalyzed by superoxide dismutase (SOD). Superoxide may also react with iron–sulfur proteins releasing ferrous iron (Fe+2). Hydrogen peroxide can be catalyzed into water by antioxidant enzymes in the mitochondrial matrix which include peroxiredoxin, catalase, and glutathione peroxidase.10
Figure 1.
Possible mitochondrial hydrogen peroxide production. The production of H2O2 is a potential redox signal. The generation of H2O2 can modulate the activity of target proteins via the oxidation of critical protein thiols. Redox signaling can alter the enzyme, kinase, and phosphatase activities and also can affect the transcription factors in the mitochondria, cytosol, or nucleus. GPX, glutathione peroxidases; GR, glutathione reductase; GSH, reduced form glutathione; GSSG, oxidized form glutathione; HO2 •, hydroperoxyl radical; O2−, superoxide; Prx, peroxiredoxin; SOD, superoxide dismutase; Trx, thioredoxin; TrxR, NADPH-thioredoxin reductase.
Mitochondrial Redox Biology
The mitochondrial respiratory chain is important in the generation of reactive species production because of the electron leaks in complex I and complex III.12 Oxygen is converted to water in mitochondria as a result of ETC reactions. In this metabolic process, 2%-3% of the oxygen in the mitochondria does not turn into water but forms a source for the formation of oxygen-derived radicals.13 As shown in Figure 1, with a wide range of reactions, including proliferation, differentiation, and cellular death pathways, mitochondrial H2O2 plays a significant role in the regulation of cellular redox status.
Low to intermediate levels of H2O2 contribute to redox-sensitive signaling and transcriptional regulation, while high levels cause oxidative damage to cell components.10 Free radicals can also originate from nitric oxide (NO). There is evidence to suggest that reactive nitrogen species, such as NO, can be formed in the mitochondria. Superoxide can react with NO produced by mitochondrial nitric oxide synthase, resulting in the formation of peroxynitrite. Peroxynitrite causes protein nitration and this modification leads to conformational changes in proteins, which triggers cell necrosis. High concentrations of NO can inhibit Cyt c oxidase and modulate mitochondrial energy metabolism by reducing oxygen consumption causing metabolic hypoxia. When cellular respiration is inhibited by NO, an outflow of Ca2+ occurs from the mitochondria and this causes activation of Ca2+-dependent proteases.14
The Effect of Mitochondrial Dysfunction in Neurogenerative Disease
Alzheimer’s Disease
Alzheimer’s disease is the most prevalent age-related neurodegenerative illness. Mutations in genes encoding amyloid precursor protein (APP), presenilin-1 and presenilin-2, or some alleles of apolipoprotein E pose a risk for AD.15 A series of enzymes known as beta (β) and gamma (γ) secretase sequentially cleave APP to produce the proteolytic product known as amyloid-beta (Aβ) peptide.16 Alzheimer's disease is characterized by extracellular senile amyloid plaques mainly composed of hydrophobic 40-42 amino acid long Aβ peptides and intracellular neurofibrillary tau tangles formed as a result of aggregation of hyperphosphorylated tau proteins.16 There are several different theories explaining why there is neuronal cell death in this disease. The most important among these is the amyloid hypothesis. According to this theory, impaired Aβ metabolism triggers Aβ peptide aggregation and causes synaptic damage, neurofibrillary tangles, inflammatory response, increased oxidative stress, cell death, and ultimately AD. In particular, increasing numbers of studies are believed to be associated with well-defined neuropathological symptoms of AD, as well as intracellular lesions such as disruption of Ca2+ homeostasis, accumulation of Aβ, presence of Aβ in mitochondria causing mitochondrial dysfunction, and increased ROS production.17
The APP indeed accumulates in mitochondrial channels and Aβ has been found to interact with some mitochondrial proteins.18 It has been argued that 2 molecular binding targets for Aβ in mitochondria, Aβ-binding alcohol dehydrogenase and cyclophilin D, are associated with this neurodegenerative disease. Decreased mitochondria numbers and reduced brain glucose metabolism are the earliest detected abnormalities in AD brains.19 Decreased activity of key enzymes in oxidative metabolism, including the pyruvate dehydrogenase complex and alpha-ketoglutarate dehydrogenase in AD has been determined and significantly correlated with clinical state.20 Since the functions of all enzymes are impaired in the presence of Aβ, a relationship between cascading amyloid theory and mitochondrial dysfunction has been proposed.21 It has been determined that oxidative damage is both a biomarker of AD and plays a role in Aβ toxicity.
Parkinson’s Disease
Parkinson’s disease is the second most common neurodegenerative disease, involving the presence of Lewy bodies composed of fibrillary α-synuclein in the substantia nigra pars compacta (SNPC) and deterioration of dopaminergic neurons.22 This death of neurons is accompanied by the loss of astrocytes and a noteworthy increase and activation of microglia in the SNPC. Both chemical and genetic evidence suggest that mitochondrial dysfunction is associated with PD.23 Several environmental toxins with inhibition of mitochondrial complex I have been shown to be involved in the epidemiology of sporadic PD.23 The first link between mitochondrial dysfunction and PD emerged in 1983 and higher expression of Complex I and Complex IV was found in PD with α-synuclein pathology and parkin mutations in aged substantia nigra neurons.24 This condition may cause impaired mitochondrial membrane potential, decreased ATP synthesis, and increased ROS release leading to oxidative stress in isolated mitochondria or intact cells.25 It may cause impaired mitochondrial membrane potential, decreased ATP synthesis, and increased ROS release leading to oxidative stress in aged substantia nigra neurons.25 Oxidative stress and mitochondrial dysfunction have primary roles in the pathogenesis of PD. In addition, mutations in PD-related genes such as the Parkinson’s disease protein 7 (PARK7) gene, parkin, and phosphatase and tensin homolog (PTEN)-induced kinase 1 (PINK1) are also known to be associated with dopaminergic neuronal loss.26 Studies have shown that mitochondrial respiration is ineffective, abnormal mitochondrial morphology occurs, and oxidative stress develops in mouse models with PINK1 deficiency.26
Huntington’s Disease
Huntington’s disease is an inherited disorder that causes the gradual degeneration of nerves. The protein called huntingtin (Htt) is thought to be essential for normal embryonic and neuronal development and is important in many cellular pathways, ion transport, vesicular trafficking, and production and transport of brain-derived neurotrophic factor.27 One of the important mechanisms involved in the pathogenesis of HD is mitochondrial dysfunction.28 Mitochondrial homeostasis and dynamics must be intact for continued ATP generation, calcium homeostasis, and antioxidant effects. Disturbances in this balance occur in the presence of mutant Htt protein. As a result, a decrease in ATP production, an increase in the level of reactive oxygen radicals, and a tendency to apoptosis are observed.28 Transport, fusion, and fission mechanisms play a role in the control of mitochondrial homeostasis. During fusion, 1 mitochondrion combines with another mitochondria and exchanges content. In this way, damaged mitochondria can have contents that will help to repair itself by fusion with normal mitochondria.28 On the contrary, in the event of fission, the mitochondrial content is released into the cytoplasm as a result of fragmentation, and in this case, factors that induce apoptosis such as Cyt c are revealed.29 Huntingtin protein, which is localized in the outer mitochondrial membrane, interacts with proteins such as Htt-interacting protein 1, dynamin, and clathrin under normal conditions, keeping the fusion–fission cycles—in other words, mitochondrial dynamics under control.30
Another important factor associated with mitochondrial dysfunction in HD is decreased expression of peroxisome proliferator-activated receptor γ co-activator-1 alpha (PGC-1α) protein, which is thought to be related to transcriptional dysregulation caused by mutant Htt, and impaired function.31
Amyotrophic Lateral Sclerosis
Amyotrophic lateral sclerosis is a progressive disease characterized by degeneration of the motor neurons.32 About 90%-95% of the disease are sporadic, while 3%-10% are familial. In familial cases, the age of onset of the disease is earlier.33 In 20% of the familial cases and 1%-5% of the sporadic cases, there is a mutation on the gene encoded by SOD1 enzyme.32 The motor neuron damage as a result of oxidative stress is the key hypothesis in ALS. There are many studies showing that oxidatively damaged metabolites are increased in the neuronal tissues of postmortem sporadic and familial ALS.34 Increased protein carbonyl levels were observed in the spinal cord and motor cortex. In the transgenic mutant SOD1 mouse model, mitochondrial dysfunction has been shown to occur at much earlier stages of ALS, before other manifestations of motor neuron damage. Mitochondrial abnormalities such as swelling and vacuolization have been observed in patients with ALS.35
Resveratrol
The natural phytocompound resveratrol (trans-3,5,4-trihydoxystilbene; C14H12O3) has come to the attention of neuroscientists for its neuroprotective effects. Resveratrol has antioxidant activity and prevents or delays cellular damage and diseases caused by oxidative stress.36 Resveratrol has many significant biological activities such as inhibition of lipid peroxidation, copper chelation, free radical scavenging, modulation of eicosanoid synthesis inhibition of platelet aggregation, vasodilator effect, modulation of lipid metabolism, anticancer effect, cardioprotective effect, and nervous system protective effect.37 Draczynska-Lusiak et al38 showed that antioxidants such as vitamin E or vitamin C and resveratrol defend neuronal cells from oxidative stress damage in vitro. Zini et al39 examined the potential consequences of resveratrol treatment on mitochondrial respiratory chain in rat brain. They showed that resveratrol decreased the activity of complex III in competition with coenzyme Q in the rat brain. With the decrease of Complex III activity, resveratrol reduces the production of ROS products and clears them.39 Resveratrol also decreases the production of reactive species by upregulating antioxidant enzymes in mitochondria.7
Resveratrol is an activator of sirtuin1 (SIRT1), known as NAD-dependent deacetylase sirtuin-1, a DNA repair protein which prolongs cell life span. The SIRT1 deacetylates histone and nonhistone proteins, including transcription factors. It helps in the regulation of immune functions and contributes to prolonging the life span.40 Resveratrol inhibits the nuclear factor κB signaling pathway thereby regulating the immune response to inflammation, infection, and cellular response to stimuli.41 In addition, it has been determined to significantly inhibit the insulin-like growth factor-1 receptor (IGF-1R)/serine/threonine kinase (Akt)/Wnt pathways.42
Resveratrol and Mitochondrial Biogenesis
Numerous clinical diseases, including cellular energy imbalance, oxidative stress, and endothelial dysfunction, are caused by impairment in mitochondrial biogenesis.43 Mitochondria are constantly being made and destroyed, and their density changes according to changes in energy requirements. Many endogenous and exogenous factors regulate mitochondrial biogenesis through PGC-1α. Resveratrol increases the activation of PGC-1α, and the expression level of PGC-1α is directly related to mitochondrial biogenesis. It has been shown that this pathway slows down the aging process, prevents many chronic diseases, and increases endurance in the muscles.44 Possible mechanisms of the neuroprotective effects of resveratrol are summarized in Figure 2.
Figure 2.
Possible mechanisms of the neuroprotective effects of resveratrol. Bax, Bcl-2 associated X protein; Bcl-2, B-cell lymphoma/leukemia-2; CAT, catalase; NF κB, nuclear factor κB; PGC1α, peroxisome proliferator-activated receptor γ co-activator-1 alpha; PI3K, phosphoinositide 3-kinase; ROS, reactive oxygen species; SIRT1, sirtuin1; SOD, superoxide dismutase.
Resveratrol’s Neuroprotective Effects Against Neurodegenerative Diseases
The neuroprotective effect of resveratrol supplementation was investigated at the neurobehavioral, neurochemical, and cerebrovascular levels. Studies have shown that resveratrol was effective in maintaining cognitive function.45 It has been stated that resveratrol may have beneficial effects for neurological disorders due to its antioxidant effect. 45
Recent studies have also demonstrated the immunomodulatory effects of resveratrol. Resveratrol effectively blocks gene expression of lipopolysaccharide-derived proinflammatory molecules in microglial cells.46 Resveratrol has been able to provide a neuroprotective effect by controlling neuroinflammation and enhancing adaptive immunity in people with AD.47 Antitumor activity of resveratrol has also been reported.48 Genetic and/or epigenetic changes cause cancer initiation and progression. Acetylation of histone and nonhistone proteins plays a crucial role in the epigenetic regulation of gene expression. The acetylation state of histones is controlled by the balance between the activity of histone acetyltransferases and histone deacetylases. An imbalance between the activity of these enzymes is associated with various types of cancer. Deacetylated histones promote genomic stability, while general hyperacetylation of histones can impair genomic stability and create a desirable environment for tumor cells. Resveratrol can activate SIRT1 which has deacetylase activity and thus exert antitumor activity.49 SIRT1 also exerts antiaging effects through histone deacetylation and neuroprotection.50 Some protective effects of resveratrol against atherosclerosis-associated endothelial dysfunction have also been demonstrated through the SIRT1, adiponectin, and calprotectin proteins.51 In summary, resveratrol may have a protective effect against neurodegenerative diseases due to its SIRT-1-mediated anti-inflammatory and neuroprotective effects.
The beneficial effects of resveratrol for AD have emerged as a result of reducing 𝛽 amyloid plaque as well as improving mitochondrial function, decreasing ROS activity and increasing neuronal cell survival by activating the SIRT1 pathway.47 A study investigated the effects of resveratrol in mild-to-moderate cases of AD. This phase 2 randomized, placebo-controlled trial showed that resveratrol was detectable in the cerebrospinal fluid (CSF) and stabilized the progressive decline of Aβ40 in CSF and plasma of AD patients compared to placebo-treated controls. It was demonstrated that resveratrol stabilized CSF Aβ42 levels.52
Resveratrol was also demonstrated to significantly reduce CSF metalloproteinase 9 and modulate neuro-inflammation in AD subjects compared to placebo-treated controls. It was concluded that SIRT 1 activation by resveratrol could be key for the treatment or prevention of neurodegenerative diseases.47 These results suggest that resveratrol can be used safely in Alzheimer’s patients. Resveratrol was demonstrated to modulate astroglial functions. The neuroprotective impact of resveratrol on neuroglia cells is hypothesized to occur subsequent to the discharge of glutamate at the synaptic gap.53 Resveratrol can enhance glutamate uptake by astrocytes, a course which promotes glutamine synthetase, an enzyme that converts glutamate into glutamine. Resveratrol can also promote the synthesis of glutathione.53 Although the molecular mechanisms underlying the neuroprotective effect of resveratrol have not been definitively determined, experiments suggest that it may be important that it selectively upregulates mitochondrial antioxidant enzyme gene expression, particularly manganese superoxide dismutase.43
It was observed that resveratrol modulates mRNA levels and protein expression of apoptotic regulators Bax and Bcl-2 genes and promotes cell survival in rat PC12 cells. In addition, the use of resveratrol has a curative effect on the dopaminergic neurons.54 Studies established that resveratrol could elicit neuroprotective effects on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-generated PD mice through free radical scavenging. Resveratrol treatment notably protected PD mice from MPTP-induced OH. production, motor coordination destruction, and neuronal death.55 Likewise, resveratrol decreased 6-hydroxydopamine-stimulated oxidative injury and dopamine reduction in a rat model of PD.56 In addition, resveratrol decreased dopamine-induced apoptosis in neuroblastoma cells.57 Studies also implicate that resveratrol may possibly minimize mitochondrial dysfunction induced by impairment of parkin function. Parkin is a protein in humans and mice that is encoded by the PARK2 gene.58 Parkin plays a critical role in ubiquitination, whereby it recognizes damaged proteins on the outer membrane of mitochondria and intervenes in the clearing out of injured mitochondria through proteasomal mechanisms.59 Parkin increases cell survival by repressing both mitochondria-reliant and mitochondria-independent apoptosis. Mutations in the PARK2 gene are connected with mitochondrial dysfunction, directing neuronal death in PD.60 A study which used fibroblast cultures commencing from 2 patients diagnosed with an early-onset form of PD with PARK2 heterozygous mutations showed that resveratrol caused a limited salvage of mitochondrial functions, possibly associated with the activation of the AMP-activated protein kinase/SIRT1/PGC-1α pathway.61
Resveratrol significantly improved motor and cognitive impairments by inhibiting cyclooxygenase-1 activity in the HD model.62 In addition, it protects against the cytotoxicity of mutant polyglutamine Htt through SIRT1 activation. It has been suggested that mutant polyglutamine Htt triggers striatal neurodegeneration by various mechanisms, such as mitochondrial dysfunction, oxidative stress, and apoptosis. Resveratrol protects cells against the toxic effects of mutant polyglutamine Htt by potentiating SIRT1 activity and induces indirect inhibition of p53.63 Sirtuin1 interacts with p53 and deacetylates it. The activity of deacetylated p53 is reduced and p53-dependent apoptosis is inhibited. p53 activation, occurring in HD, is associated with enhanced mitochondrial oxidation.64 Due to antioxidant properties, resveratrol can prevent impaired mitochondrial function through activation of the SIRT1–peroxisome proliferator-activated receptor γ co-activator-1 alpha (PGC1α) pathway. The PGC1α regulates the expression of antioxidant enzymes that inhibit ROS and consequently inhibits oxidative stress.65
Resveratrol has revealed a number of favorable effects that can slow down mitochondrial dysfunction associated with motor neuron damage observed in ALS. The antioxidant properties of resveratrol can reduce mitochondrial damage and ROS production; and in this context, Song et al66 showed suppression of ROS levels following resveratrol treatment in ALS mice. As mentioned earlier, PGC-1α activation mediated by the resveratrol-SIRT1 interaction protects against mitochondrial fragmentation.67 Zhao et al68 reported that resveratrol increased the expression of PGC-1α, improving motor performance and survival in a mouse model of ALS. Resveratrol-induced SIRT1 activation also increased neuronal survival in ALS cell models.69
Negative and Side Effects of Resveratrol
There are studies showing that resveratrol can act as a prooxidant agent and therefore pathological effects may also develop.70 Resveratrol can undergo auto-oxidation to semiquinones and to the more stable 4′-phenoxyl radical causing ROS formation.71 High doses of resveratrol induce apoptosis, while low doses induce cell proliferation.72 Acting as a prooxidant, resveratrol can reversibly or irreversibly interrupt the cell cycle and also cause DNA damage.73 Resveratrol is considered to be a phytoestrogen due to its structural resemblance to diethylstilbestrol and thus can act as an estrogen receptor agonist.73 Studies underline the importance of dose dependence in resveratrol-induced cellular responses.74
No significant adverse effects of resveratrol have been reported in long-term clinical trials in healthy populations.75 Up to 5 g daily has been reported to be well tolerated in healthy populations.76 Orally administered resveratrol is metabolized by the gut microbiota77, so observed effects may result not only from resveratrol but also from resveratrol metabolites. Studies to date have not found definite doses and administration intervals for resveratrol. An important drawback in the clinical use of resveratrol is its bioavailability. Although demonstrated safe, resveratrol does not have the best possible pharmacokinetic and pharmacodynamic properties.78
Conclusion
In studies on neurodegenerative diseases, the importance of mitochondrial dysfunction and oxidative stress has been frequently stated. Resveratrol, an antioxidant, is an important phytoalexin found in abundance in grapes and its products. It modulates mitochondrial function and dynamics with its cytoprotective, anti-inflammatory, and antioxidant effects. Identifying and targeting redox-regulated pathways in neurodegenerative diseases may provide a pathway for new treatment strategies. In this review, the literature on the effect of resveratrol on neurodegenerative diseases and mitochondrial dysfunction was reviewed. As a result, resveratrol has been shown to have a curative role on both cell and animal models of neurodegenerative diseases as a modulator of mitochondrial function, redox biology and dynamics. Despite the considerable advancement in the knowledge of resveratrol’s effects on the brain, our understanding in this area is still in progress. The greater part of the research has been carried out in cell culture or in experimental animal models. Further studies are necessary to comprehend the effect of resveratrol on mitochondrial biogenesis and function. It is imperative that the acquired knowledge is transferred into actual treatments for neurodegenerative diseases.
Funding Statement
The authors declared that this study has received no financial support.
Footnotes
Peer-review: Externally peer-reviewed.
Author Contributions: Concept – B.D., M.A.; Design – B.D., S.E.K., M.A.; Supervision – S.E.K., M.A.; Resources – B.D., M.A.; Materials – B.D., S.E.K.; Data Collection and/or Processing – B.D., M.A.; Analysis and/or Interpretation – M.A., S.E.K.; Literature Search –B.D., M.A.; Writing – B.D., M.A.; Critical Review – S.E.K., M.A.
Declaration of Interests: The authors have no conflict of interest to declare.
References
- 1. Peng C, Trojanowski JQ, Lee VMY. Protein transmission in neurodegenerative disease. Nat Rev Neurol. 2020;16(4):199 212. ( 10.1038/s41582-020-0333-7) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Reddy PH. Mitochondrial medicine for aging and neurodegenerative diseases. NeuroMolecular Med. 2008;10(4):291 315. ( 10.1007/s12017-008-8044-z) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Singh A, Kukreti R, Saso L, Kukreti S. Oxidative stress: A key modulator in neurodegenerative diseases. Molecules. 2019;24(8):1583. ( 10.3390/molecules24081583) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Yaribeygi H, Panahi Y, Javadi B, Sahebkar A. The underlying role of oxidative stress in neurodegeneration: A mechanistic review. CNS Neurol Disord Drug Targets. 2018;17(3):207 215. ( 10.2174/1871527317666180425122557) [DOI] [PubMed] [Google Scholar]
- 5. Sun AY, Wang Q, Simonyi A, Sun GY. Botanical phenolics and brain health. NeuroMolecular Med. 2008;10(4):259 274. ( 10.1007/s12017-008-8052-z) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. de Oliveira MR, Nabavi SF, Manayi A, Daglia M, Hajheydari Z, Nabavi SM. Resveratrol and the mitochondria: From triggering the intrinsic apoptotic pathway to inducing mitochondrial biogenesis, a mechanistic view. Biochim Biophys Acta. 2016;1860(4):727 745. ( 10.1016/j.bbagen.2016.01.017) [DOI] [PubMed] [Google Scholar]
- 7. Gibellini L, Bianchini E, De Biasi S, Nasi M, Cossarizza A, Pinti M. Natural compounds modulating mitochondrial functions. Evid Based Complement Alternat Med. 2015;2015:527209. ( 10.1155/2015/527209) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Jardim FR, de Rossi FT, Nascimento MX, et al. Resveratrol and brain mitochondria: A review. Mol Neurobiol. 2018;55(3):2085 2101. ( 10.1007/s12035-017-0448-z) [DOI] [PubMed] [Google Scholar]
- 9. Duchen MR. Mitochondria in health and disease: Perspectives on a new mitochondrial biology. Mol Aspects Med. 2004;25(4):365 451. ( 10.1016/j.mam.2004.03.001) [DOI] [PubMed] [Google Scholar]
- 10. Kohen R, Nyska A. Oxidation of biological systems: Oxidative stress phenomena, antioxidants, redox reactions, and methods for their quantification. Toxicol Pathol. 2002;30(6):620 650. ( 10.1080/01926230290166724) [DOI] [PubMed] [Google Scholar]
- 11. Poljsak B, Šuput D, Milisav I. Achieving the balance between ROS and antioxidants: When to use the synthetic antioxidants. Oxid Med Cell Longev. 2013;2013:956792. ( 10.1155/2013/956792) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Dröse S, Brandt U. Molecular mechanisms of superoxide production by the mitochondrial respiratory chain. Adv Exp Med Biol. 2012;748:145 169. ( 10.1007/978-1-4614-3573-0_6) [DOI] [PubMed] [Google Scholar]
- 13. Rigoulet M, Yoboue ED, Devin A. Mitochondrial ROS generation and its regulation: Mechanisms involved in H2O2 signaling. Antioxid Redox Signal. 2011;14(3):459 468. ( 10.1089/ars.2010.3363) [DOI] [PubMed] [Google Scholar]
- 14. Levine AB, Punihaole D, Levine TB. Characterization of the role of nitric oxide and its clinical applications. Cardiology. 2012;122(1):55 68. ( 10.1159/000338150) [DOI] [PubMed] [Google Scholar]
- 15. Cuyvers E, Sleegers K. Genetic variations underlying Alzheimer's disease: Evidence from genome-wide association studies and beyond. Lancet Neurol. 2016;15(8):857 868. ( 10.1016/S1474-4422(16)00127-7) [DOI] [PubMed] [Google Scholar]
- 16. Walsh DM, Selkoe DJ. Aβ oligomers–a decade of discovery. J Neurochem. 2007;101(5):1172 1184. ( 10.1111/j.1471-4159.2006.04426.x) [DOI] [PubMed] [Google Scholar]
- 17. Rajmohan R, Reddy PH. Amyloid-beta and phosphorylated tau accumulations cause abnormalities at synapses of Alzheimer’s disease neurons. J Alzheimers Dis. 2017;57(4):975 999. ( 10.3233/JAD-160612) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Devi L, Prabhu BM, Galati DF, Avadhani NG, Anandatheerthavarada HK. Accumulation of amyloid precursor protein in the mitochondrial import channels of human Alzheimer's disease brain is associated with mitochondrial dysfunction. J Neurosci. 2006;26(35):9057 9068. ( 10.1523/JNEUROSCI.1469-06.2006) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Zhu X, Perry G, Smith MA, Wang X. Abnormal mitochondrial dynamics in the pathogenesis of Alzheimer's disease. J Alzheimers Dis. 2013;33(S 1):S253 S262. ( 10.3233/JAD-2012-129005) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Cho DH, Nakamura T, Lipton SA. Mitochondrial dynamics in cell death and neurodegeneration. Cell Mol Life Sci. 2010;67(20):3435 3447. ( 10.1007/s00018-010-0435-2) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Tillement L, Lecanu L, Papadopoulos V. Alzheimer's disease: Effects of β-amyloid on mitochondria. Mitochondrion. 2011;11(1):13 21. ( 10.1016/j.mito.2010.08.009) [DOI] [PubMed] [Google Scholar]
- 22. Foffani G, Obeso JA. A cortical pathogenic theory of Parkinson’s disease. Neuron. 2018;99(6):1116 1128. ( 10.1016/j.neuron.2018.07.028) [DOI] [PubMed] [Google Scholar]
- 23. Chen C, Turnbull DM, Reeve AK. Mitochondrial dysfunction in Parkinson’s disease—cause or consequence? Biology. 2019;8(2):38. ( 10.3390/biology8020038) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Reeve AK, Park TK, Jaros E, et al. Relationship between mitochondria and α-synuclein: A study of single substantia nigra neurons. Arch Neurol. 2012;69(3):385 393. ( 10.1001/archneurol.2011.2675) [DOI] [PubMed] [Google Scholar]
- 25. Suski JM, Lebiedzinska M, Bonora M, Pinton P, Duszynski J, Wieckowski MR. Relation between mitochondrial membrane potential and ROS formation. Methods Mol Biol. 2012;810:183 205. ( 10.1007/978-1-61779-382-0_12) [DOI] [PubMed] [Google Scholar]
- 26. Belin AC, Westerlund M. Parkinson’s disease: A genetic perspective. FEBS Journal. 2008;275(7):1377 1383. ( 10.1111/j.1742-4658.2008.06301.x) [DOI] [PubMed] [Google Scholar]
- 27. Jamwal S, Kumar P. Antidepressants for neuroprotection in Huntington's disease: A review. Eur J Pharmacol. 2015;769:33 42. ( 10.1016/j.ejphar.2015.10.033) [DOI] [PubMed] [Google Scholar]
- 28. Jodeiri Farshbaf M, Ghaedi K. Huntington’s disease and mitochondria. Neurotox Res. 2017;32(3):518 529. ( 10.1007/s12640-017-9766-1) [DOI] [PubMed] [Google Scholar]
- 29. Estaquier J, Arnoult D. Inhibiting Drp1-mediated mitochondrial fission selectively prevents the release of cytochrome c during apoptosis. Cell Death Differ. 2007;14(6):1086 1094. ( 10.1038/sj.cdd.4402107) [DOI] [PubMed] [Google Scholar]
- 30. Tilokani L, Nagashima S, Paupe V, Prudent J. Mitochondrial dynamics: Overview of molecular mechanisms. Essays Biochem. 2018;62(3):341 360. ( 10.1042/EBC20170104) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Weydt P, Pineda VV, Torrence AE, et al. Thermoregulatory and metabolic defects in Huntington's disease transgenic mice implicate PGC-1α in Huntington's disease neurodegeneration. Cell Metab. 2006;4(5):349 362. ( 10.1016/j.cmet.2006.10.004) [DOI] [PubMed] [Google Scholar]
- 32. Muyderman H, Chen T. Mitochondrial dysfunction in amyotrophic lateral sclerosis–a valid pharmacological target? Br J Pharmacol. 2014;171(8):2191 2205. ( 10.1111/bph.12476) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Bozzoni V, Pansarasa O, Diamanti L, Nosari G, Cereda C, Ceroni M. Amyotrophic lateral sclerosis and environmental factors. Funct Neurol. 2016;31(1):7-19. ( 10.11138/fneur/2016.31.1.007) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Simpson EP, Henry YK, Henkel JS, Smith RG, Appel SH. Increased lipid peroxidation in sera of ALS patients: A potential biomarker of disease burden. Neurology. 2004;62(10):1758 1765. ( 10.1212/wnl.62.10.1758) [DOI] [PubMed] [Google Scholar]
- 35. Liu J, Lillo C, Jonsson PA, et al. Toxicity of familial ALS-linked SOD1 mutants from selective recruitment to spinal mitochondria. Neuron. 2004;43(1):5 17. ( 10.1016/j.neuron.2004.06.016) [DOI] [PubMed] [Google Scholar]
- 36. King RE, Bomser JA, Min DB. Bioactivity of resveratrol. Compr Rev Food Sci Food Saf. 2006;5(3):65 70. ( 10.1111/j.1541-4337.2006.00001.x) [DOI] [Google Scholar]
- 37. Belguendouz L, Fremont L, Linard A. Resveratrol inhibits metal ion-dependent and independent peroxidation of porcine low-density lipoproteins. Biochem Pharmacol. 1997;53(9):1347 1355. ( 10.1016/s0006-2952(96)00820-9) [DOI] [PubMed] [Google Scholar]
- 38. Draczynska-Lusiak B, Doung A, Sun AY. Oxidized lipoproteins may play a role in neuronal cell death in Alzheimer disease. Mol Chem Neuropathol. 1998;33(2):139 148. ( 10.1007/BF02870187) [DOI] [PubMed] [Google Scholar]
- 39. Zini R, Morin C, Bertelli A, Bertelli AA, Tillement JP. Effects of resveratrol on the rat brain respiratory chain. Drugs Exp Clin Res. 1999;25(2-3):87 97. [PubMed] [Google Scholar]
- 40. Chung S, Yao H, Caito S, Hwang JW, Arunachalam G, Rahman I. Regulation of SIRT1 in cellular functions: Role of polyphenols. Arch Biochem Biophys. 2010;501(1):79 90. ( 10.1016/j.abb.2010.05.003) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Ren Z, Wang L, Cui J, et al. Resveratrol inhibits NF-κB signaling through suppression of p65 and IkappaB kinase activities. Pharmazie. 2013;68(8):689 694. [PubMed] [Google Scholar]
- 42. Zhang LX, Li CX, Kakar MU, et al. Resveratrol (RV): A pharmacological review and call for further research. Biomed Pharmacother. 2021;143:112164. ( 10.1016/j.biopha.2021.112164) [DOI] [PubMed] [Google Scholar]
- 43. Csiszar A, Labinskyy N, Pinto JT, et al. Resveratrol induces mitochondrial biogenesis in endothelial cells. Am J Physiol Heart Circ Physiol. 2009;297(1):H13 H20. ( 10.1152/ajpheart.00368.2009) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Smoliga JM, Baur JA, Hausenblas HA. Resveratrol and health–a comprehensive review of human clinical trials. Mol Nutr Food Res. 2011;55(8):1129 1141. ( 10.1002/mnfr.201100143) [DOI] [PubMed] [Google Scholar]
- 45. Andrade S, Ramalho MJ, Pereira MDC, Loureiro JA. Resveratrol brain delivery for neurological disorders prevention and treatment. Front Pharmacol. 2018;9:1261. ( 10.3389/fphar.2018.01261) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Wiedemann J, Rashid K, Langmann T. Resveratrol induces dynamic changes to the microglia transcriptome, inhibiting inflammatory pathways and protecting against microglia-mediated photoreceptor apoptosis. Biochem Biophys Res Commun. 2018;501(1):239 245. ( 10.1016/j.bbrc.2018.04.223) [DOI] [PubMed] [Google Scholar]
- 47. Moussa C, Hebron M, Huang X, et al. Resveratrol regulates neuro-inflammation and induces adaptive immunity in Alzheimer’s disease. J Neuroinflammation. 2017;14(1):1. ( 10.1186/s12974-016-0779-0) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Aggarwal BB, Bhardwaj A, Aggarwal RS, Seeram NP, Shishodia S, Takada Y. Role of resveratrol in prevention and therapy of cancer: Preclinical and clinical studies. Anticancer Res. 2004;24(5A):2783 2840. [PubMed] [Google Scholar]
- 49. Farhan M, Ullah MF, Faisal M, et al. Differential methylation and acetylation as the epigenetic basis of resveratrol's anticancer activity. Medicines (Basel) 2019;6(1):24. ( 10.3390/medicines6010024) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Pallàs M, Casadesús G, Smith MA, et al. Resveratrol and neurodegenerative diseases: Activation of SIRT1 as the potential pathway towards neuroprotection. Curr Neurovasc Res. 2009;6(1):70 81. ( 10.2174/156720209787466019) [DOI] [PubMed] [Google Scholar]
- 51. Wiciński M, Malinowski B, Węclewicz MM, Grześk E, Grześk G. Anti-atherogenic properties of resveratrol: 4-week resveratrol administration associated with serum concentrations of SIRT1, adiponectin, S100A8/A9 and VSMCs contractility in a rat model. Exp Ther Med. 2017;13(5):2071 2078. ( 10.3892/etm.2017.4180) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Turner RS, Thomas RG, Craft S, et al. A randomized, double-blind, placebo-controlled trial of resveratrol for Alzheimer disease. Neurology. 2015;85(16):1383 1391. ( 10.1212/WNL.0000000000002035) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Quincozes-Santos A, Gottfried C. Resveratrol modulates astroglial functions: Neuroprotective hypothesis. Ann N Y Acad Sci. 2011;1215(1):72 78. ( 10.1111/j.1749-6632.2010.05857.x) [DOI] [PubMed] [Google Scholar]
- 54. Bournival J, Quessy P, Martinoli MG. Protective effects of resveratrol and quercetin against MPP+-induced oxidative stress act by modulating markers of apoptotic death in dopaminergic neurons. Cell Mol Neurobiol. 2009;29(8):1169 1180. ( 10.1007/s10571-009-9411-5) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Lu KT, Ko MC, Chen BY, et al. Neuroprotective effects of resveratrol on MPTP-induced neuron loss mediated by free radical scavenging. J Agric Food Chem. 2008;56(16):6910 6913. ( 10.1021/jf8007212) [DOI] [PubMed] [Google Scholar]
- 56. Khan MM, Ahmad A, Ishrat T, et al. Resveratrol attenuates 6-hydroxydopamine-induced oxidative damage and dopamine depletion in rat model of Parkinson's disease. Brain Res. 2010;1328:139 151. ( 10.1016/j.brainres.2010.02.031) [DOI] [PubMed] [Google Scholar]
- 57. Lee MK, Kang SJ, Poncz M, Song KJ, Park KS. Resveratrol protects SH-SY5Y neuroblastoma cells from apoptosis induced by dopamine. Exp Mol Med. 2007;39(3):376 384. ( 10.1038/emm.2007.42) [DOI] [PubMed] [Google Scholar]
- 58. Kitada T, Asakawa S, Hattori N, et al. Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism. Nature. 1998;392(6676):605 608. ( 10.1038/33416) [DOI] [PubMed] [Google Scholar]
- 59. Seirafi M, Kozlov G, Gehring K. Parkin structure and function. FEBS Journal. 2015;282(11):2076 2088. ( 10.1111/febs.13249) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Dawson TM, Dawson VL. The role of parkin in familial and sporadic Parkinson's disease. Mov Disord. 2010;25:S32 S39. ( 10.1002/mds.22798) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Ferretta A, Gaballo A, Tanzarella P, et al. Effect of resveratrol on mitochondrial function: Implications in parkin-associated familiar Parkinson's disease. Biochim Biophys Acta. 2014;1842(7):902 915. ( 10.1016/j.bbadis.2014.02.010) [DOI] [PubMed] [Google Scholar]
- 62. Kumar P, Padi SS, Naidu PS, Kumar A. Cyclooxygenase inhibition attenuates 3‐nitropropionic acid‐induced neurotoxicity in rats: Possible antioxidant mechanisms. Fundam Clin Pharmacol. 2007;21(3):297 306. ( 10.1111/j.1472-8206.2007.00485.x) [DOI] [PubMed] [Google Scholar]
- 63. Parker JA, Arango M, Abderrahmane SE, et al. Resveratrol rescues mutant polyglutamine cytotoxicity in nematode and mammalian neurons. Nat Genet. 2005;37(4):349 350. ( 10.1038/ng1534) [DOI] [PubMed] [Google Scholar]
- 64. Zhou S, Kachhap S, Singh KK. Mitochondrial impairment in p53-deficient human cancer cells. Mutagenesis. 2003;18(3):287 292. ( 10.1093/mutage/18.3.287) [DOI] [PubMed] [Google Scholar]
- 65. Wang N, Luo Z, Jin M, et al. Exploration of age-related mitochondrial dysfunction and the anti-aging effects of resveratrol in zebrafish retina. Aging (Albany NY) 2019;11(10):3117-3137. ( 10.18632/aging.101966) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Song L, Chen L, Zhang X, Li J, Le W. Resveratrol ameliorates motor neuron degeneration and improves survival in SOD1(G93A) mouse model of amyotrophic lateral sclerosis. BioMed Res Int. 2014;2014:483501. ( 10.1155/2014/483501) [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 67. Higashida K, Kim SH, Jung SR, Asaka M, Holloszy JO, Han DH. Effects of resveratrol and SIRT1 on PGC-1α activity and mitochondrial biogenesis: A reevaluation. PLOS Biol. 2013;11(7):e1001603. ( 10.1371/journal.pbio.1001603) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Zhao W, Varghese M, Yemul S, et al. Peroxisome proliferator activator receptor gamma coactivator-1alpha (PGC-1α) improves motor performance and survival in a mouse model of amyotrophic lateral sclerosis. Mol Neurodegener. 2011;6(1):51. ( 10.1186/1750-1326-6-51) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Kim D, Nguyen MD, Dobbin MM, et al. SIRT1 deacetylase protects against neurodegeneration in models for Alzheimer's disease and amyotrophic lateral sclerosis. EMBO J. 2007;26(13):3169 3179. ( 10.1038/sj.emboj.7601758) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. de la Lastra CA, Villegas I. Resveratrol as an antioxidant and pro-oxidant agent: Mechanisms and clinical implications. Biochem Soc Trans. 2007;35(5):1156 1160. ( 10.1042/BST0351156) [DOI] [PubMed] [Google Scholar]
- 71. Li DD, Han RM, Liang R, et al. Hydroxyl radical reaction with trans-resveratrol: Initial carbon radical adduct formation followed by rearrangement to phenoxyl radical. J Phys Chem B. 2012;116(24):7154 7161. ( 10.1021/jp3033337) [DOI] [PubMed] [Google Scholar]
- 72. Rüweler M, Gülden M, Maser E, Murias M, Seibert H. Cytotoxic, cytoprotective and antioxidant activities of resveratrol and analogues in C6 astroglioma cells in vitro. Chem Biol Interact. 2009;182(2-3):128 135. ( 10.1016/j.cbi.2009.09.003) [DOI] [PubMed] [Google Scholar]
- 73. Gehm BD, McAndrews JM, Chien PY, Jameson JL. Resveratrol, a polyphenolic compound found in grapes and wine, is an agonist for the estrogen receptor. Proc Natl Acad Sci U S A. 1997;94(25):14138 14143. ( 10.1073/pnas.94.25.14138) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Mukherjee S, Dudley JI, Das DK. Dose-dependency of resveratrol in providing health benefits. Dose Response. 2010;8(4):478 500. ( 10.2203/dose-response.09-015.Mukherjee) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Brown VA, Patel KR, Viskaduraki M, et al. Repeat dose study of the cancer chemopreventive agent resveratrol in healthy volunteers: Safety, pharmacokinetics, and effect on the insulin-like growth factor axis. Cancer Res. 2010;70(22):9003 9011. ( 10.1158/0008-5472.CAN-10-2364) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Patel KR, Scott E, Brown VA, Gescher AJ, Steward WP, Brown K. Clinical trials of resveratrol. Ann N Y Acad Sci. 2011;1215:161 169. ( 10.1111/j.1749-6632.2010.05853.x) [DOI] [PubMed] [Google Scholar]
- 77. Bode LM, Bunzel D, Huch M, et al. In vivo and in vitro metabolism of trans-resveratrol by human gut microbiota. Am J Clin Nutr. 2013;97(2):295 309. ( 10.3945/ajcn.112.049379) [DOI] [PubMed] [Google Scholar]
- 78. Calvo-Castro LA, Schiborr C, David F, et al. 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(9):e1701057. ( 10.1002/mnfr.201701057) [DOI] [PubMed] [Google Scholar]

Content of this journal is licensed under a 
