Skip to main content
. 2020 Mar 14;9(3):340. doi: 10.3390/foods9030340

Table 2.

Bioactivities and potential mechanisms of resveratrol from experimental studies.

Study Type Subject Dose Main Findings Ref.
Antioxidative activities
In vitro HepG2 cells 0–100 μM Dose-dependently increasing antioxidant effects by enhancing SIRT2’s activity to deacetylate Prx1 [24]
In vitro HepG2,
C2C12, and HEK293 cells
10, 25 μM Activating AMPK to maintain the structural stability of FoxO1 [25]
In vitro MCF-7 cells 1 nM, 0.02 μM, 0.1 μM, 0.5 μM, 1.5 μM Upregulating PTEN (except at the highest dose, 1.5 μM), which decreased Akt phosphorylation, leading to an upregulation of antioxidant enzyme mRNA levels such as CAT and SOD [26]
In vivo Rats 20 mg kg/b.w./day Improving the antioxidant defense system by modulating antioxidant enzymes through downregulation of ERK activated by ROS [27]
In vivo Rats 10 mg/kg b.w. Reducing the ischemia-reperfusion injury-induced oxidative stress by inhibiting the activation of p38 MAPK pathway to increase antioxidants like GSH and scavenge free radicals [28]
In vivo Rats 5, 10 mg/kg Activating SIRT1 to scavenge ROS [29]
In vivo Mice 15, 30, 60 mg/kg Activating AMPK, SIRT1, and Nrf2 associated antioxidant defense pathways to improve systemic oxidative and nitrosative stress [30]
In vivo Sows 300 mg/kg Regulating antioxidant gene expression via Keap1/Nrf2 pathway and SIRT1 [31]
In vitro HUVECs 10 μM Inducing autophagy via the activation of TFEB [32]
In vitro HEK293 cells or HEK293T 5 μg/mL Inducing autophagy via the AMPK-mediated inhibition of mTOR signaling [33]
Anti-inflammatory activities
In vivo Mice 8 mg/kg/day Inhibiting the activation of NALP3 inflammasome and inducing autophagy via SIRT1 upregulation [34]
In vitro J774 mouse macrophages,
Mouse bone-marrow cells
0.5–100 μM Inhibiting the activation of NALP3 inflammasome [35]
In vitro;
In vivo
BEAS-2B cells,
Mice
25 μM,
20 mg/kg
Inducing NF-κB inhibition, decreasing IL-6 secretion, suppressing STAT3 activation, blocking ERK1/2 activation, and upregulating MyD88 Short [36]
In vitro RAW264.7 macrophages 0–20 μM Inhibiting the production of pro-inflammatory cytokines, such as TNF-α and IL-1β, but also by inducing anti-inflammatory HO-1 [38]
In vitro RAW264.7 macrophages, MCF-7 cells 10 μM Suppressing IL-6 transcription, modulating the inflammatory responses as an ERα ligand mediated by SIRT1. [39]
In vitro Mouse C2C12 myoblasts 20, 50, 100 μM Inhibiting NF-κB signaling independent of SIRT1 [40]
In vitro RAW264.7 macrophages 1, 5, 10, 20, 40 μM Downregulating HMGB1 as well as suppressing NF-κB and JAK/STAT signaling pathways [41]
In vitro U937 monocytic cells 15, 30, 50 μM Inhibiting NF-κB and JAK/STAT signaling pathways [42]
In vitro
In vivo
NRK-52E,
Rat
100 μmol/mL,
0.23 μg/kg
Inhibiting TLR4/NF-κB signaling cascade [43]
In vivo Rats 30, 10 and 3 mg/kg, Inhibiting TLR4/NF-κBp65/MAPKs signaling cascade [44]
In vitro Primary chondrocytes and macrophages 10, 25, 50, 100 μM Interrupting an inflammatory amplification loop [45]
Immunomodulating effects
In vitro A549 cells 56.25, 112.5 μg/mL Triggering an immune response to protect against non-typeable Haemophilus influenzae without developing resistance [46]
In vitro H1HeLa cells, Human nasal epithelia 0–300 μM Inhibiting human rhinoviruses-16 replication and normalized virus-induced IL-6, IL-8, and RANTES as well as the expression of ICAM-1 [47]
In vitro Rhabdosarcoma cells 2.5–100 μg/mL Preventing EV71 replication, reducing the virus-induced elevated IL-6 and TNF-α secretion via suppressing IKK/NF-κB signaling pathway [48]
In vivo Chickens 200, 400, 800 mg/kg Reducing immunocyte apoptosis in chickens receiving conventional vaccinations, and improving the growth of young chickens [49]
In vivo Piglets 3, 10, 30 mg/kg/d Maintaining the immune function and attenuating diarrhea and inflammation [51]
In vitro Atlantic salmon macrophages 10, 30, 50 μM Reducing bacterial and inflammatory biomarkers in LPS-challenged primary Atlantic salmon macrophages [52]
In vivo Mice 30 mg/kg Upregulating SIRT1 and reducing cytokines such as TNF-α, IFN-γ, IL-6, and MCP-1 [53]
In vivo Mice 30 mg/kg Enhancing immune activity in immunosuppressive mice, showing a bidirectional regulatory effect on immunity [54]
In vitro Human CD4+ T cells 10, 30, or 50 μM Suppressing the AhR pathway, resulting in the reversal of imbalanced Th17/Treg [56]
Cardiovascular diseases
In vivo Rhesus monkeys 80 mg/day (1st year), 480 mg/day (2nd year) Improving central arterial wall stiffening based on its antioxidative and anti-inflammation [7]
In vivo Rabbits 2.5 mg/kg Mitigating atrial fibrillation by upregulating PI3K/AKT/eNOS [8]
In vitro Peripheral blood mononuclear
cells
3–80 μM Blocking atherosclerotic plaque progression by acting against pro-atherogenic oxysterol signaling in M1 and M2 macrophages [57]
In vitro
In vivo
THP-1 monocytes,
Mice
0, 25, 50, 100 μM (dose-dependent),
10 mg/kg/day
Ameliorating atherosclerosis partially through restoring intracellular GSH via AMPK-α activation, inhibiting monocyte differentiation, and reducing pro-inflammatory cytokine production [59]
In vivo Rats 50 mg/L Preventing the pathological progression of hypertension through Nrf2 activation [60]
In vitro;
In vivo
Rat aortic smooth muscle cells;
Mice
100 μmol/L,
~320 mg/kg
Lowering blood pressure by inducing oxidative activation of cGMP-dependent PKG1α [61]
In vivo Rats 50 mg/kg/day Preventing the activation of inflammasome via downregulating NF-κB p65 and p38 MAPK expression, and upregulating SIRT1 expression [62]
In vivo Mice 20 mg/kg Regulated the FERM-kinase and Nrf2 interaction, decreasing the expression of ICAM-1, and inhibiting monocyte adhesion [63]
In vivo Rats 1.24 μg/d Improving the cardiac and vascular autonomic function [65]
In vitro Human RBCs 100 μM Protecting the erythrocytes via interacting with hemoglobin and reducing heme-iron oxidation [66]
Cancers
In vitro LNCaP cells 5, 10, 20, 50 μM Inducing the expression of COX-2, promoting ERK1/2 activation, and facilitating p53-dependent anti-proliferation gene expression [14]
In vitro;
In vivo
tBregs;
Mice
12.5 μM;
20, 50, 500 μg/mouse
Preventing breast cancer metastasis by promoting antitumor immune responses via blunting STAT3, leading to inhibited generation and function of tBregs as well as decreased production of TGF-β [67]
In vivo Mice 150, 300 ppm Inhibiting the formation and growth of colorectal cancer by downregulating oncogenic KRAS expression [68]
In vitro;
In vivo
NSCLC cells
Mice
25, 50, 100 μM,
30 mg/kg every 3 days
Preventing tumorigenesis and progression by interrupting glycolysis via inhibition of hexokinase II expression, which was mediated by downregulation of EGFR/Akt/ERK1/2 signaling pathway [69]
In vitro MCF-7 cells
MVLN cells
Low: 0.1 and 1 μM; High: 10 and 25 μM; Low concentrations: Increasing the growth of ERα+ cells
High concentrations: Inhibiting the proliferation of eERα+ breast cancer
[75]
In vitro KPL-1, MCF-7, MKL-F cells Low (KPL-1, ≤22 μM; MCF-7, ≤4 μM); High: ≥44 μM Low concentrations: Causing cell proliferation ER+ cells
High concentrations: Suppressing cell growth
[76]
In vitro
In vivo
Apc10.1 cells;
Mice;
Humans
0.001–1 μM;
0.7, 14.3 mg/kg diet;
5 mg, 1 g
Lower doses of resveratrol: Showing superior efficacy than high doses due to the pro-oxidant activity and AMPK signaling upregulation [79]
In vitro A2780, OVCAR-3, SKOV-3 cells 10, 50, 100 μM Decreasing the efficiency of ovarian cancer cells adhering to peritoneal mesothelium by downregulating the production of α5β1 integrins and upregulating the release of soluble hyaluronic acid [70]
In vitro Hela cells 0.1, 1, 10 μM,
10, 20, 50, 100 μM
Inhibiting the expression of PLSCR1, leading to the growth inhibition of HeLa cells [71]
In vitro HepG2 cells 25, 50, 100, 200 μM Inhibiting proliferation and inducing apoptosis by activating caspase-3 and caspase-9, upregulating the Bax/Bcl-2 ratio, and inducing p53 expression [72]
In vitro SGC7901 and BGC823 cells 5, 10, 25, 50, 100, 200, and 400 μM Inhibiting the invasion and migration of human gastric cancer cells by blocking the MALAT1-mediated epithelial-to-mesenchymal transition [73]
Liver diseases
In vivo Mice 0.2% of diet Improving HFD-induced fatty liver by downregulating adipose differentiation-related proteins and increasing the numbers of CD68+ Kupffer cells [9]
In vivo Rats 10 mg/kg Attenuating hepatic fibrosis by restoring the architecture and normalizing collagen deposition, mainly due to its antioxidative activities and downregulation of α-SMA [80]
In vivo Rats 50, 100 mg/kg Alleviating NAFLD by upregulating LDLR and SRB1 gene expressions [83]
In vivo Rats 250 mg/kg/day Downregulating HIF-1α expression and mitochondrial ROS production [85]
In vitro;
In vivo
HepG2 cells;
Mice
45 μmol
10, 30, 100 mg/kg
Restoring the morphology and function of alcohol-injured liver through inducing autophagy [86]
In vivo Rats 10 mg/kg Mitigating liver cirrhosis by improving the homing of bone marrow-derived mesenchymal stem cells [87]
Diabetes
In vivo Rats 20 mg/kg Increasing insulin action and glucose utilization due to visfatin expression restoration, SIRT1 activation, and glucose transporter modulation [89]
In vivo Mice 50 mg/kg Increasing glucose uptake to improve insulin resistance in the muscle by decreasing DAG accumulation and PKC-θ translocation, and preventing lipolysis under the condition of adipose hypoxia [90]
In vivo Rats 147.6 mg/kg/day Preventing the offspring’s glucose intolerance and islet dysfunction [91]
In vivo Mice 0.3% diet Reducing blood glucose levels, plasma lipids, and free fatty acids, inhibiting the expression of inflammatory mediators both in the aorta and in the blood, by inhibiting the NF-κB pathway [92]
In vivo Mice 50 mg/kg Preventing ROS-mediated mitochondrial fission via AMPK-dependent upregulation of Drp1 phosphorylation, and blocking the activation of NALP3 inflammasome via inhibition of ERS [93]
Obesity
In vivo Zebrafish 40 mg/kg/day Inhibiting transcriptional regulators such as EP300 [95]
In vivo Mice 0.06% diet Decreasing the body weight and fat mass, reducing leptin and lipids in plasma, modulating metabolism of glucose and insulin, and restoring immune dysfunction by activating PI3K/SIRT1 and Nrf2 signaling pathway [96]
In vitro;
In vivo
3T3-L1 cells;
Mice
0.03 to 100 μM;
1, 10, 30 mg/kg
In vitro: low concentrations of resveratrol (1-10 μM) suppressed adipogenic differentiation in pre-adipocytes, downregulated the expression of PPAR-γ and perilipin protein in differentiated adipocytes, and inhibiting TNF-α-induced lipolysis in mature adipocytes
In vivo: Dose-dependently decreasing weight gain and lipid deposition in the liver and adipose tissue
[97]
In vitro RAW 264.7 macrophage cells 25 μM Enhancing the catecholamine production, accompanying by suppressing the pro-inflammatory M1 macrophages, and activating anti-inflammatory M2 macrophages in white adipose tissue [98]
In vivo Mice 0.2% diet Promoting white adipose browning and thermogenesis in the male descendants, and these health benefits persisted and prevented obesity in their future life [99]
In vitro;
In vivo
L6 myogenic cell line;
Rats
1, 5, 10, 25 or 50 μM;
0.4% diet
In vitro: Improving mitochondrial function and reducing oxidative stress through the PKA/LKB1/AMPK pathway;
In vivo: Preventing muscle loss and myofiber size decrease, improving grip strength, and abolishing excessive fat accumulation
[100]
In vivo Mice 0.06% diet Improving obesity-related complications by restoring plasma thyroid hormone levels, and attenuating oxidative stress in the heart [101]
In vitro Human sperm 2.6, 6, 15, 30, 50, 100 μmol/L Improving obesity-related complications by restoring reproductive dysfunction like infertility [102]
Alzheimer’s disease and Parkinson’s disease
In vivo Rats 20 mg/kg/day Ameliorating ERS by downregulating the gene expression of CHOP and GRP78, inhibiting caspase-3 activity, and ameliorating oxidative damage via suppressing xanthine oxidase activity and protein carbonyl formation as well as activating glutathione peroxidase and Nrf2 signaling pathway [10]
In vitro CL2006 cells 100 μM Inhibiting the aggregation of Aβ by modulating specific proteins such as UBL/XBP-1 involved in proteostasis [103]
In vivo Mice 16 mg/kg/day Preventing memory loss by decreasing elevated levels of mitochondrial complex IV protein in the mouse brain via the activation of SIRT1 and AMPK pathways [104]
In vivo Mice 100 mg/kg/day Preventing memory loss via the activation of SIRT1 and AMPK pathways [105]
In vitro;
In vivo
SH-SY5Y cells;
Mice
50 μM;
50 mg/kg
Elevating miR-214 expression, leading to decreased mRNA expression of α-synuclein [106]
Sex-dependent effects of resveratrol
In vivo Rats 2.5 mg/kg/day Superior improvements of MI in females in terms of IVSDs, ESV, EF, FS, and IVRT, among which IVRT is purely sex-dependent [109]
In vivo Rats 50 mg/L in drinking water Increasing the relaxations to estrogen in aortae, more potent in males, probably due to resveratrol’s promoting nitric oxide and/or suppressing superoxide effects [110]
In vitro;
In vivo
MESC2.10 and SN4741 cells; Mice 20 mg/kg;
10 μM
Increasing DAT in the striatum in females but not in males;
Upregulating DAT in the dopaminergic cells by inducing its gene transcription
[111]
In vivo Mice 100 mg/kg Adverse effects in females but not in males, regarding weight loss, stool consistency, and discomfort [112]

Abbreviations used in the table: AC, acetyl; AhR, aryl hydrocarbon receptor; Akt, protein Kinase B; AMPK, AMP-activated protein kinase; Aβ, amyloid β; cAMP, cyclic adenosine monophosphate; CAT, catalase; cGMP, cyclic guanosine monophosphate; CHOP, C/EBP homologous protein; COX-2, cyclooxygenase-2; DAG, diacylglycerol; DAT, dopamine transporter; EF, ejection fraction; EGFR, epidermal growth factor receptor; eNOS, endothelial nitric oxide synthase; ERK, extracellular signal-regulated kinases; ERRα, estrogen related receptor α; ERS, endoplasmic reticulum stress; Erα, estrogen receptor α; ERα+, estrogen receptor alpha positive; ESV, end systolic volume; EV71, enterovirus 71; FERM, band 4.1, ezrin, radixin, and moesin; FoxO1, forkhead box protein O1; FS, fractional shortening; GPx, glutathione peroxidase; GRP78, glucose-regulated protein 78; GβL, G protein beta subunit-like; HFD, high-fat diet; HIF-1α, hypoxia-inducible factor 1α; HMGB1, high mobility group box 1; HMGB1, high mobility group box 1; HO-1, heme oxygenase (decycling) 1; HSL, hormone-sensitive lipase; ICAM-1, intercellular adhesion molecule-1; IFN-γ, interferon γ; IKK, IκB kinase; IL-1β, interleukin-1β; IVRT, isovolumic relaxation time; IVSDs, interventricular septal wall dimension at systole; IκBα, nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor α; JAK, Janus kinase; Keap1, Kelch-like ECH-associated protein 1; LDLR, low-density lipoprotein receptor; LKB1, liver kinase B1; LPS, lipopolysaccharides; MALAT1, metastasis-associated lung adenocarcinoma transcript 1; MAP2K, mitogen-activated protein kinase kinase; MAPK, mitogen-activated protein kinase; MCP-1, monocyte chemoattractant protein-1; MI, myocardial infarction; mSIN1, mammalian stress-activated protein kinase interacting protein 1; mTOR, mammalian target of rapamycin; mTORC2, mTOR Complex 2; NAD, nicotinamide adenine dinucleotide; NAFLD, non-alcoholic fatty liver disease; NALP3, NACHT, LRR, and PYD domains-containing protein 3; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; Nrf2, nuclear factor (erythroid-derived 2)-like 2; p53, phosphoprotein p53; PDE 3B, phosphodiesterase 3B expression; PDK1, phosphoinositide dependent kinase 1; PGC, peroxisome proliferator-activated receptor gamma coactivator 1α; PI3K, phosphatidylinositol 3-kinase; PIP2, phosphatidylinositol 4,5-bisphosphate; PIP3, phosphatidylinositol-3,4,5--trisphosphate; PKA, protein kinase A; PKC-θ, protein kinase C θ; PKG1α, cGMP-dependent protein kinase 1α; PLSCR1, phospholipid scramblase 1; PPAR-γ, peroxisome proliferator-activated receptor γ; PTEN, phosphatase and tensin homolog; RANTES, regulated on activation normal T cell expressed and secreted; RICTOR, the rapamycin-insensitive companion of mTOR; SARM, sterile α and armadillo motif protein; SIRT, sirtuin 1; α-SMA, smooth muscle actin; SOD, superoxide dismutase; SRB1, scavenger receptor class B type I; STAT, signal transducer and activator of transcription; tBregs, tumor-evoked regulatory B cells; TF, transcription factor; TGF-β, transforming growth factor β; TLR4, toll-like receptor 4; TNF-α, tumor necrosis factor α; TRIF, toll/IL-1 receptor domain-containing adaptor inducing β interferon; UBL, ubiquitin-like protein; XBP-1, X-box binding protein 1.