Skip to main content
. 2020 Feb 25;9(3):193. doi: 10.3390/antiox9030193

Table 2.

Systematic presentation of studies on possible use of natural Nrf2 activators in therapy with possible mechanisms of action connected to Nrf2 activation pathways.

Natural Products
Type of Study Experimental Model Treatment Doses and Duration Observed Mechanism of Action/Effects Proposed Application in Therapy Ref.
In vivo male Albino rats Wistar strain 200 mg/kg dose of curcumin for four consecutive days oral administration
  • enhanced nuclear translocation and ARE-binding of Nrf2

  • curcumin protects against dimethylnitrosamine (DMN)-induced hepatic injury

chemopreventive agent [360]
In vivo male C57BL/6J mice daily treated with curcumin at the dose of 50 mg/kg body weight by oral gavage
  • mediated nuclear translocation of Nrf2, followed by induction of HO-1

  • curcumin intervention dramatically reversed the defects in Nrf2 signaling induced by high fat diet

improving glucose intolerance [361]
In vivo male Sprague–Dawley rats supplemented with curcumin (1 g/kg diet) for 16 weeks
  • increased HO-1 expression was increased along with suppressed oxidative stress as well as reduced hepatic fat accumulation and fibrotic changes

attenuating the pathogenesis of fatty liver induced metabolic diseases [362]
In vivo/In vitro female specific pathogen-free BALB/c mice
mouse macrophage RAW264.7 cells
on day 22, the mice were treated with curcumin (200 mg/kg) 1 h before ovalbumin challenge.
cells treated with different concentrations (0, 5, 10, 25, and 50 μmol/L) of curcumin for 24 h or with 50-μmol/L curcumin for different lengths of time (0, 4, 8, 12 and 24 h)
  • Nrf2 and HO-1 levels in lung tissues were significantly increased

  • heme oxygenase-1 and nuclear Nrf2 levels were enhanced in dose- and time-dependent manners in curcumin-treated RAW264.7 cells.

potentially effective drug in asthma treatment (alleviate airway inflammation in asthma through the Nrf2/HO-1 pathway) [363]
In vitro primary cultures of cerebellar granule neurons(CGNs) of rats pretreated with 5–30 μM curcumin
  • mediated neuro-protection against hemin induced damage via Nrf2 dependent HO-1 expression

neuroprotective agent [364]
In vitro human breast cancer cell line MCF-7 treatment with DMSO (vehicle) or various concentrations of curcumin for 12, 24 or 48 h
  • inhibition of the proliferation of breast cancer cells through Nrf2-mediated down-regulation of Fen1 expression

chemotherapeutic agent [365]
In vivo female Sprague-Dawley rats food supplemented with resveratrol equivalent to 50, 100, or 300 mg/kg body weight/d
  • elevated protein and mRNA expression of hepatic Nrf2, attenuation of oxidative stress and suppression of inflammatory response mediated by Nrf2

prevention and intervention of human hepatocellular carcinoma [366]
In vivo/In vitro female August Copenhagen Irish rats non-tumorigenic human breast epithelial cell line MCF-10A resveratrol given as a 50 mg subcutaneous pellet every other month during 8 months to animals subcutaneously treated with 3 mg E2 pellets prepared in cholesterol cells were treated with E2 (10 and 50 nM) and Res (50 µM) for up to 48 h
  • resveratrol treatment alone or in combination with E2 significantly upregulated expression of nuclear factor erythroid 2-related factor 2 (NRF2) in mammary tissues; increased expression of NRF2-regulated genes codying for antioxidant enzymes (NQO1, SOD3 and OGG1) involved in protection against oxidative DNA damage

  • inhibition of suppression of FMO1 and AOX1 genes expression via regulation of NRF2

a potential chemopreventive agent in the development of therapeutic strategies for the prevention of estrogen-induced breast neoplasia [367]
In vitro MCF-10F and MCF-7 cells retreated with 0.1 to 30 nmol/L TCDD with or without 25 μmol/L resveratrol for 72 h and then incubated with E2 (0.1–10 μmol/L) for 24 h
  • induction of NAD(P)H quinone oxidoreductase 1 (NQO1) in MCF-10F cells exposed to resveratrol may involve the Nrf2-Keap1-ARE pathway (dissociation of Nrf2 from Keap 1 and translocation of Nrf2 to the nucleus, where it binds to the ARE to activate the transcription of NQO1 mRNA)

a potential chemopreventive agent against estrogen-initiated breast cancer [368]
In vitro primary rat hepatocytes were obtained from Sprague–Dawley male rats cells were incubated in the presence of resveratrol for 24 and 48 h ( at concentrations of 25, 50 and 75 µM)
  • increase in the level of Nrf2 and induction of its translocation to the nucleus, and the increase in the concentration of the coding mRNA for Nrf2

protection of liver cells from oxidative stress induced damage (chemopreventive agents) [115]
In vitro human type II alveolar epithelial cell line, A549 cells were treated with various concentrations of native EGCG (5, 10, 20, 40, 60, 80 and 100 μM) and nano EGCG (1, 2, 4, 6, 8, 10 and 12 μM) and allowed to grow for 48 h
  • increased expression of Nrf2 and Keap1 in native and nano EGCG treated A549 cells that could regulate apoptosis

chemotherapeutic in lung cancer [369]
In vitro bovine aortic endothelial cells (BAECs) cells treated with various concentrations of EGCG
  • upregulates Nrf2 levels in nuclear extracts and increases ARE-luciferase activity

therapeutic targets in a variety of oxidant- and inflammatory-mediated vascular diseases [370]
In vivo male albino Wistar rats; animal model of bleomycin-induced pulmonary fibrosis intraperitoneally treated with EGCG at a dosage of 20 mg/kg body weight, once daily throughout 28 days
  • EGCG enhances antioxidant activities and Phase II enzymes involving Nrf2–Keap1 signaling, with subsequent restraint inflammation during bleomycin-induced pulmonary fibrosis

  • no significant change in the expression of Keap1

treatment of diseases such as pulmonary fibrosis [371]
In vitro human hepatocytes (HHL5) and hepatoma (HepG2) cells exposed to various concentrations of sulforaphane for different times with DMSO (0.1%) as control
  • increased nuclear Nrf2 levels and intracellular GSH levels in both cell lines but with slightly different pattern indicating a potential risk of chemo-resistance of using sulforaphane for chemoprevention

possible induction of pro-survival effects in cancer cells [128]
In vivo male BALB/c mice (6 weeks) 5 μmol/animal sulforaphane plus different doses of microcystin-LR
  • Nrf2 translocation to the nucleus in mouse livers

  • induction of Nrf2 downstream target genes, NQO1 and HO-1, two phase II enzymes

effective in cytoprotection against MC-LR-induced hepatotoxicity [372]
In vitro adult rat cardiomyocytes exposed to 5μM sulforaphane with or without H2O2
  • upregulation of Nrf2 by sulforaphane occurs at 1 h of incubation

  • increased protein expression of PGC-1α

protective action against oxidative damage, however, timeline of the sulforaphane actions needs to be established [373]
In vivo male BALB/c mice (6 weeks) 5 μmol/animal sulforaphane plus different doses of microcystin-LR
  • Nrf2 translocation to the nucleus in mouse livers

  • induction of Nrf2 downstream target genes, NQO1 and HO-1, two phase II enzymes

effective in cytoprotection against MC-LR-induced hepatotoxicity [372]
Electrophilic/Covalent
Triterpenoids
In vitro K562 myeloid leukemia cells Exposed to 0.05–10 μM CDDO-Me for 24–48 h
  • Down-regulated Na+/K+ ATPase

  • Arrested cells in G2/M and S phases

  • Increased mitochondria and death receptor-dependent and ER-mediated apoptosis

  • Triggered activation of autophagy

Activated and potentiated the effects of the apoptosis and autophagy pathways to kill K562 cancer cells [374]
In vitro and In vivo SKOV3, OVCAR3, A2780, A2780/CP70 and Hey2 ovarian cancer cells 0–50 μM CDDO-Me depending on cell assays
20 mg/kg CDDO-Me in xenograft model using nude mice
  • Binds to USP7 cells, decreasing MDM2, MDMX and UHRF1

  • Suppresses tumor growth in a xenograft model

Targets apoptosis-related substrates, increasing apoptosis and reducing growth of ovarian cancer cells [375]
In vitro MiaPaCa-2 and BxPC-3 cell lines
6 week old Scid/Ner mice
0.625–5μM CDDO-Me in cell culture
CDDO-Me 7.5 mg/kg × 5 days/wk by oral gavage until day 40 (to treat primary tumor) or day 100 (to treat residual disease)
  • Decreased proliferation and increased apoptosis in K-ras normal and mutant cells

  • Inhibits antiapoptotic pathways

  • Inhibited tumor growth in mice

  • Increased survival time of mice

Combination of in vitro and in vivo effects demonstrate that CDDO-Me will increase apoptosis in pancreatic ductal adenoma carcinoma cell lines and improve the survival of animals [376]
In vitro MDA-MB 435, MDA-MB 231, MDA-MB 468,
BT-549, T47D and MCF-7 breast cancer cells
CDDO-Me 1.5 μM for 4 h
  • Induces endoplasmic reticulum vacuolation

  • Induces apoptotic pathways

  • Increases intracellular calcium and generation of reactive oxygen species

CDDO-Me-induced c-FLIPL downregulation and relationship between Ca2+ influx and ROS generation are keys in controlling breast cancer growth. [377]
In vitro HO8910 and SKOV3 ovarian cancer cells CDDO-Me concentration range of 0–100 μM
5 μM CDDO-Me in assays requiring a single concentration
  • Upregulates Hsp70

  • Degrades Hsp90-associated protein—ErbB2 and Akt

  • CDDO-Me reacts with nucleophiles on Hsp90 to form Michael adducts

May provide added insight to CDDO-Me action, with Hsp90 as a novel target [378]
In vivo C57BL/6 WT mice
LSL-KrasG12D/+; Pdx-1-Cre (KC) mice
Polyoma-middle T (PyMT) mice
CDDO-Im (100 mg/kg diet) fed 2 days prior to LPS injections
  • Reduced the lethal effects of LPS injection in mutant mice

  • Increased migration of CD45+ cells

  • Increased percentage of Gr1+ myeloid-derived suppressor cells

  • CDDO-Im decreased IL-6, CCL-2, VEGF, and G-CSF in mutant mice

Postulating the use of CDDO-Im as prophylaxes in the development of pancreatic cancer within susceptible populations. This is due to the reduction in proinflammatory mediators. [379]
In vitro Human Jurkat E6-1 cells CDDO-Im 1 nM and 10 nM for 30 min prior to activation with anti-CD3/anti-CD28
  • Inhibited production of IL-2

  • Suppression of CD25 in Nrf2-dependent manner with no effect on CD69

Nrf2 activation by CDDO-Im reduces IL-2 secretion and CD25 expression suggesting a role in potential anticancer therapy. [380]
Dithiolethiones
In vitro Mouse carcinoma Hepa-1c1c7 cells Oltipraz 5–60 μM
anetholedithione (ADT) 3–15 μM
1,2-dithiole-3-thione (D3T) 1–5 μM
  • Dithiolethiones are reduced in Hepa-1c1c7 cells leading to generation of superoxide radicals

  • Superoxide dismutates to form hydrogen peroxide promoting translocation of Nrf2 to nucleus

  • Translocated Nrf2 results in the upregulation of various Phase II enzyme expression.

Use of D3T and members of this family may be able to modify KEAP-1 activity and upregulate the expression of Phase II enzymes [381]
In vivo Male Fischer 344 (100 g) D3T administered by oral gavage 0.5 mmol/kg (in distilled water with 1% Cremaphor, and 25% glycerol
  • Increased heme oxygenase (HO-1) activity

  • Increased expression of HO-1

  • Increased levels of ferritin

Very early paper describing the potential utility of Dithiolethiones, like D3T, may offer protection against pro-carcinogenic compounds that increase oxidative stress [382]
In vitro HT29 colon adenocarcinoma cells 30μM D3T in DMSO vehicle (less than 0.1% final DMSO concentration)
  • Increased expression and activity of multiple reductases—Thioredoxin reductase 1, prostaglandin reductase 1, NAD(P)Quinone oxidoreductase 1

  • Potentiation of hydroxymethylacylfulvene action in alkylating DNA

D3T was a much stronger inducer of reductases compared to selenite and increased potency of the anticancer drug, hydroxymethylacylfulvene [383]
In vivo Male Fisher 344 rats (90–100 g) Oral gavage of DST at 0.03 to 0.3 mmol/kg body wt at 3 days/week for 3 weeks
Also 0.1 mmol/kg for measuring hepatic protein expression
  • Reduced the pre-cancer potency of aflatoxin B by inhibiting the expression of glutathione S-transferase-placental isoform

  • Blocked aflatoxin-mediated increase in

  • Induced multiple hepatic genes associated with detoxifying aflatoxin—including glutathione

  • S-transferase A5 (GSTA5) and AFB1 aldehyde reductase

D3T is more potent than older Dithiolethiones like oltipraz and could be a probe for measuring anticancer potencies of this drug class [384]
In vitro HepG2 hepatic and LS180 colon cells S-diclofenac and S-sulindac in range of 0–100 μM
  • Inhibited activity and expression of CYP1A1, 1B1 and 1A2

  • Regulates aryl hydrocarbon receptor pathway

  • Blocked binding of aryl hydrocarbon to the responsive element

  • Increased expression of anticancer enzymes, glutathione S transferase, glutamate cysteine ligase, and glutathione reductase

The NSAIDs with the dithiolethione group, S-diclofenac and S-sulindac, may function as effective anticancer agents [385]
In vitro HT29 and HCT116 colon adenocarcinoma 100 μM Oltipraz with 0.2% MeSO as a solvent control
  • Induction of p65, IκB kinase α (IKKα), IκB kinase β (IKKβ), and NF-κB-inducing kinase (NIK)

  • IκBα phosphorylation was also induce

  • Induction of protein binding to a consensus NF-κB element

  • Transcriptional activation of QR was decreased

  • Both MEKK3 and NIK exert effects on IKKα/β activation via different pathways

This is a novel pathway involved in QR gene regulation and may provide insight to the actions of oltipraz as an anticancer agent [386]
Non-Electrophilic/Non-Covalent
In vivo and in vitro Male C57/Bl6 mice (22 g) and bone marrow-derived mouse macrophage cells RA839 was dissolved in a vehicle of 95% (v/v) hydroxyethyl cellulose (0.5% (w/v))/5% (v/v) solutol—injected IP at 30 mg/kg.
General RA839 interactions measured at a concentration of 10 μM
  • Binds to KELCH domain with affinity of 6 μM

  • Blocks Nrf2-Keap-1 protein-protein interaction

  • Modified activity of multiple genes in mouse macrophage cells

  • Reduced the induction of nitric oxide release and inducible nitric oxide synthase activity

  • Induced Nrf2 gene expression in mouse liver

RA839 may be a useful tool in developing anticancer drugs that target the prevention of Nrf2-Keap1 protein interaction. [80]
In vitro THP-1 cells Cells were exposed to TAT14 using a concentration range of 0–75 μM
  • Activates heme oxygenase 1 expression and activity

  • Did not alter Nrf2 mRNA expression

  • Reduced LPS-induced TNF expression

The 14-mer TAT fragment interacts with Keap1, preventing Nrf2-Keap1 association, allowing Nrf2 to activate mediators downstream [387]
In vitro HepG2 hepatic and U2OS bone cell lines ML334 and its isomers in a concentration range of 0–100 μM
  • Binds with high affinity to KEAP1

  • Strong inducer of ARE activity in both hepatic and bone cell lines

First description of ML334 as a potent inhibitor of Nrf2-Keap1 interaction. Highly potent. [388]
In vitro Immortalized baby mouse kidney epithelial cells (iBMK) and MDA-MB-231 breast cancer cells Geopyxin F and other “geopyxin” isomers were used in a concentration range of 0–70 μM depending on assay
  • Geopyxin F was the most potent of Geopyxin compounds at inducing Nrf2 activity

  • Geopyxin F displayed almost not toxicity/lethality in MDA-MB-231 cells

  • Geopyxin F increased autophagosome formation in iBMK cells

  • Geopyxin F prevents ubiquitination and stabilizes Nrf2 (KEAP1-dependent)—but is independent of interactions at the Cys151 in KEAP1

Geopyxin F, demonstrated a higher level of protection compared to electrophilic Nrf2 activators. Heightened potency suggests that Geopyxin F may be a useful anticancer compound. [78]
In vitro MCF-7 breast cancer cells Multiple drugs were used as “off-label” activators of Nrf2
Astemizole 8 μM
Tamoxifen 1 μM
Trifluoperazine 10 μM
  • Astemizole, Tamoxifen and Trifluoperazine increased expression of the NQO1, HO-1, and GCLM genes

  • After 24 h, the stimulated gene expression to basal was highest in Astemizole compared to sulforaphane

  • The genes for the detoxifying enzymes—NAD(P)H quinone oxidoreductase 1 (NQO1), heme oxygenase 1 (HO1) where most sensitive to upregulation

After large-scale screening, select drugs were chosen based on their ability to activate Nrf2. This shows that ‘off-label’ mechanisms may have benefit as anticancer drugs. Astemizole was the best candidate. [79]