Next Article in Journal
Antioxidants, Gut Microbiota, and Cardiovascular Programming: Unraveling a Triad of Early-Life Interactions
Previous Article in Journal
Genetic Study of Total Phenolic Content and Antioxidant Activity Traits in Tetraploid Wheat via Genome-Wide Association Mapping
Previous Article in Special Issue
Ferroptosis and Nrf2 Signaling in Head and Neck Cancer: Resistance Mechanisms and Therapeutic Prospects
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Non-Electrophilic Activation of NRF2 in Neurological Disorders: Therapeutic Promise of Non-Pharmacological Strategies

1
Translational Brain Research Laboratory, Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
2
Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
3
Department of Neurosurgery, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
4
The Litwin-Zucker Center for Alzheimer’s Disease & Memory Disorders, Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
5
Institute for Neurology and Neurosurgery, Northwell Health, Manhasset, NY 11030, USA
6
Department of Biology, Hofstra University, Hempstead, NY 11549, USA
*
Author to whom correspondence should be addressed.
Antioxidants 2025, 14(9), 1047; https://doi.org/10.3390/antiox14091047
Submission received: 15 July 2025 / Revised: 20 August 2025 / Accepted: 21 August 2025 / Published: 25 August 2025
(This article belongs to the Special Issue Oxidative Stress and NRF2 in Health and Disease—2nd Edition)

Abstract

Nuclear factor erythroid 2-related factor 2 (NRF2) serves as a master transcriptional regulator of cellular antioxidant responses through orchestration of cytoprotective gene expression, establishing its significance as a therapeutic target in cerebral pathophysiology. Classical electrophilic NRF2 activators, despite potent activation potential, exhibit paradoxically reduced therapeutic efficacy relative to single antioxidants, attributable to concurrent oxidative stress generation, glutathione depletion, mitochondrial impairment, and systemic toxicity. Although emerging non-electrophilic pharmacological activators offer therapeutic potential, their utility remains limited by bioavailability and suboptimal potency, underscoring the imperative for innovative therapeutic strategies to harness this cytoprotective pathway. Non-pharmacological interventions, including neuromodulation, physical exercise, and lifestyle modifications, activate NRF2 through non-canonical, non-electrophilic pathways involving protein–protein interaction inhibition, KEAP1 degradation, post-translational and transcriptional modulation, and protein stabilization, though mechanistic characterization remains incomplete. Such interventions utilize multi-mechanistic approaches that synergistically integrate multiple non-electrophilic NRF2 pathways or judiciously combine electrophilic and non-electrophilic mechanisms while mitigating electrophile-induced toxicity. This strategy confers neuroprotective effects without the contraindications characteristic of classical electrophilic activators. This review comprehensively examines the mechanistic underpinnings of non-pharmacological NRF2 modulation, highlighting non-electrophilic activation pathways that bypass the limitations inherent to electrophilic activators. The evidence presented herein positions non-pharmacological interventions as viable therapeutic approaches for achieving non-electrophilic NRF2 activation in the treatment of cerebrovascular and neurodegenerative pathologies.

1. Introduction

Nuclear factor erythroid 2-related factor 2 (NRF2) functions as a master transcriptional regulator of cellular antioxidant defense, governing the expression of hundreds of cytoprotective and detoxifying genes [1,2,3,4]. While NRF2 activation presents an attractive therapeutic strategy capable of coordinating multi-antioxidant responses, emerging evidence challenges this paradigm. Despite documented beneficial effects, accumulating data demonstrate that classical NRF2 activators exhibit suboptimal efficacy against oxidative damage when compared to single antioxidant interventions [5,6,7] (Figure 1), necessitating critical re-evaluation of current approaches. Under normal physiological conditions, pharmacological NRF2 activators have paradoxically been shown to deplete glutathione [6], compromise mitochondrial function [8], and elevate reactive oxygen species (ROS) levels despite potent NRF2 pathway activation. In chronic pathological settings, where these disturbances intersect with pre-existing cellular damage, their effects become highly variable, ranging from diminished therapeutic benefit to an aggravation of tissue injury and acceleration of disease progression [6,7,9,10,11,12]. Notably, dimethyl fumarate (DMF) accelerates glutathione depletion in multiple sclerosis [9], further driving the observed downregulation of glutathione in multiple sclerosis [10], and potentially causing hepatic injury even with acute application. This paradox intensifies in severe, acute, neurological injury: DMF and sulforaphane exacerbate protein oxidation and infarct expansion [6,13,14,15] relative to glutathione [7] or superoxide dismutase (SOD) mimetics [11,12], occasionally yielding inferior outcomes versus untreated controls in stroke models [15]. Traumatic brain injury studies corroborate these findings, with DMF augmenting lipid peroxidation and compromising neuroprotection [16,17,18]. These observations suggest that, under specific conditions, classical NRF2 activators may fail to achieve anticipated therapeutic benefits and potentially compromise neural integrity. Consequently, mechanistic dissection of this paradox and identification of alternative NRF2 activation strategies constitute essential steps toward realizing the full therapeutic potential of this critical cytoprotective pathway.
Current NRF2 activators predominantly employ electrophilic mechanisms (Figure 2), requiring ROS generation to dissociate NRF2 from the NRF2:Kelch-like ECH-associated protein 1 (KEAP1) complex [19]. This ROS-mediated activation depletes glutathione reserves, increases protein oxidation and lipid peroxidation, and disrupts mitochondrial bioenergetics [1,2,3,4,5,6,7,20,21,22,23,24]. While metabolically intact cells may tolerate this oxidative burden, it poses significant risks to cells experiencing oxidative stress, particularly during conditions demanding rapid, pronounced NRF2 activation and in highly vulnerable tissues such as the brain. The brain exhibits exceptional metabolic demands, consuming 20% of total oxygen despite comprising only ~2% of body mass [5]. This disproportionate oxygen utilization for glucose-dependent ATP synthesis inherently generates substantial ROS. Combined with high lipid content, limited antioxidant capacity, and dense neuronal populations, these characteristics render the brain uniquely susceptible to oxidative damage [25]. Neurons demonstrate particular vulnerability due to their dependence on mitochondrial metabolism and elevated mitochondrial ROS production [20,21,22,23,24,26,27,28,29,30], making them highly sensitive to electrophile-induced bioenergetic dysfunction. Glutathione depletion precipitates neurodegeneration [31,32], contributing to neurodegenerative diseases, chronic neurological dysfunction, and psychological disorders while increasing susceptibility to acute traumatic injury [33,34]. The brain’s intrinsic oxidative vulnerability may therefore explain the compromised efficacy of electrophilic NRF2 activators, as their ROS-dependent mechanism [19,35,36] exacerbates existing oxidative stress. Consequently, developing non-electrophilic NRF2 activation strategies represents an essential requirement for safely exploiting NRF2’s neuroprotective potential.
Recent investigations have increasingly focused on non-electrophilic NRF2 activation strategies. Unlike conventional approaches that induce NRF2 through non-specific ROS generation, non-electrophilic activators employ targeted mechanisms including protein–protein interaction (PPI) disruption, KEAP1 degradation, protein stabilization to enhance NRF2 bioavailability, and post-translational/post-transcriptional modifications to augment NRF2 function [37,38,39] (Figure 2). Despite theoretical advantages, these approaches exhibit notable limitations. Compounds promoting KEAP1 proteasomal degradation demonstrate efficacy comparable to electrophilic activators but remain scarce. While PPI inhibitors show therapeutic potential in diabetic wound healing and lipopolysaccharide-induced inflammation [39,40], they typically exhibit reduced potency relative to electrophilic compounds, similar to the efficacy observed with modulators targeting post-translational/transcriptional processes [38]. This diminished efficacy likely reflects the critical role of NRF2 bioavailability as a rate-limiting factor, which selective PPI inhibitors and post-translational/transcriptional modulators fail to enhance sufficiently compared to electrophilic agents. Consequently, identifying novel non-electrophilic activators that either promote KEAP1 degradation or integrate multiple mechanisms to simultaneously enhance NRF2 bioavailability and activity remains imperative. Non-pharmacological interventions represent a promising research direction, having demonstrated clinical efficacy in oxidative-stress-mediated cerebral pathologies [41] and NRF2 activation capacity [42,43,44,45,46,47]. Substantial evidence indicates these interventions operate through non-electrophilic pathways, including modulation of glycogen synthase kinase 3β (GSK3β), AMP-activated protein kinase (AMPK), and p38 mitogen-activated protein kinase (p38 MAPK), among others [42,43,44]. Their multi-mechanistic action may circumvent efficacy limitations inherent to single-target non-electrophilic approaches. Despite compelling evidence supporting their therapeutic utility and non-electrophilic mechanisms, however, non-pharmacological NRF2 activators remain inadequately investigated for neurological applications.
This review comprehensively examines the therapeutic potential of non-pharmacological NRF2 activation in neurological disorders, with particular emphasis on non-electrophilic mechanisms and multi-target approaches. Our investigation addresses three critical questions: (1) whether non-pharmacological interventions operate through non-electrophilic pathways; (2) whether they exhibit limitations similar to electrophilic activators; and (3) whether they overcome limitations inherent to current non-electrophilic agents. We systematically evaluate effects and mechanisms across distinct pathophysiological contexts—physiological conditions, chronic pathology, and acute injury. The review investigates whether non-pharmacological interventions generate distinct neurological outcomes compared to pharmacological NRF2 activators, whether their activation mechanisms and downstream signaling are optimized for the brain’s unique metabolic milieu, and whether they represent substantive mechanistic innovations beyond current strategies. Observations from peripheral tissues are analyzed as potential indicators of central nervous system efficacy. We further examine how multi-mechanistic NRF2 activation by non-pharmacological interventions confers therapeutic advantages for clinical translation. Through critical evaluation of non-electrophilic, non-pharmacological approaches, this review identifies promising directions for mechanistic research and therapeutic development in NRF2-targeted neurological interventions.

2. Mechanistic Characterization of Non-Pharmacological NRF2 Activation: Strategic Implementation of Non-Electrophilic Pathways

Non-pharmacological NRF2 activators operate through non-electrophilic mechanisms, either partially or completely, and demonstrate therapeutic efficacy in conditions where conventional electrophilic activators exhibit suboptimal therapeutic indices [42,43,44,45,46,47]. These interventions show substantial potential to circumvent the limitations of electrophilic NRF2 activators while preserving the beneficial effects of NRF2 activation in disease states. To elucidate this paradigm, we first examine the current landscape of classical electrophilic pharmacological interventions, followed by a comprehensive analysis of non-pharmacological approaches. Particular emphasis is placed on their non-electrophilic mechanisms and capacity to address inherent limitations of conventional Nrf2 activators. Non-pharmacological interventions are systematically categorized according to the oxidative stress context: (i) absence of oxidative stress, (ii) gradually developing oxidative stress, and (iii) acutely developing oxidative stress from immediate causative factors. This organizational framework directly addresses critical concerns regarding the application of classical electrophilic NRF2 activators under conditions of elevated cellular stress or during chronic therapeutic regimens.

2.1. Electrophilic NRF2 Activation: Mechanistic Paradoxes and Therapeutic Limitations

Electrophilic NRF2 activators demonstrate substantial potency, inducing 1.5- to 3-fold increases in NRF2 activity [1,2,3,4,6,19], conferring theoretical therapeutic efficacy. However, the reactive promiscuity inherent to non-selective electrophiles constrains their clinical utility in neurological disorders (Figure 1). In cerebral pathologies, electrophilic activators paradoxically elevate oxidative stress markers/mediators concurrent with NRF2 induction and antioxidant upregulation [6], compromising therapeutic indices and potentially inducing organ toxicity. Mechanistically, oxidative modification of cysteine 151 dissociates NRF2 from the NRF2:KEAP1 complex, preventing ubiquitination and proteasomal degradation. This process generates elevated electrophile concentrations that may deplete antioxidant reserves before therapeutic benefits manifest in neural tissue [37,38]. Indeed, glutathione depletion represents the primary mechanism underlying DMF’s efficacy in psoriasis [48,49]. In neural tissue, this depletion proves particularly detrimental through downstream mitochondrial dysfunction [9]. Glutathione depletion restricts antioxidative capacity, elevating mitochondrial ROS and enhancing mitochondrial stress responses, ultimately compromising organellar function. This particularly affects neurons given their mitochondrial dependence and inherent oxidative vulnerability [50]. Consequently, under elevated oxidative stress conditions, electrophilic NRF2 activators yield negligible benefits, null effects, or paradoxically exacerbate pathological presentations [6,7,13,14,15]. Furthermore, chronic administration of electrophilic activators may induce organ toxicity, even under conditions of minimal baseline oxidative burden [38,51,52,53,54].

2.2. Non-Pharmacological NRF2 Activation Under Physiological Conditions: Mechanistic Insights in the Absence of Oxidative Stress

Unlike electrophilic NRF2 activators associated with organ toxicity, non-pharmacological approaches demonstrate favorable safety profiles without organ damage or mitochondrial bioenergetic disruption. Electrophilic NRF2 activators present limitations even under physiological conditions despite their potency. Notably, electrophilic compounds can substantially reduce glutathione levels within 60 min of administration [55,56,57]. Sulforaphane administration, while effectively inducing NRF2 [58], produces a 6-fold increase in brain nitrotyrosine in healthy animals [6]. However, absent pre-existing pathology, these paradoxical effects produce minimal adverse outcomes, as evidenced by widespread consumption of NRF2-activating polyphenolic compounds as dietary supplements [59,60,61,62].
Multiple non-pharmacological interventions demonstrate NRF2 activation in physiological contexts, with many exhibiting partially or completely non-electrophilic mechanisms (Table 1, Figure 3). These include physical exercise, transcranial magnetic stimulation (TMS), therapeutic hypothermia (TH), diving reflex (DR), remote ischemic conditioning (RIPreC), photobiomodulation (PBM), intermittent fasting (IF), and dietary modifications. These interventions consistently induce significant NRF2 pathway activation across various metrics. Short-term physical exercise (≤2 days) elevates NRF2 levels and NRF2/antioxidant response element (ARE) binding activity, significantly increasing superoxide dismutase 1 and 2 (SOD1, SOD2), catalase, and heme oxygenase-1 (HO-1) expression [63,64,65]. The response demonstrates duration-dependence, with 6-h treadmill sessions producing more pronounced effects than 1-h sessions [64]. Similarly, DR exhibits time-dependent NRF2 activation, with enhanced NRF2 phosphorylation after 28-day treatment compared to single sessions, though acute treatment produces greater antioxidant gene upregulation [66].
Analysis of 19 documented non-pharmacological interventions under physiological conditions demonstrates consistent NRF2 activation. However, mechanistic understanding in healthy contexts remains limited. TMS and PBM activate p62, which competitively binds KEAP1 to release NRF2 without electrophile generation, while promoting autophagic KEAP1 degradation [44,67]. PBM upregulates NRF2 and LC3 expression in adipose-tissue-derived stem cells (ADSCs) [67] and reduces KEAP1 in human periodontal ligament stem cells [68]. DR employs similar non-electrophilic mechanisms through p62, AMPK, sirtuin 1 (SIRT1), and phosphoinositide 3-kinase (PI3K) induction, resulting in quinquepartite modulation: KEAP1 degradation, NRF2 protein–protein interaction inhibition, enhanced NRF2 stability, and increased DNA binding and transcription [66]. In contrast to TMS, PBM, and DR, physical exercise and IF exhibit dual activation mechanisms, incorporating both electrophilic and non-electrophilic pathways through ROS generation [63,64,69]. However, these interventions do not induce glutathione depletion, potentially due to concurrent non-electrophilic mechanisms including TLR4 and ZEB1 upregulation with ZEB2 downregulation [70,71].

2.3. Non-Pharmacological NRF2 Activation Under Conditions of Progressive Oxidative Stress

The presence of progressive oxidative stress significantly affects the efficacy and safety profile of electrophilic NRF2 activators. A critical translational limitation of electrophilic NRF2 activators involves cumulative cytotoxicity during chronic administration, manifesting as hepatotoxicity, nephrotoxicity, and cardiotoxicity [72,73,74,75]. DMF treatment in multiple sclerosis exemplifies this phenomenon, where patients exhibit elevated serum aminotransferases and bilirubin, progressing to severe acute hepatitis with hepatocellular bridging necrosis, even without pre-existing hepatic pathology [72,74,75,76]. Despite immediate withdrawal of DMF upon detection of elevated hepatic enzymes, liver injury may nonetheless progress. Notably, the incidence of DMF-associated hepatotoxicity or related adverse events increases markedly after at least three months of exposure, whereas patients with shorter treatment durations exhibit a substantially lower risk [73]. By contrast, omaveloxolone therapy in Friedreich’s ataxia has, to date, been accompanied only by mild aminotransferase elevations that are readily managed by dose modification [77]. Paradoxically, DMF administration in psoriasis patients induces only minor hepatic enzyme increases [78], whereas in multiple sclerosis populations it is associated with a higher rate of acute hepatotoxicity [76]. This differential toxicity correlates with varying oxidative stress burdens: despite both existing as autoimmune diseases, multiple sclerosis increases advanced oxidation protein products by ~24%, reduces thiol groups by ~49%, and diminishes ferric reducing ability by ~31% [75], whereas psoriasis demonstrates inconsistent advanced oxidation protein product modulation and only ~12% thiol group reduction [79,80,81]. Omaveloxolone’s favorable safety profile may reflect the relatively low oxidative-stress milieu in Friedreich’s ataxia. Although oxidative stress contributes to disease pathology, this disorder is not characterized by excessive reactive oxygen species; rather, frataxin-deficient cells display increased sensitivity to physiological oxidant levels [82]. By contrast, Phase III trials of bardoxolone methyl in chronic kidney disease were terminated due to serious cardiovascular adverse events [72]. The oxidative-stress-associated sarcopenia characteristic of chronic kidney disease [83] potentially increases cardiac vulnerability to cytotoxic insults. Collectively, these observations support a dose–response relationship between baseline oxidative stress levels and the toxicity of electrophilic activators, implying the existence of a threshold of oxidant burden beyond which these agents become contraindicated. In contrast, non-pharmacological NRF2 activators exhibit a favorable safety profile and preliminary signs of efficacy in pre-clinical models of chronic disorders characterized by progressive oxidative stress (Table 2), with particularly promising results in the brain.
Non-pharmacological interventions effectively induce NRF2 activation across diverse chronic pathologies, including non-alcoholic fatty liver disease, parkinsonism, Huntington’s disease, vascular dementia, Alzheimer’s disease (AD), post-traumatic stress disorder (PTSD), and chronic unpredictable mild stress (CUMS) (Table 2, Figure 3). Electroacupuncture activates NRF2 in complex regional pain syndrome type-1, colitis, sepsis, and diabetic encephalopathy [84,85,86,87]. Physical exercise demonstrates therapeutic benefit in parkinsonism, high-fat diet (HFD)-induced non-alcoholic fatty liver disease, and senescence, upregulating NRF2 and SOD1 expression while reducing lipid peroxidation and maintaining glutathione homeostasis [43,88,89,90]. Combined exercise and L-dopa therapy in Parkinson’s disease models demonstrates that exercise-induced NRF2 upregulation mitigates L-dopa-associated oxidative stress, illustrating the potential of non-pharmacological adjunctive approaches. Neurodegenerative disorders demonstrate responsiveness to non-pharmacological NRF2 activation, attributed to non-electrophilic mechanisms that upregulate glutathione peroxidase 4 (GPX4), glutathione reductase, HO-1, and manganese superoxide dismutase. Beyond physical exercise, electroacupuncture ameliorates motor impairment in models of hemiparkinsonism and Parkinson’s disease models through striatal NRF2 and HO-1 upregulation [88,90,91,92]. NRF2 activation effectively reduces symptomatology in models of vascular dementia using TMS, EA, and remote ischemic pre-conditioning [47,93,94], Huntington’s disease using TMS [95], and neuropsychiatric models including anxiety, depression, and PTSD using TMS, EA, and electroconvulsive therapy (ECT) [96,97,98,99]. Additionally, olive-oil-enriched diets and caloric restriction (CR) attenuate age-related degeneration through NRF2 upregulation and reduction of oxidative markers including malondialdehyde (MDA) and C-reactive protein (CRP), resulting in enhanced cognitive performance [100,101,102,103,104].
Table 1. Non-pharmacological NRF2 interventions investigated in physiological conditions.
Table 1. Non-pharmacological NRF2 interventions investigated in physiological conditions.
ModelIntervention TypeSpeciesIntervention ParametersFocal Organ/CellNRF2 Measurement TimingNRF2 ModulationOther EffectsMechanismRef.
HealthyPhysical exerciseMiceTreadmill running; 2 consecutive days; 60 min per day; 14 m/min; 10% slopeHeartDirectly after AESAES: ↑ NRF2NRF2−/− mice: ↓ GSHROS production[63]
Physical exerciseMiceTreadmill running; 20 m/min; 5% slope; one or six hoursHind limbDirectly after PE6H PE: ↑ NRF2/ARE binding activity
1H PE: ↔ NRF2/ARE binding activity
6H: ↑ GCLm, GCLc, SOD1, SOD2, CAT, HO-1ROS → KEAP1[64]
Physical exerciseYoung human malesCycling; 30 min at 70% VO2max OR 7 cycles at 90% VO2maxPeripheral blood mononuclear cellDirectly after PE↑ NRF2
Intensity had no effect
Cycling for 7 cycles ↑ 8-isoprostanes and glutathione reductase in comparison to cycling for 30 minNot specified[65]
Physical exerciseMiceMuscle stimulation on the right leg, high intensity OR low intensityGastrocnemius and soleus muscles30 min after muscle stimulationHigh intensity: local and systemic ↑NRF2
Low intensity: systemic ↓ NRF2 local ↑ NRF2
↔ glutathione
↑ NQO1
KEAP1[105]
Physical exerciseMiceEndurance exercise (EES): 90 min/per day for 2 days OR moderate exercise training (MET): 50 min/day for 6 weeksHeartDirectly after PEEES: ↑ NRF2 in young mice
Prolonged MET: ↑ NRF2 in aging mice
MET: NQO1, HO-1, and GSR were similar in young and old miceNot specified[106]
TMSIn vitroOne session of
10 series at 5, 10, and 15 Hz
THP-1-derived macrophages4 h, 6 h, and 24 hTime-dependent ↑ NRF25 min treatment: ↔ inflammatory factors
Pre-treatment: ↓ IL-1β and TNF-β
KEAP1
p62
[44]
THIn vitro8 h at 37 °C or 32 °CHepG2-ARE stable cellsDirectly after TH↓ antioxidant genes after treatment with THLower temperatures (27 °C and 22 °C): did not activate NRF2 and HIF1A pathways as efficiently as mild hypothermiaPossibly post-translational mechanism[107]
DRRatsAcute: single diving session
Chronic: daily diving sessions for 4 weeks
Brain, kidney, lungDirectly after DR↑ NRF2 phosphorylation and nuclear translocation↑ GSH/GSSG
↑ SOD, ↑ HO-1, ↑ NQO1, ↓ 4HNE, ↓ nitrotyrosine,
↔ MDA
CGRP→
KEAP1/p62,
AMPK,
SIRT1, PI3K
[66]
RIPreCRats1 session of 60 min ischemia followed by 60 min reperfusionSkin tissueAfter flap surgery↑ NRF2Viable flap area was smaller in groups with serum transfer than those w/o RICNot specified[108]
PBMIn vitroBisphenol A (BPA) + photobiomodulation (PHT) wavelength: 660 + 10, output power: 35, energy density (J/cm2): 0.28, for 12 minADSCsDirectly after PBM↑ NRF2Low concentrations of BPA + PHT: induced autophagyp62[67]
PBMIn vitro100 mW, wavelength of 808 nm, spot area of 0.5 cm2, and irradiation at 1, 2, and 3 J/cm2s
OR PBM + metformin
HPDLSCsDirectly after PBM3 J/cm2 PBM + metformin: ↑ NRF2. Was more than 2 J/cm2 PBM + metformin3 J/cm2 PBM: ↑ PIK3
3 J/cm2 PBM + metformin: ↓ TNF-α
KEAP1[68]
PBMIn vitroBlue light-500 mW/cm2A431 epidermoid carcinoma cellsDirectly after PBM↑ NRF2Average volume of light-treated tumors was significantly lower than that in the untreated controlsNot specified[109]
PBMIn vitro250 mW, 500 mW, 1000 mW for 30 s/8 h for 12 daysSchwann cellsDirectly after PBM↑ NRF2250 mW and 500 Mw: ↓ apoptosisPI3K/Akt signaling pathway[110]
Dietary change—CRRats24 mo old with lifelong 40% CRCMVECsAfter establishment of CMVECs↑ NRF2↓ age-related impairment of angiogenic processesmiR-144[103]
Intermittent fastingMiceMice fasted for 24 hSkeletal gastrocnemius muscleAfter fasting↑ NRF2↑ Cat, Gclc, Gclm, Gsr, HO-1 and Ucp3, GPX4Not specified[111]
Intermittent fastingHumansFasted during Ramadan. Daily fasting between 23–30 daysBlood sampleDirectly before RIF and after Ramadan↑ NRF2↑ SOD2, TFAM ↓ SIRT3ROS production[69]
Intermittent fastingMice, WT and Tlr4-/-Food deprived for 24 h every other day, for 30 daysHippocampusAfter regimenTlr4-/-: ↓ NRF2
Tlr4-/- + IF:
↓ NRF2
Tlr4-/- + IF:
↓ memory, depressive-like behavior
Not specified[71]
Intermittent fastingMice with ZEB1 or ZEB2 knockoutDeprived of food but free access to water for 3 days ZEB1-/- + IF: ↓ NRF2
ZEB12-/- + IF: ↑ NRF2
Not specified[70]
Table 2. Non-pharmacological NRF2 interventions investigated in chronic pathological states.
Table 2. Non-pharmacological NRF2 interventions investigated in chronic pathological states.
ModelIntervention TypeSpeciesIntervention ParametersFocal Organ/CellNRF2 Measurement TimingNRF2 ModulationOther EffectsMechanismRef.
HFD-induced non-alcoholic fatty liver disease (NAFLD)Physical exerciseZebrafishPlaced in swimming tunnel. Exercised for 5 days per weekLiverFresh liver tissue samples↑ NRF2↓ oxidative stress and apoptosisSIRT1/AMPK signaling[43]
HemiparkinsonismPhysical exercise pre-treatmentMiceTreadmill, 6 weeks, 5 times/week, time-out on weekendsStriatumDirectly after hemiparkinsonism was inducedPE: ↑ NRF2↓ nigrostriatal neurodegeneration, functional impairment, and supersensitivity of DA receptorsNot specified[92]
ParkinsonismPhysical exercise pre-treatmentRatsTreadmill, at 70% of maximal oxygen consumption for 60 min/day, 5 days/week for 4 weeksStriatum, substantia nigraDirectly after parkinsonism was induced↑ NRF2↔ glutathione or ratio of GSSGROS production[90]
Physical exerciseRatsAfter induction of PD. Treadmill, 30 min/day, 5 times a week for 4 weeksStriatumAfter behavioral tests↑ NRF2↑ NQO1 and TFAMNot specified[88]
A53T α-syn, related to PDEAA53T mice4 weeks starting at 2 months of age at the 2t36 and SP6 acupoints. Intensity increased stepwiseBrain—midbrain and striatum ↑ NRF2↑ HO-1 and glutamate-cysteine ligase modifier subunitsNot specified[91]
Heart failure with preserved ejection fraction (HFpEF)VNS (non-invasive)Rats20 Hz, 0.2 ms, 2 mA, daily for 30 min over a 4-week periodSubfornical organ, spinal trigeminal nucleusAfter VNS↑ NRF2↑ NQO1Not specified[112]
Huntington’s diseaseTMSRats60 Hz, 0.7 mT, 2 h in the morning and 2 h in the afternoon, 8 consecutive days, starting 4 days before the first injection of 3-NPStriatumAfter ELFEF↑ NRF2 Not specified[95]
Vascular dementiaTMSRats10 days, 10 Hz for 2 h per dayBrain tissueAfter TMS↑ NRF2↑ GPx4 and learning memory abilityNot specified[47]
EARatsThird day after 2VO. Administered at DU20 and ST36 acupoints for two weeksHippocampusAfter EA↑ NRF2↓ microglia activation and cognitive deficitsNot specified[94]
RIPreCMiceFour 10-min cyclesBrainAfter RIPreC↑ NRF2↑ glutathione reductaseNot specified[93]
SenescenceTMSSAMP89 mice25 Hz for durations of 14 and 28 daysHippocampus ↑ NRF2↓ MDA, ↑ GPX4Not specified[46]
Dietary change—olive oil phenolicsSAMP8 miceAte diet with high or low amounts of olive oil phenolics for 4.5 monthsHeart High olive oil phenolics:
↑ NRF2
High olive oil phenolics: ↑ GST, γ-GCS, NQO1, and PON2 mRNA lSIRT1[101]
Dietary change—Med dietHumans4 weeks.
Med diet, Med diet with CoQ (Med + CoQ), Western diet rich with saturated fats (SFA)
Blood samples—peripheral blood mononuclear cells4 h after dietMed + CoQ diet: ↑ cytoplasmic NRF2, ↓ nuclear NRF2
SFA diet: ↓ cytoplasmic NRF2, ↑ nuclear NRF2
Med diet: intermediate effects
↓ SOD1 and SOD2, TrxR, NADPH-oxidase (p22phox and p47phox subunits)KEAP1[102]
Physical exerciseRatsTreadmill exercise. 5 days/week for 6 weeksRenal proximal tubules48 h after PE↑ NRF2↓ MDA, CRP
↑ SOD1, IL-10
Not specified[89]
CUMSTMSRats15 Hz for 15 min for 7 consecutive daysHippocampusAfter TMS↑ NRF2↓ depressive and anxiety-like behaviorNot specified[96]
EA pre-treatmentRats1 h before CUMS protocol. Administered at GV23 and GV16 acupoints, every other day for 4 weeksHippocampusAfter inducing CUMS↑NRF2↓depressive behaviors, oxidative stress, and MDANot specified[97]
ECTRatsOnce daily for 10 days at 100 Hz for 0.5 s at 80 mAHippocampusAfter ECTECT: ↑ NRF2ECT: ↓ depressive-like behaviors and hippocampal neuronal ferroptosisBDNF[98]
Enhanced single prolonged stress (ESPS)–PTSDEA pre-treatmentRatsGiven at GV20 acupoint for 30 min daily (frequency: 2/15 Hz, intensity: 1 mA)Hippocampus14 days after ESPS↑ NRF2↑ HO-1, BDNF, AMPK, hippocampal neurogenesis
↓ anxiety-like behaviors
KEAP1[99]
Sleep deprivation (SD)TMS–cTBSMice600 pulses of 3 stimuli of 40 s for 7 sessionsHippocampusAfter cTBS↑ NRF2↑ spatial learning and memory abilities
↓ oxidative stress, inflammation, and autophagy of hippocampal tissues
Not specified[113]
SepsisEARatsGiven at ST36 0.5 mA and 15 Hz for 30 min once daily for five daysHippocampusAfter EA↑ NRF2EA: ↓ MDAα7nAChR[87]
THRats10 h. Maintained at 32–33.9 °CLungs5 days after model is induced↑ NRF2TH: ↑ GPX4 via the KEAP1/NRF2/SLC7A11 signaling pathwayPI3K/Akt/GSK3β signaling pathway[114]
Diabetic encephalopathyEARats30 min alternately at ST36 and EXB3 acupoints once a day for 4 weeksHippocampusAfter EA↑ NRF2↑ HO-1
↑ learning and memory abilities
Not specified[84]
Complex regional pain syndrome type-IEARatsGiven at ST36 and BL60 acupoints on a daily basis for 7 daysHind pawAfter EA↑ NRF2↓ mechanical pain responseNot specified[85]
Ventilator-induced lung injury (VILI)EAMiceGiven at BL13 and ST36 acupoints five times a week for 2 weeksLungAfter EA↑ NRF2↑ HO-1. ↓ activation of NLRP3 inflammasomeNot specified[115]
CarcinogenesisDietary change—CRMiceCR mice were fed 40% less than controlLiverAfter carcinogenesisCR required NRF2 for protection against induced tumorsCR: ↑ HO-1, GCLC, GST A1, and GPx-1Not specified[104]
ObesityDietary change—diets with virgin olive oil (VOO)HumansEach group ingested one breakfast every 2 weeks, until completing the four breakfastsBlood samples—peripheral blood mononuclear cellsAfter 12 h fasting and at 2 and 4 h after ingestion of each breakfast4 h after VOO, SOX, or SOP: ↓ NRF2
4 h after SFO: ↑ NRF2
4 h after VOO: ↓ SOD1, GPx
4 h after SOX: ↓ GPx
4 h after SFO: ↑ NRF2
GSH ↑ higher with VOO, SOX, or SOP in comparison to SFO
Not specified[116]
Intermittent fastingHumansRamadan fasting periods—28 to 30 consecutive days abstaining from food and drink from dawn to sunsetBlood samples2–7 days before fasting and after RamadanIF: ↑ NRF2IF: ↑ SOD2, TFAM, CD163Not specified[117]
Adriamycin (ADR)-induced nephropathyIntermittent fastingRatsAlternate 24-h fasting and feeding periods for 8 weeksKidneyAfter regimen↑ NRF2
↑ SIRT1, HO-1,
↑ AQP2
Not specified[118]
The mechanistic basis for the largely beneficial non-pharmacological NRF2 activation in chronic cerebral dysfunction involves predominantly non-electrophilic pathways operating through both KEAP1-dependent and KEAP1-independent mechanisms. Physical exercise exhibits particularly diverse mechanistic profiles. In high-fat diet zebrafish models, swimming exercise upregulates SIRT1 expression and AMPK phosphorylation, subsequently enhancing p-AKT (protein kinase B) and NRF2 expression through non-electrophilic pathways involving NRF2 nuclear translocation and post-translational modifications [43], effectively attenuating hepatic lipid accumulation and steatosis. Conversely, in 1-methyl-4-phenylpyridine-induced parkinsonian rat models, striatal NRF2 activation occurs via exercise-induced mild oxidative stress, preserving nigrostriatal dopaminergic neurons [90]. This suggests concurrent electrophilic and non-electrophilic mechanisms, providing therapeutic benefit without electrophile-associated cytotoxicity. Electroacupuncture and Mediterranean dietary patterns demonstrate multi-mechanistic NRF2 activation in moderate cerebral dysfunction. Electroacupuncture activates NRF2 through α7nAChR in sepsis models [87] and via KEAP1 modulation in PTSD [91]. Mediterranean diets enhance KEAP1/NRF2 dissociation and SIRT1 activation in senescence models [101,102]. In contrast, electroconvulsive therapy and therapeutic hypothermia demonstrate singular mechanistic pathways in moderate cerebral dysfunction: electroconvulsive therapy modulates brain-derived neurotrophic factor (BDNF) in chronic unpredictable mild stress models [98,102], while TH activates the survival-mediated PI3K/Akt/GSK3β signaling cascade. Although additional interventions demonstrate efficacy in chronic disease contexts, their mechanistic characterization remains incomplete.

2.4. Non-Pharmacological NRF2 Activation in Acute Oxidative Stress Paradigms

Consistent with observations in chronic oxidative stress conditions, electrophilic activators demonstrate substantial translational limitations in acute, high-oxidative stress paradigms, contrasting markedly with the therapeutic benefits of non-pharmacological NRF2 activators. Sulforaphane administration in middle cerebral artery occlusion (MCAO) ischemia/reperfusion models produces a 1.3-fold increase in protein oxidation relative to vehicle controls [6]. Despite concomitant 2.5-fold HO-1 upregulation, lesion volume reduction reaches only 50% [6], compared to 70% reduction achieved with glutathione administration [7]. This phenomenon extends beyond sulforaphane; in stroke models, DMF fails to consistently reduce lesion volume or oxidative stress, potentially exacerbating lesion damage within three days of administration [13,14,15]. These findings indicate that electrophilic-activator-mediated NRF2 induction and subsequent antioxidant upregulation, including glutathione synthesis, yield limited therapeutic benefit under acute oxidative stress conditions. Furthermore, despite NRF2’s established role in oxidative stress and inflammatory modulation, DMF administration in traumatic brain injury models provides neither antioxidative nor anti-inflammatory benefits [16,17], paradoxically increasing cerebral lipid peroxidation [66]. Given the brain’s high lipid content, this effect poses particular neurotoxicological risks [25].
Conversely, non-pharmacological interventions demonstrate robust NRF2 activation efficacy in acute/traumatic pathologies characterized by rapid oxidative stress escalation, including ischemia/reperfusion (I/R) injury (cutaneous, hepatic, myocardial, pulmonary, retinal), cerebral ischemia, traumatic brain injury, intracerebral and subarachnoid hemorrhage, hemorrhagic shock, cardiac arrest/hypoxia, acute lung injury, and acute inflammation (Table 3, Figure 3). This therapeutic efficacy persists despite heightened stress sensitivity and increased probability of contraindications or diminished effectiveness in these contexts. Remote ischemic conditioning (RIC) represents a predominant intervention for I/R injury mitigation. Remote ischemic post-conditioning (RIPostC) activates NRF2 across cerebral, myocardial, renal, and retinal I/R models, while remote ischemic pre-conditioning (RIPreC) demonstrates age-dependent effects, upregulating NRF2 in young but not aged hearts following myocardial I/R injury [119,120,121,122,123,124]. Additional NRF2-mediated I/R protective interventions include: pre-ischemic exercise reducing stroke severity, therapeutic hypothermia attenuating cerebral cell death, electroacupuncture upregulating pulmonary HO-1 in pulmonary and cerebral I/R, olive oil dietary supplementation enhancing wound healing post cutaneous I/R, and combined vagus nerve stimulation with therapeutic hypothermia increasing hepatic antioxidant enzyme expression [125,126,127,128,129,130,131]. Following traumatic cardiac injury, secondary organ damage is ameliorated through NRF2 activation via RIPreC, therapeutic hypothermia (4–24 h), non-invasive vagus nerve stimulation, electroacupuncture pretreatment, and PBM [112,132,133,134,135,136,137,138]. Plasma-mediated effects of RIC, PBM, and dietary modifications demonstrate systemic NRF2 activation, reducing acute inflammatory responses in transfer experiments [139,140,141]. In traumatic brain injury, therapeutic hypothermia and median nerve stimulation enhance NRF2 expression, ameliorating cognitive deficits [142,143]. Intermittent fasting upregulates NRF2 following intracerebral hemorrhage and nephropathy [118,144]. Additionally, RIPreC attenuates neurological dysfunction in hemorrhagic shock through NRF2 activation [145], while acute pulmonary injury is modulated by electroacupuncture and therapeutic hypothermia-induced NRF2 expression [115,146].
Table 3. Non-pharmacological NRF2 interventions investigated in acute injury and traumatic pathologies.
Table 3. Non-pharmacological NRF2 interventions investigated in acute injury and traumatic pathologies.
ModelIntervention TypeSpeciesIntervention ParametersFocal Organ/CellNRF2 Measurement TimingNRF2 ModulationOther EffectsMechanismRef.
Cerebral
ischemia—MCAO
Physical exerciseRats30-min treadmill training with either Constraint-induced movement therapy (CIMT) OR unconstrained exercise (UE)BrainDirectly after PECIMT in comparison to UE: ↑ NRF2CIMT in comparison to UE: ↓ MDAKEAP1[147]
Physical exercise—pre-conditionalMiceHoused for 6 weeks in a cage with a running wheelBrain—microgliaDirectly after PEPE: ↑ NRF2PE w/o stroke:
↔ CB2R, P2Y12, mafk, and p-NRF2
PE w/stroke:
↑ CB2R, P2Y12, mafk, and p-NRF2
CB2R[125]
EARats30 min for 7 days; continuous 2/100 Hz, ~2–4 V and 0.5~1.5 mA at the PC6, DU26, SP6, and DU20 acupointsBrain—cerebral cortexDirectly after EA↑ NRF2↑GPX4 and SLC7A11,
↓neuronal damage and neuronal mitochondrial injury, ↓ iron
Not specified[128]
EA pre-treatmentMouse30 min, 1 mA, 2/15 Hz at the GV20 acupointBrain—cerebral cortex2 h after MCAO↑ NRF2↑ HO-1 and NQO1GSK-3β[42]
THRatsCold condition (4 °C), isolated cortical temperature of 33 ± 1 °C during ischemiaBrain24 h after reperfusion↑ NRF2
↓ neurological deficit and cerebral cell deathPPARs[126]
RIPreCRatsLRIC (3 cycles) was applied every day up to 14 days before MCAOBrain24 h after reperfusion↑ NRF2↑ SOD1 and HO-1Not specified[148]
RIPostCMice4 cycles lasting 40 min in total and continued every 12 h until execution (treated twice daily for 1, 3, or 7 days)Brain—cortexAfter behavioral testing↑ NRF2↑ HO-1, NLRP3, cleaved caspase-1, TAC, SOD, GSH/GSSG levels, neurological function, ↓ MDAKEAP1[119]
RIPostCMice3 cycles immediately after stroke onsetBrain—cerebral cortex24 h after reperfusion↑ NRF2↑ in neurological outcome, HO-1, NQO-1, SOD, ↓ MDANot specified[120]
Subarachnoid hemorrhagePhysical exerciseRatsPre-conditioning exercise, treadmill, 30 min/day, 5 days/week for 3 weeksBrain—motor cortex ↑ NRF2↓ neurological deficits, sensorimotor dysfunction, and consciousness disorderNot specified[149]
Intracerebral hemorrhage (ICH)Intermittent fastingMiceEvery-other-day feedingMicroglia of ipsilateral basal gangliaDays 1, 2Day 1 and 3 after ICH: ↑ NRF2
Day 7: returned to baseline
↓ CD16+Iba-1+ microglia activation, IL-1β, TNF-αSIRT3[144]
I/R on skinDietary change = soybean oil and olive oilMice6 weeks subjected to either regimenBlood samples—proteins of wound lysate14 days after second IR cycle↑ NRF2Promoted wound closure at 7, 10, and 14 daysNot specified[129]
MIRI
Delayed RIPreC (DRIPC)Rats4 cycles once per day for 3 days before heart isolationHeart—left ventricle30 min after stabilized perfusion↑ NRF2↑ HO-1Did not activate PKB/Akt or ERK 1/2[150]
RIPreCRatsFour cycles for a total of 40 minHeart24 h after RIPreC↑ NRF2 in young hearts, ↓ in old hearts HIF-1 α[151]
VNSRatsIntensity: 0.5 V, frequency: 2.5 Hz, pulse width: 5 ms, duration: 5 minHeartAfter I/R↑ NRF2↑ GRPRKEAP1[152]
RIPostCMice3 cycles at the start of reperfusion periodHeartAfter 2 h if cardiac reperfusion↑ NRF2↑ Akt, HO-1, SOD1, ↓ MDASTAT3[121]
Hepatic I/R
THRatsInduced by the superfusion of cooled saline at 26 °C onto the ischemic lobesLiverDirectly after TH↓ NRF2↑ NQO1KEAP1[153]
VNSRats20 Hz for 0.1 millisecondLiverDirectly after VNS↑ NRF2↑ HO-1Not specified[130]
VNSRatsInterval of 1 s, a duration of 1 ms, and a frequency of 5 HzLungAfter 6 h of reperfusion↑ NRF2
↓ MPO and MDA
Not specified[131]
VNSRats20 Hz, 0.2 ms in durationKidneysAfter I/R↑ NRF2↑ HO-1Not specified[154]
Lung I/REARabbits15 min once a day for 5 days at BL13 and ST36 acupointsLungAfter I/R↑ NRF2↑ SOD, GPx, and CAT
↓ MDA
p38 MAPK[127]
Renal I/RRIPostCMice3 cycles of 5 min during the reperfusion periodKidney24 h after reperfusion↑ NRF2↑ HO-1, SOD
↓ MDA
TOPK/Akt[122]
RIPostCRatsBegan on second post-operative day, lasted 10 minRetinaAfter RIPostC↑ NRF2↑ HO-1, ↓ GFAPNot specified[123]
Hemorrhagic shock/resuscitation (S/R)RIPreCMice, zebrafishMice underwent 4 cycles for 10 min. Zebrafish were treated with RIC bloodLiver of mice. Plasma from zebrafishAfter hemorrhage (S/R)RIPreC for mice and zebrafish: ↑ NRF2
KEAP1 and ERK 1/2[145]
Tailfin-cut inflammation
RIPostCZebrafishFish were treated with RIC plasma injection from wildtype mice (4 cycles of 5 min) and NRF2-knockout mice ↑ NRF2↑ hmox1a, ↓ neutrophil migrationROS[139]
Endotoxic-shock-induced acute lung injuryEARabbitsPerformed throughout the operating steps for 6 h during the experimental day at ST36 and BL13 acupointsLungAfter EA↑ NRF2↑ HO-1, SOD, GPx, and CAT, ↓ MDANot specified[146]
Hydrogen-peroxide-induced oxidative stressRIPreCIn vitroHuman umbilical vein endothelial cells (HUVECs) were treated with rat sera. Rats underwent 3 cycles of 10 minHUVECsAfter sera was injected↑ NRF2↓ MDANot specified[155]
Inflammation—stimulation with 2,4-dinitrochlorobenzene (DNCB)PBMIn vitro660 nm (red light) or 520 nm (green light), 20 min after DNCB treatment. Exposure time of 250 sPrimary human KCsAfter PBM↑ NRF2↑ HO-1, NQO1, and GCLCNot specified[141]
DSS-induced acute colitisEAMice30 min at ST36 acupointsColon—macrophagesAfter EA↑ NRF2EA: ↑ NRF2 HO-1, ↓ NLRP3/IL-1β activationNot specified[86]
TBI
MNSRats300 microseconds at 40 Hz for 20 s/min. Continued 8 h per day for 2 weeksHippocampusAfter MNS↑ NRF2
↑ GPX4, SLC7A11, VEGFNot specified[142]
THMice4 h at 37 degrees CelsiusCortex24 h after TBI↑ NRF2↑ HO-1 and NQO-1
↓ MDA
Not specified[143]
Hypothermic circulatory arrest (HCA)RIPreCPiglets4 cycles of 5-min ischemia followed by 5-min reperfusionCortex, hippocampus, thalamus, brainstem, cerebellumAfter RIPreC↑ NRF2↓ 8-OHdGHIF-1-α[133]
Cardiac hypoxiaPBM 8 W/cm2–12 W/cm2 0.15–250 s, varied energy intensityH9C2 cardiomyocytesAfter PBM↑ NRF2↓ SOD2, PGC-1αp62[134]
Cardiac arrestTHRats4 h after return of spontaneous circulationHippocampusAfter TH↑ NRF2↓ MDA, caspase-3 ↑ SODGSK-3β[135]
THMinipigs24 h after ROSC. TH was maintained for 12 hFrontal cortexAfter TH↑ NRF2↓ MDAOxidative stress[136]
THRatsHypothermia was maintained at 33 ± 0.5 °C for four hoursLumber spinal cord—motor neurons and glial-like cells24 h after CA↑ NRF2↑ HO-1Not specified[137]
CA-induced renal I/RTHRatsAdministered for 2, 4, 6 hKidneyAfter TH↑ NRF2, more ↑ with time↑ HO-1, ↓ MDANot specified[138]
Cardiopulmonary bypassEA pre-treatmentRatsEA at PC6 and LI4 acupoints for 30 min before CPBLungAfter EA↑ NRF2↓ pulmonary neutrophil infiltrations
↓ TNF-α, IL-18, and IL-1β
ROS[132]
Limited mechanistic investigations of non-pharmacological NRF2 activation in acute contexts reveal a multi-mechanistic regulatory paradigm. RIPostC demonstrates pathway-specific activation across different pathologies: in myocardial I/R injury, RIPostC induces STAT3 phosphorylation, subsequently activating Akt and upregulating NRF2 expression [121]. Conversely, in renal I/R models, T-LAK-cell-originated protein kinase (TOPK) mediates NRF2 expression, preventing renal dysfunction and tubular necrosis [122]. TOPK phosphorylation inactivates phosphatase and tensin homolog (PTEN), thereby activating PI3K/Akt signaling and NRF2 regulation. In cerebral ischemia, RIPostC downregulates KEAP1, attenuating neuronal damage [119]. RIPreC employs combined mechanisms, inducing ROS and electrophile production for KEAP1 dissociation while modulating HIF-1α in aged myocardium [124], integrating multiple non-electrophilic pathways. Temporal analysis reveals mechanistic complexity: delayed RIPreC effects (24–72 h post-administration) occur independently of PKB/Akt and ERK1/2 pathways, despite documented ERK1/2 dependence in other studies [145,150]. Furthermore, NRF2 activation by sera from RIC-treated rats proceeds independently of MEK/ERK and PI3K/Akt pathways [155], suggesting context-dependent activation mechanisms requiring further investigation. Electroacupuncture integrates multiple non-electrophilic pathways, including GSK-3β downregulation and p38 MAPK upregulation [42,127], concurrent with electrophilic NRF2 activation. This dual mechanism likely explains clinical efficacy without organ toxicity despite electrophilic components [126,135,136,137,138]. Other non-pharmacological interventions demonstrate singular mechanistic pathways in acute/traumatic conditions: intermittent fasting upregulates sirtuin 3 (SIRT3), exercise modulates cannabinoid receptor 2 (CB2R), and PBM increases PGC-1α [125,134,140,144]. Therapeutic hypothermia employs dual mechanisms, modulating PPARs and PI3K/Akt/GSK3β signaling, exemplifying the multi-mechanistic potential of non-pharmacological activators.

3. Discussion and Future Perspectives

3.1. Multi-Mechanistic NRF2 Activation: A Strategy to Circumvent Constraints of Current Electrophilic and Non-Electrophilic Activators

Multi-mechanistic NRF2 activation strategies demonstrate superior therapeutic potential compared to singular pathway approaches. Classical electrophilic activators employ exclusively electrophilic mechanisms [1,2,3,4], whereas interventions including physical exercise, remote ischemic conditioning, intermittent fasting, and electroacupuncture integrate ROS generation with protein–protein interaction inhibition, KEAP1 degradation, protein stability modulation, or post-translational modifications (Figure 3), thereby reducing potential adverse events. Physical exercise exemplifies this multi-mechanistic approach, combining electrophilic ROS production with SIRT1/AMPK signaling modulation [43], where non-electrophilic pathways potentially counterbalance electrophilic toxicity. Therapeutic hypothermia similarly integrates electrophilic ROS generation with PPAR targeting and post-translational modifications [107,126,153].
Non-pharmacological activators inherently employ multiple NRF2-targeting mechanisms, potentially yielding more nuanced or pronounced effects than current pharmacological agents (Figure 3). This suggests that combining interventions with distinct mechanistic profiles could address limitations inherent to both classical electrophilic and non-pharmacological activators. Despite potential cytotoxicity, electrophilic activation remains among the most potent NRF2 activation methods, providing a greater enhancement to NRF2 bioavailability [37,38]. However, electrophilic mechanisms constrain pharmacological intervention utility. Subtherapeutic pharmacological dosing to enhance NRF2 bioavailability [37,38], combined with non-pharmacological activators to improve NRF2 efficiency, presents a viable strategy. This approach shows particular promise when employing interventions like electroacupuncture or dietary restriction, which utilize multiple non-electrophilic pathways, including MAPK/p38, α7nAChR, p62, AMPK, and SIRT1 [87,127].
Non-pharmacological activator combinations may address limitations of both classical electrophilic activators and individual non-pharmacological interventions. Dietary modifications alone produce insufficient sustained NRF2 activation [102], with miR-144 and miR-146a silencing showing limited efficacy [102,156]. However, combination with pharmacological coenzyme Q10 produces synergistic effects exceeding either intervention alone. Similarly, combining dietary interventions with PBM, which targets NRF2 availability through PGC-1α rather than efficiency [134], may yield enhanced NRF2 activation. Multi-factorial approaches thus exemplify the adjunctive potential of non-pharmacological NRF2 activators, combining interventions with complementary mechanisms to overcome individual limitations and optimize therapeutic efficacy. Realizing this potential requires characterizing individual intervention limitations and examining interintervention interactions. Despite these challenges, the therapeutic promise for NRF2 activation and cerebral dysfunction treatment justifies intensive investigation of these multi-mechanistic strategies.

3.2. Cell-Specific Oxidative Stress Vulnerability and NRF2 Utilization: Mechanistic Advantages of Non-Electrophilic Activation Pathways

NRF2 activator efficacy in the brain exhibits cell-type-dependent variability determined by differential cellular interactions. Distinct brain cell populations demonstrate heterogeneous oxidative stress responses and NRF2 pathway utilization, reflecting cell-specific oxidative stress sensitivities (Figure 4) and variable basal intracellular NRF2 concentrations [50]. Therefore, the therapeutic efficacy of NRF2-targeted interventions in cerebral pathology is determined not by absolute NRF2 activation levels but by cell-type-specific responses. This phenomenon becomes particularly relevant under chronic administration or acute/traumatic conditions, where electrophilic compound limitations manifest. Cell-type-specific characterization of non-pharmacological interventions remains limited. Nevertheless, mechanistic profiles and documented downstream antioxidative effects enable prediction of cell-type-specific therapeutic suitability. This analysis systematically examines cerebral cellular populations and their NRF2 utilization patterns, stratified by oxidative stress sensitivity, correlating specific activation mechanisms and antioxidative outcomes with appropriate cellular targets. This framework facilitates identification of pathological contexts where non-pharmacological NRF2 activators demonstrate optimal therapeutic potential, particularly through predominant non-electrophilic pathway activation.

3.2.1. Oligodendrocytes

Oligodendrocytes function as the primary myelinating cells of the central nervous system, essential for myelination, remyelination processes, and metabolic efficiency [157]. Among cerebral cell populations, oligodendrocyte precursor cells and mature oligodendrocytes demonstrate maximal oxidative stress susceptibility, exceeding that of cortical neurons and astrocytes [158] (Figure 4). This heightened vulnerability results from elevated ROS generation, increased metabolic demands, and enhanced lipid peroxidation susceptibility [157]. Consequently, oligodendrocytes exhibit pronounced NRF2 dependence, maintaining the highest basal NRF2 expression among cerebral cell types [159]. NRF2-mediated glutathione synthesis prevents metabolic cell death [160], ameliorating mitochondrial dysfunction and endoplasmic reticulum stress [161], while promoting mitochondrial biogenesis through PGC-1α activation [162]. Therefore, glutathione depletion by electrophilic activators [9] poses particular risks to oligodendrocyte viability.
The critical role of PGC-1α specifically implicates PBM, dietary restriction, dietary modifications, and physical exercise as therapeutic interventions. PBM directly upregulates PGC-1α [134], while the latter three modulate AMPK and/or SIRT1, essential regulators of PGC-1α biosynthesis [163]. These interventions, particularly those exhibiting dual AMPK/SIRT1 modulation, demonstrate enhanced potential for mediating oligodendrocyte oxidative stress resistance. This suggests therapeutic applicability in neurodegenerative pathologies including multiple sclerosis, Alzheimer’s disease, and Parkinson’s disease, where oxidative-stress-induced oligodendrocyte precursor cell differentiation failure impairs remyelination and adaptive myelination [164]. The chronic nature of these pathologies particularly favors non-electrophilic non-pharmacological NRF2 activators over electrophilic compounds, given the documented drawbacks of electrophilic activators in chronic administration [74,76].

3.2.2. Neurons

Neuronal death in cerebral dysfunction occurs predominantly through oxidative-stress-mediated excitotoxicity, apoptosis, and ferroptosis [50]. Neurons exhibit the second-highest oxidative stress vulnerability among brain cells (Figure 4). Neurons demonstrate exceptional mitochondrial dependence for bioenergetic requirements, exceeding other cell types. Consequently, oxidative-stress-induced mitochondrial damage significantly contributes to neuronal death, suggesting heightened vulnerability to electrophilic activators that disrupt mitochondrial bioenergetics [1,2,3,4,5,6,7,20,21,22,23,24,26,27,28,29,30]. Oxidative stress induces mitochondrial membrane hyperpermeabilization [26,27,28,29], initiating caspase-dependent apoptosis through ROS release, resulting in DNA fragmentation and cellular death [30]. Lipid peroxidation products, particularly 4-hydroxynonenal [22,23,24], generated through glutathione peroxidase inhibition and glutathione depletion [20,21], exacerbate this process—representing key mechanisms of electrophilic activator toxicity, including sulforaphane [165].
Therapeutic targeting of mitochondrial oxidative stress and lipid peroxidation represents optimal neuroprotective strategy. This requires selective upregulation of SOD2 for mitochondrial ROS scavenging and HO-1 and GPX4 for ferroptosis inhibition [166]. Additionally, PGC-1α modulation provides therapeutic benefit through mitochondrial biogenesis regulation [163]. Among interventions enhancing PGC-1α synthesis, dietary restriction and physical exercise, which concurrently modulate HO-1, glutathione, and/or SOD2, demonstrate superior neuroprotective potential [64,66]. These approaches show particular relevance in neurodegenerative pathologies including Alzheimer’s and Parkinson’s diseases, characterized by oxidative-stress-mediated neuronal dysfunction [60,167,168]. Physical exercise’s efficacy in Parkinson’s disease exemplifies this principle, where multi-mechanistic NRF2 activation neutralizes potential electrophilic activation-associated toxicity [88,90,92].

3.2.3. Pericytes and Endothelial Cells

Pericytes and endothelial cells constitute critical components of cerebral microvasculature, BBB integrity, and angiogenic processes [169] (Figure 4). Pericytes demonstrate temporal-dependent oxidative stress sensitivity: acute exposure induces relaxation, 30-min exposure triggers constriction, and prolonged exposure results in cellular death [170]. Endothelial cells similarly exhibit heightened oxidative stress vulnerability, manifesting as disrupted intracellular signaling, compromised nitric oxide synthesis, and increased vascular permeability [171,172,173,174]. NRF2 directly mediates endothelial integrity and BBB protection, preventing post-cerebral-insult leukocyte infiltration through HO-1-dependent mechanisms that inhibit endothelial apoptosis [27,173]. In pericyte–endothelial interactions, NRF2 promotes angiogenesis via BDNF upregulation [28], an effect replicated by electroacupuncture [99] and electroconvulsive therapy [98,102]. Additionally, NRF2 regulates pericyte stemness through nestin and E-cadherin mRNA upregulation, facilitating pericyte differentiation [175].
Paradoxically, DMF increases leukocyte infiltration in experimental traumatic brain injury, suggesting BBB dysfunction and endothelial/pericyte damage despite NRF2 upregulation [16]. Sulforaphane demonstrates dose-dependent inhibition of endothelial and pericyte proliferation, suppressing tube formation [176] and disrupting intercellular communication through angiogenic factor inhibition, indicating NRF2 pathway interference. Conversely, the NRF2–E-cadherin–pericyte differentiation axis suggests electroacupuncture therapeutic potential, given its documented NRF2 activation [99] and E-cadherin/GSK-3β upregulation [177]. Oxidative-stress-induced pericyte contraction dysregulates cerebral blood flow, exacerbating hypoperfusion [178]. The Wnt signaling involvement particularly implicates electroacupuncture efficacy, given its pathway-mediated effects [177,179]. Electroacupuncture’s mechanistic profile, integrating Wnt/β-catenin, E-cadherin, and GSK-3β modulation [99,177,179], suggests specific efficacy in vascular degenerative pathologies including vascular cognitive impairment and dementia, characterized by microvascular loss requiring regenerative intervention [180]. Similarly, dietary restriction’s beneficial effects in vascular dementia models, including pericyte preservation, angiogenesis, and vascular maintenance [180], may result from potent non-electrophilic NRF2 activation [66].

3.2.4. Microglia

Microglia function as the brain’s resident immune cells, mediating tissue repair, neurogenesis, soluble factor release, synaptic remodeling, and phagocytosis [181]. Microglial cells maintain higher NRF2 expression than neurons, exhibiting enhanced ARE activity and elevated NRF2 transcript levels, with functionality predominantly oriented toward inflammatory modulation [181,182,183]. In microglia, NRF2 inhibits NFκB signaling, modulating innate and adaptive immune responses while suppressing proinflammatory cytokine and chemokine release (Figure 4). This mechanism prevents NLRP3 inflammasome assembly, disrupting inflammatory cascades and inhibiting pyroptotic cell death [184]. NRF2-mediated NADPH oxidase 4 (NOX4) blockade specifically prevents NLRP3 formation [185,186,187,188]. This pathway particularly implicates Mediterranean dietary patterns in microglial NRF2 modulation; elevated polyphenol intake downregulates NOX via NRF2, attenuating oxidative stress and preventing inflammatory accumulation [102]. Furthermore, downstream effectors HO-1 and NQO1 specifically reduce NLRP3 formation and caspase-1 expression, suppressing inflammatory cytokine release [189]. These mechanisms suggest that interventions including electroacupuncture [42], vagus nerve stimulation [112], and dietary restriction [66] represent appropriate therapeutic strategies for neuroinflammatory conditions including multiple sclerosis, encephalitis, meningitis, and acute trauma-induced inflammation [190,191,192].

3.2.5. Astrocytes

Astrocytes represent the predominant glial cell population within the brain, executing diverse supportive functions including nutrient transport, BBB maintenance, ionic homeostasis, and tissue repair processes [193] (Figure 4). Under controlled culture conditions, astrocytes demonstrate enhanced NRF2 expression and superior oxidative stress resilience, mediating protective responses against oxidative insults [194]. During oxidative stress exposure, astrocytes upregulate antioxidant molecules including HO-1 and glutathione, facilitating protective neuronal–astrocytic intercellular communication [50]. Astrocytic NRF2 overexpression potentially maintains neuronal metabolic homeostasis following mitochondrial complex II inhibition and glutamatergic excitation, mediating HO-1 upregulation while suppressing ferroptosis through SOD1 and GPX4 enhancement. Therefore, astrocyte-targeted therapeutic strategies may confer neuroprotection through paracrine mechanisms. Interventions selectively upregulating HO-1 and SOD1 demonstrate preferential astrocytic targeting, including physical exercise [64] and remote ischemic conditioning [148]. Astrocyte-focused therapeutic approaches demonstrate particular relevance in acute traumatic pathologies, where neuronal oxidative stress generation resulting from bioenergetic dysfunction represents a critical pathophysiological mechanism [195].

3.3. Therapeutic Applications of Non-Pharmacological NRF2 Activators in Neurodegenerative Pathologies and Psychological Disorders

Current therapeutic strategies for neurodegenerative diseases remain predominantly symptomatic, highlighting the critical need for disease-modifying interventions. Oxidative stress represents a fundamental pathogenic mechanism across neurodegenerative disorders [60,167,168]. In Parkinson’s disease, α-synuclein oligomer aggregation induces cytotoxicity, ferroptosis, and mitochondrial ROS generation, resulting in dopaminergic neuronal depletion [196,197,198,199]. In amyotrophic lateral sclerosis, by contrast, excessive ROS generation, arising from compromised antioxidant defenses and pathogenic mutations, serves as a principal mechanism driving motor neuron degeneration [200,201]. Similarly, in Alzheimer’s disease pathogenesis, β-amyloid peptide aggregates interact with mitochondria, compromising mitochondrial function and enhancing ROS production [202,203,204], initiating progressive neuronal death cascades [205,206]. Tau accumulation, in tauopathies, leads to similar structural and metabolic neuronal and microenvironmental compromise, by disrupting physiological microtubule function [207,208,209,210], all the way to mitochondrial impairment.
Comparable oxidative stress mechanisms characterize Huntington’s disease and multiple sclerosis pathophysiology, suggesting conserved pathogenic pathways across neurodegenerative conditions [168]. The pivotal role of ROS implicates dysregulated NRF2 signaling in disease progression [167], indicating that NRF2 upregulation and subsequent antioxidant pathway enhancement may confer neuroprotection. However, clinical application of pharmacological NRF2 activators remains constrained, necessitating alternative therapeutic approaches. Non-pharmacological NRF2 activators targeting oxidative stress mechanisms may provide preventive benefits not yet comprehensively characterized. These interventions demonstrate sustained efficacy under chronic administration without the organ toxicity and mitochondrial dysfunction associated with pharmacological agents, supporting their application as long-term prophylactic strategies. Pre-clinical evidence substantiates the potential therapeutic efficacy, in neurodegenerative conditions, for several interventions, including TMS [47], electroacupuncture [94], remote ischemic pre-conditioning [93], and DR [180] in vascular dementia; TMS in Huntington’s disease [95]; and physical exercise in Parkinson’s disease [88,90,92].
Non-pharmacological NRF2 activators also demonstrate therapeutic potential extending beyond neurodegenerative pathologies to encompass psychological disorders. NRF2 functions as an upstream regulator of critical factors implicated in psychological dysfunction pathogenesis [211], including oxidative stress, neurotransmitter systems targeted by conventional psychotropic medications, apoptosis, and neuroinflammation. This multi-factorial modulation positions NRF2 as a promising therapeutic target for psychological disorders [177], as demonstrated by pharmacological interventions including CDDO-Im in post-stroke depression models [177] and TP-500 in intracerebral hemorrhage models [212]. Correspondingly, TMS and electroconvulsive therapy demonstrate preliminary efficacy in attenuating depressive and anxiety-like behaviors in chronic unpredictable mild stress models through hippocampal NRF2 upregulation [96,98]. Electroacupuncture pre-treatment in enhanced single prolonged stress models of post-traumatic stress disorder, modulating AMPK, BDNF, and KEAP1, upregulates NRF2 activation and HO-1 expression, correlating with reduced anxiety manifestations [99]. These findings suggest that additional non-pharmacological interventions enhancing cerebral NRF2 expression may demonstrate efficacy in psychological disorders, including dietary restriction (increasing brain-wide NRF2 activation, particularly hippocampal) [66], vagus nerve stimulation (enhancing NRF2 and NAD(P)H:quinone oxidoreductase expression in the subfornical organ and spinal trigeminal nucleus) [112], and remote ischemic pre-conditioning [93]. Given psychological dysfunction prevalence affecting 5–15% of the global population and 30–50% of individuals with cerebral pathology [211], novel interventions targeting fundamental pathophysiological mechanisms warrant prioritized investigation, particularly considering their demonstrated potential in acute cerebral injury contexts.

4. Conclusions

This review addresses two central topics: (i) the paradox that classical electrophilic NRF2 activators can be less effective than individual downstream antioxidants—and may, under certain conditions, exacerbate pathology—and (ii) the prospect that non-pharmacological interventions provide a viable solution. A systematic evaluation of the literature reveals that non-canonical, non-electrophilic pathways circumvent the shortcomings of electrophilic agents, particularly their limited neuroprotective efficacy, especially when combined in a multi-mechanistic manner. We delineate the multi-mechanistic activation profiles characteristic of non-pharmacological modalities, identifying this mechanistic plurality as critical to their therapeutic capacity. The combination of non-electrophilic action and pathway multiplicity affords cell-type-specific regulation, enabling precision strategies across a broad spectrum of neurological disorders. Furthermore, the convergence of multiple activation mechanisms lays the groundwork for combinatorial therapeutic approaches that surpass the constraints of traditional electrophilic compounds and single-mechanism non-pharmacological tactics. Collectively, this evidence underscores the promise of non-electrophilic, multi-mechanistic, non-pharmacological NRF2 activators and outlines essential research directions to advance their clinical translation.

Author Contributions

Conceptualization, C.L. (Chunyan Li); investigation, K.P., L.G., C.L. (Christopher LeDoux), and C.L. (Chunyan Li); writing—original draft preparation, C.L. (Chunyan Li) and K.P.; writing—review and editing, L.G., C.d., D.S., Y.A.A., and C.L. (Chunyan Li); supervision, C.L. (Chunyan Li); project administration, C.L. (Chunyan Li); funding acquisition, C.L. (Chunyan Li). All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the United States Army Medical Research Acquisition Activity (USAMRAA) under award #HT9425-24-1-1017.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data was generated.

Acknowledgments

The authors would like to acknowledge the contributions of Julia Jinu, Steven Wadolowski, and Willians Tambo, who aided in the acquisition of information for this literature review.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ADSCAdipose-tissue-derived stem cell
α7nACHrAlpha7 nicotinic acetylcholine receptor
ADAlzheimer’s disease
AMPKAMP-activated protein kinase
AREAntioxidant response element
BDNFBrain-derived neurotrophic factor
CRCaloric restriction
CB2RCannabinoid receptor 2
CACardiac arrest
CMVECsCerebromicrovascular endothelial cells
CUMSChronic unpredictable mild stress
cTBSContinuous theta burst stimulation
CRPC-reactive protein
DMFDimethyl fumarate
DRDiving reflex
EAElectropuncture
ECTElectroconvulsive therapy
ERKExtracellular signal-regulated kinase
GPX4Glutathione peroxidase 4
GSK3βGlycogen synthase kinase 3 beta
HO-1Heme oxygenase-1
HFDHigh-fat diet
I/RIschemia/reperfusion
KEAP1Kelch-like ECH-associated protein 1
MDAMalondialdehyde
MNSMedian nerve stimulation
MCAOMiddle cerebral artery occlusion
MIRIMyocardial ischemia–reperfusion injury
NQO1NAD(P)H:quinone oxidoreductase 1
NRF2Nuclear factor erythroid 2-related factor 2
PDParkinson’s disease
PI3KPhosphoinositide-3-kinase
PBMPhotobiomodulation
PTSDPost-traumatic stress disorder
AktProtein kinase B
p38 MAPKp38 mitogen-activated protein kinase
ROSReactive oxygen species
RICRemote ischemic conditioning
RIPreCRemote ischemic pre-conditioning
RIPostCRemote ischemic post-conditioning
STAT3Signal transducer and activator of transcription 3
SIRT1Sirtuin 1
SIRT3Sirtuin 3
SOD1Superoxide dismutase 1
SOD2Superoxide dismutase 2
THTherapeutic hypothermia
TOPKT-LAK cell-originated protein kinase
TMSTranscranial magnetic stimulation
TBITraumatic brain injury
VNSVagus nerve stimulation

References

  1. Johnson, J.A.; Johnson, D.A.; Kraft, A.D.; Calkins, M.J.; Jakel, R.J.; Vargas, M.R.; Chen, P.-C. The Nrf2-ARE Pathway: An Indicator and Modulator of Oxidative Stress in Neurodegeneration. Ann. N. Y. Acad. Sci. 2008, 1147, 61–69. [Google Scholar] [CrossRef]
  2. Lee, J.-M.; Calkins, M.J.; Chan, K.; Kan, Y.W.; Johnson, J.A. Identification of the NF-E2-Related Factor-2-Dependent Genes Conferring Protection against Oxidative Stress in Primary Cortical Astrocytes Using Oligonucleotide Microarray Analysis. J. Biol. Chem. 2003, 278, 12029–12038. [Google Scholar] [CrossRef]
  3. Shih, A.Y.; Johnson, D.A.; Wong, G.; Kraft, A.D.; Jiang, L.; Erb, H.; Johnson, J.A.; Murphy, T.H. Coordinate Regulation of Glutathione Biosynthesis and Release by Nrf2-Expressing Glia Potently Protects Neurons from Oxidative Stress. J. Neurosci. 2003, 23, 3394–3406. [Google Scholar] [CrossRef] [PubMed]
  4. Gan, L.; Johnson, J.A. Oxidative Damage and the Nrf2-ARE Pathway in Neurodegenerative Diseases. Biochim. Biophys. Acta (BBA)—Mol. Basis Dis. 2014, 1842, 1208–1218. [Google Scholar] [CrossRef]
  5. Navarro, E.; Esteras, N. Multitarget Effects of Nrf2 Signalling in the Brain: Common and Specific Functions in Different Cell Types. Antioxidants 2024, 13, 1502. [Google Scholar] [CrossRef] [PubMed]
  6. Alfieri, A.; Srivastava, S.; Siow, R.C.M.; Cash, D.; Modo, M.; Duchen, M.R.; Fraser, P.A.; Williams, S.C.R.; Mann, G.E. Sulforaphane Preconditioning of the Nrf2/HO-1 Defense Pathway Protects the Cerebral Vasculature against Blood–Brain Barrier Disruption and Neurological Deficits in Stroke. Free Radic. Biol. Med. 2013, 65, 1012–1022. [Google Scholar] [CrossRef]
  7. Song, J.; Park, J.; Oh, Y.; Lee, J.E. Glutathione Suppresses Cerebral Infarct Volume and Cell Death after Ischemic Injury: Involvement of FOXO3 Inactivation and Bcl2 Expression. Oxid. Med. Cell. Longev. 2015, 2015, 426069. [Google Scholar] [CrossRef]
  8. Dinkova-Kostova, A.T.; Abramov, A.Y. The Emerging Role of Nrf2 in Mitochondrial Function. Free Radic. Biol. Med. 2015, 88, 179–188. [Google Scholar] [CrossRef]
  9. Liebmann, M.; Korn, L.; Janoschka, C.; Albrecht, S.; Lauks, S.; Herrmann, A.M.; Schulte-Mecklenbeck, A.; Schwab, N.; Schneider-Hohendorf, T.; Eveslage, M.; et al. Dimethyl Fumarate Treatment Restrains the Antioxidative Capacity of T Cells to Control Autoimmunity. Brain 2021, 144, 3126–3141. [Google Scholar] [CrossRef] [PubMed]
  10. Mirshafiey, A.; Mohsenzadegan, M. Antioxidant Therapy in Multiple Sclerosis. Immunopharmacol. Immunotoxicol. 2009, 31, 13–29. [Google Scholar] [CrossRef]
  11. Dohare, P.; Hyzinski-García, M.C.; Vipani, A.; Bowens, N.H.; Nalwalk, J.W.; Feustel, P.J.; Keller, R.W.; Jourd’heuil, D.; Mongin, A.A. The Neuroprotective Properties of the Superoxide Dismutase Mimetic Tempol Correlate with Its Ability to Reduce Pathological Glutamate Release in a Rodent Model of Stroke. Free Radic. Biol. Med. 2014, 77, 168–182. [Google Scholar] [CrossRef]
  12. Huang, H.; Guo, F.; Cao, Y.; Shi, W.; Xia, Q. Neuroprotection by Manganese Superoxide Dismutase (MnSOD) Mimics: Antioxidant Effect and Oxidative Stress Regulation in Acute Experimental Stroke. CNS Neurosci. Ther. 2012, 18, 811–818. [Google Scholar] [CrossRef]
  13. Owjfard, M.; Bigdeli, M.R.; Safari, A.; Haghani, M.; Namavar, M.R. Effect of Dimethyl Fumarate on the Motor Function and Spatial Arrangement of Primary Motor Cortical Neurons in the Sub-Acute Phase of Stroke in a Rat Model. J. Stroke. Cerebrovasc. Dis. 2021, 30, 105630. [Google Scholar] [CrossRef]
  14. Hou, X.; Xu, H.; Chen, W.; Zhang, N.; Zhao, Z.; Fang, X.; Zhang, X.; Chen, H.; Xu, Y. Neuroprotective Effect of Dimethyl Fumarate on Cognitive Impairment Induced by Ischemic Stroke. Ann. Transl. Med. 2020, 8, 375. [Google Scholar] [CrossRef]
  15. Bo, C.; Li, J.; Wang, J.; Zhang, Y.; Wu, T.; Wang, M.; Hou, S.; Liang, Y.; Zhang, X.; Zhao, S.; et al. Dimethyl Fumarate Modulates the Immune Environment and Improves Prognosis in the Acute Phase after Ischemic Stroke. Neuroimmunomodulation 2024, 31, 126–141. [Google Scholar] [CrossRef]
  16. Krämer, T.; Grob, T.; Menzel, L.; Hirnet, T.; Griemert, E.; Radyushkin, K.; Thal, S.C.; Methner, A.; Schaefer, M.K.E. Dimethyl Fumarate Treatment after Traumatic Brain Injury Prevents Depletion of Antioxidative Brain Glutathione and Confers Neuroprotection. J. Neurochem. 2017, 143, 523–533. [Google Scholar] [CrossRef] [PubMed]
  17. Casili, G.; Campolo, M.; Paterniti, I.; Lanza, M.; Filippone, A.; Cuzzocrea, S.; Esposito, E. Dimethyl Fumarate Attenuates Neuroinflammation and Neurobehavioral Deficits Induced by Experimental Traumatic Brain Injury. J. Neurotrauma. 2018, 35, 1437–1451. [Google Scholar] [CrossRef]
  18. Powell, K.; Wadolowski, S.; Tambo, W.; Chang, E.H.; Kim, D.; Jacob, A.; Sciubba, D.; AlAbed, Y.; Wang, P.; Li, C. Endogenous CGRP Activates NRF2 Signaling via Non-Electrophilic Mechanisms. bioRxiv 2025. [Google Scholar] [CrossRef]
  19. Zhang, Q.-Y.; Chu, X.-Y.; Jiang, L.-H.; Liu, M.-Y.; Mei, Z.-L.; Zhang, H.-Y. Identification of Non-Electrophilic Nrf2 Activators from Approved Drugs. Molecules 2017, 22, 883. [Google Scholar] [CrossRef] [PubMed]
  20. Chan, J.Y.; Kwong, M. Impaired Expression of Glutathione Synthetic Enzyme Genes in Mice with Targeted Deletion of the Nrf2 Basic-Leucine Zipper Protein. Biochim. Biophys. Acta (BBA)—Gene Struct. Expr. 2000, 1517, 19–26. [Google Scholar] [CrossRef]
  21. Ishii, T.; Itoh, K.; Takahashi, S.; Sato, H.; Yanagawa, T.; Katoh, Y.; Bannai, S.; Yamamoto, M. Transcription Factor Nrf2 Coordinately Regulates a Group of Oxidative Stress-Inducible Genes in Macrophages. J. Biol. Chem. 2000, 275, 16023–16029. [Google Scholar] [CrossRef] [PubMed]
  22. Kwon, J.H.; Lee, J.; Kim, J.; Kirchner, V.A.; Jo, Y.H.; Miura, T.; Kim, N.; Song, G.-W.; Hwang, S.; Lee, S.-G.; et al. Upregulation of Carbonyl Reductase 1 by Nrf2 as a Potential Therapeutic Intervention for Ischemia/Reperfusion Injury during Liver Transplantation. Mol. Cells 2019, 42, 672–685. [Google Scholar] [CrossRef]
  23. Zhang, C.; Zhao, M.; Wang, B.; Su, Z.; Guo, B.; Qin, L.; Zhang, W.; Zheng, R. The Nrf2-NLRP3-Caspase-1 Axis Mediates the Neuroprotective Effects of Celastrol in Parkinson’s Disease. Redox Biol. 2021, 47, 102134. [Google Scholar] [CrossRef] [PubMed]
  24. Yang, T.; Li, Q.; Fadoul, G.; Alraqmany, N.; Ikonomovic, M.; Zhang, F. Aldo-Keto Reductase 1C15 Characterization and Protection in Ischemic Brain Injury. Antioxidants 2023, 12, 909. [Google Scholar] [CrossRef]
  25. Lee, K.H.; Cha, M.; Lee, B.H. Neuroprotective Effect of Antioxidants in the Brain. Int. J. Mol. Sci. 2020, 21, 7152. [Google Scholar] [CrossRef] [PubMed]
  26. Li, Y.; Chu, L.; Liu, C.; Zha, Z.; Shu, Y. Protective Effect of GSK-3β/Nrf2 Mediated by Dimethyl Fumarate in Middle Cerebral Artery Embolization Reperfusion Rat Model. Curr. Neurovasc. Res. 2021, 18, 456–464. [Google Scholar] [CrossRef]
  27. Mao, L.; Yang, T.; Li, X.; Lei, X.; Sun, Y.; Zhao, Y.; Zhang, W.; Gao, Y.; Sun, B.; Zhang, F. Protective Effects of Sulforaphane in Experimental Vascular Cognitive Impairment: Contribution of the Nrf2 Pathway. J. Cereb. Blood Flow Metab. 2019, 39, 352–366. [Google Scholar] [CrossRef]
  28. Chen, Y.; Zhang, X.; Yang, Y.; Zhang, L.; Cui, L.; Zhang, C.; Chen, R.; Xie, Y.; He, J.; He, W. Tert-Butylhydroquinone Enhanced Angiogenesis and Astrocyte Activation by Activating Nuclear Factor-E2-Related Factor 2/Heme Oxygenase-1 after Focal Cerebral Ischemia in Mice. Microvasc. Res. 2019, 126, 103891. [Google Scholar] [CrossRef]
  29. Dordoe, C.; Wang, X.; Lin, P.; Wang, Z.; Hu, J.; Wang, D.; Fang, Y.; Liang, F.; Ye, S.; Chen, J.; et al. Non-Mitogenic Fibroblast Growth Factor 1 Protects against Ischemic Stroke by Regulating Microglia/Macrophage Polarization through Nrf2 and NF-ΚB Pathways. Neuropharmacology 2022, 212, 109064. [Google Scholar] [CrossRef]
  30. Wu, H.; Che, X.; Zheng, Q.; Wu, A.; Pan, K.; Shao, A.; Wu, Q.; Zhang, J.; Hong, Y. Caspases: A Molecular Switch Node in the Crosstalk between Autophagy and Apoptosis. Int. J. Biol. Sci. 2014, 10, 1072–1083. [Google Scholar] [CrossRef]
  31. Aoyama, K. Glutathione in the Brain. Int. J. Mol. Sci. 2021, 22, 5010. [Google Scholar] [CrossRef]
  32. Dringen, R.; Hirrlinger, J. Glutathione Pathways in the Brain. Biol. Chem. 2003, 384, 505–516. [Google Scholar] [CrossRef]
  33. Baba, M.Z.; Gomathy, S.; Wahedi, U. Role of Nrf2 Pathway Activation in Neurological Disorder: A Brief Review. J. Pharmacol. Pharmacother. 2022, 13, 229–238. [Google Scholar] [CrossRef]
  34. Morris, G.; Walker, A.J.; Walder, K.; Berk, M.; Marx, W.; Carvalho, A.F.; Maes, M.; Puri, B.K. Increasing Nrf2 Activity as a Treatment Approach in Neuropsychiatry. Mol. Neurobiol. 2021, 58, 2158–2182. [Google Scholar] [CrossRef]
  35. Tutakhail, A.; Nazary, Q.A.; Lebsir, D.; Kerdine-Romer, S.; Coudore, F. Induction of Brain Nrf2-HO-1 Pathway and Antinociception after Different Physical Training Paradigms in Mice. Life Sci. 2018, 209, 149–156. [Google Scholar] [CrossRef] [PubMed]
  36. Shen, K.; Wang, X.; Wang, Y.; Jia, Y.; Zhang, Y.; Wang, K.; Luo, L.; Cai, W.; Li, J.; Li, S.; et al. MiR-125b-5p in Adipose Derived Stem Cells Exosome Alleviates Pulmonary Microvascular Endothelial Cells Ferroptosis via Keap1/Nrf2/GPX4 in Sepsis Lung Injury. Redox Biol. 2023, 62, 102655. [Google Scholar] [CrossRef]
  37. Baird, L.; Yamamoto, M. The Molecular Mechanisms Regulating the KEAP1-NRF2 Pathway. Mol. Cell. Biol. 2020, 40, e00099-20. [Google Scholar] [CrossRef] [PubMed]
  38. Dinkova-Kostova, A.T.; Copple, I.M. Advances and Challenges in Therapeutic Targeting of NRF2. Trends. Pharmacol. Sci. 2023, 44, 137–149. [Google Scholar] [CrossRef]
  39. Barakat, M.; Han, C.; Chen, L.; David, B.P.; Shi, J.; Xu, A.; Skowron, K.J.; Johnson, T.; Woods, R.A.; Ankireddy, A.; et al. Non-Electrophilic NRF2 Activators Promote Wound Healing in Human Keratinocytes and Diabetic Mice and Demonstrate Selective Downstream Gene Targeting. Sci. Rep. 2024, 14, 25258. [Google Scholar] [CrossRef]
  40. Zhou, H.; Wang, Y.; You, Q.; Jiang, Z. Recent Progress in the Development of Small Molecule Nrf2 Activators: A Patent Review (2017–Present). Expert Opin. Ther. Pat. 2020, 30, 209–225. [Google Scholar] [CrossRef]
  41. Davis, C.K.; Arruri, V.; Joshi, P.; Vemuganti, R. Non-Pharmacological Interventions for Traumatic Brain Injury. J. Cereb. Blood Flow Metab. 2024, 44, 641–659. [Google Scholar] [CrossRef]
  42. Ni, C.; Huang, B.; Huang, Y.; Wen, Z.; Luo, S. Keap1-Independent GSK-3β/Nrf2 Signaling Mediates Electroacupuncture Inhibition of Oxidative Stress to Induce Cerebral Ischemia-Reperfusion Tolerance. Brain Res. Bull. 2024, 217, 111071. [Google Scholar] [CrossRef]
  43. Zou, Y.; Chen, Z.; Sun, C.; Yang, D.; Zhou, Z.; Peng, X.; Zheng, L.; Tang, C. Exercise Intervention Mitigates Pathological Liver Changes in NAFLD Zebrafish by Activating SIRT1/AMPK/NRF2 Signaling. Int. J. Med. Stud. 2021, 22, 10940. [Google Scholar] [CrossRef] [PubMed]
  44. Deramaudt, T.B.; Chehaitly, A.; Charrière, T.; Arnaud, J.; Bonay, M. High-Frequency Repetitive Magnetic Stimulation Activates Bactericidal Activity of Macrophages via Modulation of P62/Keap1/Nrf2 and P38 MAPK Pathways. Antioxidants 2023, 12, 1695. [Google Scholar] [CrossRef] [PubMed]
  45. Gómez-García, E.F.; Del Campo, F.M.; Cortés-Sanabria, L.; Mendoza-Carrera, F.; Avesani, C.M.; Stenvinkel, P.; Lindholm, B.; Cueto-Manzano, A.M. Transcription Factor NRF2 as Potential Therapeutic Target for Preventing Muscle Wasting in Aging Chronic Kidney Disease Patients. J. Nephrol. 2022, 35, 2215–2225. [Google Scholar] [CrossRef] [PubMed]
  46. Xu, Y.; Xu, M.; Zhou, C.; Sun, L.; Cai, W.; Li, X. Ferroptosis and Its Implications in Treating Cognitive Impairment Caused by Aging: A Study on the Mechanism of Repetitive Transcranial Magnetic Stimulation. Exp. Gerontol. 2024, 192, 112443. [Google Scholar] [CrossRef]
  47. Jin, W.-J.; Zhu, X.-X.; Luo, K.-T.; Wang, S.; Li, J.-A.; Qian, L.-F.; Xu, G.-X. Enhancement of Cognitive Function in Rats with Vascular Dementia Through Modulation of the Nrf2/GPx4 Signaling Pathway by High-Frequency Repetitive Transcranial Magnetic Stimulation. Physiol. Res. 2024, 73, 857–868. [Google Scholar] [CrossRef]
  48. Campione, E.; Mazzilli, S.; Di Prete, M.; Dattola, A.; Cosio, T.; Lettieri Barbato, D.; Costanza, G.; Lanna, C.; Manfreda, V.; Gaeta Schumak, R.; et al. The Role of Glutathione-S Transferase in Psoriasis and Associated Comorbidities and the Effect of Dimethyl Fumarate in This Pathway. Front. Med. 2022, 9, 760852. [Google Scholar] [CrossRef]
  49. Glutathione-S-transferase T1 Genotyping and Phenotyping in Psoriasis Patients Receiving Treatment with Oral Fumaric Acid Esters—Gambichler—2014—Journal of the European Academy of Dermatology and Venereology—Wiley Online Library. Available online: https://onlinelibrary.wiley.com/doi/10.1111/jdv.12137 (accessed on 2 May 2025).
  50. Fadoul, G.; Ikonomovic, M.; Zhang, F.; Yang, T. The Cell-specific Roles of Nrf2 in Acute and Chronic Phases of Ischemic Stroke. CNS Neurosci. Ther. 2023, 30, e14462. [Google Scholar] [CrossRef]
  51. Mathis, B.J.; Kato, H.; Hiramatsu, Y. Induction of Cardiac Pathology: Endogenous versus Exogenous Nrf2 Upregulation. Cells 2022, 11, 3855. [Google Scholar] [CrossRef]
  52. Lynch, D.R.; Chin, M.P.; Delatycki, M.B.; Subramony, S.H.; Corti, M.; Hoyle, J.C.; Boesch, S.; Nachbauer, W.; Mariotti, C.; Mathews, K.D.; et al. Safety and Efficacy of Omaveloxolone in Friedreich Ataxia (MOXIe Study). Ann. Neurol. 2021, 89, 212–225. [Google Scholar] [CrossRef] [PubMed]
  53. Lewis, J.H.; Jadoul, M.; Block, G.A.; Chin, M.P.; Ferguson, D.A.; Goldsberry, A.; Meyer, C.J.; O’Grady, M.; Pergola, P.E.; Reisman, S.A.; et al. Effects of Bardoxolone Methyl on Hepatic Enzymes in Patients with Type 2 Diabetes Mellitus and Stage 4 CKD. Clin. Transl. Sci. 2021, 14, 299–309. [Google Scholar] [CrossRef]
  54. Aranda-Rivera, A.K.; Cruz-Gregorio, A.; Amador-Martínez, I.; Hernández-Cruz, E.Y.; Tapia, E.; Pedraza-Chaverri, J. Antioxidants Targeting Mitochondria Function in Kidney Diseases. Mitochondrial Commun. 2024, 2, 21–37. [Google Scholar] [CrossRef]
  55. Satoh, T.; McKercher, S.R.; Lipton, S.A. Nrf2/ARE-Mediated Antioxidant Actions of Pro-Electrophilic Drugs. Free Radic. Biol. Med. 2013, 65, 645–657. [Google Scholar] [CrossRef] [PubMed]
  56. Schmidt, M.M.; Dringen, R. Fumaric Acid Diesters Deprive Cultured Primary Astrocytes Rapidly of Glutathione. Neurochem. Int. 2010, 57, 460–467. [Google Scholar] [CrossRef]
  57. Dibbert, S.; Clement, B.; Skak-Nielsen, T.; Mrowietz, U.; Rostami-Yazdi, M. Detection of Fumarate–Glutathione Adducts in the Portal Vein Blood of Rats: Evidence for Rapid Dimethylfumarate Metabolism. Arch. Dermatol. Res. 2013, 305, 447–451. [Google Scholar] [CrossRef]
  58. Kubo, E.; Chhunchha, B.; Singh, P.; Sasaki, H.; Singh, D.P. Sulforaphane Reactivates Cellular Antioxidant Defense by Inducing Nrf2/ARE/Prdx6 Activity during Aging and Oxidative Stress. Sci. Rep. 2017, 7, 14130. [Google Scholar] [CrossRef]
  59. Ngo, V.; Duennwald, M.L. Nrf2 and Oxidative Stress: A General Overview of Mechanisms and Implications in Human Disease. Antioxidants 2022, 11, 2345. [Google Scholar] [CrossRef]
  60. Brandes, M.S.; Gray, N.E. NRF2 as a Therapeutic Target in Neurodegenerative Diseases. ASN Neuro. 2020, 12, 1759091419899782. [Google Scholar] [CrossRef] [PubMed]
  61. Cuadrado, A.; Rojo, A.I.; Wells, G.; Hayes, J.D.; Cousin, S.P.; Rumsey, W.L.; Attucks, O.C.; Franklin, S.; Levonen, A.-L.; Kensler, T.W.; et al. Therapeutic Targeting of the NRF2 and KEAP1 Partnership in Chronic Diseases. Nat. Rev. Drug Discov. 2019, 18, 295–317. [Google Scholar] [CrossRef]
  62. Ulasov, A.V.; Rosenkranz, A.A.; Georgiev, G.P.; Sobolev, A.S. Nrf2/Keap1/ARE Signaling: Towards Specific Regulation. Life Sci. 2022, 291, 120111. [Google Scholar] [CrossRef] [PubMed]
  63. Muthusamy, V.R.; Kannan, S.; Sadhaasivam, K.; Gounder, S.S.; Davidson, C.J.; Boeheme, C.; Hoidal, J.R.; Wang, L.; Rajasekaran, N.S. Acute Exercise Stress Activates Nrf2/ARE Signaling and Promotes Antioxidant Mechanisms in the Myocardium. Free Radic. Biol. Med. 2012, 52, 366–376. [Google Scholar] [CrossRef]
  64. Li, T.; He, S.; Liu, S.; Kong, Z.; Wang, J.; Zhang, Y. Effects of Different Exercise Durations on Keap1-Nrf2-ARE Pathway Activation in Mouse Skeletal Muscle. Free Radic. Res. 2015, 49, 1269–1274. [Google Scholar] [CrossRef]
  65. Done, A.J.; Newell, M.J.; Traustadóttir, T. Effect of Exercise Intensity on Nrf2 Signalling in Young Men. Free Radic. Res. 2017, 51, 646–655. [Google Scholar] [CrossRef]
  66. Powell, K.; Wadolowski, S.; Tambo, W.; Strohl, J.J.; Kim, D.; Turpin, J.; Al-Abed, Y.; Brines, M.; Huerta, P.T.; Li, C. Intrinsic Diving Reflex Induces Potent Antioxidative Response by Activation of NRF2 Signaling. bioRxiv 2024. [Google Scholar] [CrossRef]
  67. Ebrahimi-Kia, Y.; Noori-Zadeh, A.; Rajaei, F.; Darabi, S.; Darabi, L.; Ghasemi Hamidabadi, H. The Effect of Bisphenol A and Photobiomodulation Therapy on Autophagy-Related Genes Induction in Adipose Tissue-Derived Stem Cells. J. Lasers Med. Sci. 2022, 13, e15. [Google Scholar] [CrossRef]
  68. Mohamed Abdelgawad, L.; Abd El-hamed, M.M.; Sabry, D.; Abdelgwad, M. Efficacy of Photobiomodulation and Metformin on Diabetic Cell Line of Human Periodontal Ligament Stem Cells through Keap1/Nrf2/Ho-1 Pathway. Rep. Biochem. Mol. Biol. 2021, 10, 30–40. [Google Scholar] [CrossRef] [PubMed]
  69. Madkour, M.I.; El-Serafi, A.; Jahrami, H.A.; Sherif, N.M.; Hassan, R.E.; Awadallah, S.; Al-Islam, M.E.; Faris, E. Ramadan Diurnal Intermittent Fasting Modulates SOD2, TFAM, Nrf2, and Sirtuins (SIRT1, SIRT3) Gene Expressions in Subjects with Overweight and Obesity. Diabetes Res. Clin. Pract. 2019, 155, 107801. [Google Scholar] [CrossRef] [PubMed]
  70. Ninfali, C.; Cortés, M.; Martínez-Campanario, M.C.; Domínguez, V.; Han, L.; Tobías, E.; Esteve-Codina, A.; Enrich, C.; Pintado, B.; Garrabou, G.; et al. The Adaptive Antioxidant Response during Fasting-Induced Muscle Atrophy Is Oppositely Regulated by ZEB1 and ZEB2. Proc. Natl. Acad. Sci. USA 2023, 120, e2301120120. [Google Scholar] [CrossRef] [PubMed]
  71. Vasconcelos, A.R.; Da Paixão, A.G.; Kinoshita, P.F.; Orellana, A.M.; Scavone, C.; Kawamoto, E.M. Toll-like Receptor 4 Signaling Is Critical for the Adaptive Cellular Stress Response Effects Induced by Intermittent Fasting in the Mouse Brain. Neuroscience 2021, 465, 142–153. [Google Scholar] [CrossRef]
  72. Wang, Y.-Y.; Yang, Y.-X.; Zhe, H.; He, Z.-X.; Zhou, S.-F. Bardoxolone Methyl (CDDO-Me) as a Therapeutic Agent: An Update on Its Pharmacokinetic and Pharmacodynamic Properties. Drug Des. Devel. Ther. 2014, 8, 2075–2088. [Google Scholar] [CrossRef] [PubMed]
  73. Ng, H.S.; Zhu, F.; Zhao, Y.; Yao, S.; Lu, X.; Ekuma, O.; Evans, C.; Fisk, J.D.; Marrie, R.A.; Tremlett, H. Adverse Events Associated With Disease-Modifying Drugs for Multiple Sclerosis: A Multiregional Population-Based Study. Neurology 2024, 102, e208006. [Google Scholar] [CrossRef]
  74. Muñoz, M.A.; Kulick, C.G.; Kortepeter, C.M.; Levin, R.L.; Avigan, M.I. Liver Injury Associated with Dimethyl Fumarate in Multiple Sclerosis Patients. Mult. Scler. 2017, 23, 1947–1949. [Google Scholar] [CrossRef]
  75. Pasquali, L.; Pecori, C.; Lucchesi, C.; LoGerfo, A.; Iudice, A.; Siciliano, G.; Bonuccelli, U. Plasmatic Oxidative Stress Biomarkers in Multiple Sclerosis: Relation with Clinical and Demographic Characteristics. Clin. Biochem. 2015, 48, 19–23. [Google Scholar] [CrossRef] [PubMed]
  76. Jüngst, C.; Kim, Y.-J.; Lammert, F. Severe Drug-Induced Liver Injury Related to Therapy with Dimethyl Fumarate. Hepatology 2016, 64, 1367–1369. [Google Scholar] [CrossRef]
  77. Omaveloxolone. In LiverTox: Clinical and Research Information on. Drug-Induced Liver Injury; National Institute of Diabetes and Digestive and Kidney Diseases: Bethesda, MD, USA, 2012.
  78. Hoefnagel, J.J.; Thio, H.B.; Willemze, R.; Bouwes Bavinck, J.N. Long-Term Safety Aspects of Systemic Therapy with Fumaric Acid Esters in Severe Psoriasis. Br. J. Dermatol. 2003, 149, 363–369. [Google Scholar] [CrossRef]
  79. Bakić, M.; Klisić, A.; Kocić, G.; Kocić, H.; Karanikolić, V. Oxidative Stress and Metabolic Biomarkers in Patients with Psoriasis. J. Med. Biochem. 2024, 43, 97–105. [Google Scholar] [CrossRef]
  80. Skoie, I.M.; Dalen, I.; Omdal, R.; Jonsson, G. Malondialdehyde and Advanced Oxidation Protein Products Are Not Increased in Psoriasis: A Controlled Study. Arch. Dermatol. Res. 2019, 311, 299–308. [Google Scholar] [CrossRef]
  81. Aksoy, M.; Kirmit, A. Thiol/Disulphide Balance in Patients with Psoriasis. Postep. Dermatol. Alergol. 2020, 37, 52–55. [Google Scholar] [CrossRef] [PubMed]
  82. R Rodríguez, L.; Lapeña, T.; Calap-Quintana, P.; Moltó, M.D.; Gonzalez-Cabo, P.; Navarro Langa, J.A. Antioxidant Therapies and Oxidative Stress in Friedreich’s Ataxia: The Right Path or Just a Diversion? Antioxidants 2020, 9, 664. [Google Scholar] [CrossRef] [PubMed]
  83. Kato, H.; Watanabe, H.; Imafuku, T.; Arimura, N.; Fujita, I.; Noguchi, I.; Tanaka, S.; Nakano, T.; Tokumaru, K.; Enoki, Y.; et al. Advanced Oxidation Protein Products Contribute to Chronic Kidney Disease-Induced Muscle Atrophy by Inducing Oxidative Stress via CD36/NADPH Oxidase Pathway. J. Cachexia Sarcopenia Muscle 2021, 12, 1832–1847. [Google Scholar] [CrossRef] [PubMed]
  84. Wang, W.; Liu, M.; Miao, H.; Gong, X.; Han, F.; Shi, L.; Yan, X.; Xu, Z. Electroacupuncture Improves Learning and Memory Deficits in Diabetic Encephalopathy Rats by Regulating the Nrf2/HO-1 Pathway. Brain Res. 2025, 1847, 149309. [Google Scholar] [CrossRef]
  85. Li, X.; Yin, C.; Hu, Q.; Wang, J.; Nie, H.; Liu, B.; Tai, Y.; Fang, J.; Du, J.; Shao, X.; et al. Nrf2 Activation Mediates Antiallodynic Effect of Electroacupuncture on a Rat Model of Complex Regional Pain Syndrome Type-I through Reducing Local Oxidative Stress and Inflammation. Oxid. Med. Cell. Longev. 2022, 2022, 8035109. [Google Scholar] [CrossRef]
  86. Song, S.; An, J.; Li, Y.; Liu, S. Electroacupuncture at ST-36 Ameliorates DSS-Induced Acute Colitis via Regulating Macrophage Polarization Induced by Suppressing NLRP3/IL-1β and Promoting Nrf2/HO-1. Mol. Immunol. 2019, 106, 143–152. [Google Scholar] [CrossRef]
  87. Zhou, Z.; Yang, Y.; Wei, Y.; Xie, Y. Remimazolam Attenuates LPS-Derived Cognitive Dysfunction via Subdiaphragmatic Vagus Nerve Target A7nAChR-Mediated Nrf2/HO-1 Signal Pathway. Neurochem. Res. 2024, 49, 1306–1321. [Google Scholar] [CrossRef]
  88. Monir, D.M.; Mahmoud, M.E.; Ahmed, O.G.; Rehan, I.F.; Abdelrahman, A. Forced Exercise Activates the NrF2 Pathway in the Striatum and Ameliorates Motor and Behavioral Manifestations of Parkinson’s Disease in Rotenone-Treated Rats. Behav. Brain Funct. 2020, 16, 9. [Google Scholar] [CrossRef] [PubMed]
  89. Asghar, M.; George, L.; Lokhandwala, M.F. Exercise Decreases Oxidative Stress and Inflammation and Restores Renal Dopamine D1 Receptor Function in Old Rats. Am. J. Physiol.-Ren. Physiol. 2007, 293, F914–F919. [Google Scholar] [CrossRef]
  90. Tsou, Y.-H.; Shih, C.-T.; Ching, C.-H.; Huang, J.-Y.; Jen, C.J.; Yu, L.; Kuo, Y.-M.; Wu, F.-S.; Chuang, J.-I. Treadmill Exercise Activates Nrf2 Antioxidant System to Protect the Nigrostriatal Dopaminergic Neurons from MPP+ Toxicity. Exp. Neurol. 2015, 263, 50–62. [Google Scholar] [CrossRef]
  91. Deng, J.; Lv, E.; Yang, J.; Gong, X.; Zhang, W.; Liang, X.; Wang, J.; Jia, J.; Wang, X. Electroacupuncture Remediates Glial Dysfunction and Ameliorates Neurodegeneration in the Astrocytic α-Synuclein Mutant Mouse Model. J. Neuroinflamm. 2015, 12, 103. [Google Scholar] [CrossRef] [PubMed]
  92. Aguiar, A.S.; Duzzioni, M.; Remor, A.P.; Tristão, F.S.M.; Matheus, F.C.; Raisman-Vozari, R.; Latini, A.; Prediger, R.D. Moderate-Intensity Physical Exercise Protects Against Experimental 6-Hydroxydopamine-Induced Hemiparkinsonism Through Nrf2-Antioxidant Response Element Pathway. Neurochem. Res. 2016, 41, 64–72. [Google Scholar] [CrossRef]
  93. He, J.-T.; Li, H.; Yang, L.; Cheng, K.-L. Involvement of Endothelin-1, H2S and Nrf2 in Beneficial Effects of Remote Ischemic Preconditioning in Global Cerebral Ischemia-Induced Vascular Dementia in Mice. Cell. Mol. Neurobiol. 2019, 39, 671–686. [Google Scholar] [CrossRef] [PubMed]
  94. Wang, X.-R.; Shi, G.-X.; Yang, J.-W.; Yan, C.-Q.; Lin, L.-T.; Du, S.-Q.; Zhu, W.; He, T.; Zeng, X.-H.; Xu, Q.; et al. Acupuncture Ameliorates Cognitive Impairment and Hippocampus Neuronal Loss in Experimental Vascular Dementia through Nrf2-Mediated Antioxidant Response. Free Radic. Biol. Med. 2015, 89, 1077–1084. [Google Scholar] [CrossRef]
  95. Tasset, I.; Pérez-Herrera, A.; Medina, F.J.; Arias-Carrión, Ó.; Drucker-Colín, R.; Túnez, I. Extremely Low-Frequency Electromagnetic Fields Activate the Antioxidant Pathway Nrf2 in a Huntington’s Disease-like Rat Model. Brain Stimul. 2013, 6, 84–86. [Google Scholar] [CrossRef]
  96. Tian, L.; Sun, S.-S.; Cui, L.-B.; Wang, S.-Q.; Peng, Z.-W.; Tan, Q.-R.; Hou, W.-G.; Cai, M. Repetitive Transcranial Magnetic Stimulation Elicits Antidepressant- and Anxiolytic-like Effect via Nuclear Factor-E2-Related Factor 2-Mediated Anti-Inflammation Mechanism in Rats. Neuroscience 2020, 429, 119–133. [Google Scholar] [CrossRef] [PubMed]
  97. Cheng, W.-J.; Li, P.; Huang, W.-Y.; Huang, Y.; Chen, W.-J.; Chen, Y.-P.; Shen, J.-L.; Chen, J.-K.; Long, N.-S.; Meng, X.-J. Acupuncture Relieves Stress-Induced Depressive Behavior by Reducing Oxidative Stress and Neuroapoptosis in Rats. Front. Behav. Neurosci. 2022, 15. [Google Scholar] [CrossRef]
  98. Li, X.; Hu, J.; Zang, X.; Xing, J.; Mo, X.; Hei, Z.; Gong, C.; Chen, C.; Zhou, S. Etomidate Improves the Antidepressant Effect of Electroconvulsive Therapy by Suppressing Hippocampal Neuronal Ferroptosis via Upregulating BDNF/Nrf2. Mol. Neurobiol. 2023, 60, 6584–6597. [Google Scholar] [CrossRef]
  99. Zhou, C.; Xue, F.; Xue, S.; Sang, H.; Liu, L.; Wang, Y.; Cai, M.; Zhang, Z.-J.; Tan, Q.; Wang, H.; et al. Electroacupuncture Pretreatment Ameliorates PTSD-Like Behaviors in Rats by Enhancing Hippocampal Neurogenesis via the Keap1/Nrf2 Antioxidant Signaling Pathway. Front. Cell. Neurosci. 2019, 13, 275. [Google Scholar] [CrossRef]
  100. Morrison, C.D.; Pistell, P.J.; Ingram, D.K.; Johnson, W.D.; Liu, Y.; Fernandez-Kim, S.O.; White, C.L.; Purpera, M.N.; Uranga, R.M.; Bruce-Keller, A.J.; et al. High Fat Diet Increases Hippocampal Oxidative Stress and Cognitive Impairment in Aged Mice: Implications for Decreased Nrf2 Signaling. J. Neurochem. 2010, 114, 1581–1589. [Google Scholar] [CrossRef]
  101. Bayram, B.; Ozcelik, B.; Grimm, S.; Roeder, T.; Schrader, C.; Ernst, I.M.A.; Wagner, A.E.; Grune, T.; Frank, J.; Rimbach, G. A Diet Rich in Olive Oil Phenolics Reduces Oxidative Stress in the Heart of SAMP8 Mice by Induction of Nrf2-Dependent Gene Expression. Rejuvenation Res. 2012, 15, 71–81. [Google Scholar] [CrossRef]
  102. Yubero-Serrano, E.M.; Gonzalez-Guardia, L.; Rangel-Zuñiga, O.; Delgado-Casado, N.; Delgado-Lista, J.; Perez-Martinez, P.; Garcia-Rios, A.; Caballero, J.; Marin, C.; Gutierrez-Mariscal, F.M.; et al. Postprandial Antioxidant Gene Expression Is Modified by Mediterranean Diet Supplemented with Coenzyme Q10 in Elderly Men and Women. Age (Dordr.) 2013, 35, 159–170. [Google Scholar] [CrossRef]
  103. Csiszar, A.; Gautam, T.; Sosnowska, D.; Tarantini, S.; Banki, E.; Tucsek, Z.; Toth, P.; Losonczy, G.; Koller, A.; Reglodi, D.; et al. Caloric Restriction Confers Persistent Anti-Oxidative, pro-Angiogenic, and Anti-Inflammatory Effects and Promotes Anti-Aging miRNA Expression Profile in Cerebromicrovascular Endothelial Cells of Aged Rats. Am. J. Physiol.-Heart Circ. Physiol. 2014, 307, H292–H306. [Google Scholar] [CrossRef]
  104. Pearson, K.J.; Lewis, K.N.; Price, N.L.; Chang, J.W.; Perez, E.; Cascajo, M.V.; Tamashiro, K.L.; Poosala, S.; Csiszar, A.; Ungvari, Z.; et al. Nrf2 Mediates Cancer Protection but Not Prolongevity Induced by Caloric Restriction. Proc. Natl. Acad. Sci. USA 2008, 105, 2325–2330. [Google Scholar] [CrossRef]
  105. Ostrom, E.L.; Valencia, A.P.; Marcinek, D.J.; Traustadóttir, T. High Intensity Muscle Stimulation Activates a Systemic Nrf2-Mediated Redox Stress Response. Free Radic. Biol. Med. 2021, 172, 82–89. [Google Scholar] [CrossRef]
  106. Gounder, S.S.; Kannan, S.; Devadoss, D.; Miller, C.J.; Whitehead, K.J.; Odelberg, S.J.; Firpo, M.A.; Paine, R.; Hoidal, J.R.; Abel, E.D.; et al. Impaired Transcriptional Activity of Nrf2 in Age-Related Myocardial Oxidative Stress Is Reversible by Moderate Exercise Training. PLoS ONE 2012, 7, e45697. [Google Scholar] [CrossRef]
  107. Eskla, K.-L.; Porosk, R.; Reimets, R.; Visnapuu, T.; Vasar, E.; Hundahl, C.A.; Luuk, H. Hypothermia Augments Stress Response in Mammalian Cells. Free Radic. Biol. Med. 2018, 121, 157–168. [Google Scholar] [CrossRef]
  108. Orhan, E.; Gündüz, Ö.; Kaya, O.; Öznur, M.; Şahin, E. Transferring the Protective Effect of Remote Ischemic Preconditioning on Skin Flap among Rats by Blood Serum. J. Plast. Surg. Hand Surg. 2019, 53, 198–203. [Google Scholar] [CrossRef]
  109. Patel, A.D.; Rotenberg, S.; Messer, R.L.W.; Wataha, J.C.; Ogbureke, K.U.E.; Mccloud, V.V.; Lockwood, P.; Hsu, S.; Lewis, J.B. Blue Light Activates Phase 2 Response Proteins and Slows Growth of A431 Epidermoid Carcinoma Xenografts. Anticancer Res. 2014, 34, 6305–6313. [Google Scholar] [PubMed]
  110. Li, B.; Wang, X. Photobiomodulation Enhances Facial Nerve Regeneration via Activation of PI3K/Akt Signaling Pathway–Mediated Antioxidant Response. Lasers Med. Sci. 2022, 37, 993–1006. [Google Scholar] [CrossRef] [PubMed]
  111. Lettieri-Barbato, D.; Minopoli, G.; Caggiano, R.; Izzo, R.; Santillo, M.; Aquilano, K.; Faraonio, R. Fasting Drives Nrf2-Related Antioxidant Response in Skeletal Muscle. Int. J. Mol. Sci. 2020, 21, 7780. [Google Scholar] [CrossRef]
  112. Subramanian, M.; Edwards, L.; Melton, A.; Branen, L.; Herron, A.; Sivasubramanian, M.K.; Monteiro, R.; Stansbury, S.; Balasubramanian, P.; Morris, L.; et al. Non-Invasive Vagus Nerve Stimulation Attenuates Proinflammatory Cytokines and Augments Antioxidant Levels in the Brainstem and Forebrain Regions of Dahl Salt Sensitive Rats. Sci. Rep. 2020, 10, 17576. [Google Scholar] [CrossRef] [PubMed]
  113. Zhang, Y.; Zhang, C.; Dai, Q.; Ma, R. Continuous Theta Burst Stimulation Inhibits Oxidative Stress-Induced Inflammation and Autophagy in Hippocampal Neurons by Activating Glutathione Synthesis Pathway, Improving Cognitive Impairment in Sleep-Deprived Mice. Neuromol. Med. 2024, 26, 40. [Google Scholar] [CrossRef]
  114. Xu, J.; Tao, L.; Jiang, L.; Lai, J.; Hu, J.; Tang, Z. Moderate Hypothermia Alleviates Sepsis-Associated Acute Lung Injury by Suppressing Ferroptosis Induced by Excessive Inflammation and Oxidative Stress via the Keap1/GSK3β/Nrf2/GPX4 Signaling Pathway. J. Inflamm. Res. 2024, 17, 7687–7704. [Google Scholar] [CrossRef]
  115. Luo, J.; Yan, R.; Ding, L.; Ning, J.; Chen, M.; Guo, Y.; Liu, J.; Chen, Z.; Zhou, R. Electroacupuncture Attenuates Ventilator-Induced Lung Injury by Modulating the Nrf2/HO-1 Pathway. J. Surg. Res. 2024, 295, 811–819. [Google Scholar] [CrossRef] [PubMed]
  116. Perez-Herrera, A.; Rangel-Zuñiga, O.A.; Delgado-Lista, J.; Marin, C.; Perez-Martinez, P.; Tasset, I.; Tunez, I.; Quintana-Navarro, G.M.; Lopez-Segura, F.; Luque De Castro, M.D.; et al. The Antioxidants in Oils Heated at Frying Temperature, Whether Natural or Added, Could Protect against Postprandial Oxidative Stress in Obese People. Food Chem. 2013, 138, 2250–2259. [Google Scholar] [CrossRef] [PubMed]
  117. Madkour, M.I.; Hassan, R.E.; Sherif, N.M.; Awadallah, S.; Farahat, N.M.; Abdelrahim, D.N.; AlHasan, F.A.; Taneera, J.; Faris, M.E. Changes in Haptoglobin Genotype-Based Gene Expressions upon the Observance of Dawn-to-Dusk Intermittent Fasting: A Prospective Cohort Study on Overweight and Obese Individuals. Front. Nutr. 2024, 11. [Google Scholar] [CrossRef] [PubMed]
  118. Elsaid, F.H.; Hussein, A.M.; Eid, E.A.; Ammar, O.A.; Khalil, A.A. Effect of Intermittent Fasting on Adriamycin-Induced Nephropathy: Possible Underlying Mechanisms. Tissue Cell 2024, 88, 102360. [Google Scholar] [CrossRef]
  119. Sun, Y.-Y.; Zhu, H.-J.; Zhao, R.-Y.; Zhou, S.-Y.; Wang, M.-Q.; Yang, Y.; Guo, Z.-N. Remote Ischemic Conditioning Attenuates Oxidative Stress and Inflammation via the Nrf2/HO-1 Pathway in MCAO Mice. Redox Biol. 2023, 66, 102852. [Google Scholar] [CrossRef]
  120. Li, P.; Su, L.; Li, X.; Di, W.; Zhang, X.; Zhang, C.; He, T.; Zhu, X.; Zhang, Y.; Li, Y. Remote Limb Ischemic Postconditioning Protects Mouse Brain against Cerebral Ischemia/Reperfusion Injury via Upregulating Expression of Nrf2, HO-1 and NQO-1 in Mice. Int. J. Neurosci. 2015, 1–8. [Google Scholar] [CrossRef]
  121. Gao, S.; Zhan, L.; Yang, Z.; Shi, R.; Li, H.; Xia, Z.; Yuan, S.; Wu, Q.; Wang, T.; Yao, S. Remote Limb Ischaemic Postconditioning Protects Against Myocardial Ischaemia/Reperfusion Injury in Mice: Activation of JAK/STAT3-Mediated Nrf2-Antioxidant Signalling. Cell Physiol. Biochem. 2017, 43, 1140–1151. [Google Scholar] [CrossRef]
  122. Gao, S.; Zhu, Y.; Li, H.; Xia, Z.; Wu, Q.; Yao, S.; Wang, T.; Yuan, S. Remote Ischemic Postconditioning Protects against Renal Ischemia/Reperfusion Injury by Activation of T-LAK-Cell-Originated Protein Kinase (TOPK)/PTEN/Akt Signaling Pathway Mediated Anti-Oxidation and Anti-Inflammation. Int. Immunopharmacol. 2016, 38, 395–401. [Google Scholar] [CrossRef]
  123. Zhang, X.; Jizhang, Y.; Xu, X.; Kwiecien, T.D.; Li, N.; Zhang, Y.; Ji, X.; Ren, C.; Ding, Y. Protective Effects of Remote Ischemic Conditioning against Ischemia/Reperfusion-Induced Retinal Injury in Rats. Vis. Neurosci. 2014, 31, 245–252. [Google Scholar] [CrossRef]
  124. Wang, H.; Shi, X.; Cheng, L.; Han, J.; Mu, J. Hydrogen Sulfide Restores Cardioprotective Effects of Remote Ischemic Preconditioning in Aged Rats via HIF-1α/Nrf2 Signaling Pathway. Korean J. Physiol. Pharmacol. 2021, 25, 239–249. [Google Scholar] [CrossRef]
  125. He, X.; Yang, X.; Li, G.; Zhao, Y.; Luo, J.; Xu, J.; Zheng, H.; Zhang, L.; Hu, X. Physical Exercise Improves the Neuronal Function in Ischemic Stroke Via Microglial CB2R/P2Y12 Signaling. Mol. Neurobiol. 2025, 62, 2039–2057. [Google Scholar] [CrossRef] [PubMed]
  126. Dai, M.; Chen, B.; Wang, X.; Gao, C.; Yu, H. Icariin Enhance Mild Hypothermia-Induced Neuroprotection via Inhibiting the Activation of NF-ΚB in Experimental Ischemic Stroke. Metab. Brain Dis. 2021, 36, 1779–1790. [Google Scholar] [CrossRef] [PubMed]
  127. Gong, L.; Kan, Y.; Lian, Y.; Dong, S.; Zhao, D.; Shi, J.; Yu, J. Electroacupuncture Attenuates Limb Ischemia-Reperfusion-Induced Lung Injury Via P38 Mitogen-Activated Protein Kinase-Nuclear Factor Erythroid-2-Related Factor-2/Heme Oxygenase Pathway. J. Surg. Res. 2020, 246, 170–181. [Google Scholar] [CrossRef] [PubMed]
  128. Zhu, W.; Dong, J.; Han, Y. Electroacupuncture Downregulating Neuronal Ferroptosis in MCAO/R Rats by Activating Nrf2/SLC7A11/GPX4 Axis. Neurochem. Res. 2024, 49, 2105–2119. [Google Scholar] [CrossRef] [PubMed]
  129. Schanuel, F.S.; Saguie, B.O.; Monte-Alto-Costa, A. Olive Oil Promotes Wound Healing of Mice Pressure Injuries through NOS-2 and Nrf2. Appl. Physiol. Nutr. Metab. 2019, 44, 1199–1208. [Google Scholar] [CrossRef]
  130. Zhang, Q.; Lai, Y.; Deng, J.; Wang, M.; Wang, Z.; Wang, M.; Zhang, Y.; Yang, X.; Zhou, X.; Jiang, H. Vagus Nerve Stimulation Attenuates Hepatic Ischemia/Reperfusion Injury via the Nrf2/HO-1 Pathway. Oxid. Med. Cell. Longev. 2019, 2019, 9549506. [Google Scholar] [CrossRef]
  131. Deng, J.; Jiang, Y.; Wang, M.; Shao, L.; Deng, C. Activation of Vagovagal Reflex Prevents Hepatic Ischaemia–Reperfusion-Induced Lung Injury via Anti-Inflammatory and Antioxidant Effects. Exp. Physiol. 2021, 106, 2210–2222. [Google Scholar] [CrossRef]
  132. Dhar, R.; Zhang, L.; Li, Y.; Rana, M.N.; Hu, Z.; Li, Z.; Cui, H.; Tang, H. Electroacupuncture Ameliorates Cardiopulmonary Bypass Induced Apoptosis in Lung via ROS/Nrf2/NLRP3 Inflammasome Pathway. Life Sci. 2019, 238, 116962. [Google Scholar] [CrossRef]
  133. Arvola, O.; Haapanen, H.; Herajärvi, J.; Anttila, T.; Puistola, U.; Karihtala, P.; Tuominen, H.; Anttila, V.; Juvonen, T. Remote Ischemic Preconditioning Reduces Cerebral Oxidative Stress Following Hypothermic Circulatory Arrest in a Porcine Model. Semin. Thorac. Cardiovasc. Surg. 2016, 28, 92–102. [Google Scholar] [CrossRef] [PubMed]
  134. Bahr, A.C.; Naasani, L.I.S.; De Gregório, E.; Wink, M.R.; Da Rosa Araujo, A.S.; Turck, P.; Dal Lago, P. Photobiomodulation Improves Cell Survival and Death Parameters in Cardiomyocytes Exposed to Hypoxia/Reoxygenation. J. Photochem. B Biol. 2024, 258, 112991. [Google Scholar] [CrossRef] [PubMed]
  135. Diao, M.-Y.; Zheng, J.; Shan, Y.; Xi, S.; Zhu, Y.; Hu, W.; Lin, Z. Hypothermia Prevents Hippocampal Oxidative Stress and Apoptosis via the GSK-3β/Nrf2/HO-1 Signaling Pathway in a Rat Model of Cardiac Arrest-Induced Brain Damage. Neurol. Res. 2020, 42, 773–782. [Google Scholar] [CrossRef] [PubMed]
  136. Gong, P.; Li, C.-S.; Hua, R.; Zhao, H.; Tang, Z.-R.; Mei, X.; Zhang, M.-Y.; Cui, J. Mild Hypothermia Attenuates Mitochondrial Oxidative Stress by Protecting Respiratory Enzymes and Upregulating MnSOD in a Pig Model of Cardiac Arrest. PLoS ONE 2012, 7, e35313. [Google Scholar] [CrossRef]
  137. Ahn, J.H.; Lee, T.-K.; Kim, D.W.; Shin, M.C.; Cho, J.H.; Lee, J.-C.; Tae, H.-J.; Park, J.H.; Hong, S.; Lee, C.-H.; et al. Therapeutic Hypothermia after Cardiac Arrest Attenuates Hindlimb Paralysis and Damage of Spinal Motor Neurons and Astrocytes through Modulating Nrf2/HO-1 Signaling Pathway in Rats. Cells 2023, 12, 414. [Google Scholar] [CrossRef]
  138. Jawad, A.; Yoo, Y.-J.; Cho, J.-H.; Yoon, J.C.; Tian, W.; Islam, M.S.; Lee, E.-Y.; Shin, H.-Y.; Kim, S.E.; Kim, K.; et al. Therapeutic Hypothermia Effect on Asphyxial Cardiac Arrest-Induced Renal Ischemia/Reperfusion Injury via Change of Nrf2/HO-1 Levels. Exp. Ther. Med. 2021, 22, 1031. [Google Scholar] [CrossRef]
  139. Guan, R.; Wen, X.; Leung, C.H.; Ciano-Oliveira, C.D.; Lam, S.; Dai, S.Y.; Karbassi, F.; Mauro, A.; Wang, Y.; Rotstein, O. Plasma Obtained Following Murine Hindlimb Ischemic Conditioning Protects against Oxidative Stress in Zebrafish Models through Activation of Nrf2a and Downregulation of Duox. PLoS ONE 2021, 16, e0260442. [Google Scholar] [CrossRef]
  140. Bigagli, E.; Cinci, L.; Paccosi, S.; Parenti, A.; D’Ambrosio, M.; Luceri, C. Nutritionally Relevant Concentrations of Resveratrol and Hydroxytyrosol Mitigate Oxidative Burst of Human Granulocytes and Monocytes and the Production of Pro-Inflammatory Mediators in LPS-Stimulated RAW 264.7 Macrophages. Int. Immunopharmacol. 2017, 43, 147–155. [Google Scholar] [CrossRef]
  141. Salman, S.; Guermonprez, C.; Peno-Mazzarino, L.; Lati, E.; Rousseaud, A.; Declercq, L.; Kerdine-Römer, S. Photobiomodulation Controls Keratinocytes Inflammatory Response through Nrf2 and Reduces Langerhans Cells Activation. Antioxidants 2023, 12, 766. [Google Scholar] [CrossRef]
  142. Zhong, Y.; Liu, L.; Zhao, Y.; Feng, Z.; Liu, Y. Elucidating the Molecular Mechanisms behind the Therapeutic Impact of Median Nerve Stimulation on Cognitive Dysfunction Post-Traumatic Brain Injury. Exp. Gerontol. 2024, 194, 112500. [Google Scholar] [CrossRef]
  143. Yan, C.; Mao, J.; Yao, C.; Liu, Y.; Yan, H.; Jin, W. Neuroprotective Effects of Mild Hypothermia against Traumatic Brain Injury by the Involvement of the Nrf2/ARE Pathway. Brain Behav. 2022, 12, e2686. [Google Scholar] [CrossRef]
  144. Dai, S.; Wei, J.; Zhang, H.; Luo, P.; Yang, Y.; Jiang, X.; Fei, Z.; Liang, W.; Jiang, J.; Li, X. Intermittent Fasting Reduces Neuroinflammation in Intracerebral Hemorrhage through the Sirt3/Nrf2/HO-1 Pathway. J. Neuroinflamm. 2022, 19, 122. [Google Scholar] [CrossRef]
  145. Leung, C.H.; Caldarone, C.A.; Guan, R.; Wen, X.-Y.; Ailenberg, M.; Kapus, A.; Szaszi, K.; Rotstein, O.D. Nuclear Factor (Erythroid-Derived 2)-Like 2 Regulates the Hepatoprotective Effects of Remote Ischemic Conditioning in Hemorrhagic Shock. Antioxid. Redox Signal. 2019, 30, 1760–1773. [Google Scholar] [CrossRef] [PubMed]
  146. Yu, J.; Shi, J.; Gong, L.; Dong, S.; Xu, Y.; Zhang, Y.; Cao, X.; Wu, L. Role of Nrf2/ARE Pathway in Protective Effect of Electroacupuncture against Endotoxic Shock-Induced Acute Lung Injury in Rabbits. PLoS ONE 2014, 9, e104924. [Google Scholar] [CrossRef]
  147. Wang, D.; Li, L.; Pan, H.; Huang, L.; Sun, X.; He, C.; Wei, Q. Comparison of the Effects of Constraint-Induced Movement Therapy and Unconstraint Exercise on Oxidative Stress and Limb Function—A Study on Human Patients and Rats with Cerebral Infarction. Brain Sci. 2022, 13, 4. [Google Scholar] [CrossRef] [PubMed]
  148. Ren, C.; Li, S.; Liu, K.; Rajah, G.B.; Zhang, A.; Han, R.; Liu, Y.; Huang, Q.; Li, H.; Ding, Y.; et al. Enhanced Oxidative Stress Response and Neuroprotection of Combined Limb Remote Ischemic Conditioning and Atorvastatin after Transient Ischemic Stroke in Rats. Brain Circ. 2017, 3, 204–212. [Google Scholar] [CrossRef] [PubMed]
  149. Otsuka, S.; Setoyama, K.; Takada, S.; Nakanishi, K.; Terashi, T.; Norimatsu, K.; Tani, A.; Sakakima, H.; Maruyama, I.; Tancharoen, S.; et al. Preconditioning Exercise in Rats Attenuates Early Brain Injury Resulting from Subarachnoid Hemorrhage by Reducing Oxidative Stress, Inflammation, and Neuronal Apoptosis. Mol. Neurobiol. 2021, 58, 5602–5617. [Google Scholar] [CrossRef]
  150. Zhou, C.; Li, H.; Yao, Y.; Li, L. Delayed Remote Ischemic Preconditioning Produces an Additive Cardioprotection to Sevoflurane Postconditioning Through an Enhanced Heme Oxygenase 1 Level Partly Via Nuclear Factor Erythroid 2-Related Factor 2 Nuclear Translocation. J. Cardiovasc. Pharmacol. Ther. 2014, 19, 558–566. [Google Scholar] [CrossRef]
  151. Wang, Y.; Wang, S.; Xin, Y.; Zhang, J.; Wang, S.; Yang, Z.; Liu, C. Hydrogen Sulfide Alleviates the Anxiety-like and Depressive-like Behaviors of Type 1 Diabetic Mice via Inhibiting Inflammation and Ferroptosis. Life Sci. 2021, 278, 119551. [Google Scholar] [CrossRef]
  152. Zhang, J.; Du, Y.; Xiong, Z.; Cheng, H.; Du, Y.; Xiong, Y.; Lv, J.; Huang, W.; Qiu, K.; Zhang, S. Bombesin Protects Myocardium against Ischemia/Reperfusion Injury via Activation of the Keap1-Nrf2-HO-1 Signaling Pathway. Peptides 2024, 180, 171279. [Google Scholar] [CrossRef]
  153. Longo, L.; Sinigaglia-Fratta, L.X.; Weber, G.R.; Janz-Moreira, A.; Kretzmann, N.A.; Grezzana-Filho, T.D.J.M.; Possa-Marroni, N.; Corso, C.O.; Schmidt-Cerski, C.T.; Reverbel-da-Silveira, T.; et al. Hypothermia Is Better than Ischemic Preconditioning for Preventing Early Hepatic Ischemia/Reperfusion in Rats. Ann. Hepatol. 2016, 15, 110–120. [Google Scholar] [CrossRef]
  154. Deng, S.; Zhang, Y.; Xin, Y.; Hu, X. Vagus Nerve Stimulation Attenuates Acute Kidney Injury Induced by Hepatic Ischemia/Reperfusion Injury in Rats. Sci. Rep. 2022, 12, 21662. [Google Scholar] [CrossRef] [PubMed]
  155. Chen, M.; Zhang, M.; Zhang, X.; Li, J.; Wang, Y.; Fan, Y.; Shi, R. Limb Ischemic Preconditioning Protects Endothelium from Oxidative Stress by Enhancing Nrf2 Translocation and Upregulating Expression of Antioxidases. PLoS ONE 2015, 10, e0128455. [Google Scholar] [CrossRef]
  156. Yang, H.-L.; Lin, M.-W.; Korivi, M.; Wu, J.-J.; Liao, C.-H.; Chang, C.-T.; Liao, J.-W.; Hseu, Y.-C. Coenzyme Q0 Regulates NFκB/AP-1 Activation and Enhances Nrf2 Stabilization in Attenuation of LPS-Induced Inflammation and Redox Imbalance: Evidence from in Vitro and in Vivo Studies. Biochim. Biophys. Acta (BBA)—Gene Regul. Mech. 2016, 1859, 246–261. [Google Scholar] [CrossRef] [PubMed]
  157. Bradl, M.; Lassmann, H. Oligodendrocytes: Biology and Pathology. Acta Neuropathol. 2010, 119, 37–53. [Google Scholar] [CrossRef]
  158. Kim, J.Y.; Kim, J.-H.; Kim, Y.-D.; Seo, J.H. High Vulnerability of Oligodendrocytes to Oxidative Stress Induced by Ultrafine Urban Particles. Antioxidants 2020, 10, 4. [Google Scholar] [CrossRef]
  159. Moubarak, M.M.; Pagano Zottola, A.C.; Larrieu, C.M.; Cuvellier, S.; Daubon, T.; Martin, O.C.B. Exploring the Multifaceted Role of NRF2 in Brain Physiology and Cancer: A Comprehensive Review. Neuro-Oncol. Adv. 2024, 6, vdad160. [Google Scholar] [CrossRef]
  160. Lim, J.L.; Van der Pol, S.M.A.; Baron, W.; McCord, J.M.; De Vries, H.E.; Van Horssen, J. Protandim Protects Oligodendrocytes against an Oxidative Insult. Antioxidants 2016, 5, 30. [Google Scholar] [CrossRef]
  161. Teske, N.; Liessem, A.; Fischbach, F.; Clarner, T.; Beyer, C.; Wruck, C.; Fragoulis, A.; Tauber, S.C.; Victor, M.; Kipp, M. Chemical Hypoxia-Induced Integrated Stress Response Activation in Oligodendrocytes Is Mediated by the Transcription Factor Nuclear Factor (Erythroid-Derived 2)-like 2 (NRF2). J. Neurochem. 2018, 144, 285–301. [Google Scholar] [CrossRef]
  162. De Nuccio, C.; Bernardo, A.; Troiano, C.; Brignone, M.S.; Falchi, M.; Greco, A.; Rosini, M.; Basagni, F.; Lanni, C.; Serafini, M.M.; et al. NRF2 and PPAR-γ Pathways in Oligodendrocyte Progenitors: Focus on ROS Protection, Mitochondrial Biogenesis and Promotion of Cell Differentiation. Int. J. Mol. Sci. 2020, 21, 7216. [Google Scholar] [CrossRef] [PubMed]
  163. Abu Shelbayeh, O.; Arroum, T.; Morris, S.; Busch, K.B. PGC-1α Is a Master Regulator of Mitochondrial Lifecycle and ROS Stress Response. Antioxidants 2023, 12, 1075. [Google Scholar] [CrossRef]
  164. Spaas, J.; van Veggel, L.; Schepers, M.; Tiane, A.; van Horssen, J.; Wilson, D.M.; Moya, P.R.; Piccart, E.; Hellings, N.; Eijnde, B.O.; et al. Oxidative Stress and Impaired Oligodendrocyte Precursor Cell Differentiation in Neurological Disorders. Cell. Mol. Life Sci. 2021, 78, 4615–4637. [Google Scholar] [CrossRef]
  165. Sharma, R.; Sharma, A.; Chaudhary, P.; Sahu, M.; Jaiswal, S.; Awasthi, S.; Awasthi, Y.C. Role of 4-Hydroxynonenal in Chemopreventive Activities of Sulforaphane. Free Radic. Biol. Med. 2012, 52, 2177–2185. [Google Scholar] [CrossRef]
  166. Xie, S.; Zou, W.; Liu, S.; Yang, Q.; Hu, T.; Zhu, W.; Tang, H.; Wang, C. Site 1 Protease Aggravates Acute Kidney Injury by Promoting Tubular Epithelial Cell Ferroptosis through SIRT3-SOD2-mtROS Signaling. FEBS J. 2024, 291, 1575–1592. [Google Scholar] [CrossRef] [PubMed]
  167. Amoroso, R.; Maccallini, C.; Bellezza, I. Activators of Nrf2 to Counteract Neurodegenerative Diseases. Antioxidants 2023, 12, 778. [Google Scholar] [CrossRef]
  168. Chen, W.-T.; Dodson, M. The Untapped Potential of Targeting NRF2 in Neurodegenerative Disease. Front. Aging 2023, 4, 1270838. [Google Scholar] [CrossRef] [PubMed]
  169. Pisani, F.; Castagnola, V.; Simone, L.; Loiacono, F.; Svelto, M.; Benfenati, F. Role of Pericytes in Blood–Brain Barrier Preservation during Ischemia through Tunneling Nanotubes. Cell Death Dis. 2022, 13, 1–14. [Google Scholar] [CrossRef]
  170. Kerkar, S.; Speyer, C.; Tyburski, J.; Steffes, C. Reactive Oxygen Metabolites Induce a Biphasic Contractile Response in Microvascular Lung Pericytes. J. Trauma 2001, 51, 440–445. [Google Scholar] [CrossRef] [PubMed]
  171. Li, W.; Suwanwela, N.C.; Patumraj, S. Curcumin by Down-Regulating NF-κB and Elevating Nrf2, Reduces Brain Edema and Neurological Dysfunction after Cerebral I/R. Microvasc. Res. 2016, 106, 117–127. [Google Scholar] [CrossRef]
  172. Kunze, R.; Urrutia, A.; Hoffmann, A.; Liu, H.; Helluy, X.; Pham, M.; Reischl, S.; Korff, T.; Marti, H.H. Dimethyl Fumarate Attenuates Cerebral Edema Formation by Protecting the Blood-Brain Barrier Integrity. Exp. Neurol. 2015, 266, 99–111. [Google Scholar] [CrossRef]
  173. Zhao, X.; Li, S.; Mo, Y.; Li, R.; Huang, S.; Zhang, A.; Ni, X.; Dai, Q.; Wang, J. DCA Protects against Oxidation Injury Attributed to Cerebral Ischemia-Reperfusion by Regulating Glycolysis through PDK2-PDH-Nrf2 Axis. Oxid. Med. Cell. Longev. 2021, 2021, 5173035. [Google Scholar] [CrossRef]
  174. Zhao, J.; Moore, A.N.; Redell, J.B.; Dash, P.K. Enhancing Expression of Nrf2-Driven Genes Protects the Blood Brain Barrier after Brain Injury. J. Neurosci. 2007, 27, 10240–10248. [Google Scholar] [CrossRef]
  175. Sakuma, R.; Kobayashi, M.; Kobashi, R.; Onishi, M.; Maeda, M.; Kataoka, Y.; Imaoka, S. Brain Pericytes Acquire Stemness via the Nrf2-Dependent Antioxidant System. Stem Cells 2022, 40, 641–654. [Google Scholar] [CrossRef]
  176. Wang, Y.; Zhou, Z.; Wang, W.; Liu, M.; Bao, Y. Differential Effects of Sulforaphane in Regulation of Angiogenesis in a Co-Culture Model of Endothelial Cells and Pericytes. Oncol. Rep. 2017, 37, 2905–2912. [Google Scholar] [CrossRef]
  177. Li, J.-W.; Xia, L.-J.; Cui, C.-T.; Cheng, J.; Xia, Y.-B. Mechanism of Electroacupuncture in Treating Uterine Endometrial Fibrosis in Intrauterine Adhesions Rats Based on Wnt/β-Catenin Pathway-Mediated Epithelial-Mesenchymal Transition. Zhen Ci Yan Jiu 2024, 49, 566–576. [Google Scholar] [CrossRef] [PubMed]
  178. Yemisci, M.; Gursoy-Ozdemir, Y.; Vural, A.; Can, A.; Topalkara, K.; Dalkara, T. Pericyte Contraction Induced by Oxidative-Nitrative Stress Impairs Capillary Reflow despite Successful Opening of an Occluded Cerebral Artery. Nat. Med. 2009, 15, 1031–1037. [Google Scholar] [CrossRef]
  179. Andersson-Sjöland, A.; Karlsson, J.C.; Rydell-Törmänen, K. ROS-Induced Endothelial Stress Contributes to Pulmonary Fibrosis through Pericytes and Wnt Signaling. Lab. Investig. 2016, 96, 206–217. [Google Scholar] [CrossRef] [PubMed]
  180. Tambo, W.; Powell, K.; Wadolowski, S.; Unadkat, P.; Chang, E.H.; LeDoux, C.; Sciubba, D.; Wang, P.; Huerta, P.; Li, C. Vasoactive Neuropeptide Dysregulation: A Novel Mechanism of Microvascular Dysfunction in Vascular Cognitive Impairment. bioRxiv 2025, 2025.04.25.650644. [Google Scholar] [CrossRef]
  181. Paolicelli, R.C.; Sierra, A.; Stevens, B.; Tremblay, M.-E.; Aguzzi, A.; Ajami, B.; Amit, I.; Audinat, E.; Bechmann, I.; Bennett, M.; et al. Microglia States and Nomenclature: A Field at Its Crossroads. Neuron 2022, 110, 3458–3483. [Google Scholar] [CrossRef]
  182. He, F.; Ru, X.; Wen, T. NRF2, a Transcription Factor for Stress Response and Beyond. Int. J. Mol. Sci. 2020, 21, 4777. [Google Scholar] [CrossRef]
  183. Thimmulappa, R.K.; Lee, H.; Rangasamy, T.; Reddy, S.P.; Yamamoto, M.; Kensler, T.W.; Biswal, S. Nrf2 Is a Critical Regulator of the Innate Immune Response and Survival during Experimental Sepsis. J. Clin. Investig. 2016, 116, 984–995. [Google Scholar] [CrossRef]
  184. Wang, Z.; Zhang, S.; Xiao, Y.; Zhang, W.; Wu, S.; Qin, T.; Yue, Y.; Qian, W.; Li, L. NLRP3 Inflammasome and Inflammatory Diseases. Oxidative. Med. Cell. Longev. 2020, 2020, 4063562. [Google Scholar] [CrossRef]
  185. Tufekci, K.U.; Ercan, I.; Isci, K.B.; Olcum, M.; Tastan, B.; Gonul, C.P.; Genc, K.; Genc, S. Sulforaphane Inhibits NLRP3 Inflammasome Activation in Microglia through Nrf2-Mediated MiRNA Alteration. Immunol. Lett. 2021, 233, 20–30. [Google Scholar] [CrossRef]
  186. Tastan, B.; Arioz, B.I.; Tufekci, K.U.; Tarakcioglu, E.; Gonul, C.P.; Genc, K.; Genc, S. Dimethyl Fumarate Alleviates NLRP3 Inflammasome Activation in Microglia and Sickness Behavior in LPS-Challenged Mice. Front. Immunol. 2021, 12, 737065. [Google Scholar] [CrossRef]
  187. Arioz, B.I.; Tastan, B.; Tarakcioglu, E.; Tufekci, K.U.; Olcum, M.; Ersoy, N.; Bagriyanik, A.; Genc, K.; Genc, S. Melatonin Attenuates LPS-Induced Acute Depressive-Like Behaviors and Microglial NLRP3 Inflammasome Activation Through the SIRT1/Nrf2 Pathway. Front. Immunol. 2019, 10, 1511. [Google Scholar] [CrossRef]
  188. Gao, Z.; Luo, K.; Hu, Y.; Niu, Y.; Zhu, X.; Li, S.; Zhang, H. Melatonin Alleviates Chronic Stress-Induced Hippocampal Microglia Pyroptosis and Subsequent Depression-like Behaviors by Inhibiting Cathepsin B/NLRP3 Signaling Pathway in Rats. Transl. Psychiatry 2024, 14, 166. [Google Scholar] [CrossRef] [PubMed]
  189. Liu, X.; Zhang, X.; Ding, Y.; Zhou, W.; Tao, L.; Lu, P.; Wang, Y.; Hu, R. Nuclear Factor E2-Related Factor-2 Negatively Regulates NLRP3 Inflammasome Activity by Inhibiting Reactive Oxygen Species-Induced NLRP3 Priming. Antioxid. Redox Signal. 2017, 26, 28–43. [Google Scholar] [CrossRef] [PubMed]
  190. Perry, V.H. The Influence of Systemic Inflammation on Inflammation in the Brain: Implications for Chronic Neurodegenerative Disease. Brain Behav. Immun. 2004, 18, 407–413. [Google Scholar] [CrossRef] [PubMed]
  191. Dantzer, R.; O’Connor, J.C.; Freund, G.G.; Johnson, R.W.; Kelley, K.W. From Inflammation to Sickness and Depression: When the Immune System Subjugates the Brain. Nat. Rev. Neurosci. 2008, 9, 46–56. [Google Scholar] [CrossRef]
  192. Zipp, F.; Aktas, O. The Brain as a Target of Inflammation: Common Pathways Link Inflammatory and Neurodegenerative Diseases. Trends. Neurosci. 2006, 29, 518–527. [Google Scholar] [CrossRef]
  193. Barros, L.F. How Expensive Is the Astrocyte? J. Cereb. Blood Flow Metab. 2022, 42, 738–745. [Google Scholar] [CrossRef]
  194. Narayanan, S.V.; Dave, K.R.; Perez-Pinzon, M.A. Ischemic Preconditioning Protects Astrocytes against Oxygen Glucose Deprivation Via the Nuclear Erythroid 2-Related Factor 2 Pathway. Transl. Stroke Res. 2018, 9, 99–109. [Google Scholar] [CrossRef]
  195. Czyżewski, W.; Mazurek, M.; Sakwa, L.; Szymoniuk, M.; Pham, J.; Pasierb, B.; Litak, J.; Czyżewska, E.; Turek, M.; Piotrowski, B.; et al. Astroglial Cells: Emerging Therapeutic Targets in the Management of Traumatic Brain Injury. Cells 2024, 13, 148. [Google Scholar] [CrossRef]
  196. Poewe, W.; Seppi, K.; Tanner, C.M.; Halliday, G.M.; Brundin, P.; Volkmann, J.; Schrag, A.-E.; Lang, A.E. Parkinson Disease. Nat. Rev. Dis. Primers 2017, 3, 1–21. [Google Scholar] [CrossRef]
  197. Mehra, S.; Sahay, S.; Maji, S.K. α-Synuclein Misfolding and Aggregation: Implications in Parkinson’s Disease Pathogenesis. Biochim. Biophys. Acta (BBA)—Proteins Proteom. 2019, 1867, 890–908. [Google Scholar] [CrossRef]
  198. Chinta, S.J.; Mallajosyula, J.K.; Rane, A.; Andersen, J.K. Mitochondrial Alpha-Synuclein Accumulation Impairs Complex I Function in Dopaminergic Neurons and Results in Increased Mitophagy In Vivo. Neurosci. Lett. 2010, 486, 235–239. [Google Scholar] [CrossRef]
  199. Brown, D.R. α-Synuclein as a Ferrireductase. Biochem. Soc. Trans. 2013, 41, 1513–1517. [Google Scholar] [CrossRef]
  200. Cunha-Oliveira, T.; Montezinho, L.; Mendes, C.; Firuzi, O.; Saso, L.; Oliveira, P.J.; Silva, F.S.G. Oxidative Stress in Amyotrophic Lateral Sclerosis: Pathophysiology and Opportunities for Pharmacological Intervention. Oxid. Med. Cell. Longev. 2020, 2020, 5021694. [Google Scholar] [CrossRef]
  201. Hemerková, P.; Vališ, M. Role of Oxidative Stress in the Pathogenesis of Amyotrophic Lateral Sclerosis: Antioxidant Metalloenzymes and Therapeutic Strategies. Biomolecules 2021, 11, 437. [Google Scholar] [CrossRef] [PubMed]
  202. Wang, X.; Su, B.; Zheng, L.; Perry, G.; Smith, M.A.; Zhu, X. The Role of Abnormal Mitochondrial Dynamics in the Pathogenesis of Alzheimer’s Disease. J. Neurochem. 2009, 109, 153–159. [Google Scholar] [CrossRef] [PubMed]
  203. Hensley, K.; Hall, N.; Subramaniam, R.; Cole, P.; Harris, M.; Aksenov, M.; Aksenova, M.; Gabbita, S.P.; Wu, J.F.; Carney, J.M.; et al. Brain Regional Correspondence Between Alzheimer’s Disease Histopathology and Biomarkers of Protein Oxidation. J. Neurochem. 1995, 65, 2146–2156. [Google Scholar] [CrossRef]
  204. Chen, Z.; Zhong, C. Oxidative Stress in Alzheimer’s Disease. Neurosci. Bull. 2014, 30, 271–281. [Google Scholar] [CrossRef] [PubMed]
  205. Xie, H.; Hou, S.; Jiang, J.; Sekutowicz, M.; Kelly, J.; Bacskai, B.J. Rapid Cell Death Is Preceded by Amyloid Plaque-Mediated Oxidative Stress. Proc. Natl. Acad. Sci. USA 2013, 110, 7904–7909. [Google Scholar] [CrossRef]
  206. Cheignon, C.; Tomas, M.; Bonnefont-Rousselot, D.; Faller, P.; Hureau, C.; Collin, F. Oxidative Stress and the Amyloid Beta Peptide in Alzheimer’s Disease. Redox Biol. 2018, 14, 450–464. [Google Scholar] [CrossRef]
  207. Hong, X.; Huang, L.; Lei, F.; Li, T.; Luo, Y.; Zeng, M.; Wang, Z. The Role and Pathogenesis of Tau Protein in Alzheimer’s Disease. Biomolecules 2025, 15, 824. [Google Scholar] [CrossRef]
  208. Liu, Z.; Li, T.; Li, P.; Wei, N.; Zhao, Z.; Liang, H.; Ji, X.; Chen, W.; Xue, M.; Wei, J. The Ambiguous Relationship of Oxidative Stress, Tau Hyperphosphorylation, and Autophagy Dysfunction in Alzheimer’s Disease. Oxid. Med. Cell. Longev. 2015, 2015, 352723. [Google Scholar] [CrossRef]
  209. Szabo, L.; Eckert, A.; Grimm, A. Insights into Disease-Associated Tau Impact on Mitochondria. Int. J. Mol. Sci. 2020, 21, 6344. [Google Scholar] [CrossRef]
  210. Pérez, M.J.; Jara, C.; Quintanilla, R.A. Contribution of Tau Pathology to Mitochondrial Impairment in Neurodegeneration. Front. Neurosci. 2018, 12, 441. [Google Scholar] [CrossRef]
  211. Unadkat, P.; Rebeiz, T.; Ajmal, E.; De Souza, V.; Xia, A.; Jinu, J.; Powell, K.; Li, C. Neurobiological Mechanisms Underlying Psychological Dysfunction After Brain Injuries. Cells 2025, 14, 74. [Google Scholar] [CrossRef]
  212. Ren, H.; Han, R.; Liu, X.; Wang, L.; Koehler, R.C.; Wang, J. Nrf2-BDNF-TrkB Pathway Contributes to Cortical Hemorrhage-Induced Depression, but Not Sex Differences. J. Cereb. Blood Flow Metab. 2021, 41, 3288–3301. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Paradoxical consequences of electrophilic NRF2 activation. Although NRF2 coordinates the transcription of hundreds of antioxidant and cytoprotective genes, single antioxidant agents can, paradoxically, outperform classical electrophilic NRF2 inducers in therapeutic settings. This diminished efficacy arises from liabilities inherent to electrophilic compounds, including (1) depletion of intracellular glutathione, (2) exacerbation of protein oxidation, (3) amplification of reactive oxygen species, (4) disruption of mitochondrial bioenergetics, and (5) off-target organ toxicity—most notably hepatic, cardiac, and renal injury. These contraindications highlight the imperative to investigate non-electrophilic NRF2 activators. (Created with BioRender.com).
Figure 1. Paradoxical consequences of electrophilic NRF2 activation. Although NRF2 coordinates the transcription of hundreds of antioxidant and cytoprotective genes, single antioxidant agents can, paradoxically, outperform classical electrophilic NRF2 inducers in therapeutic settings. This diminished efficacy arises from liabilities inherent to electrophilic compounds, including (1) depletion of intracellular glutathione, (2) exacerbation of protein oxidation, (3) amplification of reactive oxygen species, (4) disruption of mitochondrial bioenergetics, and (5) off-target organ toxicity—most notably hepatic, cardiac, and renal injury. These contraindications highlight the imperative to investigate non-electrophilic NRF2 activators. (Created with BioRender.com).
Antioxidants 14 01047 g001
Figure 2. Mechanistic spectrum of NRF2 activation. (A) NRF2 can be activated through six principal routes: (1) non-specific electrophile/ROS generation, (2) disruption of the NRF2–KEAP1 protein–protein interface, (3) autophagy-driven KEAP1 degradation, (4) direct stabilization of NRF2, (5) post-transcriptional regulation, and (6) post-translational modification. Despite the fact that only pathway 1 is electrophilic, >90% of currently available small-molecule activators exploit electrophilic chemistry. (B) Non-pharmacological NRF2 inducers display a markedly different mechanistic distribution: they are enriched for non-electrophilic pathways and often engage multiple mechanisms concurrently. Of the twelve non-pharmacological interventions assessed in this review—excluding the two whose mechanisms have yet to be elucidated—only therapeutic hypothermia operates purely through an electrophilic mechanism. The four other interventions that exhibit some degree of electrophilicity do so in conjunction with additional, non-electrophilic pathways, highlighting their mechanistic divergence from conventional electrophile-based activators.
Figure 2. Mechanistic spectrum of NRF2 activation. (A) NRF2 can be activated through six principal routes: (1) non-specific electrophile/ROS generation, (2) disruption of the NRF2–KEAP1 protein–protein interface, (3) autophagy-driven KEAP1 degradation, (4) direct stabilization of NRF2, (5) post-transcriptional regulation, and (6) post-translational modification. Despite the fact that only pathway 1 is electrophilic, >90% of currently available small-molecule activators exploit electrophilic chemistry. (B) Non-pharmacological NRF2 inducers display a markedly different mechanistic distribution: they are enriched for non-electrophilic pathways and often engage multiple mechanisms concurrently. Of the twelve non-pharmacological interventions assessed in this review—excluding the two whose mechanisms have yet to be elucidated—only therapeutic hypothermia operates purely through an electrophilic mechanism. The four other interventions that exhibit some degree of electrophilicity do so in conjunction with additional, non-electrophilic pathways, highlighting their mechanistic divergence from conventional electrophile-based activators.
Antioxidants 14 01047 g002
Figure 3. Non-pharmacological modalities predominantly engage multiple NRF2-activating pathways. NRF2 can be induced via: (i) non-specific electrophile/ROS generation, (ii) disruption of the NRF2–KEAP1 protein–protein interaction, (iii) autophagy-mediated KEAP1 degradation, (iv) direct modulation of NRF2 protein stability, and (v) post-transcriptional/post-translational modifications. Except for a single intervention, therapeutic hypothermia, every non-pharmacological strategy with defined mechanisms employs more than one of these routes, most frequently pairing post-translational modification with either protein-stability regulation or limited electrophile production. This combinatorial activation elevates both NRF2 abundance and transcriptional competence while minimizing the liabilities of purely electrophilic agents and circumventing the efficacy limitations reported for current single-mechanism, non-electrophilic compounds. (Created with BioRender.com) (abbreviations: α7nACHr: alpha7 nicotinic acetylcholine receptor; Akt: protein kinase B; AMPK: AMP-activated protein kinase; BDNF: brain-derived neurotrophic factor; CB2r: Cannabinoid receptor 2; ERK: extracellular signal-regulated kinase; GSK-3β: glycogen synthase kinase 3 beta; miR-144: microRNA-144; p38 MAPK: p38 mitogen-activated protein kinase; p62; ROS: reactive oxygen species; SIRT1: sirtuin 1; SIRT3: sirtuin 3; STAT3: signal transducer and activator of transcription 3; TOPK: T-LAK cell-originated protein kinase).
Figure 3. Non-pharmacological modalities predominantly engage multiple NRF2-activating pathways. NRF2 can be induced via: (i) non-specific electrophile/ROS generation, (ii) disruption of the NRF2–KEAP1 protein–protein interaction, (iii) autophagy-mediated KEAP1 degradation, (iv) direct modulation of NRF2 protein stability, and (v) post-transcriptional/post-translational modifications. Except for a single intervention, therapeutic hypothermia, every non-pharmacological strategy with defined mechanisms employs more than one of these routes, most frequently pairing post-translational modification with either protein-stability regulation or limited electrophile production. This combinatorial activation elevates both NRF2 abundance and transcriptional competence while minimizing the liabilities of purely electrophilic agents and circumventing the efficacy limitations reported for current single-mechanism, non-electrophilic compounds. (Created with BioRender.com) (abbreviations: α7nACHr: alpha7 nicotinic acetylcholine receptor; Akt: protein kinase B; AMPK: AMP-activated protein kinase; BDNF: brain-derived neurotrophic factor; CB2r: Cannabinoid receptor 2; ERK: extracellular signal-regulated kinase; GSK-3β: glycogen synthase kinase 3 beta; miR-144: microRNA-144; p38 MAPK: p38 mitogen-activated protein kinase; p62; ROS: reactive oxygen species; SIRT1: sirtuin 1; SIRT3: sirtuin 3; STAT3: signal transducer and activator of transcription 3; TOPK: T-LAK cell-originated protein kinase).
Antioxidants 14 01047 g003
Figure 4. Cell-type-dependent NRF2 engagement. Neurons, oligodendrocytes, astrocytes, microglia, and vascular cells possess distinct NRF2 regulatory baselines that reflect their intrinsic oxidative stress susceptibility. Consequently, the optimal route of NRF2 induction varies across cell types and, by extension, across neurological diseases dominated by those cells. Although direct cell-specific data for non-pharmacological interventions are not yet available, the signaling pathways they recruit provide a rationale for therapeutic matching. Modalities such as physical exercise and DR activation, which primarily engage neuronal and oligodendroglial NRF2 signaling, may be preferable for disorders characterized by parenchymal injury. In contrast, interventions that robustly influence vascular NRF2 networks, e.g., electroacupuncture or electroconvulsive stimulation, may prove more advantageous in pathologies driven by cerebrovascular dysfunction. (Created with BioRender.com).
Figure 4. Cell-type-dependent NRF2 engagement. Neurons, oligodendrocytes, astrocytes, microglia, and vascular cells possess distinct NRF2 regulatory baselines that reflect their intrinsic oxidative stress susceptibility. Consequently, the optimal route of NRF2 induction varies across cell types and, by extension, across neurological diseases dominated by those cells. Although direct cell-specific data for non-pharmacological interventions are not yet available, the signaling pathways they recruit provide a rationale for therapeutic matching. Modalities such as physical exercise and DR activation, which primarily engage neuronal and oligodendroglial NRF2 signaling, may be preferable for disorders characterized by parenchymal injury. In contrast, interventions that robustly influence vascular NRF2 networks, e.g., electroacupuncture or electroconvulsive stimulation, may prove more advantageous in pathologies driven by cerebrovascular dysfunction. (Created with BioRender.com).
Antioxidants 14 01047 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Li, C.; Powell, K.; Giliberto, L.; LeDoux, C.; d’Abramo, C.; Sciubba, D.; Al Abed, Y. Non-Electrophilic Activation of NRF2 in Neurological Disorders: Therapeutic Promise of Non-Pharmacological Strategies. Antioxidants 2025, 14, 1047. https://doi.org/10.3390/antiox14091047

AMA Style

Li C, Powell K, Giliberto L, LeDoux C, d’Abramo C, Sciubba D, Al Abed Y. Non-Electrophilic Activation of NRF2 in Neurological Disorders: Therapeutic Promise of Non-Pharmacological Strategies. Antioxidants. 2025; 14(9):1047. https://doi.org/10.3390/antiox14091047

Chicago/Turabian Style

Li, Chunyan, Keren Powell, Luca Giliberto, Christopher LeDoux, Cristina d’Abramo, Daniel Sciubba, and Yousef Al Abed. 2025. "Non-Electrophilic Activation of NRF2 in Neurological Disorders: Therapeutic Promise of Non-Pharmacological Strategies" Antioxidants 14, no. 9: 1047. https://doi.org/10.3390/antiox14091047

APA Style

Li, C., Powell, K., Giliberto, L., LeDoux, C., d’Abramo, C., Sciubba, D., & Al Abed, Y. (2025). Non-Electrophilic Activation of NRF2 in Neurological Disorders: Therapeutic Promise of Non-Pharmacological Strategies. Antioxidants, 14(9), 1047. https://doi.org/10.3390/antiox14091047

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop