Your new experience awaits. Try the new design now and help us make it even better

REVIEW article

Front. Nutr., 05 December 2025

Sec. Nutrition, Psychology and Brain Health

Volume 12 - 2025 | https://doi.org/10.3389/fnut.2025.1667421

This article is part of the Research TopicThe Foundational Components and Elements of Plant Foods for Neurological Nutrition and Well-beingView all 13 articles

Plant food-derived antioxidant nutrients in neuroprotection against chemotherapy-induced neurotoxicity: from molecular mechanisms to clinical applications

Weizhong Li&#x;Weizhong Li1Dan Yang&#x;Dan Yang2Zihan Zhang&#x;Zihan Zhang3Meizi WangMeizi Wang3Senlei XuSenlei Xu3Ruiyang JiangRuiyang Jiang3Jin ChenJin Chen2Chaokui Wu
Chaokui Wu4*Jun Qian
Jun Qian1*
  • 1Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
  • 2Department of Obstetrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
  • 3Nanjing University of Chinese Medicine, Nanjing, China
  • 4The First Affiliated Hospital of Guangzhou University of Chinese Medicine (Chongqing Beibei District Hospital of Traditional Chinese Medicine), Chongqing, China

Chemotherapy-induced peripheral neuropathy (CIPN) is one of the most challenging dose-limiting toxicities in contemporary oncology, affecting 19–85% of patients receiving neurotoxic chemotherapy agents. The pathophysiological mechanisms of CIPN are complex, involving multiple interconnected processes including oxidative stress, neuroinflammation, neurotrophic factor depletion, and mitochondrial dysfunction. Plant-derived antioxidant nutrients have emerged as promising candidates for CIPN prevention due to their unique multi-target neuroprotective capabilities. This comprehensive review systematically analyzes the molecular protective mechanisms of plant-derived nutrients, evaluates existing clinical evidence, and discusses practical application strategies. The focus is on the neuroprotective effects of curcumin, green tea catechins, vitamin E, and other plant compounds. Evidence indicates that these compounds exert protective effects through activation of endogenous antioxidant systems, modulation of inflammatory pathways, enhancement of neurotrophic factors, and protection of mitochondrial function. While clinical evidence is still accumulating, preliminary studies show encouraging results, providing a scientific basis for developing plant food-based CIPN prevention strategies.

1 Introduction

Chemotherapy-induced peripheral neuropathy (CIPN) is a common and debilitating dose-limiting toxicity, affecting 19–85% of patients treated with neurotoxic agents (13). Its sensorimotor symptoms—including numbness, tingling, and burning pain—often force dose reductions or discontinuation, compromising cancer outcomes (4, 5). Current therapies offer only modest symptom relief and fail to address the underlying mechanisms (6, 7) (Table 1).

Table 1
www.frontiersin.org

Table 1. Summary of chemotherapy-induced neurotoxicity mechanisms by drug class.

The pathogenesis of CIPN is multifactorial, involving oxidative stress, neuroinflammation, neurotrophic factor depletion, and mitochondrial dysfunction (610). These convergent processes overwhelm the limited regenerative capacity of the peripheral nervous system, indicating that effective interventions must target multiple pathways simultaneously rather than isolated mechanisms (11).

Plant-derived foods have emerged as promising candidates for CIPN prevention due to their unique multi-target antioxidant properties. Their bioactive compounds can alleviate oxidative stress, modulate inflammatory cascades, preserve mitochondrial integrity, and enhance neurotrophic signaling, while offering safety, accessibility, and broad patient acceptance (12, 13). Advances in precision nutrition and nutrigenomics further expand their potential by enabling personalized interventions tailored to genetic and metabolic profiles (1416).

This comprehensive review examines the current state of knowledge regarding plant food-derived nutrients in CIPN prevention and management. We analyze molecular mechanisms, evaluate clinical evidence, and discuss practical applications while identifying key research priorities for clinical translation.

2 Pathophysiological foundations of chemotherapy-induced neurotoxicity

The development of CIPN involves multiple convergent mechanisms that fundamentally alter the neurobiological landscape of peripheral nervous system function. These mechanisms arise from direct chemotherapy-induced cellular damage but subsequently evolve into self-perpetuating cycles of dysfunction and impaired repair (68, 17). Understanding these interconnected pathways provides essential context for evaluating how plant food-derived nutrients can effectively intervene in disease progression and support neural recovery (11, 18, 19).

2.1 Oxidative stress and cellular antioxidant depletion

The peripheral nervous system demonstrates exceptional vulnerability to chemotherapy-induced oxidative damage due to its unique structural and metabolic characteristics, including long axonal transport distances, high polyunsaturated fatty acid content in neural membranes as major targets for lipid peroxidation, and weaker blood–nerve barrier protection compared with central structures (9, 2023). Chemotherapy agents initiate oxidative stress through diverse pathways that collectively overwhelm cellular antioxidant defenses, with platinum-based compounds generating excessive reactive oxygen species through direct DNA interaction and mitochondrial dysfunction while rapidly depleting glutathione stores that serve as the primary intracellular antioxidant defense system (6, 24) (Figure 1).

Figure 1
Diagram illustrating the generation and role of reactive oxygen species (ROS) in cellular processes. The NADPH oxidase complex produces superoxide (O2•−), which is converted by SOD1 to hydrogen peroxide (H2O2). H2O2 can diffuse to the mitochondria and undergo the Fenton reaction, creating hydroxyl radicals (OH•) and causing ROS diffusion to the nucleus. Mitochondrial complex I/III also generates superoxide, which is further converted by SOD2. Protective enzymes GSH, GPx, CTA, and GR are involved in detoxifying ROS.

Figure 1. Chemotherapy exposure generates reactive oxygen species (ROS) via nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and mitochondrial dysfunction, overwhelming antioxidant systems including glutathione (GSH), superoxide dismutase (SOD), and catalase. Abbreviations: ROS, reactive oxygen species; NADPH, nicotinamide adenine dinucleotide phosphate; GSH, glutathione; SOD, superoxide dismutase. Created with Figdraw (figdraw.com).

Taxane agents contribute through distinct mechanisms involving microtubule disruption and impaired axonal transport of antioxidant enzymes and protective molecules. This creates localized oxidative stress hotspots in distal nerve regions, explaining the characteristic length-dependent pattern of taxane-induced neuropathy (9, 25). Anthracycline compounds directly target mitochondrial electron transport chains, generating superoxide radicals and hydrogen peroxide while simultaneously compromising ATP-dependent antioxidant regeneration systems. This creates vicious cycles where mitochondrial damage produces more oxidants that further impair cellular function and perpetuate neuronal dysfunction (24, 26).

The temporal dynamics of chemotherapy-induced oxidative stress involve both acute phases occurring during and immediately after treatment administration, characterized by rapid antioxidant depletion and damage marker accumulation, and chronic phases persisting long after treatment completion, driven by sustained mitochondrial dysfunction, impaired antioxidant enzyme expression, and ongoing inflammatory processes (6, 27). This persistent oxidative imbalance contributes significantly to the chronic nature of CIPN and its resistance to conventional therapeutic approaches (7).

2.2 Neuroinflammatory networks and immune system activation

Neuroinflammation represents a critical pathophysiological mechanism that amplifies and perpetuates neural damage beyond the direct cytotoxic effects of chemotherapy treatment through complex interactions between immune cells, neural tissue, and inflammatory mediators that create self-reinforcing cycles of damage and impaired repair (10, 28). The inflammatory cascade begins with chemotherapy-induced tissue damage that releases damage-associated molecular patterns, including high mobility group box 1, ATP, and DNA fragments, which activate pattern recognition receptors on resident macrophages and Schwann cells that respond by producing pro-inflammatory cytokines including tumor necrosis factor-alpha, interleukin-1 beta, and interleukin-6 (2830).

These inflammatory mediators recruit additional immune cells and disrupt the blood-nerve barrier, allowing systemic inflammatory mediators to enter peripheral nerve tissue and further exacerbate local inflammation while creating a permissive environment for ongoing neural damage (29, 3133). Nuclear factor-kappa B serves as a central mediator of chemotherapy-induced neuroinflammation, coordinating the expression of multiple pro-inflammatory genes in response to cellular stress and damage through multiple activation pathways including oxidative stress, DNA damage responses, and cytokine signaling cascades (28, 34).

Once activated, nuclear factor-kappa B translocates to the nucleus and initiates transcription of genes encoding inflammatory cytokines, chemokines, and enzymes such as cyclooxygenase-2 and inducible nitric oxide synthase that perpetuate inflammatory processes and directly damage neurons while impairing neurotrophic factor signaling and axonal regeneration (28, 34). Critically, chemotherapy-induced neuroinflammation often persists long after treatment completion, contributing to the chronic nature of CIPN and representing a potential therapeutic target for interventions aimed at breaking the cycle of ongoing inflammation and neural dysfunction.

2.3 Neurotrophic factor dysregulation and regenerative capacity impairment

The depletion of neurotrophic factors represents a crucial mechanism contributing to CIPN development and progression, as these essential proteins for neural survival, growth, differentiation, and synaptic plasticity demonstrate marked reductions during chemotherapy treatment through both direct suppression of gene expression by chemotherapy agents and indirect consequences of inflammation and oxidative stress that create a hostile environment for neurotrophic factor synthesis and signaling (6, 8, 3538). Brain-derived neurotrophic factor and nerve growth factor, which are particularly important for maintaining sensory neurons predominantly affected in CIPN, show dramatic decreases that not only impair neuronal survival and increase susceptibility to chemotherapy-induced damage but also compromise the regenerative capacity of peripheral nerves, contributing to the persistent nature of CIPN and limiting recovery potential even after treatment completion (17, 3943).

This neurotrophic factor deficiency creates a cascade of dysfunction that extends from direct effects on neuronal survival and growth to broader impacts on synaptic plasticity, pain processing, and neural adaptation mechanisms that are essential for maintaining normal sensory function (4447). The spatial and temporal patterns of neurotrophic factor depletion correlate closely with the characteristic distal-to-proximal progression of neuropathy symptoms, suggesting that restoration of neurotrophic support represents a critical therapeutic target for both prevention and treatment of CIPN (4850).

2.4 Mitochondrial bioenergetic crisis and cellular energy metabolism

Mitochondrial dysfunction emerges as a fundamental mechanism underlying chemotherapy-induced neurotoxicity, as these organelles serve as both primary targets of chemotherapy-induced damage and critical determinants of neuronal survival and function, with the peripheral nervous system’s exceptionally high energy demands making it particularly vulnerable to mitochondrial impairment since neurons rely heavily on oxidative phosphorylation for ATP generation and lack the glycolytic capacity to compensate for mitochondrial dysfunction (39, 5154). Chemotherapy agents affect mitochondrial function through multiple interconnected mechanisms, including direct accumulation in mitochondria with formation of DNA crosslinks that impair replication and transcription, disruption of mitochondrial dynamics and transport processes, and direct inhibition of electron transport complexes, leading to ATP depletion and enhanced reactive oxygen species production that triggers apoptotic pathways and neuronal death (5561) (Figure 2).

Figure 2
Diagram illustrating mitochondrial metabolism and related processes. Glucose is converted to ATP, lactate, and acetyl-CoA, feeding into the TCA cycle. Outputs include citrate, succinyl-CoA, and oxaloacetate. Other processes involve cholesterol, lipid, and glutamine synthesis. ATP production affects mtDNA, with damage linked to mutations. Reactive oxygen species (ROS) and superoxide (O2-) are shown as byproducts. A secondary box outlines the conversion of O2- to H2O2 mediated by enzymes. Components like Cyt C, ATP synthase, MCU, ANT, and complexes I/III are labeled.

Figure 2. Chemotherapy disrupts mitochondrial electron transport chain complexes and the tricarboxylic acid (TCA) cycle, leading to mitochondrial DNA (mtDNA) damage, adenosine triphosphate (ATP) depletion, calcium (Ca2+) overload, and loss of mitochondrial membrane potential (ΔΨm). Abbreviations: TCA, tricarboxylic acid; mtDNA, mitochondrial DNA; ATP, adenosine triphosphate;ΔΨm, mitochondrial membrane potential. Created with Figdraw (figdraw.com).

The bioenergetic crisis particularly affects distal nerve segments where energy demands are highest and mitochondrial transport distances are greatest, explaining the characteristic length-dependent pattern of chemotherapy-induced neuropathy and its predilection for sensory neurons that have particularly long axons and high metabolic demands (6264) (Figure 3). Mitochondrial dysfunction also impairs the cell’s ability to maintain calcium homeostasis, support axonal transport, and synthesize essential proteins and lipids needed for membrane maintenance and repair, creating a cascade of cellular dysfunction that extends far beyond simple energy depletion to encompass fundamental aspects of neuronal structure and function (6568).

Figure 3
Diagram illustrating inflammatory pathways in the brain. It shows neutrophils and leukocytes moving from a capillary into the brain, causing DNA damage and infiltration. Drugs, ATP, and HMGB1 are labeled as influencing these cells. Reactive Schwann cells and activated macrophages release cytokines affecting NF-κB pathways, leading to oxidative stress and mitochondrial dysfunction. This process impacts sensory neurons, causing distal axon degeneration and affecting the blood-nerve barrier.

Figure 3. Chemotherapy damages the blood–nerve barrier, releasing damage-associated molecular patterns (DAMPs) such as high-mobility group box 1 (HMGB1), which activate Schwann cells and macrophages and induce nuclear factor-kappa B (NF-κB)-mediated cytokine production including tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β), and interleukin-6 (IL-6), ultimately injuring dorsal root ganglion (DRG) neurons. Abbreviations: DAMPs, damage-associated molecular patterns; HMGB1, high-mobility group box 1; NF-κB, nuclear factor-kappa B; TNF-α, tumor necrosis factor-alpha; IL-1β, interleukin-1 beta; IL-6, interleukin-6; DRG, dorsal root ganglion. Created with Figdraw (figdraw.com).

3 Plant food Neuroprotective mechanisms: molecular targets and pathways

3.1 Endogenous antioxidant system activation

Plant polyphenolic compounds provide neuroprotection primarily through activation of the nuclear factor erythroid 2-related factor 2/antioxidant response element (Nrf2/ARE) pathway (69, 70) (Table 2). This pathway serves as the master regulator of cellular antioxidant defenses (71). Under normal conditions, Nrf2 remains sequestered in the cytoplasm by Kelch-like ECH-associated protein 1 (Keap1) (72). Keap1 facilitates rapid Nrf2 degradation through the ubiquitin-proteasome system (73).

Table 2
www.frontiersin.org

Table 2. Comparative analysis of plant food compounds for neuroprotection.

Plant polyphenolic compounds disrupt this regulatory interaction through multiple mechanisms. They form direct covalent modifications of Keap1 cysteine residues (70, 74). They also promote phosphorylation of Nrf2 by upstream kinases and create competitive inhibition of protein–protein binding interactions (75, 76). Once released from Keap1-mediated repression, Nrf2 translocates to the nucleus (70, 74). There it forms heterodimers with small Maf proteins and binds to antioxidant response element sequences (77, 78).

This transcriptional activation upregulates numerous cytoprotective enzymes. These collectively enhance cellular resistance to oxidative stress and toxic insults (79, 80). Key enzymes include heme oxygenase-1, which catalyzes heme degradation to produce powerful antioxidant and anti-inflammatory products (81, 82). NAD(P)H: quinone oxidoreductase 1 functions as a two-electron reductase preventing quinone participation in redox cycling reactions (83, 84). Glutathione S-transferases facilitate glutathione conjugation to electrophilic compounds for detoxification and elimination (85, 86).

Curcumin exemplifies the potent Nrf2-activating potential of plant polyphenols. Its diferuloylmethane structure contains multiple electrophilic sites that form covalent adducts with Keap1 cysteine residues (87, 88). This leads to conformational changes that disrupt Nrf2 binding. Curcumin simultaneously activates upstream kinases including protein kinase C and mitogen-activated protein kinases. These phosphorylate Nrf2 and promote its nuclear translocation (8991). This mechanism is particularly relevant in CIPN, as Nrf2-driven upregulation of glutathione and phase II detoxification enzymes directly counteracts platinum-induced glutathione depletion, while enhancement of antioxidant enzymes such as SOD and catalase mitigates the distal oxidative stress hotspots characteristic of taxane-induced neuropathy.

3.2 Anti-inflammatory pathway modulation

Plant polyphenols demonstrate potent anti-inflammatory activity through inhibition of nuclear factor-kappa B signaling (92). This represents the key transcriptional pathway coordinating inflammatory gene expression in response to cellular stress (93). Curcumin functions as a particularly effective nuclear factor-kappa B inhibitor. It directly binds to nuclear factor-kappa B subunits, preventing their nuclear translocation and DNA binding (94). It also inhibits IκB kinase activity, preventing phosphorylation and degradation of IκB proteins (95).

The dual action of curcumin creates synergistic protective effects particularly relevant to chemotherapy-induced neuroinflammation prevention. It simultaneously activates anti-inflammatory Nrf2 pathways while inhibiting pro-inflammatory nuclear factor-kappa B signaling (9698). This mechanistic duality explains curcumin’s demonstrated efficacy in reducing inflammatory cytokine expression and preserving nerve function in experimental neuropathy models (99101).

Resveratrol from grapes and berries demonstrates anti-inflammatory effects through sirtuin 1 pathway activation (102). This deacetylates and inactivates nuclear factor-kappa B subunits while promoting cellular stress resistance (103, 104). Enhanced DNA repair and metabolic optimization provide sustained anti-inflammatory effects that promote cellular adaptation to stress (105109).

Green tea catechins provide complementary anti-inflammatory activity through multiple pathways. These include direct inhibition of inflammatory enzymes such as cyclooxygenase-2 and lipoxygenase (110, 111). They also suppress inflammatory cytokine production and modulate immune cell activation patterns (112, 113). These compounds enhance the production of anti-inflammatory mediators and promote active resolution of inflammatory processes. This anti-inflammatory action directly suppresses the NF-κB–mediated cytokine cascades that drive persistent neuroinflammation and amplify chemotherapy-induced neural damage.

3.3 Neurotrophic factor enhancement systems

Plant foods contain numerous compounds that specifically support neurotrophic factor synthesis and signaling. These provide essential nutritional support for neural survival, growth, and repair processes (114117). Flavonoid compounds from various plant sources demonstrate the ability to cross the blood–brain barrier (116). They directly stimulate brain-derived neurotrophic factor synthesis in neural tissue through activation of cAMP response element-binding protein pathways (115, 116, 118, 119). This transcriptional activation drives brain-derived neurotrophic factor gene transcription via phosphorylation-dependent mechanisms. The result is increased brain-derived neurotrophic factor expression that enhances neuronal survival. It also promotes axonal growth and supports synaptic plasticity essential for neural adaptation and recovery (120123).

Soy isoflavones, particularly genistein and daidzein, demonstrate unique neurotrophic properties (124126). These occur through their selective estrogen receptor modulator activities (127129). They bind preferentially to estrogen receptor beta, which is highly expressed in neural tissue (130, 131). This promotes brain-derived neurotrophic factor gene transcription via estrogen response elements (132135). The result is increased brain-derived neurotrophic factor protein synthesis and release (133, 135137).

The neurotrophic effects of soy isoflavones extend beyond brain-derived neurotrophic factor modulation. They enhance nerve growth factor synthesis in Schwann cells, the primary supportive cells of peripheral nerves (138140). They also promote expression of neurotrophin-3 and neurotrophin-4 that support different populations of sensory neurons (141). This makes these compounds particularly valuable for preventing and treating peripheral neuropathy. This neurotrophic support directly addresses the depletion of BDNF and NGF that limits peripheral nerve survival and regenerative capacity during chemotherapy.

3.4 Mitochondrial protection and bioenergetic enhancement

Plant foods provide mitochondrial protective nutrients that deliver essential bioenergetic support (142145). These can prevent chemotherapy-induced mitochondrial damage and promote neural recovery through enhanced cellular energy production and reduced oxidative stress. α-Lipoic acid, found naturally in spinach, broccoli, and other green vegetables, functions as a unique mitochondrial antioxidant (146, 147).

This compound possesses both lipophilic and hydrophilic properties that allow comprehensive protection in both aqueous and lipid phases (148). It serves as an essential cofactor for mitochondrial enzyme complexes including pyruvate dehydrogenase and α-ketoglutarate dehydrogenase (149). These catalyze crucial steps in cellular energy production. α-Lipoic acid also functions as a potent antioxidant that directly scavenges reactive oxygen species while regenerating other antioxidants (150, 151).

The protective effects extend to activation of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α). This serves as the master regulator of mitochondrial biogenesis (152, 153). PGC-1α activation promotes transcription of nuclear genes encoding mitochondrial proteins while enhancing mitochondrial DNA replication and protein synthesis (154, 155).

Coenzyme Q10, obtained from plant foods including spinach, cauliflower, and whole grains, serves as an essential component of the mitochondrial electron transport chain (156). It shuttles electrons between Complex I/II and Complex III while playing a crucial role in oxidative phosphorylation and ATP synthesis (157, 158). Coenzyme Q10 also functions as a powerful lipid-phase antioxidant that protects mitochondrial membranes from oxidative damage (158, 159).

Pyrroloquinoline quinone (PQQ), found in fermented soy products, green tea, and various fruits and vegetables, demonstrates unique mitochondrial protective properties (160). It functions as a redox cofactor for various enzymes while demonstrating potent antioxidant activity and mitochondrial biogenesis-promoting effects (161). PQQ can directly activate PGC-1α through cAMP/cAMP response element-binding protein signaling pathways. This leads to enhanced mitochondrial biogenesis and improved cellular bioenergetic capacity (162). These protective effects directly oppose the mitochondrial bioenergetic failure caused by platinum- and taxane-induced impairment of oxidative phosphorylation, calcium homeostasis, and axonal energy supply.

4 Clinical evidence landscape for plant food interventions

The translation of preclinical neuroprotective mechanisms into clinical applications has generated substantial research interest and a growing body of evidence. However, direct clinical evidence for plant food interventions in chemotherapy-induced peripheral neuropathy prevention remains limited compared to the extensive and consistently positive preclinical foundation (163165). This highlights the critical need for well-designed clinical trials that can bridge the gap between mechanistic understanding and therapeutic application while addressing the unique challenges associated with studying complex, multi-component dietary interventions in cancer patient populations.

4.1 Turmeric and curcumin: evidence chain validation

Curcumin is the plant-derived compound with the most comprehensive validation for CIPN prevention, anchored by a landmark Level I pediatric randomized trial and further supported by cross-disease and mechanistic studies (166). This double-blind study enrolled 141 children aged 5–15 years with acute lymphoblastic leukemia receiving vincristine-based chemotherapy protocols. Patients randomized to oral curcumin supplementation at 3 mg/kg twice daily for 3 months demonstrated significant reduction in vincristine-induced peripheral neuropathy incidence from 70.0% in the placebo group to 39.4% in the treatment group (Table 3). Beyond incidence reduction, curcumin treatment resulted in significantly improved Total Neuropathy Score-Pediatric Vincristine (TNS-PV) evaluations and reduced motor nerve electrophysiological abnormalities, providing objective validation of genuine neuroprotective effects.

Table 3
www.frontiersin.org

Table 3. Clinical evidence summary for plant food interventions.

Cross-disease validation arises from neuropathies that share convergent mechanisms. In patients with type 2 diabetes-associated sensorimotor polyneuropathy, nano-curcumin 80 mg once daily for 8 weeks significantly reduced HbA1c and fasting plasma glucose while improving the total neuropathy score, reflexes and skin temperature (167). Although median nerve conduction velocity was not assessed, the concordant amelioration of clinical signs and symptoms provides indirect yet compelling support for curcumin’s capacity to counter peripheral nerve dysfunction via antioxidant and anti-inflammatory pathways. Such cross-condition efficacy strengthens the translational case for curcumin as a broad neuroprotective agent against disorders that share common pathophysiological drivers (168170).

Extensive animal data establish curcumin’s pharmacological foundation across multiple chemotherapy-induced neuropathy models. In paclitaxel-induced peripheral neuropathy mice, dietary supplementation with 1.5% curcumin-phospholipid complex (Meriva®) completely blocked both mechanical and cold pain hypersensitivity (171). Mechanistic investigations revealed that this protection operates through the α7 nicotinic acetylcholine receptor (α7 nAChR) signaling axis, which suppresses spinal cord inflammatory responses. The specificity of this mechanism was definitively established through α7 nAChR gene knockout mice studies, where curcumin’s protective effects were completely eliminated. Administration of α7 nAChR receptor antagonists similarly abolished the neuroprotective benefits, confirming α7 nAChR as the critical molecular target.

Validation across different chemotherapy agents is robustly demonstrated in models of cisplatin-, vincristine-, and oxaliplatin-induced neuropathy. In oxaliplatin models, curcumin dose-dependently alleviates mechanical and cold hypersensitivity, improves both motor (MNCV) and sensory (SNCV) nerve conduction velocities, and repairs damaged spinal neurons. This protection is mechanistically linked to its ability to enhance endogenous antioxidant enzymes (SOD, GSH-Px, CAT) while reducing lipid peroxidation (MDA), and critically, to suppress neuroinflammation by inhibiting the activation of the NF-κB pathway and its downstream inflammatory cytokines (TNF-α, IL-1β, IL-6) (98). Similarly, in vincristine-induced neuropathy, advanced formulations like curcumin nano-emulsions significantly reduce cold and thermal hyperalgesia by boosting antioxidant (SOD, CAT) and anti-inflammatory (IL-10) markers, while downregulating NF-κB expression in the sciatic nerve (172). In cisplatin models, curcumin nanoparticles effectively counteract neurotoxicity by normalizing levels of lipid peroxidation, TNF-α, and caspase-3, and restoring depleted glutathione and Na+, K + -ATPase activity (173, 174). Collectively, these studies confirm that curcumin’s efficacy stems from its multi-target ability to mitigate oxidative stress, suppress the NF-κB-driven neuroinflammatory cascade, and preserve both the structure and function of peripheral nerves.

The curcumin evidence represents the most comprehensive clinical validation among plant-derived neuroprotective compounds, with the pediatric randomized controlled trial providing Level I evidence for efficacy, cross-condition validation demonstrating broader therapeutic potential, and robust preclinical data elucidating specific molecular mechanisms through α7 nAChR-Nrf2 dual pathway targeting. This evidence integration demonstrates completion of a comprehensive “clinical-preclinical-mechanistic” validation loop that positions curcumin as having achieved the most complete validation profile among plant-derived neuroprotective compounds.

4.2 Green tea: epidemiological and intervention evidence

Green tea catechins represent an emerging intervention with promising evidence from related neuropathies and ongoing clinical trials. Green tea is a widely consumed polyphenolic beverage with substantial neuroprotective potential (175177). Green tea contains a complex mixture of bioactive compounds with catechins representing the predominant polyphenolic constituents. Epigallocatechin gallate (EGCG) comprises 30–60% of total catechins and demonstrates the most potent neuroprotective activities through its ability to cross the blood–brain barrier and accumulate in neural tissue (175, 178).

Robust epidemiological evidence provides a strong population-level basis for green tea’s neuroprotective effects. Large-scale, long-term observational studies consistently link green tea consumption with superior nervous system health, showing an inverse association with the risk of cognitive decline and neurodegenerative diseases such as Alzheimer’s and Parkinson’s disease (179182). These real-world data offer a compelling rationale for investigating green tea as a targeted intervention.

Preclinical research provides clear mechanistic support for EGCG’s protective role against CIPN and related chemotherapy-induced toxicities. In mouse models of cisplatin-induced nephrotoxicity, a condition sharing oxidative stress pathways with CIPN, EGCG was shown to significantly mitigate tissue damage by protecting mitochondrial function, restoring electron transport chain activity, enhancing mitochondrial antioxidant enzymes (e.g., GPX and MnSOD), and suppressing inflammation via the NF-κB pathway (183185). Similar protective effects against cisplatin-induced hepatotoxicity have also been observed when EGCG is combined with Coenzyme Q10 (186). Furthermore, demonstrating a direct neuro-regenerative potential, green tea polyphenols, particularly EGCG, have been shown to enhance nerve growth factor (NGF)-induced neurite outgrowth in cell culture models (187, 188).

While large-scale clinical trials for CIPN are still emerging, compelling evidence comes from studies on related neuropathies. A randomized controlled trial in patients with mild-to-moderate diabetic peripheral neuropathy (DPN), a condition with similar pathological mechanisms to CIPN, found that 16 weeks of green tea extract supplementation resulted in significant improvements compared to placebo (189). Specifically, patients experienced reductions in pain (Visual Analogue Scale, VAS), total symptoms (Total Symptom Score, TCSS), and vibration perception thresholds (VPT), with benefits becoming apparent as early as 8 weeks into treatment.

In the context of oncology, the evidence is moving from indirect support to direct validation. A preliminary clinical study in liver cancer patients undergoing chemotherapy found that daily supplementation with green tea polyphenol tablets (equivalent to ~474 mg of polyphenols) significantly reduced treatment-induced oxidative stress (190). More significantly, a frontier of clinical research has been opened with the initiation of a Phase 1/2 clinical trial (NCT06524609) specifically designed to evaluate a topical EGCG solution for the prevention and treatment of taxane-induced peripheral neuropathy (TIPN). The launch of this trial marks a critical step in translating EGCG’s potential into a viable clinical intervention for CIPN.

A particularly compelling aspect of EGCG is its dual functionality in cancer therapy: it not only protects normal tissues from chemotherapy-induced damage but can also enhance the cytotoxic effects of chemotherapy on cancer cells (191, 192). This chemosensitizing effect is achieved through multiple mechanisms. For instance, EGCG has been shown to enhance the efficacy of platinum-based drugs like cisplatin and oxaliplatin by inducing autophagy in colorectal cancer cells (193). Other studies have demonstrated that EGCG can increase cancer cell sensitivity to cisplatin by inhibiting the DNA repair enzyme ERCC1/XPF, or by upregulating the platinum drug transporter CTR1, thereby increasing intracellular drug accumulation in lung and ovarian cancer cells (194). This unique ability to both protect nerves and potentiate anti-cancer treatment makes EGCG a highly attractive candidate for adjuvant therapy.

4.3 Nuts and seeds: vitamin E clinical trial lessons

Despite promising mechanistic rationale, the clinical evidence for Vitamin E presents a more complex and sometimes contradictory picture. Evidence for vitamin E’s neuroprotective potential comes from multiple randomized trials with mixed results but important methodological insights (195204). These studies highlight important considerations regarding supplement forms, dosing strategies, patient populations, and study methodologies that have significant implications for future research and clinical application.

The landmark positive study by Pace et al. randomized 108 patients receiving cisplatin-based chemotherapy to vitamin E supplementation at 400 mg daily or placebo. The study demonstrated significantly lower incidence of peripheral neuropathy in the vitamin E group compared to placebo with better preserved nerve conduction parameters and reduced symptom severity scores (197). However, larger subsequent trials have produced conflicting results that highlight the complexity of translating promising preliminary findings into consistent clinical benefits (200). These contradictory results highlight several important methodological considerations including the use of different vitamin E forms (synthetic α-tocopherol versus natural mixed tocopherols), varying dosing regimens, diverse patient populations with different baseline nutritional status, different chemotherapy agents with distinct mechanisms of neurotoxicity, and heterogeneous neuropathy assessment methods (195, 196, 205). Synthetic α-tocopherol supplementation may deplete or competitively inhibit γ-tocopherol and tocotrienols, thereby diminishing overall antioxidant defense, whereas whole-food sources provide these compounds in physiologic ratios with additional polyphenols, minerals, and unsaturated fatty acids that act synergistically to stabilize neuronal membranes and modulate inflammation (206210).

The contradictory clinical evidence strongly emphasizes the importance of obtaining vitamin E from whole food sources rather than isolated synthetic supplements. Nuts and seeds provide vitamin E in natural ratios with other tocopherols and tocotrienols along with complementary antioxidants, minerals, and bioactive compounds that may enhance protective effects through synergistic interactions (211, 212). Natural vitamin E from food sources also provides more sustained tissue levels through gradual absorption compared to rapid absorption and clearance associated with high-dose supplement administration (213216).

Plant-based omega-3 sources from walnuts, flaxseeds, and chia seeds contribute to anti-inflammatory neuroprotection through α-linolenic acid that serves as a precursor for longer-chain omega-3 fatty acids (217, 218). A double-blind RCT (NCT01049295) showed that 640 mg omega-3 TID during paclitaxel therapy kept 70% of breast-cancer patients neuropathy-free versus 40.7% with placebo, underscoring the protective potential of essential n-3 fatty acids (219).

4.4 Fruits and vegetables: carotenoid and anthocyanin evidence

For carotenoids and anthocyanins, clinical data remain limited, but preclinical and cognitive studies provide supportive evidence. Colorful fruits and vegetables provide carotenoids and anthocyanins with specialized neuroprotective properties that demonstrate significant promise in preclinical studies. Direct clinical evidence in chemotherapy-induced peripheral neuropathy remains limited, but supportive evidence from neurodegeneration studies suggests broader protective potential (220222). Dark leafy greens provide lutein and zeaxanthin, xanthophyll carotenoids that cross the blood–brain barrier and accumulate in neural tissue (223225).

A meta-analysis of 4,402 older adults demonstrated that carotenoid supplementation significantly improved global cognition (226); large cross-sectional datasets such as NHANES likewise associate higher intakes of α- and β-carotene, lutein and zeaxanthin with better performance in memory, processing speed and attention tests (227, 228). The mechanistic foundation for carotenoid neuroprotection stems from their unique molecular structure and tissue distribution patterns (229231). These compounds demonstrate preferential accumulation in neural tissue regions with high metabolic activity and oxidative stress vulnerability. The conjugated double-bond systems characteristic of carotenoids provide exceptional capacity for singlet oxygen quenching and free radical neutralization, while their lipophilic nature enables integration into neural membrane phospholipids where they stabilize membrane architecture and protect against lipid peroxidation (230233). This mechanism is particularly relevant to chemotherapy-induced neurotoxicity, where oxidative membrane damage represents a primary pathological process.

Berry anthocyanins represent another compelling class of neuroprotective compounds with emerging evidence for peripheral nerve protection (234). These deeply pigmented flavonoid compounds demonstrate sophisticated abilities to modulate neuroinflammatory cascades while simultaneously enhancing neurotrophic factor expression and supporting cellular repair mechanisms (234, 235). Preclinical studies have shown that anthocyanin-rich extracts from blueberries, blackberries, and other dark berries can reduce oxidative stress markers and inflammatory cytokine expression in neural tissue while promoting nerve growth factor synthesis (236, 237). The protective mechanisms of anthocyanins operate through multiple convergent pathways including potent nuclear factor-κB pathway inhibition, enhancement of brain-derived neurotrophic factor expression in neural tissue, and direct antioxidant effects that complement carotenoid protection through different molecular targets (236, 237).

5 Practical application of plant food neuroprotection

The convergence of mechanistic understanding and clinical evidence detailed in the preceding sections provides a compelling rationale for the clinical application of plant-based neuroprotective strategies. However, translating these research findings into effective, safe, and feasible dietary protocols for patients undergoing chemotherapy requires careful consideration. This section, therefore, aims to delineate evidence-based principles for the selection, preparation, and combination of neuroprotective plant foods, thereby bridging the gap between scientific knowledge and practical implementation in patient care.

5.1 Evidence-based food selection and prioritization

Given the varying levels of clinical validation across different plant foods, a tiered approach to food selection provides the most rational foundation for practical implementation.

Tier 1 foods include those with direct clinical evidence in neuropathy prevention, primarily turmeric-containing foods and green tea, which should form the foundation of any plant food neuroprotection strategy (163, 179, 238, 239) (Table 4). Daily consumption of turmeric-spiced foods prepared with fat and black pepper, along with 2–3 cups of green tea, provides clinically relevant doses of curcumin and EGCG with established safety profiles (239241). An illustrative example of nutrient synergy is the combination of curcumin with piperine, a bioactive alkaloid from black pepper (242). Piperine inhibits hepatic and intestinal glucuronidation, thereby slowing curcumin metabolism and increasing its systemic exposure (243). This interaction provides a mechanistic rationale for pairing turmeric with black pepper in culinary and clinical contexts, underscoring that the form and combination of bioactive compounds can be as critical as their individual properties for achieving therapeutic efficacy.

Table 4
www.frontiersin.org

Table 4. Evidence-based tiered food selection guidelines.

Tier 2 foods encompass those with strong preclinical evidence and supportive clinical data from related conditions, including nuts and seeds rich in vitamin E and omega-3 fatty acids, colorful berries high in anthocyanins, and dark leafy greens providing multiple complementary compounds (244249). These foods should be consumed regularly as part of a diverse dietary pattern that maximizes synergistic interactions while ensuring nutritional adequacy (246, 249251).

Tier 3 foods include those with primarily mechanistic evidence but limited clinical validation, such as specific vegetables high in individual antioxidants or specialized fermented products (249, 252254). While these foods may provide additional benefits, they should supplement rather than replace the evidence-based Tier 1 and 2 selections.

5.2 Addressing treatment-related dietary challenges

Chemotherapy treatment creates unique dietary challenges that require adaptive strategies to maintain neuroprotective food intake while accommodating treatment-related side effects and safety considerations (255). Nausea and altered taste perception, common with many chemotherapy regimens, can be addressed through strategic use of ginger-containing warm preparations and mild, room-temperature foods that minimize aversive sensory experiences (256, 257). Well-cooked vegetable soups combining leafy greens, root vegetables, and gentle spices provide concentrated nutrition in easily tolerated liquid forms that accommodate swallowing difficulties and reduced appetite.

Immunocompromised status during chemotherapy necessitates careful attention to food safety, requiring thorough cooking of all plant foods to eliminate potential pathogens (258, 259). Raw fruits and vegetables should be avoided, with emphasis on cooked preparations including steamed vegetables, baked fruits, and thoroughly heated plant-based soups and stews. Frozen vegetables and fruits can provide year-round access to neuroprotective compounds while offering convenience and safety advantages through processing that reduces microbial contamination risk.

Reduced appetite and early satiety require emphasis on nutrient-dense foods in smaller portions, with frequent consumption of small amounts of high-quality plant foods rather than attempting to maintain normal portion sizes (260262). Concentrated sources such as cooked nut butters, well-cooked dried fruits, and warm herbal teas can provide meaningful neuroprotective compounds without requiring large food volumes. The incorporation of neuroprotective spices into familiar comfort foods can maintain nutritional goals while supporting psychological well-being during treatment.

Oral mucositis and swallowing difficulties, common with certain chemotherapy regimens, require modifications toward softer textures and neutral temperatures (263). Pureed vegetable soups, well-cooked mashed vegetables with olive oil, and lukewarm herbal teas provide gentle nutrition delivery that minimizes oral discomfort while maintaining neuroprotective compound intake. Room-temperature foods often provide better tolerance than hot or cold preparations during periods of oral sensitivity.

5.3 Long-term sustainability and patient-centered implementation

Successful long-term implementation requires integration of neuroprotective foods into sustainable dietary patterns that account for individual preferences, cultural backgrounds, and economic constraints (264, 265). Rather than prescriptive dietary protocols, flexible frameworks that allow for personal adaptation while maintaining core neuroprotective principles demonstrate superior adherence and long-term sustainability.

Cultural adaptation involves identifying traditional foods and preparation methods that naturally provide neuroprotective compounds, such as curry dishes in South Asian cuisines, tea ceremonies in East Asian cultures, and Mediterranean dietary patterns that emphasize olive oil, nuts, and vegetables. This approach respects cultural preferences while achieving therapeutic objectives through familiar and acceptable foods.

Economic considerations require emphasis on affordable, accessible plant foods that provide optimal neuroprotective value per unit cost. Dried legumes, seasonal vegetables, frozen berries, and basic spices often provide superior nutritional value compared to expensive supplements or exotic foods, while also being more widely available and culturally acceptable across diverse populations.

Patient education should focus on practical skills including food selection, preparation techniques, and meal planning that empowers individuals to make informed choices rather than relying on complex dietary protocols. Simple guidelines such as “eat colorful vegetables daily,” “include nuts or seeds with meals,” and “drink green tea regularly” provide actionable direction without overwhelming complexity.

6 Research limitations and future priorities

A critical limitation for the clinical translation of plant-derived polyphenols is their inherently low oral bioavailability (266). Compounds such as curcumin undergo rapid metabolism and systemic clearance, which significantly reduces their effective plasma and tissue concentrations (267). This pharmacokinetic barrier explains why the consistently positive results observed in preclinical models often fail to translate into equivalent efficacy in human trials using simple extracts (268). Advanced formulation strategies—including phospholipid complexes, nanoparticle encapsulation, and co-administration with piperine from black pepper—are therefore not merely technical details but essential requirements for achieving therapeutic exposure (243, 268). These delivery approaches enhance intestinal absorption, inhibit rapid metabolism, and prolong systemic circulation, thereby providing a mechanistic basis for the observed improvements in clinical outcomes (267, 268). Consequently, the form of the intervention must be considered as critically important as the compound itself when evaluating translational potential.

Another limitation relates to safety nuances that require careful consideration. While plant-derived antioxidants are generally regarded as safe in dietary amounts, some polyphenols may exert pro-oxidant effects under specific conditions, such as high doses in the presence of free transition metal ions (269271). Moreover, these compounds have the potential to interact with chemotherapy drugs by modulating cytochrome P450 enzymes and drug transporters such as P-glycoprotein (272). Although such risks remain largely theoretical at nutritional doses, they highlight the importance of systematic pharmacokinetic and interaction studies to ensure patient safety in clinical applications.

Despite promising mechanistic foundations and preliminary clinical evidence, significant limitations constrain the clinical application of plant food neuroprotection strategies. The most critical limitation is the scarcity of large-scale, well-controlled clinical trials specifically designed for chemotherapy-induced peripheral neuropathy prevention (2, 163, 164, 273). Current evidence relies primarily on single studies, cross-condition extrapolation, and preclinical models that cannot provide the definitive evidence required for clinical practice guidelines. Methodological challenges include the difficulty of standardizing complex plant food interventions and the natural variability in bioactive compound content across different sources, seasons, and preparation methods. The lack of validated biomarkers for early neuroprotective effects and the reliance on subjective symptom reporting further complicate intervention assessment and optimization.

Future research priorities center on three critical areas. First, large-scale multicenter randomized controlled trials are urgently needed to evaluate plant food interventions in diverse patient populations receiving different chemotherapy regimens. These studies should utilize standardized outcome measures and incorporate biomarker assessments to provide objective evidence of neuroprotective efficacy. Second, mechanistic research should focus on individual variability in plant food metabolism and response, including genetic polymorphisms affecting compound absorption and effectiveness. This research can inform personalized nutrition approaches that optimize interventions based on individual characteristics and treatment protocols. Third, standardization efforts must address intervention reproducibility through development of quality control methods for plant food characterization, establishment of preparation protocols, and creation of practical implementation guidelines that translate research findings into clinical recommendations while maintaining safety standards. The integration of these research priorities with systematic safety evaluations and long-term outcome studies will provide the evidence base necessary for widespread clinical adoption of plant food neuroprotection strategies.

7 Conclusion

The Level I evidence for curcumin in pediatric patients is particularly compelling, providing definitive proof-of-concept for plant-based neuroprotection. This comprehensive review demonstrates that plant food-derived antioxidant nutrients represent a promising approach to neuroprotection against chemotherapy-induced neurotoxicity. The mechanistic foundations are well-established, with plant foods providing multi-target protection through antioxidant system activation, anti-inflammatory effects, neurotrophic factor support, and mitochondrial preservation. The clinical evidence, while requiring further validation, provides encouraging proof-of-concept, with curcumin demonstrating the strongest validation and other plant foods showing supportive evidence from related conditions.

The practical advantages of plant food approaches include established safety profiles, widespread accessibility, and cost-effectiveness compared to pharmaceutical alternatives. However, significant limitations constrain clinical implementation, particularly the scarcity of large-scale clinical trials specifically designed for chemotherapy-induced peripheral neuropathy prevention. Future research priorities include large-scale multicenter randomized controlled trials and development of standardized intervention protocols. Plant food-derived antioxidant nutrients are a safe, accessible, and evidence-based strategy that warrants continued investigation as a complement to conventional cancer therapy, with the potential to transform supportive oncology care.

Author contributions

WL: Writing – original draft, Writing – review & editing, Conceptualization, Visualization. DY: Writing – original draft, Writing – review & editing, Data curation. ZZ: Writing – original draft, Writing – review & editing, Formal analysis. MW: Writing – original draft, Investigation. SX: Writing – original draft, Visualization. RJ: Writing – review & editing, Investigation. JC: Writing – review & editing, Data curation. CW: Writing – review & editing, Formal analysis. JQ: Writing – review & editing, Funding acquisition, Supervision.

Funding

The author(s) declare that financial support was received for the research and/or publication of this article. This work was supported by the Jiangsu Provincial Administration of Traditional Chinese Medicine Program (No. MS2023014).

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The authors declare that Gen AI was used in the creation of this manuscript. Specifically, generative AI technology was employed to assist in drafting the abstract and refining certain sections of the text. All outputs were carefully reviewed, verified for accuracy and originality, and the authors take full responsibility for the content of this manuscript.

Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Bae, EH, Greenwald, MK, and Schwartz, AG. Chemotherapy-induced peripheral neuropathy: mechanisms and therapeutic avenues. Neurotherapeutics. (2021) 18:2384–96. doi: 10.1007/s13311-021-01142-2

Crossref Full Text | Google Scholar

2. Szklener, K, Szklener, S, Michalski, A, Żak, K, Kuryło, W, Rejdak, K, et al. Dietary supplements in chemotherapy-induced peripheral neuropathy: a new Hope? Nutrients. (2022) 14:625. doi: 10.3390/nu14030625

Crossref Full Text | Google Scholar

3. Zajączkowska, R, Kocot-Kępska, M, Leppert, W, Wrzosek, A, Mika, J, and Wordliczek, J. Mechanisms of chemotherapy-induced peripheral neuropathy. Int J Mol Sci. (2019):20. doi: 10.3390/ijms20061451

Crossref Full Text | Google Scholar

4. Klafke, N, Bossert, J, Kröger, B, Neuberger, P, Heyder, U, Layer, M, et al. Prevention and treatment of chemotherapy-induced peripheral neuropathy (Cipn) with non-pharmacological interventions: clinical recommendations from a systematic scoping review and an expert consensus process. Med Sci. (2023) 11. doi: 10.3390/medsci11010015

Crossref Full Text | Google Scholar

5. Preti, K, and Davis, ME. Chemotherapy-induced peripheral neuropathy: assessment and treatment strategies for advanced practice providers. Clin J Oncol Nurs. (2024) 28:351–7. doi: 10.1188/24.Cjon.351-357

Crossref Full Text | Google Scholar

6. Flatters, SJL, Dougherty, PM, and Colvin, LA. Clinical and preclinical perspectives on chemotherapy-induced peripheral neuropathy (Cipn): a narrative review. Br J Anaesth. (2017) 119:737–49. doi: 10.1093/bja/aex229

Crossref Full Text | Google Scholar

7. Starobova, H, and Vetter, I. Pathophysiology of chemotherapy-induced peripheral neuropathy. Front Mol Neurosci. (2017) 10:174. doi: 10.3389/fnmol.2017.00174

Crossref Full Text | Google Scholar

8. Boyette-Davis, JA, Cata, JP, Driver, LC, Novy, DM, Bruel, BM, Mooring, DL, et al. Persistent Chemoneuropathy in patients receiving the plant alkaloids paclitaxel and vincristine. Cancer Chemother Pharmacol. (2013) 71:619–26. doi: 10.1007/s00280-012-2047-z

Crossref Full Text | Google Scholar

9. Flatters, SJ, and Bennett, GJ. Studies of peripheral sensory nerves in paclitaxel-induced painful peripheral neuropathy: evidence for mitochondrial dysfunction. Pain. (2006) 122:245–57. doi: 10.1016/j.pain.2006.01.037

Crossref Full Text | Google Scholar

10. Zhang, H, Yoon, SY, Zhang, H, and Dougherty, PM. Evidence that spinal astrocytes but not microglia contribute to the pathogenesis of paclitaxel-induced painful neuropathy. J Pain. (2012) 13:293–303. doi: 10.1016/j.jpain.2011.12.002

Crossref Full Text | Google Scholar

11. Mohd Sairazi, NS, and Sirajudeen, KNS. Natural products and their bioactive compounds: Neuroprotective potentials against neurodegenerative diseases. Evid Based Complement Alternat Med. (2020) 2020:6565396. doi: 10.1155/2020/6565396

Crossref Full Text | Google Scholar

12. Trushina, E, and McMurray, CT. Oxidative stress and mitochondrial dysfunction in neurodegenerative diseases. Neuroscience. (2007) 145:1233–48. doi: 10.1016/j.neuroscience.2006.10.056

Crossref Full Text | Google Scholar

13. Medawar, E, Huhn, S, Villringer, A, and Veronica, WA. The effects of plant-based diets on the body and the brain: a systematic review. Transl Psychiatry. (2019) 9:226. doi: 10.1038/s41398-019-0552-0

Crossref Full Text | Google Scholar

14. Ordovas, JM, Ferguson, LR, Tai, ES, and Mathers, JC. Personalised nutrition and health. BMJ (2018) 361:bmj.k2173. doi:doi: 10.1136/bmj.k2173

Crossref Full Text | Google Scholar

15. Ferguson, LR. Nutrigenomics approaches to functional foods. J Am Diet Assoc. (2009) 109:452–8. doi: 10.1016/j.jada.2008.11.024

Crossref Full Text | Google Scholar

16. Corella, D, and Ordovas, JM. Nutrigenomics in cardiovascular medicine. Circ Cardiovasc Genet. (2009) 2:637–51. doi: 10.1161/circgenetics.109.891366

Crossref Full Text | Google Scholar

17. Cavaletti, G, and Marmiroli, P. Chemotherapy-induced peripheral neurotoxicity. Nat Rev Neurol. (2010) 6:657–66. doi: 10.1038/nrneurol.2010.160

Crossref Full Text | Google Scholar

18. Shoaib, S, Ansari, MA, Fatease, AA, Safhi, AY, Hani, U, Jahan, R, et al. Plant-derived bioactive compounds in the Management of Neurodegenerative Disorders: challenges, future directions and molecular mechanisms involved in Neuroprotection. Pharmaceutics. (2023) 15. doi: 10.3390/pharmaceutics15030749

Crossref Full Text | Google Scholar

19. Pachman, DR, Barton, DL, Watson, JC, and Loprinzi, CL. Chemotherapy-induced peripheral neuropathy: prevention and treatment. Clin Pharmacol Ther. (2011) 90:377–87. doi: 10.1038/clpt.2011.115

Crossref Full Text | Google Scholar

20. Jessen, K, and Mirsky, R. The repair Schwann cell and its function in regenerating nerves. J Physiol. (2016) 594:3521–31. doi: 10.1113/JP270874

Crossref Full Text | Google Scholar

21. Millecamps, S, and Julien, JP. Axonal transport deficits and neurodegenerative diseases. Nat Rev Neurosci. (2013) 14:161–76. doi: 10.1038/nrn3380

Crossref Full Text | Google Scholar

22. Lin, MT, and Beal, MF. Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature. (2006) 443:787–95. doi: 10.1038/nature05292

Crossref Full Text | Google Scholar

23. Abbott, NJ, Rönnbäck, L, and Hansson, E. Astrocyte–endothelial interactions at the blood–brain barrier. Nat Rev Neurosci. (2006) 7:41–53. doi: 10.1038/nrn1824

Crossref Full Text | Google Scholar

24. Podratz, JL, Knight, AM, Ta, LE, Staff, NP, Gass, JM, Genelin, K, et al. Cisplatin induced mitochondrial DNA damage in dorsal root ganglion neurons. Neurobiol Dis. (2011) 41:661–8. doi: 10.1016/j.nbd.2010.11.017

Crossref Full Text | Google Scholar

25. Zheng, H, Xiao, W, and Bennett, G. Mitotoxicity and Bortezomib-induced chronic painful peripheral neuropathy. Exp Neurol. (2012) 238:225–34. doi: 10.1016/j.expneurol.2012.08.023

Crossref Full Text | Google Scholar

26. Xiao, W, Zheng, H, and Bennett, G. Characterization of Oxaliplatin-induced chronic painful peripheral neuropathy in the rat and comparison with the neuropathy induced by paclitaxel. Neuroscience. (2012) 203:194–206. doi: 10.1016/j.neuroscience.2011.12.023

Crossref Full Text | Google Scholar

27. Areti, A, Yerra, VG, Naidu, V, and Kumar, A. Oxidative stress and nerve damage: role in chemotherapy induced peripheral neuropathy. Redox Biol. (2014) 2:289–95. doi: 10.1016/j.redox.2014.01.006

Crossref Full Text | Google Scholar

28. Ji, RR, Xu, ZZ, and Gao, YJ. Emerging targets in Neuroinflammation-driven chronic pain. Nat Rev Drug Discov. (2014) 13:533–48. doi: 10.1038/nrd4334

Crossref Full Text | Google Scholar

29. Grace, PM, Hutchinson, MR, Maier, SF, and Watkins, LR. Pathological pain and the Neuroimmune Interface. Nat Rev Immunol. (2014) 14:217–31. doi: 10.1038/nri3621

Crossref Full Text | Google Scholar

30. Wang, H, Bloom, O, Zhang, M, Vishnubhakat, JM, Ombrellino, M, Che, J, et al. Hmg-1 as a late mediator of endotoxin lethality in mice. Science. (1999) 285:248–51. doi: 10.1126/science.285.5425.248

Crossref Full Text | Google Scholar

31. Gutiérrez-Ibarluzea, I, and Arana-Arri, E. Nutrition, a health technology that deserves increasing interest among Hta doers. A systematic review. Front Pharmacol (2015) Volume 6–2015. doi:doi: 10.3389/fphar.2015.00156

Crossref Full Text | Google Scholar

32. Pacifico, P, Coy-Dibley, JS, Miller, RJ, and Menichella, DM. Peripheral mechanisms of peripheral neuropathic pain. Front Mol Neurosci. (2023) 16:1252442. doi: 10.3389/fnmol.2023.1252442

Crossref Full Text | Google Scholar

33. Havelin, J, Imbert, I, Cormier, J, Allen, J, Porreca, F, and King, T. Central sensitization and neuropathic features of Ongoing pain in a rat model of advanced osteoarthritis. J Pain. (2016) 17:374–82. doi: 10.1016/j.jpain.2015.12.001

Crossref Full Text | Google Scholar

34. Lawrence, T. The nuclear factor Nf-Κb pathway in inflammation. Cold Spring Harb Perspect Biol. (2009) 1:a001651. doi: 10.1101/cshperspect.a001651

Crossref Full Text | Google Scholar

35. Sekiguchi, F, and Kawabata, A. Role of Hmgb1 in chemotherapy-induced peripheral neuropathy. Int J Mol Sci. (2020) 22. doi: 10.3390/ijms22010367

Crossref Full Text | Google Scholar

36. Quasthoff, S, and Hartung, HP. Chemotherapy-induced peripheral neuropathy. J Neurol. (2002) 249:9–17. doi: 10.1007/pl00007853

Crossref Full Text | Google Scholar

37. Staff NPGrisold, A, Grisold, W, and Windebank, AJ. Chemotherapy-induced peripheral neuropathy: a current review. Ann Neurol. (2017) 81:772–81. doi: 10.1002/ana.24951

Crossref Full Text | Google Scholar

38. García-Domínguez, M. Ngf in neuropathic pain: understanding its role and therapeutic opportunities. Curr Issues Mol Biol. (2025) 47. doi: 10.3390/cimb47020093

Crossref Full Text | Google Scholar

39. Doyle, TM, and Salvemini, D. Mini-review: mitochondrial dysfunction and chemotherapy-induced neuropathic pain. Neurosci Lett. (2021) 760:136087. doi: 10.1016/j.neulet.2021.136087

Crossref Full Text | Google Scholar

40. Geremia, NM, Pettersson, LM, Hasmatali, JC, Hryciw, T, Danielsen, N, Schreyer, DJ, et al. Endogenous Bdnf regulates induction of intrinsic neuronal growth programs in injured sensory neurons. Exp Neurol. (2010) 223:128–42. doi: 10.1016/j.expneurol.2009.07.022

Crossref Full Text | Google Scholar

41. Samaddar, S, Redhwan, MAM, Eraiah, MM, and Koneri, R. Neurotrophins in peripheral neuropathy: exploring pathophysiological mechanisms and emerging therapeutic opportunities. CNS Neurol Disord Drug Targets. (2025) 24:91–101. doi: 10.2174/0118715273327121240820074049

Crossref Full Text | Google Scholar

42. Aromolaran, KA, and Goldstein, PA. Ion channels and neuronal Hyperexcitability in chemotherapy-induced peripheral neuropathy; cause and effect? Mol Pain. (2017) 13:1744806917714693. doi: 10.1177/1744806917714693

Crossref Full Text | Google Scholar

43. Apfel, SC. Neurotrophic factors and diabetic peripheral neuropathy. Eur Neurol. (1999) 41:27–34. doi: 10.1159/000052077

Crossref Full Text | Google Scholar

44. Kotliarova, A, and Sidorova, YA. Glial cell line-derived Neurotrophic factor family ligands, players at the Interface of Neuroinflammation and Neuroprotection: focus onto the glia. Front Cell Neurosci. (2021) 15:679034. doi: 10.3389/fncel.2021.679034

Crossref Full Text | Google Scholar

45. Sikandar, S, Minett, MS, Millet, Q, Santana-Varela, S, Lau, J, Wood, JN, et al. Brain-derived Neurotrophic factor derived from sensory neurons plays a critical role in chronic pain. Brain. (2018) 141:1028–39. doi: 10.1093/brain/awy009

Crossref Full Text | Google Scholar

46. Numakawa, T, and Kajihara, R. The role of brain-derived Neurotrophic factor as an essential mediator in neuronal functions and the therapeutic potential of its Mimetics for Neuroprotection in neurologic and psychiatric disorders. Molecules. (2025) 30. doi: 10.3390/molecules30040848

Crossref Full Text | Google Scholar

47. Mitroshina, EV, Mishchenko, TA, Shishkina, TV, and Vedunova, MV. Role of Neurotrophic factors Bdnf and Gdnf in nervous system adaptation to the influence of ischemic factors. Bull Exp Biol Med. (2019) 167:574–9. doi: 10.1007/s10517-019-04574-1

Crossref Full Text | Google Scholar

48. Youk, J, Kim, YS, Lim, JA, Shin, DY, Koh, Y, Lee, ST, et al. Depletion of nerve growth factor in chemotherapy-induced peripheral neuropathy associated with hematologic malignancies. PLoS One. (2017) 12:e0183491. doi: 10.1371/journal.pone.0183491

Crossref Full Text | Google Scholar

49. Siniscalco, D, Giordano, C, Rossi, F, Maione, S, and de Novellis, V. Role of Neurotrophins in neuropathic pain. Curr Neuropharmacol. (2011) 9:523–9. doi: 10.2174/157015911798376208

Crossref Full Text | Google Scholar

50. Gaur, A, Varatharajan, S, Balan, Y, Taranikanti, M, John, NA, Umesh, M, et al. Brain-derived Neurotrophic factor (Bdnf) and other Neurotrophic factors in type 2 diabetes mellitus and their association with neuropathy. Ir J Med Sci. (2024) 193:2287–92. doi: 10.1007/s11845-024-03716-3

Crossref Full Text | Google Scholar

51. Canta, A, Pozzi, E, and Carozzi, VA. Mitochondrial dysfunction in chemotherapy-induced peripheral neuropathy (Cipn). Toxics. (2015) 3:198–223. doi: 10.3390/toxics3020198

Crossref Full Text | Google Scholar

52. Rovini, A. Tubulin-Vdac interaction: molecular basis for mitochondrial dysfunction in chemotherapy-induced peripheral neuropathy. Front Physiol. (2019) 10:671. doi: 10.3389/fphys.2019.00671

Crossref Full Text | Google Scholar

53. Cabezas-Opazo, FA, Vergara-Pulgar, K, Pérez, MJ, Jara, C, Osorio-Fuentealba, C, and Quintanilla, RA. Mitochondrial dysfunction contributes to the pathogenesis of Alzheimer's disease. Oxidative Med Cell Longev. (2015) 2015:509654. doi: 10.1155/2015/509654

Crossref Full Text | Google Scholar

54. Stanga, S, Caretto, A, Boido, M, and Vercelli, A. Mitochondrial dysfunctions: a red thread across neurodegenerative diseases. Int J Mol Sci. (2020) 21. doi: 10.3390/ijms21103719

Crossref Full Text | Google Scholar

55. Griffiths, LA, and Flatters, SJ. Pharmacological modulation of the mitochondrial electron transport chain in paclitaxel-induced painful peripheral neuropathy. J Pain. (2015) 16:981–94. doi: 10.1016/j.jpain.2015.06.008

Crossref Full Text | Google Scholar

56. Lu, B. Mitochondrial dynamics and Neurodegeneration. Curr Neurol Neurosci Rep. (2009) 9:212–9. doi: 10.1007/s11910-009-0032-7

Crossref Full Text | Google Scholar

57. Babu, A, and Urulangodi, M. Chemotherapy-induced neuronal DNA damage: An intriguing toolbox to elucidate DNA repair mechanisms in the brain. Genome Instab Dis. (2023) 4:315–32. doi: 10.1007/s42764-023-00110-8

Crossref Full Text | Google Scholar

58. Gorini, S, De Angelis, A, Berrino, L, Malara, N, Rosano, G, and Ferraro, E. Chemotherapeutic drugs and mitochondrial dysfunction: focus on doxorubicin, Trastuzumab, and Sunitinib. Oxidative Med Cell Longev. (2018) 2018:7582730. doi: 10.1155/2018/7582730

Crossref Full Text | Google Scholar

59. Baek, ML, Lee, J, Pendleton, KE, Berner, MJ, Goff, EB, Tan, L, et al. Mitochondrial structure and function adaptation in residual triple negative breast cancer cells surviving chemotherapy treatment. Oncogene. (2023) 42:1117–31. doi: 10.1038/s41388-023-02596-8

Crossref Full Text | Google Scholar

60. Marullo, R, Werner, E, Degtyareva, N, Moore, B, Altavilla, G, Ramalingam, SS, et al. Cisplatin induces a mitochondrial-Ros response that contributes to cytotoxicity depending on mitochondrial redox status and bioenergetic functions. PLoS One. (2013) 8:e81162. doi: 10.1371/journal.pone.0081162

Crossref Full Text | Google Scholar

61. Guerra, F, Arbini, AA, and Moro, L. Mitochondria and cancer Chemoresistance. Biochim Biophys Acta Bioenerg. (2017) 1858:686–99. doi: 10.1016/j.bbabio.2017.01.012

Crossref Full Text | Google Scholar

62. Cheng, X-T, Huang, N, and Sheng, Z-H. Programming axonal mitochondrial maintenance and bioenergetics in Neurodegeneration and regeneration. Neuron. (2022) 110:1899–923. doi: 10.1016/j.neuron.2022.03.015

Crossref Full Text | Google Scholar

63. Wang, B, Huang, M, Shang, D, Yan, X, Zhao, B, and Zhang, X. Mitochondrial behavior in axon degeneration and regeneration. Front Aging Neurosci. (2021) 13:650038. doi: 10.3389/fnagi.2021.650038

Crossref Full Text | Google Scholar

64. Lim, TK, Rone, MB, Lee, S, Antel, JP, and Zhang, J. Mitochondrial and bioenergetic dysfunction in trauma-induced painful peripheral neuropathy. Mol Pain (2015) 11:s12990-015-0057-7. doi:doi: 10.1186/s12990-015-0057-7

Crossref Full Text | Google Scholar

65. Sharma, Y, Gupta, JK, Babu, MA, Singh, S, and Sindhu, RK. Signaling pathways concerning mitochondrial dysfunction: implications in Neurodegeneration and possible molecular targets. J Mol Neurosci. (2024) 74:101. doi: 10.1007/s12031-024-02269-5

Crossref Full Text | Google Scholar

66. Bathina, S, and Das, UN. Role of mitochondrial dysfunction in cellular lipid homeostasis and disease. Discov Med. (2023) 35:653–63. doi: 10.24976/Discov.Med.202335178.64

Crossref Full Text | Google Scholar

67. Suomalainen, A, and Nunnari, J. Mitochondria at the crossroads of health and disease. Cell. (2024) 187:2601–27. doi: 10.1016/j.cell.2024.04.037

Crossref Full Text | Google Scholar

68. Moro, L. Mitochondria at the crossroads of physiology and pathology. J Clin Med. (2020) 9. doi: 10.3390/jcm9061971

Crossref Full Text | Google Scholar

69. Tkachev, VO, Menshchikova, EB, and Zenkov, NK. Mechanism of the Nrf2/Keap1/are Signaling system. Biochemistry (Mosc). (2011) 76:407–22. doi: 10.1134/s0006297911040031

Crossref Full Text | Google Scholar

70. Sykiotis, GP. Keap1/Nrf2 Signaling pathway. Antioxidants. (2021) 10. doi: 10.3390/antiox10060828

Crossref Full Text | Google Scholar

71. Dinkova-Kostova, AT, Kostov, RV, and Canning, P. Keap1, the cysteine-based mammalian intracellular sensor for electrophiles and oxidants. Arch Biochem Biophys. (2017) 617:84–93. doi: 10.1016/j.abb.2016.08.005

Crossref Full Text | Google Scholar

72. Stewart, JD, Hengstler, JG, and Bolt, HM. Control of oxidative stress by the Keap1-Nrf2 pathway. Arch Toxicol. (2011) 85:239. doi: 10.1007/s00204-011-0694-1

Crossref Full Text | Google Scholar

73. Taguchi, K, Motohashi, H, and Yamamoto, M. Molecular mechanisms of the Keap1–Nrf2 pathway in stress response and cancer evolution. Genes Cells. (2011) 16:123–40. doi: 10.1111/j.1365-2443.2010.01473.x

Crossref Full Text | Google Scholar

74. Eggler, AL, and Savinov, SN. Chemical and biological mechanisms of phytochemical activation of Nrf2 and importance in disease prevention. Recent Adv Phytochem. (2013) 43:121–55. doi: 10.1007/978-3-319-00581-2_7

Crossref Full Text | Google Scholar

75. Li, M, Huang, W, Jie, F, Wang, M, Zhong, Y, Chen, Q, et al. Discovery of Keap1-Nrf2 small-molecule inhibitors from phytochemicals based on molecular docking. Food Chem Toxicol. (2019) 133:110758. doi: 10.1016/j.fct.2019.110758

Crossref Full Text | Google Scholar

76. Alzain, AA, Mukhtar, RM, Abdelmoniem, N, Shoaib, TH, Osman, W, Alsulaimany, M, et al. Modulation of Nrf2/Keap1-mediated oxidative stress for cancer treatment by natural products using Pharmacophore-based screening, molecular docking, and molecular dynamics studies. Molecules. (2023) 28. doi: 10.3390/molecules28166003

Crossref Full Text | Google Scholar

77. Hayes, JD, McMahon, M, Chowdhry, S, and Dinkova-Kostova, AT. Cancer chemoprevention mechanisms mediated through the Keap1-Nrf2 pathway. Antioxid Redox Signal. (2010) 13:1713–48. doi: 10.1089/ars.2010.3221

Crossref Full Text | Google Scholar

78. Sun, Z, Wu, T, Zhao, F, Lau, A, Birch, CM, and Zhang, DD. Kpna6 (Importin {alpha}7)-mediated nuclear import of Keap1 represses the Nrf2-dependent antioxidant response. Mol Cell Biol. (2011) 31:1800–11. doi: 10.1128/mcb.05036-11

Crossref Full Text | Google Scholar

79. Biswas, C, Shah, N, Muthu, M, La, P, Fernando, AP, Sengupta, S, et al. Nuclear Heme Oxygenase-1 (Ho-1) modulates subcellular distribution and activation of Nrf2, impacting metabolic and anti-oxidant Defenses. J Biol Chem. (2014) 289:26882–94. doi: 10.1074/jbc.M114.567685

Crossref Full Text | Google Scholar

80. Collinson, EJ, Wimmer-Kleikamp, S, Gerega, SK, Yang, YH, Parish, CR, Dawes, IW, et al. The yeast homolog of Heme Oxygenase-1 affords cellular antioxidant protection via the transcriptional regulation of known antioxidant genes. J Biol Chem. (2011) 286:2205–14. doi: 10.1074/jbc.M110.187062

Crossref Full Text | Google Scholar

81. Piantadosi, CA, Withers, CM, Bartz, RR, MacGarvey, NC, Fu, P, Sweeney, TE, et al. Heme Oxygenase-1 couples activation of mitochondrial biogenesis to anti-inflammatory cytokine expression. J Biol Chem. (2011) 286:16374–85. doi: 10.1074/jbc.M110.207738

Crossref Full Text | Google Scholar

82. Ryter, SW. Heme Oxygenase-1/carbon monoxide as modulators of autophagy and inflammation. Arch Biochem Biophys. (2019) 678:108186. doi: 10.1016/j.abb.2019.108186

Crossref Full Text | Google Scholar

83. Rashid, MH, Babu, D, and Siraki, AG. Interactions of the antioxidant enzymes Nad(P)H: Quinone Oxidoreductase 1 (Nqo1) and Nrh: Quinone Oxidoreductase 2 (Nqo2) with pharmacological agents, endogenous biochemicals and environmental contaminants. Chem Biol Interact. (2021) 345:109574. doi: 10.1016/j.cbi.2021.109574

Crossref Full Text | Google Scholar

84. den Braver-Sewradj, SP, den Braver, MW, Toorneman, RM, van Leeuwen, S, Zhang, Y, Dekker, SJ, et al. Reduction and scavenging of chemically reactive drug metabolites by Nad(P)H:Quinone Oxidoreductase 1 and Nrh:Quinone Oxidoreductase 2 and variability in hepatic concentrations. Chem Res Toxicol. (2018) 31:116–26. doi: 10.1021/acs.chemrestox.7b00289

Crossref Full Text | Google Scholar

85. Chandrasena, RE, Edirisinghe, PD, Bolton, JL, and Thatcher, GR. Problematic detoxification of Estrogen Quinones by Nad(P)H-dependent Quinone Oxidoreductase and glutathione-S-Transferase. Chem Res Toxicol. (2008) 21:1324–9. doi: 10.1021/tx8000797

Crossref Full Text | Google Scholar

86. Wu, X, Li, X, Li, Z, Yu, Y, You, Q, and Zhang, X. Discovery of Nonquinone substrates for Nad(P)H: Quinone Oxidoreductase 1 (Nqo1) as effective intracellular Ros generators for the treatment of drug-resistant non-small-cell lung cancer. J Med Chem. (2018) 61:11280–97. doi: 10.1021/acs.jmedchem.8b01424

Crossref Full Text | Google Scholar

87. Shin, JW, Chun, KS, Kim, DH, Kim, SJ, Kim, SH, Cho, NC, et al. Curcumin induces stabilization of Nrf2 protein through Keap1 cysteine modification. Biochem Pharmacol. (2020) 173:113820. doi: 10.1016/j.bcp.2020.113820

Crossref Full Text | Google Scholar

88. Deck, LM, Hunsaker, LA, Vander Jagt, TA, Whalen, LJ, Royer, RE, and Vander Jagt, DL. Activation of anti-oxidant Nrf2 Signaling by Enone analogues of Curcumin. Eur J Med Chem. (2018) 143:854–65. doi: 10.1016/j.ejmech.2017.11.048

Crossref Full Text | Google Scholar

89. Pany, S, You, Y, and Das, J. Curcumin inhibits protein kinase cα activity by binding to its C1 domain. Biochemistry. (2016) 55:6327–36. doi: 10.1021/acs.biochem.6b00932

Crossref Full Text | Google Scholar

90. Han, X, Xu, B, Beevers, CS, Odaka, Y, Chen, L, Liu, L, et al. Curcumin inhibits protein phosphatases 2a and 5, leading to activation of mitogen-activated protein kinases and death in tumor cells. Carcinogenesis. (2012) 33:868–75. doi: 10.1093/carcin/bgs029

Crossref Full Text | Google Scholar

91. Ayli, EE, Dugas-Breit, S, Li, W, Marshall, C, Zhao, L, Meulener, M, et al. Curcuminoids activate P38 map kinases and promote Uvb-dependent signalling in keratinocytes. Exp Dermatol. (2010) 19:493–500. doi: 10.1111/j.1600-0625.2010.01081.x

Crossref Full Text | Google Scholar

92. Aggarwal, BB, and Harikumar, KB. Potential therapeutic effects of Curcumin, the anti-inflammatory agent, against neurodegenerative, cardiovascular, pulmonary, metabolic, autoimmune and neoplastic diseases. Int J Biochem Cell Biol. (2009) 41:40–59. doi: 10.1016/j.biocel.2008.06.010

Crossref Full Text | Google Scholar

93. Hayden, MS, and Ghosh, S. Shared principles in Nf-Kappab Signaling. Cell. (2008) 132:344–62. doi: 10.1016/j.cell.2008.01.020

Crossref Full Text | Google Scholar

94. Singh, S, and Aggarwal, BB. Activation of transcription factor Nf-Κb is suppressed by Curcumin (Diferuloylmethane)(∗). J Biol Chem. (1995) 270:24995–5000. doi: 10.1074/jbc.270.42.24995

Crossref Full Text | Google Scholar

95. Jobin, C, Bradham, CA, Russo, MP, Juma, B, Narula, AS, Brenner, DA, et al. Curcumin blocks cytokine-mediated Nf-Κb activation and Proinflammatory gene expression by inhibiting inhibitory factor I-Κb kinase activity. J Immunol. (1999) 163:3474–83. doi: 10.4049/jimmunol.163.6.3474

Crossref Full Text | Google Scholar

96. Zhang, MW, Sun, X, Xu, YW, Meng, W, Tang, Q, Gao, H, et al. Curcumin relieves Oxaliplatin-induced neuropathic pain via reducing inflammation and activating antioxidant response. Cell Biol Int. (2024) 48:872–82. doi: 10.1002/cbin.12153

Crossref Full Text | Google Scholar

97. Kunnumakkara, AB, Bordoloi, D, Harsha, C, Banik, K, Gupta, SC, and Aggarwal, BB. Curcumin mediates anticancer effects by modulating multiple cell Signaling pathways. Clin Sci. (2017) 131:1781–99. doi: 10.1042/cs20160935

Crossref Full Text | Google Scholar

98. Zhang, X, Guan, Z, Wang, X, Sun, D, Wang, D, Li, Y, et al. Curcumin alleviates Oxaliplatin-induced peripheral neuropathic pain through inhibiting oxidative stress-mediated activation of Nf-Κb and mitigating inflammation. Biol Pharm Bull. (2020) 43:348–55. doi: 10.1248/bpb.b19-00862

Crossref Full Text | Google Scholar

99. Ashrafizadeh, M, Ahmadi, Z, Mohammadinejad, R, Farkhondeh, T, and Samarghandian, S. Curcumin activates the Nrf2 pathway and induces cellular protection against oxidative injury. Curr Mol Med. (2020) 20:116–33. doi: 10.2174/1566524019666191016150757

Crossref Full Text | Google Scholar

100. Iqbal, B, Ghildiyal, A, Singh, S, Siddiqui, S, Kumari, P, Arshad, M, et al. Combinatorial effect of Curcumin and tumor necrosis factor-Α-related apoptosis-inducing ligand (trail) in induction of apoptosis via inhibition of nuclear factor kappa activity and enhancement of Caspase-3 activity in chronic myeloid cells: An in-vitro study. J Cancer Res Ther. (2018) 14:S1193–s200. doi: 10.4103/jcrt.JCRT_348_18

Crossref Full Text | Google Scholar

101. Dong, HJ, Shang, CZ, Peng, DW, Xu, J, Xu, PX, Zhan, L, et al. Curcumin attenuates ischemia-like injury induced Il-1β elevation in brain microvascular endothelial cells via inhibiting Mapk pathways and nuclear factor-Κb activation. Neurol Sci. (2014) 35:1387–92. doi: 10.1007/s10072-014-1718-4

Crossref Full Text | Google Scholar

102. Baur, JA, and Sinclair, DA. Therapeutic potential of resveratrol: the in vivo evidence. Nat Rev Drug Discov. (2006) 5:493–506. doi: 10.1038/nrd2060

Crossref Full Text | Google Scholar

103. Lei, M, Wang, JG, Xiao, DM, Fan, M, Wang, DP, Xiong, JY, et al. Resveratrol inhibits interleukin 1β-mediated inducible nitric oxide synthase expression in articular chondrocytes by activating Sirt1 and thereby suppressing nuclear factor-Κb activity. Eur J Pharmacol. (2012) 674:73–9. doi: 10.1016/j.ejphar.2011.10.015

Crossref Full Text | Google Scholar

104. Zhu, X, Liu, Q, Wang, M, Liang, M, Yang, X, Xu, X, et al. Activation of Sirt1 by resveratrol inhibits Tnf-Α induced inflammation in fibroblasts. PLoS One. (2011) 6:e27081. doi: 10.1371/journal.pone.0027081

Crossref Full Text | Google Scholar

105. Owjfard, M, Rahimian, Z, Karimi, F, Borhani-Haghighi, A, and Mallahzadeh, A. A comprehensive review on the Neuroprotective potential of resveratrol in ischemic stroke. Heliyon. (2024) 10:e34121. doi: 10.1016/j.heliyon.2024.e34121

Crossref Full Text | Google Scholar

106. Morselli, E, Maiuri, MC, Markaki, M, Megalou, E, Pasparaki, A, Palikaras, K, et al. Caloric restriction and resveratrol promote longevity through the Sirtuin-1-dependent induction of autophagy. Cell Death Dis. (2010) 1:e10. doi: 10.1038/cddis.2009.8

Crossref Full Text | Google Scholar

107. Cao, W, Dou, Y, and Li, A. Resveratrol boosts cognitive function by targeting Sirt1. Neurochem Res. (2018) 43:1705–13. doi: 10.1007/s11064-018-2586-8

Crossref Full Text | Google Scholar

108. Denissova, NG, Nasello, CM, Yeung, PL, Tischfield, JA, and Brenneman, MA. Resveratrol protects mouse embryonic stem cells from ionizing radiation by accelerating recovery from DNA Strand breakage. Carcinogenesis. (2012) 33:149–55. doi: 10.1093/carcin/bgr236

Crossref Full Text | Google Scholar

109. Wei, G, Wang, J, Wu, Y, Zheng, X, Zeng, Y, Li, Y, et al. Sirtuin 1 alleviates Neuroinflammation-induced apoptosis after traumatic brain injury. J Cell Mol Med. (2021) 25:4478–86. doi: 10.1111/jcmm.16534

Crossref Full Text | Google Scholar

110. Fechtner, S, Singh, A, Chourasia, M, and Ahmed, S. Molecular insights into the differences in anti-inflammatory activities of green tea Catechins on Il-1β Signaling in rheumatoid arthritis synovial fibroblasts. Toxicol Appl Pharmacol. (2017) 329:112–20. doi: 10.1016/j.taap.2017.05.016

Crossref Full Text | Google Scholar

111. Yoshida, S, Inaba, H, Nomura, R, Nakano, K, and Matsumoto-Nakano, M. Green tea Catechins inhibit Porphyromonas Gulae Lps-induced inflammatory responses in human gingival epithelial cells. J Oral Biosciences. (2022) 64:352–8. doi: 10.1016/j.job.2022.05.006

Crossref Full Text | Google Scholar

112. Ohishi, T, Goto, S, Monira, P, Isemura, M, and Nakamura, Y. Anti-inflammatory action of green tea. Anti-inflammatory Anti-allergy Agents Medicinal Chemistry. (2016) 15:74–90. doi: 10.2174/1871523015666160915154443

Crossref Full Text | Google Scholar

113. Babu, P, and Liu, D. Green tea Catechins and cardiovascular health: An update. Curr Med Chem. (2008) 15:1840–50. doi: 10.2174/092986708785132979

Crossref Full Text | Google Scholar

114. Moosavi, F, Hosseini, R, Saso, L, and Firuzi, O. Modulation of Neurotrophic Signaling pathways by polyphenols. Drug Des Devel Ther. (2015) 10:23–42. doi: 10.2147/DDDT.S96936

Crossref Full Text | Google Scholar

115. Cao, C, Xiao, J, Liu, M, Ge, Z, Huang, R, Qi, M, et al. Active components, derived from Kai-Xin-san, a herbal formula, increase the expressions of Neurotrophic factor Ngf and Bdnf on mouse astrocyte primary cultures via camp-dependent Signaling pathway. J Ethnopharmacol. (2018) 224:554–62. doi: 10.1016/j.jep.2018.06.007

Crossref Full Text | Google Scholar

116. Xu, S, Bi, C, Choi, R, Zhu, K, Miernisha, A, Dong, T, et al. Flavonoids induce the synthesis and secretion of Neurotrophic factors in cultured rat astrocytes: a Signaling response mediated by Estrogen receptor. Evid Based Complement Alternat Med. (2013) 2013:127075. doi: 10.1155/2013/127075

Crossref Full Text | Google Scholar

117. Numakawa, T, and Odaka, H. Brain-derived Neurotrophic factor Signaling in the pathophysiology of Alzheimer’s disease: beneficial effects of flavonoids for Neuroprotection. Int J Mol Sci. (2021) 22:5719. doi: 10.3390/ijms22115719

Crossref Full Text | Google Scholar

118. He, Y, Chen, S, Tsoi, B, Qi, S, Gu, B, Wang, Z, et al. And its active compound P-Coumaric acid promote brain-derived Neurotrophic factor Signaling for inducing hippocampal neurogenesis and improving post-cerebral ischemic spatial cognitive functions. Front Cell Develop Biol. (2021) 8:577790. doi: 10.3389/fcell.2020.577790

Crossref Full Text | Google Scholar

119. Naoi, M, Inaba-Hasegawa, K, Shamoto-Nagai, M, and Maruyama, W. Neurotrophic function of phytochemicals for Neuroprotection in aging and neurodegenerative disorders: modulation of intracellular Signaling and gene expression. J Neural Transm. (2017) 124:1515–27. doi: 10.1007/s00702-017-1797-5

Crossref Full Text | Google Scholar

120. Colucci-D'Amato, L, Volpicelli, F, and Speranza, L. Neurotrophic factor Bdnf, physiological functions and therapeutic potential in depression, Neurodegeneration and brain cancer. Int J Mol Sci. (2020) 21:7777. doi: 10.3390/ijms21207777

Crossref Full Text | Google Scholar

121. Moya-Alvarado, G, Guerra, M, Wu, C, Mobley, W, Perlson, E, and Bronfman, F. Bdnf/Trkb Signaling endosomes in axons coordinate Creb/Mtor activation and protein synthesis in the cell body to induce dendritic growth in cortical neurons. eLife. (2023) 12:e77455. doi: 10.7554/eLife.77455

Crossref Full Text | Google Scholar

122. Azman, KF, and Zakaria, R. Recent advances on the role of brain-derived Neurotrophic factor (Bdnf) in neurodegenerative diseases. Int J Mol Sci. (2022) 23:6827. doi: 10.3390/ijms23126827

Crossref Full Text | Google Scholar

123. Costa, R, Martins, L, Tahiri, E, and Duarte, C. Brain-derived Neurotrophic factor-induced regulation of Rna metabolism in neuronal development and synaptic plasticity. Wiley Interdiscip Rev. (2022) 13:e1713. doi: 10.1002/wrna.1713

Crossref Full Text | Google Scholar

124. Setchell, KD, and Cassidy, A. Dietary Isoflavones: biological effects and relevance to human health. J Nutr. (1999) 129:758s–67s. doi: 10.1093/jn/129.3.758S

Crossref Full Text | Google Scholar

125. Viscardi, G, Back, S, Ahmed, A, Yang, S, Mejia, SB, Zurbau, A, et al. Effect of soy Isoflavones on measures of Estrogenicity: a systematic review and meta-analysis of randomized controlled trials. Adv Nutr. (2025) 16:100327. doi: 10.1016/j.advnut.2024.100327

Crossref Full Text | Google Scholar

126. Simons, R, Gruppen, H, Bovee, TF, Verbruggen, MA, and Vincken, JP. Prenylated Isoflavonoids from plants as selective Estrogen receptor modulators (Phytoserms). Food Funct. (2012) 3:810–27. doi: 10.1039/c2fo10290k

Crossref Full Text | Google Scholar

127. Adams, S, Aksenova, M, Aksenov, M, Mactutus, C, and Booze, R. Soy Isoflavones Genistein and Daidzein exert anti-apoptotic actions via a selective Er-mediated mechanism in neurons following Hiv-1 Tat1–86 exposure. PLoS One. (2012) 7:e37540. doi: 10.1371/journal.pone.0037540

Crossref Full Text | Google Scholar

128. Valeri, A, Fiorenzani, P, Rossi, R, Aloisi, A, Valoti, M, and Pessina, F. The soy phytoestrogens Genistein and Daidzein as Neuroprotective agents against anoxia-Glucopenia and reperfusion damage in rat urinary bladder. Pharmacol Res. (2012) 66:309–16. doi: 10.1016/j.phrs.2012.06.007

Crossref Full Text | Google Scholar

129. Kuiper, GGJM, Lemmen, JG, Carlsson, B, Corton, JC, Safe, SH, van der Saag, PT, et al. Interaction of estrogenic chemicals and phytoestrogens with Estrogen receptor Β. Endocrinology. (1998) 139:4252–63. doi: 10.1210/endo.139.10.6216

Crossref Full Text | Google Scholar

130. McCarty, M. Isoflavones made simple - Genistein's agonist activity for the Beta-type Estrogen receptor mediates their health benefits. Med Hypotheses. (2006) 66:1093–114. doi: 10.1016/J.MEHY.2004.11.046

Crossref Full Text | Google Scholar

131. Ajdžanović, V, Medigović, I, Živanović, J, Mojić, M, and Milošević, V. Membrane steroid receptor-mediated action of soy Isoflavones: tip of the iceberg. J Membr Biol. (2015) 248:1–6. doi: 10.1007/s00232-014-9745-x

Crossref Full Text | Google Scholar

132. Nakaya, M, Tachibana, H, and Yamada, K. Isoflavone Genistein and Daidzein up-regulate Lps-induced inducible nitric oxide synthase activity through Estrogen receptor pathway in Raw264.7 cells. Biochem Pharmacol. (2005) 71:108–14. doi: 10.1016/J.BCP.2005.10.002

Crossref Full Text | Google Scholar

133. Sohrabji, F, Miranda, R, and Toran-Allerand, C. Identification of a putative Estrogen response element in the gene encoding brain-derived Neurotrophic factor. Proc Natl Acad Sci USA. (1995) 92:11110–4. doi: 10.1073/PNAS.92.24.11110

Crossref Full Text | Google Scholar

134. Chini, A, Deb, P, Guha, P, Rishi, A, Bhan, A, Brady, B, et al. Abstract 2068 Bdnf is transcriptionally regulated by Estradiol and Lncrna Hotair. J Biol Chem. (2024) 300. doi: 10.1016/j.jbc.2024.106593

Crossref Full Text | Google Scholar

135. Rishi, A, Bhan, A, Deb, P, Mandal, S, Guha, P, Perrotti, L, et al. Dynamic regulation of Bdnf gene expression by Estradiol and Lncrna Hotair. Gene. (2024) 897:148055. doi: 10.1016/j.gene.2023.148055

Crossref Full Text | Google Scholar

136. Chow, R, Wessels, J, and Foster, W. Brain-derived Neurotrophic factor (Bdnf) expression and function in the mammalian reproductive tract. Hum Reprod Update. (2020) 26:545–64. doi: 10.1093/humupd/dmaa008

Crossref Full Text | Google Scholar

137. Tse, MCL, Pang, BPS, Bi, X, Ooi, TX, Chan, WS, Zhang, J, et al. Estrogen regulates mitochondrial activity through inducing brain-derived Neurotrophic factor expression in skeletal muscle. J Cell Physiol. (2025) 240:e31483. doi: 10.1002/jcp.31483

Crossref Full Text | Google Scholar

138. Lopez-Verrilli, MA, Picou, F, and Court, FA. Schwann cell-derived Exosomes enhance axonal regeneration in the peripheral nervous system. Glia. (2013) 61:1795–806. doi: 10.1002/glia.22558

Crossref Full Text | Google Scholar

139. Hyung, S, Im, SK, Lee, BY, Shin, J, Park, JC, Lee, C, et al. Dedifferentiated Schwann cells secrete Progranulin that enhances the survival and axon growth of motor neurons. Glia. (2019) 67:360–75. doi: 10.1002/glia.23547

Crossref Full Text | Google Scholar

140. Webber, CA, Christie, KJ, Cheng, C, Martinez, JA, Singh, B, Singh, V, et al. Schwann cells direct peripheral nerve regeneration through the Netrin-1 receptors, dcc and Unc5h2. Glia. (2011) 59:1503–17. doi: 10.1002/glia.21194

Crossref Full Text | Google Scholar

141. Liu, D, Liu, Z, Liu, H, Li, H, Pan, X, and Li, Z. Brain-derived Neurotrophic factor promotes vesicular glutamate transporter 3 expression and Neurite outgrowth of dorsal root ganglion neurons through the activation of the transcription factors Etv4 and Etv5. Brain Res Bull. (2016) 121:215–26. doi: 10.1016/j.brainresbull.2016.02.010

Crossref Full Text | Google Scholar

142. Naoi, M, Wu, Y, Shamoto-Nagai, M, and Maruyama, W. Mitochondria in Neuroprotection by phytochemicals: bioactive polyphenols modulate mitochondrial apoptosis system, function and structure. Int J Mol Sci. (2019) 20:2451. doi: 10.3390/ijms20102451

Crossref Full Text | Google Scholar

143. Biasutto, L, Szabo, I, and Zoratti, M. Mitochondrial effects of plant-made compounds. Antioxid Redox Signal. (2011) 15:3039–59. doi: 10.1089/ars.2011.4021

Crossref Full Text | Google Scholar

144. Varghese, N, Werner, S, Grimm, A, and Eckert, A. Dietary Mitophagy enhancer: a strategy for healthy brain aging? Antioxidants. (2020) 9. doi: 10.3390/antiox9100932

Crossref Full Text | Google Scholar

145. Vásquez-Reyes, S, Velázquez-Villegas, LA, Vargas-Castillo, A, Noriega, LG, Torres, N, and Tovar, AR. Dietary bioactive compounds as modulators of mitochondrial function. J Nutr Biochem. (2021) 96:108768. doi: 10.1016/j.jnutbio.2021.108768

Crossref Full Text | Google Scholar

146. Tripathi, A, Ray, A, Mishra, S, Bishen, SM, Mishra, H, and Khurana, A. Molecular and therapeutic insights of alpha-Lipoic acid as a potential molecule for disease prevention. Rev Bras. (2023) 33:272–87. doi: 10.1007/s43450-023-00370-1

Crossref Full Text | Google Scholar

147. Song, G, Liu, Z, Wang, L, Shi, R, Chu, C, Xiang, M, et al. Protective effects of Lipoic acid against acrylamide-induced neurotoxicity: involvement of mitochondrial energy metabolism and autophagy. Food Funct. (2017) 8:4657–67. doi: 10.1039/c7fo01429e

Crossref Full Text | Google Scholar

148. Superti, F, and Russo, R. Alpha-Lipoic acid: biological mechanisms and health benefits. Antioxidants. (2024) 13. doi: 10.3390/antiox13101228

Crossref Full Text | Google Scholar

149. Hu, Q, and Sun, A-J. Cardioprotective effect of alpha-Lipoic acid and its mechanisms. Cardiology Plus. (2020) 5:109–17. doi: 10.4103/cp.cp_16_20

Crossref Full Text | Google Scholar

150. Shanaida, M, Lysiuk, R, Mykhailenko, O, Hudz, N, Abdulsalam, A, Gontova, T, et al. Alpha-Lipoic acid: An antioxidant with anti-aging properties for disease therapy. Curr Med Chem. (2025) 32:23–54. doi: 10.2174/0109298673300496240416114827

Crossref Full Text | Google Scholar

151. Golbidi, S, Badran, M, and Laher, I. Diabetes and alpha Lipoic acid. Front Pharmacol. (2011) 2:69. doi: 10.3389/fphar.2011.00069

Crossref Full Text | Google Scholar

152. Ventura-Clapier, R, Garnier, A, and Veksler, V. Transcriptional control of mitochondrial biogenesis: the central role of Pgc-1alpha. Cardiovasc Res. (2008) 79:208–17. doi: 10.1093/cvr/cvn098

Crossref Full Text | Google Scholar

153. Liang, H, and Ward, W. Pgc-1alpha: a key regulator of energy metabolism. Adv Physiol Educ. (2006) 30:145–51. doi: 10.1152/advan.00052.2006

Crossref Full Text | Google Scholar

154. Srivastava, S, Diaz, F, Iommarini, L, Aure, K, Lombes, A, and Moraes, CT. Pgc-1alpha/Beta induced expression partially compensates for respiratory chain defects in cells from patients with mitochondrial disorders. Hum Mol Genet. (2009) 18:1805–12. doi: 10.1093/hmg/ddp093

Crossref Full Text | Google Scholar

155. Rambout, X, Cho, H, Blanc, R, Lyu, Q, Miano, JM, Chakkalakal, JV, et al. Pgc-1α senses the Cbc of pre-Mrna to dictate the fate of promoter-proximally paused Rnapii. Mol Cell. (2023) 83:186–202.e11. doi: 10.1016/j.molcel.2022.12.022

Crossref Full Text | Google Scholar

156. Hernández-Camacho, J, Bernier, M, López-Lluch, G, and Navas, P. Coenzyme Q10 supplementation in aging and disease. Front Physiol. (2018) 9:44. doi: 10.3389/fphys.2018.00044

Crossref Full Text | Google Scholar

157. Rizzardi, N, Liparulo, I, Antonelli, G, Orsini, F, Riva, A, Bergamini, C, et al. Coenzyme Q10 Phytosome formulation improves Coq10 bioavailability and mitochondrial functionality in cultured cells. Antioxidants. (2021) 10:927. doi: 10.3390/antiox10060927

Crossref Full Text | Google Scholar

158. Gutierrez-Mariscal, F, Larriva, AA-D, Limia-Pérez, L, Romero-Cabrera, J, Yubero-Serrano, E, and López-Miranda, J. Coenzyme Q10 supplementation for the reduction of oxidative stress: clinical implications in the treatment of chronic diseases. Int J Mol Sci. (2020) 21:7870. doi: 10.3390/ijms21217870

Crossref Full Text | Google Scholar

159. Pradhan, N, Singh, C, and Singh, A. Coenzyme Q10 a mitochondrial restorer for various brain disorders. Naunyn Schmiedeberg's Arch Pharmacol. (2021) 394:2197–222. doi: 10.1007/s00210-021-02161-8

Crossref Full Text | Google Scholar

160. Kumazawa, T, Sato, K, Seno, H, Ishii, A, and Suzuki, O. Levels of Pyrroloquinoline Quinone in various foods. Biochem J (1995) 307:331–3. doi:doi: 10.1042/bj3070331

Crossref Full Text | Google Scholar

161. Jonscher, K, Chowanadisai, W, and Rucker, R. Pyrroloquinoline-Quinone is more than an antioxidant: a vitamin-like accessory factor important in health and disease prevention. Biomolecules. (2021) 11:1441. doi: 10.3390/biom11101441

Crossref Full Text | Google Scholar

162. Chowanadisai, W, Bauerly, K, Tchaparian, E, Wong, A, Cortopassi, G, and Rucker, R. Pyrroloquinoline Quinone stimulates mitochondrial biogenesis through camp response element-binding protein phosphorylation and increased Pgc-1α expression*. J Biol Chem. (2010) 285:142–52. doi: 10.1074/jbc.M109.030130

Crossref Full Text | Google Scholar

163. Daniel, M, and Smith, EL. Promising roles of Phytocompounds and nutrients in interventions to mitigate chemotherapy-induced peripheral neuropathy. Semin Oncol Nurs. (2024) 40:151713. doi: 10.1016/j.soncn.2024.151713

Crossref Full Text | Google Scholar

164. Brami, C, Bao, T, and Deng, G. Natural products and complementary therapies for chemotherapy-induced peripheral neuropathy: a systematic review. Crit Rev Oncol Hematol. (2016) 98:325–34. doi: 10.1016/j.critrevonc.2015.11.014

Crossref Full Text | Google Scholar

165. Xu, X, Jia, L, Xiaoran, LH, and Sun, C. Application potential of plant-derived medicines in prevention and treatment of platinum-induced peripheral neurotoxicity. Front Pharmacol. (2022) 13:792331. doi: 10.3389/fphar.2021.792331

Crossref Full Text | Google Scholar

166. Eghbali, A, Adibifar, M, Ghasemi, A, Afzal, RR, Moradi, K, Eghbali, A, et al. The effect of Oral Curcumin on vincristine-induced neuropathy in Pediatric acute lymphoblastic Leukemia: a double-blind randomized controlled clinical trial. BMC Cancer. (2025) 25:344. doi: 10.1186/s12885-025-13751-7

Crossref Full Text | Google Scholar

167. Asadi, S, Gholami, MS, Siassi, F, Qorbani, M, Khamoshian, K, and Sotoudeh, G. Nano Curcumin supplementation reduced the severity of diabetic sensorimotor polyneuropathy in patients with type 2 diabetes mellitus: a randomized double-blind placebo- controlled clinical trial. Complement Ther Med (2019) 43:253–60. doi:doi: 10.1016/j.ctim.2019.02.014.

Crossref Full Text | Google Scholar

168. Zhao, W, Zhang, B, Liao, M, Zhang, W, He, W-Y, Wang, H-B, et al. Curcumin ameliorated diabetic neuropathy partially by inhibition of Nadph oxidase mediating oxidative stress in the spinal cord. Neurosci Lett. (2014) 560:81–5. doi: 10.1016/j.neulet.2013.12.019

Crossref Full Text | Google Scholar

169. Jia, T, Rao, J, Zou, L, Zhao, S, Yi, Z, Wu, B, et al. Nanoparticle-encapsulated Curcumin inhibits diabetic neuropathic pain involving the P2y12 receptor in the dorsal root ganglia. Front Neurosci. (2018) 25:755. doi: 10.3389/fnins.2017.00755

Crossref Full Text | Google Scholar

170. Elsayed, H, Rabei, M, Elshaer, M, Nashar, EE, Alghamdi, M, Al-Qahtani, Z, et al. Suppression of neuronal apoptosis and glial activation with modulation of Nrf2/Ho-1 and Nf-kb Signaling by Curcumin in Streptozotocin-induced diabetic spinal cord central neuropathy. Front Neuroanat. (2023) 9:1094301. doi: 10.3389/fnana.2023.1094301

Crossref Full Text | Google Scholar

171. Caillaud, M, Thompson, D, Toma, W, White, A, Mann, J, Roberts, J, et al. Formulated Curcumin prevents paclitaxel-induced peripheral neuropathy through reduction in Neuroinflammation by modulation of Α7 nicotinic acetylcholine receptors. Pharmaceutics. (2022) 14:1296. doi: 10.3390/pharmaceutics14061296

Crossref Full Text | Google Scholar

172. Fouda, W, Aboyoussef, A, Messiha, B, Salman, M, and Taye, A. New insights into the Cytoprotective mechanism of Nano Curcumin on neuronal damage in a vincristine-induced neuropathic pain rat model. Pak J Pharm Sci. (2023) 36:149–57.

Google Scholar

173. Kuhad, A, Pilkhwal, S, Sharma, S, Tirkey, N, and Chopra, K. Effect of Curcumin on inflammation and oxidative stress in Cisplatin-induced experimental nephrotoxicity. J Agric Food Chem. (2007) 55:10150–5. doi: 10.1021/jf0723965

Crossref Full Text | Google Scholar

174. Khadrawy, YA, El-Gizawy, MM, Sorour, SM, Sawie, HG, and Hosny, EN. Effect of Curcumin nanoparticles on the Cisplatin-induced neurotoxicity in rat. Drug Chem Toxicol. (2019) 42:194–202. doi: 10.1080/01480545.2018.1504058

Crossref Full Text | Google Scholar

175. Khalatbary, A, and Khademi, E. The green tea Polyphenolic Catechin Epigallocatechin Gallate and Neuroprotection. Nutr Neurosci. (2018) 23:281–94. doi: 10.1080/1028415X.2018.1500124

Crossref Full Text | Google Scholar

176. Mandel, S, Avramovich-Tirosh, Y, Reznichenko, L, Zheng, H, Weinreb, O, Amit, T, et al. Multifunctional activities of green tea Catechins in Neuroprotection. Neurosignals. (2005) 14:46–60. doi: 10.1159/000085385

Crossref Full Text | Google Scholar

177. Pervin, M, Unno, K, Takagaki, A, Isemura, M, and Nakamura, Y. Function of green tea Catechins in the brain: Epigallocatechin Gallate and its metabolites. Int J Mol Sci. (2019) 20:3630. doi: 10.3390/ijms20153630

Crossref Full Text | Google Scholar

178. Nain, CW, Mignolet, E, Herent, MF, Quetin-Leclercq, J, Debier, C, Page, MM, et al. The Catechins profile of green tea extracts affects the antioxidant activity and degradation of Catechins in Dha-rich oil. Antioxidants. (2022) 11:1844. doi: 10.3390/antiox11091844

Crossref Full Text | Google Scholar

179. Pervin, M, Unno, K, Ohishi, T, Tanabe, H, Miyoshi, N, and Nakamura, Y. Beneficial effects of green tea Catechins on neurodegenerative diseases. Molecules. (2018) 23:1297. doi: 10.3390/molecules23061297

Crossref Full Text | Google Scholar

180. Afzal, O, Dalhat, M, Altamimi, A, Rasool, R, Alzarea, S, Almalki, W, et al. Green tea Catechins attenuate neurodegenerative diseases and cognitive deficits. Molecules. (2022) 27:7604. doi: 10.3390/molecules27217604

Crossref Full Text | Google Scholar

181. Mandel, S, Weinreb, O, Amit, T, and Youdim, M. Cell Signaling pathways in the Neuroprotective actions of the green tea polyphenol (−)-Epigallocatechin-3-Gallate: implications for neurodegenerative diseases. J Neurochem. (2004) 88:1555–69. doi: 10.1046/j.1471-4159.2003.02291.x

Crossref Full Text | Google Scholar

182. Akbarialiabad, H, Dahroud, MD, Khazaei, M, Razmeh, S, and Zarshenas, M. Green tea, a medicinal food with promising neurological benefits. Curr Neuropharmacol. (2020) 19:349–59. doi: 10.2174/1570159X18666200529152625

Crossref Full Text | Google Scholar

183. Arafa, M, and Atteia, H. Protective role of Epigallocatechin Gallate in a rat model of Cisplatin-induced cerebral inflammation and oxidative damage: impact of modulating Nf-Κb and Nrf2. Neurotox Res. (2019) 37:380–96. doi: 10.1007/s12640-019-00095-x

Crossref Full Text | Google Scholar

184. El-Mowafy, A, Al-Gayyar, M, Salem, H, El-Mesery, M, and Darweish, M. Novel chemotherapeutic and renal protective effects for the green tea (Egcg): role of oxidative stress and inflammatory-cytokine Signaling. Phytomedicine. (2010) 17:1067–75. doi: 10.1016/j.phymed.2010.08.004

Crossref Full Text | Google Scholar

185. Huang, Y-J, Wang, K-L, Chen, H-Y, Chiang, Y-F, and Hsia, S. Protective effects of Epigallocatechin Gallate (Egcg) on endometrial, breast, and ovarian cancers. Biomolecules. (2020) 10:1481. doi: 10.3390/biom10111481

Crossref Full Text | Google Scholar

186. Fatima, S, Suhail, N, Alrashed, M, Wasi, S, Aljaser, FS, AlSubki, RA, et al. Epigallocatechin Gallate and coenzyme Q10 attenuate Cisplatin-induced hepatotoxicity in rats via targeting mitochondrial stress and apoptosis. J Biochem Mol Toxicol. (2021) 35:e22701. doi: 10.1002/jbt.22701

Crossref Full Text | Google Scholar

187. Gundimeda, U, McNeill, T, Schiffman, J, Hinton, D, and Gopalakrishna, R. Green tea polyphenols potentiate the action of nerve growth factor to induce Neuritogenesis: possible role of reactive oxygen species. J Neurosci Res. (2010) 88:3644–55. doi: 10.1002/jnr.22519

Crossref Full Text | Google Scholar

188. Zhang, Y, Han, M, Sun, X, Gao, G, Yu, G, Huang, L, et al. Egcg promotes Neurite outgrowth through the integrin Β1/Fak/P38 Signaling pathway after subarachnoid Hemorrhage. Evid Based Complement Alternat Med. (2021) 25:8810414. doi: 10.1155/2021/8810414

Crossref Full Text | Google Scholar

189. Essmat, A, and Hussein, MS. Green tea extract for mild-to-moderate diabetic peripheral neuropathy a randomized controlled trial. Complement Ther Clin Pract. (2021) 43:101317. doi: 10.1016/j.ctcp.2021.101317

Crossref Full Text | Google Scholar

190. Baba, Y, Sonoda, J, Hayashi, S, Tosuji, N, Sonoda, S, Makisumi, K, et al. Reduction of oxidative stress in liver cancer patients by Oral green tea polyphenol tablets during hepatic arterial infusion chemotherapy. Exp Ther Med. (2012) 4:452–8. doi: 10.3892/etm.2012.602

Crossref Full Text | Google Scholar

191. Wei, R, Wirkus, J, Yang, Z, Machuca, J, Esparza, Y, and Mackenzie, G. Egcg sensitizes chemotherapeutic-induced cytotoxicity by targeting the Erk pathway in multiple cancer cell lines. Arch Biochem Biophys. (2020) 692:108546. doi: 10.1016/j.abb.2020.108546

Crossref Full Text | Google Scholar

192. Li, H, Krstin, S, and Wink, M. Modulation of multidrug resistant in cancer cells by Egcg, tannic acid and Curcumin. Phytomedicine. (2018) 50:213–22. doi: 10.1016/j.phymed.2018.09.169

Crossref Full Text | Google Scholar

193. Hu, F, Wei, F, Wang, Y, Wu, B, Fang, Y, and Xiong, B. Egcg synergizes the therapeutic effect of Cisplatin and Oxaliplatin through Autophagic pathway in human colorectal cancer cells. J Pharmacol Sci. (2015) 128:27–34. doi: 10.1016/j.jphs.2015.04.003

Crossref Full Text | Google Scholar

194. Heyza, JR, Arora, S, Zhang, H, Conner, KL, Lei, W, Floyd, AM, et al. Targeting the DNA repair endonuclease Ercc1-Xpf with green tea polyphenol Epigallocatechin-3-Gallate (Egcg) and its Prodrug to enhance Cisplatin efficacy in human cancer cells. Nutrients. (2018) 10:1644. doi: 10.3390/nu10111644

Crossref Full Text | Google Scholar

195. Miao, H, Li, R, Chen, D, Hu, J, Chen, Y, Xu, C, et al. Protective effects of vitamin E on chemotherapy-induced peripheral neuropathy: a meta-analysis of randomized controlled trials. Ann Nutr Metab. (2020) 77:127–37. doi: 10.1159/000515620

Crossref Full Text | Google Scholar

196. Chen, J, Shan, H, Yang, W, Zhang, J, Dai, H, and Ye, Z. Vitamin E for the prevention of chemotherapy-induced peripheral neuropathy: a meta-analysis. Front Pharmacol. (2021) 12:684550. doi: 10.3389/fphar.2021.684550

Crossref Full Text | Google Scholar

197. Pace, A, Giannarelli, D, Galiè, E, Savarese, A, Carpano, S, Giulia, M, et al. Vitamin E Neuroprotection for Cisplatin neuropathy. Neurology. (2010) 74:762–6. doi: 10.1212/WNL.0b013e3181d5279e

Crossref Full Text | Google Scholar

198. Argyriou, A, Chroni, E, Koutras, A, Iconomou, G, Papapetropoulos, S, Polychronopoulos, P, et al. A randomized controlled trial evaluating the efficacy and safety of vitamin E supplementation for protection against Cisplatin-induced peripheral neuropathy: final results. Support Care Cancer. (2006) 14:1134–40. doi: 10.1007/s00520-006-0072-3

Crossref Full Text | Google Scholar

199. Fu, X, Wu, H, Li, J, Wang, C, Li, M, Qianqian,, et al. Efficacy of drug interventions for chemotherapy-induced chronic peripheral neurotoxicity: a network meta-analysis. Front Neurol. (2017) 8:223. doi: 10.3389/fneur.2017.00223

Crossref Full Text | Google Scholar

200. Kottschade, L, Sloan, J, Mazurczak, M, Johnson, D, Murphy, B, Rowland, K, et al. The use of vitamin E for the prevention of chemotherapy-induced peripheral neuropathy: results of a randomized phase iii clinical trial. Support Care Cancer. (2011) 19:1769–77. doi: 10.1007/s00520-010-1018-3

Crossref Full Text | Google Scholar

201. Heiba, MA, Ismail, S, Sabry, M, Bayoumy, W, and Kamal, K. The use of vitamin E in preventing Taxane-induced peripheral neuropathy. Cancer Chemother Pharmacol. (2021) 88:931–9. doi: 10.1007/s00280-021-04347-6

Crossref Full Text | Google Scholar

202. Kottschade, L, Sloan, J, Mazurczak, M, Johnson, D, Murphy, B, Rowland, K, et al. The use of vitamin E for prevention of chemotherapy-induced peripheral neuropathy: a phase iii double-blind, placebo controlled study-N05c31. J Clin Oncol. (2016):27 15_suppl:9532. doi: 10.1200/jco.2009.27.15_suppl.9532

Crossref Full Text | Google Scholar

203. Khan, A, Samad, F, Samad, S, Khan, A, Arif, S, and Zahid, R. Role of vitamin E in prevention of chemotherapy induced peripheral neuropathy. Int J Approx Reason. (2020) 8:1260–5. doi: 10.21474/ijar01/11213

Crossref Full Text | Google Scholar

204. Huang, H-P, He, M, Liu, L, and Huang, L. Vitamin E does not decrease the incidence of chemotherapy-induced peripheral neuropathy: a meta-analysis. Contemp Oncol. (2016) 20:237–41. doi: 10.5114/wo.2016.61567

Crossref Full Text | Google Scholar

205. Eum, S, Choi, H, Chang, M, Choi, H, Ko, Y-J, Ahn, J, et al. Protective effects of vitamin E on chemotherapy-induced peripheral neuropathy: a meta-analysis of randomized controlled trials. Int J Vitamin Nutr Res. (2013) 83:101–11. doi: 10.1024/0300-9831/a000149

Crossref Full Text | Google Scholar

206. Huang, HY, and Appel, LJ. Supplementation of diets with alpha-Tocopherol reduces serum concentrations of gamma- and Delta-Tocopherol in humans. J Nutr. (2003) 133:3137–40. doi: 10.1093/jn/133.10.3137

Crossref Full Text | Google Scholar

207. Handelman, GJ, Machlin, LJ, Fitch, K, Weiter, JJ, and Dratz, EA. Oral alpha-Tocopherol supplements decrease plasma gamma-Tocopherol levels in humans. J Nutr. (1985) 115:807–13. doi: 10.1093/jn/115.6.807

Crossref Full Text | Google Scholar

208. Reiter, E, Jiang, Q, and Christen, S. Anti-inflammatory properties of alpha- and gamma-Tocopherol. Mol Asp Med. (2007) 28:668–91. doi: 10.1016/j.mam.2007.01.003

Crossref Full Text | Google Scholar

209. Ahsan, H, Ahad, A, Iqbal, J, and Siddiqui, WA. Pharmacological potential of Tocotrienols: a review. Nutr Metab. (2014) 11:52. doi: 10.1186/1743-7075-11-52

Crossref Full Text | Google Scholar

210. Razali, RA, Ngah, WZW, Makpol, S, Yanagisawa, D, Kato, T, and Tooyama, I. Shifting perspectives on the role of Tocotrienol Vs. Tocopherol in brain health: a scoping review. Int J Mol Sci. (2025) 26:6339. doi: 10.3390/ijms26136339

Crossref Full Text | Google Scholar

211. Zaaboul, F, and Liu, Y. Vitamin E in foodstuff: nutritional, analytical, and food technology aspects. Compr Rev Food Sci Food Saf. (2022) 21:964–98. doi: 10.1111/1541-4337.12924

Crossref Full Text | Google Scholar

212. Shahidi, F, Fuentes, J, De Camargo, AC, Speisky, H, and Pinaffi-Langley, A. Vitamin E as an essential micronutrient for human health: common, novel, and unexplored dietary sources. Free Radic Biol Med. (2021) 176:312–21. doi: 10.1016/j.freeradbiomed.2021.09.025

Crossref Full Text | Google Scholar

213. Burton, G, Traber, M, Acuff, R, Walters, D, Kayden, H, Hughes, L, et al. Human plasma and tissue alpha-Tocopherol concentrations in response to supplementation with Deuterated natural and synthetic vitamin E. Am J Clin Nutr. (1998) 67:669–84. doi: 10.1093/AJCN/67.4.669

Crossref Full Text | Google Scholar

214. Traber, M. Utilization of vitamin E. Biofactors. (1999) 10:115–20. doi: 10.1002/biof.5520100205

Crossref Full Text | Google Scholar

215. Clemente, H, Ramalho, H, Lima, M, Grilo, E, and Dimenstein, R. Maternal supplementation with natural or synthetic vitamin E and its levels in human colostrum. J Pediatr Gastroenterol Nutr. (2015) 60:533–7. doi: 10.1097/MPG.0000000000000635

Crossref Full Text | Google Scholar

216. Gaur, S, and Sherry, C. Alpha-Tocopherol stereoisomer profiles of lactating women supplemented with natural and synthetic vitamin E. FASEB J. (2016) 30:1150. doi: 10.1096/fasebj.30.1_supplement.1150.2

Crossref Full Text | Google Scholar

217. Zhang, AC, De Silva, M, MacIsaac, R, Roberts, L, Kamel, J, Craig, J, et al. Omega-3 polyunsaturated fatty acid Oral supplements for improving peripheral nerve health: a systematic review and meta-analysis. Nutr Rev. (2020) 78:323–41. doi: 10.1093/nutrit/nuz054

Crossref Full Text | Google Scholar

218. Samuels, N, and Ben-Arye, E. Integrative approaches to chemotherapy-induced peripheral neuropathy. Curr Oncol Rep. (2020) 22:23. doi: 10.1007/s11912-020-0891-2

Crossref Full Text | Google Scholar

219. Ghoreishi, Z, Esfahani, A, Djazayeri, A, Djalali, M, Golestan, B, Ayromlou, H, et al. Omega-3 fatty acids are protective against paclitaxel-induced peripheral neuropathy: a randomized double-blind placebo controlled trial. BMC Cancer (2012) 12:355-. doi:doi: 10.1186/1471-2407-12-355

Crossref Full Text | Google Scholar

220. Kucharska, E, Majsterek, I, Mrowicka, M, and Mrowicki, J. Lutein and zeaxanthin and their roles in age-related macular degeneration—neurodegenerative disease. Nutrients. (2022) 14:827. doi: 10.3390/nu14040827

Crossref Full Text | Google Scholar

221. Şanlıer, N, Yildiz, E, and Ozler, E. An overview on the effects of some carotenoids on health: lutein and Zeaxanthin. Current Nutrition Reports. (2024) 13:828–44. doi: 10.1007/s13668-024-00579-z

Crossref Full Text | Google Scholar

222. Tan, L, Zhang, Y, Dawson, R, and Kong, L. Roles of macular carotenoids in brain function throughout the lifespan: a review of recent research. J Agriculture Food Research. (2023) 14:100785. doi: 10.1016/j.jafr.2023.100785

Crossref Full Text | Google Scholar

223. Lindbergh, C, Renzi-Hammond, L, Hammond, B, Terry, D, Mewborn, C, Puente, A, et al. Lutein and Zeaxanthin influence brain function in older adults: a randomized controlled trial. J Int Neuropsychol Soc. (2017) 24:77–90. doi: 10.1017/S1355617717000534

Crossref Full Text | Google Scholar

224. Johnson, E. Role of lutein and Zeaxanthin in visual and cognitive function throughout the lifespan. Nutr Rev. (2014) 72:605–12. doi: 10.1111/nure.12133

Crossref Full Text | Google Scholar

225. Lopresti, A, Smith, S, and Drummond, P. The effects of lutein and Zeaxanthin supplementation on cognitive function in adults with self-reported mild cognitive complaints: a randomized, double-blind, placebo-controlled study. Front Nutr. (2022) 9:843512. doi: 10.3389/fnut.2022.843512

Crossref Full Text | Google Scholar

226. Davinelli, S, Ali, S, Solfrizzi, V, Scapagnini, G, and Corbi, G. Carotenoids and cognitive outcomes: a meta-analysis of randomized intervention trials. Antioxidants. (2021) 10:223. doi: 10.3390/antiox10020223

Crossref Full Text | Google Scholar

227. Zhong, Q, Sun, W-H, Qin, Y, and Xu, H. Association of Dietary Α-carotene and Β-carotene intake with low cognitive performance in older adults: a cross-sectional study from the National Health and nutrition examination survey. Nutrients. (2023) 15:239. doi: 10.3390/nu15010239

Crossref Full Text | Google Scholar

228. Christensen, K, Gleason, C, and Mares, J. Dietary carotenoids and cognitive function among us adults, Nhanes 2011–2014. Nutr Neurosci. (2020) 23:554–62. doi: 10.1080/1028415X.2018.1533199

Crossref Full Text | Google Scholar

229. Shanaida, M, Mykhailenko, O, Lysiuk, R, Hudz, N, Balwierz, R, Shulhai, A, et al. Carotenoids for Antiaging: Nutraceutical, pharmaceutical, and Cosmeceutical applications. Pharmaceuticals. (2025) 18:403. doi: 10.3390/ph18030403

Crossref Full Text | Google Scholar

230. Pietrasik, S, Cichoń, N, Bijak, M, Gorniak, L, and Saluk-Bijak, J. Carotenoids from marine sources as a new approach in neuroplasticity enhancement. Int J Mol Sci. (2022) 23:1990. doi: 10.3390/ijms23041990

Crossref Full Text | Google Scholar

231. Cho, K, Shin, M, Kim, S, and Lee, SB. Recent advances in studies on the therapeutic potential of dietary carotenoids in neurodegenerative diseases. Oxidative Med Cell Longev 2018;2018:4120458. doi:doi: 10.1155/2018/4120458

Crossref Full Text | Google Scholar

232. Manochkumar, J, Doss, C, El-Seedi, H, Efferth, T, and Ramamoorthy, S. The Neuroprotective potential of carotenoids in vitro and in vivo. Phytomedicine. (2021) 91:153676. doi: 10.1016/j.phymed.2021.153676

Crossref Full Text | Google Scholar

233. Park, H-A, Hayden, MM, Bannerman, S, Jansen, J, and Crowe-White, K. Anti-apoptotic effects of carotenoids in Neurodegeneration. Molecules. (2020) 25:3453. doi: 10.3390/molecules25153453

Crossref Full Text | Google Scholar

234. Zaa, C, Marcelo, Á, An, Z, Medina-Franco, J, and Velasco-Velázquez, M. Anthocyanins: molecular aspects on their Neuroprotective activity. Biomolecules. (2023) 13:1598. doi: 10.3390/biom13111598

Crossref Full Text | Google Scholar

235. Zhang, J, Wu, J, Liu, F, Tong, L, Chen, Z, Chen, J, et al. Neuroprotective effects of Anthocyanins and its major component Cyanidin-3-O-Glucoside (C3g) in the central nervous system: An outlined review. Eur J Pharmacol. (2019) 858:172500. doi: 10.1016/j.ejphar.2019.172500

Crossref Full Text | Google Scholar

236. Ma, H, Johnson, SL, Liu, W, DaSilva, NA, Meschwitz, S, Dain, JA, et al. Evaluation of polyphenol anthocyanin-enriched extracts of blackberry, black raspberry, blueberry, cranberry, red raspberry, and strawberry for free radical scavenging, reactive carbonyl species trapping, anti-Glycation, anti-Β-amyloid aggregation, and microglial Neuroprotective effects. Int J Mol Sci. 2018;19:461. doi:doi: 10.3390/ijms19020461

Crossref Full Text | Google Scholar

237. Singh, L, Wani, AW, Shaifali, SRK, Kaur, H, Bashir, O, Farid, A, et al. A comprehensive review on Neurotrophic receptors and their implications in brain health: exploring the Neuroprotective potential of berries. J Berry Research. (2024) 15:48–64. doi: 10.1177/18785093241301881

Crossref Full Text | Google Scholar

238. Abbas, S, Latif, M, Shafie, N, Ghazali, M, and Kormin, F. Neuroprotective expression of turmeric and Curcumin. J Food Sci. (2020) 4:2366–81. doi: 10.26656/fr.2017.4(6).363

Crossref Full Text | Google Scholar

239. Silva, G, Da Cruz Monteiro Santana, T, ACL, V, APA, M, Gonçalves, M, Couto, RD, et al. Green tea intake reduces high-fat diet-induced sensory neuropathy in mice by Upregulating the antioxidant Defense system in the spinal cord. Antioxidants. (2025) 14:452. doi: 10.3390/antiox14040452

Crossref Full Text | Google Scholar

240. Somers-Edgar, T, Scandlyn, M, Stuart, E, Nedelec, L, Valentine, S, and Rosengren, R. The combination of Epigallocatechin Gallate and Curcumin suppresses Erα-breast cancer cell growth in vitro and in vivo. Int J Cancer. (2008) 122:1966–71. doi: 10.1002/ijc.23328

Crossref Full Text | Google Scholar

241. Hewlings, S, and Kalman, D. Curcumin: a review of its’ effects on human health. Foods. (2017) 6:92. doi: 10.3390/foods6100092

Crossref Full Text | Google Scholar

242. Kesarwani, K, Gupta, R, and Mukerjee, A. Bioavailability enhancers of herbal origin: An overview. Asian Pac J Trop Biomed. (2013) 3:253–66. doi: 10.1016/s2221-1691(13)60060-x

Crossref Full Text | Google Scholar

243. Shoba, G, Joy, D, Joseph, T, Majeed, M, Rajendran, R, and Srinivas, PS. Influence of Piperine on the pharmacokinetics of Curcumin in animals and human volunteers. Planta Med. (1998) 64:353–6. doi: 10.1055/s-2006-957450

Crossref Full Text | Google Scholar

244. Gao, Q, Rong, SL, Wang, S, Liang, Y, and Zhang, Z. Anti-Glycation and anti-inflammatory activities of Anthocyanins from purple vegetables. Food Funct. (2023) 14:2034–44. doi: 10.1039/d2fo03645b

Crossref Full Text | Google Scholar

245. Khoo, HE, Azlan, A, Tang, S, and Lim, SM. Anthocyanidins and Anthocyanins: Colored pigments as food, pharmaceutical ingredients, and the potential health benefits. Food Nutr Res. (2017) 61:1361779. doi: 10.1080/16546628.2017.1361779

Crossref Full Text | Google Scholar

246. Mattioli, R, Francioso, A, Mosca, L, and Silva, P. Anthocyanins: a comprehensive review of their chemical properties and health effects on cardiovascular and neurodegenerative diseases. Molecules. (2020) 25:3809. doi: 10.3390/molecules25173809

Crossref Full Text | Google Scholar

247. Heidari, H, Hajhashemy, Z, and Saneei, P. A meta-analysis of effects of vitamin E supplementation alone and in combination with Omega-3 or magnesium on polycystic ovary syndrome. Sci Rep. (2022) 12:19927. doi: 10.1038/s41598-022-24467-0

Crossref Full Text | Google Scholar

248. Jamilian, M, Shojaei, A, Samimi, M, Ebrahimi, FA, Aghadavod, E, Karamali, M, et al. The effects of Omega-3 and vitamin E co-supplementation on parameters of mental health and gene expression related to insulin and inflammation in subjects with polycystic ovary syndrome. J Affect Disord. (2018) 229:41–7. doi: 10.1016/j.jad.2017.12.049

Crossref Full Text | Google Scholar

249. Shahidi, F, and de Camargo, AC. Tocopherols and Tocotrienols in Common and Emerging Dietary Sources: Occurrence, Applications, and Health Benefits. Int J Mol Sci. (2016) 17:1745. doi: 10.3390/ijms17101745

Crossref Full Text | Google Scholar

250. Blesso, C. Dietary Anthocyanins and human health. Nutrients. (2019) 11:2107. doi: 10.3390/nu11092107

Crossref Full Text | Google Scholar

251. Panchal, S, John, O, Mathai, M, and Brown, L. Anthocyanins in chronic diseases: the power of purple. Nutrients. (2022) 14:2161. doi: 10.3390/nu14102161

Crossref Full Text | Google Scholar

252. Eweys, A, Zhao, Y, Zhang, J, Xiao, X, Bai, J, Darwesh, O, et al. Fermentation affects the antioxidant activity of plant-based food material through the release and production of bioactive components. Antioxidants. (2021) 10:2004. doi: 10.3390/antiox10122004

Crossref Full Text | Google Scholar

253. Etu, R, Offia-Olua, B, and Ukom, A. Fermentation and Sauteing enhances the physicochemical properties, carotenoids and the antioxidant activity of some food vegetables. Measurement: Food. (2024) 15:100177. doi: 10.1016/j.meafoo.2024.100177

Crossref Full Text | Google Scholar

254. Sarıtaş, S, Portocarrero, A, Lopez, JM, Lombardo, M, Koch, W, Raposo, A, et al. The impact of fermentation on the antioxidant activity of food products. Molecules. (2024) 29:3941. doi: 10.3390/molecules29163941

Crossref Full Text | Google Scholar

255. Steffens, JP, and Costa, CAD. The use of dietary supplements for the Management of Adverse Effects of treatment in children and adolescents with Leukemia: a scoping review. Nutr Cancer. (2025) 77:334–340. doi: 10.1080/01635581.2024.2435079

Crossref Full Text | Google Scholar

256. Gala, D, Wright, H, Zigori, B, Marshall, S, and Crichton, M. Dietary strategies for chemotherapy-induced nausea and vomiting: a systematic review. Clin Nutr. (2022) 41:2147–55. doi: 10.1016/j.clnu.2022.08.003

Crossref Full Text | Google Scholar

257. Abene, J, and Deng, J. Evaluating the role of dietary interventions in reducing chemotherapy toxicities in cancer patients: a systematic review. J Cancer Surviv. (2025). doi: 10.1007/s11764-025-01777-6

Crossref Full Text | Google Scholar

258. Leardini, D, Bossù, G, Venturelli, F, Baccelli, F, Muratore, E, Grasso, A, et al. Food safety practices and foodborne illness in Italian Pediatric oncology and Hematology Centers: a survey on behalf of the infectious disease working Group of Aieop. Pediatr Blood Cancer. (2025) 72:e31782. doi: 10.1002/pbc.31782

Crossref Full Text | Google Scholar

259. Morris, A, O'Connor, G, and Renshaw, G. A cross-sectional survey to review food safety practices within Pediatric oncology and stem cell transplant Centers in the United Kingdom. J Pediatr Hematol Oncol. (2023):45. doi: 10.1097/MPH.0000000000002649

Crossref Full Text | Google Scholar

260. Vries, Y, Berg, M, De Vries, JHM, Boesveldt, S, Kruif, J, Buist, N, et al. Differences in dietary intake during chemotherapy in breast cancer patients compared to women without cancer. Support Care Cancer. (2017) 25:2581–91. doi: 10.1007/s00520-017-3668-x

Crossref Full Text | Google Scholar

261. Putri, S, Adriani, M, and Estuningsih, Y. Hubungan Antara Nafsu Makan Dengan Asupan Energi Dan protein Pada Pasien Kanker Payudara post Kemoterapi[correlation between appetite with energy and protein intake of post chemotherapy breast cancer patients]. Media Gizi Indones. (2019) 14:170. doi: 10.20473/mgi.v14i2.170-176

Crossref Full Text | Google Scholar

262. Regyna, SD, Adriani, M, and Rachmah, Q. A systematic review: Asupan Zat Gizi Makro dan Status Gizi Pasien Kanker yang Menjalani Kemoterapi – A systematic review: Macro nutrient intake and nutritional status of cancer patients undergoing chemotherapy. Media Gizi Indonesia. (2021) 16:182–193. doi: 10.20473/mgi.v16i2.182-193

Crossref Full Text | Google Scholar

263. Anderson, PM, Thomas, SM, Sartoski, S, Scott, JG, Sobilo, K, Bewley, S, et al. Strategies to mitigate chemotherapy and radiation toxicities that affect eating. Nutrients. (2021) 13:4397. doi: 10.3390/nu13124397

Crossref Full Text | Google Scholar

264. Armeli, F, Bonucci, A, Maggi, E, Pinto, A, and Businaro, R. Mediterranean diet and neurodegenerative diseases: the neglected role of nutrition in the modulation of the Endocannabinoid system. Biomolecules. (2021) 11:790. doi: 10.3390/biom11060790

Crossref Full Text | Google Scholar

265. Chen, L, Zhong, W, Chen, H, Zhou, Y, Ran, W, He, Y, et al. Orient diet: a potential Neuroprotective dietary pattern for Chinese stroke high-risk population. J Am Med Dir Assoc. (2025) 26:105331. doi: 10.1016/j.jamda.2024.105331

Crossref Full Text | Google Scholar

266. Di Lorenzo, C, Colombo, F, Biella, S, Stockley, C, and Restani, P. Polyphenols and human health: the role of bioavailability. Nutrients. (2021) 13:273. doi: 10.3390/nu13010273

Crossref Full Text | Google Scholar

267. Jäger, R, Lowery, RP, Calvanese, AV, Joy, JM, Purpura, M, and Wilson, JM. Comparative absorption of Curcumin formulations. Nutr J. (2014) 13:11. doi: 10.1186/1475-2891-13-11

Crossref Full Text | Google Scholar

268. Tabanelli, R, Brogi, S, and Calderone, V. Improving Curcumin bioavailability: current strategies and future perspectives. Pharmaceutics. (2021) 13:1715. doi: 10.3390/pharmaceutics13101715

Crossref Full Text | Google Scholar

269. Halliwell, B. Are polyphenols antioxidants or pro-oxidants? What do we learn from cell culture and in vivo studies? Arch Biochem Biophys. (2008) 476:107–12. doi: 10.1016/j.abb.2008.01.028

Crossref Full Text | Google Scholar

270. Babich, H, Schuck, AG, Weisburg, JH, and Zuckerbraun, HL. Research strategies in the study of the pro-oxidant nature of polyphenol Nutraceuticals. J Toxicol. (2011) 2011:467305. doi: 10.1155/2011/467305

Crossref Full Text | Google Scholar

271. Duda-Chodak, A, and Tarko, T. Possible side effects of polyphenols and their interactions with medicines. Molecules. (2023) 28:2536. doi: 10.3390/molecules28062536

Crossref Full Text | Google Scholar

272. Nabekura, T, Kawasaki, T, Furuta, M, Kaneko, T, and Uwai, Y. Effects of natural polyphenols on the expression of drug efflux transporter P-glycoprotein in human intestinal cells. Acs Omega. (2018) 3:1621–6. doi: 10.1021/acsomega.7b01679

Crossref Full Text | Google Scholar

273. Santos, W, Guimarães, J, Pina, L, Serafini, M, and Guimarães, A. Antinociceptive effect of plant-based natural products in chemotherapy-induced peripheral neuropathies: a systematic review. Front Pharmacol. (2022) 13:1001276. doi: 10.3389/fphar.2022.1001276

Crossref Full Text | Google Scholar

Keywords: chemotherapy-induced peripheral neuropathy, plant-derived antioxidants, neuroprotection, curcumin, green tea, oxidative stress

Citation: Li W, Yang D, Zhang Z, Wang M, Xu S, Jiang R, Chen J, Wu C and Qian J (2025) Plant food-derived antioxidant nutrients in neuroprotection against chemotherapy-induced neurotoxicity: from molecular mechanisms to clinical applications. Front. Nutr. 12:1667421. doi: 10.3389/fnut.2025.1667421

Received: 16 July 2025; Accepted: 30 September 2025;
Published: 05 December 2025.

Edited by:

Jianmei Li, Nanjing Normal University, China

Reviewed by:

Rahul Kumar Singh, Banaras Hindu University, India
Yun Li, Beijing Tiantan Hospital, China

Copyright © 2025 Li, Yang, Zhang, Wang, Xu, Jiang, Chen, Wu and Qian. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Jun Qian, anVuX3FpYW5Abmp1Y20uZWR1LmNu; Chaokui Wu, MTM2OTY0OTE2NjBAMTYzLmNvbQ==

These authors have contributed equally to this work

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.