IMR Press / FBL / Volume 30 / Issue 10 / DOI: 10.31083/FBL39800
Open Access Review
Ionizing Radiation in Clinical Diagnostics and Radiotherapy: The Dual Role of NRF2 in Cell Protection and Carcinogenesis
Show Less
Affiliation
1 Unit of Cellular Networks and Molecular Therapeutic Targets, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
2 Departmental Faculty of Medicine, Unicamillus-Saint Camillus, University of Health Sciences, 00131 Rome, Italy
*Correspondence: alessandra.verdina@ifo.it (Alessandra Verdina)
Front. Biosci. (Landmark Ed) 2025, 30(10), 39800; https://doi.org/10.31083/FBL39800
Submitted: 17 April 2025 | Revised: 16 July 2025 | Accepted: 21 July 2025 | Published: 31 October 2025
(This article belongs to the Special Issue NRF2 and Cancer)
Copyright: © 2025 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Ionizing radiations (IRs), commonly used in both diagnostic imaging and cancer therapy, generate reactive oxygen species (ROS) and free radicals, causing significant DNA damage that can lead to genetic mutations, cell death, and tissue injury in both normal and tumor tissues. In response to the oxidative stress, the nuclear factor erythroid 2-related factor 2 (NRF2) is activated to induce target genes involved in antioxidant and detoxifying pathways, thereby playing a pivotal role in protecting cells from IR-induced oxidative damage. In clinical diagnostics, IR exposure from imaging techniques can result in DNA damage, inflammation, and increased risk of IR-induced pathologies, including cancer. NRF2 activation in response to these diagnostic exposures can help to protect normal tissues from damage by boosting antioxidant defenses. In radiotherapy, IR induces DNA damage to kill malignant cells, although it may also harm surrounding healthy tissue. Cancer cells exploit NRF2 activation to resist IR-induced cell damage, thereby maintaining redox balance and protecting themselves from oxidative stress. In that case, NRF2 inhibition could sensitize cancer cells to IR effects by disrupting their antioxidant defense, leading to increased ROS accumulation, enhanced DNA damage, and greater cell death. This review will summarize the role of NRF2 in mediating the response to IR in both healthy and cancerous cells, with a focus on its effects in clinical diagnostic and radiotherapy.

Keywords
nuclear factor erythroid 2-related factor 2 (NRF2)
kelch-like ECH-associated protein 1 (Keap1)
reactive oxygen species (ROS)
ionizing radiation (IR)
oxidative stress
DNA repair
cancer therapy
inflammation
cancer fibrosis
X-ray imaging
1. Introduction

Nuclear factor erythroid 2-related factor 2 (NRF2) is a key transcription factor that protects cells from oxidative stress and cell damage caused by environmental stressors, including ionizing radiations (IRs). Under normal conditions, NRF2 is kept inactive in the cytoplasm by its interaction with Kelch-like ECH-associated protein 1 (Keap1), an E3 ubiquitin ligase which drives NRF2 degradation [1]. Under oxidative stress, the interaction between Keap1 and NRF2 is impaired, allowing NRF2 to stabilize and translocate to the nucleus to activate target gene promoters with antioxidant response elements (ARE) whose encoded proteins help protect cells from oxidative stress [2, 3]. In tumour, NRF2 plays a double role. During the first phases of carcinogenesis its activation can be mainly cytoprotective by suppressing oxidative stress and tumor-promoting inflammation, both responsible of increased DNA mutations. On the other hand, NRF2 hyperactivation in many cancers, by different mechanisms, may support tumorigenesis, resistance to chemo- and radiotherapy, and enhanced cancer cell survival by strengthening antioxidant defenses and detoxification pathways that counteract cell death induced by oxidative stress and increase genetic instability [4]. Such dysregulation supports cancer progression, metastasis, and resistance to therapies, making NRF2 a potential attractive therapeutic target to enhance the efficacy of the current cancer treatments [5].

IRs, including those used in medical diagnostic and cancer therapy, can cause oxidative stress and DNA damage [6]. In diagnostic imaging, low doses of IR may increase cancer risk over time. The presence of oxidatively-induced clustered DNA lesions (OCDLs) challenges the linear no-threshold (LNT) model, as even low radiation doses can cause complex, hard-to-repair damage. Due to their high biological impact, OCDLs may disproportionately drive mutations and cancer, suggesting low doses could be more harmful than previously assumed [7, 8, 9, 10]. In radiotherapy, higher doses of IR target cancer cells but can also damage healthy tissues, leading to DNA breaks, inflammation, and eventually secondary cancers [11]. IR can directly interact with DNA causing single-strand or double-strand breaks, or induce indirect DNA damage through water ionization and reactive oxygen species (ROS) generation, which further harm cellular components [11, 12]. Emerging research now shows that NRF2-mediated transcription can protect cells and tissues from the pathogenic consequences of hydroxyl radicals that are directly generated by IR as well as of the hydrogen peroxide and superoxide that are generated as a secondary consequence of irradiation which can have an impact on both normal and cancer tissues.

In this review, we explore the role of NRF2 in the response to the damage caused by IR, with a focus on its effects in clinical diagnostic and radiotherapy.

2. NRF2

NRF2 is a transcription factor encoded by the NFE2L2 gene and belonging to the cap “n” collar subfamily of basic-region leucine zipper proteins [13]. It has a central role in cytoprotection by reducing oxidative stress and maintaining redox homeostasis. NRF2 regulates the expression of numerous target genes critical for cellular processes like detoxification and cytoprotection, particularly those involved in protecting against oxidative stress and supporting cellular homeostasis including heme oxygenase 1 (HO-1), Aldo-keto reductase (AKR), NADPH quinone oxidoreductase 1 (NQO-1), superoxide dismutase (SOD), catalase, multidrug resistance-associated protein (MRP), and ATP-binding cassette (ABC) transporters [14]. Additionally, it exerts anti-inflammatory activity by regulating a variety of downstream genes such as Interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α), inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX2), and plays a vital role in DNA repair mechanisms encoding DNA repair proteins and enzymes involved in non-homologous end joining (NHEJ), homologous recombination (HR), base excision repair (BER), and nucleotide excision repair (NER) [15, 16, 17, 18]. Additionally, NRF2 regulates genes linked to autophagy, apoptosis, and metabolism, further supporting its role in cellular protection and stress adaptation (Table 1) [16, 19, 20].

Table 1. NRF2 target genes and biological activity.
Biological activity Target genes
Antioxidant HO-1, GPx, Trx, TrxR, PrxR, SOD, catalase, GSR
Detoxyfication and Transport AKR, NQO-1, MRP, ABC transporters, GST, GCLC, GCLM
Inflammation IL-6, TNF-α, iNOS, COX2, MCP-1, Mip2
DNA repair 53BP1, XRCC1, GADD45, OGG1, PARP-1
Cell death Bcl-2, Caspase-3, SOD1, cIAP1

Abbreviations: 53BP1, p53 binding protein-1; ABC, (ATP-binding cassette) transporters; AKR, Aldo-keto reductase; Bcl-2, B-cell lymphoma 2; cIAP1, cellular inhibitor of apoptosis protein-1; COX2, cyclooxygenase-2; GADD45, growth-arrest and DNA-damage inducible 45; GCLC, glutamate-cysteine ligase; GCLM, glutamate-cysteine ligase modifier; GPx, gluthathione peroxidase; GSR, glutathione reductase; GST, glutathione S-transferase; HO-1, heme oxygenase-1; IL-6, interleukin-6; iNOS, inducible nitric oxide synthase; MCP-1, monocyte chemoattractant protein-1; Mip2, macrophage inflammatory protein-2; MRP, multidrug resistance-associated protein; NQO-1, NADPH quinone oxidoreductase-1; NRF-2, nuclear factor erythroid 2-related factor 2; OGG1, 8-Oxoguanine glycosylase; PARP-1, Poly (ADP-ribose) polymerase-1; PrxR, peroxiredoxin; SOD, superoxide dismutase; TNF-α, tumor necrosis factor alpha; Trx, thioredoxin; TrxR, thioredoxin reductase; XRCC1, X-ray repair cross complementing group 1.

2.1 NRF2 Activation

Under normal conditions, NRF2 is retained in the cytoplasm through its interaction with two proteins: Keap1, an actin cytoskeleton-associated protein with 25 cysteines that function as “sensors” for the NFR2/Keap1 regulatory system [1], and Cullin 3 (CUL3), a scaffold protein that is part of the E3 ubiquitin ligase complex [21]. This complex mediates NRF2 ubiquitination, which is then targeted for proteasomal degradation (Fig. 1A, left side). As a reaction to oxidative stress, cysteines 273 and 288 on Keap1 are modified by reactive oxygen species and electrophiles leading to protein conformational changes. As a result, the Keap1-CUL3-NRF2 ubiquitination system is impaired, hindering the ubiquitination of NRF2 and causing its accumulation in the cytoplasm [22]. Afterward, NRF2 translocates to the nucleus, where it forms a heterodimer with one of the small musculoaponeurotic fibrosarcoma (MAF) proteins and binds to antioxidant response elements (AREs) in the promoter regions of target genes (Fig. 1A, right side) [4]. Besides this canonical activation, NRF2 can be activated via a non-canonical mechanism by Ser349 phosphorylated p62/sequestome 1 (SQSTM1) (p-STGE), a key autophagyc adaptor that interacts with Keap1, inducing its degradation through autophagy, thereby triggering NRF2 stabilization and activation (Fig. 1B) [23]. Additionally, NRF2 activation can be regulated by a feedback mechanism involving (BTB Domain and CNC Homolog 1) BACH1 [24], a physiological repressor of NRF2 that is itself stabilized by NRF2 (Fig. 1C) [25, 26]. Both NRF2 and BACH1 associate to form a heterodimer with MAF proteins and bind to ARE sites in the promoters of cytoprotective genes. Under basal conditions or low oxidative stress, MAF-BACH1 heterodimers occupy these ARE sites, acting as transcriptional repressors (Fig. 1C, left side). As a reaction to oxidative stress, NRF2 is activated and its increased availability and affinity for AREs displace the BACH1/MAF complex from the ARE sites leading to activation of protective gene expression [27]. Notably, NRF2 activation can also induce BACH1 expression via ARE sites in its promoter, establishing a negative feedback circuit that limits the duration and intensity of the NRF2 response (Fig. 1C, right side) [28].

Fig. 1.

Nuclear factor erythroid 2-related factor 2 (NRF2) activation. (A) Under normal conditions, NRF2 is retained in the cytoplasm through its interaction with Keap1 and Cullin3 (CUL3) that allows proteasomal ubiquitination and degradation. During oxidative stress, the Keap1-CUL3-NRF2 ubiquitination system is disrupted, allowing NRF2 to accumulate in the cytoplasm and translocate to the nucleus. In the nucleus, NRF2 forms a heterodimer with MAF proteins and binds to antioxidant response elements (AREs) in the promoter regions of target genes promoting transcription (canonical activation). (B) In a non-canonical activation pathway, p-STGE (Ser349 phosphorylated p62 motif) competes with NRF2 for Keap1 binding, leading to the dissociation of NRF2. This allows NRF2 to accumulate in the cytoplasm, translocate to the nucleus, bind to target genes, and promote transcription. (C) Under basal conditions, BACH1 (BTB Domain and CNC Homolog 1) occupies ARE sites, repressing transcription of target genes. Following oxidative stress, NRF2 is activated and displaces the BACH1/MAF complex from the ARE sites, activating the expression of protective gene, including BACH1, creating a negative feedback loop (dotted arrow). ROS, reactive oxygen species; IR, ionizing radiation.

2.2 The Dual Role of NRF2 in Cancer

NRF2 plays a complex and dual role in cancer, influencing different stages of tumorigenesis in either protective or pro-tumorigenic ways and acting on both healthy and tumor cells [4, 29]. Despite the progress in the knowledge of NRF2 in recent years, there is still no clear consensus on its exact role during carcinogenesis [30, 31]. In normal cells, including the very early stages of neoplastic transformation, NRF2, through the expression of antioxidant and detoxifying enzymes, protects the cells against oxidative stress and cellular damage and therefore blocks carcinogenesis [32] (Fig. 2, upper panel). However, a tumor-promoting role for NRF2 in cancer initiation has been reported linked, for instance, to its protection against redox stress in cells with mutations in kirsten rat sarcoma viral oncogene homolog (KRAS) and/or serine/threonine kinase 11 (STK11) [33]. In the subsequent stages of tumor promotion and in the neoplastic progression, the role of NRF2 shifts, as its persistent activation becomes a key driver of cancer cell survival and proliferation and resistance to cancer treatments, often in an interplay with oncogenic pathways such as signal transducer and activator of transcription 3 (STAT3) or mutant p53 [34, 35] (Fig. 2, lower panel). By upregulating a variety of detoxification and antioxidant genes, NRF2 supports the adaptation of tumor cells to the harsh tumor microenvironment, providing resistance to cytotoxic effects of chemo- and radio-therapy in many types of tumors including breast, lung, pancreatic, head and neck, colon, etc., [36, 37, 38, 39, 40, 41, 42, 43, 44]. NRF2 activation also protects cancer cells from ferroptosis, an iron-dependent and lipid peroxidation-driven cell death cascade, occurring when there is an imbalance of redox homeostasis in the cell [45]. NRF2 upregulates multiple genes that block ferroptosis such as SLC7A11 (xCT) or glutamate-cysteine ligase (GCLC)/glutamate-cysteine ligase modifier (GCLM), modulates iron metabolism genes to limit iron-driven ROS, and limit the availability of polyunsaturated fatty acids (PUFAs)—the main substrates for lipid peroxidation [46]. NRF2 also protects cells from apoptosis, allowing cancer cells to escape death especially the one induced by anticancer treatments [20, 47]. Furthermore, NRF2 has been implicated in promoting metastasis, partly by facilitating epithelial-to-mesenchymal transition (EMT), which enhances the invasive capacity of tumor cells [26, 48, 49, 50]. Thus, while NRF2 initially serves as a protective response to cellular oxidative stress to block carcinogenesis, its chronic activation in established cancers contributes to tumor growth, metastasis, and therapy resistance. In summary, this dual role makes NRF2 a challenging molecular target for therapeutic intervention in cancer [30], as its inhibition may on one hand improve cancer cell response to cytotoxic cancer treatments [51, 52, 53], but, on the other hand, may impair its normal protective functions in healthy cells contributing to carcinogenesis. Therefore, more precise targeting strategies need to be taken in consideration [31, 54].

Fig. 2.

Dual role of NRF2 in both normal and cancer cells. In normal cells or early stage carcinogenesis, NRF2 acts as a protector against oxidative stress-induced cellular damage. In established tumors, NRF2 can acquire a pro-tumorigenic function. Its persistent activation can support tumor growth and proliferation by inducing the expression of antioxidant and detoxifying enzymes. “” means increase.

3. IR Injury and Cell Damage

IR interacts with biological tissues generating either direct or indirect effects [12, 55]. The extent of DNA damage caused by IR depends primarily on the ionization density, absorbed dose, dose rate, and linear energy transfer (LET) [56]. The high linear energy transfer (LET) IRs, i.e., alpha-particles and heavy ions, induce damage mostly by direct action, whereas the low LET IR, i.e., X-Rays and gamma-rays, by indirect one. Direct effects occur by the direct ionization and excitation of DNA molecules, which disrupt the molecular structure. The direct interaction can result in DNA single-strand breaks (SSBs) or double-strand breaks (DSBs) which, if not properly repaired, can result in mutations, genomic instability, or cellular death [57]. Indirect effects, in contrast, occur through the radiolysis of water, leading to the formation of highly reactive species like hydroxyl radicals (OH•), hydrogen peroxide (H2O2), and superoxide anion (O2•-). These reactive oxygen species (ROS) can cause damage to cellular components, including DNA, proteins, and lipids. In addition, reactive nitrogen species (RNS) such as nitric oxide (NO•) and peroxynitrite (ONOO-) may form, contributing to further cellular damage. This cascade of oxidative and nitrosative stress can result in DNA mutations, cell death, and in a broad range of biological effects, from immediate tissue damage to long-term outcomes such as cancer development [58].

The most severe damage resulting from IR exposure is DNA DSBs, which is mainly repaired through non-homologous end joining (NHEJ) and homologous recombination (HR) mechanisms [59, 60, 61, 62]. These repair processes rely on a network of factors that detect, repair, and maintain genomic stability, ensuring the cell can recover from IR-induced DNA injury or, if necessary, trigger appropriate cell death to prevent genomic instability. Key sensors like ataxia telangiectasia mutated (ATM) and ATM and Rad3-related (ATR) detect DSBs and SSBs, respectively, and initiate a signaling cascade involving checkpoint proteins like checkpoint kinase 1 (CHK1) and CHK2 to arrest the cell cycle and allow DNA damage repair [63, 64]. If the damage is irreparable, oncosuppressor p53 triggers permanent cell cycle arrest by replicative senescence or cell death by apoptosis [65]. Additionally, proteins such as breast cancer gene 1 (BRCA1) and BRCA2 play crucial roles in DNA repair and checkpoint regulation, particularly in the HR pathway [66].

IR used in clinical setting have distinct effects depending on whether they are employed for diagnosis purposes or radiotherapy and also present risks because of their ability to induce molecular and cellular damage, as seen above [67]. In diagnostic applications, IR are primarily used to create detailed scans of internal bodily organs. Techniques such as X-rays, computed tomography (CT), mammography and nuclear medicine aid in the diagnosis of various medical conditions. X-rays are commonly employed to obtain images of bones, detect fractures, and diagnose lung conditions like tuberculosis [68]. CT scans, which involve a series of X-ray images combined to create detailed cross-sectional views, provide comprehensive information on complex issues such as tumors, internal bleeding, and organ diseases [69]. Mammography, is a critical imaging modality in the screening of breast carcinoma, utilizing IR to obtain detailed breast tissue images that can identify nodules, calcifications, or other abnormalities indicative of breast cancer [70]. In nuclear medicine, small amounts of radioactive isotopes are used to create images of organs and tissues, providing functional information about the body’s physiology through diagnostic procedures like positron emission tomography (PET) and single-photon emission computed tomography (SPECT) [71] (Fig. 3). These procedures are generally quick, non-invasive, and offer valuable diagnostic insights. However, despite the relatively low doses used, this exposure can potentially cause cellular damage or increase the risk of cancer over time. It is therefore essential to carefully assess the frequency of diagnostic tests and IR doses used to manage and minimize the risk of potential harm [72, 73]. At the moment, unlike CT or X-Ray exposure, PET and SPECT do not have large-scale epidemiological studies linking them directly to increase cancer incidence.

Fig. 3.

IR in diagnostic imaging and therapy. Diagnostic and therapeutic application of Low and High doses of IR in clinical applications. PET, positron emission tomography; SPECT, single-photon emission computed tomography; IMRT, intensity-modulated IR therapy; SBRT, stereotactic body radiotherapy.

On the other hand, radiotherapy involves higher doses of IR aimed at destroying cancerous cells or shrinking tumors. Indeed, radiotherapy is a cornerstone in the treatment of various cancers, including lung [74, 75], breast [76], cervical [77], prostate [78] and colorectal cancers [79], often as part of a multi-modal approach, combined with surgery, chemotherapy, or immunotherapy to improve outcomes [80]. It is used in curative treatment, adjuvant therapy post-surgery, or in palliative care to alleviate symptoms in advanced cancer stages. In Europe, the cancers with the highest number of patients in radiotherapy departments are breast cancer, lung cancer, prostate cancer, head and neck cancer, and rectal cancer [81]. Advances in IR technology, such as intensity-modulated IR therapy (IMRT), stereotactic body radiotherapy (SBRT), and proton therapy, have significantly improved precision, enabling targeted IR delivery to tumors while minimizing exposure to surrounding healthy tissues [82]. However, despite the precision of modern radiotherapy techniques, the higher doses used can cause side effects, including damage to normal tissues, skin reactions, fatigue, or long-term effects such as secondary cancers [83, 84]. While the therapeutic benefits of radiotherapy are substantial, the potential risks necessitate careful monitoring and planning to balance efficacy with patient safety.

4. NRF2 in Response to IR

NRF2 is involved in cellular response to IR and promotes a pro-survival response in irradiated cells and tissues, both healthy and cancer ones, through ROS detoxification, supporting DNA repair and modulating cytokine responses [85]. Literature data show that the activation of NRF2 signaling, either through electrophilic modification of Keap1 or through deficiency in Keap1 expression, leads to a reduction in intracellular ROS levels, increased cell viability and resistance to treatments including radiotherapy, in almost healthy and tumor cells [30, 86, 87, 88] The involvement of NRF2 in protecting from IR damage was first demonstrated in a preclinical study using a lung cancer cell line carrying Keap1 gene mutation with consequent NRF2 hyperactivation. The authors showed that the constitutive overexpression of NRF2 leads to low intracellular ROS levels and IR-resistant cancer cell phenotype [89]. Importantly, shRNA-mediated reduction of NRF2 expression induces ROS generation and increases protein oxidation resulting in enhanced sensitivity to radiation-induced cell death [89]. Similar results were obtained in mouse embryo fibroblasts (MEF) with gain of NRF2 function (Keap1-⁣/-) as well loss of NRF2 function (NRF2-⁣/-). The Keap1-⁣/- MEF cells with high antioxidant capacity and low endogenous ROS levels were shown to be resistant to ionizing radiation-induced cell death. On the contrary, NRF2-⁣/- MEF with low antioxidant capacity and high endogenous ROS levels resulted in enhanced sensitivity to γ-irradiation, leading to decreased survival. Overall, the results from cancer cells and nontumorigenic MEF cells unequivocally establish that high levels of NRF2 confer resistance to radiotherapy [89].

Repeated pretreatment of human fibroblasts exposed to IR with sulforaphane (SFN), a natural substance present in cruciferous vegetables that activates NRF2 [90], has been demonstrated to protect fibroblasts from IR. This leads to reduced ROS levels, decreased DNA damage and increased cell survival [91]. In agreement, SFN treatment was unable to protect NRF2 knockout mouse embryonic fibroblasts, indicating that the sulforaphane-induced radioprotection was NRF2-dependent. Conversely, the use of RNA interference or pharmacological inhibition of NRF2 in these same cell types results in increased ROS production and a radiosensitive phenotype [91]. Similar results were obtained in colon cancer cells, with or without NRF2 knockdown with the CRISPR-Cas9 technology, treated with sulforaphane and chemotherapy. The results showed that SFN reduces cisplatin-induced cell death only in NRF2-proficient cells compared to NRF2-Cas9 cells. Mechanistically, the authors found that NRF2 activation protects NRF2-proficient cells from the drug-induced DNA damage and the apoptotic function of the unfolded protein response (UPR) [92], suggesting a common function of NRF2 in response to chemo- and radiotherapy.

In the study by McDonald and colleagues [88], the authors report that IR activates NRF2-ARE pathway but this occurs only after a significant delay of five days. This activation was detected in breast cancer cells, both with single doses of IR (2–8 Gy) and with clinically relevant daily dose fractions (0.5–4 Gy), and it was shown to be dose-dependent [88]. An important aspect is that no ARE activation was found in the 24 hours following IR exposure, even at high doses of up to 10 Gy. This suggests that the NRF2-ARE response to IR exposure is a second-tier antioxidant response, which is activated only a little later after exposure. Moreover, they observed that NRF2 activation prior to IR exposure does not improve the survival of cells or animals after irradiation, indicating that early activation of the pathway does not offer protection against IR damage under normal conditions. Conversely, the loss of NRF2 makes the cells significantly more radiosensitive, both in vitro and in vivo [88].

IR was also shown to activate NRF2 in a dose-dependent manner in non-small cell lung cancer (NSCLC) cells. RNAi-mediated reduction of NRF2 significantly increases endogenous ROS levels, and decreases the expression of NRF2 target genes, dampening Notch1 expression and inducing IR-induced cellular apoptosis. These authors conclude that the NRF2-mediated Notch signaling is an important determinant in radio-resistance of lung cancer cells [93].

Knockdown of NRF2 shows potential DNA damage after X-rays irradiation in lung cancers. The authors showed that NRF2 knockdown disrupts damaged DNA repair by inhibiting DNA-dependent protein kinase catalytic subunit and interfering with Rad51 expression, suggesting that NRF2 plays a critical role in the development of radio-resistance by upregulating DNA damage response via the mitogen-activated protein kinase (MAPK) pathway [94].

In a more recent study, the authors showed that NRF2-⁣/- MEFs are more sensitive to spontaneous and to IR-induced transformation and that NRF2 deficiency increases IR-induced NF-κB pro-inflammatory responses most robustly late after exposure. The tendency of NRF2 to restrain inflammation is also reflected in the reprogramming of tumor antigen-specific lymphocyte responses in mice where NRF2 knockout switches Th2 responses to Th1 polarity, suggesting that targeting NRF2, especially in those tumors that constitutively express it, is an appealing way to enhance the therapeutic benefit of radiation and immunotherapy [95].

Altogether, these data highlight the key role of NRF2 in cellular protection from the IR-induced damage and in pro-survival and radio-resistance in tumors.

4.1 Mechanisms of NRF2-Mediated Response to IR

In this paragraph the different mechanisms of NRF2-mediated response to IR will be analysed.

4.1.1 Reduction of Oxidative Stress

Exposure to IR induces oxidative stress leading to the production of ROS responsible for DNA damage. The activation of NRF2 following IR exposure induces the increased expression of antioxidant enzymes that neutralize ROS, thereby preventing cellular damage caused by IR [88] as above described. IR-induced damage can activate also Sirtuins such as Sirtuin 1 (SIRT1), that induces DNA repair and antioxidant defense. NRF2 and SIRT1 are therefore important players in the defense of normal tissues from IR damage, working through antioxidant and DNA repair mechanisms. On the other hand, NRF2 and SIRT1 overactivation in tumors induces radio-resistance. Their interconnected pathways highlight the complexity of cellular responses to radiation and offer potential targets for therapeutic interventions to protect against radiation-induced damage [4, 96].

4.1.2 Modulation of DNA Repair

In addition to its antioxidant activity, NRF2 plays a crucial role in repairing the DNA damage induced by IR through the HR pathway. Jayakumar and colleagues [97] investigated NRF2 role in DNA repair using two cancer cell models, where they inhibited NRF2 both pharmacologically and genetically. Their study confirmed that the inhibition of NRF2 results in increased radiosensitization in tumor cells, and consequent reduced survival fraction after IR exposure. Notably, NRF2 inhibition caused a significant delay in DNA repair, as evidenced by the persistence of residual DNA damage. This effect was not directly linked to NRF2-mediated antioxidant function. Even when ROS levels were reduced with N-acetyl cysteine (NAC), NRF2 inhibition still impaired DNA repair, suggesting that NRF2 involvement in DNA repair is independent of its activity in redox homeostasis. The authors found that NRF2 primarily affects the repair of DNA through the HR pathway, a critical pathway for DNA DSB repair. In support of this, NRF2 inhibition resulted in a significant reduction in IR-induced Rad51 foci formation, with Rad51 being a key protein in HR and its foci serving as a marker of HR pathway activation. Additionally, NRF2 inhibition decreased the mRNA levels of Rad51, indicating that NRF2 regulates Rad51 expression. However, NRF2 did not appear to affect the NHEJ repair pathway. When both NRF2 and DNA-dependent protein kinase (DNA-PK) (a key enzyme in NHEJ) were inhibited, there was a synergistic reduction in cell survival, suggesting that NRF2 does not act through this pathway [97].

According to Kim and colleagues [98], NRF2 repairs the broken DNA duplexes primarily by regulating p53 binding protein-1 (53BP1), a key component of DNA damage response. They found that NRF2 binds to the three ARE core sequences present in the 53BP1 promoter, resulting in an increase in 53BP1 expression. As described by Sekhar and Freeman [85], NRF2 plays a role in IR-induced DNA repair, also through base excision repair (BER). Chromatin immunoprecipitation (ChIP) assays have shown that NRF2 binds to the promoter of 8-Oxoguanine glycosylase 1 (OGG1), a glycosylase involved in base excision repair [99], and RNA interference experiments have demonstrated that NRF2 deficiencies suppress OGG1 expression [100]. The authors showed that OGG1 deficiency increases IR sensitivity in human cells, thus supporting the hypothesis of a mechanistic link between NRF2, DNA base damage repair, and IR sensitivity [100].

In addition, it was shown that NRF2 preserves genomic integrity by facilitating ATR activation and G2 cell cycle arrest [101]. Consistent with previous reports [97, 102], NRF2 influenced the protein levels of BRCA1 and Rad51. The inhibition of NRF2 by brusatol increases the radio-sensitivity of tumor cells in xenografts by perturbing ATR and CHK1 activation [101].

4.1.3 Modulation of IR-Induced Inflammation

IR-induced damage that results from both indirect and direct interactions between IR and biological molecules leads to tissue response including inflammation, fibrosis, ferroptosis, and endothelial dysfunction [103, 104, 105, 106, 107, 108]. NRF2 activation in response to IR-induced oxidative stress may also modulate inflammation, either by stimulating or suppressing inflammatory signals [103]. NRF2 achieves the anti-inflammatory effects by modulating antioxidant and anti-inflammatory genes, such as for instance the HO-1 axis, which is a potent anti-inflammatory target, by modulating macrophages, and by interacting with other key inflammatory pathways like NF-κB, playing an important role in the tumor microenvironment [103].

Several studies suggest a complex interaction between NRF2 and NF-κB-induced pathways, which regulate oxidative stress and inflammation, respectively. NRF2 deficiency can enhance NF-κB activity, increasing inflammatory cytokine production, while NF-κB can negatively modulate NRF2 activation. This interaction is influenced by factors like Keap1, IKKβ, and p62, and is cell- and tissue-specific. For instance, NF-κB can increase Keap1 expression that promotes NRF2 proteasomal degradation, can induce p62 expression that indirectly promotes NRF2 activity and therefore tumor progression and resistance to therapies; finally, IKKβ activates NF-κB pathway by phosphorylating and inducing IκB-α (an inhibitor of NF-κB) degradation, and also inhibits NRF2 through direct and indirect mechanisms [109]. Therefore, inhibiting inhibitor of nuclear factor kappa-B kinase subunit beta (IKKβ) can restore NRF2 activity, promoting antioxidant defense and radioprotection. Since ROS activate IKKβ, NRF2-mediated ROS suppression may inhibit this cascade. In the tumor microenvironment, NRF2 activation can promote the M2 macrophage phenotype which is associated with tissue repair and reduced inflammation, while NRF2 deficiency can exacerbate radiation-induced inflammation and tissue damage by potentially promoting the M1 phenotype [15]. In cancers where NRF2 is constitutively active (for example because Keap1 or NRF2 mutation) chronic NRF2 activity can desensitize cells to NF-κB regulation, resulting in dysregulated inflammatory gene expression and compensatory activation of alternative inflammatory signaling pathways (e.g., STAT3, activator protein-1 (AP-1)) [103]. Unrepaired intracellular damage caused by oxidative stress after IR exposure can lead to cell death through necrosis. Necrotic cells resulting from IR-induced damage release components such as high mobility group box 1 (HMGB1), a damage-associated molecular pattern (DAMP) molecule that triggers inflammatory response. Upon stimulation with pro-inflammatory agents such as lipopolysaccharide (LPS), tumor necrosis factor-α (TNF-α), or Interleukin 1 (IL-1), HMGB1 is released also by macrophages and monocytes during inflammatory processes [110]. Experimental research has demonstrated that NRF2-ARE pathway regulates HMGB1 release from these cells, impacting inflammation also through this mechanism [111]. Under oxidative stress, HMGB1 undergoes oxidation in Cys 23 and Cys 45, forming disulfide bonds that enable its binding to the receptor for advanced glycation end-products (RAGE) on immune cells, activating NF-κB/extracellular signal-regulated kinase (ERK) signaling and promoting inflammation [112].

4.1.4 Modulation of IR-Induced Fibrosis

IR-induced tissue fibrosis (RIF) is a common and delayed detrimental outcome of delayed IR exposure [104, 113]. It is characterized by the abnormal activation of myofibroblasts and excessive deposition of extracellular matrix components. Differentiation of fibroblasts into myofibroblasts plays a critical role in the development of fibrosis [114]. This process is promoted by increased levels of ROS in fibroblasts, which result from oxidative stress induced by IR exposure [115]. RIF can impact various organs, including lung, skin, liver, and kidney. Extensive research has elucidated that RIF involves a complex interplay of extracellular signals, such as immune cell activation and dysregulated cytokine release, as well as intracellular signaling pathways, including cyclic GMP-AMP synthase/stimulator of interferon genes (cGAS/STING), oxidative stress responses, metabolic reprogramming, and proteasomal pathway activation, all contributing to myofibroblast activation [105]. Clinical studies have highlighted the association between NRF2 and IR-induced fibrosis [116]. Traver and colleagues [117] demonstrated that upon IR exposure, NRF2 deficiency inhibits the mobilization of ΔNp63-positive stem cells, while enhances the conversion of alveolar type 2 cells into myofibroblasts under IR induction.

NRF2 exerts a protective role in fibrosis by inhibiting fibroblast-to-myofibroblast differentiation. Artaud-Macari and colleagues [118] found that reduced NRF2 expression correlates with a myofibroblastic phenotype in idiopathic pulmonary fibrosis, and NRF2 activation by sulforaphane led to the dedifferentiation of myofibroblasts. Han and colleagues [119] observed that NRF2 activation by dimethyl itaconate protects against pulmonary fibrosis by inhibiting Thioredoxin- interacting protein (TXNIP), an α-arrestin family protein that regulates intracellular ROS levels. NRF2 mediates anti-fibrotic activity by means of the inhibition of epithelial-mesenchymal transition (EMT), a major source of myofibroblasts, and TGF-β/Smad signaling, a key driver pathway of EMT. As evidence of this, NRF2 deficiency enhances TGF-β/Smad signaling, further promoting EMT and fibrotic processes. In the context of ionizing radiation, the interaction between NRF2 and TGF-β is multifaceted and often antagonistic. Oxidative stress activates NRF2, which can suppress TGF-β signaling and help mitigate radiation-induced fibrosis. On the other hand, TGF-β may influence NRF2 function, occasionally dampening its cytoprotective response [105, 120, 121].

4.1.5 Modulation of IR-Induced Ferroptosis

Ferroptosis is a programmed, oxidative stress- dependent cell death process, important in IR-induced cell death [122]. It presents with iron accumulation, lipid peroxidation [45], and excess of ROS, as it kills cells by amplifying oxidative stress or inhibiting the antioxidant system [123]. Ling and colleagues [124] have suggested that ferroptosis plays a fundamental role in regulating EMT in pulmonary fibrosis. Studies have shown that treatment of acute IR damage with ferroptosis inhibitors results in a significant decrease in ROS levels and serum inflammatory cytokines (TNF-α, IL-6), thereby reducing damage [107]. Ferroptosis can be induced by inhibiting glutathione synthesis and disrupting redox balance, thus increasing the radio-sensitivity of tumor cells [108]. A recent report also demonstrated that exosomes derived from menstrual blood stem cells and containing the microRNA miR-let-7 can inhibit ferroptosis and ameliorate pulmonary fibrosis through the specificity protein 3/histone deacetylase2/nuclear factor erythroid 2-related factor 2 (SP3/HDAC2/NRF2) signaling pathway [125].

According to recent research, NRF2 is closely involved in inhibiting ferroptosis by interfering with iron metabolism, inhibiting glutathione synthesis, and enhancing DNA repair [126, 127, 128]. NRF2 positively regulates the transcription of heme oxygenase 1 (HMOX1), increases iron storage, and reduces intracellular free iron by rapidly upregulating the transcription of ferritin heavy (FTH) and light (FTL) chains, thereby controlling the FTL/FTH ratio [129]. On the other hand, NRF2 directly promotes the expression of glutathione peroxidase 4 (GPX4), a central regulator in ferroptosis, by protecting cells against membrane lipid peroxidation and regulates the GTP cyclohydrolase-1/tetrahydrobiopterin (GCH1/BH4) pathway to mediate cellular redox reactions and inhibit ferroptosis [130, 131]. Studies have revealed that NRF2 can also mediate glutathione synthesis by promoting the expression of solute carrier family 7 member 11 (SLC7A11), glutamate-cysteine ligase (GCLC), and glutathione synthetase (GSS), which play crucial roles in preventing ferroptosis [132, 133, 134]. Hypoxia-inducible factor-1α (HIF-1α) acts as a regulator of ferroptosis. Upregulation of HIF-1α can buffer IR-induced ROS and reduce ferroptosis, thus enhancing cell radio-resistance [135]. NRF2 is also involved in the HIF-1α-mediated inhibition of ferroptosis. NRF2 silencing blocks the accumulation of HIF-1α in hypoxic tumor cells, weakening its regulatory effect on cell metabolism and leading to imbalance in ROS homeostasis [136].

These data indicate that NRF2 may prevent ferroptosis by modulating proteins involved in iron metabolism and ROS detoxification pathways, ultimately reducing oxidative stress and inflammation caused by IR and enhancing cellular radio-sensitivity.

5. NRF2 Modulation and IR-Induced Injury

Modulation of NRF2 activity through genetic or pharmacologic approaches, as described above [54, 92] significantly affects cellular and tissue injury caused by IR, including radiotherapy. Below are described the main NRF2 activating or inhibiting molecules that are used in pre-clinical studies given that at the moment there are not molecules approved for clinical use. NRF2 activators enhance resistance to IR-induced damage by increasing antioxidants and detoxifying enzymes, reducing oxidative stress and DNA damage, and improving cell survival [137]. NRF2 activation is a key mechanism for many chemopreventive agents, helping to prevent tumorigenesis, although an excessive NRF2 activation may protect tumor cells from radiotherapy, reducing treatment efficacy [138]. NRF2 activators include a variety of natural and synthetic compounds, including sulforaphane, curcumin, and seleno-hormetic molecules.

Sulforaphane is one of the most extensively studied natural compounds that targets the NRF2-Keap1 signaling pathway, recognized for its antioxidant, chemopreventive and antiproliferative properties [139]. Mathew and colleagues [91] demonstrated the radioprotective effect of sulforaphane in human skin fibroblasts. Through site-directed mutagenesis and mass spectrometry analysis, it was shown that sulforaphane can directly modify the critical cysteine residue at position 151 of Keap1, causing the activation of the NRF2-Keap1-ARE signaling pathway [140]. Curcumin is another extensively studied natural compound with many therapeutic properties, including antioxidant and anticancer activities [52, 53, 141, 142]. Notably, it was shown to act as a sensitizer for IR and chemotherapy in several human cancers, including prostate cancer, by oncogene mouse double minute2 (MDM2) downregulation [143] and colorectal cancer, by suppressing NF-κB and NF-κB-regulated gene products [144]. Khor and colleagues [145] showed that Curcumin exerts demethylating effects on the promoter region of NRF2, resulting in increased expression of NRF2 and its target genes. In addition, curcumin may indirectly phosphorylate NRF2 at serine- and/or threonine-rich regions, promoting its nuclear translocation. Furthermore, curcumin can directly interact with the cysteine thiol(s) groups of Keap1, reducing its inhibitory effect on NRF2 [146].

Seleno-hormetic compounds exhibit a broad spectrum of biological activities including anticancer and antiproliferative effects and modulate hormetic genes and antioxidant enzyme functions. Studies have demonstrated the redox-modulating and IR-protective properties of seleno-hormetic compounds showing that seleno-compounds protect against IR-induced toxicity activating NRF2 transcription factor and, consequently, upregulating the adaptive stress response to IR [147].

While several compounds that activate NRF2 have been identified, there are only a few agents recognized as NRF2 inhibitors with suitable pharmacokinetic properties for in vivo use [137]. Given the aberrant activation and the complex and varied role of NRF2 in cancer, these inhibitors are gaining interest as potential anticancer agents. Indeed, they might enhance the responsiveness of cancer cells to radio and chemotherapy and reduce chemo- and radio-resistance by downregulating the activity of enzymes responsible for detoxification and drug elimination through the inhibition of NRF2. However, NRF2 inhibitors might also increase damage to healthy tissues, causing side effects like inflammation and fibrosis [103, 115, 120, 121]. NRF2 inhibitors exert their activity at different levels depending on their characteristics: they can regulate NRF2 at the post-transcriptional level by degrading its mRNA or affecting NRF2 translation and post-translational regulation [147, 148]. They can block the translocation of NRF2 to the nucleus, or alter the binding between NRF2 and DNA [140, 149, 150]. NRF2 inhibitors can be identified in various classes of compounds, including flavonoids, natural compounds found in fruits, vegetables, and plants, with antioxidant, anti-inflammatory, and anticancer properties. While many flavonoids have been identified as NRF2 activators, some have exhibited inhibitory effects. For instance, in vivo studies have shown that luteolin reduces NRF2 mRNA and protein levels, sensitizing cells to anticancer drugs [151]. However, the effects of flavonoids on NRF2 activity are cell-type specific and concentration-dependent, and their application in cancer therapy requires further investigation. A known NRF2 inhibitor is brusatol, a natural compound extracted from the plant Brucea javanica [148]. Brusatol has been shown to reduce NRF2 protein levels by affecting NRF2 translation and post-translational regulation in various cell lines, and sensitizes A549 lung tumor cells to the anticancer drug cisplatin, both in vitro and in vivo [148, 152, 153]. However, the application of brusatol in humans has encountered challenges due to unresolved issues such as toxicity, transient nature of the reduction in NRF2 protein levels, and difficulties in drug delivery [152, 154]. Another interesting inhibitor of NRF2 is metformin (1,1-dimethylbiguanide hydrochloride), a drug used to treat type 2 diabetes and approved as an anticancer agent for certain tumors [155]. Metformin was shown to increase sensitivity to anticancer treatments by reducing NRF2 mRNA and protein levels through suppression of the rapidly accelerated fibrosarcoma (RAF)-ERK-NRF2 signaling pathway or induction of the microRNA-34a, which decreases NRF2 protein expression via the SIRT1/peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α)/NRF2 pathway [156]. However, given the complexity and variety of actions of this molecule, more research is needed to better understand its role in the NRF2 pathway and in modulating the response to anticancer treatments.

In conclusion, NRF2 modulation presents a promising yet complex therapeutic strategy, requiring a careful balance between protecting normal tissues and enhancing tumor response to treatment, particularly radiotherapy. Moreover, the double role of NRF2 in both carcinogenesis and tumor progression adds further complexity to its therapeutic targeting [157]. Therefore, the use of specific NRF2 activators or inhibitors in cancer therapy must be approached with prudence. It is also important to note that, at present, no NRF2-targeting molecules have been approved for medical use.

6. Conclusions and Future Perspectives

NRF2 plays a crucial role in the cellular response to IR used both in clinical diagnostic imaging and radiotherapy. While NRF2 activation offers protection against IR-induced oxidative damage in healthy tissues and helps prevent the onset of long-term damages, including malignant transformation, it may also promote tumor viability and resistance to radiation therapy. Inhibiting NRF2 represents an encouraging strategy to improve sensitivity of tumor cells to IR and the development of NRF2 inhibitors for clinical use as adjuncts to radiotherapy holds great potential. However, several challenges remain. One of the main obstacles is the selective targeting of NRF2 pathways in tumor cells without affecting normal tissues, which could increase the risk of side effects. Moreover, the potential for compensatory mechanisms in the cell, where other stress response pathways may take over when NRF2 is inhibited, must be carefully considered in the design of combination therapies. Further research is needed to better understand the precise role of NRF2 in IR responses and to identify the optimal timing and dosing strategies for NRF2 inhibition. Clinical trials assessing the safety and efficacy of NRF2 inhibitors, either alone or in combination with existing treatments, are essential to translating these findings into therapeutic practice.

Author Contributions

AV and GD designed the outline, wrote the original draft, and reviewed the literature. Both authors contributed to editorial changes in the manuscript. Both authors read and approved the final manuscript. Both authors have participated sufficiently in the work and agreed to be accountable for all aspects of the work.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

The authors wish to thank S. Soddu for critical reading of the manuscript.

Funding

This work was supported by the institutional “Ricerca Corrente” granted by the Italian Ministry of Health.

Conflict of Interest

The authors declare no conflict of interest. Given her role as the Guest Editor, Gabriella D’Orazi had no involvement in the peer-review of this article and has no access to information regarding its peer review. Full responsibility for the editorial process for this article was delegated to Alexandros G. Georgakilas and Graham Pawelec.

References
[1]
McMahon M, Itoh K, Yamamoto M, Hayes JD. Keap1-dependent proteasomal degradation of transcription factor Nrf2 contributes to the negative regulation of antioxidant response element-driven gene expression. The Journal of Biological Chemistry. 2003; 278: 21592–21600. https://doi.org/10.1074/jbc.M300931200.
[2]
Xiao JL, Liu HY, Sun CC, Tang CF. Regulation of Keap1-Nrf2 signaling in health and diseases. Molecular Biology Reports. 2024; 51: 809. https://doi.org/10.1007/s11033-024-09771-4.
[3]
Murakami S, Kusano Y, Okazaki K, Akaike T, Motohashi H. NRF2 signalling in cytoprotection and metabolism. British Journal of Pharmacology. 2023; 10.1111/bph.16246. https://doi.org/10.1111/bph.16246.
[4]
Rojo de la Vega M, Chapman E, Zhang DD. NRF2 and the Hallmarks of Cancer. Cancer Cell. 2018; 34: 21–43. https://doi.org/10.1016/j.ccell.2018.03.022.
[5]
Sivinski J, Zhang DD, Chapman E. Targeting NRF2 to treat cancer. Seminars in Cancer Biology. 2021; 76: 61–73. https://doi.org/10.1016/j.semcancer.2021.06.003.
[6]
Ibáñez B, Melero A, Montoro A, San Onofre N, Soriano JM. Molecular Insights into Radiation Effects and Protective Mechanisms: A Focus on Cellular Damage and Radioprotectors. Current Issues in Molecular Biology. 2024; 46: 12718–12732. https://doi.org/10.3390/cimb46110755.
[7]
Tao SM, Wang LL, Li MD, Wang J, Gu HM, Zhang LJ. Cancer risk associated with low-dose ionizing radiation: A systematic review of epidemiological and biological evidence. Mutation Research. Reviews in Mutation Research. 2024; 794: 108517. https://doi.org/10.1016/j.mrrev.2024.108517.
[8]
Nowsheen S, Wukovich RL, Aziz K, Kalogerinis PT, Richardson CC, Panayiotidis MI, et al. Accumulation of oxidatively induced clustered DNA lesions in human tumor tissues. Mutation Research. 2009; 674: 131–136. https://doi.org/10.1016/j.mrgentox.2008.09.010.
[9]
Bukowska B, Karwowski BT. The Clustered DNA Lesions - Types, Pathways of Repair and Relevance to Human Health. Current Medicinal Chemistry. 2018; 25: 2722–2735. https://doi.org/10.2174/0929867325666180226110502.
[10]
Valente D, Gentileschi MP, Valenti A, Burgio M, Soddu S, Bruzzaniti V, et al. Cumulative Dose from Recurrent CT Scans: Exploring the DNA Damage Response in Human Non-Transformed Cells. International Journal of Molecular Sciences. 2024; 25: 7064. https://doi.org/10.3390/ijms25137064.
[11]
Gilbert ES. Ionising radiation and cancer risks: what have we learned from epidemiology? International Journal of Radiation Biology. 2009; 85: 467–482. https://doi.org/10.1080/09553000902883836.
[12]
Averbeck D, Candéias S, Chandna S, Foray N, Friedl AA, Haghdoost S, et al. Establishing mechanisms affecting the individual response to ionizing radiation. International Journal of Radiation Biology. 2020; 96: 297–323. https://doi.org/10.1080/09553002.2019.1704908.
[13]
Moi P, Chan K, Asunis I, Cao A, Kan YW. Isolation of NF-E2-related factor 2 (Nrf2), a NF-E2-like basic leucine zipper transcriptional activator that binds to the tandem NF-E2/AP1 repeat of the beta-globin locus control region. Proceedings of the National Academy of Sciences of the United States of America. 1994; 91: 9926–9930. https://doi.org/10.1073/pnas.91.21.9926.
[14]
Tonelli C, Chio IIC, Tuveson DA. Transcriptional Regulation by Nrf2. Antioxidants & Redox Signaling. 2018; 29: 1727–1745. https://doi.org/10.1089/ars.2017.7342.
[15]
Kobayashi EH, Suzuki T, Funayama R, Nagashima T, Hayashi M, Sekine H, et al. Nrf2 suppresses macrophage inflammatory response by blocking proinflammatory cytokine transcription. Nature Communications. 2016; 7: 11624. https://doi.org/10.1038/ncomms11624.
[16]
He F, Antonucci L, Karin M. NRF2 as a regulator of cell metabolism and inflammation in cancer. Carcinogenesis. 2020; 41: 405–416. https://doi.org/10.1093/carcin/bgaa039.
[17]
Li J, Xu C, Liu Q. Roles of NRF2 in DNA damage repair. Cellular Oncology (Dordrecht, Netherlands). 2023; 46: 1577–1593. https://doi.org/10.1007/s13402-023-00834-5.
[18]
Gureev AP, Chernyshova EV, Krutskikh EP, Sadovnikova IS, Tekutskaya EE, Dorohova AA. Role of the Nrf2/ARE Pathway in the mtDNA Reparation. Frontiers in Bioscience (Landmark Edition). 2024; 29: 218. https://doi.org/10.31083/j.fbl2906218.
[19]
Tang Z, Hu B, Zang F, Wang J, Zhang X, Chen H. Nrf2 drives oxidative stress-induced autophagy in nucleus pulposus cells via a Keap1/Nrf2/p62 feedback loop to protect intervertebral disc from degeneration. Cell Death & Disease. 2019; 10: 510. https://doi.org/10.1038/s41419-019-1701-3.
[20]
Niture SK, Jaiswal AK. Nrf2 protein up-regulates antiapoptotic protein Bcl-2 and prevents cellular apoptosis. The Journal of Biological Chemistry. 2012; 287: 9873–9886. https://doi.org/10.1074/jbc.M111.312694.
[21]
Villeneuve NF, Lau A, Zhang DD. Regulation of the Nrf2-Keap1 antioxidant response by the ubiquitin proteasome system: an insight into cullin-ring ubiquitin ligases. Antioxidants & Redox Signaling. 2010; 13: 1699–1712. https://doi.org/10.1089/ars.2010.3211.
[22]
Kobayashi A, Kang MI, Watai Y, Tong KI, Shibata T, Uchida K, et al. Oxidative and electrophilic stresses activate Nrf2 through inhibition of ubiquitination activity of Keap1. Molecular and Cellular Biology. 2006; 26: 221–229. https://doi.org/10.1128/MCB.26.1.221-229.2006.
[23]
Riz I, Hawley TS, Marsal JW, Hawley RG. Noncanonical SQSTM1/p62-Nrf2 pathway activation mediates proteasome inhibitor resistance in multiple myeloma cells via redox, metabolic and translational reprogramming. Oncotarget. 2016; 7: 66360–66385. https://doi.org/10.18632/oncotarget.11960.
[24]
Oyake T, Itoh K, Motohashi H, Hayashi N, Hoshino H, Nishizawa M, et al. Bach proteins belong to a novel family of BTB-basic leucine zipper transcription factors that interact with MafK and regulate transcription through the NF-E2 site. Molecular and Cellular Biology. 1996; 16: 6083–6095. https://doi.org/10.1128/MCB.16.11.6083.
[25]
Dhakshinamoorthy S, Jain AK, Bloom DA, Jaiswal AK. Bach1 competes with Nrf2 leading to negative regulation of the antioxidant response element (ARE)-mediated NAD(P)H:quinone oxidoreductase 1 gene expression and induction in response to antioxidants. The Journal of Biological Chemistry. 2005; 280: 16891–16900. https://doi.org/10.1074/jbc.M500166200.
[26]
Lignitto L, LeBoeuf SE, Homer H, Jiang S, Askenazi M, Karakousi TR, et al. Nrf2 Activation Promotes Lung Cancer Metastasis by Inhibiting the Degradation of Bach1. Cell. 2019; 178: 316–329.e18. https://doi.org/10.1016/j.cell.2019.06.003.
[27]
Hu D, Zhang Z, Luo X, Li S, Jiang J, Zhang J, et al. Transcription factor BACH1 in cancer: roles, mechanisms, and prospects for targeted therapy. Biomarker Research. 2024; 12: 21. https://doi.org/10.1186/s40364-024-00570-4.
[28]
Ahuja M, Kaidery NA, Dutta D, Attucks OC, Kazakov EH, Gazaryan I, et al. Harnessing the Therapeutic Potential of the Nrf2/Bach1 Signaling Pathway in Parkinson’s Disease. Antioxidants (Basel, Switzerland). 2022; 11: 1780. https://doi.org/10.3390/antiox11091780.
[29]
Schmidlin CJ, Shakya A, Dodson M, Chapman E, Zhang DD. The intricacies of NRF2 regulation in cancer. Seminars in Cancer Biology. 2021; 76: 110–119. https://doi.org/10.1016/j.semcancer.2021.05.016.
[30]
Wu S, Lu H, Bai Y. Nrf2 in cancers: A double-edged sword. Cancer Medicine. 2019; 8: 2252–2267. https://doi.org/10.1002/cam4.2101.
[31]
Zimta AA, Cenariu D, Irimie A, Magdo L, Nabavi SM, Atanasov AG, et al. The Role of Nrf2 Activity in Cancer Development and Progression. Cancers. 2019; 11: 1755. https://doi.org/10.3390/cancers11111755.
[32]
Baird L, Dinkova-Kostova AT. The cytoprotective role of the Keap1-Nrf2 pathway. Archives of Toxicology. 2011; 85: 241–272. https://doi.org/10.1007/s00204-011-0674-5.
[33]
Galan-Cobo A, Sitthideatphaiboon P, Qu X, Poteete A, Pisegna MA, Tong P, et al. LKB1 and KEAP1/NRF2 Pathways Cooperatively Promote Metabolic Reprogramming with Enhanced Glutamine Dependence in KRAS-Mutant Lung Adenocarcinoma. Cancer Research. 2019; 79: 3251–3267. https://doi.org/10.1158/0008-5472.CAN-18-3527.
[34]
Cirone M, D’Orazi G. NRF2 in Cancer: Cross-Talk with Oncogenic Pathways and Involvement in Gammaherpesvirus-Driven Carcinogenesis. International Journal of Molecular Sciences. 2022; 24: 595. https://doi.org/10.3390/ijms24010595.
[35]
Gilardini Montani MS, Cecere N, Granato M, Romeo MA, Falcinelli L, Ciciarelli U, et al. Mutant p53, Stabilized by Its Interplay with HSP90, Activates a Positive Feed-Back Loop Between NRF2 and p62 that Induces Chemo-Resistance to Apigenin in Pancreatic Cancer Cells. Cancers. 2019; 11: 703. https://doi.org/10.3390/cancers11050703.
[36]
Vartanian S, Lee J, Klijn C, Gnad F, Bagniewska M, Schaefer G, et al. ERBB3 and IGF1R Signaling Are Required for Nrf2-Dependent Growth in KEAP1-Mutant Lung Cancer. Cancer Research. 2019; 79: 4828–4839. https://doi.org/10.1158/0008-5472.CAN-18-2086.
[37]
Kamble D, Mahajan M, Dhat R, Sitasawad S. Keap1-Nrf2 Pathway Regulates ALDH and Contributes to Radioresistance in Breast Cancer Stem Cells. Cells. 2021; 10: 83. https://doi.org/10.3390/cells10010083.
[38]
Noh JK, Woo SR, Yun M, Lee MK, Kong M, Min S, et al. SOD2- and NRF2-associated Gene Signature to Predict Radioresistance in Head and Neck Cancer. Cancer Genomics & Proteomics. 2021; 18: 675–684. https://doi.org/10.21873/cgp.20289.
[39]
Silva MM, Rocha CRR, Kinker GS, Pelegrini AL, Menck CFM. The balance between NRF2/GSH antioxidant mediated pathway and DNA repair modulates cisplatin resistance in lung cancer cells. Scientific Reports. 2019; 9: 17639. https://doi.org/10.1038/s41598-019-54065-6.
[40]
Purohit V, Wang L, Yang H, Li J, Ney GM, Gumkowski ER, et al. ATDC binds to KEAP1 to drive NRF2-mediated tumorigenesis and chemoresistance in pancreatic cancer. Genes & Development. 2021; 35: 218–233. https://doi.org/10.1101/gad.344184.120.
[41]
Srivastava R, Fernández-Ginés R, Encinar JA, Cuadrado A, Wells G. The current status and future prospects for therapeutic targeting of KEAP1-NRF2 and β-TrCP-NRF2 interactions in cancer chemoresistance. Free Radical Biology & Medicine. 2022; 192: 246–260. https://doi.org/10.1016/j.freeradbiomed.2022.09.023.
[42]
Feng L, Zhao K, Sun L, Yin X, Zhang J, Liu C, et al. SLC7A11 regulated by NRF2 modulates esophageal squamous cell carcinoma radiosensitivity by inhibiting ferroptosis. Journal of Translational Medicine. 2021; 19: 367. https://doi.org/10.1186/s12967-021-03042-7.
[43]
Koppula P, Lei G, Zhang Y, Yan Y, Mao C, Kondiparthi L, et al. A targetable CoQ-FSP1 axis drives ferroptosis- and radiation-resistance in KEAP1 inactive lung cancers. Nature Communications. 2022; 13: 2206. https://doi.org/10.1038/s41467-022-29905-1.
[44]
Matsuoka Y, Yoshida R, Kawahara K, Sakata J, Arita H, Nkashima H, et al. The antioxidative stress regulator Nrf2 potentiates radioresistance of oral squamous cell carcinoma accompanied with metabolic modulation. Laboratory Investigation; a Journal of Technical Methods and Pathology. 2022; 102: 896–907. https://doi.org/10.1038/s41374-022-00776-w.
[45]
Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell. 2012; 149: 1060–1072. https://doi.org/10.1016/j.cell.2012.03.042.
[46]
Chen Y, Jiang Z, Li X. New insights into crosstalk between Nrf2 pathway and ferroptosis in lung disease. Cell Death & Disease. 2024; 15: 841. https://doi.org/10.1038/s41419-024-07224-1.
[47]
Xie W, Tan B, Yang Z, Yu X, Chen L, Ran D, et al. Nrf2/ARE pathway activation is involved in negatively regulating heat-induced apoptosis in non-small cell lung cancer cells. Acta Biochimica et Biophysica Sinica. 2020; 52: 439–445. https://doi.org/10.1093/abbs/gmaa013.
[48]
Chang LC, Fan CW, Tseng WK, Chein HP, Hsieh TY, Chen JR, et al. The Ratio of Hmox1/Nrf2 mRNA Level in the Tumor Tissue Is a Predictor of Distant Metastasis in Colorectal Cancer. Disease Markers. 2016; 2016: 8143465. https://doi.org/10.1155/2016/8143465.
[49]
Wiel C, Le Gal K, Ibrahim MX, Jahangir CA, Kashif M, Yao H, et al. BACH1 Stabilization by Antioxidants Stimulates Lung Cancer Metastasis. Cell. 2019; 178: 330–345.e22. https://doi.org/10.1016/j.cell.2019.06.005.
[50]
Zhou XL, Zhu CY, Wu ZG, Guo X, Zou W. The oncoprotein HBXIP competitively binds KEAP1 to activate NRF2 and enhance breast cancer cell growth and metastasis. Oncogene. 2019; 38: 4028–4046. https://doi.org/10.1038/s41388-019-0698-5.
[51]
Garufi A, Pettinari R, Monteonofrio L, Puliani G, Virdia I, Appetecchia M, et al. NRF2 activation in BON 1 neuroendocrine cancer cells reduces the cytotoxic effects of a novel Ruthenium(II) curcumin compound: A pilot study. Oncology Reports. 2024; 51: 36. https://doi.org/10.3892/or.2024.8695.
[52]
Garufi A, Giorno E, Gilardini Montani MS, Pistritto G, Crispini A, Cirone M, et al. P62/SQSTM1/Keap1/NRF2 Axis Reduces Cancer Cells Death-Sensitivity in Response to Zn(II)-Curcumin Complex. Biomolecules. 2021; 11: 348. https://doi.org/10.3390/biom11030348.
[53]
Garufi A, Baldari S, Pettinari R, Gilardini Montani MS, D’Orazi V, Pistritto G, et al. A ruthenium(II)-curcumin compound modulates NRF2 expression balancing the cancer cell death/survival outcome according to p53 status. Journal of Experimental & Clinical Cancer Research: CR. 2020; 39: 122. https://doi.org/10.1186/s13046-020-01628-5.
[54]
Lin L, Wu Q, Lu F, Lei J, Zhou Y, Liu Y, et al. Nrf2 signaling pathway: current status and potential therapeutic targetable role in human cancers. Frontiers in Oncology. 2023; 13: 1184079. https://doi.org/10.3389/fonc.2023.1184079.
[55]
Talapko J, Talapko D, Katalinić D, Kotris I, Erić I, Belić D, et al. Health Effects of Ionizing Radiation on the Human Body. Medicina (Kaunas, Lithuania). 2024; 60: 653. https://doi.org/10.3390/medicina60040653.
[56]
Huang RX, Zhou PK. DNA damage response signaling pathways and targets for radiotherapy sensitization in cancer. Signal Transduction and Targeted Therapy. 2020; 5: 60. https://doi.org/10.1038/s41392-020-0150-x.
[57]
Pouget JP. Basics of radiobiology. Nuclear Medicine and Molecular Imaging. 2022; 1: 30–51. https://doi.org/10.1016/B978-0-12-822960-6.00137-X.
[58]
Brieger K, Schiavone S, Miller FJ, Jr, Krause KH. Reactive oxygen species: from health to disease. Swiss Medical Weekly. 2012; 142: w13659. https://doi.org/10.4414/smw.2012.13659.
[59]
Deng S, Vlatkovic T, Li M, Zhan T, Veldwijk MR, Herskind C. Targeting the DNA Damage Response and DNA Repair Pathways to Enhance Radiosensitivity in Colorectal Cancer. Cancers. 2022; 14: 4874. https://doi.org/10.3390/cancers14194874.
[60]
Gerelchuluun A, Manabe E, Ishikawa T, Sun L, Itoh K, Sakae T, et al. The major DNA repair pathway after both proton and carbon-ion radiation is NHEJ, but the HR pathway is more relevant in carbon ions. Radiation Research. 2015; 183: 345–356. https://doi.org/10.1667/RR13904.1.
[61]
Mladenov E, Mladenova V, Stuschke M, Iliakis G. New Facets of DNA Double Strand Break Repair: Radiation Dose as Key Determinant of HR versus c-NHEJ Engagement. International Journal of Molecular Sciences. 2023; 24: 14956. https://doi.org/10.3390/ijms241914956.
[62]
Soni A, Murmann-Konda T, Siemann-Loekes M, Pantelias GE, Iliakis G. Chromosome breaks generated by low doses of ionizing radiation in G2-phase are processed exclusively by gene conversion. DNA Repair. 2020; 89: 102828. https://doi.org/10.1016/j.dnarep.2020.102828.
[63]
Maréchal A, Zou L. DNA damage sensing by the ATM and ATR kinases. Cold Spring Harbor Perspectives in Biology. 2013; 5: a012716. https://doi.org/10.1101/cshperspect.a012716.
[64]
Shibata A, Jeggo PA. ATM’s Role in the Repair of DNA Double-Strand Breaks. Genes. 2021; 12: 1370. https://doi.org/10.3390/genes12091370.
[65]
Abuetabh Y, Wu HH, Chai C, Al Yousef H, Persad S, Sergi CM, et al. DNA damage response revisited: the p53 family and its regulators provide endless cancer therapy opportunities. Experimental & Molecular Medicine. 2022; 54: 1658–1669. https://doi.org/10.1038/s12276-022-00863-4.
[66]
Prakash R, Zhang Y, Feng W, Jasin M. Homologous recombination and human health: the roles of BRCA1, BRCA2, and associated proteins. Cold Spring Harbor Perspectives in Biology. 2015; 7: a016600. https://doi.org/10.1101/cshperspect.a016600.
[67]
Furdui CM. Ionizing radiation: mechanisms and therapeutics. Antioxidants & Redox Signaling. 2014; 21: 218–220. https://doi.org/10.1089/ars.2014.5935.
[68]
Buchberger B, Scholl K, Krabbe L, Spiller L, Lux B. Radiation exposure by medical X-ray applications. German Medical Science: GMS E-journal. 2022; 20: Doc06. https://doi.org/10.3205/000308.
[69]
Brenner DJ, Hall EJ. Computed tomography–an increasing source of radiation exposure. The New England Journal of Medicine. 2007; 357: 2277–2284. https://doi.org/10.1056/NEJMra072149.
[70]
Løberg M, Lousdal ML, Bretthauer M, Kalager M. Benefits and harms of mammography screening. Breast Cancer Research: BCR. 2015; 17: 63. https://doi.org/10.1186/s13058-015-0525-z.
[71]
Valotassiou V, Malamitsi J, Papatriantafyllou J, Dardiotis E, Tsougos I, Psimadas D, et al. SPECT and PET imaging in Alzheimer’s disease. Annals of Nuclear Medicine. 2018; 32: 583–593. https://doi.org/10.1007/s12149-018-1292-6.
[72]
Schauer DA, Linton OW. National Council on Radiation Protection and Measurements report shows substantial medical exposure increase. Radiology. 2009; 253: 293–296. https://doi.org/10.1148/radiol.2532090494.
[73]
Pozzessere C, von Garnier C, Beigelman-Aubry C. Radiation Exposure to Low-Dose Computed Tomography for Lung Cancer Screening: Should We Be Concerned? Tomography (Ann Arbor, Mich.). 2023; 9: 166–177. https://doi.org/10.3390/tomography9010015.
[74]
Vinod SK, Hau E. Radiotherapy treatment for lung cancer: Current status and future directions. Respirology (Carlton, Vic.). 2020; 25 Suppl 2: 61–71. https://doi.org/10.1111/resp.13870.
[75]
Xu Y, Trach C, Tessier T, Sinha R, Skarsgard D. Outcomes of patients receiving urgent palliative radiotherapy for advanced lung cancer: an observational study. BMC Palliative Care. 2024; 23: 296. https://doi.org/10.1186/s12904-024-01628-8.
[76]
Hennequin C, Belkacémi Y, Bourgier C, Cowen D, Cutuli B, Fourquet A, et al. Radiotherapy of breast cancer. Cancer Radiotherapie: Journal De La Societe Francaise De Radiotherapie Oncologique. 2022; 26: 221–230. https://doi.org/10.1016/j.canrad.2021.11.013.
[77]
Chino J, Annunziata CM, Beriwal S, Bradfield L, Erickson BA, Fields EC, et al. Radiation Therapy for Cervical Cancer: Executive Summary of an ASTRO Clinical Practice Guideline. Practical Radiation Oncology. 2020; 10: 220–234. https://doi.org/10.1016/j.prro.2020.04.002.
[78]
Gay HA, Michalski JM. Radiation Therapy for Prostate Cancer. Missouri Medicine. 2018; 115: 146–150.
[79]
Häfner MF, Debus J. Radiotherapy for Colorectal Cancer: Current Standards and Future Perspectives. Visceral Medicine. 2016; 32: 172–177. https://doi.org/10.1159/000446486.
[80]
Faiena I, Kim IY, Jang TL. Multimodal treatments for advanced prostate cancer. Oncotarget. 2019; 10: 255–256. https://doi.org/10.18632/oncotarget.26525.
[81]
Borras JM, Lievens Y, Barton M, Corral J, Ferlay J, Bray F, et al. How many new cancer patients in Europe will require radiotherapy by 2025? An ESTRO-HERO analysis. Radiotherapy and Oncology: Journal of the European Society for Therapeutic Radiology and Oncology. 2016; 119: 5–11. https://doi.org/10.1016/j.radonc.2016.02.016.
[82]
Shirai K, Aoki S, Endo M, Takahashi Y, Fukuda Y, Akahane K, et al. Recent developments in the field of radiotherapy for the management of lung cancer. Japanese Journal of Radiology. 2025; 43: 186–199. https://doi.org/10.1007/s11604-024-01663-8.
[83]
Akhunzianov AA, Rozhina EV, Filina YV, Rizvanov AA, Miftakhova RR. Resistance to Radiotherapy in Cancer. Diseases (Basel, Switzerland). 2025; 13: 22. https://doi.org/10.3390/diseases13010022.
[84]
Busato F, Khouzai BE, Mognato M. Biological Mechanisms to Reduce Radioresistance and Increase the Efficacy of Radiotherapy: State of the Art. International Journal of Molecular Sciences. 2022; 23: 10211. https://doi.org/10.3390/ijms231810211.
[85]
Sekhar KR, Freeman ML. Nrf2 promotes survival following exposure to ionizing radiation. Free Radical Biology & Medicine. 2015; 88: 268–274. https://doi.org/10.1016/j.freeradbiomed.2015.04.035.
[86]
Hayes JD, McMahon M. NRF2 and KEAP1 mutations: permanent activation of an adaptive response in cancer. Trends in Biochemical Sciences. 2009; 34: 176–188. https://doi.org/10.1016/j.tibs.2008.12.008.
[87]
Singh A, Boldin-Adamsky S, Thimmulappa RK, Rath SK, Ashush H, Coulter J, et al. RNAi-mediated silencing of nuclear factor erythroid-2-related factor 2 gene expression in non-small cell lung cancer inhibits tumor growth and increases efficacy of chemotherapy. Cancer Research. 2008; 68: 7975–7984. https://doi.org/10.1158/0008-5472.CAN-08-1401.
[88]
McDonald JT, Kim K, Norris AJ, Vlashi E, Phillips TM, Lagadec C, et al. Ionizing radiation activates the Nrf2 antioxidant response. Cancer Research. 2010; 70: 8886–8895. https://doi.org/10.1158/0008-5472.CAN-10-0171.
[89]
Singh A, Bodas M, Wakabayashi N, Bunz F, Biswal S. Gain of Nrf2 function in non-small-cell lung cancer cells confers radioresistance. Antioxidants & Redox Signaling. 2010; 13: 1627–1637. https://doi.org/10.1089/ars.2010.3219.
[90]
Matsagar SV, Singh RK. Protective Effects of NRF2 Activator Sulforaphane in Polyinosinic:Polycytidylic Acid-Induced In Vitro and In Vivo Model. Journal of Biochemical and Molecular Toxicology. 2024; 38: e70086. https://doi.org/10.1002/jbt.70086.
[91]
Mathew ST, Bergström P, Hammarsten O. Repeated Nrf2 stimulation using sulforaphane protects fibroblasts from ionizing radiation. Toxicology and Applied Pharmacology. 2014; 276: 188–194. https://doi.org/10.1016/j.taap.2014.02.013.
[92]
Garufi A, Pistritto G, D’Orazi V, Cirone M, D’Orazi G. The Impact of NRF2 Inhibition on Drug-Induced Colon Cancer Cell Death and p53 Activity: A Pilot Study. Biomolecules. 2022; 12: 461. https://doi.org/10.3390/biom12030461.
[93]
Zhao Q, Mao A, Yan J, Sun C, Di C, Zhou X, et al. Downregulation of Nrf2 promotes radiation-induced apoptosis through Nrf2 mediated Notch signaling in non-small cell lung cancer cells. International Journal of Oncology. 2016; 48: 765–773. https://doi.org/10.3892/ijo.2015.3301.
[94]
Xu Q, Zhang P, Han X, Ren H, Yu W, Hao W, et al. Role of ionizing radiation activated NRF2 in lung cancer radioresistance. International Journal of Biological Macromolecules. 2023; 241: 124476. https://doi.org/10.1016/j.ijbiomac.2023.124476.
[95]
Schaue D, Micewicz ED, Ratikan JA, Iwamoto KS, Vlashi E, McDonald JT, et al. NRF2 Mediates Cellular Resistance to Transformation, Radiation, and Inflammation in Mice. Antioxidants (Basel, Switzerland). 2022; 11: 1649. https://doi.org/10.3390/antiox11091649.
[96]
Qin H, Zhang H, Zhang S, Zhu S, Wang H. Protective Effect of Sirt1 against Radiation-Induced Damage. Radiation Research. 2021; 196: 647–657. https://doi.org/10.1667/RADE-20-00139.1.
[97]
Jayakumar S, Pal D, Sandur SK. Nrf2 facilitates repair of radiation induced DNA damage through homologous recombination repair pathway in a ROS independent manner in cancer cells. Mutation Research. 2015; 779: 33–45. https://doi.org/10.1016/j.mrfmmm.2015.06.007.
[98]
Kim SB, Pandita RK, Eskiocak U, Ly P, Kaisani A, Kumar R, et al. Targeting of Nrf2 induces DNA damage signaling and protects colonic epithelial cells from ionizing radiation. Proceedings of the National Academy of Sciences of the United States of America. 2012; 109: E2949–55. https://doi.org/10.1073/pnas.1207718109.
[99]
Singh B, Chatterjee A, Ronghe AM, Bhat NK, Bhat HK. Antioxidant-mediated up-regulation of OGG1 via NRF2 induction is associated with inhibition of oxidative DNA damage in estrogen-induced breast cancer. BMC Cancer. 2013; 13: 253. https://doi.org/10.1186/1471-2407-13-253.
[100]
Hyun JW, Cheon GJ, Kim HS, Lee YS, Choi EY, Yoon BH, et al. Radiation sensitivity depends on OGG1 activity status in human leukemia cell lines. Free Radical Biology & Medicine. 2002; 32: 212–220. https://doi.org/10.1016/s0891-5849(01)00793-6.
[101]
Sun X, Wang Y, Ji K, Liu Y, Kong Y, Nie S, et al. NRF2 preserves genomic integrity by facilitating ATR activation and G2 cell cycle arrest. Nucleic Acids Research. 2020; 48: 9109–9123. https://doi.org/10.1093/nar/gkaa631.
[102]
Wang Q, Li J, Yang X, Sun H, Gao S, Zhu H, et al. Nrf2 is associated with the regulation of basal transcription activity of the BRCA1 gene. Acta Biochimica et Biophysica Sinica. 2013; 45: 179–187. https://doi.org/10.1093/abbs/gmt001.
[103]
Saha S, Buttari B, Panieri E, Profumo E, Saso L. An Overview of Nrf2 Signaling Pathway and Its Role in Inflammation. Molecules (Basel, Switzerland). 2020; 25: 5474. https://doi.org/10.3390/molecules25225474.
[104]
Kolek V, Vašáková M, Šterclová M, Cwiertka K, Vrána D, Kudláček A, et al. Radiotherapy of Lung Tumours in Idiopathic Pulmonary Fibrosis. Klinicka Onkologie: Casopis Ceske a Slovenske Onkologicke Spolecnosti. 2017; 30: 303–306. https://doi.org/10.14735/amko2017303.
[105]
Yu Z, Xu C, Song B, Zhang S, Chen C, Li C, et al. Tissue fibrosis induced by radiotherapy: current understanding of the molecular mechanisms, diagnosis and therapeutic advances. Journal of Translational Medicine. 2023; 21: 708. https://doi.org/10.1186/s12967-023-04554-0.
[106]
Li X, Zhuang X, Qiao T. Role of ferroptosis in the process of acute radiation-induced lung injury in mice. Biochemical and Biophysical Research Communications. 2019; 519: 240–245. https://doi.org/10.1016/j.bbrc.2019.08.165.
[107]
Li X, Duan L, Yuan S, Zhuang X, Qiao T, He J. Ferroptosis inhibitor alleviates Radiation-induced lung fibrosis (RILF) via down-regulation of TGF-β1. Journal of Inflammation (London, England). 2019; 16: 11. https://doi.org/10.1186/s12950-019-0216-0.
[108]
Zhang Z, Lu M, Chen C, Tong X, Li Y, Yang K, et al. Holo-lactoferrin: the link between ferroptosis and radiotherapy in triple-negative breast cancer. Theranostics. 2021; 11: 3167–3182. https://doi.org/10.7150/thno.52028.
[109]
Wardyn JD, Ponsford AH, Sanderson CM. Dissecting molecular cross-talk between Nrf2 and NF-κB response pathways. Biochemical Society Transactions. 2015; 43: 621–626. https://doi.org/10.1042/BST20150014.
[110]
Chen R, Kang R, Tang D. The mechanism of HMGB1 secretion and release. Experimental & Molecular Medicine. 2022; 54: 91–102. https://doi.org/10.1038/s12276-022-00736-w.
[111]
Anuranjani, Bala M. Concerted action of Nrf2-ARE pathway, MRN complex, HMGB1 and inflammatory cytokines - implication in modification of radiation damage. Redox Biology. 2014; 2: 832–846. https://doi.org/10.1016/j.redox.2014.02.008.
[112]
Jung AR, Kim GE, Kim MY, Ha US, Hong SH, Lee JY, et al. HMGB1 promotes tumor progression and invasion through HMGB1/TNFR1/NF-κB axis in castration-resistant prostate cancer. American Journal of Cancer Research. 2021; 11: 2215–2227.
[113]
Giridhar P, Mallick S, Rath GK, Julka PK. Radiation induced lung injury: prediction, assessment and management. Asian Pacific Journal of Cancer Prevention: APJCP. 2015; 16: 2613–2617. https://doi.org/10.7314/apjcp.2015.16.7.2613.
[114]
Merkt W, Zhou Y, Han H, Lagares D. Myofibroblast fate plasticity in tissue repair and fibrosis: Deactivation, apoptosis, senescence and reprogramming. Wound Repair and Regeneration: Official Publication of the Wound Healing Society [and] the European Tissue Repair Society. 2021; 29: 678–691. https://doi.org/10.1111/wrr.12952.
[115]
Wang Y, Wei J, Deng H, Zheng L, Yang H, Lv X. The Role of Nrf2 in Pulmonary Fibrosis: Molecular Mechanisms and Treatment Approaches. Antioxidants (Basel, Switzerland). 2022; 11: 1685. https://doi.org/10.3390/antiox11091685.
[116]
Hao W, Li M, Cai Q, Wu S, Li X, He Q, et al. Roles of NRF2 in Fibrotic Diseases: From Mechanisms to Therapeutic Approaches. Frontiers in Physiology. 2022; 13: 889792. https://doi.org/10.3389/fphys.2022.889792.
[117]
Traver G, Mont S, Gius D, Lawson WE, Ding GX, Sekhar KR, et al. Loss of Nrf2 promotes alveolar type 2 cell loss in irradiated, fibrotic lung. Free Radical Biology & Medicine. 2017; 112: 578–586. https://doi.org/10.1016/j.freeradbiomed.2017.08.026.
[118]
Artaud-Macari E, Goven D, Brayer S, Hamimi A, Besnard V, Marchal-Somme J, et al. Nuclear factor erythroid 2-related factor 2 nuclear translocation induces myofibroblastic dedifferentiation in idiopathic pulmonary fibrosis. Antioxidants & Redox Signaling. 2013; 18: 66–79. https://doi.org/10.1089/ars.2011.4240.
[119]
Han YY, Gu X, Yang CY, Ji HM, Lan YJ, Bi YQ, et al. Protective effect of dimethyl itaconate against fibroblast-myofibroblast differentiation during pulmonary fibrosis by inhibiting TXNIP. Journal of Cellular Physiology. 2021; 236: 7734–7744. https://doi.org/10.1002/jcp.30456.
[120]
Zhou W, Mo X, Cui W, Zhang Z, Li D, Li L, et al. Nrf2 inhibits epithelial-mesenchymal transition by suppressing snail expression during pulmonary fibrosis. Scientific Reports. 2016; 6: 38646. https://doi.org/10.1038/srep38646.
[121]
Zhang Z, Qu J, Zheng C, Zhang P, Zhou W, Cui W, et al. Nrf2 antioxidant pathway suppresses Numb-mediated epithelial-mesenchymal transition during pulmonary fibrosis. Cell Death & Disease. 2018; 9: 83. https://doi.org/10.1038/s41419-017-0198-x.
[122]
Lang X, Green MD, Wang W, Yu J, Choi JE, Jiang L, et al. Radiotherapy and Immunotherapy Promote Tumoral Lipid Oxidation and Ferroptosis via Synergistic Repression of SLC7A11. Cancer Discovery. 2019; 9: 1673–1685. https://doi.org/10.1158/2159-8290.CD-19-0338.
[123]
Zhu J, Xiong Y, Zhang Y, Wen J, Cai N, Cheng K, et al. The Molecular Mechanisms of Regulating Oxidative Stress-Induced Ferroptosis and Therapeutic Strategy in Tumors. Oxidative Medicine and Cellular Longevity. 2020; 2020: 8810785. https://doi.org/10.1155/2020/8810785.
[124]
Ling H, Xiao H, Luo T, Lin H, Deng J. Role of Ferroptosis in Regulating the Epithelial-Mesenchymal Transition in Pulmonary Fibrosis. Biomedicines. 2023; 11: 163. https://doi.org/10.3390/biomedicines11010163.
[125]
Sun L, He X, Kong J, Yu H, Wang Y. Menstrual blood-derived stem cells exosomal miR-let-7 to ameliorate pulmonary fibrosis through inhibiting ferroptosis by Sp3/HDAC2/Nrf2 signaling pathway. International Immunopharmacology. 2024; 126: 111316. https://doi.org/10.1016/j.intimp.2023.111316.
[126]
Shakya A, McKee NW, Dodson M, Chapman E, Zhang DD. Anti-Ferroptotic Effects of Nrf2: Beyond the Antioxidant Response. Molecules and Cells. 2023; 46: 165–175. https://doi.org/10.14348/molcells.2023.0005.
[127]
Dodson M, Castro-Portuguez R, Zhang DD. NRF2 plays a critical role in mitigating lipid peroxidation and ferroptosis. Redox Biology. 2019; 23: 101107. https://doi.org/10.1016/j.redox.2019.101107.
[128]
Anandhan A, Dodson M, Schmidlin CJ, Liu P, Zhang DD. Breakdown of an Ironclad Defense System: The Critical Role of NRF2 in Mediating Ferroptosis. Cell Chemical Biology. 2020; 27: 436–447. https://doi.org/10.1016/j.chembiol.2020.03.011.
[129]
Alam J, Stewart D, Touchard C, Boinapally S, Choi AM, Cook JL. Nrf2, a Cap’n’Collar transcription factor, regulates induction of the heme oxygenase-1 gene. The Journal of Biological Chemistry. 1999; 274: 26071–26078. https://doi.org/10.1074/jbc.274.37.26071.
[130]
Xue J, Yu C, Sheng W, Zhu W, Luo J, Zhang Q, et al. The Nrf2/GCH1/BH4 Axis Ameliorates Radiation-Induced Skin Injury by Modulating the ROS Cascade. The Journal of Investigative Dermatology. 2017; 137: 2059–2068. https://doi.org/10.1016/j.jid.2017.05.019.
[131]
Zhu K, Zhu X, Liu S, Yu J, Wu S, Hei M. Glycyrrhizin Attenuates Hypoxic-Ischemic Brain Damage by Inhibiting Ferroptosis and Neuroinflammation in Neonatal Rats via the HMGB1/GPX4 Pathway. Oxidative Medicine and Cellular Longevity. 2022; 2022: 8438528. https://doi.org/10.1155/2022/8438528.
[132]
Chan JY, Kwong M. Impaired expression of glutathione synthetic enzyme genes in mice with targeted deletion of the Nrf2 basic-leucine zipper protein. Biochimica et Biophysica Acta. 2000; 1517: 19–26. https://doi.org/10.1016/s0167-4781(00)00238-4.
[133]
Kwak MK, Itoh K, Yamamoto M, Kensler TW. Enhanced expression of the transcription factor Nrf2 by cancer chemopreventive agents: role of antioxidant response element-like sequences in the nrf2 promoter. Molecular and Cellular Biology. 2002; 22: 2883–2892. https://doi.org/10.1128/MCB.22.9.2883-2892.2002.
[134]
Li M, Chiu JF, Kelsen A, Lu SC, Fukagawa NK. Identification and characterization of an Nrf2-mediated ARE upstream of the rat glutamate cysteine ligase catalytic subunit gene (GCLC). Journal of Cellular Biochemistry. 2009; 107: 944–954. https://doi.org/10.1002/jcb.22197.
[135]
Semenza GL. Intratumoral hypoxia, radiation resistance, and HIF-1. Cancer Cell. 2004; 5: 405–406. https://doi.org/10.1016/s1535-6108(04)00118-7.
[136]
Kim TH, Hur EG, Kang SJ, Kim JA, Thapa D, Lee YM, et al. NRF2 blockade suppresses colon tumor angiogenesis by inhibiting hypoxia-induced activation of HIF-1α. Cancer Research. 2011; 71: 2260–2275. https://doi.org/10.1158/0008-5472.CAN-10-3007.
[137]
Pouremamali F, Pouremamali A, Dadashpour M, Soozangar N, Jeddi F. An update of Nrf2 activators and inhibitors in cancer prevention/promotion. Cell Communication and Signaling: CCS. 2022; 20: 100. https://doi.org/10.1186/s12964-022-00906-3.
[138]
Sporn MB, Liby KT. NRF2 and cancer: the good, the bad and the importance of context. Nature Reviews. Cancer. 2012; 12: 564–571. https://doi.org/10.1038/nrc3278.
[139]
Zhang Y, Talalay P, Cho CG, Posner GH. A major inducer of anticarcinogenic protective enzymes from broccoli: isolation and elucidation of structure. Proceedings of the National Academy of Sciences of the United States of America. 1992; 89: 2399–2403. https://doi.org/10.1073/pnas.89.6.2399.
[140]
Hu C, Eggler AL, Mesecar AD, van Breemen RB. Modification of keap1 cysteine residues by sulforaphane. Chemical Research in Toxicology. 2011; 24: 515–521. https://doi.org/10.1021/tx100389r.
[141]
Verma V. Relationship and interactions of curcumin with radiation therapy. World Journal of Clinical Oncology. 2016; 7: 275–283. https://doi.org/10.5306/wjco.v7.i3.275.
[142]
de Waure C, Bertola C, Baccarini G, Chiavarini M, Mancuso C. Exploring the Contribution of Curcumin to Cancer Therapy: A Systematic Review of Randomized Controlled Trials. Pharmaceutics. 2023; 15: 1275. https://doi.org/10.3390/pharmaceutics15041275.
[143]
Li M, Zhang Z, Hill DL, Wang H, Zhang R. Curcumin, a dietary component, has anticancer, chemosensitization, and radiosensitization effects by down-regulating the MDM2 oncogene through the PI3K/mTOR/ETS2 pathway. Cancer Research. 2007; 67: 1988–1996. https://doi.org/10.1158/0008-5472.CAN-06-3066.
[144]
Kunnumakkara AB, Diagaradjane P, Guha S, Deorukhkar A, Shentu S, Aggarwal BB, et al. Curcumin sensitizes human colorectal cancer xenografts in nude mice to gamma-radiation by targeting nuclear factor-kappaB-regulated gene products. Clinical Cancer Research: an Official Journal of the American Association for Cancer Research. 2008; 14: 2128–2136. https://doi.org/10.1158/1078-0432.CCR-07-4722.
[145]
Khor TO, Huang Y, Wu TY, Shu L, Lee J, Kong ANT. Pharmacodynamics of curcumin as DNA hypomethylation agent in restoring the expression of Nrf2 via promoter CpGs demethylation. Biochemical Pharmacology. 2011; 82: 1073–1078. https://doi.org/10.1016/j.bcp.2011.07.065.
[146]
Hong F, Freeman ML, Liebler DC. Identification of sensor cysteines in human Keap1 modified by the cancer chemopreventive agent sulforaphane. Chemical Research in Toxicology. 2005; 18: 1917–1926. https://doi.org/10.1021/tx0502138.
[147]
Bartolini D, Tew KD, Marinelli R, Galli F, Wang GY. Nrf2-modulation by seleno-hormetic agents and its potential for radiation protection. BioFactors (Oxford, England). 2020; 46: 239–245. https://doi.org/10.1002/biof.1578.
[148]
Ren D, Villeneuve NF, Jiang T, Wu T, Lau A, Toppin HA, et al. Brusatol enhances the efficacy of chemotherapy by inhibiting the Nrf2-mediated defense mechanism. Proceedings of the National Academy of Sciences of the United States of America. 2011; 108: 1433–1438. https://doi.org/10.1073/pnas.1014275108.
[149]
Sova M, Saso L. Design and development of Nrf2 modulators for cancer chemoprevention and therapy: a review. Drug Design, Development and Therapy. 2018; 12: 3181–3197. https://doi.org/10.2147/DDDT.S172612.
[150]
Verma AK, Yadav A, Dewangan J, Singh SV, Mishra M, Singh PK, et al. Isoniazid prevents Nrf2 translocation by inhibiting ERK1 phosphorylation and induces oxidative stress and apoptosis. Redox Biology. 2015; 6: 80–92. https://doi.org/10.1016/j.redox.2015.06.020.
[151]
Chian S, Thapa R, Chi Z, Wang XJ, Tang X. Luteolin inhibits the Nrf2 signaling pathway and tumor growth in vivo. Biochemical and Biophysical Research Communications. 2014; 447: 602–608. https://doi.org/10.1016/j.bbrc.2014.04.039.
[152]
Olayanju A, Copple IM, Bryan HK, Edge GT, Sison RL, Wong MW, et al. Brusatol provokes a rapid and transient inhibition of Nrf2 signaling and sensitizes mammalian cells to chemical toxicity-implications for therapeutic targeting of Nrf2. Free Radical Biology & Medicine. 2015; 78: 202–212. https://doi.org/10.1016/j.freeradbiomed.2014.11.003.
[153]
Cai SJ, Liu Y, Han S, Yang C. Brusatol, an NRF2 inhibitor for future cancer therapeutic. Cell & Bioscience. 2019; 9: 45. https://doi.org/10.1186/s13578-019-0309-8.
[154]
Tao S, Wang S, Moghaddam SJ, Ooi A, Chapman E, Wong PK, et al. Oncogenic KRAS confers chemoresistance by upregulating NRF2. Cancer Research. 2014; 74: 7430–7441. https://doi.org/10.1158/0008-5472.CAN-14-1439.
[155]
Chae YK, Arya A, Malecek MK, Shin DS, Carneiro B, Chandra S, et al. Repurposing metformin for cancer treatment: current clinical studies. Oncotarget. 2016; 7: 40767–40780. https://doi.org/10.18632/oncotarget.8194.
[156]
Do MT, Kim HG, Khanal T, Choi JH, Kim DH, Jeong TC, et al. Metformin inhibits heme oxygenase-1 expression in cancer cells through inactivation of Raf-ERK-Nrf2 signaling and AMPK-independent pathways. Toxicology and Applied Pharmacology. 2013; 271: 229–238. https://doi.org/10.1016/j.taap.2013.05.010.
[157]
Milkovic L, Zarkovic N, Saso L. Controversy about pharmacological modulation of Nrf2 for cancer therapy. Redox Biology. 2017; 12: 727–732. https://doi.org/10.1016/j.redox.2017.04.013.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top