Abstract
Nuclear factor erythroid 2-related factor 2 (NRF2) is a basic leucine zipper protein that participates in a complex regulatory network in the body. The activation of NRF2 can prevent and treat colorectal cancer (CRC). A variety of natural compounds can activate NRF2 to inhibit oxidative stress and inflammation to prevent the occurrence and development of CRC, inhibit the proliferation of CRC cells and induce their apoptosis. However, some studies have also shown that it also has negative effects on CRC, such as overexpression of NRF2 can promote the growth of colorectal tumors and increase the drug resistance of chemotherapeutic drugs such as 5-fluorouracil and oxaliplatin. Therefore, inhibition of NRF2 can also be helpful in the treatment of CRC. In this study, we analyze the current research progress of NRF2 in CRC from various aspects to provide new ideas for prevention and treatment based on the NRF2 signaling pathway in CRC.
Keywords: nuclear factor E2-related factor 2, colorectal cancer, prevention, treatment, inflammatory-associated colorectal cancer
Introduction
Colorectal cancer (CRC) is a common malignant tumor, with the third morbidity and the second mortality in the world, and the incidence is increasing year by year.1 The occurrence of CRC leads to a reduced quality of life and significantly increases the risk of death, making CRC prevention and treatment extremely important. Nuclear factor E2-related factor 2 (NFE2L2/NRF2) is a basic leucine zipper protein that regulates the expression of antioxidant proteins. It is involved in a complex regulatory network and plays multiple roles in the regulation of metabolism, inflammation, autophagy, proteostasis, mitochondrial physiology, and immune responses.2 Under normal conditions, the activation of NRF2 can prevent the development of CRC, including reducing the proliferation of colon cancer cells and increasing the apoptosis of colon cancer cells. However, overexpression of NRF2 may also have negative effects, by creating the best environment for cell growth to promote the survival of normal cells and cancer cells, protect tumor cells from oxidative stress, chemotherapeutic drugs, and radiotherapy, and promote tumor development.3 In this paper, we summarize the complexity of NRF2 regulation in CRC and clarify the significance of NRF2 in CRC prevention and treatment through different perspectives.
Structure and Function of NRF2
NRF2 is encoded by the NFE2L2 gene and belongs to the Cap’n’Collar subfamily of basic leucine zipper (bZIP) transcription factors, which contains nuclear factor 2 (erythroid-derived 2 NFE2) and NRF1, NRF2, and NRF3.4 NRF2 has seven conserved NRF2-ECH (Neh) homologous domains and has different functions to control NRF2 transcriptional activity. BZIP in the Neh1 domain is heterodimerized with small Musculo aponeurotic fibrosarcoma (sMaf) oncogene homolog K, G, F, and other bZIP proteins to identify antioxidant response elements (AREs) to activate gene transcription, while the Neh2 domain contains ETGE and DLG motifs, which specifically interact with the Kelch domain of Kelch-like ECH-related protein 1 (Kelch-like ECH associated protein, Keap1) to ubiquitinate and degrade NRF2.5 The Neh3-5 structural domain functions as a transcriptional activation structural domain by binding to various components of the transcriptional machinery.6 The Neh6 structural domain contains 2 redox-independent degraders, DSGIS and DSAPGS, which bind to the E3 ubiquitin ligase β-transducing repeat protein and mediates the degradation of NRF2 in oxidatively stressed cells.7 The Neh7 structural domain mediates the interaction with the Recombinant Retinoid X Receptor Alpha (RXRa), which inhibits NRF2 activity.8
Under normal conditions, NRF2 binds to Keap1 through its 2 motifs (ETGE and DLG). Keap1 promotes the ubiquitination of NRF2 in the cytoplasm and keeps it at a low level in the cytoplasm.9 When exposed to oxidative stress or using an NRF2 activator, NRF2 is dissociated from Keap1 binding and then translocated to the nucleus, where it transcribes with sMaf protein polymer and ARE gene to activate the expression of multiple genes10 (Figure 1). These include glutathione S-transferase (GST), NAD(P)H dehydrogenase [quinone] 1 (NQO1), heme oxygenase-1 (HO-1), glutathione peroxidase (GSH-Px), and superoxide dismutase (SOD).11
Figure 1.
Under normal conditions, NRF2 binds to Keap1 through its two motifs (ETGE and DLG), and Keap1 promotes the ubiquitination of NRF2 in the cytoplasm. Under oxidative stress, NRF2 dissociates from Keap1 and translocates to the nucleus, which activates the expression of many genes with sMaf protein polymer and ARE gene transcription.
Abbreviations: NRF2, nuclear factor erythroid 2-related factor 2; ARE, antioxidant response element.
The Role of NRF2 in CRC
Studies have shown that long-term chronic inflammation leads to increased levels of pro-inflammatory cytokines in the colonic mucosa, such as tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6), etc, thereby promoting inflammation and cancer.12 IL-6 promotes tumor growth and inhibits apoptosis by activating the JAK/STAT signaling pathway. IL-1β and TNF-α cause cyclooxygenase-2 (COX-2) to be overexpressed in the early stages of carcinogenesis and also lead to angiogenesis, cell proliferation, and apoptosis. Pro-inflammatory cytokines are also able to promote the production of reactive oxygen species (ROS) and reactive nitrogen intermediates, both of which cause DNA damage by way of oxidative stress, thereby causing the development of cancer.13 In addition, after stimulation by TNF-α, the expression of metastasis-associated in colon cancer 1 (MACC1), which induces cancer cell proliferation and increases metastasis, is significantly increased at both gene and protein levels.14 Therefore, controlling inflammation is an effective means to inhibit the occurrence and development of CRC.
Nuclear factor kappa-B (NF-κB) is a central regulator of inflammation that regulates a variety of biological processes. Upon inflammation, NF-κB is activated, leading to the transcription of various cytokines such as COX-2, TNF-α, IL-1β, and IL-6.15 TNF-α and IL-1β stimulate matrix metalloproteinase (MMP) production, leading to the extracellular matrix and mucosal damage. In colonic lamina propria fibroblasts, activation of NF-κB can lead to increased expression of IL-8, IL-6, and chemokines, which are attracted by neutrophils to sites of inflammation and play a key role in their development, promoting tumor development through tissue infiltration and metastasis.16 Tumor cell growth requires the continuous formation of new blood vessels, and IL-6 and TNF-α has been shown to promote angiogenesis. Persistent activation of NF-κB also inhibits the transcription of apoptosis-related genes such as Cellular inhibitors of apoptosis (cIAPs), Caspase-8/FADD-like IL-1beta-converting enzyme inhibitory protein (c-FLIP) and members of the bcl2 family (eg A1/BFL1 and bcl-xl).17 In conclusion, activation of NF-κB promotes tumorigenesis by promoting cell proliferation and angiogenesis, inhibiting apoptosis, and promoting cell invasion and metastasis.
Therefore, preventing the activation of NF-κB and the expression of downstream cytokines is an effective means to resist the occurrence and development of tumors. The activation of NRF2 leads to the expression of HO-1, and the upregulation of HO-1 expression decreased the release of proinflammatory cytokines such as TNF-α, IL-6, etc.18 HO-1 converts heme into biliverdin and CO, which play an important role in cytoprotection against oxidative stress.19 Low concentration of CO can attenuate NF-κB p65 nuclear translocation.20 Thus reducing the release of proinflammatory cytokines. Therefore, inflammation induced by oxidative stress leads to further activation of NF-κB and excessive production of cytokines, and the activation of NRF2 plays an important role in interrupting this cycle (Figure 2).21
Figure 2.
When stimulated by oxidative stress, NRF2 transcripts to activate the expression of HO-1, and HO-1 can inhibit TNF-α and IL-6. And it can induce heme decomposition to produce CO, which can further inhibit the production of TNF-α and IL-6 by NF-κB, and finally prevent the occurrence and development of CRC.
Abbreviations: NRF2, nuclear factor erythroid 2-related factor 2; CRC, colorectal cancer.
In AOM/DSS-induced colorectal cancer mice, NRF2 knockout mice were more likely to develop colorectal cancer than wild-type mice. The main factors for this increased susceptibility were impaired antioxidant/detoxification mechanisms and increased pro-inflammatory arachidonic acid metabolism, and the lack of NRF2 significantly reduced the diversity of colon cancer gene expression.22,23 Lee et al found that the activation of the NRF2 signaling pathway by CyCl can reduce proliferation and colony formation in colon cancer cells by inhibiting the NF-κB signaling pathway and inducing apoptosis.24 Shukla et al25 indicated that AR inhibitor prevents CRC growth by increasing mitochondrial biogenesis via increasing the expression of NRF2/HO-1/AMPK/p53 and decreasing the mitochondrial DNA damage. Activation of NRF2 also affects matrix metalloproteinase (MMP). The NRF2/HO-1 axis inhibits MMP-7 in human intestinal epithelial cells. Expression of MMP-3 induced by oxidative stress is elevated in NRF2-deficient mice. In contrast, MMP-3 levels were also elevated in patients with inflammatory bowel disease, which may suggest that NRF2 has an inhibitory effect on MMP-3, resulting in a therapeutic effect on inflammatory bowel disease.26 In addition, NRF2 induces the expression of biphasic enzymes and reduces the risk of tissue carcinogenesis. Higher NRF2 levels were positively correlated with antioxidant enzymes such as glutathione S-transferase A4 (GST-A4) and peroxiredoxin protein-1 (PRDX1), and with the Interleukin-17A (IL-17A) was negatively correlated.27 In summary, the development of CRC can be inhibited by activating NRF2. In recent years, most studies have shown that some natural compounds can treat and prevent CRC by activating NRF2 (Table 1).
Table 1.
Natural Compounds in the Prevention and Treatment of Colorectal Cancer.
| Chemical compound | Cell types/animal model | Result | Reference |
|---|---|---|---|
| Luteolin | AOM/DSS mouse model HCT116 cells HT29 cells |
luteolin upregulated NRF2 and UDP-GT and GST levels and increased GST-a and GST-μ expression, thereby inhibiting tumorigenic development during AOM/DSS-induced CRC. It also inhibited the proliferation and transformation of HCT116 cells and HT29 cells by increasing the expression of NRF2. | 28,29 |
| Sulforaphane (SFN) | HT-29 cells SW480 cells |
SFN increases nuclear translocation of NRF2, inhibits cell proliferation and colony formation, and induces apoptosis in colon cancer HT-29 and SW480 cells. | 30 |
| Shaoyao decoction (SYD) | AOM/DSS mouse model HT-29 cells |
By activating the NRF2 pathway, it can up-regulate the expression of mRNA and proteins such as NRF2 and its downstream HO-1, NQO-1 in vivo and in vitro, effectively enhance the antioxidant capacity of the body, and reduce the expression of inflammatory factors such as NF-κB, TNF-α, and IL-1β, so as to prevent the occurrence of colitis-associated CRC. | 31 |
| Ginnalin A | HCT116 cells SW480 cells SW620 cells |
Ginnalin A showed a good inhibitory effect on CRC cells (HCT116, SW480, and SW620), significantly reducing cell proliferation. Ginnalin A inhibited tumor proliferation by inducing cell cycle arrest in the S phase. And it upregulated the expression levels of mRNA and protein of NRF2-associated antioxidant genes HO-1 and NQO1. | 32 |
| Digitoflavone | AOM/DSS mouse model Caco-2 cells |
Digitalis ketones induce the G2 phase cell cycle arrest, inhibit angiogenesis and down-regulate NF-κB expression, and increase NRF2 expression, nuclear translocation, and antioxidant enzyme expression. In addition, digitalis ketones reduce H2O2-induced oxidative stress and cell death through the p38 MAPK-NTF2/ARE pathway. In vivo studies showed that 50 mg/kg of digitalis significantly reduced the incidence, number, and size of AOM-DSS-induced tumors. | 33 |
| Tagitinin C | HCT116 cells | Tagitinin C inhibits the growth of CRC cells including HCT116 cells and induced an oxidative cellular microenvironment resulting in ferroptosis of HCT116 cells. Tagitinin C-induced ferroptosis was accompanied by the attenuation of glutathione (GSH) levels and an increased in lipid peroxidation. Mechanistically, tagitinin C induced endoplasmic reticulum (ER) stress and oxidative stress, thus activating nuclear translocation of NRF2. HO-1 expression increased significantly with the treatment of tagitinin C. Upregulated HO-1 led to an increase in the labile iron pool, which promoted lipid peroxidation, meanwhile tagitinin C showed a synergistic anti-tumor effect together with erastin. | 34 |
| Wogonin | AOM/DSS mouse model HTC116 cells |
Wogonin inhibits the expression of NF-κB, IL-6, and IL-1, promotes the expression of NRF2, reduces the incidence of inflammatory-related colorectal cancer tumors, and inhibits the growth of HCT116 cells. | 35 |
| Cylon cinnamon | AOM/DSS mouse model | CA increases the expression of NRF2 in mouse colonic epithelial cells, thereby inhibiting colorectal carcinogenesis in the AOM/DSS mouse model. | 36 |
| Quercetin | Rats with DMH induced colon carcinogenesis. | quercetin supplementation effectively reversed DMH-mediated oxidative stress and DNA damage by targeting the NRF2/Keap1 signaling pathway. | 37 |
Negative Role of NRF2 in CRC
However, studies have found that NRF2 can also promote cancer. Tao et al38 treated mice with NRF2 regulators to test the role of NRF2 in cancer. The results showed that the activation of NRF2 could prevent the occurrence of chemically induced cancer. However, whether caused by chemical or genetic cancer, can promote the progression of existing tumors. If the tumor has already occurred, the continued development of the tumor can be prevented by inhibiting NRF2. High expression of human telomerase reverse transcriptase (hTERT) is associated with the progression of CRC and negatively correlates with survival in CRC patients. Previous studies have shown that hTERT reduces ROS levels in cancer cells and accelerates cancer progression. hTERT promotes CRC proliferation and migration through upregulation of NRF2 expression by recruiting the transcription factor YBX1 to activate the NRF2 promoter.39 Evans et al40 applied tissue microarrays and found that, relative to normal colon tissue, NRF2 in primary CRC and metastatic cancer tissues were highly expressed (P < 0.01), and NRF2 expression in matched primary and metastatic specimens was positively correlated. In vitro experiments showed that NRF2 siRNA and NRF2 inhibitor Brusatol reduced cell survival and sensitized cells by irinotecan. In addition, Brusatol was effective in inhibiting CRC development in subcutaneous and allograft CRC mice. Studies by Torrente et al41 have shown that NRF2 is overactivated in colorectal tumors, which is associated with poor prognosis. These studies have shown that in existing CRC, the activation of NRF2 promotes the development of cancer.
The activation of NRF2 is associated with resistance to chemotherapy. 5-Fluorouracil is the standard drug for CRC chemotherapy. Zhao et al42 found that resistance to 5-fluorouracil treatment was associated with higher ROS production, upregulation of NRF2, and increased expression of HO-1 in CRC cell lines. Programmed Cell Death Receptor-Ligand 1 (PD-L1) is one of the downstream targets of the NRF2. This molecule has some beneficial effects on tumor growth by suppressing the immune system. Payandeh et al43 examined the expression of NRF2, PD-L1, and CD80 in tumors and cut edge tissues of CRC patients. The role of the NRF2-PD-L1 axis in promoting oxaliplatin resistance was investigated. The results showed that NRF2 and PD-L1 mRNA expression was significantly higher in tumor tissues than in tangential margin tissues. PD-L1mRNA expression levels were also elevated in drug-resistant cells. While NRF2 expression was decreased in SW480/RES cells, NRF2 expression was increased in LS174T/RES cells. Inhibition of NRF2 by siRNA treatment in SW480/RES and LS174T/RES cells decreased the IC50 values of oxaliplatin. Inhibition of NRF2 caused a significant increase in oxaliplatin-induced apoptosis and a decrease in migration in SW480/RES cells. Effective inhibition of the NRF2-PD-L1 signaling pathway could be a new way to improve the efficacy of oxaliplatin in the treatment of CRC. Therefore, in the process of traditional chemotherapy, inhibition of NRF2 is an effective means to reduce drug resistance, and many studies have shown its effectiveness (Table 2).
Table 2.
Different Types of Compounds/Genes Reduce Chemotherapeutic Drug Resistance by Inhibiting NRF2 in CRC.
| Compound/gene | Result | Reference |
|---|---|---|
| FoxO3 | FoxO3 can reverse CRC resistance to 5-FU by inhibiting the NRF2/TR1 signaling pathway and increasing intracellular levels of reactive oxygen species | 44 |
| Quinacrine(QC) | Treatment of mouse CRC xenografts with a combination of QC and 5-FU inhibited tumor growth more effectively than QC or 5-FU alone.QC enhances the sensitivity of CRC cells to 5-FU under hypoxic conditions by enhancing JNK1-dependent degradation of NRF2. | 45 |
| Trigonelline | Trigonelline inhibits NRF2 and ARE expression in CRC cells, and trigonelline micelles increase oxaliplatin-induced apoptosis in an NRF2/ARE-dependent manner. | 46 |
| RV-59 | A nude mouse xenograft tumor model showed that RV-59 efficiently suppressed tumor growth induced by transplanted NLS-mutated NRF2-transfected shNRF2-HCT116 stable clones without affecting the bodyweight of the nude mice over the 37-day experimental period. | 47 |
| Manuka honey (MH) | Downregulation of transcription factor (NF-κB and NRF2) and antioxidant enzyme activity (SOD, catalase, glutathione peroxidase, and glutathione reductase) and expression (SOD, catalase, and HO-1) were more evident after the combined treatment, leading to more cell death by oxidative stress. | 48 |
| Dihydromyricetin (DMY) | DMY restored chemosensitivity (OXA and VCR) by inhibiting both MRP2 expression and its promoter activity in HCT116/OXA and HCT8/VCR cell lines. Furthermore, DMY could inhibit NF-κB/p65 expression, reducing NF-κB/p65 translocation to the nucleus to silence NRF2 signaling, which is necessary for MRP2 expression. | 49 |
Discussion
NRF2 participates in various physiological processes and in complex regulatory mechanisms in cancer and plays a key role in intestinal physiological function and the prevention and treatment of CRC. In recent years, NRF2 has received more and more attention. In inflammation-related CRC, a large number of studies have shown that the expression of NRF2 and the activation of downstream genes can protect intestinal cells from oxidative stress, inflammation, and other damage, and many studies have proved that lack of NRF2 is more likely to suffer from CRC. Therefore, activating NRF2 is effective in preventing CRC. As activators of NRF2, many natural compounds can effectively prevent the occurrence and development of inflammation-related colorectal cancer. In a word, the activation of NRF2 is very important for the prevention of CRC, and further study on the preventive effect of activating NRF2 on CRC should be paid attention to in the future.
On the contrary, the negative role of NRF2 in CRC should not be ignored, because the expression of NRF2 can lead to drug resistance to some chemotherapeutic drugs, when using traditional chemotherapy to treat CRC, we should also inhibit NRF2 to reduce drug resistance to chemotherapy to improve the efficacy of chemotherapy. The overexpression of NRF2 in colorectal cancer may also promote the proliferation and migration of CRC. Inhibition of its overexpression in CRC may be effective in the treatment of CRC. However, there are few studies on NRF2 inhibitors in the treatment of CRC, so more studies on NRF2 inhibitors in CRC are needed in the future to clarify the role of NRF2 in CRC.
Abbreviations
- CRC
colorectal cancer
- NRF2/NFE2L2
nuclear factor E2-related factor 2
- bZIP
basic leucine zipper
- sMaf
small musculoaponeurotic fibrosarcoma
- ARE
antioxidant response element
- Keap1
Kelch-like ECH associated protein
- RXRa
recombinant retinoid X receptor alpha
- NQO1
NAD(P)H dehydrogenase [quinone] 1
- GST
glutathione S-transferase
- HO-1
heme oxygenase-1
- GSH-Px
glutathione peroxidase
- SOD
superoxide dismutase
- TNF-α
tumor necrosis factor-α
- IL-1β
interleukin-1β
- IL-6
interleukin-6
- COX-2
cyclooxygenase-2
- ROS
reactive oxygen species
- MACC1
metastasis-associated in colon cancer 1
- NF-κB
nuclear factor kappa-B
- MMP
matrix metalloproteinase
- cIAPs
cellular inhibitors of apoptosis
- c-FLIP
caspase-8/FADD-like IL-1beta-converting enzyme inhibitory protein
- FADD
FAS-associated death domain
- GST-A4
glutathione S-transferase A4
- PRDX1
peroxiredoxin protein-1
- IL-17A
Interleukin-17A
- hTERT
human telomerase reverse transcriptase
- PD-L1
programmed cell death receptor-ligand 1
Footnotes
The authors declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Funding: The authors disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This work was funded by grants from the Medical School of Facial Features of Hubei University of Science and Technology (No. 2020XZ35), the Xianning Municipal Science and Technology Plan Project (No. 2021ZRKX025), and the innovation and Entrepreneurship Project for College Students in Hubei Province (S202110927026).
ORCID iD: Jiaxiang Li https://orcid.org/0000-0002-8769-0440
References
- 1.Sung H, Ferlay J, Siegel R, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209‐249. doi: 10.3322/caac.21660. [DOI] [PubMed] [Google Scholar]
- 2.He F, Ru X, Wen T. NRF2, A transcription factor for stress response and beyond. Int J Mol Sci. 2020;21(13):4777. doi: 10.3390/ijms21134777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Gonzalez-Donquiles C, Alonso-Molero J, Fernandez-Villa T, Vilorio-Marqués L, Molina A, Martín V. The NRF2 transcription factor plays a dual role in colorectal cancer: a systematic review. PLoS One. 2017;12(5):e0177549. doi: 10.1371/journal.pone.0177549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Jaramillo M, Zhang D. The emerging role of the NRF2-Keap1 signaling pathway in cancer. Genes Dev. 2013;27(20):2179‐2191. doi: 10.1101/gad.225680.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Taguchi K, Yamamoto M. The KEAP1-NRF2 system in cancer. Front Oncol. 2017;7:80. doi: 10.3389/fonc.2017.00085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Canning P, Sorrell F, Bullock A. Structural basis of Keap1 interactions with NRF2. Free Radical Biol Med. 2015;88:101‐107. doi: 10.1016/j.freeradbiomed.2015.05.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.McMahon M, Thomas N, Itoh K, Yamamoto M, Hayes J. Redox-regulated turnover of NRF2 is determined by at least two separate protein domains, the redox-sensitive Neh2 degron and the redox-insensitive Neh6 degron. J Biol Chem. 2004;279(30):31556‐31567. doi: 10.1074/jbc.m403061200. [DOI] [PubMed] [Google Scholar]
- 8.Wang H, Liu K, Geng M, et al. RXRα inhibits the NRF2-are signaling pathway through a direct interaction with the Neh7 domain of NRF2. Cancer Res. 2013;73(10):3097‐3108. doi: 10.1158/0008-5472.can-12-3386. [DOI] [PubMed] [Google Scholar]
- 9.Tong K, Katoh Y, Kusunoki H, Itoh K, Tanaka T, Yamamoto M. Keap1 recruits Neh2 through binding to ETGE and DLG motifs: characterization of the two-site molecular recognition model. Mol Cell Biol. 2006;26(8):2887‐2900. doi: 10.1128/mcb.26.8.2887-2900.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Itoh K, Chiba T, Takahashi S, et al. An NRF2/small maf heterodimer mediates the induction of phase II detoxifying enzyme genes through antioxidant response elements. Biochem Biophys Res Commun. 1997;236(2):313‐322. doi: 10.1006/bbrc.1997.6943. [DOI] [PubMed] [Google Scholar]
- 11.Niture S, Khatri R, Jaiswal A. Regulation of NRF2—an update. Free Radical Biol Med. 2014;66:36‐44. doi: 10.1016/j.freeradbiomed.2013.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Muthusami S, Ramachandran I, Babu K, et al. Role of inflammation in the development of colorectal cancer. Endocrine, Metabolic & Immune Disorders – Drug Targets. 2021;21(1):77‐90. doi: 10.2174/1871530320666200909092908. [DOI] [PubMed] [Google Scholar]
- 13.Lopez A, Pouillon L, Beaugerie L, Danese S, Peyrin-Biroulet L. Colorectal cancer prevention in patients with ulcerative colitis. Best Practice & Research Clinical Gastroenterology. 2018;32–33:103‐109. doi: 10.1016/j.bpg.2018.05.010. [DOI] [PubMed] [Google Scholar]
- 14.Kobelt D, Zhang C, Clayton-Lucey I, et al. Pro-inflammatory TNF-α and IFN-γ promote tumor growth and metastasis via induction of MACC1. Front Immunol. 2020;11. doi: 10.3389/fimmu.2020.00980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.McDaniel D, Eden K, Ringel V, Allen I. Emerging roles for noncanonical NF-κB signaling in the modulation of inflammatory bowel disease pathobiology. Inflamm Bowel Dis. 2016;22(9):2265‐2279. doi: 10.1097/mib.0000000000000858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Azmy Nabeh O, Ishak Attallah M, El-Sayed El-Gawhary N. The pivotal relation between glucagon-like peptides, NF-κB and inflammatory bowel disease. Clin Exp Pharmacol Physiol. 2020. doi: 10.1111/1440-1681.13361 [DOI] [PubMed] [Google Scholar]
- 17.Park M, Hong J. Roles of NF-κB in cancer and inflammatory diseases and their therapeutic approaches. Cells. 2016;5(2):15. doi: 10.3390/cells5020015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Lee S, Ju I, Choi Y, Park S, Oh M. Trichosanthis semen suppresses lipopolysaccharide-induced neuroinflammation by regulating the NF-κB signaling pathway and HO-1 expression in microglia. Toxins (Basel). 2021;13(12):898. doi: 10.3390/toxins13120898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Parfenova H, Leffler CW, Basuroy S, Fedinec AL. Antioxidant roles of heme oxygenase, carbon monoxide and bilirubin in cerebral circulation during epileptic seizures. J Cereb Blood Flow Metab. 2012;32(6):1024-1034. doi: 10.1096/fasebj.26.1_supplement.685.24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Huang Y, Ma T, Ye Z, et al. Carbon monoxide (CO) inhibits hydrogen peroxide (H2O2)-induced oxidative stress and the activation of NF-κB signaling in lens epithelial cells. Exp Eye Res. 2018;166:29‐39. doi: 10.1016/j.exer.2017.08.016. [DOI] [PubMed] [Google Scholar]
- 21.Ahmed S, Luo L, Namani A, Wang X, Tang X. NRF2 signaling pathway: pivotal roles in inflammation. Biochimica et Biophysica Acta (BBA) – Molecular Basis of Disease. 2017;1863(2):585‐597. doi: 10.1016/j.bbadis.2016.11.005. [DOI] [PubMed] [Google Scholar]
- 22.Khor T, Huang M, Prawan A, et al. Increased susceptibility of NRF2 knockout mice to colitis-associated colorectal cancer. Cancer Prevention Research. 2008;1(3):187‐191. doi: 10.1158/1940-6207.capr-08-0028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Hammad A, Zheng Z, Gao Y, Namani A, Shi H, Tang X. Identification of novel NRF2 target genes as prognostic biomarkers in colitis-associated colorectal cancer in NRF2-deficient mice. Life Sci. 2019;238:116968. doi: 10.1016/j.lfs.2019.116968. [DOI] [PubMed] [Google Scholar]
- 24.Lee D, Yun S, Song M, Jung K, Kim E. Cyanidin chloride induces apoptosis by inhibiting NF-κB signaling through activation of NRF2 in colorectal cancer cells. Antioxidants. 2020;9(4):285. doi: 10.3390/antiox9040285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Shukla K, Sonowal H, Saxena A, Ramana K, Srivastava S. Aldose reductase inhibitor, fidarestat regulates mitochondrial biogenesis via NRF2/HO-1/AMPK pathway in colon cancer cells. Cancer Lett. 2017;411:57‐63. doi: 10.1016/j.canlet.2017.09.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Piotrowska M, Swierczynski M, Fichna J, Piechota-Polanczyk A. The NRF2 in the pathophysiology of the intestine: molecular mechanisms and therapeutic implications for inflammatory bowel diseases. Pharmacol Res. 2021;163:105243. doi: 10.1016/j.phrs.2020.105243. [DOI] [PubMed] [Google Scholar]
- 27.Sabzevary-Ghahfarokhi M, Shohan M, Shirzad H, et al. The regulatory role of NRF2 in antioxidants phase2 enzymes and IL-17A expression in patients with ulcerative colitis. Pathology – Research and Practice. 2018;214(8):1149‐1155. doi: 10.1016/j.prp.2018.06.001. [DOI] [PubMed] [Google Scholar]
- 28.Pandurangan A, Ananda Sadagopan S, Dharmalingam P, Ganapasam S. Luteolin, a bioflavonoid inhibits azoxymethane-induced colorectal cancer through activation of NRF2 signaling. Toxicol Mech Methods. 2013;24(1):13‐20. doi: 10.3109/15376516.2013.843111. [DOI] [PubMed] [Google Scholar]
- 29.Zuo Q, Wu R, Xiao X, et al. The dietary flavone luteolin epigenetically activates the NRF2 pathway and blocks cell transformation in human colorectal cancer HCT116 cells. J Cell Biochem. 2018;119(11):9573‐9582. doi: 10.1002/jcb.27275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Hao Q, Wang M, Sun NX, et al. Sulforaphane suppresses carcinogenesis of colorectal cancer through the ERK/NRF2-UDP glucuronosyltransferase 1A metabolic axis activation. Oncol Rep. 2020;43(4):1067-1080. doi: 10.3892/or.2020.7495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Wang X, Saud S, Wang F, et al. Protective effect of ShaoYao decoction on colitis-associated colorectal cancer by inducing NRF2 signaling pathway. J Ethnopharmacol. 2020;252:112600. doi: 10.1016/j.jep.2020.112600. [DOI] [PubMed] [Google Scholar]
- 32.Bi W, He C, Li X, et al. Ginnalin A from Kujin tea (Acer tataricum subsp. ginnala) exhibits a colorectal cancer chemoprevention effect via activation of the NRF2/HO-1 signaling pathway. Food Funct. 2018;9(5):2809‐2819. doi: 10.1039/c8fo00054a. [DOI] [PubMed] [Google Scholar]
- 33.Yang Y, Cai X, Yang J, et al. Chemoprevention of dietary digitoflavone on colitis-associated colon tumorigenesis through inducing NRF2 signaling pathway and inhibition of inflammation. Mol Cancer. 2014;13(1). doi: 10.1186/1476-4598-13-48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Wei R, Zhao Y, Wang J, et al. Tagitinin C induces ferroptosis through PERK-NRF2-HO-1 signaling pathway in colorectal cancer cells. Int J Biol Sci. 2021;17(11):2703‐2717. doi: 10.7150/ijbs.59404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Yao J, Zhao L, Zhao Q, et al. NF-κB and NRF2 signaling pathways contribute to wogonin-mediated inhibition of inflammation-associated colorectal carcinogenesis. Cell Death Dis. 2014;5(6):e1283‐e1283. doi: 10.1038/cddis.2014.221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Long M, Tao S, de la Vega M R, et al. NRF2-dependent suppression of azoxymethane/dextran sulfate sodium-induced colon carcinogenesis by the cinnamon-derived dietary factor cinnamaldehyde. Cancer Prevention Research. 2015;8(5):444‐454. doi: 10.1158/1940-6207.capr-14-0359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Darband S, Sadighparvar S, Yousefi B, et al. Quercetin attenuated oxidative DNA damage through NRF2 signaling pathway in rats with DMH induced colon carcinogenesis. Life Sci. 2020;253:117584. doi: 10.1016/j.lfs.2020.117584. [DOI] [PubMed] [Google Scholar]
- 38.Tao S, de la Vega M R, Chapman E, Ooi A, Zhang D. The effects of NRF2 modulation on the initiation and progression of chemically and genetically induced lung cancer. Mol Carcinog. 2017;57(2):182‐192. doi: 10.1002/mc.22745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Gong C, Yang H, Wang S, et al. hTERT promotes CRC proliferation and migration by recruiting YBX1 to increase NRF2 expression. Front Cell Dev Biol. 2021;9:658110. doi: 10.3389/fcell.2021.658101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Evans J, Winiarski B, Sutton P, et al. Correction: the NRF2 inhibitor brusatol is a potent antitumour agent in an orthotopic mouse model of colorectal cancer. Oncotarget. 2019;10(6):685‐685. doi: 10.18632/oncotarget.26625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Torrente L, Maan G, Oumkaltoum Rezig A, et al. High NRF2 levels correlate with poor prognosis in colorectal cancer patients and with sensitivity to the kinase inhibitor AT9283 in vitro. Biomolecules. 2020;10(10):1365. doi: 10.3390/biom10101365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Zhao X, Zhang Y, Xia Y, Zhou Z, Cao Y. Promoter demethylation of nuclear factor-erythroid 2-related factor 2 gene in drug-resistant colon cancer cells. Oncol Lett. 2015;10(3):1287‐1292. doi: 10.3892/ol.2015.3468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Payandeh Z, Pirpour Tazehkand A, Mansoori B, et al. The impact of NRF2 silencing on NRF2-PD-L1 axis to overcome oxaliplatin resistance and migration in colon cancer cells. Avicenna J Med Biotechnol. 2021;13(3):116-122. doi: 10.18502/ajmb.v13i3.6371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Liu C, Zhao Y, Wang J, et al. Foxo3 reverses 5-fluorouracil resistance in human colorectal cancer cells by inhibiting the NRF2/TR1 signaling pathway. Cancer Lett. 2020;470:29‐42. doi: 10.1016/j.canlet.2019.11.042. [DOI] [PubMed] [Google Scholar]
- 45.Kim H, Kim C, Lee D, Lee J, Oh E, Park H. Quinacrine-mediated inhibition of NRF2 reverses hypoxia-induced 5-fluorouracil resistance in colorectal cancer. Int J Mol Sci. 2019;20(18):4366. doi: 10.3390/ijms20184366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Pirpour Tazehkand A, Salehi R, Velaei K, Samadi N. The potential impact of trigonelline loaded micelles on NRF2 suppression to overcome oxaliplatin resistance in colon cancer cells. Mol Biol Rep. 2020;47(8):5817‐5829. doi: 10.1007/s11033-020-05650-w. [DOI] [PubMed] [Google Scholar]
- 47.Shen CJ, Lin PL, Lin HC, Cheng YW, Huang HS, Lee H. RV-59 suppresses cytoplasmic NRF2-mediated 5-fluorouracil resistance and tumor growth in colorectal cancer. Am J Cancer Res. 2019;9(12):2789‐2796. [PMC free article] [PubMed] [Google Scholar]
- 48.Afrin S, Giampieri F, Forbes-Hernández T, et al. Manuka honey synergistically enhances the chemopreventive effect of 5-fluorouracil on human colon cancer cells by inducing oxidative stress and apoptosis, altering metabolic phenotypes and suppressing metastasis ability. Free Radical Biol Med. 2018;126:41‐54. doi: 10.1016/j.freeradbiomed.2018.07.014. [DOI] [PubMed] [Google Scholar]
- 49.Wang Z, Sun X, Feng Y, et al. Dihydromyricetin reverses MRP2-induced multidrug resistance by preventing NF-κB-NRF2 signaling in colorectal cancer cell. Phytomedicine. 2021;82:153414. doi: 10.1016/j.phymed.2020.153414. [DOI] [PubMed] [Google Scholar]


