Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1805 2023-06-13 18:12:31 |
2 update references and layout Meta information modification 1805 2023-06-14 03:58:34 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Gjorgieva Ackova, D.; Maksimova, V.; Smilkov, K.; Buttari, B.; Arese, M.; Saso, L. Nuclear Factor Erythroid 2-Related Factor 2. Encyclopedia. Available online: https://encyclopedia.pub/entry/45523 (accessed on 02 July 2024).
Gjorgieva Ackova D, Maksimova V, Smilkov K, Buttari B, Arese M, Saso L. Nuclear Factor Erythroid 2-Related Factor 2. Encyclopedia. Available at: https://encyclopedia.pub/entry/45523. Accessed July 02, 2024.
Gjorgieva Ackova, Darinka, Viktorija Maksimova, Katarina Smilkov, Brigitta Buttari, Marzia Arese, Luciano Saso. "Nuclear Factor Erythroid 2-Related Factor 2" Encyclopedia, https://encyclopedia.pub/entry/45523 (accessed July 02, 2024).
Gjorgieva Ackova, D., Maksimova, V., Smilkov, K., Buttari, B., Arese, M., & Saso, L. (2023, June 13). Nuclear Factor Erythroid 2-Related Factor 2. In Encyclopedia. https://encyclopedia.pub/entry/45523
Gjorgieva Ackova, Darinka, et al. "Nuclear Factor Erythroid 2-Related Factor 2." Encyclopedia. Web. 13 June, 2023.
Nuclear Factor Erythroid 2-Related Factor 2
Edit

Being a controller of cytoprotective actions, inflammation, and mitochondrial function through participating in the regulation of multiple genes in response to stress-inducing endogenous or exogenous stressors, the transcription factor Nuclear Factor Erythroid 2-Related Factor 2 (NRF2) is considered the main cellular defense mechanism to maintain redox balance at cellular and tissue level. While a transient activation of NRF2 protects normal cells under oxidative stress, the hyperactivation of NRF2 in cancer cells may help them to survive and to adapt under oxidative stress. This can be detrimental and related to cancer progression and chemotherapy resistance. Therefore, inhibition of NRF2 activity may be an effective approach for sensitizing cancer cells to anticancer therapy.

NRF2 inhibitors alkaloids plant bioactive compound

1. Introduction

Cells are continuously exposed to exogenous stressors such as pollution, smoke, drugs, xenobiotics, ionizing radiation, and endogenous stressors derived by cellular metabolism. The presence of electrophiles or xenobiotics generates reactive oxygen species (ROS) which can induce alterations in the redox balance and promote cell death by damaging essential macromolecules such as lipids, proteins, and DNA. When ROS production increases or their scavenging by antioxidants decreases, cells undergo a process of oxidative stress [1][2]. If present in moderate concentrations, ROS are crucial for cellular life; indeed, they act as a second messengers in the transduction of extracellular signals and in the control of gene expression related to cellular proliferation, differentiation, and survival [3]. In fact, ROS have double importance in cancer. On the one hand, they can contribute to the occurrence of the modification of normal to cancer cells, on the other hand, a persistent oxidative stress renders cancer cells increasingly vulnerable to additional stressors and reverses drug resistance [4][5]. Elevated levels of ROS create conditions appropriate for the promotion of carcinogenesis through various mechanisms including generation of oxidative stress, which may favor oncogene activation, remodeling of tumor-suppressor genes, mitochondrial malfunction, and pro-inflammatory microenvironment [6][7]. In addition, cancer cells increase their unique antioxidative capacities [8][9][10][11]. In this way, cancer cells keep proliferation high and avoid ROS thresholds that would otherwise reduce the survival of cancer cells. In response to excessive ROS production, cancer cells activate several transcriptional programs for maintaining redox homeostasis [12][13][14][15][16][17].
Nuclear Factor Erythroid 2-Related Factor 2 (NRF2) is considered as the principal controller of cytoprotective activities in response to xenobiotics, electrophiles, and oxidants [5][18][19]. NRF2 plays important role in the control of oxidative stress, inflammation, mitochondrial function [20][21][22], and in the regulation of different genes. These genes are encoding antioxidant and Phase II metabolic enzymes and related proteins, linked to antioxidant response system (glutathione synthesis, xenobiotics detoxification, etc.) [23][24].
In healthy tissues, a transient activation of the NRF2 signaling pathway prevents cancer initiation by regulating several diverse transcriptional networks. More than 1000 genes possessing an Antioxidant Response Element (ARE) in their promoters can be activated by NRF2 during oxidative stress [25][26].
NRF2 is regulated by Kelch-like ECH-associated protein 1 (KEAP1), an adaptor subunit of Cullin 3-based E3 ubiquitin ligase (Cul3). Under normal conditions, KEAP1 binds to NRF2 in the cytoplasm and promotes the ubiquitination and proteasomal degradation of NRF2 and, thus, acts as a negative regulator of NRF2 [27][28][29]. In stress-inducing conditions, KEAP1, a cysteine-rich protein, readily reacts with intracellular ROS or electrophiles and alters NRF2/KEAP1 complex conformation thus inhibiting NRF2 ubiquitylation. Thus, when the interaction between NRF2 and KEAP1 is disrupted, proteasomal degradation of NRF2 decreases, causing de novo NRF2 synthesis within the cell and its increased translocation into the nucleus [30][31][32]. When NRF2 is imported to the nucleus, it heterodimerizes with small Maf proteins (sMaf) and binds to a regulatory enhancer sequence ARE, thus promoting the expression of antioxidant and detoxifying genes and down-modulating the production of pro-inflammatory mediators [21][33][34].
NRF2 is found in almost all cell types and tissues, but it is more abundant in tissues such as the intestine, lung, liver, and kidney, where detoxification reactions occur on site [35].
Recently, prooncogenic activity of NRF2 in cancer development, cancer progression, cancer cell growth and angiogenesis, and NRF2 involvement in anticancer therapy resistance is also under observation [2][36][37][38][39]. While NRF2 defends normal cells from oxidative stress, the excess of NRF2 in cancer cells may help them in adjusting to oxidative stress, surviving and resisting the redox environment (redox adaptation) [40][41]. In cancer cells, oncogenic signaling may affect the behavior of NRF2 by increasing its mRNA levels and generating constitutive NRF2 activation. The result is stress adaptation, drug resistance, cells proliferation, and activation of metabolic reprogramming and promotion of the expression of genes related to tumor progression. A cytoprotective role of NRF2 activation has been reported in many diseases associated with oxidative stress; thus, various natural sources (food and medicinal plants) have been tested for their ability to activate NRF2 and for their use as antioxidants and chemopreventive agents [42][43]. In many types of cancers, constitutive activation of NRF2 can be detrimental and responsible for cancer progression and chemotherapy resistance [44]; therefore, inhibition of NRF2 activity may be an effective approach for sensitizing cancer cells to anticancer drugs. Still, there is a scientific debate on whether activation or inhibition of NRF2 might represent an appropriate approach for the prevention and/or treatment of cancer. Regarding NRF2 modulatory effects, its activity as a detoxification agent of anticancer drugs (that possibly leads to chemoresistance) is well known; thus, favorable or adverse repercussions in cancer cells depend on a few factors, such as action control, tumor microenvironment, and cell type [19].
Among the known inhibitors of NRF2, alkaloids—natural bioactive components—are poorly investigated to date despite their pharmacological potential related to their high pro-oxidant and cytotoxic properties.
Alkaloids are secondary plant metabolites defined by Pelletier as a “cyclic organic compound containing nitrogen in a negative oxidation state which is of limited distribution among living organisms” [45].
Based on their complex structure and pharmacological properties, different classifications have been proposed. Several physiological pathways are involved in their biosynthesis. Most of them are derived from amino acids (tyrosine, tryptphan, ornithine, lysine, L-aspartate, anthranilic acid, and nicotinic acid), but there are also some classes of alkaloids that have been synthetized from steroids or terpenoids, and they are so-called pseudoalkaloids [46].
The most widely accepted method of classification of alkaloids is the chemical classification, where the main criterion is the presence of the basic heterocyclic nucleus (i.e., the chemical entity). According to their chemical structure, alkaloids have been grouped based on the nature of the heterocyclic ring in the following groups: mononuclear heterocyclic alkaloids represented as pyrrolidine alkaloids (hygrine) and aporphine alkaloids (boldine); polynuclear heterocyclic alkaloids given as tropane alkaloids (atropine); quinoline alkaloids (quinine); and Rauwolfia alkaloids (reserpine). Non-heterocyclic alkaloids have been found often in plants as atypical alkaloids and they are grouped in the phenylethylamine skeleton (ephedrine and capsaicin), tropolone skeleton (colchicine), and modified diterpene alkaloids (aconitine) [47].
The diversity in their chemical entity is the basic factor for the wide spectrum of associated pharmacological effects (antibacterial/antiviral activity, antitumor activity, antioxidant, anti-inflammatory, anticholinergic, neurological, and cardiovascular effects) [48].

2. The Cancer-Preventing Role of NRF2

Numerous studies confirm the regulatory role of NRF2 as a reflection of its cytoprotective action in xenobiotic/oxidative stress. It is reported that this factor regulates the expression of several ARE genes, such as glutathione-S-transferase (GST), heme oxygenase-1 (HO-1), NAD(P)H: quinone oxidoreductase 1 (NQO1), and γ-glutamylcysteine ligase (γ-GCS), thus exerting a protective function [49][50]. For this reason, it is assumed that by inducing gene expression via NRF2, a detoxification from ROS occurs, which leads to mitigation of the cell oxidative damage.
In addition, NRF2 was also shown to play a role in the response to inflammatory conditions. Namely, NRF2 was able to inhibit the oxidative-stress-derived activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) by decreasing ROS levels. NRF2 also inhibits its nuclear translocation, which in turn leads to a lowered expression of proinflammatory cytokines, as interleukin-1 (IL-1), interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α), and other mediators of inflammation [21]. Once its work has been completed, NRF2 is degraded in the cytoplasm, therefore allowing the cellular antioxidant defense to return to normal levels.
Indeed, the NRF2-related response to oxidative stress has been proposed in the pathogenesis of various diseases, such as various respiratory diseases [51], neurodegenerative diseases [52], inflammatory diseases [21], cardiovascular diseases [53], and metabolic diseases [54]. The NRF2 involvement in various types of cancer was also studied, including breast cancer [55], prostate cancer [56][57], urinary bladder cancer [58], hepatic carcinoma [59], pancreatic cancer [60], non-small cell lung cancer [61], and colorectal cancer [62].
The modulation of NRF2 can present an interesting option, both in the prevention and treatment of cancer as well as of other conditions, since its activation can lead to the promotion of antioxidant defense, a reduction in inflammation, detoxification of carcinogens, and inducing the apoptosis of cancer cells.

3. The Cancer-Promoting Role of NRF2 and Its Role in Chemoresistance

NRF2 is involved in increased sensitivity to a number of xenobiotic-induced toxicities and disease pathologies. An overactivation of NRF2 may lead to a negative impact, causing chemoresistance to therapy in cancer cells [63][64], thus highlighting the possibility of representing “a double-edged sword” [65]. The negative role of NRF2 highlighted by the promotion of the growth and survival of various types of cancer cells (NRF2-addicted/activated cancers) has been thoroughly studied and reviewed in the light of a deeper understanding of the mechanisms involved, and to explore the possibility of therapeutic approaches [66][67]. In these NRF2-addicted cell types, somatic mutation in NRF2 or its regulatory proteins (KEAP1) are on site, which blocks suppression of NRF2 levels (NRF2 is overexpressed and constitutively activated). These recurrent KEAP1 and NRF2 mutations (and in addition, missense mutations in Cul3) lead to metabolic reprogramming and allow cytoprotective action to cancer cells resulting in enormous proliferation and resistance to anticancer therapy [68]. Several mechanisms underlying the cancer-promoting role of NRF2 have been revealed (Figure 1):
Figure 1. Dual role of NRF2: during physiological ROS challenge, and during pathological overactivation.
-
Overactivation of NRF2 in an ROS-enriched environment and participation in cancer cell proliferation and tumorigenesis. In various cancers, the cell metabolism increases in order to meet the demands of high cell proliferation, which in turn leads to the generation of ROS that aggravate oxidative stress and disturb the redox balance of cancer cells [57][69]. NRF2 is able to adapt itself to operate in a high-oxidizing environment, thus becoming overactivated. The accumulation of NRF2 in cancer cells was shown to promote their survival and proliferation and confer a support to tumorigenesis [69]. Some studies report that cancer cells can take control of the overactivated NRF2, with this factor even being a key player in the modulation of anabolic metabolism required for cancer cell growth and proliferation [70].
-
Overexpression of NRF2 resulting in chemoresistance. Early studies have shown that overexpression of NRF2 is responsible for the resistance of cancer cells to chemotherapeutic agents as doxorubicin, cisplatin, and etoposide [71]. Recent studies have also reviewed the involvement of NRF2 in the activation of multidrug resistance proteins [72], in particular to modulate the expression of efflux transporters, of phase II metabolic enzymes [64], and of antiapoptotic genes (Bcl-2) [73][74], thus supporting the chemoresistance ability of cancer cells.

References

  1. Redza-Dutordoir, M.; Averill-Bates, D.A. Activation of apoptosis signaling pathways by reactive oxygen species. Biochim. Biophys. Acta 2016, 1863, 2977–2992.
  2. Ghareghomi, S.; Habibi-Rezaei, M.; Arese, M.; Saso, L.; Moosavi-Movahedi, A.A. Nrf2 modulation in breast cancer. Biomedicines 2022, 10, 2668.
  3. Murphy, M.P.; Holmgren, A.; Larsson, N.-G.; Halliwell, B.; Chang, C.J.; Kalyanaraman, B.; Rhee, S.G.; Thornalley, P.J.; Partridge, L.; Gems, D.; et al. Unraveling the biological roles of reactive oxygen species. Cell Metab. 2011, 13, 361–366.
  4. Kashyap, D.; Tuli, H.S.; Sak, K.; Garg, V.K.; Goel, N.; Punia, S.; Chaudhary, A. Role of reactive oxygen species in cancer progression. Curr. Pharmacol. Rep. 2019, 5, 79–86.
  5. Panieri, E.; Saso, L. Potential applications of Nrf2 inhibitors in cancer therapy. Oxid. Med. Cell. Longev. 2019, 2019, 8592348.
  6. Ogrunc, M.; Di Micco, R.; Liontos, M.; Bombardelli, L.; Mione, M.; Fumagalli, M.; Gorgoulis, V.G.; d’Adda di Fagagna, F. Oncogene-induced reactive oxygen species fuel hyperproliferation and DNA damage response activation. Cell Death Differ. 2014, 21, 998–1012.
  7. Cinat, D.; Coppes, R.P.; Barazzuol, L. DNA Damage-Induced Inflammatory Microenvironment and Adult Stem Cell Response. Front. Cell Dev. Biol. 2021, 9, 729136.
  8. Abreu, I.A.; Cabelli, D.E. Superoxide dismutases-a review of the metal-associated mechanistic variations. Biochim. Biophys. Acta 2010, 1804, 263–274.
  9. Chandimali, N.; Jeong, D.K.; Kwon, T. Peroxiredoxin II Regulates Cancer Stem Cells and Stemness-Associated Properties of Cancers. Cancers 2018, 10, 305.
  10. Rabilloud, T.; Heller, M.; Rigobello, M.P.; Bindoli, A.; Aebersold, R.; Lunardi, J. The mitochondrial antioxidant defence system and its response to oxidative stress. Proteomics 2001, 1, 1105–1110.
  11. Ahmadinejad, F.; Geir Møller, S.; Hashemzadeh-Chaleshtori, M.; Bidkhori, G.; Jami, M.-S. Molecular Mechanisms behind Free Radical Scavengers Function against Oxidative Stress. Antioxidants 2017, 6, 51.
  12. Ryter, S.W.; Kim, H.P.; Hoetzel, A.; Park, J.W.; Nakahira, K.; Wang, X.; Choi, A.M.K. Mechanisms of cell death in oxidative stress. Antioxid. Redox Signal. 2007, 9, 49–89.
  13. Yamamoto, M.; Kensler, T.W.; Motohashi, H. The KEAP1-NRF2 System: A Thiol-Based Sensor-Effector Apparatus for Maintaining Redox Homeostasis. Physiol. Rev. 2018, 98, 1169–1203.
  14. Klotz, L.-O.; Sánchez-Ramos, C.; Prieto-Arroyo, I.; Urbánek, P.; Steinbrenner, H.; Monsalve, M. Redox regulation of FoxO transcription factors. Redox Biol. 2015, 6, 51–72.
  15. Zhao, T.; Zhu, Y.; Morinibu, A.; Kobayashi, M.; Shinomiya, K.; Itasaka, S.; Yoshimura, M.; Guo, G.; Hiraoka, M.; Harada, H. HIF-1-mediated metabolic reprogramming reduces ROS levels and facilitates the metastatic colonization of cancers in lungs. Sci. Rep. 2014, 4, 3793.
  16. Yang, J.-Y.; Zong, C.S.; Xia, W.; Yamaguchi, H.; Ding, Q.; Xie, X.; Lang, J.-Y.; Lai, C.-C.; Chang, C.-J.; Huang, W.-C.; et al. ERK promotes tumorigenesis by inhibiting FOXO3a via MDM2-mediated degradation. Nat. Cell Biol. 2008, 10, 138–148.
  17. Pouremamali, F.; Pouremamali, A.; Dadashpour, M.; Soozangar, N.; Jeddi, F. An update of Nrf2 activators and inhibitors in cancer prevention/promotion. Cell Commun. Signal. 2022, 20, 100.
  18. Abed, D.A.; Goldstein, M.; Albanyan, H.; Jin, H.; Hu, L. Discovery of directinhibitors of Keap1–Nrf2 protein–protein interaction as potential therapeutic and preventive agents. Acta Pharm. Sin. B 2015, 5, 285–299.
  19. Catanzaro, E.; Calcabrini, C.; Turrini, E.; Sestili, P.; Fimognari, C. Nrf2: A potential therapeutic target for naturally occurring anticancer drugs? Expert Opin. Ther. Targets 2017, 21, 781–793.
  20. Gray, N.E.; Farina, M.; Tucci, P.; Saso, L. The Role of the NRF2 Pathway in Maintaining and Improving Cognitive Function. Biomedicines 2022, 10, 2043.
  21. Saha, S.; Buttari, B.; Panieri, E.; Profumo, E.; Saso, L. An overview of Nrf2 signaling pathway and its role in inflammation. Molecules 2020, 25, 5474.
  22. Panieri, E.; Pinho, S.A.; Afonso, G.J.M.; Oliveira, P.J.; Cunha-Oliveira, T.; Saso, L. NRF2 and Mitochondrial Function in Cancer and Cancer Stem Cells. Cells 2022, 11, 2401.
  23. Taguchi, K.; Motohashi, H.; Yamamoto, M. Molecular mechanisms of the Keap 1-Nrf2 pathway in stress response and cancer evolution. Genes Cells 2011, 16, 123–140.
  24. Rahban, M.; Habibi-Rezaei, M.; Mazaheri, M.; Saso, L.; Moosavi-Movahedi, A.A. Anti-viral potential and modulation of Nrf2 by curcumin: Pharmacological implications. Antioxidants 2020, 9, 1228.
  25. Malhotra, D.; Portales-Casamar, E.; Singh, A.; Srivastava, S.; Arenillas, D.; Happel, C.; Shyr, C.; Wakabayashi, N.; Kensler, T.W.; Wasserman, W.W.; et al. Global mapping of binding sites for Nrf2 identifies novel targets in cell survival response through ChIP-Seq profiling and network analysis. Nucleic Acids Res. 2010, 38, 5718–5734.
  26. Tonelli, C.; Chio, I.I.C.; Tuveson, D.A. Transcriptional Regulation by Nrf2. Antioxid. Redox Signal. 2018, 29, 1727–1745.
  27. Russo, G.L.; Moccia, S.; Russo, M.; Spagnuolo, C. Redox regulation by carotenoids: Evidence and conflicts for their application in cancer. Biochem. Pharmacol. 2021, 194, 114838.
  28. ATBC Cancer Prevention Study Group. The alpha-tocopherol, beta-carotene lung cancer prevention study: Design, methods, participant characteristics, and compliance. Ann. Epidemiol. 1994, 4, 1–10.
  29. Klein, E.A.; Thompson, I.M.; Tangen, C.M.; Crowley, J.J.; Lucia, M.S.; Goodman, P.J.; Minasian, L.M.; Ford, L.G.; Parnes, H.L.; Gaziano, J.M.; et al. Vitamin E and the risk of prostate cancer: The Selenium and Vitamin E Cancer Prevention Trial (SELECT). JAMA 2011, 306, 1549–1556.
  30. Kaspar, J.W.; Niture, S.K.; Jaiswal, A.K. Nrf2:INrf2 (Keap1) signaling in oxidative stress. Free Radic. Biol. Med. 2009, 47, 1304–1309.
  31. Kobayashi, M.; Li, L.; Iwamoto, N.; Nakajima-Takagi, Y.; Kaneko, H.; Nakayama, Y.; Eguchi, M.; Wada, Y.; Kumagai, Y.; Yamamoto, M. The antioxidant defense system Keap1-Nrf2 comprises a multiple sensing mechanism for responding to a wide range of chemical compounds. Mol. Cell. Biol. 2009, 29, 493–502.
  32. Shaw, P.; Chattopadhyay, A. Nrf2-ARE signaling in cellular protection: Mechanism of action and the regulatory mechanisms. J. Cell. Physiol. 2020, 235, 3119–3130.
  33. Zhu, J.; Wang, H.; Chen, F.; Fu, J.; Xu, Y.; Hou, Y.; Kou, H.H.; Zhai, C.; Nelson, M.B.; Zhang, Q.; et al. An overview of chemical inhibitors of the Nrf2-ARE signaling pathway and their potential applications in cancer therapy. Free Radic. Biol. Med. 2016, 99, 544–556.
  34. Zhang, D.D.; Hannink, M. Distinct cysteine residues in Keap1 are required for Keap1-dependent ubiquitination of Nrf2 and for stabilization of Nrf2 by chemopreventive agents and oxidative stress. Mol. Cell. Biol. 2003, 23, 8137–8151.
  35. Itoh, K.; Chiba, T.; Takahashi, S.; Ishii, T.; Igarashi, K.; Katoh, Y.; Oyake, T.; Hayashi, N.; Satoh, K.; Hatayama, I.; 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, 313–322.
  36. Jaramillo, M.C.; Zhang, D.D. The emerging role of the Nrf2–Keap1 signaling pathway in cancer. Genes Dev. 2013, 27, 2179–2191.
  37. Rojo, A.I.; Rada, P.; Mendiola, M.; Ortega-Molina, A.; Wojdyla, K.; Rogowska-Wrzesinska, A.; Hardisson, D.; Serrano, M.; Cuadrado, A. The PTEN/NRF2 axis promotes human carcinogenesis. Antioxid. Redox Signal. 2014, 21, 2498–2514.
  38. Zhang, C.; Wang, H.-J.; Bao, Q.-C.; Wang, L.; Guo, T.-K.; Chen, W.-L.; Xu, L.-L.; Zhou, H.-S.; Bian, J.-L.; Yang, Y.-R.; et al. NRF2 promotes breast cancer cell proliferation and metastasis by increasing RhoA/ROCK pathway signal transduction. Oncotarget 2016, 7, 73593–73606.
  39. Dauer, P.; Sharma, N.S.; Gupta, V.K.; Nomura, A.; Dudeja, V.; Saluja, A.; Banerjee, S. GRP78-mediated antioxidant response and ABC transporter activity confers chemoresistance to pancreatic cancer cells. Mol. Oncol. 2018, 12, 1498–1512.
  40. Sporn, M.B.; Liby, K.T. NRF2 and cancer: The good, the bad and the importance of context. Nat. Rev. Cancer 2012, 12, 564–571.
  41. Telkoparan-Akillilar, P.; Suzen, S.; Saso, L. Pharmacological Applications of Nrf2 Inhibitors as Potential Antineoplastic Drugs. Int. J. Mol. Sci. 2019, 20, 2025.
  42. No, J.H.; Kim, Y.-B.; Song, Y.S. Targeting Nrf2 signaling to combat chemoresistance. J. Cancer Prev. 2014, 19, 111–117.
  43. Panieri, E.; Buha, A.; Telkoparan-Akillilar, P.; Cevik, D.; Kouretas, D.; Veskoukis, A.; Skaperda, Z.; Tsatsakis, A.; Wallace, D.; Suzen, S.; et al. Potential applications of Nrf2 modulators in cancer therapy. Antioxidants 2020, 9, 193.
  44. Sukumari-Ramesh, S.; Prasad, N.; Alleyne, C.H., Jr.; Vender, J.R.; Dhandapani, K.M. Overexpression of Nrf2 attenuates Carmustine-induced cytotoxicity in U87MG human glioma cells. BMC Cancer 2015, 15, 118.
  45. Nakagawa, A.; Matsumura, E.; Sato, F.; Minami, H. Bioengineering of isoquinoline alkaloid production in microbial systems. Adv. Bot. Res. 2013, 68, 183–203.
  46. Gutiérrez-Grijalva, E.P.; López-Martínez, L.X.; Contreras-Angulo, L.A.; Elizalde-Romero, C.A.; Heredia, J.B. Plant Alkaloids: Structures and Bioactive Properties. In Plant-Derived Bioactives; Swamy, M., Ed.; Springer: Singapore, 2020.
  47. Bhambhani, S.; Kondhare, K.R.; Giri, A.P. Diversity in Chemical Structures and Biological Properties of Plant Alkaloids. Molecules 2021, 26, 3374.
  48. Thawabteh, A.; Juma, S.; Bader, M.; Karaman, D.; Scrano, L.; Bufo, S.A.; Karaman, R. The Biological Activity of Natural Alkaloids against Herbivores, Cancerous Cells and Pathogens. Toxins 2019, 11, 656.
  49. Kim, J.; Keum, Y.-S. NRF2, a Key Regulator of Antioxidants with Two Faces towards Cancer. Oxid. Med. Cell. Longev. 2016, 2016, 2746457.
  50. Armagan, G.; Sevgiliv, E.; Gürkan, F.T.; Köse, F.A.; Bilgiç, T.; Dagcı, T.; Saso, L. Regulation of the Nrf2 Pathway by Glycogen Synthase Kinase-3β in MPP+-Induced Cell Damage. Molecules 2019, 24, 1377.
  51. Mizumura, K.; Maruoka, S.; Shimizu, T.; Gon, Y. Role of Nrf2 in the pathogenesis of respiratory diseases. Respir. Investig. 2020, 58, 28–35.
  52. Saha, S.; Buttari, B.; Profumo, E.; Tucci, P.; Saso, L. A Perspective on Nrf2 Signaling Pathway for Neuroinflammation: A Potential Therapeutic Target in Alzheimer’s and Parkinson’s Diseases. Front. Cell. Neurosci. 2022, 15, 787258.
  53. Satta, S.; Mahmoud, A.M.; Wilkinson, F.L.; Alexander, M.Y.; White, S.J. The Role of Nrf2 in Cardiovascular Function and Disease. Oxid. Med. Cell. Longev. 2017, 2017, 9237263.
  54. Zhang, Z.; Zhou, S.; Jiang, X.; Wang, Y.H.; Li, F.; Wang, Y.G.; Zheng, Y.; Cai, L. The role of the Nrf2/Keap1 pathway in obesity and metabolic syndrome. Rev. Endocr. Metab. Disord. 2015, 16, 35–45.
  55. Tascioglu Aliyev, A.; Panieri, E.; Stepanic, V.; Gurer-Orhan, H.; Saso, L. Involvement of NRF2 in Breast Cancer and Possible Therapeutical Role of Polyphenols and Melatonin. Molecules 2021, 26, 1853.
  56. Han, C.; Wang, Z.; Xu, Y.; Chen, S.; Han, Y.; Li, L.; Wang, M.; Jin, X. Roles of reactive oxygen species in biological behaviors of prostate cancer. Biomed Res. Int. 2020, 2020, 1269624.
  57. Buttari, B.; Arese, M.; Oberley-Deegan, R.E.; Saso, L.; Chatterjee, A. NRF2: A crucial regulator for mitochondrial metabolic shift and prostate cancer progression. Front. Physiol. 2022, 13, 989793.
  58. Iida, K.; Itoh, K.; Maher, J.M.; Kumagai, Y.; Oyasu, R.; Mori, Y.; Shimazui, T.; Akaza, H.; Yamamoto, M. Nrf2 and p53 cooperatively protect against BBN-induced urinary bladder carcinogenesis. Carcinogenesis 2007, 28, 2398–2403.
  59. Kitamura, Y.; Umemura, T.; Kanki, K.; Kodama, Y.; Kitamoto, S.; Saito, K.; Itoh, K.; Yamamoto, M.; Masegi, T.; Nishikawa, A.; et al. Increased susceptibility to hepatocarcinogenicity of Nrf2-deficient mice exposed to 2-amino-3-methylimidazo quinoline. Cancer Sci. 2007, 98, 19–24.
  60. Cykowiak, M.; Krajka-Kuźniak, V. Role of Nrf2 in Pancreatic Cancer. Antioxidants 2022, 11, 98.
  61. Sánchez-Ortega, M.; Carrera, A.C.; Garrido, A. Role of NRF2 in Lung Cancer. Cells 2021, 10, 1879.
  62. Sadeghi, M.R.; Jeddi, F.; Soozangar, N.; Somi, M.H.; Samadi, N. The role of Nrf2-Keap1 axis in colorectal cancer, progression, and chemoresistance. Tumour Biol. 2017, 39, 1010428317705510.
  63. Solis, L.M.; Behrens, C.; Dong, W.; Suraokar, M.; Ozburn, N.C.; Moran, C.A.; Corvalan, A.N.; Biswal, S.; Swisher, S.G.; Nebiyou Bekele, B.; et al. Nrf2 and Keap1 abnormalities in non-small cell lung carcinoma and association with clinicopathologic features. Clin. Cancer Res. 2010, 16, 3743–3753.
  64. Bai, X.; Chen, Y.; Hou, X.; Huang, M.; Jin, J. Emerging role of NRF2 in chemoresistance by regulating drug-metabolizing enzymes and efflux transporters. Drug Metab. Rev. 2016, 48, 541–567.
  65. Wu, S.; Lu, H.; Bai, Y. Nrf2 in cancers: A double-edged sword. Cancer Med. 2019, 8, 2252–2267.
  66. Syu, J.-P.; Chi, J.-T.; Kung, H.-N. Nrf2 Contributes to the Poor Prognosis and Chemoresistance. In The Transcription Factor Nrf2; Morales-Gonzalez, J.A., Morales-Gonzalez, A., Madrigal-Santillan, E.O., Eds.; InTech Open: Rijeka, Croatia, 2016.
  67. Telkoparan-Akillilar, P.; Panieri, E.; Cevik, D.; Suzen, S.; Saso, L. Therapeutic targeting of the NRF2 signaling pathway in cancer. Molecules 2021, 26, 1417.
  68. Panda, H.; Wen, H.; Suzuki, M.; Yamamoto, M. Multifaceted roles of the KEAP1-NRF2 system in cancer and inflammatory disease milieu. Antioxidants 2022, 11, 538.
  69. DeNicola, G.M.; Karreth, F.A.; Humpton, T.J.; Gopinathan, A.; Wei, C.; Frese, K.; Mangal, D.; Yu, K.H.; Yeo, C.J.; Calhoun, E.S.; et al. Oncogene-induced Nrf2 transcription promotes ROS detoxification and tumorigenesis. Nature 2011, 475, 106–109.
  70. Mitsuishi, Y.; Taguchi, K.; Kawatani, Y.; Shibata, T.; Nukiwa, T.; Aburatani, H.; Yamamoto, M.; Motohashi, H. Nrf2 redirects glucose and glutamine into anabolic pathways in metabolic reprogramming. Cancer Cell 2012, 22, 66–79.
  71. Wang, X.J.; Sun, Z.; Villeneuve, N.F.; Zhang, S.; Zhao, F.; Li, Y.; Chen, W.; Yi, X.; Zheng, W.; Wondrak, G.T.; et al. Nrf2 enhances resistance of cancer cells to chemotherapeutic drugs, the dark side of Nrf2. Carcinogenesis 2008, 29, 1235–1243.
  72. Tian, B.; Lu, Z.-N.; Guo, X.-L. Regulation and role of nuclear factor-E2-related factor 2 (Nrf2) in multidrug resistance of hepatocellular carcinoma. Chem. Biol. Interact. 2018, 280, 70–76.
  73. Niture, S.K.; Jaiswal, A.K. Nrf2 protein up-regulates antiapoptotic protein Bcl-2 and prevents cellular apoptosis. J. Biol. Chem. 2012, 287, 9873–9886.
  74. Shelton, P.; Jaiswal, A.K. The transcription factor NF-E2-related factor 2 (Nrf2): A protooncogene? FASEB J. 2013, 27, 414–423.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , ,
View Times: 351
Revisions: 2 times (View History)
Update Date: 14 Jun 2023
1000/1000
Video Production Service