DOI QR코드

DOI QR Code

Transcriptional Regulation of the AP-1 and Nrf2 Target Gene Sulfiredoxin

  • Soriano, Francesc X. (Centre for Integrative Physiology, University of Edinburgh) ;
  • Baxter, Paul (Centre for Integrative Physiology, University of Edinburgh) ;
  • Murray, Lyndsay M. (Centre for Integrative Physiology, University of Edinburgh) ;
  • Sporn, Michael B. (Dartmouth Medical School) ;
  • Gillingwater, Thomas H. (Centre for Integrative Physiology, University of Edinburgh) ;
  • Hardingham, Giles E. (Centre for Integrative Physiology, University of Edinburgh)
  • Received : 2009.02.12
  • Accepted : 2009.02.15
  • Published : 2009.03.31

Abstract

"Two-cysteine" peroxiredoxins are antioxidant enzymes that exert a cytoprotective effect in many models of oxidative stress. However, under highly oxidizing conditions they can be inactivated through hyperoxidation of their peroxidatic active site cysteine residue. Sulfiredoxin can reverse this hyperoxidation, thus reactivating peroxiredoxins. Here we review recent investigations that have shed further light on sulfiredoxin's role and regulation. Studies have revealed sulfiredoxin to be a dynamically regulated gene whose transcription is induced by a variety of signals and stimuli. Sulfiredoxin expression is regulated by the transcription factor AP-1, which mediates its up-regulation by synaptic activity in neurons, resulting in protection against oxidative stress. Furthermore, sulfiredoxin has been identified as a new member of the family of genes regulated by Nuclear factor erythroid 2-related factor (Nrf2) via a conserved cis-acting antioxidant response element (ARE). As such, sulfiredoxin is likely to contribute to the net antioxidative effect of small molecule activators of Nrf2. As discussed here, the proximal AP-1 site of the sulfiredoxin promoter is embedded within the ARE, as is common with Nrf2 target genes. Other recent studies have shown that sulfiredoxin induction via Nrf2 may form an important part of the protective response to oxidative stress in the lung, preventing peroxiredoxin hyperoxidation and, in certain cases, subsequent degradation. We illustrate here that sulfiredoxin can be rapidly induced in vivo by administration of CDDO-TFEA, a synthetic triterpenoid inducer of endogenous Nrf2, which may offer a way of reversing peroxiredoxin hyperoxidation in vivo following chronic or acute oxidative stress.

Keywords

Acknowledgement

Supported by : Royal Society

References

  1. Bae, S.H., Woo, H.A., Sung, S.H., Lee, H.E., Lee, S.K., Kil, I.S., and Rhee, S.G. (2009). Induction of sulfiredoxin via an Nrf2-dependent pathway and hyperoxidation of peroxiredoxin III in the lungs of mice exposed to hyperoxia. Antioxid. Redox Signal. [Epub ahead of print] https://doi.org/10.1089/ars.2008.2325
  2. Biteau, B., Labarre, J., and Toledano, M.B. (2003). ATP-dependent reduction of cysteine-sulphinic acid by S. cerevisiae sulphiredoxin. Nature 425, 980-984 https://doi.org/10.1038/nature02075
  3. Boulos, S., Meloni, B.P., Arthur, P.G., Bojarski, C., and Knuckey, N.W. (2007). Peroxiredoxin 2 overexpression protects cortical neuronal cultures from ischemic and oxidative injury but not glutamate excitotoxicity, whereas Cu/Zn superoxide dismutase 1 overexpression protects only against oxidative injury. J. Neurosci. Res. 85, 3089-3097 https://doi.org/10.1002/jnr.21429
  4. Brown, P.H., Alani, R., Preis, L.H., Szabo, E., and Birrer, M.J.(1993). Suppression of oncogene-induced transformation by a deletion mutant of c-jun. Oncogene 8, 877-886
  5. Budanov, A.V., Sablina, A.A., Feinstein, E., Koonin, E.V., and Chumakov, P.M. (2004). Regeneration of peroxiredoxins by p53-regulated sestrins, homologs of bacterial AhpD. Science 304, 596-600 https://doi.org/10.1126/science.1095569
  6. Chang, T.S., Jeong, W., Woo, H.A., Lee, S.M., Park, S., and Rhee, S.G. (2004). Characterization of mammalian sulfiredoxin and its reactivation of hyperoxidized peroxiredoxin through reduction of cysteine sulfinic acid in the active site to cysteine. J. Biol. Chem. 279, 50994-51001 https://doi.org/10.1074/jbc.M409482200
  7. Fang, J., Nakamura, T., Cho, D.H., Gu, Z., and Lipton, S.A. (2007). S-nitrosylation of peroxiredoxin 2 promotes oxidative stressinduced neuronal cell death in Parkinson's disease. Proc. Natl. Acad. Sci. USA 104,18742-18747 https://doi.org/10.1073/pnas.0705904104
  8. Findlay, V.J., Townsend, D.M., Morris, T.E., Fraser, J.P., He, L., and Tew, K.D. (2006). A novel role for human sulfiredoxin in the reversal of glutathionylation. Cancer Res. 66, 6800-6806 https://doi.org/10.1158/0008-5472.CAN-06-0484
  9. Giudice, A., and Montella, M. (2006). Activation of the Nrf2-ARE signaling pathway: a promising strategy in cancer prevention. Bioessays 28,169-181 https://doi.org/10.1002/bies.20359
  10. Glauser, D.A., Brun, T., Gauthier, B.R., and Schlegel, W. (2007). Transcriptional response of pancreatic beta cells to metabolic stimulation: large scale identification of immediate-early and secondary response genes. BMC Mol. Biol. 8, 54 https://doi.org/10.1186/1471-2199-8-54
  11. Hattori, F., Murayama, N., Noshita, T., and Oikawa, S. (2003). Mitochondrial peroxiredoxin-3 protects hippocampal neurons from excitotoxic injury in vivo. J. Neurochem. 86, 860-868 https://doi.org/10.1046/j.1471-4159.2003.01918.x
  12. Immenschuh, S., and Baumgart-Vogt, E. (2005). Peroxiredoxins, oxidative stress, and cell proliferation. Antioxid. Redox Signal. 7, 768-777 https://doi.org/10.1089/ars.2005.7.768
  13. Jeong, W., Park, S.J., Chang, T.S., Lee, D.Y., and Rhee, S.G. (2006). Molecular mechanism of the reduction of cysteine sulfinic acid of peroxiredoxin to cysteine by mammalian sulfiredoxin. J. Biol. Chem. 281,14400-14407 https://doi.org/10.1074/jbc.M511082200
  14. Jonsson, T.J., Johnson, L.C., and Lowther, W.T. (2008a). Structure of the sulphiredoxin-peroxiredoxin complex reveals an essential repair embrace. Nature 451, 98-101 https://doi.org/10.1038/nature06415
  15. Jonsson, T.J., Murray, M.S., Johnson, L.C., and Lowther, W.T.(2008b). Reduction of cysteine sulfinic acid in peroxiredoxin by sulfiredoxin proceeds directly through a sulfinic phosphoryl ester intermediate. J. Biol. Chem. 283, 23846-23851 https://doi.org/10.1074/jbc.M803244200
  16. Kensler, T.W., Wakabayashi, N., and Biswal, S. (2007). Cell survival responses to environmental stresses via the Keap1-Nrf2-ARE pathway. Annu. Rev. Pharmacol. Toxicol. 47, 89-116 https://doi.org/10.1146/annurev.pharmtox.46.120604.141046
  17. Lee, J.M., Li, J., Johnson, D.A., Stein, T.D., Kraft, A.D., Calkins, M.J., Jakel, R.J., and Johnson, J.A. (2005). Nrf2, a multi-organ protector? FASEB J. 19, 1061-1066 https://doi.org/10.1096/fj.04-2591hyp
  18. Liby, K.T., Yore, M.M., and Sporn, M.B. (2007). Triterpenoids and rexinoids as multifunctional agents for the prevention and treatment of cancer. Nat. Rev. Cancer 7, 357-369 https://doi.org/10.1038/nrc2129
  19. Nguyen, T., Yang, C.S., and Pickett, C.B. (2004). The pathways and molecular mechanisms regulating Nrf2 activation in response to chemical stress. Free Radic. Biol. Med. 37, 433-441 https://doi.org/10.1016/j.freeradbiomed.2004.04.033
  20. Nioi, P., McMahon, M., Itoh, K., Yamamoto, M., and Hayes, J.D.(2003). Identification of a novel Nrf2-regulated antioxidant response element (ARE) in the mouse NAD(P)H:quinone oxidoreductase 1 gene: reassessment of the ARE consensus sequence. Biochem. J. 374, 337-348 https://doi.org/10.1042/BJ20030754
  21. Noh, Y.H., Baek, J.Y., Jeong, W., Rhee, S.G., and Chang, T.S.(2009). Sulfiredoxin translocation into mitochondria plays a crucial role in reducing hyperoxidized peroxiredoxin III. J. Biol. Chem. doi:10.1074/jbc.M808981200
  22. Papadia, S., Soriano, F.X., Leveille, F., Martel, M.A., Dakin, K.A., Hansen, H.H., Kaindl, A., Sifringer, M., Fowler, J., Stefovska, V., et al. (2008). Synaptic NMDA receptor activity boosts intrinsic antioxidant defenses. Nat. Neurosci. 11, 476-487 https://doi.org/10.1038/nn2071
  23. Qu, D., Rashidian, J., Mount, M.P., Aleyasin, H., Parsanejad, M., Lira, A., Haque, E., Zhang, Y., Callaghan, S., Daigle, M., et al.(2007). Role of Cdk5-mediated phosphorylation of Prx2 in MPTP toxicity and Parkinson's disease. Neuron 55, 37-52 https://doi.org/10.1016/j.neuron.2007.05.033
  24. Rhee, S.G., Chae, H.Z., and Kim, K. (2005). Peroxiredoxins: a historical overview and speculative preview of novel mechanisms and emerging concepts in cell signaling. Free Radic. Biol. Med. 38, 1543-1552 https://doi.org/10.1016/j.freeradbiomed.2005.02.026
  25. Rhee, S.G., Jeong, W., Chang, T.S., and Woo, H.A. (2007). Sulfiredoxin, the cysteine sulfinic acid reductase specific to 2-Cys peroxiredoxin: its discovery, mechanism of action, and biological significance. Kidney Int. 106, S3-8 https://doi.org/10.1038/sj.ki.5002380
  26. Rhee, S.G., Woo, H.A., Bae, S.H., and Park, S. (2009) Sestrin 2 is not a reductase for cysteine sulfinic acid of peroxiredoxins. Antioxid. Redox Signal. 11, 739-745. (in press) https://doi.org/10.1089/ars.2008.2360
  27. Sanchez-Font, M.F., Sebastia, J., Sanfeliu, C., Cristofol, R., Marfany, G., and Gonzalez-Duarte, R. (2003). Peroxiredoxin 2(PRDX2), an antioxidant enzyme, is under-expressed in Down syndrome fetal brains. Cell Mol. Life Sci. 60,1513-1523 https://doi.org/10.1007/s00018-003-3048-1
  28. Shih, A.Y., Li, P., and Murphy, T.H. (2005). A small-moleculeinducible Nrf2-mediated antioxidant response provides effective prophylaxis against cerebral ischemia in vivo. J. Neurosci. 25, 10321-10335 https://doi.org/10.1523/JNEUROSCI.4014-05.2005
  29. Singh, A., Ling, G., Suhasini, A.N., Zhang, P., Yamamoto, M., Navas-Acien, A., Cosgrove, G., Tuder, R.M., Kensler, T.W., Watson, W.H., et al. (2009). Nrf2-dependent sulfiredoxin-1 expression protects against cigarette smoke-induced oxidative stress in lungs. Free Radic. Biol. Med. 46, 376-386 https://doi.org/10.1016/j.freeradbiomed.2008.10.026
  30. Soriano, F.X., Leveille, F., Papadia, S., Higgins, L.G., Varley, J., Baxter, P., Hayes, J.D., and Hardingham, G.E. (2008). Induction of sulfiredoxin expression and reduction of peroxiredoxin hyperoxidation by the neuroprotective Nrf2 activator 3H-1,2-dithiole-3-thione. J. Neurochem. 107, 533-543 https://doi.org/10.1111/j.1471-4159.2008.05648.x
  31. Wasserman, W.W., and Fahl, W.E. (1997). Comprehensive analysis of proteins which interact with the antioxidant responsive element: correlation of ARE-BP-1 with the chemoprotective induction response. Arch. Biochem. Biophys. 344, 387-396 https://doi.org/10.1006/abbi.1997.0215
  32. Wei, Q., Jiang, H., Matthews, C.P., and Colburn, N.H. (2008). Sulfiredoxin is an AP-1 target gene that is required for transformation and shows elevated expression in human skin malignancies. Proc. Natl. Acad. Sci. USA 105,19738-19743 https://doi.org/10.1073/pnas.0810676105
  33. Wood, Z.A., Schroder, E., Robin Harris, J., and Poole, L.B. (2003). Structure, mechanism and regulation of peroxiredoxins. Trends Biochem. Sci. 28, 32-40 https://doi.org/10.1016/S0968-0004(02)00003-8
  34. Yao, J., Taylor, M., Davey, F., Ren, Y., Aiton, J., Coote, P., Fang, F., Chen, J.X., Yan, S.D., and Gunn-Moore, F.J. (2007). Interaction of amyloid binding alcohol dehydrogenase/Abeta mediates upregulation of peroxiredoxin II in the brains of Alzheimer's disease patients and a transgenic Alzheimer's disease mouse model. Mol. Cell. Neurosci. 35, 377-382 https://doi.org/10.1016/j.mcn.2007.03.013
  35. Yates, M.S., Tauchi, M., Katsuoka, F., Flanders, K.C., Liby, K.T., Honda, T., Gribble, G.W., Johnson, D.A., Johnson, J.A., Burton, N.C., et al. (2007). Pharmacodynamic characterization of chemopreventive triterpenoids as exceptionally potent inducers of Nrf2-regulated genes. Mol. Cancer Ther. 6, 154-162 https://doi.org/10.1158/1535-7163.MCT-06-0516
  36. Zhang, D.D. (2006). Mechanistic studies of the Nrf2-Keap1 signaling pathway. Drug Metab. Rev. 38, 769-789 https://doi.org/10.1080/03602530600971974

Cited by

  1. Nrf2 the rescue: Effects of the antioxidative/electrophilic response on the liver vol.244, pp.1, 2010, https://doi.org/10.1016/j.taap.2010.01.013
  2. Reexamination of the electrophile response element sequences and context reveals a lack of consensus in gene function vol.1799, pp.7, 2009, https://doi.org/10.1016/j.bbagrm.2010.05.003
  3. Triterpenoids CDDO-ethyl amide and CDDO-trifluoroethyl amide improve the behavioral phenotype and brain pathology in a transgenic mouse model of Huntington's disease vol.49, pp.2, 2010, https://doi.org/10.1016/j.freeradbiomed.2010.03.017
  4. Redox Regulation of Lipopolysaccharide-mediated Sulfiredoxin Induction, Which Depends on Both AP-1 and Nrf2 vol.285, pp.45, 2009, https://doi.org/10.1074/jbc.m110.126839
  5. Reduction of Cysteine Sulfinic Acid in Eukaryotic, Typical 2-Cys Peroxiredoxins by Sulfiredoxin vol.15, pp.1, 2009, https://doi.org/10.1089/ars.2010.3564
  6. Activation of Nrf2-Regulated Glutathione Pathway Genes by Ischemic Preconditioning vol.2011, pp.None, 2011, https://doi.org/10.1155/2011/689524
  7. Nuclear factor E2-related factor 2 Dependent Overexpression of Sulfiredoxin and Peroxiredoxin III in Human Lung Cancer vol.26, pp.3, 2011, https://doi.org/10.3904/kjim.2011.26.3.304
  8. hTERT Overexpression Alleviates Intracellular ROS Production, Improves Mitochondrial Function, and Inhibits ROS-Mediated Apoptosis in Cancer Cells vol.71, pp.1, 2009, https://doi.org/10.1158/0008-5472.can-10-1588
  9. CNS Peroxiredoxins and Their Regulation in Health and Disease vol.14, pp.8, 2009, https://doi.org/10.1089/ars.2010.3567
  10. Gene expression analysis of precision-cut human liver slices indicates stable expression of ADME-Tox related genes vol.253, pp.1, 2009, https://doi.org/10.1016/j.taap.2011.03.010
  11. Role of sulfiredoxin as a regulator of peroxiredoxin function and regulation of its expression vol.53, pp.3, 2012, https://doi.org/10.1016/j.freeradbiomed.2012.05.020
  12. Bardoxolone Methyl Decreases Megalin and Activates Nrf2 in the Kidney vol.23, pp.10, 2009, https://doi.org/10.1681/asn.2012050457
  13. The Subtype of GluN2 C-terminal Domain Determines the Response to Excitotoxic Insults vol.74, pp.3, 2009, https://doi.org/10.1016/j.neuron.2012.03.021
  14. Genetic Polymorphisms and Protein Expression of NRF2 and Sulfiredoxin Predict Survival Outcomes in Breast Cancer vol.72, pp.21, 2009, https://doi.org/10.1158/0008-5472.can-12-1474
  15. Loss of sulfiredoxin renders mice resistant to azoxymethane/dextran sulfate sodium-induced colon carcinogenesis vol.34, pp.6, 2009, https://doi.org/10.1093/carcin/bgt059
  16. Human stem cell-derived astrocytes and their application to studying Nrf2-mediated neuroprotective pathways and therapeutics in neurodegeneration vol.75, pp.4, 2013, https://doi.org/10.1111/bcp.12022
  17. SMRT-mediated co-shuttling enables export of class IIa HDACs independent of their CaM kinase phosphorylation sites vol.124, pp.1, 2009, https://doi.org/10.1111/jnc.12058
  18. Sulforaphane Enhances the Ability of Human Retinal Pigment Epithelial Cell against Oxidative Stress, and Its Effect on Gene Expression Profile Evaluated by Microarray Analysis vol.2013, pp.None, 2013, https://doi.org/10.1155/2013/413024
  19. Epigenetic Mechanisms in Stroke and Epilepsy vol.38, pp.1, 2009, https://doi.org/10.1038/npp.2012.134
  20. Global Transcriptome Profile of Cryptococcus neoformans during Exposure to Hydrogen Peroxide Induced Oxidative Stress vol.8, pp.1, 2009, https://doi.org/10.1371/journal.pone.0055110
  21. Studies onIn VivoFunction of Peroxiredoxins in Knockout Mice vol.33, pp.2, 2013, https://doi.org/10.7599/hmr.2013.33.2.97
  22. Telomerase reverse transcriptase promotes the proliferation of human laryngeal carcinoma cells through activation of the activator protein 1 vol.6, pp.1, 2009, https://doi.org/10.3892/ol.2013.1344
  23. Influence of GluN2 subunit identity on NMDA receptor function vol.74, pp.None, 2009, https://doi.org/10.1016/j.neuropharm.2013.01.016
  24. Recovery of NMDA receptor currents from MK-801 blockade is accelerated by Mg 2+ and memantine under conditions of agonist exposure vol.74, pp.None, 2009, https://doi.org/10.1016/j.neuropharm.2013.01.024
  25. Role of sulfiredoxin in systemic diseases influenced by oxidative stress vol.2, pp.None, 2009, https://doi.org/10.1016/j.redox.2014.09.002
  26. Effect of dietary α-lipoic acid on the mRNA expression of genes involved in drug metabolism and antioxidation system in rat liver vol.112, pp.3, 2009, https://doi.org/10.1017/s0007114514000841
  27. Tumor promoter-induced sulfiredoxin is required for mouse skin tumorigenesis vol.35, pp.5, 2009, https://doi.org/10.1093/carcin/bgu035
  28. The Cryptococcus neoformans Transcriptome at the Site of Human Meningitis vol.5, pp.1, 2009, https://doi.org/10.1128/mbio.01087-13
  29. Topical Application of the Synthetic Triterpenoid RTA 408 Protects Mice from Radiation-Induced Dermatitis vol.181, pp.5, 2009, https://doi.org/10.1667/rr13578.1
  30. Nuclear Heme Oxygenase-1 (HO-1) Modulates Subcellular Distribution and Activation of Nrf2, Impacting Metabolic and Anti-oxidant Defenses vol.289, pp.39, 2009, https://doi.org/10.1074/jbc.m114.567685
  31. Quantitative structure-activity relationship studies of dibenzo[a,d]cycloalkenimine derivatives for non-competitive antagonists of N-methyl-d-aspartate based on density functional theory with electron vol.9, pp.2, 2009, https://doi.org/10.1016/j.jtusci.2014.10.006
  32. Effect of MK-801 and Clozapine on the Proteome of Cultured Human Oligodendrocytes vol.10, pp.None, 2009, https://doi.org/10.3389/fncel.2016.00052
  33. Hepatic 3D spheroid models for the detection and study of compounds with cholestatic liability vol.6, pp.None, 2016, https://doi.org/10.1038/srep35434
  34. Thioredoxin 1 protects astrocytes from oxidative stress by maintaining peroxiredoxin activity vol.13, pp.3, 2009, https://doi.org/10.3892/mmr.2016.4855
  35. Adaptive regulation of the brain’s antioxidant defences by neurons and astrocytes vol.100, pp.None, 2009, https://doi.org/10.1016/j.freeradbiomed.2016.06.027
  36. Gene Expression Profiling in Human Lung Cells Exposed to Isoprene-Derived Secondary Organic Aerosol vol.51, pp.14, 2009, https://doi.org/10.1021/acs.est.7b01967
  37. The promising potential role of ketones in inflammatory dermatologic disease: a new frontier in treatment research vol.28, pp.6, 2017, https://doi.org/10.1080/09546634.2016.1276259
  38. Nrf2-dependent persistent oxidative stress results in stress-induced vulnerability to depression vol.22, pp.12, 2009, https://doi.org/10.1038/mp.2016.144
  39. A c-Jun N-terminal kinase inhibitor, JNK-IN-8, sensitizes triple negative breast cancer cells to lapatinib vol.8, pp.62, 2009, https://doi.org/10.18632/oncotarget.20581
  40. The Multifaceted Impact of Peroxiredoxins on Aging and Disease vol.29, pp.13, 2009, https://doi.org/10.1089/ars.2017.7452
  41. Reactive oxygen species regulate activity-dependent neuronal plasticity in Drosophila vol.7, pp.None, 2009, https://doi.org/10.7554/elife.39393
  42. XBP1-KLF9 Axis Acts as a Molecular Rheostat to Control the Transition from Adaptive to Cytotoxic Unfolded Protein Response vol.25, pp.1, 2009, https://doi.org/10.1016/j.celrep.2018.09.013
  43. Metabolism-Based Therapeutic Strategies Targeting Cancer Stem Cells vol.10, pp.None, 2009, https://doi.org/10.3389/fphar.2019.00203
  44. Earliest Mechanisms of Dopaminergic Neurons Sufferance in a Novel Slow Progressing Ex Vivo Model of Parkinson Disease in Rat Organotypic Cultures of Substantia Nigra vol.20, pp.9, 2009, https://doi.org/10.3390/ijms20092224
  45. SRXN1 Is Necessary for Resolution of GnRH-Induced Oxidative Stress and Induction of Gonadotropin Gene Expression vol.160, pp.11, 2009, https://doi.org/10.1210/en.2019-00283
  46. Neuronal Activity and Its Role in Controlling Antioxidant Genes vol.21, pp.6, 2009, https://doi.org/10.3390/ijms21061933
  47. Tanshinone IIA pretreatment promotes cell survival in human lung epithelial cells under hypoxia via AP-1-Nrf2 transcription factor vol.25, pp.3, 2009, https://doi.org/10.1007/s12192-020-01083-3
  48. Bacteroides fragilis Enterotoxin Induces Sulfiredoxin-1 Expression in Intestinal Epithelial Cell Lines Through a Mitogen-Activated Protein Kinases- and Nrf2-Dependent Pathway, Leading to the Suppress vol.21, pp.15, 2009, https://doi.org/10.3390/ijms21155383
  49. Oxidative Stress in Cancer vol.38, pp.2, 2009, https://doi.org/10.1016/j.ccell.2020.06.001
  50. NRF2 and the Ambiguous Consequences of Its Activation during Initiation and the Subsequent Stages of Tumourigenesis vol.12, pp.12, 2009, https://doi.org/10.3390/cancers12123609
  51. Targeting Reactive Oxygen Species Capacity of Tumor Cells with Repurposed Drug as an Anticancer Therapy vol.2021, pp.None, 2009, https://doi.org/10.1155/2021/8532940
  52. Increased oxidative stress and cancer biomarkers in the ventral prostate of older rats submitted to maternal malnutrition vol.523, pp.None, 2021, https://doi.org/10.1016/j.mce.2020.111148
  53. Changes in Glutathione Content in Liver Diseases: An Update vol.10, pp.3, 2009, https://doi.org/10.3390/antiox10030364
  54. Activation of Nrf2 signaling pathway by natural and synthetic chalcones: a therapeutic road map for oxidative stress vol.14, pp.4, 2009, https://doi.org/10.1080/17512433.2021.1901578
  55. The Phenolic Antioxidant 3,5-dihydroxy-4-methoxybenzyl Alcohol (DHMBA) Prevents Enterocyte Cell Death under Oxygen-Dissolving Cold Conditions through Polyphyletic Antioxidant Actions vol.10, pp.9, 2009, https://doi.org/10.3390/jcm10091972