Generic placeholder image

Current Neurovascular Research

Editor-in-Chief

ISSN (Print): 1567-2026
ISSN (Online): 1875-5739

Research Article

Keap1 as Target of Genistein on Nrf2 Signaling Pathway Antagonizing Aβ induced Oxidative Damage of Cerebrovascular Endothelial Cells

Author(s): Yuan-Di Xi, Xiao-Ying Li, Ya-Fei Chi, Jing Han, Hong-Rui Li, Xian-Yun Wang, Xuan Wang, Tian-Tian Li, Hui-Yan Yu and Rong Xiao*

Volume 19, Issue 1, 2022

Published on: 15 June, 2022

Page: [73 - 82] Pages: 10

DOI: 10.2174/1567202619666220406100320

Price: $65

Abstract

Background: β-amyloid peptides (Aβ) induced oxidative damage contributes to the pathogenesis of neurodegenerative diseases, and the cerebrovascular system is more vulnerable to oxidative stress. Our earlier study showed a clue that Genistein (Gen) might activate the Nf-E2 related factor 2 (Nrf2) pathway to protect cerebrovascular cells from oxidative damage induced by Aβ, but the specific mechanisms and regulation targets are unclear.

Objective: In this study, the anti-oxidative effects and the possible targets of Gen on regulating the Nrf2 pathway in bEnd.3 cells were investigated. Cells were divided into control, Aβ25-35, Gen, and Gen+Aβ25-35 groups.

Methods: Cell viability, levels of malondialdehyde (MDA), Superoxide Dismutase (SOD) activity, and nitrotyrosine were evaluated. Moreover, mRNA and/or protein expressions of Nrf2 and kelchlike ECH-associated protein 1 (Keap1) were measured. Then we transfected Keap1 over-expressed plasmid into bEnd.3 cells and measured the protein expressions of Nrf2 pathway related factors.

Results: Data showed that Gen could inhibit the over-production of MDA and nitrotyrosine and activate SOD activity. Furthermore, we discovered that Gen could up-regulate Nrf2 mRNA and protein expression while down-regulating Keap1 protein expression. The Keap1 over-expressed plasmid study revealed that the up-regulation of Nrf2 protein expression induced by Gen pretreatment could be blocked by transfection of Keap1 over-expressed plasmid, and the same results were observed in Nrf2 downstream factors.

Conclusion: Gen could alleviate the cerebrovascular cells' oxidative damage induced by Aβ25-35 by regulating the Nrf2 pathway, and Keap1 might be one of the targets of Gen in activating the Nrf2 pathway.

Keywords: Genistein, β-amyloid peptides 25-35, cerebrovascular endothelial cell, Keap1, Nrf2, oxidative damage.

[1]
Bangen KJ, Nation DA, Delano-Wood L, et al. Aggregate effects of vascular risk factors on cerebrovascular changes in autopsy-confirmed Alzheimer’s disease. Alzheimers Dement 2015; 11(4): 394-403.e1.
[http://dx.doi.org/10.1016/j.jalz.2013.12.025] [PMID: 25022538]
[2]
Vaz M, Machireddy N, Irving A, et al. Oxidant-induced cell death and Nrf2-dependent antioxidative response are controlled by Fra-1/AP-1. Mol Cell Biol 2012; 32(9): 1694-709.
[http://dx.doi.org/10.1128/MCB.06390-11] [PMID: 22393254]
[3]
Muresanu DF, Popa-Wagner A, Stan A, Buga AM, Popescu BO. The vascular component of Alzheimer’s disease. Curr Neurovasc Res 2014; 11(2): 168-76.
[http://dx.doi.org/10.2174/1567202611666140408105333] [PMID: 24712646]
[4]
Nagaraju GP, Zafar SF, El-Rayes BF. Pleiotropic effects of genistein in metabolic, inflammatory, and malignant diseases. Nutr Rev 2013; 71(8): 562-72.
[http://dx.doi.org/10.1111/nure.12044] [PMID: 23865800]
[5]
Hsieh HM, Wu WM, Hu ML. Genistein attenuates D-galactose-induced oxidative damage through decreased reactive oxygen species and NF-κB binding activity in neuronal PC12 cells. Life Sci 2011; 88(1-2): 82-8.
[http://dx.doi.org/10.1016/j.lfs.2010.10.021] [PMID: 21056584]
[6]
Petry FDS, Coelho BP, Gaelzer MM, et al. Genistein protects against amyloid-beta-induced toxicity in SH-SY5Y cells by regulation of Akt and Tau phosphorylation. Phytother Res 2020; 34(4): 796-807.
[http://dx.doi.org/10.1002/ptr.6560] [PMID: 31795012]
[7]
Petry FDS, Hoppe JB, Klein CP, et al. Genistein attenuates amyloid-beta-induced cognitive impairment in rats by modulation of hippo-campal synaptotoxicity and hyperphosphorylation of Tau. J Nutr Biochem 2021; 87: 108525.
[http://dx.doi.org/10.1016/j.jnutbio.2020.108525] [PMID: 33065257]
[8]
Ma W, Yuan L, Yu H, et al. Genistein as a neuroprotective antioxidant attenuates redox imbalance induced by beta-amyloid peptides 25-35 in PC12 cells. Int J Dev Neurosci 2010; 28(4): 289-95.
[http://dx.doi.org/10.1016/j.ijdevneu.2010.03.003] [PMID: 20362658]
[9]
Ma WW, Hou CC, Zhou X, et al. Genistein alleviates the mitochondria-targeted DNA damage induced by β-amyloid peptides 25-35 in C6 glioma cells. Neurochem Res 2013; 38(7): 1315-23.
[http://dx.doi.org/10.1007/s11064-013-1019-y] [PMID: 23519932]
[10]
Ding J, Yu HL, Ma WW, et al. Soy isoflavone attenuates brain mitochondrial oxidative stress induced by β-amyloid peptides 1-42 injec-tion in lateral cerebral ventricle. J Neurosci Res 2013; 91(4): 562-7.
[http://dx.doi.org/10.1002/jnr.23163] [PMID: 23239252]
[11]
Gan L, Johnson JA. Oxidative damage and the Nrf2-ARE pathway in neurodegenerative diseases. Biochim Biophys Acta 2014; 1842(8): 1208-18.
[http://dx.doi.org/10.1016/j.bbadis.2013.12.011] [PMID: 24382478]
[12]
Anuranjani BM, Bala M. Concerted action of Nrf2-ARE pathway, MRN complex, HMGB1 and inflammatory cytokines - implication in modification of radiation damage. Redox Biol 2014; 2: 832-46.
[http://dx.doi.org/10.1016/j.redox.2014.02.008] [PMID: 25009785]
[13]
Xi YD, Yu HL, Ding J, et al. Flavonoids protect cerebrovascular endothelial cells through Nrf2 and PI3K from β-amyloid peptide-induced oxidative damage. Curr Neurovasc Res 2012; 9(1): 32-41.
[http://dx.doi.org/10.2174/156720212799297092] [PMID: 22272764]
[14]
Zlokovic BV. Neurodegeneration and the neurovascular unit. Nat Med 2010; 16(12): 1370-1.
[http://dx.doi.org/10.1038/nm1210-1370] [PMID: 21135839]
[15]
Subash S, Essa MM, Al-Asmi A, Al-Adawi S, Vaishnav R. Chronic dietary supplementation of 4% figs on the modification of oxidative stress in Alzheimer’s disease transgenic mouse model. BioMed Res Int 2014; 2014: 546357.
[http://dx.doi.org/10.1155/2014/546357] [PMID: 25050360]
[16]
Ma B, Meng X, Wang J, et al. Notoginsenoside R1 attenuates amyloid-β-induced damage in neurons by inhibiting reactive oxygen species and modulating MAPK activation. Int Immunopharmacol 2014; 22(1): 151-9.
[http://dx.doi.org/10.1016/j.intimp.2014.06.018] [PMID: 24975829]
[17]
He JT, Zhao X, Xu L, Mao CY. Vascular risk factors and Alzheimer’s disease: Blood-brain barrier disruption, metabolic syndromes, and molecular links. J Alzheimers Dis 2020; 73(1): 39-58.
[http://dx.doi.org/10.3233/JAD-190764] [PMID: 31815697]
[18]
Fonseca AC, Moreira PI, Oliveira CR, Cardoso SM, Pinton P, Pereira CF. Amyloid-beta disrupts calcium and redox homeostasis in brain endothelial cells. Mol Neurobiol 2015; 51(2): 610-22.
[http://dx.doi.org/10.1007/s12035-014-8740-7] [PMID: 24833600]
[19]
Javanbakht MH, Sadria R, Djalali M, Derakhshanian H, Hosseinzadeh P, Zarei M. Soy protein and genistein improves renal antioxidant status in experimental nephrotic syndrome. Nefrologia 2014; 34(4): 483-90.
[20]
Gong DK, Liu BH, Tan XH. Genistein prevents cadmium-induced neurotoxic effects through its antioxidant mechanisms. Drug Res (Stuttg) 2015; 65(2): 65-9.
[PMID: 24918346]
[21]
Erba D, Casiraghi MC, Martinez-Conesa C, Goi G, Massaccesi L. Isoflavone supplementation reduces DNA oxidative damage and in-creases O-β-N-acetyl-D-glucosaminidase activity in healthy women. Nutr Res 2012; 32(4): 233-40.
[http://dx.doi.org/10.1016/j.nutres.2012.03.007] [PMID: 22575035]
[22]
Ayala A, Muñoz MF, Argüelles S. Lipid peroxidation: Production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. Oxid Med Cell Longev 2014; 2014: 360438.
[http://dx.doi.org/10.1155/2014/360438] [PMID: 24999379]
[23]
López N, Tormo C, De Blas I, Llinares I, Alom J. Oxidative stress in Alzheimer’s disease and mild cognitive impairment with high sensi-tivity and specificity. J Alzheimers Dis 2013; 33(3): 823-9.
[http://dx.doi.org/10.3233/JAD-2012-121528] [PMID: 23076075]
[24]
Polak-Szabela A, Dziembowska I, Bracha M, Pedrycz-Wieczorska A, Kedziora-Kornatowska K, Kozakiewicz M. The analysis of oxidative stress markers may increase the accuracy of the differential diagnosis of Alzheimer’s disease with and without depression. Clin Interv Aging 2021; 16: 1105-17.
[http://dx.doi.org/10.2147/CIA.S310750] [PMID: 34163154]
[25]
Bagheri M, Joghataei MT, Mohseni S, Roghani M. Genistein ameliorates learning and memory deficits in amyloid β(1-40) rat model of Alzheimer’s disease. Neurobiol Learn Mem 2011; 95(3): 270-6.
[http://dx.doi.org/10.1016/j.nlm.2010.12.001] [PMID: 21144907]
[26]
Radak Z, Zhao Z, Koltai E, Ohno H, Atalay M. Oxygen consumption and usage during physical exercise: The balance between oxidative stress and ROS-dependent adaptive signaling. Antioxid Redox Signal 2013; 18(10): 1208-46.
[http://dx.doi.org/10.1089/ars.2011.4498] [PMID: 22978553]
[27]
Bandookwala M, Sengupta P. 3-Nitrotyrosine: A versatile oxidative stress biomarker for major neurodegenerative diseases. Int J Neurosci 2020; 130(10): 1047-62.
[http://dx.doi.org/10.1080/00207454.2020.1713776] [PMID: 31914343]
[28]
Shi GX, Liu CZ, Wang LP, Guan LP, Li SQ. Biomarkers of oxidative stress in vascular dementia patients. Can J Neurol Sci 2012; 39(1): 65-8.
[http://dx.doi.org/10.1017/S0317167100012701]
[29]
Truran S, Franco DA, Roher AE, et al. Adipose and leptomeningeal arteriole endothelial dysfunction induced by β-amyloid peptide: A practical human model to study Alzheimer’s disease vasculopathy. J Neurosci Methods 2014; 235: 123-9.
[http://dx.doi.org/10.1016/j.jneumeth.2014.06.014] [PMID: 25004204]
[30]
Lim JL, Wilhelmus MM, de Vries HE, Drukarch B, Hoozemans JJ, van Horssen J. Antioxidative defense mechanisms controlled by Nrf2: State-of-the-art and clinical perspectives in neurodegenerative diseases. Arch Toxicol 2014; 88(10): 1773-86.
[http://dx.doi.org/10.1007/s00204-014-1338-z] [PMID: 25164826]
[31]
Zhang W, Feng C, Jiang H. Novel target for treating Alzheimer’s diseases: Crosstalk between the Nrf2 pathway and autophagy. Ageing Res Rev 2021; 65: 101207.
[http://dx.doi.org/10.1016/j.arr.2020.101207] [PMID: 33144123]
[32]
Siow RC, Mann GE. Dietary isoflavones and vascular protection: Activation of cellular antioxidant defenses by SERMs or hormesis? Mol Aspects Med 2010; 31(6): 468-77.
[http://dx.doi.org/10.1016/j.mam.2010.09.003] [PMID: 20837051]
[33]
Kanninen K, Malm TM, Jyrkkänen HK, et al. Nuclear factor erythroid 2-related factor 2 protects against beta amyloid. Mol Cell Neurosci 2008; 39(3): 302-13.
[http://dx.doi.org/10.1016/j.mcn.2008.07.010] [PMID: 18706502]
[34]
Meng Y, Feng R, Yang Z, Liu T, Huo T, Jiang H. Oxidative stress induced by realgar in neurons: p38 MAPK and ERK1/2 perturb autoph-agy and induce the p62-Keap1-Nrf2 feedback loop to activate the Nrf2 signalling pathway. J Ethnopharmacol 2022; 282: 114582.
[http://dx.doi.org/10.1016/j.jep.2021.114582] [PMID: 34492322]
[35]
Uruno A, Matsumaru D, Ryoke R, et al. Nrf2 suppresses oxidative stress and inflammation in App knock-in Alzheimer’s disease model mice. Mol Cell Biol 2020; 40(6): e00467-19.
[http://dx.doi.org/10.1128/MCB.00467-19] [PMID: 31932477]
[36]
Fu Y, Gao J, Li Y, Yang X, Zhang Y. TRIM21 deficiency confers protection from OGD/R-induced oxidative and inflammatory damage in cultured hippocampal neurons through regulation of the Keap1/Nrf2 pathway. Int Immunopharmacol 2022; 103: 108414.
[http://dx.doi.org/10.1016/j.intimp.2021.108414] [PMID: 34929478]
[37]
Ulasov AV, Rosenkranz AA, Georgiev GP, Sobolev AS. Nrf2/Keap1/ARE signaling: Towards specific regulation. Life Sci 2022; 291: 120111.
[http://dx.doi.org/10.1016/j.lfs.2021.120111] [PMID: 34732330]
[38]
Li MY, Dai XH, Yu XP, Zou W, Teng W, Liu P. Scalp acupuncture protects against neuronal ferroptosis by activating the p62-Keap1-Nrf2 pathway in rat models of intracranial haemorrhage. J Mol Neurosci 2022; 72(1): 82-96.
[http://dx.doi.org/10.1007/s12031-021-01890-y]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy