Drug Res (Stuttg) 2023; 73(05): 251-254
DOI: 10.1055/a-2008-7948
Review

AGEs RAGE Pathways: Alzheimer’s Disease

Shubhrat Maheshwari
1   Faculty of Pharmaceutical Sciences, Rama University, Kanpur, Uttar Pradesh, India
› Author Affiliations

Abstract

Neurofibrillary tangles and plaques containing tau serve as the biological markers for Alzheimer disease (AD) and pathogenesis is widely believed to be driven by the production and deposition of the β-amyloid peptide (Aβ). The β-amyloid peptide (Aβ) that results from the modification of the amyloid precursor protein (APP) by builds up as amyloid deposits in neuronal cells. Thus, a protein misfolding process is involved in the production of amyloid. In a native, aqueous buffer, amyloid fibrils are usually exceedingly stable and nearly insoluble. Although amyloid is essentially a foreign substance made of self-proteins, the immune system has difficulty identifying and eliminating it as such for unknown reasons. While the amyloidal deposit may have a direct role in the disease mechanism in some disease states involving amyloidal deposition, this is not always the case. Current research has shown that PS1 (presenilin 1) and BACE (beta-site APP-cleaving enzyme) have – and -secretase activity that increases β-amyloid peptide (Aβ). Wealth of data has shown that oxidative stress and AD are closely connected that causes the death of neuronal cells by producing reactive oxygen species (ROS). Additionally, it has been demonstrated that advanced glycation end products (AGEs) and β-amyloidal peptide (Aβ) together increase neurotoxicity. The objective of this review is to compile the most recent and intriguing data of AGEs and receptor for advanced glycation end products (RAGE) pathways which are responsible for AD.



Publication History

Received: 20 December 2022

Accepted: 03 January 2023

Article published online:
20 March 2023

© 2023. Thieme. All rights reserved.

Georg Thieme Verlag
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Munch G, Mayer S, Michaelis J. et al. Influence of advanced glycation end-products and AGE-inhibitors on nucleation-dependent polymerization of beta-amyloid peptide. Biochim Biophys Acta 1997; 1360: 17-29
  • 2 Ko SY, Lin YP, Lin YS. et al. Advanced glycation end products enhance amyloid precursor protein expression by inducing reactive oxygen species. Free Radic Biol Med 2010; 49: 474-480
  • 3 Jiang M, Wang J, Fu J. et al. Neuroprotective role of Sirt1 in mammalian models of Huntington's disease through activation of multiple Sirt1 targets. Nat Med 18:153–158. 31. Kim D, Nguyen MD, Dobbin MM, Fischer A, Sananbenesi F, Rodgers JT, et al. (2007) SIRT1 deacetylase protects against neurodegeneration in models for Alzheimer's disease and amyotrophic lateral sclerosis. EMBO J 2012; 26: 3169-3179
  • 4 Zhao Y, Luo P, Guo Q. et al. Interactions between SIRT1 and MAPK/ ERK regulate neuronal apoptosis induced by traumatic brain injury in vitro and in vivo. Exp Neurol 2012; 237: 489-498
  • 5 Khan RS, Dine K, Das Sarma J. et al. SIRT1 activating compounds reduce oxidative stress mediated neuronal loss in viral induced CNS demyelinating disease. Acta Neuropathol Commun 2014; 2: 3
  • 6 Khan RS, Fonseca-Kelly Z, Callinan C. et al. SIRT1 activating compounds reduce oxidative stress and prevent cell death in neuronal cells. Front Cell Neurosci 2012; 6: 63
  • 7 Li J, Feng L, Xing Y. et al. Radioprotective and antioxidant effect of resveratrol in hippocampus by activating Sirt1. Int J Mol Sci 2014; 15: 5928-5939
  • 8 Ye Q, Ye L, Xu X. et al. Epigallocatechin-3-gallate suppresses 1- methyl-4-phenyl-pyridine-induced oxidative stress in PC12 cells via the SIRT1/PGC-1alpha signaling pathway. BMC Complement Altern Med 2012; 12: 82
  • 9 Aso E, Semakova J, Joda L. et al. Triheptanoin supplementation to ketogenic diet curbs cognitive impairment in APP/PS1 mice used as a model of familial Alzheimer's disease. Curr Alzheimer Res 2013; 10: 290-297
  • 10 Donmez G, Wang D, Cohen DE. et al. SIRT1 suppresses beta-amyloid production by activating the alpha-secretase gene ADAM10. Cell 2010; 142: 320-332
  • 11 Qin W, Yang T, Ho L. et al. Neuronal SIRT1 activation as a novel mechanism underlying the prevention of Alzheimer disease amyloid neuropathology by calorie restriction. J Biol Chem 2006; 281: 21745-21754
  • 12 Huang K, Chen C, Hao J. et al. Polydatin promotes Nrf2-ARE anti-oxidative pathway through activating Sirt1 to resist AGEs-induced upregulation of fibronetin and transforming growth factor-beta1 in rat glomerular messangial cells. Mol Cell Endocrinol 2014; 399: 178-189
  • 13 Huang K, Huang J, Xie X. et al. Sirt1 resists advanced glycation end products-induced expressions of fibronectin and TGF-beta1 by activating the Nrf2/ARE pathway in glomerular mesangial cells. Free Radic Biol Med 2013; 65: 528-540
  • 14 Uribarri J, Cai W, Pyzik R. et al. Suppression of native defense mechanisms, SIRT1 and PPARgamma, by dietary glycoxidants precedes disease in adult humans; relevance to lifestyle-engendered chronic diseases. Amino Acids 2013; 46: 301-309
  • 15 LeBel CP, Ischiropoulos H, Bondy SC. Evaluation of the probe 2',7'-dichlorofluorescin as an indicator of reactive oxygen species formation and oxidative stress. Chem Res Toxicol 1992; 5: 227-231
  • 16 Olivieri G, Brack C, Muller-Spahn F. et al. Mercury induces cell cytotoxicity and oxidative stress and increases beta-amyloid secretion and tau phosphorylation in SHSY5Y neuroblastoma cells. J Neurochem 2000; 74: 231-236
  • 17 Sun Q, Hu H, Wang W. et al. Taurine attenuates amyloid beta 1-42-induced mitochondrial dysfunction by activating of SIRT1 in SK-N-SH cells. Biochem Biophys Res Commun 2014; 447: 485-489
  • 18 Yuyun X, Jinjun Q, Minfang X. et al. Effects of Low Concentrations of Rotenone upon Mitohormesis in SH-SY5Y Cells. Dose Response 2013; 11: 270-280
  • 19 Yang Y. et al. Target discovery from data mining approaches. Drug Discov. Today 2009; 14: 147-154
  • 20 Xie L. et al. Drug discovery using chemical systems biology: identification of the protein – ligand binding network to explain the side effects of CETP inhibitors. PLoS Comput Biol 2009; 5: E1000387
  • 21 Goh KI. et al. The human disease network. Proc Natl Acad Sci USA 2007; 104: 8685-8690
  • 22 Loscalzo J. et al. Human disease classification in the postgenomic era: a complex systems approach to human pathobiology. Mol Syst Biol 2007; 3: 124
  • 23 Hu G, Agarwal P.. Human disease-drug network based on genomic expression profiles. PLoS One 2009; 4: E6536
  • 24 Stegmaier P. et al. Molecular mechanistic associations of human diseases. BMC Syst Biol 2010; 4: 124
  • 25 Vidal M. et al. Interactome networks and human disease. Cell 2011; 144: 986-998
  • 26 Hidalgo CA. et al. A dynamic network approach for the study of human phenotypes. PLoS Comput Biol 2009; 5: E1000353
  • 27 Li GW, Xie XS.. Central dogma at the single-molecule level in living cells. Nature 2011; 475: 308-315
  • 28 Fenimore PW, Frauenfelder H, McMahon BH. et al. Slaving: solvent fluctuations dominate protein dynamics and functions. Proceedings of the National Academy of Sciences 2002; 99: 16047-16051
  • 29 Fitzpatrick AW, Debelouchina GT, Bayro MJ. et al. Atomic structure and hierarchical assembly of a cross-β amyloid fibril. Proceedings of the National Academy of Sciences 2013; 110: 5468-5473
  • 30 Jimenez JL, Guijarro JI, Orlova E. et al. Cryo-electron microscopy structure of an SH3 amyloid fibril and model of the molecular packing. The EMBO journal 1999; 18: 815-821
  • 31 Hussain R, Zubair H, Pursell S. et al. Neurodegenerative diseases: Regenerative mechanisms and novel therapeutic approaches. Brain sciences 2018; 8: 177
  • 32 Hussain R, Zubair H, Pursell S. et al. Neurodegenerative diseases: Regenerative mechanisms and novel therapeutic approaches. Brain sciences 2018; 8: 177
  • 33 Cicero CE, Mostile G, Vasta R. et al. Metals and neurodegenerative diseases. A systematic review. Environmental research 2017; 159: 82-94
  • 34 Gardner RC, Yaffe K.. Epidemiology of mild traumatic brain injury and neurodegenerative disease. Molecular and Cellular Neuroscience 2015; 66: 75-80
  • 35 Rezazadeh M, Khorrami A, Yeghaneh T. et al. Genetic factors affecting late-onset Alzheimer’s disease susceptibility. Neuromolecular medicine 2016; 18: 37-49