Drug Res (Stuttg) 2023; 73(07): 408-411
DOI: 10.1055/a-2047-3896
Review

AGE RAGE Pathways: Cardiovascular Disease and Oxidative Stress

Neeraj Sharma
1   Department of Pharmacy, Bhagwant University, Ajmer, India
,
Pavan Kumar
2   Ph.D scholar of Department of Pharmacy, Bhagwant University, Ajmer, India
,
Karuna Shanker Shukla
3   Goel Institute of Pharmaceutical Sciences, Uttar Pradesh, Luckow, India
,
Shubhrat Maheshwari
4   Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Rama University, Kanpur, Uttar Pradesh, India
› Author Affiliations

Abstract

It is well established that Advanced Glycation End Products (AGEs) and their receptor (RAGE) are primarily responsible for the development of cardiovascular disease. As a result, diabetic therapy is very interested in therapeutic strategies that can target the AGE-RAGE axis. The majority of the AGE-RAGE inhibitors showed encouraging outcomes in animal experiments, but more information is needed to completely understand their clinical effects. The main mechanism implicated in the aetiology of cardiovascular disease in people with diabetes is oxidative stress and inflammation mediated by AGE-RAGE interaction. Numerous PPAR-agonists have demonstrated favourable outcomes in the treatment of cardio-metabolic illness situations by inhibiting the AGE-RAGE axis. The body’s ubiquitous phenomena of inflammation occur in reaction to environmental stressors such tissue damage, infection by pathogens, or exposure to toxic substances. Rubor (redness), calor (heat), tumour (swelling), colour (pain), and in severe cases, loss of function, are its cardinal symptoms. When exposed, the lungs develop silicotic granulomas with the synthesis of collagen and reticulin fibres. A natural flavonoid called chyrsin has been found to have PPAR-agonist activity as well as antioxidant and anti-inflammatory properties. The RPE insod2+/animals underwent mononuclear phagocyte-induced apoptosis, which was accompanied with decreased superoxide dismutase 2 (SOD2) and increased superoxide generation. Injections of the serine proteinase inhibitor SERPINA3K decreased proinflammatory factor expression in mice with oxygen-induced retinopathy, decreased ROS production, and increased levels of SOD and GSH.



Publication History

Received: 07 February 2023

Accepted: 28 February 2023

Article published online:
12 June 2023

© 2023. Thieme. All rights reserved.

Georg Thieme Verlag
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Münch G, Mayer S, Michaelis J. et al. Influence of advanced glycation end-products and AGE-inhibitors on nucleation-dependent polymerization of β-amyloid peptide. Biochimica et Biophysica Acta (BBA)-Molecular Basis of Disease 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 Radical Biology and Medicine 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. Nature medicine 2012; 18: 153-158
  • 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. Experimental neurology 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 neuropathologica communications 2014; 2: 1-4
  • 6 Khan RS, Fonseca-Kelly Z, Callinan C. et al. SIRT1 activating compounds reduce oxidative stress and prevent cell death in neuronal cells. Frontiers in Cellular Neuroscience 2012; 6: 63
  • 7 Li J, Feng L, Xing Y. et al. Radioprotective and antioxidant effect of resveratrol in hippocampus by activating Sirt1. International journal of molecular sciences 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-1α signaling pathway. BMC complementary and alternative medicine 2012; 12: 1-7
  • 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. Current Alzheimer Research 2013; 10: 290-297
  • 10 Donmez G, Wang D, Cohen DE. et al. RETRACTED: SIRT1 Suppresses β-Amyloid Production by Activating the α-Secretase Gene ADAM10.
  • 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. Journal of Biological Chemistry 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-β1 in rat glomerular messangial cells. Molecular and cellular endocrinology 2015; 399: 178-189
  • 13 Huang K, Huang J, Xie X. et al. Sirt1 resists advanced glycation end products-induced expressions of fibronectin and TGF-β1 by activating the Nrf2/ARE pathway in glomerular mesangial cells. Free Radical Biology and Medicine 2013; 65: 528-540
  • 14 Uribarri J, Cai W, Pyzik R. et al. Suppression of native defense mechanisms, SIRT1 and PPARγ, by dietary glycoxidants precedes disease in adult humans; relevance to lifestyle-engendered chronic diseases. Amino Acids 2014; 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. Chemical research in toxicology 1992; 5: 227-231
  • 16 Olivieri G, Brack C, Müller-Spahn F. et al. Mercury induces cell cytotoxicity and oxidative stress and increases β-amyloid secretion and tau phosphorylation in SHSY5Y neuroblastoma cells. Journal of Neurochemistry 2000; 74: 231-236
  • 17 Sun Q, Hu H, Wang W. et al. Taurine attenuates amyloid β 1–42-induced mitochondrial dysfunction by activating of SIRT1 in SK-N-SH cells. Biochemical and Biophysical Research Communications 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 dose-response
  • 19 Yang H, Chen J, Tang S. et al. New drug R&D of traditional Chinese medicine: Role of data mining approaches. Journal of biological systems 2009; 17: 329-347
  • 20 Xie L, Li J, 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 computational biology 2009; 5: e1000387
  • 21 Goh KI, Cusick ME, Valle D. et al. The human disease network. Proceedings of the National Academy of Sciences 2007; 104: 8685-8690
  • 22 Loscalzo J, Kohane I, Barabasi AL. Human disease classification in the postgenomic era: a complex systems approach to human pathobiology. Molecular systems biology 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, Krull M, Voss N. et al. Molecular mechanistic associations of human diseases. BMC systems biology 2010; 4: 1-9
  • 25 Vidal M, Cusick ME, Barabási AL. Interactome networks and human disease. Cell. 2011; 144: 986-998
  • 26 Hidalgo CA, Blumm N, Barabási AL. et al. A dynamic network approach for the study of human phenotypes. PLoS computational biology 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