Understanding the genetic, molecular, and cellular basis of ageing as the biggest risk factor of Alzheimer's disease

Authors

  • Meena Yadav Department of Zoology, Maitreyi College, University of Delhi, Delhi, India
  • Prama Pandey Department of Zoology, Gargi College, University of Delhi, Delhi, India
  • Poonam Sharma Department of Zoology, Gargi College, University of Delhi, Delhi, India

Keywords:

Alzheimer's disease, Ageing, Dementia, Neurodegenerative, Telomeres, DNA methylation

Abstract

Alzheimer’s disease (AD) is one of the leading causes of dementia. The disease is characterized by atrophy of brain tissue, with major physiological, molecular, and anatomical changes being observed in the hippocampus and entorhinal region of the temporal lobe. The risk of developing this disease increases with advancing age. Ageing is a chronological phenomenon wherein a considerable decline is observed in physiological functions due to the complex interplay of various exogenous and endogenous factors such as genetic construction, elevated levels of ROS, decrease in the telomerase activity, and epigenetic factors such as methylation of DNA, histone modification etc. The physiological and molecular changes in an ageing person especially in neurons overlap considerably with those observed during the progression of AD. This article highlights various factors responsible for ageing as well as AD with the latest review of literature. Understanding the factors that bring about the fated changes and how they are associated with the progression of disease can open new doors to bring about better treatment options and help cure an otherwise incurable disease.

DOI: http://dx.doi.org/10.5281/zenodo.6323799

References

World Health Organization. https://www.who.int/news-room/fact-sheets/detail/dementia.

Breijyeh Z, Karaman R. Comprehensive Review on Alzheimer's Disease: Causes and Treatment. Molecules. 2020; 25(24): 5789.

Pandey P, Sharma P. Analysis of Early Onset of Alzheimer’s Disease Genes: Disease Causing and Risk Factors. Eur J Biol Res. 2021; 11: 251-259.

Jarmolowicz AI, Chen HY, Panegyres PK. The patterns of inheritance in early-onset dementia: Alzheimer's disease and frontotemporal dementia. Am J Alzheimer's Dis Other Demen. 2015; 30(3): 299-306.

Xia X, Jiang Q, McDermott J, Han JJ. Aging and Alzheimer's disease: Comparison and associations from molecular to system level. Aging Cell. 2018; 17(5): e12802.

Masters CL. Major risk factors for Alzheimer's disease: age and genetics. Lancet Neurol. 2020; 19(6): 475-476.

Alzheimer’s Association Report. Alzheimer's disease facts and figures. Alzheimer's Dement. 2020; 16(3): 391-460.

Fandos N, Perez-Grijalba V, Pesini P, Olmos S, Bossa M, Villemagne VL, et al. Plasma amyloid beta 42/40 ratios as biomarkers for amyloid beta cerebral deposition in cognitively normal individuals. Alzheimers Dement. 2017; 8: 179-187.

Shi J, Sabbagh MN, Vellas B. Alzheimer's disease beyond amyloid: strategies for future therapeutic interventions. BMJ. 2020; 371: m3684.

Guerreiro R, Bras J. The age factor in Alzheimer's disease. Genome Med. 2015; 7: 106.

DiLoreto R, Murphy CT. The cell biology of aging. Mol Biol Cell. 2015; 26(25): 4524-4531.

Cai Z, Yan LJ, Ratka A. Telomere shortening and Alzheimer's disease. Neuromolecular Med. 2013; 15(1): 25-48.

Fani L, Hilal S, Sedaghat S, Broer L, Licher S, Arp PP, et al. Telomere Length and the Risk of Alzheimer's Disease: The Rotterdam Study. J Alzheimer's Dis. 2020; 73(2): 707-714.

Tanokashira D, Fukuokaya W, Taguchi A. Involvement of insulin receptor substrates in cognitive impairment and Alzheimer's disease. Neural Regen Res. 2019; 14(8): 1330-1334.

Lorenzini A, Salmon AB, Lerner C, Torres C, Ikeno Y, Motch S, et al. Mice producing reduced levels of insulin-like growth factor type 1 display an increase in maximum, but not mean, life span. J Gerontol A Biol Sci Med Sci. 2014; 69(4): 410-419.

Junnila RK, List EO, Berryman DE, Murrey JW, Kopchick JJ. The GH/IGF-1 axis in ageing and longevity. Nat Rev Endocrinol. 2013; 9(6): 366-376.

Hipp MS, Kasturi P, Hartl FU. The proteostasis network and its decline in ageing. Nat Rev Mol Cell Biol. 2019; 20(7): 421-435.

Labbadia J, Morimoto RI. The biology of proteostasis in aging and disease. Annu Rev Biochem. 2015; 84: 435-464.

Korovila I, Hugo M, Castro JP, Weber D, Höhn A, Grune T, Jung T. Proteostasis, oxidative stress and aging. Redox Biol. 2017; 13: 550-567.

Mathew R, Bhadra MP, Bhadra U. Insulin/insulin-like growth factor-1 signaling (IIS) based regulation of lifespan across species. Biogerontology. 2017; 18(1): 35-53.

Barbosa MC, Grosso RA, Fader CM. Hallmarks of Aging: An Autophagic Perspective. Front Endocrinol. 2019; 9: 790.

Sun N, Youle RJ, Finkel T. The Mitochondrial Basis of Aging. Mol Cell. 2016; 61(5): 654-666.

Kounakis K, Tavernarakis N. The Cytoskeleton as a Modulator of Aging and Neurodegeneration. Adv Exp Med Biol. 2019; 1178: 227-245.

Lai WF, Wong WT. Roles of the actin cytoskeleton in aging and age-associated diseases. Ageing Res Rev. 2020; 58: 101021.

Centre for Disease Control and Prevention. https://www.cdc.gov/aging/aginginfo/alzheimers.htm (Accessed on May 20, 2021).

Shinohara M, Kanekiyo T, Tachibana M, Kurti A, Shinohara M, Fu Y, et al. APOE2 is associated with longevity independent of Alzheimer's disease. eLife. 2020; 9: e62199.

Gharibyan AL, Islam T, Pettersson N, Golchin SA, Lundgren J, Johansson G, et al. Apolipoprotein E Interferes with IAPP Aggregation and Protects Pericytes from IAPP-Induced Toxicity. Biomol. 2020; 10(1): 134.

Kenyon CJ. The genetics of ageing. Nature. 2010; 464: 504-512.

Guo T, Zhang D, Zeng Y, Huang TY, Xu H, Zhao Y. Molecular and cellular mechanisms underlying the pathogenesis of Alzheimer’s disease. Mol Neurodegener. 2020; ID 40.

Lambert JC, Ibrahim-Verbaas CA, Harold D, Naj AC, Sims R, Bellenguez C, et al. Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer’s disease. Nat Genet. 2013; 45(12): 1452-1458.

Wirz KT, Keitel S, Swaab DF, Verhaagen J, Bossers K. Early molecular changes in Alzheimer disease: can we catch the disease in its presymptomatic phase? J Alzheimers Dis. 2014; 38(4): 719-740.

Liu Y, Weng W, Gao R, Liu Y. New Insights for Cellular and Molecular Mechanisms of Aging and Aging-Related Diseases: Herbal Medicine as Potential Therapeutic Approach. Oxid Med Cell Longev. 2019; ID 4598167.

Mueed Z, Tandon P, Maurya SK, Deval R, Kamal MA, Poddar NK. Tau and mTOR: The Hotspots for Multifarious Diseases in Alzheimer's Development. Front Neurosci. 2019; 12: 1017.

Siddappaji KK, Gopal S. Molecular mechanisms in Alzheimer's disease and the impact of physical exercise with advancements in therapeutic approaches. AIMS Neurosci. 2021; 8(3): 357-389.

Burrinha T, Martinsson I, Gomes R, Terrasso AP, Gouras GK, Almeida CG. Upregulation of APP endocytosis by neuronal aging drives amyloid- dependent synapse loss. J Cell Sci. 2021; 134(9): jcs255752.

Angelova DM, Brown DR. Microglia and the aging brain: are senescent microglia the key to neurodegeneration? J Neurochem. 2019; 151(6): 676-688.

Harrison TM, Maass A, Adams JN, Du R, Baker SL, Jagust WJ. Tau deposition is associated with functional isolation of the hippocampus in aging. Nat Commun. 2019; 10: 4900.

Zhao Y, Zeng CY, Li XH, Yang TT, Kuang X, Du JR. Klotho overexpression improves amyloid-β clearance and cognition in the APP/PS1 mouse model of Alzheimer's disease. Aging Cell. 2020; 19(10): e13239.

Alapin JM, Dines M, Lamprecht R. EphB2 receptor forward signaling is needed for normal long-term memory formation in aged mice. Neurobiol Aging. 2020; 86: 11-15.

Han S, Brunet A. Histone methylation makes its mark on longevity. Trends Cell Biol. 2012; 22: 42-49.

Khan SS, Singer BD, Vaughan DE. Molecular and physiological manifestations and measurement of aging in humans. Aging Cell. 2017; 16(4): 624-633.

Koga H, Kaushik S, Cuervo AM. Protein homeostasis and aging: the importance of exquisite quality control. Ageing Res Rev. 2011; 10: 205-215.

Santra M, Dill KA, de Graff AMR. Proteostasis collapse is a driver of cell aging and death. PNAS. 2019; 116(44): 22173-22178.

Calamini B, Silva MC, Madoux F, Hutt DM, Khanna S, Chalfant MA, et al. Small‐molecule proteostasis regulators for protein conformational diseases. Nat Chem Biol. 2012; 8: 185-196.

Tomaru U, Takahashi S, Ishizu A, Miyatake Y, Gohda A, Suzuki S, et al. Decreased proteasomal activity causes age‐related phenotypes and promotes the development of metabolic abnormalities. Am J Pathol. 2012; 180: 963- 972.

Wang W, Zhao F, Ma X, Perry G, Zhu X. Mitochondria dysfunction in the pathogenesis of Alzheimer's disease: recent advances. Mol Neurodegener. 2020; 15(1): 30.

Houtkooper RH, Williams RW, Auwerx J. Metabolic networks of longevity. Cell. 2010; 142(1): 9-14.

Barzilai N, Huffman DM, Muzumdar RH, Bartke A. The critical role of metabolic pathways in aging. Diabetes. 2012; 61: 1315-1322.

Green DR, Galluzzi L, Kroemer G. Mitochondria and the autophagy‐inflammation‐cell death axis in organismal aging. Science. 2011; 333: 1109-1112.

Cosacak MI, Bhattarai P, Kizil C. Alzheimer’s disease, neural stem cells and neurogenesis: cellular phase at single-cell level. Neural Regen Res. 2020; 15: 824-827.

Kuilman T, Peeper DS. Senescence‐messaging secretome: SMS‐ing cellular stress. Nat Rev Cancer. 2009; 9: 81-94.

Ozcan S, Alessio N, Acar MB, Mert E, Omerli F, Peluso G, Galderisi U. Unbiased analysis of senescence associated secretory phenotype (SASP) to identify common components following different genotoxic stresses. Aging. 2016; 8: 1316-1329.

Cullen NC, Mälarstig AN, Stomrud E, Hansson O, Mattsson-Carlgren N. Accelerated inflammatory aging in Alzheimer’s disease and its relation to amyloid, tau, and cognition. Sci Rep. 2021; 11: 1965.

Checa J, Aran JM. Reactive Oxygen species: Drivers of physiological and pathological processes. J Inflamm Res. 2020; 13: 1057-1073.

Viswanathan A, Greenberg SM. Cerebral amyloid angiopathy in the elderly. Ann Neurol. 2011; 70(6): 871-880.

Fyfe I. Brain waste clearance reduced by ageing. Nature Rev Neurol. 2020; 16: 128.

Ciccarone F, Tagliatesta S, Caiafa P, Zampieri M. DNA methylation dynamics in aging: how far are we from understanding the mechanisms? Mech Ageing Dev. 2018; 174: 3-17.

Smith AD, Refsum H, Bottiglieri T, Fenech M, Hooshmand B, McCaddon A, et al. Homocysteine and Dementia: An International Consensus Statement. J Alzheimer's Dis. 2018; 62(2): 561-570.

Mastroeni D, McKee A, Grover A, Rogers J, Coleman PD. Epigenetic differences in cortical neurons from a pair of monozygotic twins discordant for Alzheimer's disease. PloS One. 2009; 4(8): e6617.

Coppieters N, Dieriks BV, Lill C, Faull RL, Curtis MA, Dragunow M. Global changes in DNA methylation and hydroxymethylation in Alzheimer's disease human brain. Neurobiol Aging. 2014; 35(6): 1334-1344.

Lashley T, Gami P, Valizadeh N, Li A, Revesz T, Balazs R. Alterations in global DNA methylation and hydroxymethylation are not detected in Alzheimer's disease. Neuropathol Appl Neurobiol. 2015; 41(4): 497-506.

Phipps AJ, Vickers JC, Taberlay PC, Woodhouse A. Neurofilament-labeled pyramidal neurons and astrocytes are deficient in DNA methylation marks in Alzheimer's disease. Neurobiol Aging. 2016; 45: 30-42.

Rao JS, Keleshian VL, Klein S, Rapoport SI. Epigenetic modifications in frontal cortex from Alzheimer's disease and bipolar disorder patients. Transl Psychiatry. 2012; 2(7): e132.

Barrachina M, Ferrer I. DNA methylation of Alzheimer disease and tauopathy-related genes in postmortem brain. J Neuropathol Exp Neurol. 2009; 68(8): 880-891.

Iwata A, Nagata K, Hatsuta H, Takuma H, Bundo M, Iwamoto K, et al. Altered CpG methylation in sporadic Alzheimer's disease is associated with APP and MAPT dysregulation. Hum Mol Genet. 2014; 23(3): 648-656.

Yi SJ, Kim K. New Insights into the Role of Histone Changes in Aging. Int J Mol Sci. 2020; 21(21): 8241.

O'Brien J, Hayder H, Zayed Y, Peng C. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front Endocrinol. 2018; 9: 402.

Martilla S, Chatsirisupachai K, Palmerm D, de Magalhaes JP. Ageing-associated changes in the expression of lncRNAs in human tissues reflect a transcriptional modulation in ageing pathways. Mech Ageing Dev. 2020; 185: 111-177.

Siedlecki-Wullich D, Miñano-Molina AJ, Rodríguez-Álvarez J. microRNAs as Early Biomarkers of Alzheimer's Disease: A Synaptic Perspective. Cells. 2021; 10(1): 113.

Peters R. Ageing and the brain. Postgraduate Med J. 2006; 82(964): 84-88.

Nicaise AM, Willis CM, Crocker SJ, Pluchino S. Stem Cells of the Aging Brain. Front Aging Neurosci. 2020; 12: 247.

Hollands C, Bartolotti N, Lazarov O. Alzheimer's Disease and Hippocampal Adult Neurogenesis; Exploring Shared Mechanisms. Front Neurosci. 2016; 10: 178.

Miranda M, Morici JF, Zanoni MB, Bekinschtein P. Brain-Derived Neurotrophic Factor: A Key Molecule for Memory in the Healthy and the Pathological Brain. Front Cell Neurosci. 2019; 13: 363.

Verkerke M, Hol EM, Middeldorp J. Physiological and Pathological Ageing of Astrocytes in the Human Brain. Neurochem Res. 2021; 46(10): 2662-2675.

Mattson MP, Arumugam TV. Hallmarks of Brain Aging: Adaptive and Pathological Modification by Metabolic States. Cell Metabol. 2018; 27(6): 1176-1199.

Yin F, Sancheti H, Patil I, Cadenas E. Energy metabolism and inflammation in brain aging and Alzheimer's disease. Free Radic Biol Med. 2016; 100: 108-122.

Rea IM, Gibson DS, McGilligan V, McNerlan SE, Alexander HD, Ross OA. Age and Age-Related Diseases: Role of Inflammation Triggers and Cytokines. Front Immunol. 2018; 9: 586.

Stamouli EC, Politis AM. [Pro-inflammatory cytokines in Alzheimer's disease]. Psychiatriki. 2016; 27(4): 264-275.

Su F, Bai F, Zhang Z. Inflammatory Cytokines and Alzheimer's Disease: A Review from the Perspective of Genetic Polymorphisms. Neurosci Bull. 2016; 32(5): 469-480.

Downloads

Published

03/02/2022

How to Cite

Yadav , M. ., Pandey , P. ., & Sharma , P. . (2022). Understanding the genetic, molecular, and cellular basis of ageing as the biggest risk factor of Alzheimer’s disease. European Journal of Biological Research, 12(1), 62–76. Retrieved from http://jbrodka.com/index.php/ejbr/article/view/8

Issue

Section

Review Articles