Reappraisal of the Concept of Accelerated Aging in Neurodegeneration and Beyond
Abstract
:1. Introduction
2. Biomolecular Aspects of Aging
3. Aging of Organs and Systems beyond Neurodegeneration
4. Limitations of the AA Concept
5. Recommendations for Further Development and Improvement of AA Concept
5.1. Statistical Models
- ○
- Sample size. The number of individuals in the study can affect the statistical power of the analysis, and larger sample sizes generally provide more robust results.
- ○
- Biomarker types. The choice of biomarkers can impact the diagnostic model used, as different types of biomarkers may require different statistical analyses.
- ○
- Age range. The age range of the study population can influence the types of biomarkers identified, as some biomarkers may be more prevalent in certain age groups.
- ○
- Data normalization. Normalization of the data is critical to ensure that data collection or processing differences do not affect the analysis.
- ○
- Statistical methods. The choice of statistical methods used can impact the sensitivity and specificity of the analysis, and different methods may be more appropriate for different types of data.
5.2. Molecular Clocks
5.3. Single-Cell Epigenomics
5.4. New Epigenetic Biomarkers
5.5. Consideration of Ageotypes
5.6. Genetic Predisposition to AA
5.7. Application of AA Animal Models
6. Conclusions
- i.
- The concept of an increased rate of age-related changes has certain weaknesses and limitations that are considered in the current review. In particular, so far, no unified methodology and terminology has been established in the field. The studies that justify the AA concept have too low sample sizes. Some age-related changes appear to be reversible under certain conditions.
- ii.
- Aging MBM help to estimate the aging rate increase due to a developed pathology or the exhaustion of individual reserves. A large variety of MBM candidates in different combinations can be associated with the aging brain; however, their validation, clinical interpretation and use in disease subtyping remain a challenge.
- iii.
- Activation of the regenerative mechanisms, and restoring metabolic and energy molecular reserves with novel therapeutic options could be potential ways to decelerate aging in the CNS. For example, sex hormone replacement, antioxidant-based and target therapy, and environmental and lifestyle factors’ improvement may delay ND. Future longitudinal studies could provide clinics and society with more options to prevent AA and slow the aging rate.
7. Afterword: Aging Science History and Theories
Author Contributions
Funding
Conflicts of Interest
Abbreviations
5-mC | 5-methylcytosine |
AA | accelerated aging |
AAG | gene related to accelerated aging |
AD | Alzheimer’s disease |
BA | biological age |
BM | biomarker |
circRNA | circular RNA |
HC | healthy control |
lncRNA | long non-coding RNA |
MBM | molecular biomarkers |
MCI | mild cognitive impairment |
miRNA | microRNA |
ncRNA | non-coding RNA |
ND | neurodegeneration |
NDG | gene that predispose to neurodegeneration |
NV | neurovascular |
NVU | neurovascular unit |
PD | Parkinson’s disease |
References
- Isaev, N.K.; Stelmashook, E.V.; Genrikhs, E.E. Neurogenesis and brain aging. Rev. Neurosci. 2019, 30, 573–580. [Google Scholar] [CrossRef] [PubMed]
- Brivio, P.; Paladini, M.S.; Racagni, G.; Riva, M.A.; Calabrese, F.; Molteni, R. From healthy aging to frailty: In search of the underlying mechanisms. Curr. Med. Chem. 2019, 26, 3685–3701. [Google Scholar] [CrossRef] [PubMed]
- Feltes, B.C.; de Faria Poloni, J.; Bonatto, D. Development and aging: Two opposite but complementary phenomena. Aging Health-A Syst. Biol. Perspect. 2015, 40, 74–84. [Google Scholar]
- Bogeska, R.; Mikecin, A.M.; Kaschutnig, P.; Fawaz, M.; Büchler-Schäff, M.; Le, D.; Ganuza, M.; Vollmer, A.; Paffenholz, S.V.; Asada, N.; et al. Inflammatory exposure drives long-lived impairment of hematopoietic stem cell self-renewal activity and accelerated aging. Cell Stem Cell 2022, 29, 1273–1284. [Google Scholar] [CrossRef] [PubMed]
- Adelman, E.R.; Figueroa, M.E. Human hematopoiesis: Aging and leukemogenic risk. Curr. Opin. Hematol. 2021, 28, 57. [Google Scholar] [CrossRef]
- Hooten, N.N.; Pacheco, N.L.; Smith, J.T.; Evans, M.K. The accelerated aging phenotype: The role of race and social determinants of health on aging. Ageing Res. Rev. 2022, 73, 101536. [Google Scholar] [CrossRef]
- Forrester, S.N.; Zmora, R.; Schreiner, P.J.; Jacobs, D.R., Jr.; Roger, V.L.; Thorpe, R.J., Jr.; Kiefe, C.I. Accelerated aging: A marker for social factors resulting in cardiovascular events? SSM-Popul. Health 2021, 13, 100733. [Google Scholar] [CrossRef]
- Hamczyk, M.R.; Nevado, R.M.; Barettino, A.; Fuster, V.; Andres, V. Biological versus chronological aging: Jacc focus seminar. J. Am. Coll. Cardiol. 2020, 75, 919–930. [Google Scholar] [CrossRef]
- Vaquer-Alicea, J.; Diamond, M.I. Propagation of protein aggregation in neurodegenerative diseases. Annu. Rev. Biochem. 2019, 88, 785–810. [Google Scholar] [CrossRef]
- Armada-Moreira, A.; Gomes, J.I.; Pina, C.C.; Savchak, O.K.; Gonçalves-Ribeiro, J.; Rei, N.; Pinto, S.; Morais, T.P.; Martins, R.S.; Ribeiro, F.F.; et al. Going the extra (synaptic) mile: Excitotoxicity as the road toward neurodegenerative diseases. Front. Cell. Neurosci. 2020, 14, 90. [Google Scholar] [CrossRef]
- Mehta, A.; Prabhakar, M.; Kumar, P.; Deshmukh, R.; Sharma, P. Excitotoxicity: Bridge to various triggers in neurodegenerative disorders. Eur. J. Pharmacol. 2013, 698, 6–18. [Google Scholar] [CrossRef] [PubMed]
- Margolick, J.B.; Ferrucci, L. Accelerating aging research: How can we measure the rate of biologic aging? Exp. Gerontol. 2015, 64, 78–80. [Google Scholar] [CrossRef] [PubMed]
- Melzer, D.; Pilling, L.C.; Ferrucci, L. The genetics of human ageing. Nat. Rev. Genet. 2020, 21, 88–101. [Google Scholar] [CrossRef] [PubMed]
- Miller, M.W.; Sadeh, N. Traumatic stress, oxidative stress and post-traumatic stress disorder: Neurodegeneration and the accelerated-aging hypothesis. Mol. Psychiatry 2014, 19, 1156–1162. [Google Scholar] [CrossRef] [PubMed]
- Ghosh, C.; De, A. Basics of aging theories and disease related aging-an overview. PharmaTutor 2017, 5, 16–23. [Google Scholar]
- Wadhwa, R.; Gupta, R.; Maurya, P.K. Oxidative stress and accelerated aging in neurodegenerative and neuropsychiatric disorder. Curr. Pharm. Des. 2018, 24, 4711–4725. [Google Scholar] [CrossRef]
- Bersani, F.S.; Mellon, S.H.; Reus, V.I.; Wolkowitz, O.M. Accelerated aging in serious mental disorders. Curr. Opin. Psychiatry 2019, 32, 381. [Google Scholar] [CrossRef]
- Hou, Y.; Dan, X.; Babbar, M.; Wei, Y.; Hasselbalch, S.G.; Croteau, D.L.; Bohr, V.A. Ageing as a risk factor for neurodegenerative disease. Nat. Rev. Neurol. 2019, 15, 565–581. [Google Scholar] [CrossRef]
- Wang, X.; Ma, Z.; Cheng, J.; Lv, Z. A genetic program theory of aging using an RNA population model. Ageing Res. Rev. 2014, 13, 46–54. [Google Scholar] [CrossRef]
- Kovacs, G.G. Concepts and classification of neurodegenerative diseases. Handb. Clin. Neurol. 2018, 145, 301–307. [Google Scholar]
- Sanz, A.; Stefanatos, R.K. The mitochondrial free radical theory of aging: A critical view. Curr. Aging Sci. 2008, 1, 10–21. [Google Scholar] [CrossRef] [PubMed]
- Libertini, G.; Shubernetskaya, O.; Corbi, G.; Ferrara, N. Is evidence supporting the subtelomere–telomere theory of aging? Biochemistry 2021, 86, 1526–1539. [Google Scholar] [CrossRef]
- Xie, L.; Wu, S.; He, R.; Li, S.; Lai, X.; Wang, Z. Identification of epigenetic dysregulation gene markers and immune landscape in kidney renal clear cell carcinoma by comprehensive genomic analysis. Front. Immunol. 2022, 13, 901662. [Google Scholar] [CrossRef] [PubMed]
- Růžička, M.; Kulhánek, P.; Radová, L.; Čechová, A.; Špačková, N.; Fajkusová, L.; Réblová, K. Dna mutation motifs in the genes associated with inherited diseases. PLoS ONE 2017, 12, e0182377. [Google Scholar] [CrossRef] [PubMed]
- Korb, M.K.; Kimonis, V.E.; Mozaffar, T. Multisystem proteinopathy: Where myopathy and motor neuron disease converge. Muscle Nerve 2021, 63, 442–454. [Google Scholar] [CrossRef]
- Barja, G. The mitochondrial free radical theory of aging. Prog. Mol. Biol. Transl. Sci. 2014, 127, 1–27. [Google Scholar]
- Amorim, J.A.; Coppotelli, G.; Rolo, A.P.; Palmeira, C.M.; Ross, J.M.; Sinclair, D.A. Mitochondrial and metabolic dysfunction in ageing and age-related diseases. Nat. Rev. Endocrinol. 2022, 18, 243–258. [Google Scholar] [CrossRef]
- Esmaeili, Y.; Yarjanli, Z.; Pakniya, F.; Bidram, E.; Łos, M.J.; Eshraghi, M.; Klionsky, D.J.; Ghavami, S.; Zarrabi, A. Targeting autophagy, oxidative stress, and er stress for neurodegenerative diseases treatment. J. Control. Release 2022, 345, 147–175. [Google Scholar] [CrossRef]
- Pomatto, L.C.; Davies, K.J. Adaptive homeostasis and the free radical theory of ageing. Free Radic. Biol. Med. 2018, 124, 420–430. [Google Scholar] [CrossRef]
- Simpson, D.J.; Chandra, T. Epigenetic age prediction. Aging Cell 2021, 20, e13452. [Google Scholar] [CrossRef]
- Schaeffer, S.; Iadecola, C. Revisiting the neurovascular unit. Nat. Neurosci. 2021, 24, 1198–1209. [Google Scholar] [CrossRef] [PubMed]
- Campisi, J. Cancer, aging and cellular senescence. In Vivo 2000, 14, 183–188. [Google Scholar] [PubMed]
- Zlokovic, B.V. New therapeutic targets in the neurovascular pathway in Alzheimer’s disease. Neurotherapeutics 2008, 5, 409–414. [Google Scholar] [CrossRef]
- Hu, X.; De Silva, T.M.; Chen, J.; Faraci, F.M. Cerebral vascular disease and neurovascular injury in ischemic stroke. Circ. Res. 2017, 120, 449–471. [Google Scholar] [CrossRef] [PubMed]
- Lähteenvuo, J.; Rosenzweig, A. Effects of aging on angiogenesis. Circ. Res. 2012, 110, 1252–1264. [Google Scholar] [CrossRef]
- Montagne, A.; Barnes, S.R.; Sweeney, M.D.; Halliday, M.R.; Sagare, A.P.; Zhao, Z.; Toga, A.W.; Jacobs, R.E.; Liu, C.Y.; Amezcua, L.; et al. Blood-brain barrier breakdown in the aging human hippocampus. Neuron 2015, 85, 296–302. [Google Scholar] [CrossRef]
- Nelson, A.R.; Sweeney, M.D.; Sagare, A.P.; Zlokovic, B.V. Neurovascular dysfunction and neurodegeneration in dementia and Alzheimer’s disease. Biochim. Biophys. Acta Mol. Basis Dis. 2016, 1862, 887–900. [Google Scholar] [CrossRef]
- Wilhelm, I.; Nyúl-Tóth, Á.; Kozma, M.; Farkas, A.E.; Krizbai, I.A. Role of pattern recognition receptors of the neurovascular unit in inflamm-aging. Am. J. Physiol. Heart Circ. Physiol. 2017, 313, H1000–H1012. [Google Scholar] [CrossRef]
- Zhou, Z.D.; Wang, D.Q.; Tan, E.K. The role of neurovascular unit in neurodegeneration. Front. Cell. Neurosci. 2022, 16, 870631. [Google Scholar] [CrossRef]
- Spitzer, D.; Guérit, S.; Puetz, T.; Khel, M.I.; Armbrust, M.; Dunst, M.; Macas, J.; Zinke, J.; Devraj, G.; Jia, X.; et al. Profiling the neurovascular unit unveils detrimental effects of osteopontin on the blood–brain barrier in acute ischemic stroke. Acta Neuropathol. 2022, 144, 305–337. [Google Scholar] [CrossRef]
- Jeong, H.W.; Diéguez-Hurtado, R.; Arf, H.; Song, J.; Park, H.; Kruse, K.; Sorokin, L.; Adams, R.H. Single-cell transcriptomics reveals functionally specialized vascular endothelium in brain. eLife 2022, 11, e57520. [Google Scholar] [CrossRef] [PubMed]
- Zhao, Y.; Xie, Y.Z.; Liu, Y.S. Accelerated aging-related transcriptome alterations in neurovascular unit cells in the brain of Alzheimer’s disease. Front. Aging Neurosci. 2022, 14, 949074. [Google Scholar] [CrossRef]
- Xia, X.; Chen, W.; McDermott, J.; Han, J.D.J. Molecular and phenotypic biomarkers of aging. F1000Research 2017, 6, 860. [Google Scholar] [CrossRef] [PubMed]
- Ahadi, S.; Zhou, W.; Schüssler-Fiorenza Rose, S.M.; Sailani, M.R.; Contrepois, K.; Avina, M.; Ashland, M.; Brunet, A.; Snyder, M. Personal aging markers and ageotypes revealed by deep longitudinal profiling. Nat. Med. 2020, 26, 83–90. [Google Scholar] [CrossRef] [PubMed]
- Song, Z.; von Figura, G.; Liu, Y.; Kraus, J.M.; Torrice, C.; Dillon, P.; Rudolph-Watabe, M.; Ju, Z.; Kestler, H.A.; Sanoff, H.; et al. Lifestyle impacts on the aging-associated expression of biomarkers of dna damage and telomere dysfunction in human blood. Aging Cell 2010, 9, 607–615. [Google Scholar] [CrossRef]
- Horvath, S.; Zhang, Y.; Langfelder, P.; Kahn, R.S.; Boks, M.P.; van Eijk, K.; van den Berg, L.H.; Ophoff, R.A. Aging effects on dna methylation modules in human brain and blood tissue. Genome Biol. 2012, 13, 1–18. [Google Scholar] [CrossRef]
- Day, K.; Waite, L.L.; Thalacker-Mercer, A.; West, A.; Bamman, M.M.; Brooks, J.D.; Myers, R.M.; Absher, D. Differential dna methylation with age displays both common and dynamic features across human tissues that are influenced by cpg landscape. Genome Biol. 2013, 14, 1–19. [Google Scholar] [CrossRef]
- Greer, E.L.; Shi, Y. Histone methylation: A dynamic mark in health, disease and inheritance. Nat. Rev. Genet. 2012, 13, 343–357. [Google Scholar] [CrossRef]
- Greer, E.L.; Maures, T.J.; Hauswirth, A.G.; Green, E.M.; Leeman, D.S.; Maro, G.S.; Han, S.; Banko, M.R.; Gozani, O.; Brunet, A. Members of the h3k4 trimethylation complex regulate lifespan in a germline-dependent manner in C. elegans. Nature 2010, 466, 383–387. [Google Scholar] [CrossRef]
- Li, L.; Greer, C.; Eisenman, R.N.; Secombe, J. Essential functions of the histone demethylase lid. PLoS Genet. 2010, 6, e1001221. [Google Scholar] [CrossRef]
- Djeghloul, D.; Kuranda, K.; Kuzniak, I.; Barbieri, D.; Naguibneva, I.; Choisy, C.; Bories, J.C.; Dosquet, C.; Pla, M.; Vanneaux, V.; et al. Age-associated decrease of the histone methyltransferase suv39h1 in hsc perturbs heterochromatin and b lymphoid differentiation. Stem Cell Rep. 2016, 6, 970–984. [Google Scholar] [CrossRef] [PubMed]
- Li, C.L.; Pu, M.; Wang, W.; Chaturbedi, A.; Emerson, F.J.; Lee, S.S. Region-specific h3k9me3 gain in aged somatic tissues in caenorhabdi-tis elegans. PLoS Genet. 2021, 17, e1009432. [Google Scholar] [CrossRef] [PubMed]
- Lee, J.H.; Kim, E.W.; Croteau, D.L.; Bohr, V.A. Heterochromatin: An epigenetic point of view in aging. Exp. Mol. Med. 2020, 52, 1466–1474. [Google Scholar] [CrossRef] [PubMed]
- Cao, R.; Wang, L.; Wang, H.; Xia, L.; Erdjument-Bromage, H.; Tempst, P.; Jones, R.S.; Zhang, Y. Role of histone h3 lysine 27 methylation in polycomb-group silencing. Science 2002, 298, 1039–1043. [Google Scholar] [CrossRef] [PubMed]
- Siebold, A.P.; Banerjee, R.; Tie, F.; Kiss, D.L.; Moskowitz, J.; Harte, P.J. Polycomb repressive complex 2 and trithorax modulate Drosophila longevity and stress resistance. Proc. Natl. Acad. Sci. USA 2010, 107, 169–174. [Google Scholar] [CrossRef]
- Ni, Z.; Ebata, A.; Alipanahiramandi, E.; Lee, S.S. Two set domain containing genes link epigenetic changes and aging in caenorhabditis elegans. Aging Cell 2012, 11, 315–325. [Google Scholar] [CrossRef]
- Maures, T.J.; Greer, E.L.; Hauswirth, A.G.; Brunet, A. The h3k27 demethylase utx-1 regulates C. elegans lifespan in a germline-independent, insulin-dependent manner. Aging Cell 2011, 10, 980–990. [Google Scholar] [CrossRef]
- Liu, L.; Cheung, T.H.; Charville, G.W.; Hurgo, B.M.; Leavitt, T.; Shih, J.; Brunet, A.; Rando, T.A. Chromatin modifications as determinants of muscle stem cell quiescence and chronological aging. Cell Rep. 2013, 4, 189–204. [Google Scholar] [CrossRef]
- Baumgart, M.; Groth, M.; Priebe, S.; Savino, A.; Testa, G.; Dix, A.; Ripa, R.; Spallotta, F.; Gaetano, C.; Ori, M.; et al. Rna-seq of the aging brain in the short-lived fish N. furzeri–conserved pathways and novel genes associated with neurogenesis. Aging Cell 2014, 13, 965–974. [Google Scholar] [CrossRef]
- Peters, M.J.; Joehanes, R.; Pilling, L.C.; Schurmann, C.; Conneely, K.N.; Powell, J.; Reinmaa, E.; Sutphin, G.L.; Zhernakova, A.; Schramm, K.; et al. The transcriptional landscape of age in human peripheral blood. Nat. Commun. 2015, 6, 8570. [Google Scholar] [CrossRef]
- Li, X.; Khanna, A.; Li, N.; Wang, E. Circulatory mir-34a as an RNA-based, noninvasive biomarker for brain aging. Aging 2011, 3, 985. [Google Scholar] [CrossRef] [PubMed]
- Dhahbi, J.M. Circulating small noncoding rnas as biomarkers of aging. Ageing Res. Rev. 2014, 17, 86–98. [Google Scholar] [CrossRef] [PubMed]
- Grammatikakis, I.; Panda, A.C.; Abdelmohsen, K.; Gorospe, M. Long noncoding rnas (lncrnas) and the molecular hallmarks of aging. Aging 2014, 6, 992. [Google Scholar] [CrossRef] [PubMed]
- Kour, S.; Rath, P.C. Long noncoding rnas in aging and age-related diseases. Ageing Res. Rev. 2016, 26, 1–21. [Google Scholar] [CrossRef]
- Finkel, D.; Pedersen, N.L.; Plomin, R.; McClearn, G.E. Longitudinal and cross-sectional twin data on cognitive abilities in adulthood: The swedish adoption/twin study of aging. Dev. Psychol. 1998, 34, 1400. [Google Scholar] [CrossRef]
- Reynolds, C.A.; Finkel, D. A meta-analysis of heritability of cognitive aging: Minding the “missing heritability” gap. Neuropsychol. Rev. 2015, 25, 97–112. [Google Scholar] [CrossRef]
- Blauwendraat, C.; Pletnikova, O.; Geiger, J.T.; Murphy, N.A.; Abramzon, Y.; Rudow, G.; Mamais, A.; Sabir, M.S.; Crain, B.; Ahmed, S.; et al. Genetic analysis of neurodegenerative diseases in a pathology cohort. Neurobiol. Aging 2019, 76, 214.e1–214.e9. [Google Scholar] [CrossRef]
- Cochran, J.N.; Geier, E.G.; Bonham, L.W.; Newberry, J.S.; Amaral, M.D.; Thompson, M.L.; Lasseigne, B.N.; Karydas, A.M.; Roberson, E.D.; Cooper, G.M.; et al. Non-coding and loss-of-function coding variants in tet2 are associated with multiple neurodegenerative diseases. Am. J. Hum. Genet. 2020, 106, 632–645. [Google Scholar] [CrossRef]
- Cirulli, E.T.; Lasseigne, B.N.; Petrovski, S.; Sapp, P.C.; Dion, P.A.; Leblond, C.S.; Couthouis, J.; Lu, Y.F.; Wang, Q.; Krueger, B.J.; et al. Exome sequencing in amyotrophic lateral sclerosis identifies risk genes and pathways. Science 2015, 347, 1436–1441. [Google Scholar] [CrossRef]
- Chung, S.J.; Kim, M.J.; Kim, J.; Kim, Y.J.; You, S.; Koh, J.; Kim, S.Y.; Lee, J.H. Exome array study did not identify novel variants in Alzheimer’s disease. Neurobiol. Aging 2014, 35, 1958.e13–1958.e14. [Google Scholar] [CrossRef]
- Nikolac Perkovic, M.; Pivac, N. Genetic Markers of Alzheimer’s Disease. Adv. Exp. Med. Biol. 2019, 1192, 27–52. [Google Scholar] [PubMed]
- Song, W.; Hooli, B.; Mullin, K.; Jin, S.C.; Cella, M.; Ulland, T.K.; Wang, Y.; Tanzi, R.E.; Colonna, M. Alzheimer’s disease-associated trem2 variants exhibit either decreased or in-creased ligand-dependent activation. Alzheimer’s Dement. 2017, 13, 381–387. [Google Scholar] [CrossRef] [PubMed]
- Ruiz, A.; Dols-Icardo, O.; Bullido, M.J.; Pastor, P.; Rodríguez-Rodríguez, E.; López de Munain, A.; de Pancorbo, M.M.; Pérez-Tur, J.; Alvarez, V.; Antonell, A.; et al. Assessing the role of the trem2 p. r47h variant as a risk factor for Alzheimer’s disease and frontotemporal dementia. Neurobiol. Aging 2014, 35, 444.e1–444.e4. [Google Scholar] [CrossRef] [PubMed]
- Mehrjoo, Z.; Najmabadi, A.; Abedini, S.S.; Mohseni, M.; Kamali, K.; Najmabadi, H.; Khorram Khorshid, H.R. Association study of the trem2 gene and identification of a novel variant in exon 2 in iranian patients with late-onset Alzheimer’s disease. Med. Princ. Pract. 2015, 24, 351–354. [Google Scholar] [CrossRef] [PubMed]
- Guerreiro, R.; Wojtas, A.; Bras, J.; Carrasquillo, M.; Rogaeva, E.; Majounie, E.; Cruchaga, C.; Sassi, C.; Kauwe, J.S.; Younkin, S.; et al. Trem2 variants in Alzheimer’s disease. N. Engl. J. Med. 2013, 368, 117–127. [Google Scholar] [CrossRef]
- Jonsson, T.; Stefansson, H.; Steinberg, S.; Jonsdottir, I.; Jonsson, P.V.; Snaedal, J.; Bjornsson, S.; Huttenlocher, J.; Levey, A.I.; Lah, J.J.; et al. Variant of trem2 associated with the risk of Alzheimer’s disease. N. Engl. J. Med. 2013, 368, 107–116. [Google Scholar] [CrossRef]
- Jiang, T.; Tan, L.; Chen, Q.; Tan, M.S.; Zhou, J.S.; Zhu, X.C.; Lu, H.; Wang, H.F.; Zhang, Y.D.; Yu, J.T. A rare coding variant in trem2 increases risk for Alzheimer’s disease in han chinese. Neurobiol. Aging 2016, 42, 217.e1–217.e3. [Google Scholar] [CrossRef]
- Jin, S.C.; Carrasquillo, M.M.; Benitez, B.A.; Skorupa, T.; Carrell, D.; Patel, D.; Lincoln, S.; Krishnan, S.; Kachadoorian, M.; Reitz, C.; et al. Trem2 is associated with increased risk for Alzheimer’s disease in african amer-icans. Mol. Neurodegener. 2015, 10, 1–7. [Google Scholar] [CrossRef]
- Berge, G.; Sando, S.B.; Rongve, A.; Aarsland, D.; White, L.R. Onset of dementia with lewy bodies is delayed for carriers of the apolipoprotein e ε2 genotype in a norwegian cohort. Mov. Disord. 2014, 29, S220. [Google Scholar]
- Calvo, A.; Chiò, A. Sclerosi laterale amiotrofica come modello di gestione interdisciplinare. SALUTE E SOCIETÀ 2015, 3, 173–184. [Google Scholar] [CrossRef]
- Borroni, B.; Ferrari, F.; Galimberti, D.; Nacmias, B.; Barone, C.; Bagnoli, S.; Fenoglio, C.; Piaceri, I.; Archetti, S.; Bonvicini, C.; et al. Heterozygous trem2 mutations in frontotemporal dementia. Neurobiol. Aging 2014, 35, 934.e7–934.e10. [Google Scholar] [CrossRef] [PubMed]
- Rayaprolu, S.; Mullen, B.; Baker, M.; Lynch, T.; Finger, E.; Seeley, W.W.; Hatanpaa, K.J.; Lomen-Hoerth, C.; Kertesz, A.; Bigio, E.H.; et al. Trem2 in neurodegeneration: Evidence for association of the p. r47h variant with frontotemporal dementia and parkinson’s disease. Mol. Neurodegener. 2013, 8, 1–5. [Google Scholar] [CrossRef] [PubMed]
- Cady, J.; Koval, E.D.; Benitez, B.A.; Zaidman, C.; Jockel-Balsarotti, J.; Allred, P.; Baloh, R.H.; Ravits, J.; Simpson, E.; Appel, S.H.; et al. Trem2 variant p. r47h as a risk factor for sporadic amyotrophic lateral sclerosis. JAMA Neurol. 2014, 71, 449–453. [Google Scholar] [CrossRef] [PubMed]
- Slattery, C.F.; Beck, J.A.; Harper, L.; Adamson, G.; Abdi, Z.; Uphill, J.; Campbell, T.; Druyeh, R.; Mahoney, C.J.; Rohrer, J.D.; et al. R47h trem2 variant increases risk of typical early-onset Alzheimer’s disease but not of prion or frontotemporal dementia. Alzheimer’s Dement. 2014, 10, 602–608. [Google Scholar] [CrossRef] [PubMed]
- Gonzalez Murcia, J.D.; Schmutz, C.; Munger, C.; Perkes, A.; Gustin, A.; Peterson, M.; Ebbert, M.T.; Norton, M.C.; Tschanz, J.T.; Munger, R.G.; et al. Assessment of trem2 rs75932628 association with Alzheimer’s disease in a population-based sample: The cache county study. Neurobiol. Aging 2013, 34, 2889. [Google Scholar] [CrossRef]
- Walton, R.L.; Soto-Ortolaza, A.I.; Murray, M.E.; Lorenzo-Betancor, O.; Ogaki, K.; Heckman, M.G.; Rayaprolu, S.; Rademakers, R.; Ertekin-Taner, N.; Uitti, R.J.; et al. Trem2 p. r47h substitution is not associated with dementia with lewy bodies. Neurol. Genet. 2016, 2, e85. [Google Scholar] [CrossRef]
- Sun, J.; Zhu, Z.; Chen, K.; Wei, D.; Li, X.; Li, H.; Zhang, J.; Chen, X.; Chen, Y.; Zhang, Z. Apoe ε4 allele accelerates age-related multi-cognitive decline and white matter damage in non-demented elderly. Aging 2020, 12, 12019. [Google Scholar] [CrossRef]
- Goel, N.; Karir, P.; Garg, V.K. Role of DNA methylation in human age prediction. Mech. Ageing Dev. 2017, 166, 33–41. [Google Scholar] [CrossRef]
- Jung, M.; Pfeifer, G.P. Aging and DNA methylation. BMC Biol. 2015, 13, 1–8. [Google Scholar] [CrossRef]
- Lister, R.; Pelizzola, M.; Dowen, R.H.; Hawkins, R.D.; Hon, G.; Tonti-Filippini, J.; Nery, J.R.; Lee, L.; Ye, Z.; Ngo, Q.M.; et al. Human DNA methylomes at base resolution show widespread epigenomic differences. Nature 2009, 462, 315–322. [Google Scholar] [CrossRef]
- Jones, P.A. Functions of DNA methylation: Islands, start sites, gene bodies and beyond. Nat. Rev. Genet. 2012, 13, 484–492. [Google Scholar] [CrossRef] [PubMed]
- Zampieri, M.; Ciccarone, F.; Calabrese, R.; Franceschi, C.; Bürkle, A.; Caiafa, P. Reconfiguration of DNA methylation in aging. Mech. Ageing Dev. 2015, 151, 60–70. [Google Scholar] [CrossRef] [PubMed]
- Hannum, G.; Guinney, J.; Zhao, L.; Zhang, L.; Hughes, G.; Sadda, S.; Klotzle, B.; Bibikova, M.; Fan, J.B.; Gao, Y.; et al. Genome-wide methylation profiles reveal quantitative views of human aging rates. Mol. Cell 2013, 49, 359–367. [Google Scholar] [CrossRef]
- Fraga, M.F.; Ballestar, E.; Paz, M.F.; Ropero, S.; Setien, F.; Ballestar, M.L.; Heine-Suñer, D.; Cigudosa, J.C.; Urioste, M.; Benitez, J.; et al. Epigenetic differences arise during the lifetime of monozygotic twins. Proc. Natl. Acad. Sci. USA 2005, 102, 10604–10609. [Google Scholar] [CrossRef] [PubMed]
- Ryan, C.P. “Epigenetic clocks”: Theory and applications in human biology. Am. J. Hum. Biol. 2021, 33, e23488. [Google Scholar] [CrossRef]
- Martino, D.; Loke, Y.J.; Gordon, L.; Ollikainen, M.; Cruickshank, M.N.; Saffery, R.; Craig, J.M. Longitudinal, genome-scale analysis of dna methylation in twins from birth to 18 months of age reveals rapid epigenetic change in early life and pair-specific effects of discordance. Genome Biol. 2013, 14, R42. [Google Scholar] [CrossRef]
- Bjornsson, H.T.; Sigurdsson, M.I.; Fallin, M.D.; Irizarry, R.A.; Aspelund, T.; Cui, H.; Yu, W.; Rongione, M.A.; Ekström, T.J.; Harris, T.B.; et al. Intra-individual change over time in dna methylation with familial clustering. JAMA 2008, 299, 2877–2883. [Google Scholar] [CrossRef]
- Wynford-Thomas, D. Telomeres, p53 and cellular senescence. Oncol Res. 1996, 8, 387–398. [Google Scholar]
- von Zglinicki, T. Telomeres: Influencing the rate of aging. Ann. N. Y. Acad. Sci. 1998, 854, 318–327. [Google Scholar] [CrossRef]
- Teschendorff, A.E.; West, J.; Beck, S. Age-associated epigenetic drift: Implications, and a case of epigenetic thrift? Hum. Mol. Genet. 2013, 22, R7–R15. [Google Scholar] [CrossRef]
- Horvath, S. Dna methylation age of human tissues and cell types. Genome Biol. 2013, 14, R115. [Google Scholar] [CrossRef] [PubMed]
- Levine, M.E.; Lu, A.T.; Quach, A.; Chen, B.H.; Assimes, T.L.; Bandinelli, S.; Hou, L.; Baccarelli, A.A.; Stewart, J.D.; Li, Y.; et al. An epigenetic biomarker of aging for lifespan and healthspan. Aging 2018, 10, 573–591. [Google Scholar] [CrossRef] [PubMed]
- Marioni, R.E.; Shah, S.; McRae, A.F.; Ritchie, S.J.; Muniz-Terrera, G.; Harris, S.E.; Gibson, J.; Redmond, P.; Cox, S.R.; Pattie, A.; et al. The epigenetic clock is correlated with physical and cognitive fitness in the lothian birth cohort 1936. Int. J. Epidemiol. 2015, 44, 1388–1396. [Google Scholar] [CrossRef] [PubMed]
- Levine, M.E.; Lu, A.T.; Bennett, D.A.; Horvath, S. Epigenetic age of the pre-frontal cortex is associated with neuritic plaques, amyloid load, and Alzheimer’s disease related cognitive functioning. Aging 2015, 7, 1198. [Google Scholar] [CrossRef]
- Horvath, S.; Langfelder, P.; Kwak, S.; Aaronson, J.; Rosinski, J.; Vogt, T.F.; Eszes, M.; Faull, R.L.; Curtis, M.A.; Waldvogel, H.J.; et al. Huntington’s disease accelerates epigenetic aging of human brain and disrupts dna methylation levels. Aging 2016, 8, 1485. [Google Scholar] [CrossRef]
- Grodstein, F.; Lemos, B.; Yu, L.; Klein, H.U.; Iatrou, A.; Buchman, A.S.; Shireby, G.L.; Mill, J.; Schneider, J.A.; De Jager, P.L.; et al. The association of epigenetic clocks in brain tissue with brain pathologies and common aging phenotypes. Neurobiol. Dis. 2021, 157, 105428. [Google Scholar] [CrossRef]
- Grodstein, F.; Lemos, B.; Yu, L.; Iatrou, A.; De Jager, P.L.; Bennett, D.A. Characteristics of epigenetic clocks across blood and brain tissue in older women and men. Front. Neurosci. 2021, 14, 555307. [Google Scholar] [CrossRef]
- Fraga, M.F.; Esteller, M. Epigenetics and aging: The targets and the marks. Trends Genet. 2007, 23, 413–418. [Google Scholar] [CrossRef]
- Han, S.; Brunet, A. Histone methylation makes its mark on longevity. Trends Cell Biol. 2012, 22, 42–49. [Google Scholar] [CrossRef]
- López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The hallmarks of aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef]
- Gjoneska, E.; Pfenning, A.R.; Mathys, H.; Quon, G.; Kundaje, A.; Tsai, L.H.; Kellis, M. Conserved epigenomic signals in mice and humans reveal immune basis of Alzheimer’s disease. Nature 2015, 518, 365–369. [Google Scholar] [CrossRef] [PubMed]
- Cao, Q.; Wang, W.; Williams, J.B.; Yang, F.; Wang, Z.J.; Yan, Z. Targeting histone K4 trimethylation for treatment of cognitive and synaptic deficits in mouse models of Alzheimer’s disease. Sci. Adv. 2020, 6, eabc8096. [Google Scholar] [CrossRef] [PubMed]
- Nativio, R.; Donahue, G.; Berson, A.; Lan, Y.; Amlie-Wolf, A.; Tuzer, F.; Toledo, J.B.; Gosai, S.J.; Gregory, B.D.; Torres, C.; et al. Dysregulation of the epigenetic landscape of normal aging in Alzheimer’s disease. Nat. Neurosci. 2018, 21, 497–505. [Google Scholar] [CrossRef]
- Santana, D.A.; Smith MD, A.C.; Chen, E.S. Histone modifications in alzheimer’s disease. Genes 2023, 14, 347. [Google Scholar] [CrossRef]
- Tang, B.; Dean, B.; Thomas, E. Disease-and age-related changes in histone acetylation at gene promoters in psychiatric disorders. Transl. Psychiatry 2011, 1, e64. [Google Scholar] [CrossRef]
- Chaput, D.; Kirouac, L.; Stevens, S.M., Jr.; Padmanabhan, J. Potential role of PCTAIRE-2, PCTAIRE-3 and P-Histone H4 in amyloid precursor protein-dependent Alzheimer pathology. Oncotarget 2016, 7, 8481. [Google Scholar] [CrossRef] [PubMed]
- Ogawa, O.; Zhu, X.; Lee, H.G.; Raina, A.; Obrenovich, M.E.; Bowser, R.; Smith, M.A. Ectopic localization of phosphorylated histone H3 in Alzheimer’s disease: A mitotic catastrophe? Acta Neuropathol. 2003, 105, 524–528. [Google Scholar] [CrossRef] [PubMed]
- D’haene, E.; Vergult, S. Interpreting the impact of noncoding structural variation in neurodevelopmental disorders. Genet. Med. 2021, 23, 34–46. [Google Scholar] [CrossRef] [PubMed]
- Sherazi, S.A.M.; Abbasi, A.; Jamil, A.; Uzair, M.; Ikram, A.; Qamar, S.; Olamide, A.A.; Arshad, M.; Fried, P.J.; Ljubisavljevic, M.; et al. Molecular hallmarks of long non-coding RNAs in aging and its significant effect on aging-associated diseases. Neural Regen. Res. 2023, 18, 959–968. [Google Scholar]
- Wang, D.Q.; Fu, P.; Yao, C.; Zhu, L.S.; Hou, T.Y.; Chen, J.G.; Lu, Y.; Liu, D.; Zhu, L.Q. Long non-coding RNAs, novel culprits, or bodyguards in neurodegenerative diseas-es. Mol. Ther. Nucleic Acids 2018, 10, 269–276. [Google Scholar] [CrossRef]
- Mishra, P.; Kumar, S. Association of lncRNA with regulatory molecular factors in brain and their role in the pathophysiology of schizophrenia. Metab. Brain Dis. 2021, 36, 849–858. [Google Scholar] [CrossRef] [PubMed]
- Coccia, E.M.; Cicala, C.; Charlesworth, A.; Ciccarelli, C.; Rossi, G.B.; Philipson, L.; Sorrentino, V. Regulation and expression of a growth arrest-specific gene (gas5) during growth, differentiation, and development. Mol. Cell. Biol. 1992, 12, 3514–3521. [Google Scholar]
- Pickard, M.; Mourtada-Maarabouni, M.; Williams, G. Long non-coding RNA gas5 regulates apoptosis in prostate cancer cell lines. Biochim. Biophys. Acta Mol. Basis Dis. 2013, 1832, 1613–1623. [Google Scholar] [CrossRef] [PubMed]
- Mourtada-Maarabouni, M.; Pickard, M.; Hedge, V.; Farzaneh, F.; Williams, G. Gas5, a non-protein-coding RNA, controls apoptosis and is downregulated in breast cancer. Oncogene 2009, 28, 195–208. [Google Scholar] [CrossRef] [PubMed]
- Tang, S.; Buchman, A.S.; De Jager, P.L.; Bennett, D.A.; Epstein, M.P.; Yang, J. Novel variance-component TWAS method for studying complex human diseases with applications to Alzheimer’s dementia. PLoS Genet. 2021, 17, e1009482. [Google Scholar] [CrossRef]
- Liang, W.S.; Dunckley, T.; Beach, T.G.; Grover, A.; Mastroeni, D.; Walker, D.G.; Caselli, R.J.; Kukull, W.A.; McKeel, D.; Morris, J.C.; et al. Gene expression profiles in anatomically and functionally distinct regions of the normal aged human brain. Physiol. Genom. 2007, 28, 311–322. [Google Scholar] [CrossRef]
- Mus, E.; Hof, P.R.; Tiedge, H. Dendritic bc200 RNA in aging and in Alzheimer’s disease. Proc. Natl. Acad. Sci. USA 2007, 104, 10679–10684. [Google Scholar] [CrossRef]
- Maoz, R.; Garfinkel, B.P.; Soreq, H. Alzheimer’s disease and ncRNAs. Neuroepigenomics Aging Dis. 2017, 978, 337–361. [Google Scholar]
- Fiore, R.; Khudayberdiev, S.; Saba, R.; Schratt, G. Micro-RNA function in the nervous system. Prog. Mol. Biol. Transl. Sci. 2011, 102, 47–100. [Google Scholar]
- Goodall, E.F.; Heath, P.R.; Bandmann, O.; Kirby, J.; Shaw, P.J. Neuronal dark matter: The emerging role of microRNAs in neurodegeneration. Front. Cell. Neurosci. 2013, 7, 178. [Google Scholar] [CrossRef]
- Dickson, J.R.; Kruse, C.; Montagna, D.R.; Finsen, B.; Wolfe, M.S. Alternative polyadenylation and mir-34 family members regulate tau expression. J. Neurochem. 2013, 127, 739–749. [Google Scholar] [CrossRef] [PubMed]
- Smith, P.Y.; Hernandez-Rapp, J.; Jolivette, F.; Lecours, C.; Bisht, K.; Goupil, C.; Dorval, V.; Parsi, S.; Morin, F.; Planel, E.; et al. Mir-132/212 deficiency impairs tau metabolism and promotes pathological aggregation in vivo. Hum. Mol. Genet. 2015, 24, 6721–6735. [Google Scholar] [CrossRef] [PubMed]
- Santa-Maria, I.; Alaniz, M.E.; Renwick, N.; Cela, C.; Fulga, T.A.; Van Vactor, D.; Tuschl, T.; Clark, L.N.; Shelanski, M.L.; McCabe, B.D.; et al. Dysregulation of microRNA-219 promotes neurodegeneration through post-transcriptional regulation of tau. J. Clin. Investig. 2015, 125, 681–686. [Google Scholar] [CrossRef] [PubMed]
- Hébert, S.S.; Papadopoulou, A.S.; Smith, P.; Galas, M.C.; Planel, E.; Silahtaroglu, A.N.; Sergeant, N.; Buée, L.; De Strooper, B. Genetic ablation of dicer in adult forebrain neurons results in abnormal tau hyperphosphorylation and neurodegeneration. Hum. Mol. Genet. 2010, 19, 3959–3969. [Google Scholar] [CrossRef] [PubMed]
- Cai, Z.; Zhao, Y.; Zhao, B. Roles of glycogen synthase kinase 3 in Alzheimer’s disease. Curr. Alzheimer Res. 2012, 9, 864–879. [Google Scholar] [CrossRef] [PubMed]
- Mohamed, J.S.; Lopez, M.A.; Boriek, A.M. Mechanical stretch up-regulates microRNA-26a and induces human airway smooth muscle hypertrophy by suppressing glycogen synthase kinase-3β. J. Biol. Chem. 2010, 285, 29336–29347. [Google Scholar] [CrossRef]
- Zhao, Z.B.; Wu, L.; Xiong, R.; Wang, L.L.; Zhang, B.; Wang, C.; Li, H.; Liang, L.; Chen, S.D. MicroRNA-922 promotes tau phosphorylation by downregulating ubiquitin carboxy-terminal hydrolase l1 (uchl1) expression in the pathogenesis of Alzheimer’s disease. Neuroscience 2014, 275, 232–237. [Google Scholar] [CrossRef]
- Law, P.T.; Ching, A.K.; Chan, A.W.; Wong, Q.W.; Wong, C.K.; To, K.F.; Wong, N. Mir-145 modulates multiple components of the insulin-like growth factor pathway in hepatocellular carcinoma. Carcinogenesis 2012, 33, 1134–1141. [Google Scholar] [CrossRef]
- El Ouaamari, A.; Baroukh, N.; Martens, G.A.; Lebrun, P.; Pipeleers, D.; Van Obberghen, E. mir-375 targets 3-phosphoinositide–dependent protein kinase-1 and regulates glucose-induced biological responses in pancreatic β-cells. Diabetes 2008, 57, 2708–2717. [Google Scholar] [CrossRef]
- Inukai, S.; de Lencastre, A.; Turner, M.; Slack, F. Novel microRNAs differentially expressed during aging in the mouse brain. PLoS ONE 2012, 7, e40028. [Google Scholar] [CrossRef]
- Yang, G.; Song, Y.; Zhou, X.; Deng, Y.; Liu, T.; Weng, G.; Yu, D.; Pan, S. MicroRNA-29c targets β-site amyloid precursor protein-cleaving enzyme 1 and has a neuroprotective role in vitro and in vivo. Mol. Med. Rep. 2015, 12, 3081–3088. [Google Scholar] [CrossRef] [PubMed]
- Lei, X.; Lei, L.; Zhang, Z.; Zhang, Z.; Cheng, Y. Downregulated mir-29c correlates with increased bace1 expression in sporadic Alzheimer’s disease. Int. J. Clin. Exp. Pathol. 2015, 8, 1565. [Google Scholar] [PubMed]
- Zong, Y.; Wang, H.; Dong, W.; Quan, X.; Zhu, H.; Xu, Y.; Huang, L.; Ma, C.; Qin, C. mir-29c regulates bace1 protein expression. Brain Res. 2011, 1395, 108–115. [Google Scholar] [CrossRef] [PubMed]
- Hébert, S.S.; Horré, K.; Nicolaï, L.; Bergmans, B.; Papadopoulou, A.S.; Delacourte, A.; De Strooper, B. MicroRNA regulation of Alzheimer’s amyloid precursor protein expression. Neurobiol. Dis. 2009, 33, 422–428. [Google Scholar] [CrossRef]
- Zhu, H.C.; Wang, L.M.; Wang, M.; Song, B.; Tan, S.; Teng, J.F.; Duan, D.X. MicroRNA-195 downregulates Alzheimer’s disease amyloid-β production by targeting bace1. Brain Res. Bull. 2012, 88, 596–601. [Google Scholar] [CrossRef]
- Wang, W.X.; Rajeev, B.W.; Stromberg, A.J.; Ren, N.; Tang, G.; Huang, Q.; Rigoutsos, I.; Nelson, P.T. The expression of microRNA mir-107 decreases early in Alzheimer’s disease and may accelerate disease progression through regulation of β-site amyloid precursor protein-cleaving enzyme 1. J. Neurosci. 2008, 28, 1213–1223. [Google Scholar] [CrossRef]
- Kennerdell, J.R.; Liu, N.; Bonini, N.M. Mir-34 inhibits polycomb repressive complex 2 to modulate chaperone expression and promote healthy brain aging. Nat. Commun. 2018, 9, 4188. [Google Scholar] [CrossRef]
- Cheng, C.; Li, W.; Zhang, Z.; Yoshimura, S.; Hao, Q.; Zhang, C.; Wang, Z. MicroRNA-144 is regulated by activator protein-1 (ap-1) and decreases expression of Alzheimer disease-related a disintegrin and metalloprotease 10 (adam10). J. Biol. Chem. 2013, 288, 13748–13761. [Google Scholar] [CrossRef]
- Zhang, Y.; Kim, M.S.; Jia, B.; Yan, J.; Zuniga-Hertz, J.P.; Han, C.; Cai, D. Hypothalamic stem cells control ageing speed partly through exosomal miRNAs. Nature 2017, 548, 52–57. [Google Scholar] [CrossRef]
- Zhang, G.; Li, J.; Purkayastha, S.; Tang, Y.; Zhang, H.; Yin, Y.; Li, B.; Liu, G.; Cai, D. Hypothalamic programming of systemic ageing involving ikk-β, nf-κb and gnrh. Nature 2013, 497, 211–216. [Google Scholar] [CrossRef]
- Mohammed, C.P.D.; Park, J.S.; Nam, H.G.; Kim, K. MicroRNAs in brain aging. Mech. Ageing Dev. 2017, 168, 3–9. [Google Scholar] [CrossRef]
- Abdelmohsen, K.; Panda, A.C.; De, S.; Grammatikakis, I.; Kim, J.; Ding, J.; Noh, J.H.; Kim, K.M.; Mattison, J.A.; de Cabo, R.; et al. Circular RNAs in monkey muscle: Age-dependent changes. Aging 2015, 7, 903. [Google Scholar] [CrossRef]
- Rybak-Wolf, A.; Stottmeister, C.; Glažar, P.; Jens, M.; Pino, N.; Giusti, S.; Hanan, M.; Behm, M.; Bartok, O.; Ashwal-Fluss, R.; et al. Circular RNAs in the mammalian brain are highly abundant, conserved, and dynami-cally expressed. Mol. Cell 2015, 58, 870–885. [Google Scholar] [CrossRef]
- Hansen, T.B.; Jensen, T.I.; Clausen, B.H.; Bramsen, J.B.; Finsen, B.; Damgaard, C.K.; Kjems, J. Natural RNA circles function as efficient microRNA sponges. Nature 2013, 495, 384–388. [Google Scholar] [CrossRef] [PubMed]
- Lukiw, W.J. Circular RNA (circRNA) in Alzheimer’s disease (ad). Front. Genet. 2013, 4, 307. [Google Scholar] [CrossRef]
- Bao, N.; Liu, J.; Peng, Z.; Zhang, R.; Ni, R.; Li, R.; Wu, J.; Liu, Z.; Pan, B. Identification of circRNA-miRNA-mRNA networks to explore the molecular mechanism and immune regulation of postoperative neurocognitive disorder. Aging 2022, 14, 8374. [Google Scholar] [CrossRef] [PubMed]
- Mahmoudi, E.; Fitzsimmons, C.; Geaghan, M.P.; Shannon Weickert, C.; Atkins, J.R.; Wang, X.; Cairns, M.J. Circular RNA biogenesis is decreased in postmortem cortical gray matter in schizophrenia and may alter the bioavailability of associated miRNA. Neuropsychopharmacology 2019, 44, 1043–1054. [Google Scholar] [CrossRef] [PubMed]
- Broekmans, F.; Soules, M.; Fauser, B. Ovarian aging: Mechanisms and clinical consequences. Endocr. Rev. 2009, 30, 465–493. [Google Scholar] [CrossRef]
- Frungieri, M.B.; Calandra, R.S.; Bartke, A.; Matzkin, M.E. Male and female gonadal ageing: Its impact on health span and life span. Mech. Ageing Dev. 2021, 197, 111519. [Google Scholar] [CrossRef]
- De la Rochebrochard, E.; Thonneau, P. Paternal age 40 years: An important risk factor for infertility. Am. J. Obstet. Gynecol. 2003, 189, 901–905. [Google Scholar] [CrossRef]
- Brahem, S.; Mehdi, M.; Elghezal, H.; Saad, A. The effects of male aging on semen quality, sperm dna fragmentation and chromosomal abnormalities in an infertile population. J. Assist. Reprod. Genet. 2011, 28, 425–432. [Google Scholar] [CrossRef] [PubMed]
- Paul, C.; Robaire, B. Ageing of the male germ line. Nat. Rev. Urol. 2013, 10, 227–234. [Google Scholar] [CrossRef] [PubMed]
- Frungieri, M.B.; Calandra, R.S.; Bartke, A.; Matzkin, M.E. Ageing and inflammation in the male reproductive tract. Andrologia 2018, 50, e13034. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Y.; Ni, S.; Li, C.; Song, L.; Zhang, S. Gonadal rejuvenation of mice by growth differentiation factor 11. J. Gerontol. Ser. A 2022, 77, 892–901. [Google Scholar] [CrossRef]
- Mularoni, V.; Esposito, V.; Di Persio, S.; Vicini, E.; Spadetta, G.; Berloco, P.; Fanelli, F.; Mezzullo, M.; Pagotto, U.; Pelusi, C.; et al. Age-related changes in human Leydig cell status. Hum. Reprod. 2020, 35, 2663–2676. [Google Scholar] [CrossRef]
- Mahmoud, A.; Goemaere, S.; El-Garem, Y.; Van Pottelbergh, I.; Comhaire, F.; Kaufman, J. Testicular volume in relation to hormonal indices of gonadal function in community-dwelling elderly men. J. Clin. Endocrinol. Metab. 2003, 88, 179–184. [Google Scholar] [CrossRef]
- Golan, R.; Scovell, J.M.; Ramasamy, R. Age-related testosterone decline is due to waning of both testicular and hypothalamic-pituitary function. Aging Male 2015, 18, 201–204. [Google Scholar] [CrossRef]
- Handelsman, D.J.; Staraj, S. Testicular size: The effects of aging, malnutrition, and illness. J. Androl. 1985, 6, 144–151. [Google Scholar] [CrossRef]
- Ilacqua, A.; Izzo, G.; Emerenziani, G.P.; Baldari, C.; Aversa, A. Lifestyle and fertility: The influence of stress and quality of life on male fertility. Reprod. Biol. Endocrinol. 2018, 16, 1–11. [Google Scholar] [CrossRef]
- Kaufman, J. Ageing of the hypothalamo-pituitary-testicular axis in men. Horm. Res. Paediatr. 1995, 43, 25–28. [Google Scholar]
- Harman, S.M.; Metter, E.J.; Tobin, J.D.; Pearson, J.; Blackman, M.R. Longitudinal effects of aging on serum total and free testosterone levels in healthy men. J. Clin. Endocrinol. Metab. 2001, 86, 724–731. [Google Scholar] [CrossRef] [PubMed]
- Spinelli, R.; Parrillo, L.; Longo, M.; Florese, P.; Desiderio, A.; Zatterale, F.; Miele, C.; Raciti, G.A.; Beguinot, F. Molecular basis of ageing in chronic metabolic diseases. J. Endocrinol. Investig. 2020, 43, 1373–1389. [Google Scholar] [CrossRef] [PubMed]
- Emami, M.; Agbaedeng, T.A.; Thomas, G.; Middeldorp, M.E.; Thiyagarajah, A.; Wong, C.X.; Elliott, A.D.; Gallagher, C.; Hendriks, J.M.L.; Lau, D.H.; et al. Accelerated biological aging secondary to cardiometabolic risk factors is a predictor of cardiovascular mortality: A systematic review and meta-analysis. Can. J. Cardiol. 2022, 38, 365–375. [Google Scholar] [CrossRef]
- Russell, S.J.; Kahn, C.R. Endocrine regulation of ageing. Nat. Rev. Mol. Cell Biol. 2007, 8, 681–691. [Google Scholar] [CrossRef] [PubMed]
- Van den Beld, A.W.; Kaufman, J.M.; Zillikens, M.C.; Lamberts, S.W.; Egan, J.M.; van der Lely, A.J. The physiology of endocrine systems with ageing. Lancet Diabetes Endocrinol. 2018, 6, 647–658. [Google Scholar] [CrossRef] [PubMed]
- Park, J.; Cho, B.; Kwon, H.; Lee, C. Developing a biological age assessment equation using principal component analysis and clinical biomarkers of aging in korean men. Arch. Gerontol. Geriatr. 2009, 49, 7–12. [Google Scholar] [CrossRef]
- Nakamura, E.; Moritani, T.; Kanetaka, A. Effects of habitual physical exercise on physiological age in men aged 20–85 years as estimated using principal component analysis. Eur. J. Appl. Physiol. Occup. Physiol. 1996, 73, 410–418. [Google Scholar] [CrossRef]
- Nakamura, E.; Moritani, T.; Kanetaka, A. Biological age versus physical fitness age in women. Eur. J. Appl. Physiol. Occup. Physiol. 1990, 61, 202–208. [Google Scholar] [CrossRef]
- Nunn, A.V.; Bell, J.D.; Guy, G.W. Lifestyle-induced metabolic inflexibility and accelerated ageing syndrome: Insulin resistance, friend or foe? Nutr. Metab. 2009, 6, 1–26. [Google Scholar] [CrossRef]
- Chahal, H.; Drake, W. The endocrine system and ageing. J. Pathol. A J. Pathol. Soc. Great Br. Irel. 2007, 211, 173–180. [Google Scholar] [CrossRef]
- Chandra, A.; Rajawat, J. Skeletal aging and osteoporosis: Mechanisms and therapeutics. Int. J. Mol. Sci. 2021, 22, 3553. [Google Scholar] [CrossRef] [PubMed]
- Gaffney-Stomberg, E.; Hughes, J.M.; Guerriere, K.I.; Staab, J.S.; Cable, S.J.; Bouxsein, M.L.; McClung, J.P. Once daily calcium (1000 mg) and vitamin d (1000 iu) supplementation during military training prevents increases in biochemical markers of bone resorption but does not affect tibial microarchitecture in army recruits. Bone 2022, 155, 116269. [Google Scholar] [CrossRef] [PubMed]
- Wang, S.; Luo, Z.; Luo, H.; Li, Z.; Yuan, Z.; Tang, J.; Lin, L.; Du, Z.; Zhou, J.R. Effects of a calcium/vitamin d/zinc combination on anti-osteoporosis in ovariectomized rats. J. Trace Elem. Med. Biol. 2023, 77, 127138. [Google Scholar] [CrossRef]
- Sfeir, J.G.; Drake, M.T.; Khosla, S.; Farr, J.N. Skeletal aging. Mayo Clin. Proc. 2022, 97, 1194–1208. [Google Scholar] [CrossRef] [PubMed]
- Carter, M.I.; Hinton, P.S. Physical activity and bone health. MO Med. 2014, 111, 59. [Google Scholar]
- Orwoll, E.S.; Adler, R.A.; Amin, S.; Binkley, N.; Lewiecki, E.M.; Petak, S.M.; Shapses, S.A.; Sinaki, M.; Watts, N.B.; Sibonga, J.D. Skeletal health in long-duration astronauts: Nature, assessment, and management recommendations from the NASA bone summit. J. Bone Miner. Res. 2013, 28, 1243–1255. [Google Scholar] [CrossRef]
- Boismal, F.; Serror, K.; Dobos, G.; Zuelgaray, E.; Bensussan, A.; Michel, L. Skin aging: Pathophysiology and innovative therapies. Med. Sci. M/S 2020, 36, 1163–1172. [Google Scholar] [CrossRef]
- Baumann, L. Skin ageing and its treatment. J. Pathol. A J. Pathol. Soc. Great Br. Irel. 2007, 211, 241–251. [Google Scholar] [CrossRef]
- Kohl, E.; Steinbauer, J.; Landthaler, M.; Szeimies, R.M. Skin ageing. J. Eur. Acad. Dermatol. Venereol. 2011, 25, 873–884. [Google Scholar] [CrossRef]
- Brincat, M.; Muscat Baron, Y.; Galea, R. Estrogens and the skin. Climacteric 2005, 8, 110–123. [Google Scholar] [CrossRef]
- Park, H.Y.; Kim, J.H.; Jung, M.; Chung, C.H.; Hasham, R.; Park, C.S.; Choi, E.H. A long-standing hyperglycaemic condition impairs skin barrier by accelerating skin ageing process. Exp. Dermatol. 2011, 20, 969–974. [Google Scholar] [CrossRef] [PubMed]
- Bonté, F.; Girard, D.; Archambault, J.C.; Desmoulière, A. Skin changes during ageing. Biochem. Cell Biol. Ageing Pt II Clin. Sci. 2019, 91, 249–280. [Google Scholar]
- Shpichka, A.; Butnaru, D.; Bezrukov, E.A.; Sukhanov, R.B.; Atala, A.; Burdukovskii, V.; Zhang, Y.; Timashev, P. Skin tissue regeneration for burn injury. Stem Cell Res. Ther. 2019, 10, 1–16. [Google Scholar] [CrossRef] [PubMed]
- Bae, C.Y.; Kang, Y.G.; Piao, M.H.; Cho, B.; Cho, K.H.; Park, Y.K.; Yu, B.Y.; Lee, S.W.; Kim, M.J.; Lee, S.H.; et al. Models for estimating the biological age of five organs using clinical biomarkers that are commonly measured in clinical practice settings. Maturitas 2013, 75, 253–260. [Google Scholar] [CrossRef] [PubMed]
- Naskalova, S.; Shatilo, V.; Pisaruk, A.; Antoniuk-Shcheglova, I.; Bondarenko, O.; Bodretska, L.; Shapovalenko, I. Estimating the functional age of the respiratory system. Ageing Longev. 2022, 3, 71–76. [Google Scholar] [CrossRef]
- Sprung, J.; Gajic, O.; Warner, D.O. Age related alterations in respiratory function–anesthetic considerations. Can. J. Anesth. 2006, 53, 1244. [Google Scholar] [CrossRef]
- Charansonney, O.L. Physical activity and aging: A life-long story. Discov. Med. 2011, 12, 177–185. [Google Scholar]
- Negri, A.L. The klotho gene: A gene predominantly expressed in the kidney is a fundamental regulator of aging and calcium/phosphorus metabolism. J. Nephrol. 2005, 18, 654–658. [Google Scholar]
- Wei, S.Y.; Pan, S.Y.; Li, B.; Chen, Y.M.; Lin, S.L. Rejuvenation: Turning back the clock of aging kidney. J. Formos. Med. Assoc. 2020, 119, 898–906. [Google Scholar] [CrossRef]
- Alcedo, J.; Flatt, T.; Pasyukova, E.G. The role of the nervous system in aging and longevity. Front. Genet. 2013, 4, 124. [Google Scholar] [CrossRef]
- Bouchard, J.; Villeda, S.A. Aging and brain rejuvenation as systemic events. J. Neurochem. 2015, 132, 5–19. [Google Scholar] [CrossRef] [PubMed]
- Kimura, K.; Ieda, M.; Kanazawa, H.; Yagi, T.; Tsunoda, M.; Ninomiya, S.; Kurosawa, H.; Yoshimi, K.; Mochizuki, H.; Yamazaki, K.; et al. Cardiac sympathetic rejuvenation: A link between nerve function and cardiac hypertrophy. Circ. Res. 2007, 100, 1755–1764. [Google Scholar] [CrossRef] [PubMed]
- Martin, J.; Sheaff, M. Renal ageing. J. Pathol. A J. Pathol. Soc. Great Br. Irel. 2007, 211, 198–205. [Google Scholar] [CrossRef] [PubMed]
- Harciarek, M.; Biedunkiewicz, B.; Lichodziejewska-Niemierko, M.; Dębska-Ślizień, A.; Rutkowski, B. Continuous cognitive improvement 1 year following successful kidney transplant. Kidney Int. 2011, 79, 1353–1360. [Google Scholar] [CrossRef] [PubMed]
- Griva, K.; Thompson, D.; Jayasena, D.; Davenport, A.; Harrison, M.; Newman, S.P. Cognitive functioning pre-to post-kidney transplantation—A prospective study. Nephrol. Dial. Transplant. 2006, 21, 3275–3282. [Google Scholar] [CrossRef] [PubMed]
- Stern, Y. Cognitive reserve in ageing and Alzheimer’s disease. Lancet Neurol. 2012, 11, 1006–1012. [Google Scholar] [CrossRef]
- Amanvermez, R.; Tosun, M. An update on ovarian aging and ovarian reserve tests. Int. J. Fertil. Steril. 2016, 9, 411. [Google Scholar]
- Chilovi, B.V.; Caratozzolo, S.; Mombelli, G.; Zanetti, M.; Rozzini, L.; Padovani, A. Does reversible mci exist? Alzheimer’s Dement. 2011, 7, 5. [Google Scholar] [CrossRef]
- Vermunt, L.; van Paasen, A.J.L.; Teunissen, C.E.; Scheltens, P.; Visser, P.J.; Tijms, B.M.; Alzheimer’s Disease Neuroimaging Initiative. Alzheimer disease biomarkers may aid in the prognosis of mci cases initially reverted to normal. Neurology 2019, 92, e2699–e2705. [Google Scholar] [CrossRef]
- Zarahn, E.; Rakitin, B.; Abela, D.; Flynn, J.; Stern, Y. Age-related changes in brain activation during a delayed item recognition task. Neurobiol. Aging 2007, 28, 784–798. [Google Scholar] [CrossRef]
- Oosterhuis, E.J.; Slade, K.; May, P.J.; Nuttall, H.E. Towards an understanding of healthy cognitive ageing: The importance of lifestyle in cognitive reserve and the scaffolding theory of aging and cognition. J. Gerontol. Ser. B 2022, 78, 777–788. [Google Scholar] [CrossRef] [PubMed]
- Shimada, H.; Doi, T.; Lee, S.; Makizako, H. Reversible predictors of reversion from mild cognitive impairment to normal cognition: A 4-year longitudinal study. Alzheimer’s Res. Ther. 2019, 11, 1–9. [Google Scholar] [CrossRef] [PubMed]
- Valenzuela, M.J.; Sachdev, P.; Wen, W.; Chen, X.; Brodaty, H. Lifespan mental activity predicts diminished rate of hippocampal atrophy. PLoS ONE 2008, 3, e2598. [Google Scholar] [CrossRef] [PubMed]
- Aycheh, H.M.; Seong, J.K.; Shin, J.H.; Na, D.L.; Kang, B.; Seo, S.W.; Sohn, K.A. Biological brain age prediction using cortical thickness data: A large scale cohort study. Front. Aging Neurosci. 2018, 10, 252. [Google Scholar] [CrossRef] [PubMed]
- Franke, K.; Gaser, C. Ten years of brainage as a neuroimaging biomarker of brain aging: What insights have we gained? Front. Neurol. 2019, 10, 789. [Google Scholar] [CrossRef]
- Elliott, M.L.; Belsky, D.W.; Knodt, A.R.; Ireland, D.; Melzer, T.R.; Poulton, R.; Ramrakha, S.; Caspi, A.; Moffitt, T.E.; Hariri, A.R. Brain-age in midlife is associated with accelerated biological aging and cognitive decline in a longitudinal birth cohort. Mol. Psychiatry 2021, 26, 3829–3838. [Google Scholar] [CrossRef]
- Saxon, S.V.; Etten, M.J.; Perkins, E.A. Physical Change and Aging: A Guide for Helping Professions; Springer Publishing Company: Berlin/Heidelberg, Germany, 2021. [Google Scholar]
- Berezina, T.N.; Rybtsov, S.A. Use of personal resources may influence the rate of biological aging depending on individual typology. Eur. J. Investig. Health Psychol. Educ. 2022, 12, 1793–1811. [Google Scholar] [CrossRef]
- Bethlehem, R.A.I.; Seidlitz, J.; White, S.R.; Vogel, J.W.; Anderson, K.M.; Adamson, C.; Adler, S.; Alexopoulos, G.S.; Anagnostou, E.; Areces-Gonzalez, A. Brain charts for the human lifespan. Nature 2022, 604, 525–533. [Google Scholar] [CrossRef]
- Gaser, C.; Franke, K.; Klöppel, S.; Koutsouleris, N.; Sauer, H.; Initiative, A.D.N. Brainage in mild cognitive impaired patients: Predicting the conversion to Alzheimer’s disease. PLoS ONE 2013, 8, e67346. [Google Scholar] [CrossRef]
- Eickhoff, C.R.; Hoffstaedter, F.; Caspers, J.; Reetz, K.; Mathys, C.; Dogan, I.; Amunts, K.; Schnitzler, A.; Eickhoff, S.B. Advanced brain ageing in Parkinson’s disease is related to disease duration and individual impairment. Brain Commun. 2021, 3, fcab191. [Google Scholar] [CrossRef]
- Koutsouleris, N.; Davatzikos, C.; Borgwardt, S.; Gaser, C.; Bottlender, R.; Frodl, T.; Falkai, P.; Riecher-Rössler, A.; Möller, H.J.; Reiser, M.; et al. Accelerated brain aging in schizophrenia and beyond: A neuroanatomical marker of psychiatric disorders. Schizophr. Bull. 2014, 40, 1140–1153. [Google Scholar] [CrossRef] [PubMed]
- Franke, K.; Gaser, C. Longitudinal changes in individual brainage in healthy aging, mild cognitive impairment, and Alzheimer’s disease. GeroPsych 2012, 25, 235–245. [Google Scholar] [CrossRef]
- Scahill, R.I.; Frost, C.; Jenkins, R.; Whitwell, J.L.; Rossor, M.N.; Fox, N.C. A longitudinal study of brain volume changes in normal aging using serial registered magnetic resonance imaging. Arch. Neurol. 2003, 60, 989–994. [Google Scholar] [CrossRef]
- Coffey, C.E.; Wilkinson, W.E.; Parashos, I.A.; Soady, S.A.; Sullivan, R.J.; Patterson, L.J.; Figiel, G.S.; Webb, M.C.; Spritzer, C.E.; Djang, W.T. Quantitative cerebral anatomy of the aging human brain: A cross-sectional study using magnetic resonance imaging. Neurology 1992, 42, 527. [Google Scholar] [CrossRef]
- Good, C.D.; Johnsrude, I.S.; Ashburner, J.; Henson, R.N.; Friston, K.J.; Frackowiak, R.S. A voxel-based morphometric study of ageing in 465 normal adult human brains. Neuroimage 2001, 14, 21–36. [Google Scholar] [CrossRef] [PubMed]
- Iwasaki, A.; Foxman, E.F.; Molony, R.D. Early local immune defences in the respiratory tract. Nat. Rev. Immunol. 2017, 17, 7–20. [Google Scholar] [CrossRef]
- Sarkar, T.; Patro, N.; Patro, I.K. Cumulative multiple early life hits-a potent threat leading to neurological disorders. Brain Res. Bull. 2019, 147, 58–68. [Google Scholar] [CrossRef]
- Hawkes, C.H.; Del Tredici, K.; Braak, H. A timeline for parkinson’s disease. Park. Relat. Disord. 2010, 16, 79–84. [Google Scholar] [CrossRef]
- Turknett, J.; Wood, T.R. Demand coupling drives neurodegeneration: A model of age-related cognitive decline and dementia. Cells 2022, 11, 2789. [Google Scholar] [CrossRef]
- Schneider, C.B.; Donix, M.; Linse, K.; Werner, A.; Fauser, M.; Klingelhoefer, L.; Löhle, M.; von Kummer, R.; Reichmann, H.; Storch, A.; et al. Accelerated age-dependent hippocampal volume loss in parkinson disease with mild cognitive impairment. Am. J. Alzheimer’s Dis. Other Dement. 2017, 32, 313–319. [Google Scholar] [CrossRef]
- Smith, S.M.; Elliott, L.T.; Alfaro-Almagro, F.; McCarthy, P.; Nichols, T.E.; Douaud, G.; Miller, K.L. Brain aging comprises many modes of structural and functional change with distinct genetic and biophysical associations. eLife 2020, 9, e52677. [Google Scholar] [CrossRef] [PubMed]
- Bayati, A.; Berman, T. Localized vs. systematic neurodegeneration: A paradigm shift in understanding neurodegenerative diseases. Front. Syst. Neurosci. 2017, 11, 62. [Google Scholar] [CrossRef] [PubMed]
- Beheshti, I.; Mishra, S.; Sone, D.; Khanna, P.; Matsuda, H. T1-weighted mri-driven brain age estimation in Alzheimer’s disease and parkinson’s disease. Aging Dis. 2020, 11, 618. [Google Scholar] [CrossRef]
- McCartney, D.L.; Stevenson, A.J.; Walker, R.M.; Gibson, J.; Morris, S.W.; Campbell, A.; Murray, A.D.; Whalley, H.C.; Porteous, D.J.; McIntosh, A.M.; et al. Investigating the relationship between dna methylation age acceleration and risk factors for Alzheimer’s disease. Alzheimer’s Dement. Diagn. Assess. Dis. Monit. 2018, 10, 429–437. [Google Scholar] [CrossRef] [PubMed]
- Norrara, B.; Doerl, J.G.; Guzen, F.P.; Cavalcanti, J.R.L.P.; Freire, M.A.M. Commentary: Localized vs. systematic neurodegeneration: A paradigm shift in understanding neurodegenerative diseases. Front. Syst. Neurosci. 2017, 11, 91. [Google Scholar] [CrossRef] [PubMed]
- Kim, Y.E.; Jung, Y.S.; Ock, M.; Yoon, S.J. Daly estimation approaches: Understanding and using the incidence-based approach and the prevalence-based approach. J. Prev. Med. Public Health 2022, 55, 10. [Google Scholar] [CrossRef]
- Gao, T.; Wang, X.C.; Chen, R.; Ngo, H.H.; Guo, W. Disability adjusted life year (daly): A useful tool for quantitative assessment of environmental pollution. Sci. Total Environ. 2015, 511, 268–287. [Google Scholar] [CrossRef]
- Jonsson, B.A.; Bjornsdottir, G.; Thorgeirsson, T.E.; Ellingsen, L.M.; Walters, G.B.; Gudbjartsson, D.F.; Stefansson, H.; Stefansson, K.; Ulfarsson, M.O. Brain age prediction using deep learning uncovers associated sequence variants. Nat. Commun. 2019, 10, 5409. [Google Scholar] [CrossRef]
- Hung, C.W.; Chen, Y.C.; Hsieh, W.L.; Chiou, S.H.; Kao, C.L. Ageing and neurodegenerative diseases. Ageing Res. Rev. 2010, 9, S36–S46. [Google Scholar] [CrossRef]
- Sibbett, R.A.; Altschul, D.M.; Marioni, R.E.; Deary, I.J.; Starr, J.M.; Russ, T.C. Dna methylation-based measures of accelerated biological ageing and the risk of dementia in the oldest-old: A study of the lothian birth cohort 1921. BMC Psychiatry 2020, 20, 1–15. [Google Scholar] [CrossRef]
- Soldan, A.; Pettigrew, C.; Li, S.; Wang, M.C.; Moghekar, A.; Selnes, O.A.; Albert, M.; O’Brien, R. Relationship of cognitive reserve and cerebrospinal fluid biomarkers to the emergence of clinical symptoms in preclinical Alzheimer’s disease. Neurobiol. Aging 2013, 34, 2827–2834. [Google Scholar] [CrossRef]
- Fries, G.R.; Bauer, I.E.; Scaini, G.; Valvassori, S.S.; Walss-Bass, C.; Soares, J.C.; Quevedo, J. Accelerated hippocampal biological aging in bipolar disorder. Bipolar Disord. 2020, 22, 498–507. [Google Scholar] [CrossRef] [PubMed]
- Tønnesen, S.; Kaufmann, T.; de Lange, A.G.; Richard, G.; Doan, N.T.; Alnæs, D.; van der Meer, D.; Rokicki, J.; Moberget, T.; Maximov, I.I.; et al. Brain age prediction reveals aberrant brain white matter in schizophrenia and bipolar disorder: A multisample diffusion tensor imaging study. Biol. Psychiatry Cogn. Neurosci. Neuroimaging 2020, 5, 1095–1103. [Google Scholar] [CrossRef] [PubMed]
- Heather, C.W.; Jude, G.; Riccardo, M.; Rosie, M.W.; Toni-Kim, C.; David, M.H.; Mark, J.A.; Lynsey, H.; Stewart, M.; Ian, J.D.; et al. Accelerated epigenetic ageing in major depressive disorder. bioRxiv 2017. [Google Scholar] [CrossRef]
- Dudley, J.A.; Lee, J.H.; Durling, M.; Strakowski, S.M.; Eliassen, J.C. Age-dependent decreases of high energy phosphates in cerebral gray matter of patients with bipolar i disorder: A preliminary phosphorus-31 magnetic resonance spectroscopic imaging study. J. Affect. Disord. 2015, 175, 251–255. [Google Scholar] [CrossRef] [PubMed]
- Masuda, Y.; Okada, G.; Takamura, M.; Shibasaki, C.; Yoshino, A.; Yokoyama, S.; Ichikawa, N.; Okuhata, S.; Kobayashi, T.; Yamawaki, S.; et al. Age-related white matter changes revealed by a whole-brain fiber-tracking method in bipolar disorder compared to major depressive disorder and healthy controls. Psychiatry Clin. Neurosci. 2021, 75, 46–56. [Google Scholar] [CrossRef] [PubMed]
- Muangpaisan, W.; Petcharat, C.; Srinonprasert, V. Prevalence of potentially reversible conditions in dementia and mild cognitive impairment in a geriatric clinic. Geriatr. Gerontol. Int. 2012, 12, 59–64. [Google Scholar] [CrossRef]
- Gauthier, S.; Touchon, J. Mild cognitive impairment is not a clinical entity and should not be treated. Arch. Neurol. 2005, 62, 1164–1166. [Google Scholar] [CrossRef]
- Bates, K.; Harvey, A.R.; Carruthers, M.; Martins, R. Androgens, andropause and neurodegeneration: Exploring the link between steroidogenesis, androgens and Alzheimer’s disease. Cell Mol. Life Sci. 2005, 62, 281–292. [Google Scholar] [CrossRef]
- Liu, Z.; Zhou, T.; Ziegler, A.C.; Dimitrion, P.; Zuo, L. Oxidative stress in neurodegenerative diseases: From molecular mechanisms to clinical applications. Oxidative Med. Cell. Longev. 2017, 2017, 2525967. [Google Scholar] [CrossRef]
- Biswas, S.K. Does the interdependence between oxidative stress and inflammation explain the antioxidant paradox? Oxidative Med. Cell. Longev. 2016, 2016, 5698931. [Google Scholar] [CrossRef] [PubMed]
- Bordoni, M.; Scarian, E.; Rey, F.; Gagliardi, S.; Carelli, S.; Pansarasa, O.; Cereda, C. Biomaterials in neurodegenerative disorders: A promising therapeutic approach. Int. J. Mol. Sci. 2020, 21, 3243. [Google Scholar] [CrossRef] [PubMed]
- Bell, C.G.; Lowe, R.; Adams, P.D.; Baccarelli, A.A.; Beck, S.; Bell, J.T.; Christensen, B.C.; Gladyshev, V.N.; Heijmans, B.T.; Horvath, S.; et al. DNA methylation aging clocks: Challenges and recommendations. Genome Biol. 2019, 20, 1–24. [Google Scholar] [CrossRef] [PubMed]
- Stubbs, T.M.; Bonder, M.J.; Stark, A.K.; Krueger, F.; von Meyenn, F.; Stegle, O.; Reik, W. Multi-tissue DNA methylation age predictor in mouse. Genome Biol. 2017, 18, 68. [Google Scholar] [CrossRef] [PubMed]
- Thompson, M.J.; Chwiałkowska, K.; Rubbi, L.; Lusis, A.J.; Davis, R.C.; Srivastava, A.; Korstanje, R.; Churchill, G.A.; Horvath, S.; Pellegrini, M. A multi-tissue full lifespan epigenetic clock for mice. Aging 2018, 10, 2832. [Google Scholar] [CrossRef]
- Wang, T.; Tsui, B.; Kreisberg, J.F.; Robertson, N.A.; Gross, A.M.; Yu, M.K.; Carter, H.; Brown-Borg, H.M.; Adams, P.D.; Ideker, T. Epigenetic aging signatures in mice livers are slowed by dwarfism, calorie restriction and rapamycin treatment. Genome Biol. 2017, 18, 57. [Google Scholar] [CrossRef]
- Petkovich, D.A.; Podolskiy, D.I.; Lobanov, A.V.; Lee, S.G.; Miller, R.A.; Gladyshev, V.N. Using DNA methylation profiling to evaluate biological age and longevity interventions. Cell Metab. 2017, 25, 954–960. [Google Scholar] [CrossRef]
- Meer, M.V.; Podolskiy, D.I.; Tyshkovskiy, A.; Gladyshev, V.N. A whole lifespan mouse multi-tissue DNA methylation clock. eLife 2018, 7, e40675. [Google Scholar] [CrossRef]
- Bahar, R.; Hartmann, C.H.; Rodriguez, K.A.; Denny, A.D.; Busuttil, R.A.; Dollé, M.E.; Calder, R.B.; Chisholm, G.B.; Pollock, B.H.; Klein, C.A.; et al. Increased cell-to-cell variation in gene expression in ageing mouse heart. Nature 2006, 441, 1011–1014. [Google Scholar] [CrossRef]
- Rimmelé, P.; Bigarella, C.L.; Liang, R.; Izac, B.; Dieguez-Gonzalez, R.; Barbet, G.; Donovan, M.; Brugnara, C.; Blander, J.M.; Sinclair, D.A.; et al. Aging-like phenotype and defective lineage specification in sirt1-deleted hematopoietic stem and progenitor cells. Stem Cell Rep. 2014, 3, 44–59. [Google Scholar] [CrossRef]
- Cheung, P.; Vallania, F.; Warsinske, H.C.; Donato, M.; Schaffert, S.; Chang, S.E.; Dvorak, M.; Dekker, C.L.; Davis, M.M.; Utz, P.J.; et al. Single-cell chromatin modification profiling reveals increased epigenetic variations with aging. Cell 2018, 173, 1385–1397. [Google Scholar] [CrossRef] [PubMed]
- Hernando-Herraez, I.; Evano, B.; Stubbs, T.; Commere, P.H.; Jan Bonder, M.; Clark, S.; Andrews, S.; Tajbakhsh, S.; Reik, W. Ageing affects DNA methylation drift and transcriptional cell-to-cell variability in mouse muscle stem cells. Nat. Commun. 2019, 10, 4361. [Google Scholar] [CrossRef] [PubMed]
- Trapp, A.; Kerepesi, C.; Gladyshev, V.N. Profiling epigenetic age in single cells. Nat. Aging 2021, 1, 1189–1201. [Google Scholar] [CrossRef] [PubMed]
- Hu, Y.; An, Q.; Guo, Y.; Zhong, J.; Fan, S.; Rao, P.; Liu, X.; Liu, Y.; Fan, G. Simultaneous profiling of mRNA transcriptome and DNA methylome from a single cell. Single Cell Methods Seq. Proteom. 2019, 1979, 363–377. [Google Scholar]
- Angermueller, C.; Lee, H.J.; Reik, W.; Stegle, O. Deepcpg: Accurate prediction of single-cell DNA methylation states using deep learning. Genome Biol. 2017, 18, 1–13. [Google Scholar]
- Hamidouche, Z.; Rother, K.; Przybilla, J.; Krinner, A.; Clay, D.; Hopp, L.; Fabian, C.; Stolzing, A.; Binder, H.; Charbord, P.; et al. Bistable epigenetic states explain age-dependent decline in mesenchymal stem cell heterogeneity. Stem Cells 2017, 35, 694–704. [Google Scholar] [CrossRef]
- de Lima Camillo, L.P.; Lapierre, L.R.; Singh, R. A pan-tissue DNA-methylation epigenetic clock based on deep learning. npj Aging 2022, 8, 4. [Google Scholar] [CrossRef]
- Peleg, S.; Sananbenesi, F.; Zovoilis, A.; Burkhardt, S.; Bahari-Javan, S.; Agis-Balboa, R.C.; Cota, P.; Wittnam, J.L.; Gogol-Doering, A.; Opitz, L.; et al. Altered histone acetylation is associated with age-dependent memory impairment in mice. Science 2010, 328, 753–756. [Google Scholar] [CrossRef]
- Stefanelli, G.; Azam, A.B.; Walters, B.J.; Brimble, M.A.; Gettens, C.P.; Bouchard-Cannon, P.; Cheng, H.M.; Davidoff, A.M.; Narkaj, K.; Day, J.J.; et al. Learning and age-related changes in genome-wide h2a.z binding in the mouse hippocampus. Cell Rep. 2018, 22, 1124–1131. [Google Scholar] [CrossRef]
- Klein, H.U.; McCabe, C.; Gjoneska, E.; Sullivan, S.E.; Kaskow, B.J.; Tang, A.; Smith, R.V.; Xu, J.; Pfenning, A.R.; Bernstein, B.E.; et al. Epigenome-wide study uncovers tau pathology-driven changes of chromatin organization in the aging human brain. Biorxiv 2018, 22, 273789. [Google Scholar]
- Roichman, A.; Elhanati, S.; Aon, M.A.; Abramovich, I.; Di Francesco, A.; Shahar, Y.; Avivi, M.Y.; Shurgi, M.; Rubinstein, A.; Wiesner, Y.; et al. Restoration of energy homeostasis by sirt6 extends healthy lifespan. Nat. Commun. 2021, 12, 3208. [Google Scholar] [CrossRef] [PubMed]
- Grootaert, M.O.; Finigan, A.; Figg, N.L.; Uryga, A.K.; Bennett, M.R. Sirt6 protects smooth muscle cells from senescence and reduces atherosclerosis. Circ. Res. 2021, 128, 474–491. [Google Scholar] [CrossRef] [PubMed]
- Soto-Palma, C.; Niedernhofer, L.J.; Faulk, C.D.; Dong, X. Epigenetics, DNA damage, and aging. J. Clin. Investig. 2022, 132, e158446. [Google Scholar] [CrossRef]
- Hartmann, A.; Hartmann, C.; Secci, R.; Hermann, A.; Fuellen, G.; Walter, M. Ranking biomarkers of aging by citation profiling and effort scoring. Front. Genet. 2021, 12, 686320. [Google Scholar] [CrossRef]
- Bürkle, A.; Moreno-Villanueva, M.; Bernhard, J.; Blasco, M.; Zondag, G.; Hoeijmakers, J.H.; Toussaint, O.; Grubeck-Loebenstein, B.; Mocchegiani, E.; Collino, S.; et al. MARK-AGE biomarkers of ageing. Mech. Ageing Dev. 2015, 151, 2–12. [Google Scholar] [CrossRef] [PubMed]
- Mirzayans, R.; Murray, D. Human Genetic Disorders Associated with Genome Instability, Premature Aging and Cancer Predisposition. Open Cancer J. 2008, 2, 42–52. [Google Scholar] [CrossRef]
- Tiwari, V.; Wilson, D.M. DNA damage and associated DNA repair defects in disease and premature aging. Am. J. Hum. Genet. 2019, 105, 237–257. [Google Scholar] [CrossRef]
- Rizza, E.R.; DiGiovanna, J.J.; Khan, S.G.; Tamura, D.; Jeskey, J.D.; Kraemer, K.H. Xeroderma pigmentosum: A model for human premature aging. J. Investig. Dermatol. 2021, 141, 976–984. [Google Scholar] [CrossRef]
- Han, Z.Z.; Fleet, A.; Larrieu, D. Can accelerated ageing models inform us on age-related tauopathies? Aging Cell 2023, 22, e13830. [Google Scholar] [CrossRef]
- Platzer, M.; Englert, C. Nothobranchius furzeri: A model for aging research and more. Trends Genet. 2016, 32, 543–552. [Google Scholar] [CrossRef]
- Reichwald, K.; Petzold, A.; Koch, P.; Downie, B.R.; Hartmann, N.; Pietsch, S.; Baumgart, M.; Chalopin, D.; Felder, M.; Bens, M.; et al. Insights into sex chromosome evolution and aging from the genome of a short-lived fish. Cell 2015, 163, 1527–1538. [Google Scholar] [CrossRef]
- Valenzano, D.R.; Benayoun, B.A.; Singh, P.P.; Zhang, E.; Etter, P.D.; Hu, C.K.; Clément-Ziza, M.; Willemsen, D.; Cui, R.; Harel, I.; et al. The African turquoise killifish genome provides insights into evolution and genetic architecture of lifespan. Cell 2015, 163, 1539–1554. [Google Scholar] [CrossRef]
- Petzold, A.; Reichwald, K.; Groth, M.; Taudien, S.; Hartmann, N.; Priebe, S.; Shagin, D.; Englert, C.; Platzer, M. The transcript catalogue of the short-lived fish Nothobranchius furzeri provides insights into age-dependent changes of mRNA levels. BMC Genom. 2013, 14, 185. [Google Scholar] [CrossRef] [PubMed]
- Genade, T.; Benedetti, M.; Terzibasi, E.; Roncaglia, P.; Valenzano, D.R.; Cattaneo, A.; Cellerino, A. Annual fishes of the genus Nothobranchius as a model system for aging research. Aging Cell 2005, 4, 223–233. [Google Scholar] [CrossRef] [PubMed]
- Di Cicco, E.; Tozzini, E.T.; Rossi, G.; Cellerino, A. The shortlived annual fish Nothobranchius furzeri shows a typical teleost aging process reinforced by high incidence of age-dependent neoplasias. Exp. Gerontol. 2011, 46, 249–256. [Google Scholar] [CrossRef]
- Dodzian, J.; Kean, S.; Seidel, J.; Valenzano, D.R. A protocol for laboratory housing of Turquoise Killifish (Nothobranchius furzeri). J. Vis. Exp. 2018, 134, e57073. [Google Scholar] [CrossRef]
- Tan, L.; Ke, Z.; Tombline, G.; Macoretta, N.; Hayes, K.; Tian, X.; Lv, R.; Ablaeva, J.; Gilbert, M.; Bhanu, N.V.; et al. Naked mole rat cells have a stable epigenome that resists iPSC reprogramming. Stem Cell Rep. 2017, 9, 1721–1734. [Google Scholar] [CrossRef]
- Dammann, P.; Šumbera, R.; Massmann, C.; Scherag, A.; Burda, H. Extended longevity of reproductives appears to be common in Fukomys molerats (Rodentia. Bathyergidae). PLoS ONE 2011, 6, e18757. [Google Scholar] [CrossRef]
- Ruby, J.G.; Smith, M.; Buffenstein, R. Naked mole-rat mortality rates defy gompertzian laws by not increasing with age. Elife 2018, 7, e31157. [Google Scholar] [CrossRef]
- Munshi-South, J.; Wilkinson, G.S. Bats and birds: Exceptional longevity despite high metabolic rates. Ageing Res. Rev. 2010, 9, 12–19. [Google Scholar] [CrossRef]
- Seim, I.; Fang, X.; Xiong, Z.; Lobanov, A.V.; Huang, Z.; Ma, S.; Feng, Y.; Turanov, A.A.; Zhu, Y.; Lenz, T.L.; et al. Genome analysis reveals insights into physiology and longevity of the Brandt’s bat Myotis brandtii. Nat. Commun. 2013, 4, 2212. [Google Scholar] [CrossRef] [PubMed]
- Podlutsky, A.J.; Khritankov, A.M.; Ovodov, N.D.; Austad, S.N. A new field record for bat longevity. J. Gerontol. A Biol. Sci. Med. Sci. 2005, 60, 1366–1368. [Google Scholar] [CrossRef] [PubMed]
- Holtze, S.; Lukač, M.; Cizelj, I.; Mutschmann, F.; Szentiks, C.A.; Jelić, D.; Hermes, R.; Göritz, F.; Braude, S.; Hildebrandt, T.B. Monitoring health and reproductive status of olms (Proteus anguinus) by ultrasound. PLoS ONE 2017, 12, e0182209. [Google Scholar] [CrossRef] [PubMed]
- Mulec, J. Welcome to the -omics era of the 21st century: Will Proteus anguinus finally reveal all its mysteries? Acta Carsol. 2020, 1, 49. [Google Scholar] [CrossRef]
- Voituron, Y.; de Fraipont, M.; Issartel, J.; Guillaume, O.; Clobert, J. Extreme lifespan of the human fish (Proteus anguinus): A challenge for ageing mechanisms. Biol. Lett. 2011, 7, 105–107. [Google Scholar] [CrossRef]
- Philipp, E.E.; Wessels, W.; Gruber, H.; Strahl, J.; Wagner, A.E.; Ernst, I.M.; Rimbach, G.; Kraemer, L.; Schreiber, S.; Abele, D.; et al. Gene expression and physiological changes of different populations of the long-lived bivalve Arctica islandica under low oxygen conditions. PLoS ONE 2012, 7, e44621. [Google Scholar] [CrossRef]
- Lutz, R.A.; Goodsell, J.G.; Mann, R.; Castagna, M. Experimental culture of the ocean quahog, Arctica islandica. J. World Maricult. Soc. 1981, 12, 196–205. [Google Scholar] [CrossRef]
- Chapman, J.A.; Kirkness, E.F.; Simakov, O.; Hampson, S.E.; Mitros, T.; Weinmaier, T.; Rattei, T.; Balasubramanian, P.G.; Borman, J.; Busam, D.; et al. The dynamic genome of Hydra. Nature 2010, 464, 592–596. [Google Scholar] [CrossRef]
- Schaible, R.; Sussman, M.; Kramer, B.H. Aging and potential for self-renewal: Hydra living in the age of aging—A mini-review. Gerontology 2014, 60, 548–556. [Google Scholar] [CrossRef]
- Bellantuono, A.J.; Bridge, D.; Martínez, D.E. Hydra as a tractable, long-lived model system for senescence. Invert. Reprod. Dev. 2015, 59, 39–44. [Google Scholar] [CrossRef]
- Klimovich, A.; Rehm, A.; Wittlieb, J.; Herbst, E.-M.; Benavente, R.; Bosch, T.C.G. Non-senescent Hydra tolerates severe disturbances in the nuclear lamina. Aging 2018, 10, 951–972. [Google Scholar] [CrossRef] [PubMed]
- Grohme, M.A.; Schloissnig, S.; Rozanski, A.; Pippel, M.; Young, G.R.; Winkler, S.; Brandl, H.; Henry, I.; Dahl, A.; Powell, S.; et al. The genome of Schmidtea mediterranea and the evolution of core cellular mechanisms. Nature 2018, 554, 56–61. [Google Scholar] [CrossRef] [PubMed]
- Fincher, C.T.; Wurtzel, O.; de Hoog, T.; Kravarik, K.M.; Reddien, P.W. Cell type transcriptome atlas for the planarian Schmidtea mediterranea. Science 2018, 360, eaaq1736. [Google Scholar] [CrossRef]
- Merryman, M.S.; Alvarado, A.S.; Jenkin, J.C. Culturing Planarians in the Laboratory. Methods Mol. Biol. 2018, 1774, 241–258. [Google Scholar] [CrossRef]
- Lipsky, M.S.; King, M. Biological theories of aging. Dis.-A-Mon. 2015, 61, 460–466. [Google Scholar] [CrossRef] [PubMed]
- Jin, K. Modern biological theories of aging. Aging Dis. 2010, 1, 72. [Google Scholar] [PubMed]
- Weinert, B.T.; Timiras, P.S. Invited review: Theories of aging. J. Appl. Physiol. 2003, 95, 1706–1716. [Google Scholar] [CrossRef] [PubMed]
- Northrop, J.H. The influence of the intensity of light on the rate of growth and duration of life of Drosophila. J. Gen. Physiol. 1925, 9, 81. [Google Scholar] [CrossRef]
- Northrop, J.H. Duration of life of an aseptic Drosophila culture inbred in the dark for 230 generations. J. Gen. Physiol. 1926, 9, 763–765. [Google Scholar] [CrossRef]
- NCBI PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/ (accessed on 31 August 2023).
- Tilney, F. The Aging of the Human Brain. Bull. N. Y. Acad. Med. 1928, 4, 1125–1143. [Google Scholar]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Statsenko, Y.; Kuznetsov, N.V.; Morozova, D.; Liaonchyk, K.; Simiyu, G.L.; Smetanina, D.; Kashapov, A.; Meribout, S.; Gorkom, K.N.-V.; Hamoudi, R.; et al. Reappraisal of the Concept of Accelerated Aging in Neurodegeneration and Beyond. Cells 2023, 12, 2451. https://doi.org/10.3390/cells12202451
Statsenko Y, Kuznetsov NV, Morozova D, Liaonchyk K, Simiyu GL, Smetanina D, Kashapov A, Meribout S, Gorkom KN-V, Hamoudi R, et al. Reappraisal of the Concept of Accelerated Aging in Neurodegeneration and Beyond. Cells. 2023; 12(20):2451. https://doi.org/10.3390/cells12202451
Chicago/Turabian StyleStatsenko, Yauhen, Nik V. Kuznetsov, Daria Morozova, Katsiaryna Liaonchyk, Gillian Lylian Simiyu, Darya Smetanina, Aidar Kashapov, Sarah Meribout, Klaus Neidl-Van Gorkom, Rifat Hamoudi, and et al. 2023. "Reappraisal of the Concept of Accelerated Aging in Neurodegeneration and Beyond" Cells 12, no. 20: 2451. https://doi.org/10.3390/cells12202451
APA StyleStatsenko, Y., Kuznetsov, N. V., Morozova, D., Liaonchyk, K., Simiyu, G. L., Smetanina, D., Kashapov, A., Meribout, S., Gorkom, K. N. -V., Hamoudi, R., Ismail, F., Ansari, S. A., Emerald, B. S., & Ljubisavljevic, M. (2023). Reappraisal of the Concept of Accelerated Aging in Neurodegeneration and Beyond. Cells, 12(20), 2451. https://doi.org/10.3390/cells12202451