Epigenetic Mechanisms of Aging and Aging-Associated Diseases
Abstract
:1. Introduction
2. Epigenetic Mechanisms
2.1. DNA Methylation
2.2. Histone Modifications
2.3. Histone Methylation
2.4. Histone Acetylation
2.5. Histone Phosphorylation
2.6. Histone Ribosylation
2.7. Histone Ubiquitination and SUMOylation
3. Epigenetic Changes in Cellular Senescence
4. Epigenetic Changes in Aging-Related Diseases
4.1. Tumors
4.2. Cardiovascular Disease
4.3. Neurodegenerative Diseases
4.4. Alzheimer’s Disease
4.5. Parkinson’s Disease
4.6. Diabetes
4.7. Sarcopenia
4.8. Osteoporosis
5. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Kennedy, B.K.; Berger, S.L.; Brunet, A.; Campisi, J.; Cuervo, A.M.; Epel, E.S.; Franceschi, C.; Lithgow, G.J.; Morimoto, R.I.; Pessin, J.E.; et al. Geroscience: Linking aging to chronic disease. Cell 2014, 159, 709–713. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The hallmarks of aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Harman, D. The aging process. Proc. Natl. Acad. Sci. USA 1981, 78, 7124–7128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hamilton, J.P. Epigenetics: Principles and practice. Dig. Dis. 2011, 29, 130–135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Papait, R.; Cattaneo, P.; Kunderfranco, P.; Greco, C.; Carullo, P.; Guffanti, A.; Viganò, V.; Stirparo, G.G.; Latronico, M.V.G.; Hasenfuss, G.; et al. Genome-wide analysis of histone marks identifying an epigenetic signature of promoters and enhancers underlying cardiac hypertrophy. Proc. Natl. Acad. Sci. USA 2013, 110, 20164–20169. [Google Scholar] [CrossRef] [Green Version]
- Papait, R.; Kunderfranco, P.; Stirparo, G.G.; Latronico, M.V.G.; Condorelli, G. Long noncoding RNA: A new player of heart failure? J. Cardiovasc. Transl. Res. 2013, 6, 876–883. [Google Scholar] [CrossRef] [Green Version]
- Gibney, E.R.; Nolan, C.M. Epigenetics and gene expression. Heredity 2010, 105, 4–13. [Google Scholar] [CrossRef] [Green Version]
- Heerboth, S.; Lapinska, K.; Snyder, N.; Leary, M.; Rollinson, S.; Sarkar, S. Use of epigenetic drugs in disease: An overview. Genet. Epigenet. 2014, 6, 9–19. [Google Scholar] [CrossRef]
- Sanchez-Mut, J.V.; Gräff, J. Epigenetic Alterations in Alzheimer’s Disease. Front. Behav. Neurosci. 2015, 9, 347. [Google Scholar] [CrossRef] [Green Version]
- Nikolac Perkovic, M.; Videtic Paska, A.; Konjevod, M.; Kouter, K.; Svob Strac, D.; Nedic Erjavec, G.; Pivac, N. Epigenetics of Alzheimer’s Disease. Biomolecules 2021, 11, 195. [Google Scholar] [CrossRef]
- Moore, L.D.; Le, T.; Fan, G. DNA methylation and its basic function. Neuropsychopharmacology 2013, 38, 23–38. [Google Scholar] [CrossRef] [Green Version]
- Hervouet, E.; Peixoto, P.; Delage-Mourroux, R.; Boyer-Guittaut, M.; Cartron, P.-F. Specific or not specific recruitment of DNMTs for DNA methylation, an epigenetic dilemma. Clin. Epigenet. 2018, 10, 17. [Google Scholar] [CrossRef]
- Jeltsch, A.; Ehrenhofer-Murray, A.; Jurkowski, T.P.; Lyko, F.; Reuter, G.; Ankri, S.; Nellen, W.; Schaefer, M.; Helm, M. Mechanism and biological role of Dnmt2 in Nucleic Acid Methylation. RNA Biol. 2017, 14, 1108–1123. [Google Scholar] [CrossRef] [Green Version]
- Du, X.; Han, L.; Guo, A.-Y.; Zhao, Z. Features of methylation and gene expression in the promoter-associated CpG islands using human methylome data. Comp. Funct. Genom. 2012, 2012, 598987. [Google Scholar] [CrossRef] [Green Version]
- Ziller, M.J.; Müller, F.; Liao, J.; Zhang, Y.; Gu, H.; Bock, C.; Boyle, P.; Epstein, C.B.; Bernstein, B.E.; Lengauer, T.; et al. Genomic distribution and inter-sample variation of non-CpG methylation across human cell types. PLoS Genet. 2011, 7, e1002389. [Google Scholar] [CrossRef] [Green Version]
- Ramsahoye, B.H.; Biniszkiewicz, D.; Lyko, F.; Clark, V.; Bird, A.P.; Jaenisch, R. Non-CpG methylation is prevalent in embryonic stem cells and may be mediated by DNA methyltransferase 3a. Proc. Natl. Acad. Sci. USA 2000, 97, 5237–5242. [Google Scholar] [CrossRef] [Green Version]
- Takai, D.; Jones, P.A. Comprehensive analysis of CpG islands in human chromosomes 21 and 22. Proc. Natl. Acad. Sci. USA 2002, 99, 3740–3745. [Google Scholar] [CrossRef] [Green Version]
- Lander, E.S.; Linton, L.M.; Birren, B.; Nusbaum, C. International Human Genome Sequencing Consortium. Initial sequencing and analysis of the human genome. Nature 2001, 409, 860–921. [Google Scholar]
- Jones, P.A. Functions of DNA methylation: Islands, start sites, gene bodies and beyond. Nat. Rev. Genet. 2012, 13, 484–492. [Google Scholar] [CrossRef]
- Torres, R.F.; Kouro, R.; Kerr, B. Writers and Readers of DNA Methylation/Hydroxymethylation in Physiological Aging and Its Impact on Cognitive Function. Neural Plast. 2019, 2019, 5982625. [Google Scholar] [CrossRef] [Green Version]
- Moen, E.L.; Mariani, C.J.; Zullow, H.; Jeff-Eke, M.; Litwin, E.; Nikitas, J.N.; Godley, L.A. New themes in the biological functions of 5-methylcytosine and 5-hydroxymethylcytosine. Immunol. Rev. 2015, 263, 36–49. [Google Scholar] [CrossRef] [PubMed]
- Koh, K.P.; Yabuuchi, A.; Rao, S.; Huang, Y.; Cunniff, K.; Nardone, J.; Laiho, A.; Tahiliani, M.; Sommer, C.A.; Mostoslavsky, G.; et al. Tet1 and Tet2 regulate 5-hydroxymethylcytosine production and cell lineage specification in mouse embryonic stem cells. Cell Stem Cell 2011, 8, 200–213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kumar, S.; Chinnusamy, V.; Mohapatra, T. Epigenetics of Modified DNA Bases: 5-Methylcytosine and Beyond. Front. Genet. 2018, 9, 640. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Miller, J.L.; Grant, P.A. The role of DNA methylation and histone modifications in transcriptional regulation in humans. Subcell Biochem. 2013, 61, 289–317. [Google Scholar] [PubMed]
- Richmond, T.J.; Davey, C.A. The structure of DNA in the nucleosome core. Nature 2003, 423, 145–150. [Google Scholar] [CrossRef]
- Peterson, C.L.; Laniel, M.A. Histones and histone modifications. Curr. Biol. 2004, 14, R546–R551. [Google Scholar] [CrossRef] [Green Version]
- Kooistra, S.M.; Helin, K. Molecular mechanisms and potential functions of histone demethylases. Nat. Rev. Mol. Cell Biol. 2012, 13, 297–311. [Google Scholar] [CrossRef]
- Gujral, P.; Mahajan, V.; Lissaman, A.C.; Ponnampalam, A.P. Histone acetylation and the role of histone deacetylases in normal cyclic endometrium. Reprod. Biol. Endocrinol. 2020, 18, 84. [Google Scholar] [CrossRef]
- Rossetto, D.; Truman, A.W.; Kron, S.J.; Côté, J. Epigenetic modifications in double-strand break DNA damage signaling and repair. Clin. Cancer Res. 2010, 16, 4543–4552. [Google Scholar] [CrossRef] [Green Version]
- Rossetto, D.; Avvakumov, N. Histone phosphorylation: A chromatin modification involved in diverse nuclear events. Epigenetics 2012, 7, 1098–1108. [Google Scholar] [CrossRef] [Green Version]
- Turinetto, V.; Giachino, C. Histone variants as emerging regulators of embryonic stem cell identity. Epigenetics 2015, 10, 563–573. [Google Scholar] [CrossRef]
- Zha, J.J.; Tang, Y.; Wang, Y.L. Role of mono-ADP-ribosylation histone modification (Review). Exp. Ther. Med. 2021, 21, 577. [Google Scholar] [CrossRef]
- Messner, S.; Hottiger, M.O. Histone ADP-ribosylation in DNA repair, replication and transcription. Trends Cell Biol. 2011, 21, 534–542. [Google Scholar] [CrossRef]
- Mattiroli, F.; Penengo, L. Histone Ubiquitination: An Integrative Signaling Platform in Genome Stability. Trends Genet. 2021, 37, 566–581. [Google Scholar] [CrossRef]
- Ryu, H.-Y.; Hochstrasser, M. Histone sumoylation and chromatin dynamics. Nucleic Acids Res. 2021, 49, 6043–6052. [Google Scholar] [CrossRef]
- Shiio, Y.; Eisenman, R.N. Histone sumoylation is associated with transcriptional repression. Proc. Natl. Acad. Sci. USA 2003, 100, 13225–13230. [Google Scholar] [CrossRef] [Green Version]
- Mazzio, E.A.; Soliman, K.F. Basic concepts of epigenetics: Impact of environmental signals on gene expression. Epigenetics 2012, 7, 119–130. [Google Scholar] [CrossRef] [Green Version]
- Pal, S.; Tyler, J.K. Epigenetics and aging. Sci. Adv. 2016, 2, e1600584. [Google Scholar] [CrossRef] [Green Version]
- Hu, Z.; Chen, K.; Xia, Z.; Chavez, M.; Pal, S.; Seol, J.-H.; Chen, C.-C.; Li, W.; Tyler, J.K. Nucleo-some loss leads to global transcriptional up-regulation and genomic instability during yeast aging. Genes Dev. 2014, 28, 396–408. [Google Scholar] [CrossRef] [Green Version]
- Yi, S.-J.; Kim, K. New Insights into the Role of Histone Changes in Aging. Int. J. Mol. Sci. 2020, 21, 8241. [Google Scholar] [CrossRef]
- Moskalev, A.; Aliper, A.; Smit-McBride, Z.; Buzdin, A.; Zhavoronkov, A. Genetics and epigenetics of aging and longevity. Cell Cycle 2014, 13, 1063–1077. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sen, P.; Shah, P.P.; Nativio, R.; Berger, S.L. Epigenetic Mechanisms of Longevity and Aging. Cell 2016, 166, 822–839. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Miwa, S.; Kashyap, S.; Chini, E.; von Zglinicki, T. Mitochondrial dysfunction in cell senescence and aging. J. Clin. Invest. 2022, 132, e158447. [Google Scholar] [CrossRef] [PubMed]
- Vasileiou, P.V.; Evangelou, K.; Vlasis, K.; Fildisis, G.; Panayiotidis, M.I.; Chronopoulos, E.; Passias, P.-G.; Kouloukoussa, M.; Gorgoulis, V.G.; Havaki, S. Mitochondrial Homeostasis and Cellular Senescence. Cells 2019, 8, 686. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nagano, T.; Nakano, M.; Nakashima, A.; Onishi, K.; Yamao, S.; Enari, M.; Kikkawa, U.; Kamada, S. Identification of cellular senescence-specific genes by comparative transcriptomics. Sci. Rep. 2016, 6, 31758. [Google Scholar] [CrossRef] [Green Version]
- O’Sullivan, R.J.; Kubicek, S.; Schreiber, S.L.; Karlseder, J. Reduced histone biosynthesis and chromatin changes arising from a damage signal at telomeres. Nat. Struct. Mol. Biol. 2010, 17, 1218–1225. [Google Scholar] [CrossRef] [Green Version]
- Saul, D.; Kosinsky, R. Epigenetics of Aging and Aging-Associated Diseases. Int. J. Mol. Sci. 2021, 22, 401. [Google Scholar] [CrossRef]
- Tsurumi, A.; Li, W.X. Global heterochromatin loss: A unifying theory of aging? Epigenetics 2012, 7, 680–688. [Google Scholar] [CrossRef] [Green Version]
- Köhler, F.; Bormann, F.; Raddatz, G.; Gutekunst, J.; Corless, S.; Musch, T.; Lonsdorf, A.S.; Erhardt, S.; Lyko, F.; Rodríguez-Paredes, M. Epigenetic deregulation of lami-na-associated domains in Hutchinson-Gilford progeria syndrome. Genome Med. 2020, 12, 46. [Google Scholar] [CrossRef]
- Chen, Z.; Chang, W.Y.; Etheridge, A.; Strickfaden, H.; Jin, Z.; Palidwor, G.; Cho, J.-H.; Wang, K.; Kwon, S.Y.; Doré, C.; et al. Reprogramming progeria fibroblasts re-establishes a normal epigenetic landscape. Aging Cell 2017, 16, 870–887. [Google Scholar] [CrossRef]
- Chandra, T.; Narita, M. High-order chromatin structure and the epigenome in SAHFs. Nucleus 2013, 4, 23–28. [Google Scholar] [CrossRef] [Green Version]
- Aird, K.M.; Zhang, R. Detection of senescence-associated heterochromatin foci (SAHF). Methods Mol. Biol. 2013, 965, 185–196. [Google Scholar]
- Di Micco, R.; Sulli, G. Interplay between oncogene-induced DNA damage response and heterochromatin in senescence and cancer. Nat. Cell Biol. 2011, 13, 292–302. [Google Scholar] [CrossRef] [Green Version]
- Creighton, S.; Stefanelli, G.; Reda, A.; Zovkic, I. Epigenetic Mechanisms of Learning and Memory: Implications for Aging. Int. J. Mol. Sci. 2020, 21, 6918. [Google Scholar] [CrossRef]
- Rübe, C.E.; Bäumert, C.; Schuler, N.; Isermann, A.; Schmal, Z.; Glanemann, M.; Mann, C.; Scherthan, H. Human skin aging is associated with increased expression of the his-tone variant H2A.J in the epidermis. NPJ Aging Mech. Dis. 2021, 7, 7. [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] [Green Version]
- Li, L.; Greer, C.; Eisenman, R.N.; Secombe, J. Essential functions of the histone demethylase lid. PLoS Genet. 2010, 6, e1001221. [Google Scholar] [CrossRef] [Green Version]
- 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] [Green Version]
- Coppé, J.-P.; Desprez, P.-Y.; Krtolica, A.; Campisi, J. The senescence-associated secretory phenotype: The dark side of tumor suppression. Annu. Rev. Pathol. 2010, 5, 99–118. [Google Scholar] [CrossRef] [Green Version]
- 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] [Green Version]
- 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 Reports. 2017, 9, 1721–1734. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kawahara, T.L.A.; Michishita, E.; Adler, A.S.; Damian, M.; Berber, E.; Lin, M.; McCord, R.A.; Ongaigui, K.C.L.; Boxer, L.D.; Chang, H.Y.; et al. SIRT6 links histone H3 lysine 9 deacetylation to NF-kappaB-dependent gene expression and organismal lifespan. Cell 2009, 136, 62–74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mostoslavsky, R.; Chua, K.F.; Lombard, D.B.; Pang, W.W.; Fischer, M.R.; Gellon, L.; Liu, P.; Mostoslavsky, G.; Franco, S.; Murphy, M.M.; et al. Genomic instability and aging-like phenotype in the absence of mammalian SIRT6. Cell 2006, 124, 315–329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maleszewska, M.; Mawer, J.S.P.; Tessarz, P. Histone Modifications in Ageing and Lifespan Regulation. Curr. Mol. Bio. Rep. 2016, 2, 26–35. [Google Scholar] [CrossRef] [Green Version]
- Rao, J.S.; Keleshian, V.L.; Klein, S.; Rapoport, S.I. Epigenetic modifications in frontal cortex from Alzheimer’s disease and bipolar disorder patients. Transl. Psychiatry 2012, 2, e132. [Google Scholar] [CrossRef] [Green Version]
- Voss, J.L.; Bridge, D.J.; Cohen, N.J.; Walker, J.A. A Closer Look at the Hippocampus and Memory. Trends Cogn. Sci. 2017, 21, 577–588. [Google Scholar] [CrossRef]
- Morita, K.; Kantarjian, H.M.; Wang, F.; Yan, Y.; Bueso-Ramos, C.; Sasaki, K.; Issa, G.C.; Wang, S.; Jorgensen, J.; Song, X.; et al. Clearance of Somatic Mutations at Remission and the Risk of Relapse in Acute Myeloid Leukemia. J. Clin. Oncol. 2018, 36, 1788–1797. [Google Scholar] [CrossRef]
- Lee, J.-S.; Shukla, A.; Schneider, J.; Swanson, S.K.; Washburn, M.P.; Florens, L.; Bhaumik, S.R.; Shilatifard, A. Histone crosstalk between H2B monoubiquitination and H3 methyla-tion mediated by COMPASS. Cell 2007, 131, 1084–1096. [Google Scholar] [CrossRef] [Green Version]
- Rhie, B.-H.; Song, Y.-H.; Ryu, H.-Y.; Ahn, S.H. Cellular aging is associated with increased ubiquitylation of histone H2B in yeast telomeric heterochromatin. Biochem. Biophys Res. Commun. 2013, 439, 570–575. [Google Scholar] [CrossRef]
- Cruickshanks, H.A.; McBryan, T.; Nelson, D.M.; VanderKraats, N.D.; Shah, P.P.; van Tuyn, J.; Singh Rai, T.; Brock, C.; Donahue, G.; Dunican, D.S.; et al. Senescent cells harbour features of the cancer epigenome. Nat. Cell Biol. 2013, 15, 1495–1506. [Google Scholar] [CrossRef]
- 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]
- Booth, L.N.; Brunet, A. The Aging Epigenome. Mol. Cell 2016, 62, 728–744. [Google Scholar] [CrossRef] [Green Version]
- Campisi, J.; Yaswen, P. Aging and cancer cell biology, 2009. Aging Cell 2009, 8, 221–225. [Google Scholar] [CrossRef]
- Fraga, M.F.; Agrelo, R.; Esteller, M. Cross-talk between aging and cancer: The epigenetic language. Ann. NY Acad. Sci. 2007, 1100, 60–74. [Google Scholar] [CrossRef]
- Petronis, A. Epigenetics as a unifying principle in the aetiology of complex traits and diseases. Nature 2010, 465, 721–727. [Google Scholar] [CrossRef]
- Yu, M.; Hazelton, W.D.; Luebeck, G.E.; Grady, W.M. Epigenetic Aging: More Than Just a Clock When It Comes to Cancer. Cancer Res. 2020, 80, 367–374. [Google Scholar] [CrossRef] [Green Version]
- Yang, J.; Liu, M.; Hong, D.; Zeng, M.; Zhang, X. The Paradoxical Role of Cellular Senescence in Cancer. Front. Cell Dev. Biol. 2021, 9, 722205. [Google Scholar] [CrossRef]
- Regulski, M.J. Cellular Senescence: What, Why, and How. Wounds 2017, 29, 168–174. [Google Scholar]
- van Deursen, J.M. The role of senescent cells in ageing. Nature 2014, 509, 439–446. [Google Scholar] [CrossRef] [Green Version]
- Krtolica, A.; Parrinello, S.; Lockett, S.; Desprez, P.-Y.; Campisi, J. Senescent fibroblasts promote epithelial cell growth and tumorigenesis: A link between cancer and aging. Proc. Natl. Acad. Sci. USA 2001, 98, 12072–12077. [Google Scholar] [CrossRef] [Green Version]
- Davalos, A.R.; Coppe, J.-P.; Campisi, J.; Desprez, P.-Y. Senescent cells as a source of inflammatory factors for tumor progression. Cancer Metastasis Rev. 2010, 29, 273–283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lowe, R.; Overhoff, M.G.; Ramagopalan, S.V.; Garbe, J.C.; Koh, J.; Stampfer, M.R.; Beach, D.H.; Rakyan, V.K.; Bishop, C.L. The senescent methylome and its relationship with cancer, ageing and germline genetic variation in humans. Genome Biol. 2015, 16, 194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bellanger, A.; Madsen-Østerbye, J.; Galigniana, N.M.; Collas, P. Restructuring of Lamina-Associated Domains in Senescence and Cancer. Cells 2022, 11, 1846. [Google Scholar] [CrossRef] [PubMed]
- Sogabe, Y.; Seno, H. Unveiling epigenetic regulation in cancer, aging, and rejuvenation with in vivo reprogramming technology. Cancer Sci. 2018, 109, 2641–2650. [Google Scholar] [CrossRef]
- Kuipers, E.J.; Grady, W.M. Colorectal cancer. Nat. Rev. Dis. Primers. 2015, 1, 15065. [Google Scholar] [CrossRef] [Green Version]
- Zhu, Y.-M.; Huang, Q.; Lin, J.; Hu, Y.; Chen, J.; Lai, M.D. Expression of human DNA methyl-transferase 1 in colorectal cancer tissues and their corresponding distant normal tissues. Int. J. Colorectal. Dis. 2007, 22, 661–666. [Google Scholar] [CrossRef]
- Luo, Y.; Xie, C.; Brocker, C.N.; Fan, J.; Wu, X.; Feng, L.; Wang, Q.; Zhao, J.; Lu, D.; Tandon, M.; et al. Intestinal PPARα Protects Against Colon Carcinogenesis via Regulation of Methyltransferases DNMT1 and PRMT6. Gastroenterology 2019, 157, 744–759.e4. [Google Scholar] [CrossRef]
- Pyper, S.R.; Viswakarma, N.; Yu, S.; Reddy, J.K. PPARalpha: Energy combustion, hypolipidemia, inflammation and cancer. Nucl. Recept. Signal. 2010, 8, e002. [Google Scholar] [CrossRef] [Green Version]
- Toyota, M.; Ahuja, N.; Ohe-Toyota, M.; Herman, J.G.; Baylin, S.B.; Issa, J.P. CpG island methylator phenotype in colorectal cancer. Proc. Natl. Acad. Sci. USA 1999, 96, 8681–8686. [Google Scholar] [CrossRef] [Green Version]
- Yamashita, K.; Dai, T.; Dai, Y.; Yamamoto, F.; Perucho, M. Genetics supersedes epigenetics in colon cancer phenotype. Cancer Cell 2003, 4, 121–131. [Google Scholar] [CrossRef] [Green Version]
- Nazemalhosseini Mojarad, E.; Kuppen, P.J.; Aghdaei, H.A.; Zali, M.R. The CpG island methylator phenotype (CIMP) in colorectal cancer. Gastroenterol. Hepatol. Bed Bench 2013, 6, 120–128. [Google Scholar]
- Fedorova, M.S.; Krasnov, G.S.; Lukyanova, E.N.; Zaretsky, A.R.; Dmitriev, A.A.; Melnikova, N.V.; Moskalev, A.A.; Kharitonov, S.L.; Pudova, E.A.; Guvatova, Z.G.; et al. The CIMP-high phenotype is associated with energy metabolism alterations in colon adenocarcinoma. BMC Med. Genet. 2019, 20, 52. [Google Scholar] [CrossRef]
- Maugeri, A.; Barchitta, M.; Magnano San Lio, R.; Li Destri, G.; Agodi, A.; Basile, G. Epigenetic Aging and Colorectal Cancer: State of the Art and Perspectives for Future Research. Int. J. Mol. Sci. 2020, 22, 200. [Google Scholar] [CrossRef]
- Cervena, K.; Siskova, A.; Buchler, T.; Vodicka, P.; Vymetalkova, V. Methylation-Based Therapies for Colorectal Cancer. Cells 2020, 9, 1540. [Google Scholar] [CrossRef]
- Hsieh, C.J.; Klump, B.; Holzmann, K.; Borchard, F.; Gregor, M.; Porschen, R. Hypermethylation of the p16INK4a promoter in colectomy specimens of patients with long-standing and extensive ulcerative colitis. Cancer Res. 1998, 58, 3942–3945. [Google Scholar]
- O’Hagan, H.M. Chromatin modifications during repair of environmental exposure-induced DNA damage: A potential mechanism for stable epigenetic alterations. Environ. Mol. Mutagen. 2014, 55, 278–291. [Google Scholar] [CrossRef] [Green Version]
- Wei, S.; Li, C.; Yin, Z.; Wen, J.; Meng, H.; Xue, L.; Wang, J. Histone methylation in DNA repair and clinical practice: New findings during the past 5-years. J. Cancer 2018, 9, 2072–2081. [Google Scholar] [CrossRef]
- Treviño, L.S.; Wang, Q. Phosphorylation of epigenetic "readers, writers and erasers": Implications for developmental reprogramming and the epigenetic basis for health and disease. Prog. Biophys. Mol. Biol. 2015, 118, 8–13. [Google Scholar] [CrossRef] [Green Version]
- Karczmarski, J.; Rubel, T.; Paziewska, A.; Mikula, M.; Bujko, M.; Kober, P.; Dadlez, M.; Ostrowski, J. Histone H3 lysine 27 acetylation is altered in colon cancer. Clin. Proteom. 2014, 11, 24. [Google Scholar] [CrossRef] [Green Version]
- Ashktorab, H.; Belgrave, K.; Hosseinkhah, F.; Brim, H.; Nouraie, M.; Takkikto, M.; Hewitt, S.; Lee, E.L.; Dashwood, R.H.; Smoot, D. Global histone H4 acetylation and HDAC2 expression in colon adenoma and carcinoma. Dig. Dis. Sci. 2009, 54, 2109–2117. [Google Scholar] [CrossRef] [Green Version]
- Jun, J.-I.; Lau, L.F. Taking aim at the extracellular matrix: CCN proteins as emerging therapeutic targets. Nat. Rev. Drug. Discov. 2011, 10, 945–963. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kuonen, F.; Secondini, C.; Rüegg, C. Molecular pathways: Emerging pathways me-diating growth, invasion, and metastasis of tumors progressing in an irradiated mi-croenvironment. Clin. Cancer Res. 2012, 18, 5196–5202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jeong, D.; Heo, S.; Sung Ahn, T.; Lee, S.; Park, S.; Kim, H.; Park, D.; Byung Bae, S.; Lee, S.S.; Lee, M.S.; et al. Cyr61 expression is associated with prognosis in pa-tients with colorectal cancer. BMC Cancer 2014, 14, 164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tanaka, F.; Rizqiawan, A.; Higashikawa, K.; Tobiume, K.; Okui, G.; Shigeishi, H.; Ono, S.; Shimasue, H.; Kamata, N. Snail promotes Cyr61 secretion to prime collective cell migration and form invasive tumor nests in squamous cell carcinoma. Cancer Lett. 2013, 329, 243–252. [Google Scholar] [CrossRef] [PubMed]
- Xie, L.; Song, X.; Lin, H.; Chen, Z.; Li, Q.; Guo, T.; Xu, T.; Su, T.; Xu, M.; Chang, X.; et al. Aberrant activation of CYR61 enhancers in colorectal cancer de-velopment. J. Exp. Clin. Cancer Res. 2019, 38, 213. [Google Scholar] [CrossRef] [Green Version]
- Yokoyama, Y.; Hieda, M.; Nishioka, Y.; Matsumoto, A.; Higashi, S.; Kimura, H.; Yamamoto, H.; Mori, M.; Matsuura, S.; Matsuura, N. Cancer-associated upregulation of histone H3 lysine 9 trimethylation promotes cell motility in vitro and drives tumor formation in vivo. Cancer Sci. 2013, 104, 889–895. [Google Scholar] [CrossRef]
- Sharma, A.K.; Apurva, N.; Kumar, A.; Ali, A.; Saluja, S.S.; Prasad, B. Elevated Expression of Notch 2 & Notch 3 is associated with Disease Progression in Colorectal Cancer. Int. J. Appl. Biol. Pharm. Technol. 2022, 13, 033–050. [Google Scholar]
- Wang, Q.; Chen, X.; Jiang, Y.; Liu, S.; Liu, H.; Sun, X.; Zhang, H.; Liu, Z.; Tao, Y.; Li, C.; et al. Elevating H3K27me3 level sensitizes colo-rectal cancer to oxaliplatin. J. Mol. Cell Biol. 2020, 12, 125–137. [Google Scholar] [CrossRef]
- Noushmehr, H.; Weisenberger, D.J.; Diefes, K.; Phillips, H.S.; Pujara, K.; Berman, B.P.; Pan, F.; Pelloski, C.E.; Sulman, E.P.; Bhat, K.P.; et al. Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma. Cancer Cell. 2010, 17, 510–522. [Google Scholar] [CrossRef] [Green Version]
- Toyota, M.; Ahuja, N.; Suzuki, H.; Itoh, F.; Ohe-Toyota, M.; Imai, K.; Baylin, S.B.; Issa, J.-P. Aberrant methylation in gastric cancer associated with the CpG island methylator phenotype. Cancer Res. 1999, 59, 5438–5442. [Google Scholar]
- Huang, K.K.; Ramnarayanan, K.; Zhu, F.; Srivastava, S.; Xu, C.; Tan, A.L.K.; Lee, M.; Tay, S.; Das, K.; Xing, M.; et al. Genomic and epigenomic profiling of high-risk intestinal metaplasia reveals molecular determinants of progression to gastric cancer. Cancer Cell 2018, 33, 137–150. [Google Scholar] [CrossRef] [Green Version]
- Toyota, M.; Kopecky, K.J.; Toyota, M.O.; Jair, K.W.; Willman, C.L.; Issa, J.P. Methylation profiling in acute myeloid leukemia. Blood 2001, 97, 2823–2829. [Google Scholar] [CrossRef] [Green Version]
- Halperin, E.C.; Herndon, J.; Schold, S.; Brown, M.; Vick, N.; Cairncross, J.; Macdonald, D.R.; Gaspar, L.; Fischer, B.; Dropcho, E.; et al. A phase III randomized prospective trial of external beam radiotherapy, mitomycin C, carmustine, and 6-mercaptopurine for the treatment of adults with anaplastic glioma of the brain. CNS Cancer Consortium. Int. J. Radiat. Oncol. Biol. Phys. 1996, 34, 793–802. [Google Scholar] [CrossRef]
- Stewart, L.A. Chemotherapy in adult high-grade glioma: A systematic review and meta-analysis of individual patient data from 12 randomised trials. Lancet 2002, 359, 1011–1018. [Google Scholar]
- Bady, P.; Marosi, C.; Weller, M.; Grønberg, B.H.; Schultz, H.; Taphoorn, M.J.B.; Gijtenbeek, J.M.M.; van den Bent, M.J.; von Deimling, A.; Stupp, R.; et al. DNA methylation-based age acceleration observed in IDH wild-type glioblastoma is associated with better outcome-including in elderly patients. Acta Neuropathol. Commun. 2022, 10, 39. [Google Scholar] [CrossRef]
- Garagnani, P.; Bacalini, M.G.; Pirazzini, C.; Gori, D.; Giuliani, C.; Mari, D.; Di Blasio, A.M.; Gentilini, D.; Vitale, G.; Collino, S.; et al. Methylation of ELOVL2 gene as a new epigenetic marker of age. Aging Cell 2012, 11, 1132–1134. [Google Scholar] [CrossRef] [Green Version]
- Leonard, A.E.; Kelder, B.; Bobik, E.G.; Chuang, L.T.; Lewis, C.J.; Kopchick, J.J.; Mukerji, P.; Huang, Y.-S. Identification and expression of mammalian long-chain PUFA elongation enzymes. Lipids 2002, 37, 733–740. [Google Scholar] [CrossRef]
- Gimple, R.C.; Kidwell, R.L.; Kim, L.J.Y.; Sun, T.; Gromovsky, A.D.; Wu, Q.; Wolf, M.; Lv, D.; Bhargava, S.; Jiang, L.; et al. Glioma Stem Cell-Specific Superenhancer Promotes Polyunsaturated Fatty-Acid Synthesis to Support EGFR Signaling. Cancer Discov. 2019, 9, 1248–1267. [Google Scholar] [CrossRef]
- Behbahani, T.E.; Kahl, P.; von der Gathen, J.; Heukamp, L.C.; Baumann, C.; Gütgemann, I.; Walter, B.; Hofstädter, F.; Bastian, P.J.; von Ruecker, A.; et al. Alterations of global histone H4K20 methylation during prostate carcino-genesis. BMC Urol. 2012, 12, 5. [Google Scholar] [CrossRef] [Green Version]
- Yang, H.; Pesavento, J.J.; Starnes, T.W.; Cryderman, D.E.; Wallrath, L.L.; Kelleher, N.L.; Mizzen, C.A. Preferential dimethylation of histone H4 lysine 20 by Suv4-20. J. Biol. Chem. 2008, 283, 12085–12092. [Google Scholar] [CrossRef] [Green Version]
- López, V.; Tejedor, J.R.; Carella, A.; García, M.G.; Santamarina-Ojeda, P.; Pérez, R.F.; Mangas, C.; Urdinguio, R.G.; Aranburu, A.; de la Nava, D.; et al. Epigenetic Deregulation of the Histone Methyltransferase KMT5B Contributes to Malignant Transformation in Glioblastoma. Front. Cell Dev. Biol. 2021, 9, 671838. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Liang, J.; Hou, P. Hypermethylation in gastric cancer. Clin. Chim. Acta 2015, 448, 124–132. [Google Scholar] [CrossRef] [PubMed]
- Shenoy, S. CDH1 (E-cadherin) mutation and gastric cancer: Genetics, molecular mechanisms and guidelines for management. Cancer Manag. Res. 2019, 11, 10477–10486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, X.; Chu, K.M. E-cadherin and gastric cancer: Cause, consequence, and applications. Biomed Res. Int. 2014, 2014, 637308. [Google Scholar] [CrossRef] [Green Version]
- Graziano, F.; Arduini, F.; Ruzzo, A.; Mandolesi, A.; Bearzi, I.; Silva, R.; Muretto, P.; Testa, E.; Mari, D.; Magnani, M.; et al. Combined analysis of E-cadherin gene (CDH1) promoter hypermethylation and E-cadherin protein expression in patients with gastric cancer: Implications for treatment with demethylating drugs. Ann. Oncol. 2004, 15, 489–492. [Google Scholar] [CrossRef]
- Shin, C.M.; Kim, N.; Park, J.H.; Kang, G.H.; Kim, J.S.; Jung, H.C.; Song, I.S. Prediction of the risk for gastric cancer using candidate methylation markers in the non-neoplastic gastric mucosae. J. Pathol. 2012, 226, 654–665. [Google Scholar] [CrossRef]
- Abbaszadegan, M.R.; Moaven, O.; Sima, H.R.; Ghafarzadegan, K.; A’Rabi, A.; Forghani, M.N.; Raziee, H.R.; Mashhadinejad, A.; Jafarzadeh, M.; Esmaili-Shandiz, E.; et al. p16 promoter hypermethylation: A useful serum marker for early detection of gastric cancer. World. J. Gastroenterol. 2008, 14, 2055–2060. [Google Scholar] [CrossRef]
- Ichikawa, D.; Koike, H.; Ikoma, H.; Ikoma, D.; Tani, N.; Otsuji, E.; Kitamura, K.; Yamagishi, H. Detection of aberrant methylation as a tumor marker in serum of patients with gastric cancer. Anticancer Res. 2004, 24, 2477–2481. [Google Scholar]
- Dong, C.X.; Deng, D.J.; Pan, K.F.; Zhang, L.; Zhang, Y.; Zhou, J.; You, W.C. Promoter methylation of p16 associated with Helicobacter pylori infection in precancerous gastric lesions: A population-based study. Int. J. Cancer. 2009, 124, 434–439. [Google Scholar] [CrossRef]
- Ryan, J.L.; Jones, R.J.; Kenney, S.C.; Rivenbark, A.G.; Tang, W.; Knight, E.R.; Coleman, W.B.; Gulley, M.L. Epstein-Barr virus-specific methylation of human genes in gastric cancer cells. Infect. Agents Cancer 2010, 5, 27. [Google Scholar] [CrossRef] [Green Version]
- Li, G.M. Mechanisms and functions of DNA mismatch repair. Cell Res. 2008, 18, 85–98. [Google Scholar] [CrossRef] [Green Version]
- Harfe, B.D.; Jinks-Robertson, S. Mismatch repair proteins and mitotic genome stability. Mutat Res. 2000, 451, 151–167. [Google Scholar] [CrossRef]
- Li, Y.; Yang, Y.; Lu, Y.; Herman, J.G.; Brock, M.V.; Zhao, P.; Guo, M. Predictive value of CHFR and MLH1 methylation in human gastric cancer. Gastric Cancer 2015, 18, 280–287. [Google Scholar] [CrossRef]
- Yang, Q.; Wang, B.; Gao, W.; Huang, S.; Liu, Z.; Li, W.; Jia, J. SIRT1 is downregulated in gas-tric cancer and leads to G1-phase arrest via NF-κB/Cyclin D1 signaling. Mol. Cancer Res. 2013, 11, 1497–1507. [Google Scholar] [CrossRef] [Green Version]
- He, L.J.; Cai, M.Y.; Xu, G.L.; Li, J.J.; Weng, Z.J.; Xu, D.Z.; Luo, G.Y.; Zhu, S.L.; Xie, D. Prognostic sig-nificance of overexpression of EZH2 and H3k27me3 proteins in gastric cancer. Asian. Pac. J. Cancer Prev. 2012, 13, 3173–3178. [Google Scholar] [CrossRef] [Green Version]
- Yoruker, E.E.; Mert, U.; Bugra, D.; Yamaner, S.; Dalay, N. Promoter and histone methylation and p16(INK4A) gene expression in colon cancer. Exp. Ther. Med. 2012, 4, 865–870. [Google Scholar] [CrossRef] [Green Version]
- Lee, S.H.; Kim, J.; Kim, W.H.; Lee, Y.M. Hypoxic silencing of tumor suppressor RUNX3 by histone modification in gastric cancer cells. Oncogene 2009, 28, 184–194. [Google Scholar] [CrossRef] [Green Version]
- Ito, K.; Liu, Q.; Salto-Tellez, M.; Yano, T.; Tada, K.; Ida, H.; Huang, C.; Shah, N.; Inoue, M.; Rajnakova, A.; et al. RUNX3, a novel tumor suppressor, is frequently inactivated in gastric cancer by protein mislocalization. Cancer Res. 2005, 65, 7743–7750. [Google Scholar] [CrossRef] [Green Version]
- Fahrner, J.A.; Eguchi, S.; Herman, J.G.; Baylin, S.B. Dependence of histone modifications and gene expression on DNA hypermethylation in cancer. Cancer Res. 2002, 62, 7213–7218. [Google Scholar]
- Zjablovskaja, P.; Florian, M.C. Acute Myeloid Leukemia: Aging and Epigenetics. Cancers 2019, 12, 103. [Google Scholar] [CrossRef] [Green Version]
- Silva, P.; Neumann, M.; Schroeder, M.P.; Vosberg, S.; Schlee, C.; Isaakidis, K.; Ortiz-Tanchez, J.; Fransecky, L.R.; Hartung, T.; Türkmen, S.; et al. Acute myeloid leukemia in the elderly is characterized by a distinct genetic and epigenetic landscape. Leukemia 2017, 31, 1640–1644. [Google Scholar] [CrossRef] [PubMed]
- Sun, D.; Luo, M.; Jeong, M.; Rodriguez, B.; Xia, Z.; Hannah, R.; Wang, H.; Le, T.; Faull, K.F.; Chen, R.; et al. Epigenomic profiling of young and aged HSCs reveals concerted changes during aging that reinforce self-renewal. Cell Stem Cell 2014, 14, 673–688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Adelman, E.R.; Huang, H.T.; Roisman, A.; Olsson, A.; Colaprico, A.; Qin, T.; Lindsley, R.C.; Bejar, R.; Salomonis, N.; Grimes, H.L.; et al. Aging Human Hematopoietic Stem Cells Manifest Profound Epigenetic Reprogramming of Enhancers That May Predispose to Leukemia. Cancer Discov. 2019, 9, 1080–1101. [Google Scholar] [CrossRef] [PubMed]
- Djeghloul, D.; Kuranda, K.; Kuzniak, I.; Barbieri, D.; Naguibneva, I.; Choisy, C.; Bories, J.C.; Dosquet, C.; Pla, M.; Vanneaux, V. 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] [Green Version]
- Geiger, H.; de Haan, G.; Florian, M.C. The ageing haematopoietic stem cell compartment. Nat. Rev. Immunol. 2013, 13, 376–389. [Google Scholar] [CrossRef]
- de Haan, G.; Lazare, S.S. Aging of hematopoietic stem cells. Blood 2018, 131, 479–487. [Google Scholar] [CrossRef] [Green Version]
- Prassek, V.V.; Rothenberg-Thurley, M.; Sauerland, M.C.; Herold, T.; Janke, H.; Ksienzyk, B.; Konstandin, N.P.; Goerlich, D.; Krug, U.; Faldum, A.; et al. Genetics of acute myeloid leukemia in the elderly: Mutation spectrum and clinical impact in intensively treated patients aged 75 years or older. Haematologica 2018, 103, 1853–1861. [Google Scholar] [CrossRef] [Green Version]
- Sen, P.; Lan, Y.; Li, C.Y.; Sidoli, S.; Donahue, G.; Dou, Z.; Frederick, B.; Chen, Q.; Luense, L.J.; Garcia, B.A.; et al. Histone Acetyltransferase p300 Induces De Novo Super-Enhancers to Drive Cellular Senescence. Mol. Cell 2019, 73, 684–698.e8. [Google Scholar] [CrossRef] [Green Version]
- Deneberg, S.; Grövdal, M.; Karimi, M.; Jansson, M.; Nahi, H.; Corbacioglu, A.; Gaidzik, V.; Döhner, K.; Paul, C.; Ekström, T.J.; et al. Gene-specific and global methylation patterns predict outcome in patients with acute myeloid leuke-mia. Leukemia 2010, 24, 932–941. [Google Scholar] [CrossRef] [Green Version]
- Milani, L.; Lundmark, A.; Kiialainen, A.; Nordlund, J.; Flaegstad, T.; Forestier, E.; Heyman, M.; Jonmundsson, G.; Kanerva, J.; Schmiegelow, K.; et al. DNA methylation for subtype classification and prediction of treatment outcome in patients with childhood acute lymphoblastic leukemia. Blood 2010, 115, 1214–1225. [Google Scholar] [CrossRef] [Green Version]
- Corn, P.G.; Smith, B.D.; Ruckdeschel, E.S.; Douglas, D.; Baylin, S.B.; Herman, J.G. E-cadherin expression is silenced by 5’ CpG island methylation in acute leukemia. Clin. Cancer Res. 2000, 6, 4243–4248. [Google Scholar]
- Feng, M.; Huang, B.; Du, Z.; Xu, X.; Chen, Z. DLC-1 as a modulator of proliferation, apoptosis and migration in Burkitt’s lymphoma cells. Mol. Biol. Rep. 2011, 38, 1915–1920. [Google Scholar] [CrossRef]
- Kroeger, H.; Jelinek, J.; Estécio, M.R.H.; He, R.; Kondo, K.; Chung, W.; Zhang, L.; Shen, L.; Kantarjian, H.M.; Bueso-Ramos, C.E.; et al. Aberrant CpG island methylation in acute myeloid leukemia is accentuated at relapse. Blood 2008, 112, 1366–1373. [Google Scholar] [CrossRef]
- Gensous, N.; Bacalini, M.G.; Pirazzini, C.; Marasco, E.; Giuliani, C.; Ravaioli, F.; Mengozzi, G.; Bertarelli, C.; Palmas, M.G.; Franceschi, C.; et al. The epigenetic landscape of age-related diseases: The geroscience perspective. Biogerontology 2017, 18, 549–559. [Google Scholar] [CrossRef] [Green Version]
- Rodgers, J.L.; Jones, J.; Bolleddu, S.I.; Vanthenapalli, S.; Rodgers, L.E.; Shah, K.; Karia, K.; Panguluri, S.K. Cardiovascular Risks Associated with Gender and Aging. J. Cardiovasc. Dev. Dis. 2019, 6, 19. [Google Scholar] [CrossRef] [Green Version]
- North, B.J.; Sinclair, D.A. The intersection between aging and cardiovascular disease. Circ. Res. 2012, 110, 1097–1108. [Google Scholar] [CrossRef]
- Yazdanyar, A.; Newman, A.B. The burden of cardiovascular disease in the elderly: Morbidity, mortality, and costs. Clin. Geriatr. Med. 2009, 25, 563–577. [Google Scholar] [CrossRef] [Green Version]
- Xu, Q.; Lin, X.; Andrews, L.; Patel, D.; Lampe, P.D.; Veenstra, R.D. Histone deacetylase inhibition reduces cardiac connexin43 expression and gap junction communication. Front. Pharmacol. 2013, 4, 44. [Google Scholar] [CrossRef] [Green Version]
- Wang, Y.; Miao, X.; Liu, Y.; Li, F.; Liu, Q.; Sun, J.; Cai, L. Dysregulation of histone acetyltransferases and deacetylases in cardiovascular diseases. Oxid. Med. Cell. Longev. 2014, 2014, 641979. [Google Scholar] [CrossRef] [Green Version]
- Westerman, K.; Sebastiani, P.; Jacques, P.; Liu, S.; DeMeo, D.; Ordovás, J.M. DNA methylation modules associate with incident cardiovascular disease and cumulative risk factor exposure. Clin. Epigenetics. 2019, 11, 142. [Google Scholar] [CrossRef] [Green Version]
- Kennel, P.J.; Liao, X.; Saha, A.; Ji, R.; Zhang, X.; Castillero, E.; Brunjes, D.; Takayama, H.; Naka, Y.; Thomas, T.; et al. Impairment of Myocardial Glutamine Homeostasis Induced By Suppression of the Amino Acid Carrier SLC1A5 in Failing Myocardium. Circ. Heart Fail. 2019, 12, e006336. [Google Scholar] [CrossRef]
- Fernández-Sanlés, A.; Sayols-Baixeras, S.; Subirana, I.; Sentí, M.; Pérez-Fernández, S.; de Castro Moura, M.; Esteller, M.; Marrugat, J.; Elosua, R. DNA methylation biomarkers of myocardial infarction and cardiovascular disease. Clin. Epigenetics. 2021, 13, 86. [Google Scholar] [CrossRef] [PubMed]
- Glezeva, N.; Moran, B.; Collier, P.; Moravec, C.S.; Phelan, D.; Donnellan, E.; Russell-Hallinan, A.; O’Connor, D.P.; Gallagher, W.M.; Gallagher, J.; et al. Targeted DNA Methylation Profiling of Human Cardiac Tissue Reveals Novel Epigenetic Traits and Gene Deregulation Across Different Heart Failure Patient Subtypes. Circ. Heart Fail. 2019, 12, e005765. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gentilini, D.; Garagnani, P.; Pisoni, S.; Bacalini, M.G.; Calzari, L.; Mari, D.; Vitale, G.; Franceschi, C.; Di Blasio, A.M. Stochastic epigenetic mutations (DNA methylation) increase exponentially in human aging and correlate with X chromosome inactivation skewing in females. Aging 2015, 7, 568–578. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Han, P.; Hang, C.T.; Yang, J. Chromatin remodeling in cardiovascular development and physiology. Circ. Res. 2011, 108, 378–396. [Google Scholar] [CrossRef] [Green Version]
- Lee, D.Y.; Lee, C.I.; Lin, T.E.; Lim, S.H.; Zhou, J.; Tseng, Y.C.; Chien, S.; Chiu, J.J. Role of histone deacetylases in transcription factor regulation and cell cycle modulation in endo-thelial cells in response to disturbed flow. Proc. Natl. Acad. Sci. USA 2012, 109, 1967–1972. [Google Scholar] [CrossRef] [Green Version]
- Khyzha, N.; Alizada, A.; Wilson, M.D.; Fish, J.E. Epigenetics of Atherosclerosis: Emerging Mechanisms and Methods. Trends Mol. Med. 2017, 23, 332–347. [Google Scholar] [CrossRef]
- Wolf, D.; Ley, K. Immunity and Inflammation in Atherosclerosis. Circ. Res. 2019, 124, 315–327. [Google Scholar] [CrossRef]
- Zhong, H.; May, M.J.; Jimi, E.; Ghosh, S. The phosphorylation status of nuclear NF-kappa B determines its association with CBP/p300 or HDAC-1. Mol. Cell 2002, 9, 625–636. [Google Scholar] [CrossRef]
- Brown, J.D.; Lin, C.Y.; Duan, Q.; Griffin, G.; Federation, A.; Paranal, R.M.; Bair, S.; Newton, G.; Lichtman, A.; Kung, A.; et al. NF-κB directs dynamic super enhancer formation in inflammation and atherogenesis. Mol. Cell 2014, 56, 219–231. [Google Scholar] [CrossRef] [Green Version]
- Han, Y.; Nie, J.; Wang, D.W.; Ni, L. Mechanism of histone deacetylases in cardiac hypertrophy and its therapeutic inhibitors. Front. Cardiovasc. Med. 2022, 9, 931475. [Google Scholar] [CrossRef]
- Shea, T.B.; Rogers, E.; Ashline, D.; Ortiz, D.; Sheu, M.S. Apolipoprotein E deficiency pro-motes increased oxidative stress and compensatory increases in antioxidants in brain tissue. Free Radic. Biol. Med. 2002, 33, 1115–1120. [Google Scholar] [CrossRef]
- Tecalco-Cruz, A.C.; O Ramírez-Jarquín, J.; E Alvarez-Sánchez, M.; Zepeda-Cervantes, J. Epigenetic basis of Alzheimer disease. World J. Biol. Chem. 2020, 11, 62–75. [Google Scholar] [CrossRef]
- Available online: https://www.alzint.org/about/dementia-facts-figures/dementia-statistics/ (accessed on 4 November 2022).
- Kumar, A.; Sidhu, J.; Goyal, A.; Tsao, J.W. Alzheimer Disease. 2022 June 5. In StatPearls [Internet]; StatPearls Publishing: Treasure Island, FL, USA, 2023. [Google Scholar]
- 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] [Green Version]
- Fischer, A.; Sananbenesi, F.; Wang, X.; Dobbin, M.; Tsai, L.H. Recovery of learning and memory is associated with chromatin remodelling. Nature 2007, 447, 178–182. [Google Scholar] [CrossRef]
- Levenson, J.M.; O’Riordan, K.J.; Brown, K.D.; Trinh, M.A.; Molfese, D.L.; Sweatt, J.D. Regulation of histone acetylation during memory formation in the hippocampus. J. Biol. Chem. 2004, 279, 40545–40559. [Google Scholar] [CrossRef] [Green Version]
- 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] [Green Version]
- Gräff, J.; Rei, D.; Guan, J.S.; Wang, W.Y.; Seo, J.; Hennig, K.M.; Nieland, T.J.F.; Fass, D.M.; Kao, P.F.; Kahn, M.; et al. An epigenetic blockade of cognitive functions in the neurodegenerating brain. Nature 2012, 483, 222–226. [Google Scholar] [CrossRef] [Green Version]
- Walker, M.P.; LaFerla, F.M.; Oddo, S.S.; Brewer, G.J. Reversible epigenetic histone modifications and Bdnf expression in neurons with aging and from a mouse model of Alzheimer’s disease. Age 2013, 35, 519–531. [Google Scholar] [CrossRef]
- Dolinar, A.; Ravnik-Glavač, M.; Glavač, D. Epigenetic mechanisms in amyotrophic lateral sclerosis: A short review. Mech. Ageing Dev. 2018, 174, 103–110. [Google Scholar] [CrossRef]
- Ebbert, M.T.W.; Ross, C.A.; Pregent, L.J.; Lank, R.J.; Zhang, C.; Katzman, R.B.; Jansen-West, K.; Song, Y.; da Rocha, E.L.; Palmucci, C.; et al. Conserved DNA methylation combined with differential frontal cortex and cerebellar expression distinguishes C9orf72-associated and sporadic ALS, and implicates SERPINA1 in disease. Acta Neuropathol. 2017, 134, 715–728. [Google Scholar] [CrossRef] [PubMed]
- Mastroeni, D.; McKee, A.; Grover, A.; Rogers, J.; Coleman, P.D. Epigenetic differences in cortical neurons from a pair of monozygotic twins discordant for Alzheimer’s disease. PLoS ONE 2009, 4, e6617. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- DeKosky, S.T.; Scheff, S.W. Synapse loss in frontal cortex biopsies in Alzheimer’s disease: Correlation with cognitive severity. Ann. Neurol. 1990, 27, 457–464. [Google Scholar] [CrossRef] [PubMed]
- Coppieters, N.; Dieriks, B.V.; Lill, C.; Faull, R.L.M.; Curtis, M.A.; Dragunow, M. Global changes in DNA methylation and hydroxymethylation in Alzheimer’s disease human brain. Neurobiol. Aging. 2014, 35, 1334–1344. [Google Scholar] [CrossRef]
- Iwata, A.; Nagata, K.; Hatsuta, H.; Takuma, H.; Bundo, M.; Iwamoto, K.; Tamaoka, A.; Murayama, S.; Saido, T.; Tsuji, S. Altered CpG methylation in sporadic Alzheimer’s disease is associated with APP and MAPT dysregulation. Hum. Mol. Genet. 2014, 23, 648–656. [Google Scholar] [CrossRef]
- Yu, L.; Chibnik, L.B.; Srivastava, G.P.; Pochet, N.; Yang, J.; Xu, J.; Kozubek, J.; Obholzer, N.; Leurgans, S.E.; Schneider, J.A.; et al. Association of Brain DNA methylation in SORL1, ABCA7, HLA-DRB5, SLC24A4, and BIN1 with pathological diagnosis of Alzheimer disease. JAMA Neurol. 2015, 72, 15–24. [Google Scholar] [CrossRef] [Green Version]
- Sun, X.Y.; Zheng, T.; Yang, X.; Liu, L.; Gao, S.S.; Xu, H.B.; Song, Y.T.; Tong, K.; Yang, L.; Gao, Y.; et al. HDAC2 hyperexpression alters hippocampal neuronal transcription and microglial activity in neuroinflammation-induced cognitive dysfunction. J. Neuroinflamm. 2019, 16, 249. [Google Scholar] [CrossRef] [Green Version]
- Spillantini, M.G.; Schmidt, M.L.; Lee, V.M.; Trojanowski, J.Q.; Jakes, R.; Goedert, M. Alpha-synuclein in Lewy bodies. Nature 1997, 388, 839–840. [Google Scholar] [CrossRef]
- Jowaed, A.; Schmitt, I.; Kaut, O.; Wüllner, U. Methylation regulates alpha-synuclein expression and is decreased in Parkinson’s disease patients’ brains. J. Neurosci. 2010, 30, 6355–6359. [Google Scholar] [CrossRef] [Green Version]
- Matsumoto, L.; Takuma, H.; Tamaoka, A.; Kurisaki, H.; Date, H.; Tsuji, S.; Iwata, A. CpG demethylation enhances alpha-synuclein expression and affects the pathogenesis of Parkinson’s disease. PLoS ONE 2010, 5, e15522. [Google Scholar] [CrossRef] [Green Version]
- Desplats, P.; Spencer, B.; Coffee, E.; Patel, P.; Michael, S.; Patrick, C.; Adame, A.; Rockenstein, E.; Masliah, E. Alpha-synuclein sequesters Dnmt1 from the nucleus: A novel mechanism for epigenetic alterations in Lewy body diseases. J. Biol. Chem. 2011, 286, 9031–9037. [Google Scholar] [CrossRef] [Green Version]
- Jiang, W.; Li, J.; Zhang, Z.; Wang, H.; Wang, Z. Epigenetic upregulation of alpha-synuclein in the rats exposed to methamphetamine. Eur. J. Pharmacol. 2014, 745, 243–248. [Google Scholar] [CrossRef]
- Schmitt, I.; Kaut, O.; Khazneh, H.; de Boni, L.; Ahmad, A.; Berg, D.; Klein, C.; Fröhlich, H.; Wüllner, U. L-dopa increases α-synuclein DNA methylation in Parkinson’s disease patients in vivo and in vitro. Mov. Disord. 2015, 30, 1794–1801. [Google Scholar] [CrossRef]
- de Lau, L.M.; Breteler, M.M. Epidemiology of Parkinson’s disease. Lancet Neurol. 2006, 5, 525–535. [Google Scholar] [CrossRef]
- Voutsinas, G.E.; Stavrou, E.F.; Karousos, G.; Dasoula, A.; Papachatzopoulou, A.; Syrrou, M.; Verkerk, A.J.; van der Spek, P.; Patrinos, G.P.; Stöger, R.; et al. Allelic imbalance of expression and epigenetic regulation within the alpha-synuclein wild-type and p.Ala53Thr alleles in Parkinson disease. Hum. Mutat. 2010, 31, 685–691. [Google Scholar] [CrossRef] [Green Version]
- Mittal, S.; Bjørnevik, K.; Im, D.S.; Flierl, A.; Dong, X.; Locascio, J.J.; Abo, K.M.; Long, E.; Jin, M.; Xu, B.; et al. β2-Adrenoreceptor is a regulator of the α-synuclein gene driving risk of Parkinson’s disease. Science 2017, 357, 891–898. [Google Scholar] [CrossRef] [Green Version]
- Gebremedhin, K.G.; Rademacher, D.J. Histone H3 acetylation in the postmortem Parkinson’s disease primary motor cortex. Neurosci. Lett. 2016, 627, 121–125. [Google Scholar] [CrossRef] [Green Version]
- Galicia-Garcia, U.; Benito-Vicente, A.; Jebari, S.; Larrea-Sebal, A.; Siddiqi, H.; Uribe, K.; Ostolaza, H.; Martín, C. Pathophysiology of Type 2 Diabetes Mellitus. Int. J. Mol. Sci. 2020, 21, 6275. [Google Scholar] [CrossRef]
- Ling, C.; Rönn, T. Epigenetics in Human Obesity and Type 2 Diabetes. Cell Metab. 2019, 29, 1028–1044. [Google Scholar] [CrossRef] [Green Version]
- Hall, E.; Dekker Nitert, M.; Volkov, P.; Malmgren, S.; Mulder, H.; Bacos, K.; Ling, C. The effects of high glucose exposure on global gene expression and DNA methylation in human pancreatic islets. Mol. Cell Endocrinol. 2018, 472, 57–67. [Google Scholar] [CrossRef]
- Dhawan, S.; Georgia, S.; Tschen, S.I.; Fan, G.; Bhushan, A. Pancreatic β cell identity is maintained by DNA methylation-mediated repression of Arx. Dev. Cell. 2011, 20, 419–429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, B.T.; Dayeh, T.A.; Volkov, P.A.; Kirkpatrick, C.L.; Malmgren, S.; Jing, X.; Renström, E.; Wollheim, C.B.; Nitert, M.D.; Ling, C. Increased DNA methylation and decreased expression of PDX-1 in pancreatic islets from patients with type 2 diabetes. Mol. Endocrinol. 2012, 26, 1203–1212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ishikawa, K.; Tsunekawa, S.; Ikeniwa, M.; Izumoto, T.; lida, A.; Ogata, H.; Uenishi, E.; Seino, Y.; Ozaki, N.; Sugimura, Y.; et al. Long-term pancreatic beta cell exposure to high levels of glucose but not palmitate induces DNA methylation within the insulin gene promoter and represses transcriptional activity. PLoS ONE 2015, 10, e0115350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dayeh, T.; Volkov, P.; Salö, S.; Hall, E.; Nilsson, E.; Olsson, A.H.; Kirkpatrick, C.; Wollheim, C.B.; Eliasson, L.; Rönn, T.; et al. Genome-wide DNA methylation analysis of human pancreatic islets from type 2 diabetic and non-diabetic donors identifies candidate genes that influence insulin secretion. PLoS Genet. 2014, 10, e1004160. [Google Scholar] [CrossRef]
- Cruz-Jentoft, A.J.; Bahat, G. Writing Group for the European Working Group on Sarcopenia in Older People 2 (EWGSOP2), and the Extended Group for EWGSOP2. Sarcopenia: Revised European consensus on definition and diagnosis. Age Ageing 2019, 48, 16–31. [Google Scholar] [CrossRef] [Green Version]
- Matteini, A.M.; Tanaka, T.; Karasik, D.; Atzmon, G.; Chou, W.; Eicher, J.D.; Johnson, A.D.; Arnold, A.M.; Callisaya, M.L.; Davies, G.; et al. GWAS analysis of handgrip and lower body strength in older adults in the CHARGE consortium. Aging Cell 2016, 15, 792–800. [Google Scholar] [CrossRef] [Green Version]
- Turner, D.C.; Gorski, P.P.; Maasar, M.F.; Seaborne, R.A.; Baumert, P.; Brown, A.D.; Kitchen, M.O.; Erskine, R.M.; Dos-Remedios, I.; Voisin, S.; et al. DNA methylation across the genome in aged human skeletal muscle tissue and muscle-derived cells: The role of HOX genes and physical activity. Sci. Rep. 2020, 10, 15360. [Google Scholar] [CrossRef]
- Zykovich, A.; Hubbard, A.; Flynn, J.M.; Tarnopolsky, M.; Fraga, M.F.; Kerksick, C.; Ogborn, D.; MacNeil, L.; Mooney, S.D.; Melov, S. Genome-wide DNA methylation changes with age in disease-free human skeletal muscle. Aging Cell 2014, 13, 360–366. [Google Scholar] [CrossRef] [Green Version]
- Voisin, S.; Jacques, M.; Landen, S.; Harvey, N.R.; Haupt, L.M.; Griffiths, L.R.; Gancheva, S.; Ouni, M.; Jähnert, M.; Ashton, K.J.; et al. Meta-analysis of genome-wide DNA methylation and integrative omics of age in human skeletal muscle. J. Cachexia Sarcopenia Muscle 2021, 12, 1064–1078. [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, R102. [Google Scholar] [CrossRef] [Green Version]
- Xu, F.; Li, W.; Yang, X.; Na, L.; Chen, L.; Liu, G. The Roles of Epigenetics Regulation in Bone Metabolism and Osteoporosis. Front. Cell Dev. Biol. 2021, 8, 619301. [Google Scholar] [CrossRef]
- Wang, R.; Wang, Y.; Zhu, L.; Liu, Y.; Li, W. Epigenetic Regulation in Mesenchymal Stem Cell Aging and Differentiation and Osteoporosis. Stem Cells Int. 2020, 2020, 8836258. [Google Scholar] [CrossRef]
- Delgado-Calle, J.; Fernández, A.F.; Sainz, J.; Zarrabeitia, M.T.; Sañudo, C.; García-Renedo, R.; Pérez-Núñez, M.I.; García-Ibarbia, C.; Fraga, M.F.; Riancho, J.A. Genome-wide profiling of bone reveals differentially methylated regions in osteoporosis and osteoarthritis. Arthritis Rheum. 2013, 65, 197–205. [Google Scholar] [CrossRef]
- Migliavacca, E.; Tay, S.K.H.; Patel, H.P.; Sonntag, T.; Civiletto, G.; McFarlane, C.; Forrester, T.; Barton, S.J.; Leow, M.K.; Antoun, E.; et al. Mitochondrial oxidative capacity and NAD+ biosynthesis are reduced in human sarcopenia across ethnicities. Nat. Commun. 2019, 10, 5808. [Google Scholar] [CrossRef] [Green Version]
- Nuti, R.; Brandi, M.L.; Checchia, G.; Di Munno, O.; Dominguez, L.; Falaschi, P.; Fiore, C.E.; Iolascon, G.; Maggi, S.; Michieli, R.; et al. Guidelines for the management of osteoporosis and fragility fractures. Intern. Emerg. Med. 2019, 14, 85–102. [Google Scholar] [CrossRef] [Green Version]
- Antoun, E.; Garratt, E.S.; Taddei, A.; Burton, M.A.; Barton, S.J.; Titcombe, P.; Westbury, L.D.; Baczynska, A.; Migliavacca, E.; Feige, J.N.; et al. EpiGen Global Research Consortium. Epigenome-wide association study of sarcopenia: Findings from the Hertfordshire Sarcopenia Study (HSS). J. Cachexia Sarcopenia Muscle 2022, 13, 240–253. [Google Scholar] [CrossRef]
- Reppe, S.; Noer, A.; Grimholt, R.M.; Halldórsson, B.V.; Medina-Gomez, C.; Gautvik, V.T.; Olstad, O.K.; Berg, J.P.; Datta, H.; Estrada, K.; et al. Methylation of bone SOST, its mRNA, and serum sclerostin levels correlate strongly with fracture risk in postmenopausal women. J. Bone Miner. Res. 2015, 30, 249–256. [Google Scholar] [CrossRef]
- Delgado-Calle, J.; Sañudo, C.; Bolado, A.; Fernández, A.F.; Arozamena, J.; Pascual-Carra, M.A.; Rodriguez-Rey, J.C.; Fraga, M.F.; Bonewald, L.; A Riancho, J. DNA methylation contributes to the regulation of sclerostin expression in human osteocytes. J. Bone Miner. Res. 2012, 27, 926–937. [Google Scholar] [CrossRef]
- Del Real, A.; Pérez-Campo, F.M.; Fernández, A.F.; Sañudo, C.; Ibarbia, C.G.; Pérez-Núñez, M.I.; Criekinge, W.V.; Braspenning, M.; Alonso, M.A.; Fraga, M.F.; et al. Differential analysis of genome-wide methylation and gene expression in mesenchymal stem cells of patients with fractures and osteoarthritis. Epigenetics 2017, 12, 113–122. [Google Scholar] [CrossRef]
- Bork, S.; Pfister, S.; Witt, H.; Horn, P.; Korn, B.; Ho, A.D.; Wagner, W. DNA methylation pattern changes upon long-term culture and aging of human mesenchymal stromal cells. Aging Cell. 2010, 9, 54–63. [Google Scholar] [CrossRef] [Green Version]
- Cakouros, D.; Hemming, S.; Gronthos, K.; Liu, R.; Zannettino, A.; Shi, S.; Gronthos, S. Specific functions of TET1 and TET2 in regulating mesenchymal cell lineage determination. Epigenetics Chromatin 2019, 12, 3. [Google Scholar] [CrossRef] [PubMed]
- Cakouros, D.; Gronthos, S. Epigenetic Regulators of Mesenchymal Stem/Stromal Cell Lineage Determination. Curr. Osteoporos. Rep. 2020, 18, 597–605. [Google Scholar] [CrossRef] [PubMed]
- Sinha, K.M.; Zhou, X. Genetic and molecular control of osterix in skeletal formation. J. Cell. Biochem. 2013, 114, 975–984. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, R.; Yu, T.; Kou, X.; Gao, X.; Chen, C.; Liu, D.; Zhou, Y.; Shi, S. Tet1 and Tet2 maintain mesenchymal stem cell homeostasis via demethylation of the P2rX7 promoter. Nat. Commun. 2018, 9, 2143. [Google Scholar] [CrossRef]
- Ren, J.; Huang, D.; Li, R.; Wang, W.; Zhou, C. Control of mesenchymal stem cell biology by histone modifications. Cell Biosci. 2020, 10, 11. [Google Scholar] [CrossRef]
- de Nigris, F.; Ruosi, C.; Colella, G.; Napoli, C. Epigenetic therapies of osteoporosis. Bone 2021, 142, 115680. [Google Scholar] [CrossRef]
- Jing, H.; Liao, L.; An, Y.; Su, X.; Liu, S.; Shuai, Y.; Zhang, X.; Jin, Y. Suppression of EZH2 Prevents the Shift of Osteoporotic MSC Fate to Adipocyte and Enhances Bone Formation During Osteoporosis. Mol. Ther. 2016, 24, 217–229. [Google Scholar] [CrossRef] [Green Version]
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
la Torre, A.; Lo Vecchio, F.; Greco, A. Epigenetic Mechanisms of Aging and Aging-Associated Diseases. Cells 2023, 12, 1163. https://doi.org/10.3390/cells12081163
la Torre A, Lo Vecchio F, Greco A. Epigenetic Mechanisms of Aging and Aging-Associated Diseases. Cells. 2023; 12(8):1163. https://doi.org/10.3390/cells12081163
Chicago/Turabian Stylela Torre, Annamaria, Filomena Lo Vecchio, and Antonio Greco. 2023. "Epigenetic Mechanisms of Aging and Aging-Associated Diseases" Cells 12, no. 8: 1163. https://doi.org/10.3390/cells12081163