Driver or Passenger: Epigenomes in Alzheimer’s Disease
Abstract
:1. Introduction
2. The Genetic Architecture of AD
3. Epigenomics—Beyond Sequence Variation
4. A Multifaceted Brain Methylome
4.1. Canonical DNA Methylation
4.2. Non-CG Methylation
4.3. Hydroxymethylcytosine
5. Epigenomics Landscapes in Alzheimer’s Disease
6. Conclusions
Acknowledgments
Author Contributions
Conflicts of Interest
References
- Ames, D.; O’Brien, J.; Burns, A. Dementia, 5th ed.; CRC Pr I Llc: Boca Raton, FL, USA, 2016. [Google Scholar]
- Selkoe, D.J. The therapeutics of Alzheimer’s disease: Where we stand and where we are heading. Ann. Neurol. 2013, 74, 328–336. [Google Scholar] [CrossRef] [PubMed]
- World Health Organization. Dementia Fact Sheet N°362; WHO: Geneva, Switzerland, 2016. [Google Scholar]
- Burns, A.; Iliffe, S. Alzheimer’s disease. BMJ 2009, 338, b158. [Google Scholar] [CrossRef] [PubMed]
- Brayne, C. The elephant in the room—Healthy brains in later life, epidemiology and public health. Nat. Rev. Neurosci. 2007, 8, 233–239. [Google Scholar] [CrossRef] [PubMed]
- Sperling, R.; Mormino, E.; Johnson, K. The evolution of preclinical Alzheimer’s disease: Implications for prevention trials. Neuron 2014, 84, 608–622. [Google Scholar] [CrossRef] [PubMed]
- Goedert, M. Oskar Fischer and the study of dementia. Brain 2009, 132, 1102–1111. [Google Scholar] [CrossRef] [PubMed]
- Holtzman, D.M.; Morris, J.C.; Goate, A.M. Alzheimer’s disease: The challenge of the second century. Sci. Transl. Med. 2011, 3, 77sr1. [Google Scholar] [CrossRef] [PubMed]
- Arendt, T.; Stieler, J.T.; Holzer, M. Tau and tauopathies. Brain Res. Bull. 2016, 126, 238–292. [Google Scholar] [CrossRef] [PubMed]
- Herrup, K. The case for rejecting the amyloid cascade hypothesis. Nat. Neurosci. 2015, 18, 794–799. [Google Scholar] [CrossRef] [PubMed]
- Musiek, E.S.; Holtzman, D.M. Three dimensions of the amyloid hypothesis: Time, space and “wingmen”. Nat. Neurosci. 2015, 18, 800–806. [Google Scholar] [CrossRef] [PubMed]
- Selkoe, D.J.; Hardy, J. The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Mol. Med. 2016, 8, 595–608. [Google Scholar] [CrossRef] [PubMed]
- Heppner, F.L.; Ransohoff, R.M.; Becher, B. Immune attack: The role of inflammation in Alzheimer disease. Nat. Rev. Neurosci. 2015, 16, 358–372. [Google Scholar] [CrossRef] [PubMed]
- Zlokovic, B.V. Neurovascular pathways to neurodegeneration in Alzheimer’s disease and other disorders. Nat. Rev. Neurosci. 2011, 12, 723–738. [Google Scholar] [CrossRef] [PubMed]
- Di Paolo, G.; Kim, T.-W. Linking lipids to Alzheimer’s disease: Cholesterol and beyond. Nat. Rev. Neurosci. 2011, 12, 284–296. [Google Scholar] [CrossRef] [PubMed]
- LaFerla, F.M. Calcium dyshomeostasis and intracellular signalling in Alzheimer’s disease. Nat. Rev. Neurosci. 2002, 3, 862–872. [Google Scholar] [CrossRef] [PubMed]
- Stanley, M.; Macauley, S.L.; Holtzman, D.M. Changes in insulin and insulin signaling in Alzheimer’s disease: Cause or consequence? J. Exp. Med. 2016, 213, 1375–1385. [Google Scholar] [CrossRef] [PubMed]
- Thathiah, A.; De Strooper, B. The role of G protein-coupled receptors in the pathology of Alzheimer’s disease. Nat. Rev. Neurosci. 2011, 12, 73–87. [Google Scholar] [CrossRef] [PubMed]
- Palop, J.J.; Mucke, L. Network abnormalities and interneuron dysfunction in Alzheimer disease. Nat. Rev. Neurosci. 2016, 17, 777–792. [Google Scholar] [CrossRef] [PubMed]
- Sullivan, P.F.; Daly, M.J.; O’Donovan, M. Genetic architectures of psychiatric disorders: The emerging picture and its implications. Nat. Rev. Genet. 2012, 13, 537–551. [Google Scholar] [CrossRef] [PubMed]
- Jaenisch, R.; Bird, A. Epigenetic regulation of gene expression: How the genome integrates intrinsic and environmental signals. Nat. Genet. 2003, 33 (Suppl.), 245–254. [Google Scholar] [CrossRef] [PubMed]
- Sweatt, J.D. The emerging field of neuroepigenetics. Neuron 2013, 80, 624–632. [Google Scholar] [CrossRef] [PubMed]
- Day, J.J.; Sweatt, J.D. Epigenetic mechanisms in cognition. Neuron 2011, 70, 813–829. [Google Scholar] [CrossRef] [PubMed]
- Hoffmann, A.; Spengler, D. The lasting legacy of social stress on the epigenome of the hypothalamic-pituitary-adrenal axis. Epigenomics 2012, 4, 431–444. [Google Scholar] [CrossRef] [PubMed]
- Gatz, M.; Reynolds, C.A.; Fratiglioni, L.; Johansson, B.; Mortimer, J.A.; Berg, S.; Fiske, A.; Pedersen, N.L. Role of genes and environments for explaining Alzheimer disease. Arch. Gen. Psychiatry 2006, 63, 168–174. [Google Scholar] [CrossRef] [PubMed]
- Wingo, T.S.; Lah, J.J.; Levey, A.I.; Cutler, D.J. Autosomal recessive causes likely in early-onset Alzheimer disease. Arch. Neurol. 2012, 69, 59–64. [Google Scholar] [CrossRef] [PubMed]
- Bertram, L.; Tanzi, R.E. Thirty years of Alzheimer’s disease genetics: The implications of systematic meta-analyses. Nat. Rev. Neurosci. 2008, 9, 768–778. [Google Scholar] [CrossRef] [PubMed]
- Seshadri, S.; Fitzpatrick, A.L.; Ikram, M.A.; DeStefano, A.L.; Gudnason, V.; Boada, M.; Bis, J.C.; Smith, A.V.; Carassquillo, M.M.; Lambert, J.C.; et al. Genome-wide analysis of genetic loci associated with Alzheimer disease. JAMA 2010, 303, 1832–1840. [Google Scholar] [CrossRef] [PubMed]
- Strittmatter, W.J.; Saunders, A.M.; Schmechel, D.; Pericak-Vance, M.; Enghild, J.; Salvesen, G.S.; Roses, A.D. Apolipoprotein E: High-avidity binding to beta-amyloid and increased frequency of type 4 allele in late-onset familial Alzheimer disease. Proc. Natl. Acad. Sci. USA 1993, 90, 1977–1981. [Google Scholar] [CrossRef] [PubMed]
- 1000 Genomes Project Consortium; Auton, A.; Brooks, L.D.; Durbin, R.M.; Garrison, E.P.; Kang, H.M.; Korbel, J.O.; Marchini, J.L.; McCarthy, S.; McVean, G.A. A global reference for human genetic variation. Nature 2015, 526, 68–74. [Google Scholar] [CrossRef] [PubMed]
- Harold, D.; Abraham, R.; Hollingworth, P.; Sims, R.; Gerrish, A.; Hamshere, M.L.; Pahwa, J.S.; Moskvina, V.; Dowzell, K.; Williams, A.; et al. Genome-wide association study identifies variants at CLU and PICALM associated with Alzheimer’s disease. Nat. Genet. 2009, 41, 1088–1093. [Google Scholar] [CrossRef] [PubMed]
- Hollingworth, P.; Harold, D.; Sims, R.; Gerrish, A.; Lambert, J.-C.; Carrasquillo, M.M.; Abraham, R.; Hamshere, M.L.; Pahwa, J.S.; Moskvina, V.; et al. Common variants at ABCA7, MS4A6A/MS4A4E, EPHA1, CD33 and CD2AP are associated with Alzheimer’s disease. Nat. Genet. 2011, 43, 429–435. [Google Scholar] [CrossRef] [PubMed]
- Lambert, J.-C.; Heath, S.; Even, G.; Campion, D.; Sleegers, K.; Hiltunen, M.; Combarros, O.; Zelenika, D.; Bullido, M.J.; Tavernier, B.; et al. Genome-wide association study identifies variants at CLU and CR1 associated with Alzheimer’s disease. Nat. Genet. 2009, 41, 1094–1099. [Google Scholar] [CrossRef] [PubMed]
- Naj, A.C.; Jun, G.; Beecham, G.W.; Wang, L.-S.; Vardarajan, B.N.; Buros, J.; Gallins, P.J.; Buxbaum, J.D.; Jarvik, G.P.; Crane, P.K.; et al. Common variants at MS4A4/MS4A6E, CD2AP, CD33 and EPHA1 are associated with late-onset Alzheimer’s disease. Nat. Genet. 2011, 43, 436–441. [Google Scholar] [CrossRef] [PubMed]
- Lambert, J.C.; Ibrahim-Verbaas, C.A.; Harold, D.; Naj, A.C.; Sims, R.; Bellenguez, C.; DeStafano, A.L.; Bis, J.C.; Beecham, G.W.; Grenier-Boley, B.; et al. Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer’s disease. Nat. Genet. 2013, 45, 1452–1458. [Google Scholar] [CrossRef] [PubMed]
- Roadmap Epigenomics Consortium; Kundaje, A.; Meuleman, W.; Ernst, J.; Bilenky, M.; Yen, A.; Heravi-Moussavi, A.; Kheradpour, P.; Zhang, Z.; Wang, J.; et al. Integrative analysis of 111 reference human epigenomes. Nature 2015, 518, 317–330. [Google Scholar] [CrossRef] [PubMed]
- Armstrong, L. Epigenetics; Garland Science: New York, NY, USA, 2014. [Google Scholar]
- Beck, S.; Olek, A.; Walter, J. From genomics to epigenomics: A loftier view of life. Nat. Biotechnol. 1999, 17, 1144. [Google Scholar] [CrossRef] [PubMed]
- Bird, A. Perceptions of epigenetics. Nature 2007, 447, 396–398. [Google Scholar] [CrossRef] [PubMed]
- Bernstein, B.E.; Meissner, A.; Lander, E.S. The mammalian epigenome. Cell 2007, 128, 669–681. [Google Scholar] [CrossRef] [PubMed]
- Rakyan, V.K.; Hildmann, T.; Novik, K.L.; Lewin, J.; Tost, J.; Cox, A.V.; Andrews, T.D.; Howe, K.L.; Otto, T.; Olek, A.; et al. DNA methylation profiling of the human major histocompatibility complex: A pilot study for the human epigenome project. PLoS Biol. 2004, 2, e405. [Google Scholar] [CrossRef] [PubMed]
- Pujadas, E.; Feinberg, A.P. Regulated noise in the epigenetic landscape of development and disease. Cell 2012, 148, 1123–1131. [Google Scholar] [CrossRef] [PubMed]
- Ziller, M.J.; Gu, H.; Müller, F.; Donaghey, J.; Tsai, L.T.-Y.; Kohlbacher, O.; De Jager, P.L.; Rosen, E.D.; Bennett, D.A.; Bernstein, B.E.; et al. Charting a dynamic DNA methylation landscape of the human genome. Nature 2013, 500, 477–481. [Google Scholar] [CrossRef] [PubMed]
- Lister, R.; Mukamel, E.A.; Nery, J.R.; Urich, M.; Puddifoot, C.A.; Johnson, N.D.; Lucero, J.; Huang, Y.; Dwork, A.J.; Schultz, M.D.; et al. Global epigenomic reconfiguration during mammalian brain development. Science (New York, N.Y.) 2013, 341, 1237905. [Google Scholar] [CrossRef] [PubMed]
- Gabel, H.W.; Kinde, B.; Stroud, H.; Gilbert, C.S.; Harmin, D.A.; Kastan, N.R.; Hemberg, M.; Ebert, D.H.; Greenberg, M.E. Disruption of DNA-methylation-dependent long gene repression in Rett syndrome. Nature 2015, 522, 89–93. [Google Scholar] [CrossRef] [PubMed]
- Guo, J.U.; Su, Y.; Shin, J.H.; Shin, J.; Li, H.; Xie, B.; Zhong, C.; Hu, S.; Le, T.; Fan, G.; et al. Distribution, recognition and regulation of non-CpG methylation in the adult mammalian brain. Nat. Neurosci. 2014, 17, 215–222. [Google Scholar] [CrossRef] [PubMed]
- Xie, W.; Barr, C.L.; Kim, A.; Yue, F.; Lee, A.Y.; Eubanks, J.; Dempster, E.L.; Ren, B. Base-resolution analyses of sequence and parent-of-origin dependent DNA methylation in the mouse genome. Cell 2012, 148, 816–831. [Google Scholar] [CrossRef] [PubMed]
- Feng, S.; Cokus, S.J.; Zhang, X.; Chen, P.-Y.; Bostick, M.; Goll, M.G.; Hetzel, J.; Jain, J.; Strauss, S.H.; Halpern, M.E.; et al. Conservation and divergence of methylation patterning in plants and animals. Proc. Natl. Acad. Sci. USA 2010, 107, 8689–8694. [Google Scholar] [CrossRef] [PubMed]
- Schübeler, D. Function and information content of DNA methylation. Nature 2015, 517, 321–326. [Google Scholar] [CrossRef] [PubMed]
- Suzuki, M.M.; Bird, A. DNA methylation landscapes: Provocative insights from epigenomics. Nat. Rev. Genet. 2008, 9, 465–476. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Weber, M.; Hellmann, I.; Stadler, M.B.; Ramos, L.; Pääbo, S.; Rebhan, M.; Schübeler, D. Distribution, silencing potential and evolutionary impact of promoter DNA methylation in the human genome. Nat. Genet. 2007, 39, 457–466. [Google Scholar] [CrossRef] [PubMed]
- Meissner, A.; Mikkelsen, T.S.; Gu, H.; Wernig, M.; Hanna, J.; Sivachenko, A.; Zhang, X.; Bernstein, B.E.; Nusbaum, C.; Jaffe, D.B.; et al. Genome-scale DNA methylation maps of pluripotent and differentiated cells. Nature 2008, 454, 766–770. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Schultz, M.D.; He, Y.; Whitaker, J.W.; Hariharan, M.; Mukamel, E.A.; Leung, D.; Rajagopal, N.; Nery, J.R.; Urich, M.A.; Chen, H.; et al. Human body epigenome maps reveal noncanonical DNA methylation variation. Nature 2015, 523, 212–216. [Google Scholar] [CrossRef] [PubMed]
- Kriaucionis, S.; Heintz, N. The nuclear DNA base 5-hydroxymethylcytosine is present in Purkinje neurons and the brain. Science 2009, 324, 929–930. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Varley, K.E.; Gertz, J.; Bowling, K.M.; Parker, S.L.; Reddy, T.E.; Pauli-Behn, F.; Cross, M.K.; Williams, B.A.; Stamatoyannopoulos, J.A.; Crawford, G.E.; et al. Dynamic DNA methylation across diverse human cell lines and tissues. Genome Res. 2013, 23, 555–567. [Google Scholar] [CrossRef] [PubMed]
- Mellén, M.; Ayata, P.; Dewell, S.; Kriaucionis, S.; Heintz, N. MeCP2 binds to 5hmC enriched within active genes and accessible chromatin in the nervous system. Cell 2012, 151, 1417–1430. [Google Scholar] [CrossRef] [PubMed]
- Szulwach, K.E.; Li, X.; Li, Y.; Song, C.-X.; Wu, H.; Dai, Q.; Irier, H.; Upadhyay, A.K.; Gearing, M.; Levey, A.I.; et al. 5-hmC-mediated epigenetic dynamics during postnatal neurodevelopment and aging. Nat. Neurosci. 2011, 14, 1607–1616. [Google Scholar] [CrossRef] [PubMed]
- Wu, H.; Zhang, Y. Reversing DNA methylation: Mechanisms, genomics, and biological functions. Cell 2014, 156, 45–68. [Google Scholar] [CrossRef] [PubMed]
- Feng, J.; Shao, N.; Szulwach, K.E.; Vialou, V.; Huynh, J.; Zhong, C.; Le, T.; Ferguson, D.; Cahill, M.E.; Li, Y.; et al. Role of Tet1 and 5-hydroxymethylcytosine in cocaine action. Nat. Neurosci. 2015, 18, 536–544. [Google Scholar] [CrossRef] [PubMed]
- Kaas, G.A.; Zhong, C.; Eason, D.E.; Ross, D.L.; Vachhani, R.V.; Ming, G.-L.; King, J.R.; Song, H.; Sweatt, J.D. TET1 controls CNS 5-methylcytosine hydroxylation, active DNA demethylation, gene transcription, and memory formation. Neuron 2013, 79, 1086–1093. [Google Scholar] [CrossRef] [PubMed]
- Li, X.; Wei, W.; Zhao, Q.-Y.; Widagdo, J.; Baker-Andresen, D.; Flavell, C.R.; D’Alessio, A.; Zhang, Y.; Bredy, T.W. Neocortical Tet3-mediated accumulation of 5-hydroxymethylcytosine promotes rapid behavioral adaptation. Proc. Natl. Acad. Sci. USA 2014, 111, 7120–7125. [Google Scholar] [CrossRef] [PubMed]
- Yu, H.; Su, Y.; Shin, J.; Zhong, C.; Guo, J.U.; Weng, Y.-L.; Gao, F.; Geschwind, D.H.; Coppola, G.; Ming, G.; Song, H. Tet3 regulates synaptic transmission and homeostatic plasticity via DNA oxidation and repair. Nat. Neurosci. 2015, 18, 836–843. [Google Scholar] [CrossRef] [PubMed]
- Hahn, M.A.; Qiu, R.; Wu, X.; Li, A.X.; Zhang, H.; Wang, J.; Jui, J.; Jin, S.-G.; Jiang, Y.; Pfeifer, G.P.; et al. Dynamics of 5-hydroxymethylcytosine and chromatin marks in Mammalian neurogenesis. Cell Rep. 2013, 3, 291–300. [Google Scholar] [CrossRef] [PubMed]
- Hoffmann, A.; Spengler, D. DNA memories of early social life. Neuroscience 2014, 264, 64–75. [Google Scholar] [CrossRef] [PubMed]
- Labonté, B.; Suderman, M.; Maussion, G.; Lopez, J.P.; Navarro-Sánchez, L.; Yerko, V.; Mechawar, N.; Szyf, M.; Meaney, M.J.; Turecki, G. Genome-wide methylation changes in the brains of suicide completers. Am. J. Psychiatry 2013, 170, 511–520. [Google Scholar] [CrossRef] [PubMed]
- Murgatroyd, C.; Wu, Y.; Bockmühl, Y.; Spengler, D. Genes learn from stress: How infantile trauma programs us for depression. Epigenetics 2010, 5, 194–199. [Google Scholar] [CrossRef] [PubMed]
- De Jager, P.L.; Srivastava, G.; Lunnon, K.; Burgess, J.; Schalkwyk, L.C.; Yu, L.; Eaton, M.L.; Keenan, B.T.; Ernst, J.; McCabe, C.; et al. Alzheimer’s disease: Early alterations in brain DNA methylation at ANK1, BIN1, RHBDF2 and other loci. Nat. Neurosci. 2014, 17, 1156–1163. [Google Scholar] [CrossRef] [PubMed]
- Lunnon, K.; Smith, R.; Hannon, E.; De Jager, P.L.; Srivastava, G.; Volta, M.; Troakes, C.; Al-Sarraj, S.; Burrage, J.; Macdonald, R.; et al. Methylomic profiling implicates cortical deregulation of ANK1 in Alzheimer’s disease. Nat. Neurosci. 2014, 17, 1164–1170. [Google Scholar] [CrossRef] [PubMed]
- Boiko, T.; Vakulenko, M.; Ewers, H.; Yap, C.C.; Norden, C.; Winckler, B. Ankyrin-dependent and -independent mechanisms orchestrate axonal compartmentalization of L1 family members neurofascin and L1/neuron-glia cell adhesion molecule. J. Neurosci. 2007, 27, 590–603. [Google Scholar] [CrossRef] [PubMed]
- Shin, J.-B.; Gillespie, P.G. Unraveling cadherin 23’s role in development and mechanotransduction. Proc. Natl. Acad. Sci. USA 2009, 106, 4959–4960. [Google Scholar] [CrossRef] [PubMed]
- Tanaka, M.; Murakami, K.; Ozaki, S.; Imura, Y.; Tong, X.-P.; Watanabe, T.; Sawaki, T.; Kawanami, T.; Kawabata, D.; Fujii, T.; et al. DIP2 disco-interacting protein 2 homolog A (Drosophila) is a candidate receptor for follistatin-related protein/follistatin-like 1—Analysis of their binding with TGF-β superfamily proteins. FEBS J. 2010, 277, 4278–4289. [Google Scholar] [CrossRef] [PubMed]
- Siggs, O.M.; Grieve, A.; Xu, H.; Bambrough, P.; Christova, Y.; Freeman, M. Genetic interaction implicates iRhom2 in the regulation of EGF receptor signalling in mice. Biol. Open 2014, 3, 1151–1157. [Google Scholar] [CrossRef] [PubMed]
- Almonte, A.G.; Sweatt, J.D. Serine proteases, serine protease inhibitors, and protease-activated receptors: roles in synaptic function and behavior. Brain Res. 2011, 1407, 107–122. [Google Scholar] [CrossRef] [PubMed]
- Watson, C.T.; Roussos, P.; Garg, P.; Ho, D.J.; Azam, N.; Katsel, P.L.; Haroutunian, V.; Sharp, A.J. Genome-wide DNA methylation profiling in the superior temporal gyrus reveals epigenetic signatures associated with Alzheimer’s disease. Genome Med. 2016, 8, 5. [Google Scholar] [CrossRef] [PubMed]
- Bernstein, B.E.; Mikkelsen, T.S.; Xie, X.; Kamal, M.; Huebert, D.J.; Cuff, J.; Fry, B.; Meissner, A.; Wernig, M.; Plath, K.; et al. A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell 2006, 125, 315–326. [Google Scholar] [CrossRef] [PubMed]
- Rakyan, V.K.; Down, T.A.; Maslau, S.; Andrew, T.; Yang, T.-P.; Beyan, H.; Whittaker, P.; McCann, O.T.; Finer, S.; Valdes, A.M.; et al. Human aging-associated DNA hypermethylation occurs preferentially at bivalent chromatin domains. Genome Res. 2010, 20, 434–439. [Google Scholar] [CrossRef] [PubMed]
- Watson, C.T.; Disanto, G.; Sandve, G.K.; Breden, F.; Giovannoni, G.; Ramagopalan, S.V. Age-associated hyper-methylated regions in the human brain overlap with bivalent chromatin domains. PLoS ONE 2012, 7, e43840. [Google Scholar] [CrossRef] [PubMed]
- Chibnik, L.B.; Yu, L.; Eaton, M.L.; Srivastava, G.; Schneider, J.A.; Kellis, M.; Bennett, D.A.; De Jager, P.L. Alzheimer’s loci: Epigenetic associations and interaction with genetic factors. Ann. Clin. Transl. Neurol. 2015, 2, 636–647. [Google Scholar] [CrossRef] [PubMed]
- Guintivano, J.; Aryee, M.J.; Kaminsky, Z.A. A cell epigenotype specific model for the correction of brain cellular heterogeneity bias and its application to age, brain region and major depression. Epigenetics 2013, 8, 290–302. [Google Scholar] [CrossRef] [PubMed]
- Houseman, E.A.; Accomando, W.P.; Koestler, D.C.; Christensen, B.C.; Marsit, C.J.; Nelson, H.H.; Wiencke, J.K.; Kelsey, K.T. DNA methylation arrays as surrogate measures of cell mixture distribution. BMC Bioinform. 2012, 13, 86. [Google Scholar] [CrossRef] [PubMed]
- Farlik, M.; Sheffield, N.C.; Nuzzo, A.; Datlinger, P.; Schönegger, A.; Klughammer, J.; Bock, C. Single-cell DNA methylome sequencing and bioinformatic inference of epigenomic cell-state dynamics. Cell Rep. 2015, 10, 1386–1397. [Google Scholar] [CrossRef] [PubMed]
- Macosko, E.Z.; Basu, A.; Satija, R.; Nemesh, J.; Shekhar, K.; Goldman, M.; Tirosh, I.; Bialas, A.R.; Kamitaki, N.; Martersteck, E.M.; et al. Highly Parallel Genome-wide Expression Profiling of Individual Cells Using Nanoliter Droplets. Cell 2015, 161, 1202–1214. [Google Scholar] [CrossRef] [PubMed]
- Gutierrez-Arcelus, M.; Ongen, H.; Lappalainen, T.; Montgomery, S.B.; Buil, A.; Yurovsky, A.; Bryois, J.; Padioleau, I.; Romano, L.; Planchon, A.; et al. Tissue-specific effects of genetic and epigenetic variation on gene regulation and splicing. PLoS Genet. 2015, 11, e1004958. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.I.; van de Geijn, B.; Raj, A.; Knowles, D.A.; Petti, A.A.; Golan, D.; Gilad, Y.; Pritchard, J.K. RNA splicing is a primary link between genetic variation and disease. Science 2016, 352, 600–604. [Google Scholar] [CrossRef] [PubMed]
- Bellini, E.; Pavesi, G.; Barbiero, I.; Bergo, A.; Chandola, C.; Nawaz, M.S.; Rusconi, L.; Stefanelli, G.; Strollo, M.; Valente, M.M.; et al. MeCP2 post-translational modifications: A mechanism to control its involvement in synaptic plasticity and homeostasis? Front. Cell. Neurosci. 2014, 8, 236. [Google Scholar] [CrossRef] [PubMed]
- Li, H.; Chang, Q. Regulation and function of stimulus-induced phosphorylation of MeCP. Front. Biol. 2014, 9, 367–375. [Google Scholar]
- Chen, L.; Chen, K.; Lavery, L.A.; Baker, S.A.; Shaw, C.A.; Li, W.; Zoghbi, H.Y. MeCP2 binds to non-CG methylated DNA as neurons mature, influencing transcription and the timing of onset for Rett syndrome. Proc. Natl. Acad. Sci. USA 2015, 112, 5509–5514. [Google Scholar] [CrossRef] [PubMed]
- Chahrour, M.; Zoghbi, H.Y. The story of Rett syndrome: From clinic to neurobiology. Neuron 2007, 56, 422–437. [Google Scholar] [CrossRef] [PubMed]
- Zimmermann, C.A.; Hoffmann, A.; Raabe, F.; Spengler, D. Role of MECP2 in experience-dependent epigenetic programming. Genes (Basel) 2015, 6, 60–86. [Google Scholar] [CrossRef] [PubMed]
- Zovkic, I.B.; Guzman-Karlsson, M.C.; Sweatt, J.D. Epigenetic regulation of memory formation and maintenance. Learn. Mem. 2013, 20, 61–74. [Google Scholar] [CrossRef] [PubMed]
- Baker-Andresen, D.; Ratnu, V.S.; Bredy, T.W. Dynamic DNA methylation: A prime candidate for genomic metaplasticity and behavioral adaptation. Trends Neurosci. 2013, 36, 3–13. [Google Scholar] [CrossRef] [PubMed]
- Zeltner, N.; Studer, L. Pluripotent stem cell-based disease modeling: Current hurdles and future promise. Curr. Opin. Cell Biol. 2015, 37, 102–110. [Google Scholar] [CrossRef] [PubMed]
- Brennand, K.; Savas, J.N.; Kim, Y.; Tran, N.; Simone, A.; Hashimoto-Torii, K.; Beaumont, K.G.; Kim, H.J.; Topol, A.; Ladran, I.; et al. Phenotypic differences in hiPSC NPCs derived from patients with schizophrenia. Mol. Psychiatry 2015, 20, 361–368. [Google Scholar] [CrossRef] [PubMed]
- Jo, J.; Xiao, Y.; Sun, A.X.; Cukuroglu, E.; Tran, H.-D.; Göke, J.; Tan, Z.Y.; Saw, T.Y.; Tan, C.-P.; Lokman, H.; et al. Midbrain-like Organoids from Human Pluripotent Stem Cells Contain Functional Dopaminergic and Neuromelanin-Producing Neurons. Cell Stem Cell 2016, 19, 248–257. [Google Scholar] [CrossRef] [PubMed]
- Qian, X.; Nguyen, H.N.; Song, M.M.; Hadiono, C.; Ogden, S.C.; Hammack, C.; Yao, B.; Hamersky, G.R.; Jacob, F.; Zhong, C.; et al. Brain-Region-Specific Organoids Using Mini-bioreactors for Modeling ZIKV Exposure. Cell 2016, 165, 1238–1254. [Google Scholar] [CrossRef] [PubMed]
- Camp, J.G.; Badsha, F.; Florio, M.; Kanton, S.; Gerber, T.; Wilsch-Bräuninger, M.; Lewitus, E.; Sykes, A.; Hevers, W.; Lancaster, M.; et al. Human cerebral organoids recapitulate gene expression programs of fetal neocortex development. Proc. Natl. Acad. Sci. USA 2015, 112, 15672–15677. [Google Scholar] [CrossRef] [PubMed]
- Srikanth, P.; Young-Pearse, T.L. Stem cells on the brain: Modeling neurodevelopmental and neurodegenerative diseases using human induced pluripotent stem cells. J. Neurogenet. 2014, 28, 5–29. [Google Scholar] [CrossRef] [PubMed]
- Kadoshima, T.; Sakaguchi, H.; Nakano, T.; Soen, M.; Ando, S.; Eiraku, M.; Sasai, Y. Self-organization of axial polarity, inside-out layer pattern, and species-specific progenitor dynamics in human ES cell-derived neocortex. Proc. Natl. Acad. Sci. USA 2013, 110, 20284–20289. [Google Scholar] [CrossRef] [PubMed]
- Lancaster, M.A.; Renner, M.; Martin, C.-A.; Wenzel, D.; Bicknell, L.S.; Hurles, M.E.; Homfray, T.; Penninger, J.M.; Jackson, A.P.; Knoblich, J.A. Cerebral organoids model human brain development and microcephaly. Nature 2013, 501, 373–379. [Google Scholar] [CrossRef] [PubMed]
- Byers, B.; Cord, B.; Nguyen, H.N.; Schüle, B.; Fenno, L.; Lee, P.C.; Deisseroth, K.; Langston, J.W.; Pera, R.R.; Palmer, T.D. SNCA triplication Parkinson’s patient’s iPSC-derived DA neurons accumulate α-synuclein and are susceptible to oxidative stress. PLoS ONE 2011, 6, e26159. [Google Scholar] [CrossRef] [PubMed]
- Cooper, O.; Seo, H.; Andrabi, S.; Guardia-Laguarta, C.; Graziotto, J.; Sundberg, M.; McLean, J.R.; Carrillo-Reid, L.; Xie, Z.; Osborn, T.; et al. Pharmacological rescue of mitochondrial deficits in iPSC-derived neural cells from patients with familial Parkinson’s disease. Sci. Transl. Med. 2012, 4, 141ra90. [Google Scholar] [CrossRef] [PubMed]
- Liu, G.-H.; Qu, J.; Suzuki, K.; Nivet, E.; Li, M.; Montserrat, N.; Yi, F.; Xu, X.; Ruiz, S.; Zhang, W.; et al. Progressive degeneration of human neural stem cells caused by pathogenic LRRK. Nature 2012, 491, 603–607. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, H.N.; Byers, B.; Cord, B.; Shcheglovitov, A.; Byrne, J.; Gujar, P.; Kee, K.; Schüle, B.; Dolmetsch, R.E.; Langston, W.; et al. LRRK2 mutant iPSC-derived DA neurons demonstrate increased susceptibility to oxidative stress. Cell Stem Cell 2011, 8, 267–280. [Google Scholar] [CrossRef] [PubMed]
- Reinhardt, P.; Schmid, B.; Burbulla, L.F.; Schöndorf, D.C.; Wagner, L.; Glatza, M.; Höing, S.; Hargus, G.; Heck, S.A.; Dhingra, A.; et al. Genetic correction of a LRRK2 mutation in human iPSCs links parkinsonian neurodegeneration to ERK-dependent changes in gene expression. Cell Stem Cell 2013, 12, 354–367. [Google Scholar] [CrossRef] [PubMed]
- Seibler, P.; Graziotto, J.; Jeong, H.; Simunovic, F.; Klein, C.; Krainc, D. Mitochondrial Parkin recruitment is impaired in neurons derived from mutant PINK1 induced pluripotent stem cells. J. Neurosci. 2011, 31, 5970–5976. [Google Scholar] [CrossRef] [PubMed]
- Miller, J.D.; Ganat, Y.M.; Kishinevsky, S.; Bowman, R.L.; Liu, B.; Tu, E.Y.; Mandal, P.K.; Vera, E.; Shim, J.; Kriks, S.; et al. Human iPSC-based modeling of late-onset disease via progerin-induced aging. Cell Stem Cell 2013, 13, 691–705. [Google Scholar] [CrossRef] [PubMed]
- Huh, C.J.; Zhang, B.; Victor, M.B.; Dahiya, S.; Batista, L.F.; Horvath, S.; Yoo, A.S. Maintenance of age in human neurons generated by microRNA-based neuronal conversion of fibroblasts. eLife 2016, 5, e18648. [Google Scholar] [CrossRef] [PubMed]
- Mertens, J.; Paquola, A.C. M.; Ku, M.; Hatch, E.; Böhnke, L.; Ladjevardi, S.; McGrath, S.; Campbell, B.; Lee, H.; Herdy, J.R.; et al. Directly Reprogrammed Human Neurons Retain Aging-Associated Transcriptomic Signatures and Reveal Age-Related Nucleocytoplasmic Defects. Cell Stem Cell 2015, 17, 705–718. [Google Scholar] [CrossRef] [PubMed]
- Horvath, S. DNA methylation age of human tissues and cell types. Genome Biol. 2013, 14, R115. [Google Scholar] [CrossRef] [PubMed]
- Choudhury, S.R.; Cui, Y.; Lubecka, K.; Stefanska, B.; Irudayaraj, J. CRISPR-dCas9 mediated TET1 targeting for selective DNA demethylation at BRCA1 promoter. Oncotarget. 2016, 7, 46545–46556. [Google Scholar] [CrossRef] [PubMed]
- Liu, X.S.; Wu, H.; Ji, X.; Stelzer, Y.; Wu, X.; Czauderna, S.; Shu, J.; Dadon, D.; Young, R.A.; Jaenisch, R. Editing DNA Methylation in the Mammalian Genome. Cell 2016, 167, 233–247. [Google Scholar] [CrossRef] [PubMed]
- Vojta, A.; Dobrinić, P.; Tadić, V.; Bočkor, L.; Korać, P.; Julg, B.; Klasić, M.; Zoldoš, V. Repurposing the CRISPR-Cas9 system for targeted DNA methylation. Nucleic Acids Res. 2016, 44, 5615–5628. [Google Scholar] [CrossRef] [PubMed]
- Xu, X.; Tao, Y.; Gao, X.; Zhang, L.; Li, X.; Zou, W.; Ruan, K.; Wang, F.; Xu, G.-L.; Hu, R. A CRISPR-based approach for targeted DNA demethylation. Cell Discov. 2016, 2, 16009. [Google Scholar] [CrossRef] [PubMed]
- Harel, I.; Benayoun, B.A.; Machado, B.; Singh, P.P.; Hu, C.-K.; Pech, M.F.; Valenzano, D.R.; Zhang, E.; Sharp, S.C.; Artandi, S.E.; et al. A platform for rapid exploration of aging and diseases in a naturally short-lived vertebrate. Cell 2015, 160, 1013–1026. [Google Scholar] [CrossRef] [PubMed]
© 2017 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 (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Hoffmann, A.; Sportelli, V.; Ziller, M.; Spengler, D. Driver or Passenger: Epigenomes in Alzheimer’s Disease. Epigenomes 2017, 1, 5. https://doi.org/10.3390/epigenomes1010005
Hoffmann A, Sportelli V, Ziller M, Spengler D. Driver or Passenger: Epigenomes in Alzheimer’s Disease. Epigenomes. 2017; 1(1):5. https://doi.org/10.3390/epigenomes1010005
Chicago/Turabian StyleHoffmann, Anke, Vincenza Sportelli, Michael Ziller, and Dietmar Spengler. 2017. "Driver or Passenger: Epigenomes in Alzheimer’s Disease" Epigenomes 1, no. 1: 5. https://doi.org/10.3390/epigenomes1010005
APA StyleHoffmann, A., Sportelli, V., Ziller, M., & Spengler, D. (2017). Driver or Passenger: Epigenomes in Alzheimer’s Disease. Epigenomes, 1(1), 5. https://doi.org/10.3390/epigenomes1010005