A. Abbott and E. Dolgin, Failed Alzheimer's trial does not kill leading theory of disease, Nature, vol.540, pp.15-16, 2016.

M. Achour, L. Gras, S. Keime, C. Parmentier, F. Lejeune et al., Neuronal identity genes regulated by super-enhancers are preferentially down-regulated in the striatum of Huntington's disease mice, Hum Mol Genet, vol.24, pp.3481-3496, 2015.

T. Ahmed, D. Blum, S. Burnouf, D. Demeyer, V. Buee-scherrer et al., Rescue of impaired late-phase long-term depression in a tau transgenic mouse model, Neurobiol Aging, vol.36, pp.730-739, 2015.

J. M. Alarcon, G. Malleret, K. Touzani, S. Vronskaya, S. Ishii et al., Chromatin acetylation, memory, and LTP are impaired in CBP+/mice: a model for the cognitive deficit in Rubinstein-Taybi syndrome and its amelioration, Neuron, vol.42, pp.947-959, 2004.

S. Anders and W. Huber, Differential expression analysis for sequence count data, Genome Biol, vol.11, p.106, 2010.

S. Anders, D. J. Mccarthy, Y. Chen, M. Okoniewski, G. K. Smyth et al., Count-based differential expression analysis of RNA sequencing data using R and Bioconductor, Nat Protoc, vol.8, pp.1765-1786, 2013.

G. Bai, I. Cheung, H. P. Shulha, J. E. Coelho, P. Li et al., Epigenetic dysregulation of hairy and enhancer of split 4 (HES4) is associated with striatal degeneration in postmortem Huntington brains, Hum Mol Genet, vol.24, pp.1441-1456, 2015.

R. M. Barrett, M. Malvaez, E. Kramar, D. P. Matheos, A. Arrizon et al., Hippocampal focal knockout of CBP affects specific histone modifications, long-term potentiation, and longterm memory, Neuropsychopharmacology, vol.36, pp.1545-1556, 2011.

E. Benito, H. Urbanke, B. Ramachandran, J. Barth, R. Halder et al., HDAC inhibitordependent transcriptome and memory reinstatement in cognitive decline models, J Clin Investig, vol.125, pp.3572-3584, 2015.

Y. Benjamini and Y. Hochberg, Controlling the false discovery rate: a practical and powerful approach to multiple testing, Journal of the Royal Statistical Society, vol.57, pp.289-300, 1995.

D. A. Bennett, L. Yu, J. Yang, G. P. Srivastava, C. Aubin et al., Epigenomics of Alzheimer's disease, Transl Res, vol.165, pp.200-220, 2015.

O. Bousiges, A. P. Vasconcelos, R. Neidl, B. Cosquer, K. Herbeaux et al., Spatial memory consolidation is associated with induction of several lysine-acetyltransferase (histone acetyltransferase) expression levels and H2B/H4 acetylation-dependent transcriptional events in the rat hippocampus, Neuropsychopharmacology, vol.35, pp.2521-2537, 2010.
URL : https://hal.archives-ouvertes.fr/inserm-00516665

O. Bousiges, R. Neidl, M. Majchrzak, M. A. Muller, A. Barbelivien et al., Detection of histone acetylation levels in the dorsal hippocampus reveals early tagging on specific residues of H2B and H4 histones in response to learning, PLoS One, vol.8, p.57816, 2013.
URL : https://hal.archives-ouvertes.fr/hal-02106640

M. A. Burlot, J. Braudeau, K. Michaelsen-preusse, B. Potier, S. Ayciriex et al., Cholesterol 24-hydroxylase defect is implicated in memory impairments associated with Alzheimer-like Tau pathology, Hum Mol Genet, vol.24, pp.5965-5976, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01779042

A. Caccamo, M. A. Maldonado, A. F. Bokov, S. Majumder, and S. Oddo, CBP gene transfer increases BDNF levels and ameliorates learning and memory deficits in a mouse model of Alzheimer's disease, Proc Natl Acad Sci, vol.107, pp.22687-22692, 2010.

R. G. Canter, J. Penney, and L. H. Tsai, The road to restoring neural circuits for the treatment of Alzheimer's disease, Nature, vol.539, pp.187-196, 2016.

P. Caroni, A. Chowdhury, and M. Lahr, Synapse rearrangements upon learning: from divergent-sparse connectivity to dedicated sub-circuits, Trends Neurosci, vol.37, pp.604-614, 2014.

S. Chatterjee, P. Mizar, R. Cassel, R. Neidl, B. R. Selvi et al., A novel activator of CBP/p300 acetyltransferases promotes neurogenesis and extends memory duration in adult mice, J Neurosci, vol.33, pp.10698-10712, 2013.
URL : https://hal.archives-ouvertes.fr/hal-02106635

F. Coppede, The potential of epigenetic therapies in neurodegenerative diseases, Front Genet, vol.5, p.220, 2014.

A. Fischer, Targeting histone-modifications in Alzheimer's disease. What is the evidence that this is a promising therapeutic avenue, Neuropharmacology, vol.80, pp.95-102, 2014.

L. Francelle, C. Lotz, T. Outeiro, E. Brouillet, and K. Merienne, Contribution of neuroepigenetics to Huntington's disease, Front Hum Neurosci, vol.11, p.17, 2017.
DOI : 10.3389/fnhum.2017.00017

URL : https://hal.archives-ouvertes.fr/cea-01751739

M. D. Gahete, A. Rubio, J. Córdoba-chacón, F. Gracia-navarro, R. D. Kineman et al., Expression of the ghrelin and neurotensin systems is altered in the temporal lobe of Alzheimer's disease patients, J Alzheimers Dis, vol.22, pp.819-828, 2010.

L. Gil, C. Federico, F. Pinedo, F. Bruno, A. B. Rebolledo et al., Aging dependent effect of nuclear tau, Brain Res, vol.1677, pp.129-137, 2017.

E. Gjoneska, A. R. Pfenning, H. Mathys, G. Quon, A. Kundaje et al., Conserved epigenomic signals in mice and humans reveal immune basis of Alzheimer's disease, Nature, vol.518, pp.365-369, 2015.

J. Graff, D. Rei, J. S. Guan, W. Y. Wang, J. Seo et al., An epigenetic blockade of cognitive functions in the neurodegenerating brain, Nature, vol.483, pp.222-226, 2012.

J. Graff and L. H. Tsai, The potential of HDAC inhibitors as cognitive enhancers, Annu Rev Pharmacol Toxicol, vol.53, pp.311-330, 2013.

D. Guiretti, A. Sempere, J. P. Lopez-atalaya, A. Ferrer-montiel, A. Barco et al., Specific promoter deacetylation of histone H3 is conserved across mouse models of Huntington's disease in the absence of bulk changes, Neurobiol Dis, vol.89, pp.190-201, 2016.

R. Halder, M. Hennion, R. O. Vidal, O. Shomroni, R. U. Rahman et al., DNA methylation changes in plasticity genes accompany the formation and maintenance of memory, Nat Neurosci, vol.19, pp.102-110, 2016.

G. E. Hardingham, S. Chawla, F. H. Cruzalegui, and H. Bading, Control of recruitment and transcription-activating function of CBP determines gene regulation by NMDA receptors and L-type calcium channels, Neuron, vol.22, pp.789-798, 1999.

K. M. Harris, F. E. Jensen, and B. Tsao, Three-dimensional structure of dendritic spines and synapses in rat hippocampus (CA1) at postnatal day 15 and adult ages: implications for the maturation of synaptic physiology and long-term potentiation, J Neurosci, vol.12, pp.2685-2705, 1992.

C. A. Hazzalin and L. C. Mahadevan, Dynamic acetylation of all lysine 4methylated histone H3 in the mouse nucleus: analysis at c-fos and c-jun, 2005.

, PLoS Biol, vol.3, p.393

S. Heinz, C. Benner, N. Spann, E. Bertolino, Y. C. Lin et al., Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities, Mol Cell, vol.38, pp.576-589, 2010.

W. Da-huang, B. T. Sherman, and R. A. Lempicki, Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources, Nat Protoc, vol.4, pp.44-57, 2009.

B. T. Hyman, C. H. Phelps, T. G. Beach, E. H. Bigio, N. J. Cairns et al., National Institute on Aging-Alzheimer's Association guidelines for the neuropathologic assessment of Alzheimer's disease, Alzheimers Dement, vol.8, pp.1-13, 2012.

S. Ito, A. Magalska, M. Alcaraz-iborra, J. P. Lopez-atalaya, V. Rovira et al., Loss of neuronal 3D chromatin organization causes transcriptional and behavioural deficits related to serotonergic dysfunction, Nat Commun, vol.5, p.4450, 2014.

Q. Jin, L. R. Yu, L. Wang, Z. Zhang, L. H. Kasper et al., Distinct roles of GCN5/PCAF-mediated H3K9ac and CBP/ p300-mediated H3K18/27ac in nuclear receptor transactivation, EMBO J, vol.30, pp.249-262, 2011.

J. Y. Joo, K. Schaukowitch, L. Farbiak, G. Kilaru, and T. K. Kim, Stimulus-specific combinatorial functionality of neuronal c-fos enhancers, Nat Neurosci, vol.19, pp.75-83, 2016.

R. Karli-c, H. R. Chung, J. Lasserre, K. Vlahovicek, and M. Vingron, Histone modification levels are predictive for gene expression, Proc Natl Acad Sci, vol.107, pp.2926-2931, 2010.

T. K. Kim, M. Hemberg, J. M. Gray, A. M. Costa, D. M. Bear et al., Widespread transcription at neuronal activity-regulated enhancers, Nature, vol.465, pp.182-187, 2010.
DOI : 10.1038/nature09033

URL : http://europepmc.org/articles/pmc3020079?pdf=render

D. Kim, G. Pertea, C. Trapnell, H. Pimentel, R. Kelley et al., TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions, Genome Biol, vol.14, p.36, 2013.

X. Kuang, H. J. Zhou, A. H. Thorne, X. N. Chen, L. J. Li et al., Neuroprotective effect of ligustilide through induction of a-secretase processing of both APP and Klotho in a mouse model of Alzheimer's disease, Front Aging Neurosci, vol.9, p.353, 2017.

V. Kumar, N. A. Rayan, M. Muratani, S. Lim, B. Elanggovan et al., Comprehensive benchmarking reveals H2BK20 acetylation as a distinctive signature of cell-state-specific enhancers and promoters, Genome Res, vol.26, pp.612-623, 2016.

H. Kurosu, M. Yamamoto, J. D. Clark, J. V. Pastor, A. Nandi et al., Suppression of aging in mice by the hormone Klotho, Science, vol.309, pp.1829-1833, 2005.

B. Langmead, C. Trapnell, M. Pop, and S. L. Salzberg, Ultrafast and memoryefficient alignment of short DNA sequences to the human genome, 2009.

, Genome Biol, vol.10, p.25

C. Laurent, G. Dorothée, S. Hunot, E. Martin, Y. Monnet et al., Hippocampal T cell infiltration promotes neuroinflammation and cognitive decline in a mouse model of tauopathy, Brain, vol.140, pp.184-200, 2017.
URL : https://hal.archives-ouvertes.fr/inserm-01833350

L. Gras, S. Keime, C. Anthony, A. Lotz, C. et al., , 2017.

, Huntington's disease striatal transcriptional signature, Sci Rep, vol.7, p.42875

J. Leon, A. J. Moreno, B. I. Garay, R. J. Chalkley, A. L. Burlingame et al., Peripheral elevation of a Klotho fragment enhances brain function and resilience in young, aging, and a-synuclein transgenic mice, Cell Rep, vol.20, pp.1360-1371, 2017.

J. P. Lopez-atalaya, S. Ito, L. M. Valor, E. Benito, and A. Barco, Genomic targets, and histone acetylation and gene expression profiling of neural HDAC inhibition, Nucleic Acids Res, vol.41, pp.8072-8084, 2013.

J. P. Lopez-atalaya and A. Barco, Can changes in histone acetylation contribute to memory formation?, Trends Genet, vol.30, pp.529-539, 2014.

M. I. Love, W. Huber, and S. Anders, Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2, Genome Biol, vol.15, p.550, 2014.

R. Madabhushi, F. Gao, A. R. Pfenning, L. Pan, S. Yamakawa et al., Activity-induced DNA breaks govern the expression of neuronal early-response genes, Cell, vol.161, pp.1592-1605, 2015.

A. N. Malik, T. Vierbuchen, M. Hemberg, A. A. Rubin, E. Ling et al., Genome-wide identification and characterization of functional neuronal activity-dependent enhancers, 2014.

A. Massó, A. Sánchez, A. Bosch, L. Giménez-llort, and M. Chillón, Secreted aKlotho isoform protects against age-dependent memory deficits, Mol Psychiatry, 2017.

C. Y. Mclean, D. Bristor, M. Hiller, S. L. Clarke, B. T. Schaar et al., GREAT improves functional interpretation of cisregulatory regions, Nat Biotechnol, vol.28, pp.495-501, 2010.

A. Medrano-fernández and A. Barco, Nuclear organization and 3D chromatin architecture in cognition and neuropsychiatric disorders, Mol Brain, vol.9, p.83, 2016.

A. C. Mitchell, R. Bharadwaj, C. Whittle, W. Krueger, K. Mirnics et al., The genome in three dimensions: a new frontier in human brain research, Biol Psychiat, vol.75, pp.961-969, 2014.

L. Peixoto and T. Abel, The role of histone acetylation in memory formation and cognitive impairments, Neuropsychopharmacology, vol.38, pp.62-76, 2013.

H. Qi, F. X. Cantrelle, H. Benhelli-mokrani, C. Smet-nocca, L. Buee et al., Nuclear magnetic resonance spectroscopy characterization of interaction of Tau with DNA and its regulation by phosphorylation, Biochemistry, vol.54, pp.1525-1533, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01145987

A. R. Quinlan and I. M. Hall, BEDTools: a flexible suite of utilities for comparing genomic features, Bioinformatics, vol.26, pp.841-842, 2010.

L. Restivo, G. Vetere, B. Bontempi, and M. Ammassari-teule, The formation of recent and remote memory is associated with time-dependent formation of dendritic spines in the hippocampus and anterior cingulate cortex, J Neurosci, vol.29, pp.8206-8214, 2009.
URL : https://hal.archives-ouvertes.fr/hal-01161790

C. Rouaux, N. Jokic, C. Mbebi, S. Boutillier, J. P. Loeffler et al., Critical loss of CBP/p300 histone acetylase activity by caspase-6 during neurodegeneration, EMBO J, vol.22, pp.6537-6549, 2003.

C. Rouaux, J. P. Loeffler, and A. L. Boutillier, Targeting CREB-binding protein (CBP) loss of function as a therapeutic strategy in neurological disorders, Biochem Pharmacol, vol.68, pp.1157-1164, 2004.

K. Schindowski, A. Bretteville, K. Leroy, S. Begard, J. P. Brion et al., Alzheimer's disease-like tau neuropathology leads to memory deficits and loss of functional synapses in a novel mutated tau transgenic mouse without any motor deficits, Am J Pathol, vol.169, pp.599-616, 2006.

A. Schneider, S. Chatterjee, O. Bousiges, B. R. Selvi, A. Swaminathan et al., Acetyltransferases (HATs) as targets for neurological therapeutics, Neurotherapeutics, vol.10, pp.568-588, 2013.
URL : https://hal.archives-ouvertes.fr/hal-02106628

A. Serrano-pozo, M. P. Frosch, E. Masliah, and B. T. Hyman, Neuropathological alterations in Alzheimer disease, Cold Spring Harb Perspect Med, vol.1, p.6189, 2011.

M. W. Shiflett, M. Gavin, and T. S. Tran, Altered hippocampal-dependent memory and motor function in neuropilin 2-deficient mice, Transl Psychiat, vol.5, p.521, 2015.

A. Sultan, F. Nesslany, M. Violet, S. Bégard, A. Loyens et al., Nuclear tau, a key player in neuronal DNA protection, J Biol Chem, vol.286, pp.4566-4575, 2011.

, Solanezumab: too late in mild Alzheimer's disease?, Lancet Neurol, vol.16, p.97, 2017.

F. Tie, R. Banerjee, A. R. Saiakhova, B. Howard, K. E. Monteith et al., Trithorax monomethylates histone H3K4 and interacts directly with CBP to promote H3K27 acetylation and antagonize Polycomb silencing, Development, vol.141, pp.1129-1139, 2014.

L. M. Valor, M. M. Pulopulos, M. Jimenez-minchan, R. Olivares, B. Lutz et al., Ablation of CBP in forebrain principal neurons causes modest memory and transcriptional defects and a dramatic reduction of histone acetylation but does not affect cell viability, J Neurosci, vol.31, pp.1652-1663, 2011.

L. M. Valor, J. Viosca, J. P. Lopez-atalaya, and A. Barco, Lysine acetyltransferases CBP and p300 as therapeutic targets in cognitive and neurodegenerative disorders, Curr Pharm Des, vol.19, pp.5051-5064, 2013.

A. Van-der-jeugd, T. Ahmed, S. Burnouf, K. Belarbi, M. Hamdame et al., Hippocampal tauopathy in tau transgenic mice coincides with impaired hippocampusdependent learning and memory, and attenuated late-phase long-term depression of synaptic transmission, Neurobiol Learn Mem, vol.95, pp.296-304, 2011.

M. Vashishtha, C. W. Ng, F. Yildirim, T. A. Gipson, I. H. Kratter et al., Targeting H3K4 trimethylation in Huntington disease, Proc Natl Acad Sci, vol.110, pp.3027-3036, 2013.

C. G. Vecsey, J. D. Hawk, K. M. Lattal, J. M. Stein, S. A. Fabian et al., Histone deacetylase inhibitors enhance memory and synaptic plasticity via CREB:CBPdependent transcriptional activation, J Neurosci, vol.27, pp.6128-6140, 2007.
DOI : 10.1523/jneurosci.0296-07.2007

URL : http://www.jneurosci.org/content/27/23/6128.full.pdf

L. A. Watson and L. H. Tsai, the loop: how chromatin topology links genome structure to function in mechanisms underlying learning and memory, 2017.

, Curr Opin Neurobiol, vol.43, pp.48-55

Y. Wei, M. H. Qu, X. S. Wang, L. Chen, D. L. Wang et al., Binding to the minor groove of the double-strand, tau protein prevents DNA from damage by peroxidation, PLoS One, vol.3, p.2600, 2008.

A. E. West and M. E. Greenberg, Neuronal activity-regulated gene transcription in synapse development and cognitive function, Cold Spring Harb Perspect Biol, vol.3, p.5744, 2011.

Z. Xiao, N. I. Cilz, L. Kurada, B. Hu, C. Yang et al., Activation of neurotensin receptor 1 facilitates neuronal excitability and spatial learning and memory in the entorhinal cortex: beneficial actions in an Alzheimer's disease model, J Neurosci, vol.34, pp.7027-7042, 2014.

T. Ye, A. R. Krebs, M. A. Choukrallah, C. Keime, F. Plewniak et al., ) seqMINER: an integrated ChIP-seq data interpretation platform, Nucleic Acids Res, vol.39, p.35, 2011.
DOI : 10.1093/nar/gkq1287

URL : https://academic.oup.com/nar/article-pdf/39/6/e35/16782420/gkq1287.pdf

C. Zang, D. E. Schones, C. Zeng, K. Cui, K. Zhao et al., A clustering approach for identification of enriched domains from histone modification ChIP-Seq data, Bioinformatics, vol.25, pp.1952-1958, 2009.

I. B. Zovkic, M. C. Guzman-karlsson, and J. D. Sweatt, Epigenetic regulation of memory formation and maintenance, Learn Mem, vol.20, pp.61-74, 2013.

, License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited