Front. Mol. Neurosci. Frontiers in Molecular Neuroscience Front. Mol. Neurosci. 1662-5099 Frontiers Media S.A. 10.3389/fnmol.2021.664912 Neuroscience Original Research Environmental Enrichment Induces Epigenomic and Genome Organization Changes Relevant for Cognition Espeso-Gil Sergio 1 2 3 * Holik Aliaksei Z. 1 Bonnin Sarah 1 Jhanwar Shalu 1 2 Chandrasekaran Sandhya 4 5 Pique-Regi Roger 6 Albaigès-Ràfols Júlia 1 2 Maher Michael 1 $ Permanyer Jon 1 Irimia Manuel 1 2 7 Friedländer Marc R. 8 Pons-Espinal Meritxell 1 2 Akbarian Schahram 5 Dierssen Mara 1 2 Maass Philipp G. 3 9 Hor Charlotte N. 1 2 * Ossowski Stephan 1 2 1Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain 2Universitat Pompeu Fabra (UPF), Barcelona, Spain 3Genetics and Genome Biology Program, SickKids Research Institute, Toronto, ON, Canada 4MD/PhD Program in the Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States 5Department of Psychiatry and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States 6Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, United States 7ICREA, Pg. Lluis Companys 23, Barcelona, Spain 8Science for Life Laboratory, Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden 9Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada

Edited by: Ildikó Rácz, University Hospital Bonn, Germany

Reviewed by: Eran A. Mukamel, University of California, San Diego, United States; Ricardo Marcos Pautassi, Medical Research Institute Mercedes and Martín Ferreyra (INIMEC), Argentina

*Correspondence: Sergio Espeso-Gil, sergio.espeso.gil@gmail.com Charlotte N. Hor, charlottenhor@gmail.com

Present address: Michael Maher, Joseph Carreras Leukaemia Research Institute (IJC), Campus Can Ruti, Barcelona, Spain; Meritxell Pons-Espinal, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain; Charlotte N. Hor, Centre for Integrative Genomics, University of Lausanne, Lausanne, Switzerland; Stephan Ossowski, Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany

These authors have contributed equally to this work

$ORCID: Michael Maher, orcid.org/0000-0002-4956-7185

This article was submitted to Molecular Signalling and Pathways, a section of the journal Frontiers in Molecular Neuroscience

05 05 2021 2021 14 664912 06 02 2021 09 04 2021 Copyright © 2021 Espeso-Gil, Holik, Bonnin, Jhanwar, Chandrasekaran, Pique-Regi, Albaigès-Ràfols, Maher, Permanyer, Irimia, Friedländer, Pons-Espinal, Akbarian, Dierssen, Maass, Hor and Ossowski. 2021 Espeso-Gil, Holik, Bonnin, Jhanwar, Chandrasekaran, Pique-Regi, Albaigès-Ràfols, Maher, Permanyer, Irimia, Friedländer, Pons-Espinal, Akbarian, Dierssen, Maass, Hor and Ossowski

This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

In early development, the environment triggers mnemonic epigenomic programs resulting in memory and learning experiences to confer cognitive phenotypes into adulthood. To uncover how environmental stimulation impacts the epigenome and genome organization, we used the paradigm of environmental enrichment (EE) in young mice constantly receiving novel stimulation. We profiled epigenome and chromatin architecture in whole cortex and sorted neurons by deep-sequencing techniques. Specifically, we studied chromatin accessibility, gene and protein regulation, and 3D genome conformation, combined with predicted enhancer and chromatin interactions. We identified increased chromatin accessibility, transcription factor binding including CTCF-mediated insulation, differential occupancy of H3K36me3 and H3K79me2, and changes in transcriptional programs required for neuronal development. EE stimuli led to local genome re-organization by inducing increased contacts between chromosomes 7 and 17 (inter-chromosomal). Our findings support the notion that EE-induced learning and memory processes are directly associated with the epigenome and genome organization.

environmental enrichment epigenetics 3D genome organization learning postnatal development chromatin accessibility Hi-C inter-chromosomal contacts

香京julia种子在线播放

    1. <form id=HxFbUHhlv><nobr id=HxFbUHhlv></nobr></form>
      <address id=HxFbUHhlv><nobr id=HxFbUHhlv><nobr id=HxFbUHhlv></nobr></nobr></address>

      Introduction

      Exposure to environmental stimuli influences developmental programs of organisms by modulating gene regulatory networks. These programs direct early postnatal neuronal development, particularly during the “critical period” that is key to establish brain functions that are kept throughout the lifetime of an individual (Hübener and Bonhoeffer, 2014). Environmental enrichment (EE) is a commonly used paradigm to study the behavioral and electrophysiological mechanisms of neuronal development (van Praag et al., 2000). Widely considered positively influencing cognition, it is a potential treatment application for a wide source of human traits (McDonald et al., 2018; Ball et al., 2019; Kempermann, 2019). On the counterpart, other studies found multiple sources of variability, confounds or even deleterious effects of EE (Berardo et al., 2016; Sparling et al., 2020). Generally, EE represents external factors, such as sensory, physical, cognitive, and social stimulation to provide and to maintain constant novelty and complexity, thereby laying a key foundation for future learning processes (Rountree-Harrison et al., 2018).

      The coalescing mechanisms of gene regulation, epigenetics, and genome organization leading to learning and memory formation still remain largely unknown. Thus far, studies on how EE affects gene regulatory elements are sparse, but some findings point toward the involvement of epigenetic mechanisms, both at the level of DNA methylation and histone modifications and chromatin modifiers (Irier et al., 2014; Morse et al., 2015). Recently, advances in brain research indicated that three-dimensional (3D) genome organization can be causally involved in gene-regulatory networks and chromatin conformation dynamics that impact brain function, learning, and memory formation (Fernandez-Albert et al., 2019; Rajarajan et al., 2019; Yamada et al., 2019; Beagan et al., 2020; Peter et al., 2020). These findings imply that a comprehensive molecular analysis of the processes happening during EE is needed to understand how neuronal circuits are refined by environmental cues. To accomplish this aim, we leveraged multiple genomic techniques to identify regulatory changes leading to learning and memory formation by EE during early postnatal neuronal development. We assessed chromatin accessibility, chromatin modifications, transcriptomic and proteomic changes, as well as 3D genome conformation. Our results reveal for the first time a comprehensive genome-wide perspective of global and neuronal-specific regulatory epigenetic modifications under EE in whole cerebral cortex, followed by neuron-specific and pyramidal-neuron-specific profiling. Our present study demonstrates that EE-induced early learning experience changes the neuronal epigenome and causes altered genomic conformations, especially between different chromosomes.

      Results Study Outline

      EE significantly influences learning and memory and leads to cognitive improvement, as demonstrated by multiple studies (Ohline and Abraham, 2019). Our EE-protocol was successfully established and validated by behavioral testing (Morris water maze) in an earlier study (Pons-Espinal et al., 2013). Briefly it consisted in a constantly changing environmental stimulation (every 48 h) over the course of 1 week (postnatal day P28) or 1 month (P51), an important stage of the critical period (Sztainberg and Chen, 2010; Hübener and Bonhoeffer, 2014) (Method Details, Figure 1A). For detailed insights into cell heterogeneity in the context of EE and the cerebral cortex microenvironment, we used whole cerebral cortex, sorted neuronal, and non-neuronal cells and performed epigenomic, transcriptomic, and proteomic profiling, as well as capturing of genomic interactions to provide a communal resource (Figures 1B,C).

      Experimental study design. (A) After weaning (P21), mice were exposed to environmental enrichment (EE) for 7 days (P28), and 30 days (P51, Methods). (B) Experimental workflow. Cortical tissue was homogenized from five different animals and split for the following protocols: ATACseq/SONOseq, ChIP-seq, RNAseq, label free and iTRAQ proteomics, and in situ Hi-C (2 biological replicates per condition, Nt = 20 animals in total). Neuronal and glial populations were sorted by the neuronal marker NeuN (Rbfox3) and pyramidal neurons by Thy+ (Tg[Thy1-YFP] mice). NeuN+ and NeuN (3 biological replicates per condition, Nt = 30 of animals; for Thy+ 2 individual biological replicates per condition, Nt = 4 animals; see Methods. (C) Datasets available per technique and per different cell population (dark gray).

      To analyze and intersect our multiple datasets, we devised a computational pipeline to determine activity and interplay of epigenomic marks in gene-regulatory regions, namely between enhancers predicted by the tool GEP (Jhanwar et al., 2018), and annotated promoters (Methods Details, Figure 2A and Supplementary Table 1).

      EE epigenetic changes during postnatal development. (A) Genomic features studied in the present study: left, enhancers predicted by GEP (Jhanwar et al., 2018) (Nt = 347112), middle: promoters 1,500 bp up- and 500 bp down-stream of TSS (Nt = 113,286); right: gene body regions (Nt = 46,833; see Methods). (B–D) Summary of differential changes (%) upon EE of chromatin accessibility and epigenetic marks over the total number of features in (B) enhancers, (C) promoters, (D) and gene-body regions (FDR < 0.05). (E) Top 100 enhancers, (F) promoters, and (G) Gene-body regions scaled in RPKM of the most important marks. Blue = increased; red = decreased signal upon EE, black = CTL samples. (H,I) Cell deconvolution of transcription-associated gene body marks: H3K36me3 and H3K79me2 in both whole cortex, neuronal, and non-neuronal datasets. Marker gene profile score (MPG) represents the first principal component regarding gene expression of cell-specific genes curated from single-cell studies involving GABAergic and pyramidal neurons, astrocytes, oligodendrocytes, microglia and endothelial cells (Mancarci et al., 2017) (Methods Details). (J) Overlap of differential H3K79me2 enrichment at P51 of whole cortex with NeuN+ and NeuN (at FDR < 0.05). (K) Time-course plot showing the progressive increase of differential binding sites (DBS) of H3K79me2 (P51 vs. P28) in CTL and EE samples (FDR < 0.05). (L) NeuN+ CTCF footprint plot. Y-axis corresponds to the Tn5 insertion rate over the background, x-axis distance in bp from the motif center (upper plot: bins over nucleotide position). Blue line designates increased CTCF binding in EE samples. Right plot: GO analysis (p-adj < 0.05 with Benjamini-Holchberg correction, Supplementary Table 2).

      EE Induces Increased Chromatin Accessibility and Insulation Targeting Synaptic-Associated Genes in Cortical Tissue

      EE is non-invasive in comparison to invasive neuronal stimulation which leads to increased chromatin accessibility in gene-regulatory regions to induce gene transcription (Su et al., 2017; Koberstein et al., 2018; Fernandez-Albert et al., 2019). Therefore, we asked if non-invasive EE could lead to quantifiable effects on gene and genome function during cortical cell postnatal development.

      First, we studied EE in whole cortical tissue (Pons-Espinal et al., 2013). In ATAC-seq experiments investigating chromatin accessibility (Buenrostro et al., 2013), we observed that distinct ATAC-seq peaks (macs2, fseq) were increased genome-wide in EE samples compared to controls (CTLs), suggesting a global increase in chromatin accessibility after EE (FCcortex1.17X, Supplementary Figure 1A). To validate these findings, a differential analysis of enhancers and promoters further confirmed increased chromatin accessibility in a very specific set of 0.13% of enhancers and 0.22% of the total promoters (FDR < 0.05, Figures 2B,C). To link intra-chromosomal interactions of enhancers to their corresponding promoters, we used a modified version of EpiTensor (Zhu et al., 2016; Supplementary Table 1, Method details). We found regulatory regions showing increased accessibility specific to genes that could be linked to neurogenesis and differentiation (Speisman et al., 2013; Clemenson et al., 2015), angiogenesis (Yu et al., 2014), synapse organization (Ohline and Abraham, 2019), and pathways associated to memory and learning such as Wnt (He et al., 2018), and Rho signaling (Martino et al., 2013; Supplementary Figures 1B,C and Supplementary Table 2). To further confirm previous ATAC-seq results, we used SONO-seq (Auerbach et al., 2009), a method based solely on sonicated and sequenced chromatin. We validated 76 genes showing consistent increased accessibility in their enhancers and promoters (p-adj < 0.01, Supplementary Figure 1E). Additionally, SONO-seq showed differential accessibility on pathways that are important in neuronal function such as MAPK and JNK (Coffey, 2014), neural maturation BMP (Bond et al., 2012), synaptic plasticity PI3K-Akt (Tan et al., 2017), cellular aging prevention and telomere protective role of oxytocin (Faraji et al., 2018; Stevenson et al., 2019), and neurotransmission function by GPCR (Betke et al., 2012; Supplementary Table 2).

      Accessible regions of chromatin regions encompass characteristic posttranslational modifications in surrounding histones (Fu et al., 2018). Due to the relationship between these histone marks and the increased chromatin accessibility in enhancers and promoters upon EE, we hypothesized that EE could also modulate posttranslational histone modifications and CTCF binding in gene-regulatory regions. We investigated a variety of histone marks from active (H3K27ac, H3K4me3, H3K4me1) to repressed regions (H3K27me3, H3K9me3), in addition to CTCF and DNA methylation. Interestingly, we detected relevant changes in H3K4me3, H3K27ac, DNA methylation, and CTCF upon EE (Supplementary Table 3). The regions represented about 0.2–0.4% of enhancers and 2–5% of the promoters depending on the mark being analyzed (FDR < 0.05, Figures 2B,C,E,F and Supplementary Table 3). With the exception of hypermethylated sites and weak changes in H3K27me3, the majority of changes occurred in activity-associated marks. We did not find significant changes in the heterochromatic mark H3K9me3, indicating that EE impacts the modulation of active gene sets rather than repressed regions. To investigate this in detail, we profiled transcription-associated marks such as H3K36me3 and H3K79me2 as potential readouts of gene expression (Huff et al., 2010), and determined a ∼20 and 10% differential binding of gene body marks, respectively, confirming that EE impacts transcriptional programs (FDR < 0.05, Figures 2D,G and Supplementary Table 3). Remarkably, the modulation of transcription-associated marks post EE induction was also observed in ∼13 and 8% of distal enhancers bearing H3K36me3 and H3K79me2, respectively, suggesting that enhancer-derived RNA genes are also differentially expressed (Kim et al., 2010) (FDR < 0.05, Supplementary Figures 1F,G). Genes associated to EE-induced cortical epigenetic marks changes were linked to the extracellular matrix (ECM) important for shaping synapses during postnatal development (Bikbaev et al., 2015; Ferrer-Ferrer and Dityatev, 2018), to circadian clock genes known to be required for proper healthy adult behavior (Brooks and Canal, 2013), and glutamatergic receptor complexes key for neuronal plasticity (Lüscher and Malenka, 2012; Supplementary Figures 1H–N and Supplementary Table 3).

      Having determined that EE induced differential chromatin accessibility and modulation of histone modifications in postnatal cortical tissue, we next addressed potential cross-talk mechanisms. We explored the overlap across all differential epigenetically modified and accessible chromatin regions identified previously (Supplementary Figure 1O). The strongest overlap corresponded to increased CTCF binding (18.7% of total sites) co-occurring with a decrease of the gene body activity marks H3K79me2 and H3K36me3 (at FDR < 0.1, Supplementary Figure 1P). A highly relevant example of this priming state is the early-life stress gene Met (Heun-Johnson and Levitt, 2018), and the memory regulating phosphodiesterase Pde8b (Tsai et al., 2012), both showing increased chromatin accessibility of interacting enhancers upon EE, as well as increased CTCF insulation in promoters, but decreased occupancy of H3K79me2 and H3K36me3 when compared to CTLs (Supplementary Figures 2A,B). This result could indicate a state where genes are poised to be transcribed, but are temporarily repressed by insulation, a specific state due to changes in chromatin architecture (Kim et al., 2015). But, it could also indicate mixed signals coming from the process of synaptic tuning, where some synapses gain strength meanwhile others are lost as consequence of the learning mechanism (Turrigiano, 2008).

      Molecular Phenotypic Changes by EE Mainly Target the Glutamatergic Synapse and Extracellular Matrix Elements

      Next, we determined how the previously described epigenetic changes alter transcriptional (coding and non-coding) and translational landscapes upon EE. Expression analysis revealed a total of 473 differentially expressed genes (FDR < 0.05, Supplementary Figure 2C and Supplementary Table 4). Additional biological replicates and gene ontology analysis recapitulated previously described pathways and terms, such as: BMP, JNK, MAPK, AMPAR signaling, and elements of the ECM (Supplementary Figures 2D–F). Moreover, to corroborate these results we performed a comparative analysis with previous published RNA-seq studies under the influence of EE (Zhang et al., 2016, 2018; Grégoire et al., 2018; Wassouf et al., 2018). We found 41 differential expressed genes in our data (∼10% of DEGs) that overlap with previous studies, showing consistent enrichment to ECM and elements from the postsynaptic density important for AMPA regulation such as Arc gene (Chowdhury et al., 2006; Shepherd et al., 2006; Supplementary Figure 3). By investigating the non-coding fraction of RNA, we identified 200 microRNAs and 52 long non-coding RNAs (lncRNAs) differentially expressed upon EE (Supplementary Figures 4A–F and Supplementary Table 5). Top microRNAs were validated in a new set of biological replicates (Supplementary Figure 4B). Using a multi-source microRNA target predictor (Friedman et al., 2010), we observed specificity for synaptic-associated mRNA targets in a gene ontology analysis (p-adj < 0.05, Supplementary Figures 4C,D and Supplementary Table 5). Of note, we found Meg3 and Rian (Meg8) as downregulated lncRNAs upon EE (p-adj < 0.05, Supplementary Figure 4E). Both are known for their ability to regulate glutamatergic neurotransmission, potentially in collaboration with microRNAs (Tan et al., 2017). We then explored the potential interactions between both non-coding elements (microRNAs and lnRNAs) with LncBase (Paraskevopoulou et al., 2016), and observed that 20 of our differentially expressed microRNAs could interact with Meg3 (Supplementary Figure 4F). Particularly interesting is the up-regulated miR125b-p5 reported to be involved in synaptic strength and Grin2a downregulation (Edbauer et al., 2010).

      To recapitulate EE-induced changes by quantitative protein expression, we used iTRAQ and LCMS mass spectrometry, finding about 73 and 145 differential proteins, respectively (p < 0.05, Supplementary Table 6). Gene ontology analysis of intersected EE-induced transcriptomic and proteomic changes identified pathways highlighting the importance of the ECM and neurotransmission receptor complexes, particularly involving the glutamatergic synapse (Supplementary Figure 4G).

      EE Stimulation in NeuN<sup>+</sup> Sorted Neurons

      To further understand the poised state of genes observed in cortical tissue and to avoid cell bias composition, we decided to investigate EE-induced influence in a cell-specific manner. We performed a cell deconvolution analysis to specify which cell types are primarily responding to EE stimulation (Mancarci et al., 2017). Remarkably, we observed that H3K36me3 and H3K79me2 were enriched in non-neuronal populations in whole cortex data (Figure 2H). However, to address the neuronal extent of EE-induced epigenetic changes observed in whole cortex, we performed FACSorting by nuclei immunostaining of the neuronal specific marker NeuN (Jiang et al., 2008) (Rbfox3, Supplementary Figure 5A). Another deconvolution of H3K79me2 in sorted populations demonstrated the neuronal-specific identification of EE-stimulatory effects (Figure 2I, Supplementary Figure 5B). We observed that differential analysis on H3K79me2 data between EE vs. CTLs showed two times more non-neuronal than neuronal H3K79me2 enrichment when compared to whole cortex, pointing the importance of non-neuronal for future studies (Figure 2J, FDR < 0.05). Neurons specifically, showed a total of 0.35% (P28) and 7.1% (P51) of genes with differential H3K79me2 gene-body occupation (FDR < 0.05, Figure 2D and Supplementary Table 3). Interestingly, we found that H3K79me2 occupancy changes from P28 to P51 in both CTL and EE samples (Figure 2K, FDR < 0.05, Supplementary Table 3). But the influence of EE amplifies this effect by affecting ∼2× more genes that significantly enrich for neurodevelopmental terms (Supplementary Figure 5C). Between these two time points of postnatal development, H3K79me2 changes in control samples are associated to axon sprouting, mitochondrial activity, complement cascade and cerebral cortical regionalization with associated genes such as Pcdh18, identified as a potential candidate for intellectual disability (Kasnauskiene et al., 2012; Supplementary Figure 5D). In other hand, EE samples significantly enrich for learning processes, including both glutamatergic and GABAergic genes such as Gabra2, key modulator in multiple brain traits (Mulligan et al., 2019; Supplementary Figure 5E).

      We revisited our previous findings in sorted neurons by addressing chromatin accessibility and gene-body epigenetic profiling (Figures 2C,D). Similar to whole cortex, we observed increased chromatin accessibility sites in enhancers and promoters after EE in NeuN+ cells (FCneurons = 1.61X, Supplementary Figure 6A). But differential sites represented around 0.01 and 0.006%, respectively, a lower rate when compared to whole cortex (FDR < 0.05, Figures 2B,C and Supplementary Table 2). As NeuN+ marker is specific for a broad number of different neurons (Figure 2I), we extended our study to sorted pyramidal neurons overexpressing a yellow fluorescent protein (YFP) under the control of the Thy1 gene promoter (Method Details, Supplementary Figure 6B). We found strongly increased chromatin accessibility upon EE (0.36% of enhancers, 0.39% of promoters), similar to the proportions in whole cortex (at FDR < 0.05, Figures 2B,C, Supplementary Figures 6C,D, and Supplementary Table 3).

      Our findings confirm that EE leads to increased chromatin accessibility in whole cortex, in NeuN+ neurons, and more specifically in pyramidal neurons. We further confirm that these differential accessible enhancers are active forebrain enhancers at P0 and active in pyramidal neurons both in mouse and human (Supplementary Figure 6E). Particular genes could be linked to human cognition in the context of schizophrenia and autism, such as Nrg3 and Ank2, respectively (Kao et al., 2010; Yang et al., 2019; Supplementary Figure 6F).

      Because higher chromatin accessibility may allow increased TF binding, we ran a transcription factor binding site (TFBS) footprint analysis on whole cortex and NeuN+ populations using Centipede which screens for all putative TFBS (Pique-Regi et al., 2011). We confirmed that more TFs were significantly bound in EE compared to CTLs, such as Lhx3, AP1, Nr5a2, and Phox2B (Supplementary Figure 6G and Supplementary Table 2). Interestingly, we observed that CTCF displayed one of the strongest instances bound in EE-stimulated neurons, similar to CTCF ChIPseq results (p-adj < 0.05, Figure 2C). This finding supports the idea that EE leads to increased genomic insulation and plays a role in genome organization during postnatal development.

      Overall, we found that EE in neurons recapitulates cortical results inducing increased chromatin accessibility and CTCF binding. However in neurons, H3K79me2 increased upon EE which was not observed in the poised state of whole cortex. Noteworthy, GO terms of neuronal EE-induced changes show again genes associated with learning and memory targeting glutamatergic transmission predominantly, but also GABAergic and cholinergic transmission (Figure 2L, Supplementary Figures 6I,J, and Supplementary Table 2).

      EE Stimulation Prompts 3D Genome Changes

      The described EE-related changes implicate that the epigenome plays an important role in 3D genome organization (Rao et al., 2014; Guo et al., 2015). The evidence of increased chromatin accessibility and CTCF binding may suggest that environmental stimuli impact higher-order genome organization. To further assess chromatin interactions in sorted neurons (NeuN+), we performed Hi-C experiments to explore the 3D genome organization upon EE. By comparing intra-chromosomal interactions at 100 kilobase (kb) resolution, we determined significant changes: 94 interactions increased and 544 decreased upon EE stimulation (FDR < 0.05, Figure 3A and Supplementary Table 7). A decrease of intra-chromosomal interactions was also observed when calculating the number of chromatin loops using HICCUPs between CTL and EE (FDR < 0.05, Figure 3B; Durand et al., 2016b). We found differential intra-chromosomal bins to be clustered in particular chromosomal regions, specially involving chromosomes 8 and 14 (Figure 3C). In these regions, we find genes such as the synaptic-linked gene Ngr1 linked to cognitive function improvement (Chen et al., 2008; Xu et al., 2016); and the synaptic vesicle exocytosis regulator Cadps (Sadakata et al., 2007).

      3D genome interaction changes upon EE. (A) Differential analysis of intra and inter-chromosomal interactions at 100 kb and 1 Mb, respectively (FDR < 0.05). (B) Significant chromatin loops computed with HICCUPs at 5 and 10 kb resolution (FDR < 0.05). (C) Manhattan plot of differential intra-chromosomal interactions at 100 kb. (D) Juicebox heatmaps at 250 kb showing the extraction of EE vs. CTL of inter-chromosomal interactions. (E) Circos-plot of differential inter-chromosomal interactions (blue arcs-increased interactions, pink-decreased) together with concentric bedfiles representing the differential analysis of ATACseq, H3K79me2, H3K36me3 and RNAseq at 1 MB using Diffreps (increased regions upon EE = blue, decreased = red). (F) GO analysis of genes in the differential inter-chromosomal interactions at 1 MB upon EE stimulation (p-adj < 0.05 Bonferroni-step down). (G,H) In silico chrom3D models for EE and CTL samples showing significant increase of inter-chromosomal interactions. (I) A/B compartmentalization measured by eigenvector scores in chromosomes 7, 8, and 17. *** denotes p≤0.001.

      While these intra-chromosomal contacts were in the focus of chromatin biology in recent years, contacts between different chromosomes (inter-chromosomal) also occur and are involved in important biological functions (Quinodoz et al., 2018; Maass et al., 2019; Monahan et al., 2019), but they remain less studied. Therefore, we asked if EE is associated with large scale changes in genome organization by tracing chimeric inter-chromosomal Hi-C reads. Indeed, we identified 241 increased and 40 decreased interactions at 1 megabase (Mb) resolution (FDR < 0.05, Figure 3A and Supplementary Table 7). We determined a significant accumulation of inter-chromosomal contacts between chromosome 7 and 17 in EE vs. CTL (Figures 3D–F, arrow). We observed that these differential interactions form a clear genome architectural stripe, also termed Greek islands (Monahan et al., 2019; Figure 3D). Among bins involving these two chromosomes, we found relevant genes associated with the synaptic vesicle cycle, such as Ap2a[1-2] being important for AMPAR endocytosis (DaSilva et al., 2016), Acat[2-3] acetyl-CoA C-acetyltransferase playing a key role in neuronal metabolism (Ronowska et al., 2018), and Cacng8 modulating AMPAR receptor complexes in the plasmatic membrane which are important for synaptic plasticity (Maher et al., 2016; Figure 3F and Supplementary Table 7). We validated our differential analysis by an independent method, called Chrom3D, and generated in silico models for pooled EE and CTL samples detecting significant proximity of chromosomes 7 and 17 (Paulsen et al., 2017; Figures 3G,H). We further corroborated the previous association of inter-chromosomal changes with gene-activity by studying chromatin compartmentalization (Lieberman-Aiden et al., 2009). As expected, A/B compartments do not change between CTL and EE, except for local changes in the strongest hubs of both intra- and inter-chromosomal contacts (chromosomes 7, 8, and 17), pointing to EE-related local chromatin changes in specific regions associated with active epigenetic modifications and gene expression changes (Figure 3I).

      EE Causes Coordinated Regulatory Changes That Cluster in Inter-Chromosomal Interactions Implicated in Memory-Related Functions

      The multiple “omics” datasets to study the molecular basis of EE allowed us to conduct an intersection of all EE-induced changes determined in this study (Figure 4A). Interestingly, GO analysis revealed synapse organization as the strongest ranked term (p-adj < 0.05, Supplementary Figure 7A). We decided to explore this finding further using SynGO synaptic gene curator tool to identify overrepresented genes (hits > 4 showing intersection in different sets, Figure 4B; Koopmans et al., 2019). We found a significant enrichment of postsynaptic and presynaptic genes, particularly targeting the glutamatergic synapse (Figures 4C,D).

      Data integration and EE implications in brain cognition. (A) Full intersection of differential changes induced by EE (FDR < 0.05). Pink arcs—differential expressed genes intersected with the rest of the data, blue proteomic, and green inter-chromosomal changes. (B) Intersection hits plot, representing the number of times each gene is represented in the current study. Dashed lines—genes > 4 times intersected. (C) SynGO analysis showing the enrichment of the most intersected genes which represent postsynaptic and presynaptic genes (right bar-plot, p-adj < 0.05). (D) String-db analysis interactome at 0.99 confidence of the most intersected genes. (E) Transcriptomic and proteomic changes represented in other differential sets at FDR < 0.05. “Pink + green” and “blue + green”—total percentages of transcriptomic and proteomic changes found in other differential datasets, where green specifically represents the portion of these changes found in inter-chromosomal changes. (F) Differential inter-chromosomal changes association with the rest of the marks (Npermutations = 100 k, **p < 0.01, *p < 0.05). (G) Pearson correlation of EE-induced epigenetic marks changes in enhancers and promoters with differentially expressed genes (DEG). (H) Differential inter-chromosomal changes association with human brain GWAS traits. Differential bins were lifted to the human genome for the permutation analysis (Npermutations = 100 k, **p < 0.01, *p < 0.05). To illustrate the likelihood of the results, an example of the random shuffling is provided bellow to show the strength of the analysis.

      Further analysis of our merged data showed that about 60% of transcriptomic and 84% of proteomic changes are found in our other datasets, whilst 20% of changes were determined by EE-induced inter-chromosomal changes (Figure 4E). This enrichment together with the prior observation that differentially expressed genes tend to cluster in specific inter-chromosomal bins (Figure 3E), led us to the hypothesis that EE mainly induces changes locally in the genome, especially where active transcription occurs. To test this, we permuted the background genome at 1 Mb resolution 100 k times and calculated the likelihood of differential inter-chromosomal interactions to be associated with the differential epigenomic, transcriptomic, and proteomic changes found in the rest of the study. Strikingly, we observed that microRNA target genes, proteins (iTRAQ MS data), and gene-body associated histone marks were the strongest associated features within the specific inter-chromosomal hubs (Figure 4F). This finding confirms that EE orchestrates local changes of the nuclear architecture, especially inter-chromosomal communication.

      Our intersection and permutation analysis indicated that chromatin conformation might connect the epigenome with the molecular phenotypes. We now asked how different marks influence others by estimating the linear dependency of EE-induced enhancers and promoters with transcriptomic and proteomic changes by Pearson and Spearman correlations (Figure 4G and Supplementary Figure 7B, Method details). We observed that enhancer RNA and promoter H3K79me2 activity drive transcriptomic and proteomic changes due EE. Particularly, we found H3K79me2 mark at P28 in promoters the most correlated modification indicating that transcripts and proteins observed at P51 are dependent on earlier stages of postnatal neuronal development, thereby underlining the temporal aspect of the critical period.

      The cognitive and behavioral effects of EE could be a beneficial strategy for cognitive human disorders (Ball et al., 2019). Thus far, it is unclear if the molecular effects of EE that we found in mice can be retrieved in human. Therefore, we addressed the local changes in 3D genome organization in the human genome. Due our findings of clustered induced EE changes in differential inter-chromosomal bins, we performed a lift-over of these regions at 1 Mb from mouse to the human genome and ran a permutation analysis to test the association with 33 genome wide association studies (GWAS) relevant for human brain traits (Supplementary Table 8). We observed that the top associated traits were memory-related, such as memory performance (p < 0.01, Figure 4H). These findings not only validate our previous results, but point to conserved mechanisms between mouse and human that drive EE-related molecular effects by epigenomic, transcriptomic, and proteomic changes locally in specific regions of the genome that are important for both human and mouse cognition.

      Discussion

      We have characterized the regulatory response to EE by using omics both in whole cortex tissue and in two neuronal cell populations and provide a valuable resource for other scientists. We demonstrate that EE induces coordinated changes of gene-regulatory networks that involve epigenetics and genome organization to adapt to constant cognitive stimulation and social interaction. EE induced increased enhancer and promoter chromatin accessibility in neurons, corroborating previous studies showing increased open chromatin upon invasive neuronal stimulation (Su et al., 2017; Koberstein et al., 2018; Fernandez-Albert et al., 2019). These studies also highlighted the importance of CTCF shaping the 3D genome during postnatal development for memory formation (Sams et al., 2016; Kim et al., 2018). Here, we demonstrated that CTCF tends to bind preferentially to synaptic-associated genes upon EE, particularly glutamatergic associated pathways.

      Our results also revealed, that gene body marks show differential activity in distal active enhancers upon EE, pointing to a potential role for these marks at transcriptionally active enhancers (Zentner et al., 2011). This is conform with the finding some DNA methyltransferases depend on H3K36me3 to exert their function at enhancers (Rinaldi et al., 2016). We also observed that active transcription in regulatory regions during early stages of postnatal neuronal development may influence local transcriptomic and proteomic changes at later stages. Furthermore, studying gene body marks in sorted neuronal populations allowed us to identify the molecular effects during the postnatal critical period, reflected by a constant increase in H3K79me2 occupation. We found it exacerbated in an experience-dependent manner, with a greater number of increased binding sites in EE compared to CTL samples across time.

      Despite the caveat of cell heterogeneity potentially skewing observations in whole tissue-related experiments, particularly involving epigenetic gene body marks, it has been shown that these marks can be anticorrelated with expressed genes during aging (Pu et al., 2015). Another conflicting example that could be influenced by cell composition was the observed results involving increased chromatin accessibility and decreased H3K27ac in whole cortex enhancers. Both marks are highly related with gene transcription and could share some degree of linear dependency due the influence of EE (Karlić et al., 2010; Klemm et al., 2019). However, a recent study might indicate the contrary, as changes in H3K27ac coverage do not influence ATAC-seq levels in the same genomic regions (Zhang et al., 2020). Moreover, we didn’t observe overlap between the differential regions of both marks, supporting the hypothesis that changes due EE in both marks, could be totally independent. Even though, we cannot discard cell composition as a factor influencing those results. Future investigation using single cell/nuclei approaches is needed to better determine these epigenetic relationships due external factors such as EE. In fact, we have shown by cell-deconvolution the importance of other cell types which are often ignored in learning-memory studies. Additionally, EE-induced directional regulation particularly of epigenetic marks could reflect the discrepancy occurring upon cognitive stimulation, such as pruning and synaptic tuning, where both synaptic strength and loss are part of the learning process during postnatal development (Turrigiano, 2008; Stephan et al., 2012).

      Furthermore, by applying Hi-C to neurons, we elucidated for the first time intra- and inter-chromosomal interactions sensitive to EE. Especially the mnemonic inter-chromosomal 3D conformation map with its major inter-chromosomal hub involving chromosomes 7 and 17 shows that the environmental stimulus EE affects local epigenomic regulation and chromatin interactions in a coordinated manner. EE-induced changes relate to both synapse strengthening and pruning genes, affecting cytoskeletal rearrangements and ECM associated genes (Wright and Harding, 2009; Smagin et al., 2018). These synaptic rearrangements need pathways, such as Rho, GPCR, and PKC/Akt signaling which we found enriched in our study, with special enrichment of Wnt signaling (Hu et al., 2013; Lichti et al., 2014; Tan et al., 2017).

      Our results indicate that environmental cues in postnatal development, particularly stimulation provided by EE, modulates epigenomic and 3D genome landscapes in a coordinated manner relevant for cognition.

      Materials and Methods Experimental Procedures Animals: Housing and Enrichment Conditions

      All experimental procedures were approved by the local ethical committee (Procedure Code: ISA-11-1358). Wild type mice (C57BL/6J) and Tg(Thy1-YFP) [strain B6.Cg-Tg(Thy1-YFPH)2Jrs/J No. 003782; The Jackson Laboratories] were kept and bred according to local (Catalan law 5/1995 and Decrees 214/97, 32/2007) and European regulations (EU directives 86/609 and 2001-486).

      After weaning (21 days of age), mice were randomly reared under either non-enriched (NE) or enriched (EE) conditions for 30 days. In NE conditions, animals were reared in conventional Plexiglas cages (20 × 12 × 12 cm height) in groups of two to three animals. The EE group was reared in spacious (55 × 80 × 50 cm height) Plexiglas cages with toys, small houses, tunnels, and platforms. The arrangement was changed every 3 days to maintain the novelty of the environment. To stimulate social interactions, six to eight mice were housed in each EE cage. All groups of animals were maintained under the same 12 h (8:00 to 20:00) light-dark cycle in controlled environmental conditions of humidity (60%) and temperature (22°C), with free access to food and water. The experiments were conducted using only females, since male mice showed hierarchical behavior similar to that observed previously that may affect the outcome of EE (Martínez-Cué et al., 2002). To stimulate social interactions, six to eight mice were housed in each EE cage. The use of female mice allowed to mix different mice in the same cage from different litters.

      Cortical data analysis derived from the same samples and EE protocol that was used by Pons-Espinal et al. (2013) and PhD thesis “Role of DYRK1A in hippocampal neuroplasticity: Implications for Down syndrome1,” where behavioral studies (Morris water maze testing) showed successful EE treatment. At 5 (P28) or 8 weeks of age (P51), mice were euthanized by carbon dioxide, and the cerebral cortex was dissected within 1 min of death. The tissue was immediately flash frozen in liquid nitrogen. In the case of Tg(Thy1-YFP) animals, the tissue was immersed in Hank’s Balanced Salt Solution (HBSS 1X, Gibco 14065-049) before proceeding with the sample preparation for the FAC-sorting (see section below). For each condition and replicate, we pooled the cortices of five mice with some exceptions: mice for insitu-HiC where single replicas as well as Thy-YFP mice for ATACseq (Ncortex = 20 mice in 4 bioreplicates; Nsorted = 30 mice in 6 bioreplicates, NNeuN+_HiC = 4 mice in 4 bioreplicates, NThy+ = 4 mice in 4 bioreplicates). The frozen cortices for pooled animals were ground together in a frozen mortar containing liquid nitrogen, to obtain a fine powder of pooled cortex tissue. The powder was aliquoted and stored at −80°C.

      Nucleic Acid Extraction

      DNA was extracted using Phenol:chroloform:iso-amyl alcohol (25:24:1) according to manufacturer guidelines (Sigma 77617-500ml). RNA was extracted using Qiazol total RNA (Qiagen Cat No:79306) kit according to the manufacturer’s instructions. The RNA was quantified by Qubit® 2.0 Fluorometer (Life Technologies) and the quality was assessed using a Nanodrop 2000c (Thermo Fisher Scientific) and a 2100 Bioanalyzer (Agilent Technologies, CA, United States).

      Nucleus Isolation

      To obtain fresh nuclei, ground frozen tissue was resuspended in tissue lysis buffer (1x PBS containing 0.1% Triton X-100, 1x Complete Protease inhibitor cocktail tablette (cOMPLETE mini EDTA-free, Roche Cat No. 11836170001) and 1 mg/ml AEBSF (Pefabloc, Roche Cat No. 11585916001) and dissociated by 60–90 strokes in a glass douncer (7 ml tissue grinder Tenbroek, Wheaton Cat No. 357424). Nuclei were counted using a hemacytometer and constantly checked under a microscope (Leica DM-IL).

      FAC-Sorting

      Two different procedure were performed: (1) Sorting total neurons using NeuN (Rbfox3) marker and (2) Sorting pyramidal neurons in Tg(Thy1-YFP) mice. Briefly, after the nucleus preparation, nuclei were resuspended in 1 ml of PBS-PI 1X [1X-PBS, 1x Complete Protease inhibitor cocktail (cOMPLETE mini EDTA-free, Roche Cat No. 11836170001), 1 mg/ml AEBSF Pefabloc (Pefabloc, Roche Cat No. 11585916001) and 0.1 mg/ml of BSA (BSA, Molecular Biology Grade NEB, Cat No. B9000S)]. 1.5 μl of anti-NeuN, clone A60, Alexa Fluor® 555 Conjugate (Merk Millipore Cat No. MAB377A5) was added to the solution and incubated at 4°C for 1.5 h protected from light. The sample was centrifuged for 10 min, 500 g at 4°C and washed with 1 ml of PBS-PI 1X. Next, 1μl of 4′,6-Diamidine-2′-phenylindole dihydrochloride was added (DAPI, Roche Cat. No. 10236276001) and the sample was given immediately to the FACS-sorting Facility. Samples were sorted at 12PSI in cold condition in an INFLUX sorter (BD Biosciences INFLUXTM). The sorted samples were centrifuged for 40 min at 700 g at 4°C to collect the nuclei before proceeding with the desired technique. For sorting pyramidal neurons Tg(Thy1-YFP), animals were dissected and tissue was immediately submerged in Hank’s Balanced Salt Solution (HBSS 1X, Gibco 14065-049). Brain samples were dissociated using the Neural Dissociation Kit (P) (MACS Milteny Biotec Cat. No. 130-092-628; LS Columns Cat. No 130-042-401; Myelin removal beads Cat No. 130-096-731; MidiMACSTM separator Cat. No. 130-042-302), according to manufacturers’ instructions. Cells were sorted using an INFLUXTM sorter (BD Biosciences INFLUXTM). After sorting, samples were centrifuged for 40 min at 700 g at 4°C to collect the nuclei before proceeding with the desired technique.

      Whole Genome Bisulfite-Sequencing

      WGBS was performed by CNAG Genome Facility on two independent sets of biological replicates. Briefly, genomic DNA (1–2 μg) was spiked with unmethylated λ DNA (5 ng of λ DNA per microgram of genomic DNA; Promega). DNA was sheared by sonication to 50–500 bp in size using a Covaris E220 sonicator, and fragments of 150–300 bp were selected using AMPure XP beads (Agencourt Bioscience). Genomic DNA libraries were constructed using the Illumina TruSeq Sample Preparation kit following Illumina’s standard protocol. DNA was treated with sodium bisulfite after adaptor ligation, using the EpiTexy Bisulfite kit (Qiagen), following the manufacturer’s instructions for formalin-fixed, paraffin-embedded tissue samples. Two rounds of bisulfite conversion were performed to ensure a conversion rate of >99%. Enrichment for adaptor-ligated DNA was carried out through seven PCR cycles using PfuTurboCx Hot-Start DNA polymerase (Stratagene). Library quality was monitored using the Agilent 2100 Bioanalyzer, and the concentration was determined by quantitative PCR with the library quantification kit from Kapa Biosystems. Paired-end DNA sequencing (2 × 100 bp) was then performed using the Illumina HiSeq 2000 platform.

      Chromatin Accessibility

      Open chromatin studies were performed by ATAC-seq and SONO-seq procedures. Briefly, ATAC-seq was performed with minor modifications from Buenrostro et al. (2013). 100,000 nuclei were treated with 2.5 μl Tn5 at 37°C for 30 min, followed by cleanup on a Qiagen Minelute column. Fragments >1 kb in size were removed using AmpureXP beads (Agencourt AMPure XP, Beckman Coulter Cat. No. A63881). DNA fragments were amplified by 11 cycles of PCR with custom adapter primers from Buenrostro et al. (2013). PCR reactions were cleaned up with AmpureXP beads, quantified by Qubit and quality controlled by Bioanalyzer (Agilent Technologies). SONO-seq consists of isolating and fragmenting crosslinked chromatin, before reversing crosslinks, purifying the DNA and preparing it for sequencing (Auerbach et al., 2009). Chromatin was fragmented by sonication using the same COVARIS specifications as for ChIP-seq (see above) to obtain a median fragment size of 200 bp. Sequencing libraries were prepared using the NEBNext Ultra (New England Cat. No. E7370L) kit according to the manufacturer’s protocol. Both techniques were sequenced on a HiSeq2000 sequencer (Illumina).

      Chromatin Immunoprecipitation: ChIP-Seq

      Nuclei obtained in section “Nucleus Isolation” were cross- linked with 0.5% formaldehyde (Sigma F8775-25ml) for 5 min at room temperature (RT). Residual formaldehyde was quenched by addition of glycine (MAGnifyTM Glycine P/N 100006373) to a final concentration of 0.125 M and incubation for 5 min at RT. Nuclei were pelleted by centrifugation at 500 g during 10 min at 4°C and resuspended in 300 μl lysis buffer (MAGnifyTM Chromatin Immunoprecipitation System, Cat no. 49-2024). Chromatin was fragmented by sonication in a Covaris S2 [Duty Cycle: 20, Intensity: 8, Cycles per Burst:200, for 15 min (histone marks), for 25 min (FACS-sorted nuclei)] to a median size of 200 bp, aliquoted and stored at −80°C until further use. For non-histonic proteins such CTCF, no nuclei preparation was performed. Homogenized tissue was crosslinked with 0.5% formaldehyde for 10 min at RT, quenched and fragmented as above (Duty Cycle: 5, Intensity: 2, Cycles per Burst: 200, Time: 25 min). Chromatin immunoprecipitation was performed using antibodies against histone modifications (H3K27ac, H3K4me3, H3K4me1, H3K79me2, H3K36me3, H3K27me3, H3K9me2, and CTCF) with the MAGnifyTM Chromatin Immunoprecipitation System (Invitrogen Cat no. 49-2024), according to manufacturer’s instructions. For whole cortex and NeuN histonic ChIP-seq a total amount of 50 k nuclei was used per ChIP (∼330 ng), 700 k nuclei (∼4 μg) for non-histonic ChIP-seq. Recovered ChIP DNA was used to construct sequencing libraries, using the NEBNext Ultra (New England Cat. No. E7370L) kit according to the manufacturer’s protocol, and sequenced on a HiSeq2000 sequencer (Illumina). The quality of the ChIP-seq was determined by qPCR, using positive and negative primers to detect the regions where the histones should be placed in the genome (Supplementary Table 1). Primers were diluted to a final concentration of 300 ng in Power SYBR Green PCR MM (Applied Biosystems Cat. No 4367659). Samples were run in a Applied Biosystem qPCR system (7900 HT Fast Real-Time PCR System) as follows: 50°C/2 min, 95°C/10 min, 40 cycles of 95°C/15 s, 60°C/1 min, 95°C/15 s, 60°C-15 s, and 95–15 s.

      Transcriptomics

      Transcriptome study involved both poly-A RNA, directional RNA and small RNA libraries. Poly-A RNA sequencing libraries were prepared from total RNA using the TruSeqTM RNA sample preparation kit (Illumina Inc., Cat. No. RS-122-2001) according to the manufacturer’s protocol. Directional RNA libraries were prepared using the ScriptSeqTM Complete Gold Kit (Human/Mouse/Rat) (Epicentre Biotechnologies), according to the manufacturer’s protocol. Briefly, 3 μg of total RNA were depleted of both cytoplasmic and mitochondrial rRNAs using the Ribo-ZeroTM Gold rRNA Removal Reagents. The total rRNA depletion of the samples was confirmed on a 2100 Bioanalyzer RNA 6000 Pico Chip. For the library preparation we used 50 ng of Ribo-Zero-treated RNA as starting material for the ScriptSeqTM v2 RNA-Seq Library Preparation Kit, followed by amplification by 12 cycles of PCR, using the FailSafeTM PCR Enzyme Mix (Epicentre Biotechnologies) before purification with AMPure XP beads (Agencourt, Beckman Coulter Cat. No. A63881). Both the directional mRNA and the Poly-A RNA libraries were sequenced in paired end mode with read length 2 × 101 bp on a HiSeq2000 (Illumina, Inc.) following the manufacturer’s protocol. After computational analysis, we validated 21 differential expressed genes in a new batch of biological replicates following the method of Schmittgen and Livak 2008 (see Supplementary Table 4). To validate the data generated in the RNA-seq analysis, 1 μg of the sequenced RNAs were used to prepare cDNA with SuperScriptIII (Invitrogen) according to manufacturer’s instructions, cDNAs were normalized to 100 ng/μl. RT-PCRs for the alternative splicing events were performed using oligos annealing to the adjacent constitutive exons and performed under standard conditions; 2% agarose gels were used to resolve the different bands. Image J software was used for quantification of the observed bands and determination of the PSIs for each event. Small RNA libraries were generated using the TruSeq (Illumina, Cat. No. RS-122-2001) kit according to the manufacturer’s protocol. The resulting 22 bp insert libraries were sequenced on a HiSeq2000 sequencer (Illumina), yielding 15–20 million reads per sample. After the analysis, we validated 6 out of 10 miRNA in a second independent group of animals, following Chen et al. protocol with minor modifications: instead of using Taqman probes we designed our own RT-miRNA oligonucleotides and performed qPCRs with Power SYBR Green PCR MM (Applied Biosystems Cat. No. 4367659, see Supplementary Table 5; Chen et al., 2005).

      Proteomics

      Samples were minced with RIPA-M buffer (1% NP40, 1% Sodium deoxycholate, 0.15 M NaCl, 0.001 M EDTA, 0.05 TrisHCl pH = 7.5, 1X cOMPLETE Mini EDTA free, 0.01 M NaF, 0.01 M Sodium pyrophosphate, 0.005 M β-glycerolphophate), sonicated with a Diagenode Bioruptor (cycles of 0.5 min ON 0.5 min OFF, medium intensity during 5 min). Samples were centrifuged during 10 min 16,000 rpm at 4°C and precipitated with acetone at -20°C for 1 hour. Samples were pelleted by centrifugation during 10 min 16,000 rpm at 4°C, dried and resuspended in Urea/200 mM ABC, sonicated again during 10 min (cycles of 0.5 min ON 0.5 min OFF, medium intensity) and quantified prior to mass spectrophotometry injection following isobaric tags for relative and absolute quantitation (iTRAQ) or Liquid Chromatography/Mass-Spectophotometry (LC/MS).

      <italic>In situ</italic>-HiC

      Cerebral cortex samples from individual C57BL/6J mice (2 bio-replicates per EE and CTL conditions) were sorted using NeuN+ (Rbfox3+) as described above. After sorting, approximately 1 million of nuclei were used for in situ HiC following previous specifications (Rao et al., 2014). Libraries were sequenced on a HiSeq2000 (PE × 125 bp) yielding approximately 300 M of reads per sample.

      Quantification and Statistical Analysis <italic>In silico</italic> Identification of Active Enhancers

      Active enhancers for EE and CTL cortex were predicted and annotated using a machine learning approach called Generalized Enhancer Predictor (GEP)2 (Jhanwar et al., 2018). The method performed classification of epigenetic patterns coming from cortical ATAC-seq, H3K4me1, H3K4me3, H3K27ac, H3K36me3, and H3K27me3 using Random Forest (RF) and Support Vector Machine (SVM) classifiers to build predictive models for identification of active enhancers. The total amount of enhancers used in the present study corresponded to the consensus of GEP prediction for EE, control and ENCODE data, resulting in 347112 enhancers (Supplementary Table 1).

      <italic>In silico</italic> Prediction of Chromatin Interactions

      Chromatin interactions were predicted from chromatin modifications using Epitensor with minor modifications to adapt the script to the mouse genome (Zhu et al., 2016). We used epigenetic data from following tissues from the mouse ENCODE project (forebrain, heart, hindbrain, kidney, liver, lung, midbrain, stomach) (Yue et al., 2014). As well as in-house generated data from cortex of animals with and without environmental enrichment. We used the following epigenetic marks: H3K9me3, H3K4me3, H3K4me1, H3K36me3, H3K27me3, H3K27ac, CTCF as well as RNA-seq coverage data. Chromatin accessibility was measured by DNase-seq for ENCODE tissues and ATAC-seq for in-house cortex samples. Promoter regions were defined using the TxDb.Mmusculus.UCSC.mm10.knownGene package in BioConductor as 1,500 up- and 500 down-stream of any possible TSS. Active enhancers in each of the input tissues were defined by training a machine learning classifier on a list of validated enhancers using core epigenetic mark intensities as features (DNase, H3K27ac, H3K27me3, H3K36me3, H4K4me1, H3K4me3 as well as intensity ratio between the last two marks). The trained classifier was then applied to the epigenetic data from ENCODE tissues and local cortex datasets. To limit computation load, possible interactions were limited to intra-TAD interactions, which were based on the set of mouse cortex TADs3 (Dixon et al., 2012). Differential activity in enhancers was annotated using annotate.enhancers.with.genes.sh utility4.

      Whole Genome Bisulfite-Sequencing

      Methylated CpGs were called from the raw reads using Bismark (Krueger and Andrews, 2011) following the user guide. Differential methylation analysis was carried out using bsseq (Hansen et al., 2012). Briefly, CpG methylation values were locally smoothed using the BSmooth function and CpGs that had a coverage of less than 5 reads in any of the samples were removed. We calculated the t statistic for the smoothed CpG values using the BSmooth.tstat function with paired design in local correction mode and the dmrFinder function was used to identify differentially methylated regions (DMRs) that contained multiple CpGs with a t statistic below -4.5 or above 4.5. DMRs located inside or within 5 kb of a gene or enhancer were annotated accordingly. Additionally, DMRs that overlapped enhancers, were annotated with the enhancer’s target gene according to the Epitensor predictions.

      Chromatin Accessibility

      ATAC-seq libraries were aligned to mm10 using bwa-mem with predefined parameters (Li and Durbin, 2010). Duplicate read pairs were marked using the MarkDuplicates command from the Picard software suite5. We used a peak-independent approach on the one hand using our predicted enhancers and promoters as defined above, and on the other hand a peak-dependent method using F-seq or MACS2 with default parameters with and without duplicates (Boyle et al., 2008; Zhang et al., 2008). We also used MACS2 with the shifted strategy with the following parameters: “–nomodel –shift -75 –extsize 150 –broad –keep-dup all.” Together with enhancer and promoter regions, each annotation was loaded into DiffBind (Ross-Innes et al., 2012) providing as background input SONO-seq chromatin (Ross-Innes et al., 2012). For SONO-seq libraries, the pipeline followed the same steps as the ChIP-seq (see below).

      Footprinting analysis was done using the Centipede software using the core transcription factor binding motifs from the Jaspar database (version 2016-03-02) (Pique-Regi et al., 2011; Khan et al., 2018). Instances of each motif in the genome were kept if the PWM score was superior or equal to 13. We created and used a mm10 mappability file to filter out instances that are located in regions of the genome with low mappability (gem-mappability-retriever; Marco-Sola et al., 2012). A motif was considered bound if the P-value-Z-score-combined statistics was inferior to 0.05. A motif was considered differentially bound if the ANOVA p-value was inferior to 0.05. Individual instances of a motif were considered bound when their posterior probabilities were superior to 0.99.

      Chromatin Immunoprecipitation: ChIP-Seq

      Samples were mapped to mouse mm10 (peak-independent) using Bowtie (Langmead et al., 2009) with “–quiet –sam –best –strata -m 1” parameters. Sam files were converted to bam files allowing only reads with mapping quality greater than 30 (“-q 30”). Files were visualized using bamCoverage utility from deepTools (Ramírez et al., 2016). We initially used a peak-dependent strategy as a benchmarking method and also as a tool to define our peak-independent approach (data not shown). The quality of ChIP-seq was assessed by the irreproducible discovery rate (IDR) (Li et al., 2011). The peak-independent method used in the present study consisted in the calculation of the coverage at defined regions as follows. The broad marks H3K36me3 and H3K79me2 were measured along the full gene body, defined as the distance from the TSS to the TES. Enhancer regions were provided by the GEP enhancer predictor in combination with ENCODE enhancers (Jhanwar et al., 2018; Supplementary Table 1). Promoter regions were defined as the interval from 1,500 bp upstream to 500bp downstream of the TSS. Reads were quantified by featurecounts (Liao et al., 2014) with the following parameters: “–ignoreDup –minReadOverlap 25 -Q 1 –O.” Counts tables were supplied to the batch effect corrector RUVseq for further differential analysis using edgeR (Robinson et al., 2010; Risso et al., 2014). A binned approach was used to determine differential changes in 1Mb bins to be associated with inter-chromosomal interactions hubs. This differential analysis was performed with Diffreps (Shen et al., 2013). Using the following parameters: “-pval 0.001 -frag 150 -window 1000.” Batch effects are a major issue in sequencing studies. In this study, we acknowledge the lack of bio-replicates in the cortical tissue by having a wide set of different techniques as well as a pooling strategy of animals per bio-replicate (5 individuals per sample). To minimize batch effects, we randomized the samples, applied standardized procedures and parallelized the experiments as much as possible. However, FAC-sorting experiments could not always be parallelized for different reasons (i.e., availability of the sorter). To remove batch effects, we decided to use the strategy of Russo et al. that was also used in data different from RNAseq (Risso et al., 2014; Koberstein et al., 2018). A principal component analysis was the main criterion to evaluate each dataset and the requirement for batch correction. If a PCA on uncorrected data could clearly separate the conditions, we ruled that there was no need for batch correction. However, if required, we then selected the method(s) (among RUVg, RUVs, and/or RUVr) that were able to separate conditions and intersected the results with a FDR threshold of 0.1 to obtain a conservative final gene list of changes induced by EE. Coverage plots were produced using the function bamCoverage of deepTools to produce bigwig files (Ramírez et al., 2016) (v2.0 with Python 2.7.14). These files were supplied to the function computeMatrix using the “scale-regions” parameter that allows to plot the coverage profile using the plotProfile function along regions of interest. The cell-specificity of sorted populations was assessed by the tool MakerGeneProfile that estimates cell specificity using a curated single-cell mouse brain RNAseq database6. We transformed gene-body assigned reads of H3K79me2 NeuN+ and NeuN into RPKM values and ran MakerGeneProfile to compare neuronal and non-neuronal enrichments to oligodendrocytes, astrocytes and pyramidal neurons specific markers. MGP states for MakerGeneProfile and it is described as a correlate value of cell type proportions. Specifically, MGP scores consist in the summarized expression profiles into the first principal component to estimate cell ratios (Mancarci et al., 2017). Gene ontology term enrichment analysis was performed using the Cytoscape tools clueGO and cluepedia, Metascape and SynGO (Bindea et al., 2009; Koopmans et al., 2019; Zhou et al., 2019). We performed the analysis individually for each histone mark or together, supplying upregulated and downregulated DBS separately. We used Bonferroni step down or Benjamini-Hochberg with p-value thresholds of 0.05 or 0.01 depending of the amount of data provided. In general, larger datasets needed more stringency in the statistical correction (Benjamini and Hochberg, 1995).

      Transcriptomics

      mRNA reads were aligned to mm10 using STAR with standard parameters (Dobin et al., 2013). Reads were counted using featureCounts by “gene_name” (Liao et al., 2014), batch corrected by RUV-seq before differential analysis in edgeR (Robinson et al., 2010; Risso et al., 2014). We also provide a splicing analysis results compiled in Supplementary Table 4. To identify and quantify alternative splicing, we used vast-tools v1.0.0 (Tapial et al., 2017). To increase effective read coverage at splice junctions, we pooled all replicates for polyA, respectively ribominus, samples using vast-tools merge. Differential splicing analysis was performed using vast-tools compare, comparing EE with Ctl samples in a paired manner, using the parameters –min_dPSI 10 –min_range 5 –p_IR –noVLOW. This resulted in, respectively, 40 and 53, cassette exons being up- or downregulated with EE (including 2 and 1 microexons; Irimia et al., 2014), 49 and 43 retained introns, 27 and 22 alternative 3’ss choices and 28 and 23 alternative 5’ss splice choices.

      Regarding the lncRNA analysis, total RNA reads were aligned to mm10 using Subjunc splice-aware aligner with default settings (Liao et al., 2013), and reads overlapping exons were summarized at the gene-level to the corresponding genes using featureCounts (Liao et al., 2014). Read assignment to exons was carried out in a strand-aware manner, only fragments with both mates correctly aligned were considered and genomic regions with multiple overlapping exons on the same strand were disregarded. The count matrix was further filtered to retain only GENCODE long non-coding RNA (GRCm38.p5_M15). Reads with an average log2 CPM < 0.9 across all samples were filtered out yielding 4,472 genes. Normalization factors were calculated using the TMM method from edgeR package (Robinson et al., 2010). Observational-level weights were calculated using voom (Law et al., 2014) and used to fit gene-wise linear models (Smyth, 2004). Multiple testing p-value adjustment was performed using the Benjamini-Hochberg method (Benjamini and Hochberg, 1995).

      Small RNA reads with homo-polymer and low PHRED scores were removed using FASTQ-Toolkit and a custom script. SeqBuster was used to remove adapters and align using miraligner.jar with mouse miRbase v18 annotation (Pantano et al., 2010). The small RNA dataset presented a strong batch effect which could not be corrected by RUVseq (Risso et al., 2014). We therefore devised an alternative strategy which consisted of filtering microRNAs for low read coverage (<50 counts), normalizing the libraries by read counts per million (rpm), before contrasting EE against CTL batch-wise and considering exclusively reproducible direction of change. Additionally, we calculated z-scores validating partially previous strategy (Supplementary Table 5).

      Proteomics

      The analysis of iTRAQ data consisted in sorting the discrepancy (δ = φ/β) between both biological contrasts (φ = EE1/CTL1 and β = EE2/CTL2), following the premise that consistent results will be indicated by:

      l i m δ = 1 φ β

      Discrepant results then will be considered as values far from δ = 1. Based on this consideration we select all the proteins that follow the condition and we established a threshold of ±0.1. The protein expression values from the LC/MS were log2-transformed and loess normalized using the normalize.ExpressionSet.loess function from the BioConductor package AffyPML. Differential expression analysis was conducted with a standard limma (BioConductor) pipeline by calculating sample weights, fitting a linear model for each gene across all samples and calculating moderated F-statistics. Unadjusted p-values were used to rank the proteins (Smyth, 2004).

      <italic>In situ</italic> Hi-C

      For quality check, sequencing reads were mapped to the mouse reference genome assembly (mm10), artifacts were filtered, and library was ICED normalized using the Hi-C-Pro (Servant et al., 2015) (v2.9.0) (Supplementary Table 7). For visualization we used the hicpro2juicebox.sh utility to convert the previous into a .hic format to visualize heatmap interaction matrices in Juicebox (Durand et al., 2016a). Juicer_tools were used to calculate A/B compartments using eigenvector utility (Durand et al., 2016b). We called chromatin loops with HICCUP at 5 and 10 kb of resolution (Durand et al., 2016a). For differential analysis, resulting interactions from Hi-C-Pro were splitted into intra and interchromsomal interactions, sex chromosomes and self-interacting bins were removed. Then piped into the edgeR wrapper RUVseq were RUVr for intra and RUVg for inter were used to normalized batch effects.

      Required EE and CTL gtrack file to run chrom3D (Paulsen et al., 2017) was produced using the chrom3D wrapper automat_chrom3D utility7. Domains were called using Arrowhead (Juicer tools 1.7.6; Durand et al., 2016b). “–ignore_sparsity” parameter was used, and calls could be only produced at not lower than 10 kb. For the present study, we finally selected 5 M iterations including the parameter “–nucleus” to force the beads to remain confined inside the designed radius: “-r 3.0.” Domain coloring was produced by automat_color8 that allows to color any region of interest in the model. Both in silico models are available Supplementary Table 7.

      Data Integration

      We used Metascape to intersect the totality of the data. Some of the results were clumped as the maximum amount of sets allowed is 30 (Zhou et al., 2019). Transcriptomic and proteomic changes percentages explain by the rest of the data were calculated out of the evidences table reported as Metascape result (Supplementary Table 8).

      Linear dependency test was performed by using Pearson and Spearman correlations to test how enhancer and promoter activity of different marks influence differential changes observed in the transcriptome and proteome. Briefly, normalized counts by RPKM mapping into enhancers and promoters were first averaged by target gene name they interact with using EpiTensor (Zhu et al., 2016). Then a fold change was calculated of EE vs. CTL samples. These values then are correlated with the corresponding fold changes found in differential RNA-seq and proteomic analysis. We used Spearman for proteomic changes as the scale of the fold changes were considerably different coming directly from iTRAQ data and the method is less sensitive for these outliers.

      In order to test the association of differential regions induced by EE with inter-chromosomal interactions we run a permutation analysis using the package regioneR. Both permutations shown in the study were performed with a total number of 100,000 iterations. GWAS brain trait studies are collected in detail in Supplementary Table 8.

      Data Availability Statement

      Datasets are currently accessible in the SRA repository: https://www.ncbi.nlm.nih.gov/sra/SRP154319. Processed data and other resources can be found in the dedicated website: www.mousecortexregulome.org.

      Ethics Statement

      The animal study was reviewed and approved by Comité Ético de Experimentación Animal del PRBB (Procedure Code: ISA-11-1358).

      Author Contributions

      SE-G performed most of the experimental procedures (sorted ATAC-seq samples, all ChIP-seq, RNA-seq and miRNA validations, in situ HiC, and proteome preparation for LS/MS), carried out most of the analysis (ATAC-seq, ChIP-seq, RNA-seq, miRNA, proteome, and in situ HiC), and wrote the manuscript. CNH performed part of the experimental procedures (bisulfite-seq, cortex homogenate ATAC-seq, RNA-seq preparation, and proteome preparation for iTRAQ). AH performed the bisulfite-seq analysis, differential lncRNAs analysis, and run Epitensor. SB and RP-R performed the footprint analysis. SJ run the GEP enhancer predictor analysis. SC performed Diffreps and epigenetic relationship analysis with HiC datasets. JA-R and MP-E performed animal environmental enrichment procedures, breedings, and dissections. MM performed library preparations. MI carried out the splicing analysis. JP did the validation. MF effectuated the miRNA mapping. SA, MD, PM, CNH, and SO worked in the elaboration of the manuscript and funded the study. All authors contributed to the article and approved the submitted version.

      Conflict of Interest

      The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

      Funding. We acknowledge support of the Spanish Ministry of Economy and Competitiveness (SAF2011-26216), “Centro de Excelencia Severo Ochoa 2017-2021,” SEV-2016-0571, the CERCA Programme/Generalitat de Catalunya and Jerome Lejeune Foundation, Swiss National Science Foundation Fellowship (PBLAP3_136878) and Co-funded by Marie Curie Actions to CNH. Resources for analyses conducted by SE-G were partially supported by the U.S. National Institutes of Mental Health Funds R01MH104341 and R01MH117790 and by the Social Sciences and Humanities Research Council of Canada (NFRFE-2018-01305). We acknowledge support of the Spanish Ministry of Science and Innovation to the EMBL partnership, Agencia Estatal de Investigaci n (PID2019-110755RB-I00/AEI / 10.13039/501100011033), the European Union’s Horizon 2020 Research and Innovation programme under grant agreement No 848077, Jerôme Lejeune Foundation, NIH (Grant Number: 1R01EB 028159-01), Marató TV3 (#2016/20-30). RP-R resources were supported by R01GM109215. We thank the support of the University of Tübingen for the Open Access Publication Funds contribution.

      Special thanks to the “Centre for Genomic Regulation Core Facilities”: Irene González Navarrete, María Angustias Aguilar Morón, Anna Menoyo Vilalta, Núria Andreu Somavilla, and Jochen Hecht from the Genomics Facility and Òscar Fornas, Eva Julià Arteaga, Erika Ramírez Bautista, and Alexandre Bote Tronchoni from the CRG FAC-sorting Unit. Also, we would like to thank to Domenica Marchese for helping with microRNA validation and Jekaterina Kokatjuhha and Mattia Bosio for their computational help support.

      Supplementary Material

      The Supplementary Material for this article can be found online at: /articles/10.3389/fnmol.2021.664912/full#supplementary-material

      References Auerbach R. K. Euskirchen G. Rozowsky J. Lamarre-Vincent N. Moqtaderi Z. Lefrançois P. (2009). Mapping accessible chromatin regions using Sono-Seq. Proc. Natl. Acad. Sci. U.S.A. 106 1492614931. 10.1073/pnas.0905443106 19706456 Ball N. J. Mercado E. Orduña I. (2019). Enriched environments as a potential treatment for developmental disorders: a critical assessment. Front. Psychol. 10:466. 10.3389/fpsyg.2019.00466 30894830 Beagan J. A. Pastuzyn E. D. Fernandez L. R. Guo M. H. Feng K. Titus K. R. (2020). Three-dimensional genome restructuring across timescales of activity-induced neuronal gene expression. Nat. Neurosci. 23 707717. 10.1038/s41593-020-0634-6 32451484 Benjamini Y. Hochberg Y. (1995). Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. Ser. B 57 289300. 10.2307/2346101 Berardo L. R. Fabio M. C. Pautassi R. M. (2016). Post-weaning environmental enrichment, but not chronic maternal isolation, enhanced ethanol intake during periadolescence and early adulthood. Front. Behav. Neurosci. 10:195. 10.3389/fnbeh.2016.00195 27790100 Betke K. M. Wells C. A. Hamm H. E. (2012). GPCR mediated regulation of synaptic transmission. Prog. Neurobiol. 96 304321. 10.1016/j.pneurobio.2012.01.009 22307060 Bikbaev A. Frischknecht R. Heine M. (2015). Brain extracellular matrix retains connectivity in neuronal networks. Sci. Rep. 5:14527. 10.1038/srep14527 26417723 Bindea G. Mlecnik B. Hackl H. Charoentong P. Tosolini M. Kirilovsky A. (2009). ClueGO: a Cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks. Bioinformatics 25 10911093. 10.1093/bioinformatics/btp101 19237447 Bond A. M. Bhalala O. G. Kessler J. A. (2012). The dynamic role of bone morphogenetic proteins in neural stem cell fate and maturation. Dev. Neurobiol. 72 10681084. 10.1002/dneu.22022 22489086 Boyle A. P. Guinney J. Crawford G. E. Furey T. S. (2008). F-Seq: a feature density estimator for high-throughput sequence tags. Bioinformatics 24 25372538. 10.1093/bioinformatics/btn480 18784119 Brooks E. Canal M. M. (2013). Development of circadian rhythms: role of postnatal light environment. Neurosci. Biobehav. Rev. 37 551560. 10.1016/j.neubiorev.2013.02.012 23454636 Buenrostro J. D. Giresi P. G. Zaba L. C. Chang H. Y. Greenleaf W. J. (2013). Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat. Methods 10 12131218. 10.1038/nmeth.2688 24097267 Chen C. Ridzon D. A. Broomer A. J. Zhou Z. Lee D. H. Nguyen J. T. (2005). Real-time quantification of microRNAs by stem-loop RT-PCR. Nucleic Acids Res. 33:e179. 10.1093/nar/gni178 16314309 Chen Y.-J. J. Johnson M. A. Lieberman M. D. Goodchild R. E. Schobel S. Lewandowski N. (2008). Type III neuregulin-1 is required for normal sensorimotor gating, memory-related behaviors, and corticostriatal circuit components. J. Neurosci. 28 68726883. 10.1523/JNEUROSCI.1815-08.2008 18596162 Chowdhury S. Shepherd J. D. Okuno H. Lyford G. Petralia R. S. Plath N. (2006). Arc/Arg3.1 interacts with the endocytic machinery to regulate AMPA receptor trafficking. Neuron 52 445459. 10.1016/j.neuron.2006.08.033 17088211 Clemenson G. D. Deng W. Gage F. H. (2015). Environmental enrichment and neurogenesis: from mice to humans. Curr. Opin. Behav. Sci. 4 5662. 10.1016/j.cobeha.2015.02.005 Coffey E. T. (2014). Nuclear and cytosolic JNK signalling in neurons. Nat. Rev. Neurosci. 15 285299. 10.1038/nrn3729 24739785 DaSilva L. L. P. Wall M. J. de Almeida L. P. Wauters S. C. Januário Y. C. Müller J. (2016). Activity-regulated cytoskeleton-associated protein controls AMPAR endocytosis through a direct interaction with clathrin-adaptor protein 2. eNeuro 3 125140. 10.1523/ENEURO.0144-15.2016 27257628 Dixon J. R. Selvaraj S. Yue F. Kim A. Li Y. Shen Y. (2012). Topological domains in mammalian genomes identified by analysis of chromatin interactions. Nature 485 376380. 10.1038/nature11082 22495300 Dobin A. Davis C. A. Schlesinger F. Drenkow J. Zaleski C. Jha S. (2013). STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29 1521. 10.1093/bioinformatics/bts635 23104886 Durand N. C. Robinson J. T. Shamim M. S. Machol I. Mesirov J. P. Lander E. S. (2016a). Juicebox provides a visualization system for Hi-C contact maps with unlimited zoom. Cell Syst. 3 99101. 10.1016/j.cels.2015.07.012 27467250 Durand N. C. Shamim M. S. Machol I. Rao S. S. P. Huntley M. H. Lander E. S. (2016b). Juicer provides a one-click system for analyzing loop-resolution Hi-C experiments. Cell Syst. 3 9598. 10.1016/j.cels.2016.07.002 27467249 Edbauer D. Neilson J. R. Foster K. A. Wang C.-F. Seeburg D. P. Batterton M. N. (2010). Regulation of synaptic structure and function by FMRP-associated microRNAs miR-125b and miR-132. Neuron 65 373384. 10.1016/j.neuron.2010.01.005 20159450 Faraji J. Karimi M. Soltanpour N. Moharrerie A. Rouhzadeh Z. Lotfi H. (2018). Oxytocin-mediated social enrichment promotes longer telomeres and novelty seeking. Elife 7:e40262. 10.7554/eLife.40262 30422111 Fernandez-Albert J. Lipinski M. Lopez-Cascales M. T. Rowley M. J. Martin-Gonzalez A. M. del Blanco B. (2019). Immediate and deferred epigenomic signature of neuronal activation. bioRxiv [Preprint]. 10.1101/534115 Ferrer-Ferrer M. Dityatev A. (2018). Shaping synapses by the neural extracellular matrix. Front. Neuroanat. 12:40. 10.3389/fnana.2018.00040 29867379 Friedman Y. Naamati G. Linial M. (2010). MiRror: a combinatorial analysis web tool for ensembles of microRNAs and their targets. Bioinformatics 26 19201921. 10.1093/bioinformatics/btq298 20529892 Fu S. Wang Q. Moore J. E. Purcaro M. J. Pratt H. E. Fan K. (2018). Differential analysis of chromatin accessibility and histone modifications for predicting mouse developmental enhancers. Nucleic Acids Res. 46 1118411201. 10.1093/nar/gky753 30137428 Grégoire C.-A. Tobin S. Goldenstein B. L. Samarut É Leclerc A. Aumont A. (2018). RNA-sequencing reveals unique transcriptional signatures of running and running-independent environmental enrichment in the adult mouse dentate gyrus. Front. Mol. Neurosci. 11:126. 10.3389/fnmol.2018.00126 29706867 Guo Y. Xu Q. Canzio D. Shou J. Li J. Gorkin D. U. (2015). CRISPR inversion of CTCF sites alters genome topology and enhancer/promoter function. Cell 162 900910. 10.1016/j.cell.2015.07.038 26276636 Hansen K. D. Langmead B. Irizarry R. A. (2012). BSmooth: from whole genome bisulfite sequencing reads to differentially methylated regions. Genome Biol. 13:R83. 10.1186/gb-2012-13-10-r83 23034175 He C. W. Liao C. P. Pan C. L. (2018). Wnt signalling in the development of axon, dendrites and synapses. Open Biol. 8:180116. 10.1098/rsob.180116 30282660 Heun-Johnson H. Levitt P. (2018). Differential impact of Met receptor gene interaction with early-life stress on neuronal morphology and behavior in mice. Neurobiol. Stress 8 1020. 10.1016/j.ynstr.2017.11.003 29255778 Hu Y.-S. Long N. Pigino G. Brady S. T. Lazarov O. (2013). Molecular mechanisms of environmental enrichment: impairments in Akt/GSK3β, neurotrophin-3 and CREB signaling. PLoS One 8:e64460. 10.1371/journal.pone.0064460 23700479 Hübener M. Bonhoeffer T. (2014). Neuronal plasticity: beyond the critical period. Cell 159 727737. 10.1016/j.cell.2014.10.035 25417151 Huff J. T. Plocik A. M. Guthrie C. Yamamoto K. R. (2010). Reciprocal intronic and exonic histone modification regions in humans. Nat. Struct. Mol. Biol. 17 14951499. 10.1038/nsmb.1924 21057525 Irier H. Street R. C. Dave R. Lin L. Cai C. Davis T. H. (2014). Environmental enrichment modulates 5-hydroxymethylcytosine dynamics in hippocampus. Genomics 104 376382. 10.1016/j.ygeno.2014.08.019 25205305 Irimia M. Weatheritt R. J. Ellis J. D. Parikshak N. N. Gonatopoulos-Pournatzis T. Babor M. (2014). A highly conserved program of neuronal microexons is misregulated in autistic brains. Cell 159 15111523. 10.1016/J.CELL.2014.11.035 25525873 Jhanwar S. Ossowski S. Davila-Velderrain J. (2018). Genome-wide active enhancer identification using cell type-specific signatures of epigenomic activity. bioRxiv [Preprint]. 10.1101/421230 Jiang Y. Matevossian A. Huang H.-S. Straubhaar J. Akbarian S. (2008). Isolation of neuronal chromatin from brain tissue. BMC Neurosci. 9:42. 10.1186/1471-2202-9-42 18442397 Kao W. T. Wang Y. Kleinman J. E. Lipska B. K. Hyde T. M. Weinberger D. R. (2010). Common genetic variation in neuregulin 3 (NRG3) influences risk for schizophrenia and impacts NRG3 expression in human brain. Proc. Natl. Acad. Sci. U.S.A. 107 1561915624. 10.1073/pnas.1005410107 20713722 Karlić R. Chung H. R. Lasserre J. Vlahoviček K. Vingron M. (2010). Histone modification levels are predictive for gene expression. Proc. Natl. Acad. Sci. U.S.A. 107 29262931. 10.1073/pnas.0909344107 20133639 Kasnauskiene J. Ciuladaite Z. Preiksaitiene E. Matulevičiene A. Alexandrou A. Koumbaris G. (2012). A single gene deletion on 4q28.3: PCDH18 – a new candidate gene for intellectual disability? Eur. J. Med. Genet. 55 274277. 10.1016/j.ejmg.2012.02.010 22450339 Kempermann G. (2019). Environmental enrichment, new neurons and the neurobiology of individuality. Nat. Rev. Neurosci. 20 235245. 10.1038/s41583-019-0120-x 30723309 Khan A. Fornes O. Stigliani A. Gheorghe M. Castro-Mondragon J. A. van der Lee R. (2018). JASPAR 2018: update of the open-access database of transcription factor binding profiles and its web framework. Nucleic Acids Res. 46 D260D266. 10.1093/nar/gkx1126 29140473 Kim S. Yu N. K. Kaang B.-K. (2015). CTCF as a multifunctional protein in genome regulation and gene expression. Exp. Mol. Med. 47:e166. 10.1038/emm.2015.33 26045254 Kim S. Yu N. K. Shim K. W. Kim J. I. Kim H. Han D. H. (2018). Remote memory and cortical synaptic plasticity require neuronal CCCTC-binding factor (CTCF). J. Neurosci. 38 50425052. 10.1523/JNEUROSCI.2738-17.2018 29712785 Kim T. K. Hemberg M. Gray J. M. Costa A. M. Bear D. M. Wu J. (2010). Widespread transcription at neuronal activity-regulated enhancers. Nature 465 182187. 10.1038/nature09033 20393465 Klemm S. L. Shipony Z. Greenleaf W. J. (2019). Chromatin accessibility and the regulatory epigenome. Nat. Rev. Genet. 20 207220. 10.1038/s41576-018-0089-8 30675018 Koberstein J. N. Poplawski S. G. Wimmer M. E. Porcari G. Kao C. Gomes B. (2018). Learning-dependent chromatin remodeling highlights noncoding regulatory regions linked to autism. Sci. Signal. 11:eaan6500. 10.1126/scisignal.aan6500 29339533 Koopmans F. van Nierop P. Andres-Alonso M. Byrnes A. Cijsouw T. Coba M. P. (2019). SynGO: an evidence-based, expert-curated knowledge base for the synapse. Neuron 103 217234.e4. 10.1016/j.neuron.2019.05.002 31171447 Krueger F. Andrews S. R. (2011). Bismark: a flexible aligner and methylation caller for Bisulfite-Seq applications. Bioinformatics 27 15711572. 10.1093/bioinformatics/btr167 21493656 Langmead B. Trapnell C. Pop M. Salzberg S. L. (2009). Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10:R25. 10.1186/gb-2009-10-3-r25 19261174 Law C. W. Chen Y. Shi W. Smyth G. K. (2014). voom: precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol. 15:R29. 10.1186/gb-2014-15-2-r29 24485249 Li H. Durbin R. (2010). Fast and accurate long-read alignment with Burrows–Wheeler transform. Bioinformatics 26 589595. 10.1093/bioinformatics/btp698 20080505 Li Q. Brown J. B. Huang H. Bickel P. J. (2011). Measuring reproducibility of high-throughput experiments. Ann. Appl. Stat. 5 17521779. 10.1214/11-AOAS466 24568719 Liao Y. Smyth G. K. Shi W. (2013). The subread aligner: fast, accurate and scalable read mapping by seed-and-vote. Nucleic Acids Res. 41:e108. 10.1093/nar/gkt214 23558742 Liao Y. Smyth G. K. Shi W. (2014). featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30 923930. 10.1093/bioinformatics/btt656 24227677 Lichti C. F. Fan X. English R. D. Zhang Y. Li D. Kong F. (2014). Environmental enrichment alters protein expression as well as the proteomic response to cocaine in rat nucleus accumbens. Front. Behav. Neurosci. 8:246. 10.3389/fnbeh.2014.00246 25100957 Lieberman-Aiden E. van Berkum N. L. Williams L. Imakaev M. Ragoczy T. Telling A. (2009). Comprehensive mapping of long-range interactions reveals folding principles of the human genome. Science 326 289293. 10.1126/science.1181369 19815776 Lüscher C. Malenka R. C. (2012). NMDA receptor-dependent long-term potentiation and long-term depression (LTP/LTD). Cold Spring Harb. Perspect. Biol. 4:a005710. 10.1101/cshperspect.a005710 22510460 Maass P. G. Barutcu A. R. Rinn J. L. (2019). Interchromosomal interactions: a genomic love story of kissing chromosomes. J. Cell Biol. 218 2738. 10.1083/jcb.201806052 30181316 Maher M. P. Wu N. Ravula S. Ameriks M. K. Savall B. M. Liu C. (2016). Discovery and characterization of AMPA receptor modulators selective for TARP-γ8. J. Pharmacol. Exp. Ther. 357 394414. 10.1124/jpet.115.231712 26989142 Mancarci B. O. Toker L. Tripathy S. J. Li B. Rocco B. Sibille E. (2017). Cross-laboratory analysis of brain cell type transcriptomes with applications to interpretation of bulk tissue data. eNeuro 4:ENEURO.0212-17.2017. 10.1523/ENEURO.0212-17.2017 29204516 Marco-Sola S. Sammeth M. Guigó R. Ribeca P. (2012). The GEM mapper: fast, accurate and versatile alignment by filtration. Nat. Methods 9 11851188. 10.1038/nmeth.2221 23103880 Martínez-Cué C. Baamonde C. Lumbreras M. Paz J. Davisson M. T. Schmidt C. (2002). Differential effects of environmental enrichment on behavior and learning of male and female Ts65Dn mice, a model for Down syndrome. Behav. Brain Res. 134 185200. 10.1016/S0166-4328(02)00026-8 Martino A. Ettorre M. Musilli M. Lorenzetto E. Buffelli M. Diana G. (2013). Rho GTPase-dependent plasticity of dendritic spines in the adult brain. Front. Cell. Neurosci. 7:62. 10.3389/fncel.2013.00062 23734098 McDonald M. W. Hayward K. S. Rosbergen I. C. M. Jeffers M. S. Corbett D. (2018). Is environmental enrichment ready for clinical application in human post-stroke rehabilitation? Front. Behav. Neurosci. 12:135. 10.3389/fnbeh.2018.00135 30050416 Monahan K. Horta A. Lomvardas S. (2019). LHX2- and LDB1-mediated trans interactions regulate olfactory receptor choice. Nature 565 448453. 10.1038/s41586-018-0845-0 30626972 Morse S. Butler A. Davis R. Soller I. Lubin F. (2015). Environmental enrichment reverses histone methylation changes in the aged hippocampus and restores age-related memory deficits. Biology (Basel) 4 298313. 10.3390/biology4020298 25836028 Mulligan M. K. Abreo T. Neuner S. M. Parks C. Watkins C. E. Houseal M. T. (2019). Identification of a functional non-coding variant in the GABAA receptor α2 subunit of the C57BL/6J mouse reference genome: major implications for neuroscience research. Front. Genet. 10:188. 10.3389/fgene.2019.00188 30984232 Ohline S. M. Abraham W. C. (2019). Environmental enrichment effects on synaptic and cellular physiology of hippocampal neurons. Neuropharmacology 145(Pt A) 312. 10.1016/j.neuropharm.2018.04.007 29634984 Pantano L. Estivill X. Martí E. (2010). SeqBuster, a bioinformatic tool for the processing and analysis of small RNAs datasets, reveals ubiquitous miRNA modifications in human embryonic cells. Nucleic Acids Res. 38:e34. 10.1093/nar/gkp1127 20008100 Paraskevopoulou M. D. Vlachos I. S. Karagkouni D. Georgakilas G. Kanellos I. Vergoulis T. (2016). DIANA-LncBase v2: indexing microRNA targets on non-coding transcripts. Nucleic Acids Res. 44 D231D238. 10.1093/nar/gkv1270 26612864 Paulsen J. Sekelja M. Oldenburg A. R. Barateau A. Briand N. Delbarre E. (2017). Chrom3D: three-dimensional genome modeling from Hi-C and nuclear lamin-genome contacts. Genome Biol. 18:21. 10.1186/s13059-016-1146-2 28137286 Peter M. Aschauer D. F. Pandolfo R. V. Sinning A. Grössl F. Kargl D. (2020). Rapid nucleus-scale reorganization of chromatin in neurons enables transcriptional adaptation for memory consolidation. bioRxiv [Preprint]. 10.1101/2020.12.03.409623 Pique-Regi R. Degner J. F. Pai A. A. Gaffney D. J. Gilad Y. Pritchard J. K. (2011). Accurate inference of transcription factor binding from DNA sequence and chromatin accessibility data. Genome Res. 21 447455. 10.1101/gr.112623.110 21106904 Pons-Espinal M. Martinez de Lagran M. Dierssen M. (2013). Environmental enrichment rescues DYRK1A activity and hippocampal adult neurogenesis in TgDyrk1A. Neurobiol. Dis. 60 1831. 10.1016/j.nbd.2013.08.008 23969234 Pu M. Ni Z. Wang M. Wang X. Wood J. G. Helfand S. L. (2015). Trimethylation of Lys36 on H3 restricts gene expression change during aging and impacts life span. Genes Dev. 29 718731. 10.1101/gad.254144.114 25838541 Quinodoz S. A. Ollikainen N. Tabak B. Palla A. Schmidt J. M. Detmar E. (2018). Higher-order inter-chromosomal hubs shape 3D genome organization in the nucleus. Cell 174 744757.e24. 10.1016/j.cell.2018.05.024 29887377 Rajarajan P. Borrman T. Liao W. Espeso-Gil S. Chandrasekaran S. Jiang Y. (2019). Spatial genome exploration in the context of cognitive and neurological disease. Curr. Opin. Neurobiol. 59 112119. 10.1016/j.conb.2019.05.007 31255842 Ramírez F. Ryan D. P. Grüning B. Bhardwaj V. Kilpert F. Richter A. S. (2016). deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 44 W160W165. 10.1093/nar/gkw257 27079975 Rao S. S. P. Huntley M. H. Durand N. C. Stamenova E. K. Bochkov I. D. Robinson J. T. (2014). A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell 159 16651680. 10.1016/j.cell.2014.11.021 25497547 Rinaldi L. Datta D. Serrat J. Morey L. Solanas G. Avgustinova A. (2016). Dnmt3a and Dnmt3b associate with enhancers to regulate human epidermal stem cell homeostasis. Cell Stem Cell 19 491501. 10.1016/j.stem.2016.06.020 27476967 Risso D. Ngai J. Speed T. P. Dudoit S. (2014). Normalization of RNA-seq data using factor analysis of control genes or samples. Nat. Biotechnol. 32 896902. 10.1038/nbt.2931 25150836 Robinson M. D. McCarthy D. J. Smyth G. K. (2010). edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26 139140. 10.1093/bioinformatics/btp616 19910308 Ronowska A. Szutowicz A. Bielarczyk H. Gul-Hinc S. Klimaszewska-Łata J. Dyś A. (2018). The regulatory effects of Acetyl-CoA distribution in the healthy and diseased brain. Front. Cell. Neurosci. 12:169. 10.3389/fncel.2018.00169 30050410 Ross-Innes C. S. Stark R. Teschendorff A. E. Holmes K. A. Ali H. R. Dunning M. J. (2012). Differential oestrogen receptor binding is associated with clinical outcome in breast cancer. Nature 481 389393. 10.1038/nature10730 22217937 Rountree-Harrison D. Burton T. J. Leamey C. A. Sawatari A. (2018). Environmental enrichment expedites acquisition and improves flexibility on a temporal sequencing task in mice. Front. Behav. Neurosci. 12:51. 10.3389/fnbeh.2018.00051 29599712 Sadakata T. Washida M. Iwayama Y. Shoji S. Sato Y. Ohkura T. (2007). Autistic-like phenotypes in Cadps2-knockout mice and aberrant CADPS2 splicing in autistic patients. J. Clin. Invest. 117 931943. 10.1172/JCI29031 17380209 Sams D. S. Nardone S. Getselter D. Raz D. Tal M. Rayi P. R. (2016). Neuronal CTCF is necessary for basal and experience-dependent gene regulation, memory formation, and genomic structure of BDNF and Arc. Cell Rep. 17 24182430. 10.1016/j.celrep.2016.11.004 27880914 Schmittgen T. D. Livak K. J. (2008). Analyzing real-time PCR data by the comparative CT method. Nat. Protoc. 3 11011108. 10.1038/nprot.2008.73 18546601 Servant N. Varoquaux N. Lajoie B. R. Viara E. Chen C.-J. Vert J.-P. (2015). HiC-Pro: an optimized and flexible pipeline for Hi-C data processing. Genome Biol. 16:259. 10.1186/s13059-015-0831-x 26619908 Shen L. Shao N.-Y. Liu X. Maze I. Feng J. Nestler E. J. (2013). diffReps: detecting differential chromatin modification sites from ChIP-seq data with biological replicates. PLoS One 8:e65598. 10.1371/journal.pone.0065598 23762400 Shepherd J. D. Rumbaugh G. Wu J. Chowdhury S. Plath N. Kuhl D. (2006). Arc/Arg3.1 mediates homeostatic synaptic scaling of AMPA receptors. Neuron 52 475484. 10.1016/j.neuron.2006.08.034 17088213 Smagin D. A. Galyamina A. G. Kovalenko I. L. Babenko V. N. Kudryavtseva N. N. (2018). Aberrant expression of collagen family genes in the brain regions developing under agonistic interactions in male mice. bioRxiv [Preprint], 120. 10.1101/276063 Smyth G. K. (2004). Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat. Appl. Genet. Mol. Biol. 3 125. 10.2202/1544-6115.1027 16646809 Sparling J. E. Barbeau K. Boileau K. Konkle A. T. M. (2020). Environmental enrichment and its influence on rodent offspring and maternal behaviours, a scoping style review of indices of depression and anxiety. Pharmacol. Biochem. Behav. 197:172997. 10.1016/j.pbb.2020.172997 32702399 Speisman R. B. Kumar A. Rani A. Pastoriza J. M. Severance J. E. Foster T. C. (2013). Environmental enrichment restores neurogenesis and rapid acquisition in aged rats. Neurobiol. Aging 34 263274. 10.1016/j.neurobiolaging.2012.05.023 22795793 Stephan A. H. Barres B. A. Stevens B. (2012). The complement system: an unexpected role in synaptic pruning during development and disease. Annu. Rev. Neurosci. 35 369389. 10.1146/annurev-neuro-061010-113810 22715882 Stevenson J. R. McMahon E. K. Boner W. Haussmann M. F. (2019). Oxytocin administration prevents cellular aging caused by social isolation. Psychoneuroendocrinology 103 5260. 10.1016/j.psyneuen.2019.01.006 30640038 Su Y. Shin J. Zhong C. Wang S. Roychowdhury P. Lim J. (2017). Neuronal activity modifies the chromatin accessibility landscape in the adult brain. Nat. Neurosci. 20 476483. 10.1038/nn.4494 28166220 Sztainberg Y. Chen A. (2010). An environmental enrichment model for mice. Nat. Protoc. 5 15351539. 10.1038/nprot.2010.114 20725068 Tan M. C. Widagdo J. Chau Y. Q. Zhu T. Wong J. J.-L. Cheung A. (2017). The activity-induced long non-coding RNA Meg3 modulates AMPA receptor surface expression in primary cortical neurons. Front. Cell. Neurosci. 11:124. 10.3389/fncel.2017.00124 28515681 Tapial J. Ha K. C. H. Sterne-Weiler T. Gohr A. Braunschweig U. Hermoso-Pulido A. (2017). An atlas of alternative splicing profiles and functional associations reveals new regulatory programs and genes that simultaneously express multiple major isoforms. Genome Res. 27 17591768. 10.1101/gr.220962.117 28855263 Tsai L.-C. L. Chan G. C.-K. Nangle S. N. Shimizu-Albergine M. Jones G. L. Storm D. R. (2012). Inactivation of Pde8b enhances memory, motor performance, and protects against age-induced motor coordination decay. Genes Brain Behav. 11 837847. 10.1111/j.1601-183X.2012.00836.x 22925203 Turrigiano G. G. (2008). The self-tuning neuron: synaptic scaling of excitatory synapses. Cell 135 422435. 10.1016/j.cell.2008.10.008 18984155 van Praag H. Kempermann G. Gage F. H. (2000). Neural consequences of enviromental enrichment. Nat. Rev. Neurosci. 1 191198. 10.1038/35044558 11257907 Wassouf Z. Hentrich T. Samer S. Rotermund C. Kahle P. J. Ehrlich I. (2018). Environmental enrichment prevents transcriptional disturbances induced by alpha-synuclein overexpression. Front. Cell. Neurosci. 12:112. 10.3389/fncel.2018.00112 29755323 Wright J. W. Harding J. W. (2009). Contributions of matrix metalloproteinases to neural plasticity, habituation, associative learning and drug addiction. Neural Plast. 2009:579382. 10.1155/2009/579382 20169175 Xu J. de Winter F. Farrokhi C. Rockenstein E. Mante M. Adame A. (2016). Neuregulin 1 improves cognitive deficits and neuropathology in an Alzheimer’s disease model. Sci. Rep. 6:31692. 10.1038/srep31692 27558862 Yamada T. Yang Y. Valnegri P. Juric I. Abnousi A. Markwalter K. H. (2019). Sensory experience remodels genome architecture in neural circuit to drive motor learning. Nature 569 708713. 10.1038/s41586-019-1190-7 31068695 Yang R. Walder-Christensen K. K. Kim N. Wu D. Lorenzo D. N. Badea A. (2019). ANK2 autism mutation targeting giant ankyrin-B promotes axon branching and ectopic connectivity. Proc. Natl. Acad. Sci. U.S.A. 116 1526215271. 10.1073/pnas.1904348116 31285321 Yu K. Wu Y. Zhang Q. Xie H. Liu G. Guo Z. (2014). Enriched environment induces angiogenesis and improves neural function outcomes in rat stroke model. J. Neurol. Sci. 347 275280. 10.1016/j.jns.2014.10.022 25455300 Yue F. Cheng Y. Breschi A. Vierstra J. Wu W. Ryba T. (2014). A comparative encyclopedia of DNA elements in the mouse genome. Nature 515 355364. 10.1038/nature13992 25409824 Zentner G. E. Tesar P. J. Scacheri P. C. (2011). Epigenetic signatures distinguish multiple classes of enhancers with distinct cellular functions. Genome Res. 21 12731283. 10.1101/gr.122382.111 21632746 Zhang T. Zhang Z. Dong Q. Xiong J. Zhu B. (2020). Histone H3K27 acetylation is dispensable for enhancer activity in mouse embryonic stem cells. Genome Biol. 21:45. 10.1186/s13059-020-01957-w 32085783 Zhang T.-Y. Keown C. L. Wen X. Li J. Vousden D. A. Anacker C. (2018). Environmental enrichment increases transcriptional and epigenetic differentiation between mouse dorsal and ventral dentate gyrus. Nat. Commun. 9:298. 10.1038/s41467-017-02748-x 29352183 Zhang Y. Kong F. Crofton E. J. Dragosljvich S. N. Sinha M. Li D. (2016). Transcriptomics of environmental enrichment reveals a role for retinoic acid signaling in addiction. Front. Mol. Neurosci. 9:119. 10.3389/fnmol.2016.00119 27899881 Zhang Y. Liu T. Meyer C. A. Eeckhoute J. Johnson D. S. Bernstein B. E. (2008). Model-based analysis of ChIP-Seq (MACS). Genome Biol. 9:R137. 10.1186/gb-2008-9-9-r137 18798982 Zhou Y. Zhou B. Pache L. Chang M. Khodabakhshi A. H. Tanaseichuk O. (2019). Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat. Commun. 10:1523. 10.1038/s41467-019-09234-6 30944313 Zhu Y. Chen Z. Zhang K. Wang M. Medovoy D. Whitaker J. W. (2016). Constructing 3D interaction maps from 1D epigenomes. Nat. Commun. 7:10812. 10.1038/ncomms10812 26960733

      www.tdx.cat/handle/10803/124485#page=1

      https://github.com/ShaluJhanwar/GEP

      http://chromosome.sdsc.edu/mouse/hi-c/download.html

      https://github.com/ophiothrix/enhancer.annotator

      http://broadinstitute.github.io/picard

      https://github.com/PavlidisLab/markerGeneProfile

      https://github.com/sespesogil/automat_chrom3D

      https://github.com/sespesogil/automat_chrom3D_colors

      ‘Oh, my dear Thomas, you haven’t heard the terrible news then?’ she said. ‘I thought you would be sure to have seen it placarded somewhere. Alice went straight to her room, and I haven’t seen her since, though I repeatedly knocked at the door, which she has locked on the inside, and I’m sure it’s most unnatural of her not to let her own mother comfort her. It all happened in a moment: I have always said those great motor-cars shouldn’t be allowed to career about the streets, especially when they are all paved with cobbles as they are at Easton Haven, which are{331} so slippery when it’s wet. He slipped, and it went over him in a moment.’ My thanks were few and awkward, for there still hung to the missive a basting thread, and it was as warm as a nestling bird. I bent low--everybody was emotional in those days--kissed the fragrant thing, thrust it into my bosom, and blushed worse than Camille. "What, the Corner House victim? Is that really a fact?" "My dear child, I don't look upon it in that light at all. The child gave our picturesque friend a certain distinction--'My husband is dead, and this is my only child,' and all that sort of thing. It pays in society." leave them on the steps of a foundling asylum in order to insure [See larger version] Interoffice guff says you're planning definite moves on your own, J. O., and against some opposition. Is the Colonel so poor or so grasping—or what? Albert could not speak, for he felt as if his brains and teeth were rattling about inside his head. The rest of[Pg 188] the family hunched together by the door, the boys gaping idiotically, the girls in tears. "Now you're married." The host was called in, and unlocked a drawer in which they were deposited. The galleyman, with visible reluctance, arrayed himself in the garments, and he was observed to shudder more than once during the investiture of the dead man's apparel. HoME香京julia种子在线播放 ENTER NUMBET 0016jiemenzi.org.cn
      fupwdo.com.cn
      www.happyfc500.com.cn
      www.hhoyqf.com.cn
      www.ffokkx.com.cn
      kguedm.com.cn
      www.lianlao.org.cn
      www.tlbqem.com.cn
      www.mtwu.com.cn
      nbfxj.net.cn
      处女被大鸡巴操 强奸乱伦小说图片 俄罗斯美女爱爱图 调教强奸学生 亚洲女的穴 夜来香图片大全 美女性强奸电影 手机版色中阁 男性人体艺术素描图 16p成人 欧美性爱360 电影区 亚洲电影 欧美电影 经典三级 偷拍自拍 动漫电影 乱伦电影 变态另类 全部电 类似狠狠鲁的网站 黑吊操白逼图片 韩国黄片种子下载 操逼逼逼逼逼 人妻 小说 p 偷拍10幼女自慰 极品淫水很多 黄色做i爱 日本女人人体电影快播看 大福国小 我爱肏屄美女 mmcrwcom 欧美多人性交图片 肥臀乱伦老头舔阴帝 d09a4343000019c5 西欧人体艺术b xxoo激情短片 未成年人的 插泰国人夭图片 第770弾み1 24p 日本美女性 交动态 eee色播 yantasythunder 操无毛少女屄 亚洲图片你懂的女人 鸡巴插姨娘 特级黄 色大片播 左耳影音先锋 冢本友希全集 日本人体艺术绿色 我爱被舔逼 内射 幼 美阴图 喷水妹子高潮迭起 和后妈 操逼 美女吞鸡巴 鸭个自慰 中国女裸名单 操逼肥臀出水换妻 色站裸体义术 中国行上的漏毛美女叫什么 亚洲妹性交图 欧美美女人裸体人艺照 成人色妹妹直播 WWW_JXCT_COM r日本女人性淫乱 大胆人艺体艺图片 女同接吻av 碰碰哥免费自拍打炮 艳舞写真duppid1 88电影街拍视频 日本自拍做爱qvod 实拍美女性爱组图 少女高清av 浙江真实乱伦迅雷 台湾luanlunxiaoshuo 洛克王国宠物排行榜 皇瑟电影yy频道大全 红孩儿连连看 阴毛摄影 大胆美女写真人体艺术摄影 和风骚三个媳妇在家做爱 性爱办公室高清 18p2p木耳 大波撸影音 大鸡巴插嫩穴小说 一剧不超两个黑人 阿姨诱惑我快播 幼香阁千叶县小学生 少女妇女被狗强奸 曰人体妹妹 十二岁性感幼女 超级乱伦qvod 97爱蜜桃ccc336 日本淫妇阴液 av海量资源999 凤凰影视成仁 辰溪四中艳照门照片 先锋模特裸体展示影片 成人片免费看 自拍百度云 肥白老妇女 女爱人体图片 妈妈一女穴 星野美夏 日本少女dachidu 妹子私处人体图片 yinmindahuitang 舔无毛逼影片快播 田莹疑的裸体照片 三级电影影音先锋02222 妻子被外国老头操 观月雏乃泥鳅 韩国成人偷拍自拍图片 强奸5一9岁幼女小说 汤姆影院av图片 妹妹人艺体图 美女大驱 和女友做爱图片自拍p 绫川まどか在线先锋 那么嫩的逼很少见了 小女孩做爱 处女好逼连连看图图 性感美女在家做爱 近距离抽插骚逼逼 黑屌肏金毛屄 日韩av美少女 看喝尿尿小姐日逼色色色网图片 欧美肛交新视频 美女吃逼逼 av30线上免费 伊人在线三级经典 新视觉影院t6090影院 最新淫色电影网址 天龙影院远古手机版 搞老太影院 插进美女的大屁股里 私人影院加盟费用 www258dd 求一部电影里面有一个二猛哥 深肛交 日本萌妹子人体艺术写真图片 插入屄眼 美女的木奶 中文字幕黄色网址影视先锋 九号女神裸 和骚人妻偷情 和潘晓婷做爱 国模大尺度蜜桃 欧美大逼50p 西西人体成人 李宗瑞继母做爱原图物处理 nianhuawang 男鸡巴的视屏 � 97免费色伦电影 好色网成人 大姨子先锋 淫荡巨乳美女教师妈妈 性nuexiaoshuo WWW36YYYCOM 长春继续给力进屋就操小女儿套干破内射对白淫荡 农夫激情社区 日韩无码bt 欧美美女手掰嫩穴图片 日本援交偷拍自拍 入侵者日本在线播放 亚洲白虎偷拍自拍 常州高见泽日屄 寂寞少妇自卫视频 人体露逼图片 多毛外国老太 变态乱轮手机在线 淫荡妈妈和儿子操逼 伦理片大奶少女 看片神器最新登入地址sqvheqi345com账号群 麻美学姐无头 圣诞老人射小妞和强奸小妞动话片 亚洲AV女老师 先锋影音欧美成人资源 33344iucoom zV天堂电影网 宾馆美女打炮视频 色五月丁香五月magnet 嫂子淫乱小说 张歆艺的老公 吃奶男人视频在线播放 欧美色图男女乱伦 avtt2014ccvom 性插色欲香影院 青青草撸死你青青草 99热久久第一时间 激情套图卡通动漫 幼女裸聊做爱口交 日本女人被强奸乱伦 草榴社区快播 2kkk正在播放兽骑 啊不要人家小穴都湿了 www猎奇影视 A片www245vvcomwwwchnrwhmhzcn 搜索宜春院av wwwsee78co 逼奶鸡巴插 好吊日AV在线视频19gancom 熟女伦乱图片小说 日本免费av无码片在线开苞 鲁大妈撸到爆 裸聊官网 德国熟女xxx 新不夜城论坛首页手机 女虐男网址 男女做爱视频华为网盘 激情午夜天亚洲色图 内裤哥mangent 吉沢明歩制服丝袜WWWHHH710COM 屌逼在线试看 人体艺体阿娇艳照 推荐一个可以免费看片的网站如果被QQ拦截请复制链接在其它浏览器打开xxxyyy5comintr2a2cb551573a2b2e 欧美360精品粉红鲍鱼 教师调教第一页 聚美屋精品图 中韩淫乱群交 俄罗斯撸撸片 把鸡巴插进小姨子的阴道 干干AV成人网 aolasoohpnbcn www84ytom 高清大量潮喷www27dyycom 宝贝开心成人 freefronvideos人母 嫩穴成人网gggg29com 逼着舅妈给我口交肛交彩漫画 欧美色色aV88wwwgangguanscom 老太太操逼自拍视频 777亚洲手机在线播放 有没有夫妻3p小说 色列漫画淫女 午间色站导航 欧美成人处女色大图 童颜巨乳亚洲综合 桃色性欲草 色眯眯射逼 无码中文字幕塞外青楼这是一个 狂日美女老师人妻 爱碰网官网 亚洲图片雅蠛蝶 快播35怎么搜片 2000XXXX电影 新谷露性家庭影院 深深候dvd播放 幼齿用英语怎么说 不雅伦理无需播放器 国外淫荡图片 国外网站幼幼嫩网址 成年人就去色色视频快播 我鲁日日鲁老老老我爱 caoshaonvbi 人体艺术avav 性感性色导航 韩国黄色哥来嫖网站 成人网站美逼 淫荡熟妇自拍 欧美色惰图片 北京空姐透明照 狼堡免费av视频 www776eom 亚洲无码av欧美天堂网男人天堂 欧美激情爆操 a片kk266co 色尼姑成人极速在线视频 国语家庭系列 蒋雯雯 越南伦理 色CC伦理影院手机版 99jbbcom 大鸡巴舅妈 国产偷拍自拍淫荡对话视频 少妇春梦射精 开心激动网 自拍偷牌成人 色桃隐 撸狗网性交视频 淫荡的三位老师 伦理电影wwwqiuxia6commqiuxia6com 怡春院分站 丝袜超短裙露脸迅雷下载 色制服电影院 97超碰好吊色男人 yy6080理论在线宅男日韩福利大全 大嫂丝袜 500人群交手机在线 5sav 偷拍熟女吧 口述我和妹妹的欲望 50p电脑版 wwwavtttcon 3p3com 伦理无码片在线看 欧美成人电影图片岛国性爱伦理电影 先锋影音AV成人欧美 我爱好色 淫电影网 WWW19MMCOM 玛丽罗斯3d同人动画h在线看 动漫女孩裸体 超级丝袜美腿乱伦 1919gogo欣赏 大色逼淫色 www就是撸 激情文学网好骚 A级黄片免费 xedd5com 国内的b是黑的 快播美国成年人片黄 av高跟丝袜视频 上原保奈美巨乳女教师在线观看 校园春色都市激情fefegancom 偷窥自拍XXOO 搜索看马操美女 人本女优视频 日日吧淫淫 人妻巨乳影院 美国女子性爱学校 大肥屁股重口味 啪啪啪啊啊啊不要 操碰 japanfreevideoshome国产 亚州淫荡老熟女人体 伦奸毛片免费在线看 天天影视se 樱桃做爱视频 亚卅av在线视频 x奸小说下载 亚洲色图图片在线 217av天堂网 东方在线撸撸-百度 幼幼丝袜集 灰姑娘的姐姐 青青草在线视频观看对华 86papa路con 亚洲1AV 综合图片2区亚洲 美国美女大逼电影 010插插av成人网站 www色comwww821kxwcom 播乐子成人网免费视频在线观看 大炮撸在线影院 ,www4KkKcom 野花鲁最近30部 wwwCC213wapwww2233ww2download 三客优最新地址 母亲让儿子爽的无码视频 全国黄色片子 欧美色图美国十次 超碰在线直播 性感妖娆操 亚洲肉感熟女色图 a片A毛片管看视频 8vaa褋芯屑 333kk 川岛和津实视频 在线母子乱伦对白 妹妹肥逼五月 亚洲美女自拍 老婆在我面前小说 韩国空姐堪比情趣内衣 干小姐综合 淫妻色五月 添骚穴 WM62COM 23456影视播放器 成人午夜剧场 尼姑福利网 AV区亚洲AV欧美AV512qucomwwwc5508com 经典欧美骚妇 震动棒露出 日韩丝袜美臀巨乳在线 av无限吧看 就去干少妇 色艺无间正面是哪集 校园春色我和老师做爱 漫画夜色 天海丽白色吊带 黄色淫荡性虐小说 午夜高清播放器 文20岁女性荫道口图片 热国产热无码热有码 2015小明发布看看算你色 百度云播影视 美女肏屄屄乱轮小说 家族舔阴AV影片 邪恶在线av有码 父女之交 关于处女破处的三级片 极品护士91在线 欧美虐待女人视频的网站 享受老太太的丝袜 aaazhibuo 8dfvodcom成人 真实自拍足交 群交男女猛插逼 妓女爱爱动态 lin35com是什么网站 abp159 亚洲色图偷拍自拍乱伦熟女抠逼自慰 朝国三级篇 淫三国幻想 免费的av小电影网站 日本阿v视频免费按摩师 av750c0m 黄色片操一下 巨乳少女车震在线观看 操逼 免费 囗述情感一乱伦岳母和女婿 WWW_FAMITSU_COM 偷拍中国少妇在公车被操视频 花也真衣论理电影 大鸡鸡插p洞 新片欧美十八岁美少 进击的巨人神thunderftp 西方美女15p 深圳哪里易找到老女人玩视频 在线成人有声小说 365rrr 女尿图片 我和淫荡的小姨做爱 � 做爱技术体照 淫妇性爱 大学生私拍b 第四射狠狠射小说 色中色成人av社区 和小姨子乱伦肛交 wwwppp62com 俄罗斯巨乳人体艺术 骚逼阿娇 汤芳人体图片大胆 大胆人体艺术bb私处 性感大胸骚货 哪个网站幼女的片多 日本美女本子把 色 五月天 婷婷 快播 美女 美穴艺术 色百合电影导航 大鸡巴用力 孙悟空操美少女战士 狠狠撸美女手掰穴图片 古代女子与兽类交 沙耶香套图 激情成人网区 暴风影音av播放 动漫女孩怎么插第3个 mmmpp44 黑木麻衣无码ed2k 淫荡学姐少妇 乱伦操少女屄 高中性爱故事 骚妹妹爱爱图网 韩国模特剪长发 大鸡巴把我逼日了 中国张柏芝做爱片中国张柏芝做爱片中国张柏芝做爱片中国张柏芝做爱片中国张柏芝做爱片 大胆女人下体艺术图片 789sss 影音先锋在线国内情侣野外性事自拍普通话对白 群撸图库 闪现君打阿乐 ady 小说 插入表妹嫩穴小说 推荐成人资源 网络播放器 成人台 149大胆人体艺术 大屌图片 骚美女成人av 春暖花开春色性吧 女亭婷五月 我上了同桌的姐姐 恋夜秀场主播自慰视频 yzppp 屄茎 操屄女图 美女鲍鱼大特写 淫乱的日本人妻山口玲子 偷拍射精图 性感美女人体艺木图片 种马小说完本 免费电影院 骑士福利导航导航网站 骚老婆足交 国产性爱一级电影 欧美免费成人花花性都 欧美大肥妞性爱视频 家庭乱伦网站快播 偷拍自拍国产毛片 金发美女也用大吊来开包 缔D杏那 yentiyishu人体艺术ytys WWWUUKKMCOM 女人露奶 � 苍井空露逼 老荡妇高跟丝袜足交 偷偷和女友的朋友做爱迅雷 做爱七十二尺 朱丹人体合成 麻腾由纪妃 帅哥撸播种子图 鸡巴插逼动态图片 羙国十次啦中文 WWW137AVCOM 神斗片欧美版华语 有气质女人人休艺术 由美老师放屁电影 欧美女人肉肏图片 白虎种子快播 国产自拍90后女孩 美女在床上疯狂嫩b 饭岛爱最后之作 幼幼强奸摸奶 色97成人动漫 两性性爱打鸡巴插逼 新视觉影院4080青苹果影院 嗯好爽插死我了 阴口艺术照 李宗瑞电影qvod38 爆操舅母 亚洲色图七七影院 被大鸡巴操菊花 怡红院肿么了 成人极品影院删除 欧美性爱大图色图强奸乱 欧美女子与狗随便性交 苍井空的bt种子无码 熟女乱伦长篇小说 大色虫 兽交幼女影音先锋播放 44aad be0ca93900121f9b 先锋天耗ばさ无码 欧毛毛女三级黄色片图 干女人黑木耳照 日本美女少妇嫩逼人体艺术 sesechangchang 色屄屄网 久久撸app下载 色图色噜 美女鸡巴大奶 好吊日在线视频在线观看 透明丝袜脚偷拍自拍 中山怡红院菜单 wcwwwcom下载 骑嫂子 亚洲大色妣 成人故事365ahnet 丝袜家庭教mp4 幼交肛交 妹妹撸撸大妈 日本毛爽 caoprom超碰在email 关于中国古代偷窥的黄片 第一会所老熟女下载 wwwhuangsecome 狼人干综合新地址HD播放 变态儿子强奸乱伦图 强奸电影名字 2wwwer37com 日本毛片基地一亚洲AVmzddcxcn 暗黑圣经仙桃影院 37tpcocn 持月真由xfplay 好吊日在线视频三级网 我爱背入李丽珍 电影师傅床戏在线观看 96插妹妹sexsex88com 豪放家庭在线播放 桃花宝典极夜著豆瓜网 安卓系统播放神器 美美网丝袜诱惑 人人干全免费视频xulawyercn av无插件一本道 全国色五月 操逼电影小说网 good在线wwwyuyuelvcom www18avmmd 撸波波影视无插件 伊人幼女成人电影 会看射的图片 小明插看看 全裸美女扒开粉嫩b 国人自拍性交网站 萝莉白丝足交本子 七草ちとせ巨乳视频 摇摇晃晃的成人电影 兰桂坊成社人区小说www68kqcom 舔阴论坛 久撸客一撸客色国内外成人激情在线 明星门 欧美大胆嫩肉穴爽大片 www牛逼插 性吧星云 少妇性奴的屁眼 人体艺术大胆mscbaidu1imgcn 最新久久色色成人版 l女同在线 小泽玛利亚高潮图片搜索 女性裸b图 肛交bt种子 最热门有声小说 人间添春色 春色猜谜字 樱井莉亚钢管舞视频 小泽玛利亚直美6p 能用的h网 还能看的h网 bl动漫h网 开心五月激 东京热401 男色女色第四色酒色网 怎么下载黄色小说 黄色小说小栽 和谐图城 乐乐影院 色哥导航 特色导航 依依社区 爱窝窝在线 色狼谷成人 91porn 包要你射电影 色色3A丝袜 丝袜妹妹淫网 爱色导航(荐) 好男人激情影院 坏哥哥 第七色 色久久 人格分裂 急先锋 撸撸射中文网 第一会所综合社区 91影院老师机 东方成人激情 怼莪影院吹潮 老鸭窝伊人无码不卡无码一本道 av女柳晶电影 91天生爱风流作品 深爱激情小说私房婷婷网 擼奶av 567pao 里番3d一家人野外 上原在线电影 水岛津实透明丝袜 1314酒色 网旧网俺也去 0855影院 在线无码私人影院 搜索 国产自拍 神马dy888午夜伦理达达兔 农民工黄晓婷 日韩裸体黑丝御姐 屈臣氏的燕窝面膜怎么样つぼみ晶エリーの早漏チ○ポ强化合宿 老熟女人性视频 影音先锋 三上悠亚ol 妹妹影院福利片 hhhhhhhhsxo 午夜天堂热的国产 强奸剧场 全裸香蕉视频无码 亚欧伦理视频 秋霞为什么给封了 日本在线视频空天使 日韩成人aⅴ在线 日本日屌日屄导航视频 在线福利视频 日本推油无码av magnet 在线免费视频 樱井梨吮东 日本一本道在线无码DVD 日本性感诱惑美女做爱阴道流水视频 日本一级av 汤姆avtom在线视频 台湾佬中文娱乐线20 阿v播播下载 橙色影院 奴隶少女护士cg视频 汤姆在线影院无码 偷拍宾馆 业面紧急生级访问 色和尚有线 厕所偷拍一族 av女l 公交色狼优酷视频 裸体视频AV 人与兽肉肉网 董美香ol 花井美纱链接 magnet 西瓜影音 亚洲 自拍 日韩女优欧美激情偷拍自拍 亚洲成年人免费视频 荷兰免费成人电影 深喉呕吐XXⅩX 操石榴在线视频 天天色成人免费视频 314hu四虎 涩久免费视频在线观看 成人电影迅雷下载 能看见整个奶子的香蕉影院 水菜丽百度影音 gwaz079百度云 噜死你们资源站 主播走光视频合集迅雷下载 thumbzilla jappen 精品Av 古川伊织star598在线 假面女皇vip在线视频播放 国产自拍迷情校园 啪啪啪公寓漫画 日本阿AV 黄色手机电影 欧美在线Av影院 华裔电击女神91在线 亚洲欧美专区 1日本1000部免费视频 开放90后 波多野结衣 东方 影院av 页面升级紧急访问每天正常更新 4438Xchengeren 老炮色 a k福利电影 色欲影视色天天视频 高老庄aV 259LUXU-683 magnet 手机在线电影 国产区 欧美激情人人操网 国产 偷拍 直播 日韩 国内外激情在线视频网给 站长统计一本道人妻 光棍影院被封 紫竹铃取汁 ftp 狂插空姐嫩 xfplay 丈夫面前 穿靴子伪街 XXOO视频在线免费 大香蕉道久在线播放 电棒漏电嗨过头 充气娃能看下毛和洞吗 夫妻牲交 福利云点墦 yukun瑟妃 疯狂交换女友 国产自拍26页 腐女资源 百度云 日本DVD高清无码视频 偷拍,自拍AV伦理电影 A片小视频福利站。 大奶肥婆自拍偷拍图片 交配伊甸园 超碰在线视频自拍偷拍国产 小热巴91大神 rctd 045 类似于A片 超美大奶大学生美女直播被男友操 男友问 你的衣服怎么脱掉的 亚洲女与黑人群交视频一 在线黄涩 木内美保步兵番号 鸡巴插入欧美美女的b舒服 激情在线国产自拍日韩欧美 国语福利小视频在线观看 作爱小视颍 潮喷合集丝袜无码mp4 做爱的无码高清视频 牛牛精品 伊aⅤ在线观看 savk12 哥哥搞在线播放 在线电一本道影 一级谍片 250pp亚洲情艺中心,88 欧美一本道九色在线一 wwwseavbacom色av吧 cos美女在线 欧美17,18ⅹⅹⅹ视频 自拍嫩逼 小电影在线观看网站 筱田优 贼 水电工 5358x视频 日本69式视频有码 b雪福利导航 韩国女主播19tvclub在线 操逼清晰视频 丝袜美女国产视频网址导航 水菜丽颜射房间 台湾妹中文娱乐网 风吟岛视频 口交 伦理 日本熟妇色五十路免费视频 A级片互舔 川村真矢Av在线观看 亚洲日韩av 色和尚国产自拍 sea8 mp4 aV天堂2018手机在线 免费版国产偷拍a在线播放 狠狠 婷婷 丁香 小视频福利在线观看平台 思妍白衣小仙女被邻居强上 萝莉自拍有水 4484新视觉 永久发布页 977成人影视在线观看 小清新影院在线观 小鸟酱后丝后入百度云 旋风魅影四级 香蕉影院小黄片免费看 性爱直播磁力链接 小骚逼第一色影院 性交流的视频 小雪小视频bd 小视频TV禁看视频 迷奸AV在线看 nba直播 任你在干线 汤姆影院在线视频国产 624u在线播放 成人 一级a做爰片就在线看狐狸视频 小香蕉AV视频 www182、com 腿模简小育 学生做爱视频 秘密搜查官 快播 成人福利网午夜 一级黄色夫妻录像片 直接看的gav久久播放器 国产自拍400首页 sm老爹影院 谁知道隔壁老王网址在线 综合网 123西瓜影音 米奇丁香 人人澡人人漠大学生 色久悠 夜色视频你今天寂寞了吗? 菲菲影视城美国 被抄的影院 变态另类 欧美 成人 国产偷拍自拍在线小说 不用下载安装就能看的吃男人鸡巴视频 插屄视频 大贯杏里播放 wwwhhh50 233若菜奈央 伦理片天海翼秘密搜查官 大香蕉在线万色屋视频 那种漫画小说你懂的 祥仔电影合集一区 那里可以看澳门皇冠酒店a片 色自啪 亚洲aV电影天堂 谷露影院ar toupaizaixian sexbj。com 毕业生 zaixian mianfei 朝桐光视频 成人短视频在线直接观看 陈美霖 沈阳音乐学院 导航女 www26yjjcom 1大尺度视频 开平虐女视频 菅野雪松协和影视在线视频 华人play在线视频bbb 鸡吧操屄视频 多啪啪免费视频 悠草影院 金兰策划网 (969) 橘佑金短视频 国内一极刺激自拍片 日本制服番号大全magnet 成人动漫母系 电脑怎么清理内存 黄色福利1000 dy88午夜 偷拍中学生洗澡磁力链接 花椒相机福利美女视频 站长推荐磁力下载 mp4 三洞轮流插视频 玉兔miki热舞视频 夜生活小视频 爆乳人妖小视频 国内网红主播自拍福利迅雷下载 不用app的裸裸体美女操逼视频 变态SM影片在线观看 草溜影院元气吧 - 百度 - 百度 波推全套视频 国产双飞集合ftp 日本在线AV网 笔国毛片 神马影院女主播是我的邻居 影音资源 激情乱伦电影 799pao 亚洲第一色第一影院 av视频大香蕉 老梁故事汇希斯莱杰 水中人体磁力链接 下载 大香蕉黄片免费看 济南谭崔 避开屏蔽的岛a片 草破福利 要看大鸡巴操小骚逼的人的视频 黑丝少妇影音先锋 欧美巨乳熟女磁力链接 美国黄网站色大全 伦蕉在线久播 极品女厕沟 激情五月bd韩国电影 混血美女自摸和男友激情啪啪自拍诱人呻吟福利视频 人人摸人人妻做人人看 44kknn 娸娸原网 伊人欧美 恋夜影院视频列表安卓青青 57k影院 如果电话亭 avi 插爆骚女精品自拍 青青草在线免费视频1769TV 令人惹火的邻家美眉 影音先锋 真人妹子被捅动态图 男人女人做完爱视频15 表姐合租两人共处一室晚上她竟爬上了我的床 性爱教学视频 北条麻妃bd在线播放版 国产老师和师生 magnet wwwcctv1024 女神自慰 ftp 女同性恋做激情视频 欧美大胆露阴视频 欧美无码影视 好女色在线观看 后入肥臀18p 百度影视屏福利 厕所超碰视频 强奸mp magnet 欧美妹aⅴ免费线上看 2016年妞干网视频 5手机在线福利 超在线最视频 800av:cOm magnet 欧美性爱免播放器在线播放 91大款肥汤的性感美乳90后邻家美眉趴着窗台后入啪啪 秋霞日本毛片网站 cheng ren 在线视频 上原亚衣肛门无码解禁影音先锋 美脚家庭教师在线播放 尤酷伦理片 熟女性生活视频在线观看 欧美av在线播放喷潮 194avav 凤凰AV成人 - 百度 kbb9999 AV片AV在线AV无码 爱爱视频高清免费观看 黄色男女操b视频 观看 18AV清纯视频在线播放平台 成人性爱视频久久操 女性真人生殖系统双性人视频 下身插入b射精视频 明星潜规测视频 mp4 免賛a片直播绪 国内 自己 偷拍 在线 国内真实偷拍 手机在线 国产主播户外勾在线 三桥杏奈高清无码迅雷下载 2五福电影院凸凹频频 男主拿鱼打女主,高宝宝 色哥午夜影院 川村まや痴汉 草溜影院费全过程免费 淫小弟影院在线视频 laohantuiche 啪啪啪喷潮XXOO视频 青娱乐成人国产 蓝沢润 一本道 亚洲青涩中文欧美 神马影院线理论 米娅卡莉法的av 在线福利65535 欧美粉色在线 欧美性受群交视频1在线播放 极品喷奶熟妇在线播放 变态另类无码福利影院92 天津小姐被偷拍 磁力下载 台湾三级电髟全部 丝袜美腿偷拍自拍 偷拍女生性行为图 妻子的乱伦 白虎少妇 肏婶骚屄 外国大妈会阴照片 美少女操屄图片 妹妹自慰11p 操老熟女的b 361美女人体 360电影院樱桃 爱色妹妹亚洲色图 性交卖淫姿势高清图片一级 欧美一黑对二白 大色网无毛一线天 射小妹网站 寂寞穴 西西人体模特苍井空 操的大白逼吧 骚穴让我操 拉好友干女朋友3p