Comparative chromatin accessibility upon BDNF-induced neuronal activity delineates neuronal regulatory elements

Neuronal activity induced by brain-derived neurotrophic factor (BDNF) triggers gene expression, which is crucial for neuronal survival, differentiation, synaptic plasticity, memory formation, and neurocognitive health. However, its role in chromatin regulation is unclear. Here, using temporal profiling of chromatin accessibility and transcription in mouse primary cortical neurons upon either BDNF stimulation or depolarization (KCl), we identify features that define BDNF-specific chromatin-to-gene expression programs. Enhancer activation is an early event in the regulatory control of BDNF-treated neurons, where the bZIP motif-binding Fos protein pioneered chromatin opening and cooperated with co-regulatory transcription factors (Homeobox, EGRs, and CTCF) to induce transcription. Deleting cis-regulatory sequences decreased BDNF-mediated Arc expression, a regulator of synaptic plasticity. BDNF-induced accessible regions are linked to preferential exon usage by neurodevelopmental disorder-related genes and heritability of neuronal complex traits, which were validated in human iPSC-derived neurons. Thus, we provide a comprehensive view of BDNF-mediated genome regulatory features using comparative genomic approaches to dissect mammalian neuronal activity.

[1]  Manolis Kellis,et al.  Regulatory genomic circuitry of human disease loci by integrative epigenomics , 2021, Nature.

[2]  Panagiotis K. Papasaikas,et al.  A Unique Bipartite Polycomb Signature Regulates Stimulus-Response Transcription during Development , 2021, Nature Genetics.

[3]  Fidencio J. Neri,et al.  Global reference mapping of human transcription factor footprints , 2020, Nature.

[4]  Jennifer E. Phillips-Cremins,et al.  Three-dimensional genome restructuring across timescales of activity-induced neuronal gene expression , 2020, Nature Neuroscience.

[5]  Judith B. Zaugg,et al.  Mechanistic insights into transcription factor cooperativity and its impact on protein-phenotype interactions , 2020, Nature Communications.

[6]  Anne-Laure Hemonnot,et al.  Microglia in Alzheimer Disease: Well-Known Targets and New Opportunities , 2019, Front. Aging Neurosci..

[7]  S. Carr,et al.  Trio Haploinsufficiency Causes Neurodevelopmental Disease–Associated Deficits , 2019, Cell reports.

[8]  Sophie Lèbre,et al.  Probing transcription factor combinatorics in different promoter classes and in enhancers , 2019, BMC Genomics.

[9]  Helen E. Parkinson,et al.  The NHGRI-EBI GWAS Catalog of published genome-wide association studies, targeted arrays and summary statistics 2019 , 2018, Nucleic Acids Res..

[10]  Michael E. Greenberg,et al.  Activity-Regulated Transcription: Bridging the Gap between Neural Activity and Behavior , 2018, Neuron.

[11]  A. McCallion,et al.  Leveraging mouse chromatin data for heritability enrichment informs common disease architecture and reveals cortical layer contributions to schizophrenia , 2018, bioRxiv.

[12]  Eunhee Kim,et al.  Combined adult neurogenesis and BDNF mimic exercise effects on cognition in an Alzheimer’s mouse model , 2018, Science.

[13]  R. Dierckx,et al.  Brain-Derived Neurotrophic Factor in Brain Disorders: Focus on Neuroinflammation , 2018, Molecular Neurobiology.

[14]  Michael A. Beer,et al.  Parkinson-associated SNCA enhancer variants revealed by open chromatin in mouse dopamine neurons , 2018, bioRxiv.

[15]  Serena M. Dudek,et al.  Different Neuronal Activity Patterns Induce Different Gene Expression Programs , 2018, Neuron.

[16]  I. Weissman,et al.  A Roadmap for Human Liver Differentiation from Pluripotent Stem Cells , 2018, Cell reports.

[17]  D. Geschwind,et al.  The Dynamic Landscape of Open Chromatin during Human Cortical Neurogenesis , 2018, Cell.

[18]  Judith B. Zaugg,et al.  CTCF-Mediated Chromatin Loops between Promoter and Gene Body Regulate Alternative Splicing across Individuals. , 2017, Cell systems.

[19]  M. Greenberg,et al.  AP-1 Transcription Factors and the BAF Complex Mediate Signal-Dependent Enhancer Selection. , 2017, Molecular cell.

[20]  A. Tanay,et al.  Multiscale 3D Genome Rewiring during Mouse Neural Development , 2017, Cell.

[21]  M. Bulyk,et al.  Identification of Human Lineage-Specific Transcriptional Coregulators Enabled by a Glossary of Binding Modules and Tunable Genomic Backgrounds. , 2017, Cell systems.

[22]  Keji Zhao,et al.  CTCF-Mediated Enhancer-Promoter Interaction Is a Critical Regulator of Cell-to-Cell Variation of Gene Expression. , 2017, Molecular cell.

[23]  V. Katritch,et al.  An autism spectrum disorder-related de novo mutation hotspot discovered in the GEF1 domain of Trio , 2017, Nature Communications.

[24]  A. Smit,et al.  Tomosyn associates with secretory vesicles in neurons through its N- and C-terminal domains , 2017, PloS one.

[25]  G. Ming,et al.  Neuronal activity modifies the chromatin accessibility landscape in the adult brain , 2017, Nature Neuroscience.

[26]  H. Bading,et al.  Networks of Cultured iPSC-Derived Neurons Reveal the Human Synaptic Activity-Regulated Adaptive Gene Program , 2017, Cell reports.

[27]  H. Kaphzan,et al.  Neuronal CTCF Is Necessary for Basal and Experience-Dependent Gene Regulation, Memory Formation, and Genomic Structure of BDNF and Arc. , 2016, Cell reports.

[28]  Athar N. Malik,et al.  Evolution of Osteocrin as an activity-regulated factor in the primate brain , 2016, Nature.

[29]  Kyo Seon Hwang,et al.  Ultra-sensitive detection of brain-derived neurotrophic factor (BDNF) in the brain of freely moving mice using an interdigitated microelectrode (IME) biosensor , 2016, Scientific Reports.

[30]  D. Baralle,et al.  Mutations specific to the Rac-GEF domain of TRIO cause intellectual disability and microcephaly , 2016, Journal of Medical Genetics.

[31]  L. Monteggia,et al.  BDNF – a key transducer of antidepressant effects , 2016, Neuropharmacology.

[32]  Minghui Wang,et al.  Efficient Test and Visualization of Multi-Set Intersections , 2015, Scientific Reports.

[33]  A. Jolma,et al.  DNA-dependent formation of transcription factor pairs alters their binding specificity , 2015, Nature.

[34]  Gabor T. Marth,et al.  A global reference for human genetic variation , 2015, Nature.

[35]  C. Duarte,et al.  Regulation of hippocampal synaptic plasticity by BDNF , 2015, Brain Research.

[36]  Yakir A Reshef,et al.  Partitioning heritability by functional annotation using genome-wide association summary statistics , 2015, Nature Genetics.

[37]  Howard Y. Chang,et al.  ATAC‐seq: A Method for Assaying Chromatin Accessibility Genome‐Wide , 2015, Current protocols in molecular biology.

[38]  W. Huber,et al.  Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2 , 2014, Genome Biology.

[39]  Thomas Vierbuchen,et al.  Genome-wide identification and characterization of functional neuronal activity–dependent enhancers , 2014, Nature Neuroscience.

[40]  Kate B. Cook,et al.  Determination and Inference of Eukaryotic Transcription Factor Sequence Specificity , 2014, Cell.

[41]  M. Webster,et al.  Decreased BDNF and TrkB mRNA expression in multiple cortical areas of patients with schizophrenia and mood disorders , 2014, Translational Psychiatry.

[42]  C. Bramham,et al.  BDNF mechanisms in late LTP formation: A synthesis and breakdown , 2014, Neuropharmacology.

[43]  Sarah Seton-Rogers,et al.  Therapeutics: Delving deeper into resistance , 2013, Nature Reviews Cancer.

[44]  David A. Scott,et al.  Genome engineering using the CRISPR-Cas9 system , 2013, Nature Protocols.

[45]  Praveen Sethupathy,et al.  Promoter-proximal CCCTC-factor binding is associated with an increase in the transcriptional pausing index , 2013, Bioinform..

[46]  Cole Trapnell,et al.  TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions , 2013, Genome Biology.

[47]  Atina G. Coté,et al.  Evaluation of methods for modeling transcription factor sequence specificity , 2013, Nature Biotechnology.

[48]  Shane J. Neph,et al.  Systematic Localization of Common Disease-Associated Variation in Regulatory DNA , 2012, Science.

[49]  J. Doudna,et al.  A Programmable Dual-RNA–Guided DNA Endonuclease in Adaptive Bacterial Immunity , 2012, Science.

[50]  Eric R Kandel,et al.  The molecular biology of memory: cAMP, PKA, CRE, CREB-1, CREB-2, and CPEB , 2012, Molecular Brain.

[51]  W. Huber,et al.  Detecting differential usage of exons from RNA-seq data , 2012, Genome research.

[52]  Steven L Salzberg,et al.  Fast gapped-read alignment with Bowtie 2 , 2012, Nature Methods.

[53]  D. Leprince,et al.  Hypermethylated in Cancer 1 (HIC1) Recruits Polycomb Repressive Complex 2 (PRC2) to a Subset of Its Target Genes through Interaction with Human Polycomb-like (hPCL) Proteins* , 2012, The Journal of Biological Chemistry.

[54]  Manolis Kellis,et al.  ChromHMM: automating chromatin-state discovery and characterization , 2012, Nature Methods.

[55]  R. Sandberg,et al.  CTCF-promoted RNA polymerase II pausing links DNA methylation to splicing , 2011, Nature.

[56]  Myong-Hee Sung,et al.  Transcription factor AP1 potentiates chromatin accessibility and glucocorticoid receptor binding. , 2011, Molecular cell.

[57]  William Stafford Noble,et al.  FIMO: scanning for occurrences of a given motif , 2011, Bioinform..

[58]  C. Glass,et al.  Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. , 2010, Molecular cell.

[59]  Cory Y. McLean,et al.  GREAT improves functional interpretation of cis-regulatory regions , 2010, Nature Biotechnology.

[60]  G. Kreiman,et al.  Widespread transcription at neuronal activity-regulated enhancers , 2010, Nature.

[61]  Y. Loh,et al.  Carboxypeptidase E knockout mice exhibit abnormal dendritic arborization and spine morphology in central nervous system neurons , 2010, Journal of neuroscience research.

[62]  L. Minichiello TrkB signalling pathways in LTP and learning , 2009, Nature Reviews Neuroscience.

[63]  V. Corces,et al.  CTCF: Master Weaver of the Genome , 2009, Cell.

[64]  D. Geschwind,et al.  Neuroprotective effects of brain-derived neurotrophic factor in rodent and primate models of Alzheimer's disease , 2009, Nature Medicine.

[65]  Michael E. Greenberg,et al.  From Synapse to Nucleus: Calcium-Dependent Gene Transcription in the Control of Synapse Development and Function , 2008, Neuron.

[66]  Clifford A. Meyer,et al.  Model-based Analysis of ChIP-Seq (MACS) , 2008, Genome Biology.

[67]  Steven W. Flavell,et al.  Signaling mechanisms linking neuronal activity to gene expression and plasticity of the nervous system. , 2008, Annual review of neuroscience.

[68]  M. P. Howell,et al.  The immediate early gene early growth response gene 3 mediates adaptation to stress and novelty , 2007, Neuroscience.

[69]  A. Whitmarsh Regulation of gene transcription by mitogen-activated protein kinase signaling pathways. , 2007, Biochimica et biophysica acta.

[70]  Y. Barde,et al.  Generation of a defined and uniform population of CNS progenitors and neurons from mouse embryonic stem cells , 2007, Nature Protocols.

[71]  Claus Nerlov,et al.  Neurotrophin/Trk receptor signaling mediates C/EBPα, -β and NeuroD recruitment to immediate-early gene promoters in neuronal cells and requires C/EBPs to induce immediate-early gene transcription , 2007, Neural Development.

[72]  T. Bliss,et al.  Arc/Arg3.1 Is Essential for the Consolidation of Synaptic Plasticity and Memories , 2006, Neuron.

[73]  Anastassios V. Tzingounis,et al.  Arc/Arg3.1: Linking Gene Expression to Synaptic Plasticity and Memory , 2006, Neuron.

[74]  Thomas Lengauer,et al.  Improved scoring of functional groups from gene expression data by decorrelating GO graph structure , 2006, Bioinform..

[75]  A. Beckmann,et al.  Egr transcription factors in the nervous system , 1997, Neurochemistry International.