Transcriptional and functional consequences of alterations to MEF2C and its topological organization in neuronal models

Point mutations and structural variants directly disrupting the coding sequence of MEF2C have been associated with a spectrum of neurodevelopmental disorders (NDDs), while recent studies have also implicated altered noncoding regulation of MEF2C expression in NDDs. However, the impact of haploinsufficiency of MEF2C on neurodevelopmental pathways and synaptic processes is not well understood, nor are the complex mechanisms that govern regulation of MEF2C. To explore the transcriptional and functional changes associated with coding and noncoding structural variants, we generated an allelic series of 204 isogenic iPSC-derived neuronal cell lines harboring CRISPR-engineered mutations that directly delete predominant isoforms of MEF2C, as well as deletions to the boundaries of topologically associating domains (TADs) and chromatin loops encompassing MEF2C. We then performed systematic profiling of mutation-specific alterations to transcriptional signatures, regulatory interactions, chromatin contacts, and electrophysiological effects. Our analyses reveal that direct deletion of MEF2C causes differential expression of genes enriched for neurodevelopmental and synaptic-associated pathways, accompanied by a significant reduction in synaptic firing and synchrony in neurons. By contrast, we observe robust buffering against MEF2C regulatory disruption upon deletion of a distal 5q14.3 TAD and loop boundary; however, homozygous loss of proximal loop boundary resulted in significant down-regulation of MEF2C expression and significantly reduced electrophysiological activity that was comparable to direct MEF2C disruption. Collectively, our findings demonstrate the functional impact of MEF2C haploinsufficiency in human-derived neural models and highlight the complex interactions of gene regulation and chromatin topology that challenge a priori regulatory predictions of structural variant disruption to three-dimensional genome organization.

[1]  Ryan L. Collins,et al.  Rare coding variation provides insight into the genetic architecture and phenotypic context of autism , 2022, Nature Genetics.

[2]  Ryan L. Collins,et al.  Rare coding variation illuminates the allelic architecture, risk genes, cellular expression patterns, and phenotypic context of autism , 2021, medRxiv.

[3]  Jacob C. Ulirsch,et al.  Direct characterization of cis-regulatory elements and functional dissection of complex genetic associations using HCR-FlowFISH , 2021, Nature Genetics.

[4]  Evin M. Padhi,et al.  Coding and noncoding variants in EBF3 are involved in HADDS and simplex autism , 2021, Human genomics.

[5]  L. Pasquali,et al.  UMI4Cats: an R package to analyze chromatin contact profiles obtained by UMI-4C , 2021, Bioinform..

[6]  S. Mundlos,et al.  Diagnostic Yield of Whole Genome Sequencing After Nondiagnostic Exome Sequencing or Gene Panel in Developmental and Epileptic Encephalopathies , 2021, Neurology.

[7]  J. de Ridder,et al.  svMIL: predicting the pathogenic effect of TAD boundary-disrupting somatic structural variants through multiple instance learning. , 2020, Bioinformatics.

[8]  David R. Kelley,et al.  Predicting 3D genome folding from DNA sequence with Akita , 2020, Nature Methods.

[9]  E. Tsvetkov,et al.  MEF2C Hypofunction in Neuronal and Neuroimmune Populations Produces MEF2C Haploinsufficiency Syndrome–like Behaviors in Mice , 2020, Biological Psychiatry.

[10]  B. Menten,et al.  A neuronal enhancer network upstream of MEF2C is compromised in patients with Rett like characteristics. , 2020, Human molecular genetics.

[11]  Ryan L. Collins,et al.  The mutational constraint spectrum quantified from variation in 141,456 humans , 2020, Nature.

[12]  B. Ng,et al.  Disruption of chromatin organisation causes MEF2C gene overexpression in intellectual disability: a case report , 2019, BMC Medical Genomics.

[13]  M. T. B. Geller,et al.  Molecular , 2019, Modern Pathology.

[14]  S. Mundlos,et al.  Functional dissection of the Sox9–Kcnj2 locus identifies nonessential and instructive roles of TAD architecture , 2019, Nature Genetics.

[15]  Hagen U. Tilgner,et al.  SynGO: An Evidence-Based, Expert-Curated Knowledge Base for the Synapse , 2019, Neuron.

[16]  E. Furlong,et al.  Highly rearranged chromosomes reveal uncoupling between genome topology and gene expression , 2019, Nature Genetics.

[17]  Miguel A. Soler,et al.  AMPA receptor GluA2 subunit defects are a cause of neurodevelopmental disorders , 2019, Nature Communications.

[18]  D. Goldstein,et al.  Noncoding Deletions Expose a Novel Gene Critical for Intestinal Function , 2019, Nature.

[19]  J. Baptista,et al.  Modeling the Pathological Long-Range Regulatory Effects of Human Structural Variation with Patient-Specific hiPSCs. , 2019, Cell stem cell.

[20]  S. Weiss,et al.  MEF2C-related epilepsy: Delineating the phenotypic spectrum from a novel mutation and literature review , 2019, Seizure.

[21]  R. Touraine,et al.  Whole genome paired-end sequencing elucidates functional and phenotypic consequences of balanced chromosomal rearrangement in patients with developmental disorders , 2019, Journal of Medical Genetics.

[22]  Fereydoun Hormozdiari,et al.  TAD fusion score: discovery and ranking the contribution of deletions to genome structure , 2019, Genome Biology.

[23]  Ilya M. Flyamer,et al.  Developmentally regulated Shh expression is robust to TAD perturbations , 2019, Development.

[24]  B. Black,et al.  Cardiovascular development and survival require Mef2c function in the myocardial but not the endothelial lineage. , 2019, Developmental biology.

[25]  Jan Gorodkin,et al.  Cytoscape stringApp: Network analysis and visualization of proteomics data , 2018, bioRxiv.

[26]  Stephan J Sanders,et al.  Integrative functional genomic analysis of human brain development and neuropsychiatric risks , 2018, Science.

[27]  Ryan L. Collins,et al.  Genome-wide de novo risk score implicates promoter variation in autism spectrum disorder , 2018, Science.

[28]  X. Bao,et al.  Novel MEF2C point mutations in Chinese patients with Rett (−like) syndrome or non-syndromic intellectual disability: insights into genotype-phenotype correlation , 2018, BMC medical genetics.

[29]  Ryan L. Collins,et al.  An analytical framework for whole-genome sequence association studies and its implications for autism spectrum disorder , 2018, Nature Genetics.

[30]  Ryan L. Collins,et al.  Dissecting the Causal Mechanism of X-Linked Dystonia-Parkinsonism by Integrating Genome and Transcriptome Assembly , 2018, Cell.

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

[32]  Sören Müller,et al.  Single-cell Co-expression Subnetwork Analysis , 2017, Scientific Reports.

[33]  François Dubeau,et al.  High Rate of Recurrent De Novo Mutations in Developmental and Epileptic Encephalopathies. , 2017, American journal of human genetics.

[34]  Erez Lieberman Aiden,et al.  Cohesin Loss Eliminates All Loop Domains , 2017, Cell.

[35]  Nuno A. Fonseca,et al.  Two independent modes of chromatin organization revealed by cohesin removal , 2017, Nature.

[36]  E. Olson,et al.  Notch Inhibition Enhances Cardiac Reprogramming by Increasing MEF2C Transcriptional Activity , 2017, Stem cell reports.

[37]  Edwin Cuppen,et al.  The genomic landscape of balanced cytogenetic abnormalities associated with human congenital anomalies , 2016, Nature Genetics.

[38]  Damian Szklarczyk,et al.  The STRING database in 2017: quality-controlled protein–protein association networks, made broadly accessible , 2016, Nucleic Acids Res..

[39]  S. Mundlos,et al.  Formation of new chromatin domains determines pathogenicity of genomic duplications , 2016, Nature.

[40]  Genevieve Konopka,et al.  MEF2C regulates cortical inhibitory and excitatory synapses and behaviors relevant to neurodevelopmental disorders , 2016, eLife.

[41]  R. Rocha,et al.  MEF2C haploinsufficiency syndrome: Report of a new MEF2C mutation and review. , 2016, European journal of medical genetics.

[42]  R. Schiestl,et al.  MEF2C protects bone marrow B-lymphoid progenitors during stress haematopoiesis , 2016, Nature Communications.

[43]  Pedro Olivares-Chauvet,et al.  UMI-4C for quantitative and targeted chromosomal contact profiling , 2016, Nature Methods.

[44]  A. Righini,et al.  Prenatal detection of 5q14.3 duplication including MEF2C and brain phenotype , 2016, American journal of medical genetics. Part A.

[45]  J. Mesirov,et al.  The Molecular Signatures Database Hallmark Gene Set Collection , 2015 .

[46]  A. Visel,et al.  Disruptions of Topological Chromatin Domains Cause Pathogenic Rewiring of Gene-Enhancer Interactions , 2015, Cell.

[47]  I. Rigoutsos,et al.  “Off-Spotter”: very fast and exhaustive enumeration of genomic lookalikes for designing CRISPR/Cas guide RNAs , 2015, Biology Direct.

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

[49]  Tomas W. Fitzgerald,et al.  Large-scale discovery of novel genetic causes of developmental disorders , 2014, Nature.

[50]  Neva C. Durand,et al.  A 3D Map of the Human Genome at Kilobase Resolution Reveals Principles of Chromatin Looping , 2014, Cell.

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

[52]  Kali T. Witherspoon,et al.  Refining analyses of copy number variation identifies specific genes associated with developmental delay , 2014, Nature Genetics.

[53]  J. Leek svaseq: removing batch effects and other unwanted noise from sequencing data , 2014, bioRxiv.

[54]  Edwin Cuppen,et al.  Mate pair sequencing for the detection of chromosomal aberrations in patients with intellectual disability and congenital malformations , 2013, European Journal of Human Genetics.

[55]  S. Horvath,et al.  Integrative Functional Genomic Analyses Implicate Specific Molecular Pathways and Circuits in Autism , 2013, Cell.

[56]  U. Stephani,et al.  Mutations in GRIN2A cause idiopathic focal epilepsy with rolandic spikes , 2013, Nature Genetics.

[57]  Eli J. Fine,et al.  DNA targeting specificity of RNA-guided Cas9 nucleases , 2013, Nature Biotechnology.

[58]  T. Südhof,et al.  Rapid Single-Step Induction of Functional Neurons from Human Pluripotent Stem Cells , 2013, Neuron.

[59]  S. Esposito,et al.  MEF2C deletions and mutations versus duplications: a clinical comparison. , 2013, European journal of medical genetics.

[60]  Marni J. Falk,et al.  MEF2C Haploinsufficiency features consistent hyperkinesis, variable epilepsy, and has a role in dorsal and ventral neuronal developmental pathways , 2013, neurogenetics.

[61]  V. Vedantham,et al.  Spatiotemporal regulation of an Hcn4 enhancer defines a role for Mef2c and HDACs in cardiac electrical patterning. , 2013, Developmental biology.

[62]  Thomas R. Gingeras,et al.  STAR: ultrafast universal RNA-seq aligner , 2013, Bioinform..

[63]  H. Mikkola,et al.  Mef2C Maintains B Cell Homeostasis Through the Regulation of DNA Repair Machinery , 2012 .

[64]  Gary D Bader,et al.  A travel guide to Cytoscape plugins , 2012, Nature Methods.

[65]  J. Chelly,et al.  Refining the phenotype associated with MEF2C point mutations , 2012, neurogenetics.

[66]  Data production leads,et al.  An integrated encyclopedia of DNA elements in the human genome , 2012 .

[67]  B. Porter,et al.  MEF2C haploinsufficiency syndrome: A severe neurodevelopmental disorder with variable epilepsy , 2012, Journal of Pediatric Epilepsy.

[68]  H. Mori,et al.  De novo microdeletion of 5q14.3 excluding MEF2C in a patient with infantile spasms, microcephaly, and agenesis of the corpus callosum , 2012, American journal of medical genetics. Part A.

[69]  H. Kurahashi,et al.  Clinical spectrum of SCN2A mutations , 2012, Brain and Development.

[70]  ENCODEConsortium,et al.  An Integrated Encyclopedia of DNA Elements in the Human Genome , 2012, Nature.

[71]  Chris Williams,et al.  RNA-SeQC: RNA-seq metrics for quality control and process optimization , 2012, Bioinform..

[72]  Jesse R. Dixon,et al.  Topological Domains in Mammalian Genomes Identified by Analysis of Chromatin Interactions , 2012, Nature.

[73]  Evan T. Geller,et al.  Patterns and rates of exonic de novo mutations in autism spectrum disorders , 2012, Nature.

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

[75]  N. Miyake,et al.  De novo 5q14.3 translocation 121.5‐kb upstream of MEF2C in a patient with severe intellectual disability and early‐onset epileptic encephalopathy , 2011, American journal of medical genetics. Part A.

[76]  Peter J. Bickel,et al.  Measuring reproducibility of high-throughput experiments , 2011, 1110.4705.

[77]  M. Vikkula,et al.  5q14.3 neurocutaneous syndrome: A novel continguous gene syndrome caused by simultaneous deletion of RASA1 and MEF2C , 2011, American journal of medical genetics. Part A.

[78]  Golder N Wilson,et al.  Interstitial deletion 5q14.3q21.3 with MEF2C haploinsufficiency and mild phenotype: When more is less , 2011, American journal of medical genetics. Part A.

[79]  Helga Thorvaldsdóttir,et al.  Molecular signatures database (MSigDB) 3.0 , 2011, Bioinform..

[80]  C. Martin,et al.  5 q 14 . 3 Neurocutaneous Syndrome : A Novel Continguous Gene Syndrome Caused by Simultaneous Deletion of RASA 1 and MEF 2 C , 2011 .

[81]  E. Ortibus,et al.  Refining the phenotype associated with MEF2C haploinsufficiency , 2010, Clinical genetics.

[82]  P. Stankiewicz,et al.  Severe mental retardation, seizures, and hypotonia due to deletions of MEF2C , 2010, American journal of medical genetics. Part B, Neuropsychiatric genetics : the official publication of the International Society of Psychiatric Genetics.

[83]  T. Vuocolo,et al.  A gene network switch enhances the oxidative capacity of ovine skeletal muscle during late fetal development , 2010, BMC Genomics.

[84]  H. Firth,et al.  Mutations in MEF2C from the 5q14.3q15 microdeletion syndrome region are a frequent cause of severe mental retardation and diminish MECP2 and CDKL5 expression , 2010, Human mutation.

[85]  M. Venere,et al.  The Rho Guanine Nucleotide Exchange Factor AKAP13 (BRX) Is Essential for Cardiac Development in Mice* , 2010, The Journal of Biological Chemistry.

[86]  H. Firth,et al.  Mutations in MEF2C from the 5q14.3q15 microdeletion syndrome region are a frequent cause of severe mental retardation and diminish MECP2 and CDKL5 expression , 2010, Human mutation.

[87]  I. Amit,et al.  Comprehensive mapping of long range interactions reveals folding principles of the human genome , 2011 .

[88]  S. Auvin,et al.  MEF2C haploinsufficiency caused by either microdeletion of the 5q14.3 region or mutation is responsible for severe mental retardation with stereotypic movements, epilepsy and/or cerebral malformations , 2009, Journal of Medical Genetics.

[89]  Dagmar Wieczorek,et al.  A novel microdeletion syndrome involving 5q14.3-q15: clinical and molecular cytogenetic characterization of three patients , 2009, European Journal of Human Genetics.

[90]  Manuel Corpas,et al.  DECIPHER: Database of Chromosomal Imbalance and Phenotype in Humans Using Ensembl Resources. , 2009, American journal of human genetics.

[91]  I. Scheffer,et al.  Periventricular heterotopia, mental retardation, and epilepsy associated with 5q14.3-q15 deletion , 2009, Neurology.

[92]  Steve Horvath,et al.  WGCNA: an R package for weighted correlation network analysis , 2008, BMC Bioinformatics.

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

[94]  Mi-Sung Kim,et al.  MEF2C, a transcription factor that facilitates learning and memory by negative regulation of synapse numbers and function , 2008, Proceedings of the National Academy of Sciences.

[95]  Jiankun Cui,et al.  Myocyte Enhancer Factor 2C as a Neurogenic and Antiapoptotic Transcription Factor in Murine Embryonic Stem Cells , 2008, The Journal of Neuroscience.

[96]  A. Cao,et al.  Two patients with balanced translocations and autistic disorder: CSMD3 as a candidate gene for autism found in their common 8q23 breakpoint area , 2008, European Journal of Human Genetics.

[97]  Michael L. Creech,et al.  Integration of biological networks and gene expression data using Cytoscape , 2007, Nature Protocols.

[98]  Steven W. Flavell,et al.  Activity-Dependent Regulation of MEF2 Transcription Factors Suppresses Excitatory Synapse Number , 2006, Science.

[99]  Terrence S. Furey,et al.  The UCSC Genome Browser Database: update 2006 , 2005, Nucleic Acids Res..

[100]  Pablo Tamayo,et al.  Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[101]  P. Shannon,et al.  Cytoscape: a software environment for integrated models of biomolecular interaction networks. , 2003, Genome research.

[102]  M. Gómez-Ferrería,et al.  Functional domains of FOXJ2. , 2003, Journal of molecular biology.

[103]  C. Bucana,et al.  Control of mouse cardiac morphogenesis and myogenesis by transcription factor MEF2C. , 1997, Science.