An atlas of lamina-associated chromatin across thirteen human cell types reveals cell-type-specific and multiple subtypes of peripheral heterochromatin

Three-dimensional genome organization, specifically organization of heterochromatin at the nuclear periphery, coordinates cell type-specific gene regulation. While defining various histone modifications and chromatin-associated proteins in multiple cell types has provided important insights into epigenetic regulation of gene expression and cellular identity, peripheral heterochromatin has not been mapped comprehensively and relatively few examples have emerged detailing the role of peripheral heterochromatin in cellular identity, cell fate choices, and/or organogenesis. In this study, we define nuclear peripheral heterochromatin organization signatures based on association with LAMIN B1 and/or dimethylation of lysine 9 on H3 (H3K9me2) across thirteen human cell types encompassing pluripotent stem cells, intermediate progenitors and differentiated cells from all three germ layers. Genomic analyses across this atlas reveal that lamin-associated chromatin is organized into at least two different compartments, defined by differences in genome coverage, chromatin accessibility, residence of transposable elements, replication timing domains, and gene complements. Our datasets reveal that only a small subset of lamin-associated chromatin domains are cell type invariant, underscoring the complexity of peripheral heterochromatin organization. Moreover, by integrating peripheral chromatin maps with transcriptional data, we find evidence of cooperative shifts between chromatin structure and gene expression associated with each cell type. This atlas of peripheral chromatin provides the largest resource to date for peripheral chromatin organization and a deeper appreciation for how this organization may impact the establishment and maintenance of cellular identity.

[1]  H. Akaike A new look at the statistical model identification , 1974 .

[2]  L. Swanson,et al.  Expression of a large family of POU-domain regulatory genes in mammalian brain development , 1989, Nature.

[3]  A. Joyner,et al.  Multiple developmental defects in Engrailed-1 mutant mice: an early mid-hindbrain deletion and patterning defects in forelimbs and sternum. , 1994, Development.

[4]  R. Eisenman,et al.  Mnt, a novel Max-interacting protein is coexpressed with Myc in proliferating cells and mediates repression at Myc binding sites. , 1997, Genes & development.

[5]  J. Rubenstein,et al.  Homeobox gene Nkx2.2 and specification of neuronal identity by graded Sonic hedgehog signalling , 1999, Nature.

[6]  M. Tsai,et al.  The orphan nuclear receptor COUP-TFII is required for angiogenesis and heart development. , 1999, Genes & development.

[7]  Concepción Rodríguez-Esteban,et al.  Role of the Bicoid-related homeodomain factor Pitx1 in specifying hindlimb morphogenesis and pituitary development. , 1999, Genes & development.

[8]  H. Kazazian Mobile Elements: Drivers of Genome Evolution , 2004, Science.

[9]  C. Stewart,et al.  The laminopathies: the functional architecture of the nucleus and its contribution to disease. , 2006, Annual review of genomics and human genetics.

[10]  S. Grewal,et al.  Transcription and RNA interference in the formation of heterochromatin , 2007, Nature.

[11]  John D. Hunter,et al.  Matplotlib: A 2D Graphics Environment , 2007, Computing in Science & Engineering.

[12]  H. Worman,et al.  "Laminopathies": a wide spectrum of human diseases. , 2007, Experimental cell research.

[13]  Jeannie T. Lee,et al.  Identification of a Ctcf cofactor, Yy1, for the X chromosome binary switch. , 2007, Molecular cell.

[14]  D. Haussler,et al.  Species-specific endogenous retroviruses shape the transcriptional network of the human tumor suppressor protein p53 , 2007, Proceedings of the National Academy of Sciences.

[15]  A. Visel,et al.  Combinatorial Regulation of Endothelial Gene Expression by Ets and Forkhead Transcription Factors , 2008, Cell.

[16]  L. Wessels,et al.  Domain organization of human chromosomes revealed by mapping of nuclear lamina interactions , 2008, Nature.

[17]  E. Liu,et al.  Evolution of the mammalian transcription factor binding repertoire via transposable elements. , 2008, Genome research.

[18]  H. Cedar,et al.  De novo DNA methylation promoted by G9a prevents reprogramming of embryonically silenced genes , 2008, Nature Structural &Molecular Biology.

[19]  Athar N. Malik,et al.  Activity-dependent regulation of inhibitory synapse development by Npas4 , 2008, Nature.

[20]  R. Roeder,et al.  30 nm chromatin fibre decompaction requires both H4-K16 acetylation and linker histone eviction. , 2008, Journal of molecular biology.

[21]  B. Gaut,et al.  Epigenetic silencing of transposable elements: a trade-off between reduced transposition and deleterious effects on neighboring gene expression. , 2009, Genome research.

[22]  D. Gilbert,et al.  Replication timing and transcriptional control: beyond cause and effect--part II. , 2002, Current Opinion in Genetics and Development.

[23]  Gonçalo R. Abecasis,et al.  The Sequence Alignment/Map format and SAMtools , 2009, Bioinform..

[24]  Richard Durbin,et al.  Sequence analysis Fast and accurate short read alignment with Burrows – Wheeler transform , 2009 .

[25]  A. Feinberg,et al.  Large histone H3 lysine 9 dimethylated chromatin blocks distinguish differentiated from embryonic stem cells , 2009, Nature Genetics.

[26]  M. Ashburner,et al.  Fragile regions and not functional constraints predominate in shaping gene organization in the genus Drosophila. , 2010, Genome research.

[27]  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.

[28]  Aaron R. Quinlan,et al.  BIOINFORMATICS APPLICATIONS NOTE , 2022 .

[29]  G. Bourque,et al.  Transposable elements have rewired the core regulatory network of human embryonic stem cells , 2010, Nature Genetics.

[30]  Guillaume J. Filion,et al.  Systematic Protein Location Mapping Reveals Five Principal Chromatin Types in Drosophila Cells , 2010, Cell.

[31]  Tjerk P. Straatsma,et al.  NWChem: A comprehensive and scalable open-source solution for large scale molecular simulations , 2010, Comput. Phys. Commun..

[32]  Richard Durbin,et al.  Fast and accurate long-read alignment with Burrows–Wheeler transform , 2010, Bioinform..

[33]  D. Kavanagh,et al.  Cell-specific and lamin-dependent targeting of novel transmembrane proteins in the nuclear envelope , 2010, Cellular and Molecular Life Sciences.

[34]  J. Bednar,et al.  Single-base resolution mapping of H1–nucleosome interactions and 3D organization of the nucleosome , 2010, Proceedings of the National Academy of Sciences.

[35]  P. Flicek,et al.  Molecular maps of the reorganization of genome-nuclear lamina interactions during differentiation. , 2010, Molecular cell.

[36]  Gaël Varoquaux,et al.  Scikit-learn: Machine Learning in Python , 2011, J. Mach. Learn. Res..

[37]  Brent S. Pedersen,et al.  Pybedtools: a flexible Python library for manipulating genomic datasets and annotations , 2011, Bioinform..

[38]  Gaël Varoquaux,et al.  The NumPy Array: A Structure for Efficient Numerical Computation , 2011, Computing in Science & Engineering.

[39]  Tanneguy Redarce,et al.  Automatic Lip-Contour Extraction and Mouth-Structure Segmentation in Images , 2011, Computing in Science & Engineering.

[40]  Jun S. Song,et al.  Statistical Applications in Genetics and Molecular Biology Normalization , bias correction , and peak calling for ChIP-seq , 2012 .

[41]  Jianrong Wang,et al.  Genome-wide prediction and analysis of human chromatin boundary elements , 2011, Nucleic acids research.

[42]  F. Wang,et al.  Influences of lamin A levels on induction of pluripotent stem cells , 2012, Biology Open.

[43]  H. Worman Nuclear lamins and laminopathies , 2012, The Journal of pathology.

[44]  J. Sedat,et al.  Spatial partitioning of the regulatory landscape of the X-inactivation centre , 2012, Nature.

[45]  David R. Kelley,et al.  Transposable elements reveal a stem cell-specific class of long noncoding RNAs , 2012, Genome Biology.

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

[47]  N. Rhind,et al.  DNA replication timing. , 2013, Cold Spring Harbor perspectives in biology.

[48]  Zev N. Kronenberg,et al.  Transposable Elements Are Major Contributors to the Origin, Diversification, and Regulation of Vertebrate Long Noncoding RNAs , 2013, PLoS genetics.

[49]  Guillaume J. Filion,et al.  A network model of the molecular organization of chromatin in Drosophila. , 2013, Molecular cell.

[50]  L. Peichl,et al.  LBR and Lamin A/C Sequentially Tether Peripheral Heterochromatin and Inversely Regulate Differentiation , 2013, Cell.

[51]  Manolis Kellis,et al.  Constitutive nuclear lamina–genome interactions are highly conserved and associated with A/T-rich sequence , 2013, Genome research.

[52]  Petra C. Schwalie,et al.  Co-binding by YY1 identifies the transcriptionally active, highly conserved set of CTCF-bound regions in primate genomes , 2013, Genome Biology.

[53]  M. Greenberg,et al.  The activity-dependent transcription factor NPAS4 regulates domain-specific inhibition , 2013, Nature.

[54]  W. Bickmore,et al.  Single-Cell Dynamics of Genome-Nuclear Lamina Interactions , 2013, Cell.

[55]  Kimara L. Targoff,et al.  Nkx genes are essential for maintenance of ventricular identity , 2013, Development.

[56]  R. Lanz,et al.  Atrial identity is determined by a COUP-TFII regulatory network. , 2013, Developmental cell.

[57]  A. Oldenburg,et al.  Enriched domain detector: a program for detection of wide genomic enrichment domains robust against local variations , 2014, Nucleic acids research.

[58]  Fidel Ramírez,et al.  deepTools: a flexible platform for exploring deep-sequencing data , 2014, Nucleic Acids Res..

[59]  I. Weissman,et al.  Efficient endoderm induction from human pluripotent stem cells by logically directing signals controlling lineage bifurcations. , 2014, Cell stem cell.

[60]  J. Han,et al.  The transcription factor Pou3f1 promotes neural fate commitment via activation of neural lineage genes and inhibition of external signaling pathways , 2014, eLife.

[61]  Hanspeter Pfister,et al.  UpSet: Visualization of Intersecting Sets , 2014, IEEE Transactions on Visualization and Computer Graphics.

[62]  Yanli Wang,et al.  Topologically associating domains are stable units of replication-timing regulation , 2014, Nature.

[63]  Björn Usadel,et al.  Trimmomatic: a flexible trimmer for Illumina sequence data , 2014, Bioinform..

[64]  Alan R. Mardinly,et al.  Npas4 Regulates Excitatory-Inhibitory Balance within Neural Circuits through Cell-Type-Specific Gene Programs , 2014, Cell.

[65]  S. Wheelan,et al.  Directed targeting of chromatin to the nuclear lamina is mediated by chromatin state and A-type lamins , 2015, The Journal of cell biology.

[66]  Z. Chang,et al.  PU.1 Is Identified as a Novel Metastasis Suppressor in Hepatocellular Carcinoma Regulating the miR-615-5p/IGF2 Axis. , 2015, Asian Pacific journal of cancer prevention : APJCP.

[67]  Matthew A. Hibbs,et al.  Affinity-seq detects genome-wide PRDM9 binding sites and reveals the impact of prior chromatin modifications on mammalian recombination hotspot usage , 2015, Epigenetics & Chromatin.

[68]  Panagiota Arampatzi,et al.  Common stemness regulators of embryonic and cancer stem cells. , 2015, World journal of stem cells.

[69]  Alice Barateau,et al.  The p.R482W substitution in A-type lamins deregulates SREBP1 activity in Dunnigan-type familial partial lipodystrophy. , 2015, Human molecular genetics.

[70]  Siddharth S. Dey,et al.  Genome-wide Maps of Nuclear Lamina Interactions in Single Human Cells , 2015, Cell.

[71]  M. Garcia-Parajo,et al.  Chromatin Fibers Are Formed by Heterogeneous Groups of Nucleosomes In Vivo , 2015, Cell.

[72]  Moonsup Lee,et al.  FOXKs promote Wnt/β-catenin signaling by translocating DVL into the nucleus. , 2015, Developmental cell.

[73]  E. C. Schirmer,et al.  Tissue-Specific Gene Repositioning by Muscle Nuclear Membrane Proteins Enhances Repression of Critical Developmental Genes during Myogenesis , 2016, Molecular cell.

[74]  Pang Wei Koh,et al.  An atlas of transcriptional, chromatin accessibility, and surface marker changes in human mesoderm development , 2016, Scientific Data.

[75]  Md. Abul Hassan Samee,et al.  Complex Interdependence Regulates Heterotypic Transcription Factor Distribution and Coordinates Cardiogenesis , 2016, Cell.

[76]  Pang Wei Koh,et al.  Mapping the Pairwise Choices Leading from Pluripotency to Human Bone, Heart, and Other Mesoderm Cell Types , 2016, Cell.

[77]  M. Tolstorukov,et al.  Enhancer regions show high histone H3.3 turnover that changes during differentiation , 2016, eLife.

[78]  Lior Pachter,et al.  Near-optimal probabilistic RNA-seq quantification , 2016, Nature Biotechnology.

[79]  Krzysztof J. Gorgolewski,et al.  A phenome-wide examination of neural and cognitive function , 2016, Scientific Data.

[80]  L. Studer,et al.  A Modular Platform for Differentiation of Human PSCs into All Major Ectodermal Lineages. , 2017, Cell stem cell.

[81]  Howard Y. Chang,et al.  Genome-Wide Temporal Profiling of Transcriptome and Open Chromatin of Early Cardiomyocyte Differentiation Derived From hiPSCs and hESCs , 2017, Circulation research.

[82]  Jacob Schreiber,et al.  Pomegranate: fast and flexible probabilistic modeling in python , 2017, J. Mach. Learn. Res..

[83]  Daniel S. Day,et al.  YY1 Is a Structural Regulator of Enhancer-Promoter Loops , 2017, Cell.

[84]  J. Epstein,et al.  Genome-Nuclear Lamina Interactions Regulate Cardiac Stem Cell Lineage Restriction , 2017, Cell.

[85]  A. Sharp,et al.  Foxa2 identifies a cardiac progenitor population with ventricular differentiation potential , 2017, Nature Communications.

[86]  M. Hetzer,et al.  Nucleolar expansion and elevated protein translation in premature aging , 2017, Nature Communications.

[87]  Lior Pachter,et al.  Differential analysis of RNA-seq incorporating quantification uncertainty , 2016, Nature Methods.

[88]  L. Mirny,et al.  Targeted Degradation of CTCF Decouples Local Insulation of Chromosome Domains from Genomic Compartmentalization , 2017, Cell.

[89]  Jennifer E. Phillips-Cremins,et al.  YY1 and CTCF orchestrate a 3D chromatin looping switch during early neural lineage commitment , 2017, Genome research.

[90]  M. Hetzer,et al.  Coaching from the sidelines: the nuclear periphery in genome regulation , 2018, Nature Reviews Genetics.

[91]  S. Armstrong,et al.  HOXA9 Reprograms the Enhancer Landscape to Promote Leukemogenesis. , 2018, Cancer cell.

[92]  V. Corces,et al.  Organizational principles of 3D genome architecture , 2018, Nature Reviews Genetics.

[93]  William Stafford Noble,et al.  Integrative detection and analysis of structural variation in cancer genomes , 2018, Nature Genetics.

[94]  Jonas Paulsen,et al.  The lipodystrophic hotspot lamin A p.R482W mutation deregulates the mesodermal inducer T/Brachyury and early vascular differentiation gene networks , 2018, Human molecular genetics.

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

[96]  Deniz M. Ozata,et al.  PIWI-interacting RNAs: small RNAs with big functions , 2018, Nature Reviews Genetics.

[97]  David R. Kelley,et al.  Predicting 3D genome folding from DNA sequence , 2019, bioRxiv.

[98]  Elie N. Farah,et al.  Transcriptionally Active HERV-H Retrotransposons Demarcate Topologically Associating Domains in Human Pluripotent Stem Cells , 2019, Nature Genetics.

[99]  M. Cosma,et al.  Super-resolution microscopy reveals how histone tail acetylation affects DNA compaction within nucleosomes in vivo , 2019, Nucleic acids research.

[100]  Jianfeng Pei,et al.  H2A.Z facilitates licensing and activation of early replication origins , 2019, Nature.

[101]  Nezar Abdennur,et al.  Cooler: scalable storage for Hi-C data and other genomically-labeled arrays , 2019, bioRxiv.

[102]  D. Gilbert,et al.  Control of DNA replication timing in the 3D genome , 2019, Nature Reviews Molecular Cell Biology.

[103]  Wei Xie,et al.  The role of 3D genome organization in development and cell differentiation , 2019, Nature Reviews Molecular Cell Biology.

[104]  Howard Y. Chang,et al.  Activation of PDGF Pathway Links LMNA Mutation to Dilated Cardiomyopathy , 2019, Nature.

[105]  Jinchuan Xing,et al.  SIRT7 mediates L1 elements transcriptional repression and their association with the nuclear lamina , 2019, Nucleic acids research.

[106]  Dong Hun Lee,et al.  ETV2/ER71 Transcription Factor as a Therapeutic Vehicle for Cardiovascular Disease , 2019, Theranostics.

[107]  L. Mirny,et al.  Heterochromatin drives compartmentalization of inverted and conventional nuclei , 2019, Nature.

[108]  Z. Duan,et al.  Replication Timing Becomes Intertwined with 3D Genome Organization , 2019, Cell.

[109]  Joshua M. Stuart,et al.  Modeling Human TBX5 Haploinsufficiency Predicts Regulatory Networks for Congenital Heart Disease. , 2019, Developmental cell.

[110]  H3K9me2 orchestrates inheritance of spatial positioning of peripheral heterochromatin through mitosis , 2019, eLife.

[111]  J. Wysocka,et al.  Transposable elements as a potent source of diverse cis-regulatory sequences in mammalian genomes , 2020, Philosophical Transactions of the Royal Society B.

[112]  P. Ellinor,et al.  Myocyte Specific Upregulation of ACE2 in Cardiovascular Disease: Implications for SARS-CoV-2 mediated myocarditis , 2020, medRxiv.

[113]  D. Gilbert,et al.  SPIN reveals genome-wide landscape of nuclear compartmentalization , 2020, Genome Biology.

[114]  K. Loh,et al.  Improving the safety of human pluripotent stem cell therapies using genome-edited orthogonal safeguards , 2020, Nature Communications.

[115]  N. Briand,et al.  Lamina-associated domains: peripheral matters and internal affairs , 2020, Genome Biology.

[116]  M. Pallen,et al.  Assembly of hundreds of novel bacterial genomes from the chicken caecum , 2020, Genome Biology.

[117]  L. Weinberger,et al.  Chromatin remodeler Brahma safeguards canalization in cardiac mesoderm differentiation , 2020, bioRxiv.

[118]  Nezar Abdennur,et al.  Cooler: scalable storage for Hi-C data and other genomically labeled arrays , 2020, Bioinform..

[119]  Ting Wang,et al.  Co-opted transposons help perpetuate conserved higher-order chromosomal structures , 2020, Genome Biology.

[120]  V. Nizet,et al.  Upon microbial challenge, human neutrophils undergo rapid changes in nuclear architecture and chromatin folding to orchestrate an immediate inflammatory gene program , 2020, Genes & development.