KLF4 is involved in the organization and regulation of pluripotency-associated three-dimensional enhancer networks

Cell fate transitions are accompanied by global transcriptional, epigenetic and topological changes driven by transcription factors, as is exemplified by reprogramming somatic cells to pluripotent stem cells through the expression of OCT4, KLF4, SOX2 and cMYC. How transcription factors orchestrate the complex molecular changes around their target gene loci remains incompletely understood. Here, using KLF4 as a paradigm, we provide a transcription-factor-centric view of chromatin reorganization and its association with three-dimensional enhancer rewiring and transcriptional changes during the reprogramming of mouse embryonic fibroblasts to pluripotent stem cells. Inducible depletion of KLF factors in PSCs caused a genome-wide decrease in enhancer connectivity, whereas disruption of individual KLF4 binding sites within pluripotent-stem-cell-specific enhancers was sufficient to impair enhancer–promoter contacts and reduce the expression of associated genes. Our study provides an integrative view of the complex activities of a lineage-specifying transcription factor and offers novel insights into the nature of the molecular events that follow transcription factor binding. Di Giammartino, Kloetgen, Polyzos, Liu et al. probe chromatin organization, enhancer status and transcriptional changes and show that KLF4 acts as a transcriptional regulator and chromatin organizer during induced pluripotent stem cell reprogramming and in pluripotent stem cells.

[1]  J. Ernst,et al.  Cooperative Binding of Transcription Factors Orchestrates Reprogramming , 2017, Cell.

[2]  K. Plath,et al.  Identification and Single-Cell Functional Characterization of an Endodermally Biased Pluripotent Substate in Human Embryonic Stem Cells , 2018, Stem cell reports.

[3]  Daniel Capurso,et al.  Multiplex chromatin interactions with single-molecule precision , 2019, Nature.

[4]  Jill M Dowen,et al.  Control of Cell Identity Genes Occurs in Insulated Neighborhoods in Mammalian Chromosomes , 2014, Cell.

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

[6]  A. Hutchins,et al.  Chromatin Accessibility Dynamics during iPSC Reprogramming. , 2017, Cell stem cell.

[7]  D. C. Di Giammartino,et al.  The Chromatin Signature of Pluripotency: Establishment and Maintenance , 2016, Current Stem Cell Reports.

[8]  P. Rigollet,et al.  Optimal-Transport Analysis of Single-Cell Gene Expression Identifies Developmental Trajectories in Reprogramming , 2019, Cell.

[9]  T. Graf,et al.  Historical origins of transdifferentiation and reprogramming. , 2011, Cell stem cell.

[10]  G. Natoli Maintaining cell identity through global control of genomic organization. , 2010, Immunity.

[11]  Christian L. Müller,et al.  4C-ker: A Method to Reproducibly Identify Genome-Wide Interactions Captured by 4C-Seq Experiments , 2016, bioRxiv.

[12]  Greg Donahue,et al.  Facilitators and Impediments of the Pluripotency Reprogramming Factors' Initial Engagement with the Genome , 2012, Cell.

[13]  Howard Y. Chang,et al.  Enhancer connectome in primary human cells identifies target genes of disease-associated DNA elements , 2017, Nature Genetics.

[14]  R. Maehr,et al.  Functional annotation of native enhancers with a Cas9 -histone demethylase fusion , 2015, Nature Methods.

[15]  Jennifer A. Mitchell,et al.  Preferential associations between co-regulated genes reveal a transcriptional interactome in erythroid cells , 2010, Nature Genetics.

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

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

[18]  L. Di Croce,et al.  The role of Polycomb in stem cell genome architecture. , 2016, Current opinion in cell biology.

[19]  Wange Lu,et al.  Klf4 organizes long-range chromosomal interactions with the oct4 locus in reprogramming and pluripotency. , 2013, Cell stem cell.

[20]  Charles H. Li,et al.  Mediator and RNA polymerase II clusters associate in transcription-dependent condensates , 2018, Science.

[21]  Sheng Zhong,et al.  A core Klf circuitry regulates self-renewal of embryonic stem cells , 2008, Nature Cell Biology.

[22]  Mike J. Mason,et al.  Role of the Murine Reprogramming Factors in the Induction of Pluripotency , 2009, Cell.

[23]  Michael P. Snyder,et al.  Mango: a bias-correcting ChIA-PET analysis pipeline , 2015, Bioinform..

[24]  Thomas G. Gilgenast,et al.  Local Genome Topology Can Exhibit an Incompletely Rewired 3D-Folding State during Somatic Cell Reprogramming. , 2016, Cell stem cell.

[25]  Louise S. Matheson,et al.  Polycomb repressive complex PRC1 spatially constrains the mouse embryonic stem cell genome , 2015, Nature Genetics.

[26]  David A. Orlando,et al.  Master Transcription Factors and Mediator Establish Super-Enhancers at Key Cell Identity Genes , 2013, Cell.

[27]  S. Heath,et al.  Transcription Factors Drive Tet2-Mediated Enhancer Demethylation to Reprogram Cell Fate. , 2018, Cell stem cell.

[28]  Aristotelis Tsirigos,et al.  HiC-bench: comprehensive and reproducible Hi-C data analysis designed for parameter exploration and benchmarking , 2016, BMC Genomics.

[29]  Tomohiro Kono,et al.  Aberrant silencing of imprinted genes on chromosome 12qF1 in mouse induced pluripotent stem cells , 2010, Nature.

[30]  Guillaume J. Filion,et al.  Transcription factors and 3D genome conformation in cell-fate decisions , 2019, Nature.

[31]  Mark D. Robinson,et al.  edgeR: a Bioconductor package for differential expression analysis of digital gene expression data , 2009, Bioinform..

[32]  Michael Levine,et al.  Enhancer Control of Transcriptional Bursting , 2016, Cell.

[33]  S. Ambs,et al.  Epigenetic regulation of REX1 expression and chromatin binding specificity by HMGNs. , 2019, Nucleic acids research.

[34]  Howard Y. Chang,et al.  HiChIP: efficient and sensitive analysis of protein-directed genome architecture , 2016, Nature Methods.

[35]  K. Hochedlinger,et al.  A reprogrammable mouse strain from gene-targeted embryonic stem cells , 2010, Nature Methods.

[36]  O. Elemento,et al.  Widespread Mitotic Bookmarking by Histone Marks and Transcription Factors in Pluripotent Stem Cells. , 2017, Cell reports.

[37]  G. Pan,et al.  Resolving Cell Fate Decisions during Somatic Cell Reprogramming by Single-Cell RNA-Seq. , 2019, Molecular cell.

[38]  P. Park,et al.  Ascorbic acid prevents loss of Dlk1-Dio3 imprinting and facilitates generation of all-iPS cell mice from terminally differentiated B cells , 2012, Nature Genetics.

[39]  Nathan C. Sheffield,et al.  LOLA: enrichment analysis for genomic region sets and regulatory elements in R and Bioconductor , 2015, Bioinform..

[40]  Jonathan M. Cairns,et al.  Long-Range Enhancer Interactions Are Prevalent in Mouse Embryonic Stem Cells and Are Reorganized upon Pluripotent State Transition , 2018, Cell reports.

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

[42]  Daniel S. Kim,et al.  Lineage-specific dynamic and pre-established enhancer–promoter contacts cooperate in terminal differentiation , 2017, Nature Genetics.

[43]  Eli R. Zunder,et al.  A continuous molecular roadmap to iPSC reprogramming through progression analysis of single-cell mass cytometry. , 2015, Cell stem cell.

[44]  Hong Wang,et al.  Hierarchical Oct4 Binding in Concert with Primed Epigenetic Rearrangements during Somatic Cell Reprogramming. , 2016, Cell reports.

[45]  Luke A. Gilbert,et al.  CRISPR interference (CRISPRi) for sequence-specific control of gene expression , 2013, Nature Protocols.

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

[47]  Francesco Ferrari,et al.  Genome-wide chromatin interactions of the Nanog locus in pluripotency, differentiation, and reprogramming. , 2013, Cell stem cell.

[48]  Anthony D. Schmitt,et al.  A Compendium of Chromatin Contact Maps Reveals Spatially Active Regions in the Human Genome. , 2016, Cell reports.

[49]  Simona Bianco,et al.  Single-allele chromatin interactions identify regulatory hubs in dynamic compartmentalized domains , 2018, Nature Genetics.

[50]  S. Ramaswamy,et al.  A Molecular Roadmap of Reprogramming Somatic Cells into iPS Cells , 2012, Cell.

[51]  J. Carroll,et al.  Rapid immunoprecipitation mass spectrometry of endogenous proteins (RIME) for analysis of chromatin complexes , 2016, Nature Protocols.

[52]  S. Yamanaka,et al.  Induction of Pluripotent Stem Cells from Mouse Embryonic and Adult Fibroblast Cultures by Defined Factors , 2006, Cell.

[53]  Pedro P. Rocha,et al.  Identification of multi-loci hubs from 4C-seq demonstrates the functional importance of simultaneous interactions , 2016, Nucleic acids research.

[54]  Britta A. M. Bouwman,et al.  Enhancer hubs and loop collisions identified from single-allele topologies , 2018, Nature Genetics.

[55]  M. Stadtfeld,et al.  Cellular trajectories and molecular mechanisms of iPSC reprogramming. , 2018, Current opinion in genetics & development.

[56]  F. Grosveld,et al.  The active spatial organization of the beta-globin locus requires the transcription factor EKLF. , 2004, Genes & development.

[57]  V. Corces,et al.  Architectural proteins: regulators of 3D genome organization in cell fate. , 2014, Trends in cell biology.

[58]  K. Hochedlinger,et al.  Defining molecular cornerstones during fibroblast to iPS cell reprogramming in mouse. , 2008, Cell stem cell.

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

[60]  Pedro Olivares-Chauvet,et al.  Capturing pairwise and multi-way chromosomal conformations using chromosomal walks , 2016, Nature.

[61]  M. Pellegrini,et al.  Pioneer Transcription Factors Target Partial DNA Motifs on Nucleosomes to Initiate Reprogramming , 2015, Cell.

[62]  Guo-Cheng Yuan,et al.  Dissecting super-enhancer hierarchy based on chromatin interactions , 2017, Nature Communications.

[63]  K. Hochedlinger,et al.  Chromatin dynamics during cellular reprogramming , 2013, Nature.

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

[65]  Kin Chung Lam,et al.  High-resolution TADs reveal DNA sequences underlying genome organization in flies , 2017, Nature Communications.

[66]  Javier Quilez,et al.  Transcription factors orchestrate dynamic interplay between genome topology and gene regulation during cell reprogramming , 2017, Nature Genetics.

[67]  Richard A Young,et al.  Control of the Embryonic Stem Cell State , 2011, Cell.

[68]  Peter H. L. Krijger,et al.  Cell-of-Origin-Specific 3D Genome Structure Acquired during Somatic Cell Reprogramming , 2016, Cell stem cell.

[69]  R. Lister,et al.  Transient and Permanent Reconfiguration of Chromatin and Transcription Factor Occupancy Drive Reprogramming. , 2017, Cell stem cell.

[70]  Matteo Pellegrini,et al.  Long-range chromatin contacts in embryonic stem cells reveal a role for pluripotency factors and polycomb proteins in genome organization. , 2013, Cell stem cell.

[71]  N. Hannett,et al.  Transcription Factors Activate Genes through the Phase-Separation Capacity of Their Activation Domains , 2018, Cell.

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

[73]  Aristotelis Tsirigos,et al.  Stratification of TAD boundaries reveals preferential insulation of super-enhancers by strong boundaries , 2018, Nature Communications.

[74]  Brad T. Sherman,et al.  Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources , 2008, Nature Protocols.

[75]  Yun Zhu,et al.  The pluripotent genome in three dimensions is shaped around pluripotency factors , 2013, Nature.

[76]  Rob Patro,et al.  Salmon provides fast and bias-aware quantification of transcript expression , 2017, Nature Methods.