Distinct roles of cohesin-SA1 and cohesin-SA2 in 3D chromosome organization

Two variant cohesin complexes containing SMC1, SMC3, RAD21 and either SA1 (also known as STAG1) or SA2 (also known as STAG2) are present in all cell types. We report here their genomic distribution and specific contributions to genome organization in human cells. Although both variants are found at CCCTC-binding factor (CTCF) sites, a distinct population of the SA2-containing cohesin complexes (hereafter referred to as cohesin-SA2) localize to enhancers lacking CTCF, are linked to tissue-specific transcription and cannot be replaced by the SA1-containing cohesin complex (cohesin-SA1) when SA2 is absent, a condition that has been observed in several tumors. Downregulation of each of these variants has different consequences for gene expression and genome architecture. Our results suggest that cohesin-SA1 preferentially contributes to the stabilization of topologically associating domain boundaries together with CTCF, whereas cohesin-SA2 promotes cell-type-specific contacts between enhancers and promoters independently of CTCF. Loss of cohesin-SA2 rewires local chromatin contacts and alters gene expression. These findings provide insights into how cohesin mediates chromosome folding and establish a novel framework to address the consequences of mutations in cohesin genes in cancer.Depletion, ChIP and Hi-C analyses of the genomic distribution of the two variant cohesin complexes in human cells reveal non-redundant functions and differential contributions to 3D genome organization.

[1]  Timothy J. Durham,et al.  Systematic analysis of chromatin state dynamics in nine human cell types , 2011, Nature.

[2]  Boris Lenhard,et al.  Spatial enhancer clustering and regulation of enhancer-proximal genes by cohesin , 2015 .

[3]  A. Valencia,et al.  Recurrent inactivation of STAG2 in bladder cancer is not associated with aneuploidy , 2013, Nature Genetics.

[4]  David J. Reiss,et al.  CTCF physically links cohesin to chromatin , 2008, Proceedings of the National Academy of Sciences.

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

[6]  M. Rubin,et al.  Frequent truncating mutations of STAG2 in bladder cancer , 2013, Nature Genetics.

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

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

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

[10]  D. Pisano,et al.  A unique role of cohesin‐SA1 in gene regulation and development , 2012, The EMBO journal.

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

[12]  Hongtao Yu,et al.  ZNF198 Stabilizes the LSD1–CoREST–HDAC1 Complex on Chromatin through Its MYM-Type Zinc Fingers , 2008, PloS one.

[13]  J. Ellenberg,et al.  Topologically associating domains and chromatin loops depend on cohesin and are regulated by CTCF, WAPL, and PDS5 proteins , 2017, The EMBO journal.

[14]  Ilya M. Flyamer,et al.  Single-nucleus Hi-C reveals unique chromatin reorganization at oocyte-to-zygote transition , 2017, Nature.

[15]  Pedro P. Rocha,et al.  CTCF establishes discrete functional chromatin domains at the Hox clusters during differentiation , 2015, Science.

[16]  Maxim I Molodtsov,et al.  Rapid movement and transcriptional re‐localization of human cohesin on DNA , 2016, The EMBO journal.

[17]  A. Losada,et al.  Cohesin Mutations in Cancer. , 2016, Cold Spring Harbor perspectives in medicine.

[18]  Sally Temple,et al.  A Systematic Approach to Identify Candidate Transcription Factors that Control Cell Identity , 2015, Stem cell reports.

[19]  Nuno A. Fonseca,et al.  Two independent modes of chromosome organization are revealed by cohesin removal , 2016, bioRxiv.

[20]  Jennifer E. Phillips-Cremins,et al.  Architectural Protein Subclasses Shape 3D Organization of Genomes during Lineage Commitment , 2013, Cell.

[21]  Neva C. Durand,et al.  Chromatin extrusion explains key features of loop and domain formation in wild-type and engineered genomes , 2015, Proceedings of the National Academy of Sciences.

[22]  R. Kobayashi,et al.  Identification and Characterization of Sa/Scc3p Subunits in the Xenopus and Human Cohesin Complexes , 2000, The Journal of cell biology.

[23]  G. Mandel,et al.  REST and Its Corepressors Mediate Plasticity of Neuronal Gene Chromatin throughout Neurogenesis , 2005, Cell.

[24]  J. Ellenberg,et al.  Wapl is an essential regulator of chromatin structure and chromosome segregation , 2013, Nature.

[25]  Pedro P. Rocha,et al.  Cohesin loss alters adult hematopoietic stem cell homeostasis, leading to myeloproliferative neoplasms , 2015, The Journal of experimental medicine.

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

[27]  H. Aburatani,et al.  Cohesin mediates transcriptional insulation by CCCTC-binding factor , 2008, Nature.

[28]  M. Corces,et al.  The three-dimensional cancer genome. , 2016, Current opinion in genetics & development.

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

[30]  François Serra,et al.  Structural features of the fly chromatin colors revealed by automatic three-dimensional modeling , 2016, bioRxiv.

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

[32]  Chris Berdik Bladder cancer: 4 big questions , 2017, Nature.

[33]  David A. Orlando,et al.  Mediator and Cohesin Connect Gene Expression and Chromatin Architecture , 2010, Nature.

[34]  M. Mann,et al.  MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification , 2008, Nature Biotechnology.

[35]  Andre J. Faure,et al.  3D structure of individual mammalian genomes studied by single cell Hi-C , 2017, Nature.

[36]  Susan Smith,et al.  Differential regulation of telomere and centromere cohesion by the Scc3 homologues SA1 and SA2, respectively, in human cells , 2009, The Journal of cell biology.

[37]  Masao Nagasaki,et al.  Recurrent mutations in multiple components of the cohesin complex in myeloid neoplasms , 2012, Nature Genetics.

[38]  Christopher J. Ott,et al.  Dose-dependent role of the cohesin complex in normal and malignant hematopoiesis , 2015, The Journal of experimental medicine.

[39]  R. Shiekhattar,et al.  A core–BRAF35 complex containing histone deacetylase mediates repression of neuronal-specific genes , 2002, Proceedings of the National Academy of Sciences of the United States of America.

[40]  Marco Y. Hein,et al.  The Perseus computational platform for comprehensive analysis of (prote)omics data , 2016, Nature Methods.

[41]  C. Glass,et al.  Functional roles of enhancer RNAs for oestrogen-dependent transcriptional activation , 2013, Nature.

[42]  F. Uhlmann,et al.  Establishment of DNA-DNA Interactions by the Cohesin Ring , 2018, Cell.

[43]  Michael D. Wilson,et al.  Cohesin regulates tissue-specific expression by stabilizing highly occupied cis-regulatory modules , 2012, Genome research.

[44]  Petra C. Schwalie,et al.  A CTCF-independent role for cohesin in tissue-specific transcription. , 2010, Genome research.

[45]  Jill P. Mesirov,et al.  GSEA-P: a desktop application for Gene Set Enrichment Analysis , 2007, Bioinform..

[46]  M. Blasco,et al.  Cohesin‐SA1 deficiency drives aneuploidy and tumourigenesis in mice due to impaired replication of telomeres , 2012, The EMBO journal.

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

[48]  D. Pisano,et al.  The specific contributions of cohesin-SA1 to cohesion and gene expression , 2012, Cell cycle.

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

[50]  Jesse R. Dixon,et al.  Cohesin and CTCF differentially affect chromatin architecture and gene expression in human cells , 2013, Proceedings of the National Academy of Sciences.

[51]  A. Bishop,et al.  Cohesin SA2 is a sequence-independent DNA-binding protein that recognizes DNA replication and repair intermediates , 2017, The Journal of Biological Chemistry.

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

[53]  Boris Lenhard,et al.  Cohesin-based chromatin interactions enable regulated gene expression within preexisting architectural compartments , 2013, Genome research.

[54]  Kim Nasmyth,et al.  Biological chromodynamics: a general method for measuring protein occupancy across the genome by calibrating ChIP-seq , 2015, Nucleic acids research.

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

[56]  Mikael Bodén,et al.  MEME Suite: tools for motif discovery and searching , 2009, Nucleic Acids Res..

[57]  L. Mirny,et al.  Formation of Chromosomal Domains in Interphase by Loop Extrusion , 2015, bioRxiv.

[58]  Michael Q. Zhang,et al.  CRISPR Inversion of CTCF Sites Alters Genome Topology and Enhancer/Promoter Function , 2015, Cell.

[59]  A. Cuadrado,et al.  Essential role of p18Hamlet/SRCAP‐mediated histone H2A.Z chromatin incorporation in muscle differentiation , 2010, The EMBO journal.

[60]  Susan Smith,et al.  Protein requirements for sister telomere association in human cells , 2007, The EMBO journal.

[61]  A. Losada,et al.  Pds5B is required for cohesion establishment and Aurora B accumulation at centromeres , 2013, The EMBO journal.

[62]  M. Mann,et al.  Universal sample preparation method for proteome analysis , 2009, Nature Methods.

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

[64]  Marc A. Martí-Renom,et al.  Automatic analysis and 3D-modelling of Hi-C data using TADbit reveals structural features of the fly chromatin colors , 2017, PLoS Comput. Biol..

[65]  Kim Nasmyth,et al.  Cohesin: its roles and mechanisms. , 2009, Annual review of genetics.

[66]  Jing Liang,et al.  Chromatin architecture reorganization during stem cell differentiation , 2015, Nature.

[67]  Raymond K. Auerbach,et al.  Extensive Promoter-Centered Chromatin Interactions Provide a Topological Basis for Transcription Regulation , 2012, Cell.

[68]  P. Fraser,et al.  Cohesins form chromosomal cis-interactions at the developmentally regulated IFNG locus , 2009, Nature.

[69]  Fidel Ramírez,et al.  deepTools2: a next generation web server for deep-sequencing data analysis , 2016, Nucleic Acids Res..

[70]  Peter H. L. Krijger,et al.  The Cohesin Release Factor WAPL Restricts Chromatin Loop Extension , 2017, Cell.

[71]  Bruce Stillman,et al.  Chromatin Association of Human Origin Recognition Complex, Cdc6, and Minichromosome Maintenance Proteins during the Cell Cycle: Assembly of Prereplication Complexes in Late Mitosis , 2000, Molecular and Cellular Biology.

[72]  Catarina P. Santos,et al.  Synthetic lethality between the cohesin subunits STAG1 and STAG2 in diverse cancer contexts , 2017, bioRxiv.

[73]  M. Mann,et al.  Andromeda: a peptide search engine integrated into the MaxQuant environment. , 2011, Journal of proteome research.

[74]  Amos Tanay,et al.  Robust 4C-seq data analysis to screen for regulatory DNA interactions , 2012, Nature Methods.

[75]  D. Lim,et al.  Mechanical cue‐induced YAP instructs Skp2‐dependent cell cycle exit and oncogenic signaling , 2017, The EMBO journal.

[76]  Stephan Sauer,et al.  Cohesins Functionally Associate with CTCF on Mammalian Chromosome Arms , 2008, Cell.

[77]  Peter H. L. Krijger,et al.  CTCF Binding Polarity Determines Chromatin Looping. , 2015, Molecular cell.