DNA Sequence-Dependent Compartmentalization and Silencing of Chromatin at the Nuclear Lamina

A large fraction of the mammalian genome is organized into inactive chromosomal domains along the nuclear lamina. The mechanism by which these lamina associated domains (LADs) are established remains to be elucidated. Using genomic repositioning assays, we show that LADs, spanning the developmentally regulated IgH and Cyp3a loci contain discrete DNA regions that associate chromatin with the nuclear lamina and repress gene activity in fibroblasts. Lamina interaction is established during mitosis and likely involves the localized recruitment of Lamin B during late anaphase. Fine-scale mapping of LADs reveals numerous lamina-associating sequences (LASs), which are enriched for a GAGA motif. This repeated motif directs lamina association and is bound by the transcriptional repressor cKrox, in a complex with HDAC3 and Lap2β. Knockdown of cKrox or HDAC3 results in dissociation of LASs/LADs from the nuclear lamina. These results reveal a mechanism that couples nuclear compartmentalization of chromatin domains with the control of gene activity.

[1]  Kyewon Park,et al.  The role of ThPOK in control of CD4/CD8 lineage commitment. , 2010, Annual review of immunology.

[2]  E. Bertolino,et al.  Transcriptional repression mediated by repositioning of genes to the nuclear lamina , 2008, Nature.

[3]  M. Groudine,et al.  The locus control region is required for association of the murine beta-globin locus with engaged transcription factories during erythroid maturation. , 2006, Genes & development.

[4]  Joseph Rosenecker,et al.  Transcription-dependent spatial arrangements of CFTR and adjacent genes in human cell nuclei , 2004, The Journal of cell biology.

[5]  D. Spector,et al.  A genetic locus targeted to the nuclear periphery in living cells maintains its transcriptional competence , 2008, The Journal of cell biology.

[6]  Bas van Steensel,et al.  Detection of in vivo protein–DNA interactions using DamID in mammalian cells , 2007, Nature Protocols.

[7]  O. Cohen-Fix,et al.  Sizing up the nucleus: nuclear shape, size and nuclear-envelope assembly , 2009, Journal of Cell Science.

[8]  Wendy A. Bickmore,et al.  Transcription factories: gene expression in unions? , 2009, Nature Reviews Genetics.

[9]  David L. Spector,et al.  Nuclear speckles: a model for nuclear organelles , 2003, Nature Reviews Molecular Cell Biology.

[10]  N. Amariglio,et al.  The nuclear-envelope protein and transcriptional repressor LAP2β interacts with HDAC3 at the nuclear periphery, and induces histone H4 deacetylation , 2005, Journal of Cell Science.

[11]  Thomas Cremer,et al.  The A- and B-type nuclear lamin networks: microdomains involved in chromatin organization and transcription. , 2008, Genes & development.

[12]  P. Fraser,et al.  Antisense intergenic transcription in V(D)J recombination , 2004, Nature Immunology.

[13]  Antonio Ortega,et al.  Sparse representation and Bayesian detection of genome copy number alterations from microarray data , 2008, Bioinform..

[14]  Charles Elkan,et al.  Fitting a Mixture Model By Expectation Maximization To Discover Motifs In Biopolymer , 1994, ISMB.

[15]  S. Hart,et al.  Dynamic Patterns of Histone Methylation Are Associated with Ontogenic Expression of the Cyp3a Genes during Mouse Liver Maturation , 2009, Molecular Pharmacology.

[16]  A S Belmont,et al.  In vivo localization of DNA sequences and visualization of large-scale chromatin organization using lac operator/repressor recognition , 1996, The Journal of cell biology.

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

[18]  S. Fiering,et al.  Site-specific chromosomal integration in mammalian cells: highly efficient CRE recombinase-mediated cassette exchange. , 1999, Journal of molecular biology.

[19]  J. Wellink,et al.  Non-specific interactions are sufficient to explain the position of heterochromatic chromocenters and nucleoli in interphase nuclei , 2009, Nucleic acids research.

[20]  J. Licht,et al.  Critical Residues within the BTB Domain of PLZF and Bcl-6 Modulate Interaction with Corepressors , 2002, Molecular and Cellular Biology.

[21]  B. van Steensel,et al.  Genome-nuclear lamina interactions and gene regulation. , 2010, Current opinion in cell biology.

[22]  Florence Hediger,et al.  Separation of silencing from perinuclear anchoring functions in yeast Ku80, Sir4 and Esc1 proteins , 2004, The EMBO journal.

[23]  Elizabeth Kerr,et al.  Recruitment to the Nuclear Periphery Can Alter Expression of Genes in Human Cells , 2008, PLoS genetics.

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

[25]  J. Ihle,et al.  Regulation of interleukin 7–dependent immunoglobulin heavy-chain variable gene rearrangements by transcription factor STAT5 , 2005, Nature Immunology.

[26]  Yoichi Shinkai,et al.  G9a selectively represses a class of late-replicating genes at the nuclear periphery , 2009, Proceedings of the National Academy of Sciences.

[27]  Yosef Gruenbaum,et al.  Barrier-to-autointegration factor--a BAFfling little protein. , 2007, Trends in cell biology.

[28]  G. Karsenty,et al.  c-Krox, a transcriptional regulator of type I collagen gene expression, is preferentially expressed in skin. , 1994, Proceedings of the National Academy of Sciences of the United States of America.

[29]  Andrew J. Wilson,et al.  Hdac3 is essential for the maintenance of chromatin structure and genome stability. , 2010, Cancer cell.

[30]  A. Ponti,et al.  The spatial dynamics of tissue-specific promoters during C. elegans development. , 2010, Genes & development.

[31]  P. Georgel,et al.  GAGA protein: a multi-faceted transcription factor. , 2006, Biochemistry and cell biology = Biochimie et biologie cellulaire.

[32]  E. C. Schirmer,et al.  Proteins that associate with lamins: many faces, many functions. , 2007, Experimental cell research.

[33]  Navneet Matharu,et al.  Vertebrate homologue of Drosophila GAGA factor. , 2010, Journal of molecular biology.

[34]  Sara Ahmed,et al.  DNA zip codes control an ancient mechanism for gene targeting to the nuclear periphery , 2010, Nature Cell Biology.

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

[36]  F. Karch,et al.  The GAGA factor is required in the early Drosophila embryo not only for transcriptional regulation but also for nuclear division. , 1996, Development.

[37]  S. Gasser,et al.  The budding yeast nucleus. , 2010, Cold Spring Harbor perspectives in biology.

[38]  P. Silver,et al.  Transcriptional regulation at the nuclear pore complex. , 2007, Current opinion in genetics & development.

[39]  B. Alberts,et al.  The Drosophila GAGA transcription factor is associated with specific regions of heterochromatin throughout the cell cycle. , 1994, The EMBO journal.

[40]  M. Guenther,et al.  A SANT motif in the SMRT corepressor interprets the histone code and promotes histone deacetylation , 2003, The EMBO journal.

[41]  T. Misteli,et al.  The emerging role of nuclear architecture in DNA repair and genome maintenance , 2009, Nature Reviews Molecular Cell Biology.

[42]  Roy Riblet,et al.  Subnuclear Compartmentalization of Immunoglobulin Loci During Lymphocyte Development , 2002, Science.

[43]  C. Schildkraut,et al.  Sites That Direct Nuclear Compartmentalization Are near the 5′ End of the Mouse Immunoglobulin Heavy-Chain Locus , 2005, Molecular and Cellular Biology.