Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Nat Methods ; 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38969721

ABSTRACT

The systematic determination of protein function is a key goal of modern biology, but remains challenging with current approaches. Here we present ORFtag, a versatile, cost-effective and highly efficient method for the massively parallel tagging and functional interrogation of proteins at the proteome scale. ORFtag uses retroviral vectors bearing a promoter, peptide tag and splice donor to generate fusions between the tag and endogenous open reading frames (ORFs). We demonstrate the utility of ORFtag through functional screens for transcriptional activators, repressors and posttranscriptional regulators in mouse embryonic stem cells. Each screen recovers known and identifies new regulators, including long ORFs inaccessible by other methods. Among other hits, we find that Zfp574 is a highly selective transcriptional activator and that oncogenic fusions often function as transactivators.

2.
Nature ; 626(7997): 207-211, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38086418

ABSTRACT

Enhancers control gene expression and have crucial roles in development and homeostasis1-3. However, the targeted de novo design of enhancers with tissue-specific activities has remained challenging. Here we combine deep learning and transfer learning to design tissue-specific enhancers for five tissues in the Drosophila melanogaster embryo: the central nervous system, epidermis, gut, muscle and brain. We first train convolutional neural networks using genome-wide single-cell assay for transposase-accessible chromatin with sequencing (ATAC-seq) datasets and then fine-tune the convolutional neural networks with smaller-scale data from in vivo enhancer activity assays, yielding models with 13% to 76% positive predictive value according to cross-validation. We designed and experimentally assessed 40 synthetic enhancers (8 per tissue) in vivo, of which 31 (78%) were active and 27 (68%) functioned in the target tissue (100% for central nervous system and muscle). The strategy of combining genome-wide and small-scale functional datasets by transfer learning is generally applicable and should enable the design of tissue-, cell type- and cell state-specific enhancers in any system.


Subject(s)
Deep Learning , Drosophila melanogaster , Embryo, Nonmammalian , Enhancer Elements, Genetic , Neural Networks, Computer , Organ Specificity , Animals , Chromatin/genetics , Chromatin/metabolism , Datasets as Topic , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Enhancer Elements, Genetic/genetics , Organ Specificity/genetics , Reproducibility of Results , Single-Cell Analysis , Transposases/metabolism , Synthetic Biology/methods
3.
Science ; 381(6654): 198-204, 2023 07 14.
Article in English | MEDLINE | ID: mdl-37440660

ABSTRACT

Gene expression is controlled by the precise activation and repression of transcription. Repression is mediated by specialized transcription factors (TFs) that recruit co-repressors (CoRs) to silence transcription, even in the presence of activating cues. However, whether CoRs can dominantly silence all enhancers or display distinct specificities is unclear. In this work, we report that most enhancers in Drosophila can be repressed by only a subset of CoRs, and enhancers classified by CoR sensitivity show distinct chromatin features, function, TF motifs, and binding. Distinct TF motifs render enhancers more resistant or sensitive to specific CoRs, as we demonstrate by motif mutagenesis and addition. These CoR-enhancer compatibilities constitute an additional layer of regulatory specificity that allows differential regulation at close genomic distances and is indicative of distinct mechanisms of transcriptional repression.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Enhancer Elements, Genetic , Repressor Proteins , Animals , Chromatin/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Gene Expression Regulation, Developmental , Repressor Proteins/chemistry , Repressor Proteins/genetics , Repressor Proteins/metabolism , Amino Acid Motifs
4.
Sci Rep ; 13(1): 6745, 2023 04 25.
Article in English | MEDLINE | ID: mdl-37185990

ABSTRACT

Enhancers are important cis-regulatory elements controlling cell-type specific expression patterns of genes. Furthermore, combinations of enhancers and minimal promoters are utilized to construct small, artificial promoters for gene delivery vectors. Large-scale functional screening methodology to construct genomic maps of enhancer activities has been successfully established in cultured cell lines, however, not yet applied to terminally differentiated cells and tissues in a living animal. Here, we transposed the Self-Transcribing Active Regulatory Region Sequencing (STARR-seq) technique to the mouse brain using adeno-associated-viruses (AAV) for the delivery of a highly complex screening library tiling entire genomic regions and covering in total 3 Mb of the mouse genome. We identified 483 sequences with enhancer activity, including sequences that were not predicted by DNA accessibility or histone marks. Characterizing the expression patterns of fluorescent reporters controlled by nine candidate sequences, we observed differential expression patterns also in sparse cell types. Together, our study provides an entry point for the unbiased study of enhancer activities in organisms during health and disease.


Subject(s)
Enhancer Elements, Genetic , Genomics , Animals , Mice , Genomics/methods , Chromosome Mapping/methods , Promoter Regions, Genetic , Brain
5.
Nature ; 606(7913): 406-413, 2022 06.
Article in English | MEDLINE | ID: mdl-35650434

ABSTRACT

All multicellular organisms rely on differential gene transcription regulated by genomic enhancers, which function through cofactors that are recruited by transcription factors1,2. Emerging evidence suggests that not all cofactors are required at all enhancers3-5, yet whether these observations reflect more general principles or distinct types of enhancers remained unknown. Here we categorized human enhancers by their cofactor dependencies and show that these categories provide a framework to understand the sequence and chromatin diversity of enhancers and their roles in different gene-regulatory programmes. We quantified enhancer activities along the entire human genome using STARR-seq6 in HCT116 cells, following the rapid degradation of eight cofactors. This analysis identified different types of enhancers with distinct cofactor requirements, sequences and chromatin properties. Some enhancers were insensitive to the depletion of the core Mediator subunit MED14 or the bromodomain protein BRD4 and regulated distinct transcriptional programmes. In particular, canonical Mediator7 seemed dispensable for P53-responsive enhancers, and MED14-depleted cells induced endogenous P53 target genes. Similarly, BRD4 was not required for the transcription of genes that bear CCAAT boxes and a TATA box (including histone genes and LTR12 retrotransposons) or for the induction of heat-shock genes. This categorization of enhancers through cofactor dependencies reveals distinct enhancer types that can bypass broadly utilized cofactors, which illustrates how alternative ways to activate transcription separate gene expression programmes and provide a conceptual framework to understand enhancer function and regulatory specificity.


Subject(s)
Enhancer Elements, Genetic , Transcription Factors , Cell Cycle Proteins/metabolism , Chromatin/genetics , Enhancer Elements, Genetic/genetics , Humans , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism
6.
Nat Genet ; 54(5): 613-624, 2022 05.
Article in English | MEDLINE | ID: mdl-35551305

ABSTRACT

Enhancer sequences control gene expression and comprise binding sites (motifs) for different transcription factors (TFs). Despite extensive genetic and computational studies, the relationship between DNA sequence and regulatory activity is poorly understood, and de novo enhancer design has been challenging. Here, we built a deep-learning model, DeepSTARR, to quantitatively predict the activities of thousands of developmental and housekeeping enhancers directly from DNA sequence in Drosophila melanogaster S2 cells. The model learned relevant TF motifs and higher-order syntax rules, including functionally nonequivalent instances of the same TF motif that are determined by motif-flanking sequence and intermotif distances. We validated these rules experimentally and demonstrated that they can be generalized to humans by testing more than 40,000 wildtype and mutant Drosophila and human enhancers. Finally, we designed and functionally validated synthetic enhancers with desired activities de novo.


Subject(s)
Drosophila melanogaster , Enhancer Elements, Genetic , Animals , Base Sequence , Binding Sites/genetics , Drosophila/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Curr Protoc Mol Biol ; 128(1): e105, 2019 09.
Article in English | MEDLINE | ID: mdl-31503413

ABSTRACT

The identification of transcriptional enhancers and the quantitative assessment of enhancer activities is essential to understanding how regulatory information for gene expression is encoded in animal and human genomes. Further, it is key to understanding how sequence variants affect enhancer function. STARR-seq enables the direct and quantitative assessment of enhancer activity for millions of candidate sequences of arbitrary length and origin in parallel, allowing the screening of entire genomes and the establishment of genome-wide enhancer activity maps. In STARR-seq, the candidate sequences are cloned downstream of the core promoter into a reporter gene's transcription unit (i.e., the 3' UTR). Candidates that function as active enhancers lead to the transcription of reporter mRNAs that harbor the candidates' sequences. This direct coupling of enhancer sequence and enhancer activity in cis enables the straightforward and efficient cloning of complex candidate libraries and the assessment of enhancer activities of millions of candidates in parallel by quantifying the reporter mRNAs by deep sequencing. This article describes how to create focused and genome-wide human STARR-seq libraries and how to perform STARR-seq screens in mammalian cells, and also describes a novel STARR-seq variant (UMI-STARR-seq) that allows the accurate counting of reporter mRNAs for STARR-seq libraries of low complexity. © 2019 The Authors. Basic Protocol 1: STARR-seq plasmid library cloning Basic Protocol 2: Mammalian STARR-seq screening protocol Alternate Protocol: UMI-STARR-seq screening protocol-unique molecular identifier integration Support Protocol: Transfection of human cells using the MaxCyte STX scalable transfection system.


Subject(s)
Enhancer Elements, Genetic , Genetic Techniques , Genomic Library , Animals , Cloning, Molecular , HeLa Cells , Humans , Plasmids , Transfection
8.
Nature ; 570(7759): 122-126, 2019 06.
Article in English | MEDLINE | ID: mdl-31092928

ABSTRACT

Transcriptional cofactors (COFs) communicate regulatory cues from enhancers to promoters and are central effectors of transcription activation and gene expression1. Although some COFs have been shown to prefer certain promoter types2-5 over others (for example, see refs 6,7), the extent to which different COFs display intrinsic specificities for distinct promoters is unclear. Here we use a high-throughput promoter-activity assay in Drosophila melanogaster S2 cells to screen 23 COFs for their ability to activate 72,000 candidate core promoters (CPs). We observe differential activation of CPs, indicating distinct regulatory preferences or 'compatibilities'8,9 between COFs and specific types of CPs. These functionally distinct CP types are differentially enriched for known sequence elements2,4, such as the TATA box, downstream promoter element (DPE) or TCT motif, and display distinct chromatin properties at endogenous loci. Notably, the CP types differ in their relative abundance of H3K4me3 and H3K4me1 marks (see also refs 10-12), suggesting that these histone modifications might distinguish trans-regulatory factors rather than promoter- versus enhancer-type cis-regulatory elements. We confirm the existence of distinct COF-CP compatibilities in two additional Drosophila cell lines and in human cells, for which we find COFs that prefer TATA-box or CpG-island promoters, respectively. Distinct compatibilities between COFs and promoters can explain how different enhancers specifically activate distinct sets of genes9, alternative promoters within the same genes, and distinct transcription start sites within the same promoter13. Thus, COF-promoter compatibilities may underlie distinct transcriptional programs in species as divergent as flies and humans.


Subject(s)
Promoter Regions, Genetic/genetics , Transcription Factors/metabolism , Transcription, Genetic , Transcriptional Activation , Animals , Cell Line , Chromatin/genetics , CpG Islands/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Enhancer Elements, Genetic/genetics , Histones/metabolism , Humans , Substrate Specificity , TATA Box/genetics , Transcription Initiation Site
9.
EMBO J ; 37(16)2018 08 15.
Article in English | MEDLINE | ID: mdl-30006452

ABSTRACT

Even though transcription factors (TFs) are central players of gene regulation and have been extensively studied, their regulatory trans-activation domains (tADs) often remain unknown and a systematic functional characterization of tADs is lacking. Here, we present a novel high-throughput approach tAD-seq to functionally test thousands of candidate tADs from different TFs in parallel. The tADs we identify by pooled screening validate in individual luciferase assays, whereas neutral regions do not. Interestingly, the tADs are found at arbitrary positions within the TF sequences and can contain amino acid (e.g., glutamine) repeat regions or overlap structured domains, including helix-loop-helix domains that are typically annotated as DNA-binding. We also identified tADs in the non-native reading frames, confirming that random sequences can function as tADs, albeit weakly. The identification of tADs as short protein sequences sufficient for transcription activation will enable the systematic study of TF function, which-particularly for TFs of different transcription activating functionalities-is still poorly understood.


Subject(s)
Drosophila Proteins , Trans-Activators , Transcription, Genetic , Animals , Cell Line , Drosophila Proteins/biosynthesis , Drosophila Proteins/genetics , Drosophila melanogaster , Protein Domains , Trans-Activators/biosynthesis , Trans-Activators/genetics
10.
Nat Methods ; 15(2): 141-149, 2018 02.
Article in English | MEDLINE | ID: mdl-29256496

ABSTRACT

The identification of transcriptional enhancers in the human genome is a prime goal in biology. Enhancers are typically predicted via chromatin marks, yet their function is primarily assessed with plasmid-based reporter assays. Here, we show that such assays are rendered unreliable by two previously reported phenomena relating to plasmid transfection into human cells: (i) the bacterial plasmid origin of replication (ORI) functions as a conflicting core promoter and (ii) a type I interferon (IFN-I) response is activated. These cause confounding false positives and negatives in luciferase assays and STARR-seq screens. We overcome both problems by employing the ORI as core promoter and by inhibiting two IFN-I-inducing kinases, enabling genome-wide STARR-seq screens in human cells. In HeLa-S3 cells, we uncover strong enhancers, IFN-I-induced enhancers, and enhancers endogenously silenced at the chromatin level. Our findings apply to all episomal enhancer activity assays in mammalian cells and are key to the characterization of human enhancers.


Subject(s)
Chromatin/genetics , Enhancer Elements, Genetic , Gene Expression Regulation , Genes, Reporter , Promoter Regions, Genetic , Chromosome Mapping , False Negative Reactions , Genome, Human , HeLa Cells , Humans
11.
Nat Biotechnol ; 35(2): 136-144, 2017 02.
Article in English | MEDLINE | ID: mdl-28024147

ABSTRACT

Gene expression is controlled by enhancers that activate transcription from the core promoters of their target genes. Although a key function of core promoters is to convert enhancer activities into gene transcription, whether and how strongly they activate transcription in response to enhancers has not been systematically assessed on a genome-wide level. Here we describe self-transcribing active core promoter sequencing (STAP-seq), a method to determine the responsiveness of genomic sequences to enhancers, and apply it to the Drosophila melanogaster genome. We cloned candidate fragments at the position of the core promoter (also called minimal promoter) in reporter plasmids with or without a strong enhancer, transfected the resulting library into cells, and quantified the transcripts that initiated from each candidate for each setup by deep sequencing. In the presence of a single strong enhancer, the enhancer responsiveness of different sequences differs by several orders of magnitude, and different levels of responsiveness are associated with genes of different functions. We also identify sequence features that predict enhancer responsiveness and discuss how different core promoters are employed for the regulation of gene expression.


Subject(s)
Base Pairing/genetics , Chromosome Mapping/methods , Enhancer Elements, Genetic/genetics , Gene Expression Regulation/genetics , Sequence Analysis, DNA/methods , Transcription Initiation, Genetic , Algorithms , Animals , Drosophila melanogaster , Promoter Regions, Genetic/genetics , Software
12.
PLoS One ; 11(8): e0161997, 2016.
Article in English | MEDLINE | ID: mdl-27575958

ABSTRACT

Hox homeodomain transcription factors are key regulators of animal development. They specify the identity of segments along the anterior-posterior body axis in metazoans by controlling the expression of diverse downstream targets, including transcription factors and signaling pathway components. The Drosophila melanogaster Hox factor Ultrabithorax (Ubx) directs the development of thoracic and abdominal segments and appendages, and loss of Ubx function can lead for example to the transformation of third thoracic segment appendages (e.g. halters) into second thoracic segment appendages (e.g. wings), resulting in a characteristic four-wing phenotype. Here we present a Drosophila melanogaster strain with a V5-epitope tagged Ubx allele, which we employed to obtain a high quality genome-wide map of Ubx binding sites using ChIP-seq. We confirm the sensitivity of the V5 ChIP-seq by recovering 7/8 of well-studied Ubx-dependent cis-regulatory regions. Moreover, we show that Ubx binding is predictive of enhancer activity as suggested by comparison with a genome-scale resource of in vivo tested enhancer candidates. We observed densely clustered Ubx binding sites at 12 extended genomic loci that included ANTP-C, BX-C, Polycomb complex genes, and other regulators and the clustered binding sites were frequently active enhancers. Furthermore, Ubx binding was detected at known Polycomb response elements (PREs) and was associated with significant enrichments of Pc and Pho ChIP signals in contrast to binding sites of other developmental TFs. Together, our results show that Ubx targets developmental regulators via strongly clustered binding sites and allow us to hypothesize that regulation by Ubx might involve Polycomb group proteins to maintain specific regulatory states in cooperative or mutually exclusive fashion, an attractive model that combines two groups of proteins with prominent gene regulatory roles during animal development.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Enhancer Elements, Genetic , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Animals , Binding Sites , Chromatin Immunoprecipitation , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Expression Regulation, Developmental , Genome , High-Throughput Nucleotide Sequencing , Polycomb-Group Proteins/genetics , Sequence Analysis, DNA
13.
Nature ; 518(7540): 556-9, 2015 Feb 26.
Article in English | MEDLINE | ID: mdl-25517091

ABSTRACT

Gene transcription in animals involves the assembly of RNA polymerase II at core promoters and its cell-type-specific activation by enhancers that can be located more distally. However, how ubiquitous expression of housekeeping genes is achieved has been less clear. In particular, it is unknown whether ubiquitously active enhancers exist and how developmental and housekeeping gene regulation is separated. An attractive hypothesis is that different core promoters might exhibit an intrinsic specificity to certain enhancers. This is conceivable, as various core promoter sequence elements are differentially distributed between genes of different functions, including elements that are predominantly found at either developmentally regulated or at housekeeping genes. Here we show that thousands of enhancers in Drosophila melanogaster S2 and ovarian somatic cells (OSCs) exhibit a marked specificity to one of two core promoters--one derived from a ubiquitously expressed ribosomal protein gene and another from a developmentally regulated transcription factor--and confirm the existence of these two classes for five additional core promoters from genes with diverse functions. Housekeeping enhancers are active across the two cell types, while developmental enhancers exhibit strong cell-type specificity. Both enhancer classes differ in their genomic distribution, the functions of neighbouring genes, and the core promoter elements of these neighbouring genes. In addition, we identify two transcription factors--Dref and Trl--that bind and activate housekeeping versus developmental enhancers, respectively. Our results provide evidence for a sequence-encoded enhancer-core-promoter specificity that separates developmental and housekeeping gene regulatory programs for thousands of enhancers and their target genes across the entire genome.


Subject(s)
Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental/genetics , Genes, Essential/genetics , Promoter Regions, Genetic/genetics , Animals , Base Sequence , Cell Line , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Genome, Insect/genetics , Models, Genetic , Organ Specificity , Substrate Specificity/genetics , Transcription Factors/metabolism , Transcriptional Activation/genetics
14.
Nat Genet ; 46(7): 685-92, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24908250

ABSTRACT

Phenotypic differences between closely related species are thought to arise primarily from changes in gene expression due to mutations in cis-regulatory sequences (enhancers). However, it has remained unclear how frequently mutations alter enhancer activity or create functional enhancers de novo. Here we use STARR-seq, a recently developed quantitative enhancer assay, to determine genome-wide enhancer activity profiles for five Drosophila species in the constant trans-regulatory environment of Drosophila melanogaster S2 cells. We find that the functions of a large fraction of D. melanogaster enhancers are conserved for their orthologous sequences owing to selection and stabilizing turnover of transcription factor motifs. Moreover, hundreds of enhancers have been gained since the D. melanogaster-Drosophila yakuba split about 11 million years ago without apparent adaptive selection and can contribute to changes in gene expression in vivo. Our finding that enhancer activity is often deeply conserved and frequently gained provides functional insights into regulatory evolution.


Subject(s)
Drosophila Proteins/genetics , Drosophila/genetics , Enhancer Elements, Genetic/genetics , Evolution, Molecular , Genome , Animals , Cells, Cultured , Drosophila/classification , Drosophila/growth & development , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Luciferases/metabolism , Transcription Factors/metabolism
15.
Nature ; 512(7512): 91-5, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-24896182

ABSTRACT

Transcriptional enhancers are crucial regulators of gene expression and animal development and the characterization of their genomic organization, spatiotemporal activities and sequence properties is a key goal in modern biology. Here we characterize the in vivo activity of 7,705 Drosophila melanogaster enhancer candidates covering 13.5% of the non-coding non-repetitive genome throughout embryogenesis. 3,557 (46%) candidates are active, suggesting a high density with 50,000 to 100,000 developmental enhancers genome-wide. The vast majority of enhancers display specific spatial patterns that are highly dynamic during development. Most appear to regulate their neighbouring genes, suggesting that the cis-regulatory genome is organized locally into domains, which are supported by chromosomal domains, insulator binding and genome evolution. However, 12 to 21 per cent of enhancers appear to skip non-expressed neighbours and regulate a more distal gene. Finally, we computationally identify cis-regulatory motifs that are predictive and required for enhancer activity, as we validate experimentally. This work provides global insights into the organization of an animal regulatory genome and the make-up of enhancer sequences and confirms and generalizes principles from previous studies. All enhancer patterns are annotated manually with a controlled vocabulary and all results are available through a web interface (http://enhancers.starklab.org), including the raw images of all microscopy slides for manual inspection at arbitrary zoom levels.


Subject(s)
Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Embryonic Development/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental/genetics , Genome, Insect/genetics , Animals , Internet , Nucleotide Motifs/genetics , Organ Specificity/genetics , Regulatory Sequences, Nucleic Acid/genetics , Reproducibility of Results , User-Computer Interface
16.
Nat Struct Mol Biol ; 19(10): 1023-30, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22983563

ABSTRACT

Heterochromatin is important for genome integrity and stabilization of gene-expression programs. We have identified the transcription factors Pax3 and Pax9 as redundant regulators of mouse heterochromatin, as they repress RNA output from major satellite repeats by associating with DNA within pericentric heterochromatin. Simultaneous depletion of Pax3 and Pax9 resulted in dramatic derepression of major satellite transcripts, persistent impairment of heterochromatic marks and defects in chromosome segregation. Genome-wide analyses of methylated histone H3 at Lys9 showed enrichment at intergenic major satellite repeats only when these sequences retained intact binding sites for Pax and other transcription factors. Additionally, bioinformatic interrogation of all histone methyltransferase Suv39h-dependent heterochromatic repeat regions in the mouse genome revealed a high concordance with the presence of transcription factor binding sites. These data define a general model in which reiterated arrangement of transcription factor binding sites within repeat sequences is an intrinsic mechanism of the formation of heterochromatin.


Subject(s)
Heterochromatin/metabolism , Paired Box Transcription Factors/metabolism , Animals , Base Sequence , Binding Sites , Cell Cycle/genetics , Chromosome Segregation , DNA, Satellite/metabolism , Fibroblasts/metabolism , Genome , Heterochromatin/genetics , Histones/metabolism , Lysine/metabolism , Methylation , Methyltransferases/genetics , Methyltransferases/metabolism , Mice , Mice, Mutant Strains , Molecular Sequence Data , PAX3 Transcription Factor , PAX5 Transcription Factor/genetics , PAX5 Transcription Factor/metabolism , PAX7 Transcription Factor/genetics , PAX7 Transcription Factor/metabolism , PAX9 Transcription Factor , Paired Box Transcription Factors/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism
17.
Genes Dev ; 22(15): 2048-61, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18676810

ABSTRACT

H4K20 methylation is a broad chromatin modification that has been linked with diverse epigenetic functions. Several enzymes target H4K20 methylation, consistent with distinct mono-, di-, and trimethylation states controlling different biological outputs. To analyze the roles of H4K20 methylation states, we generated conditional null alleles for the two Suv4-20h histone methyltransferase (HMTase) genes in the mouse. Suv4-20h-double-null (dn) mice are perinatally lethal and have lost nearly all H4K20me3 and H4K20me2 states. The genome-wide transition to an H4K20me1 state results in increased sensitivity to damaging stress, since Suv4-20h-dn chromatin is less efficient for DNA double-strand break (DSB) repair and prone to chromosomal aberrations. Notably, Suv4-20h-dn B cells are defective in immunoglobulin class-switch recombination, and Suv4-20h-dn deficiency impairs the stem cell pool of lymphoid progenitors. Thus, conversion to an H4K20me1 state results in compromised chromatin that is insufficient to protect genome integrity and to process a DNA-rearranging differentiation program in the mouse.


Subject(s)
Chromatin/metabolism , Gene Rearrangement , Genome , Histone-Lysine N-Methyltransferase/metabolism , Alleles , Animals , Chromatin/chemistry , Chromatin/genetics , Chromosome Painting , Crosses, Genetic , Heterozygote , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/genetics , Methylation , Mice , Mice, Knockout , Protein Methyltransferases , Repressor Proteins/genetics , Repressor Proteins/metabolism
18.
Nat Genet ; 30(1): 77-80, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11740497

ABSTRACT

Post-translational modifications of histone amino termini are an important regulatory mechanism that induce transitions in chromatin structure, thereby contributing to epigenetic gene control and the assembly of specialized chromosomal subdomains. Methylation of histone H3 at lysine 9 (H3-Lys9) by site-specific histone methyltransferases (Suv39h HMTases) marks constitutive heterochromatin. Here, we show that H3-Lys9 methylation also occurs in facultative heterochromatin of the inactive X chromosome (Xi) in female mammals. H3-Lys9 methylation is retained through mitosis, indicating that it might provide an epigenetic imprint for the maintenance of the inactive state. Disruption of the two mouse Suv39h HMTases abolishes H3-Lys9 methylation of constitutive heterochromatin but not that of the Xi. In addition, HP1 proteins, which normally associate with heterochromatin, do not accumulate with the Xi. These observations suggest the existence of an Suv39h-HP1-independent pathway regulating H3-Lys9 methylation of facultative heterochromatin.


Subject(s)
Dosage Compensation, Genetic , Heterochromatin/chemistry , Histone-Lysine N-Methyltransferase , Histones/metabolism , Lysine/analogs & derivatives , Lysine/chemistry , Protein Isoforms/metabolism , X Chromosome/metabolism , Amniocentesis , Aneuploidy , Animals , Binding Sites , Cells, Cultured/ultrastructure , Chromosome Painting , Chromosomes, Human, Pair 4/genetics , Female , Fibroblasts/ultrastructure , Gene Expression Regulation , Heterochromatin/genetics , Histone Methyltransferases , Histones/chemistry , Histones/immunology , Humans , Male , Metaphase , Methylation , Methyltransferases/physiology , Mice , Microscopy, Fluorescence , Mouth Mucosa/ultrastructure , Precipitin Tests , Pregnancy , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/immunology , Protein Methyltransferases , Repressor Proteins/physiology , Translocation, Genetic , X Chromosome/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...