Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
bioRxiv ; 2023 Apr 20.
Article in English | MEDLINE | ID: mdl-37131839

ABSTRACT

Aberrant transcriptional programming and chromatin dysregulation are common to most cancers. Whether by deranged cell signaling or environmental insult, the resulting oncogenic phenotype is typically manifested in transcriptional changes characteristic of undifferentiated cell growth. Here we analyze targeting of an oncogenic fusion protein, BRD4-NUT, composed of two normally independent chromatin regulators. The fusion causes the formation of large hyperacetylated genomic regions or megadomains, mis-regulation of c-MYC , and an aggressive carcinoma of squamous cell origin. Our previous work revealed largely distinct megadomain locations in different NUT carcinoma patient cell lines. To assess whether this was due to variations in individual genome sequences or epigenetic cell state, we expressed BRD4-NUT in a human stem cell model and found that megadomains formed in dissimilar patterns when comparing cells in the pluripotent state with the same cell line following induction along a mesodermal lineage. Thus, our work implicates initial cell state as the critical factor in the locations of BRD4-NUT megadomains. These results, together with our analysis of c-MYC protein-protein interactions in a patient cell line, are consistent with a cascade of chromatin misregulation underlying NUT carcinoma.

2.
Genetics ; 224(3)2023 Jul 06.
Article in English | MEDLINE | ID: mdl-37119804

ABSTRACT

Aberrant transcriptional programming and chromatin dysregulation are common to most cancers. Whether by deranged cell signaling or environmental insult, the resulting oncogenic phenotype is typically manifested in transcriptional changes characteristic of undifferentiated cell growth. Here we analyze targeting of an oncogenic fusion protein, BRD4-NUT, composed of 2 normally independent chromatin regulators. The fusion causes the formation of large hyperacetylated genomic regions or megadomains, mis-regulation of c-MYC, and an aggressive carcinoma of squamous cell origin. Our previous work revealed largely distinct megadomain locations in different NUT carcinoma patient cell lines. To assess whether this was due to variations in individual genome sequences or epigenetic cell state, we expressed BRD4-NUT in a human stem cell model and found that megadomains formed in dissimilar patterns when comparing cells in the pluripotent state with the same cell line following induction along a mesodermal lineage. Thus, our work implicates initial cell state as the critical factor in the locations of BRD4-NUT megadomains. These results, together with our analysis of c-MYC protein-protein interactions in a patient cell line, are consistent with a cascade of chromatin misregulation underlying NUT carcinoma.


Subject(s)
Carcinoma , Chromatin , Humans , Chromatin/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Cell Line, Tumor , Carcinoma/genetics , Carcinoma/pathology , Cell Cycle Proteins/genetics
3.
Mol Cancer Res ; 19(11): 1818-1830, 2021 11.
Article in English | MEDLINE | ID: mdl-34285087

ABSTRACT

NUT carcinoma (NC), characterized most commonly by the BRD4-NUTM1 fusion, is a rare, aggressive variant of squamous carcinoma with no effective treatment. BRD4-NUT drives growth and maintains the poorly differentiated state of NC by activating pro-growth genes such as MYC, through the formation of massive, hyperacetylated, superenhancer-like domains termed megadomains. BRD4-NUT-mediated hyperacetylation of chromatin is facilitated by the chromatin-targeting tandem bromodomains of BRD4, combined with NUT, which recruits the histone acetyltransferase, p300. Here, we developed a high-throughput small-molecule screen to identify inhibitors of transcriptional activation by NUT. In this dCAS9-based GFP-reporter assay, the strongest hits were diverse histone deacetylase (HDAC) inhibitors. Two structurally unrelated HDAC inhibitors, panobinostat and the novel compound, IRBM6, both repressed growth and induced differentiation of NC cells in proportion to their inhibition of NUT transcriptional activity. These two compounds repressed transcription of megadomain-associated oncogenic genes, such as MYC and SOX2, while upregulating pro-differentiation, non-megadomain-associated genes, including JUN, FOS, and key cell-cycle regulators, such as CDKN1A. The transcriptional changes correlate with depletion of BRD4-NUT from megadomains, and redistribution of the p300/CBP-associated chromatin acetylation mark, H3K27ac, away from megadomains toward regular enhancer regions previously populated by H3K27ac. In NC xenograft models, we demonstrated that suppression of tumor growth by panobinostat was comparable with that of bromodomain inhibition, and when combined they improved both survival and growth suppression. IMPLICATIONS: The findings provide mechanistic and preclinical rationale for the use of HDAC inhibitors, alone or combined with other agents, in the treatment of NUT carcinoma.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Cell Cycle Proteins/genetics , Early Detection of Cancer/methods , High-Throughput Nucleotide Sequencing/methods , Histone Deacetylase Inhibitors/therapeutic use , Nuclear Proteins/genetics , Transcription Factors/genetics , Animals , Cell Line, Tumor , Female , Histone Deacetylase Inhibitors/pharmacology , Humans , Male , Mice
4.
Mol Cancer Res ; 16(12): 1826-1833, 2018 12.
Article in English | MEDLINE | ID: mdl-30139738

ABSTRACT

Nuclear protein in testis (NUT) carcinoma (NC) is a rare, distinctly aggressive subtype of squamous carcinoma defined by the presence of NUT-fusion oncogenes resulting from chromosomal translocation. In most cases, the NUT gene (NUTM1) is fused to bromodomain containing 4 (BRD4) forming the BRD4-NUT oncogene. Here, a novel fusion partner to NUT was discovered using next-generation sequencing and FISH from a young patient with an undifferentiated malignant round cell tumor. Interestingly, the NUT fusion identified involved ZNF592, a zinc finger containing protein, which was previously identified as a component of the BRD4-NUT complex. In BRD4-NUT-expressing NC cells, wild-type ZNF592 and other associated "Z4" complex proteins, including ZNF532 and ZMYND8, colocalize with BRD4-NUT in characteristic nuclear foci. Furthermore, ectopic expression of BRD4-NUT in a non-NC cell line induces sequestration of Z4 factors to BRD4-NUT foci. Finally, the data demonstrate the specific dependency of NC cells on Z4 modules, ZNF532 and ZNF592. IMPLICATIONS: This study establishes the oncogenic role of Z4 factors in NC, offering potential new targeted therapeutic strategies in this incurable cancer.Visual Overview: http://mcr.aacrjournals.org/content/molcanres/16/12/1826/F1.large.jpg.


Subject(s)
Carcinoma, Squamous Cell/genetics , DNA-Binding Proteins/genetics , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Oncogene Proteins, Fusion/genetics , Tumor Suppressor Proteins/metabolism , Adolescent , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cell Survival , DNA-Binding Proteins/metabolism , Female , High-Throughput Nucleotide Sequencing , Humans , In Situ Hybridization, Fluorescence , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , Sequence Analysis, DNA
5.
Proc Natl Acad Sci U S A ; 114(21): E4184-E4192, 2017 05 23.
Article in English | MEDLINE | ID: mdl-28484033

ABSTRACT

To investigate the mechanism that drives dramatic mistargeting of active chromatin in NUT midline carcinoma (NMC), we have identified protein interactions unique to the BRD4-NUT fusion oncoprotein compared with wild-type BRD4. Using cross-linking, affinity purification, and mass spectrometry, we identified the EP300 acetyltransferase as uniquely associated with BRD4 through the NUT fusion in both NMC and non-NMC cell types. We also discovered ZNF532 associated with BRD4-NUT in NMC patient cells but not detectable in 293T cells. EP300 and ZNF532 are both implicated in feed-forward regulatory loops leading to propagation of the oncogenic chromatin complex in BRD4-NUT patient cells. Adding key functional significance to our biochemical findings, we independently discovered a ZNF532-NUT translocation fusion in a newly diagnosed NMC patient. ChIP sequencing of the major players NUT, ZNF532, BRD4, EP300, and H3K27ac revealed the formation of ZNF532-NUT-associated hyperacetylated megadomains, distinctly localized but otherwise analogous to those found in BRD4-NUT patient cells. Our results support a model in which NMC is dependent on ectopic NUT-mediated interactions between EP300 and components of BRD4 regulatory complexes, leading to a cascade of misregulation.


Subject(s)
Carcinoma, Squamous Cell/pathology , Chromatin/metabolism , E1A-Associated p300 Protein/metabolism , Lung Neoplasms/pathology , Nuclear Proteins/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Carcinoma, Squamous Cell/genetics , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation/genetics , Epithelial Cells/pathology , Female , HEK293 Cells , Humans , In Situ Hybridization, Fluorescence , Lung Neoplasms/genetics , Middle Aged , Multiprotein Complexes/genetics , Neoplasm Proteins , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Protein Domains/genetics , RNA Interference , RNA, Small Interfering/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , Zinc Fingers/genetics
6.
PLoS One ; 11(10): e0163820, 2016.
Article in English | MEDLINE | ID: mdl-27698495

ABSTRACT

Defects in chromatin proteins frequently manifest in diseases. A striking case of a chromatin-centric disease is NUT-midline carcinoma (NMC), which is characterized by expression of NUT as a fusion partner most frequently with BRD4. ChIP-sequencing studies from NMC patients revealed that BRD4-NUT (B4N) covers large genomic regions and elevates transcription within these domains. To investigate how B4N modulates chromatin, we performed affinity purification of B4N when ectopically expressed in 293-TREx cells and quantified the associated histone posttranslational modifications (PTM) using proteomics. We observed significant enrichment of acetylation particularly on H3 K18 and of combinatorial patterns such as H3 K27 acetylation paired with K36 methylation. We postulate that B4N complexes override the preexisting histone code with new PTM patterns that reflect aberrant transcription and that epigenetically modulate the nucleosome environment toward the NMC state.


Subject(s)
Carcinoma, Squamous Cell/genetics , DNA Methylation/genetics , Histones/genetics , Nuclear Proteins/genetics , Oncogene Proteins, Fusion/genetics , Acetylation , Carcinoma, Squamous Cell/pathology , Cell Differentiation/genetics , Cell Line, Tumor , Chromatin/genetics , Genome, Human , Histone Code/genetics , Histones/metabolism , Humans , Nuclear Proteins/metabolism , Nucleosomes/genetics , Oncogene Proteins, Fusion/metabolism , Proteomics , Transcription, Genetic
7.
Proc Natl Acad Sci U S A ; 113(7): 1784-9, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26831069

ABSTRACT

Posttranslational modifications (PTMs) are key contributors to chromatin function. The ability to comprehensively link specific histone PTMs with specific chromatin factors would be an important advance in understanding the functions and genomic targeting mechanisms of those factors. We recently introduced a cross-linked affinity technique, BioTAP-XL, to identify chromatin-bound protein interactions that can be difficult to capture with native affinity techniques. However, BioTAP-XL was not strictly compatible with similarly comprehensive analyses of associated histone PTMs. Here we advance BioTAP-XL by demonstrating the ability to quantify histone PTMs linked to specific chromatin factors in parallel with the ability to identify nonhistone binding partners. Furthermore we demonstrate that the initially published quantity of starting material can be scaled down orders of magnitude without loss in proteomic sensitivity. We also integrate hydrophilic interaction chromatography to mitigate detergent carryover and improve liquid chromatography-mass spectrometric performance. In summary, we greatly extend the practicality of BioTAP-XL to enable comprehensive identification of protein complexes and their local chromatin environment.


Subject(s)
Chromatin/chemistry , Histones/chemistry , Mass Spectrometry/methods , Animals , Chromatography, Liquid , Drosophila , Humans , Proteomics
8.
Genes Dev ; 29(14): 1507-23, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-26220994

ABSTRACT

NUT midline carcinoma (NMC), a subtype of squamous cell cancer, is one of the most aggressive human solid malignancies known. NMC is driven by the creation of a translocation oncoprotein, BRD4-NUT, which blocks differentiation and drives growth of NMC cells. BRD4-NUT forms distinctive nuclear foci in patient tumors, which we found correlate with ∼100 unprecedented, hyperacetylated expanses of chromatin that reach up to 2 Mb in size. These "megadomains" appear to be the result of aberrant, feed-forward loops of acetylation and binding of acetylated histones that drive transcription of underlying DNA in NMC patient cells and naïve cells induced to express BRD4-NUT. Megadomain locations are typically cell lineage-specific; however, the cMYC and TP63 regions are targeted in all NMCs tested and play functional roles in tumor growth. Megadomains appear to originate from select pre-existing enhancers that progressively broaden but are ultimately delimited by topologically associating domain (TAD) boundaries. Therefore, our findings establish a basis for understanding the powerful role played by large-scale chromatin organization in normal and aberrant lineage-specific gene transcription.


Subject(s)
Carcinoma, Squamous Cell/physiopathology , Gene Expression Regulation, Neoplastic , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Transcription Factors/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Enhancer Elements, Genetic , Humans , Neoplasm Proteins , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Protein Structure, Tertiary , Transcription Factors/genetics
9.
Genes Dev ; 29(11): 1136-50, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-26063573

ABSTRACT

The Polycomb group (PcG) proteins are key regulators of development in Drosophila and are strongly implicated in human health and disease. How PcG complexes form repressive chromatin domains remains unclear. Using cross-linked affinity purifications of BioTAP-Polycomb (Pc) or BioTAP-Enhancer of zeste [E(z)], we captured all PcG-repressive complex 1 (PRC1) or PRC2 core components and Sex comb on midleg (Scm) as the only protein strongly enriched with both complexes. Although previously not linked to PRC2, we confirmed direct binding of Scm and PRC2 using recombinant protein expression and colocalization of Scm with PRC1, PRC2, and H3K27me3 in embryos and cultured cells using ChIP-seq (chromatin immunoprecipitation [ChIP] combined with deep sequencing). Furthermore, we found that RNAi knockdown of Scm and overexpression of the dominant-negative Scm-SAM (sterile α motif) domain both affected the binding pattern of E(z) on polytene chromosomes. Aberrant localization of the Scm-SAM domain in long contiguous regions on polytene chromosomes revealed its independent ability to spread on chromatin, consistent with its previously described ability to oligomerize in vitro. Pull-downs of BioTAP-Scm captured PRC1 and PRC2 and additional repressive complexes, including PhoRC, LINT, and CtBP. We propose that Scm is a key mediator connecting PRC1, PRC2, and transcriptional silencing. Combined with previous structural and genetic analyses, our results strongly suggest that Scm coordinates PcG complexes and polymerizes to produce broad domains of PcG silencing.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Polycomb-Group Proteins/metabolism , Animals , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Histones/metabolism , Polycomb-Group Proteins/genetics , Polytene Chromosomes/metabolism , Protein Binding , Protein Transport , Repressor Proteins/metabolism
10.
Curr Protoc Mol Biol ; 109: 21.30.1-21.30.32, 2015 Jan 05.
Article in English | MEDLINE | ID: mdl-25559106

ABSTRACT

In order to understand how chromatin complexes function in the nucleus, it is important to obtain a comprehensive picture of their protein, DNA, and RNA components, as well as their mutual interactions. This unit presents a chromatin cross-linking approach (BioTAP-XL) that utilizes a special BioTAP-tagged transgenic protein bait along with mass spectrometry to identify protein complex components, and high-throughput sequencing to identify RNA components and DNA binding sites. Full protocols are provided for Drosophila cells and for human cells in culture, along with an additional protocol for Drosophila embryos as the source material. A key element of the approach in all cases is the generation of control data from input chromatin samples.


Subject(s)
Chromatin/metabolism , Cytological Techniques/methods , DNA/metabolism , Molecular Biology/methods , Proteins/metabolism , RNA/metabolism , Animals , Binding Sites , Cell Line , Chromatin/genetics , DNA/genetics , Drosophila , Humans
11.
Nature ; 512(7515): 449-52, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-25164756

ABSTRACT

Genome function is dynamically regulated in part by chromatin, which consists of the histones, non-histone proteins and RNA molecules that package DNA. Studies in Caenorhabditis elegans and Drosophila melanogaster have contributed substantially to our understanding of molecular mechanisms of genome function in humans, and have revealed conservation of chromatin components and mechanisms. Nevertheless, the three organisms have markedly different genome sizes, chromosome architecture and gene organization. On human and fly chromosomes, for example, pericentric heterochromatin flanks single centromeres, whereas worm chromosomes have dispersed heterochromatin-like regions enriched in the distal chromosomal 'arms', and centromeres distributed along their lengths. To systematically investigate chromatin organization and associated gene regulation across species, we generated and analysed a large collection of genome-wide chromatin data sets from cell lines and developmental stages in worm, fly and human. Here we present over 800 new data sets from our ENCODE and modENCODE consortia, bringing the total to over 1,400. Comparison of combinatorial patterns of histone modifications, nuclear lamina-associated domains, organization of large-scale topological domains, chromatin environment at promoters and enhancers, nucleosome positioning, and DNA replication patterns reveals many conserved features of chromatin organization among the three organisms. We also find notable differences in the composition and locations of repressive chromatin. These data sets and analyses provide a rich resource for comparative and species-specific investigations of chromatin composition, organization and function.


Subject(s)
Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , Chromatin/genetics , Chromatin/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Animals , Cell Line , Centromere/genetics , Centromere/metabolism , Chromatin/chemistry , Chromatin Assembly and Disassembly/genetics , DNA Replication/genetics , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic , Heterochromatin/chemistry , Heterochromatin/genetics , Heterochromatin/metabolism , Histones/chemistry , Histones/metabolism , Humans , Molecular Sequence Annotation , Nuclear Lamina/metabolism , Nucleosomes/chemistry , Nucleosomes/genetics , Nucleosomes/metabolism , Promoter Regions, Genetic/genetics , Species Specificity
12.
Genes Dev ; 28(13): 1445-60, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24990964

ABSTRACT

Heterochromatin protein 1 (HP1a) has conserved roles in gene silencing and heterochromatin and is also implicated in transcription, DNA replication, and repair. Here we identify chromatin-associated protein and RNA interactions of HP1a by BioTAP-XL mass spectrometry and sequencing from Drosophila S2 cells, embryos, larvae, and adults. Our results reveal an extensive list of known and novel HP1a-interacting proteins, of which we selected three for validation. A strong novel interactor, dADD1 (Drosophila ADD1) (CG8290), is highly enriched in heterochromatin, harbors an ADD domain similar to human ATRX, displays selective binding to H3K9me2 and H3K9me3, and is a classic genetic suppressor of position-effect variegation. Unexpectedly, a second hit, HIPP1 (HP1 and insulator partner protein-1) (CG3680), is strongly connected to CP190-related complexes localized at putative insulator sequences throughout the genome in addition to its colocalization with HP1a in heterochromatin. A third interactor, the histone methyltransferase MES-4, is also enriched in heterochromatin. In addition to these protein-protein interactions, we found that HP1a selectively associated with a broad set of RNAs transcribed from repetitive regions. We propose that this rich network of previously undiscovered interactions will define how HP1a complexes perform their diverse functions in cells and developing organisms.


Subject(s)
Carrier Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Heterochromatin/metabolism , RNA/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Animals , Carrier Proteins/genetics , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Life Cycle Stages/physiology , Protein Binding , RNA/genetics , Sequence Analysis, RNA , Sterol Regulatory Element Binding Protein 1/genetics
13.
Proc Natl Acad Sci U S A ; 111(7): 2488-93, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24550272

ABSTRACT

Understanding the composition of epigenetic regulators remains an important challenge in chromatin biology. Traditional biochemical analysis of chromatin-associated complexes requires their release from DNA under conditions that can also disrupt key interactions. Here we develop a complementary approach (BioTAP-XL), in which cross-linking (XL) enhances the preservation of protein interactions and also allows the analysis of DNA targets under the same tandem affinity purification (BioTAP) regimen. We demonstrate the power of BioTAP-XL through analysis of human EZH2, a core subunit of polycomb repressive complex 2 (PRC2). We identify and validate two strong interactors, C10orf12 and C17orf96, which display enrichment with EZH2-BioTAP at levels similar to canonical PRC2 components (SUZ12, EED, MTF2, JARID2, PHF1, and AEBP2). ChIP-seq analysis of BioTAP-tagged C10orf12 or C17orf96 revealed the similarity of each binding pattern with the location of EZH2 and the H3K27me3-silencing mark, validating their physical interaction with PRC2 components. Interestingly, analysis by mass spectrometry of C10orf12 and C17orf96 interactions revealed that these proteins may be mutually exclusive PRC2 subunits that fail to interact with each other or with JARID2 and AEBP2. C10orf12, in addition, shows a strong and unexpected association with components of the EHMT1/2 complex, thus potentially connecting PRC2 to another histone methyltransferase. Similarly, results from CBX4-BioTAP protein pulldowns are consistent with reports of a diversity of PRC1 complexes. Our results highlight the importance of reciprocal analyses of multiple subunits and suggest that iterative use of BioTAP-XL has strong potential to reveal networks of chromatin-based interactions in higher organisms.


Subject(s)
DNA-Binding Proteins/metabolism , Genetic Variation , Nerve Tissue Proteins/metabolism , Polycomb Repressive Complex 2/isolation & purification , Polycomb Repressive Complex 2/metabolism , Polycomb-Group Proteins/metabolism , Proteins/metabolism , Chromatin Immunoprecipitation , Chromatography, Liquid , Chromosomal Proteins, Non-Histone , Cross-Linking Reagents/metabolism , DNA-Binding Proteins/genetics , Enhancer of Zeste Homolog 2 Protein , Formaldehyde/metabolism , HEK293 Cells , Humans , Nerve Tissue Proteins/genetics , Polycomb-Group Proteins/genetics , Proteins/genetics , Repressor Proteins , Sequence Analysis, RNA , Tandem Mass Spectrometry
14.
Nat Struct Mol Biol ; 21(2): 118-25, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24500429

ABSTRACT

Males and females of many animal species differ in their sex-chromosome karyotype, and this creates imbalances between X-chromosome and autosomal gene products that require compensation. Although distinct molecular mechanisms have evolved in three highly studied systems, they all achieve coordinate regulation of an entire chromosome by differential RNA-polymerase occupancy at X-linked genes. High-throughput genome-wide methods have been pivotal in driving the latest progress in the field. Here we review the emerging models for dosage compensation in mammals, flies and nematodes, with a focus on mechanisms affecting RNA polymerase II activity on the X chromosome.


Subject(s)
Chromosomes/genetics , Dosage Compensation, Genetic , Gene Expression Regulation , Models, Genetic , Animals , Caenorhabditis elegans/genetics , Drosophila melanogaster/genetics , Gene Silencing , Genomics , Mammals/genetics , X Chromosome Inactivation
15.
Cell Rep ; 5(3): 629-36, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24183666

ABSTRACT

Dosage compensation in Drosophila is mediated by the MSL complex, which increases male X-linked gene expression approximately 2-fold. The MSL complex preferentially binds the bodies of active genes on the male X, depositing H4K16ac with a 3' bias. Two models have been proposed for the influence of the MSL complex on transcription: one based on promoter recruitment of RNA polymerase II (Pol II), and a second featuring enhanced transcriptional elongation. Here, we utilize nascent RNA sequencing to document dosage compensation during transcriptional elongation. We also compare X and autosomes from published data on paused and elongating polymerase in order to assess the role of Pol II recruitment. Our results support a model for differentially regulated elongation, starting with release from 5' pausing and increasing through X-linked gene bodies. Our results highlight facilitated transcriptional elongation as a key mechanism for the coordinated regulation of a diverse set of genes.


Subject(s)
Dosage Compensation, Genetic , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , X Chromosome , Animals , Cells, Cultured , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Female , Male , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
16.
PLoS Biol ; 11(11): e1001711, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24265597

ABSTRACT

Sex chromosomes originated from autosomes but have evolved a highly specialized chromatin structure. Drosophila Y chromosomes are composed entirely of silent heterochromatin, while male X chromosomes have highly accessible chromatin and are hypertranscribed as a result of dosage compensation. Here, we dissect the molecular mechanisms and functional pressures driving heterochromatin formation and dosage compensation of the recently formed neo-sex chromosomes of Drosophila miranda. We show that the onset of heterochromatin formation on the neo-Y is triggered by an accumulation of repetitive DNA. The neo-X has evolved partial dosage compensation and we find that diverse mutational paths have been utilized to establish several dozen novel binding consensus motifs for the dosage compensation complex on the neo-X, including simple point mutations at pre-binding sites, insertion and deletion mutations, microsatellite expansions, or tandem amplification of weak binding sites. Spreading of these silencing or activating chromatin modifications to adjacent regions results in massive mis-expression of neo-sex linked genes, and little correspondence between functionality of genes and their silencing on the neo-Y or dosage compensation on the neo-X. Intriguingly, the genomic regions being targeted by the dosage compensation complex on the neo-X and those becoming heterochromatic on the neo-Y show little overlap, possibly reflecting different propensities along the ancestral chromosome that formed the sex chromosome to adopt active or repressive chromatin configurations. Our findings have broad implications for current models of sex chromosome evolution, and demonstrate how mechanistic constraints can limit evolutionary adaptations. Our study also highlights how evolution can follow predictable genetic trajectories, by repeatedly acquiring the same 21-bp consensus motif for recruitment of the dosage compensation complex, yet utilizing a diverse array of random mutational changes to attain the same phenotypic outcome.


Subject(s)
Dosage Compensation, Genetic , Drosophila/genetics , Heterochromatin/genetics , Sex Chromosomes/genetics , Animals , Base Sequence , Binding Sites , Evolution, Molecular , Female , Gene Expression , Male , Molecular Sequence Annotation , Polymorphism, Single Nucleotide
17.
Genes Dev ; 27(8): 853-8, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23630075

ABSTRACT

Dosage compensation has arisen in response to the evolution of distinct male (XY) and female (XX) karyotypes. In Drosophila melanogaster, the MSL complex increases male X transcription approximately twofold. X-specific targeting is thought to occur through sequence-dependent binding to chromatin entry sites (CESs), followed by spreading in cis to active genes. We tested this model by asking how newly evolving sex chromosome arms in Drosophila miranda acquired dosage compensation. We found evidence for the creation of new CESs, with the analogous sequence and spacing as in D. melanogaster, providing strong support for the spreading model in the establishment of dosage compensation.


Subject(s)
Dosage Compensation, Genetic , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Sex Chromosomes/genetics , Animals , Evolution, Molecular , Female , Karyotype , Male , Molecular Sequence Data , Sex Chromosomes/metabolism
18.
Nat Struct Mol Biol ; 20(2): 202-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23295261

ABSTRACT

X-chromosome dosage compensation by the MSL (male-specific lethal) complex is required in Drosophila melanogaster to increase gene expression from the single male X to equal that of both female X chromosomes. Instead of focusing solely on protein complexes released from DNA, here we used chromatin-interacting protein MS (ChIP-MS) to identify MSL interactions on cross-linked chromatin. We identified MSL-enriched histone modifications, including histone H4 Lys16 acetylation and histone H3 Lys36 methylation, and CG4747, a putative Lys36-trimethylated histone H3 (H3K36me3)-binding protein. CG4747 is associated with the bodies of active genes, coincident with H3K36me3, and is mislocalized in the Set2 mutant lacking H3K36me3. CG4747 loss of function in vivo results in partial mislocalization of the MSL complex to autosomes, and RNA interference experiments confirm that CG4747 and Set2 function together to facilitate targeting of the MSL complex to active genes, validating the ChIP-MS approach.


Subject(s)
Dosage Compensation, Genetic/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Histones/metabolism , Mass Spectrometry/methods , Oxidoreductases/metabolism , Acetylation , Animals , Animals, Genetically Modified , Blotting, Western , Chromatin Immunoprecipitation , Female , Histone-Lysine N-Methyltransferase/metabolism , Male , Methylation , Nuclear Proteins , RNA Interference
19.
PLoS Genet ; 8(9): e1002954, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23028361

ABSTRACT

Chromatin environments differ greatly within a eukaryotic genome, depending on expression state, chromosomal location, and nuclear position. In genomic regions characterized by high repeat content and high gene density, chromatin structure must silence transposable elements but permit expression of embedded genes. We have investigated one such region, chromosome 4 of Drosophila melanogaster. Using chromatin-immunoprecipitation followed by microarray (ChIP-chip) analysis, we examined enrichment patterns of 20 histone modifications and 25 chromosomal proteins in S2 and BG3 cells, as well as the changes in several marks resulting from mutations in key proteins. Active genes on chromosome 4 are distinct from those in euchromatin or pericentric heterochromatin: while there is a depletion of silencing marks at the transcription start sites (TSSs), HP1a and H3K9me3, but not H3K9me2, are enriched strongly over gene bodies. Intriguingly, genes on chromosome 4 are less frequently associated with paused polymerase. However, when the chromatin is altered by depleting HP1a or POF, the RNA pol II enrichment patterns of many chromosome 4 genes shift, showing a significant decrease over gene bodies but not at TSSs, accompanied by lower expression of those genes. Chromosome 4 genes have a low incidence of TRL/GAGA factor binding sites and a low T(m) downstream of the TSS, characteristics that could contribute to a low incidence of RNA polymerase pausing. Our data also indicate that EGG and POF jointly regulate H3K9 methylation and promote HP1a binding over gene bodies, while HP1a targeting and H3K9 methylation are maintained at the repeats by an independent mechanism. The HP1a-enriched, POF-associated chromatin structure over the gene bodies may represent one type of adaptation for genes embedded in repetitive DNA.


Subject(s)
Chromosomal Proteins, Non-Histone , Heterochromatin/genetics , Histone-Lysine N-Methyltransferase , Histones , Animals , Animals, Genetically Modified , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomes/metabolism , DNA-Directed RNA Polymerases/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , Euchromatin/metabolism , Gene Expression Regulation/genetics , Heterochromatin/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Histones/metabolism , Humans , Methylation , Mutation
20.
Genome Res ; 22(11): 2188-98, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22767387

ABSTRACT

Chromatin insulator elements and associated proteins have been proposed to partition eukaryotic genomes into sets of independently regulated domains. Here we test this hypothesis by quantitative genome-wide analysis of insulator protein binding to Drosophila chromatin. We find distinct combinatorial binding of insulator proteins to different classes of sites and uncover a novel type of insulator element that binds CP190 but not any other known insulator proteins. Functional characterization of different classes of binding sites indicates that only a small fraction act as robust insulators in standard enhancer-blocking assays. We show that insulators restrict the spreading of the H3K27me3 mark but only at a small number of Polycomb target regions and only to prevent repressive histone methylation within adjacent genes that are already transcriptionally inactive. RNAi knockdown of insulator proteins in cultured cells does not lead to major alterations in genome expression. Taken together, these observations argue against the concept of a genome partitioned by specialized boundary elements and suggest that insulators are reserved for specific regulation of selected genes.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Genome, Insect , Insulator Elements , Microtubule-Associated Proteins/metabolism , Nuclear Proteins/metabolism , Animals , Binding Sites , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Epigenesis, Genetic , Histones/metabolism , Methylation , Microtubule-Associated Proteins/genetics , Nuclear Proteins/genetics , Polycomb-Group Proteins/metabolism , Protein Processing, Post-Translational , RNA, Small Interfering , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...