Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Nat Commun ; 14(1): 724, 2023 02 09.
Article in English | MEDLINE | ID: mdl-36759620

ABSTRACT

The PML::RARA fusion protein is the hallmark driver of Acute Promyelocytic Leukemia (APL) and disrupts retinoic acid signaling, leading to wide-scale gene expression changes and uncontrolled proliferation of myeloid precursor cells. While known to be recruited to binding sites across the genome, its impact on gene regulation and expression is under-explored. Using integrated multi-omics datasets, we characterize the influence of PML::RARA binding on gene expression and regulation in an inducible PML::RARA cell line model and APL patient ex vivo samples. We find that genes whose regulatory elements recruit PML::RARA are not uniformly transcriptionally repressed, as commonly suggested, but also may be upregulated or remain unchanged. We develop a computational machine learning implementation called Regulatory Element Behavior Extraction Learning to deconvolute the complex, local transcription factor binding site environment at PML::RARA bound positions to reveal distinct signatures that modulate how PML::RARA directs the transcriptional response.


Subject(s)
Leukemia, Promyelocytic, Acute , Humans , Cell Line , Gene Expression Regulation , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/metabolism , Multiomics , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Tretinoin/pharmacology
2.
Front Genet ; 13: 786501, 2022.
Article in English | MEDLINE | ID: mdl-35198004

ABSTRACT

It is now evident that DNA forms an organized nuclear architecture, which is essential to maintain the structural and functional integrity of the genome. Chromatin organization can be systematically studied due to the recent boom in chromosome conformation capture technologies (e.g., 3C and its successors 4C, 5C and Hi-C), which is accompanied by the development of computational pipelines to identify biologically meaningful chromatin contacts in such data. However, not all tools are applicable to all experimental designs and all structural features. Capture Hi-C (CHi-C) is a method that uses an intermediate hybridization step to target and select predefined regions of interest in a Hi-C library, thereby increasing effective sequencing depth for those regions. It allows researchers to investigate fine chromatin structures at high resolution, for instance promoter-enhancer loops, but it introduces additional biases with the capture step, and therefore requires specialized pipelines. Here, we compare multiple analytical pipelines for CHi-C data analysis. We consider the effect of retaining multi-mapping reads and compare the efficiency of different statistical approaches in both identifying reproducible interactions and determining biologically significant interactions. At restriction fragment level resolution, the number of multi-mapping reads that could be rescued was negligible. The number of identified interactions varied widely, depending on the analytical method, indicating large differences in type I and type II error rates. The optimal pipeline depends on the project-specific tolerance level of false positive and false negative chromatin contacts.

3.
Nat Genet ; 53(10): 1443-1455, 2021 10.
Article in English | MEDLINE | ID: mdl-34556857

ABSTRACT

Altered transcription is a cardinal feature of acute myeloid leukemia (AML); however, exactly how mutations synergize to remodel the epigenetic landscape and rewire three-dimensional DNA topology is unknown. Here, we apply an integrated genomic approach to a murine allelic series that models the two most common mutations in AML: Flt3-ITD and Npm1c. We then deconvolute the contribution of each mutation to alterations of the epigenetic landscape and genome organization, and infer how mutations synergize in the induction of AML. Our studies demonstrate that Flt3-ITD signals to chromatin to alter the epigenetic environment and synergizes with mutations in Npm1c to alter gene expression and drive leukemia induction. These analyses also allow the identification of long-range cis-regulatory circuits, including a previously unknown superenhancer of Hoxa locus, as well as larger and more detailed gene-regulatory networks, driven by transcription factors including PU.1 and IRF8, whose importance we demonstrate through perturbation of network members.


Subject(s)
Chromatin Assembly and Disassembly/genetics , DNA, Neoplasm/chemistry , Gene Expression Regulation, Leukemic , Histones/metabolism , Leukemia, Myeloid, Acute/genetics , Mutation/genetics , Protein Processing, Post-Translational , Animals , Base Sequence , Disease Models, Animal , Enhancer Elements, Genetic/genetics , Gene Regulatory Networks , Genetic Loci , Humans , Mice, Inbred C57BL , Nuclear Proteins/metabolism , Nucleophosmin , Principal Component Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic , fms-Like Tyrosine Kinase 3/metabolism
5.
Cell Rep ; 28(12): 3022-3031.e7, 2019 Sep 17.
Article in English | MEDLINE | ID: mdl-31533028

ABSTRACT

Acute myeloid leukemia (AML) is associated with mutations in transcriptional and epigenetic regulator genes impairing myeloid differentiation. The t(8;21)(q22;q22) translocation generates the RUNX1-ETO fusion protein, which interferes with the hematopoietic master regulator RUNX1. We previously showed that the maintenance of t(8;21) AML is dependent on RUNX1-ETO expression. Its depletion causes extensive changes in transcription factor binding, as well as gene expression, and initiates myeloid differentiation. However, how these processes are connected within a gene regulatory network is unclear. To address this question, we performed Promoter-Capture Hi-C assays, with or without RUNX1-ETO depletion and assigned interacting cis-regulatory elements to their respective genes. To construct a RUNX1-ETO-dependent gene regulatory network maintaining AML, we integrated cis-regulatory element interactions with gene expression and transcription factor binding data. This analysis shows that RUNX1-ETO participates in cis-regulatory element interactions. However, differential interactions following RUNX1-ETO depletion are driven by alterations in the binding of RUNX1-ETO-regulated transcription factors.


Subject(s)
CCAAT-Enhancer-Binding Proteins , Core Binding Factor Alpha 2 Subunit , Enhancer Elements, Genetic , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute , Oncogene Proteins, Fusion , Promoter Regions, Genetic , RUNX1 Translocation Partner 1 Protein , Transcription Factor AP-1 , Translocation, Genetic , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Chromosomes, Human, Pair 21/genetics , Chromosomes, Human, Pair 21/metabolism , Chromosomes, Human, Pair 8/genetics , Chromosomes, Human, Pair 8/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Gene Deletion , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , RUNX1 Translocation Partner 1 Protein/genetics , RUNX1 Translocation Partner 1 Protein/metabolism , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism
6.
Blood ; 134(24): 2195-2208, 2019 12 12.
Article in English | MEDLINE | ID: mdl-31515253

ABSTRACT

Cohesin complex disruption alters gene expression, and cohesin mutations are common in myeloid neoplasia, suggesting a critical role in hematopoiesis. Here, we explore cohesin dynamics and regulation of hematopoietic stem cell homeostasis and differentiation. Cohesin binding increases at active regulatory elements only during erythroid differentiation. Prior binding of the repressive Ets transcription factor Etv6 predicts cohesin binding at these elements and Etv6 interacts with cohesin at chromatin. Depletion of cohesin severely impairs erythroid differentiation, particularly at Etv6-prebound loci, but augments self-renewal programs. Together with corroborative findings in acute myeloid leukemia and myelodysplastic syndrome patient samples, these data suggest cohesin-mediated alleviation of Etv6 repression is required for dynamic expression at critical erythroid genes during differentiation and how this may be perturbed in myeloid malignancies.


Subject(s)
Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Gene Expression Regulation, Leukemic , Mutation , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Biomarkers, Tumor , Cell Line, Tumor , Female , Gene Dosage , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Histones/metabolism , Humans , Male , Myeloproliferative Disorders/diagnosis , Neoplasm Grading , Protein Binding , Proto-Oncogene Proteins c-ets/metabolism , Regulatory Sequences, Nucleic Acid , Repressor Proteins/metabolism , Cohesins , ETS Translocation Variant 6 Protein
7.
PLoS One ; 14(8): e0217532, 2019.
Article in English | MEDLINE | ID: mdl-31412036

ABSTRACT

Transcriptome analyses show a surprisingly large proportion of the mammalian genome is transcribed; much more than can be accounted for by genes and introns alone. Most of this transcription is non-coding in nature and arises from intergenic regions, often overlapping known protein-coding genes in sense or antisense orientation. The functional relevance of this widespread transcription is unknown. Here we characterize a promoter responsible for initiation of an intergenic transcript located approximately 3.3 kb and 10.7 kb upstream of the adult-specific human ß-globin genes. Mutational analyses in ß-YAC transgenic mice show that alteration of intergenic promoter activity results in ablation of H3K4 di- and tri-methylation and H3 hyperacetylation extending over a 30 kb region immediately downstream of the initiation site, containing the adult δ- and ß-globin genes. This results in dramatically decreased expression of the adult genes through position effect variegation in which the vast majority of definitive erythroid cells harbor inactive adult globin genes. In contrast, expression of the neighboring ε- and γ-globin genes is completely normal in embryonic erythroid cells, indicating a developmentally specific variegation of the adult domain. Our results demonstrate a role for intergenic non-coding RNA transcription in the propagation of histone modifications over chromatin domains and epigenetic control of ß-like globin gene transcription during development.


Subject(s)
Chromatin/genetics , DNA, Intergenic/genetics , Gene Expression Regulation, Developmental , Histones/chemistry , Promoter Regions, Genetic , RNA, Untranslated/genetics , beta-Globins/genetics , Adult , Animals , Chromosomes, Artificial, Yeast , Erythroid Cells/metabolism , Humans , Mice , Mice, Transgenic , Transcription, Genetic
8.
Nat Genet ; 50(10): 1375-1380, 2018 10.
Article in English | MEDLINE | ID: mdl-30224643

ABSTRACT

Efforts are being directed to systematically analyze the non-coding regions of the genome for cancer-driving mutations1-6. cis-regulatory elements (CREs) represent a highly enriched subset of the non-coding regions of the genome in which to search for such mutations. Here we use high-throughput chromosome conformation capture techniques (Hi-C) for 19,023 promoter fragments to catalog the regulatory landscape of colorectal cancer in cell lines, mapping CREs and integrating these with whole-genome sequence and expression data from The Cancer Genome Atlas7,8. We identify a recurrently mutated CRE interacting with the ETV1 promoter affecting gene expression. ETV1 expression influences cell viability and is associated with patient survival. We further refine our understanding of the regulatory effects of copy-number variations, showing that RASL11A is targeted by a previously identified enhancer amplification1. This study reveals new insights into the complex genetic alterations driving tumor development, providing a paradigm for employing chromosome conformation capture to decipher non-coding CREs relevant to cancer biology.


Subject(s)
Cell Transformation, Neoplastic/genetics , Chromosomes, Human/chemistry , Codon, Nonsense , Colorectal Neoplasms/genetics , Computational Biology/methods , High-Throughput Nucleotide Sequencing/methods , Caco-2 Cells , Colorectal Neoplasms/epidemiology , DNA, Neoplasm/chemistry , Databases, Genetic , Gene Frequency , HT29 Cells , HeLa Cells , Hep G2 Cells , Humans , K562 Cells , MCF-7 Cells , Nucleic Acid Conformation , Promoter Regions, Genetic/genetics , Regulatory Sequences, Nucleic Acid/genetics , Tumor Cells, Cultured
9.
Elife ; 62017 03 23.
Article in English | MEDLINE | ID: mdl-28332981

ABSTRACT

Long-range cis-regulatory elements such as enhancers coordinate cell-specific transcriptional programmes by engaging in DNA looping interactions with target promoters. Deciphering the interplay between the promoter connectivity and activity of cis-regulatory elements during lineage commitment is crucial for understanding developmental transcriptional control. Here, we use Promoter Capture Hi-C to generate a high-resolution atlas of chromosomal interactions involving ~22,000 gene promoters in human pluripotent and lineage-committed cells, identifying putative target genes for known and predicted enhancer elements. We reveal extensive dynamics of cis-regulatory contacts upon lineage commitment, including the acquisition and loss of promoter interactions. This spatial rewiring occurs preferentially with predicted changes in the activity of cis-regulatory elements and is associated with changes in target gene expression. Our results provide a global and integrated view of promoter interactome dynamics during lineage commitment of human pluripotent cells.


Subject(s)
Cell Differentiation , Gene Expression Regulation , Human Embryonic Stem Cells/physiology , Humans , Promoter Regions, Genetic , Regulatory Elements, Transcriptional , Transcription, Genetic
10.
Chromosome Res ; 25(1): 15-24, 2017 03.
Article in English | MEDLINE | ID: mdl-28078515

ABSTRACT

There is a strong interrelationship within the cell nucleus between form and function of the genome. This connection is exhibited across multiple hierarchies, ranging from grand-scale positioning of chromosomes and their intersection with specific nuclear functional activities, the segregation of chromosome structure into distinct domains and long-range regulatory contacts that drive spatial and temporal expression patterns of genes. Fifteen years ago, the development of the chromosome conformation capture method placed the nature of specific, long-range regulatory interactions under scrutiny. However, its development and integration with next-generation sequencing technologies has greatly expanded the breadth and scope of what is detected. The sheer scale of data offered by these important advances has come with new and challenging bottlenecks that are both experimental and bioinformatical. Here, we discuss the recent and prospective development and implementation of new methodologies and analytical tools that are allowing an in-depth, yet focussed characterisation of genomic contacts that are associated with functional activities in the nucleus.


Subject(s)
Chromosomes/ultrastructure , Genome/genetics , Genomics/methods , Animals , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Humans , Methods
11.
Elife ; 52016 08 04.
Article in English | MEDLINE | ID: mdl-27490482

ABSTRACT

Lymphomagenesis in the presence of deregulated MYC requires suppression of MYC-driven apoptosis, often through downregulation of the pro-apoptotic BCL2L11 gene (Bim). Transcription factors (EBNAs) encoded by the lymphoma-associated Epstein-Barr virus (EBV) activate MYC and silence BCL2L11. We show that the EBNA2 transactivator activates multiple MYC enhancers and reconfigures the MYC locus to increase upstream and decrease downstream enhancer-promoter interactions. EBNA2 recruits the BRG1 ATPase of the SWI/SNF remodeller to MYC enhancers and BRG1 is required for enhancer-promoter interactions in EBV-infected cells. At BCL2L11, we identify a haematopoietic enhancer hub that is inactivated by the EBV repressors EBNA3A and EBNA3C through recruitment of the H3K27 methyltransferase EZH2. Reversal of enhancer inactivation using an EZH2 inhibitor upregulates BCL2L11 and induces apoptosis. EBV therefore drives lymphomagenesis by hijacking long-range enhancer hubs and specific cellular co-factors. EBV-driven MYC enhancer activation may contribute to the genesis and localisation of MYC-Immunoglobulin translocation breakpoints in Burkitt's lymphoma.


Subject(s)
Bcl-2-Like Protein 11/metabolism , Epstein-Barr Virus Nuclear Antigens/metabolism , Gene Silencing , Herpesvirus 4, Human/enzymology , Herpesvirus 4, Human/physiology , Proto-Oncogene Proteins c-myc/metabolism , Transcriptional Activation , Bcl-2-Like Protein 11/genetics , DNA Helicases/metabolism , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-myc/genetics , Repressor Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism
12.
Nat Genet ; 47(10): 1179-1186, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26323060

ABSTRACT

The Polycomb repressive complexes PRC1 and PRC2 maintain embryonic stem cell (ESC) pluripotency by silencing lineage-specifying developmental regulator genes. Emerging evidence suggests that Polycomb complexes act through controlling spatial genome organization. We show that PRC1 functions as a master regulator of mouse ESC genome architecture by organizing genes in three-dimensional interaction networks. The strongest spatial network is composed of the four Hox gene clusters and early developmental transcription factor genes, the majority of which contact poised enhancers. Removal of Polycomb repression leads to disruption of promoter-promoter contacts in the Hox gene network. In contrast, promoter-enhancer contacts are maintained in the absence of Polycomb repression, with accompanying widespread acquisition of active chromatin signatures at network enhancers and pronounced transcriptional upregulation of network genes. Thus, PRC1 physically constrains developmental transcription factor genes and their enhancers in a silenced but poised spatial network. We propose that the selective release of genes from this spatial network underlies cell fate specification during early embryonic development.


Subject(s)
Embryonic Stem Cells/metabolism , Genome , Polycomb-Group Proteins/physiology , Animals , Mice , Promoter Regions, Genetic
13.
Nat Genet ; 47(6): 598-606, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25938943

ABSTRACT

Transcriptional control in large genomes often requires looping interactions between distal DNA elements, such as enhancers and target promoters. Current chromosome conformation capture techniques do not offer sufficiently high resolution to interrogate these regulatory interactions on a genomic scale. Here we use Capture Hi-C (CHi-C), an adapted genome conformation assay, to examine the long-range interactions of almost 22,000 promoters in 2 human blood cell types. We identify over 1.6 million shared and cell type-restricted interactions spanning hundreds of kilobases between promoters and distal loci. Transcriptionally active genes contact enhancer-like elements, whereas transcriptionally inactive genes interact with previously uncharacterized elements marked by repressive features that may act as long-range silencers. Finally, we show that interacting loci are enriched for disease-associated SNPs, suggesting how distal mutations may disrupt the regulation of relevant genes. This study provides new insights and accessible tools to dissect the regulatory interactions that underlie normal and aberrant gene regulation.


Subject(s)
Promoter Regions, Genetic , Cell Line , Chromosome Mapping , Epistasis, Genetic , Gene Expression Regulation , Gene Regulatory Networks , Genome, Human , Humans , Polymorphism, Single Nucleotide
14.
Genome Res ; 25(4): 582-97, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25752748

ABSTRACT

The mammalian genome harbors up to one million regulatory elements often located at great distances from their target genes. Long-range elements control genes through physical contact with promoters and can be recognized by the presence of specific histone modifications and transcription factor binding. Linking regulatory elements to specific promoters genome-wide is currently impeded by the limited resolution of high-throughput chromatin interaction assays. Here we apply a sequence capture approach to enrich Hi-C libraries for >22,000 annotated mouse promoters to identify statistically significant, long-range interactions at restriction fragment resolution, assigning long-range interacting elements to their target genes genome-wide in embryonic stem cells and fetal liver cells. The distal sites contacting active genes are enriched in active histone modifications and transcription factor occupancy, whereas inactive genes contact distal sites with repressive histone marks, demonstrating the regulatory potential of the distal elements identified. Furthermore, we find that coregulated genes cluster nonrandomly in spatial interaction networks correlated with their biological function and expression level. Interestingly, we find the strongest gene clustering in ES cells between transcription factor genes that control key developmental processes in embryogenesis. The results provide the first genome-wide catalog linking gene promoters to their long-range interacting elements and highlight the complex spatial regulatory circuitry controlling mammalian gene expression.


Subject(s)
Binding Sites/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental/genetics , Promoter Regions, Genetic/genetics , Animals , Chromatin/genetics , Embryonic Stem Cells/cytology , Epigenesis, Genetic , Histones/genetics , Liver/cytology , Liver/embryology , Mice , Mice, Inbred C57BL , Transcription Factors/genetics , Transcription Factors/metabolism
15.
Clin Cancer Res ; 20(2): 296-300, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24166911

ABSTRACT

The mammalian nucleus is a highly complex structure that carries out a diverse range of functions such as DNA replication, cell division, RNA processing, and nuclear export/import. Many of these activities occur at discrete subcompartments that intersect with specific regions of the genome. Over the past few decades, evidence has accumulated to suggest that RNA transcription also occurs in specialized sites, called transcription factories, that may influence how the genome is organized. There may be certain efficiency benefits to cluster transcriptional activity in this way. However, the clustering of genes at transcription factories may have consequences for genome stability, and increase the susceptibility to recurrent chromosomal translocations that lead to cancer. The relationships between genome organization, transcription, and chromosomal translocation formation will have important implications in understanding the causes of therapy-related cancers.


Subject(s)
Transcription Factors/metabolism , Translocation, Genetic , Animals , Humans , Neoplasms/etiology , Neoplasms/therapy
16.
PLoS One ; 7(7): e38983, 2012.
Article in English | MEDLINE | ID: mdl-22802932

ABSTRACT

Although somatic homologous pairing is common in Drosophila it is not generally observed in mammalian cells. However, a number of regions have recently been shown to come into close proximity with their homologous allele, and it has been proposed that pairing might be involved in the establishment or maintenance of monoallelic expression. Here, we investigate the pairing properties of various imprinted and non-imprinted regions in mouse tissues and ES cells. We find by allele-specific 4C-Seq and DNA FISH that the Kcnq1 imprinted region displays frequent pairing but that this is not dependent on monoallelic expression. We demonstrate that pairing involves larger chromosomal regions and that the two chromosome territories come close together. Frequent pairing is not associated with imprinted status or DNA repair, but is influenced by chromosomal location and transcription. We propose that homologous pairing is not exclusive to specialised regions or specific functional events, and speculate that it provides the cell with the opportunity of trans-allelic effects on gene regulation.


Subject(s)
Gene Expression Regulation , Genomic Imprinting , KCNQ1 Potassium Channel/genetics , Mice/genetics , Alleles , Animals , Chromosomes, Mammalian/genetics , Embryonic Stem Cells , In Situ Hybridization, Fluorescence
17.
PLoS One ; 6(1): e16607, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21305042

ABSTRACT

Massively parallel DNA sequencing is capable of sequencing tens of millions of DNA fragments at the same time. However, sequence bias in the initial cycles, which are used to determine the coordinates of individual clusters, causes a loss of fidelity in cluster identification on Illumina Genome Analysers. This can result in a significant reduction in the numbers of clusters that can be analysed. Such low sample diversity is an intrinsic problem of sequencing libraries that are generated by restriction enzyme digestion, such as e4C-seq or reduced-representation libraries. Similarly, this problem can also arise through the combined sequencing of barcoded, multiplexed libraries. We describe a procedure to defer the mapping of cluster coordinates until low-diversity sequences have been passed. This simple procedure can recover substantial amounts of next generation sequencing data that would otherwise be lost.


Subject(s)
Gene Library , Sequence Analysis, DNA/methods , Cluster Analysis , DNA Barcoding, Taxonomic , Data Collection , Restriction Mapping , Sequence Analysis, DNA/standards
18.
Brief Funct Genomics ; 10(1): 11-7, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21258046

ABSTRACT

The eukaryotic cell nucleus displays a high degree of spatial organization, with discrete functional subcompartments that provide microenvironments where specialized processes take place. Concordantly, the genome also adopts defined conformations that, in part, enable specific genomic regions to interface with these functional centers. Yet the roles of many subcompartments and the genomic regions that contact them have not been explored fully. More fundamentally, it is not entirely clear how genome organization impacts function, and vice versa. The past decade has witnessed the development of a new breed of methods that are capable of assessing the spatial organization of the genome. These stand to further our understanding of the relationship between genome structure and function, and potentially assign function to various nuclear subcompartments. Here, we review the principal techniques used for analyzing genomic interactions, the functional insights they have afforded and discuss the outlook for future advances in nuclear structure and function dynamics.


Subject(s)
Cell Nucleus/genetics , Cell Nucleus/metabolism , Genomics/methods , Animals , Chromatin Immunoprecipitation , DNA, Circular/genetics , Genome/genetics , Humans , Nucleic Acid Conformation
19.
J Cell Biochem ; 104(5): 1553-61, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18384074

ABSTRACT

A recent spate of examples of specific interactions between loci on separate chromosomes in mammalian nuclei has illuminated another layer of complexity in gene regulation. As the specifics of the cross-talk between interacting loci are worked out, it is also important to consider exactly how, when and where loci can ever reliably find each other within such an intricate environment. Answers may lie in how the genome is organised in relation to itself and to specialised nuclear sub-compartments. Here, we discuss how such specialised nuclear bodies may have the potential to specifically sequester loci and provide a context where interchromosomal communications can occur.


Subject(s)
Cell Nucleus/metabolism , Chromosome Positioning/physiology , Chromosomes/metabolism , Genome , Animals , Cell Nucleolus/metabolism , Humans , Transcription, Genetic
20.
PLoS Biol ; 5(8): e192, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17622196

ABSTRACT

Transcription in mammalian nuclei is highly compartmentalized in RNA polymerase II-enriched nuclear foci known as transcription factories. Genes in cis and trans can share the same factory, suggesting that genes migrate to preassembled transcription sites. We used fluorescent in situ hybridization to investigate the dynamics of gene association with transcription factories during immediate early (IE) gene induction in mouse B lymphocytes. Here, we show that induction involves rapid gene relocation to transcription factories. Importantly, we find that the Myc proto-oncogene on Chromosome 15 is preferentially recruited to the same transcription factory as the highly transcribed Igh gene located on Chromosome 12. Myc and Igh are the most frequent translocation partners in plasmacytoma and Burkitt lymphoma. Our results show that transcriptional activation of IE genes involves rapid relocation to preassembled transcription factories. Furthermore, the data imply a direct link between the nonrandom interchromosomal organization of transcribed genes at transcription factories and the incidence of specific chromosomal translocations.


Subject(s)
Gene Expression Regulation , Genes, Immediate-Early , Genes, Immunoglobulin Heavy Chain , Immunoglobulin Heavy Chains , Proto-Oncogene Proteins c-myc/metabolism , Transcription, Genetic , Alleles , Animals , B-Lymphocytes/cytology , B-Lymphocytes/physiology , Cell Nucleus/metabolism , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/metabolism , In Situ Hybridization, Fluorescence , Mice , Mice, Inbred BALB C , Neoplasms/genetics , Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/genetics , RNA Polymerase II/metabolism , Transcriptional Activation , Translocation, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...