Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
2.
Wellcome Open Res ; 8: 165, 2023.
Article in English | MEDLINE | ID: mdl-37736013

ABSTRACT

Background: Resolving causal genes for type 2 diabetes at loci implicated by genome-wide association studies (GWAS) requires integrating functional genomic data from relevant cell types. Chromatin features in endocrine cells of the pancreatic islet are particularly informative and recent studies leveraging chromosome conformation capture (3C) with Hi-C based methods have elucidated regulatory mechanisms in human islets. However, these genome-wide approaches are less sensitive and afford lower resolution than methods that target specific loci. Methods: To gauge the extent to which targeted 3C further resolves chromatin-mediated regulatory mechanisms at GWAS loci, we generated interaction profiles at 23 loci using next-generation (NG) capture-C in a human beta cell model (EndoC-ßH1) and contrasted these maps with Hi-C maps in EndoC-ßH1 cells and human islets and a promoter capture Hi-C map in human islets. Results: We found improvements in assay sensitivity of up to 33-fold and resolved ~3.6X more chromatin interactions. At a subset of 18 loci with 25 co-localised GWAS and eQTL signals, NG Capture-C interactions implicated effector transcripts at five additional genetic signals relative to promoter capture Hi-C through physical contact with gene promoters. Conclusions: High resolution chromatin interaction profiles at selectively targeted loci can complement genome- and promoter-wide maps.

4.
Nat Protoc ; 18(6): 1687-1711, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36991220

ABSTRACT

Micro Capture-C (MCC) is a chromatin conformation capture (3C) method for visualizing reproducible three-dimensional contacts of specified regions of the genome at base pair resolution. These methods are an established family of techniques that use proximity ligation to assay the topology of chromatin. MCC can generate data at substantially higher resolution than previous techniques through multiple refinements of the 3C method. Using a sequence agnostic nuclease, the maintenance of cellular integrity and full sequencing of the ligation junctions, MCC achieves subnucleosomal levels of resolution, which can be used to reveal transcription factor binding sites analogous to DNAse I footprinting. Gene dense regions, close-range enhancer-promoter contacts, individual enhancers within super-enhancers and multiple other types of loci or regulatory regions that were previously challenging to assay with conventional 3C techniques, are readily observed using MCC. MCC requires training in common molecular biology techniques and bioinformatics to perform the experiment and analyze the data. The protocol can be expected to be completed in a 3 week timeframe for experienced molecular biologists.


Subject(s)
Chromatin , Chromosomes , Chromatin/genetics , Genome , Computational Biology/methods , Regulatory Sequences, Nucleic Acid
5.
Bioinformatics ; 38(18): 4255-4263, 2022 09 15.
Article in English | MEDLINE | ID: mdl-35866989

ABSTRACT

MOTIVATION: Genome sequencing experiments have revolutionized molecular biology by allowing researchers to identify important DNA-encoded elements genome wide. Regions where these elements are found appear as peaks in the analog signal of an assay's coverage track, and despite the ease with which humans can visually categorize these patterns, the size of many genomes necessitates algorithmic implementations. Commonly used methods focus on statistical tests to classify peaks, discounting that the background signal does not completely follow any known probability distribution and reducing the information-dense peak shapes to simply maximum height. Deep learning has been shown to be highly accurate for many pattern recognition tasks, on par or even exceeding human capabilities, providing an opportunity to reimagine and improve peak calling. RESULTS: We present the peak calling framework LanceOtron, which combines deep learning for recognizing peak shape with multifaceted enrichment calculations for assessing significance. In benchmarking ATAC-seq, ChIP-seq and DNase-seq, LanceOtron outperforms long-standing, gold-standard peak callers through its improved selectivity and near-perfect sensitivity. AVAILABILITY AND IMPLEMENTATION: A fully featured web application is freely available from LanceOtron.molbiol.ox.ac.uk, command line interface via python is pip installable from PyPI at https://pypi.org/project/lanceotron/, and source code and benchmarking tests are available at https://github.com/LHentges/LanceOtron. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Deep Learning , Humans , Sequence Analysis, DNA/methods , Software , Chromatin Immunoprecipitation Sequencing , Base Sequence , High-Throughput Nucleotide Sequencing/methods
6.
Methods Mol Biol ; 2532: 95-112, 2022.
Article in English | MEDLINE | ID: mdl-35867247

ABSTRACT

Tri-C is a chromosome conformation capture (3C) approach that can efficiently identify multiway chromatin interactions with viewpoints of interest. As opposed to pair-wise interactions identified in methods such as Hi-C, 4C, and Capture-C, the detection of multiway interactions allows researchers to investigate how multiple cis-regulatory elements interact together in higher-order structures in single nuclei and address questions regarding structural cooperation between these elements. Here, we describe the procedure for designing and performing a Tri-C experiment.


Subject(s)
Chromatin , Chromosomes , Chromatin/genetics , Regulatory Sequences, Nucleic Acid
7.
Nat Commun ; 13(1): 3485, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35710802

ABSTRACT

The chromatin remodeller ATRX interacts with the histone chaperone DAXX to deposit the histone variant H3.3 at sites of nucleosome turnover. ATRX is known to bind repetitive, heterochromatic regions of the genome including telomeres, ribosomal DNA and pericentric repeats, many of which are putative G-quadruplex forming sequences (PQS). At these sites ATRX plays an ancillary role in a wide range of nuclear processes facilitating replication, chromatin modification and transcription. Here, using an improved protocol for chromatin immunoprecipitation, we show that ATRX also binds active regulatory elements in euchromatin. Mutations in ATRX lead to perturbation of gene expression associated with a reduction in chromatin accessibility, histone modification, transcription factor binding and deposition of H3.3 at the sequences to which it normally binds. In erythroid cells where downregulation of α-globin expression is a hallmark of ATR-X syndrome, perturbation of chromatin accessibility and gene expression occurs in only a subset of cells. The stochastic nature of this process suggests that ATRX acts as a general facilitator of cell specific transcriptional and epigenetic programmes, both in heterochromatin and euchromatin.


Subject(s)
Chromatin , Heterochromatin , DNA Helicases/genetics , DNA Helicases/metabolism , Euchromatin/genetics , Heterochromatin/genetics , Histones/metabolism , Mental Retardation, X-Linked , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , X-linked Nuclear Protein/genetics , X-linked Nuclear Protein/metabolism , alpha-Thalassemia
8.
Annu Rev Genomics Hum Genet ; 23: 73-97, 2022 08 31.
Article in English | MEDLINE | ID: mdl-35472292

ABSTRACT

The successful development and ongoing functioning of complex organisms depend on the faithful execution of the genetic code. A critical step in this process is the correct spatial and temporal expression of genes. The highly orchestrated transcription of genes is controlled primarily by cis-regulatory elements: promoters, enhancers, and insulators. The medical importance of this key biological process can be seen by the frequency with which mutations and inherited variants that alter cis-regulatory elements lead to monogenic and complex diseases and cancer. Here, we provide an overview of the methods available to characterize and perturb gene regulatory circuits. We then highlight mechanisms through which regulatory rewiring contributes to disease, and conclude with a perspective on how our understanding of gene regulation can be used to improve human health.


Subject(s)
Gene Expression Regulation , Gene Regulatory Networks , Enhancer Elements, Genetic , Humans , Mutation , Promoter Regions, Genetic
9.
Nat Commun ; 13(1): 773, 2022 02 09.
Article in English | MEDLINE | ID: mdl-35140205

ABSTRACT

The transcription factor RUNX1 is a critical regulator of developmental hematopoiesis and is frequently disrupted in leukemia. Runx1 is a large, complex gene that is expressed from two alternative promoters under the spatiotemporal control of multiple hematopoietic enhancers. To dissect the dynamic regulation of Runx1 in hematopoietic development, we analyzed its three-dimensional chromatin conformation in mouse embryonic stem cell (ESC) differentiation cultures. Runx1 resides in a 1.1 Mb topologically associating domain (TAD) demarcated by convergent CTCF motifs. As ESCs differentiate to mesoderm, chromatin accessibility, Runx1 enhancer-promoter (E-P) interactions, and CTCF-CTCF interactions increase in the TAD, along with initiation of Runx1 expression from the P2 promoter. Differentiation to hematopoietic progenitor cells is associated with the formation of tissue-specific sub-TADs over Runx1, a shift in E-P interactions, P1 promoter demethylation, and robust expression from both Runx1 promoters. Deletion of promoter-proximal CTCF sites at the sub-TAD boundaries has no obvious effects on E-P interactions but leads to partial loss of domain structure, mildly affects gene expression, and delays hematopoietic development. Together, our analysis of gene regulation at a large multi-promoter developmental gene reveals that dynamic sub-TAD chromatin boundaries play a role in establishing TAD structure and coordinated gene expression.


Subject(s)
Chromatin/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Gene Expression , Animals , Cell Cycle Proteins/metabolism , Cell Differentiation , DNA/chemistry , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/metabolism , Mesoderm/metabolism , Mice , Nucleic Acid Conformation , Promoter Regions, Genetic
10.
Nat Protoc ; 17(2): 445-475, 2022 02.
Article in English | MEDLINE | ID: mdl-35121852

ABSTRACT

Chromosome conformation capture (3C) methods measure the spatial proximity between DNA elements in the cell nucleus. Many methods have been developed to sample 3C material, including the Capture-C family of protocols. Capture-C methods use oligonucleotides to enrich for interactions of interest from sequencing-ready 3C libraries. This approach is modular and has been adapted and optimized to work for sampling of disperse DNA elements (NuTi Capture-C), including from low cell inputs (LI Capture-C), as well as to generate Hi-C like maps for specific regions of interest (Tiled-C) and to interrogate multiway interactions (Tri-C). We present the design, experimental protocol and analysis pipeline for NuTi Capture-C in addition to the variations for generation of LI Capture-C, Tiled-C and Tri-C data. The entire procedure can be performed in 3 weeks and requires standard molecular biology skills and equipment, access to a next-generation sequencing platform, and basic bioinformatic skills. Implemented with other sequencing technologies, these methods can be used to identify regulatory interactions and to compare the structural organization of the genome in different cell types and genetic models.


Subject(s)
Chromosomes
11.
Nat Genet ; 53(11): 1606-1615, 2021 11.
Article in English | MEDLINE | ID: mdl-34737427

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS­CoV­2) disease (COVID-19) pandemic has caused millions of deaths worldwide. Genome-wide association studies identified the 3p21.31 region as conferring a twofold increased risk of respiratory failure. Here, using a combined multiomics and machine learning approach, we identify the gain-of-function risk A allele of an SNP, rs17713054G>A, as a probable causative variant. We show with chromosome conformation capture and gene-expression analysis that the rs17713054-affected enhancer upregulates the interacting gene, leucine zipper transcription factor like 1 (LZTFL1). Selective spatial transcriptomic analysis of lung biopsies from patients with COVID-19 shows the presence of signals associated with epithelial-mesenchymal transition (EMT), a viral response pathway that is regulated by LZTFL1. We conclude that pulmonary epithelial cells undergoing EMT, rather than immune cells, are likely responsible for the 3p21.31-associated risk. Since the 3p21.31 effect is conferred by a gain-of-function, LZTFL1 may represent a therapeutic target.


Subject(s)
COVID-19/complications , Chromosomes, Human, Pair 3/genetics , Epithelial-Mesenchymal Transition , Lung/virology , Polymorphism, Single Nucleotide , SARS-CoV-2/isolation & purification , Transcription Factors/genetics , COVID-19/transmission , COVID-19/virology , Case-Control Studies , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/virology , Female , Genome-Wide Association Study , Humans , Lung/metabolism , Lung/pathology , Male , Transcription Factors/metabolism
12.
Nat Commun ; 12(1): 4439, 2021 07 21.
Article in English | MEDLINE | ID: mdl-34290235

ABSTRACT

The α- and ß-globin loci harbor developmentally expressed genes, which are silenced throughout post-natal life. Reactivation of these genes may offer therapeutic approaches for the hemoglobinopathies, the most common single gene disorders. Here, we address mechanisms regulating the embryonically expressed α-like globin, termed ζ-globin. We show that in embryonic erythroid cells, the ζ-gene lies within a ~65 kb sub-TAD (topologically associating domain) of open, acetylated chromatin and interacts with the α-globin super-enhancer. By contrast, in adult erythroid cells, the ζ-gene is packaged within a small (~10 kb) sub-domain of hypoacetylated, facultative heterochromatin within the acetylated sub-TAD and that it no longer interacts with its enhancers. The ζ-gene can be partially re-activated by acetylation and inhibition of histone de-acetylases. In addition to suggesting therapies for severe α-thalassemia, these findings illustrate the general principles by which reactivation of developmental genes may rescue abnormalities arising from mutations in their adult paralogues.


Subject(s)
Gene Expression Regulation, Developmental , Gene Silencing , Transcriptional Activation , zeta-Globins/genetics , Acetylation , Animals , Chromatin/metabolism , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Erythroid Cells/metabolism , Gene Expression Regulation, Developmental/drug effects , Gene Silencing/drug effects , Histone Deacetylase Inhibitors/pharmacology , Humans , Mice , Repressor Proteins/metabolism , Transcription Factors/metabolism , Transcriptional Activation/drug effects , alpha-Globins/genetics
13.
Nat Commun ; 12(1): 3806, 2021 06 21.
Article in English | MEDLINE | ID: mdl-34155213

ABSTRACT

Many single nucleotide variants (SNVs) associated with human traits and genetic diseases are thought to alter the activity of existing regulatory elements. Some SNVs may also create entirely new regulatory elements which change gene expression, but the mechanism by which they do so is largely unknown. Here we show that a single base change in an otherwise unremarkable region of the human α-globin cluster creates an entirely new promoter and an associated unidirectional transcript. This SNV downregulates α-globin expression causing α-thalassaemia. Of note, the new promoter lying between the α-globin genes and their associated super-enhancer disrupts their interaction in an orientation-dependent manner. Together these observations show how both the order and orientation of the fundamental elements of the genome determine patterns of gene expression and support the concept that active genes may act to disrupt enhancer-promoter interactions in mammals as in Drosophila. Finally, these findings should prompt others to fully evaluate SNVs lying outside of known regulatory elements as causing changes in gene expression by creating new regulatory elements.


Subject(s)
Enhancer Elements, Genetic/genetics , Gain of Function Mutation/genetics , Promoter Regions, Genetic/genetics , Gene Expression Regulation , Humans , Multigene Family , Point Mutation , Transcription, Genetic/genetics , alpha-Globins/genetics , alpha-Thalassemia/genetics
14.
mBio ; 12(3): e0076821, 2021 06 29.
Article in English | MEDLINE | ID: mdl-34154419

ABSTRACT

Fungi, bacteria, and plants, but not animals, synthesize the branched-chain amino acids: leucine, isoleucine, and valine. While branched-chain amino acid (BCAA) biosynthesis has been well characterized in the yeast Saccharomyces cerevisiae, it is incompletely understood in filamentous fungi. The three BCAAs share several early biosynthesis steps before divergence into specific pathways. In Aspergillus nidulans, the genes for the first two dedicated steps in leucine biosynthesis have been characterized, but the final two have not. We used sequence searches of the A. nidulans genome to identify two genes encoding ß-isopropylmalate dehydrogenase, which catalyzes the penultimate step of leucine biosynthesis, and six genes encoding BCAA aminotransferase, which catalyzes the final step in biosynthesis of all three BCAA. We have used combinations of gene knockouts to determine the relative contribution of each of these genes to BCAA biosynthesis. While both ß-isopropylmalate dehydrogenase genes act in leucine biosynthesis, the two most highly expressed BCAA aminotransferases are responsible for BCAA biosynthesis. We have also characterized the expression of leucine biosynthesis genes using reverse transcriptase-quantitative PCR and found regulation in response to leucine availability is mediated through the Zn(II)2Cys6 transcription factor LeuB. IMPORTANCE Branched-chain amino acid (BCAA) biosynthesis is important for pathogenic fungi to successfully cause disease in human and plant hosts. The enzymes for their production are absent from humans and, therefore, provide potential antifungal targets. While BCAA biosynthesis is well characterized in yeasts, it is poorly understood in filamentous fungal pathogens. Developing a thorough understanding of both the genes encoding the metabolic enzymes for BCAA biosynthesis and how their expression is regulated will inform target selection for antifungal drug development.


Subject(s)
Amino Acids, Branched-Chain/genetics , Amino Acids, Branched-Chain/metabolism , Aspergillus nidulans/genetics , Biosynthetic Pathways/genetics , Amino Acids, Branched-Chain/biosynthesis , Aspergillus nidulans/chemistry , Gene Expression Regulation, Fungal , Leucine/biosynthesis , Transaminases/genetics , Transaminases/metabolism
15.
Nature ; 595(7865): 125-129, 2021 07.
Article in English | MEDLINE | ID: mdl-34108683

ABSTRACT

In higher eukaryotes, many genes are regulated by enhancers that are 104-106 base pairs (bp) away from the promoter. Enhancers contain transcription-factor-binding sites (which are typically around 7-22 bp), and physical contact between the promoters and enhancers is thought to be required to modulate gene expression. Although chromatin architecture has been mapped extensively at resolutions of 1 kilobase and above; it has not been possible to define physical contacts at the scale of the proteins that determine gene expression. Here we define these interactions in detail using a chromosome conformation capture method (Micro-Capture-C) that enables the physical contacts between different classes of regulatory elements to be determined at base-pair resolution. We find that highly punctate contacts occur between enhancers, promoters and CCCTC-binding factor (CTCF) sites and we show that transcription factors have an important role in the maintenance of the contacts between enhancers and promoters. Our data show that interactions between CTCF sites are increased when active promoters and enhancers are located within the intervening chromatin. This supports a model in which chromatin loop extrusion1 is dependent on cohesin loading at active promoters and enhancers, which explains the formation of tissue-specific chromatin domains without changes in CTCF binding.


Subject(s)
Base Pairing/genetics , Genome/genetics , Animals , Binding Sites , CCCTC-Binding Factor/metabolism , Cell Cycle Proteins/metabolism , Cells, Cultured , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Enhancer Elements, Genetic/genetics , Erythroid Cells/cytology , Erythroid Cells/metabolism , Gene Expression Regulation , Mice , Mice, Inbred C57BL , Organ Specificity , Promoter Regions, Genetic/genetics , alpha-Globins/genetics , Cohesins
16.
Commun Biol ; 4(1): 623, 2021 05 25.
Article in English | MEDLINE | ID: mdl-34035422

ABSTRACT

Tracking and understanding data quality, analysis and reproducibility are critical concerns in the biological sciences. This is especially true in genomics where next generation sequencing (NGS) based technologies such as ChIP-seq, RNA-seq and ATAC-seq are generating a flood of genome-scale data. However, such data are usually processed with automated tools and pipelines, generating tabular outputs and static visualisations. Interpretation is normally made at a high level without the ability to visualise the underlying data in detail. Conventional genome browsers are limited to browsing single locations and do not allow for interactions with the dataset as a whole. Multi Locus View (MLV), a web-based tool, has been developed to allow users to fluidly interact with genomics datasets at multiple scales. The user is able to browse the raw data, cluster, and combine the data with other analysis and annotate the data. User datasets can then be shared with other users or made public for quick assessment from the academic community. MLV is publically available at https://mlv.molbiol.ox.ac.uk .


Subject(s)
Sequence Analysis, DNA/methods , Chromatin Immunoprecipitation Sequencing/methods , Computational Biology/methods , Genomics/methods , High-Throughput Nucleotide Sequencing/methods , Internet , Numerical Analysis, Computer-Assisted , RNA-Seq/methods , Reproducibility of Results , Sequence Analysis, RNA/methods , Software
17.
Nucleic Acids Res ; 49(5): 2488-2508, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33533919

ABSTRACT

The ubiquitous family of dimeric transcription factors AP-1 is made up of Fos and Jun family proteins. It has long been thought to operate principally at gene promoters and how it controls transcription is still ill-understood. The Fos family protein Fra-1 is overexpressed in triple negative breast cancers (TNBCs) where it contributes to tumor aggressiveness. To address its transcriptional actions in TNBCs, we combined transcriptomics, ChIP-seqs, machine learning and NG Capture-C. Additionally, we studied its Fos family kin Fra-2 also expressed in TNBCs, albeit much less. Consistently with their pleiotropic effects, Fra-1 and Fra-2 up- and downregulate individually, together or redundantly many genes associated with a wide range of biological processes. Target gene regulation is principally due to binding of Fra-1 and Fra-2 at regulatory elements located distantly from cognate promoters where Fra-1 modulates the recruitment of the transcriptional co-regulator p300/CBP and where differences in AP-1 variant motif recognition can underlie preferential Fra-1- or Fra-2 bindings. Our work also shows no major role for Fra-1 in chromatin architecture control at target gene loci, but suggests collaboration between Fra-1-bound and -unbound enhancers within chromatin hubs sometimes including promoters for other Fra-1-regulated genes. Our work impacts our view of AP-1.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins c-fos/metabolism , Triple Negative Breast Neoplasms/genetics , Binding Sites , Cell Line, Tumor , Chromatin/chemistry , Chromatin/metabolism , Epigenesis, Genetic , Fos-Related Antigen-2/metabolism , Humans , Nucleotide Motifs , Promoter Regions, Genetic , Proto-Oncogene Proteins c-fos/physiology , Transcription Factor AP-1/metabolism , Triple Negative Breast Neoplasms/metabolism , p300-CBP Transcription Factors/metabolism
18.
Mol Cell ; 81(5): 983-997.e7, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33539786

ABSTRACT

Gene transcription occurs via a cycle of linked events, including initiation, promoter-proximal pausing, and elongation of RNA polymerase II (Pol II). A key question is how transcriptional enhancers influence these events to control gene expression. Here, we present an approach that evaluates the level and change in promoter-proximal transcription (initiation and pausing) in the context of differential gene expression, genome-wide. This combinatorial approach shows that in primary cells, control of gene expression during differentiation is achieved predominantly via changes in transcription initiation rather than via release of Pol II pausing. Using genetically engineered mouse models, deleted for functionally validated enhancers of the α- and ß-globin loci, we confirm that these elements regulate Pol II recruitment and/or initiation to modulate gene expression. Together, our data show that gene expression during differentiation is regulated predominantly at the level of initiation and that enhancers are key effectors of this process.


Subject(s)
Enhancer Elements, Genetic , Promoter Regions, Genetic , RNA Polymerase II/genetics , Transcription Initiation, Genetic , alpha-Globins/genetics , beta-Globins/genetics , Animals , Cell Differentiation , Exons , Fetus , Gene Expression Regulation , Gene Library , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Humans , Introns , K562 Cells , Liver/cytology , Liver/metabolism , Mice , Mice, Knockout , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA Polymerase II/metabolism , Signal Transduction , alpha-Globins/deficiency , beta-Globins/deficiency
19.
Nat Commun ; 12(1): 531, 2021 01 22.
Article in English | MEDLINE | ID: mdl-33483495

ABSTRACT

Chromosome conformation capture (3C) provides an adaptable tool for studying diverse biological questions. Current 3C methods generally provide either low-resolution interaction profiles across the entire genome, or high-resolution interaction profiles at limited numbers of loci. Due to technical limitations, generation of reproducible high-resolution interaction profiles has not been achieved at genome-wide scale. Here, to overcome this barrier, we systematically test each step of 3C and report two improvements over current methods. We show that up to 30% of reporter events generated using the popular in situ 3C method arise from ligations between two individual nuclei, but this noise can be almost entirely eliminated by isolating intact nuclei after ligation. Using Nuclear-Titrated Capture-C, we generate reproducible high-resolution genome-wide 3C interaction profiles by targeting 8055 gene promoters in erythroid cells. By pairing high-resolution 3C interaction calls with nascent gene expression we interrogate the role of promoter hubs and super-enhancers in gene regulation.


Subject(s)
Cell Nucleus/genetics , Chromatin/genetics , Erythroid Cells/metabolism , Genome, Human/genetics , Genome-Wide Association Study/methods , Regulatory Sequences, Nucleic Acid/genetics , Animals , Cells, Cultured , Chromosome Mapping/methods , Computational Biology/methods , Gene Expression Regulation , Genomics/methods , Humans , Mice, Inbred C57BL , Mice, Inbred CBA
20.
J Med Genet ; 58(3): 185-195, 2021 03.
Article in English | MEDLINE | ID: mdl-32518175

ABSTRACT

BACKGROUND: Congenital dyserythropoietic anaemia type I (CDA-I) is a hereditary anaemia caused by biallelic mutations in the widely expressed genes CDAN1 and C15orf41. Little is understood about either protein and it is unclear in which cellular pathways they participate. METHODS: Genetic analysis of a cohort of patients with CDA-I identifies novel pathogenic variants in both known causative genes. We analyse the mutation distribution and the predicted structural positioning of amino acids affected in Codanin-1, the protein encoded by CDAN1. Using western blotting, immunoprecipitation and immunofluorescence, we determine the effect of particular mutations on both proteins and interrogate protein interaction, stability and subcellular localisation. RESULTS: We identify six novel CDAN1 mutations and one novel mutation in C15orf41 and uncover evidence of further genetic heterogeneity in CDA-I. Additionally, population genetics suggests that CDA-I is more common than currently predicted. Mutations are enriched in six clusters in Codanin-1 and tend to affect buried residues. Many missense and in-frame mutations do not destabilise the entire protein. Rather C15orf41 relies on Codanin-1 for stability and both proteins, which are enriched in the nucleolus, interact to form an obligate complex in cells. CONCLUSION: Stability and interaction data suggest that C15orf41 may be the key determinant of CDA-I and offer insight into the mechanism underlying this disease. Both proteins share a common pathway likely to be present in a wide variety of cell types; however, nucleolar enrichment may provide a clue as to the erythroid specific nature of CDA-I. The surprisingly high predicted incidence of CDA-I suggests that better ascertainment would lead to improved patient care.


Subject(s)
Anemia, Dyserythropoietic, Congenital/genetics , Genetic Predisposition to Disease , Glycoproteins/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Anemia, Dyserythropoietic, Congenital/pathology , Female , Gene Expression Regulation/genetics , Genetic Testing , Genetics, Population , Humans , Male , Multiprotein Complexes/genetics , Mutation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...