Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
Genes (Basel) ; 15(7)2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39062704

ABSTRACT

The identification of structural variants (SVs) in genomic data represents an ongoing challenge because of difficulties in reliable SV calling leading to reduced sensitivity and specificity. We prepared high-quality DNA from 9 parent-child trios, who had previously undergone short-read whole-genome sequencing (Illumina platform) as part of the Genomics England 100,000 Genomes Project. We reanalysed the genomes using both Bionano optical genome mapping (OGM; 8 probands and one trio) and Nanopore long-read sequencing (Oxford Nanopore Technologies [ONT] platform; all samples). To establish a "truth" dataset, we asked whether rare proband SV calls (n = 234) made by the Bionano Access (version 1.6.1)/Solve software (version 3.6.1_11162020) could be verified by individual visualisation using the Integrative Genomics Viewer with either or both of the Illumina and ONT raw sequence. Of these, 222 calls were verified, indicating that Bionano OGM calls have high precision (positive predictive value 95%). We then asked what proportion of the 222 true Bionano SVs had been identified by SV callers in the other two datasets. In the Illumina dataset, sensitivity varied according to variant type, being high for deletions (115/134; 86%) but poor for insertions (13/58; 22%). In the ONT dataset, sensitivity was generally poor using the original Sniffles variant caller (48% overall) but improved substantially with use of Sniffles2 (36/40; 90% and 17/23; 74% for deletions and insertions, respectively). In summary, we show that the precision of OGM is very high. In addition, when applying the Sniffles2 caller, the sensitivity of SV calling using ONT long-read sequence data outperforms Illumina sequencing for most SV types.


Subject(s)
Benchmarking , Nanopore Sequencing , Whole Genome Sequencing , Humans , Whole Genome Sequencing/methods , Whole Genome Sequencing/standards , Nanopore Sequencing/methods , Benchmarking/methods , Genomic Structural Variation/genetics , Chromosome Mapping/methods , Genome, Human/genetics , Genomics/methods , Software , High-Throughput Nucleotide Sequencing/methods , High-Throughput Nucleotide Sequencing/standards , Female , Nanopores , Male , Sequence Analysis, DNA/methods , Sequence Analysis, DNA/standards
2.
Nucleic Acids Res ; 51(13): 6509-6527, 2023 07 21.
Article in English | MEDLINE | ID: mdl-36940725

ABSTRACT

Telomere maintenance is a hallmark of malignant cells and allows cancers to divide indefinitely. In some cancers, this is achieved through the alternative lengthening of telomeres (ALT) pathway. Whilst loss of ATRX is a near universal feature of ALT-cancers, it is insufficient in isolation. As such, other cellular events must be necessary - but the exact nature of the secondary events has remained elusive. Here, we report that trapping of proteins (such as TOP1, TOP2A and PARP1) on DNA leads to ALT induction in cells lacking ATRX. We demonstrate that protein-trapping chemotherapeutic agents, such as etoposide, camptothecin and talazoparib, induce ALT markers specifically in ATRX-null cells. Further, we show that treatment with G4-stabilising drugs cause an increase in trapped TOP2A levels which leads to ALT induction in ATRX-null cells. This process is MUS81-endonuclease and break-induced replication dependent, suggesting that protein trapping leads to replication fork stalling, with these forks being aberrantly processed in the absence of ATRX. Finally, we show ALT-positive cells harbour a higher load of genome-wide trapped proteins, such as TOP1, and knockdown of TOP1 reduced ALT activity. Taken together, these findings suggest that protein trapping is a fundamental driving force behind ALT-biology in ATRX-deficient malignancies.


A key feature of all cancer cells is their ability to divide indefinitely, and this is dependent on circumvention of telomere shortening through induction of a telomere maintenance mechanism, such as the telomerase-independent, Alternative Lengthening of Telomeres (ALT) pathway. The ALT pathway is characterised by loss of the ATRX chromatin remodeler. The current study provides evidence that, in the absence of ATRX, increased trapping of proteins on DNA leads to replication fork stalling and collapse. At telomeres, this leads to ALT pathway activity. These results help to better understand ALT tumours and might, eventually, be instrumental in developing new therapeutic strategies.


Subject(s)
Neoplasms , Telomere , Humans , DNA , Neoplasms/genetics , Telomerase/genetics , Telomere/genetics , Telomere/metabolism , Telomere Homeostasis , X-linked Nuclear Protein/genetics , X-linked Nuclear Protein/metabolism
3.
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
4.
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
5.
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
6.
Blood ; 136(3): 269-278, 2020 07 16.
Article in English | MEDLINE | ID: mdl-32396940

ABSTRACT

The oxygen transport function of hemoglobin (HB) is thought to have arisen ∼500 million years ago, roughly coinciding with the divergence between jawless (Agnatha) and jawed (Gnathostomata) vertebrates. Intriguingly, extant HBs of jawless and jawed vertebrates were shown to have evolved twice, and independently, from different ancestral globin proteins. This raises the question of whether erythroid-specific expression of HB also evolved twice independently. In all jawed vertebrates studied to date, one of the HB gene clusters is linked to the widely expressed NPRL3 gene. Here we show that the nprl3-linked hb locus of a jawless vertebrate, the river lamprey (Lampetra fluviatilis), shares a range of structural and functional properties with the equivalent jawed vertebrate HB locus. Functional analysis demonstrates that an erythroid-specific enhancer is located in intron 7 of lamprey nprl3, which corresponds to the NPRL3 intron 7 MCS-R1 enhancer of jawed vertebrates. Collectively, our findings signify the presence of an nprl3-linked multiglobin gene locus, which contains a remote enhancer that drives globin expression in erythroid cells, before the divergence of jawless and jawed vertebrates. Different globin genes from this ancestral cluster evolved in the current NPRL3-linked HB genes in jawless and jawed vertebrates. This provides an explanation of the enigma of how, in different species, globin genes linked to the same adjacent gene could undergo convergent evolution.


Subject(s)
Erythrocytes/metabolism , Evolution, Molecular , Fish Proteins , Gene Expression Regulation/physiology , Hemoglobins , Lampreys , Animals , Fish Proteins/biosynthesis , Fish Proteins/genetics , Hemoglobins/biosynthesis , Hemoglobins/genetics , Lampreys/genetics , Lampreys/metabolism , Multigene Family
7.
Int J Mol Sci ; 20(21)2019 Oct 29.
Article in English | MEDLINE | ID: mdl-31671722

ABSTRACT

Methyl-CpG binding protein 2 (MeCP2) is a multi-function factor involved in locus-specific transcriptional modulation and the regulation of genome architecture, e.g., pericentric heterochromatin (PCH) organization. MECP2 mutations are responsible for Rett syndrome (RTT), a devastating postnatal neurodevelopmental disorder, the pathogenetic mechanisms of which are still unknown. MeCP2, together with Alpha-thalassemia/mental retardation syndrome X-linked protein (ATRX), accumulates at chromocenters, which are repressive PCH domains. As with MECP2, mutations in ATRX cause ATR-X syndrome which is associated with severe intellectual disability. We exploited two murine embryonic stem cell lines, in which the expression of MeCP2 or ATRX is abolished. Through immunostaining, chromatin immunoprecipitation and western blot, we show that MeCP2 and ATRX are reciprocally dependent both for their expression and targeting to chromocenters. Moreover, ATRX plays a role in the accumulation of members of the heterochromatin protein 1 (HP1) family at PCH and, as MeCP2, modulates their expression. Furthermore, ATRX and HP1 targeting to chromocenters depends on an RNA component. 3D-DNA fluorescence in situ hybridization (FISH) highlighted, for the first time, a contribution of ATRX in MeCP2-mediated chromocenter clustering during neural differentiation. Overall, we provide a detailed dissection of the functional interplay between MeCP2 and ATRX in higher-order PCH organization in neurons. Our findings suggest molecular defects common to RTT and ATR-X syndrome, including an alteration in PCH.


Subject(s)
Cell Differentiation/physiology , Heterochromatin/metabolism , Methyl-CpG-Binding Protein 2/metabolism , Neurons/metabolism , X-linked Nuclear Protein/metabolism , Animals , Cell Differentiation/genetics , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Disease Models, Animal , Embryonic Stem Cells , Gene Expression Regulation , Gene Knockout Techniques , Heterochromatin/chemistry , Heterochromatin/genetics , In Situ Hybridization, Fluorescence , Intellectual Disability/genetics , Mental Retardation, X-Linked/genetics , Methyl-CpG-Binding Protein 2/genetics , Mice , Mutation , Rett Syndrome/genetics , X-linked Nuclear Protein/chemistry , X-linked Nuclear Protein/genetics , alpha-Thalassemia/genetics
9.
Nat Struct Mol Biol ; 26(3): 213-219, 2019 03.
Article in English | MEDLINE | ID: mdl-30833786

ABSTRACT

The growth of telomerase-deficient cancers depends on the alternative lengthening of telomeres (ALT), a homology-directed telomere-maintenance pathway. ALT telomeres exhibit a unique chromatin environment and generally lack the nucleosome remodeler ATRX, pointing to an epigenetic basis for ALT. Recently, we identified a protective role for the ATRX-interacting macroH2A1.2 histone variant during homologous recombination and replication stress (RS). Consistent with an inherent susceptibility to RS, we show that human ALT telomeres are highly enriched for macroH2A1.2. However, in contrast to ATRX-proficient cells, ALT telomeres transiently lose macroH2A1.2 during acute RS to facilitate DNA double-strand break (DSB) formation, a process that is almost completely prevented by ectopic ATRX expression. Telomeric macroH2A1.2 is re-deposited in a DNA damage response (DDR)-dependent manner to promote homologous recombination-associated ALT pathways. Our findings thus identify the dynamic exchange of macroH2A1.2 on chromatin as an epigenetic link among ATRX loss, RS-induced DDR initiation and telomere maintenance via homologous recombination.


Subject(s)
Chromatin/metabolism , DNA Repair/genetics , Histones/genetics , Homologous Recombination/genetics , Telomere Homeostasis/genetics , X-linked Nuclear Protein/genetics , Cell Line, Tumor , DNA Breaks, Double-Stranded , HEK293 Cells , HeLa Cells , Humans , Telomerase/metabolism
10.
Am J Med Genet A ; 179(3): 344-349, 2019 03.
Article in English | MEDLINE | ID: mdl-30637921

ABSTRACT

Primrose syndrome is a rare autosomal dominant condition caused by heterozygous missense variants within ZBTB20. Through an exome sequencing approach (as part of the Deciphering Developmental Disorders [DDD] study) we have identified five unrelated individuals with previously unreported, de novo ZBTB20 pathogenic missense variants. All five missense variants targeted the C2H2 zinc finger domains. This genotype-up approach has allowed further refinement of the Primrose syndrome phenotype. Major characteristics (>90% individuals) include an intellectual disability (most frequently in the moderate range), a recognizable facial appearance and brain MRI abnormalities, particularly abnormalities of the corpus callosum. Other frequent clinical associations (in 50-90% individuals) include sensorineural hearing loss (83%), hypotonia (78%), cryptorchidism in males (75%), macrocephaly (72%), behavioral issues (56%), and dysplastic/hypoplastic nails (57%). Based upon these clinical data we discuss our current management of patients with Primrose syndrome.


Subject(s)
Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/genetics , Calcinosis/diagnosis , Calcinosis/genetics , Ear Diseases/diagnosis , Ear Diseases/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Genetic Variation , Intellectual Disability/diagnosis , Intellectual Disability/genetics , Muscular Atrophy/diagnosis , Muscular Atrophy/genetics , Nerve Tissue Proteins/genetics , Phenotype , Transcription Factors/genetics , Child , Child, Preschool , Facies , Female , Genetic Loci , Genotype , Humans , Magnetic Resonance Imaging , Male , Mutation
11.
Methods Mol Biol ; 1832: 105-130, 2018.
Article in English | MEDLINE | ID: mdl-30073524

ABSTRACT

Chromatin immunoprecipitation coupled with high-throughput, next-generation DNA sequencing (ChIP-seq) has enabled researchers to establish the genome-wide patterns of chromatin modifications and binding of chromatin-associated proteins. Well-established protocols produce robust ChIP-seq data for many proteins by sequencing the DNA obtained following immunoprecipitation of fragmented chromatin using a wide range of specific antibodies. In general, the quality of these data mainly depends on the specificity and avidity of the antibody used. However, even using optimal antibodies, ChIP-seq can become more challenging when the protein associates with chromatin via protein-protein interactions rather than directly binding DNA. An example of such a protein is the alpha-thalassaemia mental retardation X-linked (ATRX) protein; a chromatin remodeler that associates with the histone chaperone DAXX, in the deposition of the replication-independent histone variant H3.3 and plays an important role in maintaining chromatin integrity. Inherited mutations of ATRX cause syndromal mental retardation (ATR-X Syndrome) whereas acquired mutations are associated with myelodysplasia, acute myeloid leukemia (ATMDS syndrome), and a range of solid tumors. Therefore, high quality ChIP-seq data have been needed to analyze the genome-wide distribution of ATRX, to advance our understanding of its normal role and to comprehend how mutations contribute to human disease. Here, we describe an optimized ChIP-seq protocol for ATRX which can also be used to produce high quality data sets for other challenging proteins which are indirectly associated with DNA and complement the ChIP-seq toolkit for genome-wide analyses of histone chaperon complexes and associated chromatin remodelers. Although not a focus of this chapter, we will also provide some insight for the analysis of the large dataset generated by ChIP-seq. Even though this protocol has been fully optimized for ATRX, it should also provide guidance for efficient ChIP-seq analysis, using the appropriate antibodies, for other proteins interacting indirectly with DNA.


Subject(s)
Chromatin Immunoprecipitation/methods , Cross-Linking Reagents/chemistry , X-linked Nuclear Protein/metabolism , Gene Library , Humans , Real-Time Polymerase Chain Reaction , Reproducibility of Results , Sonication
12.
Cell Rep ; 23(5): 1553-1564, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29719265

ABSTRACT

Isocitrate dehydrogenase 1 (IDH1) is the most commonly mutated gene in grade II-III glioma and secondary glioblastoma (GBM). A causal role for IDH1R132H in gliomagenesis has been proposed, but functional validation in vivo has not been demonstrated. In this study, we assessed the role of IDH1R132H in glioma development in the context of clinically relevant cooperating genetic alterations in vitro and in vivo. Immortal astrocytes expressing IDH1R132H exhibited elevated (R)-2-hydroxyglutarate levels, reduced NADPH, increased proliferation, and anchorage-independent growth. Although not sufficient on its own, IDH1R132H cooperated with PDGFA and loss of Cdkn2a, Atrx, and Pten to promote glioma development in vivo. These tumors resembled proneural human mutant IDH1 GBM genetically, histologically, and functionally. Our findings support the hypothesis that IDH1R132H promotes glioma development. This model enhances our understanding of the biology of IDH1R132H-driven gliomas and facilitates testing of therapeutic strategies designed to combat this deadly disease.


Subject(s)
Astrocytes/enzymology , Carcinogenesis/metabolism , Glioma/enzymology , Isocitrate Dehydrogenase/metabolism , Mutation, Missense , Neoplasm Proteins/metabolism , Amino Acid Substitution , Animals , Astrocytes/pathology , Carcinogenesis/genetics , Carcinogenesis/pathology , Glioma/genetics , Glioma/pathology , Humans , Isocitrate Dehydrogenase/genetics , Mice , Mice, Transgenic , Neoplasm Proteins/genetics
13.
Eur J Med Genet ; 61(5): 273-279, 2018 May.
Article in English | MEDLINE | ID: mdl-29307788

ABSTRACT

Pontocerebellar hypoplasia type 10 (PCH10) is a progressive autosomal recessive neurodegenerative disorder that has been recently described in association with cleavage and polyadenylation factor I subunit 1 (CLP1) mutations. To date, all reported cases have the same homozygous missense mutation in the CLP1 gene suggesting a founder mutation. CLP1 is an RNA kinase involved in tRNA splicing and maturation. There is evidence that the mutation is associated with functionally impaired kinase activity and subsequent defective tRNA processing. Through whole exome sequencing, we identified the same mutation in an extended family of Turkish origin. Both children presented with severe psychomotor delay, progressive microcephaly, and constipation. However, intrafamilial phenotypic variability is suggested due to the variability in their brain abnormalities and clinical features.


Subject(s)
Cerebellar Diseases/genetics , Cerebellar Diseases/diagnosis , Child , Female , Humans , Mutation, Missense , Nuclear Proteins/genetics , Pedigree , Phosphotransferases/genetics , Transcription Factors/genetics , Exome Sequencing
14.
Exp Hematol ; 60: 10-20, 2018 04.
Article in English | MEDLINE | ID: mdl-29329925

ABSTRACT

In vitro erythroid differentiation systems are used to study the mechanisms underlying normal and abnormal erythropoiesis and to test the effects of various extracellular factors on erythropoiesis. The use of serum or conditioned medium in liquid cultures and the seeding of cultures with heterogeneous peripheral blood mononuclear cells confound the reproducibility of these systems. Newer erythroid differentiation culture systems have overcome some of these limitations by using a fully defined, serum-free medium and initiating cultures using purified CD34+ cells. Although widely used in bulk cultures, these protocols have not been rigorously tested in high-throughput or single-cell assays. Here, we describe a serum-free erythroid differentiation system suitable for small-scale and single-cell experiments. This system generates large numbers of terminally differentiated erythroid cells of very high purity. Here we have adapted this culture system to a 96-well format and have developed a protocol to grow erythroid colonies from single erythroid progenitors in minute culture volumes.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation , Erythroid Cells/cytology , Erythroid Cells/metabolism , Erythropoiesis , Culture Media, Serum-Free/chemistry , Culture Media, Serum-Free/pharmacology , Humans
15.
Methods Protoc ; 1(3)2018 Jul 30.
Article in English | MEDLINE | ID: mdl-31164570

ABSTRACT

The study of cellular processes and gene regulation in terminal erythroid development has been greatly facilitated by the generation of an immortalised erythroid cell line derived from Human Umbilical Derived Erythroid Precursors, termed HUDEP-2 cells. The ability to efficiently genome edit HUDEP-2 cells and make clonal lines hugely expands their utility as the insertion of clinically relevant mutations allows study of potentially every genetic disease affecting red blood cell development. Additionally, insertion of sequences encoding short protein tags such as Strep, FLAG and Myc permits study of protein behaviour in the normal and disease state. This approach is useful to augment the analysis of patient cells as large cell numbers are obtainable with the additional benefit that the need for specific antibodies may be circumvented. This approach is likely to lead to insights into disease mechanisms and provide reagents to allow drug discovery. HUDEP-2 cells provide a favourable alternative to the existing immortalised erythroleukemia lines as their karyotype is much less abnormal. These cells also provide sufficient material for a broad range of analyses as it is possible to generate in vitro-differentiated erythroblasts in numbers 4-7 fold higher than starting cell numbers within 9-12 days of culture. Here we describe an efficient, robust and reproducible plasmid-based methodology to introduce short (<20 bp) DNA sequences into the genome of HUDEP-2 cells using the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR associated protein 9 Cas9 system combined with single-stranded oligodeoxynucleotide (ssODN) donors. This protocol produces genetically modified lines in ~30 days and could also be used to generate knock-out and knock-in mutations.

16.
Nat Commun ; 8(1): 424, 2017 09 04.
Article in English | MEDLINE | ID: mdl-28871148

ABSTRACT

ß-Thalassemia is one of the most common inherited anemias, with no effective cure for most patients. The pathophysiology reflects an imbalance between α- and ß-globin chains with an excess of free α-globin chains causing ineffective erythropoiesis and hemolysis. When α-thalassemia is co-inherited with ß-thalassemia, excess free α-globin chains are reduced significantly ameliorating the clinical severity. Here we demonstrate the use of CRISPR/Cas9 genome editing of primary human hematopoietic stem/progenitor (CD34+) cells to emulate a natural mutation, which deletes the MCS-R2 α-globin enhancer and causes α-thalassemia. When edited CD34+ cells are differentiated into erythroid cells, we observe the expected reduction in α-globin expression and a correction of the pathologic globin chain imbalance in cells from patients with ß-thalassemia. Xenograft assays show that a proportion of the edited CD34+ cells are long-term repopulating hematopoietic stem cells, demonstrating the potential of this approach for translation into a therapy for ß-thalassemia.ß-thalassemia is characterised by the presence of an excess of α-globin chains, which contribute to erythrocyte pathology. Here the authors use CRISP/Cas9 to reduce α-globin expression in hematopoietic precursors, and show effectiveness in xenograft assays in mice.


Subject(s)
Enhancer Elements, Genetic/genetics , Gene Editing , Hematopoietic Stem Cells/metabolism , alpha-Globins/genetics , beta-Thalassemia/genetics , beta-Thalassemia/therapy , Animals , Antigens, CD34/metabolism , Base Sequence , CRISPR-Cas Systems , Cells, Cultured , Female , Gene Knockdown Techniques , Genome, Human , Heterografts , Humans , Mice , Sequence Deletion/genetics , Single-Cell Analysis
17.
EMBO Rep ; 18(6): 914-928, 2017 06.
Article in English | MEDLINE | ID: mdl-28487353

ABSTRACT

ATRX is a chromatin remodelling factor found at a wide range of tandemly repeated sequences including telomeres (TTAGGG)n ATRX mutations are found in nearly all tumours that maintain their telomeres via the alternative lengthening of telomere (ALT) pathway, and ATRX is known to suppress this pathway. Here, we show that recruitment of ATRX to telomeric repeats depends on repeat number, orientation and, critically, on repeat transcription. Importantly, the transcribed telomeric repeats form RNA-DNA hybrids (R-loops) whose abundance correlates with the recruitment of ATRX Here, we show loss of ATRX is also associated with increased R-loop formation. Our data suggest that the presence of ATRX at telomeres may have a central role in suppressing deleterious DNA secondary structures that form at transcribed telomeric repeats, and this may account for the increased DNA damage, stalling of replication and homology-directed repair previously observed upon loss of ATRX function.


Subject(s)
Chromatin Assembly and Disassembly , DNA/genetics , RNA/genetics , Telomere/genetics , Telomere/metabolism , X-linked Nuclear Protein/metabolism , Chromatin , DNA/chemistry , DNA Damage , DNA Replication , G-Quadruplexes , Humans , Telomere Homeostasis/genetics , Transcription Factors/metabolism , Transcription, Genetic , X-linked Nuclear Protein/deficiency , X-linked Nuclear Protein/genetics
19.
Nat Genet ; 48(8): 895-903, 2016 08.
Article in English | MEDLINE | ID: mdl-27376235

ABSTRACT

Many genes determining cell identity are regulated by clusters of Mediator-bound enhancer elements collectively referred to as super-enhancers. These super-enhancers have been proposed to manifest higher-order properties important in development and disease. Here we report a comprehensive functional dissection of one of the strongest putative super-enhancers in erythroid cells. By generating a series of mouse models, deleting each of the five regulatory elements of the α-globin super-enhancer individually and in informative combinations, we demonstrate that each constituent enhancer seems to act independently and in an additive fashion with respect to hematological phenotype, gene expression, chromatin structure and chromosome conformation, without clear evidence of synergistic or higher-order effects. Our study highlights the importance of functional genetic analyses for the identification of new concepts in transcriptional regulation.


Subject(s)
Enhancer Elements, Genetic/genetics , Erythroid Cells/metabolism , Gene Expression Regulation , Transcription Factors/metabolism , Transcription, Genetic/genetics , alpha-Globins/genetics , Animals , Chromatin/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Mice , Mice, Knockout
20.
Cell Mol Life Sci ; 73(9): 1871-9, 2016 May.
Article in English | MEDLINE | ID: mdl-26883803

ABSTRACT

Imprinted genes are an exceptional cluster of genes which are expressed in a parent-of-origin dependent fashion. This allele-specific expression is dependent on differential DNA methylation which is established in the parental germlines in a sex-specific manner. The DNA methylation imprint is accompanied by heterochromatin modifications which must be continuously maintained through development. This review summarises the factors which are important for protecting the epigenetic modifications at imprinted differentially methylated regions (DMRs), including PGC7, ZFP57 and the ATRX/Daxx/H3.3 complex. We discuss how these factors maintain heterochromatin silencing, not only at imprinted DMRs, but also other heterochromatic regions in the genome.


Subject(s)
Gene Silencing , Genomic Imprinting , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Chromosomal Proteins, Non-Histone , Co-Repressor Proteins , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Methylation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Heterochromatin/metabolism , Histones/genetics , Histones/metabolism , Humans , Molecular Chaperones , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Proteins/genetics , Proteins/metabolism , Repressor Proteins , Transcription Factors/genetics , Transcription Factors/metabolism , X-linked Nuclear Protein
SELECTION OF CITATIONS
SEARCH DETAIL