LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 10 of total 19

Search options

  1. Article ; Online: Predicting Chromatin Interactions from DNA Sequence Using DeepC.

    Schwessinger, Ron

    Methods in molecular biology (Clifton, N.J.)

    2023  Volume 2624, Page(s) 19–42

    Abstract: The genome 3D structure is central to understanding how disease-associated genetic variants in the noncoding genome regulate their target genes. Genome architecture spans large-scale structures determined by fine-grained regulatory elements, making it ... ...

    Abstract The genome 3D structure is central to understanding how disease-associated genetic variants in the noncoding genome regulate their target genes. Genome architecture spans large-scale structures determined by fine-grained regulatory elements, making it challenging to predict the effects of sequence and structural variants. Experimental approaches for chromatin interaction mapping remain costly and time-consuming, limiting their use for interrogating changes of chromatin architecture associated with genomic variation at scale. Computational models to predict chromatin interactions have either interpreted chromatin at coarse resolution or failed to capture the long-range dependencies of larger sequence contexts. To bridge this gap, we previously developed deepC, a deep neural network approach to predict chromatin interactions from DNA sequence at megabase scale. deepC employs dilated convolutional layers to achieve simultaneously a large sequence context while interpreting the DNA sequence at single base pair resolution. Using transfer learning of convolutional weights trained to predict a compendium of chromatin features across cell types allows deepC to predict cell type-specific chromatin interactions from DNA sequence alone. Here, we present a detailed workflow to predict chromatin interactions with deepC. We detail the necessary data pre-processing steps, guide through deepC model training, and demonstrate how to employ trained models to predict chromatin interactions and the effect of sequence variations on genome architecture.
    MeSH term(s) Chromatin/genetics ; Base Sequence ; Regulatory Sequences, Nucleic Acid ; Neural Networks, Computer ; Genome
    Chemical Substances Chromatin
    Language English
    Publishing date 2023-02-01
    Publishing country United States
    Document type Journal Article
    ISSN 1940-6029
    ISSN (online) 1940-6029
    DOI 10.1007/978-1-0716-2962-8_3
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article ; Online: Direct correction of haemoglobin E β-thalassaemia using base editors.

    Badat, Mohsin / Ejaz, Ayesha / Hua, Peng / Rice, Siobhan / Zhang, Weijiao / Hentges, Lance D / Fisher, Christopher A / Denny, Nicholas / Schwessinger, Ron / Yasara, Nirmani / Roy, Noemi B A / Issa, Fadi / Roy, Andi / Telfer, Paul / Hughes, Jim / Mettananda, Sachith / Higgs, Douglas R / Davies, James O J

    Nature communications

    2023  Volume 14, Issue 1, Page(s) 2238

    Abstract: Haemoglobin E (HbE) β-thalassaemia causes approximately 50% of all severe thalassaemia worldwide; equating to around 30,000 births per year. HbE β-thalassaemia is due to a point mutation in codon 26 of the human HBB gene on one allele (GAG; glutamatic ... ...

    Abstract Haemoglobin E (HbE) β-thalassaemia causes approximately 50% of all severe thalassaemia worldwide; equating to around 30,000 births per year. HbE β-thalassaemia is due to a point mutation in codon 26 of the human HBB gene on one allele (GAG; glutamatic acid → AAG; lysine, E26K), and any mutation causing severe β-thalassaemia on the other. When inherited together in compound heterozygosity these mutations can cause a severe thalassaemic phenotype. However, if only one allele is mutated individuals are carriers for the respective mutation and have an asymptomatic phenotype (β-thalassaemia trait). Here we describe a base editing strategy which corrects the HbE mutation either to wildtype (WT) or a normal variant haemoglobin (E26G) known as Hb Aubenas and thereby recreates the asymptomatic trait phenotype. We have achieved editing efficiencies in excess of 90% in primary human CD34 + cells. We demonstrate editing of long-term repopulating haematopoietic stem cells (LT-HSCs) using serial xenotransplantation in NSG mice. We have profiled the off-target effects using a combination of circularization for in vitro reporting of cleavage effects by sequencing (CIRCLE-seq) and deep targeted capture and have developed machine-learning based methods to predict functional effects of candidate off-target mutations.
    MeSH term(s) Humans ; Animals ; Mice ; beta-Thalassemia/genetics ; Hemoglobin E/genetics ; Thalassemia/genetics ; Mutation ; Point Mutation
    Chemical Substances Hemoglobin E (9034-61-1)
    Language English
    Publishing date 2023-04-19
    Publishing country England
    Document type Journal Article ; Research Support, Non-U.S. Gov't ; Research Support, N.I.H., Extramural
    ZDB-ID 2553671-0
    ISSN 2041-1723 ; 2041-1723
    ISSN (online) 2041-1723
    ISSN 2041-1723
    DOI 10.1038/s41467-023-37604-8
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article ; Online: The chromatin remodeller ATRX facilitates diverse nuclear processes, in a stochastic manner, in both heterochromatin and euchromatin.

    Truch, Julia / Downes, Damien J / Scott, Caroline / Gür, E Ravza / Telenius, Jelena M / Repapi, Emmanouela / Schwessinger, Ron / Gosden, Matthew / Brown, Jill M / Taylor, Stephen / Cheong, Pak Leng / Hughes, Jim R / Higgs, Douglas R / Gibbons, Richard J

    Nature communications

    2022  Volume 13, Issue 1, Page(s) 3485

    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 ... ...

    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.
    MeSH term(s) Chromatin ; 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
    Chemical Substances Chromatin ; Euchromatin ; Heterochromatin ; Histones ; Nuclear Proteins ; DNA Helicases (EC 3.6.4.-) ; X-linked Nuclear Protein (EC 3.6.4.12)
    Language English
    Publishing date 2022-06-17
    Publishing country England
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 2553671-0
    ISSN 2041-1723 ; 2041-1723
    ISSN (online) 2041-1723
    ISSN 2041-1723
    DOI 10.1038/s41467-022-31194-7
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  4. Article ; Online: DeepC: predicting 3D genome folding using megabase-scale transfer learning.

    Schwessinger, Ron / Gosden, Matthew / Downes, Damien / Brown, Richard C / Oudelaar, A Marieke / Telenius, Jelena / Teh, Yee Whye / Lunter, Gerton / Hughes, Jim R

    Nature methods

    2020  Volume 17, Issue 11, Page(s) 1118–1124

    Abstract: Predicting the impact of noncoding genetic variation requires interpreting it in the context of three-dimensional genome architecture. We have developed deepC, a transfer-learning-based deep neural network that accurately predicts genome folding from ... ...

    Abstract Predicting the impact of noncoding genetic variation requires interpreting it in the context of three-dimensional genome architecture. We have developed deepC, a transfer-learning-based deep neural network that accurately predicts genome folding from megabase-scale DNA sequence. DeepC predicts domain boundaries at high resolution, learns the sequence determinants of genome folding and predicts the impact of both large-scale structural and single base-pair variations.
    MeSH term(s) Base Sequence ; CCCTC-Binding Factor/genetics ; Chromatin/genetics ; Computer Simulation ; Genome, Human/genetics ; Genomic Structural Variation ; Genomics/methods ; Humans ; Models, Genetic ; Neural Networks, Computer
    Chemical Substances CCCTC-Binding Factor ; CTCF protein, human ; Chromatin
    Language English
    Publishing date 2020-10-12
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 2169522-2
    ISSN 1548-7105 ; 1548-7091
    ISSN (online) 1548-7105
    ISSN 1548-7091
    DOI 10.1038/s41592-020-0960-3
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  5. Article: Enhancers predominantly regulate gene expression during differentiation via transcription initiation

    Larke, Martin S.C / Schwessinger, Ron / Nojima, Takayuki / Telenius, Jelena / Beagrie, Robert A / Downes, Damien J / Oudelaar, A. Marieke / Truch, Julia / Graham, Bryony / Bender, M.A / Proudfoot, Nicholas J / Higgs, Douglas R / Hughes, Jim R

    Elsevier Inc. Molecular cell. 2021 Mar. 04, v. 81, no. 5

    2021  

    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, ...

    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 β-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.
    Keywords DNA-directed RNA polymerase ; gene expression ; gene expression regulation ; genetic engineering ; mice ; transcription initiation
    Language English
    Dates of publication 2021-0304
    Size p. 983-997.e7.
    Publishing place Elsevier Inc.
    Document type Article
    Note NAL-AP-2-clean
    ZDB-ID 1415236-8
    ISSN 1097-4164 ; 1097-2765
    ISSN (online) 1097-4164
    ISSN 1097-2765
    DOI 10.1016/j.molcel.2021.01.002
    Database NAL-Catalogue (AGRICOLA)

    More links

    Kategorien

  6. Article ; Online: Enhancers predominantly regulate gene expression during differentiation via transcription initiation.

    Larke, Martin S C / Schwessinger, Ron / Nojima, Takayuki / Telenius, Jelena / Beagrie, Robert A / Downes, Damien J / Oudelaar, A Marieke / Truch, Julia / Graham, Bryony / Bender, M A / Proudfoot, Nicholas J / Higgs, Douglas R / Hughes, Jim R

    Molecular cell

    2021  Volume 81, Issue 5, Page(s) 983–997.e7

    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, ...

    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 β-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.
    MeSH term(s) Animals ; Cell Differentiation ; Enhancer Elements, Genetic ; 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 ; Promoter Regions, Genetic ; Proto-Oncogene Proteins c-myc/genetics ; Proto-Oncogene Proteins c-myc/metabolism ; RNA Polymerase II/genetics ; RNA Polymerase II/metabolism ; Signal Transduction ; Transcription Initiation, Genetic ; alpha-Globins/deficiency ; alpha-Globins/genetics ; beta-Globins/deficiency ; beta-Globins/genetics
    Chemical Substances HSP70 Heat-Shock Proteins ; HSPA1A protein, human ; MYC protein, human ; Proto-Oncogene Proteins c-myc ; alpha-Globins ; beta-Globins ; RNA Polymerase II (EC 2.7.7.-)
    Language English
    Publishing date 2021-02-03
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 1415236-8
    ISSN 1097-4164 ; 1097-2765
    ISSN (online) 1097-4164
    ISSN 1097-2765
    DOI 10.1016/j.molcel.2021.01.002
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  7. Article ; Online: Iron overload induces dysplastic erythropoiesis and features of myelodysplasia in Nrf2-deficient mice.

    Duarte, Tiago L / Lopes, Marta / Oliveira, Mónica / Santos, Ana G / Vasco, Catarina / Reis, Joana P / Antunes, Ana Rita / Gonçalves, Andreia / Chacim, Sérgio / Oliveira, Cláudia / Porto, Beatriz / Teles, Maria José / Moreira, Ana C / Silva, André M N / Schwessinger, Ron / Drakesmith, Hal / Henrique, Rui / Porto, Graça / Duarte, Delfim

    Leukemia

    2023  Volume 38, Issue 1, Page(s) 96–108

    Abstract: Iron overload (IOL) is hypothesized to contribute to dysplastic erythropoiesis. Several conditions, including myelodysplastic syndrome, thalassemia and sickle cell anemia, are characterized by ineffective erythropoiesis and IOL. Iron is pro-oxidant and ... ...

    Abstract Iron overload (IOL) is hypothesized to contribute to dysplastic erythropoiesis. Several conditions, including myelodysplastic syndrome, thalassemia and sickle cell anemia, are characterized by ineffective erythropoiesis and IOL. Iron is pro-oxidant and may participate in the pathophysiology of these conditions by increasing genomic instability and altering the microenvironment. There is, however, lack of in vivo evidence demonstrating a role of IOL and oxidative damage in dysplastic erythropoiesis. NRF2 transcription factor is the master regulator of antioxidant defenses, playing a crucial role in the cellular response to IOL in the liver. Here, we crossed Nrf2
    MeSH term(s) Animals ; Humans ; Mice ; Anemia/metabolism ; Erythropoiesis/genetics ; Hemochromatosis/genetics ; Hemochromatosis/metabolism ; Iron Overload/genetics ; Iron Overload/metabolism ; Mice, Knockout ; Myelodysplastic Syndromes/genetics ; NF-E2-Related Factor 2/genetics ; NF-E2-Related Factor 2/metabolism
    Chemical Substances NF-E2-Related Factor 2 ; Nfe2l2 protein, mouse
    Language English
    Publishing date 2023-10-19
    Publishing country England
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 807030-1
    ISSN 1476-5551 ; 0887-6924
    ISSN (online) 1476-5551
    ISSN 0887-6924
    DOI 10.1038/s41375-023-02067-9
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  8. Article ; Online: Dynamic Runx1 chromatin boundaries affect gene expression in hematopoietic development.

    Owens, Dominic D G / Anselmi, Giorgio / Oudelaar, A Marieke / Downes, Damien J / Cavallo, Alessandro / Harman, Joe R / Schwessinger, Ron / Bucakci, Akin / Greder, Lucas / de Ornellas, Sara / Jeziorska, Danuta / Telenius, Jelena / Hughes, Jim R / de Bruijn, Marella F T R

    Nature communications

    2022  Volume 13, Issue 1, Page(s) 773

    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 ... ...

    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.
    MeSH term(s) Animals ; Cell Cycle Proteins/metabolism ; Cell Differentiation ; Chromatin/metabolism ; Core Binding Factor Alpha 2 Subunit/genetics ; Core Binding Factor Alpha 2 Subunit/metabolism ; DNA/chemistry ; Gene Expression ; Gene Expression Regulation, Developmental ; Hematopoietic Stem Cells/metabolism ; Mesoderm/metabolism ; Mice ; Nucleic Acid Conformation ; Promoter Regions, Genetic
    Chemical Substances Cell Cycle Proteins ; Chromatin ; Core Binding Factor Alpha 2 Subunit ; Runx1 protein, mouse ; DNA (9007-49-2)
    Language English
    Publishing date 2022-02-09
    Publishing country England
    Document type Journal Article ; Research Support, Non-U.S. Gov't ; Research Support, U.S. Gov't, Non-P.H.S.
    ZDB-ID 2553671-0
    ISSN 2041-1723 ; 2041-1723
    ISSN (online) 2041-1723
    ISSN 2041-1723
    DOI 10.1038/s41467-022-28376-8
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  9. Article ; Online: Defining genome architecture at base-pair resolution.

    Hua, Peng / Badat, Mohsin / Hanssen, Lars L P / Hentges, Lance D / Crump, Nicholas / Downes, Damien J / Jeziorska, Danuta M / Oudelaar, A Marieke / Schwessinger, Ron / Taylor, Stephen / Milne, Thomas A / Hughes, Jim R / Higgs, Doug R / Davies, James O J

    Nature

    2021  Volume 595, Issue 7865, Page(s) 125–129

    Abstract: In higher eukaryotes, many genes are regulated by enhancers that are ... ...

    Abstract In higher eukaryotes, many genes are regulated by enhancers that are 10
    MeSH term(s) Animals ; Base Pairing/genetics ; 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 ; Genome/genetics ; Mice ; Mice, Inbred C57BL ; Organ Specificity ; Promoter Regions, Genetic/genetics ; alpha-Globins/genetics ; Cohesins
    Chemical Substances CCCTC-Binding Factor ; Cell Cycle Proteins ; Chromatin ; Chromosomal Proteins, Non-Histone ; alpha-Globins
    Language English
    Publishing date 2021-06-09
    Publishing country England
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 120714-3
    ISSN 1476-4687 ; 0028-0836
    ISSN (online) 1476-4687
    ISSN 0028-0836
    DOI 10.1038/s41586-021-03639-4
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  10. Article ; Online: Sasquatch: predicting the impact of regulatory SNPs on transcription factor binding from cell- and tissue-specific DNase footprints.

    Schwessinger, Ron / Suciu, Maria C / McGowan, Simon J / Telenius, Jelena / Taylor, Stephen / Higgs, Doug R / Hughes, Jim R

    Genome research

    2017  Volume 27, Issue 10, Page(s) 1730–1742

    Abstract: In the era of genome-wide association studies (GWAS) and personalized medicine, predicting the impact of single nucleotide polymorphisms (SNPs) in regulatory elements is an important goal. Current approaches to determine the potential of regulatory SNPs ... ...

    Abstract In the era of genome-wide association studies (GWAS) and personalized medicine, predicting the impact of single nucleotide polymorphisms (SNPs) in regulatory elements is an important goal. Current approaches to determine the potential of regulatory SNPs depend on inadequate knowledge of cell-specific DNA binding motifs. Here, we present Sasquatch, a new computational approach that uses DNase footprint data to estimate and visualize the effects of noncoding variants on transcription factor binding. Sasquatch performs a comprehensive
    MeSH term(s) DNA Footprinting/methods ; Deoxyribonucleases/chemistry ; Erythroid Cells/metabolism ; Humans ; Nucleotide Motifs ; Polymorphism, Single Nucleotide ; Predictive Value of Tests ; Response Elements ; Sequence Analysis, DNA/methods ; Transcription Factors/metabolism
    Chemical Substances Transcription Factors ; Deoxyribonucleases (EC 3.1.-)
    Language English
    Publishing date 2017-09-13
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 1284872-4
    ISSN 1549-5469 ; 1088-9051 ; 1054-9803
    ISSN (online) 1549-5469
    ISSN 1088-9051 ; 1054-9803
    DOI 10.1101/gr.220202.117
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top