Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Am J Hum Genet ; 111(6): 1206-1221, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38772379

ABSTRACT

Utilizing trio whole-exome sequencing and a gene matching approach, we identified a cohort of 18 male individuals from 17 families with hemizygous variants in KCND1, including two de novo missense variants, three maternally inherited protein-truncating variants, and 12 maternally inherited missense variants. Affected subjects present with a neurodevelopmental disorder characterized by diverse neurological abnormalities, mostly delays in different developmental domains, but also distinct neuropsychiatric signs and epilepsy. Heterozygous carrier mothers are clinically unaffected. KCND1 encodes the α-subunit of Kv4.1 voltage-gated potassium channels. All variant-associated amino acid substitutions affect either the cytoplasmic N- or C-terminus of the channel protein except for two occurring in transmembrane segments 1 and 4. Kv4.1 channels were functionally characterized in the absence and presence of auxiliary ß subunits. Variant-specific alterations of biophysical channel properties were diverse and varied in magnitude. Genetic data analysis in combination with our functional assessment shows that Kv4.1 channel dysfunction is involved in the pathogenesis of an X-linked neurodevelopmental disorder frequently associated with a variable neuropsychiatric clinical phenotype.


Subject(s)
Neurodevelopmental Disorders , Adolescent , Adult , Child , Child, Preschool , Female , Humans , Infant , Male , Epilepsy/genetics , Exome Sequencing , Genetic Diseases, X-Linked/genetics , Heterozygote , Mutation, Missense/genetics , Neurodevelopmental Disorders/genetics , Pedigree , Phenotype , Shal Potassium Channels/genetics
2.
Hum Mutat ; 43(12): 1844-1851, 2022 12.
Article in English | MEDLINE | ID: mdl-35904126

ABSTRACT

TATA-binding protein associated factor 4 (TAF4) is a subunit of the Transcription Factor IID (TFIID) complex, a central player in transcription initiation. Other members of this multimeric complex have been implicated previously as monogenic disease genes in human developmental disorders. TAF4 has not been described to date as a monogenic disease gene. We here present a cohort of eight individuals, each carrying de novo putative loss-of-function (pLoF) variants in TAF4 and expressing phenotypes consistent with a neuro-developmental disorder (NDD). Common features include intellectual disability, abnormal behavior, and facial dysmorphisms. We propose TAF4 as a novel dominant disease gene for NDD, and coin this novel disorder "TAF4-related NDD" (T4NDD). We place T4NDD in the context of other disorders related to TFIID subunits, revealing shared features of T4NDD with other TAF-opathies.


Subject(s)
Neurodevelopmental Disorders , TATA-Binding Protein Associated Factors , Transcription Factor TFIID , Child , Humans , Developmental Disabilities/genetics , Neurodevelopmental Disorders/diagnosis , Neurodevelopmental Disorders/genetics , Phenotype , TATA-Binding Protein Associated Factors/genetics , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/genetics , Transcription Factor TFIID/metabolism
3.
Genet Med ; 24(7): 1583-1591, 2022 07.
Article in English | MEDLINE | ID: mdl-35499524

ABSTRACT

PURPOSE: CTR9 is a subunit of the PAF1 complex (PAF1C) that plays a crucial role in transcription regulation by binding CTR9 to RNA polymerase II. It is involved in transcription-coupled histone modification through promoting H3K4 and H3K36 methylation. We describe the clinical and molecular studies in 13 probands, harboring likely pathogenic CTR9 missense variants, collected through GeneMatcher. METHODS: Exome sequencing was performed in all individuals. CTR9 variants were assessed through 3-dimensional modeling of the activated human transcription complex Pol II-DSIF-PAF-SPT6 and the PAF1/CTR9 complex. H3K4/H3K36 methylation analysis, mitophagy assessment based on tetramethylrhodamine ethyl ester perchlorate immunofluorescence, and RNA-sequencing in skin fibroblasts from 4 patients was performed. RESULTS: Common clinical findings were variable degrees of intellectual disability, hypotonia, joint hyperlaxity, speech delay, coordination problems, tremor, and autism spectrum disorder. Mild dysmorphism and cardiac anomalies were less frequent. For 11 CTR9 variants, de novo occurrence was shown. Three-dimensional modeling predicted a likely disruptive effect of the variants on local CTR9 structure and protein interaction. Additional studies in fibroblasts did not unveil the downstream functional consequences of the identified variants. CONCLUSION: We describe a neurodevelopmental disorder caused by (mainly) de novo variants in CTR9, likely affecting PAF1C function.


Subject(s)
Autism Spectrum Disorder , Intellectual Disability , Neurodevelopmental Disorders , Phosphoproteins , Transcription Factors , Gene Expression Regulation , Heterozygote , Humans , Intellectual Disability/genetics , Neurodevelopmental Disorders/genetics , Phosphoproteins/genetics , Transcription Factors/genetics
4.
Genet Med ; 24(8): 1774-1780, 2022 08.
Article in English | MEDLINE | ID: mdl-35567594

ABSTRACT

PURPOSE: SRRM2 encodes the SRm300 protein, a splicing factor of the SR-related protein family characterized by its serine- and arginine-enriched domains. It promotes interactions between messenger RNA and the spliceosome catalytic machinery. This gene, predicted to be highly intolerant to loss of function (LoF) and very conserved through evolution, has not been previously reported in constitutive human disease. METHODS: Among the 1000 probands studied with developmental delay and intellectual disability in our database, we found 2 patients with de novo LoF variants in SRRM2. Additional families were identified through GeneMatcher. RESULTS: Here, we report on 22 patients with LoF variants in SRRM2 and provide a description of the phenotype. Molecular analysis identified 12 frameshift variants, 8 nonsense variants, and 2 microdeletions of 66 kb and 270 kb. The patients presented with a mild developmental delay, predominant speech delay, autistic or attention-deficit/hyperactivity disorder features, overfriendliness, generalized hypotonia, overweight, and dysmorphic facial features. Intellectual disability was variable and mild when present. CONCLUSION: We established SRRM2 as a gene responsible for a rare neurodevelopmental disease.


Subject(s)
Intellectual Disability , Neurodevelopmental Disorders , RNA-Binding Proteins/genetics , Child , Developmental Disabilities/genetics , Humans , Intellectual Disability/genetics , Muscle Hypotonia/genetics , Neurodevelopmental Disorders/genetics , Phenotype
5.
Cell Stem Cell ; 28(10): 1822-1837.e10, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34129813

ABSTRACT

AXIN2 and LGR5 mark intestinal stem cells (ISCs) that require WNT/ß-Catenin signaling for constant homeostatic proliferation. In contrast, AXIN2/LGR5+ pericentral hepatocytes show low proliferation rates despite a WNT/ß-Catenin activity gradient required for metabolic liver zonation. The mechanisms restricting proliferation in AXIN2+ hepatocytes and metabolic gene expression in AXIN2+ ISCs remained elusive. We now show that restricted chromatin accessibility in ISCs prevents the expression of ß-Catenin-regulated metabolic enzymes, whereas fine-tuning of WNT/ß-Catenin activity by ZNRF3 and RNF43 restricts proliferation in chromatin-permissive AXIN2+ hepatocytes, while preserving metabolic function. ZNRF3 deletion promotes hepatocyte proliferation, which in turn becomes limited by RNF43 upregulation. Concomitant deletion of RNF43 in ZNRF3 mutant mice results in metabolic reprogramming of periportal hepatocytes and induces clonal expansion in a subset of hepatocytes, ultimately promoting liver tumors. Together, ZNRF3 and RNF43 cooperate to safeguard liver homeostasis by spatially and temporally restricting WNT/ß-Catenin activity, balancing metabolic function and hepatocyte proliferation.


Subject(s)
Liver , Ubiquitin-Protein Ligases/genetics , Animals , Cell Proliferation , Hepatocytes/metabolism , Liver/growth & development , Liver/metabolism , Mice , Stem Cells/metabolism , Ubiquitin-Protein Ligases/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism
6.
Cell Rep ; 31(11): 107767, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32553152

ABSTRACT

The mammalian precerebellar pontine nucleus (PN) has a main role in relaying cortical information to the cerebellum. The molecular determinants establishing ordered connectivity patterns between cortical afferents and precerebellar neurons are largely unknown. We show that expression of Hox5 transcription factors is induced in specific subsets of postmitotic PN neurons at migration onset. Hox5 induction is achieved by response to retinoic acid signaling, resulting in Jmjd3-dependent derepression of Polycomb chromatin and 3D conformational changes. Hoxa5 drives neurons to settle posteriorly in the PN, where they are monosynaptically targeted by cortical neuron subsets mainly carrying limb somatosensation. Furthermore, Hoxa5 postmigratory ectopic expression in PN neurons is sufficient to attract cortical somatosensory inputs regardless of position and avoid visual afferents. Transcriptome analysis further suggests that Hoxa5 is involved in circuit formation. Thus, Hoxa5 coordinates postmitotic specification, migration, settling position, and sub-circuit assembly of PN neuron subsets in the cortico-cerebellar pathway.


Subject(s)
Cerebellum/metabolism , Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/metabolism , Neurons/metabolism , Transcription Factors/metabolism , Animals , Cell Movement/physiology , Cerebral Cortex/metabolism
7.
Cell Stem Cell ; 26(1): 97-107.e6, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31866224

ABSTRACT

The existence of specialized liver stem cell populations, including AXIN2+ pericentral hepatocytes, that safeguard homeostasis and repair has been controversial. Here, using AXIN2 lineage tracing in BAC-transgenic mice, we confirm the regenerative potential of intestinal stem cells (ISCs) but find limited roles for pericentral hepatocytes in liver parenchyma homeostasis. Liver regrowth following partial hepatectomy is enabled by proliferation of hepatocytes throughout the liver, rather than by a pericentral population. Periportal hepatocyte injury triggers local repair as well as auxiliary proliferation in all liver zones. DTA-mediated ablation of AXIN2+ pericentral hepatocytes transiently disrupts this zone, which is reestablished by conversion of pericentral vein-juxtaposed glutamine synthetase (GS)- hepatocytes into GS+ hepatocytes and by compensatory proliferation of hepatocytes across liver zones. These findings show hepatocytes throughout the liver can upregulate AXIN2 and LGR5 after injury and contribute to liver regeneration on demand, without zonal dominance by a putative pericentral stem cell population.


Subject(s)
Hepatocytes , Liver , Animals , Axin Protein , Homeostasis , Liver Regeneration , Mice , Stem Cells
8.
Nat Commun ; 10(1): 3739, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31431624

ABSTRACT

Transcription factor networks shape the gene expression programs responsible for normal cell identity and pathogenic state. Using Core Regulatory Circuitry analysis (CRC), we identify PAX8 as a candidate oncogene in Renal Cell Carcinoma (RCC) cells. Validation of large-scale functional genomic screens confirms that PAX8 silencing leads to decreased proliferation of RCC cell lines. Epigenomic analyses of PAX8-dependent cistrome demonstrate that PAX8 largely occupies active enhancer elements controlling genes involved in various metabolic pathways. We selected the ferroxidase Ceruloplasmin (CP) as an exemplary gene to dissect PAX8 molecular functions. PAX8 recruits histone acetylation activity at bound enhancers looping onto the CP promoter. Importantly, CP expression correlates with sensitivity to PAX8 silencing and identifies a subset of RCC cases with poor survival. Our data identifies PAX8 as a candidate oncogene in RCC and provides a potential biomarker to monitor its activity.


Subject(s)
Carcinoma, Renal Cell/genetics , Ceruloplasmin/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Neoplastic/genetics , Kidney Neoplasms/genetics , PAX8 Transcription Factor/genetics , Acetylation , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Ceruloplasmin/metabolism , Histones/metabolism , Humans , Promoter Regions, Genetic/genetics , RNA Interference , RNA, Small Interfering/genetics
9.
Elife ; 62017 11 06.
Article in English | MEDLINE | ID: mdl-29106373

ABSTRACT

High-resolution daylight vision is mediated by cone photoreceptors. The molecular program responsible for the formation of their light sensor, the outer segment, is not well understood. We correlated daily changes in ultrastructure and gene expression in postmitotic mouse cones, between birth and eye opening, using serial block-face electron microscopy (EM) and RNA sequencing. Outer segments appeared rapidly at postnatal day six and their appearance coincided with a switch in gene expression. The switch affected over 14% of all expressed genes. Genes that switched off were rich in transcription factors and neurogenic genes. Those that switched on contained genes relevant for cone function. Chromatin rearrangements in enhancer regions occurred before the switch was completed, but not after. We provide a resource comprised of correlated EM, RNAseq, and ATACseq data, showing that the growth of a key compartment of a postmitotic cell involves an extensive switch in gene expression and chromatin accessibility.


Subject(s)
Cell Differentiation , Gene Expression Regulation , Retina/growth & development , Retinal Cone Photoreceptor Cells/physiology , Transcription, Genetic , Animals , Gene Expression Profiling , Mice , Microscopy, Electron , Retinal Cone Photoreceptor Cells/ultrastructure , Sequence Analysis, RNA
10.
Science ; 355(6332)2017 03 31.
Article in English | MEDLINE | ID: mdl-28360266

ABSTRACT

The cranial neural crest cells are multipotent cells that provide head skeletogenic mesenchyme and are crucial for craniofacial patterning. We analyzed the chromatin landscapes of mouse cranial neural crest subpopulations in vivo. Early postmigratory subpopulations contributing to distinct mouse craniofacial structures displayed similar chromatin accessibility patterns yet differed transcriptionally. Accessible promoters and enhancers of differentially silenced genes carried H3K27me3/H3K4me2 bivalent chromatin marks embedded in large enhancer of zeste homolog 2-dependent Polycomb domains, indicating transcriptional poising. These postmigratory bivalent chromatin regions were already present in premigratory progenitors. At Polycomb domains, H3K27me3 antagonized H3K4me2 deposition, which was restricted to accessible sites. Thus, bivalent Polycomb domains provide a chromatin template for the regulation of cranial neural crest cell positional identity in vivo, contributing insights into the epigenetic regulation of face morphogenesis.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/genetics , Epigenesis, Genetic , Face/embryology , Gene Expression Regulation, Developmental , Neural Crest/embryology , Skull/embryology , Animals , Cell Movement , Cell Plasticity/genetics , Chromatin/metabolism , Enhancer Elements, Genetic , Homeodomain Proteins/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Mice , Neural Crest/cytology , Promoter Regions, Genetic , Protein Domains
11.
Genome Res ; 27(6): 922-933, 2017 06.
Article in English | MEDLINE | ID: mdl-28341771

ABSTRACT

The spatial arrangement of chromatin is linked to the regulation of nuclear processes. One striking aspect of nuclear organization is the spatial segregation of heterochromatic and euchromatic domains. The mechanisms of this chromatin segregation are still poorly understood. In this work, we investigated the link between the primary genomic sequence and chromatin domains. We analyzed the spatial intranuclear arrangement of a human artificial chromosome (HAC) in a xenospecific mouse background in comparison to an orthologous region of native mouse chromosome. The two orthologous regions include segments that can be assigned to three major chromatin classes according to their gene abundance and repeat repertoire: (1) gene-rich and SINE-rich euchromatin; (2) gene-poor and LINE/LTR-rich heterochromatin; and (3) gene-depleted and satellite DNA-containing constitutive heterochromatin. We show, using fluorescence in situ hybridization (FISH) and 4C-seq technologies, that chromatin segments ranging from 0.6 to 3 Mb cluster with segments of the same chromatin class. As a consequence, the chromatin segments acquire corresponding positions in the nucleus irrespective of their chromosomal context, thereby strongly suggesting that this is their autonomous property. Interactions with the nuclear lamina, although largely retained in the HAC, reveal less autonomy. Taken together, our results suggest that building of a functional nucleus is largely a self-organizing process based on mutual recognition of chromosome segments belonging to the major chromatin classes.


Subject(s)
Cell Nucleus/genetics , Chromosomes, Artificial, Human/metabolism , Euchromatin/metabolism , Fibroblasts/metabolism , Heterochromatin/metabolism , Retina/metabolism , Animals , Cell Line, Transformed , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Chromosomes, Artificial, Human/ultrastructure , Euchromatin/classification , Euchromatin/ultrastructure , Fibroblasts/ultrastructure , Gene Expression Profiling , Gene Expression Regulation , Heterochromatin/classification , Heterochromatin/ultrastructure , Humans , In Situ Hybridization, Fluorescence , Mice , Primary Cell Culture , Retina/ultrastructure
12.
Mol Cell ; 60(4): 676-84, 2015 Nov 19.
Article in English | MEDLINE | ID: mdl-26527277

ABSTRACT

CCCTC-binding factor (CTCF) is an architectural protein involved in the three-dimensional (3D) organization of chromatin. In this study, we assayed the 3D genomic contact profiles of a large number of CTCF binding sites with high-resolution 4C-seq. As recently reported, our data also suggest that chromatin loops preferentially form between CTCF binding sites oriented in a convergent manner. To directly test this, we used CRISPR/Cas9 genome editing to delete core CTCF binding sites in three loci, including the CTCF site in the Sox2 super-enhancer. In all instances, CTCF and cohesin recruitment were lost, and chromatin loops with distal, convergent CTCF sites were disrupted or destabilized. Re-insertion of oppositely oriented CTCF recognition sequences restored CTCF and cohesin recruitment, but did not re-establish chromatin loops. We conclude that CTCF binding polarity plays a functional role in the formation of higher-order chromatin structure.


Subject(s)
Chromatin/chemistry , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Animals , Binding Sites , CCCTC-Binding Factor , CRISPR-Cas Systems , Cell Cycle Proteins/metabolism , Cell Line , Chromosomal Proteins, Non-Histone/metabolism , Embryonic Stem Cells/cytology , Mice , Protein Binding , Cohesins
13.
Immunity ; 39(2): 229-44, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23973221

ABSTRACT

The immunoglobulin heavy-chain (Igh) locus undergoes large-scale contraction in pro-B cells, which facilitates VH-DJH recombination by juxtaposing distal VH genes next to the DJH-rearranged gene segment in the 3' proximal Igh domain. By using high-resolution mapping of long-range interactions, we demonstrate that local interaction domains established the three-dimensional structure of the extended Igh locus in lymphoid progenitors. In pro-B cells, these local domains engaged in long-range interactions across the Igh locus, which depend on the regulators Pax5, YY1, and CTCF. The large VH gene cluster underwent flexible long-range interactions with the more rigidly structured proximal domain, which probably ensures similar participation of all VH genes in VH-DJH recombination to generate a diverse antibody repertoire. These long-range interactions appear to be an intrinsic feature of the VH gene cluster, because they are still generated upon mutation of the Eµ enhancer, IGCR1 insulator, or 3' regulatory region in the proximal Igh domain.


Subject(s)
Antibody Diversity/genetics , Gene Rearrangement, B-Lymphocyte, Heavy Chain/genetics , Genes, Immunoglobulin Heavy Chain , Immunoglobulin Variable Region/genetics , Precursor Cells, B-Lymphoid/immunology , Animals , Base Sequence , Binding Sites , CCCTC-Binding Factor , Chromosome Mapping , Gene Rearrangement , Mice , Mice, Inbred C57BL , PAX5 Transcription Factor/metabolism , Protein Binding , Repressor Proteins/metabolism , Sequence Analysis, DNA , YY1 Transcription Factor/metabolism
14.
Nucleic Acids Res ; 41(14): 6905-16, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23748562

ABSTRACT

In developing B cells, the immunoglobulin heavy chain (IgH) locus is thought to move from repressive to permissive chromatin compartments to facilitate its scheduled rearrangement. In mature B cells, maintenance of allelic exclusion has been proposed to involve recruitment of the non-productive IgH allele to pericentromeric heterochromatin. Here, we used an allele-specific chromosome conformation capture combined with sequencing (4C-seq) approach to unambigously follow the individual IgH alleles in mature B lymphocytes. Despite their physical and functional difference, productive and non-productive IgH alleles in B cells and unrearranged IgH alleles in T cells share many chromosomal contacts and largely reside in active chromatin. In brain, however, the locus resides in a different repressive environment. We conclude that IgH adopts a lymphoid-specific nuclear location that is, however, unrelated to maintenance of allelic exclusion. We additionally find that in mature B cells-but not in T cells-the distal VH regions of both IgH alleles position themselves away from active chromatin. This, we speculate, may help to restrict enhancer activity to the productively rearranged VH promoter element.


Subject(s)
Alleles , B-Lymphocytes/immunology , Genes, Immunoglobulin Heavy Chain , Animals , Cell Nucleus/chemistry , Chromatin/chemistry , Chromosomes, Mammalian , Genetic Loci , Immunoglobulin Heavy Chains/analysis , Immunoglobulin Heavy Chains/biosynthesis , Immunoglobulin Variable Region/genetics , Mice , Recombination, Genetic , Sequence Analysis, DNA , Spleen/immunology , T-Lymphocytes/immunology , Transcription, Genetic
15.
Philos Trans R Soc Lond B Biol Sci ; 368(1620): 20120369, 2013.
Article in English | MEDLINE | ID: mdl-23650640

ABSTRACT

CTCF has it all. The transcription factor binds to tens of thousands of genomic sites, some tissue-specific, others ultra-conserved. It can act as a transcriptional activator, repressor and insulator, and it can pause transcription. CTCF binds at chromatin domain boundaries, at enhancers and gene promoters, and inside gene bodies. It can attract many other transcription factors to chromatin, including tissue-specific transcriptional activators, repressors, cohesin and RNA polymerase II, and it forms chromatin loops. Yet, or perhaps therefore, CTCF's exact function at a given genomic site is unpredictable. It appears to be determined by the associated transcription factors, by the location of the binding site relative to the transcriptional start site of a gene, and by the site's engagement in chromatin loops with other CTCF-binding sites, enhancers or gene promoters. Here, we will discuss genome-wide features of CTCF binding events, as well as locus-specific functions of this remarkable transcription factor.


Subject(s)
Chromatin/metabolism , Genetic Loci , Repressor Proteins/metabolism , Animals , Binding Sites , CCCTC-Binding Factor , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromatin/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Enhancer Elements, Genetic , Humans , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Promoter Regions, Genetic , Protein Binding , Repressor Proteins/genetics , Transcriptional Activation , beta-Globins/genetics , beta-Globins/metabolism , Cohesins
16.
Front Genet ; 3: 217, 2012.
Article in English | MEDLINE | ID: mdl-23087710

ABSTRACT

Technological developments and intense research over the last years have led to a better understanding of the 3D structure of the genome and its influence on genome function inside the cell nucleus. We will summarize topological studies performed on four model gene loci: the α- and ß-globin gene loci, the antigen receptor loci, the imprinted H19-Igf2 locus and the Hox gene clusters. Collectively, these studies show that regulatory DNA sequences physically contact genes to control their transcription. Proteins set up the 3D configuration of the genome and we will discuss the roles of the key structural organizers CTCF and cohesin, the nuclear lamina and the transcription machinery. Finally, genes adopt non-random positions in the nuclear interior. We will review studies on gene positioning and propose that cell-specific genome conformations can juxtapose a regulatory sequence on one chromosome to a responsive gene on another chromosome to cause altered gene expression in subpopulations of cells.

17.
Nat Methods ; 9(10): 969-72, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22961246

ABSTRACT

Regulatory DNA elements can control the expression of distant genes via physical interactions. Here we present a cost-effective methodology and computational analysis pipeline for robust characterization of the physical organization around selected promoters and other functional elements using chromosome conformation capture combined with high-throughput sequencing (4C-seq). Our approach can be multiplexed and routinely integrated with other functional genomics assays to facilitate physical characterization of gene regulation.


Subject(s)
DNA/chemistry , High-Throughput Nucleotide Sequencing/methods , Gene Expression Regulation , Locus Control Region , Polymerase Chain Reaction , Promoter Regions, Genetic
18.
Methods Enzymol ; 513: 89-112, 2012.
Article in English | MEDLINE | ID: mdl-22929766

ABSTRACT

Chromosome conformation capture (3C) technology and its genome-wide derivatives have revolutionized our knowledge on chromatin folding and nuclear organization. 4C-seq Technology combines 3C principles with high-throughput sequencing (4C-seq) to enable for unbiased genome-wide screens for DNA contacts made by single genomic sites of interest. Here, we discuss in detail the design, application, and data analysis of 4C-seq experiments. Based on many hundreds of different 4C-seq experiments, we define criteria to assess data quality and show how different restriction enzymes and cross-linking conditions affect results. We describe in detail the mapping strategy of 4C-seq reads and show advanced strategies for data analysis.


Subject(s)
Chromatin/chemistry , Chromosome Mapping/methods , DNA/chemistry , Sequence Analysis, DNA/methods , Statistics as Topic/methods , Chromatin Assembly and Disassembly , Cross-Linking Reagents , DNA/genetics , DNA Restriction Enzymes/chemistry , Formaldehyde/chemistry , High-Throughput Nucleotide Sequencing/methods , Nucleic Acid Conformation , Polymerase Chain Reaction/methods , beta-Globins/chemistry , beta-Globins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...