Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Clin Epigenetics ; 9: 89, 2017.
Article in English | MEDLINE | ID: mdl-28855971

ABSTRACT

BACKGROUND: Cornelia de Lange syndrome (CdLS) is a multisystem developmental disorder frequently associated with heterozygous loss-of-function mutations of Nipped-B-like (NIPBL), the human homolog of Drosophila Nipped-B. NIPBL loads cohesin onto chromatin. Cohesin mediates sister chromatid cohesion important for mitosis but is also increasingly recognized as a regulator of gene expression. In CdLS patient cells and animal models, expression changes of multiple genes with little or no sister chromatid cohesion defect suggests that disruption of gene regulation underlies this disorder. However, the effect of NIPBL haploinsufficiency on cohesin binding, and how this relates to the clinical presentation of CdLS, has not been fully investigated. Nipbl haploinsufficiency causes CdLS-like phenotype in mice. We examined genome-wide cohesin binding and its relationship to gene expression using mouse embryonic fibroblasts (MEFs) from Nipbl+/- mice that recapitulate the CdLS phenotype. RESULTS: We found a global decrease in cohesin binding, including at CCCTC-binding factor (CTCF) binding sites and repeat regions. Cohesin-bound genes were found to be enriched for histone H3 lysine 4 trimethylation (H3K4me3) at their promoters; were disproportionately downregulated in Nipbl mutant MEFs; and displayed evidence of reduced promoter-enhancer interaction. The results suggest that gene activation is the primary cohesin function sensitive to Nipbl reduction. Over 50% of significantly dysregulated transcripts in mutant MEFs come from cohesin target genes, including genes involved in adipogenesis that have been implicated in contributing to the CdLS phenotype. CONCLUSIONS: Decreased cohesin binding at the gene regions is directly linked to disease-specific expression changes. Taken together, our Nipbl haploinsufficiency model allows us to analyze the dosage effect of cohesin loading on CdLS development.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , De Lange Syndrome/genetics , Gene Expression Profiling/methods , Haploinsufficiency , Proteins/genetics , Animals , Binding Sites , CCCTC-Binding Factor/chemistry , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism , DNA Methylation , De Lange Syndrome/metabolism , Disease Models, Animal , Gene Expression , Gene Expression Regulation , Genome-Wide Association Study , Humans , Mice , Promoter Regions, Genetic , Protein Binding , Transcriptional Activation , Cohesins
2.
Development ; 141(23): 4537-47, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25359723

ABSTRACT

Nodal/TGFß signaling regulates diverse biological responses. By combining RNA-seq on Foxh1 and Nodal signaling loss-of-function embryos with ChIP-seq of Foxh1 and Smad2/3, we report a comprehensive genome-wide interaction between Foxh1 and Smad2/3 in mediating Nodal signaling during vertebrate mesendoderm development. This study significantly increases the total number of Nodal target genes regulated by Foxh1 and Smad2/3, and reinforces the notion that Foxh1-Smad2/3-mediated Nodal signaling directly coordinates the expression of a cohort of genes involved in the control of gene transcription, signaling pathway modulation and tissue morphogenesis during gastrulation. We also show that Foxh1 may function independently of Nodal signaling, in addition to its role as a transcription factor mediating Nodal signaling via Smad2/3. Finally, we propose an evolutionarily conserved interaction between Foxh1 and PouV, a mechanism observed in Pou5f1-mediated regulation of pluripotency in human embryonic stem and epiblast cells.


Subject(s)
Endoderm/embryology , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental/physiology , Mesoderm/embryology , Transforming Growth Factor beta/metabolism , Xenopus Proteins/metabolism , Xenopus/embryology , Animals , Blotting, Western , Chromatin Immunoprecipitation , Computational Biology , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Developmental/genetics , Gene Knockdown Techniques , Humans , Immunoprecipitation , Morpholinos/genetics , Nodal Protein/genetics , Nodal Protein/metabolism , Real-Time Polymerase Chain Reaction , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Statistics, Nonparametric , Xenopus Proteins/genetics
3.
PLoS One ; 9(3): e92317, 2014.
Article in English | MEDLINE | ID: mdl-24651522

ABSTRACT

BACKGROUND: Deregulation of canonical Wnt/CTNNB1 (beta-catenin) pathway is one of the earliest events in the pathogenesis of colon cancer. Mutations in APC or CTNNB1 are highly frequent in colon cancer and cause aberrant stabilization of CTNNB1, which activates the transcription of Wnt target genes by binding to chromatin via the TCF/LEF transcription factors. Here we report an integrative analysis of genome-wide chromatin occupancy of CTNNB1 by chromatin immunoprecipitation coupled with high-throughput sequencing (ChIP-seq) and gene expression profiling by microarray analysis upon RNAi-mediated knockdown of CTNNB1 in colon cancer cells. RESULTS: We observed 3629 CTNNB1 binding peaks across the genome and a significant correlation between CTNNB1 binding and knockdown-induced gene expression change. Our integrative analysis led to the discovery of a direct Wnt target signature composed of 162 genes. Gene ontology analysis of this signature revealed a significant enrichment of Wnt pathway genes, suggesting multiple feedback regulations of the pathway. We provide evidence that this gene signature partially overlaps with the Lgr5+ intestinal stem cell signature, and is significantly enriched in normal intestinal stem cells as well as in clinical colorectal cancer samples. Interestingly, while the expression of the CTNNB1 target gene set does not correlate with survival, elevated expression of negative feedback regulators within the signature predicts better prognosis. CONCLUSION: Our data provide a genome-wide view of chromatin occupancy and gene regulation of Wnt/CTNNB1 signaling in colon cancer cells.


Subject(s)
Chromatin Immunoprecipitation , Colonic Neoplasms/genetics , Gene Expression Profiling , Intestines/pathology , Oligonucleotide Array Sequence Analysis , Stem Cells/metabolism , beta Catenin/metabolism , Cell Line, Tumor , Cell Survival , Chromatin/metabolism , Feedback, Physiological , Gene Expression Regulation, Neoplastic , Gene Ontology , Humans , Kaplan-Meier Estimate , Reproducibility of Results , Software , Stem Cells/pathology , Wnt Signaling Pathway/genetics
4.
Genesis ; 51(12): 827-34, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24123579

ABSTRACT

Gene inactivation is an important tool for correlation of phenotypic and genomic data, allowing researchers to infer normal gene function based on the phenotype when the gene is impaired. New and better approaches are needed to overcome the shortfalls of existing methods for any significant acceleration of scientific progress. We have adapted the CRISPR/Cas system for use in Xenopus tropicalis and report on the efficient creation of mutations in the gene encoding the enzyme tyrosinase, which is responsible for oculocutaneous albinism. Biallelic mutation of this gene was detected in the F0 generation, suggesting targeting efficiencies similar to that of TALENs. We also find that off-target mutagenesis seems to be negligible, and therefore, CRISPR/Cas may be a useful system for creating genome modifications in this important model organism.


Subject(s)
CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Monophenol Monooxygenase/genetics , Xenopus Proteins/genetics , Xenopus/embryology , Xenopus/genetics , Albinism/genetics , Alleles , Animals , Base Sequence , Embryo, Nonmammalian/metabolism , Female , Gene Dosage , Gene Knockout Techniques , Genome , INDEL Mutation , Monophenol Monooxygenase/metabolism , Phenotype , Xenopus Proteins/metabolism
5.
BMC Bioinformatics ; 14 Suppl 5: S4, 2013.
Article in English | MEDLINE | ID: mdl-23734743

ABSTRACT

New biological techniques and technological advances in high-throughput sequencing are paving the way for systematic, comprehensive annotation of many genomes, allowing differences between cell types or between disease/normal tissues to be determined with unprecedented breadth. Epigenetic modifications have been shown to exhibit rich diversity between cell types, correlate tightly with cell-type specific gene expression, and changes in epigenetic modifications have been implicated in several diseases. Previous attempts to understand chromatin state have focused on identifying combinations of epigenetic modification, but in cases of multiple cell types, have not considered the lineage of the cells in question.We present a Bayesian network that uses epigenetic modifications to simultaneously model 1) chromatin mark combinations that give rise to different chromatin states and 2) propensities for transitions between chromatin states through differentiation or disease progression. We apply our model to a recent dataset of histone modifications, covering nine human cell types with nine epigenetic modifications measured for each. Since exact inference in this model is intractable for all the scale of the datasets, we develop several variational approximations and explore their accuracy. Our method exhibits several desirable features including improved accuracy of inferring chromatin states, improved handling of missing data, and linear scaling with dataset size. The source code for our model is available at http:// http://github.com/uci-cbcl/tree-hmm.


Subject(s)
Chromatin/metabolism , Epigenesis, Genetic , Bayes Theorem , Cell Differentiation/genetics , Chromatin Assembly and Disassembly , Chromatin Immunoprecipitation , Disease Progression , Genome, Human , Histones/metabolism , Humans , Markov Chains , Models, Statistical , Sequence Analysis, DNA
6.
Proc Natl Acad Sci U S A ; 109(46): 18773-8, 2012 Nov 13.
Article in English | MEDLINE | ID: mdl-23112178

ABSTRACT

Cleavage stimulation factor 64 kDa (CstF64) is an essential pre-mRNA 3' processing factor and an important regulator of alternative polyadenylation (APA). Here we characterized CstF64-RNA interactions in vivo at the transcriptome level and investigated the role of CstF64 in global APA regulation through individual nucleotide resolution UV crosslinking and immunoprecipitation sequencing and direct RNA sequencing analyses. We observed highly specific CstF64-RNA interactions at poly(A) sites (PASs), and we provide evidence that such interactions are widely variable in affinity and may be differentially required for PAS recognition. Depletion of CstF64 by RNAi has a relatively small effect on the global APA profile, but codepletion of the CstF64 paralog CstF64τ leads to greater APA changes, most of which are characterized by the increased relative use of distal PASs. Finally, we found that CstF64 binds to thousands of dormant intronic PASs that are suppressed, at least in part, by U1 small nuclear ribonucleoproteins. Taken together, our findings provide insight into the mechanisms of PAS recognition and identify CstF64 as an important global regulator of APA.


Subject(s)
Polyadenylation/physiology , RNA 3' Polyadenylation Signals/physiology , RNA-Binding Proteins/metabolism , Transcriptome/physiology , Cleavage Stimulation Factor , HeLa Cells , Humans , Protein Binding , RNA-Binding Proteins/genetics , Ribonucleoprotein, U1 Small Nuclear/genetics , Ribonucleoprotein, U1 Small Nuclear/metabolism
7.
BMC Genomics ; 13: 51, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22296850

ABSTRACT

BACKGROUND: In a previous genome-wide analysis of FXR binding to hepatic chromatin, we noticed that an extra nuclear receptor (NR) half-site was co-enriched close to the FXR binding IR-1 elements and we provided limited support that the monomeric LRH-1 receptor that binds to NR half-sites might function together with FXR to activate gene expression. RESULTS: To analyze the global pattern for LRH-1 binding and to determine whether it might associate with FXR on a whole genome-wide scale, we analyzed LRH-1 binding to the entire hepatic genome using a non-biased genome-wide ChIP-seq approach. We identified over 10,600 LRH-1 binding sites in hepatic chromatin and over 20% were located within 2 kb of the 5' end of a known mouse gene. Additionally, the results demonstrate that a significant fraction of the genome sites occupied by LRH-1 are located close to FXR binding sites revealed in our earlier study. A Gene ontology analysis revealed that genes preferentially enriched in the LRH-1/FXR overlapping gene set are related to lipid metabolism. These results demonstrate that LRH-1 recruits FXR to lipid metabolic genes. A significant fraction of FXR binding peaks also contain a nuclear receptor half-site that does not bind LRH-1 suggesting that additional monomeric nuclear receptors such as RORs and NR4As family members may also target FXR to other pathway selective genes related to other areas of metabolism such as glucose metabolism where FXR has also been shown to play an important role. CONCLUSION: These results document an important role for LRH-1 in hepatic metabolism through acting predominantly at proximal promoter sites and working in concert with additional nuclear receptors that bind to neighboring sites.


Subject(s)
Gene Expression Regulation , Genome-Wide Association Study , Lipid Metabolism/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Binding Sites , Chromatin Immunoprecipitation , Genome , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Protein Binding , Sequence Analysis, DNA
8.
J Comput Biol ; 18(11): 1495-505, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22035330

ABSTRACT

High-throughput sequencing coupled to chromatin immunoprecipitation (ChIP-Seq) is widely used in characterizing genome-wide binding patterns of transcription factors, cofactors, chromatin modifiers, and other DNA binding proteins. A key step in ChIP-Seq data analysis is to map short reads from high-throughput sequencing to a reference genome and identify peak regions enriched with short reads. Although several methods have been proposed for ChIP-Seq analysis, most existing methods only consider reads that can be uniquely placed in the reference genome, and therefore have low power for detecting peaks located within repeat sequences. Here, we introduce a probabilistic approach for ChIP-Seq data analysis that utilizes all reads, providing a truly genome-wide view of binding patterns. Reads are modeled using a mixture model corresponding to K enriched regions and a null genomic background. We use maximum likelihood to estimate the locations of the enriched regions, and implement an expectation-maximization (E-M) algorithm, called AREM (aligning reads by expectation maximization), to update the alignment probabilities of each read to different genomic locations. We apply the algorithm to identify genome-wide binding events of two proteins: Rad21, a component of cohesin and a key factor involved in chromatid cohesion, and Srebp-1, a transcription factor important for lipid/cholesterol homeostasis. Using AREM, we were able to identify 19,935 Rad21 peaks and 1,748 Srebp-1 peaks in the mouse genome with high confidence, including 1,517 (7.6%) Rad21 peaks and 227 (13%) Srebp-1 peaks that were missed using only uniquely mapped reads. The open source implementation of our algorithm is available at http://sourceforge.net/projects/arem.


Subject(s)
Algorithms , Computer Simulation , Models, Genetic , Sequence Alignment/methods , Animals , Base Sequence , Cell Cycle Proteins , Chromatin Immunoprecipitation , DNA-Binding Proteins , High-Throughput Nucleotide Sequencing , Likelihood Functions , Mice , Nuclear Proteins/genetics , Phosphoproteins/genetics , Protein Binding , Sequence Analysis, DNA , Sterol Regulatory Element Binding Protein 1/genetics
9.
Cell Metab ; 13(4): 367-375, 2011 Apr 06.
Article in English | MEDLINE | ID: mdl-21459322

ABSTRACT

Sterol regulatory element-binding proteins (SREBPs) are key transcriptional regulators of lipid metabolism. To define functional differences between the three mammalian SREBPs we used genome-wide ChIP-seq with isoform-specific antibodies and chromatin from select tissues of mice challenged with different dietary conditions that enrich for specific SREBPs. We show that hepatic SREBP-2 binds preferentially to two different gene-proximal motifs. A Gene Ontology (GO) analysis suggests SREBP-2 targets lipid metabolic processes as expected, but apoptosis and autophagy gene categories were also enriched. We show that SREBP-2 directly activates autophagy genes during cell-sterol depletion, conditions known to induce both autophagy and nuclear SREBP-2 levels. Additionally, SREBP-2 knockdown during nutrient depletion decreased autophagosome formation and lipid droplet association of the autophagosome targeting protein LC3. Thus, the lipid droplet could be viewed as a third source of cellular cholesterol, which along with sterol synthesis and uptake, is also regulated by SREBP-2.


Subject(s)
Autophagy , Chromatin/metabolism , Liver/metabolism , Sterol Regulatory Element Binding Protein 2/analysis , Animals , Cell Line , Chromatin Immunoprecipitation , Gene Expression Regulation , Genome , Humans , Male , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Molecular Sequence Data , RNA Interference , RNA, Small Interfering/metabolism , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/metabolism
10.
Cancer Res ; 69(18): 7459-65, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19738052

ABSTRACT

MicroRNAs regulate diverse cellular processes and play an integral role in cancer pathogenesis. Genomic variation within miRNA target sites may therefore be important sources for genetic differences in cancer risk. To investigate this possibility, we mapped HapMap single nucleotide polymorphisms (SNP) to putative miRNA recognition sites within genes dysregulated in estrogen receptor-stratified breast tumors and used local linkage disequilibrium patterns to identify high-ranking SNPs in the Cancer Genetic Markers of Susceptibility (CGEMS) breast cancer genome-wide association study for further testing. Two SNPs, rs1970801 and rs11097457, scoring in the top 100 from the CGEMS study, were in strong linkage disequilibrium with rs1434536, an SNP that resides within a miR-125b target site in the 3' untranslated region of the bone morphogenic receptor type 1B (BMPR1B) gene encoding a transmembrane serine/threonine kinase. We validated the CGEMS association findings for rs1970801 in an independent cohort of admixture-corrected cases identified from families with multiple case histories. Subsequent association testing of rs1434536 for these cases and CGEMS controls with imputed genotypes supported the association. Furthermore, luciferase reporter assays and overexpression of miR-125b-mimics combined with quantitative reverse transcription-PCR showed that BMPR1B transcript is a direct target of miR-125b and that miR-125b differentially regulates the C and T alleles of rs1434536. These results suggest that allele-specific regulation of BMPR1B by miR-125b explains the observed disease risk. Our approach is general and can help identify and explain the mechanisms behind disease association for alleles that affect miRNA regulation.


Subject(s)
Bone Morphogenetic Protein Receptors, Type I/genetics , Breast Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , MicroRNAs/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Alleles , Binding Sites , Bone Morphogenetic Protein Receptors, Type I/biosynthesis , Bone Morphogenetic Protein Receptors, Type I/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/metabolism , Middle Aged , Polymorphism, Single Nucleotide , Receptors, Estrogen/genetics , Transfection , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...