Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 15 de 15
1.
Genetics ; 222(4)2022 11 30.
Article En | MEDLINE | ID: mdl-36218393

Transcription factors play important roles in the development of the intestinal epithelium and its ability to respond to endocrine, nutritional, and microbial signals. Hepatocyte nuclear factor 4 family nuclear receptors are liganded transcription factors that are critical for the development and function of multiple digestive organs in vertebrates, including the intestinal epithelium. Zebrafish have 3 hepatocyte nuclear factor 4 homologs, of which, hnf4a was previously shown to mediate intestinal responses to microbiota in zebrafish larvae. To discern the functions of other hepatocyte nuclear factor 4 family members in zebrafish development and intestinal function, we created and characterized mutations in hnf4g and hnf4b. We addressed the possibility of genetic redundancy amongst these factors by creating double and triple mutants which showed different rates of survival, including apparent early lethality in hnf4a; hnf4b double mutants and triple mutants. RNA sequencing performed on digestive tracts from single and double mutant larvae revealed extensive changes in intestinal gene expression in hnf4a mutants that were amplified in hnf4a; hnf4g mutants, but limited in hnf4g mutants. Changes in hnf4a and hnf4a; hnf4g mutants were reminiscent of those seen in mice including decreased expression of genes involved in intestinal function and increased expression of cell proliferation genes, and were validated using transgenic reporters and EdU labeling in the intestinal epithelium. Gnotobiotics combined with RNA sequencing also showed hnf4g has subtler roles than hnf4a in host responses to microbiota. Overall, phenotypic changes in hnf4a single mutants were strongly enhanced in hnf4a; hnf4g double mutants, suggesting a conserved partial genetic redundancy between hnf4a and hnf4g in the vertebrate intestine.


Hepatocyte Nuclear Factor 4 , Intestinal Mucosa , Zebrafish Proteins , Zebrafish , Animals , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/physiology , Intestinal Mucosa/embryology , Intestinal Mucosa/metabolism , Intestines/embryology , Intestines/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/physiology
2.
Cell Mol Gastroenterol Hepatol ; 14(2): 465-493, 2022.
Article En | MEDLINE | ID: mdl-35533983

BACKGROUND & AIMS: The intestine constantly interprets and adapts to complex combinations of dietary and microbial stimuli. However, the transcriptional strategies by which the intestinal epithelium integrates these coincident sources of information remain unresolved. We recently found that microbiota colonization suppresses epithelial activity of hepatocyte nuclear factor 4 nuclear receptor transcription factors, but their integrative regulation was unknown. METHODS: We compared adult mice reared germ-free or conventionalized with a microbiota either fed normally or after a single high-fat meal. Preparations of unsorted jejunal intestinal epithelial cells were queried using lipidomics and genome-wide assays for RNA sequencing and ChIP sequencing for the activating histone mark H3K27ac and hepatocyte nuclear factor 4 alpha. RESULTS: Analysis of lipid classes, genes, and regulatory regions identified distinct nutritional and microbial responses but also simultaneous influence of both stimuli. H3K27ac sites preferentially increased by high-fat meal in the presence of microbes neighbor lipid anabolism and proliferation genes, were previously identified intestinal stem cell regulatory regions, and were not hepatocyte nuclear factor 4 alpha targets. In contrast, H3K27ac sites preferentially increased by high-fat meal in the absence of microbes neighbor targets of the energy homeostasis regulator peroxisome proliferator activated receptor alpha, neighbored fatty acid oxidation genes, were previously identified enterocyte regulatory regions, and were hepatocyte factor 4 alpha bound. CONCLUSIONS: Hepatocyte factor 4 alpha supports a differentiated enterocyte and fatty acid oxidation program in germ-free mice, and that suppression of hepatocyte factor 4 alpha by the combination of microbes and high-fat meal may result in preferential activation of intestinal epithelial cell proliferation programs. This identifies potential transcriptional mechanisms for intestinal adaptation to multiple signals and how microbiota may modulate intestinal lipid absorption, epithelial cell renewal, and systemic energy balance.


Duodenum , Gastrointestinal Microbiome , Intestinal Mucosa , Animals , Duodenum/metabolism , Duodenum/microbiology , Fatty Acids/metabolism , Hepatocyte Nuclear Factor 4/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Lipids , Mice
3.
BMC Genomics ; 23(1): 225, 2022 Mar 22.
Article En | MEDLINE | ID: mdl-35317738

BACKGROUND: The ability of animals and their microbiomes to adapt to starvation and then restore homeostasis after refeeding is fundamental to their continued survival and symbiosis. The intestine is the primary site of nutrient absorption and microbiome interaction, however our understanding of intestinal adaptations to starvation and refeeding remains limited. Here we used RNA sequencing and 16S rRNA gene sequencing to uncover changes in the intestinal transcriptome and microbiome of zebrafish subjected to long-term starvation and refeeding compared to continuously fed controls. RESULTS: Starvation over 21 days led to increased diversity and altered composition in the intestinal microbiome compared to fed controls, including relative increases in Vibrio and reductions in Plesiomonas bacteria. Starvation also led to significant alterations in host gene expression in the intestine, with distinct pathways affected at early and late stages of starvation. This included increases in the expression of ribosome biogenesis genes early in starvation, followed by decreased expression of genes involved in antiviral immunity and lipid transport at later stages. These effects of starvation on the host transcriptome and microbiome were almost completely restored within 3 days after refeeding. Comparison with published datasets identified host genes responsive to starvation as well as high-fat feeding or microbiome colonization, and predicted host transcription factors that may be involved in starvation response. CONCLUSIONS: Long-term starvation induces progressive changes in microbiome composition and host gene expression in the zebrafish intestine, and these changes are rapidly reversed after refeeding. Our identification of bacterial taxa, host genes and host pathways involved in this response provides a framework for future investigation of the physiological and ecological mechanisms underlying intestinal adaptations to food restriction.


Microbiota , Transcriptome , Animals , Intestines/microbiology , RNA, Ribosomal, 16S , Zebrafish/genetics
4.
Sci Adv ; 7(30)2021 07.
Article En | MEDLINE | ID: mdl-34301599

Bile salt synthesis, secretion into the intestinal lumen, and resorption in the ileum occur in all vertebrate classes. In mammals, bile salt composition is determined by host and microbial enzymes, affecting signaling through the bile salt-binding transcription factor farnesoid X receptor (Fxr). However, these processes in other vertebrate classes remain poorly understood. We show that key components of hepatic bile salt synthesis and ileal transport pathways are conserved and under control of Fxr in zebrafish. Zebrafish bile salts consist primarily of a C27 bile alcohol and a C24 bile acid that undergo multiple microbial modifications including bile acid deconjugation that augments Fxr activity. Using single-cell RNA sequencing, we provide a cellular atlas of the zebrafish intestinal epithelium and uncover roles for Fxr in transcriptional and differentiation programs in ileal and other cell types. These results establish zebrafish as a nonmammalian vertebrate model for studying bile salt metabolism and Fxr signaling.


Bile Acids and Salts , Zebrafish , Animals , Bile Acids and Salts/metabolism , Intestines , Liver/metabolism , Mammals/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Zebrafish/metabolism
5.
Cell Host Microbe ; 29(2): 179-196.e9, 2021 02 10.
Article En | MEDLINE | ID: mdl-33352109

The intestinal epithelium senses nutritional and microbial stimuli using epithelial sensory enteroendocrine cells (EEC). EECs communicate nutritional information to the nervous system, but whether they also relay signals from intestinal microbes remains unknown. Using in vivo real-time measurements of EEC and nervous system activity in zebrafish, we discovered that the bacteria Edwardsiella tarda activate EECs through the receptor transient receptor potential ankyrin A1 (Trpa1) and increase intestinal motility. Microbial, pharmacological, or optogenetic activation of Trpa1+EECs directly stimulates vagal sensory ganglia and activates cholinergic enteric neurons by secreting the neurotransmitter 5-hydroxytryptamine (5-HT). A subset of indole derivatives of tryptophan catabolism produced by E. tarda and other gut microbes activates zebrafish EEC Trpa1 signaling. These catabolites also directly stimulate human and mouse Trpa1 and intestinal 5-HT secretion. These results establish a molecular pathway by which EECs regulate enteric and vagal neuronal pathways in response to microbial signals.


Edwardsiella tarda/metabolism , Enteric Nervous System/metabolism , Enteroendocrine Cells/physiology , Intestinal Mucosa/metabolism , TRPA1 Cation Channel/metabolism , Animals , Animals, Genetically Modified , Cholinergic Neurons/metabolism , Enteric Nervous System/cytology , Gastrointestinal Motility/physiology , Intestinal Mucosa/cytology , Intestinal Mucosa/innervation , Proto-Oncogene Proteins c-ret/genetics , Serotonin/metabolism , Signal Transduction , Tryptophan/metabolism , Zebrafish , Zebrafish Proteins/genetics
6.
Nat Rev Gastroenterol Hepatol ; 18(1): 7-23, 2021 01.
Article En | MEDLINE | ID: mdl-33024279

The intestinal epithelium serves the unique and critical function of harvesting dietary nutrients, while simultaneously acting as a cellular barrier separating tissues from the luminal environment and gut microbial ecosystem. Two salient features of the intestinal epithelium enable it to perform these complex functions. First, cells within the intestinal epithelium achieve a wide range of specialized identities, including different cell types and distinct anterior-posterior patterning along the intestine. Second, intestinal epithelial cells are sensitive and responsive to the dynamic milieu of dietary nutrients, xenobiotics and microorganisms encountered in the intestinal luminal environment. These diverse identities and responsiveness of intestinal epithelial cells are achieved in part through the differential transcription of genes encoded in their shared genome. Here, we review insights from mice and other vertebrate models into the transcriptional regulatory mechanisms underlying intestinal epithelial identity and microbial responsiveness, including DNA methylation, chromatin accessibility, histone modifications and transcription factors. These studies are revealing that most transcription factors involved in intestinal epithelial identity also respond to changes in the microbiota, raising both opportunities and challenges to discern the underlying integrative transcriptional regulatory networks.


Cell Differentiation/genetics , Cellular Microenvironment/physiology , Gastrointestinal Microbiome , Intestinal Mucosa/physiology , Animals , Cell Differentiation/physiology , Cellular Microenvironment/genetics , Gastrointestinal Microbiome/genetics , Gastrointestinal Microbiome/physiology , Gene Expression Regulation , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/microbiology , Intestines/microbiology , Intestines/physiology , Mice , Models, Animal , Nutrigenomics , Nutritional Physiological Phenomena/genetics , Nutritional Physiological Phenomena/physiology , Transcription, Genetic/genetics , Transcription, Genetic/physiology , Zebrafish
7.
Dev Cell ; 51(1): 7-20.e6, 2019 10 07.
Article En | MEDLINE | ID: mdl-31474562

The guts of neonatal mammals and stomachless fish have a limited capacity for luminal protein digestion, which allows oral acquisition of antibodies and antigens. However, how dietary protein is absorbed during critical developmental stages when the gut is still immature is unknown. Here, we show that specialized intestinal cells, which we call lysosome-rich enterocytes (LREs), internalize dietary protein via receptor-mediated and fluid-phase endocytosis for intracellular digestion and trans-cellular transport. In LREs, we identify a conserved endocytic machinery, composed of the scavenger receptor complex Cubilin/Amnionless and Dab2, that is required for protein uptake by LREs and for growth and survival of larval zebrafish. Moreover, impairing LRE function in suckling mice, via conditional deletion of Dab2, leads to stunted growth and severe protein malnutrition reminiscent of kwashiorkor, a devastating human malnutrition syndrome. These findings identify digestive functions and conserved molecular mechanisms in LREs that are crucial for vertebrate growth and survival.


Dietary Proteins/metabolism , Enterocytes/metabolism , Intestinal Absorption , Intestines/embryology , Lysosomes/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Disease Models, Animal , Female , Gastrointestinal Microbiome , Gene Deletion , Gene Expression Regulation, Developmental , Ileum/embryology , Ileum/metabolism , Kwashiorkor/metabolism , Ligands , Male , Membrane Proteins/metabolism , Mice , Receptors, Cell Surface/metabolism , Zebrafish , Zebrafish Proteins/metabolism
8.
Chem Senses ; 44(8): 615-630, 2019 10 17.
Article En | MEDLINE | ID: mdl-31403159

Sensory systems such as the olfactory system detect chemical stimuli and thereby determine the relationships between the animal and its surroundings. Olfaction is one of the most conserved and ancient sensory systems in vertebrates. The vertebrate olfactory epithelium is colonized by complex microbial communities, but microbial contribution to host olfactory gene expression remains unknown. In this study, we show that colonization of germ-free zebrafish and mice with microbiota leads to widespread transcriptional responses in olfactory organs as measured in bulk tissue transcriptomics and RT-qPCR. Germ-free zebrafish olfactory epithelium showed defects in pseudostratification; however, the size of the olfactory pit and the length of the cilia were not different from that of colonized zebrafish. One of the mechanisms by which microbiota control host transcriptional programs is by differential expression and activity of specific transcription factors (TFs). REST (RE1 silencing transcription factor, also called NRSF) is a zinc finger TF that binds to the conserved motif repressor element 1 found in the promoter regions of many neuronal genes with functions in neuronal development and differentiation. Colonized zebrafish and mice showed increased nasal expression of REST, and genes with reduced expression in colonized animals were strongly enriched in REST-binding motifs. Nasal commensal bacteria promoted in vitro differentiation of Odora cells by regulating the kinetics of REST expression. REST knockdown resulted in decreased Odora cell differentiation in vitro. Our results identify a conserved mechanism by which microbiota regulate vertebrate olfactory transcriptional programs and reveal a new role for REST in sensory organs.


Microbiota/physiology , Nerve Tissue Proteins/genetics , Olfactory Mucosa/metabolism , Olfactory Receptor Neurons/metabolism , Repressor Proteins/genetics , Smell/genetics , Animals , Cell Line , Conserved Sequence , Gene Expression Profiling , Gene Expression Regulation , Germ-Free Life , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Olfactory Mucosa/cytology , Olfactory Mucosa/microbiology , Olfactory Receptor Neurons/cytology , Olfactory Receptor Neurons/microbiology , Promoter Regions, Genetic , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rats , Repressor Proteins/metabolism , Symbiosis/physiology , Zebrafish
9.
PLoS Biol ; 15(8): e2002054, 2017 Aug.
Article En | MEDLINE | ID: mdl-28850571

The intestinal epithelium serves critical physiologic functions that are shared among all vertebrates. However, it is unknown how the transcriptional regulatory mechanisms underlying these functions have changed over the course of vertebrate evolution. We generated genome-wide mRNA and accessible chromatin data from adult intestinal epithelial cells (IECs) in zebrafish, stickleback, mouse, and human species to determine if conserved IEC functions are achieved through common transcriptional regulation. We found evidence for substantial common regulation and conservation of gene expression regionally along the length of the intestine from fish to mammals and identified a core set of genes comprising a vertebrate IEC signature. We also identified transcriptional start sites and other putative regulatory regions that are differentially accessible in IECs in all 4 species. Although these sites rarely showed sequence conservation from fish to mammals, surprisingly, they drove highly conserved IEC expression in a zebrafish reporter assay. Common putative transcription factor binding sites (TFBS) found at these sites in multiple species indicate that sequence conservation alone is insufficient to identify much of the functionally conserved IEC regulatory information. Among the rare, highly sequence-conserved, IEC-specific regulatory regions, we discovered an ancient enhancer upstream from her6/HES1 that is active in a distinct population of Notch-positive cells in the intestinal epithelium. Together, these results show how combining accessible chromatin and mRNA datasets with TFBS prediction and in vivo reporter assays can reveal tissue-specific regulatory information conserved across 420 million years of vertebrate evolution. We define an IEC transcriptional regulatory network that is shared between fish and mammals and establish an experimental platform for studying how evolutionarily distilled regulatory information commonly controls IEC development and physiology.


Fish Proteins/metabolism , Gene Expression Regulation , Intestinal Mucosa/metabolism , RNA, Messenger/metabolism , Smegmamorpha/metabolism , Zebrafish/metabolism , Animals , California , Colon/cytology , Colon/growth & development , Colon/metabolism , Duodenum/cytology , Duodenum/growth & development , Duodenum/metabolism , Female , Fish Proteins/genetics , Gene Expression Profiling/veterinary , Genomics/methods , Humans , Ileum/cytology , Ileum/growth & development , Ileum/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/growth & development , Jejunum/cytology , Jejunum/growth & development , Jejunum/metabolism , Larva/growth & development , Larva/metabolism , Male , Mice , Organ Specificity , Rivers , Smegmamorpha/growth & development , Species Specificity , Zebrafish/growth & development
10.
Genome Res ; 27(7): 1195-1206, 2017 07.
Article En | MEDLINE | ID: mdl-28385711

Microbiota influence diverse aspects of intestinal physiology and disease in part by controlling tissue-specific transcription of host genes. However, host genomic mechanisms mediating microbial control of intestinal gene expression are poorly understood. Hepatocyte nuclear factor 4 (HNF4) is the most ancient family of nuclear receptor transcription factors with important roles in human metabolic and inflammatory bowel diseases, but a role in host response to microbes is unknown. Using an unbiased screening strategy, we found that zebrafish Hnf4a specifically binds and activates a microbiota-suppressed intestinal epithelial transcriptional enhancer. Genetic analysis revealed that zebrafish hnf4a activates nearly half of the genes that are suppressed by microbiota, suggesting microbiota negatively regulate Hnf4a. In support, analysis of genomic architecture in mouse intestinal epithelial cells disclosed that microbiota colonization leads to activation or inactivation of hundreds of enhancers along with drastic genome-wide reduction of HNF4A and HNF4G occupancy. Interspecies meta-analysis suggested interactions between HNF4A and microbiota promote gene expression patterns associated with human inflammatory bowel diseases. These results indicate a critical and conserved role for HNF4A in maintaining intestinal homeostasis in response to microbiota.


Gastrointestinal Microbiome , Gene Expression Regulation , Hepatocyte Nuclear Factor 4/biosynthesis , Inflammatory Bowel Diseases , Zebrafish Proteins/biosynthesis , Zebrafish , Animals , Humans , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/microbiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mice , Species Specificity , Zebrafish/metabolism , Zebrafish/microbiology
11.
Genome Res ; 24(9): 1504-16, 2014 Sep.
Article En | MEDLINE | ID: mdl-24963153

Microbiota regulate intestinal physiology by modifying host gene expression along the length of the intestine, but the underlying regulatory mechanisms remain unresolved. Transcriptional specificity occurs through interactions between transcription factors (TFs) and cis-regulatory regions (CRRs) characterized by nucleosome-depleted accessible chromatin. We profiled transcriptome and accessible chromatin landscapes in intestinal epithelial cells (IECs) from mice reared in the presence or absence of microbiota. We show that regional differences in gene transcription along the intestinal tract were accompanied by major alterations in chromatin accessibility. Surprisingly, we discovered that microbiota modify host gene transcription in IECs without significantly impacting the accessible chromatin landscape. Instead, microbiota regulation of host gene transcription might be achieved by differential expression of specific TFs and enrichment of their binding sites in nucleosome-depleted CRRs near target genes. Our results suggest that the chromatin landscape in IECs is preprogrammed by the host in a region-specific manner to permit responses to microbiota through binding of open CRRs by specific TFs.


Chromatin Assembly and Disassembly , Intestinal Mucosa/metabolism , Microbiota , Transcription, Genetic , Animals , Intestinal Mucosa/microbiology , Mice , Mice, Inbred C57BL , Organ Specificity , Promoter Regions, Genetic , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome
12.
Nat Protoc ; 8(7): 1337-53, 2013.
Article En | MEDLINE | ID: mdl-23764940

Competition chromatin immunoprecipitation (competition ChIP) enables experimenters to measure protein-DNA dynamics at a single locus or across the entire genome, depending on the detection method. Competition ChIP relies on a cell containing two copies of a single DNA-associated factor, with each copy of the factor differentially epitope tagged. One of the copies is expressed constitutively and the second is induced as a competitor. The ratio of isoforms associated with discrete genomic locations is detected by ChIP-on-chip (ChIP-chip) or ChIP-sequencing (ChIP-seq). The rate at which the resident isoform of the protein is replaced by the competitor at each binding location enables the calculation of residence time for that factor at each site of interaction genome wide. Here we provide a detailed protocol for designing and performing competition ChIP experiments in Saccharomyces cerevisiae, which takes ∼5 d to complete (not including strain production and characterizations, which may take as long as 6 months). Included in this protocol are guidelines for downstream bioinformatic analysis to extract residence times throughout the genome.


Chromatin Immunoprecipitation/methods , DNA/metabolism , Proteins/analysis , Proteins/metabolism , Binding Sites , DNA/analysis , Epitopes , Genome , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Single-Cell Analysis/methods
13.
Science ; 340(6129): 190-5, 2013 Apr 12.
Article En | MEDLINE | ID: mdl-23470731

RNA chaperones are ubiquitous, heterogeneous proteins essential for RNA structural biogenesis and function. We investigated the mechanism of chaperone-mediated RNA folding by following the time-resolved dimerization of the packaging domain of a retroviral RNA at nucleotide resolution. In the absence of the nucleocapsid (NC) chaperone, dimerization proceeded through multiple, slow-folding intermediates. In the presence of NC, dimerization occurred rapidly through a single structural intermediate. The RNA binding domain of heterogeneous nuclear ribonucleoprotein A1 protein, a structurally unrelated chaperone, also accelerated dimerization. Both chaperones interacted primarily with guanosine residues. Replacing guanosine with more weakly pairing inosine yielded an RNA that folded rapidly without a facilitating chaperone. These results show that RNA chaperones can simplify RNA folding landscapes by weakening intramolecular interactions involving guanosine and explain many RNA chaperone activities.


Guanosine/metabolism , Molecular Chaperones/metabolism , Moloney murine leukemia virus/metabolism , Nucleocapsid Proteins/metabolism , RNA, Viral/chemistry , Base Sequence , Dimerization , Guanosine/chemistry , Heterogeneous Nuclear Ribonucleoprotein A1 , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/chemistry , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Inosine/chemistry , Inosine/metabolism , Kinetics , Models, Molecular , Molecular Chaperones/chemistry , Moloney murine leukemia virus/genetics , Nucleic Acid Conformation , Nucleocapsid Proteins/chemistry , Protein Binding , RNA, Viral/metabolism
14.
Nature ; 484(7393): 251-5, 2012 Apr 11.
Article En | MEDLINE | ID: mdl-22498630

Dynamic access to genetic information is central to organismal development and environmental response. Consequently, genomic processes must be regulated by mechanisms that alter genome function relatively rapidly. Conventional chromatin immunoprecipitation (ChIP) experiments measure transcription factor occupancy, but give no indication of kinetics and are poor predictors of transcription factor function at a given locus. To measure transcription-factor-binding dynamics across the genome, we performed competition ChIP (refs 6, 7) with a sequence-specific Saccharomyces cerevisiae transcription factor, Rap1 (ref. 8). Rap1-binding dynamics and Rap1 occupancy were only weakly correlated (R(2) = 0.14), but binding dynamics were more strongly linked to function than occupancy. Long Rap1 residence was coupled to transcriptional activation, whereas fast binding turnover, which we refer to as 'treadmilling', was linked to low transcriptional output. Thus, DNA-binding events that seem identical by conventional ChIP may have different underlying modes of interaction that lead to opposing functional outcomes. We propose that transcription factor binding turnover is a major point of regulation in determining the functional consequences of transcription factor binding, and is mediated mainly by control of competition between transcription factors and nucleosomes. Our model predicts a clutch-like mechanism that rapidly engages a treadmilling transcription factor into a stable binding state, or vice versa, to modulate transcription factor function.


DNA, Fungal/metabolism , Genome, Fungal , Models, Biological , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Telomere-Binding Proteins/metabolism , Transcription Factors/metabolism , Base Sequence , Binding Sites , Binding, Competitive , Chromatin Immunoprecipitation , DNA, Fungal/genetics , Gene Expression Regulation, Fungal , Histone Acetyltransferases/metabolism , Nucleosomes/genetics , Nucleosomes/metabolism , Protein Binding , RNA Polymerase II/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Saccharomyces cerevisiae/classification , Shelterin Complex , Time Factors
15.
PLoS One ; 4(3): e4886, 2009.
Article En | MEDLINE | ID: mdl-19295910

In cells lacking the histone methyltransferase Set2, initiation of RNA polymerase II transcription occurs inappropriately within the protein-coding regions of genes, rather than being restricted to the proximal promoter. It was previously reported that this "cryptic" transcription occurs preferentially in long genes, and in genes that are infrequently transcribed. Here, we mapped the transcripts produced in an S. cerevisiae strain lacking Set2, and applied rigorous statistical methods to identify sites of cryptic transcription at high resolution. We find that suppression of cryptic transcription occurs independent of gene length or transcriptional frequency. Our conclusions differ with those reported previously because we obtained a higher-resolution dataset, we accounted for the fact that gene length and transcriptional frequency are not independent variables, and we accounted for several ascertainment biases that make cryptic transcription easier to detect in long, infrequently transcribed genes. These new results and conclusions have implications for many commonly used genomic analysis approaches, and for the evolution of high-fidelity RNA polymerase II transcriptional initiation in eukaryotes.


Histone Deacetylases/metabolism , Methyltransferases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Transcription, Genetic , Algorithms , Histone Deacetylases/genetics , Methyltransferases/genetics , Microarray Analysis , Promoter Regions, Genetic , RNA/genetics , RNA/metabolism , RNA Polymerase II/metabolism , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics
...