Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Mol Biol Cell ; 35(7): ar96, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38717453

ABSTRACT

Cell type-specific enhancers are critically important for lineage specification. The mechanisms that determine cell-type specificity of enhancer activity, however, are not fully understood. Most current models for how enhancers function invoke physical proximity between enhancer elements and their target genes. Here, we use an imaging-based approach to examine the spatial relationship of cell type-specific enhancers and their target genes with single-cell resolution. Using high-throughput microscopy, we measure the spatial distance from target promoters to their cell type-specific active and inactive enhancers in individual pancreatic cells derived from distinct lineages. We find increased proximity of all promoter-enhancer pairs relative to non-enhancer pairs separated by similar genomic distances. Strikingly, spatial proximity between enhancers and target genes was unrelated to tissue-specific enhancer activity. Furthermore, promoter-enhancer proximity did not correlate with the expression status of target genes. Our results suggest that promoter-enhancer pairs exist in a distinctive chromatin environment but that genome folding is not a universal driver of cell-type specificity in enhancer function.


Subject(s)
Chromatin , Enhancer Elements, Genetic , Promoter Regions, Genetic , Transcription, Genetic , Enhancer Elements, Genetic/genetics , Promoter Regions, Genetic/genetics , Chromatin/metabolism , Animals , Mice , Cell Lineage , Pancreas/metabolism
2.
BMC Genomics ; 25(1): 76, 2024 Jan 18.
Article in English | MEDLINE | ID: mdl-38238687

ABSTRACT

BACKGROUND: The human pancreas is composed of specialized cell types producing hormones and enzymes critical to human health. These specialized functions are the result of cell type-specific transcriptional programs which manifest in cell-specific gene expression. Understanding these programs is essential to developing therapies for pancreatic disorders. Transcription in the human pancreas has been widely studied by single-cell RNA technologies, however the diversity of protocols and analysis methods hinders their interpretability in the aggregate. RESULTS: In this work, we perform a meta-analysis of pancreatic single-cell RNA sequencing data. We present a database for reference transcriptome abundances and cell-type specificity metrics. This database facilitates the identification and definition of marker genes within the pancreas. Additionally, we introduce a versatile tool which is freely available as an R package, and should permit integration into existing workflows. Our tool accepts count data files generated by widely-used single-cell gene expression platforms in their original format, eliminating an additional pre-formatting step. Although we designed it to calculate expression specificity of pancreas cell types, our tool is agnostic to the biological source of count data, extending its applicability to other biological systems. CONCLUSIONS: Our findings enhance the current understanding of expression specificity within the pancreas, surpassing previous work in terms of scope and detail. Furthermore, our database and tool enable researchers to perform similar calculations in diverse biological systems, expanding the applicability of marker gene identification and facilitating comparative analyses.


Subject(s)
Pancreatic Diseases , Software , Humans , Single-Cell Analysis/methods , Transcriptome , Pancreas , Gene Expression Profiling/methods , Sequence Analysis, RNA/methods
3.
Mol Cancer Ther ; 23(4): 464-477, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38151817

ABSTRACT

Histone deacetylase inhibitors (HDACi) are part of a growing class of epigenetic therapies used for the treatment of cancer. Although HDACis are effective in the treatment of T-cell lymphomas, treatment of solid tumors with this class of drugs has not been successful. Overexpression of the multidrug resistance protein P-glycoprotein (P-gp), encoded by ABCB1, is known to confer resistance to the HDACi romidepsin in vitro, yet increased ABCB1 expression has not been associated with resistance in patients, suggesting that other mechanisms of resistance arise in the clinic. To identify alternative mechanisms of resistance to romidepsin, we selected MCF-7 breast cancer cells with romidepsin in the presence of the P-gp inhibitor verapamil to reduce the likelihood of P-gp-mediated resistance. The resulting cell line, MCF-7 DpVp300, does not express P-gp and was found to be selectively resistant to romidepsin but not to other HDACis such as belinostat, panobinostat, or vorinostat. RNA-sequencing analysis revealed upregulation of the mRNA coding for the putative methyltransferase, METTL7A, whose paralog, METTL7B, was previously shown to methylate thiol groups on hydrogen sulfide and captopril. As romidepsin has a thiol as the zinc-binding moiety, we hypothesized that METTL7A could inactivate romidepsin and other thiol-based HDACis via methylation of the thiol group. We demonstrate that expression of METTL7A or METTL7B confers resistance to thiol-based HDACis and that both enzymes are capable of methylating thiol-containing HDACis. We thus propose that METTL7A and METTL7B confer resistance to thiol-based HDACis by methylating and inactivating the zinc-binding thiol.


Subject(s)
Histone Deacetylase Inhibitors , Neoplasms , Humans , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Methyltransferases/metabolism , Neoplasms/drug therapy , Panobinostat/pharmacology , Panobinostat/therapeutic use , Zinc
4.
Proc Natl Acad Sci U S A ; 119(26): e2201267119, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35733248

ABSTRACT

Delineating gene regulatory networks that orchestrate cell-type specification is a continuing challenge for developmental biologists. Single-cell analyses offer opportunities to address these challenges and accelerate discovery of rare cell lineage relationships and mechanisms underlying hierarchical lineage decisions. Here, we describe the molecular analysis of mouse pancreatic endocrine cell differentiation using single-cell transcriptomics, chromatin accessibility assays coupled to genetic labeling, and cytometry-based cell purification. We uncover transcription factor networks that delineate ß-, α-, and δ-cell lineages. Through genomic footprint analysis, we identify transcription factor-regulatory DNA interactions governing pancreatic cell development at unprecedented resolution. Our analysis suggests that the transcription factor Neurog3 may act as a pioneer transcription factor to specify the pancreatic endocrine lineage. These findings could improve protocols to generate replacement endocrine cells from renewable sources, like stem cells, for diabetes therapy.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Chromatin , Islets of Langerhans , Nerve Tissue Proteins , Transcriptome , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Chromatin/genetics , Chromatin/metabolism , Gene Expression Regulation, Developmental , Islets of Langerhans/growth & development , Islets of Langerhans/metabolism , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Single-Cell Analysis
6.
Cancer Res ; 81(15): 3958-3970, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34049974

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) tumors can originate either from acinar or ductal cells in the adult pancreas. We re-analyze multiple pancreas and PDAC single-cell RNA-seq datasets and find a subset of nonmalignant acinar cells, which we refer to as acinar edge (AE) cells, whose transcriptomes highly diverge from a typical acinar cell in each dataset. Genes upregulated among AE cells are enriched for transcriptomic signatures of pancreatic progenitors, acinar dedifferentiation, and several oncogenic programs. AE-upregulated genes are upregulated in human PDAC tumors, and consistently, their promoters are hypomethylated. High expression of these genes is associated with poor patient survival. The fraction of AE-like cells increases with age in healthy pancreatic tissue, which is not explained by clonal mutations, thus pointing to a nongenetic source of variation. The fraction of AE-like cells is also significantly higher in human pancreatitis samples. Finally, we find edge-like states in lung, liver, prostate, and colon tissues, suggesting that subpopulations of healthy cells across tissues can exist in pre-neoplastic states. SIGNIFICANCE: These findings show "edge" epithelial cell states with oncogenic transcriptional activity in human organs without oncogenic mutations. In the pancreas, the fraction of acinar cells increases with age.


Subject(s)
Acinar Cells/metabolism , Carcinoma, Pancreatic Ductal/physiopathology , Carcinoma, Pancreatic Ductal/mortality , Humans , Survival Analysis
7.
Cancer Discov ; 11(3): 638-659, 2021 03.
Article in English | MEDLINE | ID: mdl-33060108

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is characterized by extensive desmoplasia, which challenges the molecular analyses of bulk tumor samples. Here we FACS-purified epithelial cells from human PDAC and normal pancreas and derived their genome-wide transcriptome and DNA methylome landscapes. Clustering based on DNA methylation revealed two distinct PDAC groups displaying different methylation patterns at regions encoding repeat elements. Methylationlow tumors are characterized by higher expression of endogenous retroviral transcripts and double-stranded RNA sensors, which lead to a cell-intrinsic activation of an interferon signature (IFNsign). This results in a protumorigenic microenvironment and poor patient outcome. Methylationlow/IFNsignhigh and Methylationhigh/IFNsignlow PDAC cells preserve lineage traits, respective of normal ductal or acinar pancreatic cells. Moreover, ductal-derived Kras G12D/Trp53 -/- mouse PDACs show higher expression of IFNsign compared with acinar-derived counterparts. Collectively, our data point to two different origins and etiologies of human PDACs, with the aggressive Methylationlow/IFNsignhigh subtype potentially targetable by agents blocking intrinsic IFN signaling. SIGNIFICANCE: The mutational landscapes of PDAC alone cannot explain the observed interpatient heterogeneity. We identified two PDAC subtypes characterized by differential DNA methylation, preserving traits from normal ductal/acinar cells associated with IFN signaling. Our work suggests that epigenetic traits and the cell of origin contribute to PDAC heterogeneity.This article is highlighted in the In This Issue feature, p. 521.


Subject(s)
Carcinoma, Pancreatic Ductal/etiology , Carcinoma, Pancreatic Ductal/metabolism , DNA Methylation , Interferons/metabolism , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/metabolism , Repetitive Sequences, Nucleic Acid , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , CpG Islands , Disease Progression , Disease Susceptibility , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Models, Biological , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Prognosis , Reproducibility of Results , Signal Transduction , Transcriptome , Tumor Microenvironment/genetics
8.
Cell Syst ; 7(3): 310-322.e4, 2018 09 26.
Article in English | MEDLINE | ID: mdl-30145115

ABSTRACT

Understanding the genomic logic that underlies cellular diversity and developmental potential in the human pancreas will accelerate the growth of cell replacement therapies and reveal genetic risk mechanisms in diabetes. Here, we identified and characterized thousands of chromatin regions governing cell-specific gene regulation in human pancreatic endocrine and exocrine lineages, including islet ß cells, α cells, duct, and acinar cells. Our findings have captured cellular ontogenies at the chromatin level, identified lineage-specific regulators potentially acting on these sites, and uncovered hallmarks of regulatory plasticity between cell types that suggest mechanisms to regenerate ß cells from pancreatic endocrine or exocrine cells. Our work shows that disease risk variants related to pancreas are significantly enriched in these regulatory regions and reveals previously unrecognized links between endocrine and exocrine pancreas in diabetes risk.


Subject(s)
Chromatin/physiology , Diabetes Mellitus/genetics , Insulin-Secreting Cells/physiology , Pancreas, Exocrine/pathology , Pancreas/physiology , Cell Differentiation , Cell Lineage , Cell Plasticity , Cells, Cultured , Chromatin Assembly and Disassembly , Diabetes Mellitus/pathology , Gene Expression Regulation , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Organ Specificity , Pancreas/pathology , Polymorphism, Single Nucleotide , Regeneration , Risk
9.
Cell ; 171(2): 321-330.e14, 2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28965763

ABSTRACT

As organisms age, cells accumulate genetic and epigenetic errors that eventually lead to impaired organ function or catastrophic transformation such as cancer. Because aging reflects a stochastic process of increasing disorder, cells in an organ will be individually affected in different ways, thus rendering bulk analyses of postmitotic adult cells difficult to interpret. Here, we directly measure the effects of aging in human tissue by performing single-cell transcriptome analysis of 2,544 human pancreas cells from eight donors spanning six decades of life. We find that islet endocrine cells from older donors display increased levels of transcriptional noise and potential fate drift. By determining the mutational history of individual cells, we uncover a novel mutational signature in healthy aging endocrine cells. Our results demonstrate the feasibility of using single-cell RNA sequencing (RNA-seq) data from primary cells to derive insights into genetic and transcriptional processes that operate on aging human tissue.


Subject(s)
Aging/pathology , Cellular Senescence , Mutation , Pancreas/pathology , Single-Cell Analysis , Adult , Child , Child, Preschool , Humans , Infant , Middle Aged , Pancreas/cytology , Pancreas/physiology , Polymorphism, Single Nucleotide , Sequence Analysis, RNA , Transcription, Genetic
10.
Sci Rep ; 7(1): 5899, 2017 07 19.
Article in English | MEDLINE | ID: mdl-28724969

ABSTRACT

Three-dimensional tissue-structural relationships are not well captured by typical thin-section histology, posing challenges for the study of tissue physiology and pathology. Moreover, while recent progress has been made with intact methods for clearing, labeling, and imaging whole organs such as the mature brain, these approaches are generally unsuitable for soft, irregular, and heterogeneous tissues that account for the vast majority of clinical samples and biopsies. Here we develop a biphasic hydrogel methodology, which along with automated analysis, provides for high-throughput quantitative volumetric interrogation of spatially-irregular and friable tissue structures. We validate and apply this approach in the examination of a variety of developing and diseased tissues, with specific focus on the dynamics of normal and pathological pancreatic innervation and development, including in clinical samples. Quantitative advantages of the intact-tissue approach were demonstrated compared to conventional thin-section histology, pointing to broad applications in both research and clinical settings.


Subject(s)
Disease , Imaging, Three-Dimensional/methods , Organogenesis , Animals , Female , Humans , Hydrogels/chemistry , Mice, Inbred C57BL , Neural Crest/cytology , Neurosecretory Systems/cytology , Pancreas/cytology
11.
Cell Metab ; 23(5): 909-20, 2016 05 10.
Article in English | MEDLINE | ID: mdl-27133132

ABSTRACT

Intensive efforts are focused on identifying regulators of human pancreatic islet cell growth and maturation to accelerate development of therapies for diabetes. After birth, islet cell growth and function are dynamically regulated; however, establishing these age-dependent changes in humans has been challenging. Here, we describe a multimodal strategy for isolating pancreatic endocrine and exocrine cells from children and adults to identify age-dependent gene expression and chromatin changes on a genomic scale. These profiles revealed distinct proliferative and functional states of islet α cells or ß cells and histone modifications underlying age-dependent gene expression changes. Expression of SIX2 and SIX3, transcription factors without prior known functions in the pancreas and linked to fasting hyperglycemia risk, increased with age specifically in human islet ß cells. SIX2 and SIX3 were sufficient to enhance insulin content or secretion in immature ß cells. Our work provides a unique resource to study human-specific regulators of islet cell maturation and function.


Subject(s)
Aging/genetics , Gene Expression Regulation, Developmental , Insulin-Secreting Cells/metabolism , Adult , Cell Differentiation , Cell Separation , Child , Child, Preschool , Chromatin/metabolism , Chromatin Immunoprecipitation , Diabetes Mellitus/genetics , Histone Code , Homeodomain Proteins/metabolism , Humans , Infant , Middle Aged , Transcription Factors/metabolism , Transcriptome/genetics , Young Adult
12.
Cell Syst ; 1(2): 152-162, 2015 Aug 26.
Article in English | MEDLINE | ID: mdl-26430702

ABSTRACT

A wealth of physical interaction data between transcription factors (TFs) and DNA has been generated, but these interactions often do not have apparent regulatory consequences. Thus, equating physical interaction data with gene regulatory networks (GRNs) is problematic. Here, we comprehensively assay TF activity, rather than binding, to construct a network of gene regulatory interactions in the C. elegans intestine. By manually observing the in vivo tissue-specific knockdown of 921 TFs on a panel of 19 fluorescent transcriptional reporters, we identified a GRN of 411 interactions between 19 promoters and 177 TFs. This GRN shows only modest overlap with physical interactions, indicating that many regulatory interactions are indirect. We applied nested effects modeling to uncover information flow between TFs in the intestine that converges on a small set of physical TF-promoter interactions. We found numerous cell nonautonomous regulatory interactions, illustrating tissue-to-tissue communication. Altogether, our study illuminates the complexity of gene regulation in the context of a living animal.

13.
PLoS Genet ; 10(10): e1004645, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25330008

ABSTRACT

The regulatory logic underlying global transcriptional programs controlling development of visceral organs like the pancreas remains undiscovered. Here, we profiled gene expression in 12 purified populations of fetal and adult pancreatic epithelial cells representing crucial progenitor cell subsets, and their endocrine or exocrine progeny. Using probabilistic models to decode the general programs organizing gene expression, we identified co-expressed gene sets in cell subsets that revealed patterns and processes governing progenitor cell development, lineage specification, and endocrine cell maturation. Purification of Neurog3 mutant cells and module network analysis linked established regulators such as Neurog3 to unrecognized gene targets and roles in pancreas development. Iterative module network analysis nominated and prioritized transcriptional regulators, including diabetes risk genes. Functional validation of a subset of candidate regulators with corresponding mutant mice revealed that the transcription factors Etv1, Prdm16, Runx1t1 and Bcl11a are essential for pancreas development. Our integrated approach provides a unique framework for identifying regulatory genes and functional gene sets underlying pancreas development and associated diseases such as diabetes mellitus.


Subject(s)
Cell Separation/methods , Gene Expression Regulation, Developmental , Pancreas/cytology , Pancreas/embryology , Pancreas/growth & development , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Genomics/methods , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/physiology , Mice, Mutant Strains , Mice, Transgenic , Models, Statistical , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Reproducibility of Results , SOX9 Transcription Factor/genetics , Stem Cells/cytology , Stem Cells/physiology
14.
Cell ; 153(1): 240-52, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23540701

ABSTRACT

Dietary composition has major effects on physiology. Here, we show that developmental rate, reproduction, and lifespan are altered in C. elegans fed Comamonas DA1877 relative to those fed a standard E. coli OP50 diet. We identify a set of genes that change in expression in response to this diet and use the promoter of one of these (acdh-1) as a dietary sensor. Remarkably, the effects on transcription and development occur even when Comamonas DA1877 is diluted with another diet, suggesting that Comamonas DA1877 generates a signal that is sensed by the nematode. Surprisingly, the developmental effect is independent from TOR and insulin signaling. Rather, Comamonas DA1877 affects cyclic gene expression during molting, likely through the nuclear hormone receptor NHR-23. Altogether, our findings indicate that different bacteria elicit various responses via distinct mechanisms, which has implications for diseases such as obesity and the interactions between the human microbiome and intestinal cells.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/physiology , Insulin/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Acyl-CoA Dehydrogenase/metabolism , Animals , Betaproteobacteria , Caenorhabditis elegans/metabolism , Diet , Escherichia coli , Gene Expression , Longevity , Molting , Receptors, Cytoplasmic and Nuclear/metabolism , Starvation , Transcriptome
15.
Cell ; 153(1): 253-66, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23540702

ABSTRACT

Expression profiles are tailored according to dietary input. However, the networks that control dietary responses remain largely uncharacterized. Here, we combine forward and reverse genetic screens to delineate a network of 184 genes that affect the C. elegans dietary response to Comamonas DA1877 bacteria. We find that perturbation of a mitochondrial network composed of enzymes involved in amino acid metabolism and the TCA cycle affects the dietary response. In humans, mutations in the corresponding genes cause inborn diseases of amino acid metabolism, most of which are treated by dietary intervention. We identify several transcription factors (TFs) that mediate the changes in gene expression upon metabolic network perturbations. Altogether, our findings unveil a transcriptional response system that is poised to sense dietary cues and metabolic imbalances, illustrating extensive communication between metabolic networks in the mitochondria and gene regulatory networks in the nucleus.


Subject(s)
Caenorhabditis elegans/metabolism , Gene Regulatory Networks , Metabolic Networks and Pathways , Acyl-CoA Dehydrogenase/metabolism , Animals , Betaproteobacteria , Caenorhabditis elegans Proteins/metabolism , Diet , Escherichia coli , Humans , Insulin/metabolism , Metabolism, Inborn Errors , Mitochondria/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , RNA Interference , Signal Transduction , Transcription Factors/metabolism
16.
Dev Cell ; 25(1): 5-13, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23597482

ABSTRACT

Elucidation of cellular and gene regulatory networks (GRNs) governing organ development will accelerate progress toward tissue replacement. Here, we have compiled reference GRNs underlying pancreas development from data mining that integrates multiple approaches, including mutant analysis, lineage tracing, cell purification, gene expression and enhancer analysis, and biochemical studies of gene regulation. Using established computational tools, we integrated and represented these networks in frameworks that should enhance understanding of the surging output of genomic-scale genetic and epigenetic studies of pancreas development and diseases such as diabetes and pancreatic cancer. We envision similar approaches would be useful for understanding the development of other organs.


Subject(s)
Gene Regulatory Networks , Pancreas/cytology , Pancreas/growth & development , Acinar Cells/cytology , Acinar Cells/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Dedifferentiation , Cell Differentiation , Computational Biology/methods , Data Mining , Diabetes Mellitus/genetics , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nucleotide Motifs , Stem Cells/cytology , Stem Cells/metabolism
17.
Mol Syst Biol ; 6: 367, 2010 May 11.
Article in English | MEDLINE | ID: mdl-20461074

ABSTRACT

Gene regulatory networks (GRNs) provide insights into the mechanisms of differential gene expression at a systems level. GRNs that relate to metazoan development have been studied extensively. However, little is still known about the design principles, organization and functionality of GRNs that control physiological processes such as metabolism, homeostasis and responses to environmental cues. In this study, we report the first experimentally mapped metazoan GRN of Caenorhabditis elegans metabolic genes. This network is enriched for nuclear hormone receptors (NHRs). The NHR family has greatly expanded in nematodes: humans have 48 NHRs, but C. elegans has 284, most of which are uncharacterized. We find that the C. elegans metabolic GRN is highly modular and that two GRN modules predominantly consist of NHRs. Network modularity has been proposed to facilitate a rapid response to different cues. As NHRs are metabolic sensors that are poised to respond to ligands, this suggests that C. elegans GRNs evolved to enable rapid and adaptive responses to different cues by a concurrence of NHR family expansion and modular GRN wiring.


Subject(s)
Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Gene Regulatory Networks/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Gene Expression Regulation , Models, Genetic , Promoter Regions, Genetic , RNA Interference , Two-Hybrid System Techniques , Yeasts
18.
Brief Funct Genomics ; 9(1): 4-12, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20008400

ABSTRACT

Differential gene expression plays a critical role in the development and physiology of multicellular organisms. At a 'systems level' (e.g. at the level of a tissue, organ or whole organism), this process can be studied using gene regulatory network (GRN) models that capture physical and regulatory interactions between genes and their regulators. In the past years, significant progress has been made toward the mapping of GRNs using a variety of experimental and computational approaches. Here, we will discuss gene-centered approaches that we employed to characterize GRNs and describe insights that we have obtained into the global design principles of gene regulation in complex metazoan systems.


Subject(s)
Gene Regulatory Networks/genetics , Genes/physiology , Animals , Binding Sites , Caenorhabditis elegans/genetics , Gene Expression Regulation/genetics , Gene Regulatory Networks/physiology , Models, Biological , Transcription Factors/metabolism , Two-Hybrid System Techniques , Yeasts
19.
Nucleic Acids Res ; 37(11): 3689-98, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19372275

ABSTRACT

Snail-type transcription factors (TFs) are found in numerous metazoan organisms and function in a plethora of cellular and developmental processes including mesoderm and neuronal development, apoptosis and cancer. So far, Snail-type TFs are exclusively known as transcriptional repressors. They repress gene expression by recruiting transcriptional co-repressors and/or by preventing DNA binding of activators from the basic helix-loop-helix (bHLH) family of TFs to CAGGTG E-box sequences. Here we report that the Caenorhabditis elegans Snail-type TF CES-1 can activate transcription in vivo. Moreover, we provide results that suggest that CES-1 can share its binding site with bHLH TFs, in different tissues, rather than only occluding bHLH DNA binding. Together, our data indicate that there are at least two types of CES-1 target genes and, therefore, that the molecular function of Snail-type TFs is more plastic than previously appreciated.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , DNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Binding Sites , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Pharynx/metabolism , Promoter Regions, Genetic , Regulatory Elements, Transcriptional
20.
Nat Methods ; 4(8): 659-64, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17589517

ABSTRACT

Yeast one-hybrid (Y1H) assays provide a gene-centered method for the identification of interactions between gene promoters and regulatory transcription factors (TFs). To date, Y1H assays have involved library screens that are relatively expensive and laborious. We present two Y1H strategies that allow immediate prey identification: matrix assays that use an array of 755 individual Caenorhabditis elegans TFs, and smart-pool assays that use TF multiplexing. Both strategies simplify the Y1H pipeline and reduce the cost of protein-DNA interaction identification. We used a Steiner triple system (STS) to create smart pools of 4-25 TFs. Notably, we uniplexed a small number of highly connected TFs to allow efficient assay deconvolution. Both strategies outperform library screens in terms of coverage, confidence and throughput. These versatile strategies can be adapted both to TFs in other systems and, likely, to other biomolecules and assays as well.


Subject(s)
Transcription, Genetic , Animals , Caenorhabditis elegans/genetics , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...