Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
Add more filters

Publication year range
1.
Mol Cell ; 84(5): 822-838.e8, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38157845

ABSTRACT

Chromatin loops between gene pairs have been observed in diverse contexts in both flies and vertebrates. Combining high-resolution Capture-C, DNA fluorescence in situ hybridization, and genetic perturbations, we dissect the functional role of three loops between genes with related function during Drosophila embryogenesis. By mutating the loop anchor (but not the gene) or the gene (but not loop anchor), we disentangle loop formation and gene expression and show that the 3D proximity of paralogous gene loci supports their co-regulation. Breaking the loop leads to either an attenuation or enhancement of expression and perturbs their relative levels of expression and cross-regulation. Although many loops appear constitutive across embryogenesis, their function can change in different developmental contexts. Taken together, our results indicate that chromatin gene-gene loops act as architectural scaffolds that can be used in different ways in different contexts to fine-tune the coordinated expression of genes with related functions and sustain their cross-regulation.


Subject(s)
Chromatin , Chromosomes , Animals , In Situ Hybridization, Fluorescence , Chromatin/genetics , Drosophila/genetics
2.
Nat Rev Mol Cell Biol ; 20(6): 327-337, 2019 06.
Article in English | MEDLINE | ID: mdl-30886333

ABSTRACT

The spatial organization of the genome into compartments and topologically associated domains can have an important role in the regulation of gene expression. But could gene expression conversely regulate genome organization? Here, we review recent studies that assessed the requirement of transcription and/or the transcription machinery for the establishment or maintenance of genome topology. The results reveal different requirements at different genomic scales. Transcription is generally not required for higher-level genome compartmentalization, has only moderate effects on domain organization and is not sufficient to create new domain boundaries. However, on a finer scale, transcripts or transcription does seem to have a role in the formation of subcompartments and subdomains and in stabilizing enhancer-promoter interactions. Recent evidence suggests a dynamic, reciprocal interplay between fine-scale genome organization and transcription, in which each is able to modulate or reinforce the activity of the other.


Subject(s)
Chromatin/metabolism , Enhancer Elements, Genetic , Genome, Human , Promoter Regions, Genetic , Transcription, Genetic , Animals , Chromatin/genetics , Humans
3.
Nature ; 626(7997): 207-211, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38086418

ABSTRACT

Enhancers control gene expression and have crucial roles in development and homeostasis1-3. However, the targeted de novo design of enhancers with tissue-specific activities has remained challenging. Here we combine deep learning and transfer learning to design tissue-specific enhancers for five tissues in the Drosophila melanogaster embryo: the central nervous system, epidermis, gut, muscle and brain. We first train convolutional neural networks using genome-wide single-cell assay for transposase-accessible chromatin with sequencing (ATAC-seq) datasets and then fine-tune the convolutional neural networks with smaller-scale data from in vivo enhancer activity assays, yielding models with 13% to 76% positive predictive value according to cross-validation. We designed and experimentally assessed 40 synthetic enhancers (8 per tissue) in vivo, of which 31 (78%) were active and 27 (68%) functioned in the target tissue (100% for central nervous system and muscle). The strategy of combining genome-wide and small-scale functional datasets by transfer learning is generally applicable and should enable the design of tissue-, cell type- and cell state-specific enhancers in any system.


Subject(s)
Deep Learning , Drosophila melanogaster , Embryo, Nonmammalian , Enhancer Elements, Genetic , Neural Networks, Computer , Organ Specificity , Animals , Chromatin/genetics , Chromatin/metabolism , Datasets as Topic , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Enhancer Elements, Genetic/genetics , Organ Specificity/genetics , Reproducibility of Results , Single-Cell Analysis , Transposases/metabolism , Synthetic Biology/methods
4.
Mol Cell ; 82(16): 2922-2924, 2022 08 18.
Article in English | MEDLINE | ID: mdl-35985301

ABSTRACT

By systematically assessing the effects of depleting eight cofactors on enhancer activity, Neumayr et al. (2022) found that different enhancers have different requirements for some perceived universal cofactors. While some cofactors influence enhancer strength, others affect enhancer-promoter specificity.


Subject(s)
Enhancer Elements, Genetic , Promoter Regions, Genetic
5.
Cell ; 148(3): 473-86, 2012 Feb 03.
Article in English | MEDLINE | ID: mdl-22304916

ABSTRACT

Cell fate decisions are driven through the integration of inductive signals and tissue-specific transcription factors (TFs), although the details on how this information converges in cis remain unclear. Here, we demonstrate that the five genetic components essential for cardiac specification in Drosophila, including the effectors of Wg and Dpp signaling, act as a collective unit to cooperatively regulate heart enhancer activity, both in vivo and in vitro. Their combinatorial binding does not require any specific motif orientation or spacing, suggesting an alternative mode of enhancer function whereby cooperative activity occurs with extensive motif flexibility. A fraction of enhancers co-occupied by cardiogenic TFs had unexpected activity in the neighboring visceral mesoderm but could be rendered active in heart through single-site mutations. Given that cardiac and visceral cells are both derived from the dorsal mesoderm, this "dormant" TF binding signature may represent a molecular footprint of these cells' developmental lineage.


Subject(s)
Drosophila melanogaster/cytology , Gene Regulatory Networks , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Mesoderm/cytology , Mesoderm/metabolism , Myocardium/cytology , Myocardium/metabolism , Transcription Factors/metabolism
6.
Nat Rev Genet ; 21(10): 581-596, 2020 10.
Article in English | MEDLINE | ID: mdl-32839576

ABSTRACT

In celebration of the 20th anniversary of Nature Reviews Genetics, we asked 12 leading researchers to reflect on the key challenges and opportunities faced by the field of genetics and genomics. Keeping their particular research area in mind, they take stock of the current state of play and emphasize the work that remains to be done over the next few years so that, ultimately, the benefits of genetic and genomic research can be felt by everyone.


Subject(s)
Disease/genetics , Genetics/trends , Genome, Human , Genome-Wide Association Study , Genomics/trends , Humans
7.
Genes Dev ; 32(1): 42-57, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29378788

ABSTRACT

Gene expression is regulated by promoters, which initiate transcription, and enhancers, which control their temporal and spatial activity. However, the discovery that mammalian enhancers also initiate transcription questions the inherent differences between enhancers and promoters. Here, we investigate the transcriptional properties of enhancers during Drosophila embryogenesis using characterized developmental enhancers. We show that while the timing of enhancer transcription is generally correlated with enhancer activity, the levels and directionality of transcription are highly varied among active enhancers. To assess how this impacts function, we developed a dual transgenic assay to simultaneously measure enhancer and promoter activities from a single element in the same embryo. Extensive transgenic analysis revealed a relationship between the direction of endogenous transcription and the ability to function as an enhancer or promoter in vivo, although enhancer RNA (eRNA) production and activity are not always strictly coupled. Some enhancers (mainly bidirectional) can act as weak promoters, producing overlapping spatio-temporal expression. Conversely, bidirectional promoters often act as strong enhancers, while unidirectional promoters generally cannot. The balance between enhancer and promoter activity is generally reflected in the levels and directionality of eRNA transcription and is likely an inherent sequence property of the elements themselves.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Promoter Regions, Genetic , RNA, Untranslated/biosynthesis , Transcription, Genetic , Animals , Drosophila/embryology , Drosophila/genetics , Embryonic Development/genetics , Humans , K562 Cells
8.
Mol Cell ; 66(4): 439-441, 2017 May 18.
Article in English | MEDLINE | ID: mdl-28525737

ABSTRACT

Although H3K4me1 is a pervasive "mark" of enhancers, its functional requirement for enhancer activity remains unclear. In this issue of Molecular Cell, Dorighi et al. (2017) show that in some contexts, the methyltransferase complex, rather than the H3K4me1 mark, is required for gene expression.


Subject(s)
Enhancer Elements, Genetic , Histones/genetics , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/genetics , Humans , Methylation
9.
Genes Dev ; 31(6): 590-602, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28381411

ABSTRACT

Developmental gene expression is tightly regulated through enhancer elements, which initiate dynamic spatio-temporal expression, and Polycomb response elements (PREs), which maintain stable gene silencing. These two cis-regulatory functions are thought to operate through distinct dedicated elements. By examining the occupancy of the Drosophila pleiohomeotic repressive complex (PhoRC) during embryogenesis, we revealed extensive co-occupancy at developmental enhancers. Using an established in vivo assay for PRE activity, we demonstrated that a subset of characterized developmental enhancers can function as PREs, silencing transcription in a Polycomb-dependent manner. Conversely, some classic Drosophila PREs can function as developmental enhancers in vivo, activating spatio-temporal expression. This study therefore uncovers elements with dual function: activating transcription in some cells (enhancers) while stably maintaining transcriptional silencing in others (PREs). Given that enhancers initiate spatio-temporal gene expression, reuse of the same elements by the Polycomb group (PcG) system may help fine-tune gene expression and ensure the timely maintenance of cell identities.


Subject(s)
Drosophila Proteins/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Polycomb-Group Proteins/metabolism , Response Elements , Animals , Drosophila/embryology , Drosophila/genetics , Drosophila/metabolism , Embryonic Development/genetics
10.
Genome Res ; 31(9): 1573-1581, 2021 09.
Article in English | MEDLINE | ID: mdl-34266978

ABSTRACT

Inter-species comparisons of both morphology and gene expression within a phylum have revealed a period in the middle of embryogenesis with more similarity between species compared with earlier and later time points. This "developmental hourglass" pattern has been observed in many phyla, yet the evolutionary constraints on gene expression, as well as the underlying mechanisms of how this is regulated, remain elusive. Moreover, the role of positive selection on gene regulation in the more diverged earlier and later stages of embryogenesis remains unknown. Here, using DNase-seq to identify regulatory regions in two distant Drosophila species (D. melanogaster and D. virilis), we assessed the evolutionary conservation and adaptive evolution of enhancers throughout multiple stages of embryogenesis. This revealed a higher proportion of conserved enhancers at the phylotypic period, providing a regulatory basis for the hourglass expression pattern. Using an in silico mutagenesis approach, we detect signatures of positive selection on developmental enhancers at early and late stages of embryogenesis, with a depletion at the phylotypic period, suggesting positive selection as one evolutionary mechanism underlying the hourglass pattern of animal evolution.


Subject(s)
Drosophila melanogaster , Evolution, Molecular , Animals , Drosophila/genetics , Drosophila melanogaster/genetics , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Regulatory Sequences, Nucleic Acid
11.
Genome Res ; 31(2): 211-224, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33310749

ABSTRACT

Precise patterns of gene expression are driven by interactions between transcription factors, regulatory DNA sequences, and chromatin. How DNA mutations affecting any one of these regulatory "layers" are buffered or propagated to gene expression remains unclear. To address this, we quantified allele-specific changes in chromatin accessibility, histone modifications, and gene expression in F1 embryos generated from eight Drosophila crosses at three embryonic stages, yielding a comprehensive data set of 240 samples spanning multiple regulatory layers. Genetic variation (allelic imbalance) impacts gene expression more frequently than chromatin features, with metabolic and environmental response genes being most often affected. Allelic imbalance in cis-regulatory elements (enhancers) is common and highly heritable, yet its functional impact does not generally propagate to gene expression. When it does, genetic variation impacts RNA levels through two alternative mechanisms involving either H3K4me3 or chromatin accessibility and H3K27ac. Changes in RNA are more predictive of variation in H3K4me3 than vice versa, suggesting a role for H3K4me3 downstream from transcription. The impact of a substantial proportion of genetic variation is consistent across embryonic stages, with 50% of allelic imbalanced features at one stage being also imbalanced at subsequent developmental stages. Crucially, buffering, as well as the magnitude and evolutionary impact of genetic variants, is influenced by regulatory complexity (i.e., number of enhancers regulating a gene), with transcription factors being most robust to cis-acting, but most influenced by trans-acting, variation.

12.
Nature ; 555(7697): 538-542, 2018 03 22.
Article in English | MEDLINE | ID: mdl-29539636

ABSTRACT

Understanding how gene regulatory networks control the progressive restriction of cell fates is a long-standing challenge. Recent advances in measuring gene expression in single cells are providing new insights into lineage commitment. However, the regulatory events underlying these changes remain unclear. Here we investigate the dynamics of chromatin regulatory landscapes during embryogenesis at single-cell resolution. Using single-cell combinatorial indexing assay for transposase accessible chromatin with sequencing (sci-ATAC-seq), we profiled chromatin accessibility in over 20,000 single nuclei from fixed Drosophila melanogaster embryos spanning three landmark embryonic stages: 2-4 h after egg laying (predominantly stage 5 blastoderm nuclei), when each embryo comprises around 6,000 multipotent cells; 6-8 h after egg laying (predominantly stage 10-11), to capture a midpoint in embryonic development when major lineages in the mesoderm and ectoderm are specified; and 10-12 h after egg laying (predominantly stage 13), when each of the embryo's more than 20,000 cells are undergoing terminal differentiation. Our results show that there is spatial heterogeneity in the accessibility of the regulatory genome before gastrulation, a feature that aligns with future cell fate, and that nuclei can be temporally ordered along developmental trajectories. During mid-embryogenesis, tissue granularity emerges such that individual cell types can be inferred by their chromatin accessibility while maintaining a signature of their germ layer of origin. Analysis of the data reveals overlapping usage of regulatory elements between cells of the endoderm and non-myogenic mesoderm, suggesting a common developmental program that is reminiscent of the mesendoderm lineage in other species. We identify 30,075 distal regulatory elements that exhibit tissue-specific accessibility. We validated the germ-layer specificity of a subset of these predicted enhancers in transgenic embryos, achieving an accuracy of 90%. Overall, our results demonstrate the power of shotgun single-cell profiling of embryos to resolve dynamic changes in the chromatin landscape during development, and to uncover the cis-regulatory programs of metazoan germ layers and cell types.


Subject(s)
Drosophila melanogaster/cytology , Drosophila melanogaster/embryology , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Single-Cell Analysis , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Chromatin/genetics , Chromatin/metabolism , Drosophila melanogaster/genetics , Endoderm/cytology , Endoderm/metabolism , Enhancer Elements, Genetic/genetics , Female , Gastrulation/genetics , Genome, Insect/genetics , Male , Mesoderm/cytology , Mesoderm/metabolism , Organ Specificity/genetics , Organisms, Genetically Modified/cytology , Organisms, Genetically Modified/genetics , Reproducibility of Results
13.
Nature ; 541(7637): 402-406, 2017 01 19.
Article in English | MEDLINE | ID: mdl-28024300

ABSTRACT

Embryonic development is driven by tightly regulated patterns of gene expression, despite extensive genetic variation among individuals. Studies of expression quantitative trait loci (eQTL) indicate that genetic variation frequently alters gene expression in cell-culture models and differentiated tissues. However, the extent and types of genetic variation impacting embryonic gene expression, and their interactions with developmental programs, remain largely unknown. Here we assessed the effect of genetic variation on transcriptional (expression levels) and post-transcriptional (3' RNA processing) regulation across multiple stages of metazoan development, using 80 inbred Drosophila wild isolates, identifying thousands of developmental-stage-specific and shared QTL. Given the small blocks of linkage disequilibrium in Drosophila, we obtain near base-pair resolution, resolving causal mutations in developmental enhancers, validated transcription-factor-binding sites and RNA motifs. This fine-grain mapping uncovered extensive allelic interactions within enhancers that have opposite effects, thereby buffering their impact on enhancer activity. QTL affecting 3' RNA processing identify new functional motifs leading to transcript isoform diversity and changes in the lengths of 3' untranslated regions. These results highlight how developmental stage influences the effects of genetic variation and uncover multiple mechanisms that regulate and buffer expression variation during embryogenesis.


Subject(s)
Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Genetic Variation , 3' Untranslated Regions/genetics , Alleles , Animals , Binding Sites , Enhancer Elements, Genetic , Linkage Disequilibrium , Mutation , Quantitative Trait Loci , RNA 3' End Processing , Transcription Factors/metabolism , Transcription, Genetic
14.
Development ; 146(19)2019 09 25.
Article in English | MEDLINE | ID: mdl-31554625

ABSTRACT

In the past decade, two major advances in our understanding of nuclear organization have taken the field of gene regulation by storm. First, technologies that can analyze the three-dimensional conformation of chromatin have revealed how the genome is organized and have provided novel insights into how regulatory regions in the genome interact. Second, the recognition that many proteins can form membraneless compartments through liquid-liquid phase separation (LLPS) has challenged long-standing notions of how proteins within the nucleus are organized and has offered a tantalizing general mechanism by which many aspects of nuclear function may be regulated. However, the functional roles of chromatin topology and LLPS in regulating gene expression remain poorly understood. These topics were discussed with great fervor during an open discussion held at a recent workshop titled 'Chromatin-based regulation of development' organized by The Company of Biologists. Here, we summarize the major points covered during this debate and discuss how they tie into current thinking in the field of gene regulation.


Subject(s)
Chromatin/chemistry , Chromatin/metabolism , Gene Expression Regulation, Developmental , Nucleic Acid Conformation , Animals , Enhancer Elements, Genetic/genetics , Humans , Repressor Proteins/metabolism
15.
PLoS Genet ; 15(9): e1008382, 2019 09.
Article in English | MEDLINE | ID: mdl-31553718

ABSTRACT

Comprehensive information on the timing and location of gene expression is fundamental to our understanding of embryonic development and tissue formation. While high-throughput in situ hybridization projects provide invaluable information about developmental gene expression patterns for model organisms like Drosophila, the output of these experiments is primarily qualitative, and a high proportion of protein coding genes and most non-coding genes lack any annotation. Accurate data-centric predictions of spatio-temporal gene expression will therefore complement current in situ hybridization efforts. Here, we applied a machine learning approach by training models on all public gene expression and chromatin data, even from whole-organism experiments, to provide genome-wide, quantitative spatio-temporal predictions for all genes. We developed structured in silico nano-dissection, a computational approach that predicts gene expression in >200 tissue-developmental stages. The algorithm integrates expression signals from a compendium of 6,378 genome-wide expression and chromatin profiling experiments in a cell lineage-aware fashion. We systematically evaluated our performance via cross-validation and experimentally confirmed 22 new predictions for four different embryonic tissues. The model also predicts complex, multi-tissue expression and developmental regulation with high accuracy. We further show the potential of applying these genome-wide predictions to extract tissue specificity signals from non-tissue-dissected experiments, and to prioritize tissues and stages for disease modeling. This resource, together with the exploratory tools are freely available at our webserver http://find.princeton.edu, which provides a valuable tool for a range of applications, from predicting spatio-temporal expression patterns to recognizing tissue signatures from differential gene expression profiles.


Subject(s)
Gene Expression Profiling/methods , Gene Expression Regulation, Developmental/genetics , Genome-Wide Association Study/methods , Algorithms , Animals , Computational Biology/methods , Computer Simulation , Drosophila/genetics , Embryonic Development/genetics , Forecasting/methods , Gene Expression Regulation, Developmental/physiology , Genes, Developmental/genetics , Machine Learning , Spatio-Temporal Analysis , Transcriptome/genetics
16.
Genes Dev ; 28(2): 167-81, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24402316

ABSTRACT

The transcription factors of the Snail family are key regulators of epithelial-mesenchymal transitions, cell morphogenesis, and tumor metastasis. Since its discovery in Drosophila ∼25 years ago, Snail has been extensively studied for its role as a transcriptional repressor. Here we demonstrate that Drosophila Snail can positively modulate transcriptional activation. By combining information on in vivo occupancy with expression profiling of hand-selected, staged snail mutant embryos, we identified 106 genes that are potentially directly regulated by Snail during mesoderm development. In addition to the expected Snail-repressed genes, almost 50% of Snail targets showed an unanticipated activation. The majority of "Snail-activated" genes have enhancer elements cobound by Twist and are expressed in the mesoderm at the stages of Snail occupancy. Snail can potentiate Twist-mediated enhancer activation in vitro and is essential for enhancer activity in vivo. Using a machine learning approach, we show that differentially enriched motifs are sufficient to predict Snail's regulatory response. In silico mutagenesis revealed a likely causative motif, which we demonstrate is essential for enhancer activation. Taken together, these data indicate that Snail can potentiate enhancer activation by collaborating with different activators, providing a new mechanism by which Snail regulates development.


Subject(s)
Drosophila/genetics , Drosophila/metabolism , Transcription Factors/metabolism , Amino Acid Motifs , Animals , Drosophila/embryology , Drosophila Proteins/metabolism , Embryo, Nonmammalian , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental , Mesoderm/metabolism , Protein Binding , Snail Family Transcription Factors , Transcription Factors/genetics , Twist-Related Protein 1/metabolism
17.
Nature ; 512(7512): 96-100, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-25043061

ABSTRACT

Developmental enhancers initiate transcription and are fundamental to our understanding of developmental networks, evolution and disease. Despite their importance, the properties governing enhancer-promoter interactions and their dynamics during embryogenesis remain unclear. At the ß-globin locus, enhancer-promoter interactions appear dynamic and cell-type specific, whereas at the HoxD locus they are stable and ubiquitous, being present in tissues where the target genes are not expressed. The extent to which preformed enhancer-promoter conformations exist at other, more typical, loci and how transcription is eventually triggered is unclear. Here we generated a high-resolution map of enhancer three-dimensional contacts during Drosophila embryogenesis, covering two developmental stages and tissue contexts, at unprecedented resolution. Although local regulatory interactions are common, long-range interactions are highly prevalent within the compact Drosophila genome. Each enhancer contacts multiple enhancers, and promoters with similar expression, suggesting a role in their co-regulation. Notably, most interactions appear unchanged between tissue context and across development, arising before gene activation, and are frequently associated with paused RNA polymerase. Our results indicate that the general topology governing enhancer contacts is conserved from flies to humans and suggest that transcription initiates from preformed enhancer-promoter loops through release of paused polymerase.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , Drosophila melanogaster/enzymology , Drosophila melanogaster/genetics , Embryonic Development/genetics , Enhancer Elements, Genetic/genetics , Promoter Regions, Genetic/genetics , Animals , Binding Sites , Chromosomes, Insect/genetics , Chromosomes, Insect/metabolism , Drosophila melanogaster/embryology , Gene Expression Regulation, Developmental/genetics , Genetic Loci/genetics , Genome, Insect/genetics , Humans , Transcription Initiation, Genetic , Transcriptional Activation
18.
Development ; 143(23): 4533-4542, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27899510

ABSTRACT

Developmental patterning and tissue formation are regulated through complex gene regulatory networks (GRNs) driven through the action of transcription factors (TFs) converging on enhancer elements. Here, as a point of entry to dissect the poorly defined GRN underlying cardiomyocyte differentiation, we apply an integrated approach to identify active enhancers and TFs involved in Drosophila heart development. The Drosophila heart consists of 104 cardiomyocytes, representing less than 0.5% of all cells in the embryo. By modifying BiTS-ChIP for rare cells, we examined H3K4me3 and H3K27ac chromatin landscapes to identify active promoters and enhancers specifically in cardiomyocytes. These in vivo data were complemented by a machine learning approach and extensive in vivo validation in transgenic embryos, which identified many new heart enhancers and their associated TF motifs. Our results implicate many new TFs in late stages of heart development, including Bagpipe, an Nkx3.2 ortholog, which we show is essential for differentiated heart function.


Subject(s)
Drosophila/embryology , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental/genetics , Heart/embryology , Myocytes, Cardiac/cytology , Organogenesis/genetics , Promoter Regions, Genetic/genetics , Animals , Animals, Genetically Modified , Cell Differentiation/genetics , Chromatin , Drosophila/genetics , Gene Regulatory Networks/genetics , Histones/metabolism , Organogenesis/physiology , Transcription Factors/genetics
19.
Nucleic Acids Res ; 45(21): 12285-12300, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-29036346

ABSTRACT

A special class of poorly characterized architectural proteins is required for chromatin topology and enhancer-promoter interactions. Here, we identify Opbp as a new Drosophila architectural protein, interacting with CP190 both in vivo and in vitro. Opbp binds to a very restrictive set of genomic regions, through a rare sequence specific motif. These sites are co-bound by CP190 in vivo, and generally located at bidirectional promoters of ribosomal protein genes. We show that Opbp is essential for viability, and loss of opbp function, or destruction of its motif, leads to reduced ribosomal protein gene expression, indicating a functional role in promoter activation. As characteristic of architectural/insulator proteins, the Opbp motif is sufficient for distance-dependent reporter gene activation and enhancer-blocking activity, suggesting an Opbp-mediated enhancer-promoter interaction. Rather than having a constitutive role, Opbp represents a new type of architectural protein with a very restricted, yet essential, function in regulation of housekeeping gene expression.


Subject(s)
Drosophila Proteins/metabolism , Gene Expression Regulation , Genes, rRNA , Transcription Factors/metabolism , Animals , CRISPR-Cas Systems , Chromatin/metabolism , Drosophila/genetics , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila Proteins/physiology , Enhancer Elements, Genetic , Gene Deletion , Microtubule-Associated Proteins/metabolism , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Ribosomal Proteins/genetics , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/physiology , Transcriptional Activation
20.
Anal Chem ; 90(10): 5982-5988, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29688703

ABSTRACT

Partitioning and sorting particles, including molecules, cells and organisms, is an essential prerequisite for a diverse range of applications. Here, we describe a very economical microfluidic platform (built from parts costing about U.S. $6800 for a stand-alone system or U.S. $3700, when mounted on an existing fluorescence microscope connected to a computer) to sort droplets, cells and embryos, based on imaging data. Valves operated by a Braille display are used to open and close microfluidic channels, enabling sorting at rates of >2 Hz. Furthermore, we show microfluidic 8-way sorting for the first time, facilitating the simultaneous separation and collection of objects with diverse characteristics/phenotypes. Due to the high flexibility in the size of objects that can be sorted, the low cost, and the many possibilities enabled by imaging technology, we believe that our approach nicely complements existing FACS and µFACS technology.


Subject(s)
Cell Separation/economics , Drosophila/embryology , Microfluidic Analytical Techniques/economics , Adsorption , Animals , Cell Line, Tumor , Humans , Microfluidic Analytical Techniques/instrumentation , Microscopy, Fluorescence , Particle Size , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL