Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Curr Top Dev Biol ; 159: 406-437, 2024.
Article in English | MEDLINE | ID: mdl-38729683

ABSTRACT

Transcriptional regulation plays a pivotal role in orchestrating the intricate genetic programs governing embryonic development. The expression of developmental genes relies on the combined activity of several cis-regulatory elements (CREs), such as enhancers and silencers, which can be located at long linear distances from the genes that they regulate and that interact with them through establishment of chromatin loops. Mutations affecting their activity or interaction with their target genes can lead to developmental disorders and are thought to have importantly contributed to the evolution of the animal body plan. The income of next-generation-sequencing approaches has allowed identifying over a million of sequences with putative regulatory potential in the human genome. Characterizing their function and establishing gene-CREs maps is essential to decode the logic governing developmental gene expression and is one of the major challenges of the post-genomic era. Chromatin 3D organization plays an essential role in determining how CREs specifically contact their target genes while avoiding deleterious off-target interactions. Our understanding of these aspects has greatly advanced with the income of chromatin conformation capture techniques and fluorescence microscopy approaches to visualize the organization of DNA elements in the nucleus. Here we will summarize relevant aspects of how the interplay between CRE activity and chromatin 3D organization regulates developmental gene expression and how it relates to pathological conditions and the evolution of animal body plan.


Subject(s)
Chromatin , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Humans , Chromatin/metabolism , Chromatin/genetics , Animals , Evolution, Molecular
2.
Nat Genet ; 56(4): 686-696, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38467791

ABSTRACT

To regulate expression, enhancers must come in proximity to their target gene. However, the relationship between the timing of enhancer-promoter (E-P) proximity and activity remains unclear, with examples of uncoupled, anticorrelated and correlated interactions. To assess this, we selected 600 characterized enhancers or promoters with tissue-specific activity in Drosophila embryos and performed Capture-C in FACS-purified myogenic or neurogenic cells during specification and tissue differentiation. This enabled direct comparison between E-P proximity and activity transitioning from OFF-to-ON and ON-to-OFF states across developmental conditions. This showed remarkably similar E-P topologies between specified muscle and neuronal cells, which are uncoupled from activity. During tissue differentiation, many new distal interactions emerge where changes in E-P proximity reflect changes in activity. The mode of E-P regulation therefore appears to change as embryogenesis proceeds, from largely permissive topologies during cell-fate specification to more instructive regulation during terminal tissue differentiation, when E-P proximity is coupled to activation.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Animals , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental/genetics , Promoter Regions, Genetic/genetics , Drosophila/genetics , Cell Differentiation/genetics
3.
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
4.
Dev Cell ; 56(16): 2348-2363.e8, 2021 08 23.
Article in English | MEDLINE | ID: mdl-34363757

ABSTRACT

Many developmental regulators have complex and context-specific roles in different tissues and stages, making the dissection of their function extremely challenging. As regulatory processes often occur within minutes, perturbation methods that match these dynamics are needed. Here, we present the improved light-inducible nuclear export system (iLEXY), an optogenetic loss-of-function approach that triggers translocation of proteins from the nucleus to the cytoplasm. By introducing a series of mutations, we substantially increased LEXY's efficiency and generated variants with different recovery times. iLEXY enables rapid (t1/2 < 30 s), efficient, and reversible nuclear protein depletion in embryos, and is generalizable to proteins of diverse sizes and functions. Applying iLEXY to the Drosophila master regulator Twist, we phenocopy loss-of-function mutants, precisely map the Twist-sensitive embryonic stages, and investigate the effects of timed Twist depletions. Our results demonstrate the power of iLEXY to dissect the function of pleiotropic factors during embryogenesis with unprecedented temporal precision.


Subject(s)
Cell Nucleus/metabolism , Optogenetics/methods , Active Transport, Cell Nucleus , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , Embryo, Nonmammalian/metabolism , Loss of Function Mutation , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism
5.
Dev Cell ; 55(5): 648-664.e9, 2020 12 07.
Article in English | MEDLINE | ID: mdl-33171098

ABSTRACT

Enhancers are essential drivers of cell states, yet the relationship between accessibility, regulatory activity, and in vivo lineage commitment during embryogenesis remains poorly understood. Here, we measure chromatin accessibility in isolated neural and mesodermal lineages across a time course of Drosophila embryogenesis. Promoters, including tissue-specific genes, are often constitutively open, even in contexts where the gene is not expressed. In contrast, the majority of distal elements have dynamic, tissue-specific accessibility. Enhancer priming appears rarely within a lineage, perhaps reflecting the speed of Drosophila embryogenesis. However, many tissue-specific enhancers are accessible in other lineages early on and become progressively closed as embryogenesis proceeds. We demonstrate the usefulness of this tissue- and time-resolved resource to definitively identify single-cell clusters, to uncover predictive motifs, and to identify many regulators of tissue development. For one such predicted neural regulator, l(3)neo38, we generate a loss-of-function mutant and uncover an essential role for neuromuscular junction and brain development.


Subject(s)
Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Embryonic Development/genetics , Enhancer Elements, Genetic , Promoter Regions, Genetic , Animals , Cell Lineage/genetics , Chromatin , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Mesoderm/embryology , Muscles/embryology , Neurons/cytology , Organ Specificity/genetics , Protein Binding , Single-Cell Analysis , Time Factors , Transcription Factors/metabolism
6.
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
7.
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
8.
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
9.
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
10.
J Biol Chem ; 290(45): 26927-26942, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26378230

ABSTRACT

A well integrated and hierarchically organized gene regulatory network is responsible for the progressive specification of the forebrain. The transcription factor Six3 is one of the central components of this network. As such, Six3 regulates several components of the network, but its upstream regulators are still poorly characterized. Here we have systematically identified such regulators, taking advantage of the detailed functional characterization of the regulatory region of the medaka fish Six3.2 ortholog and of a time/cost-effective trans-regulatory screening, which complemented and overcame the limitations of in silico prediction approaches. The candidates resulting from this search were validated with dose-response luciferase assays and expression pattern criteria. Reconfirmed candidates with a matching expression pattern were also tested with chromatin immunoprecipitation and functional studies. Our results confirm the previously proposed direct regulation of Pax6 and further demonstrate that Msx2 and Pbx1 are bona fide direct regulators of early Six3.2 distribution in distinct domains of the medaka fish forebrain. They also point to other transcription factors, including Tcf3, as additional regulators of different spatial-temporal domains of Six3.2 expression. The activity of these regulators is discussed in the context of the gene regulatory network proposed for the specification of the forebrain.


Subject(s)
Eye Proteins/genetics , Fish Proteins/genetics , Homeodomain Proteins/genetics , Nerve Tissue Proteins/genetics , Oryzias/embryology , Oryzias/genetics , Prosencephalon/embryology , Prosencephalon/metabolism , Animals , Animals, Genetically Modified , Body Patterning/genetics , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Oryzias/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/genetics , Homeobox Protein SIX3
11.
Nucleic Acids Res ; 42(12): 7793-806, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24895435

ABSTRACT

Vertebrate organogenesis is critically sensitive to gene dosage and even subtle variations in the expression levels of key genes may result in a variety of tissue anomalies. MicroRNAs (miRNAs) are fundamental regulators of gene expression and their role in vertebrate tissue patterning is just beginning to be elucidated. To gain further insight into this issue, we analysed the transcriptomic consequences of manipulating the expression of miR-204 in the Medaka fish model system. We used RNA-Seq and an innovative bioinformatics approach, which combines conventional differential expression analysis with the behavior expected by miR-204 targets after its overexpression and knockdown. With this approach combined with a correlative analysis of the putative targets, we identified a wider set of miR-204 target genes belonging to different pathways. Together, these approaches confirmed that miR-204 has a key role in eye development and further highlighted its putative function in neural differentiation processes, including axon guidance as supported by in vivo functional studies. Together, our results demonstrate the advantage of integrating next-generation sequencing and bioinformatics approaches to investigate miRNA biology and provide new important information on the role of miRNAs in the control of axon guidance and more broadly in nervous system development.


Subject(s)
Axons/physiology , Gene Expression Profiling , MicroRNAs/metabolism , Neurogenesis/genetics , Oryzias/genetics , Animals , Axons/ultrastructure , Computational Biology , Gene Knockdown Techniques , High-Throughput Nucleotide Sequencing , Models, Animal , Oryzias/embryology , Oryzias/metabolism , Retina/embryology , Retina/metabolism , Retina/ultrastructure , Sequence Analysis, RNA
12.
Mech Dev ; 130(2-3): 95-111, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23111324

ABSTRACT

The vertebrate forebrain or prosencephalon is patterned at the beginning of neurulation into four major domains: the telencephalic, hypothalamic, retinal and diencephalic anlagen. These domains will then give rise to the majority of the brain structures involved in sensory integration and the control of higher intellectual and homeostatic functions. Understanding how forebrain pattering arises has thus attracted the interest of developmental neurobiologists for decades. As a result, most of its regulators have been identified and their hierarchical relationship is now the object of active investigation. Here, we summarize the main morphogenetic pathways and transcription factors involved in forebrain specification and propose the backbone of a possible gene regulatory network (GRN) governing its specification, taking advantage of the GRN principles elaborated by pioneer studies in simpler organisms. We will also discuss this GRN and its operational logic in the context of the remarkable morphological and functional diversification that the forebrain has undergone during evolution.


Subject(s)
Gene Regulatory Networks , Prosencephalon/embryology , Animals , Body Patterning , Gene Expression Regulation, Developmental , Humans , Signal Transduction , Transcription Factors/physiology , Vertebrates
13.
Genome Biol ; 13(3): R24, 2012.
Article in English | MEDLINE | ID: mdl-22458515

ABSTRACT

Here we present the Transcription Factor Encyclopedia (TFe), a new web-based compendium of mini review articles on transcription factors (TFs) that is founded on the principles of open access and collaboration. Our consortium of over 100 researchers has collectively contributed over 130 mini review articles on pertinent human, mouse and rat TFs. Notable features of the TFe website include a high-quality PDF generator and web API for programmatic data retrieval. TFe aims to rapidly educate scientists about the TFs they encounter through the delivery of succinct summaries written and vetted by experts in the field. TFe is available at http://www.cisreg.ca/tfe.


Subject(s)
Computational Biology , Databases, Protein/supply & distribution , Transcription Factors/genetics , Access to Information , Animals , Encyclopedias as Topic , Humans , Internet , Mice , Rats , Transcription, Genetic
14.
Proc Natl Acad Sci U S A ; 107(35): 15491-6, 2010 Aug 31.
Article in English | MEDLINE | ID: mdl-20713703

ABSTRACT

MicroRNAs (miRNAs) are small noncoding RNAs that have important roles in the regulation of gene expression. The roles of individual miRNAs in controlling vertebrate eye development remain, however, largely unexplored. Here, we show that a single miRNA, miR-204, regulates multiple aspects of eye development in the medaka fish (Oryzias latipes). Morpholino-mediated ablation of miR-204 expression resulted in an eye phenotype characterized by microphthalmia, abnormal lens formation, and altered dorsoventral (D-V) patterning of the retina, which is associated with optic fissure coloboma. Using a variety of in vivo and in vitro approaches, we identified the transcription factor Meis2 as one of the main targets of miR-204 function. We show that, together with altered regulation of the Pax6 pathway, the abnormally elevated levels of Meis2 resulting from miR-204 inactivation are largely responsible for the observed phenotype. These data provide an example of how a specific miRNA can regulate multiple events in eye formation; at the same time, they uncover an as yet unreported function of Meis2 in the specification of D-V patterning of the retina.


Subject(s)
Fish Proteins/genetics , Lens, Crystalline/metabolism , MicroRNAs/genetics , Retina/metabolism , Transcription Factors/genetics , Animals , Cell Line , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Humans , In Situ Hybridization , Lens, Crystalline/embryology , Models, Genetic , Organogenesis/genetics , Oryzias/embryology , Oryzias/genetics , Retina/embryology , Reverse Transcriptase Polymerase Chain Reaction
15.
Development ; 137(14): 2307-17, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20534668

ABSTRACT

Timely generation of distinct neural cell types in appropriate numbers is fundamental for the generation of a functional retina. In vertebrates, the transcription factor Six6 is initially expressed in multipotent retina progenitors and then becomes restricted to differentiated retinal ganglion and amacrine cells. How Six6 expression in the retina is controlled and what are its precise functions are still unclear. To address this issue, we used bioinformatic searches and transgenic approaches in medaka fish (Oryzias latipes) to characterise highly conserved regulatory enhancers responsible for Six6 expression. One of the enhancers drove gene expression in the differentiating and adult retina. A search for transcription factor binding sites, together with luciferase, ChIP assays and gain-of-function studies, indicated that NeuroD, a bHLH transcription factor, directly binds an 'E-box' sequence present in this enhancer and specifically regulates Six6 expression in the retina. NeuroD-induced Six6 overexpression in medaka embryos promoted unorganized retinal progenitor proliferation and, most notably, impaired photoreceptor differentiation, with no apparent changes in other retinal cell types. Conversely, Six6 gain- and loss-of-function changed NeuroD expression levels and altered the expression of the photoreceptor differentiation marker Rhodopsin. In addition, knockdown of Six6 interfered with amacrine cell generation. Together, these results indicate that Six6 and NeuroD control the expression of each other and their functions coordinate amacrine cell generation and photoreceptor terminal differentiation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Transcription Factors/metabolism , Amacrine Cells/metabolism , Animals , Animals, Genetically Modified , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , Chromatin Immunoprecipitation , Gene Expression , Neurons/metabolism , Oryzias/genetics , Oryzias/metabolism , Photoreceptor Cells , Retina/cytology , Retina/metabolism , Retinal Ganglion Cells/metabolism , Rhodopsin/genetics , Rhodopsin/metabolism , Transcription Factors/genetics
16.
Dev Biol ; 337(1): 16-28, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19835855

ABSTRACT

Although tight quantitative control of gene expression is required to ensure that organs and tissues function correctly, the transcriptional mechanisms underlying this process still remain poorly understood. Here, we describe novel and evolutionary conserved secondary enhancers that are needed for the regulation of the expression of Troponin I genes. Secondary enhancers are silent when tested individually in electroporated muscles but interact with the primary enhancers and are required to precisely control the appropriate timing, the tissue and fibre specificity, and the quantitative expression of these genes during muscle differentiation. Synergism is completely dependent of the fully conserved MEF2 site present on the primary enhancers core of skeletal muscle Troponin I genes. Thus, while each of these paired enhancers has a different function, the concerted action of both is crucial to recapitulate endogenous gene expression. Through comparative genomics, we predict that this mechanism has also arisen in other mammalian muscle genes. Our results reveal the existence of a novel mechanism, conserved from flies to mammals, to fine-tune gene expression in each muscle and probably other tissues.


Subject(s)
Enhancer Elements, Genetic/physiology , Gene Expression Regulation , Muscle, Skeletal/metabolism , Myocytes, Cardiac/metabolism , Troponin I/genetics , Animals , Binding Sites , Cells, Cultured , Conserved Sequence , Lac Operon , MEF2 Transcription Factors , Male , Mice , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Slow-Twitch/metabolism , Myogenic Regulatory Factors/metabolism , Rats , Rats, Wistar
17.
J Biol Chem ; 283(18): 12333-42, 2008 May 02.
Article in English | MEDLINE | ID: mdl-18308726

ABSTRACT

Characterization of the basal transcription machinery of mitochondrial DNA (mtDNA) is critical to understand mitochondrial pathophysiology. In mammalian in vitro systems, mtDNA transcription requires mtRNA polymerase, transcription factor A (TFAM), and either transcription factor B1 (TFB1M) or B2 (TFB2M). We have silenced the expression of TFB2M by RNA interference in Drosophila melanogaster. RNA interference knockdown of TF2BM causes lethality by arrest of larval development. Molecular analysis demonstrates that TF2BM is essential for mtDNA transcription during Drosophila development and is not redundant with TFB1M. The impairment of mtDNA transcription causes a dramatic decrease in oxidative phosphorylation and mitochondrial ATP synthesis in the long-lived larvae, and a metabolic shift to glycolysis, which partially restores ATP levels and elicits a compensatory response at the nuclear level that increases mitochondrial mass. At the cellular level, the mitochondrial dysfunction induced by TFB2M knockdown causes a severe reduction in cell proliferation without affecting cell growth, and increases the level of apoptosis. In contrast, cell differentiation and morphogenesis are largely unaffected. Our data demonstrate the essential role of TFB2M in mtDNA transcription in a multicellular organism, and reveal the complex cellular, biochemical, and molecular responses induced by impairment of oxidative phosphorylation during Drosophila development.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Mitochondria/metabolism , Adenosine Triphosphate/biosynthesis , Animals , Apoptosis , Body Patterning , Body Weight , Cell Proliferation , DNA, Mitochondrial/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Energy Metabolism , Gene Expression Regulation, Developmental , Gene Silencing , Glycolysis , Larva/cytology , Larva/growth & development , Longevity , Oxidative Phosphorylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic , Wings, Animal/cytology
18.
Mech Dev ; 122(5): 681-94, 2005 May.
Article in English | MEDLINE | ID: mdl-15817225

ABSTRACT

The distinct muscles of an organism accumulate different quantities of structural proteins, but always maintaining their stoichiometry. However, the mechanisms that control the levels of these proteins and that co-ordinate muscle gene expression remain to be defined. The paramyosin/miniparamyosin gene encodes two thick filament proteins transcribed from two different promoters. We have analysed the regulatory regions that control expression of this gene and that are situated in the two promoters, the 5' and the internal promoters, both in vivo and in silico. A distal muscle enhancer containing three conserved MEF2 motifs is essential to drive high levels of paramyosin expression in all the major embryonic, larval and adult muscles. This enhancer shares sequence motifs, as well as its structure and organisation, with at least four co-regulated muscle enhancers that direct similar patterns of expression. However, other elements located downstream of the enhancer are also required for correct gene expression. Other muscle genes with different patterns of expression, such as miniparamyosin, are regulated by other basic mechanisms. The expression of miniparamyosin is controlled by two enhancers, AB and TX, but a BF modulator is required to ensure the correct levels of expression in each particular muscle. We propose a mechanism of transcriptional regulation in which similar enhancers are responsible for the spatio-temporal expression of co-regulated genes. However, it is the interaction between enhancers which ensures that the correct amounts of protein are expressed at any particular time in a cell, adapting these levels to their specific needs. These mechanisms may not be exclusive to neural or muscle tissue and might represent a general mechanism for genes that are spatially and temporally co-regulated.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Muscles/embryology , Muscles/metabolism , Promoter Regions, Genetic , Tropomyosin/genetics , Amino Acid Motifs , Animals , Base Sequence , Binding Sites , Drosophila melanogaster , Genes, Reporter , Genome , Models, Genetic , Molecular Sequence Data , Neurons/metabolism , Nucleotides/chemistry , Phylogeny , Plasmids/metabolism , Protein Biosynthesis , Sequence Homology, Nucleic Acid , Tissue Distribution , Transgenes , Tropomyosin/biosynthesis , Two-Hybrid System Techniques , beta-Galactosidase/metabolism
19.
Biochem J ; 386(Pt 1): 145-52, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15469415

ABSTRACT

Formation of the contractile apparatus in muscle cells requires co-ordinated activation of several genes and the proper assembly of their products. To investigate the role of TnT (troponin T) in the mechanisms that control and co-ordinate thin-filament formation, we generated transgenic Drosophila lines that overexpress TnT in their indirect flight muscles. All flies that overexpress TnT were unable to fly, and the loss of thin filaments themselves was coupled with ultrastructural perturbations of the sarcomere. In contrast, thick filaments remained largely unaffected. Biochemical analysis of these lines revealed that the increase in TnT levels could be detected only during the early stages of adult muscle formation and was followed by a profound decrease in the amount of this protein as well as that of other thin-filament proteins such as tropomyosin, troponin I and actin. The decrease in thin-filament proteins is not only due to degradation but also due to a decrease in their synthesis, since accumulation of their mRNA transcripts was also severely diminished. This decrease in expression levels of the distinct thin-filament components led us to postulate that any change in the amount of TnT transcripts might trigger the down-regulation of other co-regulated thin-filament components. Taken together, these results suggest the existence of a mechanism that tightly co-ordinates the expression of thin-filament genes and controls the correct stoichiometry of these proteins. We propose that the high levels of unassembled protein might act as a sensor in this process.


Subject(s)
Actin Cytoskeleton/metabolism , Drosophila Proteins/physiology , Drosophila melanogaster/metabolism , Gene Expression Regulation, Developmental/physiology , Muscle Proteins/biosynthesis , Muscles/metabolism , Troponin T/physiology , Animals , Animals, Genetically Modified , Drosophila Proteins/biosynthesis , Drosophila Proteins/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/ultrastructure , Feedback, Physiological , Flight, Animal , Muscles/ultrastructure , Muscular Diseases/etiology , Muscular Diseases/pathology , Pupa , RNA, Messenger/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/physiology , Sarcomeres/ultrastructure , Troponin T/biosynthesis , Troponin T/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...