Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
PLoS Genet ; 17(4): e1009536, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33901190

RESUMO

Several distinct activities and functions have been described for chromatin insulators, which separate genes along chromosomes into functional units. Here, we describe a novel mechanism of functional separation whereby an insulator prevents gene repression. When the homie insulator is deleted from the end of a Drosophila even skipped (eve) locus, a flanking P-element promoter is activated in a partial eve pattern, causing expression driven by enhancers in the 3' region to be repressed. The mechanism involves transcriptional read-through from the flanking promoter. This conclusion is based on the following. Read-through driven by a heterologous enhancer is sufficient to repress, even when homie is in place. Furthermore, when the flanking promoter is turned around, repression is minimal. Transcriptional read-through that does not produce anti-sense RNA can still repress expression, ruling out RNAi as the mechanism in this case. Thus, transcriptional interference, caused by enhancer capture and read-through when the insulator is removed, represses eve promoter-driven expression. We also show that enhancer-promoter specificity and processivity of transcription can have decisive effects on the consequences of insulator removal. First, a core heat shock 70 promoter that is not activated well by eve enhancers did not cause read-through sufficient to repress the eve promoter. Second, these transcripts are less processive than those initiated at the P-promoter, measured by how far they extend through the eve locus, and so are less disruptive. These results highlight the importance of considering transcriptional read-through when assessing the effects of insulators on gene expression.


Assuntos
Proteínas de Drosophila/genética , Elementos Facilitadores Genéticos/genética , Proteínas de Homeodomínio/genética , Elementos Isolantes/genética , Regiões Promotoras Genéticas/genética , Fatores de Transcrição/genética , Animais , Cromatina/genética , Drosophila melanogaster/genética , Regulação da Expressão Gênica/genética , Proteínas de Choque Térmico HSP70/genética , RNA Antissenso/genética , Transcrição Gênica
2.
J Biol Chem ; 297(1): 100901, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34157281

RESUMO

Alx1, a homeodomain-containing transcription factor, is a highly conserved regulator of skeletogenesis in echinoderms. In sea urchins, Alx1 plays a central role in the differentiation of embryonic primary mesenchyme cells (PMCs) and positively regulates the transcription of most biomineralization genes expressed by these cells. The alx1 gene arose via duplication and acquired a skeletogenic function distinct from its paralog (alx4) through the exonization of a 41-amino acid motif (the D2 domain). Alx1 and Alx4 contain glutamine-50 paired-type homeodomains, which interact preferentially with palindromic binding sites in vitro. Chromatin immunoprecipitation sequencing (ChIP-seq) studies have shown, however, that Alx1 binds both to palindromic and half sites in vivo. To address this apparent discrepancy and explore the function of the D2 domain, we used an endogenous cis-regulatory module associated with Sp-mtmmpb, a gene that encodes a PMC-specific metalloprotease, to analyze the DNA-binding properties of Alx1. We find that Alx1 forms dimeric complexes on TAAT-containing half sites by a mechanism distinct from the well-known mechanism of dimerization on palindromic sites. We used transgenic reporter assays to analyze the functional roles of half sites in vivo and demonstrate that two sites with partially redundant functions are essential for the PMC-specific activity of the Sp-mtmmpb cis-regulatory module. Finally, we show that the D2 domain influences the DNA-binding properties of Alx1 in vitro, suggesting that the exonization of this motif may have facilitated the acquisition of new transcriptional targets and consequently a novel developmental function.


Assuntos
Biomineralização , Equinodermos/genética , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição/metabolismo , Animais , Sítios de Ligação , Sequência Conservada , DNA/metabolismo , Equinodermos/metabolismo , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/genética , Ligação Proteica , Multimerização Proteica , Fatores de Transcrição/química , Fatores de Transcrição/genética
3.
Mol Cell ; 44(1): 51-61, 2011 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-21981918

RESUMO

The Drosophila ecdysone receptor (EcR/Usp) is thought to activate or repress gene transcription depending on the presence or absence, respectively, of the hormone ecdysone. Unexpectedly, we found an alternative mechanism at work in salivary glands during the ecdysone-dependent transition from larvae to pupae. In the absense of ecdysone, both ecdysone receptor subunits localize to the cytoplasm, and the heme-binding nuclear receptor E75A replaces EcR/Usp at common target sequences in several genes. During the larval-pupal transition, a switch from gene activation by EcR/Usp to gene repression by E75A is triggered by a decrease in ecdysone concentration and by direct repression of the EcR gene by E75A. Additional control is provided by developmentally timed modulation of E75A activity by NO, which inhibits recruitment of the corepressor SMRTER. These results suggest a mechanism for sequential modulation of gene expression during development by competing nuclear receptors and their effector molecules, ecdysone and NO.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Ecdisona/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Óxido Nítrico/metabolismo , Receptores de Esteroides/metabolismo , Fatores de Transcrição/metabolismo , Animais , Drosophila melanogaster , Inativação Gênica , Histona-Lisina N-Metiltransferase/metabolismo , Larva , Modelos Genéticos , Regiões Promotoras Genéticas , Pupa
4.
Bioessays ; 39(3)2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28133765

RESUMO

Chromosomes in multicellular animals are subdivided into a series of looped domains. In addition to being the underlying principle for organizing the chromatin fiber, looping is critical for processes ranging from gene regulation to recombination and repair. The subdivision of chromosomes into looped domains depends upon a special class of architectural elements called boundaries or insulators. These elements are distributed throughout the genome and are ubiquitous building blocks of chromosomes. In this review, we focus on features of boundaries that are critical in determining the topology of the looped domains and their genetic properties. We highlight the properties of fly boundaries that are likely to have an important bearing on the organization of looped domains in vertebrates, and discuss the functional consequences of the observed similarities and differences.


Assuntos
Cromossomos de Mamíferos/genética , Elementos Isolantes , Animais , Cromatina , Eucariotos/genética , Regulação da Expressão Gênica , Humanos , Sequências Repetidas Invertidas , Homologia de Sequência do Ácido Nucleico
5.
PLoS Genet ; 12(2): e1005889, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26910731

RESUMO

The chromosomes of multicellular animals are organized into a series of topologically independent looped domains. This domain organization is critical for the proper utilization and propagation of the genetic information encoded by the chromosome. A special set of architectural elements, called boundaries or insulators, are responsible both for subdividing the chromatin into discrete domains and for determining the topological organization of these domains. Central to the architectural functions of insulators are homologous and heterologous insulator:insulator pairing interactions. The former (pairing between copies of the same insulator) dictates the process of homolog alignment and pairing in trans, while the latter (pairing between different insulators) defines the topology of looped domains in cis. To elucidate the principles governing these architectural functions, we use two insulators, Homie and Nhomie, that flank the Drosophila even skipped locus. We show that homologous insulator interactions in trans, between Homie on one homolog and Homie on the other, or between Nhomie on one homolog and Nhomie on the other, mediate transvection. Critically, these homologous insulator:insulator interactions are orientation-dependent. Consistent with a role in the alignment and pairing of homologs, self-pairing in trans is head-to-head. Head-to-head self-interactions in cis have been reported for other fly insulators, suggesting that this is a general principle of self-pairing. Homie and Nhomie not only pair with themselves, but with each other. Heterologous Homie-Nhomie interactions occur in cis, and we show that they serve to delimit a looped chromosomal domain that contains the even skipped transcription unit and its associated enhancers. The topology of this loop is defined by the heterologous pairing properties of Homie and Nhomie. Instead of being head-to-head, which would generate a circular loop, Homie-Nhomie pairing is head-to-tail. Head-to-tail pairing in cis generates a stem-loop, a configuration much like that observed in classical lampbrush chromosomes. These pairing principles provide a mechanistic underpinning for the observed topologies within and between chromosomes.


Assuntos
Cromossomos de Insetos/química , Proteínas de Drosophila/genética , Proteínas de Homeodomínio/genética , Elementos Isolantes/genética , Fatores de Transcrição/genética , Animais , Animais Geneticamente Modificados , Cromossomos de Insetos/fisiologia , Drosophila melanogaster/embriologia , Drosophila melanogaster/genética , Embrião não Mamífero/fisiologia , Genes Reporter , Proteínas de Fluorescência Verde/genética , Óperon Lac
6.
Biochim Biophys Acta Gen Subj ; 1861(11 Pt A): 2789-2801, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28774855

RESUMO

BACKGROUND: The affinities of DNA binding proteins for target sites can be used to model the regulation of gene expression. These proteins can bind to DNA cooperatively, strongly impacting their affinity and specificity. However, current methods for measuring cooperativity do not provide the means to accurately predict binding behavior over a wide range of concentrations. METHODS: We use standard computational and mathematical methods, and develop novel methods as described in Results. RESULTS: We explore some complexities of cooperative binding, and develop an improved method for relating in vitro measurements to in vivo function, based on ternary complex formation. We derive expressions for the equilibria among the various complexes, and explore the limitations of binding experiments that model the system using a single parameter. We describe how to use single-ligand binding and ternary complex formation in tandem to determine parameters that have thermodynamic relevance. We develop an improved method for finding both single-ligand dissociation constants and concentrations simultaneously. We show how the cooperativity factor can be found when only one of the single-ligand dissociation constants can be measured. CONCLUSIONS: The methods that we develop constitute an optimized approach to accurately model cooperative binding. GENERAL SIGNIFICANCE: The expressions and methods we develop for modeling and analyzing DNA binding and cooperativity are applicable to most cases where multiple ligands bind to distinct sites on a common substrate. The parameters determined using these methods can be fed into models of higher-order cooperativity to increase their predictive power.


Assuntos
Proteínas de Ligação a DNA/química , Termodinâmica , Fatores de Transcrição/química , Regulação Alostérica/genética , Sítios de Ligação , Fenômenos Biofísicos , Biologia Computacional , Proteínas de Ligação a DNA/genética , Cinética , Ligantes , Modelos Teóricos , Ligação Proteica , Fatores de Transcrição/genética
7.
PLoS Genet ; 9(10): e1003883, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24204298

RESUMO

Insulators can block the action of enhancers on promoters and the spreading of repressive chromatin, as well as facilitating specific enhancer-promoter interactions. However, recent studies have called into question whether the activities ascribed to insulators in model transgene assays actually reflect their functions in the genome. The Drosophila even skipped (eve) gene is a Polycomb (Pc) domain with a Pc-group response element (PRE) at one end, flanked by an insulator, an arrangement also seen in other genes. Here, we show that this insulator has three major functions. It blocks the spreading of the eve Pc domain, preventing repression of the adjacent gene, TER94. It prevents activation of TER94 by eve regulatory DNA. It also facilitates normal eve expression. When Homie is deleted in the context of a large transgene that mimics both eve and TER94 regulation, TER94 is repressed. This repression depends on the eve PRE. Ubiquitous TER94 expression is "replaced" by expression in an eve pattern when Homie is deleted, and this effect is reversed when the PRE is also removed. Repression of TER94 is attributable to spreading of the eve Pc domain into the TER94 locus, accompanied by an increase in histone H3 trimethylation at lysine 27. Other PREs can functionally replace the eve PRE, and other insulators can block PRE-dependent repression in this context. The full activity of the eve promoter is also dependent on Homie, and other insulators can promote normal eve enhancer-promoter communication. Our data suggest that this is not due to preventing promoter competition, but is likely the result of the insulator organizing a chromosomal conformation favorable to normal enhancer-promoter interactions. Thus, insulator activities in a native context include enhancer blocking and enhancer-promoter facilitation, as well as preventing the spread of repressive chromatin.


Assuntos
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Proteínas de Homeodomínio/genética , Elementos Isolantes/genética , Proteínas do Grupo Polycomb/genética , Fatores de Transcrição/genética , Animais , Animais Geneticamente Modificados , Proteínas de Ciclo Celular/biossíntese , Proteínas de Ciclo Celular/genética , Cromatina/genética , Proteínas de Drosophila/biossíntese , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/biossíntese , Regiões Promotoras Genéticas , Elementos de Resposta/genética , Retroelementos/genética , Fatores de Transcrição/biossíntese , Proteína com Valosina
8.
Dev Biol ; 388(1): 117-33, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24512689

RESUMO

Hb9 is a homeodomain-containing transcription factor that acts in combination with Nkx6, Lim3, and Tail-up (Islet) to guide the stereotyped differentiation, connectivity, and function of a subset of neurons in Drosophila. The role of Hb9 in directing neuronal differentiation is well documented, but the lineage of Hb9(+) neurons is only partly characterized, its regulation is poorly understood, and most of the downstream genes through which it acts remain at large. Here, we complete the lineage tracing of all embryonic Hb9(+) neurons (to eight neuronal lineages) and provide evidence that hb9, lim3, and tail-up are coordinately regulated by a common set of upstream factors. Through the parallel use of micro-array gene expression profiling and the Dam-ID method, we searched for Hb9-regulated genes, uncovering transcription factors as the most over-represented class of genes regulated by Hb9 (and Nkx6) in the CNS. By a nearly ten-to-one ratio, Hb9 represses rather than activates transcription factors, highlighting transcriptional repression of other transcription factors as a core mechanism by which Hb9 governs neuronal determination. From the small set of genes activated by Hb9, we characterized the expression and function of two - fd59a/foxd, which encodes a transcription factor, and Nitric oxide synthase. Under standard lab conditions, both genes are dispensable for Drosophila development, but Nos appears to inhibit hyper-active behavior and fd59a appears to act in octopaminergic neurons to control egg-laying behavior. Together our data clarify the mechanisms through which Hb9 governs neuronal specification and differentiation and provide an initial characterization of the expression and function of Nos and fd59a in the Drosophila CNS.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição/metabolismo , Alelos , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Diferenciação Celular , Linhagem da Célula , Sistema Nervoso Central/embriologia , Elementos Facilitadores Genéticos , Fatores de Transcrição Forkhead/metabolismo , Estudos de Associação Genética , Genótipo , Hibridização In Situ , Dados de Sequência Molecular , Mutagênese , Neurônios/metabolismo , Óxido Nítrico Sintase/metabolismo , Regiões Promotoras Genéticas , Homologia de Sequência de Aminoácidos , Transcriptoma
9.
Dev Biol ; 362(2): 309-19, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22178246

RESUMO

In order to investigate regulation and redundancy within the sloppy paired (slp) locus, we analyzed 30 kilobases of DNA encompassing the tandem, coordinately regulated slp1 and slp2 transcription units. We found a remarkable array of stripe enhancers with overlapping activities surrounding the slp1 transcription unit, and, unexpectedly, glial cell enhancers surrounding slp2. The slp stripe regulatory region generates 7 stripes at blastoderm, and later 14 stripes that persist throughout embryogenesis. Phylogenetic analysis among drosophilids suggests that the multiplicity of stripe enhancers did not evolve through recent duplication. Most of the direct integration among cis-regulatory modules appears to be simply additive, with one notable exception. Despite the apparent redundancy among stripe enhancers, transgenic rescue suggests that most are required for full function, to maintain wingless expression and parasegment boundaries throughout embryogenesis. Transgenic rescue also reveals indirect positive autoregulation by the 7 early stripes, without which alternate stripes within the 14-stripe pattern are lost, leading to embryos with a pair-rule phenotype.


Assuntos
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Elementos Facilitadores Genéticos/genética , Evolução Molecular , Regulação da Expressão Gênica no Desenvolvimento/genética , Genes Duplicados/genética , Fatores de Transcrição/genética , Animais , Sequência de Bases , Biologia Computacional , Sequência Conservada/genética , Elementos Facilitadores Genéticos/fisiologia , Hibridização In Situ , Dados de Sequência Molecular , Neuroglia/metabolismo , Filogenia , Análise de Sequência de DNA
10.
Dev Biol ; 366(2): 382-92, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22537495

RESUMO

Even skipped (Eve) and Engrailed (En) are homeodomain-containing transcriptional repressors with similar DNA binding specificities that are sequentially expressed in Drosophila embryos. The sloppy-paired (slp) locus is a target of repression by both Eve and En. At blastoderm, Eve is expressed in 7 stripes that restrict the posterior border of slp stripes, allowing engrailed (en) gene expression to be initiated in odd-numbered parasegments. En, in turn, prevents expansion of slp stripes after Eve is turned off. Prior studies showed that the two tandem slp transcription units are regulated by cis-regulatory modules (CRMs) with activities that overlap in space and time. An array of CRMs that generate 7 stripes at blastoderm, and later 14 stripes, surround slp1 (Fujioka and Jaynes, 2012). Surprisingly given their similarity in DNA binding specificity and function, responsiveness to ectopic Eve and En indicates that most of their direct target sites are either in distinct CRMs, or in different parts of coregulated CRMs. We localized cooperative binding sites for En, with the homeodomain-containing Hox cofactors Extradenticle (Exd) and Homothorax (Hth), within two CRMs that drive similar expression patterns. Functional analysis revealed two distinct, redundant sites within one CRM. The other CRM contains a single cooperative site that is both necessary and sufficient for repression in the en domain. Correlating in vivo and in vitro analysis suggests that cooperativity with Exd and Hth is a key ingredient in the mechanism of En-dependent repression, and that apparent affinity in vitro is an unreliable predictor of in vivo function.


Assuntos
Sítios de Ligação/genética , Proteínas de Drosophila/genética , Drosophila/genética , Drosophila/metabolismo , Proteínas de Homeodomínio/genética , Fatores de Transcrição/genética , Animais , Drosophila/embriologia , Proteínas de Drosophila/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , Ligação Proteica , Sequências Reguladoras de Ácido Nucleico , Fatores de Transcrição/metabolismo
11.
Development ; 136(18): 3077-87, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19675129

RESUMO

Insulator sequences help to organize the genome into discrete functional regions by preventing inappropriate cross-regulation. This is thought to be mediated in part through associations with other insulators located elsewhere in the genome. Enhancers that normally drive Drosophila even skipped (eve) expression are located closer to the TER94 transcription start site than to that of eve. We discovered that the region between these genes has enhancer-blocking activity, and that this insulator region also mediates homing of P-element transgenes to the eve-TER94 genomic neighborhood. Localization of these activities to within 0.6 kb failed to separate them. Importantly, homed transgenic promoters respond to endogenous eve enhancers from great distances, and this long-range communication depends on the homing/insulator region, which we call Homie. We also find that the eve promoter contributes to long-distance communication. However, even the basal hsp70 promoter can communicate with eve enhancers across distances of several megabases, when the communication is mediated by Homie. These studies show that, while Homie blocks enhancer-promoter communication at short range, it facilitates long-range communication between distant genomic regions, possibly by organizing a large chromosomal loop between endogenous and transgenic Homies.


Assuntos
Cromatina/genética , Drosophila melanogaster/genética , Elementos Facilitadores Genéticos , Regiões Promotoras Genéticas , Transgenes , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteína com Valosina
12.
Cell Mol Gastroenterol Hepatol ; 13(4): 1276-1296, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34954189

RESUMO

BACKGROUND & AIMS: Sporadic colorectal cancers arise from initiating mutations in APC, producing oncogenic ß-catenin/TCF-dependent transcriptional reprogramming. Similarly, the tumor suppressor axis regulated by the intestinal epithelial receptor GUCY2C is among the earliest pathways silenced in tumorigenesis. Retention of the receptor, but loss of its paracrine ligands, guanylin and uroguanylin, is an evolutionarily conserved feature of colorectal tumors, arising in the earliest dysplastic lesions. Here, we examined a mechanism of GUCY2C ligand transcriptional silencing by ß-catenin/TCF signaling. METHODS: We performed RNA sequencing analysis of 4 unique conditional human colon cancer cell models of ß-catenin/TCF signaling to map the core Wnt-transcriptional program. We then performed a comparative analysis of orthogonal approaches, including luciferase reporters, chromatin immunoprecipitation sequencing, CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats) knockout, and CRISPR epigenome editing, which were cross-validated with human tissue chromatin immunoprecipitation sequencing datasets, to identify functional gene enhancers mediating GUCY2C ligand loss. RESULTS: RNA sequencing analyses reveal the GUCY2C hormones as 2 of the most sensitive targets of ß-catenin/TCF signaling, reflecting transcriptional repression. The GUCY2C hormones share an insulated genomic locus containing a novel locus control region upstream of the guanylin promoter that mediates the coordinated silencing of both genes. Targeting this region with CRISPR epigenome editing reconstituted GUCY2C ligand expression, overcoming gene inactivation by mutant ß-catenin/TCF signaling. CONCLUSIONS: These studies reveal DNA elements regulating corepression of GUCY2C ligand transcription by ß-catenin/TCF signaling, reflecting a novel pathophysiological step in tumorigenesis. They offer unique genomic strategies that could reestablish hormone expression in the context of canonical oncogenic mutations to reconstitute the GUCY2C axis and oppose transformation.


Assuntos
Neoplasias Colorretais , beta Catenina , Carcinogênese/genética , Cateninas/genética , Cateninas/metabolismo , Neoplasias Colorretais/patologia , Humanos , Ligantes , Região de Controle de Locus Gênico , Receptores de Enterotoxina/genética , Receptores de Enterotoxina/metabolismo , Fatores de Transcrição TCF/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
13.
Nature ; 426(6962): 78-83, 2003 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-14603321

RESUMO

Steroid hormones fulfil important functions in animal development. In Drosophila, ecdysone triggers moulting and metamorphosis through its effects on gene expression. Ecdysone works by binding to a nuclear receptor, EcR, which heterodimerizes with the retinoid X receptor homologue Ultraspiracle. Both partners are required for binding to ligand or DNA. Like most DNA-binding transcription factors, nuclear receptors activate or repress gene expression by recruiting co-regulators, some of which function as chromatin-modifying complexes. For example, p160 class coactivators associate with histone acetyltransferases and arginine histone methyltransferases. The Trithorax-related gene of Drosophila encodes the SET domain protein TRR. Here we report that TRR is a histone methyltransferases capable of trimethylating lysine 4 of histone H3 (H3-K4). trr acts upstream of hedgehog (hh) in progression of the morphogenetic furrow, and is required for retinal differentiation. Mutations in trr interact in eye development with EcR, and EcR and TRR can be co-immunoprecipitated on ecdysone treatment. TRR, EcR and trimethylated H3-K4 are detected at the ecdysone-inducible promoters of hh and BR-C in cultured cells, and H3-K4 trimethylation at these promoters is decreased in embryos lacking a functional copy of trr. We propose that TRR functions as a coactivator of EcR by altering the chromatin structure at ecdysone-responsive promoters.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila/efeitos dos fármacos , Drosophila/embriologia , Ecdisona/farmacologia , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Animais , Montagem e Desmontagem da Cromatina/efeitos dos fármacos , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Olho/embriologia , Olho/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Proteínas Hedgehog , Histona-Lisina N-Metiltransferase/genética , Masculino , Metilação/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Ligação Proteica , Receptores de Esteroides/metabolismo
14.
Genetics ; 216(3): 689-700, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32948625

RESUMO

Polycomb group (PcG) proteins are an important group of transcriptional repressors that act by modifying chromatin. PcG target genes are covered by the repressive chromatin mark H3K27me3. Polycomb repressive complex 2 (PRC2) is a multiprotein complex that is responsible for generating H3K27me3. In Drosophila, PRC2 is recruited by Polycomb Response Elements (PREs) and then trimethylates flanking nucleosomes, spreading the H3K27me3 mark over large regions of the genome, the "Polycomb domains." What defines the boundary of a Polycomb domain? There is experimental evidence that insulators, PolII, and active transcription can all form the boundaries of Polycomb domains. Here we divide the boundaries of larval Polycomb domains into six different categories. In one category, genes are transcribed toward the Polycomb domain, where active transcription is thought to stop the spreading of H3K27me3. In agreement with this, we show that introducing a transcriptional terminator into such a transcription unit causes an extension of the Polycomb domain. Additional data suggest that active transcription of a boundary gene may restrict the range of enhancer activity of a Polycomb-regulated gene.


Assuntos
Histonas/metabolismo , Complexo Repressor Polycomb 2/metabolismo , Animais , Drosophila melanogaster , Elementos Facilitadores Genéticos , Elementos Isolantes
15.
Nat Genet ; 50(9): 1296-1303, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30038397

RESUMO

A long-standing question in gene regulation is how remote enhancers communicate with their target promoters, and specifically how chromatin topology dynamically relates to gene activation. Here, we combine genome editing and multi-color live imaging to simultaneously visualize physical enhancer-promoter interaction and transcription at the single-cell level in Drosophila embryos. By examining transcriptional activation of a reporter by the endogenous even-skipped enhancers, which are located 150 kb away, we identify three distinct topological conformation states and measure their transition kinetics. We show that sustained proximity of the enhancer to its target is required for activation. Transcription in turn affects the three-dimensional topology as it enhances the temporal stability of the proximal conformation and is associated with further spatial compaction. Furthermore, the facilitated long-range activation results in transcriptional competition at the locus, causing corresponding developmental defects. Our approach offers quantitative insight into the spatial and temporal determinants of long-range gene regulation and their implications for cellular fates.


Assuntos
Elementos Facilitadores Genéticos , Regiões Promotoras Genéticas , Ativação Transcricional , Animais , Cromatina/genética , Drosophila/genética , Feminino , Edição de Genes/métodos , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Masculino , Transcrição Gênica
16.
Curr Biol ; 12(2): 125-30, 2002 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-11818063

RESUMO

Inhibitors of apoptosis proteins (IAPs) interact with caspases and inhibit their protease activity, whereas the IAP-inhibitory proteins Smac/DIABLO in mammals and Reaper, Hid, and Grim in flies relieve IAP-mediated inhibition to induce cell death. Here we describe the functional characterization of the novel Drosophila cell death protein Sickle (Skl), which binds to IAPs and neutralizes their apoptotic inhibitory activity. Skl exhibits no sequence homology to Reaper, Hid, Grim, or Smac/DIABLO, except within the 4 residue N-terminal IAP binding motif. Skl interacts with Drosophila and mammalian IAPs and can promote caspase activation in the presence of IAPs. Consistent with these findings, expression of Skl in Drosophila and mammalian cell lines or in Drosophila embryos induces apoptosis. Skl can also synergize with Grim to induce cell death in the Drosophila eye imaginal disc. Based on biochemical and structural data, the N terminus of Skl, like that of the mammalian Smac/DIABLO, is absolutely required for its apoptotic and caspase-promoting activities and its ability to interact with IAPs. These findings point to conservation in the structure and function of the IAP-inhibitory proteins across species and suggest the existence of other family members.


Assuntos
Apoptose/genética , Proteínas de Drosophila/genética , Drosophila/genética , Proteínas de Insetos/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Células Cultivadas , Clonagem Molecular , Drosophila/embriologia , Proteínas de Drosophila/química , Proteínas de Drosophila/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Insetos/genética , Modelos Moleculares , Dados de Sequência Molecular , Neuropeptídeos/genética , Peptídeos/genética , Ligação Proteica , Conformação Proteica
17.
PLoS Biol ; 1(2): E41, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14624243

RESUMO

The organisational principles of locomotor networks are less well understood than those of many sensory systems, where in-growing axon terminals form a central map of peripheral characteristics. Using the neuromuscular system of the Drosophila embryo as a model and retrograde tracing and genetic methods, we have uncovered principles underlying the organisation of the motor system. We find that dendritic arbors of motor neurons, rather than their cell bodies, are partitioned into domains to form a myotopic map, which represents centrally the distribution of body wall muscles peripherally. While muscles are segmental, the myotopic map is parasegmental in organisation. It forms by an active process of dendritic growth independent of the presence of target muscles, proper differentiation of glial cells, or (in its initial partitioning) competitive interactions between adjacent dendritic domains. The arrangement of motor neuron dendrites into a myotopic map represents a first layer of organisation in the motor system. This is likely to be mirrored, at least in part, by endings of higher-order neurons from central pattern-generating circuits, which converge onto the motor neuron dendrites. These findings will greatly simplify the task of understanding how a locomotor system is assembled. Our results suggest that the cues that organise the myotopic map may be laid down early in development as the embryo subdivides into parasegmental units.


Assuntos
Dendritos/metabolismo , Drosophila melanogaster/anatomia & histologia , Drosophila melanogaster/fisiologia , Embrião não Mamífero/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Neurônios Motores/fisiologia , Músculos/embriologia , Animais , Animais Geneticamente Modificados , Padronização Corporal , Diferenciação Celular , Sistema Nervoso Central/patologia , Cruzamentos Genéticos , Proteínas de Drosophila/metabolismo , Imuno-Histoquímica , Modelos Anatômicos , Modelos Biológicos , Neurônios Motores/metabolismo , Músculos/patologia , Neurônios/metabolismo , Neurônios Aferentes/patologia , Plasmídeos/metabolismo , Terminações Pré-Sinápticas , Fatores de Tempo , Transgenes
18.
Circ Res ; 97(11): 1108-14, 2005 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-16239588

RESUMO

The Drosophila pair-rule gene even skipped (eve) is required for embryonic segmentation and later in specific cell lineages in both the nervous system and the mesoderm. We previously generated eve mesoderm-specific mutants by combining an eve null mutant with a rescuing transgene that includes the entire locus, but with the mesodermal enhancer removed. This allowed us to analyze in detail the defects that result from a precisely targeted elimination of mesodermal eve expression in the context of an otherwise normal embryo. Absence of mesodermal eve causes a highly selective loss of the entire eve-expressing lineage in this germ layer, including those progeny that do not continue to express eve, suggesting that mesodermal eve precursor specification is not implemented. Despite the resulting absence of a subset of muscles and pericardial cells, mesoderm-specific eve mutants survive to fertile adulthood, providing an opportunity to examine the effects of these developmental abnormalities on adult fitness and heart function. We find that in these mutants, flying ability, myocardial performance under normal and stressed conditions, and lifespan are severely reduced. These data imply a nonautonomous role of the affected pericardial cells and body wall muscles in developing and/or maintaining cardiac performance and possibly other functions contributing to normal lifespan. Given the similarities of molecular-genetic control between Drosophila and vertebrates, these findings suggest that peri/epicardial influences may well be important for proper myocardial function.


Assuntos
Proteínas de Drosophila/fisiologia , Coração/embriologia , Proteínas de Homeodomínio/fisiologia , Músculos/embriologia , Fatores de Transcrição/fisiologia , Envelhecimento , Animais , Drosophila , Regulação da Expressão Gênica no Desenvolvimento , Coração/fisiologia , Mesoderma/fisiologia , Mutação , Pericárdio/embriologia
19.
Genetics ; 160(4): 1561-71, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11973310

RESUMO

Regulatory DNA from the Drosophila gene engrailed causes silencing of a linked reporter gene (mini-white) in transgenic Drosophila. This silencing is strengthened in flies homozygous for the transgene and has been called "pairing-sensitive silencing." The pairing-sensitive silencing activities of a large fragment (2.6 kb) and a small subfragment (181 bp) were explored. Since pairing-sensitive silencing is often associated with Polycomb group response elements (PREs), we tested the activities of each of these engrailed fragments in a construct designed to detect PRE activity in embryos. Both fragments were found to behave as PREs in a bxd-Ubx-lacZ reporter construct, while the larger fragment showed additional silencing capabilities. Using the mini-white reporter gene, a 139-bp minimal pairing-sensitive element (PSE) was defined. DNA mobility-shift assays using Drosophila nuclear extracts suggested that there are eight protein-binding sites within this 139-bp element. Mutational analysis showed that at least five of these sites are important for pairing-sensitive silencing. One of the required sites is for the Polycomb group protein Pleiohomeotic and another is GAGAG, a sequence bound by the proteins GAGA factor and Pipsqueak. The identity of the other proteins is unknown. These data suggest a surprising degree of complexity in the DNA-binding proteins required for PSE function.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Inativação Gênica/fisiologia , Proteínas de Homeodomínio/genética , Elementos de Resposta , Fatores de Transcrição/genética , Animais , Animais Geneticamente Modificados , Sequência de Bases , Sítios de Ligação , Análise Mutacional de DNA , Drosophila/embriologia , Drosophila/genética , Proteínas de Drosophila , Regulação da Expressão Gênica , Genes Reporter , Dados de Sequência Molecular , Fenótipo , Análise de Sequência de DNA
20.
Development ; 135(24): 4131-9, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19029043

RESUMO

Although epigenetic maintenance of either the active or repressed transcriptional state often involves overlapping regulatory elements, the underlying basis of this is not known. Epigenetic and pairing-sensitive silencing are related properties of Polycomb-group proteins, whereas their activities are generally opposed by the trithorax group. Both groups modify chromatin structure, but how their opposing activities are targeted to allow differential maintenance remains a mystery. Here, we identify a strong pairing-sensitive silencing (PSS) element at the 3' border of the Drosophila even skipped (eve) locus. This element can maintain repression during embryonic as well as adult eye development. Transgenic dissection revealed that silencing activity depends on a binding site for the Polycomb-group protein Pleiohomeotic (Pho) and on pho gene function. Binding sites for the trithorax-group protein GAGA factor also contribute, whereas sites for the known Polycomb response element binding factors Zeste and Dsp1 are dispensible. Normally, eve expression in the nervous system is maintained throughout larval stages. An enhancer that functions fully in embryos does not maintain expression, but the adjacent PSS element confers maintenance. This positive activity also depends on pho gene activity and on Pho binding. Thus, a DNA-binding complex requiring Pho is differentially regulated to facilitate epigenetic transcriptional memory of both the active and the repressed state.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila/genética , Drosophila/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Animais Geneticamente Modificados , Sequência de Bases , Sítios de Ligação/genética , Sistema Nervoso Central/embriologia , Sistema Nervoso Central/crescimento & desenvolvimento , Sistema Nervoso Central/metabolismo , DNA/genética , DNA/metabolismo , Drosophila/embriologia , Drosophila/crescimento & desenvolvimento , Epigênese Genética , Olho/embriologia , Olho/crescimento & desenvolvimento , Olho/metabolismo , Inativação Gênica , Genes de Insetos , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Dados de Sequência Molecular , Proteínas do Grupo Polycomb
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa