Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Curr Biol ; 24(12): 1341-1346, 2014 Jun 16.
Article in English | MEDLINE | ID: mdl-24909324

ABSTRACT

Zygotic genome activation (ZGA) is a major genome programming event whereby the cells of the embryo begin to adopt specified fates. Experiments in Drosophila and zebrafish have revealed that ZGA depends on transcription factors that provide large-scale control of gene expression by direct and specific binding to gene regulatory sequences. Zelda (Zld) plays such a role in the Drosophila embryo, where it has been shown to control the action of patterning signals; however, the mechanisms underlying this effect remain largely unclear. A recent model proposed that Zld binding sites act as quantitative regulators of the spatiotemporal expression of genes activated by Dorsal (Dl), the morphogen that patterns the dorsoventral axis. Here we tested this model experimentally, using enhancers of brinker (brk) and short gastrulation (sog), both of which are directly activated by Dl, but at different concentration thresholds. In agreement with the model, we show that there is a clear positive correlation between the number of Zld binding sites and the spatial domain of enhancer activity. Likewise, the timing of expression could be advanced or delayed. We present evidence that Zld facilitates binding of Dl to regulatory DNA, and that this is associated with increased chromatin accessibility. Importantly, the change in chromatin accessibility is strongly correlated with the change in Zld binding, but not Dl. We propose that the ability of genome activators to facilitate readout of transcriptional input is key to widespread transcriptional induction during ZGA.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Nuclear Proteins/genetics , Phosphoproteins/genetics , Transcription Factors/genetics , Animals , Chromatin/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Embryo, Nonmammalian/metabolism , Embryonic Development , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Polymerase Chain Reaction , Transcription Factors/metabolism
2.
PLoS Genet ; 7(10): e1002339, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22028675

ABSTRACT

In past years, much attention has focused on the gene networks that regulate early developmental processes, but less attention has been paid to how multiple networks and processes are temporally coordinated. Recently the discovery of the transcriptional activator Zelda (Zld), which binds to CAGGTAG and related sequences present in the enhancers of many early-activated genes in Drosophila, hinted at a mechanism for how batteries of genes could be simultaneously activated. Here we use genome-wide binding and expression assays to identify Zld target genes in the early embryo with the goal of unraveling the gene circuitry regulated by Zld. We found that Zld binds to genes involved in early developmental processes such as cellularization, sex determination, neurogenesis, and pattern formation. In the absence of Zld, many target genes failed to be activated, while others, particularly the patterning genes, exhibited delayed transcriptional activation, some of which also showed weak and/or sporadic expression. These effects disrupted the normal sequence of patterning-gene interactions and resulted in highly altered spatial expression patterns, demonstrating the significance of a timing mechanism in early development. In addition, we observed prevalent overlap between Zld-bound regions and genomic "hotspot" regions, which are bound by many developmental transcription factors, especially the patterning factors. This, along with the finding that the most over-represented motif in hotspots, CAGGTA, is the Zld binding site, implicates Zld in promoting hotspot formation. We propose that Zld promotes timely and robust transcriptional activation of early-gene networks so that developmental events are coordinated and cell fates are established properly in the cellular blastoderm embryo.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Embryonic Development/genetics , Gene Regulatory Networks , Transcription Factors/genetics , Transcriptional Activation/genetics , Animals , Binding Sites/genetics , Blastoderm/embryology , Blastoderm/growth & development , Body Patterning/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental , Neurogenesis/genetics , Nuclear Proteins , Nucleotide Motifs/genetics , Promoter Regions, Genetic , Protein Binding/genetics , Sex Determination Processes/genetics , Transcription Factors/metabolism , Zygote/growth & development
3.
Nature ; 456(7220): 400-3, 2008 Nov 20.
Article in English | MEDLINE | ID: mdl-18931655

ABSTRACT

In all animals, the initial events of embryogenesis are controlled by maternal gene products that are deposited into the developing oocyte. At some point after fertilization, control of embryogenesis is transferred to the zygotic genome in a process called the maternal-to-zygotic transition. During this time, many maternal RNAs are degraded and transcription of zygotic RNAs ensues. There is a long-standing question as to which factors regulate these events. The recent findings that microRNAs and Smaug mediate maternal transcript degradation have shed new light on this aspect of the problem. However, the transcription factor(s) that activate the zygotic genome remain elusive. The discovery that many of the early transcribed genes in Drosophila share a cis-regulatory heptamer motif, CAGGTAG and related sequences, collectively referred to as TAGteam sites raised the possibility that a dedicated transcription factor could interact with these sites to activate transcription. Here we report that the zinc-finger protein Zelda (Zld; Zinc-finger early Drosophila activator) binds specifically to these sites and is capable of activating transcription in transient transfection assays. Mutant embryos lacking zld are defective in cellular blastoderm formation, and fail to activate many genes essential for cellularization, sex determination and pattern formation. Global expression profiling confirmed that Zld has an important role in the activation of the early zygotic genome and suggests that Zld may also regulate maternal RNA degradation during the maternal-to-zygotic transition.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Genome, Insect/genetics , Transcription Factors/metabolism , Zinc Fingers , Zygote/metabolism , Animals , Blastoderm/cytology , Blastoderm/embryology , Blastoderm/metabolism , Body Patterning/genetics , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Female , Gene Deletion , Gene Expression Profiling , Male , Nuclear Proteins , RNA Stability , RNA, Messenger, Stored/genetics , RNA, Messenger, Stored/metabolism , Sex Determination Processes , Transcription Factors/deficiency , Transcription Factors/genetics , Transcriptional Activation , Zygote/cytology , Zygote/growth & development
4.
Development ; 133(24): 4805-13, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17092951

ABSTRACT

Morphogen gradients determine a range of cell fates by specifying multiple transcriptional threshold responses. In the dorsal ectoderm of the Drosophila embryo, a BMP gradient is translated into an activated Smad transcription factor gradient, which elicits at least three threshold responses - high, intermediate and low. However, the mechanism underlying differential response to Dpp is poorly understood, due in part to the insufficient number of well-studied target genes. We analyzed the regulation of the C15 gene, which can be activated in cells containing intermediate levels of Dpp. We show that C15 expression requires both dpp and zen, thus forming a genetic feed-forward loop. The C15 regulatory element contains clusters of Smad- and Zen-binding sites in close proximity. Mutational analysis shows that the number of intact Smad- and Zen-binding sites is essential for the C15 transcriptional response, and that the spatial limits of C15 expression are established through a repression mechanism in the dorsolateral cells of the embryo. Thus, the combinatorial action of Smad and Zen activators bound to a number of adjacent sites, and competing negative cues allows for proper gene response to lower than peak levels of the Dpp morphogen.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila/embryology , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Smad Proteins/metabolism , Animals , Base Sequence , Binding Sites , Blastoderm/metabolism , Drosophila/genetics , Drosophila/metabolism , Enhancer Elements, Genetic , Introns , Molecular Sequence Data , Protein Structure, Tertiary , Regulatory Elements, Transcriptional , Transcriptional Activation
5.
Development ; 132(7): 1637-47, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15728670

ABSTRACT

Gradients of morphogens determine cell fates by specifying discrete thresholds of gene activities. In the Drosophila embryo, a BMP gradient subdivides the dorsal ectoderm into amnioserosa and dorsal epidermis, and also inhibits neuroectoderm formation. A number of genes are differentially expressed in response to the gradient, but how their borders of expression are established is not well understood. We present evidence that the BMP gradient, via the Smads, provides a two-fold input in regulating the amnioserosa-specific target genes such as Race. Peak levels of Smads in the presumptive amnioserosa set the expression domain of zen, and then Smads act in combination with Zen to directly activate Race. This situation resembles a feed-forward mechanism of transcriptional regulation. In addition, we demonstrate that ectopically expressed Zen can activate targets like Race in the presence of low level Smads, indicating that the role of the highest activity of the BMP gradient is to activate zen.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Drosophila/genetics , Gene Expression Regulation, Developmental/physiology , Peptidyl-Dipeptidase A/genetics , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Peptidyl-Dipeptidase A/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Smad Proteins , Trans-Activators/genetics , Trans-Activators/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...