Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cell ; 186(18): 3810-3825.e18, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37552983

ABSTRACT

A ubiquitous feature of eukaryotic transcriptional regulation is cooperative self-assembly between transcription factors (TFs) and DNA cis-regulatory motifs. It is thought that this strategy enables specific regulatory connections to be formed in gene networks between otherwise weakly interacting, low-specificity molecular components. Here, using synthetic gene circuits constructed in yeast, we find that high regulatory specificity can emerge from cooperative, multivalent interactions among artificial zinc-finger-based TFs. We show that circuits "wired" using the strategy of cooperative TF assembly are effectively insulated from aberrant misregulation of the host cell genome. As we demonstrate in experiments and mathematical models, this mechanism is sufficient to rescue circuit-driven fitness defects, resulting in genetic and functional stability of circuits in long-term continuous culture. Our naturally inspired approach offers a simple, generalizable means for building high-fidelity, evolutionarily robust gene circuits that can be scaled to a wide range of host organisms and applications.


Subject(s)
Gene Regulatory Networks , Transcription Factors , Transcription Factors/genetics , Saccharomyces cerevisiae/genetics , Genome
2.
G3 (Bethesda) ; 10(12): 4473-4482, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33037064

ABSTRACT

Enhancers are DNA sequences composed of transcription factor binding sites that drive complex patterns of gene expression in space and time. Until recently, studying enhancers in their genomic context was technically challenging. Therefore, minimal enhancers, the shortest pieces of DNA that can drive an expression pattern that resembles a gene's endogenous pattern, are often used to study features of enhancer function. However, evidence suggests that some enhancers require sequences outside the minimal enhancer to maintain function under environmental perturbations. We hypothesized that these additional sequences also prevent misexpression caused by a transcription factor binding site mutation within a minimal enhancer. Using the Drosophila melanogastereven-skipped stripe 2 enhancer as a case study, we tested the effect of a Giant binding site mutation (gt-2) on the expression patterns driven by minimal and extended enhancer reporter constructs. We found that, in contrast to the misexpression caused by the gt-2 binding site deletion in the minimal enhancer, the same gt-2 binding site deletion in the extended enhancer did not have an effect on expression. The buffering of expression levels, but not expression pattern, is partially explained by an additional Giant binding site outside the minimal enhancer. Deleting the gt-2 binding site in the endogenous locus had no significant effect on stripe 2 expression. Our results indicate that rules derived from mutating enhancer reporter constructs may not represent what occurs in the endogenous context.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mutation , Transcription Factors/genetics , Transcription Factors/metabolism
3.
G3 (Bethesda) ; 9(7): 2171-2182, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31048401

ABSTRACT

Complex spatiotemporal gene expression patterns direct the development of the fertilized egg into an adult animal. Comparisons across species show that, in spite of changes in the underlying regulatory DNA sequence, developmental programs can be maintained across millions of years of evolution. Reciprocally, changes in gene expression can be used to generate morphological novelty. Distinguishing between changes in regulatory DNA that lead to changes in gene expression and those that do not is therefore a central goal of evolutionary developmental biology. Quantitative, spatially-resolved measurements of developmental gene expression patterns play a crucial role in this goal, enabling the detection of subtle phenotypic differences between species and the development of computations models that link the sequence of regulatory DNA to expression patterns. Here we report the generation of two atlases of cellular resolution gene expression measurements for the primary anterior-posterior patterning genes in Drosophila simulans and Drosophila virilis By combining these data sets with existing atlases for three other Drosophila species, we detect subtle differences in the gene expression patterns and dynamics driving the highly conserved axis patterning system and delineate inter-species differences in the embryonic morphology. These data sets will be a resource for future modeling studies of the evolution of developmental gene regulatory networks.


Subject(s)
Body Patterning , Drosophila/embryology , Embryonic Development , Animals , Biomarkers , Body Patterning/genetics , Embryo, Nonmammalian , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Organ Specificity , Species Specificity , Transcriptome
4.
PLoS Genet ; 14(9): e1007644, 2018 09.
Article in English | MEDLINE | ID: mdl-30192762

ABSTRACT

Hunchback is a bifunctional transcription factor that can activate and repress gene expression in Drosophila development. We investigated the regulatory DNA sequence features that control Hunchback function by perturbing enhancers for one of its target genes, even-skipped (eve). While Hunchback directly represses the eve stripe 3+7 enhancer, we found that in the eve stripe 2+7 enhancer, Hunchback repression is prevented by nearby sequences-this phenomenon is called counter-repression. We also found evidence that Caudal binding sites are responsible for counter-repression, and that this interaction may be a conserved feature of eve stripe 2 enhancers. Our results alter the textbook view of eve stripe 2 regulation wherein Hb is described as a direct activator. Instead, to generate stripe 2, Hunchback repression must be counteracted. We discuss how counter-repression may influence eve stripe 2 regulation and evolution.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Animals, Genetically Modified , Binding Sites/genetics , DNA-Binding Proteins/genetics , Drosophila melanogaster/growth & development , Embryo, Nonmammalian , Enhancer Elements, Genetic/genetics , Female , Homeodomain Proteins/metabolism , Male
6.
Cell Rep ; 12(11): 1740-7, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26344774

ABSTRACT

Many developmental genes are controlled by shadow enhancers­pairs of enhancers that drive overlapping expression patterns. We hypothesized that compensatory evolution can maintain the total expression of a gene, while individual shadow enhancers diverge between species. To test this hypothesis, we analyzed expression driven by orthologous pairs of shadow enhancers from Drosophila melanogaster, Drosophila yakuba, and Drosophila pseudoobscura that control expression of Krüppel, a transcription factor that patterns the anterior-posterior axis of blastoderm embryos. We found that the expression driven by the pair of enhancers is conserved between these three species, but expression levels driven by the individual enhancers are not. Using sequence analysis and experimental perturbation, we show that each shadow enhancer is regulated by different transcription factors. These results support the hypothesis that compensatory evolution can occur between shadow enhancers, which has implications for mechanistic and evolutionary studies of gene regulation.


Subject(s)
Drosophila Proteins/biosynthesis , Drosophila Proteins/genetics , Drosophila/genetics , Drosophila/metabolism , Enhancer Elements, Genetic , Kruppel-Like Transcription Factors/biosynthesis , Kruppel-Like Transcription Factors/genetics , Animals , Female , Gene Expression Regulation, Developmental , Male
7.
Mol Cell ; 58(5): 718-21, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-26046646

ABSTRACT

The National Institutes of Health (NIH) encourages trainees to make Individualized Development Plans to help them prepare for academic and nonacademic careers. We describe our approach to building an Individualized Development Plan, the reasons we find them useful and empowering for both PIs and trainees, and resources to help other labs implement them constructively.


Subject(s)
Biomedical Research/organization & administration , National Institutes of Health (U.S.) , Goals , Group Processes , Humans , Motivation , Personnel Management , United States
8.
Proc Natl Acad Sci U S A ; 112(3): 785-90, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25564665

ABSTRACT

Hunchback (Hb) is a bifunctional transcription factor that activates and represses distinct enhancers. Here, we investigate the hypothesis that Hb can activate and repress the same enhancer. Computational models predicted that Hb bifunctionally regulates the even-skipped (eve) stripe 3+7 enhancer (eve3+7) in Drosophila blastoderm embryos. We measured and modeled eve expression at cellular resolution under multiple genetic perturbations and found that the eve3+7 enhancer could not explain endogenous eve stripe 7 behavior. Instead, we found that eve stripe 7 is controlled by two enhancers: the canonical eve3+7 and a sequence encompassing the minimal eve stripe 2 enhancer (eve2+7). Hb bifunctionally regulates eve stripe 7, but it executes these two activities on different pieces of regulatory DNA--it activates the eve2+7 enhancer and represses the eve3+7 enhancer. These two "shadow enhancers" use different regulatory logic to create the same pattern.


Subject(s)
DNA-Binding Proteins/physiology , Drosophila Proteins/physiology , Drosophila/embryology , Enhancer Elements, Genetic , Transcription Factors/physiology , Animals , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Transcription Factors/genetics
9.
Development ; 142(3): 587-96, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25605785

ABSTRACT

In developing embryos, gene regulatory networks drive cells towards discrete terminal fates, a process called canalization. We studied the behavior of the anterior-posterior segmentation network in Drosophila melanogaster embryos by depleting a key maternal input, bicoid (bcd), and measuring gene expression patterns of the network at cellular resolution. This method results in a gene expression atlas containing the levels of mRNA or protein expression of 13 core patterning genes over six time points for every cell of the blastoderm embryo. This is the first cellular resolution dataset of a genetically perturbed Drosophila embryo that captures all cells in 3D. We describe the technical developments required to build this atlas and how the method can be employed and extended by others. We also analyze this novel dataset to characterize the degree and timing of cell fate canalization in the segmentation network. We find that in two layers of this gene regulatory network, following depletion of bcd, individual cells rapidly canalize towards normal cell fates. This result supports the hypothesis that the segmentation network directly canalizes cell fate, rather than an alternative hypothesis whereby cells are initially mis-specified and later eliminated by apoptosis. Our gene expression atlas provides a high resolution picture of a classic perturbation and will enable further computational modeling of canalization and gene regulation in this transcriptional network.


Subject(s)
Body Patterning/genetics , Cell Lineage/genetics , Databases, Genetic , Drosophila melanogaster/embryology , Gene Regulatory Networks/genetics , Transcriptome/genetics , Animals , Drosophila Proteins , Homeodomain Proteins , In Situ Hybridization , RNA Interference , Real-Time Polymerase Chain Reaction , Trans-Activators/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...