Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Development ; 150(1)2023 01 01.
Article in English | MEDLINE | ID: mdl-36645371

ABSTRACT

Developing Future Biologists (DFB) is an inclusive, trainee-run organization that strives to excite and engage the next generation of biologists, regardless of race, gender or socioeconomic status, in the field of developmental biology. DFB offers a week-long course consisting of active lectures, hands-on laboratory sessions, and professional development opportunities through interactions with scientists from a variety of backgrounds and careers. A major goal of DFB is to propel undergraduate students from underserved communities to pursue biomedical research opportunities and advanced degrees in science. To achieve this goal, we provide DFB participants with continuing access to a diverse network of scientists that students can utilize to secure opportunities and foster success throughout multiple stages of their research careers. Here, we describe the flourishing DFB program at the University of Michigan to encourage other institutions to create their own DFB programs.


Subject(s)
Developmental Biology , Students , Humans
4.
Elife ; 52016 05 05.
Article in English | MEDLINE | ID: mdl-27146892

ABSTRACT

The Hedgehog signaling pathway is part of the ancient developmental-evolutionary animal toolkit. Frequently co-opted to pattern new structures, the pathway is conserved among eumetazoans yet flexible and pleiotropic in its effects. The Hedgehog receptor, Patched, is transcriptionally activated by Hedgehog, providing essential negative feedback in all tissues. Our locus-wide dissections of the cis-regulatory landscapes of fly patched and mouse Ptch1 reveal abundant, diverse enhancers with stage- and tissue-specific expression patterns. The seemingly simple, constitutive Hedgehog response of patched/Ptch1 is driven by a complex regulatory architecture, with batteries of context-specific enhancers engaged in promoter-specific interactions to tune signaling individually in each tissue, without disturbing patterning elsewhere. This structure-one of the oldest cis-regulatory features discovered in animal genomes-explains how patched/Ptch1 can drive dramatic adaptations in animal morphology while maintaining its essential core function. It may also suggest a general model for the evolutionary flexibility of conserved regulators and pathways.


Subject(s)
Gene Expression Regulation, Developmental , Hedgehog Proteins/metabolism , Patched-1 Receptor/metabolism , Signal Transduction , Animals , Cell Line , Drosophila , Mice
5.
PLoS One ; 10(12): e0145225, 2015.
Article in English | MEDLINE | ID: mdl-26710299

ABSTRACT

The Hedgehog (Hh) signaling pathway directs a multitude of cellular responses during embryogenesis and adult tissue homeostasis. Stimulation of the pathway results in activation of Hh target genes by the transcription factor Ci/Gli, which binds to specific motifs in genomic enhancers. In Drosophila, only a few enhancers (patched, decapentaplegic, wingless, stripe, knot, hairy, orthodenticle) have been shown by in vivo functional assays to depend on direct Ci/Gli regulation. All but one (orthodenticle) contain more than one Ci/Gli site, prompting us to directly test whether homotypic clustering of Ci/Gli binding sites is sufficient to define a Hh-regulated enhancer. We therefore developed a computational algorithm to identify Ci/Gli clusters that are enriched over random expectation, within a given region of the genome. Candidate genomic regions containing Ci/Gli clusters were functionally tested in chicken neural tube electroporation assays and in transgenic flies. Of the 22 Ci/Gli clusters tested, seven novel enhancers (and the previously known patched enhancer) were identified as Hh-responsive and Ci/Gli-dependent in one or both of these assays, including: Cuticular protein 100A (Cpr100A); invected (inv), which encodes an engrailed-related transcription factor expressed at the anterior/posterior wing disc boundary; roadkill (rdx), the fly homolog of vertebrate Spop; the segment polarity gene gooseberry (gsb); and two previously untested regions of the Hh receptor-encoding patched (ptc) gene. We conclude that homotypic Ci/Gli clustering is not sufficient information to ensure Hh-responsiveness; however, it can provide a clue for enhancer recognition within putative Hedgehog target gene loci.


Subject(s)
Computational Biology/methods , DNA-Binding Proteins/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Hedgehog Proteins/genetics , Oncogene Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , Binding Sites/genetics , Chick Embryo , Drosophila melanogaster/genetics , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/genetics , Hedgehog Proteins/metabolism , Neural Tube/metabolism , Regulatory Sequences, Nucleic Acid , Sequence Analysis, DNA , Signal Transduction/genetics , Wings, Animal/embryology , Zinc Finger Protein GLI1
6.
Curr Biol ; 25(7): R290-3, 2015 Mar 30.
Article in English | MEDLINE | ID: mdl-25829016

ABSTRACT

Some transcriptional enhancers work best with one type of promoter, while ignoring others. How widespread is such specificity across the genome? A new study finds that, in a fair fight, most enhancers prefer to activate promoters resembling those of their parent genes.


Subject(s)
Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental/genetics , Genes, Essential/genetics , Promoter Regions, Genetic/genetics , Animals
7.
Curr Biol ; 24(15): R695-7, 2014 Aug 04.
Article in English | MEDLINE | ID: mdl-25093562

ABSTRACT

As genomes evolve, proteins with novel functions arise primarily from gene duplication and divergence events. A new study identifies several molecular mechanisms by which related transcription factors diverge over time to control new sets of target genes and novel cellular functions.


Subject(s)
Candida albicans/genetics , Evolution, Molecular , Gene Duplication , Gene Expression Regulation/genetics , Genes, Fungal/genetics , Transcription Factors/genetics , Animals , Humans
8.
Curr Biol ; 23(23): R1054-6, 2013 Dec 02.
Article in English | MEDLINE | ID: mdl-24309285

ABSTRACT

Why do some genes seem to respond in a 'digital', on/off manner to a graded signal, while others produce an 'analog', graded response? A new study suggests that the DNA-binding properties of transcription factors can strongly influence the response patterns of gene networks.


Subject(s)
Cell Communication/genetics , DNA-Binding Proteins/genetics , Drosophila/embryology , Transcription Factors/genetics , Animals , DNA-Binding Proteins/metabolism , Drosophila/growth & development , Gene Expression Regulation , Protein Binding , Signal Transduction
9.
Philos Trans R Soc Lond B Biol Sci ; 368(1632): 20130018, 2013 Dec 19.
Article in English | MEDLINE | ID: mdl-24218631

ABSTRACT

In the era of functional genomics, the role of transcription factor (TF)-DNA binding affinity is of increasing interest: for example, it has recently been proposed that low-affinity genomic binding events, though frequent, are functionally irrelevant. Here, we investigate the role of binding site affinity in the transcriptional interpretation of Hedgehog (Hh) morphogen gradients. We noted that enhancers of several Hh-responsive Drosophila genes have low predicted affinity for Ci, the Gli family TF that transduces Hh signalling in the fly. Contrary to our initial hypothesis, improving the affinity of Ci/Gli sites in enhancers of dpp, wingless and stripe, by transplanting optimal sites from the patched gene, did not result in ectopic responses to Hh signalling. Instead, we found that these enhancers require low-affinity binding sites for normal activation in regions of relatively low signalling. When Ci/Gli sites in these enhancers were altered to improve their binding affinity, we observed patterning defects in the transcriptional response that are consistent with a switch from Ci-mediated activation to Ci-mediated repression. Synthetic transgenic reporters containing isolated Ci/Gli sites confirmed this finding in imaginal discs. We propose that the requirement for gene activation by Ci in the regions of low-to-moderate Hh signalling results in evolutionary pressure favouring weak binding sites in enhancers of certain Hh target genes.


Subject(s)
Binding Sites/genetics , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila/growth & development , Drosophila/genetics , Evolution, Molecular , Gene Expression Regulation, Developmental/genetics , Transcription Factors/genetics , Wings, Animal/growth & development , Animals , Base Sequence , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Imaginal Discs/metabolism , Immunohistochemistry , Models, Genetic , Molecular Sequence Data , Protein Binding/genetics , Selection, Genetic , Sequence Alignment , Wings, Animal/metabolism
10.
Curr Biol ; 22(23): R1009-11, 2012 Dec 04.
Article in English | MEDLINE | ID: mdl-23218008

ABSTRACT

How do gene regulatory networks evolve? A new study in yeasts shows that cis- and trans-regulatory changes resulted in a hybrid state of coexisting ancestral and derived regulatory circuits. This hybrid state then diversified into a variety of modern networks.


Subject(s)
Evolution, Molecular , Gene Regulatory Networks , Yeasts/genetics , Animals
11.
Mol Syst Biol ; 8: 614, 2012.
Article in English | MEDLINE | ID: mdl-23010997

ABSTRACT

Morphogens control patterns of transcription in development, often by establishing concentration gradients of a single transcriptional activator. However, many morphogens, including Hedgehog, create opposing activator and repressor gradients (OARGs). In contrast to single activator gradients, it is not well understood how OARGs control transcriptional patterns. We present a general thermodynamic model that explains how spatial patterns of gene expression are established within OARGs. The model predicts that differences in enhancer binding site affinities for morphogen-responsive transcription factors (TFs) produce discrete transcriptional boundaries, but only when either activators or repressors bind cooperatively. This model quantitatively predicts the boundaries of gene expression within OARGs. When trained on experimental data, our model accounts for the counterintuitive observation that increasing the affinity of binding sites in enhancers of Hedgehog target genes produces more restricted transcription within Hedgehog gradients in Drosophila. Because our model is general, it may explain the role of low-affinity binding sites in many contexts, including mammalian Hedgehog gradients.


Subject(s)
Drosophila melanogaster/genetics , Models, Biological , Repressor Proteins/metabolism , Trans-Activators/metabolism , Transcription, Genetic , Animals , Base Sequence , Binding Sites , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Embryo, Nonmammalian/metabolism , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental , Molecular Sequence Data
12.
Curr Top Dev Biol ; 98: 97-120, 2012.
Article in English | MEDLINE | ID: mdl-22305160

ABSTRACT

This review focuses on a single cis-regulatory element: the sparkling eye enhancer of the Drosophila dPax2 gene. sparkling responds to Notch and EGFR signaling, along with other direct regulatory inputs, to drive gene expression that is restricted to cone cells of the developing fly retina. Functional, genetic, biochemical, evolutionary, and bioinformatic analyses have revealed surprising properties of sparkling, which may provide new insights into cis-regulatory logic and mechanisms of transcriptional activation. These properties include: a very high density of regulatory information and a correspondingly low "junk" content; an unexpectedly complex combinatorial code; tight functional constraints on enhancer organization, paradoxically coupled with high turnover of DNA sequence and binding site position; a requirement for weak binding of the transcription factor Su(H) to low-affinity sites in order to maintain a cell-type-specific response to Notch signaling; and multiple specialized regulatory sequences conferring functionally distinct activation activities, all of which are required in concert to achieve proper gene expression in vivo.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Evolution, Molecular , Eye Proteins/metabolism , Animals , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Eye Proteins/genetics , Gene Expression Regulation, Developmental , Humans , Receptors, Notch/metabolism , Signal Transduction
13.
Bioessays ; 34(2): 135-41, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22083793

ABSTRACT

This paper, in the form of a frequently asked questions page (FAQ), addresses outstanding questions about "shadow enhancers", quasi-redundant cis-regulatory elements, and their proposed roles in transcriptional control. Questions include: What exactly are shadow enhancers? How many genes have shadow/redundant/distributed enhancers? How redundant are these elements? What is the function of distributed enhancers? How modular are enhancers? Is it useful to study a single enhancer in isolation? In addition, a revised definition of "shadow enhancers" is proposed, and possible mechanisms of shadow enhancer function and evolution are discussed.


Subject(s)
Enhancer Elements, Genetic , Transcription, Genetic , Gene Expression Regulation , Genomics , Humans
14.
Curr Biol ; 21(14): 1186-96, 2011 Jul 26.
Article in English | MEDLINE | ID: mdl-21737276

ABSTRACT

BACKGROUND: Enhancers are genomic cis-regulatory sequences that integrate spatiotemporal signals to control gene expression. Enhancer activity depends on the combination of bound transcription factors as well as-in some cases-the arrangement and spacing of binding sites for these factors. Here, we examine evolutionary changes to the sequence and structure of sparkling, a Notch/EGFR/Runx-regulated enhancer that activates the dPax2 gene in cone cells of the developing Drosophila eye. RESULTS: Despite functional and structural constraints on its sequence, sparkling has undergone major reorganization in its recent evolutionary history. Our data suggest that the relative strengths of the various regulatory inputs into sparkling change rapidly over evolutionary time, such that reduced input from some factors is compensated by increased input from different regulators. These gains and losses are at least partly responsible for the changes in enhancer structure that we observe. Furthermore, stereotypical spatial relationships between certain binding sites ("grammar elements") can be identified in all sparkling orthologs-although the sites themselves are often recently derived. We also find that low binding affinity for the Notch-regulated transcription factor Su(H), a conserved property of sparkling, is required to prevent ectopic responses to Notch in noncone cells. CONCLUSIONS: Rapid DNA sequence turnover does not imply either the absence of critical cis-regulatory information or the absence of structural rules. Our findings demonstrate that even a severely constrained cis-regulatory sequence can be significantly rewired over a short evolutionary timescale.


Subject(s)
DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila/growth & development , Drosophila/genetics , Evolution, Molecular , Eye Proteins/genetics , Animals , Base Sequence , DNA-Binding Proteins/metabolism , Drosophila/metabolism , Drosophila Proteins/metabolism , Enhancer Elements, Genetic , ErbB Receptors/genetics , ErbB Receptors/metabolism , Eye/growth & development , Eye/metabolism , Eye Proteins/metabolism , Gene Expression Regulation, Developmental , Receptors, Invertebrate Peptide/genetics , Receptors, Invertebrate Peptide/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Sequence Homology, Nucleic Acid
15.
Sci Signal ; 4(176): ra38, 2011 Jun 07.
Article in English | MEDLINE | ID: mdl-21653228

ABSTRACT

Gradients of diffusible signaling proteins control precise spatial patterns of gene expression in the developing embryo. Here, we use quantitative expression measurements and thermodynamic modeling to uncover the cis-regulatory logic underlying spatially restricted gene expression in a Hedgehog (Hh) gradient in Drosophila. When Hh signaling is low, the Hh effector Gli, known as Cubitus interruptus (Ci) in Drosophila, acts as a transcriptional repressor; when Hh signaling is high, Gli acts as a transcriptional activator. Counterintuitively and in contrast to previous models of Gli-regulated gene expression, we found that low-affinity binding sites for Ci were required for proper spatial expression of the Hh target gene decapentaplegic (dpp) in regions of low Hh signal. Three low-affinity Ci sites enabled expression of dpp in response to low signal; increasing the affinity of these sites restricted dpp expression to regions of maximal signaling. A model incorporating cooperative repression by Ci correctly predicted the in vivo expression of a reporter gene controlled by a single Ci site. Our work clarifies how transcriptional activators and repressors, competing for common binding sites, can transmit positional information to the genome. It also provides an explanation for the widespread presence of conserved, nonconsensus Gli binding sites in Hh target genes.


Subject(s)
Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Transcription Factors/physiology , Animals , Binding Sites , Binding, Competitive , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Drosophila Proteins/metabolism , Drosophila Proteins/physiology , Drosophila melanogaster , Oncogene Proteins/metabolism , Oncogene Proteins/physiology , Repressor Proteins , Trans-Activators/metabolism , Trans-Activators/physiology , Transcription Factors/metabolism , Zinc Finger Protein GLI1
17.
Dev Cell ; 18(3): 359-70, 2010 Mar 16.
Article in English | MEDLINE | ID: mdl-20230745

ABSTRACT

Enhancers integrate spatiotemporal information to generate precise patterns of gene expression. How complex is the regulatory logic of a typical developmental enhancer, and how important is its internal organization? Here, we examine in detail the structure and function of sparkling, a Notch- and EGFR/MAPK-regulated, cone cell-specific enhancer of the Drosophila Pax2 gene, in vivo. In addition to its 12 previously identified protein-binding sites, sparkling is densely populated with previously unmapped regulatory sequences, which interact in complex ways to control gene expression. One segment is essential for activation at a distance, yet dispensable for other activation functions and for cell type patterning. Unexpectedly, rearranging sparkling's regulatory sites converts it into a robust photoreceptor-specific enhancer. Our results show that a single combination of regulatory inputs can encode multiple outputs, and suggest that the enhancer's organization determines the correct expression pattern by facilitating certain short-range regulatory interactions at the expense of others.


Subject(s)
DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila/growth & development , Drosophila/genetics , ErbB Receptors/metabolism , Eye Proteins/genetics , Eye/growth & development , Receptors, Invertebrate Peptide/metabolism , Receptors, Notch/metabolism , Animals , Animals, Genetically Modified , Base Sequence , Binding Sites/genetics , DNA/genetics , DNA-Binding Proteins/metabolism , Drosophila/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Enhancer Elements, Genetic , ErbB Receptors/genetics , Evolution, Molecular , Eye/metabolism , Eye Proteins/metabolism , Gene Expression Regulation, Developmental , Genes, Insect , MAP Kinase Signaling System , Molecular Sequence Data , Mutagenesis , PAX2 Transcription Factor/genetics , PAX2 Transcription Factor/metabolism , Photoreceptor Cells, Invertebrate/cytology , Photoreceptor Cells, Invertebrate/metabolism , Receptors, Invertebrate Peptide/genetics , Receptors, Notch/genetics , Sequence Homology, Nucleic Acid
18.
Dev Biol ; 323(1): 41-52, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18786525

ABSTRACT

The highly conserved Wingless/Wnt signaling pathway controls many developmental processes by regulating the expression of target genes, most often through members of the TCF family of DNA-binding proteins. In the absence of signaling, many of these targets are silenced, by mechanisms involving TCFs that are not fully understood. Here we report that the chromatin remodeling proteins ISWI and ACF1 are required for basal repression of WG target genes in Drosophila. This regulation is not due to global repression by ISWI and ACF1 and is distinct from their previously reported role in chromatin assembly. While ISWI is localized to the same regions of Wingless target gene chromatin as TCF, we find that ACF1 binds much more broadly to target loci. This broad distribution of ACF1 is dependent on ISWI. ISWI and ACF1 are required for TCF binding to chromatin, while a TCF-independent role of ISWI-ACF1 in repression of Wingless targets is also observed. Finally, we show that Wingless signaling reduces ACF1 binding to WG targets, and ISWI and ACF1 regulate repression by antagonizing histone H4 acetylation. Our results argue that WG signaling activates target gene expression partly by overcoming the chromatin barrier maintained by ISWI and ACF1.


Subject(s)
Adenosine Triphosphatases/metabolism , Drosophila Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Wnt1 Protein/metabolism , Adenosine Triphosphatases/genetics , Animals , Cells, Cultured , Chromatin/metabolism , Drosophila/cytology , Drosophila/genetics , Drosophila/metabolism , Drosophila/physiology , Drosophila Proteins/genetics , Mutation , Protein Binding , Repressor Proteins/genetics , Transcription Factors/genetics , Transcription, Genetic , Wnt1 Protein/genetics
19.
Tissue Eng Part A ; 14(9): 1549-59, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18687053

ABSTRACT

Enhancers, or cis-regulatory elements, are the principal determinants of spatiotemporal patterning of gene expression. For reasons of clinical and research utility, it is desirable to build customized enhancers that drive novel gene expression patterns, but currently, we largely rely on "found" genomic elements. Synthetic enhancers, assembled from transcription factor binding sites taken from natural signal-regulated enhancers, generally fail to behave like their wild-type counterparts when placed in transgenic animals, suggesting that important aspects of enhancer function are still unexplored. As a step toward the creation of a truly synthetic regulatory element, we have undertaken an extensive structure-function study of an enhancer of the Drosophila decapentaplegic (dpp) gene that drives expression in the developing visceral mesoderm (VM). Although considerable past efforts have been made to dissect the dppVM enhancer, transgenic experiments presented here indicate that its activity cannot be explained by the known regulators alone. dppVM contains multiple, previously uncharacterized, regulatory sites, some of which exhibit functional redundancy. The results presented here suggest that even the best-studied enhancers must be further dissected before they can be fully understood, and before faithful synthetic elements based on them can be created. Implications for developmental genetics, mathematical modeling, and therapeutic applications are discussed.


Subject(s)
Enhancer Elements, Genetic/genetics , Transcription, Genetic , Animals , Base Sequence , Drosophila , Drosophila Proteins/genetics , Gene Expression Regulation , Genes, Insect/genetics , Molecular Sequence Data , Sequence Homology, Nucleic Acid
20.
Dev Biol ; 321(2): 446-54, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18585374

ABSTRACT

Signaling pathways usually activate transcriptional targets in a cell type-specific manner. Notable exceptions are pathway-specific feedback antagonists, which serve to restrict the range or duration of the signal. These factors are often activated by their respective pathways in a broad array of cell types. For example, the Wnt ligand Wingless (Wg) activates the naked cuticle (nkd) gene in all tissues examined throughout Drosophila development. How does the nkd gene respond in such an unrestricted manner to Wg signaling? Analysis in cell culture revealed regions of the nkd locus that contain Wg response elements (WREs) that are directly activated by the pathway via the transcription factor TCF. In flies, Wg signaling activates these WREs in multiple tissues, in distinct but overlapping patterns. These WREs are necessary and largely sufficient for nkd expression in late stage larval tissues, but only contribute to part of the embryonic expression pattern of nkd. These results demonstrate that nkd responsiveness to Wg signaling is achieved by several WREs which are broadly (but not universally) activated by the pathway. The existence of several WREs in the nkd locus may have been necessary to allow the Wg signaling-Nkd feedback circuit to remain intact as Wg expression diversified during animal evolution.


Subject(s)
Drosophila Proteins/metabolism , Gene Expression Regulation, Developmental/physiology , Signal Transduction/physiology , Wnt1 Protein/metabolism , Animals , Chromatin Immunoprecipitation , Drosophila , In Situ Hybridization , RNA Interference , Response Elements/physiology , T Cell Transcription Factor 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...