Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
G3 (Bethesda) ; 10(9): 3015-3024, 2020 09 02.
Article in English | MEDLINE | ID: mdl-32601058

ABSTRACT

This study has taken advantage of the availability of the assembled genomic sequence of flies, mosquitos, ants and bees to explore the presence of ultraconserved sequence elements in these phylogenetic groups. We compared non-coding sequences found within and flanking Drosophila developmental genes to homologous sequences in Ceratitis capitata and Musca domestica Many of the conserved sequence blocks (CSBs) that constitute Drosophila cis-regulatory DNA, recognized by EvoPrinter alignment protocols, are also conserved in Ceratitis and Musca Also conserved is the position but not necessarily the orientation of many of these ultraconserved CSBs (uCSBs) with respect to flanking genes. Using the mosquito EvoPrint algorithm, we have also identified uCSBs shared among distantly related mosquito species. Side by side comparison of bee and ant EvoPrints of selected developmental genes identify uCSBs shared between these two Hymenoptera, as well as less conserved CSBs in either one or the other taxon but not in both. Analysis of uCSBs in these dipterans and Hymenoptera will lead to a greater understanding of their evolutionary origin and function of their conserved non-coding sequences and aid in discovery of core elements of enhancers.This study applies the phylogenetic footprinting program EvoPrinter to detection of ultraconserved non-coding sequence elements in Diptera, including flies and mosquitos, and Hymenoptera, including ants and bees. EvoPrinter outputs an interspecies comparison as a single sequence in terms of the input reference sequence. Ultraconserved sequences flanking known developmental genes were detected in Ceratitis and Musca when compared with Drosophila species, in Aedes and Culex when compared with Anopheles, and between ants and bees. Our methods are useful in detecting and understanding the core evolutionarily hardened sequences required for gene regulation.


Subject(s)
Diptera , Hymenoptera , Animals , Bees , Conserved Sequence , DNA , Diptera/genetics , Drosophila/genetics , Hymenoptera/genetics , Phylogeny
2.
Elife ; 92020 04 14.
Article in English | MEDLINE | ID: mdl-32286225

ABSTRACT

Existing genetic methods of neuronal targeting do not routinely achieve the resolution required for mapping brain circuits. New approaches are thus necessary. Here, we introduce a method for refined neuronal targeting that can be applied iteratively. Restriction achieved at the first step can be further refined in a second step, if necessary. The method relies on first isolating neurons within a targeted group (i.e. Gal4 pattern) according to their developmental lineages, and then intersectionally limiting the number of lineages by selecting only those in which two distinct neuroblast enhancers are active. The neuroblast enhancers drive expression of split Cre recombinase fragments. These are fused to non-interacting pairs of split inteins, which ensure reconstitution of active Cre when all fragments are expressed in the same neuroblast. Active Cre renders all neuroblast-derived cells in a lineage permissive for Gal4 activity. We demonstrate how this system can facilitate neural circuit-mapping in Drosophila.


In humans ­ as well as flies and most other animals ­ the brain controls how we move and behave, and regulates heartbeat, breathing and other core processes. To perform these different roles, cells known as neurons form large networks that quickly carry messages around the brain and to other parts of the body. In order to fully understand how the brain works, it is important to first understand how individual neurons connect to each other and operate within these networks. Fruit flies and other animals with small brains are often used as models to study how the brain works. There are several methods currently available that allow researchers to manipulate small groups of fruit fly neurons for study, and in some cases it is even possible to target individual neurons. However, it remains an aspirational goal to be able to target every neuron in the fly brain individually. The Gal4-UAS system is a way of manipulating gene activity widely used to study neurons in fruit flies. The system consists of two parts: a protein that can bind DNA and control the activity of genes (Gal4); and a genetic sequence (the UAS) that tells Gal4 where to bind and therefore which genes to activate. Fruit flies can be genetically engineered so that only specific cells make Gal4. This makes it possible, for example, to limit the activity of a gene under the control of the UAS to a specific set of neurons and therefore to identify or target these neurons. Luan et al. developed a new technique named SpaRCLIn that allows the targeting of a subset of neurons within a group already identified with the Gal4-UAS system. During embryonic development, all neurons originate from a small pool of cells called neuroblasts, and it is possible to target the descendants of particular neuroblasts. SpaRCLIn exploits this strategy to limit the activity of Gal4 to smaller and smaller numbers of neuroblast descendants. In this way, Luan et al. found that SpaRCLIn was routinely capable of limiting patterns of Gal4 activity to one, or a few, neurons at a time. Further experiments used SpaRCLIn to identify two pairs of neurons that trigger a well-known feeding behavior in fruit flies. Luan et al. also developed a SpaRCLIn toolkit that will form the basis of a community resource other researchers can use to study neurons in fruit flies. These findings could also benefit researchers developing similar tools in mice and other animals.


Subject(s)
Genetic Techniques , Integrases , Neurons , Animals , Animals, Genetically Modified , Drosophila , Neural Pathways , Neural Stem Cells/metabolism
3.
Genesis ; 56(8): e23237, 2018 08.
Article in English | MEDLINE | ID: mdl-30005136

ABSTRACT

While developmental studies of Drosophila neural stem cell lineages have identified transcription factors (TFs) important to cell identity decisions, currently only an incomplete understanding exists of the cis-regulatory elements that control the dynamic expression of these TFs. Our previous studies have identified multiple enhancers that regulate the POU-domain TF paralogs nubbin and pdm-2 genes. Evolutionary comparative analysis of these enhancers reveals that they each contain multiple conserved sequence blocks (CSBs) that span TF DNA-binding sites for known regulators of neuroblast (NB) gene expression in addition to novel sequences. This study functionally analyzes the conserved DNA sequence elements within a NB enhancer located within the nubbin gene and highlights a high level of complexity underlying enhancer structure. Mutational analysis has revealed CSBs that are important for enhancer activation and silencing in the developing CNS. We have also observed that adjusting the number and relative positions of the TF binding sites within these CSBs alters enhancer function.


Subject(s)
Drosophila Proteins/genetics , Drosophila/genetics , Homeodomain Proteins/genetics , POU Domain Factors/genetics , Animals , Base Sequence , Binding Sites , Central Nervous System/embryology , Conserved Sequence/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental/genetics , Genes, Reporter , Neural Stem Cells/metabolism , Phylogeny , Transcription Factors/genetics , Transcription, Genetic
4.
Genesis ; 56(3): e23094, 2018 03.
Article in English | MEDLINE | ID: mdl-29417712

ABSTRACT

Evolutionary analysis of cis-regulatory DNA reveals that enhancers consist of clusters of conserved sequence blocks (CSBs) that are made up of both unique and repeated sequence elements. This study seeks to address the basis for spatial and temporal regulation of neuroblas. A search for temporally restricted CNS NB enhancers identified one within the transcription factor grainyhead (grh) gene locus. The intronic enhancer, grh-15, contains two separable semi-autonomous activities, one that drives expression predominantly within the developing brain NBs and another in ventral cord NBs. To gain insight into the function of the CSBs constituting the brain-specific enhancer, we have systematically deleted each CSB and compared the activity of the altered enhancer to that of the full brain-specific enhancer. While our results indicate that information regulating enhancer activity is highly redundant, we have found that individual CSBs convey expression in subsets of larval lineages that are generated from either Type I or Type II NBs. These studies also highlight how evolutionary sequence conservation can be used as a guide the functional analysis of cis-regulatory DNA.


Subject(s)
DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila/genetics , Enhancer Elements, Genetic , Regulatory Sequences, Nucleic Acid , Transcription Factors/genetics , Animals , Animals, Genetically Modified , Base Sequence , Conserved Sequence , Fluorescent Antibody Technique , Gene Deletion , Gene Expression , Gene Expression Regulation , Genes, Reporter , Genome, Insect , Genomics/methods , Immunohistochemistry , Phenotype , Sequence Analysis, DNA
5.
PLoS Negl Trop Dis ; 11(6): e0005673, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28622346

ABSTRACT

BACKGROUND: Flavivirus and Filovirus infections are serious epidemic threats to human populations. Multi-genome comparative analysis of these evolving pathogens affords a view of their essential, conserved sequence elements as well as progressive evolutionary changes. While phylogenetic analysis has yielded important insights, the growing number of available genomic sequences makes comparisons between hundreds of viral strains challenging. We report here a new approach for the comparative analysis of these hemorrhagic fever viruses that can superimpose an unlimited number of one-on-one alignments to identify important features within genomes of interest. METHODOLOGY/PRINCIPAL FINDING: We have adapted EvoPrinter alignment algorithms for the rapid comparative analysis of Flavivirus or Filovirus sequences including Zika and Ebola strains. The user can input a full genome or partial viral sequence and then view either individual comparisons or generate color-coded readouts that superimpose hundreds of one-on-one alignments to identify unique or shared identity SNPs that reveal ancestral relationships between strains. The user can also opt to select a database genome in order to access a library of pre-aligned genomes of either 1,094 Flaviviruses or 460 Filoviruses for rapid comparative analysis with all database entries or a select subset. Using EvoPrinter search and alignment programs, we show the following: 1) superimposing alignment data from many related strains identifies lineage identity SNPs, which enable the assessment of sublineage complexity within viral outbreaks; 2) whole-genome SNP profile screens uncover novel Dengue2 and Zika recombinant strains and their parental lineages; 3) differential SNP profiling identifies host cell A-to-I hyper-editing within Ebola and Marburg viruses, and 4) hundreds of superimposed one-on-one Ebola genome alignments highlight ultra-conserved regulatory sequences, invariant amino acid codons and evolutionarily variable protein-encoding domains within a single genome. CONCLUSIONS/SIGNIFICANCE: EvoPrinter allows for the assessment of lineage complexity within Flavivirus or Filovirus outbreaks, identification of recombinant strains, highlights sequences that have undergone host cell A-to-I editing, and identifies unique input and database SNPs within highly conserved sequences. EvoPrinter's ability to superimpose alignment data from hundreds of strains onto a single genome has allowed us to identify unique Zika virus sublineages that are currently spreading in South, Central and North America, the Caribbean, and in China. This new set of integrated alignment programs should serve as a useful addition to existing tools for the comparative analysis of these viruses.


Subject(s)
Computational Biology/methods , Filoviridae/classification , Filoviridae/genetics , Flavivirus/classification , Flavivirus/genetics , Sequence Alignment/methods , Animals , Evolution, Molecular , Humans , Molecular Epidemiology/methods
6.
BMC Genomics ; 16: 700, 2015 Sep 16.
Article in English | MEDLINE | ID: mdl-26377945

ABSTRACT

BACKGROUND: One of the major challenges in developmental biology is to understand the regulatory events that generate neuronal diversity. During Drosophila embryonic neural lineage development, cellular temporal identity is established in part by a transcription factor (TF) regulatory network that mediates a cascade of cellular identity decisions. Two of the regulators essential to this network are the POU-domain TFs Nubbin and Pdm-2, encoded by adjacent genes collectively known as pdm. The focus of this study is the discovery and characterization of cis-regulatory DNA that governs their expression. RESULTS: Phylogenetic footprinting analysis of a 125 kb genomic region that spans the pdm locus identified 116 conserved sequence clusters. To determine which of these regions function as cis-regulatory enhancers that regulate the dynamics of pdm gene expression, we tested each for in vivo enhancer activity during embryonic development and postembryonic neurogenesis. Our screen revealed 77 unique enhancers positioned throughout the noncoding region of the pdm locus. Many of these activated neural-specific gene expression during different developmental stages and many drove expression in overlapping patterns. Sequence comparisons of functionally related enhancers that activate overlapping expression patterns revealed that they share conserved elements that can be predictive of enhancer behavior. To facilitate data accessibility, the results of our analysis are catalogued in cisPatterns, an online database of the structure and function of these and other Drosophila enhancers. CONCLUSIONS: These studies reveal a diversity of modular enhancers that most likely regulate pdm gene expression during embryonic and adult development, highlighting a high level of temporal and spatial expression specificity. In addition, we discovered clusters of functionally related enhancers throughout the pdm locus. A subset of these enhancers share conserved elements including sequences that correspond to known TF DNA binding sites. Although comparative analysis of the nubbin and pdm-2 encoding sequences indicate that these two genes most likely arose from a duplication event, we found only partial evidence of sequence duplication between their enhancers, suggesting that after the putative duplication their cis-regulatory DNA diverged at a higher rate than their coding sequences.


Subject(s)
Drosophila Proteins/genetics , Drosophila/genetics , Enhancer Elements, Genetic , Genetic Loci , Homeodomain Proteins/genetics , Neurogenesis/genetics , POU Domain Factors/genetics , Animals , Base Sequence , Brain/metabolism , Cluster Analysis , Conserved Sequence , Drosophila Proteins/chemistry , Gene Expression Regulation , Gene Expression Regulation, Developmental , Gene Order , Genes, Reporter , Homeodomain Proteins/chemistry , Molecular Sequence Data , Organ Specificity/genetics , POU Domain Factors/chemistry , Sequence Alignment , Sequence Analysis, DNA , Transgenes
7.
Nat Neurosci ; 17(4): 631-7, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24561995

ABSTRACT

The Drosophila cerebrum originates from about 100 neuroblasts per hemisphere, with each neuroblast producing a characteristic set of neurons. Neurons from a neuroblast are often so diverse that many neuron types remain unexplored. We developed new genetic tools that target neuroblasts and their diverse descendants, increasing our ability to study fly brain structure and development. Common enhancer-based drivers label neurons on the basis of terminal identities rather than origins, which provides limited labeling in the heterogeneous neuronal lineages. We successfully converted conventional drivers that are temporarily expressed in neuroblasts, into drivers expressed in all subsequent neuroblast progeny. One technique involves immortalizing GAL4 expression in neuroblasts and their descendants. Another depends on loss of the GAL4 repressor, GAL80, from neuroblasts during early neurogenesis. Furthermore, we expanded the diversity of MARCM-based reagents and established another site-specific mitotic recombination system. Our transgenic tools can be combined to map individual neurons in specific lineages of various genotypes.


Subject(s)
Cell Lineage , Cerebrum/cytology , Drosophila Proteins , Drosophila/cytology , Genetic Techniques , Neural Stem Cells/cytology , Animals , Cell Lineage/physiology , Cerebrum/physiology , Drosophila/physiology , Drosophila Proteins/biosynthesis , Drosophila Proteins/genetics , Neural Stem Cells/metabolism , Neural Stem Cells/physiology , Receptors, Notch/biosynthesis , Receptors, Notch/genetics , Recombination, Genetic , Transgenes
8.
Development ; 141(2): 253-8, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24306106

ABSTRACT

Drosophila type II neuroblasts (NBs), like mammalian neural stem cells, deposit neurons through intermediate neural progenitors (INPs) that can each produce a series of neurons. Both type II NBs and INPs exhibit age-dependent expression of various transcription factors, potentially specifying an array of diverse neurons by combinatorial temporal patterning. Not knowing which mature neurons are made by specific INPs, however, conceals the actual variety of neuron types and limits further molecular studies. Here we mapped neurons derived from specific type II NB lineages and found that sibling INPs produced a morphologically similar but temporally regulated series of distinct neuron types. This suggests a common fate diversification program operating within each INP that is modulated by NB age to generate slightly different sets of diverse neurons based on the INP birth order. Analogous mechanisms might underlie the expansion of neuron diversity via INPs in mammalian brain.


Subject(s)
Drosophila/cytology , Drosophila/growth & development , Neural Stem Cells/cytology , Animals , Animals, Genetically Modified , Brain/cytology , Brain/growth & development , Brain/metabolism , Cell Lineage , Drosophila/genetics , Models, Neurological , Neural Stem Cells/classification , Neural Stem Cells/metabolism , Neurogenesis
9.
PLoS One ; 8(4): e60137, 2013.
Article in English | MEDLINE | ID: mdl-23613719

ABSTRACT

Analysis of cis-regulatory enhancers has revealed that they consist of clustered blocks of highly conserved sequences. Although most characterized enhancers reside near their target genes, a growing number of studies have shown that enhancers located over 50 kb from their minimal promoter(s) are required for appropriate gene expression and many of these 'long-range' enhancers are found in genomic regions that are devoid of identified exons. To gain insight into the complexity of Drosophila cis-regulatory sequences within exon-poor regions, we have undertaken an evolutionary analysis of 39 of these regions located throughout the genome. This survey revealed that within these genomic expanses, clusters of conserved sequence blocks (CSBs) are positioned once every 1.1 kb, on average, and that a typical cluster contains multiple (5 to 30 or more) CSBs that have been maintained for at least 190 My of evolutionary divergence. As an initial step toward assessing the cis-regulatory activity of conserved clusters within gene-free genomic expanses, we have tested the in-vivo enhancer activity of 19 consecutive CSB clusters located in the middle of a 115 kb gene-poor region on the 3(rd) chromosome. Our studies revealed that each cluster functions independently as a specific spatial/temporal enhancer. In total, the enhancers possess a diversity of regulatory functions, including dynamically activating expression in defined patterns within subsets of cells in discrete regions of the embryo, larvae and/or adult. We also observed that many of the enhancers are multifunctional-that is, they activate expression during multiple developmental stages. By extending these results to the rest of the Drosophila genome, which contains over 70,000 non-coding CSB clusters, we suggest that most function as enhancers.


Subject(s)
Drosophila/genetics , Enhancer Elements, Genetic/genetics , Genome, Insect/genetics , Animals , Base Sequence , Chromosomes, Insect/genetics , Conserved Sequence/genetics , Genes, Insect/genetics , Genes, Reporter/genetics , Genomics , Molecular Sequence Data , Multigene Family/genetics , Species Specificity
10.
PLoS One ; 7(7): e39026, 2012.
Article in English | MEDLINE | ID: mdl-22859937

ABSTRACT

Coding region alterations of ZIC2 are the second most common type of mutation in holoprosencephaly (HPE). Here we use several complementary bioinformatic approaches to identify ultraconserved cis-regulatory sequences potentially driving the expression of human ZIC2. We demonstrate that an 804 bp element in the 3' untranslated region (3'UTR) is highly conserved across the evolutionary history of vertebrates from fish to humans. Furthermore, we show that while genetic variation of this element is unexpectedly common among holoprosencephaly subjects (6/528 or >1%), it is not present in control individuals. Two of six proband-unique variants are de novo, supporting their pathogenic involvement in HPE outcomes. These findings support a general recommendation that the identification and analysis of key ultraconserved elements should be incorporated into the genetic risk assessment of holoprosencephaly cases.


Subject(s)
3' Untranslated Regions , Holoprosencephaly/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Animals , Base Sequence , Body Patterning , Conserved Sequence , DNA Mutational Analysis , Gene Regulatory Networks , Genetic Association Studies , Humans , Molecular Sequence Data , Prosencephalon/growth & development , Sequence Alignment , Zebrafish
11.
Gene Expr Patterns ; 12(7-8): 261-72, 2012.
Article in English | MEDLINE | ID: mdl-22691242

ABSTRACT

In the developing CNS, unique functional identities among neurons and glia are, in part, established as a result of successive transitions in gene expression programs within neural precursor cells. One of the temporal-identity windows within Drosophila CNS neural precursor cells or neuroblasts (NBs) is marked by the expression of a zinc-finger transcription factor (TF) gene, castor (cas). Our analysis of cis-regulatory DNA within a cas loss-of-function rescue fragment has identified seven enhancers that independently activate reporter transgene expression in specific sub-patterns of the wild-type embryonic cas gene expression domain. Most of these enhancers also regulate different aspects of cas expression within the larval and adult CNS. Phylogenetic footprinting reveals that each enhancer is made up of clusters of highly conserved DNA sequence blocks that are flanked by less-conserved inter-cluster spacer sequences. Comparative analysis of the conserved DNA also reveals that cas enhancers share different combinations of sequence elements and many of these shared elements contain core DNA-binding recognition motifs for characterized temporal-identity TFs. Intra-species alignments show that two of the sub-pattern enhancers originated from an inverted duplication and that this repeat is unique to the cas locus in all sequenced Drosophila species. Finally we show that three of the enhancers differentially require cas function for their wild-type regulatory behavior. Cas limits the expression of one enhancer while two others require cas function for full expression. These studies represent a starting point for the further analysis of cas gene expression and the TFs that regulate it.


Subject(s)
Central Nervous System/metabolism , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila/genetics , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Animals , Base Sequence , Central Nervous System/cytology , Conserved Sequence , DNA-Binding Proteins/metabolism , Drosophila/cytology , Drosophila/embryology , Drosophila Proteins/metabolism , Evolution, Molecular , Female , Genes, Reporter , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Larva/genetics , Larva/growth & development , Larva/metabolism , Male , Molecular Sequence Data , Neural Stem Cells/metabolism , Transcription, Genetic
12.
Dev Dyn ; 241(1): 169-89, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22174086

ABSTRACT

BACKGROUND: Phylogenetic footprinting has revealed that cis-regulatory enhancers consist of conserved DNA sequence clusters (CSCs). Currently, there is no systematic approach for enhancer discovery and analysis that takes full-advantage of the sequence information within enhancer CSCs. RESULTS: We have generated a Drosophila genome-wide database of conserved DNA consisting of >100,000 CSCs derived from EvoPrints spanning over 90% of the genome. cis-Decoder database search and alignment algorithms enable the discovery of functionally related enhancers. The program first identifies conserved repeat elements within an input enhancer and then searches the database for CSCs that score highly against the input CSC. Scoring is based on shared repeats as well as uniquely shared matches, and includes measures of the balance of shared elements, a diagnostic that has proven to be useful in predicting cis-regulatory function. To demonstrate the utility of these tools, a temporally-restricted CNS neuroblast enhancer was used to identify other functionally related enhancers and analyze their structural organization. CONCLUSIONS: cis-Decoder reveals that co-regulating enhancers consist of combinations of overlapping shared sequence elements, providing insights into the mode of integration of multiple regulating transcription factors. The database and accompanying algorithms should prove useful in the discovery and analysis of enhancers involved in any developmental process.


Subject(s)
Databases, Genetic , Drosophila melanogaster/genetics , Enhancer Elements, Genetic , Genome, Insect , Algorithms , Animals , Base Sequence , Computational Biology/methods , Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/embryology , Gene Expression Regulation, Developmental , Molecular Sequence Data , Phylogeny , Transgenes
13.
Mech Dev ; 128(3-4): 165-77, 2011.
Article in English | MEDLINE | ID: mdl-21315151

ABSTRACT

Many of the key regulators of Drosophila CNS neural identity are expressed in defined temporal orders during neuroblast (NB) lineage development. To begin to understand the structural and functional complexity of enhancers that regulate ordered NB gene expression programs, we have undertaken the mutational analysis of the temporally restricted nerfin-1 NB enhancer. Our previous studies have localized the enhancer to a region just proximal to the nerfin-1 transcription start site. Analysis of this enhancer, using the phylogenetic footprint program EvoPrinter, reveals the presence of multiple sequence blocks that are conserved among drosophilids. cis-Decoder alignments of these conserved sequence blocks (CSBs) has identified shorter elements that are conserved in other Drosophila NB enhancers. Mutagenesis of the enhancer reveals that although each CSB is required for wild-type expression, neither position nor orientation of the CSBs within the enhancer is crucial for enhancer function; removal of less-conserved or non-conserved sequences flanking CSB clusters also does not significantly alter enhancer activity. While all three conserved E-box transcription factor (TF) binding sites (CAGCTG) are required for full function, adding an additional site at different locations within non-conserved sequences interferes with enhancer activity. Of particular note, none of the mutations resulted in ectopic reporter expression outside of the early NB expression window, suggesting that the temporally restricted pattern is defined by transcriptional activators and not by direct DNA binding repressors. Our work also points to an unexpectedly large number of TFs required for optimal enhancer function - mutant TF analysis has identified at least four that are required for full enhancer regulation.


Subject(s)
Drosophila Proteins/genetics , Drosophila/genetics , Nerve Tissue Proteins/genetics , Neural Stem Cells/cytology , Neurons/cytology , Transcription Factors/genetics , Animals , Base Sequence , Central Nervous System/cytology , Central Nervous System/embryology , Conserved Sequence , Drosophila/embryology , Drosophila Proteins/metabolism , E-Box Elements , Enhancer Elements, Genetic , Genes, Reporter , In Situ Hybridization , Molecular Sequence Data , Mutagenesis, Site-Directed , Nerve Tissue Proteins/metabolism , Neural Stem Cells/metabolism , Neurons/metabolism , Sequence Deletion , Transcription Factors/metabolism , Transcription, Genetic
14.
Gene Expr Patterns ; 9(2): 65-72, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19056518

ABSTRACT

We have identified clusters of conserved sequences constituting discrete modular enhancers within the Drosophilanerfin-1 locus. nerfin-1 encodes a Zn-finger transcription factor that directs pioneer interneuron axon guidance. nerfin-1 mRNA is detected in many early delaminating neuroblasts, ganglion mother cells and transiently in nascent neurons. The comparative genomics analysis program EvoPrinter revealed conserved sequence blocks both upstream and downstream of the transcribed region. By using the aligning regions of different drosophilids as the reference DNA, EvoPrinter detects sequence length flexibility between clusters of conserved sequences and thus facilitates differentiation between closely associated modular enhancers. Expression analysis of enhancer-reporter transgenes identified enhancers that drive expression in different regions of the developing embryonic and adult nervous system, including subsets of embryonic CNS neuroblasts, GMCs, neurons and PNS neurons. In summary, EvoPrinter facilitates the discovery and analysis of enhancers that control crucial aspects of nerfin-1 expression.


Subject(s)
Central Nervous System/embryology , Conserved Sequence , Drosophila Proteins/genetics , Drosophila/genetics , Enhancer Elements, Genetic/genetics , Multigene Family/physiology , Transcription Factors/genetics , 5' Flanking Region/genetics , Animals , Animals, Genetically Modified , Base Sequence , Central Nervous System/metabolism , Comparative Genomic Hybridization , Computational Biology , Drosophila/embryology , Drosophila/metabolism , Drosophila Proteins/metabolism , Embryo, Nonmammalian , Enhancer Elements, Genetic/physiology , Gene Expression Regulation, Developmental , Genetic Variation/physiology , Molecular Sequence Data , Software , Transcription Factors/metabolism
15.
Neuron ; 60(2): 328-42, 2008 Oct 23.
Article in English | MEDLINE | ID: mdl-18957224

ABSTRACT

Drosophila vision is mediated by inputs from three types of photoreceptor neurons; R1-R6 mediate achromatic motion detection, while R7 and R8 constitute two chromatic channels. Neural circuits for processing chromatic information are not known. Here, we identified the first-order interneurons downstream of the chromatic channels. Serial EM revealed that small-field projection neurons Tm5 and Tm9 receive direct synaptic input from R7 and R8, respectively, and indirect input from R1-R6, qualifying them to function as color-opponent neurons. Wide-field Dm8 amacrine neurons receive input from 13-16 UV-sensing R7s and provide output to projection neurons. Using a combinatorial expression system to manipulate activity in different neuron subtypes, we determined that Dm8 neurons are necessary and sufficient for flies to exhibit phototaxis toward ultraviolet instead of green light. We propose that Dm8 sacrifices spatial resolution for sensitivity by relaying signals from multiple R7s to projection neurons, which then provide output to higher visual centers.


Subject(s)
Color Vision/physiology , Compound Eye, Arthropod/physiology , Drosophila melanogaster/physiology , Interneurons/physiology , Photoreceptor Cells, Invertebrate/physiology , Visual Pathways/physiology , Amacrine Cells/cytology , Amacrine Cells/physiology , Amacrine Cells/radiation effects , Animals , Color Vision/radiation effects , Compound Eye, Arthropod/cytology , Compound Eye, Arthropod/radiation effects , Drosophila melanogaster/cytology , Interneurons/cytology , Interneurons/radiation effects , Light Signal Transduction/physiology , Light Signal Transduction/radiation effects , Optic Lobe, Nonmammalian/cytology , Optic Lobe, Nonmammalian/physiology , Photic Stimulation , Photoreceptor Cells, Invertebrate/cytology , Photoreceptor Cells, Invertebrate/radiation effects , Synapses/physiology , Synapses/radiation effects , Synapses/ultrastructure , Synaptic Transmission/physiology , Synaptic Transmission/radiation effects , Ultraviolet Rays , Visual Pathways/cytology , Visual Pathways/radiation effects
16.
PLoS One ; 3(8): e3074, 2008 Aug 27.
Article in English | MEDLINE | ID: mdl-18728754

ABSTRACT

BACKGROUND: Acquisition of virulence factors and antibiotic resistance by many clinically important bacteria can be traced to horizontal gene transfer (HGT) between related or evolutionarily distant microflora. Comparative genomic analysis has become an important tool for identifying HGT DNA in emerging pathogens. We have adapted the multi-genome alignment tool EvoPrinter to facilitate discovery of HGT DNA sequences within bacterial genomes and within their mobile genetic elements. PRINCIPAL FINDINGS: EvoPrinter analysis of 13 different Staphylococcus aureus genomes revealed that one of the human isolates, the hospital epidemic methicillin-resistant MRSA252 strain, uniquely shares multiple putative HGT DNA sequences with different causative agents of bovine mastitis that are not found in the other human S. aureus isolates. MRSA252 shares over 14 different DNA sequence blocks with the bovine mastitis ET3 S. aureus strain RF122, and many of the HGT DNAs encode virulence factors. EvoPrinter analysis of the MRSA252 chromosome also uncovered virulence-factor encoding HGT events with the genome of Listeria monocytogenes and a Staphylococcus saprophyticus associated plasmid. Both bacteria are also causal agents of contagious bovine mastitis. CONCLUSIONS: EvoPrinter analysis reveals that the human MRSA252 strain uniquely shares multiple DNA sequence blocks with different causative agents of bovine mastitis, suggesting that HGT events may be occurring between these pathogens. These findings have important implications with regard to animal husbandry practices that inadvertently enhance the contact of human and livestock bacterial pathogens.


Subject(s)
Gene Transfer, Horizontal/genetics , Mastitis, Bovine/transmission , Staphylococcal Infections/transmission , Staphylococcus aureus/genetics , Animals , Cattle , Cattle Diseases/microbiology , Chromosomes, Bacterial/genetics , Community-Acquired Infections/microbiology , Community-Acquired Infections/transmission , Cross Infection/microbiology , Cross Infection/transmission , Female , Genome, Bacterial , Humans , Methicillin Resistance/genetics , Staphylococcal Infections/veterinary
17.
BMC Genomics ; 9: 371, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18673565

ABSTRACT

BACKGROUND: The presence of highly conserved sequences within cis-regulatory regions can serve as a valuable starting point for elucidating the basis of enhancer function. This study focuses on regulation of gene expression during the early events of Drosophila neural development. We describe the use of EvoPrinter and cis-Decoder, a suite of interrelated phylogenetic footprinting and alignment programs, to characterize highly conserved sequences that are shared among co-regulating enhancers. RESULTS: Analysis of in vivo characterized enhancers that drive neural precursor gene expression has revealed that they contain clusters of highly conserved sequence blocks (CSBs) made up of shorter shared sequence elements which are present in different combinations and orientations within the different co-regulating enhancers; these elements contain either known consensus transcription factor binding sites or consist of novel sequences that have not been functionally characterized. The CSBs of co-regulated enhancers share a large number of sequence elements, suggesting that a diverse repertoire of transcription factors may interact in a highly combinatorial fashion to coordinately regulate gene expression. We have used information gained from our comparative analysis to discover an enhancer that directs expression of the nervy gene in neural precursor cells of the CNS and PNS. CONCLUSION: The combined use EvoPrinter and cis-Decoder has yielded important insights into the combinatorial appearance of fundamental sequence elements required for neural enhancer function. Each of the 30 enhancers examined conformed to a pattern of highly conserved blocks of sequences containing shared constituent elements. These data establish a basis for further analysis and understanding of neural enhancer function.


Subject(s)
Conserved Sequence , Drosophila/genetics , Enhancer Elements, Genetic , Neurons , Software , Adaptor Proteins, Signal Transducing/genetics , Animals , Base Sequence , Computational Biology , Drosophila Proteins/genetics , Gene Expression Regulation, Developmental , Gene Library , Genes, Insect , Sequence Alignment , Stem Cells , Transcription Factors/genetics
18.
BMC Genomics ; 9: 106, 2008 Feb 28.
Article in English | MEDLINE | ID: mdl-18307801

ABSTRACT

BACKGROUND: Multi-genome comparative analysis has yielded important insights into the molecular details of gene regulation. We have developed EvoPrinter, a web-accessed genomics tool that provides a single uninterrupted view of conserved sequences as they appear in a species of interest. An EvoPrint reveals with near base-pair resolution those sequences that are essential for gene function. RESULTS: We describe here EvoPrinterHD, a 2nd-generation comparative genomics tool that automatically generates from a single input sequence an enhanced view of sequence conservation between evolutionarily distant species. Currently available for 5 nematode, 3 mosquito, 12 Drosophila, 20 vertebrate, 17 Staphylococcus and 20 enteric bacteria genomes, EvoPrinterHD employs a modified BLAT algorithm [enhanced-BLAT (eBLAT)], which detects up to 75% more conserved bases than identified by the BLAT alignments used in the earlier EvoPrinter program. The new program also identifies conserved sequences within rearranged DNA, highlights repetitive DNA, and detects sequencing gaps. EvoPrinterHD currently holds over 112 billion bp of indexed genomes in memory and has the flexibility of selecting a subset of genomes for analysis. An EvoDifferences profile is also generated to portray conserved sequences that are uniquely lost in any one of the orthologs. Finally, EvoPrinterHD incorporates options that allow for (1) re-initiation of the analysis using a different genome's aligning region as the reference DNA to detect species-specific changes in less-conserved regions, (2) rapid extraction and curation of conserved sequences, and (3) for bacteria, identifies unique or uniquely shared sequences present in subsets of genomes. CONCLUSION: EvoPrinterHD is a fast, high-resolution comparative genomics tool that automatically generates an uninterrupted species-centric view of sequence conservation and enables the discovery of conserved sequences within rearranged DNA. When combined with cis-Decoder, a program that discovers sequence elements shared among tissue specific enhancers, EvoPrinterHD facilitates the analysis of conserved sequences that are essential for coordinate gene regulation.


Subject(s)
Computational Biology/methods , Conserved Sequence/genetics , DNA/analysis , DNA/genetics , Genomics/methods , Software , Algorithms , Animals , Gene Duplication , Gene Rearrangement/genetics , Genome, Bacterial/genetics , Humans , Repetitive Sequences, Nucleic Acid/genetics , Sequence Alignment , Sequence Homology, Nucleic Acid , Species Specificity , Time Factors
19.
Dev Biol ; 310(1): 35-43, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17714701

ABSTRACT

The mRNA encoding the Drosophila Zn-finger transcription factor Nerfin-1, required for CNS axon pathfinding events, is subject to post-transcriptional silencing. Although nerfin-1 mRNA is expressed in many neural precursor cells including all early delaminating CNS neuroblasts, the encoded Nerfin-1 protein is detected only in the nuclei of neural precursors that divide just once to generate neurons and then only transiently in nascent neurons. Using a nerfin-1 promoter-controlled reporter transgene, replacement of the nerfin-1 3' UTR with the viral SV-40 3' UTR releases the neuroblast translational block and prolongs reporter protein expression in neurons. Comparative genomics analysis reveals that the nerfin-1 mRNA 3' UTR contains multiple highly conserved sequence blocks that either harbor and/or overlap 21 predicted binding sites for 18 different microRNAs. To determine the functional significance of these microRNA-binding sites and less conserved microRNA target sites, we have studied their ability to block or limit the expression of reporter protein in nerfin-1-expressing cells during embryonic development. Our results indicate that no single microRNA is sufficient to fully inhibit protein expression but rather multiple microRNAs that target different binding sites are required to block ectopic protein expression in neural precursor cells and temporally restrict expression in neurons. Taken together, these results suggest that multiple microRNAs play a cooperative role in the post-transcriptional regulation of nerfin-1 mRNA, and the high degree of microRNA-binding site evolutionary conservation indicates that all members of the Drosophila genus employ a similar strategy to regulate the onset and extinction dynamics of Nerfin-1 expression.


Subject(s)
Drosophila Proteins/biosynthesis , Drosophila/embryology , Nervous System/embryology , Transcription Factors/biosynthesis , Animals , Base Sequence , Drosophila/physiology , Gene Expression Regulation, Developmental , MicroRNAs/physiology , Molecular Sequence Data , Nervous System/metabolism , Organogenesis/genetics , Protein Biosynthesis , RNA Processing, Post-Transcriptional , RNA, Messenger/physiology , Sequence Alignment
20.
Genetics ; 177(1): 89-100, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17603097

ABSTRACT

Ferritin is a symmetric, 24-subunit iron-storage complex assembled of H and L chains. It is found in bacteria, plants, and animals and in two classes of mutations in the human L-chain gene, resulting in hereditary hyperferritinemia cataract syndrome or in neuroferritinopathy. Here, we examined systemic and cellular ferritin regulation and trafficking in the model organism Drosophila melanogaster. We showed that ferritin H and L transcripts are coexpressed during embryogenesis and that both subunits are essential for embryonic development. Ferritin overexpression impaired the survival of iron-deprived flies. In vivo expression of GFP-tagged holoferritin confirmed that iron-loaded ferritin molecules traffic through the Golgi organelle and are secreted into hemolymph. A constant ratio of ferritin H and L subunits, secured via tight post-transcriptional regulation, is characteristic of the secreted ferritin in flies. Differential cellular expression, conserved post-transcriptional regulation via the iron regulatory element, and distinct subcellular localization of the ferritin subunits prior to the assembly of holoferritin are all important steps mediating iron homeostasis. Our study revealed both conserved features and insect-specific adaptations of ferritin nanocages and provides novel imaging possibilities for their in vivo characterization.


Subject(s)
Apoferritins/genetics , Drosophila melanogaster/genetics , Image Processing, Computer-Assisted , Iron/metabolism , Animals , Animals, Genetically Modified , Apoferritins/metabolism , Base Sequence , Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Female , Gene Expression Regulation, Developmental , Genetic Techniques , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeostasis , In Situ Hybridization , Larva/growth & development , Larva/metabolism , Male , Molecular Sequence Data , RNA Probes , Response Elements/physiology , Subcellular Fractions
SELECTION OF CITATIONS
SEARCH DETAIL
...