Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Development ; 151(6)2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38493800

ABSTRACT

This is a personal, non-linear summary of the discovery of the homeobox, a short DNA sequence encoding a DNA-binding domain conserved in developmental control genes. It is based on our recollections, a few decaying lab notebooks and letters, the early research papers we published, and conversations with a few colleagues who were in Basel at the time. It presents a simple story, when the research we did was anything but, with failed experiments, blind alleys and dumb ideas. Homeobox DNA sequences were independently discovered by Matt Scott and Amy Weiner in Thomas Kaufmann's lab at Indiana University ( Scott and Weiner, 1984). The accompanying Perspective from Scott (2024), provides their fascinating story.


Subject(s)
DNA-Binding Proteins , Genes, Homeobox , Humans , Amino Acid Sequence , Base Sequence
2.
G3 (Bethesda) ; 12(1)2022 01 04.
Article in English | MEDLINE | ID: mdl-34791161

ABSTRACT

Gene drives are programmable genetic elements that can spread beneficial traits into wild populations to aid in vector-borne pathogen control. Two different drives have been developed for population modification of mosquito vectors. The Reckh drive (vasa-Cas9) in Anopheles stephensi displays efficient allelic conversion through males but generates frequent drive-resistant mutant alleles when passed through females. In contrast, the AgNosCd-1 drive (nos-Cas9) in Anopheles gambiae achieves almost complete allelic conversion through both genders. Here, we examined the subcellular localization of RNA transcripts in the mosquito germline. In both transgenic lines, Cas9 is strictly coexpressed with endogenous genes in stem and premeiotic cells of the testes, where both drives display highly efficient conversion. However, we observed distinct colocalization patterns for the two drives in female reproductive tissues. These studies suggest potential determinants underlying efficient drive through the female germline. We also evaluated expression patterns of alternative germline genes for future gene-drive designs.


Subject(s)
Anopheles , Gene Drive Technology , Animals , Anopheles/genetics , CRISPR-Cas Systems , Female , Germ Cells , Male , Mosquito Vectors/genetics
3.
G3 (Bethesda) ; 12(1)2022 01 04.
Article in English | MEDLINE | ID: mdl-34791185

ABSTRACT

Long noncoding RNAs (lncRNAs) have been implicated in a variety of processes in development, differentiation, and disease. In Drosophila melanogaster, the bithorax Hox cluster contains three Hox genes [Ultrabithorax (Ubx), abdominal-A, and Abdominal-B], along with a number of lncRNAs, most with unknown functions. Here, we investigated the function of a lncRNA, lncRNA:PS4 that originates in the second intron of Ubx and is transcribed in the antisense orientation to Ubx. The expression pattern of lncRNA:PS4 is complementary to Ubx in the thoracic primordia, and the lncRNA:PS4 coding region overlaps the location of the large insertion that causes the dominant homeotic mutation Contrabithorax-1 (UbxCbx-1), which partially transforms Drosophila wings into halteres via ectopic activation of Ubx. This led us to investigate the potential role of this lncRNA in regulation of Ubx expression. The UbxCbx-1 mutation dramatically changes the pattern of lncRNA:PS4, eliminating the expression of most lncRNA:PS4 sequences from parasegment 4 (where Ubx protein is normally absent) and ectopically activating lncRNA:PS4 at high levels in the abdomen (where Ubx is normally expressed). These changes, however, did not lead to changes in the Ubx embryonic transcription pattern. Targeted deletion of the two promoters of lncRNA:PS4 did not result in the change of Ubx expression in the embryos. In the genetic background of a UbxCbx-1 mutation, the lncRNA:PS4 mutation does slightly enhance the ectopic activation of Ubx protein expression in wing discs and also slightly enhances the wing phenotype seen in UbxCbx-1 heterozygotes.


Subject(s)
Drosophila Proteins , RNA, Long Noncoding , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Gene Expression Regulation, Developmental , Genes, Homeobox , Genetic Background , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Introns/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Sci Adv ; 7(46): eabk1003, 2021 Nov 12.
Article in English | MEDLINE | ID: mdl-34757777

ABSTRACT

Hox genes determine positional codes along the head-to-tail axis. Here, we replaced the entire Drosophila melanogaster proboscipedia (pb) Hox locus, which controls the development of the proboscis and maxillary palps, with that from Drosophila mimica, a related species with highly modified mouthparts. The D. mimica replacement rescues most aspects of adult proboscis morphology; however, the shape and orientation of maxillary palps were modified, resembling D. mimica and closely related species. Expressing the D. mimica Pb protein in the D. melanogaster pattern fully rescued D. melanogaster morphology. However, the expression pattern directed by D. mimica pb cis-regulatory sequences differed from that of D. melanogaster pb in cells that produce altered maxillary structures, indicating that pb regulatory sequences can evolve in related species to alter morphology.

6.
Proc Natl Acad Sci U S A ; 114(13): E2682-E2688, 2017 03 28.
Article in English | MEDLINE | ID: mdl-28289197

ABSTRACT

The epidermis serves as a protective barrier in animals. After epidermal injury, barrier repair requires activation of many wound response genes in epidermal cells surrounding wound sites. Two such genes in Drosophila encode the enzymes dopa decarboxylase (Ddc) and tyrosine hydroxylase (ple). In this paper we explore the involvement of the Toll/NF-κB pathway in the localized activation of wound repair genes around epidermal breaks. Robust activation of wound-induced transcription from ple and Ddc requires Toll pathway components ranging from the extracellular ligand Spätzle to the Dif transcription factor. Epistasis experiments indicate a requirement for Spätzle ligand downstream of hydrogen peroxide and protease function, both of which are known activators of wound-induced transcription. The localized activation of Toll a few cell diameters from wound edges is reminiscent of local activation of Toll in early embryonic ventral hypoderm, consistent with the hypothesis that the dorsal-ventral patterning function of Toll arose from the evolutionary cooption of a morphogen-responsive function in wound repair. Furthermore, the combinatorial activity of Toll and other signaling pathways in activating epidermal barrier repair genes can help explain why developmental activation of the Toll, ERK, or JNK pathways alone fail to activate wound repair loci.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/metabolism , Toll-Like Receptors/physiology , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Gene Expression Regulation , Models, Biological , NF-kappa B/metabolism , Signal Transduction , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism , Wound Healing/genetics
7.
J Thorac Oncol ; 12(4): 697-703, 2017 04.
Article in English | MEDLINE | ID: mdl-28089762

ABSTRACT

INTRODUCTION: This phase I/II trial was designed to determine the maximally tolerated dose of thoracic radiotherapy as part of a combined modality approach. This report includes the long-term outcomes of patients treated on this study. The phase II portion was never completed, as RTOG-0617 opened before it was concluded. METHODS: In this study, the maximally tolerated dose was defined as 74 Gy of radiation in 37 fractions. Twenty-five patients with unresectable NSCLC were treated with 2-Gy daily fractions and concurrent weekly carboplatin and paclitaxel. Of these patients, 20 had stage III disease and five had stage I or II disease. RESULTS: Patients were followed until death or for a minimum of 5 years in the case of survivors. The median and 5-year survivals were 42.5 months and 20% for all patients, 52.9 months and 40% in patients with stages I or II disease, and 39.8 months and 15% in patients with stage III disease. CONCLUSIONS: The median survival of the stage III patients was quite favorable. We believe that this may have been due to a robust central review program of radiotherapy plans before treatment, ensuring compliance with protocol guidelines along with very low exposure of the heart to radiotherapy. Further improvements in 5-year survival will likely require research on both systemic therapy and thoracic radiotherapy. Potential therapeutic modalities that may aid in these efforts include immunotherapy, targeted therapy, improved imaging, adaptive radiotherapy, simultaneous integrated boost techniques, novel dose fractionation regimens, and charged particle therapy.


Subject(s)
Adenocarcinoma/therapy , Carcinoma, Large Cell/therapy , Carcinoma, Non-Small-Cell Lung/therapy , Carcinoma, Squamous Cell/therapy , Chemoradiotherapy , Lung Neoplasms/therapy , Adenocarcinoma/pathology , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carboplatin/administration & dosage , Carcinoma, Large Cell/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/pathology , Female , Follow-Up Studies , Humans , Lung Neoplasms/pathology , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Staging , Paclitaxel/administration & dosage , Prognosis , Radiotherapy Dosage , Radiotherapy, Conformal , Survival Rate
8.
J Vis Exp ; (81): e50750, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24300796

ABSTRACT

The Drosophila embryo develops a robust epidermal layer that serves both to protect the internal cells from a harsh external environment as well as to maintain cellular homeostasis. Puncture injury with glass needles provides a direct method to trigger a rapid epidermal wound response that activates wound transcriptional reporters, which can be visualized by a localized reporter signal in living embryos or larvae. Puncture or laser injury also provides signals that promote the recruitment of hemocytes to the wound site. Surprisingly, severe (through and through) puncture injury in late stage embryos only rarely disrupts normal embryonic development, as greater than 90% of such wounded embryos survive to adulthood when embryos are injected in an oil medium that minimizes immediate leakage of hemolymph from puncture sites. The wound procedure does require micromanipulation of the Drosophila embryos, including manual alignment of the embryos on agar plates and transfer of the aligned embryos to microscope slides. The Drosophila epidermal wound response assay provides a quick system to test the genetic requirements of a variety of biological functions that promote wound healing, as well as a way to screen for potential chemical compounds that promote wound healing. The short life cycle and easy culturing routine make Drosophila a powerful model organism. Drosophila clean wound healing appears to coordinate the epidermal regenerative response, with the innate immune response, in ways that are still under investigation, which provides an excellent system to find conserved regulatory mechanisms common to Drosophila and mammalian epidermal wounding.


Subject(s)
Drosophila/embryology , Epidermis/injuries , Epidermis/physiology , Wound Healing/physiology , Animals , Drosophila/genetics , Female , Male , Microinjections/methods , Models, Animal , Regeneration/physiology , Wound Healing/genetics
9.
PLoS One ; 8(4): e61773, 2013.
Article in English | MEDLINE | ID: mdl-23637905

ABSTRACT

After injury to the animal epidermis, a variety of genes are transcriptionally activated in nearby cells to regenerate the missing cells and facilitate barrier repair. The range and types of diffusible wound signals that are produced by damaged epidermis and function to activate repair genes during epidermal regeneration remains a subject of very active study in many animals. In Drosophila embryos, we have discovered that serine protease function is locally activated around wound sites, and is also required for localized activation of epidermal repair genes. The serine protease trypsin is sufficient to induce a striking global epidermal wound response without inflicting cell death or compromising the integrity of the epithelial barrier. We developed a trypsin wounding treatment as an amplification tool to more fully understand the changes in the Drosophila transcriptome that occur after epidermal injury. By comparing our array results with similar results on mammalian skin wounding we can see which evolutionarily conserved pathways are activated after epidermal wounding in very diverse animals. Our innovative serine protease-mediated wounding protocol allowed us to identify 8 additional genes that are activated in epidermal cells in the immediate vicinity of puncture wounds, and the functions of many of these genes suggest novel genetic pathways that may control epidermal wound repair. Additionally, our data augments the evidence that clean puncture wounding can mount a powerful innate immune transcriptional response, with different innate immune genes being activated in an interesting variety of ways. These include puncture-induced activation only in epidermal cells in the immediate vicinity of wounds, or in all epidermal cells, or specifically in the fat body, or in multiple tissues.


Subject(s)
Drosophila/genetics , Drosophila/metabolism , Regeneration/genetics , Serine Proteases/metabolism , Signal Transduction , Wound Healing/genetics , Animals , Cell Death/drug effects , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Epidermis/injuries , Epidermis/metabolism , Gene Expression Profiling , Gene Expression Regulation , Genes, Reporter , Proteolysis , Transcription, Genetic , Trypsin/metabolism , Trypsin/pharmacology
10.
PLoS One ; 7(5): e36254, 2012.
Article in English | MEDLINE | ID: mdl-22590528

ABSTRACT

The Grainy head (GRH) family of transcription factors are crucial for the development and repair of epidermal barriers in all animals in which they have been studied. This is a high-level functional conservation, as the known structural and enzymatic genes regulated by GRH proteins differ between species depending on the type of epidermal barrier being formed. Interestingly, members of the CP2 superfamily of transcription factors, which encompasses the GRH and LSF families in animals, are also found in fungi--organisms that lack epidermal tissues. To shed light on CP2 protein function in fungi, we characterized a Neurospora crassa mutant lacking the CP2 member we refer to as grainy head-like (grhl). We show that Neurospora GRHL has a DNA-binding specificity similar to that of animal GRH proteins and dissimilar to that of animal LSF proteins. Neurospora grhl mutants are defective in conidial-spore dispersal due to an inability to remodel the cell wall, and we show that grhl mutants and the long-known conidial separation-2 (csp-2) mutants are allelic. We then characterized the transcriptomes of both Neurospora grhl mutants and Drosophila grh mutant embryos to look for similarities in the affected genes. Neurospora grhl appears to play a role in the development and remodeling of the cell wall, as well as in the activation of genes involved in defense and virulence. Drosophila GRH is required to activate the expression of many genes involved in cuticular/epidermal-barrier formation. We also present evidence that GRH plays a role in adult antimicrobial defense. These results, along with previous studies of animal GRH proteins, suggest the fascinating possibility that the apical extracellular barriers of some animals and fungi might share an evolutionary connection, and that the formation of physical barriers in the last common ancestor was under the control of a transcriptional code that included GRH-like proteins.


Subject(s)
Drosophila Proteins , Evolution, Molecular , Fungal Proteins , Neurospora , Transcription Factors , Alleles , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , Fungal Proteins/genetics , Fungal Proteins/metabolism , Mutation , Neurospora/genetics , Neurospora/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
11.
PLoS One ; 7(2): e31365, 2012.
Article in English | MEDLINE | ID: mdl-22393361

ABSTRACT

The discovery of microRNAs has resulted in a major expansion of the number of molecules known to be involved in gene regulation. Elucidating the functions of animal microRNAs has posed a significant challenge as their target interactions with messenger RNAs do not adhere to simple rules. Of the thousands of known animal microRNAs, relatively few microRNA:messenger RNA regulatory interactions have been biologically validated in an normal organismal context. Here we present evidence that three microRNAs from the Hox complex in Drosophila (miR-10-5p, miR-10-3p, miR-iab-4-5p) do not have significant effects during embryogenesis on the expression of Hox genes that contain high confidence microRNAs target sites in the 3' untranslated regions of their messenger RNAs. This is significant, in that it suggests that many predicted microRNA-target interactions may not be biologically relevant, or that the outcomes of these interactions may be so subtle that mutants may only show phenotypes in specific contexts, such as in environmental stress conditions, or in combinations with other microRNA mutations.


Subject(s)
Drosophila melanogaster/metabolism , Gene Expression Regulation , Genes, Homeobox , Homeodomain Proteins/metabolism , MicroRNAs/metabolism , 3' Untranslated Regions , Animals , Conserved Sequence , Environment , Evolution, Molecular , Gene Expression Regulation, Developmental , Genotype , Mice , MicroRNAs/genetics , Mutation , Oligonucleotide Array Sequence Analysis , Phenotype , RNA, Messenger/metabolism , Species Specificity
12.
Dev Biol ; 360(1): 230-40, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21920356

ABSTRACT

An important question in developmental biology is how relatively shallow gradients of morphogens can reliably establish a series of distinct transcriptional readouts. Current models emphasize interactions between transcription factors binding in distinct modes to cis-acting sequences of target genes. Another recent idea is that the cis-acting interactions may amplify preexisting biases or prepatterns to establish robust transcriptional responses. In this study, we examine the possible contribution of one such source of prepattern, namely gene length. We developed quantitative imaging tools to measure gene expression levels for several loci at a time on a single-cell basis and applied these quantitative imaging tools to dissect the establishment of a gene expression border separating the mesoderm and neuroectoderm in the early Drosophila embryo. We first characterized the formation of a transient ventral-to-dorsal gradient of the Snail (Sna) repressor and then examined the relationship between this gradient and repression of neural target genes in the mesoderm. We found that neural genes are repressed in a nested pattern within a zone of the mesoderm abutting the neuroectoderm, where Sna levels are graded. While several factors may contribute to the transient graded response to the Sna gradient, our analysis suggests that gene length may play an important, albeit transient, role in establishing these distinct transcriptional responses. One prediction of the gene-length-dependent transcriptional patterning model is that the co-regulated genes knirps (a short gene) and knirps-related (a long gene) should be transiently expressed in domains of differing widths, which we confirmed experimentally. These findings suggest that gene length may contribute to establishing graded responses to morphogen gradients by providing transient prepatterns that are subsequently amplified and stabilized by traditional cis-regulatory interactions.


Subject(s)
Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Genes, Insect , Transcription, Genetic , Animals , Body Patterning/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Evolution, Molecular , Gene Expression Regulation, Developmental , Genetic Complementation Test , Mesoderm/embryology , Mesoderm/metabolism , Models, Genetic , Neurogenesis/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism
13.
Proc Natl Acad Sci U S A ; 108(2): 650-5, 2011 Jan 11.
Article in English | MEDLINE | ID: mdl-21187384

ABSTRACT

Grainy head (GRH) is a key transcription factor responsible for epidermal barrier formation and repair, whose function is highly conserved across diverse animal species. However, it is not known how GRH function is reactivated to repair differentiated epidermal barriers after wounding. Here, we show that GRH is directly regulated by extracellular signal-regulated kinase (ERK) phosphorylation, which is required for wound-dependent expression of GRH target genes in epidermal cells. Serine 91 is the principal residue in GRH that is phosphorylated by ERK. Although mutations of the ERK phosphorylation sites in GRH do not impair its DNA binding function, the ERK sites in GRH are required to activate Dopa decarboxylase (Ddc) and misshapen (msn) epidermal wound enhancers as well as functional regeneration of an epidermal barrier upon wounding. This result indicates that the phosphorylation sites are essential for damaged epidermal barrier repair. However, GRH with mutant ERK phosphorylation sites can still promote barrier formation during embryonic epidermal development, suggesting that ERK sites are dispensable for the GRH function in establishing epidermal barrier integrity. These results provide mechanistic insight into how tissue repair can be initiated by posttranslational modification of a key transcription factor that normally mediates the developmental generation of that tissue.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Epidermis/embryology , Epidermis/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Developmental , Transcription Factors/metabolism , Animals , Developmental Biology , Drosophila melanogaster , Genotype , Microscopy, Fluorescence/methods , Models, Genetic , Mutation , Permeability , Phosphorylation , Wound Healing
14.
PLoS Genet ; 7(12): e1002424, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22242003

ABSTRACT

The epidermis is the largest organ of the body for most animals, and the first line of defense against invading pathogens. A breach in the epidermal cell layer triggers a variety of localized responses that in favorable circumstances result in the repair of the wound. Many cellular and genetic responses must be limited to epidermal cells that are close to wounds, but how this is regulated is still poorly understood. The order and hierarchy of epidermal wound signaling factors are also still obscure. The Drosophila embryonic epidermis provides an excellent system to study genes that regulate wound healing processes. We have developed a variety of fluorescent reporters that provide a visible readout of wound-dependent transcriptional activation near epidermal wound sites. A large screen for mutants that alter the activity of these wound reporters has identified seven new genes required to activate or delimit wound-induced transcriptional responses to a narrow zone of cells surrounding wound sites. Among the genes required to delimit the spread of wound responses are Drosophila Flotillin-2 and Src42A, both of which are transcriptionally activated around wound sites. Flotillin-2 and constitutively active Src42A are also sufficient, when overexpressed at high levels, to inhibit wound-induced transcription in epidermal cells. One gene required to activate epidermal wound reporters encodes Dual oxidase, an enzyme that produces hydrogen peroxide. We also find that four biochemical treatments (a serine protease, a Src kinase inhibitor, methyl-ß-cyclodextrin, and hydrogen peroxide) are sufficient to globally activate epidermal wound response genes in Drosophila embryos. We explore the epistatic relationships among the factors that induce or delimit the spread of epidermal wound signals. Our results define new genetic functions that interact to instruct only a limited number of cells around puncture wounds to mount a transcriptional response, mediating local repair and regeneration.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Membrane Proteins/genetics , NADPH Oxidases/genetics , Proto-Oncogene Proteins pp60(c-src)/genetics , Wound Healing/genetics , Animals , Drosophila melanogaster/physiology , Epidermis/physiology , Gene Expression Regulation , Hydrogen Peroxide/chemistry , Mutation , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Wound Healing/drug effects , beta-Cyclodextrins/pharmacology
15.
Dev Biol ; 344(1): 358-62, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20435033

ABSTRACT

The enormous diversity of extant animal forms is a testament to the power of evolution, and much of this diversity has been achieved through the emergence of novel morphological traits. The origin of novel morphological traits is an extremely important issue in biology, and a frequent source of this novelty is co-option of pre-existing genetic systems for new purposes (Carroll et al., 2008). Appendages, such as limbs, fins and antennae, are structures common to many animal body plans which must have arisen at least once, and probably multiple times, in lineages which lacked appendages. We provide evidence that appendage proximodistal patterning genes are expressed in similar registers in the anterior embryonic neurectoderm of Drosophila melanogaster and Saccoglossus kowalevskii (a hemichordate). These results, in concert with existing expression data from a variety of other animals suggest that a pre-existing genetic system for anteroposterior head patterning was co-opted for patterning of the proximodistal axis of appendages of bilaterian animals.


Subject(s)
Body Patterning , Extremities/embryology , Animals , Biological Evolution , Chordata/embryology , Developmental Biology/methods , Drosophila melanogaster/embryology , Gene Expression Regulation, Developmental , Genes, Insect , In Situ Hybridization , Models, Biological , Molecular Sequence Data , Neural Plate/metabolism , Oligonucleotides/metabolism
16.
Evol Dev ; 12(2): 131-43, 2010.
Article in English | MEDLINE | ID: mdl-20433454

ABSTRACT

We tested whether Artemia abd-A could repress limbs in Drosophila embryos, and found that although abd-A transcripts were produced, ABD-A protein was not. Similarly, developing Artemia epidermal cells showed expression of abd-A transcripts without accumulation of ABD-A protein. This finding in Artemia reveals a new variation in Hox gene function that is associated with morphological evolution. In this case, a HOX protein expression pattern is completely absent during early development, although the HOX protein is expressed at later stages in the central nervous system in a "homeotic-like" pattern. The combination of an absence of ABD-A protein expression in the Artemia limb primordia and the weak repressive function of Artemia UBX protein on the limb-promoting gene Dll are likely to be two reasons why homonomous limbs develop throughout the entire Artemia trunk.


Subject(s)
Artemia/genetics , Drosophila melanogaster/embryology , Embryo, Nonmammalian/metabolism , Extremities/growth & development , Gene Expression Regulation, Developmental , Homeodomain Proteins/physiology , Animals , Artemia/growth & development , Artemia/metabolism , Casein Kinase II/genetics , Casein Kinase II/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Female , Gene Silencing , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , In Situ Hybridization , Insect Proteins/genetics , Insect Proteins/metabolism , Male , Phosphorylation , Transcription Factors/genetics , Transcription Factors/metabolism
17.
Nucleic Acids Res ; 38(10): e115, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20164092

ABSTRACT

Fluorescent in situ hybridization (FISH) techniques are becoming extremely sensitive, to the point where individual RNA or DNA molecules can be detected with small probes. At this level of sensitivity, the elimination of 'off-target' hybridization is of crucial importance, but typical probes used for RNA and DNA FISH contain sequences repeated elsewhere in the genome. We find that very short (e.g. 20 nt) perfect repeated sequences within much longer probes (e.g. 350-1500 nt) can produce significant off-target signals. The extent of noise is surprising given the long length of the probes and the short length of non-specific regions. When we removed the small regions of repeated sequence from either short or long probes, we find that the signal-to-noise ratio is increased by orders of magnitude, putting us in a regime where fluorescent signals can be considered to be a quantitative measure of target transcript numbers. As the majority of genes in complex organisms contain repeated k-mers, we provide genome-wide annotations of k-mer-uniqueness at http://cbio.mskcc.org/ approximately aarvey/repeatmap.


Subject(s)
In Situ Hybridization, Fluorescence/methods , RNA Probes/chemistry , RNA, Messenger/analysis , Animals , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Embryo, Nonmammalian/chemistry , Nuclear Proteins/genetics , RNA, Messenger/chemistry , Repetitive Sequences, Nucleic Acid , Transcription Factors/genetics
18.
Curr Biol ; 19(23): 2037-42, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19931455

ABSTRACT

The detection and counting of transcripts within single cells via fluorescent in situ hybridization (FISH) has allowed researchers to ask quantitative questions about gene expression at the level of individual cells. This method is often preferable to quantitative RT-PCR, because it does not necessitate destruction of the cells being probed and maintains spatial information that may be of interest. Until now, studies using FISH at single-molecule resolution have only been rigorously carried out in isolated cells (e.g., yeast cells or mammalian cell culture). Here, we describe the detection and counting of transcripts within single cells of fixed, whole-mount Drosophila embryos via a combination of FISH, immunohistochemistry, and image segmentation. Our method takes advantage of inexpensive, long RNA probes detected with antibodies, and we present novel evidence to show that we can robustly detect single mRNA molecules. We use this method to characterize transcription at the endogenous locus of the Hox gene Sex combs reduced (Scr), by comparing a stably expressing group of cells to a group that only transiently expresses the gene. Our data provide evidence for transcriptional bursting, as well for divergent "accumulation" and "maintenance" phases of gene activity at the Scr locus.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/embryology , RNA, Messenger/metabolism , Transcription Factors/metabolism , Transcription, Genetic/physiology , Animals , Drosophila Proteins/genetics , Gene Expression Regulation, Developmental/physiology , Immunohistochemistry , In Situ Hybridization, Fluorescence , RNA, Messenger/genetics , Transcription Factors/genetics
19.
Proc Natl Acad Sci U S A ; 106(7): 2224-9, 2009 Feb 17.
Article in English | MEDLINE | ID: mdl-19168633

ABSTRACT

Wounds in Drosophila and mouse embryos induce similar genetic pathways to repair epidermal barriers. However, the transcription factors that transduce wound signals to repair epidermal barriers are largely unknown. We characterize the transcriptional regulatory enhancers of 4 genes-Ddc, ple, msn, and kkv-that are rapidly activated in epidermal cells surrounding wounds in late Drosophila embryos and early larvae. These epidermal wound enhancers all contain evolutionarily conserved sequences matching binding sites for JUN/FOS and GRH transcription factors, but vary widely in trans- and cis-requirements for these inputs and their binding sites. We propose that the combination of GRH and FOS is part of an ancient wound-response pathway still used in vertebrates and invertebrates, but that other mechanisms have evolved that result in similar transcriptional output. A common, but largely untested assumption of bioinformatic analyses of gene regulatory networks is that transcription units activated in the same spatial and temporal patterns will require the same cis-regulatory codes. Our results indicate that this is an overly simplistic view.


Subject(s)
Epidermis/pathology , Gene Expression Regulation , Transcription Factors/metabolism , Wound Healing , Animals , Binding Sites , Drosophila , Drosophila melanogaster , Enhancer Elements, Genetic , Microscopy, Fluorescence , Models, Biological , Models, Genetic , Mutation , Time Factors , Transcription, Genetic
20.
Dev Genes Evol ; 218(6): 321-32, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18504607

ABSTRACT

Variations in Hox protein sequences and functions have been proposed to contribute to evolutionary changes in appendage shape and number in crustaceans and insects. One model is that insect Hox proteins of the Ultrabithorax (UBX) ortholog class evolved increased abilities to repress Distal-less (Dll) transcription and appendage development in part through the loss of serine and threonine residues in casein kinase 2 (CK2) phosphorylation sites. To explore this possibility, we constructed and tested the appendage repression function of chimeric proteins with insertions of different CK2 consensus sites or phosphomimetics of CK2 sites in C-terminal regions of Drosophila melanogaster UBX. Our results indicate that CK2 sites C-terminal to the homeodomain can inhibit the appendage repression functions of UBX proteins, but only in the context of specific amino acid sequences. Our results, combined with previous findings on evolutionary changes in Hox protein, suggest how intra-protein regulatory changes can diversify Hox protein function, and thus animal morphology.


Subject(s)
Casein Kinase II/metabolism , Homeodomain Proteins/metabolism , Homeodomain Proteins/physiology , Amino Acid Sequence , Animals , Animals, Genetically Modified , Binding Sites , Body Patterning/genetics , Chimera/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Molecular Sequence Data , Phosphorylation , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Thorax/embryology , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...